524
Views
9
CrossRef citations to date
0
Altmetric
Review

Improving the targeting of therapeutics with single-domain antibodies

, , &
Pages 561-570 | Received 23 Jul 2015, Accepted 15 Dec 2015, Published online: 25 Jan 2016

References

  • Craik DJ, Fairlie DP, Liras S, et al. The future of peptide-based drugs. Chem Biol Drug Des. 2013;81:136–147.
  • Carter PJ. Introduction to current and future protein therapeutics: A protein engineering perspective. Exp Cell Res. 2011;317:1261–1269.
  • Doane TL, Burda C. The unique role of nanoparticles in nanomedicine: Imaging, drug delivery and therapy. Chem Soc Rev. 2012;41:2885–2911.
  • Gosu V, Basith S, Kwon O-P, et al. Therapeutic applications of nucleic acids and their analogues in toll-like receptor signaling. Molecules. 2012;17:13503–13529.
  • Dash A, Knapp FF, Pillai MR. Targeted radionuclide therapy–an overview. Curr Radiopharm. 2013;6:152–180.
  • Yuan Z, Yi X, Zhang J, et al. A prodrug nanoassembly entrapping drugs as a tumor-targeted delivery system. Chem Commun (Camb). 2013;49:801–803.
  • Xing H, Wong NY, Xiang Y, et al. DNA aptamer functionalized nanomaterials for intracellular analysis, cancer cell imaging and drug delivery. Curr Opin Chem Biol. 2012;16:429–435.
  • Yang X, Liu X, Liu Z, et al. Near-infrared light-triggered, targeted drug delivery to cancer cells by aptamer gated nanovehicles. Adv Mater. 2012;24:2890–2895.
  • Zhu G, Zheng J, Song E, et al. Self-assembled, aptamer-tethered DNA nanotrains for targeted transport of molecular drugs in cancer theranostics. Proc Natl Acad Sci U S A. 2013;110:7998–8003.
  • Jin E, Zhang B, Sun X, et al. Acid-active cell-penetrating peptides for in vivo tumor-targeted drug delivery. J Am Chem Soc. 2013;135:933–940.
  • Pan L, He Q, Liu J, et al. Nuclear-targeted drug delivery of tat peptide-conjugated monodisperse mesoporous silica nanoparticles. J Am Chem Soc. 2012;134:5722–5725.
  • Wu AM, Senter PD. Arming antibodies: Prospects and challenges for immunoconjugates. Nat Biotechnol. 2005;23:1137–1146.
  • Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–497.
  • Goldman ER, Brozozog-Lee PA, Zabetakis D, et al. Negative tail fusions can improve ruggedness of single domain antibodies. Protein Expr Purif. 2014;95:226–232.
  • Liu JL, Zabetakis D, Brown JC, et al. Thermal stability and refolding capability of shark derived single domain antibodies. Mol Immunol. 2014;59:194–199.
  • Liu JL, Zabetakis D, Walper SA, et al. Bioconjugates of rhizavidin with single domain antibodies as bifunctional immunoreagents. J Immunol Methods. 2014;411:37–42.
  • Walper SA, Battle SR, Audrey Brozozog Lee P, et al. Thermostable single domain antibody-maltose binding protein fusion for bacillus anthracis spore protein bcla detection. Anal Biochem. 2014;447:64–73.
  • Holliger P, Hudson PJ. Engineered antibody fragments and the rise of single domains. Nat Biotechnol. 2005;23:1126–1136.
  • Anderson GP, Legler PM, Zabetakis D, et al. Comparison of immunoreactivity of staphylococcal enterotoxin b mutants for use as toxin surrogates. Anal Chem. 2012;84:5198–5203.
  • Shia WW, Bailey RC. Single domain antibodies for the detection of ricin using silicon photonic microring resonator arrays. Anal Chem. 2013;85:805–810.
  • Swain MD, Anderson GP, Serrano-González J, et al. Immunodiagnostic reagents using llama single domain antibody-alkaline phosphatase fusion proteins. Anal Biochem. 2011;417:188–194.
  • Walper SA, Lee PAB, Goldman ER, et al. Comparison of single domain antibody immobilization strategies evaluated by surface plasmon resonance. J Immunol Methods. 2013;388:68–77.
  • Harmsen MM, De Haard HJ. Properties, production, and applications of camelid single-domain antibody fragments. Appl Microbiol Biotechnol. 2007;77:13–22.
