Engineered nanomaterials that exploit blood-brain barrier dysfunction for delivery to the brain

Advanced Drug Delivery Reviews - Tập 197 - Trang 114820 - 2023
Jason R. Wu1, Yazmin Hernandez1, Katelyn F. Miyasaki1, Ester J. Kwon1,2
1Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
2Sanford Consortium for Regenerative Medicine, United States

Tài liệu tham khảo

Pardridge, 2012, Drug transport across the blood-brain barrier, J. Cereb. Blood Flow Metab., 32, 1959, 10.1038/jcbfm.2012.126 Wong, 2013, The blood-brain barrier: an engineering perspective, Front. Neuroeng., 6, 10.3389/fneng.2013.00007 Kadry, 2020, A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity, Fluids Barriers CNS, 17, 69, 10.1186/s12987-020-00230-3 Abbott, 2010, Structure and function of the blood–brain barrier, Neurobiol. Dis., 37, 13, 10.1016/j.nbd.2009.07.030 Daneman, 2012, The blood-brain barrier in health and disease, Ann. Neurol., 72, 648, 10.1002/ana.23648 Zlokovic, 2008, The blood-brain barrier in health and chronic neurodegenerative disorders, Neuron, 57, 178, 10.1016/j.neuron.2008.01.003 Walter, 2021, Surface charge, glycocalyx, and blood-brain barrier function, Tissue Barriers, 9, 1904773, 10.1080/21688370.2021.1904773 Helms, 2016, In vitro models of the blood–brain barrier: an overview of commonly used brain endothelial cell culture models and guidelines for their use, J. Cereb. Blood Flow Metab., 36, 862, 10.1177/0271678X16630991 Naik, 2012, In vitro blood-brain barrier models: current and perspective technologies, J. Pharm. Sci., 101, 1337, 10.1002/jps.23022 Bergers, 2005, The role of pericytes in blood-vessel formation and maintenance, Neuro Oncol., 7, 452, 10.1215/S1152851705000232 Brown, 2019, Pericytes and neurovascular function in the healthy and diseased brain, Front. Cell. Neurosci., 13, 282, 10.3389/fncel.2019.00282 A. Ben-Zvi et al., Mfsd2a is critical for the formation and function of the blood–brain barrier, Nature 509 (7501) 507–511, 2014, doi: 10.1038/nature13324. In this paper, the function of mfsd2a in the BBB was identified for the first time. Mfsd2a is highly upregulated specifically in brain vasculature over peripheral vasculature and has a role in suppressing transcytosis in the BBB. Obermeier, 2013, Development, maintenance and disruption of the blood-brain barrier, Nat. Med., 19, 1584, 10.1038/nm.3407 Kutuzov, 2018, Contributions of the glycocalyx, endothelium, and extravascular compartment to the blood–brain barrier, PNAS, 115, 10.1073/pnas.1802155115 Luissint, 2012, Tight junctions at the blood brain barrier: physiological architecture and disease-associated dysregulation, Fluids Barriers CNS, 9, 23, 10.1186/2045-8118-9-23 Liu, 2012, Tight junction in blood-brain barrier: an overview of structure, regulation, and regulator substances: tight junction in blood-brain barrier, CNS Neurosci. Ther., 18, 609, 10.1111/j.1755-5949.2012.00340.x Stamatovic, 2016, Junctional proteins of the blood-brain barrier: new insights into function and dysfunction, Tissue Barriers, 4, e1154641, 10.1080/21688370.2016.1154641 Gomez-Zepeda, 2019, ABC transporters at the blood-brain interfaces, their study models, and drug delivery implications in gliomas, Pharmaceutics, 12, 20, 10.3390/pharmaceutics12010020 Jones, 2007, Blood–brain barrier transport of therapeutics via receptor-mediation, Pharm. Res., 24, 1759, 10.1007/s11095-007-9379-0 Patching, 2017, Glucose transporters at the blood-brain barrier: function, regulation and gateways for drug delivery, Mol. Neurobiol., 54, 1046, 10.1007/s12035-015-9672-6 Pulgar, 2019, Transcytosis to cross the blood brain barrier, new advancements and challenges, Front. Neurosci., 12, 1019, 10.3389/fnins.2018.01019 Gray, 2017, Unravelling the regulation of insulin transport across the brain endothelial cell, Diabetologia, 60, 1512, 10.1007/s00125-017-4285-4 Roberts, 1993, Receptor-mediated endocytosis of transferrin at the blood-brain barrier, J. Cell Sci., 104, 521, 10.1242/jcs.104.2.521 Hervé, 2008, CNS delivery via adsorptive transcytosis, AAPS J., 10, 455, 10.1208/s12248-008-9055-2 Bickel, 2001, Delivery of peptides and proteins through the blood–brain barrier, Adv. Drug Deliv. Rev., 46, 247, 10.1016/S0169-409X(00)00139-3 Thöle, 2002, Uptake of cationized albumin coupled liposomes by cultured porcine brain microvessel endothelial cells and intact brain capillaries, J. Drug Target., 10, 337, 10.1080/10611860290031840 Andreone, 2017, Blood-brain barrier permeability is regulated by lipid transport-dependent suppression of caveolae-mediated transcytosis, Neuron, 94, 581, 10.1016/j.neuron.2017.03.043 Ng, 2019, Traumatic brain injuries: pathophysiology and potential therapeutic targets, Front. Cell. Neurosci., 13, 528, 10.3389/fncel.2019.00528 Achar, 2021, Drug delivery challenges in brain disorders across the blood-brain barrier: novel methods and future considerations for improved therapy, Biomedicines, 9, 1834, 10.3390/biomedicines9121834 Yoo, 2019, Quantitative dynamic contrast-enhanced MR imaging shows widespread blood-brain barrier disruption in mild traumatic brain injury patients with post-concussion syndrome, Eur. Radiol., 29, 1308, 10.1007/s00330-018-5656-z Johnson, 2018, Mechanical disruption of the blood-brain barrier following experimental concussion, Acta Neuropathol., 135, 711, 10.1007/s00401-018-1824-0 Tagge, 2018, Concussion, microvascular injury, and early tauopathy in young athletes after impact head injury and an impact concussion mouse model, Brain, 141, 422, 10.1093/brain/awx350 Hay, 2015, Blood-brain barrier disruption is an early event that