专利摘要:
Disclosed are formulations of rasagiline base which are designed to delay the release of rasagiline while retaining specific pharmacokinetic properties. Also disclosed are rasagiline citrate salt and its use and method of preparation thereof.
公开号:AT12508U1
申请号:TGM76/2011U
申请日:2010-01-22
公开日:2012-06-15
发明作者:Daniella Licht;Rachel Cohen;Anton Frenkel;Marina Zholkovsky;Tamas Koltai;Muhammad Safadi
申请人:Teva Pharma;
IPC主号:
专利说明:

Austrian Patent Office AT12 508U1 2012-06-15
description
RASAGILINE FORMULATION WITH DELAYED RELEASE BACKGROUND OF THE INVENTION
[0001] US Patents 5,532,415, 5,387,612, 5,453,446, 5,457,133, 5,599,991, 5,744,500, 5,891,923, 5,668,181,5,576,353, 5,519,061, 5,786,390, 6,316,504, 6,630,514 disclose R (+) - N-propargyl-1-aminoindan (" R- " PAI "), also known as Rasagiline. Rasagiline has been reported to be a selective inhibitor of the B form of the enzyme monoamine oxidase ("MAO-B") and is useful in the treatment of Parkinson's disease and various other conditions by inhibiting MAO-B in the brain ,
U.S. Patent No. 6,126,968 discloses pharmaceutical formulations containing rasagiline. A formulation of rasagiline mesylate is approved for the treatment of Parkinson's disease either as monotherapy or as an adjunct to other treatments. See, for example, AZILECT®, Physicians' Desk Reference 2009 (PRD, 63rd Edition).
AZILECT® is a commercially available rapid release rasagiline mesylate formulation indicated for the treatment of the signs and symptoms of idiopathic Parkinson's disease as an initial monotherapy and as an adjunctive therapy for levodopa. The currently marketed formulation of Rasagiline (AZILECT®) is rapidly absorbed, reaching the maximum plasma concentration (tmax) in about one hour. The absolute bioavailability of rasagiline is about 36%. (AZILECT® product label, May 2006).
However, there are some concerns associated with the commercially available form of rasagiline mesylate. A concern with using monoamine oxidase inhibitors (" MAO "), for example, is the risk of hypertensive crises, often referred to as the " Cheese Effect " referred to as. (Simpson, GM and White K. " Tyramine studies and the sat-fety of MAOI drugs. &Quot; J Clin Psychiatry, 1984 July; 45 (7 part 2): 59-91.) This effect is mediated by inhibition of peripheral MAO caused. A high concentration of peripheral MAO is found in the stomach.
Another concern in patients with Parkinson's disease is that many patients suffer from delayed gastric emptying (Pfeiffer, RF and Quigley, EMM Gastrointestinal motility problems in patients with Parkinson's disease: Epidemiology, pathophysiology, and guidelines for Management, " CNS Drugs, 1999, 11 (6): 435-448; Jost, WH; " Gastrointestinal motility problems in patients with Parkinson's disease: Effects of antiparkinsonian treatment and guidelines for management ", Drugs and Aging, 1997, 10 (4): 249-258). Delayed gastric emptying (prolonged gastric residence time) may be a cause of increased inhibition of peripheral MAO and may contribute to the Cheese effect.
There is also a concern regarding the potential for the formation of alkyl mesylates during the treatment of the free base of a drug substance with MSA, if any residues of short chain alcohols are present. (Snodin D., "Residues of genotoxic alkyl mesylates in mesylate salt drug substances:" Real or imaginary problems "" Regulatory Toxicology and Pharmacology, Vol. 45, 2006, pp. 79-90).
Efforts to overcome such concerns and to improve the commercially available form of rasagiline mesylate are described in the literature. For example, International Application Publication No. WO 2006/057912 describes orally disintegrating rasagiline compositions; International Application Publication No. WO 2006/014973 discloses delayed release rasagiline compositions; International Application Publication No. WO 2008/076348 discloses a crystalline solid form of rasagiline base; International Application Publication No. WO 2008/076315 discloses the tannate salt of rasagiline. Other efforts to achieve certain improvements are described in International Application Publication No. WO 2008/019871 and International Application Publication No. WO 2008/131961.
However, the previous efforts do not disclose formulations as described herein, especially formulations using the solid Rasagiline base described herein. Also, the past efforts do not disclose the citric acid salt of rasagiline or the benefits of formulations utilizing the citric acid salt of rasagiline as described herein.
SUMMARY OF THE INVENTION
The present invention provides a stable oral dosage form containing a core having a form of rasagiline resulting from a manufacturing process and at least one pharmaceutically acceptable excipient; and an acid resistant, pharmaceutically acceptable coating, wherein the manufacturing process comprises a) preparing the core by mixing rasagiline base, citric acid and / or
Malic acid, and a pharmaceutically acceptable excipient; and b) coating the core with the acid-resistant pharmaceutically acceptable coating.
[0012] The present invention also provides a stable oral dosage form containing a core, rasagiline base, rasagiline citrate, rasagiline malate, or a mixture of at least two of rasagiline base, rasagiline citrate, and rasagiline. Malate, and at least one pharmaceutically acceptable excipient; and an acid resistant, pharmaceutically acceptable coating.
The present invention further provides a method of treating a patient suffering from Parkinson's disease comprising administering to the patient the dosage form described herein.
The present invention still further provides rasagiline citrate.
The present invention still further provides a composition containing the rasagiline citrate and a carrier described herein.
The present invention still further provides a method of making the rasagiline citrate or composition described herein, comprising: a) combining a citric acid solution with rasagiline base to form a first mixture; B) adding a solvent to the first mixture to form a second mixture; C) complete removal of liquid from the second mixture; and [0020] d) recovering the rasagiline citrate or preparing the composition.
The present invention still further provides a process for preparing the composition described herein, comprising: a) obtaining rosaniline citrate in isolated form; and b) mixing the rasagiline citrate with a carrier.
The present invention still further provides a method for treating a human suffering from Parkinson's disease (PD), brain ischemia, stroke, traumatic brain injury, spinal cord injury, neurotrauma, neurodegenerative disease, neurotoxic injury, nerve injury , Dementia, Alzheimer's dementia, senile dementia, depressive disorder, memory disorders, hyperactivity disorder, attention deficit disorder, multiple sclerosis (MS), AT 12 508 Ul 2012-06-15 Schizophrenia, affective disorder, amyotrophic lateral sclerosis; Restless legs syndrome (RLS); hearing loss; Multisystem atrophy (MSA), glaucoma, glucoma, modifying Parkinson's disease, and progressive vision palsy (PSP), comprising administering to the human an amount of the rasagiline citrate or composition described herein which is effective in treating humans.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a stable oral dosage form comprising a core having a form of rasagiline resulting from a manufacturing process and at least one pharmaceutically acceptable excipient; and an acid resistant, pharmaceutically acceptable coating, wherein the manufacturing process comprises a) preparing the core by mixing rasagiline base, citric acid and / or
Malic acid, and a pharmaceutically acceptable excipient; and b) coating the core with the acid-resistant pharmaceutically acceptable coating.
In another embodiment of the dosage form, step a) of the process comprises preparing wet granules of rasagiline base, citric acid and / or malic acid, and a pharmaceutically acceptable excipient.
In a further embodiment of the dosage form, step a) of the process further comprises: i) drying the wet granules to form a dry granulate; ii) disintegrating the dry granules to form particles; and [0032] iii) Mixing the particles with at least one lubricant.
In another embodiment of the dosage form, in step iii) of the method, the lubricant is talc or stearic acid or a combination thereof.
In a further embodiment of the dosage form, in step i) of the process, the wet granules are dried in a fluid bed dryer at an air inlet temperature of 40 ° to 50 ° C, and at an air outlet temperature of not higher than 37 ° C.
In a further embodiment of the dosage form, in step i) of the process, the air inlet temperature is 45 ° C.
In a further embodiment of the dosage form, in step ii) of the process, the dry granules are comminuted by an oscillating granulator.
In another embodiment of the dosage form, step a) of the method further comprises a step of forming the core by compression.
In another embodiment of the dosage form, in step a) of the method, the core is prepared by mixing rasagiline base, citric acid and a pharmaceutically acceptable excipient.
In another embodiment of the dosage form, in step a) of the method, the core is prepared by mixing rasagiline base, malic acid and a pharmaceutically acceptable excipient.
In another embodiment of the dosage form, in step a) of the method, the core is prepared by mixing rasagiline base, citric acid and malic acid, and a pharmaceutically acceptable excipient.
The present invention also provides a stable oral dosage form containing a core, rasagiline base, rasagiline citrate, rasagiline malate, or a mixture at least two of rasagiline base, rasagiline citrate and rasagiline malate, and at least one pharmaceutically acceptable excipient; and an acid resistant pharmaceutically acceptable coating.
In one embodiment of the dosage form, the rasagiline base described herein is crystalline rasagiline base.
In one embodiment of the dosage form, the dosage form contains a core comprising rasagiline citrate and at least one pharmaceutically acceptable excipient; and an acid resistant, pharmaceutically acceptable coating.
In a further embodiment of the dosage form, the dosage form contains a core comprising rasagiline malate and at least one pharmaceutically acceptable excipient; and an acid resistant, pharmaceutically acceptable coating.
In yet another embodiment of the dosage form, the dosage form, when taken by a human, provides an AUC value of rasagiline that is 80-130% that of the corresponding amount of rasagiline, which is more rapid as a formulation Release is recorded.
In yet another embodiment of the dosage form, the dosage form, when administered to a human, provides an AUC of rasagiline that is 80-125% that of the corresponding amount of rasagiline, which is more rapid as a formulation Release is recorded.
In yet another embodiment of the dosage form described herein, the dosage form, when ingested by a human in a saturated state, provides an AUC value of rasagiline greater than that of the corresponding amount of rasagiline, which a rapid release formulation is included.
In yet another embodiment of the dosage form described herein, the dosage form, when taken by a human, provides a Cmax of rasagiline that is 80-145% that of the corresponding amount of rasagiline, which is more rapid as a formulation Release is recorded.
In yet another embodiment of the dosage form described herein, the dosage form, when taken by a human, provides a Cmax of rasagiline that is 80-125% of that of the corresponding dosage of rasagiline, which is faster than a formulation Release is recorded.
In yet another embodiment of the dosage form described herein, the dosage form, when ingested by a human in a saturated state, provides a Cmax of rasagiline greater than that of the corresponding amount of rasagiline used as a formulation rapid release is recorded.
In yet another embodiment of the dosage form, the core further contains at least one antioxidant.
In yet another embodiment of the dosage form, the antioxidant is citric acid.
In yet another embodiment of the dosage form, the antioxidant is malic acid.
In yet another embodiment of the dosage form, the antioxidant is citric acid and malic acid.
In yet another embodiment of the dosage form described herein, the core is in the form of a tablet.
In yet another embodiment of the dosage form, the core further contains at least one disintegrant or disintegrant.
In yet another embodiment of the dosage form, the disintegrants are present in the core in an amount between 0.5% and 20% by weight.
In yet another embodiment of the dosage form, the disintegrant is pregelatinized starch.
In yet another embodiment of the dosage form described herein, the acid-resistant coating layer comprises methacrylic acid-ethyl acrylate copolymer (1: 1) and a plasticizer.
In yet another embodiment of the dosage form, the ratio of methacrylic acid-ethyl acrylate copolymer (1: 1) to plasticizer in the acid-resistant coating layer is between 10 to 1 and 2 to 1.
In yet another embodiment of the dosage form, the ratio of methacrylic acid-ethyl acrylate copolymer (1: 1) to plasticizer in the coating is about 5 to 1.
In yet another embodiment of the dosage form, the plasticizer is triethyl citrate.
In yet another embodiment of the dosage form, the acid-resistant coating further contains talc.
In yet another embodiment of the dosage form, the acid resistant coating is between 3% and 12% by weight of the dosage form.
In yet another embodiment of the dosage form, the acid resistant coating is about 8% by weight of the dosage form.
In yet another embodiment of the dosage form, the acid-resistant coating contains two coating layers.
In yet another embodiment of the dosage form, the interior of the two coating layers contains hypromellose.
In yet another embodiment of the dosage form, the dosage form has a weight of less than 150 mg.
In yet another embodiment of the dosage form, in addition to the rasagiline base and citric and / or malic acid, the dosage form contains mannitol, colloidal silica, starch NF, pregelatinized starch, stearic acid, talc, hypromellose, methacrylic acid-ethyl acrylate copolymer, talc extra fine and triethyl citrate.
In yet another embodiment of the dosage form described herein, the content of rasagiline citrate is from 0.74 mg to 3.63 mg.
In yet another embodiment of the dosage form, in addition to the rasagiline citrate, the dosage form contains mannitol, colloidal silica, starch NF, pregelatinized starch, stearic acid, talc, hypromellose, methacrylic acid-ethyl acrylate copolymer, talc extrafine and triethyl citrate.
In yet another embodiment of the dosage form described herein, the content of rasagiline malate is 0.66 mg to 3.05 mg.
In yet another embodiment of the dosage form, in addition to the rasagiline malate, the dosage form contains mannitol, colloidal silica, starch NF, pregelatinized starch, stearic acid, talc, hypromellose, methacrylic acid-ethyl acrylate copolymer, talc extrafine and triethyl citrate.
In yet another embodiment of the dosage form described herein, the rasagiline content is 1.0 mg. In yet another embodiment of the dosage form, the dosage form contains 79.8 mg of mannitol, 0.6 mg of colloidal silica, 10.0 mg of NF, 20.0 mg of talc, 4 , 8 mg hypromellose, 6.25 mg methacrylic acid-ethyl acrylate copolymer, 1.25 mg triethyl citrate and 3.1 mg talc extra fine.
In yet another embodiment of the dosage form, the dosage form contains 67.8 mg mannitol, 0.6 mg Aerosil, 10.0 mg NF, 20.0 mg pregelatinized starch, 2.0 mg
Stearic acid, 2.0 mg talc, 4.8 mg hypromellose, 4.0 mg methacrylic acid-ethyl acrylate
Copolymer, 0.8 mg triethyl citrate and 1.9 mg talc extra fine.
In yet another embodiment of the dosage form, the dosage form contains 45.0 mg mannitol, 0.4 mg Aerosil, 5.0% NF, 20.0 mg pregelatinized starch, 1.5 mg stearic acid, 1.5 mg talc, 3.5 mg hypromellose, 4.0 mg methacrylic acid-ethyl acrylate copolymer, 0.8 mg triethyl citrate and 1.9 mg talc extra fine.
In yet another embodiment of the dosage form described herein, the rasagiline content is 0.5 mg.
In yet another embodiment of the dosage form, the dosage form contains 80.3 mg mannitol, 0.6 mg Aerosil, 10.0 mg NF, 20.0 mg pregelatinized starch, 2.0 mg
Stearic acid, 2.0 mg talc, 4.8 mg hypromellose, 6.25 mg methacrylic acid-ethyl acrylate
Copolymer, 1.25 mg triethyl citrate and 3.1 mg talc extra fine.
In yet another embodiment of the dosage form, the dosage form contains 68.3 mg mannitol, 0.6 mg Aerosil, 10.0 mg NF, 20.0 mg pregelatinized starch, 2.0 mg
Stearic acid, 2.0 mg talc, 4.8 mg hypromellose, 4.0 mg methacrylic acid-ethyl acrylate
Copolymer, 0.8 mg triethyl citrate and 1.9 mg talc extra fine.
In yet another embodiment of the dosage form, the dosage form contains 45.5 mg mannitol, 0.4 mg Aerosil, 5.0 mg NF, 20.0 mg pregelatinized starch, 1.5 mg stearic acid, 1.5 mg talc , 3.5 mg hypromellose, 4.0 mg methacrylic acid-ethyl acrylate
Copolymer, 0.8 mg triethyl citrate and 1.9 mg talc extra fine.
In yet another embodiment of the dosage form described herein, the dosage form further contains 2.0 mg of a colored coating agent.
In yet another embodiment of the dosage form, the dosage form releases between 80 and 100% rasagiline when placed in a basket apparatus in 500 ml buffered aqueous medium at pH 6.8 at 37 ° C at 75 revolutions per minute for 20 minutes Minute is given.
In yet another embodiment of the dosage form, the total amount of nonpolar impurities is less than 0.3% by weight based on the amount of rasagiline.
In yet another embodiment of the dosage form, the amount of N- (2-chloroallyl) -1 (R) -aminoindan in the dosage form is less than 20 ppm relative to the amount of rasagiline.
In yet another embodiment of the dosage form, the amount of N- (2-chloroallyl) -1 (R) -aminoindane in the dosage form is less than 4 ppm based on the amount of rasagiline.
In yet another embodiment of the dosage form, the dosage form, when taken by a human, achieves a MAO-B inhibition substantially the same as that of the corresponding rasagiline dosage taken as a rapid release formulation becomes.
The present invention further provides rasagiline citrate.
In one embodiment of the rasagiline citrate, the rasagiline citrate is isolated rasagiline citrate or is substantially pure.
In yet another embodiment of the rasagiline citrate described herein is the 6/70 Austrian Patent Office AT12 508U1 2012-06-15
Rasagiline citrate amorphous.
In yet another embodiment of the rasagiline citrate described herein, the rasagiline citrate is mono-rasagiline citrate.
In yet another embodiment of the rasagiline citrate described herein, the rasagiline content in the rasagiline citrate is between 42% and 52% by weight based on the total weight of the rasagiline citrate.
By a range between 42% and 52%, all tenths and integer percentages within the range are expressly disclosed as part of the invention. Thus, 43%, 44%, ..., 50%, 51% and 42.1%, 42.2%, ..., 51.8%, 51.9% are included as embodiments of this invention.
In yet another embodiment of the rasagiline citrate described herein, the water content in the rasagiline citrate, as determined by Karl Fischer analysis, is less than 5%, less than 4%, less than 3%, less than 2%, or less than one %.
The present invention further provides a composition containing the rasagiline citrate and a carrier described herein.
In one embodiment of the composition, the composition further contains rasagiline base.
In yet another embodiment of the described composition, the rasagiline base is present in an amount of less than 5%, less than 4%, less than 3%, less than 2%, or less than 1% based on the composition Total content of rasagiline in the composition.
In yet another embodiment of the composition described herein, the rasagiline base present in the composition is crystalline rasagiline base.
In yet another embodiment of the composition described herein, the composition does not contain rasagiline base.
In yet another embodiment of the composition described herein, the rasagiline content present in the form of rasagiline citrate is greater than 50%, 60%, 70%, 80%, 90%, 95%, 96 %, 97%, 98% or 99% of the total rasagiline content in the composition.
In yet another embodiment of the composition described herein, the composition is a pharmaceutical composition and the carrier is a pharmaceutically acceptable carrier.
In yet another embodiment of the composition described herein, the composition is in the form of an oral dosage form.
In yet another embodiment of the composition described herein, the composition is in the form of a tablet.
In yet another embodiment of the composition described herein, the composition further contains stearic acid.
In yet another embodiment of the composition described herein, the composition is in the form of a transdermal patch.
In yet another embodiment of the composition described herein, the rasagiline citrate is mixed with a polymer.
[00107] The present invention further provides a method of making the rasagiline citrate or composition described herein, comprising: a) combining a citric acid solution with rasagiline base to form a first mixture; [00109] b) adding a solvent to the first mixture to form a second mixture; [00110] c) complete removal of liquid from the second mixture; and [00111] d) recovering the rasagiline citrate or preparing the composition.
In one embodiment of the process, the solvent added in step b) is acetone.
In a further embodiment of the method described here, the liquid is removed at ambient temperature and at reduced pressure in step c).
The present invention further provides a method of making the composition described herein, comprising: a) obtaining rasagiline citrate in isolated form; and [00116] b) mixing the rasagiline citrate with a carrier.
The present invention further provides a method of treating a human suffering from Parkinson's disease (PD), brain ischemia, stroke, traumatic brain injury, spinal cord injury, neurotrauma, neurodegenerative disease, neurotoxic injury, nerve injury , Dementia, Alzheimer's disease, senile dementia, depression, memory disorders, hyperactivity disorder, attention deficit disorder, multiple sclerosis (MS), schizophrenia, affective disorder, amyotrophic lateral sclerosis, restless legs syndrome (RLS), hearing loss, multisystem atrophy (MSA ), Glaucoma, modifying Parkinson's disease and progressive visual impairment (PSP), comprising administering to the human an amount of the dosage form or composition described herein which is effective in treating humans.
In one embodiment of the method, humans suffer from delayed emptying of the stomach.
In a further embodiment of the method, the administering step is performed on humans in a saturated state.
Each of the embodiments described herein may be combined with any other embodiment disclosed herein.
Through each area disclosed herein, all hundredths, tenths, and integer values within the range are expressly intended to be part of the invention. Thus, for example, 0.01 mg to 50 mg means 0.02, 0.03 ... 0.09; 0.1, 0.2 ... 0.9; and 1.2 ... 49 mg values are included as embodiments of this invention.
As used herein, an example of a rapid release rasagiline formulation is an AZILECT® tablet containing rasagiline mesylate.
As used herein, a polymer is a large molecule composed of repeating structural units, typically linked together by covalent chemical bonds.
As used herein, a " pharmaceutically acceptable " A carrier or excipient which is suitable for use in humans and / or animals without undesirable negative side effects (such as toxicity, irritation and allergic reaction) according to a reasonable benefit / risk ratio.
As used herein, " isolated " Compound a compound which was separated from the crude reaction mixture in which it was formed by an active Isolierungsakt. The isolation act necessarily involves separating the compound from the other known components of the crude reaction mixture, leaving some impurities, unknown by-products and residual amounts of the other known components of the crude reaction mixture therein. Cleaning is an example of an active isolation clock.
As used herein, a composition that means " free " of a chemical entity is that the composition, if any, contains an amount of the chemical entity that can not be avoided in the performance of an active act intended to separate the chemical entity and the composition.
As used herein, " approximately " in the context of a numerical value or range ± 10% of the said or claimed numerical value or range.
Citric acid is a weak organic acid and is tribasic. Therefore, the rasagiline citrate described herein may exist as a mono-, di- or tri-rasagiline citrate form or a mixture thereof.
A rapid release rasagiline formulation are AZILECT® tablets containing rasagiline (as the mesylate), a propargylamine-based drug indicated for the treatment of idiopathic Parkinson's disease. It is chemically referred to as: 1H-inden-1-amine, 2,3-dihydro-N-2-propynyl, (1R) -, methanesulfonate.
MAO inhibitors that selectively inhibit MAO-B largely lack the potential of " Cheese effect " to cause. However, there is a possibility that delayed emptying of R-PAI from the stomach contributes to this phenomenon. Therefore, it was an objective in developing the formulations of the present invention to develop an enterically coated sustained-release formulation containing rasagiline in an amount equivalent to 1 mg of rasagiline base, leaving the active ingredient in the duodenum and / or jejunum releases the stomach.
During development of the formulations of the present invention, the formulations were determined to comply with the bioequivalence criteria of the known rapid release rasagiline mesylate formulations (e.g. as disclosed in Example 1) in a single dose bioequivalence study healthy people. These criteria include a similarity of Cmax and AUC0-t (area under the curve) within the range of 80-125% within a 90% confidence interval between the new formulations and the known rapid release formulations. The difference between the two formulations should be evident in bioequivalence studies as a difference in tmax. In other words, the mean pharmacokinetic profile of the formulations of the present invention should be substantially the same as the mean pharmacokinetic profile of the formulations of the known rapid release formulation, except that the tmax for the sustained release formulation should be greater than that for the formulations Formulation with rapid release.