  • Muruganandam A, Tanha J, Narang S, et al. Selection of phage-displayed llama single-domain antibodies that transmigrate across human blood-brain barrier endothelium. Faseb J. 2001;15:240–242.
  • Rissiek B, Koch-Nolte F, Magnus T. Nanobodies as modulators of inflammation: Potential applications for acute brain injury. Front Cell Neurosci. 2014;8:1–7.
  • Van der Linden RH, Frenken LG, De Geus B, et al. Comparison of physical chemical properties of llama vhh antibody fragments and mouse monoclonal antibodies. Biochim Biophys Acta. 1999;1431:37–46.
  • Harmsen MM, van Solt CB, Van Zijderveld-Van Bemmel AM, et al. Selection and optimization of proteolytically stable llama single-domain antibody fragments for oral immunotherapy. Appl Microbiol Biotechnol. 2006;72:544–551.
  • Desmyter A, Transue TR, Ghahroudi MA, et al. Crystal structure of a camel single-domain vh antibody fragment in complex with lysozyme. Nat Struct Biol. 1996;3:803–811.
  • Anderson GP, Glaven RH, Algar WR, et al. Single domain antibody-quantum dot conjugates for ricin detection by both fluoroimmunoassay and surface plasmon resonance. Analytica Chimica Acta. 2013;786:132–138.
  • Glaven RH, Anderson GP, Zabetakis D, et al. Linking single domain antibodies that recognize different epitopes on the same target. Biosensors. 2012;2:43–56.
  • Stone E, Hirama T, Tanha J, et al. The assembly of single domain antibodies into bispecific decavalent molecules. J Immunol Methods. 2007;318:88–94.
  • Sherwood LJ, Hayhurst A. Hapten mediated display and pairing of recombinant antibodies accelerates assay assembly for biothreat countermeasures. Sci Rep. 2012;2:807.
  • Raphael MP, Christodoulides JA, Byers JM, et al. Optimizing nanoplasmonic biosensor sensitivity with orientated single domain antibodies. Plasmonics (Norwell, Mass). 2015;10:1649–1655.
  • Oliveira S, Heukers R, Sornkom J, et al. Targeting tumors with nanobodies for cancer imaging and therapy. J Control Release. 2013;172:607–617.
  • Feng Y, Zhu Z, Chen W, et al. Conjugates of small molecule drugs with antibodies and other proteins. Biomedicines. 2014;2:1–13.
  • Alves NJ, Champion MM, Stefanick JF, et al. Selective photocrosslinking of functional ligands to antibodies via the conserved nucleotide binding site. Biomaterials. 2013;34:5700–5710.
  • Ramakrishnan B, Boeggeman E, Manzoni M, et al. Multiple site-specific in vitro labeling of single-chain antibody. Bioconjug Chem. 2009;20:1383–1389.
  • Ramakrishnan B, Boeggeman E, Qasba PK. Novel method for in vitro o-glycosylation of proteins: application for bioconjugation. Bioconjug Chem. 2007;18:1912–1918.
  • Hofer T, Skeffington LR, Chapman CM, et al. Molecularly defined antibody conjugation through a selenocysteine interface. Biochemistry. 2009;48:12047–12057.
  • Axup JY, Bajjuri KM, Ritland M, et al. Synthesis of sitespecific antibody-drug conjugates using unnatural amino acids. Proc Natl Acad Sci USA. 2012;109:16101–16106.
  • Francisco JA, Cerveny CG, Meyer DL, et al. Cac10-vcmmae, an anti-cd30-monomethyl auristatin e conjugate with potent and selective antitumor activity. Blood. 2003;102:1458–1465.
  • Heukers R, van Bergen en Henegouwen PMP, Oliveira S. Nanobody-photosensitizer conjugates for targeted photodynamic therapy. Nanomedicine. 2014;10:1441–1451.
  • Sha H, Zou Z, Xin K, et al. Tumor-penetrating peptide fused egfr single-domain antibody enhances cancer drug penetration into 3d multicellular spheroids and facilitates effective gastric cancer therapy. J Control Release. 2015;200:188–200.
  • Hirsch LR, Stafford RJ, Bankson JA, et al. Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance. Proc Natl Acad Sci U S A. 2003;100:13549–13554.
  • Huang X, El-Sayed IH, Qian W, et al. Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. J Am Chem Soc. 2006;128:2115–2120.
