• No results found

University of Groningen G23 peptide-mediated delivery of biodegradable nanocarriers across an in vitro blood-brain barrier model de Jong, Edwin

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen G23 peptide-mediated delivery of biodegradable nanocarriers across an in vitro blood-brain barrier model de Jong, Edwin"

Copied!
15
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

G23 peptide-mediated delivery of biodegradable nanocarriers across an in vitro blood-brain

barrier model

de Jong, Edwin

DOI:

10.33612/diss.132284892

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

de Jong, E. (2020). G23 peptide-mediated delivery of biodegradable nanocarriers across an in vitro

blood-brain barrier model. University of Groningen. https://doi.org/10.33612/diss.132284892

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

CHAPTER 1

General introduction and scope of the

thesis

Edwin de Jong

University of Groningen, University Medical Center Groningen, Department of Biomedical

Engineering, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands

(3)

THE BLOOD-BRAIN BARRIER

Brain homeostasis is largely dependent on proper functioning of the blood-brain barrier

(BBB), which is a cellular barrier that separates the circulating blood from the interstitial

fluid in the brain. The BBB is formed by a polarized layer of brain capillary endothelial

cells and supporting cell types, such as astrocytes and pericytes, that are located in close

proximity to the abluminal surface of brain capillaries [1, 2]. Neurons reside on average

about 10 to 30 µm from the nearest brain capillary [3−5]. Adjacent endothelial cells are

interconnected by transmembrane tight junction (TJ) proteins that are linked through

cytosolic scaffolding TJ proteins to the actin cytoskeleton and separate the plasma

membrane into a luminal (apical) and abluminal (basolateral) domain [6−8]. The TJ

complexes limit paracellular diffusion of biomolecules into the brain (Figure 1). Gaseous

lipophilic molecules, such as oxygen and carbon dioxide, are able to cross the plasma

membrane of brain endothelial cells by passive diffusion (Figure 1). The BBB actively

regulates the transcellular transport of biomolecules that can’t freely diffuse across the

endothelium in order to provide neurons with essential nutrients and metabolites while

preventing the passage of dangerous toxins and pathogens.

Figure 1. Transport across the blood-brain barrier. Gaseous lipophilic molecules are able to cross

the plasma membrane by passive diffusion. Tight junction complexes between adjacent endothelial

cells limit paracellular diffusion of biomolecules into the brain. Transporters and receptors facilitate

the translocation of specific biomolecules across the endothelium in order to provide the brain with

essential nutrients and metabolites.

The brain endothelium expresses transporters and receptors that facilitate the

translocation of specific biomolecules across the BBB. Carrier-mediated transport enables

BBB translocation of small hydrophilic molecules via membrane-embedded transporters

(4)

(Figure 1). The bidirectional glucose transporter GLUT1, which is present in both the

luminal and abluminal membrane domain of endothelial cells, mediates the delivery of

glucose to the brain by facilitated diffusion [7, 9, 10]. Other facilitative transporters that

enable transport of amino acids and lactate across both membrane domains of the brain

endothelium are the large neutral amino acid transporter LAT1 and the monocarboxylate

transporter MCT1, respectively [7, 10]. In contrast to facilitative transporters that mediate

translocation of molecules across the BBB in an energy-independent manner,

receptor-mediated transcytosis is an energy-dependent vesicular transport process that allows for

the delivery of relatively large molecules across the BBB (Figure 1). Upon interaction of a

ligand with the relevant membrane receptor, such as the transferrin receptor [11−13], the

low density lipoprotein (LDL) receptor [14, 15] or the insulin receptor [16−18], internalization

occurs via endocytosis. The uptake of iron-loaded transferrin is primarily facilitated by

clathrin-mediated endocytosis, a process which involves the formation of a clathrin-coated

pit at the plasma membrane that pinches off into the cytosol, forming clathrin-coated

vesicles. Caveolae-mediated endocytosis is the major route for internalization of LDL by

brain endothelial cells. Caveolae are invaginated membrane microdomains with a

flask-shaped morphology that bud from the plasma membrane and form caveolar vesicles upon

ligand-receptor interaction. After endocytosis of the ligands, subsequent intracellular

vesicular transport and exocytosis at the opposite cell surface of the brain endothelium

results in the transcellular transport of the ligands, e.g. nutrients and metabolites.

DRUG DELIVERY INTO THE BRAIN

Brain diseases have an enormous impact on the daily life of patients and pose a large

economic and social burden on society. The number of people diagnosed with a brain

disease, such as Alzheimer’s [19], continues to increase due to the ageing population.

The treatment of brain diseases is greatly hampered by the presence of the BBB, which

actively prevents the delivery of therapeutics from the blood into the brain. Temporary

disruption of TJ integrity upon intra-arterial infusion of a hyperosmolar solution, e.g.

mannitol, enables paracellular diffusion of chemotherapeutics through the BBB [20, 21].

However, besides the invasiveness of drug administration via the carotid or vertebral

artery, TJ disruption by mannitol is associated with the occurrence of seizures [22, 23].

Other highly invasive delivery techniques enable direct administration of a drug into

the brain via an injection or implant, but come with disadvantages such as the limited

volume of drug distribution, the potential increase of intracranial pressure and the risk

(5)

of infection [24, 25]. Intravenous administration of drugs that cross brain endothelial

cells without compromising BBB integrity is considered a less invasive alternative to treat

brain diseases.

