Skip to main content

Advertisement

Log in

Targeting Therapeutics Across the Blood Brain Barrier (BBB), Prerequisite Towards Thrombolytic Therapy for Cerebrovascular Disorders—an Overview and Advancements

  • Review Article
  • Published:
AAPS PharmSciTech Aims and scope Submit manuscript

ABSTRACT

Cerebral tissues possess highly selective and dynamic protection known as blood brain barrier (BBB) that regulates brain homeostasis and provides protection against invading pathogens and various chemicals including drug molecules. Such natural protection strictly monitors entry of drug molecules often required for the management of several diseases and disorders including cerebral vascular and neurological disorders. However, in recent times, the ischemic cerebrovascular disease and clinical manifestation of acute arterial thrombosis are the most common causes of mortality and morbidity worldwide. The management of cerebral Ischemia requires immediate infusion of external thrombolytic into systemic circulation and must cross the blood brain barrier. The major challenge with available thrombolytic is their poor affinity towards the blood brain barrier and cerebral tissue subsequently. In the clinical practice, a high dose of thrombolytic often prescribed to deliver drugs across the blood brain barrier which results in drug dependent toxicity leading to damage of neuronal tissues. In recent times, more emphasis was given to utilize blood brain barrier transport mechanism to deliver drugs in neuronal tissue. The blood brain barrier expresses a series of receptor on membrane became an ideal target for selective drug delivery. In this review, the author has given more emphasis molecular biology of receptor on blood brain barrier and their potential as a carrier for drug molecules to cerebral tissues. Further, the use of nanoscale design and real-time monitoring for developed therapeutic to encounter drug dependent toxicity has been reviewed in this study.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

REFERENCES

  1. Abbott NJ. Physiology of the blood–brain barrier and its consequences for drug transport to the brain. Int Congr Ser. 2005;1277:3–18.

    Article  CAS  Google Scholar 

  2. Jones AR, Shusta EV. Blood–brain barrier transport of therapeutics via receptor mediation. Pharm Res. 2007;24:1759–71.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Ballabh P, Braun A, Nedergaard M. The blood–brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16(1):1–13.

    Article  CAS  PubMed  Google Scholar 

  4. Cardoso FL, Brites D, Brito MA. Looking at the blood–brain barrier: molecular anatomy and possible investigation approaches. Brain Res Rev. 2010;64:328–63.

    Article  CAS  PubMed  Google Scholar 

  5. Pardridge WM. Blood–brain barrier drug targeting: the future of brain drug development. Mol Interv. 2003;3:90–105.

    Article  CAS  PubMed  Google Scholar 

  6. Eyal S, Hsiao P, Unadkat JD. Drug interactions at the blood–brain barrier: fact or fantasy? Pharmacol Ther. 2009;123:80–104.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. Shilo M, Motiei M, Hana P, Popovtzer R. Transport of nanoparticles through the blood–brain barrier for imaging and therapeutic applications. Nanoscale. 2014;6(4):2146–52.

    Article  CAS  PubMed  Google Scholar 

  8. Hawkins BT, Davis TP. The blood–brain barrier/neurovascular unit in health and disease. Pharmacol Rev. 2005;57:173–85.

    Article  CAS  PubMed  Google Scholar 

  9. Ji H, Wang L, Bi H, Sun L, Cai B, Wang Y, et al. Mechanisms of lumbrokinase in protection of cerebral ischemia. Eur J Pharmacol. 2008;590:281–9.

    Article  CAS  PubMed  Google Scholar 

  10. Furlan AJ, Abou-Chebi A. The role of recombinant pro-urokinase (r-pro-UK) and intraarterial thrombolysis in acute ischemic stroke: the PROACT trials. Prolyse in acute cerebral thromboembolism. Curr Med Res Opin. 2002;18 Suppl 2:44–7.

    Article  Google Scholar 

  11. Cesarman-Maus G, Hajjar KA. Molecular mechanisms of fibrinolysis. Br J Haematol. 2005;129(3):307–21.

    Article  CAS  PubMed  Google Scholar 

  12. Huai Q, Mazar A, Kuo A, Parry GC, Shaw DE, Callahan J, et al. Structure of human urokinase plasminogen activator in complex with its receptor. Science. 2006;311:656–9.

    Article  CAS  PubMed  Google Scholar 

  13. Lakhan SE, Kirchgessner A, Hofer M. Inflammatory mechanisms in ischemic stroke: therapeutic approaches. J Transl Med. 2009;7:97.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  14. Edwards MT, Murphy MM, Geraghty JJ, Wulf JA, Konzen JP. Intraarterial cerebral thrombolysis for acute ischemic stroke in a community hospital. AJNR Am J Neuroradiol. 1999;20(9):1682–7.

