Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
3704 Current Pharmaceutical Design, 2012, 18, 3704-3720 Delivery of Neurotherapeutics Across the Blood Brain Barrier in Stroke Xiaoming Hua,b, Meijuan Zhangb, Rehana K Leakc, Yu Gana,b, Peiying Lia,b, Yanqin Gaoa,* and Jun Chena,b,d,* a State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China; bDepartment of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; c Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, 15282; dGeriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261 Abstract: Stroke is a devastating disease with few therapeutic options. Despite our growing understanding of the critical mechanistic events in post-stroke brain injury, the clinical translation of these findings has been less effective. A monumental hurdle to the field has been the inability of many systemically applied therapies to efficiently cross the blood brain barrier (BBB) and enter brain cells. Over the last two decades, however, significant technological achievements have overcome this obstacle to facilitate central nervous system (CNS) drug delivery. Noninvasive drug carriers, especially cell penetrating peptide (CPP) show great potential to deliver neurotherapeutics across the BBB for the treatment of ischemic brain injury. This review begins with a brief introduction to the BBB in relation to drug delivery and then provides an overview of the development of drug carriers for neurotherapeutics, with a focus on CPP-mediated transduction. We discuss recent advances and limitations in this field, as well as mechanisms underlying CPP-mediated brain targeting. We also summarize the application of CPPs in stroke research. Continuing modifications and improvements of CPPs are expected to enhance both their feasibility in clinical stroke management and their specificity towards particular cell types. Keywords: Cell penetrating peptide, TAT protein transduction domains, blood brain barrier, stroke, neuroprotection. INTRODUCTION Stroke is one of the most common causes of death and the leading cause of serious long-term adult disability in the United States. Currently approved therapies for ischemic stroke are limited by a short temporal window of opportunity and low success rate. The development of new therapeutic strategies for stroke is imperative. In recent years, an increasing number of molecules have been identified as potential therapeutic agents for central nervous system (CNS) diseases such as stroke. However, the blood brain barrier (BBB) prevents the free passage of many of these substances into the CNS and has stymied drug development in this area. For example, nearly all peptide- or protein-based drugs fail to penetrate into brain tissue to an appreciable extent and have led to very few clinical applications. Thus, the development of drug delivery and targeting strategies to overcome the obstacle of the BBB is more urgent now than ever before. Here, we discuss recent developments in drug carriers for neurotherapeutics, with particular focus on cell penetrating peptide (CPP)-mediated transduction. We review the mechanisms, recent achievements, and limitations of CPP-mediated delivery and the application of CPP to stroke research. BLOOD BRAIN BARRIER: AN OBSTACLE TO EFFECTIVE CNS DRUG DELIVERY The presence of a barrier between the blood and the brain was first observed by Paul Ehrlich and Edwin Goldman a century ago with intravenous versus intracranial infusions of dyes such as Trypan Blue (for reviews see [1, 2]). However, it is Lewandowski that is often credited by researchers for coining the term “Bluthirnshranke” (BBB in German) and Lina Stern for coining the term “hematoencephalic barrier” [1, 2]. Although there is also a bloodcerebrospinal fluid barrier, an arachnoid barrier, and a blood-retinal barrier, the study of the BBB for the past century has overshadowed that of all the others. As a whole, these studies confirm that the *Address correspondence to these authors at the Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Tel: 412-648-1263; Fax: 412-383-9985; E-mail: [email protected] OR State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China; E-mail: [email protected] 1381-6128/12 $58.00+.00 BBB is a highly effective physical and metabolic barrier segregating CNS parenchyma from the blood circulation. It prevents the passage of non-CNS cells, various foreign substances and microbes from the bloodstream to the CNS. As a result of its efficacy in protecting vulnerable post-mitotic neurons and resident glia from potentially harmful peripheral substances, it also acts as a natural obstacle for the systemic delivery of neurotherapeutics. The BBB is composed principally of specialized capillary endothelial cells sealed by highly restrictive tight junctions not found elsewhere in the circulation. These endothelial cells are, in turn, surrounded by pericytes that cover 32% of the brain capillaries [3]. Pericytes synthesize most elements of the basement membrane and share this membrane with the endothelial cells. Astrocytes embrace the outer surface of the endothelium and respective basement membrane with their end-feet, also known as the glia limitans perivascularis. Altogether, these densely packed cells and structure as whole maintains and stabilizes the BBB Fig. (1). In general, a CNS-penetrating drug has to be hydrophobic or small enough to cross this impressive fortification effectively. However, most newly-discovered neuroprotective substances are hydrophilic peptides or proteins that cannot cross the BBB on their own. In addition to its restrictive physical properties, the BBB has an active enzymatic system to curtail the effective half-life of perceived toxins and to remove infiltrating foreign substances Fig. (1). For example, the enkephalin metabolizing enzymes, aminopeptidase M, angiotensin converting enzymes and neutral endopeptidase are all active enzymatic barriers to prevent the free passage of many peptides through the BBB [4]. Finally, the BBB is also reinforced by a group of active efflux transport proteins Fig. (1), including P-glycoprotein (P-gp), multidrug resistance protein (MRP), and breast cancer resistance protein (BCRP), all of which belong to the ATP-binding cassette protein family [5]. A high concentration of P-gp is expressed in the luminal membranes of the cerebral capillary endothelium. It is the most important efflux system and actively removes many compounds from the endothelial cell cytoplasm before they penetrate the brain parenchyma. BCRP and several members in the MRP family (MRP-1, MRP3 and MRP5) are also expressed in the BBB where they help to maintain brain homeostasis and remove toxic compounds. © 2012 Bentham Science Publishers Delivery of Neurotherapeutics Current Pharmaceutical Design, 2012, Vol. 18, No. 25 3705 Fig. (1). Blood brain barrier as an obstacle for CNS drug delivery. The BBB is a physical and metabolic barrier segregating the CNS parenchyma from blood circulation. BBB is composed principally of specialized capillary endothelial cells joined by highly restrictive tight junctions. The endothelial cells are surrounded by pericytes, which synthesize most elements of the basement membrane and share it with the endothelial cells. Astrocytes embrace the outer surface of the endothelium and respective basement membrane by their end-feet. All these densely packed cells and structure form a membranous barrier to maintain and stabilize the BBB. In addition to its restrictive physical property, the BBB is reinforced by an active enzymatic system and a group of active efflux transport proteins. DRUG CARRIERS TO ENABLE BBB PENETRATION To promote the passage of therapeutic agents into the CNS, a variety of approaches have been attempted, including disruption of the BBB [6], bypassing the BBB [7], blocking efflux transporters [8], and using drug carriers such as cell-penetrating peptides (CPPs) [9], antibodies or ligands based on receptors on the BBB [10], liposomes [11], and nanoparticles (NPs) [12]. The current review will focus on non-invasive drug carriers, with a special emphasis on the CPPs. CELL-PENETRATING PEPTIDES DERIVED FROM PROTEIN TRANSDUCTION DOMAINS FOR CNS DRUG DELIVERY Analysis of naturally occurring proteins that are able to circumvent membranous barriers has led to the discovery of several protein sequences known as protein transduction domains (PTD). CPPs are small peptides derived from PTD that retain the cell-membranepermeable properties of the native proteins. When attached to the molecule of interest via either chemical or genetic means, CPPs confer their cargo with the ability to cross cellular membranes and gain access to CNS targets. CPPs are derived from various sources (Table 1). Naturally derived CPPs, including the TAT peptide from Human Immunodeficiency Virus (HIV) transcription-activating factor [13] and penetratin peptide from the Drosophila antennapedia protein [14] are prototypical CPPs and have been used extensively. Synthetic CPPs, such as homoarginine peptides and transportan, are designed with predetermined structures and are easily modified with different chemical entities, which give rise to novel and more efficient thera- peutic agents [15, 16]. Chimeric CPPs, such as the sequence signalbased peptide, combine sequences with different functions to achieve synergistic translocation efficiency [17]. Although heterogeneous in size and sequence, CPPs share a few common properties. (1) All CPPs are small peptides between 6 to 30 amino acids long. (2) Nearly all CPPs are positively net charged at physiological pH due to the high content of basic amino acids (eg. arginine and lysine) in their protein sequences. This positive charge seems to be critical for the cellular uptake of CPPs. The substitution of basic residues in CPPs with neutral alanines reduces cell penetration, while substitution of neutral residues with arginines actually enhances internalization [18, 19]. The cationic nature itself, however, is not sufficient for optimal translocation of CPPs. Other features, such as the number of arginine residues are also important for their translocation activity [20]. (3) A vast majority of CPPs are characterized by an amphipathic structure while showing overall hydrophobicity. Of course, a certain degree of hydrophobicity can help the cationic peptide sequences enter target cells, but a highly hydrophobic peptide would be trapped in the membrane rather than being internalized [21]. In short, an amphipathic structure is critical for the internalization of CPPs. In addition to its BBB permeability, several other advantages of the CPP delivery system have made it one of the most widely exploited drug carriers in CNS studies. First, CPP-mediated delivery is surprisingly independent of cargo size. Molecules range in size from small peptides, proteins, oligonucleotides, imaging agents to massive structures such as nanoparticles and liposomes have all been successfully delivered into the brain using CPPs. Second, the 3706 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 Table 1. Hu et al. Selected Cell Penetrating Peptides CPP Sequence Source Refs Naturally-derived TAT GRKKRRQRRRPPQ HIV transcription-activating factor [22] Penetratin RQIKIWFQNRRMKWKK Drosophila homeoprotein antennapedia [23] VP22 DAATATRGRSAASRPTERPRAPARSASRPRRPVD HSV VP22 protein [24] SynB1 RGGRLSYSRRRFSTSTGR Protegrin [25] pVEC LLIILRRRIRKQAHAHSK Murine vascular endothelial cadherin [26] SAP (VRLPPP) 3 Modified maize -zein-related sequence [27] hCT LGTYTQDFNKFHTFPQTAIGVGAP Human calcitonion [28] BagP MKKKTRRRST Human protein bag-1 [29] HP4 RRRRPRRRTTRRRRR Herring portamine [19] Lactoferrin (hLF) KCFQWQRNMRKVRGPPVSCIKR Human milk protein lactoferrin [30] Nucleolar targeting peptides YKQCHKKGGKKGSG Crotamine, a toxin in venom of the South American rattlesnake [31] Cytosol Localizing Peptide-1 (CyLoP-1) CRWRWKCCKK Crotamine, a toxin in venom of the South American rattlesnake [32] gH625 HGLASTLTRWAHYNALIRAFGGG glycoprotein gH of herpes simplex type I virus [33] Model amphipathic peptide (MAP), KLALKLALKALKAALKLA Synthetic [34] Homoarginine peptides RRRRRRR(RR) Synthetic [20] C105Y CSIPPEVKFNKPFVYLI Synthesized based on the amino acid sequence corresponding to residues 359-374 of alpha1-antitrypsin [35] M918 MVTVLFRRLRIRRACGPPRVRV Synthetic [36] S41 CVQWSLLRGYQPC Synthetic [37] CADY GLWRALWRLLRSLWRLLWRA Synthetic [38] MPG GALFLGFLGAAGSTMGAWSQPKSKRKV A hydrophobic domain from the fusion sequence of HIV gp41 and a hydrophilic domain from the NLS of