Most small-molecule drugs that are currently available for the treatment of brain-related

diseases cross the endothelium by passive diffusion. In addition to their low molecular

weight, the drug molecules must be moderately lipophilic and have a low

hydrogen-bonding potential to diffuse transcellularly [26]. However, the presence of ATP-binding

cassette (ABC) efflux transporters at the plasma membrane of brain endothelial cells

may prevent the transcellular transport of lipophilic molecules by exporting them

back into the circulation, and thereby reducing their delivery to the brain [27, 28]. The

polarized brain endothelium is characterized by an asymmetric distribution of efflux

transporters, including p-glycoprotein, between the luminal and abluminal membrane

domains [7]. P-glycoprotein, which is present in the luminal membrane domain, limits

the entry of various substrates, such as opiates, antipsychotics, antidepressants and

chemotherapeutics, into the brain [27, 28]. Although combined treatment with drugs and

inhibitors of ABC efflux transporters may stimulate drug transport across the BBB [29, 30],

the use of efflux pump inhibitors also increases the risk of concomitant entry of other,

potentially toxic, compounds from the blood into the brain.

Several hydrophilic small-molecule drugs, which are structurally similar to endogenous

substrates of the facilitative transporter LAT1 [31], are delivered across the brain

endothelium via LAT1-mediated transport. Alternatively, an endogenous substrate, e.g.

glucose, can be conjugated to the drug molecule in order to facilitate its BBB translocation

via a facilitative transporter, e.g. the GLUT1 transporter [32]. Also, the transport of certain

nanocarriers across the BBB is enhanced by conjugation of GLUT1-substrates [33−35].

However, transport via facilitative transporters is thought to be primarily suited for the

delivery of single hydrophilic drug molecules [36].

The treatment of brain diseases with therapeutic macromolecules, such as nucleic acids and

proteins, necessitates the development of delivery platforms to deliver these drugs into the

brain [37]. Nanocarriers are able to transport drugs across the BBB via the so-called

Trojan-horse mechanism whereby the nanoparticles cross the brain endothelium as passengers

on a vesicular transport route. Although adsorptive-mediated transcytosis enables

transendothelial transport of nanoparticles, e.g. cationic serum albumin-conjugated

polymersomes [38], it is considered to be a non-specific transport process. The conjugation

(6)

of ligands to nanoparticles that specifically target native receptors on the brain endothelium

and promote receptor-mediated transcytosis represents a more promising strategy for drug

delivery into the brain [39, 40]. Decoration of nanoparticles with moieties that target the

transferrin receptor, such as OX26 antibodies or B6 peptides, has been demonstrated to

enhance drug delivery across the BBB [41, 42]. Likewise, HIRMAb-functionalized liposomes,

which are targeted towards the insulin receptor, were shown to induce transendothelial

transport of macromolecular cargo into the brain [43]. Besides the transcytosis of targeted

nanoparticles across the endothelium, receptor-mediated transport may result in recycling

or degradation in lysosomes. The fate of a targeting ligand and its associated cargo is

affected by its capacity to dissociate from the receptor after internalization [44−46].

POLYMERSOMES FOR DRUG DELIVERY

Polymersomes are spherical bilayer structures composed of amphiphilic block copolymers

that protect the therapeutic cargo from degradation during systemic circulation. These

polymeric vesicles can accommodate hydrophilic and hydrophobic molecules within

their aqueous core and polymer bilayer (Figure 2), respectively. Decoration with targeting

ligands facilitates receptor-targeted delivery of drugs, e.g. peptides, proteins, siRNA and

doxorubicin, into the brain [41, 47−56]. Polymersomes are of great interest for biomedical

applications due to the high chemical versatility of block copolymers that allows for tuning

of nanoparticle properties, such as membrane thickness and stimulus-responsiveness [57].

The morphology of polymersomes closely resembles that of liposomes, which are bilayer

structures of amphiphilic phospholipids with a thickness of several nanometers. The thicker

membrane of polymersomes, in which the entanglement of block copolymers results in

low lateral diffusivity, contributes to a lower permeability and higher stability compared to

liposomes [57−59]. However, the superiority of polymersomes over liposomes in terms of

permeability and stability remains a matter of debate [60].

(7)

Figure 2. Different morphologies of diblock copolymer assemblies in aqueous solutions. Amphiphilic

block copolymers can self-assemble into various ordered structures depending on the ratio of the

hydrophilic and hydrophobic segments. As the hydrophilic fraction of the amphiphile decreases, the

morphology of the polymer assemblies generally shifts from spherical micelles to worm-like micelles

and eventually to polymersomes. Spherical and worm-like micelles can only encapsulate hydrophobic

cargo molecules. Polymersomes can accommodate both hydrophilic and hydrophobic cargo molecules

within their aqueous core and polymer bilayer, respectively.

Upon intravenous administration, nanoparticles are prone to be cleared from the

circulation by the mononuclear phagocyte system [61], thereby hampering the delivery

of drugs to the target site. Phagocytosis is triggered by opsonisation of the nanoparticle

surface with plasma proteins. Poly(ethylene glycol) (PEG) is a biocompatible and

hydrophilic polymer that is used to avoid opsonisation and subsequent degradation

of nanoparticles by opsonin-recognising cells [62]. Amphiphilic block copolymers,

in which PEG comprises the hydrophilic segment, allow for the assembly of 100%

pegylated polymersomes. Due to the potential immunogenicity of PEG following

repeated administration of pegylated liposomes [63−65], other synthetic polymers,

such as poly(vinylpyrrolidone) or poly(glycerol), as well as biopolymers, e.g. hyaluronic

acid, and poly(amino acids) have been applied to prolong the blood circulation time of

nanoparticles. However, the available alternatives for PEG are considered suboptimal for

application in drug delivery [66−68]. In contrast to the limited number of polymers for

the hydrophilic segment, a variety of polymers can comprise the hydrophobic segment of

block copolymers [57], enabling the assembly of polymersomes with specific membrane

properties. For polymersome-mediated drug delivery, biodegradable polymers, such

as poly(caprolactone) (PCL) and poly(trimethylene carbonate) (PTMC), are preferred

because of their susceptibility to hydrolytic and/or enzymatic degradation [69, 70].

