CN113831338A - Histone deacetylase inhibitor and preparation method and application thereof - Google Patents

Histone deacetylase inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN113831338A
CN113831338A CN202111103830.3A CN202111103830A CN113831338A CN 113831338 A CN113831338 A CN 113831338A CN 202111103830 A CN202111103830 A CN 202111103830A CN 113831338 A CN113831338 A CN 113831338A
Authority
CN
China
Prior art keywords
compound
cancer
pharmaceutically acceptable
acceptable salt
acid
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
CN202111103830.3A
Other languages
Chinese (zh)
Other versions
CN113831338B (en
Inventor
李晓杨
江余祺
岳凯瑞
徐文方
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
Priority to CN202111103830.3A priority Critical patent/CN113831338B/en
Publication of CN113831338A publication Critical patent/CN113831338A/en
Application granted granted Critical
Publication of CN113831338B publication Critical patent/CN113831338B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C241/00Preparation of compounds containing chains of nitrogen atoms singly-bound to each other, e.g. hydrazines, triazanes
    • C07C241/04Preparation of hydrazides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • C07D217/26Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen

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 discloses a histone deacetylase inhibitor, and a preparation method and application thereof. Deacetylation of said histoneThe enzyme inhibitor is a compound which takes six-membered fused ring as a parent nucleus and hydrazide as a zinc ion chelating group and has a structure shown in the following general formula (I). The compound of the invention has obvious inhibition effect on Histone Deacetylase (HDAC) and Acute Myelogenous Leukemia (AML). Therefore, the compound of the invention can be used for preparing medicaments for treating diseases such as AML and the like related to abnormal expression of HDAC.

Description

Histone deacetylase inhibitor and preparation method and application thereof
Technical Field
The invention relates to the technical field of chemical synthetic drugs, and particularly relates to a hydrazide-containing histone deacetylase inhibitor, and a preparation method and application thereof.
Background
Histone Deacetylases (HDACs) participate in a variety of physiological reactions through histone deacetylation and non-histone lysine post-translational modification. Targeting HDACs has shown significant clinical effects, especially for cancer treatment, five HDAC inhibitors (HDACI) have been approved by the U.S. and chinese food and drug administration for the treatment of cutaneous T-cell lymphoma, peripheral T-cell lymphoma, and multiple myeloma (Journal of clinical chemistry,2020,63, 12460-. Class I HDAC inhibitors play a crucial role in the treatment of myeloid and lymphoid malignancies. Combined genetic and pharmacological deletions of HDAC1 and HDAC2 mediate pro-apoptotic responses in leukemia (Blood,2015,126, 2392-2403). Abnormal expression of HDAC3 is closely associated with the development of Leukemia and mediates resistance to chemotherapeutic drugs by modulating DNA damage repair in acute Leukemia (Leukemia,2017,31, 2761-2770). Previous studies have shown that class I HDAC inhibition can induce AML apoptosis by indirectly inhibiting the FLT3/STAT5 signaling pathway and down-regulating major anti-apoptotic proteins such as c-Flip and XIAP (Journal of medicinal chemistry,2020,63, 5501-.
Acute Myeloid Leukemia (AML) is a heterogeneous hematological disease characterized by uncontrolled expansion of immature malignant myeloid cells. AML is the most common form of leukemia in adults and is the most fatal of all leukemia types. Since 1973, chemotherapy for AML has been predominantly performed using the antimetabolites cytarabine and anthracycline antibiotics. Three-quarters of the patients with AML over the age of 60 are intolerant to large doses of chemotherapeutic drugs. In recent years, targeted drugs such as FLT3 inhibitor midodoxalin, IDH2 inhibitor imazalcitonin and Bcl-2 inhibitor venetoclax have been approved for the treatment of AML; however, their use is limited by the particular patient population and high drug resistance (Genes Chromosomes Cancer,2019,58, 903-. Under current treatment, the prognosis of elderly AML patients is very poor, with a 5-year relative survival rate of less than 10% (CA Cancer J Clin,2019,69, 363-385). Therefore, there is an urgent need for more effective and well-tolerated AML therapeutics. These studies above indicate that HDACs 1,2 and 3 are potential molecular targets for the treatment of AML.