SV40 T-antigen [39] Signal sequence- based peptide MGLGLHLLVLAAALQGAWSQPKKKRKV A hydrophobic domain from Caiman crocodylus Ig(v) light chain and a hydrophilic domain from the NLS of SV40 large T antigen [17] Transportan 10 GWTLNSAGYLLGKINLKALAALAKKIL 12 amino acids from the N-terminal of neuropeptide galanin and 14 amino acid long wasp venom peptide, mastoparan, connected via a lysine residue [40] PEP-1 KETWWETWWTEWSQPKKKRKV a hydrophobic tryptophan-rich motif and a hydrophilic lysine-rich domain derived from the nuclear localization sequence (NLS) of simian virus 40 (SV-40) large T antigen, separated by a spacer domain (SQP) [41] S4(13)-PV ALWKTLLKKVLKAPKKKRKV a 13 amino acid cell-penetrating sequence derived from the dermaseptin S4 peptide and the NLS of SV40 large T antigen [42] Synthesized Chimeric Delivery of Neurotherapeutics in vivo systemic delivery of CPPs lead to rapid expression of cargo molecules in the CNS cells. For example, certain proteins constructed using TAT-PTD technology can cross the BBB and transduce into CNS cells in a matter of mere hours after systemic administration [9, 43]. Finally, peptide-mediated delivery of macromolecules is superior to other commonly used delivery systems in many aspects, such as relatively low toxicity, high delivery efficiency, and the possibility for peptide backbone modifications. Hence, the use of CPPs opened a new gateway for CNS drug delivery and is a promising approach for treating neurological diseases. In this section, select CPPs used frequently for BBB penetration and their application in CNS diseases will be discussed. HIV TRANSCRIPTION-ACTIVATING FACTOR TAT PEPTIDE The transactivator of transcription protein of the HIV is an 86 amino acid protein that plays an essential role in virus replication. In 1988, two research groups independently discovered that the HIV TAT protein can penetrate cells efficiently when added exogenously in tissue culture [13, 44]. It was also found that the transduction property could be attributed to the amino acid counterpart of the gene sequence position 37-72 [44]. A subsequent study [22] more precisely located the 11-amino acid sequence (YGRKKRR QRRR at amino acids 47–57), now known as the TAT peptide as the essential CPP for TAT protein. It was then demonstrated in 1994 that several large proteins, when conjugated to TAT transduction peptides at amino acids 1-72 or 37-72, could enter various types of cells in culture [45]. For example, TAT-conjugated -galactosidase was shown to distribute to cells of multiple organs, but not the brain, after systemic delivery [46]. Later on, by constructing fusion proteins with the essential TAT-PTD at their amino- or carboxy-terminus, an improved TAT delivery system was shown to mediate an efficient and rapid expression of fusion protein in various tissues after intraperitoneal injection [9]. Spectacularly, the cargo protein activity was detected in brain neurons without impairment of the BBB, as manifested by the lack of Evans blue dye brain infiltration [9]. This attraction of TAT-PTD for the brain and non-dividing neurons has particularly important implications in neuroscience research and in CNS drug design. In recent years, different modifications of TAT-CPP have successfully enhanced its efficient transduction into cultured cells and increase its bioavailability and activity in vivo [47-49]. The application of the TAT-based delivery system has extended from peptides/proteins to a wide variety of cargoes (Table 2). In the later section, we will discuss in detail the application of TAT in stroke. PEP-1 Pep-1 is another CPP frequently used in CNS studies. It is a chimeric CPP with an amphipathic sequence. It contains a hydrophobic tryptophan-rich motif (KETWWETWWTEW), which is responsible for the interaction with proteins and cell membranes, a hydrophilic lysine-rich domain derived from the nuclear localization sequence (NLS) of simian virus 40 (SV-40) large T antigen (KKKRKV), which improves solubility, and a spacer domain in between (SQP), which improves the flexibility and the integrity of the other two domains [41]. When mixed with target proteins (GFP, -gal), Pep-1 peptide led the nondenatured target protein into cultured cells. A number of studies have demonstrated that Pep-1 is able to deliver a variety of proteins and peptides into the brain. A fusion protein of antioxidant enzyme Cu,Zn-superoxide dismutase (SOD) and Pep-1 can cross the BBB after intraperitoneal injection and transduce into neurons [50]. A series of studies reported that this Pep-1-SOD fusion protein protected against transient forebrain ischemia [50-52] and spinal cord injury [53, 54]. Pep-1 also successfully delivered other neuroprotective proteins into the brain, including copper chaperone for SOD (CCS) [55], ribosomal protein Current Pharmaceutical Design, 2012, Vol. 18, No. 25 3707 S3 (rPS3) [51], HSP27 [56], catalase [57] and Frataxin [58] to reduce post-ischemic injury. PENETRATIN Penetratin is derived from Drosophila homeoprotein Antennapedia (Antp) and belongs to a class of trans-activating factors that are critical in several morphological processes including neuronal morphogenesis [14, 59]. These homeoproteins bind DNA through a specific sequence of 60 amino acids called the homeodomain. It was found that the homeodomain of Antp has the capacity to translocate across the neuronal membrane [14]. Structurally, the homeodomain consists of three -helices with one turn between helix 2 and 3 [60]. Subsequent studies identified a 16-amino-acid sequence (KETWWETWWTEWSQPKKKRKV), so-called penetratin, in the third helix of the Antp homeodomain that is involved in the translocation process and is the minimal CPP of Antp [23]. The ability of penetratin to deliver cargoes across the BBB was first shown by Rousselle and colleagues. In their study, penetratin CPP enabled a covalently coupled chemotherapeutic agent to cross the cellular membranes of the BBB with high efficiency without compromising BBB integrity [25]. It has also been shown that penetratin can help small proteins, such as C-terminal structure BIR3-RING of X-linked inhibitor of apoptosis protein (XIAP) [61] and calbindin D [62], enter rat brains after systemic application and thereby decrease transient middle cerebral artery occlusion (MCAO) induced cell apoptosis. Penetratin was also conjugated with polyethylenimine (PEI) and polyamidoamine (PAMAM) to prepare Antp-modified DNA-loaded nanoparticles, which enhances the transfection and expression efficiency of DNA/PAMAM nanoparticle in brain capillary endothelial cells [63]. SYNB VECTORS The SynB peptide family is derived from a natural antimicrobial peptide called protegrin 1 (PG1) [64]. PG1 is 18 amino acids long (RGGRLCYCRRRFCVCVGR), forming an antiparallel sheet structure constrained by two disulfide bridges [65]. It interacts with and disrupts the lipid matrix of bacterial membranes [66]. SynB1 is a linear analogue of PG1 with the four cysteine residues replaced by serines and two valines replaced by threonines [25]. SynB1 is able to cross cell membranes efficiently and is devoid of any cytolytic effect of PG-1. Further optimizations have led to the development of shorter SynB peptides with improved translocation properties [67]. Several drugs conjugated to SynB vectors have demonstrated increased brain uptake and in vivo activities [25, 68, 69]. For example, the BBB non-permeable doxorubicin can gain access to the brain after systemic application when it is covalently linked to SynB1 peptide [25]. Interestingly, this delivery strategy may reduce the cardiotoxicity of doxorubicin, as the heart had significantly lower SynB1-doxorubicin levels. The SynB vectors have also been used to deliver peptide molecules such as enkephalin and streptavidin to the brain [70]. A recent study designed a novel delivery system that contains SynB1 and a thermally responsive elastin-like polypeptide (ELP). It was shown that this SynB-ELP polypeptide could enhance thermal-based therapeutics targeting to various regions of the brain, particularly the cerebellum [71]. OTHER CPPs FOR CNS DELIVERY Several other CPPs can penetrate the BBB after systemic application. For example, a synthetic arginine-rich CPP ((RXRRBR)XB) facilitated the brain uptake of antisense morpholino oligonucleotides [72], which can induce alternative pre-mRNA splicing and thereby offer a potential therapeutic tool for neurogenetic disorders such as ataxia-telangiectasia [73]. In another study, a basic amino acid-rich nucleolar localization signal in LIM kinase 2 (LIMK2 NoLS) showed potential to transport peptides and proteins across 3708 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 Table 2. Hu et al. Cargoes Delivered Across Blood Brain Barrier by TAT Cell Penetrating Peptides Delivery System in Stroke and other CNS Disease Models Cargo Formula Route Animal Model Biological Effect Refs TAT-Bcl-x(L) fusion protein iv or ip Mouse, Ischemia reperfusion (I/R) Anti-apoptotic; attenuates ischemia-induced brain injury; increases survival of neural precursor cells [43, 97, 102, 103] ip Rat, neonatal HI Anti-apoptotic; decreases cerebral tissue loss [126] ip Gerbils, transient forebrain ischemia Prevents delayed neuronal death in the hippocampus [102] Peptide and Protein Bcl-x(L) X chromosome-linked inhibitor of apoptosis (XIAP) TAT-XIAP fusion protein iv Mouse, permanent ischemia Anti-apoptotic; efficiently targets the lesion, reduces infarct volumes, reverses long-term impairments [100] Glial-cell-line-derived neurotrophic factor (GDNF) TAT-GDNF fusion protein iv Mouse, I/R Neurotrophic factor; prevents neuronal death and reduces infarct volume [99] ip Mouse, MPTP model of Parkinson's disease Neurotrophic factor; reaches dopaminergic neurons but elicits no protection [127] Erythropoietin (EPO) TAT-EPO fusion protein ip or iv Mouse, TAT enhances the capacity of EPO to cross the BBB at least twofold when given IP and up to five-fold when given IV, TAT-EPO protects against ischemic brain injury. [108] Inhibitor of PKC (V1-1) V1-1-TAT fusion protein ip Rat, I/R Improves microvascular pathology, increases cerebral blood flow, reduces infarct size [101] -Receptors for Activated C Kinase (RACK) TAT-RACK fusion protein iv Rat, Global cerebral ischemia Activation of PKC by TAT-- RACK protects the brain against ischemic damage by regulating cerebral blood flow [114] c-Jun N-terminal kinase-inhibitor (DJNKI1) TAT-DJNKI1 fusion protein ip Rat/mouse, permanent ischemia JNK inhibitor; reduces lesion volume and improves neurological function [115-117] JNK-inhibitor JBD TAT-JBD fusion protein ip Rat, neonatal HI Reduces neonatal HI brain damage with long-lasting anatomical and behavioral improvements [128] Postsynaptic density95 (PSD95) inhibitor Tat-NR2B9c fusion protein iv Rat, I/R and permanent ischemia Blocks NMDAR-PSD95 signaling, reduces infarct volumes and improves long-term neurobehavioral functions [119, 120] vein. infusion Rat, status epilepticus Reduces cell loss in hippocampus [129] I/R Neuroglobin TATNeuroglobin fusion protein iv Mouse, I/R Efficiently transduces into neurons and protects the brain from ischemia [97] Nogo-A extracellular peptide residues 1-40 (NEP1-40) TAT-NEP140 fusion protein ip Mouse/rat, I/R Inhibits neuronal death, promotes axonal regrowth and functional recovery [122, 123] Hsp70 TAT-Hsp70 fusion protein iv Mouse, I/R Chaperone; reduces infarct volume and increases survival of neural precursor cells [124] ip Mouse, MPTP model of Parkinson's disease Chaperone; prevents neuronal cell death [130] ip Rat, I/R Increases SOD activity in the brain and protects the brain from I/R injury [125] Superoxide dismutase TAT-SOD fusion protein Delivery of Neurotherapeutics Current Pharmaceutical Design, 2012, Vol. 18, No. 25 3709 (Table 2) Contd..... Cargo Formula Route Animal Model ip Gerbils, transient forebrain ischemia Biological Effect Refs Prevents neuronal death in hippocampus [131] Apaf-1-inhibitory protein (AIP) TAT-AIP fusion protein ip Rat, neonatal hypoxia-ischemia (HI) Anti-apoptotic; decreases brain damage and improves neurological functions [132] NF-B essential modulator (NEMO) binding domain (NBD) TAT-NBD fusion protein ip Rat, neonatal HI NF-B inhibitor; causes reduction in brain tissue loss and improvement in sensorimotor and cognitive functions [133] Sensitive to apoptosis gene (SAG) TAT-SAG fusion protein ip Gerbils, transient forebrain ischemia Prevents neuronal death in hippocampus, decreases lipid peroxidation in the brain [135] Leptin TAT-leptin ip