Full length article
Pain relief devoid of opioid side effects following central action of a silylated neurotensin analog

https://doi.org/10.1016/j.ejphar.2020.173174Get rights and content

Abstract

Neurotensin (NT) exerts naloxone-insensitive antinociceptive action through its binding to both NTS1 and NTS2 receptors and NT analogs provide stronger pain relief than morphine on a molecular basis. Here, we examined the analgesic/adverse effect profile of a new NT(8–13) derivative denoted JMV2009, in which the Pro10 residue was substituted by a silicon-containing unnatural amino acid silaproline. We first report the synthesis and in vitro characterization (receptor-binding affinity, functional activity and stability) of JMV2009. We next examined its analgesic activity in a battery of acute, tonic and chronic pain models. We finally evaluated its ability to induce adverse effects associated with chronic opioid use, such as constipation and analgesic tolerance or related to NTS1 activation, like hypothermia. In in vitro assays, JMV2009 exhibited high binding affinity for both NTS1 and NTS2, improved proteolytic resistance as well as agonistic activities similar to NT, inducing sustained activation of p42/p44 MAPK and receptor internalization. Intrathecal injection of JMV2009 produced dose-dependent antinociceptive responses in the tail-flick test and almost completely abolished the nociceptive-related behaviors induced by chemical somatic and visceral noxious stimuli. Likewise, increasing doses of JMV2009 significantly reduced tactile allodynia and weight bearing deficits in nerve-injured rats. Importantly, repeated agonist treatment did not result in the development of analgesic tolerance. Furthermore, JMV2009 did not cause constipation and was ineffective in inducing hypothermia. These findings suggest that NT drugs can act as an effective opioid-free medication for the management of pain or can serve as adjuvant analgesics to reduce the opioid adverse effects.

Introduction

Today, opioids remain at the forefront of the pharmacological treatment of chronic pain (de Leon-Casasola, 2013; Gomes et al., 2014). However, opioid therapy is associated with a wide range of side effects, such as constipation, nausea, drowsiness, dizziness, sleep disturbance, dry mouth, respiratory depression and tolerance that negatively affects patients’ well-being and even discourages them for continuing their medications (Benyamin et al., 2008; Bruneau et al., 2018; Devulder et al., 2005; McNicol et al., 2003). In the ongoing opioid crisis, involving drug misuse, abuse, overdose and death (Coyle et al., 2018), there is therefore an urgent need for new, effective and safe non-opioid analgesics to improve the care of patients suffering from chronic pain (Besserer-Offroy and Sarret, 2019).

Among the alternative to opioids, neurotensin (NT) receptors recently emerged as attractive targets to develop new painkillers (Perez de Vega et al., 2018). Through its binding with two G protein-coupled receptors (GPCRs), referred to as NTS1 and NTS2, the tridecapeptide NT(1–13) was indeed found to induce a potent opioid-independent analgesia in a variety of pain paradigms following systemic or central administration (Dobner, 2006; Feng et al., 2014; Kleczkowska and Lipkowski, 2013; Sarret and Cavelier, 2018). Accordingly, it was pharmacologically demonstrated that the antinociceptive responses induced by NT and its analogs, designed and synthesized based on the minimal biologically active C-terminal region (Arg8-Arg9-Pro10-Tyr11-Ile12-Leu13), could not be reversed by the opioid antagonists, naloxone and naltrexone (Behbehani and Pert, 1984; Bredeloux et al., 2006; Osbahr et al., 1981; Wustrow et al., 1995). Importantly, NT was also reported to provide stronger pain relief than morphine on a molecular basis (al-Rodhan et al., 1991; Nemeroff et al., 1979).

