US20110105551A1 - Analgesics for nasal administration - Google Patents

Analgesics for nasal administration Download PDF

Info

Publication number
US20110105551A1
US20110105551A1 US12/929,279 US92927911A US2011105551A1 US 20110105551 A1 US20110105551 A1 US 20110105551A1 US 92927911 A US92927911 A US 92927911A US 2011105551 A1 US2011105551 A1 US 2011105551A1
Authority
US
United States
Prior art keywords
buprenorphine
solution
solution according
analgesic
pectin
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/929,279
Inventor
Phillip John Birch
Ann Gail Hayes
Peter James Watts
Jonathan David Castile
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kyowa Kirin Services Ltd
Original Assignee
Vernalis R&D Ltd
Archimedes Development Ltd
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 GB0206448A external-priority patent/GB0206448D0/en
Priority claimed from GB0225042A external-priority patent/GB0225042D0/en
Priority claimed from GB0225040A external-priority patent/GB0225040D0/en
Priority claimed from GB0225041A external-priority patent/GB0225041D0/en
Application filed by Vernalis R&D Ltd, Archimedes Development Ltd filed Critical Vernalis R&D Ltd
Priority to US12/929,279 priority Critical patent/US20110105551A1/en
Publication of US20110105551A1 publication Critical patent/US20110105551A1/en
Assigned to ARCHIMEDES DEVELOPMENT LIMITED reassignment ARCHIMEDES DEVELOPMENT LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VERNALIS (R&D) LIMITED
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/5415Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof

Definitions

  • the invention relates to analgesic compositions and their use.
  • analgesics A wide variety of compounds can act as analgesics. Two important classes of analgesics are opioid analgesics and non-steroidal anti-inflammatory drugs (NSAIDs).
  • opioid analgesics opioid analgesics and non-steroidal anti-inflammatory drugs (NSAIDs).
  • NSAIDs non-steroidal anti-inflammatory drugs
  • Opioid analgesics exhibit morphine-like properties. Opioids can be sub-classified on the basis of their receptor specificity. Mu-agonist opioids provide intense analgesia. These opioids can be long-acting (e.g. methadone) or short-acting (e.g. remifentanil). Mixed agonist/antagonist opioids (e.g. butorphanol and buprenorphine) are partial agonists (the former at mu and kappa receptors and the latter at the mu receptor) and can produce good quality analgesia. They produce less respiratory depression and constipation than high efficacy mu agonists.
  • Mu-agonist opioids provide intense analgesia. These opioids can be long-acting (e.g. methadone) or short-acting (e.g. remifentanil).
  • Mixed agonist/antagonist opioids e.g. butorphanol and buprenorphine
  • are partial agonists the former at
  • opioids are associated with a number of undesirable side-effects, including respiratory depression, nausea, vomiting, dizziness, mental clouding, dysphoria, pruritus, constipation, increased biliary tract pressure, urinary retention and hypotension.
  • side-effects including respiratory depression, nausea, vomiting, dizziness, mental clouding, dysphoria, pruritus, constipation, increased biliary tract pressure, urinary retention and hypotension.
  • Buprenorphine is unusual in exhibiting a low maximum effect for respiratory depression and also a bell-shaped dose response curve where the effect first increases with larger doses, reaches a ceiling and then diminishes as the dosage is further increased, which makes it a safer drug than morphine, where respiratory depression will ultimately lead to death.
  • Buprenorphine has also been shown to have a lower incidence of other side-effects like constipation in man, and it has a lower abuse potential than full mu agonists.
  • NSAIDs have anti-inflammatory action and are effective on pain associated with the release of prostaglandins (PG) and other mediators of inflammation. They act by blocking the action of cyclooxygenase (COX), which converts arachidonic acid to eicosanoids.
  • COX cyclooxygenase
  • the eicosanoids include the prostanoids, prostacyclin (PGI 2 ), PGE 2 and the thromboxanes.
  • COX enzymes a constitutively-expressed COX-1 responsible for producing homeostatic prostaglandin and thromboxane mediators and an inducible COX-2 that is produced in large quantities in response to stimuli such as infection and inflammation.
  • NSAIDs Since the prostaglandins and thromboxanes mediate a number of homeostatic and protective mechanisms, toxic side effects often arise from the use of NSAIDs as a result of disruption of these mechanisms. These include clotting disorders (leading to prolonged bleeding times) and gastric irritation (including ulceration). NSAIDs may also cause salt and water retention and may therefore exacerbate hypertension. They may also be teratogenic at high doses during pregnancy.
  • the NSAIDs are associated with a number of adverse effects on the kidneys, although most are rare.
  • the kidney produces PGI 2 , PGE 2 and some PGF 2 ⁇ . These are involved in local modulation of renal blood flow, glomerular filtration rate, renin release, the concentrating mechanism for urine and the excretion of sodium and potassium.
  • the unwanted effects of NSAIDs result from the decrease in production of the prostaglandins and are summarized below:
  • pain relief should follow immediately upon administration of an analgesic.
  • the relief should be maintained for an extended period that is at least long enough to permit normal unbroken sleep patterns and avoid complicated dosage regimes.
  • intravenous injection is generally associated with rapid offset of pain relief as the circulating analgesic is cleared from the plasma. Prolonged analgesia requires multiple injections which is inconvenient and expensive. Intravenous injection is also usually associated with relatively high C max values, which can trigger (or amplify) any side effects associated with the analgesic.
  • transdermal patches can cause skin irritation and the drug dosage is difficult to control.
  • Sublingual tablets have an unpleasant taste and must be maintained in the mouth for relatively long periods of time (often 30 minutes or more), leading to compliance problems.
  • Buprenorphine has previously been administered via the intravenous, intramuscular and sublingual routes to human subjects. There are limited reports of nasal administration. Eriksen et al, J. Pharm. Pharmacol. 41, 803-805, 1989 report administration to human volunteers of a nasal spray. The spray consisted of 2 mg/ml of buprenorphine hydrochloride dissolved in 5% dextrose and the pH of the solution was adjusted to pH 5.
  • WO 90/09870 describes a composition for administration to mucosa comprising a pharmacologically active compound and a polycationic substance such as DEAE-dextran or chitosan.
  • WO 98/47535 discloses a single component liquid pharmaceutical composition for administration to a mucosal surface.
  • the composition comprises a therapeutic agent, a pectin with a low degree of esterification and an aqueous carrier that gels or can be adapted to gel at the site of application.
  • WO 90/09780 nor WO 98/47535 mentions buprenorphine.
  • analgesic formulations for nasal administration have now been devised. Rapid uptake of the analgesic across the nasal mucosa into the plasma can be achieved, which results in fast onset of analgesia. Further, the residence time of the analgesic in the nasal cavity can be increased, which results in prolonged analgesia. An improved profile of absorption of the analgesic into the systemic circulation can thus be achieved.
  • the present invention provides use of an analgesic and a delivery agent for the manufacture of a medicament for administration intranasally for the treatment of pain whereby, on introduction into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration C ther of 0.2 ng/ml or greater which is maintained for a duration T maint of at least 2 hours.
  • the invention thus enables a therapeutic blood plasma concentration of an analgesic, i.e. a concentration that produces pain relief or pain amelioration, to be attained within 30 minutes and maintained for up to 24 hours.
  • an analgesic i.e. a concentration that produces pain relief or pain amelioration
  • C ther denotes a therapeutic blood plasma concentration.
  • T maint denotes the duration for which C ther is maintained. Also provided are:
  • the invention further provides:
  • the invention also provides:
  • FIGS. 1 to 3 show the pharmacokinetic profiles that were obtained when buprenorphine formulations according to the invention (Formulations A to C) were administered intranasally to healthy volunteers at a dose of 800 ⁇ g of buprenorphine hydrochloride, calculated as buprenorphine.
  • Formulation A buprenorphine hydrochloride-pectin solution.
  • Formulation B buprenorphine hydrochloride-chitosan/hydroxypropylmethylcellulose (HPMC) solution.
  • Formulation C buprenorphine hydrochloride-chitosan/poloxamer 188 solution.
  • FIG. 4 shows a pharmacokinetic profile for a 400 ⁇ g dose of Formulation A. This profile was calculated from the data for the 800 ⁇ g dose of Formulation A. The pharmacokinetic profile for the 400 ⁇ g dose of Formulation D is also shown for comparison.
  • the invention is concerned with analgesic compositions that are delivered intranasally to achieve fast onset of analgesia and also prolonged analgesia.
  • An advantageous pharmacokinetic profile can therefore be attained.
  • An analgesic is combined with a delivery agent which is selected so that, on introduction into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration C ther of 0.2 ng/ml or greater which is maintained for a duration T maint of at least 2 hours. This effect may be achieved by administration of a single dose of the analgesic, or following multiple dosing.
  • compositions of the invention are adapted to enable the analgesic to be delivered such that a C ther of 0.2 ng/ml or more, for example 0.4 ng/ml or more, is attained within 30 minutes after introduction into the nasal cavity such as within 0.5 to 20 minutes, for example 2 to 15 minutes or 5 to 10 minutes.
  • C ther defines a therapeutic plasma concentration (or range thereof).
  • the term is used herein to define a blood plasma concentration (or range of plasma concentrations) of the analgesic that produces pain relief or pain amelioration.
  • C ther may be up to 100 ng/ml such as from 0.4 ng/ml to 80 mg/ml.
  • C ther may be from 0.4 ng/ml to 20 ng/ml.
  • C ther may be from 20 to 100 ng/ml such as from 50 to 80 ng/ml.
  • C ther values apply especially to opioid analgesics.
  • C ther may be from 0.4 to 5 ng/ml, for example 0.5 to 4 ng/ml or 0.8 to 2 ng/ml.
  • T maint is typically at least 2 hours.
  • the term T maint defines the duration of maintenance of C ther after administration of the analgesic.
  • the T maint can be from up to 24 hours, up to 12 hours or up to 6 hours such as from 2 to 4 hours or 2 to 3 hours.
  • a C ther of at least 0.4 ng/ml may be attained within 2 to 15′ minutes and maintained for a time period T maint of from 2 to 4 hours.
  • the analgesic delivery profile that can be attained may avoid the relatively high C max values associated with intravenous administration and so lead to an improved therapeutic index.
  • the peak plasma concentration of an analgesic that is attained after administration is defined as C max .
  • the invention can permit reduction or elimination of some or all of the side effects associated with the analgesic.
  • C max depends upon the analgesic.
  • C max is typically from 1 to 500 ng/ml or higher, for example from 1.5 to 400 ng/ml or from 1.5 to 100 ng/ml.
  • C max may be from 1.5 to 50 ng/ml.
  • C max may be from 50 to 500 ng/ml such as from 50 to 200 ng/ml or 50 to 100 ng/ml.
  • These C max values apply especially to opioid analgesics.
  • C max is typically from 1 to 5 ng/ml, for example from 1 to 4 ng/ml or from 1.5 to 3 ng/ml.
  • C max may be from 1 to 2 ng/ml, especially for lower doses.
  • the time at which C max is reached (T max ) is typically 10 to 40 minutes after administration, for example 10 to 30 minutes or 15 to 25 minutes such as 15 to 20 minutes.
  • C opt is used in relation to analgesic drugs which exhibit a dose-response curve to analgesia which is displaced to the left with respect to the dose-response curve for side-effects.
  • the term defines a therapeutic plasma concentration or range thereof which produces acceptable pain relief or pain amelioration but which does not produce side-effects or produces side effects which are less than those associated with higher plasma concentrations.
  • the analgesic may be an opioid analgesic. It may be a mixed agonist/antagonist such as a mixed mu-agonist/antagonist (also known as a partial agonist) or a mixed mu- and kappa-agonist/antagonist. It may be a mu-agonist.
  • a preferred opioid analgesic is buprenorphine or a physiologically acceptable salt or ester thereof. Buprenorphine (Chemical Abstracts Registration No.
  • the buprenorphine salt may be an acid addition salt or a salt with a base.
  • Suitable acid addition salts include the hydrochloride, sulphate, methane sulphonate, stearate, tartrate and lactate salts.
  • the hydrochloride salt is preferred (CAS RN 53152-21-9)
  • opioid analgesics include alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, butorphanol, clonitazene, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetylbutyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentanyl, heroin, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphanol, levophenacylmorphan, lofentanil, meperidine, meptazinol,
  • the salts for use in the invention may be any physiologically acceptable salts, including pharmaceutically acceptable acid addition salts.
  • examples include hydrochloride salts (for example the hydrochloride salts of nalbuphine, profadol, buprenorphine, morphine, pentazocine, naloxone and nalorphine) as well as levorphanol tartrate, nalorphine hydrobromide, levallorphan tartrate, morphine sulfate, butorphanol tartrate, pentazocine lactate and phenazocine hydrobromide.
  • hydrochloride salts for example the hydrochloride salts of nalbuphine, profadol, buprenorphine, morphine, pentazocine, naloxone and nalorphine
  • levorphanol tartrate for example the hydrochloride salts of nalbuphine, profadol, buprenorphine, morphine, pen
  • the analgesic may be a non-opioid analgesic such as a NSAID, a tricylic antidepressant (e.g. amitryptyline), an anticonvulsant (e.g. gabapentin) or an antimigraine compound (e.g. sumatriptan or naratriptan).
  • a NSAID may be a cyclooxygenase (COX) COX-1 or COX-2 inhibitor.
  • NSAIDs include ibuprofen, flurbiprofen, diclofenac, indomethacin, piroxicam, ketoprofen, etodolac, diflusinal, meloxicam, aceclofenac, fenoprofen, naproxen, tiaprofenic acid, tolmetin, celecoxib and rofecoxib, and their physiologically acceptable salts and esters.
  • Suitable salts are alkali addition salts such as the potassium or sodium salt.
  • a preferred NSAID is diclofenac (CAS RN 15307-86-5; 2-[(2,6-dichlorophenyl)amino]benzeneacetic acid) or a physiologically acceptable salt or ester thereof.
  • Diclofenac has the formula:
  • Diclofenac is available in various forms.
  • Diclofenac Sodium (CAS RN 15307-79-60; 2-[(2,6-dichlorophenyl)amino]benzeneacetic acid sodium salt) is sold under the following trade marks: Allorvan, Benfofen, Dealgic, Deflamat, Delphinac, Diclomax, Diclometin, Dichlophlogont, Diclo-Puren, Dicloreum, Diclo-Spondyril, Dolobasan, Duravolten, Ecofenac, brieflyon, Lexobene, Motifene, Neriodin, Novapirina, Primofenac, Prophenatin, Rewodina, Rhumalgan, Trabona, Tsudohmin, Valetan, Voldal, Voltaren and Xenid.
  • Diclofenac Potassium (CAS RN 15307-81-0; 2-[(2,6-dichlorophenyl)amino]benzene
  • etodolac (CAS RN 41340-25-4; 1,8-Diethyl-1,3,4,9-tetrahydropyrano[3,4-b]indole-1-acetic acid) or a physiologically acceptable salt or ester thereof.
  • Etodolac is sold under the trade marks Etogesic, Lodine, Tedolan and Ultradol.
  • Etodolac has the formula:
  • NSAIDs are piroxicam and meloxicam, and physiologically acceptable salts and esters thereof.
  • Piroxicam (CAS RN 36322-90-4; 4-Hydroxy-2-methyl-N-2-pyridinyl-2H-1,2-benzothiazine-3-carboxamide. 1,1-dioxide) is sold under the trade mark Feldine, Sinartrol, Zelis, Zen, Brexin, Cicladol or Cycladol and has the formula:
  • the analgesic is provided in a formulation suitable for nasal administration in combination with a delivery agent.
  • the formulation is typically a liquid formulation, especially as an aqueous solution.
  • the formulation may be in the form of a powder or microspheres.
  • the concentration of buprenorphine or buprenorphine salt or ester is from 0.1 to 10 mg/ml, for example from 0.5 to 8 mg/ml. Preferred concentrations are 1 to 6 mg/ml, for example 1 to 4 mg/ml calculated as buprenorphine.
  • Suitable formulations can contain buprenorphine or a buprenorphine salt or ester in an amount of 1 mg/ml or 4 mg/ml, calculated as buprenorphine.
  • the delivery agent is selected so that rapid onset and prolonged analgesia is obtained.
  • the delivery agent acts to deliver the analgesic to the bloodstream.
  • the delivery agent acts as an analgesic absorption modifier and any of a wide variety of delivery agents may be used providing that this functional requirement is met.
  • the delivery agent may comprise an absorption promoting agent.
  • Such agents promote uptake of the analgesic component into the bloodstream. They may act via a variety of different mechanisms. Particularly preferred are mucosal adhesives. Such adhesives maintain an intimate association between the bulk analgesic composition and the nasal mucosa, so enhancing absorption and extending the T maint of the analgesic component. They can also be used to lower the analgesic C max which may be important in applications where the minimization or elimination of side-effects is desired.
  • Suitable absorption promoting agents include cationic polymers (particularly chitosans), surface active agents, fatty acids, chelating agents, mucolytic agents, cyclodextrins, diethylaminoethyl-dextran (DEAE-dextran; a polycationic derivative of dextran) or combinations thereof.
  • cationic polymers particularly chitosans
  • surface active agents particularly fatty acids, chelating agents, mucolytic agents, cyclodextrins, diethylaminoethyl-dextran (DEAE-dextran; a polycationic derivative of dextran) or combinations thereof.
  • DAE-dextran diethylaminoethyl-dextran
  • pectins having a degree of esterification of less than 50%, especially from 10 to 35%, and chitosans.
  • the composition of the invention takes the form of an aqueous solution when the delivery agent is a pectin.
  • the pectin acts as a gelling agent.
  • a pectin-containing composition of the invention gels on the mucosal surfaces of the nasal cavity after delivery without the need for an extraneous source of divalent metal ions.
  • the analgesic that is formulated with the pectin is thus retained for longer on the surfaces of the nasal epithelium.
  • the resulting sustained release of the analgesic into the bloodstream enables prolonged analgesia to be achieved. Improved delivery of the analgesic can consequently be obtained. Rapid uptake of the analgesic also results, which leads to fast onset of analgesia.
  • the pectins employed in the invention have a degree of esterification of less than 50%.
  • a pectin is a polysaccharide substance present in the cell walls of all plant tissues. Commercially pectins are generally obtained from the dilute acid extract of the inner portion of the rind of citrus fruits or from apple pomace.
  • a pectin consists of partially methoxylated polygalacturonic acids. The proportion of galacturonic acid moieties in the methyl ester form represents the degree of esterification (DE).
  • DE is well understood by those skilled in the art and may be represented as the percentage of the total number of carboxyl groups that are esterified, i.e.
  • a degree of esterification of 80% or as the methoxyl content of the pectin.
  • DE refers to the total percentage of carboxyl groups that are esterified.
  • Pectins can be categorised into those having a low degree of esterification (low methoxylation) or a high degree of esterification (high methoxylation).
  • a “low DE” or “LM” pectin has a degree of esterification below 50% whereas a “high DE” or “HM” pectin has a degree of esterification of 50% or above.
  • the gelling properties of aqueous pectin solutions can be controlled by the concentration of pectin, the type of pectin, especially the degree of esterification of the galacturonic acid units, and the presence of added salts.
  • Low DE pectins are used in the present invention.
  • the degree of esterification of the pectin used in the invention is preferably less than 35%. The degree of esterification may thus be from 10 to 35%, for example from 15 to 25%.
  • Low DE pectins may be purchased commercially.
  • An example of a low DE pectin is SLENDID (trade mark) 100, supplied by CP Kelco (Lille Skenved) which has a degree of esterification of around 15 to 25%.
  • a pectin-containing solution of the invention must not gel on storage. It should not gel prior to application to the nasal cavity. It must therefore be substantially free of agents which would cause the solution to gel.
  • a solution of the invention must be substantially free of divalent metal ions and especially calcium ions. The content of divalent metal ions in the solution must therefore be minimised.
  • a pectin-containing solution of the invention may therefore contain a negligible concentration of divalent metal ions or there may no detectable divalent metal ions.
  • a pectin is typically present in a solution of the invention at a concentration of from 5 to 40 mg/ml, for example from 5 to 30 mg/ml. More preferably, the pectin concentration is from 10 to 30 ng/ml or from 10 to 25 mg/ml.
  • the pectin and the pectin concentration are selected such that the solution gels on delivery to the nasal mucosa.
  • composition of the invention also takes the form of an aqueous solution when the delivery agent is a chitosan.
  • Chitosans are cationic polymers that have mucoadhesive properties. The mucoadhesion is thought to result from an interaction between the positively charged chitosan molecule and the negatively charged sialic acid group's on mucin (Soane et al, Int. J. Pharm 178, 55-65, 1999).
  • chitosan we include all derivatives of chitin, or poly-N-acetyl-D-glucosamine, including all polyglucosamines and oligomers of glucosamine materials of different molecular weights, in which the greater proportion of the N-acetyl groups have been removed through hydrolysis (deacetylation).
  • the chitosan is produced from chitin by deacetylation to a degree of greater than 40%, preferably between 50 and 98%, more preferably between 70% and 90%.
  • the chitosan typically has a molecular weight of 4,000 Da or more, preferably from 10,000 to 1,000,000 Da, more preferably from 15,000 to 750,000 Da and most preferably from 50,000 to 500,000 Da.
  • the chitosan may thus be a deacetylated chitin. It may be a physiologically acceptable salt. Suitable physiologically acceptable salts include salts with a pharmaceutically acceptable mineral or organic acid such as the nitrate, phosphate, lactate, citrate, hydrochloride and acetate salts. Preferred salts are chitosan glutamate and chitosan hydrochloride.
  • the chitosan may be a derivative of a deacetylated chitin.
  • Suitable derivatives include, but are not limited to, ester, ether or other derivatives formed by bonding of acyl and/or alkyl groups with the hydroxy groups, but not the amino groups, of a deacetylated chitin. Examples are O—(C 1 -C 6 alkyl)ethers of deacetylated chitin and O-acyl esters of deacetylated chitin.
  • Derivatives also include modified forms of a deacetylated chitin for example a deacetylated chitin conjugated to polyethylene glycol.
  • Low and medium viscosity chitosans suitable for use in the present invention may be obtained from various sources, including FMC Biopolymer, Drammen, Norway; Seigagaku America Inc., Maryland, USA; Meron (India) Pvt, Ltd., India; Vanson Ltd, Virginia, USA; and AMS Biotechnology Ltd., UK.
  • Suitable derivatives include those that are disclosed in Roberts, Chitin Chemistry, MacMillan Press Ltd., London (1992).
  • Particularly preferred chitosan compounds that may be mentioned include “Protosan” (trade mark) available from FMC Biopolymer, Drammen, Norway.
  • the chitosan is preferably water-soluble.
  • An aqueous solution of chitosan may be prepared by dissolving chitosan base or a derivative of chitosan base in a pharmaceutically acceptable mineral or organic acid such as hydrochloric, lactic, citric or glutamic acid or by dissolving a chitosan salt in water.
  • the chitosan is typically present in solution at a concentration of from 0.1 to 20 mg/ml, for example from 0.5 to 20 mg/ml.
  • the solution contains from 1 to 15 mg/ml, more preferably from 2 to 10 mg/ml, of chitosan.
  • a chitosan concentration of 5 mg/ml is particularly suitable.
  • Chitosan-containing solutions of the invention may also contain hydroxypropylmethylcellulose (HPMC) or a polyoxyethylene-polyoxypropylene copolymer.
  • HPMC hydroxypropylmethylcellulose
  • Polyoxyethylene-polyoxypropylene copolymer a polyoxyethylene-polyoxypropylene copolymer
  • HPMC any suitable HPMC may be employed.
  • Several grades of HPMC are available.
  • Dow Chemical Company produces a range of HPMC polymers under the trade mark Methocel.
  • the grade and concentration of HPMC is chosen such that the solution of the invention preferably has a viscosity, at 25° C. as measured by a cone and plate viscometer (e.g. Brookfield), in the range from 1 to 200 cps, more preferably from 3 to 150 cps and most preferably from 5 to 100 cps.
  • Producing a solution having a particular viscosity is within the capability of one skilled in the at and can be achieved, for example, by using a high concentration of a low viscosity HPMC or a low concentration of a high viscosity HPMC.
  • the HPMC is preferably one having an apparent viscosity (measured as a 2% solution in water at 20° C.) in the range from 3000 to 6000 cps.
  • the concentration of the HPMC having a viscosity of from 3000 to 6000 cps is typically in the range from 0.1 to 15 mg/ml, for example from 0.5 to 10 mg/ml and preferably from 1 to 5 mg/ml.
  • the polyoxyethylene-polyoxypropylene copolymer typically has a molecular weight of from 2,500 to 18,000 for example from 7,000 to 15,000.
  • the copolymer is a block copolymer of the general formula:
  • a is from 2 to 130 and b is from 15 to 67.
  • the value for a may be from 40 to 100 such as from 60 to 90 or from 70 to 95.
  • the value for b may be from 20 to 40 such as from 25 to 35.
  • poloxamers Such copolymers are known as poloxamers.
  • poloxamers are available commercially, from suppliers such as BASF, and vary with respect to molecular weight and the proportions of ethylene oxide “a” units acid propylene oxide “b” units.
  • a commercially available poloxamer suitable for use in the present invention is poloxamer 188 which structurally contains 80 “a” units and 27 “b” units and has a molecular weight of 7680-9510 (Handbook of Pharmaceutical Excipients, editor A. H. Kippe, third edition, Pharmaceutical Press, London, UK, 2000).
  • the poloxamer is poloxamer 188.
  • the polyoxyethylene-polyoxypropylene copolymer is typically present in an amount of from 50 to 200 mg/ml, preferably from 65 to 160 mg/ml and more preferably from 80 to 120 mg/ml. A preferred concentration is 100 mg/ml.
  • cationic polymers besides chitosans suitable for use as absorption promoting agents include polycationic carbohydrates.
  • the polycationic substances preferably have a molecular weight of at least 10,000. They may be in liquid formulations at concentrations of 0.01 to 50% w/v, preferably 0.1 to 50% w/v and more preferably 0.2 to 30% w/v.
  • polycationic polymers examples include polyaminoacids (e.g. polylysine), polyquaternary compounds, protamine, polyamine, DEAE-imine, polyvinylpyridine, polythiodiethyl-aminomethylethylene, polyhistidine, DEAE-methacrylate, DEAF-acrylamide, poly-p-aminostyrene, polyoxethane, co-polymethacrylates (e.g. copolymers of HPMA, N-(2-hydroxypropyl)-methacrylamide), GAFQUAT (see for example U.S. Pat. No. 3,910,862) and polyamidoamines.
  • polyaminoacids e.g. polylysine
  • polyquaternary compounds e.g. polyquaternary compounds
  • protamine polyamine
  • polyamine DEAE-imine
  • polyvinylpyridine polythiodiethyl-aminomethylethylene
  • polyhistidine DEAE-methacrylate
  • Suitable surface active agents for use according to the present invention are bile salts (for example sodium deoxycholate and cholylsarcosine, a synthetic N-acyl conjugate of cholic acid with sarcosine [N-methylglycine]).
  • bile salt derivatives for example sodium tauro dihydrofusidate.
  • Any of a wide range of non-ionic surfactants e.g. polyoxyethylene-9 lauryl ether), phospholipids and lysophosphatidyl compounds (e.g.
  • lysolecithin lysophosphatidyl-ethanolamine, lysophosphatidylcholine, lysophosphatidylglycerol, lysophosphatidylserine and lysophosphatidic acid
  • Water-soluble phospholipids may also be employed (e.g. short chain phosphatidylglycerol and phosphatidylcholines).
  • concentration of surface active agents used according to the invention varies according to the physico-chemical properties of the surface active agent selected, but typical concentrations are in the range 0.02 to 10% w/v.
  • Particularly preferred surface active agents for use as absorption promoting materials are phospholipids and lysophosphatides (hydrolysis products of phospholipids), both of which form micellar structures.
  • microspheres When microspheres are used as the delivery agent, they are preferably prepared from a biocompatible material that will gel in contact with the mucosal surface: Substantially uniform solid microspheres are preferred. Starch microspheres (crosslinked if necessary) are preferred.
  • Microspheres may also be prepared from starch derivatives, modified starches (such as amylodextrin), gelatin, albumin, collagen, dextran and dextran derivatives, polyvinyl alcohol, polylactide-co-glycolide, hyaluronic acid and derivatives thereof (such as benzyl and ethyl esters), gellan gum and derivatives thereof (such as benzyl and ethyl esters) and pectin and derivatives thereof (such as benzyl and ethyl esters).
  • modified starches such as amylodextrin
  • gelatin such as amylodextrin
  • albumin such as a polyvinyl alcohol, polylactide-co-glycolide
  • hyaluronic acid and derivatives thereof such as benzyl and ethyl esters
  • gellan gum and derivatives thereof such as benzyl and ethyl esters
  • pectin and derivatives thereof such
  • starch derivatives including hydroxyethyl starch, hydroxypropyl starch, carboxymethyl starch, cationic starch, acetylated starch, phosphorylated starch, succinate derivatives of starch and grafted starches.
  • Suitable dextran derivatives include, diethylaminoethyl-dextran (DEAE-dextran), dextran sulphate, dextran methyl-benzylamide sulphonates, dextran methyl-benzylamide carboxylates, carboxymethyl dextran, diphosphonate dextran, dextran hydrazide, palmitoyldextran and dextran phosphate.
  • DEAE-dextran diethylaminoethyl-dextran
  • dextran sulphate dextran methyl-benzylamide sulphonates
  • dextran methyl-benzylamide carboxylates carboxymethyl dextran
  • diphosphonate dextran dextran hydrazide
  • palmitoyldextran dextran phosphate
  • microspheres for use according to the invention may be carried out by known processes, including emulsion and phase separation methods (see for example Davis et al., (Eds), “Microspheres and Drug Therapy”, Elsevier Biomedical Press, 1984, which parts relating to microsphere preparation are incorporated herein by reference).
  • albumin microspheres may be made using the water-in-oil emulsification method where a dispersion of albumin in oil is produced by homogenization or stirring, with the addition if necessary of small amounts of an appropriate surface active agent.
  • the size of the microspheres is largely determined by the speed of stirring or the homogenization conditions. Agitation can be provided by a simple laboratory stirrer or by more sophisticated devices (such as microfluidizers or homogenisers). Emulsification techniques may also be used to produce starch microspheres (as described in GB 1518121 and EP 223303) and for the preparation of gelatin microspheres.
  • Proteinaceous microspheres may be prepared by coacervation methods. Such methods include simple or complex coacervation as well as phase separation techniques (using solvents or electrolyte solutions). Such methods are well known to those skilled in the art and details may be found in standard textbooks (for example Florence and Attwood, Physicochemical Principles of Pharmacy 2nd Ed., MacMillan Press, 1988, Chapter 8).
  • microspheres may advantageously have controlled-release properties, which may be conferred by modifications of the microspheres (for example by controlling the degree of cross-linking or by the incorporation of excipients that alter the diffusional properties of the analgesic component).
  • controlled release properties may be incorporated by exploiting ion-exchange chemistry (for example DEAE-dextran and chitosan are positively charged and can be used for an ion-exchange interaction with metabolites that are negatively charged).
  • the maximum amount of analgesic component that can be carried by the microspheres is termed the loading capacity. It is determined by the physico-chemical properties of the analgesic component and in particular its size and affinity for the matrix of the microspheres. High loading capacities can be achieved when the analgesic is incorporated into the microspheres during microsphere manufacture.
  • Microcapsules may also be employed as an absorption promoting agent in the compositions of the invention. These microcapsules can be produced by a variety of methods.
  • the surface of the capsule may be inherently adhesive or can be modified by standard coating methods known to those skilled in the art. Suitable coating materials include bioadhesive polymers such as polycarbophil, carbopol, DEAE-dextran, alginate, microcrystalline cellulose, dextran, polycarbophils and chitosan).
  • Oil-in-water formulations can provide for the effective nasal delivery of analgesics that are poorly soluble in water. In such applications nasal irritation may also be reduced.
  • the oil phase of the emulsions of the invention may comprise a hydroxylated oil, particularly a hydroxylated vegetable oil.
  • hydroxylated oil is intended to cover any oil that contains hydroxylated fatty acids.
  • Preferred hydroxylated oils are hydroxylated vegetable oils, and a preferred hydroxylated vegetable oil for use in the present composition is castor oil.
  • castor oil is intended to include ricinus oil, oil of Palma Christie, tangantargon oil and Neoloid (as described in Merck Index, 12th Edition, p: 311), as well as the oil from Ricinus Zanzibarinus.
  • the latter has a high content of glycerides of ricinoleic acid.
  • castor oil comprises glycerides of ricinoleic acid (a hydroxy fatty acid).
  • castor oil When castor oil is used in the present invention, it may conveniently be obtained by cold pressing of the seeds of Ricinus Communis L. (family: Euphorbiaceae).
  • the oil phase in the emulsions of the invention may constitute 1 to 50% v/v of the emulsion.
  • a preferred concentration of oil in the emulsion is from 10 to 40% v/v. Particularly preferred are concentrations of 20 to 30% v/v.
  • the emulsion compositions of the invention can be prepared using conventional methods such as by homogenisation of a mixture of the oil and analgesic component with an aqueous phase (optionally together with a stabilizing agent).
  • Any suitable device may be used, including a microfluidizer or ultrasonic device, though microfluidizers are preferred for large scale production.
  • Suitable stabilizers for use in the emulsions of the invention include block copolymers containing a polyoxyethylene block (i.e. a block made up of repeating ethylene oxide moieties).
  • a suitable stabilizer of this type is PoloxamerTM.
  • Other suitable stabilizers include phospholipid emulsifiers (for example soy and egg lecithins). Particularly preferred is the egg lecithin Lipoid E80TM (from LipoidTM), which contains both phosphatidylcholine and phosphatidyl ethanoline.
  • suitable phospholipids include phospholipid-polyethylene glycol (PEG) conjugates (see for example Litzinger et al., Biochem Biophys Acta, 1190 (1994) 99-107).
  • any suitable concentration of stabilizer/emulsifier may be used, and it typically falls within the range 0.1 to 10% w/v in the aqueous phase of the emulsion. Particularly preferred are concentrations of 1 to 5% w/v.
  • Suitable pharmaceutically-acceptable co-emulsifiers include fatty acids, bile acids and salts thereof. Preferred fatty acids have greater than 8 carbon atoms, and particularly preferred is oleic acid. Of the suitable bile acids, preferred is deoxycholic acid. Suitable salts of the foregoing include the alkali metal (e.g. Na and K) salts. Co-emulsifiers can be added at a concentration of 1% w/v or less on the aqueous phase.
  • Buffering agents may also be used in the composition.
  • a buffer may be used to maintain a pH that is compatible with nasal fluid, to preserve emulsion stability and/or to ensure that the analgesic component does not partition from the emulsion oil phase into the aqueous phase.
  • thickening and gelling agents such as cellulose polymers, particularly sodium carboxymethyl cellulose, alginates, gellans, pectins, acrylic polymers, agar-agar, gum tragacanth, gum xanthan, hydroxyethyl cellulose, chitosan, as well as block copolymers of polyoxyethylene-polyoxypropylene.
  • Preservative agents such as methyl parabenzoates, benzylalcohol and chlorobutanol may also be added.
  • the delivery agent may comprise a liposome.