Mouse, fed with high-fat diet Increases leptin in hippocampus, reduces body weight [136] Lipoamide dehydrogenase (LAD, E3) TAT-LAD fusion protein iv E3-deficient mice Causes significant increase in the enzymatic activity of the mitochondrial multienzyme complex pyruvate dehydrogenase complex within the liver, heart and brain [137] Ubiquitin c-terminal hydrolase L1 (UchL1) TAT-HAUch-L1 fusion protein s.c Mouse, APP/PS1 Alzheimer model Restores spine density [138] Ciliary neurotrophic factor (CNTF) TAT-CNTF Ip; per rectum Mouse, A-induced dementia Neurotrophic factor, reduces? learning and memory impairments [139] Choline acetyltransferase TAT-choline acetyltransferase fusion protein iv Mouse, A-induced dementia Neurotransmitter, improves memory and cognitive dysfunction [140] Cy5.5 Cy5.5-TAT iv Rat, experimental autoimmune encephalomyelitis Labels and tracks T lymphocyte [141] CdS ratio Mn/ZnS quantum dots (Qdots), TAT-Qdots Intraartery Rat Reaches the brain parenchyma for imaging [142] FITC doped silica nanoparticles TAT-FSNPs Intraartery rat Used for brain imaging [143] TAT conjugated with cholesterol for doxorubicin- loaded liposome iv Rat, Brain glioma Increases suvival time of glioma-bearing rats; distribution of doxorubicin increases in the brain and decreases in heart [144] Coumarinloaded MPEG-PCL TAT Intranasal Rat Nano-sized micelles modified with Tat facilitate direct intranasal brain delivery [145] Imaging agents Liposomes Doxorubicin-loaded liposome Nanoparticles Coumarin-loaded methoxy poly(ethylene glycol) (MPEG)/poly(caprolactone) (PCL) 3710 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 Hu et al. (Table 2) Contd..... Cargo Formula Route Animal Model Biological Effect Refs Cholesterolconjugated antimicrobial peptides G(3)R(6) nanoparticle Cholesterolconjugated G(3)R(6)TAT iv Rabbit. C.neoformans meningitis model Antimicrobial; Crosses the BBB and suppresses yeast growth in brain tissues [146] Ciprofloxacin-loaded cholesterol-conjugated poly(ethylene glycol) (PEG) TAT-PEG-bColCiprofloxacin iv Rat Ritonavir-loaded Poly (L-lactide) (PLA) nanoparticle, RitonavirPLA-TAT iv Mouse the BBB [74]. With rapid advancements in technology, an increasing number of CPPs will be discovered, designed, or modified to enable brain delivery of therapeutic biomolecules. RECEPTOR-MEDIATED SPECIFIC TRANSCYTOSIS Receptor-mediated specific transcytosis is a delivery method based on the physiological receptors expressed on endothelial cells forming the BBB. The molecule of interest is conjugated with the monoclonal antibodies or ligands to these physiological receptors, which lead to the translocation of fusion products through the BBB. So far, several receptors, including the insulin receptor and transferrin receptor, have been exploited for delivering compounds across the BBB in experimental stroke studies. TRANSFERRIN RECEPTOR Serum transferrin functions as a transporter carrying iron from the site of intake to various tissues and cells. Transferrin draws wide attention in the drug delivery area due to its nonimmunogenicity, low toxicity, as well as biodegradability [75]. In addition, the high expression levels of transferrin receptors (TfR) on CNS endothelial cells relative to other organs makes this vector an ideal candidate for CNS-targeted delivery [75]. One disadvantage, however, is that the transferrin vector applied in vivo has to compete with large amounts of endogenous transferrin for BBB TfR binding sites, which significantly diminishes its delivery efficiency [76]. To overcome this problem, a monoclonal antibody against the TfR named OX26 was generated. This antibody binds to a distinct site on TfR from the binding site for endogenous transferrin and thus cannot be outcompeted. Using this approach, nerve growth factor (NGF) was found to penetrate the BBB in a dose-dependent fashion following intravenous injection and prevented the degeneration of cholinergic striatal neurons in a rat model of Huntington’s disease [10]. In another study, a single intravenous injection of BDNF-OX26 protected neurons in rats subjected to 60-min MCAO [77]. In order to further strengthen the binding between the drug and OX26 as well as to reduce the drug modification, the high affinity of streptavidin for the biotin molecule was exploited in this system. Briefly, a conjugate of OX26 and streptavidin was produced in parallel with the monobiotinylated drug [78]. The subsequent conjugate of biotinylated FGF and OX26-streptavidin was shown to be effective in reducing brain infarct volume and in improving neurological deficits in rats subjected to permanent MCAO [79]. INSULIN RECEPTOR Small amounts of circulating insulin can cross the BBB into the CNS through insulin receptors on endothelial cells. The monoclonal antibody to human insulin receptor (HIRMAb) thus acts as a Trojan horse to ferry a molecule of interest across the BBB when this Antimicrobial; crosses the BBB and enters the brain Enhances the CNS bioavailability of the encapsulated PI and maintains therapeutic drug levels in the brain for a sustained period [147, 148] [149] molecule is fused to the carboxyl terminus of the heavy chain of HIRMAb. Exemplary HIRMAb fusion proteins used for the uptake of various drugs into the brain are listed in (Table 3). Among these fusion proteins, HIRMAb-EPO and HIRMAb-GDNF have been shown to cross the BBB after intravenous injection in rhesus monkeys [80] and both protect against cerebral ischemia following intracerebral administration in rat model of stroke [81, 82]. Although many studies observed an increase in brain uptake of the receptor ligand/antibody-conjugated proteins from the periphery compared to the corresponding unmodified proteins and reported the effectiveness of these conjugated proteins in models of neurological diseases after systemic application, several issues remain poorly addressed. For instance, it is unclear if and how fusion with antibodies or ligands alters the activity of the modified protein. In addition, given that all these targeted BBB receptors are also expressed on peripheral organs and play important physiological roles, this raises the concern that the effective dosage to enter brain parenchyma may lead to unbearable peripheral side effects. Furthermore there is also the possibility that chronic repeated injection of the fusion protein in vivo may cause the down-regulation of targeted receptors during the therapeutic period. NANOPARTICLE-MEDIATED DELIVERY Recently, various types of nanoparticles (NPs) infused with therapeutic agents have been developed as efficient, site-specific and control-released modes of drug delivery. NPs used for drug delivery consist of different materials such as polymers, lipids or metals. In principle, the biodegradable and stable polymers such as poly (alkyl cyanoacrylate) (PBCA), poly (lactide-co-glycolide) (PLGA), human serum albumin [83] are excellent candidates. Drugs can either be integrated into the matrix of the particle or attached to the particle surface. These NPs allow peptide cargo to gain access to the surface of the endothelial cells forming the BBB and to penetrate cell membranes. Coating the NPs with certain surfactants such as polysorbate 80 (PS 80) [84] or attaching the NPs with certain ligands such as transferrin [85] or lipoprotein [86] could redistribute the NPs and enhance their accumulation in the cerebral parenchyma. These pre-coating modified NPs also reduce kidney and liver toxicity because of their targeted distribution [87]. This system has already been effective against in vivo models of ischemic stroke [88], brain tumors [87, 89], Alzheimer’s disease [90], and Parkinson’s disease [91]. The internalization of NPs depends on the cell type, the size of the particles, the surface charge, and the hydrophobicity of the particles [92]. For instance, small particles below 200 nm are probably internalized through clathrin-mediated endocytosis, while the uptake of larger particles up to 500 nm may be caveolae-mediated. It Delivery of Neurotherapeutics Table 3. Current Pharmaceutical Design, 2012, Vol. 18, No. 25 3711 Insulin Receptor-Mediated CNS Delivery Fusion protein Route Animal Model Functions Refs HIRMAb-EPO fusion protein (Erythropoietin conjugated to human insulin receptor monoclonal antibody ) iv rhesus monkeys HIRMAb-EPO fusion protein is rapidly transported into brain [80] HIRMAb-EPO fusion protein icv rat permanent MCAO Reduces infarct volume and reduces neurological deficits [81] HIRMAb-GDNF (Glial-derived factor bind to HIRMAb) neurotrophic icv rat permanent MCAO The fusion protein is active in vivo and reduces infarct volume [82] HIRMAb-IDS fusion protein sulfatase conjugated to HIRMAb) (iduronate-2- iv Mucopolysaccharidosis [150] Type II Produces therapeutic concentrations of IDS in the brain [151] HIRMAb-TNFR fusion protein (type II TNF receptor conjugated to HIRMAb ) iv rhesus monkeys Fusion protein selectively targets brain more than peripheral organs; 3% of the injected dose is taken up by the primate brain [80] HIRMAb-ScFv (a single chain Fv antibody against the Abeta peptide conjugated to HIRMAb) iv rhesus monkeys Can be transported across the primate BBB in vivo [152] has also been reported that PS 80 coated PBCA NPs have the ability to adsorb apolipoprotein E and B in the plasma after intravenous injection and therefore cross the BBB through low-density lipoprotein receptor-related protein (LRP) [93]. IMMUNE CELL-BASED DELIVERY Resent research suggests that immune cells might also be feasible drug delivery vectors [94, 95]. This approach exploits the natural ability of peripheral immune cells such as macrophages to pass the BBB and home to sites of neuronal injury or degeneration to endocytose cellular debris. For example, transplantation of genetically modified macrophages overexpressing GDNF protects against MPTP-induced dopaminergic neurodegeneration [94]. Another study demonstrated that macrophages could deliver the vectors of NPs into gliomas for photothermal ablation [95]. Finally, it is well known that the infiltration of peripheral immune cells into the ischemic core and penumbra occurs immediately after stroke and remains prominent for several days [96]. In this regard, immune cell-based delivery is a highly promising approach to rapidly deliver neurotherapeutics to the ischemic brain for an early rescue of injured neurons. A distinct advantage of this approach is its cellular specificity, as the immune cells are not likely to target uninjured sites, either in the brain or the periphery. TAT-MEDIATED CNS DELIVERY OF THERAPEUTIC PEPTIDES AND PROTEINS IN PRE-CLINICAL STROKE RESEARCH Despite our growing understanding of the critical mechanistic events in post-stroke brain injury, there has been little clinical translation of these findings. One important limiting factor is the inability of systemically applied strategies to efficiently cross the BBB and enter brain cells. Several viral vectors, especially the adenoassociated virus (AAV), have shown great promise to achieve robust transgene expression in the brain. However, given the relatively acute nature of cell death after stroke and the small time window of opportunity for stroke treatment, gene delivery, which necessitates a relatively long time for protein expression, is not a feasible option. Additionally, the expression of viral vector-delivered genes relies on host machinery for protein synthesis, which is often severely compromised during ischemic injury. Thus far, the application of viral vectors is therefore mostly in the research arena. An alternative approach for CNS delivery of neurotherapeutics after stroke is to directly send the gene product itself into the brain. CPP is a commonly used strategy under this category. As a nonviral system, the in vivo delivery of CPP does not require a prolonged incubation period to achieve gene expression. This advantage allows for its therapeutic application in acute CNS diseases such as stroke. According to most studies, TAT coupled proteins are detected in the brain as early as 4 hours after systemic injection [43, 97-99], a timeframe that is within the therapeutic window for stroke. More intriguingly, some of these proteins have proven protective even when applied after ischemic onset [43, 98-101]. All of these advantages encourage the translation of these treatments into clinical use. Quite a few peptides and proteins have been successfully delivered into the brain by CPP for the treatment of stroke and other neurological disorders (Table 2). In this