CN107501359B - Fondaparinux sodium impurity compound and preparation method and application thereof - Google Patents

Fondaparinux sodium impurity compound and preparation method and application thereof Download PDF

Info

Publication number
CN107501359B
CN107501359B CN201710732953.0A CN201710732953A CN107501359B CN 107501359 B CN107501359 B CN 107501359B CN 201710732953 A CN201710732953 A CN 201710732953A CN 107501359 B CN107501359 B CN 107501359B
Authority
CN
China
Prior art keywords
formula
compound
solution
water
fondaparinux sodium
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.)
Active
Application number
CN201710732953.0A
Other languages
Chinese (zh)
Other versions
CN107501359A (en
Inventor
任鹏
唐天声
汪小华
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.)
JOINCARE PHARMACEUTICAL INDUSTRY GROUP Co.,Ltd.
SHENZHEN HAIBIN PHARMACEUTICAL Co.,Ltd.
XINXIANG HAIBIN PHARMACEUTICAL Co.,Ltd.
Original Assignee
XINXIANG HAIBIN PHARMACEUTICAL CO Ltd
Shenzhen Haibin Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by XINXIANG HAIBIN PHARMACEUTICAL CO Ltd, Shenzhen Haibin Pharmaceutical Co ltd filed Critical XINXIANG HAIBIN PHARMACEUTICAL CO Ltd
Priority to CN201710732953.0A priority Critical patent/CN107501359B/en
Publication of CN107501359A publication Critical patent/CN107501359A/en
Application granted granted Critical
Publication of CN107501359B publication Critical patent/CN107501359B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/04Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N2030/022Column chromatography characterised by the kind of separation mechanism
    • G01N2030/027Liquid chromatography

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a fondaparinux sodium impurity compound and a preparation method and application thereof. The impurity compound is shown as a formula (I), and can be used as a reference substance or a standard substance of related substances for quality control of fondaparinux sodium.
Figure DDA0001387491960000011