The reason for attempting to agree with the mean tmax and AUC0.1 of the known rapid release formulation (ie formulating a bioequivalent sustained release formulation) is that the efficacy of the rapid release formulation has been demonstrated, and that it is likely that the efficacy of the formulation is related to their mean Cmax and / or AUC. (Arch Neurol., 2002; 59: 1937-1943.) To achieve this goal, development has been directed to enteric coated tablets having a rapidly disintegrating core with an enteric coating, which results in release of rasagiline in a very specific manner pH range allows. This specific pH range would prevent the formulation from releasing rasagiline in the stomach and would allow the formulation to rapidly release rasagiline under the physiological conditions of the gut.
[00134] International application publication number WO 2006/014973 discloses delayed release pharmaceutical rasagiline mesylate formulations. In the disclosed formulations (Examples 1, 2 and 4) was a 30% dispersion of 9/70 Austrian Patent Office AT 12 508 Ul 2012-06-15
Methacrylic acid-ethyl acrylate copolymer (1: 1), which is known as Eudragil® L-30 D-55. Indeed, as disclosed in WO 2006/014973, these formulations are in fact sustained release formulations, as demonstrated by their dissolution profiles and in vivo data, however, their pharmacokinetic profile with respect to the mean Cmax is not consistent with the pharmacokinetic profile of Rasagiline mesylate rapid release formulations.
The 30% propellant carrier dispersion of methacrylic acid-ethyl acrylate copolymer (1: 1) known as Eudragil® L-30 D-55, which was used in the above-mentioned publication WO 2006/014973, prevents when applied as an aqueous dispersion either on tablets or on beads, the dissolution of the coated composition at low acid pH. This polymer has the following structure:
C = 0
O C2H5 The ratio of the free carboxyl groups to the ester groups is about 1: 1. The average molecular weight is about 250,000.
When this excipient is used in an aqueous dispersion or in an organic solution and formed into a film coating of a pharmaceutical formulation, it should dissolve at a pH of about 5.5. (Aqueous Polymer Coatings for Pharmaceutical Dosage Forms, Second Edition, Revised and Expanded, Ed. James W. McGinity, 1997.) Without wishing to be bound by theory, it is possible that these prior art formulations began at the lower end pH in the stomach, possibly in the presence of a food that can raise the pH in the stomach and continue to dissolve in the duodenum and jejunum for a prolonged period of time. The extended dissolution time could explain why the Cmax of these prior art formulations was significantly less than the Cmax of the rapid release formulations to which they were compared.
In general, the release process involves three major steps: 1. Transport to the site where the pH is high enough to allow release from the
Initiate dosage form.
2. dissolving the coating; and decay and release of the drug from the nucleus.
For highly soluble compounds, the third step is the most important. In contrast, for enteric-coated pellets, where depletion takes place gradually, not all at once, the first step has a greater impact on the PK profile. As pellets deflate at different times, they also reach the second step at different times. Therefore, the PK profile is a superposition of multiple " mini " -PK profiles.
The sustained-release compositions of the present invention are intended to withstand pH conditions of 6.0 and are said to release the active ingredient only above this pH. This specific pH was chosen to try to minimize any possible dissolution of the pharmaceutical compositions of the invention in the stomach in the saturated state and rapid dissolution of the pharmaceutical compositions according to the invention into the stomach in the duodenum and / or to allow the jejunum. The ability of a pharmaceutical formulation to enter the duodenum before releasing rasagiline and then rapidly release the rasagiline to the stomach provides a pharmacokinetic profile, and in particular a Cmax and AUCo t, similar to that of the known rapid release formulation.
To achieve the goal of a sustained release pharmaceutical formulation in which the Cmax is similar to the corresponding rapid release formulation is not easily achieved. In general, when compared to sustained release formulations with their counterparts in biostudies, the Cmax of the sustained release formulations is lower than the Cmax in the corresponding rapid release formulations. (Mäscher, et al., Arzneimittelforschung, 2001; 51 (6): 465-9, Behr, et al J. Clin Pharmacol., 2002; 42 (7): 791-7.) In addition, the present invention provides provides a solution to the problem of peripheral MAO inhibition by providing rasagiline-containing pharmaceutical dosage forms adapted to inhibit the release or absorption of rasagiline in the stomach (ie delaying the release of rasagiline to at least a portion of the dosage form has crossed the stomach). This avoids or minimizes absorption of rasagiline in the stomach, thereby avoiding or minimizing the potential Cheese effect.
The pharmaceutical dosage form may contain an acid-resistant excipient which prevents the dosage form or portions thereof from contacting the acidic environment of the stomach. The acid-resistant excipient may coat the rasagiline in the form of an enteric-coated tablet, capsule, hard or soft gelatin capsule. Enteric coating is, in the context of this invention, a coating which prevents the dissolution of an active ingredient in the stomach. This is determined by measuring the dissolution of the pharmaceutical dosage form in acidic solution as defined by USP methods. Even with enteric-pharmaceutical dosage forms, some of the dosage form may dissolve in the stomach; however, the dosage form may still be considered enteric-coated according to USP standards.
In all its aspects, the present invention provides pharmaceutical dosage forms useful for treating a condition selected from the group consisting of: Parkinson's disease (PD), cerebral ischemia, stroke, cerebral palsy Trauma, spinal cord injury, neurotrauma, neurodegenerative disease, neurotoxic injury, nerve injury, dementia, Alzheimer's disease, senile dementia, depression, memory disorders, hyperactivity disorder, attention deficit disorder, multiple sclerosis (MS), schizophrenia, affective disorder, amyotrophic lateral sclerosis , Restless legs syndrome (RLS), hearing loss, multisystem atrophy (MSA), glaucoma, modifying Parkinson's disease, and progressive visual palsy (PSP), but with a reduced risk of peripheral MAO inhibition, typically associated with the administration of rasagiline oral dosage forms.
Specific examples of pharmaceutically acceptable carriers and excipients which may be used to formulate oral dosage forms of the invention are described, for example, in U.S. Pat. Pat. No. 6,126,968 to Peskin et al., Published Oct. 3, 2000. Techniques and compositions for preparing dosage forms useful in the present invention are described, for example, in the following references: 7 Modern Pharmaceutics, Chapter 9 and 10 (Banker & Rhodes, Editors, 1979); Pharmaceutical Dosage Forms: Tablets (Lieberman et al., 1981); Ansei, Introduction to Pharmaceutical Dosage Form; second edition (1976); Remington's Pharmaceutical Sciences, 17th Edition (Mack Publishing Company, Easton, Pa., 1985); Advances in Pharmaceutical Sciences (David Ganderton, Trevor Jones, Eds., 1992); Advances in 11/70 Austrian Patent Office AT 12 508 Ul 2012-06-15
Pharmaceutical Sciences Volume 7 (David Ganderton, Trevor Jones, James McGinity, Eds., 1995); Aqueous Polymeric Coatings for Pharmaceutical Dosage Form (Drugs and the Pharmaceutical Sciences, Series 36 (James McGinity, Eds., 1989); Pharmaceutical Particulate Carriers: Therapeutic Applications: Drugs and the Pharmaceutical Sciences, Volume 61 (Alain Rolland, Eds., 1993); Drug Delivery to the Gastrointestinal Tract (Ellis Hor-wood Books in the Biological Sciences, Series in Pharmaceutical Technology; JG Hardy, SS Davis, Clive G. Wilson, Ed.); Modern Pharmaceutics Drugs and the Pharmaceutical Sciences, Volume 40 (Gilbert S. Banker , Christopher T. Rhodes, Editor).
The pharmaceutical dosage forms may be prepared as medicaments to be administered orally, parenterally, rectally or transdermally. Suitable forms for oral administration include tablets, pressed or coated pills, dragees, sachets, hard or soft gelatin capsules, sublingual tablets, syrups and suspensions; for parenteral administration, the invention provides ampoules or vials containing an aqueous or nonaqueous solution or emulsion; for rectal administration, the invention provides suppositories with hydrophilic or hydrophobic vehicles; Ointments for tropical use; and for transdermal delivery, the invention provides suitable delivery systems known in the art.
Tablets may contain suitable binders, lubricants, disintegrants, colorants, flavors, flow aids, melt aids, stabilizers, dissolution aids, antioxidants, buffers, chelants, fillers and plasticizers. For example, for oral administration in the unit dosage form of a tablet or capsule, the active drug component may be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as gelatin, agar, starch, methyl cellulose, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, microcrystalline Cellulose and the like combined. Suitable binders include starch, gelatin, natural sugars such as corn starch, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, povidone, carboxymethyl cellulose, polyethylene glycol, waxes and the like. Antioxidants include ascorbic acid, fumaric acid, citric acid, hybridsuccinic acid, gallic acid and its salts and esters, butylated hydroxyanisole, edetic acid. Lubricants used in these dosage forms include sodium oleate, sodium stearate, sodium benzoate, sodium acetate, stearic acid, sodium stearyl fumarate, talc and the like. Disintegrators include but are not limited to starch, methyl cellulose, agar, bentonite, xanthan gum, croscarmellose sodium, sodium starch glycolate, and the like; suitable plasticizers include triacetin, triethyl citrate, dibutyl sebacate, polyethylene glycol, and the like.
One type of oral dosage forms of the invention relates to sustained release formulations. Such formulations may contain an acid-resistant excipient which prevents the dosage form or portions thereof from contacting the acidic environment of the stomach. The acid-resistant excipient may coat the rasagiline in the form of an enteric-coated tablet, capsule or gelatin capsule. Enteric coating is, in the context of this invention, a coating which prevents the dissolution of an active ingredient in the stomach. Specific examples of pharmaceutically acceptable carriers and excipients that can be used to formulate such sustained release formulations are described, for example, in International Application Publication No. WO 06/014973, which is hereby incorporated by reference in its entirety.
Another type of oral dosage forms of the present invention relates to rapidly disintegrating formulations which provide a means for preventing the absorption of rasagiline in the stomach and which eliminate the need to swallow tablets by absorbing rasagiline into the body before it reached the stomach. Such absorption of rasagiline can be achieved by contact with the buccal, sublingual, pharyngeal and / or esophageal mucosal membranes. To achieve this, the rapidly disintegrating formulations are formed so that they rapidly disperse within the mouth to maximize the contact of rasagiline with the buccal, sublingual, pharyngeal and / or esophageal To allow mucosal membranes. Specific examples of pharmaceutically acceptable carriers and excipients that can be used to formulate such rapidly disintegrating formulations are described, for example, in International Application Publication No. WO 03/051338, which is hereby incorporated by reference in its entirety.
Other pharmaceutical compositions of the invention include transdermal patches. Transdermal patches are drug-adherent adhesive patches that are placed on the skin to deliver a timed dose of drug through the skin and into the blood stream. A wide variety of drugs can be delivered by transdermal patches. Some medicines need to be combined with other substances, such as alcohol, to increase their ability to penetrate the skin. Transdermal patches have several important components, including a liner to protect the patch during storage, the remedy, adhesive, a membrane (to control the release of the drug from the reservoir), and a backsheet to hold the patch against the patch protect external environment. The two most common types of transdermal patches are the matrix type and reservoir type. (Reservoir type plasters have a remedy combined with a non-volatile, inert liquid such as mineral oil, whereas in plasters the matrix Type of remedy is dispersed in a lipophilic or hydrophilic polymer matrix, such as acrylic or vinyl polymers. Adherent polymers, such as polyisobutylene, are used to fix the patch to the skin. (Stanley Scheindlin, (2004) "Transdermal Drug Delivery: PAST, PRESENT, FUTURE," Molecular Interventions, 4: 308-312) The greatest limitation on transdermal drug delivery is the intrinsic barrier property of the skin. Frequently, penetration enhancers are added to transdermal drug formulations to disrupt the skin surface and to induce faster drug delivery. Typical penetration enhancers include high boiling alcohols, diols, fatty acid esters, oleic acid and glycerin based solvents, and are usually added in a concentration of one to 20 percent (w / w). (Melinda Hopp, " Developing Custom Adhesive Systems for Transdermal Drug Delivery Products, " Drug Delivery) The basket-like apparatus used in this invention is that described in the United States Pharmacopeia, 29th Edition (2006), Apparatus 7 described in Chapter 711. The apparatus is constructed as follows: The structure consists of the following: A jacketed housing of glass or other inert transparent material; an engine; a metallic drive shaft; and a cylindrical basket. The housing is partially immersed in a suitable water bath of a suitable size or placed in a heating jacket. The water bath or heating mantle makes it possible to maintain the temperature within the vessel at 37 ± 0.5 during the test, and to keep the liquid of the bath in a constant, gentle motion. No part of the structure, including the environment in which the structure is incorporated, significantly contributes to movement, agitation or vibration beyond that caused by the gently rotating stirring element. An apparatus which allows observation of the sample and the stirring element during the test is preferred. The container is cylindrical with a hemispherical bottom and one of the following dimensions and capacities: for a nominal capacity of 1 liter, the height is 160 mm to 210 mm and its inner diameter is 98 mm to 106 mm; for a nominal capacity of 2 liters, the height is 280 mm to 300 mm and its inner diameter is 98 mm to 106 mm; and for a nominal capacity of 4 liters, the height is 280 mm to 300 mm and its inner diameter is 145 mm to 155 mm. Its sides are flanged at the top. A suitable cover can be used to reduce evaporation. 13/70 Austrian Patent Office AT12 508U1 2012-06-15
The shaft is arranged so that its axis is no more than 2 mm away from the vertical axis of the vessel at any point and rotates smoothly and without significant wobble. A speed regulating device is used which allows the rotational speed of the shaft to be selected and maintained within ± 4% of the speed specified in the individual monograph. The shaft and basket components of the agitator are made of Type 316 stainless steel or equivalent.
Unless otherwise specified in the individual monograph, use a 40-mesh cloth. A basket with a gold coating of 0.0001 inch (2.5 μm) thick can be used. The dosage unit is placed in a dry basket at the beginning of each test. The distance between the inner bottom of the container and the basket is maintained at 25 ± 2 mm during the test.
Owing to the sensitivity of rasagiline base to UV radiation and light in general, it is advisable during the preparation of the formulations described in the following examples to perform the process in a low UV environment, preferably in a non-UV environment -Radiation.
The present invention is also intended to include all isotopes of atoms found in the compounds disclosed herein. Isotopes include those having the same atomic number but different mass numbers. As a general example, and without limitation, hydrogen isotopes include tritium and deuterium. Carbon isotopes include C-13 and C-14.
It should be noted that any indication of a carbon in structures in this application, when given without further indication, is intended to represent all isotopes of carbon, such as 12C, 13C or 14C. In addition, all compounds containing 13C or 14C may expressly have the structure of each of the compounds disclosed herein.
It should also be noted that any indication of a hydrogen atom in structures within this application, when otherwise given, is intended to represent all isotopes of hydrogen, such as 1H, 2H or 3H. In addition, all compounds containing 2H or 3H may expressly have the structure of all the compounds disclosed herein.
Radiolabeled compounds can generally be prepared by conventional techniques known to those skilled in the art, or by methods analogous to those disclosed in the examples disclosed herein, using appropriate radiolabelled reagents in place of the present invention used unlabelled reagents.
This invention will be better understood by the following experimental details. However, one skilled in the art will readily recognize that the specific methods and results discussed are illustrative only of the invention as further described in the claims thereafter.
EXPERIMENTAL DETAILS
EXAMPLE 1. RASAGILIN TABLETS WITH RAPID RELEASE
Rapid release rasagiline tablets were prepared using the ingredients listed in Table 1. 14/70 Austrian Patent Office AT12 508U1 2012-06-15 [00166] Table 1
Component Function Per tablet (mg) (0.5 mg rasagiline base) Per tablet (mg) (1 mg rasagiline base) Rasagiline mesylate 0.78 1.56 Mannitol Filler 79.62 159.24 Aerosil Fluid 0.6 1,2 starch NF binder 10,0 20,0 starch, pregelatinized (starch STA-RX 1500) disintegrant 10,0 20,0 talc lubricant 2,0 4,0 stearic acid lubricant 2,0 4,0 total weight of the tablet core 105 210 Rasagiline mesylate, mannitol, half of the colloidal silica, starch and pregelatinized starch were mixed in a Diosna P-800 mixer for about 5 minutes. Water was added and the mixture was further mixed. The granules were dried and the remainder of the colloidal silica was added. The granules were crushed in a Frewitt mill and stearic acid and talc were added. The granules were mixed in a tumbler for 5 minutes and tabletted.
EXAMPLE 2. TABLETS WITH RASAGILINE BASE
An attempt was made to formulate tablet cores having a pharmacokinetic profile (Cma * and AUC) similar to that of the rapid release formulation of Example 1.
A method of making crystalline rasagiline base is disclosed in U.S. Pat. Patent Application Publication No. 2008/0161408 (corresponding to WO 2008/076348). In particular, the document describes a process for producing crystalline rasagiline base, which comprises: a) dissolving a salt of R (+) - N-propargyl-1-aminoindan in water to form a solution; B) cooling the solution to a temperature of about 0-15 KD; C) basifying the solution to a pH of about 11 to form a suspension; and [00173] d) obtaining the crystalline rasagiline base from the suspension.
[0017] Five preliminary formulations of rasagiline base API were prepared based on the rapid release rasagiline formulation of Example 1 using standard tabletting technique. Different reagents were added to stabilize the API within the formulation. 15/70 Austrian Patent Office AT12 508U1 2012-06-15 Table 2: Compositions of tablet cores with rasagiline base:
Composition 1 Composition 2 Composition 3 Composition 4 Composition 5 Rasagiline Base Rasagiline Base Rasagiline Base Rasagiline Base Rasagiline Base Citric Acid Maleic Acid Succinic Acid Maleic BHT Mannitol USP / EP Mannitol USP / EP Mannitol USP / EP Mannitol USP / EP Mannitol USP / EP Colloidal Silica Colloidal Silica Colloidal Silica Colloidal Silica Colloidal Silica Pregelatinized Starch Pregelatinized Starch Pregelatinized Starch Pregelatinised Starch Pregelatinized Starch Starch NF / EP Starch NF / EP Starch NF / EP Starch NF / EP Starch NF / EP Stearic Acid Stearic Acid Stearic Acid Stearic Acid Stearic Acid Talc Talk Talc Talk Talc Each composition was prepared in a laboratory scale batch of ~ 500 tablets using laboratory equipment with non-GMP portion of API.
Stability results for all five formulations (final mixtures) were obtained by short term stability studies under accelerated and room temperature conditions. Stability results, content of each formulation, and dissolution results of tablets pressed using an eccentric press are shown in the following tables.
Composition 1 mg / tablet starting materials Stability results of the test (%) Time 0 2 weeks 25 ° C 2 weeks 40 <€ 1 month 40-C 0.82 Citric Acid 101.6% 94.2% 94.8% 98.0% water 1.00 Rasagiline base 80.0 Mannitol USP / EP 0.3 Aerosil 200 Stability results - (5 vials of impurity ^ _ 10.0 strength NF / EP time 0 2 weeks 25 ° C 2 weeks 40 <€ 1 month 40 <O 20.0 Strength STA-RX 1500 Total Contamination <0.04 (NG) <0.04 (NG) <0.1 (QG) <0.2 (QG ) 0.3 Aerosil 200 2.0 stearic acid 2.0 talc 116.42 total weight [00179] (translator's note: NG = detection limit; QG = limit of quantification) 16/70 Austrian Patent Office AT12 508U1 2012-06-15 [00180] Composition 2 mg / tablet starting material Stability results of the test (%) Time 0 2 weeks 25 ° C 2 weeks 40 ° C 1 month 40 ° C 0.7 Maleic acid 82.3 84.6 79.8 80.8 Water 1.00 Rasagiline base 8 0.0 Mannitol USP / EP Stability Results - (5 Level of Impurity Q_ 0.3 Aerosil 200 Time 0 2 Weeks 25 ° C 2 Weeks 40 <€ 1 Month 40 <€ 10.0 Strength NF / EP Total Contamination & lt 0.1 (QG) 0.1 0.4 0.8 20.0 Starch STA-RX 1500 0.3 Aerosil 200 2.0 Stearic acid 2.0 Talc 116.3 Total Weight Composition 3 mg / tablet starting material alien Stability results of the test (%) Time 0 2 weeks 25 ° C 2 weeks 40 ° C 1 month 40 ° C 0.7 Succinic acid 102.9 99.4 100.6 101.9 Water 1.00 Rasagiline base Stability results - _ Level of contamination Q_ 80.0 Mannitol USP / EP Time 0 2 weeks 25 ° C 2 weeks 40 ° C 1 month 40 ° C 0.3 Aerosil 200 Total Contamination 0.4 0.4 0.6 1.2 10.0 Starch NF / EP 20.0 Starch STA-RX 1500 0.3 Aerosil 200 2.0 Stearic acid 2.0 Talc 116.3 Total Weight 17/70 Austrian Patent Office AT12 508U1 2012-06-15 Composition 4 mg / tablet Starting materials Stability results of the test (%) Time 0 2 weeks 25 ° C 2 weeks 40 ° C 1 month 40 ° C 0.8 Maleic acid 103.4 101.5 101.5 102.2 Water 1.00 Rasaqiline base 80, 0 Mannitol USP / EP 0.3 Aerosil 200 Stability results - _ (5 Level of impurity Q_ 10.0 Strength NF / EP Time 0 2 weeks 25 ° C 2 weeks 40 ° C 1 month 40 ° C 20.0 Starch STA-RX 1500 total impurity <0.04 (NG) <0.04 (NG) <0.1 (QG) <0.2 (QG) 0.3 Aerosil 200 2.0 stearic acid 2.0 talc 116, 4 Total weight Composition 5 mg / tablet Starting materials Stability results of the test (%) Time 0 2 weeks 25 ° C 2 weeks 40 ° C 1 month 40 ° C Ethanol 95% 67.8 65.7 48.5 31.9 0.02 BHT 1.00 Rasagiline Base Stability Results - _ (5 Level of Impurity Q_ 80.0 Mannitol USP / EP Time 0 2 Weeks 25 ° C 2 Weeks 40 ° C 1 Month 40 ° C 0.3 Aerosil 200 Total Contamination <0.1 (QG) <0.1 (QG) 2, 9 5.7 10.0 Starch NF / EP 20.0 Starch STA-RX 1500 0.3 Aerosil 200 2.0 Stearic acid 2.0 Talc 115.62 Total weight 18/70 Austrian Patent Office AT12 508U1 2012-06-15 [00184 ] Dissolution results (% in 0.1 N HCl) 5 min 10 min 15 min Composition 1 85 99 100 Composition 2 49 82 90 Composition 3 62 98 103 Composition 4 59 100 107 Composition 5 70 70 70
Dissolution results (% in phosphate buffer pH 6.8) 5 min 10 min 15 min Composition 1 78 92 94 Composition 2 40 77 82 Composition 3 59 98 101 Composition 4 59 95 102 Composition 5 70 70 70 DISCUSSION: Compositions 1 and 4 containing citric acid or maleic acid as the antioxidant, gave the best stability results and a satisfactory dissolution profile. Therefore, they were selected for further development.
