MXPA00008596A - Pharmaceutical compositions containing the phospholipase inhibitor sodium [[3-(2-amino-1, 2-dioxoethyl)- 2-ethyl-1-phenylmethyl) -1h-indol-4-yl]oxy]acetate - Google Patents

Pharmaceutical compositions containing the phospholipase inhibitor sodium [[3-(2-amino-1, 2-dioxoethyl)- 2-ethyl-1-phenylmethyl) -1h-indol-4-yl]oxy]acetate

Info

Publication number
MXPA00008596A
MXPA00008596A MXPA/A/2000/008596A MXPA00008596A MXPA00008596A MX PA00008596 A MXPA00008596 A MX PA00008596A MX PA00008596 A MXPA00008596 A MX PA00008596A MX PA00008596 A MXPA00008596 A MX PA00008596A
Authority
MX
Mexico
Prior art keywords
composition
lyophilized
acid
solubilizer
percent
Prior art date
Application number
MXPA/A/2000/008596A
Other languages
Spanish (es)
Inventor
William Lester Confer
Hideaki Tai
Original Assignee
William Lester Confer
Shionogi & Co Ltd
Hideaki Tai
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by William Lester Confer, Shionogi & Co Ltd, Hideaki Tai filed Critical William Lester Confer
Publication of MXPA00008596A publication Critical patent/MXPA00008596A/en

Links

Abstract

A lyophilized pharmaceutical composition is described which contains Sodium [[3- (2-amino- 1, 2-dioxoethyl)- 2-ethyl-1-phenylmethyl)- 1H-indol-4- yl]oxy]acetate, a Solubilizer, and Stabilizer. Such compositions are storage stable and readily dissolve in aqueous medium to give injectable solution for treatment of sepsis, etc.