may persist for many years after traumatic brain injury in humans, J. Neuropathol. Exp. Neurol., 74, 1147 Tomkins, 2011, Blood-brain barrier breakdown following traumatic brain injury: a possible role in posttraumatic epilepsy, Cardiovasc. Psychiatry Neurol., 2011, 1, 10.1155/2011/765923 Sandsmark, 2019, Cerebral microvascular injury: a potentially treatable endophenotype of traumatic brain injury-induced neurodegeneration, Neuron, 103, 367, 10.1016/j.neuron.2019.06.002 Bashir, 2020, Increased severity of the CHIMERA model induces acute vascular injury, sub-acute deficits in memory recall, and chronic white matter gliosis, Exp. Neurol., 324, 10.1016/j.expneurol.2019.113116 Ichkova, 2020, Early cerebrovascular and long-term neurological modifications ensue following juvenile mild traumatic brain injury in male mice, Neurobiol. Dis., 141, 10.1016/j.nbd.2020.104952 Chodobski, 2011, Blood-brain barrier pathophysiology in traumatic brain injury, Transl. Stroke Res., 2, 492, 10.1007/s12975-011-0125-x Minta, 2020, Dynamics of cerebrospinal fluid levels of matrix metalloproteinases in human traumatic brain injury, Sci. Rep., 10, 18075, 10.1038/s41598-020-75233-z Kempuraj, 2020, Mast cell activation, neuroinflammation, and tight junction protein derangement in acute traumatic brain injury, Mediators Inflamm., 2020, 1, 10.1155/2020/4243953 E.J. Kwon, M. Skalak, R. Lo Bu, S.N. Bhatia, Neuron-targeted nanoparticle for sirna delivery to traumatic brain injuries, ACS Nano 10 (8) (2016) 7926–7933, doi: 10.1021/acsnano.6b03858. Simon, 2017, The far-reaching scope of neuroinflammation after traumatic brain injury, Nat. Rev. Neurol., 13, 171, 10.1038/nrneurol.2017.13 Nian, 2020, Blood-Brain Barrier Damage in Ischemic Stroke and Its Regulation by Endothelial Mechanotransduction, Front. Physiol., 11, 10.3389/fphys.2020.605398 Bernardo-Castro, 2020, Pathophysiology of blood-brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery, Front. Neurol., 11, 10.3389/fneur.2020.594672 Okada, 2020, The stroke-induced blood-brain barrier disruption: current progress of inspection technique, mechanism, and therapeutic target, CN, 18, 1187, 10.2174/1570159X18666200528143301 Shi, 2016, Rapid endothelial cytoskeletal reorganization enables early blood-brain barrier disruption and long-term ischaemic reperfusion brain injury, Nat. Commun., 7, 10523, 10.1038/ncomms10523 Knowland, 2014, Stepwise recruitment of transcellular and paracellular pathways underlies blood-brain barrier breakdown in stroke, Neuron, 82, 603, 10.1016/j.neuron.2014.03.003 Liu, 2018, Time-course investigation of blood–brain barrier permeability and tight junction protein changes in a rat model of permanent focal ischemia, J. Physiol. Sci., 68, 121, 10.1007/s12576-016-0516-6 Merali, 2017, Evolution of blood-brain-barrier permeability after acute ischemic stroke, PLoS One, 12, e0171558, 10.1371/journal.pone.0171558 Li, 2018, The peripheral immune response after stroke—A double edge sword for blood-brain barrier integrity, CNS Neurosci. Ther., 24, 1115, 10.1111/cns.13081 Zhang, 2018, Autophagy- and MMP-2/9-mediated reduction and redistribution of ZO-1 contribute to hyperglycemia-increased blood-brain barrier permeability during early reperfusion in stroke, Neuroscience, 377, 126, 10.1016/j.neuroscience.2018.02.035 Liu, 2018, Cytokine MIF enhances blood-brain barrier permeability: impact for therapy in ischemic stroke, Sci. Rep., 8, 743, 10.1038/s41598-017-16927-9 Revuelta, 2019, Ischemic stroke in neonatal and adult astrocytes, Mech. Ageing Dev., 183, 10.1016/j.mad.2019.111147 K. Schoknecht et al., Monitoring stroke progression: in vivo imaging of cortical perfusion, blood—brain barrier permeability and cellular damage in the rat photothrombosis model, J. Cereb. Blood Flow Metab. 34 (11) (2014) 1791–1801, doi: 10.1038/jcbfm.2014.147. D’Souza, 2021, Targeting the blood-brain barrier for the delivery of stroke therapies, Adv. Drug Deliv. Rev., 171, 332, 10.1016/j.addr.2021.01.015 Lochhead, 2020, Structure, function, and regulation of the blood-brain barrier tight junction in central nervous system disorders, Front. Physiol., 11, 914, 10.3389/fphys.2020.00914 Kelleher, 2013, Evidence of endothelial dysfunction in the development of Alzheimer’s disease: Is Alzheimer’s a vascular disorder?, Am. J. Cardiovasc. Dis., 3, 197 Narayan, 2015, Assessing fibrinogen extravasation into Alzheimer’s disease brain using high-content screening of brain tissue microarrays, J. Neurosci. Methods, 247, 41, 10.1016/j.jneumeth.2015.03.017 Llorens, 2020, Plasma total prion protein as a potential biomarker for neurodegenerative dementia: diagnostic accuracy in the spectrum of prion diseases, Neuropathol. Appl. Neurobiol., 46, 240, 10.1111/nan.12573 van de Haar, 2016, Blood-brain barrier leakage in patients with early Alzheimer disease, Radiology, 281, 527, 10.1148/radiol.2016152244 Kook, 2012, A 1–42-RAGE interaction disrupts tight junctions of the blood-brain barrier via Ca2+-calcineurin signaling, J. Neurosci., 32, 8845, 10.1523/JNEUROSCI.6102-11.2012 Keaney, 2015, Autoregulated paracellular clearance of amyloid-β across the blood-brain barrier, Sci. Adv., 1, e1500472, 10.1126/sciadv.1500472 Yamazaki, 2019, Selective loss of cortical endothelial tight junction proteins during Alzheimer’s disease progression, Brain, 142, 1077, 10.1093/brain/awz011 Liu, 2020, Tau and apolipoprotein E modulate cerebrovascular tight junction integrity independent of cerebral amyloid angiopathy in Alzheimer’s disease, Alzheimer’s Dementia, 16, 1372, 10.1002/alz.12104 Nehra, 2022, Blood-brain barrier