Sie sind auf Seite 1von 26

ADR-12215; No of Pages 26

Advanced Drug Delivery Reviews xxx (2011) xxxxxx

Contents lists available at SciVerse ScienceDirect

Advanced Drug Delivery Reviews


journal homepage: www.elsevier.com/locate/addr

Modern methods for delivery of drugs across the bloodbrain barrier


Yan Chen a,, Lihong Liu b, 1
a b

School of Pharmacy, CHIRI, WABRI, Curtin University, GPO Box U1987, Perth, Western Australia 6845, Australia Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669, Singapore

a r t i c l e

i n f o

a b s t r a c t
The bloodbrain barrier (BBB) is a highly regulated and efcient barrier that provides a sanctuary to the brain. It is designed to regulate brain homeostasis and to permit selective transport of molecules that are essential for brain function. Unfortunately, drug transport to the brain is hampered by this almost impermeable, highly selective and well coordinated barrier. With progress in molecular biology, the BBB is better understood, particularly under different pathological conditions. This review will discuss the barrier issue from a biological and pathological perspective to provide a better insight to the challenges and opportunities associated with the BBB. Modern methods which can take advantage of these opportunities will be reviewed. Applications of nanotechnology in drug transport, receptor-mediated targeting and transport, and nally cell-mediated drug transport will also be covered in the review. The challenge of delivering an effective therapy to the brain is formidable; solutions will likely involve concerted multidisciplinary approaches that take into account BBB biology as well as the unique features associated with the pathological condition to be treated. Crown Copyright 2011 Published by Elsevier B.V. All rights reserved.

Article history: Received 6 August 2011 Accepted 21 November 2011 Available online xxxx Keywords: Bloodbrain barrier Drug delivery Receptor-mediated transport Cell-mediated transport Nanoparticles Liposomes Pathological conditions

Contents 1. 2. 3. 4. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . Physiology and biology of the bloodbrain barrier . . . . . . . Transport routes across the bloodbrain barrier . . . . . . . . Biological and pathological properties of BBB for drug transport . 4.1. Physical barrier . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 0 0 0 0

Abbreviations: a2M, alpha-2 macroglobulin; A, amyloid ; ABC, ATP binding cassette; AD, Alzheimer's disease; AIDS, autoimmunodeciency syndrome; AJ, adherens junction; AMT, adsorptive-mediated transport; AMP, adenosine monophosphate; ANG1005, angiopep 2 conjugated with 3 molecules of paclitaxel; Antp, Antennapedia; APP, amyloid beta precursor protein; ApoE, Apolipoprotein E; ATP, adenosine triphosphate; AUC, area under curve; BBB, bloodbrain barrier; BCSFB, bloodcerebrospinal uid barrier; BSA-NP, bovine serum albumin conjugated nanoparticles; cAMP, cyclic AMP; CBSA, cationic bovine serum albumin; CBSA-NP, CBSA conjugated PEG-PLA nanoparticles; CED, convection enhanced diffusion; CHP, hydrophobic cholesterol groups; CMC, critical micelle concentration; CMT, carrier-mediated transport; CNS, central nervous system; CPP, cell penetrating peptide; CRM, cross reacting material; CSF, cerebrospinal uid; DT, diphtheria toxin; DTR, diphtheria toxin receptor; EAE, experimental autoimmune encephalomyelitis; EO, ethylene oxide; EC, endothelial cell; EMF, electromagnetic elds; FBP, fusion sequence-based peptide; g7, similopioid peptide; GMP, guanosine monophosphate; HB-EGF, heparin binding epidermal growth factor; HIRMAb, human insulin receptor monoclonal antibody; HIV, human immunodeciency virus; HLB, hydrophobichydrophilic balance; HSA, human serum albumin; HSP-96, heat shock protein 96; HUVEC, human umbilical vein endothelial cells; ICH, intercerebral haemorrhage; ICV, intracerebroventricular; IgG, immunoglobulin G; IL, interleukin; INF, interferon; JAM, junction adhesion molecules; LDL, low density lipoprotein; LDLR, low density lipoprotein receptor; Lf, lactoferrin; LMV, large multilamellar vesicles; LPA, lysophosphatidic acid; LRP, lipoprotein receptor protein; LUV, large unilamellar vesicles; MAP, model amphipathic peptide; MAPK, mitogen activated protein kinase; MCP, monocyte chemotactic protein; MHC, major histocompatibility complex; MLCK, myosin light chain kinase; MP, mononuclear phagocytes; MRP, multidrug resistant protein; MS, multiple sclerosis; NOS, nitric oxide syntheses; NP, nanoparticles; NVU, neurovascular unit; P97, melanotransferrin; PAI-1, plasminogen activator inhibitor 1; PHDCA, poly(hexadecylcyanoacrylate); PBCA, poly(butylcyanoacrylate); PEG, polyethylene glycol; PEG-PCL, PEG-polycaprolactone; PEG-G-CSF, PEGylated-recombinant methionyl human granulocyted colony stimulating factor; PEG-PLA, polyethylene glycol-polylactic acid; P-gp, P-glycoprotein; PKA, protein kinase A; PKC, protein kinase C; PKG, protein kinase G; PLGA, poly(D,L-lactide-co-glycolide); PO, propylene oxide; PTD, protein transduction domain; PTK, protein tyrosine kinase; Qdots, quantum dots; RAP, receptor associated protein; RES, reticuloendothelial system; REV, reverse phase evaporation vesicles; RMT, receptor-mediated transport; R123, rhodamine 123; SA, sialic acid residue; SBP, sequence signal-based peptide; SUV, small unilamellar vesicles; TAT, HIV-1 trans-activating transcriptor; TEM, transmission electron microscopy; TER, transendothelial electrical resistance; TfR, transferrin receptor; TJ, tight junction; TNF, tumour necrosis factors; tPA, tissue plasminogen activator; VE, vascular endothelial; VEGF, vascular endothelial growth factor; ZO, zonula occludens. This review is part of the Advanced Drug Delivery Reviews theme issue on Delivery of Therapeutics to the Central Nervous System. Corresponding author at: School of Pharmacy, Curtin University, GPO Box U1987, Perth, Western Australia 6845, Australia. Tel.: +61 8 9266 2738; fax. +61 89266 2769. E-mail address: y.chen@curtin.edu.au (Y. Chen). 1 L Liu is currently funded as an Australian Postdoctoral Fellow by ARC Discovery Project DP110104599 at Chemical Engineering, Curtin University. 0169-409X/$ see front matter. Crown Copyright 2011 Published by Elsevier B.V. All rights reserved. doi:10.1016/j.addr.2011.11.010

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

4.1.1. Tight junctions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1.2. Adherens junctions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1.3. Tight junctions regulation and signalling pathways . . . . . . . . . . . . . . . . . . . . . . . . 4.2. Impact of pathological conditions on the properties of the bloodbrain barrier . . . . . . . . . . . . . . . . 4.2.1. Changes in permeability of the bloodbrain barrier and drug transport under pathological conditions . 4.2.2. Changes in the bloodbrain barrier transport systems under pathological conditions . . . . . . . . . 4.2.3. Monocyte and macrophage trafcking across the bloodbrain barrier . . . . . . . . . . . . . . . . 5. Modern methods of transporting drugs across the bloodbrain barrier . . . . . . . . . . . . . . . . . . . . . . . 5.1. Drug transport across the bloodbrain barrier via tight junction opening. . . . . . . . . . . . . . . . . . . 5.1.1. Enhanced by biological stimuli . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1.2. Enhanced by chemical stimuli . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1.3. Enhanced by physical stimuli. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2. Transport system-mediated drug delivery across the bloodbrain barrier . . . . . . . . . . . . . . . . . . 5.2.1. Nanocarriers for brain drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2.2. Supramolecular architectures of aggregated amphiphilic molecules . . . . . . . . . . . . . . . . . 5.2.3. PEGylation of nanocarriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2.4. Functionalization of nanocarriers for drug transport across the bloodbrain barrier . . . . . . . . . 5.3. Drug transport across the bloodbrain barrier via transport vectors . . . . . . . . . . . . . . . . . . . . . . 5.4. Drug transport across the bloodbrain barrier via adsorptive-mediated transcytosis . . . . . . . . . . . . . 5.5. Drug transport across the bloodbrain barrier via endogenous receptor-mediated transcytosis . . . . . . . . 5.5.1. Insulin receptor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.5.2. Transferrin receptor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.5.3. Low-density lipoprotein receptor related proteins 1 and 2 (LRP1 and LRP2 receptors) . . . . . . . . 5.5.4. Diphtheria toxin receptor (DTR) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.6. Drug transport across the bloodbrain barrier via inhibition of efux Pumps . . . . . . . . . . . . . . . . . 6. Cell-mediated drug transport across the bloodbrain barrier . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7. Conclusions and remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . .

0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

1. Introduction The bloodbrain barrier (BBB) is a dynamic barrier protecting the brain against invading organisms and unwanted substances. It is also the most important barrier impeding drug transport into the brain via the blood circulation. Despite the rapid development in our understanding of the molecular structure of components of the BBB, our knowledge in receptor expression at the BBB, advances in medical technology, and breakthroughs in nanotechnology-based approaches, many of the brain or central nervous system (CNS) associated diseases remain under-treated by effective therapies. This is not because there is a lack of candidate drugs but due to the inability of many therapeutic molecules to cross the BBB, the bloodcerebrospinal uid barrier (BCSFB), or other specialised CNS barriers to reach the specic areas of brain [1]. Such a difculty in delivering therapeutic molecules to the brain or CNS can only be overcome by a concerted effort in understanding the physiology of BBB, its permeability under different pathological or disease conditions, and its response to physical and chemical stimuli, as well as the various transport receptors at the BBB and available delivery technologies. As many attempts to transport drugs across the BBB could be against the natural function of the BBB, effective approaches or methods should be cautiously assessed with regards to their impact on the overall protective function of BBB. The purpose of this review is to explore molecular and biological opportunities at the BBB for transport of therapeutic molecules across BBB under physiologic and pathological conditions. In addition, any changes in BBB that can be utilised, when exposed to physical and chemical stimuli, are analysed for opportunities to maximise drug delivery. In this review, we also highlight the modern approaches and present insights into using ligand-conjugation and nanotechnology to target the BBB via adsorptive or receptor-mediated transport of drug molecules into the brain. Particular attention has also been paid to the cellmediated approach which takes advantage of the immunological surveillance system of the brain, using circulating phagocytic cells

such as monocytes or macrophages as Trojan horse to deliver drug molecules into the brain. 2. Physiology and biology of the bloodbrain barrier The brain is well protected and dynamically regulated to provide a sanctuary for the central nervous system (CNS). There are several gateways to enter brain parenchyma, the most important two are blood circulation and cerebrospinal uid (CSF) circulation. In the human brain, there are about 100 billion capillaries in total, providing a combined length of brain capillary endothelium of approximately 650 km and a total surface area of approximately 20 m 2 [2]. Any molecules' entry into the brain via parenteral administration is strictly controlled by the BBB and the BCSFB. As the surface of BCSFB faces the ventricle that is lled with CSF, not the blood [3], this, in combination with the high turnover rate of CSF, leads to continuously ushing the injected drug (i.e. those injected into the ventricle) back to the blood [4]. The BBB, therefore, is universally considered as the most important barrier in preventing molecules from reaching the brain parenchyma via extensive branches of blood capillary networks. The chief anatomical and functional site of the BBB is the brain endothelium. Physiologically, in addition to brain capillary endothelial cells, extracellular base membrane, adjoining pericytes, astrocytes, and microglia are all integral parts of the BBB supporting system. Together with surrounding neurons, these components form a complex and functional neurovascular unit [5] (Fig. 1). A feature of the BBB is its low and selective permeability to molecules which can be attributed to its unique biological characteristics. These include: 1) the lack of fenestrationsand with very few pinocytotic vesicles, but a greater number and volume of mitochondria in endothelial cells [68]; 2) the presence of tight junctions (TJ) between adjacent endothelial cells, formed by an intricate complex of transmembrane proteins

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

Fig. 1. Schematic representation of the bloodbrain barrier (BBB) and other components of a neurovascular unit (NVU). Reproduced with permission from reference [11].

(junctional adhesion molecule-1, occludin, and claudins) with cytoplasmic accessory proteins (zonula occludens-1 and -2, cingulin, AF-6, and 7H6). They are linked to the actin cytoskeleton [9], thereby forming the most intimate cell to cell connection. The TJ are further strengthened and maintained by the interaction or communication of astrocytes and pericytes with brain endothelia cells [10]; 3) the expression of various transporters including GLUT1 glucose carrier, amino acid carrier LAT1, transferring receptors, insulin receptors, lipoprotein receptors and ATP family of efux transporters such as p-glycoprotein (P-gp) and multidrug resistancerelated proteins MRPs [3,11]. Some of these aid the transport into the brain while others prevent the entry of many molecules; 4) the synergistic inductive functions and upregulating of BBB features by astrocytes, astrocytic perivascular endfeet, pericytes, perivascular macrophages and neurons, as suggested by the strong evidence from cell culture studies [1214]; 5) the lack of lymphatic drainage, and absence of major histocompatibility complex (MHC) antigens in CNS with immune reactivity inducible on temporary demand in order to provide maximum protection to neuronal function [15]. The BBB has a strict limit for the passage of immune cells, especially lymphocytes [16] and its immune barrier is made by the association between BBB endothelia cells and perivascular macrophages and mast cells [17]. Additionally, this immune barrier is reinforced by local microglial cells [18]. All these characteristics lead to BBB to possess multiple functions as a physical barrier (TJ), a transport barrier (P-gp), a metabolic or enzymatic barrier (specialised enzyme systems [11,19] and an immunological barrier. 3. Transport routes across the bloodbrain barrier It has been well established that there are several transport routes by which solute molecules move across the BBB [11,20]. Diffusion of substances into the brain can be divided into paracellular and transcellular. As illustrated in Fig. 2.a, small water-soluble molecules simply diffuse through the TJ but not to any great extent. Small lipid soluble substances like alcohol and steroid hormones penetrate transcellularly by dissolving in their lipid plasma membrane (Fig. 2.b).

However, for almost all other substances, including essential materials such as glucose and amino acids, transport proteins (carriers), specic receptor-mediated or vesicular mechanisms (adsorptive transcytosis) are required to pass the BBB. In the case of transport proteins or known as carrier-mediated transport (Fig. 2.c), there is binding of a solute such as glucose or amino acids to a protein transporter on one side of the membrane that triggers a conformational change in the protein, resulting in the transport of the substance to the other side of the membrane, from high to low concentration. If compounds need to be moved against a concentration gradient, ATP may provide the energy to facilitate the process. Efux pumps or transporters (Fig. 2.d) are responsible for extruding drugs from the brain and this mechanism is a major obstacle for the accumulation of a wide range of biologically active molecules in the brain, with the ATP binding cassette (ABC) transporter P-gp and multidrug resistant protein (MRP) being the principle efux mechanism of these agents [21]. Inhibition of P-gp in pre-clinical studies has enhanced the penetration of paclitaxel into the brain, indicating the feasibility of achieving improved drug delivery to the brain by suppression of P-gp [22]. Receptor-mediated transcytosis (RMT) (Fig. 2.e) provides a means for selective uptake of macromolecules. Endothelial cells have receptors for the uptake of many different types of ligands, including growth factors, enzymes and plasma proteins. For example, insulin molecules rst bind to receptors that collect in specialized areas of the plasma membrane known as coated pits. When bound to ligand these pits invaginate into the cytoplasm and then pinch free of the plasma membrane to form coated vesicles. After acidication of the endosome, the ligand will dissociate from the receptor and cross the other side of membrane. RMT has been extensively studied for brain targeting [23]. Those well-characterised systems include transferring receptor (TfR), insulin receptor, lipoprotein receptors, scavenger receptors class B type I, diphtheria toxin receptor and glutathione transporter [3]. Adsorptive-mediated transcytosis (AMT), also known as the pinocytosis route (Fig. 2.f), is triggered by an electrostatic interaction between a positively charged substance, usually the charged moiety of a peptide, and the negatively charged plasma membrane surface (i.e. heparin sulphate proteoglycans). Adsorptive-mediated transport has a lower afnity but higher capacity than RMT. The development of many new drug delivery technologies focuses on AMT [24]. AMT-

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

Fig. 2. Transport routes across the bloodbrain barrier. Pathways a to f are commonly for solute molecules; and the route g involves monocytes, macrophages and other immune cells and can be used for any drugs or drugs incorporated liposomes or nanoparticles. Adapted from reference [11].

based drug delivery typically involves either cationic proteins or cellpenetrating peptide such as Tat-derived peptides and Syn-B vectors. Last, but not least, cell-mediated transcytosis (Fig. 2.g) is a more recently identied route of drug transport across the BBB [25], although it is a well established mechanism for some pathogens such as Cryptococcus neoformans and HIV entry into the brain, known as Trojan horse model [26,27]. This transport route relies on immune cells such as monocytes or macrophages to cross the intact BBB. Unlike aforementioned transport pathways which normally permit only solute molecules with specic properties, cell-mediated transcytosis is unique in that it can be used virtually for any type of molecules or materials as well as particulate carrier systems [28]. Due to the unique properties of the TJs, paracellular transport of hydrophilic drugs is virtually absent and transcellular transport by passive diffusion is only available to molecules which full certain criteria [4,29,30] such as: 1) molecular weight is less than 500 Da; 2) compounds are unionised; 3) log P value of the drug is close to 2; 4) cumulative number of hydrogen bonds is not more than 10. Unfortunately only a small percentage of drugs t these criteria [2]. For other therapeutic molecules, their transport across the BBB will then have to rely on either the integrity of the BBB or the drug or drug carrier properties and their interaction with or afnity for receptors expressed at the BBB, as well as other biological or immunological processes occurring at the BBB. In other words, the BBB properties and related biological processes, and their roles in trafcking various types of molecules are fundamental to the success of drug transport across the BBB. This is the reason for the need to gain a thorough understanding of the biological and pathological properties and processes of the BBB. 4. Biological and pathological properties of BBB for drug transport Recent progress in the study of the molecular biology of the BBB has led to a greater understanding of the barrier functions under normal physiological and pathological conditions, as well as when the BBB is subjected to external stimuli. More importantly, this knowledge empowers researchers to develop new strategies for therapeutic molecules to target and transport across the BBB for treatment of

various CNS associated diseases. This section is focused on the physical barrier and properties of the BBB undergoing pathological changes which may present potential opportunities for drug transport. 4.1. Physical barrier The physical barrier of the BBB is a result of the formation of an elaborated junctional complex by TJ and adherens junctions (AJ) between adjacent endothelial cells [31]. 4.1.1. Tight junctions TJ are located on the apical region of endothelia cells and structurally formed by an intricate complex network made of a series of parallel, interconnected, transmembrane and cytoplasmatic strands of proteins [32,31]. The high level of integrity of TJ is reected by the high electrical resistance of the BBB (15002000 cm 2), which depends on a proper extracellular Ca 2 + ion concentration. There are extensive reviews on the TJ elsewhere [3133]. Here the focus of this review is placed on some key molecules involved in the formation and maintenance of TJ and the regulation of the permeability of TJ. Among the identied molecules associated with TJ, the transmembrane proteins claudins and occludin are most well studied. Claudins form dimmers and bind homotypically to other claudin molecules in an adjacent brain capillary endothelia cell [34,35] thus forming the primary seal of the TJ [31]. On the other hand, occludin is not essential for the formation of TJ, as indicted in the knockout and knockdown experiments [9] and its main function appears to be for TJ regulation and as an additional support structure [10,36]. Of claudins, Claudin-5 has been shown to be involved in size-selective loosening the permeability of BBB in mice [37] with permeability of molecules of size less than 800 Da affected. However, similar effects were not observed with barrier function of non-BBB endothelium, such as human umbilical vein endothelial cells (HUVEC) [38]. In another experiment, treatment of claudin-5 by cyclic AMP (cAMP) lead to enhancement of claudin-5 activity along cell borders, rapid reduction in transendothelial electrical resistance (TER), and loosening of the claudin-5-based endothelial barrier against mannitol, but not inulin [39]. These suggest that manipulation of claudin-5, or potentially other TJ proteins

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

may permit drug transport by altering the molecular sieve function at the BBB but without its total disruption [40]. Junctional adhesion molecules JAM-A, JAM-B, and JAM-C, which are present in brain endothelial cells, also take part in the formation and maintenance of the TJ [11] , together with occludin and claudins. Although functions of JAM at the BBB are still largely unknown, it has been suggested that their involvement is not limited only to the junctional tightness, they may also associate with leukocyte trafcking, with implications for immune activity in CNS diseases [41]. Submembranous TJ-associated proteins, also known as peripheral zonula occludens proteins, such as ZO proteins ( ZO-1, ZO-2 and ZO-3) are another integral part of TJ with possible functions of forming a scaffold to link TJ to the cytoskeleton [31]. Certainly there is no TJ without ZO-1 as its molecules are delimiting the interendothelial cleft and connecting transmembranous TJ proteins with the actin cytoskeleton [42]. A recent study in multiple sclerosis (MS) model experimental autoimmune encephalomyelitis (EAE) suggests that the relocalization of ZO-1 from cell boundary to cytosol directly increased permeability of the endothelial monolayer, leading to disruption of the BBB and development of clinical diseases such as MS [43]. Other cytoplasmic proteins such as cingulin (140 kDa) and 7H6 phosphoprotein (155 kDa) have also been implicated in playing a role in regulating the permeability of TJ [32]. 7H6 phosphoprotein has been associated with TJ impermeability to ions and large molecules and evidence suggests that the potential detachment of 7H6 protein from TJ when ATP levels reduce can result in increased paracellular permeability of the BBB [33]. 4.1.2. Adherens junctions Adherens junctions (AJ) are located below the TJ in the basal region of the lateral plasma membrane. They are composed of transmembrane glycoproteins (represented by the large family of cadherins) linked to the cytoskeleton by cytoplasmatic proteins, thus providing additional tightening structure between the adjacent endothelial cells at the BBB [31]. In addition to supporting the barrier function, AJ mediate the adhesion of brain endothelial cells to each other, the initiation of cell polarity and the regulation of paracellular permeability [9]. Cadherins' homotypical interaction with each other takes place when Ca2 + is present. Cadherin-5, also known as VE-cadherin (vascular endothelial cadherin), is an important determinant of microvascular integrity. When VEcadherin is over expressed, Ca2 +-dependent cell adhesion, inhibition of cell proliferation and reduction in cell permeability and migration are observed [44]. In order for cadherins to work as adhesion molecules, catenin proteins have to play the role of anchor molecules, linking cadherin complex to the actin cytoskeleton [33]. One of the catenins, -catenin, is a structural protein and also serves as a mediator in regulation of P-gp and other multidrug efux transporters in brain vasculature [45]. It has also been suggested that its up-regulation is important for the maintenance of TJ protein assembly and barrier function [46]. 4.1.3. Tight junctions regulation and signalling pathways TJ are highly dynamic structures that undergo changes (breakdown or reassemble) in response to physiological and pathological conditions, which provides the opportunities for reversible opening of the membranous barrier, via the use of TJ modulators, to improve drug delivery across the BBB [47]. Intensive research has been conducted to understand the relationship between TJ function or disruption and the state of TJ protein phosphorylation or dephosphorylation [48]. Physiologically the opening and closing of the BBB paracellular pathway is controlled by the dynamic interaction among TJ and AJ structure elements. It has been suggested that changes in adhesive properties of TJ and AJ proteins and reorganization of the actin cytoskeleton would result in the formation of intercellular gaps at the BBB [48]. A correlation between the changes in the adhesive property of TJ and AJ, and alterations in their phosphorylation status has been

established [31,48]. Phosphorylation, sometimes dephosphorylation may occur to the TJ proteins as they are phosphoproteins, depending on the type of stimuli and local microenvironment. For instance, vascular endothelial growth factor (VEGF) induces Ser/Thr phosphorylation, causing redistribution of occludin and ZO-1 in murine brain endothelial cells [49], whereas, in conditions like calcium depletion or bacterial infection, TJ proteins (occludin) would undergo dephosphorylation rather than additional phosphorylation during TJ disruption [50]. Similarly, AJ proteins such as VE-cadherin and -catenin undergo phosphorylation of Ser/Thr and Tyr residues, which is correlated to opening of the BBB [51]. Such changes in phosphorylation state of TJ and AJ proteins weaken their interaction, alter transmembrane protein localization and induce their redistribution, eventually causing the dissociation of junction complex from its cytoskeleton anchor and leading to the increased permeability of the BBB [52,53]. In addition to TJ and AJ proteins, mediators of signalling pathways, such as protein kinase C (PKC), protein tyrosine kinases (PTKs), nitric oxide syntheses (NOS), protein kinase A and G (PKA, PKG), myosin light chain kinase (MLCK) and mitogen activated protein kinase (MAPK), are also involved in regulation of TJ and TJ-mediated BBB permeability [31]. They could potentially be exploited for drug delivery. The mechanisms of their function in altering the permeability of the BBB can be complex and sometimes manifold. For instance, activation of PKC may stimulate phosphorylation of TJ proteinsoccludin or directly stimulate the cytoskeletal contractile machinery by phosphorylating MLCK [54], leading to an increase in the BBB paracellular permeability. Such activation of PKC may take place in a wide range of pathological and physiological conditions, such as removal of extracellular Ca 2 +, elevation of intracellular Ca 2 +, and the presence of Ca 2 + chelators, oxidative stress, inammatory mediators (e.g. tumour necrosis factors-: TNF-), vasogenic agent (e.g. VEGF) and human immunodeciency virus 120 kDa ligase (HIV gp 120) [49,55]. Their action on PKC leads to opening the TJ. Likewise, enhanced PTK's activities have also been associated with increased endothelial permeability under several pathological conditions [58]. Similar to PKC and PTK, activation of MLCK (e.g. by vasoactive mediator histamine and lysophosphatidic acid: LPA) leads to weakening of the integrity of TJ [56]. Bacteria or viral pathogens, cytokines and even bile acids are some examples of pathological and physiological agents that can trigger TJ opening via MLCK [55]. In contrast, pathological conditions such as hypoxia, ischemia or excessive nitric oxide (NO) release can decrease BBB paracellular barrier function via activation of PKG, which results in soluble guanylate cyclase activation and elevation of cyclic GMP [57]. Although many signalling pathways participate in TJ opening, there are exceptions. For example, upon elevation of intracellular cAMP, PKA can be activated and it stabilises endothelial cytoskeletal and adhesive structures, strengthens cell-matrix adhesion and inhibits leukocyte adhesion, which eventually results in increased transendothelial electric resistance (TEER) and decreased TJ permeability [57,58]. There are numerous other signalling pathways and modulators which can act directly on the TJ components, modifying permeability of TJ. More comprehensive reviews on this topic can be found elsewhere [47,31]. To date almost all of the abovementioned studies were carried out in vitro on brain capillary endothelia cells. Although some of these processes can be expected to be more complex in vivo, these studies are important in understanding the development and progression of many CNS diseases, their pathological conditions with drug transport opportunities and responses of the BBB to chemical, physical and biological stimuli. Knowledge of the molecular features of tight junctions regulation and signalling pathways gained from in vitro studies has been translated into in vivo studies. For instance, in vivo studies have been used to conrm the location of tight junction-specic protein ZO-1 in the BBB of rats and humans [59] and to study the underlying mechanism