  • van de Broek B, Devoogdt N, D’Hollander A, et al. Specific cell targeting with nanobody conjugated branched gold nanoparticles for photothermal therapy. ACS Nano. 2011;5:4319–4328.
  • Kirui DK, Rey DA, Batt CA. Gold hybrid nanoparticles for targeted phototherapy and cancer imaging. Nanotechnology. 2010;21:105105.
  • Day ES, Morton JG, West JL. Nanoparticles for thermal cancer therapy. J Biomech Eng. 2009;131:074001.
  • Arruebo M, Valladares M, Gonzalez-Fernandez A. Antibody-conjugated nanoparticles for biomedical applications. J Nanomater. 2009;439389:1–24.
  • Sapsford KE, Algar WR, Berti L, et al. Functionalizing nanoparticles with biological molecules: Developing chemistries that facilitate nanotechnology. Chem Rev. 2013;113:1904–2074.
  • Gujrati V, Kim S, Kim S-H, et al. Bioengineered bacterial outer membrane vesicles as cell-specific drug-delivery vehicles for cancer therapy. ACS Nano. 2014;8:1525–1537.
  • Altintas I, Heukers R, van der Meel R, et al. Nanobody-albumin nanoparticles (nanaps) for the delivery of a multikinase inhibitor 17864 to egfr overexpressing tumor cells. J Control Release. 2013;165:110–118.
  • Goeddel DV, Kleid DG, Bolivar F, et al. Expression in Escherichia coli of chemically synthesized genes for human insulin. Proc Natl Acad Sci U S A. 1979;76:106–110.
  • Hermanson GT. Bioconjugate techniques, 3rd edition. London: Academic Press; 2013. p. 1–1146.
  • Rashidian M, Dozier JK, Distefano MD. Enzymatic labeling of proteins: Techniques and approaches. Bioconjug Chem. 2013;24:1277–1294.
  • Kim CH, Axup JY, Schultz PG. Protein conjugation with genetically encoded unnatural amino acids. Curr Opin Chem Biol. 2013;17:412–419.
  • Alewine C, Hassan R, Pastan I. Advances in anticancer immunotoxin therapy. Oncologist. 2015;20:176–185.
  • Behdani M, Zeinali S, Karimipour M, et al. Development of vegfr2-specific nanobody pseudomonas exotoxin a conjugated to provide efficient inhibition of tumor cell growth. N Biotechnol. 2013;30:205–209.
  • Altintas I, Kok RJ, Schiffelers RM. Targeting epidermal growth factor receptor in tumors: From conventional monoclonal antibodies via heavy chain-only antibodies to nanobodies. Eur J Pharm Sci. 2012;45:399–407.
  • Cortez-Retamozo V, Backmann N, Senter PD, et al. Efficient cancer therapy with a nanobody-based conjugate. Cancer Res. 2004;64:2853–2857.
  • Tian B, Wong WY, Hegmann E, et al. Production and characterization of a camelid single domain antibody–urease enzyme conjugate for the treatment of cancer. Bioconjug Chem. 2015;26(6):1144–1155.
  • Shi J, Kundrat L, Pishesha N, et al. Engineered red blood cells as carriers for systemic delivery of a wide array of functional probes. Proc Natl Acad Sci U S A. 2014;111:10131–10136.
  • De Vos J, Devoogdt N, Lahoutte T, et al. Camelid single-domain antibody-fragment engineering for (pre)clinical in vivo molecular imaging applications: Adjusting the bullet to its target. Expert Opin Biol Ther. 2013;13:1149–1160.
  • D’Huyvetter M, Xavier C, Caveliers V, et al. Radiolabeled nanobodies as theranostic tools in targeted radionuclide therapy of cancer. Expert Opin Drug Deliv. 2014;11:1939–1954.
  • D’Huyvetter M, Vincke C, Xavier C, et al. Targeted radionuclide therapy with a 177lu-labeled anti-her2 nanobody. Theranostics. 2014;4:708–720.
  • Chames P, Van Regenmortel M, Weiss E, et al. Therapeutic antibodies: Successes, limitations and hopes for the future. Br J Pharmacol. 2009;157:220–233.
  • Cortez-Retamozo V, Lauwereys M, Gh GH, et al. Efficient tumor targeting by single-domain antibody fragments of camels. Int J Cancer. 2002;98:456–462.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.