leakage in Alzheimer’s disease: from discovery to clinical relevance, Pharmacol. Ther., 234, 10.1016/j.pharmthera.2022.108119 Uemura, 2020, Brain microvascular pericytes in vascular cognitive impairment and dementia, Front. Aging Neurosci., 12, 80, 10.3389/fnagi.2020.00080 Halliday, 2016, Accelerated pericyte degeneration and blood–brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease, J. Cereb. Blood Flow Metab., 36, 216, 10.1038/jcbfm.2015.44 Giannoni, 2016, Cerebrovascular pathology during the progression of experimental Alzheimer’s disease, Neurobiol. Dis., 88, 107, 10.1016/j.nbd.2016.01.001 Sengillo, 2013, Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer’s disease: pericytes in Alzheimer’s disease, Brain Pathol., 23, 303, 10.1111/bpa.12004 Oikari, 2020, Altered brain endothelial cell phenotype from a familial alzheimer mutation and its potential implications for amyloid clearance and drug delivery, Stem Cell Rep., 14, 924, 10.1016/j.stemcr.2020.03.011 Tao, 2022, Discerning the role of blood brain barrier dysfunction in Alzheimer’s disease, Aging Dis., 13, 1391, 10.14336/AD.2022.0130-1 Nation, 2019, Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction, Nat. Med., 25, 270, 10.1038/s41591-018-0297-y Alajangi, 2022, Blood–brain barrier: emerging trends on transport models and new-age strategies for therapeutics intervention against neurological disorders, Mol. Brain, 15, 49, 10.1186/s13041-022-00937-4 Pisani, 2012, Increased blood-cerebrospinal fluid transfer of albumin in advanced Parkinson’s disease, J. Neuroinflammation, 9, 670, 10.1186/1742-2094-9-188 Gray, 2015, Striatal blood-brain barrier permeability in Parkinson’S disease, J. Cereb. Blood Flow Metab., 35, 747, 10.1038/jcbfm.2015.32 Pienaar, 2015, Deep-brain stimulation associates with improved microvascular integrity in the subthalamic nucleus in Parkinson’s disease, Neurobiol. Dis., 74, 392, 10.1016/j.nbd.2014.12.006 Huang, 2016, β -asarone and levodopa co-administration increase striatal dopamine level in 6-hydroxydopamine induced rats by modulating P-glycoprotein and tight junction proteins at the blood-brain barrier and promoting levodopa into the brain, Clin. Exp. Pharmacol. Physiol., 43, 634, 10.1111/1440-1681.12570 Al-Bachari, 2020, Blood–brain barrier leakage is increased in Parkinson’s disease, Front. Physiol., 11, 10.3389/fphys.2020.593026 Zhao, 2007, TNF-α knockout and minocycline treatment attenuates blood–brain barrier leakage in MPTP-treated mice, Neurobiol. Dis., 26, 36, 10.1016/j.nbd.2006.11.012 Droździk, 2003, Polymorphism in the P-glycoprotein drug transporter MDR1 gene: a possible link between environmental and genetic factors in Parkinson’s disease, Pharmacogenetics, 13, 259, 10.1097/00008571-200305000-00004 Kortekaas, 2005, Blood-brain barrier dysfunction in parkinsonian midbrain in vivo, Ann. Neurol., 57, 176, 10.1002/ana.20369 Arvold, 2016, Updates in the management of brain metastases, Neuro Oncol., 18, 1043, 10.1093/neuonc/now127 Stelzer, 2013, Epidemiology and prognosis of brain metastases, Surg. Neurol. Int., 4, 192, 10.4103/2152-7806.111296 Davis, 2012, Toward determining the lifetime occurrence of metastatic brain tumors estimated from 2007 United States cancer incidence data, Neuro Oncol., 14, 1171, 10.1093/neuonc/nos152 Gerstner, 2009, VEGF inhibitors in the treatment of cerebral edema in patients with brain cancer, Nat. Rev. Clin. Oncol., 6, 229, 10.1038/nrclinonc.2009.14 Jain, 2014, Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia, Cancer Cell, 26, 605, 10.1016/j.ccell.2014.10.006 Yuan, 1996, Time-dependent vascular regression and permeability changes in established human tumor xenografts induced by an anti-vascular endothelial growth factor/vascular permeability factor antibody, PNAS, 93, 14765, 10.1073/pnas.93.25.14765 Argaw, 2009, VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown, PNAS, 106, 1977, 10.1073/pnas.0808698106 Wen, 2017, VEGF-mediated tight junctions pathological fenestration enhances doxorubicin-loaded glycolipid-like nanoparticles traversing BBB for glioblastoma-targeting therapy, Drug Deliv., 24, 1843, 10.1080/10717544.2017.1386731 Akter, 2021, Pre-clinical tumor models of primary brain tumors: challenges and opportunities, Biochim. Biophys. Acta (BBA) – Rev. Cancer, 1875 Sarkaria, 2018, Is the blood–brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data, Neuro Oncol., 20, 184, 10.1093/neuonc/nox175 de Gooijer, 2021, ATP-binding cassette transporters restrict drug delivery and efficacy against brain tumors even when blood-brain barrier integrity is lost, Cell Reports Med., 2, 10.1016/j.xcrm.2020.100184 Mason, 2015, Blood-brain barrier-associated efflux transporters: a significant but underappreciated obstacle to drug development in glioblastoma, Neuro Oncol., p. nov122, 10.1093/neuonc/nov122 Cheng, 2013, Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth, Cell, 153, 139, 10.1016/j.cell.2013.02.021 Brandao, 2019, Astrocytes, the rising stars of the glioblastoma microenvironment, Glia, 67, 779, 10.1002/glia.23520 Sarin, 2008, Effective transvascular delivery of nanoparticles across the blood-brain tumor barrier into malignant glioma cells, J. Transl. Med., 6, 80, 10.1186/1479-5876-6-80 Yonemori, 2010, Disruption of the blood brain barrier by brain metastases of triple-negative and basal-type breast cancer but not HER2/neu -positive breast cancer, Cancer, 116, 302, 10.1002/cncr.24735 Arvanitis, 2020, The blood–brain barrier and blood–tumour barrier in brain tumours and metastases, Nat. Rev. Cancer, 20, 26, 10.1038/s41568-019-0205-x Başkaya, 