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

of drug action in the brain [60]. In addition, in vivo studies provided direct insights into the relationship between pathological conditions and tight junctions regulation or change of signalling pathways [34,61]. Willis et al. [62] revealed the relationship between transitory astrocyte loss and the disruption of tight junction complexes in the rat BBB in an in vivo study using a gliotoxin and three vascular tight junction markers, providing a better understanding of tight junctions regulation and the integrity of the BBB. 4.2. Impact of pathological conditions on the properties of the bloodbrain barrier The properties of the BBB undergo signicant changes when the brain is developing a neurological disorder, in inammatory conditions or under attack by pathogens. These changes affect the integrity as well as the functions of the BBB, including transport pathways. In vitro studies have shown that altered drug delivery under pathological conditions may be mediated through changes in a number of transport pathways including paracellular transport and transcellular transport (RMT and adsorptive transcytosis) [3]. Indeed, it has been anticipated that the impaired BBB may provide a window of opportunity for those drugs which normally are unable to traverse the BBB to reach the target in the diseased brain. In Table 1, we highlight some of the examples and the implications of pathological conditions on the drug delivery opportunities into the brain. 4.2.1. Changes in permeability of the bloodbrain barrier and drug transport under pathological conditions Analysing the functions of TJ structural components, signalling pathways and regulation of TJ under different conditions, a large body of evidence suggests that the permeability of the BBB is strongly inuenced by the stimuli produced by physiological and pathological conditions such as oxidative stress (e.g. free radical nitric oxide:NO; peroxide:H2O2), inammatory mediators (e.g. interleukin-1, IL-1; interferon-, INF-; TNF-), lipid mediators (prostaglandin E2 and F2a), vasogenic agents (e.g. histamine; VEGF), infective agents (e.g. bacteria and bacteria toxin; viruses and virus components; parasites

and fungal pathogens), as well as physiological and immunological stimuli (e.g. intracellular Ca 2 + and leukocytes, respectively) [47,48]. There is growing evidence that the BBB's integrity is compromised in brain disorders or diseases such as stroke, Alzheimer's disease (AD), MS, HIV, Parkinson's, ischemia and brain tumours [11,34,63]. This often leads to disordering, impairment or even breakdown of the BBB. Often the changes in brain endothelia phenotype, junctional complex remodelling and a progressive increase in leukocyte inltration [64] are observed with the breakdown of the BBB. In patients suffering from AD, the level of BBB impairment is closely correlated to increased rates of neurodegeneration. It was reported by Bowman et al. [65] that BBB disruption, due to amyloid angiopathy and possible hyperhomocysteinemia, was persistently presented in a subgroup of patients with AD, which permitted peripheral immunoglobulin G (IgG) to have greater access to the CNS peripherally. However, the researchers did point out that although the BBB impairment may benet the delivery of drug treatment, the increase of the BBB impairment could lead to increased rates of neurodegeneration. In other words, in the case of AD, restoring BBB integrity maybe more important than taking advantage of the impaired BBB for drug transport. In brain disorders such as Parkinson's disease and epileptic seizures, transient opening of the BBB occurs due to the transient secretion of inammatory mediators [66,67]. On the other hand, both primary and secondary brain tumours can increase BBB permeability, likely caused by disturbance of TJ complex and/or accumulation of growth factors such as VEGF and proinammatory cytokines [68]. Analysing all neuropathological conditions reveals one common factor: inammation and inammatory mediators are involved in BBB disruption. It has been suggested that the process of inammation and inammatory mediators can be targeted or controlled to open or close the BBB [48]. It is generally anticipated that BBB interruption that occurs under various pathological conditions may provide an opportunity for enhancement of drug transport into the brain via the paracellular route. This, however, has proved to be a more complex issue. Due to limited in vivo results obtained by different investigators under pathological conditions, the answer is not clear. For instance, Fang and colleagues recently showed that the enhanced BBB permeability

Table 1 Pathological conditions, their impact on the bloodbrain barrier, and drug transport opportunities into the brain. Pathological condition Multiple sclerosis Impact on the BBB Disruption of TJ; enhanced leukocyte activity; release of inammatory cytokines/chemokines BBB disruption and permitted the greater access of peripheral IgG to the CNS Overexpression of efux pumps BBB disruption Increase in the diameter of cortical vessels, thinning of basal lamina, loss of glycoproteins, apoptosis of endothelial cells and tight junction disruption Leukocyte invasion, elevated CSF-to-serum albumin ratio, and BBB impairment Increased BBB permeability BBB disruption Upregulation of DTR Trauma Pain BBB breakdown Alternation of BBB chemokine receptor due to activated astrocytes Decreased TJ proteins and BBB perturbation Loss of the tight junctions in the tumour vascular system, enhanced retention effect Overexpression receptors of folate, insulin and transferrin Upregulation of DTR Implication for drug traversing the BBB Potentially it may enhance paracellular transport of drugs. References [291] [43] [63] [65] [292] [293] [294]

Alzheimer's disease

Parkinson's disease HIV

Potentially it may enhance paracellular transport of drugs that have afnity for albumin and IgG into the CNS Efux pump inhibitors may improve drug deliver into the brain. It enhanced therapeutic agent concentration in the brain. Potentially it may increase drug transport into the brain due to the leaky barrier. It may enhance paracellular transport of drugs and drugs with afnity for albumin. It may facilitate paracellular drug transportation. It enhanced paracellular drug, e.g. Ginkgolide B, passage into the brain It may provide disease-induced specic drug targeting of the BBB and receptor mediated transcytosis. It enhanced therapeutic agent concentration in brain It may lead to astrocyte-targeted therapy. It may facilitate paracellular drug transportation. Angiogenic vessels are permeable to nano-sized materials. It enhanced folic acid, insulin and transferrin-attached nanoparticles across the BBB. Potentially it may increase disease-induced specic drug targeting of BBB and receptor mediated transcytosis.

Infectious disease Inammation Stroke

[295,296] [297] [69] [102] [30] [298] [10] [299] [300] [301,302] [303]

Brain tumour

Ischemia/seizures

DTR: diphtheria toxin receptor.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

associated with stoke or ischemia/reperfusion injury facilitated a significant increase in penetration of Ginkgolide B, a potential neuroprotective agent, through the BBB in a ischemia/reperfusion injury rat model compared to that in normal rats [69]. However, in a separate study using MS, AD and, AD related animal models (experimental autoimmune encephalomyelitis, EAE, induced mice for MS, streptozotocininduced mice and TASTPM transgenic mice for AD) showed a signicantly enhanced brain penetration rate in EAE mice compared to nave mice 10 min after intravenous bolus injection for both paracellular markershydrophilic molecule sodium uorescein (NaF) and atenolol [70]. At 2.5 h post intravenous infusion, concentration of atenolol was found to be almost identical in EAE and nave mice and there was no data reported for other time points post intravenous injection and for NaF at 2.5 h post intravenous infusion in the study. Data presented on NaF in nave and TASTPM mice also showed no differences at 0.5 h post injection. Researchers of the study concluded that despite reported BBB disruption, CNS penetration for small molecule therapeutic agents does not increase in MS- and AD-related animal models. It is worth noting that the study, however, did not characterise the level of BBB disruption in these diseased mice and the comparison data of paracellular markers was collected, in most cases, from a single time point from a different types of administration (bolus injection versus infusion). It is, therefore, difcult to ascertain the true level of the BBB penetration rate and extent of paracellular molecules given as intravenous bolus injection in AD and MS conditions. 4.2.2. Changes in the bloodbrain barrier transport systems under pathological conditions 4.2.2.1. P-glycoprotein transporters. P-gp is one of the most important transport systems as it plays a crucial role in preventing the passage of drugs and toxins across the BBB as well as facilitating their transport from brain to blood [71]. P-gp expression changes as a result of pathogenesis, a phenomenon that is associated with a number of neurological disorders [10]. An inverse correlation was found between P-gp expression and deposition of -amyloid in AD [72]. Likewise, it was found that there was a decrease in P-gp functional activity in the brain tissue collected from patients with Parkinson's disease [73]. However, in some diseases, P-gp is reported to be upregulated. For instance, the overexpression of P-gp was described in epilepsy and P-gp was found with both endothelia cells and perivascular astrocytes, which was suspected to have contributed to the multiple drug resistance of the treatment [74]. A similar upgrading of P-gp was also found at the BBB after focal cerebral ischemia [75]. Sometimes active efux transport by P-gp can be the major obstacle for delivering a new drug therapy, therefore, to facilitate the treatment, either P-gp inhibitor should be considered or the drug has to be chemically modied or formulated in such a way that it will not be the substrate of P-gp. One of the success stories of an approach with P-gp inhibitors can be highlighted by the research using Pluronic block copolymers as an inhibitor of drug efux transporters expressed at BBB, for drug delivery into the brain. When co-administrated with drugs, Pluronic P85 increased the transport of the drugs that are substrates of these efux transporters into the brain [76]. It has been pointed out that the P-gp inhibitor approach is better suited to improve the delivery of the treatment to acute diseases such as brain tumour when the aim of the delivery is to maximise the drug concentration for a relatively short duration. The prolonged inhibition of P-gp, in the case of chronic administration, may lead to the interference of physiological homeostatic regulation provided by P-gp [77]. Furthermore, blockade of P-gp in the BBB by application of P-gp inhibitor can lead to a significant increase of brain concentrations of various drugs. This is due to P-gp's function as efux transporters in the BBB, which if blocked may cause dramatic toxicity of drugs in brain so that doses of drugs that are normally well tolerated may become neurotoxic [78]. Given the

protective role of P-gp in various cells and organs, the prolonged application of P-gp inhibitors, especially if they are poorly selective, also poses the risk of systemic toxicity caused by the reduction in elimination of drugs [79]. Thus the strategy of using P-gp inhibitors to enhance drug transport into the CNS should be applied with caution and restricted to transient application. 4.2.2.2. Adsorptive-mediated transcytosis (AMT). Changes in AMT (pinocytosis) and RMT in diseased states were also reported in cell studies and animal models. These changes will not only affect the process itself but also affect the drug delivery strategy. An in vitro study with brain endothelia cells showed an increase in AMT after treatment with proinammatory cytokines TNF- and interleukin-6 (IL-6), which are important cytokines presented in several neurological diseases including MS and AIDS [80]. Increased endocytosis and immunological responses were reported in rats after head injury [81]. In another study, Fillebeen and colleagues demonstrated a marked increase of transcytosis of lactoferrin (Lf), an iron-binding protein against infection and severe inammation, at the BBB in vitro in the presence of TNF- [82]. It was concluded that such enhancement of transcytosis did not involve the up-regulation of the Lf receptor but rather an increase in the rate of transcytosis transport. Furthermore, enhanced endocytotic activity has also been reported in a rat ischemia/stroke model in which transient focal ischemia was induced using the lament occlusion of middle cerebral arteries [83]. Although this enhanced endocytosis or transcytosis mechanism is yet to be conrmed in human with CNS disorders, it is hypothesized that the same may be expected given that the same inammatory mediators or responses are associated with the CNS diseases in human. Thus, this enhanced endocytosis activity associated with diseased brain supports the rationale for designing drug delivery systems with cationic proteins or peptides (e.g. cell-penetrating peptides) as a means to maximise the drug transport into the diseased brain. 4.2.2.3. Receptor-mediated transcytosis (RMT). RMT is an important transport pathway for endogenous peptides such as insulin, insulinlike growth factor and transferrin [84]. It is highly specic and it uptakes macromolecules presented on the luminal side of the brain endothelia cells and delivers them to the brain with the receptor recycled back to the luminal membrane [29]. Although more and more receptors have continuously been discovered, differential expression, distribution and regulation of various receptors at BBB in the diseased brain is not fully understood or characterised. This is an issue that requires urgent attention as it has a profound inuence on the effectiveness of the targeting strategy of many emerging drug delivery systems, such as the molecular Trojan horse approach. Currently there is only limited knowledge of how different CNS disorders impact on the expression of receptors and their regulation despite this being an important factor which must be considered in the early stage of designing a BBB targeting approach for a particular CNS disorder. Here the review will focus on some of the most commonly known receptors involved in RMT. 1. TfR. TfR mediates the transcytosis of transferrin-bound iron through the brain capillary endothelial cell in humans [85]. It is the most well characterised receptor. As there is often an association between iron accumulation and cellular damage, iron has long been considered to play an important role in exacerbating the brain tissue degradation process in many neurodegenerative disorders. There is an increased expression of TfR in brain ischemia by brain capillary endothelia cells [86]. However, a decreased expression of TfR was observed in the hippocampus of humans with AD compared to that of normal human [87], which supports an early nding by Kalaria et al. [88] that TfR density was decreased in some cortical areas including the hippocampus in AD

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

but relatively unchanged in cerebral microvessels. In contrast, evidence indicates that there is a marked increase in the brain TfR level during brain injury after intracerebral haemorrhage (ICH) [89]. Recht et al. reported that an immunohistochemistry study carried out on normal human brain-tissue and brain tumour biopsy specimens from patients revealed TfR primarily in brain endothelia cells with a differential immunostaining for TfR between normal and neoplastic tissue, suggesting TfR could be exploited as a target for delivering drug across the BBB for brain tumour treatment [90]. 2. Insulin receptor: This is the receptor that plays a crucial role in diabetes and obesity. Its density and sensitivity could be altered during the progression of diseases such as AD. Frolich et al. [91] reported in 1998 that brain insulin receptor density was increased in patients with sporadic AD compared to the age-matched control but decreased compared to middle-aged control. It has also been reported that AD pathological mediators such as amyloid- (A) peptide can also compete for insulin binding to the insulin receptor, interfering with insulin metabolism and leading to impaired glucose utilisation in the AD brain [92]. This evidence strongly suggests that targeting the insulin receptor for the purpose of treating a brain disease may result in disturbance of insulin metabolism and may affect insulin utilisation in the brain, therefore, it must be used with caution. 3. Lipoprotein receptors: Low density lipoprotein receptor (LDLR), lipoprotein receptor-related protein (LRP) 1 & 2 are all related and expressed on the BBB. They are multifunctional, multi-ligand scavenger and signalling receptors [93]. LRP has been reported to serve as a receptor for amyloid beta precursor protein (APP), apolipoprotein E (ApoE) and alpha-2-macroglobulin (2M), all of which have been genetically linked to AD [94]. LRP is therefore, thought to contribute to the pathobiology of AD. The levels of LRP that are downregulated substantially with age [95] are, a major risk factor for nonfamilial AD. Such down regulation of expression of LRP decreased the LRP-mediated clearance of Abetaanti-Abeta complexes, contributing to AD development [96]. On the other hand, increased expression of LDLR was observed in the hippocampus of stroke-prone spontaneously hypertensive rats associated with BBB impairment [97]. It has been suggested that because LRP inhibits the inammatory process, the expression of LRP may be affected by most brain diseases which are associated with inammatory process such as AD, Parkinson's disease, MS and encephalitis [3]. This may explain some of the success seen with LRPtargeted systems such as melanotransferrin/P97 and RAP transport systems 4. Diphtheria toxin receptor (DTR): DTR also known as the precursor of heparin-binding epidermal growth factor ( HB-EGF). It is a well characterised internalizing transport receptor on the BBB, neuro, and glial cells [98]. DTR involves receptor-mediated endocytosis with some unique properties. It has no endogenous ligands, thus, neither competes with nor interferes with endogenous ligands or transport of essential nutrients into the brain [99]. Since upregulation of DTR expression has been reported in many inammatory conditions induced by brain diseases [100102], it has been proposed as a useful receptor for site-specic disease targeting [99]. From the above review, it can be concluded that designing or selecting an appropriate drug delivery system for transporting drugs across the BBB, should take into consideration the impact of the diseased brain on the transporter or receptor systems. If a receptor is amplied or up-regulated under pathological conditions, it will aid drug transport into the brain via RMT. If the receptor is responsible for transporting nutrients or metabolites, it will not be a good candidate target for drug delivery across the BBB. One also needs to be aware that receptor-mediated endocytosis has high afnity but may

not necessary guarantee efcient transcytosis. Yu and colleagues demonstrated recently that low-afnity anti-TfR antibody variants showed a higher level of transcytosis and permitted more to reach the mouse brain to produce therapeutically relevant concentration compared to high afnity anti-TfR antibody [103]. Their design of bispecic antibody proved to be effective in delivering the antibody against enzyme beta-secretase into the brain for treating AD. 4.2.3. Monocyte and macrophage trafcking across the bloodbrain barrier Perivascular macrophages, which reside on the parenchymal side of endothelia cells, close to astrocyte endfeet, originally come from circulating phagocytes such as monocytes and have shown a remarkable capability to cross an intact BBB with 80% turnover in 3 months [104]. Such frequent migration of perivascular macrophages across the BBB plays an important role in the innate and adaptive immune response for protecting the CNS from pathogens. Interestingly monocytes/macrophages have been suggested as being utilised by pathogens as a vehicle to enter the CNS [26]. This transport route is also named as Trojan Horse mechanism and accepted as one of the means for pathogens invasion of the CNS [27]. In a recent rodent infection study Charlier et al. demonstrated that bone marrowderived monocytes infected with C. neoformans produced a 3.9 fold increase in CNS infection compared to the treatment with free yeast [105], highlighting the prominent role that monocyte trafcking plays in CNS infection by C. neoformans. In addition, there is compelling evidence to suggest that transmigration of infected monocytes through the BBB is one of the major mechanisms for HIV infection of the brain [106]. When the brain is overtaken by neuroinammation caused by disorders such as stroke and MS there is evidence showing that the inammation process can lead to a breakdown of the BBB and increased trafcking or migration of some immune cells, such as leukocytes including monocytes and macrophages [107,108]. It has been suggested that monocyte migration through the disrupted cerebral endothelial cell (EC) junctions plays an essential role in the formation of MS demyelinating lesions [108]. Furthermore, monocyte adhesion and subsequent migration in MS was found to be predominantly regulated by VCAM-1, an adhesion molecule expressed on brain capillaries [109]. In the case of brain tumours, histological analysis of gliomas has shown a high level of macrophage and microglia inltration [110,111]. Using erythrocyte bound IgG and tumour homogenate, Morantz et al. [112,113] quantied the macrophage content in 11 glioblastomas and found values ranging from 8 to 78% with a mean of 45%. In another study, endogenous macrophages were detected within the tumour or tumour periphery but not in the nontumoural hemisphere 2 weeks after implantation of a tumour in a rat brain [114]. These ndings suggest that macrophage and microglia accumulation in tumours such as glioma occurs naturally and could be a part of the immune defence mechanism in response to pathological conditions. It was suspected that a chemokine, monocyte chemotactic protein (MCP-1), which was found in glioma cells might have promoted monocyte and macrophage inltration of glioma [115]. One can speculate that this naturally preferential brain tumour targeting property of monocytes and macrophages provides a promising opportunity for delivery of anticancer drug across the BBB via monocyte or macrophage cell-mediated transcytosis, and directly into the tumours. Furthermore, Djukic et al. demonstrated that in the case of bacterial meningitis, there is an increased transmigration of monocytes across the BBB and their differentiation to microglia [116]. By tracking uorescently labelled monocytes in the diseased brain, the study also showed that these newly recruited monocytes became an integral part of the pool of parenchymal microglia and contributed to the clearance of damaged tissue.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

These studies provide evidence to support the rationale that the increased trafcking of circulating cells such as monocytes/macrophages across the BBB in the disease brain should be fully utilised and exploited as a potential cell-mediated transport mechanism and their role in maximizing the effectiveness of a drug delivery system for the treatment of CNS diseases warrants further investigation and exploration. Recently there are increasing efforts directed towards this area and this will be one of the focuses in this review on modern approaches of drug delivery across the BBB. 5. Modern methods of transporting drugs across the bloodbrain barrier As previously stated, the various transport mechanisms across the BBB can be summarised as per Fig. 2. The paracellular pathway, transcellular pathway, transport proteins, efux pumps, RMT and adsorptive transcytosis described in Fig. 2 have long been accepted as potential transport routes for drug entering the brain [20]. The cellmediated transcytosis is a relatively new approach for transporting therapeutical materials that emerged in last decade [25,117]. The rapid progress in molecular biology has propelled the development of novel drug delivery systems that take advantage of our better understanding of the BBB, the brain and various brain disorders. There is an increase in multi-discipline approaches combing biology, nanotechnology and even biophysics to achieve the common goal. With this in mind, the following section will focus on the latest developments in TJ opening, receptor-mediated, adsorptive-mediated, efux pump inhibition and cell-mediated transportation approaches. 5.1. Drug transport across the bloodbrain barrier via tight junction opening In the past two decades, there has been gradually emerging knowledge and understanding of molecules involved in TJ and AJ, and parallel discovery of modulators which can be used for opening the BBB, ranging from chemical and biological substances to physical stimuli such as high frequency focused ultrasound and electromagnetic elds (EMF) [47,118,119]. Table 2 presents examples of a wide range of these modulators and their impact on the BBB and drugs or tracers passage through the BBB. The rationale for modulating TJ opening to enhance the paracellular approach is fourfold: 1) the TJ opening or BBB leakage is a phenomena
Table 2 Effect of selected stimuli/agents on the bloodbrain barrier (BBB) and drug transport. Stimuli/agents Chemical Cyclodextrin Poloxamers (Pluronic block copolymers) Cell penetrating Peptides Peptide Biological Virus Effect on the BBB Extracting cholesterol from BCEC membrane leading to opening of tight junctions Inhibiting P-gp and MRP efux transporters at the concentration below critical micelle concentration inconsistent Binding to neuron cell acetylcholine receptor Upregulation of chemokines to open tight junctions

associated with many brain diseases (Table 1) and stimuli, and many modulators have already been characterised; 2) enhanced paracellular transport will increase the delivery of small water soluble molecules into the brain; 3) modulated TJ opening may also improve the BBB passage of macromolecules and drug delivery systems including liposomes, nanoparticles, micelles, polymer conjugates, and their distribution in the brain; 4) using physical stimuli such as ultrasound and EMF will temporarily provide local BBB disruption, therefore, concentrated drug can be delivered locally.

5.1.1. Enhanced by biological stimuli Several modulators with capacity to temporarily open TJ to enhance the transport of drugs and traces have been reported and have been mostly studied on in vitro cell models (for review see [57]). It is interesting to note that a 45 kDa biological molecule zonula occludens toxin (Zot), an active TJ modulator at the BBB, can induce a reversible, concentration-dependent TJ opening (measured by TEER decrease), which increases the paracellular transport of sucrose and inulin (permeability markers) without detectable short-term toxicity in cultured bovine brain capillary endothelia cells [120]. In addition, it also permits an enhanced transport of the therapeutic agents doxorubicin and paclitaxel that are substrates of P-gp and otherwise would have very low transportation across the BBB [120]. Other groups of biological compounds which can act on TJ are vasoactive compounds and inammatory stimuli such as histamine, bradykinin and VEGF. They are products or mediators of the inammation process and can increase BBB permeability [54,121]. In the case of brain tumours such as gliomas, microvascular permeability in tumour tissue is more sensitive to the effects of these biological compounds than the normal brain endothelia cells [122]. Therefore, these stimuli, when used in combination with imaging materials, gene or anticancer drugs, can potentially boost the preferential delivery of these materials to the brain tumour to achieve tumour diagnosis, tumour gene therapy or chemotherapeutic treatment. This hypothesis is supported by the experimental results obtained on a rat glioma model which showed Intracarotid histamine infusion selectively increases permeability of Evans blue albumin and enhanced the transport of -aminoisobutyric acid in brain tumours without affecting BBB permeability in the normal brain tissues [123,124]. It is suggested that the signalling pathway for histamine's effect involves H2 receptors, NO and cyclic GMP production [124126].

