Transportation of Single-Domain Antibodies through the Blood–Brain Barrier
Abstract
:1. Introduction
2. The Blood–Brain Barrier
3. Antibodies, Nanobodies and the BBB
4. Brain Delivery of Nanobodies through the Blood–Brain Barrier
4.1. Physiopathological Disruption of the BBB
4.2. Device-Based and Physicochemical Disruption of the BBB
4.3. Receptor-Mediated Transcytosis
4.4. Adsorptive-Mediated Transcytosis
4.5. Shuttle-Mediated Transcytosis
4.5.1. Cell-Penetrating Peptides (CPPs)
4.5.2. Liposomes
4.5.3. Nanoparticles
4.6. Somatic Gene Transfer of Nanobodies into the Brain Parenchyma
5. Discussion and Future Perspectives
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Muyldermans, S. Nanobodies: Natural single-domain antibodies. Annu. Rev. Biochem. 2013, 82, 775–797. [Google Scholar] [CrossRef] [Green Version]
- Steeland, S.; Vandenbroucke, R.E.; Libert, C. Nanobodies as therapeutics: Big opportunities for small antibodies. Drug Discov. Today 2016, 21, 1076–1113. [Google Scholar] [CrossRef] [PubMed]
- Jovčevska, I.; Muyldermans, S. The Therapeutic Potential of Nanobodies. BioDrugs 2020, 34, 11–26. [Google Scholar] [CrossRef] [Green Version]
- Pothin, E.; Lesuisse, D.; Lafaye, P. Brain delivery of single-domain antibodies: A focus on VHH and VNAR. Pharmaceutics 2020, 12, 937. [Google Scholar] [CrossRef] [PubMed]
- Gao, Y.; Zhu, J.; Lu, H. Single domain antibody-based vectors in the delivery of biologics across the blood–brain barrier: A review. Drug Deliv. Transl. Res. 2020. [Google Scholar] [CrossRef] [PubMed]
- Bélanger, K.; Iqbal, U.; Tanha, J.; MacKenzie, R.; Moreno, M.; Stanimirovic, D. Single-Domain Antibodies as Therapeutic and Imaging Agents for the Treatment of CNS Diseases. Antibodies 2019, 8, 27. [Google Scholar] [CrossRef] [PubMed] [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]
- CM de Lange, E. The Physiological Characteristics and Transcytosis Mechanisms of the Blood-Brain Barrier (BBB). Curr. Pharm. Biotechnol. 2012, 13, 2319–2327. [Google Scholar] [CrossRef] [PubMed]
- Muoio, V.; Persson, P.B.; Sendeski, M.M. The neurovascular unit-concept review. Acta Physiol. 2014, 210, 790–798. [Google Scholar] [CrossRef]
- Pulgar, V.M. Transcytosis to cross the blood brain barrier, new advancements and challenges. Front. Neurosci. 2019, 13, 1–9. [Google Scholar] [CrossRef]
- Pardridge, W.M. Drug transport across the blood-brain barrier. J. Cereb. Blood Flow Metab. 2012, 32, 1959–1972. [Google Scholar] [CrossRef] [PubMed]
- López-Otín, C.; Kroemer, G. Hallmarks of Health. Cell 2021, 184, 33–63. [Google Scholar] [CrossRef] [PubMed]
- Širochmanová, I.; Čomor, Ľ.; Káňová, E.; Jiménez-Munguía, I.; Tkáčová, Z.; Bhide, M. Permeability of the Blood-Brain Barrier and Transport of Nanobodies Across the Blood-Brain Barrier. Folia Vet. 2018, 62, 59–66. [Google Scholar] [CrossRef] [Green Version]
- Papadopoulos, M.C.; Saadoun, S.; Davies, D.C.; Bell, B.A. Emerging molecular mechanisms of brain tumour oedema. Br. J. Neurosurg. 2001, 15, 101–108. [Google Scholar] [CrossRef]
- Davies, D.C. Blood-brain barrier breakdown in septic encephalopathy and brain tumours. J. Anat. 2002, 200, 639–646. [Google Scholar] [CrossRef]
- Abbott, N.J.; Rönnbäck, L.; Hansson, E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [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] [Green Version]
- Margolin, K.; Ernstoff, M.S.; Hamid, O.; Lawrence, D.; McDermott, D.; Puzanov, I.; Wolchok, J.D.; Clark, J.I.; Sznol, M.; Logan, T.F.; et al. Ipilimumab in patients with melanoma and brain metastases: An open-label, phase 2 trial. Lancet Oncol. 2012, 13, 459–465. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. The Microenvironmental Landscape of Brain Tumors. Cancer Cell 2017, 31, 326–341. [Google Scholar] [CrossRef] [Green Version]
- Amaral, T.; Kiecker, F.; Schaefer, S.; Stege, H.; Kaehler, K.; Terheyden, P.; Gesierich, A.; Gutzmer, R.; Haferkamp, S.; Uttikal, J.; et al. Combined immunotherapy with nivolumab and ipilimumab with and without local therapy in patients with melanoma brain metastasis: A DeCOG∗ study in 380 patients. J. Immunother. Cancer 2020, 8, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Montagne, A.; Nation, D.A.; Sagare, A.P.; Barisano, G.; Sweeney, M.D.; Chakhoyan, A.; Pachicano, M.; Joe, E.; Nelson, A.R.; D’Orazio, L.M.; et al. APOE4 leads to blood–brain barrier dysfunction predicting cognitive decline. Nature 2020, 581, 71–76. [Google Scholar] [CrossRef]
- Halliday, M.R.; Rege, S.V.; Ma, Q.; Zhao, Z.; Miller, C.A.; Winkler, E.A.; Zlokovic, B.V. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. J. Cereb. Blood Flow Metab. 2016, 36, 216–227. [Google Scholar] [CrossRef]
- Sevenich, L.; Bowman, R.L.; Mason, S.D.; Quail, D.F.; Elie, B.T.; Brogi, E.; Brastianos, P.K.; Hahn, W.C.; Holsinger, J.; Massagué, J.; et al. Analysis of tumor- and stroma-supplied proteolytic networks reveals a brain metastasis-promoting role for cathepsin S. Nat. Cell Biol. 2014, 16, 876–888. [Google Scholar] [CrossRef] [Green Version]
- Eichler, A.F.; Chung, E.; Kodack, D.P.; Loeffler, J.S.; Fukumura, D.; Jain, R.K. The biology of brain metastases-translation to new therapies. Nat. Rev. Clin. Oncol. 2011, 8, 344–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steeg, P.S.; Camphausen, K.A.; Smith, Q.R. Brain metastases as preventive and therapeutic targets. Nat. Rev. Cancer 2011, 11, 352–363. [Google Scholar] [CrossRef] [PubMed]
- Potter, M.; Lieberman, R. Genetics of Immunoglobulins in the Mouse. Adv. Immunol. 1967, 7, 91–145. [Google Scholar] [CrossRef]
- Edelman, G.M.; Gall, W.E. The antibody problem. Annu. Rev. Biochem. 1969, 38, 415–466. [Google Scholar] [CrossRef] [PubMed]