1997, The biphasic opening of the blood–brain barrier in the cortex and hippocampus after traumatic brain injury in rats, Neurosci. Lett., 226, 33, 10.1016/S0304-3940(97)00239-5 Hatashita, 1990, Brain edema and cerebrovascular permeability during cerebral ischemia in rats, Stroke, 21, 582, 10.1161/01.STR.21.4.582 Strbian, 2008, The blood–brain barrier is continuously open for several weeks following transient focal cerebral ischemia, Neuroscience, 153, 175, 10.1016/j.neuroscience.2008.02.012 Bharadwaj, 2020, Sex-dependent macromolecule and nanoparticle delivery in experimental brain injury, Tissue Eng. A, 26, 688, 10.1089/ten.tea.2020.0040 Matsumura, 1986, A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs, Cancer Res., 46, 6387 Fang, 2011, The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect, Adv. Drug Deliv. Rev., 63, 136, 10.1016/j.addr.2010.04.009 Ishii, 2012, Amelioration of cerebral ischemia–reperfusion injury based on liposomal drug delivery system with asialo-erythropoietin, J. Control. Release, 160, 81, 10.1016/j.jconrel.2012.02.004 Boyd, 2015, Traumatic brain injury opens blood–brain barrier to stealth liposomes via an enhanced permeability and retention (EPR)-like effect, J. Drug Target., 23, 847, 10.3109/1061186X.2015.1034280 Hersh, 2022, Crossing the blood-brain barrier: advances in nanoparticle technology for drug delivery in neuro-oncology, IJMS, 23, 4153, 10.3390/ijms23084153 Ribovski, 2021, Polymeric nanoparticles properties and brain delivery, Pharmaceutics, 13, 2045, 10.3390/pharmaceutics13122045 Nowak, 2020, Size, shape, and flexibility influence nanoparticle transport across brain endothelium under flow, Bioeng. Transl. Med., 5, 10.1002/btm2.10153 De Jong, 2008, Particle size-dependent organ distribution of gold nanoparticles after intravenous administration, Biomaterials, 29, 1912, 10.1016/j.biomaterials.2007.12.037 Cruz, 2016, Effect of PLGA NP size on efficiency to target traumatic brain injury, J. Control. Release, 223, 31, 10.1016/j.jconrel.2015.12.029 Bharadwaj, 2016, Temporal assessment of nanoparticle accumulation after experimental brain injury: effect of particle size, Sci. Rep., 6, 29988, 10.1038/srep29988 Cai, 2016, Biocompatible red fluorescent organic nanoparticles with tunable size and aggregation-induced emission for evaluation of blood-brain barrier damage, Adv. Mater., 28, 8760, 10.1002/adma.201601191 Thorne, 2006, In vivo diffusion analysis with quantum dots and dextrans predicts the width of brain extracellular space, PNAS, 103, 5567, 10.1073/pnas.0509425103 Hobbs, 1998, Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment, PNAS, 95, 4607, 10.1073/pnas.95.8.4607 Petros, 2010, Strategies in the design of nanoparticles for therapeutic applications, Nat. Rev. Drug Discov., 9, 615, 10.1038/nrd2591 Brown, 2020, Effect of nanoparticle composition, size, shape, and stiffness on penetration across the blood-brain barrier, ACS Biomater. Sci. Eng., 6, 4916, 10.1021/acsbiomaterials.0c00743 Decuzzi, 2010, Size and shape effects in the biodistribution of intravascularly injected particles, J. Control. Release, 141, 320, 10.1016/j.jconrel.2009.10.014 Champion, 2007, Making polymeric micro- and nanoparticles of complex shapes, PNAS, 104, 11901, 10.1073/pnas.0705326104 Kolhar, 2013, Using shape effects to target antibody-coated nanoparticles to lung and brain endothelium, PNAS, 110, 10753, 10.1073/pnas.1308345110 Da Silva-Candal, 2019, Shape effect in active targeting of nanoparticles to inflamed cerebral endothelium under static and flow conditions, J. Control. Release, 309, 94, 10.1016/j.jconrel.2019.07.026 Mailänder, 2009, Interaction of nanoparticles with cells, Biomacromolecules, 10, 2379, 10.1021/bm900266r Blanco, 2015, Principles of nanoparticle design for overcoming biological barriers to drug delivery, Nat. Biotechnol., 33, 941, 10.1038/nbt.3330 Moscariello, 2019, Unraveling in vivo brain transport of protein-coated fluorescent nanodiamonds, Small, 15, 1902992, 10.1002/smll.201902992 Lockman, 2004, Nanoparticle surface charges alter blood-brain barrier integrity and permeability, J. Drug Target., 12, 635, 10.1080/10611860400015936 Tenzer, 2013, Rapid formation of plasma protein corona critically affects nanoparticle pathophysiology, Nat. Nanotech, 8, 772, 10.1038/nnano.2013.181 Sachais, 2002, Platelet factor 4 binds to low-density lipoprotein receptors and disrupts the endocytic itinerary, resulting in retention of low-density lipoprotein on the cell surface, Blood, 99, 3613, 10.1182/blood.V99.10.3613 Z.P. Lin, W. Ngo, S.M. Mladjenovic, J.L.Y. Wu, W.C.W. Chan, Nanoparticles bind to endothelial cells in injured blood vessels via a transient protein corona, Nano Lett. (2023) p. acs.nanolett.2c04501, doi: 10.1021/acs.nanolett.2c04501. This recent work investigates the interaction of nanoparticles with protein coronas containing platelet factors and heparan sulfate proteoglycans on the surface of endothelial cells. Waggoner, 2021, Pharmacokinetic analysis of peptide-modified nanoparticles with engineered physicochemical properties in a mouse model of traumatic brain injury, AAPS J., 23, 100, 10.1208/s12248-021-00626-5 Nance, 2012, A dense poly(ethylene glycol) coating improves penetration of large polymeric nanoparticles within brain tissue, Sci. Transl. Med., 4, 10.1126/scitranslmed.3003594 Mishra, 2004, PEGylation significantly affects cellular uptake and intracellular trafficking of non-viral gene delivery particles, Eur. J. Cell Biol., 83, 97, 10.1078/0171-9335-00363 Suk, 2016, PEGylation as a strategy for improving nanoparticle-based