Effect on drug/marker transport across the BBB High percentage of DOX across in vitro brain endothelia cell monolayer Its co-administration with digoxin enhanced the drug delivery into the brain. Cannot cross intact BBB; transport drug via adsorptivemediated transcytosis Delivered siRNA to the neuronal cells Permitted passage of Glutamic acid decarboxylase (GAD) gene with adeno-associated virus (AAV) safely improved condition in AD patients May increase paracellular transport Facilitated brain delivery of didanosine and indinavir Enhanced the CNS delivery of carboplatin, loperamide and cyclosporin-A Enhanced delivery of antibody, chemotherapy and gene Enhanced horseradish peroxidase transport in a rat model Increased BBB permeability of saquinavir in an in vitro BBB model

References [155] [276] [304] [305] [139,145]

HIV-1clade-sepcic Tat protein B Macrophage Cereport (RMP-7) Physical Ultrasound Microwave Electromagnetic eld BCEC: brain capillary endothelial cells.

Disrupt BBB integrity Trojan horses effect Open tight junctions

[306] [307,308] [130,135,128,127,134]

Transcytosis ; transendothelial openings; partial opening of tight junctions Increasing BBB permeability via thermal effects Opening tight junctions via protein kinase C signalling and tight junctions protein translocation

[309311,118] [165] [177,178]

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

10

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

Cereport (also known as RMP-7 or Lobradimil), a synthetic peptide analogue of bradykinin, has been extensively studied for facilitating drug transport via the paracellular route [127130]. Cereport increases the permeability of the BBB by transiently disrupting the TJ [131]. It shows specic time, dose and size dependent actions on human brain microvascular endothelial cells and its effect can be modulated through changes in cAMP and cGMP second messenger systems [132]. Cereport is also unique in that it is effective when it is given via either intracarotid or i.v. administration, although the latter does require a higher dose [128,133]. Studies have shown that Cereport is capable of enhancing the BBB transport of a number of drugs including carboplatin, loperamide, and acyclovir in different types of diseased animal models [127,128,134]. Because Cereport has no toxicity by itself, can selectively increase drug uptake in the brain tumour and showed less effect in non-permeable normal brain, it was once held as one of the most effective BBB modulators despite its inability to inhibit the P-gp efux pump, therefore no effect on BBB transport of drugs which are P-gp substrates such as paclitaxel [135]. Nevertheless, at a high dose, Cereport did show an enhanced survival rate in rat glioma model when combined with carboplatin [135]. When attached to the surface of a liposome, Cereport is even more effective in facilitating Evans blue transport into the brain compared to free Cereport with liposome [136]. The latter was ineffective due to the different arrival times of free Cereport and Evans blue incorporated liposomes at the BBB, which results in inefcient opening of BBB for Evans blue incorporated liposomes. This study also compared the bioactivity of Cereport-attached liposomes with free Cereport in vitro on mouse brain microendothelial vessel cells and showed that the former was a little stronger than that of free Cereport. This study also demonstrated that Cereport attached liposomes can be potentially used for transporting different types of drugs, including P-gp substrates via transient TJ opening. Despite positive results obtained with Cereport in facilitating carboplatin in animal models and phase I clinical trial, the outcomes of a phase II clinic trial were inconsistent. The combination showed a signicant activity in recurrent malignant glioma patients following radiotherapy [137], whereas, it was found to be inactive in childhood high-grade gliomas and brainstem gliomas [138]. This highlights the challenge of translating experimental research into a clinical therapy. Nevertheless, Cereport still warrants further research in enhancing drug transport across the BBB. Virus is also one type of biological material which can act as stimuli and open the TJ via upregulation of chemokines as a precursor for inltration of inammatory cells into the CNS [139]. Immunohistochemical analysis of CNS tissue from HIV-1-seronegative and HIV-1-infected patients, both with and without encephalitis, revealed signicant tight junction disruption in patients who died with HIV encephalitis, as shown by fragmentation or absence of immunoreactivity for occludin and ZO-1 [140]. These phenomena were associated with accumulation of activated HIV-1-infected brain macrophages, brinogen leakage, and marked astrocytosis, suggesting that the main route of HIV-1infected monocyte entry into the CNS could be the disrupted BBB structure [140]. Further evidence was also reported in a study with human brain microvascular endothelial cells where the increased BBB microvascular permeability was found to be due to the degradation of tight junction proteins ZO-1 and ZO-2 caused by HIV type 1 gp120 [141]. In addition to change of tight junction proteins, other alterations, such as expression of matrix metalloproteinases in virus-infected BBB can result in the BBB integrity being compromised, allowing unrestricted entry of immune cells into the brain, thereby contributing to virus induced neuropathogenesis [142]. A study conducted by Verma et al. [143] indicates that a cell-free virus, such as West Nile virus, has the ability to enter the CNS without compromising the BBB integrity. It gained access across the BBB via the Trojan horse mechanism through induction and increased expression of cell adhesion molecules that

permit migration of monocytes/macrophages through the BBB. Recently, Foust and colleagues [144] demonstrated the intravenously administered adeno-associated virus (AAV9) can efciently target cells of the CNS such as neonatal neurons and adult astrocytes without disruption of the BBB. Although the precise mechanism by which AAV9 bypasses the BBB is unclear, it was speculated that AAV9 may utilise transport proteins, receptor mediated transcytosis, adsorptive transcytosis or other mechanisms to cross the BBB [144]. This unique property of AAV9 is of great interest for development of a non-invasive method to achieve efcient gene delivery to the CNS. When Kaplitt et al. [145] studied the delivery of glutamic acid decarboxylase (GAD) gene with (AAVinto the subthalamic nucleus of patients with Parkinson's disease, they found this gene therapy was well tolerated by patients and safely improved the symptoms of patients with advanced Parkinson's disease. The use of viruses for gene delivery to the CNS is still in its infancy, and its long term effects and toxicity are yet to be fully understood. 5.1.2. Enhanced by chemical stimuli There are several examples of chemical stimuli used to modulate the TJ to increase the permeability of the BBB for drug transport [47]. An often employed BBB opening practice is via arterial injection of hyperosmolar solution (e.g., mannitol, arabinose). The shrinkage of endothelial cells results in transient opening gaps between cells. One major limitation of this approach is its invasiveness, which requires considerable expertise [146]. Here we will highlight actions of some pharmaceutical excipients on the BBB. For instance, oleic acid, a PKC activator, showed reversible opening of the BBB to Evans blue albumin and -aminoisobutyric acid when it was given via arterial infusion in a rat model [147]. Lysophosphatidic acid was also reported to increase TJ permeability in cultured brain endothelia cells [148] via the activation of PKC-alpha channels which reduces caudin-5 expression and F-actin recombination [149]. Like some biological stimuli, its action is also rapid, dose-dependent and reversible, and its effect can be attenuated by activation of protein kinase C [148]. One of the most commonly used pharmaceutical excipients, sodium dodecyl sulphate (SDS) has also been shown to induce an extensive but reversible, dose-dependent Evans blue extravasations and increased transport of -aminoisobutyric acid at a dose of 25100 g/kg in rats [150]. This is not surprising considering SDS is an anionic surfactant with function as a solubilisation agent and may interact with lipids or protein in the cell membrane [151]. Cyclodextrins (CDs) are cyclic oligosaccharides composed of 6, 7 and 8 glucose units; namely -, -, -CD, respectively. They are known for their ability to form inclusion molecular complexes to increase the water solubility of hydrophobic drugs (guest molecules) [152]. Monnaert and colleagues studied the endothelial permeability and toxicity of native, methylated, and hydroxypropylated -, -, -CD in an in vitro cell model [153]. Native -, -CD elicited a rapid increase in sucrose permeability of cerebral endothelial cell monolayers which correlated with their ability to extract phospholipids [153]. Among those tested, -CD had the least toxicity. High concentrations of hydroxypropyl -CD and -CD increased doxorubicin passage through the brain endothelia cell monolayers but at the expense of a loss of BBB integrity and decreased junctional staining of occludin [154]. This nding suggests that oligosaccharide units are likely to be responsible for the toxicity of CDs in the brain because of their extraction of lipophilic components of the BBB, phospholipids and, in some cases, cholesterol, which may break down the brain endothelial cell monolayers or lead to decreased P-gp activity [155]. Interestingly, ammonium CD derivatives (monosubstituted nalkyldimethylammonium--CD derivatives, DMA-Cn-CDs 2 b n b 16) showed much lower toxicity than native -CD. In particular, DMAC12-CD always remained non-toxic even at a concentration as high as 10 mM [156]. It appears that a long alkyl chain is responsible for producing non-toxic effects by obstructing the CD cavity, therefore reducing the extraction ability of CDs for phospholipids and cholesterol. In

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

11

addition, the masked amphiphilic structure of DMA-C12-CD was also thought to be responsible for its permeability through the BBB. 30% of DMA-C12-CD passed through the endothelial cells at 5 mM [156]. Unfortunately, all these studies were conducted in vitro, and there is yet no conrmation if CDs would work in vivo for enhancing drug transport across the BBB. 5.1.3. Enhanced by physical stimuli The ability of energy-based physical methods, such as ultrasound, microwave or electromagnetic elds, to open the BBB has been investigated. Extensive reviews can be found elsewhere [118,119]. Development of acoustic technology has enabled ultrasound to become not only a diagnostic tool but also a therapeutic modality. Focused ultrasound techniques concentrate acoustic energy in a focal spot deep in the body with minimal effect to tissues outside the eld of focus [157]. This allows it to non-invasively induce local biological effects deep inside the body and removes the need for surgical intervention. Hynynen et al. showed that introduction of a preformed gas bubble before focused ultrasound exposure would allow transient opening of the BBB locally without causing acute damage to the neurons [158] or delayed ischemia. The gas bubble not only connes the ultrasound effect to the vasculature, but also reduces the power needed to open the BBB, making it possible for the ultrasound to be applied through the intact skull. By combining with an imaging device such as magnetic resonance imaging (MRI) scanner, ultrasound becomes a non-invasive approach to open targeted regions of the BBB to permit the delivery of drugs and other therapeutic molecules across the BBB [118]. Sheikov et al. studied the effect of focused ultrasound in a rabbit model and revealed that at the acoustic power applied (0.55 W and 3 W), TJ opening occurred with leakage of dye and contrast matter [159]. Characteristic ultrastructural changes of the brain microvasculature were seen under electron microscopy, however they were conned to sonicated locations. The electron microscopic data show that sonication produced acoustic power-dependent tissue damage with the damage by 0.55 W equivalent to categories 12 and a low level of red blood cell extravasations [159]. In a separate study, sonication with the same parameter, 1 MPa (0.55 W), induced no delayed damage to neurons for up to a month. [160]. Using goat antirabbit IgG conjugated to 10 nm gold particles and electron microscopic analysis, the group also demonstrated that the ultrasound induction of fenestration appeared to be rapid with fenestrate-like openings seen on samples in 1 and 2 h after the sonication. The cytoplasmic channels were observed in the endothelia cells and appear to be formations for transendothelial passage [159], a phenomenon similar to that reported in mouse brain capillaries in response to heat stress [161]. Evidence suggests that the local, reversible disruption of the BBB by bursts of low frequency MRI-guided ultrasound enhances the brain delivery of monoclonal antibodyHerceptin (trastuzumab) in mice [162] and doxorubicin in rats [163]. It was suggested that the mechanisms for transport of molecules by focused ultrasound may involve transcytosis, transendothelial openingsfenestration and channel formation, widening of interendothelial clefts and partial opening of TJs and free passage through the injured endothelium [159]. In a recent report [164], MRI-guided focused ultrasound facilitated the delivery of anti-Abeta antibody, BAM-10 to Abeta plaques in targeted cortical areas following intravenous injection in a mouse AD model. The reduction in Abeta pathology became apparent four days post treatment [164]. It was suggested that the mechanisms for transport of molecules by focused ultrasound may involve transcytosis, transendothelial openingsfenestration and channel formation, widening of interendothelial clefts and partial opening of TJs and free passage through the injured endothelium [159]. Such rapid reduction in plaque pathology indicates MRI-guided focused ultrasound has great potential in enhancing the transport of both small and large molecules across the BBB for delivery of a target therapy for

treatment of CNS disorders. There is an urgent need to further develop and ne tune the technology to allow the repeat application of ultrasound bursts without producing sustained damage to the tissue and the BBB. Microwave energy has also been studied for drug transport into the CNS [165]. It was reported that Chinese hamsters exposed to low level microwave energy exhibited reversible permeability of the BBB to horseradish peroxidase (HRP) [166]. In a separate study, microwave irradiation facilitated central effects of domperidone by altering the permeability of the BBB and increasing the entry of the drug into the CNS [167]. Since sufciently strong microwave energy can lead to tissue heating, the resultant brain temperature increase may produce increased BBB permeability. This was shown in a recent study where neuronal albumin uptake in brain was shown to be correlated with brain temperature, with more apparent effects observed when temperature was raised 1 C or more [168]. Indeed, reports show that exposure of the rat head to microwave frequencies (2.5 3.2 GHz) leads to an increase of brain temperature above 40 C, which can enhance the BBB permeability to HRP [165], Evans blue [169,170] and sodium uorescein [171]. It is interesting to note that when brain temperature was cooled down below 40 C, microwave irradiation failed to open the BBB suggesting the mechanism of barrier opening is not related to the non-thermal effect of microwaves [165]. Stam reviewed all evidence reported with laboratory animals and came to the conclusion that acute, non-thermal microwave exposure does not increase BBB permeability [119]. If that is true, one has to be cautious about exposure to microwave at thermal levels as it could put the brain at risk of infection [172]. Exposure to the electric magnetic eld of MRI can also potentially reach thermal levels under certain circumstances [119]. Although there are studies carried out on assessment of the effects of MRI on BBB permeability, the ndings are inconsistent: some showed an increased BBB permeability [173,174], others showed no change [175,176]. To date there is insufcient evidence to make a conclusion on the impact of MRI on BBB permeability and drug transport. There are also examples of using electromagnetic eld (EMF) pulses to induce the permeability of the BBB. Qiu et al. showed that electromagnetic pulses induce BBB permeability via regulating protein kinase C signalling and translocation of tight junction's protein ZO-1 [177]. When the antiretroviral agent saquinavir was loaded in polymeric and solid lipid nanoparticles and investigated for BBB permeability under the inuence of an EMF in an in vitro BBB model, enhanced transport of antiviral drug across BBB was observed. The level of enhancement was dependent on wave shape, frequency and amplitude of EMF [178]. The study demonstrated the combination of nanoparticle carrier and EMF strongly benets drug transport and could have potential clinical application to the therapy of brainrelated disease. Overall, ultrasound as physical stimuli to transiently open the BBB to enhance the drug transport into the brain has received signicantly more research effort than other methods. Although its mechanisms for drug transport and sustained impact on tissue are not fully understood yet, MRI-guided focused ultrasound does have attractive features that it can provide diagnosis and treatment at the same time and can be used to enhance not just small molecules, but also large proteins or even particulate delivery systems such as liposomes and nanoparticles [118]. It would appear to have the potential to be an important drug delivery modality for treatment of CNS diseases. In summary, the TJ opening approach is a double-edged sword. On one hand, it provides enhanced BBB permeability to a large number and variety of drugs, proteins, peptides, genetic materials, and even drug delivery systems such as liposomes or nanoparticles, without the need to modify the chemical structure of the drug. On the other hand, opening of TJ for a drug molecule may also lead to the enhanced passage of other molecules and unwanted substances such as pathogens through the protective barrier because this paracellular route is

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

12

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

not specic enough to exclude CNS entry of toxins and other unwanted molecules. It is, therefore, important to be able to control the time and duration of reversible opening of TJ and to ensure the frequent application of stimuli will not have an impact on the BBB and brain conditions, thus this approach can provide both efcacy and safety for a brain therapy. 5.2. Transport system-mediated drug delivery across the bloodbrain barrier The combination of endogenous transport systems present in the BBB and macromolecular conjugates or surface enhanced nanoparticulate delivery systems such as liposomes, nanoparticles and supermolecular complexes has been utilised to access the brain. As discussed previously, transporter proteins, specic receptors or adsorptive endocytosis can be used to realize drug delivery. The characteristics of these systems are 1. the drug is either chemically modied, for instance, conjugated to a ligand or polymer as a homing device thereby masking its intrinsic properties; 2. the drug is encapsulated in a surface modied drug delivery system, such as liposomes, nanoparticles or niosomes. The surface of the drug delivery system is often modied to contain a homing device and a hydrophilic polymer such as PEG to prolong the circulation time of the delivery system. The latter is necessary for achieving a high drug concentration gradient at the BBB [29]; 3. the homing device and drug delivery system should be nonimmunogenic (unless it is targeting the monocytes/macrophages) and capable of interacting with receptors presented at the BBB to facilitate the uptake of the drug by the BBB; 4. the homing device must be receptor specic, thereby reducing potential side-effects and increasing transport efciency; 5. all systems must have controlled size, therefore their properties are uniform and consistent and their biological fate can be controlled. The best system will be the one which can full all these characteristics and have a homing device that is specic for a target which is induced or up-regulated by the pathological conditions (see Section 4.2). 5.2.1. Nanocarriers for brain drug delivery Nanocarriers are an emerging class of drug delivery systems that can be easily tailored to delivery drugs to various parts of the body, including the brain. In the past decade, it has been attracting increasing attention for its use in transport of drug across the BBB due to the rapid increase in our understanding of receptors and the fast development in polymer chemistry and nanotechnology. Nanocarriers are unique because of their size and easily tailored structures due to the material used. They can behave like macromolecules in certain circumstances but they can carry much more drug payload and are capable of controlling drug release. They can carry a range of drugs and their surface properties can be modied. These properties make nanocarriers an attractive alternative for transporting drug across the BBB. Nanoscale drug carriers consist of particles in the size range from 10 to 1000 nm. Ideal properties of nanocarriers for drug delivery across the BBB are listed below [179,180]: Nontoxic, biodegradable and biocompatible Particle size less than 100 nm (except if transport via monocytes or macrophages) Stable in blood (no aggregation and dissociation) Prolonged blood circulation time Non-immunogenic

BBB-targeted moiety (receptor or adsorptive mediated mechanism, or uptake by monocytes or macrophages) Well maintained parent drug stability Tunable drug release proles Applicable to carry small molecules, proteins, peptides or nucleic acids Despite a large variety of nanocarriers developed so far, it is noteworthy that only amphiphilic molecule-formed liposomes and polymeric nanoparticles have been extensively exploited for brain drug delivery [181]. Several such systems are now in clinical trials for anticancer drug delivery. For instance, the University of Regensburg in collaboration with Essex Pharma (Schering-Plough) Germany has completed a phase II clinical trial of pegylated liposomal doxorubicin and prolonged temozolomide in combination with radiotherapy in newly diagnosed glioblastoma [182]. Compound 2B3-101, a brain targeted doxorubicin liposomes coated with the tripeptide glutathione at the tips of polyethylene glycol (PEG), is in the Phase I/II clinical trial in the Antoni van Leeuwenhoek hospital, the Netherlands. Nonamphiphilic colloidal drug carrier systems such as dendrimers and microemulsions are still at relatively early development stage and therefore will not be covered in this review. In the following sections the structure and compositions of liposomes and micelles will be briey introduced with emphasis on polymeric nanoparticles for drug delivery to the brain. 5.2.2. Supramolecular architectures of aggregated amphiphilic molecules An amphiphile, by denition, is a chemical species having a polar (hydrophilic) head group and hydrophobic tails. Amphiphiles interact very strongly with water. At low concentration, amphiphiles accumulate at the surface of the water because the free energy of the air-water interface is reduced. The head group of the amphiphile orients to the water, and the hydrophobic tail group is adjacent to the air. Above a certain critical concentration the air-water interface is saturated and the amphiphilic molecules aggregate in the bulk of the water forming various morphologies, such as lamellar bilayer, micelles, rods, vesicles or larger hexagonal aggregates. Vesicles are hollow, lamellar spherical structures. In cell biology, vesicles are relatively small and enclosed compartments, separated from the cytosol by at least one lipid bilayer. Their size ranges from 20 nm to 100 m, whereas the thickness of the membranes is around 3.5 nm. Liposomes are phospholipids vesicles frequently used for drug delivery purposes [183,184]. Niosomes are non-phospholipidbased synthetic vesicles/micelles that have properties and functions like liposomes [185,186]. Micelles are basically spherical aggregates of amphiphilic molecules dispersing in water with their hydrophilic head groups on the surface of the sphere, and their hydrophobic tails collected inside. An important property of micelles is their ability to increase the solubility and bioavailability of poorly soluble pharmaceuticals. The amphiphilic molecules in micelles are in constant exchange with those in the bulk solution. On the other hand, polymeric micelles, also known as polymersomes, are self-assembled polymer shells composed of block copolymer amphiphiles [187] such as polyethylene glycol-polylactic acid (PEG-PLA) and PEG-polycaprolactone (PEG-PCL). Block copolymers have the same basic amphiphilic property as lipids (Fig. 3) but they consist of distinct polymer chains covalently linked in a series of two or more segments [187]. Polymeric micelles differ from nanoparticles that are either more solid or monolithic (nanospheres) or contain an oily or aqueous core and are surrounded by a polymer shell (nanocapsules). However, in practice, polymeric micelles also be referred to as nanoparticle or nanocarriers because of their particle size. Amphiphilic block copolymers have recently emerged as a special class of materials. This is because polymer molecular weights can be orders of magnitude greater than those of lipids, self-assembled structures of which are rather stable. In addition, the diversity of

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

13

Fig. 3. (a) Natural lipid versus synthetic polymer assemblies. (b) Self-assembled block copolymers from hydrated lms (polymeric micelles). (c) Block copolymer labelled with uorescent dyes and hydrated (uoro-polymeric micelles). (d) Cryogenic transmission electron microscopy of approximately 100-nm polymeric micelles. The two arrows point to spherical and rod-like micelles that sometimes coexist with polymeric micelles. Reproduced from reference [187].

block composition and block length offers the possibility of designing the most desirable drug delivery systems. The most commonly used hydrophobic core-forming polymers are poly(propylene glycol), poly(D,L-lactide) and poly(caprolactone). Cholesterol is often included in membrane phospholipids to stabilize the membrane and reduce the membrane permeability towards encapsulated materials. In this context, cholesterol is an intriguing component of cell membranes because it both toughens and uidizes the membrane. Cholesterol is often employed as the hydrophobic segment because cholesterol possesses good bio-compatibility and the potential for interaction with cholesterol receptors on the cell surface, and strong ability to drive self-assembly of cholesterol containing polymers [188]. 5.2.3. PEGylation of nanocarriers Most studied brain-targeted systems are liposomes, polymeric micelles and nanoparticles. These systems are able to protect enclosed drug from chemical and biological degradation in the blood stream, control their release, permit surface modication with targeting ligands and allow for steric stabilisation or PEGylation. Liposomes are made of natural biological lipids with structural similarity to the cell membrane and are therefore considered to be biologically compatible with low toxicity [189]. Without the use of surface modication by a hydrophilic polymer such as polyethylene glycol (PEG), the biological half-lives of liposomes is very short due to a number of factors including the tendency of the liposome to exchange lipid materials with cell membranes and their uptake by phagocytes [189]. The grafting of PEG to the surface of liposomes signicantly changed their fate and extended their residence time in vivo. Thus PEGylated liposomes have become a standard preparation particularly when tissue/receptor targeting is desired [190192]. A highly exible and hydrated PEG chain attached to the liposomes surface is assumed to have an effective

opsonins-resistant property due to its steric repulsion effect [189]. The PEGylation or stealth technology has also been applied to all other drug delivery systems, including macromolecular conjugates, nanoparticles, dendrimers and polymeric micelles [187]. Grafting PEG to polymer or including PEG as part of the block copolymers essentially also made the polymer molecules amphiphilic. PEG-containing surfactants, poly (oxy-ethylene)-poly(oxy-propylene) block copolymers, (poloxamer 338 = PluronicF108 and poloxamine 908 = Tetronic ) were also found to be effective in prolonging the blood circulation time of nanoparticles, even by coating [193,194]. Compared to PEG-surfactant coating micelles or nanoparticles, self-assembly from PEGylated polymers provides a better choice due to the chemical binding of PEG onto the nanoparticles. The PEG chains were rst bound to poly(lactic acid) nanoparticles by Gref et al. [195] and Bazile et al. [196]. Later PEG was chemically linked to poly(hexadecyl cyanoacrylate (PHDCA) nanoparticles [197]. Both PEGylated poly(lactic acid) and PHDCA nanoparticles, signicantly prolonged the blood circulation time and reduced liver uptake. Calvo et al. investigated the distribution of PEG-PHDCA nanoparticles in EAE rats [198]. Analysis by confocal microscopy showed evidence that uorescent PEG-PHDCA nanoparticles were present in the epithelial cells of the pia mater, ventricles, spinal cord surface and in the ependymal cells of the choroid plexus, with less found in the endothelia cells of the BBB. In the same study, Poloxamine 908-coated PHDCA and polysorbate-80 coated nanoparticles exhibited less penetration of the BBB. It was hypothesized that PEG-PHDCA nanoparticles reached the brain by two mechanisms: passive diffusion due to the increase of the BBB permeability and transport by nanoparticles-containing macrophages, which inltrated these inammatory tissues. The PEGylated PHDCA nanoparticles accumulated in the brain at a 48 fold higher concentration than the non-PEGylated PHDCA nanoparticles