    CAS  PubMed  Google Scholar 

  15. Wu JH, Diamond SL. Tissue plasminogen activator (t-PA) inhibits plasmin degradation of fibrin. A mechanism that slows t-PA-mediated fibrinolysis but does not require alpha 2-antiplasmin or leakage of intrinsic plasminogen. J Clin Invest. 1995;95(6):2483–90.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Rhim T, Lee DY, Lee M. Drug delivery systems for the treatment of ischemic stroke. Pharm Res. 2013;30(10):2429–44.

    Article  CAS  PubMed  Google Scholar 

  17. Abbott NJ. Prediction of blood–brain barrier permeation in drug discovery from in vivo, in vitro and in silico models. Drug Discov Today Technol. 2004;1:407–16.

    Article  CAS  PubMed  Google Scholar 

  18. Mruk DD, Su L, Cheng CY. Emerging role for drug transporters at the blood brain barrier. Trends Pharmacol Sci. 2010;32:99–106.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  19. Palmer AM. The role of the blood-CNS barrier in CNS disorders and their treatment. Neurobiol Dis. 2010;37:3–12.

    Article  CAS  PubMed  Google Scholar 

  20. Pardridge WM. Introduction to the blood–brain barrier: methodology, biology, and pathology. Cambridge University PRESS. 1998; 24(9):521–81.

  21. Pardridge WM. Blood–brain barrier delivery. Drug Discov Today. 2007;12:54–61.

    Article  CAS  PubMed  Google Scholar 

  22. Anstrom JA, Brown WR, Moody DM, Thore CR, Challa VR, Block SM. Anatomical analysis of the developing cerebral vasculature in premature neonates: absence of precapillary arteriole to-venous shunts. Pediatr Res. 2002;52:554–60.

    Article  PubMed  Google Scholar 

  23. Angelow S, Yu AS. Claudins and paracellular transport: an update. Curr Opin Nephrol Hypertens. 2007;16:459–64.

    Article  CAS  PubMed  Google Scholar 

  24. Findley MK, Koval M. Regulation and roles for claudin-family tight junction proteins. IUBMB Life. 2009;61(4):431–7.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Abbott NJ, Ronnback L, Hansson E. Astrocyte–endothelial interactions at the blood brain barrier. Nat Rev Neurosci. 2006;7:41–53.

    Article  CAS  PubMed  Google Scholar 

  26. Pardridge WM. Molecular biology of the blood–brain barrier. Mol Biotechnol. 2005;30:57–70.

    Article  CAS  PubMed  Google Scholar 

  27. Asotra K, Ningaraj N, Black KL. Measurement of blood–brain and blood-tumor barrier permeabilities with (14C)-labeled tracers. Methods Mol Med. 2003;89:177–90.

    PubMed  Google Scholar 

  28. Pardridge WM. The blood–brain barrier: bottleneck in brain drug development. NeuroRx. 2005;2(1):3–14.

    Article  PubMed Central  PubMed  Google Scholar 

  29. Abulrob A, Sprong H, Bergen Henegouwen PV, Stanimirovic D. The blood–brain barrier transmigrating single domain antibody: mechanisms of transport and antigenic epitopes in human brain endothelial cells. J Neurochem. 2005;95:1201–14.

    Article  CAS  PubMed  Google Scholar 

  30. Schlossauer B, Steuer H. Comparative anatomy, physiology and in vitro models of the blood–brain and blood-retina barrier. Curr Med Chem. 2002;2:175–86.

    Google Scholar 

  31. Ferguson CS, Tyndale RF. Cytochrome P450 enzymes in the brain: emerging evidence of biological significance. Trends Pharmacol Sci. 2011;32:708–14.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  32. Miksys S, Tyndale RF. Brain drug-metabolizing cytochrome P450 enzymes are active in vivo, demonstrated by mechanism-based enzyme inhibition. Neuropsychopharmacology. 2009;34(3):634–40.

    Article  CAS  PubMed  Google Scholar 

  33. Born J, Lange T, Kern W, Mc-Gregor GP, Bickel U, Fehm HL. Sniffing neuropeptides a transnasal approach to the human brain. Nat Neurosci. 2002;5:514–6.

    Article  CAS  PubMed  Google Scholar 

  34. Huang CC, Lee CC, Hsu KS. The role of insulin receptor signalling in synaptic plasticity and cognitive function. Chang Gung Med J. 2010;33(2):115–25.

    PubMed  Google Scholar 

  35. Zhang Y, Pardridge WM. Rapid transferrin efflux from brain to blood across the blood–brain barrier. J Neurochem. 2001;76:1597–600.

