Current Update on Transcellular Brain Drug Delivery
Abstract
:1. Introduction
2. Crossing the BBB—A Major Obstacle in Attaining Transcellular Brain Drug Delivery
3. Parameters Governing BBB Permeability and Access to the Brain
3.1. Mechanical Stress
3.2. Basement Membrane and Junction Remodeling
3.3. Cytokines
3.4. Physiochemical Properties
3.5. Age
3.6. Neuroinflammation
3.7. Density of the BBB Receptors
4. Mechanistic Pathways in Transcellular Brain Drug Delivery
4.1. Receptor-Mediated Transcytosis
4.2. Transcytosis
4.3. Cell-Mediated Transcytosis
4.4. Adsorptive Mediated Transcytosis
4.5. Efflux Pumps and Drug Transport
5. Formulation Consideration in Developing Transcellular Brain Drug Delivery Device
5.1. Particle Size
5.2. Solubility of the Drug
5.3. Molecular Weight
5.4. Lipophilicity
5.5. Surface Charge Distribution
6. Strategies for Transcellular Brain Drug Delivery
6.1. Nanomedicines for Transcellular Brain Drug Delivery
Types of Nanocarriers
- Polymeric nanoparticle
- Lipidic nanoparticles
- Inorganic nanoparticles
6.2. Nose-to-Brain Transcellular Brain Drug Delivery
6.3. Peptide as Brain Drug Delivery Carrier
6.4. Exosome as Brain Drug Delivery Carrier
6.5. Drug Delivery via Active Transporters in BBB
6.5.1. Influx Transporters
6.5.2. Efflux Transporters
6.6. Permeability Enhancers for Brain
6.7. Prodrug Approach for Transcellular Brain Drug Delivery
6.8. Redox-Responsive Brain Drug Delivery
6.9. Stem Cell as a Brain Delivery System
7. Body Clearance of Drug following Brain Drug Delivery
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
ABC | ATP binding cassette |
ABP | albumin binding proteins (ABPs) |
AD | Alzheimer’s disease |
AFM | atomic force microscopy |
AMF | alternating magnetic field |
AMT | adsorptive-mediated transcytosis |
AQP4 | aquaporin 4 |
ATP | adenosine triphosphate |
BBB | blood–brain barrier |
BCRP | breast cancer resistance protein |
BDNF | brain-derived neurotrophic factor |
CFTR | cystic fibrosis transmembrane conductance regulator |
CLSM | confocal laser scanning microscopy |
CNS | central nervous system |
CPP | cell penetrating peptide |
CSF | cerebrospinal-fluid |
CX43 | connexin 43 |
DGL-PEG | Dendrigraft-Poly L-lysine-Polyethylene glycol |
DHED | 10β,17β-Dihydroxyestra-1,4-dien-3-one |
DOX | doxorubicin |
DTR | diphtheria toxin receptor |
ECM | extracellular matrix |
ECS | extracellular space |
FCS | fluorescence correlation spectroscopy |
FSS | fluid shear stress |
GIT | gastrointestinal fluid |
GOF | gain of function |
IL | interleukin |
iNOS | inducible nitric oxide synthase |
ISF | interstitial fluid |
IV | intravenous |
LATs | large neutral amino acid transporters |
LDL | lipoprotein |
LIF | leukemia inhibitory factor |
LOF | loss of function |
LRP1 | low-density receptor-related protein |
MFS | major facilitator superfamily |
MMP | matrix metalloproteinase |
MPO | myeloperoxidase |
MRP | multidrug resistance protein |
MSN | mesoporous silica nanoparticles |
MTC | monocarboxylic acid transporters |
NBDs | nucleotide-binding domains |
NO | nitric oxide |
NP | nanoparticle |
NSCs | Neural stem cells |
NTA | nanoparticle tracking analysis |
OAT | organic anion transporters |
OATPs | organic anion transporting polypeptide transporters |
OATs | organic anion transporters |
OCT | organic cation transporters |
OCTs | organic cation transporters |
PAA | polyacrylic acid |
PAMAM | polyamidoamine |
PDPA | poly(2-diisopropyl methacrylate) |
PEG | polyethylene glycol |
P-gp | P-glycoprotein |
PLA | polylactic acid |
PLGA | poly (D, L-lactide-co-glycolide) |
PNP | polymeric nanoparticle |
PT | proximal tubule |
PTX | paclitaxel |
PVS | perivascular space |
RABV | rabies virus |
RES | reticuloendothelial system |
RMT | receptor-mediated transcytosis |
SIT | saturable influx transport |
SLC | solute carrier |
SLCO | solute carrier organic anion |
SLNP | solid lipid nanoparticles |
SOCS3 | suppressor of cytokine signaling 3 |
SPIONs | superparamagnetic iron oxide nanoparticles |
SPR | surface plasmon resonance detection |
SURs | sulfonylurea receptors |
TBI | traumatic brain injury |
TEM | transmission electron microscopy |
TfR | transferrin receptor |
TGF | transforming growth factor |
TJ | tight junctions |
TNF-α | tumor necrosis factor-α |
References
- Kim, J.; Ahn, S.I.; Kim, Y. Nanotherapeutics engineered to cross the blood-brain barrier for advanced drug delivery to the central nervous system. J. Ind. Eng. Chem. 2019, 73, 8–18. [Google Scholar] [CrossRef] [PubMed]
- Reddy, S.; Tatiparti, K.; Sau, S.; Iyer, A.K. Recent advances in nano delivery systems for blood-brain barrier (BBB) penetration and targeting of brain tumors. Drug Discov. Today 2021, 26, 1944–1952. [Google Scholar] [CrossRef] [PubMed]
- Zeiadeh, I.; Najjar, A.; Karaman, R. Strategies for Enhancing the Permeation of CNS-Active Drugs through the Blood-Brain Barrier: A Review. Molecules 2018, 23, 1289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anthony, D.P.; Hegde, M.; Shetty, S.S.; Rafic, T.; Mutalik, S.; Rao, B.S. Targeting receptor-ligand chemistry for drug delivery across blood-brain barrier in brain diseases. Life Sci. 2021, 274, 119326. [Google Scholar] [CrossRef]
- Betterton, R.D.; Davis, T.P.; Ronaldson, P.T. Organic Cation Transporter (OCT/OCTN) expression at brain barrier sites: Focus on CNS drug delivery. In Organic Cation Transporters in the Central Nervous System; Springer: Berlin/Heidelberg, Germany, 2021; pp. 301–328. [Google Scholar]
- Islam, Y.; Leach, A.G.; Smith, J.; Pluchino, S.; Coxon, C.R.; Sivakumaran, M.; Downing, J.; Fatokun, A.A.; Teixidò, M.; Ehtezazi, T. Physiological and pathological factors affecting drug delivery to the brain by nanoparticles. Adv. Sci. 2021, 8, 2002085. [Google Scholar] [CrossRef]
- Ban, J.; Li, S.; Zhan, Q.; Li, X.; Xing, H.; Chen, N.; Long, L.; Hou, X.; Zhao, J.; Yuan, X. PMPC modified PAMAM dendrimer enhances brain tumor-targeted drug delivery. Macromol. Biosci. 2021, 21, 2000392. [Google Scholar] [CrossRef]
- Caraway, C.A.; Gaitsch, H.; Wicks, E.E.; Kalluri, A.; Kunadi, N.; Tyler, B.M. Polymeric nanoparticles in brain cancer therapy: A review of current approaches. Polymers 2022, 14, 2963. [Google Scholar] [CrossRef]
- Jiang, T.; Qiao, Y.; Ruan, W.; Zhang, D.; Yang, Q.; Wang, G.; Chen, Q.; Zhu, F.; Yin, J.; Zou, Y. Cation-Free siRNA Micelles as Effective Drug Delivery Platform and Potent RNAi Nanomedicines for Glioblastoma Therapy. Adv. Mater. 2021, 33, 2104779. [Google Scholar] [CrossRef]
- Yue, Q.; Peng, Y.; Zhao, Y.; Lu, R.; Fu, Q.; Chen, Y.; Yang, Y.; Hai, L.; Guo, L.; Wu, Y. Dual-targeting for brain-specific drug delivery: Synthesis and biological evaluation. Drug Deliv. 2018, 25, 426–434. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Y.; Wang, F.; Wang, K.; Zhong, Y.; Wei, X.; Wang, Q.; Zhang, H. Engineered Exosomes: A Promising Drug Delivery Strategy for Brain Diseases. Curr. Med. Chem. 2022, 29, 3111–3124. [Google Scholar] [CrossRef]
- Barchet, T.M.; Amiji, M.M. Challenges and opportunities in CNS delivery of therapeutics for neurodegenerative diseases. Expert Opin. Drug Deliv. 2009, 6, 211–225. [Google Scholar] [CrossRef] [PubMed]
