Sunteți pe pagina 1din 19

Journal of Controlled Release 261 (2017) 4361

Contents lists available at ScienceDirect

Journal of Controlled Release


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

Review article

Self-assembled amphiphilic core-shell nanocarriers in line with the modern MARK


strategies for brain delivery
Reham S. Elezaby, Heba A. Gad, Abdelkader A. Metwally, Ahmed S. Geneidi, Gehanne A. Awad
Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt

A R T I C L E I N F O A B S T R A C T

Keywords: Disorders of the central nervous system (CNS) represent increasing social and economic problems all over the
Brain targeting world which makes the eective transport of drugs to the brain a crucial need. In the last decade, many strategies
Core-shell nanocarriers were introduced to deliver drugs to the brain trying to overcome the challenge of the blood brain barrier (BBB)
Micelles using both invasive and non-invasive methods. Non-invasive strategy represented in the application of nano-
Nanotechnology
carriers became very common. One of the most hopeful nanoscopic carriers for brain delivery is core-shell
nanocarriers or polymeric micelles (PMs). They are more advantageous than other nanocarriers. They oer small
size, ease of preparation, ease of sterilization and the possibility of surface modication with various ligands.

Abbreviations: 17-AAG, 17-Allylamino-17-demethoxy geldanamycin; 5-FU, 5-ourouracil; AAV, adeno-associated virus; ABC, ATP binding cassette; Ach, acetyl choline; AFM, atomic
force microscopy; AmB, amphotericin B; AMT, adsorbtive-mediated transcytosis; APN, aminopeptidase N; Apo, apolipoprotein; ATRA, all-trans retinoic acid; BBB, blood brain barrier;
BCECs, brain capillary endothelial cells; BCNU, bis-chloroethylnitrosourea; BCSFB, bloodcerebrospinal uid barrier; BMECs, brain microvascular endothelial cells; BS, bile salt; BVECs,
brain vascular endothelial cells; CA8, dendritic octamer of cholic acid; CAC, critical aggregation concentration; CD, choline derivatives; CDs, cyclodextrins; cl-micelles, cross linked
micelles; CIMS, core-inversible micelles; CMC, critical micelle concentration; CMT, carrier-mediated transport; CNS, central nervous system; CPPs, cell penetrating peptides; CPT,
captothecin; cRAD, cyclic-arginine-alanine-aspartic acid; CREKA, cysteine-arginine-glutamic acid-lysine-alanine; cRGD, cyclic Arginine-Glycine-Aspartic acid; CRM, cross reacting ma-
terial; CSF, cerebrospinal uid; CS-SA, stearic acid-grafted chitosan; CT, X-ray computed tomography; DACHPt, (1,2-diaminocyclohexane) platinum(II); DHA, dehydroascorbic acid; DiR,
near-infrared uorescent dye; DL, drug loading; DLS, dynamic light scattering; DOM, domperidone; DOPE, dioleolyphosphatidyl ethanolamine; DOX, doxycycline; DPA, 3,30-dithio-
dipropionic acid; DPc, dendrimerphthalocyanine; DSPE, distearoylphophatidyl ethanolamine; DSPE-PEG, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-methoxy(polyethylene
glycol); DT, diphtheria toxin; DTR, diphtheria toxin receptor; DTSSP, 3,3-dithiobis(sulfosuccinimidylpropionate); DTX, docetaxel; ED, 1,2-ethylenediamine; EDC, 1-(3-dimethylami-
nopropyl)-3-ethylcarbodiimidehydrochloride; EE, encapsulation eciency; EPR, enhanced permeability and retention eect; FA, folic acid; GBM, glioblastoma multiforme; GLUT,
glucose transporter; GLUT-1 scFv, GLUT1 antibody single chain fragment variable; GTA, glycidyltrimethylammonium chloride; HIV, human immunodeciency virus; HRP, horseradish
peroxidase; HSA, human serum albumin; HSVtk, herpes simplex virus thymidine kinase gene; IAs, inhaled anaesthetics; ITZ, itraconazole; LAT1, large neutral amino-acid transporter type
1; LCST, lower critical solution temperature; LINCL, late infantile neuronal ceroidlipofuscinosis; LRP, lipoprotein receptor-related protein; Lt, lactoferrin; LTG, lamotrigine; LtR, lacto-
ferrin receptor; mAbs, monoclonal antibodies; Mal, maliemide; MAN, aminophenyl--d-mannopyranoside; mPEG-b-P(Glu), poly(ethyleneglycol)-b-poly(L-glutamic acid); mPEG-b-PCL-b-
PPEEA, monomethoxy poly(ethylene glycol)-block-poly(-caprolactone)-block-poly(2-aminoethyl ethylene phosphate); mPEG-OA, monomethoxy poly(ethylene glycol)-oleate; mPEG-
PAHy-GTA, methoxy poly(ethylene glycol)-,-polyasparthydrazide-glycidyltrimethylammonium; m-PEG-PDMAEMA, methoxy poly(ethylene glycol)-poly(2-(dimethylamino)ethyl
methacrylate; mPEG-St, mPEGylated starch; MRI, magnetic resonance imaging; MRP, multidrug resistant protein; MRSA, methicillin-resistant Staphylococcus aureus; NGR, Asn-Gly-Arg
peptide; NHS, n-hydroxysuccinimidyl; NIR, near-infrared; NPs, nanoparticles; OX26, an anti-TfR antibody; PC, phosphatidylcholine; PCL-b-p(NIPAAm-co-DMA), poly(-caprolactone)-
block-poly(N-isopropylacrylamide-co-N,N-dimethylacrylamide); PCL-PEEP, poly(-caprolactone)-block-poly(ethyl ethylene phosphate); PDI, polydispersity index; PEG, poly(ethylene
glycol); PEG-b-Chol, poly(ethylene glycol)-block-cholesterol; PEG-b-P(NIPAAm-co-NHPAAm), poly(ethylene glycol)-block-poly(N-isopropylacrylamide-co-N-(4-hydroxyphenethyl)acry-
lamide); PEG-b-PCL, poly(ethylene glycol)-block-poly(-caprolactone); PEG-b-PLA, poly(ethylene glycol)-block-poly(lactic acid); PEG-b-PLL, poly(ethylene glycol)-block-poly(L-lysine);
PEG-b-PMA, poly(ethylene glycol)-block-poly(methacrylic acid); PEG-PAE-API, poly(ethylene glycol)-poly(b-amino ester)-1-(3-aminopropyl) imidazole; PEG-PBLA, poly(ethylene glycol)-
poly(b-benzyl-L-aspartate; PEGPGluPPhe, poly(ethylene glycol)-poly(glutamic acid)-poly(phenylalanine); PEG-pLys-pPhe, poly(ethylene glycol)-b-poly(L-lysine)-b-poly(L-phenylala-
nine); PE-mPEG, 1,2 distearoyl-snglycero-3-phosphoethanolamine-N-(methoxy(polyethylene glycol)); PEO, polyethylene oxide; P-gp, P-glycoprotein; PHEA-EDA-PLA, ,-poly((N-hy-
droxyethyl)-DL-aspartamide)-ethylenediamine-poly(lactic acid); pHPMA-co-pLMA, poly(N-(2-hydroxypropyl)-methacrylamide)-poly(lauryl methacrylate); PIC, polyion complex; PLA-b-
PDMAEMA, poly(lactic acid)-poly(dimethylaminoethyl methacrylate; PMs, polymeric micelles; pNIPAAm, poly(N-isopropyl acrylamide); PPO, polypropylene oxide; PS80, polysorbate 80;
PTX, paclitaxel; R3V6, 3-arginine and 6-valine; R7L10, 7-arginine and 10-leucine; RAFT, reversible addition-fragmentation chain transfer; RES, reticuloendothelal system; RMT, receptor-
mediated transcytosis; ROP, metal-free organo catalyticpolymerization; RT, radiation therapy; RVG, rabies virus glycoprotein; SACDs, sodium alginate cholesterol derivatives; SA-g-
pNIPAAm, sodium alginate-graft-poly(N-isopropylacrylamide); SAH, subarachnoid hemorrhage; SDS, sodium dodecyl sulphate; siRNA, small interfering RNA; SMA, poly(styrene-co-
maleic anhydride); SPIONs, superparamagnetic iron oxide nanoparticles; sPMI, stapled peptide antagonist of both MDM2 and MDMX; T7, His-Ala-Ile-Tyr-Pro-Arg-His peptide; Tat, trans-
activating transcriptional activator; Td, tryptophan derivative; TEER, transendothelial electrical resistance; TEM, transmission electron microscope; Tf, transferrin; TFF3, trefoil factor 3;
TfR, transferrin receptor; TGN, TGNYKALHPHNG peptide; Tjs, tight junctions; TMZ, temozolomide; TPGS, D-alphatocopheryl polyethylene glycol succinate; UV, ultra-violet; VEGF-
siRNA, vascular endothelial growth factor-small interfering RNA; YC1, 5-cholestane-24-methylene-3,5,6,19-tetraol; Zot, zonulaoccludens toxin

Corresponding author.
E-mail address: reham_samir@pharma.asu.edu.eg (R.S. Elezaby).

http://dx.doi.org/10.1016/j.jconrel.2017.06.019
Received 17 March 2017; Received in revised form 16 June 2017; Accepted 20 June 2017
Available online 23 June 2017
0168-3659/ 2017 Elsevier B.V. All rights reserved.
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

Hence, the aim of this review is to discuss modern strategies for brain delivery, micelles as a successful delivery
system for the brain and how micelles could be modied to act as magic bullets for brain delivery.

1. Introduction 2. Challenge of the BBB

Diseases can aect both brain and spinal cord causing central ner- Being of selective nature, BBB oers to the brain protective func-
vous system (CNS) neurological or psychiatric disorders. CNS diseases tions against noxious circulating substances, while at the same time
are numerous and can be classied under three categories [1,2]. The aording needed nutrients for an adequate brain function [5]. It is
rst category includes neurological infectious diseases caused by bac- physiologically made up of three layers; the rst is the inner endothelial
teria, viruses and fungi such as meningitis, encephalitis, Lyme en- cell layer forming the capillary wall and containing tight junctions
cephalopathy, CreutzfeldJakob and cerebral toxoplasmosis. The (Tjs); the second is the basement membrane and nally the third layer
second category includes neurological disorders such as Alzheimer's composed of pericytes and astrocytic feet processes lying on the base-
disease, Parkinson's disease, epilepsy, seizures, dementia, depression, ment membrane [2] (Fig. 1). These whole anatomical and physiological
multiple sclerosis, mononeuropathy and polyneuropathy. The last ca- properties generate the concept of BBB.
tegory involves brain tumors and physical injuries such as cerebral Tjs, acting as a physical barrier, are formed and regulated by claudins
tumors, thrombosis, embolism, hemorrhage and vasculitis. CNS dis- (e.g. claudins 1 and 5), occluding, annexin-1 and junction adhesives,
orders represent an increasing problem both socially and economically which are protein molecules linked together by zonulaoccludens and
all over the word with regard to both the number of patients and the cingulin to the beta-actin cytoskeleton. They are lling the gap within
cost of therapy. brain vascular endothelial cells creating the tight junctions. This will
So far the CNS disorders are responsible for almost 11% of disease not only reduce the paracellular movement of substances, but also will
burden - as they aect 1.5 billion people - and about 1% of deaths contribute to the high transendothelial electrical resistance (TEER)
worldwide [3]. Hence, ecient treatment of CNS diseases became a of > 1500 cm2 [2,8].
crucial issue. Despite the presence of relatively high blood ow to the Astrocytes and pericytes play essential role in maintaining BBB
CNS (total surface area of 20 m2 and a total length of 400 miles), the function in healthy CNS and following injuries. Astrocytes main roles
eective targeting presents a great problem [4]. are in the healthy CNS and include blood ow regulation, nutrients
The two main barriers hinder the brain delivery are the bloodbrain provision and assistance of extracellular ion balance. Moreover, fol-
barrier (BBB) and the bloodcerebrospinal uid barrier (BCSFB), with lowing traumatic injuries, it has a fundamental eect on the reforma-
the rst one regarded as the most important obstacle to the brain tion of the damaged brain and spinal cord [5,14]. Pericytes, responsible
parenchyma [57]. Generally, invasive strategies were oftenly used [7]. for growth factors secretion or changes in the extracellular matrix, are
Lately, non-invasive strategies involving nanocarriers delivery became important in the maturation, renovation and conservation of the vas-
the most targeted strategy [8]. Polymeric micelles (PMs) as self-as- cular system. Besides, their participation in molecules delivery across
sembled amphiphilic core-shell nanocarriers are among the promising the BBB and vascular permeability control is well dened [5,15].
nanoscopic carriers [9,10]. During the last decade, numerous studies The concept of biochemical barrier is established from the occupancy
utilized PMs as an ecient drug delivery system for brain delivery of many receptors, transporter proteins and eux pumps that permit
[1113]. In this review, we will highlight the modern strategies for the passage of certain vital substances into the brain and prevent the
brain delivery. Likewise, an in-depth focus on PMs as promising na- others [17]. Actually, during physiological and pathological processes,
nocarriers for brain targeting will be upraised. The review will em- BBB is included in several functions like tissue homeostasis, brinolysis
phasize at the end the use of PMs as magic bullets in brain ther- and coagulation, adjustment of vascular tone, blood cell movement and
apeutics. activation, preservation of neuronal environment and vascularization of
normal neoplastic tissues [5].

Fig. 1. Schematic illustration of blood brain barrier.


Reprinted from [16] after permission of Elsevier.

44
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

3. Transport across the BBB

The last two decades have witnessed many researchers focusing on


the deep understanding of dierent transport pathways across the BBB
[18]. As illustrated in Fig. 2, there are dierent transport routs through
which molecules can cross the BBB.
Passive diusion of substances can be divided into paracellular and
transcellular. The paracellular pathway across the tiny spaces in be-
tween cells always provides a pathway for small water soluble mole-
cules. In contrast to other cell membranes, brain paracellular pathway Fig. 3. Schematic illustration of modern strategies for brain delivery.
is virtually absent due to presence of Tjs. The transcellular pathway is
available for molecules, which fulll certain criteria such as [1921]: i-
gp) [29], ATP binding cassette (ABC) transporter [30,31] and multidrug
molecular weight < 400500 Da; ii- unionized compounds; iii- log P
resistant protein (MRP) [32].These account for forcing out a vast ma-
value close to 2; iv- maximum number of cumulative hydrogen bonds in
jority of biologically active molecules.
molecule equal ten; and v- no anity to eux pumps. However, only
Finally, the cell-mediated transcytosis [33] is a recent dened route
few molecules meet these criteria and are capable to bypass the BBB
depending on monocytes and leukocytes to cross the BBB. Drug mole-
transcellulary by dissolving in lipid plasma membrane such as alcohol
cules or carriers can be uptaken by the inammatory cells and cross
and steroid hormones. A paradigm rarely encountered in drug mole-
through the BBB where inammatory cells are taking the function of
cules [21,22].
Trojan horses [34]. Cell-mediated transcytosis is distinctive for being
Carrier-mediated transport (CMT) involves some transporter proteins
practically suitable for both any type of molecules and particulate de-
on the BBB that bind to certain substrates followed by conformational
livery systems.
changes at the protein and transportation of the substance across the
So, the prohibited eects of the BBB on various important drugs
membrane. The most common transporter proteins are the glucose
delineated several modern strategies for eective drug transport to the
transporters (GLUT). There are ve members of the sodium-in-
brain.
dependent glucose transporters (GLUT1-5). Each GLUT transports glu-
cose, galactose, mannose, 2-deoxyglucose and 3-O-methylglucose to the
CNS [17]. The large neutral amino-acid transporter type 1 (LAT1), is
4. Modern strategies for brain delivery
also responsible for transportation of > 10 amino acids across the BBB
[18].
Multi-discipline approaches have lately emerged to elevate drug
Receptor-mediated transcytosis (RMT) [23] provides a mean for se-
concentrations within the brain. As shown in Fig. 3, these advances are
lective uptake of macromolecules via the reaction with their selective
based either on the route of delivery, modulation of drug chemistry or
receptors such as transferrin receptor (TfR) [24], insulin receptor [25],
BBB physiology or the use of nanocarriers. Herein we will discuss
lipoprotein receptor-related proteins (LRP1, 2) [26], and diphtheria
briey these access roads.
toxin receptor [27].
Adsorptive-mediated transcytosis (AMT) [28], involves an electro-
static interaction between a solute molecule carries a positive charge
4.1. Approaches based on the route of delivery
and the plasma membrane which carries a negative charge.
Eux pumps or transporters are the primary cause for drug expul-
To circumvent the BBB, scientists tried to overcome the barriers
sion from the brain. They are considered the main restriction for the
either by invasive routes through direct injection into the brain such as
retention of a large number of therapeutics in the brain. The most well
intracerebral, intraventricular and intrathecal injections or non-in-
characterized eux pumps or transporters are the P-glycoprotein (P-
vasively using the intranasal route [5].

Fig. 2. Transport routes across the blood brain barrier.

45
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

4.1.1. Invasive routes more sobetirome brain distribution in comparison to unmodied one.
Intracerebral (intraparenchymal) delivery necessitates direct drug
injection into the parenchymal space of the brain through cranium 4.2.2. Chimeric peptides
holes [35]. Therapeutic agents are administered as bolus or infusion via It was used since many decades ago to deliver peptides through the
intrathecal catheters [36,37]. Likewise, the intraventricular route im- BBB [54,55]. It depends on covalent binding of a non-transportable
plicates direct injection into the cerebral ventricle, providing rapid drug peptide to a peptide vector which is able to pass the BBB by RMT or
distribution in high concentrations, into ventricles and subarachnoidal AMT. The conjugated vector may be endogenous peptides [56],
area within the brain, therefore highly applicable in meningioma and monoclonal antibodies (mAbs) [57], or modied proteins [58], etc.
metastatic cells of cerebrospinal uid (CSF) [38]. In comparison to the Chimeric peptides dier from prodrugs in being biologically active so
intracerebral delivery, the absence of interconnection of this route with they don't need any extra biological steps to start their action. Pardridge
the interstitial uid is an asset [39]. However, the ependymal brain et al. coupled beta-endorphins (non- transportable peptides) to cationic
exterior is subjected to large drug amounts, which may provoke sub- albumin -which pass the BBB by AMT- via disulphide linkage [54,55].
ependymal astrogliatic reaction. They found that beta-endorphin-cationized albumin chimera was ra-
Though less invasive than intracerebral or intraventricular admin- pidly embraced by the brain; conversely, the BBB uptake of native beta-
istration, intrathecal route (direct into the cistern mana of the brain) endorphin was negligible. Recently, Condamine et al. [59] have studied
doesn't allow drug accumulation deeply in the brain parenchyma which the structural-activity relationship of some chimeric peptides obtained
is greatly imperative for drug sustainment. Spinal and disseminated from peptide E and beta-endorphin with and without various spacers.
meningeal diseases, but not glioblastoma, are conveniently treated by They deduced that the length and physicochemical properties of spacers
this route [40,41]. had a minimal eect on the analgesic potency.
However, all direct approaches suer from infection hazards, high
neurosurgical cost, need of experts and low eciency in diuse tumors, 4.2.3. Cationic peptides
metastasis, Alzheimer's disease, epilepsy, etc. This technique depends on increasing the positivity of the poly-
peptide chain to pass the BBB by AMT pathway [60]. The free carboxyl
4.1.2. Non-invasive route groups of the polypeptide (e.g., IGF-I, IGF-II, NGF) may be cationized
Recently, the intranasal route became successful for both systemic using polylysine (PLL), hexamethylenediamine, diazomethane or PLL
and brain delivery. The nose is divided into two nasal cavities via the with cleavable ester bonds [61]. Protamine, histone, avidin and catio-
septum, with a total surface area of 150 cm2 for nasal epithelium and a nized polyclonal bovine immunoglobulin are examples on cationic
total volume of 15 ml [42]. Nasal cavities are further subdivided into peptides with a profound ability to cross the BBB [61].
three distinct areas namely the nasal vestibule, respiratory and olfac-
tory regions. As shown in Fig. 4, systemic drug absorption occurs via 4.3. Approaches based on the physiology of the BBB
the respiratory region either transcellularly or paracellularly. CMT and
other transcytosis pathways are also available [43]. Direct brain ab- The noticed increased BBB permeability in pathological conditions
sorption takes place through the olfactory region by diverse mechan- such as ischemic stroke, cerebral edema and inammatory disorders
isms, embracing transcellular, paracellular, olfactory and trigeminal encouraged the scientists to similarly use the same gated enhancement
neural pathways [43]. Many recent projects targeted the olfactory area of drug permeation [62]. Meanwhile, Achareya et al. showed increased
intranasally for direct brain delivery [4450].The convenience of the BBB permeability after administration of inhaled anaesthetics (IAs), like
intranasal route resides in [43]: i- non-invasive nature, lacking risks of sevourane and isourane [63]. Subsequently, scientists succeeded to
infection or disease transmission, ii-facile self-administration, iii- high open the Tjs temporarily using dierent stimuli: chemical, physical or
surface area of absorption, iv- fast onset of action, v- a direct path to the biological. Such phenomenon is known as BBB leakage or CNS dis-
brain bypassing the BBB. On the other side, its limitations are: i- small ruption.
volume (25200 l) for administration, so limited for drugs of low dose,
ii- individual variability, iii- low pH of nasal cavity (5.56.5 in adults), 4.3.1. Chemical stimuli
iv- mucocilliary clearance, v- enzymatic degradation by nasal cyto- These are hyperosmolar solutions injected intra-arterially, causing
chrome P-450/peptidases/proteases (pseudo rst pass eect). shrinkage of endothelial cell accompanied by Tjs opening. Mannitol
hyperosmolar solution is the most commonly used. Foley et al. deliv-
4.2. Approaches based on drug chemistry ered adeno - associated virus (AAV) vectors to the brain using mannitol
[64]. The AAV vector encodes the human CLN2 gene, the gene of in-
These rely on performing chemical drug modication allowing its terest in Late Infantile Neuronal Ceroid Lipofuscinosis (LINCL). They
transfer across the BBB. Chemistry may increase drug hydrophobicity to
facilitate paracellular transport or attach a certain moiety to the drug
molecule to help in their brain uptake. Chemical changes embrace the
preparation of prodrugs, chimeric peptides and cationic peptides.