drug and gene delivery, Adv. Drug Deliv. Rev., 99, 28, 10.1016/j.addr.2015.09.012 Koffie, 2011, Nanoparticles enhance brain delivery of blood–brain barrier-impermeable probes for in vivo optical and magnetic resonance imaging, PNAS, 108, 18837, 10.1073/pnas.1111405108 W. Li et al., BBB pathophysiology–independent delivery of siRNA in traumatic brain injury, Sci. Adv. 7 (1) (2021) eabd6889, doi: 10.1126/sciadv.abd6889. In this work, multiple surface chemistries were compared for nanomaterial uptake in the brain after traumatic brain injury. Interestingly, they observed polysorbate 80 coated nanomaterials could still transport into the brain, even 2 weeks after injury. Qin, 2023, Ultrasound nanotheranostics: toward precision medicine, J. Control. Release, 353, 105, 10.1016/j.jconrel.2022.11.021 Ohta, 2020, Investigating the optimum size of nanoparticles for their delivery into the brain assisted by focused ultrasound-induced blood–brain barrier opening, Sci. Rep., 10, 18220, 10.1038/s41598-020-75253-9 Curley, 2020, Augmentation of brain tumor interstitial flow via focused ultrasound promotes brain-penetrating nanoparticle dispersion and transfection, Sci. Adv., 6, eaay1344, 10.1126/sciadv.aay1344 Ogawa, 2022, Focused ultrasound/microbubbles-assisted BBB opening enhances LNP-mediated mRNA delivery to brain, J. Control. Release, 348, 34, 10.1016/j.jconrel.2022.05.042 Liu, 2022, Strategies to overcome/penetrate the BBB for systemic nanoparticle delivery to the brain/brain tumor, Adv. Drug Deliv. Rev., 191, 10.1016/j.addr.2022.114619 Gorick, 2020, Sonoselective transfection of cerebral vasculature without blood–brain barrier disruption, PNAS, 117, 5644, 10.1073/pnas.1914595117 Caspani, 2020, Magnetic nanomaterials as contrast agents for MRI, Materials (Basel), 13, 2586, 10.3390/ma13112586 Felfoul, 2016, Simultaneous steering and imaging of magnetic particles using MRI toward delivery of therapeutics, Sci. Rep., 6, 33567, 10.1038/srep33567 Israel, 2020, Magnetic iron oxide nanoparticles for imaging, targeting and treatment of primary and metastatic tumors of the brain, J. Control. Release, 320, 45, 10.1016/j.jconrel.2020.01.009 Qiu, 2017, Magnetic forces enable controlled drug delivery by disrupting endothelial cell-cell junctions, Nat. Commun., 8, 15594, 10.1038/ncomms15594 Heydarheydari, 2021, Pulsed high magnetic field-induced reversible blood-brain barrier permeability to enhance brain-targeted drug delivery, Electromagn. Biol. Med., 40, 361, 10.1080/15368378.2021.1925905 Li, 2021, Reversibly modulating the blood-brain barrier by laser stimulation of molecular-targeted nanoparticles, Nano Lett., 21, 9805, 10.1021/acs.nanolett.1c02996 Li, 2023, Mechanobiological modulation of blood–brain barrier permeability by laser stimulation of endothelial-targeted nanoparticles, Nanoscale, 15, 3387, 10.1039/D2NR05062E Salcman, 1983, Interstitial microwave hyperthermia for brain tumors, J Neuro-Oncol, 1, 225, 10.1007/BF00165607 Kiyatkin, 2009, Permeability of the blood–brain barrier depends on brain temperature, Neuroscience, 161, 926, 10.1016/j.neuroscience.2009.04.004 Patel, 2020, The effect of thermal therapy on the blood-brain barrier and blood-tumor barrier, Int. J. Hyperth., 37, 35, 10.1080/02656736.2020.1783461 Tabatabaei, 2015, Remote control of the permeability of the blood-brain barrier by magnetic heating of nanoparticles: a proof of concept for brain drug delivery, J. Control. Release, 206, 49, 10.1016/j.jconrel.2015.02.027 Yarmush, 2014, Electroporation-based technologies for medicine: principles, applications, and challenges, Annu. Rev. Biomed. Eng., 16, 295, 10.1146/annurev-bioeng-071813-104622 Sharabi, 2020, The effects of point-source electroporation on the blood-brain barrier and brain vasculature in rats: An MRI and histology study, Bioelectrochemistry, 134, 10.1016/j.bioelechem.2020.107521 Lorenzo, 2019, Temporal characterization of blood-brain barrier disruption with high-frequency electroporation, Cancers (Basel), 11, 1850, 10.3390/cancers11121850 Sharabi, 2021, Non-invasive low pulsed electrical fields for inducing BBB disruption in mice-feasibility demonstration, Pharmaceutics, 13, 169, 10.3390/pharmaceutics13020169 Hersh, 2016, Evolving drug delivery strategies to overcome the blood brain barrier, Curr. Pharm. Des., 22, 1177, 10.2174/1381612822666151221150733 Rapoport, 2000, Osmotic opening of the blood-brain barrier: principles, mechanism, and therapeutic applications, Cell. Mol. Neurobiol., 20, 217, 10.1023/A:1007049806660 Linville, 2020, Modeling hyperosmotic blood–brain barrier opening within human tissue-engineered in vitro brain microvessels, J. Cereb. Blood Flow Metab., 40, 1517, 10.1177/0271678X19867980 Pandit, 2020, The blood-brain barrier: physiology and strategies for drug delivery, Adv. Drug Deliv. Rev., 165–166, 1, 10.1016/j.addr.2019.11.009 Foley, 2014, Intra-arterial delivery of AAV vectors to the mouse brain after mannitol mediated blood brain barrier disruption, J. Control. Release, 196, 71, 10.1016/j.jconrel.2014.09.018 Chu, 2020, Optimization of osmotic blood-brain barrier opening to enable intravital microscopy studies on drug delivery in mouse cortex, J. Control. Release, 317, 312, 10.1016/j.jconrel.2019.11.019 Burks, 2021, Blood–brain barrier opening by intracarotid artery hyperosmolar mannitol induces sterile inflammatory and innate immune responses, PNAS, 118, 10.1073/pnas.2021915118 Chu, 2022, Hyperosmolar blood–brain barrier opening using intra-arterial injection of hyperosmotic mannitol in mice under real-time MRI guidance, Nat. Protoc., 17, 76, 