CN115286632A - Preparation process of valganciclovir hydrochloride - Google Patents

Preparation process of valganciclovir hydrochloride Download PDF

Info

Publication number
CN115286632A
CN115286632A CN202211128373.8A CN202211128373A CN115286632A CN 115286632 A CN115286632 A CN 115286632A CN 202211128373 A CN202211128373 A CN 202211128373A CN 115286632 A CN115286632 A CN 115286632A
Authority
CN
China
Prior art keywords
stirring
valganciclovir
cbz
filtering
temperature
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202211128373.8A
Other languages
Chinese (zh)
Other versions
CN115286632B (en
Inventor
张恺
薛娜
张玉辉
华园
陈兴贺
李林
闫弘毅
孙飞
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hebei Renhe Yikang Pharmaceutical Co ltd
Hebei Medical University
Hebei Chemical and Pharmaceutical College
Original Assignee
Hebei Renhe Yikang Pharmaceutical Co ltd
Hebei Medical University
Hebei Chemical and Pharmaceutical College
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 Hebei Renhe Yikang Pharmaceutical Co ltd, Hebei Medical University, Hebei Chemical and Pharmaceutical College filed Critical Hebei Renhe Yikang Pharmaceutical Co ltd
Priority to CN202211128373.8A priority Critical patent/CN115286632B/en
Publication of CN115286632A publication Critical patent/CN115286632A/en
Application granted granted Critical
Publication of CN115286632B publication Critical patent/CN115286632B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

The invention relates to a preparation process of valganciclovir hydrochloride, which takes ganciclovir as a raw material, reacts with trimethyl orthoacetate in the presence of p-toluenesulfonic acid to obtain monoacetylganciclovir, then reacts with Cbz-L-valine in DMSO, is treated by hydrochloric acid, is adjusted by ammonia water to pH 1.5 to obtain Cbz-L-valganciclovir, and is further prepared by hydrogenation and post-treatment.

Description

Preparation process of valganciclovir hydrochloride
Technical Field
The invention belongs to the technical field of medicine synthesis, and particularly relates to a preparation process of valganciclovir hydrochloride.
Background
Cytomegalovirus (CMV) is a virus of the herpesvirus family, also known as the cellular inclusion body virus, infection of which causes swelling of the cells and the production of large intranuclear inclusions. CMV is a very widespread infection in humans, usually a recessive infection, is present in almost all organs and tissues of the human body, and after infection in healthy humans, it is mostly asymptomatic, but under certain conditions it can invade multiple organs and systems, causing severe infections mainly of diseases of the genitourinary system, the central nervous system and the liver. CMV infection is a common cause of disease in pathologically and physiologically immunocompromised persons (including fetuses and newborns with developmental immunodeficiency) and is one of the most important causes of advanced disease and increased mortality in patients with aids and organ and bone marrow transplants.
Ganciclovir (ganciclovir) is one of the most effective drugs for resisting CMV infection clinically at present, but has poor oral bioavailability and great side effect. Valganciclovir hydrochloride is an oral cell-resistant (CMV) infection drug developed by roche switzerland and approved by FDA in us5 months in 2001 for marketing. Clinically used for treating acute retinitis of patients with acquired immunodeficiency syndrome caused by CMV infection; preventing and treating CMV infection in organ transplant patients. The product is a prodrug of ganciclovir, is rapidly hydrolyzed into ganciclovir by esterase in intestinal tract and liver cells after being taken orally, thereby exerting the drug effect, has the oral absorption bioavailability 10 times that of ganciclovir, greatly reduces the toxicity, and has wide market prospect. In China, valganciclovir hydrochloride belongs to a clinically urgent medicine, but no domestic variety is on the market, the valganciclovir hydrochloride completely depends on import, the price is high, and the economic burden of patients is heavy.
Valganciclovir hydrochloride has a complex structure, needs to overcome multiple technical barriers in the research and development process, and has high synthesis difficulty, and the synthesis of valganciclovir hydrochloride in the prior art is mainly as follows:
patent WO9727194 discloses a preparation method of valganciclovir hydrochloride, which comprises the steps of taking ganciclovir as a raw material, firstly reacting with trimethylchlorosilane to generate an N-silane compound, protecting 2-amino of the ganciclovir, then reacting with Cbz-valine, and obtaining the valganciclovir hydrochloride through Cbz protection removal. The synthetic route is as follows:
Figure BDA0003849078590000021
the patent US5700936 discloses a preparation method of valganciclovir hydrochloride, which also takes ganciclovir as an initial raw material to react with active L-valine derivatives under the catalysis of triethylamine to generate a diester product, then the diester product is hydrolyzed under an alkaline condition to hydrolyze L-valine groups on one of hydroxyl groups, and finally the valganciclovir hydrochloride is obtained through Cbz removal protection. The difficulty is that the mixture of ganciclovir, monoester and diester can be obtained in the second step hydrolysis reaction process, the separation is difficult, the yield is low, and the synthetic route is as follows:
Figure BDA0003849078590000031
patent US5756736 discloses that ganciclovir is used as a starting material, and reacts with triphenylchloromethane under the catalysis of 4-dimethylaminopyridine and triethylamine to generate an N, O-bis (triphenylmethyl) ganciclovir derivative, one alcoholic hydroxyl group and 2-amino group of ganciclovir are protected, then esterification is carried out with Cbz-L-valine, and then the protecting group trityl is removed under the action of trifluoroacetic acid to obtain a monoester. The main problems are that the activity of two hydroxyl groups of ganciclovir is consistent, the introduction of a single protecting group is not easy, the route is long, the yield is difficult to improve, the product quality is poor, and the synthetic route is as follows:
Figure BDA0003849078590000032
because the two hydroxyl groups of the ganciclovir have consistent activity and no selectivity, the route can not realize the high-selectivity mono-esterification of the ganciclovir, and can only separate the mixture of the ganciclovir, the monoester and the diester by different purification methods, and the final product has poor quality and lower yield.
US5840890 discloses a process which also uses ganciclovir as starting material, reacts with trimethyl orthoformate under the catalysis of trifluoroacetic acid, is acidified by formic acid to obtain substituted ganciclovir with introduced protecting group, then is esterified with active anhydride compound under the catalysis of triethylamine to obtain diester, is hydrolyzed by hydrochloric acid to remove formyl group, and finally is hydrogenated and reduced to obtain the target product. According to the method, ortho-acid ester is introduced to construct mono-substituted ganciclovir in a breakthrough manner, asymmetric diester is obtained by esterifying the mono-substituted ganciclovir, the asymmetric diester can be hydrolyzed at high selectivity under the control of hydrochloric acid concentration and reaction temperature to obtain a monoester product, the total yield can be greatly improved to more than 30%, but the problems of uncontrollable diastereoisomer ratio, N-methylated impurities, excessive palladium residue and the like in valganciclovir hydrochloride are not concerned.
Disclosure of Invention
The invention aims to overcome the defects in the prior art and provide a method for preparing valganciclovir hydrochloride with controllable purity and considerable yield.
In order to realize the purpose, the technical scheme adopted by the invention is as follows:
a preparation method of valganciclovir hydrochloride specifically comprises the following steps:
s1: stirring ganciclovir, p-toluenesulfonic acid, trimethyl orthoacetate and DMSO at normal temperature, reacting for 2-4 h, adding purified water, stirring at normal temperature for 0.25-0.5 h, adding ethanol into the reaction solution, separating out white solid, filtering, and drying to obtain monoacetylganciclovir;
s2: adding monoacetylganciclovir into DMSO, heating to 60-70 ℃, stirring for 0.5-1 h, cooling to 20-30 ℃, adding Cbz-L-valine and DMAP, stirring for 0.25-0.5 h, adding DCC, stirring for 3-6 h, adding dichloromethane and purified water after the reaction is finished, stirring for 0.5h, filtering to retain an organic phase, decompressing and evaporating to dryness, adding methanol and dilute hydrochloric acid, stirring for 8-10 h at 20-30 ℃, then adding dichloromethane for extraction, separating to retain a water phase, adjusting the pH to 1.5-3 by using ammonia water, stirring, and filtering to obtain Cbz-L-valganciclovir;
s3: dissolving Cbz-L-valganciclovir in methanol, adjusting pH to be less than 5 by hydrochloric acid, adding 10% palladium carbon, exchanging nitrogen, introducing hydrogen, reacting for 6-12 h at 25-35 ℃, filtering, adding TulsionCH-97 resin into filtrate, stirring for 2.5-3 h at 20-30 ℃, filtering, concentrating the filtrate under reduced pressure to be dry, and carrying out post-treatment to obtain valganciclovir hydrochloride.
As a further improvement of the invention, the mass ratio of ganciclovir to p-toluenesulfonic acid and trimethyl orthoacetate in step S1 is 50-10: 150-400 mL.
As a further improvement of the invention, the adding amount of the pure water in the step S1 is 0.15-0.25 time of the quality of the ganciclovir, and the adding amount of the ethanol is 1-1.5 times of the quality of the dimethyl sulfoxide.
As a further improvement of the invention, the mass ratio of the monoacetylganciclovir, the Cbz-L-valine, the DMAP and the DCC in the step S2 is 45-80.
As a further improvement of the invention, the mass ratio of the monoacetylganciclovir, the Cbz-L-valine, the DMAP and the DCC in the step S2 is 45-55.
As a further improvement of the invention, the addition amount of the dilute hydrochloric acid in the step S2 is 5-8 times of the mass of the monoacetylganciclovir, the concentration is 4mol/L, and the volume ratio of the methanol to the dilute hydrochloric acid is 1-1.5.
As a further improvement of the invention, the 10% palladium on carbon and the Cbz-L-valganciclovir in step S3: the mass ratio of (A) is 1:5-20, and the mass ratio of the TulsionCH-97 resin to the Cbz-L-valganciclovir is 0.5-5.
As a further improvement of the invention, the mass ratio of the 10% palladium carbon to the Cbz-L-valganciclovir in the step S3 is 1:8-15, and the mass ratio of the TulsionCH-97 resin to the Cbz-L-valganciclovir is 0.8-2:1.
As a further improvement of the present invention, the post-processing step in S3 is: adding purified water with the mass of 1-2 times of that of the concentrate, controlling the temperature to be 40-50 ℃, adding isopropanol with the volume of 7-9 times of that of the pure water, cooling to 20-30 ℃, keeping the temperature for reaction for 1 hour, continuously dropwise adding isopropanol with the volume of 8-16 times of that of the pure water, cooling to 0-10 ℃, stirring for 1.5-2 hours, filtering, washing a filter cake with the isopropanol, and drying to obtain the filter cake.
As a further improvement of the present invention, the post-processing step in S3 is: adding purified water with the mass of 1.5 times of that of the concentrate, controlling the temperature to be 40-50 ℃, adding isopropanol with the volume of 8 times of that of the pure water, cooling to 20-30 ℃, keeping the temperature for reaction for 1 hour, continuously dropwise adding isopropanol with the volume of 16 times of that of the pure water, cooling to 0-10 ℃, stirring for 1.5-2 hours, filtering, washing a filter cake with the isopropanol, and drying to obtain the filter cake.
Adopt the produced beneficial effect of above-mentioned technical scheme to lie in:
the method provided by the invention inhibits the generation of byproducts, realizes the removal of the byproducts and impurities, ensures the purity of products, improves the yield of the reaction and ensures that the total yield can reach more than 60 percent by controlling the conditions of each reaction step.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the embodiments or the prior art descriptions will be briefly described below, and it is obvious that the drawings in the following description are some embodiments of the present invention, and other drawings can be obtained by those skilled in the art without creative efforts.
FIG. 1 is an HPLC profile of monoacetylganciclovir of example 1 of the invention;
FIG. 2 is an HPLC chromatogram of example 1 Cbz-L-valganciclovir of the invention;
FIG. 3 is an HPLC chromatogram of valbixilol hydrochloride of example 1 of the present invention.
Detailed Description
In order to make the objects, technical solutions and advantages of the present invention more apparent, the present invention will be described in detail and fully with reference to the following embodiments.
Example 1
S1: 50g of ganciclovir, 200ml of dimethyl sulfoxide, 2.61g of p-toluenesulfonic acid and 45.08g of trimethyl orthoacetate are added into a reaction bottle, and the reaction is stirred at 25 ℃ for 4 hours. Adding 10.58g of purified water into the reaction solution, stirring for 0.5h, adding 240ml of ethanol, separating out a white solid, crystallizing for 14h at 25 ℃, performing suction filtration, washing with ethanol, and performing vacuum drying at 45-55 ℃ to obtain 49.5g of monoacetylganciclovir, wherein the yield is 85.0%, the purity is 99.7%, an HPLC (high performance liquid chromatography) spectrum is shown in figure 1, and peak table data is shown in table 1.
Table 1 monoacetylganciclovir HPLC peak table data
Figure BDA0003849078590000071
Figure BDA0003849078590000081
S2: adding 45g of monoacetylganciclovir and 225mL of DMSO into a three-neck bottle, heating to 66 ℃, stirring for 0.5h, cooling to 25 ℃, adding 47.93g of Cbz-L-valine and 3.14g of DMAP, stirring for 15 min, controlling the temperature to be below 20 ℃, adding 44.98g of DCC, controlling the temperature to be 25 ℃ for reaction for 5h, controlling the temperature to be 25 ℃, adding 225mL of DCM, 225mL of purified water, stirring for 30min, filtering, washing a filter cake with dichloromethane, standing, separating the filtrate, concentrating and evaporating an organic phase at 40 ℃ under reduced pressure.
Adding 315mL of methanol into the concentrated residue, stirring, controlling the temperature to be below 30 ℃, adding 315g of 4mol/L dilute hydrochloric acid, controlling the temperature to be 20 ℃ after the addition is finished, reacting for 9h, adding 200mL of DCM for extraction, retaining the water phase, controlling the temperature to be 10 ℃, adjusting the pH to 1.5 by using ammonia water, stirring for 30min, filtering, washing by purified water, and drying in vacuum at 45 ℃ to obtain 63.89g of Cbz-L-valganciclovir, wherein the yield is 86.4%, and the purity is 99.2%, which is shown in figure 2.
S3: adding 500ml of methanol and 50g of Cbz-L-valganciclovir into a reaction bottle, stirring, slowly adding 11g of hydrochloric acid at the temperature of 20 ℃, stirring for dissolving, adding 5g of 10% palladium-carbon, displacing with nitrogen for three times, introducing hydrogen, carrying out hydrogenation reaction at the temperature of 25 ℃ for 8h, controlling the pH of a reaction process system to be less than 5, filtering, washing a filter cake by 32ml of multiplied by 2 methanol, adding 50g of TulsionCH-97 resin into the filtrate, stirring for 3h at the temperature of 20 ℃, filtering, concentrating the filtrate at the temperature of 40 ℃ under reduced pressure to be nearly dry, adding 31.25g of purified water, dropwise adding 250ml of isopropanol at the temperature of 40-50 ℃, cooling to 20-30 ℃, stirring for 1h, continuously dropwise adding 500ml of isopropanol, cooling to 0-10 ℃ for crystallization for 2h, carrying out suction filtration, leaching the filter cake, carrying out vacuum drying at the temperature of 55 ℃ to obtain 35.0g of valganciclovir hydrochloride, obtaining the yield of 87.5%, purity of 99.2%, residual ppm of palladium 4ppm, and the HPLC data are shown in a table 2.
TABLE 2 Peak Table data for valganciclovir hydrochloride
Peak number Retention time Area of Height Area% Height%
1 4.088 75242 12243 0.938 1.162
2 4.875 2802 367 0.035 0.035
3 5.174 1231 149 0.015 0.014
4 9.935 4261113 526209 53.095 49.938
5 10.665 3677438 513697 45.823 48.751
6 11.782 1021 143 0.013 0.014
7 11.898 1104 139 0.014 0.013
8 13.959 5447 768 0.068 0.073
Total of 8025398 1053715 100.000 100.000
Note: the correction factor of the impurity A ganciclovir is 0.71, the actual content of the impurity A is 0.938% multiplied by 0.71=0.666%, and the purity of the corrected valganciclovir hydrochloride is 99.2%.
Example 2
S1: 50g of ganciclovir, 150ml of dimethyl sulfoxide, 1g of p-toluenesulfonic acid and 40g of trimethyl orthoacetate are added into a reaction bottle, and stirred and reacted for 4 hours at 25 ℃. Adding 12g of purified water into the reaction solution, stirring for 0.5h, adding 200ml of ethanol, separating out a white solid, crystallizing for 14h at 25 ℃, filtering, washing with ethanol, and drying in vacuum at 50 ℃ to obtain 48.4g of monoacetylganciclovir with the yield of 83.1 percent and the purity equivalent to that of example 1.
S2: adding 45g of monoacetylganciclovir and dmso130ml into a three-neck bottle, heating to 60 ℃, stirring for 0.5h, cooling to 30 ℃, adding 40g of Cbz-L-valine and 1g of DMAP, stirring for 15 min, controlling the temperature to be below 20 ℃, adding 40g of DCC, controlling the temperature to be 25 ℃ for reaction for 4h, controlling the temperature to be 25 ℃, adding 225mL of DCM and 225mL of purified water, stirring for 30min, filtering, washing a filter cake by dichloromethane, standing and separating the filtrate, and concentrating and evaporating the organic phase at 40 ℃ under reduced pressure.
Adding 200mL of methanol into the concentrated residue, stirring, controlling the temperature to be below 30 ℃, adding 200g of 4mol/L dilute hydrochloric acid, controlling the temperature to be 20 ℃ after the addition is finished, reacting for 6h, adding 200mL of DCM for extraction, retaining the water phase, controlling the temperature to be 10 ℃, adjusting the pH to 3 by using ammonia water, stirring for 30min, filtering, washing by using purified water, and drying in vacuum at 45 ℃ to obtain 61.40g of Cbz-L-valganciclovir, wherein the yield is 83.0%, and the purity is equivalent to that of the example 1.
S3: adding 500ml of methanol and 50g of Cbz-L-valganciclovir into a reaction bottle, stirring, controlling the temperature to be 20 ℃, slowly adding 11g of hydrochloric acid, stirring for dissolving, adding 10g of 10% palladium-carbon, replacing with nitrogen for three times, introducing hydrogen, carrying out hydrogenation reaction for 6h at 30 ℃, controlling the pH of a reaction process control system to be less than 5, filtering, washing a filter cake by 32ml of multiplied-2 methanol, adding 25g of TulsionCH-97 resin into the filtrate, stirring for 2.5h at 20 ℃, filtering, concentrating the filtrate under reduced pressure at 40 ℃ to be nearly dry, adding 41g of purified water, controlling the temperature to be 40-50 ℃, dropwise adding 328ml of isopropanol, cooling to be 20-30 ℃, stirring for 1h, continuously dropwise adding 328ml of isopropanol, cooling to be 5 ℃ for crystallization for 2h, carrying out suction filtration, carrying out leaching, carrying out vacuum drying on the filter cake at 55 ℃, obtaining 35.5g of valganciclovir hydrochloride, obtaining the yield of 88.8%, the purity of 99.3%, the palladium residue of 4ppm, and the purity is equivalent to that of the embodiment 1.
Example 3
S1: 50g of ganciclovir, 400ml of dimethyl sulfoxide, 10g of p-toluenesulfonic acid and 60g of trimethyl orthoacetate are added into a reaction bottle, and stirred and reacted for 4 hours at 25 ℃. Adding 7.2g of purified water into the reaction solution, stirring for 0.5h, adding 200ml of ethanol, separating out a white solid, crystallizing for 14h at 25 ℃, performing suction filtration, washing with ethanol, and performing vacuum drying at 45 ℃ to obtain 50.4g of monoacetylganciclovir, wherein the yield is 86.5%, and the purity is equivalent to that of example 1.
S2: adding 45g of monoacetylganciclovir and 450mL of DMSO450mL into a three-mouth bottle, heating to 70 ℃, stirring for 0.5h, cooling to 30 ℃, adding 80g of Cbz-L-valine and 10g of DMAP, stirring for 15 min, controlling the temperature to be below 20 ℃, adding 100g of DCC, controlling the temperature to be 25 ℃ for reaction for 4h, controlling the temperature to be 25 ℃, adding 225mL of DCM and 225mL of purified water, stirring for 30min, filtering, washing a filter cake by dichloromethane, standing and separating the filtrate, and concentrating and evaporating the organic phase at 40 ℃ under reduced pressure.
Adding 200mL of methanol into the concentrated residue, stirring, controlling the temperature to be below 30 ℃, adding 500g of 4mol/L dilute hydrochloric acid, controlling the temperature to be 20 ℃ after the addition is finished, reacting for 6h, adding 200mL of DCM for extraction, retaining the water phase, controlling the temperature to be 10 ℃, adjusting the pH to 1.5 by using ammonia water, stirring for 30min, filtering, washing by using purified water, and drying in vacuum at 45 ℃ to obtain 60.26g of Cbz-L-valganciclovir, wherein the yield is 81.5%, and the purity is equivalent to that of example 1.
S3: adding 500ml of methanol and 50g of Cbz-L-valganciclovir into a reaction bottle, stirring, controlling the temperature to be 20 ℃, slowly adding 11g of hydrochloric acid, stirring for dissolving, adding 20g of 10% palladium-carbon, replacing with nitrogen for three times, introducing hydrogen, carrying out hydrogenation reaction at 35 ℃ for 6h, controlling the pH of a reaction process control system to be less than 5, filtering, washing a filter cake by 32ml of multiplied-2 methanol, adding 250g of TulsionCH-97 resin into the filtrate, stirring for 2.5h at 20 ℃, filtering, concentrating the filtrate at 40 ℃ under reduced pressure to be nearly dry, adding 20.8g of purified water, controlling the temperature to be 40-50 ℃, dropwise adding 167ml of isopropanol, cooling to 20-30 ℃, stirring for 1h, continuously dropwise adding 167ml of isopropanol, cooling to 5 ℃ for crystallization for 2h, carrying out suction filtration, leaching the isopropanol, carrying out vacuum drying on the filter cake at 55 ℃, obtaining 34.2g of valganciclovir hydrochloride, obtaining, the yield being 85.5%, purity being 99.2%, and 3ppm of palladium residue, which are equivalent to the purity of the example 1.
Comparative example 1
The reaction conditions of step S1 were examined and the yield and purity were recorded, the results are shown in table 3, and the specific reaction conditions were set as follows:
comparative example 1-1: reference is made to the procedure of example 1, except that DMSO is replaced with DMF;
comparative examples 1 to 2: the procedure of example 1 was followed except that the reaction temperature was 45 ℃;
comparative examples 1 to 3: the procedure of example 1 was followed except that methanol was added after the reaction.
TABLE 3 yield and purity of step S1
Yield of Purity of
Example 1 85.0% 99.7%
Comparative examples 1 to 1 75.3% 91.5%
Comparative examples 1 to 2 60.5% 70.5%
Comparative examples 1 to 3 10.4% 99.2%
Comparative example 2
The reaction conditions of step S2 were examined and the yield and purity were recorded, the results are shown in table 4, and the specific reaction conditions were set as follows:
comparative example 2-1: reference is made to the procedure of example 1, except that DMSO is replaced with DMF;
comparative examples 2 to 2: the procedure of example 1 was followed except that ammonia was used to adjust the pH to 4;
comparative examples 2 to 3: reference is made to the procedure of example 1, except that the pH is adjusted to 6 with aqueous ammonia.
TABLE 4
Figure BDA0003849078590000121
Figure BDA0003849078590000131
Comparative example 3
The reaction conditions of step S3 were examined and the yield and purity were recorded, the results are shown in table 5, and the specific reaction conditions were set as follows:
comparative example 3-1: the procedure of example 1 was followed except that no resin treatment was used;
comparative example 3-2: reference is made to the procedure of example 1, except that the hydrogenation temperature is 40 ℃ and the time is 2h;
comparative examples 3 to 3: the procedure of example 1 was followed, except that 10g of 5% palladium-carbon was used;
comparative examples 3 to 4: the procedure of example 1 was followed, except that the post-treatment steps were: adding 31.25g of purified water, controlling the temperature to be 25 ℃, dropwise adding 250ml of isopropanol, stirring for 1h, cooling to 5 ℃, crystallizing for 2h, performing suction filtration, and leaching with 56ml of isopropanol;
comparative examples 3 to 5: the procedure of example 1 was followed except that the post-treatment steps were: adding 31.25g of purified water, controlling the temperature to be 25 ℃, dropwise adding 750ml of isopropanol, stirring for 1h, cooling to 5 ℃, crystallizing for 2h, filtering, and leaching with 56ml of isopropanol.
TABLE 5
Figure BDA0003849078590000132
Figure BDA0003849078590000141
Finally, it should be noted that: the above examples are only intended to illustrate the technical solution of the present invention, but not to limit it; although the present invention has been described in detail with reference to the foregoing embodiments, it will be understood by those of ordinary skill in the art that: the technical solutions described in the foregoing embodiments may still be modified, or some technical features may be equivalently replaced; and such modifications or substitutions do not depart from the spirit and scope of the corresponding technical solutions of the embodiments of the present invention.

Claims (10)

1. A preparation process of valganciclovir hydrochloride is characterized by comprising the following steps:
s1: stirring ganciclovir, p-toluenesulfonic acid, trimethyl orthoacetate and DMSO at normal temperature, reacting for 2-4 h, adding purified water, stirring at normal temperature for 0.25-0.5 h, adding ethanol into the reaction solution, precipitating white solid, filtering, and drying to obtain monoacetylganciclovir;
s2: adding monoacetylganciclovir into DMSO, heating to 60-70 ℃, stirring for 0.5-1h, cooling to 20-30 ℃, adding Cbz-L-valine and DMAP, stirring for 0.25-0.5h, adding DCC, stirring for 3-6 h, adding dichloromethane and purified water after the reaction is finished, stirring for 0.5h, filtering to retain an organic phase, decompressing and evaporating to dryness, adding methanol and diluted hydrochloric acid, stirring for 8-10h at 20-30 ℃, then adding dichloromethane for extraction, separating to retain a water phase, adjusting the pH to 1.5-3 with ammonia water, stirring, and filtering to obtain Cbz-L-valganciclovir;
s3: dissolving Cbz-L-valganciclovir in methanol, adjusting the pH to be less than 5 by hydrochloric acid, adding 10% palladium carbon, replacing with nitrogen, introducing hydrogen, reacting at 25 to 35 ℃ for 6 to 12h, filtering, adding TulsionCH-97 resin into filtrate, stirring at 20 to 30 ℃ for 2.5h to 3h, filtering, concentrating the filtrate under reduced pressure to dryness, and performing post-treatment to obtain valganciclovir hydrochloride.
2. The preparation process of valganciclovir hydrochloride according to claim 1, wherein the mass ratio of ganciclovir to p-toluenesulfonic acid and trimethyl orthoacetate in step S1 is 50 to 10 to 40 to 60, and the mass-volume ratio of ganciclovir to DMSO is 50g:150 to 400mL.
3. The process for preparing valganciclovir hydrochloride according to claim 1, wherein the addition amount of pure water in step S1 is 0.15 to 0.25 times of the mass of ganciclovir, and the addition amount of ethanol is 1 to 1.5 times of dimethyl sulfoxide.
4. The process for preparing valganciclovir hydrochloride according to claim 1, wherein the mass ratio of the monoacetylganciclovir to the Cbz-L-valine to the DMAP to the DCC in the step S2 is 45 to 80 to 40 to 10.
5. The process for preparing valganciclovir hydrochloride according to claim 1, wherein the mass ratio of the monoacetylganciclovir, the Cbz-L-valine, the DMAP and the DCC in the step S2 is 45 to 55.
6. The process for preparing valganciclovir hydrochloride according to claim 1, wherein the amount of dilute hydrochloric acid added in step S2 is 5~8 times the mass of monoacetylganciclovir, the concentration is 4mol/L, and the volume ratio of methanol to dilute hydrochloric acid is 1 to 1.5.
7. The process of claim 1, wherein the ratio of palladium on carbon 10% to the ratio of Cbz-L-valganciclovir hydrochloride in step S3: the mass ratio of the TulsionCH-97 resin to the Cbz-L-valganciclovir is 1 to 5, and the mass ratio of the TulsionCH-97 resin to the Cbz-L-valganciclovir is 0.5 to 5.
8. The process for preparing valganciclovir hydrochloride according to claim 1, wherein the mass ratio of 10% palladium on carbon to the Cbz-L-valganciclovir in the step S3 is 1 to 8 to 15, and the mass ratio of the TulsionCH-97 resin to the Cbz-L-valganciclovir is 0.8 to 2.
9. The process of claim 1, wherein the post-treatment step in S3 comprises: adding purified water which is 1~2 times of the mass of the concentrate, controlling the temperature at 40-50 ℃, adding isopropanol which is 7~9 times of the volume of pure water, cooling to 20-30 ℃, keeping the temperature for reaction for 1h, continuously adding isopropanol which is 8-16 times of the volume of pure water, cooling to 0-10 ℃, stirring for 1.5-2h, filtering, washing a filter cake with isopropanol, and drying to obtain the filter cake.
10. The process of claim 1, wherein the post-treatment step in S3 comprises: adding purified water with the mass being 1.5 times of that of the concentrate, controlling the temperature to be 40-50 ℃, adding isopropanol with the volume being 8 times of that of pure water, cooling to 20-30 ℃, keeping the temperature for reaction for 1h, continuously dropwise adding isopropanol with the volume being 16 times of that of the pure water, cooling to 0-10 ℃, stirring for 1.5-2h, filtering, washing a filter cake with the isopropanol, and drying to obtain the water-soluble glass.
CN202211128373.8A 2022-09-16 2022-09-16 Preparation process of valganciclovir hydrochloride Active CN115286632B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211128373.8A CN115286632B (en) 2022-09-16 2022-09-16 Preparation process of valganciclovir hydrochloride