4.2.1. Prodrugs
Prodrug is a chemically modied form of the drug that has neither
biological toxicity nor activity. Although the concept of prodrugs for
brain delivery was introduced many decades ago [51], yet they are the
center of many researches till now. Peura et al. [52] used prodrug ap-
proach to increase cationic dopamine brain uptake by LAT1 receptors
by synthesizing three amino acid prodrugs of dopamine. They observed
that the phenylalanine promoiety of the prodrug had greater binding
ability for LAT1 and hence higher brain uptake in comparison to those
having an alkyl amino acid promoiety. Recently, Placzek et al. [53]
developed ester prodrugs of sobetirome, the selective thyromimetic that
trigger myelin repair to be used in demyelinating diseases such as
multiple sclerosis. They found that ethanolamine ester 11 resulted in Fig. 4. Schematic illustration showing the fate of drug after intranasal delivery.

46
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

reported that the mannitol volume was a determining factor that af- fresh mouse serum, RAW 264.7 murine macrophages were found to
fected the degree of BBB disruption. The combination of mannitol and engulf NPs with high negative or positive surface charges more ade-
stem cell was reported by Gonzales et al. to enhance the therapeutic quately [87]. Likewise, the same group found that there was a direct
eects in adult stroke and neonatal cerebral palsy [65]. Substances like proportionality between the magnitude of positive surface charge and
alkylglycerols [66,67], Lysophosphatidic acid [68], Cyclodextrins (CDs) the hemolytic activity and cytotoxicity against RAW 264.7cells;
[69], surfactants like sodium dodecyl sulfate (SDS) [70], and poly- whereas negatively charged NPs were devoided from any eect. In the
sorbate-80 (Tween-80) [71], endocannabinoids [72] revealed also BBB blood stream, cationic nanoparticles undergo opsonization, i.e. they
permeability modication. adsorb, on their surface, the protein opsonin from the blood stream
forming a corona [86]. Such opsonized NPs are cleared from the
4.3.2. Physical stimuli circulation by the RES very fast [86].
Ultrasound [73], microwave [74] and electromagnetic radiations Meanwhile, increasing surface hydrophobicity facilitates phago-
[75] are considered as physical stimuli for tight junctions opening in cytes uptake and hemolysis [88]. The PEGylation as a very common and
the BBB. popular example on stealth technology has been applied to all nano-
carriers to solve this problem [8991]. This technology provides the
4.3.3. Biological stimuli coating of nanoparticles with neutral, hydrophilic, highly exible PEG
They are exemplied by zonulaoccludens toxin (Zot) which was polymer chains imparting protection against plasma protein opsoniza-
found to reversibly promote - in a reversible concentration-dependent tion. Grafting PEG to a polymer (e.g., PEG-PLA and PEG-PLGA) or in-
manner - the Tjs opening [76]. Similarly, inammatory stimuli and cluding it as a main fragment of the copolymer (e.g., poloxamers) has
vasoactive compounds such as histamine [77], bradykinin [78] and its the same eects [92]. Coating the NPs with poloxamers, a PEG con-
analogues such as labradimil [79] were found to act as transient Tjs taining surfactants, was also adequate for extending the circulation
openers. time [93]. The diminished opsonization provided by PEGylation was
Although being eective in enhancing drug passage across the BBB, found to be due to steric hindrance as well as charge shielding [94,95].
opening of Tjs may reinforce the crossing of noxious pathogens. Besides Recently, it was reported that PEGylated NPs showed an enhanced
and more seriously, blood plasma protein leakage may occur producing extracellular bioavailability due to the escape from brain cell popula-
astrocytes toxicity, a condition known as astrogliosis [80]. Therefore, to tions (immune and non-immune) [96]. However, a PEGylated NPs
ensure a safe and ecient use of this approach in brain therapeutics, it prepared with long and dense PEG chains suered from a reduced ef-
deemed necessary to accurately estimate the time and extent of brain ciency due to the prohibited interaction with target cells [97,98]. The
exposure to such stimulations. removal of PEG coating after reaching the target site have been the
work of various authors [99,100]. Generally, minimal PEG coverage is
4.4. Approaches based on nanotechnology required to maximize circulation times. Although PEGylation is the
most common strategy applied for stealth technology, other strategies
CNS drug delivery has found appealing promises in nanotechnology. are available. For example, the use of various stealth lipids contained in
Due to their nano size range, they can overthrow the restraints of liposomes to suppress complement-dependent phagocytosis [101]. Al-
conventional drug delivery to access the brain [7]. An ideal nanocarrier though not fully understood, the authors suggested that the suppression
for CNS delivery should fulll the following criteria: of complement-induced phagocytosis induced by lipids occurs at a step
after liposome opsonization [101].
Particle size < 200 nm [81]. However, some researches preferred a
size < 100 nm [7]. 4.4.2. NPs for brain delivery
Long circulating, controlled-release particles with low im- Several classes of nanocarriers were applied for brain delivery
munogenicity, good biocompatibility and bioavailability, averting during the last decades such as liposomes [102,103], solid lipid nano-
eux transport. particles [104,105], polymeric nanoparticles [106,107], polymeric
Protects the therapeutic cargo from enzymatic degradation. micelles [108,109], cubosomes [110], nanospheres [111,112], den-
Preferably, attached to a BBB-targeting moiety. drimers [113,114], cyclodextrins [115,116], drug polymer conjugates
[117], gold nanoparticles [118,119], silver nanoparticles [120], carbon
4.4.1. Optimization of nanoparticles blood circulation time nanotubes [121,122] and quantum dots [123,124]. Many review arti-
The proximity of the nanoparticles (NPs) to their target organ for cles discussed the dierent classes of nanocarriers for brain delivery
long time will ensure maximum eciency. After oral or intravenous before [8,125132].
administration, the NPs circulating in the blood suer from two main Despite the fact that NPs can pass BBB owing to their small particle
challenges viz. renal clearance and phagocytosis. size, many researches pinpoint on specic delivery of NPs to CNS de-
NPs with size (< 25 nm) are immediately cleared from blood by pending on their conjugation to a particular ligand specic to cross the
kidneys [82].On the other hand, increasing size will result in re- BBB via active targeting, e.g. of ligands specic to certain pathways
ticuloendothelial system (RES) clearance due to protein adsorption. include:
Actually, Fang et al. found that the 80 nm particles had slower blood
clearance compared t0 the larger nanoparticles (171 and 243 nm) due Ligands specic for CMT: The most commonly used ligands for this
to fewer protein adsorptions [83]. pathway are the ligands that target glucose transporters such as
NPs with particle size (< 100 nm) can pass BBB without active glucose and dehydroascorbic acid (DHA) [133,134] that target the
targeting. Sonavane et al. showed that gold nanoparticles with particle GLUT1 and aminophenyl--d-mannopyranoside (MAN) [135,136]
size 15 and 50 nm could pass BBB, but those with size of 200 nm which targets GLUT1 and GLUT3.
couldn't [84]. On a similar basis, Shilo et al. demonstrated a great re- Ligands specic for RMT: this pathway is very common and is the
liance of the intracellular uptake of gold NPs on size [85]. They com- most targeted pathway. Many receptors are present on the surface of
pared four sizes viz. 20, 50, 70 and 110 nm, and found that the size of BBB. Here are examples on the receptors found on the BBB with
70 nm was the optimum. Consequently, the range of 25100 nm is their targeting ligands:
optimum for crossing BBB, avoiding renal clearance and phagocytosis. 1. Insulin receptors: Amyloid -peptide [137], HIRMAb [138]
Zeta potential (), an expression of the net total charge on the 2. Transferrin receptor (TfR): Transferin (Tf) [139,140] or its anti-
particle's outside surface, is an important physical factor controlling its bodies (e.g. 8D3 [141] OX26 [142] and R17 [143])
pharmacokinetics and biodistribution [8,86]. After opsonization in 3. Lactoferrin receptors (LfR): Lactoferrin (Lf) [144]

47
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

4. LRP1 and LRP2 receptors: Polysorbate 80 (PS80) [145,146], accompanied by their association together with the formation of core -
lactoferrin (Lf) [147149], apolipoproteins (ApoA, E or B) shell micellar structure [169]. PMs were rst introduced as drug de-
[150152] and angiopeps family (specially angiopep 2) livery vehicles by Helmut Ringsdorf in the early 1980s [170]. However,
[153,154]. it was used for the rst time for brain delivery in 2008 by Liu et al. [11].
5. Diphtheria toxin receptor (DTR): Cross reacting material (CRM Table 1 summarizes the studies using PMs for brain delivery since that
197) which is the benign mutant of diphtheria toxin (DT) [155]. time.
6. Acetyl choline (Ach) receptors: Rabies virus glycoprotein (RVG)
[108]. 4.4.3.1. Advantages of PMs as nanocarriers for brain delivery. PMs are
Ligands specic for AMT: Cell penetrating peptides (CPPs) [156] more advantageous than other nanocarriers for brain delivery.
which are small peptides of < 30 amino acids. They dier from each Generally, it has been shown that PMs enjoy the following advantages:
other in the peptide chain length, sequence of amino acids and the
net charge on the peptide chain. Their positive charge makes them 1) Small size (10100 nm), which is in the ideal range to pass BBB.
attracted to the negative charge on the cell membrane followed by 2) Ease of preparation and handling (just by self-assembly).
transcytosis. Examples of CPPs applied for brain delivery include 3) Ease of incorporation of hydrophobic drugs.
lectin [157,158], odorranalectin [110,159], penetratin [160,161] 4) High encapsulation eciency (EE %) of hydrophobic drugs.
and trans-activating transcriptional activator (Tat) [119,162]. 5) Ease of sterilization by ltration due to small size [171].
Ligands specic for inhibition of eux pumps: An ecient strategy 6) The hydrophilic shell prevents non-specic interactions with plasma
for drugs having anity to any of the eux pumps. Poloxamers proteins or uptake by RES (inherited stealth property).
were commonly found to block P-gp by interacting with the cell 7) Possibility of surface modication by various ligands for ecient
membrane, and uidizating it; inhibiting of P-gp ATPase activity; brain targeting.
and attenuating cellular ATP [163,164].
Ligands specic for cell-mediated transcytosis: Cyclic Arginine- 4.4.3.2. Classication of PMs. PMs can be classied on a dierent basis
Glycine-Aspartic acid (cRGD) was used by Jain et al. to target in- as follows:
tegrins expressed on monocytes and neutrophils [165]. The in-
ammatory cells carried liposomes loaded with diclofenac to the A) Classication based on the type of copolymer used
brain eciently. Qin et al. formulated negatively charged nanosized
liposome loaded with trefoil factor 3 (TFF3) [166] for treatment of PMs can be classied as polymer-polymer micelles or polymer-lipid
depression. In-vitro studies proved that their negative charge en- micelles. In polymer-polymer micelles, both the core and shell are
abled their success transfer inside the bound monocytes across the formed of polymers. These polymers may be of c-c chains (Table 1) as
BBB. This approach doesn't require nanosized carriers and impedes PLA, PLGA, PEG, Poloxamer etc., or it may be of polypeptide backbone
one of the routes by which pathogens intrude the brain. as poly-glutamic acid [172], poly-L-phenylalanine (PPhe) [133] and
poly-asparthydrazide [108]. In polymer-lipid micelles, the corona is
A special class of NPs which arised in the last decade as a really formed of hydrophilic polymer mostly PEG while the core is formed of
promising nanocarrier system for brain delivery is polymeric micelles lipid. The commonly used lipids were synthetic phospholipids such as
(PMs) [1113]. In the upcoming section, we will discuss PMs as na- dioleolyphophatidyl ethanolamine (DOPE) and distearoylphophatidyl
nocarriers, its history with brain delivery, its advantages over other ethanolamine (DSPE) [173]. Cholesterol [11] and oleic acid [174] were
nanoparticles and its applications in brain delivery. The modications also used (Table 1).
performed on micelles to be in line with modern strategies for ecient
brain delivery will be focused. B) Classication based on the type of interaction between drug and
polymer
4.4.3. Micelles as a type of nanocarries: history
PMs are nanoscopic carriers of amphiphilic copolymers formed by PMs can be classied on this basis to conventional micelles; drug
their self-assembly in an aqueous medium [167] presenting their hy- conjugated micelles and polyion complex (PIC) micelles. In conven-
drophilic shell or corona to the medium, while their core is composed tional micelles, no interaction exists between drug and copolymer.
from their hydrophobic portion (Fig. 5). The aggregation occurs in Hydrophobic drugs are easily included in micellar cores by direct
aqueous medium when the amphiphilic block copolymer concentration mixing. A technique only suitable for PMs with soft cores (i.e. with a Tg
surpasses what's called critical aggregation concentration (CAC) or lower than the temperature used for drug loading) such as pluronic
critical micelle concentration (CMC) [168]. For entropy considerations, micelles. On the other hand, PMs with solid cores (crystalline or with a
the hydrophobic segments of the amphiphilic copolymer should ar- Tg higher than the temperature used for drug loading) need more
range themselves away from water molecules. This will be complex techniques for loading drugs; examples include poly(ethylene

Fig. 5. Schematic illustration of polymeric micelles struc-


ture and formation.

48
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

Table 1
Summary of studies discussing micelles for brain delivery.

Drug Disease Copolymer Method of preparation Ref.

ATRA GBM PEG-chitosan Simple addition [187]


Ciprooxacin Encephalitis PEG-b-Col Dialysis [11]
PHEA-EDA-PLA Dialysis [175]
Zolmitriptan Migrain Pluronic F127 + PEG-400 and vitamin E-TPGS Simple addition [188]
Coumarin PCL-PEEP Dialysis method [189]
GBM PEG-b-PLA Co-solvent evaporation [190]
PTX GBM PEG-b-PLA Thin lm hydration [191]
Flurbiprofen Alzheimer PHEA-EDA-PLA Dialysis method [192]
PTX GBM DSPE-PEG Thin lm hydration [173]
siRNA Cancer mPEG-b-PCL-b-PPEEA Co-solvent evaporation [184]
DIR PEG-b-PLA Thin lm hydration [193]
Coumarin PEG-b-PCL W/O emulsion [194]
HSA PEG-PAE-API Simple addition [183]
Doxurobicin Cancer CS-SA Dialysis [195]
AmB Meningoencephalitis PE-PEG Thin lm hydration [196,197]
PTX GBM PCL-PEEP + PEG-PCL Dialysis [12,171]
Plasmid DNA mPEG-PDMAEMA Direct addition [198]
DACHPt GBM mPEG-b-P(Glu) Dialysis [172]
17-AAG GBM Pluronic P123/F127 Thin lm hydration [164]
Efavirenz HIV Pluronic F127 + poloxamine T904 Simple addition [185]
Flurbiprofen Alzheimer PEG-b-PLA Co-solvent evaporation [199]
BCNU and VEGF-siRNA GBM R3V6 Direct addition [200]
miR-21 inhibitor and doxorubicin GBM PLA-b-PDMAEMA Co-solvent evaporation [201]
siRNA and captothecin GBM PEG-b-PCL Thin lm hydration [202]
Captothecin GBM PEG-b-PCL W/O emulsion [203]
Curcumin Cancer mPEG-OA Co-solvent evaporation [174]
YC1 Neuroprotection SACDs Co-solvent evaporation [177]
GBM DSPE-PEG Thin lm hydration [204]
Doxorubicin GBM Pluronic105 Dialysis [205]
WIN55, 212-2 Neuropathic pain SMA Co-solvent evaporation [178]
Clonazepam Epilepsy BS: sodium glycocholatehydrate + soya PC Thin lm hydration [206]
LTG Epilepsy Pluronic P123 Thin lm hydration [163]
Pluronic P123/F127 [207]
Docetaxel Cancer PEG-b-PLA Thin lm hydration [109]
ITZ Intracranial fungal infections PEG-pLys-pPhe Dialysis [133]
siRNA Neurodegenerative diseases mPEG-g-PAHy-GTA Simple addition [108]
SPIONs GBM PEG-b-PCL Co-solvent evaporation [208]
Curcumin and HSVtk GBM R7L10 O/W emulsion [209]
Nimodipine SAH Pluronic F127 and P123 + PC Thin lm hydration [186,210]
sPMI GBM PEG-b-PLA Thin Film hydration [211]
Curcumin and rapamycin GBM PEG-b-PCL Dialysis [212]
Docetaxel Cancer TPGS Thin lm hydration [13]
DOM Psychosis pHPMA- co -pLMA Co-solvent evaporation [213]
TMZ GBM R7L10 O/W emulsion [214]
AmB Intracranial fungal infection PEG-b-PLA Co-solvent evaporation [215]
Myricetin GBM Pluronic P123/F68 + Chitosan Thin lm hydration [216]
Quercetin GBM Soy lecithin/Pluronic F68/Labrasol Thin lm hydration [217]
Doxorubicin GBM PEG-b-PCL Dialysis [218]
Doxorubicin and paclitaxel GBM Pluronics Thin lm hydration [219]
Carmustine GBM PEG-PLGA Thin lm hydration [220]
Gold and SPIONs GBM PEG-b-PCL O/W emulsion [221]
Curcumin and doxorubicin GBM PE-PEG Thin lm hydration [222]
Curcumin GBM PEG-b-PLA Thin lm hydration [223]
Clonazepam Status epilepticus Pluronic P123/L121 Thin lm hydration [224]
Myricetin GBM Pluronic P123/F68 Thin lm hydration [225]
Vinpocetine Cerebrovascular and cerebral degenerative diseases Pluronic P123 + PEG-PCL Thin lm hydration [226]

