Elsevier

Clinical Immunology

Volume 121, Issue 2, November 2006, Pages 159-176
Clinical Immunology

Review
The transferrin receptor part II: Targeted delivery of therapeutic agents into cancer cells

https://doi.org/10.1016/j.clim.2006.06.006Get rights and content

Abstract

Traditional anti-cancer treatments consist of chemotherapeutic drugs that effectively eliminate rapidly dividing tumor cells. However, in many cases chemotherapy fails to eliminate the tumor and even when chemotherapy is successful, its systemic cytotoxicity often results in detrimental side effects. To overcome these problems, many laboratories have focused on the design of novel therapies that exhibit tumor specific toxicity. The transferrin receptor (TfR), a cell membrane-associated glycoprotein involved in iron homeostasis and cell growth, has been explored as a target to deliver therapeutics into cancer cells due to its increased expression on malignant cells, accessibility on the cell surface, and constitutive endocytosis. The TfR can be targeted by direct interaction with conjugates of its ligand transferrin (Tf) or by monoclonal antibodies specific for the TfR. In this review we summarize the strategies of targeting the TfR in order to deliver therapeutic agents into tumor cells by receptor-mediated endocytosis.

Introduction

The TfR (also known as CD71), a type II transmembrane glycoprotein found as a homodimer (180 kDa) on the surface of cells, is a vital protein involved in iron homeostasis and the regulation of cell growth (recently reviewed in [1]). The TfR monomer contains a large extracellular C-terminal domain, a single-pass transmembrane domain, and a short intracellular N-terminal domain. The TfR is ubiquitously expressed on normal cells and expression is increased on cells with a high proliferation rate or on cells that require large amounts of iron [1]. Little or no TfR expression has been detected on pluripotent hematopoietic stem cells, while late erythroid and myeloid progenitor cells demonstrate TfR expression. Expression of the TfR is significantly upregulated in a variety of malignant cells and in many cases, increased expression correlates with tumor stage and is associated with poor prognosis [1].

Iron is involved in a variety of cellular processes and is a required co-factor for many enzymatic reactions including those involved in metabolism, respiration, and DNA synthesis [2], [3]. Delivery and cellular uptake of iron occurs through the interaction and internalization of iron-loaded Tf mediated by the TfR [1] (Fig. 1). Tf is a monomeric glycoprotein (apo-Tf) that can transport one (monoferric Tf) or two (diferric Tf) iron atoms. Diferric Tf has the highest affinity for the TfR and is 10- to 100-fold greater than that of apo-Tf at physiological pH [3]. Upon binding the TfR, the Tf/TfR complex is internalized in clathrin-coated pits through receptor-mediated endocytosis. Due to the decrease in pH, iron is released from transferrin in the endosome. Tf remains bound to the receptor at this pH and the apo-Tf/TfR complex is recycled back to the cell surface where apo-Tf is then released. The TfR is constitutively recycled independently of Tf binding.

A second transferrin receptor (TfR2) was identified and has a 25-fold lower affinity for Tf than TfR1 [1], [4], [5], [6]. The human TfR2 α and β transcripts are produced by alternative splicing [6]. The TfR2 α and TfR1 only show similarity in their extracellular domains. In contrast to TfR1, TfR2α expression appears to be limited to hepatocytes and enterocytes of the small intestine and is not regulated by intracellular iron levels. High surface expression of TfR2α was detected in many solid and hematopoietic malignant human cell lines. The intracellular TfR2 β (lacks the transmembrane and cytoplasmic domains) is ubiquitously expressed at low levels and its function remains unclear.

Traditional cancer therapy consists of chemotherapeutic drugs that can be successful in irradicating the tumor, but are often toxic to normal cells as well. Targeting the TfR is a promising strategy actively being explored as a drug alternative to offset these dangerous side effects. The high levels of expression of TfR in cancer cells, which may be up to 100-fold higher than the average expression of normal cells [7], [8], [9], its extracellular accessibility, its ability to internalize, and its central role in the cellular pathology of human cancer, make this receptor an attractive target for cancer therapy. In fact, the TfR can be successfully used to deliver cytotoxic agents into malignant cells including chemotherapeutic drugs, cytotoxic proteins, or high molecular weight compounds including liposomes, viruses, or nanoparticles (Fig. 2).