Description

Fondaparinux sodium impurity compound and preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a fondaparinux sodium impurity compound and a preparation method and application thereof.
Background
Fondaparinux sodium Injection (Fondaparinux sodium Injection) was the first targeted anti-factor Xa inhibitor developed and produced by Sanofi corporation of francisco. The product was approved by EMEA in 12 months of 2011 and approved by the U.S. FDA in 2002. The medicine has no side effect of thrombocytopenia, is an excellent anticoagulant, and does not need monitoring in clinical application.
Fondaparinux sodium is a novel highly selective factor Xa inhibitor, a pure chemically synthesized methyl derivative of pentosan sodium, and exerts therapeutic effects mainly through specific inhibition of Xa by antithrombin (AT III). It contains five sugar building blocks, the sulfate group of which can be specifically and firmly combined with AT III and is the main endogenous regulator of blood coagulation process.
The fondaparinux sodium injection has the advantages of high bioavailability, less adverse reaction, quick response, long half-life period and more convenient clinical application. Is safer and more effective for preventing postoperative venous thrombosis. The curative effect of treating acute coronary syndrome is at least equal to that of low molecular heparin.
Therefore, the fondaparinux sodium prepared by chemical synthesis has wide prospect in the whole anticoagulation field, and the share can be continuously increased.
Figure BDA0001387491940000011
Any substance that affects the purity of the drug is collectively referred to as an impurity. The research on impurities is an important content of drug development. It includes selecting proper analysis method, accurately distinguishing and measuring impurity content and integrating the results of pharmaceutical, toxicological and clinical research to determine reasonable limit of impurity. This study is throughout the entire process of drug development. The adverse reaction generated by the medicine in clinical use is not only related to the pharmacological activity of the medicine, but also has a great relationship with impurities in the medicine.
The fondaparinux sodium compound has a complex molecular structure and a plurality of active functional groups such as hydroxyl groups, amino groups and the like, so that a plurality of impurities are generated in the synthesis reaction process, different impurities in a synthesis route are different, different impurities can be generated even if the same route has different reaction conditions, the structures of different impurities are different, and the impurities and the fondaparinux sodium structure have similar places, so that the separation difficulty of the impurities is quite high, and the product quality is difficult to predict due to the fact that specific impurities actually influence the product quality.
Nine fondaparinux sodium impurities are reported in patent US20050020536a1, and RRT is 0.8, 0.93, 1.2, 1.3, 1.4, 1.5, 1.58 and 1.6 respectively, and the impurities are very similar to the molecular structure of fondaparinux sodium, for example, the impurity compound of RRT ═ 0.8 is one sulfate group less than that of fondaparinux sodium, and the impurity compound of RRT ═ 0.93 is one sulfate group more than that of fondaparinux sodium. These impurities are inactive or poorly active, but are not very safe for administration.
Therefore, is new impurity still generated in the preparation of fondaparinux sodium by different processes? The method is very important for quality control, and also very important for the separation and purification research of impurities, and comprehensive research and strict quality control are necessary, so that the reliable quality of the fondaparinux sodium medicine and the medication safety of patients are ensured.
Disclosure of Invention
The inventor unexpectedly discovers the fondaparinux sodium impurity compound (I) in the preparation research of fondaparinux sodium, and the discovery of the compound provides possibility for carrying out the research on the impurities in a standardized way, controlling the impurities within a safe and reasonable limit range and ensuring the quality and the safety of the marketed medicines.
The invention is realized by the following technical scheme:
in one aspect, the present invention provides a compound having the structure of formula (I):
Figure BDA0001387491940000031
preferably, the compound of formula (i) has an RRT of 0.60 relative to fondaparinux sodium in the fondaparinux sodium HP L C assay.
In another aspect, the present invention provides a process for the preparation of a compound of formula (i), said process comprising the steps of:
(1) dissolving a compound shown as a formula (II) in water, carrying out reduction hydrogenation reaction, and concentrating after the reaction is finished to obtain a concentrated solution;
Figure BDA0001387491940000032
(2) the concentrated solution obtained in the step (1)Adding pyridine-sulfur trioxide (Py-SO)3) Reacting under an alkaline condition, adding ethanol, stirring, separating out a solid, performing suction filtration, and performing vacuum drying to obtain a white solid;
(3) dissolving the white solid obtained in the step (2) in water, and loading the white solid on anion exchange resin for gradient elution;
(4) and monitoring and collecting the eluent with RRT of 0.60 by ultraviolet detection, and desalting to obtain the final product.
Preferably, the step (1) comprises dissolving the compound represented by the formula (II) in water, adding Pd/C, and charging H2Carrying out reduction hydrogenation reaction, washing the carbon cake with water after the reaction is finished, filtering reaction liquid, and carrying out vacuum concentration to obtain concentrated solution;
preferably, said H2Having a pressure of 0.3-0.4 MPa;
preferably, the reaction temperature is 40 ℃.
Preferably, in the step (2), the alkaline condition is that NaOH aqueous solution is added to adjust the pH to 10.5-11.5;
preferably, in the step (2), after reacting for 1-3 hours under alkaline conditions, adding ethanol and stirring;
preferably, in step (2), the reaction temperature is 20 ℃.
Preferably, in step (3), the white solid obtained in step (2) is dissolved in water at 20-35 ℃;
preferably, in step (3), the pH value of the aqueous solution obtained by dissolving the white solid obtained in step (2) in water is 6.0 to 9.0, preferably 7.0 to 8.0;
preferably, in step (3), the anion exchange resin is based on a cross-linked polyacrylate;
preferably, in step (3), the anion exchange resin is selected from
Figure BDA0001387491940000041
Q-50S (aperture 500 angstrom, particle size 67 +/-7 um),
Figure BDA0001387491940000042
PMM Q-30S (pore diameter 500 angstroms, particle size 33. + -.3 u)m)、
Figure BDA0001387491940000043
SP-30S (aperture 500 angstrom, particle size 55 +/-5 um),
Figure BDA0001387491940000044
Q-50XS (pore size 300 angstroms, particle size 55. + -. 5um), preferably
Figure BDA0001387491940000045
Q-50S (pore diameter 500 angstrom, particle size 67 +/-7 um);
preferably, in step (3), the column efficiency of the anion exchange resin column is not less than 2000, and the tailing factor is not more than 1.4;
preferably, in step (3), the gradient elution is performed with 2 mol/L NaCl as mobile phase A and H2Taking O as mobile phase B, the following conditions are carried out:
Figure BDA0001387491940000046
Figure BDA0001387491940000051
preferably, in step (4), the ultraviolet detector wavelength is 200nm-254nm, preferably 210 nm;
preferably, in step (4), desalting is performed by column chromatography or concentration.
In addition, the invention also provides the application of the compound shown in the formula (I) as a reference substance or a standard substance of related substances in the quality control of fondaparinux sodium;
preferably, in the quality control of fondaparinux sodium, the content of the compound shown in the formula (I) is not more than 0.3 percent calculated by a peak area according to an external standard method.
Preferably, in the quality control of fondaparinux sodium, the method for measuring the content of the compound shown in the formula (I) comprises the following steps:
(1) preparation of a test solution: taking 50mg of fondaparinux sodium, placing the fondaparinux sodium in a 10ml volumetric flask, dissolving the fondaparinux sodium in water, quantitatively diluting the fondaparinux sodium to a scale, and shaking up to be used as a test solution;
(2) preparation of impurity reference substance solution: precisely weighing a proper amount of the compound shown in the formula (I), dissolving in water, and quantitatively diluting to prepare a solution containing 5mg per 1ml as a control solution;
(3) content determination: precisely measuring 100 mu l of each of the reference solution and the test solution, injecting into a liquid chromatograph, recording a chromatogram, wherein if an impurity peak exists in the chromatogram of the test solution, the retention time is not counted before 3 minutes, namely the chromatogram contains the compound shown in the formula (I), and calculating the content of the compound by using a peak area according to an external standard method;
wherein, HP L C chromatographic conditions are as follows:
column Dionex CarboPac PA1 column 4 × 250mm,
column temperature: 35 deg.C
Mobile phase A: water;
mobile phase B:2M sodium chloride solution
The flow rate is 1m L/min;
wavelength: 210nm
The elution gradient was as follows:
Figure BDA0001387491940000061
drawings
FIG. 1 shows NMR of a compound of formula (I)1H NMR spectrum;
FIG. 2 shows NMR of a compound of formula (I)13C NMR spectrum;
FIG. 3 is a high resolution mass spectrum of a compound of formula (I);
FIG. 4 is a plot of the infrared spectrum (KBr pellet) of the compound of formula (I);
FIG. 5 is a HPLC chromatogram of a crude fondaparinux sodium product (containing the compound of formula (I));
figure 6 is a HP L C spectrum (RRT 0.6) of a compound of formula (I);
FIG. 7 is a HP L C spectrum of the compound of formula (I) as a fondaparinux sodium impurity detection reference substance for detecting fondaparinux sodium.
Detailed Description
The present invention is described in further detail below with reference to specific embodiments, which are given for the purpose of illustration only and are not intended to limit the scope of the invention.
The experimental procedures in the following examples are conventional unless otherwise specified. The raw materials and reagents used in the following examples are all commercially available products unless otherwise specified.
The apparatus used was:
nuclear magnetic resonance (Bruker AVANCE III HD 500)
Mass spectrum (L TQ Orbitrap Elite)
Infrared spectrum (NICO L ET6700)
Ultraviolet spectrum (Perkin Elmer L ambda 950)
Liquid chromatography (Agilent 1260).
Preparation example: preparation method of fondaparinux sodium crude product
Dissolving 12g of the compound of formula (II) in 300m L purified water, adding 13g Pd/C, charging 0.30-0.40MPa of H2And (3) reacting at 40 ℃, washing the carbon cake with purified water after the reaction is finished, filtering the reaction solution, and concentrating in vacuum to obtain a concentrated solution of about 280m L.
Adding 21g of pyridine-sulfur trioxide into the concentrated solution, controlling the pH value to be 10.5-11.5 by using 15% sodium hydroxide aqueous solution, reacting for 1-3 hours at 20 ℃ after the addition is finished, adding ethanol, stirring, separating out a solid, filtering, and drying in vacuum to obtain 27g of white solid, namely the fondaparinux sodium crude product (containing the compound of formula (I) and having the molecular formula C)31H44N3Na9O46S7)。
Figure BDA0001387491940000071
The conditions of crude fondaparinux sodium HP L C are as follows:
chromatography column Dionex CarboPac PA1 chromatography column 4 × 250mm
Column temperature: 35 deg.C
Mobile phase A: water (W)
Mobile phase B2M sodium chloride solution
Flow rate 1.0m L/min
Wavelength: 210nm
Elution gradient:
time (minutes) Mobile phase A (%) Mobile phase B (%)
0 50 50
5 50 50
25 10 90
30 10 90
35 50 50
50 50 50
The liquid phase detection results of the obtained product are as follows:
< Peak Table > Detector: 210nm
Figure BDA0001387491940000081
Figure BDA0001387491940000091
FIG. 5 is a high performance liquid chromatography chromatogram of a crude fondaparinux sodium product; wherein the impurity with retention time of 10.281min (RRT ═ 0.60) is the compound of formula (I), and retention time 17.076 is fondaparinux sodium peak.
EXAMPLE 1 preparation of the Compound of formula (I)
At 20-35 ℃, mixing
Figure BDA0001387491940000092
Packing of Q-50S (pore size 500 a, particle size 67 ± 7um) was loaded into a preparative column with column efficiency 2000 and tailing factor 1.4, 12.0g of crude fondaparinux sodium (wherein HP L C area% of the compound of formula (i) is normalized to 3.44%) was dissolved in 1.0L water, pH of the solution was adjusted to 7.0-8.0 with dilute hydrochloric acid (5.0%, w/w), after loading, gradient elution (gradient elution conditions are shown below) with purified water washing 0.9 times column volume in the first 45 minutes, uv detection (wavelength 210nm) was monitored to collect the compound salt solution with RRT 0.60, concentration desalination, freeze drying to obtain 0.26g of white compound of formula (i) solid (HP L C area% normalized to 99.3%).
Wherein, the gradient elution conditions are as follows:
mobile phase A2 mol/L NaCl
Mobile phase B: h2O
Figure BDA0001387491940000093
Structural determination of the compounds of formula (I):
the structural data of the compound of formula (I) obtained are as follows, wherein D, E, F, G, H represents the different sugar groups respectively:
Figure BDA0001387491940000101
TABLE 1 high resolution Mass Spectrometry data for Compounds of formula (I)
Figure BDA0001387491940000102
From the measured value (m/z) of the negative ion, it can be seen that the molecule contains an odd number of nitrogen atoms, and the nitrogen rule is satisfied.
FIG. 3 is a high resolution mass spectrum of a compound of formula (I).
Impurity compound formula (i) the process for mass spectrometric cleavage of a + B is as follows:
Figure BDA0001387491940000111
(1) the structure A is as follows: pieces of
Figure BDA0001387491940000112
m/z:1047.66;
Figure BDA0001387491940000113
(2) The structure of B is as follows: pieces of+C13H19NNa3O16S2,m/z:578.39;
Figure BDA0001387491940000121
(3) The designation "H" in Table 1 is hydrogen.
TABLE 2 NMR of the compounds of formula (I) (, ppm, D)2O) detection data
Figure BDA0001387491940000122
Figure BDA0001387491940000131
Nuclear magnetic resonance of the compound of formula (I) (, ppm, D) due to the shift of the G sugar unit 2 of the compound of formula (I) to high field (the structure of the compound of formula (I) is just one sulfonic sodium salt less at the 2-position than fondaparinux sodium)2O) at position 2 of the corresponding G sugar unit in the spectrum1H NMR、13The C NMR chemical shift is relatively reduced.
A compound of formula (I) C31H44N3Na9O46S7In that13C NMR(D2O) shows a chemical shift of 22C, 9 less than the actual; and fondaparinux sodium C31H43N3Na10O49S8In that13C NMR(D2O) shows 29C, indicating that the structure of the impurity compound of formula (i) has some symmetry.
A compound of formula (I) C31H44N3Na9O46S7 1H NMR(D2O) shows a chemical shift of 27H, where 6H are overlapped since 11 active H are not present; and fondaparinux sodium C31H43N3Na10O49S8In that1H NMR(D2O) shows 34H, of which 9 active H are not shown1H NMR(D2O) also illustrates that the impurity compound of formula (i) has some symmetry.
FIG. 1 and FIG. 2 are nuclear magnetic resonances for compounds of formula (I), respectively1H NMR spectrum and13c NMR spectrum;
TABLE 3 Infrared Spectroscopy (KBr solid tablet) data for compounds of formula (I)
Figure BDA0001387491940000141
Figure BDA0001387491940000151
From the above infrared spectrum data, the compound contains a secondary amine, a methylene group, a carboxyl group (salt), a sulfonic acid group (salt), a hydroxyl group, a saturated ether, no aldehyde group, and no primary amine; thus, the structure of the impurity compound of formula (I) is consistent with the IR spectrum. FIG. 4 is a plot of the infrared spectrum (KBr pellet) of the compound of formula (I).
Fig. 6 is a HP L C spectrum of a compound of formula (I) with retention time 10.578min (RRT 0.60).
Example 2 selection of pH
At 20-35 deg.C
Figure BDA0001387491940000152
Q-50S packing (pore diameter 500 angstroms, particle size 67 + -7 um) was loaded into a preparative column with column efficiency 2000 and tailing factor 1.4, 12.0g of crude fondaparinux sodium (wherein HP L C area% of the compound of formula (I) is normalized to 3.44%) was dissolved in 1.0L water, the pH of the solution was adjusted with dilute hydrochloric acid (5.0%, w/w), after loading, gradient elution (gradient elution conditions as in example 1) with 0.9 column volume of purified water washed in the first 45 minutes, ultraviolet detection (wavelength 210nm) monitored to collect a compound salt solution with RRT 0.60, concentration, desalting, and freeze-drying to obtain a white solid compound of formula (I).
Wherein, after dissolving the fondaparinux sodium crude product in water, dilute hydrochloric acid (5.0%, w/w) is used to adjust the pH value of the solution, and the influence of different pH values on the compound of formula (I) collected by the method is shown in the following table:
selection of pH
pH 6.0-6.9 7.0-8.0 8.1-9.0
To obtain the weight (g) of the compound of formula (I) 0.16 0.26 0.18
It is known from experiments that peracids or overbasings destroy the molecular structure of the compound of formula (i), and therefore, different solution pH values result in variations in the weight (g) of the compound of formula (i) obtained.
Therefore, when the crude fondaparinux sodium is dissolved in water, it is preferable to adjust the solution to 7.0 to 8.0 with dilute hydrochloric acid (5.0%, w/w).
EXAMPLE 3 selection of column Effect
At 20-35 deg.C
Figure BDA0001387491940000161
Q-50S packing (pore diameter 500 angstroms, particle size 67 + -7 um) was loaded into preparative chromatography columns, and when column efficiency was different values, tailing factor 1.4, 12.0g of crude fondaparinux sodium (wherein HP L C area% of the compound of formula (I) was normalized to 3.44%) was dissolved in 1.0L water, pH was adjusted to 7.0-8.0 with dilute hydrochloric acid (5.0%, w/w), after loading, gradient elution (gradient elution conditions as in example 1) with 0.9 column volume of purified water in the first 45 minutes, and solution of the compound salt at RRT 0.60 was collected by monitoring with ultraviolet detection (wavelength 210nm), concentrated for desalting, and freeze-drying to obtain white solid compound of formula (I) varying with column efficiency as shown in the following table.
Selection of column efficiency
Figure BDA0001387491940000162
As can be seen from the results in the table, when the column efficiency was not less than 2000, the isolated solid compound of formula (I) had HP L C (%) purity which was superior to that of the solid compound of formula (I) obtained when the column efficiency was less than 2000.
Example 4 selection of tailing factors
At 20-35 ℃, mixing
Figure BDA0001387491940000163
Q-50S packing (pore diameter 500 angstroms, particle size 67 + -7 um) was loaded into a preparative column, column efficiency 2000, and tailing factor for different values, 12.0g of crude fondaparinux sodium (wherein HP L C area percent of the compound of formula (I) is normalized to 3.44%) was dissolved in 1.0L water, pH of the solution was adjusted to 7.0-8.0 with dilute hydrochloric acid (5.0%, w/w), after loading, gradient elution (gradient elution conditions same as in example 1) in which purified water was washed 0.9 times column volume over the first 45 minutes, and solution of the compound salt was collected at RRT 0.60 monitored by UV detection (wavelength 210nm), concentrated for desalting, and freeze-dried to obtain white solid compound of formula (I) HP L C (%) with tailing factor variation.
Selection of tailing factors
Figure BDA0001387491940000171
Because the fondaparinux sodium has similar structures with the impurities, the fondaparinux sodium is very difficult to separate, so that the separation of a plurality of impurities with similar structures is very difficult; because the liquid chromatogram peak positions of the impurities and the fondaparinux sodium are very similar.
Therefore, the column tail factor is important, and the tail factor is serious, which directly affects the purity of the product, so that the purity of the solid compound of formula (I) (HP L C%) is significantly reduced when the column tail factor is more than 1.4, and the purity of the solid compound of formula (I) (HP L C%) is more than 99.3% when the column tail factor is less than 1.4.
EXAMPLE 5 selection of wavelength
At 20-35 ℃, mixing
Figure BDA0001387491940000173
Q-50S packing (pore diameter 500 angstroms, particle size 67 + -7 um) was loaded into a preparative column with column efficiency of 2000 and tailing factor of 1.4, and 12.0g of crude fondaparinux sodium (wherein the HP L C area% of the compound of formula (I) is normalized to 3.44%) was dissolved in 1.0L of water and usedAdjusting the pH value of the solution to 7.0-8.0 by using dilute hydrochloric acid (5.0%, w/w); after loading, gradient elution (same gradient elution conditions as in example 1) was performed, a compound salt solution with RRT of 0.60 was collected under monitoring at different uv detection wavelengths, concentrated for desalination, and freeze-dried to obtain a white solid compound of formula (i) whose weight (g) varied with different uv detection wavelengths as shown in the following table.
Selection of wavelength
Wavelength (nm) 200 210 254
To obtain the weight (g) of the compound of formula (I) 0.13 0.26 0.06
Since different compounds absorb different amounts of UV light, the compound of formula (I) is more advantageous for observation and collection when the wavelength is set to 210 nm.
EXAMPLE 6 preparation of the Compound of formula (I)
At 20-35 ℃, mixing
Figure BDA0001387491940000172
Loading PMM Q-30S (pore diameter 500 angstrom, particle diameter 33 + -3 um) filler into a medium-pressure preparative chromatographic column, dissolving tailing factor 1.4.15.0 g crude fondaparinux sodium in 1.5L water, controlling pH to 7.0-8.0, loading, and gradient eluting (gradient eluting conditions are the same as in example 1), wherein purified water is washed with 0m in the first 45 min9 times column volume, flow rate 230 ml/min; the salt solution of the compound with RRT of 0.60 was collected under ultraviolet detection (wavelength 210nm), concentrated for desalting, and freeze-dried to obtain 0.14g of a white solid compound of formula (i) with a purity of 99.0%.
EXAMPLE 7 preparation of the Compound of formula (I)
At 20-35 ℃, mixing
Figure BDA0001387491940000181
SP-30S (pore diameter 500 angstrom, particle size 55 +/-5 um) filler is filled into a medium-pressure preparative chromatographic column, column efficiency is 2000, tailing factor is 1.4, 9.0g of crude fondaparinux sodium is dissolved in 0.9L water, pH is controlled to be 7.0-8.0, after sample loading, gradient elution (the gradient elution condition is the same as that of example 1), wherein purified water is flushed for the first 45 minutes for 0.9 times of column volume, flow rate is 230ml/min, compound salt solution with RRT of 0.60 is collected by ultraviolet detection (wavelength is 210nm) monitoring, concentrated and desalted, and freeze-dried to obtain 0.102g of white solid compound with the purity of 99.1 percent of the formula (I).
EXAMPLE 8 preparation of the Compound of formula (I)
At 20-35 ℃, mixing
Figure BDA0001387491940000182
Q-50XS (pore diameter 300 angstroms, particle size 55 +/-5 microns) is filled into a preparative chromatographic column, the column efficiency is 2000, a tailing factor 1.4.12.0 g of crude fondaparinux sodium is dissolved in 1.0L water, the pH is controlled to be 7.0-8.0, after the sample is loaded, gradient elution is carried out (the gradient elution condition is the same as that of example 1), wherein purified water is washed for the first 45 minutes for 0.9 times of the column volume, the flow rate is 230ml/min, compound salt solution with the RRT of 0.60 is collected by monitoring through ultraviolet detection (210nm), and the white solid compound with the formula (I) with the purity of 99.0 percent is obtained by concentration, desalination and freeze drying, namely 0.185 g.
Example 9 use of Compounds of formula (I) as reference for detection of fondaparinux sodium impurities
HP L C chromatographic conditions were as follows:
column Dionex CarboPac PA1 column 4 × 250mm,
column temperature: 35 deg.C
Mobile phase A: water;
mobile phase B:2M sodium chloride solution
The flow rate is 1m L/min;
wavelength: 210nm
The elution gradient was as follows:
time (minutes) Mobile phase A (%) Mobile phase B (%)
0 50 50
5 50 50
25 10 90
30 10 90
35 50 50
50 50 50
Test solution: taking 50mg of fondaparinux sodium, putting the fondaparinux sodium into a 10ml volumetric flask, dissolving the fondaparinux sodium in water, quantitatively diluting the fondaparinux sodium to a scale, and shaking up to obtain a test solution.
Impurity control solution: an appropriate amount of the impurity control substance represented by the formula (I) (prepared in example 1) was precisely weighed, dissolved in water and quantitatively diluted to prepare a solution containing 5mg per 1ml as a control solution.
The determination method comprises the following steps: precisely measuring 100 mu l of each of the reference solution and the test solution, injecting into a liquid chromatograph, recording a chromatogram, and calculating according to an external standard method by using peak area, wherein if an impurity peak (the retention time is not counted before 3 minutes) exists in the chromatogram of the test solution, the compound shown in the formula (I) is contained, and the content of the compound is not more than 0.3%.
The results are shown in FIG. 7. Therefore, the analysis method can well separate the compound shown in the formula (I) from fondaparinux sodium and other impurities, and the compound shown in the formula (I) can be used as a fondaparinux sodium impurity detection reference substance.
The reasonable limit of the compound of formula (I) is determined to be no more than 0.3% by a combination of the results of pharmacological, toxicological and clinical studies.

Claims (12)

1. A process for the preparation of a compound of formula (I),
Figure FDF0000008696140000011
the method comprises the following steps:
(1) dissolving a compound shown as a formula (II) in water, carrying out reduction hydrogenation reaction, and concentrating after the reaction is finished to obtain a concentrated solution;
Figure FDF0000008696140000012
(2) adding pyridine-sulfur trioxide into the concentrated solution obtained in the step (1), reacting under an alkaline condition, adding ethanol, stirring, separating out a solid, performing suction filtration, and performing vacuum drying to obtain a white solid;
(3) dissolving the white solid obtained in the step (2) in water, and loading the white solid on anion exchange resin for gradient elution;
(4) monitoring and collecting the eluent with RRT of 0.60 by ultraviolet detection, and desalting to obtain the final product;
wherein in the step (3), the pH value of an aqueous solution obtained by dissolving the white solid obtained in the step (2) in water is 7.0-8.0, the column efficiency of the anion exchange resin column is not less than 2000, the tailing factor is not more than 1.4, the anion exchange resin takes crosslinked polyacrylate as a matrix, the gradient elution takes 2 mol/L NaCl as a mobile phase A, and H is used as a mobile phase A2Taking O as mobile phase B, the following conditions are carried out:
Figure FDF0000008696140000021
in the step (4), the wavelength of the ultraviolet detector is 210 nm.
2. The method according to claim 1, wherein the step (1) comprises dissolving the compound represented by the formula (II) in water, adding Pd/C, and charging H2Carrying out reduction hydrogenation reaction, washing the carbon cake with water after the reaction is finished, filtering the reaction solution, and carrying out vacuum concentration to obtain a concentrated solution.
3. The method of claim 2, wherein the H is2Has a pressure of 0.3-0.4 MPa.
4. The process according to claim 2, characterized in that the reaction temperature is 40 ℃.
5. The method according to claim 1, wherein in step (2), the alkaline condition is that an aqueous NaOH solution is added to adjust the pH to 10.5-11.5.
6. The method of claim 1, wherein in the step (2), ethanol is added and stirred after the reaction is carried out for 1 to 3 hours under alkaline conditions.
7. The method according to claim 1, wherein in step (2), the reaction temperature is 20 ℃.
8. The method according to claim 1, wherein in step (3), the white solid obtained in step (2) is dissolved in water at 20-35 ℃.
9. The method according to claim 1, wherein in step (3), the anion exchange resin is selected from the group consisting of those having a pore diameter of 500 angstroms and a particle diameter of 67 ± 7 μm
Figure FDF0000008696140000031
Q-50S, pore diameter of 500 angstroms, particle diameter of 33 +/-3 microns
Figure FDF0000008696140000032
PMM Q-30S, pore diameter 500 angstrom, particle size 55 + -5 μm
Figure FDF0000008696140000033
SP-30S, pore diameter of 300 angstroms, and particle diameter of 55 +/-5 microns
Figure FDF0000008696140000034
Q-50XS。
10. The method according to claim 1, wherein in step (3), the anion exchange resin has a pore diameter of 500 angstroms and a particle diameter of 67 ± 7 μm
Figure FDF0000008696140000035
Q-50S。
11. The method of claim 1, wherein in step (4), desalting is performed by column chromatography or concentration.
12. A method for measuring the content of a compound represented by the formula (I) obtained by the production method according to any one of claims 1 to 11, which comprises the steps of:
(1) preparation of a test solution: taking 50mg of fondaparinux sodium, placing the fondaparinux sodium in a 10ml volumetric flask, dissolving the fondaparinux sodium in water, quantitatively diluting the fondaparinux sodium to a scale, and shaking up to be used as a test solution;
(2) preparation of impurity reference substance solution: precisely weighing a proper amount of the compound shown in the formula (I), dissolving in water, and quantitatively diluting to prepare a solution containing 5mg per 1ml as a control solution;
(3) content determination: precisely measuring 100 mu l of each of the reference solution and the test solution, injecting into a liquid chromatograph, recording a chromatogram, wherein if an impurity peak exists in the chromatogram of the test solution, the retention time is not counted before 3 minutes, namely the chromatogram contains the compound shown in the formula (I), and calculating the content of the compound by using a peak area according to an external standard method;
wherein, HP L C chromatographic conditions are as follows:
column Dionex CarboPac PA1 column 4 × 250mm,
column temperature: 35 deg.C
Mobile phase A: water;
mobile phase B:2M sodium chloride solution
The flow rate is 1m L/min;
wavelength: 210nm
The elution gradient was as follows:
Figure FDF0000008696140000036
Figure FDF0000008696140000041
CN201710732953.0A 2017-08-24 2017-08-24 Fondaparinux sodium impurity compound and preparation method and application thereof Active CN107501359B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201710732953.0A CN107501359B (en) 2017-08-24 2017-08-24 Fondaparinux sodium impurity compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710732953.0A CN107501359B (en) 2017-08-24 2017-08-24 Fondaparinux sodium impurity compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN107501359A CN107501359A (en) 2017-12-22
CN107501359B true CN107501359B (en) 2020-07-31