section, we will use TAT as a prototypical CPP and review several specific TAT fusion proteins that cross the BBB and exert neuroprotection against cerebral ischemia. ANTI-APOPTOTIC MOLECULES In 2002, three independent research groups [43, 83, 98] reported that systemically administered TAT-Bcl-xL penetrated the brain and protected neurons against focal ischemia/reperfusion injury. Our study [43] showed that intraperitoneally applied TATHA-Bcl-xL could be detected in brain tissue by Western blot at 4 hours after injection. Dose-dependent increases in the amount of TAT-HA-Bcl-xL were detected by either anti-HA or anti-Bcl-xL antibody in various brain regions including the cortex, caudoputamen, hippocampus, cerebellum and spinal cord. In contrast, the HA-Bcl-xL without TAT could not achieve detectable protein transduction. Quantitative ELISA confirmed that transduced Bcl-xL reached concentrations of 4 ng/mg in cortex tissue. Importantly, this TAT-HA-Bcl-xL fusion protein was protective against 90-min transient focal ischemia even when applied 45 min after ischemia onset. Similarly, Kilic and colleagues showed that intravenously injected TAT-Bcl-xL entered the brain within 4 hours. This TAT-Bcl-xL greatly reduced infarct size and improved neurological performance after 90-min of MCAO [98]. TAT-Bcl-xL also exerted neuroprotec- 3712 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 tion against a 30-min or a 2-hour focal ischemia. In another study, a derivative of Bcl-xL with a more flexible and mobile pore-forming domain was generated and designated as FNK [83]. FNK has a higher protective activity than wild-type Bcl-xL. The TAT-mycFNK fusion protein accessed brain neurons after intraperitoneal injection and prevented delayed neuronal death in the hippocampus caused by 5-min global ischemia. Subsequent studies using TATBcl-xL fusion protein demonstrated that this protein not only inhibits ischemia-induced cell death, but also enhances the survival of neural precursor cells and improves hippocampal neurogenesis after transient ischemia [102, 103]. X chromosome-linked inhibitor of apoptosis (XIAP), a potent inhibitor of both caspase-3 and apoptosis, is a 62-kDa protein that does not cross BBB by itself. TAT fused XIAP, however, could transduce cortical cells even when applied topically. Shorter constructs could successfully target brain lesions after intravenous delivery and significantly reduced infarct volumes induced by distal occlusion of the MCA [100]. TROPHIC FACTORS Many trophic factors have shown high potential for neuroprotective applications. However, they usually do not readily cross the BBB. For example, glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor that promotes neuronal survival [104]. Kilic and colleagues showed robust GDNF transduction in the brain of TAT-GDNF-treated animals 4 hours after intravenous application [99]. TAT-GDNF administered either before or 30 minutes after onset of ischemia was neuroprotective in a MCAO model of ischemic stroke, suggesting that this protein may be a powerful tool for the immediate treatment of stroke. It is now widely known that erythropoietin [41] is not only a haematopoietic factor, but also a multifunctional trophic factor with neuroprotective effects in animal models of ischemia [105] and in stroke patients [106]. However, the BBB permeability of EPO is very low as only 0.5-1% of systemically administered EPO crosses the BBB [107]. To reach therapeutic concentrations in the brain, either large amounts or multiple doses of EPO have to be applied, which may induce undesired side effects such as polycythemia and secondary stroke [105, 106]. Research from our group reported that a TAT–derived peptide enhanced the capacity of EPO to cross the BBB in animals to at least two fold higher levels than unmodified EPO when intraperitoneally administered and up to five fold higher levels when intravenously administered. Not surprisingly, the therapeutic dose of the TAT fused EPO was about ten fold lower than regular EPO to achieve equivalent neuroprotection in terms of reducing infarct volume induced by MCAO in mice [108]. PROTEIN KINASES PKC is known to be activated by ischemia/reperfusion stress in multiple cell types of the brain [109] and is involved in cellular responses such as cell apoptosis, mitochondrial dysfunction, and oxidative stress [110, 111]. TAT-mediated delivery of V1-1, a peptide inhibitor selective for PKC, was shown to be protective even when given intraperitoneally 6 hours after onset of reperfusion following 2-hour MCAO [101]. Furthermore, TAT-V1-1 improved microvascular pathology, characterized as an increased number of patent microvessels and increased cerebral blood flow following acute focal ischemia [101]. Indeed, TAT-V1-1 fusion protein (also named KAI-9803) has already shown promise in a clinical trial of myocardial ischemia [112]. Clinical trials are now underway to test the effect of TAT-V1-1 fusion protein on ischemic stroke. In contrast to PKC, the activation of PKC, a different member of the PKC family, was documented to be neuroprotective against cerebral ischemia [113, 114]. Intravenous pretreatment with a fusion protein of TAT--Receptors for Activated C Kinase (RACK), a PKC-selective peptide activator, 30 min prior to global Hu et al. cerebral ischemia conferred protection to the CA1 region of the rat hippocampus by regulating cerebral blood flow [114]. Protection against permanent or transient cerebral ischemia was also achieved when D-JNKI1 (also named XG102), a fusion protein containing a TAT sequence and a 20-amino acid sequence of the cJun N-terminal kinase (JNK) binding motif of Islet-Brain-1/JNKinteracting protein-1 (JIP-1), was applied intraventricularly or systemically with a 6-hour therapeutic window [115]. D-JNKI1afforded neuroprotection is associated with caspase-3 activation and potent inhibition of phospho-c-Jun [116]. A subsequent study further showed that D-JNKI1 could also protect against ischemic damage in the presence of tPA [117]. POSTSYNAPTIC DENSITY-95 (PSD95) INHIBITOR It has been known for a long time that activation of the NMDA receptor by pathologically high levels of excitatory neurotransmitter glutamate causes excitotoxicity and plays a key role in ischemic brain injury. However, direct targeting of the NMDA receptor is not practical for stroke treatment because blocking NMDA receptors causes substantial side effects in humans. Recent studies revealed that the postsynaptic density protein PSD95 is a prominent organizing protein coupling the NMDA receptor with other intracellular proteins and signaling enzymes including nNOS [118, 119]. A TAT coupled peptide (TAT-NR2B9c) targeting the NMDA receptor binding site on PSD95 has been shown to perturb the PSD95NMDA receptor interaction. Intravenously or intraperitoneally injected TAT-NR2B9c crossed the BBB in both rats and mice [119] and potently alleviated ischemic brain damage in rats even when applied 3 hours after onset of permanent or transient MCAO. Behavioral studies showed that the reduction in infarct size was accompanied by improved long term neurological functions [120]. NEUROGLOBIN Neuroglobin is an oxygen-binding protein found in cerebral neurons. Neuronal expression of neuroglobin enhances neuronal survival upon hypoxic challenge [121]. However, like most other protein neurotherapeutics, neuroglobin is too big to cross the BBB. Although intracerebral administration of a neuroglobin-expressing AAV vector has been shown to reduce infarct volume and improve functional outcome after focal ischemia, its invasive application and delayed expression after transduction render it clinically unfeasible. Recently, a TAT-fused neuroglobin has been developed and tested in the MCAO model of stroke [97]. TAT-neuroglobin fusion protein was delivered efficiently (at least 10-fold higher levels than endogenous neuroglobin) into mouse brain 4 hours after intravenous injection. Pretreatment with TAT-neuroglobin led to prominently reduced post-ischemic brain damage and improved neurological functions. Post-treatment with TAT-neuroglobin immediately after reperfusion, however, did not show significant protection against 2-hour ischemia. OTHERS Several other proteins such as Nogo-A extracellular peptide residues 1-40 (NEP1-40) [122, 123], heat shock protein 70 (HSP70) [124], and superoxide dismutase (SOD) [125] were show to penetrate the brain when fused with TAT and exert neuroprotective effects in models of cerebral ischemia. With ever-growing knowledge on the mechanism of neuronal damage in ischemic stroke and further improvements in the TAT transduction technique, greater numbers of therapeutic proteins or peptides will likely be tested against stroke treatment and eventually brought into the clinic for use in humans. MECHANISMS OF CPP INTERNALIZATION The mechanisms by which CPPs cross cell membranes are not completely understood. So far, three main entry mechanisms have been proposed: (1) direct penetration across the membrane; (2) Delivery of Neurotherapeutics endocytosis-mediated entry; and (3) translocation through inverted micelle formation. DIRECT PENETRATION Early studies on the mechanism of CPP internalization suggested an energy-independent mechanism involving the direct translocation of CPPs across the cell membrane. Lowering the incubation temperature to 4°C or depletion of cellular ATP did not inhibit the rapid transduction of TAT into cells, suggesting that TAT-mediated protein transduction is endocytosis independent [22, 153, 154]. Other arguments in favor of an energy-independent mechanism include the absence of effect of metabolic or endocytosis inhibitors on CPPs internalization and the failure of radiolabeled CPPs to label endocytotic vesicles [155]. Moreover, structureactivity studies indicate that the internalization was equally effective whether the CPPs were in inverse or retro forms, which implies receptor-independent recognition [156]. A direct penetration mechanism was thus proposed which emphasizes the positive charges on most CPPs including TAT and Antp. It was suggested that the unfolded fusion protein interacts with the membrane through electrostatic interactions between the basic amino acids on CPPs and the negatively charged cell membrane polysaccharides or phospholipids, and then nucleates a pore, which enables protein translocation across the membrane [156-159]. According to this theory, once inside the cell, the fusion protein is refolded into its active form with the help of the endogenous chaperone system. However, several studies have challenged the energy- and receptor-independent CPP translocation theory. It has been argued that artifactual uptake of CPPs arose following fixation procedures used for fluorescence microscopy. In addition, CPPs such as TAT or penetratin strongly bind to the plasma membrane and cannot be removed by the standard washing protocol used in FACS analysis. These membrane-bound peptides might be mistaken by FACS as internalized peptide, leading to questionable conclusions with respect to direct CPPs penetration [72]. The application of procedures avoiding these experimental artifacts yielded an alternative inter- Current Pharmaceutical Design, 2012, Vol. 18, No. 25 3713 nalization mechanism favoring endocytosis-mediated entry (discussed below). However, there are still experiments supporting the direct penetration of TAT peptide across the membrane even in conditions devoid of the artifacts described above. For example, Ter-Avetisyan and colleagues demonstrated that a low temperature did not abrogate TAT uptake or change its intracellular distribution in live unfixed cells. They also showed that genetically engineered cells incapable of performing caveolin or clathrin-mediated endocytosis showed an equal capacity to take up TAT as control cells [160]. ENDOCYTOSIS-MEDIATED ENTRY Endocytosis is a process of cellular ingestion by which patches of plasma membrane invaginate to engulf substances into the cell. Richard and colleagues demonstrated in their fluorescence microscopy study that live unfixed cells exhibit characteristic endosomal distribution of TAT or (Arg)9. Flow cytometry analysis further indicated that the kinetics of TAT uptake were similar to the kinetics of endocytosis. These data are all consistent with the involvement of endocytosis in the cellular internalization of CPPs [72]. Endocytosis is now believed to be the major route for CPP internalization. There are three types of endocytosis: clathrinmediated, caveolae-mediated