Peripheralization Strategies Applied to Morphinans: Comparison
Please note this is a comparison between Version 3 by Camila Xu and Version 2 by Camila Xu.

Among therapeutically valuable opioids, morphinans are of the utmost clinical importance as analgesic drugs because of their agonistic actions to the mu-opioid receptor. They include powerful pain relieving agents, such as naturally occurring alkaloids (e.g., morphine and codeine), semisynthetic analogues (e.g., hydrocodone, hydromorphone, oxycodone, oxymorphone and buprenorphine), and synthetic derivatives (e.g., levorphanol).

  • pain
  • analgesia
  • opioid receptors
  • peripheral analgesia
  • peripherally restricted opioids
  • morphinans
  • morphine

1. Introduction

Effective and adequate management of pain, particularly chronic pain, is still an area of unmet medical need at the beginning of the third millennium. Opioids are the gold standard for the treatment of moderate to severe acute and chronic pain [1]. However, strong opioid analgesics, such as morphine, oxycodone and fentanyl, are not effective for pain relief in all patients, nor are they well-tolerated by all patients, because of an array of severe side effects, including respiratory depression, constipation, sedation, nausea and dizziness [2]. With prolonged use, opioid safety is dramatically reduced because of analgesic tolerance, and risks of physical dependence and addiction, promoting the development of opioid use disorders and overdose deaths [2]. Moreover, the misuse of prescription opioids (i.e., oxycodone, hydrocodone and fentanyl) [3], as well as over-the-counter opioids (i.e., codeine, hydrocodeine and loperamide) [4], has led to the current opioid epidemic, particularly in North America and Europe. In the USA, the opioid-involved overdose deaths had increased to 80,411 in 2021 [5]. The recent explosion in fatalities resulting from overdose of prescription and synthetic opioids, especially fentanyl and its various analogs [3][5], has dramatically increased the need for safer analgesics.
Opioid receptors represent the most important players in pain modulation and are the molecular targets of clinically used opioids. There are three main opioid receptor types, mu (MOR), delta (DOR) and kappa (KOR), and the non-classical receptor, nociceptin/orphanin FQ (NOP) receptor [1]. All opioid receptor types belong to the family of G protein-coupled receptors (GPCRs) with seven transmembrane domains, and are expressed throughout the central and peripheral nervous systems (CNS and PNS, respectively), and in various non-neuronal tissues [1][6][7]. Because of their therapeutic relevance, opioid receptors are among the few GPCRs determined in different activation states, providing important information on the type-specific binding characteristics of ligands [8]. Although opioid receptors contribute to pain inhibition, the MOR is recognized as the dominant type for its pain-relieving effects [2][9][10]. The major drawback of targeting the MOR for clinical analgesia is that it is also responsible for the undesirable side effects. Most of the detrimental side effects, including respiratory depression, sedation, analgesic tolerance, reward and dependence, are mediated by the MOR in the CNS, whereas constipation is mainly mediated by the MOR in the intestinal myenteric plexus [2][11][12].
Among therapeutically valuable opioids, morphinans are of the utmost clinical importance as analgesic drugs because of their agonistic actions to the MOR. They include powerful pain relieving agents, such as naturally occurring alkaloids (e.g., morphine and codeine), semisynthetic analogues (e.g., hydrocodone, hydromorphone, oxycodone, oxymorphone and buprenorphine), and synthetic derivatives (e.g., levorphanol) (Figure 1) [9][13][14][15]. Morphine and structurally related MOR agonists also share similar side effects, including addictive liability. Because of their outstanding medicinal relevance in combination with an attractive chemical scaffold, morphinan alkaloids represent attractive synthetic targets. Therefore, diverse research approaches toward morphine and its congeners have been devised for mitigating their deleterious side effects and limiting abuse and misuse (for reviews, see [9][13][14][15][16][17][18][19][20]).
Figure 1. Examples of clinically used opioid analgesic drugs from the class of morphinans.
Experimental and clinical studies provide substantial evidence that opioid analgesia is not exclusively mediated via the central opioid receptors (for reviews, see [21][22][23][24]). Pharmacological, neuroanatomical, molecular and electrophysiological studies have shown that peripheral opioid receptors are expressed on the peripheral terminals of sensory neurons, where they can modulate both afferent and efferent neuronal functions, resulting in potent and clinically measurable analgesia. Early clinical studies using intra-articular morphine administration in conjunction with arthroscopy in the knee joint supported the notion that the activation of peripheral opioid receptors induced pain relief by a peripheral mechanism and did so without side effects [25][26]. These findings have led to new research directions aiming on targeting the peripheral opioid receptors for superior pain management.

2. Peripheralization Strategies Applied to Morphinans

Different chemical strategies have been developed to limit the ability of opioids to cross the BBB, including (a) chemical modifications to the morphinan skeleton to increase hydrophilicity of known and new opioids, and (b) nanocarrier-based approaches to selectively deliver opioids, such as morphine to the peripheral tissue. The following sections discuss significant representatives, including design strategies, synthetical procedures, pharmacology and structure–activity relationships (SAR).