  • Liposomes are microscopic vesicles composed of an aqueous compartment surrounded by a phospholipid bilayer that acts as a permeable entrapment barrier. Many different classes of liposomes are known (see Gregoriadis (ed.) in Liposome Technology, 2nd edition, vol I-III, CRC Press, Boca Ranto, Fla., 1993). Some liposomes can provide controlled sustained release of the encapsulated drug. In such systems, the rate of drug release is determined by the liposome's physicochemical properties.
  • Liposomes can be tailored for a specific application by modification of size, composition, and surface charge to provide the desired rate of drug delivery (see Meisner D, et al: In Proceedings, 15th International Symposium on Controlled Release of Bioactive Materials. 15:262-263, 1988; Mezei M: In Drug Permeation Enhancement, Theory and Application. Hsieh DS (ed.): Marcel Dekker Inc., New York, 1993, pp 171-198; and Meisner D, et al: J Microencapsulation 6:379-387, 1989).
  • liposome-encapsulation can act as an effective and safe delivery agent in the compositions of the invention.
  • the sustained release property of the liposomal product can be regulated by the nature of the lipid membrane and by the inclusion of other excipients in the composition of the liposomal products.
  • Current liposome technology permits a reasonable prediction on the rate of drug release based on the composition of the liposome formulation.
  • the rate of drug release is primarily dependent on the nature of the phospholipids, e.g. hydrogenated (—H) or unhydrogenated (-G), or the phospholipid/cholesterol ratio (the higher this ratio, the faster the rate of release), the hydrophilic/lipophilic properties of the active ingredients and by the method of liposome manufacturing.
  • liposomes Materials and procedures for forming liposomes are well known to those skilled in the art and include ethanol or ether injection methods.
  • the lipid is dissolved in a solvent and the solvent evaporated (often under reduced pressure) to produce a thin film.
  • the film is then hydrated with agitation.
  • the analgesic component is incorporated at the lipid film forming stage (if lipophilic) or at the hydration phase as part of the aqueous hydrating phase (if hydrophilic).
  • the liposomes can be multilamellar lipid vesicles (MLV), unilamellar lipid vesicles (including small unilamellar vesicles (SUV) and large unilamellar vesicles (LUV)) and as multivesicular liposomes.
  • MLV multilamellar lipid vesicles
  • SUV small unilamellar vesicles
  • LUV large unilamellar vesicles
  • Lipid components typically comprise phospholipids and cholesterol while excipients may comprise tocopherol, antioxidants, viscosity inducing agents and/or preservatives.
  • Phospholipids are particularly useful, such as those selected from the group consisting of phosphatidylcholines, lysophosphatidylcholines, phosphatidylserines, phosphatidylethanolamines, and phosphatidylinositols.
  • Such phospholipids may be modified using, for example, cholesterols, stearylamines, stearic acid, and tocopherols.
  • compositions of the invention may further comprise other suitable excipients, including for example inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservatives.
  • suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc.
  • Excipients such as humectants, isotonicity agents, antioxidants, buffers and/or preservatives are preferably used.
  • Formulation and dosage will depend on, amongst other things, whether the analgesic is to be used in the form of drops or as a spray (aerosol).
  • suspensions, ointments and gels can be applied to the nasal cavity.
  • nasal mucous membranes are also capable of tolerating slightly hypertonic solutions.
  • appropriate oily or gel vehicles may be used or one or more polymeric materials may be included, which desirably should be capable of conferring bioadhesive characteristics to the vehicle.
  • nasal dosage forms are solutions, suspensions and gels, which contain a major amount of water (preferably purified water) in addition to the active ingredient. Minor amounts of other ingredients such as pH adjusters (e.g. a base such as NaOH), emulsifiers or dispersing agents, buffering agents, preservatives, wetting agents and jelling agents (e.g., methylcellulose) may also be present.
  • pH adjusters e.g. a base such as NaOH
  • emulsifiers or dispersing agents e.g. a base such as NaOH
  • buffering agents e.g., preservatives
  • wetting agents and jelling agents e.g., methylcellulose
  • the nasal compositions of the invention may be isotonic, hypertonic or hypotonic.
  • sustained release nasal compositions e.g. sustained release gels, can be readily prepared, preferably by employing the desired drug in one of its relatively insoluble forms, such as the free base or an insoluble salt.
  • composition of the present invention may therefore be adjusted, if necessary, to approximately the same osmotic pressure as that of the body fluids (i.e. isotonic).
  • Hypertonic solutions can irritate the delicate nasal membranes, while isotonic compositions do not.
  • Isotonicity can be achieved by adding glycerol or an ionic compound to the composition (for example, sodium chloride).
  • the compositions may take the form of a kit of parts, which kit may comprise the intranasal composition together with instructions for use and/or unit dosage containers and/or an intranasal delivery device.
  • compositions of the invention are administered intranasally to a patient in order to induce analgesia.
  • An effective amount of the analgesic is delivered to a patient.
  • a unit dose can be delivered to one nostril. Alternatively, half of a dose or two doses can be delivered to each nostril each time administration occurs. The dose will depend upon a number of factors including the analgesic that is being delivered, the age and sex of the patient, the nature and extent of the pain to be treated and the period of treatment.
  • a suitable dose for an opioid analgesic may be from 0.02 to 100 mg such as 0.1 to 50 mg.
  • a suitable dose is from 0.02 to 1.2 mg, such as from 50 to 600 ⁇ g or from 100 to 400 ⁇ g, calculated as buprenorphine.
  • the rapid onset analgesia produced by the solution of the invention may permit self-titration of analgesic by the patient.
  • the analgesic effect of an initial dose can be quickly and reliably gauged by the patient and, if insufficient, can be immediately supplemented by further dose(s) (often alternating between each nostril) until the required level of analgesia is attained.
  • Multiple dosing may also be used in order to extend pain relief. For example, from 1 to 4 doses, for example 2 to 4 doses, per day may be indicated.
  • compositions of the invention may be used to treat an existing pain condition or to prevent a pain condition from occurring.
  • An existing pain may be alleviated.
  • Compositions can be used to treat or manage chronic or acute pain, for example the management of post-operative pain (e.g. abdominal surgery, back surgery, cesarean section, hip replacement or knee replacement).
  • Other medical uses include: pre-operative intranasal administration of the solution of the invention; therapy or prophylaxis adjunctive to anesthesia; post-operative analgesia; the management of trauma pain; the management of cancer pain; the management of endometriosis; the management of inflammatory pain; the management of arthritis pain (including pain associated with rheumatoid arthritis and osteoarthritis); the management of back pain; the management of myocardial pain (for example ischaemic or infarction pain); the Management of dental pain; the management of neuropathic pain (e.g. diabetic neuropathy, post-herpetic neuralgia or trigeminal neuralgia); the management of colic (e.g.
  • renal colic or gallstones headache, migraine, fibromyalgia or dysmenorrhoea; the management of breakthrough pain associated with malignant and non-malignant disease; and the management of acute procedural pain (e.g. bone marrow aspiration or lumber puncture).
  • compositions according to the invention may be administered to the nasal cavity in forms including drops or sprays.
  • the preferred method of administration is using a spray device.
  • Spray devices can be single (unit) dose or multiple dose systems, for example comprising a bottle, pump and actuator. Suitable spray devices are available from various commercial sources including Pfeiffer, Valois, Bespak and Becton-Dickinson.
  • a nasal insufflator device When in the form of powder or microspheres, a nasal insufflator device may be employed. Such devices are already in use for commercial powder systems intended for nasal application.
  • the insufflator may be used to produce a fine, dispersed plume of the dry powder or microspheres.
  • the insufflator is preferably provided with means for administering a predetermined dose of the analgesic composition.
  • Powder or microspheres may be contained in a bottle or container adapted to be used with the insufflator.
  • powders or microspheres may be provided in capsules (e.g. gelatin capsules) or other single dose devices adapted for nasal administration, in which embodiments the insufflator may comprise means for breaking open the capsule (or other single dose device).
  • a first solution of the invention consists essentially of 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, from 0.1 to 20 mg/ml of a chitosan, from 0.1 to 15 mg/ml of HPMC, and water.
  • a second solution of the invention consists essentially of 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, from 0.1 to 20 mg/ml of chitosan, from 50 to 200 mg/ml of a polyoxyethylene-polyoxypropylene copolymer of the general formula HO(C 2 H 4 O) a (C 3 H 6 O) b (C 2 H 4 O) a H wherein a is from 2 to 130 and b is from 15 to 67, and water.
  • the buprenorphine salt may be an acid addition salt or a salt with a base.
  • Suitable acid addition salts the hydrochloride, sulphate, methane sulphonate, stearate, tartrate and lactate salts.
  • the hydrochloride salt is preferred.
  • the concentration of buprenorphine or buprenorphine salt or ester in either solution is from 0.1 to 10 mg/ml, for example from 0.5 to 8 mg/ml. Preferred concentrations are 1 to 6 mg/ml, for example 1 to 4 mg/ml. Suitable solutions can contain the buprenorphine or buprenorphine salt or ester at a concentration of 1 mg/ml or 4 mg/ml, calculated as buprenorphine. Each solution is typically delivered as a nasal spray.
  • a 100 ⁇ l spray of a solution containing 1 to 4 mg/ml of buprenorphine or a buprenorphine salt or ester, calculated as buprenorphine thus results in a clinical dose of 100 to 400 ⁇ g of the buprenorphine or buprenorphine salt or ester, calculated as buprenorphine.
  • Two such sprays may be given per nostril per administration time to deliver a dose of up to 4 ⁇ 400 ⁇ g, i.e. up to 1600 ⁇ g, of buprenorphine or the buprenorphine salt or ester, calculated as buprenorphine.
  • chitosans are described above.
  • the chitosan is present in solution at a concentration of from 0.1 to 20 mg/ml, for example from 0.5 to 20 mg/ml.
  • the solution contains from 1 to 15 mg/ml, more preferably from 2 to 10 mg/ml, of chitosan.
  • a chitosan concentration of 5 mg/ml is particularly suitable.
  • HPMC used in the solution of the invention is preferably one having an apparent viscosity (measured as a 2% solution in water at 20° C.) in the range from 3000 to 6000 cps.
  • concentration of the HPMC having a viscosity of from 3000 to 6000 cps is in the range from 0.1 to 15 mg/ml, preferably from 0.5 to 10 mg/ml and preferably from 1 to 5 mg/ml.
  • Suitable polyoxyethylene-polyoxypropylene copolymers typically are described above.
  • the polyoxyethylene-polyoxypropylene copolymer is present in an amount of from 50 to 200 mg/ml, preferably from 65 to 160 mg/ml and more preferably from 80 to 120 mg/ml.
  • a preferred concentration is 100 mg/ml.
  • any suitable preservative may be present in the solutions, in particular a preservative that prevents microbial spoilage of the solution.
  • the preservative must be compatible with the other components of the solution.
  • the preservative may be any pharmaceutically acceptable preservative, for example a quaternary ammonium compound such as benzalkonium chloride.
  • the solution has a pH of from 3 to 4.8. Any pH within this range may be employed provided the buprenorphine or buprenorphine salt or ester remains dissolved in the solution.
  • the pH may be from 3.2 to 4.2, for example from 3.2 to 4.0 or 3.5 to 4.0.
  • a suitable pH is from 3.6 to 3.8.
  • the pH may be adjusted to an appropriate value by addition of a physiologically acceptable acid and/or physiologically acceptable buffer.
  • the pH may thus be adjusted solely by means of a physiologically acceptable mineral acid or solely by means of a physiologically acceptable organic acid.
  • the use of hydrochloric acid is preferred.
  • a tonicity adjustment agent may be included in the solution.
  • the tonicity adjustment agent may be a sugar, for example dextrose, or a polyhydric alcohol, for example mannitol.
  • a solution may be hypertonic, substantially isotonic or hypotonic.
  • a substantially isotonic solution can have an osmolality of from 0.28 to 0.32 osmol/kg.
  • An exactly isotonic solution is 0.29 osmol/kg.
  • a sufficient amount of a tonicity adjustment agent such as dextrose or mannitol may therefore be present to achieve a desired osmolality.
  • a solution contains 50 mg/ml dextrose or mannitol.
  • the osmolality of a solution containing chitosan and HPMC or a poloxamer may be from 0.1 to 0.8 osmol/kg such as from 0.2 to 0.6 osmol/kg or preferably from 0.32 to 0.4 osmol/kg.
  • the solutions may also contain other ingredients such as an antioxidant, chelating agent or other agent generally used in pharmaceutical liquid preparations.
  • the solution can be a sterile solution.
  • the solution containing chitosan and HPMC is prepared by dissolving buprenorphine or a physiologically acceptable salt or ester thereof, a chitosan and HPMC in water, typically Water for Injections.
  • the amount of the buprenorphine or salt or ester thereof is selected so that from 0.1 to 10 mg/ml of buprenorphine or the buprenorphine salt or ester is dissolved in the solution.
  • the required concentrations of the chitosan and of HPMC are provided too.
  • a preservative can be dissolved in the solution.
  • the pH of the solution can be adjusted to a value within the range from 3 to 4.8 as required. Preferably the pH is adjusted by means of hydrochloric acid.
  • a solution containing chitosan and a polyoxyethylene-polyoxypropylene copolymer is prepared by dissolving buprenorphine or a physiologically acceptable salt or ester thereof, a chitosan and the polyoxyethylene-polyoxypropylene copolymer in water, typically Water for Injections.
  • the amount of the buprenorphine or salt or ester thereof is selected so that from 0.1 to 10 mg/ml of buprenorphine or the buprenorphine salt or ester is dissolved in the solution.
  • the required concentrations of the chitosan and of the polyoxyethylene-polyoxypropylene copolymer are provided too.
  • a preservative can be dissolved in the solution.
  • the pH of the solution can be adjusted to a value within the range from 3 to 4.8 as required. Preferably, the pH is adjusted by means of hydrochloric acid.
  • dextrose or mannitol may be dissolved in the water in which the buprenorphine or buprenorphine salt or ester is being dissolved.
  • a sterile solution can be obtained either by using sterile starting materials and operating under sterile conditions and/or by using standard sterilising techniques such as passing the final solution through a sterilising filter.
  • a pyrogen-free solution can thus be provided.
  • the solution can then be introduced into a nasal delivery device, typically a sterile such device. If required, prior to sealing the device, the solution may be overlaid with an inert gas such as nitrogen to protect it from oxidation.
  • a third pharmaceutical solution of the invention consists essentially of 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, from 5 to 40 mg/ml of a pectin having a low degree of esterification, in particular a degree of esterification of less than 50%, and water.
  • the buprenorphine salt may be an acid addition salt or a salt with a base. Suitable acid addition salts include the hydrochloride, sulphate, methane sulphonate, stearate, tartrate and lactate salts. The hydrochloride salt is preferred.
  • the concentration of buprenorphine or buprenorphine salt or ester is from 0.1 to 10 mg/ml, for example from 0.5 to 8 mg/ml. Preferred concentrations are 1 to 6 mg/ml, for example 1 to 4 mg/ml calculated as buprenorphine. Suitable solutions can contain buprenorphine or a buprenorphine salt or ester in an amount of 1 mg/ml or 4 mg/ml, calculated as buprenorphine.
  • the solution is typically delivered as a nasal spray.
  • a 100 ⁇ l spray of a solution containing 1 to 4 mg/ml of buprenorphine or a buprenorphine salt or ester, calculated as buprenorphine thus results in a clinical dose of 100 to 400 ⁇ g of the buprenorphine or buprenorphine salt or ester, calculated as buprenorphine.
  • Two such sprays may be given per nostril per administration time to deliver a dose of up to 4 ⁇ 400 ⁇ g, i.e. up to 1600 ⁇ g, of buprenorphine or the buprenorphine salt or ester, calculated as buprenorphine.
  • the solutions gels on the mucosal surfaces of the nasal cavity without the need for extraneous source of divalent metal ions.
  • the pectin is present in the solution of the invention at a concentration of from 5 to 40 mg/ml, for example from 5 to 30 mg/ml. More preferably, the pectin concentration is from 10 to 30 mg/ml or from 10 to 25 mg/ml.
  • a pectin-containing solution of the invention has a pH of from 3 to 4.2. Any pH within this range may be employed provided the buprenorphine or buprenorphine salt or esteremains dissolved in the solution.
  • the pH may be from 3.2 to 4.0, for example from 3.5 to 4.0.
  • a particularly suitable pH is from 3.6 to 3.8.
  • the pH may be adjusted to an appropriate value by addition of a physiologically acceptable acid and/or physiologically acceptable buffer.
  • the pH may thus be adjusted solely by means of a physiologically acceptable mineral acid or solely by means of a physiologically acceptable organic acid.
  • the use of hydrochloric acid is preferred.
  • any suitable preservative may be present in the pectin-containing solution, in particular a preservative that prevents microbial spoilage of the solution.
  • the preservative may be any pharmaceutically acceptable preservative, for example phenylethyl alcohol or propyl hydroxybenzoate (propylparaben) or one of its salts.
  • the phenylethyl alcohol and the propylparaben or propylparaben salt are preferably used in combination.
  • the preservative must be compatible with the other components of the solution and, in particular, must not cause gelling of the solution.
  • Pectin-containing solutions may include a tonicity adjustment agent such as a sugar, for example dextrose, or a polyhydric alcohol for example mannitol.
  • a solution may be hypertonic, substantially isotonic or hypotonic.
  • a substantially isotonic solution can have an osmolality of from 0.28 to 0.32 osmol/kg.
  • An exactly isotonic solution is 0.29 osmol/kg.
  • the osmolality of a solution may be from 0.1 to 0.8 osmol/kg such as from 0.2 to 0.6 osmol/kg or preferably from 0.35 to 0.5 osmol/kg.
  • a suitable osmolality range is from 0.32 to 0.36 osmol/kg.
  • a sufficient amount of a tonicity adjustment agent such as dextrose or mannitol may therefore be present to achieve such osmolalities.
  • a solution contains 50 mg/ml dextrose or mannitol
  • the pectin-containing solution is prepared by dissolving buprenorphine or a physiologically acceptable salt or ester thereof in water, typically Water for Injections, and the resulting solution is mixed with a solution of a suitable pectin in water, again typically Water for Injections.
  • the amount of the buprenorphine or salt or ester thereof and of the pectin are selected so that from 0.1 to 10 mg/ml of buprenorphine or the buprenorphine salt or ester and from 5 to 40 mg/ml of pectin are dissolved in the mixed solution.
  • a preservative or combination of preservatives may be dissolved in the solution.
  • the pH of the mixed, solution can be adjusted to a value within the range from 3 to 4.2 as required.
  • the pH is adjusted with hydrochloric acid if pH adjustment is required.
  • dextrose or mannitol may be dissolved in the water in which the buprenorphine or buprenorphine salt or ester is being dissolved.
  • a sterile solution can be obtained either by using sterile starting materials and operating under sterile conditions and/or by using standard sterilising techniques such as passing the final solution through a sterilising filter.
  • a pyrogen-free solution can thus be provided.
  • the solution can then be introduced into a nasal delivery device, typically a sterile such device. If required, prior to sealing the device, the solution may be overlaid with an inert gas such as nitrogen to protect it from oxidation.
  • Each of the three solutions of the invention is administered intranasally to a patient in order to induce analgesia. Rapid onset of analgesia and prolonged analgesia can thus be obtained.
  • An effective amount of buprenorphine or a salt or ester thereof is delivered to a patient.
  • a unit dose can be delivered to one nostril. Alternatively, half of a dose or two doses can be delivered to each nostril each administration time. The dose will depend upon a number of factors including the age and sex of the patient, the nature and extent of the pain to be treated and the period of treatment.
  • a suitable dose of buprenorphine or a buprenorphine salt or ester is from 0.02 to 1.2 mg, such as from 50 to 600 ⁇ g or from 100 to 400 ⁇ g, calculated as buprenorphine.
  • the rapid onset analgesia produced by the solution of the invention may permit self-titration of analgesic by the patient.
  • the analgesic effect of an initial dose can be quickly and reliably gauged by the patient and, if insufficient, can be immediately supplemented by further dose(s) (often alternating between each nostril) until the required level of analgesia is attained.
  • Multiple dosing may also be used in order to extend pain relief For example, from 2 to 4 doses per day may be indicated.
  • solutions of the invention may be used to treat an existing pain condition or to prevent a pain condition from occurring.
  • An existing pain may be alleviated.
  • Solutions of the invention can be used to treat or manage chronic or acute pain, for example the management of post-operative pain (e.g. abdominal surgery, back surgery, cesarean section, hip replacement or knee replacement). Other medical uses have been described above.
  • the solutions according to the invention may be administered to the nasal cavity in forms including drops or sprays.
  • the preferred method of administration is using a spray device.
  • Spray devices can be single (unit) dose or multiple dose systems, for example comprising a bottle, pump and actuator. Suitable spray devices are available from various commercial sources including Pfeiffer, Valois, Bespak and Becton-Dickinson.
  • the analgesic delivery profile that can be attained may avoid the relatively high C max values associated with intravenous administration and so lead to an improved therapeutic index.
  • the peak plasma concentration of an analgesic that is attained after administration is defined as C max .
  • the invention can permit reduction or elimination of some or all of the side effects associated with the analgesic.
  • C max is typically from 1 to 5 ng/ml, for example from 1 to 4 ng/ml or from 1.5 to 3 ng/ml. C max may be from 1 to 2 ng/ml, especially for lower doses of buprenorphine.
  • the time at which C max is reached (T max ) is typically 10 to 40 minutes after administration, for example 10 to 30 minutes or 15 to 25 minutes such as 15 to 20 minutes.
  • C opt is used in relation to analgesic drugs which exhibit a dose-response curve to analgesia which is displaced to the left with respect to the dose-response curve for side-effects.
  • the term defines a therapeutic plasma concentration or range thereof which produces acceptable pain relief or pain amelioration but which does not produce side-effects or produces side effects which are less than those associated with higher plasma concentrations.
  • the solutions of the invention enable the buprenorphine or salt or ester thereof to be delivered such that a C ther of 0.2 ng/ml or more, for example 0.4 ng/ml or more, is attained within 30 minutes after introduction into the nasal cavity within 30 minutes, for example within 0.5 to 20 minutes, such as 2 to 15 minutes or 5 to 10 minutes.
  • C ther defines a therapeutic plasma concentration (or range thereof).
  • the term is used herein to define a blood plasma concentration (or range of plasma concentrations) of the buprenorphine or salt or ester thereof that produces pain relief or pain amelioration.
  • C ther may be from 0.4 to 5 ng/ml, for example 0.4 to 1 ng/ml or 0.5 to 4 ng/ml or 0.8 to 2 ng/ml.
  • the T maint is typically at least 2 hours.
  • T maint defines the duration of maintenance of C ther after administration of the analgesic.
  • the T maint can be from up to 24 hours, up to 12 hours or up to 6 hours such as from 2 to 4 hours or 2 to 3 hours.
  • a C ther of at least 0.4 ng/ml may be attained within 2 to 15 minutes and maintained for a time period T maint of from 2 to 4 hours.
  • pectin 5 g of pectin (SLENDID (trade mark) 100, CP Kelco, Denmark) was dissolved by stirring into approximately 180 ml of water for injection (WFI) (Baxter, UK). 1075 mg of buprenorphine hydrochloride (MacFarlan Smith, UK) and 12.5 g of dextrose (Roquette) were dissolved into the pectin solution. 1:25 ml of phenylethyl alcohol (R. C. Treat, UK) and 50 mg of propyl hydroxybenzoate (Nipa, UK) were dissolved into the pectin/buprenorphine solution. The solution was adjusted to 250 ml using WFI. 1M hydrochloric acid (BDH, UK) was added to adjust the pH to 3.6.
  • WFI water for injection
  • the final product was a slightly turbid solution 4.3 mg/ml buprenorphine hydrochloride (corresponding to 4 mg/ml buprenorphine), 20 mg/ml pectin, 50 mg/ml dextrose, 5 ⁇ l/ml phenylethyl alcohol and 0.2 mg/ml propyl hydroxybenzoate.
  • the pH of the solution was 3.6, as mentioned above.
  • the osmolality of the solution was 0.46 osmol/kg.
  • Single dose nasal spray devices (Pfeiffer, Germany) were filled with the solution. Each device was filled with 123 ⁇ l of liquid. Actuation of the device delivered a dose of 100 ⁇ l of liquid containing 400 ⁇ g of buprenorphine and 2 mg of pectin.
  • pectin 5 g is dissolved by stirring into approximately 180 ml of WFI. 538 mg of buprenorphine hydrochloride and 12.5 g of dextrose are dissolved into the pectin solution. 1.25 ml of phenylethyl alcohol and 50 mg of propyl hydroxybenzoate are dissolved into the pectin/buprenorphine solution. The solution is adjusted to 250 ml using WFI.
  • the final product is a slightly turbid solution containing 2.16 mg/ml buprenorphine hydrochloride (corresponding to 2 mg/ml buprenorphine), 20 mg/ml pectin, 50 mg/ml dextrose, 5 ⁇ l/ml phenylethyl alcohol and 0.2 mg/ml propyl hydroxybenzoate.
  • 123 ⁇ l of the above solution is filled into a Valois Monospray single dose nasal spray device (Pfeiffer, Germany). Actuation of the device will deliver a dose of 100 ⁇ l of liquid containing 200 ⁇ g of buprenorphine and 2 mg of pectin.
  • HPMC Metal-heated (70-80° C.) water for injection
  • WFI water for injection
  • the HPMC dispersion was stirred in an ice bath until a clear solution had formed.
  • 1.25 g of chitosan glutamate Protosan (trade mark) UPG213, Pronova, Norway
  • 75 mg of 50% w/w benzalkonium chloride solution was dispersed in 10 ml of WFI and transferred with an additional 40 ml of WFI to a 250 ml volumetric flask.
  • the final product was a clear colourless solution containing a 4.3 mg/ml buprenophine hydrochloride (corresponding to 4 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 3 mg/ml HPMC, 50 mg/ml dextrose and 0.15 mg/ml benzalkonium chloride.
  • the osmolality of the final solution was 0.34 osmol/kg and the viscosity, as measured using a Brookfield CP70 cone and plate viscometer was 84.7 cps at 2.5 rpm and 25° C.
  • Single dose nasal spray devices (Pfeiffer, Germany) were filled with the solution. Each device was filled with 123 ⁇ l of liquid. Actuation of the device delivered a dose of 100 ⁇ l of liquid containing 400 ⁇ g of buprenorphine, 0.5 mg of chitosan and 0.3 mg of HPMC. Hence, a dose of 400 ⁇ g buprenorphine is provided by a single spray into one nostril. A dose of 800 ⁇ g is provided by a single spray into each nostril.
  • a solution containing HPMC, chitosan glutamate and benzalkonium chloride is prepared according to Example 3. 269 mg of buprenorphine hydrochloride and 12.5 g of mannitol (Sigma, UK) are transferred into the volumetric flask. The chitosan/HPMC solution and an additional 40 ml of WFI are added to the flask. The pH of the solution is adjusted to pH 3.6 using 1M hydrochloric acid solution and the flask contents adjusted to 250 ml using WFI.
  • the final product is a clear colourless solution containing 1.08 mg/ml buprenorphine hydrochloride (corresponding to 1 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 3 mg/ml HPMC, 50 mg/ml mannitol and 0.15 mg/ml benzalkonium chloride.
  • 123 ⁇ l of the above solution is filled into a single dose nasal spray device (Pfeiffer, Germany). Actuation of the device will deliver a dose of 100 ⁇ l of liquid containing 100 ⁇ g of buprenorphine, 0.5 mg of chitosan and 0.3 mg of HPMC.
  • the final product was a clear colourless solution containing 4.3 mg/ml buprenorphine hydrochloride (corresponding to 4 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 100 mg/ml poloxamer 188, 50 mg/ml dextrose and 0.15 mg/ml benzalkonium chloride.
  • the osmolality of the final solution was 0.60 Osmol/kg.
  • Single dose nasal spray devices (Pfeiffer, Germany) were filled with the solution. Each device was filled with 123 ⁇ l of liquid. Actuation of the device delivered a dose of 100 ⁇ l of liquid containing 400 ⁇ g of buprenorphine, 0.5 mg of chitosan and 10 mg of poloxamer 188.
  • a solution containing chitosan glutamate, poloxamer 188 and benzalkonium chloride is prepared according to Example 5. 269 mg of buprenorphine hydrochloride and 12.5 g mannitol (Sigma, UK) are transferred into the volumetric flask. The chitosan/poloxamer solution and an additional 40 ml of WFI are added to the flask. The pH of the solution is adjusted to pH 3.6 using 1M hydrochloric acid and the flask contents adjusted to 250 ml using WFI.
  • the final product is a clear colourless solution containing 1.08 mg/ml buprenorphine hydrochloride (corresponding to 1 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 100 mg/ml poloxamer 188, 50 mg/ml mannitol and 0.15 mg/ml benzalkonium chloride.
  • 123 ⁇ l of the above solution is filled into a single dose nasal spray device (Pfeiffer, Germany). Actuation of the device will deliver a dose of 100 ⁇ l of liquid containing 100 ⁇ g of buprenorphine, 0.5 mg of chitosan and 10 mg of poloxamer 188.
  • the viscosity of the solution was measured using a Brookfield Cone and Plate Rheometer. Results given are the mean of determinations at three rotation speeds appropriate to the viscosity of the solution.
  • the buprenorphine content of formulations was determined by hplc.
  • Gels were prepared by controlled mixing of 20 ml of formulation with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl 2 .2H 2 0) before standing for 1 hour at room temperature. A visual assessment of the structure, uniformity, clarity and evidence of syneresis of each gel was conducted and, in addition, the gel structure was examined with a Stable Microsystems Texture Analyser. Results (from single determinations) are expressed in terms of force (maximum penetration force) and area (total work of gel penetration).
  • Buprenorphine hydrochloride (107.5 mg) and anhydrous dextrose (1.25 g) were stirred in 18-20 ml water in a 25 ml volumetric flask together with an appropriate quantity of pectin and the mixture stirred overnight or until a solution formed. The mixture was then made up to 25 ml with water to give a solution containing 4 mg/ml buprenorphine, 50 mg/ml dextrose and 1, 5, 10, 20, 30, 40 or 80 mg/ml pectin and the pH, appearance, osmolality, viscosity were determined.
  • spray characteristics from a Pfeiffer multi-dose nasal spray device (standard nozzle, 0.1 ml pump, Cat. No.
  • Gels were prepared by controlled mixing of 20 ml of formulation with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl 2 .2H 2 0) before standing for 1 hour at room temperature. A visual assessment of the structure, uniformity, clarity and evidence of syneresis of each gel was conducted and, in addition, the gel structure was examined with a Stable Microsystems Texture Analyser.
  • SNES simulated nasal electrolyte solution
  • pectin produced mobile gels at 10-20 mg/ml and strong, inhomogeneous gels at higher concentrations.
  • gels were prepared by controlled mixing of 20 ml of formulation with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl 2 .2H 2 O) before standing for 1 hour at room temperature. A visual assessment of the structure, uniformity, clarity and evidence of syneresis of each gel was conducted and, in addition, the gel structure was examined with a Stable Microsystems Texture Analyser.
  • Buprenorphine was sparingly soluble (greater than 10 ng/ml) in aqueous solutions containing 20 mg/ml pectin+50 mg/ml dextrose at pH below 4.4 (Table 3). In general, solubility fell as pH increased above 4.5 (Table 3a). Solutions were slightly soluble (less than 10 ng/ml) at pH 4.5-6.0.
  • Buprenorphine hydrochloride (107.5 mg) and pectin (Slendid 100) (500 mg) were stirred in 18-20 ml water in a 25 ml volumetric flask together with an appropriate quantity of anhydrous dextrose or mannitol and the mixture stirred overnight or until a solution formed.
  • the mixture was then made up to 25 ml with water to give a solution containing 4 mg/ml buprenorphine, 20 mg/ml pectin and 15, 50, 87, 122, 157 or 192 mg/ml dextrose (or 15, 50, 87, 122 mg/ml mannitol) and the pH, appearance, osmolality, viscosity were determined.
  • Buprenorphine solubility in aqueous solution containing 20 mg/ml pectin was not affected significantly by dextrose (Table 9) or mannitol (Table 10) concentration across the measured pH range.
  • the pectins suitable for retaining drugs at mucosal surfaces have a low degree of esterification (also called “low methoxy” or “LM” pectins) and, in aqueous solution, will gel in the presence of ions found in mucosal fluid, especially divalent ions, in particular calcium.
  • LM low methoxy
  • a solution of “high methoxy” pectin was prepared and mixed with a solution containing calcium ions.
  • Buprenorphine hydrochloride (107.5 mg), anhydrous dextrose (1.25 g) and pectin (Genu pectin [citrus] type USP-H; CP Kelco, Lille Skenved, Denmark) (500 mg) were stirred in 18-20 ml water in a 25 ml volumetric flask overnight or until a solution formed. The mixture was then made up to 25 ml with water to give a solution containing 4 mg/ml buprenorphine, 20 mg/ml pectin and 50 mg/ml dextrose and the pH and osmolality were determined.
  • the solution had a pH of 3.3 and an osmolality of 0.35 osmol/kg. An opaque, pale yellow solution was formed when the solution was mixed with 9.44 mg/ml CaCl 2 .2H 2 0. The solution did not gel even when left for 1 hour at room temperature.
  • Unit doses of the intranasal buprenorphine formulations of Examples 1, 3 and (Formulations A to C) and one intravenous commercial buprenorphine formulation (Temgesic-trade mark; Formulation D) were administered to healthy human volunteers.
  • the unit doses administered to the volunteers were as follows:
  • the dosing was performed on twelve healthy volunteers using a randomised, complete crossover design. Each dose was separated by, at least, seven days. The volunteers were required to fast overnight prior to dosing. Subjects were admitted to a clinic the evening before each dose of administration and remained in the clinic until blood sample collection for each study day. Blood samples were collected at regular intervals up to 24 hours after each dose administration. The volunteers were discharged from the clinic after completion of all 24 hour study procedures. There was a wash out period of, at least seven days, between each dose.
  • FIG. 4 also shows the pharmacokinetic profile for the 400 ⁇ g dose of Formulation A that was administered intravenously.