- Barar, J.; Rafi, M.A.; Pourseif, M.M.; Omidi, Y. Blood-brain barrier transport machineries and targeted therapy of brain diseases. BioImpacts 2016, 6, 225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarin, H. Physiologic upper limits of pore size of different blood capillary types and another perspective on the dual pore theory of microvascular permeability. J. Angiogenesis Res. 2010, 2, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, K.K. Nanobiotechnology-based strategies for crossing the blood–brain barrier. Nanomedicine 2012, 7, 1225–1233. [Google Scholar] [CrossRef]
- Smith, N.M.; Gachulincova, I.; Ho, D.; Bailey, C.; Bartlett, C.A.; Norret, M.; Murphy, J.; Buckley, A.; Rigby, P.J.; House, M.J. An unexpected transient breakdown of the blood brain barrier triggers passage of large intravenously administered nanoparticles. Sci. Rep. 2016, 6, 22595. [Google Scholar] [CrossRef] [Green Version]
- Erickson, M.A.; Banks, W.A.J.P.R. Neuroimmune axes of the blood–brain barriers and blood–brain interfaces: Bases for physiological regulation, disease states, and pharmacological interventions. Pharmacol. Rev. 2018, 70, 278–314. [Google Scholar] [CrossRef] [Green Version]
- Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Volceanov, A.; Teleanu, R.I.J.P. Blood-brain delivery methods using nanotechnology. Pharmaceutics 2018, 10, 269. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Kaplan, J.A.; Colson, Y.L.; Grinstaff, M.W. Mechanoresponsive materials for drug delivery: Harnessing forces for controlled release. Adv. Drug Deliv. Rev. 2017, 108, 68–82. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Yu, J.; Bomba, H.N.; Zhu, Y.; Gu, Z. Mechanical Force-Triggered Drug Delivery. Chem. Rev. 2016, 116, 12536–12563. [Google Scholar] [CrossRef] [Green Version]
- Sabayan, B.; van Buchem, M.A.; Sigurdsson, S.; Zhang, Q.; Harris, T.B.; Gudnason, V.; Arai, A.E.; Launer, L.J. Cardiac hemodynamics are linked with structural and functional features of brain aging: The age, gene/environment susceptibility (AGES)-Reykjavik Study. J. Am. Heart Assoc. 2015, 4, e001294. [Google Scholar] [CrossRef]
- Ma, Y.; Shaik, M.A.; Kozberg, M.G.; Kim, S.H.; Portes, J.P.; Timerman, D.; Hillman, E.M. Resting-state hemodynamics are spatiotemporally coupled to synchronized and symmetric neural activity in excitatory neurons. Proc. Natl. Acad. Sci. USA 2016, 113, E8463–E8471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Wei, J.-Y.; Liu, H.; Wang, K.-J.; Jin, S.-N.; Su, Z.-K.; Wang, H.-J.; Shi, J.-X.; Li, B.; Shang, D.-S. An oxygen-adaptive interaction between SNHG12 and occludin maintains blood-brain barrier integrity. Cell Rep. 2022, 39, 110656. [Google Scholar] [CrossRef] [PubMed]
- Tjakra, M.; Wang, Y.; Vania, V.; Hou, Z.; Durkan, C.; Wang, N.; Wang, G. Overview of Crosstalk Between Multiple Factor of Transcytosis in Blood Brain Barrier. Front. Neurosci. 2019, 13, 1436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Long, K.R.; Shipman, K.E.; Rbaibi, Y.; Menshikova, E.V.; Ritov, V.B.; Eshbach, M.L.; Jiang, Y.; Jackson, E.K.; Baty, C.J.; Weisz, O.A. Proximal tubule apical endocytosis is modulated by fluid shear stress via an mTOR-dependent pathway. Mol. Biol. Cell 2017, 28, 2508–2517. [Google Scholar] [CrossRef]
- Han, J.; Shuvaev, V.V.; Davies, P.F.; Eckmann, D.M.; Muro, S.; Muzykantov, V.R. Flow shear stress differentially regulates endothelial uptake of nanocarriers targeted to distinct epitopes of PECAM-1. J. Control. Release 2015, 210, 39–47. [Google Scholar] [CrossRef] [Green Version]
- Sachdeva, S.; Persaud, S.; Patel, M.; Popard, P.; Colverson, A.; Doré, S. Effects of Sound Interventions on the Permeability of the Blood–Brain Barrier and Meningeal Lymphatic Clearance. Brain Sci. 2022, 12, 742. [Google Scholar] [CrossRef]
- Thomsen, M.S.; Routhe, L.J.; Moos, T. The vascular basement membrane in the healthy and pathological brain. J. Cereb. Blood Flow Metab. 2017, 37, 3300–3317. [Google Scholar] [CrossRef] [Green Version]
- Greene, C.; Hanley, N.; Campbell, M. Claudin-5: Gatekeeper of neurological function. Fluids Barriers CNS 2019, 16, 3. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Jin, X.; Liu, K.J.; Liu, W. Matrix metalloproteinase-2-mediated occludin degradation and caveolin-1-mediated claudin-5 redistribution contribute to blood-brain barrier damage in early ischemic stroke stage. J. Neurosci. 2012, 32, 3044–3057. [Google Scholar] [CrossRef] [Green Version]
- Keaney, J.; Walsh, D.M.; O’Malley, T.; Hudson, N.; Crosbie, D.E.; Loftus, T.; Sheehan, F.; McDaid, J.; Humphries, M.M.; Callanan, J.J.; et al. Autoregulated paracellular clearance of amyloid-β across the blood-brain barrier. Sci. Adv. 2015, 1, e1500472. [Google Scholar] [CrossRef]
- Prados, M.D.; Schold, S.C., Jr.; Fine, H.A.; Jaeckle, K.; Hochberg, F.; Mechtler, L.; Fetell, M.R.; Phuphanich, S.; Feun, L.; Janus, T.J.; et al. A randomized, double-blind, placebo-controlled, phase 2 study of RMP-7 in combination with carboplatin administered intravenously for the treatment of recurrent malignant glioma. Neuro-Oncology 2003, 5, 96–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brunner, J.; Ragupathy, S.; Borchard, G. Target specific tight junction modulators. Adv. Drug Deliv. Rev. 2021, 171, 266–288. [Google Scholar] [CrossRef] [PubMed]
- Emerich, D.F.; Dean, R.L.; Marsh, J.; Pink, M.; Lafreniere, D.; Snodgrass, P.; Bartus, R.T. Intravenous cereport (RMP-7) enhances delivery of hydrophilic chemotherapeutics and increases survival in rats with metastatic tumors in the brain. Pharm. Res. 2000, 17, 1212–1219. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Xie, Y.; Jin, Y.; Hou, X.; Ye, L.; Lou, J. The effect of RMP-7 and its derivative on transporting evens blue liposomes into the brain. Drug Deliv. 2004, 11, 301–309. [Google Scholar] [CrossRef] [PubMed]
- Orellana, J.A.; Figueroa, X.F.; Sánchez, H.A.; Contreras-Duarte, S.; Velarde, V.; Sáez, J.C. Hemichannels in the neurovascular unit and white matter under normal and inflamed conditions. CNS Neurol. Disord. Drug Targets 2011, 10, 404–414. [Google Scholar] [CrossRef] [PubMed]
- Bobbie, M.W.; Roy, S.; Trudeau, K.; Munger, S.J.; Simon, A.M.; Roy, S. Reduced connexin 43 expression and its effect on the development of vascular lesions in retinas of diabetic mice. Investig. Ophthalmol. Vis. Sci. 2010, 51, 3758–3763. [Google Scholar] [CrossRef]
- Liao, C.K.; Wang, S.M.; Chen, Y.L.; Wang, H.S.; Wu, J.C. Lipopolysaccharide-induced inhibition of connexin43 gap junction communication in astrocytes is mediated by downregulation of caveolin-3. Int. J. Biochem. Cell Biol. 2010, 42, 762–770. [Google Scholar] [CrossRef] [PubMed]
- Kolářová, H.; Víteček, J.; Černá, A.; Černík, M.; Přibyl, J.; Skládal, P.; Potěšil, D.; Ihnatová, I.; Zdráhal, Z.; Hampl, A. Myeloperoxidase mediated alteration of endothelial function is dependent on its cationic charge. Free Radic. Biol. Med. 2021, 162, 14–26. [Google Scholar] [CrossRef]
- Shahriary, A.; Sabzevari, M.; Jadidi, K.; Yazdani, F.; Aghamollaei, H. The role of inflammatory cytokines in neovascularization of chemical ocular injury. Ocul. Immunol. Inflamm. 2021, 30, 1149–1161. [Google Scholar] [CrossRef]
- Doğanyiğit, Z.l.; Erbakan, K.; Akyuz, E.; Polat, A.K.; Arulsamy, A.; Shaikh, M.F. The Role of Neuroinflammatory Mediators in the Pathogenesis of Traumatic Brain Injury: A Narrative Review. ACS Chem. Neurosci. 2022, 13, 1835–1848. [Google Scholar] [CrossRef]
