WO2009138734A2 - Use of opioid compounds in peripheral pain, wound healing and scar formation - Google Patents

Use of opioid compounds in peripheral pain, wound healing and scar formation Download PDF

Info

Publication number
WO2009138734A2
WO2009138734A2 PCT/GB2009/001184 GB2009001184W WO2009138734A2 WO 2009138734 A2 WO2009138734 A2 WO 2009138734A2 GB 2009001184 W GB2009001184 W GB 2009001184W WO 2009138734 A2 WO2009138734 A2 WO 2009138734A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound according
pain
wound
compound
chronic
Prior art date
Application number
PCT/GB2009/001184
Other languages
French (fr)
Other versions
WO2009138734A3 (en
Inventor
Andrew Douglas Baxter
Alan Rothaul
Original Assignee
Serentis Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0808754A external-priority patent/GB0808754D0/en
Priority claimed from GB0808753A external-priority patent/GB0808753D0/en
Priority claimed from GB0808755A external-priority patent/GB0808755D0/en
Application filed by Serentis Limited filed Critical Serentis Limited
Publication of WO2009138734A2 publication Critical patent/WO2009138734A2/en
Publication of WO2009138734A3 publication Critical patent/WO2009138734A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/10Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with radicals containing only carbon and hydrogen atoms attached to ring carbon atoms
    • C07D211/12Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with radicals containing only carbon and hydrogen atoms attached to ring carbon atoms with only hydrogen atoms attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/56Nitrogen atoms
    • C07D211/58Nitrogen atoms attached in position 4