Abstract

An analgesic and a delivery agent are combined in a pharmaceutical composition such that, on introduction into the nasal cavity of a patient to be treated, the analgesic may be delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration, Cther, of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours. The analgesic may be an opioid analgesic or a non-steroidal anti-inflammatory drug.

Description

    FIELD OF THE INVENTION
  • The invention relates to analgesic compositions and their use.
  • BACKGROUND OF THE INVENTION
  • A wide variety of compounds can act as analgesics. Two important classes of analgesics are opioid analgesics and non-steroidal anti-inflammatory drugs (NSAIDs).
  • Opioid analgesics exhibit morphine-like properties. Opioids can be sub-classified on the basis of their receptor specificity. Mu-agonist opioids provide intense analgesia. These opioids can be long-acting (e.g. methadone) or short-acting (e.g. remifentanil). Mixed agonist/antagonist opioids (e.g. butorphanol and buprenorphine) are partial agonists (the former at mu and kappa receptors and the latter at the mu receptor) and can produce good quality analgesia. They produce less respiratory depression and constipation than high efficacy mu agonists.
  • As a class, opioids are associated with a number of undesirable side-effects, including respiratory depression, nausea, vomiting, dizziness, mental clouding, dysphoria, pruritus, constipation, increased biliary tract pressure, urinary retention and hypotension. The development of tolerance and the risk of chemical dependence and abuse are further problems. Buprenorphine, however, is unusual in exhibiting a low maximum effect for respiratory depression and also a bell-shaped dose response curve where the effect first increases with larger doses, reaches a ceiling and then diminishes as the dosage is further increased, which makes it a safer drug than morphine, where respiratory depression will ultimately lead to death. Buprenorphine has also been shown to have a lower incidence of other side-effects like constipation in man, and it has a lower abuse potential than full mu agonists.
  • NSAIDs have anti-inflammatory action and are effective on pain associated with the release of prostaglandins (PG) and other mediators of inflammation. They act by blocking the action of cyclooxygenase (COX), which converts arachidonic acid to eicosanoids. The eicosanoids include the prostanoids, prostacyclin (PGI2), PGE2 and the thromboxanes. There are at least two COX enzymes: a constitutively-expressed COX-1 responsible for producing homeostatic prostaglandin and thromboxane mediators and an inducible COX-2 that is produced in large quantities in response to stimuli such as infection and inflammation.
  • Since the prostaglandins and thromboxanes mediate a number of homeostatic and protective mechanisms, toxic side effects often arise from the use of NSAIDs as a result of disruption of these mechanisms. These include clotting disorders (leading to prolonged bleeding times) and gastric irritation (including ulceration). NSAIDs may also cause salt and water retention and may therefore exacerbate hypertension. They may also be teratogenic at high doses during pregnancy. They are contra-indicated in patients with peptic ulcers, gastritis, regional enteritis, ulcerative colitis, diverticulitis, a recurrent history of gastrointestinal lesions, gastrointestinal bleeding, coagulation disorders (such as anemia, hypoprothrombinemia and haemophilia), kidney diseases and in patients about to undergo surgery or taking anticoagulants.
  • The NSAIDs are associated with a number of adverse effects on the kidneys, although most are rare. The kidney produces PGI2, PGE2 and some PGF. These are involved in local modulation of renal blood flow, glomerular filtration rate, renin release, the concentrating mechanism for urine and the excretion of sodium and potassium. The unwanted effects of NSAIDs result from the decrease in production of the prostaglandins and are summarized below:
      • 1. Acute reversible/vasomotor renal failure.
      • 2. Interference with the renal excretion of water, sodium and potassium.
      • 3. Interference with antihypertensive therapy and diuretic therapy.
      • 4. Acute interstitial nephritis with or without renal failure.
      • 5. Nephrotic syndrome with or without interstitial nephritis and renal failure.
      • 6. Chronic renal injury (“analgesic nephropathy”).
  • There is great interest in the development of NSAIDs that are COX-2 specific, since such drugs would be expected to permit the treatment of inflammation and pain without affecting COX-1-mediated gastrointestinal protection. However, COX-2 inhibitors still show the renal and cardiac effects of non-selective NSAIDs.
  • Ideally, pain relief should follow immediately upon administration of an analgesic. The relief should be maintained for an extended period that is at least long enough to permit normal unbroken sleep patterns and avoid complicated dosage regimes.
  • In practice, however, the dynamics of pain relief obtained with current analgesic administration technologies does not meet these ideals. While rapid onset of pain relief can be achieved by intravenous injection, this mode of administration cannot in general be carried out by the patient and so is relatively expensive and inconvenient. Moreover, intravenous injection is generally associated with rapid offset of pain relief as the circulating analgesic is cleared from the plasma. Prolonged analgesia requires multiple injections which is inconvenient and expensive. Intravenous injection is also usually associated with relatively high Cmax values, which can trigger (or amplify) any side effects associated with the analgesic.
  • While alternative technologies (including intramuscular injection and inhalation) have been developed for effecting rapid onset analgesia, these all rely upon rapid delivery of the bulk of the analgesic dose into the blood system and so suffer from the same rapid offset problems associated with intravenous injection.
  • Attempts have been made to obviate such problems by providing pumped analgesic into the blood supply via a patient-controlled quick-dose pump. While this apparatus has the potential for long-term effective pain management, it is expensive, does not permit ambulation, requires extensive monitoring and may interfere with normal sleep patterns (depending on the frequency with which pain prompts the patient to re-dose).
  • The problem of rapid offset of pain relief has promoted the development of sustained release technologies. Such technologies include transdermal patches and sublingual tablets. However, transdermal patches can cause skin irritation and the drug dosage is difficult to control. Sublingual tablets have an unpleasant taste and must be maintained in the mouth for relatively long periods of time (often 30 minutes or more), leading to compliance problems.
  • However, the principal problem associated with such sustained analgesia techniques stems from the fact that the onset of pain relief is slow and associated with a lag time of at least an hour (during which plasma levels of the analgesic steadily climb towards the therapeutic concentration threshold). In many applications (especially in cases where pain is intense and prolonged) such pain relief dynamics are unacceptable.
  • Buprenorphine has previously been administered via the intravenous, intramuscular and sublingual routes to human subjects. There are limited reports of nasal administration. Eriksen et al, J. Pharm. Pharmacol. 41, 803-805, 1989 report administration to human volunteers of a nasal spray. The spray consisted of 2 mg/ml of buprenorphine hydrochloride dissolved in 5% dextrose and the pH of the solution was adjusted to pH 5.
  • WO 90/09870 describes a composition for administration to mucosa comprising a pharmacologically active compound and a polycationic substance such as DEAE-dextran or chitosan. WO 98/47535 discloses a single component liquid pharmaceutical composition for administration to a mucosal surface. The composition comprises a therapeutic agent, a pectin with a low degree of esterification and an aqueous carrier that gels or can be adapted to gel at the site of application. Neither WO 90/09780 nor WO 98/47535 mentions buprenorphine.
  • SUMMARY OF THE INVENTION
  • Improved analgesic formulations for nasal administration have now been devised. Rapid uptake of the analgesic across the nasal mucosa into the plasma can be achieved, which results in fast onset of analgesia. Further, the residence time of the analgesic in the nasal cavity can be increased, which results in prolonged analgesia. An improved profile of absorption of the analgesic into the systemic circulation can thus be achieved.
  • Accordingly, the present invention provides use of an analgesic and a delivery agent for the manufacture of a medicament for administration intranasally for the treatment of pain whereby, on introduction into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration Cther of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours.
  • The invention thus enables a therapeutic blood plasma concentration of an analgesic, i.e. a concentration that produces pain relief or pain amelioration, to be attained within 30 minutes and maintained for up to 24 hours. The term Cther denotes a therapeutic blood plasma concentration. The term Tmaint denotes the duration for which Cther is maintained. Also provided are:
      • use of a pharmaceutical composition which comprises an analgesic and a delivery agent for the manufacture of a nasal delivery device for use in inducing analgesia whereby, on introduction into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration Cther of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours;
      • a pharmaceutical composition which comprises an analgesic and a delivery agent whereby, on introduction into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration Cther of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours; and
      • a method of inducing analgesia in a patient in need thereof, which method comprises administering intranasally to said patient a pharmaceutical composition which comprises an analgesic and a delivery agent whereby, on introduction into the nasal cavity of said patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration Cther of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours.
  • The invention further provides:
      • (1) an aqueous solution suitable for intranasal administration, which comprises:
        • (a) from 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof,
        • (b) from 0:1 to 20 mg/ml of a chitosan, and
        • (c) from 0.1 to 15 mg/ml of hydroxypropylmethylcellulose (HPMC);
        • which solution has a pH of from 3 to 4.8;
      • (2) an aqueous solution suitable for intranasal administration, which comprises:
        • (a) from 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof,
        • (b) from 0.1 to 20 mg/ml of a chitosan, and
        • (c) from 50 to 200 mg/ml of a polyoxyethylene-polyoxypropylene copolymer of the general formula HO(C2H4O)a(C3H6O)b(C2H4O)aH wherein a is from 2 to 130 and b is from 15 to 67;
        • which solution has a pH of from 3 to 4.8; and
      • (3) an aqueous solution suitable for intranasal administration, which comprises from 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof and from 5 to 40 mg/ml of a pectin having a degree of esterification of less than 50%; which solution has a pH of from 3 to 4.2, is substantially free from divalent metal ions and gels on the nasal mucosa.
        • A preferred solution of the invention has a pH of from 3.5 to 4.0, is substantially free from divalent metal ions and comprises:
        • (a) from 1 to 6 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, calculated as buprenorphine,
        • (b) from 10 to 40 mg/ml of a pectin which has a degree of esterification from 10 to 35%, and
        • (c) dextrose as a tonicity adjustment agent.
  • The invention also provides:
      • a process for the preparation of solution (1), which comprises dissolving buprenorphine or a physiologically acceptable salt or ester thereof, a chitosan and HPMC in water to provide a solution comprising from 0.1 to 10 mg/ml of buprenorphine or said salt or ester thereof, from 0.1 to 20 mg/ml of chitosan and from 0.1 to 15 mg/ml of HPMC; and adjusting the pH of the solution to a value from 3 to 4.8 as desired;
      • a process for the preparation of solution 2), which comprises dissolving buprenorphine or a physiologically acceptable salt or ester thereof, a chitosan and a polyoxyethylene-polyoxypropylene copolymer of the general formula HO(C2H4O)a(C3H6O)b(C2H4O)aH wherein a is from 2 to 130 and b is from 15 to 67, in water to provide a solution comprising from 0.1 to 10 mg/ml of buprenorphine or said salt or ester thereof, from 0.1 to 20 mg/ml of a chitosan and from 50 to 200 mg/ml of the polyoxyethylene-polyoxypropylene copolymer; and adjusting the pH of the solution to a value from 3 to 4.8 as desired;
      • a process for the preparation of solution (3), which comprises dissolving buprenorphine or a physiologically acceptable salt or ester thereof in water; mixing the resulting solution with a solution in water of a pectin having a degree of esterification of less than 50% such that the mixed solution comprises from 0.1 to 10 mg/ml of buprenorphine or said salt or ester thereof and from 5 to 40 mg/ml of the pectin; and adjusting the pH of the solution to a value from 3 to 4.2 if desired;
      • a nasal delivery device loaded with a solution of the invention;
      • use of a solution of the invention for the manufacture of a nasal delivery device for use in inducing analgesia; and
      • a method of inducing analgesia in a patient in need thereof, which method comprises intranasally administering a solution of the invention to the patient.
    BRIEF DESCRIPTION OF DRAWINGS
  • FIGS. 1 to 3 show the pharmacokinetic profiles that were obtained when buprenorphine formulations according to the invention (Formulations A to C) were administered intranasally to healthy volunteers at a dose of 800 μg of buprenorphine hydrochloride, calculated as buprenorphine. Formulation A: buprenorphine hydrochloride-pectin solution. Formulation B: buprenorphine hydrochloride-chitosan/hydroxypropylmethylcellulose (HPMC) solution. Formulation C: buprenorphine hydrochloride-chitosan/poloxamer 188 solution. Also shown for comparison is the pharmacokinetic profile that was obtained when a commercial solution of buprenorphine hydrochloride (Terngesic—trade mark; Formulation D) was administered intravenously to healthy volunteers in the same study at a dose of 400 μg of buprenorphine hydrochloride, calculated as buprenorphine.
  • FIG. 4 shows a pharmacokinetic profile for a 400 μg dose of Formulation A. This profile was calculated from the data for the 800 μg dose of Formulation A. The pharmacokinetic profile for the 400 μg dose of Formulation D is also shown for comparison.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention is concerned with analgesic compositions that are delivered intranasally to achieve fast onset of analgesia and also prolonged analgesia. An advantageous pharmacokinetic profile can therefore be attained. An analgesic is combined with a delivery agent which is selected so that, on introduction into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration Cther of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours. This effect may be achieved by administration of a single dose of the analgesic, or following multiple dosing.
  • The compositions of the invention are adapted to enable the analgesic to be delivered such that a Cther of 0.2 ng/ml or more, for example 0.4 ng/ml or more, is attained within 30 minutes after introduction into the nasal cavity such as within 0.5 to 20 minutes, for example 2 to 15 minutes or 5 to 10 minutes. The term Cther defines a therapeutic plasma concentration (or range thereof). Thus, the term is used herein to define a blood plasma concentration (or range of plasma concentrations) of the analgesic that produces pain relief or pain amelioration.
  • Depending upon the analgesic, Cther may be up to 100 ng/ml such as from 0.4 ng/ml to 80 mg/ml. For potent analgesics, Cther may be from 0.4 ng/ml to 20 ng/ml. For less potent analgesics, Cther may be from 20 to 100 ng/ml such as from 50 to 80 ng/ml. These Cther values apply especially to opioid analgesics. For the potent opioid analgesic buprenorphine, Cther may be from 0.4 to 5 ng/ml, for example 0.5 to 4 ng/ml or 0.8 to 2 ng/ml.
  • Tmaint is typically at least 2 hours. The term Tmaint defines the duration of maintenance of Cther after administration of the analgesic. For example, the Tmaint can be from up to 24 hours, up to 12 hours or up to 6 hours such as from 2 to 4 hours or 2 to 3 hours. By means of the invention, therefore, a Cther of at least 0.4 ng/ml may be attained within 2 to 15′ minutes and maintained for a time period Tmaint of from 2 to 4 hours.
  • As already mentioned, rapid onset of analgesia and prolonged analgesia can be achieved. The analgesic delivery profile that can be attained may avoid the relatively high Cmax values associated with intravenous administration and so lead to an improved therapeutic index. The peak plasma concentration of an analgesic that is attained after administration is defined as Cmax. The invention can permit reduction or elimination of some or all of the side effects associated with the analgesic.
  • Cmax depends upon the analgesic. Cmax is typically from 1 to 500 ng/ml or higher, for example from 1.5 to 400 ng/ml or from 1.5 to 100 ng/ml. For potent analgesics, Cmax may be from 1.5 to 50 ng/ml. For less potent analgesics, Cmax may be from 50 to 500 ng/ml such as from 50 to 200 ng/ml or 50 to 100 ng/ml. These Cmax values apply especially to opioid analgesics. For buprenorphine, Cmax is typically from 1 to 5 ng/ml, for example from 1 to 4 ng/ml or from 1.5 to 3 ng/ml. Cmax may be from 1 to 2 ng/ml, especially for lower doses. The time at which Cmax is reached (Tmax) is typically 10 to 40 minutes after administration, for example 10 to 30 minutes or 15 to 25 minutes such as 15 to 20 minutes.
  • In preferred embodiments, the delivery agent is adapted to deliver the analgesic component such that Cmax=Copt. The term Copt is used in relation to analgesic drugs which exhibit a dose-response curve to analgesia which is displaced to the left with respect to the dose-response curve for side-effects. The term defines a therapeutic plasma concentration or range thereof which produces acceptable pain relief or pain amelioration but which does not produce side-effects or produces side effects which are less than those associated with higher plasma concentrations.
  • The analgesic may be an opioid analgesic. It may be a mixed agonist/antagonist such as a mixed mu-agonist/antagonist (also known as a partial agonist) or a mixed mu- and kappa-agonist/antagonist. It may be a mu-agonist. A preferred opioid analgesic is buprenorphine or a physiologically acceptable salt or ester thereof. Buprenorphine (Chemical Abstracts Registration No. (CAS RN) 52485-79-7; [5α,7α(S)-17-(cyclopropylmethyl)-α-(1,1-dimethylethyl)-4,5-epoxy-18,19-dihydro-3-hydroxy-6-methoxy-α-methyl-6,14-ethenomorphinan-7-methanol) has the formula:
  • Figure US20110105551A1-20110505-C00001
  • The buprenorphine salt may be an acid addition salt or a salt with a base. Suitable acid addition salts include the hydrochloride, sulphate, methane sulphonate, stearate, tartrate and lactate salts. The hydrochloride salt is preferred (CAS RN 53152-21-9)
  • Other suitable opioid analgesics include alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, butorphanol, clonitazene, cyclazocine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetylbutyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene fentanyl, heroin, hydromorphone, hydroxypethidine, isomethadone, ketobemidone, levallorphan, levorphanol, levophenacylmorphan, lofentanil, meperidine, meptazinol, metazocine, methadone, metopon, morphine, myrophine, nalbuphine, narceine, nicomorphine, norlevorphanol, normethadone, nalbuphine, nalorphine, naloxone, naltrexone, normorphine, norpipanone, opium, oxycodone, oxymorphone, papavereturn, pentazocine, phenadoxone, phenomorphan, phenazocine, phenoperidine, piminodine, piritramide, profadol, propheptazine, promedol, properidine, propiram, propoxyphene, sufentanil, tilidine and tramadol. Also included are esters, salts and mixtures of any of the foregoing.
  • The salts for use in the invention may be any physiologically acceptable salts, including pharmaceutically acceptable acid addition salts. Examples include hydrochloride salts (for example the hydrochloride salts of nalbuphine, profadol, buprenorphine, morphine, pentazocine, naloxone and nalorphine) as well as levorphanol tartrate, nalorphine hydrobromide, levallorphan tartrate, morphine sulfate, butorphanol tartrate, pentazocine lactate and phenazocine hydrobromide.
  • The analgesic may be a non-opioid analgesic such as a NSAID, a tricylic antidepressant (e.g. amitryptyline), an anticonvulsant (e.g. gabapentin) or an antimigraine compound (e.g. sumatriptan or naratriptan). The NSAID may be a cyclooxygenase (COX) COX-1 or COX-2 inhibitor. Specific examples of NSAIDs include ibuprofen, flurbiprofen, diclofenac, indomethacin, piroxicam, ketoprofen, etodolac, diflusinal, meloxicam, aceclofenac, fenoprofen, naproxen, tiaprofenic acid, tolmetin, celecoxib and rofecoxib, and their physiologically acceptable salts and esters. Suitable salts are alkali addition salts such as the potassium or sodium salt.
  • A preferred NSAID is diclofenac (CAS RN 15307-86-5; 2-[(2,6-dichlorophenyl)amino]benzeneacetic acid) or a physiologically acceptable salt or ester thereof. Diclofenac has the formula:
  • Figure US20110105551A1-20110505-C00002
  • Diclofenac is available in various forms. Diclofenac Sodium (CAS RN 15307-79-60; 2-[(2,6-dichlorophenyl)amino]benzeneacetic acid sodium salt) is sold under the following trade marks: Allorvan, Benfofen, Dealgic, Deflamat, Delphinac, Diclomax, Diclometin, Dichlophlogont, Diclo-Puren, Dicloreum, Diclo-Spondyril, Dolobasan, Duravolten, Ecofenac, Effekton, Lexobene, Motifene, Neriodin, Novapirina, Primofenac, Prophenatin, Rewodina, Rhumalgan, Trabona, Tsudohmin, Valetan, Voldal, Voltaren and Xenid. Diclofenac Potassium (CAS RN 15307-81-0; 2-[(2,6-dichlorophenyl)amino]benzeneacetic acid potassium salt) is also known as Cataflam (trade mark).
  • Another preferred NSAID is etodolac (CAS RN 41340-25-4; 1,8-Diethyl-1,3,4,9-tetrahydropyrano[3,4-b]indole-1-acetic acid) or a physiologically acceptable salt or ester thereof. Etodolac is sold under the trade marks Etogesic, Lodine, Tedolan and Ultradol. Etodolac has the formula:
  • Figure US20110105551A1-20110505-C00003
  • Further preferred NSAIDs are piroxicam and meloxicam, and physiologically acceptable salts and esters thereof. Piroxicam (CAS RN 36322-90-4; 4-Hydroxy-2-methyl-N-2-pyridinyl-2H-1,2-benzothiazine-3-carboxamide. 1,1-dioxide) is sold under the trade mark Feldine, Sinartrol, Zelis, Zen, Brexin, Cicladol or Cycladol and has the formula:
  • Figure US20110105551A1-20110505-C00004
  • Meloxicam (CAS RN 71125-38-7; 4-hydroxy-2-methyl-N-(5-methyl-2-thiazolyl)-2H-1,2-benzothiazine-3-carboxamide 1,1-dioxide has the formula:
  • Figure US20110105551A1-20110505-C00005
  • The analgesic is provided in a formulation suitable for nasal administration in combination with a delivery agent. The formulation is typically a liquid formulation, especially as an aqueous solution. Alternatively, the formulation may be in the form of a powder or microspheres.
  • When the formulation is a liquid formulation, the concentration of buprenorphine or buprenorphine salt or ester is from 0.1 to 10 mg/ml, for example from 0.5 to 8 mg/ml. Preferred concentrations are 1 to 6 mg/ml, for example 1 to 4 mg/ml calculated as buprenorphine. Suitable formulations can contain buprenorphine or a buprenorphine salt or ester in an amount of 1 mg/ml or 4 mg/ml, calculated as buprenorphine.
  • The delivery agent is selected so that rapid onset and prolonged analgesia is obtained. The delivery agent acts to deliver the analgesic to the bloodstream. Thus, the delivery agent acts as an analgesic absorption modifier and any of a wide variety of delivery agents may be used providing that this functional requirement is met.
  • The delivery agent may comprise an absorption promoting agent. Such agents promote uptake of the analgesic component into the bloodstream. They may act via a variety of different mechanisms. Particularly preferred are mucosal adhesives. Such adhesives maintain an intimate association between the bulk analgesic composition and the nasal mucosa, so enhancing absorption and extending the Tmaint of the analgesic component. They can also be used to lower the analgesic Cmax which may be important in applications where the minimization or elimination of side-effects is desired.
  • Suitable absorption promoting agents include cationic polymers (particularly chitosans), surface active agents, fatty acids, chelating agents, mucolytic agents, cyclodextrins, diethylaminoethyl-dextran (DEAE-dextran; a polycationic derivative of dextran) or combinations thereof. Particularly preferred are pectins having a degree of esterification of less than 50%, especially from 10 to 35%, and chitosans.
  • The composition of the invention takes the form of an aqueous solution when the delivery agent is a pectin. The pectin acts as a gelling agent. A pectin-containing composition of the invention gels on the mucosal surfaces of the nasal cavity after delivery without the need for an extraneous source of divalent metal ions. The analgesic that is formulated with the pectin is thus retained for longer on the surfaces of the nasal epithelium. The resulting sustained release of the analgesic into the bloodstream enables prolonged analgesia to be achieved. Improved delivery of the analgesic can consequently be obtained. Rapid uptake of the analgesic also results, which leads to fast onset of analgesia.
  • The pectins employed in the invention have a degree of esterification of less than 50%. A pectin is a polysaccharide substance present in the cell walls of all plant tissues. Commercially pectins are generally obtained from the dilute acid extract of the inner portion of the rind of citrus fruits or from apple pomace. A pectin consists of partially methoxylated polygalacturonic acids. The proportion of galacturonic acid moieties in the methyl ester form represents the degree of esterification (DE). The term DE is well understood by those skilled in the art and may be represented as the percentage of the total number of carboxyl groups that are esterified, i.e. if four out of five acid groups is esterified this represents a degree of esterification of 80%, or as the methoxyl content of the pectin. DE as used herein refers to the total percentage of carboxyl groups that are esterified. Pectins can be categorised into those having a low degree of esterification (low methoxylation) or a high degree of esterification (high methoxylation). A “low DE” or “LM” pectin has a degree of esterification below 50% whereas a “high DE” or “HM” pectin has a degree of esterification of 50% or above. The gelling properties of aqueous pectin solutions can be controlled by the concentration of pectin, the type of pectin, especially the degree of esterification of the galacturonic acid units, and the presence of added salts.
  • Low DE pectins are used in the present invention. The primary mechanism by which such pectins gel in aqueous solution is through exposure to metal ions, such as those found in the nasal mucosal fluid as described in WO 98/47535. The degree of esterification of the pectin used in the invention is preferably less than 35%. The degree of esterification may thus be from 10 to 35%, for example from 15 to 25%. Low DE pectins may be purchased commercially. An example of a low DE pectin is SLENDID (trade mark) 100, supplied by CP Kelco (Lille Skenved) which has a degree of esterification of around 15 to 25%.
  • A pectin-containing solution of the invention must not gel on storage. It should not gel prior to application to the nasal cavity. It must therefore be substantially free of agents which would cause the solution to gel. In particular, a solution of the invention must be substantially free of divalent metal ions and especially calcium ions. The content of divalent metal ions in the solution must therefore be minimised. A pectin-containing solution of the invention may therefore contain a negligible concentration of divalent metal ions or there may no detectable divalent metal ions.
  • A pectin is typically present in a solution of the invention at a concentration of from 5 to 40 mg/ml, for example from 5 to 30 mg/ml. More preferably, the pectin concentration is from 10 to 30 ng/ml or from 10 to 25 mg/ml. The pectin and the pectin concentration are selected such that the solution gels on delivery to the nasal mucosa. The solution gels on the nasal mucosa in the absence of an extraneous source of divalent metal ions, e.g. Ca2+ ions.
  • The composition of the invention also takes the form of an aqueous solution when the delivery agent is a chitosan. Chitosans are cationic polymers that have mucoadhesive properties. The mucoadhesion is thought to result from an interaction between the positively charged chitosan molecule and the negatively charged sialic acid group's on mucin (Soane et al, Int. J. Pharm 178, 55-65, 1999).
  • By the term “chitosan” we include all derivatives of chitin, or poly-N-acetyl-D-glucosamine, including all polyglucosamines and oligomers of glucosamine materials of different molecular weights, in which the greater proportion of the N-acetyl groups have been removed through hydrolysis (deacetylation). Preferably, the chitosan is produced from chitin by deacetylation to a degree of greater than 40%, preferably between 50 and 98%, more preferably between 70% and 90%.
  • The chitosan typically has a molecular weight of 4,000 Da or more, preferably from 10,000 to 1,000,000 Da, more preferably from 15,000 to 750,000 Da and most preferably from 50,000 to 500,000 Da.
  • The chitosan may thus be a deacetylated chitin. It may be a physiologically acceptable salt. Suitable physiologically acceptable salts include salts with a pharmaceutically acceptable mineral or organic acid such as the nitrate, phosphate, lactate, citrate, hydrochloride and acetate salts. Preferred salts are chitosan glutamate and chitosan hydrochloride.
  • The chitosan may be a derivative of a deacetylated chitin. Suitable derivatives include, but are not limited to, ester, ether or other derivatives formed by bonding of acyl and/or alkyl groups with the hydroxy groups, but not the amino groups, of a deacetylated chitin. Examples are O—(C1-C6 alkyl)ethers of deacetylated chitin and O-acyl esters of deacetylated chitin. Derivatives also include modified forms of a deacetylated chitin for example a deacetylated chitin conjugated to polyethylene glycol.
  • Low and medium viscosity chitosans suitable for use in the present invention may be obtained from various sources, including FMC Biopolymer, Drammen, Norway; Seigagaku America Inc., Maryland, USA; Meron (India) Pvt, Ltd., India; Vanson Ltd, Virginia, USA; and AMS Biotechnology Ltd., UK. Suitable derivatives include those that are disclosed in Roberts, Chitin Chemistry, MacMillan Press Ltd., London (1992). Particularly preferred chitosan compounds that may be mentioned include “Protosan” (trade mark) available from FMC Biopolymer, Drammen, Norway. The chitosan is preferably water-soluble.
  • An aqueous solution of chitosan may be prepared by dissolving chitosan base or a derivative of chitosan base in a pharmaceutically acceptable mineral or organic acid such as hydrochloric, lactic, citric or glutamic acid or by dissolving a chitosan salt in water.
  • The chitosan is typically present in solution at a concentration of from 0.1 to 20 mg/ml, for example from 0.5 to 20 mg/ml. Preferably the solution contains from 1 to 15 mg/ml, more preferably from 2 to 10 mg/ml, of chitosan. A chitosan concentration of 5 mg/ml is particularly suitable.
  • Chitosan-containing solutions of the invention may also contain hydroxypropylmethylcellulose (HPMC) or a polyoxyethylene-polyoxypropylene copolymer.
  • Any suitable HPMC may be employed. Several grades of HPMC are available. For example, Dow Chemical Company produces a range of HPMC polymers under the trade mark Methocel. The grade and concentration of HPMC is chosen such that the solution of the invention preferably has a viscosity, at 25° C. as measured by a cone and plate viscometer (e.g. Brookfield), in the range from 1 to 200 cps, more preferably from 3 to 150 cps and most preferably from 5 to 100 cps.
  • Producing a solution having a particular viscosity is within the capability of one skilled in the at and can be achieved, for example, by using a high concentration of a low viscosity HPMC or a low concentration of a high viscosity HPMC. The HPMC is preferably one having an apparent viscosity (measured as a 2% solution in water at 20° C.) in the range from 3000 to 6000 cps. The concentration of the HPMC having a viscosity of from 3000 to 6000 cps is typically in the range from 0.1 to 15 mg/ml, for example from 0.5 to 10 mg/ml and preferably from 1 to 5 mg/ml.
  • The polyoxyethylene-polyoxypropylene copolymer typically has a molecular weight of from 2,500 to 18,000 for example from 7,000 to 15,000. The copolymer is a block copolymer of the general formula:

