US20160310480A1 - Therapeutical composition containing apomorphine as active ingredient - Google Patents

Therapeutical composition containing apomorphine as active ingredient Download PDF

Info

Publication number
US20160310480A1
US20160310480A1 US15/199,070 US201615199070A US2016310480A1 US 20160310480 A1 US20160310480 A1 US 20160310480A1 US 201615199070 A US201615199070 A US 201615199070A US 2016310480 A1 US2016310480 A1 US 2016310480A1
Authority
US
United States
Prior art keywords
apomorphine
pharmaceutical composition
solvent
test
antioxidant
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
US15/199,070
Inventor
Michael Dey
Joel Richard
Marie-Madeleine Baronnet
Nathalie Mondoly
Laurent Bertocchi
Jeremiah Harnett
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.)
Britannia Pharmaceuticals Ltd
Original Assignee
Britannia Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Britannia Pharmaceuticals Ltd filed Critical Britannia Pharmaceuticals Ltd
Priority to US15/199,070 priority Critical patent/US20160310480A1/en
Publication of US20160310480A1 publication Critical patent/US20160310480A1/en
Assigned to BRITANNIA PHARMACEUTICALS LTD. reassignment BRITANNIA PHARMACEUTICALS LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DEY, Michael, BARONNET, Marie-Madeleine, BERTOCCHI, LAURENT, HARNETT, JEREMIAH, RICHARD, JOEL, MONDOLY, NATHALIE
Assigned to BRITANNIA PHARMACEUTICALS LTD. reassignment BRITANNIA PHARMACEUTICALS LTD. CHANGE OF ADDRESS Assignors: BRITANNIA PHARMACEUTICALS LTD.
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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • 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
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs

Definitions

  • the present invention relates to a pharmaceutical composition in the form of a solution containing apomorphine as the active pharmaceutical ingredient, and more particularly a pharmaceutical formulation for parenteral administration.
  • Apomorphine is a pharmaceutical active ingredient which is used to reduce “off episodes” in patients with advanced Parkinson's disease.
  • Three solutions of apomorphine are currently marketed: a solution for subcutaneous infusion marketed under the trademark of APO-go® PFS, a solution for intermittent subcutaneous injection (bolus administration) marketed under the trademark of Apo-go® ampoules; and a solution of apomorphine for intermittent subcutaneous injection (bolus administration) marketed under the trademark of Apokyn®.
  • These three currently marketed solutions present a pH range of 3.0-4.0.
  • parenteral subcutaneous injection of an active pharmaceutical ingredient (API) formulated in acidic conditions may lead to in situ precipitation and chemical degradation under physiological conditions and pH.
  • Low pH value of the formulation, as well as potential precipitation and degradation of the drug under physiological conditions may induce local subcutaneous site reactions such as redness, itching, local indurations and nodules which are sore and troublesome. This is typically the case of apomorphine when administered in a subcutaneous (SC) infusion.
  • SC subcutaneous
  • the applicant has found a new formulation with a pH close to the physiological pH avoiding in situ precipitation, increasing chemical stability of the formulation and an increased drug concentration allowing the reduction of the injectable volume.
  • the object of the present invention is a pharmaceutical composition in the form of a solution comprising
  • pH of the composition is greater than 4.
  • co-solvent refers to a) a solvent or a mixture of solvents which allows the incorporation of the active ingredient in the composition to obtain the required dose in a suitable volume and matches the injectability criteria; or b) a water soluble solid which allows the incorporation of the active ingredient in the composition to obtain the required dose in a suitable volume and matches the injectability criteria.
  • antioxidant means a pharmaceutically acceptable compound having antioxidant properties in order to prevent oxidative degradation of the active substance and prevent oxidative degradation of the excipients.
  • surfactant refers to a compound or excipient with surface active properties, used mainly in the present formulations to improve the aqueous solubility of the active ingredient, help to protect the active substance against degradation and limit in vitro active ingredient precipitation if co-solvent alone is not sufficient.
  • pH modifier refers to a compound or excipient used mainly to adjust the pH of the formulation.
  • a composition according to the present invention comprises apomorphine as the active pharmaceutical ingredient (API) (or active substance).
  • the apomorphine API may be in the form of a salt or the free base.
  • apomorphine is in a salt form.
  • the salts of apomorphine which can be used for the invention are preferably pharmaceutically acceptable salts of organic acids, such as those of acetic, methanesulfonic, arylsulfonic, lactic, citric, tartaric, succinic, glutamic or ascorbic acid, or pharmaceutically acceptable salts of inorganic acids, such as those of hydrochloric, hydrobromic, phosphoric, sulphuric or nitric acid.
  • apomorphine is in the form of the free base.
  • apomorphine is in a salt form, preferably apomorphine is in the form of apomorphine hydrochloride.
  • the amount of apomorphine as the active ingredient is preferably between 10 to 70 mg/mL. In another preferred embodiment, the amount of apomorphine is between 10 to 65 mg/mL. In another preferred embodiment, the amount of apomorphine as the active ingredient is selected from 10, 20, 30, 40, 50 and 60 mg/mL.
  • a composition according to the present invention comprises a co-solvent which allows obtaining the required injectability criteria of the pharmaceutical composition.
  • the co-solvent is a solvent or a mixture of solvents or a water soluble solid.
  • the co-solvent is water-miscible (i.e. miscible at least at the concentration of 5% in water at 25° C.) or water-soluble, respectively. It may be selected for instance from an alcohol or a polyol such as diols, triols, mannitol, or a polyether, or a mixture thereof.
  • the co-solvent is an alcohol, it may be selected for instance from ethanol or isopropanol.
  • the co-solvent when the co-solvent is a polyol, it may be for instance a diol such as propylene glycol, or a triol such as glycerol, or may have more than 3 hydroxyl groups such as mannitol, maltitol or cyclodextrin derivatives such hydroxypropyl- ⁇ -cyclodextrin (HP ⁇ CD) or sulfobutyl- ⁇ -cyclodextrin (SB ⁇ CD).
  • the co-solvent when the co-solvent is a polyether, it may be selected for instance from polyoxyethylene glycols or polyoxyethylene glycol derivatives, such as polyoxyethylene glycol 400, Solutol® HS15 or Cremophor® ELP.
  • the co-solvent is selected from an alcohol or a polyol or a polyether or a mixture thereof, and more preferably from polyols and polyethers or a mixture thereof.
  • the co-solvent is selected from diols, triols, cyclodextrin derivatives, polyethylene glycols and polyethylene glycol derivatives or a mixture thereof.
  • the co-solvent is selected from propylene glycol, glycerol, hydroxypropyl- ⁇ -cyclodextrin (HP ⁇ CD), sulfobutyl- ⁇ -cyclodextrin (SB ⁇ CD), and polyethylene glycols derivatives or a mixture thereof.
  • the amount of co-solvent is between 0.1 to 80% (w:w) of the composition and more preferably between 0.2 to 70% (w:w).
  • a composition according to the present invention comprises also an antioxidant.
  • Said anti-oxidant is preferably soluble in the mixture consisting of apomorphine and co-solvent(s). It may be selected for instance from acids and their salts, vitamins and derivatives, amino acids, sulfites or free phenolic radical scavengers.
  • the antioxidant is an acid or a salt thereof, it may be selected for instance from ascorbic acid or its salts such as sodium ascorbate, isoascorbic acid or its salts such as sodium isoascorbate, citric acid or its salts such as sodium citrate, lactic acid or malic acid.
  • the antioxidant when the antioxidant is a vitamin or a vitamin derivative, it may be selected for instance from tocopherol (vitamin E), riboflavine (vitamin B2), tocopherol-PEG-succinate (vitamin derivative) or trolox (vitamin derivative).
  • the antioxidant when the antioxidant is an amino acid, it may be selected for instance from cysteine, tryptophane, histidine, selenocysteine, N-acetyl cysteine, taurine, glutathione or glutathione-glutathione.
  • the antioxidant when the antioxidant is a sulfite, it may be selected for instance from sodium sulfite or sodium metabisulfite.
  • the antioxidant when the antioxidant is a phenolic free radical scavenger, it may be selected for instance from butylated hydroxy toluene, butyl hydroxyl anisole or cinnamic acid.
  • the antioxidant is selected from acids and their salts, vitamins and vitamin derivatives, amino acids, sulfites or phenolic free radical scavengers, and preferably from acids and their salts or sulfites.
  • the antioxidant is selected from ascorbic acid or sodium metabisulfite, and is even more preferably ascorbic acid.
  • the antioxidant is ascorbic acid and the ratio (w:w) apomorphine/antioxidant is between 1:0.01 and 1:3.0, preferably between 1:0.03 and 1:2.0, and more preferably between 1:0.05 and 1:0.50.
  • the antioxidant is sodium metabisulfite and the ratio (w:w) apomorphine/antioxidant is between 1:0.01 and 1:0.50, preferably between 1:0.03 and 1:0.30, and more preferably between 1:0.09 and 1:0.11. In a more preferred embodiment the antioxidant is sodium metabisulfite and the ratio (w:w) apomorphine/antioxidant is 1:0.10.
  • a composition according to the present invention may contain other additives usually used in such pharmaceutical compositions such as surfactant or pH modifier.