Definitions

  • This invention relates to non-CNS-penetrant opioid compounds and to their use for the treatment of peripheral pain and, wound care, and to modify scar formation.
  • opioids act solely on the central nervous system, producing analgesia by interaction with cerebral and spinal opioid receptors. It is now understood that the number of opioid receptors in peripheral tissue is normally low but rapidly increases during inflammation (Hassan et a/., 1993; Schafer et al., 1995; Stein et al., 1996).
  • Immune cells are a major source of endogenous opioid peptides and it appears, by binding peripheral opioid receptors, they may constitute a mechanism of peripheral pain control (Stein et a/., 2003).
  • the present invention is based on the discovery that peripherally-acting analogues of opioids are capable of treating peripheral pain without CNS- mediated effects.
  • the peripherally-acting analogues can be used to boost analgesia when pain is associated with inflammation.
  • a partial agonist pharmacological profile confers both analgesic and antiinflammatory activity.
  • such compounds can be used, inter alia, in the treatment of pain conditions such as acute and chronic pain (as well as, but not limited to, pain associated with cancer, surgery, arthritis, dental surgery, trauma, musculoskeletal injury, such as osteoarthritis, back pain or repetitive strain injury or disease, visceral diseases) and migraine headache.
  • painful conditions to be treated can be neuropathic (post-herpetic neuralgia, diabetic neuropathy, drug induced neuropathy, HIV-mediated neuropathy, sympathetic reflex dystrophy or causalgia, fibromyalgia, myofacial pain, entrapment neuropathy, phantom limb pain, trigeminal neuralgia).
  • Neuropathic conditions include pain related to stroke, multiple sclerosis, spinal cord injury, arachnoiditis, neoplasms, syringomyelia, Parkinson's and epilepsia.
  • peripherally-acting analogues of opioids are capable accelerating the healing of both chronic and acute wounds.
  • These compounds are also capable of accelerating collagen deposition during scar formation and are therefore capable of increasing scar strength in acute wounds, surgical incisions and similar events and by doing so reducing scar formation.
  • they can be used to boost analgesia when such wounds are associated with pain.
  • Wounds typically associated with pain include chronic dermal wounds, diabetic ulcers, pressure wounds, ophthalmic injury, skin grafts at the graft and donor sites, abrasive wounds and burns.
  • opioids applied locally provide some pain relief, they are still susceptible to causing unwanted systemic and CNS side-effects. These side- effects can be managed by the peripherally-acting analogues of the invention as they do not penetrate the CNS.
  • a quaternary ammonium derivative of an opioid is used for wound healing, to modify scar formation or to treat peripheral pain or pain associated with an inflammatory condition, e.g. as described above.
  • a compound of or for use in the invention is an opioid which normally comprises a tertiary amine and which has been modified by introducing an alkyl group on that nitrogen atom.
  • opioids such as (but not limited to) fentanyl (1), tramadol (2), buprenorphine (3), methadone (4), morphine (5), oxycodone, hydrocodone, dihydrocodone, hydromorphone, oxymorphone, alfentanil, sufentanil, remifentanyl, dextropropxyphene, bezitramide, piritramide, pentazocine, phenazocine, nalbufine, levorphanol, loperamide and diphenoxylate, i.e. including compounds of the formulae
  • R1 is C 1 -C 4 alkyl
  • X is counterion of any pharmaceutically acceptable acid, including halide, sulphate, methosulphate, phosphate or of an organic acid such as citrate, succinate, tartarate, fumarate, maleate, acetate or methanesulphonate.
  • Analogues of opiates that act as partial agonists at ⁇ and K opioid receptors while demonstrating antagonist activity at ⁇ receptors may be preferred.
  • Compounds of the invention may have one or more chiral centres. Any reference herein to these compounds should be understood as a reference to any diastereomer, enantiomer or mixture thereof. Any enantiomer may be substantially free of others, e.g. in an enantiomeric excess of at least 80%, preferably at least 90% and more preferably at least 95%. Similarly, any mixture of diastereomers may be substantially free of the other. The form of each of formulae 1 to 5 drawn above is preferred.
  • the active agent may be administered by various routes, including parenteral, oral, intraoral, rectal, epicutaneous, transdermal, subcutaneous, intramuscular, intranasal, sublingual, buccal, vaginal, rectal, intradural, intraocular, intrarespiratory, or nasal inhalation and like forms.
  • the amount of active ingredient that is used can be chosen by the skilled person having regard to the usual factors.
  • the pharmaceutical composition containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • the composition may be in immediate or controlled release form.
  • the topical route may be preferred for compounds of formulae (1), (2) or
  • the active agent is typically formulated, e.g. with conventional diluents or carriers, or as a dressing or a patch, as a medicament adapted to be delivered by the chosen route.
  • Such formulations may be designed to retard the passage of the compound into the systemic circulation and are chosen according to such considerations as the potency of the drug, the severity of the condition and the route of administration.
  • the active agent may be administered into the incision/traumatic site in a suitable dermal or topical formulation.
  • the amount of active ingredient that is used can be chosen by the skilled person having regard to the usual factors.
  • the suture or incision closing device is impregnated with a compound of the invention.