In the last two decades, peptide therapeutics have gained increased interest from the biomedical and pharmaceutical industry, filling the gap between the two main classes of available market drugs, small molecules and biologics (Fosgerau and Hoffmann, 2015; Kaspar and Reichert, 2013). Peptides are definitely attractive drug candidates due to their high potency/efficacy, target specificity and safety/tolerability profile. Despite offering many advantages, their extensive use as therapeutics is, however, limited by their short half-life and rapid plasma clearance, which leads to low bioavailability (Fosgerau and Hoffmann, 2015; McGonigle, 2012). Several strategies have therefore been developed to overcome the undesirable physicochemical properties of therapeutic peptides. These approaches successfully applied to the C-terminal NT(8–13) hexapeptide include backbone and side-chain modifications, such as d-amino acid and non-natural amino acid substitutions, incorporation of reduced bonds, lactam bridged mimetics or peptide-peptoid hybrids, N- and C-terminal modifications as well as introduction of conformational constraints via cyclization (Bittermann et al., 2004; Boules et al., 2010; Bredeloux et al., 2008; Einsiedel et al., 2011; Fanelli et al., 2015; Hughes et al., 2010; Labbé-Jullié et al., 1994; Sarret and Cavelier, 2018; Sousbie et al., 2018; Tyler-McMahon et al., 2000; Wustrow et al., 1995).

According to recent structure-activity relationship (SAR) studies, structural modifications with unnatural amino acids were effective in producing NT(8–13) analogs with improved metabolic stability, receptor selectivity and in vivo activity (Boules et al., 2010; Fanelli et al., 2015; Hapău et al., 2016; Hughes et al., 2010; Magafa et al., 2019; Tokumura et al., 1990; Tyler-McMahon et al., 2000). In that respect, non-natural silylated amino acids are of great interest for their ability to modulate the physicochemical properties of bioactive peptides (Cavelier et al., 2002, 2004; Dalkas et al., 2010; Fanelli et al., 2015, 2017; Remond et al., 2016). Herein, we report the synthesis and in vitro/in vivo characterization of a new NT(8–13) analog, named JMV2009, which binds to both NTS1 and NTS2 receptors. In this compound, the Pro10 residue was substituted by a silicon-containing unnatural amino acid proline surrogate, denoted silaproline (Sip), which has been shown to retain peptide conformation (Cavelier et al., 2002, 2006; Remond et al., 2017; Vivet et al., 2000). This Sip-containing NT(8–13) analog was further screened in a battery of pain models to assess its putative analgesic properties and evaluate for its ability to induce adverse effects associated with chronic opioid therapy, such as constipation or analgesic tolerance.

Section snippets

Materials

PD149163 was obtained by the U.S. National Institute of Mental Health, Chemical Synthesis and Drug Supply Program. Morphine sulfate was purchased from Sandoz (Boucherville, QC, Canada).

Synthesis and in vitro characterization of JMV2009

Synthesis, chemical characterization, and in vitro and in cellulo characterization (binding, signaling and plasma stability) of JMV2009 are reported into the accompanying Data in Brief article (Besserer-Offroy et al., 2020).

Animals

Experimental procedures were approved by the Animal Care and Ethical Committee of the

JMV2009 acts as a non-selective NT receptor agonist

We report here the synthesis and pharmacological evaluation of a NT(8–13) analog (JMV2009) carrying an unnatural silicon-containing proline surrogate, the 4-(dimethyl)silaproline (Sip) in replacement of the proline residue at position 10 (Figs. 1–2 and Scheme 1 of the accompanying Data in Brief article, Besserer-Offroy et al., 2020). We first found that this silaproline-containing peptide exhibited relatively high and similar binding affinities for both NTS1 and NTS2 receptors, with only

Discussion

G-protein coupled receptors are the target of more than 25% of the FDA-approved drugs (Overington et al., 2006) and represent one of the largest classes of therapeutic targets in pain medicine (Stone and Molliver, 2009). Nowadays, validation of new molecular targets and development of new effective, non-addicting pain medications are critical, but still remain extremely slow (Kissin, 2010; Yaksh et al., 2018). In the past decades, both NTS1 and NTS2 have generated a growing interest as

Conclusion

In conclusion, we demonstrate here that the NT(8–13) peptide can accommodate the substitution of the proline residue to give rise to a powerful analgesic peptide. We further provide evidence that NT agonists do not induce the common adverse effects associated with the use of opioids (i.e. constipation and tolerance), thus offering a non-opioid option for the treatment of pain. Finally, these findings suggest that NT agonists used combined with opioid therapy might contribute to adequate pain

CRediT authorship contribution statement

Pascal Tétreault: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Writing - original draft, Writing - review & editing, Visualization. Élie Besserer-Offroy: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Writing - original draft, Writing - review & editing, Visualization. Rebecca L. Brouillette: Investigation. Adeline René: Resources. Alexandre Murza: Investigation. Roberto Fanelli: Resources. Karyn Kirby: Investigation. Alexandre J.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships which have, or could be perceived to have, influenced the work reported in this article.

