Skip to main content
Top
Published in: Fluids and Barriers of the CNS 1/2017

Open Access 01-12-2017 | Review

The opioid epidemic: a central role for the blood brain barrier in opioid analgesia and abuse

Authors: Charles P. Schaefer, Margaret E. Tome, Thomas P. Davis

Published in: Fluids and Barriers of the CNS | Issue 1/2017

Login to get access

Abstract

Opioids are currently the primary treatment method used to manage both acute and chronic pain. In the past two to three decades, there has been a surge in the use, abuse and misuse of opioids. The mechanism by which opioids relieve pain and induce euphoria is dependent on the drug crossing the blood–brain barrier and accessing the central nervous system. This suggests the blood brain barrier plays a central role in both the benefits and risks of opioid use. The complex physiological responses to opioids that provide the benefits and drive the abuse also needs to be considered in the resolution of the opioid epidemic.
Literature
2.
go back to reference Rudd RA, Aleshire N, Zibbell JE, Gladden RM. Increases in drug and opioid overdose deaths—United States, 2000–2014. MMWR Morb Mortal Wkly Rep. 2016;64:1378–82.CrossRefPubMed Rudd RA, Aleshire N, Zibbell JE, Gladden RM. Increases in drug and opioid overdose deaths—United States, 2000–2014. MMWR Morb Mortal Wkly Rep. 2016;64:1378–82.CrossRefPubMed
3.
go back to reference Joshi GP, Beck D, Emerson R, Halaszynki T, Jahr J, Lipman A, et al. Defining new directions for more effective management of surgical pain in the United States: highlights of the inaugural surgical pain congress ä. Highlights Inaug Surg Pain Congr. 2014;80:219–28. Joshi GP, Beck D, Emerson R, Halaszynki T, Jahr J, Lipman A, et al. Defining new directions for more effective management of surgical pain in the United States: highlights of the inaugural surgical pain congress ä. Highlights Inaug Surg Pain Congr. 2014;80:219–28.
7.
go back to reference Schiff PL. Opium and its alkaloids. Am J Pharm Educ. 2002;66:186–94. Schiff PL. Opium and its alkaloids. Am J Pharm Educ. 2002;66:186–94.
8.
go back to reference Sertuner F. Trommsdorff’s J Pharm. 1806;47–93. Sertuner F. Trommsdorff’s J Pharm. 1806;47–93.
9.
go back to reference Wright CRA. On the action of organic acids and their anhydrides on the natural alkaloids. Part I. J Chem Soc. 1872;27:1031–43. Wright CRA. On the action of organic acids and their anhydrides on the natural alkaloids. Part I. J Chem Soc. 1872;27:1031–43.
10.
go back to reference Bockmuhl VM, Ehrhart G. Uber eine neue Klasse von spasmolytisch und aiialgetisch wirkenden Verbindungen. I. Eur J Org Chem. 1947;561:52–85. Bockmuhl VM, Ehrhart G. Uber eine neue Klasse von spasmolytisch und aiialgetisch wirkenden Verbindungen. I. Eur J Org Chem. 1947;561:52–85.
11.
go back to reference Fugelstad A, Stenbacka M, Leifman A, Nylander M, Thiblin I. Methadone maintenance treatment: the balance between life-saving treatment and fatal poisonings. Addiction. 2007;102:406–12.CrossRefPubMed Fugelstad A, Stenbacka M, Leifman A, Nylander M, Thiblin I. Methadone maintenance treatment: the balance between life-saving treatment and fatal poisonings. Addiction. 2007;102:406–12.CrossRefPubMed
13.
go back to reference Porter J, Jick H. Addiction rare in patients treated with narcotics. N Engl J Med. 1980;302:123.PubMed Porter J, Jick H. Addiction rare in patients treated with narcotics. N Engl J Med. 1980;302:123.PubMed
14.
go back to reference Zenz M, Strumpf M, Tryba M. Long-term oral opioid therapy in patients with chronic nonmalignant pain. J Pain Symptom Manage. 1992;7:69–77.CrossRefPubMed Zenz M, Strumpf M, Tryba M. Long-term oral opioid therapy in patients with chronic nonmalignant pain. J Pain Symptom Manage. 1992;7:69–77.CrossRefPubMed
15.
go back to reference Portenoy RK, Foley KM. Chronic use of opioid analgesics in non-malignant pain [letter]. Pain. 1987;29:257–62.CrossRef Portenoy RK, Foley KM. Chronic use of opioid analgesics in non-malignant pain [letter]. Pain. 1987;29:257–62.CrossRef
16.
go back to reference Weingarten MA. Chrnoic opioid therapy in patients with a remote history of substance abuse. J Pain Symptom Manage. 1991;6:2–3.CrossRefPubMed Weingarten MA. Chrnoic opioid therapy in patients with a remote history of substance abuse. J Pain Symptom Manage. 1991;6:2–3.CrossRefPubMed
19.
go back to reference Yaksh TL, Wallace MS. Chapter 18 : opioids, analgesia, and pain management. In: Goodman and Gilman’s: the pharmacological basis of therapeutics, 12th ed. New York: McGraw-Hill; 2011. Yaksh TL, Wallace MS. Chapter 18 : opioids, analgesia, and pain management. In: Goodman and Gilman’s: the pharmacological basis of therapeutics, 12th ed. New York: McGraw-Hill; 2011.