  • a composition according to the present invention may contain a surfactant or a mixture thereof. It may be selected for instance from the family of polyoxyethylene sorbitan fatty esters or polyethylene glycol derivatives or poloxamers. When the surfactant is from the family of polyoxyethylene sorbitan fatty esters, it may be selected for instance from polyoxyethylene (80) sorbitan monooleate, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (40) sorbitan monopalmitate, polyoxyethylene (60) sorbitan monostearate.
  • the surfactant is a polyethylene glycol derivative
  • it may be selected for instance from polyethylene glycol fatty esters such as polyethylene glycol 660 12-hydroxystearate (Solutol® HS 15), or polyethylene glycol castor oil derivatives such as Cremophor® ELP.
  • a surfactant is optionally present in the composition.
  • a surfactant is present in the composition.
  • the surfactant present in the composition has a hydrophile-lipophile balance (HLB) between 8 and 20 and more preferably between 12 to 17.
  • HLB hydrophile-lipophile balance
  • the surfactant is present in a composition according to the present invention and the surfactant is selected from the family of polyoxyethylene sorbitan fatty esters or polyethylene glycol derivatives or poloxamers.
  • the surfactant is selected from polyoxyethylene (80) sorbitan monooleate, polyoxyethylene (20) sorbitan monolaurate, polyethylene glycol castor oil derivatives, and polyethylene glycol derivative and more preferably Cremophor® ELP, or polyethylene glycol 660 12-hydroxystearate (Solutol® HS 15).
  • the co-solvent is present in a composition according to the present invention and the apomorphine : co-solvent ratio (w:w) in the composition is between 1:0.1 to 1:40, and preferably between 1:0.2 and 1:30.
  • the pH of the composition is greater than 4. In a preferred embodiment, the pH of the composition is between 4 and 7, and preferably between 5 and 7. In a more preferred embodiment, the pH of the composition is between 5.5 and 6.5. In a more preferred embodiment, the pH of the composition is selected from 5.8, 5.9, 6.0, 6.1 or 6.2.
  • a pH modifier is optionally present in the composition.
  • a pH modifier is present in the composition.
  • the pH modifier may be selected for instance from sodium hydroxide, sodium bicarbonate, potassium hydroxide or magnesium hydroxide and, if necessary, acids like hydrochloric acid.
  • a pH modifier is present in the composition and it is selected from sodium hydroxide, sodium bicarbonate, potassium hydroxide or magnesium hydroxide.
  • a pharmaceutical composition according to the present invention is suitable to be administered by parenteral route.
  • the composition of the present invention is administered by subcutaneous route, and preferably by a subcutaneous infusion.
  • a pharmaceutical composition according to the present invention shows a low viscosity and is easily administered by the parenteral route through 27 Gauge (G) needle and more preferably through 29G needle.
  • a pharmaceutical composition according to the present invention may be useful in the treatment to reduce “off episodes” in patients with advanced Parkinson's disease.
  • composition of the present invention is for use as medicament intended to reduce “off episodes” in patients with advanced Parkinson's disease.
  • the present invention is further related to the use of a composition according to the invention as injection solution, in particular as injection solution in the treatment of Parkinson's disease.
  • the invention relates to a process for the preparation of a pharmaceutical composition as described above, said process being defined by the following steps with the order below:
  • the solution is preferably protected from the light.
  • Degassed water is previously obtained by circulating and bubbling dry nitrogen gas in the solution for at least 30 minutes, and the level of oxygen in the solution is monitored using an oxymeter.
  • the impurity content which was determined by the area-under-the-curve calculation, is a criteria used to evaluate the stability of the prototypes.
  • the degree of coloration was assayed using the Eur. Pharmacopoeia colour test.
  • the color grading (from 1 to 7) is inversely proportional to the colour intensity, i.e. 7 means a colourless aqueous solution, while 1 means a highly coloured solution.
  • PG Propylene glycol
  • PS 80 Polysorbate 80
  • HP ⁇ CD Hydroxypropyl ⁇ cyclodextrin
  • SM sodium metabisulfite
  • AA ascorbic acid
  • PK profiles of the selected prototypes were evaluated in rats. Six cannulated rats were used for each prototype. Animals received a subcutaneous injection of the formulation at a dose of 1 mg/kg at a first injection site, a subcutaneous injection of the corresponding vehicle in a second injection site and a subcutaneous injection of saline solution in a third injection site. The injected volumes range between 0.03 to 0.2 mL/kg depending on the concentration of the formulation (0.5 to 30 mg/mL). Blood samples were collected for a 3-hour period. Plasma concentrations were determined by HPLC-MS/MS and PK parameters were calculated.
  • tissues from the 3 injection sites were collected from each rat and examined by an anatomo-pathologist. A scoring from 0 to 4 (0: no effect, 1: minimal effect, 2: slight effect, 3: moderate effect, 4: severe effect) was attributed for each section and the tolerance was deduced.
  • PG Propylene glycol
  • PS 80 Polysorbate 80
  • SB ⁇ CD Sulfobutylether ⁇ cyclodextrin
  • HP ⁇ CD Hydroxypropyl ⁇ cyclodextrin
  • SM sodium metabisulfite
  • FIG. 1 shows the plasma profile of apomorphine over the first 14 minutes in order to obtain the PK parameters of the absorption phase.
  • FIG. 2 shows the overall plasma profile of apomorphine over a 3-hour period.
  • a formulation according to the present invention patients will be able to receive a better treatment based on a 6-fold lower injection volume, a formulation at a more physiologically acceptable pH and a formulation that tends to be more tolerated than the APO-go® reference.
  • PK profile of the selected prototypes were evaluated in rats.
  • Subcutaneous micropumps were implanted in 6 rats for each prototype.
  • Micropumps were implanted subcutaneously in the lumbar region for continuous infusion in the scapular region.
  • Animals received a subcutaneous infusion of the formulation at a dose of 6 mg/kg/day (0.3 mg/kg/hour) of the appropriate apomorphine formulations to animals over a 20 h period/day for 14 days. Blood samples were collected at the following time point:
  • Plasma concentrations were determined by HPLC-ESI-MS/MS method and PK parameters were calculated.
  • tissue from the injection site was collected from each rat and examined by an anatomo-pathologist. A scoring from 0 to 4 (0: no effect, 1: minimal effect, 2: slight effect, 3: moderate effect, 4: severe effect) was attributed for each section and the tolerance was deduced.
  • C maxss is the mean of all concentration values at day 7 (single sample time point).
  • C maxss is the mean of all concentration values at 20 h Post-SOI, i.e. at the end of infusion (EOI) (single sample time point).
  • the local tolerance data of the tested formulations in subcutaneous infusion over 20 h/day in rats at 6 mg/kg/d are presented in the Table 6.
  • the tolerance scoring was established at the 14 th day of infusion, i.e., at the EOI.
  • Test 1 Parameter (Reference) Test 2 Test 3 Test 4 Test 5 Test 6 Concentration 4.58 28.9 29.6 30.4 39.5 37.2 (mg/mL) pH 3.3 6.0 5.8 5.9 5.9 5.9 Tolerance scoring 3.0 2.8 2.3 2.7 2.4 2.6
  • Test 1 The same tolerance level than the Apo-Go ® reference (Test 1) was obtained with the test formulations that show a 6 to 8 times higher concentration.
  • a formulation according to the present invention patients will be able to receive a better treatment based on a 6 to 8 times lower injection volume, a formulation at a more physiologically acceptable pH and a formulation that tends to be more tolerated than the APO-go® reference.