Most of the currently marketed HDAC inhibitors lack subtype selectivity and use hydroxamic acid as a zinc ion chelating group, and such HDAC inhibitors have the disadvantages of low oral bioavailability, susceptibility to mutation, etc., so the research of novel zinc ion chelating groups is a research focus in the field (Recent Patents on Anti-Cancer Drug Discovery,2017, 12). In previous researches, it was found that the HDAC inhibitor using hydrazide as a zinc ion chelating group not only has stronger subtype selectivity, but also has good pharmacokinetic properties, and is a class of inhibitors with development potential (Journal of medicinal chemistry,2020,63, 5501-. However, the reported structure of the hydrazide HDAC inhibitor has Michael receptors, and the hydrazide HDAC inhibitor is easy to be off target in vivo to generate side effects. Therefore, the development of novel hydrazide HDAC inhibitors is of great significance.
Disclosure of Invention
In view of the above problems, a first object of the present invention is to provide a histone deacetylase inhibitor which has good activity against AML and exhibits excellent class I HDACs inhibitory action.
The second object of the present invention is to provide a method for preparing a histone deacetylase inhibitor compound, which can synthesize a desired compound with a low price, easily available raw materials, a high yield, and a stable property of the compound.
The third purpose of the invention is to provide the application of the compound in preparing class I HDACs inhibitors and AML-resistant medicines.
To develop potential Class I HDACs inhibitors and to treat AML and other diseases. On the basis of deep research on the defects of the current HDACs inhibitor, the invention designs and synthesizes a hydrazide compound taking naphthalene ring, quinoline, isoquinoline, quinazoline and other structures as a mother nucleus, and the hydrazide compound has obvious treatment effect in vitro experiments.
On one hand, the invention provides a histone deacetylase inhibitor which is characterized by being a hydrazide compound taking structures such as naphthalene ring, quinoline, isoquinoline and quinazoline as a parent nucleus and hydrazide as a zinc ion chelating group. The inhibitor is a compound with a structure shown as a general formula (I) or a pharmaceutically acceptable salt thereof:
Figure BDA0003271346360000021
wherein:
A. b and C are independently selected from CR2Or N; wherein R is2Selected from H, halogen, C1-C2Alkane, C1-C2Alkyl halide, amino, nitro, hydroxyl or cyano; said amino or hydroxy group being optionally substituted by 1-2C1-C2Alkoxy radical, C2-C3Alkynyloxy substituted.
The D ring is selected from:
Figure BDA0003271346360000022
wherein R is3The substituents may be one or more and may be in any position on the ring, selected from H, halogen, C1-C2Alkane, C1-C2Alkyl halide, amino, nitro, hydroxyl or cyano; said amino or hydroxy group being optionally substituted by 1-2C1-C2Alkyl radical, C2-C3And (3) alkynyl substitution.
L is selected from
Figure BDA0003271346360000031
Figure BDA0003271346360000032
Wherein R is4Selected from H, optionally substituted (C)1-C12) An alkyl group.
R1Is selected from C1-C6Alkane, C3-C6A cycloalkane.
The compound of the general formula I further has a structure shown as a general formula (II) or a general formula (III).
Figure BDA0003271346360000033
In the general formulas (II) and (III), L can be connected to para position or meta position of a benzene ring; in the general formula (III), E is a carbon atom.
In another aspect, the present invention also provides a process for the preparation of a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure BDA0003271346360000034
taking a compound 1 as a raw material, and reacting the compound with substituted or unsubstituted 4-aminomethyl methyl benzoate or 4-aminobenzoic acid methyl ester under the action of 1-ethyl- (3-dimethylaminopropyl) carbodiimide (EDCI) and 1-hydroxybenzotriazole (HOBt) to obtain an intermediate 2; the intermediate 2 reacts with hydrazine hydrate to obtain an intermediate 3; the intermediate 3 is further reacted with a substituted aldehyde to obtain the final target compound.
In addition, the invention also provides a pharmaceutical composition, which comprises the compound shown in the general formula (I), (II) or (III) or pharmaceutically acceptable salt thereof and pharmaceutically acceptable diluent or carrier; the content of the compound or the pharmaceutically acceptable salt thereof is 0.1-99.9 wt%.
Finally, the present invention provides the use of a compound of formula (I), (II) or (III) as described above, or a pharmaceutically acceptable salt thereof, for the manufacture of an HDAC inhibitor.
In another aspect, the present invention also provides the use of a compound of formula (I), (II) or (III) as described above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease associated with aberrant expression of Class I HDACs activity.