2.1. Quaternization of the Morphinan Nitrogen

The first effort to minimize the CNS effects of opioids while retaining their actions in peripheral tissue was the quaternization of the nitrogen in the clinically used morphine, oxymorphone, nalorphine, naloxone and naltrexone (Figure 2) [27][28]. Peripheral selectivity of the quaternary derivative of morphine, N-methylmorphine, was described over 50 years ago [29]. The systemic intravenous (i.v.) administration of N-methylmorphine caused the inhibition of gastrointestinal transit because of its action on the opioid receptors in the gut. In the hot-plate test, centrally mediated antinociception was produced by 15 mg/kg morphine in mice after intraperitoneal (i.p.) administration but not by N-methylmorphine at the same dose [29]. Furthermore, N-methylmorphine proved to be ineffective in the hot-plate test even in a dose 100 mg/kg [30]. In an acetic-acid-induced writhing assay, N-methylmorphine produced antinociceptive effects in mice after i.p. administration in a dose of 45 mg/kg, being 30-fold less potent than morphine [30]. N-methylmorphine was also shown to selectively inhibited phase II in the formalin test following systemic i.p. administration [31]. The antinociceptive effect of N-methylmorphine after central intracerebroventricular (i.c.v.) administration was antagonized by systemically applied naloxone but not by peripheral antagonist N-methylnaloxone, showing the peripheral site of action of N-methylmorphine [31].
Figure 2. Chemical structures of quaternary derivatives of morphine, oxymorphone, nalorphine, naloxone and naltrexone.
It is important to note that quaternization of nitrogen on morphine-based structure derivatives has been reported to have negative impact on both the affinity to the opioid receptor and the agonist activity of generated analogues [28][32]. Quaternization also trends to reduce potency in vivo. Therefore, alternative strategies to limit BBB penetration have been pursued.

2.2. Introduction of Hydrophilic Substituents at Position 6

Polar or ionizable substitutions are able to increase polarity and inhibit the crossing of the BBB. Therefore, opioids with hydrophilic groups attached to the C-6 position of the morphinan skeleton were designed. The first examples of morphinans having ionizable residues at position 6 were reported more than 30 years ago. They were synthesized from β-oxymorphamine [33], β-naltrexamine [33] and β-funaltrexamine [34]. Such compounds with zwitterionic moieties showed significantly reduced access to the CNS without substantially decreased opioid receptor in vitro and in vivo activity [33][34]. Noteworthy are the 6-amide derivatives of β-oxymorphamine (ae, Figure 3) reported as the first peripherally selective opioid agonists and effective antinociceptives [33]. All compounds have C-6 moieties that are ionized at the pH of the gut or at physiologic pH, accounting for a more restricted capability to enter the CNS than the unionized molecules. Compounds a, c and d were synthesized from β-oxymorphamine with the appropriate anhydride [33]. The fumaramic acid b was prepared by coupling the half-ester of fumaric acid with β-oxymorphamine and then subjecting the fumaramate esters to hydrolysis. The aspartyl derivative e was obtained through coupling BocAsp γ-benzyl ester with β-oxymorphamine, followed by deprotection with acid to remove the Boc group and hydrogenolysis of the benzyl function [33]. As regards biological activities, the ß-oxymorphamine derivatives ae were all full agonists in the guinea pig ileum (GPI) bioassay with potencies that were 1.5- to 6-fold higher than the potency of morphine (Table 1) [33]. In a mouse model of acute thermal nociception, the tail-flick assay, all compounds possessed potent antinociceptive activity when administered by the i.c.v route (Table 1). They also were active in inducing antinociception when given systemically by i.v. administration to mice. When compared on a body weight basis, the i.v. ED50 doses were about 1000-fold higher than the i.c.v. ED50 values. Derivatives a and c were also active when given orally (p.o.) (Table 1) [33]. The attachment of polar groups, particularly zwitterionic moieties, at the C-6 position of the morphinan structure is effective in excluding such ligands from the CNS, thereby affording peripheral selectivity.
Figure 3. Structures of β-oxymorphamine and its 6-amide derivatives.
Table 1. In vitro agonist potencies and antinociceptive activities of 6-amide derivatives of β-oxymorphamine (ae).
Other more recent examples of morphinans with ionizable groups at position 6 emerging as peripheral opioid antinociceptives with restricted penetration into the CNS are described in the following sections.

2.2.1. 6-Amino-acid-substituted 14-Alkoxymorphinans

The first synthetic efforts directed towards the development of ionizable molecules in the class of 14-alkoxymorphinans as peripherally acting opioid analgesics started with the series of six 6-amino acids, i.e., Gly-, L-Ala- and L-Phe- substituted derivatives; 2a/b (HS-730/HS-731); 3a/b (HS-935/HS-936); and 4a/b (HS-937/HS-938), respectively, of the highly potent and centrally acting MOR agonist 14-O-methyloxymorphone (14-OMO, 1) (Scheme 1) [35]. A novel synthetic procedure for the synthesis of 6-amino-acid-substituted derivatives in the morphinan series was used. The tert-butyl ester derivatives 2aa/bb, 3aa/bb, and 4aa/bb were prepared from 14-OMO (1) by reductive amination with the respective tert-butyl ester hydrochlorides and sodium cyanoborohydride in ethanol. After separating the diastereoisomers by column chromatography, esters 2aa/bb, 3aa/bb, and 4aa/bb were treated with tetrafluoroboric acid in dichloromethane to afford 6-Gly (2a and 2b), 6-Ala (3a and 3b) and 6-Phe (4a and 4b) substituted derivatives, respectively (Scheme 1) [35].