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

14

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

after intravenous injection into rats with 9 L gliosarcoma [199]. Therefore, the covalent attachment of PEG chains was a promising step forward. Other PEG-containing copolymers showing potential ability for drug delivery to the brain are depicted in Fig. 4. 5.2.4. Functionalization of nanocarriers for drug transport across the bloodbrain barrier Functionalization of nanocarriers is one of the most important steps or challenges in formulating nanocarriers for drug delivery. The challenge is not only in the chemistry, but also in selecting and designing appropriate targeting ligands that can achieve the targeting or homing function. Functionalization itself entails a thorough understanding of the target organ and transport mechanisms, and more importantly their functions in relation to a particular pathological condition, which was discussed previously. As shown in Tables 1 and 2, some of the transport mechanisms may be up regulated or down regulated depending on the type of brain disease. There are also a number of agents which can have direct effect on the BBB (Table 2) [180]. One needs to take into consideration the sensitivity of the BBB to the functional groups on the surface of the nanocarrier as well as the potential dose dependent toxicity of the ligands. In terms of a chemical approach to functionalization, PEG is a good candidate for functionalization. In addition to providing steric stabilisation, the presence of PEG on the surface of the nanocarriers also allows for the preparation of bioactive nanocarriers by attaching bioactive ligands on the surface for targeted delivery to the brain. There are obvious reasons for the design of ligand coated longcirculating drug carriers: 1) Ligand (an antibody, protein, peptide, sugar moiety, folate or carbohydrate) attached to the carrier surface may increase the rate of elimination from the blood and uptake in the liver and spleen. However, the presence of the PEG protecting polymer may compensate for this effect; 2) Longevity of the specic ligand-bearing nanocarrier may allow for its successful accumulation in targets with diminished blood ow or with low concentration of the surface antigen. Covalent linkage of targeting ligands to the nanocarriers is typically made by a simple coupling reaction between amine-functionalized nanocarriers and succinimidyl ester derivatives. To achieve better selective targeting, ligands should be attached to the nanocarrier via the PEG spacer arm, so that the ligands are extended outside of the dense

PEG corona thus excluding steric hindrance for their binding to the target receptors [29]. 5.3. Drug transport across the bloodbrain barrier via transport vectors One of the successful strategies of the delivery of molecules that are unable to pass the BBB is the use of transport vectors which activate natural transport routes. The endogenous carrier-mediated transport (CMT) for nutrients and AMT for peptides can be portals of entry to the brain for circulating drugs. The BBB expresses several transport systems for nutrients [200], but the utilisation of these transport systems for targeting drugs into the brain is mainly limited to peptide drugs, which must have a molecular structure mimicking the endogenous nutrients. The prototypical example is levodopa, a lipid-insoluble precursor of dopamine that has been used for the treatment of Parkinson's disease because it contains the carboxyl and -amino groups that allow it to compete for transport across the BBB by the large neutral amino acid carrier [201]. In addition, since nutrient carriers stay in the membrane of the cell, the size of the drugs must be close to that of the endogenous ligand if they need to be taken up and transferred into the brain [200]. This approach is generally less favoured because it may interfere with the transport of nutrients and also for certain molecules, for instance, antibiotics, that do not have structures that are close to that of endogenous ligands. 5.4. Drug transport across the bloodbrain barrier via adsorptive-mediated transcytosis AMT has recently gained signicant importance as a route for drug delivery into the brain due to growing evidence showing the success of this transport route for delivery of drugs into the brain via cationic proteins, and cell-penetrating peptides (CPPs) [24]. CPPs are positively charged peptides with amphipathic characteristics. They are capable of rapidly entering living cells without producing cytolytic effects. They have been successfully used as vectors for delivery of drugs that are P-gp substrates by effectively by-passing the P-gp in the BBB. For instance, they have increased doxorubicin transport into the rat brain up to 30-fold [202]. This approach is effective because cell-penetrating peptides utilise adsorptive-mediated endocytosis to enter the brain. Another notable success is the use of vectors such as SynB3 (RRLSYSRRRF) to increase brain uptake of poorly brainpenetrating drugs [24,203]. SynB peptides derive from a natural mammalian antimicrobial peptide with high afnity for biological membranes. It is one class of CPPs that have emerged, facilitating the intracellular delivery of polar biomolecules in vitro and in vivo. It was shown to enhance the transport of morphine-6-glucuronide to the brain in a clinical trial [30]. CPPs refer to a group of short peptides of less than 30 amino acids that are able to penetrate cell membranes and transport their cargo into cells [204]. Table 3 summarises the majority of CPPs with their principal features. While these individual CPPs differ in length and sequence, they share a few common features, which include their amphipathic nature, net positive charge, theoretical hydrophobicity and helical moment, the ability to interact with lipidic membranes, and to adopt a distinct secondary structure upon association with lipids [205]. The major dogma has been that CPPs enter cells by a receptor and energy-independent process but the exact mechanisms are not yet fully understood. For some CPPs, endocytosis is an exclusive and alternative mechanism of internalization. For instance, SynB's internalization is a temperature and energy dependent process and involves endosomal transport. It has been conrmed that SynB penetrates into cells via AMT [206]. On the other hand, TAT (HIV-1 trans-activating transcriptor) derived CPPs enter cells primarily by lipid raft-mediated macropinocytosis that is stimulated by cellsurface binding of TAT derived CPPs [207].

Fig. 4. Selected examples of PEG containing block polymers which can be selfassembled to form polymeric micelles for drug delivery.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx Table 3 Principle features of the selected cell penetrating peptides (CPPs). Peptide name MAP pAntp4368 Transportan SBP FBP TAT4860 SynB1 SynB3 Sequence KLALKLALKALKAALKLA RQIKIWFQNRRMKWKK GWTLNSAGYLLGKINLKALAALAKKIL MGLGLHLLVLAAALQGAWSQPKKKRKV GALFLGWLGAAGSTMGAWSQPKKKRKV GRKKRRQRRRPPQ RGGRLSYSRRRFSTSTGR RRLSYSRRRF Net charge +5 +8 +4 +6 +6 +8 +6 +6 Cell lytic activity Yes No Yes No No No

15

MAP: model amphipathic peptide; Antp: Antennapedia; SBP: sequence signal-based peptide; FBP, fusion sequence-based peptide; TAT, HIV-1 trans-activating transcriptor. The peptide residues in this table are expressed with one-letter-code: Klysine; L leucine; Aalanine; Rarginines; Qglutamine; Iisoleucine; Wtryptophan; F phenylalanine; Nasparagine; Mmethionine; Gglycines; Sserine; Tthreonine. Data was collected from references [204,206,221,222,312314].

The application of CPPs is based on the premises that biologically active cargo can be attached to CPPs and translocated into cells. The link between the CPPs and cargo is most commonly a covalent bond and seldom in non-covalent bond. A large variety of cargo molecules/materials have been effectively delivered into cells via CPPs, including small molecules, proteins, peptides, fragments of DNA, liposomes and nanoparticles [204]. Some can enter brain capillary endothelia cells or are even translocated into the brain tissues. Some examples are highlighted here. Adenot and colleagues studied brain uptake of a number of free and SynB3 vectorized chemotherapeutic agents using both in situ brain perfusion and in vitro BBB/cell model [203]. They reported that SynB3's conjugation with various poorly brain-penetrating drugs enhanced their brain penetration by a factor of 30 for doxorubicin, 7 for benzylpenicillin, 22 for paclitaxel, 18 for dalargin and 50 for morphine-6-glucuronide with no effect on tight junction integrity. Brain uptake of the enkaphalin analogue, dalargin, a hexapeptide, was enhanced signicantly when conjugated to SynB and injected intravenously in mice [208]. This study signalled the potential for delivery of peptides or drugs for treatment of brain cancer, through the targeting of brain tissue after systemic delivery. TAT is a HIV-1 trans-activating transcriptor with 101 amino acids. The protein consists of ve domains; probably the best-studied region of TAT is located in domain 4, which contains a highly basic region (with two lysines and six arginines in nine residues) involved in nuclear and nucleolar localization [209]. While all CPPs listed in Table 3 above have been used mainly for small cargoes such as peptides and oligonucleotides, Schwarze et al. [210] synthesized full-length fusion proteins that contained a NH2-terminal 11-amino acid protein transduction domain (PTD) from the HIV TAT protein. Transduction of the proteins evaluated was non-cell-specic, and was seen to occur even across the BBB. Further proof of this mode of peptide delivery was attained by Cao et al. [211] who fused the antiapoptotic protein Bcl-xL to TAT and injected the construct intraperitoneally into mice that were affected by stroke. The Bcl-xL protein is expressed in adult neurons of the CNS and is believed to have an important role in the prevention of neuronal apoptosis that would normally occur during brain development, or results from varying stimuli leading to pathology, including cerebral ischemia. Protein transduction with this entity occurred in a rapid, concentration-dependent fashion, with entry into cells thought to occur via the lipid bilayer component of the cellular membrane. A study by Kilic et al. [212] using the same model showed that brain tissue was progressively transduced with TAT proteins within 34 h after intravenous delivery. TAT-Bcl-xL treatment reduced infarct volume and neurological decits after long ischemic insults lasting 90 min, when applied both before and after ischemia. Studies have also shown that even relatively large particles could be delivered into various cells by TAT vector. A biocompatible 45 nm

nanoparticle with an iron core, a dextran coating, and covalently linked TAT peptides was efciently taken up by human hematopoietic CD34+ cells [213]. Even cytoplasmatic uptake of liposomes with a diameter of 200 nm has been documented [214]. Taking the TAT-mediated nanoparticles delivery approach a step further, one of the most exciting demonstrations of the effectiveness of TAT-shuttled nanocarriers across the BBB was accomplished by TAT-conjugated CdS:Mn/ZnS quantum dots (Qdots) [215]. Histological data clearly showed that TAT-Qdots migrated beyond endothelial cells and reached the brain parenchyma. TAT-mediated intracellular delivery of large molecules and nanoparticles was proved to proceed via the energy-dependent macropinocytosis with subsequent enhanced escape from endosome into the cell cytoplasm [207]. Recently, Liu et al. produced compelling evidence that TAT facilitates human brain endothelia cell uptake of nanoparticles self-assembled from TAT-PEG-b-cholesterol in vitro and more importantly, the nanoparticles with TAT were able to cross the BBB and translocate around the cell nucleus of neurons [216]. This study demonstrates the effectiveness of TAT in promoting the transport of nanoparticles across the BBB. It conrms that nanocarriers conjugated with TAT could be a promising carrier system for transporting drug across the BBB for the treatment of brain disorders. In a more recent study by Wang et al., cationic nanoparticles fabricated from cholesterol-CG3R6TAT via self-assembly showed strong antimicrobial activity [217]. Biodistribution studies of FITC-loaded nanoparticles in rabbits and efcacy studies in a C. neoformans meningitis rabbit model revealed that these nanoparticles crossed the BBB and produced antimicrobial activity against the pathological strains in the brain tissue with a similar efcacy as amphotericin B, suggesting a therapeutic dose was delivered by TAT containing nanoparticles. Furthermore, these nanoparticles avoided causing the side-effects associated with amphotericin. This study holds importance for TAT-containing nanoparticles as it has proven that it is possible to deliver a therapeutic dose, together with functional agents, via TAT-nanoparticles into the brain for treatment of brain infections and tracking of nanoparticles in vivo, a step closer to the development of a clinically applicable nanocarriers for treatment as well as monitoring meningitis and other brainrelated disorders. Recent evidence showed that TAT can also enhance the delivery of liposomes into the brain. Qin et al. prepared liposomes using cholesterol-PEG2000-TAT (TAT-LIP) and compared them to liposomes fabricated from cholesterol-PEG2000 polymer (LLIP) and conventional cholesterol formulation (LIP) in vitro and in vivo [218]. TAT-LIP accumulated most in the brain (including various regions of the brain) within 24 hr after administration via tail vein, although all were not selectively targeted to the brain. All liposomes showed a uniform distribution across the brain. The study also suggested adsorptive transcytosis could be one of the mechanisms for TAT-LIP transport across the BBB and the positive charge of the TAT-LIP played an important role in enhancing this transport [218]. In addition to CPPs, cationic protein can also enter the brain via an adsorptive-mediated mechanism and Poduslo and Curran demonstrated that polyamine modication of proteins (insulin, albumin and IgG) can dramatically increase the permeability of proteins at the BBB with 1.72.0 fold increase for insulin, 54165 folds for albumin and 111349 fold for IgG in normal adult rats [219]. It is, however, unknown, if this chemical modication may lead to toxicity or immunogenicity problems. In a study reported by Lu et al., cationic bovine serum albumin (CBSA) conjugated PEG-PLA nanoparticles (CBSA-NP) was compared to native PLA bovine serum albumin conjugated nanoparticles (BSA-NP) and CBSA unconjugated PEGylated nanoparticles (NP) in brain transcytosis across the BBB coculture and brain delivery in mice using a uorescent probe [220]. This study conrmed that AMT is the mechanism of brain delivery of CBSA-NP. Increasing the surface density of CBSA conjugated per nanoparticle promoted the transcytosis ability of nanoparticles

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

16

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

while their blood AUC decreased. With the optimized CBSA number conjugated per nanoparticle of 1:10, CBSA-NP achieved the highest % injection dose/g brain by 2.3-fold compared with NP [220]. The same study also reported an accelerated blood clearance phenomena can be induced by rst injection of NP or CSBA-NP or over a period of successive high dose of CBSA-NP, highlighting the importance of the issue and potential related toxicity and immunological responses unique to this type of nanocarrier. AMT enables many poorly brain-penetrating drugs across the BBB, and holds potential for promoting drug delivery into the brain. However, because it is a non-specic process, the adsorptive process will not only occur at the BBB but also in the blood vessels in other organs. This poses a challenge for both achieving therapeutic concentration in the brain and limiting the drug distribution in non-target organ. Toxicity and immunogenicity issues, especially with cationic proteins, should not be overlooked and must be assessed for each system. There are studies that can be carried out to assess both the membrane toxicity and tissue inammation caused by above mentioned CPPs and cationic albumin nanoparticles [220,221]. Some membrane toxicity studies have indicated that cytotoxicity of different CPPs can vary. For instance, TAT is non toxic with concentration up to 100 M, whereas Antp is signicantly more toxic [222]. Furthermore, peptides bound to TAT can trigger signicant and chain length- dependent cytotoxicity when their concentration is above 10 M, irrespective of the sequence of cargo [222]. In addition, a biocompatibility and dose-dependent cytotoxicity study should be carried out. For instance, the basic domain TAT4957 has been used in inhibiting neural death [223] and achieving biological effects by delivering large proteins, such as RNase, domain III of pseudomonas exotoxin A, horseradish peroxidase and -galactosidase [224] into the intracellular space [225]. A very high dose of TAT4660 peptide was reported to be toxic ([226]. The cytotoxicity of TAT-conjugated nano-carrier should be assessed against human brain microvascular endothelial cells and astrocytes. Cell viability could be measured by following a standard MTT assay procedure. The effect of the degree of TAT conjugation should also be investigated and optimized to gain an understanding of the toxicity of various carrier systems and their structure and dose relationship. 5.5. Drug transport across the bloodbrain barrier via endogenous receptor-mediated transcytosis Despite the huge success with some AMT based drug delivery systems, one of the biggest shortcomings of AMT is its lack of selectivity, which potentially can cause side effects of drugs in non-targeted organs. On the other hand, RMT provides an opportunity for active targeting of BBB, particularly when the target receptor is up-graded under the diseased condition, e.g. diphtheria toxin receptor under inammatory disease conditions. Cargo molecules which are associated with targeting ligands can be transported across the BBB via this approach, therefore, RMT is also known as the molecular Trojan horse approach [227,2]. Although this approach was originally applied to molecular or macromolecular cargos [228,229], it has now been extended to transport drug-carrying nanocarriers with the same level of success (see below). The progress in molecular biological and BBB genomics enables the rapid discovery of novel transporters that are expressed at the brain capillary endothelium, potentially a large number of receptors/transporters expressed at the BBB can be utilised for drug delivery across the BBB into the brain. In this section, focus will be on those most studied and potentially most effective systems. In general, there are three steps for RMT: 1) endocytosis at the luminal (blood) side after receptor-ligand binding; 2) movement through the endothelia cytoplasm; 3) exocytosis of the drug or ligand-attached drug or cargo at the abluminal (brain) side [227]. Step two however, may involve endosomal/lysosomal systems,

which potentially can degrade drug molecules, therapeutic protein and peptides and genetic materials. To escape this fate, pH-sensitive liposomes or use of cationic molecules have been applied [230,231]. Some molecules, such as diphtheria toxin, when used as a targeting ligand, have intrinsic lysosomal escape mechanisms [232]. Fortunately, lysosomal escape mechanism is not essential in brain delivery, RMT has been successful in transporting large drug molecules, drugcarrying liposomes, nanoparticles and polymeric complexes to the brain even without it [30,233]. 5.5.1. Insulin receptor The insulin receptor is a large protein having a molecular weight of 300 kDa and is a heterotetramer of two extracellular alpha and two transmembrane beta subunits [30]. Each beta chain contains a tyrosine kinase activity in its cytosolic extension. This is a widely characterised receptor-mediated transcytosis system used for transporting drugs and genes into the brain. Extensive studies of the insulin-receptor and its uses have been carried out by Pardridge and colleagues [234237]. Pardridge's group successfully developed a radiolabelled amyloid-peptide, 125I-Abeta140 conjugated to 8314 monoclonal antibody (mAb) that binds to the human insulin receptor as a diagnostic probe for AD [238]. Recently, the same group tested a genetically engineered human/mouse chimeric form of the human insulin receptor monoclonal antibody (HIRMAb) on an adult anesthetized Rhesus monkey and showed that humanized HIRMAb was rapidly transported into all parts of the primate brain after intravenous administration, suggesting its potential for delivering drug and gene across the BBB in human [239]. However, this approach is still considered as a risky one as it targets one of the most important mechanism involved in glucose homeostasis. 5.5.2. Transferrin receptor The most widely characterised RMT system for drug delivery across the BBB is the TfR, even though the intracellular trafcking of transferrin upon internalization via the TfR has not yet been elucidated. The TfR is a transmembrane glycoprotein consisting of two subunits of 90 kDa linked by a disulde bridge. Each subunit can bind one transferrin molecule [240]. The TfR is not unique to the BBB, it is also expressed on hepatocytes (mostly), erythrocytes, intestinal cells, monocytes in addition to endothelial cells of the BBB. In the brain, the TfR can also be found on choroid plexus epithelial cells and neurons. The TfR mediates cellular uptake of iron bound to transferrin. There are two ways of utilising TfR for transporting drugs into the brain: using endogenous transferrin as targeting ligand or an antibody directed against the TfR (e.g. OX-26). The latter binds to a different site from that of transferrin, therefore, it is less likely to be affected by or interfere with endogenous transferrin. da Cruz et al. [241] reported that cationic liposomes decorated with transferrin resulted in a signicant enhancement of luciferase gene expression activity in C6 glioma cells, primary hippocampal neurons and primary cortical neurons. However, the transfection efciency of this system was low. The use of transferrin as a delivery vector may not be advantageous due to the very high concentration of endogenous transferrin in the circulation, which competes for BBB transferrin binding sites. The likelihood of overdosing with iron, when one tries to displace the endogenous transferrin with exogenously applied transferrin-containing systems, is another limiting factor for TfR in vivo application. To overcome the limitations of using transferrin as the targeting ligand in vivo, antibodies binding to an epitope of the TfR that is different to the transferring binding site, have been developed. OX26 (anti-rat TfR monoclonal antibody), R17-217 and 8D3 (both antimouse TfR monoclonal antibody) have all been examined. When R17217 was compared to 8D3, the brain uptake of R17-217 was relatively low at 1.7% injected dose/g, whereas, 8D3 was 3.1% injected dose/g [242]. However, Lee et al. found that in contrast to 8D3, R17-217 was more selective for the brain and poorly taken up by the liver and kidney

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

17

[242]. Ulbrich et al. studied human serum albumin (HAS) nanoparticles with covalently coupled transferrin or transferrin receptor monoclonal antibodies (OX26 or R17-217) for brain delivery of loperamide, a molecule that is normally unable to cross the BBB on its own [243]. Results showed that signicant anti-nociceptive effects were detected with loperamide-loaded HAS nanoparticles with covalently bound transferrin or the OX20 or R17-217 antibodies following iv injection. A similar efciency in delivery of the drug into the brain was achieved with the three different types of nanoparticles but not with the IgG2a-modied HAS nanoparticles [243], suggesting that OX26 or R17-217 can be a good alternative to transferrin for transporting drugs across the BBB with similar efciency. In a more recent study conducted by Rooy et al. [244], ve different targeting ligands: transferrin, R17-217 (against TfR), COG 133 (against LDLR and LRP), Angioprep-2 (against LRP) and CRM197 (against DTR) were directly compared for their ability to target the liposomes to the brain in vitro and in vivo. Only R17-217 and CRM197 attached liposomes were found to be associated with human endothelia cells in vitro and only R17-217 signicantly enhanced the brain uptake of liposomes in vivo at all time points after tail vein injection to male Balb/c mice [244]. Using the brain capillary depletion method, the authors determined the uptake of 3H-labelled liposomes in brain capillaries and found R17-217 liposomes were 10 times higher than untargeted liposomes. The biodistribution study showed the uptake of R17-217-liposomes was 4.3 times higher than the control 6 h post injection [244]. Interestingly R17-217 liposome is the only one whose concentration in the brain was maintained over the period of 6 h and its dose in the brain at 12 h was 0.18%/g. Authors suggested that the high molecular weight (i.e. longer chain and protrude more from the surface) and high afnity for the receptor may have contributed to its strong brain targeting ability [244]. Although targeting ligand is an important factor in determining the performance of a drug delivery system, other parameters may also inuence the fate of the system, such as the matrix material, particle size, surface properties, the density and conformation of targeting ligand. More studies are needed to fully understand the function and performance of the targeting ligands on different types of nanocarriers. 5.5.3. Low-density lipoprotein receptor related proteins 1 and 2 (LRP1 and LRP2 receptors) LRP1 and 2 are multifunctional, multi-ligand scavenger and signalling receptors. Together, they can interact with a diverse range of molecules and mediators including ApoE, tissue plasminogen activator (tPA), plasminogen activator inhibitor 1 (PAI-1), amyloid precursor protein (APP), lactoferrin, melanotransferrin, 2 macroglobulin (2 M), receptor associated protein (RAP), HIV-1 TAT protein, Heparin cofactor II, heat shock protein 96 (HSP-96) and engineered angiopeps [30,233]. Over the years, the low-density lipoprotein receptor related protein-1 (LRP1) and LRP2 receptors have been exploited to target drugs to the brain in a fashion similar to that used for the transferrin and insulin receptors. There are some successful examples using LRP1 and LRP2 receptors for brain drug transport. Polysorbate 80 coated poly(butylcyanoacrylate) (PBCA) nanoparticles were rst investigated [245] in vivo to enhance the brain transport of hexapeptide dalargin. Results showed that after i.v. injection, the polysorbate 80 coating enabled the highest induction of analgesia in mice. Likewise, other drugs that normally do not penetrate the BBB, including tubocurarine, loperamide, 8-chloro-4-hydroxy-1oxol, 2-dihydropyridazino, quinoline-5-oxide choline salt (MRZ 2/576), and doxorubicin showed higher concentrations in the brain when associated with polysorbate 80-coated nanoparticles [246]. The protein ApoE or B absorption on the nanoparticles, followed by low density lipoprotein (LDL) receptor mediated endocytosis and transcytosis, is believed to facilitate the uptake of the nanoparticles by the brain. Recently Zensi reported that PEGylated albumin nanoparticles with covalently bonded ApoE showed enhanced uptake and intracellular localization in

mouse endothelia cells [247]. More importantly, following administration into the jugular vein of SV129 mice, the ApoE albumin nanoparticles transcytosed into brain parenchyma and were taken into the neurones of the mice, as supported by micrographs of different brain regions taken 30 min post injection [248]. This study is one of a few investigations used Transmission Electron Microscopy (TEM) to provide direct high quality visual evidence of transcytosis of ApoE albumin nanoparticles both in vitro and in vivo. Lactoferrin (Lf) is a mammalian cationic iron-binding glycoprotein belonging to the transferrin family. It is an interesting molecule as it has been implicated in the pathogenesis of brain lesions. An elevated level of Lf has been found to be associated with normal ageing and to be more pronounced in people with AD, Guamanian cases, Pick's disease, and Down syndrome [249]. Lf was reported to be transported into the brain via LRP mediated transcytosis by some researchers [250] and others showed evidence to support that there are Lf receptors on brain capillary endothelia cells in mouse at least [251]. Fillebeen et al. demonstrated in a bovine brain capillary endothelial cell model that when the inammatory mediator TNF- was present, Lf transport in vitro through the brain capillary endothelia cells was markedly decreased [82]. This suggests that the transport of Lf could be different under the pathological condition compared to that of the normal condition. The reduction in Lf transport under the inammatory condition may work in favour for drug transport as more Lf receptors would be free for exogenous Lf from the drug delivery systems. Interestingly, Ji et al. demonstrated that the brain uptake of Lf in rats was much greater than that of transferrin and OX26 [252]. When Lf-conjugated PEG-PLA nanoparticles were administrated to mice intravenously, there was a 3-fold increase in brain uptake of Lf-nanoparticles compared to un-conjugated nanoparticles [253]. The cell viability study also indicated that it was non-toxic [253]. Lately, the same group assessed biodistribution of coumarin-6loaded Lf-conjugated PEG-PLA nanoparticles in mice and therapeutic efcacy of urocortin-loaded Lf nanoparticles on a 6-OHDA rat model of Parkinson's disease [254]. Their data showed 2.5 times increase in AUC by Lf nanoparticles compared to the un-conjugated nanoparticles in 24 h. Drug-loaded Lf- nanoparticles were successfully taken up by the brain and produced therapeutic efcacy, as demonstrated by the signicant attenuation of striatum lesion despite the drug loading being only 0.280.33% [255]. This compelling evidence suggests that Lf-nanoparticles could be a promising drug delivery system for treatment of Parkinson's disease if their safety in human can be conrmed. Another interesting targeting molecule is melanotransferrin (P97) which has been reported to undergo active transcytosis, possibly via the LRP1 receptor [256]. It was revealed that recombinant human melanotransferrin (P97) was readily taken up by mouse brain following intravenous injection and in situ brain perfusion [256]. This P97 transcytosis across the bovine brain capillary endothelial cell monolayers was at least 14-fold higher than that of transferrin, with no apparent intra-endothelial degradation. When the effectiveness of P97 as a brain targeting ligand was tested in a mouse model, P97 and P97-adrimycin conjugates showed 68-fold higher than that of BSA or lactoferrin in terms of transport into the brain. More importantly their efcacy against intracranial rat C6 glioma and human ZR-75-1 mammary tumours in athymic mice was very signicant with improved survival rates compared to free adriamycin [257]. This technology has now been patented worldwide and the technology (NeroTrans TM transporter platform) is the proprietary property of Raptor Pharmaceutical Corp (Novato, CA). In June 2009, Raptor and Roche entered a collaboration and licensing agreement to evaluate the delivery of Roche's investigative molecules attached to Raptor's proprietary NeuroTrans transporter platform [258]. Another group of LRP ligands, known as angiopeps, has also been reported as a highly effective BBB targeting ligand. Angiopeps belong

