Skip to main content
The AAPS Journal logoLink to The AAPS Journal
. 2005 Oct 14;7(3):E560–E565. doi: 10.1208/aapsj070356

Opioid peptide-derived analgesics

Peter W Schiller 1,
PMCID: PMC2751258  PMID: 16353933

Abstract

Two recent developments of opioid peptide-based analgesics are reviewed. The first part of the review discusses the dermorphin-derived, cationic-aromatic tetrapeptide H-Dmt-D-Arg-Phe-Lys-NH2 ([Dmt1]DALDA, where Dmt indicates 2′,6′-dimethyltyrosine), which showed subnanomolar μ receptor binding affinity, extraordinary μ receptor selectivity, and high μ agonist potency in vitro. In vivo, [Dmt1]DALDA looked promising as a spinal analgesic because of its extraordinary antinociceptive effect (3000 times more potent than morphine) in the mouse tail-flick assay, long duration of action (4 times longer than morphine), and lack of effect on respiration. Unexpectedly, [Dmt1]DALDA also turned out to be a potent and long-acting analgesic in the tail-flick test when given subcutaneously (s.c.), indicating that it is capable of crossing the blood-brain barrier. Furthermore, little or no cross-tolerance was observed with s.c. [Dmt1]DALDA in morphine-tolerant mice. The second part of the review concerns the development of mixed μ agonist/δ antagonists that, on the basis of much evidence, are expected to be analgesics with a low propensity to produce tolerance and physical dependence. The prototype pseudopeptide H-Dmt-TicΨ[CH2NH]Phe-Phe-NH2 (DIPP-NH2[Ψ], where Tic indicates 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid) showed subnanomolar μ and δ receptor binding affinities and the desired μ agonist/δ antagonist profile in vitro. DIPP-NH2[Ψ] produced a potent analgesic effect after intracerebroventricular administration in the rat tail-flick assay, no physical dependence, and less tolerance than morphine. The results obtained with DIPP-NH2[Ψ] indicate that mixed μ agonist/δ antagonists look promising as analgesic drug candidates, but compounds with this profile that are systemically active still need to be developed.

Full Text

The Full Text of this article is available as a PDF (197.1 KB).