- Monhasery, N.; Moll, J.; Cuman, C.; Franke, M.; Lamertz, L.; Nitz, R.; Görg, B.; Häussinger, D.; Lokau, J.; Floss, D.M.; et al. Transcytosis of IL-11 and Apical Redirection of gp130 Is Mediated by IL-11α Receptor. Cell Rep. 2016, 16, 1067–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Williams, J.J.L.; Alotaiq, N.; Mullen, W.; Burchmore, R.; Liu, L.; Baillie, G.S.; Schaper, F.; Pilch, P.F.; Palmer, T.M. Interaction of suppressor of cytokine signalling 3 with cavin-1 links SOCS3 function and cavin-1 stability. Nat. Commun. 2018, 9, 168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sade, H.; Baumgartner, C.; Hugenmatter, A.; Moessner, E.; Freskgård, P.O.; Niewoehner, J. A human blood-brain barrier transcytosis assay reveals antibody transcytosis influenced by pH-dependent receptor binding. PLoS ONE 2014, 9, e96340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chesler, M. Regulation and modulation of pH in the brain. Physiol. Rev. 2003, 83, 1183–1221. [Google Scholar] [CrossRef] [PubMed]
- Römer, W.; Pontani, L.L.; Sorre, B.; Rentero, C.; Berland, L.; Chambon, V.; Lamaze, C.; Bassereau, P.; Sykes, C.; Gaus, K.; et al. Actin dynamics drive membrane reorganization and scission in clathrin-independent endocytosis. Cell 2010, 140, 540–553. [Google Scholar] [CrossRef] [Green Version]
- Delvendahl, I.; Vyleta, N.P.; von Gersdorff, H.; Hallermann, S. Fast, Temperature-Sensitive and Clathrin-Independent Endocytosis at Central Synapses. Neuron 2016, 90, 492–498. [Google Scholar] [CrossRef] [Green Version]
- Rankovic, Z. CNS drug design: Balancing physicochemical properties for optimal brain exposure. J. Med. Chem. 2015, 58, 2584–2608. [Google Scholar] [CrossRef]
- Greig, N.H.; Daly, E.M.; Sweeney, D.J.; Rapoport, S.I. Pharmacokinetics of chlorambucil-tertiary butyl ester, a lipophilic chlorambucil derivative that achieves and maintains high concentrations in brain. Cancer Chemother. Pharmacol. 1990, 25, 320–325. [Google Scholar] [CrossRef]
- Reichel, A. Addressing central nervous system (CNS) penetration in drug discovery: Basics and implications of the evolving new concept. Chem. Biodivers. 2009, 6, 2030–2049. [Google Scholar] [CrossRef]
- Gosselet, F.; Loiola, R.A.; Roig, A.; Rosell, A.; Culot, M. Central nervous system delivery of molecules across the blood-brain barrier. Neurochem. Int. 2021, 144, 104952. [Google Scholar] [CrossRef]
- Zhao, Z.; Nelson, A.R.; Betsholtz, C.; Zlokovic, B.V. Establishment and Dysfunction of the Blood-Brain Barrier. Cell 2015, 163, 1064–1078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof, S.R.; Begley, D.J. Structure and function of the blood-brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
- Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef] [PubMed]
- Erdő, F.; Denes, L.; de Lange, E. Age-associated physiological and pathological changes at the blood-brain barrier: A review. J. Cereb. Blood Flow Metab. 2017, 37, 4–24. [Google Scholar] [CrossRef] [Green Version]
- Senatorov, V.V., Jr.; Friedman, A.R.; Milikovsky, D.Z.; Ofer, J.; Saar-Ashkenazy, R.; Charbash, A.; Jahan, N.; Chin, G.; Mihaly, E.; Lin, J.M.; et al. Blood-brain barrier dysfunction in aging induces hyperactivation of TGFβ signaling and chronic yet reversible neural dysfunction. Sci. Transl. Med. 2019, 11, eaaw8283. [Google Scholar] [CrossRef]
- Yang, A.C.; Stevens, M.Y.; Chen, M.B.; Lee, D.P.; Stähli, D.; Gate, D.; Contrepois, K.; Chen, W.; Iram, T.; Zhang, L.; et al. Physiological blood-brain transport is impaired with age by a shift in transcytosis. Nature 2020, 583, 425–430. [Google Scholar] [CrossRef] [PubMed]
- Cerqueira, S.R.; Ayad, N.G.; Lee, J.K. Neuroinflammation Treatment via Targeted Delivery of Nanoparticles. Front. Cell Neurosci 2020, 14, 576037. [Google Scholar] [CrossRef]
- Zhang, J.; Sadowska, G.B.; Chen, X.; Park, S.Y.; Kim, J.E.; Bodge, C.A.; Cummings, E.; Lim, Y.P.; Makeyev, O.; Besio, W.G.; et al. Anti-IL-6 neutralizing antibody modulates blood-brain barrier function in the ovine fetus. FASEB J. 2015, 29, 1739–1753. [Google Scholar] [CrossRef] [Green Version]
- Miller, D.H.; Thompson, A.J.; Morrissey, S.P.; MacManus, D.G.; Moore, S.G.; Kendall, B.E.; Moseley, I.F.; McDonald, W.I. High dose steroids in acute relapses of multiple sclerosis: MRI evidence for a possible mechanism of therapeutic effect. J. Neurol. Neurosurg. Psychiatry 1992, 55, 450–453. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Zheng, M.; Shimoni, O.; Banks, W.A.; Bush, A.I.; Gamble, J.R.; Shi, B. Development of Novel Therapeutics Targeting the Blood-Brain Barrier: From Barrier to Carrier. Adv. Sci. 2021, 8, e2101090. [Google Scholar] [CrossRef]
- Rojas, O.L.; Pröbstel, A.K.; Porfilio, E.A.; Wang, A.A.; Charabati, M.; Sun, T.; Lee, D.S.W.; Galicia, G.; Ramaglia, V.; Ward, L.A.; et al. Recirculating Intestinal IgA-Producing Cells Regulate Neuroinflammation via IL-10. Cell 2019, 176, 610–624.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ito, M.; Komai, K.; Mise-Omata, S.; Iizuka-Koga, M.; Noguchi, Y.; Kondo, T.; Sakai, R.; Matsuo, K.; Nakayama, T.; Yoshie, O.; et al. Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 2019, 565, 246–250. [Google Scholar] [CrossRef] [PubMed]
- Erickson, M.A.; Dohi, K.; Banks, W.A. Neuroinflammation: A common pathway in CNS diseases as mediated at the blood-brain barrier. Neuroimmunomodulation 2012, 19, 121–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kenney-Jung, D.L.; Vezzani, A.; Kahoud, R.J.; LaFrance-Corey, R.G.; Ho, M.L.; Muskardin, T.W.; Wirrell, E.C.; Howe, C.L.; Payne, E.T. Febrile infection-related epilepsy syndrome treated with anakinra. Ann. Neurol. 2016, 80, 939–945. [Google Scholar] [CrossRef] [PubMed]
- Uchida, Y.; Ohtsuki, S.; Katsukura, Y.; Ikeda, C.; Suzuki, T.; Kamiie, J.; Terasaki, T. Quantitative targeted absolute proteomics of human blood–brain barrier transporters and receptors. J. Neurochem. 2011, 117, 333–345. [Google Scholar] [CrossRef] [PubMed]
- Guo, Q.; Zhu, Q.; Miao, T.; Tao, J.; Ju, X.; Sun, Z.; Li, H.; Xu, G.; Chen, H.; Han, L. LRP1-upregulated nanoparticles for efficiently conquering the blood-brain barrier and targetedly suppressing multifocal and infiltrative brain metastases. J. Control. Release 2019, 303, 117–129. [Google Scholar] [CrossRef] [PubMed]
- Saraiva, C.; Praça, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernardino, L. Nanoparticle-mediated brain drug delivery: Overcoming blood–brain barrier to treat neurodegenerative diseases. J. Control. Release 2016, 235, 34–47. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Liu, L. Modern methods for delivery of drugs across the blood–brain barrier. Adv. Drug Deliv. Rev. 2012, 64, 640–665. [Google Scholar] [CrossRef]
- Azarmi, M.; Maleki, H.; Nikkam, N.; Malekinejad, H. Transcellular brain drug delivery: A review on recent advancements. Int. J. Pharm. 2020, 586, 119582. [Google Scholar] [CrossRef]
- He, Q.; Liu, J.; Liang, J.; Liu, X.; Li, W.; Liu, Z.; Ding, Z.; Tuo, D. Towards Improvements for Penetrating the Blood–Brain Barrier—Recent Progress from a Material and Pharmaceutical Perspective. Cells 2018, 7, 24. [Google Scholar] [CrossRef]
- Di, L.; Kerns, E.J.D.; Kerns, E.H. (Eds.) Chapter 10—Blood-brain barrier. In Drug-Like Properties; Academic Press: Cambridge, MA, USA, 2016; pp. 141–159. [Google Scholar]
- Pardridge, W.M. Drug and gene targeting to the brain with molecular trojan horses. Nat. Rev. Drug Discov. 2002, 1, 131–139. [Google Scholar] [CrossRef]