EXAMPLE 3. PREPARATION OF MAGENTATE-RESISTANT COATED TABLETS WITH DELTA ACID WITH DELAYED RELEASE
In this example, an enteric-coated rasagiline tablet containing citric acid was prepared.
Rasagiline citrate was identified as formed in the tablet prepared as described in this example.
Example 3a -1.0 mg rasagiline base, 117 mg weight of the tablet core (Formulation I) Table 3a: Composition of enteric-coated rasagiline sustained-release tablet.
Component Function Per tablet (mg) Tablet cores Rasagiline Base Remedy Substance 1.0 Citric Acid Antioxidant / Stabilizer 1.6 Mannitol Filler 79.84 Colloidal Silica Flux 0.6 Starch NF Binder 10.0 Pregelatinized Starch (STA-RAX® 1500) Disintegrant 20.0 talc lubricant 2.0 stearic acid lubricant 2.0 total weight of tablet core 117.0 subcoating Pharmacoat® 606 (Hypromello-se USP) granules coating agent 4.8 purified water processing aid 19/70 AT12 508U1 2012-06-15 Austrian
Patent Office
Coating suspension Eudragil® L-30D-55 Coating compound 6.250 * talc USP extrafine lubricant 1.25 triethyl citrate plasticizer 3.1 Purified water processing aid Total weight of tablet 132.4 * dry matter remaining in the core.
[00189]!. Dry mixing: Mannitol, half of the aerosol, pregelatinized starch and NF starch were added to a high shear granulating mixer and premixed for one minute by mixing at mixer speed I, followed by one minute mixing at mixer speed I and chopper I ,
[00191] II. Wet Granulation: Citric acid solution was prepared using 320 g of citric acid in purified water at a weight ratio of about 1: 10.6 to 1: 6.
Rasagiline base was added with stirring over a period of about 15 minutes. Stirring was continued until a clear solution was observed. The solution was placed in a high shear granulating mixer and the contents were mixed for about two minutes at a Mixer Speed II and Chopper II. An extra amount of water was added to the granulating high shear mixer and the solution was mixed for two more minutes at a Mixer Speed II and Chopper II.
The wet granules were emptied into a carriage of a fluidized bed dryer at mixer speed I.
III. Fluid Bed Drying: The material of step II was dried in a fluid bed dryer at an air inlet temperature of 45 ° C (40 ^ -50 ° C) and an air outlet temperature of at most 37-38 ° C.
[00197] IV. Comminution: The dry granules and the remaining amount of Aerosil were comminuted by an oscillating granulator with a 0.6 mm sieve into a storage container.
Furthermore, the crushed granules were weighed.
V. Final Blending: Stearic acid and talc were screened through a 50 mesh screen and transferred to the Y cone / container.
1. The mixture was mixed for 5 minutes.
2. The final mixture was obtained and the percent yield was determined.
3. The final mixture was stored in a container using an inner transparent polyethylene bag and an outer black polyethylene bag. Two silica gel pads were placed between the two polyethylene bags.
4. Samples were taken for a homogeneity test of the mixture.
VI. Tabletting A tablet press machine (FETTE 1200) was constructed with the desired punches of 6.0 mm.
The in-process control or control tests for tablets during the 20/70 Austrian Patent Office AT12 508U1 2012-06-15
Procedures included average weight, individual weight, thickness, hardness, friability and disintegration.
Procedural Control Specifications for 1 mg Rasagiline Base DR Tablets are:
Parameter Minimum Target Maximum Maximum Average Weight (mg) 111 117 123 Individual Weight (mg) 111 117 123 Thickness (mm) 3.3 3.6 3.9 Hardness (SCU) 7 9 11 Friability (%) - - 1.0 Decay ( Minutes) - - 5 [00210] The tablets were weighed and the percent recovery was calculated.
[00211] VII. Undercoating: [00212] Tablet cores were first coated with hypromellose (Pharmacoat 606®) as a precoat followed by coating with methacrylic acid-methyl methacrylate copolymer [1: 1] (Eudragit® L-30D-55, 30%). Eudragit® L100-55 dispersion) to prevent any interaction between the rasagiline base in the core and the Eudragit L polymer.
[00213] 1. Preparation of Pharmacoat 606® Solution:
Hypromellose USP solution was prepared using hypromellose in purified water at a weight ratio of about 1:10.
[00214] 2. preheating:
The tablet cores were placed in an (Ohara) coater shovel. The tablets were heated at an air inlet temperature of 50 ° C (45 ° C-55 ° C) and an air outlet temperature of 45-50 ° C.
[00215] 3. Spray Method:
The tablet cores were sprayed with Hypromellose in the Ohara Coater Coater Shovel. The air inlet temperature was 50 ° C; the air outlet temperature was 350. The blade speed was set at 16 rpm (may vary from 14-18 rpm). The spray rate was 15-35 g / min. The tablets were dried for one hour at an air inlet temperature of 45 ° C (temperature range 400-500).
[00216] VIII. Enteric coating: [00217] 1. Preparation of enteric coating dispersion from Eudragit® L100-55:
Triethyl citrate was mixed with water for 15 minutes. Within 10 minutes, the talc was added extra fine in an Ultraturax to the dispersion of triethyl citrate and water. A 30% dispersion of Eudragit L100-55 was added to the triethyl citrate / talc dispersion, filtered and stirred.
[00218] 2. Heating:
The precoated tablets were placed in an Ohara Coater coating blade. The tablets were heated at an air inlet temperature of 50 * 0 (45-55 * 0) and an air outlet temperature of 450 (40 ° -500).
[00219] 3. Spray Method:
The tablets were sprayed with the dispersion in an Ohara coater shovel. The air inlet temperature was in the range of 40-500, the air outlet temperature was in the range of 30-400. The blade speed was set at 16 rpm in the range of 14-18 rpm and the spray rate was 5-20 g / min. The tablets were dried for 2 hours. The air inlet temperature was 500 at minimum blade speed. 21/70 Austrian Patent Office AT 12 508 Ul 2012-06-15 [00220] Eudragil® L100-55 contains an anionic copolymer based on methacrylic acid and ethyl acrylate. It is also known as methacrylic acid copolymer type C. The ratio of the free carboxyl groups to the ester groups is about 1: 1. The average molecular weight is about 250,000. CH3
O
OH
O cp,
Example 3b -1.0 mg rasagiline base, 76 mg tablet core weight (Formulation III) [00221] This formulation was prepared using similar steps as described in Example 3a.
Table 3b: Composition of enteric-coated rasagiline base tablet with delayed release
Component Function Per tablet (mg) Tablet cores Rasagiline Base Remedy Substance 1.0 Citric Acid Antioxidant / Stabilizer 1.6 Mannitol Filler 45.0 Aerosil Solvent 0.4 Starch NF Binder 5.0 Pregelatinized Starch (Thickness STA-RX 1500) Disintegrant 20 , 0 talc lubricant 1.5 stearic acid lubricant 1.5 total weight of tablet core 76.0 subcoating Pharmacoat® 606 (hypromellose USP) granules coating agent 3.5 purified water processing aid coating suspension Eudragil® L-30D-55 coating agent 4.0 * talc USP extrafine Lubricant 1,9 Triethyl citrate NF Plasticizer 0,8 Purified water Processing aids Total tablet weight 86,2 * Dry matter remaining on the core.
Example 3c - 0.5 mg rasagiline base, 117 mg tablet core weight [00223] This formulation was prepared using similar steps as described in Example 3a. [00224] Table 3c: Composition of enteric-coated rasagiline base tablet with delayed release
Component Function Per tablet (mg) Tablet cores Rasagiline Base Medicinal substance 0.5 Citric Acid Antioxidant / Stabilizer 1.6 Mannitol Filler 80.34 Aerosil Plasticizer 0.6 Starch NF Binder 10.0 Starch, pre-gelatinized (starch STA-RX 1500) Disintegrant 20 , 0 Talc Lubricant 2.0 Stearic acid Lubricant 2.0 Total weight of tablet core 117.0 Undercoating Pharmacoat® 606 (Hypromello-se USP) Granules Coating agent 4.8 Purified water Processing aid Coating suspension Eudragit® L-30D-55 Coating material 6.25 * Talc USP extrafine lubricant 3.1 triethyl citrate NF plasticizer 1.25 purified water processing aid total weight of the tablet 132.4 * dry matter remaining on the core.
Example 3d - 0.5 mg Rasagiline Base, 76 mg Tablet Core Weight [00225] This formulation was prepared using similar steps as described in Example 3a.
Table 3: Composition of enteric-coated rasagiline base tablet with delayed release
Component Function Per tablet (mg) Tablet cores Rasagiline Base Medicinal substance 0.5 Citric Acid Antioxidant / Stabilizer 1.6 Mannitol Filler 45.5 Aerosil Solvent 0.4 Starch NF Binder 5.0 Pregelatinized Starch (Thickness STA-RX 1500) Disintegrant 20 , 0 talc lubricant 1.5 stearic acid lubricant 1.5 total weight of the tablet core 76.0 subcoating Pharmacoal® 606 (Hypromello-se USP) granules coating agent 3.5 purified water processing aid 23/70 AT12 508U1 2012-06-15 Austrian
Patent Office
Coating suspension Eudragit® L-30D-55 Coating agent 4.0 * Talc USP extrafine Lubricant 1.9 Triethyl citrate NF Plasticizer 0.8 Purified water Processing aid Total weight of tablet 86.2 * Dry matter remaining on the core.
EXAMPLE 4. RESOLUTION RESULTS OF TABLETS MANUFACTURED IN ACCORDANCE WITH EXAMPLE 3A
The tablets prepared according to Example 3a were tested in terms of their dissolution profile in various media according to USP method. The following data represent an average for 4 tablets.
The statement "% Rasagiline released &quot; in the following tables refers to a standard that is 1 mg rasagiline.
[00229] Tablet cores:
Dissolution profile (% Rasagiline released) - 0.1 N HCl, 75 rpm, 2> 1 ° C 10 min. 20 min. 30 min. 45 min. 1,101,102,102,102,105,105,105,103,104 105 105 105 4 106 106 107 107% average 104 105 105 105
Dissolution profile (% rasagiline released) - phosphate buffer, 75 rpm, 37 ° C. 10 min. 20 min. 30 min. 45 min. 1 98 99 99 99 2 100 101 101 102 3 99 100 100 101 4 96 96 97 97% Average 98 99 99 100% RSD 1.9 2.0 2.0 2.2 [00232] Undercoated tablets:
Dissolution profile (% Rasagiline released) - 0.1 N HCl, 75 rpm, 37 ° C. 10 min. 20 min. 30 min. 45 min. 1 105 105 106 106 2 109 109 109 109 3 103 104 104 104 4 103 104 103 104% Average 105 105 105 106% RSD 2.5 2.3 2.3 2.3 [00234] Coated tablets: [00235] According to the USP specification for sustained release articles (enterically coated) , 29th Ed., Chapter 724, the dissolution profile of the coated tablets in 0.1 N HCl was acceptable, showing less than 10% release after 120 minutes. 24/70 Austrian Patent Office AT12 508U1 2012-06-15 [00236] Dissolution profile (% Rasagiline released) - Phosphate buffer pH 5.8 10 min. 20 min. 30 min. 40 min. 60 min. 90 min.% Average 0 0 0 0 0 [00237] Dissolution profile (% rasagiline released) - phosphate buffer pH 6.4 10 min. 20 min. 30 min. 40 min. 60 min. 90 min.% Average 0 35 93 96 96 96% RSD 2.2 1 3 1.3 1.2 [00238] Dissolution profile (% Rasagiline released) - Phosphate buffer pH 6.8 10 min. 20 min. 30 min. 40 min. 60 min. 90 min. % Average 11 92 95 95 94 94% RSD 3.7 1.6 1.6 1.5 1.6 [00239] Discussion: The tablets prepared according to Example 3a do not start the release of rasagiline at a pH of lower as 6.0. At a pH of 6.8, rapid release of rasagiline occurs and over about 20 minutes, more than 90% of the radiolabel is released from the formulation.
During development of the formulations of the present invention, the formulations were determined to comply with the bioequivalence criteria of the known rapid release rasagiline mesylate formulations (as disclosed, for example, in Example 1) in a single dose bioequivalence study healthy people. These criteria include a similarity of Cmax and AUC0. (Area under the curve) within the range of 80-125% within a 90% confidence interval between the new formulations and the known rapid release formulations. The difference between the two formulations should be evident in bioequivalence studies as a difference in tmax. In other words, the mean pharmacokinetic profile of the formulations of the present invention should be substantially the same as the mean pharmacokinetic profile of the formulations of the known rapid release formulation, except that the tmax for the sustained release formulation should be greater than that for the formulation with rapid release.
The reason for attempting to agree with the mean tmax and AUC0. Of the known rapid release formulation (ie, to formulate a sustained release formulation that is bioequivalent) is that the effectiveness of the rapid release formulation and that it is likely that the efficacy of the formulation is related to its mean Cmax and / or AUC. (Arch Neural, 2002; 59: 1937-1943.) To achieve this goal, development has been directed to enteric coated tablets having a rapidly disintegrating core with an enteric coating, which results in release of the rasagiline in a very specific manner pH range allows. This specific pH range would prevent the formulation from releasing rasagiline in the stomach and would allow the formulation to rapidly release rasagiline at the physiological conditions of the gut.
Although the tablets according to Example 3a were coated with an enteric coating containing methacrylic acid-ethyl acrylate copolymer, which were the coatings in International Application Publication No. WO 2006/014973, the tablets according to Example 3a were able to a pH of 6.0 and smaller, whereas the composition in WO 2006/014973 was not.
The difference in the dissolution profiles is based on the fact that in the composition according to the invention the formulation of the core contained a large amount of disintegrants and that the enteric film had a smaller ratio from polymer to plasticizer. The ratio of polymer to plasticizer between 10: 1 and 2: 1, and especially 5: 1, allows for improved in vitro dissolution profiles.
The dissolution profile of the formulation according to Example 3a enables the composition to have improved pharmacokinetic properties similar to those of the currently marketed rapid release formulations.
EXAMPLE 5. STABILITY RESULTS OF TABLETS MANUFACTURED ACCORDING TO EXAMPLE 3A
Stability of enteric-coated tablets prepared using citric acid-containing formulations was tested under various storage conditions. The results are summarized below.
Stability Results (Accelerated Conditions): According to the USP specification for sustained-release articles (enteric-coated), 29th edition, Chapter 724, the dissolution profile of the enteric-coated tablets in 0.1 N HCl was acceptable 10% release after 120 minutes.
[00250] The following table shows that dissolution profile for enteric-coated tablets after different storage times.
Dissolution profile of coated tablets - phosphate buffer pH 6.8, 37Ό
Dissolution rate (% release after different storage time of rasagiline) Storage period (months) 10 min. 20 min. 30 min. 40 min. 60 min. 90 min. 0 11 92 95 95 96 96 1 28 95 96 96 97 97 2 12 97 The% rasagiline released in the above table refers to a standard that is 1 mg rasagiline.
The following tables show the analytical results for different batches of the enteric coated tablets under different storage conditions.
Coated tablets - batch 1
Conditions Sample% Total Verify (%) T = 0 101.5 <NG 1 month 101.1 <NG 40 ° C, 75 rF 2 months 105.4 0.3% 3 months 104.5 0, 4% 4 months 100.9 0.4% 25 ° C, 60 rF 1 month 104.7 <NG 3 months 106.2 <NG 26/70 Austrian Patent Office AT12 508U1 2012-06-15 Coated tablets - batch 2
Conditions Sample% Total Verify (%) T = 0 98.6 <NG 1 month 99.1 0.05% 40 <€, 75 rF 2 months 96.3 0.1% 3 months 95.6 0.2% 4 months 96.6 0.3% 1 month 99.8 <NG 30 ° C, 65 ° F 2 months 98.4 <NG 3 months 96.5 <NG 1 month 98.4 &lt; NG 25 ° C, 60 rF 2 months 95.8 <NG 3 months 96.2 <NG
Coated tablets - batch 3
Conditions Sample% Total Impurities (%) T = 0 100.3 <NG 40 ° C, 75 rF 1 month 100.3 <NG 40 ° C, 75 rF 2 months 102.0 <NG 40 ° C, 75 rF 3 months <0.28 30 ° C, 65 rF 3 months <0.08 25 ° C, 60 rF 1 month 101.2 <NG 25 ° C, 60 rF 2 months 102.1 <NG 25 ° C, 60 rf 3 months <0.08 N- (2-chloroallyl) -1 (R) -aminoindane (2-CI-AAI) impurities
Batch number 2-CI-AAI content,% 1 LT 0.00004 2 LT 0.00004
EXAMPLE 6. PREPARATION OF MAGENTATE-RESISTANT COATED RASAGILIN-BASE TABLETS WITH MALIC ACID WITH DELAYED RELEASE
Example 6a -1 mg rasagiline base, 117 mg tablet core weight Table 6a: Composition of enteric-coated rasagiline base delayed release tablets
Component Function Per tablet (mg) Tablet cores Rasagiline base Remedy Substance 1.0 Maleic Acid Antioxidant / Stabilizer 1.6 Mannitol Filler 80.0 Aerosil Plasticizer 0.6 Starch NF Binder 10.0 Starch, pre-gelatinized (Thickness STA-RX 1500) Disintegrant 20 , 0 talc lubricant 2.0 stearic acid lubricant 2.0 total weight of the tablet core 117,2 27/70 AT12 508U1 2012-06-15 Austrian
Patent Office
Undercoating Pharmacoat® 606 (Hypromello-se USP) Granules Coating Composition 4.8 Purified Water Processing Aid Coating Suspension Eudragit® L-30D-55 Coating Material 6.25 * Talc USP Extra Fine Lubricant 3.1 Triethyl Citrate NF Plasticizer 1.25 Purified Water Processing Aid Total Weight tablet 132.6 * Dry matter remaining on the core.
[00260]!. Dry Mixing: Mannitol, half of the aerosol, pregelatinized starch and NF starch were added to a high shear granulating mixer and premixed for one minute by mixing at mixer speed I, followed by one minute mixing at mixer speed II and chopper II ,
II. Wet granulation: A maleic acid solution was prepared using maleic acid in purified water in the ratio of about 1: 10.6 to 1: 6.
Rasagiline base was added with stirring over a period of about 15 minutes. Stirring was continued until a clear solution was observed.
The solution was placed in a high shear granulating mixer and mixed for about 2 minutes by mixing at a Mixer Speed II and Chopper II. An extra amount of water was added to the granulating high shear mixer and the solution was mixed for a further 2 minutes at a Mixer Speed II and Chopper II.
The wet granules were discharged on the carriage of a fluidized bed dryer at mixer speed I.
III. Fluid Bed Drying: The material was dried in a fluid bed dryer at an air inlet temperature of 45 ° C (40 ° to 50 ° C) and an air outlet temperature of 37-38 ^ 0 maximum.
[00269] IV. Comminution: The dry granules were comminuted with the remaining amount of Aerosil by an oscillating granulator with a 0.6 mm sieve into a storage container.
The crushed granules were weighed.
V. Final Mixing: 1. Stearic acid and talc were sieved through a 50 mesh sieve and transferred to the Y cone or container.
2. The mixture was mixed for 5 minutes.
3. The final mixture was stored in a container using an inner transparent polyethylene bag and an outer black polyethylene bag. Two silica gel pads were placed between the two polyethylene bags.
4. Samples were taken for a homogeneity test of the mixture. 28/70 Austrian Patent Office AT 12 508 Ul 2012-06-15 [00277] VI. Tableting: A tablet press machine was constructed with the desired punches of 6.0 mm. The diameter of the punch can change by ± 10%.
The in-process control tests included average weight, individual weight, thickness, hardness, friability and disintegration.
Procedural Control Specifications for 1 mg Rasagiline Base DR Tablets are:
Parameter Minimum Target value Maximum Average weight (mg) 111 117 (Actual value 121) 123 Individual weight (mg) 111 117 123 Thickness (mm) 3.3 3.6 (Actual value 3.7) 3.9 Hardness (SCU) 7 9 (Actual value 10) 11 Friability (%) - - 1.0 Decay (minutes) - - 5 The tablet cores were weighed and the percent recovery was calculated.
VII. Undercoating: Tablet cores were first coated with hypromellose (Pharmacoat 606) as a precoat, followed by coating with methacrylic acid-methyl methacrylate copolymer [1: 1] (Eudragit® L-30D-55, 30%). dispersion of Eudragit® L100-55) to prevent any possible interaction between the rasagiline base in the core and the Eudragit L polymer.
1. Preparation of Pharmacoat 606 Solution:
Pharmacoat 606 (Hypromellose-USP) solution was prepared using Pharmacoat in purified water at a weight ratio of 1:10.
[00285] 2. preheating:
The tablet cores were placed in an Ohara Coater coating paddle and the tablets were heated at an air inlet temperature of 50 ° C (45 ° to 55 ° C) and an air outlet temperature of 40 ° to 50 ° C.
[00286] 3. Spray Method:
The tablet cores were sprayed with solution in the Ohara Coater Coating Shovel. The air inlet temperature was 50 ° C (temperature range 40-50'C); the air outlet temperature was in the range of 30-40 ^. The blade speed was set at 16 rpm in the range of 14-18 rpm; the spray rate was 15-35 g / min. The tablets were dried for one hour at an air inlet temperature of 45 ° C (temperature range 40-50 ° C).
VIII. Enteric Coating: The undercoated drug product rasagiline tablet formulation described in the previous chapter was used for the enteric coated.
1. Preparation of Eudragil® L100-55 dispersion:
Triethyl citrate was mixed with water for 15 minutes. The extra-fine talc was added to the triethyl citrate and water dispersion in an Ultraturax over a period of 10 minutes.
Eudragit® L100-55 was added to the triethyl citrate / talc dispersion, filtered and stirred until proceeding.
[00290] 2. preheating:
The tablet cores are placed in an Ohara coater coating paddle and the tablets were placed at an air inlet temperature of 50 ° C. ATP 508U1 2012-06-15 (45 ° to 55 ° C) and an air outlet temperature of 45 Ό (40 ° C) heated to 50 ° C).
[00291] 3. Spray Method
The tablet cores were sprayed with the dispersion in an Ohara coating paddle. The air inlet temperature was 45 ^: the air outlet temperature was 35 ° C (temperature range 30-40 ^). The blade speed was set at 16 rpm (in the range of 14-18 rpm) and the spray rate was 5-20 g / min. The tablets were dried for two hours at an air inlet temperature of 50 ° C (temperature range 45-55 ° C) at minimum blade speed.
Example 6b -1.0 mg rasagiline base, 76 mg tablet core weight. This formulation was prepared using similar steps as described in Example 6a.
Table 6b: Composition of enteric-coated rasagiline base tablets with delayed release
Component Function Per tablet (mg) Tablet cores Rasagiline Base Remedy Substance 1.0 Maleic Acid Antioxidant / Stabilizer 1.6 Mannitol Filler 45.0 Aerosil Solvent 0.4 Starch NF Binder 5.0 Pregelatinized Starch (Thickness STA-RX 1500) Disintegrant 20 , 0 talc lubricant 1.5 stearic acid lubricant 1.5 total weight of tablet core 76.0 subcoating Pharmacoat® 606 (Hypromello-se USP) granules coating agent 3.5 purified water processing aid coating suspension Eudragit® L-30D-55 coating agent 4.0 * talc USP extrafine Lubricant 1,9 Triethyl Citrate NF Plasticizer 0,8 Purified Water Processing Aid Total weight of tablet 86,2 * Dry matter remaining on the core.