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

18

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

to a family of peptides derived from Kunitz domains of aprotinin and other human proteins. They are known to have afnity for LRP receptors [259]. The most studied is angiopep 2 (TFFYGGSRGKRNNFKTEEY, molecular weight 2.4 kDa) which has shown greater transcytosis capacity and parenchymal accumulation than transferrin, lactoferrin, and avidin [260]. Moreover, their ability in efciently facilitating nanocarrier transport across the BBB in vivo has been conrmed with dendrimers [261] and more recently with amphotericin B-loaded polymeric micelles [262]. Chemical conjugation of angiopep 2 with 3 molecules of paclitaxel (ANG1005) was shown to be particularly effective in enhancing drug uptake into the brain, with an 86-fold increase compared to the free drug using an in situ rat brain perfusion model. It also exceeded the free drug by 454-fold following i.v injection in mice bearing metastases of breast cancer [263]. A therapeutic amount of ANG1005 was able to cross the BBB, as evidenced by the increased survival rates of mice with implanted tumour cells [264]. It is interesting to note an innovative approach reported recently by Tosi and colleagues. They developed poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles that are surface modied with two ligands: one is a BBB-penetrating peptide (similopioid peptide, g7) for transporting across the BBB, another is a sialic acid residue (SA) for the interaction with receptors in the brain tissue to prolong the nanoparticle residence time in the brain parenchyma [265]. The researchers reported a remarkably high dose (6% injected dose) in the CNS over a prolonged period of time (24 h). Surprisingly when the nanoparticles were conjugated with only g7 molecules, the brain uptake of nanoparticles increased to 14% injected dose/g at 1.5 h post injection via the tail vein, which was more than twice that of the nanoparticles with double targeting ligands. The g7-nanoparticles, however, had a much shorter opioid effect (5 h). With an additional SA ligand, the central opioid activity of loperamide increased to 24 h [265]. These remarkable results were attributed to the ability of SA g7 nanoparticles to cross the BBB and remain within the brain parenchyma. The unusually high brain uptake of g7 nanoparticles suggests that g7 is a promising BBB targeting ligand on its own. Finally, Tosi's data also reveals that the clearance of nanoparticles in the brain parenchyma is very rapid without SA ligand and we speculate that this removal of nanoparticles is more likely due to physical translocation rather than enzyme degradation of nanoparticles.

both diagnosis and treatment to be delivered at the same time by a nanocarrier system conjugated to the CRM197. 5.6. Drug transport across the bloodbrain barrier via inhibition of efux Pumps Efux pumps such as P-gp and MRPs can prevent many drugs from entering and accumulating in the brain. Evidence shows that P-gp excludes a number of lipophilic compounds that may be structurally unrelated, from cerebral endothelial cells [271]. Many MRP substrates are amphiphilic anions with at least one negatively charged group, although MRP can also transport cationic and neutral compounds. To circumvent this blockade, one strategy is to coadminister the drug with a pharmacological modulator which inhibits efux transport systems in brain capillary endothelia cells. The concept of using of Pluronic block (or poloxamers) copolymers to inhibit the P-gp efux pump was derived from an early study conducted in Kabanov laboratory by Miller et al. [272]. The study examined cellular accumulation of rhodamine 123, a selective P-gp substrate in bovine brain microvessel endothelial cell monolayers, with and without the presence of Pluronic P85. Extensive studies have now conrmed that Pluronic P85, a Pluronic block copolymer, can be used not only as an effective efux pump inhibitor but also as a drug delivery vehicle [273]. Pluronic block copolymers are simple yet unique. They consist of hydrophilic ethylene oxide (EO) and hydrophobic propylene oxide (PO) blocks. EO and PO are arranged in a basic A-BA tri-block structure. This arrangement gives rise to their amphiphilic character. The PO/EO ratio determines the hydrophobic- hydrophilic balance (HLB) of the copolymers. Due to their amphiphilic nature, these copolymers exhibit the properties of surfactants, including the ability to accumulate and interact with hydrophobic surfaces and biological membranes. When their concentration in water is above the critical micelle concentration (CMC), they self-assemble into micelles. The most studied Pluronic P85 showed the ability to enhance the BBB permeability of a wide range of drugs, including doxorubicin, etoposide, taxol, 3-azido-3-deoxythymidine, valproic acid and loperamide, in the bovine brain microvessel endothelia cell monolayer [274]. Evidence suggests that the inhibition mechanisms of Pluronic block copolymers on P-gp activity in the BBB involve 1) copolymer interaction with the cell membrane, producing a membrane uidization effect; 2) inhibition of P-gp ATPase activity; 3) depletion of cellular ATP [273]. Batrakova et al. demonstrated in their study using bovine brain microvessel endothelial cells that both HLB and PO block length are important determinants for the P-gp inhibition activities of Pluronic block copolymers [275]. They conclude that lipophilic Pluronic with intermediate length of PO (3060 units) and HLB b20 are most effective at inhibiting the P-gp efux in brain microvessel endothelia cells. The effect of Pluronic P85 on facilitating drug transport across the BBB evaluated in vitro and in vivo [276] demonstrated that coadministration of 1% Pluronic P85 increased digoxin penetration of the brain by threefold in vivo. Further studies showed that Pluronic P85 also inhibits MRP1 and MRP2 ATPases but to a lesser extent [277]. One of the interesting studies conducted by Batrakova et al. [278] showed that when Pluronic P85 was used at high concentration (>CMC), the model drug, rhodamine 123 (R123) would be solubilised inside the micelles, enter the cell and then be recycled back with a net decrease in R123 transport across the cell monolayer. In contrast, at a concentration below CMC, Pluronic P85 increased BBB permeability of R123 in vitro. However, after Pluronic P85 was conjugated to insulin, the Pluronic P85 micelles enhanced permeability of R123, suggesting that modied micelles might pass through brain microvessel endothelia cells via insulin RMT [278]. This has important implications for the use of Pluronic P85 in drug delivery systems.

5.5.4. Diphtheria toxin receptor (DTR) DTR is also known as transmembrane HB-EGF. Uniquely, it has no endogenous ligand. Although diphtheria toxin (DT) can bind to this receptor and enter the cell via endocytosis [266], DT is too toxic to be used in vivo. Cross reacting material (CRM 197) is a mutated form of DT but without the enzymatic activity that would cause the toxicity seen in DT. Because it is non-toxic, yet, retains its ability to bind to DTR [267], it makes CRM197 a useful and attractive targeting ligand. In fact, CRM 197 has been used as a safe and effective carrier protein for human vaccines for a long time and has been tested as a therapeutic protein for cancer treatment in a clinical trial [268,269]. CRM 197 uses DTR as its transport receptor and delivers its cargo molecules across the BBB by receptor mediated-transcytosis [99]. It has been shown that CRM197 can achieve brain targeting in vitro and in vivo [270,99]. One interesting feature of DTR is that it is strongly up-regulated under inammatory conditions [99], which occur often with CNS diseases such as AD, Parkinson's disease, MS, ischemia, encephalitis, epilepsy, tumour and lysosomal storage disease. This up-regulation of DTR has already been seen following seizures [100]. This provides opportunities for disease-induced targeting and consequently may enhance the transport of the drug across the BBB and maximise the therapeutic efcacy of the drug in the brain. If CRM197 is attached to a cargo of diagnostic agent, then it could be used to image the inammatory condition in patients. Therefore, it is possible to allow

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

19

These studies have profound implications as they suggest that polymers are not inert; that they may have important biological activity which could be benecial or detrimental to a drug therapy if they are used as pharmaceutical excipients or as drug delivery vehicles. Because the action of Pluronic block copolymers involves interaction with cell membrane and direct inhibition of ATP activity, a concern was raised about the potential toxicity of the system on the BBB. Several pieces of evidence, however, show that ATP depletion by Pluronic P85 was transient and that there was no compromise of BBB integrity after ATP depletion as demonstrated by the fact that there was no change in permeability of mannitol (a permeability marker) both in vitro and in vivo [274,276]. In addition to their application in enhancing drug transport into the BBB, Pluronic block copolymers have been used to enhance activity of anticancer drugs such as doxorubicin due to its ability to chemosensitize MDR tumours by inhibiting P-gp and enhancing pro-apoptotic signalling in cancer cells [279]. A Pluronic -based micelle formulation of doxorubicin (SP1049C) has now entered Phase III clinical trial and received orphan drug status from the FDA in 2008. It is possible that following the completion of the clinical trial, this formulation can also be adopted for transporting CNS drugs, possibly in combination with a nanocarrier system and BBB targeting ligands. Besides Pluronic block copolymers, liposomes, nanoparticles and even macromolecular conjugates can also be used to bypass P-gp and MDR efux pumps. Potentially, Pluronic copolymers could be used in the formulation of all these types of drug delivery systems, providing additional function as a biological response modier [76]. No doubt, this will be a sensitive research topic as we are moving into areas of using polymers with certain biological activities. Caution has to be taken. It is acceptable for the delivery of cancer treatment but it may not be suitable for treatment of chronic diseases that require long term therapy. 6. Cell-mediated drug transport across the bloodbrain barrier Cell-mediated drug transport employs specic cells that take up drug-loaded nano or microcarriers trafc them through the BBB and deliver the drugs to their target sites inside the brain. In this case, cells act as Trojan horses. The rationale for cell-mediated trafcking of drug across the blood brain barrier is based on the following: 1) During the process of inammation in the brain (commonly occurring in AD, Parkinson's disease, MS, stroke, brain tumours and HIV-1 associated dementia) there is extensive recruitment and trafcking of leukocytes (monocytes and neutrophils) in the brain. Mononuclear phagocytes (MP) and T cells would migrate towards the site of inammation involving the processes known as diapedesis and chemotaxis. 2) Cells such as macrophages and monocytes/neutrophils are phagocytic and have a tendency to endocytose colloidal materials, for example, nano or microparticles, liposomes and subsequent exocytosis to release drug and/or colloidal materials to external media [280282]. 3) A high payload of drug can be incorporated/loaded into nanocarriers or microcarriers, then taken up by Trojan horse cells. 4) Cell-mediated transport can be combined with magnetic particles and application of a magnetic eld to further enhance the brain delivery and provide the diagnosis of inammation sites and tracking of nanomaterials. 5) Potentially block one of the routes that pathogens may take for invasion of the brain. Compared to other transport pathways, cell-mediated drug delivery has attracted far less attention for brain drug transport but there have been some very promising results reported. Most noteworthy is the work carried out by Jain et al. [25] on RGD-anchored magnetic

liposomes for monocytes/neutrophils-mediated brain targeting using an IL-1 induced brain inammation rat model. In this study, the authors used RGD as the targeting ligand for integrin receptors expressed on neutrophils and monocytes to facilitate cell uptake of liposomes containing the anti-inammatory drug diclofenac. A magnetic eld of 8.0 kG strength was applied near the brain of rats receiving the treatment. The uptake of drug/liposomes by cells was improved with RGD modication, increasing to about 16%. The cell sorting study conducted on the blood samples collected from the animal study showed RGD improved both the monocyte and neutrophil count, by 6% and 20% respectively. The most striking result is that by the incorporation of magnetic particles and application of a magnetic eld, the percentage drug dose that reached the brain was elevated from 3.25% to 21.53% for RGD-modied magnetic liposomes and from 1.86% to 14.11% for unmodied magnetic liposomes. It is a huge success for BBB targeting. Moreover, the RGD-modied magnetic liposomes also dramatically reduced the liver uptake of the drug from 51.43% down to 26.58%; and the unmodied 47.32% down to 29.68%. This reduction of liver uptake is just as important as the improvement in brain targeting as it will potentially reduce the side effects in liver tissues. Furthermore, the RGD-modied magnetic liposomes also halved the dose that usually reaches the spleen. In terms of brain accumulation, RGD-modied magnetic liposomes was found to be 9.1 times of the free drug and 6.6 times of nonmagnetic RGD liposomes and 1.5 times of non-RGD magnetic liposomes [25] These results indicate that the guidance of an external magnetic eld augments the active targeting of drug to the brain as well as the recruitment of leukocytes at the site of inammation. If this targeting effect can be translated into drug efcacy without toxicity, then this combined strategy will revolutionize the treatment of all CNS diseases which have an inammatory component. The potential level of toxicity arising from repeated dosing of magnetic materials must be addressed before this approach can be developed into a clinical treatment. It is noteworthy that the vesicle size used in the above study was large, at 1.2 m. It is assumed this is to allow the incorporation of both drug and magnetic particles. Nevertheless, this also indicates that the cell-mediated approach does not require nano-sized carriers, a feature very different from that of all other BBB targeting approaches. In fact, a relatively large size may be necessary to allow a sufcient number of magnetic particles to be loaded into liposomes and later guided to the target. However, the toxicity of these vesicles needs to be assessed. Previously it has been reported that if the vesicle size is greater than 1 m this could cause adverse effects after accumulation in the lungs, causing thrombosis [283]. Qin et al. have reported a similar RGD-liposome approach for the delivery of ferulic acid to monocytes/neutrophils in brain [284] using the same inammatory animal model as described by Jain et al. [25]. The results of the study showed that 72% of RGD-liposomes (152 nm) associated with leukocytes (monocytes and neutrophils) while only 19.8% for plain liposomes (155 nm). Despite the liposomes being non-magnetic, drug-loaded RGD-liposomes reached the brain at a level 6-fold higher than that of the drug solution and 3-folds higher than that of the drug-loaded plain liposomes, indicating that the strategy of targeting leukocytes for brain delivery of drug worked. Furthermore, the drug concentration in the brain appeared to have dropped little over the period of 120 min. However, as expected, there was a very high level of drug in the liver and the spleen. As the data was reported as the concentration of drug in organ (g/g), it is impossible to compare this data with others. In contrast to the previous two studies, Afergan et al. used conventional negatively charged liposomes without a targeting ligand to deliver serotonin to the brain via monocytes [117]. Endocytosis of liposome by monocytes was found to be 60%, and 28.5% by granulocytes. Four hours post i.v. injection, drug concentration in the brain was found to be 0.138% and 0.068% of dosage for drug-loaded

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

20

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

liposomes and free drug solution, respectively. The majority of the injected dose was in the spleen, liver, blood and lungs. This study was designed elegantly using 3H-liposomes and 14C-serotonin for co-localization imaging, which conrmed that liposomes reach the brain intact. Furthermore, the group studied the liposome distribution following injection of liposomal aldendronate (a monocyte depletion agent) and found that the treatment with liposomal alendronate labelled with a uorescent probe resulted in no uorescence in brain tissue which indirectly demonstrated that the circulating monocytes are the carriers of liposomes to the brain [117]. Nevertheless, it is noted that the group used healthy rabbits and rats for brain penetration and body distribution studies which would not give an opportunity for recruitment of monocytes in the brain region. This may explain why the results obtained in this study are so different compared to previous groups'. Indirectly it demonstrates that brain targeting by cell-mediated transport is most suitable for treating brain diseases under inammation. Research has shown that both endocytosis and exocytosis process can be modulated. For instance, Panyam showed that the dynamics of endocytosis and exocytosis of PLGA nanoparticles in cells can be inuenced by concentration, time and energy [281]. Endocytosis is much faster than exocytosis and exocytosis can be almost completely inhibited by depletion of serum in the medium [281]. In the study of liposome uptake by human peripheral blood monocytes, Mehta et al. reported negatively charged liposomes have a more rapid uptake rate than positively charged liposomes (3-fold increase) and neutral liposomes (5-fold increase). The drug is released slowly from the cells after endocytosis [285]. This suggests that by surface modication of the nanocarrier, their uptake into and release from cells can be modulated. Bender et al. reported the use of nanoparticles to deliver HIV protease inhibitor saquinavir into human monocytes/macrophages [286]. An in vitro test showed that the nanoparticle formulation is more potent than the free drug. It improved the delivery of antiviral agents to mononuclear phagocyte systems. It is noteworthy that this eld research has up to now focused more on delivery of immunomodulators for activation of cells [287,288], and on the delivery of magnetic particles for diagnosis purposes [289]. This approach does have potential as it can virtually deliver any types of drugs to the brain; whether hydrophilic, hydrophobic, large, small, proteins, peptides or gene constructs. In addition, because it targets monocytes/macrophages, it will also be effective for delivering drugs against pathogens which infect leukocytes. As previously shown by Jain et al. [25], the combined strategy of magnetic liposomes and monocytes targeting ligands can maximise the monocytes' recruitment at the brain and may prove to be a very effective approach for drug delivery to the diseased brain which has an inammation component. However, the safety of this route has yet to be fully assessed. It is unknown whether its combination with the use of magnetic particles may lead to any iron toxicity in long term use. Furthermore, the selectivity and maximum capacity of this strategy for delivery of drug across the BBB warrants further study. 7. Conclusions and remarks In the past decade, we have seen tremendous attention and effort focused on the development of modern and novel drug delivery systems to circumvent the BBB. This is due to the signicant challenge faced by industry, government and academics in seeking effective drug therapies for the increasing incidence of brain disease associated with an ageing population. In this review, we looked at the barrier issue from a biological and pathological perspective to provide a better insight to the challenges and opportunities associated with the BBB. We need to remind ourselves that we are developing drug delivery systems which ultimately will transport a drug to a diseased

brain, not a healthy one. The delivery system, including the targeting ligands should be designed based on knowledge of the diseased BBB. Unfortunately most of the models used in the literature for assessing the biodistribution or pharmacokinetics of drug delivery systems are healthy mice, rats and rabbits. They could be very different to the diseased brain, therefore the data generated on healthy animals sometimes could be inaccurate or even misleading. Our rst conclusion is therefore that in vivo models that reect the true diseases should be adopted to evaluate a drug delivery system adequately. The urgent need of standardised diseased animal models that are accessible to all must be considered and met to advance the research and development in drug transport across the BBB. To successfully target the brain, the selectivity of a BBB receptor is extremely critical. This means, ideally, the receptor should be brain specic, or at least, preferentially expressed at the BBB. Unfortunately almost all the receptors are nearly non-specic as shown by percentage dose reached the brain compared to that reached by the liver, spleen or lung. To date the best targeting in the BBB we have seen is 21% (of administrated dose) in rat and 26.6% in liver by Jain et al. [25] achieved via cell-mediated transport using RGD-modied magnetic liposomes under magnetic guidance, which takes advantages of immune-response occurred at the BBB under inammatory conditions. We believe that the reason for this high level of targeting is the design of the delivery system that is based on the pathological condition of the BBB and the fact that the system was tested on a diseased model. The targeting effect was greatly enhanced further by its combination with the guidance of an external magnetic eld. In contrast, while RMT worked alone, the best brain targeting, as far as authors are aware, was achieved by g7 molecule which was characterised as a high but short lived targeting (14% at peak) [265]. Unfortunately the majority only achieved 14% [290] as a result of poor selectivity and low BBB permeability. Although the amount reaching the brain may be sufcient for a therapeutic effect, the loss of 9699% is huge from the cost point of view [290], in addition to the potential sideeffects and toxicity that can be caused by 96% of drugs. Jones and Shusta [290] suggest the solution may be in combinatory antibody library technology which allows the development of BBB-specic RMT. Tosi et al. showed that the application of double targeting ligands can provide added targeting benet [265]. Therefore our second conclusion is that use of multiple targeting ligands developed with consideration of the pathological conditions of the disease will maximise success. Ideally the treatment for a brain disease should be constantly monitored and modulated. The idea of using magnetic or bubblelled and drug-loaded nanocarriers, in combination with MRI and ultrasound, may provide a solution for achieving both the diagnosis and treatment purposes. This requires cross-discipline fertilisation of innovative ideas and multi-discipline approach to the design, characterisation and evaluation of the systems. Our third conclusion, therefore, is to take the multidiscipline approach with innovative ideas to ultimately achieve the goal of delivering both diagnosis and drug therapy, selectively and efciently, across the BBB. Our nal conclusion is that any brain-targeted delivery systems must be assessed for their safety, risk and benet for patients. Currently the safety issue has been largely overlooked during the research stage, yet this issue will become critical when the drug to be delivered is for a long term therapy. It is extremely important that any delivery systems developed should have no signicant impact, short or long term, on the functions of the brain.