    Article  CAS  PubMed  Google Scholar 

  36. Wong AD, Ye M, Levy AF, Rothstein JD, Bergles DE, Searson PC. The blood–brain barrier: an engineering perspective. Front Neuroeng. 2013;6:1–22.

    Article  CAS  Google Scholar 

  37. Moos T, Morgan EH. Transferrin and transferrin receptor function in brain barrier systems. Cell Mol Neurobiol. 2001;20:77–95.

    Article  Google Scholar 

  38. Allen DD, Lockman PR, Roder KE, Dwoskin LP, Crooks PA. Active transport of high-affinity choline and nicotine analogs into the central nervous system by the blood–brain barrier choline transporter. J Pharmacol Exp Ther. 2003;304:1268–74.

    Article  CAS  PubMed  Google Scholar 

  39. Smith QR. A review of blood brain barrier transport techniques. Methods Mol Med. 2003;89:193–208.

    CAS  PubMed  Google Scholar 

  40. Hawkins RA, O’Kane RL, Simpson IA, Viña JR. Structure of the blood–brain barrier and its role in the transport of amino acids. J Nutr. 2006;136(1):218S–26.

    CAS  PubMed  Google Scholar 

  41. Zhao F-Q, Keating AF. Functional properties and genomics of glucose transporters. Curr Genomics. 2007;8:113–28.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Augustin R, Carayannopoules MO, Dowd LO, Phay JE, Moley JF, Moley KH. Identification and characterization of human glucose transporter-like protein-9 (GLUT9). J Biol Chem. 2004;279:16229–36.

    Article  CAS  PubMed  Google Scholar 

  43. Rip J, Schenk GJ, Boer AG. Differential receptor-mediated drug targeting to the diseased brain. Expert Opin Drug Deliv. 2009;6:227–37.

    Article  CAS  PubMed  Google Scholar 

  44. Gynther M, Laine K, Ropponen J, Leppanen J, Mannila A, Nevalainen T, et al. Large neutral amino acid transporter enables brain drug delivery via prodrugs. J Med Chem. 2008;51(4):932–6.

    Article  CAS  PubMed  Google Scholar 

  45. Peura L, Malmioja K, Huttunen K, Leppanen J, Hamalainen M, Forsberg MM, et al. Design, synthesis and brain uptake of LAT1-targeted amino acid prodrugs of dopamine. Pharm Res. 2013;30(10):2523–37.

    Article  CAS  PubMed  Google Scholar 

  46. Peura L, Malmioja K, Laine K, Leppanen J, Gynther M, Isotalo A, et al. Large amino acid transporter 1 (LAT1) prodrugs of valproic acid: new prodrug design ideas for central nervous system delivery. Mol Pharm. 2011;8(5):1857–66.

    Article  CAS  PubMed  Google Scholar 

  47. Xiao G, Gan L-S. Receptor-mediated endocytosis and brain delivery of therapeutic biologics. Int J Cell Biol. 2013;2013, 703545. 14 pages.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  48. Zhang Y, Pardridge WM. Delivery of β-galactosidase to mouse brain via the blood–brain barrier transferrin receptor. J Pharmacol Exp Ther. 2005;313:1075–81.

    Article  CAS  PubMed  Google Scholar 

  49. Temsamani J, Rousselle C, Rees AR, Scherrmann JM. Vector-mediated drug delivery to the brain. Exp Opin Biol Ther. 2001;1:773–82.

    Article  CAS  Google Scholar 

  50. Maresh GA, Maness LM, Zadina JE, Kastin AJ. In vitro demonstration of a saturable transport system for leptin across the blood–brain barrier. Life Sci. 2001;69(1):67–73.

    Article  CAS  PubMed  Google Scholar 

  51. Qian ZM, Li H, Sun H, Ho K. Targeted drug delivery via the transferrin receptor mediated endocytosis pathway. Pharmacol Rev. 2002;54(4):561–87.

    Article  CAS  PubMed  Google Scholar 

  52. Li H, Qian ZM. Transferrin/transferrin receptor-mediated drug delivery. Med Res Rev. 2002;22(3):225–50.

    Article  CAS  PubMed  Google Scholar 

  53. Waheed A, Grubb JH, Zhou XY, Tomatsu S, Fleming RE, Costaldi ME, et al. Regulation of transferrin-mediated iron uptake by HFE, the protein defective in hereditary hemochromatosis. Proc Natl Acad Sci. 2002;99(5):3117–22.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  54. Karsten U, Telli H, Elisabeth H, Jorg K. Transferrin- and transferrin-receptor antibody-modified nanoparticles enable drug delivery across the blood–brain barrier (BBB). Eur J Pharm Biopharm. 2009;71(2):251–6.