10.1038/s41596-021-00634-x Zawadzki, 2019, Real-time MRI guidance for intra-arterial drug delivery in a patient with a brain tumor: technical note, BMJ Case Rep., 12, 10.1136/bcr-2018-014469 Ruoslahti, 2022, Molecular ZIP codes in targeted drug delivery, PNAS, 119, 10.1073/pnas.2200183119 Tang, 2019, A ligand motif enables differential vascular targeting of endothelial junctions between brain and retina, PNAS, 116, 2300, 10.1073/pnas.1809483116 Anthony, 2021, Targeting receptor-ligand chemistry for drug delivery across blood-brain barrier in brain diseases, Life Sci., 274, 10.1016/j.lfs.2021.119326 Pardridge, 2006, Molecular Trojan horses for blood–brain barrier drug delivery, Curr. Opin. Pharmacol., 6, 494, 10.1016/j.coph.2006.06.001 Lajoie, 2015, Targeting receptor-mediated transport for delivery of biologics across the blood-brain barrier, Annu. Rev. Pharmacol. Toxicol., 55, 613, 10.1146/annurev-pharmtox-010814-124852 Wiley, 2013, Transcytosis and brain uptake of transferrin-containing nanoparticles by tuning avidity to transferrin receptor, PNAS, 110, 8662, 10.1073/pnas.1307152110 Munji, 2019, Profiling the mouse brain endothelial transcriptome in health and disease models reveals a core blood-brain barrier dysfunction module, Nat. Neurosci., 22, 1892, 10.1038/s41593-019-0497-x DiSabato, 2016, Neuroinflammation: the devil is in the details, J. Neurochem., 139, 136, 10.1111/jnc.13607 Kwon, 2020, Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes, Transl. Neurodegener, 9, 42, 10.1186/s40035-020-00221-2 Jayaraj, 2019, Neuroinflammation: friend and foe for ischemic stroke, J. Neuroinflammation, 16, 142, 10.1186/s12974-019-1516-2 Schimmel, 2017, Neuroinflammation in traumatic brain injury: a chronic response to an acute injury, Brain Circ, 3, 135, 10.4103/bc.bc_18_17 Carvalho-Tavares, 2000, A role for platelets and endothelial selectins in tumor necrosis factor-α–induced leukocyte recruitment in the brain microvasculature, Circ. Res., 87, 1141, 10.1161/01.RES.87.12.1141 Mitroulis, 2015, Leukocyte integrins: role in leukocyte recruitment and as therapeutic targets in inflammatory disease, Pharmacol. Ther., 147, 123, 10.1016/j.pharmthera.2014.11.008 Bui, 2020, ICAM-1: a master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis, J. Leukoc. Biol., 108, 787, 10.1002/JLB.2MR0220-549R Kong, 2018, Emerging roles of vascular cell adhesion molecule-1 (VCAM-1) in immunological disorders and cancer, IJMS, 19, 1057, 10.3390/ijms19041057 Ferber, 2017, Co-targeting the tumor endothelium and P-selectin-expressing glioblastoma cells leads to a remarkable therapeutic outcome, Elife, 6, e25281, 10.7554/eLife.25281 Marcos-Contreras, 2019, Combining vascular targeting and the local first pass provides 100-fold higher uptake of ICAM-1-targeted vs untargeted nanocarriers in the inflamed brain, J. Control. Release, 301, 54, 10.1016/j.jconrel.2019.03.008 Hsu, 2014, Specific binding, uptake, and transport of ICAM-1-targeted nanocarriers across endothelial and subendothelial cell components of the blood-brain barrier, Pharm. Res., 31, 1855, 10.1007/s11095-013-1289-8 O.A. Marcos-Contreras et al., Selective targeting of nanomedicine to inflamed cerebral vasculature to enhance the blood–brain barrier, Proc. Natl. Acad. Sci. U.S.A. 117 (7) (2020) 3405–3414, doi: 10.1073/pnas.1912012117. This work evaluates anti-ICAM and VCAM antibodies in order to target liposomes to the inflamed vasculature. While many studies use fluorophores to track nanomaterials in vivo, radiolabeling is used in this to quantitate pharmacokinetic parameters. Choudhury, 2018, Transferrin receptors-targeting nanocarriers for efficient targeted delivery and transcytosis of drugs into the brain tumors: a review of recent advancements and emerging trends, Drug Deliv. and Transl. Res., 8, 1545, 10.1007/s13346-018-0552-2 Li, 2002, Transferrin/transferrin receptor-mediated drug delivery, Med. Res. Rev., 22, 225, 10.1002/med.10008 Cui, 2013, Transferrin-conjugated magnetic silica PLGA nanoparticles loaded with doxorubicin and paclitaxel for brain glioma treatment, Biomaterials, 34, 8511, 10.1016/j.biomaterials.2013.07.075 Shruthi, 2022, Elevated expression of cholesterol transporter LRP-1 is crucially implicated in the pathobiology of glioblastoma, Front. Neurol., 13, 1003730, 10.3389/fneur.2022.1003730 Régina, 2008, Antitumour activity of ANG1005, a conjugate between paclitaxel and the new brain delivery vector Angiopep-2: Antitumour activity of ANG1005, Br. J. Pharmacol., 155, 185, 10.1038/bjp.2008.260 Kumthekar, 2020, ANG1005, a brain-penetrating peptide-drug conjugate, shows activity in patients with breast cancer with leptomeningeal carcinomatosis and recurrent brain metastases, Clin. Cancer Res., 26, 2789, 10.1158/1078-0432.CCR-19-3258 Tang, 2016, ANG1005, a novel peptide-paclitaxel conjugate crosses the BBB and shows activity in patients with recurrent CNS metastasis from breast cancer, results from a phase II clinical study, Ann. Oncol., 27, p. vi103, 10.1093/annonc/mdw367.02 Drappatz, 2013, Phase I study of GRN1005 in recurrent malignant glioma, Clin. Cancer Res., 19, 1567, 10.1158/1078-0432.CCR-12-2481 Guidotti, 2019, Peptides in clinical development for the treatment of brain tumors, Curr. Opin. Pharmacol., 47, 102, 10.1016/j.coph.2019.02.007 Y. Zou et al., Blood-brain barrier–penetrating single CRISPR-Cas9 nanocapsules for effective and safe glioblastoma gene therapy, Sci. Adv. 8 (16) (2022) eabm8011, doi: 10.1126/sciadv.abm8011. In this manuscript, authors create Angiopep-2 targeted disfulfide crosslinked polymer capsules around