- Srinivasarao, M.; Galliford, C.V.; Low, P.S. Principles in the design of ligand-targeted cancer therapeutics and imaging agents. Nat. Rev. Drug Discov. 2015, 14, 203–219. [Google Scholar] [CrossRef] [PubMed]
- Strazielle, N.; Ghersi-Egea, J.F. Physiology of Blood–Brain Interfaces in Relation to Brain Disposition of Small Compounds and Macromolecules. Mol. Pharm. 2013, 10, 1473–1491. [Google Scholar] [CrossRef] [PubMed]
- Lajoie, J.M.; Shusta, E.V. Targeting receptor-mediated transport for delivery of biologics across the blood-brain barrier. Annu Rev. Pharm. Toxicol 2015, 55, 613–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pardridge, W.M. Brain Drug Targeting: The Future of Brain Drug Development; Cambridge University Press: Cambridge, UK, 2001. [Google Scholar]
- Zhang, Y.; Pardridge, W.M. Mediated efflux of IgG molecules from brain to blood across the blood–brain barrier. J. Neuroimmunol. 2001, 114, 168–172. [Google Scholar] [CrossRef] [PubMed]
- Shen, J.; Zhao, Z.; Shang, W.; Liu, C.; Zhang, B.; Xu, Z.; Cai, H. Fabrication and evaluation a transferrin receptor targeting nano-drug carrier for cerebral infarction treatment. Artif. Cells Nanomed. Biotechnol. 2019, 47, 192–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnsen, K.B.; Burkhart, A.; Thomsen, L.B.; Andresen, T.L.; Moos, T. Targeting the transferrin receptor for brain drug delivery. Prog. Neurobiol. 2019, 181, 101665. [Google Scholar] [CrossRef]
- Pardridge, W.M. Blood–brain barrier drug delivery of IgG fusion proteins with a transferrin receptor monoclonal antibody. Expert Opin. Drug Deliv. 2015, 12, 207–222. [Google Scholar] [CrossRef]
- Zhang, Y.; Pardridge, W.M. Blood–brain barrier targeting of BDNF improves motor function in rats with middle cerebral artery occlusion. Brain Res. 2006, 1111, 227–229. [Google Scholar] [CrossRef]
- Lee, H.J.; Engelhardt, B.; Lesley, J.; Bickel, U.; Pardridge, W.M. Targeting Rat Anti-Mouse Transferrin Receptor Monoclonal Antibodies through Blood-Brain Barrier in Mouse. J. Pharmacol. Exp. Ther. 2000, 292, 1048. [Google Scholar]
- Sumbria, R.K.; Zhou, Q.-H.; Hui, E.K.-W.; Lu, J.Z.; Boado, R.J.; Pardridge, W.M. Pharmacokinetics and Brain Uptake of an IgG-TNF Decoy Receptor Fusion Protein Following Intravenous, Intraperitoneal, and Subcutaneous Administration in Mice. Mol. Pharm. 2013, 10, 1425–1431. [Google Scholar] [CrossRef] [PubMed]
- Bickel, U.; Yoshikawa, T.; Pardridge, W.M. Delivery of peptides and proteins through the blood–brain barrier. Adv. Drug Deliv. Rev. 2001, 46, 247–279. [Google Scholar] [CrossRef] [PubMed]
- Boado, R.J.; Hui, E.K.-W.; Lu, J.Z.; Zhou, Q.-H.; Pardridge, W.M. Selective targeting of a TNFR decoy receptor pharmaceutical to the primate brain as a receptor-specific IgG fusion protein. J. Biotechnol. 2010, 146, 84–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sagare, A.P.; Deane, R.; Zlokovic, B.V. Low-density lipoprotein receptor-related protein 1: A physiological Aβ homeostatic mechanism with multiple therapeutic opportunities. Pharmacol. Ther. 2012, 136, 94–105. [Google Scholar] [CrossRef] [Green Version]
- Demeule, M.; Currie, J.-C.; Bertrand, Y.; Ché, C.; Nguyen, T.; Régina, A.; Gabathuler, R.; Castaigne, J.-P.; Béliveau, R. Involvement of the low-density lipoprotein receptor-related protein in the transcytosis of the brain delivery vector Angiopep-2. J. Neurochem. 2008, 106, 1534–1544. [Google Scholar] [CrossRef]
- Demeule, M.; Poirier, J.; Jodoin, J.; Bertrand, Y.; Desrosiers, R.R.; Dagenais, C.; Nguyen, T.; Lanthier, J.; Gabathuler, R.; Kennard, M.; et al. High transcytosis of melanotransferrin (P97) across the blood–brain barrier. J. Neurochem. 2002, 83, 924–933. [Google Scholar] [CrossRef]
- Kaefer, M.; Vemulapalli, S.; Freeman Michael, R. A NONTOXIC DIPHTHERIA TOXIN ANALOGUE INHIBITS NEONATAL BLADDER SMOOTH MUSCLE CELL PROLIFERATION. J. Urol. 2000, 163, 580–584. [Google Scholar] [CrossRef]
- Wang, P.; Liu, Y.; Shang, X.; Xue, Y. CRM197-Induced Blood–Brain Barrier Permeability Increase is Mediated by Upregulation of Caveolin-1 Protein. J. Mol. Neurosci. 2011, 43, 485–492. [Google Scholar] [CrossRef]
- Shir, A.; Ogris, M.; Wagner, E.; Levitzki, A. EGF Receptor-Targeted Synthetic Double-Stranded RNA Eliminates Glioblastoma, Breast Cancer, and Adenocarcinoma Tumors in Mice. PLoS Med. 2005, 3, e6. [Google Scholar] [CrossRef]
- Clark, A.J.; Davis, M.E. Increased brain uptake of targeted nanoparticles by adding an acid-cleavable linkage between transferrin and the nanoparticle core. Proc. Natl. Acad. Sci. USA 2015, 112, 12486–12491. [Google Scholar] [CrossRef] [Green Version]
- Cai, L.; Yang, C.; Jia, W.; Liu, Y.; Xie, R.; Lei, T.; Yang, Z.; He, X.; Tong, R.; Gao, H. Endo/lysosome-escapable delivery depot for improving BBB transcytosis and neuron targeted therapy of Alzheimer’s disease. Adv. Funct. Mater. 2020, 30, 1909999. [Google Scholar] [CrossRef]
- Georgieva, J.V.; Hoekstra, D.; Zuhorn, I.S. Smuggling Drugs into the Brain: An Overview of Ligands Targeting Transcytosis for Drug Delivery across the Blood–Brain Barrier. Pharmaceutics 2014, 6, 557–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pardridge, W.M.J.M.i. Blood-brain barrier drug targeting: The future of brain drug development. Mol. Interv. 2003, 3, 90. [Google Scholar] [CrossRef] [Green Version]
- Ayloo, S.; Gu, C. Transcytosis at the blood–brain barrier. Curr. Opin. Neurobiol. 2019, 57, 32–38. [Google Scholar] [CrossRef] [PubMed]
- Preston, J.E.; Joan Abbott, N.; Begley, D.J. Chapter Five—Transcytosis of Macromolecules at the Blood–Brain Barrier. In Advances in Pharmacology; Davis, T.P., Ed.; Academic Press: Cambridge, MA, USA, 2014; Volume 71, pp. 147–163. [Google Scholar]
- Hill, M.M.; Bastiani, M.; Luetterforst, R.; Kirkham, M.; Kirkham, A.; Nixon, S.J.; Walser, P.; Abankwa, D.; Oorschot, V.M.J.; Martin, S.; et al. PTRF-Cavin, a Conserved Cytoplasmic Protein Required for Caveola Formation and Function. Cell 2008, 132, 113–124. [Google Scholar] [CrossRef] [Green Version]
- Villaseñor, R.; Lampe, J.; Schwaninger, M.; Collin, L. Intracellular transport and regulation of transcytosis across the blood–brain barrier. Cell. Mol. Life Sci. 2019, 76, 1081–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, M.M.; Patel, B.M. Crossing the Blood–Brain Barrier: Recent Advances in Drug Delivery to the Brain. CNS Drugs 2017, 31, 109–133. [Google Scholar] [CrossRef]
- Rogers, W.J.; Basu, P. Factors regulating macrophage endocytosis of nanoparticles: Implications for targeted magnetic resonance plaque imaging. Atherosclerosis 2005, 178, 67–73. [Google Scholar] [CrossRef]
- Hou, J.; Yang, X.; Li, S.; Cheng, Z.; Wang, Y.; Zhao, J.; Zhang, C.; Li, Y.; Luo, M.; Ren, H.; et al. Accessing neuroinflammation sites: Monocyte/neutrophil-mediated drug delivery for cerebral ischemia. Sci. Adv. 2019, 5, eaau8301. [Google Scholar] [CrossRef] [Green Version]
- Jain, S.; Mishra, V.; Singh, P.; Dubey, P.K.; Saraf, D.K.; Vyas, S.P. RGD-anchored magnetic liposomes for monocytes/neutrophils-mediated brain targeting. Int. J. Pharm. 2003, 261, 43–55. [Google Scholar] [CrossRef]
- Batrakova, E.V.; Gendelman, H.E.; Kabanov, A.V. Cell-mediated drug delivery. Expert Opin. Drug Deliv. 2011, 8, 415–433. [Google Scholar] [CrossRef] [PubMed]
