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.
Addict Biol ; 23(5): 1010-1019, 2018 09.
Article in English | MEDLINE | ID: mdl-28944554

ABSTRACT

Cebranopadol is a novel potent analgesic agonist at the nociceptin/orphanin FQ peptide (NOP) and classical opioid receptors. As NOP receptor activation has been shown to reduce side effects related to the activation of µ-opioid peptide (MOP) receptors, the present study evaluated opioid-type physical dependence produced by cebranopadol in mice and rats. In a naloxone-precipitated withdrawal assay in mice, a regimen of seven escalating doses of cebranopadol over 2 days produced only very limited physical dependence as evidenced by very little withdrawal symptoms (jumping) even at cebranopadol doses clearly exceeding the analgesic dose range. In contrast, mice showed clear withdrawal symptoms when treated with morphine within the analgesic dose range. In the rat, spontaneous withdrawal (by cessation of drug treatment; in terms of weight loss and behavioral score) was studied after 4-week subacute administration. Naloxone-precipitated withdrawal (in terms of weight loss and behavioral score) was studied in the same groups of rats after 1-week re-administration following the spontaneous withdrawal period. In both tests, cebranopadol-treated rats showed only few signs of withdrawal, while withdrawal effects in rats treated with morphine were clearly evident. These findings demonstrate a low potential of cebranopadol to produce opioid-type physical dependence in rodents. The prospect of this promising finding into the clinical setting remains to be established.


Subject(s)
Behavior, Animal/drug effects , Indoles/pharmacology , Opioid-Related Disorders/prevention & control , Receptors, Opioid/drug effects , Spiro Compounds/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice , Rats , Rats, Wistar , Substance Withdrawal Syndrome/prevention & control
2.
ACS Med Chem Lett ; 5(8): 851-6, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25147602

ABSTRACT

We report the discovery of spiro[cyclohexane-pyrano[3,4-b]indole]-amines, as functional nociceptin/orphanin FQ peptide (NOP) and opioid receptor agonists with strong efficacy in preclinical models of acute and neuropathic pain. Utilizing 4-(dimethylamino)-4-phenylcyclo-hexanone 1 and tryptophol in an oxa-Pictet-Spengler reaction led to the formation of spiroether 2, representing a novel NOP and opioid peptide receptor agonistic chemotype. This finding initially stems from the systematic derivatization of 1, which resulted in alcohols 3-5, ethers 6 and 7, amines 8-10, 22-24, and 26-28, amides 11 and 25, and urea 12, many with low nanomolar binding affinities at the NOP and mu opioid peptide (MOP) receptors.

3.
ACS Med Chem Lett ; 5(8): 857-62, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25147603

ABSTRACT

In a previous communication, our efforts leading from 1 to the identification of spiro[cyclohexane-dihydropyrano[3,4-b]indole]-amine 2a as analgesic NOP and opioid receptor agonist were disclosed and their favorable in vitro and in vivo pharmacological properties revealed. We herein report our efforts to further optimize lead 2a, toward trans-6'-fluoro-4',9'-dihydro-N,N-dimethyl-4-phenyl-spiro[cyclohexane-1,1'(3'H)-pyrano[3,4-b]indol]-4-amine (cebranopadol, 3a), which is currently in clinical development for the treatment of severe chronic nociceptive and neuropathic pain.

4.
J Pharmacol Exp Ther ; 349(3): 535-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24713140

ABSTRACT

Cebranopadol (trans-6'-fluoro-4',9'-dihydro-N,N-dimethyl-4-phenyl-spiro[cyclohexane-1,1'(3'H)-pyrano[3,4-b]indol]-4-amine) is a novel analgesic nociceptin/orphanin FQ peptide (NOP) and opioid receptor agonist [Ki (nM)/EC50 (nM)/relative efficacy (%): human NOP receptor 0.9/13.0/89; human mu-opioid peptide (MOP) receptor 0.7/1.2/104; human kappa-opioid peptide receptor 2.6/17/67; human delta-opioid peptide receptor 18/110/105]. Cebranopadol exhibits highly potent and efficacious antinociceptive and antihypersensitive effects in several rat models of acute and chronic pain (tail-flick, rheumatoid arthritis, bone cancer, spinal nerve ligation, diabetic neuropathy) with ED50 values of 0.5-5.6 µg/kg after intravenous and 25.1 µg/kg after oral administration. In comparison with selective MOP receptor agonists, cebranopadol was more potent in models of chronic neuropathic than acute nociceptive pain. Cebranopadol's duration of action is long (up to 7 hours after intravenous 12 µg/kg; >9 hours after oral 55 µg/kg in the rat tail-flick test). The antihypersensitive activity of cebranopadol in the spinal nerve ligation model was partially reversed by pretreatment with the selective NOP receptor antagonist J-113397[1-[(3R,4R)-1-cyclooctylmethyl-3-hydroxymethyl-4-piperidyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one] or the opioid receptor antagonist naloxone, indicating that both NOP and opioid receptor agonism are involved in this activity. Development of analgesic tolerance in the chronic constriction injury model was clearly delayed compared with that from an equianalgesic dose of morphine (complete tolerance on day 26 versus day 11, respectively). Unlike morphine, cebranopadol did not disrupt motor coordination and respiration at doses within and exceeding the analgesic dose range. Cebranopadol, by its combination of agonism at NOP and opioid receptors, affords highly potent and efficacious analgesia in various pain models with a favorable side effect profile.


Subject(s)
Analgesics, Opioid/therapeutic use , Indoles/therapeutic use , Opioid Peptides/agonists , Pain/drug therapy , Receptors, Opioid/agonists , Spiro Compounds/therapeutic use , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Behavior, Animal/drug effects , Bone Neoplasms/complications , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , Female , Indoles/administration & dosage , Indoles/adverse effects , Indoles/pharmacology , Male , Pain/etiology , Pain/metabolism , Polyneuropathies/complications , Polyneuropathies/drug therapy , Polyneuropathies/metabolism , Protein Binding , Radioligand Assay , Rats , Rats, Sprague-Dawley , Rats, Wistar , Rotarod Performance Test , Spiro Compounds/administration & dosage , Spiro Compounds/adverse effects , Spiro Compounds/pharmacology , Nociceptin
5.
CNS Drugs ; 28(4): 319-29, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24578192

ABSTRACT

Tapentadol is a novel, centrally-acting analgesic drug, with an analgesic efficacy comparable to that of strong opioids such as oxycodone and morphine. Its high efficacy has been demonstrated in a range of animal models of acute and chronic, nociceptive, inflammatory, and neuropathic pain as well as in clinical studies with moderate to severe pain arising from a number of different etiologies. At the same time, a favorable gastrointestinal tolerability has been demonstrated in rodents and humans, and advantages over morphine regarding tolerance development and physical dependence were shown in animal studies. Furthermore, a low level of abuse and diversion is beginning to emerge from first post-marketing data. Tapentadol acts as a µ-opioid receptor (MOR) agonist and noradrenaline reuptake inhibitor (NRI). Both mechanisms of action have been shown to contribute to the analgesic activity of tapentadol and to produce analgesia in a synergistic manner, such that relatively moderate activity at the two target sites (MOR and noradrenaline reuptake transporter) is sufficient to produce strong analgesic effects. It has been suggested that tapentadol is the first representative of a proposed new class of analgesics, MOR-NRI. This review presents the evidence that has led to this suggestion, and outlines how the pharmacology of tapentadol can explain its broad analgesic activity profile and high analgesic potency as well as its favorable tolerability.


Subject(s)
Adrenergic Uptake Inhibitors/administration & dosage , Analgesia/methods , Pain/drug therapy , Phenols/administration & dosage , Receptors, Opioid, mu/agonists , Animals , Drug Therapy, Combination , Humans , Pain/diagnosis , Receptors, Opioid, mu/physiology , Tapentadol
6.
J Med Chem ; 55(19): 8392-408, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22957803

ABSTRACT

A series of N-(2-amino-6-trifluoromethylpyridin-3-ylmethyl)-2-(3-fluoro-4-methylsulfonylaminophenyl)propanamides were designed combining previously identified pharmacophoric elements and evaluated as hTRPV1 antagonists. The SAR analysis indicated that specific hydrophobic interactions of the 2-amino substituents in the C-region of the ligand were critical for high hTRPV1 binding potency. In particular, compound 49S was an excellent TRPV1 antagonist (K(i(CAP)) = 0.2 nM; IC(50(pH)) = 6.3 nM) and was thus approximately 100- and 20-fold more potent, respectively, than the parent compounds 2 and 3 for capsaicin antagonism. Furthermore, it demonstrated strong analgesic activity in the rat neuropathic model superior to 2 with almost no side effects. Compound 49S antagonized capsaicin induced hypothermia in mice but showed TRPV1-related hyperthermia. The basis for the high potency of 49S compared to 2 is suggested by docking analysis with our hTRPV1 homology model in which the 4-methylpiperidinyl group in the C-region of 49S made additional hydrophobic interactions with the hydrophobic region.


Subject(s)
Analgesics/chemical synthesis , Pyridines/chemical synthesis , Sulfonamides/chemical synthesis , TRPV Cation Channels/antagonists & inhibitors , Analgesics/chemistry , Analgesics/pharmacology , Animals , Body Temperature/drug effects , CHO Cells , Capsaicin/pharmacology , Cricetinae , Cricetulus , Dopamine/analogs & derivatives , Dopamine/pharmacology , Hot Temperature , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Neuralgia/drug therapy , Pyridines/chemistry , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , TRPV Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...