Description

PHARMACEUTICAL COMPOSITIONS CONTAINING THE PHOSPHOLIPASE INHIBITOR [[3 - (2-AMINO-1, 2-DIOXOETHYL) -2-ETHYL-1- (PHENYLEMETIL) -1H-INDOL-4-IL1QXI] SODIUM ACETATE BACKGROUND OF THE INVENTION Field of the Invention This invention relates to pharmaceutical compositions containing a phospholipase inhibitor, more particularly, to a more stable pharmaceutical composition containing [[3- (2-amino-1, 2-dioxoethyl) -2- ethyl-l- (phenylmethyl) -lH-indol-4-yl] oxy] sodium acetate as an Active Ingredient. BACKGROUND [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-1- (phenylmethyl) -lH-indol-4-yl] oxy] sodium acetate (hereinafter referred to as Compound ( I) or Active Ingredient in the specification) is a compound useful as an inhibitor against non-pancreatic secretory human phospholipase A2 (hereinafter called sPLA2), as described in Japanese Patent JP-A 7-285933, Patent application. European number 95302166.4 (EP publication number 0 675 110 AI) and United States of America patent number 5,654,226; whose descriptions are incorporated herein by reference. Injection medications are usually formulated not only in emulsions, suspensions or solutions, but also in lyophilized preparations to be dissolved before use which are stable, can be preserved and easily reconstituted. As it is generally known to those skilled in the art, it is easy to maintain the sterile lyophilized preparations and remove foreign insoluble matter from the lyophilized preparations. With respect to injectable preparations, Japanese Patent JP-A 7-285933 discloses an isotonic saline solution of Compound (I) at a concentration of 0.1 gram / liter and a method for intravenously infusing the saline solution but does not disclose any composition such as of the present invention. A stable pharmaceutical preparation of Compound (I), in particular, a stable injectable preparation of Compound (I) or a lyophilized preparation of Compound (I) to prepare injectable preparations has been desired for a long time. The storage of a pharmaceutical composition of Compound (I) prepared by known methods can lead to the degradation of its content and a coloration and a worsening of its reconstitution properties, especially in the case of injections to be dissolved before use. It is difficult to provide a pharmaceutical composition of Compound (I) which has good stability and a good reconstitution property, especially for injections of Compound (I) to be dissolved before use, after long storage at room temperature. In greater detail, since Compound (I) is a fairly soluble compound, it is difficult to obtain a clear solution of Compound (I) only by mixing Compound (I) with a solvent, which makes it desirable to obtain an injection solution and a solution of processing. Even if a clear solution of the Compound was obtained (I) by changing conditions such as temperature and the like, degradation of a solution of Compound (I) and clouding would occur after long storage, resulting in degradation of the non-uniform contents of Compound (I) in the solution of the lyophilized injectable preparation of Compound (I). One of the problems that are going to be solved by the present invention is to maintain the Compound (I) stable in aqueous carriers. When preparing lyophilized products by the methods of the prior art, sometimes quality problems are caused. These problems include a sublimation of the ingredients that matches the water, the content of the ingredients decreases in the lyophilized product, a partial certification of the product, and the lyophilized preparations fracture and contract and may have a thin layer in the part top of the tablet, adhesion to the upper portion of bottles and splashes, and non-uniform appearance of lyophilized tablets. Pharmaceuticals and the like require injections that are dissolved in an infusion solution before use, for example, a lyophilized injection should have good reconstitution properties. Another problem that is to be solved by the present invention is to provide an excellent lyophilized preparation of Compound (I) of good quality with a uniform appearance, good reconstitution and without degradation of its contents during storage. COMPENDIUM The inventors of the present invention studied various additive agents for preparations of Compound (I) to solve the aforementioned problems. As a result, by adding a solubilizer, preferably together with a stabilizer, in a preparation with Compound (I), the present inventors discovered improved preparations of Compound (I) having the desired qualities such as non-degradation of their content, good stability, a uniform appearance and a good reconstitution property. This invention is a storage stable solid lyophilized composition of Compound (I) suitable for making a liquid useful for the treatment of inflammatory diseases (including sepsis) in mammals. This invention is also an aqueous solution of a lyophilized composition of Compound (I) useful for the treatment of inflammatory diseases (including sepsis) in mammals.
This invention is also a method of making a lyophilized composition reconstituted easily and stable to the storage of Compound (I). This invention is also an improved method for treating inflammatory diseases in mammals using the lyophilized composition of Compound (I). This invention is also an intravenous unit dosage bag or bottle containing a lyophilized composition of Compound (I) dissolved in aqueous medium at a convenient concentration to direct injection in a mammal for the treatment of inflammatory diseases. (including sepsis). This invention is a concentrated dose sealed container containing a solid lyophilized composition of Compound (I). DETAILED DESCRIPTION Definitions: The term "lyophilized composition or compositions" refers to a cold-dried solid composition of material prepared by the process of this invention and comprising as essential ingredients (1) [[3- (2-amino-1, Sodium 2-dioxoethyl) -2-ethyl-1- (phenylmethyl) -lH-indol-4-yl] oxy] acetate; (2) a solubilizer; and (3) a stabilizer. The term, "Active Ingredient" (also called Compound (I) ") refers to the compound [[3- (2-amino-1, 2-dioxoethyl) -2-ethyl-1- (phenylmethyl) -lH-indole- Sodium 4-yl] oxy] acetate, a compound represented by the formula: The term, "mammal", includes a human as well as important veterinary species related to mammals, domesticated quadrupeds such as monkeys, dogs, cats, horses, sheep, pigs, goats, and cows. The term "inflammatory diseases" refers to diseases such as inflammatory bowel disease, sepsis, septic shock, adult respiratory distress syndrome, pancreatitis, trauma induced shock, bronchial asthma, allergic rhinitis, rheumatoid arthritis, cystic fibrosis, seizure, acute bronchitis, chronic bronchitis, acute bronchitis, chronic bronchiolitis, osteoarthritis, gout, spondylarthropathy, ankylosing spondylitis, Reiter's syndrome, psoriatic arthropathy, enteropatric spondylitis, juvenile arthropathy or juvenile ankylosing spondylitis, reactive arthropathy, infectious or postinfectious arthritis, gonococcal arthritis, arthritis due to tuberculosis, viral arthritis, fungal arthritis, syphilitic arthritis, Lyme disease, arthritis associated with "vasculitic syndromes", polyarteritis nodosa, vasculitis hypersensitivity, granulomatosis of Luegenec, polymialgina rheumatica, arteritis of joint cells, arthropathy calcium deposit, pseudo-gout, non-articular rheumatism, bursitis, tenosynovitis, epicondylitis (tennis shoulder), carpal tunnel syndrome, repetitive use damage (typing), miscellaneous forms of arthritis, neuropathic joint disease (from charcot and articulation), hemarthrosis (hemarthrosis), Henoch-Schonlein purpura, hypertrophic osteoarthropathy, multicentric reticulohistiocytosis, arthritis associated with certain diseases, sulcoilosis, hemochromatosis, sickle cell disease and other hemoglobinopathies, hyperlipoprote and inemia, hypogamagobulinemia, hyperparathyroidism, acromegaly, Mediterranean family fever, Behat's disease, systemic lupus erythematosus, or relapsing polychondritis and related diseases included or maintained by the release of fatty acid sPLA2 and the cascade of arachidonic acid and its harmful products. The term "collapse temperature" describes the glass transition temperature for amorphous solids or the eutectic temperature for crystalline solids. The collapse temperature is the temperature above which the product is not completely frozen. The dry freezing microscopy allows the measurement of the temperature at which the frozen solutions begin to lose their rigid structure during a sublimation process. For the frozen solutions of [[3 - (2-amino-1, 2-dioxoethyl) -2-ethyl-1 - (phenylmethyl) -lH-indol-4-yl] oxy] sodium acetate, the collapse temperature before annealing has been measured at about -33 ° C while the collapse temperature after annealing is about -13 ° C. The term "solubilizer" refers to a chelating agent. An "effective amount of solubilizer" is an amount of solubilizer