- Park, K.J.J.o.C.R. Trojan monocytes for improved drug delivery to the brain. J. Control. Release 2008, 2, 75. [Google Scholar] [CrossRef] [PubMed]
- Ayer, M.; Klok, H.-A. Cell-mediated delivery of synthetic nano- and microparticles. J. Control. Release 2017, 259, 92–104. [Google Scholar] [CrossRef] [PubMed]
- Zhu, X.; Jin, K.; Huang, Y.; Pang, Z. 7—Brain drug delivery by adsorption-mediated transcytosis. In Brain Targeted Drug Delivery System; Gao, H., Gao, X., Eds.; Academic Press: Cambridge, MA, USA, 2019. [Google Scholar] [CrossRef]
- Belting, M. Heparan sulfate proteoglycan as a plasma membrane carrier. Trends Biochem. Sci. 2003, 28, 145–151. [Google Scholar] [CrossRef]
- Muniswamy, V.J.; Raval, N.; Gondaliya, P.; Tambe, V.; Kalia, K.; Tekade, R.K. ‘Dendrimer-Cationized-Albumin’ encrusted polymeric nanoparticle improves BBB penetration and anticancer activity of doxorubicin. Int. J. Pharm. 2019, 555, 77–99. [Google Scholar] [CrossRef]
- Monsalve, Y.; Tosi, G.; Ruozi, B.; Belletti, D.; Vilella, A.; Zoli, M.; Vandelli, M.A.; Forni, F.; López, B.L.; Sierra, L. PEG-g-chitosan nanoparticles functionalized with the monoclonal antibody OX26 for brain drug targeting. Nanomedicine 2015, 10, 1735–1750. [Google Scholar] [CrossRef]
- Janaszewska, A.; Ziemba, B.; Ciepluch, K.; Appelhans, D.; Voit, B.; Klajnert, B.; Bryszewska, M.J. The biodistribution of maltotriose modified poly (propylene imine)(PPI) dendrimers conjugated with fluorescein—Proofs of crossing blood–brain–barrier. New J. Chem. 2012, 36, 350–353. [Google Scholar] [CrossRef]
- Hervé, F.; Ghinea, N.; Scherrmann, J.-M. CNS Delivery Via Adsorptive Transcytosis. AAPS J. 2008, 10, 455–472. [Google Scholar] [CrossRef] [Green Version]
- Kristensen, M.; Brodin, B. Routes for Drug Translocation Across the Blood-Brain Barrier: Exploiting Peptides as Delivery Vectors. J. Pharm. Sci. 2017, 106, 2326–2334. [Google Scholar] [CrossRef] [Green Version]
- Aarts, M.; Liu, Y.; Liu, L.; Besshoh, S.; Arundine, M.; Gurd, J.W.; Wang, Y.-T.; Salter, M.W.; Tymianski, M. Treatment of Ischemic Brain Damage by Perturbing NMDA Receptor- PSD-95 Protein Interactions. Science 2002, 298, 846–850. [Google Scholar] [CrossRef]
- Lim, S.; Kim, W.-J.; Kim, Y.-H.; Lee, S.; Koo, J.-H.; Lee, J.-A.; Kim, H.-M.; Park, H.-J.; Kim, D.-H.; Lee, H.-G.; et al. BBB-permeable peptide conjugated cytoplasmic domain of CTLA-4 inhibits Th1 and Th17 responses and pathogenesis of multiple sclerosis. J. Immunol. 2016, 196. [Google Scholar]
- Song, J.; Lu, C.; Leszek, J.; Zhang, J. Design and Development of Nanomaterial-Based Drug Carriers to Overcome the Blood–Brain Barrier by Using Different Transport Mechanisms. Int. J. Mol. Sci. 2021, 22, 10118. [Google Scholar] [CrossRef] [PubMed]
- Kamalinia, G.; Khodagholi, F.; Shaerzadeh, F.; Tavssolian, F.; Chaharband, F.; Atyabi, F.; Sharifzadeh, M.; Amini, M.; Dinarvand, R. Cationic Albumin-Conjugated Chelating Agent as a Novel Brain Drug Delivery System in Neurodegeneration. Chem. Biol. Drug Des. 2015, 86, 1203–1214. [Google Scholar] [CrossRef] [PubMed]
- Annilo, T.; Tammur, J.; Hutchinson, A.; Rzhetsky, A.; Dean, M.; Allikmets, R. Human and mouse orthologs of a new ATP-binding cassette gene, ABCG4. Cytogenet. Genome Res. 2001, 94, 196–201. [Google Scholar] [CrossRef]
- Eckford, P.D.W.; Sharom, F.J. ABC Efflux Pump-Based Resistance to Chemotherapy Drugs. Chem. Rev. 2009, 109, 2989–3011. [Google Scholar] [CrossRef] [PubMed]
- Tamaki, A.; Ierano, C.; Szakacs, G.; Robey, R.W.; Bates, S.E. The controversial role of ABC transporters in clinical oncology. Essays Biochem. 2011, 50, 209–232. [Google Scholar] [CrossRef]
- Rice, A.; Liu, Y.; Michaelis, M.L.; Himes, R.H.; Georg, G.I.; Audus, K.L. Chemical Modification of Paclitaxel (Taxol) Reduces P-Glycoprotein Interactions and Increases Permeation across the Blood−Brain Barrier in Vitro and in Situ. J. Med. Chem. 2005, 48, 832–838. [Google Scholar] [CrossRef]
- Ndungu, J.M.; Lu, Y.J.; Zhu, S.; Yang, C.; Wang, X.; Chen, G.; Shin, D.M.; Snyder, J.P.; Shoji, M.; Sun, A. Targeted Delivery of Paclitaxel to Tumor Cells: Synthesis and in Vitro Evaluation. J. Med. Chem. 2010, 53, 3127–3132. [Google Scholar] [CrossRef] [Green Version]
- Miller, D.W.; Kabanov, A.V. Potential applications of polymers in the delivery of drugs to the central nervous system. Colloids Surf. B: Biointerfaces 1999, 16, 321–330. [Google Scholar] [CrossRef]
- Mori, S.; Ohtsuki, S.; Takanaga, H.; Kikkawa, T.; Kang, Y.-S.; Terasaki, T. Organic anion transporter 3 is involved in the brain-to-blood efflux transport of thiopurine nucleobase analogs. J. Neurochem. 2004, 90, 931–941. [Google Scholar] [CrossRef]
- Ohtsuki, S.; Terasaki, T. Contribution of Carrier-Mediated Transport Systems to the Blood–Brain Barrier as a Supporting and Protecting Interface for the Brain; Importance for CNS Drug Discovery and Development. Pharm. Res. 2007, 24, 1745–1758. [Google Scholar] [CrossRef] [PubMed]
- Tamai, I.; Tsuji, A. Transporter-Mediated Permeation of Drugs Across the Blood–Brain Barrier. J. Pharm. Sci. 2000, 89, 1371–1388. [Google Scholar] [CrossRef] [PubMed]
- Colabufo, N.A.; Berardi, F.; Contino, M.; Niso, M.; Perrone, R. ABC Pumps and Their Role in Active Drug Transport. Curr. Top. Med. Chem. 2009, 9, 119–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Rooy, I.; Cakir-Tascioglu, S.; Hennink, W.E.; Storm, G.; Schiffelers, R.M.; Mastrobattista, E. In vivo methods to study uptake of nanoparticles into the brain. Pharm. Res. 2011, 28, 456–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shilo, M.; Sharon, A.; Baranes, K.; Motiei, M.; Lellouche, J.-P.M.; Popovtzer, R.J. The effect of nanoparticle size on the probability to cross the blood-brain barrier: An in-vitro endothelial cell model. J. Nanobiotechnol 2015, 13, 19. [Google Scholar] [CrossRef] [Green Version]
- Thorne, R.G.; Nicholson, C. In vivo diffusion analysis with quantum dots and dextrans predicts the width of brain extracellular space. Proc. Natl. Acad. Sci. USA 2006, 103, 5567–5572. [Google Scholar] [CrossRef] [Green Version]
- Nance, E.A.; Woodworth, G.F.; Sailor, K.A.; Shih, T.-Y.; Xu, Q.; Swaminathan, G.; Xiang, D.; Eberhart, C.; Hanes, J. A dense poly (ethylene glycol) coating improves penetration of large polymeric nanoparticles within brain tissue. Sci. Transl. Med. 2012, 4, 149ra119. [Google Scholar] [CrossRef] [Green Version]
- Yu, H.; Kim, K. Direct nose-to-brain transfer of a growth hormone releasing neuropeptide, hexarelin after intranasal administration to rabbits. Int. J. Pharm. 2009, 378, 73–79. [Google Scholar] [CrossRef]
- Aiassa, V.; Garnero, C.; Longhi, M.R.; Zoppi, A. Cyclodextrin multicomponent complexes: Pharmaceutical applications. Pharmaceutics 2021, 13, 1099. [Google Scholar] [CrossRef]
- Laddha, U.D.; Tagalpallewar, A.A. Physicochemical, biopharmaceutical, and practical considerations for efficient nose-to-brain drug delivery. In Direct Nose-to-Brain Drug Delivery; Elsevier: Piscataway, NJ, USA, 2021; pp. 39–54. [Google Scholar]
- Bahadur, S.; Pathak, K. Physicochemical and physiological considerations for efficient nose-to-brain targeting. Expert Opin. Drug Deliv. 2012, 9, 19–31. [Google Scholar] [CrossRef]
- Costantino, H.R.; Illum, L.; Brandt, G.; Johnson, P.H.; Quay, S.C. Intranasal delivery: Physicochemical and therapeutic aspects. Int. J. Pharm. 2007, 337, 1–24. [Google Scholar] [CrossRef] [PubMed]