Scheme 1. Synthesis of 6-Gly-, 6-Ala- and 6-Phe-substituted N-methyl-14-methoxymorphinans (2a/b, 3a/b, and 4a/b, respectively). 14-OMO (1.HBr) was used as starting material. The tert-butyl ester derivatives 2aa/bb, 3aa/bb, and 4aa/bb were prepared from 1 by reductive amination with Gly tert-butyl ester hydrochloride, Ala tert-butyl ester hydrochloride and Phe tert-butyl ester hydrochloride, respectively. Esters were hydrolyzed to yield the corresponding 6-Gly (2a and 2b)-, 6-Ala (3a and 3b)- and 6-Phe (4a and 4b)- substituted derivatives.
The further targeted derivatization of 14-OMO (1) through introduction of other amino acid residues of the L- and/or D-series at position 6, including natural amino acids, i.e., Ser, Val, Lys, Tyr, Trp, Asn, Gln, Asp and Glu (5a13b, Scheme 2), and unnatural amino acids, i.e., D-Ala, D-Val, D-Phe, L-Chg (L-cyclohexylglycine), L-Abu (L-2-aminobutyric acid), β-Ala and GABA (γ-aminobutyric acid) (14a20b, Scheme 2) [36]. Additionally, three zwitterionic molecules with a dipeptide substitution at position 6, i.e., L-Val-L-Tyr and Gly-Gly in 14-OMO (21a22a, Scheme 3) were synthesized [36]. The reductive amination of 14-OMO (1) was performed using amino acid tert-butyl ester hydrochlorides or dipeptide benzyl ester hydrochlorides, and NaBH3CN in CH3OH. Medium-pressure liquid chromatography (MPLC) was used to separate the diastereoisomers, providing ester derivatives 5aa22aa (Scheme 2 and Scheme 3). Typically, the ratio of 6β-amino to 6α amino epimers was between 4:1 and 2:1. The 6-amino-acid (5a20b)-substituted derivatives were obtained through ester cleavage of the tert-butyl derivatives in dioxane/HCl (Scheme 2). Catalytic hydrogenation of the benzyl esters 21aa/bb and 22aa in CH3OH using 10% Pd/C catalyst provided the 6-dipeptide-substituted 21a/b and 22a, respectively (Scheme 3) [36].
Scheme 2. Synthesis of different natural- and unnatural-6-amino-acid-substituted N-methyl-14-methoxymorphinans (5a20b). 14-OMO (1.HBr) was used as starting material. The amino acid tert-butyl ester derivatives 5aa20bb were prepared from 1 by reductive amination with the respective tert-butyl ester hydrochloride. Esters were hydrolyzed to yield the corresponding 6-amino-acid-substituted derivatives 5a20b. Ph: Phenyl.
Scheme 3. Synthesis of 6-dipeptide-substituted N-methyl-14-methoxymorphinans (21a/b and 22b). 14-OMO (1.HBr) was used as starting material. Dipeptide benzyl ester derivatives (21aa/bb and 22aa/bb) were prepared from 1 by reductive amination with the respective benzyl ester hydrochloride. Esters were hydrolyzed to yield the corresponding 6-dipeptide-substituted derivatives 21a22b. Ph: Phenyl.
Synthetic work also targeted the combination of 6-amino amino (i.e., Gly) and 14-arylalkoxy (e.g., phenylpropoxy) substitutions in N-methyl-morphinans (Scheme 4) [37]. The reductive amination of the 14-phenylpropoxyoxymorphone (POMO, 23) was performed with Gly tert-butyl ester hydrochloride and NaCNBH3 in DMF/MeOH at room temperature. The diastereoisomers were separated by column chromatography to obtain tert-butyl esters 24aa and 24bb. Ester cleavage in dioxane/HCl generated the amino acids 24a and 24b (Scheme 4) [37].
Scheme 4. Synthesis of 6-Gly-substituted derivatives of N-methyl-14-O-phenylpropylmorphinan-6-one (24a and 24b). POMO (23) was used as starting material. The amino acid tert-butyl ester derivatives 24aa and 24bb were prepared from 23 by reductive amination with the tert-butyl ester hydrochloride. Esters were hydrolyzed to yield the corresponding 6-Gly-substituted derivatives 24a and 24b.