Example 6c - 0.5 mg rasagiline base, 117 mg tablet core weight [00294] This formulation was prepared using similar steps as described in Example 6a. 30/70 Austrian Patent Office AT12 508U1 2012-06-15 [00295] Table 6c: Composition of enteric-coated rasagiline base tablets with delayed release
Component Function Per tablet (mg) Tablet cores Rasagiline base Medicinal substance 0.5 Maleic acid Antioxidant / Stabilizer 1.6 Mannitol Filler 80.34 Aerosil Plasticizer 0.6 Starch NF Binder 10.0 Starch, pre-gelatinized (starch STA-RX 1500) Disintegration agent 20 , 0 talc lubricant 2.0 stearic acid lubricant 2.0 total weight of tablet core 117.0 subcoating Pharmacoal® 606 (Hypromello-se USP) granules coating agent 4.8 purified water processing aid coating suspension Eudragit® L-30D-55 coating agent 6.25 * talc USP Extra Fine Lubricant 3,1 Triethyl Citrate NF Plasticizer 1,25 Purified Water Processing Aid Total weight of tablet 132,4 * Dry matter remaining on the core.
Example 6d. Preparation of an enteric coated sustained release tablet with 0.5 mg rasagiline base In this example, an enteric coated sustained release tablet was administered with 0.5 mg rasagiline base containing maleic acid (76 mg tablet core weight) using similar steps as described in Example 6a.
Table 6d: Composition of enteric-coated rasagiline base tablets with delayed release
Component Function Per tablet (mg) Tablet cores Rasagiline base Remedy substance 0.5 Maleic acid Antioxidant / Stabilizer 1.6 Mannitol Filler 45.5 Aerosil Plasticizer 0.4 Starch NF Binder 5.0 Pregelatinized starch (Thickness STA-RX 1500) Disintegrant 20 , 0 talc lubricant 1.5 stearic acid lubricant 1.5 total weight of the tablet core 76.0 31/70 AT12 508U1 2012-06-15 Austrian
Patent Office
Undercoating Pharmacoat® 606 (Hypromello-se USP) Granules Coating Composition 3.5 Purified Water Processing Aid Coating Suspension Eudragit® L-30D-55 Coating 4.0 * Talc USP Extra Fine Lubricant 1.9 Triethyl Citrate NF Plasticizer 0.8 Purified Water Processing Aid Total Weight Tablet 86.2 * Dry matter remaining on the core.
EXAMPLE 7. STABILITY RESULTS OF TABLETS MANUFACTURED ACCORDING TO EXAMPLE 6A
Stability of enteric-coated tablets prepared using citric acid-containing formulations was tested under various storage conditions. The results are summarized below.
Stability Results (Accelerated Conditions): [00300] According to the USP specification for sustained-release articles (enteric coated) 29th Edition, Chapter 724, the dissolution profile of the enteric-coated tablets in 0.1 N HCl was acceptable 10% release after 120 minutes.
The following tables show the analysis results for tablets under different storage conditions.
[00302] Tablet cores:
Conditions Sample% Total Incidence T = 0 101.2 <NG 40 <€, 75rF 1 month 101.1 0.1 2 months 98.3 0.3 3 months 93.3 0.5 4 months 93.1 0.4 30'C, 65rF 1 month 101.4 <NG 2 months 101.9 <QD 3 months 98.3 <QD 25 ° C, 60 rF 1 month 101.5 <NG 2 months 102.0 &lt; QG 3 months 100.3 &lt; QG
Enteric coated tablets:
Conditions Sample% Total Interferences T = 0 98.2 <QG 40 ° C, 75 rF 1 month 100.5 0.2 2 months 96.4 0.3 3 months 96.6 0.5 30 ° C, 65 rF 1 Month 98.2 <QG 2 months 100.2 <QG 3 months 101.0 0.1 25 ° C, 60 rF 1 month 101.5 <QG 2 months 96.7 <QG 3 months 99.5 &lt; QG 32/70 Austrian Patent Office AT12 508U1 2012-06-15 N- (2-chloroallyl) -1 (R) -aminoindane (2-CI-AAI) impurities
Batch number 2-CI-AAI content,% 1 &lt; 0.00004
EXAMPLE 8. PREPARATION OF RASAGILIN BASE TABLET CORE WITH CITRIC ACID
Starting material mg / tablet Percentage Part I, granulating solution Citric acid 1,6 2,0 Rasagiline base 1 1,25 Purified water 12,35 15,44 Part II Mannitol 48,5 60,63 Aerosil 200 0,18 0,22 Starch NF / BP 6.1 7.62 Pregelatinized starch NF / Ph. Eur 20.0 25.0 Part III Aerosil 200 0.18 0.22 part IV Stearic acid 1.22 1.52 Talc 1.22 1.52 Total: 80 The above composition can also be used to prepare rasagiline base tablets with maleic acid by replacing the citric acid with the same amount of maleic acid.
Calculated amounts of external excipients according to the actual amount of granules: mg / tablet Starting material Percentage Part III Granules 0.18 Aerosil 200 0.22 part IV 1.22 Stearic acid 1.52 1.22 Talc 1.52 [00307] I Preparation of Granulating Solution: [00308] 1. 80% of the required amount of purified water is poured into a glass.
2. In the same glass, the citric acid is weighed.
[00310] 3. A stirrer is inserted into the glass and stirring is started for about 5-10 minutes until complete dissolution.
[00311] 4. Rasagiline base is weighed and gege ben in the resulting citric acid solution.
[00312] 5. Stirring is continued for about 30 minutes until complete dissolution of API.
[00313] II. Preparation of Granules: [00314] 1. Mannitol, Aerosil 200, starch and pregelatinized starch are weighed and 33/70 Austrian Patent Office [00315 [00316 [00317 [00318 [00319 [00320 [00321 [00322 [00323 00324 [00325 [00329 [00329 [00329 [00330 [00331 [00332 [00333 [00334 [00335 [00336 [00337 [00338 [00339 [00340 [00341 [00342 [00343 AT 12 508 Ul 2012-06-15 all excipients transferred to Diosna P-6 (Diosna) and mixed for 1 minute with Mixer I (270 rpm). 2. The excipient is mixed for an additional minute with Mixer I (270 rpm) and Chopper I (1500 rpm). 3. Granulation solution is added to the Diosna P-6 (Diosna) and mixed for 2 minutes with Mixer II (540 rpm) and Chopper II (2200 rpm). 4. The glass is cleaned after the granulating solution with 46.563 g of purified water and it is placed in the Diosna P-6 (Diosna). 5. Mix for 2 minutes with Mixer II (540 rpm) and Chopper II (2200 rpm). 6. The granules obtained are dried in the Glatt 1.1 (fluidized bed) to dry at 37 ° C inlet air to a residual moisture content of not more than (orig .: L.O.D. NMT) 1.5%. T rocking conditions:
Admission: Min. - 35 ° C; Target - δΟ'Ό; Max. - 55 ° C Outlet: product temperature - 37 ° C Flow: min. - 25; Goal - 60; Max. -1000 III. crushing:
The granules are crushed through a 0.6 mm sieve using Frewitt. IV. Final mixing: 1. The amount of granules obtained is weighed. 2. The amounts of Aerosil 200, stearic acid and talc are calculated according to the actual weight of the granules. 3. Aerosil 200 is screened through a 50 mesh screen. 4. The required amount of Aerosil 200 is weighed after sieving. 5. The crushed granules and the Aerosil 200 are transferred to the Y-cone after sieving. 6. It is mixed for 2 minutes. 7. Stearic acid and talc are weighed. 8. These excipients are sieved through a 50 mesh sieve. 9. They are transferred to the Y-cone. 10. It is mixed for 5 minutes. V. tabletting:
Machine: Sviac
Diameter of the punch: 5.0 mm (may vary by ± 10%)
Tablet weight: 80 mg ± 5%
Hardness: 3-7 kP
Friability: not more than 1%
Disintegration: not more than 5 minutes 34/70 Austrian Patent Office AT12 508U1 2012-06-15
EXAMPLE 9. PREPARATION OF RASAGILIN BASE TABLET CORE WITH MALEACIC ACID
Starting material mg / tablet Percentage Part I, granulating solution Maleic acid 1.6 3.72 Rasagiline base 1 2.33 part II Mannitol 25.8 60.0 Aerosil 200 0.1 0.24 Starch NF / BP 3.0 6.98 Pregelatinized starch NF / Ph. Eur 10.0 23.26 Part III Aerosil 200 0.1 0.23 part IV Stearic acid 0.7 1.63 Talc 0.7 1.63 Total 43.0 100 [00344] The above composition can also be used to make rasagiline base tablets with citric acid by replacing the maleic acid with the same amount of citric acid.
[00341] [00346] [00347] [00348] [00349] [00350] [00351] [00352] [00353] [00354] [00355] [00356] [00357] [00358] [00359] I. Preparation of granulating solution : 1. 80% of the required amount of purified water is weighed into a glass. 2. In the same glass, the maleic acid is weighed. 3. A stirrer is placed in the jar and stirring is started for about 5-10 minutes until completely dissolved. 4. Rasagiline base is weighed and added to the resulting maleic acid solution. 5. Stirring is continued for about 30 minutes until complete dissolution of API. II. Preparation of Granules: 1. Weigh out mannitol, Aerosil 200, starch and pregelatinized starch and transfer all excipients to Diosna P-10 (Diosna) and mix for 1 minute with Mixer I. 2. The excipient is mixed for an additional minute with Mixer I and Chopper I rpm. 3. The granulation solution is placed in the Diosna P-10 (Diosna) and mixed for 2 minutes with Mixer II and Chopper II. 4. Add additional purified water to the Diosna P-10 (Diosna) and mix for 2 minutes with Mixer II and Chopper II. 5. The granules obtained are dried in the Glatt 5 (fluidized bed) to dry at 37 ^ 0 inlet air to a residual moisture content of not more than 1.5%.
Drying conditions:
Admission: Min. - 35 ° C; Target - 50 * Ό; Max. - 55 ^ 0
Outlet: product temperature - 37 ° C 35/70 Austrian Patent Office AT 12 508 Ul 2012-06-15 [00360] III. Crushing: Aerosil 200 is weighed and added to the granules and the granules are minced through a 0.6 mm sieve using Frewitt.
[00362] IV. Final Mixing: [00363] 1. Stearic acid and talc are weighed.
2. The excipients are sieved through a 50-mesh sieve.
3. The crushed granules and sieved stearic acid and talc are transferred to the Y-cone.
[00366] 4. Mix for 5 minutes.
[00367] V. tabletting: [00368] machine: Sviac [00369] diameter of the punch: 4.0 mm (may vary by ± 10%) [00370] tablet weight: 43 mg ± 5%
Hardness: 3-5 kP
Friability: not more than 1% [00373] Decay: not more than 5 minutes
EXAMPLE 10. PREPARATION OF RASAGILIN BASE TABLET CORE WITH CITRIC ACID AND MALEIC ACID
Starting material mg / tablet Percentage Part I, granulating solution Citric acid 0.8 0.68 Maleic acid 0.8 0.68 Rasagiline base 1.0 0.85 part II Mannitol 79.8 68.2 Aerosil 200 0.3 0.26 Starch NF / BP 10.0 8.55 Pregelatinized starch NF / Ph. Eur 20.0 17.09 Part III Aerosil 200 0.3 0.26 part IV Stearic acid 2.0 1.71 Talc 2.0 1.71 Total 117 , 0 100 Calculated amounts of external excipients according to the actual amount of granules:
Starting material mg / tablet Percentage Part III Granules Aerosil 200 0.3 0.26 part IV Stearic acid 2.0 1.71 Talc 2.0 1.71 36/70 Austrian Patent Office AT 12 508 Ul 2012-06-15 [00375] I [00376] 1. [00377] 2. [00379] 3. [00379] 4.
[00381] II. [00382] 1.
[00383] 2.
[00385 [00386 III 1.
[00387 [00388 [00389 [00390 [00391 2. 3. 4. 5. 6.
Preparation of granulating solution 1: 80% of the required amount of purified water is weighed into a glass. The citric acid is weighed into the same glass. A stirrer is placed in the jar and stirring is started for about 5-10 minutes until completely dissolved. Rasagiline base is weighed and added to the resulting citric acid solution. Stirring is continued for about 30 minutes until complete dissolution of API. Preparation of granulating solution 2: 20% of the required amount of purified water is weighed into the glass. Into this glass is added the weighed amount of maleic acid. A stirrer is placed in the jar and stirring is started for about 5-10 minutes until completely dissolved. Preparation of granules: Mannitol, Aerosil 200, starch and pregelatinized starch are weighed and all excipients transferred to Diosna P-6 (Diosna) and mixed for 1 minute with Mixer I (270 rpm). The excipient is mixed for an additional minute with Mixer I (270 rpm) and Chopper I (1500 rpm). Granulation Solution 1 is added to the Diosna P-6 (Diosna) and mixed for 2 minutes with Mixer II (540 rpm) and Chopper II (2200 rpm). The glass is cleaned after the granulating solution 1 with granulating solution 2 and it is placed in the Diosna P-6 (Diosna). Mix for 2 minutes with Mixer II (540 rpm) and Chopper II (2200 rpm). The granules obtained are dried in the Glatt 1.1 (fluidized bed) for drying in 37 * Ό inlet air to a residual moisture content of not more than 1.5%.
Drying conditions: [00393] inlet: min. - 35 ° C; Target - 50 * Ό; Max. - 55 Exit: Product temperature - 37 ° C [00395] Flow: Min. - 25; Goal - 60; Max. -1000 IV. Crushing: The granules obtained are comminuted through a 0.6 mm sieve using Frewitt.
[00398] 1. [00399] 1. [00400] 2.
Final mixing: The granules obtained are weighed. The amounts of Aerosil 200, stearic acid and talc are calculated according to the actual weight of the granules.
3. Aerosil 200 is screened through a 50 mesh screen. [00404] [00404] [00405] [00406] [00407] [00408] [00409] [00410] [00411] [00412] [00413] [00414 ] [00415] [00416] 4. The required amount of Aerosil 200 is weighed after sieving. 5. The crushed granules and the Aerosil 200 are transferred to the Y-cone after sieving. 6. It is mixed for 2 minutes. 7. Stearic acid and talc are weighed. 8. These excipients are sieved through a 50 mesh sieve. 9. They are transferred to the Y-cone. 10. It is mixed for 5 minutes. VI. tableting:
Machine: Sviac
Diameter of the punch: 6.0 mm (may vary by ± 10%)
Tablet weight: 117 mg ± 5%
Hardness: 6-8 kP
Friability: not more than 1%
Disintegration: not more than 5 minutes VII. Subcoating: mq / T ablette starting material 4.8 Pharmacoat 606 (Hypromellose USP) [00417] Apparatus: O'HARA, peristaltic pump [00418] 1. Preparation of Undercoating Solution:
Pharmacoat 606 (Hypromellose USP) was added to the jar along with 1510 g of purified water and mixed for 30 minutes using a stirrer.
[00419] 2. Heating:
The tablet cores were placed in the 2.5 kg bucket of the O'HARA coater and preheated:
Air inlet temperature - 50 ° C (45 ° to 55 ° C)
Air outlet temperature - 45 ° C (40 ° to 50 ° C)
Pressure difference -50 Pa [00420] 3. Spray method (the process was continued until the desired tablet weight was reached):
Undercoating solution was sprayed on the preheated tablet cores under the following conditions:
Number of sprayers -1 nozzle hole - 1 mm
Distance tablet bed / spray device -15 cm bucket speed 10 rpm (8-12 rpm)
Air inlet temperature - 50 ° C (45 ° to 55 ° C)
Air outlet temperature - 35 ° C (30 ° to 40 ° C)
Spray rate -10-20 g / min pressure difference - -50 Pa atomization air pressure - 30 psi forming air pressure - 30 psi [00421] 4. Drying process:
Air inlet temperature - 45 ° C (40 ° to 50 ° C) 38/70 Austrian Patent Office AT12 508U1 2012-06-15
Air outlet temperature - 40 ° C-50 ° C Blade speed - 5 rpm Jogging operation Drying time - 60 min
EXAMPLE 11. ADDITIONAL ADHESIVE-COATED RASAGILINE-BASE FORMULATIONS WITH CITRIC ACID
Example 11a - 0.5 mg rasagiline base [00422] This example describes 0.5 mg rasagiline base formulations with variations in the amount of citric acid and other excipients. These formulations have a dissolution and pharmacokinetic profile (Cmax and AUC) similar to those of Example 1.
Component Function Per tablet (mg) Per tablet (mg) Per tablet (mg) Per tablet (mg) Tablet cores Rasagiline base Remedy substance 0.5 0.5 0.5 0.5 Citric Acid Antioxidant 1.6 or 0.8 1 , 6 or 0.8 1,6 or 0,8 1,6 or 0,8 mannitol filler 45,5 68,3 50,5 80,3 Aerosil flow agent 0,4 0,6 0,4 0,6 starch NF Binder 5.0 10.0 5.0 10 Pre-gelatinized starch (starch STA-RX 1500) disintegrant 20.0 20.0 15.0 20.0 talc lubricant 1.5 2.0 1.5 2.0 stearic acid lubricant 1 , 5 2.0 1.5 2.0 Total weight of the tablet core 76.0 (± 10%) 105.0 (± 10%) 76.0 (± 10%) 117.0 (± 10%) Undercoating Pharmacoat 606 ( Hypromellose USP) Granules Coating Medium 3.5 (± 10%) 4.8 (± 10%) 3.5 (± 10%) 4.8 (± 10%) Purified Water Processing Aid Coating Suspension Eudragit L-30D-55 Coating Composition 4, 0 (± 10%) 4.0 (± 10%) 4.0 (± 10%) 6.25 (± 10%) talc USP extrafine lubricant 1.9 (± 10%) 1.9 (± 10%) 1.9 (± 1 0%) 3.1 (± 10%) triethyl citrate NF plasticizer 0.8 (± 10%) 0.8 (± 10%) 0.8 (± 10%) 1.25 (± 10%) Purified water processing aids
Example 11b - 1.0 mg rasagiline base [00423] This example describes 1 mg rasagiline base formulations with variations in the amount of citric acid and other excipients. These formulations have a dissolution and pharmacokinetic profile (Cmax and AUC) similar to those of Example 1.
Component Function Per tablet (mg) Per tablet (mg) Per tablet (mg) Per tablet (mg) Tablet cores Rasagiline base Medicinal substance 1.0 1.0 1.0 1.0 Citric Acid Antioxidant 1.6 or 1.6 or 1,6 or 1,6 or 39/70 AT 12 508 Ul 2012-06-15 Austrian
Patent Office 0,8 0,8 0,8 0,8 Mannitol Filler 45,0 67,8 50,0 79,8 Aerosil Plasticizer 0,4 0,6 0,4 0,6 Starch NF Binder 5,0 10,0 5.0 10.0 Pre-gelatinized starch (starch STA-RX 1500) disintegrant 20.0 20.0 15.0 20.0 talc lubricant 1.5 2.0 1.5 2.0 stearic acid lubricant 1.5 2.0 1.5 2.0 Total weight of tablet core 76.0 (± 10%) 105.0 (± 10%) 76.0 (± 10%) 117.0 (± 10%) Undercoating Pharmacoat 606 (Hypromellose USP) Granules Coating 3.5 (± 10%) 4.8 (± 10%) 3.5 (± 10%) 4.8 (± 10%) Purified Water Processing Aid Coating Suspension Eudragit L-30D-55 Coating 4.0 (± 10%) ) 4.0 (± 10%) 4.0 (± 10%) 6.25 (± 10%) talc USP extrafine lubricant 1.9 (± 10%) 1.9 (± 10%) 1.9 (± 10%) 3.1 (± 10%) triethyl citrate NF plasticizer 0.8 (± 10%) 0.8 (± 10%) 0.8 (± 10%) 1.25 (± 10%) Purified water processing aids
EXAMPLE 12. ADDITIONAL MAGENTATE-RESISTANT COATED RASAGILINE-BASE FORMULATIONS WITH MALEACIC ACID
Example 12a - 0.5 mg rasagiline base [00424] This example describes 0.5 mg rasagiline base formulations with variations in the amount of maleic acid and other drug carriers. These formulations have a dissolution and pharmacokinetic profile (Cmax and AUC) similar to those of Example 1.
Component Function Per tablet (mg) Per tablet (mg) Per tablet (mg) Per tablet (mg) Tablet cores Rasagiline base Remedy substance 0.5 0.5 0.5 0.5 Maleic Antioxidant 1.6 or 0.8 1, 6 or 0.8 1,6 or 0,8 1,6 or 0,8 mannitol filler 45,5 68,3 50,5 80,3 Aerosil flow agent 0,4 0,6 0,4 0,6 starch NF binder 5.0 10.0 5.0 10 Pre-gelatinized starch (starch STA-RX 1500) disintegrant 20.0 20.0 15.0 20.0 talc lubricant 1.5 2.0 1.5 2.0 stearic acid lubricant 1, 5 2.0 1.5 2.0 Total weight of the tablet core 76.0 (± 10%) 105.0 (± 10%) 76.0 (± 10%) 117.0 (± 10%) Undercoating Pharmacoat 606 (Hypromellose USP) Granules Coating Material 3.5 (± 10%) 4.8 (± 10%) 3.5 (± 10%) 4.8 (± 10%) Purified Water Processing Aids 40/70 AT12 508U1 2012-06-15 Austrian
Patent Office
Coating suspension Eudragit L-30D-55 Coating agent 4.0 (± 10%) 4.0 (± 10%) 4.0 (± 10%) 6.25 (± 10%) Talc USP extrafine Lubricant 1.9 (± 10%) %) 1.9 (± 10%) 1.9 (± 10%) 3.1 (± 10%) triethyl citrate NF plasticizer 0.8 (± 10%) 0.8 (± 10%) 0.8 (± 10%) 1.25 (± 10%) Purified Water Processing Aid
Example 12b - 1.0 mg rasagiline base [00425] This example describes 1 mg rasagiline base formulations with variations in the amount of maleic acid and other excipients. These formulations have a dissolution and pharmacokinetic profile (Cma * and AUC) similar to those of Example 1.
