Elsevier

Methods in Enzymology

Volume 503, 2012, Pages 269-292
Methods in Enzymology

Chapter eleven - Reengineering Biopharmaceuticals for Targeted Delivery Across the Blood–Brain Barrier

https://doi.org/10.1016/B978-0-12-396962-0.00011-2Get rights and content

Abstract

Recombinant protein therapeutics cannot enter brain drug development because these large molecule drugs do not cross the blood–brain barrier (BBB). However, recombinant proteins can be reengineered as BBB-penetrating IgG fusion proteins, where the IgG part is a genetically engineered monoclonal antibody (MAb) against an endogenous BBB receptor, such as the human insulin receptor (HIR) or the transferrin receptor (TfR). The IgG binds the endogenous insulin receptor or TfR to trigger transport across the BBB and acts as a molecular Trojan horse (MTH) to ferry into brain the fused protein therapeutic. The most potent MTH to date is a MAb against the HIR, designated the HIRMAb, which is active in humans and Old World primates, such as the Rhesus monkey. There is no known MAb against the mouse insulin receptor. For drug delivery in the mouse, protein therapeutics are fused to a chimeric MAb against the mouse TfR, designated the cTfRMAb. The HIRMAb or cTfRMAb Trojan horses have been engineered and expressed as fusion proteins with multiple classes of protein therapeutics, including lysosomal enzymes, neurotrophins, decoy receptors, single chain Fv therapeutic antibodies, and avidin. The pharmacokinetic (PK) properties of the IgG fusion proteins differ from that of typical MAb drugs and resemble the PK profiles of small molecules due to rapid uptake by peripheral tissues, as well as brain. The brain uptake of the IgG fusion proteins, 2–3% of injected dose/brain, is comparable to the brain uptake of small molecules. The IgG fusion proteins have been administered chronically in mouse models, and the immune response is low titer and has no effect on the fusion protein clearance from blood or brain uptake in vivo. The BBB MTH technology enables the reengineering of a wide spectrum of recombinant protein therapeutics for targeted drug delivery to the brain.

Introduction

Treatment of the brain with the products of biotechnology, for example, recombinant proteins and monoclonal antibodies (MAbs), or even short interfering RNA (siRNA), is not possible without fundamental solutions to the problem of blood–brain barrier (BBB) delivery. All biotechnological pharmaceutics are large molecules that do not cross the BBB. In the absence of an effective BBB drug delivery technology, the brain drug developer is left with the traditional, yet ineffective, brain drug delivery strategies, including transcranial drug delivery to the brain, BBB disruption, or small molecules. Transcranial drug delivery, such as convection-enhanced diffusion, only delivers drug to the local injection site (Pardridge, 2010) and is ineffective as a brain drug delivery technology for the 1200 g human brain. BBB disruption leads to chronic neuropathologic changes (Salahuddin et al., 1988) and is too toxic to be widely used in humans. Small molecules are hardly an alternative strategy because 98% of small molecules tested do not cross the BBB. To be brain penetrating, the small molecule must be lipid soluble, form < 8–10 hydrogen bonds with water, and have a molecular weight < 400–500 Daltons (Da) (Pardridge, 2005). Few small molecule pharmaceutical candidates have these molecular properties. Thus, even if a peptidomimetic small molecule were produced in lieu of drug development of a recombinant protein, the small molecule would most likely still need a BBB drug targeting technology to advance in clinical drug development.

The failure of biotechnology to treat the brain is illustrated with the biologic tumor necrosis factor (TNF)-α inhibitors (TNFI). Etanercept, the TNF receptor (TNFR); decoy receptor:Fc fusion protein, infliximab; the chimeric anti-TNFα MAb; and adalimumab, the human anti-TNFα MAb, had combined revenues of $16 billion in 2008 (Tansey and Szymbowski, 2009). However, there was no penetration of the CNS markets with the biologic TNFIs, despite the primary role played by TNFα in chronic brain diseases such as Alzheimer's disease (AD) or Parkinson's disease (PD) (Park and Bowers, 2010). The biologic TNFIs have not been developed for the brain because these molecules do not cross the BBB, and no BBB drug targeting technology has been developed within the pharmaceutical industry. The purpose of this chapter is to review the BBB molecular Trojan horse (MTH) technology for BBB-targeted delivery of recombinant protein pharmaceuticals.