Acknowledgements The authors wish to thank Dr Heather Benson for reviewing and proofreading the manuscript; Sonya Chu for preparation of the graphic abstract; Michelle Robert-Libia for drawing Figs. 1 and 2.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx

21

References
[1] E. Neuwelt, N.J. Abbott, L. Abrey, W.A. Banks, B. Blakley, T. Davis, B. Engelhardt, P. Grammas, M. Nedergaard, J. Nutt, W. Pardridge, G.A. Rosenberg, Q. Smith, L.R. Drewes, Strategies to advance translational research into brain barriers, Lancet Neurol. 7 (2008) 8496. [2] W.M. Pardridge, Bloodbrain barrier drug targeting: the future of brain drug development, Mol. Interv. 3 (2003) 90105 151. [3] J. Rip, G.J. Schenk, A.G. de Boer, Differential receptor-mediated drug targeting to the diseased brain, Expert Opin. Drug Deliv. 6 (2009) 227237. [4] S.A. Pathan, Z. Iqbal, S.M. Zaidi, S. Talegaonkar, D. Vohra, G.K. Jain, A. Azeem, N. Jain, J.R. Lalani, R.K. Khar, F.J. Ahmad, CNS drug delivery systems: novel approaches, Recent Pat. Drug Deliv. Formul. 3 (2009) 7189. [5] B.T. Hawkins, R.D. Egleton, Pathophysiology of the bloodbrain barrier: animal models and methods, Curr. Top. Dev. Biol. 80 (2008) 277309. [6] P.A. Stewart, Endothelial vesicles in the bloodbrain barrier: are they related to permeability? Cell. Mol. Neurobiol. 20 (2000) 149163. [7] N.J. Abbott, Dynamics of CNS barriers: evolution, differentiation, and modulation, Cell. Mol. Neurobiol. 25 (2005) 523. [8] W.H. Oldendorf, M.E. Cornford, W.J. Brown, The large apparent work capability of the bloodbrain barrier: a study of the mitochondrial content of capillary endothelial cells in brain and other tissues of the rat, Ann. Neurol. 1 (1977) 409417. [9] B.T. Hawkins, T.P. Davis, The bloodbrain barrier/neurovascular unit in health and disease, Pharmacol. Rev. 57 (2005) 173185. [10] Y. Persidsky, S.H. Ramirez, J. Haorah, G.D. Kanmogne, Bloodbrain barrier: structural components and function under physiologic and pathologic conditions, J. Neuroimmune Pharmacol. 1 (2006) 223236. [11] N.J. Abbott, L. Ronnback, E. Hansson, Astrocyte-endothelial interactions at the bloodbrain barrier, Nat. Rev. Neurosci. 7 (2006) 4153. [12] M. Ramsauer, J. Kunz, D. Krause, R. Dermietzel, Regulation of a bloodbrain barrier-specic enzyme expressed by cerebral pericytes (pericytic aminopeptidase N/pAPN) under cell culture conditions, J. Cereb. Blood Flow Metab. 18 (1998) 12701281. [13] M. Ramsauer, D. Krause, R. Dermietzel, Angiogenesis of the bloodbrain barrier in vitro and the function of cerebral pericytes, FASEB J. 16 (2002) 12741276. [14] S. Dohgu, F. Takata, A. Yamauchi, S. Nakagawa, T. Egawa, M. Naito, T. Tsuruo, Y. Sawada, M. Niwa, Y. Kataoka, Brain pericytes contribute to the induction and upregulation of bloodbrain barrier functions through transforming growth factorbeta production, Brain Res. 1038 (2005) 208215. [15] H. Wekerle, Immune protection of the brain-efcient and delicate, J. Infect. Dis. 186 (Suppl 2) (2002) S140S144. [16] R. Daneman, M. Rescigno, The gut immune barrier and the bloodbrain barrier: are they so different? Immunity 31 (2009) 722735. [17] K. Williams, X. Alvarez, A.A. Lackner, Central nervous system perivascular cells are immunoregulatory cells that connect the CNS with the peripheral immune system, Glia 36 (2001) 156164. [18] W.J. Streit, J.R. Conde, S.E. Fendrick, B.E. Flanary, C.L. Mariani, Role of microglia in the central nervous system's immune response, Neurol. Res. 27 (2005) 685691. [19] W.M. Pardridge, Molecular biology of the bloodbrain barrier, Mol. Biotechnol. 30 (2005) 5770. [20] N.J. Abbott, I.A. Romero, Transporting therapeutics across the bloodbrain barrier, Mol. Med. Today 2 (1996) 106113. [21] G. Lee, S. Dallas, M. Hong, R. Bendayan, Drug transporters in the central nervous system: brain barriers and brain parenchyma considerations, Pharmacol. Rev. 53 (2001) 569596. [22] E.M. Kemper, W. Boogerd, I. Thuis, J.H. Beijnen, O. van Tellingen, Modulation of the bloodbrain barrier in oncology: therapeutic opportunities for the treatment of brain tumours? Cancer Treat. Rev. 30 (2004) 415423. [23] W.M. Pardridge, Drug and gene targeting to the brain via bloodbrain barrier receptor-mediated transport systems, Int. Congr. Ser. 1277 (2005) 4962. [24] F. Herve, N. Ghinea, J.M. Scherrmann, CNS delivery via adsorptive transcytosis, AAPS J. 10 (2008) 455472. [25] S. Jain, V. Mishra, P. Singh, P.K. Dubey, D.K. Saraf, S.P. Vyas, RGD-anchored magnetic liposomes for monocytes/neutrophils-mediated brain targeting, Int. J. Pharm. 261 (2003) 4355. [26] F. Chretien, O. Lortholary, I. Kansau, S. Neuville, F. Gray, F. Dromer, Pathogenesis of cerebral Cryptococcus neoformans infection after fungemia, J. Infect. Dis. 186 (2002) 522530. [27] F. Gonzalez-Scarano, J. Martin-Garcia, The neuropathogenesis of AIDS, Nat. Rev. Immunol. 5 (2005) 6981. [28] K. Park, Trojan monocytes for improved drug delivery to the brain, J. Control. Release 132 (2008) 75. [29] Y. Chen, G. Dalwadi, H. Benson, Drug delivery across the bloodbrain barrier, Curr. Drug Deliv. 1 (2004) 361376. [30] A.G. de Boer, P.J. Gaillard, Drug targeting to the brain, Annu. Rev. Pharmacol. Toxicol. 47 (2007) 323355. [31] F.L. Cardoso, D. Brites, M.A. Brito, Looking at the bloodbrain barrier: molecular anatomy and possible investigation approaches, Brain Res. Rev. 64 (2010) 328363. [32] H. Wolburg, A. Lippoldt, Tight junctions of the bloodbrain barrier: development, composition and regulation, Vascul. Pharmacol. 38 (2002) 323337. [33] J. Bernacki, A. Dobrowolska, K. Nierwinska, A. Malecki, Physiology and pharmacological role of the bloodbrain barrier, Pharmacol. Rep. 60 (2008) 600622.

[34] J.D. Huber, R.D. Egleton, T.P. Davis, Molecular physiology and pathophysiology of tight junctions in the bloodbrain barrier, Trends Neurosci. 24 (2001) 719725. [35] M.A. Petty, E.H. Lo, Junctional complexes of the bloodbrain barrier: permeability changes in neuroinammation, Prog. Neurobiol. 68 (2002) 311323. [36] A.S. Yu, K.M. McCarthy, S.A. Francis, J.M. McCormack, J. Lai, R.A. Rogers, R.D. Lynch, E.E. Schneeberger, Knockdown of occludin expression leads to diverse phenotypic alterations in epithelial cells, Am. J. Physiol. Cell Physiol. 288 (2005) C1231C1241. [37] T. Nitta, M. Hata, S. Gotoh, Y. Seo, H. Sasaki, N. Hashimoto, M. Furuse, S. Tsukita, Size-selective loosening of the bloodbrain barrier in claudin-5-decient mice, J. Cell Biol. 161 (2003) 653660. [38] R.D. Fontijn, J. Rohlena, J. van Marle, H. Pannekoek, A.J. Horrevoets, Limited contribution of claudin-5-dependent tight junction strands to endothelial barrier function, Eur. J. Cell Biol. 85 (2006) 11311144. [39] T. Soma, H. Chiba, Y. Kato-Mori, T. Wada, T. Yamashita, T. Kojima, N. Sawada, Thr (207) of claudin-5 is involved in size-selective loosening of the endothelial barrier by cyclic AMP, Exp. Cell Res. 300 (2004) 202212. [40] K. Matter, M.S. Balda, Holey barrier: claudins and the regulation of brain endothelial permeability, J. Cell Biol. 161 (2003) 459460. [41] B.V. Zlokovic, The bloodbrain barrier in health and chronic neurodegenerative disorders, Neuron 57 (2008) 178201. [42] A.W. Vorbrodt, D.H. Dobrogowska, Molecular anatomy of interendothelial junctions in human bloodbrain barrier microvessels, Folia Histochem. Cytobiol. 42 (2004) 6775. [43] J. Bennett, J. Basivireddy, A. Kollar, K.E. Biron, P. Reickmann, W.A. Jefferies, S. McQuaid, Bloodbrain barrier disruption and enhanced vascular permeability in the multiple sclerosis model EAE, J. Neuroimmunol. 229 (2010) 180191. [44] B.D. Cook, G. Ferrari, G. Pintucci, P. Mignatti, TGF-beta1 induces rearrangement of FLK-1-VE-cadherin-beta-catenin complex at the adherens junction through VEGF-mediated signaling, J. Cell. Biochem. 105 (2008) 13671373. [45] J.C. Lim, K.D. Kania, H. Wijesuriya, S. Chawla, J.K. Sethi, L. Pulaski, I.A. Romero, P.O. Couraud, B.B. Weksler, S.B. Hladky, M.A. Barrand, Activation of beta-catenin signalling by GSK-3 inhibition increases p-glycoprotein expression in brain endothelial cells, J. Neurochem. 106 (2008) 18551865. [46] A.W. Vorbrodt, S. Li, W.T. Brown, N. Ramakrishna, Increased expression of betacatenin in brain microvessels of a segmentally trisomic (Ts65Dn) mouse model of Down syndrome, Brain Cell Biol. 36 (2008) 203211. [47] M.A. Deli, Potential use of tight junction modulators to reversibly open membranous barriers and improve drug delivery, Biochim. Biophys. Acta 1788 (2009) 892910. [48] S.M. Stamatovic, R.F. Keep, A.V. Andjelkovic, Brain endothelial cellcell junctions: how to open the blood brain barrier, Curr. Neuropharmacol. 6 (2008) 179192. [49] N.S. Harhaj, E.A. Felinski, E.B. Wolpert, J.M. Sundstrom, T.W. Gardner, D.A. Antonetti, VEGF activation of protein kinase C stimulates occludin phosphorylation and contributes to endothelial permeability, Invest. Ophthalmol. Vis. Sci. 47 (2006) 51065115. [50] H. Clarke, A.P. Soler, J.M. Mullin, Protein kinase C activation leads to dephosphorylation of occludin and tight junction permeability increase in LLC-PK1 epithelial cell sheets, J. Cell Sci. 113 (Pt 18) (2000) 31873196. [51] M.D. Potter, S. Barbero, D.A. Cheresh, Tyrosine phosphorylation of VE-cadherin prevents binding of p120- and beta-catenin and maintains the cellular mesenchymal state, J. Biol. Chem. 280 (2005) 3190631912. [52] J. Haorah, S.H. Ramirez, K. Schall, D. Smith, R. Pandya, Y. Persidsky, Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to bloodbrain barrier dysfunction, J. Neurochem. 101 (2007) 566576. [53] C. Lohmann, M. Krischke, J. Wegener, H.J. Galla, Tyrosine phosphatase inhibition induces loss of bloodbrain barrier integrity by matrix metalloproteinasedependent and -independent pathways, Brain Res. 995 (2004) 184196. [54] P. Kumar, Q. Shen, C.D. Pivetti, E.S. Lee, M.H. Wu, S.Y. Yuan, Molecular mechanisms of endothelial hyperpermeability: implications in inammation, Expert Rev. Mol. Med. 11 (2009) e19. [55] L. Gonzalez-Mariscal, R. Tapia, D. Chamorro, Crosstalk of tight junction components with signaling pathways, Biochim. Biophys. Acta 1778 (2008) 729756. [56] T. Hirase, S. Kawashima, E.Y. Wong, T. Ueyama, Y. Rikitake, S. Tsukita, M. Yokoyama, J.M. Staddon, Regulation of tight junction permeability and occludin phosphorylation by Rhoa-p160ROCK-dependent and -independent mechanisms, J. Biol. Chem. 276 (2001) 1042310431. [57] M.A. Deli, C.S. Abraham, Y. Kataoka, M. Niwa, Permeability studies on in vitro bloodbrain barrier models: physiology, pathology, and pharmacology, Cell. Mol. Neurobiol. 25 (2005) 59127. [58] S.Y. Yuan, Protein kinase signaling in the modulation of microvascular permeability, Vascul. Pharmacol. 39 (2002) 213223. [59] P.M. Watson, J.M. Anderson, C.M. Vanltallie, S.R. Doctrow, The tight-junctionspecic protein ZO-1 is a component of the human and rat bloodbrain barriers, Neurosci. Lett. 129 (1991) 610. [60] X.K. Tu, W.Z. Yang, S.S. Shi, Y. Chen, C.H. Wang, C.M. Chen, Z. Chen, Baicalin inhibits TLR2/4 signaling pathway in rat brain following permanent cerebral ischemia, Inammation 34 (2011) 463470. [61] J.D. Huber, K.A. Witt, S. Hom, R.D. Egleton, K.S. Mark, T.P. Davis, Inammatory pain alters bloodbrain barrier permeability and tight junctional protein expression, Am. J. Physiol. Heart Circ. Physiol. 280 (2001) H1241H1248. [62] C.L. Willis, L. Leach, G.J. Clarke, C.C. Nolan, D.E. Ray, Reversible disruption of tight junction complexes in the rat bloodbrain barrier, following transitory focal astrocyte loss, Glia 48 (2004) 113.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

22

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx [93] B. Dehouck, L. Fenart, M.P. Dehouck, A. Pierce, G. Torpier, R. Cecchelli, A new function for the LDL receptor: transcytosis of LDL across the bloodbrain barrier, J. Cell Biol. 138 (1997) 877889. [94] J. Herz, D.K. Strickland, LRP: a multifunctional scavenger and signaling receptor, J. Clin. Invest. 108 (2001) 779784. [95] M. Shibata, S. Yamada, S.R. Kumar, M. Calero, J. Bading, B. Frangione, D.M. Holtzman, C.A. Miller, D.K. Strickland, J. Ghiso, B.V. Zlokovic, Clearance of Alzheimer's amyloidss(140) peptide from brain by LDL receptor-related protein-1 at the bloodbrain barrier, J. Clin. Invest. 106 (2000) 14891499. [96] R. Deane, A. Sagare, K. Hamm, M. Parisi, B. LaRue, H. Guo, Z. Wu, D.M. Holtzman, B.V. Zlokovic, IgG-assisted age-dependent clearance of Alzheimer's amyloid beta peptide by the bloodbrain barrier neonatal Fc receptor, J. Neurosci. 25 (2005) 1149511503. [97] M. Ueno, B. Wu, T. Nakagawa, Y. Nagai, M. Onodera, C.L. Huang, T. Kusaka, K. Kanenishi, H. Sakamoto, The expression of LDL receptor in vessels with blood brain barrier impairment in a stroke-prone hypertensive model, Histochem. Cell Biol. 133 (2010) 669676. [98] K. Mishima, S. Higashiyama, Y. Nagashima, Y. Miyagi, A. Tamura, N. Kawahara, N. Taniguchi, A. Asai, Y. Kuchino, T. Kirino, Regional distribution of heparin-binding epidermal growth factor-like growth factor mRNA and protein in adult rat forebrain, Neurosci. Lett. 213 (1996) 153156. [99] P.J. Gaillard, A. Brink, A.G. de Boer, Diphtheria toxin receptor-targeted brain drug delivery, Int. Congr. Ser. 1277 (2005) 185198. [100] L.A. Opanashuk, R.J. Mark, J. Porter, D. Damm, M.P. Mattson, K.B. Seroogy, Heparinbinding epidermal growth factor-like growth factor in hippocampus: modulation of expression by seizures and anti-excitotoxic action, J. Neurosci. 19 (1999) 133146. [101] N. Kawahara, K. Mishima, S. Higashiyama, N. Taniguchi, A. Tamura, T. Kirino, The gene for heparin-binding epidermal growth factor-like growth factor is stressinducible: its role in cerebral ischemia, J. Cereb. Blood Flow Metab. 19 (1999) 307320. [102] K. Jin, Y. Sun, L. Xie, J. Childs, X.O. Mao, D.A. Greenberg, Post-ischemic administration of heparin-binding epidermal growth factor-like growth factor (HB-EGF) reduces infarct size and modies neurogenesis after focal cerebral ischemia in the rat, J. Cereb. Blood Flow Metab. 24 (2004) 399408. [103] Y.J. Yu, Y. Zhang, M. Kenrick, K. Hoyte, W. Luk, Y. Lu, J. Atwal, J.M. Elliott, S. Prabhu, R.J. Watts, M.S. Dennis, Boosting brain uptake of a therapeutic antibody by reducing its afnity for a transcytosis target, Sci. Transl. Med. 3 (2011) 84ra44. [104] K. Vass, W.F. Hickey, R.E. Schmidt, H. Lassmann, Bone marrow-derived elements in the peripheral nervous system. An immunohistochemical and Ultrastructural investigation in Chimeric rats, Lab. Invest. 69 (1993) 275282. [105] C. Charlier, K. Nielsen, S. Daou, M. Brigitte, F. Chretien, F. Dromer, Evidence of a role for monocytes in dissemination and brain invasion by Cryptococcus neoformans, Infect. Immun. 77 (2009) 120127. [106] M. Strazza, V. Pirrone, B. Wigdahl, M.R. Nonnemacher, Breaking down the barrier: the effects of HIV-1 on the bloodbrain barrier, Brain Res. 1399 (2011) 96115. [107] J. Huang, U.M. Upadhyay, R.J. Tamargo, Inammation in stroke and focal cerebral ischemia, Surg. Neurol. 66 (2006) 232245. [108] J. Jimenez, W. Jy, L.M. Mauro, L.L. Horstman, E.R. Ahn, Y.S. Ahn, A. Minagar, Elevated endothelial microparticlemonocyte complexes induced by multiple sclerosis plasma and the inhibitory effects of interferon-beta 1b on release of endothelial microparticles, formation and transendothelial migration of monocyteendothelial microparticle complexes, Mult. Scler. 11 (2005) 310315. [109] S. Floris, S.R. Ruuls, A. Wierinckx, S.M. van der Pol, E. Dopp, P.H. van der Meide, C.D. Dijkstra, H.E. De Vries, Interferon-beta directly inuences monocyte inltration into the central nervous system, J. Neuroimmunol. 127 (2002) 6979. [110] W. Roggendorf, S. Strupp, W. Paulus, Distribution and characterization of microglia/macrophages in human brain tumors, Acta Neuropathol. 92 (1996) 288293. [111] W.J. Streit, Cellular immune response in brain tumors, Neuropathol. Appl. Neurobiol. 20 (1994) 205206. [112] R.A. Morantz, G.W. Wood, M. Foster, M. Clark, K. Gollahon, Macrophages in experimental and human brain tumors. Part 2: studies of the macrophage content of human brain tumors, J. Neurosurg. 50 (1979) 305311. [113] R.A. Morantz, G.W. Wood, M. Foster, M. Clark, K. Gollahon, Macrophages in experimental and human brain tumors. Part 1: studies of the macrophage content of experimental rat brain tumors of varying immunogenicity, J. Neurosurg. 50 (1979) 298304. [114] A. Bajetto, F. Barbieri, A. Dorcaratto, S. Barbero, A. Daga, C. Porcile, J.L. Ravetti, G. Zona, R. Spaziante, G. Corte, G. Schettini, T. Florio, Expression of CXC chemokine receptors 15 and their ligands in human glioma tissues: role of CXCR4 and SDF1 in glioma cell proliferation and migration, Neurochem. Int. 49 (2006) 423432. [115] I. Desbaillets, M. Tada, N. de Tribolet, A.C. Diserens, M.F. Hamou, E.G. Van Meir, Human astrocytomas and glioblastomas express monocyte chemoattractant protein-1 (MCP-1) in vivo and in vitro, Int. J. Cancer 58 (1994) 240247. [116] M. Djukic, A. Mildner, H. Schmidt, D. Czesnik, W. Bruck, J. Priller, R. Nau, M. Prinz, Circulating monocytes engraft in the brain, differentiate into microglia and contribute to the pathology following meningitis in mice, Brain 129 (2006) 23942403. [117] E. Afergan, H. Epstein, R. Dahan, N. Koroukhov, K. Rohekar, H.D. Danenberg, G. Golomb, Delivery of serotonin to the brain by monocytes following phagocytosis of liposomes, J. Control. Release 132 (2008) 8490. [118] K. Hynynen, Ultrasound for drug and gene delivery to the brain, Adv. Drug Deliv. Rev. 60 (2008) 12091217.

[63] D.W. Holman, R.S. Klein, R.M. Ransohoff, The bloodbrain barrier, chemokines and multiple sclerosis, Biochim. Biophys. Acta 1812 (2011) 220230. [64] N. Tsao, H.P. Hsu, C.M. Wu, C.C. Liu, H.Y. Lei, Tumour necrosis factor-alpha causes an increase in bloodbrain barrier permeability during sepsis, J. Med. Microbiol. 50 (2001) 812821. [65] G.L. Bowman, J.A. Kaye, M. Moore, D. Waichunas, N.E. Carlson, J.F. Quinn, Blood brain barrier impairment in Alzheimer disease: stability and functional signicance, Neurology 68 (2007) 18091814. [66] B.S. Desai, A.J. Monahan, P.M. Carvey, B. Hendey, Bloodbrain barrier pathology in Alzheimer's and Parkinson's disease: implications for drug therapy, Cell Transplant. 16 (2007) 285299. [67] N. Marchi, L. Angelov, T. Masaryk, V. Fazio, T. Granata, N. Hernandez, K. Hallene, T. Diglaw, L. Franic, I. Najm, D. Janigro, Seizure-promoting effect of bloodbrain barrier disruption, Epilepsia 48 (2007) 732742. [68] S. Sato, S. Suga, K. Yunoki, B. Mihara, Effect of barrier opening on brain edema in human brain tumors, Acta. Neurochir. Suppl. (Wien) 60 (1994) 116118. [69] W. Fang, Y. Deng, Y. Li, E. Shang, F. Fang, P. Lv, L. Bai, Y. Qi, F. Yan, L. Mao, Blood brain barrier permeability and therapeutic time window of Ginkgolide B in ischemiareperfusion injury, Eur. J. Pharm. Sci. 39 (2010) 814. [70] Z. Cheng, J. Zhang, H. Liu, Y. Li, Y. Zhao, E. Yang, Central nervous system penetration for small molecule therapeutic agents does not increase in multiple sclerosis- and Alzheimer's disease-related animal models despite reported bloodbrain barrier disruption, Drug Metab. Dispos. 38 (2010) 13551361. [71] G. Lee, R. Bendayan, Functional expression and localization of P-glycoprotein in the central nervous system: relevance to the pathogenesis and treatment of neurological disorders, Pharm. Res. 21 (2004) 13131330. [72] J.R. Cirrito, R. Deane, A.M. Fagan, M.L. Spinner, M. Parsadanian, M.B. Finn, H. Jiang, J.L. Prior, A. Sagare, K.R. Bales, S.M. Paul, B.V. Zlokovic, D. PiwnicaWorms, D.M. Holtzman, P-glycoprotein deciency at the bloodbrain barrier increases amyloid-beta deposition in an Alzheimer disease mouse model, J. Clin. Invest. 115 (2005) 32853290. [73] R. Kortekaas, K.L. Leenders, J.C. van Oostrom, W. Vaalburg, J. Bart, A.T. Willemsen, N.H. Hendrikse, Bloodbrain barrier dysfunction in parkinsonian midbrain in vivo, Ann. Neurol. 57 (2005) 176179. [74] M. Marroni, N. Marchi, L. Cucullo, N.J. Abbott, K. Signorelli, D. Janigro, Vascular and parenchymal mechanisms in multiple drug resistance: a lesson from human epilepsy, Curr. Drug Targets 4 (2003) 297304. [75] A. Spudich, E. Kilic, H. Xing, U. Kilic, K.M. Rentsch, H. Wunderli-Allenspach, C.L. Bassetti, D.M. Hermann, Inhibition of multidrug resistance transporter-1 facilitates neuroprotective therapies after focal cerebral ischemia, Nat. Neurosci. 9 (2006) 487488. [76] E.V. Batrakova, A.V. Kabanov, Pluronic block copolymers: evolution of drug delivery concept from inert nanocarriers to biological response modiers, J. Control. Release 130 (2008) 98106. [77] D.J. Begley, ABC transporters and the bloodbrain barrier, Curr. Pharm. Des. 10 (2004) 12951312. [78] A.H. Schinkel, J.W. Jonker, Mammalian drug efux transporters of the ATP binding cassette (ABC) family: an overview, Adv. Drug Deliv. Rev. 55 (2003) 329. [79] W. Lscher, H. Potschka, Bloodbrain barrier active efux transporters: ATPbinding cassette gene family, NeuroRx 2 (2005) 8698. [80] A. Duchini, S. Govindarajan, M. Santucci, G. Zampi, F.M. Hofman, Effects of tumor necrosis factor-alpha and interleukin-6 on uid-phase permeability and ammonia diffusion in CNS-derived endothelial cells, J. Investig. Med. 44 (1996) 474482. [81] J. Lu, S. Moochhala, C. Kaur, E.A. Ling, Cellular inammatory response associated with breakdown of the bloodbrain barrier after closed head injury in rats, J. Neurotrauma 18 (2001) 399408. [82] C. Fillebeen, B. Dehouck, M. Benaissa, I. Dhennin-Duthille, R. Cecchelli, A. Pierce, Tumor necrosis factor-alpha increases lactoferrin transcytosis through the bloodbrain barrier, J. Neurochem. 73 (1999) 24912500. [83] M.J. Cipolla, R. Crete, L. Vitullo, R.D. Rix, Transcellular transport as a mechanism of bloodbrain barrier disruption during stroke, Front. Biosci. 9 (2004) 777785. [84] W.M. Pardridge, Drug delivery to the brain, J. Cereb. Blood Flow Metab. 17 (1997) 713731. [85] W.M. Pardridge, J. Eisenberg, J. Yang, Human bloodbrain barrier transferrin receptor, Metabolism 36 (1987) 892895. [86] T. Moos, E.H. Morgan, The metabolism of neuronal iron and its pathogenic role in neurological disease: review, Ann. N. Y. Acad. Sci. 1012 (2004) 1426. [87] C.M. Morris, J.M. Candy, J.M. Kerwin, J.A. Edwardson, Transferrin receptors in the normal human hippocampus and in Alzheimer's disease, Neuropathol. Appl. Neurobiol. 20 (1994) 473477. [88] R.N. Kalaria, S.M. Sromek, I. Grahovac, S.I. Harik, Transferrin receptors of rat and human brain and cerebral microvessels and their status in Alzheimer's disease, Brain Res. 585 (1992) 8793. [89] J. Wu, Y. Hua, R.F. Keep, T. Nakamura, J.T. Hoff, G. Xi, Iron and iron-handling proteins in the brain after intracerebral hemorrhage, Stroke 34 (2003) 29642969. [90] L. Recht, C.O. Torres, T.W. Smith, V. Raso, T.W. Grifn, Transferrin receptor in normal and neoplastic brain tissue: implications for brain-tumor immunotherapy, J. Neurosurg. 72 (1990) 941945. [91] L. Frolich, D. Blum-Degen, H.G. Bernstein, S. Engelsberger, J. Humrich, S. Laufer, D. Muschner, A. Thalheimer, A. Turk, S. Hoyer, R. Zochling, K.W. Boissl, K. Jellinger, P. Riederer, Brain insulin and insulin receptors in aging and sporadic Alzheimer's disease, J. Neural Transm. 105 (1998) 423438. [92] L. Xie, E. Helmerhorst, K. Taddei, B. Plewright, W. Van Bronswijk, R. Martins, Alzheimer's beta-amyloid peptides compete for insulin binding to the insulin receptor, J. Neurosci. 22 (2002) RC221.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx [119] R. Stam, Electromagnetic elds and the bloodbrain barrier, Brain Res. Rev. 65 (2010) 8097. [120] C.S. Karyekar, A. Fasano, S. Raje, R. Lu, T.C. Dowling, N.D. Eddington, Zonula occludens toxin increases the permeability of molecular weight markers and chemotherapeutic agents across the bovine brain microvessel endothelial cells, J. Pharm. Sci. 92 (2003) 414423. [121] N.J. Abbott, Inammatory mediators and modulation of bloodbrain barrier permeability, Cell. Mol. Neurobiol. 20 (2000) 131147. [122] P. Vajkoczy, M.D. Menger, Vascular microenvironment in gliomas, Cancer Treat. Res. 117 (2004) 249262. [123] T. Inamura, T. Nomura, K. Ikezaki, M. Fukui, G. Pollinger, K.L. Black, Intracarotid histamine infusion increases blood tumour permeability in RG2 glioma, Neurol. Res. 16 (1994) 125128. [124] T. Nomura, K. Ikezaki, K. Matsukado, M. Fukui, Effect of histamine on the bloodtumor barrier in transplanted rat brain tumors, Acta. Neurochir. Suppl. (Wien) 60 (1994) 400402. [125] M.H. Sarker, P.A. Fraser, The role of guanylyl cyclases in the permeability response to inammatory mediators in pial venular capillaries in the rat, J. Physiol. 540 (2002) 209218. [126] A. Yamauchi, S. Dohgu, T. Nishioku, H. Shuto, M. Naito, T. Tsuruo, Y. Sawada, Y. Kataoka, An inhibitory role of nitric oxide in the dynamic regulation of the bloodbrain barrier function, Cell. Mol. Neurobiol. 27 (2007) 263270. [127] D.F. Emerich, P. Snodgrass, M. Pink, F. Bloom, R.T. Bartus, Central analgesic actions of loperamide following transient permeation of the blood brain barrier with Cereport (RMP-7), Brain Res. 801 (1998) 259266. [128] D.F. Emerich, P. Snodgrass, R. Dean, M. Agostino, B. Hasler, M. Pink, H. Xiong, B.S. Kim, R.T. Bartus, Enhanced delivery of carboplatin into brain tumours with intravenous Cereport (RMP-7): dramatic differences and insight gained from dosing parameters, Br. J. Cancer 80 (1999) 964970. [129] D.F. Emerich, R.L. Dean, J. Marsh, M. Pink, D. Lafreniere, P. Snodgrass, R.T. Bartus, Intravenous cereport (RMP-7) enhances delivery of hydrophilic chemotherapeutics and increases survival in rats with metastatic tumors in the brain, Pharm. Res. 17 (2000) 12121219. [130] C.V. Borlongan, D.F. Emerich, Facilitation of drug entry into the CNS via transient permeation of blood brain barrier: laboratory and preliminary clinical evidence from bradykinin receptor agonist, Cereport. Brain Res. Bull. 60 (2003) 297306. [131] E. Sanovich, R.T. Bartus, P.M. Friden, R.L. Dean, H.Q. Le, M.W. Brightman, Pathway across bloodbrain barrier opened by the bradykinin agonist, RMP-7, Brain Res. 705 (1995) 125135. [132] J.B. Mackic, M. Stins, S. Jovanovic, K.S. Kim, R.T. Bartus, B.V. Zlokovic, Cereport (RMP-7) increases the permeability of human brain microvascular endothelial cell monolayers, Pharm. Res. 16 (1999) 13601365. [133] R.F. Barth, W. Yang, R.T. Bartus, M.L. Moeschberger, J.H. Goodman, Enhanced delivery of boronophenylalanine for neutron capture therapy of brain tumors using the bradykinin analog Cereport (Receptor-Mediated Permeabilizer-7), Neurosurgery 44 (1999) 351359 discussion 359360. [134] D.J. Bidanset, L. Placidi, R. Rybak, J. Palmer, J.P. Sommadossi, E.R. Kern, Intravenous infusion of cereport increases uptake and efcacy of acyclovir in herpes simplex virus-infected rat brains, Antimicrob. Agents Chemother. 45 (2001) 23162323. [135] R.T. Bartus, P. Snodgrass, J. Marsh, M. Agostino, A. Perkins, D.F. Emerich, Intravenous cereport (RMP-7) modies topographic uptake prole of carboplatin within rat glioma and brain surrounding tumor, elevates platinum levels, and enhances survival, J. Pharmacol. Exp. Ther. 293 (2000) 903911. [136] X. Zhang, Y. Xie, Y. Jin, X. Hou, L. Ye, J. Lou, The effect of RMP-7 and its derivative on transporting Evans blue liposomes into the brain, Drug Deliv. 11 (2004) 301309. [137] A. Gregor, M. Lind, H. Newman, R. Grant, D.M. Hadley, T. Barton, C. Osborn, Phase II studies of RMP-7 and carboplatin in the treatment of recurrent high grade glioma. RMP-7 European Study Group, J. Neurooncol 44 (1999) 137145. [138] K. Warren, R. Jakacki, B. Widemann, A. Aikin, M. Libucha, R. Packer, G. Vezina, G. Reaman, D. Shaw, M. Krailo, C. Osborne, J. Cehelsky, D. Caldwell, J. Stanwood, S.M. Steinberg, F.M. Balis, Phase II trial of intravenous lobradimil and carboplatin in childhood brain tumors: a report from the Children's Oncology Group, Cancer Chemother. Pharmacol. 58 (2006) 343347. [139] Y. Kuang, S.N. Lackay, L. Zhao, Z.F. Fu, Role of chemokines in the enhancement of BBB permeability and inammatory inltration after rabies virus infection, Virus Res. 144 (2009) 1826. [140] L.M. Dallasta, L.A. Pisarov, J.E. Esplen, J.V. Werley, A.V. Moses, J.A. Nelson, C.L. Achim, Bloodbrain barrier tight junction disruption in human immunodeciency virus-1 encephalitis, Am. J. Pathol. 155 (1999) 19151927. [141] S. Nakamuta, H. Endo, Y. Higashi, A. Kousaka, H. Yamada, M. Yano, H. Kido, Human immunodeciency virus type 1 gp120-mediated disruption of tight junction proteins by induction of proteasome-mediated degradation of zonula occludens-1 and -2 in human brain microvascular endothelial cells, J. Neurovirol. 14 (2008) 186195. [142] S. Verma, M. Kumar, U. Gurjav, S. Lum, V.R. Nerurkar, Reversal of West Nile virus-induced bloodbrain barrier disruption and tight junction proteins degradation by matrix metalloproteinases inhibitor, Virology 397 (2010) 130138. [143] S. Verma, Y. Lo, M. Chapagain, S. Lum, M. Kumar, U. Gurjav, H. Luo, A. Nakatsuka, V.R. Nerurkar, West nile virus infection modulates human brain microvascular endothelial cells tight junction proteins and cell adhesion molecules: transmigration across the in vitro bloodbrain barrier, Virology 385 (2009) 425433. [144] K.D. Foust, E. Nurre, C.L. Montgomery, A. Hernandez, C.M. Chan, B.K. Kaspar, Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes, Nat. Biotechnol. 27 (2009) 5965.