Section snippets

Doxorubicin (Adriamycin®)

Doxorubicin (Adriamycin®) (ADR) is an anthracycline anticancer drug that blocks DNA synthesis and also blocks the activity of topoisomerase II, an enzyme that helps to relax the coil and extend the DNA molecule prior to DNA synthesis or RNA transcription. ADR is used to treat leukemia, breast cancer, and many other cancers. When used alone ADR often exhibits side effects including cardiotoxicity, myelosuppression, nephrotoxicity, and extravasation [10]. Systemic drug toxicity is often

Delivery of toxic proteins

In addition to chemotherapeutic drugs, the TfR has been used for the targeted delivery of toxic proteins into malignant cells. An immunotoxin describes a cell-specific ligand linked to a plant or bacterial toxin or modified toxin subunit [35], [36]. The cell-specific ligand can either be an antibody, antibody fragment, cytokine, or other ligand that binds specifically to target cells and results in the internalization of the immunotoxin [35], [36]. The TfR has been targeted by many immunotoxins

Delivery of high molecular weight compounds

Many tumor therapies seek to deliver therapeutic high molecular weight compounds including genes, which either restore the normal function of a defective gene and/or are capable of destroying the malignant cell. One of the major hurdles of these therapies is adequate delivery of the therapeutic agent into target cells. Genetically engineered viral vectors are highly efficient in delivery. However, viral vectors face potential problems including the induction the host immune response. There is a

Conclusion

The TfR is an attractive targeting molecule that can be used to treat a variety of malignancies. Targeting the TfR can occur via one of two ways: either through Tf itself, which targets both TfR1 and TfR2, or through the use of monoclonal antibodies specific for TfR1 and potentially specific for TfR2. Targeting the TfR has been shown to be effective in delivering therapeutic agents, including chemotherapeutic drugs, toxic proteins, and high molecular weight compounds into cells and causing

Acknowledgments

This work was supported in part by grants K01 CA86915 and R01 CA107023 from NCI/NIH, the 2004 Brian D. Novis International Myeloma Foundation Senior Grant Award, and the 2003 Jonsson Cancer Center Foundation Interdisciplinary Grant “Targeted Therapy of Multiple Myeloma.”

References (113)

  • M. Fritzer et al.

    Cytotoxic effects of a doxorubicin–transferrin conjugate in multidrug-resistant KB cells

    Biochem. Pharmacol.

    (1996)
  • S. Neidle

    The molecular basis for the action of some DNA-binding drugs

    Prog. Med. Chem.

    (1979)
  • K. Barabas et al.

    Transferrin conjugates of adriamycin are cytotoxic without intercalating nuclear DNA

    J. Biol. Chem.

    (1992)
  • T. Hoshino et al.

    In vitro cytotoxicities and in vivo distribution of transferrin-platinum (II) complexes

    J. Pharm. Sci.

    (1995)
  • J.F. Head et al.

    Antineoplastic drugs that interfere with iron metabolism in cancer cells

    Advan Enzyme Regul.

    (1997)
  • T. Tanaka et al.

    Tumor targeting based on the effect of enhanced permeability and retention (EPR) and the mechanism of receptor-mediated endocytosis (RME)

    Int. J. Pharm.

    (2004)
  • A.E. Frankel et al.

    Immunotoxin therapy of hematologic malignancies

    Semin. Oncol.

    (2003)
  • V. Raso et al.

    A highly cytotoxic human transferrin-ricin A chain conjugate used to select receptor-modified cells

    J. Biol. Chem.

    (1984)
  • J. Lesley et al.

    Effect of an anti-murine transferrin receptor-ricin A conjugate on bone marrow stem and progenitor cells treated in vitro

    Exp. Cell Res.

    (1984)
  • S. Ramakrishnan et al.

    Characterization of a translation inhibitory protein from Luffa aegyptiaca

    Biochem. Biophys. Res. Commun.

    (1989)
  • D. Rathore et al.

    Generation of active immunotoxins containing recombinant restrictocin

    Biochem. Biophys. Res. Commun.

    (1996)
  • D. Rathore et al.

    Overproduction of fungal ribotoxin alpha-sarcin in Escherichia coli: generation of an active immunotoxin

    Gene

    (1997)
  • D.O. O'Keefe et al.

    Characterization of a transferrin-diphtheria toxin conjugate

    J. Biol. Chem.

    (1985)
  • V.G. Johnson et al.

    The role of the diphtheria toxin receptor in cytosol translocation

    J. Biol. Chem.

    (1988)
  • S. Leppla et al.

    Inhibition of diphtheria toxin degradation and cytotoxic action by chloroquine

    J. Biol. Chem.

    (1980)
  • S.M. Rybak et al.