Abstract

A pharmaceutical composition contains apomorphine as the active pharmaceutical ingredient, a water-miscible co-solvent, an antioxidant, and water. The solution has a pH greater than 4. The pharmaceutical composition is suitable for parenteral administration for the treatment of Parkinson's disease. The process for the manufacture of the pharmaceutical composition includes weighing the apomorphine and introducing it into a container with the co-solvent and the antioxidant under agitation until complete dissolution takes place.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional application of U.S. patent application Ser. No. 14/152,000, filed Jan. 10, 2014, which, in turn, is a continuation application of international patent application PCT/EP2012/002916, filed Jul. 11, 2012, designating the United States and claiming priority from European application 11290320.8, filed Jul. 11, 2011, and the entire content of the above applications is incorporated herein by reference.
  • FIELD OF THE DISCLOSURE
  • The present invention relates to a pharmaceutical composition in the form of a solution containing apomorphine as the active pharmaceutical ingredient, and more particularly a pharmaceutical formulation for parenteral administration.
  • BACKGROUND OF THE DISCLOSURE
  • Apomorphine is a pharmaceutical active ingredient which is used to reduce “off episodes” in patients with advanced Parkinson's disease. Three solutions of apomorphine are currently marketed: a solution for subcutaneous infusion marketed under the trademark of APO-go® PFS, a solution for intermittent subcutaneous injection (bolus administration) marketed under the trademark of Apo-go® ampoules; and a solution of apomorphine for intermittent subcutaneous injection (bolus administration) marketed under the trademark of Apokyn®. These three currently marketed solutions present a pH range of 3.0-4.0.
  • However, these formulations induce injection site reactions. Therefore the medical precaution for their infusion is to change the injection site every 12 hours and for the bolus administration is to rotate the injection site.
  • Furthermore, parenteral subcutaneous injection of an active pharmaceutical ingredient (API) formulated in acidic conditions may lead to in situ precipitation and chemical degradation under physiological conditions and pH. Low pH value of the formulation, as well as potential precipitation and degradation of the drug under physiological conditions, may induce local subcutaneous site reactions such as redness, itching, local indurations and nodules which are sore and troublesome. This is typically the case of apomorphine when administered in a subcutaneous (SC) infusion.
  • SUMMARY OF THE DISCLOSURE
  • To increase the local tolerance of the treatment, the applicant has found a new formulation with a pH close to the physiological pH avoiding in situ precipitation, increasing chemical stability of the formulation and an increased drug concentration allowing the reduction of the injectable volume.
  • The object of the present invention is a pharmaceutical composition in the form of a solution comprising
      • i) apomorphine as the active substance,
      • ii) a water miscible co-solvent,
      • iii) an anti-oxidant, in particular an anti-oxidant which is soluble in the mixture consisting of apomorphine and co-solvent, and
      • iv) water,
  • wherein the pH of the composition is greater than 4.
  • Unless otherwise indicated the following definitions are set forth to illustrate and define the meaning and scope of the various terms used to describe the invention herein.
  • The term “co-solvent” as used in the present application refers to a) a solvent or a mixture of solvents which allows the incorporation of the active ingredient in the composition to obtain the required dose in a suitable volume and matches the injectability criteria; or b) a water soluble solid which allows the incorporation of the active ingredient in the composition to obtain the required dose in a suitable volume and matches the injectability criteria.
  • The term “anti-oxidant” means a pharmaceutically acceptable compound having antioxidant properties in order to prevent oxidative degradation of the active substance and prevent oxidative degradation of the excipients.
  • The term “surfactant” as used herein refers to a compound or excipient with surface active properties, used mainly in the present formulations to improve the aqueous solubility of the active ingredient, help to protect the active substance against degradation and limit in vitro active ingredient precipitation if co-solvent alone is not sufficient.
  • The term “pH modifier” as used herein refers to a compound or excipient used mainly to adjust the pH of the formulation.
  • A composition according to the present invention comprises apomorphine as the active pharmaceutical ingredient (API) (or active substance). The apomorphine API may be in the form of a salt or the free base.
  • According to the present invention it is preferred that apomorphine is in a salt form. The salts of apomorphine which can be used for the invention are preferably pharmaceutically acceptable salts of organic acids, such as those of acetic, methanesulfonic, arylsulfonic, lactic, citric, tartaric, succinic, glutamic or ascorbic acid, or pharmaceutically acceptable salts of inorganic acids, such as those of hydrochloric, hydrobromic, phosphoric, sulphuric or nitric acid.
  • According to another preferred embodiment of the invention apomorphine is in the form of the free base.
  • According to another preferred embodiment of the invention apomorphine is in a salt form, preferably apomorphine is in the form of apomorphine hydrochloride.
  • In a pharmaceutical composition according to the present invention, the amount of apomorphine as the active ingredient is preferably between 10 to 70 mg/mL. In another preferred embodiment, the amount of apomorphine is between 10 to 65 mg/mL. In another preferred embodiment, the amount of apomorphine as the active ingredient is selected from 10, 20, 30, 40, 50 and 60 mg/mL.
  • A composition according to the present invention comprises a co-solvent which allows obtaining the required injectability criteria of the pharmaceutical composition. The co-solvent is a solvent or a mixture of solvents or a water soluble solid. The co-solvent is water-miscible (i.e. miscible at least at the concentration of 5% in water at 25° C.) or water-soluble, respectively. It may be selected for instance from an alcohol or a polyol such as diols, triols, mannitol, or a polyether, or a mixture thereof. When the co-solvent is an alcohol, it may be selected for instance from ethanol or isopropanol. When the co-solvent is a polyol, it may be for instance a diol such as propylene glycol, or a triol such as glycerol, or may have more than 3 hydroxyl groups such as mannitol, maltitol or cyclodextrin derivatives such hydroxypropyl-β-cyclodextrin (HPβCD) or sulfobutyl-β-cyclodextrin (SBβCD). When the co-solvent is a polyether, it may be selected for instance from polyoxyethylene glycols or polyoxyethylene glycol derivatives, such as polyoxyethylene glycol 400, Solutol® HS15 or Cremophor® ELP.
  • In a preferred embodiment, the co-solvent is selected from an alcohol or a polyol or a polyether or a mixture thereof, and more preferably from polyols and polyethers or a mixture thereof. In another preferred embodiment, the co-solvent is selected from diols, triols, cyclodextrin derivatives, polyethylene glycols and polyethylene glycol derivatives or a mixture thereof. In a more preferred embodiment, the co-solvent is selected from propylene glycol, glycerol, hydroxypropyl-β-cyclodextrin (HPβCD), sulfobutyl-β-cyclodextrin (SBβCD), and polyethylene glycols derivatives or a mixture thereof.
  • In a preferred embodiment, the amount of co-solvent is between 0.1 to 80% (w:w) of the composition and more preferably between 0.2 to 70% (w:w).
  • A composition according to the present invention comprises also an antioxidant. Said anti-oxidant is preferably soluble in the mixture consisting of apomorphine and co-solvent(s). It may be selected for instance from acids and their salts, vitamins and derivatives, amino acids, sulfites or free phenolic radical scavengers. When the antioxidant is an acid or a salt thereof, it may be selected for instance from ascorbic acid or its salts such as sodium ascorbate, isoascorbic acid or its salts such as sodium isoascorbate, citric acid or its salts such as sodium citrate, lactic acid or malic acid. When the antioxidant is a vitamin or a vitamin derivative, it may be selected for instance from tocopherol (vitamin E), riboflavine (vitamin B2), tocopherol-PEG-succinate (vitamin derivative) or trolox (vitamin derivative). When the antioxidant is an amino acid, it may be selected for instance from cysteine, tryptophane, histidine, selenocysteine, N-acetyl cysteine, taurine, glutathione or glutathione-glutathione. When the antioxidant is a sulfite, it may be selected for instance from sodium sulfite or sodium metabisulfite. When the antioxidant is a phenolic free radical scavenger, it may be selected for instance from butylated hydroxy toluene, butyl hydroxyl anisole or cinnamic acid.
  • In a preferred embodiment, the antioxidant is selected from acids and their salts, vitamins and vitamin derivatives, amino acids, sulfites or phenolic free radical scavengers, and preferably from acids and their salts or sulfites.
  • In a more preferred embodiment, the antioxidant is selected from ascorbic acid or sodium metabisulfite, and is even more preferably ascorbic acid.
  • In another preferred embodiment, the antioxidant is ascorbic acid and the ratio (w:w) apomorphine/antioxidant is between 1:0.01 and 1:3.0, preferably between 1:0.03 and 1:2.0, and more preferably between 1:0.05 and 1:0.50.
  • In another preferred embodiment, the antioxidant is sodium metabisulfite and the ratio (w:w) apomorphine/antioxidant is between 1:0.01 and 1:0.50, preferably between 1:0.03 and 1:0.30, and more preferably between 1:0.09 and 1:0.11. In a more preferred embodiment the antioxidant is sodium metabisulfite and the ratio (w:w) apomorphine/antioxidant is 1:0.10.