17-AAG: 17-Allylamino-17-demethoxy geldanamycin; AmB: amphotericin B; APN: aminopeptidase N; ATRA: all-trans retinoic acid; BCNU: bis-chloroethylnitrosourea; BS: bile salt; CS-SA:
stearic acid-grafted chitosan; DACHPt: (1,2-diaminocyclohexane) platinum(II); DiR: near-infrared uorescent dye; DOM: domperidone; DSPE-PEG: 1,2-distearoyl-sn-glycero-3-phos-
phoethanolamine-N-methoxy(polyethylene glycol); DTX: docetaxel; FA: folic acid; GBM: glioblastoma multiforme; HIV: human immunodeciency virus; HSA: human serum albumin;
HSVtk: herpes simplex virus thymidine kinase gene; ITZ: itraconazole; LTG: lamotrigine; mPEG-b-P(Glu): poly(ethylene glycol)-b-poly(L-glutamic acid); mPEG-b-PCL-b-PPEEA: mono-
methoxy poly(ethylene glycol)-block-poly(-caprolactone)-block-poly(2-aminoethyl ethylene phosphate); mPEG-OA: monomethoxy poly(ethylene glycol)-oleate; mPEG-PAHy-GTA:
methoxy poly(ethylene glycol)-,-polyasparthydrazide-glycidyltrimethylammonium; m-PEG-PDMAEMA: methoxy poly(ethylene glycol)-poly(2-(dimethylamino)ethyl methacrylate;
PC: phosphatidylcholine; PCL-PEEP: poly(-caprolactone)-block-poly(ethyl ethylene phosphate); PEG: poly(ethylene glycol); PEG-b-Chol: poly(ethylene glycol)-block-cholesterol; PEG-b-
PCL: poly(ethylene glycol)-block-poly(-caprolactone); PEG-b-PLA: poly(ethylene glycol)-block-poly(lactic acid); PEG-PAE-API: poly(ethylene glycol)-poly(b-amino ester)-1-(3-amino-
propyl) imidazole; PEG-pLys-pPhe: poly(ethylene glycol)-b-poly(L-lysine)-b-poly(L-phenylalanine); PE-mPEG: 1,2 distearoyl-snglycero-3-phosphoethanolamine-N-(methoxy(polyethylene
glycol); PHEA-EDA-PLA: ,-poly((N-hydroxyethyl)-DL-aspartamide)-ethylenediamine-poly(lactic acid); pHPMA-co-pLMA: poly(N-(2-hydroxypropyl)-methacrylamide)-poly(lauryl me-
thacrylate); PLA-b-PDMAEMA: poly(lactic acid)-poly(dimethylaminoethyl methacrylate); PTX: paclitaxel; R3V6: 3-arginine and 6-valine; R7L10: 7-arginine and 10-leucine; SACDs:
sodium alginate cholesterol derivatives; SAH: subarachnoid hemorrhage; siRNA: small interfering RNA; SMA: poly(styrene-co-maleic anhydride); SPIONs: superparamagnetic iron oxide
nanoparticles; sPMI; stapled peptide antagonist of both MDM2 and MDMX; TMZ: temozolomide; TPGS: D-alphatocopheryl polyethylene glycol succinate; VEGF-siRNA: vascular en-
dothelial growth factor-small interfering RNA; YC1: 5-cholestane-24-methylene-3,5,6,19-tetraol

49
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

glycol)-poly(lactic acid) (PEG-PLA), poly(ethylene glycol)-poly(-ca- C) Classication based on the number of copolymers incorporated in
prolactone) (PEG-b-PCL) and poly(ethylene glycol)-poly(b-benzyl-L-as- PMs fabrication
partate) (PEG-PBLA) micelles. Incorporating drugs into conventional
micelles can be done by ve dierent methods: On this basis, PMs can be classied into purebred micelles and
mixed or hybrid micelles (Table 1). Purebred micelles are composed of
(1) Dialysis method: it is an extensively used method [11,175] done by only one type of amphiphilic copolymers. On the other hand, for mixed
preparing a drug solution in a water miscible organic solvent in or hybrid micelles, there is a combination of more than one type of
which the copolymer is soluble too, followed by dialysis against copolymers [171,185]. Mixing of more than one copolymer during
water. During dialysis, water as a non-solvent will replace the or- micelle preparation aims always at improving their physicochemical
ganic solvent provoking the hydrophobic blocks to self- assemble properties. Mixing pluronic F127 with pluronic P123 produced PMs
into the micelle cores followed by drug encapsulation. Micelles with enhanced kinetic stability due to the long polyethylene oxide
diusion out is prohibited by the semi-permeable membrane while (PEO) chains of Pluronic F127, counteracting the stacking of cylindrical
unloaded free drug will be removed. aggregates of hydrophobic polypropylene oxide (PPO) chains of
(2) Emulsion method (O/W or W/O): PMs are formed due to the eva- Pluronic P123 [164]. Mixing these micelles with phosphatidylcholine
poration of a volatile water immiscible organic solvent, in which (PC) lowered CMC and multiplied blood micellar stability as noticed by
the drug is dissolved, after the formation of an emulsion due to the Baselious et al. [186].
vigorous mixing with the aqueous solution [176]. The copolymer
may be present in the organic or aqueous phase according to its 4.4.3.3. Eect of physicochemical properties of PMs on its behavior in brain
solubility. delivery.
(3) Solvent evaporation method (i.e., thin lm evaporation or solvent
casting method): a drug-impregnated polymer lm is formed due to A) Critical micellar concentration
heat and vacuum aided evaporation of a volatile organic solvent
containing both components [163,164]. The drug loaded PMs are As we mentioned before PMs are formed when the copolymer
formed thereafter due to lm hydration accompanied with vigorous concentration reaches a certain value called CAC or CMC. The CMC
shaking. aects greatly the thermodynamic stability of PMs due to the extreme
(4) Co-solvent evaporation method: similar to dialysis method both dilution eect of the circulation after IV injection. If micellar con-
drug and polymer are dissolved in a water miscible organic solvent centration drops below the CMC, the encapsulated drug will be early
[177,178]. Water is added to this solution with vigorous shaking released into the circulation before getting to its target site with the
causing organic phase evaporation, forming the drug-loaded mi- possibility of drug precipitation inside the blood vessels [167]. Gen-
celles. erally, the blood stability improves as the CMC value decreases.
(5) Freeze-drying method: a freeze-dryable organic solvent such as tert- For CMC determination denite concentrations of pyrene (hydro-
butanol is used in this method to dissolve polymer and drug [179]. phobic uorescent probe) are mixed with serial copolymer dilutions by
After water addition, the mixture is freeze-dried then reconstituted ultrasonication to ensure complete pyrene lodging into the hydrophobic
with aqueous media. micro-domains [177]. The uorescence spectra are recorded on a
uorometer at room temperature. The intensity ratios of its rst peak in
A drug covalently bonded to one segment of the copolymer to build water (I1) to its third peak in the hydrophobic medium (I3), obtained
the hydrophobic core of a PM forms what's known as conjugated mi- from the pyrene emission spectra, are plotted versus the logarithmic
celles [180]. To achieve a high drug load a linear dendritic drug- function of polymer concentrations [192]. The intersection of the hor-
polymer conjugates modied with choline derivate (CD) was synthe- izontal tangent with that of the curve at the inection is taken as the
sized then self-assembled into PM [181]. Eight molecules of doxor- CMC value.
ubicin (DOX) were attached to PEG, which was linked to CD. The CD-
PEG-DOX8 micelles showed higher cellular uptake and antitumor ac- Table 2
tivity against glioma in comparison to the non-modied conjugate. CMC values for some of copolymers used for brain delivery.
Likewise, a water soluble PEG-PLA-PTX micelles were prepared by
Copolymer CMC Ref. Copolymer CMC (mg/ Ref.
Wang et al. after conjugation of paclitaxel (PTX) with PEG-b-PLA [182]. (mg/L) L)
They were tested in C6 glioma model in Wistar rats. After tail injection,
the micelles could cross the BBB and showed an enhanced therapeutic mPEG-OA 30 [174] PHEA-EDA- 25 [192]
PLA
eect when compared to Taxol.
SACDs [177] PEG2000- 107 [167]
PIC or micelleplex depends on electrostatic interaction between the PLA700
drug and the copolymer with no covalent bond formation. A very (Ch pretreated with 0.8 PEG2000- 22
popular example on PIC was reported between negatively charged triphosgene) PLA1000
nucleic acids and proteins and positively charged amphiphilic copoly- (Ch not pretreated 2 PEG2000- 12
with triphosgene) PLA1300
mers [183,184]. Huo et al. prepared a novel PIC micelles from PEG-
CS-SA 65 [195] PCL-PEEP 0.51 [189]
polyasparthydrazide-glycidyltrimethylammonium (mPEG-g-PAHy- PEG-b-Col 6.2 [11] DSPE-PEG700 28 [227]
GTA) copolymer to deliver siRNA to the brain [108]. PAHy was the Tat-PEG-b-Col 7.8 DSPE- 30.85
core, while PEG corona shielded the surface charges of the micelles for PEG2000 17.39
stability and prolonged circulation. The negative charged siRNA inter- DSPE-
PEG5000
acted with the positive charged GTA connected to PAHy. The PIC mi- Pluronic F127 35.228 [228]
celles decorated with RVG (a 29 amino acid peptide with the sequence Pluronic P123 25.3
(YTIWMPENPRPGTPCDIFTNSRGKRASNGC) - as a specic binder for Pluronic P105 40.3
Ach receptors expressed by neuronal cells - were easily internalized in
CS-SA: stearic acid-grafted chitosan; DSPE-PEG: 1,2-distearoyl-sn-glycero-3-phos-
the neuro2a cells. In neuro2a cells, Huo et al. proved that cellular up-
phoethanolamine-N-methoxy(polyethylene glycol); mPEG-OA: monomethoxy poly(ethy-
take of the RVG-decorated micelles was 1.52.6 times that of the un-
lene glycol)-oleate; PCL-PEEP: poly(-caprolactone)-block-poly(ethyl ethylene phos-
modied micelles. By this study, Huo et al. introduced an interesting phate); PEG-b-Chol: poly(ethylene glycol)-block-cholesterol; PEG-b-PLA: poly(ethylene
siRNA carrier to the brain delivery. glycol)-block-poly(lactic acid); PHEA-EDA-PLA: ,-poly((N-hydroxyethyl)-DL-asparta-
mide)-ethylenediamine-poly(lactic acid); SACDs: sodium alginate cholesterol derivatives;

50
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

Table 2 shows that CMC is greatly aected by hydrophilic and hy- formulation and process related factors. The formulation-related de-
drophobic chain length as well as the ratio between them, copolymer terminants include the type of copolymer, percentage of grafting of
type and ligand conjugation [167]. Generally, CMC value reduces as the hydrophilic and hydrophobic segments and the drug/polymer feeding
hydrophobic block length increases [167]. ratio. While those associated to the process comprise method of pre-
paration and conditions of the preparation including solvent nature, its
B) Size, shape and zeta potential ratio to aqueous phase and so on.
Huo et al. showed that increasing PEG grafting degree on COOH-
Crossing the BBB by NPs has been known to be particle size de- PEG-g-PAHy-GTA copolymer decreased the encapsulation of siRNA
pendent. Dynamic light scattering (DLS), atomic force microscopy [108]. Flexible PEG chains could shield the positive charge on the
(AFM) and transmission electron microscope (TEM) are used to de- polymer which decreased its interaction with siRNA. Niu et al. used
termine and assist PMs particle size [189,205]. Those in the range of dierent drug DOX to polymer ratios viz. 2/200, 4/200, 6/200, 8/200
10100 nm are optimally uptaken by the brain. Micellar particle size and 10/200 [205]. The increase in DOX content, up to a certain limit,
depends on many factors viz.; copolymer, hydrophilic/hydrophobic raised the EE% from 18.0% to 45.3%. The initial increase in drug
segment's ratio, drug loading, drug/polymer ratio, ligand conjugation concentrations enhanced drug diusion from the organic to the aqu-
and density. Generally, particle size increases after drug loading due to eous phase with an increased tendency to reside in the core. However,
expansion of hydrophobic core volume accommodating the drug mo- drug amounts surpassing a certain limit precipitate before micelle for-
lecules [205]. Leyva-Gomez et al. showed that particle size increased mation, which lowered the EE% signicantly. When studying the level
from 12.4 1.8 to 16.5 1.4 nm after drug loading [206]. Con- of the organic phase (DMSO), they found that both EE% and DL% de-
versely, Mao et al. observed a decrease in particle size of micelles from creased with higher DMSO concentrations due to micelles disruption
60.3 0.3 to 47.7 0.4 nm after drug loading ascribable to the very induced by DMSO. Besides, DMSO enhanced DOX solubilization in the
high interaction between the siRNA and copolymers [184]. In a further aqueous phase hindering its diusion into the micellar core. The au-
study, Zhang et al. showed that particle size increased from thors recommended a ne-tuning of the organic solvent amounts during
33.31 0.61 to 87.85 2.32 nm after Tf conjugation [171]. How- PM preparation.
ever, Niu et al. showed no PMs size increase after conjugation with
glucose or folic acid (FA) because of their small size [205]. Moreover, D) Cytotoxicity and hemolysis assays
Mu et al. observed a particle size increase from 22.2 1.5 to
111.3 17.2 nm after raising the feeding ratio from 5% to 20% [199]. Cytotoxicity test is performed on in-vitro BBB model to examine the
The shape of PMs can be examined using TEM [108,177,198,205] toxicity of the formulation on brain cells [12,189,191,198]. Using MTT
and AFM [13,133,189]. Almost in all studies for brain targeting, mi- assay, Shao et al. tested the cytotoxicity of AmB-incorporated PE-PEG
celles were found to be spherical in shape with no aggregates micelles against brain capillary endothelial cells (BCECs) [197]. The
[133,198]. percentage of viable cells was determined using the following equation:
Zeta potential is also an important parameter for longer blood cir-
Viability% = (ABst ABsc) 100
culation. It is measured depending on DLS using zetasizer. It is aected
by the type of copolymer, drug loading, ligand conjugation and ligand where Abst and Absc are the absorbances of drug-treated and control
density. Chung et al. showed that zeta potential increased from wells, respectively.
27.4 4.5 to 12.6 1.6 mV upon conjugation of CREKA pep- Hemolysis test is performed to examine the damaging eect of mi-
tide to the surface of PEG-PE micelles [204]. Miura et al. showed non- cellar solution on RBCs. It is very critical if the copolymer or ligand is
signicant zeta potential variation upon increasing in cRGD conjuga- used for the rst time. Zhang et al. performed this test on dierent
tion ratio from 5% to 20% on the surface of poly(ethyleneglycol)-b-poly concentrations of blank PMs and their Tf modied analogs on isolated
(L-glutamic acid) (PEG-b-P(Glu)) micelles [172]. erythrocytes from SD rat blood using a positive control (Triton X-100)
[171]. After centrifugation, the supernatant was analyzed for he-
C) Encapsulation eciency and drug loading moglobin content and the percentage of hemolysis was calculated as
follows:
Micelles are generally separated from unentrapped drug by ltra-
Hemolysis% = (A s A 0) (A100 A 0) 100
tion using 0.22 m nylon lter [164,173,193,205]. To calculate en-
capsulation eciency (EE%) and drug loading (DL%), three methods where As, A0and A100 are the respective absorbances of the sample,
are reported: the rst involves the mixing of an aliquot of the ltrate unlysed sample treated with saline and fully lysed sample treated with
with organic solvent to dissolve the micelles and calculate the released deionized water. All copolymers and ligands listed in (Tables 1 and 4)
drug [11,211]. However, this method is not accurate in calculating DL are safe for brain delivery.
as it doesn't consider the micelles yield. In the second method which is
considered to be the most accurate, the ltrate is subjected to freeze E) Stability of PMs
drying followed by organic solvent addition to dissolve the micelles
powder [171,189,199]. Finally, a third method stated only in one study Both shelf and serum stability should be tested for PMs. Shelf sta-
by Sonali et al. in which they removed the water in the ltrate via bility is performed by leaving PMs at 4 C followed by examination of
evaporation by rotary evaporator under high temperature and reduced the particle size, zeta potential and EE% at specied time intervals.
pressure to form a lm [13], which was then dissolved in an organic Zheng et al. showed that negligible changes in particle size and EE%
solvent. EE % and DL % are then calculated using to the following occurred in PMs at 4 C after 15 days, 30 days and 60 days [177].
equations: However, PMs can't retain its stability > 60 days, and that's why lyo-
philization is very common just after the preparation [164,174]. Serum
Weight of drug in micelles
EE% = 100 stability test is performed in conditions mimicking the physiological
Weight of the feeding drug
conditions (i.e. PBS 7.4, 37 C, in presence [108] or absence of serum
Weight of drug in micelles [192] with moderate stirring). Sarpietro et al. performed the test
DL% = 100 through a 48 h period in PBS pH 7.4 [192] and found non-signicant
Weight of the micelles
dierence in the particle size or PDI. Additionally, Huo et al. performed
Almost in all studies, PMs showed high EE% for hydrophobic drugs. the test in presence of 50% fetal bovine serum for 24 h [108] and
Factors aecting EE% and DL% of certain drugs include both conrmed the sequence integrity of siRNA.

51
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

F) In-vitro drug release As we mentioned before, PMs consist of hydrophilic shell and hy-
drophobic core. They encapsulate hydrophobic drugs such as paclitaxel,
Studies are generally done using dialysis method where 12 ml of docetaxel, vincristine and many other hydrophobic drugs eciently in
micellar solution is introduced in pre-swollen dialysis bag and im- their cores (Table 1). They are not suitable for encapsulation and de-
mersed in a buer from which samples are withdrawn at predetermined livery of hydrophilic drugs. However, Huang et al. prepared core-in-
time intervals [13,177]. Generally, a sustained drug release was usually versible micelles (CIMs) for delivery of hydrophilic drugs [231]. They
obtained preceded by an initial burst phase. The release proles depend developed a novel biocompatible linear-dendritic amphiphilic co-
mainly on the type of copolymer used. PEG-PLA copolymer achieved polymer viz. PEG5kCA8 which is composed of polyethylene glycol (PEG
sustained release of PTX up to 72 h [191]. In another study, 65% of 5000) and a dendritic octamer of cholic acid (CA8). This amphiphilic
lamotrigine was released from Pluronic P123 during the rst 4 h copolymer was found to self-assemble into micelles in aqueous solution
[163].Similarly, Sonali et al. and Zhan et al. reported the same beha- for ecient delivery of hydrophobic drugs and into CIMs selectively in
viors with docetaxel from Tf receptor-targeted vitamin E TPGS micelles certain apolar solvents with a PEG shell and a hydrophilic core for ef-
and PTX from PEG-PLA micelles, respectively [13,191]. cient delivery of hydrophilic drugs. Despite the trials of applying PMs
In addition, drug type, pH and ligand conjugation have also high for hydrophilic drugs delivery, so far PMs are used mainly for ecient
inuence on drug release. Dierent authors showed that the release of hydrophobic drugs delivery.
DOX was commonly fastened by the decrease in pH [205,229,230]. An
observation which was likely due to its 30-NH2 group protonation and B) Shelf stability
azomethine bond breakdown in its dimer, which increases its solubility
in water [205]. In addition, Sonali et al. showed that decoration of PMs PMs like all nanoparticles have poor shelf stability. They tend to
surface with Tf slowed the drug release kinetics, in comparison to non- aggregate with time leading to an increase in the particle size [232].
targeted micelles [13]. They also showed that the increase in the ligand Undoubtedly, this particle growth will aect the physicochemical,
concentration extended the release sustainment. The t50% (time at pharmacodynamic and pharmacokinetic properties of the PMs. Lyo-
which 50% of the drug was released) of the formulations were 12, 16, philization is a popular and common solution for this problem. Many
24 and 48 h for the unconjugated micelles, DTX-TPGS-Tf1, DTX-TPGS- studies subjected PMs to lyophilization just after the preparation to
Tf2 and DTXTPGS-Tf3, respectively where DTX-TPGS-Tf1(contains keep it in the dry solid form with high stability [164,186]. Lyophili-
50 mg of TPGS-Tf and 100 mg of TPGS), DTXTPGS- Tf2 (contains 75 mg zation can be performed in absence or presence of cryoprotectants such
of TPGS-Tf and 75 mg of TPGS) and DTX-TPGS-Tf3 (contains 100 mg of as glucose, sucrose, maltose, mannitol and trihalose. Cesur et al. could
TPGS-Tf and 75 mg of TPGS). On the opposite side, Zhang et al. showed successfully prepare freeze-dried DSPE-PEG2000 micelles without the
that Tf conjugation didn't aect release behavior [12]. Likewise, Mu addition of any cryoprotectants [233]. They found that particle size and
et al. showed that aptamer conjugation didn't destroy the integrity and encapsulation eciency didn't change signicantly before and after the
compactness of micelles and didn't aect the release [199]. lyophilization process. However, Moretton et al. showed that the ad-
dition of a cryoprotectant to PCL-PEG-PCL ower-like PMs was a must
G) Cellular uptake and in-vivo study to prevent the particle size growth during lyophilization [234].

Cellular uptake is performed on in-vitro BBB models to study the C) Serum stability
ability of crossing the barrier in-vivo [8]. McCarthy et al. described two
types of models; primary cell models utilizing brain microvascular en- The main challenge of PMs is the possibility of premature release of
dothelial cells (BMECs or BMVECs) or also called BCECs. The second drugs in the circulation due to the high dilution [167]. The premature
suggested model was immortalized cell lines isolated from mice, rats or drug release results in inability to deliver a sucient amount of the
humans. After proper incubation of cells, micellar solutions application drug to the targeted site, increased systemic side eects and the pos-
and drug quantitation is performed. Table 3 shows a comparison be- sibility of precipitation of the drug in the blood vessels. Many ap-
tween the two types and examples used with micelles for brain delivery. proaches were performed to increase the serum stability of PMs which
As shown, most studies used primary cell lines due to their similarity are discussed in details in 4.4.3.6 Section.
with in-vivo conditions. In-vivo study and pharmacokinetic analysis Despite all the aforementioned limitations, PMs were able to reach
should follow cellular studies [173,205]. the clinical studies for treatment of dierent types of cancer [235].