CRISPR-Cas9 complexes. The result is a ∼30 nm nanomaterial that can accumulate into an orthotopic glioblastoma tumor and release payload in the reducing environment inside the cell. Jia, 2014, The role of claudin-5 in blood-brain barrier (BBB) and brain metastases (Review), Mol. Med. Rep., 9, 779, 10.3892/mmr.2013.1875 Sladojevic, 2019, Claudin-1-dependent destabilization of the blood-brain barrier in chronic stroke, J. Neurosci., 39, 743, 10.1523/JNEUROSCI.1432-18.2018 Bony, 2021, Claudin-1-targeted nanoparticles for delivery to aging-induced alterations in the blood-brain barrier, ACS Nano, 15, 18520, 10.1021/acsnano.1c08432 Yanagida, 2017, Size-selective opening of the blood–brain barrier by targeting endothelial sphingosine 1–phosphate receptor 1, PNAS, 114, 4531, 10.1073/pnas.1618659114 Wang, 2016, Mfsd2a-based pharmacological strategies for drug delivery across the blood–brain barrier, Pharmacol. Res., 104, 124, 10.1016/j.phrs.2015.12.024 Ju, 2021, Overcoming Mfsd2a-mediated low transcytosis to boost nanoparticle delivery to brain for chemotherapy of brain metastases, Adv. Healthc. Mater., 10, 2001997, 10.1002/adhm.202001997 Yang, 2017, Mfsd2a (major facilitator superfamily domain containing 2a) attenuates intracerebral hemorrhage-induced blood–brain barrier disruption by inhibiting vesicular transcytosis, JAHA, 6, e005811, 10.1161/JAHA.117.005811 Smith-Cohn, 2022, Transient opening of the blood-brain barrier by vasoactive peptides to increase CNS drug delivery: reality versus wishful thinking?, Curr. Neuropharmacol., 20, 1383, 10.2174/1570159X20999220131163504 Carman, 2011, Adenosine receptor signaling modulates permeability of the blood-brain barrier, J. Neurosci., 31, 13272, 10.1523/JNEUROSCI.3337-11.2011 Jackson, 2017, The effect of regadenoson on the integrity of the human blood-brain barrier, a pilot study, J. Neurooncol, 132, 513, 10.1007/s11060-017-2404-1 Raymond, 1986, Pharmacological modification of bradykinin induced breakdown of the blood-brain barrier, Can. J. Neurol. Sci., 13, 214, 10.1017/S0317167100036301 Borlongan, 2003, Facilitation of drug entry into the CNS via transient permeation of blood brain barrier: laboratory and preliminary clinical evidence from bradykinin receptor agonist, Cereport Brain Res. Bull., 60, 297, 10.1016/S0361-9230(03)00043-1 Bartus, 1996, Permeability of the blood brain barrier by the bradykinin agonist, RMP-7: evidence for a sensitive, auto-regulated, receptor-mediated system, Immunopharmacology, 33, 270, 10.1016/0162-3109(96)00070-7 Prados, 2003, 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 Oncol., 5, 96, 10.1093/neuonc/5.2.96 Li, 2021, From blood to brain: blood cell-based biomimetic drug delivery systems, Drug Deliv., 28, 1214, 10.1080/10717544.2021.1937384 Chen, 2020, Biomimetic drug-delivery systems for the management of brain diseases, Biomater. Sci., 8, 1073, 10.1039/C9BM01395D Banks, 2010, The blood–brain barrier and immune function and dysfunction, Neurobiol. Dis., 37, 26, 10.1016/j.nbd.2009.07.031 Matthews, 2019, Chronic inflammation in multiple sclerosis — seeing what was always there, Nat. Rev. Neurol., 15, 582, 10.1038/s41582-019-0240-y Hodes, 2015, Neuroimmune mechanisms of depression, Nat. Neurosci., 18, 1386, 10.1038/nn.4113 Xie, 2019, Nanomaterial-based blood-brain-barrier (BBB) crossing strategies, Biomaterials, 224, 10.1016/j.biomaterials.2019.119491 Doshi, 2011, Cell-based drug delivery devices using phagocytosis-resistant backpacks, Adv. Mater., 23, H105, 10.1002/adma.201004074 Klyachko, 2017, Macrophages with cellular backpacks for targeted drug delivery to the brain, Biomaterials, 140, 79, 10.1016/j.biomaterials.2017.06.017 Pang, 2018, Primary M1 macrophages as multifunctional carrier combined with PLGA nanoparticle delivering anticancer drug for efficient glioma therapy, Drug Deliv., 25, 1922, 10.1080/10717544.2018.1502839 Xue, 2017, Neutrophil-mediated anticancer drug delivery for suppression of postoperative malignant glioma recurrence, Nat. Nanotech, 12, 692, 10.1038/nnano.2017.54 Lu, 2020, Targeted immunomodulation of inflammatory monocytes across the blood-brain barrier by curcumin-loaded nanoparticles delays the progression of experimental autoimmune encephalomyelitis, Biomaterials, 245, 10.1016/j.biomaterials.2020.119987 Hou, 2019, Accessing neuroinflammation sites: Monocyte/neutrophil-mediated drug delivery for cerebral ischemia, Sci. Adv., 5, 10.1126/sciadv.aau8301 Anselmo, 2013, Delivering nanoparticles to lungs while avoiding liver and spleen through adsorption on red blood cells, ACS Nano, 7, 11129, 10.1021/nn404853z Pan, 2016, The effect of polymeric nanoparticles on biocompatibility of carrier red blood cells, PLoS One, 11, e0152074, 10.1371/journal.pone.0152074 Brenner, 2018, Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude, Nat. Commun., 9, 2684, 10.1038/s41467-018-05079-7 Xia, 2019, Red blood cell membrane-camouflaged nanoparticles: a novel drug delivery system for antitumor application, Acta Pharm. Sin. B, 9, 675, 10.1016/j.apsb.2019.01.011 Fu, 2015, Programmed co-delivery of paclitaxel and doxorubicin boosted by camouflaging with erythrocyte membrane, Nanoscale, 7, 4020, 10.1039/C4NR07027E Wei, 2015, A D -peptide ligand of nicotine acetylcholine receptors for brain-targeted drug delivery, Angew. Chem. Int. Ed., 54, 3023, 10.1002/anie.201411226 Chai, 2017, A facile approach to functionalizing cell membrane-coated nanoparticles with neurotoxin-derived peptide for brain-targeted drug delivery, J. Control. Release, 264, 102, 10.1016/j.jconrel.2017.08.027 Doyle, 