that allows the Active Ingredient to form stable aqueous solutions suitable for medical use. The term "stabilizer" refers to a solid sugar or an alcohol sugar. An "effective amount of stabilizer" is a stabilizing amount that allows the lyophilized composition to dissolve easily to form aqueous solutions suitable for medical use. The term, "dose concentrate" refers to a pharmaceutical formulation solution. The dose concentrate can be maintained in the container where it was formed by adding aqueous solvent to the pharmaceutical formulation or it can be removed and maintained externally. The dose concentrate may be used as is, but is generally further diluted to a unit dose concentration for administration to a mammal. The entire volume of dose concentrate or aliquots thereof can be used to prepare unit doses for treatment by the method of this invention. The term "the equivalent acid of the Active Ingredient" or "the equivalent acid of Compound (I)" means [[3- (2-amino-1, 2-dioxoethyl) -2-ethyl-l- (phenylmethyl) -1H] -indol-4-yl] oxy] acetic acid. In this specification, the weight of the Active Ingredient is shown by the actual weight of [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-l- (phenylmethyl) -lH-indole-4-yl] oxy] sodium acetate. To show the property (percentage by weight) of the Active Ingredient, however, this is calculated based on the equivalent acid of the Active Ingredient. In this way the weights of the Active Ingredient must be multiplied by the factor 100 / 105.8 to calculate the equivalent weight of the equivalent acid. Methods for Making the Active Ingredient: Compound (I) for use in the present invention can be synthesized by the known process described in JP-A 7 285933 and can be used for the compositions and preparations of the present invention in any condition, including crystal, amorphous, hydrate, solvate, or a mixture of these forms. Similarly, the Compound (I) can be prepared by synthesis schemes taught in the United States Patent Number ,654,326; whose description is incorporated herein by reference. Another method of making Compound (I) is described in the United States patent application of North America serial number 09/105381, filed June 26, 1998 and entitled, "Process for preparing 4-substituted l-H-Indol-3-glycoxyamides", the entire disclosure of which is incorporated herein by reference. An illustrative synthesis of Compound (I) is as follows in steps (A) through (F): Preparation of [[3- (2-amino-1, 2-dioxoethyl) -2-ethyl-1- (phenylmethyl) - sodium lH-indol-4-yl] oxy] acetate, a compound represented by the formula: Step A. Preparation of 2-Ethyl-4-methoxy-1H-indole: A solution of 140 milliliters (0.18 mole) of 1.3M sec-butyl lithium in cyclohexane was slowly added to N-tert-butoxycarbonyl-3-methoxy-2 -metilaniline (21.3 grams 0.09 mol) in 250 milliliters of THF keeping the temperature below -40 ° C and with an ethanol bath on dry ice. The bath was removed and the temperature allowed to rise to 0 ° C and then the bath was replaced. After the temperature was cooled to -60 ° C, 18.5 grams (018 mol) of N-methoxy-N-methylpropanamide in equal volume of THF was added dropwise. The reaction mixture was stirred for 5 minutes, the cooling bath was stirred and stirred for an additional 18 hours. It was then poured into a mixture of 300 milliliters of ether and 400 milliliters of 0.5N HCl. The organic layer was separated, washed with water, brine, dried over MgSO 4 and concentrated under reduced pressure to give 25.5 grams of a crude l- [2- (tert-butoxycarbonylamino) -6-methoxyphenyl] -2- butanone. this material was dissolved in 250 milliliters of methylene chloride and 50 milliliters of trifluoroacetic acid and stirred for a total of 17 hours. The mixture was concentrated under reduced pressure and ethyl acetate and water were added to the remaining oil. The ethyl acetate was separated, washed with brine, dried (MgSO 4) and concentrated. The residue was chromatographed three times on silica oxide eluting with 20 percent EtOAc / hexane to give 13.9 grams of 2-ethyl-4-methoxy-1H-indole.
Step B. Preparation of 2-ethyl-4-methoxy-1- (phenylmethyl) -lH-indole: 2-Ethyl-4-methoxy-1H-indole (4.2 grams, 24 mmol) was dissolved in 30 milliliters of DMF and 960 milligrams (24 mmol) of 60 percent NaH / mineral oil was added. After 1.5 hours, 2.9 milliliters (24 mmol) of benzyl bromide was added. After 4 hours, the mixture was diluted with water and extracted twice with ethyl acetate. The combined ethyl acetate was washed with brine, dried (MgSO 4) and concentrated under reduced pressure. The residue was chromatographed on silica gel and eluted with 20 percent EtOAc / hexane to give 3.1 grams (49 percent yield) of 2-ethyl-4-methoxy-1- (phenylmethyl) -1H-indole. .
Step C. Preparation of 2-ethyl-4-hydroxy-1- (phenylmethyl) -1H-indole: 3.1 grams (11.7 mmol) of 2-ethyl-4-hydroxy-1- (phenylmethyl) -lH-indole is O- demethylated by treating with 48.6 milliliters of 1M BBr 3 in methylene chloride with stirring at room temperature for 5 hours, followed by concentration under reduced pressure. The residue was dissolved in ethyl acetate, washed with brine and dried (MgSO 4). After concentrating under reduced pressure, the residue was chromatographed on silica gel eluting with 29 percent EtOAc / hexane to give 1.58 grams (54 percent yield) of 2-ethyl-4-hydroxy-1- (phenylmethyl) - lH-indole.
Step D: preparation of [[2-ethyl-1- (phenylmethyl) -lH-indol-4-yl] oxy] acetic acid methyl ester. 2-ethyl-4-hydroxy-1- (phenylmethyl) -lH-indole (1.56 grams 5.2 mmol) was added to a mixture of 248 milligrams (6.2 mmol) of 60 percent NaH / mineral oil in 20 milliliters of DMF and stirred for 0.67 hours. Then 0.6 milliliters (6.2 mmol) of methyl bromoacetate was added and stirring was continued for 17 hours.
The mixture was diluted with water and extracted with ethyl acetate. The ethyl acetate solution was washed with brine, dried (MgSO4), and concentrated under reduced pressure. The residue was chromatographed on silica gel eluting with 20 percent EtOAc / hexane to yield 1.37 grams (69 percent yield) of methyl ester of [[2-ethyl-1- (phenylmethyl) -lH-] acid. indole-4-yl] oxy] acetic acid.
Step E. Preparation of [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-1- (phenylmethyl) -lH-indol-4-yl] oxy] acetic acid methyl ester: Oxalyl chloride (0.4 milliliters, 4.2 mmol) was added to 1.36 grams (4.2 mmol) of [[2-ethyl-1- (phenylmethyl) -lH-indol-4-yl] oxy] acetic acid methyl ester in 10 milliliters of chloride of methylene and the mixture was stirred for 1.5 hours. The mixture was concentrated under reduced pressure and the residue was taken up in 10 milliliters of methylene chloride. Anhydrous ammonia was bubbled for 0.25 hours, the mixture was stirred for 1.5 hours and evaporated under reduced pressure. The residue was stirred with 20 milliliters of ethyl acetate and the mixture was filtered. The filtrate was concentrated to give 1.37 grams of a mixture of [[3- (2-amino-1,2-d-oxoethyl) -2- ethyl-1- (phenylmethyl) -lH-indole-4-methyl ester. il] oxy] acetic acid and ammonium chloride This mixture melted at 172-187 ° C.
Step F. Preparation of [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-1- (phenylmethyl) -lH-indol-4-yl] oxy] acetic acid, sodium salt: A mixture of 788 milligrams (2 mmol) of [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-1- (phenylmethyl) -1H-indol-4-yl] oxy] acetic acid methyl ester, 10 milliliters in NaOH and 30 milliliters of MeOH was heated to maintain reflux for 0.5 hours, stirred at room temperature for 0.5 hours and concentrated under reduced pressure.
Identity and proportion of ingredients in the pharmaceutical compositions and lyophilized compositions of the invention: In one embodiment, the present invention is a pharmaceutical composition comprising Compound (I) as an Active Ingredient and an effective amount of a solubilizer that acts as an active ingredient. chelating agent, for example, preferably at least one compound selected from citric acid, edetic acid (e.g., disodium EDTA), polyphosphoric acid and its salts, more preferably sodium citrate. Examples of polyphosphoric acid and its salts are potassium polyphosphate as described in the Japanese standards of food additives, sixth edition, and sodium polyphosphate as described in the Japanese standards of food additives, sixth edition, or Japanese standards of cosmetic ingredients, second edition. Sodium citrate is available as anhydrous trisodium citrate, trisodium citrate dihydrate, and trisodium citrate pentahydrate, but it is more convenient and preferred to use it in the form of trisodium citrate dihydrate (molecular weight 294.10). The amount of solubilizer varies with the class of solubilizer and with the concentration of Compound (I), and can be from about 1 percent to about 400 percent (w / w), preferably 1 to 200 percent (w / w) ), more preferably 100 percent (weight / weight) of the equivalent acid amount of the Compound (I) For pharmaceutical compositions using sodium citrate the weight of the solubilizer is from 10 percent to 150 percent (w / w) and more preferably 25 percent to 100 percent (w / w) of the equivalent acid amount of the Compound (I) Preferably, the pharmaceutical composition described in the preceding paragraph also has an effective amount of stabilizer. The stabilizer is at least one pharmaceutically acceptable compound selected from solid sugars and sugar alcohols, more preferably at least one compound selected from mannitol, xylitol, sorbitol, glucose, fructose, lactose and maltose. Mannitol is the most preferred stabilizing ingredient. The amount of stabilizer varies with the class of stabilizer and with the concentration of Compound (I), and can be 40 percent to 500 percent (w / w), preferably 50 percent to 300 percent (w / w), more preferably 50 to 250 percent (w / w), more preferably 50 percent to 200 percent (w / w) of the equivalent acid amount of Compound (I). Without departing from the object and scope of the present invention, other pharmaceutically acceptable additive agents may be optionally added to the preparations of the present invention. When a solution according to the invention is prepared for injection, an isotonizing agent, a calming agent or other additives may be added thereto. Preferably, the pharmaceutical compositions described above have no salt except for the Active Ingredient, the solubilizer and the stabilizer.
Lyophilized compositions of the invention: Preferably, the pharmaceutical compositions described in the preceding section are lyophilized. More preferably the lyophilized composition is prepared with an annealing step employing the collapse temperature characteristics of Compound (I). The lyophilized composition contains solubilizer from about 1 to about 200 percent (weight / weight) of the equivalent acid amount of the Compound (I) The proportions of the solubilizer are the same as those presented in the previous section for the pharmaceutical composition. When the solubilizer is disodium EDTA (or its acid or other salts) preferably from about 1 percent to about 15 percent (w / w) of the equivalent amount of the acid is used.
Compound (I). When the solubilizer is trisodium citrate dihydrate preferably from about 10 percent to about 100 percent of the amount of the Active ingredient. The identity and proportions of the stabilizer are the same as those established in the previous section for the pharmaceutical composition. Mannitol is most preferred as the stabilizing ingredient of the lyophilized compositions of the invention. Table 1. List of the preferred lyophilized compositions specific to the invention (all amounts in milligrams) Table 1 I.A. = Active Ingredient, Citrate Na = trisodium citrate dihydrate, EDTA = ethylenediaminetetraacetic acid, disodium salt. Preferably, the solid lyophilized compositions of the invention are substantially free of salt, except for Compound (I) and the stabilizer and solubilizer contained therein. The lyophilized pharmaceutical formulation can be dissolved in a pharmaceutically acceptable carrier, such as sterile water, optionally sterile water containing saline and / or sterile water containing sugars. For example, for intravenous injection the compositions of the invention can be dissolved in a concentration of 2 milligrams / milliliter in an aqueous solution of 4 percent dextrose / 0.5 percent sodium citrate.
As used herein, the terms "lyophilized compositions", "pharmaceutical compositions" and "pharmaceutical preparations" refer to all preparations described in "general rules for preparation" in the Japanese Pharmacopoeia, preferably those that are preparations of injection solutions, more preferably solutions for injection and lyophilized preparations for injection. Methods for Making the Lyophilized Compositions of the Invention: The lyophilized compositions of the present invention relate to a preparation prepared by freeze drying a solution containing Compound (I), which is optionally subjected to the heat treatment process, and drying it at high vacuum to sublimate the water. These lyophilized preparations include lyophilized preparations for injection as mentioned above. The lyophilized preparation can be produced by conventional methods including pan lyophilization, spray lyophilization and bottle lyophilization methods. Bottle lyophilization is advantageous for preparing units from multiple doses of the invention as described, below. In order to obtain a solution of a Compound (I) by the process of the present invention,. Compound (I), a solubilizer and a solvent are mixed and stirred until the mixture becomes clear. The solvent is preferably an aqueous solvent such as water, purified water, water for injection, only isotonic sodium chloride or glucose injection as described in the Japanese Pharmacopoeia, more preferably a salt-free aqueous solvent such as water, purified water , water for injection or glucose solutions for injection. Alternatively, a suitable solvent for forming a solution from the composition of the invention is any injectable solution as exemplified below by those described in the Pharmacopeia of the United States of America (1995, ISBN 0195-7996), for example, " sterile water for injection "," dextrose and sodium chloride injection "," dextrose injection ", mannitol injection" or "mannitol injection in sodium chloride." In order to obtain a lyophilized solution of Compound (I) prior to lyophilization of the present invention, first, a processing solution prior to lyophilization is prepared.The processing solution before lyophilization is a solution prepared by mixing and stirring Compound (I), a solubilizer and a solvent, preferably Compound (I), a solubilizer and a stabilizer and a solvent, until the mixture becomes clear For the sequence of addition of the ingredients to the solvent and it first prefers to dissolve the solubilizer and stabilizer, and thereafter dissolve Compound (I). The solvent is preferably an aqueous solvent such as that previously presented above and described in the Japanese Pharmacopoeia, and more preferably an aqueous, salt-free solvent, such as water, water for injection or glucose injection. The processing solution before lyophilization of Compound (I) may contain Compound (I) at a concentration of from about 0.5 percent to 2 percent (w / w). If desired, the processing solution before lyophilization can be subjected to a filtration process. The filtration process includes, for example in the case of injection preparations, a sterilizing filtration and / or an ultrafiltration of the processing solution prior to lyophilization to remove micro-organisms or other contaminating materials from the processing solution before the lyophilization. If desired, the processing solution before lyophilization can be subjected to a distribution process. The distribution process includes, for example in the case of bottle lyophilisations, a process that distributes a suitable volume of the processing solution before freeze-drying into bottles taking the concentration of Compound (I) into consideration so that the The products of the bottle carry a desired amount of Compound (I).
A lyophilization process is carried out as follows: Preferably, the lyophilized composition is prepared by a heating and cooling process. A process for preparing a lyophilized composition comprises the sequential steps of: (a) dissolving the ingredients of the lyophilized composition comprising [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-1-acetate] sodium (phenylmethyl) -lH-indol-4-yl] oxy] acetate, solubilizer and stabilizer in an aqueous solvent; (b) cooling the processing solution from step (a) to a temperature below 33 ° C; (c) heating the product from step (b) to a temperature above -33 ° C; (d) cooling the product of step (c) to a temperature below -33 ° C; (e) heating the product from step (d) to a temperature above -13 ° C, under subatmospheric pressure for a sufficient time to remove the water from the aqueous solvent and produce a solid lyophilized product. Preferably, step (a) is carried out by dissolving in an aqueous solvent: [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-1- (phenylmethyl) -lH-indol-4-yl ] oxi] sodium acetate, -solubilizer selected from citric acid, edetic acid, polyphosphoric acid and its salts, the amount of which is 1 to 100 percent (w / w) of the equivalent acid amount of Compound (I); and stabilizer selected from mannitol, xylitol, sorbitol, glucose, fructose, lactose and maltose, the amount of which is 50 to 200 percent (w / w) of the equivalent acid of Compound (I). Moreover, each of steps (b), (c), (d) and (e) is preferably carried out for a period of at least half an hour, and step (e) is carried out at a lower subatmospheric pressure of approximately 133 Pa (1000 milliTorr). The preferred parameters in the lyophilization process are those in which Compound (I) is frozen by