CN112851648A - Application of brefeldin A ester derivatives in antitumor drugs - Google Patents

Application of brefeldin A ester derivatives in antitumor drugs Download PDF

Info

Publication number
CN112851648A
CN112851648A CN202011327932.9A CN202011327932A CN112851648A CN 112851648 A CN112851648 A CN 112851648A CN 202011327932 A CN202011327932 A CN 202011327932A CN 112851648 A CN112851648 A CN 112851648A
Authority
CN
China
Prior art keywords
cancer
compound
compounds
pharmaceutically acceptable
ring
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
CN202011327932.9A
Other languages
Chinese (zh)
Other versions
CN112851648B (en
Inventor
邵长伦
王长云
魏美燕
姜瑶瑶
刘明
王翠芳
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.)
Ocean University of China
Original Assignee
Ocean University of China
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 Ocean University of China filed Critical Ocean University of China
Publication of CN112851648A publication Critical patent/CN112851648A/en
Application granted granted Critical
Publication of CN112851648B publication Critical patent/CN112851648B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to the field of medicinal chemistry, in particular to application of a brefeldin A ester derivative in inhibiting tumor proliferation activity and preventing or treating hyperproliferative diseases, wherein the structure of the brefeldin A ester derivative is shown as a formula (I). The hyperproliferative diseases comprise liver cancer, leukemia, breast cancer, colon adenocarcinoma, gastric cancer, lung cancer, Bart's esophagus cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymph cancer, kidney cancer, brain cancer, nerve cancer, nasopharyngeal cancer, oral cancer and colorectal cancer, so the brefeldin A ester derivatives have the potential of being developed into novel antitumor drugs.

Description

Application of brefeldin A ester derivatives in antitumor drugs
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a brefeldin A ester derivative, a composition and application thereof, wherein the compound or the composition has the application of inhibiting tumor proliferation activity and can be used for preventing or treating hyperproliferative diseases.
Background
Brefeldin A (British name: Brefeldin A, abbreviated as BFA) is a class of macrolide fungal metabolites. Isolated from Penicillium decembens by Singleton et al in 1958 (Singleton, V.L. et, al. Nature, 1958, 181, 1072-. BFA has a wide range of biological activities including antiviral, antifungal, nematicidal, antimitotic and antitumor etc. (Tamura, G, et, al. J. Antibilt.1968, 21, 160-. The American tumor research institute finds that BFA can induce tumor cell differentiation and apoptosis, and has wide application prospect when being used as a chemotherapeutic agent for tumor treatment because BFA has wide biological activity and a unique macrolide structure.
BFA, however, is not ideal as a drug for clinical use due to its own pharmacokinetic properties (low bioavailability, poor water solubility, low plasma exposure, short plasma half-life, high toxicity) (Sausville, e.a.et al cancer. j. sci. Am. 1996, 2, 52-58). In order to overcome the defects, attention is paid to structural modification of BFA, and a compound which can keep high-activity antitumor effect and has good pharmacokinetic property is expected to be found to be used as a medicine for clinical application. However, many of the reported BFA derivatives have been reported to retain in vitro anti-tumor effects (CN 103788053A; CN 103772342A; CN 105153136A; CN 106928213A), and there are no reports of BFA derivatives that are effective in improving pharmacokinetic properties, as well as other properties such as increased solubility and reduced toxicity.
Disclosure of Invention
The following is a summary of some aspects of the invention only and is not intended to be limiting. These aspects and others are described more fully below. All references in this specification are incorporated herein by reference in their entirety. When the disclosure of the present specification differs from the cited documents, the disclosure of the present specification controls.
The invention provides a novel class of brefeldin A ester derivatives, which are used for preventing or treating hyperproliferative diseases, such as liver cancer, leukemia, breast cancer, colon adenocarcinoma, stomach cancer, lung cancer, Bart's esophagus cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymph cancer, kidney cancer, brain cancer, nerve cancer, nasopharyngeal cancer, oral cancer and colorectal cancer. The compound has stable property and good safety, maintains relatively high content (high blood plasma Cmax and blood plasma exposure) in a mammal body, can greatly improve the in-vivo efficacy of the medicament, and has good clinical application prospect.
The invention also provides pharmaceutical compositions comprising these compounds and methods of using these compounds or compositions in the treatment of the above-mentioned diseases in mammals, especially humans.
Specifically, the method comprises the following steps:
in one aspect, the invention relates to a compound of formula (I) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, ester, pharmaceutically acceptable salt or prodrug thereof of a compound of formula (I),
Figure 751217DEST_PATH_IMAGE001
(I);
wherein:
z is a substituted or unsubstituted pyridine ring, quinoline ring or isoquinoline ring, wherein the pyridine ring, quinoline ring and isoquinoline ring may each independently optionally be substituted with 1,2,3 or 4 substituents selected from deuterium, F, Cl, Br, I, hydroxy, amino, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C1-3Alkoxy radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, aryl, or 5-6 membered heteroaryl.
In some embodiments, Z is a substituted or unsubstituted pyridine, quinoline, or isoquinoline ring, wherein the pyridine, quinoline, and isoquinoline rings may each be independently optionally substituted with 1,2,3, or 4 substituents selected from deuterium, F, Cl, Br, I, hydroxy, amino, cyano, nitro, methyl, ethyl, n-propyl, isopropyl, difluoromethyl, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, phenyl, thiazolyl, pyrazolyl, oxazolyl, pyridinyl, or pyrimidinyl.
In another aspect, the invention relates to a pharmaceutical composition comprising a compound disclosed herein.
In some embodiments, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle, or combination thereof.
In another aspect, the present invention relates to the use of a compound or pharmaceutical composition disclosed herein for the prevention or treatment of hyperproliferative diseases in mammals, including humans; wherein the hyperproliferative disease comprises liver cancer, leukemia, breast cancer, colon adenocarcinoma, stomach cancer, lung cancer, Bart's esophageal cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymph cancer, kidney cancer, brain cancer, nerve cancer, nasopharyngeal cancer, oral cancer, colorectal cancer.
Biological test results show that the compound provided by the invention can inhibit tumor proliferation.
Any embodiment of any aspect of the invention may be combined with other embodiments, as long as they do not contradict. Furthermore, in any embodiment of any aspect of the invention, any feature may be applicable to that feature in other embodiments, so long as they do not contradict.
The foregoing merely summarizes certain aspects of the invention and is not intended to be limiting. These and other aspects will be more fully described below.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
The following definitions as used herein should be applied unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be found in the descriptions of "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B. Smith and Jerry March, John Wiley & Sons, New York: 2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "subject" as used herein refers to an animal. Typically the animal is a mammal. Subjects, e.g., also primates (e.g., humans, males or females), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, etc. In certain embodiments, the subject is a primate. In other embodiments, the subject is a human.
The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.
"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"chiral" is a molecule having the property of not overlapping its mirror image; and "achiral" refers to a molecule that can overlap with its mirror image.
"enantiomer" refers to two isomers of a compound that are not overlapping but are in mirror image relationship to each other.
"diastereomer" refers to a stereoisomer having two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow the general definitions of S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E.and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
Many organic compounds exist in an optically active form, i.e., they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of a molecule with respect to one or more of its chiral centers. The prefixes d and l or (+) and (-) are the symbols used to specify the rotation of plane polarized light by the compound, where (-) or l indicates that the compound is left-handed. Compounds prefixed with (+) or d are dextrorotatory. A particular stereoisomer is an enantiomer and a mixture of such isomers is referred to as an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity during the chemical reaction.
Any asymmetric atom (e.g., carbon, etc.) of a compound disclosed herein can exist in racemic or enantiomerically enriched forms, such as the (R) -, (S) -or (R, S) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
Depending on the choice of starting materials and methods, the compounds of the invention may exist as one of the possible isomers or as mixtures thereof, for example as racemates and diastereomeric mixtures (depending on the number of asymmetric carbon atoms). Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituents may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may have cis or trans configuration.
Any resulting mixture of stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, depending on differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
The racemates of any of the resulting end products or intermediates can be resolved into the optical enantiomers by known methods using methods familiar to those skilled in the art, e.g., by separation of the diastereomeric salts obtained. The racemic product can also be separated by chiral chromatography, e.g., High Performance Liquid Chromatography (HPLC) using a chiral adsorbent. In particular, Enantiomers can be prepared by asymmetric synthesis, for example, see Jacques, et al, Enantiomers, racemes and solutions (Wiley Interscience, New York, 1981); principles of Asymmetric Synthesis (2)nd Ed. Robert E. Gawley, Jeffrey Aubé, Elsevier, Oxford, UK, 2012); Eliel, E.L. Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); Wilen, S.H. Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972);Chiral Separation Techniques: A Practical Approach (Subramanian, G. Ed., Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, Germany, 2007)。
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, in compounds of the general formula above, or as specifically exemplified, sub-classes, and classes of compounds encompassed by the invention.
In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced by a substituent described herein. Unless otherwise indicated, a substituted group may have one substituent substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently.
The term "unsubstituted" means that the specified group bears no substituents.
The term "optionally substituted with … …" is used interchangeably with the term "unsubstituted or substituted with … …", i.e., the structure is unsubstituted or substituted with one or more substituents described herein. Substituents described herein include, but are not limited to, D, F, Cl, Br, I, N3、CN、NO2、OH、SH、NH2Alkyl, haloalkyl, alkenyl, alkynyl, alkoxy, alkylamino, cycloalkyl, heterocyclyl, aryl, heteroaryl, and the like.
In addition, unless otherwise explicitly indicated, the descriptions of the terms "… independently" and "… independently" and "… independently" used in the present invention are interchangeable and should be understood in a broad sense to mean that the specific items expressed between the same symbols do not affect each other in different groups or that the specific items expressed between the same symbols in the same groups do not affect each other.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-6Alkyl radical'In particular to independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group.
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein, denotes a saturated straight or branched chain monovalent hydrocarbon radical, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1-20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 3 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH)3) Ethyl group (Et, -CH)2CH3) N-propyl (n-Pr, -CH)2CH2CH3) Isopropyl group (i-Pr, -CH (CH)3)2) N-butyl (n-Bu, -CH)2CH2CH2CH3) Isobutyl (i-Bu, -CH)2CH(CH3)2) Sec-butyl (s-Bu, -CH (CH)3)CH2CH3) Tert-butyl (t-Bu, -C (CH)3)3) N-pentyl (-CH)2CH2CH2CH2CH3) 2-pentyl (-CH (CH)3)CH2CH2CH3) 3-pentyl (-CH (CH)2CH3)2) 2-methyl-2-butyl (-C (CH)3)2CH2CH3) 3-methyl-2-butyl (-CH (CH)3)CH (CH3)2),3-methyl-1-butyl (-CH)2CH2CH(CH3)2) 2-methyl-1-butyl (-CH)2CH(CH3)CH2CH3) N-hexyl (-CH)2CH2CH2CH2CH2CH3) 2-hexyl (-CH (CH)3)CH2CH2CH2CH3) 3-hexyl (-CH (CH)2CH3)(CH2CH2CH3) 2-methyl-2-pentyl (-C (CH))3)2CH2CH2CH3) 3-methyl-2-pentyl (-CH (CH)3)CH(CH3)CH2CH3) 4-methyl-2-pentyl (-CH (CH)3)CH2CH(CH3)2) 3-methyl-3-pentyl (-C (CH)3)(CH2CH3)2) 2-methyl-3-pentyl (-CH (CH)2CH3)CH(CH3)2) 2, 3-dimethyl-2-butyl (-C (CH)3)2CH(CH3)2) 3, 3-dimethyl-2-butyl (-CH (CH)3)C(CH3)3) N-heptyl, n-octyl, and the like.
The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein.
Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH)3) Ethoxy (EtO, -OCH)2CH3) 1-propoxy (n-PrO, n-propoxy, -OCH)2CH2CH3) 2-propoxy (i-PrO, i-propoxy, -OCH (CH)3)2) 1-butoxy (n-BuO, n-butoxy, -OCH)2CH2CH2CH3) 2-methyl radical-l-propoxy (i-BuO, i-butoxy, -OCH2CH(CH3)2) 2-butoxy (s-BuO, s-butoxy, -OCH (CH)3)CH2CH3) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH)3)3) 1-pentyloxy (n-pentyloxy, -OCH)2CH2CH2CH2CH3) 2-pentyloxy (-OCH (CH)3)CH2CH2CH3) 3-pentyloxy (-OCH (CH))2CH3)2) 2-methyl-2-butoxy (-OC (CH))3)2CH2CH3) 3-methyl-2-butoxy (-OCH (CH)3)CH(CH3)2) 3-methyl-l-butoxy (-OCH)2CH2CH(CH3)2) 2-methyl-l-butoxy (-OCH)2CH(CH3)CH2CH3) And so on.
The term "haloalkyl" denotes an alkyl group substituted with one or more halogen atoms, wherein the alkyl group has the meaning as described herein, examples of which include, but are not limited to, trifluoromethyl, 2,3, 3-tetrafluoropropyl, and the like.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic cyclic hydrocarbon radical containing from 3 to 12 carbon atoms. In one embodiment, cycloalkyl groups contain 7 to 12 carbon atoms; in yet another embodiment, the cycloalkyl group contains 3 to 8 carbon atoms; in yet another embodiment, the cycloalkyl group contains 3 to 6 carbon atoms. The cycloalkyl groups may be independently unsubstituted or substituted with one or more substituents described herein.
The term "heterocyclyl" refers to a saturated or partially unsaturated, non-aromatic, monovalent or polyvalent, monocyclic, bicyclic, or tricyclic ring containing 3 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms. Unless otherwise indicated, a heterocyclyl group may be attached to other groups in the molecule through a carbon atom, may be attached to other groups in the molecule through a nitrogen atom, and-CH2The-group may optionally be replaced by-C (= O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may beOptionally oxidized to an N-oxygen compound. Examples of heterocyclyl groups include, but are not limited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl, homopiperazinyl, homopiperidinyl, diazepanyl, oxepanyl, thiepanyl, oxazepinyl, diazepin, thiazepinyl, 2-oxa-5-azabicyclo [ 2.2.1.1]Hept-5-yl. In heterocyclic radicals of-CH2Examples of the substitution of the-group by-C (= O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl, 3, 5-dioxopiperidinyl and pyrimidinedione. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, sulfolane group, 1-dioxothiomorpholinyl group. The heterocyclyl group may be optionally substituted with one or more substituents as described herein.
The terms "r-atom composition" and "r-member" are used interchangeably, where r is an integer typically describing the number of ring-forming atoms in a molecule in which the number of ring-forming atoms is r. For example, piperidinyl is heterocycloalkyl or 6-membered heterocyclyl consisting of 6 atoms, and decahydronaphthyl is cycloalkyl or 10-membered cycloalkyl consisting of 10 atoms.
The term "unsaturated" as used herein means that the group contains one or more unsaturations.
The term "heteroatom" refers to O, S, N, P and Si, including N, S and any oxidation state form of P; primary, secondary, tertiary amines and quaternary ammonium salt forms; or hydrogen on nitrogen atoms in the heterocycle substituted, e.g. N (like N in 3, 4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR10(like NR in N-substituted pyrrolidinyl10)。
The term "halogen" or "halogen atom" means a fluorine atom (F), chlorine atom (Cl), bromine atom (Br) or iodine atom (I).
The term "cyano" or "CN" denotes a cyano structure, which group may be attached to another group.
The term "nitro" or "NO2"denotes a nitro structure, which may be linked to other groups.
The term "aryl" denotes a monovalent or polyvalent mono-, bi-, or tricyclic all carbocyclic ring system containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring is aromatic and has one or more attachment points to the rest of the molecule. In one embodiment, aryl is a monovalent or polyvalent carbocyclic ring system consisting of 6 to 10 ring atoms and containing at least one aromatic ring. Examples of the aryl group may include phenyl, naphthyl and anthracenyl. The aryl group may independently be optionally substituted with one or more substituents described herein.
The term "heteroaryl" denotes a monovalent or polyvalent monocyclic, bicyclic or tricyclic ring containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic and at least one aromatic ring contains one or more heteroatoms and has one or more attachment points to the rest of the molecule. The heteroaryl group is optionally substituted with one or more substituents described herein. In one embodiment, heteroaryl is a 5-12 atom heteroaryl comprising 1,2,3, or 4 heteroatoms independently selected from O, S and N; in another embodiment, heteroaryl is 5-6 atom consisting of 1,2,3, or 4 heteroatoms independently selected from O, S and N.
Examples of heteroaryl groups include, but are not limited to, 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, oxadiazolyl (e.g., 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl), oxadiazolyl (e.g., 1,2,3, 4-oxadiazolyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, isothiazolyl, 2-thiadiazolyl (e.g., 1,3, 4-thiadiazolyl, 1,2, 3-thiadiazolyl, 1,2, 5-thiadiazolyl) Thiatriazolyl (e.g., 1,2,3, 4-thiatriazolyl), tetrazolyl (e.g., 2H-1,2,3, 4-tetrazolyl, 1H-1,2,3, 4-tetrazolyl), triazolyl (e.g., 2H-1,2, 3-triazolyl, 1H-1,2, 4-triazolyl, 4H-1,2, 4-triazolyl), 2-thienyl, 3-thienyl, 1H-pyrazolyl (e.g., 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl), N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 2-triazolyl, triazolyl (e.g., 2H-1,2,3, 4-pyridyl, 4-pyrimidinyl, triazolyl, triazol, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl, 4-pyridazinyl), 2-pyrazinyl, triazinyl (e.g., 1,3, 5-triazine), tetrazinyl (e.g., 1,2,4, 5-tetrazine, 1,2,3, 5-tetrazine); the following bicyclic rings are also included, but are in no way limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl), imidazo [1,2-a ] pyridyl, pyrazolo [1,5-a ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, [1,2,4] triazolo [4,3-b ] pyridazinyl, [1,2,4] triazolo [1,5-a ] pyrimidinyl, [1,2,4] triazolo [1,5-a ] pyridyl, and the like.
The term "protecting group" or "PG" refers to a substituent that, when reacted with other functional groups, is generally used to block or protect a particular functionality. For example, "amino protecting group" means a substituent attached to an amino group to block or protect the functionality of the amino group in a compound, and suitable amino protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC ), benzyloxycarbonyl (CBZ ) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). Similarly, "hydroxyl protecting group" refers to the functionality of a substituent of a hydroxyl group to block or protect the hydroxyl group, and suitable protecting groups include acetyl and silyl groups. "carboxy protecting group" refers to the functionality of a substituent of a carboxy group to block or protect the carboxy group, and typical carboxy protecting groups include-CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) silaneAlkyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl, and the like. General descriptions of protecting groups can be found in the literature: t W. Greene, Protective Groups in Organic Synthesis, John Wiley&Sons, New York, 1991; and P. J. Kocienski, Protecting Groups, Thieme, Stuttgart, 2005.
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-C24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: t, Higuchi and V, Stella, Pro-drugs as Novel Delivery Systems, Vol, 14 of the A.C.S. Symposium Series, Edward B.Roche, ed., Bioreversible Carriers in Drug designs, American Pharmaceutical Association and Pergamon Press, 1987, J. Rautio et al, Prodrugs in Design and Clinical Applications, Nature Review Drug Discovery, 2008, 7, 255-.
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: S.M. Berge et al, description of the descriptive Pharmaceutical acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66: 1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids formed by reaction with amino groups such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or those obtained by other methods described in the literature above, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, thiocyanates, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. The alkali metal or alkaline earth metal salt includesSodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
When the solvent is water, the term "hydrate" may be used. In some embodiments, a molecule of a compound of the present invention may be associated with a molecule of water, such as a monohydrate; in other embodiments, one molecule of the compound of the present invention may be associated with more than one molecule of water, such as a dihydrate, and in still other embodiments, one molecule of the compound of the present invention may be associated with less than one molecule of water, such as a hemihydrate. It should be noted that the hydrates of the present invention retain the biological effectiveness of the compound in its non-hydrated form.
The term "solubility" as used herein refers to the mass of solute dissolved in a solid material at a certain temperature to reach saturation in 100g of solvent. The solubility tests are referenced in the following documents: (ZHU Shang-bin, et al Preparation, chromatography, and physiochemical properties of aryl phosphopeptide complex [ J ]. Chinese medicinal Traditional and Herbal drugs.2020: 1-10).
The term "acute toxicity" as used herein refers to the toxic effect or even death caused by exposure of the body (human or experimental animal) to a foreign compound once (or more than once within 24 hours). However, it should be noted that the degree of the toxic effect of the compound on the experimental animal may vary depending on the nature and amount of the compound. Some compounds may develop toxic symptoms, or even death, within minutes of exposure of the test animal to the lethal dose. Some compounds show toxic symptoms and death after several days, i.e. late death. In addition, the experimental animals will be exposed to the compound in different ways or by different routes, and the meaning of "once" will be different. By "once" is meant that the test compound is delivered into the body of the test animal at an instant, for example, by oral contact or by various means of injection. By "once" is meant the process by which the test animal is continuously exposed to the test compound for a particular period of time, and therefore "once" contains a time factor.
The term "treating" any disease or condition, as used herein, means all that can slow, halt, arrest, control or halt the progression of the disease or condition, but does not necessarily mean that all the symptoms of the disease or condition have disappeared, and also includes prophylactic treatment of the symptoms, particularly in patients susceptible to such disease or disorder. In some of these embodiments, refers to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
The term "therapeutically effective amount" or "therapeutically effective dose" as used herein refers to an amount of a compound of the invention that is capable of eliciting a biological or medical response (e.g., reducing or inhibiting enzyme or protein activity, or ameliorating symptoms, alleviating a disorder, slowing or delaying the progression of a disease, or preventing a disease, etc.) in a subject. In one non-limiting embodiment, the term "therapeutically effective amount" refers to an amount that, when administered to a subject, is effective for: (1) at least partially alleviating, inhibiting, preventing and/or ameliorating tumor proliferation. In another embodiment, the term "therapeutically effective amount" refers to an amount of a compound of the invention that is effective to at least partially reduce or inhibit tumor proliferation when administered to a cell, or organ, or non-cellular biological substance, or medium.
The terms "administration" and "administering" of a compound as used herein shall be understood as providing a compound of the invention or a prodrug of a compound of the invention to a subject in need thereof. It will be appreciated that one skilled in the art may have an effect on tumor proliferation by treating a patient currently suffering from such a disorder, or prophylactically treating a patient suffering from such a disorder, with an effective amount of a compound of the present invention.
The term "composition" as used herein refers to a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The meaning of such terms in relation to pharmaceutical compositions includes products comprising the active ingredient(s) and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from mixing, complexation or aggregation of any two or more of the ingredients, or from decomposition of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention include any composition prepared by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
Description of the Compounds of the invention
The invention discloses a brefeldin A ester derivative, a stereoisomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, a pharmaceutical preparation and a composition thereof, which can be used for preventing or treating hyperproliferative diseases, such as liver cancer, leukemia, breast cancer, colon adenocarcinoma, stomach cancer, lung cancer, Bart's esophagus cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymph cancer, kidney cancer, brain cancer, nerve cancer, nasopharyngeal cancer, oral cancer and colorectal cancer.
In one aspect, the invention relates to a compound of formula (I) or a stereoisomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (I) or a prodrug thereof,
Figure 536201DEST_PATH_IMAGE002
(I);
z is a substituted or unsubstituted pyridine ring, quinoline ring or isoquinoline ring, wherein the pyridine ring, quinoline ring and isoquinoline ring may each independently optionally be substituted with 1,2,3 or 4 substituents selected from deuterium, F, Cl, Br, I, hydroxy, amino, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C1-3Alkoxy radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, aryl, or 5-6 membered heteroaryl;
in some embodiments, Z is a substituted or unsubstituted pyridine, quinoline, or isoquinoline ring, wherein the pyridine, quinoline, and isoquinoline rings may each be independently optionally substituted with 1,2,3, or 4 substituents selected from deuterium, F, Cl, Br, I, hydroxy, amino, cyano, nitro, methyl, ethyl, n-propyl, isopropyl, difluoromethyl, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, phenyl, thiazolyl, pyrazolyl, oxazolyl, pyridinyl, or pyrimidinyl.
In some embodiments, the present invention relates to compounds, or stereoisomers, nitroxides, solvates, metabolites, pharmaceutically acceptable salts or prodrugs thereof, of one of the following, but in no way limited to these compounds:
Figure 100002_DEST_PATH_IMAGE003
Figure 112676DEST_PATH_IMAGE004
the compounds of the present disclosure may contain asymmetric or chiral centers and thus may exist in different stereoisomeric forms. The present invention contemplates that all stereoisomeric forms of the compounds of formula (I), including but not limited to diastereomers, enantiomers, atropisomers and geometric (or conformational) isomers, and mixtures thereof, such as racemic mixtures, are integral to the invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not specified, then all stereoisomers of that structure are contemplated as within this invention and are included as disclosed compounds in this invention. When stereochemistry is indicated by a solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of the structure are so well-defined and defined.
The compounds of formula (I) may exist in different tautomeric forms and all such tautomers are included within the scope of the invention.
The compounds of formula (I) may be present in the form of salts. In one embodiment, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be compatible chemically and/or toxicologically with the other ingredients comprising the formulation and/or the mammal being treated therewith. In another embodiment, the salts need not be pharmaceutically acceptable salts and may be intermediates useful in the preparation and/or purification of compounds of formula (I) and/or in the isolation of enantiomers of compounds of formula (I).
Pharmaceutically acceptable acid addition salts may be formed from the disclosed compounds of the invention by the action of an inorganic or organic acid, for example, acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheyl salt, citrate, edisylate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, phosphate, Polysilonolactates, propionates, stearates, succinates, sulfosalicylates, tartrates, tosylates and trifluoroacetates.
Pharmaceutically acceptable base addition salts may be formed from the disclosed compounds by reaction with an inorganic or organic base.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of groups I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include primary, secondary and tertiary amines, and substituted amines include naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Some organic amines include, for example, isopropylamine, benzathine (benzathine), choline salts (cholinate), diethanolamine, diethylamine, lysine, meglumine (meglumine), piperazine, and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "handbook of pharmaceutically acceptable salts: properties, Selection and application (Handbook of Pharmaceutical Salts: Properties, Selection, and Use) ", Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) may find some additional lists of suitable Salts.
In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents containing them (e.g., ethanol, DMSO, etc.), for their crystallization. The compounds disclosed herein may form solvates with pharmaceutically acceptable solvents (including water), either inherently or by design; thus, the present invention is intended to include both solvated and unsolvated forms of the disclosed compounds.
Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、11C、13C、14C、15N、17O、18O、18F、31P、32P、35S、36Cl and125I。
in another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, e.g. wherein a radioisotope, e.g. is present3H、14C and18those compounds of F, or in which a non-radioactive isotope is present, e.g.2H and13those of C. The isotopically enriched compounds can be used for metabolic studies (use)14C) Reaction kinetics study (using, for example2H or3H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient.18F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples described in the present specification using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.
In addition, heavier isotopes are, in particular, deuterium (i.e.,2substitution of H or D) may provide certain therapeutic advantages resulting from greater metabolic stability. For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is to be considered as a substituent of the compound of formula (I). The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. The term "isotopic enrichment factor" as used herein refers to the ratio between the isotopic and natural abundance of a given isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D2O, acetone-d6、DMSO-d6Those solvates of (a).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention. In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle or combination thereof. In another embodiment, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The present invention provides a pharmaceutical composition comprising a compound disclosed herein, for example, as set forth in the examples; and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof.
The present invention provides methods of treating, preventing or ameliorating a disease or condition comprising administering a safe and effective amount of a combination comprising a compound of the present disclosure and one or more therapeutically active agents. Wherein, the combined medicine contains one or more medicines for preventing or treating liver cancer, leukemia, breast cancer, colon adenocarcinoma, gastric cancer, lung cancer, Bart's esophagus cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymph cancer, renal cancer, brain cancer, nerve cancer, nasopharyngeal cancer, oral cancer and colorectal cancer, and the active ingredients of the medicines are different from the compounds disclosed by the invention.
Drugs for preventing or treating liver cancer, leukemia, breast cancer, colon adenocarcinoma, stomach cancer, lung cancer, Bart's esophagus cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymph cancer, kidney cancer, brain cancer, nerve cancer, nasopharyngeal cancer, oral cancer, colorectal cancer include, but are not limited to: aromatase inhibitors, topoisomerase I inhibitors, topoisomerase II inhibitors, microtubule active agents, alkylating agents, histone deacetylase inhibitors, compounds that induce cellular differentiation processes, cyclooxygenase inhibitors, MMP inhibitors, Mtor inhibitors, antitumor antimetabolites, platinum compounds, compounds that target/reduce the activity of protein or lipid phosphokinase and other anti-angiogenic compounds, antiproliferative antibodies, heparanase inhibitors, Ras oncogenic inhibitors, telomerase inhibitors, proteasome inhibitors, or any combination thereof.
The dosage of the active ingredient in the composition of the present invention may vary, however, the amount of the active ingredient must be such that a suitable dosage form is obtained. The active ingredient may be administered to patients (animals and humans) in need of such treatment at dosages that provide optimal pharmaceutical efficacy. The selected dosage depends on the desired therapeutic effect, on the route of administration and on the duration of the treatment. The dosage will vary from patient to patient depending on the nature and severity of the disease, the weight of the patient, the particular diet of the patient, the concurrent use of drugs, and other factors that will be recognized by those skilled in the art. The dosage range is generally about 0.5 mg to 1.0 g per patient per day and may be administered in a single dose or in multiple doses. In one embodiment, the dosage range is from about 0.5 mg to 500 mg per patient per day; from about 0.5 mg to 200 mg per patient per day in another embodiment; and in yet another embodiment from about 5 mg to 50 mg per patient per day.
It will also be appreciated that certain compounds of the invention may be present in free form and used in therapy, or if appropriate in the form of a pharmaceutically acceptable derivative thereof. Pharmaceutically acceptable derivatives include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any additional adduct or derivative that upon administration to a patient in need thereof provides, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
The medicaments or pharmaceutical compositions disclosed herein may be prepared and packaged in bulk (bulk) form, wherein a safe and effective amount of the compound of formula (I) may be extracted and then administered to a patient in the form of a powder or syrup. Typically, the administration to a patient is at a dosage level of between 0.0001 to 10 mg/kg body weight per day to achieve an effective effect. Alternatively, the pharmaceutical compositions disclosed herein can be prepared and packaged in unit dosage forms, wherein each physically discrete unit contains a safe and effective amount of a compound of formula (I). When prepared in unit dosage form, the disclosed pharmaceutical compositions can generally contain, for example, from 0.5 mg to 1 g, or from 1 mg to 700 mg, or from 5 mg to 100 mg of the disclosed compounds.
When the pharmaceutical composition of the invention contains one or more other active ingredients in addition to the compound of the invention, the compound weight ratio of the compound of the invention to the second active ingredient may vary and depends on the effective dose of each ingredient. Generally, an effective dose of each is used. Thus, for example, when a compound of the present invention is mixed with another pharmaceutical agent, the weight ratio of the compound of the present invention to the other pharmaceutical agent typically ranges from about 1000: 1 to about 1: 1000, e.g., from about 200: 1 to about 1: 200. Mixtures of the compounds of the invention with other active ingredients are generally also within the above-mentioned ranges, but in each case an effective dose of each active ingredient should be used.
As used herein, "pharmaceutically acceptable excipient" means a pharmaceutically acceptable material, mixture or vehicle, which is compatible with the dosage form or pharmaceutical composition to be administered. Each excipient, when mixed, must be compatible with the other ingredients of the pharmaceutical composition to avoid interactions that would substantially reduce the efficacy of the disclosed compounds and which would result in a pharmaceutical composition that is not pharmaceutically acceptable when administered to a patient. Furthermore, each excipient must be pharmaceutically acceptable, e.g., of sufficiently high purity.
Suitable pharmaceutically acceptable excipients will vary depending on the particular dosage form selected. In addition, pharmaceutically acceptable excipients may be selected for their specific function in the composition. For example, certain pharmaceutically acceptable excipients may be selected to aid in the production of a uniform dosage form. Certain pharmaceutically acceptable excipients may be selected to aid in the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be selected to facilitate carrying or transporting the disclosed compounds from one organ or portion of the body to another organ or portion of the body when administered to a patient. Certain pharmaceutically acceptable excipients may be selected that enhance patient compliance.
Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants and buffers. The skilled artisan will recognize that certain pharmaceutically acceptable excipients may provide more than one function, and provide alternative functions, depending on how many such excipients are present in the formulation and which other excipients are present in the formulation.
The skilled person is knowledgeable and skilled in the art to enable them to select suitable amounts of suitable pharmaceutically acceptable excipients for use in the present invention. Furthermore, there is a large amount of resources available to the skilled person, who describes pharmaceutically acceptable excipients and is used to select suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (The American Pharmaceutical Association and The Pharmaceutical Press).
Various carriers for The formulation of pharmaceutically acceptable compositions, and well known techniques for their preparation, are disclosed in Remington, The Science and Practice of Pharmacy, 21st edition, 2005, ed. D.B. Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J.C. Boylan, 1988-1999, Marcel Dekker, New York, The contents of each of which are incorporated herein by reference. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention (e.g., exhibits any undesirable biological effect or otherwise interacts in a deleterious manner with any other component of a pharmaceutically acceptable composition), its use is contemplated as falling within the scope of the present invention.
The pharmaceutical compositions disclosed herein are prepared using techniques and methods known to those skilled in the art. Some commonly used methods in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company).
Thus, in another aspect, the invention relates to a process for preparing a pharmaceutical composition comprising a compound of the present disclosure and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof, which process comprises admixing the ingredients. Pharmaceutical compositions comprising the disclosed compounds may be prepared by mixing, for example, at ambient temperature and atmospheric pressure.
The compounds disclosed herein are generally formulated in a dosage form suitable for administration to a patient by a desired route. For example, dosage forms include those suitable for the following routes of administration: (1) oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets and cachets; (2) parenteral administration, such as sterile solutions, suspensions, and reconstituted powders; (3) transdermal administration, such as transdermal patches; (4) rectal administration, e.g., suppositories; (5) inhalation, such as aerosols, solutions, and dry powders; and (6) topical administration, such as creams, ointments, lotions, solutions, pastes, sprays, foams and gels.
In one embodiment, the compounds disclosed herein may be formulated in oral dosage forms. In another embodiment, the compounds disclosed herein may be formulated in an inhalation dosage form. In another embodiment, the compounds disclosed herein can be formulated for nasal administration. In yet another embodiment, the compounds disclosed herein can be formulated for transdermal administration. In yet another embodiment, the compounds disclosed herein may be formulated for topical administration.
The pharmaceutical compositions provided by the present invention may be provided as compressed tablets, milled tablets, chewable lozenges, fast-dissolving tablets, double-compressed tablets, or enteric-coated, sugar-coated or film-coated tablets. Enteric coated tablets are compressed tablets coated with a substance that is resistant to the action of gastric acid but dissolves or disintegrates in the intestine, thereby preventing the active ingredient from contacting the acidic environment of the stomach. Enteric coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalate. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which can help to mask unpleasant tastes or odors and prevent oxidation of the tablet. Film-coated tablets are compressed tablets covered with a thin layer or film of a water-soluble substance. Film coatings include, but are not limited to, hydroxyethyl cellulose, sodium carboxymethyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coatings are endowed with the same general characteristics as sugar coatings. A tabletted tablet is a compressed tablet prepared over more than one compression cycle, including a multi-layer tablet, and a press-coated or dry-coated tablet.
Tablet dosage forms may be prepared from the active ingredient in powder, crystalline or granular form, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled release polymers, lubricants, diluents and/or colorants. Flavoring and sweetening agents are particularly useful in forming chewable tablets and lozenges.
The pharmaceutical composition provided by the present invention may be provided in soft or hard capsules, which may be prepared from gelatin, methylcellulose, starch or calcium alginate. The hard gelatin capsules, also known as Dry Fill Capsules (DFC), consist of two segments, one inserted into the other, thus completely encapsulating the active ingredient. Soft Elastic Capsules (SEC) are soft, spherical shells, such as gelatin shells, which are plasticized by the addition of glycerol, sorbitol or similar polyols. The soft gelatin shell may contain a preservative to prevent microbial growth. Suitable preservatives are those as described herein, including methyl and propyl parabens, and sorbic acid. The liquid, semi-solid and solid dosage forms provided by the present invention may be encapsulated in a capsule. Suitable liquid and semi-solid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils or triglycerides. Capsules containing such solutions may be prepared as described in U.S. Pat. nos. 4,328,245, 4,409,239 and 4,410,545. The capsules may also be coated as known to those skilled in the art to improve or maintain dissolution of the active ingredient.
The pharmaceutical compositions provided herein may be provided in liquid and semi-solid dosage forms, including emulsions, solutions, suspensions, elixirs and syrups. Emulsions are two-phase systems in which one liquid is dispersed throughout another in the form of globules, which can be either oil-in-water or water-in-oil. Emulsions may include pharmaceutically acceptable non-aqueous liquids and solvents, emulsifiers and preservatives. Suspensions may include a pharmaceutically acceptable suspending agent and a preservative. The aqueous alcoholic solution may comprise pharmaceutically acceptable acetals, such as di (lower alkyl) acetals of lower alkyl aldehydes, e.g. acetaldehyde diethyl acetal; and water-soluble solvents having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, hydroalcoholic solutions. Syrups are concentrated aqueous solutions of sugars, such as sucrose, and may also contain preservatives. For liquid dosage forms, for example, a solution in polyethylene glycol may be diluted with a sufficient amount of a pharmaceutically acceptable liquid carrier, such as water, for precise and convenient administration.
Other useful liquid and semi-solid dosage forms include, but are not limited to, those comprising the active ingredients provided herein and a secondary mono-or poly-alkylene glycol, including: 1, 2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, where 350, 550, 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations may further include one or more antioxidants, such as Butylated Hydroxytoluene (BHT), Butylated Hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
Dosage unit formulations for oral administration may be microencapsulated, where appropriate. They may also be prepared as extended or sustained release compositions, for example by coating or embedding the particulate material in a polymer, wax or the like.
The oral pharmaceutical composition provided by the invention can also be provided in the form of liposome, micelle, microsphere or nano system. Micellar dosage forms can be prepared using the methods described in U.S. Pat. No. 6,350,458.
The pharmaceutical compositions provided herein can be provided as non-effervescent or effervescent granules and powders for reconstitution into liquid dosage forms. Pharmaceutically acceptable carriers and excipients used in non-effervescent granules or powders may include diluents, sweeteners and wetting agents. Pharmaceutically acceptable carriers and excipients used in effervescent granules or powders may include organic acids and sources of carbon dioxide.
Coloring and flavoring agents may be used in all of the above dosage forms.
The disclosed compounds may also be conjugated to soluble polymers as targeted drug carriers. Such polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol or polyoxyethylene polylysine substituted with palmitoyl residues. In addition, the disclosed compounds may be combined with a class of biodegradable polymers used in achieving controlled release of a drug, such as polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and crosslinked or amphiphilic block copolymers of hydrogels.
The pharmaceutical compositions provided by the present invention may be formulated into immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed release forms.
The pharmaceutical compositions provided by the present invention may be co-formulated with other active ingredients that do not impair the intended therapeutic effect, or with substances that supplement the intended effect.
The pharmaceutical compositions provided by the present invention may be administered parenterally by injection, infusion or implantation for local or systemic administration. Parenteral administration as used herein includes intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous administration.
The pharmaceutical compositions provided herein can be formulated in any dosage form suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems and solid forms suitable for solution or suspension in a liquid prior to injection. Such dosage forms may be prepared according to conventional methods known to those skilled in The art of pharmaceutical Science (see Remington: The Science and Practice of Pharmacy, supra).
Pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives to inhibit microbial growth, stabilizers, solubility enhancers, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, thickening agents, pH adjusting agents, and inert gases.
Suitable aqueous carriers include, but are not limited to: water, saline, normal saline or Phosphate Buffered Saline (PBS), sodium chloride injection, Ringers injection, isotonic glucose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and the medium chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1, 3-butanediol, liquid polyethylene glycols (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerol, N-methyl-2-pyrrolidone, N-dimethylacetamide, and dimethylsulfoxide.
Suitable antimicrobial agents or preservatives include, but are not limited to, phenol, cresol, mercurial, benzyl alcohol, chlorobutanol, methyl and propyl parabens, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl and propyl parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerol and glucose. Suitable buffers include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcellulose, hydroxypropylmethylcellulose and polyvinylpyrrolidone. Suitable emulsifiers include those described herein, including polyoxyethylene sorbitan monolaurate. Polyoxyethylene sorbitan monooleate 80 and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to, EDTA. Suitable pH adjusters include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta-cyclodextrin, sulfobutyl ether-beta-cyclodextrin, and sulfobutyl ether 7-beta-Cyclodextrin (CAPTISOL)®, CyDex, Lenexa, KS)。
The pharmaceutical compositions provided herein may be formulated for single or multiple dose administration. The single dose formulations are packaged in ampoules, vials or syringes. The multi-dose parenteral formulation must contain a bacteriostatic or fungistatic concentration of the antimicrobial agent. All parenteral formulations must be sterile, as is known and practiced in the art.
In one embodiment, the pharmaceutical composition is provided as a ready-to-use sterile solution. In another embodiment, the pharmaceutical compositions are provided as sterile dried soluble products, including lyophilized powders and subcutaneous injection tablets, which are reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a ready-to-use sterile suspension. In yet another embodiment, the pharmaceutical composition is formulated as a sterile, dry, insoluble product that is reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a sterile emulsion ready for use.
The pharmaceutical composition may be formulated as a suspension, solid, semi-solid, or thixotropic liquid for depot administration for implantation. In one embodiment, the disclosed pharmaceutical compositions are dispersed in a solid internal matrix surrounded by an outer polymeric membrane that is insoluble in body fluids but allows diffusion therethrough of the active ingredient in the pharmaceutical composition.
Suitable internal matrices include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, silicone carbonate copolymers, hydrogels of hydrophilic polymers such as esters of acrylic and methacrylic acid, collagen, crosslinked polyvinyl alcohol, and partially hydrolyzed polyvinyl acetate of the class of copolymers.
Suitable outer polymeric films include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, neoprene, chlorinated polyethylene, polyvinyl chloride, copolymers of chlorinated ethylene and vinyl acetate, vinylidene chloride, ethylene and propylene, ionomers polyethylene terephthalate, butyl rubber chlorohydrin rubber, ethylene/vinyl alcohol copolymers, ethylene/vinyl acetate/vinyl alcohol terpolymers, and ethylene/ethyleneoxyethanol copolymers.
In another aspect, the disclosed pharmaceutical compositions may be formulated in any dosage form suitable for administration to a patient by inhalation, such as a dry powder, aerosol, suspension, or solution composition. In one embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient as a dry powder. In yet another embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient via a nebulizer. Dry powder compositions for delivery to the lung by inhalation typically comprise a finely powdered compound disclosed herein and one or more finely powdered pharmaceutically acceptable excipients. Pharmaceutically acceptable excipients that are particularly suitable for use as dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-and polysaccharides. Fine powders may be prepared, for example, by micronization and milling. Generally, the size-reduced (e.g., micronized) compound may pass through a D of about 1 to 10 microns50Values (e.g., measured by laser diffraction).
Aerosols can be formulated by suspending or dissolving the disclosed compounds in a liquefied propellant. Suitable propellants include chlorinated hydrocarbons, hydrocarbons and other liquefied gases. Representative propellants include: trichlorofluoromethane (propellant 11), dichlorofluoromethane (propellant 12), dichlorotetrafluoroethane (propellant 114), tetrafluoroethane (HFA-134 a), 1-difluoroethane (HFA-152 a), difluoromethane (HFA-32), pentafluoroethane (HFA-12), heptafluoropropane (HFA-227 a), perfluoropropane, perfluorobutane, perfluoropentane, butane, isobutane and pentane. Aerosols comprising the compounds disclosed herein are typically administered to a patient via a Metered Dose Inhaler (MDI). Such devices are known to those skilled in the art
The aerosol may contain additional pharmaceutically acceptable excipients that may be used by MDIs, such as surfactants, lubricants, co-solvents, and other excipients, to improve the physical stability of the formulation, to improve valve characteristics, to improve solubility, or to improve taste.
Pharmaceutical compositions suitable for transdermal administration may be prepared as discrete patches intended to remain in intimate contact with the epidermis of the patient for an extended period of time. For example, the active ingredient may be delivered from a patch agent by iontophoresis, as generally described in Pharmaceutical Research, 3(6), 318 (1986).
Pharmaceutical compositions suitable for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For example, ointments, creams and gels may be formulated with a water or oil base, and suitable thickeners and/or gelling agents and/or solvents. Such bases may include, water, and/or oils such as liquid paraffin and vegetable oils (e.g., peanut oil or castor oil), or solvents such as polyethylene glycol. Thickeners and gelling agents used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycol, lanolin, beeswax, carbopol and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifiers.
Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
Powders for external use may be formed in the presence of any suitable powder base, for example talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base containing one or more dispersing agents, solubilising agents, suspending agents or preservatives.
Topical formulations may be administered by application to the affected area one or more times per day; an occlusive dressing covering the skin is preferably used. Adhesive depot systems allow for continuous or extended administration.
Use of the Compounds and compositions of the invention
The disclosed compounds or pharmaceutical compositions can be used in the preparation of a medicament for the treatment, prevention, amelioration, control or alleviation of hyperproliferative disorders in a mammal, including a human.
In particular, the compounds of the present invention can be used for preventing or treating human hyperproliferative diseases, including liver cancer, leukemia, breast cancer, colon adenocarcinoma, stomach cancer, lung cancer, Bart's esophagus cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymph cancer, kidney cancer, brain cancer, nerve cancer, nasopharyngeal cancer, oral cancer, colorectal cancer.
The compounds or compositions of the present invention can be used in, but are in no way limited to, the prevention, treatment, or alleviation of hyperproliferative disorders in mammals, including humans, by administering to a patient an effective amount of a compound or composition of the present invention.
In addition to being beneficial for human therapy, the compounds and pharmaceutical compositions of the present invention may also find application in veterinary therapy for pets, animals of the introduced species and mammals in farm animals. Examples of other animals include horses, dogs, and cats. Herein, the compound of the present invention includes pharmaceutically acceptable derivatives thereof.
Method of treatment
In one embodiment, the presently disclosed methods of treatment comprise administering to a patient in need thereof a safe and effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the present invention. Various embodiments of the present disclosure include methods of treating the above-mentioned diseases by administering to a patient in need thereof a safe and effective amount of a disclosed compound or a pharmaceutical composition comprising a disclosed compound.
In one embodiment, the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds may be administered by any suitable route of administration, including systemic and topical administration. Systemic administration includes oral, parenteral, transdermal and rectal administration. Typical parenteral administration refers to administration by injection or infusion, including intravenous, intramuscular, and subcutaneous injection or infusion. Topical administration includes application to the skin and intraocular, otic, intravaginal, inhalation, and intranasal administration. In one embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered orally. In another embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered by inhalation. In yet another embodiment, the presently disclosed compounds or compositions comprising the presently disclosed compounds may be administered intranasally.
In one embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered once or several times at different time intervals over a specified period of time according to a dosing regimen. For example, once, twice, three times or four times daily. In one embodiment, the administration is once daily. In yet another embodiment, the administration is twice daily. The administration may be carried out until the desired therapeutic effect is achieved or the desired therapeutic effect is maintained indefinitely. Suitable dosing regimens for the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds depend on the pharmacokinetic properties of the compound, such as dilution, distribution and half-life, which can be determined by the skilled person. In addition, suitable dosing regimens for the compounds or pharmaceutical compositions comprising the disclosed compounds, including the duration of the regimen, will depend on the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient being treated, the nature of concurrent therapy, the desired therapeutic effect, and other factors within the knowledge and experience of the skilled artisan. Such a skilled artisan will also appreciate that adjustments to the subject's response to the dosage regimen, or the need for changes in the subject's patient over time, may be required.
The compounds disclosed herein may be administered simultaneously, or before or after, one or more other therapeutic agents. The compounds of the present invention may be administered separately, or together with other therapeutic agents, by the same or different routes of administration, in the form of pharmaceutical compositions.
For an individual of about 50-70 kg, the disclosed pharmaceutical compositions and combinations may be in unit dosage form containing from about 1-1000 mg, or from about 1-500 mg, or from about 1-250 mg, or from about 1-150 mg, or from about 0.5-100 mg, or from about 1-50 mg of the active ingredient. The therapeutically effective amount of the compound, pharmaceutical composition or combination thereof will depend on the species, weight, age and condition of the individual, the disease (disorder) or illness (disease) being treated, or the severity thereof. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each active ingredient to prevent, treat or inhibit the progression of the disease (disorder) or condition (disease).
The above cited dose profiles have been demonstrated in vitro and in vivo tests using beneficial mammals (e.g., mice, rats, dogs, monkeys) or isolated organs, tissues and specimens thereof. The compounds disclosed herein are used in vitro in the form of solutions, e.g. aqueous solutions, and also enterally, parenterally, especially intravenously, in vivo, e.g. in the form of suspensions or aqueous solutions.
In one embodiment, a therapeutically effective dose of a compound of the present disclosure is from about 0.1 mg to about 2,000 mg per day. The pharmaceutical composition thereof should provide a dose of the compound of about 0.1 mg to about 2,000 mg. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide from about 1 mg to about 2,000 mg, from about 10 mg to about 1,000 mg, from about 20 mg to about 500 mg, or from about 25 mg to about 250 mg of the principal active ingredient or a combination of principal ingredients per dosage unit form. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide about 10 mg, 20 mg, 25 mg, 50 mg, 100 mg, 250 mg, 500 mg, 1000 mg or 2000 mg of the primary active ingredient.
In addition, the compounds disclosed herein may be administered in the form of a prodrug. In the present invention, a "prodrug" of a disclosed compound is a functional derivative that, when administered to a patient, is ultimately released in vivo. When administering the compounds disclosed herein in the form of a prodrug, one skilled in the art can practice one or more of the following: (a) altering the in vivo onset time of the compound; (b) altering the duration of action of the compound in vivo; (c) altering the in vivo delivery or distribution of the compound; (d) altering the in vivo solubility of the compound; and (e) overcoming side effects or other difficulties faced by the compounds. Typical functional derivatives useful for preparing prodrugs comprise variants of the compounds which are cleaved in vivo either chemically or enzymatically. These variants, which involve the preparation of phosphates, amides, esters, thioesters, carbonates and carbamates, are well known to those skilled in the art.
The compounds, pharmaceutical compositions and uses thereof provided by the present invention are further illustrated below in connection with the examples.
Examples
Example 1 4-O-Nicotinyl BFA (Compound 1)
Figure 100002_DEST_PATH_IMAGE005
Under the protection of nitrogen, BFA (40 mg, 1 eq), DMAP (1 eq) and EDC.HCl (4 eq) are dissolved in 8ml of anhydrous DCM, stirred for 10 min at 40 ℃, added with a nicotinic acid (2 eq) DCM solution, continuously reacted for 2h, added with water for extraction after TLC detection reaction is finished, organic phase is decompressed and concentrated, and crude extract is separated and purified to obtain white solid with the yield of 40%.1H NMR (400 MHz, Chloroform-d) δ = 9.23 (dd, J=2.2, 0.9, 1H), 8.78 (dd, J=4.9, 1.7, 1H), 8.30 (dt, J=8.0, 2.0, 1H), 7.40 (ddd, J=7.9, 4.9, 0.9, 1H), 7.32 (dd, J=15.7, 3.4, 1H), 5.79 – 5.66 (m, 2H), 5.51 (ddd, J=10.5, 3.4, 1.9, 1H), 5.33 (dd, J=15.2, 9.5, 1H), 4.83 (dqd, J=10.9, 6.2, 1.8, 1H), 4.32 (tt, J=5.3, 2.5, 1H), 2.48 (qd, J=9.2, 6.7, 1H), 2.35 (tt, J=10.5, 8.2, 1H), 2.23 (ddd, J=14.1, 9.3, 5.1, 1H), 2.12 (d, J=3.1, 1H), 2.05 – 1.94 (m, 2H), 1.90 – 1.78 (m, 2H), 1.78 – 1.62 (m, 2H), 1.58 – 1.47 (m, 2H), 1.22 (d, J=6.3, 3H), 1.00 – 0.86 (m, 1H). 13C NMR (150 MHz, Chloroform-d) δ = 165.7, 164.3, 154.0, 151.1, 146.9, 137.4, 136.5, 131.0, 125.7, 123.7, 118.7, 77.7, 72.5, 72.2, 49.8, 44.5, 43.3, 41.2, 34.2, 32.0, 26.8, 21.0. HRESIMS m/z 386.1955 [M + H]+ (calcd. for [C22H28O5N]+, 386.1962)。
Example 2 4-O- (2-Picolinic acid) acyl BFA (Compound 2)
Figure 193371DEST_PATH_IMAGE006
The preparation method of reference example 1 gave a white solid with a yield of 45%.1H NMR (400 MHz, Chloroform-d) δ = 8.82 – 8.74 (m, 1H), 8.17 – 8.08 (d, J=7.8, 1H), 7.90 – 7.80 (td, J=7.8, 1.7, 1H), 7.53 – 7.45 (dd, J=7.7, 4.8, 1H), 7.39 – 7.28 (dd, J=15.7, 3.3, 1H), 5.86 – 5.77 (dt, J=15.7, 1.7, 1H), 5.77 – 5.67 (ddd, J=14.9, 10.1, 4.5, 1H), 5.64 – 5.55 (ddd, J=10.3, 3.4, 1.7, 1H), 5.38 – 5.26 (dd, J=15.2, 9.2, 1H), 4.88 – 4.77 (m, 1H), 4.37 – 4.27 (p, J=4.6, 1H), 2.55 – 2.35 (ddd, J=31.1, 13.8, 8.4, 2H), 2.28 – 2.17 (ddd, J=14.1, 9.0, 5.2, 1H), 2.07 – 1.99 (m, 2H), 1.89 – 1.82 (dd, J=11.4, 4.5, 2H), 1.77 – 1.66 (dt, J=13.3, 8.4, 2H), 1.59 – 1.47 (m, 2H), 1.25 – 1.21 (d, J=6.3, 3H), 1.00 – 0.87 (ddt, J=17.8, 10.2, 4.6, 1H). 13C NMR (100 MHz, Chloroform-d) δ 165.7, 164.0, 150.2, 147.5, 146.9, 137.2, 136.5, 130.8, 127.3, 125.4, 118.6, 77.9, 72.4, 72.0, 49.5, 44.5, 43.2, 41.1, 34.1, 31.9, 26.7, 20.9. HRESIMS m/z 386.1956 [M + H]+(calcd. for [C22H28O5N]+, 386.1962)。
Example 3 4-O- (4-Pyridinecarboxylic acid) acyl BFA (Compound 3)
Figure 538902DEST_PATH_IMAGE007
The preparation method of reference example 1 gave a white solid with a yield of 47%.1H NMR (400 MHz, Chloroform-d) δ = 8.86 – 8.69 (d, J=5.0, 2H), 7.93 – 7.82 (m, 2H), 7.36 – 7.28 (dd, J=15.7, 3.4, 1H), 5.80 – 5.66 (m, 2H), 5.57 – 5.48 (ddd, J=10.5, 3.4, 1.8, 1H), 5.40 – 5.28 (dd, J=15.2, 9.5, 1H), 4.90 – 4.78 (dqd, J=12.5, 6.2, 1.7, 1H), 4.38 – 4.29 (td, J=5.1, 2.4, 1H), 2.56 – 2.43 (qd, J=9.2, 6.6, 1H), 2.42 – 2.32 (m, 1H), 2.30 – 2.20 (ddd, J=14.3, 9.4, 5.2, 1H), 2.07 – 1.94 (m, 2H), 1.93 – 1.80 (m, 3H), 1.79 – 1.61 (dddd, J=28.3, 14.0, 9.2, 3.5, 2H), 1.59 – 1.46 (m, 2H), 1.29 – 1.20 (d, J=6.3, 3H), 1.00 – 0.87 (m, 1H). 13C NMR (100 MHz, Chloroform-d) δ 165.6, 164.1, 150.8, 146.5, 136.9, 136.4, 131.0, 123.0, 118.7, 78.0, 72.4, 72.2, 49.8, 44.5, 43.3, 41.2, 34.2, 31.9, 26.7, 20.9. HRESIMS m/z 386.1957 [M + H]+ (calcd. for [C22H28O5N]+, 386.1962)。
Example 4-O- (5-Fluoronicotinic acid) acyl BFA (Compound 4)
Figure DEST_PATH_IMAGE008
The preparation method of reference example 1 gave a white solid with a yield of 32%.1H NMR (400 MHz, Chloroform-d) δ = 9.13 – 9.03 (d, J=1.5, 1H), 8.72 – 8.65 (d, J=2.8, 1H), 8.06 – 7.96 (ddd, J=8.5, 2.9, 1.6, 1H), 7.36 – 7.27 (dd, J=15.7, 3.4, 1H), 5.80 – 5.68 (m, 2H), 5.57 – 5.49 (ddd, J=10.5, 3.4, 1.8, 1H), 5.39 – 5.31 (dd, J=15.2, 9.5, 1H), 4.92 – 4.79 (dtd, J=9.3, 6.3, 4.5, 1H), 4.42 – 4.31 (q, J=4.4, 1H), 2.56 – 2.44 (qd, J=9.2, 6.6, 1H), 2.44 – 2.31 (p, J=9.3, 1H), 2.31 – 2.22 (ddd, J=14.3, 9.4, 5.2, 1H), 2.10 – 1.95 (m, 2H), 1.94 – 1.81 (m, 2H), 1.78 – 1.62 (m, 2H), 1.60 – 1.48 (dddd, J=14.8, 12.4, 6.1, 4.4, 3H), 1.27 – 1.21 (d, J=6.3, 3H), 1.01 – 0.89 (m, 1H). 13C NMR (100 MHz, Chloroform-d) δ 165.6, 163.3, 146.8, 146.4, 142.9, 142.7, 136.4, 131.1, 123.9, 123.7, 118.8, 78.1, 72.5, 72.2, 49.7, 44.5, 43.3, 41.2, 34.2, 32.0, 26.7, 20.9. HRESIMS m/z 404.1861 [M + H]+ (calcd. for [C22H27O5NF]+, 404.1868)。
Example 5 4-O- (5-methylnicotinic acid) acyl BFA (Compound 28)
Figure 892523DEST_PATH_IMAGE009
The preparation method of reference example 1 gave a white solid with a yield of 48%.1H NMR (400 MHz, Chloroform-d) δ = 9.08 – 8.99 (s, 1H), 8.66 – 8.57 (s, 1H), 8.13 – 8.07 (d, J=2.0, 1H), 7.38 – 7.28 (dd, J=15.7, 3.3, 1H), 5.80 – 5.67 (m, 2H), 5.55 – 5.48 (ddd, J=10.5, 3.3, 1.8, 1H), 5.38 – 5.27 (m, 1H), 4.89 – 4.78 (m, 1H), 4.37 – 4.29 (t, J=4.6, 1H), 2.54 – 2.43 (qd, J=9.2, 6.8, 1H), 2.43 – 2.38 (s, 3H), 2.38 – 2.30 (q, J=9.0, 1H), 2.29 – 2.19 (ddd, J=14.3, 9.4, 5.2, 1H), 2.04 – 1.97 (m, 2H), 1.90 – 1.81 (m, 2H), 1.76 – 1.61 (dddd, J=22.6, 13.9, 8.1, 5.1, 2H), 1.59 – 1.47 (m, 2H), 1.25 – 1.20 (d, J=6.2, 3H), 1.00 – 0.86 (m, 1H). 13C NMR (100 MHz, Chloroform-d) δ 165.7, 164.5, 154.3, 148.2, 146.9, 137.6, 136.5, 133.5, 130.8, 125.2, 118.5, 77.5, 72.3, 72.1, 49.7, 44.5, 43.3, 41.1, 34.1, 31.9, 26.7, 20.9, 18.4. HRESIMS m/z 400.2114 [M + H]+ (calcd. for [C23H30O5N]+, 400.2118)。
Example 6 solubility testing
The test purpose is as follows: to compare the solubility differences of the compounds of the invention with BFA
The test method comprises the following steps: excess BFA, compound of the invention were added to 10 mL of methanol, acetone and 5% DMSO +95% H, respectively2And oscillating the mixture in three solvents O for 24 hours at constant temperature of 100 r/min on a shaker at 37 ℃. After 24 h, the sample was centrifuged at 15000 r/min for 15 min to remove undissolved compounds. The supernatant was filtered using a 0.45 μm membrane filter. The supernatant was diluted appropriately with methanol, and the content of the compound was measured by an HPLC method to calculate the solubility.
As a result:
table 1 solubility of the compounds of the invention (x ± s, n = 3)
Figure DEST_PATH_IMAGE010
Note: the invention tests compounds with Z as benzene ring or alkyl substituent and disubstituted compounds, namely compound A (A)
Figure 457758DEST_PATH_IMAGE011
) Compound B: (A)
Figure DEST_PATH_IMAGE012
) Compound C (A), (B) and (C)
Figure 453396DEST_PATH_IMAGE013
) For comparison.
The results show that the compounds of the invention are in methanol, acetone and 5% DMSO +95% H2The solubility of the O solvent is far higher than that of BFA, and Z is a benzene ring or alkyl substituent compound or a disubstituted compound, thereby showing remarkable advantages.
Example 7 biological Activity assay
Test example 1 cell proliferation test
1) The detection principle is as follows:
CCK-8 method: the CCK-8 method is a highly sensitive, radioactivity-free colorimetric assay for determining the number of viable cells in a cell proliferation or cytotoxicity assay. The orange formazan dye generated after CCK-8 is biologically reduced by intracellular dehydrogenase can be dissolved in a cell culture medium, and the generated formazan amount is in direct proportion to the number of living cells. The kit used in the method is tetrazolium salt-WST-8 (2- (2-Methoxy-4-nitrophenyl) -3- (4-nitrophenyl) -5- (2, 4-disulfophenyl) -2H-tetrazole monosodium salt) developed by Dojindo research institute (Dojindo), and the tetrazolium salt can be reduced into water-soluble formazan dye in the presence of an electron carrier 1-Methoxy PMS.
2) Cell culture and test compound preparation
Human cervical cancer cell Hela, pancreatic cancer cell PANC-1, esophageal cancer cell Eca-109, human leukemia cell K562, human lung cancer cell A549, human liver cancer cell HepG2, human gastric cancer cell MKN-45, human breast cancer cell MCF7 were cultured in DMEM medium containing 10% FBS (fetal bovine serum), 100U/ml penicillin and 100. mu.g/ml streptomycin. Placing hepatoma carcinoma cell Bel-7402 in a medium containing 10% FBS(fetal bovine serum), 100U/ml penicillin and 100. mu.g/ml streptomycin in RPMI-1640 medium. All cells were incubated at 37 ℃ in 5% CO2Cultured in a cell culture box. And (3) changing the liquid of the cells once every 3-5 days, after 80% of the cells are fused, digesting with pancreatin, carrying out passage, and keeping the cells in a good logarithmic growth phase.
The samples to be tested were dissolved in DMSO.
3) The detection method comprises the following steps:
cells in the logarithmic growth phase are respectively inoculated into a 96-well plate at 5000/well, and after 24 h of culture, samples to be tested (the final concentration is shown in the table) are added, wherein each sample is provided with 2 multiple wells. The dosage of the solvent DMSO is not higher than 2 per mill. After the drug acts for 72 hours, 10 mul of CCK-8 solution is added into each hole, and the incubation is carried out for 1.5 to 3 hours at the temperature of 37 ℃. OD at 450 nm was measured on a microplate reader.
Inhibition rate (%) - (1- [ (OD)450Dosing well-OD450Blank well)/(OD450Control well-OD450Blank hole)])×100%
Calculation of the fitted IC Using the Lo (inhibitor) vs. response-Variable slope analysis method in GraphPad Prism software50The value is obtained.
4) And (3) detection results:
TABLE 2 Effect of the Compounds of the invention on tumor cell proliferation
Figure DEST_PATH_IMAGE014
In the table, "A" represents IC50The value is less than 0.1. mu.M, "B" denotes IC50The value is between 0.1 and 1. mu.M, "C" denotes IC50The value is between 1 and 10. mu.M, "D" denotes IC50The value is greater than 10. mu.M. In addition, the present invention tests the effect of compounds a, B, C on tumor cell proliferation in parallel.
Through in vitro cell experiment screening, the compound of the invention shows moderate to strong cytotoxic activity to a plurality of cancer cell lines, the activity is stronger than that of positive drug adriamycin or equivalent to the positive drug adriamycin, and is slightly better than that of compounds A, B and C, and part of compounds simultaneously show better selectivity. Therefore, the compound has better potential application prospect in the aspect of inhibiting the tumor proliferation activity.
Test example 2 acute toxicity test
The purpose of the test is as follows: the toxic effects of the compounds after intraperitoneal (ip.) administration to mice were observed.
Test materials:
1) medicine preparation: the compounds of the invention are formulated at a suitable concentration for use.
1) Animals: kunming mouse, 18-22 g weight, male. Supplied by experimental animal breeding limited of Jinnanpunyue, animal license number: SCXK (Lu 20140007).
The test method comprises the following steps:
kunming mice are randomly grouped according to body weight, and each group comprises 6 mice, namely a normal control group, a BFA high-low dose group (100 mg/kg and 50 mg/kg respectively), and a part of compound high-low dose group (BFA effective dose conversion). After administration, the animals were observed for behavioral activity, mental status, food intake, drinking, and other status responses and death. After 14 days of continuous dosing, all animals were weighed, sacrificed, dissected and visually observed for abnormalities.
And (3) test results:
a) the high dose group of BFA died at 12 d of administration and the low dose group died at 14 d of administration.
b) Animals with high and low doses of the compounds 1,2,3,4 and 28 of the invention have good mental states and no death, and the skin and hair of the animals are smoother when the compounds of the invention are administrated compared with those of the animals administrated with BFA in the administration process. The 14 d dissection result shows that all animals do not have obvious abnormality when dissecting and observing organs.
c) The body weight of the animals in the administration group is slightly lower than that of the normal control group, but no significant difference exists through statistical comparison.
The results show that compared with BFA, the compound of the invention has obviously reduced toxicity after being administrated to mice and has certain attenuation effect.
Test example 3 pharmacokinetic evaluation
1) Test method
SD rats were weighed after overnight fasting for 15 hours and randomly grouped according to body weight. Accurately weighing a proper amount of test compound, adding 5% DMSO, 10% Solutol and 85% Saline in final volume, and fully and uniformly mixing by vortex or ultrasound to obtain 0.5 mg/mL administration solution for intravenous injection administration (iv). Then, venous blood was taken at time points of 0, 0.083, 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24 hours, blood was collected via jugular vein or other suitable means, about 0.20 mL of each sample was collected, heparin sodium was anticoagulated (6.25. mu.L of 200 mM DDVP/tube was added), placed on ice after collection, and plasma was centrifuged within 2 hours (centrifugation conditions: 6800g, 6 minutes, 2-8 ℃). The collected plasma samples were stored in a-70 ℃ freezer before analysis, and the remaining plasma samples after analysis were stored at-20 ℃ or-70 ℃ until LC/MS analysis. The evaluation of the accuracy of the quality control sample is carried out while analyzing the sample, and the accuracy of the quality control sample exceeding 66% is required to be between 80 and 120%. When plasma drug concentration-time curves were plotted, BLQ was scored as 0. When calculating the drug-induced parameters, the concentration before administration is calculated according to 0; BLQ (including "No peak") before Cmax is calculated as 0; BLQ (including "No peak") appearing after Cmax does not participate in the calculation uniformly. Pharmacokinetic parameters such as AUC (0-T), T1/2, Cmax, Tmax, MRT, etc. were calculated using WinNonlin from data on plasma concentrations of Compound 1 of the present invention at various time points.
The pharmacokinetic properties of the compounds of the invention were tested by the above assay.
Test results
TABLE 3 pharmacokinetic Activity of the Compounds of the invention
Figure 653434DEST_PATH_IMAGE015
And (4) conclusion: the blood concentration data of the compound in the blood at each time period show that the Cmax and AUC values of the compound in the body are higher, namely, the compound has better absorption rate and absorption degree in the body, higher content and longer half-life period, and is beneficial to the exertion of the in-vivo drug effect.
In conclusion, the compound realizes the remarkable progress of attenuation and synergy on the basis of BFA, and has further research value and wide development prospect.
Finally, it should be noted that there are other ways of implementing the invention. Accordingly, the embodiments of the present invention will be described by way of illustration, but not limitation to the description of the present invention, and modifications made within the scope of the present invention or equivalents added to the claims are possible. All publications or patents cited herein are incorporated by reference.

Claims (7)

1. A brefeldin A ester compound is characterized in that the brefeldin A ester compound is a compound shown in a formula (I) or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, an ester, a pharmaceutically acceptable salt or a prodrug thereof of the compound shown in the formula (I),
Figure DEST_PATH_IMAGE001
(I);
wherein:
z is a substituted or unsubstituted pyridine ring, quinoline ring or isoquinoline ring, wherein the pyridine ring, quinoline ring and isoquinoline ring may each independently optionally be substituted with 1,2,3 or 4 substituents selected from deuterium, F, Cl, Br, I, hydroxy, amino, cyano, nitro, C1-3Alkyl radical, C1-3Haloalkyl, C1-3Alkoxy radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, aryl, or 5-6 membered heteroaryl.
2. The compound of claim 1, wherein Z is a substituted or unsubstituted pyridine, quinoline or isoquinoline ring, wherein the pyridine, quinoline or isoquinoline ring may each be independently optionally substituted with 1,2,3 or 4 substituents selected from deuterium, F, Cl, Br, I, hydroxy, amino, cyano, nitro, methyl, ethyl, n-propyl, isopropyl, difluoromethyl, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, phenyl, thiazolyl, pyrazolyl, oxazolyl, pyridinyl or pyrimidinyl.
3. The compound of claim 1, having a structure of one of the following or a stereoisomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt, or a prodrug thereof:
Figure DEST_PATH_IMAGE003
Figure DEST_PATH_IMAGE005
4. a pharmaceutical composition comprising a compound according to any one of claims 1 to 3.
5. The pharmaceutical composition of claim 4, further comprising a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle, or combination thereof.
6. Use of a compound according to any one of claims 1 to 3 or a pharmaceutical composition according to any one of claims 4 to 5 for the manufacture of a medicament for the prevention or treatment of a hyperproliferative disease in a mammal, including a human.
7. The use of claim 6, wherein the hyperproliferative disease is liver cancer, leukemia, breast cancer, colon adenocarcinoma, stomach cancer, lung cancer, Bart's esophageal cancer, cervical cancer, pancreatic cancer, endometrial cancer, bone cancer, lymphatic cancer, kidney cancer, brain cancer, neural cancer, nasopharyngeal cancer, oral cancer, colorectal cancer.
CN202011327932.9A 2019-11-28 2020-11-24 Application of brefeldin A ester derivatives in antitumor drugs Active CN112851648B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019111887596 2019-11-28
CN201911188759 2019-11-28

Publications (2)

Publication Number Publication Date
CN112851648A true CN112851648A (en) 2021-05-28
CN112851648B CN112851648B (en) 2022-11-15

Family

ID=75996394

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011327932.9A Active CN112851648B (en) 2019-11-28 2020-11-24 Application of brefeldin A ester derivatives in antitumor drugs

Country Status (1)

Country Link
CN (1) CN112851648B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116178350A (en) * 2021-08-16 2023-05-30 中国海洋大学 Natural macrolide derivative, preparation method thereof and application of natural macrolide derivative as antitumor drug

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103788053A (en) * 2012-10-30 2014-05-14 浙江工业大学 Brefeldin A ester derivatives and their preparation method and use
CN105153136A (en) * 2015-09-16 2015-12-16 浙江工业大学 Brefeldin A ester derivatives, and preparation and application thereof
CN110028477A (en) * 2018-01-11 2019-07-19 沈阳药科大学 The preparation method and purposes of the 4- position split nitrogen mustard derivatives of a kind of brefeldin A

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103788053A (en) * 2012-10-30 2014-05-14 浙江工业大学 Brefeldin A ester derivatives and their preparation method and use
CN105153136A (en) * 2015-09-16 2015-12-16 浙江工业大学 Brefeldin A ester derivatives, and preparation and application thereof
CN110028477A (en) * 2018-01-11 2019-07-19 沈阳药科大学 The preparation method and purposes of the 4- position split nitrogen mustard derivatives of a kind of brefeldin A

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116178350A (en) * 2021-08-16 2023-05-30 中国海洋大学 Natural macrolide derivative, preparation method thereof and application of natural macrolide derivative as antitumor drug

Also Published As

Publication number Publication date
CN112851648B (en) 2022-11-15

Similar Documents

Publication Publication Date Title
CN108135912B (en) Pregnansterol and methods of use thereof
WO2019127008A1 (en) Compound for targeted degradation of btk and application thereof
EP2729448B1 (en) Compounds for the treatment of hiv
JP5827962B2 (en) Cdc7 kinase inhibitors and uses thereof
CN106589055B (en) Substituted cell acyl dipeptide compound and preparation method and application thereof
KR102634308B1 (en) Pyrrolopyrimidine compound
CN105339351A (en) Arginine methyltransferase inhibitors and uses thereof
WO2017101791A1 (en) Compound and use thereof in preparing drug
CN104918934A (en) 3-substituted pyrazoles and use as DLK inhibitors
WO2020188467A1 (en) Condensed tricyclic compound used as kinase inhibitor
CN111333648A (en) 8-substituted aromatic ring vinyl xanthine derivative and application thereof
CN106831614B (en) Substituted benzodiazacyclo compound and its preparation method and use
WO2020156189A1 (en) Camptothecin derivative and water-soluble prodrug thereof, pharmaceutical composition containing same, preparation method, and use
CN110612286A (en) Cyano-substituted fused bicyclic derivatives, their preparation and use
CN112851648B (en) Application of brefeldin A ester derivatives in antitumor drugs
CN112851647B (en) Brefeldin A derivative and preparation method and application thereof
CN116178350A (en) Natural macrolide derivative, preparation method thereof and application of natural macrolide derivative as antitumor drug
WO2023109751A1 (en) Pyrimidine or pyridine derivative and medicinal use thereof
CN107089955B (en) Sulfonamide derivative and preparation method and application thereof
CN112851622B (en) Macrolide brefeldin A ester derivative and application thereof
CN111072676B (en) Nitrogen-containing fused tricyclic derivatives and uses thereof
CN111018856B (en) 8-substituted styrylxanthine derivatives and uses thereof
CN112679497B (en) Azaindole amide compound and preparation method and application thereof
CN108864114B (en) Selectivity A2AReceptor antagonists
CN106243052B (en) Substituted heterocyclic compounds, process for their preparation and their use

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