4.4.3.4. Limitations of PMs. 4.4.3.5. Applications of PMs in brain delivery.

A) Delivery of hydrophilic drugs A) Drug delivery

Table 3
Comparison of dierent in-vitro BBB models used with micelles for brain delivery.

Primary cell models Immortalized cell lines

Denition They are isolated from blood vessels of dierent species of animals such as rats, mice, calves, pigs They have been immortalized through various techniques
and human
Advantages 1- High TEER values 1- Easily subcultured
2- With various receptors and enzymes resembling in-vivo conditions 2- Used for number of passages without changing their
properties.
Disadvantages 1- Their procurement is a challenge 1- Lower TEER values
2- Short time use 2- Higher paracellular permeability in comparison to
3- Exhibit batch to batch variation primary cell models
Examples BMECs [12,133,171,173,189,196198,205] b.END3 [195,199],
Neuro2a [108,175],
hCMEC/D3 [206]

BMECs: brain microvascular endothelial cells

52
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

As shown in Table 1, many studies utilized PMs as promising suc- core cl-micelles as follows: 1- radical polymerization: used for poly-
cessful nanocarriers for the transportation of dierent therapeutic merizable groups containing PMs; 2- the addition of a bifunctional
agents in dierent diseases across the BBB. Obviously, PMs have crosslinker: used for reactive groups containing PMs; and 3- disulphide
especially many applications in brain cancer treatment adopting the bridges: used for thiol groups containing PMs [246]. In their study,
dierent techniques of controlled dissociation and active targeting. Oberoi et al. prepared cisplatin core cl-micelles using poly(ethylene
Many review articles discussed the advances and applications of PMs in glycol)-b-poly(methacrylic acid) (PEG-b-PMA) where cross linking was
cancer [236240]. Although a signicant number of PMs formulations performed using 1,2-ethylenediamine (ED) and 1-(3-dimethylamino-
for treatment of cancer reached the clinical trials phase, none of them propyl)-3-ethylcarbodiimidehydrochloride (EDC) in presence of CaCl2.
was for brain cancer [235,241]. The obtained cl-micelles were a nanogel assembly with a particle size
ranging from 100 to 200 nm with a narrow PDI. The drug incorporation
B) Imaging reached 42% and the tuned cross linking controlled the drug release
eciently [243]. Desale et al. used the same bifunctional cross linker
Earlier disease detection, assessment of therapy and disease condi- for the preparation of PMs with multicompartment morphology to in-
tions have been widely improved using magnetic resonance imaging clude both the hydrophilic cisplatin and the hydrophobic PTX. The
(MRI), nuclear imaging, and X-ray computed tomography (CT) [235]. amphiphilic poly(ethylene glycol)-poly(glutamic acid)-poly(phenylala-
Zhou et al. developed optical MRI bifunctional targeted PMs for glioma nine) (PEGPGluPPhe) triblock copolymer was used [247]. Wu et al.
to determine its location before and during tumor resection operation were able to synthesize a phenol-containing amphiphilic diblock co-
[208]. The PMs were prepared from PEG-b-PCL encapsulating hydro- polymer poly(ethylene glycol)-block-poly(N-isopropylacrylamide-co-N-
phobic superparamagnetic iron oxide nanoparticles (SPIONs). The PMs (4-hydroxyphenethyl)acrylamide)(PEG-b-P(NIPAAm-co-NHPAAm)) by
were decorated with Lf which is a glioma- targeting ligand, in addition reversible addition-fragmentation chain transfer (RAFT) polymeriza-
to the modication with Cy5.5 which is a near-infrared uorescent tion of NHPAAm. This was followed by conjugation of the phenolic
probe. The glioma was obviously delineated due to Cy5.5 and Lf en- derivatives using horseradish peroxidase (HRP) in presence of H2O2.
sured high tumor specicity. Hence, good imaging properties were of- Finally, cl-micelles with cross linked core were self - aggregated at
fered by the newly synthesized contrast agent combining both magnetic ~37 C [248].
and uorescent properties. Finally the remarkable hydrophobicity of
PCL15000 in the used PEG-b-PCL made it possible to follow the brain 2- Stimuli responsive PMs
tumor after surgery or radiation even after the decrease of the nano
probe concentration in the brain. Similarly, Sun et al. prepared mixed i- Temperature-responsive PMs
SPIONs and gold micelles (GSMs) utilizing PEG-b-PCL copolymer for
both imaging and treatment of glioblastoma [221]. They found that the The amphiphilic copolymers with thermo-responsive blocks, are
combination between GSMs and radiation therapy (RT) resulted in ~ 2- smart copolymers that respond to temperature change by decreasing
fold increase in double-stranded DNA breaks density in tumor cells. their aqueous solubility, disrupting the micelles and provoking drug
They also showed that MRI but not CT was sensitive enough to detect release [238]. Most of the studies dealt with thermoresponsive poly-
and delineate borders of the tumor after GSMs administration and ac- mers presenting with lower critical solution temperature (LCST) or
cumulation. cloud point (CP) [235]. Below the LCST and due to hydrogen bonds, the
polymer is clear aqueous solutions. While, above this temperature the
4.4.3.6. Micelles in line with the modern strategies for brain targeting. polymer solution developed into a cloudy one (due to polymer pre-
cipitation). Higher temperature causes hydrogen bonds cleavage driven
A) Controlled dissociation PMs by an energetically more favorable situation rendering water less ar-
ranged with higher entropy. The polymer LCST regulation is usually
It is well known that PMs are easily dissociated when diluted in the done by incorporating either hydrophilic or hydrophobic monomers for
blood causing an early drug release. Accordingly, many approaches higher or lower temperatures, respectively [235]. One of the most
were developed to prevent this premature dissociation, e.g. cross linked widely used thermoresponsive polymer with LCST of 32 C is poly(N-
PMs, stimuli responsive PMs and cross linked- stimuli responsive PMs. isopropyl acrylamide) (PNIPAAm). pNIPAAm can form either the core
of PMs with PEG as the corona [249,250] or the shell with PCL as the
1- Cross linked PMs hydrophobic core [251,252]. Recently, Yu et al. used divalent metal
ions such as (Ba+ 2, Zn+ 2, Co+ 2) to graft sodium alginate with
Cross-linked micelles (cl-micelles) are prepared to prevent PMs de- pNIPAAm. The cross-linked alginate formed the core, while the corona
stabilization [242]. It can be hypothesized that cl-micelles behave as was made of PNIPAAm chains [253]. At 37 C, the hydrophobic
nanogels [242,243]. They can be prepared by: PNIPAAm chains helped in micellar dissociation with 5-uorouracil (5-
FU) release. However, no drug release was detected at 25 C.
i- Cross linking the shell
ii- pH-responsive PMs
This was done by Li et al. with the PMs prepared from poly(styrene-
b-butadiene-b-styrene) copolymer. They used 1,6-hexanediamine as a In normal extracellular matrix and blood, pH maintains at about
linker and produced clearly more stable micelles than the original ones 7.4, while the tumor microenvironment shows acidic pH (~ 6.5) due to
[244]. However, intermicellar cross linking is a common drawback of low oxygen supply in the intercellular environment [254]. The en-
this method, which would require the preparation at high dilution, a dosomal and lysosomal pHs are still lower ranging from 4.5 to 6 [254].
fact which decreases the process eciency [242]. Numerous studies have tried to prepare pH responsive PMs to deliver
drugs to tumors, endosomes or lysosomes [235,255257]. At acidic pH,
ii- Cross linking the core the carboxylate groups of orthoester, acetal, hydrazone, cis-acotinyl,
imine and oximeare easily protonated. Hence their covalent bonds
Core crosslinking was reported to have a dual action: rst avoidance break down releasing the drug in acidic environment only [255]. Cao
of PMs destabilization upon dilution and second the cross-linked core et al. prepared pH-sensitive PMs for DOX delivery from PEG-b-PCL
would control the burst release frequently encountered in PMs [245]. containing citraconic amide as pH-sensitive bond (PEG-pH-PCL) [258].
Talelli et al. summarized the most widely used methods for preparing The drug was released faster in the endosomes pH 5.5.This was

53
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

visualized by confocal laser microscopy and ow cytometry measure- Photo-responsive PMs are prepared from light-sensitive groups
ments containing copolymers, included in the core, corona, or at their inter-
face [238]. Upon exposure to the specic light stimulus which may be
iii- Redox-responsive PMs at the region of ultra-violet (UV), visible or near-infrared (NIR), the
photo-responsive polymers undergo structural modication which
Due to the abundance of glutathione (GSH) tripeptide in- causes sudden drug release. For example, azobenzene group and its
tracellularly, the intracellular uids are known to be in a less oxidized derivatives undergo reversible photoisomerization from trans to cis and
state than the extracellular uids. This will create a redox potential from cis to trans upon exposure to UV (300380 nm) and visible
[255]. Amphiphilic copolymers with disulphide linkage will be reduced (420490) light stimuli respectively. This phase change resulted in PMs
at low redox potential and hence will be redox responsive [255]. Hence, assembly disruption due to the more hydrophobicity of the trans-azo
contrary to pH-responsive micelles, redox-responsive micelles destabi- bond than the cis-azo bond [259261]. Due to its high skin absorption
lize and release drugs in the cytoplasm not in endosomes or lysosomes. UV light cannot inltrate the deep body tissue. Hence, it is only used for
The PMs core with a disulphide linkage will be cross-linked and when topical treatment. On the other side, NIR can go deep into the body up
present intracellularly drug release is initiated by the drop in redox to 10 cm, helped by the low absorption in the NIR region (650900 nm)
potential. Shoe et al. prepared itraconazole redox- responsive PMs from of hemoglobin, water and lipids [235]. Additionally, healthy tissues can
poly(ethylene glycol)-b-poly(L-lysine)-b-poly(L-phenylalanine) (PEG- be severely damaged due to the high absorbance of UV light by many
pLys-pPhe) for treatment of intracranial infections [133].The segment biological molecules [262]. Cabral et al. developed two groups of PMs,
of pPhe oered the hydrophobic core for hydrophobic drug itracona- one encapsulate captothecin (CPT), an antitumor drug, while the other
zole. The amino groups of pLys segments could react with a cross- encapsulate dendrimer phthalocyanine (DPc) [262]. The CPT-PMs were
linking agent containing disulphide bonds viz. 3,3-Dithiobis(sulfo- formed by a disulphide bond conjugation between a thiolated-camp-
succinimidylpropionate) (DTSSP) to produce a shell cl-micelles. The tothecin derivative (CPT-DP) and a thiolated poly(ethylene glycol)-poly
prepared redox-responsive PMs were able to shield the leakage of drug (glutamic acid) (PEG-b-P(Glu-DP)) block copolymer. As mentioned
in the circulation with enhanced drug release intracellulary. before, the disulphide bond is stable in blood and cleaved specically in
the intracellular reductive environment. DPc is a photosensitizer which
iv- pH and redox dual responsive PMs stimulates reactive oxygen formation when exposed to suitable wave-
lengths. DPc-PMs were prepared from poly(ethylene glycol)b-poly(L-
PMs can be designed to be responsive to both change in the pH and Lysine) (PEG-b-PLL). PMs were able to accumulate at the tumor site by
redox potential. Teo et al. prepared diblock copolymers of PEG and enhanced permeability and retention eect (EPR) and entered the cell
polycarbonate containing both disulphide bonds and carboxylic acid by endocytosis. The presence of DPc-PMs in the endosome allows a
groups (on the same monomer unit) via metal-free organo cataly- selective photo-damage of the endosomal membrane upon photo-irra-
ticpolymerization (ROP) of functional cyclic carbonate, and employed diation releasing the CPT-PMs in the cytosol. CPT is released in the
it to form pH and redox dual-responsive micelles [254]. cytosol upon the reduction in the redox potential. Huang et al. used NIR
irradiation to enhance the cellular uptake of nely tuned temperature-
v- Photo- responsive PMs responsive PMs [263]. They prepared PMs from poly(-caprolactone)-
block-poly(N-isopropylacrylamide-co-N,N-dimethylacrylamide) (PCL-b-

Table 4
History of surface modication of micelles for brain targeting.

Targeting ligand Pre- or post-conjugation Targeted moiety Targeted pathway Ref.

Tat Pre-conjugation ve charge on epithelial cells AMT [11,194,202,203]


PS80 Pre-conjugation LRP1 and LRP2 RMT [175,192]
Angiopep-2 Pre-conjugation LRP RMT [193]
NGR Pre-conjugation APN RMT [173]
cRGD Pre-conjugation v3 and v5 integrins RMT [109,191,211,218,219]
Angiopep-2 Post-conjugation LRP RMT [196,197]
Tf Post-conjugation TfR RMT [171,190]
Tf Post-conjugation TfR RMT [12]
cRGD Pre-conjugation v3 and v5 integrins
cRGD Post-conjugation v3 and v5 integrins RMT [172]
TGN Pre-conjugation NA RMT [198]
FB4 aptamer Post-conjugation TfR RMT [199]
DHA Pre-conjugation GLUT1 CMT [133]
Glucose and FA Pre-conjugation GLUT1 CMT [205]
FA-receptors RMT
Lt Pre-conjugation LfR RMT [177]
CREKA Pre-conjugation Fibrin deposited in tumors EPR [204]
RVG Post-conjugation Ach receptors RMT [108]
Lt Post-conjugation LfR RMT [208]
PS80 Post-conjugation LRP1 and LRP2 RMT [186]
Tf Pre-conjugation TfR RMT [13]
Td Pre-conjugation LAT1 CMT [207]
OX26 Pre-conjugation TfR/CD71 RMT [215]
T7 Pre-conjugation TfR RMT [220]
GLUT-1 scFv Post-conjugation GLUT1 CMT [222]

Ach: acetyl choline; AMT: adsorbtive- mediated transcytosis; APN: aminopeptidase N; CMT: carrier-mediated transport; cRGD: cyclic Arginine-Glycine-Aspartic acid; DHA: dehy-
droascorbic acid; FA: folic acid; GLUT-1 scFv: GLUT1 antibody single chain fragment variable; LAT1: large neutral amino-acid transporter type 1; LRP: lipoprotein receptor-related
protein; Lt: lactoferrin; LtR: lactoferrin receptor; NGR: Asn-Gly-Arg peptide; OX26: an anti-TfR antibody; PS80: polysorbate 80; RMT: receptor- mediated transcytosis; RVG: rabies virus
glycoprotein; T7: His-Ala-Ile-Tyr-Pro-Arg-His peptide; Tat: trans-activating transcriptional activator; Td: tryptophan derivative; Tf: transferrin; TfR: transferrin receptor; TGN:
TGNYKALHPHNG peptide.

54
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

p(NIPAAm-co-DMA)), with nely tuned compositions of p(NIPAAm-co- et al. with cRGD based on the same chemistry [172]. v3 and v5
DMA) for desirable LCST. NIR irradiation increased PMs and cell integrins are generally over expressed on endothelial cells of tumor
membranes interactions due to the hydrophilic to hydrophobic transi- angiogenic vessels and glioblastoma cells have been selectively targeted
tion of p(NIPAAm-co-DMA) shells. The cellular uptake of the PMs and by cRGD [172]. The ecacy of a platinum anticancer drug was found
therapeutic benets of loaded anticancer drugs were dramatically highly enhanced when loaded in 20% cRGD-linked micelles in contrast
promoted. to non-targeted PMs.
PepTGN, a specic peptide capable of binding to a specic brain
3- Cross linked-stimuli responsive PMs. protein, was prepared by phage display techniques and introduced
during the last ve years [21,198,265270]. TGN is a 12-amino acid
PMs starting drug release upon subjection to stimuli were also cross peptide, TGNYKALHPHNG which when conjugated to PEG-PLGA na-
linked for a higher stability upon dilution. pH and redox sensitive core noparticles proved signicant superiority in crossing the BBB [21].
cl-micelles encapsulating DOX were prepared by Zhang et al. [245].The Qian et al. conjugated TGN simply to mal-PEG through its cysteine end
hydrophobic moiety and the amino group of DOX enabled its tight and [198]. The decorated polyplexes enhanced signicantly the BCECS
high core encapsulation in cl-micelles formed from PEG as the shell uptake and higher cell uorescence intensity was evident in YOYO-1-
while the core contained (n-butyl methacrylate) and carboxylic acid labeled TGN-PEG-PDMAEMA/DNA polyplexes in comparison to the
groups (methacrylic acid), by both hydrophobic and ionic interactions. unmodied polyplexes [198].
The long circulation was ascertained by the PEG shell. Moreover, the Another simple technique of conjugation, that depends on N-hy-
core was cross linked via disulphide linkage to beat the noxious eect of droxysuccinimidyl PEG (NHS-PEG) rather than mal-PEG was presented
CMC rise, resulting from the presence of the hydrophilic carboxylic acid in many projects [173]. A stable amide bond was obtained due to the
groups inside the micellar core. After entering the cancer cells, a high reaction between the terminal amino group of the cyclic Asn-Gly-Arg
drug release rate was possible due to: rst, the reduction of the PMs (NGR) and NHS-PEG [173] (Fig. 6). A twice higher cellular uptake of
disulphide cross linkages in the existence of high amount of in- the modied micelles with a signicant inhibition in the cell pro-
tracellular glutathione; second, the low pH environment protonation of liferation was proven when compared to the unmodied micelles
the carboxylate groups breaking the bond with the DOX amino group. [173]. Additionally, Mu et al. conjugated FB4 aptamer to PEG-PLA
Recently, a novel redox-responsive core cl-micelles was prepared by Wu micelles via this technique [199]. They showed that the surface mod-
et al. using a cross linker with a disulphide bond, 3,30-dithiodipro- ication increased the cellular uptake in bEND5 by a 2.2-fold.
pionic acid (DPA), with mPEGylated starch (mPEG-St) copolymer Tf is another popular ligand eectively trancytosed across the BBB
[264]. The authors concluded that core-cross linked redox-responsive via binding TfR. Zhang et al. conjugated Tf to mal-PEG-PCL copolymer
mPEG-St-DPA micelles have high abilities for cancer therapy. [171]. According to the general scheme described before (Fig. 6),
briey, Tf was activated with 2-iminothiolane hydrochloride (ITH) at a
B) Active targeted PMs molar ratio 1:20. The thiolated Tf was then puried, and extent of
thiolation was determined [12,171]. Both cell and brain uptake proved
In addition to their various advantages, the propensity of PMs almost a two-fold increase in the tagged micelles than their unmodied
decoration with ligands able to target the brain gives them a special counterpart. The TfR mediated transcytosis was further conrmed by
appeal. In the last decade, the two principle brain targeted pathways inhibited cellular uptake in the presence of free Tf [171].
were RMT and CMT pathways. Both pre- and post-formulation con- Thereafter, Zhang et al. conjugated PTX to cRGD to obtain cRGD-
jugation strategies were applied. Table 4 summarizes the surface PTX conjugate (RP) which was encapsulated into Tf modied PCL-PEEP
modications on the micellar surface in the last decade for brain de- polymeric micelles (TRPM). The study proved an enhanced cellular
livery. uptake in BMECs (2.4-fold in comparison to unmodied micelles) with
a specic superior accumulation of the drug in mice brain glioma tumor
1- Micellar targeting for RMT pathway and peritumor areas following intravenous injection [12].
Lf is a multifunctional protein of Tf family. Lf proved to be more
Angiopep-2 - a member of angiopeps peptide family obtained from a advantageous for brain targeting than Tf due to its much lower plasma
natural occurring protein - proved to cross the BBB at LRP-1 better than concentration than the saturation value of its receptors, ability to allow
other proteins such as aprotinin and transferrin. Being entangled in more chemicals across the BBB and participation in the brain lesions
several cellular processes, LRP-1 has been used as a therapeutic pathogenesis [177]. Besides, its receptors (LfR) expression was higher
gateway to the brain. The fact of being over-expressed in malignant
astrocytomas, especially in glioblastomas encouraged scientists to use it
for brain cancer therapy [193]. Shen et al. synthesized cysteine an-
giopep-2 (TFFYGGSRGKRNNFKTEEYC) and conjugated it to mal-PEG-
PLA copolymer via simple one-step chemistry (click chemistry) ap-
plying the pre-conjugation strategy [193]. Shen et al. showed very
rapid brain uptake of angiopep-2 conjugated PMs immediately after IV
injection in comparison to the unmodied PMs. On the same chemical
basis, Shao et al. conjugated angiopep-2 to mal-PEG-PE copolymer
applying post-conjugation strategy [196]. The interaction between an-
giopep-2 and mal-PEG portion of any copolymer is simple click chem-
istry too. A stable thioether conjugate was formed between the thiol
group in angiopep-2 and the mal-PEG. Simply, angiopep-2 is added to
mal-copolymer or to mal-conjugated micelles in phosphate buer saline
at pH = 7.27.4 with stirring for 12 h in darkness at room temperature.
Unconjugated angiopep-2 was then removed by dialysis [193,196].
Fig. 6 shows the general scheme for interaction between maliemide
moiety (usually attached to PEG) and the thiol group of any ligand,
which is very popular in ligand conjugation. Fig. 6. General scheme for interaction between A) Mal reagent and a thiol group B) NHS
Mal-PEG-b-poly-L-glutamic acid PMs were conjugated by Miura reagent and an amine group.