  • the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing treatment. Optimum dose levels and frequency of dosing will be determined by clinical trials, as is required in the pharmaceutical art. In general, the daily dose range will lie within the range of from about 0.1 ⁇ g to 1000 mg per kg body weight of a mammal, often 0.01 mg to about 100 mg per kg, for example 0.1 ⁇ g to 10 mg per kg, in single or divided doses. It may be necessary to use dosages outside these limits in some cases.
  • a compound of the invention is preferably administered via the oral, parenteral, intranasal, inhaled, sub-lingual, buccal, topical or transdermal route.
  • a typical daily dose is less than 1000 mg, e.g. 0.1 to 100 mg.
  • Quaternary ammonium derivatives of opioids can be prepared by treatment of the opioid with R1-X.
  • the following reaction may be used:
  • Peripherally active compounds of formula (5) can be prepared according to the general scheme below.
  • X is typically a reactive halide such as chloride, bromide or iodide, or methosulphate.
  • X is typically a reactive halide such as chloride, bromide or iodide, or methosulphate.
  • the transformation may be carried out using methods known to those skilled in the art.
  • Such another drug may be a local anaesthetic, anti-septic or antibiotic.
  • another drug may be a local anaesthetic, anti-septic or antibiotic.
  • Other combinations include those with an anti-convulsant, such as phenytoin, or a compound used to enhance vascular permeability, including peripheral vasodilators such as adenosine agonists or re-uptake inhibitors (e.g. dipyridamole), anti-platelets agents (e.g. ketanserin) and pentoxyfylline, and vasopeptidase inhibitors such as neutral endopeptidase (NEP) inhibitors or combined ACE/NEP inhibitors.
  • an anti-convulsant such as phenytoin
  • a compound used to enhance vascular permeability including peripheral vasodilators such as adenosine agonists or re-uptake inhibitors (e.g. dipyridamole), anti-platelets agents (e.g
  • Such another drug may be selected from corticosteroids (examples including Cortisol, cortisone, hydrocortisone, dihydrocortisone, fludrocortisone, prednisone, prednisolone, deflazacort, flunisolide, beconase, methylprednisolone, triamcinolone, betamethasone, and dexamethasone), disease modifying anti-rheumatic drugs (DMARDs) (examples including azulfidine, aurothiomalate, bucillamine, chlorambucil, cyclophosphamide, leflunomide, methotrexate, mizoribine, penicillamine and sulphasalazine), immunosuppressants (examples including azathioprine, cyclosporine and mycophenolate), COX inhibitors (examples including Cortisol, cortisone, hydrocortisone, dihydro
  • lodomethane (2 mL, 32.3 mmol) was added to a solution of methadone (0.20 g, 0.65 mmol) in ethyl acetate (10 mL). The mixture was stirred at room temperature for 4 h. The resulting precipitate was filtered and washed with a small amount of ethyl acetate. After drying under vacuum the product was obtained as white powder (0.19 g, 66%).
  • lodomethane (5 ml_, 80.2 mmol) was added to a solution of fentanyl (0.50 g, 1.49 mmol) in ethyl acetate (50 ml_). The resulting slurry was stirred at room temperature overnight an then filtered. The crude product was washed with hot ethyl acetate twice and dried under vacuum to obtain product as white powder (0.29 g, 41%).
  • lodomethane (3.5 ml_, 56.1 mmol) was added to slurry of morphine (0.32 g, 1.1 mmol) in acetonitrile (16 mL). The mixture was stirred at room temperature overnight and filtered. The crude product was purified by trituration with acetonitrile followed by trituration with ethyl acetate. After drying under vacuum the product was obtained as white powder (0.20 g, 42%).
  • Buprenorphine (5 g, 10.7 mmol) and MeI (34.2 g, 15 ml, 240 mmol) were taken up in MIBK (30 ml), halved between two sealed tubes and heated at 70 0 C for six days.
  • Buprenorphine and buprenorphine methiodide were assayed for binding to the human opiate receptors in radiolabel binding studies. The results are shown in Table 1. Buprenorphine methiodide showed significant inhibition in all three of the opiate receptor binding assays. Table 1 : Opiate receptor binding potency
  • nt not tested Buprenorphine methiodide also demonstrated partial agonism in an isolated organ bath experiment (guinea pig ileum twitch response, 50% of control at 10 ⁇ M).
  • buprenorphine is a partial ⁇ receptor agonist which also shows partial agonism for K receptors, but is a potent antagonist at ⁇ receptors.
  • the data demonstrate that buprenorphine methiodide has retained the opiate receptor profile of buprenorphine at ⁇ and K opiate receptors.
  • Buprenorphine methiodide and methyl-naloxone were co-administered each at 1 mg/kg i.v. via the tail vein.
  • Plasma and brains were prepared from 3 rats at each of the pre-dose and 30, 60, 120 and 180 minutes post dose. Samples were extracted by protein precipitation and analysed by HPLC-MS/MS for buprenorpine methiodide, and methyl-naloxone. Results are shown in Table 4.
  • Plasma cone ng 3 // ⁇ ml
  • Brain cone ng/mL
  • Time(Hrs) BupMel Methyl- BupMel Methyl- io NNaloxone Naloxone
  • Example 7 Activity in an inflammatory pain model
  • the analgesic effects of buprenorpine methiodide were evaluated in the Acute Inflammatory Pain Test (carrageenan) in the rat.
  • the method which detects analgesic/anti-inflammatory activity in rats with inflammatory pain, follows that described by Winter et a/ (Proc. Soc. Exp. Biol. Med., 111 , 544-547, 1962). Rats were injected with a suspension of carrageenan into the lower surface of the right hindpaw. Two hours later, rats were submitted consecutively to tactile and thermal stimulation of both hindpaws. Buprenorpine methiodide was evaluated at 3 doses, administered as an i.v.
  • buprenorpine methiodide significantly reversed the hyperalgesic response associated with peripheral inflammation in a dose-dependent manner.