References

  • 1.Pfeiffer A, Brantl V, Herz A, Emrich HM. Psychotomimesis mediated by kappa opiate receptors. Science. 1986;233:774–776. doi: 10.1126/science.3016896. [DOI] [PubMed] [Google Scholar]
  • 2.Walsh SL, Strain EC, Abreu ME, Bigelow GE. Enadoline, a selective kappa opioid agonist: comparison with butorphanol and hydromorphone in humans. Psychopharmacology (Berl) 2001;157:151–162. doi: 10.1007/s002130100788. [DOI] [PubMed] [Google Scholar]
  • 3.Yaksh TL. In vivo studies on spinal opiate receptor systems mediating antinociception, I: mu and delta receptor profiles in the primate. J Pharmacol Exp Ther. 1983;226:303–316. [PubMed] [Google Scholar]
  • 4.Porreca F, Mosberg HI, Hurst R, Hruby VJ, Burks TF. Roles of mu, delta and kappa opioid receptors in spinal and supraspinal mediation of gastrointestinal transit effects and hot-plate analgesia in the mouse. J Pharmacol Exp Ther. 1984;230:341–348. [PubMed] [Google Scholar]
  • 5.Cowan A, Zhu XZ, Mosberg HI, Omnaas JR, Porreca F. Direct dependence studies in rats with agents selective for different types of opioid receptor. J Pharmacol Exp Ther. 1988;246:950–955. [PubMed] [Google Scholar]
  • 6.Cheng PY, Wu D, Decena J, Soong Y, McCabe S, Szeto HH. Opioid-induced stimulation of fetal respiratory activity by [D-Ala2]deltorphin, I. Eur J Pharmacol. 1993;230:85–88. doi: 10.1016/0014-2999(93)90413-C. [DOI] [PubMed] [Google Scholar]
  • 7.Galligan JJ, Mosberg HI, Hurst R, Hruby VJ, Burks TF. Cerebral delta opioid receptors mediate analgesia but not the intestinal motility effects of intracerebroventricularly administered opioids. J Pharmacol Exp Ther. 1984;229:641–648. [PubMed] [Google Scholar]
  • 8.Weber SJ, Greene DL, Sharma SD, et al. Distribution and analgesia of [3H][D-Pen2, D-Pen5]enkephalin and two halogenated analogs after intravenous administration. J Pharmacol Exp Ther. 1991;259:1109–1117. [PubMed] [Google Scholar]
  • 9.Svensson CI, Rew Y, Malkmus S, et al. Systemic and spinal analgesic activity of a δ-opioid-selective lanthionine enkephalin analog. J Pharmacol Exp Ther. 2003;304:827–832. doi: 10.1124/jpet.102.039750. [DOI] [PubMed] [Google Scholar]
  • 10.Kamei J, Saitoh A, Ohsawa M, et al. Antinociceptive effects of the selective non-peptidic δ-opioid receptor antagonist TAN-67 in diabetic mice. Eur J Pharmacol. 1995;276:131–135. doi: 10.1016/0014-2999(95)00026-H. [DOI] [PubMed] [Google Scholar]
  • 11.Chang K-J, Rigdon GC, Howard JL, McNutt RW. A novel, potent and selective nonpeptidic delta opioid receptor agonist BW373U86. J Pharmacol Exp Ther. 1993;267:852–857. [PubMed] [Google Scholar]
  • 12.Calderon SN, Rothman RB, Porreca F, et al. Probes for narcotic receptor mediated phenomena, 19: synthesis of (+)-4-(αR)-α-((2S, 5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80): a highly selective, nonpeptide delta opioid receptor agonist. J Med Chem. 1994;37:2125–2128. doi: 10.1021/jm00040a002. [DOI] [PubMed] [Google Scholar]
  • 13.Plobeck N, Delorme D, Wei Z-Y, et al. New diarylmethylpiperazines as potent and selective nonpeptidic δ opioid receptor agonists with increased in vitro metabolic stability. J Med Chem. 2000;43:3878–3894. doi: 10.1021/jm000228x. [DOI] [PubMed] [Google Scholar]
  • 14.DeHaven-Hudkins DL, Dolle RE. Peripherally restricted opioid agonists as novel analgesic agents. Curr Pharm Des. 2004;10:743–757. doi: 10.2174/1381612043453036. [DOI] [PubMed] [Google Scholar]
  • 15.Schiller PW, Nguyen TM-D, Chung NN, Lemieux C. Dermorphin analogues carrying an increased positive net charge in their “message” domain display extremely high μ-opioid receptor selectivity. J Med Chem. 1989;32:698–703. doi: 10.1021/jm00123a035. [DOI] [PubMed] [Google Scholar]