55
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

magic bullets. The expression of magic bullet for brain delivery means
that the delivery system reaches the BBB, directly, without any
instability problems; escaping any immune reaction, overcomes the
challenge of BBB, readily uptaken by the brain and reaches the aected
area to exert its pharmacological action. PMs can be modulated to
behave as magic bullets as follows:

1) The small particle size (10100 nm) and the hydrophilic shell (helps
also in shielding of charge) of PMs help in escaping from the RES.
2) Core or shell cross-linking or stimulus responsive would help in
avoidance of instability upon circulatory dilution.
3) Decoration of the PMs surface with a ligand that targets a certain
pathway in the brain helps in the BBB passage.
4) Decoration of the PMs surface with a ligand - or being conjugated to
the drug - that targets special tissues or receptors in the brain im-
proves the eciency and minimizes the side eects.

As shown in Fig. 7, if the PMs preparation combines all the previous


Fig. 7. An illustration of the trip of magic bullet PMs after IV injection to its targeted
criteria, they will act as brain magic bullets that deliver therapeutic
location in the brain. agents directly to the desired location in the brain.

in patients with acute stroke and Parkinson's disease [271,272]. Zheng 5. Conclusion
et al. conjugated Lf to sodium alginate-cholesterol derivatives (SACDs)
simply by reaction with activated NHS-SACDs [177]. The Lf conjugated Numerous strategies have been adopted to trigger dierent ap-
PMs of the cholesterol derivative showed higher brain uptake in mice proaches to overcome limited brain transport. Nevertheless, nano-
than the unmodied micelles. technology remains of paramount signicance for CNS delivery. The
wide use of dierent nanocarriers has led to a wide array of techniques
2- Micellar targeting for CMT pathway. for improving brain targeting. PMs with a controlled, targeted and
smart drug release would help in the production of the so called magic
D-Glucose and other brain energy supply hexoses cross the brain via bullets for brain delivery. Dissociation controlled PMs which are de-
GLUT1. DHA, an oxidized form of ascorbic acid, is one a GLUT1 sub- corated with multiple ligands that target a certain brain pathway and a
strates which is rapidly reduced to ascorbic acid once its uptake by the certain area inside the brain was already tried. Collectively, this will
brain. Ascorbic acid itself isn't one of GLUT1 substrates, which will open a new era for friendly non-invasive treatment of CNS diseases.
guarantee its presence in a high concentration in the brain. Hence, DHA Besides, the shift from initial pragmatic delivery ways towards tailored
enjoys an eective one-way ux across the BBB compared with D- multifunctional PMs would oer more eective treatment of mental
glucose, which suers from bidirectional uxes. and neurobiological disorders, which account for 11% of disease
Shoa et al. conjugated DHA to poly(ethylene glycol)-b-poly(L-ly- burden all over the world.
sine)-b-poly(L-phenylalanine) (PEG-pLys-pPhe) to construct active tar-
geted PMs [133]. Propargylic DHA was linked to the terminus of N3- References
PEG-b-PLL- PPhe via a click reaction of the copper (I)-catalyzed azi-
dealkyne. In comparison to unmodied micelles, these PMs exhibited [1] R.K. Upadhyay, Drug delivery systems, CNS protection, and the blood brain bar-
rier, Biomed. Res. Int. 2014 (2014) 137.
improved cellular uptake by 6 fold in-vitro and 1.48 in-vivo [133]. [2] E. Barbu, E. Molnar, J. Tsibouklis, D.C. Gorecki, The potential for nanoparticle-
based drug delivery to the brain: overcoming the blood-brain barrier, Expert Opin.
3- Dual targeting PMs Drug Deliv. 6 (2009) 553565.
[3] A. Domnguez, A. lvarez, E. Hilario, B. Suarez-merino, F. Goi-de-Cerio, Central
nervous system diseases and the role of the blood-brain barrier in their treatment,
Another very interesting approach in brain targeting is the dual Neurosci. Discov. 1 (2013) 111.
targeting. In this approach, nanoparticles are decorated by two types of [4] P. Blasi, S. Giovagnoli, A. Schoubben, M. Ricci, C. Rossi, Solid lipid nanoparticles
for targeted brain drug delivery, Adv. Drug Deliv. Rev. 59 (2007) 454477.
ligands; one ligand targets a special pathway to cross the BBB while the
[5] M.I. Alam, S. Beg, A. Samad, S. Baboota, K. Kohli, J. Ali, A. Ahuja, M. Akbar,
other targets a certain location in the brain. Niu et al. conjugated both Strategy for eective brain drug delivery, Eur. J. Pharm. Sci. 40 (2010) 385403.
glucose and FA to Pluronic P105 PMs designed for DOX delivery named [6] C.E. Johanson, E.G. Stopa, P.N. McMillan, The blood-cerebrospinal uid barrier:
(GF-DOX). Glucose was added to target the CMT pathway via structure and functional signicance, Methods Mol. Biol. 686 (2011) 101131.
[7] Y. Chen, L. Liu, Modern methods for delivery of drugs across the blood-brain
GLUT1while FA was added to target brain tumor cells per se [205]. barrier, Adv. Drug Deliv. Rev. 64 (2012) 640665.
After 8.0 h GF-DOX showed a 2-fold increase in the uptake of C6 cells, [8] D.J. Mc Carthy, M. Malhotra, A.M. O'Mahony, J.F. Cryan, C.M. O'Driscoll,
in comparison to unmodied micelles or micelles decorated with glu- Nanoparticles and the blood-brain barrier: advancing from in-vitro models to-
wards therapeutic signicance, Pharm. Res. 32 (2015) 11611185.
cose only, encouraging such approach for successful brain targeting. [9] N. Nishiyama, K. Kataoka, Current state, achievements, and future prospects of
Dual targeting strategy was introduced dierently in some other polymeric micelles as nanocarriers for drug and gene delivery, Pharmacol. Ther.
researches, where PMs were decorated by one ligand that targets a 112 (2006) 630648.
[10] Y. Lu, K. Park, Polymeric micelles and alternative nanonized delivery vehicles for
specic brain pathway while the other ligand - which targets specic poorly soluble drugs, Int. J. Pharm. 453 (2013) 198214.
receptors in the brain - was conjugated to the drug itself. As mentioned [11] L. Liu, S.S. Venkatraman, Y.Y. Yang, K. Guo, J. Lu, B. He, S. Moochhala, L. Kan,
before, Zhang et al. applied this strategy for brain targeting where they Polymeric micelles anchored with TAT for delivery of antibiotics across the blood-
brain barrier, Biopolymers 90 (2008) 617623.
conjugated PTX to cRGD followed by encapsulation into Tf modied
[12] P. Zhang, L. Hu, Q. Yin, L. Feng, Y. Li, Transferrin-modied c[RGDfK]-paclitaxel
PCL-PEEP micelles. loaded hybrid micelle for sequential blood-brain barrier penetration and glioma
targeting therapy, Mol. Pharm. 9 (2012) 15901598.
[13] P. Agrawal Sonali, R.P. Singh, C.V. Rajesh, S. Singh, M.R. Vijayakumar,
B.L. Pandey, M.S. Muthu, Transferrin receptor-targeted vitamin E TPGS micelles
4.4.3.7. PMs as magic bullets for brain delivery. PMs are very promising for brain cancer therapy: preparation, characterization and brain distribution in
nanocarriers for brain delivery, which can be modulated to behave as

56
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

rats, Drug Deliv. 23 (2016) 17881798. system, Adv. Drug Deliv. Rev. 64 (2012) 614628.
[14] T.A. Saraan, C. Montes, T. Imura, J. Qi, G. Coppola, D.H. Geschwind, [44] C.T. Lu, R.R. Jin, Y.N. Jiang, Q. Lin, W.Z. Yu, K.L. Mao, F.R. Tian, Y.P. Zhao,
M.V. Sofroniew, Disruption of astrocyte STAT3 signaling decreases mitochondrial Y.Z. Zhao, Gelatin nanoparticle-mediated intranasal delivery of substance P pro-
function and increases oxidative stress in vitro, PLoS One 5 (2010) e9532. tects against 6-hydroxydopamine-induced apoptosis: an in vitro and in vivo study,
[15] G. Allt, J.G. Lawrenson, Pericytes: cell biology and pathology, Cells Tissues Organs Drug Des. Devel. Ther. 9 (2015) 19551962.
169 (2001) 111. [45] S. Yadav, F. Gattacceca, R. Panicucci, M.M. Amiji, Comparative biodistribution
[16] N.J. Abbott, A.A. Patabendige, D.E. Dolman, S.R. Yusof, D.J. Begley, Structure and and pharmacokinetic analysis of cyclosporine-A in the brain upon intranasal or
function of the blood-brain barrier, Neurobiol. Dis. 37 (2010) 1325. intravenous administration in an oil-in-water nanoemulsion formulation, Mol.
[17] L. Biddlestone-Thorpe, N. Marchi, K. Guo, C. Ghosh, D. Janigro, K. Valerie, Pharm. 12 (2015) 15231533.
H. Yang, Nanomaterial-mediated CNS delivery of diagnostic and therapeutic [46] J. Xi, Z. Zhang, X.A. Si, Improving intranasal delivery of neurological nanomedi-
agents, Adv. Drug Deliv. Rev. 64 (2012) 605613. cine to the olfactory region using magnetophoretic guidance of microsphere car-
[18] W.M. Pardridge, Drug transport across the blood-brain barrier, J. Cereb. Blood riers, Int. J. Nanomedicine 10 (2015) 12111222.
Flow Metab. 32 (2012) 19591972. [47] D. Sharma, R.K. Sharma, N. Sharma, R. Gabrani, S.K. Sharma, J. Ali, S. Dang,
[19] S.A. Pathan, Z. Iqbal, S.M. Zaidi, S. Talegaonkar, D. Vohra, G.K. Jain, A. Azeem, Nose-to-brain delivery of PLGA-diazepam nanoparticles, AAPS PharmSciTech 16
N. Jain, J.R. Lalani, R.K. Khar, F.J. Ahmad, CNS drug delivery systems: novel (2015) 11081121.
approaches, Recent Pat. Drug Deliv. Formul. 3 (2009) 7189. [48] S. Maimaiti, K.L. Anderson, C. DeMoll, L.D. Brewer, B.A. Rauh, J.C. Gant,
[20] A.G. de Boer, P.J. Gaillard, Drug targeting to the brain, Annu. Rev. Pharmacol. E.M. Blalock, N.M. Porter, O. Thibault, Intranasal insulin improves age-related
Toxocol. 47 (2007) 323355. cognitive decits and reverses electrophysiological correlates of brain aging, J.
[21] J. Li, L. Feng, L. Fan, Y. Zha, L. Guo, Q. Zhang, J. Chen, Z. Pang, Y. Wang, X. Jiang, Gerontol. A Biol. Sci. Med. Sci. 71 (2015) 3039.
V.C. Yang, L. Wen, Targeting the brain with PEG-PLGA nanoparticles modied [49] T. Kanazawa, Brain delivery of small interfering ribonucleic acid and drugs
with phage-displayed peptides, Biomaterials 32 (2011) 49434950. through intranasal administration with nano-sized polymer micelles, Med. Devices
[22] W.M. Pardridge, Blood-brain barrier drug targeting: the future of brain drug de- (Auckl) 8 (2015) 5764.
velopment, Mol. Interv. 3 (2003) 90105. [50] P. Kaur, T. Garg, B. Vaidya, A. Prakash, G. Rath, A.K. Goyal, Brain delivery of
[23] G. Xiao, L.-S. Gan, Receptor-mediated endocytosis and brain delivery of ther- intranasal in situ gel of nanoparticulated polymeric carriers containing anti-
apeutic biologics, Int. J. Cell Biol. 2013 (2013) (Article ID 703545). depressant drug: behavioral and biochemical assessment, J. Drug Target. 23
[24] N. Bien-Ly, Y.J. Yu, D. Bumbaca, J. Elstrott, C.A. Boswell, Y. Zhang, W. Luk, Y. Lu, (2015) 275286.
M.S. Dennis, R.M. Weimer, I. Chung, R.J. Watts, Transferrin receptor (TfR) traf- [51] A. Garzon-Aburbeh, J.H. Poupaert, M. Claesen, P. Dumont, A lymphotropic pro-
cking determines brain uptake of TfR antibody anity variants, J. Exp. Med. 211 drug of L-dopa: synthesis, pharmacological properties, and pharmacokinetic be-
(2014) 233244. havior of 1,3-dihexadecanoyl-2-[(S)-2-amino-3-(3,4-dihydroxyphenyl)prop anoyl]
[25] W.M. Pardridge, J. Eisenberg, J. Yang, Human blood-brain barrier insulin re- propane-1,2,3-triol, J. Med. Chem. 29 (1986) 687691.
ceptor, J. Neurochem. 44 (1985) 17711778. [52] L. Peura, K. Malmioja, K. Huttunen, J. Leppanen, M. Hamalainen, M.M. Forsberg,
[26] K. Yamada, T. Hashimoto, C. Yabuki, Y. Nagae, M. Tachikawa, D.K. Strickland, M. Gynther, J. Rautio, K. Laine, Design, synthesis and brain uptake of LAT1-tar-
Q. Liu, G. Bu, J.M. Basak, D.M. Holtzman, S. Ohtsuki, T. Terasaki, T. Iwatsubo, The geted amino acid prodrugs of dopamine, Pharm. Res. 30 (2013) 25232537.
low density lipoprotein receptor-related protein 1 mediates uptake of amyloid beta [53] A.T. Placzek, S.J. Ferrara, M.D. Hartley, H.S. Sanford-Crane, J.M. Meinig,
peptides in an in vitro model of the blood-brain barrier cells, J. Biol. Chem. 283 T.S. Scanlan, Sobetirome prodrug esters with enhanced blood-brain barrier per-
(2008) 3455434562. meability, Bioorg. Med. Chem. 24 (2016) 58425854.
[27] P. Wang, Y. Xue, X. Shang, Y. Liu, Diphtheria toxin mutant CRM197-mediated [54] W.M. Pardridge, A.K. Kumagai, J.B. Eisenberg, Chimeric peptides as a vehicle for
transcytosis across blood-brain barrier in vitro, Cell. Mol. Neurobiol. 30 (2010) peptide pharmaceutical delivery through the blood-brain barrier, Biochem.
717725. Biophys. Res. Commun. 146 (1987) 307313.
[28] W. Lu, Adsorptive-mediated brain delivery systems, Curr. Pharm. Biotechnol. 13 [55] W.M. Pardridge, D. Triguero, J.L. Buciak, Beta-endorphin chimeric peptides:
(2012) 23402348. transport through the blood-brain barrier in vivo and cleavage of disulde linkage
[29] M. Demeule, A. Regina, J. Jodoin, A. Laplante, C. Dagenais, F. Berthelet, by brain, Endocrinology 126 (1990) 977984.
A. Moghrabi, R. Beliveau, Drug transport to the brain: key roles for the eux [56] S. Zanin, M. Sandre, G. Cozza, D. Ottaviani, O. Marin, L.A. Pinna, M. Ruzzene,
pump P-glycoprotein in the blood-brain barrier, Vasc. Pharmacol. 38 (2002) Chimeric peptides as modulators of CK2-dependent signaling: mechanism of ac-
339348. tion and o-target eects, Biochim. Biophys. Acta 1854 (2015) 16941707.
[30] W. Loscher, H. Potschka, Blood-brain barrier active eux transporters: ATP- [57] E. Kobayashi, H. Kishi, T. Ozawa, H. Hamana, H. Nakagawa, A. Jin, Z. Lin,
binding cassette gene family, NeuroRx 2 (2005) 8698. A. Muraguchi, A chimeric antigen receptor for TRAIL-receptor 1 induces apoptosis
[31] C.J. Ek, A. Wong, S.A. Liddelow, P.A. Johansson, K.M. Dziegielewska, in various types of tumor cells, Biochem. Biophys. Res. Commun. 453 (2014)
N.R. Saunders, Eux mechanisms at the developing brain barriers: ABC-trans- 798803.
porters in the fetal and postnatal rat, Toxicol. Lett. 197 (2010) 5159. [58] B.A. Baldo, Chimeric fusion proteins used for therapy: indications, mechanisms,
[32] A. Bodo, E. Bakos, F. Szeri, A. Varadi, B. Sarkadi, The role of multidrug trans- and safety, Drug Saf. 38 (2015) 455479.
porters in drug availability, metabolism and toxicity, Toxicol. Lett. 140141 [59] E. Condamine, K. Courchay, J.C. Rego, J. Leprince, C. Mayer, D. Davoust,
(2003) 133143. J. Costentin, H. Vaudry, Structural and pharmacological characteristics of chi-
[33] R.J. Boado, E.K.W. Hui, J.Z. Lu, R.K. Sumbria, W.M. Pardridge, Blood-brain barrier meric peptides derived from peptide E and beta-endorphin reveal the crucial role
molecular trojan horse enables imaging of brain uptake of radioiodinated re- of the C-terminal YGGFL and YKKGE motifs in their analgesic properties, Peptides
combinant protein in the rhesus monkey, Bioconjug. Chem. 24 (2013) 17411749. 31 (2010) 962972.
[34] K. Park, Trojan monocytes for improved drug delivery to the brain, J. Control. [60] W.A. Banks, Peptides and the blood-brain barrier, Peptides 72 (2015) 1619.
Release 132 (2008) 75. [61] I. Brasnjevic, H.W. Steinbusch, C. Schmitz, P. Martinez-Martinez, Delivery of
[35] J.M. Larsen, D.R. Martin, M.E. Byrne, Recent advances in delivery through the peptide and protein drugs over the blood-brain barrier, Prog. Neurobiol. 87 (2009)
blood-brain barrier, Curr. Top. Med. Chem. 14 (2014) 11481160. 212251.
[36] J.C. Roth, K.A. Cassady, J.J. Cody, J.N. Parker, K.H. Price, J.M. Coleman, [62] M.A. Deli, Potential use of tight junction modulators to reversibly open mem-
J.O. Peggins, P.E. Noker, N.W. Powers, S.D. Grimes, S.L. Carroll, G.Y. Gillespie, branous barriers and improve drug delivery, Biochim. Biophys. Acta 1788 (2009)
R.J. Whitley, J.M. Markert, Evaluation of the safety and biodistribution of M032, 892910.
an attenuated herpes simplex virus type 1 expressing hIL-12, after intracerebral [63] N.K. Acharya, E.L. Goldwaser, M.M. Forsberg, G.A. Godsey, C.A. Johnson,
administration to aotus nonhuman primates, Hum. Gene Ther. Clin. Dev. 25 A. Sarkar, C. DeMarshall, M.C. Kosciuk, J.M. Dash, C.P. Hale, D.M. Leonard,
(2014) 1627. D.M. Appelt, R.G. Nagele, Sevourane and isourane induce structural changes in
[37] S. Reyes, N. Tajiri, C.V. Borlongan, Developments in intracerebral stem cell grafts, brain vascular endothelial cells and increase blood-brain barrier permeability:
Expert. Rev. Neurother. 15 (2015) 381393. possible link to postoperative delirium and cognitive decline, Brain Res. 1620
[38] D.R. Groothuis, H. Benalcazar, C.V. Allen, R.M. Wise, C. Dills, C. Dobrescu, (2015) 2941.
V. Rothholtz, R.M. Levy, Comparison of cytosine arabinoside delivery to rat brain [64] C.P. Foley, D.G. Rubin, A. Santillan, D. Sondhi, J.P. Dyke, Y.P. Gobin, R.G. Crystal,
by intravenous, intrathecal, intraventricular and intraparenchymal routes of ad- D.J. Ballon, Intra-arterial delivery of AAV vectors to the mouse brain after man-
ministration, Brain Res. 856 (2000) 281290. nitol mediated blood brain barrier disruption, J. Control. Release 196 (2014)
[39] E.H. Lo, A.B. Singhal, V.P. Torchilin, et al., Drug delivery to damaged brain, Brain 7178.
Res. Rev. 38 (2001) 140148. [65] G.S. Gonzales-Portillo, P.R. Sanberg, M. Franzblau, C. Gonzales-Portillo,
[40] C.H. Yang, X. Tian, H.B. Yin, X.H. Gao, N. Li, Sedation and analgesia with fentanyl T. Diamandis, M. Staples, C.D. Sanberg, C.V. Borlongan, Mannitol-enhanced de-
and etomidate for intrathecal injection in childhood leukemia patients, Medicine livery of stem cells and their growth factors across the blood-brain barrier, Cell
(Baltimore) 94 (2015) e361. Transplant. 23 (2014) 531539.
[41] T. Nishikawa, Y. Okamoto, S. Maruyama, T. Tanabe, K. Kurauchi, Y. Kodama, [66] P. Hulper, S. Veszelka, F.R. Walter, H. Wolburg, P. Fallier-Becker, J. Piontek,
S. Nakagawa, Y. Shinkoda, Y. Kawano, Acute encephalomyelitis complicated with I.E. Blasig, M. Lakomek, W. Kugler, M.A. Deli, Acute eects of short-chain alkyl-
severe neurological sequelae after intrathecal administration of methotrexate in a glycerols on blood-brain barrier properties of cultured brain endothelial cells, Br.
patient with acute lymphoblastic leukemia, Rinsho Ketsueki 55 (2014) J. Pharmacol. 169 (2013) 15611573.
23062310. [67] B. Erdlenbruch, C. Schinkhof, W. Kugler, D.E. Heinemann, J. Herms, H. Eibl,
[42] J. Ali, M. Ali, S. Baboota, J.K. Sahani, C. Ramassamy, L. Dao, Bhavna, Potential of M. Lakomek, Intracarotid administration of short-chain alkylglycerols for in-
nanoparticulate drug delivery systems by intranasal administration, Curr. Pharm. creased delivery of methotrexate to the rat brain, Br. J. Pharmacol. 139 (2003)
Des. 16 (2010) 16441653. 685694.
[43] J.J. Lochhead, R.G. Thorne, Intranasal delivery of biologics to the central nervous [68] N.H. On, S. Savant, M. Toews, D.W. Miller, Rapid and reversible enhancement of