(8)

Amphiphilic block copolymers can self-assemble in aqueous solutions into various

ordered structures depending on the ratio of the hydrophilic and hydrophobic

segments. In general, as the hydrophilic fraction of the amphiphile decreases, the

morphology of the polymer assemblies shifts from mostly spherical micelles to

worm-like micelles and eventually to polymersomes (Figure 2) [71−75]. Besides the chemical

composition of the block copolymers, the preparation method may have an effect on the

morphology of polymer assemblies [72]. The solvent displacement method and the thin

film rehydration method are widely used procedures for the formation of polymersomes.

Potential denaturation of the therapeutic cargo by organic solvents is a major drawback

of the solvent displacement method. The direct hydration method, which is a recently

developed procedure, does not involve the use of small molecular organic solvents

for the assembly of polymersomes. Instead, the block copolymers are blended with

oligo(ethylene glycol) prior to hydration. Furthermore, the direct hydration method has

demonstrated a higher encapsulation efficiency of cargo into polymersomes compared

to the thin film rehydration method [76].

G23 PEPTIDE-MEDIATED TRANSPORT ACROSS THE

BLOOD-BRAIN BARRIER

G23 peptide is a GM1 ganglioside-binding peptide [77] that can promote transendothelial

transport of different types of nanoparticles from the blood into the brain [77−81]. GM1 is

concentrated in caveolae at the luminal plasma membrane of endothelial cells [82]. The

enrichment of GM1 in these membrane microdomains suggests a possible involvement

of caveolae-mediated endocytosis in the transport of GM1-targeted nanoparticles

across the endothelium. In addition to GM1, the G23 peptide has binding affinity for

GT1b [77, 83, 84], which is described to promote binding of the peptide to neuronal

cells [84−88]. However, the affinity of the peptide for GT1b may also imply a role for this

trisialoganglioside in the transendothelial transport of G23-functionalized nanoparticles

across the BBB. Interestingly, in recent studies that focus on the neuronal-targeting

properties of the G23 peptide additional BBB-targeting ligands are used in order to form

dual-targeted nanoparticles for the delivery of therapeutics into the brain [52, 89, 90].

Conjugation of G23 peptide to non-biodegradable poly(ethylene

glycol)-block-poly(butadiene) (PEG-b-PBD) polymersomes has been demonstrated to enhance

polymersome transcytosis across the BBB, both in vitro and in vivo [77, 78]. Remarkably,

(9)

the PEG-b-PBD polymersomes also seemed to accumulate in the lung [78]. Other studies

have recently shown the use of the G23 peptide for brain-targeted delivery of

doxorubicin-loaded nanoparticles in glioblastoma tumour-bearing mice, and an RNA-binding protein

complexed with siRNA against β-secretase 1 in a mouse model for Alzheimer’s disease

[79−81]. The reduced levels of transcytosis observed with PEG-b-PBD polymersomes and

alginate-iron oxide nanoparticles that were decorated with scrambled versions of the

G23 peptide emphasise the significance of the peptide sequence for promoting transport

across the BBB [77−79]. Altogether, these observations make G23 a promising ligand for

drug delivery into the brain.

SCOPE OF THE THESIS

Effective delivery of therapeutic cargo from the blood into the brain necessitates nanocarriers

that are decorated with ligands, such as the G23 peptide, that allow for specific binding

to the brain endothelium and stimulate subsequent vesicular transport across the BBB.

In earlier work BBB translocation of non-biodegradable G23-PEG-b-PBD polymersomes

was shown. However, the inability to biologically degrade polymersomes composed of

PEG-b-PBD block copolymers severely limits their application in drug delivery. The aim

of the work described in this thesis was to design biodegradable polymersomes suitable

for the delivery of therapeutics into the brain, and to improve the transcytosis capacity

of G23 peptide-decorated nanocarriers across the BBB. First, we report the formation of

biodegradable PEG-b-PCL polymersomes by the direct hydration method in Chapter 2.

This preparation method is used throughout this thesis for the assembly of biodegradable

PEG-P(CL-g-TMC) polymersomes. Because the PEG-P(CL-g-TMC) polymersomes adhere to

the membrane filter of conventional Transwell

®

culture systems, which are typically used

to prepare in vitro BBB models for the quantification of transendothelial transport, we

establish a filter-free in vitro BBB model to quantitatively study transcytosis of

G23-PEG-P(CL-g-TMC) polymersomes in Chapter 3. In addition, eight other GM1-binding peptides

are conjugated to the polymersomes and their transcytosis capacity is assessed using

the filter-free BBB model. In Chapter 4, we compare the transendothelial transport of

polymersomes decorated with either G23, or the transferrin receptor-targeting peptide

THR, or a combination of both peptides, using the filter-free in vitro BBB. In Chapter 5,

we report the identification of properties that the G23 peptide has in common with

cell-penetrating peptides. Finally, a summary of this thesis and future perspectives regarding

the delivery of nanocarriers across the BBB are presented in Chapter 6.