  • 16.Schiller PW, Nguyen TM-D, Berezowska I, et al. Synthesis and in vitro opioid activity profiles of DALDA analogues. Eur J Med Chem. 2000;35:895–901. doi: 10.1016/S0223-5234(00)01171-5. [DOI] [PubMed] [Google Scholar]
  • 17.Neilan CL, Nguyen TM-D, Schiller PW, Pasternak GW. Pharmacological characterization of the dermorphim analog [Dmt1]DALDA, a highly potent and selective μ-opioid peptide. Eur J Pharmacol. 2001;419:15–23. doi: 10.1016/S0014-2999(01)00946-3. [DOI] [PubMed] [Google Scholar]
  • 18.Neilan CL, Janvey AJ, Bolan E, et al. Characterization of the binding of [3H][Dmt1]H-Dmt-D-Arg-Phe-Lys-NH2, a highly potent opioid peptide. J Pharmacol Exp Ther. 2003;306:430–436. doi: 10.1124/jpet.103.049742. [DOI] [PubMed] [Google Scholar]
  • 19.Szeto HH, Lovelace JL, Fridland G, et al. In vivo pharmacokinetics of selective μ-opioid peptide agonists. J Pharmacol Exp Ther. 2001;298:57–61. [PubMed] [Google Scholar]
  • 20.Shimoyama M, Shimoyama N, Zhao G-M, Schiller PW, Szeto HH. Antinociceptive and respiratory effects of intrathecal H-Tyr-D-Arg-Phe-Lys-NH2 (DALDA) and [Dmt1]DALDA. J Pharmacol Exp Ther. 2001;297:364–371. [PubMed] [Google Scholar]
  • 21.Reimann W, Schlutz H, Selve N. The antinociceptive effects of morphine, desipramine, and serotonin and their combinations after intrathecal injection in the rat. Anesth Analg. 1999;88:141–145. doi: 10.1097/00000539-199901000-00026. [DOI] [PubMed] [Google Scholar]
  • 22.Szeto HH, Soong Y, Wu D, Qian X, Zhao G-M. Endogenous opioid peptides contribute to antinociceptive potency of intrathecal [Dmt1]DALDA. J Pharmacol Exp Ther. 2003;305:696–702. doi: 10.1124/jpet.102.048561. [DOI] [PubMed] [Google Scholar]
  • 23.Zhao GM, Wu D, Soong Y, et al. Profound spinal tolerance after repeated exposure to a highly selective μ-opioid peptide agonist: role of δ-opioid receptors. J Pharmacol Exp Ther. 2002;302:188–196. doi: 10.1124/jpet.302.1.188. [DOI] [PubMed] [Google Scholar]
  • 24.Ben Y, Smith AP, Schiller PW, Lee NM. Tolerance develops in spinal cord, but not in brain with chronic [Dmt1]DALDA treatment. Br J Pharmacol. 2004;143:987–993. doi: 10.1038/sj.bjp.0706007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Neilan CL, King MA, Rossi G, et al. Differential sensitivities of mouse strains to morphine and [Dmt1]DALDA analgesia. Brain Res. 2003;974:254–257. doi: 10.1016/S0006-8993(03)02590-3. [DOI] [PubMed] [Google Scholar]
  • 26.Riba P, Ben Y, Nguyen TM-D, Furst S, Schiller PW, Lee NM. [Dmt1]DALDA is highly selective and potent at μ opioid receptors, but is not cross-tolerant with systemic morphine. Curr Med Chem. 2002;9:31–39. doi: 10.2174/0929867023371445. [DOI] [PubMed] [Google Scholar]
  • 27.Zhao K, Luo G, Zhao G-M, Schiller PW, Szeto HH. Transcellular transport of a highly polar 3+net charge opioid tetrapeptide. J Pharmacol Exp Ther. 2003;304:425–432. doi: 10.1124/jpet.102.040147. [DOI] [PubMed] [Google Scholar]
  • 28.Berezowska I, Chung NN, Lemieux C, Zelent B, Szeto HH, Schiller PW. Highly potent fluorescent analogues of the opioid peptide [Dmt1]DALDA. Peptides. 2003;24:1195–1200. doi: 10.1016/j.peptides.2003.07.004. [DOI] [PubMed] [Google Scholar]
  • 29.Abdelhamid EE, Sultana M, Portoghese PS, Takemori AE. Selective blockage of delta opioid receptors prevents the development of morphine tolerance and dependence in mice. J Pharmacol Exp Ther. 1991;258:299–303. [PubMed] [Google Scholar]
  • 30.Fundytus ME, Schiller PW, Shapiro M, Weltrowska G, Coderre TJ. The highly selective δ-opioid antagonist H-Tyr-Ticφ[CH2-NH]Phe-Phe-OH (TIPP[φ]) attenuates morphine tolerance and dependence. Eur J Pharmacol. 1995;286:105–108. doi: 10.1016/0014-2999(95)00554-X. [DOI] [PubMed] [Google Scholar]