and macropinocytosis Fig. (2). Studies on TAT internalization with chemical inhibition of specific endocytotic pathways point to the involvement of multiple mechanisms. In support of lipid raft-mediated endocytosis involving caveolae, Fittipaldi and colleagues showed that the internalization of TAT-EGFP endosomes was resistant to nonionic detergents and colocalized with caveolae markers. Moreover, both the disruption of lipid rafts and the disturbance of caveolar trafficking impaired the internalization of TAT-GFP [161]. They also showed that conjugated TAT was not internalized in clathrin-coated endosomes. In contrast, using unconjugated TAT, Richard and colleagues showed that specific inhibitors of clathrin-dependent endocytosis partially inhibit TAT peptide uptake, while inhibitors of raft/caveolindependent pathway or genetic deficient of caveolin-1 expression Fig. (2). Endocytosis-mediated CPP internalization. Three types of endocytosis: clathrin-mediated, caveolae-mediated and macropinocytosis, could mediate the internalization of CPPs. 3714 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 could not inhibit the internalization of TAT [162]. Taken together, these results suggest that unconjugated TAT peptide might follow a clathrin-mediated endocytic pathway, whereas conjugated TAT follows a caveolae-mediated endocytic pathway. Furthermore, lipid raft-dependent macropinocytosis, yet another form of endocytosis that is independent of either caveloe or clathrin, was also implicated in the endocytosis of large TAT-fusion proteins in excess of 30kD [163] as well as unconjugated TAT [164]. Dose-dependent inhibition of TAT uptake was observed when cells were pretreated with amiloride, an inhibitor of the Na+/K+ exchanger required for macropinocytosis and when cholesterol was removed from the cell membrane [163, 164]. Indeed, the co-existence of different routes of endocytosis is not unique for TAT but true for many other CPPs such as homoarginine peptide [150, 165] and penetratin [134, 166]. Although endocytosis is an important mechanism for CPPmediated cargo internalization, it is not surprising that this process of heterophagic internalization usually ends up with lysosomal delivery and significant degradation of CPPs and their cargoes. As a result, only a small fraction of the cargo reaches the cytoplasm in active form. In order to avoid such endosomal trapping, invasive approaches such as electroporation and microinjection were reported decades ago [167, 168]. Noninvasive methods were developed later to achieve efficient endosomal escape without physical damage to the targeted cells. One of these methods is to add peptides that can be activated in the acidic environment in the endosome and lyse the endosomal membrane to release entrapped CPP-cargoes. For example, HA2-TAT chimeric peptide, which consists of TAT and the pH-sensitive HA2 fusion peptide from influenza virus, has recently been engineered to enhance both macropinocytosis and endosomal release/delivery of TAT fused proteins and other macromolecules [163, 169]. INVERTED MICELLE FORMATION An inverted micelle refers to an aggregate of colloidal surfactants where the hydrophilic groups are concentrated inside and the hydrophobic groups extend outward. The inverted micelle model was first proposed for the internalization of penetratin [156]. The model proposes that the binding of CPPs to negatively charged lipids in the cell membrane leads to the disruption of the lipid bilayer, resulting in the formation of inverted micelles that entrap the peptide in a hydrophilic environment. Further interaction of the peptides with membrane components leads to the inverse process, resulting in the release of peptides into the cell [156, 170]. Subsequent studies showed that inverted micelles are also involved in the internalization of arginine-rich CPPs [171]. Although endocytosis is now recognized as the predominant route of CPPs internalization, mounting evidence suggested that several different mechanisms may work in concert. It is possible that different CPPs utilize different internalization mechanisms depending on their specific biophysical properties, the nature of their cargo and the experimental conditions. Furthermore, one type of CPP may use several different pathways simultaneously for efficient internalization. ADVANCES IN CPPS’ DELIVERY SYSTEM Organelle-targeted Delivery Organelle-specific CPPs are under rapid development to provide a means to target therapeutic agents not only to the cell, but also to a specific subcellular compartment to achieve optimal activity and reduce non-specific effects. So far, the nucleus and the mitochondria have been successfully targeted with CPPs for gene therapies and anti-cancer chemotherapies. Nucleus-targeted delivery is important for gene therapies. A common strategy to improve nuclear delivery of CPP-fused cargoes is to add a nuclear localization signal (NLS) to the conjugates. NLS-conjugates are highly cationic short peptides. The sequence PKKKRKV derived from SV40 large T antigens is the most often Hu et al. used NLS. For example, this NLS sequence is a component of MPG chimeric CPP and has been shown to improve the nuclear uptake of plasmid DNA and oligonucleotides without necessitating nuclear membrane breakdown [172]. The NLS-mediated nuclear delivery system has been mainly tested in tumor cell lines or in vivo tumors because a large number of anticancer drugs are DNA toxins that must bind nuclear DNA or its associated enzymes for cytotoxic effects. Recently, a nucleolin binding peptide (NBP) was reported to deliver macromolecules including fluorophores, recombinant protein and DNA to the nuclei of ocular tissues in vivo. Pegylated NBP nanoparticles could further improve nuclear delivery and the expression of transgenes. This NBP might be a useful tool for therapeutic delivery to the eyes [173]. Future studies are warranted to evaluate the potential of nuclear-targeted CPPs in gene therapies against brain diseases, such as gene therapy for enzyme/neurotransmitter replacement in neurodegenerative diseases, antisense gene therapy for brain cancers, and anti-apoptotic/anti-oxidative gene therapy for other brain injuries such as stroke. Mitochondria are another subcellular compartment specifically targeted by modified CPPs. Mitochondria are a primary source of reactive oxygen species (ROS) in the cell, and release critical apoptotic signaling molecules when damaged. Mitochondria-specific CPPs may thus provide a platform for anti-apoptotic or antioxidant therapies for many common diseases. One strategy to target mitochondria is to incorporate a mitochondrial signal sequence, such as mitochondrial malate dehydrogenase (mMDH) signal sequence into a CPP fusion protein [174]. For instance, it has been shown that TAT-mMDH-GFP was enriched in mitochondria in vitro and detectable throughout fetal and neonatal pups when injected into pregnant mice [174]. Recently, mitochondria penetrating peptides (MPP) were designed by Horton and colleagues. They examined a panel of mitochondrial transporters-synthetic cell-permeable peptides and observed efficient mitochondrial penetration in a variety of cell types [175]. According to their studies, MPP efficiency can be optimized by altering lipophilicity and charge. The application of mitochondria-specific delivery to neuronal cells was reported by Zhao and colleagues, who described that a novel mitochondriatargeted CPP antioxidant SS-31 (D-Arg-Dmt-Lys-Phe-NH2; Dmt = 2',6'-dimethyltyrosine) protected against oxidant-induced mitochondrial dysfunction and apoptosis in two neuronal cell lines [176]. The Combined Use of Peptides and other Transportation Mechanisms Many new formulations of CPPs combined with other transportation mechanisms have increased CNS delivery capacity. For example, liposomes formulated with TAT-conjugated cholesterol were recently shown to exhibit high delivery efficiency across the BBB [144]. In addition, NPs conjugated with low-molecular-weight protamine (LMWP), a CPP with high cell-translocation potency, exhibited enhanced cellular accumulation compared to unmodified NPs and increased the efficiency of brain delivery of coumarin after intranasal administration [177]. In another study, TAT was used to facilitate transport of fluorescently labeled silica NPs into the brain [143]. These studies suggested that CPP-conjugated NPs are effective carriers for brain drug delivery. With the increased interest in peptide-based drugs for the treatment and diagnosis of CNS diseases, CPP-conjugated peptidic NPs may become increasingly utilized as nanoplatforms to improve transportation across the BBB. LIMITATIONS Lack of Tissue and Cell Selectivity The lack of both tissue and cellular phenotype specificity for cargo delivery represent important caveats for using CPPs technology in the nervous system, where inappropriate targeting could lead to dire consequences for neurological function and animal behavior. Although there is a tendency for TAT-fusion proteins to favorably Delivery of Neurotherapeutics transduce neurons over glial cells [43], this delivery system is still unable to convey proteins specifically to a targeted cell type within the CNS or even to the CNS as a whole. In this regard, a doubletargeted delivery system simultaneously capable of extracellular accumulation at the appropriate cellular location through specific receptors or enzymes, and intracellular penetration through CPPs may overcome this shortcoming and render CPPs capable of distinguishing between non-target and target cells. For example, lipoplexes modified with monoclonal anti-myosin monoclonal antibody specific toward cardiac myosin and with TAT for cell penetrating can achieve targeted gene delivery to ischemic myocardium [178]. Targeted CPP delivery system to CNS cells, however, has not yet been developed, but will be essential before clinical translation is feasible. Uncertainty in Fusion Protein Activity Another caveat of using CPP as the delivery system is that there is no guarantee that the transduced fusion protein will gain its natural biological activity in target cells. Once inside the cells, it has been proposed that the CPP-fusion protein is properly refolded by chaperone proteins such as HSP 90 [179]. However, we found that the TAT system may not deliver all classes of proteins with equal success. For example, proteins that are only biologically active in their multimeric state may not be good candidates for this approach. Even the delivery of native monomeric subunits cannot ensure the proper assembly of the subunits into a functional enzyme. In this regard, the CPPs delivery system has not yet rendered the viral vectors for delivery of gene products obsolete. This is because, in many instances, the viral system utilizes host machinery for endogenous folding, enzyme assembly or post-translational modifications in a more efficient and appropriate manner. Stability For successful in vivo delivery, the CPPs must also not be metabolically degraded until they send their cargo to the appropriate destination. However, concerns about the stability of these peptide vectors still plague the field. For instance, the free TAT CPP has six potential trypsin cleavage sites [180] and thus has a short half-life of just a few minutes in the presence of trypsin [181]. Several structure-activity relationship features, including amino acid sequences, amino acid three-dimensional configurations, incorporation of non-amino acids, and type of linkage between CPPs and cargoes, have been shown to affect the stability of CPP conjugates in human serum and within cells. Generally, the D-configuration of the peptide is less sensitive to degradation by proteases and remains intact in serum and cells for a much longer time than the naturally occurring L-amino acids [182]. The replacement of protease-sensitive amino acids with their stable synthetic mimics [183, 184] or the insertion of -alanines [182] also increase CPP stability. A recent study showed that the shielding of TAT through TAT conjugated poly(ethylene glycol)-phosphatidyl ethanolamine block copolymer (TAT-PEG-PE) incorporation into PEG-PE micelles could protect TAT against trypsinolysis with an over 100-fold increase in halflife compared to free TAT [181]. With all our efforts to stabilize CPPs, one must also keep in mind that the metabolic cleavage of the CPPs after internalization is a prerequisite for the release of their cargo. Another tradeoff of gaining enhanced metabolic stability