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211128373.8A CN115286632B (en) 2022-09-16 2022-09-16 Preparation process of valganciclovir hydrochloride

Publications (2)

Publication Number Publication Date
CN115286632A true CN115286632A (en) 2022-11-04
CN115286632B CN115286632B (en) 2023-05-09

Family

ID=83833286

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211128373.8A Active CN115286632B (en) 2022-09-16 2022-09-16 Preparation process of valganciclovir hydrochloride

Country Status (1)

Country Link
CN (1) CN115286632B (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5840890A (en) * 1996-01-26 1998-11-24 Syntex (U.S.A.) Inc. Process for preparing a 2-(2-amino-1,6-dihydro-6-oxo-purin-9-yl)methoxy-1,3-propanediol derivative
CN101955481A (en) * 2010-10-09 2011-01-26 广东肇庆星湖生物科技股份有限公司 Method for preparing valganciclovir hydrochloride
CN110204505A (en) * 2019-05-31 2019-09-06 荆门医药工业技术研究院 (S) preparation process of -3- benzyloxycarbonyl group -4- isopropyl -2,5- oxazolidinedione
CN112159407A (en) * 2020-10-06 2021-01-01 湖北益泰药业股份有限公司 Preparation method of valganciclovir

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5840890A (en) * 1996-01-26 1998-11-24 Syntex (U.S.A.) Inc. Process for preparing a 2-(2-amino-1,6-dihydro-6-oxo-purin-9-yl)methoxy-1,3-propanediol derivative
CN101955481A (en) * 2010-10-09 2011-01-26 广东肇庆星湖生物科技股份有限公司 Method for preparing valganciclovir hydrochloride
CN110204505A (en) * 2019-05-31 2019-09-06 荆门医药工业技术研究院 (S) preparation process of -3- benzyloxycarbonyl group -4- isopropyl -2,5- oxazolidinedione
CN112159407A (en) * 2020-10-06 2021-01-01 湖北益泰药业股份有限公司 Preparation method of valganciclovir

Also Published As

Publication number Publication date
CN115286632B (en) 2023-05-09

Similar Documents

Publication Publication Date Title
CN106939029A (en) A kind of preparation method of Tulathromycin
EP1740564A2 (en) Process for preparation of mycophenolic acid and ester derivatives thereof
WO2010041269A1 (en) Process for preparation of mycophenolic acid, its salt and ester derivatives
CN109628541A (en) A kind of method of enzymatic clarification penicillin V salt
CN115286632A (en) Preparation process of valganciclovir hydrochloride
CN108863822B (en) Method for refining isoproterenol hydrochloride
CN111018887B (en) Method for purifying rifampicin
CN112661757A (en) Method for synthesizing valganciclovir hydrochloride
WO2023071328A1 (en) Method for synthesizing 8-amino-1-{[2-(trimethylsilyl)ethoxy]methoxy}octane-3-one
CN105085524A (en) Preparation method of high purity valganciclovir hydrochloride
CN111285914B (en) Preparation method of obeticholic acid
CN112661723A (en) Method for producing cefdinir active ester
CN112645889A (en) Refining method of Favipiravir
CN113354647A (en) Ganciclovir sodium synthesis process
CN110938075A (en) Ganciclovir preparation method
CN111961056A (en) Method for simultaneously synthesizing hypoxanthine and tetraacetyl ribose by utilizing inosine
CN112552283A (en) Preparation method of novel tinib medicine
CN112159407A (en) Preparation method of valganciclovir
CN111393444B (en) Preparation method of abacavir hydroxy acetate
CN116987148B (en) Synthesis process of acetyl hexapeptide-8
CN112300070B (en) Milrinone purification method
CN115197242B (en) Preparation method of cefpodoxime proxetil impurity I
CN116768910B (en) Refining method of rifabutin
CN114195664A (en) Preparation method of sitagliptin phosphate key intermediate
US6004801A (en) Method of purifying K-252a

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant