US20120034297A1 - Pharmaceutical dosage forms comprising 6'-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine - Google Patents

Pharmaceutical dosage forms comprising 6'-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine Download PDF

Info

Publication number
US20120034297A1
US20120034297A1 US13/198,182 US201113198182A US2012034297A1 US 20120034297 A1 US20120034297 A1 US 20120034297A1 US 201113198182 A US201113198182 A US 201113198182A US 2012034297 A1 US2012034297 A1 US 2012034297A1
Authority
US
United States
Prior art keywords
dosage form
pharmaceutical dosage
active agent
range
pharmacologically active
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
US13/198,182
Other languages
English (en)
Inventor
Nadja GRUENING
Marc Schiller
Ashish Hemani
Chris Kirby
Ingo Friedrich
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.)
Gruenenthal GmbH
Original Assignee
Gruenenthal GmbH
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=45556332&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20120034297(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Gruenenthal GmbH filed Critical Gruenenthal GmbH
Priority to US13/198,182 priority Critical patent/US20120034297A1/en
Assigned to GRUENENTHAL GMBH reassignment GRUENENTHAL GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KIRBY, CHRIS, HEMANI, ASHISH, FRIEDRICH, INGO, GRUENING, NADJA, SCHILLER, MARC
Publication of US20120034297A1 publication Critical patent/US20120034297A1/en
Assigned to GRUENENTHAL GMBH reassignment GRUENENTHAL GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KIRBY, CHRIS, HEMANI, ASHISH, BOTHMER, JOHN, SCHOLZ, ANDREAS, SCHILLER, MARC, GRUENING, NADJA, FRIEDRICH, INGO
Priority to US14/579,401 priority patent/US9289416B2/en
Priority to US15/245,626 priority patent/US10912763B2/en
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
    • A61K31/33Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • 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
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2095Tabletting processes; Dosage units made by direct compression of powders or specially processed granules, by eliminating solvents, by melt-extrusion, by injection molding, by 3D printing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate

Definitions

  • the invention relates to a pharmaceutical dosage form for preferably oral administration twice daily, once daily or less frequently, which contains a pharmacologically active agent according to general formula (I)
  • R is —H or —CH 3 , or a physiologically acceptable salt thereof.
  • the invention also relates to the use of a pharmaceutical dosage form for preferably oral administration once daily, which contains a pharmacologically active agent according to general formula (I)
  • R is —H or —CH 3 , or a physiologically acceptable salt thereof, in the treatment of neuropathic pain, preferably chronic neuropathic pain.
  • the invention additionally relates to the use of a pharmaceutical dosage form for preferably oral administration once daily, which contains a pharmacologically active agent according to general formula (I)
  • R is —H or —CH 3 , or a physiologically acceptable salt thereof, in the treatment of nociceptive pain, preferably acute or chronic nociceptive pain.
  • the pharmacologically active agents according to general formula (I) can also be referred to as 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-py-rano[3,4,b]indol]-4-amine. Unless expressly stated otherwise, this term also includes the physiologically acceptable salts.
  • the pharmacologically active agents according to the invention are known from the prior art and can be administered orally, perorally, parenterally, intravenously, intraperitoneally, intra-dermally, intramuscularly, intrathecally, epidurally, intranasally, buccally, rectally or locally, for example to the skin, the mucous membranes or into the eyes.
  • the compounds exhibit analgesic properties and are particularly suitable for the treatment of acute, visceral, neuropathic or chronic pain (cf., e.g., WO 2004/043967 and WO 2008/040481).
  • analgesics are typically available as formulations providing immediate release or as formulations providing prolonged release.
  • formulations providing immediate release upon oral administration have the advantage that they lead to a fast release of the analgesic in the gastrointestinal tract.
  • a comparatively high dose of the analgesic is quickly absorbed leading to high plasma levels within a short period of time and resulting in a rapid onset of pain relief, i.e. analgesic action begins shortly after administration. This is particularly desirable in acute pain.
  • formulations providing immediate release of analgesics typically need to be administered frequently, e.g. eight times per day. This is not only detrimental with respect to patient compliance but also may cause comparatively high peak plasma drug concentrations and high fluctuations between peak and trough plasma drug concentrations which in turn may deteriorate tolerability.
  • formulations providing prolonged release upon oral administration have the advantage that they need to be administered less frequently, typically once daily or twice daily. This improves patient compliance and also can reduce peak plasma drug concen-trations and fluctuations between peak and trough plasma drug concentrations which in turn may improve tolerability.
  • analgesic action begins quite a while after first administration.
  • formulations providing prolonged release typically contain higher doses of the analgesics than formulations providing immediate release, they bear a higher risk of being misused.
  • Older patients in particular frequently have difficulties in taking solid pharmaceutical dosage forms.
  • most elderly require adaptations in dosages due to different ADME (absorption, distribution, metablolsm excretion) characteristics in age which is another reason for the need of breakable tablets.
  • ADME absorption, distribution, metablolsm excretion
  • various apparatuses have been developed by means of which solid pharmaceutical dosage forms may be comminuted or pulverized (“tablet crushers”). Such apparatuses are used, for example, by the care staff in old people's homes. The pharmaceutical dosage forms are then administered to the people being cared for not as tablets etc.
  • the comminution of pharmaceutical dosage forms with such apparatuses is problematic if the pharmaceutical dosage forms are prolonged release formulations. As a rule, comminution then results in destruction of the inner structure of the pharmaceutical dosage form, which is responsible for the prolonged release, so doing away with the prolonged-release action. Consequently, after administration, frequently all the physiologically active substance originally contained in the pharmaceutical dosage form is released in a relatively short time, whereby a comparatively very high plasma concentration of the substance is abruptly reached for a relatively short period (dose dumping). In this way, the original prolonged-release formulations become immediate-release formulations.
  • Formulations providing a dual release mode i.e. a combination of immediate release with prolonged release
  • formulations typically rely upon immediate-release units and prolonged-release units that are locally separated from one another and therefore, such pharmaceutical dosage forms can only be prepared by specific and costly methods.
  • the treatment of chronic pain involves long-term analgesic treatment which often requires higher doses than those sufficient in acute pain episodes.
  • titration of the analgesic dose may be required at the start of therapy, especially when common ⁇ -opioid analgesics such as morphine are employed.
  • titrated long-term opioid therapy typically starts with sub-therapeutic doses which are gradually increased until adequate analgesia is reported.
  • the pharmaceutical dosage forms should provide good bioavailability and rapid and adequate pain relief already after the first administration, but also should have a high tolerability, good compliance, and safety.
  • pharmaceutical dosage forms can be prepared which provide immediate release of 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine and provide good bioavailability.
  • 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine has a relatively large pharmacokinetic terminal half life time (t 1/2 ⁇ 60-90 h) and thus, provides pharmacological activity for a comparatively extended period of time after administration (the operational half life is about 24 h).
  • the operational half life is about 24 h.
  • the pharmaceutical dosage form according to the invention combines the advantageous properties of conventional formulations providing immediate release—e.g., rapid pain relief due to adequately high concentration of active ingredient just a short time (e.g., about one hour) after administration of the pharmaceutical composition—with the advantageous properties of conventional formulations providing prolonged release—e.g., long-lasting analgesic action owing to an adequately high level of active ingredient over a prolonged time —, and at the same time even overcomes the drawbacks of said conventional formulations.
  • the patient can effectively combat his pain acutely and, at the same time, treat it effectively over a prolonged period without further measures and merely by regular administration at 12 (or e.g., 24) hourly intervals.
  • the pharmaceutical dosage form according to the invention not only allows the pharmacologically active agent to start flowing rapidly in the plasma when the pharmaceutical dosage form is first administered, leading to a rapid onset of pain relief in the patient owing to the immediate release, but at the same time ensures long-lasting therapeutic efficacy over a relatively long period (at least 12 hours, preferably at least 24 hours). Therefore, the pain suffered by a patient can rapidly be alleviated when the pharmaceutical dosage form according to the invention is administered without the analgesic action quickly fading again.
  • the dose of 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine may generally be kept at a low level, not only initially but throughout the whole analgesic therapy and accordingly, undesired side-effects may be reduced or even fully suppressed. Dose titration during the initial administration phase may be possible but is not required necessarily.
  • the pharmaceutical dosage form according to the invention has good patient compliance and safety. Even if the pharmaceutical dosage form according to the invention is tampered with, e.g. by means of tablet crushers, dose dumping cannot occur—crushing the pharmaceutical dosage form does not further accelerate the immediate release profile. This finding is supported by the pharmacokinetic profiles of three different galenic formulations (solution in macrogol, self-emulsifying capsules filled with liquid, and tablets).
  • the invention relates to a pharmaceutical dosage form containing a pharmacologically active agent according to general formula (I)
  • R is —H or —CH 3
  • said pharmaceutical dosage form being for administration twice daily, once daily or less frequently.
  • the invention also relates to a pharmaceutical dosage form for administration once daily and containing a pharmacologically active agent according to general formula (I)
  • R is —H or —CH 3 , or a physiologically acceptable salt thereof
  • the invention additionally relates to a pharmaceutical dosage form for administration once daily and containing a pharmacologically active agent according to general formula (I)
  • R is —H or —CH 3 , or a physiologically acceptable salt thereof
  • the pharmacologically active agent according to general formula (I) can also be referred to as “6′-fluoro-N-methyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine” when R is —H, and “6′-fluoro-N,N-dimethyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclo-hexane-1,1′-pyrano[3,4,b]indol]-4-amine” when R is —CH 3 ; for the purpose of the specification, the pharmacologically active agent according to general formula (I) can also be referred to as “6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-
  • the pharmacologically active agent according to general formula (I) has a stereochemistry according to general formula (I′)
  • R is —H or —CH 3 , or a physiologically acceptable salt thereof.
  • the compound of formula (I) is selected from
  • the free base according to general formula (I′a) can be systematically referred to as “1,1-(3-methylamino-3-phenylpentamethylene)-6-fluoro-1,3,4,9-tetrahydropyrano[3,4-b]indole (trans)” or as “(1r,40-6′-fluoro-N-methyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine”, respectively.
  • the free base according to general formula (I′b) can be systematically referred to as “1,1-(3-dimethylamino-3-phenylpentamethylene)-6-fluoro-1,3,4,9-tetrahydropyrano[3,4-b]indole (trans)” or as “(1r,4r)-6′-fluoro-N,N-dimethyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine”, respectively.
  • the definition of the pharmacologically active agent according to general formula (I) as used herein includes 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro-[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine, derivatives thereof and stereoisomers thereof in any possible form, thereby particularly including solvates and polymorphs, salts, in particular acid addition salts and corresponding solvates and polymorphs.
  • the pharmacologically active agent according to general formula (I) is present as the single diastereomer according to general formula (I′).
  • the pharmacologically active agent according to general formula (I) is present as mixture of diastereomers.
  • a mixture may contain the diastereomers in any ratio.
  • a diastereomeric mixture could, for example, contain the diastereomers in a ratio of 60 ⁇ 5:40 ⁇ 5, 70 ⁇ 5:30 ⁇ 5, 80 ⁇ 5:20 ⁇ 5 or 90 ⁇ 5:10 ⁇ 5.
  • the pharmaceutical dosage form according to the invention contains the diastereomer according to general formula (I′) in a diastereomeric excess (de) of at least 50% de, more preferably at least 60% de, still more preferably at least 70% de, yet more preferably at least 80% de, even more preferably at least 90% de, most preferably at least 95% de, and in particular at least 98% de, with respect to the other diastereomer (i.e. trans vs. cis and anti vs. syn, respectively).
  • diastereomer according to general formula (I′) in a diastereomeric excess (de) of at least 50% de, more preferably at least 60% de, still more preferably at least 70% de, yet more preferably at least 80% de, even more preferably at least 90% de, most preferably at least 95% de, and in particular at least 98% de, with respect to the other diastereomer (i.e. trans vs. cis and anti vs. syn, respectively).
  • 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyra-no[3,4,b]indol]-4-amine may be present in the pharmaceutical dosage form according to the invention in form of the free base or in form of an acid addition salt, whereby any suitable acid capable of forming such an addition salt may be used.
  • Suitable acids include but are not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, methanesulfonic acid, formic acid, acetic acid, oxalic acid, succinic acid, tartaric acid, mandelic acid, fumaric acid, lactic acid, citric acid, glutamic acid and/or aspartic acid.
  • Salt formation is preferably effected in a solvent, for example, diethyl ether, diisopropyl ether, alkyl acetates, acetone and/or 2-butanone.
  • trimethylchlorosilane in aqueous solution is also suitable for the preparation of hydrochlorides.
  • the pharmacologically active agent according to general formula (I) is preferably contained in the pharmaceutical dosage form in a therapeutically effective amount, i.e., in an amount that is therapeutically effective with regard to a daily administration of the dosage form in the treatment of pain, particularly in the treatment of neuropathic pain, especially chronic neuropathic pain, or in the treatment of nociceptive pain, particularly acute or chronic nociceptive pain.
  • the amount that constitutes a therapeutically effective amount varies according to the compound, the condition being treated, the severity of said condition, the patient being treated, and whether the pharmaceutical dosage form is designed for an immediate or retarded release.
  • the pharmacologically active agent according to general formula (I) is contained in the dosage form in a quantity such that single administration of the dosage form does not lead to any analgesic effect, i.e. the pharmacologically active agent according to general formula (I) is contained in the dosage form in an amount that is sub-therapeutic with regard to a single administration of the dosage form.
  • the pharmacologically active agent according to general formula (I) is contained in the dosage form in an amount that is sub-therapeutic with regard to a single administration of the dosage form.
  • once daily administration of the dosage form leads to an analgesic effect, at the latest, on the fifth day, more preferably at the latest on the fourth day and still more preferably at the latest on the third day of once daily administration.
  • neuropathic pain preferably chronic neuropathic pain
  • once daily administration of the dosage form leads to a sub-therapeutic plasma concentration of the pharmacologically active agent on the first day of administration, but to therapeutic plasma concentrations of the pharmacologically active agent after once daily administration of the dosage form for at least 3, or at least 4, or at least 5 subsequent days.
  • the pharmacologically active agent according to general formula (I) is contained in the dosage form in a quantity such that the dosage form is not effective in the treatment of acute pain, i.e. the pharmacologically active agent is contained in a quantity that is sub-therapeutic with regard to acute pain treatment.
  • the quantity is so low that even after repeated administration for several consecutive days, e.g. 5 days, a significant effect in the treatment of acute pain is not achieved.
  • nociceptive pain preferably acute or chronic nociceptive pain
  • once daily administration of the dosage form leads to a sub-therapeutic plasma concentration of the pharmacologically active agent on the first day of administration, but to therapeutic plasma concentrations of the pharmacologically active agent after once daily administration of the dosage form for at least 3, or at least 4, or at least 5 subsequent days.
  • the pharmacologically active agent according to general formula (I) is contained in the dosage form in a quantity such that initial dose titration is not required.
  • the pharmacologically active agent according to general formula (I) is contained in the dosage form in a quantity such that adverse events that occur during administration of the dosage form are decreased compared to a dosage form comprising a pure ⁇ -opioid receptor agonist, such as morphine in equianalgetic doses.
  • a pure ⁇ -opioid receptor agonist such as morphine in equianalgetic doses.
  • the content of the pharmacologically active agent according to the general formula (I) in the pharmaceutical dosage form according to the invention is at most 95 wt.-%, more preferably at most 50 wt.-%, yet more preferably at most 25 wt.-%, still more preferably at most 10 wt.-%, even more preferably at most 5 wt.-%, most preferably at most 1.0 wt.-%, and in particular at most 0.5 wt-%.
  • the content of the pharmacologically active agent according to the general formula (I) in the pharmaceutical dosage form according to the invention is at least 0.001 wt.-%, more preferably at least 0.005 wt.-%, yet more preferably at least 0.01 wt.-%, still more preferably at least 0.05 wt.-%, even more preferably at least 0.1 wt.-%, most preferably at least 0.5 wt.-%, and in particular at least 1.0 wt.-%.
  • the content of the pharmacologically active agent according to the general formula (I) in the pharmaceutical dosage form according to the invention is within the range of 0.4 ⁇ 0.3 wt.-%, for example, 0.4 ⁇ 0.2 wt.-%, or even 0.4 ⁇ 0.1 wt.-%.
  • the content of the pharmacologically active agent according to the general formula (I) in the pharmaceutical dosage form according to the invention is within the range of 0.5 ⁇ 0.3 wt.-%, for example, 0.5 ⁇ 0.2 wt.-%, or even 0.5 ⁇ 0.1 wt.-%.
  • the content of the pharmacologically active agent according to the general formula (I) in the pharmaceutical dosage form according to the invention is within the range of 0.3 ⁇ 0.3 wt-%, for example, 0.320 ⁇ 0.315 wt.-%, more preferably 0.320 ⁇ 0.310 wt.-%, still more preferably 0.320 ⁇ 0.305 wt.-%, yet more preferably 0.320 ⁇ 0.300 wt.-%, even more preferably 0.320 ⁇ 0.295 wt.-%, most preferably 0.320 ⁇ 0.290 wt.-%, and in particular 0.320 ⁇ 0.285 wt.-%.
  • the content of the pharmacologically active agent according to the general formula (I) in the pharmaceutical dosage form according to the invention is within the range of 0.04 ⁇ 0.035 wt.-%, more preferably 0.04 ⁇ 0.03 wt.-%, still more preferably 0.04 ⁇ 0.025 wt.-%, yet more preferably 0.04 ⁇ 0.02 wt.-%, even more preferably 0.04 ⁇ 0.015 wt.-%, most preferably 0.04 ⁇ 0.01 wt.-%, and in particular 0.04 ⁇ 0.005 wt.-%.
  • the content of the pharmacologically active agent according to the general formula (I) in the pharmaceutical dosage form according to the invention is within the range of 0.6 ⁇ 0.35 wt.-%, more preferably 0.6 ⁇ 0.3 wt.-%, still more preferably 0.6 ⁇ 0.25 wt.-%, yet more preferably 0.6 ⁇ 0.2 wt.-%, even more preferably 0.6 ⁇ 0.15 wt.-%, most preferably 0.6 ⁇ 0.1 wt.-%, and in particular 0.6 ⁇ 0.05 wt.-%.
  • the indication “wt.-%” shall mean weight of the respective ingredient per total weight of the pharmaceutical dosage form.
  • the pharmaceutical dosage form is film coated or encapsulated by an encapsulating medium which does not contain any amount of the pharmacologically active agent according to the general formula (I) and surrounds a core that in turn contains the total amount of the pharmacologically active agent according to the general formula (I)
  • the indication “wt.-%” shall mean weight of the respective ingredient per total weight of the composition forming said core.
  • the pharmacologically active agent according to general formula (I) is preferably homogeneously distributed in the core of the pharmaceutical dosage form.
  • the encapsulating medium or film coating does not contain any pharmacologically active agent according to general formula (I).
  • the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 0.1 ⁇ g to 5000 ⁇ g, more preferably in the range of 0.1 ⁇ g to 1000 ⁇ g, and most preferably in the range of 1.0 ⁇ g to 100 ⁇ g or in the range of 30 ⁇ g to 600 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 25 ⁇ 20 ⁇ g, more preferably 25 ⁇ 15 ⁇ g, still more preferably 25 ⁇ 10 ⁇ g, and most preferably 25 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 40 ⁇ 35 ⁇ g, more preferably 40 ⁇ 30 ⁇ g, still more preferably 40 ⁇ 25 ⁇ g, yet more preferably 40 ⁇ 20 ⁇ g, even more preferably 40 ⁇ 15 ⁇ g, most preferably 40 ⁇ 10 ⁇ g, and in particular 40 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 50 ⁇ 35 ⁇ g, more preferably 50 ⁇ 30 ⁇ g, still more preferably 50 ⁇ 25 ⁇ g, yet more preferably 50 ⁇ 20 ⁇ g, even more preferably 50 ⁇ 15 ⁇ g, most preferably 50 ⁇ 10 ⁇ g, and in particular 50 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 60 ⁇ 35 ⁇ g, more preferably 60 ⁇ 30 ⁇ g, still more preferably 60 ⁇ 25 ⁇ g, yet more preferably 60 ⁇ 20 ⁇ g, even more preferably 60 ⁇ 15 ⁇ g, most preferably 60 ⁇ 10 ⁇ g, and in particular 60 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 100 ⁇ 90 ⁇ g, more preferably 100 ⁇ 80 ⁇ g, still more preferably 100 ⁇ 60 ⁇ g, yet more preferably 100 ⁇ 40 ⁇ g, even more preferably 100 ⁇ 20 ⁇ g, most preferably 100 ⁇ 10 ⁇ g, and in particular 100 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 200 ⁇ 175 ⁇ g, more preferably 200 ⁇ 150 ⁇ g, still more preferably 200 ⁇ 125 ⁇ g, yet more preferably 200 ⁇ 100 ⁇ g, even more preferably 200 ⁇ 75 ⁇ g, most preferably 200 ⁇ 50 ⁇ g, and in particular 200 ⁇ 25 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 400 ⁇ 350 ⁇ g, more preferably 400 ⁇ 300 ⁇ g, still more preferably 400 ⁇ 250 ⁇ g, yet more preferably 400 ⁇ 200 ⁇ g, even more preferably 400 ⁇ 150 ⁇ g, most preferably 400 ⁇ 100 ⁇ g, and in particular 400 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 600 ⁇ 350 ⁇ g, more preferably 600 ⁇ 300 ⁇ g, still more preferably 600 ⁇ 250 ⁇ g, yet more preferably 600 ⁇ 200 ⁇ g, even more preferably 600 ⁇ 150 ⁇ g, most preferably 600 ⁇ 100 ⁇ g, and in particular 600 ⁇ 50 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of acute pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 50 ⁇ g to 3000 ⁇ g, more preferably in the range of 100 ⁇ g to 1000 ⁇ g, even more preferably in the range of 300 ⁇ g to 500 ⁇ g, and most preferably in the range of 350 ⁇ g to 450 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of acute pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 200 ⁇ g to 400 ⁇ g, and in particular in the range of 250 ⁇ g to 350 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of acute pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 200 ⁇ g to 400 ⁇ g, and in particular in the range of 250 ⁇ g to 350 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of acute pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 250 ⁇ g to 450 ⁇ g, and in particular in the range of 300 ⁇ g to 400 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of acute pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 300 ⁇ g to 500 ⁇ g, and in particular in the range of 350 ⁇ g to 450 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of acute pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 350 ⁇ g to 550 ⁇ g, and in particular in the range of 400 ⁇ g to 500 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of acute pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 400 ⁇ g to 600 ⁇ g, and in particular in the range of 450 ⁇ g to 550 ⁇ g.
  • the pharmaceutical dosage form is for use in the treatment of chronic pain, where the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of 0.1 ⁇ g to 500 ⁇ g, more preferably in the range of 1 ⁇ g to 250 ⁇ g, even more preferably in the range of 5 ⁇ g to 100 ⁇ g, and most preferably in the range of 10 ⁇ g to 50 ⁇ g.
  • the pharmaceutical dosage form according to the invention is for oral administration, i.e. the pharmaceutical dosage form is adapted for oral administration.
  • Suitable alternative pathways of administration of the pharmaceutical dosage form according to the invention include but are not limited to vaginal and rectal administration.
  • the pharmaceutical dosage form according to the invention is for administration twice daily, once daily or less frequently, i.e. the pharmaceutical dosage form is adapted for administration twice daily, once daily or less frequently.
  • the pharmaceutical dosage form according to the invention is for administration twice daily.
  • “administration twice daily” preferably means that the pharmaceutical dosage form is adapted for being administered according to a regimen comprising the administration of a first pharmaceutical dosage form according to the invention and the subsequent administration of a second pharmaceutical dosage form according to the invention, wherein both, the first and the second pharmaceutical dosage form are administered during a time interval of about 24 hours, but wherein the second pharmaceutical dosage form is administered not earlier than 6 hours, preferably not earlier than 8 hours, more preferably not earlier than 10 hours and in particular, about 12 hours after the first pharmaceutical dosage form has been administered.
  • the pharmaceutical dosage form according to the invention is for administration once daily.
  • “administration once daily” preferably means that the pharmaceutical dosage form is adapted for being administered according to a regimen comprising the administration of a first pharmaceutical dosage form according to the invention and the subsequent administration of a second pharmaceutical dosage form according to the invention, wherein both, the first and the second pharmaceutical dosage form are administered during a time interval of about 48 hours, but wherein the second pharmaceutical dosage form is administered not earlier than 18 hours, preferably not earlier than 20 hours, more preferably not earlier than 22 hours and in particular, about 24 hours after the first pharmaceutical dosage form has been administered.
  • the pharmaceutical dosage form according to the invention is for administration once daily or less frequently.
  • the pharmaceutical dosage form according to the invention is for administration less frequently than once daily, preferably thrice during four days (3/4), twice during three days (2/3), thrice during five days (3/5), once during two days (1/2), thrice in a week (3/7), twice during five days (2/5), once during three days (1/3), twice in a week (2/7), once during four days (1/4), once during five days (1/5), once during six days (1/6), or once in a week (1/7).
  • administration once during two days (1/2) is particularly preferred.
  • administration regimens “twice daily, once daily, or less frequently” may be realized by administering a single pharmaceutical dosage form containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at a particular point in time or, alternatively, administering a multitude of dose units, i.e. two, three or more dose units, the sum of which multitude of dose units containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at said particular point in time, where the individual dose units are for simultaneous administration or administration within a short period of time, e.g. within 5, 10 or 15 minutes.
  • the dosage form according to the invention is adapted for administration once daily and contains the pharmacologically active agent according to general formula (I) in a dose of from 10 ⁇ g to 190 ⁇ g, i.e. the dosage form according to the invention contains the pharmacologically active agent according to general formula (I) in a daily dose of from 10 ⁇ g to 190 ⁇ g.
  • the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of from 10 ⁇ g to 180 ⁇ g, preferably in the range of from 12.5 ⁇ g to 150 ⁇ g, more preferably in the range of from 15 ⁇ g to 120 ⁇ g, still more preferably in the range of from 17.5 ⁇ g to 100 ⁇ g, yet more preferably in the range of from 20 ⁇ g to 90 ⁇ g, most preferably in the range of from 25 ⁇ g to 80 ⁇ g, and in particular in the range of from 30 ⁇ g to 75 ⁇ g.
  • the dose of the pharmacologically active agent according to general formula (I) is in the range of from 10 ⁇ g to 50 ⁇ g.