57
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

blood-brain barrier permeability using lysophosphatidic acid, J. Cereb. Blood Flow [95] J.S. Zhang, F. Liu, L. Huang, Implications of pharmacokinetic behavior of lipoplex
Metab. 33 (2013) 19441954. for its inammatory toxicity, Adv. Drug Deliv. Rev. 57 (2005) 689698.
[69] V. Monnaert, D. Betbeder, L. Fenart, H. Bricout, A.M. Lenfant, C. Landry, [96] S.I. Jenkins, D. Weinberg, A.F. al-Shakli, A.R. Fernandes, H.H.P. Yiu, N.D. Telling,
R. Cecchelli, E. Monier, S. Tilloy, Eects of gamma- and hydroxypropyl-gamma- P. Roach, D.M. Chari, "Stealth" nanoparticles evade neural immune cells but also
cyclodextrins on the transport of doxorubicin across an in vitro model of blood- evade major brain cell populations: implications for PEG-based neurotherapeutics,
brain barrier, J. Pharmacol. Exp. Ther. 311 (2004) 11151120. J. Control. Release 224 (2016) 136145.
[70] A. Saija, P. Princi, D. Trombetta, M. Lanza, A. De Pasquale, Changes in the per- [97] A. Mori, A.L. Klibanov, V.P. Torchilin, L. Huang, Inuence of the steric barrier
meability of the blood-brain barrier following sodium dodecyl sulphate adminis- activity of amphipathic poly(ethyleneglycol) and ganglioside GM1 on the circu-
tration in the rat, Exp. Brain Res. 115 (1997) 546551. lation time of liposomes and on the target binding of immunoliposomes in vivo,
[71] Y.C. Kuo, C.Y. Shih-Huang, Solid lipid nanoparticles carrying chemotherapeutic FEBS Lett. 284 (1991) 263266.
drug across the blood-brain barrier through insulin receptor-mediated pathway, J. [98] J.F. Stefanick, J.D. Ashley, T. Kiziltepe, B. Bilgicer, A systematic analysis of pep-
Drug Target. 21 (2013) 730738. tide linker length and liposomal polyethylene glycol coating on cellular uptake of
[72] W.H. Hind, C. Tufarelli, M. Neophytou, S.I. Anderson, T.J. England, peptide-targeted liposomes, ACS Nano 7 (2013) 29352947.
S.E. O'Sullivan, Endocannabinoids modulate human blood-brain barrier perme- [99] X. Guo, F.C. Szoka Jr., Steric stabilization of fusogenic liposomes by a low-pH
ability in vitro, Br. J. Pharmacol. 172 (2015) 30153027. sensitive PEG-diortho ester-lipid conjugate, Bioconjug. Chem. 12 (2001) 291300.
[73] M.E. Downs, A. Buch, C. Sierra, M.E. Karakatsani, S. Chen, E.E. Konofagou, [100] W. Gao, R. Langer, O.C. Farokhzad, Poly(ethylene glycol) with observable shed-
V.P. Ferrera, Long-term safety of repeated blood-brain barrier opening via focused ding, Angew. Chem. Int. Ed. Eng. 49 (2010) 65676571.
ultrasound with microbubbles in non-human primates performing a cognitive task, [101] N.M. Wassef, G.R. Matyas, C.R. Alving, Complement-dependent phagocytosis of
PLoS One 10 (2015) e0125911. liposomes by macrophages: suppressive eects of "stealth" lipids, Biochem.
[74] J.L. Eberhardt, B.R. Persson, A.E. Brun, L.G. Salford, L.O. Malmgren, Blood-brain Biophys. Res. Commun. 176 (1991) 866874.
barrier permeability and nerve cell damage in rat brain 14 and 28 days after ex- [102] Y.C. Kuo, C.C. Lin, Rescuing apoptotic neurons in Alzheimer's disease using wheat
posure to microwaves from GSM mobile phones, Electromagn. Biol. Med. 27 germ agglutinin-conjugated and cardiolipin-conjugated liposomes with en-
(2008) 215229. capsulated nerve growth factor and curcumin, Int. J. Nanomedicine 10 (2015)
[75] R. Stam, Electromagnetic elds and the blood-brain barrier, Brain Res. Rev. 65 26532672.
(2010) 8097. [103] F. Helm, G. Fricker, Liposomal conjugates for drug delivery to the central nervous
[76] C.S. Karyekar, A. Fasano, S. Raje, R. Lu, T.C. Dowling, N.D. Eddington, Zonula system, Pharmaceutics 7 (2015) 2742.
occludens toxin increases the permeability of molecular weight markers and [104] Z. Liu, H. Zhao, L. Shu, Y. Zhang, C. Okeke, L. Zhang, J. Li, N. Li, Preparation and
chemotherapeutic agents across the bovine brain microvessel endothelial cells, J. evaluation of Baicalin-loaded cationic solid lipid nanoparticles conjugated with
Pharm. Sci. 92 (2003) 414423. OX26 for improved delivery across the BBB, Drug Dev. Ind. Pharm. 41 (2015)
[77] C. Lu, S.A. Diehl, R. Noubade, J. Ledoux, M.T. Nelson, K. Spach, J.F. Zachary, 353361.
E.P. Blankenhorn, C. Teuscher, Endothelial histamine H1 receptor signaling re- [105] S. Laserra, A. Basit, P. Sozio, L. Marinelli, E. Fornasari, I. Cacciatore, M. Ciulla,
duces blood-brain barrier permeability and susceptibility to autoimmune en- H. Turkez, F. Geyikoglu, A. Di Stefano, Solid lipid nanoparticles loaded with li-
cephalomyelitis, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 1896718972. poyl-memantine codrug: preparation and characterization, Int. J. Pharm. 485
[78] T. Ma, L. Liu, P. Wang, Y. Xue, Evidence for involvement of ROCK signaling in (2015) 183191.
bradykinin-induced increase in murine blood-tumor barrier permeability, J. [106] A. Lopalco, H. Ali, N. Denora, E. Rytting, Oxcarbazepine-loaded polymeric na-
Neuro-Oncol. 106 (2012) 291301. noparticles: development and permeability studies across in vitro models of the
[79] C.V. Borlongan, D.F. Emerich, Facilitation of drug entry into the CNS via transient blood-brain barrier and human placental trophoblast, Int. J. Nanomedicine 10
permeation of blood brain barrier: laboratory and preliminary clinical evidence (2015) 19851996.
from bradykinin receptor agonist, Cereport, Brain Res. Bull. 60 (2003) 297306. [107] P. Mastorakos, C. Zhang, S. Berry, Y. Oh, S. Lee, C.G. Eberhart, G.F. Woodworth,
[80] A.S. Lossinsky, A.W. Vorbrodt, H.M. Wisniewski, Scanning and transmission J.S. Suk, J. Hanes, Highly PEGylated DNA nanoparticles provide uniform and
electron microscopic studies of microvascular pathology in the osmotically im- widespread gene transfer in the brain, Adv. Healthc. Mater. 4 (2015) 10231033.
paired blood-brain barrier, J. Neurocytol. 24 (1995) 795806. [108] H. Huo, Y. Gao, Y. Wang, J. Zhang, Z.Y. Wang, T. Jiang, S. Wang, Polyion complex
[81] A. Mistry, S. Stolnik, L. Illum, Nanoparticles for direct nose-to-brain delivery of micelles composed of pegylated polyasparthydrazide derivatives for siRNA de-
drugs, Int. J. Pharm. 379 (2009) 146157. livery to the brain, J. Colloid Interface Sci. 447 (2015) 815.
[82] N. Bertrand, J. Wu, X. Xu, N. Kamaly, O.C. Farokhzad, Cancer nanotechnology: the [109] A.J. Li, Y.H. Zheng, G.D. Liu, W.S. Liu, P.C. Cao, Z.F. Bu, Ecient delivery of
impact of passive and active targeting in the era of modern cancer biology, Adv. docetaxel for the treatment of brain tumors by cyclic RGD-tagged polymeric mi-
Drug Deliv. Rev. 66 (2014) 225. celles, Mol. Med. Rep. 11 (2015) 30783086.
[83] C. Fang, B. Shi, Y.Y. Pei, M.H. Hong, J. Wu, H.Z. Chen, In vivo tumor targeting of [110] H. Wu, J. Li, Q. Zhang, X. Yan, L. Guo, X. Gao, M. Qiu, X. Jiang, R. Lai, H. Chen, A
tumor necrosis factor-alpha-loaded stealth nanoparticles: eect of MePEG mole- novel small Odorranalectin-bearing cubosomes: preparation, brain delivery and
cular weight and particle size, Eur. J. Pharm. Sci. 27 (2006) 2736. pharmacodynamic study on amyloid- 25-35-treated rats following intranasal
[84] G. Sonavane, K. Tomoda, K. Makino, Biodistribution of colloidal gold nano- administration, Eur. J. Pharm. Biopharm. 80 (2012) 368378.
particles after intravenous administration: eect of particle size, Colloids Surf. B: [111] M. Mozafari, Synthesis and characterisation of poly(lactide-co-glycolide) nano-
Biointerfaces 66 (2008) 274280. spheres using vitamin E emulsier prepared through one-step oil-in-water emul-
[85] M. Shilo, A. Sharon, K. Baranes, M. Motiei, J.P. Lellouche, R. Popovtzer, The eect sion and solvent evaporation techniques, IET Nanobiotechnol. 8 (2014) 257262.
of nanoparticle size on the probability to cross the blood-brain barrier: an in-vitro [112] H. Wang, W. Su, S. Wang, X. Wang, Z. Liao, C. Kang, L. Han, J. Chang, G. Wang,
endothelial cell model, J. Nanobiotechnology 13 (2015) 19. P. Pu, Smart multifunctional core-shell nanospheres with drug and gene co-loaded
[86] E. Mahon, A. Salvati, F. Baldelli Bombelli, I. Lynch, K.A. Dawson, Designing the for enhancing the therapeutic eect in a rat intracranial tumor model, Nano 4
nanoparticle-biomolecule interface for "targeting and therapeutic delivery", J. (2012) 65016508.
Control. Release 161 (2012) 164174. [113] F. Zhang, P. Mastorakos, M.K. Mishra, A. Mangraviti, L. Hwang, J. Zhou, J. Hanes,
[87] K. Xiao, Y. Li, J. Luo, J.S. Lee, W. Xiao, A.M. Gonik, R.G. Agarwal, K.S. Lam, The H. Brem, A. Olivi, B. Tyler, R.M. Kannan, Uniform brain tumor distribution and
eect of surface charge on in vivo biodistribution of PEG-oligocholic acid based tumor associated macrophage targeting of systemically administered dendrimers,
micellar nanoparticles, Biomaterials 32 (2011) 34353446. Biomaterials 52 (2015) 507516.
[88] H. Maeda, H. Nakamura, J. Fang, The EPR eect for macromolecular drug delivery [114] M.J. Serramia, S. Alvarez, E. Fuentes-Paniagua, M.I. Clemente, J. Sanchez-Nieves,
to solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and R. Gomez, J. de la Mata, M.A. Munoz-Fernandez, In vivo delivery of siRNA to the
distinct tumor imaging in vivo, Adv. Drug Deliv. Rev. 65 (2013) 7179. brain by carbosilane dendrimer, J. Control. Release 200 (2015) 6070.
[89] K. Chaturvedi, K. Ganguly, A.R. Kulkarni, W.E. Rudzinski, L. Krauss, [115] M. Gooding, M. Malhotra, D.J. McCarthy, B.M. Godinho, J.F. Cryan, R. Darcy,
M.N. Nadagouda, T.M. Aminabhavi, Oral insulin delivery using deoxycholic acid C.M. O'Driscoll, Synthesis and characterization of rabies virus glycoprotein-tagged
conjugated PEGylated polyhydroxybutyrate co-polymeric nanoparticles, amphiphilic cyclodextrins for siRNA delivery in human glioblastoma cells: in vitro
Nanomedicine (London) 10 (2015) 15691583. analysis, Eur. J. Pharm. Sci. 71 (2015) 8092.
[90] G. Marslin, A.M. Revina, V.K. Khandelwal, K. Balakumar, C.J. Sheeba, G. Franklin, [116] G. Rassu, E. Soddu, M. Cossu, A. Brundu, G. Cerri, N. Marchetti, L. Ferraro,
PEGylated ooxacin nanoparticles render strong antibacterial activity against R.F. Regan, P. Giunchedi, E. Gavini, A. Dalpiaz, Solid microparticles based on
many clinically important human pathogens, Colloids Surf. B: Biointerfaces 132 chitosan or methyl-beta-cyclodextrin: a rst formulative approach to increase the
(2015) 6270. nose-to-brain transport of deferoxamine mesylate, J. Control. Release 201 (2015)
[91] C. Conte, G. Costabile, I. d'Angelo, M. Pannico, P. Musto, G. Grassia, A. Ialenti, 6877.
P. Tirino, A. Miro, F. Ungaro, F. Quaglia, Skin transport of PEGylated poly(-ca- [117] J. Tong, X. Yi, R. Luxenhofer, W.A. Banks, R. Jordan, M.C. Zimmerman,
prolactone) nanoparticles assisted by (2-hydroxypropyl)-beta-cyclodextrin, J. A.V. Kabanov, Conjugates of superoxide dismutase 1 with amphiphilic poly(2-
Colloid Interface Sci. 454 (2015) 112120. oxazoline) block copolymers for enhanced brain delivery: synthesis, character-
[92] R.Z. Xiao, Z.W. Zeng, G.L. Zhou, J.J. Wang, F.Z. Li, A.M. Wang, Recent advances in ization and evaluation in vitro and in vivo, Mol. Pharm. 10 (2013) 360377.
PEG-PLA block copolymer nanoparticles, Int. J. Nanomedicine 5 (2010) [118] S. Dixit, T. Novak, K. Miller, Y. Zhu, M.E. Kenney, A.M. Broome, Transferrin re-
10571065. ceptor-targeted theranostic gold nanoparticles for photosensitizer delivery in brain
[93] W.L. Zhang, J.P. Liu, X.X. Liu, Z.Q. Chen, Stealth tanshinone IIA-loaded solid lipid tumors, Nano 7 (2015) 17821790.
nanoparticles: eects of poloxamer 188 coating on in vitro phagocytosis and in [119] Y. Cheng, Q. Dai, R.A. Morshed, X. Fan, M.L. Wegscheid, D.A. Wainwright, Y. Han,
vivo pharmacokinetics in rats, Yao Xue Xue Bao 44 (2009) 14211428. L. Zhang, B. Aunger, A.L. Tobias, E. Rincon, B. Thaci, A.U. Ahmed, P.C. Warnke,
[94] T.S. Levchenko, R. Rammohan, A.N. Lukyanov, K.R. Whiteman, V.P. Torchilin, C. He, M.S. Lesniak, Blood-brain barrier permeable gold nanoparticles: an ecient
Liposome clearance in mice: the eect of a separate and combined presence of delivery platform for enhanced malignant glioma therapy and imaging, Small 10
surface charge and polymer coating, Int. J. Pharm. 240 (2002) 95102. (2014) 51375150.

58
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

[120] E. Locatelli, M. Naddaka, C. Uboldi, G. Loudos, E. Fragogeorgi, V. Molinari, e0134722.


