We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Transferrin and the transferrin receptor for the targeted delivery of therapeutic agents to the brain and cancer cells

    Christine Dufès

    * Author for correspondence

    Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.

    ,
    Majed Al Robaian

    Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK

    These authors contributed equally

    Search for more papers by this author

    &
    Sukrut Somani

    Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK

    These authors contributed equally

    Search for more papers by this author

    Published Online:https://doi.org/10.4155/tde.13.21

    The potential use of many promising novel drugs is limited by their inability to specifically reach their site of action after intravenous administration, without secondary effects on healthy tissues. In order to remediate this problem, the protein transferrin (Tf) has been extensively studied as a targeting molecule for the transport of drug and gene delivery systems to the brain and cancer cells. A wide range of delivery approaches have been developed to target the Tf receptor and they have already improved the specific delivery of Tf-bearing therapeutic agents to their site of action. This review provides a summary of the numerous delivery strategies used to target the Tf receptor and focuses on recent therapeutic advances.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Qian ZM, Li H, Sun H, Ho K. Targeted drug delivery via the transferrin receptor-mediated endocytosis pathway. Pharmacol. Rev.54,561–587 (2002).
    • Jefferies WA, Brandon MR, Hunt SV, Williams AF, Gatter KC, Mason DY. Transferrin receptor on endothelium of brain capillaries. Nature312,162–163 (1984).
    • Fishman JB, Rubin JB, Handrahan JV, Connor JR, Fine RE. Receptor-mediated transcytosis of transferrin across the blood–brain barrier. J. Neurosci. Res.18,299–304 (1987).
    • Bickel U, Yoshikawa T, Pardridge WM. Delivery of peptides and proteins through the blood–brain barrier. Adv. Drug Deliv. Rev.46,247–279 (2001).
    • Pardridge WM. Drug targeting to the brain. Pharm. Res.24,1733–1744 (2007).
    • Yang DC, Wang F, Elliott RL, Head JF. Expression of transferrin receptor and ferritin H-chain mRNA are associated with clinical and histopathological prognostic indicators in breast cancer. Anticancer Res.21,541–549 (2001).
    • Seymour GJ, Walsh MD, Lavin MF, Strutton G, Gardiner RA. Transferrin receptor expression by human bladder transitional cell carcinomas. Urol. Res.15,341–344 (1987).
    • Kondo K, Noguchi M, Mukai K et al. Transferrin receptor expression in adenocarcinoma of the lung as a histopathologic indicator of prognosis. Chest97,1367–1371 (1990).
    • Prior R, Reifenberger G, Wechsler W. Transferrin receptor expression in tumours of the human nervous system: relation to tumour type, grading and tumour growth fraction. Virchows Arch. A Pathol. Anat. Histopathol.416,491–496 (1990).
    • 10  Huebers HA, Finch CA. The physiology of transferrin and transferrin receptors. Physiol. Rev.67,520–582 (1987).
    • 11  Brandsma ME, Jevnikar AM, Ma S. Recombinant human transferrin: beyond iron binding and transport. Biotechnol. Adv.29,230–238 (2011).
    • 12  Aisen P, Leibman A, Zweier J. Stoichiometric and site characteristics of the binding of iron to human transferrin. J. Biol. Chem.253,1930–1937 (1978).
    • 13  Richardson DR, Ponka P. The molecular mechanisms of the metabolism and transport of iron in normal and neoplastic cells. Biophys. Acta1331,1–40 (1997).
    • 14  Baker HM, Anderson BF, Baker EN. Dealing with iron: common structural principles in proteins that transport iron and heme. Proc. Natl Acad. Sci. USA100,3579–3583 (2003).
    • 15  Neckers LM, Trepel JB. Transferrin receptor expression and the control of cell growth. Cancer Invest.4,461–470 (1986).
    • 16  Daniels TR, Delgado T, Rodriguez JA, Helguera G, Penichet ML. The transferrin receptor part I: biology and targeting with cytotoxic antibodies for the treatment of cancer. Clin. Immunol.121,144–158 (2006).
    • 17  Lai BT, Gao JP, Lanks KW. Mechanism of action and spectrum of cell lines sensitive to a doxorubicin-transferrin conjugate. Cancer Chemother. Pharmacol.41,155–160 (1998).
    • 18  Kawabata H, Germain RS, Vuong PT, Nakamaki T, Said JW, Koeffler HP. Transferrin receptor 2-alpha supports cell growth in iron-chelated cultured cells and in vivo. J. Biol. Chem.275,16618–16625 (2000).
    • 19  Lee HJ, Engelhardt B, Lesley J, Bickel U, Pardridge WM. Targeting rat anti-mouse transferrin receptor monoclonal antibodies through blood–brain barrier in mouse. J. Pharmacol. Exp. Ther.292,1048–1052 (2000).
    • 20  Friden PM, Walus LR, Musso GF, Taylor MA, Malfroy B, Starzyk RM. Anti-transferrin receptor antibody and antibody-drug conjugates cross the blood–brain barrier. Proc. Natl Acad. Sci. USA88,4771–4775 (1991).
    • 21  Zhang Y, Pardridge WM. Blood–brain barrier targeting of BDNF improves motor function in rats with middle cerebral artery occlusion. Brain Res.1111,227–229 (2006).
    • 22  Wu D, Pardridge WM. Central nervous system pharmacologic effect in conscious rats after intravenous injection of a biotinylated vasoactive intestinal peptide analog coupled to a blood–brain barrier drug delivery system. J. Pharmacol. Exp. Ther.279,77–83 (1996).
    • 23  Li XB, Liao GS, Shu YY, Tang SX. Brain delivery of biotinylated NGF bounded to an avidin-transferrin conjugate. J. Nat. Toxins.9,73–83 (2000).
    • 24  Lee HJ, Zhang Y, Zhu CN, Duff K, Pardridge WM. Imaging brain amyloid of Alzheimer disease in vivo in transgenic mice with an A beta peptide radiopharmaceutical. J. Cereb. Blood Flow Metab.22(2),223–231 (2002).
    • 25  Fu A, Zhou QH, Hui EK, Lu JZ, Boado RJ, Pardridge WM. Intravenous treatment of experimental Parkinson’s disease in the mouse with an IgG-GDNF fusion protein that penetrates the blood–brain barrier. Brain Res.1352,208–213 (2010).
    • 26  Pardridge WM. Vector-mediated drug delivery to the brain. Adv. Drug Delivery Rev.36,299–321 (1999).
    • 27  Zhang Y, Pardridge WM. Delivery of β-galactosidase to mouse brain via the blood–brain barrier transferrin receptor. J. Pharmacol. Exp. Ther.313,1075–1081 (2005).
    • 28  Xu G, Yong KT, Roy I et al. Bioconjugated quantum rods as targeted probes for efficient transmigration across an in vitro blood–brain barrier. Bioconjugate Chem.19,1179–1185 (2008).
    • 29  Mahajan SD, Roy I, Xu G, et al. Enhancing the delivery of anti-retroviral drug “Saquinavir” across the blood–brain barrier using nanoparticles. Curr. HIV Res.8,396–404 (2010).
    • 30  Liu S, Guo Y, Huang R et al. Gene and doxorubicin co-delivery system for targeting therapy of glioma. Biomaterials33,4907–4916 (2012).
    • 31  Aragnol D, Leserman LD. Immune clearance of liposomes inhibited by an anti-Fc receptor antibody in vivo. Proc. Natl Acad. Sci. USA83,2699–2703 (1986).
    • 32  Soni V, Kohli DV, Jain SK. Transferrin-conjugated liposomal system for improved delivery of 5-fluorouracil to brain. J. Drug Target.16,73–78 (2008).
    • 33  Huwyler J, Wu D, Pardridge WM. Brain drug delivery of small molecules using immunoliposomes. Proc. Natl Acad. Sci. USA93,14164–14169 (1996).
    • 34  Béduneau A, Saulnier P, Hindré F, Clavreul A, Leroux JC, Benoit JP. Design of targeted lipid nanocapsules by conjugation of whole antibodies and antibody Fab’ fragments. Biomaterials28,4978–4990 (2007).
    • 35  Béduneau A, Hindré F, Clavreul A, Leroux JC, Saulnier P, Benoit JP. Brain targeting using novel lipid nanovectors. J. Control. Release126,44–49 (2008).
    • 36  Kreuter J. Mechanism of polymeric nanoparticle-based drug transport across the blood–brain barrier. J. Microencapsul.30,49–54 (2013).
    • 37  Olivier JC. Drug transport to brain with targeted nanoparticles. NeuroRx2,108–119 (2005).
    • 38  Bao H, Jin X, Li L, Lv F, Liu T. OX26 modified hyperbranched polyglycerol-conjugated poly(lactic-co-glycolic acid) nanoparticles: synthesis, characterization and evaluation of its brain delivery ability. J. Mater. Sci. Mater. Med.23,1891–1901 (2012).
    • 39  Lalani J, Raichandani Y, Mathur R et al. Comparative receptor based brain delivery of tramadol-loaded poly (lactic-co-glycolic acid) nanoparticles. J. Biomed. Nanotechnol.8,918–927 (2012).
    • 40  Prades R, Guerrero S, Araya E et al. Delivery of gold nanoparticles to the brain by conjugation with a peptide that recognizes the transferrin receptor. Biomaterials33,7194–7205 (2012).
    • 41  Kedar U, Phutane P, Shidhaye S, Kadam V. Advances in polymeric micelles for drug delivery and tumor targeting. Nanomedicine6,714–729 (2010).
    • 42  Zhang P, Hu L, Yin Q, Zhang Z, Feng L, Li Y. Transferrin-conjugated polyphosphoester hybrid micelle loading paclitaxel for brain-targeting delivery: synthesis, preparation and in vivo evaluation. J. Control. Release159,429–434 (2012).
    • 43  Zhang P, Hu L, Yin Q, Feng L, Li Y. Transferrin-modified c[RGDfK]-Paclitaxel loaded hybrid micelle for sequential blood–brain barrier penetration and glioma targeting therapy. Mol. Pharm.9,1590–1598 (2012).
    • 44  Yang Y, Nunes FA, Berencsi K, Furth EE, Gonczol E, Wilson JM. Cellular-immunity to viral-antigens limits E1-deleted adenoviruses for gene-therapy. Proc. Natl Acad. Sci. USA91,4407–4411 (1994).
    • 45  Shi N, Pardridge WM. Non-invasive gene targeting to the brain. Proc. Natl Acad. Sci. USA97,7567–7572 (2000).
    • 46  Zhang Y, Schlachetzki F, Zhang YF, Boado RJ, Pardridge WM. Normalization of striatal tyrosine hydroxylase and reversal of motor impairment in experimental parkinsonism with intravenous nonviral gene therapy and a brain-specific promoter. Human Gene Ther.15,339–350 (2004).
    • 47  Zhang Y, Zhu C, Pardridge WM. Antisense gene therapy of brain cancer with an artificial virus gene delivery system. Mol. Ther.6,67–72 (2002).
    • 48  Shi NY, Zhang Y, Zhu CN, Boado RJ, Pardridge WM. Brain-specific expression of an exogenous gene after i.v. administration. Proc. Natl Acad. Sci. USA98,12754–12759 (2001).▪ Demonstrates that 8D3-conjugated PEGylated liposomes encapsulating plasmid DNA encoding β-galactosidase were able to deliver their cargo to the brain of mice following intravenous administration. When the enzyme expression was driven by a brain-specific promoter, gene expression was only observed in the brain.