- Pardridge, W.M. Drug targeting to the brain. Pharm. Res. 2007, 24, 1733–1744. [Google Scholar] [CrossRef] [PubMed]
- Pepinsky, R.B.; Shao, Z.; Ji, B.; Wang, Q.; Meng, G.; Walus, L.; Lee, X.; Hu, Y.; Graff, C.; Garber, E.; et al. Exposure levels of anti-LINGO-1 Li81 antibody in the central nervous system and dose-efficacy relationships in rat spinal cord remyelination models after systemic administration. J. Pharmacol. Exp. Ther. 2011, 339, 519–529. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Pardridge, W.M. Rapid transferrin efflux from brain to blood across the blood-brain barrier. J. Neurochem. 2001, 76, 1597–1600. [Google Scholar] [CrossRef]
- Siegelman, J.; Fleit, H.B.; Peress, N.S. Characterization of immunoglobulin G-Fe receptor activity in the outflow system of the cerebrospinal fluid. Cell Tissue Res. 1987, 248, 599–605. [Google Scholar] [CrossRef]
- Schlachetzki, F.; Zhu, C.; Pardridge, W.M. Expression of the neonatal Fc receptor (FcRn) at the blood–brain barrier. J. Neurochem. 2002, 81, 203–206. [Google Scholar] [CrossRef]
- Cooper, P.R.; Ciambrone, G.J.; Kliwinski, C.M.; Maze, E.; Johnson, L.; Li, Q.; Feng, Y.; Hornby, P.J. Efflux of monoclonal antibodies from rat brain by neonatal Fc receptor, FcRn. Brain Res. 2013, 1534, 13–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abuqayyas, L.; Balthasar, J.P. Investigation of the role of FcγR and FcRn in mAb distribution to the brain. Mol. Pharm. 2013, 10, 1505–1513. [Google Scholar] [CrossRef] [PubMed]
- Ruano-Salguero, J.S.; Lee, K.H. Antibody transcytosis across brain endothelial-like cells occurs nonspecifically and independent of FcRn. Sci. Rep. 2020, 10, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Garg, A.; Balthasar, J.P. Investigation of the influence of FcRn on the distribution of IgG to the brain. AAPS J. 2009, 11, 553–557. [Google Scholar] [CrossRef] [Green Version]
- Kunik, V.; Peters, B.; Ofran, Y. Structural consensus among antibodies defines the antigen binding site. PLoS Comput. Biol. 2012, 8, e1002388. [Google Scholar] [CrossRef] [PubMed]
- Kunik, V.; Ashkenazi, S.; Ofran, Y. Paratome: An online tool for systematic identification of antigen-binding regions in antibodies based on sequence or structure. Nucleic Acids Res. 2012, 40, 521–524. [Google Scholar] [CrossRef] [Green Version]
- Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Robinson, G.; Hammers, C.; Songa, E.B.; Bendahman, N.; Hammers, R. Naturally occurring antibodies devoid of light chains. Nature 1993, 363, 446–448. [Google Scholar] [CrossRef]
- Greenberg, A.S.; Avila, D.; Hughes, M.; Hughes, A.; McKinney, E.C.; Flajnik, M.F. A New Antigen Receptor Gene Family That Undergoes Rearrangement and Extensive Somatic Diversification In Sharks. Nature 1995, 374, 168–173. [Google Scholar] [CrossRef]
- Muyldermans, S.; Baral, T.N.; Retamozzo, V.C.; De Baetselier, P.; De Genst, E.; Kinne, J.; Leonhardt, H.; Magez, S.; Nguyen, V.K.; Revets, H.; et al. Camelid immunoglobulins and nanobody technology. Vet. Immunol. Immunopathol. 2008, 128, 178–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arbabi Ghahroudi, M.; Desmyter, A.; Wyns, L.; Hamers, R.; Muyldermans, S. Selection and identification of single domain antibody fragments from camel heavy-chain antibodies. FEBS Lett. 1997, 414, 521–526. [Google Scholar] [CrossRef] [Green Version]
- Nuttall, S.D. Overview and discovery of IgNARs and generation of VNARs. Methods Mol. Biol. 2012, 911, 27–36. [Google Scholar] [CrossRef] [PubMed]
- Abskharon, R.N.N.; Giachin, G.; Wohlkonig, A.; Soror, S.H.; Pardon, E.; Legname, G.; Steyaert, J. Probing the N-terminal β-sheet conversion in the crystal structure of the human prion protein bound to a nanobody. J. Am. Chem. Soc. 2014, 136, 937–944. [Google Scholar] [CrossRef]
- De Genst, E.; Silence, K.; Decanniere, K.; Conrath, K.; Loris, R.; Kinne, J.; Muyldermans, S.; Wyns, L. Molecular basis for the preferential cleft recognition by dromedary heavy-chain antibodies. Proc. Natl. Acad. Sci. USA 2006, 103, 4586–4591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Staus, D.P.; Wingler, L.M.; Strachan, R.T.; Rasmussen, S.G.F.; Pardon, E.; Ahn, S.; Steyaert, J.; Kobilka, B.K.; Lefkowitz, R.J. Regulation of b2-Adrenergic Receptor Function by Conformationally Selective Single-Domain Intrabodies s. Mol. Pharmacol. 2013, 85, 472–481. [Google Scholar] [CrossRef] [Green Version]
- Freise, A.C.; Wu, A.M. In vivo imaging with antibodies and engineered fragments. Mol. Immunol. 2015, 67, 142–152. [Google Scholar] [CrossRef] [Green Version]
- Ackaert, C.; Smiejkowska, N.; Xavier, C.; Sterckx, Y.G.J.; Denies, S.; Stijlemans, B.; Elkrim, Y.; Devoogdt, N.; Caveliers, V.; Lahoutte, T.; et al. Immunogenicity Risk Profile of Nanobodies. Front. Immunol. 2021, 12, 1–15. [Google Scholar] [CrossRef]
- Soler, M.A.; Medagli, B.; Wang, J.; Oloketuyi, S.; Bajc, G.; Huang, H.; Fortuna, S.; De Marco, A. Effect of humanizing mutations on the stability of the llama single-domain variable region. Biomolecules 2021, 11, 163. [Google Scholar] [CrossRef]
- Rossotti, M.A.; Bélanger, K.; Henry, K.A.; Tanha, J. Immunogenicity and humanization of single-domain antibodies. FEBS J. 2021, 2021, 1–24. [Google Scholar] [CrossRef]
- Duggan, S. Caplacizumab: First Global Approval. Drugs 2018, 78, 1639–1642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scully, M.; Cataland, S.R.; Peyvandi, F.; Coppo, P.; Knöbl, P.; Kremer Hovinga, J.A.; Metjian, A.; de la Rubia, J.; Pavenski, K.; Callewaert, F.; et al. Caplacizumab Treatment for Acquired Thrombotic Thrombocytopenic Purpura. N. Engl. J. Med. 2019, 380, 335–346. [Google Scholar] [CrossRef]