SAR studies on the series of amino acid and dipeptide substitution at position 6 in 14-OMO (1) as zwitterionic molecules explored their binding and activation of the opioid receptors and antinociceptive properties (Table 2, Table 3 and Table 4). The 6-amino acid groups included natural amino acids (i.e., Gly, Ala, Phe, Ser, Val, Lys, Tyr, Trp, Asn, Gln, Asp and Glu), unnatural amino acids (i.e., D-Ala, D-Val, D-Phe, L-Chg, L-Abu, β-Ala and GABA) (2a20b) (Scheme 1, Scheme 2 and Scheme 4) [35][36][37][38], and 6-dipeptide substitution (i.e., L-Val-L-Tyr and Gly-Gly) (22a/b and 23a) (Scheme 3) [36]. In vitro receptor binding (radioligand binding assays with membranes from rodent brain and CHO cells expressing the human opioid receptors) and functional assays (mouse vas deferens (MVD) bioassay and [35S]GTPγS binding assay with membranes from CHO cells expressing the human opioid receptors) established the potent MOR/DOR agonist profile and reduced the binding and activation of the KOR for most compounds (Table 2). The replacement of the 14-methoxy group in N-methyl, 6-Gly substituted morphinans 2a (HS-730) and 2b (HS-731) with a 14-phenylpropoxy group (compounds 24a and 24b, respectively) resulted in a considerable increase in binding affinities to all three opioid receptor types, MOR, DOR and KOR, in rodent brain membranes (Table 2 and Table 3) [37]. Compared to the nonselective 14-phenylpropoxy-substituted POMO (23) [39], the 6-Gly analogues 24a and 24b showed a comparable binding profile to the opioid receptors, acting as mixed MOR/DOR/KOR ligands (Table 2) [37].
Table 2. In vitro activities to the opioid receptors of 6-amino-acid (2a20b)- and 6-dipeptide (22a/b and 23a)-substituted derivatives of 14-OMO (1).
24a/b)-substituted derivatives of POMO (23).

References

  1. Corder, G.; Castro, D.C.; Bruchas, M.R.; Scherrer, G. Endogenous and exogenous opioids in pain. Annu. Rev. Neurosci. 2018, 41, 453–473.
  2. Paul, A.K.; Smith, C.M.; Rahmatullah, M.; Nissapatorn, V.; Wilairatana, P.; Spetea, M.; Gueven, N.; Dietis, N. Opioid analgesia and opioid-induced adverse effects: A review. Pharmaceuticals 2021, 14, 1091.
  3. Volkow, N.; Benveniste, H.; McLellan, T.A. Use and misuse of opioids in chronic pain. Annu. Rev. Med. 2018, 69, 451–465.
  4. Sobczak, Ł.; Goryński, K. Pharmacological aspects of over-the-counter opioid drugs misuse. Molecules 2020, 25, 3905.
  5. National Institute on Drug Abuse (NIDA). Available online: https://nida.nih.gov/research-topics/trends-statistics/overdose-death-rates (accessed on 28 April 2023).
  6. Spetea, M. Opioid receptors and their ligands in the musculoskeletal system and relevance for pain control. Curr. Pharm. Des. 2013, 19, 7382–7390.
  7. Sein, C. Opioid receptors. Annu. Rev. Med. 2016, 67, 433–451.
  8. Che, T.; Roth, B.L. Structural insights accelerate the discovery of opioid alternatives. Annu. Rev. Biochem. 2021, 90, 739–761.
  9. Spetea, M.; Asim, M.F.; Wolber, G.; Schmidhammer, H. The opioid receptor and ligands acting at the µ opioid receptor, as therapeutics and potential therapeutics. Curr. Pharm. Des. 2013, 19, 7415–7434.
  10. Busserolles, J.; Lolignier, S.; Kerckhove, N.; Bertin, C.; Authier, N.; Eschalier, A. Replacement of current opioid drugs focusing on MOR-related strategies. Pharmacol. Ther. 2020, 210, 107519.
  11. Darcq, E.; Kieffer, B.L. Opioid receptors: Drivers to addiction? Nat. Rev. Neurosci. 2018, 19, 499–514.
  12. Imam, M.Z.; Kuo, A.; Ghassabian, S.; Smith, M.T. Progress in understanding mechanisms of opioid-induced gastrointestinal adverse effects and respiratory depression. Neuropharmacology 2018, 131, 238–255.
  13. Fürst, S.; Hosztafi, S. The chemical and pharmacological importance of morphine analogues. Acta Physiol. Hung. 2008, 95, 3–44.
  14. Pasternak, G.W.; Pan, Y.X. Mu opioids and their receptors: Evolution of a concept. Pharmacol. Rev. 2013, 65, 1257–1317.
  15. Spetea, M.; Schmidhammer, H. Recent advances in the development of 14-alkoxy substituted morphinans as potent and safer opioid analgesics. Curr. Med. Chem. 2012, 19, 2442–2457.
  16. Lewis, J.W.; Husbands, S.M. 14-Amino-4,5-epoxymorphinan derivatives and their pharmacological actions. Top. Curr. Chem. 2011, 299, 93–119.
  17. Stavitskaya, L.; Coop, A. Most recent developments and modifications of 14-alkylamino and 14-alkoxy-4,5-epoxymorphinan derivatives. Mini. Rev. Med. Chem. 2011, 11, 1002–1008.
  18. Schmidhammer, H.; Spetea, M.; Windisch, P.; Schütz, J.; Riba, P.; Al-Khrasani, M.; Fürst, S. Functionalization of the carbonyl group in position 6 of morphinan-6-ones. Development of novel 6-amino and 6-guanidino substituted 14-alkoxymorphinans. Curr. Pharm. Des. 2013, 19, 7391–7399.
  19. Devereaux, A.L.; Mercer, S.L.; Cunningham, C.W. DARK classics in chemical neuroscience: Morphine. ACS Chem. Neurosci. 2018, 9, 2395–2407.
  20. Spetea, M.; Schmidhammer, H. Recent chemical and pharmacological developments on 14-oxygenated-N-methylmorphinan-6-ones. Molecules 2021, 26, 5677.