is an impairment in the clearance of CPPs and a subsequent increase in their toxicity. Therefore, a thorough evaluation of each modified CPP, with an emphasis on safety profiles, as discussed below, is essential for the eventual translation of these CPPs into the clinic. Toxicity and Immunogenicity Various CPPs have toxic effects on cultured cells. A comprehensive characterization of the metabolic effects of CPPs was done by Kilk et al [185]. Their study suggested that transportan treatment changed the GSH redox ratio and significantly decreased cellular Current Pharmaceutical Design, 2012, Vol. 18, No. 25 3715 redox potential, while other four tested peptides (TAT, Antp, Arginine9 and MAP) showed minimal effect on metabolic profiles. Cardozo and colleagues showed that although up to 100 μM TAT (48–57) alone was essentially harmless, TAT-peptide conjugates triggered significant cytotoxicity when used at concentrations higher than 10 μM [186]. In comparison, free penetratin exhibits intrinsic toxicity and penetratin conjugates showed more cellular toxicity than TAT conjugates. Although all the factors that determine the cytotoxicity of CPPs are still not completely clear, peptide length, peptide concentration, cargo molecule and coupling strategy have all been suggested as contributing factors [187]. The immunogenicity of CPPs is not frequently reported. However, since most CPPs are derived from non-human proteins, they have the potential to induce immune responses when used in a clinical scenario [188]. Therefore, further examinations of immune responses to CPPs are highly warranted. CONCLUSIONS On the one hand, the BBB is essential to maintain the integrity of the brain and the spinal cord, and yet it also presents a vexing barrier to getting drugs to their neural destinations in human CNS diseases. What was first observed by Paul Ehrlich as an inability of vital dyes to cross into the brain has therefore motivated many researchers in the past century to overcome this formidable obstacle to CNS drug delivery. Relative to previous years, the past decade witnessed significant achievements in successfully penetrating the BBB and facilitating targeted CNS drug delivery to injured neurons, to tumors, and even to specific subcellular organelles. This is in large part due to the noninvasive drug carriers, especially CPPs, which show great potential to deliver various neurotherapeutics across the BBB for the treatment of ischemic brain injury, among other conditions. However, it behooves us to retain a balanced view of these new techniques because certain weaknesses, in particular the lack of tissue and cell type specificity, still restrict their clinical application. Future modifications of CPP sequences or the combinations of CPPs with other drug carriers are expected to improve the specificity of CPPs as well as reduce the side effects associated with the delivery of cargoes to unintended organs. With such modifications in hand, CPP research may lead the way to novel avenues of non-invasive stroke management in humans. CONFLICT OF INTEREST The author(s) confirm that this article content has no conflicts of interest. ACKNOWLEDGMENTS XIAOMING HU is supported by a grant (10POST4150028) from the American Heart Association. JUN CHEN is supported by National Institutes of Health/NINDS Grants NS36736, NS43802, NS56118, and NS45048. YANQIN GAO is supported by Chinese Natural Science Foundation grants 30670642, 30870794, and 81020108021. We thank Pat Strickler for excellent secretarial support. REFERENCES [1] [2] [3] [4] [5] Bechmann I, Galea I, Perry VH. What is the blood-brain barrier (not)? Trends Immunol 2007; 28: 5-11. Liddelow SA. Fluids and barriers of the CNS: a historical viewpoint. Fluids Barriers CNS 2011; 8: 2. Kim JA, Tran ND, Li Z, Yang F, Zhou W, Fisher MJ. Brain endothelial hemostasis regulation by pericytes. J Cereb Blood Flow Metab 2006; 26: 209-17. Brownson EA, Abbruscato TJ, Gillespie TJ, Hruby VJ, Davis TP. Effect of peptidases at the blood brain barrier on the permeability of enkephalin. J Pharmacol Exp Ther 1994; 270: 675-80. Loscher W, Potschka H. Blood-brain barrier active efflux transporters: ATP-binding cassette gene family. NeuroRx 2005; 2: 86-98. 3716 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] Neuwelt EA, Dahlborg SA. Chemotherapy administered in conjunction with osmotic blood-brain barrier modification in patients with brain metastases. J Neurooncol 1987; 4: 195-207. Lin S, Fan LW, Rhodes PG, Cai Z. Intranasal administration of IGF-1 attenuates hypoxic-ischemic brain injury in neonatal rats. Exp Neurol 2009; 217: 361-70. Namanja HA, Emmert D, Pires MM, Hrycyna CA, Chmielewski J. Inhibition of human P-glycoprotein transport and substrate binding using a galantamine dimer. Biochem Biophys Res Commun 2009; 388: 672-6. Schwarze SR, Ho A, Vocero-Akbani A, Dowdy SF. In vivo protein transduction: delivery of a biologically active protein into the mouse. Science 1999; 285: 1569-72. Kordower JH, Charles V, Bayer R, et al. Intravenous administration of a transferrin receptor antibody-nerve growth factor conjugate prevents the degeneration of cholinergic striatal neurons in a model of Huntington disease. Proc Natl Acad Sci USA 1994; 91: 9077-80. Shi N, Pardridge WM. Noninvasive gene targeting to the brain. Proc Natl Acad Sci USA 2000; 97: 7567-72. Silva GA. Nanotechnology approaches for drug and small molecule delivery across the blood brain barrier. Surg Neurol 2007; 67: 1136. Frankel AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell 1988; 55: 1189-93. Joliot A, Pernelle C, Deagostini-Bazin H, Prochiantz A. Antennapedia homeobox peptide regulates neural morphogenesis. Proc Natl Acad Sci USA 1991; 88: 1864-8. Pooga M, Hallbrink M, Zorko M, Langel U. Cell penetration by transportan. Faseb J 1998; 12: 67-77. Andaloussi SE, Lehto T, Mager I, et al. Design of a peptide-based vector, PepFect6, for efficient delivery of siRNA in cell culture and systemically in vivo. Nucleic Acids Res; 39: 3972-87. Chaloin L, Vidal P, Heitz A, et al. Conformations of primary amphipathic carrier peptides in membrane mimicking environments. Biochemistry 1997; 36: 11179-87. Wender PA, Mitchell DJ, Pattabiraman K, Pelkey ET, Steinman L, Rothbard JB. The design, synthesis, and evaluation of molecules that enable or enhance cellular uptake: peptoid molecular transporters. Proc Natl Acad Sci USA 2000; 97: 13003-8. Youn JI, Park SH, Jin HT, et al. Enhanced delivery efficiency of recombinant adenovirus into tumor and mesenchymal stem cells by a novel PTD. Cancer Gene Ther 2008; 15: 703-12. Mitchell DJ, Kim DT, Steinman L, Fathman CG, Rothbard JB. Polyarginine enters cells more efficiently than other polycationic homopolymers. J Pept Res 2000; 56: 318-25. Carrigan CN, Imperiali B. The engineering of membranepermeable peptides. Anal Biochem 2005; 341: 290-8. Vives E, Brodin P, Lebleu B. A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J Biol Chem 1997; 272: 16010-7. Derossi D, Joliot AH, Chassaing G, Prochiantz A. The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem 1994; 269: 10444-50. Elliott G, O'Hare P. Intercellular trafficking and protein delivery by a herpesvirus structural protein. Cell 1997; 88: 223-33. Rousselle C, Clair P, Lefauconnier JM, Kaczorek M, Scherrmann JM, Temsamani J. New advances in the transport of doxorubicin through the blood-brain barrier by a peptide vector-mediated strategy. Mol Pharmacol 2000; 57: 679-86. Elmquist A, Lindgren M, Bartfai T, Langel U. VE-cadherinderived cell-penetrating peptide, pVEC, with carrier functions. Exp Cell Res 2001; 269: 237-44. Foerg C, Ziegler U, Fernandez-Carneado J, et al. Decoding the entry of two novel cell-penetrating peptides in HeLa cells: lipid raft-mediated endocytosis and endosomal escape. Biochemistry 2005; 44: 72-81. Neundorf I, Rennert R, Franke J, Kozle I, Bergmann R. Detailed analysis concerning the biodistribution and metabolism of human calcitonin-derived cell-penetrating peptides. Bioconjug Chem 2008; 19: 1596-603. Niarchos DK, Perez SA, Papamichail M. Characterization of a novel cell penetrating peptide derived from Bag-1 protein. Peptides 2006; 27: 2661-9. Hu et al. [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] Duchardt F, Ruttekolk IR, Verdurmen WP, et al. A cell-penetrating peptide derived from human lactoferrin with conformationdependent uptake efficiency. J Biol Chem 2009; 284: 36099-108. Radis-Baptista G, de la Torre BG, Andreu D. A novel cellpenetrating peptide sequence derived by structural minimization of a snake toxin exhibits preferential nucleolar localization. J Med Chem 2008; 51: 7041-4. Jha D, Mishra R, Gottschalk S, et al. CyLoP-1: a novel cysteinerich cell-penetrating peptide for cytosolic delivery of cargoes. Bioconjug Chem 2011; 22: 319-28. Falanga A, Vitiello MT, Cantisani M, et al. A peptide derived from herpes simplex virus type 1 glycoprotein H: membrane translocation and applications to the delivery of quantum dots. Nanomedicine 2011; 7: 925-34. Oehlke J, Wallukat G, Wolf Y, et al. Enhancement of intracellular concentration and biological activity of PNA after conjugation with a cell-penetrating synthetic model peptide. Eur J Biochem 2004; 271: 3043-9. Rhee M, Davis P. Mechanism of uptake of C105Y, a novel cellpenetrating peptide. J Biol Chem 2006; 281: 1233-40. El-Andaloussi S, Johansson HJ, Holm T, Langel U. A novel cellpenetrating peptide, M918, for efficient delivery of proteins and peptide nucleic acids. Mol Ther 2007; 15: 1820-6. Sheng J, Oyler G, Zhou B, Janda K, Shoemaker CB. Identification and characterization of a novel cell-penetrating peptide. Biochem Biophys Res Commun 2009; 382: 236-40. Crombez L, Aldrian-Herrada G, Konate K, et al. A new potent secondary amphipathic cell-penetrating peptide for siRNA delivery into mammalian cells. Mol Ther 2009; 17: 95-103. Morris MC, Vidal P, Chaloin L, Heitz F, Divita G. A new peptide vector for efficient delivery of oligonucleotides into mammalian cells. Nucleic Acids Res 1997; 25: 2730-6. Soomets U, Lindgren M, Gallet X, et al. Deletion analogues of transportan. Biochim Biophys Acta 2000; 1467: 165-76. Morris MC, Depollier J, Mery J, Heitz F, Divita G. A peptide carrier for the delivery of biologically active proteins into mammalian cells. Nat Biotechnol 2001; 19: 1173-6. Mano M, Teodosio C, Paiva A, Simoes S, Pedroso de Lima MC. On the mechanisms of the internalization of S4(13)-PV cellpenetrating peptide. Biochem J 2005; 390: 603-12. Cao G, Pei W, Ge H, et al. 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 2002; 22: 5423-31. Green M, Loewenstein PM. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat transactivator protein. Cell 1988; 55: 1179-88. Fawell S, Seery J, Daikh Y, et al. Tat-mediated delivery of heterologous proteins into cells. Proc Natl Acad Sci USA 1994; 91: 664-8. Brooks H, Lebleu B, Vives E. Tat peptide-mediated cellular delivery: back to basics. Adv Drug Deliv Rev 2005; 57: 559-77. Vazquez J, Sun C, Du J, Fuentes L, Sumners C, Raizada MK. Transduction of a functional domain of the AT1 receptor in neurons by HIV-Tat PTD. Hypertension 2003; 41: 751-6. Flinterman M, Farzaneh F, Habib N, Malik F, Gaken J, Tavassoli M. Delivery of therapeutic proteins as secretable TAT fusion products. Mol Ther 2009; 17: 334-42. Yamano S, Dai J, Yuvienco C, Khapli S, Moursi AM, Montclare JK. Modified Tat peptide with cationic lipids enhances gene transfection efficiency via temperature-dependent and caveolaemediated endocytosis. J Control Release 2011; 152: 278-85. Eum WS, Kim DW, Hwang IK, et al. In vivo protein transduction: biologically active intact pep-1-superoxide dismutase fusion protein efficiently protects against ischemic insult. Free Radic Biol Med 2004; 37: 1656-69. Cho JH, Hwang IK, Yoo KY, et al. Effective delivery of Pep-1cargo protein into ischemic neurons and long-term neuroprotection of Pep-1-SOD1 against ischemic injury in the gerbil hippocampus. Neurochem Int 2008; 52: 659-68. Hwang IK, Eum WS, Yoo KY, et al. Copper chaperone for Cu,ZnSOD supplement potentiates the Cu,Zn-SOD function of neuroprotective effects against ischemic neuronal damage in the gerbil hippocampus. Free Radic Biol Med 2005; 39: 392-402. Delivery of Neurotherapeutics [53] [54] [55] [56] [57] [58] [59] [60] [61] [62] [63] [64] [65] [66] [67] [68] [69] [70] [71] [72] [73] [74] Yune TY, Lee JY, Jiang MH, Kim DW, Choi SY, Oh TH. Systemic administration of PEP-1-SOD1 fusion protein improves functional recovery by inhibition of neuronal cell death after spinal cord injury. Free Radic Biol