Acknowledgements

PT is the recipient of a post-doctoral fellowship from the Canadian Institutes for Health Research (CIHR, grant MFE-135472). ÉBO is the recipient of a Fond de recherche du Québec – Santé (FRQ-S, grant 255989) and a CIHR (grant MFE-164740) research fellowships. RLB has been awarded a FRQ-S and a Faculty of Medicine and Health Sciences of the Université de Sherbrooke Ph.D. scholarships. PS is the recipient of a Tier 1 Canada Research Chair in Neurophysiopharmacology of Chronic Pain and a member

References (100)

  • E.J. Cobos et al.

    Inflammation-induced Decrease in Voluntary Wheel Running in Mice: A Nonreflexive Test for Evaluating Inflammatory Pain and Analgesia

    Pain

    (2012)
  • O.A. de Leon-Casasola

    Opioids for chronic pain: new evidence, new strategies, safe prescribing

    Am. J. Med.

    (2013)
  • P.R. Dobner

    Neurotensin and pain modulation

    Peptides

    (2006)
  • I. Dubuc et al.

    JMV 449: a pseudopeptide analogue of neurotensin-(8-13) with highly potent and long-lasting hypothermic and analgesic effects in the mouse

    Eur. J. Pharmacol.

    (1992)
  • I. Dubuc et al.

    Tolerance to the hypothermic but not to the analgesic effect of [D-Trp11]neurotensin during the semichronic intracerebroventricular infusion of the peptide in rats

    Peptides

    (1994)
  • D. Dubuisson et al.

    The Formalin Test: a Quantitative Study of the Analgesic Effects of Morphine, Meperidine, and Brain Stem Stimulation in Rats and Cats

    (1977)
  • E. Eiselt et al.

    The combination of opioid and neurotensin receptor agonists improves their analgesic/adverse effect ratio

    Eur. J. Pharmacol.

    (2019)
  • R. Fanelli et al.

    Hydrophobic alpha,alpha-disubstituted disilylated TESDpg induces incipient 310-helix in short tripeptide sequence

    Org. Lett.

    (2017)
  • W.E. Fantegrossi et al.

    Antinociceptive, hypothermic, hypotensive, and reinforcing effects of a novel neurotensin receptor agonist, NT69L, in rhesus monkeys

    Pharmacol. Biochem. Behav.

    (2005)
  • D. Feifel et al.

    The acute and subchronic effects of a brain-penetrating, neurotensin-1 receptor agonist on feeding, body weight and temperature

    Neuropharmacology

    (2010)
  • K. Fosgerau et al.

    Peptide therapeutics: current status and future directions

    Drug Discov. Today

    (2015)
  • I. Gaumond et al.

    Sex differences in opioid-mediated pain inhibitory mechanisms during the interphase in the formalin test

    Neuroscience

    (2007)
  • D. Hapău et al.

    Stereoselective synthesis of β-(5-arylthiazolyl) α-amino acids and use in neurotensin analogues

    Eur. J. Org Chem.

    (2016)
  • A.A. Kaspar et al.

    Future directions for peptide therapeutics development

    Drug Discov. Today

    (2013)
  • P. Kleczkowska et al.

    Neurotensin and neurotensin receptors: characteristic, structure-activity relationship and pain modulation--a review

    Eur. J. Pharmacol.

    (2013)
  • J.L. Lau et al.

    Therapeutic peptides: historical perspectives, current development trends, and future directions

    Bioorg. Med. Chem.

    (2018)
  • P. McGonigle

    Peptide therapeutics for CNS indications

    Biochem. Pharmacol.

    (2012)
  • E. McNicol et al.