  21. Kalso, E.; Smith, L.; McQuay, H.J.; Moore, R.A. No pain, no gain: Clinical excellence and scientific rigour—Lessons learned from IA morphine. Pain 2002, 98, 269–275.
  22. Stein, C.; Schäfer, M.; Machelska, H. Attaching pain at its source: New perspectives on opioids. Nat. Med. 2003, 9, 1003–1008.
  23. Kapitzke, D.; Vetter, I.; Cabot, P.J. Endogenous opioid analgesia in peripheral tissues and the clinical implications for pain control. Ther. Clin. Risk Manag. 2005, 1, 279–297.
  24. Martinez, V.; Abalo, R. Peripherally acting opioid analgesics and peripherally-induced analgesia. Behav. Pharmacol. 2020, 31, 136–158.
  25. Stein, C.; Comisel, K.; Haimaeri, E.; Yassouridis, A.; Lehrberger, K. Analgesic effects of intraarticular morphine after arthroscopic knee surgery. N. Engl. J. Med. 1991, 325, 1123–1126.
  26. Likar, R.; Kapral, S.; Steinkellner, H.; Stein, C.; Schäfer, M. Dose-dependency of intra-articular morphine analgesia. Br. J. Anaesth. 1999, 83, 241–244.
  27. Iorio, M.A.; Frigeni, V. Narcotic agonist/antagonist properties of quaternary diastereoisomers derived from oxymorphone and naloxone. Eur. J. Med. Chem. 1984, 19, 301–303.
  28. Brown, D.R.; Goldberg, L.I. The use of quaternary narcotic antagonists in opiate research. Neuropharmacology 1985, 24, 181–191.
  29. Foster, R.S.; Jenden, D.J.; Lomax, P. A comparison of the pharmacologic effects of morphine and N-methyl morphine. J. Pharmacol. Exp. Ther. 1967, 157, 185–195.
  30. Smith, T.W.; Buchan, P.; Parsons, D.N.; Wilkinson, S. Peripheral antinociceptive effects of N-methyl morphine. Life Sci. 1982, 31, 1205–1208.
  31. Oluyomi, A.O.; Hart, S.L.; Smith, T.W. Differential antinociceptive effects of morphine and methylmorphine in the formalin test. Pain 1992, 49, 415–418.
  32. Kosterlitz, H.W.; Waterfield, A.A. In vitro models in the study of structure-activity relationships of narcotic analgesics. Annu. Rev. Pharmacol. 1975, 15, 29–47.
  33. Botros, S.; Lipkowski, A.; Larson, D.; Stark, P.; Takemori, A.; Portoghese, P.S. Opioid agonist and antagonist activities of peripherally selective derivatives of naltrexamine and oxymorphamine. J. Med. Chem. 1989, 32, 2068–2071.
  34. Larson, D.L.; Hua, M.; Takemori, A.K.; Portoghese, P.S. Possible contribution of a glutathione conjugate to the long-duration action of funaltrexamine. J. Med. Chem. 1993, 36, 3669–3673.
  35. Schütz, J.; Brandt, W.; Spetea, M.; Wurst, K.; Wunder, G.; Schmidhammer, H. Synthesis of 6-amino acid substituted derivatives of the highly potent analgesic 14-O-methyloxymorphone. Helv. Chim. Acta 2003, 86, 2142–2148.
  36. Spetea, M.; Rief, S.B.; Haddou, T.B.; Fink, M.; Kristeva, E.; Mittendorfer, H.; Haas, S.; Hummer, N.; Follia, V.; Guerrieri, E.; et al. Synthesis, biological, and structural explorations of new zwitterionic derivatives of 14-O-methyloxymorphone, as potent μ/δ opioid agonists and peripherally selective antinociceptives. J. Med. Chem. 2019, 62, 641–653.
  37. Spetea, M.; Windisch, P.; Guo, Y.; Bileviciute-Ljungar, I.; Schütz, J.; Asim, M.F.; Berzetei-Gurske, I.P.; Riba, P.; Kiraly, K.; Fürst, S.; et al. Synthesis and pharmacological activities of 6-glycine substituted 14-phenylpropoxymorphinans, a novel class of opioids with high opioid receptor affinities and antinociceptive potencies. J. Med. Chem. 2011, 54, 980–988.
  38. Spetea, M.; Friedmann, T.; Riba, P.; Schütz, J.; Wunder, G.; Langer, T.; Schmidhammer, H.; Fürst, S. In vitro opioid activity profiles of 6-amino acid substituted derivatives of 14-O-methyloxymorphone. Eur. J. Pharmacol. 2004, 483, 301–308.
  39. Lattanzi, R.; Rief, S.; Schmidhammer, H.; Negri, L.; Spetea, M. In vitro and in vivo pharmacological activities of 14-O-phenylpropyloxymorphone, a potent mixed mu/delta/kappa-opioid receptor agonist with reduced constipation in mice. Front. Pharmacol. 2018, 9, 1002.
  40. Fürst, S.; Riba, P.; Friedmann, T.; Tímar, J.; Al-Khrasani, M.; Obara, I.; Makuch, W.; Spetea, M.; Schütz, J.; Przewlocki, R.; et al. Peripheral versus central antinociceptive actions of 6-amino acid-substituted derivatives of 14-O-methyloxymorphone in acute and inflammatory pain in the rat. J. Pharmacol. Exp. Ther. 2005, 312, 609–618.
