Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 788: 176-182, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27266667

ABSTRACT

Neuropathic pain remains difficult to treat due to the involvement of various pathophysiological mechanisms in its pathogeny. Among the different opioidergic systems the enkephalinergic one is primarily recruited via activation of delta opioid receptor (DOP) in chronic pain and of mu opioid receptor (MOP) in acute pain. To investigate the role of their endogenous ligands Met and Leu-enkephalin in neuropathic pain control, a dual inhibitor of their degrading enzymes, PL265, which acts restrictively at the level of peripheral nociceptors, was administered per os to assess its efficacy in pain prevention and alleviation using a partial sciatic nerve ligation model (PSNL) in mice. We demonstrated here that the pre-injury oral administration of PL265 (50mg/kg) during the 9 days of neuropathy development reduces thermal hyperalgesia and mechanical allodynia for two weeks after the end of treatment. The repeated administration (50mg/kg daily, during 10 days) does not induce tolerance. Therefore, protecting the enkephalins released at the peripheral level during neuropathic pain with oral PL265 seems to be a promising approach to prevent and alleviate the painful symptoms of neuropathic pain in humans without the unwanted effects of exogenous opiates such as morphine.


Subject(s)
Alanine/analogs & derivatives , Biphenyl Compounds/pharmacology , Neprilysin/antagonists & inhibitors , Neuralgia/diet therapy , Neuralgia/prevention & control , Protease Inhibitors/pharmacology , Alanine/administration & dosage , Alanine/adverse effects , Alanine/pharmacology , Alanine/therapeutic use , Animals , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/adverse effects , Biphenyl Compounds/therapeutic use , Body Weight/drug effects , Disease Models, Animal , Hyperalgesia/prevention & control , Male , Mice , Neuralgia/enzymology , Protease Inhibitors/administration & dosage , Protease Inhibitors/adverse effects , Protease Inhibitors/therapeutic use , Safety , Time Factors , Touch/drug effects
2.
Eur J Med Chem ; 102: 58-67, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26241877

ABSTRACT

The endogenous opioid system, essentially constituted by two opioid receptors which are stimulated by the natural internal effectors enkephalins (Met-enkephalin and Leu-enkephalin), is present at the different sites (peripheral, spinal, central) of the control of pain. We have demonstrated that the protection of the enkephalin inactivation by the two metallopeptidases (neprilysin and neutral aminopeptidase) increases their local concentration selectively induced by pain stimuli triggering analgesic responses. With the aim of increasing the orally antinociceptive responses of the previously described disulfide DENKIs ( [Formula: see text] CH(R1)CH2-S-S-CH2-C(R2R3)CONHCH(R4)COOR5), we designed new pro-drugs, in the same chemical series, with a transient protection of the free amino group by an acyloxyalkyl carbamate, giving rise to ((CH3)2CHCO2CH(CH3)OCONHCH(R1)CH2-S-S-CH2-C(R2R3)CONHCH(R4)COOR5) pro-drugs 2a-2g. These compounds were easily prepared from their parent analogs, with a good yield. They were tested per os and shown to be highly efficient in peripherally-controlled inflammatory and neuropathic pain with long lasting effects but completely inactive in the acute centrally-controlled hot plate test, a model of pain by excess of nociception. This demonstrates that DENKIs are able to relieve pain at its source thanks to the increase of enkephalin levels.


Subject(s)
Analgesics/pharmacology , Disulfides/pharmacology , Neprilysin/antagonists & inhibitors , Pain/drug therapy , Protease Inhibitors/pharmacology , Administration, Oral , Analgesics/administration & dosage , Analgesics/chemistry , Animals , Disulfides/administration & dosage , Disulfides/chemistry , Male , Mice , Mice, Inbred Strains , Neprilysin/metabolism , Pain Measurement , Protease Inhibitors/administration & dosage , Protease Inhibitors/chemistry
3.
Pharmacol Res Perspect ; 3(2): e00116, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25692029

ABSTRACT

The peripheral endogenous opioid system is critically involved in neuropathic and inflammatory pain generation as suggested by the modulation of opioid receptors expression and enkephalins (ENKs) release observed in these painful conditions. Accordingly, an innovative approach in the treatment of these nocifensive events is to increase and maintain high local concentrations of extracellular pain-evoked ENKs, by preventing their physiological enzymatic inactivation by two Zn metallopeptidases, the neutral endopeptidase (NEP, neprilysin, EC 3.4.24.11) and the neutral aminopeptidase (APN, EC 3.4.11.2). With this aim, new orally active dual ENKephalinase inhibitors (DENKIs) were designed as soluble prodrugs by introducing a N-terminal cleavable carbamate in the previously described aminophosphinic inhibitors. This induces long-lasting antinociceptive responses after oral administration, in various rodent models of inflammatory and neuropathic pain. These responses are mediated through stimulation of peripheral opioid receptors by DENKIs-protected ENKs as demonstrated by naloxone methiodide reversion. In all tested models, the most efficient prodrug 2a (PL265) was active, at least during 150-180 min, after single oral administration of 25-50 mg/kg in mice and of 100-200 mg/kg in rats. In models of neuropathic pain, both hyperalgesia and allodynia were markedly reduced. Interestingly, combination of inactive doses of 2a (PL265) and of the anti-epileptic drug gabapentin had synergistic effect on neuropathic pain. Pharmacokinetic studies of 2a (PL265) in rats show that the active drug is the only generated metabolite produced. These encouraging results have made 2a (PL265) a suitable candidate for clinical development.

4.
J Med Chem ; 57(13): 5748-63, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24927250

ABSTRACT

Protecting enkephalins, endogenous opioid peptides released in response to nociceptive stimuli, is an innovative approach for acute and neuropathic pain alleviation. This is achieved by inhibition of their enzymatic degradation by two membrane-bound Zn-metallopeptidases, neprilysin (NEP, EC 3.4.24.11) and aminopeptidase N (APN, EC 3.4.11.2). Selective and efficient inhibitors of both enzymes, designated enkephalinases, have been designed that markedly increase extracellular concentrations and half-lives of enkephalins, inducing potent antinociceptive effects. Several chemical families of Dual ENKephalinase Inhibitors (DENKIs) have previously been developed but devoid of oral activity. We report here the design and synthesis of new pro-drugs, derived from co-drugs combining a NEP and an APN inhibitor through a disulfide bond with side chains improving oral bioavailability. Their pharmacological properties were assessed in various animal models of pain targeting central and/or peripheral opioid systems. Considering its efficacy in acute and neuropathic pain, one of these new DENKIs, 19-IIIa, was selected for clinical development.


Subject(s)
Disulfides/chemical synthesis , Neuralgia/drug therapy , Phenylpropionates/chemical synthesis , Protease Inhibitors/administration & dosage , Administration, Oral , Analgesics/administration & dosage , Analgesics/chemical synthesis , Analgesics/pharmacokinetics , Animals , CD13 Antigens/antagonists & inhibitors , Disulfides/administration & dosage , Disulfides/pharmacokinetics , Enkephalins/metabolism , Humans , Male , Mice , Neprilysin/antagonists & inhibitors , Neuralgia/enzymology , Phenylpropionates/administration & dosage , Phenylpropionates/pharmacokinetics , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Propylamines , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacokinetics , Rats
5.
Eur J Biochem ; 270(20): 4187-99, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14519131

ABSTRACT

Peptides which should be generated from the neuropeptide FF (NPFF) precursor were identified in a neuronal (human neuroblastoma SH-SY5Y) cell line and in COS-7 cells after transient transfection of the human proNPFFA cDNA and were compared with those detected in the mouse spinal cord. After reverse-phase high performance liquid chromatography of soluble material, NPFF-related peptides were immunodetected with antisera raised against NPFF and identified by using on-line capillary liquid chromatography/nanospray ion trap tandem mass spectrometry. Neuronal and non-neuronal cells generated different peptides from the same precursor. In addition to NPFF, SQA-NPFF (Ser-Gln-Ala-Phe-Leu-Phe-Gln-Pro-Gln-Arg-Phe-amide) and NPAF were identified in the human neuroblastoma while only NPFF was clearly identified in COS-7 cells. In mouse, in addition to previously detected NPFF and NPSF, SPA-NPFF (Ser-Pro-Ala-Phe-Leu-Phe-Gln-Pro-Gln-Arg-Phe-amide), the homologous peptide of SQA-NPFF, were characterized. These data on intracellular processing of proNeuropeptide FFA are discussed in regard to the known enzymatic processing mechanisms.


Subject(s)
Oligopeptides/isolation & purification , Peptides/isolation & purification , Animals , COS Cells , Chromatography, High Pressure Liquid , Humans , Mass Spectrometry , Mice , Neuroblastoma/metabolism , Spinal Cord/metabolism
6.
Peptides ; 23(6): 1107-13, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12126738

ABSTRACT

Pharmacological studies have implicated the anti-opioid neuropeptide FF (NPFF) in the modulation of pain transmission. Since its physiological role has not yet been fully elucidated, the present study examined whether antisense peptide nucleic acid (PNA) complementary to the NPFF precursor (proNPFF(A)) modified pain sensitivity. Mice received three intraperitoneal (i.p.) injections (10mg/kg) of antisense PNA (As-proNPFF(A)) over a period of 24h. As-proNPFF(A) treatment significantly increased the basal tail withdrawal latency in the tail-flick test. This analgesia persisted during 2 days and was completely reversed by naloxone. Thus, antisense PNAs, by decreasing anti-opioid effects, revealed a basal endogenous opioid activity. Our results evidence a physiological interplay between NPFF and opioid systems and further support the use of PNA as effective antisense agents, for studying gene function in vivo.


Subject(s)
Narcotics/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Nucleic Acids/metabolism , Animals , Chromatography, High Pressure Liquid , Mice , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oligonucleotides, Antisense/pharmacology , Pain/drug therapy , Radioimmunoassay , Spinal Cord/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...