Abstract

A compound which is a quaternary ammonium salt derivative of an opioid, for use in therapy.

Description

USE OF OPIOID COMPOUNDS IN PERIPHERAL PAIN, WOUND HEALING
AND SCAR FORMATION
Field of the Invention
This invention relates to non-CNS-penetrant opioid compounds and to their use for the treatment of peripheral pain and, wound care, and to modify scar formation. Background of the Invention
It had long been thought that opioids act solely on the central nervous system, producing analgesia by interaction with cerebral and spinal opioid receptors. It is now understood that the number of opioid receptors in peripheral tissue is normally low but rapidly increases during inflammation (Hassan et a/., 1993; Schafer et al., 1995; Stein et al., 1996).
Immune cells are a major source of endogenous opioid peptides and it appears, by binding peripheral opioid receptors, they may constitute a mechanism of peripheral pain control (Stein et a/., 2003).
Local delivery of opioids has been employed in pain therapy to minimize the CNS side-effects traditionally associated with systemic use. Injection of opioids into knee joints after knee surgery (Stein et a/., 1991) or local delivery after dental surgery (Likar et a/., 1998) caused prolonged post-operative analgesia in humans.
Summary of the Invention
The present invention is based on the discovery that peripherally-acting analogues of opioids are capable of treating peripheral pain without CNS- mediated effects. In addition, the peripherally-acting analogues can be used to boost analgesia when pain is associated with inflammation. Surprisingly, a partial agonist pharmacological profile confers both analgesic and antiinflammatory activity. Accordingly, such compounds can be used, inter alia, in the treatment of pain conditions such as acute and chronic pain (as well as, but not limited to, pain associated with cancer, surgery, arthritis, dental surgery, trauma, musculoskeletal injury, such as osteoarthritis, back pain or repetitive strain injury or disease, visceral diseases) and migraine headache. Additionally, painful conditions to be treated can be neuropathic (post-herpetic neuralgia, diabetic neuropathy, drug induced neuropathy, HIV-mediated neuropathy, sympathetic reflex dystrophy or causalgia, fibromyalgia, myofacial pain, entrapment neuropathy, phantom limb pain, trigeminal neuralgia). Neuropathic conditions include pain related to stroke, multiple sclerosis, spinal cord injury, arachnoiditis, neoplasms, syringomyelia, Parkinson's and epilepsia. In addition, peripherally-acting analogues of opioids are capable accelerating the healing of both chronic and acute wounds. These compounds are also capable of accelerating collagen deposition during scar formation and are therefore capable of increasing scar strength in acute wounds, surgical incisions and similar events and by doing so reducing scar formation. In addition, they can be used to boost analgesia when such wounds are associated with pain. Wounds typically associated with pain include chronic dermal wounds, diabetic ulcers, pressure wounds, ophthalmic injury, skin grafts at the graft and donor sites, abrasive wounds and burns.
While opioids applied locally provide some pain relief, they are still susceptible to causing unwanted systemic and CNS side-effects. These side- effects can be managed by the peripherally-acting analogues of the invention as they do not penetrate the CNS.
According to the invention, a quaternary ammonium derivative of an opioid is used for wound healing, to modify scar formation or to treat peripheral pain or pain associated with an inflammatory condition, e.g. as described above.
A compound of or for use in the invention is an opioid which normally comprises a tertiary amine and which has been modified by introducing an alkyl group on that nitrogen atom. Such compounds are derivatives of opioids such as (but not limited to) fentanyl (1), tramadol (2), buprenorphine (3), methadone (4), morphine (5), oxycodone, hydrocodone, dihydrocodone, hydromorphone, oxymorphone, alfentanil, sufentanil, remifentanyl, dextropropxyphene, bezitramide, piritramide, pentazocine, phenazocine, nalbufine, levorphanol, loperamide and diphenoxylate, i.e. including compounds of the formulae
Figure imgf000004_0001
(D (2)
Figure imgf000004_0002
(3)
Figure imgf000004_0003
(4) (5)
wherein R1 is C1-C4 alkyl; and X is counterion of any pharmaceutically acceptable acid, including halide, sulphate, methosulphate, phosphate or of an organic acid such as citrate, succinate, tartarate, fumarate, maleate, acetate or methanesulphonate.
Most of these compounds are new. Those that are known have never been used in therapy or, more particularly, for the treatment of peripheral pain or pain associated with an inflammatory condition, wound healing or to modify scar formation.
Analogues of opiates that act as partial agonists at μ and K opioid receptors while demonstrating antagonist activity at δ receptors may be preferred.
Description of the Invention
Compounds of the invention may have one or more chiral centres. Any reference herein to these compounds should be understood as a reference to any diastereomer, enantiomer or mixture thereof. Any enantiomer may be substantially free of others, e.g. in an enantiomeric excess of at least 80%, preferably at least 90% and more preferably at least 95%. Similarly, any mixture of diastereomers may be substantially free of the other. The form of each of formulae 1 to 5 drawn above is preferred.
The active agent may be administered by various routes, including parenteral, oral, intraoral, rectal, epicutaneous, transdermal, subcutaneous, intramuscular, intranasal, sublingual, buccal, vaginal, rectal, intradural, intraocular, intrarespiratory, or nasal inhalation and like forms. The amount of active ingredient that is used can be chosen by the skilled person having regard to the usual factors. The pharmaceutical composition containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. The composition may be in immediate or controlled release form. The topical route may be preferred for compounds of formulae (1), (2) or