(10)

REFERENCES

[1] A. Villabona-Rueda, C. Erice, C.A. Pardo, M.F. Stins, The evolving concept of the blood brain barrier (BBB): from a single static barrier to a heterogeneous and dynamic relay center, Front. Cell Neurosci. 13 (2019) 405.

[2] T.M. Mathiisen, K.P. Lehre, N.C. Danbolt, O.P. Ottersen, The perivascular astroglial sheath provides a complete covering of the brain microvessels: an electron microscopic 3D reconstruction, Glia 58 (9) (2010) 1094−1103. [3] P.S. Tsai, J.P. Kaufhold, P. Blinder, B. Friedman, P.J. Drew, H.J. Karten, P.D. Lyden, D. Kleinfeld, Correlations of neuronal

and microvascular densities in murine cortex revealed by direct counting and colocalization of nuclei and vessels, J. Neurosci. 29 (46) (2009) 14553−14570.

[4] T. Mabuchi, J. Lucero, A. Feng, J.A. Koziol, G.J. Del Zoppo, Focal cerebral ischemia preferentially affects neurons distant from their neighboring microvessels, J. Cereb. Blood Flow Metab. 25 (2) (2005) 257−266.

[5] K.E. Schlageter, P. Molnar, G.D. Lapin, D.R. Groothuis, Microvessel organization and structure in experimental brain tumors: microvessel populations with distinctive structural and functional properties, Microvasc. Res. 58 (3) (1999) 312−328.

[6] S.M. Stamatovic, A.M. Johnson, R.F. Keep, A.V. Andjelkovic, Junctional proteins of the blood-brain barrier: new insights into function and dysfunction, Tissue Barriers 4 (1) (2016) e1154641.

[7] T. Worzfeld, M. Schwaninger, Apicobasal polarity of brain endothelial cells, J. Cereb. Blood Flow Metab. 36 (2) (2016) 340−362.

[8] R.F. Haseloff, S. Dithmer, L. Winkler, H. Wolburg, I.E. Blasig, Transmembrane proteins of the tight junctions at the blood-brain barrier: structural and functional aspects, Semin. Cell Dev. Biol. 38 (2015) 16−25.

[9] S.G. Patching, Glucose transporters at the blood-brain barrier: function, regulation and gateways for drug delivery, Mol. Neurobiol. 54 (2) (2017) 1046−1077.

[10] P. Campos-Bedolla, F.R. Walter, S. Veszelka, M.A. Deli, Role of the blood-brain barrier in the nutrition of the central nervous system, Arch. Med. Res. 45 (8) (2014) 610−638.

[11] D.F. Leitner, J.R. Connor, Functional roles of transferrin in the brain, Biochim. Biophys. Acta 1820 (3) (2012) 393−402. [12] L. Descamps, M.P. Dehouck, G. Torpier, R. Cecchelli, Receptor-mediated transcytosis of transferrin through

blood-brain barrier endothelial cells, Am. J. Physiol. 270 (4 Pt 2) (1996) H1149−H1158.

[13] R.L. Roberts, R.E. Fine, A. Sandra, Receptor-mediated endocytosis of transferrin at the blood-brain barrier, J. Cell. Sci. 104 (Pt 2) (1993) 521−532.

[14] P. Candela, F. Gosselet, F. Miller, V. Buee-Scherrer, G. Torpier, R. Cecchelli, L. Fenart, Physiological pathway for low-density lipoproteins across the blood-brain barrier: transcytosis through brain capillary endothelial cells in vitro, Endothelium 15 (5−6) (2008) 254−264.

[15] B. Dehouck, L. Fenart, M.P. Dehouck, A. Pierce, G. Torpier, R. Cecchelli, A new function for the LDL receptor: transcytosis of LDL across the blood-brain barrier, J. Cell Biol. 138 (4) (1997) 877−889.

[16] M. Konishi, M. Sakaguchi, S.M. Lockhart, W. Cai, M.E. Li, E.P. Homan, C. Rask-Madsen, C.R. Kahn, Endothelial insulin receptors differentially control insulin signaling kinetics in peripheral tissues and brain of mice, Proc. Natl. Acad. Sci. USA 114 (40) (2017) E8478−E8487.

[17] R.I. Meijer, S.M. Gray, K.W. Aylor, E.J. Barrett, Pathways for insulin access to the brain: the role of the microvascular endothelial cell, Am. J. Physiol. Heart Circ. Physiol. 311 (5) (2016) H1132−H1138.

[18] W.M. Pardridge, J. Eisenberg, J. Yang, Human blood-brain barrier insulin receptor, J. Neurochem. 44 (6) (1985) 1771−1778.

[19] Alzheimer’s Association, 2019 Alzheimer’s disease facts and figures, Alzheimers Dement. 15 (3) (2019) 321−387. [20] L. Angelov, N.D. Doolittle, D.F. Kraemer, T. Siegal, G.H. Barnett, D.M. Peereboom, G. Stevens, J. McGregor, K. Jahnke,

C.A. Lacy, N.A. Hedrick, E. Shalom, S. Ference, S. Bell, L. Sorenson, R.M. Tyson, M. Haluska, E.A. Neuwelt, Blood-brain barrier disruption and intra-arterial methotrexate-based therapy for newly diagnosed primary CNS lymphoma: a multi-institutional experience, J. Clin. Oncol. 27 (21) (2009) 3503−3509.

[21] D. Fortin, A. Desjardins, A. Benko, T. Niyonsega, M. Boudrias, Enhanced chemotherapy delivery by intraarterial infusion and blood-brain barrier disruption in malignant brain tumors: the Sherbrooke experience, Cancer 103 (12) (2005) 2606−2615.