    Article  CAS  Google Scholar 

  55. Van Hoof D, Rodenburg KW, Van der Horst DJ. Receptor-mediated endocytosis and intracellular trafficking of lipoproteins and transferrin in insect cells. Insect Biochem Mol Biol. 2005;35(2):117–28.

    Article  PubMed  CAS  Google Scholar 

  56. Boado RJ, Zhang Y, Zhang Y, Pardridge WM. Humanization of anti-human insulin receptor antibody for drug targeting across the human blood–brain barrier. Biotechnol Bioeng. 2007;96(2):381–91.

    Article  CAS  PubMed  Google Scholar 

  57. Coloma MJ, Lee HJ, Landaw EM, Boado RJ, Morrison SL, Pardridge WM. Transport across the primate blood–brain barrier of a genetically engineered chimeric monoclonal antibody to the human insulin receptor. Pharm Res. 2000;17(3):266–74.

    Article  CAS  PubMed  Google Scholar 

  58. Boado RJ, Zhang Y, Zhang Y, Wang Y, Pardridge WM. GDNF fusion protein for targeted-drug delivery across the human blood–brain barrier. Biotech Bioeng. 2007;100:387–96.

    Article  CAS  Google Scholar 

  59. Chung NS, Wasan KM. Potential role of the low-density lipoprotein receptor family as mediators of cellular drug uptake. Adv Drug Deliv Rev. 2004;56(9):1315–34.

    Article  CAS  PubMed  Google Scholar 

  60. Demeule M, Currie JC, Bertrand Y, Che C, Regina A, Gabathuler R, et al. Involvement of the low-density lipoprotein receptor-related protein in the transcytosis of the brain delivery vector angiopep-2. J Neurochem. 2008;106(4):1534–44.

    Article  CAS  PubMed  Google Scholar 

  61. May P, Woldt E, Matz RL, Boucher P. The LDL receptor-related protein (LRP) family: an old family of proteins with new physiological functions. Ann Med. 2007;39(3):219–28.

    Article  CAS  PubMed  Google Scholar 

  62. Yoon IS, Chen E, Busse T, Repetto E, Lakshmana MK, Koo EH, et al. Low-density lipoprotein receptor-related protein promotes amyloid precursor protein trafficking to lipid rafts in the endocytic pathway. FASEB J. 2007;21:2742–52.

    Article  CAS  PubMed  Google Scholar 

  63. Harris-White ME, Frautschy SA. Low density lipoprotein receptor-related proteins (LRPs), Alzheimer’s and cognition. Curr Drug Targets CNS Neurol Disord. 2005;4(5):469–80.

    Article  CAS  PubMed  Google Scholar 

  64. Gaillard PJ, Brink A, Boer AG. Diphtheria toxin receptor-targeted brain drug delivery. Int Congr Ser. 2005;1277:185–95.

    Article  CAS  Google Scholar 

  65. Bu G, Maksymovitch EA, Nerbonne JM, Schwartz AL. Expression and function of the low density lipoprotein receptor-related protein (LRP) in mammalian central neuron. J Biochem. 1994;269:18521–8.

    CAS  Google Scholar 

  66. Raab G, Klagsbrun M. Heparin-binding EGF like growth factor. Biochim Biophys Acta. 1997;1333:F179–99.

    CAS  PubMed  Google Scholar 

  67. Wang YF, Tsirka SE, Strickland S, Stieg PE, Soriano SG, Lipton SA. Tissue plasminogen activator (t-PA) increases neuronal damage after focal cerebral ischemia in wild-type and tPA-deficient mice. Nat Med. 1998;4(2):228–31.

    Article  CAS  PubMed  Google Scholar 

  68. Chernyshev OY, Martin-Schild S, Albright KC, Barreto A, Misra V, Acosta Grotta JC, et al. Safety of t-PA in stroke mimics and neuroimaging-negative cerebral ischemia. Neurology. 2010;74(17):1340–5.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Hatcher MA, Starr JA. Role of tissue plasminogen activator in acute ischemic stroke. Ann Pharmacother. 2011;45(3):364–71.

    Article  CAS  PubMed  Google Scholar 

  70. Cronin CA. Intravenous tissue plasminogen activator for stroke: a review of the ECASS III results in relation to prior clinical trials. J Emerg Med. 2010;38(1):99–105.

    Article  PubMed  Google Scholar 

  71. Nakashima T, Minematsu K. Prospects of thrombolytic therapy for acute ischemic stroke. Brain Nerve. 2009;61(9):1003–12.

    PubMed  Google Scholar 

  72. Anderson JL. Development and evaluation of anisoylated plasminogen streptokinase activator complex (APSAC) as a second generation thrombolytic agent. J Am Coll Cardiol. 1987;10(5 Suppl B):22B–7.