  • the dose of the pharmacologically active agent according to general formula (I) is in the range of from 1.0 ⁇ g to 100 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 30 ⁇ 20 ⁇ g, more preferably 30 ⁇ 15 ⁇ g, most preferably 30 ⁇ 10 ⁇ g, and in particular 30 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 35 ⁇ 25 ⁇ g, more preferably 35 ⁇ 20 ⁇ g, still more preferably 35 ⁇ 15 ⁇ g, most preferably 35 ⁇ 10 ⁇ g, and in particular 35 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 40 ⁇ 30 ⁇ g, more preferably 40 ⁇ 25 ⁇ g, still more preferably 40 ⁇ 20 ⁇ g, yet more preferably 40 ⁇ 15 ⁇ g, most preferably 40 ⁇ 10 ⁇ g, and in particular 40 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 45 ⁇ 35 ⁇ g, more preferably 45 ⁇ 30 ⁇ g, still more preferably 45 ⁇ 25 ⁇ g, yet more preferably 45 ⁇ 20 ⁇ g, even more preferably 45 ⁇ 15 ⁇ g, most preferably 45 ⁇ 10 ⁇ g, and in particular 45 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 50 ⁇ 35 ⁇ g, more preferably 50 ⁇ 30 ⁇ g, still more preferably 50 ⁇ 25 ⁇ g, yet more preferably 50 ⁇ 20 ⁇ g, even more preferably 50 ⁇ 15 ⁇ g, most preferably 50 ⁇ 10 ⁇ g, and in particular 50 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 55 ⁇ 35 ⁇ g, more preferably 55 ⁇ 30 ⁇ g, still more preferably 55 ⁇ 25 ⁇ g, yet more preferably 55 ⁇ 20 ⁇ g, even more preferably 55 ⁇ 15 ⁇ g, most preferably 55 ⁇ 10 ⁇ g, and in particular 55 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 60 ⁇ 40 ⁇ g or 60 ⁇ 35 ⁇ g, more preferably 60 ⁇ 30 ⁇ g, still more preferably 60 ⁇ 25 ⁇ g, yet more preferably 60 ⁇ 20 ⁇ g, even more preferably 60 ⁇ 15 ⁇ g, most preferably 60 ⁇ 10 ⁇ g, and in particular 60 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 65 ⁇ 40 ⁇ g or 65 ⁇ 35 ⁇ g, more preferably 65 ⁇ 30 ⁇ g, still more preferably 65 ⁇ 25 ⁇ g, yet more preferably 65 ⁇ 20 ⁇ g, even more preferably 65 ⁇ 15 ⁇ g, most preferably 65 ⁇ 10 ⁇ g, and in particular 65 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 70 ⁇ 40 ⁇ g or 70 ⁇ 35 ⁇ g, more preferably 70 ⁇ 30 ⁇ g, still more preferably 70 ⁇ 25 ⁇ g, yet more preferably 70 ⁇ 20 ⁇ g, even more preferably 70 ⁇ 15 ⁇ g, most preferably 70 ⁇ 10 ⁇ g, and in particular 70 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 75 ⁇ 40 ⁇ g or 75 ⁇ 35 ⁇ g, more preferably 75 ⁇ 30 ⁇ g, still more preferably 75 ⁇ 25 ⁇ g, yet more preferably 75 ⁇ 20 ⁇ g, even more preferably 75 ⁇ 15 ⁇ g, most preferably 75 ⁇ 10 ⁇ g, and in particular 75 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 80 ⁇ 45 ⁇ g or 80 ⁇ 40 ⁇ g, more preferably 80 ⁇ 35 ⁇ g or 80 ⁇ 30 ⁇ g, still more preferably 80 ⁇ 25 ⁇ g, yet more preferably 80 ⁇ 20 ⁇ g, even more preferably 80 ⁇ 15 ⁇ g, most preferably 80 ⁇ 10 ⁇ g, and in particular 80 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 85 ⁇ 45 ⁇ g or 85 ⁇ 40 ⁇ g, more preferably 85 ⁇ 35 ⁇ g or 85 ⁇ 30 ⁇ g, still more preferably 85 ⁇ 25 ⁇ g, yet more preferably 85 ⁇ 20 ⁇ g, even more preferably 85 ⁇ 15 ⁇ g, most preferably 85 ⁇ 10 ⁇ g, and in particular 85 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 90 ⁇ 45 ⁇ g or 90 ⁇ 40 ⁇ g, more preferably 90 ⁇ 35 ⁇ g or 90 ⁇ 30 ⁇ g, still more preferably 90 ⁇ 25 ⁇ g, yet more preferably 90 ⁇ 20 ⁇ g, even more preferably 90 ⁇ 15 ⁇ g, most preferably 90 ⁇ 10 ⁇ g, and in particular 90 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 95 ⁇ 35 ⁇ g, more preferably 95 ⁇ 30 ⁇ g, still more preferably 95 ⁇ 25 ⁇ g, yet more preferably 95 ⁇ 20 ⁇ g, even more preferably 95 ⁇ 15 ⁇ g, most preferably 95 ⁇ 10 ⁇ g, and in particular 95 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 100 ⁇ 80 ⁇ g, more preferably 100 ⁇ 60 ⁇ g, still more preferably 100 ⁇ 40 ⁇ g, even more preferably 100 ⁇ 20 ⁇ g, most preferably 100 ⁇ 10 ⁇ g, and in particular 100 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 110 ⁇ 35 ⁇ g, more preferably 110 ⁇ 30 ⁇ g, still more preferably 110 ⁇ 25 ⁇ g, yet more preferably 110 ⁇ 20 ⁇ g, even more preferably 110 ⁇ 15 ⁇ g, most preferably 110 ⁇ 10 ⁇ g, and in particular 110 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 120 ⁇ 60 ⁇ g, more preferably 120 ⁇ 50 ⁇ g, still more preferably 120 ⁇ 40 ⁇ g, yet more preferably 120 ⁇ 30 ⁇ g, even more preferably 120 ⁇ 20 ⁇ g, most preferably 120 ⁇ 10 ⁇ g, and in particular 120 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 130 ⁇ 60 ⁇ g, more preferably 130 ⁇ 50 ⁇ g, still more preferably 130 ⁇ 40 ⁇ g, yet more preferably 130 ⁇ 30 ⁇ g, even more preferably 130 ⁇ 20 ⁇ g, most preferably 130 ⁇ 10 ⁇ g, and in particular 130 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 140 ⁇ 50 ⁇ g, more preferably 140 ⁇ 50 ⁇ g, still more preferably 140 ⁇ 40 ⁇ g, yet more preferably 140 ⁇ 30 ⁇ g, even more preferably 140 ⁇ 20 ⁇ g, most preferably 140 ⁇ 10 ⁇ g, and in particular 140 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 150 ⁇ 60 ⁇ g, more preferably 150 ⁇ 50 ⁇ g, still more preferably 150 ⁇ 40 ⁇ g, yet more preferably 150 ⁇ 30 ⁇ g, even more preferably 150 ⁇ 20 ⁇ g, most preferably 150 ⁇ 10 ⁇ g, and in particular 150 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 160 ⁇ 30 ⁇ g, more preferably 160 ⁇ 25 ⁇ g, still more preferably 160 ⁇ 20 ⁇ g, yet more preferably 160 ⁇ 15 ⁇ g, most preferably 160 ⁇ 10 ⁇ g, and in particular 160 ⁇ 5 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 170 ⁇ 20 ⁇ g, more preferably 170 ⁇ 10 ⁇ g, and in particular 170 ⁇ 5 ⁇ g.
  • the pharmaceutical dosage form according to the invention is adapted for oral administration.
  • Suitable alternative pathways of administration of the pharmaceutical dosage form according to the invention include but are not limited to vaginal and rectal administration.
  • the pharmaceutical dosage form according to the invention is intended for administration once daily.
  • “administration once daily” preferably means that the pharmaceutical dosage form is adapted for being administered according to a regimen comprising the administration of a first pharmaceutical dosage form according to the invention and the subsequent administration of a second pharmaceutical dosage form according to the invention, wherein both, the first and the second pharmaceutical dosage form are administered during a time interval of about 48 hours, but wherein the second pharmaceutical dosage form is administered not earlier than 18 hours, preferably not earlier than 20 hours, more preferably not earlier than 22 hours and in particular, about 24 hours after the first pharmaceutical dosage form has been administered.
  • administration regimens “once daily” may be realized by administering a single pharmaceutical dosage form containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at a particular point in time or, alternatively, administering a multitude of dose units, i.e. two, three or more dose units, the sum of which multitude of dose units containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at said particular point in time, where the individual dose units are adapted for simultaneous administration or administration within a short period of time, e.g. within 5, 10 or 15 minutes.
  • the dosage form according to the invention is for use in the treatment of neuropathic pain, preferably chronic neuropathic pain such as diabetic neuropathic pain.
  • neuropathic pain preferably chronic neuropathic pain such as diabetic neuropathic pain.
  • the pain is moderate, severe, or moderate to severe.
  • neuropathic pain is pain that originates from nerve damage or nerve malfunction.
  • the neuropathic pain is selected from acute neuropathic pain and chronic neuropathic pain.
  • Neuropathic pain may be caused by damage or disease affecting the central or peripheral portions of the nervous system involved in bodily feelings (the somatosensory system).
  • the dosage form according to the invention is for use in the treatment of chronic neuropathic pain or acute neuropathic pain, peripheral neuropathic pain or central neuropathic pain, mononeuropathic pain or polyneuropathic pain.
  • the neuropathic pain When the neuropathic pain is chronic, it may be chronic peripheral neuropathic pain or chronic central neuropathic pain, in a preferred embodiment chronic peripheral mononeuropathic pain or chronic central mononeuropathic pain, in another preferred embodiment chronic peripheral polyneuropathic pain or chronic central polyneuropathic pain.
  • the neuropathic pain When the neuropathic pain is acute, it may be acute peripheral neuropathic pain or acute central neuropathic pain, in a preferred embodiment acute peripheral mononeuropathic pain or acute central mononeuropathic pain, in another preferred embodiment acute peripheral polyneuropathic pain or acute central polyneuropathic pain.
  • the invention also relates to a pharmacologically active agent according to general formula (I) or a physiologically acceptable salt thereof for use in the treatment of neuropathic pain as described above, preferably by means of administering once daily the pharmaceutical dosage form according to the invention.
  • Fibromyalgia is potentially a central pain disorder and is responsive to medications that are effective for neuropathic pain. Aside from diabetic neuropathy and other metabolic conditions, the common causes of painful peripheral neuropathies are herpes zoster infection, HIV-related neuropathies, nutritional deficiencies, toxins, remote manifestations of malignancies, genetic, and immune mediated disorders or physical trauma to a nerve trunk. Neuropathic pain is common in cancer as a direct result of cancer on peripheral nerves (e.g., compression by a tumor), or as a side effect of chemotherapy, radiation injury or surgery.
  • the pain to be treated is selected from the group consisting of pain being or being associated with panic disorder [episodic paroxysmal anxiety] [F41.0]; dissociative [conversion] disorders [F44]; persistent somatoform pain disorder [F45.4]; pain disorders exclusively related to psychological factors [F45.41]; nonorganic dyspareunia [F52.6]; other enduring personality changes [F62.8]; sadomasochism [F65.5]; elaboration of physical symptoms for psychological reasons [F68.0]; migraine [G43]; other headache syndromes [G44]; trigeminal neuralgia [G50.0]; atypical facial pain [G50.1]; phantom limb syndrome with pain [G54.6]; phantom limb syndrome without pain [G54.7]; acute and chronic pain, not elsewhere classified [G89]; ocular pain [H57.1]; otalgia [H92.0]; angina pectoris, unspecified [120.9
  • the invention also relates to a pharmacologically active agent according to general formula (I) or a physiologically acceptable salt thereof for use in the treatment of pain, preferably neuropathic pain as described above, preferably by means of administering once daily the pharmaceutical dosage form according to the invention.
  • the dosage form according to the invention is adapted for administration once daily and contains the pharmacologically active agent according to general formula (I) in a dose of from 150 ⁇ g to 800 ⁇ g, preferably more than 190 ⁇ g to 800 ⁇ g, i.e. the dosage form according to the invention contains the pharmacologically active agent according to general formula (I) in a daily dose of from 150 ⁇ g to 800 ⁇ g.
  • the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of from 200 ⁇ g to 800 ⁇ g, preferably in the range of from 210 ⁇ g to 750 ⁇ g, more preferably in the range of from 220 ⁇ g to 700 ⁇ g, still more preferably in the range of from 230 ⁇ g to 650 ⁇ g, yet more preferably in the range of from 240 ⁇ g to 600 ⁇ g, and most preferably in the range of from 250 ⁇ g to 550 ⁇ g.
  • the dose of the pharmacologically active agent according to general formula (I) is in the range of from 200 ⁇ g to 600 ⁇ g. In a preferred embodiment, the dose of the pharmacologically active agent according to general formula (I) is in the range of from 300 ⁇ g to 500 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 200 ⁇ 50 ⁇ g, more preferably 200 ⁇ 40 ⁇ g, most preferably 200 ⁇ 30 ⁇ g, and in particular 200 ⁇ 20 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 250 ⁇ 100 ⁇ g, more preferably 250 ⁇ 80 ⁇ g, most preferably 250 ⁇ 60 ⁇ g, and in particular 250 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 300 ⁇ 150 ⁇ g, more preferably 300 ⁇ 125 ⁇ g, most preferably 300 ⁇ 100 ⁇ g, and in particular 300 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 350 ⁇ 200 ⁇ g, more preferably 350 ⁇ 175 ⁇ g, still more preferably 350 ⁇ 150 ⁇ g, most preferably 350 ⁇ 100 ⁇ g, and in particular 350 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 400 ⁇ 250 ⁇ g, more preferably 400 ⁇ 225 ⁇ g, still more preferably 400 ⁇ 200 ⁇ g, yet more preferably 400 ⁇ 150 ⁇ g, most preferably 400 ⁇ 100 ⁇ g, and in particular 400 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 450 ⁇ 300 ⁇ g, more preferably 450 ⁇ 275 ⁇ g, still more preferably 450 ⁇ 250 ⁇ g, yet more preferably 450 ⁇ 200 ⁇ g, even more preferably 450 ⁇ 150 ⁇ g, most preferably 450 ⁇ 100 ⁇ g, and in particular 450 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 500 ⁇ 350 ⁇ g, more preferably 500 ⁇ 300 ⁇ g, still more preferably 500 ⁇ 250 ⁇ g, yet more preferably 500 ⁇ 200 ⁇ g, even more preferably 500 ⁇ 150 ⁇ g, most preferably 500 ⁇ 100 ⁇ g, and in particular 500 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 550 ⁇ 350 ⁇ g, more preferably 550 ⁇ 300 ⁇ g, still more preferably 550 ⁇ 250 ⁇ g, yet more preferably 550 ⁇ 200 ⁇ g, even more preferably 550 ⁇ 150 ⁇ g, most preferably 550 ⁇ 100 ⁇ g, and in particular 550 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 600 ⁇ 400 ⁇ g or 600 ⁇ 350 ⁇ g, more preferably 600 ⁇ 300 ⁇ g, still more preferably 600 ⁇ 250 ⁇ g, yet more preferably 600 ⁇ 200 ⁇ g, even more preferably 600 ⁇ 150 ⁇ g, most preferably 600 ⁇ 100 ⁇ g, and in particular 600 ⁇ 50 ⁇ g.
  • the pharmaceutical dosage form according to the invention is adapted for oral administration.
  • Suitable alternative pathways of administration of the pharmaceutical dosage form according to the invention include but are not limited to vaginal and rectal administration.
  • the pharmaceutical dosage form according to the invention is intended for administration once daily.
  • “administration once daily” preferably means that the pharmaceutical dosage form is adapted for being administered according to a regimen comprising the administration of a first pharmaceutical dosage form according to the invention and the subsequent administration of a second pharmaceutical dosage form according to the invention, wherein both, the first and the second pharmaceutical dosage form are administered during a time interval of about 48 hours, but wherein the second pharmaceutical dosage form is administered not earlier than 18 hours, preferably not earlier than 20 hours, more preferably not earlier than 22 hours and in particular, about 24 hours after the first pharmaceutical dosage form has been administered.
  • administration regimens “once daily” may be realized by administering a single pharmaceutical dosage form containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at a particular point in time or, alternatively, administering a multitude of dose units, i.e. two, three or more dose units, the sum of which multitude of dose units containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at said particular point in time, where the individual dose units are adapted for simultaneous administration or administration within a short period of time, e.g. within 5, 10 or 15 minutes.
  • the dosage form according to the invention is for use in the treatment of nociceptive pain, preferably acute or chronic nociceptive pain.
  • nociceptive pain preferably acute or chronic nociceptive pain.
  • the pain is moderate, severe, or moderate to severe.
  • Nociceptive pain refers to the discomfort that results when a stimulus causes tissue damage to the muscles, bones, skin or internal organs.
  • nociceptive pain is caused by stimulation of peripheral nerve fibers that respond only to stimuli approaching or exceeding harmful intensity (nociceptors), and may be classified according to the mode of noxious stimulation; the most common categories being “thermal” (heat or cold), “mechanical” (crushing, tearing, etc.) and “chemical” (iodine in a cut, chili powder in the eyes).
  • Nociceptive pain may also be divided into “visceral,” “deep somatic” and “superficial somatic” pain.
  • Visceral pain describes a type of nociceptive pain originating in the body's internal organs or their surrounding tissues. This form of pain usually results from the infiltration of harmful cells, as well as the compression or extension of healthy cells. Patients suffering from visceral pain tend to feel generally achy, as this pain tends to not be localized to a specific area. Cancer is a common source of visceral pain.
  • Somatic pain is nociceptive pain that results from some injury to the body. It's generally localized to the affected area and abates when the body repairs the damage to that area. Deep somatic pain is initiated by stimulation of nociceptors in ligaments, tendons, bones, blood vessels, fasciae and muscles, and is dull, aching, poorly-localized pain. Examples include sprains and broken bones. Superficial pain is initiated by activation of nociceptors in the skin or superficial tissues, and is sharp, well-defined and clearly located.
  • nociceptive pain is preferably classified chronic if it has occurred for at least 3 months.
  • the chronic nociceptive pain is selected from chronic visceral pain, chronic deep somatic pain and chronic superficial somatic pain.
  • nociceptive pain includes post-operative pain, cancer pain, low back pain, osteoarthitic pain, and inflammatory pain.
  • the pain to be treated is selected from the group consisting of pain being or being associated with panic disorder [episodic paroxysmal anxiety] [F41.0]; dissociative [conversion] disorders [F44]; persistent somatoform pain disorder [F45.4]; pain disorders exclusively related to psychological factors [F45.41]; nonorganic dyspareunia [F52.6]; other enduring personality changes [F62.8]; sadomasochism [F65.5]; elaboration of physical symptoms for psychological reasons [F68.0]; migraine [G43]; other headache syndromes [G44]; trigeminal neuralgia [G50.0]; atypical facial pain [G50.1]; phantom limb syndrome with pain [G54.6]; phantom limb syndrome without pain [G54.7]; acute and chronic pain, not elsewhere classified [G89]; ocular pain [H57.1]; otalgia [H92.0]; angina pectoris, unspecified [120.9
  • the invention also relates to a pharmacologically active agent according to general formula (I) or a physiologically acceptable salt thereof for use in the treatment of pain, preferably neuropathic pain as described above, preferably by means of administering once daily the pharmaceutical dosage form according to the invention.
  • the pharmaceutical dosage form according to the invention provides immediate release of the pharmacologically active agent according to general formula (I).
  • the pharmaceutical dosage form is specifically designed to provide immediate release of the pharmacologically active agent according to general formula (I) in vitro in accordance with Ph. Eur.
  • the pharmaceutical dosage form is coated, e.g., with a coating that is soluble in gastric juice, the release kinetic is preferably monitored after such coating has been dissolved.
  • the term “immediate release” refers to any release profile that fulfills at least one, preferably both, of the following requirements.
  • the pharmaceutical dosage form disintegrates in 10 minutes or less following exposure to a disintegrating medium. Methods to determine the disintegration time are known to a person skilled in the art. For instance, they can be determined according to the USP XXIV disintegration test procedure, using, for example, an Erweka ZT-71 disintegration tester.
  • the pharmaceutical dosage form releases at least 70 wt.-% of the drug within 15 minutes following exposure to a dissolution medium.
  • the in vitro release properties of the pharmaceutical dosage form according to the invention are determined according to the paddle method with sinker at 50, 75 or 100 rpm, preferably under in vitro conditions at 37 ⁇ 0.5° C. in 900 mL artificial gastric juice at pH 1.2, or under the same conditions in non-artificial gastric juice.
  • the pharmaceutical dosage form releases under in vitro conditions in 900 mL artificial gastric juice at pH 1.2 and 37 ⁇ 0.5° C. after 30 minutes according to the paddle method with sinker at 100 rpm at least 50 wt.-%, more preferably at least 60 wt.-%, still more preferably at least 70 wt.-%, yet more preferably at least 80 wt.-%, most preferably at least 90 wt.-%, and in particular at least 95 wt.-% of the pharmacologically active agent according to general formula (I), based on the total amount of the pharmacologically active agent according to general formula (I) originally contained in the pharmaceutical dosage form.
  • the pharmaceutical dosage form according to the invention exhibits excellent shelf-life and storage stability, i.e. neither the chemical composition, nor the physical characteristics, nor the dissolution profile of the pharmaceutical dosage form are altered significantly upon storage.
  • the pharmaceutical dosage form according to the invention provides sufficient stability to the pharmacologically active agent according to general formula (I) contained therein, so that after storage of the pharmaceutical dosage form at 40 ⁇ 2° C. at 75% RH ⁇ 5% for a minimum time period of 6 weeks, preferably 3 months, the concentrations of undesirable degradants and impurities, respectively, preferably resulting from a degradation or decomposition of the pharmacologically active agent according to general formula (I) as such, is at most 1.0 wt.-%, more preferably at most 0.8 wt.-%, still more preferably at most 0.6 wt.-%, yet more preferably at most 0.4 wt.-%, even more preferably at most 0.2 wt.-%, most preferably at most 0.1 wt.-%, and in particular at most 0.05 wt.-%, relative to the original content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form, i.e. its content before
  • the pharmacologically active agent according to general formula (I) may be decomposed by elimination of the group —NRCH 3 thereby yielding 6′-fluoro-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohex-3-ene-1,1′-pyrano[3,4-b]indole] which appears to be pharmacologically inactive.
  • the pharmaceutical dosage form at 40 ⁇ 2° C. and 75% RH ⁇ 5%, or at 25 ⁇ 2° C.
  • the concentration of 6′-fluoro-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohex-3-ene-1,1′-pyrano[3,4-b]indole] is at most 1.0 wt.-%, more preferably at most 0.8 wt.-%, still more preferably at most 0.6 wt.-%, yet more preferably at most 0.4 wt.-%, even more preferably at most 0.2 wt.-%, most preferably at most 0.1 wt.-%, and in particular at most 0.05 wt.-%, relative to the original content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form, i.e. its content before subjecting the pharmaceutical dosage form to storage.
  • a generally accepted accelerated test for the determination of a drug's stability according to ICH and FDA guidelines relates to the storage of a pharmaceutical formulation containing the drug (e.g., in its container and packaging).
  • a so-called accelerated storage testing should be conducted for pharmaceutical formulations at 40 ⁇ 2° C. at 75% RH ⁇ 5% for a minimum time period of 6 months.
  • a so-called long-term storage testing should be conducted for pharmaceutical formulations at 25 ⁇ 2° C. at not less than 60% RH ⁇ 5% for a minimum time period of 12 months.
  • the long-time storage testing may be shortened to 6 months and the corresponding data doubled to obtain estimated data for the 12-month period.
  • samples of the pharmaceutical formulation are withdrawn at specified time intervals and analyzed in terms of their drug content, presence of impurities, their release profile and if applicable other parameters. According to the ICH guidelines, in all samples the purity of the drug should be ⁇ 98%, the drug content should be 95-105% (FDA guideline: 90-110%). Furthermore, the pharmaceutical formulation should release >80% of the drug within 30 minutes.
  • a content uniformity test should additionally be conducted for 10 randomly chosen dosage forms.
  • the pharmaceutical formulation complies if none individual content is outside the limits of 85% to 115% of the average content. In case that an individual content is outside these limits, another 30 dosage forms have to be analyzed.
  • the preparation fails to comply with the test if more than 3 individual contents are outside the limits of 85 to 115% of the average content or if one or more individual contents are outside the limits of 75% to 125% of the average content.
  • the degradation of the pharmacologically active agent according to general formula (I) does not exceed 2.0%, more preferably 1.5%, still more preferably 1.0%, and most preferably 0.5%.
  • the degradation of the pharmacologically active agent according to general formula (I) does not exceed 4%, more preferably 3%, still more preferably 2%, yet more preferably 1%, and most preferably 0.5%.
  • the pharmaceutical dosage form releases under in vitro conditions in 900 mL artificial gastric juice at pH 1.2 and 37 ⁇ 0.5° C. after 30 minutes according to the paddle method with sinker at 100 rpm at least 50 wt.-%, more preferably at least 60 wt.-%, still more preferably at least 70 wt.-%, and most preferably at least 80 wt.-% of the pharmacologically active agent according to general formula (I), based on the total amount of the pharmacologically active agent according to general formula (I) originally contained in the pharmaceutical dosage form.
  • the pharmaceutical dosage form releases under in vitro conditions in 900 mL artificial gastric juice at pH 1.2 and 37 ⁇ 0.5° C. after 30 minutes according to the paddle method with sinker at 100 rpm at least 50 wt.-%, more preferably at least 60 wt.-%, still more preferably at least 70 wt.-%, and most preferably at least 80 wt.-% of the pharmacologically active agent according to general formula (I), based on the total amount of the pharmacologically active agent according to general formula (I) originally contained in the pharmaceutical dosage form.
  • the absorption properties of a pharmacologically active agent administered by a pharmaceutical dosage form can be described by the pharmacokinetic parameters C max , t max and AUC 0-t .
  • the determination of C max and t max , as well as the calculation of an AUC are well known to a person skilled in the art and described, for example, in Bauer, Frömming, Fuhrer, “Lehrbuch der Pharmazeutica Technologie,” 6th Edition (1999), and in Shargel, Wu-Pong, Yu, “Applied Biopharmaceuticals & Pharmacokinetics”, 5 th Edition (2005). Unless expressly stated otherwise, all pharmacokinetic parameters are expressed as mean values over a population of subjects.
  • C max is the highest plasma concentration of the pharmacologically active agent reached after single administration of the pharmaceutical dosage form.
  • t max is the time needed in order to reach C max .
  • C max, n days is the highest plasma concentration of the pharmacologically active agent reached after once daily administration of the pharmaceutical dosage form for n consecutive days, wherein n can be e.g. 1, 2, 3, 4, 5, 6, etc.
  • C max, ⁇ 5 days is the highest plasma concentration of the pharmacologically active agent reached after once daily administration of the pharmaceutical dosage form for at least 5 consecutive days.
  • steady state conditions are reached after 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 or more days, i.e. continuing administration of the dosage form according to the invention on additional consecutive days does not substantially increase C max any further.
  • t max, n days is the time needed to reach C max, n days (post-dose), wherein n can be e.g. 1, 2, 3, 4, 5, 6, etc.
  • t max, ⁇ 5 days is the time needed to reach C max, ⁇ 5 days (post-dose).
  • AUC 0-t is the area under the curve after single administration to the time t of the last sample that contained an analytically quantifiable concentration of the pharmacologically active agent.
  • AUC 0-72h is the area under the curve baseline after single administration to 72 hours thereafter.
  • the ratio C max /dose is preferably within the range of from 0.01 to 3.00 m ⁇ 3 , yet more preferably within the range of from 0.02 to 2.50 m ⁇ 3 , more preferably within the range of from 0.04 to 2.00 m ⁇ 3 , and most preferably within the range of from 0.06 to 1.69 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.40 ⁇ 0.35 m ⁇ 3 , more preferably 0.40 ⁇ 0.30 m ⁇ 3 , still more preferably 0.40 ⁇ 0.25 m ⁇ 3 , yet more preferably 0.40 ⁇ 0.20 m ⁇ 3 , even more preferably 0.40 ⁇ 0.15 m ⁇ 3 , most preferably 0.40 ⁇ 0.10 m ⁇ 3 , and in particular 0.40 ⁇ 0.05 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.80 ⁇ 0.70 m ⁇ 3 , more preferably 0.80 ⁇ 0.60 m ⁇ 3 , still more preferably 0.80 ⁇ 0.50 m ⁇ 3 , yet more preferably 0.80 ⁇ 0.40 m ⁇ 3 , even more preferably 0.80 ⁇ 0.30 m ⁇ 3 , most preferably 0.80 ⁇ 0.20 m ⁇ 3 , and in particular 0.80 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 1.20 ⁇ 1.05 m ⁇ 3 , more preferably 1.20 ⁇ 0.90 m ⁇ 3 , still more preferably 1.20 ⁇ 0.75 m ⁇ 3 , yet more preferably 1.20 ⁇ 0.60 m ⁇ 3 , even more preferably 1.20 ⁇ 0.45 m ⁇ 3 , most preferably 1.20 ⁇ 0.30 m ⁇ 3 , and in particular 1.20 ⁇ 0.15 m ⁇ 3 .
  • t max is within the range of from 15 minutes to 24 h, still more preferably within the range of from 20 minutes to 20 h, yet more preferably within the range of from 0.5 to 16 h, most preferably within the range of from 1 to 12 h, and in particular within the range of from 2 to 10 h.
  • T max is within the range of 4 ⁇ 3.5 h, more preferably 4 ⁇ 3 h, still more preferably 4 ⁇ 2.5 h, yet more preferably 4 ⁇ 2 h, even more preferably 4 ⁇ 1.5 h, most preferably 4 ⁇ 1 h, and in particular 4 ⁇ 0.5 h.
  • t max is within the range of 8 ⁇ 7 h, more preferably 8 ⁇ 6 h, still more preferably 8 ⁇ 5 h, yet more preferably 8 ⁇ 4 h, even more preferably 8 ⁇ 3 h, most preferably 8 ⁇ 2 h, and in particular 8 ⁇ 1 h.
  • t max is within the range of 12 ⁇ 11 h, more preferably 12 ⁇ 9 h, still more preferably 12 ⁇ 7 h, yet more preferably 12 ⁇ 5 h, even more preferably 12 ⁇ 3 h, most preferably 12 ⁇ 2 h, and in particular 12 ⁇ 1 h.
  • the ratio AUC 0-t /dose is within the range from 0.3 to 20 h/m 3 , more preferably within the range of from 0.4 to 18 h/m 3 , still more preferably within the range of from 0.5 to 16.5 h/m 3 and most preferably within the range of from 0.55 to 12.5 h/m s .
  • the ratio AUC 0-t /dose is within the range of 3 ⁇ 2.5 h/m 3 , more preferably 3 ⁇ 2 h/m 3 , still more preferably 3 ⁇ 1.5 h/m 3 , yet more preferably 3 ⁇ 1 h/m 3 , even more preferably 3 ⁇ 0.75 h/m 3 , most preferably 3 ⁇ 0.5 h/m 3 , and in particular 3 ⁇ 0.25 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 6 ⁇ 5 h/m 3 , more preferably 6 ⁇ 4 h/m 3 , still more preferably 6 ⁇ 3 h/m 3 , yet more preferably 6 ⁇ 2 h/m 3 , even more preferably 6 ⁇ 1.5 h/m 3 , most preferably 6 ⁇ 1 h/m 3 , and in particular 6 ⁇ 0.5 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 9 ⁇ 8 h/m 3 , more preferably 9 ⁇ 7 h/m 3 , still more preferably 9 ⁇ 5 h/m 3 , yet more preferably 9 ⁇ 4 h/m 3 , even more preferably 9 ⁇ 3 h/m 3 , most preferably 9 ⁇ 2 h/m 3 , and in particular 9 ⁇ 1 h/m 3 .
  • C max may advantageously be within the range of from 1 to 250 ⁇ g/m 3 , more preferably within the range of from 5 to 200 ⁇ g/m 3 , still more preferably within the range of from 10 to 150 ⁇ g/m 3 , most preferably within the range of from 15 to 120 ⁇ g/m 3 , and in particular within the range of from 20 to 100 ⁇ g/m 3 .
  • C max is within the range of 20 ⁇ 17.5 ⁇ g/m 3 , more preferably within the range of 20 ⁇ 15 ⁇ g/m 3 , still more preferably within the range of 20 ⁇ 12.5 ⁇ g/m 3 , yet more preferably within the range of 20 ⁇ 10 ⁇ g/m 3 , and most preferably within the range of 20 ⁇ 5 ⁇ g/m 3 .
  • C max is within the range of 25 ⁇ 20 ⁇ g/m 3 , more preferably within the range of 25 ⁇ 17.5 ⁇ g/m 3 , still more preferably within the range of 25 ⁇ 15 ⁇ g/m 3 , yet more preferably within the range of 25 ⁇ 12.5 ⁇ g/m 3 , most preferably within the range of 25 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 25 ⁇ 5 ⁇ g/m 3 .
  • C max is within the range of 30 ⁇ 20 ⁇ g/m 3 , more preferably within the range of 30 ⁇ 17.5 ⁇ g/m 3 , still more preferably within the range of 30 ⁇ 15 ⁇ g/m 3 , yet more preferably within the range of 30 ⁇ 12.5 ⁇ g/m 3 , most preferably within the range of 30 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 30 ⁇ 5 ⁇ g/m 3 .