- Vugmeyster, Y.; Entrican, C.A.; Joyce, A.P.; Lawrence-Henderson, R.F.; Leary, B.A.; Mahoney, C.S.; Patel, H.K.; Raso, S.W.; Olland, S.H.; Hegen, M.; et al. Pharmacokinetic, biodistribution, and biophysical profiles of TNF nanobodies conjugated to linear or branched poly(ethylene glycol). Bioconjug. Chem. 2012, 23, 1452–1462. [Google Scholar] [CrossRef]
- Rotman, M.; Welling, M.M.; Bunschoten, A.; De Backer, M.E.; Rip, J.; Nabuurs, R.J.A.; Gaillard, P.J.; Van Buchem, M.A.; Van Der Maarel, S.M.; Van Der Weerd, L. Enhanced glutathione PEGylated liposomal brain delivery of an anti-amyloid single domain antibody fragment in a mouse model for Alzheimer’s disease. J. Control. Release 2015, 203, 40–50. [Google Scholar] [CrossRef]
- Farrington, G.K.; Caram-Salas, N.; Haqqani, A.S.; Brunette, E.; Eldredge, J.; Pepinsky, B.; Antognetti, G.; Baumann, E.; Ding, W.; Garber, E.; et al. A novel platform for engineering blood-brain barrier-crossing bispecific biologics. FASEB J. 2014, 28, 4764–4778. [Google Scholar] [CrossRef] [Green Version]
- Sehlin, D.; Stocki, P.; Gustavsson, T.; Hultqvist, G.; Walsh, F.S.; Rutkowski, J.L.; Syvänen, S. Brain delivery of biologics using a cross-species reactive transferrin receptor 1 VNAR shuttle. FASEB J. 2020, 34, 13272–13283. [Google Scholar] [CrossRef]
- Iqbal, U.; Trojahn, U.; Albaghdadi, H.; Zhang, J.; O’Connor-Mccourt, M.; Stanimirovic, D.; Tomanek, B.; Sutherland, G.; Abulrob, A. Kinetic analysis of novel mono- and multivalent VHH-fragments and their application for molecular imaging of brain tumours. Br. J. Pharmacol. 2010, 160, 1016–1028. [Google Scholar] [CrossRef]
- Hultberg, A.; Temperton, N.J.; Rosseels, V.; Koenders, M.; Gonzalez-Pajuelo, M.; Schepens, B.; Ibañez, L.I.; Vanlandschoot, P.; Schillemans, J.; Saunders, M.; et al. Llama-derived single domain antibodies to build multivalent, superpotent and broadened neutralizing anti-viral molecules. PLoS ONE 2011, 6, e17665. [Google Scholar] [CrossRef] [Green Version]
- Glassman, P.M.; Walsh, L.R.; Villa, C.H.; Marcos-Contreras, O.A.; Hood, E.D.; Muzykantov, V.R.; Greineder, C.F. Molecularly Engineered Nanobodies for Tunable Pharmacokinetics and Drug Delivery. Bioconjug. Chem. 2020, 31, 1144–1155. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Bourgeois, J.P.; Celli, S.; Glacial, F.; Le Sourd, A.M.; Mecheri, S.; Weksler, B.; Romero, I.; Couraud, P.O.; Rougeon, F.; et al. Cell-penetrating anti-GFAP VHH and corresponding fluorescent fusion protein VHH-GFP spontaneously cross the blood-brain barrier and specifically recognize astrocytes: Application to brain imaging. FASEB J. 2012, 26, 3969–3979. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Vandesquille, M.; Koukouli, F.; Dudeffant, C.; Youssef, I.; Lenormand, P.; Ganneau, C.; Maskos, U.; Czech, C.; Grueninger, F.; et al. Camelid single-domain antibodies: A versatile tool for in vivo imaging of extracellular and intracellular brain targets. J. Control. Release 2016, 243, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Dorresteijn, B.; Rotman, M.; Faber, D.; Schravesande, R.; Suidgeest, E.; Van Der Weerd, L.; Van Der Maarel, S.M.; Verrips, C.T.; El Khattabi, M. Camelid heavy chain only antibody fragment domain against β-site of amyloid precursor protein cleaving enzyme 1 inhibits β-secretase activity in vitro and in vivo. FEBS J. 2015, 282, 3618–3631. [Google Scholar] [CrossRef] [PubMed]
- Rissiek, B.; Koch-Nolte, F.; Magnus, T. Nanobodies as modulators of inflammation: Potential applications for acute brain injury. Front. Cell. Neurosci. 2014, 8, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stanimirovic, D.; Kemmerich, K.; Haqqani, A.S.; Farrington, G.K. Engineering and Pharmacology of Blood-Brain Barrier-Permeable Bispecific Antibodies, 1st ed.; Elsevier Inc.: New York, NY, USA, 2014; Volume 71. [Google Scholar]
- Keyaerts, M.; Xavier, C.; Heemskerk, J.; Devoogdt, N.; Everaert, H.; Ackaert, C.; Vanhoeij, M.; Duhoux, F.P.; Gevaert, T.; Simon, P.; et al. Phase I study of 68Ga-HER2-Nanobody for PET/CT assessment of HER2 expression in breast carcinoma. J. Nucl. Med. 2016, 57, 27–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iqbal, U.; Albaghdadi, H.; Luo, Y.; Arbabi, M.; Desvaux, C.; Veres, T.; Stanimirovic, D.; Abulrob, A. Molecular imaging of glioblastoma multiforme using anti-insulin-like growth factor-binding protein-7 single-domain antibodies. Br. J. Cancer 2010, 103, 1606–1616. [Google Scholar] [CrossRef] [Green Version]
- Varatharaj, A.; Galea, I. The blood-brain barrier in systemic inflammation. Brain. Behav. Immun. 2017, 60, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steeland, S.; Puimège, L.; Vandenbroucke, R.E.; Van Hauwermeiren, F.; Haustraete, J.; Devoogdt, N.; Hulpiau, P.; Leroux-Roels, G.; Laukens, D.; Meuleman, P.; et al. Generation and characterization of small single domain antibodies inhibiting human tumor necrosis factor receptor 1. J. Biol. Chem. 2014, 290, 4022–4037. [Google Scholar] [CrossRef] [Green Version]
- Steeland, S.; Van Ryckeghem, S.; Van Imschoot, G.; De Rycke, R.; Toussaint, W.; Vanhoutte, L.; Vanhove, C.; De Vos, F.; Vandenbroucke, R.E.; Libert, C. TNFR1 inhibition with a Nanobody protects against EAE development in mice. Sci. Rep. 2017, 7, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steeland, S.; Gorlé, N.; Vandendriessche, C.; Balusu, S.; Brkic, M.; Van Cauwenberghe, C.; Van Imschoot, G.; Van Wonterghem, E.; De Rycke, R.; Kremer, A.; et al. Counteracting the effects of TNF receptor-1 has therapeutic potential in Alzheimer’s disease. EMBO Mol. Med. 2018, 10, 1–22. [Google Scholar] [CrossRef]
- Lafaye, P.; Achour, I.; England, P.; Duyckaerts, C.; Rougeon, F. Single-domain antibodies recognize selectively small oligomeric forms of amyloid β, prevent Aβ-induced neurotoxicity and inhibit fibril formation. Mol. Immunol. 2009, 46, 695–704. [Google Scholar] [CrossRef]