20.
go back to reference Osbourne R, Joel S, Trew D, Slevin M. analgesic activity of morphine-6-glucuronide. Lancet. 1988;1:828.CrossRef Osbourne R, Joel S, Trew D, Slevin M. analgesic activity of morphine-6-glucuronide. Lancet. 1988;1:828.CrossRef
21.
go back to reference Smith MT. Neuroexcitatory effects of morphine and hydromorphone: evidence implicating the 3-glucuronide metabolites. Clin Exp Pharmacol Physiol. 2000;27:524–8.CrossRefPubMed Smith MT. Neuroexcitatory effects of morphine and hydromorphone: evidence implicating the 3-glucuronide metabolites. Clin Exp Pharmacol Physiol. 2000;27:524–8.CrossRefPubMed
22.
go back to reference Sia AT, Lim Y, Lim ECP, Goh RWC, Law HY, Landau R, et al. A118G single nucleotide polymorphism of human mu-opioid receptor gene influences pain perception and patient-controlled intravenous morphine consumption after intrathecal morphine for postcesarean analgesia. Anesthesiology. 2008;109:520–6.CrossRefPubMed Sia AT, Lim Y, Lim ECP, Goh RWC, Law HY, Landau R, et al. A118G single nucleotide polymorphism of human mu-opioid receptor gene influences pain perception and patient-controlled intravenous morphine consumption after intrathecal morphine for postcesarean analgesia. Anesthesiology. 2008;109:520–6.CrossRefPubMed
23.
go back to reference Yaksh TL. Pharmacology and mechanisms of opioid analgesic activity. Acta Anaesthesiol Scand. 1997;41(1 Pt 2):94–111.CrossRefPubMed Yaksh TL. Pharmacology and mechanisms of opioid analgesic activity. Acta Anaesthesiol Scand. 1997;41(1 Pt 2):94–111.CrossRefPubMed
24.
go back to reference Bundgaard M, Abbott NJ. All vertebrates started out with a glial blood–brain barrier 4–500 million years ago. Glia. 2008;56:699–708.CrossRefPubMed Bundgaard M, Abbott NJ. All vertebrates started out with a glial blood–brain barrier 4–500 million years ago. Glia. 2008;56:699–708.CrossRefPubMed
25.
go back to reference Mayer F, Mayer N, Chinn L, Pinsonneault RL, Bainton RJ. Evolutionary conservation of vertebrate blood–brain barrier chemoprotective mechanisms in Drosophila. J Neurosci. 2011;29:3538–50.CrossRef Mayer F, Mayer N, Chinn L, Pinsonneault RL, Bainton RJ. Evolutionary conservation of vertebrate blood–brain barrier chemoprotective mechanisms in Drosophila. J Neurosci. 2011;29:3538–50.CrossRef
26.
go back to reference Mahringer A, Ott M, Fricker G. The blood brain barrier (BBB). Heidelberg: Springer; 2014. p. 1–20. Mahringer A, Ott M, Fricker G. The blood brain barrier (BBB). Heidelberg: Springer; 2014. p. 1–20.
27.
go back to reference Seleman M, Chapy H, Cisternino S, Courtin C, Smirnova M, Schlatter J, et al. Impact of P-glycoprotein at the blood–brain barrier on the uptake of heroin and its main metabolites: behavioral effects and consequences on the transcriptional responses and reinforcing properties. Psychopharmacology. 2014;231:3139–49.CrossRefPubMed Seleman M, Chapy H, Cisternino S, Courtin C, Smirnova M, Schlatter J, et al. Impact of P-glycoprotein at the blood–brain barrier on the uptake of heroin and its main metabolites: behavioral effects and consequences on the transcriptional responses and reinforcing properties. Psychopharmacology. 2014;231:3139–49.CrossRefPubMed
28.
go back to reference Campbell AW. The blood–brain barrier. Altern Ther. 2016;22:6–7. Campbell AW. The blood–brain barrier. Altern Ther. 2016;22:6–7.