  • HO(C2H4O)a(C3H6O)b(C2H4O)aH
  • wherein a is from 2 to 130 and b is from 15 to 67. The value for a may be from 40 to 100 such as from 60 to 90 or from 70 to 95. The value for b may be from 20 to 40 such as from 25 to 35.
  • Such copolymers are known as poloxamers. Several different types of poloxamer are available commercially, from suppliers such as BASF, and vary with respect to molecular weight and the proportions of ethylene oxide “a” units acid propylene oxide “b” units. A commercially available poloxamer suitable for use in the present invention is poloxamer 188 which structurally contains 80 “a” units and 27 “b” units and has a molecular weight of 7680-9510 (Handbook of Pharmaceutical Excipients, editor A. H. Kippe, third edition, Pharmaceutical Press, London, UK, 2000). Preferably the poloxamer is poloxamer 188.
  • The polyoxyethylene-polyoxypropylene copolymer is typically present in an amount of from 50 to 200 mg/ml, preferably from 65 to 160 mg/ml and more preferably from 80 to 120 mg/ml. A preferred concentration is 100 mg/ml.
  • Other cationic polymers besides chitosans suitable for use as absorption promoting agents include polycationic carbohydrates. The polycationic substances preferably have a molecular weight of at least 10,000. They may be in liquid formulations at concentrations of 0.01 to 50% w/v, preferably 0.1 to 50% w/v and more preferably 0.2 to 30% w/v.
  • Examples of suitable polycationic polymers are polyaminoacids (e.g. polylysine), polyquaternary compounds, protamine, polyamine, DEAE-imine, polyvinylpyridine, polythiodiethyl-aminomethylethylene, polyhistidine, DEAE-methacrylate, DEAF-acrylamide, poly-p-aminostyrene, polyoxethane, co-polymethacrylates (e.g. copolymers of HPMA, N-(2-hydroxypropyl)-methacrylamide), GAFQUAT (see for example U.S. Pat. No. 3,910,862) and polyamidoamines.
  • Suitable surface active agents for use according to the present invention are bile salts (for example sodium deoxycholate and cholylsarcosine, a synthetic N-acyl conjugate of cholic acid with sarcosine [N-methylglycine]). Also suitable for use in the invention are bile salt derivatives (for example sodium tauro dihydrofusidate). Any of a wide range of non-ionic surfactants (e.g. polyoxyethylene-9 lauryl ether), phospholipids and lysophosphatidyl compounds (e.g. lysolecithin, lysophosphatidyl-ethanolamine, lysophosphatidylcholine, lysophosphatidylglycerol, lysophosphatidylserine and lysophosphatidic acid) may also be used. Water-soluble phospholipids may also be employed (e.g. short chain phosphatidylglycerol and phosphatidylcholines). The concentration of surface active agents used according to the invention varies according to the physico-chemical properties of the surface active agent selected, but typical concentrations are in the range 0.02 to 10% w/v.
  • Particularly preferred surface active agents for use as absorption promoting materials are phospholipids and lysophosphatides (hydrolysis products of phospholipids), both of which form micellar structures.
  • When microspheres are used as the delivery agent, they are preferably prepared from a biocompatible material that will gel in contact with the mucosal surface: Substantially uniform solid microspheres are preferred. Starch microspheres (crosslinked if necessary) are preferred.
  • Microspheres may also be prepared from starch derivatives, modified starches (such as amylodextrin), gelatin, albumin, collagen, dextran and dextran derivatives, polyvinyl alcohol, polylactide-co-glycolide, hyaluronic acid and derivatives thereof (such as benzyl and ethyl esters), gellan gum and derivatives thereof (such as benzyl and ethyl esters) and pectin and derivatives thereof (such as benzyl and ethyl esters). The term “derivative” covers inter alia esters and ethers of the parent compound, which can be functionalised (for example to incorporate ionic groups).
  • Any of a wide variety of commercially available starch derivatives may be used, including hydroxyethyl starch, hydroxypropyl starch, carboxymethyl starch, cationic starch, acetylated starch, phosphorylated starch, succinate derivatives of starch and grafted starches.
  • Suitable dextran derivatives include, diethylaminoethyl-dextran (DEAE-dextran), dextran sulphate, dextran methyl-benzylamide sulphonates, dextran methyl-benzylamide carboxylates, carboxymethyl dextran, diphosphonate dextran, dextran hydrazide, palmitoyldextran and dextran phosphate.
  • The preparation of microspheres for use according to the invention may be carried out by known processes, including emulsion and phase separation methods (see for example Davis et al., (Eds), “Microspheres and Drug Therapy”, Elsevier Biomedical Press, 1984, which parts relating to microsphere preparation are incorporated herein by reference). For example, albumin microspheres may be made using the water-in-oil emulsification method where a dispersion of albumin in oil is produced by homogenization or stirring, with the addition if necessary of small amounts of an appropriate surface active agent.
  • The size of the microspheres is largely determined by the speed of stirring or the homogenization conditions. Agitation can be provided by a simple laboratory stirrer or by more sophisticated devices (such as microfluidizers or homogenisers). Emulsification techniques may also be used to produce starch microspheres (as described in GB 1518121 and EP 223303) and for the preparation of gelatin microspheres.
  • Proteinaceous microspheres may be prepared by coacervation methods. Such methods include simple or complex coacervation as well as phase separation techniques (using solvents or electrolyte solutions). Such methods are well known to those skilled in the art and details may be found in standard textbooks (for example Florence and Attwood, Physicochemical Principles of Pharmacy 2nd Ed., MacMillan Press, 1988, Chapter 8).
  • The microspheres may advantageously have controlled-release properties, which may be conferred by modifications of the microspheres (for example by controlling the degree of cross-linking or by the incorporation of excipients that alter the diffusional properties of the analgesic component). Alternatively, controlled release properties may be incorporated by exploiting ion-exchange chemistry (for example DEAE-dextran and chitosan are positively charged and can be used for an ion-exchange interaction with metabolites that are negatively charged).
  • The maximum amount of analgesic component that can be carried by the microspheres is termed the loading capacity. It is determined by the physico-chemical properties of the analgesic component and in particular its size and affinity for the matrix of the microspheres. High loading capacities can be achieved when the analgesic is incorporated into the microspheres during microsphere manufacture.
  • Microcapsules (which may be bioadhesive and which may also exhibit controlled release properties) may also be employed as an absorption promoting agent in the compositions of the invention. These microcapsules can be produced by a variety of methods. The surface of the capsule may be inherently adhesive or can be modified by standard coating methods known to those skilled in the art. Suitable coating materials include bioadhesive polymers such as polycarbophil, carbopol, DEAE-dextran, alginate, microcrystalline cellulose, dextran, polycarbophils and chitosan).
  • Oil-in-water formulations can provide for the effective nasal delivery of analgesics that are poorly soluble in water. In such applications nasal irritation may also be reduced.
  • The oil phase of the emulsions of the invention may comprise a hydroxylated oil, particularly a hydroxylated vegetable oil. As used herein the term “hydroxylated oil” is intended to cover any oil that contains hydroxylated fatty acids. Preferred hydroxylated oils are hydroxylated vegetable oils, and a preferred hydroxylated vegetable oil for use in the present composition is castor oil.
  • As used herein, the term “castor oil” is intended to include ricinus oil, oil of Palma Christie, tangantargon oil and Neoloid (as described in Merck Index, 12th Edition, p: 311), as well as the oil from Ricinus Zanzibarinus. The latter has a high content of glycerides of ricinoleic acid. Thus, castor oil comprises glycerides of ricinoleic acid (a hydroxy fatty acid).
  • When castor oil is used in the present invention, it may conveniently be obtained by cold pressing of the seeds of Ricinus Communis L. (family: Euphorbiaceae).
  • The oil phase in the emulsions of the invention may constitute 1 to 50% v/v of the emulsion. A preferred concentration of oil in the emulsion is from 10 to 40% v/v. Particularly preferred are concentrations of 20 to 30% v/v.
  • The emulsion compositions of the invention can be prepared using conventional methods such as by homogenisation of a mixture of the oil and analgesic component with an aqueous phase (optionally together with a stabilizing agent). Any suitable device may be used, including a microfluidizer or ultrasonic device, though microfluidizers are preferred for large scale production.
  • Suitable stabilizers for use in the emulsions of the invention include block copolymers containing a polyoxyethylene block (i.e. a block made up of repeating ethylene oxide moieties). An example of a suitable stabilizer of this type is Poloxamer™. Other suitable stabilizers include phospholipid emulsifiers (for example soy and egg lecithins). Particularly preferred is the egg lecithin Lipoid E80™ (from Lipoid™), which contains both phosphatidylcholine and phosphatidyl ethanoline. Other suitable phospholipids include phospholipid-polyethylene glycol (PEG) conjugates (see for example Litzinger et al., Biochem Biophys Acta, 1190 (1994) 99-107).
  • Any suitable concentration of stabilizer/emulsifier may be used, and it typically falls within the range 0.1 to 10% w/v in the aqueous phase of the emulsion. Particularly preferred are concentrations of 1 to 5% w/v.
  • The stability of the emulsion can be enhanced by the addition of one or more co-emulsifier(s). Suitable pharmaceutically-acceptable co-emulsifiers include fatty acids, bile acids and salts thereof. Preferred fatty acids have greater than 8 carbon atoms, and particularly preferred is oleic acid. Of the suitable bile acids, preferred is deoxycholic acid. Suitable salts of the foregoing include the alkali metal (e.g. Na and K) salts. Co-emulsifiers can be added at a concentration of 1% w/v or less on the aqueous phase.
  • Buffering agents may also be used in the composition. For example, a buffer may used to maintain a pH that is compatible with nasal fluid, to preserve emulsion stability and/or to ensure that the analgesic component does not partition from the emulsion oil phase into the aqueous phase.
  • It will be clear to the person skilled in the art that additional components can also be added to the emulsion including thickening and gelling agents (such as cellulose polymers, particularly sodium carboxymethyl cellulose, alginates, gellans, pectins, acrylic polymers, agar-agar, gum tragacanth, gum xanthan, hydroxyethyl cellulose, chitosan, as well as block copolymers of polyoxyethylene-polyoxypropylene). Preservative agents such as methyl parabenzoates, benzylalcohol and chlorobutanol may also be added.
  • The delivery agent may comprise a liposome. Liposomes are microscopic vesicles composed of an aqueous compartment surrounded by a phospholipid bilayer that acts as a permeable entrapment barrier. Many different classes of liposomes are known (see Gregoriadis (ed.) in Liposome Technology, 2nd edition, vol I-III, CRC Press, Boca Ranto, Fla., 1993). Some liposomes can provide controlled sustained release of the encapsulated drug. In such systems, the rate of drug release is determined by the liposome's physicochemical properties. Liposomes can be tailored for a specific application by modification of size, composition, and surface charge to provide the desired rate of drug delivery (see Meisner D, et al: In Proceedings, 15th International Symposium on Controlled Release of Bioactive Materials. 15:262-263, 1988; Mezei M: In Drug Permeation Enhancement, Theory and Application. Hsieh DS (ed.): Marcel Dekker Inc., New York, 1993, pp 171-198; and Meisner D, et al: J Microencapsulation 6:379-387, 1989). Thus, liposome-encapsulation can act as an effective and safe delivery agent in the compositions of the invention.
  • The sustained release property of the liposomal product can be regulated by the nature of the lipid membrane and by the inclusion of other excipients in the composition of the liposomal products. Current liposome technology permits a reasonable prediction on the rate of drug release based on the composition of the liposome formulation. The rate of drug release is primarily dependent on the nature of the phospholipids, e.g. hydrogenated (—H) or unhydrogenated (-G), or the phospholipid/cholesterol ratio (the higher this ratio, the faster the rate of release), the hydrophilic/lipophilic properties of the active ingredients and by the method of liposome manufacturing.
  • Materials and procedures for forming liposomes are well known to those skilled in the art and include ethanol or ether injection methods. Typically, the lipid is dissolved in a solvent and the solvent evaporated (often under reduced pressure) to produce a thin film. The film is then hydrated with agitation. The analgesic component is incorporated at the lipid film forming stage (if lipophilic) or at the hydration phase as part of the aqueous hydrating phase (if hydrophilic). Depending on the hydration conditions selected and the physicochemical properties of the lipid(s) used, the liposomes can be multilamellar lipid vesicles (MLV), unilamellar lipid vesicles (including small unilamellar vesicles (SUV) and large unilamellar vesicles (LUV)) and as multivesicular liposomes.
  • Lipid components typically comprise phospholipids and cholesterol while excipients may comprise tocopherol, antioxidants, viscosity inducing agents and/or preservatives. Phospholipids are particularly useful, such as those selected from the group consisting of phosphatidylcholines, lysophosphatidylcholines, phosphatidylserines, phosphatidylethanolamines, and phosphatidylinositols. Such phospholipids may be modified using, for example, cholesterols, stearylamines, stearic acid, and tocopherols.
  • The compositions of the invention may further comprise other suitable excipients, including for example inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc.
  • Excipients such as humectants, isotonicity agents, antioxidants, buffers and/or preservatives are preferably used. Formulation and dosage will depend on, amongst other things, whether the analgesic is to be used in the form of drops or as a spray (aerosol). Alternatively, suspensions, ointments and gels can be applied to the nasal cavity. However, it is known that nasal mucous membranes are also capable of tolerating slightly hypertonic solutions. Should a suspension or gel be desired instead of a solution, appropriate oily or gel vehicles may be used or one or more polymeric materials may be included, which desirably should be capable of conferring bioadhesive characteristics to the vehicle.
  • Many other suitable pharmaceutically acceptable nasal carriers will be apparent to those skilled in the art. The choice of suitable carriers will depend on the exact nature of the particular nasal dosage form desired, for example whether the drug is to be formulated into a nasal solution (for use as drops or as a spray), a nasal suspension, a nasal ointment or a nasal gel. In another embodiment, nasal dosage forms are solutions, suspensions and gels, which contain a major amount of water (preferably purified water) in addition to the active ingredient. Minor amounts of other ingredients such as pH adjusters (e.g. a base such as NaOH), emulsifiers or dispersing agents, buffering agents, preservatives, wetting agents and jelling agents (e.g., methylcellulose) may also be present.
  • When solutions, the nasal compositions of the invention may be isotonic, hypertonic or hypotonic. If desired, sustained release nasal compositions, e.g. sustained release gels, can be readily prepared, preferably by employing the desired drug in one of its relatively insoluble forms, such as the free base or an insoluble salt.
  • The composition of the present invention may therefore be adjusted, if necessary, to approximately the same osmotic pressure as that of the body fluids (i.e. isotonic). Hypertonic solutions can irritate the delicate nasal membranes, while isotonic compositions do not. Isotonicity can be achieved by adding glycerol or an ionic compound to the composition (for example, sodium chloride). The compositions may take the form of a kit of parts, which kit may comprise the intranasal composition together with instructions for use and/or unit dosage containers and/or an intranasal delivery device.
  • The compositions of the invention are administered intranasally to a patient in order to induce analgesia. An effective amount of the analgesic is delivered to a patient. A unit dose can be delivered to one nostril. Alternatively, half of a dose or two doses can be delivered to each nostril each time administration occurs. The dose will depend upon a number of factors including the analgesic that is being delivered, the age and sex of the patient, the nature and extent of the pain to be treated and the period of treatment. A suitable dose for an opioid analgesic may be from 0.02 to 100 mg such as 0.1 to 50 mg. For buprenorphine or a buprenorphine salt or ester, a suitable dose is from 0.02 to 1.2 mg, such as from 50 to 600 μg or from 100 to 400 μg, calculated as buprenorphine.
  • Multiple doses of a composition according to the invention may be employed. For example, the rapid onset analgesia produced by the solution of the invention may permit self-titration of analgesic by the patient. The analgesic effect of an initial dose can be quickly and reliably gauged by the patient and, if insufficient, can be immediately supplemented by further dose(s) (often alternating between each nostril) until the required level of analgesia is attained. Multiple dosing may also be used in order to extend pain relief. For example, from 1 to 4 doses, for example 2 to 4 doses, per day may be indicated.
  • The compositions of the invention may be used to treat an existing pain condition or to prevent a pain condition from occurring. An existing pain may be alleviated. Compositions can be used to treat or manage chronic or acute pain, for example the management of post-operative pain (e.g. abdominal surgery, back surgery, cesarean section, hip replacement or knee replacement).
  • Other medical uses include: pre-operative intranasal administration of the solution of the invention; therapy or prophylaxis adjunctive to anesthesia; post-operative analgesia; the management of trauma pain; the management of cancer pain; the management of endometriosis; the management of inflammatory pain; the management of arthritis pain (including pain associated with rheumatoid arthritis and osteoarthritis); the management of back pain; the management of myocardial pain (for example ischaemic or infarction pain); the Management of dental pain; the management of neuropathic pain (e.g. diabetic neuropathy, post-herpetic neuralgia or trigeminal neuralgia); the management of colic (e.g. renal colic or gallstones), headache, migraine, fibromyalgia or dysmenorrhoea; the management of breakthrough pain associated with malignant and non-malignant disease; and the management of acute procedural pain (e.g. bone marrow aspiration or lumber puncture).
  • When in the form of a solution, compositions according to the invention may be administered to the nasal cavity in forms including drops or sprays. The preferred method of administration is using a spray device. Spray devices can be single (unit) dose or multiple dose systems, for example comprising a bottle, pump and actuator. Suitable spray devices are available from various commercial sources including Pfeiffer, Valois, Bespak and Becton-Dickinson.
  • When in the form of powder or microspheres, a nasal insufflator device may be employed. Such devices are already in use for commercial powder systems intended for nasal application. The insufflator may be used to produce a fine, dispersed plume of the dry powder or microspheres. The insufflator is preferably provided with means for administering a predetermined dose of the analgesic composition. Powder or microspheres may be contained in a bottle or container adapted to be used with the insufflator. Alternatively, powders or microspheres may be provided in capsules (e.g. gelatin capsules) or other single dose devices adapted for nasal administration, in which embodiments the insufflator may comprise means for breaking open the capsule (or other single dose device).
  • The invention also provides specific buprenorphine formulations which are suitable for nasal delivery. A first solution of the invention consists essentially of 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, from 0.1 to 20 mg/ml of a chitosan, from 0.1 to 15 mg/ml of HPMC, and water. A second solution of the invention consists essentially of 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, from 0.1 to 20 mg/ml of chitosan, from 50 to 200 mg/ml of a polyoxyethylene-polyoxypropylene copolymer of the general formula HO(C2H4O)a(C3H6O)b(C2H4O)aH wherein a is from 2 to 130 and b is from 15 to 67, and water.
  • In each case, the buprenorphine salt may be an acid addition salt or a salt with a base. Suitable acid addition salts the hydrochloride, sulphate, methane sulphonate, stearate, tartrate and lactate salts. The hydrochloride salt is preferred.
  • The concentration of buprenorphine or buprenorphine salt or ester in either solution is from 0.1 to 10 mg/ml, for example from 0.5 to 8 mg/ml. Preferred concentrations are 1 to 6 mg/ml, for example 1 to 4 mg/ml. Suitable solutions can contain the buprenorphine or buprenorphine salt or ester at a concentration of 1 mg/ml or 4 mg/ml, calculated as buprenorphine. Each solution is typically delivered as a nasal spray. A 100 μl spray of a solution containing 1 to 4 mg/ml of buprenorphine or a buprenorphine salt or ester, calculated as buprenorphine, thus results in a clinical dose of 100 to 400 μg of the buprenorphine or buprenorphine salt or ester, calculated as buprenorphine. Two such sprays may be given per nostril per administration time to deliver a dose of up to 4×400 μg, i.e. up to 1600 μg, of buprenorphine or the buprenorphine salt or ester, calculated as buprenorphine.
  • Suitable chitosans are described above. The chitosan is present in solution at a concentration of from 0.1 to 20 mg/ml, for example from 0.5 to 20 mg/ml. Preferably the solution contains from 1 to 15 mg/ml, more preferably from 2 to 10 mg/ml, of chitosan. A chitosan concentration of 5 mg/ml is particularly suitable.
  • Any suitable HPMC may be employed, as described above also. The HPMC used in the solution of the invention is preferably one having an apparent viscosity (measured as a 2% solution in water at 20° C.) in the range from 3000 to 6000 cps. The concentration of the HPMC having a viscosity of from 3000 to 6000 cps is in the range from 0.1 to 15 mg/ml, preferably from 0.5 to 10 mg/ml and preferably from 1 to 5 mg/ml.
  • Suitable polyoxyethylene-polyoxypropylene copolymers typically are described above. The polyoxyethylene-polyoxypropylene copolymer is present in an amount of from 50 to 200 mg/ml, preferably from 65 to 160 mg/ml and more preferably from 80 to 120 mg/ml. A preferred concentration is 100 mg/ml.
  • Any suitable preservative may be present in the solutions, in particular a preservative that prevents microbial spoilage of the solution. The preservative must be compatible with the other components of the solution. The preservative may be any pharmaceutically acceptable preservative, for example a quaternary ammonium compound such as benzalkonium chloride.
  • The solution has a pH of from 3 to 4.8. Any pH within this range may be employed provided the buprenorphine or buprenorphine salt or ester remains dissolved in the solution. The pH may be from 3.2 to 4.2, for example from 3.2 to 4.0 or 3.5 to 4.0. A suitable pH is from 3.6 to 3.8. The pH may be adjusted to an appropriate value by addition of a physiologically acceptable acid and/or physiologically acceptable buffer. The pH may thus be adjusted solely by means of a physiologically acceptable mineral acid or solely by means of a physiologically acceptable organic acid. The use of hydrochloric acid is preferred.
  • A tonicity adjustment agent may be included in the solution. The tonicity adjustment agent may be a sugar, for example dextrose, or a polyhydric alcohol, for example mannitol. A solution may be hypertonic, substantially isotonic or hypotonic. A substantially isotonic solution can have an osmolality of from 0.28 to 0.32 osmol/kg. An exactly isotonic solution is 0.29 osmol/kg. A sufficient amount of a tonicity adjustment agent such as dextrose or mannitol may therefore be present to achieve a desired osmolality. Preferably a solution contains 50 mg/ml dextrose or mannitol.
  • The osmolality of a solution containing chitosan and HPMC or a poloxamer may be from 0.1 to 0.8 osmol/kg such as from 0.2 to 0.6 osmol/kg or preferably from 0.32 to 0.4 osmol/kg.
  • The solutions may also contain other ingredients such as an antioxidant, chelating agent or other agent generally used in pharmaceutical liquid preparations. The solution can be a sterile solution.
  • The solution containing chitosan and HPMC is prepared by dissolving buprenorphine or a physiologically acceptable salt or ester thereof, a chitosan and HPMC in water, typically Water for Injections. The amount of the buprenorphine or salt or ester thereof is selected so that from 0.1 to 10 mg/ml of buprenorphine or the buprenorphine salt or ester is dissolved in the solution. The required concentrations of the chitosan and of HPMC are provided too. A preservative can be dissolved in the solution. The pH of the solution can be adjusted to a value within the range from 3 to 4.8 as required. Preferably the pH is adjusted by means of hydrochloric acid.
  • A solution containing chitosan and a polyoxyethylene-polyoxypropylene copolymer is prepared by dissolving buprenorphine or a physiologically acceptable salt or ester thereof, a chitosan and the polyoxyethylene-polyoxypropylene copolymer in water, typically Water for Injections. The amount of the buprenorphine or salt or ester thereof is selected so that from 0.1 to 10 mg/ml of buprenorphine or the buprenorphine salt or ester is dissolved in the solution. The required concentrations of the chitosan and of the polyoxyethylene-polyoxypropylene copolymer are provided too. A preservative can be dissolved in the solution. The pH of the solution can be adjusted to a value within the range from 3 to 4.8 as required. Preferably, the pH is adjusted by means of hydrochloric acid.
  • Other components can be provided in the solutions at any convenient stage. For example, dextrose or mannitol may be dissolved in the water in which the buprenorphine or buprenorphine salt or ester is being dissolved. A sterile solution can be obtained either by using sterile starting materials and operating under sterile conditions and/or by using standard sterilising techniques such as passing the final solution through a sterilising filter. A pyrogen-free solution can thus be provided. The solution can then be introduced into a nasal delivery device, typically a sterile such device. If required, prior to sealing the device, the solution may be overlaid with an inert gas such as nitrogen to protect it from oxidation.
  • A third pharmaceutical solution of the invention consists essentially of 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, from 5 to 40 mg/ml of a pectin having a low degree of esterification, in particular a degree of esterification of less than 50%, and water. The buprenorphine salt may be an acid addition salt or a salt with a base. Suitable acid addition salts include the hydrochloride, sulphate, methane sulphonate, stearate, tartrate and lactate salts. The hydrochloride salt is preferred.
  • The concentration of buprenorphine or buprenorphine salt or ester is from 0.1 to 10 mg/ml, for example from 0.5 to 8 mg/ml. Preferred concentrations are 1 to 6 mg/ml, for example 1 to 4 mg/ml calculated as buprenorphine. Suitable solutions can contain buprenorphine or a buprenorphine salt or ester in an amount of 1 mg/ml or 4 mg/ml, calculated as buprenorphine.
  • The solution is typically delivered as a nasal spray. A 100 μl spray of a solution containing 1 to 4 mg/ml of buprenorphine or a buprenorphine salt or ester, calculated as buprenorphine thus results in a clinical dose of 100 to 400 μg of the buprenorphine or buprenorphine salt or ester, calculated as buprenorphine. Two such sprays may be given per nostril per administration time to deliver a dose of up to 4×400 μg, i.e. up to 1600 μg, of buprenorphine or the buprenorphine salt or ester, calculated as buprenorphine.
  • Suitable pectins for inclusion in the solution are described above. The solutions gels on the mucosal surfaces of the nasal cavity without the need for extraneous source of divalent metal ions. The pectin is present in the solution of the invention at a concentration of from 5 to 40 mg/ml, for example from 5 to 30 mg/ml. More preferably, the pectin concentration is from 10 to 30 mg/ml or from 10 to 25 mg/ml.
  • A pectin-containing solution of the invention has a pH of from 3 to 4.2. Any pH within this range may be employed provided the buprenorphine or buprenorphine salt or esteremains dissolved in the solution. The pH may be from 3.2 to 4.0, for example from 3.5 to 4.0. A particularly suitable pH is from 3.6 to 3.8. The pH may be adjusted to an appropriate value by addition of a physiologically acceptable acid and/or physiologically acceptable buffer. The pH may thus be adjusted solely by means of a physiologically acceptable mineral acid or solely by means of a physiologically acceptable organic acid. The use of hydrochloric acid is preferred.
  • Any suitable preservative may be present in the pectin-containing solution, in particular a preservative that prevents microbial spoilage of the solution. The preservative may be any pharmaceutically acceptable preservative, for example phenylethyl alcohol or propyl hydroxybenzoate (propylparaben) or one of its salts. The phenylethyl alcohol and the propylparaben or propylparaben salt are preferably used in combination. The preservative must be compatible with the other components of the solution and, in particular, must not cause gelling of the solution.
  • Pectin-containing solutions may include a tonicity adjustment agent such as a sugar, for example dextrose, or a polyhydric alcohol for example mannitol. A solution may be hypertonic, substantially isotonic or hypotonic. A substantially isotonic solution can have an osmolality of from 0.28 to 0.32 osmol/kg. An exactly isotonic solution is 0.29 osmol/kg. The osmolality of a solution may be from 0.1 to 0.8 osmol/kg such as from 0.2 to 0.6 osmol/kg or preferably from 0.35 to 0.5 osmol/kg. A suitable osmolality range is from 0.32 to 0.36 osmol/kg. A sufficient amount of a tonicity adjustment agent such as dextrose or mannitol may therefore be present to achieve such osmolalities. Preferably a solution contains 50 mg/ml dextrose or mannitol.
  • The pectin-containing solution is prepared by dissolving buprenorphine or a physiologically acceptable salt or ester thereof in water, typically Water for Injections, and the resulting solution is mixed with a solution of a suitable pectin in water, again typically Water for Injections. The amount of the buprenorphine or salt or ester thereof and of the pectin are selected so that from 0.1 to 10 mg/ml of buprenorphine or the buprenorphine salt or ester and from 5 to 40 mg/ml of pectin are dissolved in the mixed solution. A preservative or combination of preservatives may be dissolved in the solution. The pH of the mixed, solution can be adjusted to a value within the range from 3 to 4.2 as required. Preferably, the pH is adjusted with hydrochloric acid if pH adjustment is required.
  • Other components can be provided in solution at any convenient stage. For example, dextrose or mannitol may be dissolved in the water in which the buprenorphine or buprenorphine salt or ester is being dissolved. A sterile solution can be obtained either by using sterile starting materials and operating under sterile conditions and/or by using standard sterilising techniques such as passing the final solution through a sterilising filter. A pyrogen-free solution can thus be provided. The solution can then be introduced into a nasal delivery device, typically a sterile such device. If required, prior to sealing the device, the solution may be overlaid with an inert gas such as nitrogen to protect it from oxidation.
  • Each of the three solutions of the invention is administered intranasally to a patient in order to induce analgesia. Rapid onset of analgesia and prolonged analgesia can thus be obtained. An effective amount of buprenorphine or a salt or ester thereof is delivered to a patient. A unit dose can be delivered to one nostril. Alternatively, half of a dose or two doses can be delivered to each nostril each administration time. The dose will depend upon a number of factors including the age and sex of the patient, the nature and extent of the pain to be treated and the period of treatment. A suitable dose of buprenorphine or a buprenorphine salt or ester is from 0.02 to 1.2 mg, such as from 50 to 600 μg or from 100 to 400 μg, calculated as buprenorphine.
  • Multiple doses of a solution according to the invention may be employed. For example, the rapid onset analgesia produced by the solution of the invention may permit self-titration of analgesic by the patient. The analgesic effect of an initial dose can be quickly and reliably gauged by the patient and, if insufficient, can be immediately supplemented by further dose(s) (often alternating between each nostril) until the required level of analgesia is attained. Multiple dosing may also be used in order to extend pain relief For example, from 2 to 4 doses per day may be indicated.
  • The solutions of the invention may be used to treat an existing pain condition or to prevent a pain condition from occurring. An existing pain may be alleviated. Solutions of the invention can be used to treat or manage chronic or acute pain, for example the management of post-operative pain (e.g. abdominal surgery, back surgery, cesarean section, hip replacement or knee replacement). Other medical uses have been described above.
  • The solutions according to the invention may be administered to the nasal cavity in forms including drops or sprays. The preferred method of administration is using a spray device. Spray devices can be single (unit) dose or multiple dose systems, for example comprising a bottle, pump and actuator. Suitable spray devices are available from various commercial sources including Pfeiffer, Valois, Bespak and Becton-Dickinson.
  • As already mentioned, rapid onset of analgesia and prolonged analgesia can be achieved by means of the invention. The analgesic delivery profile that can be attained may avoid the relatively high Cmax values associated with intravenous administration and so lead to an improved therapeutic index. The peak plasma concentration of an analgesic that is attained after administration is defined as Cmax. The invention can permit reduction or elimination of some or all of the side effects associated with the analgesic.
  • Cmax is typically from 1 to 5 ng/ml, for example from 1 to 4 ng/ml or from 1.5 to 3 ng/ml. Cmax may be from 1 to 2 ng/ml, especially for lower doses of buprenorphine. The time at which Cmax is reached (Tmax) is typically 10 to 40 minutes after administration, for example 10 to 30 minutes or 15 to 25 minutes such as 15 to 20 minutes.
  • In preferred embodiments, the delivery agent is adapted to deliver the analgesic component such that Cmax=Copt. The term Copt is used in relation to analgesic drugs which exhibit a dose-response curve to analgesia which is displaced to the left with respect to the dose-response curve for side-effects. The term defines a therapeutic plasma concentration or range thereof which produces acceptable pain relief or pain amelioration but which does not produce side-effects or produces side effects which are less than those associated with higher plasma concentrations.
  • Preferably, the solutions of the invention enable the buprenorphine or salt or ester thereof to be delivered such that a Cther of 0.2 ng/ml or more, for example 0.4 ng/ml or more, is attained within 30 minutes after introduction into the nasal cavity within 30 minutes, for example within 0.5 to 20 minutes, such as 2 to 15 minutes or 5 to 10 minutes. The term Cther defines a therapeutic plasma concentration (or range thereof). Thus, the term is used herein to define a blood plasma concentration (or range of plasma concentrations) of the buprenorphine or salt or ester thereof that produces pain relief or pain amelioration. Cther may be from 0.4 to 5 ng/ml, for example 0.4 to 1 ng/ml or 0.5 to 4 ng/ml or 0.8 to 2 ng/ml.
  • The Tmaint is typically at least 2 hours. The term Tmaint defines the duration of maintenance of Cther after administration of the analgesic. For example, the Tmaint can be from up to 24 hours, up to 12 hours or up to 6 hours such as from 2 to 4 hours or 2 to 3 hours. By means of the invention, therefore, a Cther of at least 0.4 ng/ml may be attained within 2 to 15 minutes and maintained for a time period Tmaint of from 2 to 4 hours.
  • The following Examples illustrate the invention.
  • Example 1 Nasal Solution Containing Buprenorphine (4 mg/ml) and Pectin
  • 5 g of pectin (SLENDID (trade mark) 100, CP Kelco, Denmark) was dissolved by stirring into approximately 180 ml of water for injection (WFI) (Baxter, UK). 1075 mg of buprenorphine hydrochloride (MacFarlan Smith, UK) and 12.5 g of dextrose (Roquette) were dissolved into the pectin solution. 1:25 ml of phenylethyl alcohol (R. C. Treat, UK) and 50 mg of propyl hydroxybenzoate (Nipa, UK) were dissolved into the pectin/buprenorphine solution. The solution was adjusted to 250 ml using WFI. 1M hydrochloric acid (BDH, UK) was added to adjust the pH to 3.6.
  • The final product was a slightly turbid solution 4.3 mg/ml buprenorphine hydrochloride (corresponding to 4 mg/ml buprenorphine), 20 mg/ml pectin, 50 mg/ml dextrose, 5 μl/ml phenylethyl alcohol and 0.2 mg/ml propyl hydroxybenzoate. The pH of the solution was 3.6, as mentioned above. The osmolality of the solution was 0.46 osmol/kg.
  • Single dose nasal spray devices (Pfeiffer, Germany) were filled with the solution. Each device was filled with 123 μl of liquid. Actuation of the device delivered a dose of 100 μl of liquid containing 400 μg of buprenorphine and 2 mg of pectin.
  • Example 2 Nasal Solution Containing Buprenorphine (2 mg/ml) and Pectin
  • 5 g of pectin is dissolved by stirring into approximately 180 ml of WFI. 538 mg of buprenorphine hydrochloride and 12.5 g of dextrose are dissolved into the pectin solution. 1.25 ml of phenylethyl alcohol and 50 mg of propyl hydroxybenzoate are dissolved into the pectin/buprenorphine solution. The solution is adjusted to 250 ml using WFI.
  • The final product is a slightly turbid solution containing 2.16 mg/ml buprenorphine hydrochloride (corresponding to 2 mg/ml buprenorphine), 20 mg/ml pectin, 50 mg/ml dextrose, 5 μl/ml phenylethyl alcohol and 0.2 mg/ml propyl hydroxybenzoate.
  • 123 μl of the above solution is filled into a Valois Monospray single dose nasal spray device (Pfeiffer, Germany). Actuation of the device will deliver a dose of 100 μl of liquid containing 200 μg of buprenorphine and 2 mg of pectin.
  • Example 3 Nasal Solution Containing Buprenorphine (4 mg/ml), Chitosan and HPMC
  • 0.75 g of HPMC (Methocel (trade mark) E4M, Colorcon, UK) was dispersed into approximately 125 ml of pre-heated (70-80° C.) water for injection (WFI) (Baxter, UK). The HPMC dispersion was stirred in an ice bath until a clear solution had formed. 1.25 g of chitosan glutamate (Protosan (trade mark) UPG213, Pronova, Norway) was dissolved in the HPMC solution. 75 mg of 50% w/w benzalkonium chloride solution (Albright and Wilson, UK) was dispersed in 10 ml of WFI and transferred with an additional 40 ml of WFI to a 250 ml volumetric flask. 1075 mg of buprenorphine hydrochloride (MacFarlan Smith, UK) and 12.5 g of dextrose (Roquette, UK) were transferred into the volumetric flask. The chitosan/HPMC solution and an additional 40 ml of WFI were added to the flask. The solution was adjusted to pH 3.4 using 1M hydrochloric acid solution (BDH, UK) and the flask contents adjusted to 250 ml using WFI.
  • The final product was a clear colourless solution containing a 4.3 mg/ml buprenophine hydrochloride (corresponding to 4 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 3 mg/ml HPMC, 50 mg/ml dextrose and 0.15 mg/ml benzalkonium chloride. The osmolality of the final solution was 0.34 osmol/kg and the viscosity, as measured using a Brookfield CP70 cone and plate viscometer was 84.7 cps at 2.5 rpm and 25° C.
  • Single dose nasal spray devices (Pfeiffer, Germany) were filled with the solution. Each device was filled with 123 μl of liquid. Actuation of the device delivered a dose of 100 μl of liquid containing 400 μg of buprenorphine, 0.5 mg of chitosan and 0.3 mg of HPMC. Hence, a dose of 400 μg buprenorphine is provided by a single spray into one nostril. A dose of 800 μg is provided by a single spray into each nostril.
  • Example 4 Nasal Solution Containing Buprenorphine (1 mg/ml), Chitosan and HPMC
  • A solution containing HPMC, chitosan glutamate and benzalkonium chloride is prepared according to Example 3. 269 mg of buprenorphine hydrochloride and 12.5 g of mannitol (Sigma, UK) are transferred into the volumetric flask. The chitosan/HPMC solution and an additional 40 ml of WFI are added to the flask. The pH of the solution is adjusted to pH 3.6 using 1M hydrochloric acid solution and the flask contents adjusted to 250 ml using WFI.
  • The final product is a clear colourless solution containing 1.08 mg/ml buprenorphine hydrochloride (corresponding to 1 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 3 mg/ml HPMC, 50 mg/ml mannitol and 0.15 mg/ml benzalkonium chloride.
  • 123 μl of the above solution is filled into a single dose nasal spray device (Pfeiffer, Germany). Actuation of the device will deliver a dose of 100 μl of liquid containing 100 μg of buprenorphine, 0.5 mg of chitosan and 0.3 mg of HPMC.
  • 5 ml of the solution is filled into a 10 ml glass bottle. A Valois VP7, 100 μl pump and actuator (Valois, France) are attached to the bottle. When primed, the pump will dispense 100 μl of solution containing 100 μg of buprenorphine.
  • Example 5 Nasal Solution Containing Buprenorphine (4 mg/ml), Chitosan and Poloxamer
  • 25 g of poloxamer 188 (Lutrol (trade mark) F-68, BASF, Germany) was dissolved by stirring into 100 ml of water for injection (WFI) (Baxter, UK) at a temperature of 2 to 8° C. 1.25 g of chitosan glutamate (Protasan (trade mark) UPG213, Pronova, Norway) was dissolved in the poloxamer solution. 75 mg of 50% w/w benzalkonium chloride solution (Albright and Wilson, UK) was dispersed in 10 ml of WFI and transferred with an additional 40 ml of WFI to a 250 ml volumetric flask. 1075 mg of buprenorphine hydrochloride (MacFarlan Smith, UK) and 12.5 g of dextrose (Roquette, UK) were transferred into the volumetric flask. The chitosan/poloxamer solution and an additional 40 ml of WFI were added to the flask. The solution was adjusted to pH 3.4 using 1M hydrochloric acid solution (BDH, and the flask contents adjusted to 250 ml using WFI.
  • The final product was a clear colourless solution containing 4.3 mg/ml buprenorphine hydrochloride (corresponding to 4 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 100 mg/ml poloxamer 188, 50 mg/ml dextrose and 0.15 mg/ml benzalkonium chloride. The osmolality of the final solution was 0.60 Osmol/kg.
  • Single dose nasal spray devices (Pfeiffer, Germany) were filled with the solution. Each device was filled with 123 μl of liquid. Actuation of the device delivered a dose of 100 μl of liquid containing 400 μg of buprenorphine, 0.5 mg of chitosan and 10 mg of poloxamer 188.
  • Example 6 Nasal Solution Containing Buprenorphine (1 mg/ml), Chitosan and Poloxamer
  • A solution containing chitosan glutamate, poloxamer 188 and benzalkonium chloride is prepared according to Example 5. 269 mg of buprenorphine hydrochloride and 12.5 g mannitol (Sigma, UK) are transferred into the volumetric flask. The chitosan/poloxamer solution and an additional 40 ml of WFI are added to the flask. The pH of the solution is adjusted to pH 3.6 using 1M hydrochloric acid and the flask contents adjusted to 250 ml using WFI.
  • The final product is a clear colourless solution containing 1.08 mg/ml buprenorphine hydrochloride (corresponding to 1 mg/ml buprenorphine), 5 mg/ml chitosan glutamate, 100 mg/ml poloxamer 188, 50 mg/ml mannitol and 0.15 mg/ml benzalkonium chloride.
  • 123 μl of the above solution is filled into a single dose nasal spray device (Pfeiffer, Germany). Actuation of the device will deliver a dose of 100 μl of liquid containing 100 μg of buprenorphine, 0.5 mg of chitosan and 10 mg of poloxamer 188.
  • 4 ml of the solution is filled into a 5 ml glass bottle. A Pfeiffer 100 μl nasal spray pump and actuator are attached to the bottle. When primed, the pump will dispense 100 μl of solution containing 100 μg of buprenorphine.
  • Example 7 Effects of Varying Parameters of Buprenorphine-Pectin Solutions General Methods
  • The appearance, pH (Mettler MP230 pH meter) and osmolality (Osmomat 030 cryoscopic osmometer) of the solutions were determined.
  • The viscosity of the solution was measured using a Brookfield Cone and Plate Rheometer. Results given are the mean of determinations at three rotation speeds appropriate to the viscosity of the solution.
  • The spray characteristics from a Pfeiffer multi-dose nasal spray device (standard nozzle, 0.1 ml pump, Cat. No. 62897) were evaluated by measurement of plume angle using image analysis. Results given are the mean of four determinations (two at one orientation and two at a 90° rotation to the first orientation).
  • The buprenorphine content of formulations was determined by hplc.
  • Gels were prepared by controlled mixing of 20 ml of formulation with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl2.2H20) before standing for 1 hour at room temperature. A visual assessment of the structure, uniformity, clarity and evidence of syneresis of each gel was conducted and, in addition, the gel structure was examined with a Stable Microsystems Texture Analyser. Results (from single determinations) are expressed in terms of force (maximum penetration force) and area (total work of gel penetration).
  • Effect of Pectin Concentration on Appearance, Solution/Gel Properties and Spray Characteristics 1. Methods
  • Buprenorphine hydrochloride (107.5 mg) and anhydrous dextrose (1.25 g) were stirred in 18-20 ml water in a 25 ml volumetric flask together with an appropriate quantity of pectin and the mixture stirred overnight or until a solution formed. The mixture was then made up to 25 ml with water to give a solution containing 4 mg/ml buprenorphine, 50 mg/ml dextrose and 1, 5, 10, 20, 30, 40 or 80 mg/ml pectin and the pH, appearance, osmolality, viscosity were determined. In addition, spray characteristics from a Pfeiffer multi-dose nasal spray device (standard nozzle, 0.1 ml pump, Cat. No. 62897) were evaluated by measurement of plume angle using image analysis. Gels were prepared by controlled mixing of 20 ml of formulation with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl2.2H20) before standing for 1 hour at room temperature. A visual assessment of the structure, uniformity, clarity and evidence of syneresis of each gel was conducted and, in addition, the gel structure was examined with a Stable Microsystems Texture Analyser.
  • An in vitro method was employed to simulate the gelling that may occur when the pectin formulation comes into contact with the nasal mucosal surface. This involved adding 2 ml of each formulation to an equal volume of simulated nasal electrolyte solution (SNES) (comprised 8.77 g/l sodium chloride, 2.98 g/l potassium chloride and 0.59 g/l calcium chloride dihydrate) and agitating gently. The mixtures were left to stand for 1 hour at room temperature before visual assessment.
  • 2. Results
  • As pectin concentration increased, solutions became increasingly turbid, osmolality and viscosity increased and plume angle decreased (Table 1). An excellent relationship was obtained between concentration and plume angle up to 30 mg/ml pectin. The pH was not significantly affected by pectin concentration.
  • Upon addition of calcium ions pectin formed visually satisfactory gels in the concentration range 5-20 mg/ml (Table 2). Correspondingly greater integrity of gel structure was noted over this range. At higher pectin concentrations texture analysis results were inconclusive because homogeneity of the gel is difficult to control and increasing syneresis was observed.
  • At a lower calcium ion concentration (SNES) pectin produced mobile gels at 10-20 mg/ml and strong, inhomogeneous gels at higher concentrations.
  • TABLE 1
    Appearance, pH, osmolality, viscosity and spray characteristics
    (plume angle) of buprenorphine solutions containing 4.3 mg/ml
    buprenorphine hydrochloride (BPN•HCl), 50 mg/ml dextrose
    and different concentrations of pectin (Slendid 100).
    Pectin Osmo- Vis- Plume
    Batch Conc. lality cosity angle
    No. (mg/ml) pH (osmol/kg) (cps) (°) Appearance
    105 1 4.4 0.32 1.4 56 Clear, colourless
    solution
    106 5 4.2 0.33 2.1 53 Very slightly turbid,
    colourless solution
    107 10 4.1 0.34 3.7 42 Slightly turbid,
    colourless solution
    108 20 4.0 0.37 9.0 29 Slightly turbid, pale
    yellow solution
    153 30 3.9 0.40 16.8 21 Turbid, pale yellow
    solution
    109 40 4.0 0.43 33.9 20 Turbid, pale yellow
    solution
    110 80 4.0 0.55 N/M* 16 Very turbid, pale
    yellow solution
    *N/M = not measurable
  • TABLE 2
    Gelling properties of buprenorphine solutions containing
    4.3 mg/ml BPN•HCl, 50 mg/ml dextrose and different
    concentrations of pectin (Slendid 100) when mixed
    with a standard calcium chloride solution.
    Pectin Texture analysis
    Batch Conc. Force Area
    No. (mg/ml) (g) (g s) Visual assessment
    105 1 Clear, slightly viscous, colourless
    solution. Did not gel.
    106 5 116 1420 Slightly opalescent, strong, uniform
    gel with minimum syneresis.
    107 10 220 3858 Semi-transparent, strong, uniform gel
    with minimum syneresis.
    108 20 279 4872 Semi-transparent, pale yellow, strong,
    uniform gel with minimum syneresis.
    153 30 190 4259 Semi-transparent, pale yellow, strong,
    uniform gel with some syneresis.
    109 40 234 2691 Semi-transparent, pale yellow, very
    strong, non-uniform gel with some
    syneresis.
    110 80 303 5356 Semi-transparent, yellow, extremely
    strong, non-uniform gel with
    significant syneresis.
  • TABLE 2a
    Gelling properties of buprenorphine solutions containing
    4.3 mg/ml BPN•HCl, 50 mg/ml dextrose and different concentrations
    of pectin (Slendid 100) when mixed with SNES.
    Pectin
    Batch Conc.
    No. (mg/ml) Visual assessment
    161 1 Clear, slightly viscous, colourless solution. Did not gel.
    162 5 Clear, viscous, colourless solution. Did not gel.
    163 10 Clear, colourless, weak diffuse gel.
    164 20 Pale yellow, semi-transparent weak diffuse gel.
    165 30 Semi-transparent pale yellow, strong gel with some
    syneresis.
    166 40 Semi-transparent pale yellow, strong gel with some
    syneresis.
    167 80 Opaque pale yellow, very strong gel with significant
    syneresis.
  • Effect of pH on Solubility and Gelling Properties of Buprenorphine Hydrochloride 1. Methods
  • Stock solutions containing pectin (Slendid 100) (20 mg/ml) and dextrose (50 mg/ml) were prepared at various pH in the range pH 3.0 to 6.0 (pH adjustments were made with 0.1M HCl or 0.1M meglumine). An excess of buprenorphine hydrochloride was then stirred overnight at 18° C. in 5 or 25 ml of each solution. Saturated solutions were recovered by passing each mixture through a 0.2 μm polycarbonate membrane filter. The concentration of buprenorphine hydrochloride in the filtrate was determined by hplc.
  • In preliminary experiments addition of excess buprenorphine hydrochloride was found to reduce the pH of the (unbuffered) solutions. In order to produce solutions at the higher end of the desired pH range, a minimal excess of buprenorphine hydrochloride was added to solutions (5 ml) containing pectin (Slendid 100) 20 mg/ml) and dextrose (50 mg/ml) adjusted to various pH values in the range pH 4.5 to 6.0 with 0.1M HCl or 0.1M meglumine. The quantity of excess buprenorphine hydrochloride added was based on preliminary findings and on reported solubility data for buprenorphine hydrochloride (Cassidy et al, J. Controlled Release 25, 21-29, 1993). Following overnight stirring at 18° C., mixtures were examined to confirm that undissolved drug remained before saturated solutions were recovered by passing each mixture through a 0.2 μm polycarbonate membrane filter.
  • For selected formulations gels were prepared by controlled mixing of 20 ml of formulation with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl2.2H2O) before standing for 1 hour at room temperature. A visual assessment of the structure, uniformity, clarity and evidence of syneresis of each gel was conducted and, in addition, the gel structure was examined with a Stable Microsystems Texture Analyser.
  • 2. Results
  • Buprenorphine was sparingly soluble (greater than 10 ng/ml) in aqueous solutions containing 20 mg/ml pectin+50 mg/ml dextrose at pH below 4.4 (Table 3). In general, solubility fell as pH increased above 4.5 (Table 3a). Solutions were slightly soluble (less than 10 ng/ml) at pH 4.5-6.0.
  • Gelling properties were largely unaffected by pH (and therefore by buprenorphine concentration) (Table 4).
  • TABLE 3
    Solubility of BPN•HCl at pH 3.2-4.0 in solutions containing
    20 mg/ml pectin (Slendid 100) and 50 mg/ml dextrose.
    Buprenorphine
    Batch No. Final pH detected* (mg/ml)
    043 3.2 12.3
    085 3.6 14.2
    086 3.8 13.5
    087 3.9 15.8
    048 4.0 14.3
    *Expressed as buprenorphine free base
  • TABLE 3a
    Solubility of BPN•HCl at pH 4.4-5.3 in solutions containing
    20 mg/ml pectin (Slendid 100) and 50 mg/ml dextrose.
    Buprenorphine
    Batch No. Final pH detected* (mg/ml)
    202 4.4 11.6
    203 4.5 9.0
    204 4.7 7.3
    205 4.7 6.0
    206 4.8 3.5
    207 5.1 2.7
    209 5.2 1.4
    208 5.3 1.3
    *Expressed as buprenorphine free base
  • TABLE 4
    Effect of pH on the gelling properties of BPN•HCl in solution
    containing 20 mg/ml pectin (Slendid 100) and 50 mg/ml dextrose
    when mixed with a standard calcium chloride solution.
    Texture analysis*
    Batch pH Force Area
    No. (actual) (g) (g s) Visual assessment
    043 3.0 (3.2) 328 4439 Semi-transparent, uniform gel
    with minimum syneresis.
    086 3.5 (3.8) 309 4018 Semi-transparent, uniform gel
    with minimum syneresis.
    048 4.0 (3.9) 371 4056 Semi-transparent, uniform gel
    with minimum syneresis.
    089 5.5 (5.1) 168 1620 Semi-transparent, uniform gel
    with some syneresis.
    *Reduced volumes (14 ml of formulation and 3.5 ml of CaCl2•2H2O) were used due to higher than expected loss of volume during filtration.
  • Effect of Osmolality (Dextrose or Mannitol Concentration) on Viscosity, Spray Characteristics and Gelling Properties of Buprenorphine Hydrochloride 1. Methods
  • Buprenorphine hydrochloride (107.5 mg) and pectin (Slendid 100) (500 mg) were stirred in 18-20 ml water in a 25 ml volumetric flask together with an appropriate quantity of anhydrous dextrose or mannitol and the mixture stirred overnight or until a solution formed. The mixture was then made up to 25 ml with water to give a solution containing 4 mg/ml buprenorphine, 20 mg/ml pectin and 15, 50, 87, 122, 157 or 192 mg/ml dextrose (or 15, 50, 87, 122 mg/ml mannitol) and the pH, appearance, osmolality, viscosity were determined. In addition, spray characteristics from a Pfeiffer multi-dose nasal spray device (standard nozzle, 0.1 ml pump, Cat. No. 62897) were evaluated by measurement of plume angle using image analysis. Gels were prepared by controlled mixing of 20 ml of formulation with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl2.2H20) before standing for 1 hour at room temperature. A visual assessment of the structure, uniformity, clarity and evidence of syneresis of each gel was conducted and, in addition, the gel structure was examined with a Stable Microsystems Texture Analyser.
  • 2. Results
  • As dextrose concentration increased from 15 to 50 mg/ml spray characteristics from a nasal spray device were affected as indicated by a decrease in plume angle associated with an increase in viscosity: a narrow plume was consistently obtained above 50 mg/ml dextrose (Table 5). As mannitol concentration increased there was a slight increase in viscosity and a slight decrease in plume angle (Table 6).
  • Gel structure may have been slightly weakened as dextrose concentration increased. This was indicated by a visual assessment but texture analysis results were inconclusive (Table 7).
  • Gel structure was affected at higher mannitol concentration. Visual assessment and texture analysis indicated that less uniform and weaker gels were produced (Table 8).
  • TABLE 5
    Osmolality, viscosity and spray characteristics of
    4.3 mg/ml BPN•HCl/20 mg/ml pectin (Slendid 100)
    solution containing varying concentrations of dextrose.
    Dextrose
    Batch concentration Osmolality Viscosity Plume
    No. (mg/ml) (osmol/kg) (cps) angle (°)
    114 15 0.15 8.0 43
    115 50 0.37 9.2 30
    116 87 0.62 10.3 22
    117 122 0.88 11.5 19
    118 157 1.18 13.0 23
    119 192 1.5 14.5 17
  • TABLE 6
    Osmolality, viscosity and spray characteristics of
    4.3 mg/ml BPN•HCl/20 mg/ml pectin (Slendid 100)
    solution containing varying concentrations of mannitol.
    Batch Mannitol conc. Osmolality Viscosity Plume
    No. (mg/ml) (osmol/kg) (cps) angle (°)
    120 15 0.16 8.4 33
    121 50 0.37 9.2 22
    122 87 0.61 10.3 22
    123 122  0.85 11.3 21
    124 157*
    125 192*
    *Did not dissolve
  • TABLE 7
    Gelling properties of 4.3 mg/ml BPN•HCl/20 mg/ml pectin (Slendid
    100) solution containing varying concentrations of dextrose.
    Texture analysis
    Batch Osmolality Force Area
    No. (osmol/kg) (g) (g s) Visual assessment
    114 0.15 574 10338 Very strong, uniform, semi-
    transparent pale yellow gel with
    minimum syneresis.
    115 0.37 359 6589 Very strong, uniform, semi-
    transparent pale yellow gel with
    minimum syneresis.
    116 0.62 280 5520 Strong, uniform, semi-
    transparent pale yellow gel with
    minimum syneresis.
    117 0.88 336 5019 Strong, uniform, semi-
    transparent pale yellow gel with
    minimum syneresis.
    118 1.18 467 7066 Strong, uniform, semi-
    transparent pale yellow gel with
    minimum syneresis.
    119 1.5 249 3435 Strong, uniform, semi-
    transparent pale yellow gel with
    some syneresis.
  • TABLE 8
    Gelling properties of 4.3 mg/ml BPN•HCl/20 mg/ml pectin
    (Slendid 100) solution containing varying concentrations of
    mannitol when mixed with a standard calcium chloride solution.
    Texture analysis
    Batch Osmolality Force Area
    No. (osmol/kg) (g) (g s) Visual assessment
    120 0.16 477 9006 Strong, uniform, semi-
    transparent pale yellow gel with
    some syneresis.
    121 0.37 497 8991 Strong, uniform, semi-
    transparent pale yellow gel with
    some syneresis.
    122 0.61 358 7160 Weak, non-uniform, semi-
    transparent pale yellow gel with
    some syneresis
    123 0.85 221 3881 Weak, non-uniform, semi-
    transparent pale yellow gel with
    some syneresis
  • Effect of Dextrose and Mannitol Concentration on Buprenorphine Solubility 1. Methods
  • Solutions containing pectin (Slendid 100) (20 mg/ml) were prepared at pH 3, 4, 5 and 6 (pH adjustments were made with 0.1M HCl or 0.1M meglumine). Into 5 ml of each solution was dissolved 0, 62.5, 125, 187.5 or 200 mg anhydrous dextrose or mannitol to give approximate dextrose/mannitol concentrations of 0, 12.5, 25, 37.5 or 50 mg/ml respectively. An excess of buprenorphine hydrochloride was then added and the mixture stirred overnight at 18° C. Saturated buprenorphine hydrochloride solutions were produced by passing each mixture through a 0.2 μm polycarbonate membrane filter. The concentration of buprenorphine hydrochloride in the filtrate was determined by hplc.
  • 2. Results
  • Buprenorphine solubility in aqueous solution containing 20 mg/ml pectin was not affected significantly by dextrose (Table 9) or mannitol (Table 10) concentration across the measured pH range.
  • TABLE 9
    Effect of dextrose concentration on the solubility of BPN•HCl
    in solution containing 20 mg/ml pectin (Slendid 100).
    Dextrose Buprenorphine
    concentration pH detected*
    Batch No. (mg/ml) (actual) (mg/ml)
    036 0 3 (3.1) 13.7
    040 12.5 3 (2.9) 13.1
    041 25 3 (2.7) 13.9
    042 37.5 3 (3.0) 13.9
    043 50 3 (3.2) 12.3
    037 0 4 (3.9) 16.4
    045 12.5 4 (3.8) 16.0
    046 25 4 (3.9) 15.6
    047 37.5 4 (4.0) 15.6
    048 50 4 (4.0) 14.3
    038 0 5 (4.9) 4.4
    050 12.5 5 (5.0) 5.7
    051 25 5 (5.0) 4.4
    052 37.5 5 (5.1) 4.4
    053 50 5 (5.2) 4.6
    039 0 6 (5.9) 1.8
    055 12.5 6 (5.7) 1.6
    056 25 6 (5.8) 1.7
    057 37.5 6 (5.7) 1.7
    058 50 6 (5.6) 1.8
    *Expressed as buprenorphine free base
  • TABLE 10
    Effect of mannitol concentration on the solubility of BPN•HCl
    in solution containing 20 mg/ml pectin (Slendid 100).
    Mannitol Buprenorphine
    concentration pH detected*
    Batch No. (mg/ml) (actual) (mg/ml)
    036 0 3 (3.2) 13.7
    060 12.5 3 (3.1) 13
    061 25 3 (3.1) 12.3
    062 37.5 3 (3.0) 12.4
    063 50 3 (3.2) 13.9
    037 0 4 (3.9) 16.4
    065 12.5 4 (4.0) 16.4
    066 25 4 (4.0) 15.8
    067 37.5 4 (4.0) 15.7
    068 50 4 (4.0) 15.5
    038 0 5 (4.9) 4.4
    070 12.5 5 (5.1) 4.9
    071 25 5 (5.2) 3.9
    072 37.5 5 (5.1) 4.6
    073 50 5 (5.2) 4.3
    039 0 6 (5.9) 1.8
    075 12.5 6 (5.9) 2.0
    076 25 6 (5.6) 1.9
    077 37.5 6 (5.6) 2.4
    078 50 6 (5.3) 1.8
    *Expressed as buprenorphine free base