    • 49  Suzuki T, Wu D, Schlachetzki F, Li JY, Boado RJ, Pardridge WM. Imaging endogenous gene expression in brain cancer in vivo with 111In-peptide nucleic acid antisense radiopharmaceuticals and brain drug-targeting technology. J. Nucl. Med.45,1766–1775 (2004).
    • 50  Xia CF, Zhang Y, Zhang Y, Boado RJ, Pardridge WM. Intravenous siRNA of brain cancer with receptor targeting and avidin-biotin technology. Pharm. Res.24,2309–2316 (2007).
    • 51  Dufès C, Uchegbu IF, Schätzlein AG. Dendrimers in gene delivery. Adv. Drug Deliv. Rev.57,2177–2202 (2005).
    • 52  Huang RQ, Qu YH, Ke WL, Zhu JH, Pei YY, Jiang C. Efficient gene delivery targeted to the brain using a transferrin-conjugated polyethyleneglycol-modified polyamidoamine dendrimer. FASEB J.21,117–1125 (2007).
    • 53  Daniels TR, Bernabeu E, Rodriguez JA et al. The transferrin receptor and the targeted delivery of therapeutic agents against cancer. Biochim. Biophys. Acta1820,291–317 (2012).▪▪ Very detailed description of transferrin (Tf), its receptor and all the Tf receptor-targeting delivery strategies attempted so far.
    • 54  Singh M, Atwal H, Micetich R. Transferrin directed delivery of Adriamycin to human cells. Anticancer Res.18,1423–1427 (1998).
    • 55  Lubgan D, Jozwiak Z, Grabenbauer GG, Distel LV. Doxorubicin-transferrin conjugate selectively overcomes multidrug resistance in leukaemia cells. Cell. Mol. Biol. Lett.14,113–127 (2009).
    • 56  Wang F, Jiang X, Yang DC, Elliott RL, Head JF. Doxorubicin-gallium-transferrin conjugate overcomes multidrug resistance: evidence for drug accumulation in the nucleus of drug resistant MCF-7/ADR cells. Anticancer Res.20,799–808 (2000).
    • 57  Head JF, Wang F, Elliott RL. Antineoplastic drugs that interfere with iron metabolism in cancer cells. Adv. Enzyme Regul.37,147–159 (1997).▪ Describes that a clinical trial for advanced breast cancer has observed a high response rate to the Tf–cisplatin conjugate in four out of 11 patients, including one complete response, with only minor side effects.
    • 58  Li X, Ding L, Xu Y, Wang Y, Ping Q. Targeted delivery of doxorubicin using stealth liposomes modified with transferrin. Int. J. Pharm.373,116–123 (2009).
    • 59  Suzuki S, Inoue K, Hongoh A, Hashimoto Y, Yamazoe Y. Modulation of doxorubicin resistance in a doxorubicin-resistant human leukaemia cell by an immunoliposome targeting transferrin receptor. Br. J. Cancer76,83–89 (1997).
    • 60  Dufès C, Muller JM, Olivier JC, Uchegbu IF, Schätzlein AG. Anticancer drug delivery with transferrin targeted polymeric chitosan vesicles. Pharm. Res.21,101–107 (2004).
    • 61  Fu JY, Blatchford DR, Tetley L, Dufès C. Tumor regression after systemic administration of tocotrienol entrapped in tumor-targeted vesicles. J. Control. Release140,95–99 (2009).
    • 62  Fu JY, Zhang W, Blatchford DR, Tetley L, McConnell G, Dufès C. Novel tocotrienol-entrapping vesicles can eradicate solid tumors after intravenous administration. J. Control. Release154,20–26 (2011).▪ Describes that intravenously administered Tf-bearing multilamellar vesicles entrapping tocotrienol led to complete tumor eradication for 40% of B16-F10 murine melanoma and 20% of A431 tumors, as well as improvement of animal survival.
    • 63  Dufès C. Delivery of the vitamin E compound tocotrienol to cancer cells. Ther. Deliv.2,1385–1389 (2011).
    • 64  Lemarié F, Chang CW, Blatchford DR et al. Anti-tumor activity of the tea polyphenol epigallocatechin gallate encapsulated in targeted vesicles after intravenous administration. Nanomedicine (Lond.)8(2),181–192 (2013).