29.
go back to reference Nakagawa S, Deli MA, Kawaguchi H, Shimizudani T, Shimono T, Kittel Á, et al. A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int. 2009;54:253–63.CrossRefPubMed Nakagawa S, Deli MA, Kawaguchi H, Shimizudani T, Shimono T, Kittel Á, et al. A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int. 2009;54:253–63.CrossRefPubMed
30.
go back to reference Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37:13–25.CrossRefPubMed Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37:13–25.CrossRefPubMed
31.
go back to reference Dick AP, Harik SI, Klip A, Walker DM. Identification and characterization of the glucose transporter of the blood–brain barrier by cytochalasin B binding and immunological reactivity. Proc Natl Acad Sci USA. 1984;81:7233–7.CrossRefPubMedPubMedCentral Dick AP, Harik SI, Klip A, Walker DM. Identification and characterization of the glucose transporter of the blood–brain barrier by cytochalasin B binding and immunological reactivity. Proc Natl Acad Sci USA. 1984;81:7233–7.CrossRefPubMedPubMedCentral
32.
go back to reference Jones PM, George AM. The ABC transporter structure and mechanism: perspectives on recent research. Cell Mol Life Sci. 2004;61:682–99.CrossRefPubMed Jones PM, George AM. The ABC transporter structure and mechanism: perspectives on recent research. Cell Mol Life Sci. 2004;61:682–99.CrossRefPubMed
33.
go back to reference Cordon-Cardo C, O’Brien JP, Casals D, Rittman-Grauer L, Biedler JL, Melamed MR, et al. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood–brain barrier sites. Proc Natl Acad Sci USA. 1989;86:695–8.CrossRefPubMedPubMedCentral Cordon-Cardo C, O’Brien JP, Casals D, Rittman-Grauer L, Biedler JL, Melamed MR, et al. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood–brain barrier sites. Proc Natl Acad Sci USA. 1989;86:695–8.CrossRefPubMedPubMedCentral
34.
go back to reference Schinkel AH, Smit JJM, van Tellingen O, Beijnen JH, Wagenaar E, van Deemter L, et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood–brain barrier and to increased sensitivity to drugs. Cell. 1994;77:491–502.CrossRefPubMed Schinkel AH, Smit JJM, van Tellingen O, Beijnen JH, Wagenaar E, van Deemter L, et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood–brain barrier and to increased sensitivity to drugs. Cell. 1994;77:491–502.CrossRefPubMed
36.
go back to reference Ambudkar SV, Kim I, Sauna ZE. The power of the pump: mechanisms of action of P-glycoprotein (ABCB1). Eur J Pharm Sci. 2006;27:392–400.CrossRefPubMed Ambudkar SV, Kim I, Sauna ZE. The power of the pump: mechanisms of action of P-glycoprotein (ABCB1). Eur J Pharm Sci. 2006;27:392–400.CrossRefPubMed
37.
go back to reference Thomas H, Coley H. Overcoming multidrug resistance in cancer: an update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control. 2003;10:159–65.CrossRefPubMed Thomas H, Coley H. Overcoming multidrug resistance in cancer: an update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control. 2003;10:159–65.CrossRefPubMed
38.
39.
go back to reference Yamada H, Ishii K, Ishii Y, Ieiri I, Nishio S, Morioka T, et al. Formation of highly analgesic morphine-6-glucuronide following physiologic concentration of morphine in human brain. J Toxicol Sci. 2003;28:395–401.CrossRefPubMed Yamada H, Ishii K, Ishii Y, Ieiri I, Nishio S, Morioka T, et al. Formation of highly analgesic morphine-6-glucuronide following physiologic concentration of morphine in human brain. J Toxicol Sci. 2003;28:395–401.CrossRefPubMed
40.
go back to reference Tournier N, Chevillard L, Megarbane B, Scherrmann J, Pirnay S, Decleves X. Interaction of drugs of abuse and maintenance treatments with human P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2). Int J Neurophychopharmacology. 2010;13:905–15.CrossRef Tournier N, Chevillard L, Megarbane B, Scherrmann J, Pirnay S, Decleves X. Interaction of drugs of abuse and maintenance treatments with human P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2). Int J Neurophychopharmacology. 2010;13:905–15.CrossRef
41.
go back to reference Fujita KI, Ando Y, Yamamoto W, Miya T, Endo H, Sunakawa Y, et al. Association of UGT2B7 and ABCB1 genotypes with morphine-induced adverse drug reactions in Japanese patients with cancer. Cancer Chemother Pharmacol. 2010;65:251–8.CrossRefPubMed Fujita KI, Ando Y, Yamamoto W, Miya T, Endo H, Sunakawa Y, et al. Association of UGT2B7 and ABCB1 genotypes with morphine-induced adverse drug reactions in Japanese patients with cancer. Cancer Chemother Pharmacol. 2010;65:251–8.CrossRefPubMed
42.