(3). For use through this route of delivery the active agent is typically formulated, e.g. with conventional diluents or carriers, or as a dressing or a patch, as a medicament adapted to be delivered by the chosen route. Such formulations may be designed to retard the passage of the compound into the systemic circulation and are chosen according to such considerations as the potency of the drug, the severity of the condition and the route of administration. The active agent may be administered into the incision/traumatic site in a suitable dermal or topical formulation. The amount of active ingredient that is used can be chosen by the skilled person having regard to the usual factors. In another aspect of the invention, the suture or incision closing device is impregnated with a compound of the invention.
It will be understood that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing treatment. Optimum dose levels and frequency of dosing will be determined by clinical trials, as is required in the pharmaceutical art. In general, the daily dose range will lie within the range of from about 0.1 μg to 1000 mg per kg body weight of a mammal, often 0.01 mg to about 100 mg per kg, for example 0.1 μg to 10 mg per kg, in single or divided doses. It may be necessary to use dosages outside these limits in some cases. A compound of the invention is preferably administered via the oral, parenteral, intranasal, inhaled, sub-lingual, buccal, topical or transdermal route. In this context, a typical daily dose is less than 1000 mg, e.g. 0.1 to 100 mg.
Quaternary ammonium derivatives of opioids can be prepared by treatment of the opioid with R1-X. For example, the following reaction may be used:
Figure imgf000006_0001
Peripherally active compounds of formula (5) can be prepared according to the general scheme below.
Figure imgf000007_0001
wherein X is typically a reactive halide such as chloride, bromide or iodide, or methosulphate. The transformation may be carried out using methods known to those skilled in the art.
It will often be advantageous to use the compounds of the invention in combination with another drug used for pain, inflammation or wound care therapy. Such another drug may be a local anaesthetic, anti-septic or antibiotic. Other combinations include those with an anti-convulsant, such as phenytoin, or a compound used to enhance vascular permeability, including peripheral vasodilators such as adenosine agonists or re-uptake inhibitors (e.g. dipyridamole), anti-platelets agents (e.g. ketanserin) and pentoxyfylline, and vasopeptidase inhibitors such as neutral endopeptidase (NEP) inhibitors or combined ACE/NEP inhibitors. It will also be often advantageous to use the compounds of the invention in combination with another drug used for pain or inflammation. Such another drug may be selected from corticosteroids (examples including Cortisol, cortisone, hydrocortisone, dihydrocortisone, fludrocortisone, prednisone, prednisolone, deflazacort, flunisolide, beconase, methylprednisolone, triamcinolone, betamethasone, and dexamethasone), disease modifying anti-rheumatic drugs (DMARDs) (examples including azulfidine, aurothiomalate, bucillamine, chlorambucil, cyclophosphamide, leflunomide, methotrexate, mizoribine, penicillamine and sulphasalazine), immunosuppressants (examples including azathioprine, cyclosporine and mycophenolate), COX inhibitors (examples including aceclofenac, acemetacin, alcofenac, alminoprofen, aloxipirin, amfenac, aminophenazone, antraphenine, aspirin, azapropazone, benorilate, benoxaprofen, benzydamine, butibufen, celecoxib, chlorthenoxacine, choline salicylate, chlometacin, dexketoprofen, diclofenac, diflunisal, emorfazone, epirizole, etodolac, feclobuzone, felbinac, fenbufen, fenclofenac, flurbiprofen, glafenine, hydroxylethyl salicylate, ibuprofen, indometacin, indoprofen, ketoprofen, ketorolac, lactyl phenetidin, loxoprofen, mefenamic acid, metamizole, mofebutazone, mofezolac, nabumetone, naproxen, nifenazone, oxametacin, phenacetin, pipebuzone, pranoprofen, propyphenazone, proquazone, rofecoxib, salicylamide, salsalate, suiindac, suprofen, tiaramide, tinoridine, tolfenamic acid and zomepirac), neutralising antibodies (examples including etanercept and infliximab), antibiotics (examples including doxycycline and minocycline), analgesics (opiate or non opiate such as baclofen), neuropathic pain agents (such as gabapentin or pregabalin), non steroidal anti-inflammatory drugs, NMDA antagonists, neuroleptics, anticonvulsants, anti-spasmodics, anti-depressants or muscle relaxants. The following Examples illustrate the invention.
Example 1 Methadone methiodide
Figure imgf000008_0001
lodomethane (2 mL, 32.3 mmol) was added to a solution of methadone (0.20 g, 0.65 mmol) in ethyl acetate (10 mL). The mixture was stirred at room temperature for 4 h. The resulting precipitate was filtered and washed with a small amount of ethyl acetate. After drying under vacuum the product was obtained as white powder (0.19 g, 66%).
1H NMR (δ, 200MHz, d6-DMSO): 0.41 (d, 3H, CH3), 0.69 (t, 3H, CH3), 2.21-2.33 (m, 4H), 3.10-3.31 (m, 1 H, CH2), 3.0 (s, 9H, CH3), 7.20-7.57 (m, 10H, aromatic). m/z 324=451 (MT842) - 127 (I"). Example 2 Fentanyl methiodide
Figure imgf000009_0001
lodomethane (5 ml_, 80.2 mmol) was added to a solution of fentanyl (0.50 g, 1.49 mmol) in ethyl acetate (50 ml_). The resulting slurry was stirred at room temperature overnight an then filtered. The crude product was washed with hot ethyl acetate twice and dried under vacuum to obtain product as white powder (0.29 g, 41%).