Family

ID=60692599

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710732953.0A Active CN107501359B (en) 2017-08-24 2017-08-24 Fondaparinux sodium impurity compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN107501359B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108896696A (en) * 2018-04-18 2018-11-27 南京健友生化制药股份有限公司 Dissociate in a kind of Fondaparinux sodium injection the detection method of sulfate radical
CN113461744B (en) * 2020-03-30 2024-03-15 鲁南制药集团股份有限公司 Purification method of fondaparinux sodium intermediate

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1558911A (en) * 2001-09-07 2004-12-29 Synthetic heparin pentasaccharides
US20050020536A1 (en) * 2003-02-27 2005-01-27 Jean-Francois Branellec Highly pure fondaparinux sodium composition, process for preparing said composition and pharmaceutical compositions containing it as active principle
CN103122012A (en) * 2011-11-17 2013-05-29 江苏恒瑞医药股份有限公司 Compound for preparing fondaparinux sodium, preparation method thereof and preparation method of fondaparinux sodium
CN104910217A (en) * 2015-06-19 2015-09-16 天津红日药业股份有限公司 Reference compound for controlling quality of fondaparinux sodium
CN105985387A (en) * 2015-02-04 2016-10-05 苏州朗科生物技术有限公司 Fondaparinux sodium impurity compound, preparation method therefor and use of Fondaparinux sodium impurity compound
CN105985388A (en) * 2015-02-04 2016-10-05 苏州朗科生物技术有限公司 Fondaparinux sodium impurity compound, preparation method therefor and application of Fondaparinux sodium impurity compound

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1558911A (en) * 2001-09-07 2004-12-29 Synthetic heparin pentasaccharides
US20050020536A1 (en) * 2003-02-27 2005-01-27 Jean-Francois Branellec Highly pure fondaparinux sodium composition, process for preparing said composition and pharmaceutical compositions containing it as active principle
CN103122012A (en) * 2011-11-17 2013-05-29 江苏恒瑞医药股份有限公司 Compound for preparing fondaparinux sodium, preparation method thereof and preparation method of fondaparinux sodium
CN105985387A (en) * 2015-02-04 2016-10-05 苏州朗科生物技术有限公司 Fondaparinux sodium impurity compound, preparation method therefor and use of Fondaparinux sodium impurity compound
CN105985388A (en) * 2015-02-04 2016-10-05 苏州朗科生物技术有限公司 Fondaparinux sodium impurity compound, preparation method therefor and application of Fondaparinux sodium impurity compound
CN104910217A (en) * 2015-06-19 2015-09-16 天津红日药业股份有限公司 Reference compound for controlling quality of fondaparinux sodium

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
3-O-Sulfated Oligosaccharide Structures Are Recognized by Anti-heparan;Gerdy B. ten Dam,等;《Journal of Biological Chemistry》;20061231;第281卷(第8期);第4654-4662页 *
Interaction of heparin and antithrombin III. The role of O-sulfate groups;Maurice PETITOU,等;《European Journal of Biochemistry》;19881231;第176卷(第3期);第638页 *

Also Published As

Publication number Publication date
CN107501359A (en) 2017-12-22

Similar Documents

Publication Publication Date Title
EP3421503B1 (en) Sugammadex preparation and purification method
CN107501359B (en) Fondaparinux sodium impurity compound and preparation method and application thereof
CN110013836B (en) Reversed phase/ion exchange mixed mode chromatographic stationary phase, preparation method and application
CN105334301B (en) Pyrrolo quinoline quinone (PQQ) disodium salt impurity separation and purification method
CN110997666A (en) N-formyl palbociclib, preparation method and application thereof, palbociclib preparation and quality control method thereof
Chi et al. Preparation of phenyl-boronic acid polymer monolith by initiator-free ring-opening polymerization for microextraction of sulfamethoxazole and trimethoprim from animal-originated foodstuffs
CN110108818B (en) High performance liquid chromatography for high performance separation detection of phthalide derivatives and application thereof
CN110709696B (en) Method for separating and analyzing mixture of oligonucleotides
WO2020238529A1 (en) Parecoxib impurity reference substance and preparation method therefor
CN106706769B (en) Separation and determination method of empagliflozin and optical isomer thereof
CN109212093B (en) HPLC detection method of valganciclovir hydrochloride intermediate condensation compound isomer
CN104650140B (en) A kind of preparation method of high-purity good fortune department Fluconazole
CN114113412B (en) Analysis method of posaconazole impurity related substances
Wang et al. Detection of two genotoxic impurities in drug substance and preparation of imatinib mesylate by LC–MS/MS
CN115215765A (en) Preparation method of kreb polymer impurity
CN112213407B (en) Detection method of levoornidazole related substances
CN107778290B (en) Impurity of quinoline derivative and preparation method thereof
EP3505929B1 (en) Packing material for hilic columns, hilic column filled with same, and method for analyzing oligosaccharides with said column
CN111217691A (en) Compound 1- (2-benzyl-3, 5-bis (benzyloxy) phenyl) ethanone, preparation method and application thereof
CN114200066A (en) Separation and detection method for ambrisentan related substances
US20080287675A1 (en) Cascade system
CN102993087A (en) Impurity compound in Tegafur/Gimeracil/Oteracil prescription, and preparation method and application thereof
CN110759948B (en) Platinum-containing compound, preparation method, separation method and application thereof
CN106632615A (en) Extraction process of vasopressin solution
CN111032635B (en) N-formyl vortioxetine, preparation method thereof and vortioxetine solid preparation

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
TR01 Transfer of patent right

Effective date of registration: 20200928

Address after: 518081 No. 2003, Sha Mo Road, Yantian District, Guangdong, Shenzhen

Patentee after: SHENZHEN HAIBIN PHARMACEUTICAL Co.,Ltd.

Patentee after: XINXIANG HAIBIN PHARMACEUTICAL Co.,Ltd.

Patentee after: JOINCARE PHARMACEUTICAL INDUSTRY GROUP Co.,Ltd.

Address before: 518081 No. 2003, Sha Mo Road, Yantian District, Guangdong, Shenzhen

Patentee before: SHENZHEN HAIBIN PHARMACEUTICAL Co.,Ltd.

Patentee before: XINXIANG HAIBIN PHARMACEUTICAL Co.,Ltd.

TR01 Transfer of patent right