(11)

[22] N.M. Elkassabany, J. Bhatia, A. Deogaonkar, G.H. Barnett, M. Lotto, M. Maurtua, Z. Ebrahim, A. Schubert, S. Ference, E. Farag, Perioperative complications of blood brain barrier disruption under general anesthesia: a retrospective review, J. Neurosurg. Anesthesiol. 20 (1) (2008) 45−48.

[23] N. Marchi, L. Angelov, T. Masaryk, V. Fazio, T. Granata, N. Hernandez, K. Hallene, T. Diglaw, L. Franic, I. Najm, D. Janigro, Seizure-promoting effect of blood-brain barrier disruption, Epilepsia 48 (4) (2007) 732−742.

[24] D. Furtado, M. Björnmalm, S. Ayton, A.I. Bush, K. Kempe, F. Caruso, Overcoming the blood-brain barrier: the role of nanomaterials in treating neurological diseases, Adv. Mater. 30 (46) (2018) e1801362.

[25] C.T. Lu, Y.Z. Zhao, H.L. Wong, J. Cai, L. Peng, X.Q. Tian, Current approaches to enhance CNS delivery of drugs across the brain barriers, Int. J. Nanomedicine 9 (2014) 2241−2257.

[26] J.L. Mikitsh, A.M. Chacko, Pathways for small molecule delivery to the central nervous system across the blood-brain barrier, Perspect. Medicin. Chem. 6 (2014) 11−24.

[27] A. Mahringer, G. Fricker, ABC transporters at the blood-brain barrier, Expert Opin. Drug Metab. Toxicol. 12 (5) (2016) 499−508.

[28] D. Gomez-Zepeda, M. Taghi, J.M. Scherrmann, X. Decleves, M.C. Menet, ABC transporters at the blood-brain interfaces, their study models, and drug delivery implications in gliomas, Pharmaceutics 12 (1) (2019) E20. [29] M. Bauer, R. Karch, M. Zeitlinger, C. Philippe, K. Römermann, J. Stanek, A. Maier-Salamon, W. Wadsak, W. Jäger, M.

Hacker, M. Müller, O. Langer, Approaching complete inhibition of p-glycoprotein at the human blood-brain barrier: an (R)-[11C]verapamil PET study, J. Cereb. Blood Flow Metab. 35 (5) (2015) 743−746.

[30] W.C. Kreisl, R. Bhatia, C.L. Morse, A.E. Woock, S.S. Zoghbi, H.U. Shetty, V.W. Pike, R.B. Innis, Increased permeability-glycoprotein inhibition at the human blood-brain barrier can be safely achieved by performing PET during peak plasma concentrations of tariquidar, J. Nucl. Med. 56 (1) (2015) 82−87.

[31] N. Singh, G.F. Ecker, Insights into the structure, function, and ligand discovery of the large neutral amino acid transporter 1, LAT1, Int. J. Mol. Sci. 19 (5) (2018) E1278.

[32] Q. Chen, T. Gong, J. Liu, X. Wang, H. Fu, Z. Zhang, Synthesis, in vitro and in vivo characterization of glycosyl derivatives of ibuprofen as novel prodrugs for brain drug delivery, J. Drug Target. 17 (4) (2009) 318−328.

[33] Y. Anraku, H. Kuwahara, Y. Fukusato, A. Mizoguchi, T. Ishii, K. Nitta, Y. Matsumoto, K. Toh, K. Miyata, S. Uchida, K. Nishina, K. Osada, K. Itaka, N. Nishiyama, H. Mizusawa, T. Yamasoba, T. Yokota, K. Kataoka, Glycaemic control boosts glucosylated nanocarrier crossing the BBB into the brain, Nat. Commun. 8 (1) (2017) 1001.

[34] K. Shao, Y. Zhang, N. Ding, S. Huang, J. Wu, J. Li, C. Yang, Q. Leng, L. Ye, J. Lou, L. Zhu, C. Jiang, Functionalized nanoscale micelles with brain targeting ability and intercellular microenvironment biosensitivity for anti-intracranial infection applications, Adv. Healthc. Mater. 4 (2) (2015) 291−300.

[35] Z. Hao, Y. Cui, M. Li, D. Du, M. Liu, H. Tao, S. Li, F. Cao, Y. Chen, X. Lei, L. Wang, D. Zhu, H. Peng, C. Jiang, Liposomes modified with p-aminophenyl-α-D-mannopyranoside: a carrier for targeting cerebral functional regions in mice, Eur. J. Pharm. Biopharm. 84 (3) (2013) 505−516.

[36] J. Barar, M.A. Rafi, M.M. Pourseif, Y. Omidi, Blood-brain barrier transport machineries and targeted therapy of brain diseases, Bioimpacts 6 (4) (2016) 225−248.

[37] W.M. Pardridge, Blood-brain barrier and delivery of protein and gene therapeutics to brain, Front. Aging Neurosci. 11 (2020) 373.

[38] Z. Pang, H. Gao, J. Chen, S. Shen, B. Zhang, J. Ren, L. Guo, Y. Qian, X. Jiang, H. Mei, Intracellular delivery mechanism and brain delivery kinetics of biodegradable cationic bovine serum albumin-conjugated polymersomes, Int. J. Nanomedicine 7 (2012) 3421−3432.