1H NMR (δ, 200MHz, d6-DMSO): 0.88 (t, 3H, CH3), 1.78-1.99 (m, 5H), 2.89 (S, 3H, CH3), 3.33 (S, 3H, CH3), 3.50-3.55 (bm, 6H), 4.58 (bm, 1H), 7.26-7.34 (m, 7H), 7.48-7.25 (m, 3H). m/z 351=478 (MT842) - 127 (I"). Example 3 Morphine methiodide
Figure imgf000009_0002
lodomethane (3.5 ml_, 56.1 mmol) was added to slurry of morphine (0.32 g, 1.1 mmol) in acetonitrile (16 mL). The mixture was stirred at room temperature overnight and filtered. The crude product was purified by trituration with acetonitrile followed by trituration with ethyl acetate. After drying under vacuum the product was obtained as white powder (0.20 g, 42%). 1H NMR (δ, 200MHz, d6-DMSO): 1.78-1.85 (bd, 1 H), 2.70-2.81 (m, 2H), 3.10- 3.40 (m, 10H), 4.14 (bs, 1 H), 4.36 (bs, 1H), 4.79 (d, 1 H), 5.05 (d,1 H), 5.25 (d, 1H)1 5.60 (bd, 1H), 6.41-6.56 (m, 2H), 9.01 (s, 1H). m/z 300=427 (MT842) - 127 (V). Example 4 Buprenorphine Methiodide
Figure imgf000010_0001
Buprenorphine (5 g, 10.7 mmol) and MeI (34.2 g, 15 ml, 240 mmol) were taken up in MIBK (30 ml), halved between two sealed tubes and heated at 700C for six days.
The tubes were allowed to cool to RT, combined and evaporated. The residue was triturated with 1PrOAc, and the solid formed was filtered off and dried. The solid was purified by column chromatography (2%-5% MeOH in CH2CI2). The product obtained was triturated with MTBE and the solid formed was filtered off and dried in vacuo to give 2 g (31% yield) of the title product as a pale yellow solid.
1H NMR (δ, 400MHz, d4-MeOH): 0.50-0.58 (m, 1H), 0.61-0.68 (m, 1 H), 0.80-0.95 (m, 2H), 1.025 (s, 9H, 'butyl CH3), 1.05-1.18 (m, 1H), 1.19-1.3 (m,2H), 1.40 (s, 3H, CH3), 1.48-1.68 (m, 2H), 1.85-1.97 (bd, 2H), 2.1 (dd, 1 H), 2.45 (dd, 1 H), 2.68-2.80 (m, 2H), 3.05 (dd, 1H), 3.15 (dd, 1H), 3.30 (m, 1H), 3.50 (s, 3H, CH3O), 3.56 (m, 1H), 3.59 (s, 3H, CH3N), 4.08 (dd, 1H), 4.18 (d, 1H), 4.65 (s, 1 H, CH2CHO), 6.65 (d, 1 H, aromatic), 6.74 (d, 1H, aromatic). Example 5 Opiate receptor pharmacology
Buprenorphine and buprenorphine methiodide were assayed for binding to the human opiate receptors in radiolabel binding studies. The results are shown in Table 1. Buprenorphine methiodide showed significant inhibition in all three of the opiate receptor binding assays. Table 1 : Opiate receptor binding potency
Figure imgf000011_0001
Both compounds were assayed for opiate receptor agonism and antagonism activity using GTPγS binding assays against human cloned μ, δ and K receptors. The methiodide analogues of methadone, morphine and fentanyl were screened for agonist and antagonist activity against μ opiate receptors in a guinea pig ileum organ bath assay. The results are shown in Tables 2 & 3.
Table 2: Opiate receptor agonist activity
Figure imgf000011_0002
nt = not tested Buprenorphine methiodide also demonstrated partial agonism in an isolated organ bath experiment (guinea pig ileum twitch response, 50% of control at 10 μM).
The results confirm that buprenorphine is a partial μ receptor agonist which also shows partial agonism for K receptors, but is a potent antagonist at δ receptors. The data demonstrate that buprenorphine methiodide has retained the opiate receptor profile of buprenorphine at μ and K opiate receptors.
The results confirm that Methadone methiodide, Morphine Methiodide and Fentanyl Methiodide behave as μ-receptor agonists. Example 6
Restriction to peripheral sites
To determine if the activity of buprenorphine methiodide is restricted to the periphery, a pharmacokinetic study was performed and the CNS penetration determined following i.v. dosing. Brains and plasma were sampled from rats that had been co-administered i.v. with buprenorphine methiodide and methyl- naloxone. Methyl-naloxone (Sigma-Aldrich N 129) was used as a control compound, to check for blood contamination during the brain collection process. Methyl-naloxone is a compound known not to cross the blood-brain barrier.
Buprenorphine methiodide and methyl-naloxone were co-administered each at 1 mg/kg i.v. via the tail vein. Plasma and brains were prepared from 3 rats at each of the pre-dose and 30, 60, 120 and 180 minutes post dose. Samples were extracted by protein precipitation and analysed by HPLC-MS/MS for buprenorpine methiodide, and methyl-naloxone. Results are shown in Table 4.
Table 4
Plasma cone (ng 3//πml) Brain cone (ng/mL) tat Time(Hrs) BupMel Methyl- BupMel Methyl- io NNaloxone Naloxone
1 P re BLQ BLQ BLQ BLQ
2 Pre BLQ BLQ BLQ BLQ
3 Pre BLQ BLQ BLQ BLQ
4 0.5 78.6 46.7 BLQ BLQ
5 0.5 84.4 52.7 BLQ BLQ
6 0.5 70.8 55.3 BLQ BLQ
7 1 18.7 32.3 BLQ BLQ
8 1 22.1 29.2 BLQ BLQ
9 1 27.5 32.9 BLQ BLQ
10 2 17.4 19.2 BLQ BLQ
11 2 7.13 13.3 BLQ BLQ
12 2 4.51 15.5 BLQ BLQ
13 3 3.60 13.2 BLQ BLQ
14 3 2.98 8.68 BLQ BLQ
15 3 BLQ 6.17 BLQ BLQ
BLQ= below limit of quantitation
The results show that buprenorpine methiodide was not found in brain tissue and does not penetrate the CNS. Example 7 Activity in an inflammatory pain model
The analgesic effects of buprenorpine methiodide were evaluated in the Acute Inflammatory Pain Test (carrageenan) in the rat. The method, which detects analgesic/anti-inflammatory activity in rats with inflammatory pain, follows that described by Winter et a/ (Proc. Soc. Exp. Biol. Med., 111 , 544-547, 1962). Rats were injected with a suspension of carrageenan into the lower surface of the right hindpaw. Two hours later, rats were submitted consecutively to tactile and thermal stimulation of both hindpaws. Buprenorpine methiodide was evaluated at 3 doses, administered as an i.v. bolus 15 minutes before testing, and compared with a vehicle control group and morphine (4 mg/kg i.v.) used as reference substance. In this model, buprenorpine methiodide significantly reversed the hyperalgesic response associated with peripheral inflammation in a dose- dependent manner.