A. Pucci, T. Tsotakos, D. Psimadas, J. Ponti, M.C. Franchini, Targeted delivery of [147] C. Fillebeen, L. Descamps, M.P. Dehouck, L. Fenart, M. Benaissa, G. Spik,
silver nanoparticles and alisertib: in vitro and in vivo synergistic eect against R. Cecchelli, A. Pierce, Receptor-mediated transcytosis of lactoferrin through the
glioblastoma, Nanomedicine (London) 9 (2014) 839849. blood-brain barrier, J. Biol. Chem. 274 (1999) 70117017.
[121] L. Zhang, H. Yuan, L.M. Burk, C.R. Inscoe, M.J. Hadsell, P. Chtcheprov, Y.Z. Lee, [148] A. Tomitaka, H. Arami, S. Gandhi, K.M. Krishnan, Lactoferrin conjugated iron
J. Lu, S. Chang, O. Zhou, Image-guided microbeam irradiation to brain tumour oxide nanoparticles for targeting brain glioma cells in magnetic particle imaging,
bearing mice using a carbon nanotube x-ray source array, Phys. Med. Biol. 59 Nano 7 (2015) 1689016898.
(2014) 12831303. [149] I. Singh, R. Swami, D. Pooja, M.K. Jeengar, W. Khan, R. Sistla, Lactoferrin bio-
[122] G. Bardi, A. Nunes, L. Gherardini, K. Bates, K.T. Al-Jamal, C. Gaillard, M. Prato, conjugated solid lipid nanoparticles: a new drug delivery system for potential
A. Bianco, T. Pizzorusso, K. Kostarelos, Functionalized carbon nanotubes in the brain targeting, J. Drug Target. 24 (2016) 212223.
brain: cellular internalization and neuroinammatory responses, PLoS One 8 [150] A. Zensi, D. Begley, C. Pontikis, C. Legros, L. Mihoreanu, C. Buchel, J. Kreuter,
(2013) e80964. Human serum albumin nanoparticles modied with apolipoprotein A-I cross the
[123] R. Agarwal, M.S. Domowicz, N.B. Schwartz, J. Henry, I. Medintz, J.B. Delehanty, blood-brain barrier and enter the rodent brain, J. Drug Target. 18 (2010) 842848.
M.H. Stewart, K. Susumu, A.L. Huston, J.R. Deschamps, P.E. Dawson, V. Palomo, [151] M. Tamaru, H. Akita, T. Nakatani, K. Kajimoto, Y. Sato, H. Hatakeyama,
G. Dawson, Delivery and tracking of quantum dot peptide bioconjugates in an H. Harashima, Application of apolipoprotein E-modied liposomal nanoparticles
intact developing avian brain, ACS Chem. Neurosci. 6 (2015) 494504. as a carrier for delivering DNA and nucleic acid in the brain, Int. J. Nanomedicine
[124] K.C. Weng, R. Hashizume, C.O. Noble, L.P. Serwer, D.C. Drummond, D.B. Kirpotin, 9 (2014) 42674276.
A.M. Kuwabara, L.X. Chao, F.F. Chen, C.D. James, J.W. Park, Convection-en- [152] J. Kreuter, T. Hekmatara, S. Dreis, T. Vogel, S. Gelperina, K. Langer, Covalent
hanced delivery of targeted quantum dot-immunoliposome hybrid nanoparticles attachment of apolipoprotein A-I and apolipoprotein B-100 to albumin nano-
to intracranial brain tumor models, Nanomedicine (London) 8 (2013) 19131925. particles enables drug transport into the brain, J. Control. Release 118 (2007)
[125] L. Costantino, Drug delivery to the CNS and polymeric nanoparticulate carriers, 5458.
Future Med. Chem. 2 (2010) 16811701. [153] L. Wang, Y. Hao, H. Li, Y. Zhao, D. Meng, D. Li, J. Shi, H. Zhang, Z. Zhang,
[126] E.F. Craparo, M.L. Bondi, G. Pitarresi, G. Cavallaro, Nanoparticulate systems for Y. Zhang, Co-delivery of doxorubicin and siRNA for glioma therapy by a brain
drug delivery and targeting to the central nervous system, CNS Neurosci. Ther. 17 targeting system: angiopep-2-modied poly(lactic-co-glycolic acid) nanoparticles,
(2011) 670677. J. Drug Target. 23 (2015) 832846.
[127] J. Kreuter, Nanoparticulate systems for brain delivery of drugs, Adv. Drug Deliv. [154] S. Ruan, M. Yuan, L. Zhang, G. Hu, J. Chen, X. Cun, Q. Zhang, Y. Yang, Q. He,
Rev. 47 (2001) 6581. H. Gao, Tumor microenvironment sensitive doxorubicin delivery and release to
[128] J. Kreuter, Drug delivery to the central nervous system by polymeric nano- glioma using angiopep-2 decorated gold nanoparticles, Biomaterials 37 (2015)
particles: what do we know? Adv. Drug Deliv. Rev. 71 (2014) 214. 425435.
[129] T. Patel, J. Zhou, J.M. Piepmeier, W.M. Saltzman, Polymeric nanoparticles for [155] G. Tosi, A. Vilella, P. Veratti, D. Belletti, F. Pederzoli, B. Ruozi, M.A. Vandelli,
drug delivery to the central nervous system, Adv. Drug Deliv. Rev. 64 (2012) M. Zoli, F. Forni, Exploiting bacterial pathways for BBB crossing with PLGA na-
701705. noparticles modied with a mutated form of diphtheria toxin (CRM197): in vivo
[130] G. Tosi, L. Costantino, B. Ruozi, F. Forni, M.A. Vandelli, Polymeric nanoparticles experiments, Mol. Pharm. 12 (2015) 36723684.
for the drug delivery to the central nervous system, Expert Opin. Drug Deliv. 5 [156] S.M. Farkhani, A. Valizadeh, H. Karami, S. Mohammadi, N. Sohrabi, F. Badrzadeh,
(2008) 155174. Cell penetrating peptides: ecient vectors for delivery of nanoparticles, nano-
[131] S. Wohlfart, S. Gelperina, J.r. Kreuter, Transport of drugs across the blood brain carriers, therapeutic and diagnostic molecules, Peptides 57 (2014) 7894.
barrier by nanoparticles, J. Control. Release 161 (2012) 264273. [157] J. Piazza, T. Hoare, L. Molinaro, K. Terpstra, J. Bhandari, P.R. Selvaganapathy,
[132] H.L. Wong, N. Chattopadhyay, X.Y. Wu, R. Bendayan, Nanotechnology applica- B. Gupta, R.K. Mishra, Haloperidol-loaded intranasally administered lectin func-
tions for improved delivery of antiretroviral drugs to the brain, Adv. Drug Deliv. tionalized poly(ethylene glycol)-block-poly(D,L)-lactic-co-glycolic acid (PEG-
Rev. 62 (2010) 503517. PLGA) nanoparticles for the treatment of schizophrenia, Eur. J. Pharm. Biopharm.
[133] K. Shao, Y. Zhang, N. Ding, S. Huang, J. Wu, J. Li, C. Yang, Q. Leng, L. Ye, J. Lou, 87 (2014) 3039.
L. Zhu, C. Jiang, Functionalized nanoscale micelles with brain targeting ability and [158] C. Zhang, J. Chen, C. Feng, X. Shao, Q. Liu, Q. Zhang, Z. Pang, X. Jiang, Intranasal
intercellular microenvironment biosensitivity for anti-intracranial infection ap- nanoparticles of basic broblast growth factor for brain delivery to treat
plications, Adv. Healthc. Mater. 4 (2015) 291300. Alzheimer's disease, Int. J. Pharm. 461 (2014) 192202.
[134] H. Gupta, P. Paul, N. Kumar, S. Baxi, D.P. Das, One pot synthesis of water-dis- [159] Z. Wen, Z. Yan, K. Hu, Z. Pang, X. Cheng, L. Guo, Q. Zhang, X. Jiang, L. Fang,
persible dehydroascorbic acid coated Fe3O4 nanoparticles under atmospheric air: R. Lai, Odorranalectin-conjugated nanoparticles: preparation, brain delivery and
blood cell compatibility and enhanced magnetic resonance imaging, J. Colloid pharmacodynamic study on Parkinson's disease following intranasal administra-
Interface Sci. 430 (2014) 221228. tion, J. Control. Release 151 (2011) 131138.
[135] I. Singh, R. Swami, M.K. Jeengar, W. Khan, R. Sistla, p-Aminophenyl--D-man- [160] H. Xia, X. Gao, G. Gu, Z. Liu, Q. Hu, Y. Tu, Q. Song, L. Yao, Z. Pang, X. Jiang,
nopyranoside engineered lipidic nanoparticles for eective delivery of docetaxel J. Chen, H. Chen, Penetratin-functionalized PEG-PLA nanoparticles for brain drug
to brain, Chem. Phys. Lipids 188 (2015) 19. delivery, Int. J. Pharm. 436 (2012) 840850.
[136] H. Peng, D. Du, J. Zhang, Liposomes modied with p-aminophenyl-alpha-D- [161] S. Chaudhary, C.A. Smith, P. Del Pino, J.M. de la Fuente, M. Mullin, A. Hursthouse,
mannopyranoside: a promising delivery system in targeting the brain, Ther. Deliv. D. Stirling, C.C. Berry, Elucidating the function of penetratin and a static magnetic
4 (2013) 14751477. eld in cellular uptake of magnetic nanoparticles, Pharmaceuticals (Basel) 6
[137] D. Wu, J. Yang, W.M. Pardridge, Drug targeting of a peptide radiopharmaceutical (2013) 204222.
through the primate blood-brain barrier in vivo with a monoclonal antibody to the [162] T. Zong, L. Mei, H. Gao, K. Shi, J. Chen, Y. Wang, Q. Zhang, Y. Yang, Q. He,
human insulin receptor, J. Clin. Invest. 100 (1997) 18041812. Enhanced glioma targeting and penetration by dual-targeting liposome co-mod-
[138] R.J. Boado, Y. Zhang, Y. Zhang, W.M. Pardridge, Humanization of anti-human ied with T7 and TAT, J. Pharm. Sci. 103 (2014) 38913901.
insulin receptor antibody for drug targeting across the human blood-brain barrier, [163] J.S. Liu, J.H. Wang, J. Zhou, X.H. Tang, L. Xu, T. Shen, X.Y. Wu, Z. Hong,
Biotechnol. Bioeng. 96 (2007) 381391. Enhanced brain delivery of lamotrigine with Pluronic P123-based nanocarrier,
[139] T. Kang, M. Jiang, D. Jiang, X. Feng, J. Yao, Q. Song, H. Chen, X. Gao, J. Chen, Int. J. Nanomedicine 9 (2014) 39323935.
Enhancing glioblastoma-specic penetration by functionalization of nanoparticles [164] V. Saxena, M.D. Hussain, Formulation and in vitro evaluation of 17-allyamino-17-
with an iron-mimic peptide targeting transferrin/transferrin receptor complex, demethoxygeldanamycin (17-AAG) loaded polymeric mixed micelles for glio-
Mol. Pharm. 12 (2015) 29472961. blastoma multiforme, Colloids Surf. B: Biointerfaces 112 (2013) 350355.
[140] X. Wang, X. Huang, Z. Yang, D. Gallego-Perez, J. Ma, X. Zhao, J. Xie, I. Nakano, [165] S. Jain, V. Mishra, P. Singh, P.K. Dubey, D.K. Saraf, S.P. Vyas, RGD-anchored
L.J. Lee, Targeted delivery of tumor suppressor microRNA-1 by transferrin-con- magnetic liposomes for monocytes/neutrophils-mediated brain targeting, Int. J.
jugated lipopolyplex nanoparticles to patient-derived glioblastoma stem cells, Pharm. 261 (2003) 4355.
Curr. Pharm. Biotechnol. 15 (2014) 839846. [166] J. Qin, X. Yang, J. Mi, J. Wang, J. Hou, T. Shen, Y. Li, B. Wang, X. Li, W. Zhu,
[141] I. Cabezon, G. Manich, R. Martin-Venegas, A. Camins, C. Pelegri, J. Vilaplana, Enhanced antidepressant-like eects of the macromolecule trefoil factor 3 by
Tracking of gold nanoparticles coated with the 8D3 anti-transferrin receptor loading into negatively charged liposomes, Int. J. Nanomedicine 9 (2014)
antibody at the mouse blood-brain barrier, Mol. Pharm. 12 (2015) 41374145. 52475257.
[142] H. Bao, X. Jin, L. Li, F. Lv, T. Liu, OX26 modied hyperbranched polyglycerol- [167] N. Rapoport, Physical stimuli-responsive polymeric micelles for anti-cancer drug
conjugated poly(lactic-co-glycolic acid) nanoparticles: synthesis, characterization delivery, Prog. Polym. Sci. 32 (2007) 962990.
and evaluation of its brain delivery ability, J. Mater. Sci. Mater. Med. 23 (2012) [168] W. Xu, P. Ling, T. Zhang, Polymeric micelles, a promising drug delivery system to
18911901. enhance bioavailability of poorly water-soluble drugs, J. Drug Deliv. 2013 (2013)
[143] I.T. Papademetriou, C. Garnacho, E.H. Schuchman, S. Muro, In vivo performance 340315.
of polymer nanocarriers dually-targeted to epitopes of the same or dierent re- [169] L.Y. Qiu, Y.H. Bae, Polymer architecture and drug delivery, Pharm. Res. 23 (2006)
ceptors, Biomaterials 34 (2013) 34593466. 130.
[144] R.Q. Huang, W.L. Ke, Y.H. Qu, J.H. Zhu, Y.Y. Pei, C. Jiang, Characterization of [170] M.K. Pratten, J.B. Lloyd, G. Hrpel, H. Ringsdorf, Micelle-forming block copoly-
lactoferrin receptor in brain endothelial capillary cells and mouse brain, J. mers: pinocytosis by macrophages and interaction with model membranes,
Biomed. Sci. 14 (2007) 121128. Makromol. Chem. 186 (1985) 725733.
[145] D. Sun, A. Xue, B. Zhang, H. Lou, H. Shi, X. Zhang, Polysorbate 80-coated PLGA [171] P. Zhang, L. Hu, Q. Yin, Z. Zhang, L. Feng, Y. Li, Transferrin-conjugated poly-
nanoparticles improve the permeability of acetylpuerarin and enhance its brain- phosphoester hybrid micelle loading paclitaxel for brain-targeting delivery:
protective eects in rats, J. Pharm. Pharmacol. 67 (2015) 16501662. synthesis, preparation and in vivo evaluation, J. Control. Release 159 (2012)
[146] Z.Y. Yuan, Y.L. Hu, J.Q. Gao, Brain localization and neurotoxicity evaluation of 429434.
polysorbate-80 modied chitosan nanoparticles in rats, PLoS One 10 (2015) [172] Y. Miura, T. Takenaka, K. Toh, S. Wu, H. Nishihara, M.R. Kano, Y. Ino, T. Nomoto,

59
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

Y. Matsumoto, H. Koyama, H. Cabral, N. Nishiyama, K. Kataoka, Cyclic RGD- PEG based polymeric micelles for amphotericin B delivery targeted to the brain, J.
linked polymeric micelles for targeted delivery of platinum anticancer drugs to Control. Release 147 (2010) 118126.
glioblastoma through the blood-brain tumor barrier, ACS Nano 7 (2013) [198] Y. Qian, Y. Zha, B. Feng, Z. Pang, B. Zhang, X. Sun, J. Ren, C. Zhang, X. Shao,
85838592. Q. Zhang, X. Jiang, PEGylated poly(2-(dimethylamino) ethyl methacrylate)/DNA
[173] B.J. Zhao, X.Y. Ke, Y. Huang, X.M. Chen, X. Zhao, B.X. Zhao, W.L. Lu, J.N. Lou, polyplex micelles decorated with phage-displayed TGN peptide for brain-targeted
X. Zhang, Q. Zhang, The antiangiogenic ecacy of NGR-modied PEG-DSPE mi- gene delivery, Biomaterials 34 (2013) 21172129.
celles containing paclitaxel (NGR-M-PTX) for the treatment of glioma in rats, J. [199] C. Mu, N. Dave, J. Hu, P. Desai, G. Pauletti, S. Bai, J. Hao, Solubilization of
Drug Target. 19 (2011) 382390. urbiprofen into aptamer-modied PEG-PLA micelles for targeted delivery to
[174] V. Erfani-Moghadam, A. Nomani, M. Zamani, Y. Yazdani, F. Naja, brain-derived endothelial cells in vitro, J. Microencapsul. 30 (2013) 701708.
M. Sadeghizadeh, A novel diblock of copolymer of (monomethoxy poly [ethylene [200] N. Yi, B. Oh, H.A. Kim, M. Lee, Combined delivery of BCNU and VEGF siRNA using
glycol]-oleate) with a small hydrophobic fraction to make stable micelles/poly- amphiphilic peptides for glioblastoma, J. Drug Target. 22 (2014) 156164.
mersomes for curcumin delivery to cancer cells, Int. J. Nanomedicine 9 (2014) [201] X. Qian, L. Long, Z. Shi, C. Liu, M. Qiu, J. Sheng, P. Pu, X. Yuan, Y. Ren, C. Kang,
55415554. Star-branched amphiphilic PLA-b-PDMAEMA copolymers for co-delivery of miR-
[175] E.F. Craparo, M.C. Ognibene, M.P. Casaletto, G. Pitarresi, G. Teresi, G. Giammona, 21 inhibitor and doxorubicin to treat glioma, Biomaterials 35 (2014) 23222335.
Biocompatible polymeric micelles with polysorbate 80 for use in brain targeting, [202] T. Kanazawa, K. Morisaki, S. Suzuki, Y. Takashima, Prolongation of life in rats
Nanotechnology 19 (2008) 485603. with malignant glioma by intranasal siRNA/drug codelivery to the brain with cell-
[176] K. Kataoka, T. Matsumoto, M. Yokoyama, T. Okano, Y. Sakurai, S. Fukushima, penetrating peptide-modied micelles, Mol. Pharm. 11 (2014) 14711478.
K. Okamoto, G.S. Kwon, Doxorubicin-loaded poly(ethylene glycol)-poly(beta- [203] H. Taki, T. Kanazawa, F. Akiyama, Y. Takashima, H. Okada, Intranasal delivery of
benzyl-L-aspartate) copolymer micelles: their pharmaceutical characteristics and camptothecin-loaded tat-modied nanomicells for treatment of intracranial brain
biological signicance, J. Control. Release 64 (2000) 143153. tumors, Pharmaceuticals (Basel) 5 (2012) 10921102.
[177] S. Zheng, Y. Xie, Y. Li, L. Li, N. Tian, W. Zhu, G. Yan, C. Wu, H. Hu, Development [204] E.J. Chung, Y. Cheng, R. Morshed, K. Nord, Y. Han, M.L. Wegscheid, B. Aunger,
of high drug-loading nanomicelles targeting steroids to the brain, Int. J. D.A. Wainwright, M.S. Lesniak, M.V. Tirrell, Fibrin-binding, peptide amphiphile
Nanomedicine 9 (2014) 5566. micelles for targeting glioblastoma, Biomaterials 35 (2014) 12491256.
[178] O. Linsell, P.W. Brownjohn, H. Neho, K. Greish, J.C. Ashton, Eect of styrene [205] J. Niu, A. Wang, Z. Ke, Z. Zheng, Glucose transporter and folic acid receptor-
maleic acid WIN55,212-2 micelles on neuropathic pain in a rat model, J. Drug mediated Pluronic P105 polymeric micelles loaded with doxorubicin for brain
Target. 23 (2015) 353359. tumor treating, J. Drug Target. 22 (2014) 712723.
[179] E. Fournier, M.H. Dufresne, D.C. Smith, M. Ranger, J.C. Leroux, A novel one-step [206] G. Leyva-Gomez, M.E. Gonzalez-Trujano, E. Lopez-Ruiz, P.O. Couraud,
drug-loading procedure for water-soluble amphiphilic nanocarriers, Pharm. Res. B. Wekslerg, I. Romero, F. Miller, F. Delie, E. Allemann, D. Quintanar-Guerrero,
21 (2004) 962968. Nanoparticle formulation improves the anticonvulsant eect of clonazepam on the
[180] N. Nishiyama, S. Okazaki, H. Cabral, M. Miyamoto, Y. Kato, Y. Sugiyama, pentylenetetrazole-induced seizures: behavior and electroencephalogram, J.
K. Nishio, Y. Matsumura, K. Kataoka, Novel cisplatin-incorporated polymeric Pharm. Sci. 103 (2014) 25092519.
micelles can eradicate solid tumors in mice, Cancer Res. 63 (2003) 89778983. [207] J. Liu, Y. He, J. Zhang, J. Li, X. Yu, Z. Cao, F. Meng, Y. Zhao, X. Wu, T. Shen,
[181] J. Li, H. Yang, Y. Zhang, X. Jiang, Y. Guo, S. An, H. Ma, X. He, C. Jiang, Choline Z. Hong, Functionalized nanocarrier combined seizure-specic vector with P-
derivate-modied doxorubicin loaded micelle for glioma therapy, ACS Appl. glycoprotein modulation property for antiepileptic drug delivery, Biomaterials 74
Mater. Interfaces 7 (2015) 2158921601. (2016) 6476.
[182] Z. Wang, X. Hu, J. Yue, X. Jing, Experimental study on biodegradable polymer- [208] Q. Zhou, K. Mu, L. Jiang, H. Xie, W. Liu, Z. Li, H. Qi, S. Liang, H. Xu, Y. Zhu,
paclitaxel conjugate micelles for chemotherapy of C6 glioma, J. Control. Release W. Zhu, X. Yang, Glioma-targeting micelles for optical/magnetic resonance dual-
152 (2011) e41e42. mode imaging, Int. J. Nanomedicine 10 (2015) 18051818.
[183] G.H. Gao, M.J. Park, Y. Li, G.H. Im, J.H. Kim, H.N. Kim, J.W. Lee, P. Jeon, [209] J.H. Park, J. Han, M. Lee, Thymidine kinase gene delivery using curcumin loaded
O.Y. Bang, J.H. Lee, D.S. Lee, The use of pH-sensitive positively charged polymeric peptide micelles as a combination therapy for glioblastoma, Pharm. Res. 32 (2015)
micelles for protein delivery, Biomaterials 33 (2012) 91579164. 528537.
[184] C.Q. Mao, J.Z. Du, T.M. Sun, Y.D. Yao, P.Z. Zhang, E.W. Song, J. Wang, A bio- [210] H.M. Rashed, R.N. Shamma, E.B. Basalious, Contribution of both olfactory and
degradable amphiphilic and cationic triblock copolymer for the delivery of siRNA systemic pathways for brain targeting of nimodipine-loaded lipo-pluronics mi-
targeting the acid ceramidase gene for cancer therapy, Biomaterials 32 (2011) celles: in vitro characterization and in vivo biodistribution study after intranasal
31243133. and intravenous delivery, Drug Deliv. 24 (2017) 181187.
[185] D.A. Chiappetta, C. Hocht, J.A. Opezzo, A. Sosnik, Intranasal administration of [211] X. Chen, L. Tai, J. Gao, J. Qian, M. Zhang, B. Li, C. Xie, L. Lu, W. Lu, W. Lu, A
antiretroviral-loaded micelles for anatomical targeting to the brain in HIV, stapled peptide antagonist of MDM2 carried by polymeric micelles sensitizes
Nanomedicine (London) 8 (2013) 223237. glioblastoma to temozolomide treatment through p53 activation, J. Control.
[186] E.B. Basalious, R.N. Shamma, Novel self-assembled nano-tubular mixed micelles of Release 218 (2015) 2935.
Pluronics P123, Pluronic F127 and phosphatidylcholine for oral delivery of ni- [212] A.K. Mohanty, G.P. Mohanta, Micelle-assisted combination therapies for eective
modipine: in vitro characterization, ex vivo transport and in vivo pharmacokinetic glioblastoma treatment: an in-vitro assessment, Anti-Cancer Drugs 26 (2015)
studies, Int. J. Pharm. 493 (2015) 347356. 312322.
[187] Y.I. Jeong, S.H. Kim, T.Y. Jung, I.Y. Kim, S.S. Kang, Y.H. Jin, H.H. Ryu, H.S. Sun, [213] M. Clemens-Hemmelmann, C. Kuner, V. Metz, L. Kircher, U. Schmitt, C. Hiemke,
S. Jin, K.K. Kim, K.Y. Ahn, S. Jung, Polyion complex micelles composed of all-trans R. Postina, R. Zentel, Amphiphilic copolymers shuttle drugs across the blood brain
retinoic acid and poly (ethylene glycol)-grafted-chitosan, J. Pharm. Sci. 95 (2006) barrier, Macromol. Biosci. 16 (2016) 655665.
23482360. [214] E. Choi, J. Han, X. Tan, J. Oh, D. Lee, T. Rhim, M. Lee, Combined delivery of
[188] R. Jain, S. Nabar, P. Dandekar, V. Patravale, Micellar nanocarriers: potential nose- temozolomide and the thymidine kinase gene for treatment of glioblastoma, J.
to-brain delivery of zolmitriptan as novel migraine therapy, Pharm. Res. 27 (2010) Drug Target. 24 (2016) 17.
655664. [215] X. Tang, Y. Liang, Y. Zhu, C. Xie, A. Yao, L. Chen, Q. Jiang, T. Liu, X. Wang,
[189] P. Zhang, L. Hu, Y. Wang, J. Wang, L. Feng, Y. Li, Poly(-caprolactone)-block-poly Y. Qian, J. Wei, W. Ni, J. Dai, Z. Jiang, W. Hou, Anti-transferrin receptor-modied
(ethyl ethylene phosphate) micelles for brain-targeting drug delivery: in vitro and amphotericin B-loaded PLA-PEG nanoparticles cure Candidal meningitis and re-
in vivo valuation, Pharm. Res. 27 (2010) 26572669. duce drug toxicity, Int. J. Nanomedicine 10 (2015) 62276241.
[190] W.H. Ren, J. Chang, C.H. Yan, X.M. Qian, L.X. Long, B. He, X.B. Yuan, C.S. Kang, [216] G. Wang, J.J. Wang, X.J. Tang, L. Du, F. Li, In vitro and in vivo evaluation of
D. Betbeder, J. Sheng, P.Y. Pu, Development of transferrin functionalized poly functionalized chitosan-Pluronic micelles loaded with myricetin on glioblastoma
(ethylene glycol)/poly(lactic acid) amphiphilic block copolymeric micelles as a cancer, Nanomedicine 12 (2016) 12631278.
potential delivery system targeting brain glioma, J. Mater. Sci. Mater. Med. 21 [217] G. Wang, J.J. Wang, X.L. Chen, L. Du, F. Li, Quercetin-loaded freeze-dried nano-
(2010) 26732681. micelles: improving absorption and anti-glioma eciency in vitro and in vivo, J.
[191] C. Zhan, B. Gu, C. Xie, J. Li, Y. Liu, W. Lu, Cyclic RGD conjugated poly(ethylene Control. Release 235 (2016) 276290.
glycol)-co-poly(lactic acid) micelle enhances paclitaxel anti-glioblastoma eect, J. [218] Y. Zhu, J. Zhang, F. Meng, C. Deng, R. Cheng, J. Feijen, Z. Zhong, cRGD-func-
Control. Release 143 (2010) 136142. tionalized reduction-sensitive shell-sheddable biodegradable micelles mediate
[192] M.G. Sarpietro, G. Pitarresi, S. Ottimo, M.C. Giurida, M.C. Ognibene, C. Fiorica, enhanced doxorubicin delivery to human glioma xenografts in vivo, J. Control.
G. Giammona, F. Castelli, Interaction between drug loaded polyaspartamide- Release 233 (2016) 2938.
polylactide-polysorbate based micelles and cell membrane models: a calorimetric [219] Y. Huang, W. Liu, F. Gao, X. Fang, Y. Chen, c(RGDyK)-decorated Pluronic micelles
study, Mol. Pharm. 8 (2011) 642650. for enhanced doxorubicin and paclitaxel delivery to brain glioma, Int. J.
[193] J. Shen, C. Zhan, C. Xie, Q. Meng, B. Gu, C. Li, Y. Zhang, W. Lu, Poly(ethylene Nanomedicine 11 (2016) 16291641.
glycol)-block-poly(D,L-lactide acid) micelles anchored with angiopep-2 for brain- [220] Y. Bi, L. Liu, Y. Lu, T. Sun, C. Shen, X. Chen, Q. Chen, S. An, X. He, C. Ruan, Y. Wu,
targeting delivery, J. Drug Target. 19 (2011) 197203. Y. Zhang, Q. Guo, Z. Zheng, Y. Liu, M. Lou, S. Zhao, C. Jiang, T7 peptide-func-
[194] T. Kanazawa, H. Taki, K. Tanaka, Y. Takashima, H. Okada, Cell-penetrating pep- tionalized PEG-PLGA micelles loading with carmustine for targeting therapy of
tide-modied block copolymer micelles promote direct brain delivery via in- glioma, ACS Appl. Mater. Interfaces 8 (2016) 2746527473.
tranasal administration, Pharm. Res. 28 (2011) 21302139. [221] L. Sun, D.Y. Joh, A. Al-Zaki, M. Stangl, S. Murty, J.J. Davis, B.C. Baumann,
[195] Y.T. Xie, Y.Z. Du, H. Yuan, F.Q. Hu, Brain-targeting study of stearic acid-grafted M. Alonso-Basanta, G.D. Kaol, A. Tsourkas, J.F. Dorsey, Theranostic application of
chitosan micelle drug-delivery system, Int. J. Nanomedicine 7 (2012) 32353244. mixed gold and superparamagnetic iron oxide nanoparticle micelles in glio-
[196] K. Shao, J. Wu, Z. Chen, S. Huang, J. Li, L. Ye, J. Lou, L. Zhu, C. Jiang, A brain- blastoma multiforme, J. Biomed. Nanotechnol. 12 (2016) 347356.
vectored angiopep-2 based polymeric micelles for the treatment of intracranial [222] C. Sarisozen, S. Dhokai, E.G. Tsikudo, E. Luther, I.M. Rachman, V.P. Torchilin,
fungal infection, Biomaterials 33 (2012) 68986907. Nanomedicine based curcumin and doxorubicin combination treatment of glio-
[197] K. Shao, R. Huang, J. Li, L. Han, L. Ye, J. Lou, C. Jiang, Angiopep-2 modied PE- blastoma with scFv-targeted micelles: in vitro evaluation on 2D and 3D tumor