  41. Bileviciute-Ljungar, I.; Spetea, M.; Guo, Y.; Schütz, J.; Windisch, P.; Schmidhammer, H. Peripherally mediated antinociception of the mu-opioid receptor agonist 2-(4,5alpha-epoxy-3-hydroxy-14beta-methoxy-17-methylmorphinan-6beta-yl)aminoacetic acid (HS-731) after subcutaneous and oral administration in rats with carrageenan-induced hindpaw inflammation. J. Pharmacol. Exp. Ther. 2006, 317, 220–227.
  42. Al-Khrasani, M.; Spetea, M.; Friedmann, T.; Riba, P.; Király, K.; Schmidhammer, H.; Fürst, S. DAMGO and 6β-glycine substituted 14-O-methyloxymorphone but not morphine show peripheral, preemptive antinociception after systemic administration in a mouse visceral pain model and high intrinsic efficacy in the isolated rat vas deferens. Brain Res. Bull. 2007, 74, 369–375.
  43. Baillie, L.D.; Schmidhammer, H.; Mulligan, S.J. Peripheral μ-opioid receptor mediated inhibition of calcium signaling and action potential-evoked calcium fluorescent transients in primary afferent CGRP nociceptive terminals. Neuropharmacology 2015, 93, 267–273.
  44. Obara, I.; Makuch, W.; Spetea, M.; Schütz, J.; Schmidhammer, H.; Przewlocki, R.; Przewlocka, B. Local peripheral antinociceptive effects of 14-O-methyloxymorphone derivatives in inflammatory and neuropathic pain in the rat. Eur. J. Pharmacol. 2007, 558, 60–67.
  45. Puls, K.; Schmidhammer, H.; Wolber, G.; Spetea, M. Mechanistic characterization of the pharmacological profile of HS-731, a peripherally acting opioid analgesic, at the µ-, δ-, κ-opioid and nociceptin receptors. Molecules 2022, 27, 919.
  46. Fürst, S.; Zádori, Z.S.; Zádor, F.; Király, K.; Balogh, M.; László, S.B.; Hutka, B.; Mohammadzadeh, A.; Calabrese, C.; Galambos, A.R.; et al. On the role of peripheral sensory and gut mu opioid receptors: Peripheral analgesia and tolerance. Molecules 2020, 25, 2473.
  47. Herz, A.; Teschemacher, H.J. Activities and sites of antinociceptive action of morphine-like analgesics and kinetics of distribution following intravenous, intracerebral and intraventricular application. In Advances in Drug Research; Harper, N., Simmonds, A.B., Eds.; Academic Press Limited: London, UK, 1971; Volume 6, pp. 79–118.
  48. Frances, B.; Gout, R.; Monsarrat, B.; Cros, J.; Zajac, J.M. Further evidence that morphine-6β-glucuronide is a more potent opioid agonist than morphine. J. Pharmacol. Exp. Ther. 1992, 262, 2531.
  49. Tukey, R.H.; Strassburg, C.P. Human UDP-glucuronosyltransferases: Metabolism, expression, and disease. Annu. Rev. Pharmacol. Toxicol. 2000, 40, 581–616.
  50. Mori, M.; Oguri, K.; Yoshimura, H.; Shimomura, K.; Kamata, O.; Ueki, S. Chemical synthesis and analgesic effect of morphine ethereal sulfates. Life Sci. 1972, 11, 525–533.
  51. Váradi, A.; Gergely, A.; Béni, S.; Jankovics, P.; Noszál, B.; Hosztafi, S. Sulfate esters of morphine derivatives: Synthesis and characterization. Eur. J. Pharm. Sci. 2011, 42, 65–72.
  52. Zuckerman, A.; Bolan, E.; de Paulis, T.; Schmidt, D.; Spector, S.; Pasternak, G.W. Pharmacological characterization of morphine-6-sulfate and codeine-6-sulfate. Brain Res. 1999, 842, 1–5.
  53. Zádor, F.; Mohammadzadeh, A.; Balogh, M.; Zádori, Z.S.; Király, K.; Barsi, S.; Galambos, A.R.; László, S.B.; Hutka, B.; Váradi, A.; et al. Comparisons of in vivo and in vitro opioid effects of newly synthesized 14-methoxycodeine-6-O-sulfate and codeine-6-O-sulfate. Molecules 2020, 25, 1370.
  54. Preechagoon, D.; Brereton, I.; Staatz, C.; Prankerd, R. Ester prodrugs of a potent analgesic, morphine-6-sulfate: Syntheses, spectroscopic and physicochemical properties. Int. J. Pharm. 1998, 163, 177–190.
  55. Crooks, P.A.; Kottayil, S.G.; Al-Ghananeem, A.M.; Byrn, S.R.; Butterfield, A.D. Opiate receptor binding properties of morphine-, dihydromorphine-, and codeine 6-O-sulfate ester congeners. Bioorg. Med. Chem. Lett. 2006, 16, 4291–4295.
  56. Kaspersen, F.M.; Van Boeckel, C.A.A. A review of the methods of chemical synthesis of sulphate and glucuronide conjugates. Xenobiotica 1987, 17, 1451–1471.
