MXPA98009960A - Process for preparing 2,2 '- (1-methyl-1,2-etanodyiliden) bis [hidrazincarboximidami - Google Patents

Process for preparing 2,2 '- (1-methyl-1,2-etanodyiliden) bis [hidrazincarboximidami

Info

Publication number
MXPA98009960A
MXPA98009960A MXPA/A/1998/009960A MX9809960A MXPA98009960A MX PA98009960 A MXPA98009960 A MX PA98009960A MX 9809960 A MX9809960 A MX 9809960A MX PA98009960 A MXPA98009960 A MX PA98009960A
Authority
MX
Mexico
Prior art keywords
methyl
bis
parts
approximately
solution
Prior art date
Application number
MXPA/A/1998/009960A
Other languages
Spanish (es)
Inventor
Philion Richard
A Elenbaas Steven
Original Assignee
Sanofi
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 Sanofi filed Critical Sanofi
Publication of MXPA98009960A publication Critical patent/MXPA98009960A/en

Links

Abstract

A process for the preparation of 2,2 '- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide] is described by: a) reacting aminoguanidine hydrochloride with methylglyoxaldehyde or methylglyoxal dimethylacetaldehyde; , 2 '- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide] by crystallization from an aqueous isopropyl alcohol medium

Description

PROCESS FOR PREPARING 2, 2 '- (1-METHYL-2-ETANODYYLIDEN) BIS [HYDRAZINCARBOXIMIDAMIDE] DESCRIPTION OF THE INVENTION The present invention relates to a process for preparing 2,2' - (1-methyl-1, 2- ethanediylidene) bis [hydrazine carboximidamide] which has greatly reduced impurities therein useful in a method for treating cancer or advanced malignancies. The compound 2, 2 '- (1-methyl-1, 2-ethanediylidene) bis [hydrazine carboximidamide] is known by various names, such as 1,1' [(methyletandiylidene) dinitrile] diguamidine, pyruvaldehyde bis (amidinohydrazone), mitoguazone and methylglycoal bis-guanylhydrazone, methyl GAG or MGBG, represented by the formula MGBG and its salts have been described in the prior art since 1950 for use against various diseases as illustrated by the following patents and publications. The antitumor activity of MGBB in leukemia L-1210 and adenocarcinoma 755 carried by rodents is reported by Freelander et al., 18 Cancer Res. 360 (1958). Japanese Patent 51044643 discloses MGBG and its acid addition salts as effective agents against viral diseases to prevent and treat infections, pancreatic necrosis and hematopoietic necrosis. Japanese Patent 50029520 describes MGBG and its salts for use against influenza viruses. U.S. Patent No. 4,201,788 describes MGBG for the treatment of non-malignant proliferative skin diseases. MGBG is known to inhibit S-adenosylmethionine decarboxylase (SMD), which is a key enzyme in the synthesis of polyamide, leading to the reduction of cellular polyamine. However, research with MGBG reveals unacceptable levels of toxicity. The toxicological effects of MGBG, some of which are peculiar to certain animal species, include gastrointestinal toxicity, delayed and fatal hypoglycemia, liver and kidney damage, bone marrow depression, diarrhea and phlebitis. These also have prevailed in human subjects suffering from treatment with MGBG. Additionally, several toxic effects are demonstrated which are unique to men. These include esophagitis, ulcerative pharyngitis, laryngitis, stomatitis, swelling of the genital mucosa, conjunctivitis, mucositis, erythema, edema, desquamative dermatitis, and deep anorexia as associated weight loss. Patients who are administered with MGBG in a daily program exhibit remission to acute leukemia only after a precarious struggle with some frequency of life of side effect treatments. In many patients, treatment has been discontinued before any beneficial results can be noticed. Knight et al., In Can. Treat. Rep. , £ 3 1933-1937 (1979) finds that the toxicity levels are dose-related programs and can be controlled. U.S. Patent 4,520,031 addresses the issue of such control related to the dose schedule in order to reduce toxicity. The control of the dose program, as described in the patent, is based on the postulation that MGBG exerts an inhibitory action in relation to the biosynthesis of polyamine. The