Med 2008; 45: 1190-200. Kim W, Kim DW, Yoo DY, et al. Neuroprotective Effects of PEP1-Cu,Zn-SOD against Ischemic Neuronal Damage in the Rabbit Spinal Cord. Neurochem Res 2012; 37(2): 307-13. Choi SH, Kim DW, Kim SY, et al. Transduced human copper chaperone for Cu,Zn-SOD (PEP-1-CCS) protects against neuronal cell death. Mol Cells 2005; 20: 401-8. An JJ, Lee YP, Kim SY, et al. Transduced human PEP-1-heat shock protein 27 efficiently protects against brain ischemic insult. Febs J 2008; 275: 1296-308. Kim DW, Jeong HJ, Kang HW, et al. Transduced human PEP-1catalase fusion protein attenuates ischemic neuronal damage. Free Radic Biol Med 2009; 47: 941-52. Kim DW, Kim DS, Kim MJ, et al. Imipramine enhances neuroprotective effect of PEP-1-Catalase against ischemic neuronal damage. BMB Rep 2011; 44: 647-52. Perez F, Joliot A, Bloch-Gallego E, Zahraoui A, Triller A, Prochiantz A. Antennapedia homeobox as a signal for the cellular internalization and nuclear addressing of a small exogenous peptide. J Cell Sci 1992; 102 ( Pt 4): 717-22. Qian YQ, Billeter M, Otting G, Muller M, Gehring WJ, Wuthrich K. The structure of the Antennapedia homeodomain determined by NMR spectroscopy in solution: comparison with prokaryotic repressors. Cell 1989; 59: 573-80. Fan YF, Lu CZ, Xie J, Zhao YX, Yang GY. Apoptosis inhibition in ischemic brain by intraperitoneal PTD-BIR3-RING (XIAP). Neurochem Int 2006; 48: 50-9. Fan Y, Shi L, Gu Y, et al. Pretreatment with PTD-calbindin D 28k alleviates rat brain injury induced by ischemia and reperfusion. J Cereb Blood Flow Metab 2007; 27: 719-28. Huang R, Yang W, Jiang C, Pei Y. Gene delivery into brain capillary endothelial cells using Antp-modified DNA-loaded nanoparticles. Chem Pharm Bull (Tokyo) 2006; 54: 1254-8. Harwig SS, Swiderek KM, Lee TD, Lehrer RI. Determination of disulphide bridges in PG-2, an antimicrobial peptide from porcine leukocytes. J Pept Sci 1995; 1: 207-15. Aumelas A, Mangoni M, Roumestand C, et al. Synthesis and solution structure of the antimicrobial peptide protegrin-1. Eur J Biochem 1996; 237: 575-83. Sokolov Y, Mirzabekov T, Martin DW, Lehrer RI, Kagan BL. Membrane channel formation by antimicrobial protegrins. Biochim Biophys Acta 1999; 1420: 23-9. Rousselle C, Smirnova M, Clair P, et al. Enhanced delivery of doxorubicin into the brain via a peptide-vector-mediated strategy: saturation kinetics and specificity. J Pharmacol Exp Ther 2001; 296: 124-31. Rousselle C, Clair P, Temsamani J, Scherrmann JM. Improved brain delivery of benzylpenicillin with a peptide-vector-mediated strategy. J Drug Target 2002; 10: 309-15. Rousselle C, Clair P, Smirnova M, et al. Improved brain uptake and pharmacological activity of dalargin using a peptide-vectormediated strategy. J Pharmacol Exp Ther 2003; 306: 371-6. Drin G, Rousselle C, Scherrmann JM, Rees AR, Temsamani J. Peptide delivery to the brain via adsorptive-mediated endocytosis: advances with SynB vectors. AAPS PharmSci 2002; 4: E26. Hearst SM, Walker LR, Shao Q, Lopez M, Raucher D, Vig PJ. The design and delivery of a thermally responsive peptide to inhibit S100B-mediated neurodegeneration. Neuroscience 2011; 197: 36980. Richard JP, Melikov K, Vives E, et al. Cell-penetrating peptides. A reevaluation of the mechanism of cellular uptake. J Biol Chem 2003; 278: 585-90. Du L, Kayali R, Bertoni C, et al. Arginine-rich cell-penetrating peptide dramatically enhances AMO-mediated ATM aberrant splicing correction and enables delivery to brain and cerebellum. Hum Mol Genet 2011; 20: 3151-60. Kobayashi N, Niwa M, Hao Y, Yoshida T. Nucleolar localization signals of LIM kinase 2 function as a cell-penetrating peptide. Protein Pept Lett 2010; 17: 1480-8. Current Pharmaceutical Design, 2012, Vol. 18, No. 25 [75] [76] [77] [78] [79] [80] [81] [82] [83] [84] [85] [86] [87] [88] [89] [90] [91] [92] [93] [94] [95] [96] 3717 Jefferies WA, Brandon MR, Hunt SV, Williams AF, Gatter KC, Mason DY. Transferrin receptor on endothelium of brain capillaries. Nature 1984; 312: 162-3. Pardridge WM. Non-invasive drug delivery to the human brain using endogenous blood-brain barrier transport systems. Pharm Sci Technolo Today 1999; 2: 49-59. Zhang Y, Pardridge WM. Neuroprotection in transient focal brain ischemia after delayed intravenous administration of brain-derived neurotrophic factor conjugated to a blood-brain barrier drug targeting system. Stroke 2001; 32: 1378-84. Wu D, Pardridge WM. Neuroprotection with noninvasive neurotrophin delivery to the brain. Proc Natl Acad Sci USA 1999; 96: 254-9. Song BW, Vinters HV, Wu D, Pardridge WM. Enhanced neuroprotective effects of basic fibroblast growth factor in regional brain ischemia after conjugation to a blood-brain barrier delivery vector. J Pharmacol Exp Ther 2002; 301: 605-10. Boado RJ, Hui EK, Lu JZ, Pardridge WM. Drug targeting of erythropoietin across the primate blood-brain barrier with an IgG molecular Trojan horse. J Pharmacol Exp Ther; 333: 961-9. Fu A, Hui EK, Lu JZ, Boado RJ, Pardridge WM. Neuroprotection in experimental stroke in the rat with an IgG-erythropoietin fusion protein. Brain Res 2010; 1360: 193-7. Boado RJ, Zhang Y, Wang Y, Pardridge WM. GDNF fusion protein for targeted-drug delivery across the human blood-brain barrier. Biotechnol Bioeng 2008; 100: 387-96. Asoh S, Ohsawa I, Mori T, et al. Protection against ischemic brain injury by protein therapeutics. Proc Natl Acad Sci USA 2002; 99: 17107-12. Kreuter J, Ramge P, Petrov V, et al. Direct evidence that polysorbate-80-coated poly(butylcyanoacrylate) nanoparticles deliver drugs to the CNS via specific mechanisms requiring prior binding of drug to the nanoparticles. Pharm Res 2003; 20: 409-16. Ulbrich K, Hekmatara T, Herbert E, Kreuter J. Transferrin- and transferrin-receptor-antibody-modified nanoparticles enable drug delivery across the blood-brain barrier (BBB). Eur J Pharm Biopharm 2009; 71: 251-6. Michaelis K, Hoffmann MM, Dreis S, et al. Covalent linkage of apolipoprotein e to albumin nanoparticles strongly enhances drug transport into the brain. J Pharmacol Exp Ther 2006; 317: 1246-53. Gelperina SE, Khalansky AS, Skidan IN, et al. Toxicological studies of doxorubicin bound to polysorbate 80-coated poly(butyl cyanoacrylate) nanoparticles in healthy rats and rats with intracranial glioblastoma. Toxicol Lett 2002; 126: 131-41. Reddy MK, Labhasetwar V. Nanoparticle-mediated delivery of superoxide dismutase to the brain: an effective strategy to reduce ischemia-reperfusion injury. FASEB J 2009; 23: 1384-95. Agemy L, Friedmann-Morvinski D, Kotamraju VR, et al. Targeted nanoparticle enhanced proapoptotic peptide as potential therapy for glioblastoma. Proc Natl Acad Sci USA; 108: 17450-5. Wilson B, Samanta MK, Santhi K, Kumar KP, Paramakrishnan N, Suresh B. Poly(n-butylcyanoacrylate) nanoparticles coated with polysorbate 80 for the targeted delivery of rivastigmine into the brain to treat Alzheimer's disease. Brain Res 2008; 1200: 159-68. Kurakhmaeva KB, Djindjikhashvili IA, Petrov VE, et al. Brain targeting of nerve growth factor using poly(butyl cyanoacrylate) nanoparticles. J Drug Target 2009; 17: 564-74. Hillaireau H, Couvreur P. Nanocarriers' entry into the cell: relevance to drug delivery. Cell Mol Life Sci 2009; 66: 2873-96. Kreuter J, Shamenkov D, Petrov V, et al. Apolipoprotein-mediated transport of nanoparticle-bound drugs across the blood-brain barrier. J Drug Target 2002; 10: 317-25. Biju K, Zhou Q, Li G, et al. Macrophage-mediated GDNF delivery protects against dopaminergic neurodegeneration: a therapeutic strategy for Parkinson's disease. Mol Ther 2010; 18: 1536-44. Baek SK, Makkouk AR, Krasieva T, Sun CH, Madsen SJ, Hirschberg H. Photothermal treatment of glioma; an in vitro study of macrophage-mediated delivery of gold nanoshells. J Neurooncol 2011; 104: 439-48. Barone FC, Feuerstein GZ. Inflammatory mediators and stroke: new opportunities for novel therapeutics. J Cereb Blood Flow Metab 1999; 19: 819-34. 3718 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 [97] [98] [99] [100] [101] [102] [103] [104] [105] [106] [107] [108] [109] [110] [111] [112] [113] [114] [115] [116] [117] Cai B, Lin Y, Xue XH, Fang L, Wang N, Wu ZY. TAT-mediated delivery of neuroglobin protects against focal cerebral ischemia in mice. Exp Neurol 2011; 227: 224-31. Kilic E, Dietz GP, Hermann DM, Bahr M. Intravenous TAT-BclXl is protective after middle cerebral artery occlusion in mice. Ann Neurol 2002; 52: 617-22. Kilic U, Kilic E, Dietz GP, Bahr M. Intravenous TAT-GDNF is protective after focal cerebral ischemia in mice. Stroke 2003; 34: 1304-10. Guegan C, Braudeau J, Couriaud C, et al. PTD-XIAP protects against cerebral ischemia by anti-apoptotic and transcriptional regulatory mechanisms. Neurobiol Dis 2006; 22: 177-86. Bright R, Steinberg GK, Mochly-Rosen D. DeltaPKC mediates microcerebrovascular dysfunction in acute ischemia and in chronic hypertensive stress in vivo. Brain Res 2007; 1144: 146-55. Doeppner TR, Dietz GP, El Aanbouri M, et al. TAT-Bcl-x(L) improves survival of neuronal precursor cells in the lesioned striatum after focal cerebral ischemia. Neurobiol Dis 2009; 34: 8794. Doeppner TR, El Aanbouri M, Dietz GP, Weise J, Schwarting S, Bahr M. Transplantation of TAT-Bcl-xL-transduced neural precursor cells: long-term neuroprotection after stroke. Neurobiol Dis; 40: 265-76. Backman CM, Shan L, Zhang YJ, et al. Gene expression patterns for GDNF and its receptors in the human putamen affected by Parkinson's disease: a real-time PCR study. Mol Cell Endocrinol 2006; 252: 160-6. Brines ML, Ghezzi P, Keenan S, et al. Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proc Natl Acad Sci USA 2000; 97: 10526-31. Ehrenreich H, Hasselblatt M, Dembowski C, et al. Erythropoietin therapy for acute stroke is both safe and beneficial. Mol Med 2002; 8: 495-505. Brines M, Grasso G, Fiordaliso F, et al. Erythropoietin mediates tissue protection through an erythropoietin and common betasubunit heteroreceptor. Proc Natl Acad Sci USA 2004; 101: 1490712. Zhang F, Signore AP, Zhou Z, Wang S, Cao G, Chen J. Erythropoietin protects CA1 neurons against global cerebral ischemia in rat: potential signaling mechanisms. J Neurosci Res 2006; 83: 1241-51. Koponen S, Goldsteins G, Keinanen R, Koistinaho J. Induction of protein kinase Cdelta subspecies in neurons and microglia after transient global brain ischemia. J Cereb Blood Flow Metab 2000; 20: 93-102. Brodie C, Blumberg PM. Regulation of cell apoptosis by protein kinase c delta. Apoptosis 2003; 8: 19-27. Majumder PK, Pandey P, Sun X, et al. Mitochondrial translocation of protein kinase C delta in phorbol ester-induced cytochrome c release and apoptosis. J Biol Chem 2000; 275: 21793-6. Bates E, Bode C, Costa M, et al. Intracoronary KAI-9803 as an adjunct to primary percutaneous coronary intervention for acute ST-segment elevation myocardial infarction. Circulation 2008; 117: 886-96. Shimohata T, Zhao H, Steinberg GK. Epsilon PKC may contribute to the protective effect of hypothermia in a rat focal cerebral ischemia model. Stroke 2007; 38: 375-80. Della-Morte D, Raval AP, Dave KR, Lin HW, Perez-Pinzon MA. Post-ischemic activation of protein kinase C epsilon protects the hippocampus from cerebral ischemic injury via alterations in cerebral blood flow. Neurosci Lett 2011; 487: 158-62. Borsello T, Clarke PG, Hirt L, et al. A peptide inhibitor of c-Jun Nterminal kinase protects against excitotoxicity and cerebral ischemia. Nat Med 2003; 9: 1180-6. Repici M, Centeno C, Tomasi S, et al. Time-course of c-Jun Nterminal kinase activation after cerebral ischemia and effect of DJNKI1 on c-Jun and caspase-3 activation. Neuroscience 2007; 150: 40-9. Wiegler K, Bonny C, Coquoz D, Hirt L. The JNK inhibitor XG102 protects from ischemic damage with delayed intravenous administration also in the presence of recombinant tissue plasminogen activator. Cerebrovasc Dis 2008; 26: 360-6. Hu et al. [118] [119] [120] [121] [122] [123] [124] [125] [126] [127] [128] [129] [130] [131] [132] [133] [134] [135] [136] [137] [138] Sheng M. The postsynaptic NMDA-receptor--PSD-95 signaling complex in excitatory synapses of the brain. J Cell Sci 2001; 114: 1251. Aarts M, Liu Y, Liu L, et al. Treatment of ischemic brain damage by perturbing NMDA receptor- PSD-95 protein interactions. Science 2002; 298: 846-50. Sun HS, Doucette TA, Liu Y, et al. Effectiveness of PSD95 inhibitors in permanent and transient focal ischemia in the rat. Stroke 2008; 39: 2544-53. Sun Y, Jin K, Mao XO, Zhu Y, Greenberg DA. Neuroglobin is upregulated by and protects neurons from hypoxic-ischemic injury. Proc Natl Acad