go back to reference De Gregori S, De Gregori M, Ranzani GN, Allegri M, Minella C, Regazzi M. Morphine metabolism, transport and brain disposition. Metab Brain Dis. 2012;27:1–5.CrossRefPubMed De Gregori S, De Gregori M, Ranzani GN, Allegri M, Minella C, Regazzi M. Morphine metabolism, transport and brain disposition. Metab Brain Dis. 2012;27:1–5.CrossRefPubMed
43.
go back to reference Bourasset F, Cisternino S, Temsamani J, Scherrmann J. Evidence for an active transport of morphine-6-β-d-glucuronide but not P-glycoprotein-mediated at the blood–brain barrier. J Neurochem. 2003;86:1564–7.CrossRefPubMed Bourasset F, Cisternino S, Temsamani J, Scherrmann J. Evidence for an active transport of morphine-6-β-d-glucuronide but not P-glycoprotein-mediated at the blood–brain barrier. J Neurochem. 2003;86:1564–7.CrossRefPubMed
44.
go back to reference Balayssac D, Cayre A, Ling B, Maublant J, Penault-Llorca F, Eschalier A, et al. Increase in morphine antinociceptive activity by a P-glycoprotein inhibitor in cisplatin-induced neuropathy. Neurosci Lett. 2009;465:108–12.CrossRefPubMed Balayssac D, Cayre A, Ling B, Maublant J, Penault-Llorca F, Eschalier A, et al. Increase in morphine antinociceptive activity by a P-glycoprotein inhibitor in cisplatin-induced neuropathy. Neurosci Lett. 2009;465:108–12.CrossRefPubMed
46.
go back to reference Wandel C, Kim R, Wood M, Ch MBB, Wood A, Ch MBB. Interaction of morphine, fentanyl, sufentanil, alfentanil, and loperamide with the efflux drug transporter P-glycoprotein. Anesthesiology. 2002;86:913–20.CrossRef Wandel C, Kim R, Wood M, Ch MBB, Wood A, Ch MBB. Interaction of morphine, fentanyl, sufentanil, alfentanil, and loperamide with the efflux drug transporter P-glycoprotein. Anesthesiology. 2002;86:913–20.CrossRef
47.
go back to reference Montesinos RN, Moulari B, Gromand J, Beduneau A, Lamprecht A. Coadministration of P-glycoprotein modulators on loperamide pharmacokinetics and brain distribution. Drug Metab Dispos. 2014;42:700–6.CrossRefPubMed Montesinos RN, Moulari B, Gromand J, Beduneau A, Lamprecht A. Coadministration of P-glycoprotein modulators on loperamide pharmacokinetics and brain distribution. Drug Metab Dispos. 2014;42:700–6.CrossRefPubMed
48.
go back to reference Regnard C, Twycross R, Mihalyo M, Wilcock A. Loperamide. J Pain Symptom Manag. 2011;42:319–23.CrossRef Regnard C, Twycross R, Mihalyo M, Wilcock A. Loperamide. J Pain Symptom Manag. 2011;42:319–23.CrossRef
49.
go back to reference Schinkel AH, Wagenaar E, Mol CAAM, Van Deemter L. P-Glycoprotein in the blood–brain barrier of mice influences the brain penetration and pharmacological activity of many drugs. J Clin Invest. 1996;97:2517–24.CrossRefPubMedPubMedCentral Schinkel AH, Wagenaar E, Mol CAAM, Van Deemter L. P-Glycoprotein in the blood–brain barrier of mice influences the brain penetration and pharmacological activity of many drugs. J Clin Invest. 1996;97:2517–24.CrossRefPubMedPubMedCentral
50.
go back to reference Kaiko RF, Wallenstein SL, Rogers A. Relative analgesic potency of intramuscular heroin and morphine in cancer patients with postoperative pain and chronic pain due to cancer. NIDA Res Minigr Ser. 1981;34:213–9. Kaiko RF, Wallenstein SL, Rogers A. Relative analgesic potency of intramuscular heroin and morphine in cancer patients with postoperative pain and chronic pain due to cancer. NIDA Res Minigr Ser. 1981;34:213–9.