    • 65  Karatas H, Aktas Y, Gursoy-Ozdemir Y et al. A nanomedicine transports a peptide caspase-3 inhibitor across the blood-brain barrier and provides neuroprotection. J. Neuroscience19,13761–13769 (2009).
    • 66  Shah N, Chaudhari K, Dantuluri P, Murthy RS, Das S. Paclitaxel-loaded PLGA nanoparticles surface modified with transferrin and Pluronic® P85, an in vitro cell line and in vivo biodistribution studies on rat model. J. Drug Target.17,533–542 (2009).
    • 67  Pulkkinen M, Pikkarainen J, Wirth T et al. Three-step tumor targeting of paclitaxel using biotinylated PLA-PEG nanoparticles and avidin-biotin technology: formulation development and in vitro anticancer activity. Eur. J. Pharm. Biopharm.70,66–74 (2008).
    • 68  Elsabahy M, Nazarali A, Foldvari M. Non-viral nucleic acid delivery: key challenges and future directions. Curr. Drug Deliv.8,235–244 (2011).
    • 69  Xu L, Frederik P, Pirollo KF et al. Self-assembly of a virus-mimicking nanostructure system for efficient tumor-targeted gene delivery. Hum. Gene Ther.13,469–481 (2002).
    • 70  Wenz G, Han BH, Muller A. Cyclodextrin rotaxanes and polyrotaxanes. Chem. Rev.106,782–817 (2006).
    • 71  Hu-Lieskovan S, Heidel JD, Bartlett DW, Davis ME, Triche TJ. Sequence-specific knockdown of EWS-FLI1 by targeted, nonviral delivery of small interfering RNA inhibits tumor growth in a murine model of metastatic Ewing’s sarcoma. Cancer Res.65,8984–8992 (2005).
    • 72  Vinogradov S, Batrakova E, Li S, Kabanov A. Polyion complex micelles with protein-modified corona for receptor-mediated delivery of oligonucleotides into cells. Bioconjug. Chem.10,851–860 (1999).
    • 73  Cotten M, Wagner E, Birnstiel ML. Receptor-mediated transport of DNA into eukaryotic cells. Method Enzymol.217,618–644 (1993).
    • 74  Kircheis R, Ostermann E, Wolschek MF et al. Tumor-targeted gene delivery of tumor necrosis factor-alpha induces tumor necrosis and tumor regression without systemic toxicity. Cancer Gene Ther.9,673–680 (2002).▪▪ Demonstrates that the systemically administered Tf-bearing polyethylenimine encoding TNFα resulted in antitumor effects on three tumor models, with complete regression of MethA tumors.
    • 75  Kursa M, Walker GF, Roessler V et al. Novel shielded transferrin-polyethylene glycol-polyethylenimine/DNA complexes for systemic tumor-targeted gene transfer. Bioconjug. Chem.14,222–231 (2003).
    • 76  Han L, Huang R, Li J, Liu S, Huang S, Jiang C. Plasmid pORF-hTRAIL and doxorubicin co-delivery targeting to tumor using peptide-conjugated polyamidoamine dendrimer. Biomaterials32,1242–1252 (2011).▪ Demonstrates that a Tf receptor-targeted PEGylated polyamidoamine dendrimer was able to co-deliver the drug doxorubicin together with a therapeutic plasmid encoding TNF-related apoptosis-inducing ligand to tumors and to inhibit 77% of tumor growth.
    • 77  Koppu S, Oh YJ, Edrada-Ebel R et al. Tumor regression after systemic administration of a novel tumor-targeted gene delivery system carrying a therapeutic plasmid DNA. J. Control. Release143,215–221 (2010).▪▪ Demonstrates that an intravenously administered Tf-bearing generation 3-diaminobutyric polypropylenimine dendriplex encoding TNFα led to complete disappearance of 90% of A431 human epidermoid carcinoma tumors in mice.
    • 78  Lemarié F, Croft DR, Tate RJ, Ryan KM, Dufès C. Tumor regression following intravenous administration of a tumor-targeted p73 gene delivery system. Biomaterials33,2701–2709 (2012).