23

[145] M.G. Kaplitt, A. Feigin, C. Tang, H.L. Fitzsimons, P. Mattis, P.A. Lawlor, R.J. Bland, D. Young, K. Strybing, D. Eidelberg, M.J. During, Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial, Lancet 369 (2007) 20972105. [146] N.D. Doolittle, M.E. Miner, W.A. Hall, T. Siegal, E. Jerome, E. Osztie, L.D. McAllister, J.S. Bubalo, D.F. Kraemer, D. Fortin, R. Nixon, L.L. Muldoon, E.A. Neuwelt, Safety and efcacy of a multicenter study using intraarterial chemotherapy in conjunction with osmotic opening of the bloodbrain barrier for the treatment of patients with malignant brain tumors, Cancer 88 (2000) 637647. [147] L. Sztriha, A.L. Betz, Oleic acid reversibly opens the bloodbrain barrier, Brain Res. 550 (1991) 257262. [148] C. Schulze, C. Smales, L.L. Rubin, J.M. Staddon, Lysophosphatidic acid increases tight junction permeability in cultured brain endothelial cells, J. Neurochem. 68 (1997) 9911000. [149] N. Gan, F. Yin, J. Peng, W.D. Wang, Effect of lysophosphatidic acid increase the permeability of bloodbrain barrier model, Zhonghua Yi Xue Za Zhi 88 (2008) 416418. [150] A. Saija, P. Princi, D. Trombetta, M. Lanza, A. De Pasquale, Changes in the permeability of the bloodbrain barrier following sodium dodecyl sulphate administration in the rat, Exp. Brain Res. 115 (1997) 546551. [151] K. Kato, S. Kokeguchi, H. Ishihara, Y. Murayama, M. Tsujimoto, H. Takada, T. Ogawa, S. Kotani, Chemical composition and immunobiological activities of sodium dodecyl sulphate extracts from the cell envelopes of Actinobacillus actinomycetemcomitans, Bacteroides gingivalis and Fusobacterium nucleatum, J. Gen. Microbiol. 133 (1987) 10331043. [152] J. Szejtli, Introduction and general overview of cyclodextrin chemistry, Chem. Rev. 98 (1998) 17431754. [153] V. Monnaert, S. Tilloy, H. Bricout, L. Fenart, R. Cecchelli, E. Monier, Behavior of alpha-, beta-, and gamma-cyclodextrins and their derivatives on an in vitro model of bloodbrain barrier, J. Pharmacol. Exp. Ther. 310 (2004) 745751. [154] V. Monnaert, D. Betbeder, L. Fenart, H. Bricout, A.M. Lenfant, C. Landry, R. Cecchelli, E. Monier, S. Tilloy, Effects of gamma- and hydroxypropyl-gamma-cyclodextrins on the transport of doxorubicin across an in vitro model of bloodbrain barrier, J. Pharmacol. Exp. Ther. 311 (2004) 11151120. [155] S. Tilloy, V. Monnaert, L. Fenart, H. Bricout, R. Cecchelli, E. Monier, Methylated beta-cyclodextrin as P-gp modulators for deliverance of doxorubicin across an in vitro model of bloodbrain barrier, Bioorg. Med. Chem. Lett. 16 (2006) 21542157. [156] C. Binkowski-Machut, F. Hapiot, P. Martin, R. Cecchelli, E. Monier, How cyclodextrins can mask their toxic effect on the bloodbrain barrier, Bioorg. Med. Chem. Lett. 16 (2006) 17841787. [157] N. Vykhodtseva, N. McDannold, K. Hynynen, Progress and problems in the application of focused ultrasound for bloodbrain barrier disruption, Ultrasonics 48 (2008) 279296. [158] K. Hynynen, N. McDannold, N. Vykhodtseva, F.A. Jolesz, Noninvasive MR imaging-guided focal opening of the bloodbrain barrier in rabbits, Radiology 220 (2001) 640646. [159] N. Sheikov, N. McDannold, N. Vykhodtseva, F. Jolesz, K. Hynynen, Cellular mechanisms of the bloodbrain barrier opening induced by ultrasound in presence of microbubbles, Ultrasound Med. Biol. 30 (2004) 979989. [160] K. Hynynen, N. McDannold, H. Martin, F.A. Jolesz, N. Vykhodtseva, The threshold for brain damage in rabbits induced by bursts of ultrasound in the presence of an ultrasound contrast agent (Optison), Ultrasound Med. Biol. 29 (2003) 473481. [161] J.A. Wijsman, R.R. Shivers, Heat stress affects bloodbrain barrier permeability to horseradish peroxidase in mice, Acta Neuropathol. 86 (1993) 4954. [162] M. Kinoshita, N. McDannold, F.A. Jolesz, K. Hynynen, Noninvasive localized delivery of Herceptin to the mouse brain by MRI-guided focused ultrasoundinduced bloodbrain barrier disruption, Proc. Natl. Acad. Sci. U.S.A. 103 (2006) 1171911723. [163] L.H. Treat, N. McDannold, N. Vykhodtseva, Y. Zhang, K. Tam, K. Hynynen, Targeted delivery of doxorubicin to the rat brain at therapeutic levels using MRIguided focused ultrasound, Int. J. Cancer 121 (2007) 901907. [164] J.F. Jordao, C.A. Ayala-Grosso, K. Markham, Y. Huang, R. Chopra, J. McLaurin, K. Hynynen, I. Aubert, Antibodies targeted to the brain with image-guided focused ultrasound reduces amyloid-beta plaque load in the TgCRND8 mouse model of Alzheimer's disease, PLoS One 5 (2010) e10549. [165] E. Moriyama, M. Salcman, R.D. Broadwell, Bloodbrain barrier alteration after microwave-induced hyperthermia is purely a thermal effect: I, temperature and power measurements, Surg. Neurol. 35 (1991) 177182. [166] E.N. Albert, J.M. Kerns, Reversible microwave effects on the bloodbrain barrier, Brain Res. 230 (1981) 153164. [167] R.M. Quock, F.J. Kouchich, T.K. Ishii, D.G. Lange, Microwave facilitation of domperidone antagonism of apomorphine-induced stereotypic climbing in mice, Bioelectromagnetics 8 (1987) 4555. [168] E.A. Kiyatkin, H.S. Sharma, Permeability of the bloodbrain barrier depends on brain temperature, Neuroscience 161 (2009) 926939. [169] J.C. Lin, M.F. Lin, Microwave hyperthermia-induced bloodbrain barrier alterations, Radiat. Res. 89 (1982) 7787. [170] Y. Ohmoto, H. Fujisawa, T. Ishikawa, H. Koizumi, T. Matsuda, H. Ito, Sequential changes in cerebral blood ow, early neuropathological consequences and bloodbrain barrier disruption following radiofrequency-induced localized hyperthermia in the rat, Int. J. Hyperthermia 12 (1996) 321334. [171] W.M. Williams, S.T. Lu, M. Del Cerro, S.M. Michaelson, Effect of 2450 MHz microwave energy on the bloodbrain barrier to hydrophilic molecules. D. Brain temperature and bloodbrain barrier permeability to hydrophilic tracers, Brain Res. 319 (1984) 191212.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

24

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx [200] A. Tsuji, I.I. Tamai, Carrier-mediated or specialized transport of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. 36 (1999) 277290. [201] L.A. Wade, R. Katzman, 3-O-methyldopa uptake and inhibition of L-dopa at the bloodbrain barrier, Life Sci. 17 (1975) 131136. [202] C. Rousselle, P. Clair, J.M. Lefauconnier, M. Kaczorek, J.M. Scherrmann, J. Temsamani, New advances in the transport of doxorubicin through the bloodbrain barrier by a peptide vector-mediated strategy, Mol. Pharmacol. 57 (2000) 679686. [203] M. Adenot, P. Merida, R. Lahana, Applications of a bloodbrain barrier technology platform to predict CNS penetration of various chemotherapeutic agents. 2. Cationic peptide vectors for brain delivery, Chemotherapy 53 (2007) 7376. [204] M. Zorko, U. Langel, Cell-penetrating peptides: mechanism and kinetics of cargo delivery, Adv. Drug Deliv. Rev. 57 (2005) 529545. [205] S. Deshayes, M.C. Morris, G. Divita, F. Heitz, Cell-penetrating peptides: tools for intracellular delivery of therapeutics, Cell. Mol. Life Sci. 62 (2005) 18391849. [206] G. Drin, S. Cottin, E. Blanc, A.R. Rees, J. Temsamani, Studies on the internalization mechanism of cationic cell-penetrating peptides, J. Biol. Chem. 278 (2003) 3119231201. [207] J.S. Wadia, R.V. Stan, S.F. Dowdy, Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis, Nat. Med. 10 (2004) 310315. [208] C. Rousselle, P. Clair, M. Smirnova, Y. Kolesnikov, G.W. Pasternak, S. Gac-Breton, A.R. Rees, J.-M. Scherrmann, J. Temsamani, Improved brain uptake and pharmacological activity of dalargin using a peptide-vector-mediated strategy, J. Pharmacol. Exp. Ther. 306 (2003) 371376. [209] C. Brigati, M. Giacca, D.M. Noonan, A. Albini, HIV Tat, its TARgets and the control of viral gene expression, FEMS Microbiol. Lett. 220 (2003) 5765. [210] S.R. Schwarze, A. Ho, A. Vocero-Akbani, S.F. Dowdy, In vivo protein transduction: delivery of a biologically active protein into the mouse, Science 285 (1999) 15691572. [211] G. Cao, W. Pei, H. Ge, Q. Liang, Y. Luo, F.R. Sharp, A. Lu, R. Ran, S.H. Graham, J. Chen, In vivo delivery of a Bcl-xL fusion protein containing the TAT protein transduction domain protects against ischemic brain injury and neuronal apoptosis, J. Neurosci. 22 (2002) 54235431. [212] E. Kilic, G.P. Dietz, D.M. Hermann, M. Bahr, Intravenous TAT-Bcl-Xl is protective after middle cerebral artery occlusion in mice, Ann. Neurol. 52 (2002) 617622. [213] M. Zhao, M.F. Kircher, L. Josephson, R. Weissleder, Differential conjugation of tat peptide to superparamagnetic nanoparticles and its effect on cellular uptake, Bioconjug. Chem. 13 (2002) 840844. [214] M.M. Fretz, E. Mastrobattista, G.A. Koning, W. Jiskoot, G. Storm, Strategies for cytosolic delivery of liposomal macromolecules, Int. J. Pharm. 298 (2005) 305309. [215] S. Santra, H. Yang, P.H. Holloway, J.T. Stanley, R.A. Mericle, Synthesis of waterdispersible uorescent, radio-opaque, and paramagnetic CdS:Mn/ZnS quantum dots: a multifunctional probe for bioimaging, J. Am. Chem. Soc. 127 (2005) 16561657. [216] L. Liu, S.S. Venkatraman, Y.Y. Yang, K. Guo, J. Lu, B. He, S. Moochhala, L. Kan, Polymeric micelles anchored with TAT for delivery of antibiotics across the blood brain barrier, Biopolymers 90 (2008) 617623. [217] H. Wang, K. Xu, L. Liu, J.P. Tan, Y. Chen, Y. Li, W. Fan, Z. Wei, J. Sheng, Y.Y. Yang, L. Li, The efcacy of self-assembled cationic antimicrobial peptide nanoparticles against Cryptococcus neoformans for the treatment of meningitis, Biomaterials 31 (2010) 28742881. [218 Y. Qin, H. Chen, W. Yuan, R. Kuai, Q. Zhang, F. Xie, L. Zhang, Z. Zhang, J. Liu, Q. He, Liposome formulated with TAT-modied cholesterol for enhancing the brain delivery. Int J Pharm 420 (2011) 304312. [219] J.F. Poduslo, G.L. Curran, Polyamine modication increases the permeability of proteins at the bloodnerve and bloodbrain barriers, J. Neurochem. 66 (1996) 15991609. [220] W. Lu, J. Wan, Z. She, X. Jiang, Brain delivery property and accelerated blood clearance of cationic albumin conjugated pegylated nanoparticle, J. Control. Release 118 (2007) 3853. [221] K. Saar, M. Lindgren, M. Hansen, E. Eiriksdottir, Y. Jiang, K. Rosenthal-Aizman, M. Sassian, U. Langel, Cell-penetrating peptides: a comparative membrane toxicity study, Anal. Biochem. 345 (2005) 5565. [222] A.K. Cardozo, V. Buchillier, M. Mathieu, J. Chen, F. Ortis, L. Ladrire, N. AllamanPillet, O. Poirot, S. Kellenberger, J.S. Beckmann, D.L. Eizirik, C. Bonny, F. Maurer, Cell-permeable peptides induce dose- and length-dependent cytotoxic effects, Biochim. Biophys. Acta(BBA) - Biomembranes 1768 (2007) 22222234. [223] E. Kilic, G.P.H. Dietz, D.M. Hermann, M. Bhr, Intravenous Tat-Bcl-XL is protective after middle cerebral artery occlusion in mice, Ann. Neurol. 52 (2002) 617622. [224] S. Fawell, J. Seery, Y. Daikh, C. Moore, L.L. Chen, B. Pepinsky, J. Barsoum, Tat-mediated delivery of heterologous proteins into cells, Proc. Natl. Acad. Sci. U.S.A. 91 (1994) 664668. [225] H. Nagahara, A.M. Vocero-Akbani, E.L. Snyder, A. Ho, D.G. Latham, N.A. Lissy, M. Becker-Hapak, S.A. Ezhevsky, S.F. Dowdy, Transduction of full-length TAT fusion proteins into mammalian cells: TAT-p27Kip1 induces cell migration, Nat. Med. 4 (1998) 14491452. [226] J.M. Sabatier, E. Vives, K. Mabrouk, A. Benjouad, H. Rochat, A. Duval, B. Hue, E. Bahraoui, Evidence for neurotoxic activity of tat from human immunodeciency virus type 1, J. Virol. 65 (1991) 961967. [227] W.M. Pardridge, Drug and gene targeting to the brain with molecular Trojan horses, Nat. Rev. Drug Discov. 1 (2002) 131139. [228] W.M. Pardridge, R.J. Boado, Y.S. Kang, Vector-mediated delivery of a polyamide (peptide) nucleic acid analogue through the bloodbrain barrier in vivo, Proc. Natl. Acad. Sci. U.S.A. 92 (1995) 55925596.

[172] D.G. Lange, J. Sedmak, Japanese encephalitis virus (JEV): potentiation of lethality in mice by microwave radiation, Bioelectromagnetics 12 (1991) 335348. [173] F.S. Prato, J.R. Frappier, R.R. Shivers, M. Kavaliers, P. Zabel, D. Drost, T.Y. Lee, Magnetic resonance imaging increases the bloodbrain barrier permeability to 153-gadolinium diethylenetriaminepentaacetic acid in rats, Brain Res. 523 (1990) 301304. [174] R.R. Shivers, M. Kavaliers, G.C. Teskey, F.S. Prato, R.M. Pelletier, Magnetic resonance imaging temporarily alters bloodbrain barrier permeability in the rat, Neurosci. Lett. 76 (1987) 2531. [175] E. Preston, K. Butler, N. Haas, Does magnetic resonance imaging compromise integrity of the bloodbrain barrier? Neurosci. Lett. 101 (1989) 4650. [176] R.P. Liburdy, D.J. de Manincor, M.S. Roos, K.M. Brennan, Permeability of the bloodbrain barrier of the rat is not signicantly altered by NMR exposure, Ann. N. Y. Acad. Sci. 649 (1992) 345349. [177] L.B. Qiu, G.R. Ding, K.C. Li, X.W. Wang, Y. Zhou, Y.C. Zhou, Y.R. Li, G.Z. Guo, The role of protein kinase C in the opening of bloodbrain barrier induced by electromagnetic pulse, Toxicology 273 (2010) 2934. [178] Y.C. Kuo, C.Y. Kuo, Electromagnetic interference in the permeability of saquinavir across the bloodbrain barrier using nanoparticulate carriers, Int. J. Pharm. 351 (2008) 271281. [179] Y.-E.L. Koo, G.R. Reddy, M. Bhojani, R. Schneider, M.A. Philbert, A. Rehemtulla, B.D. Ross, R. Kopelman, Brain cancer diagnosis and therapy with nanoplatforms, Adv. Drug Deliv. Rev. 58 (2006) 15561577. [180] S. Bhaskar, F. Tian, T. Stoeger, W. Kreyling, J.M. de la Fuente, V. Grazu, P. Borm, G. Estrada, V. Ntziachristos, D. Razansky, Multifunctional Nanocarriers for diagnostics, drug delivery and targeted treatment across bloodbrain barrier: perspectives on tracking and neuroimaging, Part. Fibre Toxicol. 7 (2010) 3. [181] E. Garcia-Garcia, K. Andrieux, S. Gil, P. Couvreur, Colloidal carriers and blood brain barrier (BBB) translocation: a way to deliver drugs to the brain? Int. J. Pharm. 298 (2005) 274292. [182] ClinicalTrials.gov, Pegylated Liposomal Doxorubicine and Prolonged Temozolomide in Addition to Radiotherapy in Newly Diagnosed Glioblastoma. http:// clinicaltrials.gov/ct2/show/NCT00944801, Last access 21st July 2011. [183] H. Chen, Y. Qin, Q. Zhang, W. Jiang, L. Tang, J. Liu, Q. He, Lactoferrin modied doxorubicin-loaded procationic liposomes for the treatment of gliomas. Eur. J. Pharm. Sci. 44 (2011) 164173. [184] B. Kateb, K. Chiu, K.L. Black, V. Yamamoto, B. Khalsa, J.Y. Ljubimova, H. Ding, R. Patil, J.A. Portilla-Arias, M. Modo, D.F. Moore, K. Farahani, M.S. Okun, N. Prakash, J. Neman, D. Ahdoot, W. Grundfest, S. Nikzad, J.D. Heiss, Nanoplatforms for constructing new approaches to cancer treatment, imaging, and drug delivery: what should be the policy? Neuroimage 54 (Suppl 1) (2011) S106S124. [185] C. Dufes, A.G. Schatzlein, L. Tetley, A.I. Gray, D.G. Watson, J.C. Olivier, W. Couet, I.F. Uchegbu, Niosomes and polymeric chitosan based vesicles bearing transferrin and glucose ligands for drug targeting, Pharm. Res. 17 (2000) 12501258. [186] P. Arunothayanun, M.S. Bernard, D.Q. Craig, I.F. Uchegbu, A.T. Florence, The effect of processing variables on the physical characteristics of non-ionic surfactant vesicles (niosomes) formed from a hexadecyl diglycerol ether, Int. J. Pharm. 201 (2000) 714. [187] D.E. Discher, F. Ahmed, Polymersomes, Annu. Rev. Biomed. Eng. 8 (2006) 323341. [188] X.M. Liu, K.P. Pramoda, Y.Y. Yang, S.Y. Chow, C. He, Cholesteryl-grafted functional amphiphilic poly(N-isopropylacrylamide-co-N-hydroxylmethylacrylamide): synthesis, temperature-sensitivity, self-assembly and encapsulation of a hydrophobic agent, Biomaterials 25 (2004) 26192628. [189] M.C. Woodle, Sterically stabilized liposome therapeutics, Adv. Drug Deliv. Rev. 16 (1995) 249265. [190] H. Hatakeyama, H. Akita, E. Ishida, K. Hashimoto, H. Kobayashi, T. Aoki, J. Yasuda, K. Obata, H. Kikuchi, T. Ishida, H. Kiwada, H. Harashima, Tumor targeting of doxorubicin by anti-MT1-MMP antibody-modied PEG liposomes, Int. J. Pharm. 342 (2007) 194200. [191] X. Li, L. Ding, Y. Xu, Y. Wang, Q. Ping, Targeted delivery of doxorubicin using stealth liposomes modied with transferrin, Int. J. Pharm. 373 (2009) 116123. [192] E. Markoutsa, G. Pampalakis, A. Niarakis, I.A. Romero, B. Weksler, P.-O. Couraud, S.G. Antimisiaris, Uptake and permeability studies of BBB-targeting immunoliposomes using the hCMEC/D3 cell line, Eur. J. Pharm. Biopharm. 77 (2011) 265274. [193] S. Stolnik, L. Illum, S.S. Davis, Long circulating microparticulate drug carriers, Adv. Drug Deliv. Rev. 16 (1995) 195214. [194] A.G.A. Coombes, P.D. Scholes, M.C. Davies, L. Illum, S.S. Davis, Resorbable polymeric microspheres for drug delivery production and simultaneous surface modication using PEO-PPO surfactants, Biomaterials 15 (1994) 673680. [195] R. Gref, Y. Minamitake, M.T. Peracchia, V. Trubetskoy, V. Torchilin, R. Langer, Biodegradable long-circulating polymeric nanospheres, Science (Wash DC) 263 (1994) 16001603. [196] D. Bazile, C. Prud'homme, M.T. Bassoullet, M. Marlard, G. Spenlehauer, M. Veillard, Stealth Me.PEG-PLA nanoparticles avoid uptake by the mononuclear phagocytes system, J. Pharm. Sci. 84 (1995) 493498. [197] M. Peracchia, R. Gref, Y. Minamitake, A. Domb, N. Lotan, R. Langer, PEG-coated nanospheres from amphiphilic diblock and multiblock copolymers: investigation of their drug encapsulation and release characteristics, J. Control. Release 46 (1997) 223231. [198] P. Calvo, B. Gouritin, H. Chacun, D. Desmaele, J. D'Angelo, J.P. Noel, D. Georgin, E. Fattal, J.P. Andreux, P. Couvreur, Long-circulating PEGylated polycyanoacrylate nanoparticles as new drug carrier for brain delivery, Pharm. Res. 18 (2001) 11571166. [199] I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer therapy and diagnosis, Adv. Drug Deliv. Rev. 54 (2002) 631651.