- Lombardo, S.; Catteau, J.; Besson, M.; Maskos, U. A role for β2* nicotinic receptors in a model of local amyloid pathology induced in dentate gyrus. Neurobiol. Aging 2016, 46, 221–234. [Google Scholar] [CrossRef] [Green Version]
- Terryn, S.; Francart, A.; Lamoral, S.; Hultberg, A.; Rommelaere, H.; Wittelsberger, A.; Callewaert, F.; Stohr, T.; Meerschaert, K.; Ottevaere, I.; et al. Protective effect of different anti-rabies virus VHH constructs against rabies disease in mice. PLoS ONE 2014, 9, e109367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boruah, B.M.; Liu, D.; Ye, D.; Gu, T.J.; Jiang, C.L.; Qu, M.; Wright, E.; Wang, W.; He, W.; Liu, C.; et al. Single Domain Antibody Multimers Confer Protection against Rabies Infection. PLoS ONE 2013, 8, e71383. [Google Scholar] [CrossRef] [PubMed]
- Caljon, G.; Caveliers, V.; Lahoutte, T.; Stijlemans, B.; Ghassabeh, G.H.; Van Den Abbeele, J.; Smolders, I.; De Baetselier, P.; Michotte, Y.; Muyldermans, S.; et al. Using microdialysis to analyse the passage of monovalent nanobodies through the blood-brain barrier. Br. J. Pharmacol. 2012, 165, 2341–2353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hunt Bobo, R.; Laske, D.W.; Akbasak, A.; Morrison, P.F.; Dedrick, R.L.; Oldfield, E.H. Convection-enhanced delivery of macromolecules in the brain. Proc. Natl. Acad. Sci. USA 1994, 91, 2076–2080. [Google Scholar] [CrossRef] [Green Version]
- Debinski, W.; Tatter, S.B. Convection-enhanced delivery for the treatment of brain tumors. Expert Rev. Neurother. 2009, 9, 1519–1527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barua, N.U.; Gill, S.S.; Love, S. Convection-enhanced drug delivery to the brain: Therapeutic potential and neuropathological considerations. Brain Pathol. 2014, 24, 117–127. [Google Scholar] [CrossRef]
- D’Amico, R.S.; Aghi, M.K.; Vogelbaum, M.A.; Bruce, J.N. Convection-enhanced drug delivery for glioblastoma: A review. J. Neurooncol. 2021, 151, 415–427. [Google Scholar] [CrossRef] [PubMed]
- Faraji, A.H.; Rajendran, S.; Jaquins-Gerstl, A.S.; Hayes, H.J.; Richardson, R.M. Convection-Enhanced Delivery and Principles of Extracellular Transport in the Brain. World Neurosurg. 2021, 151, 163–171. [Google Scholar] [CrossRef]
- Boskovitz, A.; McLendon, R.E.; Okamura, T.; Sampson, J.H.; Bigner, D.D.; Zalutsky, M.R. Treatment of HER2 Positive Breast Carcinomatous Meningitis with Intrathecal Administration of α-Particle Emitting 211 At-labeled Trastuzumab. Nucl. Med. Biol. 2009, 36, 659–669. [Google Scholar] [CrossRef] [Green Version]
- Pizzo, M.E.; Wolak, D.J.; Kumar, N.N.; Brunette, E.; Brunnquell, C.L.; Hannocks, M.J.; Abbott, N.J.; Meyerand, M.E.; Sorokin, L.; Stanimirovic, D.B.; et al. Intrathecal antibody distribution in the rat brain: Surface diffusion, perivascular transport and osmotic enhancement of delivery. J. Physiol. 2018, 596, 445–475. [Google Scholar] [CrossRef] [Green Version]
- Rapoport, S.I. Advances in osmotic opening of the blood-brain barrier to enhance CNS chemotherapy. Expert Opin. Investig. Drugs 2001, 10, 1809–1818. [Google Scholar] [CrossRef] [PubMed]
- Shin, B.J.; Burkhardt, J.K.; Riina, H.A.; Boockvar, J.A. Superselective Intra-Arterial Cerebral Infusion of Novel Agents After Blood-Brain Disruption for the Treatment of Recurrent Glioblastoma Multiforme: A Technical Case Series. Neurosurg. Clin. N. Am. 2012, 23, 323–329. [Google Scholar] [CrossRef]
- Lesniak, W.G.; Chu, C.; Jablonska, A.; Azad, B.B.; Zwaenepoel, O.; Zawadzki, M.; Lisok, A.; Pomper, M.; Walczak, P. PET imaging of distinct brain uptake of a nanobody and similarly-sized PAMAM dendrimers after intra-arterial administration. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1940–1951. [Google Scholar] [CrossRef] [PubMed]
- Nabuurs, R.J.A.; Rutgers, K.S.; Welling, M.M.; Metaxas, A.; de Backer, M.E.; Rotman, M.; Bacskai, B.J.; van Buchem, M.A.; van der Maarel, S.M.; van der Weerd, L. In vivo detection of amyloid-β deposits using heavy chain antibody fragments in a transgenic mouse model for alzheimer’s disease. PLoS ONE 2012, 7, e38284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.Q.; Chen, P.; Haimovitz-Friedman, A.; Reilly, R.M.; Wong, C.S. Endothelial apoptosis initiates acute blood-brain barrier disruption after ionizing radiation. Cancer Res. 2003, 63, 5950–5956. [Google Scholar]
- Kiyatkin, E.A.; Sharma, H.S. Permeability of the blood-brain barrier depends on brain temperature. Neuroscience 2009, 161, 926–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shanker Sharma, H. Hyperthermia induced brain oedema: Current status and future perspectives. Indian J. Med. Res. 2006, 123, 629–652. [Google Scholar]
- Hildebrandt, B.; Wust, P.; Ahlers, O.; Dieing, A.; Sreenivasa, G.; Kerner, T.; Felix, R.; Riess, H. The cellular and molecular basis of hyperthermia. Crit. Rev. Oncol. Hematol. 2002, 43, 33–56. [Google Scholar] [CrossRef]
- Dan, M.; Bae, Y.; Pittman, T.A.; Yokel, R.A. Alternating Magnetic Field-Induced Hyperthermia Increases Iron Oxide Nanoparticle Cell Association/Uptake and Flux in Blood-Brain Barrier Models. Pharm. Res. 2015, 32, 1615–1625. [Google Scholar] [CrossRef] [Green Version]
- Tabatabaei, S.N.; Girouard, H.; Carret, A.S.; Martel, S. 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 2015, 206, 49–57. [Google Scholar] [CrossRef] [PubMed]
- Gupta, R.; Sharma, D. Evolution of Magnetic Hyperthermia for Glioblastoma Multiforme Therapy. ACS Chem. Neurosci. 2019, 10, 1157–1172. [Google Scholar] [CrossRef] [PubMed]
- Zhu, S.; Huang, A.G.; Luo, F.; Li, J.; Li, J.; Zhu, L.; Zhao, L.; Zhu, B.; Ling, F.; Wang, G.X. Application of Virus Targeting Nanocarrier Drug Delivery System in Virus-Induced Central Nervous System Disease Treatment. ACS Appl. Mater. Interfaces 2019, 11, 19006–19016. [Google Scholar] [CrossRef]