  • C max is within the range of 35 ⁇ 20 ⁇ g/m 3 , more preferably within the range of 35 ⁇ 17.5 ⁇ g/m 3 , still more preferably within the range of 35 ⁇ 15 ⁇ g/m 3 , yet more preferably within the range of 35 ⁇ 12.5 ⁇ g/m 3 , most preferably within the range of 35 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 35 ⁇ 5 ⁇ g/m 3 .
  • C max is within the range of 40 ⁇ 35 ⁇ g/m 3 , more preferably within the range of 40 ⁇ 30 ⁇ g/m 3 , still more preferably within the range of 40 ⁇ 25 ⁇ g/m 3 , yet more preferably within the range of 40 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 40 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 40 ⁇ 10 ⁇ g/m 3 .
  • C max is within the range of 50 ⁇ 40 ⁇ g/m 3 , more preferably within the range of 50 ⁇ 30 ⁇ g/m 3 , still more preferably within the range of 50 ⁇ 25 ⁇ g/m 3 , yet more preferably within the range of 50 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 50 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 50 ⁇ 10 ⁇ g/m 3 .
  • C max is within the range of 60 ⁇ 40 ⁇ g/m 3 , more preferably within the range of 60 ⁇ 30 ⁇ g/m 3 , still more preferably within the range of 60 ⁇ 25 ⁇ g/m 3 , yet more preferably within the range of 60 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 60 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 60 ⁇ 10 ⁇ g/m 3 .
  • C max is within the range of 70 ⁇ 45 ⁇ g/m 3 , more preferably within the range of 70 ⁇ 40 ⁇ g/m 3 , still more preferably within the range of 70 ⁇ 30 ⁇ g/m 3 , yet more preferably within the range of 70 ⁇ 25 ⁇ g/m 3 , even more preferably within the range of 70 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 70 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 70 ⁇ 10 ⁇ g/m 3 .
  • C max is within the range of 80 ⁇ 50 ⁇ g/m 3 , more preferably within the range of 80 ⁇ 40 ⁇ g/m 3 , still more preferably within the range of 80 ⁇ 30 ⁇ g/m 3 , yet more preferably within the range of 80 ⁇ 25 ⁇ g/m 3 , even more preferably within the range of 80 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 80 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 80 ⁇ 10 ⁇ g/m 3 .
  • C max is within the range of 90 ⁇ 50 ⁇ g/m 3 , more preferably within the range of 90 ⁇ 30 ⁇ g/m 3 , still more preferably within the range of 90 ⁇ 25 ⁇ g/m 3 , yet more preferably within the range of 90 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 90 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 90 ⁇ 10 ⁇ g/m 3 .
  • C max is within the range of 100 ⁇ 50 ⁇ g/m 3 , more preferably within the range of 100 ⁇ 30 ⁇ g/m 3 , still more preferably within the range of 100 ⁇ 25 ⁇ g/m 3 , yet more preferably within the range of 100 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 100 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 100 ⁇ 10 ⁇ g/m 3 .
  • C max is within the range of 120 ⁇ 50 ⁇ g/m 3 , more preferably within the range of 120 ⁇ 30 ⁇ g/m 3 , still more preferably within the range of 120 ⁇ 25 ⁇ g/m 3 , yet more preferably within the range of 120 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 120 ⁇ 15 ⁇ g/m 3 , and in particular within the range of 120 ⁇ 10 ⁇ g/m 3 .
  • the ratio C max /dose is within the range of from 0.01 to 3.00 m ⁇ 3 , yet more preferably within the range of from 0.02 to 2.50 m ⁇ 3 , more preferably within the range of from 0.04 to 2.00 m ⁇ 3 , and most preferably within the range of from 0.06 to 1.69 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.30 ⁇ 0.25 m ⁇ 3 , more preferably 0.30 ⁇ 0.20 m ⁇ 3 , still more preferably 0.30 ⁇ 0.15 m ⁇ 3 , most preferably 0.30 ⁇ 0.10 m ⁇ 3 , and in particular 0.30 ⁇ 0.05 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.40 ⁇ 0.35 m ⁇ 3 , more preferably 0.40 ⁇ 0.30 m ⁇ 3 , still more preferably 0.40 ⁇ 0.25 m ⁇ 3 , yet more preferably 0.40 ⁇ 0.20 m ⁇ 3 , even more preferably 0.40 ⁇ 0.15 m ⁇ 3 , most preferably 0.40 ⁇ 0.10 m ⁇ 3 , and in particular 0.40 ⁇ 0.05 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.50 ⁇ 0.35 m ⁇ 3 , more preferably 0.50 ⁇ 0.30 m ⁇ 3 , still more preferably 0.50 ⁇ 0.25 m ⁇ 3 , yet more preferably 0.50 ⁇ 0.20 m ⁇ 3 , even more preferably 0.50 ⁇ 0.15 m ⁇ 3 , most preferably 0.50 ⁇ 0.10 m ⁇ 3 , and in particular 0.50 ⁇ 0.05 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.60 ⁇ 0.40 m ⁇ 3 , more preferably 0.60 ⁇ 0.30 m ⁇ 3 , still more preferably 0.60 ⁇ 0.25 m ⁇ 3 , yet more preferably 0.60 ⁇ 0.20 m ⁇ 3 , most preferably 0.60 ⁇ 0.15 m ⁇ 3 , and in particular 0.60 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.70 ⁇ 0.40 m ⁇ 3 , more preferably 0.70 ⁇ 0.35 m ⁇ 3 , still more preferably 0.70 ⁇ 0.30 m ⁇ 3 , yet more preferably 0.70 ⁇ 0.25 m ⁇ 3 , even more preferably 0.70 ⁇ 0.20 m ⁇ 3 , most preferably 0.70 ⁇ 0.15 m ⁇ 3 , and in particular 0.70 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.80 ⁇ 0.70 m ⁇ 3 , more preferably 0.80 ⁇ 0.60 m ⁇ 3 , still more preferably 0.80 ⁇ 0.50 m ⁇ 3 , yet more preferably 0.80 ⁇ 0.40 m ⁇ 3 , even more preferably 0.80 ⁇ 0.30 m ⁇ 3 , most preferably 0.80 ⁇ 0.20 m ⁇ 3 , and in particular 0.80 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.90 ⁇ 0.70 m ⁇ 3 , more preferably 0.90 ⁇ 0.60 m ⁇ 3 , still more preferably 0.90 ⁇ 0.50 m ⁇ 3 , yet more preferably 0.90 ⁇ 0.40 m ⁇ 3 , even more preferably 0.90 ⁇ 0.30 m ⁇ 3 , most preferably 0.90 ⁇ 0.20 m ⁇ 3 , and in particular 0.90 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 1.00 ⁇ 0.70 m ⁇ 3 , more preferably 1.00 ⁇ 0.60 m ⁇ 3 , still more preferably 1.00 ⁇ 0.50 m ⁇ 3 , yet more preferably 1.00 ⁇ 0.40 m ⁇ 3 , even more preferably 1.00 ⁇ 0.30 m ⁇ 3 , most preferably 1.00 ⁇ 0.20 m ⁇ 3 , and in particular 1.00 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 1.10 ⁇ 0.70 m ⁇ 3 , more preferably 1.10 ⁇ 0.60 m ⁇ 3 , still more preferably 1.10 ⁇ 0.50 m ⁇ 3 , yet more preferably 1.10 ⁇ 0.40 m ⁇ 3 , even more preferably 1.10 ⁇ 0.30 m ⁇ 3 , most preferably 1.10 ⁇ 0.20 m ⁇ 3 , and in particular 1.10 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 1.20 ⁇ 1.05 m ⁇ 3 , more preferably 1.20 ⁇ 0.90 m ⁇ 3 , still more preferably 1.20 ⁇ 0.75 m ⁇ 3 , yet more preferably 1.20 ⁇ 0.60 m ⁇ 3 , even more preferably 1.20 ⁇ 0.45 m ⁇ 3 , most preferably 1.20 ⁇ 0.30 m ⁇ 3 , and in particular 1.20 ⁇ 0.15 m ⁇ 3 .
  • C max, ⁇ 5 days is within the range of from 1 to 150 ⁇ g/m 3 , more preferably within the range of from 10 to 120 ⁇ g/m 3 , still more preferably within the range of from 15 to 100 ⁇ g/m 3 , yet more preferably within the range of from 20 to 80 ⁇ g/m 3 , most preferably within the range of from 20 to 70 ⁇ g/m 3 , and in particular within the range of from 25 to 60 ⁇ g/m 3
  • C max, ⁇ 5 days is within the range of 25 ⁇ 20 ⁇ g/m 3 , more preferably within the range of 25 ⁇ 15 ⁇ g/m 3 , still more preferably within the range of 25 ⁇ 12.5 ⁇ g/m 3 , yet more preferably within the range of 25 ⁇ 10 ⁇ g/m 3 , most preferably within the range of 25 ⁇ 7.5 ⁇ g/m 3 , and in particular within the range of 25 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 30 ⁇ 20 ⁇ g/m 3 , more preferably within the range of 30 ⁇ 15 ⁇ g/m 3 , still more preferably within the range of 30 ⁇ 12.5 ⁇ g/m 3 , yet more preferably within the range of 30 ⁇ 10 ⁇ g/m 3 , most preferably within the range of 30 ⁇ 7.5 ⁇ g/m 3 , and in particular within the range of 30 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 35 ⁇ 20 ⁇ g/m 3 , more preferably within the range of 35 ⁇ 15 ⁇ g/m 3 , still more preferably within the range of 35 ⁇ 12.5 ⁇ g/m 3 , yet more preferably within the range of 35 ⁇ 10 ⁇ g/m 3 , most preferably within the range of 35 ⁇ 7.5 ⁇ g/m 3 , and in particular within the range of 35 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 40 ⁇ 20 ⁇ g/m 3 , more preferably within the range of 40 ⁇ 15 ⁇ g/m 3 , still more preferably within the range of 40 ⁇ 12.5 ⁇ g/m 3 , yet more preferably within the range of 40 ⁇ 10 ⁇ g/m 3 , most preferably within the range of 40 ⁇ 7.5 ⁇ g/m 3 , and in particular within the range of 40 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 45 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 45 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 45 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 45 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 45 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 45 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 50 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 50 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 50 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 50 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 50 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 50 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 55 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 55 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 55 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 55 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 55 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 55 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 60 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 60 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 60 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 60 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 60 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 60 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 65 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 65 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 65 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 65 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 65 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 65 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 70 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 70 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 70 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 70 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 70 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 70 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 75 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 75 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 75 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 75 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 75 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 75 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 80 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 80 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 80 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 80 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 80 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 80 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 85 ⁇ 50 ⁇ g/m 3 , more preferably within the range of 85 ⁇ 40 ⁇ g/m 3 , still more preferably within the range of 85 ⁇ 30 ⁇ g/m 3 , yet more preferably within the range of 85 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 85 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 85 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 90 ⁇ 50 ⁇ g/m 3 , more preferably within the range of 90 ⁇ 40 ⁇ g/m 3 , still more preferably within the range of 90 ⁇ 30 ⁇ g/m 3 , yet more preferably within the range of 90 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 90 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 90 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 100 ⁇ 50 ⁇ g/m 3 , more preferably within the range of 100 ⁇ 40 ⁇ g/m 3 , still more preferably within the range of 100 ⁇ 30 ⁇ g/m 3 , yet more preferably within the range of 100 ⁇ 20 ⁇ g/m 3 , most preferably within the range of 100 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 100 ⁇ 5 ⁇ g/m 3 .
  • C max, ⁇ 5 days is within the range of 120 ⁇ 30 ⁇ g/m 3 , more preferably within the range of 120 ⁇ 25 ⁇ g/m 3 , still more preferably within the range of 120 ⁇ 20 ⁇ g/m 3 , yet more preferably within the range of 120 ⁇ 15 ⁇ g/m 3 , most preferably within the range of 120 ⁇ 10 ⁇ g/m 3 , and in particular within the range of 120 ⁇ 5 ⁇ g/m 3 .
  • the ratio C max, ⁇ 5 days /dose is within the range of from 0.25 to 1.50 m ⁇ 3 , more preferably within the range of from 0.40 to 1.10 m ⁇ 3 , still more preferably within the range of from 0.45 to 1.00 m ⁇ 3 , yet more preferably within the range of from 0.50 to 0.95 m ⁇ 3 , most preferably within the range of from 0.55 to 0.90 m ⁇ 3 , and in particular within the range of from 0.60 to 0.85 m ⁇ 3 .
  • the ratio C max, ⁇ 5 days /C max, 1 day is ⁇ 1.00, more preferably ⁇ 1.00.
  • the ratio C max, ⁇ 5 days /C max, 1 day is ⁇ 1.00, more preferably ⁇ 1.10, still more preferably ⁇ 1.20, yet more preferably ⁇ 1.30, most preferably ⁇ 1.40, and in particular ⁇ 1.50.
  • the ratio C max, ⁇ 5 days /C max, 1 day is ⁇ 1.60, more preferably ⁇ 1.70, still more preferably ⁇ 1.80, yet more preferably ⁇ 1.90, most preferably ⁇ 2.00, and in particular ⁇ 2.10.
  • the ratio C max, ⁇ 5 days /C max, 1 day is ⁇ 3.10, more preferably ⁇ 3.00, still more preferably ⁇ 2.90, yet more preferably ⁇ 2.80, most preferably ⁇ 2.70, and in particular ⁇ 2.60.
  • the ratio C max, ⁇ 5 days /C max, 1 day is ⁇ 2.50, more preferably ⁇ 2.40, still more preferably ⁇ 2.30, yet more preferably ⁇ 2.20, most preferably ⁇ 2.10, and in particular ⁇ 2.00.
  • C 0-3h is the highest plasma concentration of the pharmacologically active agent reached after a single administration of the pharmaceutical dosage form within the first 3 hours after administration. Accordingly, for the purpose of the specification, C 0-3h, 1 day is the highest plasma concentration of the pharmacologically active agent reached within the first 3 hours after once daily administration of the pharmaceutical dosage form on the very first day of an administration interval, whereas C 0-3h, 5 day is the highest plasma concentration of the pharmacologically active agent reached within the first 3 hours after once daily administration of the pharmaceutical dosage form on the fifths day of said administration interval comprising 5 consecutive days where the dosage form is administered once daily.
  • the pharmaceutical dosage form contains the pharmacologically active agent in a quantity so that C 0-3h, 5d ⁇ C 0-3h, 1d . In another preferred embodiment, the pharmaceutical dosage form contains the pharmacologically active agent in a quantity so that C 0-3h, 5d ⁇ C 0-3h, 1d .
  • the quotient (C 0-3h, 5d )/(C 0-3h, 1d ) is at most 2.5 or 2.4 or 2.3, more preferably at most 2.2 or 2.1 or 2.0, still more preferably at most 1.9 or 1.8 or 1.7, yet more preferably at most 1.6 or 1.5 or 1.4, most preferably at most 1.3 or 1.2 or 1.1, and in particular at most 1.0 or 0.9 or 0.8.
  • the quotient (C 0-3h, 5d ) (C 0-3h, 1d ) is at least 0.8 or 0.9 or 1.0, more preferably at least 1.1 or 1.2 or 1.3, still more preferably at least 1.4 or 1.5 or 1.6, yet more preferably at least 1.7 or 1.8 or 1.9, most preferably at least 2.0 or 2.1 or 2.2, and in particular at least 2.3 or 2.4 or 2.5.
  • the highest plasma concentration of the pharmacologically active agent reached on day 5 of a 5 day long period of once daily administration of the pharmaceutical dosage form is higher than the highest plasma concentrations reached on the first and/or second and/or third and/or fourth day of said period.
  • the daily mean plasma concentration of the pharmacologically active agent is steadily increased during the first 5 days of at least 5 day long period of once daily administration of the pharmaceutical dosage form.
  • the plasma concentration of the pharmacologically active agent measured 10 days after single administration of the pharmaceutical dosage form is still at least 0.5 pg/mL, more preferably at least 1.0 pg/mL, still more preferably 1.25 pg/mL, yet more preferably at least 1.5 pg/mL, most preferably at least 1.75 pg/mL, and in particular at least 2.0 pg/mL.
  • the plasma concentration of the pharmacologically active agent measured 10 drug-free days after once daily administration of the pharmaceutical dosage form for at least 5 consecutive days is still at least 0.5 pg/mL, more preferably at least 1.0 pg/mL, still more preferably 1.25 pg/mL, yet more preferably at least 1.5 pg/mL, most preferably at least 1.75 pg/mL, and in particular at least 2.0 pg/mL.
  • the pharmacokinetic parameter t max is within the range of from 0.5 to 16 h.
  • t max is within the range of from 1 to 12 h, and in particular within the range of from 2 to 10 h.
  • t max is within the range of 4 ⁇ 3.5 h, more preferably 4 ⁇ 3 h, still more preferably 4 ⁇ 2.5 h, yet more preferably 4 ⁇ 2 h, even more preferably 4 ⁇ 1.5 h, most preferably 4 ⁇ 1 h, and in particular 4 ⁇ 0.5 h.
  • t max is within the range of 5 ⁇ 3.5 h, more preferably 5 ⁇ 3 h, still more preferably 5 ⁇ 2.5 h, yet more preferably 5 ⁇ 2 h, even more preferably 5 ⁇ 1.5 h, most preferably 5 ⁇ 1 h, and in particular 5 ⁇ 0.5 h.
  • t max is within the range of 6 ⁇ 4 h, more preferably 6 ⁇ 3 h, still more preferably 6 ⁇ 2.5 h, yet more preferably 6 ⁇ 2 h, even more preferably 6 ⁇ 1.5 h, most preferably 6 ⁇ 1 h, and in particular 6 ⁇ 0.5 h.
  • T max is within the range of 8 ⁇ 7 h, more preferably 8 ⁇ 6 h, still more preferably 8 ⁇ 5 h, yet more preferably 8 ⁇ 4 h, even more preferably 8 ⁇ 3 h, most preferably 8 ⁇ 2 h, and in particular 8 ⁇ 1 h.
  • T max is within the range of 12 ⁇ 3 h, more preferably 12 ⁇ 2 h, and most preferably 12 ⁇ 1 h.
  • t max, ⁇ 5 days is within the range of from 1 to 12 h, more preferably within the range of from 1.5 to 10 h, still more preferably within the range of from 2 to 9 h, yet more preferably within the range of from 2.5 to 8 h, most preferably within the range of from 3 to 7 h, and in particular within the range of from 4 to 6 h.
  • t max, ⁇ 5 days is within the range of 4 ⁇ 3.5 h, more preferably 4 ⁇ 3 h, still more preferably 4 ⁇ 2.5 h, yet more preferably 4 ⁇ 2 h, even more preferably 4 ⁇ 1.5 h, most preferably 4 ⁇ 1 h, and in particular 4 ⁇ 0.5 h.
  • t max, ⁇ 5 days is within the range of 5 ⁇ 3.5 h, more preferably 5 ⁇ 3 h, still more preferably 5 ⁇ 2.5 h, yet more preferably 5 ⁇ 2 h, even more preferably 5 ⁇ 1.5 h, most preferably 5 ⁇ 1 h, and in particular 5 ⁇ 0.5 h.
  • t max, ⁇ 5 days is within the range of 6 ⁇ 4 h, more preferably 6 ⁇ 3 h, still more preferably 6 ⁇ 2.5 h, yet more preferably 6 ⁇ 2 h, even more preferably 6 ⁇ 1.5 h, most preferably 6 ⁇ 1 h, and in particular 6 ⁇ 0.5 h.
  • the ratio AUC 0-t /dose is within the range from 0.3 to 20 h/m 3 , more preferably within the range of from 0.4 to 18 h/m 3 , still more preferably within the range of from 0.5 to 16.5 h/m 3 and most preferably within the range of from 0.55 to 12.5 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 3 ⁇ 2.5 h/m 3 , more preferably 3 ⁇ 2 h/m 3 , still more preferably 3 ⁇ 1.5 h/m 3 , yet more preferably 3 ⁇ 1 h/m 3 , even more preferably 3 ⁇ 0.75 h/m 3 , most preferably 3 ⁇ 0.5 h/m 3 , and in particular 3 ⁇ 0.25 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 6 ⁇ 5 h/m 3 , more preferably 6 ⁇ 4 h/m 3 , still more preferably 6 ⁇ 3 h/m 3 , yet more preferably 6 ⁇ 2 h/m 3 , even more preferably 6 ⁇ 1.5 h/m 3 , most preferably 6 ⁇ 1 h/m 3 , and in particular 6 ⁇ 0.5 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 7.5 ⁇ 7 h/m 3 , more preferably 7.5 ⁇ 6 h/m 3 , still more preferably 7.5 ⁇ 5 h/m 3 , yet more preferably 7.5 ⁇ 4 h/m 3 , even more preferably 7.5 ⁇ 3 h/m 3 , most preferably 7.5 ⁇ 2 h/m 3 , and in particular 7.5 ⁇ 1 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 9 ⁇ 8 h/m 3 , more preferably 9 ⁇ 7 h/m 3 , still more preferably 9 ⁇ 5 h/m 3 , yet more preferably 9 ⁇ 4 h/m 3 , even more preferably 9 ⁇ 3 h/m 3 , most preferably 9 ⁇ 2 h/m 3 , and in particular 9 ⁇ 1 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 10 ⁇ 7 h/m 3 , more preferably 10 ⁇ 6 h/m 3 , still more preferably 10 ⁇ 5 h/m 3 , yet more preferably 10 ⁇ 4 h/m 3 , even more preferably 10 ⁇ 3 h/m 3 , most preferably 10 ⁇ 2 h/m 3 , and in particular 10 ⁇ 1 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range from 0.3 to 20 h/m 3 , more preferably within the range of from 0.4 to 18 h/m 3 , still more preferably within the range of from 0.5 to 16.5 h/m 3 and most preferably within the range of from 0.55 to 12.5 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 3 ⁇ 2.5 h/m 3 , more preferably 3 ⁇ 2 h/m 3 , still more preferably 3 ⁇ 1.5 h/m 3 , yet more preferably 3 ⁇ 1 h/m 3 , even more preferably 3 ⁇ 0.75 h/m 3 , most preferably 3 ⁇ 0.5 h/m 3 , and in particular 3 ⁇ 0.25 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 6 ⁇ 5 h/m 3 , more preferably 6 ⁇ 4 h/m 3 , still more preferably 6 ⁇ 3 h/m 3 , yet more preferably 6 ⁇ 2 h/m 3 , even more preferably 6 ⁇ 1.5 h/m 3 , most preferably 6 ⁇ 1 h/m 3 , and in particular 6 ⁇ 0.5 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 7.5 ⁇ 7 h/m 3 , more preferably 7.5 ⁇ 6 h/m 3 , still more preferably 7.5 ⁇ 5 h/m 3 , yet more preferably 7.5 ⁇ 4 h/m 3 , even more preferably 7.5 ⁇ 3 h/m 3 , most preferably 7.5 ⁇ 2 h/m 3 , and in particular 7.5 ⁇ 1 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 9 ⁇ 8 h/m 3 , more preferably 9 ⁇ 7 h/m 3 , still more preferably 9 ⁇ 5 h/m 3 , yet more preferably 9 ⁇ 4 h/m 3 , even more preferably 9 ⁇ 3 h/m 3 , most preferably 9 ⁇ 2 h/m 3 , and in particular 9 ⁇ 1 h/m 3 .
  • the pharmaceutical dosage form according to the invention is administered once daily during an administration interval comprising an initial phase, during which the plasma concentration time profile substantially changes from day to day, and a steady state phase, during which the plasma concentration time profile does not substantially change from day to day.
  • the plasma concentration time profile may still change during a day, i.e. the plasma concentration measured e.g. 1 hour after administration may substantially differ from the plasma concentration measured e.g. 2, 3, 4, 6, 12 or 20 hours after the same administration on the same day.
  • the plasma concentration measured X hours after administration on day N does not substantially differ from the plasma concentration measured X hours after the following administration on the following day N+1.
  • the initial phase lasts 1, 2, 3, 4 or 5 consecutive days until the steady state phase commences.
  • the pharmaceutical dosage form provides and maintains upon administration once daily pharmacologically effective plasma concentrations of the pharmacologically active agent according to general formula (I) for at least 12 h, preferably at least 18 h, more preferably at least 20 h, yet more preferably at least 22 h, and in particular all 24 h of at least 1.0 pg/mL, at least 2.0 pg/mL, or at least 3.0 pg/mL, more preferably at least 4.0 pg/mL, at least 5.0 pg/mL, or at least 6.0 pg/mL, still more preferably at least 7.0 pg/mL, at least 8.0 pg/mL, or at least 9.0 pg/mL, yet more preferably at least 10 pg/mL, at least 12.5 pg/mL, or at least 15 p
  • the dosage form according to the invention is adapted for administration once daily and contains the pharmacologically active agent according to general formula (I) in a dose of from 150 ⁇ g to 800 ⁇ g, preferably more than 190 ⁇ g to 800 ⁇ g, i.e. the dosage form according to the invention contains the pharmacologically active agent according to general formula (I) in a daily dose of from 150 ⁇ g to 800 ⁇ g.
  • the dose of the pharmacologically active agent according to general formula (I) preferably is in the range of from 200 ⁇ g to 800 ⁇ g, preferably in the range of from 210 ⁇ g to 750 ⁇ g, more preferably in the range of from 220 ⁇ g to 700 ⁇ g, still more preferably in the range of from 230 ⁇ g to 650 ⁇ g, yet more preferably in the range of from 240 ⁇ g to 600 ⁇ g, and most preferably in the range of from 250 ⁇ g to 550 ⁇ g.
  • the dose of the pharmacologically active agent according to general formula (I) is in the range of from 200 ⁇ g to 600 ⁇ g. In a preferred embodiment, the dose of the pharmacologically active agent according to general formula (I) is in the range of from 300 ⁇ g to 500 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 200 ⁇ 50 ⁇ g, more preferably 200 ⁇ 40 ⁇ g, most preferably 200 ⁇ 30 ⁇ g, and in particular 200 ⁇ 20 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 250 ⁇ 100 ⁇ g, more preferably 250 ⁇ 80 ⁇ g, most preferably 250 ⁇ 60 ⁇ g, and in particular 250 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 300 ⁇ 150 ⁇ g, more preferably 300 ⁇ 125 ⁇ g, most preferably 300 ⁇ 100 ⁇ g, and in particular 300 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 350 ⁇ 200 ⁇ g, more preferably 350 ⁇ 175 ⁇ g, still more preferably 350 ⁇ 150 ⁇ g, most preferably 350 ⁇ 100 ⁇ g, and in particular 350 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 400 ⁇ 250 ⁇ g, more preferably 400 ⁇ 225 ⁇ g, still more preferably 400 ⁇ 200 ⁇ g, yet more preferably 400 ⁇ 150 ⁇ g, most preferably 400 ⁇ 100 ⁇ g, and in particular 400 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 450 ⁇ 300 ⁇ g, more preferably 450 ⁇ 275 ⁇ g, still more preferably 450 ⁇ 250 ⁇ g, yet more preferably 450 ⁇ 200 ⁇ g, even more preferably 450 ⁇ 150 ⁇ g, most preferably 450 ⁇ 100 ⁇ g, and in particular 450 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 500 ⁇ 350 ⁇ g, more preferably 500 ⁇ 300 ⁇ g, still more preferably 500 ⁇ 250 ⁇ g, yet more preferably 500 ⁇ 200 ⁇ g, even more preferably 500 ⁇ 150 ⁇ g, most preferably 500 ⁇ 100 ⁇ g, and in particular 500 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 550 ⁇ 350 ⁇ g, more preferably 550 ⁇ 300 ⁇ g, still more preferably 550 ⁇ 250 ⁇ g, yet more preferably 550 ⁇ 200 ⁇ g, even more preferably 550 ⁇ 150 ⁇ g, most preferably 550 ⁇ 100 ⁇ g, and in particular 550 ⁇ 50 ⁇ g.
  • the content of the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form is within the range of 600 ⁇ 400 ⁇ g or 600 ⁇ 350 ⁇ g, more preferably 600 ⁇ 300 ⁇ g, still more preferably 600 ⁇ 250 ⁇ g, yet more preferably 600 ⁇ 200 ⁇ g, even more preferably 600 ⁇ 150 ⁇ g, most preferably 600 ⁇ 100 ⁇ g, and in particular 600 ⁇ 50 ⁇ g.
  • the pharmaceutical dosage form according to the invention is adapted for oral administration.
  • Suitable alternative pathways of administration of the pharmaceutical dosage form according to the invention include but are not limited to vaginal and rectal administration.
  • the pharmaceutical dosage form according to the invention is intended for administration once daily.
  • “administration once daily” preferably means that the pharmaceutical dosage form is adapted for being administered according to a regimen comprising the administration of a first pharmaceutical dosage form according to the invention and the subsequent administration of a second pharmaceutical dosage form according to the invention, wherein both, the first and the second pharmaceutical dosage form are administered during a time interval of about 48 hours, but wherein the second pharmaceutical dosage form is administered not earlier than 18 hours, preferably not earlier than 20 hours, more preferably not earlier than 22 hours and in particular, about 24 hours after the first pharmaceutical dosage form has been administered.
  • administration regimens “once daily” may be realized by administering a single pharmaceutical dosage form containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at a particular point in time or, alternatively, administering a multitude of dose units, i.e. two, three or more dose units, the sum of which multitude of dose units containing the full amount of the pharmacologically active agent according to general formula (I) to be administered at said particular point in time, where the individual dose units are adapted for simultaneous administration or administration within a short period of time, e.g. within 5, 10 or 15 minutes.
  • the dosage form according to the invention is for use in the treatment of nociceptive pain, preferably acute or chronic nociceptive pain.
  • nociceptive pain preferably acute or chronic nociceptive pain.
  • the pain is moderate, severe, or moderate to severe.
  • Nociceptive pain refers to the discomfort that results when a stimulus causes tissue damage to the muscles, bones, skin or internal organs.
  • nociceptive pain is caused by stimulation of peripheral nerve fibers that respond only to stimuli approaching or exceeding harmful intensity (nociceptors), and may be classified according to the mode of noxious stimulation; the most common categories being “thermal” (heat or cold), “mechanical” (crushing, tearing, etc.) and “chemical” (iodine in a cut, chili powder in the eyes).
  • Nociceptive pain may also be divided into “visceral,” “deep somatic” and “superficial somatic” pain.
  • Visceral pain describes a type of nociceptive pain originating in the body's internal organs or their surrounding tissues. This form of pain usually results from the infiltration of harmful cells, as well as the compression or extension of healthy cells. Patients suffering from visceral pain tend to feel generally achy, as this pain tends to not be localized to a specific area. Cancer is a common source of visceral pain.
  • Somatic pain is nociceptive pain that results from some injury to the body. It's generally localized to the affected area and abates when the body repairs the damage to that area. Deep somatic pain is initiated by stimulation of nociceptors in ligaments, tendons, bones, blood vessels, fasciae and muscles, and is dull, aching, poorly-localized pain. Examples include sprains and broken bones. Superficial pain is initiated by activation of nociceptors in the skin or superficial tissues, and is sharp, well-defined and clearly located.
  • nociceptive pain is preferably classified chronic if it has occurred for at least 3 months.
  • the chronic nociceptive pain is selected from chronic visceral pain, chronic deep somatic pain and chronic superficial somatic pain.
  • nociceptive pain includes broken or fractured bones, bruises, burns, cuts, inflammation (from infection or arthritis), and sprains.
  • nociceptive pain includes post-operative pain, cancer pain, low back pain, and inflammatory pain.
  • the pain to be treated is selected from the group consisting of pain being or being associated with panic disorder [episodic paroxysmal anxiety] [F41.0]; dissociative [conversion] disorders [F44]; persistent somatoform pain disorder [F45.4]; pain disorders exclusively related to psychological factors [F45.41]; nonorganic dyspareunia [F52.6]; other enduring personality changes [F62.8]; sadomasochism [F65.5]; elaboration of physical symptoms for psychological reasons [F68.0]; migraine [G43]; other headache syndromes [G44]; trigeminal neuralgia [G50.0]; atypical facial pain [G50.1]; phantom limb syndrome with pain [G54.6]; phantom limb syndrome without pain [G54.7]; acute and chronic pain, not elsewhere classified [G89]; ocular pain [H57.1]; otalgia [H92.0]; angina pectoris, unspecified [120.9