    Cytotoxic potential of ribonuclease and ribonuclease hybrid proteins

    J. Biol. Chem.

    (1991)
  • D.L. Newton et al.

    Expression and characterization of recombinant human eosinophil-derived neurotoxin and eosinophil-derived neurotoxin-anti-transferrin receptor sFv

    J. Biol. Chem.

    (1994)
  • D.L. Newton et al.

    Antitransferrin receptor antibody-RNase fusion protein expressed in the mammary gland of transgenic mice

    J. Immunol. Methods

    (1999)
  • H. Hatakeyama et al.

    Factors governing the in vivo tissue uptake of transferrin-coupled polyethylene glycol liposomes in vivo

    Int. J. Pharm.

    (2004)
  • C. Fonseca et al.

    Targeting of sterically stabilised pH-sensitive liposomes to human T-leukaemia cells

    Eur. J. Pharm. Biopharm.

    (2005)
  • Y. Yamada et al.

    Mitochondrial delivery of mastoparan with transferrin liposomes equipped with a pH-sensitive fusogenic peptide for selective cancer therapy

    Int. J. Pharm.

    (2005)
  • N.D. Smith et al.

    The p53 tumor suppressor gene and nuclear protein: basic science review and relevance in the management of bladder cancer

    J. Urol.

    (2003)
  • A.C. Prost et al.

    Differential transferrin receptor density in human colorectal cancer: a potential probe for diagnosis and therapy

    Int. J. Oncol.

    (1998)
  • H. Shinohara et al.

    Site-specific expression of transferrin receptor by human colon cancer cells directly correlates with eradication by antitransferrin recombinant immunotoxin

    Int. J. Oncol.

    (2000)
  • C.E. Myers et al.

    Anthracyclins

  • Y.H. Takakura et al.

    Macromolecular drug carrier systems in cancer chemotherapy: macromolecular prodrugs

    Crit. Rev. Oncol. Hematol.

    (1994)
  • M. Singh et al.

    Transferrin directed delivery of adriamycin to human cells

    Anticancer Res.

    (1998)
  • M. Fritzer et al.

    Cytotoxicity of a transferrin-adriamycin conjugate to anthracycline-resistant cells

    Int. J. Cancer

    (1992)
  • T. Hatano et al.

    Cytotoxic effect of the protein–doxorubicin conjugates on the multidrug-resistant human myelogenous leukemia cell line, K562, in vitro

    Tumour Biol.

    (1993)
  • A. Berczi et al.

    Influence of conjugation of doxorubicin to transferrin on the iron uptake by K562 cells via receptor-mediated endocytosis

    Eur. J. Biochem.

    (1993)
  • B.T. Lai et al.

    Mechanism of action and spectrum of cell lines sensitive to a doxorubicin-transferrin conjugate

    Cancer Chemother. Pharmacol.

    (1998)
  • G.R. Braslawsky et al.

    Adriamycin(hydrazone)–antibody conjugates require internalization and intracellular acid hydrolysis for antitumor activity

    Cancer Immunol. Immunother.

    (1991)
  • F. Wang et al.

    Doxorubicin–gallium–transferrin conjugate overcomes multidrug resistance: evidence for drug accumulation in the nucleus of drug resistant MCF-7/ADR cells

    Anticancer Res.

    (2000)
  • C.R. Chitambar

    Gallium compounds as antineoplastic agents

    Curr. Opin. Oncol.

    (2004)
  • K. Barabas et al.

    Evidence in support of the plasma membrane as the target for transferrin–adriamycin conjugates in K562 cells

    Am. J. Reprod. Immunol.

    (1991)
  • R.L. Elliott et al.

    Preliminary evaluation of platinum transferrin (MPTC-63) as a potential nontoxic treatment for breast cancer

    Cancer Detect. Prev.

    (1988)
  • J.F. Head et al.

    Cisplatin-transferrin complex acts synergistically with doxorubicin to inhibit the growth of cultured MCF-7 cells

    International Symposium on Platinum and Other Metal Coordination Compounds in Cancer Chemotherapy

    (1991)
  • U. Beyer et al.

    Synthesis and in vitro efficacy of transferrin conjugates of the anticancer drug chlorambucil

    J. Med. Chem.

    (1998)
  • G.L. Mayers et al.

    Transferrin-gemcitabine conjugate: application to chemotherapy

  • N. Bejaoui et al.

    Cytotoxicity of transferrin–daunorubicin conjugates on small cell carcinoma of the lung (SCCL) cell line NCI-H69

    Anticancer Res.

    (1991)
  • Cited by (0)

    View full text