- Illum, L.J. Nasal drug delivery—Possibilities, problems and solutions. J. Control. Release 2003, 87, 187–198. [Google Scholar] [CrossRef]
- Ribeiro, M.M.; Domingues, M.M.; Freire, J.M.; Santos, N.C.; Castanho, M.A. Translocating the blood-brain barrier using electrostatics. Front. Cell. Neurosci. 2012, 6, 44. [Google Scholar] [CrossRef] [Green Version]
- Drin, G.; Cottin, S.; Blanc, E.; Rees, A.R.; Temsamani, J. Studies on the internalization mechanism of cationic cell-penetrating peptides. J. Biol. Chem 2003, 278, 31192–31201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gabal, Y.M.; Kamel, A.O.; Sammour, O.A.; Elshafeey, A.H. Effect of surface charge on the brain delivery of nanostructured lipid carriers in situ gels via the nasal route. Int. J. Pharm. 2014, 473, 442–457. [Google Scholar] [CrossRef] [PubMed]
- Rawal, S.U.; Patel, B.M.; Patel, M.M. New Drug Delivery Systems Developed for Brain Targeting. Drugs 2022, 82, 749–792. [Google Scholar] [CrossRef] [PubMed]
- Naqvi, S.; Panghal, A.; Flora, S.J.S. Nanotechnology: A Promising Approach for Delivery of Neuroprotective Drugs. Front. Neurosci. 2020, 14, 494. [Google Scholar] [CrossRef]
- Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnology 2018, 16, 71. [Google Scholar] [CrossRef] [Green Version]
- Zhang, F.; Trent Magruder, J.; Lin, Y.A.; Crawford, T.C.; Grimm, J.C.; Sciortino, C.M.; Wilson, M.A.; Blue, M.E.; Kannan, S.; Johnston, M.V.; et al. Generation-6 hydroxyl PAMAM dendrimers improve CNS penetration from intravenous administration in a large animal brain injury model. J. Control. Release 2017, 249, 173–182. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Sun, X.; Mei, H.; Wang, Y.; Liao, Z.; Chen, J.; Zhang, Q.; Hu, Y.; Pang, Z.; Jiang, X. LDLR-mediated peptide-22-conjugated nanoparticles for dual-targeting therapy of brain glioma. Biomaterials 2013, 34, 9171–9182. [Google Scholar] [CrossRef]
- Hu, Q.; Gao, X.; Gu, G.; Kang, T.; Tu, Y.; Liu, Z.; Song, Q.; Yao, L.; Pang, Z.; Jiang, X. Glioma therapy using tumor homing and penetrating peptide-functionalized PEG–PLA nanoparticles loaded with paclitaxel. Biomaterials 2013, 34, 5640–5650. [Google Scholar] [CrossRef] [PubMed]
- Tang, W.; Fan, W.; Lau, J.; Deng, L.; Shen, Z.; Chen, X. Emerging blood–brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem. Soc. Rev. 2019, 48, 2967–3014. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Patel, T.R.; Sirianni, R.W.; Strohbehn, G.; Zheng, M.Q.; Duong, N.; Schafbauer, T.; Huttner, A.J.; Huang, Y.; Carson, R.E.; et al. Highly penetrative, drug-loaded nanocarriers improve treatment of glioblastoma. Proc. Natl. Acad. Sci. USA 2013, 110, 11751–11756. [Google Scholar] [CrossRef] [Green Version]
- Mojarad-Jabali, S.; Farshbaf, M.; Walker, P.R.; Hemmati, S.; Fatahi, Y.; Zakeri-Milani, P.; Sarfraz, M.; Valizadeh, H. An update on actively targeted liposomes in advanced drug delivery to glioma. Int. J. Pharm. 2021, 602, 120645. [Google Scholar] [CrossRef] [PubMed]
- Kenchappa, V.; Cao, R.; Venketaraman, V.; Betageri, G.V. Liposomes as Carriers for the Delivery of Efavirenz in Combination with Glutathione—An Approach to Combat Opportunistic Infections. Appl. Sci. 2022, 12, 1468. [Google Scholar] [CrossRef]
- Shi, D.; Mi, G.; Shen, Y.; Webster, T.J. Glioma-targeted dual functionalized thermosensitive Ferri-liposomes for drug delivery through an in vitro blood–brain barrier. Nanoscale 2019, 11, 15057–15071. [Google Scholar] [CrossRef]
- Wiley, D.T.; Webster, P.; Gale, A.; Davis, M.E. Transcytosis and brain uptake of transferrin-containing nanoparticles by tuning avidity to transferrin receptor. Proc. Natl. Acad. Sci. USA 2013, 110, 8662–8667. [Google Scholar] [CrossRef] [Green Version]
- Lee, D.; Minko, T. Nanotherapeutics for Nose-to-Brain Drug Delivery: An Approach to Bypass the Blood Brain Barrier. Pharmaceutics 2021, 13, 2049. [Google Scholar] [CrossRef]
- Manta, K.; Papakyriakopoulou, P.; Chountoulesi, M.; Diamantis, D.A.; Spaneas, D.; Vakali, V.; Naziris, N.; Chatziathanasiadou, M.V.; Andreadelis, I.; Moschovou, K. Preparation and biophysical characterization of quercetin inclusion complexes with β-cyclodextrin derivatives to be formulated as possible nose-to-brain quercetin delivery systems. Mol. Pharm. 2020, 17, 4241–4255. [Google Scholar] [CrossRef]
- Papakyriakopoulou, P.; Manta, K.; Kostantini, C.; Kikionis, S.; Banella, S.; Ioannou, E.; Christodoulou, E.; Rekkas, D.M.; Dallas, P.; Vertzoni, M. Nasal powders of quercetin-β-cyclodextrin derivatives complexes with mannitol/lecithin microparticles for Nose-to-Brain delivery: In vitro and ex vivo evaluation. Int. J. Pharm. 2021, 607, 121016. [Google Scholar] [CrossRef]
- Khoury, R.; Rajamanickam, J.; Grossberg, G.T. An update on the safety of current therapies for Alzheimer’s disease: Focus on rivastigmine. Ther. Adv. Drug Saf. 2018, 9, 171–178. [Google Scholar] [CrossRef] [PubMed]
- Islam, Y.; Leach, A.G.; Smith, J.; Pluchino, S.; Coxonl, C.R.; Sivakumaran, M.; Downing, J.; Fatokun, A.A.; Teixidò, M.; Ehtezazi, T. Peptide based drug delivery systems to the brain. Nano Express 2020, 1, 012002. [Google Scholar] [CrossRef]
- Kim, G.; Kim, M.; Lee, Y.; Byun, J.W.; Hwang, D.W.; Lee, M. Systemic delivery of microRNA-21 antisense oligonucleotides to the brain using T7-peptide decorated exosomes. J. Control. Release 2020, 317, 273–281. [Google Scholar] [CrossRef] [PubMed]
- Al-azzawi, S.; Masheta, D.; Guildford, A.; Phillips, G.; Santin, M. A Peptide-Based Nanocarrier for an Enhanced Delivery and Targeting of Flurbiprofen into the Brain for the Treatment of Alzheimer’s Disease: An In Vitro Study. Nanomaterials 2020, 10, 1590. [Google Scholar] [CrossRef]
- Li, J.; Zhang, Q.; Pang, Z.; Wang, Y.; Liu, Q.; Guo, L.; Jiang, X. Identification of peptide sequences that target to the brain using in vivo phage display. Amino Acids 2012, 42, 2373–2381. [Google Scholar] [CrossRef] [PubMed]
- Patil, S.M.; Sawant, S.S.; Kunda, N.K. Exosomes as drug delivery systems: A brief overview and progress update. Eur. J. Pharm. Biopharm. 2020, 154, 259–269. [Google Scholar] [CrossRef] [PubMed]
- Liao, W.; Du, Y.; Zhang, C.; Pan, F.; Yao, Y.; Zhang, T.; Peng, Q. Exosomes: The next generation of endogenous nanomaterials for advanced drug delivery and therapy. Acta Biomater. 2018, 86, 1–14. [Google Scholar] [CrossRef]
- Maheshwari, R.; Tekade, M.; Gondaliya, P.; Kalia, K.; D’Emanuele, A.; Tekade, R.K. Recent advances in exosome-based nanovehicles as RNA interference therapeutic carriers. Nanomedicine 2017, 12, 2653–2675. [Google Scholar] [CrossRef]
- Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef]
- Peng, H.; Ji, W.; Zhao, R.; Yang, J.; Lu, Z.; Li, Y.; Zhang, X. Exosome: A significant nano-scale drug delivery carrier. J. Mater. Chem. B 2020, 8, 7591–7608. [Google Scholar] [CrossRef]
- Bunggulawa, E.; Wang, W.; Yin, T.; Wang, N.; Durkan, C.; Wang, Y.; Wang, G. Recent advancements in the use of exosomes as drug delivery systems. J. Nanobiotechnol. 2018, 16, 81. [Google Scholar] [CrossRef] [PubMed]
- Morad, G.; Carman, C.V.; Hagedorn, E.J.; Perlin, J.R.; Zon, L.I.; Mustafaoglu, N.; Park, T.-E.; Ingber, D.E.; Daisy, C.C.; Moses, M.A. Tumor-Derived Extracellular Vesicles Breach the Intact Blood–Brain Barrier via Transcytosis. ACS Nano 2019, 13, 13853–13865. [Google Scholar] [CrossRef] [PubMed]