Section snippets

Blood–Brain Barrier Receptor-Mediated Transport and Molecular Trojan Horses

l-DOPA is an effective drug for PD because this water-soluble small molecule penetrates the BBB via carrier-mediated transport (CMT) on the BBB large neutral amino-acid transporter type 1, which is LAT1 (Boado et al., 1999). Once in brain, the l-DOPA is decarboxylated to dopamine, the monoamine deficient in PD. Apart from LAT1, other BBB CMT systems include the GLUT1 glucose transporter, the CNT2 concentrative nucleoside transporter, and others (Pardridge, 2005). The CMT systems transport

Reengineering Recombinant Proteins for Targeted Brain Delivery

The most active BBB MTH is a MAb against the HIR. The BBB expresses an insulin receptor, and this has been demonstrated with human brain capillaries used as an in vitro model of the human BBB (Pardridge et al., 1985). A murine MAb against the HIR bound to the capillary endothelium in brain of the Rhesus monkey in an immunocytochemical study (Pardridge et al., 1995). This HIRMAb rapidly penetrates the BBB in the Rhesus monkey following IV administration with a brain uptake of 2–3% ID/brain (

Genetic Engineering of Expression Plasmid DNA Encoding IgG Fusion Proteins

IgG fusion proteins such as those depicted in Figure 11.1, Figure 11.2 are heterotetrameric proteins comprising two HCs and two LCs, which are glycosylated both at a single site on the C-region of the HC and at one or multiple sites on the therapeutic protein. Such proteins are generally expressed in eukaryotic host cells transfected with separate HC and LC genes. High host cell expression of the IgG fusion protein requires equally high expression of both the HC and the LC gene. In addition,

Pharmacokinetics

The HIRMAb and cTfRMAb fusion proteins are purified by protein A and protein G affinity chromatography, respectively, following collection of host cell-conditioned serum-free medium. For fusion protein manufacturing for GLP toxicology studies, or GMP production of the fusion protein, the protein A affinity column is followed by a cation exchange and/or anion exchange chromatographic step. Preclinical PK and brain uptake studies are performed in the Rhesus monkey for the HIRMAb fusion proteins

CNS Pharmacological Effects of IgG Fusion Proteins

The BBB insulin receptor and TfR are actual transport systems and cause the net movement of the ligand across the endothelial barrier followed by distribution into brain parenchyma. The delivery to brain cells beyond the BBB was demonstrated by emulsion autoradiography at the light microscopic level for insulin (Duffy and Pardridge, 1987), for transferrin (Skarlatos et al., 1995), and for HIRMAb fusion proteins (Boado and Pardridge, 2009). The capillary depletion method was developed to provide

Immune Response Against IgG Fusion Proteins

The chronic treatment of mice with cTfRMAb fusion proteins has not resulted in a preclinically significant immune response. Even in the MPSI mouse study, where the mice had never been exposed to the IDUA lysosomal enzyme, the immune response was of low titer (Boado et al., 2011). In a chronic treatment study, mice were treated for 12 weeks with twice weekly cTfRMAb-GDNF fusion protein at a dose of 2 mg/kg/dose or 4 mg/kg/week (Zhou et al., 2011d). The immune response against the fusion protein was

Summary

Biotechnology has considerable potential for the development of new treatments of brain and spinal cord disorders. However, recombinant proteins must be reengineered to enable BBB transport and brain penetration. This is possible with the MTH technology, wherein recombinant proteins are fused to genetically engineered, receptor-specific MAb. The MAb, such as the HIRMAb, crosses the BBB via RMT and carries the fused therapeutic protein into brain. The IgG fusion proteins have PK profiles very

References (65)

  • W.M. Pardridge

    The blood-brain barrier: Bottleneck in brain drug development

    NeuroRx

    (2005)
  • K.M. Park et al.