[39] F. Fang, D. Zou, W. Wang, Y. Yin, T. Yin, S. Hao, B. Wang, G. Wang, Y. Wang, Non-invasive approaches for drug delivery to the brain based on the receptor mediated transport, Mater. Sci. Eng. C Mater. Biol. Appl. 76 (2017) 1316−1327. [40] J.V. Georgieva, D. Hoekstra, I.S. Zuhorn, Smuggling drugs into the brain: an overview of ligands targeting transcytosis

for drug delivery across the blood-brain barrier, Pharmaceutics 6 (4) (2014) 557−583.

[41] Z. Pang, W. Lu, H. Gao, K. Hu, J. Chen, C. Zhang, X. Gao, X. Jiang, C. Zhu, Preparation and brain delivery property of biodegradable polymersomes conjugated with OX26, J. Control. Release 128 (2) (2008) 120−127.

[42] S. Fan, Y. Zheng, X. Liu, W. Fang, X. Chen, W. Liao, X. Jing, M. Lei, E. Tao, Q. Ma, X. Zhang, R. Guo, J. Liu, Curcumin-loaded PLGA-PEG nanoparticles conjugated with B6 peptide for potential use in Alzheimer’s disease, Drug Deliv. 25 (1) (2018) 1091−1102.

[43] Y. Zhang, F. Schlachetzki, W.M. Pardridge, Global non-viral gene transfer to the primate brain following intravenous administration, Mol. Ther. 7 (1) (2003) 11−18.

(12)

[44] A.S. Haqqani, G. Thom, M. Burrell, C.E. Delaney, E. Brunette, E. Baumann, C. Sodja, A. Jezierski, C. Webster, D.B. Stanimirovic, Intracellular sorting and transcytosis of the rat transferrin receptor antibody OX26 across the blood-brain barrier in vitro is dependent on its binding affinity, J. Neurochem. 146 (6) (2018) 735−752.

[45] N. Bien-Ly, Y.J. Yu, D. Bumbaca, J. Elstrott, C.A. Boswell, Y. Zhang, W. Luk, Y. Lu, M.S. Dennis, R.M. Weimer, I. Chung, R.J. Watts, Transferrin receptor (TfR) trafficking determines brain uptake of TfR antibody affinity variants, J. Exp. Med. 211 (2) (2014) 233−244.

[46] Y.J. Yu, Y. Zhang, M. Kenrick, K. Hoyte, W. Luk, Y. Lu, J. Atwal, J.M. Elliott, S. Prabhu, R.J. Watts, M.S. Dennis, Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target, Sci. Transl. Med. 3 (84) (2011) 84ra44.

[47] H. Qin, Y. Jiang, J. Zhang, C. Deng, Z. Zhong, Oncoprotein inhibitor rigosertib loaded in ApoE-targeted smart polymersomes reveals high safety and potency against human glioblastoma in mice, Mol. Pharm. 16 (8) (2019) 3711−3719.

[48] Y. Shi, Y. Jiang, J. Cao, W. Yang, J. Zhang, F. Meng, Z. Zhong, Boosting RNAi therapy for orthotopic glioblastoma with nontoxic brain-targeting chimaeric polymersomes, J. Control. Release 292 (2018) 163−171.

[49] Y. Jiang, J. Zhang, F. Meng, Z. Zhong, Apolipoprotein E peptide-directed chimeric polymersomes mediate an ultrahigh-efficiency targeted protein therapy for glioblastoma, ACS Nano 12 (11) (2018) 11070−11079.

[50] Y. Jiang, W. Yang, J. Zhang, F. Meng, Z. Zhong, Protein toxin chaperoned by LRP-1-targeted virus-mimicking vesicles induces high-efficiency glioblastoma therapy in vivo, Adv. Mater. 30 (30) (2018) e1800316.

[51] F. Lu, Z. Pang, J. Zhao, K. Jin, H. Li, Q. Pang, L. Zhang, Z. Pang, Angiopep-2-conjugated poly(ethylene glycol)-co-poly(ε-caprolactone) polymersomes for dual-targeting drug delivery to glioma in rats, Int. J. Nanomedicine 12 (2017) 2117−2127.

[52] T. Jia, Z. Sun, Y. Lu, J. Gao, H. Zou, F. Xie, G. Zhang, H. Xu, D. Sun, Y. Yu, Y. Zhong, A dual brain-targeting curcumin-loaded polymersomes ameliorated cognitive dysfunction in intrahippocampal amyloid-β1-42-injected mice, Int. J. Nanomedicine 11 (2016) 3765−3775.

[53] Y.C. Chen, C.F. Chiang, L.F. Chen, P.C. Liang, W.Y. Hsieh, W.L. Lin, Polymersomes conjugated with des-octanoyl ghrelin and folate as a BBB-penetrating cancer cell-targeting delivery system, Biomaterials 35 (13) (2014) 4066−4081. [54] Y. Yu, Z. Pang, W. Lu, Q. Yin, H. Gao, X. Jiang, Self-assembled polymersomes conjugated with lactoferrin as novel

drug carrier for brain delivery, Pharm. Res. 29 (1) (2012) 83−96.

[55] Z. Pang, H. Gao, Y. Yu, L. Guo, J. Chen, S. Pan, J. Ren, Z. Wen, X. Jiang, Enhanced intracellular delivery and chemotherapy for glioma rats by transferrin-conjugated biodegradable polymersomes loaded with doxorubicin, Bioconjug. Chem. 22 (6) (2011) 1171−1180.

[56] Z. Pang, L. Feng, R. Hua, J. Chen, H. Gao, S. Pan, X. Jiang, P. Zhang, Lactoferrin-conjugated biodegradable polymersome holding doxorubicin and tetrandrine for chemotherapy of glioma rats, Mol. Pharm. 7 (6) (2010) 1995−2005.