  • the invention also relates to a pharmacologically active agent according to general formula (I) or a physiologically acceptable salt thereof for use in the treatment of pain, preferably neuropathic pain as described above, preferably by means of administering once daily the pharmaceutical dosage form according to the invention.
  • C max is preferably within the range of from 1 to 250 ⁇ g/m 3 , more preferably within the range of from 10 to 220 ⁇ g/m 3 , still more preferably within the range of from 40 to 200 ⁇ g/m 3 , most preferably within the range of from 60 to 170 ⁇ g/m 3 , and in particular within the range of from 80 to 150 ⁇ g/m 3 .
  • C max is within the range of 90 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 90 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 90 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 90 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 90 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 90 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 90 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 100 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 100 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 100 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 100 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 100 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 100 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 100 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 110 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 110 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 110 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 110 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 110 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 110 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 110 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 120 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 120 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 120 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 120 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 120 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 120 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 120 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 130 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 130 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 130 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 130 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 130 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 130 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 130 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 140 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 140 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 140 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 140 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 140 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 140 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 140 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 150 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 150 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 150 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 150 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 150 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 150 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 150 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 160 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 160 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 160 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 160 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 160 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 160 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 160 ⁇ 20 ⁇ g/m 3 .
  • C max is within the range of 170 ⁇ 80 ⁇ g/m 3 , more preferably within the range of 170 ⁇ 70 ⁇ g/m 3 , still more preferably within the range of 170 ⁇ 60 ⁇ g/m 3 , yet more preferably within the range of 170 ⁇ 50 ⁇ g/m 3 , even more preferably within the range of 170 ⁇ 40 ⁇ g/m 3 , most preferably within the range of 170 ⁇ 30 ⁇ g/m 3 , and in particular within the range of 170 ⁇ 20 ⁇ g/m 3 .
  • the ratio C max /dose is within the range of from 0.01 to 3.00 m ⁇ 3 , yet more preferably within the range of from 0.02 to 2.50 m ⁇ 3 , more preferably within the range of from 0.04 to 2.00 m ⁇ 3 , and most preferably within the range of from 0.06 to 1.69 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.25 ⁇ 0.20 m ⁇ 3 , more preferably 0.25 ⁇ 0.15 m ⁇ 3 , still more preferably 0.25 ⁇ 0.10 m ⁇ 3 , and most preferably 0.25 ⁇ 0.15 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.30 ⁇ 0.25 m ⁇ 3 , more preferably 0.30 ⁇ 0.20 m ⁇ 3 , still more preferably 0.30 ⁇ 0.15 m ⁇ 3 , most preferably 0.30 ⁇ 0.10 m ⁇ 3 , and in particular 0.30 ⁇ 0.05 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.40 ⁇ 0.35 m ⁇ 3 , more preferably 0.40 ⁇ 0.30 m ⁇ 3 , still more preferably 0.40 ⁇ 0.25 m ⁇ 3 , yet more preferably 0.40 ⁇ 0.20 m ⁇ 3 , even more preferably 0.40 ⁇ 0.15 m ⁇ 3 , most preferably 0.40 ⁇ 0.10 m ⁇ 3 , and in particular 0.40 ⁇ 0.05 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.50 ⁇ 0.35 m ⁇ 3 , more preferably 0.50 ⁇ 0.30 m ⁇ 3 , still more preferably 0.50 ⁇ 0.25 m ⁇ 3 , yet more preferably 0.50 ⁇ 0.20 m ⁇ 3 , even more preferably 0.50 ⁇ 0.15 m ⁇ 3 , most preferably 0.50 ⁇ 0.10 m ⁇ 3 , and in particular 0.50 ⁇ 0.05 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.60 ⁇ 0.40 m ⁇ 3 , more preferably 0.60 ⁇ 0.30 m ⁇ 3 , still more preferably 0.60 ⁇ 0.25 m ⁇ 3 , yet more preferably 0.60 ⁇ 0.20 m ⁇ 3 , most preferably 0.60 ⁇ 0.15 m ⁇ 3 , and in particular 0.60 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.70 ⁇ 0.40 m ⁇ 3 , more preferably 0.70 ⁇ 0.35 m ⁇ 3 , still more preferably 0.70 ⁇ 0.30 m ⁇ 3 , yet more preferably 0.70 ⁇ 0.25 m ⁇ 3 , even more preferably 0.70 ⁇ 0.20 m ⁇ 3 , most preferably 0.70 ⁇ 0.15 m ⁇ 3 , and in particular 0.70 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.80 ⁇ 0.70 m ⁇ 3 , more preferably 0.80 ⁇ 0.60 m ⁇ 3 , still more preferably 0.80 ⁇ 0.50 m ⁇ 3 , yet more preferably 0.80 ⁇ 0.40 m ⁇ 3 , even more preferably 0.80 ⁇ 0.30 m ⁇ 3 , most preferably 0.80 ⁇ 0.20 m ⁇ 3 , and in particular 0.80 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 0.90 ⁇ 0.70 m ⁇ 3 , more preferably 0.90 ⁇ 0.60 m ⁇ 3 , still more preferably 0.90 ⁇ 0.50 m ⁇ 3 , yet more preferably 0.90 ⁇ 0.40 m ⁇ 3 , even more preferably 0.90 ⁇ 0.30 m ⁇ 3 , most preferably 0.90 ⁇ 0.20 m ⁇ 3 , and in particular 0.90 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 1.00 ⁇ 0.70 m ⁇ 3 , more preferably 1.00 ⁇ 0.60 m ⁇ 3 , still more preferably 1.00 ⁇ 0.50 m ⁇ 3 , yet more preferably 1.00 ⁇ 0.40 m ⁇ 3 , even more preferably 1.00 ⁇ 0.30 m ⁇ 3 , most preferably 1.00 ⁇ 0.20 m ⁇ 3 , and in particular 1.00 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 1.10 ⁇ 0.70 m ⁇ 3 , more preferably 1.10 ⁇ 0.60 m ⁇ 3 , still more preferably 1.10 ⁇ 0.50 m ⁇ 3 , yet more preferably 1.10 ⁇ 0.40 m ⁇ 3 , even more preferably 1.10 ⁇ 0.30 m ⁇ 3 , most preferably 1.10 ⁇ 0.20 m ⁇ 3 , and in particular 1.10 ⁇ 0.10 m ⁇ 3 .
  • the ratio C max /dose is within the range of 1.20 ⁇ 1.05 m ⁇ 3 , more preferably 1.20 ⁇ 0.90 m ⁇ 3 , still more preferably 1.20 ⁇ 0.75 m ⁇ 3 , yet more preferably 1.20 ⁇ 0.60 m ⁇ 3 , even more preferably 1.20 ⁇ 0.45 m ⁇ 3 , most preferably 1.20 ⁇ 0.30 m ⁇ 3 , and in particular 1.20 ⁇ 0.15 m ⁇ 3 .
  • the highest plasma concentration of the pharmacologically active agent reached on day 5 of a 5 day long period of once daily administration of the pharmaceutical dosage form is higher than the highest plasma concentrations reached on the first and/or second and/or third and/or fourth day of said period.
  • the daily mean plasma concentration of the pharmacologically active agent is steadily increased during the first 5 days of at least 5 day long period of once daily administration of the pharmaceutical dosage form.
  • the plasma concentration of the pharmacologically active agent measured 10 days after single administration of the pharmaceutical dosage form is still at least 0.5 ⁇ g/mL, more preferably at least 1.0 pg/mL, still more preferably 1.25 pg/mL, yet more preferably at least 1.5 pg/mL, most preferably at least 1.75 pg/mL, and in particular at least 2.0 pg/mL.
  • the plasma concentration of the pharmacologically active agent measured 10 drug-free days after once daily administration of the pharmaceutical dosage form for at least 5 consecutive days is still at least 0.5 pg/mL, more preferably at least 1.0 pg/mL, still more preferably 1.25 pg/mL, yet more preferably at least 1.5 pg/mL, most preferably at least 1.75 pg/mL, and in particular at least 2.0 pg/mL.
  • the pharmacokinetic parameter t max is within the range of from 0.5 to 16 h.
  • t max is within the range of from 1 to 12 h, and in particular within the range of from 2 to 10 h.
  • T max is within the range of 4 ⁇ 3.5 h, more preferably 4 ⁇ 3 h, still more preferably 4 ⁇ 2.5 h, yet more preferably 4 ⁇ 2 h, even more preferably 4 ⁇ 1.5 h, most preferably 4 ⁇ 1 h, and in particular 4 ⁇ 0.5 h.
  • T max is within the range of 5 ⁇ 3.5 h, more preferably 5 ⁇ 3 h, still more preferably 5 ⁇ 2.5 h, yet more preferably 5 ⁇ 2 h, even more preferably 5 ⁇ 1.5 h, most preferably 5 ⁇ 1 h, and in particular 5 ⁇ 0.5 h.
  • t max is within the range of 6 ⁇ 4 h, more preferably 6 ⁇ 3 h, still more preferably 6 ⁇ 2.5 h, yet more preferably 6 ⁇ 2 h, even more preferably 6 ⁇ 1.5 h, most preferably 6 ⁇ 1 h, and in particular 6 ⁇ 0.5 h.
  • t max is within the range of 7 ⁇ 6 h, more preferably 7 ⁇ 5 h, still more preferably 7 ⁇ 4 h, yet more preferably 7 ⁇ 3 h, even more preferably 7 ⁇ 2 h, most preferably 7 ⁇ 1 h, and in particular 7 ⁇ 0.5 h.
  • T max is within the range of 8 ⁇ 7 h, more preferably 8 ⁇ 6 h, still more preferably 8 ⁇ 5 h, yet more preferably 8 ⁇ 4 h, even more preferably 8 ⁇ 3 h, most preferably 8 ⁇ 2 h, and in particular 8 ⁇ 1 h.
  • t max is within the range of 12 ⁇ 3 h, more preferably 12 ⁇ 2 h, and most preferably 12 ⁇ 1 h.
  • the ratio AUC 0-t /dose is within the range from 0.3 to 20 h/m 3 , more preferably within the range of from 0.4 to 18 h/m 3 , still more preferably within the range of from 0.5 to 16.5 h/m 3 and most preferably within the range of from 0.55 to 12.5 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 3 ⁇ 2.5 h/m 3 , more preferably 3 ⁇ 2 h/m 3 , still more preferably 3 ⁇ 1.5 h/m 3 , yet more preferably 3 ⁇ 1 h/m 3 , even more preferably 3 ⁇ 0.75 h/m 3 , most preferably 3 ⁇ 0.5 h/m 3 , and in particular 3 ⁇ 0.25 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 6 ⁇ 5 h/m 3 , more preferably 6 ⁇ 4 h/m 3 , still more preferably 6 ⁇ 3 h/m 3 , yet more preferably 6 ⁇ 2 h/m 3 , even more preferably 6 ⁇ 1.5 h/m 3 , most preferably 6 ⁇ 1 h/m 3 , and in particular 6 ⁇ 0.5 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 7.5 ⁇ 7 h/m 3 , more preferably 7.5 ⁇ 6 h/m 3 , still more preferably 7.5 ⁇ 5 h/m 3 , yet more preferably 7.5 ⁇ 4 h/m 3 , even more preferably 7.5 ⁇ 3 h/m 3 , most preferably 7.5 ⁇ 2 h/m 3 , and in particular 7.5 ⁇ 1 h/m 3 .
  • the ratio AUC 0-t /dose is within the range of 9 ⁇ 8 h/m 3 , more preferably 9 ⁇ 7 h/m 3 , still more preferably 9 ⁇ 5 h/m 3 , yet more preferably 9 ⁇ 4 h/m 3 , even more preferably 9 ⁇ 3 h/m 3 , most preferably 9 ⁇ 2 h/m 3 , and in particular 9 ⁇ 1 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 10 ⁇ 7 h/m 3 , more preferably 10 ⁇ 6 h/m 3 , still more preferably 10 ⁇ 5 h/m 3 , yet more preferably 10 ⁇ 4 h/m 3 , even more preferably 10 ⁇ 3 h/m 3 , most preferably 10 ⁇ 2 h/m 3 , and in particular 10 ⁇ 1 h/m 3 .
  • AUC 0-t is within the range of 3750 ⁇ 3500 h ⁇ pg/mL, more preferably 3750 ⁇ 3000 h ⁇ pg/mL, still more preferably 3750 ⁇ 2500 h ⁇ pg/mL, yet more preferably 3750 ⁇ 2000 h ⁇ pg/mL, even more preferably 3750 ⁇ 1500 h ⁇ pg/mL, most preferably 3750 ⁇ 1000 h ⁇ pg/mL, and in particular 3750 ⁇ 500 h ⁇ pg/mL.
  • the ratio AUC 0-72h /dose is within the range from 0.3 to 20 h/m 3 , more preferably within the range of from 0.4 to 18 h/m 3 , still more preferably within the range of from 0.5 to 16.5 h/m 3 and most preferably within the range of from 0.55 to 12.5 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 3 ⁇ 2.5 h/m 3 , more preferably 3 ⁇ 2 h/m 3 , still more preferably 3 ⁇ 1.5 h/m 3 , yet more preferably 3 ⁇ 1 h/m 3 , even more preferably 3 ⁇ 0.75 h/m 3 , most preferably 3 ⁇ 0.5 h/m 3 , and in particular 3 ⁇ 0.25 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 6 ⁇ 5 h/m 3 , more preferably 6 ⁇ 4 h/m 3 , still more preferably 6 ⁇ 3 h/m 3 , yet more preferably 6 ⁇ 2 h/m 3 , even more preferably 6 ⁇ 1.5 h/m 3 , most preferably 6 ⁇ 1 h/m 3 , and in particular 6 ⁇ 0.5 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 7.5 ⁇ 7 h/m 3 , more preferably 7.5 ⁇ 6 h/m 3 , still more preferably 7.5 ⁇ 5 h/m 3 , yet more preferably 7.5 ⁇ 4 h/m 3 , even more preferably 7.5 ⁇ 3 h/m 3 , most preferably 7.5 ⁇ 2 h/m 3 , and in particular 7.5 ⁇ 1 h/m 3 .
  • the ratio AUC 0-72h /dose is within the range of 9 ⁇ 8 h/m 3 , more preferably 9 ⁇ 7 h/m 3 , still more preferably 9 ⁇ 5 h/m 3 , yet more preferably 9 ⁇ 4 h/m 3 , even more preferably 9 ⁇ 3 h/m 3 , most preferably 9 ⁇ 2 h/m 3 , and in particular 9 ⁇ 1 h/m 3 .
  • AUC 0-72 is within the range of 2800 ⁇ 2500 h ⁇ pg/mL, more preferably 2800 ⁇ 2250 h ⁇ pg/mL, still more preferably 2800 ⁇ 2000 h ⁇ pg/mL, yet more preferably 2800 ⁇ 1750 h ⁇ pg/mL, even more preferably 2800 ⁇ 1500 h ⁇ pg/mL, most preferably 2800 ⁇ 1000 h ⁇ pg/mL, and in particular 2800 ⁇ 500 h ⁇ pg/mL.
  • the pharmaceutical dosage form according to the invention is administered once daily during an administration interval comprising an initial phase, during which the plasma concentration time profile substantially changes from day to day, and a steady state phase, during which the plasma concentration time profile does not substantially change from day to day.
  • the plasma concentration time profile may still change during a day, i.e. the plasma concentration measured e.g. 1 hour after administration may substantially differ from the plasma concentration measured e.g. 2, 3, 4, 6, 12 or 20 hours after the same administration on the same day.
  • the plasma concentration measured X hours after administration on day N does not substantially differ from the plasma concentration measured X hours after the following administration on the following day N+1.
  • the initial phase lasts 1, 2, 3, 4 or 5 consecutive days until the steady state phase commences.
  • the pharmaceutical dosage form provides and maintains upon administration once daily pharmacologically effective plasma concentrations of the pharmacologically active agent according to general formula (I) for at least 12 h, preferably at least 18 h, more preferably at least 20 h, yet more preferably at least 22 h and in particular all 24 h of at least 25 pg/mL, at least 30 pg/mL, or at least 35 pg/mL, more preferably at least 40 pg/mL, at least 45 pg/mL, or at least 50 pg/mL, still more preferably at least 60 pg/mL, at least 70 pg/mL, or at least 80 pg/mL, yet more preferably at least 90 pg/mL, at least 100 pg/mL, or at least 110 pg/mL, even more preferably at
  • the ratio of the maximum plasma concentration C max to the plasma concentration measured 3 h after administration C 3h is not more than 3.9, not more than 3.8, or not more than not more than 3.7, preferably not more than 3.6, not more than 3.5, or not more than not more than 3.4, more preferably not more than 3.3, not more than 3.2, or not more than not more than 3.1, still more preferably not more than 3.0, not more than 2.9, or not more than not more than 2.8, yet more preferably not more than 2.7, not more than 2.6, or not more than not more than 2.5, even more preferably not more than 2.4, not more than 2.3, or not more than not more than 2.2, most preferably not more than 2.1, not more than 2.0, or not more than not more than 1.9, and in particular preferably not more than 1.8, not more than 1.7, or not more than not more than 1.6.
  • the pharmaceutical dosage form according to the invention is monolithic.
  • the pharmaceutical dosage form according to the invention comprises a core that is surrounded by a coating or by an encapsulating material.
  • the core is liquid and the pharmacologically active agent according to general formula (I) is dispersed, preferably dissolved in the liquid.
  • the pharmaceutical dosage form according to the invention provides the pharmacologically active agent according to general formula (I) in form of self-(micro) emulsifying drug delivery systems, solid solutions, nanoparticles, cyclodextrin complexes, liposomes, micelles, micronized and/or amorphous states.
  • the options for formulation of poorly water-soluble drugs include crystalline solid, amorphous and lipid formulations.
  • the dissolution rate of the pharmacologically active agent from crystalline formulations can be increased by particle size reduction, thereby increasing the surface area for dissolution, e.g. by conventional micronisation of the pharmacologically active agent to particle sizes of about 2-5 ⁇ m. In some cases, this is not sufficient and nanocrystal technology is applied. Nanocrystals show a particle size of 100-250 nm, which can be obtained by ball-milling or by dense gas or supercritical fluid technology.
  • Solid solutions provide and sustain the pharmacologically active agent in an amorphous or semi-amorphous state immobilized in a polymer.
  • Amorphous solutions may contain surfactants and polymers, thereby providing surface-activity during dispersion upon contact with water.
  • Solid solutions can be formed using a variety of technologies such as spray drying and melt extrusion.
  • Lipid formulations exhibiting different characteristics can be used to disperse and form micellar solutions, including simple solutions and self-emulsifying drug delivery systems (SEDDS). Depending on the excipients, some require digestion (e.g. simple oily liquids), others can easily be absorbed without digestion. Lipid formulations have been classified according to the lipid formulation classification system (LFCS) as follows:
  • Excipients Type Type Type Type Type in formulation I II IIIA IIIB IV Oil triglycerides 100 40-80 40-80 ⁇ 20 — or mixed mono- and diglycerides Water-insoluble — 20-60 — — 0-20 surfactants (HLB ⁇ 12) Water-soluble surfactants — — 20-40 20-50 30-80 (HLB > 12) Hydrophilic co-solvent — — 0-40 20-50 0-50
  • cyclodextrin complexes in which the pharmacologically active agent is located in the cavity of the cyclodextrin and is thereby molecularly present in a more soluble form in presence of aqueous media.
  • the success of the fitting strongly depends on the quality of the cyclodextrins as well as on the physicochemical properties and size of the pharmacologically active agent.
  • the pharmaceutical dosage form according to the invention can be regarded as a self emulsifying drug delivery system (SEDDS).
  • SEDDS self emulsifying drug delivery system
  • the pharmacologically active agent according to general formula (I) is preferably embedded in a self-emulsifying formulation.
  • a so called self emulsifying drug delivery system is a drug delivery system that uses an emulsion achieved by chemical rather than mechanical means. That is, by an intrinsic property of the drug formulation, rather than by special mixing and handling. Said formulation dilutes in aqueous media and results in an emulsion.
  • the self emulsifying drug delivery system SEDDS
  • SMEDDS self-micro emulsifying drug delivery system
  • these formulations are typically assigned to the group of type III formulations.
  • SEDDSs are self-emulsifying oily formulations (SEOF).
  • SEOFs typically comprise a natural or synthetic oil, surfactant and hydrophilic solvent and sometimes co-solvents.
  • the principal characteristic of SEOFs is their ability to form fine oil-in-water emulsions or micro emulsions upon mild agitation following dilution by aqueous phases. These formulations can disperse in the gastrointestinal lumen to form micro emulsions or fine emulsions, upon dilution with gastrointestinal fluids.
  • the pharmaceutical dosage form contains the pharmacologically active agent according to general formula (I) in form of a solid solution, i.e. molecularly dispersed in a solid matrix, so that preferably the pharmaceutical dosage form as such has an amorphous or semi-amorphous nature.
  • the solid solution preferably comprises the pharmacologically active agent according to general formula (I) in a molecular disperse form and an amorphous polymer matrix having a comparatively large specific surface.
  • the pharmacologically active agent according to general formula (I) is preferably present in a molecular disperse form, i.e. the compound is truly solved and evenly spread in the solidified solution.
  • the particle size of the compound is neither microcrystalline nor fine crystalline. The typical particle size is preferably from 0.1-1 ⁇ m.
  • the pharmacologically active agent according to general formula (I) is provided by means of a nanotechnological formulation with an average size of the nanoparticles of preferably less than 1 ⁇ m.
  • the pharmacologically active agent according to general formula (I) is preferably blended with the nanoparticles and thus adsorbed to the surface of the nanoparticles.
  • the nanoparticles are preferably selected from organic nanoparticles and inorganic nanoparticles.
  • Organic nanoparticles preferably contain small proteins which are present as a cluster or an agglomerate of small proteins, oligopeptides or lipids.
  • Inorganic nanoparticles preferably contain crystalline silicates. These silicates are from mineral origin or artificial silicates like metallosilicates (e.g. zeolites). In a preferred embodiment, the nanoparticles are modified in a way that they bear an electrostatic charge.
  • the nanoparticles are preferably ultra finely grounded silicates and the pharmacologically active agent according to general formula (I) is preferably bounded to the micro porous surface of the nanoparticles.
  • nanoparticles The formation of nanoparticles is known to a person skilled in the art.
  • One method is to produce colloidal nanoparticles as carriers for oral drug release by spraying the pharmacologically active agent according to general formula (I) under pressure at a defined temperature, together with a suitable carrier material like protamine, through jets, which are equipped with perforated strainers, into strongly cooled towers. The result of the fast cooling is an amorphous phase consisting of nanoparticles.
  • Another method is to blend the pharmacologically active agent according to general formula (I) with suitable macromolecules in solution. By adding hydrophobic compounds, solvent molecules are removed from the solution and desolvation occurs. For this reason the formation of very tiny particles takes place wherein the pharmacologically active agent according to general formula (I) is integrated. For a hardening of the formed nanoparticles a crosslinker may be added to the solution.
  • the method of high-pressure-homogenization and subsequent spray-cooling can be used.
  • the pharmacologically active agent according to general formula (I) is dissolved in a suitable solvent or in form of sub-micro particles.
  • a lipid vehicle and a surfactant may be added to the solution.
  • fine filler materials as outer phase as well as glidants and further surfactants may be added to fill the obtained formulation into e.g. capsules such as hard gelatin capsules.
  • the pharmacologically active agent according to general formula (I) are provided as cyclodextrin (inclusion) complexes.
  • Cyclodextrins are composed of sugar molecules forming a ring and typically comprising 5 or more ⁇ -D-glycopyranoside units which are linked via the 1-4 position. The typical number of connected sugar monomers ranges from 6 to 8 units.
  • a six membered sugar ring molecule is called ⁇ -cyclodextrin.
  • a seven membered sugar ring molecule is called ⁇ -cyclodextrin and an eight membered sugar ring molecule is called ⁇ -cyclodextrin.
  • the shape of these compounds is a toroid with the larger and the smaller openings exposed to the solvent. Due to this formation the inner part of the toroid is not hydrophobic, but considerably less hydrophilic than the aqueous environment and thus able to host hydrophobic molecules.
  • the outer part of the toroid is sufficiently hydrophilic to render cyclodextrins water solubility.
  • cyclodextrins greatly modifies the physical and chemical properties.
  • the mechanism of controlled degradation of such complexes and resultant drug release is based on pH change of aqueous solutions, leading to the cleavage of hydrogen or ionic bonds between the cyclodextrins and the included molecules.
  • Alternative means for the disruption of the complexes take advantage of heating or action of enzymes able to cleave ⁇ -1-4 linkages between ⁇ -D-glycopyranosides.
  • the pharmacologically active agent according to general formula (I) is provided in form of liposomes.
  • a liposome is preferably composed of phospholipids and is preferably of spherical shape.
  • the shell of this shape is preferably a lamellar or bilayer structure.
  • Another type of phospholipids arrangement is a monolayer.
  • Phospholipids comprise molecules with an amphiphilic character i.e. the molecules have a hydrophobic (lipophilic) and a hydrophilic (Iipophobic) part.
  • the hydrophilic part In the presence of water, the hydrophilic part is attracted to the water and forms a surface facing to the water, while the hydrophobic part is repelled by the water and forms a surface away from the water.
  • the amphiphilic molecules arrange themselves in one of the mentioned types.
  • the bilayer structures preferably arrange in a spherical shape wherein the inner part is filled with an aqueous solution.
  • This type is called “liposome”.
  • the hydrophobic parts of the molecules face each other in the middle of the layer and the hydrophilic parts of the molecules face the water molecules outside of the liposome.
  • the aqueous solution inside the liposome is the same as it is outside of the liposome.
  • Ingredients solved in this aqueous solution e.g. the pharmacologically active agents according to general formula (I), are in this way inside of the liposome.
  • a typical diameter of the liposomes is between 25 nm and 1 ⁇ m.
  • the smaller ones (25 nm-200 nm) are made of one single bilayer while the bigger ones (200 nm-1 ⁇ m) comprise more bilayer shells on the top of each other.
  • the monolayer structures also arrange in spherical shapes. Due to the amphiphilic character of the molecules and the spherical shape of the monolayer structures, the inner part of the spherical structures is filled with/formed by the hydrophobic parts of the molecules. These types are called micelles. There is no solvent inside the structure. In a preferred embodiment, the inner parts of the micelles contain the pharmacologically active agents according to general formula (I).
  • the pharmacologically active agent according to general formula (I) is provided in a micronized state.
  • particles of the pharmacologically active agent according to general formula (I) with a diameter in nanometer scale can be prepared. Said particles have a large surface to volume ratio.
  • Milling and grinding is a useful method to obtain particles in nanometer scale.
  • Sophisticated techniques for the micronization include RESS (rapid expansion of supercritical solutions), SAS (supercritical anti solvent) and the PGSS (particles from gas saturated solutions).
  • the RESS method uses a supercritical fluid wherein the pharmacologically active agent according to general formula (I) is dissolved under high pressure and temperature thereby yielding a homogenous supercritical phase. After expanding the solution through a nozzle, small particles are formed. Due to the expansion at the end of the nozzle the solved pharmacologically active agent according to general formula (I) precipitates as crystals and encloses small amounts of the solvent. The solvent changes from the supercritical fluid state to the normal state, preferred the gas phase, and breaks the crystals from inside-out. In this way and due to the fact that the crystals collide with each other, particles with a diameter in nanometer scale are formed.
  • the pharmacologically active agent according to general formula (I) is dissolved in a preferably organic solvent.
  • a supercritical fluid is added to the solution under pressure and thus forced to also dissolve in the solvent.
  • the volume of the complete system is increased and the solubility of the pharmacologically active agent according to general formula (I) is decreased. Due to its decreased solubility, the compound according to general formula (I) precipitates and forms particles having a small diameter.
  • the PGSS method is similar to the SAS method.
  • the pharmacologically active agent according to general formula (I) is melted and a supercritical fluid is dissolved in the melt. Due to the expansion through a nozzle, the pharmacologically active agent according to general formula (I) precipitates and forms particles in a nanometer scale.
  • the pharmaceutical dosage form according to the invention contains
  • the pharmacologically active agent according to general formula (I) is molecularly dispersed in a matrix.
  • the pharmacologically active agent according to general formula (I) is molecularly dispersed in a non-crystalline matrix.
  • the pharmacologically active agent according to general formula (I) is molecularly dispersed in a non-amorphous matrix.
  • the pharmacologically active agent according to general formula (I) is homogeneously distributed in the pharmaceutical dosage form according to the invention.
  • said two segments of the pharmaceutical dosage form having a volume of 1.0 mm 3 each are preferably segments of the core, i.e. do not contain any encapsulating medium or film coating, respectively.
  • the pharmaceutical dosage form according to the invention is characterized by a comparatively homogeneous distribution of density.