The diseases related to the abnormal expression of the activity of HDACs comprise tumors, neurodegenerative diseases, metabolic diseases, inflammatory reactions or heart diseases and the like.
The tumors include various types of leukemia, lymphoma, myeloma, triple negative breast cancer, lung cancer, melanoma, liver cancer, esophageal cancer, kidney cancer, oral cancer, tongue cancer, prostate cancer, breast cancer, cervical cancer, ovarian cancer, stomach cancer, pancreatic cancer, bladder cancer, colorectal cancer, nasopharyngeal cancer, brain tumor, glioma, anaplastic oligodendroglioma, adult glioblastoma, adult anaplastic astrocytoma, bone cancer, and soft tissue sarcoma.
In particular to the application of the compound of the general formula (I), (II) or (III) or the pharmaceutically acceptable salt thereof in preparing the medicine for treating Acute Myeloid Leukemia (AML).
The invention has the advantages that:
1. on the basis of the prior art, aiming at the defects of the existing HDAC inhibitor, the HDAC inhibitor of hydrazide class is optimized by using a method of computer-aided drug design, the HDAC inhibitor which takes naphthalene ring, quinoline, isoquinoline, quinazoline and other structures as a parent nucleus and takes hydrazide as a zinc ion chelating group is provided, and the compound has high subtype selectivity, HDAC targeting property and excellent pharmacokinetic property, and is an inhibitor with development potential.
2. The synthesis method provided by the invention has the following advantages: the compound can be obtained in high yield by using cheap and easily available raw materials through a three-step simple green synthesis method, and a new method is provided for the synthesis of the compound.
Detailed Description
The invention is further illustrated in detail below by means of specific examples, which are intended to facilitate a better understanding of the present invention without limiting its scope.
I. Compounds of the invention
A compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure BDA0003271346360000041
A. b and E are independently selected from CR2Or N; wherein R is2Selected from one or more of H, halogen, C1-C2Alkane, C1-C2Alkyl halide, amino, nitro, hydroxyl or cyano; said amino or hydroxy group being unsubstituted or optionally substituted by 1-2C1-C2Alkoxy radical, C2-C3Alkynyloxy substituted.
In some embodiments, 0-2 of A, B and E are selected from N.
D ring is selected from
Figure BDA0003271346360000042
Figure BDA0003271346360000043
Wherein R is3The substituents may be one or more and may be in any position on the ring, selected from H, halogen, C1-C2Alkane, C1-C2Alkyl halide, amino, nitro, hydroxyl or cyano; said amino or hydroxy group being optionally substituted by 1-2C1-C2Alkyl radical, C2-C3And (3) alkynyl substitution.
L is selected from
Figure BDA0003271346360000051
Figure BDA0003271346360000052
Wherein R is4Selected from H, optionally substituted (C)1-C12) An alkyl group.
In some embodiments, R4Is H. In some embodiments, R4Is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl or tert-butyl.
R1Is selected from C1-C6Alkane, C3-C6A cycloalkane.
In some embodiments, R1Is methyl, ethyl, propyl or butyl.
In some embodiments, R2Or R3Independently selected from one or more halogens, haloalkyl, (C)1-C2) Alkoxy, phenolic hydroxyl and cyano. In some embodiments, R2Or R3Selected from one or more halogens, haloalkyl groups, (C)1-C2) Alkoxy, phenolic hydroxyl. In some embodiments, R2Or R3Selected from one or more halogens, haloalkyl groups, (C)1-C2) An alkoxy group. In some embodiments, R2Or R3Selected from one or more halogens, haloalkyl groups. In some embodiments, R2Or R3The radicals are selected from one or more halogens. In some embodiments, R2Or R3Selected from one or more of F, Cl, Br or I. In some embodiments, R2Or R3Selected from haloalkyl groups. In some embodiments, R2Or R3Is haloalkyl, wherein the halogen atom is selected from F, Cl, Br or I. In some embodiments, R2Or R3Is a haloalkyl group in which the halogen atom is F. In some embodiments, R2Or R3Is a haloalkyl group in which the halogen atom is Cl. In some embodiments, R2Or R3Is a haloalkyl group in which the halogen atom is Br. In some embodiments, R2Or R3Is a haloalkyl group wherein the halogen atom is I. In some embodiments, R2Or R3is-CH2Cl、CH2F、-CH2Br、CHF2、CF3、-CFClBr、CH2CH2F、-CH2CH2Cl、CH2CHF2、CH2CF3、-CH2CCl3、CH2CH2CH2F、CH2CH2CHF2Or CH2CH2CF3And the like.