- Van Campenhout, R.; Muyldermans, S.; Vinken, M.; Devoogdt, N.; De Groof, T.W.M. Therapeutic nanobodies targeting cell plasma membrane transport proteins: A high-risk/high-gain endeavor. Biomolecules 2021, 11, 63. [Google Scholar] [CrossRef]
- Muruganandam, A.; Tanha, J.; Narang, S.; Stanimirovic, D. Selection of phage-displayed llama single-domain antibodies that transmigrate across human blood-brain barrier endothelium. FASEB J. 2002, 16, 240–242. [Google Scholar] [CrossRef]
- Abulrob, A.; Sprong, H.; Van Bergen En Henegouwen, P.; Stanimirovic, D. The blood-brain barrier transmigrating single domain antibody: Mechanisms of transport and antigenic epitopes in human brain endothelial cells. J. Neurochem. 2005, 95, 1201–1214. [Google Scholar] [CrossRef] [PubMed]
- Haqqani, A.S.; Caram-Salas, N.; Ding, W.; Brunette, E.; Delaney, C.E.; Baumann, E.; Boileau, E.; Stanimirovic, D. Multiplexed evaluation of serum and CSF pharmacokinetics of brain-targeting single-domain antibodies using a NanoLC-SRM-ILIS method. Mol. Pharm. 2013, 10, 1542–1556. [Google Scholar] [CrossRef] [PubMed]
- Werner, H.; LeRoith, D. Insulin and insulin-like growth factor receptors in the brain: Physiological and pathological aspects. Eur. Neuropsychopharmacol. 2014, 24, 1947–1953. [Google Scholar] [CrossRef] [Green Version]
- Sheff, J.; Wang, P.; Xu, P.; Arbour, M.; Masson, L.; van Faassen, H.; Hussack, G.; Kemmerich, K.; Brunette, E.; Stanimirovic, D.; et al. Defining the epitope of a blood–brain barrier crossing single domain antibody specific for the type 1 insulin-like growth factor receptor. Sci. Rep. 2021, 11, 1–13. [Google Scholar] [CrossRef]
- Stanimirovic, D.K.; Kemmerich, K.; Haqqani, A.S.; Sulea, T.; Arbabi-Ghahroudi, M.; Massie, B.; Gilbert, R. Insulin-Like Growth Factor 1 Receptor-Specific Antibodies and Uses Thereof. U.S. Patent US20170015748A1, 19 January 2017. [Google Scholar]
- Rutgers, K.S.; van Remoortere, A.; van Buchem, M.A.; Verrips, C.T.; Greenberg, S.M.; Bacskai, B.J.; Frosch, M.P.; van Duinen, S.G.; Maat-Schieman, M.L.; Van der Maarel, S.M. Differential recognition of vascular and parenchymal beta amyloid deposition. Neurobiol. Aging 2011, 32, 1774–1783. [Google Scholar] [CrossRef] [Green Version]
- Rutgers, K.S.; Nabuurs, R.J.A.; van den Berg, S.A.A.; Schenk, G.J.; Rotman, M.; Verrips, C.T.; van Duinen, S.G.; Maat-Schieman, M.L.; van Buchem, M.A.; de Boer, A.G.; et al. Transmigration of beta amyloid specific heavy chain antibody fragments across the in vitro blood-brain barrier. Neuroscience 2011, 190, 37–42. [Google Scholar] [CrossRef]
- Jones, D.R.; Taylor, W.A.; Bate, C.; David, M.; Tayebi, M. A camelid anti-PrP antibody abrogates PrPSc replication in prion-permissive neuroblastoma cell lines. PLoS ONE 2010, 5, e9804. [Google Scholar] [CrossRef] [Green Version]
- David, M.A.; Jones, D.R.; Tayebi, M. Potential candidate camelid antibodies for the treatment of protein-misfolding diseases. J. Neuroimmunol. 2014, 272, 76–85. [Google Scholar] [CrossRef]
- Stocki, P.; Szary, J.; Rasmussen, C.L.M.; Demydchuk, M.; Northall, L.; Logan, D.B.; Gauhar, A.; Thei, L.; Moos, T.; Walsh, F.S.; et al. Blood-brain barrier transport using a high affinity, brain-selective VNAR antibody targeting transferrin receptor 1. FASEB J. 2020, 35, e21172. [Google Scholar] [CrossRef]
- Leitner, D.F.; Connor, J.R. Functional roles of transferrin in the brain. Biochim. Biophys. Acta Gen. Subj. 2012, 1820, 393–402. [Google Scholar] [CrossRef]
- Johnsen, K.B.; Burkhart, A.; Melander, F.; Kempen, P.J.; Vejlebo, J.B.; Siupka, P.; Nielsen, M.S.; Andresen, T.L.; Moos, T. Targeting transferrin receptors at the blood-brain barrier improves the uptake of immunoliposomes and subsequent cargo transport into the brain parenchyma. Sci. Rep. 2017, 7, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Wouters, Y.; Jaspers, T.; De Strooper, B.; Dewilde, M. Identification and in vivo characterization of a brain-penetrating nanobody. Fluids Barriers CNS 2020, 17, 4–13. [Google Scholar] [CrossRef]
- Hervé, F.; Ghinea, N.; Scherrmann, J.M. CNS delivery via adsorptive transcytosis. AAPS J. 2008, 10, 455–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, W. Adsorptive-Mediated Brain Delivery Systems. Curr. Pharm. Biotechnol. 2012, 13, 2340–2348. [Google Scholar] [CrossRef] [PubMed]
- Zhu, X.; Jin, K.; Huang, Y.; Pang, Z. Brain Drug Delivery by Adsorption-Mediated Transcytosis; Elsevier Ltd.: Amsterdam, The Netherlands, 2019; ISBN 9780128140017. [Google Scholar]
- Vandesquille, M.; Li, T.; Po, C.; Ganneau, C.; Lenormand, P.; Dudeffant, C.; Czech, C.; Grueninger, F.; Duyckaerts, C.; Delatour, B.; et al. Chemically-defined camelid antibody bioconjugate for the magnetic resonance imaging of Alzheimer’s disease. MAbs 2017, 9, 1016–1027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kang, T.; Gao, X.; Chen, J. Harnessing the Capacity of Cell-Penetrating Peptides for Drug Delivery to the Central Nervous System. Curr. Pharm. Biotechnol. 2014, 15, 220–230. [Google Scholar] [CrossRef]
- Škrlj, N.; Drevenšek, G.; Hudoklin, S.; Romih, R.; Čurin Šerbec, V.; Dolinar, M. Recombinant single-Chain antibody with the trojan peptide penetratin positioned in the linker region enables cargo transfer across the blood-brain barrier. Appl. Biochem. Biotechnol. 2013, 169, 159–169. [Google Scholar] [CrossRef]
- Spencer, B.; Williams, S.; Rockenstein, E.; Valera, E.; Xin, W.; Mante, M.; Florio, J.; Adame, A.; Masliah, E.; Sierks, M.R. α-synuclein conformational antibodies fused to penetratin are effective in models of Lewy body disease. Ann. Clin. Transl. Neurol. 2016, 3, 588–606. [Google Scholar] [CrossRef]