Component Function Per tablet (mg) Per tablet (mg) Per tablet (mg) Per tablet (mg) Tablet cores Rasagiline base Medicinal substance 1.0 1.0 1.0 1.0 Maleic acid Antioxidant 1.6 or 0.8 1, 6 or 0.8 1.6 or 0.8 Mannitol filler 45.0 67.8 50.0 79.8 Aerosil flow agent 0.4 0.6 0.4 0.6 starch NF binder 5.0 10.0 5.0 10.0 Pre-gelatinized starch (starch STA-RX 1500) disintegrant 20.0 20.0 15.0 20.0 talc lubricant 1.5 2.0 1.5 2.0 stearic acid lubricant 1.5 2.0 1.5 2.0 Total weight of the tablet core 76.0 (± 10%) 105.0 (± 10%) 76.0 (± 10%) 117.0 (± 10%) Undercoating Pharmacoat 606 (Hypromellose USP) Granules Coating Medium 3.5 (± 10%) 4.8 (± 10%) 3.5 (± 10%) 4.8 (± 10%) Purified Water Processing Aid Coating Suspension Eudragit L-30D-55 Coats 4.0 (± 10%) 4.0 (± 10%) 4.0 (± 10%) 6.25 (± 10%) talc USP extrafine lubricant 1.9 (± 10%) 1.9 (± 10%) 1.9 (± 10%) 3, 1 (± 10%) triethyl citrate NF plasticizer 0.8 (± 10%) 0.8 (± 10%) 0.8 (± 10%) 1.25 (± 10%) Purified water Processing aid
EXAMPLE 13. ADDITIONAL ADHESIVE-COATED RASAGILINE-BASE FORMULATIONS WITH CITRIC ACID AND MALEIC ACID
Example 13a - 0.5 mg rasagiline base [00426] This example describes 0.5 mg rasagiline base formulations with variations in the amount of citric acid, maleic acid and other excipients. These formulations have a dissolution and pharmacokinetic profile (Cmax and AUC) equal to those of 41/70 Austrian Patent Office AT12 508U1 2012-06-15
Example 1 is similar.
Component Function Per tablet (mg) Per tablet (mg) Per tablet (mg) Per tablet (mg) Tablet cores Rasagiline base Medicinal substance 0.5 0.5 0.5 0.5 Maleic acid Antioxidant 0.8 or 0.4 0, 8 or 0.4 0.8 or 0.4 1.6 or 0.8 citric acid antioxidant 0.8 or 0.4 0.8 or 0.4 0.8 or 0.4 1.6 or 0.8 mannitol Fillers 45.5 68.3 50.5 80.3 Aerosil Flowing Agents 0.4 0.6 0.4 0.6 Starch NF Binder 5.0 10.0 5.0 10.0 Pre-gelatinized starch (starch STA-RX 1500 ) Disintegrant 20.0 20.0 15.0 20.0 talc lubricant 1.5 2.0 1.5 2.0 stearic acid lubricant 1.5 2.0 1.5 2.0 total weight of tablet core 76.0 (± 10%) 105.0 (± 10%) 76.0 (± 10%) 117.0 (± 10%) Undercoating Pharmacoat 606 (Hypromellose USP) Granules Coating Composition 3.5 (± 10%) 4.8 (± 10%) %) 3.5 (± 10%) 4.8 (± 10%) Purified Water Processing Aid Coating Suspension Eudragit L-30D-55 Coating 4.0 (± 10%) 4.0 (± 10%) 4.0 (± 10%) 6.25 (± 10%) talc USP extrafine lubricant 1.9 (± 10%) 1.9 (± 10%) 1.9 (± 10%) 3.1 (± 10%) triethyl citrate NF plasticizer 0.8 (± 10%) 0.8 (± 10%) 0.8 (± 10%) 1.25 (± 10%) Purified water Processing aid
Example 13b - 1.0 mg rasagiline base [00427] This example describes 1 mg rasagiline base formulations with variations in the amount of citric acid, maleic acid and other excipients. These formulations have a dissolution and pharmacokinetic profile (Cmax and AUC) similar to those of Example 1.
Component Function Per tablet (mg) Per tablet (mg) Per tablet (mg) Per tablet (mg) Tablet cores Rasagiline base Medicinal substance 1.0 1.0 1.0 1.0 Maleic acid Antioxidant 0.8 or 0.4 0, 8 or 0.4 0.8 or 0.4 1.6 or 0.8 citric acid antioxidant 0.8 or 0.4 0.8 or 0.4 0.8 or 0.4 1.6 or 0.8 mannitol Filler 45.0 67.8 50.0 79.8 Aerosil Plasticizer 0.4 0.6 0.4 0.6 Starch NF Binder 5.0 10.0 5.0 10.0 Pre-gelatinized starch (starch STA-RX 1500 ) Disintegrating agents 20.0 20.0 15.0 20.0 42/70 AT 12 508 U1 2012-06-15 Austrian
Patent Office
Talc lubricant 1.5 2.0 1.5 2.0 stearic acid lubricant 1.5 2.0 1.5 2.0 total weight of tablet core 76.0 (± 10%) 105.0 (± 10%) 76.0 (± 10%) 117.0 (± 10%) Undercoating Pharmacoat 606 (Hypromellose USP) Granules Coating Composition 3.5 (± 10%) 4.8 (± 10%) 3.5 (± 10%) 4.8 ( ± 10%) Purified Water Processing Aid Coating Suspension Eudragit L-30D-55 Coating 4.0 (± 10%) 4.0 (± 10%) 4.0 (± 10%) 6.25 (± 10%) talc USP extrafine Lubricant 1.9 (± 10%) 1.9 (± 10%) 1.9 (± 10%) 3.1 (± 10%) Triethyl Citrate NF Plasticizer 0.8 (± 10%) 0.8 ( ± 10%) 0.8 (± 10%) 1.25 (± 10%) Purified water Processing aids
EXAMPLE 14. POWDERED COATED RASAGILINE BASE FORMULATION WITH CITRIC ACID, COLORED COATED
Example 14a - 0.5 mg rasagiline base [00428] This example describes a 0.5 mg rasagiline base formulation containing citric acid with a colored extra coating.
Component Function Per tablet (mg) Tablet cores Rasagiline Base Medicinal substance 0.5 Citric Acid Antioxidant 1.6 Mannitol Filler 80.3 Aerosil Plasticizer 0.6 Pre-gelatinized starch (starch STA-RX 1500) Disintegrant 20.0 Starch NF Binder 10.0 Talc Lubricant 2.0 Stearic acid Lubricant 2.0 Total weight of tablet core 117.0 Undercoat Pharmacoat 606 (Hypromellose USP) Granules Coating agent 4.8 Purified water Processing aid Coating suspension Eudragit L-30D-55 Coating agent 6.25 * talc USP extrafine lubricant 3.1 Triethyl -Citrate NF Plasticizer 1.25 Purified Water Processing Aids 43/70 Austrian Patent Office AT12 508U1 2012-06-15
Top coat OPADRY II OY-GM-28900 WHITE (Catalog Number 415850005) or OPADRY II Y-30-18037 WHITE (Catalog Number 415880719 and / or OPADRY fx 63f97546 silver Coating Composition 1-5 Purified Water Processing Aid * Dry matter remaining on the core.
Example 14b - 1.0 mg rasagiline base [00430] This example describes a 1 mg rasagiline base formulation containing citric acid with a colored extra coating.
Component Function Per tablet (mg) Tablet cores Rasagiline Base Remedy Substance 1.0 Citric Acid Antioxidant 1.6 Mannitol Filler 79.8 Aerosil Plasticizer 0.6 Pregelatinised Starch (Thickness STA-RX 1500) Disintegrator 20.0 Starch NF Binder 10.0 Talc Lubricant 2.0 Stearic acid Lubricant 2.0 Total weight of tablet core 117.0 Undercoat Pharmacoat 606 (Hypromellose USP) Granules Coating agent 4.8 Purified water Processing aid Coating suspension Eudragit L-30D-55 Coating agent 6.25 * talc USP extrafine lubricant 3.1 Triethyl -Citrate NF Plasticizer 1.25 Purified water Processing aid Top coat OPADRY® II 31F20721 Blue or OPADRY® II 34G24627 Pink and / or Opadry fx 63f97546 silver Coating materials 1-5 Purified water Processing aids * Dry matter remaining on the core. 44/70 Austrian Patent Office AT 12 508 Ul 2012-06-15
Example 14c - Rasagiline base (Formulation III with colored coating) [00431] This example describes a Rasagiline base formulation (Formulation III) containing citric acid with a colored extra coating.
Component Function Per tablet (mg) -0.5 mg Rasagiline formulation Per tablet (mg) -1.0 mg Rasagiline formulation Tablet cores Rasagiline base Medicinal substance 0.5 1 Citric Acid Antioxidant 1.6 1.6 Mannitol Filler 45.5 45 Aerosil Plasticizer 0.4 0.4 Starch NF Binder 5 5 Pregelatinised Starch (Thickness STA-RX 1500) Disintegrant 20 20 Talc Lubricant 1.5 1.5 Stearic Acid Lubricant 1.5 1.5 Total Weight of Tablet Core 76 76 Undercoating Pharmacoat 606 (Hypromeless USP) Granules Coating Composition 3.5 3.5 Coating Suspension Eudragit L-30D-55 Coating Composition 4 4 Talc USP Extra Fine Lubricant 1.9 1.9 Triethyl Citrate NF Plasticizer 0.8 0.8 Overcoat Opadry® Coating Composition 2 2 total weight of the tablet 88.2 88.2
EXAMPLE 15. EMULSIVE-COATED RASAGILINE-BASE-FORMULATION-RUNG WITH MALEIC ACID, COLORED COATED
Example 15a - 0.5 mg rasagiline base [00432] This example describes a 0.5 mg rasagiline base formulation containing maleic acid with a colored extra coating.
Component Function Per tablet (mg) Tablet cores Rasagiline base Remedy substance 0.5 Maleic acid Antioxidant 1.6 Mannitol Filler 80.3 Aerosil Plasticizer 0.6 Pregelatinized starch (starch STA-RX 1500) Disintegrant 20.0 Starch NF Binder 10.0 talc Lubricant 2.0 Stearic acid Lubricant 2.0 Total weight of the tablet core 117.0 Undercoating Pharmacoat 606 (Hypromellose coating agent 4.8 45/70 AT12 508U1 2012-06-15 Austrian
Patent Office USP) Granules Purified Water Processing Aid Coating Suspension Eudragit L-30D-55 Coating Material 6.25 * Talc USP Extra Fine Lubricant 3.1 Triethyl Citrate NF Plasticizer 1.25 Purified Water Processing Aid Topcoat OPADRY II OY-GM-28900 WHITE (Catalog Number 415850005) or OPADRY II Y-30-18037 WHITE (catalog number 415880719) and / or OPADRY fx 63f97546 silver Coating materials 1-5 Purified water Processing aids * Dry matter remaining on the core.
Example 15b - 1.0 mg rasagiline base [00433] This example describes a 1.0 mg rasagiline base formulation containing maleic acid with a colored extra coating.
Component Function Per tablet (mg) Tablet cores Rasagiline Base Remedy Substance 1.0 Maleic Acid Antioxidant 1.6 Mannitol Filler 79.8 Aerosil Plasticizer 0.6 Pregelatinised Starch (Thickness STA-RX 1500) Disintegrant 20.0 Starch NF Binder 10.0 Talc Lubricant 2.0 Stearic acid Lubricant 2.0 Total weight of the tablet core 117.0 Undercoating Pharmacoat 606 (Hypromellose USP) Granules Coating agent 4.8 Purified water Processing aid Coating suspension Eudragit L-30D-55 Coating powder 6.25 * talc USP extrafine lubricant 3.1 Triethyl Citrate NF Plasticizer 1.25 Purified Water Processing Auxiliaries Topcoat OPADRY® II 31F20721 Blue or OPADRY® II 34G24627 Pink and / or Opadry fx 63f97546 silver Primer 1-5 Purified Water Processing Aid * Dry matter remaining on the core. 46/70 Austrian Patent Office AT12 508U1 2012-06-15
EXAMPLE 16. EXTRACTION OF RASAGILIN BASE FROM TABLETS
This example evaluated the amount of free rasagiline base in 1 mg tablets of citric acid formulations.
It is believed that rasagiline is present in the formulation in salt form or as the free base.
Rasagiline base is a nonpolar compound, very soluble in non-polar organic solvents such as hexane, toluene and ethyl acetate. Therefore, free rasagiline base could be extracted from the solid formulation by these solvents.
Rasagiline salts are not soluble in non-polar solvents and the likelihood of extraction of rasagiline citrate with hexane, toluene, 1-octanol or ethyl acetate is very low.
Tablet cores were tested with rasagiline prepared using the steps described in Example 9. Each tablet contained 1 mg of rasagiline base. Placebo tablets were used as a reference.
17 tablet cores each with 1 mg of rasagiline base were broken in a mortar to homogeneous fine powder and crushed.
Each powder was mixed with 20 ml of organic solvent and stirred for 1 hour at room temperature in a closed glass vessel with a magnetic stirrer. The mixture was then allowed to settle without stirring, the clear liquid was decanted off and a sample of the resulting extract was filtered through a 0.2 μ filter.
The filtered samples of the extracts were subjected to HPLC analysis for the amount of dissolved rasagiline. Samples of placebo extracts were used as controls.
The maximum possible calculated concentration of rasagiline base in the extracts is 0.85 mg / ml (17 mg in 20 ml solvent).
The results are summarized in the following Table 16 (orig .: 5).
Table 16. Extraction of rasagiline base from tablet cores with organic solvents
Experiment No. Number of Tablets Weight of the Tablets Solvent Achieved Concentration of Rasagiline in the Extract, mg / ml 1 17 2.02 Toluene 0.01 2 17 2.02 n-Hexane 0.01 3 17 2.02 DCM 0.01 4 17 2.03 1 -Octanol 0.01 5 17 2.02 Ethyl acetate 0.02 [00445] Summary of Results The experimental results in Table 16 show that the tablets have cores of &quot; citric acid &quot; Formulation of rasagiline base may contain 1 to 2 percent of the free rasagiline base extractable with nonpolar solvents.
The amount of the extractable base is not dependent on the solvent type of the nonpolar solvent such as n-hexane, toluene, 1-octanol and dichloromethane. However, a more polar solvent such as ethyl acetate extracted more rasagiline base from the tablet cores.
EXAMPLE 17. CLINICAL STUDY BASED ON TABLETS ACCORDING TO EXAMPLES 3A AND 3B
This study evaluated the bioavailability of two different enteric-coated tablet formulations with 1 mg of rasagiline base prepared according to Examples 47/70 Austrian Patent Office AT12 508U1 2012-06-15 3a (Formulation I) and 3b (Formulation III) to the marketed Rasagiline drug product (AZILECT® 1 mg) following a single-dose administration to estimate the effect of foods on each of the test formulations.
[00449] This study also assessed the safety and tolerability of each treatment.
[00450] 1. Study Design This study was a flexible two part protocol, each part testing the bioavailability of a different enteric coated 1 mg rasagiline base formulation (formulation I or formulation III) against the reference product (AZILECT® 1 mg) ,
Each part was a non-allied, three-period, three-sequence, comparative crossover study with 15 healthy men and women (5 per sequence).
Treatment A: An enteric-coated rasagiline base tablet 1 mg (Test Formulation I or Test Formulation III) in the fasted state.
Treatment B: An AZILECT® tablet (reference 1 mg rasagiline as rasagiline mesylate) in the fasted state.
Treatment C: An enteric-coated rasagiline base tablet 1 mg (Test Formulation I or Test Formulation III) following a standard high fat and high calorie meal.
The three treatments were administered during three study periods, each separated by a 14-day washout interval.
Administration to the subjects was according to one of three sequences to which they were randomly assigned: A-B-C, B-C-A or C-A-B.
In each period, subjects were enrolled for two overnight stays [at least 10.5 hours before and until administration of the dose]. Subjects returned for day 2 ambulatory blood sampling (36 hours).
In part 1 subjects received 1-15 test formulation I or reference, while in part 2 subjects received 16-30 test formulation III or reference. The decision to proceed with each part of the study was based on the availability of the test formulation.
AEs, vital signs, medical examination and clinical laboratory tests were used to assess safety and blood samples were taken during the study at scheduled, pre-defined times for the measurement of plasma rasagiline and aminoindan concentrations.
[00461] 2. Selection of Subjects Thirty (30) healthy adult (~ 50% / 50% male and female) subjects were selected from non-institutional subjects consisting of members of the community in their entirety.
3. Pharmacokinetic (PK) Sampling and Analysis [00464] A total of 80 samples (approximately 400 ml) were taken from each subject for PK purposes. Pharmacokinetic sampling took place at the following times: [00465] a) Treatment A (fasting):
Day 1 within 90 minutes before dose (hour 0) and after dose administration at 0.5, 0.75, 1, 1.33, 1.67, 2, 2.33, 2.67, 3, 3 , 33, 3, 67, 4, 4.5, 5, 6, 7, 8, 9, 12, 24 and 36 hours (22 samples).
[00466] b) Treatment B (reference, fasting):
Day 1 within 90 minutes prior to dosing (hour 0) and after dosing by 0.25, 0.5, 0.75, 1, 1.25, 1.5, 2, 3, 4, 5, 6 , 7, 8, 12, 24 and 36 hours (17 samples). 48/70 Austrian Patent Office [00467] c) AT 12 508 Ul 2012-06-15
Treatment C (test, saturated):
Day 1 within 90 minutes before dose (hour 0) and after dose administration at 1, 1.5, 2, 2.5, 3, 3.33, 3.67, 4, 4.33, 4.67 , 5, 5, 33, 5, 67, 6, 6, 33, 6, 67, 7, 7, 33, 7, 67, 8, 8, 5, 9, 10, 11, 12, 13, 14, 15 , 16, 18, 19, 20, 21, 22, 23, 24, 25, 26 and 36 hours (41 samples).
Blood was taken either by direct venipuncture or by an intravenous indwelling cannula. Whenever the latter was used, the cannula was rinsed with 1.5 ml of physiological saline after each sampling. In addition, to avoid sample dilution, 1 ml of blood was discarded before the next sample (as long as the cannula was in place). Thus, up to 5 ml of blood was collected at each time point. The total blood volume drawn per subject for pharmacokinetic sampling was approximately 400 ml over a four-week period.
Samples were collected in K2-EDTA Vacutainer of appropriate volume. The labels for all biological sample collection and storage containers contained at least the protocol number, substudy number, subject number; Dosing period; dosing day; PK time. Immediately after sampling, the samples were mixed by inverting the blood collection tube at least two to three times. The samples were cooled by an ice bath or by a cooling device until further processing. Further processing of the blood took place within two hours of sampling: the sample was centrifuged at approximately 2000 g and 4 ° C (± 3 ° C) for approximately 10 minutes, the plasma was transferred to suitably labeled duplicate polypropylene tubes , and stored at about -20 ° C until transfer or transport to the bioanalytical laboratory. At least 0.7 ml of plasma was transferred to the first polypropylene tube and the remainder of the plasma was transferred to the second polypropylene tube. The time at which samples were introduced at -20 ^ was recorded in the study documentation.
The actual sampling time was recorded directly in the data source or CRF. Sample processing procedures were documented in the PK logbook.
The rasagiline and aminoindan plasma concentrations were measured using a validated bioanalytical LC / MS / MS method and according to Bioanalytical Laboratory's Standard Operating Procedures and FDA Guidelines.
Analysis of PK data of each substudy was performed separately according to verified availability of bioanalytical data. The individual plasma concentrations of rasagiline and aminoindan were recorded in a list, graphically displayed as appropriate, and summarized using descriptive statistics for each of the treatments.
Pharmacokinetic analysis was performed on rasagiline and aminoindan concentration profiles using suitable non-compartmental methods.
The following parameters were calculated: Cmax, Tmax, Τ, ^, AUC ,, AUC ", t1 / 2, CL / F, V / F,% AUCext, regression coefficient of the terminal slope. Additional PK parameters were calculated if deemed necessary. All PK parameters were listed and summarized using descriptive statistics.
Statistical analysis was performed using SAS for each substudy based on the reception of the data. For each substudy, bioequivalence between the test and reference formulas in the fasted state and the food effect on the test formulation was evaluated for rasagiline only, according to 90% confidence intervals (CIs) of geometric mean ratios for Cmax, AUC ,, AUCoo. The ratios and CI were calculated using ANCOVA with the log-transformed data (MIXED method, SAS). The conclusion regarding bioequivalence was based on the back-transformed point estimate and CI. Tmax were using nonparametric 49/70 Austrian Patent Office AT 12 508 Ul 2012-06-15
Analysis analyzed (Wilcoxon rank sum test).
[00476] 4. Results The following Tables 17a-17d summarize the test results of this study. Bioequivalence Test The test results showed that the tested sustained release formulations (Formulation I and Formulation III) met the criteria of bioequivalence of the known rapid release formulation. Both the Cmax and AUC reached a range of 80-140% within a 90% confidence interval between the formulation tested and the rapid release reference formulation. MAO Study: The test results showed that MAO-B activity for a formulation prepared according to each of Examples 3a and 3b was comparable to that of the rapid release reference formulation.
For the enzymatic determination of MAO, the standard method was used: "Determination of monoamine oxidase (MAO) by an extraction method using radiolabelled substrates in various tissues".
Briefly, fifty (50) μΙ of homogenate was added to 100 μΙ 0.1 M phosphate buffer (pH -7.4). After pre-incubation at 37 ° C for 20 minutes, 50 μM 14C-phenylethylamine hydrochloride (10 μM final concentration) was added and incubation continued for the next 20 minutes. The reaction was then stopped by addition of 2 M citric acid.
Radioactive metabolites were extracted into toluene / ethyl acetate (1: 1 v / v), a solution of 2,5-diphenyloxazole at a final concentration of 0.4% added, and the metabolite content estimated by liquid scintillation counting ,
Rat brain homogenate activity served as the standard (positive control) for the study.
Protein determination was performed by the Lowrey method.
Safety and Tolerability The test results showed that safety and tolerability were acceptable for each treatment.
Table 17a. PK parameters in the fasted state:
Study Formulation t1 / 2 (h) tmax (h) Cmax (ng / ml) AUCinf (ng h / ml) t1 / 2 part 1 AZILECT® mean ± standard deviation 1.92 ± 1.19 0.50 ± 0.24 5.790 ± 2.731 4.281 ± 1.280 Formulation I Avg. ± Standard Deviation 2.25 ± 1.15 2.68 ± 0.80 5.078 ± 2.160 4.092 ± 1.187 Part 2 AZILECT® Average ± Standard Deviation 2.65 ± 3.31 0.50 ± 0, 21 6.22 ± 2.585 4.960 ± 1.807 Formulation III Average ± standard deviation 2.12 ± 0.78 2.33 ± 0.80 5.739 ± 1.406 5.073 ± 1.045 50/70 Austrian Patent Office AT 12 508 Ul 2012-06-15 [00488 ] Table 17b. Bioequivalence in the fasted state:
Study Formulation Single dose Fasting Score 90% CI part 1 Formulation I AUC 96 79-117 Cmax 88 58-133 part 2 Formulation III AUC 105 106-117 Cmax 99 75-130 [00489] Table 17c. PK parameters in the saturated state:
Study Formulation t1 / 2 (h) tlag (h) tmax (h) Cmax (ng / ml) AUCinf (ng h / ml) Part 1 Formulation I - rapid mean ± standard deviation 2.25 ± 1.15 1.19 ± 0 , 82 2.68 ± 0.80 5.078 ± 2.160 4.092 ± 1.187 Formulation I - saturated mean ± standard deviation 2.29 ± 1.28 11.14 ± 5.88 12.31 ± 6.53 3.696 ± 2.670 3.664 ± 1.940 part 2 Formulation III - rapid Average ± standard deviation 2.12 ± 0.78 1.33 + 0.61 2.33 ± 0.80 5.739 ± 1.406 5.073 ± 1.045 Formulation III - saturated Mean ± standard deviation 2.90 ± 2.50 5 , 57 ± 1.98 6.12 ± 2.11 7.511 ± 3.448 4.995 ± 1.710 [00490] Table 17 d. Bioequivalence in the saturated state
Formulation Single dose Fasting Score 90% CI AZILECT® AUC 81 71-85 Cmax 49 29-50 Rasagiline Base Formulation I EC AUC 82 68-100 Cmax 58 38-87 Rasagiline Base Formulation III EC AUC 95 79-113 Cmax 121 90- 164
Findings: As shown in Tables 17a-17d, Formulation III meets the requirements in both fasted and saturated states. The PK parameters and bioequivalence of Formulation III are similar to those of AZILECT®.