    Negative Control Experiment: Effect of Mixing HM (High Methoxy) Pectin (20 mg/ml Genu (Trade Mark) Pectin [Citrus] Type USP-H) Solution with Calcium
  • The pectins suitable for retaining drugs at mucosal surfaces have a low degree of esterification (also called “low methoxy” or “LM” pectins) and, in aqueous solution, will gel in the presence of ions found in mucosal fluid, especially divalent ions, in particular calcium. As a negative control, a solution of “high methoxy” pectin was prepared and mixed with a solution containing calcium ions.
  • 1. Methods
  • Buprenorphine hydrochloride (107.5 mg), anhydrous dextrose (1.25 g) and pectin (Genu pectin [citrus] type USP-H; CP Kelco, Lille Skenved, Denmark) (500 mg) were stirred in 18-20 ml water in a 25 ml volumetric flask overnight or until a solution formed. The mixture was then made up to 25 ml with water to give a solution containing 4 mg/ml buprenorphine, 20 mg/ml pectin and 50 mg/ml dextrose and the pH and osmolality were determined. A 20 ml aliquot of the formulation was mixed (under controlled conditions) with 5 ml of a standard calcium chloride solution (9.44 mg/ml CaCl2.2H2O) before standing for 1 hour at room temperature. The structure, uniformity and clarity of the product were then evaluated.
  • 2. Results
  • The solution had a pH of 3.3 and an osmolality of 0.35 osmol/kg. An opaque, pale yellow solution was formed when the solution was mixed with 9.44 mg/ml CaCl2.2H20. The solution did not gel even when left for 1 hour at room temperature.
  • Example 8 Clinical Study
  • Unit doses of the intranasal buprenorphine formulations of Examples 1, 3 and (Formulations A to C) and one intravenous commercial buprenorphine formulation (Temgesic-trade mark; Formulation D) were administered to healthy human volunteers. The unit doses administered to the volunteers were as follows:
      • 800 μg buprenorphine hydrochloride, calculated as buprenorphine, of Formulations A, B or C administered intranasally; and
      • a single slow intravenous injection of 400 μg buprenorphine hydrochloride, calculated as buprenorphine, of Formulation D.
  • The dosing was performed on twelve healthy volunteers using a randomised, complete crossover design. Each dose was separated by, at least, seven days. The volunteers were required to fast overnight prior to dosing. Subjects were admitted to a clinic the evening before each dose of administration and remained in the clinic until blood sample collection for each study day. Blood samples were collected at regular intervals up to 24 hours after each dose administration. The volunteers were discharged from the clinic after completion of all 24 hour study procedures. There was a wash out period of, at least seven days, between each dose.
  • The pharmacokinetics of each dosage regimen were evaluated. The results are shown in FIGS. 1 to 3. All three intranasal solutions showed similar pharmacokinetic profiles. The Cther was reached within 5 to 10 minutes for each formulation and the Cmax was reached in 20 minutes or less. The data indicated that the initial plasma peak was blunted for the intranasal formulations compared to intravenous administration. That appeared most pronounced for Formulation A. All three intranasal solutions gave high bioavailability (Table 11)
  • TABLE 11
    Comparison of key pharmacokinetic parameters derived from the clinical
    study data on intranasal buprenorphine with published data on the sublingual
    tablet and with a dextrose formulation of buprenorphine.
    Clinical study data (prior art) (prior art)
    Intranasal Buprenorphine sublingual intranasal
    0.8 mg 0.8 mg Buprenorphine Buprenorphine
    PK 0.8 mg Chitosan/ Chitosan/ 0.4 mg 0.5 mg 0.3 mg dextrose
    Parameter Pectin HPMC Poloxamer tablet tablet solution
    Cmax (ng/ml) 3.7 4.4 3.8 0.5 1.04 1.8
    Tmax (min) 20 18 20 210 192 31
    Bioavailability 80% 81% 72% 56% 48%
  • A pharmacokinetic profile was computed for a 400 μg intranasal dose of Formulation A, calculated as buprenorphine, from the data for the 800 μg dose of Formulation A. This profile is shown in FIG. 4. FIG. 4 also shows the pharmacokinetic profile for the 400 μg dose of Formulation A that was administered intravenously.