- Herce, H.D.; Schumacher, D.; Schneider, A.F.L.; Ludwig, A.K.; Mann, F.A.; Fillies, M.; Kasper, M.A.; Reinke, S.; Krause, E.; Leonhardt, H.; et al. Cell-permeable nanobodies for targeted immunolabelling and antigen manipulation in living cells. Nat. Chem. 2017, 9, 762–771. [Google Scholar] [CrossRef] [PubMed]
- van Lith, S.A.M.; van den Brand, D.; Wallbrecher, R.; van Duijnhoven, S.M.J.; Brock, R.; Leenders, W.P.J. A Conjugate of an Anti-Epidermal Growth Factor Receptor (EGFR) VHH and a Cell-Penetrating Peptide Drives Receptor Internalization and Blocks EGFR Activation. ChemBioChem 2017, 18, 2390–2394. [Google Scholar] [CrossRef]
- van Lith, S.A.M.; van den Brand, D.; Wallbrecher, R.; Wübbeke, L.; van Duijnhoven, S.M.J.; Mäkinen, P.I.; Hoogstad-van Evert, J.S.; Massuger, L.; Ylä-Herttuala, S.; Brock, R.; et al. The effect of subcellular localization on the efficiency of EGFR-targeted VHH photosensitizer conjugates. Eur. J. Pharm. Biopharm. 2018, 124, 63–72. [Google Scholar] [CrossRef]
- Wang, D.; El-Amouri, S.S.; Dai, M.; Kuan, C.Y.; Hui, D.Y.; Brady, R.O.; Pan, D. Engineering a lysosomal enzyme with a derivative of receptor-binding domain of apoE enables delivery across the blood-brain barrier. Proc. Natl. Acad. Sci. USA 2013, 110, 2999–3004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Markoutsa, E.; Papadia, K.; Giannou, A.D.; Spella, M.; Cagnotto, A.; Salmona, M.; Stathopoulos, G.T.; Antimisiaris, S.G. Mono and dually decorated nanoliposomes for brain targeting, in vitro and in vivo studies. Pharm. Res. 2014, 31, 1275–1289. [Google Scholar] [CrossRef]
- Papadia, K.; Giannou, A.D.; Markoutsa, E.; Bigot, C.; Vanhoute, G.; Mourtas, S.; Van der Linded, A.; Stathopoulos, G.T.; Antimisiaris, S.G. Multifunctional LUV liposomes decorated for BBB and amyloid targeting-B. In vivo brain targeting potential in wild-type and APP/PS1 mice. Eur. J. Pharm. Sci. 2017, 102, 180–187. [Google Scholar] [CrossRef] [PubMed]
- Rotman, M.; Welling, M.M.; van den Boogaard, M.L.; Moursel, L.G.; van der Graaf, L.M.; van Buchem, M.A.; van der Maarel, S.M.; van der Weerd, L. Fusion of hIgG1-Fc to 111In-anti-amyloid single domain antibody fragment VHH-pa2H prolongs blood residential time in APP/PS1 mice but does not increase brain uptake. Nucl. Med. Biol. 2015, 42, 695–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kannan, R.; Chakrabarti, R.; Tang, D.; Kim, K.J.; Kaplowitz, N. GSH transport in human cerebrovascular endothelial cells and human astrocytes: Evidence for luminal localization of Na+-dependent GSH transport in HCEC. Brain Res. 2000, 852, 374–382. [Google Scholar] [CrossRef]
- Yin, W.; Zhao, Y.; Kang, X.; Zhao, P.; Fu, X.; Mo, X.; Wan, Y.; Huang, Y. BBB-penetrating codelivery liposomes treat brain metastasis of non-small cell lung cancer with EGFRT790M mutation. Theranostics 2020, 10, 6122–6135. [Google Scholar] [CrossRef]
- Thangudu, S.; Cheng, F.Y.; Su, C.H. Advancements in the blood–brain barrier penetrating nanoplatforms for brain related disease diagnostics and therapeutic applications. Polymers 2020, 12, 3055. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Filipiak, M.S.; Rother, M.; Andoy, N.M.; Knudsen, A.C.; Grimm, S.; Bachran, C.; Swee, L.K.; Zaumseil, J.; Tarasov, A. Highly sensitive, selective and label-free protein detection in physiological solutions using carbon nanotube transistors with nanobody receptors. Sens. Actuators B Chem. 2018, 255, 1507–1516. [Google Scholar] [CrossRef]
- Mann, F.A.; Lv, Z.; Großhans, J.; Opazo, F.; Kruss, S. Nanobody-Conjugated Nanotubes for Targeted Near-Infrared In Vivo Imaging and Sensing. Angew. Chem. Int. Ed. 2019, 58, 11469–11473. [Google Scholar] [CrossRef] [PubMed]
- Kafa, H.; Wang, J.T.W.; Rubio, N.; Venner, K.; Anderson, G.; Pach, E.; Ballesteros, B.; Preston, J.E.; Abbott, N.J.; Al-Jamal, K.T. The interaction of carbon nanotubes with an invitro blood-brain barrier model and mouse brain invivo. Biomaterials 2015, 53, 437–452. [Google Scholar] [CrossRef] [Green Version]
- Hong, S.; Leroueil, P.R.; Majoros, I.J.; Orr, B.G.; Baker, J.R.; Banaszak Holl, M.M. The Binding Avidity of a Nanoparticle-Based Multivalent Targeted Drug Delivery Platform. Chem. Biol. 2007, 14, 107–115. [Google Scholar] [CrossRef] [Green Version]
- Tassa, C.; Duffner, J.L.; Lewis, T.A.; Weissleder, R.; Schreiber, S.L.; Koehler, A.N.; Shaw, S.Y. Binding affinity and kinetic analysis of targeted small molecule-modified nanoparticles. Bioconjug. Chem. 2010, 21, 14–19. [Google Scholar] [CrossRef] [Green Version]
- Qin, M.; Wang, L.; Wu, D.; Williams, C.K.; Xu, D.; Kranz, E.; Guo, Q.; Guan, J.; Vinters, H.V.; Lee, Y.J.; et al. Enhanced Delivery of Rituximab Into Brain and Lymph Nodes Using Timed-Release Nanocapsules in Non-Human Primates. Front. Immunol. 2020, 10, 1–13. [Google Scholar] [CrossRef]
- Zafir-Lavie, I.; Sherbo, S.; Goltsman, H.; Badinter, F.; Yeini, E.; Ofek, P.; Miari, R.; Tal, O.; Liran, A.; Shatil, T.; et al. Successful intracranial delivery of trastuzumab by gene-therapy for treatment of HER2-positive breast cancer brain metastases. J. Control. Release 2018, 291, 80–89. [Google Scholar] [CrossRef]
- Rincon, M.Y.; Zhou, L.; Marneffe, C.; Voytyuk, I.; Wouters, Y.; Dewilde, M.; Duqué, S.I.; Vincke, C.; Levites, Y.; Golde, T.E.; et al. AAV mediated delivery of a novel anti-BACE1 VHH reduces Abeta in an Alzheimer’s disease mouse model. bioRxiv 2019. [Google Scholar] [CrossRef]
- Asokan, A.; Conway, J.C.; Phillips, J.L.; Li, C.; Hegge, J.; Sinnott, R.; Yadav, S.; Diprimio, N.; Nam, H.J.; Agbandje-Mckenna, M.; et al. Reengineering a receptor footprint of adeno-associated virus enables selective and systemic gene transfer to muscle. Nat. Biotechnol. 2010, 28, 79–82. [Google Scholar] [CrossRef] [Green Version]
- Choudhury, S.R.; Fitzpatrick, Z.; Harris, A.F.; Maitland, S.A.; Ferreira, J.S.; Zhang, Y.; Ma, S.; Sharma, R.B.; Gray-Edwards, H.L.; Johnson, J.A.; et al. In vivo selection yields AAV-B1 capsid for central nervous system and muscle gene therapy. Mol. Ther. 2016, 24, 1247–1257. [Google Scholar] [CrossRef] [Green Version]
- Deverman, B.E.; Pravdo, P.L.; Simpson, B.P.; Ravindra Kumar, S.; Chan, K.Y.; Banerjee, A.; Wu, W.-L.; Yang, B.; Huber, N.; Pasca, S.P.; et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 2016, 34, 204–209. [Google Scholar] [CrossRef] [PubMed]
- Passeleu-Le Bourdonnec, C.; Carrupt, P.A.; Scherrmann, J.M.; Martel, S. Methodologies to assess drug permeation through the blood-brain barrier for pharmaceutical research. Pharm. Res. 2013, 30, 2729–2756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- René, C.A.; Parks, R.J. Delivery of therapeutic agents to the central nervous system and the promise of extracellular vesicles. Pharmaceutics 2021, 13, 492. [Google Scholar] [CrossRef]
- Saunders, N.R.; Liddelow, S.A.; Dziegielewska, K.M. Barrier mechanisms in the developing brain. Front. Pharmacol. 2012, 3, 1–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bartus, R.T.; Elliott, P.J.; Dean, R.L.; Hayward, N.J.; Nagle, T.L.; Huff, M.R.; Snodgrass, P.A.; Blunt, D.G. Controlled modulation of BBB permeability using the bradykinin agonist, RMP-7. Exp. Neurol. 1996, 142, 14–28. [Google Scholar] [CrossRef] [PubMed]
- Thomas, H.D.; Lind, M.J.; Ford, J.; Bleehen, N.; Calvert, A.H.; Boddy, A.V. Pharmacokinetics of carboplatin administered in combination with the bradykinin agonist Cereport (RMP-7) for the treatment of brain tumours. Cancer Chemother. Pharmacol. 2000, 45, 284–290. [Google Scholar] [CrossRef]
- Packer, R.J.; Krailo, M.; Mehta, M.; Warren, K.; Allen, J.; Jakacki, R.; Villablanca, J.G.; Chiba, A.; Reaman, G. A phase I study of concurrent RMP-7 and carboplatin with radiation therapy for children with newly diagnosed brainstem gliomas. Cancer 2005, 104, 1968–1974. [Google Scholar] [CrossRef] [PubMed]
- Carman, A.J.; Mills, J.H.; Krenz, A.; Kim, D.G.; Bynoe, M.S. Adenosine receptor signaling modulates permeability of the blood-brain barrier. J. Neurosci. 2011, 31, 13272–13280. [Google Scholar] [CrossRef]
- Jackson, S.; Anders, N.M.; Mangraviti, A.; Wanjiku, T.M.; Sankey, E.W.; Liu, A.; Brem, H.; Tyler, B.; Rudek, M.A.; Grossman, S.A. The effect of regadenoson-induced transient disruption of the blood–brain barrier on temozolomide delivery to normal rat brain. J. Neurooncol. 2016, 126, 433–439. [Google Scholar] [CrossRef] [Green Version]
- Lammers, T.; Koczera, P.; Fokong, S.; Gremse, F.; Ehling, J.; Vogt, M.; Pich, A.; Storm, G.; Van Zandvoort, M.; Kiessling, F. Theranostic USPIO-loaded microbubbles for mediating and monitoring blood-brain barrier permeation. Adv. Funct. Mater. 2015, 25, 36–43. [Google Scholar] [CrossRef] [PubMed]
- Fan, X.; Wang, L.; Guo, Y.; Tu, Z.; Li, L.; Tong, H.; Xu, Y.; Li, R.; Fang, K. Ultrasonic nanobubbles carrying anti-PSMA nanobody: Construction and application in prostate cancer-targeted imaging. PLoS ONE 2015, 10, e0127419. [Google Scholar] [CrossRef] [PubMed]
- Yu, Z.; Hu, M.; Li, Z.; Dan, X.; Zhu, L.; Guo, Y.; Liu, Q.; Lan, W.; Jiang, J.; Wang, L. Anti-G250 nanobody-functionalized nanobubbles targeting renal cell carcinoma cells for ultrasound molecular imaging. Nanotechnology 2020, 31. [Google Scholar] [CrossRef] [PubMed]
- Yu, Z.; Wang, Y.; Xu, D.; Zhu, L.; Hu, M.; Liu, Q.; Lan, W.; Jiang, J.; Wang, L. G250 antigen-targeting drug-loaded nanobubbles combined with ultrasound targeted nanobubble destruction: A potential novel treatment for renal cell carcinoma. Int. J. Nanomed. 2020, 15, 81–95. [Google Scholar] [CrossRef] [Green Version]
- Hernot, S.; Unnikrishnan, S.; Du, Z.; Shevchenko, T.; Cosyns, B.; Broisat, A.; Toczek, J.; Caveliers, V.; Muyldermans, S.; Lahoutte, T.; et al. Nanobody-coupled microbubbles as novel molecular tracer. J. Control. Release 2012, 158, 346–353. [Google Scholar] [CrossRef] [Green Version]
- Punjabi, M.; Xu, L.; Ochoa-Espinosa, A.; Kosareva, A.; Wolff, T.; Murtaja, A.; Broisat, A.; Devoogdt, N.; Kaufmann, B.A. Ultrasound Molecular Imaging of Atherosclerosis with Nanobodies: Translatable Microbubble Targeting Murine and Human VCAM (Vascular Cell Adhesion Molecule) 1. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 2520–2530. [Google Scholar] [CrossRef]
- Marty, B.; Larrat, B.; Van Landeghem, M.; Robic, C.; Robert, P.; Port, M.; Le Bihan, D.; Pernot, M.; Tanter, M.; Lethimonnier, F.; et al. Dynamic study of blood-brain barrier closure after its disruption using ultrasound: A quantitative analysis. J. Cereb. Blood Flow Metab. 2012, 32, 1948–1958. [Google Scholar] [CrossRef] [PubMed]
- Janowicz, P.W.; Leinenga, G.; Götz, J.; Nisbet, R.M. Ultrasound-mediated blood-brain barrier opening enhances delivery of therapeutically relevant formats of a tau-specific antibody. Sci. Rep. 2019, 9, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Urakawa, M.; Yamaguchi, K.; Tsuchida, E.; Kashiwagi, S.; Ito, H.; Matsuda, T. Blood-brain barrier disturbance following localized hyperthermia in rats. Int. J. Hyperth. 1995, 11, 709–718. [Google Scholar] [CrossRef]
- Praça, C.; Rai, A.; Santos, T.; Cristovão, A.C.; Pinho, S.L.; Cecchelli, R.; Dehouck, M.P.; Bernardino, L.; Ferreira, L.S. A nanoformulation for the preferential accumulation in adult neurogenic niches. J. Control. Release 2018, 284, 57–72. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Navarro, M.; Giralt, E.; Teixidó, M. Blood–brain barrier peptide shuttles. Curr. Opin. Chem. Biol. 2017, 38, 134–140. [Google Scholar] [CrossRef] [PubMed]