    Article  CAS  PubMed  Google Scholar 

  73. Monk JP, Heel RC. Anisoylated plasminogen streptokinase activator complex (APSAC). A review of its mechanism of action, clinical pharmacology and therapeutic use in acute myocardial infarction. Drugs. 1987;34(1):25–49.

    Article  CAS  PubMed  Google Scholar 

  74. Collen D. Staphylokinase: a potent, uniquely fibrin-selective thrombolytic agent. Nat Med. 1998;4(3):279–84.

    Article  CAS  PubMed  Google Scholar 

  75. Moreadith RW, Collen D. Clinical development of PEGylated recombinant staphylokinase (PEG-Sak) for bolus thrombolytic treatment of patients with acute myocardial infarction. Adv Drug Deliv Rev. 2003;55(10):1337–45.

    Article  CAS  PubMed  Google Scholar 

  76. Cooper EL, Hrzenjak TM, Grdisa T. Alternative sources of fibrinolytic, anticoagulative, antimicrobial and anticancer molecules. Int J Immunopathol Pharmacol. 2004;17:237–44.

    CAS  PubMed  Google Scholar 

  77. Wang F, Wang C, Li M, Gui L, Zhang J, Chang W. Purification, characterization and crystallization of a group of earthworm fibrinolytic enzymes from Eisenia fetida. Biotechnol Lett. 2003;25:1105–9.

    Article  CAS  PubMed  Google Scholar 

  78. Inoue T, Yaguchi I, Takayangi K, Hayashi T, Moorrks S, Eguchi Y. A new thrombolytic agent, monteplase, is independent of plasminogen activator inhibitor in patients with acute myocardial infarction: comparison with native t-PA E 610. J Am Coll Cardiol. 1997;29:1447–53.

    Article  Google Scholar 

  79. Sun HL, Jiao JD, Pan ZW, Dong DL, Yang FB. The cardio-protective effect and mechanism of lumbrokinase. Yao Xue Xue Bao. 2006;41(3):247–51.

    CAS  PubMed  Google Scholar 

  80. Justicia C, Gabriel C, Planas AM. Activation of the JAK/STAT pathway following transient focal cerebral ischemia: signaling through Jak1 and Stat3 in astrocytes. Glia. 2000;30:253–70.

    Article  CAS  PubMed  Google Scholar 

  81. Katoh M, Karasawa T, Doi H, Odawara A, Takagi M, Ikeo T, et al. Antiplatelet mechanisms of TA-993 and its metabolite MB3 in ADP-induced platelet aggregation. Eur J Pharmacol. 2000;399:91–6.

    Article  CAS  PubMed  Google Scholar 

  82. Mascareno E, Dhar M, Siddiqui MA. Signal transduction and activator of transcription (STAT) protein-dependent activation of angiotensinogen promoter: a cellular signal for hypertrophy in cardiac muscle. Proc Natl Acad Sci. 1998;95:5590–4.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  83. Strong K, Mathers C, Bonita R. Preventing stroke: saving lives around the world. Lancet Neurol. 2007;6:182–7.

    Article  PubMed  Google Scholar 

  84. Zhang X, Zhang J, Kuang P, Lang S, Wu W, et al. The effect of lumbrokinase on P-selectin and E-selectin in cerebral ischemia model of rat. J Tradit Chin Med. 2003;23:141–6.

    PubMed  Google Scholar 

  85. Kidwell CS, Saver JL, Mattiello J, Starkman S, Vinuela F, Duckwiler G, et al. Thrombolytic reversal of acute human cerebral ischemic injury shown by diffusion/perfusion magnetic resonance imaging. Ann Neurol. 2000;47(4):462–9.

    Article  CAS  PubMed  Google Scholar 

  86. Higashida RT, Furlan AJ, Roberts H, Tomsick T, Connors B, Barr J, et al. Trial design and reporting standards for intra-arterial cerebral thrombolysis for acute ischemic stroke. Stroke. 2003;34(8):e109–37.

    Article  PubMed  Google Scholar 

  87. Schellinger PD, Jansen O, Fiebach JB, Heiland S, Steiner T, Schwab S, et al. Monitoring intravenous recombinant tissue plasminogen activator thrombolysis for acute ischemic stroke with diffusion and perfusion MRI. Stroke. 2000;31(6):1318–28.

    Article  CAS  PubMed  Google Scholar 

  88. Kuntz M, Mysiorek C, Pétrault O, Petrault M, Uzbekov R, Bordet R, et al. Stroke-induced brain parenchymal injury drives blood–brain barrier early leakage kinetics: a combined in vivo/in vitro study. J Cereb Blood Flow Metab. 2014;34:95–107.