EXAMPLE 18. PREPARATION OF RAS AG I LI N-CITRATE
Solid, crystalline rasagiline base used in this example was prepared by a similar procedure as described below: A) Preparation of rasagiline base oil 100.0 g of rasagiline tartrate suspended in 458 ml of deionized water, 229 ml of toluene were added and 46 ml of 25% NaOH solution were introduced with stirring. The mixture was warmed to 45 ° C, stirred at 45 ° C for 15 minutes and allowed to sit at this temperature.
There were two phases separated. The lower aqueous phase (pH = 13-14) was discarded, the upper toluene-containing phase was washed with 140 ml of deionized water. The resulting emulsion was allowed to settle and two phases separated. The lower aqueous phase (pH = 9-10) was discarded. The toluene-containing solution was evaporated under vacuum in the evaporator.
After completion of the solvent evaporation, 60 ml of isopropanol was added to the residue, and evaporation was continued. After completion of the evaporation, 50 ml of isopropanol was added and distilled off under the same conditions. It was obtained 51/70 Austrian Patent Office AT 12 508 U1 2012-06-15 the residue, oil of R-PAI base.
B) Crystallization of Rasagiline Base The rasagiline base oil obtained in step A) above was dissolved in 56 ml of isopropanol. The solution was cooled to ΙΘ'Ό and 147.5 ml deionized water was added portionwise over 3 hours with cooling and stirring. During the addition of water, development of precipitate was observed and the batch was immediately seeded with crystalline R-PAI base.
The resulting suspension was cooled to 2 ° C, stirred at this temperature overnight and filtered. The solid was washed with water and dried at room temperature under vacuum. Solid dry R-PAI base was obtained, with a yield of 96% based on oil base.
This example describes the preparation and characterization of rasagiline citrate salt. Rasagiline citrate is an attractive drug substance. Since citric acid is a tribasic compound, there are three possible forms of rasagiline citrate: mono-, di- and tri-citrate. Therefore, the rasagiline citrate described herein may be mono-rasagiline citrate, di-rasagiline citrate or tri-rasagiline citrate or a mixture thereof.
Since rasagiline is a weak base and pKs values of citric acid are 3.13, 4.76 and 6.40, it can be expected that binding of 1st and 2nd rasagiline base molecules to citrate is much more likely is as a binding of the 3rd Rasagilin base molecule.
Starting Materials Citric acid anhydrous acid of USP purity was used to prepare citrate salts.
Rasagiline base - crystalline rasagiline base, prepared as described above in this example.
Example 18a - Preparation of Rasagiline Citrate in Ethanol-Acetone 3.02 g of citric acid were dissolved in 10 ml of absolute alcohol at room temperature. 5.38 g of rasagiline base were dissolved in 15 ml of absolute alcohol. Rasagiline base solution was added portionwise with stirring to the citric acid solution. A significant exothermic effect was recorded during the addition, during which the temperature of the solution increased from 17 ° to 24 ° during 10 minutes of addition. The resulting clear solution was stored in a freezer at -18 ° C and no precipitation was observed.
An additional 2.71 g of Rasagiline solid base was added to the above resulting clear solution. After prolonged stirring at 20-23 ° C, the solid dissolved and it was obtained a viscous clear solution. The resulting viscous clear solution was stored overnight at -18 ° C in a freezer. No solid precipitate from the solution was observed in the freezer for 20 hours.
The solution was evaporated under vacuum in a rotary evaporator, the resulting residue (11.2 g) of honey-like semi-solid material was stored over the weekend in a freezer (-18 ° C). No crystallization of solid was observed.
The semi-solid material was mixed with 40 ml of acetone with stirring, no dissolution of the semi-solid material was observed during prolonged stirring.
Absolute ethanol (3 ml) was added portionwise with stirring. Complete dissolution of the semi-solid material was observed and the resulting clear solution was stored in the freezer overnight.
Honey-type, semi-solid material precipitated from the acetone-ethanol solution was found at the bottom of the vessel. The solution was decanted off and the precipitate under 52/70 Austrian Patent Office AT12 508U1 2012-06-15
Vacuum (20 mbar) dried for 4 hours. During drying, a stable foam formed. The jar with the foam was connected to a high vacuum pump and dried at 2-3 mbar overnight.
[00510] The foam solidified under high vacuum. The vacuum was turned off and the material broken with a spatula. There were obtained 6.1 g white powder.
[00511] Analysis: [00512] Analysis of Rasagiline Base by HPLC - 60.8% [00513] Crystallinity by XRD - Amorphous [00514] Thermal analysis: [00515] DSC peak at 179.7 ° C (128 exo), TGA-LOD = 1.2% (25-100 ^), continuous weight loss at Τϊ'ΙΟΟ'Ό.
Example 18b - Mono citrate salt in water-acetone (1: 1 molar ratio) [00516] 3.02 g of citric acid were dissolved in 4 ml of deionized water. 2.69 g rasagiline base was added to the solution. An exothermic effect was observed (temperature increased from 22 to 25 ° C) and almost all of the solid was dissolved. Then the mixture was heated to 42 ° and complete dissolution of the solid was observed. The resulting viscous syrupy solution was kept in a refrigerator at + 5 ° C overnight. No precipitation was observed for 15 hours.
The solution was mixed with 15 ml of acetone and evaporated on a rotary evaporator under vacuum. The residue of honey-type semi-solid material (6.29 g) was dried under vacuum at ambient temperature (20 mbar). During drying, a foam is formed (6.11 g) and then further dried under high vacuum (2-3 mbar).
The foam was solidified under high vacuum. The vacuum was turned off and the material broken with a spatula. There were obtained 5.58 g of white powder.
[00519] Analysis: [00520] Examination of Rasagiline Base by HPLC - 44.5% [00521] Crystallinity by XRD Amorphous [00522] Thermal analysis: [00523] DSC peak at 188.6 (61 exo), TGA -LOD = 1.5% (25-100 ^), continued weight loss at Τ ^ ΟΟΌ.
Example 18c - Di-citrate salt in water-acetone (2: 1 molar ratio) [00524] 3.45 g of citric acid were dissolved in 5 ml of deionized water and preheated to 30 ° C. 6.13 g rasagiline base was added to the solution. An exothermic effect was observed (temperature increased from 30 to 36 ^) and the solid was dissolved. The resulting clear, viscous, syrupy solution was kept in a refrigerator at + 5 ° C overnight. No precipitation was observed for 15 hours.
The solution was mixed with 18 ml of acetone and evaporated on a rotary evaporator under vacuum. The residue of honey-like semi-solid material (9.7 g) was dried under vacuum (20 mbar) at ambient temperature. During drying, a foam formed, which was then further dried under high vacuum (2-3 mbar).
The foam was solidified under high vacuum. The vacuum was turned off and the material broken with a spatula. There were obtained 8.81 g of white powder.
[00527] Analysis: [00528] Analysis of Rasagiline Base by HPLC - 60.9% 53/70 Austrian Patent Office AT12 508U1 2012-06-15 [00529] Crystallinity by XRD - Amorphous [00530] Thermal analysis: [00531] DSC- Peak at 180.2 ° C (141 exo), TGA-L0D = 1.2% (25-100'O), continued weight loss at Τϊ'ΙΟΟ'Ό.
Example 18d - Tri-citrate salt in water-acetone (3: 1 molar ratio) [00532] 3.46 g of citric acid were dissolved in 5 ml of deionized water. 9.19 g rasagiline base was added to the solution. An exothermic effect was observed (temperature increased from 22 to 27) and almost all of the solid was dissolved. Then, the mixture was warmed to 46 ° C, 0.5 ml of water was added and complete dissolution of the solid was observed. The resulting viscous syrupy solution was kept in a refrigerator at + 5 ° C overnight. No precipitation was observed for 15 hours.
The solution was mixed with 18 ml of acetone and evaporated on a rotary evaporator under vacuum. The residue of honey-type semi-solid material (13.20 g) was dried under vacuum at ambient temperature (20 mbar). During drying, a foam (13.19 g) was formed, which was then further dried under high vacuum (2-3 mbar).
[00534] The foam was solidified under high vacuum. The vacuum was turned off and the material broken with a spatula. There were obtained 12.80 g of white powder.
[00535] Analysis: [00536] Analysis of Rasagiline Base by HPLC - 70.6% [00537] Crystallinity by XRD Amorphous [00538] Thermal Analysis: [00539] DSC peak at 181.8 ° C (136 exo), TGA-LOD = 1.3% (25-100 ^ 0), weight loss at T> 100 ° C.
Discussion of Example 18 [00541] Experimental observations show exothermic reactions between rasagiline base and citric acid in aqueous solutions. The fact that rasagiline base having a solubility in water of about 2 mg / ml dissolves in aqueous reaction solution of more than 10% by weight shows complete or near complete conversion of the base to salt.
At the same time, part of the base could be extracted from the brine with nonpolar organic solvent such as toluene.
The production of mono-, di- and tri-citrate salts of rasagiline can be calculated from the relative molecular weights of rasagiline (R-PAI), citric acid and water. The calculation results are shown in Table 18 below. The data presented in Table 18 also show that the R-PAI content in the citrates prepared in Examples 18a-18d is consistent with the composition of the hydrate salts.
Table 18. Calculated composition of rasagiline citrate
Salt Composition MW R-PAI content Wt.% Water content Wt.% Monocitrate (R-PAIH +) CitH2- 363.3 47.1 0 Di-Citrate (R-PAIH +) 2CitH'ii 534.5 64 , 0 0 Tri-Citrate (R-PAIH +) 3Cit'a 705.7 72.8 0 Monocitrate Monohydrate (R-PAIH +) CitH2 '* H20 381.3 44.9 4.7 54/70 Austrian Patent Office AT12 508U1 2012-06-15
Di-citrate monohydrate (R-PAIH +) 2CitH "2 * H20 552.5 61.9 3.2 Tri-citrate monohydrate (R-PAI Η +) 30ΐΓ3 · Η20 723.7 70.9 2.5 Di Citrate Dihydrate (R-PAIH +) 2CitHT 2 * 2H20 570.5 60.0 3.1 [00545] R-PAIH + Rasagiline Base (R-PAI) Cation [00546] Citn Citrate Anion [00547] In Rasagiline citrate salts prepared in Examples 18a-18d show extremely high solubility in water. Solutions of mono-di- and tricitrate salts prepared in Examples 18b, 18c and 18d had dissolved solids concentrations of 59, 66 and 70 wt%, respectively. These solutions showed no saturation and proved to be stable at low temperatures. During 15 hours no precipitation was observed at + 5 * €. These data show extremely high solubility of the citrate salts of rasagiline in water. Solutions containing more than 70% by weight of rasagiline citrate could be prepared. Rasagiline citrate salts with a water content of 3-10 wt .-% appear as syrups or honey-like semi-solid.
The most soluble rasagiline salt heretofore described is a monobasic maleate salt of rasagiline which has a solubility of not less than 1000 mg / ml of water, as disclosed in U.S. Pat. Patent No. 6,630,514. However, the phenomenon of extremely high solubility exhibited by rasagiline citrate has not been observed with any previously identified salt of rasagiline.
Such extremely high solubility is a property of practical value and enables the preparation of highly concentrated liquid and semi-solid formulations. Aqueous or alcoholic solution of rasagiline citrate containing 60-80% of active pharmaceutical ingredient (API) could be used in the preparation of transdermal patches, sublingual strips and other formulations that could benefit from such highly concentrated liquid or semi-solid. Such highly concentrated solutions are also useful for optimizing the efficiency of manufacturing processes, e.g. B. for tablets.
EXAMPLE 19. Additional Preparation of Rasagiline Citrate
Solid crystalline rasagiline base used in this example was prepared in a similar procedure as described below: A) Preparation of rasagiline base oil. 100.0 g of rasagiline tartrate were prepared in 458 of deionized water, 229 ml of toluene were added, and 46 ml of 25% NaOH solution were introduced with stirring. The mixture was warmed to 45 ° C, stirred at 45 ° C for 15 minutes and allowed to sit at this temperature.
There were two phases separated. The lower aqueous phase (pH = 13-14) was discarded, the upper toluene-containing phase was washed with 140 ml of deionized water. The resulting emulsion was allowed to settle and two phases separated. The lower aqueous phase (pH = 9-10) was discarded. The toluene-containing solution was evaporated under vacuum in an evaporator.
After completion of the evaporation of the solvent, 60 ml of isopropanol was added to the residue, and evaporation was continued. After completion of the evaporation, 50 ml of isopropanol was added and distilled off under the same conditions. The residue, oil of R-PAI base, was obtained.
B) Crystallization of Rasagiline Base The rasagiline base oil obtained in step A) above was dissolved in 56 ml of isopropanol. The solution was cooled to 16 ° C. and 147.5 ml deionized water were added in portions over 3 hours with cooling and stirring to portions. During the addition of water, evolution of precipitate was observed and the batch was immediately seeded with crystalline R-PAI base.
The resulting suspension was cooled to 2 ° C, stirred at this temperature overnight and filtered. The solid was washed with water and dried at room temperature under vacuum. Solid dry R-PAI base was obtained, with a yield of 96% based on the oil base.
This example describes the additional preparations and characterization of rasagiline citrate salt.
[00559] Starting Materials Citric acid anhydrous acid of USP purity was used to prepare citrate salts.
[00561] Rasagiline base - pure crystalline rasagiline base (DS) prepared as in Example 18 was used in this study.
Example 19.1 [00563] 3.84 g of citric acid were dissolved in 25 ml of water and 3.42 g of rasagiline base were added to the solution, which was stirred at room temperature and monitored by TLC. After 30 minutes, no traces of R-PAI were observed on the TLC. The reaction mixture was extracted after one hour with 2x30 ml of toluene. The combined toluene-containing extract was evaporated to dryness. Yield: 0.06 g (1.75%) (R-PAI).
The aqueous phase was evaporated in vacuo to dryness. A honey-like semi-solid product was obtained. Yield: 7.53 g (103.7%).
Example 19.2 [00566] 1.92 g of citric acid were dissolved in 10 ml of water and 1.71 g of rasagiline base were added to the solution. The mixture was stirred for 18 hours and then the solvent was removed by lyophilization (1-0.3 mbar, -20- + 20Ό, 46 hours). Yield: 3.69 g (101.65%).
The product was a solid foam, but became a semi-solid honey-like material after several hours. According to NMR data, the salt formed with 0.73 equivalents of acid.
Example 19.3 [00569] 1.92 g of citric acid were dissolved in 15 ml of water and 3.42 g of rasagiline base were added to the solution. The reaction mixture was stirred at room temperature for 22 hours. The water was removed by freeze-drying (1-0.3 mbar, -20- + 20 ^: 46 hours).
Crystal-like foam was obtained, which then became a semi-solid honey-like material in a few hours. According to NMR data, the salt formed with 0.48 equivalents of acid.
Example 19.4 [00572] 3.84 g of citric acid were dissolved in 30 ml of water, and 6.84 g of rasagiline base were added to the solution, which was stirred for 2 hours, and then the reaction mixture was extracted with 2x40 ml of toluene , The combined toluene-containing extract was evaporated to dryness. 20 ml of IPA was added to the residue and the solvent was evaporated in vacuo to dryness. Yield: 1.5 g (22%, R-PAI).
The aqueous phase was evaporated to dryness to give a honey-like semi-solid product. Yield: 9.47 g (103.3%). 56/70 Austrian Patent Office AT 12 508 U1 2012-06-15 1H-NMR - 0.65 equivalents of salt formed the salt.
Example 19.5 [00576] 3.84 g of citric acid were dissolved in 50 ml of water and 10.26 g of rasagiline base were added to the solution, which was stirred at room temperature for 3 hours.
The reaction mixture was extracted with 2x50 ml of toluene. The combined toluene-containing extract was evaporated to dryness in vacuo. IPA was added to the residue and then evaporated to dryness. Yield: 3.92-4.13 g (R-PAI) (38.2-40.2%).
The aqueous phase was evaporated to dryness, giving a honey-like semi-solid product. Yield: 10.54-9.73 g.
1 H-NMR - 0.58 equivalents of acid formed the salt.
Example 19.6 3.84 g of citric acid were dissolved in 50 ml of water, and 10.26 g of rasagiline base were added to the solution, which was stirred at 60 ° C for 3 hours.
The reaction mixture was extracted with 2x50 ml of toluene. The combined toluene-containing extract was evaporated to dryness in vacuo. IPA was added to the residue and then evaporated to dryness. Yield: 3.92-4.13 g (R-PAI) (38.2-40.2%).
The aqueous phase was evaporated to dryness to give a honey-like semi-solid product. Yield: 10.54-9.73 g.
1 H-NMR - 0.58 equivalents of acid formed the salt.
Example 19.7 3.84 g of citric acid were dissolved in 50 ml of water and 10.26 g of rasagiline base were added to the solution, which was stirred at 25 ° C. for 42 hours.
The reaction mixture was extracted with 2x50 ml of toluene. The combined toluene-containing extract was evaporated to dryness in vacuo. IPA was added to the residue and then evaporated to dryness. Yield: 3.92-4.13 g (R-PAI) (38.2-40.2%).
The aqueous phase was evaporated to dryness, giving a honey-like semi-solid product. Yield: 10.54-9.73 g.
1 H NMR - 0.58 equivalents of acid formed the salt.
Example 19.8 1.92 g of citric acid were dissolved in 25 ml of water and 5.13 g of rasagiline base were added to the solution, which was stirred at room temperature for 16 hours. The reaction mixture was extracted with 2x30 ml of toluene and the toluene-containing extract was evaporated to dryness. Yield: 2.19 g (R-PAI, 42.7%).
The aqueous phase was dried by freeze-drying. The product was a crystalline foam which then became honey-like semi-solid.
1 H NMR - 0.55 equivalents of acid formed the salt.
Example 19.9 [00595] 1.92 g of citric acid were dissolved in 25 ml of water and 5.13 g of rasagiline base were added to the solution, which was stirred at room temperature for 9 days. The solid was filtered off, washed with 5 ml of water and dried with air. Yield: 0.31 g (6%, R-PAI), m.p .: 39.3-41.0 ^. 57/70 Austrian Patent Office AT12 508U1 2012-06-15 [00596] The aqueous phase was freeze-dried. The crystal-like foam was formed, which became honey-like semi-solid after a few hours.
1 H-NMR - 0.35 equivalents of acid formed the salt.
Example 19.10 1.6 g of citric acid were dissolved in 10 ml of water and 1.0 g of rasagiline base was added to the solution, which was stirred at room temperature. The solvent was removed by lyophilization. The product was a crystalline foam which became semi-solid after a few hours.
[00600] 1 H NMR -1.2 equivalents of acid formed the salt. Example 19.11 Example 19.11 [00602] 1.92 g of citric acid were dissolved in 15 ml of IPA and 1.71 g of rasagiline base were added to the solution, which was stirred at room temperature for 2 hours. No R-PAI was detected by TLC. The solvent was removed under vacuum. Yield: 3.85 (106%) The foamy semi-solid product became honey-like upon contact with the humidity.
Example 19.12 [00605] 1.92 g of citric acid were dissolved in 15 ml of IPA and 3.42 g of rasagiline base were added to the solution, which was stirred at room temperature for 2 hours. The reaction mixture, which was monitored by TLC (hexane: EtOAc = 1: 1), became clear. Traces of R-PAI were detected. The solvent was removed under vacuum. The residue was slurried in 2x30 ml of toluene. The combined toluene-containing phase was evaporated to dryness. Yield: 0.65 g (19%; R-PAI).
The crude product was dissolved in IPA and the solvent was evaporated to dryness to give a honey-like product.
Example 19.13 [00608] 1.92 g of citric acid were dissolved in 15 ml of IPA and 5.13 g of rasagiline base were added to the solution, which was stirred at room temperature for 2 hours. The reaction was monitored by TLC. Free R-PAI was available. The solvent was removed under vacuum. The residue was slurried in 2x30 ml of toluene. The combined toluene-containing phase was evaporated to dryness. Yield: 2.47 g (48%; R-PAI).
The crude product was dissolved in IPA and the solution was evaporated to dryness. A honey-like product was obtained.
[00610] Example 19.14 [00611] 1.92 g of citric acid were dissolved in 15 ml of methanol and 1.71 g of rasagiline base were added to the solution, which was stirred at room temperature for 22 hours and then evaporated to dryness. Yield: 3.77 g (103.86%).
1 H NMR - 0.72 equivalents of acid formed the salt.
Example 19.15 [00614] 1.92 g of citric acid were dissolved in 20 ml of methanol and 3.42 g of rasagiline base were added to the solution, which was stirred at room temperature for 22 hours and then evaporated to dryness. Yield: 5.48 g - 103.6%. TLC was used to detect free R-PAI in the product.
1 H-NMR - 0.5 equivalents of acid formed the salt. EXAMPLE 19.16 [00617] 1.92 g of citric acid were dissolved in 25 ml of methanol and 5.13 g of rasagiline base were added to the solution, which was dissolved at room temperature, 22 ° C Stirred for hours and then evaporated to dryness. Yield: 7.32 g - 103.8%. TLC was used to detect free R-PAI in the product.
1H-NMR - 0.33 equivalents of acid formed the salt.
Example 19.17 [00620] 1.92 g of citric acid were stirred in 20 ml of EtOAc and 1.71 g of rasagiline base were added to the solution, which was stirred for a further 72 hours. The reaction was monitored by TLC. The free rasagiline base was detected.
The solution was decanted from the reaction mixture. The solution was removed under vacuum. Yield: 1.32 g (77%) of R-PAI.
The isolated R-PAI was redissolved in 20 ml of ethyl acetate and 10 ml of water was added to the mixture. The reaction mixture was stirred for 22 hours. According to the TLC data, the unreacted R-PAI remained in the EtOAc phase. The phases were separated. The organic phase was evaporated to dryness. Yield: 0.13 g (7.6%) of R-PAI.
Example 19.18 [00624] 1.92 g of citric acid were stirred in 20 ml of EtOAc and 3.42 g of rasagiline base were added. The solution was stirred for an additional 72 hours. The reaction was monitored by TLC. The free rasagiline base was detected.
The solution was decanted from the reaction mixture. The solution was removed under vacuum. Yield: 2.87 g (83.9%, R-PAI).
The isolated R-PAI was redissolved in 20 ml of ethyl acetate and 10 ml of water was added to the mixture. The reaction mixture was stirred for 22 hours. According to the TLC data, the unreacted R-PAI remained in the EtOAc phase. The phases were separated. The organic phase was evaporated to dryness. Yield: 0.62 g (18% R-PAI).
Example 19.19 [00628] 1.92 g of citric acid were stirred in 25 ml of EtOAc and 5.13 g of rasagiline base were added. The reaction mixture was stirred for a further 72 hours. The reaction was monitored by TLC. In all cases, the free rasagiline base was detected.
The solution was decanted from the reaction mixture. The solvent was removed under vacuum. Yield: 4.49 g (87.5%, R-PAI).
The isolated R-PAI was redissolved in 20 ml of ethyl acetate and 10 ml of water was added to the mixture. The reaction mixture was stirred for 22 hours. According to TLC data, the unreacted R-PAI remained in the EtOAc phase. The phases were separated. The organic phase was evaporated to dryness. Yield: 1.76 g (34.3% R-PAI).
Example 19.20 [00632] 1.92 g of citric acid were stirred in 25 ml of toluene and 1.71 g of rasagiline base were added to the mixture. The heterogeneous mixture was stirred at room temperature for 24 hours. The solution was decanted from the reaction mixture. The toluene-containing phase was evaporated to dryness. Yield: 1.58 g (92.4%); (R-PAI by TLC).
The isolated R-PAI was redissolved in 10 ml of toluene and returned to the solid 59/70 Austrian Patent Office AT12 508U1 2012-06-15
Phase given. 20 ml of water was added to the heterogeneous mixture and stirred for 3 hours. The reaction was monitored by TLC. The phases were separated. The toluene-containing phase was evaporated to dryness. Yield: 0.12 g (7%), R-PAI was detected according to TLC data. The aqueous phase was evaporated to dryness.
Example 19.21 [00635] 1.92 g of citric acid were dissolved in 20 ml of acetone and 1.71 g of rasagiline base were added to the reaction mixture, which was stirred at room temperature for 2 hours. The reaction was monitored by TLC. No R-PAI was detected.
The solution was decanted from the honey-like precipitate. Yield: 2.43 g (66.9%).
The acetone-containing solution was evaporated to dryness. A honey-like product was obtained. Yield: 1.48 (40.7%).
The overall yield was 107.6% (there remained acetone in the product).
Example 19.22 [00640] 1.92 g of citric acid were dissolved in 20 ml of acetone and 3.42 g of rasagiline base were added to the mixture, which was stirred at room temperature for 22 hours. R-PAI was detected by TLC. The acetone-containing solution was decanted from the honey-like precipitate. Yield: 4.41 g (82.6%) semi-solid product.
The acetone-containing phase was evaporated to dryness. Yield: 1.34 g (25.1%) 60/70 Austrian Patent Office AT12 508U1 2012-06-15
Table 19a. Summary of experimental results PH (after extraction of R-PAI) G CO 4.88 (4.46) 6.09 (4.66) cn - m (4.90) R-PAI by TLC l 1 + + + + + + + + 1 '1 + + -1- + + + 1 "l · Equivalent to citric acid in the salt by NMR in cn OO o 1.55 Z9'l <N iCo ZL'O IO o εε' 0 VO ΓΓ ** x 00 rn Extracted R-PAI (%) (filtriert filtered) Extr. With toluene 0.06 g- 1.75% 1.51 g (22%) 4.13 g (40.2%) 4.44 g (43.3%) 3.92 g (38.2%) of C * βχ VO OD m O 00 tJ-00 tv ri C 00, n kO o freeze-dried Extr. 2.19 g (42 , 7%) freeze-dried fractions of reagents (mol) | Rasagiline Base, Mol - - mmm CN CN (N Γ "θ '- - <N ΓΌ - - CN m - <N Citric Acid, Mol - - - - - - - - - - τ i - - - - - - - τ-H - - solvent IPA water water water water 60 ° C water 2 days | - i water water IPA IPA water water water MeOH MeOH MeOH toluene EtOAc EtOAc EtOAc acetone acetone Example 19.11 Γ6Ι | 19.4 IO 0 19.6 19.7 19.9 19.3 19.13 | 19.12 0Γ61 19.8 19.2 19.14! 19.15 1 19.16 19.20 19.17 22.18 19.19 19.21 19.22 61/70 Austrian Patent Office AT12 508U1 2012-06-15 [00642] Discussion of Example 19 [00643] Rasagiline base readily forms salts with citric acid in almost all different ones Types of solvents, but easiest in water and in alcohols.
Mono-rasagiline citrate salt forms and is stable in most solvents. A few percent of free Rasagiline can be extracted from the aqueous solution of this salt.
Di- and tri-citrates are not so stable in the aqueous and other solvents (alcohol, MEK, acetone). Free Rasagiline base can be detected by TLC and extracted with toluene.
The separation of free rasagiline base from the aqueous solution of di- and tri-rasagiline citrates resulted in a change in the pH of the solution as shown in Table 19a.
All rasagiline citrate salts are hygroscopic salts and readily absorb air moisture. The rasagiline citrates are more likely to form strong solvates with the solvents in which the formation of the salt takes place (up to 10%).
The aqueous solution of rasagiline citrates can be dried by freeze-drying.
The NMR study of rasagiline citrate in the above examples provides information on the composition (proportion) of the samples and not on the proportion of the free base and the charged base (cationic form) with citric acid.
The results of this example also show that the ratio of rasagiline base: citric acid used to the extractable &quot; extractable &quot; Rasagiline base and the amount of unreacted citric acid found in the salt by NMR. The results are summarized in Table 19b below.
Table 19b. Effect of salt composition on extractable rasagiline base content
Salt Ratio Base to Acid Equivalent to Citric Acid by 1 H NMR for Salt Prepared in: Extractable Base Content (Toluene) Mol: Mole Methanol Water% Total Base Monocatrate 0.7: 1.0 Not Available Not Available Not Available Mono Citrate 1.0: 1.0 0.72 0.73 1.75-3.6 di-citrate 2.0: 1.0 0.50 0.48 22.0 tri-citrate 3.0: 1.0 0.33 0.35 42.7 The data in Table 19b show that an excess of citric acid dramatically reduces the extractable rasagiline base content.
It can be concluded that a low content of extractable rasagiline base (or a higher content of citric acid) provides a higher stability of rasagiline in the salt. Therefore, mono-citrate salt is the most stable rasagiline citrate salt and the most stable compositions of rasagiline citrate are compositions containing less than 1 mole of rasagiline base per 1 mole of citric acid.
EXAMPLE 20. EVALUATION OF RAS AG I LI N-CITRATE ALZES
Three samples of rasagiline citrates prepared in Examples 18b, 18c and 18d were exposed in open dishes at ambient temperature to atmospheric air. The changes were observed and recorded. The results are presented in the following Table 20a: 62/70 Austrian Patent Office AT12 508U1 2012-06-15 [00655] Table 20a. Changes in citrate salts exposed to the atmosphere at ambient temperature
Example 4b Example 4c Example 4d Salt Type Mono-Di-Tri- Exposure Period (Hours: Minutes) 0:00 Powder Powder Powder 0:30 Powder Powder 0: 50-1: 00 Sticky Aggregates Powder Sticky Aggregates 1: 50-2: 00 Semisolid Powder Sticky Aggregates 5:00 Honeyy Semisolid Lumpy Powder Semisolid 6:00 Syrup Sticky Aggregates Semisolid 7:00 Syrup Sticky Aggregates Honeyy Semisolid 25:00 Not Available Sticky Aggregates + Semi-Solid Not Available [00656] Discussion [00657] The results in Table 20a show in that all three salts disclosed above are highly hygroscopic when exposed to ambient temperature atmosphere. These results also show that there is no significant difference in hygroscopicity between the mono-, di- and tri-rasagiline citrates. All three salts appear as hydrates.
Parkinson's patients suffer from dysphagia that prevents them from swallowing normal tablets or capsules. (Potulska A., "Swallowing Disorders in Parkinson's Disease", Parkinsonism Relat. Disord. (2003 Aug) vol. 9 (6), pp. 349-53). This difficulty hinders their treatment by reducing patient compliance. Patients are more likely to agree with dosage instructions when swallowing tablets or capsules is not required.
One means of avoiding the absorption of rasagiline in the stomach and eliminating the need to swallow tablets is to absorb rasagiline into the body before it reaches the stomach. Such absorption of rasagiline, and thus the solution to both problems, can be achieved by contact with buccal, sublingual, pharyngeal and / or esophageal mucosal membranes. To accomplish this, oral compositions can be formed which rapidly dissolve within the mouth to allow for maximum contact of rasagiline with the buccal, sublingual, pharyngeal and / or esophageal mucosal membranes. The unexpectedly high hygroscopicity of the citrate salts of rasagiline is particularly suitable for such oral formulations.
Another three samples of rasagiline citrates prepared in Examples 18b, 18c and 18d were stored in closed transparent paraffin-sealed glass vials in a refrigerator at 7 ± 2 ° C. The changes were observed and recorded. The results are presented in the following Table 20b: Table 20b. Appearance of citrate salts stored in the refrigerator
Example 4b Example 4d Salt Type monobasic dibasic three-season Storage Time, Month (s) Appearance: 0 white powder white powder white powder 3 white powder white powder white powder 6 white powder white powder white powder 63/70 Austrian Patent Office AT 12 508 U1 2012- Discussion [0066] Discussion The results in Table 20b show that all three salts could be stored for a long time (longer than 6 months) under occlusion at a low temperature (~ 7Ό) with no change in color or the appearance despite its high hygroscopicity at ambient temperature. This result was surprising and may be the result of the effect of temperature on the hygroscopicity point of rasagiline citrates.
The results in Table 20b also show that all three salts could be handled and processed under controlled conditions such as low temperature and low humidity without changing their physical appearance despite their high hygroscopicity.
EXAMPLE 21. CHARACTERIZATION OF RASAGILINE-CITRATE-XRD ANALYSIS
Samples were tested using a Scintag X-ray powder diffractometer, Model X'TRA, Cu X-ray tube, solid state detector.
[00666] Scan Parameters [00667] Range: 2-40 degrees two-theta.
[00668] Scan Mode: Continuous Scan [00669] Increment: 0.05 degrees.
[00670] Rate: 3 degrees / min.
[00671] Sample holder: A round standard aluminum sample holder with a round zero-background quartz plate with a diameter of 25 (diameter) * 0.5 (depth.) Mm.
[00672] Table 21. Characteristic XRD peak positions of the various samples (± 0.2 degrees two-theta) __'___
Form Amorphous form Amorphous form Amorphous form Amorphous form Sample 1 2 3 4 Tip positions not available not available not available not available [00673] Discussion The results in Table 21 show that samples of rasagiline citrate have no characteristic peaks in XRD Analysis, indicating that the rasagiline citrates produced are in amorphous form.
EXAMPLE 22. COMPARISON OF PROPERTIES OF RASAGILINE CITRATE WITH OTHER SALTS
Rasagiline citrate exhibits properties other than the properties of other citrate salts, as shown in Table 22a, and which are other than the properties of other rasagiline salts, as shown in Table 22c.
Table 22a - Summary of citrates of various drug substances
Citrate salt of drug substance Polymorph References 5,8,14-triazatetracyclohexdecqa-2 (11), 3,5,7,9-pentaene crystalline WO 02/092597 2-Hydroxy-3- [5- (morpholin-4-ylmethol ) pyridin-2-yl] 1H-indole-5-carbonitrile crystalline WO 07/089191 2- (6- {2 - [(2 (2R) -2-methyl-1-pyrrolidin-1-yl] -ethyl) -2-naphthalen-2-yl) -2H-pyridazin-3-one crystalline US 2005/0256127 4- (3,4-dichlorophenyl) -2- [2-94-methylpiperazin-1-yl] -benzylidenethiomorpholine 3-on crystalline US 2003/0181444 64/70 AT 12 508 U1 2012-06-15 Austrian
Patent Office 4 - [(4-Methyl-1-piperazinyl) methyl] -N- [4-methyl-3 - [[(4- (3-pyridinyl) -2-pyrimidinyl] -amino] -phenyl] -benzamide crystalline US 2008 / 0249104 5- (5-Fluoro-2-oxo-1,2-dihydroindol-3-ylidenemethyl) -2,4-dimethyl-1H-pyrrole-3-carboxylic acid (2-pyrrolidin-1-yl-ethyl) -amide crystalline 3 - {(3R, 4R) -4-Methyl-3- [methyl- (7H-pyrrole) 2,3-d] pyrimidin-4-yl) -amino] -piperidin-1-yl} - US Patent Application 2008/0275101 3-oxopropionitrile crystalline US 2005/0159434 Decitabine crystalline US 2006/0069060 [00678] As shown in Table 22a, unlike the citrate salts of other drug substances, the rasagiline citrate salts are amorphous. Sagilin citrate detected.
Table 22b Summary of properties of rasagiline base and citric acid
Hygroscopicity by KF Water solubility (mg / ml) References Solid R-PAI (free base) non-hygroscopic low US 2008/0161408 Low citric acid 1330 &quot; Pharmaceutical Excipients &quot; Database [00680] Table 22c - Summary of Properties of Rasagiline Salts R-PAI Base / R-PAI Salts Hygroscopicity by KF Water Solubility (mg / ml) References Chloride non-hygroscopic 238 US 5,457,133 mesylate non-hygroscopic 635 US 5,532,415 tartrate non-hygroscopic 33 US 5,532,415 maleate not available> = 1000 US 5,532,415 sulfate not available 485 US 5,532,415 tosylate not available 60-70 US 5,532,415 fumarate not available 95 US 5,532,415 phosphate not available> 720 US 5,532,415 Esylate not available> 300 US 5,532,415 Acetate not available> = 720 US 5,532,415 tannate <10% (R-PAI based on content) low US 7,547,806 citrate extremely hygroscopic extremely large (greater than rasagiline maleate) edisilate non-hygroscopic 342.5 WO 2008/019871 oxalate not hygroscopic 19.7 WO 2008/019871 The results in Tables 22b and 22c show that compared to rasagiline base and other rasagiline salts Rasagiline citrate salt shows the highest water solubility and the strongest hygroscopicity.
The following numbers characterize particular embodiments of the invention: 1. A stable oral dosage form containing a core having a form of rasagiline base resulting from a manufacturing process, and at least one pharmaceutically acceptable excipient; and an acid resistant, pharmaceutically acceptable coating, wherein the manufacturing process comprises a) preparing the core by mixing rasagiline base, citric acid and / or malic acid, and a pharmaceutically acceptable excipient; and b) coating the core with the acid-resistant pharmaceutically acceptable coating.
Number 2. The dosage form according to item 1, wherein step a) comprises preparing wet granules of rasagiline base and citric acid and a pharmaceutically acceptable excipient, and wherein step a) further comprises: i) drying the wet granules to form a dry granules, preferably in a fluidized bed dryer at an air inlet temperature of 40 ° to 50 ° C, and an air outlet temperature of not higher than 37 ° C, ii) crushing the dry granules to form particles, preferably by an oscillating granulator, and iii) mixing the particle with at least one lubricant, wherein the lubricant is preferably talc or stearic acid, or a combination thereof.
A stable oral dosage form containing a core comprising at least one pharmaceutically acceptable excipient and rasagiline base, rasagiline citrate, rasagiline malate, or a mixture of at least two of rasagiline base, rasagiline citrate and Having rasagiline malate; and an acid resistant, pharmaceutically acceptable coating.
Number 4. The stable oral dosage form of item 3 having a weight of less than 150 mg, the core consisting essentially of at least one pharmaceutically acceptable excipient and rasagiline citrate, wherein the at least one pharmaceutically acceptable excipient in the Core is at least one antioxidant and at least one disintegrant, wherein the antioxidant is preferably citric acid and the disintegrant is present in the core preferably in an amount of between 0.5% and 20% by weight, and wherein the disintegrant is preferably pregelatinized starch is.
[00687] Item 5. The dosage form according to Number 4, wherein the content of rasagiline
Citrate is 0.74 mg to 3.63 mg, and in addition to the rasagiline citrate, mannitol, colloidal silica, starch NF, pregelatinized starch, stearic acid, talc, hypromellose, methacrylic acid-ethyl acrylate copolymer, talc extra fine, and triethyl citrate contains.
Number 6. The dosage form according to item 4 or 5, wherein the core is in the form of a tablet, wherein the content of rasagiline is 1.0 mg and the tablet contains 45.0 mg of mannitol, or wherein the content of rasagiline is 0 5 mg and the tablet contains 45.5 mg of mannitol, and the tablet further contains 0.4 mg Aerosil, 5.0 mg NF, 20.0 mg pregelatinized starch, 1.5 mg stearic acid, 1.5 mg talc , 3.5 mg of hypromellose, 4.0 mg of methacrylic acid-ethyl acrylate copolymer, 0.8 mg of triethyl citrate, 1.9 mg of extra fine talc, and 2.0 mg of a colored coating agent. [00689] Number 7. The dosage form according to any one of 3-6, wherein the more acidic, pharmaceutically acceptable coating comprises methacrylic acid-ethyl acrylate copolymer (1: 1) and a plasticizer wherein the ratio of methacrylic acid-ethyl acrylate copolymer (1: 1) to plasticizer is preferably between 10 to 1 and 2 to 1, more preferably about 5 to 1, wherein the plasticizer is triethyl citrate, the acid resistant, pharmaceutically acceptable coating wherein the acid-resistant pharmaceutically acceptable coating is between 3% and 12% by weight of the dosage form, preferably about 8% by weight of the dosage form, or wherein the acid-resistant pharmaceutically acceptable coating contains two coating layers Preferably, the interior of the two coating layers contains hypromellose.
Number 8. The dosage form according to any one of items 1-7, which releases between 80 and 100% rasagiline when placed in a basket apparatus in 500 ml of a buffered aqueous medium at pH 6.8 at 37 ° for 20 minutes 0 at 75 revolutions per minute, which, when taken by a human, achieves substantially the same MAO-B inhibition as that of the corresponding rasagiline dosage, which is taken as a rapid release formulation, which when taken by a human, provides an AUC level of rasagiline of 80-130%, preferably 80-125% of that of the corresponding amount of rasagiline taken as a rapid release formulation which, when administered from a person in the saturated state, provides an AUC value of rasagiline that is greater than that of the corresponding amount of rasagiline, which is considered to be a formulation with rapid clearance which, when ingested by a human, provides a Cmax of rasagiline of 80-145%, preferably 80-125% of the corresponding amount of rasagiline taken as a rapid release formulation, or [00691] which, when taken by a human in the saturated state, one
Cmax to rasagiline, which is greater than that of the corresponding amount of rasagiline, which is taken as a formulation with rapid release.
[0069] Number 9. The dosage form according to any one of items 1-8, wherein the total amount of nonpolar impurities is less than 0.3% by weight based on the amount of rasagiline, or wherein the amount of N- (2- chloroallyl) -1 (R) -aminoindane in the dosage form is less than 20 ppm, preferably less than 4 ppm, based on the amount of rasagiline.
Number 10. Rasagiline citrate.
Number 11. The rasagiline citrate according to item 10 which is isolated rasagiline citrate which is solid amorphous rasagiline citrate or which is substantially pure. No. 12. The rasagiline citrate according to any one of items 3, 10 and 11, which is a mono-rasagiline citrate having a rasagiline content of between 42 Wt .-% and 52 wt .-% based on the total weight of rasagiline citrate, or having a determined by Karl Fischer analysis water content of less than 5%.
Number 13. A composition, preferably in the form of a tablet containing the
Rasagiline citrate according to any one of the numbers 10-12 and a carrier, the composition wherein the rasagiline content in the form of rasagiline citrate is greater than 50% of the total rasagiline content in the composition, the composition Rasagiline citrate is mixed with a polymer, or the composition which is a pharmaceutical composition and wherein the carrier is a pharmaceutically acceptable carrier.
[00697] Item 14. The composition according to Number 13, which further contains rasagiline base, preferably in an amount of less than 5% based on the total rasagiline content of the composition, or which does not contain rasagiline base, or the further stearic acid contains.
Number 15. A process for producing rasagiline citrate according to any one of
Nos. 10-12 or the composition according to number 13 or 14, comprising: a) combining a citric acid solution with rasagiline base to form a first mixture; b) adding a solvent, preferably acetone, to the first mixture to form a second mixture; c) complete removal of the liquid, preferably at ambient temperature and under reduced pressure, from the second mixture; and d) recovering the rasagiline citrate or preparing the composition.
[00699] Number 16. Use of the dosage form according to one of the numbers 1-9 or the
A composition according to number 13 or 14 in an amount effective to treat a human suffering from Parkinson's disease, wherein the human optionally suffers from delayed gastric emptying or the human being optionally in a saturated state. 68/70
权利要求:
Claims (12)
[1]
Austrian Patent Office AT12 508U1 2012-06-15 Claims 1. An oral dosage form containing a core comprising rasagiline citrate and at least one pharmaceutically acceptable excipient; and an acid resistant, pharmaceutically acceptable coating.
[2]
The oral dosage form of claim 1 which has a weight of less than 150 mg, the core consisting essentially of at least one pharmaceutically acceptable excipient and rasagiline citrate, wherein the at least one pharmaceutically acceptable excipient in the core comprises at least one antioxidant and at least one disintegrant, wherein the antioxidant is preferably citric acid and the disintegrant is preferably present in the core in an amount of between 0.5% and 20% by weight, and wherein the disintegrant is preferably pregelatinized starch.
[3]
The oral dosage form of claim 2, wherein the rasagiline citrate content is 0.74 mg to 3.63 mg, and in addition to the rasagiline citrate mannitol, colloidal silica, starch NF, pregelatinized starch, stearic acid, talc, hypromellose , Methacrylic acid-ethyl acrylate copolymer, talc extra fine and triethyl citrate.
[4]
The oral dosage form of any one of claims 1 to 3, wherein the acid resistant, pharmaceutically acceptable coating comprises methacrylic acid-ethyl acrylate copolymer 1: 1 and a plasticizer.
[5]
The oral dosage form according to any one of claims 1 to 3, wherein the core is in the form of a tablet, wherein the content of rasagiline is 1.0 mg.
[6]
6. Oral dosage form according to claim 5, containing 45.0 mg of mannitol, 0.4 mg of Aerosil, 5.0 mg of NF, 20.0 mg of pregelatinized starch, 1.5 mg of stearic acid, 1.5 mg of talc, 3.5 mg of hypromellose , 4.0 mg of methacrylic acid-ethyl acrylate copolymer, 0.8 mg of triethyl citrate, 1.9 mg of talc extra fine and 2.0 mg of a colored coating agent.
[7]
7. An oral dosage form according to claim 5, containing 79.84 mg of mannitol, 0.6 mg of colloidal silica, 10.0 mg of NF, 20.0 mg of pregelatinized starch, 2.0 mg of stearic acid, 2.0 mg of talc, 4.8 mg Hypromellose, 6.25 mg of methacrylic acid-ethyl acrylate copolymer, 1.25 mg of triethyl citrate and 3.1 mg of extra fine talc.
[8]
8. Oral dosage form according to claim 5, containing 67.8 mg of mannitol, 0.6 mg of Aerosil, 10.0 mg of starch NF, 20.0 mg of pregelatinized starch, 2.0 mg of stearic acid, 2.0 mg of talc, 4.8 mg of hypromellose , 4.0 mg of methacrylic acid-ethyl acrylate copolymer, 0.8 mg of triethyl citrate and 1.9 mg of talc extra fine.
[9]
9. An oral dosage form according to claim 5, containing 80.34 mg of mannitol, 0.6 mg of Aerosil, 10.0 mg of starch NF, 20.0 mg of pregelatinized starch, 2.0 mg of stearic acid, 2.0 mg of talc, 4.8 mg of hypromellose , 6.25 mg of methacrylic acid-ethyl acrylate copolymer, 1.25 mg of triethyl citrate and 3.1 mg of talc extra fine.