[57] C.G. Palivan, R. Goers, A. Najer, X. Zhang, A. Car, W. Meier, Bioinspired polymer vesicles and membranes for biological and medical applications, Chem. Soc. Rev. 45 (2) (2016) 377−411.

[58] E. Rideau, R. Dimova, P. Schwille, F.R. Wurm, K. Landfester, Liposomes and polymersomes: a comparative review towards cell mimicking, Chem. Soc. Rev. 47 (23) (2018) 8572−8610.

[59] L. Messager, J. Gaitzsch, L. Chierico, G. Battaglia, Novel aspects of encapsulation and delivery using polymersomes, Curr. Opin. Pharmacol. 18 (2014) 104−111.

[60] S. Matoori, J.C. Leroux, Twenty-five years of polymersomes: lost in translation?, Mater. Horiz. (2020).

[61] H.H. Gustafson, D. Holt-Casper, D.W. Grainger, H. Ghandehari, Nanoparticle uptake: the phagocyte problem, Nano Today 10 (4) (2015) 487−510.

[62] J.S. Suk, Q. Xu, N. Kim, J. Hanes, L.M. Ensign, PEGylation as a strategy for improving nanoparticle-based drug and gene delivery, Adv. Drug Deliv. Rev. 99 (Pt A) (2016) 28−51.

[63] P.H. Kierstead, H. Okochi, V.J. Venditto, T.C. Chuong, S. Kivimae, J.M. Fréchet, F.C. Szoka, The effect of polymer backbone chemistry on the induction of the accelerated blood clearance in polymer modified liposomes, J. Control. Release 213 (2015) 1−9.

[64] T. Ishida, M. Ichihara, X. Wang, K. Yamamoto, J. Kimura, E. Majima, H. Kiwada, Injection of PEGylated liposomes in rats elicits PEG-specific IgM, which is responsible for rapid elimination of a second dose of PEGylated liposomes, J. Control. Release 112 (1) (2006) 15−25.

(13)

[65] E.T. Dams, P. Laverman, W.J. Oyen, G. Storm, G.L. Scherphof, J.W. Van Der Meer, F.H. Corstens, O.C. Boerman, Accelerated blood clearance and altered biodistribution of repeated injections of sterically stabilized liposomes, J. Pharmacol. Exp. Ther. 292 (3) (2000) 1071−1079.

[66] N. d’Avanzo, C. Celia, A. Barone, M. Carafa, L. Di Marzio, H.A. Santos, M. Fresta, Immunogenicity of polyethylene glycol based nanomedicines: mechanisms, clinical implications and systematic approach, Adv. Therap. 3 (3) (2020) 1900170.

[67] T.T. Hoang Thi, E.H. Pilkington, D.H. Nguyen, J.S. Lee, K.D. Park, N.P. Truong, The importance of poly(ethylene glycol) alternatives for overcoming PEG immunogenicity in drug delivery and bioconjugation, Polymers (Basel) 12 (2) (2020) E298.

[68] K. Knop, R. Hoogenboom, D. Fischer, U.S. Schubert, Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives, Angew. Chem. Int. Ed. Engl. 49 (36) (2010) 6288−6308.

[69] F. Ahmed, D.E. Discher, Self-porating polymersomes of PEG-PLA and PEG-PCL: hydrolysis-triggered controlled release vesicles, J. Control. Release 96 (1) (2004) 37−53.

[70] C. Sanson, C. Schatz, J.F. Le Meins, A. Brûlet, A. Soum, S. Lecommandoux, Biocompatible and biodegradable poly(trimethylene carbonate)-b-poly(L-glutamic acid) polymersomes: size control and stability, Langmuir 26 (4) (2010) 2751−2760.

[71] C. Lebleu, L. Rodrigues, J.M. Guigner, A. Brûlet, E. Garanger, S. Lecommandoux, Self-assembly of PEG-b-PTMC copolymers: micelles and polymersomes size control, Langmuir 35 (41) (2019) 13364−13374.

[72] M. Dionzou, A. Morère, C. Roux, B. Lonetti, J.D. Marty, C. Mingotaud, P. Joseph, D. Goudounèche, B. Payré, M. Léonetti, A.F. Mingotaud, Comparison of methods for the fabrication and the characterization of polymer self-assemblies: what are the important parameters?, Soft Matter 12 (7) (2016) 2166−2176.

[73] J.S. Lee, J. Feijen, Polymersomes for drug delivery: design, formation and characterization, J. Control. Release 161 (2) (2012) 473−483.

[74] D.J. Adams, C. Kitchen, S. Adams, S. Furzeland, D. Atkins, P. Schuetz, C.M. Fernyhough, N. Tzokova, A.J. Ryan, M.F. Butler, On the mechanism of formation of vesicles from poly(ethylene oxide)-block-poly(caprolactone) copolymers, Soft Matter 5 (16) (2009) 3086−3096.

[75] D.A. Christian, S. Cai, D.M. Bowen, Y. Kim, J.D. Pajerowski, D.E. Discher, Polymersome carriers: from self-assembly to siRNA and protein therapeutics, Eur. J. Pharm. Biopharm. 71 (3) (2009) 463−474.

[76] C.P. O’Neil, T. Suzuki, D. Demurtas, A. Finka, J.A. Hubbell, A novel method for the encapsulation of biomolecules into polymersomes via direct hydration, Langmuir 25 (16) (2009) 9025−9029.