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx [229] Y. Zhang, W.M. Pardridge, Conjugation of brain-derived neurotrophic factor to a bloodbrain barrier drug targeting system enables neuroprotection in regional brain ischemia following intravenous injection of the neurotrophin, Brain Res. 889 (2001) 4956. [230] M. Bartsch, A.H. Weeke-Klimp, D.K. Meijer, G.L. Scherphof, J.A. Kamps, Cellspecic targeting of lipid-based carriers for ODN and DNA, J. Liposome Res. 15 (2005) 5992. [231] A. Shir, M. Ogris, E. Wagner, A. Levitzki, EGF receptor-targeted synthetic doublestranded RNA eliminates glioblastoma, breast cancer, and adenocarcinoma tumors in mice, PLoS Med. 3 (2006) e6. [232] G. Raab, M. Klagsbrun, Heparin-binding EGF-like growth factor, Biochim. Biophys. Acta 1333 (1997) F179F199. [233] R. Gabathuler, Approaches to transport therapeutic drugs across the blood brain barrier to treat brain diseases, Neurobiol. Dis. 37 (2010) 4857. [234] W.M. Pardridge, J. Eisenberg, J. Yang, Human bloodbrain barrier insulin receptor, J. Neurochem. 44 (1985) 17711778. [235] W.M. Pardridge, Y.S. Kang, J.L. Buciak, J. Yang, Human insulin receptor monoclonal antibody undergoes high afnity binding to human brain capillaries in vitro and rapid transcytosis through the bloodbrain barrier in vivo in the primate, Pharm. Res. 12 (1995) 807816. [236] Y. Zhang, R.J. Boado, W.M. Pardridge, Marked enhancement in gene expression by targeting the human insulin receptor, J. Gene Med. 5 (2003) 157163. [237] C.F. Xia, R.J. Boado, W.M. Pardridge, Antibody-mediated targeting of siRNA via the human insulin receptor using avidinbiotin technology, Mol. Pharm. 6 (2009) 747751. [238] D. Wu, J. Yang, W.M. Pardridge, Drug targeting of a peptide radiopharmaceutical through the primate bloodbrain barrier in vivo with a monoclonal antibody to the human insulin receptor, J. Clin. Invest. 100 (1997) 18041812. [239] R.J. Boado, Y. Zhang, W.M. Pardridge, Humanization of anti-human insulin receptor antibody for drug targeting across the human bloodbrain barrier, Biotechnol. Bioeng. 96 (2007) 381391. [240] T. Moos, E.H. Morgan, Transferrin and transferrin receptor function in brain barrier systems, Cell. Mol. Neurobiol. 20 (2000) 7795. [241] M.T. da Cruz, S. Simoes, M.C. de Lima, Improving lipoplex-mediated gene transfer into C6 glioma cells and primary neurons, Exp. Neurol. 187 (2004) 6575. [242] H.J. Lee, B. Engelhardt, J. Lesley, U. Bickel, W.M. Pardridge, Targeting rat antimouse transferrin receptor monoclonal antibodies through bloodbrain barrier in mouse, J. Pharmacol. Exp. Ther. 292 (2000) 10481052. [243] K. Ulbrich, T. Hekmatara, E. Herbert, J. Kreuter, Transferrin- and transferrinreceptor-antibody-modied nanoparticles enable drug delivery across the bloodbrain barrier (BBB), Eur. J. Pharm. Biopharm. 71 (2009) 251256. [244] I. van Rooy, E. Mastrobattista, G. Storm, W.E. Hennink, R.M. Schiffelers, Comparison of ve different targeting ligands to enhance accumulation of liposomes into the brain, J. Control. Release 150 (2011) 3036. [245] U. Schrder, B.A. Sabel, Nanoparticles, a drug carrier system to pass the blood brain barrier, permit central analgesic effects of i.v. dalargin injections, Brain Res. 710 (1996) 121124. [246] P. Blasi, S. Giovagnoli, A. Schoubben, M. Ricci, C. Rossi, Solid lipid nanoparticles for targeted brain drug delivery, Adv. Drug Deliv. Rev. 59 (2007) 454477. [247] A. Zensi, D. Begley, C. Pontikis, C. Legros, L. Mihoreanu, S. Wagner, C. Buchel, H. von Briesen, J. Kreuter, Albumin nanoparticles targeted with Apo E enter the CNS by transcytosis and are delivered to neurones, J. Control. Release 137 (2009) 7886. [248] S.Y. Yuan, New insights into eNOS signaling in microvascular permeability, Am. J. Physiol. Heart Circ. Physiol. 291 (2006) H1029H1031. [249] B. Leveugle, G. Spik, D.P. Perl, C. Bouras, H.M. Fillit, P.R. Hof, The iron-binding protein lactotransferrin is present in pathologic lesions in a variety of neurodegenerative disorders: a comparative immunohistochemical analysis, Brain Res. 650 (1994) 2031. [250] C. Fillebeen, L. Descamps, M.-P. Dehouck, L. Fenart, M. Benaissa, G. Spik, R. Cecchelli, A. Pierce, Receptor-mediated transcytosis of lactoferrin through the bloodbrain barrier, J. Biol. Chem. 274 (1999) 70117017. [251] R.Q. Huang, W.L. Ke, Y.H. Qu, J.H. Zhu, Y.Y. Pei, C. Jiang, Characterization of lactoferrin receptor in brain endothelial capillary cells and mouse brain, J. Biomed. Sci. 14 (2007) 121128. [252] B. Ji, J. Maeda, M. Higuchi, K. Inoue, H. Akita, H. Harashima, T. Suhara, Pharmacokinetics and brain uptake of lactoferrin in rats, Life Sci. 78 (2006) 851855. [253] K. Hu, J. Li, Y. Shen, W. Lu, X. Gao, Q. Zhang, X. Jiang, Lactoferrin-conjugated PEGPLA nanoparticles with improved brain delivery: In vitro and in vivo evaluations, J. Control. Release 134 (2009) 5561. [254] K. Hu, Y. Shi, W. Jiang, J. Han, S. Huang, X. Jiang, Lactoferrin conjugated PEGPLGA nanoparticles for brain delivery: preparation, characterization and efcacy in Parkinson's disease, Int. J. Pharm. 415 (2011) 273283. [255] K. Hu, Y. Shi, W. Jiang, J. Han, S. Huang, X. Jiang, Lactoferrin conjugated PEGPLGA nanoparticles for brain delivery: preparation, characterization and efcacy in Parkinson's disease, Int. J. Pharm. 415 (2011) 273283. [256] M. Demeule, J. Poirier, J. Jodoin, Y. Bertrand, R.R. Desrosiers, C. Dagenais, T. Nguyen, J. Lanthier, R. Gabathuler, M. Kennard, W.A. Jefferies, D. Karkan, S. Tsai, L. Fenart, R. Cecchelli, R. Beliveau, High transcytosis of melanotransferrin (P97) across the bloodbrain barrier, J. Neurochem. 83 (2002) 924933. [257] R. Gabathuler, G. Arthur, M. Kennard, Q. Chen, S. Tsai, J. Yang, W. Schoorl, T.Z. Vitalis, W.A. Jefferies, Development of a potential protein vector (NeuroTrans) to deliver drugs across the bloodbrain barrier, Int. Congr. Ser. 1277 (2005) 171184. [258 Raptor, Raptor Pharmaceutical Corp entered a research collaboration and license agreement with Roche in June 2009, http://www.raptorpharma.com/science_ neurotrans.html, Last access 27th July, 2011.

25

[259] M. Demeule, A. Regina, C. Che, J. Poirier, T. Nguyen, R. Gabathuler, J.P. Castaigne, R. Beliveau, Identication and design of peptides as a new drug delivery system for the brain, J. Pharmacol. Exp. Ther. 324 (2008) 10641072. [260] M. Demeule, J.C. Currie, Y. Bertrand, C. Che, T. Nguyen, A. Regina, R. Gabathuler, J.P. Castaigne, R. Beliveau, Involvement of the low-density lipoprotein receptor-related protein in the transcytosis of the brain delivery vector angiopep-2, J. Neurochem. 106 (2008) 15341544. [261] W. Ke, K. Shao, R. Huang, L. Han, Y. Liu, J. Li, Y. Kuang, L. Ye, J. Lou, C. Jiang, Gene delivery targeted to the brain using an Angiopep-conjugated polyethyleneglycolmodied polyamidoamine dendrimer, Biomaterials 30 (2009) 69766985. [262] K. Shao, R. Huang, J. Li, L. Han, L. Ye, J. Lou, C. Jiang, Angiopep-2 modied PE-PEG based polymeric micelles for amphotericin B delivery targeted to the brain, J. Control. Release 147 (2010) 118126. [263] F.C. Thomas, K. Taskar, V. Rudraraju, S. Goda, H.R. Thorsheim, J.A. Gaasch, R.K. Mittapalli, D. Palmieri, P.S. Steeg, P.R. Lockman, Q.R. Smith, Uptake of ANG1005, a novel paclitaxel derivative, through the bloodbrain barrier into brain and experimental brain metastases of breast cancer, Pharm. Res. 26 (2009) 24862494. [264] A. Regina, M. Demeule, C. Che, I. Lavallee, J. Poirier, R. Gabathuler, R. Beliveau, J.P. Castaigne, Antitumour activity of ANG1005, a conjugate between paclitaxel and the new brain delivery vector Angiopep-2, Br. J. Pharmacol. 155 (2008) 185197. [265] G. Tosi, A.V. Vergoni, B. Ruozi, L. Bondioli, L. Badiali, F. Rivasi, L. Costantino, F. Forni, M.A. Vandelli, Sialic acid and glycopeptides conjugated PLGA nanoparticles for central nervous system targeting: in vivo pharmacological evidence and biodistribution, J. Control. Release 145 (2010) 4957. [266] G. Giannini, R. Rappuoli, G. Ratti, The amino-acid sequence of two non-toxic mutants of diphtheria toxin: CRM45 and CRM197, Nucleic Acids Res. 12 (1984) 40634069. [267] M. Kaefer, S. Vemulapalli, M.R. Freeman, A nontoxic diphtheria toxin analogue inhibits neonatal bladder smooth muscle cell proliferation, J. Urol. 163 (2000) 580584. [268] P. Anderson, Antibody responses to Haemophilus inuenzae type b and diphtheria toxin induced by conjugates of oligosaccharides of the type b capsule with the nontoxic protein CRM197, Infect. Immun. 39 (1983) 233238. [269] S. Buzzi, D. Rubboli, G. Buzzi, A.M. Buzzi, C. Morisi, F. Pironi, CRM197 (nontoxic diphtheria toxin): effects on advanced cancer patients, Cancer Immunol. Immunother. 53 (2004) 10411048. [270] P.J. Gaillard, A.G. de Boer, A novel opportunity for targeted drug delivery to the brain, J. Control. Release 116 (2006) e60e62. [271] M.F. Fromm, P-glycoprotein: a defense mechanism limiting oral bioavailability and CNS accumulation of drugs, Int. J. Clin. Pharmacol. Ther. 38 (2000) 6974. [272] D.W. Miller, E.V. Batrakova, T.O. Waltner, V. Alakhov, A.V. Kabanov, Interactions of pluronic block copolymers with brain microvessel endothelial cells: evidence of two potential pathways for drug absorption, Bioconjug. Chem. 8 (1997) 649657. [273] A.V. Kabanov, E.V. Batrakova, D.W. Miller, Pluronic block copolymers as modulators of drug efux transporter activity in the bloodbrain barrier, Adv. Drug Deliv. Rev. 55 (2003) 151164. [274] E.V. Batrakova, S. Li, D.W. Miller, A.V. Kabanov, Pluronic P85 increases permeability of a broad spectrum of drugs in polarized BBMEC and Caco-2 cell monolayers, Pharm. Res. 16 (1999) 13661372. [275] E.V. Batrakova, S. Li, V.Y. Alakhov, D.W. Miller, A.V. Kabanov, Optimal structure requirements for pluronic block copolymers in modifying P-glycoprotein drug efux transporter activity in bovine brain microvessel endothelial cells, J. Pharmacol. Exp. Ther. 304 (2003) 845854. [276] E.V. Batrakova, D.W. Miller, S. Li, V.Y. Alakhov, A.V. Kabanov, W.F. Elmquist, Pluronic P85 enhances the delivery of digoxin to the brain: in vitro and in vivo studies, J. Pharmacol. Exp. Ther. 296 (2001) 551557. [277] E.V. Batrakova, S. Li, Y. Li, V.Y. Alakhov, A.V. Kabanov, Effect of pluronic P85 on ATPase activity of drug efux transporters, Pharm. Res. 21 (2004) 22262233. [278] E.V. Batrakova, H.Y. Han, D.W. Miller, A.V. Kabanov, Effects of pluronic P85 unimers and micelles on drug permeability in polarized BBMEC and Caco-2 cells, Pharm. Res. 15 (1998) 15251532. [279] A.K. Sharma, L. Zhang, S. Li, D.L. Kelly, V.Y. Alakhov, E.V. Batrakova, A.V. Kabanov, Prevention of MDR development in leukemia cells by micelle-forming polymeric surfactant, J. Control. Release 131 (2008) 220227. [280] W.J. Rogers, P. Basu, Factors regulating macrophage endocytosis of nanoparticles: implications for targeted magnetic resonance plaque imaging, Atherosclerosis 178 (2005) 6773. [281] J. Panyam, V. Labhasetwar, Dynamics of endocytosis and exocytosis of poly(D, Llactide-co-glycolide) nanoparticles in vascular smooth muscle cells, Pharm. Res. 20 (2003) 212220. [282] D.L. Daleke, K. Hong, D. Papahadjopoulos, Endocytosis of liposomes by macrophages: binding, acidication and leakage of liposomes monitored by a new uorescence assay, Biochim. Biophys. Acta 1024 (1990) 352366. [283] W.V. Rodrigueza, P.H. Pritchard, M.J. Hope, The inuence of size and composition on the cholesterol mobilizing properties of liposomes in vivo, Biochim. Biophys. Acta 1153 (1993) 919. [284] J. Qin, D. Chen, H. Hu, Q. Cui, M. Qiao, B. Chen, Surface modication of RGDliposomes for selective drug delivery to monocytes/neutrophils in brain, Chem. Pharm. Bull.(Tokyo) 55 (2007) 11921197. [285] K. Mehta, G. Lopez-Berestein, E.M. Hersh, R.L. Juliano, Uptake of liposomes and liposome-encapsulated muramyl dipeptide by human peripheral blood monocytes, J. Reticuloendothel. Soc. 32 (1982) 155164. [286] A.R. Bender, H. von Briesen, J. Kreuter, I.B. Duncan, H. Rubsamen-Waigmann, Efciency of nanoparticles as a carrier system for antiviral agents in human

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

26

Y. Chen, L. Liu / Advanced Drug Delivery Reviews xxx (2011) xxxxxx immunodeciency virus-infected human monocytes/macrophages in vitro, Antimicrob. Agents Chemother. 40 (1996) 14671471. W.C. Koff, W.E. Fogler, J. Gutterman, I.J. Fidler, Efcient activation of human blood monocytes to a tumoricidal state by liposomes containing human recombinant gamma interferon, Cancer Immunol. Immunother. 19 (1985) 8589. W.C. Koff, I.J. Fidler, S.D. Showalter, M.K. Chakrabarty, B. Hampar, L.M. Ceccorulli, E.S. Kleinerman, Human monocytes activated by immunomodulators in liposomes lyse herpesvirus-infected but not normal cells, Science 224 (1984) 10071009. C. von zur Muhlen, A. Fink-Petri, J. Salaklang, D. Paul, I. Neudorfer, V. Berti, A. Merkle, K. Peter, C. Bode, D. von Elverfeldt, Imaging monocytes with iron oxide nanoparticles targeted towards the monocyte integrin MAC-1 (CD11b/CD18) does not result in improved atherosclerotic plaque detection by in vivo MRI, Contrast Media Mol. Imaging 5 (2010) 268275. A.R. Jones, E.V. Shusta, Bloodbrain barrier transport of therapeutics via receptor-mediation, Pharm. Res. 24 (2007) 17591771. A. Minagar, J.S. Alexander, Bloodbrain barrier disruption in multiple sclerosis, Mult. Scler. 9 (2003) 540549. H.C. Wijesuriya, J.Y. Bullock, R.L. Faull, S.B. Hladky, M.A. Barrand, ABC efux transporters in brain vasculature of Alzheimer's subjects, Brain Res. 1358 (2010) 228238. A.M. Palmer, The role of the blood-CNS barrier in CNS disorders and their treatment, Neurobiol. Dis. 37 (2010) 312. M. Toborek, Y.W. Lee, G. Flora, H. Pu, I.E. Andras, E. Wylegala, B. Hennig, A. Nath, Mechanisms of the bloodbrain barrier disruption in HIV-1 infection, Cell. Mol. Neurobiol. 25 (2005) 181199. M. Gottfredsson, J.R. Perfect, Fungal meningitis, Semin. Neurol. 20 (2000) 307322. J.D. Lee, L.Y. Tsai, C.H. Chen, J.J. Wang, J.K. Hsiao, C.M. Yen, Bloodbrain barrier dysfunction occurring in mice infected with Angiostrongylus cantonensis, Acta Trop. 97 (2006) 204211. H.B. Stolp, P.A. Johansson, M.D. Habgood, K.M. Dziegielewska, N.R. Saunders, C.J. Ek, Effects of neonatal systemic inammation on bloodbrain barrier permeability and behaviour in juvenile and adult rats, Cardiovasc. Psychiatry Neurol. 2011 (2011) 469046. M.D. Habgood, N. Bye, K.M. Dziegielewska, C.J. Ek, M.A. Lane, A. Potter, C. MorgantiKossmann, N.R. Saunders, Changes in bloodbrain barrier permeability to large and small molecules following traumatic brain injury in mice, Eur. J. Neurosci. 25 (2007) 231238. T.A. Brooks, S.M. Ocheltree, M.J. Seelbach, R.A. Charles, N. Nametz, R.D. Egleton, T.P. Davis, Biphasic cytoarchitecture and functional changes in the BBB induced by chronic inammatory pain, Brain Res. 1120 (2006) 172182. H. Maeda, G.Y. Bharate, J. Daruwalla, Polymeric drugs for efcient tumortargeted drug delivery based on EPR-effect, Eur. J. Pharm. Biopharm. 71 (2009) 409419. [301] V. Soni, D.V. Kohli, S.K. Jain, Transferrin-conjugated liposomal system for improved delivery of 5-uorouracil to brain, J. Drug Target. 16 (2008) 7378. [302] A. Doi, S. Kawabata, K. Iida, K. Yokoyama, Y. Kajimoto, T. Kuroiwa, T. Shirakawa, M. Kirihata, S. Kasaoka, K. Maruyama, H. Kumada, Y. Sakurai, S. Masunaga, K. Ono, S. Miyatake, Tumor-specic targeting of sodium borocaptate (BSH) to malignant glioma by transferrin-PEG liposomes: a modality for boron neutron capture therapy, J. Neurooncol 87 (2008) 287294. [303] N. Tanaka, M. Sasahara, M. Ohno, S. Higashiyama, Y. Hayase, M. Shimada, Heparinbinding epidermal growth factor-like growth factor mRNA expression in neonatal rat brain with hypoxic/ischemic injury, Brain Res. 827 (1999) 130138. [304] M.J. Simon, W.H. Kang, S. Gao, S. Banta, B. Morrison III, TAT is not capable of transcellular delivery across an intact endothelial monolayer in vitro, Ann. Biomed. Eng. 39 (2011) 394401. [305] P. Kumar, H. Wu, J.L. McBride, K.E. Jung, M.H. Kim, B.L. Davidson, S.K. Lee, P. Shankar, N. Manjunath, Transvascular delivery of small interfering RNA to the central nervous system, Nature 448 (2007) 3943. [306] N. Gandhi, Z.M. Saiyed, J. Napuri, T. Samikkannu, P.V. Reddy, M. Agudelo, P. Khatavkar, S.K. Saxena, M.P. Nair, Interactive role of human immunodeciency virus type 1 (HIV-1) clade-specic Tat protein and cocaine in bloodbrain barrier dysfunction: implications for HIV-1-associated neurocognitive disorder, J. Neurovirol. 16 (2010) 294305. [307] A. Kaur, S. Jain, A.K. Tiwary, Mannan-coated gelatin nanoparticles for sustained and targeted delivery of didanosine: in vitro and in vivo evaluation, Acta Pharm. 58 (2008) 6174. [308] H. Dou, C.B. Grotepas, J.M. McMillan, C.J. Destache, M. Chaubal, J. Werling, J. Kipp, B. Rabinow, H.E. Gendelman, Macrophage delivery of nanoformulated antiretroviral drug to the brain in a murine model of neuroAIDS, J. Immunol. 183 (2009) 661669. [309] W.M. Williams, W. Hoss, M. Formaniak, S.M. Michaelson, Effect of 2450 MHz microwave energy on the bloodbrain barrier to hydrophilic molecules, A. Effect on the permeability to sodium uorescein, Brain Res. 319 (1984) 165170. [310] F.Y. Yang, W.M. Fu, R.S. Yang, H.C. Liou, K.H. Kang, W.L. Lin, Quantitative evaluation of focused ultrasound with a contrast agent on bloodbrain barrier disruption, Ultrasound Med. Biol. 33 (2007) 14211427. [311] H.L. Liu, C.H. Pan, C.Y. Ting, M.J. Hsiao, Opening of the bloodbrain barrier by low-frequency (28-kHz) ultrasound: a novel pinhole-assisted mechanical scanning device, Ultrasound Med. Biol. 36 (2010) 325335. [312] M. Lindgren, M. Hallbrink, A. Prochiantz, U. Langel, Cell-penetrating peptides, Trends Pharmacol. Sci. 21 (2000) 99103. [313] E.L. Snyder, S.F. Dowdy, Cell penetrating peptides in drug delivery, Pharm. Res. 21 (2004) 389393. [314] J. Temsamani, P. Vidal, The use of cell-penetrating peptides for drug delivery, Drug Discov. Today 9 (2004) 10121019.

[287]

[288]

[289]

[290] [291] [292]

[293] [294]

[295] [296]

[297]

[298]

[299]

[300]

Please cite this article as: Y. Chen, L. Liu, Modern methods for delivery of drugs across the bloodbrain barrier, Adv. Drug Deliv. Rev. (2011), doi:10.1016/j.addr.2011.11.010

Das könnte Ihnen auch gefallen