  57. Al-Horani, R.A.; Desai, U.R. Chemical sulfation of small molecules—Advances and challenges. Tetrahedron 2010, 66, 2907–2918.
  58. Lacko, E.; Varadi, A.; Rapavi, R.; Zador, F.; Riba, P.; Benyhe, S.; Borsodi, A.; Hosztafi, S.; Timar, J.; Noszal, B.; et al. A novel µ-opioid receptor ligand with high in vitro and in vivo agonist efficacy. Curr. Med. Chem. 2012, 54, 4699–4707.
  59. Brown, C.E.; Roerig, S.C.; Burger, V.T. Analgesic potencies of morphine 3- and 6-sulfates, after intracerebroventricular administration in mice: Relationship to structural characteristics defined by mass spectrometry and nuclear magnetic resonance. J. Pharm. Sci. 1985, 74, 821–824.
  60. Holtman, J.R.; Crooks, P.A.; Johnson-Hardy, J.; Wala, E.P. Antinociceptive effects and toxicity of morphine-6-O-sulfate sodium salt in rat models of pain. Eur. J. Pharmacol. 2010, 648, 87–94.
  61. Al-Khrasani, M.; Lackó, E.; Riba, P.; Király, K.; Sobor, M.; Timár, J.; Mousa, S.; Schäfer, M.; Fürst, S. The central versus peripheral antinociceptive effects of µ-opioid receptor agonists in the new model of rat visceral pain. Brain Res. Bull. 2012, 87, 238–243.
  62. Lackó, E.; Riba, P.; Giricz, Z.; Váradi, A.; Cornic, L.; Balogh, M.; Király, K.; Cseko, K.; Mousa, S.A.; Hosztafi, S.; et al. New morphine analogs produce peripheral antinociception within a certain dose range of their systemic administration. J. Pharmacol. Exp. Ther. 2016, 359, 171–181.
  63. Yadlapalli, J.S.K.; Ford, B.M.; Ketkar, A.; Wan, A.; Penthala, N.R.; Eoff, R.L.; Prather, P.L.; Dobretsov, M.; Crooks, P.A. Antinociceptive effects of the 6-O-sulfate ester of morphine in normal and diabetic rats: Comparative role of mu- and delta-opioid receptors. Pharmacol. Res. 2016, 13, 335–347.
  64. Yadlapalli, J.S.K.; Jai Shankar, K.; Dogra, N.; Walbaum, A.W.; Wessinger, W.D.; Prather, P.L.; Crooks, P.A.; Dobretsov, M. Evaluation of analgesia, tolerance, and the mechanism of action of morphine-6-O-sulfate across multiple pain modalities in Sprague-Dawley Rats. Anesth. Analg. 2017, 125, 1021–1031.
  65. Balogh, M.; Zádori, Z.S.; Lázár, B.; Karádi, D.; László, S.; Mousa, S.A.; Hosztafi, S.; Zádor, F.; Riba, P.; Schäfer, M.; et al. The peripheral versus central antinociception of a novel opioid agonist: Acute inflammatory pain in rats. Neurochem. Res. 2018, 43, 1250–1257.
  66. Balogh, M.; Zádor, F.; Zádori, Z.S.; Shaqura, M.; Király, K.; Mohammadzadeh, A.; Varga, B.; Lázár, B.; Mousa, S.A.; Hosztafi, S.; et al. Efficacy-based perspective to overcome reduced opioid analgesia of advanced painful diabetic neuropathy in rats. Front. Pharmacol. 2019, 10, 347.
  67. Khalefa, B.I.; Mousa, S.A.; Shaqura, M.; Lackó, E.; Hosztafi, S.; Riba, P.; Schäfer, M.; Ferdinandy, P.; Fürst, S.; Al-Khrasani, M. Peripheral antinociceptive efficacy and potency of a novel opioid compound 14-O-MeM6SU in comparison to known peptide and non-peptide opioid agonists in a rat model of inflammatory pain. Eur.J. Pharmacol. 2013, 713, 54–57.
  68. Rosow, C.E. Anesthetic drug interaction: An overview. J. Clin. Anesthn. 1997, 9, 27S–32S.
  69. Yadlapalli, J.S.K.; Albayati, Z.A.F.; Penthala, N.R.; Hendrickson, H.P.; Crooks, H.A. Stability studies of potent opioid analgesic, morphine-6-O-sulfate in various buffers and biological matrices by HPLC-DAD analysis. Biomed. Chromatogr. 2017, 31, e3957.
  70. Kiraly, K.; Caputi, F.F.; Hanuska, A.; Kató, E.; Balogh, M.; Köles, L.; Palmisano, M.; Riba, P.; Hosztafi, S.; Romualdi, P.; et al. A new potent analgesic agent with reduced liability to produce morphine tolerance. Brain Res. Bull. 2015, 117, 32–38.
  71. Paul, D.; Standifer, K.M.; Inturrisi, C.E.; Pasternak, G.W. Pharmacological characterization of morphine-6 beta-glucuronide, a very potent morphine metabolite. J. Pharmacol. Exp. Ther. 1989, 251, 477–483.