cooling it to -35 ° C to -45 ° C. This cooling step is preferably carried out for 2 to 4 hours. This process in the present is called the "primary freezing process". If desired, the frozen solution obtained in the primary freezing process is heated to -5 ° C to -25 ° C preferably -10 ° C to -20 ° C. This heating step is carried out for 3 hours, preferably 5 to 10 hours. This process in the present is referred to as the "heat treatment process". The composition obtained in the heat-heating process is refrozen, preferably from -35 ° C to -45 ° C. This cooling step is preferably carried out for 2 to 4 hours. This process in the present is referred to as the "refreezing process". The composition obtained through the primary freezing process, the heat treatment process and the refreezing process is dried under high vacuum by subliming the water according to methods known to those skilled in the art. Thus, a lyophilized preparation? The present invention is obtained. If desired, drying two steps in which the temperature and degree of vacuum are different can be done to completely remove the water. This process later in the present is called the "drying process". If two steps are dried, these processes are known as the "primary drying" process and the "secondary drying" process. The lyophilization process removes most of the water originally present, but the final product of lyophilized composition may contain some free water. Typically, the water content can vary from 0.5 percent to 5.0 percent by weight. More typically, the water content varies from 0.8 percent to 2.0 percent.
Concentrated dose and unit dose configurations of the invention: A concentrated dose configuration of the formulation of the invention is a sealed container containing a quantity of lyophilized pharmaceutical formulation of the invention employed over a standard treatment range such as from 12 to 24 hours. The concentrated dose configuration is prepared by placing the lyophilized composition in a container (e.g., glass or plastic bottles, flasks, ampoules) in an amount sufficient to treat a mammal for a period ranging from one hour to one week, but preferably from 4 hours to 48 hours. The container preferably also contains a void space of sufficient size to allow (i) the addition of aqueous solvent plus (ii) additional space as necessary to allow agitation and effect complete dissolution of the lyophilized composition in the added aqueous solvent. The container can be equipped with a penetrable upper part, for example, a rubber seal, so that the aqueous solvent can be added by penetrating the seal with a hypodermic syringe and subsequently removing the concentrate by the same method. An example of a concentrated dose setting is a glass bottle having a capacity of from about 10 to about 100 milliliters containing from 50 to 5000 milligrams of lyophilized pharmaceutical composition. A specific example is a glass bottle 20 with a rubber seal having the lyophilized pharmaceutical composition containing 105.8 milligrams of Compound (I), 50 milligrams of sodium citrate dihydrate, and 200 milligrams of mannitol. The void space on the solid composition has ample space for the addition of a solvent such as sterile water for injection plus space to stir the total contents. The addition of the aqueous solvent to the concentrated dose configuration results in a liguid concentrate which can then conveniently be used to form a unit dose of liquid pharmaceutical formulations by removing aliquots or whole contents for dilution as presented in the next section. Unit dose of the invention: The concentrated solution of lyophilized composition formed in the concentrated dose container (typically a bright yellow color) is added to an intravenous (IV) container containing a suitable aqueous solvent. Useful solvents are standard solutions for injection as previously described (e.g., 5 percent dextrose or sterile water for injection, etc.). The intravenous unit dose bags are conventional glass or plastic containers that have inlet and outlet elements and have standard capabilities (eg, 250 milliliters and 500 milliliters). The concentrated solution of lyophilized pharmaceutical formulation is added to the intravenous unit dose bag in an amount to achieve a concentration of about 0.05 to 2.0 milligrams of Compound (I) per milliliter and preferably 0.2 to 0.8 milligrams per milliliter.
Without departing from the object and scope of the present invention, other pharmaceutically acceptable additive agents can be added to the lyophilized preparations of the present invention. When the lyophilized preparation for injection is to be used, an isotonizing agent or a calming agent or other additives may be added thereto. Use of the composition of the invention for the treatment of inflammatory diseases: The improved method of treatment using the lyophilized composition can be practiced as follows: The diluted formulations of this invention are given by injection, either subcutaneously or in muscle tissue by injection in a vein Intravenous injection is the preferred mode of administration to the mammal being treated and offers the advantage of rapid effect and rapid access to the circulatory system particularly in emergency situations. Preferably the lyophilized pharmaceutical formulation of this invention is diluted with aqueous solvent suitable for injection and a prepared unit dosage form (ie, intravenous bag) per administration to a mammal. An intravenous bag for pediatric use can have a capacity of 100 milliliters. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and condition of the patient. The specific dose of a compound administered according to this invention to obtain therapeutic or prophylactic objects will, of course, be determined by the particular circumstances surrounding the case, including, for example, the compound administered, the route of administration and the disease that is being treated. trying. Typically the daily doses will contain a non-toxic dosage level of compound (I) of from about 0.01 milligrams / kilogram to about 50 milligrams / kilogram of body weight of an Active Ingredient of this invention. This invention is a method of treating or preventing sepsis by administering to a mammal in need thereof a therapeutically effective amount of a solution prepared by dissolving the pharmaceutical formulation of the present invention. The administration to a septic patient can be either continuous or intermittent. The decision to begin therapy for sepsis will be based on the appearance of clinical manifestations of sepsis or laboratory tests that show the onset of the sepsis cascade (including renal complications or coagulation abnormalities or multiple organ failure). Typical clinical manifestations are fever, tremor, taguicardia, taguipnea, altered mental status, hypothermia, hyperthermia, accelerated or repressed breathing or heart rates, increased or decreased leukocyte cell counts, and hypotension. These and other symptoms are well known in the art and are presented in standard references such as Harrison's Principles of Internal Medicine (ISBN 0-07-032270-4; 1994, pages 511-515) The decision to determine the duration of the therapy it can be applied for the results of standard clinical laboratories of commercially available tests or instrumentation that supports the eradication of the symptoms that define sepsis The method of the invention can be practiced by administering continuously or intermittently a therapeutically effective dose of the prepared solution. from the lyophilized pharmaceutical formulation as long as it is considered effective for the treatment of the septic episode.The administration can be carried out for a total of approximately 60 days with a preferred course of therapy lasting up to 10 days. the therapy by the method of the invention can be supported by laboratory results Clinical standards of commercially available trials or instrumentation of the disappearance of clinical symptoms characteristic of sepsis. Therapy can be resumed if sepsis returns. Pediatric forms of sepsis are also successfully treated by the methods and compositions of this invention.
Examples The present invention is described more specifically and is explained by the following examples, without being limited in any way: Example 1 and Comparative Example 1: Solutions were prepared by mixing the materials of Table 2 with a solvent, the solvent being one of water for injection, isotonic sodium chloride solution and 5 percent glucose solution in a volume of 20 milliliters . Table 2 Preparation of solutions I.A. = Active Ingredient, Citrate Na = trisodium citrate dihydrate The tests for solubility were carried out by inspecting the solutions immediately after they were prepared and after 24 hours of storage at room temperature with direct view. Tables 3 to 5 show the results for the solubility tests. Each table clearly shows that the addition of the solubilizer of the present invention improves the stability of the solution. Table 3 Solubility test result (20 milliliters of water 1 added to the preparation of Table 1) Table 4 Solubility test results (20 milliliters of saline 2 added to the preparation of the Table 1) Table 5 Solubility test result (20 milliliters of glucose 3 solution added to the preparation of Table 1) Explaon of the symbols in Tables 2 to 4 0 = excellent solution? = crystals appear slightly in solution 1 water = water for injection 2 saline = isotonic sodium chloride solution 3 glucose solution = 5 percent glucose solution.