    Tumor necrosis factor-alpha mediated signaling in neuronal homeostasis and dysfunction

    Cell. Signal.

    (2010)
  • S. Skarlatos et al.

    Transport of [125I]transferrin through the rat blood-brain barrier in vivo

    Brain Res.

    (1995)
  • G.C. Su et al.

    Intravascular infusions of soluble beta-amyloid compromise the blood-brain barrier, activate CNS glial cells and induce peripheral hemorrhage

    Brain Res.

    (1999)
  • M.G. Tansey et al.

    The TNF superfamily in 2009: New pathways, new indications, and new drugs

    Drug Discov. Today

    (2009)
  • Y.Q. Xue et al.

    Intrastriatal administration of erythropoietin protects dopaminergic neurons and improves neurobehavioral outcome in a rat model of Parkinson's disease

    Neuroscience

    (2007)
  • Y. Zhang et al.

    Mediated efflux of IgG molecules from brain to blood across the blood-brain barrier

    J. Neuroimmunol.

    (2001)
  • Y. Zhang et al.

    Global non-viral gene transfer to the primate brain following intravenous administration

    Mol. Ther.

    (2003)
  • Q.H. Zhou et al.

    Brain penetrating IgG-erythropoietin fusion protein is neuroprotective following intravenous treatment in Parkinson's disease in the mouse

    Brain Res.

    (2011)
  • U. Bickel et al.

    In vivo demonstration of subcellular localization of anti-transferrin receptor monoclonal antibody-colloidal gold conjugate within brain capillary endothelium

    J. Histochem. Cytochem.

    (1994)
  • R.S. Black et al.

    A single ascending dose study of bapineuzumab in patients with Alzheimer disease

    Alzheimer Dis. Assoc. Disord.

    (2010)
  • R.J. Boado et al.

    Comparison of blood-brain barrier transport of GDNF and an IgG-GDNF fusion protein in the Rhesus monkey

    Drug Metab. Dispos.

    (2009)
  • R.J. Boado et al.

    Genetic engineering of IgG-glucuronidase fusion proteins

    J. Drug Target.

    (2010)
  • R.J. Boado et al.

    Selective expression of the large neutral amino acid transporter (LAT) at the blood-brain barrier

    Proc. Natl. Acad. Sci. USA.

    (1999)
  • R.J. Boado et al.

    Humanization of anti-human insulin receptor antibody for drug targeting across the human blood-brain barrier

    Biotechnol. Bioeng.

    (2007)
  • R.J. Boado et al.

    Genetic engineering, expression, and activity of a fusion protein of a human neurotrophin and a molecular Trojan horse for delivery across the human blood-brain barrier

    Biotechnol. Bioeng.

    (2007)
  • R.J. Boado et al.

    Fusion antibody for Alzheimer's disease with bi-directional transport across the blood-brain barrier transport and Abeta fibril disaggregation

    Bioconjug. Chem.

    (2007)
  • R.J. Boado et al.

    GDNF fusion protein for targeted-drug delivery across the human blood brain barrier

    Biotechnol. Bioeng.

    (2008)
  • R.J. Boado et al.

    Genetic engineering of a lysosomal enzyme fusion protein for targeted delivery across the human blood-brain barrier

    Biotechnol. Bioeng.

    (2008)
  • R.J. Boado et al.

    Genetic engineering, expression and activity of a chimeric monoclonal antibody-avidin fusion protein for receptor-mediated delivery of biotinylated drugs in humans

    Bioconjug. Chem.

    (2008)
  • R.J. Boado et al.

    IgG-paraoxonase-1 fusion protein for targeted drug delivery across the human blood-brain barrier

    Mol. Pharm.

    (2008)
  • R.J. Boado et al.

    Engineering and expression of a chimeric transferrin receptor monoclonal antibody for blood-brain barrier delivery in the mouse

    Biotechnol. Bioeng.

    (2009)
  • Cited by (160)

    View all citing articles on Scopus
    View full text