  • A composition according to the present invention may contain other additives usually used in such pharmaceutical compositions such as surfactant or pH modifier.
  • A composition according to the present invention may contain a surfactant or a mixture thereof. It may be selected for instance from the family of polyoxyethylene sorbitan fatty esters or polyethylene glycol derivatives or poloxamers. When the surfactant is from the family of polyoxyethylene sorbitan fatty esters, it may be selected for instance from polyoxyethylene (80) sorbitan monooleate, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (40) sorbitan monopalmitate, polyoxyethylene (60) sorbitan monostearate. When the surfactant is a polyethylene glycol derivative, it may be selected for instance from polyethylene glycol fatty esters such as polyethylene glycol 660 12-hydroxystearate (Solutol® HS 15), or polyethylene glycol castor oil derivatives such as Cremophor® ELP.
  • In a preferred embodiment, a surfactant is optionally present in the composition. In another preferred embodiment, a surfactant is present in the composition. In a further preferred embodiment, the surfactant present in the composition has a hydrophile-lipophile balance (HLB) between 8 and 20 and more preferably between 12 to 17.
  • In a preferred embodiment, the surfactant is present in a composition according to the present invention and the surfactant is selected from the family of polyoxyethylene sorbitan fatty esters or polyethylene glycol derivatives or poloxamers. In a more preferred embodiment, the surfactant is selected from polyoxyethylene (80) sorbitan monooleate, polyoxyethylene (20) sorbitan monolaurate, polyethylene glycol castor oil derivatives, and polyethylene glycol derivative and more preferably Cremophor® ELP, or polyethylene glycol 660 12-hydroxystearate (Solutol® HS 15).
  • In a preferred embodiment, the co-solvent is present in a composition according to the present invention and the apomorphine : co-solvent ratio (w:w) in the composition is between 1:0.1 to 1:40, and preferably between 1:0.2 and 1:30.
  • The pH of the composition is greater than 4. In a preferred embodiment, the pH of the composition is between 4 and 7, and preferably between 5 and 7. In a more preferred embodiment, the pH of the composition is between 5.5 and 6.5. In a more preferred embodiment, the pH of the composition is selected from 5.8, 5.9, 6.0, 6.1 or 6.2.
  • In a preferred embodiment, a pH modifier is optionally present in the composition. In another preferred embodiment, a pH modifier is present in the composition. The pH modifier may be selected for instance from sodium hydroxide, sodium bicarbonate, potassium hydroxide or magnesium hydroxide and, if necessary, acids like hydrochloric acid.
  • In a preferred embodiment, a pH modifier is present in the composition and it is selected from sodium hydroxide, sodium bicarbonate, potassium hydroxide or magnesium hydroxide.
  • A pharmaceutical composition according to the present invention is suitable to be administered by parenteral route. In a preferred embodiment, the composition of the present invention is administered by subcutaneous route, and preferably by a subcutaneous infusion.
  • A pharmaceutical composition according to the present invention shows a low viscosity and is easily administered by the parenteral route through 27 Gauge (G) needle and more preferably through 29G needle.
  • A pharmaceutical composition according to the present invention may be useful in the treatment to reduce “off episodes” in patients with advanced Parkinson's disease.
  • In a preferred embodiment, the composition of the present invention is for use as medicament intended to reduce “off episodes” in patients with advanced Parkinson's disease.
  • The present invention is further related to the use of a composition according to the invention as injection solution, in particular as injection solution in the treatment of Parkinson's disease.
  • Furthermore, the invention relates to a process for the preparation of a pharmaceutical composition as described above, said process being defined by the following steps with the order below:
      • the optional surfactant is weighed followed by the co-solvent in the same container;
      • the antioxidant is weighed (alone) and dissolved in degassed water;
      • the antioxidant solution is added to the co-solvent alone or to a mixture of co-solvent with surfactant under agitation;
      • a quantity of degassed water is added and then the solution is adjusted to around pH 7.5±0.5 and more preferably close to pH 7;
      • the apomorphine is weighed and introduced into the above solution and agitated until complete dissolution takes place;
      • the pH is controlled and adjusted if necessary according to the required pH values within the range of pH 4-7, and preferably 5-7, using a pH modifier;
      • finally, the remaining quantity of degassed water is added to reach the required volume.
  • The solution is preferably protected from the light. Degassed water is previously obtained by circulating and bubbling dry nitrogen gas in the solution for at least 30 minutes, and the level of oxygen in the solution is monitored using an oxymeter.
  • Unless otherwise indicated, all technical and scientific terms used herein have the same meaning as commonly understood by a specialist in the domain associated with this invention.
  • The following examples are presented to illustrate the above procedures and should not be considered as limiting the scope of the invention.
  • EXPERIMENTAL PART EXAMPLE 1 Preparation of Apomorphine Hydrochloride Solution at 40 mg/mL, Propylene Glycol (PG) (45%), Ascorbic Acid (AA) (0.4%), Surfactant Polysorbate (PS80) (0.3%), pH 6 100 mL of Apomorphine Hydrochloride Solution at 40 mg/mL is Manufactured According to the Following Process
      • 0.3 g of PS80 are weighed in a flask. 45.0 g of PG are added in the flask containing PS80;
  • in a beaker, 0.4 g of AA or 0.4 g of sodium ascorbate are weighed. 5 g of degassed water for injection (WFI) are added to AA or sodium ascorbate, and AA or sodium ascorbate is dissolved in degassed water under magnetic stirring;
      • the solution of ascorbic acid is added in the flask containing PG and PS80;
      • The beaker containing AA solution is rinsed with 5 g of degassed WFI. This rinsing WFI is added to the flask containing PG, PS80 and AA. The solution is homogenised under magnetic stirring.
      • 30 g of degassed WFI are added in the flask containing PS80, PG and AA and the solution is homogenised under magnetic stirring. The pH is then adjusted at 7.5±0.5 using a NaOH 1M solution (to be as close as possible to pH 7.0);
      • 4 g of apomorphine hydrochloride are weighed in a glass vial. Apomorphine hydrochloride is introduced in the flask containing the vehicle adjusted at pH 7.0. The glass vial containing apomorphine hydrochloride is rinsed with 3 g of degassed WFI. The rinsing WFI is added to apomorphine solution. Apomorphine hydrochloride is dissolved under magnetic stirring until complete dissolution of apomorphine hydrochloride.
      • pH of apomorphine solution is then adjusted at pH 6.0±0.2 using a NaOH 0.1 M solution;
      • Apomorphine hydrochloride solution is maintained under magnetic stirring during introduction of NaOH 0.1 M solution.
      • Degassed WFI is added to apomorphine hydrochloride solution in order to reach a final weight of 104.8 g of apomorphine hydrochloride solution.
      • Apomorphine hydrochloride solution is homogenised under magnetic stirring and the final pH is checked.
    EXAMPLE 2 Stability Study of Apomorphine Solutions at 30 mg/mL and 40 mg/mL
  • Apomorphine content and impurities were analysed using HPLC methods. The characteristics of the HPLC method used for the apomorphine content determination are as follows:
      • HPLC equipment: Alliance 2695 Waters
      • Column: YMC ODS-A 5 μm, 46×150 mm, 200 Å
      • Mobile phase: 750 mL buffer : 0.03 M K2HPO4 pH 3 250 mL methanol 0.75 g acid L-tartaric
      • Solvent for dilution: 0.1% sodium metabisulfite (SM) 0.1 M HCl Sparging with helium for at least 30 minutes
      • Flow rate: 1 mL/min
      • Detection: UV at 210 nm
      • Run time: 20 min
      • Temperature Column: ambient-Carousel : 5° C.
      • Standard solution: 0.05 mg/mL
      • Injection volume: 25 μL
      • Retention time: ˜8.5 min
  • The characteristics of the HPLC method used for the apomorphine impurities determination are as follows:
      • HPLC equipmentL Alliance 2695 Waters
      • Column: YMC ODS-A 5 μm, 46×150 mm, 200 Å
      • Mobile phase: 875 ml buffer : 0.01 M K2HPO4 pH 3 125 mL methanol 0.75 g L-tartaric acid
      • Solvent for dilution: 0.1% SM 0.1 M HCl Sparging with helium for at least 30 minutes
      • Flow rate: 1 mL/min
      • Detection: UV at 273 nm
      • Run time: 110 min
      • Temperature Column: ambient—Carousel : 5° C.
      • Standard solution: 0.0025 mg/ml
      • Injection volume: 20 μl
      • Retention time orthoquinone: ˜10 min impurity A2: ˜27 min impurity A1: ˜28 min apomorphine: ˜35 min
  • The impurity content, which was determined by the area-under-the-curve calculation, is a criteria used to evaluate the stability of the prototypes.
  • The degree of coloration was assayed using the Eur. Pharmacopoeia colour test.
  • It has to be noted that the color grading (from 1 to 7) is inversely proportional to the colour intensity, i.e. 7 means a colourless aqueous solution, while 1 means a highly coloured solution.
  • The materials and the preparations used for this test are colour reference solutions prepared according to Eur. Pharmacopoeia.
  • Each below formulation (1 to 6) and its supportive stability data are reported in the following Table 1.
      • Formulation 1: 30 mg/mL apomorphine, 35% PG, 0.2% PS80, 0.3% SM, pH 6.0.
      • Formulation 2: 30 mg/mL apomorphine, 35% PG, 0.2% PS 80, 0.3% AA, pH 6.0.
      • Formulation 3: 30 mg/mL apomorphine, 35% PG, 0.3% AA, pH 6.0.
      • Formulation 4: 40 mg/mL apomorphine, 45% PG, 0.4% AA, pH 6.0.
      • Formulation 5: 40 mg/mL apomorphine, 45% PG, 0.3% PS 80, 0.4% AA, pH 6.0.
      • Formulation 6: 30 mg/mL apomorphine, 20% HPIβCD, 0.2% PS80, 0.3% SM, pH 6.0.