2019, Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis, Cells, 8, 727, 10.3390/cells8070727 Zheng, 2019, Harnessing exosomes for the development of brain drug delivery systems, Bioconjug. Chem., 30, 994, 10.1021/acs.bioconjchem.9b00085 Amiri, 2022, Exosomes as bio-inspired nanocarriers for RNA delivery: preparation and applications, J. Transl. Med., 20, 125, 10.1186/s12967-022-03325-7 el Andaloussi, 2013, Exosomes for targeted siRNA delivery across biological barriers, Adv. Drug Deliv. Rev., 65, 391, 10.1016/j.addr.2012.08.008 Banks, 2020, Transport of extracellular vesicles across the blood-brain barrier: brain pharmacokinetics and effects of inflammation, IJMS, 21, 4407, 10.3390/ijms21124407 Batrakova, 2015, Using exosomes, naturally-equipped nanocarriers, for drug delivery, J. Control. Release, 219, 396, 10.1016/j.jconrel.2015.07.030 Tan, 2013, Exosomes as nano-theranostic delivery platforms for gene therapy, Adv. Drug Deliv. Rev., 65, 357, 10.1016/j.addr.2012.06.014 van den Boorn, 2011, SiRNA delivery with exosome nanoparticles, Nat. Biotechnol., 29, 325, 10.1038/nbt.1830 Yuan, 2017, Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain, Biomaterials, 142, 1, 10.1016/j.biomaterials.2017.07.011 Kumar, 2007, Transvascular delivery of small interfering RNA to the central nervous system, Nature, 448, 39, 10.1038/nature05901 Liu, 2009, Brain-targeting gene delivery and cellular internalization mechanisms for modified rabies virus glycoprotein RVG29 nanoparticles, Biomaterials, 30, 4195, 10.1016/j.biomaterials.2009.02.051 Liu, 2015, Targeted exosome-mediated delivery of opioid receptor Mu siRNA for the treatment of morphine relapse, Sci. Rep., 5, 17543, 10.1038/srep17543 Alvarez-Erviti, 2011, Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes, Nat. Biotechnol., 29, 341, 10.1038/nbt.1807 Zhu, 2022, Specific anti-glioma targeted-delivery strategy of engineered small extracellular vesicles dual-functionalised by Angiopep-2 and TAT peptides, J. Extracellular Vesicle, 11, 10.1002/jev2.12255 Jia, 2018, NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo, Biomaterials, 178, 302, 10.1016/j.biomaterials.2018.06.029 Doolittle, 2000, Safety and efficacy of a multicenter study using intraarterial chemotherapy in conjunction with osmotic opening of the blood-brain barrier for the treatment of patients with malignant brain tumors, Cancer, 88, 637, 10.1002/(SICI)1097-0142(20000201)88:3<637::AID-CNCR22>3.0.CO;2-Y Boockvar, 2011, Safety and maximum tolerated dose of superselective intraarterial cerebral infusion of bevacizumab after osmotic blood-brain barrier disruption for recurrent malignant glioma: clinical article, JNS, 114, 624, 10.3171/2010.9.JNS101223 Burkhardt, 2012, Intra-arterial delivery of bevacizumab after blood-brain barrier disruption for the treatment of recurrent glioblastoma: progression-free survival and overall survival, World Neurosurg., 77, 130, 10.1016/j.wneu.2011.05.056 Chakraborty, 2016, Superselective intraarterial cerebral infusion of cetuximab after osmotic blood/brain barrier disruption for recurrent malignant glioma: phase I study, J. Neurooncol., 128, 405, 10.1007/s11060-016-2099-8 Carpentier, 2016, Clinical trial of blood-brain barrier disruption by pulsed ultrasound, Sci. Transl. Med., 8, 10.1126/scitranslmed.aaf6086 Mainprize, 2019, Blood-brain barrier opening in primary brain tumors with non-invasive MR-guided focused ultrasound: a clinical safety and feasibility study, Sci. Rep., 9, 321, 10.1038/s41598-018-36340-0 Meng, 2021, MR-guided focused ultrasound enhances delivery of trastuzumab to Her2-positive brain metastases, Sci. Transl. Med., 13, eabj4011, 10.1126/scitranslmed.abj4011 Park, 2021, Safety and feasibility of multiple blood-brain barrier disruptions for the treatment of glioblastoma in patients undergoing standard adjuvant chemotherapy, J. Neurosurg., 134, 475, 10.3171/2019.10.JNS192206 Park, 2020, One-year outcome of multiple blood-brain barrier disruptions with temozolomide for the treatment of glioblastoma, Front. Oncol., 10, 1663, 10.3389/fonc.2020.01663 Lipsman, 2018, Blood–brain barrier opening in Alzheimer’s disease using MR-guided focused ultrasound, Nat. Commun., 9, 2336, 10.1038/s41467-018-04529-6 Abrahao, 2019, First-in-human trial of blood–brain barrier opening in amyotrophic lateral sclerosis using MR-guided focused ultrasound, Nat. Commun., 10, 4373, 10.1038/s41467-019-12426-9 Todd, 2020, Secondary effects on brain physiology caused by focused ultrasound-mediated disruption of the blood–brain barrier, J. Control. Release, 324, 450, 10.1016/j.jconrel.2020.05.040 Chu, 2015, Neuromodulation accompanying focused ultrasound-induced blood-brain barrier opening, Sci. Rep., 5, 15477, 10.1038/srep15477 Todd, 2019, The neurovascular response is attenuated by focused ultrasound-mediated disruption of the blood-brain barrier, Neuroimage, 201, 10.1016/j.neuroimage.2019.116010 Jordão, 2013, Amyloid-β plaque reduction, endogenous antibody delivery and glial activation by brain-targeted, transcranial focused ultrasound, Exp. Neurol., 248, 16, 10.1016/j.expneurol.2013.05.008 Kovacs, 2017, Disrupting the blood-brain barrier by focused ultrasound induces sterile inflammation, PNAS, 114, E75, 10.1073/pnas.1614777114 Garcia, 2022, Single-cell dissection of the human brain vasculature, Nature, 603, 893, 10.1038/s41586-022-04521-7 Yang, 2022, A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk, Nature, 603, 885, 10.1038/s41586-021-04369-3 Winkler, 2022, A single-cell atlas of the normal and malformed human brain vasculature, Science, 375, eabi7377, 10.1126/science.abi7377