60
R.S. Elezaby et al. Journal of Controlled Release 261 (2017) 4361

models, Eur. J. Pharm. Biopharm. 108 (2016) 5467. Surf. B: Biointerfaces 118 (2014) 298305.
[223] S. Zheng, X. Gao, X. Liu, T. Yu, T. Zheng, Y. Wang, C. You, Biodegradable micelles [249] J. Li, Z. Zha, Z. Ge, Thermo-responsive polyplex micelles with PEG shells and
enhance the antiglioma activity of curcumin in vitro and in vivo, Int. J. PNIPAM layer to protect DNA cores for systemic gene therapy, Methods Mol. Biol.
Nanomedicine 11 (2016) 27212736. 1445 (2016) 269276.
[224] S.A. Nour, N.S. Abdelmalak, M.J. Naguib, H.M. Rashed, A.B. Ibrahim, Intranasal [250] Z. Quan, K. Zhu, K.D. Knudsen, B. Nystrom, R. Lund, Tailoring the amphiphilicity
brain-targeted clonazepam polymeric micelles for immediate control of status and self-assembly of thermosensitive polymers: end-capped PEG-PNIPAAM block
epilepticus: in vitro optimization, ex vivo determination of cytotoxicity, in vivo copolymers, Soft Matter 9 (2013) 1076810778.
biodistribution and pharmacodynamics studies, Drug Deliv. 23 (2016) 36813695. [251] R.S. Lee, C.H. Lin, I.A. Aljuali, K.Y. Hu, J.Y. Fang, Passive targeting of thermo-
[225] X.J. Tang, K.-M. Huang, H. Gui, J.-J. Wang, J.-T. Lu, L.-J. Dai, L. Zhang, G. Wang, sensitive diblock copolymer micelles to the lungs: synthesis and characterization
Pluronic-based micelle encapsulation potentiates myricetin-induced cytotoxicity of poly(N-isopropylacrylamide)-block-poly(-caprolactone), J. Nanobiotechnology
in human glioblastoma cells, Int. J. Nanomedicine 11 (2016) 49915002. 13 (2015) (Article ID 42).
[226] J. Ding, Y. Sun, J. Li, H. Wang, S. Mao, Enhanced blood brain barrier transport of [252] C.L. Peng, Y.I. Chen, H.J. Liu, P.C. Lee, T.Y. Luo, M.J. Shieh, A novel temperature-
vinpocetine by oral delivery of mixed micelles in combination with a message responsive micelle for enhancing combination therapy, Int. J. Nanomedicine 11
guider, J. Drug Target. 25 (2017) 133. (2016) 33573369.
[227] A.N. Lukyanov, V.P. Torchilin, Micelles from lipid derivatives of water-soluble [253] N. Yu, G. Li, Y. Gao, H. Jiang, Q. Tao, Thermo-sensitive complex micelles from
polymers as delivery systems for poorly soluble drugs, Adv. Drug Deliv. Rev. 56 sodium alginate-graft-poly(N-isopropylacrylamide) for drug release, Int. J. Biol.
(2004) 12731289. Macromol. 86 (2016) 296301.
[228] M. Yu, N.S. Kozlov, E.V. Melik-Nubarov, A.V. Batrakov, Relationship between [254] J.Y. Teo, W. Chin, X. Ke, S. Gao, S. Liu, W. Cheng, J.L. Hedrick, Y.Y. Yang, pH and
Pluronic block copolymer structure, critical micellization concentration and par- redox dual-responsive biodegradable polymeric micelles with high drug loading
titioning coecients of low molecular mass solutes, Macromolecules 33 (2000) for eective anticancer drug delivery, Nanomedicine 13 (2017) 431442.
33053313. [255] H. Wei, R.-X. Zhuo, X.-Z. Zhang, Design and development of polymeric micelles
[229] Y. Chan, T. Wong, F. Byrne, M. Kavallaris, V. Bulmus, Acid-labile core cross-linked with cleavable links for intracellular drug delivery, Prog. Polym. Sci. 38 (2013)
micelles for pH-triggered release of antitumor drugs, Biomacromolecules 9 (2008) 503535.
18261836. [256] G.H. Gao, J.W. Lee, M.K. Nguyen, G.H. Im, J. Yang, H. Heo, P. Jeon, T.G. Park,
[230] X.B. Xiong, A. Mahmud, H. Uludag, A. Lavasanifar, Multifunctional polymeric J.H. Lee, D.S. Lee, pH-responsive polymeric micelle based on PEG-poly(beta-
micelles for enhanced intracellular delivery of doxorubicin to metastatic cancer amino ester)/(amido amine) as intelligent vehicle for magnetic resonance imaging
cells, Pharm. Res. 25 (2008) 25552566. in detection of cerebral ischemic area, J. Control. Release 155 (2011) 1117.
[231] W. Huang, C. Shi, Y. Shao, K.S. Lam, J. Luo, The core-inversible micelles for hy- [257] K. Miller, S. Dixit, A.L. Bredlau, A. Moore, E. McKinnon, A.M. Broome, Delivery of
drophilic drug delivery, Chem. Commun. (Camb.) 49 (2013) 66746676. a drug cache to glioma cells overexpressing platelet-derived growth factor re-
[232] R. Dave, R. Patel, J. Patel, H. Chauhan, Eect of cryoprotectant on lyophlisation of ceptor using lipid nanocarriers, Nanomedicine (London) 11 (2016) 581595.
doxorubicin HCl loaded chitosan nanoparticles, Int. J. Pharm. Life Sci. 3 (2012) [258] J. Cao, T. Su, L. Zhang, R. Liu, G. Wang, B. He, Z. Gu, Polymeric micelles with
17691772. citraconic amide as pH-sensitive bond in backbone for anticancer drug delivery,
[233] H. Cesur, I. Rubinstein, A. Pai, H. nyksel, Self-associated Indisulam in Int. J. Pharm. 471 (2014) 2836.
phosphoIipid-based nanomicelles: a potential nanomedicine for cancer, [259] Y. Bi, H. Wei, Q. Hu, W. Xu, Y. Gong, L. Yu, Wormlike micelles with photo-
Nanomedicine 5 (2009) 178183. responsive viscoelastic behavior formed by surface active ionic liquid/azobenzene
[234] M.A. Moretton, D.A. Chiappetta, A. Sosnik, Cryoprotection-lyophilization and derivative mixed solution, Langmuir 31 (2015) 37893798.
physical stabilization of rifampicin-loaded ower-like polymeric micelles, J. R. [260] A.J. Harnoy, G. Slor, E. Tirosh, R.J. Amir, The eect of photoisomerization on the
Soc. Interface 9 (2012) 487502. enzymatic hydrolysis of polymeric micelles bearing photo-responsive azobenzene
[235] C. Oerlemans, W. Bult, M. Bos, G. Storm, J.F. Nijsen, W.E. Hennink, Polymeric groups at their cores, Org. Biomol. Chem. 14 (2016) 58135819.
micelles in anticancer therapy: targeting, imaging and triggered release, Pharm. [261] D. Yang, J. Zhao, A light-responsive organouid based on reverse worm-like mi-
Res. 27 (2010) 25692589. celles formed from an equi-charged, mixed, anionic gemini surfactant with an
[236] R.A. Morshed, Y. Cheng, B. Aunger, M.L. Wegscheid, M.S. Lesniak, The potential azobenzene spacer and a cationic conventional surfactant, Soft Matter 12 (2016)
of polymeric micelles in the context of glioblastoma therapy, Front Pharmacol. 4 40444051.
(2013) (Article ID 157). [262] H. Cabral, M. Nakanishi, M. Kumagai, W.D. Jang, N. Nishiyama, K. Kataoka, A
[237] M.W. Amjad, P. Kesharwani, M.C.I.M. Amin, A.K. Iyer, Recent advances in the photo-activated targeting chemotherapy using glutathione sensitive camptothecin-
design, development, and targeting mechanisms of polymeric micelles for delivery loaded polymeric micelles, Pharm. Res. 26 (2009) 8292.
of siRNA in cancer therapy, Prog. Polym. Sci. 64 (2017) 154181. [263] M. Huang, H. Li, W. Ke, J. Li, C. Zhao, Z. Ge, Finely tuned thermo-responsive block
[238] S. Biswas, P. Kumari, P.M. Lakhani, B. Ghosh, Recent advances in polymeric mi- copolymer micelles for photothermal eect-triggered ecient cellular inter-
celles for anti-cancer drug delivery, Eur. J. Pharm. Sci. 83 (2016) 184202. nalization, Macromol. Biosci. 16 (2016) 12651272.
[239] Y. Li, K. Xiao, W. Zhu, W. Deng, K.S. Lam, Stimuli-responsive cross-linked micelles [264] C. Wu, J. Yang, X. Xu, C. Gao, S. L, M. Liu, Redox-responsive core-cross-linked
for on-demand drug delivery against cancers, Adv. Drug Deliv. Rev. 66 (2014) mPEGylated starch micelles as nanocarriers for intracellular anticancer drug re-
5873. lease, Eur. Polym. J. 83 (2016) 230243.
[240] A. Gothwal, I. Khan, U. Gupta, Polymeric micelles: recent advancements in the [265] H. Gao, J. Qian, S. Cao, Z. Yang, Z. Pang, S. Pan, L. Fan, Z. Xi, X. Jiang, Q. Zhang,
delivery of anticancer drugs, Pharm. Res. 33 (2016) 1839. Precise glioma targeting of and penetration by aptamer and peptide dual-func-
[241] H. Cabral, K. Kataoka, Progress of drug-loaded polymeric micelles into clinical tioned nanoparticles, Biomaterials 33 (2012) 51155123.
studies, J. Control. Release 190 (2014) 465476. [266] H. Gao, Z. Yang, S. Zhang, Z. Pang, X. Jiang, Internalization and subcellular fate of
[242] P.V. Devarajan, S. Jain, Polymeric micelles in targeted drug delivery, in: aptamer and peptide dual-functioned nanoparticles, J. Drug Target. 22 (2014)
P.V. Devarajan, V. Padma, S. Jain, Sanyog (Eds.), Targeted drug delivery: concepts 450459.
and design, E- publishing Inc, 2015, pp. 501541. [267] H. Gao, Z. Yang, S. Zhang, Z. Pang, Q. Liu, X. Jiang, Study and evaluation of
[243] H.S. Oberoi, F.C. Laquer, L.A. Marky, A.V. Kabanov, T.K. Bronich, Core cross- mechanisms of dual targeting drug delivery system with tumor microenvironment
linked block ionomer micelles as pH-responsive carriers for cis-diamminedi- assays compared with normal assays, Acta Biomater. 10 (2014) 858867.
chloroplatinum(II), J. Control. Release 153 (2011) 6472. [268] H. Ma, Z. Gao, P. Yu, S. Shen, Y. Liu, B. Xu, A dual functional uorescent probe for
[244] T. Li, F. Ning, J. Xie, D. Chen, M. Jiang, Preparation and morphologies of shell glioma imaging mediated by blood-brain barrier penetration and glioma cell tar-
cross-linked micelles based on commercial poly (styrene-block-ethylene-co-bu- geting, Biochem. Biophys. Res. Commun. 449 (2014) 4448.
tene-block-styrene), Polym. J. 34 (2002) 529533. [269] L. Shan, Indotricarbocyanine-loaded AS1411 DNA aptamer- and TGN peptide-
[245] Y. Zhang, C. Wang, Y. Huang, H. Yan, K. Liu, Core-crosslinked polymeric micelles modied poly(ethylene glycol)-poly(-caprolactone) nanoparticles, Molecular
with high doxorubicin loading capacity and intracellular pH- and redox-triggered Imaging and Contrast Agent Database (MICAD), 2012, pp. 115.
payload release, Eur. Polym. J. 68 (2015) 104114. [270] C. Zhang, X. Zheng, X. Wan, X. Shao, Q. Liu, Z. Zhang, Q. Zhang, The potential use
[246] M. Talelli, M. Barz, C.J.F. Rijcken, F. Kiessling, W.E. Hennink, T. Lammers, Core- of H102 peptide-loaded dual-functional nanoparticles in the treatment of
crosslinked polymeric micelles: principles, preparation, biomedical applications Alzheimer's disease, J. Control. Release 192 (2014) 317324.
and clinical translation, Nano Today 10 (2015) 93117. [271] K. Hu, Y. Shi, W. Jiang, J. Han, S. Huang, X. Jiang, Lactoferrin conjugated PEG-
[247] S.S. Desale, S.M. Cohen, Y. Zhao, A.V. Kabanov, T.K. Bronich, Biodegradable hy- PLGA nanoparticles for brain delivery: preparation, characterization and ecacy
brid polymer micelles for combination drug therapy in ovarian cancer, J. Control. in Parkinson's disease, Int. J. Pharm. 415 (2011) 273283.
Release 171 (2013) 339348. [272] R. Huang, W. Ke, Y. Liu, D. Wu, L. Feng, C. Jiang, Y. Pei, Gene therapy using
[248] Y. Wu, Q. Lai, S. Lai, J. Wu, W. Wang, Z. Yuan, Facile fabrication of core cross- lactoferrin-modied nanoparticles in a rotenone-induced chronic Parkinson
linked micelles by RAFT polymerization and enzyme-mediated reaction, Colloids model, J. Neurol. Sci. 290 (2010) 123130.

61

S-ar putea să vă placă și