  • (PG: Propylene glycol; PS 80: Polysorbate 80; HPβCD: Hydroxypropyl β cyclodextrin; SM: sodium metabisulfite; AA: ascorbic acid)
  • The column headings for the tables are the following:
      • pH;
      • degree of coloration noted as “colour;”
      • apomorphine content noted as “Est. content (mg/mL)”, since data were obtained without using an apomorphine reference standard; and,
      • total impurities noted as “Total imp. (%).”
  • TABLE 1
    Test
    pH Colour Est. Content (mg/mL) Total imp. (%)
    T 3 months T 3 months T 3 months T 3 months T 3 months T 3 months T 3 months T 3 months
    Formulation T0 (25° C.) (40° C.) T0 (25° C.) (40° C.) T0 (25° C.) (40° C.) t0 (25° C.) (40° C.)
    1 6.0 5.7 5.5 <B4 <B1 <JV2 29.9 29.0 28.9 <0.1 <0.1 <0.1
    2 6.1 6.0 6.0 <J4 <J3 <JB3 29.2 30.3 30.1 <0.1 <0.1 <0.1
    3 6.1 6.0 6.0 <J4 <J3 <JB3 30.4 30.2 30.1 <0.1 <0.1 <0.1
    4 6.0 6.1 6.1 <J4 <J3 <JB3 40.7 40.1 39.9 <0.1 <0.1 <0.1
    5 6.0 6.1 6.1 <J4 <JB3 <JB3 40.5 39.7 39.9 <0.1 <0.1 <0.1
    6 6.0 5.8 5.7 GY6 R3/RY3 <RY1 30.8 31.0 30.3 <0.1 0.2 0.8
  • EXAMPLE 3 In vivo Testing
  • Pharmacokinetic (PK) profiles of the selected prototypes were evaluated in rats. Six cannulated rats were used for each prototype. Animals received a subcutaneous injection of the formulation at a dose of 1 mg/kg at a first injection site, a subcutaneous injection of the corresponding vehicle in a second injection site and a subcutaneous injection of saline solution in a third injection site. The injected volumes range between 0.03 to 0.2 mL/kg depending on the concentration of the formulation (0.5 to 30 mg/mL). Blood samples were collected for a 3-hour period. Plasma concentrations were determined by HPLC-MS/MS and PK parameters were calculated.
  • In order to evaluate the local tolerance of the tested formulations, tissues from the 3 injection sites were collected from each rat and examined by an anatomo-pathologist. A scoring from 0 to 4 (0: no effect, 1: minimal effect, 2: slight effect, 3: moderate effect, 4: severe effect) was attributed for each section and the tolerance was deduced.
  • The formulations as follows were administered to rats (bolus injection):
      • Reference: Apo-go® PFS 5 mg/mL, pH 3.3
      • Test 1: 29.12 mg/mL apomorphine, 20% SBβCD, 0.3% SM, pH 5.9.
      • Test 2: 10 mg/mL apomorphine, 0.2% PS 80, 0.1% SM, pH 4.2.
      • Test 3: 29.36 mg/mL apomorphine, 0.2% PS 80, 30% PG, 0.3% SM, pH 6.0.
      • Test 4: 29.36 mg/mL apomorphine, 20% HPβCD, 0.2% PS 80, 0.3% SM, pH 5.9.
      • Test 5: 29.36 mg/mL apomorphine, 0.2% PS 80, 30% PG, 0.3% SM, pH 4.1.
  • (PG: Propylene glycol; PS 80: Polysorbate 80; SBβCD: Sulfobutylether β cyclodextrin; HPβCD: Hydroxypropyl β cyclodextrin; SM: sodium metabisulfite)
  • These selected prototypes were compared to the reference solution (APO-go®) regarding the PK profile and the local tolerance.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The invention will now be described with reference to the drawings wherein:
  • FIG. 1 (details of the absorption phase of tested formulations (1 mg/kg, SC in rats)) shows the plasma profile of apomorphine over the first 14 minutes in order to obtain the PK parameters of the absorption phase.
  • FIG. 2 (PK profiles of tested formulations (1 mg/kg, SC in rats)) shows the overall plasma profile of apomorphine over a 3-hour period.
  • DESCRIPTION OF THE BEST AND VARIOUS EMBODIMENTS
  • The PK parameters of these tested formulations in rats (1 mg/kg—SC) are presented in the Table 2.
  • TABLE 2
    Parameter APO-go ® Test 1 Test 2 Test 3 Test 4 Test 5
    Concentration 5.0 29.1 10.0 29.4 29.4 29.4
    (mg/mL)
    pH 3.3 5.9 4.2 6.0 5.9 4.1
    Cmax (ng/mL) 242 150 211 191 357 182
    Tmax (min) 5 11 4 4 5 9
    AUC (ng.h/mL) 104 98 99 85 136 113
    Bioavailability (%) Reference 94 95 82 130 108
    Cmax: maximum value of the apomorphine concentration in the rat serum.
    Tmax: time after administration for the apomorphine concentration to reach the Cmax value.
    AUC: area under the curve of the PK profile vs time.
  • The total apomorphine exposure for the above tested formulations was quite similar with regard to the reference, and no major PK differences can be observed for the five test formulations, which are significantly more concentrated than the reference APO-go® formulation. Consequently all of them show an acceptable profile from a PK point of view.
  • EXAMPLE 4 Local Tolerance
  • The local tolerance results of the tested formulations in rats (1 mg/kg—SC) are presented in the Table 3.
  • TABLE 3
    Parameter APO-go ® Test 1 Test 2 Test 3 Test 4 Test 5
    Concentration (mg/ml) 5.0 29.1 10.0 29.4 29.4 29.4
    pH 3.3 5.9 4.2 6.0 5.9 4.1
    Tolerance formulation 2.7 2.4 3.7 2.3 3.0 3.2
    scoring placebo 0.3 0.6 0.5 0.3 0.2 0.8
    Note:
    tolerance scoring for the saline solution was 0.1.
  • The same tolerance level as the Apo-go® reference is observed for the 5 test formulations, which have a concentration from 2 to 6 times higher than the reference (from 10 to 30 mg/mL for the test formulations vs. 5 mg/mL for the reference).
  • Thus, using a formulation according to the present invention, patients will be able to receive a better treatment based on a 6-fold lower injection volume, a formulation at a more physiologically acceptable pH and a formulation that tends to be more tolerated than the APO-go® reference.
  • EXAMPLE 5 14-Day Subcutaneous Infusion in Vivo Testing
  • PK profile of the selected prototypes were evaluated in rats. Subcutaneous micropumps were implanted in 6 rats for each prototype. Micropumps were implanted subcutaneously in the lumbar region for continuous infusion in the scapular region. Animals received a subcutaneous infusion of the formulation at a dose of 6 mg/kg/day (0.3 mg/kg/hour) of the appropriate apomorphine formulations to animals over a 20 h period/day for 14 days. Blood samples were collected at the following time point:
      • Day 1 at 0 h, 6 h post Start of Infusion and 20 h post Start of Infusion (SOI),
      • Day 7 20 h post Start of Infusion (SOI),
      • Day 14 at 0 h, 20 h post Start of Infusion (SOI).
  • Plasma concentrations were determined by HPLC-ESI-MS/MS method and PK parameters were calculated.
  • In order to evaluate the local tolerance of the tested formulations, tissue from the injection site was collected from each rat and examined by an anatomo-pathologist. A scoring from 0 to 4 (0: no effect, 1: minimal effect, 2: slight effect, 3: moderate effect, 4: severe effect) was attributed for each section and the tolerance was deduced.
  • The formulations as follows were administered to rats (infusion injection):
      • Test 1 (reference): Apo-go® PFS 5 mg/mL, pH 3.3
      • Test 2: 30 mg/mL apomorphine, 35% PG, 0.2% PS80, 0.3% SM, pH 6.0.
      • Test 3: 30 mg/mL apomorphine, 35% PG, 0.2% PS 80, 0.3% AA, pH 5.8.
      • Test 4: 30 mg/mL apomorphine, 35% PG, 0.3% AA, pH 5.9.
      • Test 5: 40 mg/mL apomorphine, 45% PG, 0.4% AA, pH 5.9.
      • Test 6: 40 mg/mL apomorphine, 45% PG, 0.3% PS 80, 0.4% AA, pH 5.9.
  • (PG: Propylene glycol; PS 80: Polysorbate 80; SM: sodium metabisulfite; AA: ascorbic acid)
  • These selected prototypes were compared to the reference solution (APO-go®, Test 1) regarding the PK profile and the local tolerance.
  • The PK parameters of these tested formulations in rats (6 mg/kg/day—SC continuous infusion) after day 1, 7 and 14 are presented in the Table 4.
  • TABLE 4
    Test 1
    Parameter (Reference) Test 2 Test 3 Test 4 Test 5 Test 6
    Concentration 4.58 28.9 29.6 30.4 39.5 37.2
    (mg/mL)
    pH 3.3 6.0 5.8 5.9 5.9 5.9
    Cmaxss (ng/ml) 12.95 17.16 13.43 12.19 13.51 10.02
    on day 1
    Cmaxss (ng/ml) 10.35 13.01 9.79 11.53 7.17 9.25
    on day 7
    Cmaxss (ng/ml) 15.21 9.68 8.73 7.30 2.76 9.54
    on day 14
    Cmaxss: is the maximum concentration of apomorphine in plasma at steady-state conditions. The value on day 1 was calculated as the mean of all concentration values obtained at 6 and 20 hours Post-start of infusion (SOI) (2 sample time points). At the 7th infusion day, Cmaxss is the mean of all concentration values at day 7 (single sample time point). At the 14th infusion day, Cmaxss is the mean of all concentration values at 20 h Post-SOI, i.e. at the end of infusion (EOI) (single sample time point).
  • Apomorphine exposure data of above tested formulations were similar with regard to the reference.
  • The PK parameters of these tested formulations in rats (6 mg/kg/day—SC continuous infusion) on day 14 are presented in the Table 5.
  • TABLE 5
    Test 1
    Parameter (Reference) Test 2 Test 3 Test 4 Test 5 Test 6
    Concentration 4.58 28.9 29.6 30.4 39.5 37.2
    (mg/mL)
    pH 3.3 6.0 5.8 5.9 5.9 5.9
    Cmaxss (ng/ml) 15.2 9.68 8.73 7.30 2.76 9.54
    on day 14
    AUC (ng · h/ml) 277 210 178 183 75 173
    Cmaxss corresponds to the mean concentration obtained after 20 h Post SOI
  • No major PK differences can be observed for the five test formulations. Consequently all of the test formulations show an acceptable profile from a PK point of view.
  • EXAMPLE 6 Local Tolerance in the 14-Day Subcutaneous Infusion Experiment
  • The local tolerance data of the tested formulations in subcutaneous infusion over 20 h/day in rats at 6 mg/kg/d are presented in the Table 6. The tolerance scoring was established at the 14th day of infusion, i.e., at the EOI.
  • TABLE 6
    Test 1
    Parameter (Reference) Test 2 Test 3 Test 4 Test 5 Test 6
    Concentration 4.58 28.9 29.6 30.4 39.5 37.2
    (mg/mL)
    pH 3.3 6.0 5.8 5.9 5.9 5.9
    Tolerance scoring 3.0 2.8 2.3 2.7 2.4 2.6
    The same tolerance level than the Apo-Go ® reference (Test 1) was obtained with the test formulations that show a 6 to 8 times higher concentration.
  • Thus, using a formulation according to the present invention, patients will be able to receive a better treatment based on a 6 to 8 times lower injection volume, a formulation at a more physiologically acceptable pH and a formulation that tends to be more tolerated than the APO-go® reference.
  • It is understood that the foregoing description is that of the preferred embodiments of the invention and that various changes and modifications may be made thereto without departing from the spirit and scope of the invention as defined in the appended claims.

Claims (17)

1-14. (canceled)
15. A method of administering a pharmaceutical composition to a patient in need thereof, the method comprising:
administering the pharmaceutical composition by parenteral route,
wherein the pharmaceutical composition includes:
i) apomorphine as an active substance;
ii) a water miscible co-solvent;
iii) an antioxidant; and
iv) water; and
wherein a pH of the pharmaceutical composition is greater than 4.
16. A method of reducing “off episodes” in a patient with advanced Parkinson's disease comprising administering the pharmaceutical composition according to claim 15.
17-18. (canceled)
19. The method according to claim 15, wherein an amount of the active substance is between about 10 to 70 mg/ml.
20. The method according to claim 15, wherein an amount of the water miscible co-solvent is between 0.1 to 80% (w:w) of the pharmaceutical composition.
21. The method according to claim 15, wherein the water miscible co-solvent is selected from the group consisting of an alcohol, a polyol, and a polyether or a mixture thereof.
22. The method according to claim 15, wherein the co-solvent is selected from a diol, a triol, a cyclodextrin derivative, a polyethylene glycol, and a polyethylene glycol derivative or a mixture thereof.
23. The method according to claim 15, wherein the antioxidant is selected from the group consisting of an acid, a salt thereof, a vitamin, a vitamin derivative, an amino acid, a sulphite, and a phenolic free radical scavenger.
24. The method according to claim 15, wherein the antioxidant is selected from the group consisting of ascorbic acid and sodium metabisulfite.
25. The method according to claim 15, wherein the pharmaceutical composition further comprises a surfactant.
26. The method according to claim 25, wherein the surfactant has a hydrophile-lipophile balance (HLB) between 8 and 20.
27. The method according to claim 25, wherein the surfactant is selected from the group consisting of a polyoxyethylene sorbitan fatty ester, a polyethylene glycol derivative, and a poloxamer.
28. The method according to claim 15, wherein an apomorphine: co-solvent ratio (w:w) is between 1:0.1 to 1:40.
29. The method according to claim 15, wherein the pH of the composition is between 4 and 7.
30. The method according to claim 15, wherein the composition further comprises a pH modifier.
31. The method according to claim 30, wherein the pH modifier is selected from the group consisting of sodium hydroxide, sodium bicarbonate, potassium hydroxide, and magnesium hydroxide or a mixture thereof.
US15/199,070 2011-07-11 2016-06-30 Therapeutical composition containing apomorphine as active ingredient Abandoned US20160310480A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/199,070 US20160310480A1 (en) 2011-07-11 2016-06-30 Therapeutical composition containing apomorphine as active ingredient

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP11290320A EP2545905A1 (en) 2011-07-11 2011-07-11 A new therapeutical composition containing apomorphine as active ingredient
EP11290320.8 2011-07-11
PCT/EP2012/002916 WO2013007381A1 (en) 2011-07-11 2012-07-11 A new therapeutical composition containing apomorphine as active ingredient
US14/152,000 US11026938B2 (en) 2011-07-11 2014-01-10 Therapeutical composition containing apomorphine as active ingredient
US15/199,070 US20160310480A1 (en) 2011-07-11 2016-06-30 Therapeutical composition containing apomorphine as active ingredient

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/152,000 Division US11026938B2 (en) 2011-07-11 2014-01-10 Therapeutical composition containing apomorphine as active ingredient

Publications (1)

Publication Number Publication Date
US20160310480A1 true US20160310480A1 (en) 2016-10-27

Family

ID=46514305

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/152,000 Active US11026938B2 (en) 2011-07-11 2014-01-10 Therapeutical composition containing apomorphine as active ingredient
US15/199,070 Abandoned US20160310480A1 (en) 2011-07-11 2016-06-30 Therapeutical composition containing apomorphine as active ingredient
US15/840,585 Abandoned US20180098981A1 (en) 2011-07-11 2017-12-13 Therapeutical composition containing apomorphine as active ingredient
US17/245,209 Active 2033-02-09 US11766431B2 (en) 2011-07-11 2021-04-30 Therapeutical composition containing apomorphine as active ingredient

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/152,000 Active US11026938B2 (en) 2011-07-11 2014-01-10 Therapeutical composition containing apomorphine as active ingredient

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/840,585 Abandoned US20180098981A1 (en) 2011-07-11 2017-12-13 Therapeutical composition containing apomorphine as active ingredient
US17/245,209 Active 2033-02-09 US11766431B2 (en) 2011-07-11 2021-04-30 Therapeutical composition containing apomorphine as active ingredient

Country Status (29)

Country Link
US (4) US11026938B2 (en)
EP (2) EP2545905A1 (en)
JP (2) JP6356064B2 (en)
KR (1) KR101990897B1 (en)
CN (2) CN108434094A (en)
AU (1) AU2012283381B2 (en)
BR (1) BR112014000306B1 (en)
CA (1) CA2841807C (en)
CL (1) CL2014000050A1 (en)
CO (1) CO6870038A2 (en)
CR (1) CR20140005A (en)
CU (1) CU24159B1 (en)
DO (1) DOP2014000003A (en)
EA (1) EA025870B1 (en)
EC (1) ECSP14013200A (en)
GE (1) GEP20196969B (en)
GT (1) GT201400003A (en)
IL (1) IL230308A (en)
IN (1) IN2014DN00090A (en)
MA (1) MA35411B1 (en)
MX (2) MX2014000454A (en)
MY (1) MY168212A (en)
NI (1) NI201400003A (en)
PE (1) PE20141539A1 (en)
SG (1) SG10201605491PA (en)
TN (1) TN2014000001A1 (en)
UA (1) UA113962C2 (en)
WO (1) WO2013007381A1 (en)
ZA (1) ZA201309711B (en)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2545905A1 (en) * 2011-07-11 2013-01-16 Britannia Pharmaceuticals Limited A new therapeutical composition containing apomorphine as active ingredient
CA2875446A1 (en) 2012-06-05 2013-12-12 Neuroderm Ltd. Compositions comprising apomorphine and organic acids and uses thereof
EP2918266A1 (en) 2014-03-11 2015-09-16 CDRD Berolina AB Composition comprising a dopamine agonist and an L-DOPA derivative for treating Parkinson's disease
EP2979688A1 (en) * 2014-08-01 2016-02-03 Britannia Pharmaceuticals Limited Composition containing Apomorphine and a Divalent Metal Cation
CA2971826A1 (en) * 2014-12-23 2016-06-30 Neuroderm Ltd Crystal forms of apomorphine and uses thereof
AU2016333486B2 (en) * 2015-09-28 2022-02-03 Ever Neuro Pharma Gmbh Aqueous composition of apomorphine for subcutaneous administration
CN114007617A (en) * 2019-05-31 2022-02-01 葡瑞慕生物技术有限公司 Unit dosage form for transmucosal administration of pharmaceutically active ingredients
JP2022543803A (en) * 2019-08-07 2022-10-14 アクリプス ワン インコーポレイテッド Pharmaceutical compositions of (6aS)-6-methyl-5,6,6a,7-tetrahydro-4H-dibenzo[de,g]quinoline-10,11-diol
EP4154885A1 (en) 2020-05-20 2023-03-29 MEDRx Co., Ltd. Apomorphine-containing percutaneous absorption type preparation
WO2022271537A1 (en) * 2021-06-25 2022-12-29 President And Fellows Of Harvard College Compositions and methods relating to injectable microemulsions
WO2023242355A1 (en) 2022-06-15 2023-12-21 Ever Neuro Pharma Gmbh Apomorphine prodrugs and uses thereof

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SU1662566A1 (en) * 1988-03-05 1991-07-15 Всесоюзный Научно-Исследовательский Институт Биотехнологии Method for stabilization of injectable apomorphine solution
JP2822049B2 (en) * 1989-02-02 1998-11-05 ゼリア新薬工業株式会社 Aqueous formulation composition
DK0865789T3 (en) * 1993-03-26 2005-07-18 Franciscus Wilhelmus He Merkus Pharmaceuticals for intranasal administration of dihydroergotamine
US6121276A (en) * 1994-04-22 2000-09-19 Pentech Pharmaceuticals, Inc. Apomorphine-containing dosage forms for ameliorating male erectile dysfunction
US5562917A (en) * 1994-12-23 1996-10-08 Pentech Pharmaceuticals, Inc. Transdermal administration of apomorphine
US5945117A (en) 1998-01-30 1999-08-31 Pentech Pharmaceuticals, Inc. Treatment of female sexual dysfunction
JP3723857B2 (en) * 1998-02-04 2005-12-07 日本ケミカルリサーチ株式会社 Aqueous pharmaceutical composition containing human growth hormone
US6011043A (en) 1998-06-19 2000-01-04 Schering Corporation Combination of phentolamine and apomorphine for the treatment of human sexual function and dysfunction
US6436950B1 (en) * 1998-08-14 2002-08-20 Nastech Pharmaceutical Company, Inc. Nasal delivery of apomorphine
US5994363A (en) * 1998-08-24 1999-11-30 Pentech Pharmaceuticals, Inc. Amelioration of apomorphine adverse effects
BR0005797A (en) * 2000-03-20 2001-10-16 Abbott Lab Methods for the treatment of sexual dysfunction with apomorphine at specified plasma concentration levels
US20030008878A1 (en) 2001-03-09 2003-01-09 Cowart Marlon D. Benzimidazoles that are useful in treating sexual dysfunction
US20020169166A1 (en) 2001-03-09 2002-11-14 Cowart Marlon D. Benzimidazoles that are useful in treating sexual dysfunction
ITMI20011321A1 (en) * 2001-06-22 2002-12-22 Chiesi Farma Spa MICROEMULSION PHARMACEUTICAL COMPOSITION SUITABLE FOR TRANSMINISTRIC ADMINISTRATION OF APOMORPHINE USEFUL IN TREATMENT OF
US20040028613A1 (en) * 2001-06-25 2004-02-12 Nastech Pharmaceutical Company Inc Dopamine agonist formulations for enhanced central nervous system delivery
PE20030762A1 (en) 2001-12-18 2003-09-05 Schering Corp HETEROCYCLIC COMPOUNDS AS NK1 ANTAGONISTS
US6939094B2 (en) 2003-01-28 2005-09-06 Macro Technologies Inc. Autonomous power interface for modifying limited rotation speed of a machine
CA2520121A1 (en) 2003-04-03 2004-10-21 Protein Design Labs, Inc. Inhibitors of integrin alpha5beta1 and their use for the control of tissue granulation
US20050090518A1 (en) * 2003-10-24 2005-04-28 Nastech Pharmaceutical Company Inc. Method for treating parkinson's disease using apomorphine and apomorphine prodrugs
WO2005074909A1 (en) * 2004-02-09 2005-08-18 Aska Pharmaceutical Co., Ltd. Combination drug
EP2032605A2 (en) 2006-05-24 2009-03-11 Bayer Schering Pharma Aktiengesellschaft HIGH AFFINITY HUMAN AND HUMANIZED ANTI-alpha5ß1 INTEGRIN FUNCTION BLOCKING ANTIBODIES WITH REDUCED IMMUNOGENICITY
US20080171072A1 (en) * 2006-08-11 2008-07-17 Frank Burczynski Ocular inserts containing apomorphine
US8741918B2 (en) 2007-06-21 2014-06-03 Veroscience Llc Parenteral formulations of dopamine agonists
EP2057982A1 (en) * 2007-11-09 2009-05-13 Archimedes Development Limited Intranasal compositions
CN101712675B (en) * 2008-10-07 2013-07-03 江苏恒谊药业有限公司 Nitrogenous benzheterocycle derivate and application thereof in treating nervous and mental diseases
EP2545905A1 (en) * 2011-07-11 2013-01-16 Britannia Pharmaceuticals Limited A new therapeutical composition containing apomorphine as active ingredient

Also Published As

Publication number Publication date
CO6870038A2 (en) 2014-02-20
IN2014DN00090A (en) 2015-05-15
AU2012283381A1 (en) 2014-02-06
UA113962C2 (en) 2017-04-10
CU20140005A7 (en) 2014-04-24
JP2014518285A (en) 2014-07-28
DOP2014000003A (en) 2014-06-01
EP2545905A1 (en) 2013-01-16
US11766431B2 (en) 2023-09-26
MX2021010046A (en) 2021-09-21
IL230308A (en) 2017-04-30
CN108434094A (en) 2018-08-24
KR101990897B1 (en) 2019-06-19
BR112014000306B1 (en) 2022-03-29
BR112014000306A2 (en) 2017-02-07
CN103826612A (en) 2014-05-28
MX2014000454A (en) 2014-09-01
SG10201605491PA (en) 2016-08-30
CU24159B1 (en) 2016-03-31
PE20141539A1 (en) 2014-10-25
EA025870B1 (en) 2017-02-28
CA2841807A1 (en) 2013-01-17
EP2731589A1 (en) 2014-05-21
JP2017081947A (en) 2017-05-18
GT201400003A (en) 2017-09-28
ECSP14013200A (en) 2014-04-30
CN103826612B (en) 2020-07-28
CR20140005A (en) 2014-07-21
CL2014000050A1 (en) 2014-08-18
NI201400003A (en) 2015-10-12
US20210244724A1 (en) 2021-08-12
MY168212A (en) 2018-10-15
KR20140072861A (en) 2014-06-13
EA201400067A1 (en) 2015-02-27
MA35411B1 (en) 2014-09-01
US20140128422A1 (en) 2014-05-08
ZA201309711B (en) 2014-08-27
TN2014000001A1 (en) 2015-07-01
WO2013007381A1 (en) 2013-01-17
US20180098981A1 (en) 2018-04-12
CA2841807C (en) 2018-02-20
GEP20196969B (en) 2019-04-25
AU2012283381B2 (en) 2017-07-27
JP6356064B2 (en) 2018-07-11
US11026938B2 (en) 2021-06-08

Similar Documents

Publication Publication Date Title
US11766431B2 (en) Therapeutical composition containing apomorphine as active ingredient
KR101593579B1 (en) Therapeutic compositions
CN108135980B (en) Method for preparing stable glucagon therapeutic agent in non-proton polar solvent
RU2345772C2 (en) Lyophilised compositions cci-779
US20100267818A1 (en) Stabilized single-liquid pharmaceutical composition containing docetaxel
EA012522B1 (en) Compositions for delivering highly water soluble drugs (embodiments) and use thereof for the treatment of malignant tumor
WO2019136817A1 (en) Cabazitaxel composition for injection and preparation method therefor
EP1946747A1 (en) Pharmaceutical composition of improved stability containing taxane derivatives
EP2900216B1 (en) Stabilized aqueous compositions of neuromuscular blocking agents
KR20150112975A (en) Stable pharmaceutical composition of clopidogrel free base for oral and parenteral delivery
NZ620162A (en) A new therapeutical composition containing apomorphine as active ingredient
NZ620162B2 (en) A new therapeutical composition containing apomorphine as active ingredient
BR112019016545A2 (en) COMPOSITION FOR INTRAVESICAL ADMINISTRATION FOR TREATMENT OF PAIN IN THE BLADDER
CA3220425A1 (en) Pharmaceutical composition, and aprepitant injection and freeze-dried powder injection
JP2010502565A (en) Lyophilized formulation
WO2024075135A1 (en) &#34;stable injectable compositions of glp-2 peptide&#34;
US20160120742A1 (en) Compositions including cabazitaxel
CN116350619A (en) Oral taxane pharmaceutical composition
KR20190107987A (en) Stabilized Injectable Solution Composition Containing Polmacoxib and Preparation Method Thereof
WO2014108791A1 (en) Injectable composition containing chlorothiazide

Legal Events

Date Code Title Description
AS Assignment

Owner name: BRITANNIA PHARMACEUTICALS LTD., UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DEY, MICHAEL;RICHARD, JOEL;BARONNET, MARIE-MADELEINE;AND OTHERS;SIGNING DATES FROM 20140127 TO 20140226;REEL/FRAME:041044/0693

AS Assignment

Owner name: BRITANNIA PHARMACEUTICALS LTD., UNITED KINGDOM

Free format text: CHANGE OF ADDRESS;ASSIGNOR:BRITANNIA PHARMACEUTICALS LTD.;REEL/FRAME:041496/0180

Effective date: 20170126

STCB Information on status: application discontinuation

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