    Article  PubMed Central  PubMed  Google Scholar 

  89. O’Collins VE, Macleod MR, Donnan GA, Horky LL, Van Der Worp BH, Howells DW. 1,026 experimental treatments in acute stroke. Ann Neurol. 2006;59:467–77.

    Article  PubMed  CAS  Google Scholar 

  90. Rosenberg N, Chen M, Prabhakaran S. New devices for treating acute ischemic stroke. Recent Pat CNS Drug Discov. 2005;5:118–34.

    Article  Google Scholar 

  91. Dirnagl U. Pathobiology of injury after stroke: the neurovascular unit and beyond. Ann N Y Acad Sci. 2012;1268:21–5.

    Article  PubMed  Google Scholar 

  92. Franko J, Pomfy M, Prosbova T, Kimakova T. Temporal kinetics of calcium in neocortical neuronal nuclei after global cerebral ischemia and reperfusion. An ultrastructural study. Sb Lek. 2001;102(2):149–52.

    CAS  PubMed  Google Scholar 

  93. Summerfield SG, Read K, Begley DJ, Obradovic T, Hidalgo IJ, Coggon S. Central nervous system drug disposition: the relationship between in situ brain permeability and brain free fraction. J Pharmacol Exp Ther. 2007;322:205–13.

    Article  CAS  PubMed  Google Scholar 

  94. Avdeef A. Physicochemical profiling (solubility, permeability and charge state). Curr Top Med Chem. 2001;1:277–351.

    Article  CAS  PubMed  Google Scholar 

  95. Laquintana V. New strategies to deliver anticancer drugs to brain tumors. Expert Opin Drug Deliv. 2009;6(10):1017–32.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  96. Pardridge W. Vector mediated drug delivery to the brain. Adv Drug Deliv Rev. 1999;36:299–321.

    Article  CAS  PubMed  Google Scholar 

  97. Benedict CR, Refino CJ, Keyt BA, Pakala R, Paoni NF, Thomas GR, et al. New variant of human tissue plasminogen activator (TPA) with enhanced efficacy and lower incidence of bleeding compared with recombinant human TPA. Circulation. 1995;92:3032–40.

    Article  CAS  PubMed  Google Scholar 

  98. Armstead WM, Riley J, Yarovoi S, Cines DB, Smith DH, Higazi AA. TPA-S481A prevents neurotoxicity of endogenous tPA in traumatic brain injury. J Neurotrauma. 2012;29(9):1794–802.

    Article  PubMed Central  PubMed  Google Scholar 

  99. Graham EM, Sheldon RA, Flock DL, Ferriero DM, Martin LJ, O’Riordan DP, et al. Neonatal mice lacking functional Fas death receptors are resistant to hypoxic-ischemic brain injury. Neurobiol Dis. 2004;17(1):89–98.

    Article  CAS  PubMed  Google Scholar 

  100. Maier W, Bednorz M, Meister S, Roebroek A, Weggen S, Schmitt U, et al. LRP1 is critical for the surface distribution and internalization of the NR2B NMDA receptor subtype. Mol Neurodegener. 2013;8:25.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  101. Kanwar JR, Sriramoju B, Kanwar RK. Neurological disorders and therapeutics targeted to surmount the blood–brain barrier. Int J Nanomedicine. 2012;7:3259–78.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  102. Pardridge WM. Biopharmaceutical drug targeting to the brain. J Drug Target. 2010;18(3):157–67.

    Article  CAS  PubMed  Google Scholar 

  103. Ren X, Cui G, Zhao M, Wang C, Peng S. Coordination of thrombolytic pro-ala-lys peptides with Cu (II): leading to nanoscale self-assembly, increase of thrombolytic activity and additional vasodilation. J Phys Chem B. 2008;112(27):8174–80.

    Article  CAS  PubMed  Google Scholar 

  104. Marsh JN, Hu G, Scott MJ, Zhang H, Goette MJ, Gaffney PJ, et al. A fibrin-specific thrombolytic nanomedicine approach to acute ischemic stroke. Nanomedicine (London). 2011;6(4):605–15.

    Article  CAS  Google Scholar 

  105. Marsh JN, Partlow KC, Abendschein DR, Scott MJ, Lanza GM, Wickline SA. Molecular imaging with targeted perfluorocarbon nanoparticles: quantification of the concentration dependence of contrast enhancement for binding to sparse cellular epitopes. Ultrasound Med Biol. 2007;33(6):950–8.

    Article  PubMed Central  PubMed  Google Scholar 

  106. Alexandrov AV, Demchuk AM, Burgin WS, Robinson DJ, Grotta JC, CLOTBUST Investigators. Ultrasound-enhanced thrombolysis for acute ischemic stroke: phase I. Findings of the CLOTBUST trial. J Neuroimaging. 2004;14(2):113–7.