- Druzhkova, T.A.; Yakovlev, A.A. Exosome Drug Delivery through the Blood–Brain Barrier: Experimental Approaches and Potential Applications. Neurochem. J. 2018, 12, 195–204. [Google Scholar] [CrossRef]
- Garcia, J.; Arranz-Gibert, P.; Sánchez-Navarro, M.; Giralt, E.; Teixidó, M. Peptide Shuttle-Mediated Delivery for Brain Gene Therapies. Curr. Top. Med. Chem. 2020, 20, 2945–2958. [Google Scholar] [CrossRef]
- Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
- Van Niel, G.; Porto-Carreiro, I.; Simoes, S.; Raposo, G. Exosomes: A common pathway for a specialized function. J. Biochem. 2006, 140, 13–21. [Google Scholar] [CrossRef]
- Cocucci, E.; Racchetti, G.; Meldolesi, J. Shedding microvesicles: Artefacts no more. Trends Cell Biol. 2009, 19, 43–51. [Google Scholar] [CrossRef]
- Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as Drug Delivery Vehicles for Parkinson’s Disease Therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [Green Version]
- Yuan, D.; Zhao, Y.; Banks, W.A.; Bullock, K.M.; Haney, M.; Batrakova, E.; Kabanov, A.V. Macrophage Exosomes as Natural Nanocarriers for Protein Delivery to Inflamed Brain. Biomaterials 2017, 142, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Ye, Z.; Zhang, T.; He, W.; Jin, H.; Liu, C.; Yang, Z.; Ren, J. Methotrexate-Loaded Extracellular Vesicles Functionalized with Therapeutic and Targeted Peptides for the Treatment of Glioblastoma Multiforme. ACS Appl. Mater. Interfaces 2018, 10, 12341–12350. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
- Cooper, J.M.; Wiklander, P.B.O.; Nordin, J.Z.; Al-Shawi, R.; Wood, M.J.; Vithlani, M.; Schapira, A.H.V.; Simons, J.P.; El-Andaloussi, S.; Alvarez-Erviti, L. Systemic exosomal siRNA delivery reduced alpha-synuclein aggregates in brains of transgenic mice. Mov. Disord. 2014, 29, 1476–1485. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.; Zhang, X.; Chen, X.; Wang, L.; Yang, G. Exosome Mediated Delivery of miR-124 Promotes Neurogenesis after Ischemia. Mol. Ther. Nucleic Acids 2017, 7, 278–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Murphy, D.E.; de Jong, O.G.; Brouwer, M.; Wood, M.J.; Lavieu, G.; Schiffelers, R.M.; Vader, P. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019, 51. [Google Scholar] [CrossRef]
- Wiklander, O.P.B.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Kooijmans, S.A.A.; Fliervoet, L.A.L.; Van Der Meel, R.; Fens, M.H.A.M.; Heijnen, H.F.G.; Van Bergen En Henegouwen, P.M.P.; Vader, P.; Schiffelers, R.M. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. J. Control. Release 2016, 224, 77–85. [Google Scholar] [CrossRef]
- Bao, C.; Gao, Q.; Li, L.; Han, L.; Zhang, B.; Ding, Y.; Song, Z.; Zhang, R.; Zhang, J.; Wu, X. The Application of Nanobody in CAR-T Therapy. Biomolecules 2021, 11, 238. [Google Scholar] [CrossRef] [PubMed]
- Xie, Y.J.; Dougan, M.; Ingram, J.R.; Pishesha, N.; Fang, T.; Momin, N.; Ploegh, H.L. Improved Antitumor Efficacy of Chimeric Antigen Receptor T Cells that Secrete Single-Domain Antibody Fragments. Cancer Immunol. Res. 2020, 8, 518–529. [Google Scholar] [CrossRef]
- Smith, D.E.; Johanson, C.E.; Keep, R.F. Peptide and peptide analog transport systems at the blood-CSF barrier. Adv. Drug Deliv. Rev. 2004, 56, 1765–1791. [Google Scholar] [CrossRef] [PubMed]
- Johanson, C.E.; Johanson, N.L. Choroid Plexus Blood-CSF Barrier: Major Player in Brain Disease Modeling and Neuromedicine. J. Neurol. Neuromed. 2018, 3, 39–58. [Google Scholar] [CrossRef]
- Louveau, A.; Herz, J.; Alme, M.N.; Francesca, A.; Dong, M.Q.; Viar, K.E.; Herod, G.; Knopp, J.; Setliff, J.; Lupi, A.L.; et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat. Neurosci. 2018, 21, 1380–1391. [Google Scholar] [CrossRef]
- Goldmann, J.; Kwidzinski, E.; Brandt, C.; Mahlo, J.; Richter, D.; Bechmann, I. T cells traffic from brain to cervical lymph nodes via the cribroid plate and the nasal mucosa. J. Leukoc. Biol. 2006, 80, 797–801. [Google Scholar] [CrossRef] [PubMed]
- Mondin, V.; Ferlito, A.; Devaney, K.O.; Woolgar, J.A.; Rinaldo, A. A survey of metastatic central nervous system tumors to cervical lymph nodes. Eur. Arch. Oto-Rhino-Laryngology 2010, 267, 1657–1666. [Google Scholar] [CrossRef] [PubMed]
- Aspelund, A.; Antila, S.; Proulx, S.T.; Karlsen, T.V.; Karaman, S.; Detmar, M.; Wiig, H.; Alitalo, K. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 2015, 212, 991–999. [Google Scholar] [CrossRef] [PubMed]
- Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Sherin, J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Kevin, L.; et al. Structural and functional features of central nervous system lymphatics. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef] [PubMed]
- Absinta, M.; Ha, S.-K.; Nair, G.; Sati, P.; Luciano, N.J.; Palisoc, M.; Louveau, A.; Zaghloul, K.A.; Pittaluga, S.; Kipnis, J.; et al. Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI. Elife 2017, 6, 1–15. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Ruiz-López, E.; Schuhmacher, A.J. Transportation of Single-Domain Antibodies through the Blood–Brain Barrier. Biomolecules 2021, 11, 1131. https://doi.org/10.3390/biom11081131
Ruiz-López E, Schuhmacher AJ. Transportation of Single-Domain Antibodies through the Blood–Brain Barrier. Biomolecules. 2021; 11(8):1131. https://doi.org/10.3390/biom11081131
Chicago/Turabian StyleRuiz-López, Eduardo, and Alberto J. Schuhmacher. 2021. "Transportation of Single-Domain Antibodies through the Blood–Brain Barrier" Biomolecules 11, no. 8: 1131. https://doi.org/10.3390/biom11081131
APA StyleRuiz-López, E., & Schuhmacher, A. J. (2021). Transportation of Single-Domain Antibodies through the Blood–Brain Barrier. Biomolecules, 11(8), 1131. https://doi.org/10.3390/biom11081131