51.
go back to reference Selley DE, Cao CC, Sexton T, Schwegel JA, Martin TJ, Childers SR. μ opioid receptor-mediated G-protein activation by heroin metabolites: evidence for greater efficacy of 6-monoacetylmorphine compared with morphine. Biochem Pharmacol. 2001;62:447–55.CrossRefPubMed Selley DE, Cao CC, Sexton T, Schwegel JA, Martin TJ, Childers SR. μ opioid receptor-mediated G-protein activation by heroin metabolites: evidence for greater efficacy of 6-monoacetylmorphine compared with morphine. Biochem Pharmacol. 2001;62:447–55.CrossRefPubMed
52.
go back to reference Boix F, Andersen JM, Mørland J. Pharmacokinetic modeling of subcutaneous heroin and its metabolites in blood and brain of mice. Addict Biol. 2013;18:1–7.CrossRefPubMed Boix F, Andersen JM, Mørland J. Pharmacokinetic modeling of subcutaneous heroin and its metabolites in blood and brain of mice. Addict Biol. 2013;18:1–7.CrossRefPubMed
53.
go back to reference Boström E, Simonsson USH, Hammarlund-Udenaes M. Oxycodone pharmacokinetics and pharmacodynamics in the rat in the presence of the P-glycoprotein inhibitor PSC833. J Pharm Sci. 2005;94:1060–6.CrossRefPubMed Boström E, Simonsson USH, Hammarlund-Udenaes M. Oxycodone pharmacokinetics and pharmacodynamics in the rat in the presence of the P-glycoprotein inhibitor PSC833. J Pharm Sci. 2005;94:1060–6.CrossRefPubMed
54.
go back to reference Silvasti M, Rosenburg P, Seppala T, Svartling N, Pitkanen M. Comparison of analgesic efficacy of oxycodone and morphine in postoperative intravenous patient-controlled analgesia. Acta Anaesthesiol Scand. 1998;42:576–80.CrossRefPubMed Silvasti M, Rosenburg P, Seppala T, Svartling N, Pitkanen M. Comparison of analgesic efficacy of oxycodone and morphine in postoperative intravenous patient-controlled analgesia. Acta Anaesthesiol Scand. 1998;42:576–80.CrossRefPubMed
55.
go back to reference Bostrom E, Simonsson USH, Hamarlund-Udenases M. In vivo blood–brain barrier transport of oxycodone in the rat: indications for active influx and implications for pharmacokinetics/pharmacodynamics. Drug Metab Dispos. 2006;34:1624–31.CrossRefPubMed Bostrom E, Simonsson USH, Hamarlund-Udenases M. In vivo blood–brain barrier transport of oxycodone in the rat: indications for active influx and implications for pharmacokinetics/pharmacodynamics. Drug Metab Dispos. 2006;34:1624–31.CrossRefPubMed
56.
go back to reference Okura T, Hattori A, Takano Y, Sato T, Hammarlund-udenaes M, Terasaki T, et al. Involvement of the pyrilamine transporter, a putative organic cation transporter, in blood–brain barrier transport of oxycodone. Drug Metab Dispos. 2008;36:2005–13.CrossRefPubMed Okura T, Hattori A, Takano Y, Sato T, Hammarlund-udenaes M, Terasaki T, et al. Involvement of the pyrilamine transporter, a putative organic cation transporter, in blood–brain barrier transport of oxycodone. Drug Metab Dispos. 2008;36:2005–13.CrossRefPubMed
57.
go back to reference Hagen NA, Wasylenko E. Methadone: outpatient titration and monitoring strategies in cancer patients. J Pain Symptom Manage. 1999;18:369–75.CrossRefPubMed Hagen NA, Wasylenko E. Methadone: outpatient titration and monitoring strategies in cancer patients. J Pain Symptom Manage. 1999;18:369–75.CrossRefPubMed
58.
go back to reference Wang JS, Ruan Y, Taylor RM, Donovan JL, Markowitz JS, DeVane CL. Brain penetration of methadone (R)- and (S)-enantiomers is greatly increased by P-glycoprotein deficiency in the blood–brain barrier of Abcb1a gene knockout mice. Psychopharmacology. 2004;173:132–8.CrossRefPubMed Wang JS, Ruan Y, Taylor RM, Donovan JL, Markowitz JS, DeVane CL. Brain penetration of methadone (R)- and (S)-enantiomers is greatly increased by P-glycoprotein deficiency in the blood–brain barrier of Abcb1a gene knockout mice. Psychopharmacology. 2004;173:132–8.CrossRefPubMed
59.
go back to reference Sullivan HR, Due SL. Urinary metabolites of dl-methadone in maintenance subjects. J Med Chem. 1973;16:909–13.CrossRefPubMed Sullivan HR, Due SL. Urinary metabolites of dl-methadone in maintenance subjects. J Med Chem. 1973;16:909–13.CrossRefPubMed
61.
go back to reference Crettol S, Digon P, Powell Golay K, Brawand M, Eap CB. In vitro P-glycoprotein-mediated transport of (R)-, (S)-, (R,S)-methadone, LAAM and their main metabolites. Pharmacology. 2007;80:304–11.CrossRefPubMed Crettol S, Digon P, Powell Golay K, Brawand M, Eap CB. In vitro P-glycoprotein-mediated transport of (R)-, (S)-, (R,S)-methadone, LAAM and their main metabolites. Pharmacology. 2007;80:304–11.CrossRefPubMed
62.