Examples 2 to 4 and Comparative Example 2: The processing solution before lyophilization was prepared by dissolving the materials of Table 6 in water for injection as a solvent as described in the Japanese Pharmacopoeia so that the concentration of Compound (I) It was 10 milligrams / gram. The processing solution before lyophilization was subjected to sterilizing filtration using a membrane filter of which the pore size was 0.22 microns and was then distributed in glass jars. The lyophilization was carried out as follows. The primary freezing process was done for 3 hours at -40 ° C, the heat treatment process for 10 hours at -10 ° C, and the re-cooling process for 3 hours was at -40 CC. Then, the primary drying process was carried out for 24 hours at 0 ° C, 2.0 Pa and the secondary drying process for 5 hours at 60 ° C, 2.0 Pa. In this way, the lyophilized preparations of Examples 2 and 4 were obtained. For comparison, the preparation of Comparative Example 2 was produced by the method in which the filtration process and the distribution process was the same as that of Examples 2-4 and the lyophilization was substantially equal to the process of Examples 2- 4, this is the primary freezing process was done for 3 hours at -40 ° C, the primary drying weight for 20 hours at 10 ° C, 10 Pa, and the secondary drying step for 5 hours at 40 CC, 2.0 Pa. Table 6 Composition of lyophilized preparations (in bottle 1) I.A. = Active Ingredient, Citrate Na = trisodium citrate dihydrate Table 7 shows the appearance of lyophilization tablets immediately after lyophilization in Examples 2 to 4. The addition of the stabilizer of the present invention gives excellent lyophilized preparations in which the lyophilized tablet has no fractured parts, shrunken parts and a thin layer of the surface of the tablet and in which the tablet does not have much color. Table 7 Appearance of the tablet after lyophilization This tablet has no fractured parts, shrunken parts and a thin layer on the surface of the tablet. A uniformity test of the content of Compound (I) of the 10 bottles of the lyophilized preparation obtained in Example 4 was carried out. The content of Compound (I) was measured by the high performance, liquid chromatography method. The content of Compound (I) in a bottle was 99.0 percent up to 103.4 percent relative to the theoretical value of the amount of Compound (I), which indicates that the lyophilized preparation of the present invention had satisfactory content uniformity. The reconstitution times of 10 bottles of the lyophilized preparation obtained in Example 4 were measured. The measurement of the reconstitution time was carried out by adding the sample to 10 milliliters of water for injection, stirring the resulting mixture at 200 times / millimeter, and measuring the period of time until the mixtures were completely dissolved. The reconstitution time was 7 to 18 seconds. The lyophilized preparation of the present invention had a satisfactory reconstitution property. Three hermetic bottles of the lyophilized preparation of Example 2 were tested for storage stability at 60 ° C for 2 months. The residual percentages of the content in three bottles were all 99 percent in relation to the initial value immediately after lyophilization. The lyophilized preparation of the present invention had satisfactory storage stability. Example 5: The processing solution before lyophilization was prepared by dissolving the materials of Table 8 in water for injection as a solvent as described in the Japanese Pharmacopoeia so that the concentration of Compound (I) was 15 milligrams / gram . The processing solution before lyophilization was subjected to sterilization filtration using a membrane filter of which the pore size was 0.22 microns and was then distributed in glass jars. Table 8 Composition of lyophilized preparations (in bottle 1) I.A. = Active Ingredient, Na Citrate = trisodium citrate dihydrate The lyophilization was carried out as follows. The freezing step was done for 3 hours at -40 ° C, the heat treatment step for 10 hours at -10 ° C, and the refreezing step for 3 hours at -40 ° C. Then, the primary drying step was carried out for 60 hours at 0 ° C, 10 Pa and the secondary drying step for 5 hours at 60 ° C, 4 Pa. In this way the lyophilized preparation of Example 5 was obtained. The appearance of the tablet immediately after lyophilization in Example 5 was observed. The lyophilized tablet had no broken parts, shrunken parts and a thin layer on the surface of the tablet and the color of the tablet was dark white. The uniformity test of content on the Compound (I) of 10 bottles of the lyophilized preparation obtained in Example 5 was made. The content of Compound (I) was measured by the high performance liquid chromatography method. The content of Compound (I) per bottle was from 99.8 to 101.3 percent relative to the theoretical value of the amount of Compound (I), which indicates the lyophilized preparation of the present invention had satisfactory content uniformity. The reconstitution time of 10 bottles of the lyophilized preparation obtained in Example 5 was measured. The measurement of the reconstitution time was carried out by adding 10 milliliters of water for injection to each of the bottles, stirring the resulting mixture at 200 times / millimeter, and measuring the period of time until the tablet completely dissolved. The reconstitution time was 22 to 29 seconds. The lyophilized preparation of the present invention had a satisfactory reconstitution property. Example 6: Another example for preparing the lyophilized composition of the invention is as follows: Preparation of a convenient solution for lyophilization (processing solution): (A) A volume of water equal to about 80 percent of the final volume of the pharmaceutical lyotilized composition was placed in the manufacturing vessel. (B) Mannitol and sodium citrate were first added to the vessel and stirred until complete dissolution was carried out. (C) Sodium [3- (2-amino-1,2-dioxoethyl) -2-ethyl-l- (phenylmethyl) -lH-indol-4-yl] oxy] acetate was added to the vessel and stirred until that completely dissolved. (D) The remaining water was added to achieve a final Active Ingredient concentration of 10 milligrams per milliliter. The sequential process of lyophilization was carried out as follows: 1. Sealable bottles containing 10 milliliters of the processing solution prepared in the previous step of this example were placed on the shelves of a freeze dryer pre-cooled to 0 ° C. 2. After the bottles were loaded onto the shelves, the shelf temperature was cooled to -40 ° C. As soon as the temperature of the product of the monitored bottles is -40 ° C, the bottles are kept at this temperature for 2 hours. 3. The temperature on the shelf was raised to -10 ° C to anneal the frozen solution. This temperature was above the collapse temperature of the solution which allowed the crystallization of the solutes. The product was kept at this temperature for several hours. 4. The temperature in the rack then decreased to -40 ° C again for a short period of time to complete the annealing process (temperature cycling). 5. Primary drying was then initiated by increasing the shelf temperature to 0 ° C and decreasing the chamber pressure to approximately 13.3 Pa (100 milliTorr). The primary drying lasted for more than 24 hours until the temperature of the product began to rise, signaling that the solvent water was removed. 6. Secondary drying was achieved by raising the chamber temperature to 28 ° C, leaving the chamber pressure at 13.3 Pa (100 milliTorr) and lasting for several hours. The product was an unbleached, solid-fractured tablet with a dull white color. As the previous examples show, the present invention provides pharmaceutical compositions of Compound (I) that are stable and have good reconstitution properties. It will be apparent to those skilled in the art that various modifications and variations may be made to the compositions and processes of this invention. Thus, it is intended that the present invention cover these modifications and variations, as long as they are within the scope of the appended claims and their equivalents. The description of all publications cited above is expressly incorporated herein by reference in its entirety to the same extent as if they were incorporated by reference individually.