- Jia, G.; Han, Y.; An, Y.; Ding, Y.; He, C.; Wang, X.; Tang, Q. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials 2018, 178, 302–316. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Wu, J.; Wu, J.; Fan, Q.; Zhou, J.; Wu, J.; Liu, S.; Zang, J.; Ye, J.; Xiao, M.; et al. Exosome-mediated targeted delivery of miR-210 for angiogenic therapy after cerebral ischemia in mice. J. Nanobiotechnol. 2019, 17, 29. [Google Scholar] [CrossRef] [Green Version]
- Tian, T.; Zhang, H.-X.; He, C.-P.; Fan, S.; Zhu, Y.-L.; Qi, C.; Huang, N.-P.; Xiao, Z.-D.; Lu, Z.-H.; Tannous, B.A.; et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018, 150, 137–149. [Google Scholar] [CrossRef]
- Puris, E.; Fricker, G.; Gynther, M. Targeting Transporters for Drug Delivery to the Brain: Can We Do Better? Pharm. Res. 2022, 39, 1415–1455. [Google Scholar] [CrossRef]
- Brzica, H.; Abdullahi, W.; Ibbotson, K.; Ronaldson, P.T. Role of Transporters in Central Nervous System Drug Delivery and Blood-Brain Barrier Protection: Relevance to Treatment of Stroke. J. Cent. Nerv Syst Dis 2017, 9, 1179573517693802. [Google Scholar] [CrossRef]
- Ueno, M.; Nakagawa, T.; Wu, B.; Onodera, M.; Huang, C.l.; Kusaka, T.; Araki, N.; Sakamoto, H. Transporters in the Brain Endothelial Barrier. Curr. Med. Chem. 2010, 17, 1125–1138. [Google Scholar] [CrossRef]
- Abdul Razzak, R.; Florence, G.J.; Gunn-Moore, F.J. Approaches to CNS Drug Delivery with a Focus on Transporter-Mediated Transcytosis. Int. J. Mol. Sci. 2019, 20, 3108. [Google Scholar] [CrossRef] [Green Version]
- Lu, X. The Role of Large Neutral Amino Acid Transporter (LAT1) in Cancer. Curr. Cancer Drug Targets 2019, 19, 863–876. [Google Scholar] [CrossRef] [PubMed]
- Obaidat, A.; Roth, M.; Hagenbuch, B. The expression and function of organic anion transporting polypeptides in normal tissues and in cancer. Annu. Rev. Pharmacol. Toxicol. 2012, 52, 135–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thakkar, N.; Lockhart, A.C.; Lee, W. Role of Organic Anion-Transporting Polypeptides (OATPs) in Cancer Therapy. AAPS J. 2015, 17, 535–545. [Google Scholar] [CrossRef] [Green Version]
- Kalliokoski, A.; Niemi, M. Impact of OATP transporters on pharmacokinetics. Br. J. Pharmacol. 2009, 158, 693–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nies, A.T.; Koepsell, H.; Damme, K.; Schwab, M. Organic Cation Transporters (OCTs, MATEs), In Vitro and In Vivo Evidence for the Importance in Drug Therapy. In Drug Transporters; Fromm, M.F., Kim, R.B., Eds.; Springer: Berlin/Heidelberg, Germany, 2011; pp. 105–167. [Google Scholar] [CrossRef]
- Koepsell, H. General Overview of Organic Cation Transporters in Brain. In Organic Cation Transporters in the Central Nervous System; Daws, L.C., Ed.; Springer International Publishing: Cham, Switzerland, 2021; pp. 1–39. [Google Scholar] [CrossRef]
- Couroussé, T.; Gautron, S. Role of organic cation transporters (OCTs) in the brain. Pharmacol. Ther. 2015, 146, 94–103. [Google Scholar] [CrossRef]
- Nigam, S.K.; Bush, K.T.; Martovetsky, G.; Ahn, S.-Y.; Liu, H.C.; Richard, E.; Bhatnagar, V.; Wu, W. The organic anion transporter (OAT) family: A systems biology perspective. Physiol. Rev. 2015, 95, 83–123. [Google Scholar] [CrossRef]
- Hosoya, K.-i.; Tachikawa, M. Roles of organic anion/cation transporters at the blood–brain and blood–cerebrospinal fluid barriers involving uremic toxins. Clin. Exp. Nephrol. 2011, 15, 478–485. [Google Scholar] [CrossRef]
- Halestrap, A.P.; Wilson, M.C. The monocarboxylate transporter family—Role and regulation. IUBMB Life 2012, 64, 109–119. [Google Scholar] [CrossRef]
- Vijay, N.; Morris, M.E. Role of monocarboxylate transporters in drug delivery to the brain. Curr. Pharm. Des. 2014, 20, 1487–1498. [Google Scholar] [CrossRef]
- Sun, Y.; Sun, J.; He, Z.; Wang, G.; Wang, Y.; Zhao, D.; Wang, Z.; Luo, C.; Tian, C.; Jiang, Q. Monocarboxylate transporter 1: A promising drug-delivery target in the treatment of brain diseases and cancers. Curr. Drug Metab. 2019, 20, 855–866. [Google Scholar] [CrossRef]
- Benarroch, E.E. Brain glucose transporters. Neurology 2014, 82, 1374. [Google Scholar] [CrossRef]
- Koepsell, H. Glucose transporters in brain in health and disease. Pflug. Arch. 2020, 472, 1299–1343. [Google Scholar] [CrossRef] [PubMed]
- Patching, S.G. Glucose Transporters at the Blood-Brain Barrier: Function, Regulation and Gateways for Drug Delivery. Mol. Neurobiol. 2017, 54, 1046–1077. [Google Scholar] [CrossRef] [PubMed]
- Fletcher, J.I.; Haber, M.; Henderson, M.J.; Norris, M.D. ABC transporters in cancer: More than just drug efflux pumps. Nat. Rev. Cancer 2010, 10, 147–156. [Google Scholar] [CrossRef]
- Gomez-Zepeda, D.; Taghi, M.; Scherrmann, J.-M.; Decleves, X.; Menet, M.-C. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019, 12, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mahringer, A.; Fricker, G. ABC transporters at the blood–brain barrier. Expert Opin. Drug Metab. Toxicol. 2016, 12, 499–508. [Google Scholar] [CrossRef]
- Löscher, W.; Potschka, H. Role of drug efflux transporters in the brain for drug disposition and treatment of brain diseases. Prog. Neurobiol. 2005, 76, 22–76. [Google Scholar] [CrossRef] [PubMed]
- Fromm, M.F. Importance of P-glycoprotein for drug disposition in humans. Eur. J. Clin. Investig. 2003, 33, 6–9. [Google Scholar] [CrossRef] [PubMed]
- Sharom, F.J. The P-glycoprotein multidrug transporter. Essays Biochem. 2011, 50, 161–178. [Google Scholar] [CrossRef] [PubMed]
- Garg, P.; Dhakne, R.; Belekar, V. Role of breast cancer resistance protein (BCRP) as active efflux transporter on blood-brain barrier (BBB) permeability. Mol. Divers. 2015, 19, 163–172. [Google Scholar] [CrossRef]
- Afrouzian, M.; Al-Lahham, R.; Patrikeeva, S.; Xu, M.; Fokina, V.; Fischer, W.G.; Abdel-Rahman, S.Z.; Costantine, M.; Ahmed, M.S.; Nanovskaya, T. Role of the efflux transporters BCRP and MRP1 in human placental bio-disposition of pravastatin. Biochem. Pharm. 2018, 156, 467–478. [Google Scholar] [CrossRef] [PubMed]
- Koenderink, J.B.; van den Heuvel, J.J.M.W.; Bilos, A.; Vredenburg, G.; Vermeulen, N.P.E.; Russel, F.G.M. Human multidrug resistance protein 4 (MRP4) is a cellular efflux transporter for paracetamol glutathione and cysteine conjugates. Arch. Toxicol. 2020, 94, 3027–3032. [Google Scholar] [CrossRef] [PubMed]
- Huynh, T.; Norris, M.D.; Haber, M.; Henderson, M.J. ABCC4/MRP4: A MYCN-regulated transporter and potential therapeutic target in neuroblastoma. Front. Oncol. 2012, 2, 178. [Google Scholar] [CrossRef] [Green Version]
- Silva, A.C.; Oliveira, T.R.; Mamani, J.B.; Malheiros, S.M.F.; Malavolta, L.; Pavon, L.F.; Sibov, T.T.; Amaro, E., Jr.; Tannús, A.; Vidoto, E.L.G.; et al. Application of hyperthermia induced by superparamagnetic iron oxide nanoparticles in glioma treatment. Int. J. Nanomed. 2011, 6, 591–603. [Google Scholar] [CrossRef] [Green Version]
- Yang, F.-Y.; Wang, H.-E.; Lin, G.-L.; Lin, H.-H.; Wong, T.-T. Evaluation of the increase in permeability of the blood–brain barrier during tumor progression after pulsed focused ultrasound. Int. J. Nanomed. 2012, 7, 723–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, L.; Song, Q.; Bai, W.; Zhang, J.; Miao, D.; Jiang, M.; Wang, Y.; Shen, Z.; Hu, Q.; Gu, X.; et al. Facilitated brain delivery of poly (ethylene glycol)–poly (lactic acid) nanoparticles by microbubble-enhanced unfocused ultrasound. Biomaterials 2014, 35, 3384–3395. [Google Scholar] [CrossRef] [PubMed]