Claims (48)

1.-12. (canceled)
13. A pharmaceutical composition suitable for use as an analgesic which comprises an analgesic and a delivery agent whereby, on introduction into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration Cther of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours.
14.-15. (canceled)
16. An aqueous solution suitable for intranasal administration, which comprises:
(a) from 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof,
(b) from 0.1 to 20 mg/ml of a chitosan, and
(c) from 0.1 to 15 mg/ml of hydroxypropylmethylcellulose;
which solution has a pH of from 3 to 4.8.
17. A solution according to claim 16, wherein the
hydroxypropylmethylcellulose has an apparent viscosity of from 3000 to 6000 cps and is present in an amount of from 0.1 to 15 mg/ml.
18. A solution according to claim 17, wherein the hydroxypropylmethylcellulose is present in an amount of from 0.5 to 10 mg/ml.
19. An aqueous solution suitable for intranasal administration, which comprises:
(a) from 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof,
(b) from 0.1 to 20 mg/ml of a chitosan, and
(c) from 50 to 200 mg/ml of a polyoxyethylene-polyoxypropylene copolymer of the general formula HO(C2H4O)a(C3H6O)b(C2H4O)aH wherein a is from 2 to 130 and b is from 15 to 67;
which solution has a pH of from 3 to 4.8.
20. A solution according to claim 19, wherein the polyoxyethylene-polyoxypropylene copolymer is present in an amount of from 80 to 120 mg/ml.
21. A solution according to claim 19, wherein the polyoxyethylene-polyoxypropyene copolymer has a molecular weight of from 7,000 to 10,000.
22. A solution according to claim 19, wherein the polyoxyethylene-polyoxypropylene copolymer is one in which a is 80 and b is 27.
23. A solution according to claim 16, which has an osmolality of from 0.32 to 0.4 osmol/kg.
24. A solution according to claim 16, wherein the buprenorphine or buprenorphine salt or ester is present in an amount of from 0.5 to 8 mg/ml.
25. A solution according to claim 24, wherein the buprenorphine or buprenorphine salt or ester is present in an amount of from 1 to 6 mg/ml calculated as buprenorphine.
26. A solution according to claim 16, which comprises buprenorphine hydrochloride.
27. A solution according to claim 16, wherein the chitosan is present in an amount of from 2 to 10 mg/ml.
28. A solution according to claim 16, wherein the chitosan is a physiologically acceptable salt of a deacetylated chitin
29. A solution according to claim 28, wherein the salt is chitosan glutamate.
30. A solution according to claim 16, wherein the pH is from 3.2 to 3.8.
31. A solution according to claim 16, wherein the pH has been adjusted by means of hydrochloric acid.
32. A solution according to claim 16, which comprises a preservative.
33. A solution according to claim 32, wherein the preservative is benzalkonium chloride.
34. A solution according to claim 16, which contains dextrose as a tonicity adjustment agent.
35.-37. (canceled)
38. An aqueous solution suitable for intranasal administration, which comprises from 0.1 to 10 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof and from 5 to 40 mg/ml of a pectin having a degree of esterification of less than 50%; which solution has a pH of from 3 to 4.2, is substantially free from divalent metal ions and gels on the nasal mucosa.
39. A solution according to claim 38, wherein the buprenorphine or buprenorphine salt or ester is present in an amount of from 0.5 to 8 mg/ml.
40. A solution according to claim 39, wherein the buprenorphine or buprenorphine salt or ester is present in an amount of from 1 to 6 mg/ml calculated as buprenorphine.
41. A solution according to claim 38, which comprises buprenorphine hydrochloride.
42. A solution according to any claim 38, wherein the pectin is present in an amount of from 10 to 30 mg/ml.
43. A solution according to claim 38, wherein the pectin has a degree of esterification of from 10 to 35%.
44. A solution according to claim 38, wherein the pH is from 3.2 to 3.8.
45. A solution according to claim 38, wherein the pH has been adjusted by means of hydrochloric acid.
46. A solution according to claim 38, which comprises a preservative.
47. A solution according to claim 46, which comprises phenylethyl alcohol and propyl hydroxybenzoate as preservatives.
48. A solution according to claim 38, which has an osmolality of from 0.25 to 0.4 osmol/kg.
49. A solution according to claim 38, which contains dextrose as a tonicity adjustment agent.
50. An aqueous solution suitable for intranasal administration, which has a pH of from 3.5 to 4.0, which is substantially free from divalent metal ions and which comprises:
(a) from 1 to 6 mg/ml of buprenorphine or a physiologically acceptable salt or ester thereof, calculated as buprenorphine,
(b) from 10 to 40 mg/ml of a pectin which has a degree of esterification from 10 to 35%, and
(c) dextrose as a tonicity adjustment agent.
51.-52. (canceled)
53. A nasal delivery device loaded with a solution as claimed in claim 16.
54. A device according to claim 53, which is a spray device.
55.-56. (canceled)
57. A method of inducing analgesia in a patient in need thereof, which method comprises administering intranasally to said patient a pharmaceutical composition, which comprises an opioid analgesic and a delivery agent in the form of an aqueous solution;
whereby, on introduction of said composition into the nasal cavity of the patient to be treated, the analgesic is delivered to the bloodstream to produce within 30 minutes a therapeutic plasma concentration Cther of 0.2 ng/ml or greater which is maintained for a duration Tmaint of at least 2 hours, and
wherein the delivery agent is from 5 to 40 mg/ml, based on the composition, of a pectin having a degree of esterification of less than 50% provided that the composition is substantially free of divalent metal ions.
58. A method according to claim 57, wherein on introduction of the composition into the nasal cavity of the patient to be treated, the analgesic is delivered to the bloodstream to produce within 2 to 15 minutes a therapeutic plasma concentration Cther of 0.4 ng/ml or more which is maintained for a duration Tmaint of 2 to 4 hours.
59. A method according to claim 57, wherein the pectin has a degree of esterification of from 10 to 35%.
60. A method according to claim 57, wherein Cther is from 0.4 to 100 ng/ml and is produced within 1 to 15 minutes.
61. A method according to claim 57, wherein Cmax is reached 10 to 30 minutes after introduction of said composition into the nasal cavity of the patient to be treated.
62. A method according to claim 57, wherein the composition is delivered using a nasal delivery device.
63. A method according to claim 62, wherein on introduction of the composition into the nasal cavity of a patient to be treated, the analgesic is delivered to the bloodstream to produce within 2 to 15 minutes a therapeutic plasma concentration Cther of 0.4 ng/ml or more which is maintained for a duration Tmaint of 2 to 4 hours.
64. A method according to claim 57, wherein the opioid analgesic is fentanyl.
US12/929,279 2002-03-19 2011-01-12 Analgesics for nasal administration Abandoned US20110105551A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/929,279 US20110105551A1 (en) 2002-03-19 2011-01-12 Analgesics for nasal administration

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
GB0206448.3 2002-03-19
GB0206448A GB0206448D0 (en) 2002-03-19 2002-03-19 Analgesics
GB0225042A GB0225042D0 (en) 2002-10-28 2002-10-28 Pharmaceutical composition
GB0225042.1 2002-10-28
GB0225040.5 2002-10-28
GB0225041.3 2002-10-28
GB0225040A GB0225040D0 (en) 2002-10-28 2002-10-28 Formulation
GB0225041A GB0225041D0 (en) 2002-10-28 2002-10-28 Pharmaceutical formulation
PCT/GB2003/001184 WO2003080022A2 (en) 2002-03-19 2003-03-19 Analgesics for nasal administration
US10/508,315 US20050142072A1 (en) 2002-03-19 2003-03-19 Analgesics for nasal administration
US11/798,384 US20070231269A1 (en) 2002-03-19 2007-05-14 Analgesics
US12/929,279 US20110105551A1 (en) 2002-03-19 2011-01-12 Analgesics for nasal administration

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/798,384 Continuation US20070231269A1 (en) 2002-03-19 2007-05-14 Analgesics

Publications (1)

Publication Number Publication Date
US20110105551A1 true US20110105551A1 (en) 2011-05-05

Family

ID=28457822

Family Applications (5)

Application Number Title Priority Date Filing Date
US10/508,336 Expired - Fee Related US7666876B2 (en) 2002-03-19 2002-03-19 Buprenorphine formulations for intranasal delivery
US10/508,315 Abandoned US20050142072A1 (en) 2002-03-19 2003-03-19 Analgesics for nasal administration
US11/798,384 Abandoned US20070231269A1 (en) 2002-03-19 2007-05-14 Analgesics
US12/588,528 Abandoned US20100041624A1 (en) 2002-03-19 2009-10-19 Buprenorphine formulations for intranasal delivery
US12/929,279 Abandoned US20110105551A1 (en) 2002-03-19 2011-01-12 Analgesics for nasal administration

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US10/508,336 Expired - Fee Related US7666876B2 (en) 2002-03-19 2002-03-19 Buprenorphine formulations for intranasal delivery
US10/508,315 Abandoned US20050142072A1 (en) 2002-03-19 2003-03-19 Analgesics for nasal administration
US11/798,384 Abandoned US20070231269A1 (en) 2002-03-19 2007-05-14 Analgesics
US12/588,528 Abandoned US20100041624A1 (en) 2002-03-19 2009-10-19 Buprenorphine formulations for intranasal delivery

Country Status (19)

Country Link
US (5) US7666876B2 (en)
EP (3) EP1878446A3 (en)
JP (3) JP2005527535A (en)
KR (2) KR101030403B1 (en)
CN (1) CN1642577B (en)
AT (2) ATE422902T1 (en)
AU (2) AU2003219282A1 (en)
BR (1) BRPI0308527B1 (en)
CA (2) CA2479718A1 (en)
DE (2) DE60326231D1 (en)
DK (2) DK1513493T3 (en)
ES (2) ES2315485T3 (en)
GB (1) GB2397016B (en)
IL (2) IL164129A0 (en)
MX (1) MXPA04009055A (en)
NZ (2) NZ535973A (en)
PT (1) PT1513493E (en)
SI (1) SI1499360T1 (en)
WO (2) WO2003080022A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8889176B2 (en) 2003-01-10 2014-11-18 Depomed, Inc. Method of managing or treating pain
US9192570B2 (en) 2013-12-20 2015-11-24 AntiOP, Inc. Intranasal naloxone compositions and methods of making and using same
EP3725303A1 (en) * 2019-04-18 2020-10-21 S.I.I.T. S.r.l.-Servizio Internazionale Imballaggi Termosaldanti Method for preparing a plant-based fungal chitosan product
WO2021225973A1 (en) * 2020-05-04 2021-11-11 Amphastar Pharmaceuticals, Inc. Naloxone pharmaceutical formulations for intranasal (in) delivery