Claims (27)

1. A composition comprising [[3- (2-amino-l, 2-dioxoethyl) -2-ethyl-l- (phenylmethyl) -lH-indol-4-yl] oxy] -acetate as active ingredient; a solubilizer selected from at least one compound selected from the group consisting of citric acid, edetic acid, polyphosphoric acid and its salts, - and a stabilizer selected from at least one compound selected from the group consisting of mannitol, xylitol, sorbitol, glucose, fructose, lactose and maltose. The composition of claim 1 wherein the solubilizer is sodium citrate dihydrate or disodium EDTA; and the stabilizer is mannitol. 3. The composition of claim 1 wherein the composition is lyophilized. 4. The composition of claim 3 wherein the solubilizer is sodium citrate and the stabilizer is mannitol. The composition of any of claims 1, 2, 3, or 4 wherein by the amount of the equivalent acid of the Active Ingredient, the stabilizer is present from 1 to 100 percent (w / w) and the solubilizer is present at 50 to 200 percent (weight / weight). 6. The composition of any one of claims 1, 2, 3, 4, or 5 that is free of salt, except for the Active Ingredient, the solubilizer and the stabilizer. 7. A method for making the pharmaceutical formulation of the invention, comprising the steps of: a) dissolving in an aqueous solvent a composition comprising [[3- (2-amino-1,2-dioxoethyl) -2-ethyl- sodium l- (phenylmethyl) -lH-indol-4-yl] oxy] acetate as an Active Ingredient, a solubilizer selected from at least one compound selected from the group consisting of citric acid, edetic acid, polyphosphoric acid and its salts, - and a stabilizer selected from at least one compound selected from the group consisting of mannitol, xylitol, sorbitol, glucose, fructose, lactose and maltose; b) cooling the processing solution from step (a) to a temperature below about -33 ° C; c) heating the product from step (b) to a temperature above about -33 ° C; d) cooling the product of step (c) to a temperature below about -33 ° C; e) heating the product of step (d) to a temperature above about -13 ° C, under subatmospheric pressure for a sufficient time to remove the water from the aqueous solvent and produce a solid lyophilized product. 8. The process of claim 7 wherein: in step (a) the amount of solubilizer is 100 percent (w / w) of the amount of the eguivalent acid of the Active Ingredient and the stabilizing amount is 50 to 200 percent (weight / weight) of the amount of the eguivalent acid of the Active Ingredient. The process of claim 7 wherein: steps (b), (c), (d) and (e) each are carried out for a period of at least half an hour and step (e) is carried out at a subatmospheric pressure of less than 133 Pa (1000 milliTorr). The process of claim 7 wherein the solubilizer is sodium citrate and the stabilizer is mannitol. 11. A lyophilized composition prepared by the process of claim 7. 1
2. A method for inhibiting the release mediated by sPLA2 of fatty acid comprising contacting sPLA2 with a therapeutically effective amount of a solution prepared from the composition of claim 1 or 4. A method for treating a mammal, including a human, to alleviate the pathological effects of inflammatory diseases; wherein the method comprises parenterally administering to said mammal a therapeutically effective amount of a solution prepared from the formulation of claim 1 or 4. 14. A method for the treatment or prevention of sepsis which comprises administering a therapeutically effective amount of an aqueous solution. which comprises: (a) [[3- (2-amino-1,2-dioxoethyl) -2-ethyl-l- (phenylmethyl) -lH-indol-4-yl] oxy] sodium acetate, (b) Trisodium citrate dihydrate, and (c) Mannitol. 15. A lyophilized composition of claim 1 or 4 for the manufacture of a medicament for treating sepsis in a mammal, including a human, currently afflicted with or susceptible to sepsis. 16. A dose concentrate containing a sealed container containing the lyophilized composition of claim 1 or 4, the container having a sufficient space for the introduction of a sufficient volume of aqueous solvent to form a concentrated solution of said formulation. 17. Lyophilized compositions for the treatment of inflammatory diseases as described by any of the examples herein. 18. The process of claim 17 wherein: step (a) is carried out by dissolving an aqueous solvent: [[3- (2-amino-l, 2-dioxoethyl) -2-ethyl-l- (phenylmethyl)] -lH-indol-4-yl] oxy] sodium acetate, - a solubilizer selected from citric acid, edetic acid, polyphosphoric acid and its salts, the amount of which is from 1 to 100 percent (w / w) of the Equivalent acid amount of the Active Ingredient; and a stabilizer selected from mannitol, xylitol, sorbitol, glucose, fructose, lactose and maltose, the amount of which is 50 to 200 percent (w / w) of the amount of eguivalent acid of the Active Ingredient. The process of claim 17 wherein: steps (b), (c), (d) and (e) each are carried out for a period of at least half an hour and step (e) is performed at a subatmospheric pressure of less than 133 Pa (100 milliTorr). 20. A lyophilized composition prepared by the process of claim 17. 21. A method for inhibiting the release mediated by sPLA2 of fatty acid comprising contacting sPLA2 with a therapeutically effective amount of a solution prepared from the composition of claim 1. or 12. 22. A method for treating a mammal, including a human, to alleviate the pathological effects of inflammatory diseases; wherein the method comprises parenterally administering to said mammal a therapeutically effective amount of a solution prepared from the formulation of claim 1 or 12. 2
3. A method for the treatment or prevention of sepsis comprising administering a therapeutically effective amount of a solution aqueous comprising: (a) [[3 - (2-amino-1,2-dioxoethyl) -2-ethyl-l- (phenylmethyl) -lH-indol-4-yl] oxy] sodium acetate, (b) Trisodium citrate dihydrate, and (c) Mannitol. 2
4. A lyophilized composition of claim 1 or 12 for the manufacture of a medicament for treating sepsis in a mammal, including a human, currently afflicted with or susceptible to sepsis. 2
5. A dose concentrate containing a sealed container containing lyophilized composition of claim 1 or 12, said container having a sufficient space for the introduction of a sufficient volume of aqueous solvent to form a concentrated solution of said formulation. 2
6. An intravenous bag or a unit-dose intravenous bottle convenient for containing a liquid for intravenous injection for the treatment of inflammatory diseases wherein the improvement comprises: filling the bag with a prepared aqueous solution of the composition of claim 1 or 12. 2
7. Lyophilized compositions for the treatment of inflammatory diseases as described by any of the examples herein.
MXPA/A/2000/008596A 1998-03-03 2000-09-01 Pharmaceutical compositions containing the phospholipase inhibitor sodium [[3-(2-amino-1, 2-dioxoethyl)- 2-ethyl-1-phenylmethyl) -1h-indol-4-yl]oxy]acetate MXPA00008596A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US60/076,659 1998-03-03