    Article  PubMed  Google Scholar 

  107. Alexandrov AV. Ultrasound-enhanced thrombolysis for stroke: clinical significance. Eur J Ultrasound. 2002;16(1–2):131–40.

    Article  PubMed  Google Scholar 

  108. Alexandrov AV. Ultrasound enhanced thrombolysis for stroke. Int J Stroke. 2005;1(1):26–9.

    Article  Google Scholar 

  109. Eibel B, Rodrigues CG, Giusti II, Nesralla IA, Prates PR, Sant Anna RT, et al. Gene therapy for ischemic heart disease: review of clinical trials. Rev Bras Cir Cardiovasc. 2011;26(4):635–46.

    Article  PubMed  Google Scholar 

  110. Masumu M, Hata R. Recent advances in adenovirus-mediated gene therapy for cerebral ischemia. Curr Gene Ther. 2003;3(1):43–8.

    Article  PubMed  Google Scholar 

  111. Yenari MA, Dumas TC, Sapolsky RM, Steinberg GK. Gene therapy for treatment of cerebral ischemia using defective herpes simplex viral vectors. Ann N Y Acad Sci. 2001;939:340–57.

    Article  CAS  PubMed  Google Scholar 

  112. Coles JP, Fryer TD, Smielewski P, Chatfield DA, Steiner LA, Johnston AJ, et al. Incidence and mechanisms of cerebral ischemia in early clinical head injury. J Cereb Blood Flow Metab. 2004;24(2):202–11.

    Article  PubMed  Google Scholar 

  113. Soler EP, Ruiz VC. Epidemiology and risk factors of cerebral ischemia and ischemic heart diseases: similarities and differences. Curr Cardiol Rev. 2010;6(3):138–49.

    Article  PubMed Central  PubMed  Google Scholar 

  114. European Stroke Organisation. ESO guidelines for the management of ischaemic stroke. Basel: European Stroke Organisation; 2008.

    Google Scholar 

  115. Perry VH, Newman TA, Cunningham C. The impact of systemic infection on the progression of neurodegenerative disease. Nat Rev Neurosci. 2003;4(2):103–212.

    Article  CAS  PubMed  Google Scholar 

  116. Teeling JL, Perry VH. Systemic infection and inflammation in acute CNS injury and chronic neurodegeneration: underlying mechanisms. Neuroscience. 2009;158(3):1062–73.

    Article  CAS  PubMed  Google Scholar 

  117. Mattson MP, Duan W, Pedersen WA, Culmsee C. Neurodegenerative disorders and ischemic brain diseases. Apoptosis. 2001;6(1–2):69–81.

    Article  CAS  PubMed  Google Scholar 

  118. Goldstein LB. Modern medical management of acute ischemic stroke. Methodist Debakey Cardiovasc J. 2014;10(2):99–104.

    Article  PubMed Central  PubMed  Google Scholar 

  119. Johnston DCC, Goldstein LB. Clinical carotid endarterectomy decision making: noninvasive vascular imaging versus angiography. Neurology. 2001;56(8):1009–15.

    Article  CAS  PubMed  Google Scholar 

  120. Albers GW, Caplan LR, Easton JD, Fayad PB, Mohr JP, Saver JL, et al. Transient ischemic attack—proposal for a new definition. N Engl J Med. 2002;347(21):1713–6.

    Article  PubMed  Google Scholar 

  121. Kidwell CS, Latour L, Saver JL, Alger JR, Starkman S, Duckwiler G, et al. Thrombolytic toxicity: blood brain barrier disruption in human ischemic stroke. Cerebrovasc Dis. 2008;25(4):338–43.

    Article  CAS  PubMed  Google Scholar 

  122. Kastrup A, Groschel K, Ringer TM, Redecker C, Cordesmeyer R, Witte OW, et al. Early disruption of the blood–brain barrier after thrombolytic therapy predicts hemorrhage in patients with acute stroke. Stroke. 2008;39(8):2385–7.

    Article  CAS  PubMed  Google Scholar 

  123. Johnston SC, Gress DR, Browner WS, Sidney S. Short-term prognosis after emergency department diagnosis of TIA. JAMA. 2000;284(22):2901–6.

    Article  CAS  PubMed  Google Scholar 

  124. Gorelick PB. The burden and management of TIA and stroke in government-funded healthcare programs. Am J Manage Care. 2009;15(6 Suppl):S177–84.

    Google Scholar 

  125. Power A, Epstein D, Cohen D, Bathula R, Devine J, Kar A, et al. Renal impairment reduces the efficacy of thrombolytic therapy in acute ischemic stroke. Cerebrovasc Dis. 2013;35(1):45–52.

    Article  CAS  PubMed  Google Scholar 

  126. Masrur S, Abdullah AR, Smith EE, Hidalgo R, El-Ghandour A, Rordorf G, et al. Risk of thrombolytic therapy for acute ischemic stroke in patients with current malignancy. J Stroke Cerebrovasc Dis. 2011;20(2):124–30.

    Article  PubMed  Google Scholar 

  127. Ross OA, Worrall BB, Meschia JF. Advancing stroke therapeutics through genetic understanding. Curr Drug Targets. 2007;8(7):850–9.

    Article  CAS  PubMed  Google Scholar 

  128. Robinson T, Zaheer Z, Mistri AK. Thrombolysis in acute ischaemic stroke: an update. Ther Adv Chronic Dis. 2011;2(2):119–31.

    Article  PubMed Central  PubMed  Google Scholar 

  129. Alyautdin R, Khalin I, Nafeeza MI, Haron MH, Kuznetsov D. Nanoscale drug delivery systems and the blood–brain barrier. Int J Nanomedicine. 2014;9:795–811.

    PubMed Central  CAS  PubMed  Google Scholar 

  130. Chen Y, Liu L. Modern methods for delivery of drugs across the blood–brain barrier. Adv Drug Deliv Rev. 2012;64(7):640–65.

    Article  CAS  PubMed  Google Scholar 

  131. Alavijeh MS, Chishty M, Qaiser MZ, Palmer AM. Drug metabolism and pharmacokinetics, the blood–brain barrier, and central nervous system drug discovery. NeuroRx. 2005;2(4):554–71.

    Article  PubMed Central  PubMed  Google Scholar 

  132. Smith WS, Sung G, Saver J, Budzik R, Duckwiler G, Liebeskind DS, et al. Mechanical thrombectomy for acute ischemic stroke: final results of the multi MERCI trial. Stroke. 2008;39:1205–12.

    Article  PubMed  Google Scholar 

  133. Penumbra Pivotal Stroke Trial Investigators. The penumbra pivotal stroke trial: safety and effectiveness of a new generation of mechanical devices for clot removal in intracranial large vessel occlusive disease. Stroke. 2009;40:2761–8.

    Article  Google Scholar 

  134. Pereira VM, Gralla J, Davalos A, Bonafé A, Castaño C, Chapot R, et al. Prospective, multicenter, single-arm study of mechanical thrombectomy using solitaire flow restoration in acute ischemic stroke. Stroke. 2013;44(10):2802–7.

    Article  PubMed Central  PubMed  Google Scholar 

  135. Gralla J, Schroth G, Remonda L, Nedeltchev K, Slotboom J, Brekenfeld C. Mechanical thrombectomy for acute ischemic stroke, thrombus–device interaction, efficiency, and complications in vivo. Stroke. 2006;37:3019–24.

    Article  PubMed  Google Scholar 

  136. Mijajlovic MD, Pavlovic A, Covickovic-Sternic N. Is sonothrombolysis an effective stroke treatment? J Ultrasound Med. 2013;32:1117–23.

    Article  PubMed  Google Scholar 

  137. Rubiera M, Alexandrov AV. Sonothrombolysis in the management of acute ischemic stroke. Am J Cardiovasc Drugs. 2010;10(1):5–10.

    Article  PubMed  Google Scholar 

  138. Barlinn K, Alexandrov AV. Sonothrombolysis in ischemic stroke. Curr Treat Options Neurol. 2013;15(2):91–103.

    Article  PubMed  Google Scholar 

  139. Ahadi G, Welch CS, Grimm MJ, Fisher DJ, Zadicario E, Ernstrom K, et al. Transcranial sonothrombolysis using high-intensity focused ultrasound: impact of increasing output power on clot fragmentation. J Ther Ultrasound. 2013;1:22.

    Article  PubMed Central  PubMed  Google Scholar 

Download references

ACKNOWLEDGMENTS

The author thanks the Principal and Management, R.V.R. & J.C. College of Engineering (A), Guntur, Andhra Pradesh, India for their support to carry out the research work. Furthermore, my deep gratitude to the Department of Biotechnology, R.V.R. & J.C. College of Engineering (A) for providing facility utilized in the current study.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to K. K. Pulicherla.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pulicherla, K.K., Verma, M.K. Targeting Therapeutics Across the Blood Brain Barrier (BBB), Prerequisite Towards Thrombolytic Therapy for Cerebrovascular Disorders—an Overview and Advancements. AAPS PharmSciTech 16, 223–233 (2015). https://doi.org/10.1208/s12249-015-0287-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12249-015-0287-z

KEY WORDS

Navigation