  • the densities of two segments of the pharmaceutical dosage form having a volume of 1.0 mm 3 each deviate from one another by not more than ⁇ 10%, more preferably not more than more than ⁇ 7.5%, still more preferably not more than ⁇ 5.0%, most preferably not more than ⁇ 2.5%, and in particular not more than ⁇ 1.0%.
  • said two segments of the pharmaceutical dosage form having a volume of 1.0 mm 3 each are preferably segments of the core, i.e. do not contain any encapsulating medium or film coating.
  • the pharmaceutical dosage form further contains a surfactant.
  • surfactant refers to any compound that contains at least one hydrophobic group and at least one hydrophilic group.
  • the surfactant contains at least one terminal hydrophobic group (tail) and at least one terminal hydrophilic group (head).
  • the hydrophobic group is preferably selected from the group consisting of hydrocarbon, alkyl ether, fluorocarbon and siloxan groups.
  • the surfactant contains at least one aliphatic group comprising at least 3 carbon atoms, more preferably at least 4 carbon atoms, still more preferably at least 6 carbon atoms, yet more preferably 6 to 30 carbon atoms, and most preferably 8 to 24 carbon atoms.
  • the aliphatic group may be a saturated or unsaturated, branched or unbranched (linear), terminal or internal aliphatic group.
  • the surfactant contains at least one group derivable from a saturated or unsaturated fatty acid or from a saturated or unsaturated fatty alcohol, which group is preferably an ether, carboxylic acid ester or sulfuric acid ester group.
  • the saturated or unsaturated fatty acid or fatty alcohol contains at least 6 carbon atoms, yet more preferably 6 to 30 carbon atoms, and most preferably 8 to 24 carbon atoms.
  • the surfactant contains at least one group derivable from a saturated or unsaturated fatty acid, preferably C 6 to C 30 fatty acid, more preferably C 8 to C 24 fatty acid, and most preferably C 12 to C 22 fatty acid.
  • suitable fatty acids are lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, lignoceric acid, 12-hydroxystearic acid, oleic acid and ricinoleic acid.
  • the surfactant contains at least one group derivable from a saturated or unsaturated fatty alcohol, preferably C 6 to C 30 fatty alcohol, more preferably C 8 to C 24 fatty alcohol, and most preferably C 12 to C 22 fatty alcohol.
  • suitable fatty alcohols are cetyl alcohol, stearyl alcohol, 2-octyldodecane-1-ol and 2-hexyldecane-1-ol.
  • the surfactant has a molecular weight of at most 20,000 g/mol, more preferably at most 15,000 g/mol, still more preferably at most 10,000 g/mol, yet more preferably at most 5,000 g/mol, even more preferably at most 4,000 g/mol, most preferably at most 3,000 g/mol, and in particular within the range of from 100 g/mol to 2,500 g/mol.
  • the surfactant is contained in a matrix in which the pharmacologically active agent according to general formula (I) is dispersed, preferably molecularly.
  • the pharmacologically active agent according to general formula (I) and the surfactant are intimately homogeneously distributed in a matrix so that the matrix does not contain any segments where either the pharmacologically active agent according to general formula (I) is present in the absence of the surfactant or where the surfactant is present in the absence of the pharmacologically active agent according to general formula (I).
  • the pharmaceutical dosage form contains a surfactant. In another preferred embodiment, the pharmaceutical dosage form contains a mixture of two or more surfactants.
  • the surfactant acts as an oil-in-water (O/W) emulsifier. In another preferred embodiment, the surfactant acts as a water-in-oil (W/O) emulsifier.
  • O/W oil-in-water
  • W/O water-in-oil
  • the pharmaceutical dosage form contains a surfactant having a hydrophilic-lipophilic balance (HLB) of at least 10 or at least 11. More preferably, the hydrophilic-lipophilic balance (HLB) is at least 12 or at least 13. Most preferably, the hydrophilic-lipophilic balance (HLB) ranges within 14 and 16.
  • HLB hydrophilic-lipophilic balance
  • the hydrophilic-lipophilic balance (HLB) of the surfactant is at most 30, more preferably at most 28, still more preferably at most 26, yet more preferably at most 24, even more preferably at most 22, most preferably at most 20 and in particular at most 18.
  • the hydrophilic-lipophilic balance (HLB) of the surfactant is at least 27, more preferably at least 29, still more preferably at least 31, yet more preferably at least 33, even more preferably at least 35, most preferably at least 37 and in particular at least 39.
  • An especially preferred surfactant of this type is sodium lauryl sulfave having an HLB value of about 40.
  • the HLB value of the surfactant is within the range of 10 ⁇ 3.5, more preferably 10 ⁇ 3, still more preferably 10 ⁇ 2.5, yet more preferably 10 ⁇ 2, even more preferably 10 ⁇ 1.5, most preferably 10 ⁇ 1, and in particular 10 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 12 ⁇ 3.5, more preferably 12 ⁇ 3, still more preferably 12 ⁇ 2.5, yet more preferably 12 ⁇ 2, even more preferably 12 ⁇ 1.5, most preferably 12 ⁇ 1, and in particular 12 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 14 ⁇ 3.5, more preferably 14 ⁇ 3, still more preferably 14 ⁇ 2.5, yet more preferably 14 ⁇ 2, even more preferably 14 ⁇ 1.5, most preferably 14 ⁇ 1, and in particular 14 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 15 ⁇ 3.5, more preferably 15 ⁇ 3, still more preferably 15 ⁇ 2.5, yet more preferably 15 ⁇ 2, even more preferably 15 ⁇ 1.5, most preferably 15 ⁇ 1, and in particular 15 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 16 ⁇ 3.5, more preferably 16 ⁇ 3, still more preferably 16 ⁇ 2.5, yet more preferably 16 ⁇ 2, even more preferably 16 ⁇ 1.5, most preferably 16 ⁇ 1, and in particular 16 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 18 ⁇ 3.5, more preferably 18 ⁇ 3, still more preferably 18 ⁇ 2.5, yet more preferably 18 ⁇ 2, even more preferably 18 ⁇ 1.5, most preferably 18 ⁇ 1, and in particular 18 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 30 ⁇ 3.5, more preferably 30 ⁇ 3, still more preferably 30 ⁇ 2.5, yet more preferably 30 ⁇ 2, even more preferably 30 ⁇ 1.5, most preferably 30 ⁇ 1, and in particular 30 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 32 ⁇ 3.5, more preferably 32 ⁇ 3, still more preferably 32 ⁇ 2.5, yet more preferably 32 ⁇ 2, even more preferably 32 ⁇ 1.5, most preferably 32 ⁇ 1, and in particular 32 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 34 ⁇ 3.5, more preferably 34 ⁇ 3, still more preferably 34 ⁇ 2.5, yet more preferably 34 ⁇ 2, even more preferably 34 ⁇ 1.5, most preferably 34 ⁇ 1, and in particular 34 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 36 ⁇ 3.5, more preferably 36 ⁇ 3, still more preferably 36 ⁇ 2.5, yet more preferably 36 ⁇ 2, even more preferably 36 ⁇ 1.5, most preferably 36 ⁇ 1, and in particular 36 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 38 ⁇ 3.5, more preferably 38 ⁇ 3, still more preferably 38 ⁇ 2.5, yet more preferably 38 ⁇ 2, even more preferably 38 ⁇ 1.5, most preferably 38 ⁇ 1, and in particular 38 ⁇ 0.5.
  • the HLB value of the surfactant is within the range of 40 ⁇ 3.5, more preferably 40 ⁇ 3, still more preferably 40 ⁇ 2.5, yet more preferably 40 ⁇ 2, even more preferably 40 ⁇ 1.5, most preferably 40 ⁇ 1, and in particular 40 ⁇ 0.5.
  • the surfactant can be ionic, amphoteric or non-ionic.
  • Suitable amphoteric surfactants include phospholipids, in particular lecithins such as soya bean lecithins.
  • the pharmaceutical dosage form contains an ionic surfactant, in particular an anionic surfactant.
  • Suitable anionic surfactants include but are not limited to sulfuric acid esters such as sodium lauryl sulfate (sodium dodecyl sulfate, e.g. Texapon® K12), sodium cetyl sulfate (e.g. Lanette E®), sodium cetylstearyl sulfate, sodium stearyl sulfate, sodium dioctylsulfosuccinate (docusate sodium), di-[2-ethylhexyl]-succinate; and the corresponding potassium or calcium salts thereof.
  • sulfuric acid esters such as sodium lauryl sulfate (sodium dodecyl sulfate, e.g. Texapon® K12), sodium cetyl sulfate (e.g. Lanette E®), sodium cetylstearyl sulfate, sodium stearyl sulfate, sodium dioctylsulfosuccinate (doc
  • the anionic surfactant has the general formula (II-a)
  • anionic surfactants include salts of cholic acid including sodium glycocholate (e.g. Konakion® MM, Cernevit®), sodium taurocholate and the corresponding potassium or ammonium salts.
  • the pharmaceutical dosage form contains a non-ionic surfactant.
  • Suitable non-ionic surfactants include but are not limited to
  • the pharmaceutical dosage form according to the invention comprises a surfactant or mixture of different surfactants obtainable by
  • the surfactant is selected from the group consisting of macrogolhydroxystearate, macrogolglycerylhydroxystearate and macrogolglyceryllaurate, wherein the macrogol moiety preferably comprises 15 to 45 ethylene oxide units.
  • Especially preferred surfactants of this class that are contained in the pharmaceutical dosage form according to the invention are non-ionic surfactants having a hydrophilic-lipophilic balance (HLB) of at least 10, in particular non-ionic surfactants having an HLB value of at least 12, more in particular non-ionic surfactant's having an HLB value within 14 and 16.
  • HLB hydrophilic-lipophilic balance
  • examples for this type of surfactants are the above-listed surfactants “polysorbate 80” (Tween® 80) and “Solutol® HS 15”.
  • Solutol® HS-15 is a mixture of polyethyleneglycol 660 12-hydroxystearate and polyethylene glycol. It is a white paste at room temperature that becomes liquid at about 30° C. and has an HLB of about 15.
  • Tween® 80 polyoxyethylene(20)sorbitan monooleate
  • Tween® 80 is liquid at room temperature, has a viscosity of 375-480 mPa ⁇ s and has an HLB of about 15.
  • the pharmaceutical dosage form according to the invention contains a mixture of at least one surfactant having a HLB value of at least 10 (hydrophilic surfactant) and at least one surfactant having a HLB value below 10 (lipophilic surfactant).
  • the dosage form may contain macrogol-glycerolhydroxystearat 40 (e.g., Cremophor® RH 40) as the hydrophilic surfactant component and glyceryl monooleate 40 (e.g., Peceol®) as the lipophilic surfactant component.
  • the relative weight ratio of the surfactant having a HLB value of at least 10 (hydrophilic surfactant) and the surfactant having a HLB value below 10 (lipophilic surfactant) is within the range of 15:1 to 1:20, more preferably 10:1 to 1:15, still more preferably 8:1 to 1:12, yet more preferably 6:1 to 1:10, even more preferably 5:1 to 1:7, most preferably 4:1 to 1:4 and in particular 2:1 to 1:2.
  • the content of the surfactant is at least 0.001 wt.-% or at least 0.005 wt.-%, more preferably at least 0.01 wt.-% or at least 0.05 wt.-%, still more preferably at least 0.1 wt.-%, at least 0.2 wt.-%, or at least 0.3 wt.-%, yet more preferably at least 0.4 wt.-%, at least 0.5 wt.-%, or at least 0.6 wt.-%, and in particular at least 0.7 wt.-%, at least 0.8 wt.-%, at least 0.9 wt.-%, or at least 1.0 wt.-%, based on the total weight of the pharmaceutical dosage form.
  • the content of the surfactant is at least 10 wt.-%, more preferably at least 15 wt.-%, still more preferably at least 20 wt.-%, yet more preferably at least 25 wt.-% and in particular at least 30 wt.-%, based on the total weight of the composition forming the core.
  • the content of the surfactant ranges preferably from 0.1 wt.-% to 95 wt.-%, more preferably from 1 wt.-% to 95 wt.-%, still more preferably from 5 wt.-% to 90 wt.-%, yet more preferably from 10 wt.-% to 80 wt.-%, most preferably from 20 wt.-% to 70 wt.-%, and in particular from 30 wt.-% to 75 wt.-%, based on the total weight of the composition forming the core.
  • the pharmaceutical dosage form contains a core that is encapsulated by an encapsulating medium.
  • the core can be liquid, semi-liquid or solid.
  • said encapsulating medium is a soft gelatin capsule or a hard gelatin capsule, in particular a hard gelatin capsule.
  • the pharmaceutical dosage form comprises a liquid core encapsulated by a solid material, wherein the pharmacologically active agent according to general formula (I) is dispersed in the liquid core.
  • the solid material is a hard gelatin capsule.
  • the pharmaceutical dosage form according to the invention contains a self-emulsifying formulation in which the pharmacologically active agent according to general formula (I) is preferably embedded.
  • the pharmacologically active agent according to general formula (I) is molecularly dispersed in the other ingredients of liquid core.
  • “molecularly dispersed in a liquid core”, e.g. in the other ingredients of the liquid core, means that a substantial portion of the overall content of the pharmacologically active agent according to general formula (I) is present in non-crystalline form, i.e. does not provide X-ray reflexes.
  • the pharmacologically active agent according to general formula (I) is dissolved in the other ingredients of the core.
  • the content of non-crystalline pharmacologically active agent according to general formula (I) is at least 60 wt.-%, more preferably at least 65 wt.-%, still more preferably at least 70 wt.-%, yet more preferably at least 75 wt.-%, even more preferably at least 80 wt.-%, most preferably at least 85 wt.-%, and in particular at least 90 wt.-%, based on the total content of pharmacologically active agent according to general formula (I).
  • the self-emulsifying formulation contains the surfactant and an oil.
  • the self-emulsifying formulation is a self-emulsifying oily formulation (SEOF), i.e. it comprises the surfactant, the oil and additionally a hydrophilic solvent.
  • SEOF self-emulsifying oily formulation
  • an oil is preferably to be regarded as any substance that is liquid at ambient temperatures or has a melting point below 70° C. and is hydrophobic but soluble in organic solvents.
  • the oil is a C 12 -C 18 -fatty acid ester of a monoalcohol (e.g. C 1 -C 12 -alkylalcohols), a di-C 12 -C 18 -fatty acid ester of a dialcohol (e.g. ethylene glycol) or tri-C 12 -C 18 -fatty acid ester of a trialcohol (e.g. glycerol).
  • a monoalcohol e.g. C 1 -C 12 -alkylalcohols
  • a di-C 12 -C 18 -fatty acid ester of a dialcohol e.g. ethylene glycol
  • tri-C 12 -C 18 -fatty acid ester of a trialcohol e.g. glycerol
  • the oil has a melting point below 60° C., more preferably below 55° C., still more preferably below 50° C., yet more preferably below 45° C., even more preferably below 40° C., most preferably below 35° C. and in particular below 30° C.
  • the pure oil has a density within the range of 0.94 ⁇ 0.07 g/cm 3 , more preferably 0.94 ⁇ 0.06 g/cm 3 , still more preferably 0.94 ⁇ 0.05 g/cm 3 , yet more preferably 0.94 ⁇ 0.04 g/cm 3 , even more preferably 0.94 ⁇ 0.03 g/cm 3 , most preferably 0.94 ⁇ 0.02 g/cm 3 , and in particular 0.94 ⁇ 0.01 g/cm 3 .
  • the pure oil has a viscosity at 20° C. measured in accordance with Ph. Eur. 2.2.8, within the range of 30 ⁇ 9 mPas, more preferably 30 ⁇ 8 mPas, still more preferably 30 ⁇ 7 mPas, yet more preferably 30 ⁇ 6 mPas, even more preferably 30 ⁇ 5 mPas, most preferably 30 ⁇ 4 mPas, and in particular 30 ⁇ 3 mPas.
  • the oil is selected from the group consisting of
  • medium-chain triglycerides according to Ph. Eur. or USP such as said caprylic/capric triglyceride mixtures.
  • the content of the oil in the pharmaceutical dosage form is within the range of from 1 wt.-% to 90 wt.-%, preferably from 2 wt.-% to 80 wt.-%, more preferably from 5 wt.-% to 60 wt.-%, still more preferably from 10 wt.-% to 50 wt.-% and most preferably from 15 wt.-% to 30 wt.-%, preferably based on the total weight of the core.
  • the relative weight ratio of the surfactant to the oil is within the range of from 20:1 to 1:20, more preferably 10:1 to 1:10, still more preferably 7.5:1 to 1:5, yet more preferably 7:1 to 1:1, most preferably 5:1 to 1.5:1 and in particular 4:1 to 2:1.
  • the content of the surfactant ranges preferably from 0.001 wt.-% to 95 wt.-%, more preferably from 0.01 wt.-% to 50 wt.-%, still more preferably from 0.1 wt.-% to 20 wt.-%, yet more preferably from 0.15 wt.-% to 15 wt.-%, most preferably from 0.2 wt.-% to 10 wt.-%, and in particular from 0.25 wt.-% to 5 wt.-%, based on the total weight of the dosage form.
  • the content of the surfactant is at most 25 wt.-%, more preferably at most 20 wt.-%, still more preferably at most 15 wt.-%, yet more preferably at most 10 wt.-% and in particular at most 5 wt.-%, based on the total weight of the pharmaceutical dosage form.
  • the indication “wt.-%” preferably refers to the weight of the surfactant per total weight of the composition forming the core, i.e. the pharmaceutical dosage form without its coating.
  • the content of the surfactant is within the range of 1 ⁇ 0.7 wt.-%, more preferably 1 ⁇ 0.6 wt.-%, still more preferably 1 ⁇ 0.5 wt.-%, yet more preferably 1 ⁇ 0.4 wt.-%, even more preferably 1 ⁇ 0.3 wt.-%, most preferably 1 ⁇ 0.2 wt.-%, and in particular 1 ⁇ 0.1 wt.-%
  • the self-emulsifying formulation is present as the liquid core, encapsulated by a hard gelatin capsule.
  • the self-emulsifying formulation further contains a hydrophilic solvent.
  • the hydrophilic solvent is an organic alcohol such as an organic monoalcohol, organic dialcohol or organic trialcohol.
  • the pure hydrophilic solvent has a boiling point at ambient pressure within the range of 78 ⁇ 22° C., more preferably 78 ⁇ 18° C., still more preferably 78 ⁇ 15° C., yet more preferably 78 ⁇ 12° C., even more preferably 78 ⁇ 8° C., most preferably 78 ⁇ 5° C., and in particular 78 ⁇ 2° C.
  • the hydrophilic solvent is selected from the group ethanol, isopropanol, glycerol and propylene glycol; especially preferred is ethanol.
  • the content of the hydrophilic solvent is within the range of from about 1 wt.-% to about 90 wt.-%, preferably from about 2 wt.-% to about 80 wt.-%, more preferably from about 5 wt.-% to about 60 wt.-%, still more preferably from about 10 wt.-% to about 50 wt.-%, most preferably from about 15 wt.-% to about 30 wt.-%, preferably based on the total weight of the core.
  • the pharmaceutical dosage form contains a liquid core comprising the pharmacologically active agent according to general formula (I), a surfactant, an oil and a hydrophilic solvent, wherein the relative weight ratio of surfactant:oil:hydrophilic solvent is within the range of 60:20 ⁇ 17.5:20 ⁇ 17.5, more preferably 60:20 ⁇ 15:20 ⁇ 15, still more preferably 60:20 ⁇ 12.5:20 ⁇ 12.5, yet more preferably 60:20 ⁇ 10:20 ⁇ 10, even more preferably 60:20 ⁇ 7.5:20 ⁇ 7.5, most preferably 60:20 ⁇ 5:20 ⁇ 5, and in particular 60:20 ⁇ 2.5:20 ⁇ 2.5.
  • the relative weight ratio of surfactant:oil:hydrophilic solvent is within the range of 60:20 ⁇ 17.5:20 ⁇ 17.5, more preferably 60:20 ⁇ 15:20 ⁇ 15, still more preferably 60:20 ⁇ 12.5:20 ⁇ 12.5, yet more preferably 60:20 ⁇ 10:20 ⁇ 10, even more preferably 60:20 ⁇ 7.5:20 ⁇ 7.5, most preferably 60:
  • the pharmaceutical dosage form contains a liquid core comprising the pharmacologically active agent according to general formula (I), a surfactant having a HLB value of at least 10 (hydrophilic surfactant), an oil and a surfactant having a HLB value below 10 (lipophilic surfactant), wherein the relative weight ratio of hydrophilic:oil:lipophilic solvent is within the range of 60:20 ⁇ 17.5:20 ⁇ 17.5, more preferably 60:20 ⁇ 15:20 ⁇ 15, still more preferably 60:20 ⁇ 12.5:20 ⁇ 12.5, yet more preferably 60:20 ⁇ 10:20 ⁇ 10, even more preferably 60:20 ⁇ 7.5:20 ⁇ 7.5, most preferably 60:20 ⁇ 5:20 ⁇ 5, and in particular 60:20 ⁇ 2.5:20 ⁇ 2.5.
  • hydrophilic surfactant having a HLB value of at least 10
  • lipophilic surfactant an oil and a surfactant having a HLB value below 10
  • the relative weight ratio of hydrophilic:oil:lipophilic solvent is within the range of
  • the pharmaceutical dosage form contains a liquid core comprising the pharmacologically active agent according to general formula (I), a surfactant having a HLB value of at least 10 (hydrophilic surfactant), an oil and a surfactant having a HLB value below 10 (lipophilic surfactant), wherein the relative weight ratio of hydrophilic:oil:lipophilic solvent is within the range of 40:40 ⁇ 35:20 ⁇ 17.5, more preferably 40:40 ⁇ 30:20 ⁇ 15, still more preferably 40:40 ⁇ 25:20 ⁇ 12.5, yet more preferably 40:40 ⁇ 20:20 ⁇ 10, even more preferably 40:40 ⁇ 15:20 ⁇ 7.5, most preferably 40:40 ⁇ 10:20 ⁇ 5, and in particular 40:40 ⁇ 5:20 ⁇ 2.5.
  • a surfactant having a HLB value of at least 10 hydrophilic surfactant
  • lipophilic surfactant an oil and a surfactant having a HLB value below 10
  • the relative weight ratio of hydrophilic:oil:lipophilic solvent is within
  • embodiment A 9 relates to a pharmaceutical dosage according to the invention, which contains a pharmacologically active agent according to general formula (I′) or a physiologically acceptable salt thereof in an amount of 0.25 ⁇ 0.24 wt.-%, a non-ionic surfactant having a HLB value of between 14 and 16 in an amount of 50 ⁇ 15 wt.-%, triglycerides of the C 6 to C 12 fatty acids in an amount of 25 ⁇ 7.5% and a hydrophilic solvent selected from organic monoalcohol, dialcohol or trialcohol in an amount of 25 ⁇ 7.5%, based on the total weight of the liquid core.
  • a pharmaceutical dosage according to the invention which contains a pharmacologically active agent according to general formula (I′) or a physiologically acceptable salt thereof in an amount of 0.25 ⁇ 0.24 wt.-%, a non-ionic surfactant having a HLB value of between 14 and 16 in an amount of 50 ⁇ 15 wt.-%, triglycerides of the C
  • the self-emulsifying formulation is a lipid formulation of type IIIA or type IIIB, according to the lipid formulation classification system (LFCS).
  • LFCS lipid formulation classification system
  • the self emulsifying formulation gives emulsions with an average droplet size smaller than or equal to 10 micrometers, more preferably smaller than or equal to 1000 nanometers, most preferably smaller than or equal to 100 nanometers, when exposed to aqueous media.
  • the self-emulsifying formulation is a self-micro emulsifying drug delivery system (SMEDDS), i.e. when exposed to aqueous media, the formulation gives microemulsions with an average droplet size smaller than or equal to 50 nanometers, which contain the pharmacologically active agent according to general formula (I). In another preferred embodiment, the average droplet size is smaller than or equal to 10 nanometers
  • SMEDDS self-micro emulsifying drug delivery system
  • the average droplet size is within the range of 50 ⁇ 70 nm, more preferably 50 ⁇ 60 nm, still more preferably 50 ⁇ 50 nm, yet more preferably 50 ⁇ 40 nm, even more preferably 50 ⁇ 30 nm, most preferably 50 ⁇ 20 nm, and in particular 50 ⁇ 10 nm.
  • the average droplet size is within the range of 75 ⁇ 70 nm, more preferably 75 ⁇ 60 nm, still more preferably 75 ⁇ 50 nm, yet more preferably 75 ⁇ 40 nm, even more preferably 75 ⁇ 30 nm, most preferably 75 ⁇ 20 nm, and in particular 75 ⁇ 10 nm.
  • the average droplet size is within the range of 100 ⁇ 70 nm, more preferably 100 ⁇ 60 nm, still more preferably 100 ⁇ 50 nm, yet more preferably 100 ⁇ 40 nm, even more preferably 100 ⁇ 30 nm, most preferably 100 ⁇ 20 nm, and in particular 100 ⁇ 10 nm.
  • the average droplet size is within the range of 125 ⁇ 70 nm, more preferably 125 ⁇ 60 nm, still more preferably 125 ⁇ 50 nm, yet more preferably 125 ⁇ 40 nm, even more preferably 125 ⁇ 30 nm, most preferably 125 ⁇ 20 nm, and in particular 125 ⁇ 10 nm.
  • the average droplet size is within the range of 150 ⁇ 70 nm, more preferably 150 ⁇ 60 nm, still more preferably 150 ⁇ 50 nm, yet more preferably 150 ⁇ 40 nm, even more preferably 150 ⁇ 30 nm, most preferably 150 ⁇ 20 nm, and in particular 150 ⁇ 10 nm.
  • the pharmaceutical dosage form according to the invention particularly when it contains the pharmacologically active agent according to general formula (I) in form of a solid solution, i.e. molecularly dispersed in a solid matrix, may further contain at least one matrix material.
  • said matrix material comprises a polymer selected from the group consisting of polyvinylpyrrolidone, vinylpyrrolidone-polyvinylacetate copolymers, cellulose derivatives, preferably cellulose esters or cellulose ethers, such as for example hydroxypropyl methylcellulose, hydroxypropyl methylcellulose acetate succinate, hydroxy-propyl methylcellulose phthalate, ethylcellulose, polymethacrylates, polyethylene oxides, polyethylene glycols and any combinations thereof.
  • polyvinyl-pyrrolidone are commercialized as Kollidon® 90 and examples of vinylpyrrolidone-polyvinyl acetate copolymer are commercialized as Kollidon® VA64.
  • molecularly dispersed in a solid matrix means that a substantial portion of the overall content of the pharmacologically active agent according to general formula (I) is present in non-crystalline form, i.e. does not provide X-ray reflexes.
  • the content of non-crystalline pharmacologically active agent according to general formula (I) is at least 60 wt.-%, more preferably at least 65 wt.-%, still more preferably at least 70 wt.-%, yet more preferably at least 75 wt.-%, even more preferably at least 80 wt.-%, most preferably at least 85 wt.-%, and in particular at least 90 wt.-%, based on the total content of pharmacologically active agent corresponding to formula (I).
  • the pharmaceutical dosage form according to the invention contains a polymer with a weight average molecular weight of preferably at least 50,000 g/mol, more preferably at least 100,000 g/mol, yet more preferably at least 250,000 g/mol, still more preferably at least 500,000 g/mol, most preferably at least 750,000 g/mol and in particularly at least 800,000 g/mol.
  • the pharmaceutical dosage form according to the invention contains a polymer with a weight average molecular weight of preferably at least 5000 g/mol, more preferably at least 10,000 g/mol, yet more preferably at least 20,000 g/mol, still more preferably at least 30,000 g/mol, even more preferably at least 40,000 g/mol, most preferably at least 50,000 g/mol and in particular within the range of from 50,000 g/mol to 250,000 g/mol.
  • the pharmaceutical dosage form according to the invention contains the pharmacologically active agent according to general formula (I) in form of a solid solution, i.e. molecularly dispersed in a solid matrix, wherein the matrix comprises one or more polymers and wherein the content of the polymer(s) is within the range of 25 ⁇ 22.5 wt.-%, more preferably 25 ⁇ 20 wt.-%, still more preferably 25 ⁇ 17.5 wt.-%, yet more preferably 25 ⁇ 15 wt.-%, even more preferably 25 ⁇ 12.5 wt.-%, most preferably 25 ⁇ 10 wt.-% and in particular 25 ⁇ 7.5 wt.-%, based on the total weight of the pharmaceutical dosage form.
  • a solid solution i.e. molecularly dispersed in a solid matrix
  • the matrix comprises one or more polymers and wherein the content of the polymer(s) is within the range of 25 ⁇ 22.5 wt.-%, more preferably 25 ⁇ 20 wt.-
  • the pharmaceutical dosage form according to the invention contains the pharmacologically active agent according to general formula (I) in form of a solid solution, i.e. molecularly dispersed in a solid matrix, wherein the matrix comprises one or more polymers and wherein the content of the polymer(s) is within the range of 50 ⁇ 22.5 wt.-%, more preferably 50 ⁇ 20 wt.-%, still more preferably 50 ⁇ 17.5 wt.-%, yet more preferably 50 ⁇ 15 wt.-%, even more preferably 50 ⁇ 12.5 wt.-%, most preferably 50 ⁇ 10 wt.-% and in particular 50 ⁇ 7.5 wt.-%, based on the total weight of the pharmaceutical dosage form.
  • a solid solution i.e. molecularly dispersed in a solid matrix
  • the matrix comprises one or more polymers and wherein the content of the polymer(s) is within the range of 50 ⁇ 22.5 wt.-%, more preferably 50 ⁇ 20 wt.-
  • the pharmaceutical dosage form according to the invention contains the pharmacologically active agent according to general formula (I) in form of a solid solution, i.e. molecularly dispersed in a solid matrix, wherein the matrix comprises one or more polymers and wherein the content of the polymer(s) is within the range of 75 ⁇ 22.5 wt.-%, more preferably 75 ⁇ 20 wt.-%, still more preferably 75 ⁇ 17.5 wt-%, yet more preferably 75 ⁇ 15 wt.-%, even more preferably 75 ⁇ 12.5 wt.-%, most preferably 75 ⁇ 10 wt.-% and in particular 75 ⁇ 7.5 wt.-%, based on the total weight of the pharmaceutical dosage form.
  • a solid solution i.e. molecularly dispersed in a solid matrix
  • the matrix comprises one or more polymers and wherein the content of the polymer(s) is within the range of 75 ⁇ 22.5 wt.-%, more preferably 75 ⁇ 20 wt.-%
  • the pharmaceutical dosage form according to the invention contains a polymer which comprises repeating units derived from vinylpyrrolidones.
  • the polymer comprises monomer units derived from vinyl acetates.
  • the polymer is a copolymer comprising repeating units derived from vinylpyrrolidones and repeating units derived from vinyl acetates, wherein the weight ratio of repeating units derived from vinylpyrrolidones:repeating units derived from vinyl acetates is preferably at most 10:1, more preferably at most 4.5:1, still more preferably at most 4:1, most preferably at most 2:1 and in particular at most 1.5:1.
  • the relative weight ratio of the polymer related to the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form according to the invention ranges from 1:1 to 70:1, more preferably from 2:1 to 50:1, still more preferably from 3:1 to 40:1, most preferably from 3.5:1 to 30:1 and in particular from 4:1 to 19:1.
  • the relative weight ratio of the polymer related to the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form according to the invention is at least 3:1 or at least 4:1, more preferably at least 5:1 or at least 6:1, still more preferably at least 7:1 or at least 8:1, yet more preferably at least 9:1 or at least 10:1, even more preferably at least 11:1 or at least 12:1, most preferably at least 13:1 or at least 14:1 and in particular at least 15:1 or at least 16:1.
  • the relative weight ratio of the polymer, especially of polyvinyl-pyrrolidone, related to the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form according to the invention ranges from 1:1 to 70:1, more preferably from 2:1 to 45:1, still more preferably from 3:1 to 20:1, most preferably from 3.5:1 to 10:1 and in particular from 4:1 to 5:1.