[10]
10. Oral dosage form according to claim 5, containing 68.3 mg of mannitol, 0.6 mg of Aerosil, 10.0 mg of starch NF, 20.0 mg of pregelatinized starch, 2.0 mg of stearic acid, 2.0 mg of talc, 4.8 mg of hypromellose , 4.0 mg of methacrylic acid-ethyl acrylate copolymer, 0.8 mg of triethyl citrate and 1.9 mg of talc extra fine.
[11]
The oral dosage form according to any one of claims 1 to 3, wherein the core is in the form of a tablet, wherein the content of rasagiline is 0.5 mg.
[12]
12. Oral dosage form according to claim 11, containing 45.5 mg of mannitol, 0.4 mg Aerosil, 5.0 mg of starch NF, 20.0 mg of pregelatinized starch, 1.5 mg of stearic acid, 1.5 mg of talc, 3.5 mg of hypromellose , 4.0 mg of methacrylic acid-ethyl acrylate copolymer, 0.8 mg of triethyl citrate, 1.9 mg of talc extra fine and 2.0 mg of a colored coating agent. 69/70
类似技术:
公开号 | 公开日 | 专利标题
AT12508U1|2012-06-15|RASAGILINE FORMULATION WITH DELAYED RELEASE
EP1892233A1|2008-02-27|New salts of the active component rasagiline
JP2013537530A|2013-10-03|Rasagiline citrate dispersion
WO2011087791A1|2011-07-21|3-keto-n-propargyl-1-aminoindan
EP1848417A1|2007-10-31|Formulations of ladostigil tartrate
EP2218444A2|2010-08-18|Delayed release rasagiline formulation
JP5766713B2|2015-08-19|Transnorsertraline formulations, salts, and polymorphs and uses thereof
EP2416761B1|2015-02-11|Desfesoterodin in the form of a tartaric acid salt
WO2009129913A1|2009-10-29|Solid pharmaceutical preparation comprising 1-[|-amide]-2-{[4-|-phenyl]-amide}-4-hydroxy-pyrrolidine-1,2-dicarboxylic acid
同族专利:
公开号 | 公开日
EA201170962A1|2012-03-30|
PT2308477E|2012-08-09|
AT518527T|2011-08-15|
CA2799515A1|2010-11-23|
US20120263789A1|2012-10-18|
EP2308477A1|2011-04-13|
US20100189787A1|2010-07-29|
AT556703T|2012-05-15|
JP5774495B2|2015-09-09|
RS52421B|2013-02-28|
US8080584B2|2011-12-20|
EA029262B1|2018-02-28|
CN102333442A|2012-01-25|
EP2246321B1|2011-08-03|
IL214220A|2016-03-31|
HK1150308A1|2011-11-25|
DK201100050U1|2011-07-08|
CA2727899A1|2010-11-23|
PL2246321T3|2011-12-30|
CY1112486T1|2015-12-09|
US20100190859A1|2010-07-29|
AU2010206970B2|2016-05-05|
DK2308477T3|2012-08-13|
US7855233B2|2010-12-21|
CA2713292C|2011-08-02|
PT2246321E|2011-10-19|
JP2015091813A|2015-05-14|
PL2308477T3|2012-10-31|
EP2381767B1|2016-01-20|
ES2564317T3|2016-03-21|
DK201100050U4|2014-10-24|
AU2010206970A1|2011-08-18|
US20100189788A1|2010-07-29|
DE202010017129U1|2011-09-14|
SI2246321T1|2011-11-30|
DK2246321T3|2011-11-21|
RS52033B|2012-04-30|
US20120003310A1|2012-01-05|
US20100189791A1|2010-07-29|
EP2308477B1|2012-05-09|
EP2381767A4|2014-12-24|
HRP20120633T1|2012-09-30|
CY1112954T1|2016-04-13|
AR075148A1|2011-03-09|
ES2389889T3|2012-11-02|
SI2308477T1|2012-09-28|
CA2727899C|2013-02-26|
US20140186514A1|2014-07-03|
EP2381767A1|2011-11-02|
US20120100189A1|2012-04-26|
CZ22819U1|2011-10-24|
ES2370980T3|2011-12-26|
WO2010085354A1|2010-07-29|
HRP20110745T1|2011-12-31|
JP2012515775A|2012-07-12|
EP2246321A1|2010-11-03|
IL214220D0|2011-08-31|
US20100189790A1|2010-07-29|
CN102333442B|2015-08-19|
CA2713292A1|2010-11-23|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
US20060018957A1|2004-07-26|2006-01-26|Lerner E I|Pharmaceutical dosage forms including rasagiline|
WO2006012634A1|2004-07-26|2006-02-02|Teva Pharmaceutical Indudstries, Ltd.|Dosage forms with an enterically coated core tablet|
EP1987816A1|2007-04-30|2008-11-05|Ratiopharm GmbH|Adsorbate of a rasagiline salt with a water-soluble inactive ingredient|
US761917A|1903-02-11|1904-06-07|Alphonse King|Telegraph-transmitter.|
IL92952A|1990-01-03|1994-06-24|Teva Pharma|R-enantiomers of n-propargyl-1-aminoindan compounds, their preparation and pharmaceutical compositions containing them|
US5744500A|1990-01-03|1998-04-28|Teva Pharmaceutical Industries, Ltd.|Use of R-enantiomer of N-propargyl-1-aminoindan, salts, and compositions thereof|
IL99759A|1991-10-16|1997-06-10|Teva Pharma|Mono-fluorinated derivatives of n-propargyl-1-aminoindan, their preparation and pharmaceutical compositions containing them|
IL111240A|1993-10-18|2001-10-31|Teva Pharma|Salts of r - enantiomers of n- propargyl-1-aminoindan and pharmaceutical compositions comprising them|
IL115357A|1995-09-20|2000-01-31|Teva Pharma|Stable compositions containing N-propargyl-1-aminoindan and polyhydric alcohols|
US5663415A|1996-06-28|1997-09-02|Jame Fine Chemicals, Inc.|Process for preparing antihistamine tannates|
IL118836A|1996-07-11|2001-01-11|Teva Pharma|Pharmaceutical compositions comprising s--n-propargyl-1-aminoindan|
DE69732984T2|1996-12-18|2006-02-16|Teva Pharmaceutical Industries Ltd.|AMINOINDANDERIVATE|
AU6417198A|1997-03-13|1998-09-29|Bioresearch Ireland a division of Eolas - The Irish Science and Technology Agency|Cytoprotective agents comprising monoamine oxidase inhibitors|
US6548706B2|1999-12-23|2003-04-15|Aerojet Fine Chemicals Llc|Preparation of 2S,3S-N-isobutyl-N- -p-nitrobenzenesulfonylamide hydrochloride and other derivatives of 2-hydroxy-1,3-diamines|
DE10041478A1|2000-08-24|2002-03-14|Sanol Arznei Schwarz Gmbh|New pharmaceutical composition|
US20070197484A1|2001-05-03|2007-08-23|Ching Song|Method of treating disorder related to high cholesterol concentration|
SK13372003A3|2001-05-14|2004-08-03|Pfizer Products Inc.|Citrate salt of 5, 8, 14-triazatetracyclo[10.3.1.02.11.04.9]- hexadeca-2,3,5,7,9-pentaene, and pharmaceutical compositions thereof|
GT200200234A|2001-12-06|2003-06-27|NEW CRYSTAL COMPOUNDS|
MXPA04005418A|2001-12-07|2005-04-19|Pfizer Prod Inc|Citric acid salt of a therapeutic compound and pharmaceutical compositions thereof.|
US7118765B2|2001-12-17|2006-10-10|Spi Pharma, Inc.|Co-processed carbohydrate system as a quick-dissolve matrix for solid dosage forms|
SI1567152T1|2002-11-15|2013-10-30|Teva Pharmaceutical Industries Ltd.|Use of rasagiline with riluzole to treat amyotrophic lateral sclerosis|
AR044007A1|2003-04-11|2005-08-24|Newron Pharmaceuticals Inc|METHODS FOR THE TREATMENT OF PARKINSON'S DISEASE|
US20060177507A1|2003-05-22|2006-08-10|Joaquina Faour|Controlled release device containing lercanidipine|
AR047530A1|2004-02-04|2006-01-25|Novartis Ag|FORMS OF SALT OF 4- -N- PIRIMIDIN-2-ILAMINO) PHENYL) -BENZAMIDA|
US20050214372A1|2004-03-03|2005-09-29|Simona Di Capua|Stable pharmaceutical composition comprising an acid labile drug|
US7145005B2|2004-05-12|2006-12-05|Abbott Laboratories|2--2-Methyl-1-pyrrolidin-1-yl]-ethyl}-2-naphthalen-2-yl)-2H-pyridazin-3-one salts and their preparation|
US20060069060A1|2004-09-27|2006-03-30|Sanjeev Redkar|Salts of decitabine|
NZ555470A|2004-11-24|2011-02-25|Teva Pharma|Rasagiline orally disintegrating compositions|
CN101098685A|2004-11-24|2008-01-02|特瓦制药工业有限公司|Rasagiline orally disintegrating compositions|
WO2006089164A1|2005-02-17|2006-08-24|Teva Pharmaceutical Industries, Ltd.|Combination therapy with glatiramer acetate and rasagiline for the treatment of multiple sclerosis|
NZ560660A|2005-02-23|2010-11-26|Teva Pharma|Rasagiline -N-propargyl-1-aminodan ) formulations of improved content uniformity|
US8349951B2|2005-05-31|2013-01-08|Prc Desoto International, Inc.|Polythioether polymers and curable compositions containing them|
US20070078172A1|2005-06-16|2007-04-05|Jenrin Discovery|Mao-b inhibitors useful for treating obesity|
GB0514593D0|2005-07-15|2005-08-24|Davy Process Techn Ltd|Process|
MX2008002415A|2005-09-19|2008-03-27|Pfizer Prod Inc|Solid salt forms of a pyrrole substituted 2-indolinone.|
FR2891459B1|2005-09-30|2007-12-28|Flamel Technologies Sa|MICROPARTICLES WITH MODIFIED RELEASE OF AT LEAST ONE ACTIVE INGREDIENT AND ORAL GALENIC FORM COMPRISING THE SAME|
AU2006316585B2|2005-11-17|2012-09-20|Teva Pharmaceutical Industries, Ltd.|Methods for isolating propargylated aminoindans|
HU0501084A2|2005-11-23|2008-05-28|Richter Gedeon Nyrt|New pharmaceutical compositions of high effectivity|
US7572834B1|2005-12-06|2009-08-11|Teva Pharmaceutical Industries, Ltd.|Rasagiline formulations and processes for their preparation|
SA2797B1|2006-02-02|2011-10-29|استرازينيكا ايه بي|Citrate Salt of 2-Hydroxy-3-[5-Pyridin-2-Yl]1H-Indole-5-Carbonitrile Citrate|
ES2551481T3|2006-02-21|2015-11-19|Teva Pharmaceutical Industries, Ltd.|Use of rasagiline for the treatment of multisystemic atrophy|
WO2007102999A2|2006-02-21|2007-09-13|Ampla Pharmaceuticals Inc.|Cb1 antagonists and inverse agonists|
CN101032474B|2006-03-06|2011-02-16|重庆医药工业研究院有限责任公司|Rasagiline transparent patch for curing and preventing neurological diseases and the preparing method thereof|
EP2007369A4|2006-04-03|2009-07-01|Teva Pharma|Use of rasagiline for the treatment of restless legs syndrome|
US8524695B2|2006-12-14|2013-09-03|Knopp Neurosciences, Inc.|Modified release formulations of -4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine and methods of using the same|
WO2008010768A1|2006-07-17|2008-01-24|Cereuscience Ab|Method of treating and diagnosing restless legs syndrome and periodic limb movements during sleep and means for carrying out the method|
EP1892233A1|2006-08-18|2008-02-27|Ratiopharm GmbH|New salts of the active component rasagiline|
CN1911211B|2006-08-25|2010-04-14|重庆医药工业研究院有限责任公司|Solid oral preparation of leishajilan|
CN100542524C|2006-09-29|2009-09-23|北京德众万全药物技术开发有限公司|A kind of Pharmaceutical composition that contains rasagiline|
CL2007003409A1|2006-11-28|2008-04-11|Wyeth Corp|CONTROLLED RELEASE PEARL THAT INCLUDES A CENTRAL NUCLEUS UNIT, A FIRST LAYER WITH A PHARMACOLOGICAL AGENT THAT IS A COMPOSITE DERIVED FROM PIPERAZINE-PIPERIDINE, AN ACCIDIFIER, A SECOND LAYER; PREPARATION PROCEDURE; FORMULATION|
JP5623081B2|2006-12-14|2014-11-12|テバ ファーマシューティカル インダストリーズ リミティド|Crystalline solid rasagiline base|
EP2101570B1|2006-12-14|2013-02-13|Teva Pharmaceutical Industries, Ltd.|Tannate salt of rasagiline|
US7949403B2|2007-02-27|2011-05-24|Accelerated Care Plus Corp.|Electrical stimulation device and method for the treatment of neurological disorders|
WO2008139984A1|2007-04-26|2008-11-20|Eisai R & D Management Co., Ltd.|Cinnamide compounds for dementia|
JP5577012B2|2007-05-03|2014-08-20|株式会社カネカ|Multilayer substrate and manufacturing method thereof|
WO2009032273A1|2007-09-05|2009-03-12|Teva Pharmaceutical Industries, Ltd.|Method of treating glaucoma using rasagiline|
US8188149B2|2007-09-17|2012-05-29|Teva Pharmaceutical Industries, Ltd.|Use of R-N-propargy1-1-aminoindan to treat or prevent hearing loss|
NZ585752A|2007-12-24|2011-11-25|Cipla Ltd|Process for the synthesis of propargylated aminoindan derivatives|
NZ586025A|2008-01-11|2012-08-31|Teva Pharma|Rasagiline mesylate tablet coated with a composisiton comprising methacrylic acid - ethyl acrylate copolymer and triethyl citrate|
US20090247537A1|2008-03-25|2009-10-01|William Dale Overfield|Methods for preventing or treating bruxism using dopaminergic agents|
EP2271612B1|2008-03-31|2016-08-10|Actavis Group PTC EHF|Rasagiline mesylate particles and process for the preparation thereof|
WO2009151594A1|2008-06-10|2009-12-17|Teva Pharmaceutical Industries, Ltd.|Rasagiline soft gelatin capsules|
MX2010013766A|2008-06-13|2011-03-15|Teva Pharmaceutical Ind Ltd Star|Rasagiline for parkinson's disease modification.|
NZ589547A|2008-06-19|2013-03-28|Teva Pharma|Dried rasagiline base having a water content less than 0.5 % by weight|
WO2009154782A1|2008-06-19|2009-12-23|Teva Pharmaceutical Industries, Ltd.|Process for purifying rasagiline base|
CN101606923B|2008-06-20|2013-01-09|重庆医药工业研究院有限责任公司|Stable controlled-release rasagiline transdermal patch and preparation method thereof|
WO2011012140A2|2008-07-11|2011-02-03|Synthon Bv|Polymorphs of rasagiline hydrochloride|
WO2010007181A2|2008-07-18|2010-01-21|Medichem, S.A.|New salt forms of an aminoindan derivative|
US20100029987A1|2008-07-29|2010-02-04|Dipharma Francis S.R.I.|Crystalline Form of Rasagiline and Process for the Preparation Thereof|
JP2011529480A|2008-07-30|2011-12-08|ジェネリクス・(ユーケー)・リミテッド|Polymorphism of rasagiline mesylate|
EP2181980A1|2008-10-28|2010-05-05|Chemo Ibérica, S.A.|A process for the preparation of -1-aminoindanes|
DE102008064061A1|2008-12-19|2010-06-24|Ratiopharm Gmbh|Solid composition with the active ingredient rasagiline|
US20100189791A1|2009-01-23|2010-07-29|Teva Pharmaceutical Industries, Ltd.|Delayed release rasagiline malate formulation|
AU2010270254A1|2009-07-09|2012-02-02|Ratiopharm Gmbh|Salts of rasagiline and pharmaceutical preparations thereof|
US20110015274A1|2009-07-20|2011-01-20|Ester Masllorens Llinas|Form of an aminoindan mesylate derivative|
WO2011010324A1|2009-07-23|2011-01-27|Alkem Laboratories Ltd.|Oral pharmaceutical composition of rasagiline and process for preparing thereof|
CA2777185A1|2009-10-09|2011-04-14|Teva Pharmaceutical Industries Ltd.|Use of rasagiline for the treatment of progressive supranuclear palsy|
CN102048717B|2009-10-29|2014-02-19|重庆医药工业研究院有限责任公司|Stable rasagiline composition|
EP2939669A1|2009-12-22|2015-11-04|Teva Pharmaceutical Industries, Ltd.|3-keto-n-propargyl-1-aminoindan|CA2574925A1|2004-07-26|2006-02-09|Teva Pharmaceutical Industries Ltd.|Pharmaceutical dosage forms including rasagiline|
CN101098685A|2004-11-24|2008-01-02|特瓦制药工业有限公司|Rasagiline orally disintegrating compositions|
ES2551481T3|2006-02-21|2015-11-19|Teva Pharmaceutical Industries, Ltd.|Use of rasagiline for the treatment of multisystemic atrophy|
EP2007369A4|2006-04-03|2009-07-01|Teva Pharma|Use of rasagiline for the treatment of restless legs syndrome|
EP1892233A1|2006-08-18|2008-02-27|Ratiopharm GmbH|New salts of the active component rasagiline|
JP5623081B2|2006-12-14|2014-11-12|テバ ファーマシューティカル インダストリーズ リミティド|Crystalline solid rasagiline base|
EP1987816A1|2007-04-30|2008-11-05|Ratiopharm GmbH|Adsorbate of a rasagiline salt with a water-soluble inactive ingredient|
WO2009032273A1|2007-09-05|2009-03-12|Teva Pharmaceutical Industries, Ltd.|Method of treating glaucoma using rasagiline|
US8188149B2|2007-09-17|2012-05-29|Teva Pharmaceutical Industries, Ltd.|Use of R-N-propargy1-1-aminoindan to treat or prevent hearing loss|
NZ586025A|2008-01-11|2012-08-31|Teva Pharma|Rasagiline mesylate tablet coated with a composisiton comprising methacrylic acid - ethyl acrylate copolymer and triethyl citrate|
WO2009151594A1|2008-06-10|2009-12-17|Teva Pharmaceutical Industries, Ltd.|Rasagiline soft gelatin capsules|
MX2010013766A|2008-06-13|2011-03-15|Teva Pharmaceutical Ind Ltd Star|Rasagiline for parkinson's disease modification.|
WO2009154782A1|2008-06-19|2009-12-23|Teva Pharmaceutical Industries, Ltd.|Process for purifying rasagiline base|
NZ589547A|2008-06-19|2013-03-28|Teva Pharma|Dried rasagiline base having a water content less than 0.5 % by weight|
US20100189791A1|2009-01-23|2010-07-29|Teva Pharmaceutical Industries, Ltd.|Delayed release rasagiline malate formulation|
ITPR20090047A1|2009-06-16|2010-12-17|Agricola Cerasaro S S Soc|PROCEDURE FOR THE PRODUCTION OF A TRITO OR GARLIC CREAM AND TRITO OR GARLIC CREAM SO IT IS OBTAINED|
AU2010270254A1|2009-07-09|2012-02-02|Ratiopharm Gmbh|Salts of rasagiline and pharmaceutical preparations thereof|
US9675566B2|2009-07-16|2017-06-13|Pathologica Llc|Method of treatment with anti-inflammatory and analgesic compounds which are GI-, renal-, and platelet-sparing|
CA2777185A1|2009-10-09|2011-04-14|Teva Pharmaceutical Industries Ltd.|Use of rasagiline for the treatment of progressive supranuclear palsy|
CN102048717B|2009-10-29|2014-02-19|重庆医药工业研究院有限责任公司|Stable rasagiline composition|
EP2939669A1|2009-12-22|2015-11-04|Teva Pharmaceutical Industries, Ltd.|3-keto-n-propargyl-1-aminoindan|
CA2806740A1|2010-07-27|2012-02-02|Teva Pharmaceutical Industries Ltd.|Use of rasagiline for the treatment of olfactory dysfunction|
EP2598136A4|2010-07-27|2015-03-25|Teva Pharma|Dispersions of rasagiline citrate|
JP2014503480A|2010-10-26|2014-02-13|テバファーマシューティカルインダストリーズリミティド|Deuterium-rich rasagiline|
HU231054B1|2010-11-18|2020-04-28|Egis Gyógyszergyár Nyrt.|Novel salts suitable for the preparation of pharmaceutical compositions|
EP2665471B1|2011-01-19|2017-12-27|Pathologica, LLC.|Controlled release oral pharmaceutical dosage forms comprising mgbg|
MX2014004201A|2011-10-10|2015-01-12|Teva Pharma|R-n-methyl-propargyl-aminoindan.|
US9346746B2|2011-10-10|2016-05-24|Teva Pharmaceutical Industries, Ltd.|R-N-formyl-propargyl-aminoindan|
EA201490756A1|2011-10-10|2014-09-30|Тева Фармасьютикал Индастриз, Лтд.|CITRAMID RAZAGLININA|
ES2584059T3|2012-03-21|2016-09-23|Synthon Bv|Stabilized pharmaceutical compositions comprising rasagiline salts|
BR112015003451A2|2012-08-17|2017-07-04|Teva Pharma|parenteral formulation of rasagiline|
JP2014050039A|2012-09-03|2014-03-17|Sony Corp|Image processor, image processing method and computer program|
CA2896977C|2013-01-08|2021-12-07|Pathologica Llc|Methods and compositions for treatment of demyelinating diseases|
JP6173098B2|2013-07-30|2017-08-02|キヤノン株式会社|Optical equipment|
JP5897196B1|2015-10-05|2016-03-30|大同化成工業株式会社|Compound granulated product containing sugar or sugar alcohol, swelling binder, disintegrant and superabsorbent excipient, and production method thereof|
CN107049985B|2017-06-07|2020-06-19|广州帝奇医药技术有限公司|Long-acting sustained-release preparation of anti-Parkinson disease drug and preparation method thereof|
CN109028765A|2018-07-02|2018-12-18|无锡富泽药业有限公司|A kind of drying means of tirofiban hydrochloride|
CN109832262A|2019-01-25|2019-06-04|袁隆平农业高科技股份有限公司|Live plant store method|
法律状态:
2019-09-15| MM01| Lapse because of not paying annual fees|Effective date: 20190131 |
优先权:
申请号 | 申请日 | 专利标题
US20583309P| true| 2009-01-23|2009-01-23|
US12/456,031|US20100189788A1|2009-01-23|2009-06-09|Delayed release rasagiline base formulation|
US12/456,001|US20100189791A1|2009-01-23|2009-06-09|Delayed release rasagiline malate formulation|
US12/456,029|US20100189790A1|2009-01-23|2009-06-09|Delayed release rasagiline formulation|
US12/455,976|US8080584B2|2009-01-23|2009-06-09|Delayed release rasagiline citrate formulation|
US12/689,044|US7855233B2|2009-01-23|2010-01-18|Citrate salt of Rasagiline|
[返回顶部]