Publications (1)

Publication Number Publication Date
MXPA00008596A true MXPA00008596A (en) 2002-03-05

Family

ID=

Similar Documents

Publication Publication Date Title
JP2011021042A (en) Pharmaceutical composition containing phospholipase inhibitor sodium [[3-(2-amino-1,2-dioxoethyl)-2-ethyl-1-(phenylmethyl)-1h-indol-4-yl]oxy]acetate
FI70794B (en) PROCEDURE FOR THE STABILIZATION OF THE PHARMACEUTICALS
JP5406197B2 (en) Co-solvent compositions and methods for improving delivery of dantrolene therapeutics
US5036060A (en) Cyclophosphamide
US10869848B2 (en) Carmustine pharmaceutical composition
CH695185A5 (en) injectable pharmaceutical compositions containing piperacillin and tazobactam and process for their production.
AU738413B2 (en) Human growth hormone-containing aqueous pharmaceutical composition
CZ292423B6 (en) Crystalline anhydrous mofetil mycophenolate and intravenous formulation thereof
US20080103121A1 (en) Cephalosporin derivative formulation
KR900003491B1 (en) Lyophilized composition containing isofamide and process for its preparation
US5972912A (en) Method for lyophilizing ifosfamide
IE60453B1 (en) Parenteral suspensions of diclofenac
KR100725076B1 (en) SOLUTION OF N-[o-p-PIVALOYLOXYBENZENESULFONYLAMINOBENZOYL]GLYCINE MONOSODIUM SALT TETRA-HYDRATE AND DRUG PRODUCT THEREOF
MXPA00008596A (en) Pharmaceutical compositions containing the phospholipase inhibitor sodium [[3-(2-amino-1, 2-dioxoethyl)- 2-ethyl-1-phenylmethyl) -1h-indol-4-yl]oxy]acetate
EP0682522B1 (en) Process to prepare pharmaceutical compositions containing vecuronium bromide
CN117651548A (en) Freeze-dried preparation solution, freeze-dried preparation, method and application
CZ20003208A3 (en) Pharmaceutical preparations containing sodium [[3-(2-amino-1,2-dioxoethyl)-2-ethyl-1-(phenylmethyl)-1H-indol-4-yl]oxy]acetate inhibitor of phospholipase
JP3956654B2 (en) N- [o- (p-pivaloyloxybenzenesulfonylamino) benzoyl] glycine monosodium salt tetrahydrate-containing solution and preparation
US9765108B2 (en) Formulation of 5-azacytidine
EP0139024B1 (en) Injectable compositions of bbm-928a
CN112752569A (en) Freeze-dried substance of trooshusan
CN114306578A (en) A pharmaceutical composition containing human blood coagulation factor IX and its preparation method