  • the relative weight ratio of the polymer, especially of vinylpyrrolidone-vinyl acetate copolymer, related to the pharmacologically active agent according to general formula (I) in the pharmaceutical dosage form according to the invention ranges from 1:1 to 70:1, more preferably from 2:1 to 55:1, still more preferably from 4:1 to 40:1, most preferably from 10:1 to 25:1 and in particular from 15:1 to 25:1.
  • the pharmaceutical dosage form according to the invention particularly when it contains the pharmacologically active agent according to general formula (I) in form of a solid solution, i.e. molecularly dispersed in a solid matrix, may further contain at least one surfactant selected from the group containing partial fatty acid esters of polyoxyethylene sorbitan (polyoxyethylene-sorbitan-fatty acid esters), preferably a fatty acid monoester of polyoxyethylene sorbitan, a fatty acid diester of polyoxyethylene sorbitan, or a fatty acid triester of polyoxyethylene sorbitan; sulfuric acid esters, or the alkali or earthalkali salts thereof; and poloxamers.
  • polyoxyethylene sorbitan polyoxyethylene-sorbitan-fatty acid esters
  • a fatty acid monoester of polyoxyethylene sorbitan preferably a fatty acid monoester of polyoxyethylene sorbitan, a fatty acid diester of polyoxyethylene sorbitan, or a
  • the pharmaceutical dosage form according to the invention contains a surfactant with a weight average molecular weight of preferably at least 500 g/mol, more preferably at least 1,000 g/mol, yet more preferably at least 2,500 g/mol, still more preferably at least 5,000 g/mol, most preferably at least 7,000 g/mol and in particularly at least 8,000 g/mol.
  • the pharmaceutical dosage form according to the invention contains a surfactant with a weight average molecular weight of preferably at least 100 g/mol, more preferably at least 250 g/mol, yet more preferably at least 500 g/mol, still more preferably at least 750 g/mol, most preferably at least 1,000 g/mol and in particularly at least 1,250 g/mol.
  • the dosage form according to the invention contains a surfactant, preferably [polyoxyethylene(20)sorbitan monooleate] or polyoxyethylene-polyoxypropylene block co-polymer, in a content of preferably 0.5 wt-% to 80 wt-%, more preferably 1.5 wt-% to 60 wt-%, still more preferably 2.5 wt-% to 50 wt-%, yet more preferably 3.0 wt-% to 40 wt-%, most preferably 3.5 wt-% to 20 wt-%, and in particular 4 wt-% to 10 wt-%, based on the total weight of the pharmaceutical dosage form.
  • a surfactant preferably [polyoxyethylene(20)sorbitan monooleate] or polyoxyethylene-polyoxypropylene block co-polymer
  • Preferred embodiment A 1 to A 20 of the pharmaceutical dosage form according to the invention are summarized in the following Table 2:
  • embodiment A 9 relates to a pharmacologically active agent according to general formula (I′) or a physiologically acceptable salt thereof, a vinylpyrrolidone-polyvinylacetate copolymer and a surfactant selected from the group of polysorbitanes, wherein the weight ratio is 2:38:5.
  • the content of the surfactant is within the range of 1.00 ⁇ 0.70 wt.-%, more preferably 1.00 ⁇ 0.60 wt.-%, still more preferably 1.00 ⁇ 0.50 wt.-%, yet more preferably 1.00 ⁇ 0.40 wt.-%, even more preferably 1.00 ⁇ 0.30 wt.-%, most preferably 1.00 ⁇ 0.20 wt.-%, and in particular 1.00 ⁇ 0.10 wt.-%
  • embodiment A 9 relates to a pharmaceutical dosage according to the invention, which contains the pharmacologically active agent according to general formula (I) in an amount of 0.50 ⁇ 0.45 wt.-% and a surfactant having a HLB value of at least 10 in an amount of 2.75 ⁇ 2.50 wt.-%, based on the total weight of the dosage form.
  • the pharmacologically active agent according to general formula (I) in an amount of 0.50 ⁇ 0.45 wt.-% and a surfactant having a HLB value of at least 10 in an amount of 2.75 ⁇ 2.50 wt.-%, based on the total weight of the dosage form.
  • the pharmaceutical dosage form according to the invention is a tablet, chewable tablet, chewing gum, coated tablet or powder, optionally filled into a capsule, but particularly preferably a tablet.
  • the pharmaceutical dosage form according to the invention is present in multi-particulate form, preferably in form of a micro-tablet, micro-capsule, granulate, pellet or active-substance crystal, particularly preferably in form of a micro-tablet, granulate or pellet, optionally filled into a capsule or compressed to form a tablet.
  • the pharmaceutical dosage form according to the invention is a tablet.
  • the tablet can be of any desired size.
  • the tablet has a diameter of 6 ⁇ 3 mm, more preferably of 6 ⁇ 2.5 mm, even more preferably of 6 ⁇ 2 mm, still more preferably of 6 ⁇ 1.5 mm, most preferably of 6 ⁇ 1 mm, and in particular of 6 ⁇ 0.5 mm.
  • the pharmaceutical dosage form according to the invention may contain pharmaceutical excipients including conventional antiadherents, binders, disintegrants, fillers, diluents, glidants, lubricants and preservatives, known to a person skilled in the art.
  • One suitable antiadherent that may be contained in the pharmaceutical dosage form is magnesium stearate.
  • the content of the antiadherent is within the range of from 0.001 to 5.0 wt.-%, for example 0.01 to 5 wt.-%, 0.1 to 5 wt.-%, 0.1 to 3 wt.-%, 0.1 to 2 wt.-%, or even 0.5 to 1.5 wt.-%.
  • the pharmaceutical dosage form according to the invention further contains a binder.
  • Suitable binders include but are not limited to gelatin, cellulose, modified cellulose such as microcrystalline cellulose, methyl cellulose, polyvinyl pyrrolidone, starch, sucrose and polyethylene glycol; especially preferred are polyvinyl pyrrolidone and/or microcrystalline cellulose.
  • the content of antiadherent is within the range of from 0.001 to 30 wt.-%, more preferably 0.1 to 25 wt.-%.
  • the dosage form comprises 1 to 20 wt.-%, 5 to 20 wt.-%, or 10 to 20 wt.-% of binder(s).
  • the pharmaceutical dosage form according to the invention further contains a filler and/or diluent, preferably selected from the group consisting of but are not limited to cellulose, calcium diphosphate, lactose, sucrose, glucose, mannitol, sorbitol, and calcium carbonate; especially preferred are micro-crystalline cellulose and lactose.
  • the content of filler and/or diluent is within the range of from 0.001 to 90 wt.-%, 0.01 to 85 wt.-%, more preferably 0.1 to 80 wt.-%, most preferably 10 to 75 wt.-%.
  • the pharmaceutical dosage form according to the invention further contains a lubricant such as magnesium stearate, stearic acid and stearin.
  • a lubricant such as magnesium stearate, stearic acid and stearin.
  • the content of the lubricant is within the range of from 0.001 to 5 wt.-%, e.g., from 0.1 to 3 wt. %.
  • the pharmaceutical dosage form according to the invention further contains a disintegrant such as cross-linked sodium carboxymethyl cellulose (croscarmellose sodium), cross-linked polyvinyl pyrrolidone and sodium starch glycolate.
  • a disintegrant such as cross-linked sodium carboxymethyl cellulose (croscarmellose sodium), cross-linked polyvinyl pyrrolidone and sodium starch glycolate.
  • the content of the disintegrant is within the range of from 0.001 to 5 wt.-%, e.g., from 0.1 to 3 wt. %.
  • the pharmaceutical dosage form may further contain at least one preservative.
  • Suitable preservatives include but are not limited to antioxidants, such as vitamin A, vitamin E, vitamin C, retinyl palmitate and selenium; cysteine, methionine, citric acid, sodium citrate, methyl paraben and propyl paraben.
  • the pharmaceutical dosage form further contains a coating, in particular a polymer-based coating, more in particular a polyvinyl alcohol-based coating such as the ones commercially available under the trade name “Opadry”.
  • a coating in particular a polymer-based coating, more in particular a polyvinyl alcohol-based coating such as the ones commercially available under the trade name “Opadry”.
  • the pharmaceutical dosage form is a tablet which comprises the pharmacologically active agent according to the general formula (I) (e.g., in an amount from 0.6 ⁇ 0.4 wt.-%, 0.6 ⁇ 0.3 wt.-%, 0.6 ⁇ 0.2 wt.-%, 0.6 ⁇ 0.1 wt.-%, 0.04 ⁇ 0.03 wt.-%, 0.04 ⁇ 0.02 wt-%, or 0.04 ⁇ 0.01 wt.-%), one or more antiadherents (e.g., magnesium stearate) in an amount from 0.001 to 5.0 wt.-% (e.g., 0.01 to 5 wt.-%, 0.1 to 5 wt.-%, 0.1 to 3 wt.-%, 0.1 to 2 wt.-%, or even 0.5 to 1.5 wt.-%), one a more binders (e.g., polyvinyl pyrrolidone and/or microcrystalline cellulose) in an adherent
  • the tablet also comprises one or more lubricants (e.g., magnesium stearate, stearic acid and/or stearin) in an amount from 0.001 to 5 wt.-% (e.g., from 0.1 to 3 wt. %) and/or one or more disintegrants (e.g., croscarmellose sodium, cross-linked polyvinyl pyrrolidone and/or sodium starch glycolate) in an amount from 0.001 to 5 wt.-% (e.g., from 0.1 to 3 wt. %).
  • lubricants e.g., magnesium stearate, stearic acid and/or stearin
  • disintegrants e.g., croscarmellose sodium, cross-linked polyvinyl pyrrolidone and/or sodium starch glycolate
  • the coating protects the pharmaceutical dosage form from moisture, but dissolves rapidly in gastric juice. More preferably, the coated dosage form has a disintegration time of less than 5 minutes in gastric juice, more preferably of at most 4.5 minutes, still more preferably at most 4 minutes, yet more preferably at most 3.5 minutes, even more preferably at most 3 minutes, most preferably at most 2.5 minutes and in particular at most 2 minutes.
  • the various solid auxiliary substances and the pharmacologically active agent according to general formula (I) are preferably homogenized, processed by means of wet, dry or fusion granulation to form granulates, and compressed to form tablets. Alternatively, they are manufactured by direct tabletting of the auxiliary substances and the pharmacologically active agent according to general formula (I).
  • the pharmaceutical dosage form is prepared by means of wet granulation from a granulating fluid containing the pharmacologically active agent according to general formula (I), in particular from an aqueous granulating fluid containing said pharmacologically active agent and the surfactant.
  • a granulating fluid containing the pharmacologically active agent according to general formula (I)
  • the resulting granulating fluid is then top-sprayed or bottom-sprayed onto a solid formulation containing at least one auxiliary substance to yield compressible granules, which may optionally be mixed with further auxiliary substances before being compressed to tablets.
  • a further aspect of the invention relates to the pharmaceutical dosage form according to the invention as described above for use in the treatment of pain.
  • a further aspect of the invention relates to a method of treating pain comprising the twice daily, once daily, or less frequently, preferably oral administration of the pharmaceutical dosage form according to the invention to a subject in need thereof.
  • the pain is selected from acute, visceral, neuropathic or chronic pain.
  • a further aspect of the invention relates to a method of treating neuropathic pain, preferably chronic neuropathic pain comprising the administration of a pharmacologically effective amount of the pharmacologically active agent according to general formula (I) or a physiologically acceptable salt thereof; preferably the once daily, preferably oral administration of the pharmaceutical dosage form according to the invention; to a subject in need thereof.
  • the pain is chronic neuropathic pain or acute neuropathic pain, peripheral neuropathic pain or central neuropathic pain, mononeuropathic pain or polyneuropathic pain.
  • the neuropathic pain is chronic, it may be chronic peripheral neuropathic pain or chronic central neuropathic pain, in a preferred embodiment chronic peripheral mononeuropathic pain or chronic central mononeuropathic pain, in another preferred embodiment chronic peripheral polyneuropathic pain or chronic central polyneuropathic pain.
  • the neuropathic pain is acute, it may be acute peripheral neuropathic pain or acute central neuropathic pain, in a preferred embodiment acute peripheral mononeuropathic pain or acute central mononeuropathic pain, in another preferred embodiment acute peripheral polyneuropathic pain or acute central polyneuropathic pain.
  • the pain is chronic neuropathic pain.
  • neuropathic pain is pain that originates from nerve damage or nerve malfunction. It becomes classified as chronic neuropathic pain when it is present for more than 3 months.
  • a further aspect of the invention relates to a method of treating nociceptive pain, preferably acute or chronic nociceptive pain, comprising the once daily, preferably oral administration of the pharmaceutical dosage form according to the invention to a subject in need thereof.
  • FIG. 1 shows the averaged numerical rating scale (NRS) values measured over a 24 hour period after administration of different single doses of the compound according to formula (I′b) (200, 400, 600 ⁇ g) compared to slow release morphine and placebo in patients with acute post-operative pain following orthopedic surgery (bunionectomy).
  • NRS numerical rating scale
  • FIG. 2 shows the release profile of the pharmacologically active agent according to general formula (I′b) from the pure solid itself (A), from solid formulations containing the active agent and a polymer (Kollidon 90; B) and from solid solutions containing the active agent, a polymer (Kollidon 90 or Kollidon VA 64) and a surfactant (Pluronic F68 or Tween 80; C-E) in hydrochloric acid (0.1 N).
  • the X-Axis refers to the time in minutes and the Y-Axis refers to the amount of dissolved active agent in percent in relation to the whole amount of active agent originally contained in the dosage form.
  • FIG. 3 shows the release profile of the pharmacologically active agent according to general formula (I′b) from the pure solid itself (A), from a solid formulation containing the active agent and a polymer (Kollidon VA 64; B) and from solid solutions containing the active agent, a polymer (Kollidon VA 64) and a surfactant (Tween 80; C) in hydrochloric acid (0.1 N).
  • the X-Axis refers to the time in minutes and the Y-Axis refers to the amount of dissolved active agent in percent in relation to the whole amount of active agent originally contained in the dosage form.
  • FIG. 4 shows the X-ray powder diffractogram of a solid solution containing the pharmacologically active agent according to general formula (I′b), polymer Kollidon VA64 and surfactant Tween 80 before being objected to a storage stability test.
  • FIG. 5 shows the X-ray powder diffractogram of a solid solution containing the pharmacologically active agent according to general formula (I′b), polymer Kollidon VA64 and surfactant Tween 80 after 4 weeks of storage at 25° C. and 60% relative humidity.
  • FIG. 6 shows the X-ray powder diffractogram of a solid solution containing the pharmacologically active agent according to general formula (I′b), polymer Kollidon VA64 and surfactant Tween 80 after 4 weeks of storage at 30° C. and 65% relative humidity.
  • FIG. 7 shows the average daily pain changes (change of NRS value) over a 5-day period after administration of daily doses of the compound according to formula (I′b) (40 ⁇ g, 120 ⁇ g) compared to placebo in patients with painful diabetic neuropathy.
  • FIG. 8 shows the average daily pain changes (change of NRS value) over a 5-day period after administration of daily doses of the compound according to formula (I′b) (80 ⁇ g, 200 ⁇ g) compared to placebo in patients with painful diabetic neuropathy.
  • FIG. 9 shows the average daily pain changes (change of NRS value) over a 5-day period after administration of daily doses of the compound according to formula (I′b) (100 ⁇ g) compared to placebo and morphine slow release (60 mg) in patients with painful diabetic neuropathy.
  • FIG. 10 shows the mean maximum plasma concentration of the compound according to formula (I′b) measured on the last day of a 5-day once daily dosing period in comparison to the plasma concentration measured 8 to 10 days later at the end of a wash-out phase.
  • a saturated solution of said pharmacologically active agent was made by suspending an appropriate amount in 5 g of the respective medium. After removing any trace of undissolved material by filtration through a sintered glass filter (0.45 ⁇ m and 0.22 ⁇ m), the remaining solution was concentrated under vacuum. The vacuum was maintained until all obvious traces of solvent had been removed, and the flask was then stored over night under high vacuum. The weight of the remaining compound was determined and the saturation concentration of (I′b) in the respective oil was calculated.
  • oils oleic acid, caprylic acid and capric acid were identified (solubility>20 mg/g solvent), whereas labrasol, solutol HS 15 and gelucire 50/13 were identified as suitable surfactants (solubility>10 mg/g solvent).
  • the saturation concentration of compound (I′b) was determined in two-component formulations of one oil (oleic acid, caprylic acid or capric acid) and one surfactant (labrasol, tween 60, solutol HS 15 and gelucire 50/13). Since these two-component formulations are solid at room-temperature, these solubility studies were conducted at 50° C. If these two-component formulations contain sufficient amounts of (I′b), they are also liquid at room temperature. A saturated solution of the two-component formulation is a liquid, as well.
  • the saturation concentration of (I′b) in said formulation was determined to be 1.3 mg/g.
  • phase behavior of the oil/surfactant mixtures according to example 1b was determined in presence of an aqueous medium.
  • 20 g of the respective two-component mixture was made by mixing the respective amounts of oil and surfactant. 0.5 g of the mixture was then added to 250 g of water or 250 g of gastric juice, respectively.
  • turbidity is an indication for the size of the droplets of an emulsion. Micro- or nanoemulsions appear clear to the naked eye and do not show turbidity, since the size of the droplets is too small for refracting visible light.
  • Emulsions which gave promising results in the previous study were then subjected to particle size analysis (using a laser-based particle size analyzer type Zetasizer NanoZS, Malvern Instruments) with a particle size measurement range of 0.02 to 2,000 ⁇ m. Particle size was calculated from the volume size distribution.
  • Labrasol-oleic acid 40 60 in 250 g H 2 O Mea- Peak 1 Peak 1 Peak 2 Peak 2 Peak 3 Peak 3 sure- diame- occurence diame- occurence diame- occurence ment ter [nm] [%] ter [nm] [%] ter [nm] [%] 1 326.2 3.1 100.2 96.9 0 0 2 169.8 100 0 0 0 0 3 2956 0.1 152 99.9 0 0 0 Labrasol-oleic acid 40:60 in 250 g gastric juice Mea- Peak 1 Peak 1 Peak 2 Peak 2 Peak 3 Peak 3 sure- diame- occurence diame- occurence diame- occurence ment ter [nm] [%] ter [nm] [%] ter [nm] [%] 1 242.6 100 0 0 0 0 2 243.8 100 0 0 0 0 3 265.8 100 0 0 0 0 0 Labrasol-ole
  • a formulation was formulated from the following composition:
  • SMEDDS self-microemulsifying drug delivery system
  • composition capsules of two dosages were prepared (50 ⁇ g and 400 ⁇ g):
  • the primary efficacy assessment endpoint was the absolute pain intensity over a 24 hour period. Pain intensity was measured using an 11-point numerical rating scale (NRS). At each time point, patients were instructed to evaluate their current pain intensity relative to an 11-point numerical rating scale. A score of zero represented no pain and a score of 10 represented worst possible pain. Missing scheduled pain assessments for the patients were imputed with the last observation carried forward (LOCF). The resulting averaged NRS values over the 24 hour period are depicted in FIG. 1 .
  • Tables 25 and 26 summarize the treatment emergent adverse events (TEAE(s)) experienced by the five treatment groups.
  • Clinical studies were conducted to determine the bioavailability of a liquid filled capsule formulation containing compound (I′b) in a dose strength of 400 ⁇ g compared to a hemicitrate oral solution of compound (I′b) (400 ⁇ g, 400 ⁇ g/mL oral solution) in a Macrogol 400 formulation after single oral administration.
  • 24 healthy white male subjects were included in a randomized, open-label, 3-way crossover, single-center clinical trial.
  • the main pharmacokinetic parameters were AUC 0-t , AUC 0-72h and C max .
  • the relative bioavailability of the 400 ⁇ g capsule and 400 ⁇ g/mL oral solution based on AUC 0-72h was 105%, with 90%-Cl within the 80% to 125% range conventionally used for assessing bioequivalence.
  • compositions are intended to be exemplary and it should be understood that the ingredients, the amount thereof and the procedure to obtain the dosage form may be varied.
  • hard gelatin capsules containing compound (I′b) in a dosage of, for instance, 40 or 400 ⁇ g can also be produced by means of the following SMEDDS formulations:
  • Active pharmaceutical ingredient i.e. the pharmacologically active agent according to general formula (I)
  • a polymer were dispersed in dichloromethane in a flask.
  • a surfactant was added.
  • the flask was heated and subjected to ultrasound in order to dissolve the ingredients properly.
  • the solution was filtered through a sintered glass filter in order to remove traces of undissolved material.
  • the solvent was evaporated by means of a rotary evaporator at a temperature of 60° C.
  • the solid residue was further dried under high vacuum overnight.
  • the dried material was transferred to a sealed glass vial for the analysis for amorphous contents by using XRPD and DSC.
  • the dissolution behavior was analyzed in 0.1N HCl.
  • FIG. 2 shows the release profile of the API.
  • the X-Axis refers to the time in minutes and the Y-Axis refers to the amount of dissolved API in percent in relation to the whole amount API originally contained in the dosage form.
  • FIG. 3 shows the release profile of the API.
  • the X-Axis refers to the time in minutes and the Y-Axis refers to the amount of dissolved API in percent in relation to the whole amount API originally contained in the dosage form.
  • the storage stability of a solid solution was tested under various conditions and analyzed by using XRPD.
  • the solid solution was prepared in accordance with examples 1 and 2 and had the following composition:
  • Unit doses of the blended formulation were filled into size 0 hard gelatin capsules.
  • the encapsulated formulations were stored at 25° C. and 60% relative humidity (RH) at 30° C. and 65% relative humidity, respectively. After predetermined time periods of 1, 2, and 4 weeks, the capsules were removed from the storage and analyzed.
  • the solid solution of the API especially the formulation with API:V64 in a ratio of 1:19, revealed to be physically stable over a time period of 4 weeks at 25° C. and 30° C. demonstrated via XRPD.
  • FIG. 4 shows the XRPD result of the API solid solution at time zero.
  • FIGS. 5 and 6 show the result after 4 weeks of storage at 25° C. and 60% relative humidity and at 30° C. and 65% relative humidity, respectively.
  • the drug product has amorphous or at least semi-amorphous nature.
  • Clinical studies were conducted to determine the analgesic efficacy and tolerability of single doses of the compound according to formula (I′b) (200 ⁇ g, 400 ⁇ g and 600 ⁇ g, based on the amount of the free base; hemicitrate oral solution of compound (I′b) in Macrogol 400) compared to that of morphine (60 mg, controlled-released form) and placebo in patients with acute post-operative pain following orthopedic surgery (bunionectomy).
  • the primary efficacy assessment endpoint was the absolute pain intensity over a 24 hour period. Pain intensity was measured using an 11-point numerical rating scale (NRS). At each time point, patients were instructed to evaluate their current pain intensity relative to an 11-point numerical rating scale. A score of zero represented no pain and a score of 10 represented worst possible pain. Missing scheduled pain assessments for the patients were imputed with the last observation carried forward (LOCF). The resulting averaged NRS values over the 24 hour period are depicted in FIG. 6 .
  • Tables 32 and 33 summarize the treatment emergent adverse events (TEAE(s)) experienced by the five treatment groups.
  • compositions are intended to be exemplary and it should be understood that the ingredients, the amount thereof and the procedure to obtain the dosage form may be varied.
  • compositions can be prepared in accordance with the general procedure:
  • ingredients of the following examples may be split up into intragranular and extragranular ingredients as well as ingredients from which a granulating solution is formed in order to process dosage forms by fluid bed granulation according to inventive example 1; or they may be processed by an alternative process, such as dry granulation or direct compression.
  • compositions were manufactured by fluid bed granulation according to the following compositions:
  • Avicel® PH101 and Avicel® PH102 are microcrystalline celluloses with different mean particle sizes (50 and 100 microns). Purified water was used as part of the granulating fluid, but was removed during the granulation process.
  • Clinical studies were conducted to determine the analgesic efficacy and tolerability of single doses of the compound according to formula (I′b) (200 ⁇ g, 400 ⁇ g and 600 ⁇ g, based on the content of the free base; hemicitrate oral solution of compound (I′b) in Macrogol 400) compared to that of morphine (60 mg, controlled-released form) and placebo in patients with acute post-operative pain following orthopedic surgery (bunionectomy).
  • the primary efficacy assessment endpoint was the absolute pain intensity over a 24 hour period. Pain intensity was measured using an 11-point numerical rating scale (NRS). At each time point, patients were instructed to evaluate their current pain intensity relative to an 11-point numerical rating scale. A score of zero represented no pain and a score of 10 represented worst possible pain. Missing scheduled pain assessments for the patients were imputed with the last observation carried forward (LOCF). The resulting averaged NRS values over the 24 hour period are depicted in FIG. 1 .
  • Tables 38 and 39 summarize the treatment emergent adverse events (TEAE(s)) experienced by the five treatment groups.
  • Clinical studies were conducted to determine the bioavailability of a tablet formulation containing compound (I′b) in a dose strength of 400 ⁇ g compared to a hemicitrate oral solution of compound (I′b) (400 ⁇ g, 400 ⁇ g/mL oral solution) in a Macrogol 400 formulation after single oral administration.
  • 24 healthy white male subjects were included in a randomized, open-label, 3-way crossover, single-center clinical trial.
  • the main pharmacokinetic parameters were AUC 0-t , AUC 0-72h and C max .
  • the relative bioavailability of the 400 ⁇ g tablet and 400 ⁇ g/mL oral solution based on AUC 0-72h was 108%, with 90%-Cl within the 80% to 125% range used for assessing bioequivalence.
  • the relative bioavailability of the 400 ⁇ g tablet and 400 ⁇ g/mL oral solution based on C max was also 108%, with 90%-Cl within the 80% to 125% range used for assessing bioequivalence.
  • Clinical studies were conducted to determine the analgesic efficacy and tolerability of multiple doses of compound (I′b) (40 ⁇ g, 80 ⁇ g, 100 ⁇ g, 120 ⁇ g and 200 ⁇ g; all dosages as weight equivalent dosages relative to the free base, in the form of the hemicitrate,) compared to that of morphine (60 mg, controlled-release) and placebo in patients with painful diabetic neuropathy.
  • the primary endpoint criterion was the reduction from baseline in average pain intensity score over the last 24 hours measured with the 11-point numerical rating scale (NRS) at the final treatment day of each treatment period, in comparison to placebo. Pain assessments were done 5 times a day starting at 7 h a.m. almost every 4 hours. At each time point, patients were instructed to evaluate their current pain intensity relative to the 11-point numerical rating scale via e-diary. A score of zero represented no pain and a score of 10 represented worst possible pain. Missing scheduled pain assessments for the patients were imputed with the last observation carried forward (LOCF). The resulting average daily pain changes (average change of the NRS value) over the 5-day treatment periods are depicted in FIGS. 7 to 9 .
  • Pain intensity changes from the period baseline and the overall baseline were analyzed using descriptive statistics and an analysis of covariance (ANCOVA) model with factors for treatment and site effects, treatment sequence and period effects, and baseline pain effects.
  • ANCOVA covariance
  • Table 48 summarizes the plasma concentrations that were measured at different points in time during the five days administration regimen:
  • FIG. 10 shows a comparison of the mean C max values measured on day 5 in comparison to the plasma concentration that was observed before the next dose was administered (C next predose ), i.e. 8 to 10 days after administration of the fifth dose at the end of the wash-out phase.
  • Tables 49 and 50 summarize the treatment emergent adverse events (TEAE(s)) experienced by the treatment groups.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Dispersion Chemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Neurosurgery (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US13/198,182 2010-08-04 2011-08-04 Pharmaceutical dosage forms comprising 6'-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine Abandoned US20120034297A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/198,182 US20120034297A1 (en) 2010-08-04 2011-08-04 Pharmaceutical dosage forms comprising 6'-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine
US14/579,401 US9289416B2 (en) 2010-08-04 2014-12-22 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
US15/245,626 US10912763B2 (en) 2010-08-04 2016-08-24 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US37063410P 2010-08-04 2010-08-04
US37064810P 2010-08-04 2010-08-04
US37064310P 2010-08-04 2010-08-04
EP10008115 2010-08-04
EP10008116.5 2010-08-04
EP10008117.3 2010-08-04
EP10008117 2010-08-04
EP10008115.7 2010-08-04
EP10008116 2010-08-04
US13/198,182 US20120034297A1 (en) 2010-08-04 2011-08-04 Pharmaceutical dosage forms comprising 6'-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/579,401 Division US9289416B2 (en) 2010-08-04 2014-12-22 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
US15/245,626 Continuation US10912763B2 (en) 2010-08-04 2016-08-24 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine

Publications (1)

Publication Number Publication Date
US20120034297A1 true US20120034297A1 (en) 2012-02-09

Family

ID=45556332

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/198,182 Abandoned US20120034297A1 (en) 2010-08-04 2011-08-04 Pharmaceutical dosage forms comprising 6'-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine
US14/579,401 Active US9289416B2 (en) 2010-08-04 2014-12-22 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
US15/245,626 Active US10912763B2 (en) 2010-08-04 2016-08-24 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/579,401 Active US9289416B2 (en) 2010-08-04 2014-12-22 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
US15/245,626 Active US10912763B2 (en) 2010-08-04 2016-08-24 Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine

Country Status (28)

Country Link
US (3) US20120034297A1 (ru)
EP (3) EP2600851B1 (ru)
JP (2) JP5792299B2 (ru)
KR (2) KR20130097173A (ru)
CN (2) CN103237545A (ru)
AU (2) AU2011287954B2 (ru)
BR (2) BR112013002734A2 (ru)
CA (2) CA2805525C (ru)
CL (2) CL2013000218A1 (ru)
CO (2) CO6670579A2 (ru)
DK (2) DK2600850T3 (ru)
EC (2) ECSP13012423A (ru)
ES (1) ES2675316T3 (ru)
HR (2) HRP20180904T1 (ru)
HU (1) HUE039328T2 (ru)
IL (2) IL224154B (ru)
LT (2) LT2600851T (ru)
MX (2) MX354686B (ru)
NZ (2) NZ605888A (ru)
PE (2) PE20131084A1 (ru)
PL (2) PL2600850T3 (ru)
PT (1) PT2600851T (ru)
RS (2) RS57344B1 (ru)
RU (2) RU2631481C2 (ru)
SI (2) SI2600851T1 (ru)
TR (1) TR201808244T4 (ru)
WO (2) WO2012016697A2 (ru)
ZA (1) ZA201300655B (ru)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130231381A1 (en) * 2011-07-08 2013-09-05 Gruenenthal Gmbh Crystalline (1r,4r)-6'-fluoro-N,N-dimethyl-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine
US8614245B2 (en) 2011-07-08 2013-12-24 Gruenenthal Gmbh Crystalline (1r,4r)-6′-fluoro-N,N-dimethyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
WO2018219847A1 (en) 2017-05-29 2018-12-06 Universita' Degli Studi Di Camerino Co-activators of mop and nop receptors in the treatment of addiction to drugs and/or psychostimulants

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2631481C2 (ru) 2010-08-04 2017-09-22 Грюненталь Гмбх ЛЕКАРСТВЕННАЯ ДОЗИРОВАННАЯ ФОРМА, КОТОРАЯ СОДЕРЖИТ 6'-ФТОР-(N-МЕТИЛ-ИЛИ N, N-ДИМЕТИЛ-)-4-ФЕНИЛ-4', 9'-ДИГИДРО-3' Н-СПИРО[ЦИКЛОГЕКСАН-1, 1'-ПИРАНО[3, 4, b] ИНДОЛ]-4-АМИН ДЛЯ ЛЕЧЕНИЯ НОЦИЦЕПТИВНОЙ БОЛИ
GB2514054A (en) 2011-09-29 2014-11-12 Gw Pharma Ltd A pharmaceutical composition comprising the phytocannabinoids cannabidivarin (CBDV) and cannabidiol (CBD)
US9320729B2 (en) 2012-05-18 2016-04-26 Gruenenthal Gmbh Pharmaceutical composition comprising (1r,4r)-6′-flouro-N,N-dimethyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano-[3,4,b]indol]-4-amine and a propionic acid derivative
US9320725B2 (en) 2012-05-18 2016-04-26 Gruenenthal Gmbh Pharmaceutical composition comprising (1r,4r)-6′-fluoro-N,N-dimethyl-4-phenyl-4′9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine and a gabapentinoid
US9345689B2 (en) * 2012-05-18 2016-05-24 Gruenenthal Gmbh Pharmaceutical composition comprising (1r,4r)-6′-fluoro-N, N-dimethyl-4-phenyl-4,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine and an anticonvulsant
US9308196B2 (en) * 2012-05-18 2016-04-12 Gruenenthal Gmbh Pharmaceutical composition comprising (1 r,4r) -6'-fluoro-N ,N-dimethyl-4-phenyl-4',9'-d ihydro-3'H-spiro[cyclohexane-1,1'-pyrano-[3,4,b]indol]-4-amine and an oxicam
US20130310435A1 (en) * 2012-05-18 2013-11-21 Gruenenthal Gmbh Pharmaceutical Composition Comprising (1r,4r)-6'-fluoro-N, N-dimethyl-4-phenyl-4,9' -dihydro-3'H-spiro[cyclohexane-1,1' -pyrano[3,4,b]indol]-4-amine and Paracetamol or Propacetamol
US9855286B2 (en) * 2012-05-18 2018-01-02 Gruenenthal Gmbh Pharmaceutical composition comprising (1r,4r)-6′-fluoro-N,N-di methyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano-[3,4,b]indol]-4-amine and a salicylic acid component
US20130310385A1 (en) * 2012-05-18 2013-11-21 Gruenenthal Gmbh Pharmaceutical Composition Comprising (1r,4r)-6'-fluoro-N,N-dimethyl-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine and Antidepressants
US8912226B2 (en) * 2012-05-18 2014-12-16 Gruenenthal Gmbh Pharmaceutical composition comprising (1r,4r) -6′-fluoro-N,N-dimethyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine and a NSAR
AU2013369649B2 (en) 2012-12-31 2018-07-26 Sunovion Pharmaceuticals Inc. Heterocyclic compounds and methods of use thereof
GB2530001B (en) 2014-06-17 2019-01-16 Gw Pharma Ltd Use of cannabidiol in the reduction of convulsive seizure frequency in treatment-resistant epilepsy
KR102354582B1 (ko) * 2014-08-12 2022-01-24 삼성전자 주식회사 전자 장치의 동작 방법 및 장치
GB2531278A (en) 2014-10-14 2016-04-20 Gw Pharma Ltd Use of cannabidiol in the treatment of intractable epilepsy
GB2531282A (en) 2014-10-14 2016-04-20 Gw Pharma Ltd Use of cannabinoids in the treatment of epilepsy
GB2531281A (en) 2014-10-14 2016-04-20 Gw Pharma Ltd Use of cannabidiol in the treatment of intractable epilepsy
AU2016208779B2 (en) 2015-01-23 2020-11-12 Grunenthal Gmbh Cebranopadol for treating pain in subjects with impaired hepatic and/or impaired renal function
CN106278994B (zh) * 2015-05-26 2019-06-21 复旦大学 螺环化合物及其制备方法和用途
GB2539472A (en) 2015-06-17 2016-12-21 Gw Res Ltd Use of cannabinoids in the treatment of epilepsy
GB2541191A (en) 2015-08-10 2017-02-15 Gw Pharma Ltd Use of cannabinoids in the treatment of epilepsy
WO2017148595A1 (en) 2016-02-29 2017-09-08 Grünenthal GmbH Titration of cebranopadol
GB2548873B (en) 2016-03-31 2020-12-02 Gw Res Ltd Use of Cannabidiol in the Treatment of SturgeWeber Syndrome
GB2551986A (en) 2016-07-01 2018-01-10 Gw Res Ltd Parenteral formulations
GB2551987A (en) 2016-07-01 2018-01-10 Gw Res Ltd Oral cannabinoid formulations
GB2553139A (en) 2016-08-25 2018-02-28 Gw Res Ltd Use of cannabinoids in the treatment of multiple myeloma
GB2557921A (en) 2016-12-16 2018-07-04 Gw Res Ltd Use of cannabinoids in the treatment of angelman syndrome
GB2559774B (en) 2017-02-17 2021-09-29 Gw Res Ltd Oral cannabinoid formulations
CN109134479A (zh) * 2017-06-27 2019-01-04 石家庄蒎格医药科技有限公司 结晶聚乙二醇纳洛酮草酸盐及制备方法
GB2569961B (en) * 2018-01-03 2021-12-22 Gw Res Ltd Pharmaceutical
GB2572125B (en) * 2018-01-03 2021-01-13 Gw Res Ltd Pharmaceutical
GB2584341B (en) 2019-05-31 2023-03-01 Gw Res Ltd Cannabinoid formulations
GB202002754D0 (en) 2020-02-27 2020-04-15 Gw Res Ltd Methods of treating tuberous sclerosis complex with cannabidiol and everolimus
US11160757B1 (en) 2020-10-12 2021-11-02 GW Research Limited pH dependent release coated microparticle cannabinoid formulations

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5356896A (en) * 1991-12-12 1994-10-18 Sandoz Ltd. Stabilized pharmaceutical compositions comprising an HMG-CoA reductase inhibitor compound
US20060004034A1 (en) * 2002-11-11 2006-01-05 Gruenenthal Gmbh Spirocyclic cyclohexane compounds

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL109944A (en) 1993-07-01 1998-12-06 Euro Celtique Sa Continuous release dosage form containing morphine and a method of preparing such sustained release unit dosage forms
FR2782006B1 (fr) 1998-08-07 2002-04-19 Gattefosse Ets Sa Composition a liberation prolongee apte a former une micro-emulsion
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
EP1195160B1 (en) 2000-10-05 2009-09-16 USV Ltd. Sustained release trimetazidine pharmaceutical compositions and a method of their preparation
KR100441167B1 (ko) 2001-12-27 2004-07-21 씨제이 주식회사 마이크로에멀젼 예비 농축액 조성물
AR038681A1 (es) 2002-02-14 2005-01-26 Solvay Pharm Bv Formulacion oral de solucion solida de una sustancia activa pobremente soluble en agua
SI1374906T1 (sl) 2002-06-17 2007-10-31 Chiesi Farma Spa Postopek za pripravo inkluzijskih spojin piroksakama: beta-ciklodekstrina
DE102005005446A1 (de) 2005-02-04 2006-08-10 Grünenthal GmbH Bruchfeste Darreichungsformen mit retardierter Freisetzung
US20070048228A1 (en) 2003-08-06 2007-03-01 Elisabeth Arkenau-Maric Abuse-proofed dosage form
WO2005062722A2 (en) 2003-11-21 2005-07-14 Sun Pharmaceutical Industries Limited Fexofenadine containing pharmaceutical formulation
DE10360793A1 (de) 2003-12-23 2005-07-28 Grünenthal GmbH Spirocyclische Cyclohexan-Derivate
EA012570B1 (ru) 2004-03-10 2009-10-30 Шеринг Акциенгезельшафт Твердая или полутвердая композиция, содержащая молекулярно диспергированный дроспиренон, и способ ее получения
WO2006063048A2 (en) * 2004-12-06 2006-06-15 Avigen, Inc. Ibudilast for treating neuropathic pain and associated syndromes
DE102006046745A1 (de) 2006-09-29 2008-04-03 Grünenthal GmbH Gemischte ORL1/µ-Agonisten zur Behandlung von Schmerz
KR101448050B1 (ko) * 2006-10-02 2014-10-14 랍테크 게젤샤프트 퓌르 테히놀로기슈 포르슝 운트 엔트빅클룽 엠베하 점막 비점착성 필름 제형
SI2260042T1 (sl) 2008-03-27 2011-11-30 Gruenenthal Gmbh Substituirani spirociklični cikloheksanski derivati
CN100560061C (zh) 2008-06-20 2009-11-18 海南锦瑞制药股份有限公司 一种氯诺昔康冻干粉针剂及其制备方法
CN103079548A (zh) 2010-08-04 2013-05-01 格吕伦塔尔有限公司 含有6’-氟-(N-甲基-或N,N-二甲基)-4-苯基-4’,9’-二氢-3’H-螺[环己烷-1,1’-吡喃并[3,4,b]吲哚]-4-胺的药物剂型
HUE025322T2 (en) 2010-08-04 2016-02-29 Gruenenthal Gmbh A pharmaceutical dosage form comprising 6'-fluoro- (N-methyl or N, N-dimethyl) -4-phenyl-4 ', 9'-dihydro-3'H-spiro [cyclohexane-1,1'-pyran [3, 4, b] indole] -4-amine
RU2631481C2 (ru) 2010-08-04 2017-09-22 Грюненталь Гмбх ЛЕКАРСТВЕННАЯ ДОЗИРОВАННАЯ ФОРМА, КОТОРАЯ СОДЕРЖИТ 6'-ФТОР-(N-МЕТИЛ-ИЛИ N, N-ДИМЕТИЛ-)-4-ФЕНИЛ-4', 9'-ДИГИДРО-3' Н-СПИРО[ЦИКЛОГЕКСАН-1, 1'-ПИРАНО[3, 4, b] ИНДОЛ]-4-АМИН ДЛЯ ЛЕЧЕНИЯ НОЦИЦЕПТИВНОЙ БОЛИ

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5356896A (en) * 1991-12-12 1994-10-18 Sandoz Ltd. Stabilized pharmaceutical compositions comprising an HMG-CoA reductase inhibitor compound
US20060004034A1 (en) * 2002-11-11 2006-01-05 Gruenenthal Gmbh Spirocyclic cyclohexane compounds

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Lee et al. (Arch Pharm Res Vol 28, No 1, 120-126, 2005). *
Mandawgadea et al. (International Journal of Pharmaceutics 362 (2008): 179-183). *
Sudheer et al. (Asian Journal of Pharmacy and Life Sciences; 2(2). April-June, 2012: pp.214-225) *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130231381A1 (en) * 2011-07-08 2013-09-05 Gruenenthal Gmbh Crystalline (1r,4r)-6'-fluoro-N,N-dimethyl-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine
US8614245B2 (en) 2011-07-08 2013-12-24 Gruenenthal Gmbh Crystalline (1r,4r)-6′-fluoro-N,N-dimethyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
US8618156B2 (en) * 2011-07-08 2013-12-31 Gruenenthal Gmbh Crystalline (1r,4r)-6'-fluoro-N,N-dimethyl-4-phenyl-4',9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine
US8765800B2 (en) 2011-07-08 2014-07-01 Gruenenthal Gmbh Crystalline (1r,4r)-6′-fluoro-N,N-dimethyl-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
WO2018219847A1 (en) 2017-05-29 2018-12-06 Universita' Degli Studi Di Camerino Co-activators of mop and nop receptors in the treatment of addiction to drugs and/or psychostimulants

Also Published As

Publication number Publication date
US20150111939A1 (en) 2015-04-23
WO2012016697A3 (en) 2012-08-16
AU2011287954A1 (en) 2013-01-31
RS58017B1 (sr) 2019-02-28
BR112013003677A2 (pt) 2016-09-06
RU2631481C2 (ru) 2017-09-22
ECSP13012422A (es) 2013-03-28
ZA201300655B (en) 2013-09-25
AU2011287955B2 (en) 2015-07-09
LT2600850T (lt) 2018-12-10
IL224154B (en) 2018-10-31
JP5792299B2 (ja) 2015-10-07
ECSP13012423A (es) 2013-03-28
AU2011287954B2 (en) 2015-07-09
DK2600851T3 (en) 2018-06-25
HRP20181856T1 (hr) 2018-12-28
PL2600851T3 (pl) 2018-08-31
NZ605888A (en) 2014-08-29
RS57344B1 (sr) 2018-08-31
SI2600851T1 (en) 2018-07-31
EP2600851A2 (en) 2013-06-12
US10912763B2 (en) 2021-02-09
SI2600850T1 (sl) 2018-12-31
DK2600850T3 (en) 2018-12-03
CN103260610A (zh) 2013-08-21
CL2013000225A1 (es) 2013-04-01
ES2675316T3 (es) 2018-07-10
JP2013532697A (ja) 2013-08-19
CA2805523C (en) 2018-06-12
IL224155A (en) 2017-05-29
RU2013109135A (ru) 2014-09-10
PE20131096A1 (es) 2013-10-10
JP5792298B2 (ja) 2015-10-07
TR201808244T4 (tr) 2018-07-23
MX2013001289A (es) 2013-03-22
MX350154B (es) 2017-08-29
KR20130097173A (ko) 2013-09-02
AU2011287955A1 (en) 2013-02-07
WO2012016697A2 (en) 2012-02-09
LT2600851T (lt) 2018-06-25
BR112013002734A2 (pt) 2016-06-07
NZ605887A (en) 2014-08-29
EP3459533A1 (en) 2019-03-27
PE20131084A1 (es) 2013-10-10
US9289416B2 (en) 2016-03-22
US20160361294A1 (en) 2016-12-15
MX2013001345A (es) 2013-03-22
CA2805525A1 (en) 2012-02-09
CO6670579A2 (es) 2013-05-15
PT2600851T (pt) 2018-06-19
CL2013000218A1 (es) 2013-03-15
CA2805523A1 (en) 2012-02-09
RU2013109137A (ru) 2014-09-10
CN103237545A (zh) 2013-08-07
HUE039328T2 (hu) 2018-12-28
CO6680626A2 (es) 2013-05-31
EP2600851B1 (en) 2018-05-02
KR20130137600A (ko) 2013-12-17
PL2600850T3 (pl) 2019-06-28
HRP20180904T1 (hr) 2018-08-24
JP2013532696A (ja) 2013-08-19
MX354686B (es) 2018-03-15
WO2012016698A3 (en) 2012-08-16
RU2638818C2 (ru) 2017-12-15
CA2805525C (en) 2018-10-09
EP2600850B1 (en) 2018-10-17
EP2600850A2 (en) 2013-06-12
WO2012016698A2 (en) 2012-02-09

Similar Documents

Publication Publication Date Title
US10912763B2 (en) Pharmaceutical dosage forms comprising 6′-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4′,9′-dihydro-3′H-spiro[cyclohexane-1,1′-pyrano[3,4,b]indol]-4-amine
DK2600846T3 (en) PHARMACEUTICAL DOSAGE FORM COMPRISING 6'-fluoro- (N-methyl or N, N-dimethyl -) - 4-phenyl-4 ', 9'-dihydro-3'H-spiro [cyclohexane-1,1'-pyrano [3 , 4, b] indole] -4-amine
EP2600838B1 (en) Pharmaceutical dosage form comprising 6'-fluoro-(n-methyl- or n,n-dimethyl-)-4-phenyl-4',9'-dihydro-3'h-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine
AU2011287956B2 (en) Pharmaceutical dosage form comprising 6'-fluoro-(N-methyl- or N,N-dimethyl-)-4-phenyl-4,9'-dihydro-3'H-spiro[cyclohexane-1,1'-pyrano[3,4,b]indol]-4-amine

Legal Events

Date Code Title Description
AS Assignment

Owner name: GRUENENTHAL GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GRUENING, NADJA;SCHILLER, MARC;HEMANI, ASHISH;AND OTHERS;SIGNING DATES FROM 20110905 TO 20110920;REEL/FRAME:027053/0058

AS Assignment

Owner name: GRUENENTHAL GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GRUENING, NADJA;SCHILLER, MARC;HEMANI, ASHISH;AND OTHERS;SIGNING DATES FROM 20120201 TO 20120210;REEL/FRAME:027796/0695

STCB Information on status: application discontinuation

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