go back to reference Mercer SL, Coop A. Opioid analgesics and P-glycoprotein efflux transporters: a potential systems-level contribution to analgesic tolerance. Curr Top Med Chem. 2011;11:1157–64.CrossRefPubMedPubMedCentral Mercer SL, Coop A. Opioid analgesics and P-glycoprotein efflux transporters: a potential systems-level contribution to analgesic tolerance. Curr Top Med Chem. 2011;11:1157–64.CrossRefPubMedPubMedCentral
64.
go back to reference Henthorn TK, Liu Y, Mahapatro M, Ng K. Active transport of fentanyl by the blood–brain barrier 1. J Pharmacol Exp Ther. 1999;289:1084–9.PubMed Henthorn TK, Liu Y, Mahapatro M, Ng K. Active transport of fentanyl by the blood–brain barrier 1. J Pharmacol Exp Ther. 1999;289:1084–9.PubMed
65.
go back to reference Klepstad P, Dale O, Skorpen F, Borchgrevink PC, Kaasa S. Genetic variability and clinical efficacy of morphine. Acta Anaesthesiol Scand. 2005;49:902–8.CrossRefPubMed Klepstad P, Dale O, Skorpen F, Borchgrevink PC, Kaasa S. Genetic variability and clinical efficacy of morphine. Acta Anaesthesiol Scand. 2005;49:902–8.CrossRefPubMed
66.
67.
go back to reference Doi K, Gibbons G. Improving postoperative pain management in orthopedic total joint surgical patients with opioid tolerance using the iowa model of evidence-based practice. ASPAN Natl Conf Abstr. 2014;29:e38. Doi K, Gibbons G. Improving postoperative pain management in orthopedic total joint surgical patients with opioid tolerance using the iowa model of evidence-based practice. ASPAN Natl Conf Abstr. 2014;29:e38.
68.
go back to reference Pasternak GW. Molecular biology of opioid analgesia. J Pain Symptom Manage. 2005;29:2–9.CrossRef Pasternak GW. Molecular biology of opioid analgesia. J Pain Symptom Manage. 2005;29:2–9.CrossRef
69.
go back to reference Yousif S, Saubamea B, Cisternino S, Marie-Claire C, Dauchy S, Scherrmann J-M, et al. Effect of chronic exposure to morphine on the rat blood–brain barrier: focus on the P-glycoprotein. J Neurochem. 2008;107:647–57.CrossRefPubMed Yousif S, Saubamea B, Cisternino S, Marie-Claire C, Dauchy S, Scherrmann J-M, et al. Effect of chronic exposure to morphine on the rat blood–brain barrier: focus on the P-glycoprotein. J Neurochem. 2008;107:647–57.CrossRefPubMed
70.
go back to reference Aquilante CL, Letrent SP, Pollack GM, Brouwer KL. Increased brain P-glycoprotein in morphine tolerant rats. Life Sci. 2000;66:L47–51. Aquilante CL, Letrent SP, Pollack GM, Brouwer KL. Increased brain P-glycoprotein in morphine tolerant rats. Life Sci. 2000;66:L47–51.
71.
go back to reference Li Y, Yue H, Xing Y, Sun H, Pan Z, Xie G. Oxymatrine inhibits development of morphine-induced tolerance associated with decreased expression of P-glycoprotein in rats. Integr Cancer Ther. 2010;9:213–8.CrossRefPubMed Li Y, Yue H, Xing Y, Sun H, Pan Z, Xie G. Oxymatrine inhibits development of morphine-induced tolerance associated with decreased expression of P-glycoprotein in rats. Integr Cancer Ther. 2010;9:213–8.CrossRefPubMed
72.
go back to reference Nakagawa T, Ozawa T, Shige K, Yamamoto R, Minami M. Inhibition of morphine tolerance and dependence by MS-153, a glutamate transporter activator. Eur J Pharmacol. 2001;419:39–45.CrossRefPubMed Nakagawa T, Ozawa T, Shige K, Yamamoto R, Minami M. Inhibition of morphine tolerance and dependence by MS-153, a glutamate transporter activator. Eur J Pharmacol. 2001;419:39–45.CrossRefPubMed
73.
go back to reference Chaves C, Gomez-Zepeda D, Auvity S, Menet M-C, Crete D, Labat L, et al. Effect of subchronic intravenous morphine infusion and naloxone-precipitated morphine withdrawal on P-gp and Bcrp at the rat blood–brain barrier. J Pharm Sci. 2015;105:350–8.CrossRef Chaves C, Gomez-Zepeda D, Auvity S, Menet M-C, Crete D, Labat L, et al. Effect of subchronic intravenous morphine infusion and naloxone-precipitated morphine withdrawal on P-gp and Bcrp at the rat blood–brain barrier. J Pharm Sci. 2015;105:350–8.CrossRef
74.
go back to reference Kahan M, Srivastava A, Wilson L, Gourlay D, Midmer D. Misuse of and dependence on opioids: study of chronic pain patients. Can Fam Physician. 2006;52(9):1081–7.PubMedPubMedCentral Kahan M, Srivastava A, Wilson L, Gourlay D, Midmer D. Misuse of and dependence on opioids: study of chronic pain patients. Can Fam Physician. 2006;52(9):1081–7.PubMedPubMedCentral
75.
go back to reference Gupta A, Christo PJ. Opioid abuse and dependence. J Nurse Pract. 2009;5:132–4.CrossRef Gupta A, Christo PJ. Opioid abuse and dependence. J Nurse Pract. 2009;5:132–4.CrossRef
76.
go back to reference Author. Substance-related and addictive disorders. In: Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013. Author. Substance-related and addictive disorders. In: Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.
77.
go back to reference Sonne S, Brady K. Substance abuse and bipolar comorbidity. Psychiatr Clin N Am. 1999;22:609–27.CrossRef Sonne S, Brady K. Substance abuse and bipolar comorbidity. Psychiatr Clin N Am. 1999;22:609–27.CrossRef
78.
go back to reference Martins SS, Keyes KM, Storr CL, Zhu H, Chilcoat HD. Pathways between nonmedical opioid use/dependence and psychiatric disorders: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Drug Alcohol Depend. 2009;103:16–24.CrossRefPubMedPubMedCentral Martins SS, Keyes KM, Storr CL, Zhu H, Chilcoat HD. Pathways between nonmedical opioid use/dependence and psychiatric disorders: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Drug Alcohol Depend. 2009;103:16–24.CrossRefPubMedPubMedCentral
84.
85.
go back to reference Han H, Kass PH, Wilsey BL, Li C-S. Increasing trends in Schedule II opioid use and doctor shopping during 1999–2007 in California. Pharmacoepidemiol Drug Saf. 2014;23:26–35.CrossRefPubMed Han H, Kass PH, Wilsey BL, Li C-S. Increasing trends in Schedule II opioid use and doctor shopping during 1999–2007 in California. Pharmacoepidemiol Drug Saf. 2014;23:26–35.CrossRefPubMed
86.
go back to reference Aldridge J, Decary-Hetu D. Hidden wholesale: the drug diffusing capacity of online drug cryptomarkets. Int J Drug Policy. 2016;35:7–15.CrossRefPubMed Aldridge J, Decary-Hetu D. Hidden wholesale: the drug diffusing capacity of online drug cryptomarkets. Int J Drug Policy. 2016;35:7–15.CrossRefPubMed
87.
go back to reference Lange A, Lasser KE, Xuan Z, Khalid L, Beers D, Heymann OD, et al. Variability in opioid prescription monitoring and evidence of aberrant medication taking behaviors in urban safety-net clinics. Pain. 2015;156:335–40.CrossRefPubMedPubMedCentral Lange A, Lasser KE, Xuan Z, Khalid L, Beers D, Heymann OD, et al. Variability in opioid prescription monitoring and evidence of aberrant medication taking behaviors in urban safety-net clinics. Pain. 2015;156:335–40.CrossRefPubMedPubMedCentral
88.
go back to reference Katz N, Panas L, Kim M, Audet A, Bilansky A, Eadie J, et al. Usefulness of prescription monitoring programs for surveillance—analysis of Schedule II opioid prescription data in Massachusetts, 1996–2006. Pharmacoepidemiol Drug Saf. 2010;19:115–23.CrossRefPubMed Katz N, Panas L, Kim M, Audet A, Bilansky A, Eadie J, et al. Usefulness of prescription monitoring programs for surveillance—analysis of Schedule II opioid prescription data in Massachusetts, 1996–2006. Pharmacoepidemiol Drug Saf. 2010;19:115–23.CrossRefPubMed
89.
91.
go back to reference Jones CM, Lurie PG, Throckmorton DC. Effect of US drug enforcement administration’s rescheduling of hydrocodone combination analgesic products on opioid analgesic prescribing. J Am Med Assoc Intern Med. 2017;176:399–402. Jones CM, Lurie PG, Throckmorton DC. Effect of US drug enforcement administration’s rescheduling of hydrocodone combination analgesic products on opioid analgesic prescribing. J Am Med Assoc Intern Med. 2017;176:399–402.
92.