physiologically achieved effects of MGBG can be related to the inhibition of the enzyme S-adenosyl methionine decarboxylase, which catalyzes the synthesis of polyamine, spermidine. It is believed that spermidine plays an important role in the initiation of DNA synthesis. Studies have shown that MGBG-mediated depression of DNA synthesis is associated with decreased spermidine and putrescine accumulation. Another area in which polyamines are thought to play a major role is in the synthesis of RNA, especially that of transfer RNA (t). TRNA methylation can be stimulated directly by polyamines, a finding of particular interest in the light of the reports is that neoplastic tissue differs from normal tissue with respect to the degree of methylated tRNA. Here, too, spermidine seems to play a critical role. The accumulation of polyamine seems to be a necessary requirement for DNA synthesis at an optimal speed, in both normal and neoplastic tissues. In this way, the toxicity of MGBG observed in rapidly changing tissues (skin, mucosa and bone marrow) may be related to the inhibition of polyamine synthesis and subsequent reduction of RNA and DNA, the agents which ultimately regulate cell replication. There is, however, strong evidence that: (1) polyamines are excreted in excess in most cancer patients; (2) polyamines, especially spermidine, are released from tumor cells during and after effective chemotherapy, with an initial increase in excretion and in serum levels and subsequent normal values decreasing; and (3) chemotherapy which produces only bone marrow toxicity (or other normal tissue), and has no antitumor efficacy, does not produce a significant increase in polyamine excretion. The latter observation may suggest either that cells with cancer have much higher levels of polyamines than normal cells, even those with higher proportions of DNA synthesis, or that which therapy is effective produces different specific effects in polyamine synthesis in cells with cancer. In this way, the decrease in spermidine is associated with the action of MGBG. In studies conducted in men, the toxicological effects observed clinically for the cumulative effects of repeated daily doses have been attributed. This accumulation or addition of toxicity is possibly explained by the unusual prolonged period required for urinary elimination of MGBG in man. Studies of bioavailability in man with MGBG-C14 have shown that following a simple intravenous infusion in a period of 20 minutes, the radioactivity disappears quickly from the plasma and that over a prolonged period of 3 weeks, 60 percent of the drug is excreted without changing in the urine. These data suggest that MGBG accumulates in the tissues and slowly filters out of tissue deposits to carry out the removal. In the patent, after a considerably large number of studies conducted with MGBG in the treatment of several tumors, it is concluded that a weekly administration schedule is more effective to achieve the higher therapeutic index while reducing the toxicity to an acceptable level. Accordingly, a dose range of 250 mg / m2 to 1000 mg / m2 of MGBG administered at intervals of weeks is established for the treatment of several tumors. While the dose range indicated above decreases toxicological side effects and produces treatments for several tumors, the long-term accumulation of MGBG is still a problem that requires additional studies and / or treatment modifications. In the present, there have been prolonged studies of MGBG in order to further reduce the toxic side effects thereof. In the course of these studies, it has been discovered that MGBG contains relatively large amounts of impurities, which may contribute to the toxicological side effects of MGBG. Accordingly, various efforts have been made to identify and reduce the amount of impurities present in MGBG and its salts. The process for preparing guanylhydrazones is described by Baiocchi et al., J. Med. Chem., 431 (1963) and Oliver, Denham., J. Pharm. Sci., 2 202 (1963). reacting the aminoguanidine salt with the corresponding carbonyl compound in an