Metabolically stable neurotensin analogs exert potent and long-acting analgesia without hypothermia

https://doi.org/10.1016/j.bbr.2021.113189Get rights and content

Highlights

  • Incorporation of reduced amide bond, silaproline and TMSAla in NT(8−13) resulted in JMV5296, a 25-fold NTS2-selective analog.

  • Combination of these three chemical modifications increased resistance toward proteases having a plasma stability over 20 h.

  • Intrathecal injection of JMV5296 induced potent antinociception in acute, tonic and chronic inflammatory pain models.

  • Central delivery of JMV5296 had no impact on body temperature.

Abstract

The endogenous tridecapeptide neurotensin (NT) has emerged as an important inhibitory modulator of pain transmission, exerting its analgesic action through the activation of the G protein-coupled receptors, NTS1 and NTS2. Whereas both NT receptors mediate the analgesic effects of NT, NTS1 activation also produces hypotension and hypothermia, which may represent obstacles for the development of new pain medications. In the present study, we implemented various chemical strategies to improve the metabolic stability of the biologically active fragment NT(8−13) and assessed their NTS1/NTS2 relative binding affinities. We then determined their ability to reduce the nociceptive behaviors in acute, tonic, and chronic pain models and to modulate blood pressure and body temperature. To this end, we synthesized a series of NT(8−13) analogs carrying a reduced amide bond at Lys8-Lys9 and harboring site-selective modifications with unnatural amino acids, such as silaproline (Sip) and trimethylsilylalanine (TMSAla). Incorporation of Sip and TMSAla respectively in positions 10 and 13 of NT(8−13) combined with the Lys8-Lys9 reduced amine bond (JMV5296) greatly prolonged the plasma half-life time over 20 h. These modifications also led to a 25-fold peptide selectivity toward NTS2. More importantly, central delivery of JMV5296 was able to induce a strong antinociceptive effect in acute (tail-flick), tonic (formalin), and chronic inflammatory (CFA) pain models without inducing hypothermia. Altogether, these results demonstrate that the chemically-modified NT(8−13) analog JMV5296 exhibits a better therapeutic profile and may thus represent a promising avenue to guide the development of new stable NT agonists and improve pain management.

Introduction

Chronic pain is an important public health problem affecting more than 20 % of the worldwide population [1]. Opioid drugs are extensively used in the treatment of chronic pain despite a long list of undesired effects and their limited long-term efficacy to relieve pain for many patients [2,3]. Even with the growing awareness of the risks associated with opioid misuse, overdose and addiction, opioid use is still rising, thus leading to the current opioid crisis in North America [4,5]. Thus, the societal and economic burden, healthcare costs and high prevalence of chronic pain encourage researchers to seek for new pain medications with an increased benefit/risk ratio [6,7]. Among the current development strategies, drugs targeting non-opioid G protein-coupled receptors (GPCR) represent a promising therapeutic avenue in pain research [7].

The development of peptide-based therapeutics is undergoing an exciting revival in the last decade, when compared to small molecule drugs [8,9]. Peptides often offer high target selectivity and specificity as well as enhanced efficacy, safety and tolerability profiles. However, naturally occurring peptides are often not directly suitable for clinical use due to low oral bioavailability, poor blood-brain barrier (BBB) penetration, and short half-life in physiological fluids related to their poor resistance to proteolytic degradation [8,10].

Among the promising alternatives to opioid pain medications, neurotensin (NT) receptors emerge as attractive targets for the treatment of pain [7,11]. Neurotensin (NT) is a small neuropeptide of 13 amino acids (pGlu-Leu-Tyr-Glu-Asn-Lys-Pro-Arg-Arg-Pro-Tyr-Ile-Leu) [12] known to mediate its physiological effects through its binding to two receptors that belongs to 7TMRs superfamily, namely NTS1 and NTS2 [13]. Peripherally, NT acts as a hormone in the cardiovascular system where it induces a drop in blood pressure [14,15], controls appetite [16] and regulates gastrointestinal motility [17]. When administered directly into the central nervous system (CNS), NT is known to play a role in the regulation of anxiety [18,19] as well as in dopaminergic (DAergic)-associated diseases, such as schizophrenia, drug abuse, and Parkinson’s disease [[20], [21], [22]]. NT and its analogs also produce persistent hypothermia [20,23] and analgesia [[24], [25], [26]]. Both NTS1 and NTS2 receptors are present in CNS regions involved in nociceptive transmission and pain modulation, such as the spinal dorsal horn, periaqueductal gray (PAG), rostroventral medulla (RVM), dorsal raphe nucleus, raphe magnus and pallidus [25,[27], [28], [29]]. Moreover, the neurotensinergic system is gaining further interests for pain relief as NT-induced analgesia is not altered by the administration of the opioid antagonists’ naloxone and naltrexone, thereby supporting that NT receptor activation mediates its antinociception action independently of the opioid system [30].

Amino acids forming the C-terminal moiety of the native NT peptide, at position 8–13 (Arg-Arg-Pro-Tyr-Ile-Leu), were identified as the minimal sequence for NT receptor binding and biological activity [31,32]. Therefore, compound synthesis and structure-activity relationship studies have allowed the development of new NT(8−13) selective analogs targeting either NTS1 or NTS2 receptors and permitted the discrimination of their respective physiological effects [33,34]. While analgesia in acute, tonic, and chronic pain paradigms was demonstrated to be mediated by the activation of both receptors [24,25,28,[34], [35], [36]], only NTS1 activation was associated with hypotension [14,37] and hypothermia [38].

These adverse effects (hypothermia and hypotension) combined with the low metabolic stability of the native NT peptide preclude its therapeutic use as non-opioid pain-relieving medications. Therefore, chemical modifications of the backbone and the incorporation of unnatural amino acids are necessary to optimize the pharmacological properties of newly synthesized NT(8−13) drug candidates. In the present study, we evaluate the impact of site-selective chemical modifications of NT(8−13) to improve the peptide metabolic stability and its analgesic efficacy in different experimental pain models as well as to reduce the unwanted effects triggered by NTS1 activation.

Section snippets

Peptide chemistry and characterization

Full synthetic procedures and characterization of the compounds presented in this study are reported in [39].

Competitive radioligand binding assay

CHO-K1 cells stably expressing hNTS1 (ES-690-C from PerkinElmer, Montréal, Canada) or 1321N1 cells stably expressing hNTS2 (ES-691-C from PerkinElmer) were cultured respectively in DMEM/F12 or DMEM. Culture media were supplemented with 10 % FBS, 100 U/mL penicillin, 100 μg/mL streptomycin, 20 mM HEPES and 0.4 mg/mL G418, and cells were incubated at 37 °C in a humidified chamber at 5% CO2

Rational design of NT(8−13) analogs

Both Arg residues in positions 8 and 9 were replaced by two Lys for ease synthesis (JMV438). It was previously demonstrated that this amino acid di-substitution had no impact on NTS1 and NTS2 receptor binding or activation [[51], [52], [53]]. Therefore, the subsequent chemical modifications of the NT(8−13) sequence were based on the JMV438 derivative.

First, the chemical modifications performed on the NT(8−13) backbone were focused on increasing the metabolic stability of NT(8−13) analogs to

Conclusion and perspectives

The present study reports the characterization of a series of NT(8−13) analogs harboring a reduced amine bond and additional substitutions with unnatural silylated amino acids to give rise to metabolically stable and powerful analgesic pseudopeptide compounds. Incorporation of a reduced amine bond between Lys8-Lys9, Sip in position 10 and a TMSAla in position 13 of NT(8−13) resulted in the generation of JMV5296. These modifications produced a fairly NTS2-selective analog with extended

Funding sources

This research was supported by a grant from the Canadian Institutes for Health Research [grant number FDN-148413].

CRediT authorship contribution statement

Mélanie Vivancos: Methodology, Investigation, Formal analysis, Writing - original draft, Visualization. Roberto Fanelli: Resources. Élie Besserer-Offroy: Validation, Writing - review & editing. Sabrina Beaulieu: Methodology, Investigation. Magali Chartier: Methodology, Investigation. Martin Resua-Rojas: Investigation. Christine E. Mona: Resources. Santo Previti: Resources. Emmanuelle Rémond: Resources. Jean-Michel Longpré: Validation, Writing - review & editing. Florine Cavelier: Validation,

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgments

The authors thank Professors Éric Marsault and Pedro D’Orléan-Juste (Dept. Pharmacology-Physiology, Université de Sherbrooke) for allowing them to use, the UPLC/MS instrument for plasma stability assays and the use of Micro-Med transducer for the blood pressure measurements, respectively.

MV was supported by a PhD scholarship from the Institut de Pharmacologie de Sherbrooke (IPS) and Centre d′Excellence en Neurosciences de l′Université de Sherbrooke (CNS). Funding from theUniversité de

References (89)

  • P.R. Dobner

    Neurotensin and pain modulation

    Peptides

    (2006)
  • P. Kleczkowska et al.

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

    Eur. J. Pharmacol.

    (2013)
  • P. Sarret et al.

    Pharmacology and functional properties of NTS2 neurotensin receptors in cerebellar granule cells

    J. Biol. Chem.

    (2002)
  • B.V. Clineschmidt et al.

    Neurotensin administered intracisternally inhibits responsiveness of mice to noxious stimuli

    Eur. J. Pharmacol.

    (1977)
  • 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)
  • A.V. Buhler et al.

    Neurotensin-produced antinociception in the rostral ventromedial medulla is partially mediated by spinal cord norepinephrine

    Pain

    (2008)
  • A. Remaury et al.

    Targeted inactivation of the neurotensin type 1 receptor reveals its role in body temperature control and feeding behavior but not in analgesia

    Brain Res.

    (2002)
  • D. Hapău et al.

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

    European J. Org. Chem.

    (2016)
  • H. Beaudry et al.

    Differential noxious and motor tolerance of chronic delta opioid receptor agonists in rodents

    Neuroscience

    (2009)
  • J.L. Hylden et al.

    Spinal opioid analgesic effects are enhanced in a model of unilateral inflammation/hyperalgesia: possible involvement of noradrenergic mechanisms

    Eur. J. Pharmacol.

    (1991)
  • C.A. Fairbanks

    Spinal delivery of analgesics in experimental models of pain and analgesia

    Adv. Drug Deliv. Rev.

    (2003)
  • A. Tjolsen et al.

    The formalin test: an evaluation of the method

    Pain

    (1992)
  • 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

    Pain

    (1977)
  • T.J. Coderre et al.

    The formalin test: a validation of the weighted-scores method of behavioural pain rating

    Pain

    (1993)
  • S.R. Chaplan et al.

    Quantitative assessment of tactile allodynia in the rat paw

    J. Neurosci. Methods

    (1994)
  • D. Lugrin et al.

    Reduced peptide bond pseudopeptide analogues of neurotensin: binding and biological activities, and in vitro metabolic stability

    Eur. J. Pharmacol.

    (1991)
  • F. Checler et al.

    Purification and characterization of a novel neurotensin-degrading peptidase from rat brain synaptic membranes

    J. Biol. Chem.

    (1986)
  • E. Besserer-Offroy et al.

    Data set describing the in vitro biological activity of JMV2009, a novel silylated neurotensin(8-13) analog

    Data Brief

    (2020)
  • M. Oishi et al.

    Release of histamine and adrenaline in vivo following intravenous administration of neurotensin

    Life Sci.

    (1983)
  • M. Oishi et al.

    Attenuation of triphasic blood pressure responses induced by shortening of amino acid sequences of neurotensin

    Neuropeptides

    (1982)
  • M. Oishi et al.

    Influence of histamine and prostaglandin on desensitization to neurotensin in rat blood pressure

    Neuropeptides

    (1984)
  • R. Quirion et al.

    Compound 48/80 inhibits neurotensin-induced hypotension in rats

    Life Sci.

    (1980)
  • X. Gu et al.

    Pharmacologically induced hypothermia attenuates traumatic brain injury in neonatal rats

    Exp. Neurol.

    (2015)
  • Y. Zhao et al.

    Pharmacological hypothermia induced neurovascular protection after severe stroke of transient middle cerebral artery occlusion in mice

    Exp. Neurol.

    (2020)
  • D.J. Wustrow et al.

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

    Bioorg. Med. Chem. Lett.

    (1995)
  • B.M. Tyler et al.

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

    Brain Res.

    (1998)
  • M. Boules et al.

    Neurotensin analog selective for hypothermia over antinociception and exhibiting atypical neuroleptic-like properties

    Brain Res.

    (2001)
  • H. Wheeler-Aceto et al.

    The rat paw formalin test: comparison of noxious agents

    Pain

    (1990)
  • C.A. Porro et al.

    Spatial and temporal aspects of spinal cord and brainstem activation in the formalin pain model

    Prog. Neurobiol.

    (1993)
  • P. Tétreault et al.

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

    Physiol. Behav.

    (2011)
  • A.J. Parent et al.

    Increased anxiety-like behaviors in rats experiencing chronic inflammatory pain

    Behav. Brain Res.

    (2012)
  • E. Eiselt et al.

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

    Eur. J. Pharmacol.

    (2019)
  • D.S. Goldberg et al.

    Pain as a global public health priority

    BMC Public Health

    (2011)
  • I. Kissin

    The development of new analgesics over the past 50 years: a lack of real breakthrough drugs

    Anesth. Analg.

    (2010)
  • Cited by (6)

    1

    Present address: Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA

    2

    Present address: Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Vial Annunziata, Messina, Italy.

    3

    Lead Author.

    View full text