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10141650C1 (en) * 2001-08-24 2002-11-28 Lohmann Therapie Syst Lts Safe transdermal therapeutic system for administration of fentanyl or analogous analgesics, having matrix layer of carboxy group-free polyacrylate adhesive providing high permeation rate
US7666876B2 (en) * 2002-03-19 2010-02-23 Vernalis (R&D) Limited Buprenorphine formulations for intranasal delivery
GB0315632D0 (en) 2003-07-04 2003-08-13 West Pharm Serv Drug Res Ltd Pharmaceutical formulations
GB0328186D0 (en) * 2003-12-05 2004-01-07 West Pharm Serv Drug Res Ltd Intranasal compositions
US20050129679A1 (en) 2003-12-15 2005-06-16 Nastech Pharmaceutical Company Inc. Method for opening tight junctions
GB0329918D0 (en) * 2003-12-24 2004-01-28 West Pharm Serv Drug Res Ltd Intranasal compositions
ES2399053T3 (en) * 2004-10-11 2013-03-25 Nasaleze Patents Limited Compositions for intranasal administration
ATE404206T1 (en) * 2004-12-06 2008-08-15 Janssen Pharmaceutica Nv ORAL SUSPENSION WITH MELOXICAM
DE602006012513D1 (en) * 2005-08-26 2010-04-08 Healthpartners Res Foundation METHOD FOR THE TREATMENT OF HEADACHES BY ADMINISTERING OXYTOCIN
GB0606124D0 (en) * 2006-03-28 2006-05-03 Reckitt Benckiser Healthcare Buprenorphine derivatives and uses thereof
WO2007115975A2 (en) * 2006-04-04 2007-10-18 Holger Lars Hermann USE OF kappa OPIOID RECEPTOR ANTAGONIST-CONTAINING COMPOSITIONS FOR THE TREATMENT OF DISSOCIATIVE DISORDERS
RS54764B1 (en) 2006-07-21 2016-10-31 Biodelivery Sciences Int Inc Transmucosal delivery devices with enhanced uptake
EP1897543A1 (en) 2006-08-30 2008-03-12 Euro-Celtique S.A. Buprenorphine- wafer for drug substitution therapy
US20090270438A1 (en) * 2006-10-18 2009-10-29 Clive Booles Novel compositions and formulations
EP2130539A4 (en) * 2007-04-02 2010-05-19 Toyo Boseki Therapeutic tablet for postherpetic neuralgia and method of treating postherpetic neuralgia
EP2057982A1 (en) * 2007-11-09 2009-05-13 Archimedes Development Limited Intranasal compositions
JP2011511674A (en) 2008-02-07 2011-04-14 ユニヴァーシティ オブ ワシントン Circumferential aerosol device
CN101297973B (en) * 2008-05-22 2010-06-09 武汉华纳生物工程有限公司 Highly bioadhesive and thermosensitive hydrogel, and preparation method and application thereof
WO2010072398A2 (en) * 2008-12-22 2010-07-01 Boehringer Ingelheim Limited Veterinary formulations
DE102009001041A1 (en) * 2009-02-20 2010-08-26 Marien-Apotheke Fulda Ohg Emergency kit for the manufacture of a medicament for the nasal use of opioids in respiratory distress
DE102009024542A1 (en) 2009-06-10 2010-12-16 Arivine Pharma Ag Compositions based on chitosan oligosaccharides
SG177752A1 (en) * 2009-07-27 2012-03-29 Trigemina Inc Methods for treatment of pain
US8475832B2 (en) 2009-08-07 2013-07-02 Rb Pharmaceuticals Limited Sublingual and buccal film compositions
CZ302789B6 (en) 2009-11-25 2011-11-09 Zentiva, K. S. Method of increasing solubility of pharmaceutically active compounds and targeted (controlled) transport thereof into intestine
WO2012077024A1 (en) * 2010-12-08 2012-06-14 Mrinmayee Kondhalkar Composition for inhibiting the growth of mammalian hair
JP6339371B2 (en) 2011-03-03 2018-06-06 インペル ニューロファーマ インコーポレイテッド Nasal drug delivery device
CN103619485B (en) 2011-05-09 2017-08-08 英倍尔药业股份有限公司 Nozzle for nasal medicament delivery
AR086391A1 (en) 2011-05-13 2013-12-11 Euro Celtique Sa INTRANASAL PHARMACEUTICAL DOSAGE FORMS THAT INCLUDE NALOXONE
ES2712072T3 (en) * 2011-07-20 2019-05-09 Torrent Pharmaceuticals Ltd Pharmaceutical composition of tapentadol
ES2660116T3 (en) 2011-08-18 2018-03-20 Biodelivery Sciences International, Inc. Mucoadhesive devices resistant to improper use for buprenorphine administration
WO2013040295A2 (en) * 2011-09-14 2013-03-21 University Of South Florida Divalent-metal coated nanoparticles for delivery of compositions into the central nervous system by nasal insufflation
US9901539B2 (en) 2011-12-21 2018-02-27 Biodelivery Sciences International, Inc. Transmucosal drug delivery devices for use in chronic pain relief
EP2991713B1 (en) 2013-04-28 2019-06-19 Impel Neuropharma Inc. Medical unit dose container
WO2015038327A1 (en) * 2013-09-10 2015-03-19 Insys Pharma, Inc. Sublingual buprenorphine spray
US9867818B2 (en) 2013-09-10 2018-01-16 Insys Development Company, Inc. Sublingual buprenorphine spray
US9839611B2 (en) 2013-09-10 2017-12-12 Insys Development Company, Inc. Sublingual buprenorphine spray
US9918981B2 (en) 2013-09-10 2018-03-20 Insys Development Company, Inc. Liquid buprenorphine formulations
EP3129013A1 (en) * 2014-04-10 2017-02-15 Crowley, Patrick Delivery of non-steroidal antiinflammatory agents to the brain via the nasal tract to treat neurological disorders
WO2015195708A1 (en) * 2014-06-16 2015-12-23 Loewi LLC Methods of anesthetizing nerve tissue in the trigeminal nerve pathway and medical uses thereof
MX2017008913A (en) 2015-01-07 2018-04-30 Trigemina Inc Magnesium-containing oxytocin formulations and methods of use.
CN107847518A (en) * 2015-07-10 2018-03-27 帝斯曼知识产权资产管理有限公司 For preventing and treating the food and/or fodder compound of inflammatory disease
US11266799B2 (en) 2015-09-10 2022-03-08 Impel Neuropharma, Inc. In-line nasal delivery device
BR112019008263A2 (en) * 2016-10-28 2019-07-09 Servier Lab liposomal formulation for use in cancer treatment
CA3077627A1 (en) * 2017-10-20 2019-04-25 Chiesi Farmaceutici S.P.A. Pharmaceutical formulations comprising opioid receptor agonist as active ingredients, methods of manufacture and therapeutic uses thereof
US11571532B2 (en) 2017-11-21 2023-02-07 Impel Pharmaceuticals Inc. Intranasal device with dip tube
JP2021503988A (en) 2017-11-21 2021-02-15 インペル ニューロファーマ インコーポレイテッド Intranasal device with inlet interface
JP2021509677A (en) 2018-01-05 2021-04-01 インペル ニューロファーマ インコーポレイテッド Internasal delivery of olanzapine by precision olfactory device
CA3088942C (en) 2018-01-05 2023-01-03 Impel Neuropharma, Inc. Intranasal delivery of dihydroergotamine by precision olfactory device
KR20210034047A (en) 2018-07-19 2021-03-29 임펠 뉴로파마 인코포레이티드 Airway delivery of levodopa and dopa decarboxylase inhibitors for the treatment of Parkinson's disease
NZ777687A (en) 2019-01-03 2023-12-22 Impel Pharmaceuticals Inc Nasal drug delivery device
WO2020236658A1 (en) 2019-05-17 2020-11-26 Impel Neuropharma, Inc. Single-use nasal delivery device

Citations (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2125212A (en) * 1938-05-03 1938-07-26 Vick Chemical Company Therapeutic composition
US2730483A (en) * 1952-08-12 1956-01-10 Nepera Chemical Co Inc Therapeutic compositions comprising neomycin, gramicidin and quaternary ammonium salt of thonzylamine
US3164600A (en) * 1961-10-10 1965-01-05 Res Lab Dr C Janssen N V 1-aralkyl-4-(n-aryl-carbonyl amino)-piperidines and related compounds
US4464378A (en) * 1981-04-28 1984-08-07 University Of Kentucky Research Foundation Method of administering narcotic antagonists and analgesics and novel dosage forms containing same
US4486423A (en) * 1983-04-21 1984-12-04 Janssen Pharmaceutica Inc. Stable fentanyl composition
US4609640A (en) * 1981-11-17 1986-09-02 Toyo Jozo Company, Ltd. Preparation having excellent absorption property
US4613500A (en) * 1983-03-09 1986-09-23 Teijin Limited Powdery pharmaceutical composition for nasal administration
US4659696A (en) * 1982-04-30 1987-04-21 Takeda Chemical Industries, Ltd. Pharmaceutical composition and its nasal or vaginal use
US4681887A (en) * 1985-06-21 1987-07-21 Richter Gedeon Vegyeszeti Gyar Rt. Pharmaceutical compositions with a neuroleptic action and process for preparing same
US4806341A (en) * 1985-02-25 1989-02-21 Rutgers, The State University Of New Jersey Transdermal absorption dosage unit for narcotic analgesics and antagonists and process for administration
US4826683A (en) * 1987-01-09 1989-05-02 Bates Harry L Decongestant
US4861760A (en) * 1985-10-03 1989-08-29 Merck & Co., Inc. Ophthalmological composition of the type which undergoes liquid-gel phase transition
US4915948A (en) * 1987-08-31 1990-04-10 Warner-Lambert Company Tablets having improved bioadhesion to mucous membranes
US4981875A (en) * 1987-08-12 1991-01-01 Bayer Aktiengesellschaft Medicaments for the region of the oral cavity
US4983385A (en) * 1985-11-22 1991-01-08 Sunstar Kabushiki Kaisha Ointment base
US5147648A (en) * 1986-01-16 1992-09-15 Christian Bannert Method of improving the adhesiveness of gels to mucosae
US5200180A (en) * 1987-06-26 1993-04-06 Christian Bannert Pharmaceutical composition for the treatment of the human eye
US5236714A (en) * 1988-11-01 1993-08-17 Alza Corporation Abusable substance dosage form having reduced abuse potential
US5238917A (en) * 1989-09-11 1993-08-24 Teikoku Seiyaku Kabushiki Kaisha High-absorbable transvaginal preparation containing biologically active polypeptide
US5318780A (en) * 1991-10-30 1994-06-07 Mediventures Inc. Medical uses of in situ formed gels
US5346703A (en) * 1990-08-07 1994-09-13 Mediventures, Inc. Body cavity drug delivery with thermo-irreversible polyoxyalkylene and ionic polysaccharide gels
US5397771A (en) * 1990-05-10 1995-03-14 Bechgaard International Research And Development A/S Pharmaceutical preparation
US5457093A (en) * 1987-09-18 1995-10-10 Ethicon, Inc. Gel formulations containing growth factors
US5456745A (en) * 1988-08-13 1995-10-10 Lts Lohmann Therapie-Systeme Gmbh & Co. Kg Flexible, hydrophilic gel film, the process for its production and the use of it
US5508042A (en) * 1991-11-27 1996-04-16 Euro-Celtigue, S.A. Controlled release oxycodone compositions
US5543434A (en) * 1994-02-25 1996-08-06 Weg; Stuart L. Nasal administration of ketamine to manage pain
US5655517A (en) * 1994-03-29 1997-08-12 Electrosols, Ltd. Dispensing device
US5707644A (en) * 1989-11-04 1998-01-13 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5725263A (en) * 1997-04-03 1998-03-10 Rodriguez; Ernest L. Door securing device
US5756483A (en) * 1993-03-26 1998-05-26 Merkus; Franciscus W. H. M. Pharmaceutical compositions for intranasal administration of apomorphine
US5855517A (en) * 1995-07-27 1999-01-05 Flex-Elektrowerkzeuge Gmbh Releasable coupling for machine tools
US5910301A (en) * 1994-05-13 1999-06-08 Aradigm Corporation Method of intrapulmonary administration of a narcotic drug
US5955502A (en) * 1994-03-30 1999-09-21 Gs Development Ab Use of fatty acid esters as bioadhesive substances
US6090368A (en) * 1998-03-03 2000-07-18 The Board Of Governors For Higher Education, State Of Rhode Island And Providence Plantations Pharmaceutical compositions for intranasal spray administration of ketorolac tromethamine
US6387917B1 (en) * 1999-10-20 2002-05-14 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Salts of opioid analgesics, particularly morphine, and methods of using same
US6432440B1 (en) * 1997-04-18 2002-08-13 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Pectin compositions and methods of use for improved delivery of drugs to mucosal surfaces
US6541021B1 (en) * 1999-03-18 2003-04-01 Durect Corporation Devices and methods for pain management
US20030077300A1 (en) * 2000-05-10 2003-04-24 Wermeling Daniel P. System and method for intranasal administration of opioids
US6610271B2 (en) * 2000-05-10 2003-08-26 University Of Kentucky Research Foundation System and method for intranasal administration of lorazepam
US6663883B1 (en) * 1999-08-26 2003-12-16 Takeda Chemical Industries, Ltd. Matrix adhering to nasal mucosa
US6667279B1 (en) * 1996-11-13 2003-12-23 Wallace, Inc. Method and composition for forming water impermeable barrier
US6777000B2 (en) * 2001-02-28 2004-08-17 Carrington Laboratories, Inc. In-situ gel formation of pectin
US20040166067A1 (en) * 2003-01-10 2004-08-26 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Pharmaceutical compositions
US20050142072A1 (en) * 2002-03-19 2005-06-30 Birch Phillip J. Analgesics for nasal administration

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8904370D0 (en) 1989-02-25 1989-04-12 Cosmas Damian Ltd Liquid delivery compositions
GB9202464D0 (en) 1992-02-05 1992-03-18 Danbiosyst Uk Composition for nasal administration
EP0571671B1 (en) 1992-05-26 1997-09-17 The Procter & Gamble Company A Pharmaceutical composition in powder form
AU5551394A (en) 1992-11-09 1994-06-08 Pharmetrix Corporation Combined analgesic delivery methods for pain management
FR2733420B1 (en) 1995-04-28 1997-06-27 Sep Tarral PECTIC PREPARATIONS FOR USE AS MEDICINAL MEDIA
US5840731A (en) 1995-08-02 1998-11-24 Virginia Commonwealth University Pain-alleviating drug composition and method for alleviating pain
GB2328443B (en) 1997-08-21 2001-09-05 Reckitt & Colmann Prod Ltd In situ formation of pharmaceutically acceptable polymeric material
RS49982B (en) 1997-09-17 2008-09-29 Euro-Celtique S.A., Synergistic analgesic combination of opioid analgesic and cyclooxygenase-2 inhibitor
AU751182B2 (en) 1997-12-02 2002-08-08 Archimedes Development Limited Compositions for nasal administration
US6007841A (en) 1998-03-13 1999-12-28 Algos Pharmaceutical Corporation Analgesic composition and method for treating pain
AU5305899A (en) 1998-08-26 2000-03-21 Teijin Limited Powdery pernasal compositions
JP2000229859A (en) 1999-02-12 2000-08-22 Teijin Ltd Stable pernasal preparation of buprenorphine
JP2001002589A (en) 1999-06-18 2001-01-09 Teijin Ltd Composition for nasal administration enabling mild raising of medicine blood concentration
AR031682A1 (en) 1999-11-19 2003-10-01 Reckitt Benckiser Helthcare Uk PHARMACEUTICAL COMPOSITIONS
PT2092936E (en) 2000-02-08 2013-06-20 Euro Celtique Sa Tamper-resistant oral opioid agonist formulations
US20020013331A1 (en) 2000-06-26 2002-01-31 Williams Robert O. Methods and compositions for treating pain of the mucous membrane
GB2378383A (en) 2001-06-06 2003-02-12 Gursharan Moonga Nasal delivery of pharmaceutical compositions in powder form

Patent Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2125212A (en) * 1938-05-03 1938-07-26 Vick Chemical Company Therapeutic composition
US2730483A (en) * 1952-08-12 1956-01-10 Nepera Chemical Co Inc Therapeutic compositions comprising neomycin, gramicidin and quaternary ammonium salt of thonzylamine
US3164600A (en) * 1961-10-10 1965-01-05 Res Lab Dr C Janssen N V 1-aralkyl-4-(n-aryl-carbonyl amino)-piperidines and related compounds
US4464378A (en) * 1981-04-28 1984-08-07 University Of Kentucky Research Foundation Method of administering narcotic antagonists and analgesics and novel dosage forms containing same
US4609640A (en) * 1981-11-17 1986-09-02 Toyo Jozo Company, Ltd. Preparation having excellent absorption property
US4659696A (en) * 1982-04-30 1987-04-21 Takeda Chemical Industries, Ltd. Pharmaceutical composition and its nasal or vaginal use
US4613500A (en) * 1983-03-09 1986-09-23 Teijin Limited Powdery pharmaceutical composition for nasal administration
US4486423A (en) * 1983-04-21 1984-12-04 Janssen Pharmaceutica Inc. Stable fentanyl composition
US4806341A (en) * 1985-02-25 1989-02-21 Rutgers, The State University Of New Jersey Transdermal absorption dosage unit for narcotic analgesics and antagonists and process for administration
US4681887A (en) * 1985-06-21 1987-07-21 Richter Gedeon Vegyeszeti Gyar Rt. Pharmaceutical compositions with a neuroleptic action and process for preparing same
US4861760A (en) * 1985-10-03 1989-08-29 Merck & Co., Inc. Ophthalmological composition of the type which undergoes liquid-gel phase transition
US4983385A (en) * 1985-11-22 1991-01-08 Sunstar Kabushiki Kaisha Ointment base
US5147648A (en) * 1986-01-16 1992-09-15 Christian Bannert Method of improving the adhesiveness of gels to mucosae
US4826683A (en) * 1987-01-09 1989-05-02 Bates Harry L Decongestant
US5200180A (en) * 1987-06-26 1993-04-06 Christian Bannert Pharmaceutical composition for the treatment of the human eye
US4981875A (en) * 1987-08-12 1991-01-01 Bayer Aktiengesellschaft Medicaments for the region of the oral cavity
US4915948A (en) * 1987-08-31 1990-04-10 Warner-Lambert Company Tablets having improved bioadhesion to mucous membranes
US5457093A (en) * 1987-09-18 1995-10-10 Ethicon, Inc. Gel formulations containing growth factors
US5456745A (en) * 1988-08-13 1995-10-10 Lts Lohmann Therapie-Systeme Gmbh & Co. Kg Flexible, hydrophilic gel film, the process for its production and the use of it
US5236714A (en) * 1988-11-01 1993-08-17 Alza Corporation Abusable substance dosage form having reduced abuse potential
US5238917A (en) * 1989-09-11 1993-08-24 Teikoku Seiyaku Kabushiki Kaisha High-absorbable transvaginal preparation containing biologically active polypeptide
US5707644A (en) * 1989-11-04 1998-01-13 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5397771A (en) * 1990-05-10 1995-03-14 Bechgaard International Research And Development A/S Pharmaceutical preparation
US5346703A (en) * 1990-08-07 1994-09-13 Mediventures, Inc. Body cavity drug delivery with thermo-irreversible polyoxyalkylene and ionic polysaccharide gels
US5318780A (en) * 1991-10-30 1994-06-07 Mediventures Inc. Medical uses of in situ formed gels
US5508042A (en) * 1991-11-27 1996-04-16 Euro-Celtigue, S.A. Controlled release oxycodone compositions
US5756483A (en) * 1993-03-26 1998-05-26 Merkus; Franciscus W. H. M. Pharmaceutical compositions for intranasal administration of apomorphine
US5543434A (en) * 1994-02-25 1996-08-06 Weg; Stuart L. Nasal administration of ketamine to manage pain
US5655517A (en) * 1994-03-29 1997-08-12 Electrosols, Ltd. Dispensing device
US5955502A (en) * 1994-03-30 1999-09-21 Gs Development Ab Use of fatty acid esters as bioadhesive substances
US5910301A (en) * 1994-05-13 1999-06-08 Aradigm Corporation Method of intrapulmonary administration of a narcotic drug
US5855517A (en) * 1995-07-27 1999-01-05 Flex-Elektrowerkzeuge Gmbh Releasable coupling for machine tools
US6667279B1 (en) * 1996-11-13 2003-12-23 Wallace, Inc. Method and composition for forming water impermeable barrier
US5725263A (en) * 1997-04-03 1998-03-10 Rodriguez; Ernest L. Door securing device
US20070092535A1 (en) * 1997-04-18 2007-04-26 Archimedes Development Limited Delivery of drugs to mucosal surfaces
US6432440B1 (en) * 1997-04-18 2002-08-13 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Pectin compositions and methods of use for improved delivery of drugs to mucosal surfaces
US20020197324A1 (en) * 1997-04-18 2002-12-26 West Pharmaceutical Drug Delivery And Research Centre Limited Delivery of drugs to mucosal surfaces
US20110182857A1 (en) * 1997-04-18 2011-07-28 Archimedes Development Limited Delivery of Drugs to Mucosal Surfaces
US20070110677A1 (en) * 1997-04-18 2007-05-17 Archimedes Development Limited Delivery of drugs to mucosal surfaces
US6090368A (en) * 1998-03-03 2000-07-18 The Board Of Governors For Higher Education, State Of Rhode Island And Providence Plantations Pharmaceutical compositions for intranasal spray administration of ketorolac tromethamine
US6541021B1 (en) * 1999-03-18 2003-04-01 Durect Corporation Devices and methods for pain management
US6663883B1 (en) * 1999-08-26 2003-12-16 Takeda Chemical Industries, Ltd. Matrix adhering to nasal mucosa
US6387917B1 (en) * 1999-10-20 2002-05-14 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Salts of opioid analgesics, particularly morphine, and methods of using same
US20030077300A1 (en) * 2000-05-10 2003-04-24 Wermeling Daniel P. System and method for intranasal administration of opioids
US6610271B2 (en) * 2000-05-10 2003-08-26 University Of Kentucky Research Foundation System and method for intranasal administration of lorazepam
US6777000B2 (en) * 2001-02-28 2004-08-17 Carrington Laboratories, Inc. In-situ gel formation of pectin
US20050142072A1 (en) * 2002-03-19 2005-06-30 Birch Phillip J. Analgesics for nasal administration
US20070231269A1 (en) * 2002-03-19 2007-10-04 Ionix Pharmaceuticals Limited Analgesics
US20040166067A1 (en) * 2003-01-10 2004-08-26 West Pharmaceutical Services Drug Delivery & Clinical Research Centre Limited Pharmaceutical compositions
US8216604B2 (en) * 2003-01-10 2012-07-10 Archimedes Development Limited Method of managing or treating pain

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8889176B2 (en) 2003-01-10 2014-11-18 Depomed, Inc. Method of managing or treating pain
US9078814B2 (en) 2003-01-10 2015-07-14 Depomed, Inc. Intranasal spray device containing pharmaceutical composition
US9814705B2 (en) 2003-01-10 2017-11-14 Depomed, Inc. Intranasal spray device containing pharmaceutical composition
US9192570B2 (en) 2013-12-20 2015-11-24 AntiOP, Inc. Intranasal naloxone compositions and methods of making and using same
US9289425B2 (en) 2013-12-20 2016-03-22 AntiOP, Inc. Intranasal naloxone compositions and methods of making and using same
EP3725303A1 (en) * 2019-04-18 2020-10-21 S.I.I.T. S.r.l.-Servizio Internazionale Imballaggi Termosaldanti Method for preparing a plant-based fungal chitosan product
WO2021225973A1 (en) * 2020-05-04 2021-11-11 Amphastar Pharmaceuticals, Inc. Naloxone pharmaceutical formulations for intranasal (in) delivery

Also Published As

Publication number Publication date
GB2397016B (en) 2004-10-27
WO2003080022A3 (en) 2004-05-13
SI1499360T1 (en) 2009-04-30
BR0308527A (en) 2005-02-01
IL164129A0 (en) 2005-12-18
ATE422902T1 (en) 2009-03-15
EP1513493B8 (en) 2009-04-22
GB0318346D0 (en) 2003-09-10
KR101030403B1 (en) 2011-04-20
GB2397016A (en) 2004-07-14
KR20050000371A (en) 2005-01-03
JP2005526094A (en) 2005-09-02
US7666876B2 (en) 2010-02-23
ES2315485T3 (en) 2009-04-01
US20100041624A1 (en) 2010-02-18
DK1513493T3 (en) 2009-05-18
EP1513493B1 (en) 2009-02-18
NZ535973A (en) 2007-09-28
CA2479718A1 (en) 2003-10-02
PT1513493E (en) 2009-05-06
NZ535972A (en) 2006-07-28
EP1878446A3 (en) 2008-08-27
US20050142072A1 (en) 2005-06-30
EP1513493A2 (en) 2005-03-16
CA2479711C (en) 2011-11-29
IL164129A (en) 2011-04-28
DE60326231D1 (en) 2009-04-02
US20070231269A1 (en) 2007-10-04
WO2003080022A2 (en) 2003-10-02
EP1499360A2 (en) 2005-01-26
JP2005527535A (en) 2005-09-15
AU2003216842B2 (en) 2007-12-13
CA2479711A1 (en) 2003-10-02
CN1642577A (en) 2005-07-20
AU2003216842A1 (en) 2003-10-08
JP4728580B2 (en) 2011-07-20
WO2003080021A2 (en) 2003-10-02
CN1642577B (en) 2010-05-12
EP1878446A2 (en) 2008-01-16
MXPA04009055A (en) 2005-07-27
EP1499360B1 (en) 2008-10-08
WO2003080021A3 (en) 2003-12-31
BRPI0308527B1 (en) 2015-07-07
ATE410187T1 (en) 2008-10-15
ES2325364T3 (en) 2009-09-02
DE60323958D1 (en) 2008-11-20
DK1499360T3 (en) 2009-02-09
JP2010174029A (en) 2010-08-12
AU2003219282A1 (en) 2003-10-08
KR20110005919A (en) 2011-01-19
US20050085440A1 (en) 2005-04-21

Similar Documents

Publication Publication Date Title
EP1513493B1 (en) Analgesics
JP6657454B2 (en) Abuse resistant mucoadhesive delivery device for buprenorphine
JP3903061B2 (en) Nanoparticles containing drug, method for producing the same, and preparation for parenteral administration comprising the nanoparticles
CA2567075C (en) Pharmaceutical suspension composition
JP2002509103A (en) Oral liquid composition
CA2511974C (en) Pharmaceutical compositions comprising fentanyl for intranasal delivery
JP5529165B2 (en) Formulation for oral mucosal administration of lipid-lowering drugs
ZA200407541B (en) Analgesics for nasal administration
CN113490485A (en) Film for oral cavity

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARCHIMEDES DEVELOPMENT LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:VERNALIS (R&D) LIMITED;REEL/FRAME:028369/0521

Effective date: 20120327

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION