CN116239513A - Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug - Google Patents

Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug Download PDF

Info

Publication number
CN116239513A
CN116239513A CN202310494927.4A CN202310494927A CN116239513A CN 116239513 A CN116239513 A CN 116239513A CN 202310494927 A CN202310494927 A CN 202310494927A CN 116239513 A CN116239513 A CN 116239513A
Authority
CN
China
Prior art keywords
product
mmae
solvent
preparation
reaction
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
CN202310494927.4A
Other languages
Chinese (zh)
Other versions
CN116239513B (en
Inventor
李九远
董博
李常峰
王金超
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.)
Tianjin Asymchem Pharmaceutical Co Ltd
Original Assignee
Tianjin Asymchem 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 Tianjin Asymchem Pharmaceutical Co Ltd filed Critical Tianjin Asymchem Pharmaceutical Co Ltd
Priority to CN202310494927.4A priority Critical patent/CN116239513B/en
Publication of CN116239513A publication Critical patent/CN116239513A/en
Application granted granted Critical
Publication of CN116239513B publication Critical patent/CN116239513B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/08Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon radicals, substituted by hetero atoms, attached to ring carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • 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

Abstract

The invention provides a preparation method of an MMAE key intermediate, a preparation method of the MMAE and an antibody coupling drug. The preparation method of the MMAE key intermediate comprises the following steps: step S1, carrying out condensation reaction on a D-alanine compound and N, O-dialkylhydroxylamine salt in an alkaline environment to obtain a first product; step S2, R on the first product 1 Removing to obtain a second product; step S3, performing condensation coupling reaction on the second product and a pyridine alkyl carboxylic acid compound to obtain a third product; step S4, coupling reaction is carried out on the third product and the phenyl compound, so as to obtain a fourth product; s5, selectively reducing the fourth product to obtain an MMAE key intermediate; by applying the technical scheme of the invention, the key intermediate of the MMAE is synthesized by adopting cheap and easily available commercial raw materials as the initial raw materials, thereby avoiding the direct participation of the norephedrine in the reaction and providing a novel synthetic method for the MMAE.

Description

Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug
Technical Field
The invention relates to the technical field of pharmaceutical chemistry, in particular to a preparation method of an MMAE key intermediate, a preparation method of the MMAE and an antibody coupling drug.
Background
The Antibody conjugated drug anti-drug Conjugate (ADC) is formed by coupling a chemotherapeutic drug with strong cytotoxicity with monoclonal Antibody through a linker, and has strong killing power of a small molecular drug and high targeting of pure monoclonal Antibody, so that the Antibody conjugated drug is a research and development hot spot of tumor targeted therapy. The ADC drug is characterized in that: the treatment efficacy is strong; the tumor cell specificity is high, and the false killing rate is low; the immunogenicity is weak, and drug resistance is not easy to generate; the circulation time in serum is long; has weak cytotoxicity to non-target cells.
The antibody class (macromolecular fraction) of ADC drugs is unlimited, whereas the class (small fraction) of toxins is limited, monomethyl auristatin E Monomethyl auristation E (MMAE; SGD-1010; CAS: 474645-27-7) being the most commonly used class of toxin compounds. MMAE is a synthetic derivative of dolastatin 10, is a novel, synthetic, antimitotic/antimitotic protein capable of blocking tubulin polymerization, shows strong activity against a range of lymphomas, leukemias and solid tumors in preclinical studies in vitro and in vivo, has extremely strong antitumor effects, belongs to the class of antimicrotubules as vinblastine, but has toxicity approximately equal to 200 times that of vinblastine. However, due to its high toxicity, it cannot be used as a drug directly. MMAE is widely used as a cytotoxic ingredient in the manufacture of antibody-conjugated drugs to treat cancer.
The existing disclosed synthetic methods of MMAE all adopt norephedrine as a starting material, but the norephedrine belongs to a class of easy-to-poison compounds in China and cannot be widely applied. Thus, there is a need in the art to develop a synthetic method of MMAE that does not require the use of norephedrine as a starting material.
Disclosure of Invention
The invention mainly aims to provide a preparation method of an MMAE key intermediate, a preparation method of the MMAE and an antibody coupling drug, so as to solve the problem that the synthesis method of the MMAE in the prior art needs to adopt an easily-toxic compound, namely, norephedrine, as a starting material, so that the MMAE cannot be widely applied.
In order to achieve the above object, according to a first aspect of the present invention, there is provided a process for preparing an MMAE-critical intermediate having a structure corresponding to the following formula (i):
Figure SMS_1
formula (I);
the preparation method comprises the following steps: step S1, carrying out condensation reaction on a D-alanine compound and N, O-dialkylhydroxylamine salt in an alkaline environment to obtain a first product system, and purifying the first product system to obtain a first product; the D-alanine compound has a structure shown in the following formula (II), the N, O-dialkylhydroxylamine salt has a structure shown in the following formula (III), and the first product has a structure shown in the following formula (IV); step S2, R on the first product 1 Removing to obtain a second product system, and purifying the second product system to obtain a second product, wherein the second product has a structure shown in the following formula (V); step S3, performing condensation coupling reaction on the second product and a pyridine alkyl carboxylic acid compound to obtain a third product system, and purifying the third product system to obtain a third product, wherein the pyrrole alkyl carboxylic acid compound has a structure shown in the following formula (VI), and the third product has a structure shown in the following formula (VI)
Figure SMS_2
) The structure is shown; step S4, coupling reaction is carried out on the third product and a phenyl compound to obtain a fourth product system, and the fourth product system is purified to obtain a fourth product, wherein the phenyl compound has the following formula (the formula:)>
Figure SMS_3
) The structure shown, the fourth product has the formula (++>
Figure SMS_4
) The structure is shown; s5, selectively reducing the fourth product to obtain a fifth product system, and purifying the fifth product system to obtain an MMAE key intermediate;
Figure SMS_5
the [ (x) ray ]Ⅱ);/>
Figure SMS_6
Formula (III); />
Figure SMS_7
Formula (IV);
Figure SMS_8
formula (V); />
Figure SMS_9
(-)>
Figure SMS_10
);
Figure SMS_11
(-)>
Figure SMS_12
);
Wherein R and R 1 Each independently represents a protecting group comprising at least one of an alkoxycarbonyl group, an acyl group or an alkyl group; z represents hydrochloric acid, nitric acid or sulfuric acid; r is R a Is methyl; r is R 2 And R is 3 Each independently represents a C1 to C6 alkyl group; m represents a metal or a halogenated metal.
Further, R and R 1 Each independently represents benzyloxycarbonyl, t-butyloxycarbonyl, fluorenylmethoxycarbonyl, allyloxycarbonyl, trimethylsilylethoxycarbonyl, benzoyl, phthaloyl, p-toluenesulfonyl, trifluoroacetyl, trityl, 2,4 dimethoxybenzyl, p-methoxybenzyl or benzyl; and/or R 2 And R is 3 Each independently represents a C1 to C4 alkyl group; and/or M represents lithium, sodium, potassium, magnesium bromide, magnesium chloride or magnesium iodide.
Further, R and R 1 Each independently represents a benzyloxycarbonyl group, a t-butyloxycarbonyl group, a fluorenylmethoxycarbonyl group or a benzoyl group; and/or, Z represents hydrochloric acid; and/or R 2 And R is 3 Each independently represents methyl;and/or M represents lithium or magnesium bromide.
Further, in the step S1, the pH value of the alkaline environment is 8.0-11.0.
Further, in the above step S1, the condensation reaction is performed in an alkaline environment formed by a first solvent including at least one of dichloromethane, tetrahydrofuran, 2-methyltetrahydrofuran, and N, N-dimethylformamide.
Further, the temperature of the condensation reaction is 0-10 ℃, and the time of the condensation reaction is 4.0-24.0 h.
Further, the purification of the first product system includes quenching, extraction and concentration, which are performed sequentially.
Further, in the step S2, R is performed on the first product and the hydrogen chloride solution in a second solvent 1 The removal reaction is carried out, and the hydrogen chloride solution is preferably at least one of a hydrogen chloride methanol solution, a hydrogen chloride dioxane solution, a hydrogen chloride ethanol solution or an isopropyl hydrogen acetate solution.
Further, the second solvent includes at least one of methanol, ethanol, dioxane or isopropyl acetate.
Further, R 1 The temperature of the removal reaction is 0-10 ℃, and the time of the removal reaction is 2.0-8.0 h.
Further, the purification of the second product system comprises solid-liquid separation, extraction and concentration, which are performed sequentially.
Further, in the step S3, the second product and the pyrrolidinyl carboxylic acid compound are subjected to condensation coupling reaction under the action of a condensation reagent, wherein the condensation reagent is preferably at least one of HOBT/EDCI, HATU, HOBT/HBTU or T3P.
Further, the second product is subjected to a condensation coupling reaction with a pyrrolidinecarboxylic acid compound in a third solvent, preferably at least one of N, N-dimethylformamide, methylene chloride, tetrahydrofuran and 2-methyltetrahydrofuran.
Further, the temperature of the condensation coupling reaction is 15-25 ℃, and the time of the condensation coupling reaction is 1.0-8.0 h.
Further, the purification of the third product system comprises quenching, crystallization, recrystallization or beating, which are performed sequentially.
Further, in the step S5, the fourth product is selectively reduced by a reducing agent, and the reducing agent preferably includes at least one of aluminum isopropoxide, tributyl lithium hydride or lithium aluminum hydride.
Further, the fourth product and the reducing agent are selectively reduced in a fifth agent, which is preferably at least one of toluene, an ether solvent or an alcohol solvent.
Further, the temperature of the selective reduction is 40-55 ℃, and the time of the selective reduction is 1.0-8.0 h.
Further, the purification of the above substrate product system includes quenching, extraction, quenching, concentration, crystallization and beating, which are sequentially performed.
In order to achieve the above object, according to a second aspect of the present invention, there is also provided a process for producing MMAE, the process comprising a process for producing MMAE-critical intermediate, the process for producing MMAE-critical intermediate being any one of the processes for producing MMAE-critical intermediate provided in the first aspect.
According to a third aspect of the present invention, there is also provided an antibody-conjugated drug, the raw material of which comprises an antibody and the MMAE key intermediate obtained by any one of the preparation methods provided in the first aspect.
By using the technical scheme of the invention, the MMAE key intermediate with the structure shown in the formula (I) is synthesized by adopting cheap and easily available commercial raw materials as starting materials, thereby avoiding the direct participation of norephedrine in the reaction and providing a new synthesis method for the MMAE. In addition, the preparation method of the MMAE key intermediate provided by the invention has mild and controllable reaction conditions, fully considers the atom economy of the reaction, effectively improves the yield of the product, and simultaneously has the advantages of green chemistry concept, low cost of the reaction solvent, environmental friendliness and good cost advantage and commercialization basis.
Drawings
The accompanying drawings, which are included to provide a further understanding of the invention and are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and together with the description serve to explain the invention.
In the drawings:
FIG. 1 shows a nuclear magnetic spectrum of a key intermediate of MMAE obtained by the preparation method provided in example 9 of the present invention;
Fig. 2 shows a high performance liquid chromatogram of the MMAE key intermediate obtained according to the preparation method provided in example 9 of the present invention.
Detailed Description
It should be noted that, in the case of no conflict, the embodiments and features in the embodiments may be combined with each other. The present invention will be described in detail with reference to examples.
As analyzed in the background of the present application, in the existing MMAE synthesis methods, norephedrine is required to be used as a starting material, but norephedrine belongs to a class of easily toxic compounds in China, so that the norephedrine cannot be widely applied. In order to solve the technical problem, the application provides a preparation method of an MMAE key intermediate, a preparation method of the MMAE and an antibody coupling drug.
In one exemplary embodiment of the present application, a process is provided for the preparation of an MMAE key intermediate having the structure of formula (i):
Figure SMS_13
formula (I);
the preparation method of the MMAE key intermediate with the structure shown in the formula (I) comprises the following steps: step S1, carrying out condensation reaction on a D-alanine compound and N, O-dialkylhydroxylamine salt in an alkaline environment to obtain a first product system, and purifying the first product system to obtain a first product; the D-alanine compound has a structure shown in the following formula (II), the N, O-dialkylhydroxylamine salt has a structure shown in the following formula (III), and the first product has a structure shown in the following formula (IV);
Figure SMS_14
Formula (II); />
Figure SMS_15
Formula (III); />
Figure SMS_16
Formula (IV);
step S2, R on the first product 1 Removing to obtain a second product system, and purifying the second product system to obtain a second product, wherein the second product has a structure shown in the following formula (V);
Figure SMS_17
formula (V);
step S3, performing condensation coupling reaction on the second product and the pyrrolidinyl carboxylic acid compound to obtain a third product system, and purifying the third product system to obtain a third product;
Figure SMS_18
formula (VI); />
Figure SMS_19
(-)>
Figure SMS_20
);
The pyrrolidinyl carboxylic acid compound has the structure shown in the following formula (VI), and the third product has the structure shown in the following formula
Figure SMS_21
) The structure is shown; step S4, coupling reaction is carried out on the third product and phenyl compound to obtain a fourth product system, and the fourth product system is purified to obtain a fourth product, wherein the phenyl compound has the following formula ()>
Figure SMS_22
) The structure shown, the fourth product has the formula (++>
Figure SMS_23
) The structure is shown;
Figure SMS_24
(-)>
Figure SMS_25
);/>
Figure SMS_26
(-)>
Figure SMS_27
);
And S5, selectively reducing the fourth product to obtain a fifth product system, and purifying the fifth product system to obtain the MMAE key intermediate.
The formula (I) to (II)
Figure SMS_28
) Wherein R and R 1 Each independently represents a protecting group comprising at least one of an alkoxycarbonyl group, an acyl group or an alkyl group; z represents hydrochloric acid, nitric acid or sulfuric acid; r is R a Is methyl; r is R 2 And R is 3 Each independently represents a C1 to C6 alkyl group; m represents a metal or a halogenated metal.
The above alkoxycarbonyl group includes, but is not limited to, any one of benzyloxycarbonyl, t-butyloxycarbonyl, fluorenylmethoxycarbonyl, or allyloxycarbonyl, and the above acyl group includes, but is not limited to, phthaloyl or p-toluenesulfonyl; the above alkyl group includes, but is not limited to, any of trityl, 2,4 dimethoxybenzyl, p-methoxybenzyl or benzyl.
In order to further increase the efficiency of the preparation of the above-mentioned MMAE key intermediates, R and R are preferably selected 1 Each independently represents benzyloxycarbonyl (Cbz), t-butyloxycarbonyl (Boc), fluorenylmethoxycarbonyl (Fmoc), allyloxycarbonyl (Alloc), trimethylsilylethoxycarbonyl (Teoc), benzoyl (Bz), phthaloyl (Pht), p-toluenesulfonyl (Tos), trifluoroacetyl (Tfa), trityl (Trt), 2,4 Dimethoxybenzyl (DMB)Any one of p-methoxybenzyl (Pmb) or benzyl (Bn); and/or, preferably R 2 And R is 3 Each independently represents a C1-C4 alkyl group such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, or the like; and/or M represents any one of lithium, sodium, potassium, magnesium bromide, magnesium chloride or magnesium iodide.
In order to further reduce the controllability of the reaction conditions and to further increase the purity and yield of MMAE key intermediates, R and R are preferred 1 Each independently represents benzyloxycarbonyl (Cbz), t-butyloxycarbonyl (Boc), fluorenylmethoxycarbonyl (Fmoc) or benzylformyl (Bz); and/or, Z represents hydrochloric acid; and/or R 2 And R is 3 Each independently represents methyl; and/or M represents lithium or magnesium bromide.
In some embodiments of the present application, in step S1, in order to further improve the efficiency of the condensation reaction, the pH of the alkaline environment is preferably 8 to 11.0.
In order to further accelerate the rate of the condensation reaction, it is preferable that the above condensation reaction is carried out in an alkaline environment formed by a first solvent including, but not limited to, a mixed solvent formed by any one or more of methylene chloride, tetrahydrofuran, 2-methyltetrahydrofuran, and N, N-dimethylformamide.
In order to further promote the condensation reaction to proceed more completely and to further enhance the efficiency of the first product production, it is preferable that the temperature of the condensation reaction is 0 to 10 ℃ and the time of the condensation reaction is 4.0 to 24.0 hours.
Typically, but not by way of limitation, the pH of the alkaline environment is, for example, 8.0, 8.2, 8.5, 9.0, 9.4, 9.8, 10.0, 10.5, 11.0 or any two values in the range; the temperature of the condensation reaction is, for example, 0 ℃,1 ℃, 2 ℃, 5 ℃, 8 ℃, 10 ℃ or a range value consisting of any two values; the time of the above-mentioned condensation reaction is, for example, 4h, 8h, 12h,15h, 18h, 20h, 24h or a range of values consisting of any two values.
The above-mentioned method for purifying the first product system is not limited, and any purification method capable of separating the first product from the first product system may be used. To further increase the yield of the first product system, it is preferred that the purification of the first product system comprises quenching, extraction and concentration, which are performed sequentially.
From the standpoint of environmental protection and cost reduction, the first product system is preferably quenched by an aqueous solution of sodium bicarbonate, and the mass concentration of the sodium bicarbonate is preferably 10% -15%. In order to further improve the yield of the first product, the quenched first product system is preferably extracted by an organic solvent, so as to obtain a first product extraction phase, the type of the organic solvent is not limited, any organic solvent capable of extracting the first product can be used, and from the aspects of environmental protection and cost reduction, the organic solvent is preferably a mixed solvent formed by any one or more of dichloromethane, tetrahydrofuran and 2-methyltetrahydrofuran. The way of concentrating the first product extract phase described above is not limited, and includes, but is not limited to, evaporation, membrane concentration, and the like.
In the step S2, in order to further improve the R1 removal reaction efficiency, it is preferable to mix the first product with a hydrogen chloride solution in a second solvent to perform the R1 removal reaction. The above hydrogen chloride solution includes, but is not limited to, at least one of a hydrogen chloride methanol solution, a hydrogen chloride dioxane solution, a hydrogen chloride ethanol solution, or an isopropyl hydrogen acetate solution; from the standpoint of environmental protection and cost reduction, it is preferable that the second solvent is a mixed solvent formed of any one or more of methanol, ethanol, dioxane or isopropyl acetate.
In order to further promote the removal of R1 more completely and improve the efficiency of the R1 removal reaction, the temperature of the R1 removal reaction is preferably 0-10 ℃, and the time of the R1 removal reaction is preferably 2.0-8.0 h.
Typically, but not by way of limitation, the temperature of the R1 removal reaction is, for example, 0 ℃, 5 ℃, 8 ℃, 10 ℃ or any two values, and the time of the R1 removal reaction is, for example, 2 hours, 4 hours, 6 hours, 8 hours or any two values.
The above-mentioned method for purifying the second product system is not limited, and any purification method capable of separating the second product from the second product system may be used. In order to further increase the yield of the second product system, it is preferred that the purification of the second product system comprises solid-liquid separation, extraction and concentration, which are performed sequentially. The solid-liquid separation mode is not limited, and includes, but is not limited to, filtration, centrifugation, etc., and the organic solvent used for the extraction is not limited, and any organic solvent capable of extracting the second product can be used, including, but not limited to, a mixed solvent formed by any one or more of methanol, ethanol, dioxane or isopropyl acetate; the manner of concentration is not limited and includes, but is not limited to, evaporation and the like.
In the above step S3, in order to further enhance the efficiency of the condensation coupling reaction, it is preferable that the second product is subjected to the above condensation coupling reaction with the pyrrolidinylcarboxylic acid compound under the action of a condensing agent, the type of the above condensing agent is not limited, and any agent capable of promoting the condensation coupling reaction of an amino group with a carboxyl group may be used, including, but not limited to, a mixed agent formed of any one or more of HOBT (1-hydroxybenzotriazole)/EDCI (1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride), HATU (2- (7-azabenzotriazole) -N, N '-tetramethylurea hexafluorophosphate), HOBT (1-hydroxybenzotriazole)/HBTU (benzotriazole-N, N' -tetramethylurea hexafluorophosphate) or T3P (1-propylphosphoric anhydride).
In order to further improve the yield and production efficiency of the third product, it is preferable that the second product and the pyrrolidinyl carboxylic acid compound are subjected to the condensation coupling reaction in a third solvent, and from the viewpoints of green chemistry and cost reduction, the third solvent is preferably a mixed solvent formed of any one or more of N, N-dimethylformamide, dichloromethane, tetrahydrofuran and 2-methyltetrahydrofuran.
In order to further promote the progress of the condensation coupling reaction more sufficiently and to further increase the yield of the third product, it is preferable that the temperature of the condensation coupling reaction is 15 to 25℃and the time of the condensation coupling reaction is 1.0 to 8.0 hours.
Typically, but not by way of limitation, the temperature of the condensation coupling reaction is, for example, 15 ℃, 18 ℃, 22 ℃, 25 ℃ or any two values, and the time of the condensation coupling reaction is, for example, 1h, 3h, 5h, 6h, 8h or any two values.
The above-mentioned method for purifying the third product system is not limited, and any purification method capable of separating the third product from the third product system may be used. To further increase the yield of the third product system, it is preferred that the purification of the third product system comprises quenching, crystallization, beating or recrystallization, which are performed sequentially. From the viewpoints of cost saving and environmental protection, it is preferable to dropwise add an acidic aqueous solution, an alkaline aqueous solution or an aqueous solution to the third product system for quenching treatment, wherein the acidic aqueous solution includes, but is not limited to, an aqueous solution of citric acid or an aqueous solution of hydrochloric acid; the above-mentioned alkaline aqueous solution includes, but is not limited to, an aqueous solution of sodium bicarbonate or an aqueous solution of sodium carbonate; the crystallization mode is not limited, and the quenched third product system is preferably subjected to crystallization. In order to further improve the purity of the third product, it is preferable to subject the crude third product obtained by the crystallization treatment to beating or recrystallization treatment.
In step S4, in order to further enhance the coupling reaction efficiency, it is preferable that the coupling reaction between the third product and the phenyl compound is performed in a fourth solvent, and from the viewpoints of green chemistry and cost reduction, the fourth solvent is preferably a mixed solvent of any one or more of tetrahydrofuran, 2-methyltetrahydrofuran, cyclopentyl methyl ether and diethyl ether.
In order to further promote the progress of the coupling reaction more completely and further improve the efficiency of the coupling reaction, it is preferable that the temperature of the coupling reaction is 15 to 25℃and the time of the coupling reaction is 4 to 24 h.
Typically, but not by way of limitation, the temperature of the coupling reaction is, for example, 15 ℃, 18 ℃, 22 ℃, 25 ℃ or any two values, and the time of the coupling reaction is, for example, 4 hours, 10 hours, 12 hours, 16 hours, 20 hours, 24 h or any two values.
The above-mentioned method for purifying the fourth product system is not limited, and any purification method capable of separating the fourth product from the fourth product system may be used. To further increase the yield of the fourth product system, it is preferred that the purification of the fourth product system comprises quenching, extraction, concentration, crystallization and beating, which are performed sequentially. From the viewpoints of cost saving and environmental protection, the fourth product system is preferably added with an ammonium chloride solution dropwise for quenching treatment, and the mass concentration of the ammonium chloride solution is preferably 2-4% to further improve the quenching efficiency. The extraction is performed by using an organic solvent, the type of the organic solvent used for extraction is not limited, and any organic solvent capable of extracting the fourth product can be used, including but not limited to a mixed solvent formed by any one or more of tetrahydrofuran, 2-methyltetrahydrofuran, cyclopentyl methyl ether or diethyl ether; the manner of concentration is not limited and includes, but is not limited to, evaporation and the like. The crystallization mode is not limited, and preferably, the concentrated fourth product system is dropwise added into a small polar solvent for crystallization to obtain solid crystals. The small polar solvent is, for example, a mixed polar solvent formed by any one or more of methyl tertiary butyl ether, n-heptane, n-hexane and isopropyl ether. In order to further improve the purity of the fourth product, it is preferable to obtain the fourth product by subjecting the solid crystals obtained by the crystallization to a pulping treatment.
In step S5, in order to further increase the efficiency of the selective reduction reaction, it is preferable that the fourth product is selectively reduced by the reducing agent. In order to further promote the selective reduction reaction more efficiently, the reducing agent is preferably selected from the group consisting of aluminum isopropoxide, tributyl lithium hydride and lithium aluminum hydride.
In order to further promote the selective reduction of the fourth product by the reducing agent in step S5, the fourth product is preferably dissolved in a fifth solvent, and then the reducing agent is added to the fifth solvent to perform the selective reduction. The type of the fifth solvent is not limited, and any solvent can be used to dissolve the fourth product. From the viewpoint of green chemistry and cost reduction, the fifth solvent is preferably toluene, an ether solvent or an alcohol solvent. The above ether solvents include, but are not limited to, mixed solvents of any several or more of methyl tertiary butyl ether, isopropyl ether, or diethyl ether; the above alcohol solvents include, but are not limited to, mixed solvents formed of any several or more of isopropanol, methanol, ethanol or n-butanol.
In order to further promote the selective reduction reaction to be more complete and further improve the preparation efficiency of the MMAE key intermediate, the temperature of the selective reduction is preferably 40-55 ℃, and the time of the selective reduction is preferably 1-8 hours.
Typically, but not by way of limitation, the temperature of the selective reduction is, for example, 40, 45, 50 or 55℃and the time of the coupling reaction is, for example, 2, 4, 6 or 8h.
The above-mentioned method for purifying the fifth product system is not limited, and any purification method capable of separating the fifth product from the fifth product system may be used. To further increase the yield of the fifth product system, it is preferred that the purification of the fifth product system comprises quenching, extraction, quenching, concentration, crystallization and beating, which are performed sequentially. From the viewpoints of cost saving and environmental protection, the quenching treatment is preferably performed by dropwise adding an ammonium chloride solution into the fifth product system, and the mass concentration of the ammonium chloride solution is preferably 2-4% so as to further improve the quenching efficiency. The extraction is performed by using an organic solvent, the type of the organic solvent used for extraction is not limited, and any organic solvent capable of extracting the fifth product can be used, including but not limited to a mixed solvent formed by any one or more of ethyl acetate, isopropyl acetate, dichloromethane and 2-methyltetrahydrofuran; the manner of the above-mentioned quenching is not limited, and it is preferable to conduct the quenching with saturated brine. The manner of concentration is not limited and includes, but is not limited to, evaporation and the like. The crystallization mode is not limited, and preferably, the concentrated fifth product system is dropwise added into a small polar solvent for crystallization to obtain solid crystals. And a mixed solvent formed by any one or more of the small polar solvents such as methyl tertiary butyl ether, n-heptane and n-hexane. In order to further improve the purity of the MMAE key intermediate, it is preferable to perform pulping treatment on the solid crystal obtained by crystallization to obtain the MMAE key intermediate.
In some embodiments of the present application, the process schematic of the preparation method of the MMAE key intermediate is as follows:
Figure SMS_29
Figure SMS_30
Figure SMS_31
Figure SMS_32
Figure SMS_33
the Me represents methyl, and the R and R 1 The same meaning as in any of the above preparation methods is used, wherein Me represents methyl, and step represents "step".
In another exemplary embodiment of the present application, there is also provided a method of preparing MMAE, the method comprising a method of preparing MMAE key intermediate, the method of preparing MMA key intermediate being any one of the methods of preparation provided in the first exemplary embodiment above.
By applying the technical scheme of the invention, the MMAE key intermediate is prepared by adopting the preparation method of the MMAE key intermediate, and then the MMAE is prepared by utilizing the MMAE key intermediate, so that the use of norephedrine as a starting material is avoided, a novel synthetic method is provided for the MMAE, and the mass production is more facilitated.
In a third exemplary embodiment of the present application, there is also provided an antibody-conjugated drug, the starting materials of which include an antibody and an MMAE-critical intermediate prepared by any one of the preparation methods provided in the first exemplary embodiment described above.
By applying the technical scheme of the invention, the antibody coupling drug provided by the application adopts the MMAE key intermediate and the antibody obtained by any one of the preparation methods provided by the first exemplary embodiment of the application as raw materials, thereby avoiding the use of norephedrine as an initial raw material, providing a novel synthesis method for the antibody coupling drug and having wide application prospect.
The advantageous effects of the present application will be further described below with reference to examples.
In the examples below, vol refers to the volume ratio, i.e., the ratio of the volume of the solvent added to the mass of the reference material, eq refers to the equivalent ratio, i.e., the ratio of the molar ratio of the material added to the reference material to which 1.0eq of the main material was added per operation.
Example 1
The embodiment provides a preparation method of an MMAE key intermediate, wherein the MMAE key intermediate has a structure shown in the following formula (I); wherein R is tert-butoxycarbonyl (Boc), R a Methyl (Me).
Figure SMS_34
Formula (I);
the preparation method of the MMAE key intermediate comprises the following steps of (1) mixing 150g (0.79 mol,1.0 eq) of Boc-D-acrylic acid with a protecting group Boc on the amino group and 96.6g (0.97 mol,1.22 eq) of N, O-dimethylhydroxylamine hydrochloride in 1500mL (10 Vol) of methylene chloride solvent, then adding 387g (1.02 mol,1.29 eq) of HATU (2- (7-azabenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate), using 257.4g (2.54 eq) of triethylamine to adjust the pH value of the system to 10.5, carrying out condensation reaction on the Boc-D-acrylic acid and N, O-dimethylhydroxylamine hydrochloride at the temperature of 15-25 ℃ for 14h, and ending the reaction to obtain a first product system. Adding 1500mL of purified water into the first product system for quenching treatment, and separating to obtain an organic phase of a first product; the organic phase of the first product is heated for 5 hours at the temperature of T less than or equal to 45 ℃ to be concentrated, 750mL of ethyl acetate solvent is adopted to be concentrated until no solvent exists, then 750mL of ethyl acetate is added into the system, pulping is carried out for 2 hours at the temperature of 15-25 ℃, 157g of the first product is obtained through filtration, and the first product is white-like solid with the yield of 85.5%.
(2) Mixing the first product with 628mL (4 Vol) of 4M (mol/L) 1, 4-dioxane solution of hydrogen chloride in 314mL (2 Vol) 1, 4-dioxane solvent, and carrying out R of the first product at 15-25 DEG C 1 And (3) carrying out radical removal reaction, wherein the reaction lasts for 6 hours, and a second product system is obtained after the reaction is finished. Concentrating the second product system at the temperature of T less than or equal to 40 ℃, and carrying out four times of concentration treatment by using 235mL of n-heptane to obtain a second product, wherein the second product is a light yellow oily matter, and the yield is 95.0%.
(3) 115.9g (0.69 mol,1.1 eq) of the second product and 180g (0.63 mol,1.0 eq) of a pyrrolidinylcarboxylic acid compound with a Boc protecting group are mixed in 1440mL (8 Vol) of dichloromethane solvent, then 325.1g (2.5 mol,4 eq) of N, N-diisopropylethylamine, 127g (0.94 mol,1.5 eq) of HOBT and 180.2g (0.94 mol,1.5 eq) of EDCI condensing reagent are added to the solvent at a temperature of 0-10 ℃ for 2h, and a condensation coupling reaction is carried out at 15-25 ℃ to obtain a third product system after the reaction is finished. And (3) dropwise adding 1800mL of citric acid aqueous solution with the mass concentration of 10% into the third product system for quenching treatment, separating the obtained quenching system of the third product to obtain an organic phase, sequentially using 1800mL of purified water and 1800mL of 10% sodium bicarbonate aqueous solution for extraction and washing, concentrating the organic phase at the temperature of less than or equal to 40 ℃ to obtain a third product crude product, adding 900mL of methyl tertiary butyl ether reagent into the third product crude product, pulping the third product crude product, and filtering to obtain 230g of the third product with the yield of 91.5%.
(4) 230g (0.57 mol,1.0 eq) of the third product is dissolved in 2760mL (12 vol) of tetrahydrofuran solvent, the system is cooled to 10 ℃, 1140mL (1.14 mol,2.0 eq) of 1M phenylmagnesium bromide tetrahydrofuran solution is added dropwise into the system to be mixed with the third product for coupling reaction, cooling liquid is removed after the addition, the reaction lasts for 13h at 15-25 ℃ and a fourth product system is obtained after the reaction is finished. And (3) dropwise adding 1150mL of ammonium chloride solution with the mass concentration of 3.7% into the fourth product system for quenching treatment to obtain a quenching system of the fourth product. Extracting a quenching system of a fourth product by adopting 1150mL of ethyl acetate solvent, continuously extracting an aqueous phase once by adopting 1150mL of ethyl acetate, merging organic phases to obtain a fourth product extract phase, concentrating the fourth product extract phase to 460mL, then dropwise adding the fourth product extract phase into 4600mL of n-heptane solvent for crystallization treatment, crystallizing and separating out a crude product of the fourth product, adding 2300mL of n-heptane into the crude product of the fourth product for pulping treatment to obtain 225g of the fourth product which is a white solid, and obtaining the yield of 93.7%.
(5) 225g (0.54 mol,1.0 eq) of the fourth product was dissolved in a mixed solvent of 810mL (3.6 Vol) toluene and 450mL (2 Vol) isopropanol, and then 198.5. 198.5 g aluminum isopropoxide (0.97 mol,1.8 eq) was added to the system three times, and the reaction was continued at 40-55℃for 5 hours, and the reaction was completed to obtain a fifth product system. And (3) dropwise adding 1125mL of ammonium chloride solution with the mass concentration of 3.7% into a fifth product system for quenching treatment, separating out solids in the system, filtering, extracting filtrate once by using 1125mL of ethyl acetate, extracting water phase once by using 1125mL of ethyl acetate after liquid separation, extracting and washing organic phases once by using 1125mL of saturated sodium chloride solution after merging, and then decompressing and desolventizing at the temperature T being less than or equal to 40 ℃ and concentrating to 2 Vol. And (3) obtaining a concentrated system of a fifth product, dropwise adding the concentrated system of the fifth product into a mixed solvent formed by 1125mL of methyl tertiary butyl ether and 1575mL of n-heptane to perform crystallization treatment, crystallizing and separating out a crude product of the fifth product, and adding 675mL of n-heptane into the crude product of the fifth product to perform pulping treatment to obtain 214.8g of the MMAE key intermediate with the yield of 95%.
The preparation process of the MMAE intermediate is schematically shown as follows:
Figure SMS_35
Figure SMS_36
Figure SMS_37
Figure SMS_38
Figure SMS_39
in the above process scheme, R 1 And R are each independently a Boc protecting group, me is methyl.
Example 2
This example differs from example 1 in that the MMAE key intermediate has the structure shown in formula (i) below; wherein R is fluorenylmethoxycarbonyl (Fmoc), R a Methyl (Me).
Figure SMS_40
Formula (I);
the preparation process differs from example 1 in that in step (3) the protecting group of the pyrrolidinyl carboxylic acid compound used is Fmoc and the amount of the pyrrolidinyl carboxylic acid compound used is 0.63mol.
Example 3
This example differs from example 1 in that the MMAE key intermediate has the structure shown in formula (i) below; wherein R is carbobenzoxy (Cbz), R a Methyl (Me).
Figure SMS_41
Formula (I);
the preparation process differs from example 1 in that in step (3) the protecting group of the pyrrolidinyl carboxylic acid compound used is Cbz and the amount of the pyrrolidinyl carboxylic acid compound used is 0.63mol.
Example 4
This example differs from example 1 in that the MMAE key intermediate has the structure shown in formula (i) below; wherein R is benzoyl (Bz), R a Methyl (Me).
Figure SMS_42
Formula (I);
the preparation process differs from example 1 in that in step (3) the protecting group of the pyrrolidinyl carboxylic acid compound used is Bz and the amount of the pyrrolidinyl carboxylic acid compound used is 0.63mol.
Example 5
This example differs from example 1 in that in step (4), phenylmagnesium bromide is replaced with phenyllithium as the formative reagent, and the amount of the substance of phenyllithium is the same as phenylmagnesium bromide.
Example 6
This example differs from example 1 in that in step (5), aluminum isopropoxide is replaced with tributyl lithium hydride, and the amount of the substance of tributyl lithium hydride is the same as that of aluminum isopropoxide.
Example 7
This example differs from example 1 in that in step (1), N, O-dimethylhydroxylamine nitrate was used instead of N, O-dimethylhydroxylamine hydrochloride, and the amount of the substance of N, O-dimethylhydroxylamine nitrate was the same as that of N, O-dimethylhydroxylamine hydrochloride.
Example 8
This example differs from example 1 in that in step (1), N, O-dimethylhydroxylamine sulfate was used instead of N, O-dimethylhydroxylamine hydrochloride, and the amount of the substance of N, O-dimethylhydroxylamine sulfate was the same as that of N, O-dimethylhydroxylamine hydrochloride.
Example 9
The present example provides a kg-grade preparation method of the MMAE key intermediate provided in example 1 above, which is performed according to the following steps:
(1) Boc-D-acrylic acid with 3.00 kg (15.8 mol,1.0 eq) amino group as Boc and 3.20 kg (19.0 mol,1.2 eq) N, O-dimethylhydroxylamine hydrochloride are mixed in 30.0L (10 Vol) methylene chloride solvent, HATU 7.81 Kg (20.5 mol,1.3 eq) is added, the pH value of the system is adjusted to 10.4 by using 5.11kg (50.6 mol,3.2 eq) triethylamine, condensation reaction is carried out at 15-25 ℃ for 12h, and the reaction is ended to obtain a first product system. Adding 30.0L of purified water into the first product system for quenching treatment, and separating to obtain an organic phase of a first product; the organic phase is heated for 5 hours at the temperature of less than or equal to 45 ℃ for concentration, 15.0L of ethyl acetate solvent is adopted for shrinking until the solvent is basically absent, then 15.0L of ethyl acetate is added into the system for pulping for 2 hours at the temperature of 15-25 ℃, the first product 3.28 and kg is obtained after filtration, and the first product is off-white solid with the yield of 89.4 percent.
(2) Mixing 3.28kg of the first product with 13.1L (4 Vol) of 4M hydrogen chloride 1, 4-dioxane solution in 6.6L (2 Vol) of 1, 4-dioxane solvent, and carrying out R of the first product at 15-25 DEG C 1 And (3) carrying out radical removal reaction, wherein the reaction lasts for 7 hours, and a second product system is obtained after the reaction is finished. Concentrating the second product system at a temperature of less than or equal to 40 ℃, and concentrating 4 times by using 4.9L of n-heptane to obtain a second product, wherein the second product is a light yellow oily matter, and the yield is 99.0%.
(3) 2.38Kg (14.1 mol,1.1 eq) of the second product and 3.68Kg (12.8 mol,1.0 eq) of a pyrrolidinylcarboxylic acid compound having a protecting group Boc were mixed in 29.5L (8 Vol) of methylene chloride solvent, then 6.60Kg (51.2 mol,4 eq) of N, N-diisopropylethylamine, 2.59Kg (19.2 mol,1.5 eq) of HOBT,3.68Kg (19.2 mol,1.5 eq) of EDCI condensing reagent were added to the solvent at a controlled temperature of 0 to 10℃for 2 hours to obtain a third product system. Dropwise adding 36.8L of 10% citric acid aqueous solution in mass concentration into a third product system for quenching treatment to obtain a quenching system of the third product, separating the quenching system to obtain an organic phase, sequentially using 1800ml of purified water and 36.8ml of 10% sodium bicarbonate aqueous solution for extraction and washing, then concentrating the organic phase at the temperature of less than or equal to 40 ℃ to obtain a third product crude product, adding 18.4L of methyl tertiary butyl ether reagent into the third product crude product, pulping the third product crude product, filtering to obtain 4.75kg of the third product, and obtaining 92.4% of yield.
(4) And (3) dissolving 4.75kg (11.8 mol,1.0 eq) of a third product in 57.0L (12 vol) tetrahydrofuran solvent, cooling the system to 10 ℃, dropwise adding 23.6L (mol/L) of 1M (mol/L) phenylmagnesium bromide tetrahydrofuran solution (23.6 mol,2.0 eq) into the system, mixing with the third product for coupling reaction, removing cooling liquid after the addition, continuing the reaction at 15-25 ℃ for 13h, and obtaining a fourth product system after the reaction is finished. And (3) dropwise adding 23.8L of ammonium chloride solution with the mass concentration of 3.7% into the fourth product system for quenching treatment to obtain a quenching system of the fourth product. Extracting a quenching system of a fourth product by using 23.8L of ethyl acetate solvent, continuously extracting a water phase by using 23.8L of ethyl acetate for one time, merging organic phases to obtain a fourth product extract phase, concentrating the fourth product extract phase to 9.5L, then dropwise adding the fourth product extract phase into 95L of n-heptane solvent for crystallization treatment, crystallizing and separating out a fourth product crude product, adding 47.5L of n-heptane into the fourth product crude product for pulping treatment to obtain a fourth product of 4.67kg, wherein the fourth product is a white solid product, and the yield is 94.4%.
(5) The fourth product 4.67Kg (11.2 mol,1.0 eq) was dissolved in 16.8L (3.6 Vol) toluene and 9.3L (2 Vol) isopropanol, then 4.12Kg aluminum isopropoxide (20.2 mol,1.8 eq) was added to the system in five portions, and the reaction was continued at 40-55℃for 6 hours, and the reaction was completed to obtain a fifth product system. And (3) dropwise adding 23.5L of ammonium chloride solution with the mass concentration of 3.7% into a fifth product system for quenching treatment, separating out solids in the system, filtering, extracting filtrate with 23.5L of ethyl acetate once, separating liquid, extracting water phase with 23.5L of ethyl acetate once, merging organic phases, extracting and washing the organic phases with 23.5L of saturated sodium chloride solution once, then carrying out reduced pressure desolventizing at the temperature T of less than or equal to 40 ℃, concentrating to 10L to obtain a concentrated system of a fifth product, dropwise adding the concentrated system of the fifth product into a mixed solvent formed by 23.5L of methyl tertiary butyl ether and 33.0L of n-heptane for crystallization treatment, crystallizing and separating out crude products of the fifth product, adding 14.0L of n-heptane into the crude products of the fifth product for pulping treatment, thus obtaining 4.48kg of the key intermediate of the MMAE, and the yield of 95.5%.
Test example 1
The nuclear magnetic resonance test and the high performance liquid phase test are performed on the MMAE key intermediate prepared in the embodiment 9, and the results are shown in fig. 1 and 2, and it can be seen from fig. 1 that the MMAE key intermediate prepared in the embodiment 9 is consistent with the expected structure of the molecule through nuclear magnetic resonance identification.
As can be seen from fig. 2, the purity of the MMAE key intermediate prepared in example 9 is 99.43%, and the maximum unknown single impurity rrt=1.04 is 0.26%, so that the requirement of downstream production of the intermediate can be met, and the requirement meets expectations.
Test example 2
The purity and yield of the first, second, third, fourth and MMAE key intermediates prepared in examples 1 to 9 were measured, respectively, and the results are shown in table 1 below.
Wherein the purity is measured by high performance liquid chromatography.
The calculation method of the yield comprises the following steps:
theoretical yield per step was calculated: theoretical yield = (starting material charge/starting material molecular weight) ×intermediate molecular weight
Actual yield calculation method per step, yield= (crude yield/theoretical yield) ×100%
TABLE 1
Figure SMS_43
From the above description, it can be seen that the above embodiments of the present invention achieve the following technical effects:
the method adopts cheap and easily available commercial raw materials as the initial raw materials to synthesize the MMAE key intermediate, avoids the direct participation of the norephedrine in the reaction, provides a novel synthetic method for the MMAE, and solves the problem that the synthetic method of the MMAE in the prior art needs to adopt the easily-toxic compound of the norephedrine as the initial raw material, so that the method cannot be widely applied.
The above description is only of the preferred embodiments of the present invention and is not intended to limit the present invention, but various modifications and variations can be made to the present invention by those skilled in the art. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (10)

1. The preparation method of the MMAE key intermediate is characterized in that the MMAE key intermediate has a structure shown in the following formula (I);
Figure QLYQS_1
formula (I);
the preparation method comprises the following steps:
step S1, carrying out condensation reaction on a D-alanine compound and N, O-dialkylhydroxylamine salt in an alkaline environment to obtain a first product system, and purifying the first product system to obtain a first product; the D-alanine compound has a structure shown in the following formula (II), the N, O-dialkylhydroxylamine salt has a structure shown in the following formula (III), and the first product has a structure shown in the following formula (IV);
Figure QLYQS_2
formula (II); />
Figure QLYQS_3
Formula (III); />
Figure QLYQS_4
Formula (IV);
step S2, R on the first product 1 Removing to obtain a second product system, and purifying the second product system to obtain a second product; the second product has a structure shown in the following formula (V);
Figure QLYQS_5
Formula (V);
step S3, performing condensation coupling reaction on the second product and a pyrrolidinyl carboxylic acid compound to obtain a third product system, and purifying the third product system to obtain a third product; the pyrrolidinyl carboxylic acid compound has the structure shown in the following formula (VI), and the third product has the structure shown in the following formula
Figure QLYQS_6
) The structure is shown;
Figure QLYQS_7
formula (VI); />
Figure QLYQS_8
(-)>
Figure QLYQS_9
);
Step S4, performing a coupling reaction on the third product and a phenyl compound to obtain a fourth product system, and purifying the fourth product system to obtain a fourth product; the phenyl compound has the following formula
Figure QLYQS_10
) The structure shown, the fourth product has the formula (/ ->
Figure QLYQS_11
) The structure is shown;
Figure QLYQS_12
(-)>
Figure QLYQS_13
);/>
Figure QLYQS_14
(-)>
Figure QLYQS_15
);
S5, selectively reducing the fourth product to obtain a fifth product system, and purifying the fifth product system to obtain the MMAE key intermediate;
wherein R and R 1 Each independently represents a protecting group, saidThe protecting group comprises at least one of an alkoxycarbonyl group, an acyl group or an alkyl group; z represents hydrochloric acid, nitric acid or sulfuric acid; r is R a Is methyl; r is R 2 And R is 3 Each independently represents a C1 to C6 alkyl group; m represents a metal or a halogenated metal.
2. The process of claim 1, wherein R and R are 1 Each independently represents benzyloxycarbonyl, t-butyloxycarbonyl, fluorenylmethoxycarbonyl, allyloxycarbonyl, trimethylsilylethoxycarbonyl, benzoyl, phthaloyl, p-toluenesulfonyl, trifluoroacetyl, trityl, 2, 4-dimethoxybenzyl, p-methoxybenzyl or benzyl;
and/or R 2 And R is 3 Each independently represents a C1 to C4 alkyl group;
and/or M represents lithium, sodium, potassium, magnesium bromide, magnesium chloride or magnesium iodide.
3. The process of claim 1, wherein R and R are 1 Each independently represents a benzyloxycarbonyl group, a t-butyloxycarbonyl group, a fluorenylmethoxycarbonyl group or a benzoyl group;
and/or, Z represents hydrochloric acid;
and/or R 2 And R is 3 Each independently represents methyl;
and/or M represents lithium or magnesium bromide.
4. The preparation method according to claim 1, wherein in the step S1, the pH value of the alkaline environment is 8.0 to 11.0;
and/or the condensation reaction is carried out in an alkaline environment formed by a first solvent, wherein the first solvent comprises at least one of dichloromethane, tetrahydrofuran, 2-methyltetrahydrofuran and N, N-dimethylformamide;
and/or the temperature of the condensation reaction is 0-10 ℃, and the time of the condensation reaction is 4.0-24.0 h;
And/or, the purification of the first product system comprises quenching, extraction and concentration performed sequentially.
5. The method according to any one of claims 1 to 4, wherein the step S2 is performed by mixing the first product with a hydrogen chloride solution in a second solvent to perform the R 1 Removing reaction;
the second solvent comprises at least one of methanol, ethanol, dioxane or isopropyl acetate;
and/or, the R 1 The temperature of the removal reaction is 0-10 ℃ and the time is 2.0-8.0 h;
and/or, the purification of the second product system comprises solid-liquid separation, extraction and concentration which are sequentially carried out.
6. The process according to any one of claims 1 to 4, wherein in step S3, the second product is subjected to the condensation coupling reaction with the pyrrolidinylcarboxylic acid compound under the action of a condensing agent comprising HOBT/EDCI, HATU, HOBT/HBTU or T 3 At least one of P;
and/or, the second product and the pyrrolidinyl carboxylic acid compound are subjected to the condensation coupling reaction in a third solvent, wherein the third solvent comprises at least one of N, N-dimethylformamide, dichloromethane, tetrahydrofuran and 2-methyltetrahydrofuran;
And/or the temperature of the condensation coupling reaction is 15-25 ℃ and the time is 1.0-8.0 h;
and/or the purification of the third product system comprises quenching, crystallization, recrystallization or beating, which are performed sequentially.
7. The preparation method according to any one of claims 1 to 4, wherein the step S4, the coupling reaction of the third product and the phenyl-based compound is performed in a fourth solvent, the fourth solvent comprising at least one of tetrahydrofuran, 2-methyltetrahydrofuran, cyclopentyl methyl ether, or diethyl ether;
and/or the temperature of the coupling reaction is 15-25 ℃ and the time is 4-24 hours;
and/or, the purification of the fourth product system comprises quenching, extraction, concentration, crystallization and beating which are sequentially carried out.
8. The method according to any one of claims 1 to 4, wherein the fourth product is subjected to the selective reduction by a reducing agent comprising at least one of aluminum isopropoxide, lithium tributylhydride or lithium aluminum tetrahydroide in step S5;
and/or, the fourth product and the reducing agent are subjected to the selective reduction in a fifth solvent comprising at least one of toluene, an ether solvent, or an alcohol solvent;
And/or the temperature of the selective reduction is 40-55 ℃ and the time is 1.0-8.0 h;
and/or, the purification of the fifth product system comprises quenching, extraction, quenching, concentration, crystallization and beating which are sequentially carried out.
9. A process for the preparation of MMAE, characterized in that the process comprises a process for the preparation of MMAE key intermediates, which process is according to any one of claims 1 to 8.
10. An antibody-conjugated drug, characterized in that the raw materials of the antibody-conjugated drug comprise an antibody and the MMAE key intermediate obtained by the preparation method of any one of claims 1 to 8.
CN202310494927.4A 2023-05-05 2023-05-05 Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug Active CN116239513B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310494927.4A CN116239513B (en) 2023-05-05 2023-05-05 Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310494927.4A CN116239513B (en) 2023-05-05 2023-05-05 Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug

Publications (2)

Publication Number Publication Date
CN116239513A true CN116239513A (en) 2023-06-09
CN116239513B CN116239513B (en) 2023-08-18

Family

ID=86631619

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310494927.4A Active CN116239513B (en) 2023-05-05 2023-05-05 Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug

Country Status (1)

Country Link
CN (1) CN116239513B (en)

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5969100A (en) * 1995-01-06 1999-10-19 Sibia Neurosciences, Inc. Peptide, peptide analog and amino acid analog protease inhibitors
CA2378600A1 (en) * 1999-07-09 2001-01-18 The Regents Of The University Of Michigan Amino ceramide-like compounds and therapeutic methods of use
US20030083263A1 (en) * 2001-04-30 2003-05-01 Svetlana Doronina Pentapeptide compounds and uses related thereto
EP1338592A1 (en) * 2002-02-22 2003-08-27 Nippon Zoki Pharmaceutical Co., Ltd. Novel 2-phenylpiperazine derivatives
WO2004010957A2 (en) * 2002-07-31 2004-02-05 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
WO2012143496A2 (en) * 2011-04-21 2012-10-26 Bayer Intellectual Property Gmbh Novel binder-drug conjugates (adcs) and their use
CN103342663A (en) * 2013-07-15 2013-10-09 凯莱英医药集团(天津)股份有限公司 Method for preparing key anastrozole intermediate
CN103435525A (en) * 2013-07-25 2013-12-11 凯莱英医药集团(天津)股份有限公司 Preparation method of ezetimibe
CN103508948A (en) * 2013-10-17 2014-01-15 凯莱英医药集团(天津)股份有限公司 Method for preparing pitavastatin calcium
CN104254342A (en) * 2011-12-14 2014-12-31 西雅图基因公司 FGRF antibody drug conjugates (ADCs) and the use thereof
CN107375941A (en) * 2017-07-17 2017-11-24 中国药科大学 A kind of anti-human DLL4 monoclonal antibodies and aplysiatoxin derivative MMAE conjugate
WO2020020365A1 (en) * 2018-07-27 2020-01-30 中国医学科学院药物研究所 Method for preparing optically active eliglustat
CN113195487A (en) * 2018-10-12 2021-07-30 杭州多禧生物科技有限公司 2, 3-diaminosuccinyl conjugate linkers
WO2021183542A1 (en) * 2020-03-09 2021-09-16 Sigma-Aldrich Co., Llc Efficient preparation of dolastatin and auristatin analogs through a common intermediate
TW202142266A (en) * 2020-02-05 2021-11-16 中國大陸商杭州多禧生物科技有限公司 Conjugates of cell-binding molecules with cytotoxic agents
CN113698335A (en) * 2015-07-04 2021-11-26 杭州多禧生物科技有限公司 Specific conjugation of cell binding molecules
WO2021252640A1 (en) * 2020-06-09 2021-12-16 Senda Biosciences, Inc. Asparagine derivatives and methods of using same
CN113842467A (en) * 2020-06-28 2021-12-28 达石药业(广东)有限公司 Intermediate for preparing Antibody and Drug Conjugate (ADC) and preparation method and application thereof
CN114040781A (en) * 2019-06-24 2022-02-11 杭州多禧生物科技有限公司 Formulation of conjugate of TUBULYSIN derivative and cell binding molecule
WO2022206870A1 (en) * 2021-03-31 2022-10-06 Remegen Co., Ltd. Preparation and purification process of monomethyl auristain e compound

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5969100A (en) * 1995-01-06 1999-10-19 Sibia Neurosciences, Inc. Peptide, peptide analog and amino acid analog protease inhibitors
CA2378600A1 (en) * 1999-07-09 2001-01-18 The Regents Of The University Of Michigan Amino ceramide-like compounds and therapeutic methods of use
US20030083263A1 (en) * 2001-04-30 2003-05-01 Svetlana Doronina Pentapeptide compounds and uses related thereto
EP1338592A1 (en) * 2002-02-22 2003-08-27 Nippon Zoki Pharmaceutical Co., Ltd. Novel 2-phenylpiperazine derivatives
US20030166616A1 (en) * 2002-02-22 2003-09-04 Takashi Ogino Novel 2-phenylpiperazine derivatives
WO2004010957A2 (en) * 2002-07-31 2004-02-05 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
CN106117312A (en) * 2011-04-21 2016-11-16 西雅图基因公司 New bonding agent drug conjugate (ADC) and application thereof
WO2012143496A2 (en) * 2011-04-21 2012-10-26 Bayer Intellectual Property Gmbh Novel binder-drug conjugates (adcs) and their use
WO2012143497A2 (en) * 2011-04-21 2012-10-26 Bayer Intellectual Property Gmbh Novel binder-drug conjugates (adcs) and their use
CN104254342A (en) * 2011-12-14 2014-12-31 西雅图基因公司 FGRF antibody drug conjugates (ADCs) and the use thereof
CN103342663A (en) * 2013-07-15 2013-10-09 凯莱英医药集团(天津)股份有限公司 Method for preparing key anastrozole intermediate
CN103435525A (en) * 2013-07-25 2013-12-11 凯莱英医药集团(天津)股份有限公司 Preparation method of ezetimibe
CN103508948A (en) * 2013-10-17 2014-01-15 凯莱英医药集团(天津)股份有限公司 Method for preparing pitavastatin calcium
CN113698335A (en) * 2015-07-04 2021-11-26 杭州多禧生物科技有限公司 Specific conjugation of cell binding molecules
CN107375941A (en) * 2017-07-17 2017-11-24 中国药科大学 A kind of anti-human DLL4 monoclonal antibodies and aplysiatoxin derivative MMAE conjugate
WO2020020365A1 (en) * 2018-07-27 2020-01-30 中国医学科学院药物研究所 Method for preparing optically active eliglustat
CN113195487A (en) * 2018-10-12 2021-07-30 杭州多禧生物科技有限公司 2, 3-diaminosuccinyl conjugate linkers
CN114040781A (en) * 2019-06-24 2022-02-11 杭州多禧生物科技有限公司 Formulation of conjugate of TUBULYSIN derivative and cell binding molecule
TW202142266A (en) * 2020-02-05 2021-11-16 中國大陸商杭州多禧生物科技有限公司 Conjugates of cell-binding molecules with cytotoxic agents
WO2021183542A1 (en) * 2020-03-09 2021-09-16 Sigma-Aldrich Co., Llc Efficient preparation of dolastatin and auristatin analogs through a common intermediate
WO2021252640A1 (en) * 2020-06-09 2021-12-16 Senda Biosciences, Inc. Asparagine derivatives and methods of using same
CN113842467A (en) * 2020-06-28 2021-12-28 达石药业(广东)有限公司 Intermediate for preparing Antibody and Drug Conjugate (ADC) and preparation method and application thereof
WO2022206870A1 (en) * 2021-03-31 2022-10-06 Remegen Co., Ltd. Preparation and purification process of monomethyl auristain e compound

Also Published As

Publication number Publication date
CN116239513B (en) 2023-08-18

Similar Documents

Publication Publication Date Title
CN103333942B (en) A synthetic method for (R)-praziquantel
CN112125805B (en) Water-soluble magnolol derivative, preparation method of honokiol derivative and intermediate thereof, and related monohydroxy protected intermediate
GB1592552A (en) Pseudopeptides used as medicaments
CN111187269A (en) Synthetic method of Reidesciclovir intermediate
SE447389B (en) NEW TRIPEPTIDES AFFECTING THE CENTRAL NERVOUS SYSTEM
Tanaka et al. Studies on 2-aziridinecarboxylic acid. IV. Total synthesis of actinomycin D (C1) via ring-opening reaction of aziridine.
CN116239513B (en) Preparation method of MMAE key intermediate, preparation method of MMAE and antibody coupling drug
JP2002507624A (en) Peptide turn mimic structure
CN112876502B (en) Preparation method of N-trimethylsiloxyethoxycarbonyl-N-methyl-L/D-leucine
Meng et al. Synthetic approaches toward glidobamine, the core structure of the glidobactin antibiotics
CN100343275C (en) Process for preparing glycyl gtutamine
WO2022017317A1 (en) Method for large-scale synthesis of tetrodotoxin
CN113072514A (en) Preparation method of cycleanine and intermediate thereof
CN112375055A (en) Preparation method of eptifibatide key raw material L-high arginine
Martin et al. A SAFE AND CONVENIENT PROCEDURE FOR THE SYNTHESIS OF POLYAMBSES via AZIDE INTERMEDIATES
Nomoto et al. First synthesis of blastidic acid, a component amino acid in an antibiotic, blasticidin S
CN110684028A (en) Preparation method of 2, 6-diazabicyclo [3, 3, 0] octane compound
CN115010639B (en) Intermediate compound and preparation method and application thereof
CN105503698A (en) Method for synthesizing Saxagliptin and intermediate
KINGSBURY et al. Synthesis of α‐thiophenylglycine peptides: Novel peptide substrates useful in the study of microbial peptide transport
CN109503430B (en) O-fluoro-o-imine benzoic acid intermediate compound and preparation method and application thereof
Shin et al. Synthetic studies on quinoxaline antibiotics. Synthesis of quinoxaline-2-carbonyl tetradepsipeptide derivatives related to triostin A.
Deeks et al. Leucine enkephalin analogues containing a conformationally restrained N-terminal amino acid residue
WO2013159283A1 (en) Method for preparing (s)-oxiracetam
CN113636980B (en) Preparation method of dexrazoxane

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