aqueous or aqueous-alcohol medium in the presence of a catalytic amount of acid The process uses commercially available aminoguanidine bicarbonate and is described as follows General Preparation of Guanilhydrazones To a solution of 0.11 moles (15 g) of aminoguanidine bicarbonate in 125 ml of water is slowly added the desired acid (add a few drops of amyl alcohol in order to avoid foam) until the pH of the solution is less than 7. It is filtered The solution of trace amounts of insoluble solids is then added to the appropriate carbonyl compound to a slightly acidic filtrate at a filtrate at 60 ° C. The amount of the carbonyl compound used is such that the proportion of aminoguanidine by carbonyl function is d 1: 1. If the carbonyl compound does not dissolve in the aqueous mixture, ethanol is added until the reaction mixture is homogeneous. The solution is then stirred at room temperature for 16 hours. If a precipitate forms, the solid product is isolated by filtration. Similarly, the reaction mixture is evaporated until a solid residue is obtained using methanol, ethanol and solvent combination. The recovery of guanilhidrazonas is low. In addition to the low yield it is found here that the products contain large amounts of impurities. Experimental Methods for the analysis of mitoguazone dichlorohydrate using various systems to identify and quantify their impurities have been developed in the present. U analysis system includes High Resolution Liquid chromatography. Considered in terms of determining chromatographic purity levels, a high degree of specificity provides confidence that all potential species of interest are detectable. Where these species, including impurities of degradation processes and products, are not available for injecting directly into the chromatographic system, the specificity test is usually performed on stressed samples using one or more techniques which can be placed in one of two categories. spacious. (a) Use of a specialized detection system to extract additional information from the peak of the analyte. (b) Some form of comparison between complementary separation techniques, the first being the low validation system and the second being a system which, by virtue of its different selectivity, can be expected to resolve co-elution of species in the first example. Examples of techniques in the first category include the use of diode array and mass spectroscopy detectors to obtain UV / Visible or mass spectra respectively from several positions across the analyte peak potentially allowing the detection of species co-eluents The techniques in the second category include the use of flux change apparatuses to invert the analyte peak in a second stationary phase with a different selectivity, or comparison of the results of the system that is validated with those of a second chromatography technique such as a thin layer chromatography (TLC).
Equipment and Chemicals HPCL data are generated using several Kontron (Watford, Herts) and Waters (atford, Herts) pumps, an autosampler, a column kiln and detector models. The HPLC data are processed using Multichrom ™ VI-82 (LabSystems, Altringham, Cheshire). The UV / visible absorption spectra are captured, using an HPO 1040 diode array detector (Hewlett Packard, Bracknell, Berks). Light tension (Xenon Source, filtered through window glass) is performed on a Haraeus Suntest ™ (Alplas Technology, Oxford). The HPLC grade acetonitrile of chemicals is obtained Rathburn (Walkerburn, Scotland), grade heptanesulfonic acid HPLC (sodium salt), and inorganic chemicals from BHD limited (Poole, Dorset) ACVA is obtained (4,4'-azobis (4-cyanovaleric acid), a radical initiator, exposure to which the oxidative contraction of Ajdrich (Gillingham, Dorset) is mimicked.) Mitoguazone dihydrochloride is obtained and purified water at home Stressed Sample Preparation Specimens of mitoguazone dichlorohydrate (approximately 370 mg, equivalent to one bq of 250 mg) are weighed out in 50 ml volumetric flasks and stressed, in accordance with the conditions given above. the tension continues for a maximum of 7 days or until the degradation has been achieved from 20 to 50% After the tension, the samples are neutralized, if necessary, and diluted to a volume with purified water to give solutions of approximately 5 m (base) / ml by TLC analysis The aliquots of these solutions are diluted with purified water to give solutions of about 1 mg / (base) / ml for use in HPLC impurity determinations. dilute aliquots of these solutions, either in the HPLC mobile phase to give a solution of approximately 0.01 mg (base) / ml for HPLC assay. Heat Tension Conditions: The sample is maintained at 80 ° C for 7 days.
Acidity: 10 ml of 0.1 M hydrochloric acid is added to the sample and the solution is kept at 70 ° C for 7 days. Basic: 10 ml of 0.1 M sodium hydroxide is added to the sample and the solution is maintained at 70 ° C for 2 days. Aqueous: 10 ml of purified water is added to the sample and the solution is maintained at 70 ° C for 7 days. Oxidative: 10 ml of a 0.1 M aqueous ACVA solution is added and the sample is kept at 40 ° C for 7 days. Light: The sample receives a total illumination of approximately 15,000 klx hours (with associated UV). Assay: The sample is chromatographed isocratically in columns of 25 cm x 0.46 d.i. Hypersil BDS C8 5 μm (Anachem, Luton, Beds.) Using a mobile phase consisting of 0.05 M potassium diacid orthophosphate buffer containing 1 g / 1 heptanesulfonic acid (sodium salt) and adjusted to pH 3.0 with acid concentrated orthophosphoric (89% by volume) and acetonitrile (11% by volume). The flow rate is 2 ml / minute, the wavelength of the detector is 283 nm, the injection volume is 20 μl and the temperature of the column is 40 ° C. The samples are quantified with respect to an accurately prepared external standard (nominally 0.01 mg (base) / ml) Impurity Method: The chromatographic conditions are com for the assay, except that a detector wavelength of 210 nm is used. uses a second HPLC system with a ratio of aqueous mobile phase to acetonitrile from 85% to 15% by volume, mainly to estimate the specific process impurities.The impurities are quantified with respect to a closely prepared external standard (nominally 0.005 mg (base ) / ml) TLC method 20 μl of each sample is placed on a silica gel TLC plate (Merck 60F254) The plate is developed at a height of 10 cm in acetone / ammonium hydroxide (SG 0.88) / water (90: 55.5% by volume) mobile phase The impurities are estimated against diluted mitoguazone dichlorohydrate stains, both with short wavelength ultraviolet light (254 nm), and after treatment with a reagent by aspersion Nitroprusside (sodium) -ferricyanide. Results Triplicate samples representing 0, 80%, 100% and 120% of the nominal mitoguazone dichlorohydrate concentration are analyzed. The results obtained in Table I are given. TABLE 1 Data Recovery TABLE I (continued) Data Recovery HPLC assay *. The Least Squares Regression analysis of the data gives an average accuracy of 99.8% (Coefficient d Correlation 0.99935). Analysis of Stressed Samples Stressed samples are assayed by HPLC while the chromatographic impurity levels are determined by TLC HPLC. The chromatographic data is summarized in Table II. TABLE II Chromatographic Assays and Impurity Data for Stressed Mitoguazone Dichlorohydrate Using HPLC, TLC and mass spectroscopy, they identify and quantify the impurities contained in the starting materials or formed during the process to manufacture the final product. It is found herein that purity related to diaminoguanidine is counted by more than 70 impurity level of MGBG-total dihydrochloride.
Then the general reaction scheme for the production of impurities and their chemical names. 1, 3 -Diaminoquanidine Impurity O: R1 = H, R2 = CH3 Impurity E ^ = CH3, R2 = H D v E Impurity F: Rx = H, R2 = CH3, R3 = CH3, R4 = H Impurity G: RX = CH3, R2 = H, R3 = H, R4 = CH3 Impurity H ^ = H, R2 = CH3, R3 = H, R4 = CH3 wherein D [2- [(aminoiminomethyl) hydrazonojpropiliden-carbonimidicdihydrazid. E = [2- [(Aminoiminomethyl) hydrazono-1-methylethylidene] -carbonimidic dihydrazide. F = Bis [2- [(Aminoiminomethyl) hydrazono-1-methylethylidene-carbonimidic dihydrazide. G = Bis [2- [(Aminoiminomethyl) hydrazono] propylidene] -carbonimidic dihydrazide.
H = [2- [(Aminoiminomethyl) hydrazono] -l methylethylidene] [2- [(aminoiminomethyl) hydrazono] propylidene] ~ carbonimidic dihydrazide. In the co-pending application D.N. 70493, Seri No. 08/655, 512 on the date with the present application, the reaction parameters are studied and modified in order to reduce the impurities in the final product. The copending application, which is incorporated by reference in its entirety, describes a process for the preparation of 2,2 '- (1-methyl-1, 2-ethandylidene) bis [hydrazine carbsximidamine] which comprises the steps of: a) removing impurities of bicarbonate d aminoguanidine by suspending bicarbonate d aminoguanidine in water and filtering the suspension; b) reacting the filtered aminoguanidine bicarbonate with methylglyoxal dimethylacetal in an aqueous reaction medium to yield 2,2 '(methyl-1,2-ethandylidene) bis [hydrazine carboximidamide]; and c) purifying 2, 2 '- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide by recrystallization from a d-isopropanol-aqueous acidic medium. An essential step in the process is the removal of the impurities from the bicarbonate d aminoguanidine starting material by suspending the aminoguanidine bicarbonate in water or by filtering out the impurities from the filtration. It has now been discovered that instead of using aminoguanidine bicarbonate as a starting material, the aminoguanidine hydrochloride can be used to react with methylglyoxal dimethylacetal in an aqueous reaction medium to produce 2, 2 '- (1- methyl-l, 2-ethandylidene) bis [hydrazine, caroximidamide]. The process including a subsequent purification step provides a final product which is at least 95.5% pure. The process for obtaining highly purified 2, 2 '- (1-methyl-1,2-ethandylidene) bis [hydrazine carboximidamide] involves reacting aminoguanidine chlorohydrate with methylglyoxal dimethylacetal comprising the steps of: a) dissolving a part of aminoguanidine hydrochloride in a mixture of about 0.5 to 3 parts, preferably about 0.93 parts of water, and about 0 to 3 parts of a miscible organic solvent, preferably about 0.75 of isopropyl alcohol. b) adjusting the pH of the solution to about 0 5, preferably 0 to 1 with concentrated hydrochloric acid. c) adding to the solution about 0.25 part of methylglyoxaldehyde, preferably about 0.5 part of methylglyoxa dimethylacetal, at a temperature of about 0 to 50 ° C, preferably of 25-30 ° C; d) stirring the reaction mixture for about 48 hours, preferably about 1 to 1 hours at room temperature. e) adding approximately 0.5 to 20 parts of a water miscible organic solvent, preferably about 5 parts of isopropyl alcohol to the reaction mixture to produce 2,2 '- (1-methyl-1,2-ethandylidene) bis [hydrazine] carboximidamide]; f) collecting 2, 2 '- (1-methyl-1, 2-ethandylidene) bis [hydrazine carboximidamide] without purifying solid and washing the same with 1 to 10 parts of an organic solvent, preferably with 1 to 2 parts of alcohol isopropyl; and g) optionally drying the final product without purification before purification in a vacuum oven at 45 ° C. The purification process of 2, 2 '- (1-methyl-1, 2-ethandylidene) bis [hydrazine carboximidamide] without purification comprises the steps of: h) dissolving the unpurified compound from about 0.5 to 4 parts of deionized water, preferably about 3 parts of deionized water; 5) add approximately 0.5 to 2 parts of a water miscible organic solvent, preferably about 1 part isopropanol; j) adjusting the pH of the solution to about 0 to 5, and preferably 0 to 1 with concentrated hydrochloric acid; k) stir the solution for approximately 0.5 to 2 hours while adding 0.1 to 2 parts of deionized water and maintaining the temperature of the solution at approximately 28-32 ° C. 15 1) filter the solution to remove insoluble impurities; m) adding to the filtered solution about 0.5 to 10 parts of a water miscible organic solvent, preferably about 5 parts of isopropyl alcohol to precipitate the compound; n) cooling the mixture to about 0 to 25 ° C, preferably about 10 ° C; o) collecting the final product by filtration and washing the filtrate with 0.5 to 10 parts of an organic solvent, preferably with a part of isopropyl alcohol; and p) drying the purified product. As used herein the process for synthesizing and purifying 2, 2 '- (1-methyl-1,2-ethandylidene) bis [carboximid hydrazine] includes synthesis and purification of its various forms including its hydrochloride, monohydrate, dihydrate and hemihydrated. In the process for synthesizing and purifying 2,2 '- (1-methyl-1, 2-ethandylidene) bis [hydrazine carboximidamide] the use of isopropyl alcohol is preferred. However, other organic solvents miscible in water may be used which include methanol, ethanol, n-propyl alcohol, tetrahydrofuran, acetic acid, dimethylformamide, acetonitrile and dimethyl sulfoxide or mixtures thereof. Aminoguanidine hydrochloride, methylglyoxaldehyde and methylglyoxal dimethylacetal are commercially available such as from Aldrich Chemical Co. , and can also be prepared by processes known in the art. A representative example (Example 1) for synthesizing and purifying 2, 2 '- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide] illustrates the invention. Example 1 (A) Synthesis 60.0 grams of aminoguanidine hydrochloride is dissolved in 56 milliliters of deionized water and 36 grams of isopropyl alcohol. The pH of the solution is adjusted to approximately 0 to 1 by adding concentrated hydrochloric acid. 30.96 grams of methylglyoxal dimethyl acetal are added in 1.5 to 3 hours while maintaining the reaction temperature between 25 and 30 ° C. The reaction mixture is then stirred at room temperature for an additional 16 hours. 240 grams of isopropyl alcohol are added and the reaction mixture is cooled to 10 ° C. The unpurified product which is formed during the reaction process is collected by filtration and washed with 90 milliliters of isopropyl alcohol. The washed filtrate is then dried in an oven at 45 ° C overnight. The yield is 87% of the theoretical yield. (B) Purification Dissolve 79.3 grams of the unpurified product obtained (A) in 159 grams of deionized water at 37 ° C. Add 60.7 grams of isopropyl alcohol and adjust the pH to 0-1 by adding concentrated hydrochloric acid. The pH-adjusted solution is stirred for 1 hour while maintaining its temperature at 28-32 ° C, and adding 10 ml of deionized water. The mixture is filtered to remove impurities, such as dirty mechanical particles. 312 grams of isopropyl alcohol are then added to the solution to precipitate the product. The mixture containing the precipitated product is cooled to 8-12 ° C and stirred for about 15 minutes. The purified product is then washed with 68 milliliters with isopropyl alcohol and dried in a vacuum oven for 45 ° C. The yield of the purified product was 67.5 of the theoretical yield. Samples of 2, 2 '- (1-methyl-1, 2-ethanediidine) bis [hydrazine carboximidamide] produced ,: 1 reacting aminoguanidine bicarbonate with methylglyoxamethyl acetal or 2) Purify by HPLC to react aminoguanidine hydrochloride with dimethylglyoxamine dimethyl acetal . The comparative results are shown in l Taba II and Table III. TABLE II Test Results for Batch of Bicarbonate Aminoguanidine used in the Production of MGBG and Test Results of MGBG Produced by the same.
TABLE III Test Results for Lots of Aminoguanidine Hydrochloride used in the Production of MGBG and MGBG Test Results Produced by the same.
Aminoguanidine hydrochloride contains lower levels of impurities than aminoguanidine bicarbonate gives a superior quality of MGBG. The purity of MGBG produced by the use of aminoguanidine hydrochloride and according to the present invention is found to be as high as 99.9%. Highly pure MGBG is well suited for pharmaceutical compositions for the treatment of cancer and other diseases. The invention has been described in detail with particular reference to certain preferred embodiments thereof, but it will be understood that variations and modifications may be made within the spirit and scope of the invention.

Claims (6)

1. A process for preparing 2, 2 '- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide] characterized in that it comprises the steps of: a) dissolving a part of the chlorohydrate d aminoguanidine in a mixture of approximately 0.5 to 3 parts, of water and about 0 to parts of an organic solvent miscible with water; b) adjusting the pH of the solution to about 0 5; with concentrated hydrochloric acid; c) adding to the solution about 0.25 to parts of methylglyoxaldehyde or methylglyoxa dimethylacetal, at a temperature of about 0 to 50 ° C to obtain a reaction mixture; d) stirring the reaction mixture for approximately 48 hours, at room temperature. e) adding about 0.5 to 20 parts of a water-miscible organic solvent to the reaction mixture, to produce 2,2 '- (1-methyl-1,2-ethandylidene) bis [hydrazine carboximidamide]; f) collecting 2, 2 '- (1-methyl-1, 2-ethandylidene) bis [hydrazine carboximidamide] if purifying by filtration and washing the same with 1 10 parts of an organic solvent; g) dissolving 2, 2 '- (1-methyl-1,2-ethanediylidene) is [hydrazine carboximidamide] if purifying in about 0.5 to 4 parts of water; h) adding approximately 0.5 to the solution to parts of a water-miscible organic solvent; and) adjusting the pH of the solution to about 0 5; 10 j) stir the solution for approximately 0.5 to hours while adding approximately 0 to parts of water and maintain the temperature of the solution at approximately 28 to 32 ° C; k) add approximately 0.5 to 1 to the solution 15 parts of an organic solvent miscible in agu to precipitate 2, 2 '- (1-methyl-1, 2-ethanediylidene) bis [hydrazine carboximidamide], - 1) cool the mixture to about 0 to 25 ° C; m) collecting the 2, 2 '- (1-methyl-1, 2- 20 ethandylidene) bis [hydrazine carboximidamide] solid by filtration and washing with 0.5 to 10 parts of an organic solvent; and n) drying purified 2,2 '- (1-methyl-1, 2-ethanediylidene) bis [hydrazine carboxamide].
2. 2,2 '- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide] prepared by the process according to claim 1.
3. A method for treating cancer or malignant disease in a mammal characterized because it comprises administering to the mammal an effective amount of a pharmaceutical composition containing the compound d according to claim 2.
4. A process for preparing 2,2 '- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide] characterized because it comprises the steps of: a) dissolving a part of the chlorohydrate d aminoguanidine in a mixture of approximately 0.93 parts, water and approximately 0.75 part isopropyl alcohol; b) adjust the pH of the solution to approximately 0 1, with concentrated hydrochloric acid; c) adding approximately 0.5 d methylglyoxal dimethylacetal to the solution at a temperature of about 25 to 30 ° C; d) shaking the solution for about 1 to 1 hours at room temperature; e) adding approximately 5 parts of isopropyl alcohol to the reaction mixture to produce 2,2'- (1-methyl-1,2-ethanediylidene) bis [hydrazine carboximidamide]; f) collecting 2, 2 '- (1-methyl-1, 2-ethanediylidene) bis [hydrazine carboximidamide] without purification by filtration and washing the same with 1 to 2 parts of isopropyl alcohol; 5 g) dissolving 2, 2 '- (1-methyl-1, 2-ethanediylidene) bis [hydrazine carboximidamide] without purification in about 2 parts of water; h) add approximately one part of isopropyl alcohol; 10 i) adjust the pH of the solution to 0 to 1 with concentrated hydrochloric acid; ) stirring the solution for approximately 0.5 to 2 hours while adding approximately 0 to 2 parts of water and maintaining the temperature of the solution at approximately 28-32 ° C; k) filter the solution to eliminate the soluble impurities thereof; 1) add approximately 5 parts of isopropyl alcohol to the filtered solution to precipitate 2,2 '- (1-methyl-1,2-ethoylidene) bis [hydrazine carboximidamide]; m) cooling the mixture to about 10 ° C; n) collecting the 2, 2 '- (1-methyl-1, 2-ethandylidene) bis [hydrazine carboximidamide] solid 25 by filtration and washing it with a portion of isopropyl alcohol; and o) drying 2, 2 '- (1-methyl-1, 2-ethanediylidene) bis [hydrazine carboxamidamide].
5. The 2, 2 '- (1-methyl-1, 2-ethanediylidene) bis [hydrazine carboximidamide] prepared by the process of compliance with claim 4.
6. A method for treating cancer or malignant diseases in a mammal, characterized because it comprises: administering to the mammal an effective amount of a pharmaceutical composition containing the compound according to claim 5.
MXPA/A/1998/009960A 1996-05-30 1998-11-27 Process for preparing 2,2 '- (1-methyl-1,2-etanodyiliden) bis [hidrazincarboximidami MXPA98009960A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US08655518 1996-05-30

Publications (1)

Publication Number Publication Date
MXPA98009960A true MXPA98009960A (en) 1999-04-27

Family

ID=

Similar Documents

Publication Publication Date Title
CN101282946B (en) Cysteine protease inhibitors and their therapeutic applications
EP3505526B1 (en) Myricetin derivative and preparing method thereof, and application of same for treating colitis, preventing and treating colitis tumorigenesis, and treating colorectal cancer
JP2002512233A (en) Use of dihydropyrimidines as drugs and novel substances
ES2517871T3 (en) Trazodone and trazodone hydrochloride in purified form
JP2983669B2 (en) 2,4-pyrimidinedione derivatives and pharmaceutical compositions
EP0031128A1 (en) (E)-5-(2-halogenovinyl)-arabinofuranosyluracil, process for preparation thereof, and uses thereof
Baiocchi et al. Studies on methylglyoxal bis (guanylhydrazone) 1 analogs. II. Structural variations on methylglyoxal bis (guanylhydrazone) 2
SK287814B6 (en) Azithromycin in the form of a monohydrate, a process for its preparation and pharmaceutical compositions comprising same
CN108299255B (en) Histone deacetylase 8 selective inhibitor and preparation method and application thereof
EP2102172B1 (en) Synthesis of 4-amino-pyrimidines
JPH08277262A (en) Mercaptoethanesulfonic acid derivative and its production
US5693673A (en) Process of preparing 2,2'-(1-methyl-1,2-ethanediylidene)bis hydrazine caroximidamide!
MXPA98009960A (en) Process for preparing 2,2 '- (1-methyl-1,2-etanodyiliden) bis [hidrazincarboximidami
US5659083A (en) Process of preparing 2,2'-(1-methyl-1,2-ethanediylidene) bis[hydrazinecarboximidamide]
JP2519205B2 (en) All-cis-1,3,5-triamino-2,4,6-cyclohexanetriol derivative
JP2598609B2 (en) 6,6-disubstituted pteridine derivatives
JPS6344577A (en) Pyrimidine derivative
EP3763728A1 (en) Glycopeptide compounds having activity of resisting drug-resistant bacteria, and preparation method therefor and application thereof
Jensen et al. Use of acetylacetone to prepare a prodrug of cycloserine
MXPA98009959A (en) Process for preparing 2,2 '- (1-methyl-1,2-etanodyiliden) bis [hidrazincarboximidami
CN103601673B (en) The fluorinated derivatives of 3-pyridone-4-ketone
HUT71240A (en) Process for the preparation of 9-amino camptothecin
JP2561531B2 (en) Purification method of 7-dimethylamino-6-demethyl-6-deoxytetracycline
RU2708590C1 (en) Method of purifying glycoluril from hydantoin impurity
EP0307788A1 (en) Taurine derivative, its preparation and pharmaceutical compositions containing it