[77] J.V. Georgieva, R.P. Brinkhuis, K. Stojanov, C.A. Weijers, H. Zuilhof, F.P. Rutjes, D. Hoekstra, J.C. Van Hest, I.S. Zuhorn, Peptide-mediated blood-brain barrier transport of polymersomes, Angew. Chem. Int. Ed. Engl. 51 (33) (2012) 8339−8342.

[78] K. Stojanov, J.V. Georgieva, R.P. Brinkhuis, J.C. Van Hest, F.P. Rutjes, R.A. Dierckx, E.F. De Vries, I.S. Zuhorn, In vivo biodistribution of prion- and GM1-targeted polymersomes following intravenous administration in mice, Mol. Pharm. 9 (6) (2012) 1620−1627.

[79] C.H. Su, C.Y. Tsai, B. Tomanek, W.Y. Chen, F.Y. Cheng, Evaluation of blood-brain barrier-stealth nanocomposites for in situ glioblastoma theranostics applications, Nanoscale 8 (15) (2016) 7866−7870.

[80] M.M. Haroon, K. Saba, V.H. Boddedda, J.M. Kumar, A.B. Patel, V. Gopal, Delivery of BACE1 siRNA mediated by TARBP-BTP fusion protein reduces β-amyloid deposits in a transgenic mouse model of Alzheimer’s disease, J. Biosci. 44 (1) (2019) 1.

[81] M.M. Haroon, G.H. Dar, D. Jeyalakshmi, U. Venkatraman, K. Saba, N. Rangaraj, A.B. Patel, V. Gopal, A designed recombinant fusion protein for targeted delivery of siRNA to the mouse brain, J. Control. Release 228 (2016) 120−131. [82] J.E. Schnitzer, D.P. McIntosh, A.M. Dvorak, J. Liu, P. Oh, Separation of caveolae from associated microdomains of

GPI-anchored proteins, Science 269 (5229) (1995) 1435−1439.

[83] T. Federici, J.K. Liu, Q. Teng, J. Yang, N.M. Boulis, A means for targeting therapeutics to peripheral nervous system neurons with axonal damage, Neurosurgery 60 (5) (2007) 911−918.

[84] J.K. Liu, Q. Teng, M. Garrity-Moses, T. Federici, D. Tanase, M.J. Imperiale, N.M. Boulis, A novel peptide defined through phage display for therapeutic protein and vector neuronal targeting, Neurobiol. Dis. 19 (3) (2005) 407−418. [85] A.S. Davis, T. Federici, W.C. Ray, N.M. Boulis, D. O’Connor, K.R. Clark, J.S. Bartlett, Rational design and engineering of a

modified adeno-associated virus (AAV1)-based vector system for enhanced retrograde gene delivery, Neurosurgery 76 (2) (2015) 216−225.

(14)

[86] J. Zhang, X. Zhou, Q. Yu, L. Yang, D. Sun, Y. Zhou, J. Liu, Epigallocatechin-3-gallate (EGCG)-stabilized selenium nanoparticles coated with Tet-1 peptide to reduce amyloid-β aggregation and cytotoxicity, ACS Appl. Mater. Interfaces 6 (11) (2014) 8475−8487.

[87] E.J. Kwon, J. Lasiene, B.E. Jacobson, I.K. Park, P.J. Horner, S.H. Pun, Targeted nonviral delivery vehicles to neural progenitor cells in the mouse subventricular zone, Biomaterials 31 (8) (2010) 2417−2424.

[88] I.K. Park, J. Lasiene, S.H. Chou, P.J. Horner, S.H. Pun, Neuron-specific delivery of nucleic acids mediated by Tet1-modified poly(ethylenimine), J. Gene Med. 9 (8) (2007) 691−702.

[89] Q. Guo, S. Xu, P. Yang, P. Wang, S. Lu, D. Sheng, K. Qian, J. Cao, W. Lu, Q. Zhang, A dual-ligand fusion peptide improves the brain-neuron targeting of nanocarriers in Alzheimer’s disease mice, J. Control. Release 320 (2020) 347−362. [90] P. Wang, X. Zheng, Q. Guo, P. Yang, X. Pang, K. Qian, W. Lu, Q. Zhang, X. Jiang, Systemic delivery of BACE1 siRNA

through neuron-targeted nanocomplexes for treatment of Alzheimer’s disease, J. Control. Release 279 (2018) 220−233.

(15)

Referenties

GERELATEERDE DOCUMENTEN

Olke en Anja, dank voor jullie interesse in mijn werkzaamheden en natuurlijk voor het allerbelangrijkste in mijn leven, Jolanda. Last but definitely not least, Jolanda, dank voor

G23 peptide-mediated delivery of biodegradable nanocarriers across an in vitro blood- brain barrier model.. University

 Without an effort in the development of drug delivery systems capable of traversing the blood-brain barrier that is equal to the effort in drug discovery for brain diseases, the

Here, we propose a mathematical model for the transport of paclitaxel across the blood-brain barrier, based on ordinary differential equations, which considers (1) passive diffu-

The Transferrin Receptor at the Blood-Brain Barrier - exploring the possibilities for brain drug delivery..

Physiological drug delivery strategies aim to use endogenous transport mechanisms at the BBB, such as adsorptive-mediated, carrier-mediated or receptor mediated

The obj ective of our research was to determine the expression level of the TfR on brain capillary endothelial cells (BCEC), as well as the endocytosis of 125

For both the concentration and the time dependent experiments, non-specific association was approximately 40 % of total association, which was independent of the applied