  72. Osborne, R.; Joel, S.; Trew, D.; Slevin, M. Morphine and metabolite behavior after different routes of morphine administration: Demonstration of the importance of the active metabolite morphine-6-glucuronide. Clin. Pharmacol. Ther. 1990, 47, 12–19.
  73. Tegeder, I.; Meier, S.; Burian, M.; Schmidt, H.; Geisslinger, G.; Lötsch, J. Peripheral opioid analgesia in experimental human pain models. Brain 2003, 126, 1092–1102.
  74. Hanna, M.H.; Elliott, K.M.; Fung, M. Randomized, double-blind study of the analgesic efficacy of morphine-6-glucuronide versus morphine sulfate for postoperative pain in major surgery. Anesthesiology 2005, 102, 815–821.
  75. Binning, A.R.; Przesmycki, K.; Sowinski, P.; Morrison, L.M.M.; Smith, T.W.; Marcus, P.; Lees, J.P.; Dahan, A. A randomised controlled trial on the efficacy and side-effect profile (nausea/vomiting/sedation) of morphine-6-glucuronide versus morphine for post-operative pain relief after major abdominal surgery. Eur. J. Pain 2011, 15, 402–408.
  76. Sverrisdóttir, E.; Lund, T.M.; Olesen, A.E.; Drewes, A.M.; Christrup, L.L.; Kreilgaard, M. A review of morphine and morphine-6-glucuronide’s pharmacokinetic-pharmacodynamic relationships in experimental and clinical pain. Eur. J. Pharm. Sci. 2015, 74, 45–62.
  77. Kilpatrick, G.J.; Smith, T.W. Morphine-6-glucuronide: Actions and mechanisms. Med. Res. Rev. 2005, 25, 521–544.
  78. Lötsch, J.; Geisslinger, G. Morphine-6-glucuronide: An analgesic of the future? Clin. Pharmacokinet. 2001, 40, 485–499.
  79. Bickel, U.; Schumacher, O.P.; Kang, Y.S.; Voigt, K. Poor permeability of morphine 3-glucuronide and morphine 6-glucuronide through the blood barrier in the rat. J. Pharmacol. Exp. Ther. 1996, 278, 107–113.
  80. Huwyler, J.; Drewe, J.; Klusemann, C.; Fricker, G. Evidence for P-glycoprotein-modulated penetration of morphine-6-glucuronide into brain capillary endothelium. Br. J. Pharmacol. 1996, 118, 1879–1885.
  81. Sattari, M.; Routledge, P.; Mashayekhi, S. The influence of active transport systems on morphine -6-glucuronide transport in MDCKII and MDCK-PGP cells. Daru 2011, 19, 412–416.
  82. Bourasset, F.; Cisternino, S.; Temsamani, J.; Scherrmann, J.-M. 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, 1564–1567.
  83. Koning, G.A.; Schiffelers, R.M.; Storm, G. Endothelial cells at inflammatory sites as target for therapeutic intervention. Endothelium 2002, 9, 161–171.
  84. Fleige, E.; Quadir, M.A.; Haag, R. Stimuli-responsive polymeric nanocarriers for the controlled transport of active compounds: Concepts and applications. Adv. Drug Deliv. Rev. 2012, 64, 866–884.
  85. Kraft, J.C.; Freeling, J.P.; Wang, Z.; Ho, R.J. Emerging research and clinical development trends of liposome and lipid nanoparticle drug delivery systems. J. Pharm. Sci. 2014, 103, 29–52.
  86. Nehoff, H.; Parayath, N.N.; Domanovitch, L.; Taurin, S.; Greish, K. Nanomedicine for drug targeting: Strategies beyond the enhanced permeability and retention effect. Int. J. Nanomed. 2014, 22, 2539–2555.
  87. Gurunathan, S.; Kang, M.H.; Qasim, M.; Kim, J.H. Nanoparticle-mediated combination therapy: Two-in-one approach for cancer. Int. J. Mol. Sci. 2018, 19, 3264.
  88. Moradkhani, M.R.; Karimi, A.; Negahdari, B. Nanotechnology application for pain therapy. Artif. Cells Nanomed. Biotechnol. 2018, 46, 368–373.
  89. Chakravarthy, K.V.; Boehm, F.J.; Christo, P.J. Nanotechnology: A promising new paradigm for the control of pain. Pain Med. 2018, 19, 232–243.
  90. González-Rodríguez, S.; Quadir, M.A.; Gupta, S.; Walker, K.A.; Zhang, X.; Spahn, V. Polyglycerol-opioid conjugate produces analgesia devoid of side effects. Elife 2017, 6, e27081.
  91. Martin, C.; Oyen, E.; Mangelschots, J.; Bibian, M.; Ben Haddou, T.; Andrade, J.; Gardiner, J.; Van Mele, B.; Madder, A.; Hoogenboom, R.; et al. Injectable peptide hydrogels for controlled-release of opioids. MedChemComm 2016, 7, 542–549.
  92. Martin, C.; Oyen, E.; Van Wanseele, Y.; Haddou, T.B.; Schmidhammer, H.; Andrade, J.; Waddington, L.; Van Eeckhaut, A.; Van Mele, B.; Gardiner, J.; et al. Injectable peptide-based hydrogel formulations for the extended in vivo release of opioids. Mater. Today Chem. 2017, 3, 49–59.
More
Video Production Service