    Management of opioid side effects in cancer-related and chronic noncancer pain: a systematic review

    J. Pain

    (2003)
  • E. Nakazato-Imasato et al.

    Pharmacological characteristics of the hind paw weight bearing difference induced by chronic constriction injury of the sciatic nerve in rats

    Life Sci.

    (2009)
  • S. Pellissier et al.

    Peripheral neurotensin participates in the modulation of pre- and postprandial intestinal motility in rats

    Neuropeptides

    (1996)
  • M.J. Perez de Vega et al.

    Recent progress in non-opioid analgesic peptides

    Arch. Biochem. Biophys.

    (2018)
  • K. Thor et al.

    Neurotensin increases colonic motility

    Gastroenterology

    (1986)
  • B.M. Tyler et al.

    Evidence for additional neurotensin receptor subtypes: neurotensin analogs that distinguish between neurotensin-mediated hypothermia and antinociception

    Brain Res.

    (1998)
  • B.M. Tyler-McMahon et al.

    Highly potent neurotensin analog that causes hypothermia and antinociception

    Eur. J. Pharmacol.

    (2000)
  • P. Tétreault et al.

    Weight bearing evaluation in inflammatory, neuropathic and cancer chronic pain in freely moving rats

    Physiol. Behav.

    (2011)
  • D.J. Wustrow et al.

    Reduced amide bond neurotensin 8-13 mimetics with potent in vivo activity

    Bioorg. Med. Chem. Lett

    (1995)
  • T.L. Yaksh et al.

    Development of new analgesics: an answer to opioid epidemic

    Trends Pharmacol. Sci.

    (2018)
  • R. Yamauchi et al.

    Antinociception induced by beta-lactotensin, a neurotensin agonist peptide derived from beta-lactoglobulin, is mediated by NT2 and D1 receptors

    Life Sci.

    (2003)
  • K. Angeby-Moller et al.

    Using the CatWalk method to assess weight-bearing and pain behaviour in walking rats with ankle joint monoarthritis induced by carrageenan: effects of morphine and rofecoxib

    J. Neurosci. Methods

    (2008)
  • R. Benyamin et al.

    Opioid complications and side effects

    Pain Physician

    (2008)
  • E. Besserer-Offroy et al.

    The signaling signature of the neurotensin type 1 receptor with endogenous ligands

    Eur. J. Pharmacol.

    (2017)
  • E. Besserer-Offroy et al.

    Sending out biased signals: an appropriate proposition for pain?

    Douleur Analgésie

    (2019)
  • É. Besserer-Offroy et al.

    Data Set Describing the in Vitro Biological Activity of JMV2009, a Novel Silylated Neurotensin(8-13) Analog

    (2020)
  • G. Bissette et al.

    Hypothermia and intolerance to cold induced by intracisternal administration of the hypothalamic peptide neurotensin

    Nature

    (1976)
  • H. Bittermann et al.

    Evaluation of lactam-bridged neurotensin analogues adjusting psi(Pro10) close to the experimentally derived bioactive conformation of NT(8-13)

    J. Med. Chem.

    (2004)
  • M. Boules et al.

    Analgesic synergy of neurotensin receptor subtype 2 agonist NT79 and morphine

    Behav. Pharmacol.

    (2011)
  • B.L. Bowers et al.

    The evolving role of long-term pharmacotherapy for opioid-induced constipation in patients being treated for noncancer pain

    J. Pharm. Pract.

    (2017)
  • P. Bredeloux et al.

    Synthesis and biological effects of c(Lys-Lys-Pro-Tyr-Ile-Leu-Lys-Lys-Pro-Tyr-Ile-Leu) (JMV2012), a new analogue of neurotensin that crosses the blood-brain barrier

    J. Med. Chem.

    (2008)
  • J. Bruneau et al.

    Management of opioid use disorders: a national clinical practice guideline

    CMAJ (Can. Med. Assoc. J.)

    (2018)
  • F. Cavelier et al.

    Biological activity of silylated amino acid containing substance P analogues

    J. Pept. Res.

    (2004)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    2

    Present address: Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada.

    3

    Lead Authors.

    View full text