  • 31.Rothman RB, Danks JA, Jacobson AE, Burke TR, Rice KE, Holaday JW. Morphine tolerance increases mu-noncompetitive delta binding sites. Eur J Pharmacol. 1986;124:113–119. doi: 10.1016/0014-2999(86)90130-5. [DOI] [PubMed] [Google Scholar]
  • 32.Yukhananov RY, Klodt PM, Riva AD, Zeitsev SV, Masky AI. Opiate withdrawal correlates with the presence of DSLET high-affinity binding. Pharmacol Biochem Behav. 1994;49:1109–1112. doi: 10.1016/0091-3057(94)90273-9. [DOI] [PubMed] [Google Scholar]
  • 33.Kest B, Lee CY, McLemore GL, Inturrisi CE. An antisense oligodeoxynucleotide to the delta opioid receptor (DOR-1) inhibits morphine tolerance and acute dependence in mice. Brain Res Bull. 1996;39:185–188. doi: 10.1016/0361-9230(95)02092-6. [DOI] [PubMed] [Google Scholar]
  • 34.Zhu Y, King MA, Schuller AG, et al. Retention of supraspinal delta-like analgesia and loss of morphine tolerance in delta opioid receptor knock-out mice. Neuron. 1999;24:243–252. doi: 10.1016/S0896-6273(00)80836-3. [DOI] [PubMed] [Google Scholar]
  • 35.Gomes I, Gupta A, Filipovska J, Szeto HH, Pintar JE, Devi LA. A role for heterodimerization of μ and δ opiate receptors in enhancing morphine analgesia. Proc Natl Acad Sci USA. 2004;101:5135–5139. doi: 10.1073/pnas.0307601101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Freye E, Latasch L, Portoghese PS. The delta receptor is involved in sufentanil-induced respiratory depression—opioid subreceptors mediate different effects. Eur J Anaesthesiol. 1992;9:457–462. [PubMed] [Google Scholar]
  • 37.Foxx-Orenstein AE, Jin JG, Grider JR. 5TH4 receptor agonists and delta opioid receptor antagonists act synergistically to stimulate colonic propulsion. Am J Physiol. 1998;275:G979–G983. doi: 10.1152/ajpgi.1998.275.5.G979. [DOI] [PubMed] [Google Scholar]
  • 38.Schiller PW, Nguyen TM-D, Weltrowska G, et al. Differential stereochemical requirements of μ vs δ opioid receptors for ligand binding and signal transduction: development of a class of potent and highly δ-selective peptide antagonists. Proc Natl Acad Sci USA. 1992;89:11871–11875. doi: 10.1073/pnas.89.24.11871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Schiller PW, Fundytus ME, Merovitz L, et al. The opioid μ agonist/δ antagonist DIPP-NH2[φ] produces a potent analgesic effect, no physical dependence, and less tolerance than morphine in rats. J Med Chem. 1999;42:3520–3526. doi: 10.1021/jm980724+. [DOI] [PubMed] [Google Scholar]
  • 40.Schiller PW, Weltrowska G, Berezowska I, et al. The TIPP opioid peptide family: development of δ antagonists, δ agonists and mixed μ agonist/δ antagonists. Biopolymers (Peptide Science) 1999;51:411–425. doi: 10.1002/(SICI)1097-0282(1999)51:6<411::AID-BIP4>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
  • 41.Weltrowska G, Lemieux C, Chung NN, Schiller PW. A chimeric opioid peptide with mixed μ agonist/δ antagonist properties. J Pept Res. 2004;63:63–68. doi: 10.1111/j.1399-3011.2003.00108.x. [DOI] [PubMed] [Google Scholar]
  • 42.Ananthan S, Kezar HS, Carter RL, et al. Synthesis, opioid receptor binding, and biological activities of naltrexone-derived pyrido-and pyrimidomorphinans. J Med Chem. 1999;42:3527–3538. doi: 10.1021/jm990039i. [DOI] [PubMed] [Google Scholar]
  • 43.Ananthan S, Khare NK, Saini SK, et al. Identification of opioid ligands possessing mixed μ agonist/δ antagonist activity among pyridomorphinans derived from naloxone, oxymorphone, and hydromorphone. J Med Chem. 2004;47:1400–1412. doi: 10.1021/jm030311v. [DOI] [PubMed] [Google Scholar]

Articles from The AAPS Journal are provided here courtesy of American Association of Pharmaceutical Scientists

RESOURCES