94.
go back to reference Cicero TJ, Ellis MS, Harney J. Shifting patterns of prescription opioid and heroin abuse in the United States. N Engl J Med. 2015;373:1789–90.CrossRefPubMed Cicero TJ, Ellis MS, Harney J. Shifting patterns of prescription opioid and heroin abuse in the United States. N Engl J Med. 2015;373:1789–90.CrossRefPubMed
96.
go back to reference Spahn V, Massaly N, Temp J, Durmaz V, Sabri P, Reidelbach M, et al. A nontoxic pain killer designed by modeling of pathological receptor conformations. Science (80 −). 2017;355:966–9.CrossRef Spahn V, Massaly N, Temp J, Durmaz V, Sabri P, Reidelbach M, et al. A nontoxic pain killer designed by modeling of pathological receptor conformations. Science (80 −). 2017;355:966–9.CrossRef
97.
go back to reference Kieffer BL, Gavériaux-ruff C. Exploring the opioid system by gene knockout. Prog Neurobiol. 2002;66:285–306.CrossRefPubMed Kieffer BL, Gavériaux-ruff C. Exploring the opioid system by gene knockout. Prog Neurobiol. 2002;66:285–306.CrossRefPubMed
98.
go back to reference Oeltjenbruns J, Schäfer M. Peripheral opioid analgesia: clinical applications. Anesth Tech Pain Manag. 2005;9:36–44. Oeltjenbruns J, Schäfer M. Peripheral opioid analgesia: clinical applications. Anesth Tech Pain Manag. 2005;9:36–44.
99.
go back to reference Arti H, Mehdinasab SA. The comparison effects of intra-articular injection of different opioids on postoperative pain relieve after arthroscopic anterior cruciate ligament reconstruction: a randomized clinical trial study. J Res Med Sci. 2011;16:1176–82.PubMedPubMedCentral Arti H, Mehdinasab SA. The comparison effects of intra-articular injection of different opioids on postoperative pain relieve after arthroscopic anterior cruciate ligament reconstruction: a randomized clinical trial study. J Res Med Sci. 2011;16:1176–82.PubMedPubMedCentral
100.
go back to reference Reuben SS, Sklar J. Current concepts review pain management in patients who undergo outpatient arthroscopic surgery of the knee. J Bone Jt Surg. 2000;82:1754–66.CrossRef Reuben SS, Sklar J. Current concepts review pain management in patients who undergo outpatient arthroscopic surgery of the knee. J Bone Jt Surg. 2000;82:1754–66.CrossRef
101.
go back to reference Ding H, Czoty PW, Kiguchi N, Cami-kobeci G, Sukhtankar DD, Nader MA. A novel orvinol analog, BU08028, as a safe opioid analgesic without abuse liability in primates. Proc Natl Acad Sci USA. 2016;113:5511–8.CrossRef Ding H, Czoty PW, Kiguchi N, Cami-kobeci G, Sukhtankar DD, Nader MA. A novel orvinol analog, BU08028, as a safe opioid analgesic without abuse liability in primates. Proc Natl Acad Sci USA. 2016;113:5511–8.CrossRef
104.
go back to reference Ong CKS, Seymour RA, Lirk P, Merry AF. Combining paracetamol (acetaminophen) with nonsteroidal antiinflammatory drugs: a qualitative systematic review of analgesic efficacy for acute postoperative pain. Anesth Analg. 2010;110:1170–9.PubMed Ong CKS, Seymour RA, Lirk P, Merry AF. Combining paracetamol (acetaminophen) with nonsteroidal antiinflammatory drugs: a qualitative systematic review of analgesic efficacy for acute postoperative pain. Anesth Analg. 2010;110:1170–9.PubMed
105.
go back to reference Hayden JA, Van Tulder MW, Tomlinson G. Systematic review: strategies for using exercise therapy to improve outcomes in chronic low back pain. Ann Intern Med. 2005;142:776–85.CrossRefPubMed Hayden JA, Van Tulder MW, Tomlinson G. Systematic review: strategies for using exercise therapy to improve outcomes in chronic low back pain. Ann Intern Med. 2005;142:776–85.CrossRefPubMed
106.
go back to reference Ibrahim MM, Patwardhan A, Gilbraith KB, Moutal A, Yang X, Chew LA. Long-lasting antinociceptive effects of green light in acute and chronic pain in rats. Pain. 2017;158:347–60.CrossRefPubMed Ibrahim MM, Patwardhan A, Gilbraith KB, Moutal A, Yang X, Chew LA. Long-lasting antinociceptive effects of green light in acute and chronic pain in rats. Pain. 2017;158:347–60.CrossRefPubMed
Metadata
Title
The opioid epidemic: a central role for the blood brain barrier in opioid analgesia and abuse
Authors
Charles P. Schaefer
Margaret E. Tome
Thomas P. Davis
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Fluids and Barriers of the CNS / Issue 1/2017
Electronic ISSN: 2045-8118
DOI
https://doi.org/10.1186/s12987-017-0080-3

Other articles of this Issue 1/2017

Fluids and Barriers of the CNS 1/2017 Go to the issue