Claims

1. A compound which is a quaternary ammonium salt derivative of an opioid, for use in therapy.
2. A compound according to claim 1 , wherein the opioid is selected from fentanyl, tramadol, buprenorphine, methadone, morphine, oxycodone, hydrocodone, dihydrocodone, hydromorphone, oxymorphone, alfentanil, sufentanil, remifentanyl, dextropropxyphene, bezitramide, piritramide, pentazocine, phenazocine, nalbufine, levorphanol, loperamide and diphenoxylate.
3. A compound of formula (1)
Figure imgf000015_0001
(D wherein R1 is CrC4 alkyl and X is the counterion of a pharmaceutically acceptable acid.
4. A compound of formula (2)
Figure imgf000015_0002
(2)
wherein R1 and X are as defined in claim 3.
5. A compound of formula (3)
Figure imgf000016_0001
(3) wherein R1 and X are as defined in claim 3.
6. A compound of formula (4)
Figure imgf000016_0002
wherein R1 and X are as defined in claim 3.
7. A compound of formula (5)
Figure imgf000016_0003
(5) wherein R1 and X are as defined in claim 3.
8. A compound according to any preceding claim, which has a counterion of the quaternary or which is sulphate, methosulphate or phosphate or of an organic acid such as citrate, succinate, tartarate, fumarate, maleate, acetate or methanesulphonate.
9. A compound according to any preceding claim, wherein the salt or X is a halide.
10. A compound according to any preceding claim e.g. as defined in claim 7, wherein the treatment is of a pain condition.
11. A compound according to claim 10, for the treatment of peripheral pain.
12. A compound according to claim 11 , wherein the pain is inflammatory pain.
13. A compound according to claim 10, for the treatment of chronic pain.
14. A compound according to claim 13, wherein the pain is back pain, malignant pain, chronic headache (including migraine and cluster headaches) or arthritic pain.
15. A compound according to claim 10, for the treatment of acute pain.
16. A compound according to claim 15, wherein the pain is post-operative pain, post-traumatic pain or acute disease-induced pain.
17. A compound according to claims 10, for the treatment of neuropathic pain.
18. A compound according to any of claims 1 to 9, for use in wound care.
19. A compound according to claim 18, for use in scar formation and increase of scar strength in a wound.
20. A compound according to claim 18, wherein the wound is a chronic dermal wound.
21. A compound according to claim 18, wherein the wound is a diabetic ulcer.
22. A compound according to claim 18, wherein the wound is caused by a skin graft.
23. A compound according to claim 18, wherein the wound is a chronic dermal wound on a graft or donor site.
24. A compound according to claim 18, wherein the wound is a burn.
25. A compound according to claim 18, wherein the wound is a chronic dermal wound or an acute or chronic abrasive injury.
26. A compound according to claim 18, wherein the wound is a surgical incision.
27. A compound according to claim 18, wherein the wound is a traumatic incision.
28. A compound according to claim 18, wherein the wound is laceration.
29. A compound according to claim 18, wherein the wound is caused by acne, chicken pox or a dermatological condition.
30. A compound according to any of claims 18 to 29, for administration via direct installation into the wound.
31. A compound according to any of claims 18 to 29, for administration via direct installation into the dermis of the skin.
32. A compound according to any of claims 18 to 29, used in combination with another agent capable of facilitating or accelerating wound healing.
33. A compound according to any of claims 18 to 29, used in combination with an anti-convulsant, a peripheral vasodilator, a vasopeptidase inhibitor or an anti-platelets agent.
34. A compound according to any of claims 18 to 29, used in combination with an antiseptic or an antibiotic agent.
35. A compound according to any preceding claim, used in combination with an analgesic, a local anesthetic or an anti-inflammatory agent.
36. A compound according to any preceding claim, for administration via a topical or percutaneous route.
37. A compound according to any of claims 1 to 35, for administration via the oral route.
PCT/GB2009/001184 2008-05-14 2009-05-11 Use of opioid compounds in peripheral pain, wound healing and scar formation WO2009138734A2 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GB0808753.8 2008-05-14
GB0808754.6 2008-05-14
GB0808754A GB0808754D0 (en) 2008-05-14 2008-05-14 Use of opiod compounds in peripheral pain
GB0808755.3 2008-05-14
GB0808753A GB0808753D0 (en) 2008-05-14 2008-05-14 Use of opioid compounds in wound healing and scar formation
GB0808755A GB0808755D0 (en) 2008-05-14 2008-05-14 Use of opioid compounds in peripheral pain