Sci USA 2001; 98: 15306-11. Gou X, Wang Q, Yang Q, Xu L, Xiong L. TAT-NEP1-40 as a novel therapeutic candidate for axonal regeneration and functional recovery after stroke. J Drug Target 2011; 19: 86-95. Wang Q, Gou X, Xiong L, et al. Trans-activator of transcriptionmediated delivery of NEP1-40 protein into brain has a neuroprotective effect against focal cerebral ischemic injury via inhibition of neuronal apoptosis. Anesthesiology 2008; 108: 107180. Doeppner TR, Nagel F, Dietz GP, et al. TAT-Hsp70-mediated neuroprotection and increased survival of neuronal precursor cells after focal cerebral ischemia in mice. J Cereb Blood Flow Metab 2009; 29: 1187-96. Ye N, Liu S, Lin Y, Rao P. Protective effects of intraperitoneal injection of TAT-SOD against focal cerebral ischemia/reperfusion injury in rats. Life Sci 2011; 89: 868-74. Yin W, Cao G, Johnnides MJ, et al. TAT-mediated delivery of BclxL protein is neuroprotective against neonatal hypoxic-ischemic brain injury via inhibition of caspases and AIF. Neurobiol Dis 2006; 21: 358-71. Dietz GP, Valbuena PC, Dietz B, et al. Application of a bloodbrain-barrier-penetrating form of GDNF in a mouse model for Parkinson's disease. Brain Res 2006; 1082: 61-6. Nijboer CH, van der Kooij MA, van Bel F, Ohl F, Heijnen CJ, Kavelaars A. Inhibition of the JNK/AP-1 pathway reduces neuronal death and improves behavioral outcome after neonatal hypoxicischemic brain injury. Brain Behav Immun 2010; 24: 812-21. Dykstra CM, Ratnam M, Gurd JW. Neuroprotection after status epilepticus by targeting protein interactions with postsynaptic density protein 95. J Neuropathol Exp Neurol 2009; 68: 823-31. Nagel F, Falkenburger BH, Tonges L, et al. Tat-Hsp70 protects dopaminergic neurons in midbrain cultures and in the substantia nigra in models of Parkinson's disease. J Neurochem 2008; 105: 853-64. Kim DW, Eum WS, Jang SH, et al. Transduced Tat-SOD fusion protein protects against ischemic brain injury. Mol Cells 2005; 19: 88-96. Gao Y, Liang W, Hu X, et al. Neuroprotection against hypoxicischemic brain injury by inhibiting the apoptotic protease activating factor-1 pathway. Stroke 2010; 41: 166-72. Nijboer CH, Heijnen CJ, Groenendaal F, van Bel F, Kavelaars A. Alternate pathways preserve tumor necrosis factor-alpha production after nuclear factor-kappaB inhibition in neonatal cerebral hypoxia-ischemia. Stroke 2009; 40: 3362-8. Alves ID, Jiao CY, Aubry S, et al. Cell biology meets biophysics to unveil the different mechanisms of penetratin internalization in cells. Biochim Biophys Acta 2010; 1798: 2231-9. Kim DW, Lee SH, Jeong MS, et al. Transduced Tat-SAG fusion protein protects against oxidative stress and brain ischemic insult. Free Radic Biol Med 2010; 48: 969-77. Zhang C, Su Z, Zhao B, et al. Tat-modified leptin is more accessible to hypothalamus through brain-blood barrier with a significant inhibition of body-weight gain in high-fat-diet fed mice. Exp Clin Endocrinol Diabetes 2010; 118: 31-7. Rapoport M, Salman L, Sabag O, Patel MS, Lorberboum-Galski H. Successful TAT-mediated enzyme replacement therapy in a mouse model of mitochondrial E3 deficiency. J Mol Med (Berl) 2011; 89: 161-70. Smith DL, Pozueta J, Gong B, Arancio O, Shelanski M. Reversal of long-term dendritic spine alterations in Alzheimer disease models. Proc Natl Acad Sci USA 2009; 106: 16877-82. Delivery of Neurotherapeutics [139] [140] [141] [142] [143] [144] [145] [146] [147] [148] [149] [150] [151] [152] [153] [154] [155] [156] [157] [158] [159] Qu HY, Zhang T, Li XL, et al. Transducible P11-CNTF rescues the learning and memory impairments induced by amyloid-beta peptide in mice. Eur J Pharmacol 2008; 594: 93-100. Fu AL, Li Q, Dong ZH, Huang SJ, Wang YX, Sun MJ. Alternative therapy of Alzheimer's disease via supplementation with choline acetyltransferase. Neurosci Lett 2004; 368: 258-62. Berger C, Gremlich HU, Schmidt P, et al. In vivo monitoring the fate of Cy5.5-Tat labeled T lymphocytes by quantitative nearinfrared fluorescence imaging during acute brain inflammation in a rat model of experimental autoimmune encephalomyelitis. J Immunol Methods 2007; 323: 65-77. Santra S, Yang H, Holloway PH, Stanley JT, Mericle RA. Synthesis of water-dispersible fluorescent, radio-opaque, and paramagnetic CdS:Mn/ZnS quantum dots: a multifunctional probe for bioimaging. J Am Chem Soc 2005; 127: 1656-7. Santra S, Yang H, Dutta D, et al. TAT conjugated, FITC doped silica nanoparticles for bioimaging applications. Chem Commun (Camb) 2004: 2810-1. Qin Y, Chen H, Yuan W, et al. Liposome formulated with TATmodified cholesterol for enhancing the brain delivery. Int J Pharm 2011; 419: 85-95. Kanazawa T, Taki H, Tanaka K, Takashima Y, Okada H. Cellpenetrating peptide-modified block copolymer micelles promote direct brain delivery via intranasal administration. Pharm Res 2011; 28: 2130-9. Wang H, Xu K, Liu L, et al. The efficacy of self-assembled cationic antimicrobial peptide nanoparticles against Cryptococcus neoformans for the treatment of meningitis. Biomaterials 2010; 31: 2874-81. Liu L, Venkatraman SS, Yang YY, et al. Polymeric micelles anchored with TAT for delivery of antibiotics across the bloodbrain barrier. Biopolymers 2008; 90: 617-23. Liu L, Guo K, Lu J, et al. Biologically active core/shell nanoparticles self-assembled from cholesterol-terminated PEGTAT for drug delivery across the blood-brain barrier. Biomaterials 2008; 29: 1509-17. Rao KS, Reddy MK, Horning JL, Labhasetwar V. TAT-conjugated nanoparticles for the CNS delivery of anti-HIV drugs. Biomaterials 2008; 29: 4429-38. Nakase I, Niwa M, Takeuchi T, et al. Cellular uptake of argininerich peptides: roles for macropinocytosis and actin rearrangement. Mol Ther 2004; 10: 1011-22. Lu JZ, Boado RJ, Hui EK, Zhou QH, Pardridge WM. Expression in CHO cells and pharmacokinetics and brain uptake in the Rhesus monkey of an IgG-iduronate-2-sulfatase fusion protein. Biotechnol Bioeng 2011; 108: 1954-64. Boado RJ, Lu JZ, Hui EK, Pardridge WM. IgG-single chain Fv fusion protein therapeutic for Alzheimer's disease: Expression in CHO cells and pharmacokinetics and brain delivery in the rhesus monkey. Biotechnol Bioeng; 105: 627-35. Futaki S, Suzuki T, Ohashi W, et al. Arginine-rich peptides. An abundant source of membrane-permeable peptides having potential as carriers for intracellular protein delivery. J Biol Chem 2001; 276: 5836-40. Mai JC, Shen H, Watkins SC, Cheng T, Robbins PD. Efficiency of protein transduction is cell type-dependent and is enhanced by dextran sulfate. J Biol Chem 2002; 277: 30208-18. Suzuki T, Futaki S, Niwa M, Tanaka S, Ueda K, Sugiura Y. Possible existence of common internalization mechanisms among arginine-rich peptides. J Biol Chem 2002; 277: 2437-43. Derossi D, Calvet S, Trembleau A, Brunissen A, Chassaing G, Prochiantz A. Cell internalization of the third helix of the Antennapedia homeodomain is receptor-independent. J Biol Chem 1996; 271: 18188-93. Deshayes S, Gerbal-Chaloin S, Morris MC, et al. On the mechanism of non-endosomial peptide-mediated cellular delivery of nucleic acids. Biochim Biophys Acta 2004; 1667: 141-7. Prochiantz A. Messenger proteins: homeoproteins, TAT and others. Curr Opin Cell Biol 2000; 12: 400-6. Herce HD, Garcia AE. Molecular dynamics simulations suggest a mechanism for translocation of the HIV-1 TAT peptide across lipid membranes. Proc Natl Acad Sci USA 2007; 104: 20805-10. Current Pharmaceutical Design, 2012, Vol. 18, No. 25 [160] [161] [162] [163] [164] [165] [166] [167] [168] [169] [170] [171] [172] [173] [174] [175] [176] [177] [178] [179] [180] [181] [182] 3719 Ter-Avetisyan G, Tunnemann G, Nowak D, et al. Cell entry of arginine-rich peptides is independent of endocytosis. J Biol Chem 2009; 284: 3370-8. Fittipaldi A, Ferrari A, Zoppe M, et al. Cell membrane lipid rafts mediate caveolar endocytosis of HIV-1 Tat fusion proteins. J Biol Chem 2003; 278: 34141-9. Richard JP, Melikov K, Brooks H, Prevot P, Lebleu B, Chernomordik LV. Cellular uptake of unconjugated TAT peptide involves clathrin-dependent endocytosis and heparan sulfate receptors. J Biol Chem 2005; 280: 15300-6. Wadia JS, Stan RV, Dowdy SF. Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat Med 2004; 10: 310-5. Kaplan IM, Wadia JS, Dowdy SF. Cationic TAT peptide transduction domain enters cells by macropinocytosis. J Control Release 2005; 102: 247-53. Khalil IA, Kogure K, Futaki S, Harashima H. High density of octaarginine stimulates macropinocytosis leading to efficient intracellular trafficking for gene expression. J Biol Chem 2006; 281: 3544-51. Saalik P, Elmquist A, Hansen M, et al. Protein cargo delivery properties of cell-penetrating peptides. A comparative study. Bioconjug Chem 2004; 15: 1246-53. Chakrabarti R, Wylie DE, Schuster SM. Transfer of monoclonal antibodies into mammalian cells by electroporation. J Biol Chem 1989; 264: 15494-500. Arnheiter H, Haller O. Antiviral state against influenza virus neutralized by microinjection of antibodies to interferon-induced Mx proteins. Embo J 1988; 7: 1315-20. Lee YJ, Johnson G, Peltier GC, Pellois JP. A HA2-Fusion tag limits the endosomal release of its protein cargo despite causing endosomal lysis. Biochim Biophys Acta 2011; 1810: 752-8. Derossi D, Chassaing G, Prochiantz A. Trojan peptides: the penetratin system for intracellular delivery. Trends Cell Biol 1998; 8: 84-7. Kawamoto S, Takasu M, Miyakawa T, et al. Inverted micelle formation of cell-penetrating peptide studied by coarse-grained simulation: importance of attractive force between cell-penetrating peptides and lipid head group. J Chem Phys 2011; 134: 095103. Simeoni F, Morris MC, Heitz F, Divita G. Insight into the mechanism of the peptide-based gene delivery system MPG: implications for delivery of siRNA into mammalian cells. Nucleic Acids Res 2003; 31: 2717-24. Binder C, Read SP, Cashman SM, Kumar-Singh R. Nuclear targeted delivery of macromolecules to retina and cornea. J Gene Med 2011; 13: 158-70. Del Gaizo V, Payne RM. A novel TAT-mitochondrial signal sequence fusion protein is processed, stays in mitochondria, and crosses the placenta. Mol Ther 2003; 7: 720-30. Horton KL, Stewart KM, Fonseca SB, Guo Q, Kelley SO. Mitochondria-penetrating peptides. Chem Biol 2008; 15: 375-82. Zhao K, Luo G, Giannelli S, Szeto HH. Mitochondria-targeted peptide prevents mitochondrial depolarization and apoptosis induced by tert-butyl hydroperoxide in neuronal cell lines. Biochem Pharmacol 2005; 70: 1796-806. Xia H, Gao X, Gu G, L et al. Low molecular weight protaminefunctionalized nanoparticles for drug delivery to the brain after intranasal administration. Biomaterials 2011; 32: 9888-98. Ko YT, Hartner WC, Kale A, Torchilin VP. Gene delivery into ischemic myocardium by double-targeted lipoplexes with antimyosin antibody and TAT peptide. Gene Ther 2009; 16: 52-9. Nagahara H, Vocero-Akbani AM, Snyder EL, et al. Transduction of full-length TAT fusion proteins into mammalian cells: TATp27Kip1 induces cell migration. Nat Med 1998; 4: 1449-52. Chauhan A, Tikoo A, Kapur AK, Singh M. The taming of the cell penetrating domain of the HIV Tat: myths and realities. J Control Release 2007; 117: 148-62. Grunwald J, Rejtar T, Sawant R, Wang Z, Torchilin VP. TAT peptide and its conjugates: proteolytic stability. Bioconjug Chem 2009; 20: 1531-7. Pujals S, Fernandez-Carneado J, Ludevid MD, Giralt E. D-SAP: a new, noncytotoxic, and fully protease resistant cell-penetrating peptide. ChemMedChem 2008; 3: 296-301. 3720 Current Pharmaceutical Design, 2012, Vol. 18, No. 25 [183] [184] [185] Achilefu S, Srinivasan A, Schmidt MA, Jimenez HN, Bugaj JE, Erion JL. Novel bioactive and stable neurotensin peptide analogues capable of delivering radiopharmaceuticals and molecular beacons to tumors. J Med Chem 2003; 46: 3403-11. Meng H, Kumar K. Antimicrobial activity and protease stability of peptides containing fluorinated amino acids. J Am Chem Soc 2007; 129: 15615-22. Kilk K, Mahlapuu R, Soomets U, Langel U. Analysis of in vitro toxicity of five cell-penetrating peptides by metabolic profiling. Toxicology 2009; 265: 87-95. Received: January 11, 2012 Accepted: January 24, 2012 Hu et al. [186] [187] [188] Cardozo AK, Buchillier V, Mathieu M, et al. Cell-permeable peptides induce dose- and length-dependent cytotoxic effects. Biochim Biophys Acta 2007; 1768: 2222-34. El-Andaloussi S, Jarver P, Johansson HJ, Langel U. Cargodependent cytotoxicity and delivery efficacy of cell-penetrating peptides: a comparative study. Biochem J 2007; 407: 285-92. Herve F, Ghinea N, Scherrmann JM. CNS delivery via adsorptive transcytosis. Aaps J 2008; 10: 455-72.