- Sİrav, B.; Seyhan, N. Blood-Brain Barrier Disruption by Continuous-Wave Radio Frequency Radiation. Electromagn. Biol. Med. 2009, 28, 215–222. [Google Scholar] [CrossRef]
- Zhang, F.; Xu, C.-L.; Liu, C.-M. Drug delivery strategies to enhance the permeability of the blood-brain barrier for treatment of glioma. Drug Des. Dev. 2015, 9, 2089–2100. [Google Scholar] [CrossRef] [Green Version]
- Kiessling, M.; Herchenhan, E.; Eggert, H.R. Cerebrovascular and metabolic effects on the rat brain of focal Nd:YAG laser irradiation. J. Neurosurg. 1990, 73, 909–917. [Google Scholar] [CrossRef]
- Feng, W.; Zhang, C.; Yu, T.; Semyachkina-Glushkovskaya, O.; Zhu, D. In vivo monitoring blood-brain barrier permeability using spectral imaging through optical clearing skull window. J. Biophotonics 2019, 12, e201800330. [Google Scholar] [CrossRef]
- Zou, L.-L.; Ma, J.-L.; Wang, T.; Yang, T.-B.; Liu, C.-B. Cell-penetrating Peptide-mediated therapeutic molecule delivery into the central nervous system. Curr. Neuropharmacol. 2013, 11, 197–208. [Google Scholar] [CrossRef] [PubMed]
- Khan, M.M.; Filipczak, N.; Torchilin, V.P. Cell penetrating peptides: A versatile vector for co-delivery of drug and genes in cancer. J. Control. Release 2021, 330, 1220–1228. [Google Scholar] [CrossRef]
- Zhou, X.; Smith, Q.R.; Liu, X. Brain penetrating peptides and peptide–drug conjugates to overcome the blood–brain barrier and target CNS diseases. WIREs Nanomed. Nanobiotechnol. 2021, 13, e1695. [Google Scholar] [CrossRef] [PubMed]
- Nair, K.; Ramaiyan, V.; Kumar, S. Enhancement of drug permeability across blood brain barrier using nanoparticles in meningitis. Inflammopharmacology 2018, 26, 675–684. [Google Scholar] [CrossRef] [PubMed]
- Chai, Q.; He, W.Q.; Zhou, M.; Lu, H.; Fu, Z.F. Enhancement of blood-brain barrier permeability and reduction of tight junction protein expression are modulated by chemokines/cytokines induced by rabies virus infection. J. Virol. 2014, 88, 4698–4710. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, L.N.; Ma, D.; Shui, G.; Wong, P.; Cazenave-Gassiot, A.; Zhang, X.; Wenk, M.R.; Goh, E.L.; Silver, D.L. Mfsd2a is a transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature 2014, 509, 503–506. [Google Scholar] [CrossRef]
- Hagan, N.; Ben-Zvi, A. The molecular, cellular, and morphological components of blood–brain barrier development during embryogenesis. In Seminars in Cell & Developmental Biology; Elsevier: Amsterdam, The Netherlands, 2015. [Google Scholar]
- Ju, X.; Miao, T.; Chen, H.; Ni, J.; Han, L. Overcoming Mfsd2a-Mediated Low Transcytosis to Boost Nanoparticle Delivery to Brain for Chemotherapy of Brain Metastases. Adv. Healthc. Mater. 2021, 10, 2001997. [Google Scholar] [CrossRef]
- Prokai-Tatrai, K.; Prokai, L. Prodrug Design for Brain Delivery of Small- and Medium-Sized Neuropeptides. In Neuropeptides: Methods and Protocols; Merighi, A., Ed.; Humana Press: Totowa, NJ, USA, 2011; pp. 313–336. [Google Scholar] [CrossRef]
- Alaofi, A.; Siahaan, T. Prodrug strategies for brain delivery. In Advances in Non-Invasive Drug Delivery to the Brain; Future Science: London, UK, 2015; pp. 144–156. [Google Scholar] [CrossRef]
- Prokai-Tatrai, K.; Prokai, L. A Novel Prodrug Approach for Central Nervous System-Selective Estrogen Therapy. Molecules 2019, 24, 4197. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Wang, W.; Li, L.; Perry, G.; Lee, H.-g.; Zhu, X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2014, 1842, 1240–1247. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.-Y.; Hu, J.-J.; Xu, Q.; Chen, S.; Jia, H.-Z.; Sun, Y.-X.; Zhuo, R.-X.; Zhang, X.-Z. A redox-responsive drug delivery system based on RGD containing peptide-capped mesoporous silica nanoparticles. J. Mater. Chem. B 2015, 3, 39–44. [Google Scholar] [CrossRef]
- Alessandrini, M.; Preynat-Seauve, O.; De Bruin, K.; Pepper, M.S. Stem cell therapy for neurological disorders. S. Afr. Med. J. 2019, 109, S71–S78. [Google Scholar] [CrossRef]
- Altanerova, V.; Cihova, M.; Babic, M.; Rychly, B.; Ondicova, K.; Mravec, B.; Altaner, C. Human adipose tissue-derived mesenchymal stem cells expressing yeast cytosinedeaminase: Uracil phosphoribosyltransferase inhibit intracerebral rat glioblastoma. Int. J. Cancer 2012, 130, 2455–2463. [Google Scholar] [CrossRef]
- Cristofanilli, M.; Harris, V.K.; Zigelbaum, A.; Goossens, A.M.; Lu, A.; Rosenthal, H.; Sadiq, S.A. Mesenchymal stem cells enhance the engraftment and myelinating ability of allogeneic oligodendrocyte progenitors in dysmyelinated mice. Stem Cells Dev. 2011, 20, 2065–2076. [Google Scholar] [CrossRef]
- Shigematsu, K.; Takeda, T.; Komori, N.; Tahara, K.; Yamagishi, H. Hypothesis: Intravenous administration of mesenchymal stem cells is effective in the treatment of Alzheimer’s disease. Med. Hypotheses 2021, 150, 110572. [Google Scholar] [CrossRef]
- Hladky, S.B.; Barrand, M.A. The glymphatic hypothesis: The theory and the evidence. Fluids Barriers CNS 2022, 19, 9. [Google Scholar] [CrossRef]
- Liu, J.; Guo, Y.; Zhang, C.; Zeng, Y.; Luo, Y.; Wang, G. Clearance Systems in the Brain, From Structure to Function. Front. Cell Neurosci 2021, 15, 729706. [Google Scholar] [CrossRef]
- Nedergaard, M.; Goldman, S.A. Glymphatic failure as a final common pathway to dementia. Science 2020, 370, 50–56. [Google Scholar] [CrossRef]
- Benveniste, H.; Liu, X.; Koundal, S.; Sanggaard, S.; Lee, H.; Wardlaw, J. The glymphatic system and waste clearance with brain aging: A review. Gerontology 2019, 65, 106–119. [Google Scholar] [CrossRef]
- Rasmussen, M.K.; Mestre, H.; Nedergaard, M. The glymphatic pathway in neurological disorders. Lancet Neurol. 2018, 17, 1016–1024. [Google Scholar] [CrossRef] [Green Version]
- Pardridge, W.M. CSF, blood-brain barrier, and brain drug delivery. Expert Opin. Drug Deliv. 2016, 13, 963–975. [Google Scholar] [CrossRef]
- Hammarlund-Udenaes, M.; Fridén, M.; Syvänen, S.; Gupta, A. On the rate and extent of drug delivery to the brain. Pharm. Res. 2008, 25, 1737–1750. [Google Scholar] [CrossRef] [Green Version]
- Han, L. Modulation of the Blood–Brain Barrier for Drug Delivery to Brain. Pharmaceutics 2021, 13, 2024. [Google Scholar]
- Han, L.; Jiang, C. Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharm. Sin. B 2021, 11, 2306–2325. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pawar, B.; Vasdev, N.; Gupta, T.; Mhatre, M.; More, A.; Anup, N.; Tekade, R.K. Current Update on Transcellular Brain Drug Delivery. Pharmaceutics 2022, 14, 2719. https://doi.org/10.3390/pharmaceutics14122719
Pawar B, Vasdev N, Gupta T, Mhatre M, More A, Anup N, Tekade RK. Current Update on Transcellular Brain Drug Delivery. Pharmaceutics. 2022; 14(12):2719. https://doi.org/10.3390/pharmaceutics14122719
Chicago/Turabian StylePawar, Bhakti, Nupur Vasdev, Tanisha Gupta, Mahi Mhatre, Anand More, Neelima Anup, and Rakesh Kumar Tekade. 2022. "Current Update on Transcellular Brain Drug Delivery" Pharmaceutics 14, no. 12: 2719. https://doi.org/10.3390/pharmaceutics14122719
APA StylePawar, B., Vasdev, N., Gupta, T., Mhatre, M., More, A., Anup, N., & Tekade, R. K. (2022). Current Update on Transcellular Brain Drug Delivery. Pharmaceutics, 14(12), 2719. https://doi.org/10.3390/pharmaceutics14122719