Publications (2)

Publication Number Publication Date
WO2009138734A2 true WO2009138734A2 (en) 2009-11-19
WO2009138734A3 WO2009138734A3 (en) 2010-01-07

Family

ID=40825230

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2009/001184 WO2009138734A2 (en) 2008-05-14 2009-05-11 Use of opioid compounds in peripheral pain, wound healing and scar formation

Country Status (1)

Country Link
WO (1) WO2009138734A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013084060A1 (en) * 2011-12-08 2013-06-13 Purdue Pharma L.P. Quaternized buprenorphine analogs
US20150352100A1 (en) * 2013-01-21 2015-12-10 Avant Derma Pte. Ltd. Use of selective delta-opioid receptor antagonists and specific sensory receptor ligands
WO2017070017A1 (en) * 2015-10-23 2017-04-27 Rush University Medical Center Topical compositions providing pain relief and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0897726A1 (en) * 1996-11-25 1999-02-24 Toray Industries, Inc. Antipruritic agent

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0897726A1 (en) * 1996-11-25 1999-02-24 Toray Industries, Inc. Antipruritic agent

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013084060A1 (en) * 2011-12-08 2013-06-13 Purdue Pharma L.P. Quaternized buprenorphine analogs
JP2015502952A (en) * 2011-12-08 2015-01-29 パーデュー、ファーマ、リミテッド、パートナーシップ Quaternized buprenorphine analogues
US9096606B2 (en) 2011-12-08 2015-08-04 Purdue Pharma, L.P. Quarternized buprenorphine analogs
US20150352100A1 (en) * 2013-01-21 2015-12-10 Avant Derma Pte. Ltd. Use of selective delta-opioid receptor antagonists and specific sensory receptor ligands
WO2017070017A1 (en) * 2015-10-23 2017-04-27 Rush University Medical Center Topical compositions providing pain relief and methods of use thereof

Also Published As

Publication number Publication date
WO2009138734A3 (en) 2010-01-07

Similar Documents

Publication Publication Date Title
AU784541B2 (en) Novel methods and compositions involving opioids and antagonists thereof
US6451806B2 (en) Methods and compositions involving opioids and antagonists thereof
AU738743B2 (en) Antipruritic
JP5460771B2 (en) A novel method for the treatment and prevention of ileus
US20050159453A1 (en) Novel methods for the treatment and prevention of ileus
JP5860895B2 (en) 5-Methyl-1- (naphthalen-2-yl) -1H-pyrazole derivative
JP2006502190A (en) Opiate analogs selective for delta-opioid receptors
WO2010103312A1 (en) Hydroxymorpholiness and their use for the treatment of inflammatory disorders and pain
WO2009138734A2 (en) Use of opioid compounds in peripheral pain, wound healing and scar formation
EA006507B1 (en) Novel substituted 4-phenyl-4-[1h-imidazol-2-yl]-piperidine derivatives and their use as selective non-peptide delta opioid agonists
US20100076068A1 (en) Aminoalcohol Derivatives and Their Therapeutic Use
EP2475361B1 (en) N-substituted benzenepropanamides for use in the treatment of pain and inflammation
US20100016357A1 (en) Use of Beta-Aminoalcohols for the Treatment of Inflammatory Disorders and Pain
EP3860582B1 (en) Compounds for use in the treatment or prophylaxis of pain, inflammation and/or autoimmunity
US20120219592A1 (en) Use of B-Aminoalcohols in the Treatment of Inflammatory Disorders and Pain
AU2018215466B2 (en) Small molecule antagonists of SUMO related modification of CRMP2 and uses thereof
JP3681770B2 (en) Treatment for senile dementia or Alzheimer's disease
US20080306162A1 (en) Treatment of Inflammatory Disorders and Pain
WO2010013044A1 (en) Nalmefene and derivatives thereof for the treatment of skin disorders
WO1996008468A1 (en) Carbamoyloxy amine compounds

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09746051

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 31.05.11)

122 Ep: pct application non-entry in european phase

Ref document number: 09746051

Country of ref document: EP

Kind code of ref document: A2