In some embodiments, the D ring is selected from the group consisting of
Figure BDA0003271346360000053
Figure BDA0003271346360000054
Figure BDA0003271346360000055
In some embodiments, the D ring is
Figure BDA0003271346360000056
A. B, C is CH, R1Is propyl, L is
Figure BDA0003271346360000057
The general formula (I) has the following structure:
Figure BDA0003271346360000061
in some embodiments, the D ring is
Figure BDA0003271346360000062
A and C are CH, B is N, R1Is propyl, L is
Figure BDA0003271346360000063
The general formula (I) has the following structure:
Figure BDA0003271346360000064
in some embodiments, the D ring is
Figure BDA0003271346360000065
A. B, C is CH, R1Is propyl, L is
Figure BDA0003271346360000066
The general formula (I) has the following structure:
Figure BDA0003271346360000067
in some embodiments, the D ring is
Figure BDA0003271346360000068
A and B are CH, C is N, R1Is propyl, L is
Figure BDA0003271346360000069
The general formula (I) has the following structure:
Figure BDA00032713463600000610
in some embodiments, the D ring is
Figure BDA00032713463600000611
A. B and C are CH, R1Is propyl, L is
Figure BDA00032713463600000612
The general formula (I) has the following structure:
Figure BDA00032713463600000613
d ring is
Figure BDA0003271346360000071
A. B and C are CH, R1Is propyl, L is
Figure BDA0003271346360000072
The general formula (I) has the following structure:
Figure BDA0003271346360000073
the invention also provides a compound of general formula (I) as described above, or a pharmaceutically acceptable salt thereof, for use as a medicament.
Preparation method II
A process for the preparation of a compound of formula (II) comprising the steps of:
Figure BDA0003271346360000074
in reaction I, the initial material carboxylic acid 1 is condensed with substituted or unsubstituted 4-aminomethyl methyl benzoate hydrochloride under the action of EDCI and HOBt to obtain an intermediate 2, and the reaction conditions are as follows: EDCI, HOBt, DCM, r.t.,6 h.
In reaction II, intermediate 2 reacts with hydrazine hydrate to obtain hydrazide key intermediate 3, the reaction conditions are as follows: hydrazine hydrate, MeOH, 80 ℃, 24 h.
In reaction III, the intermediate 3 is first reacted with an aldehyde to form a Schiff base, followed by NaBH3Reduction of CN to give a compound of formula (I) under the reaction conditions: aldehyde, MeOH, NaBH3CN,r.t.,4h。
Definition of
Prefix "Cx-Cy"means that subsequent groups have from x (e.g., 1) to y (e.g., 12) carbon atoms, in certain groups, one or more of which may be replaced by one or more heteroatoms or heteroatom groups. For example, "(C)1-C12) Alkyl "means that the alkyl has 1 to 12 carbon atoms. Similarly, the term "x-y membered" ring, where x and y are a range of values, e.g., "5-6 membered heterocyclic ring," refers to a ring containing x-y atoms (e.g., 5-6), such as N, O, S, with the remaining atoms being carbon.
"alkyl" refers to any group derived from a straight or branched chain saturated hydrocarbon. Alkyl groups include, but are not limited to, methyl, ethyl, propyl such as propan-1-yl, propan-2-yl (isopropyl), butyl such as butan-1-yl (n-butyl), butan-2-yl (sec-butyl), 2-methyl-propan-1-yl (isobutyl), 2-methyl-propan-2-yl (tert-butyl), pentyl, hexyl, octyl, decyl, and the like. Unless otherwise specified, an alkyl group has 1 to 12 carbon atoms, such as 1 to 8 carbon atoms, such as 1 to 6 carbon atoms, such as 1 to 4 carbon atoms, such as 1 to 2 carbon atoms.
"alkenyl" means a radical composed of at least two carbon atoms and at least one carbon-carbon double bond as defined aboveAlkyl of "C1-C12Alkenyl "refers to straight or branched chain alkenyl groups containing 1 to 12 carbons, including but not limited to ethenyl, 1-propenyl 5 alkenyl, 2-propenyl, 1-, 2-, or 3-butenyl, and the like, unless otherwise specified, alkenyl groups having 1 to 12 carbon atoms, e.g., 1 to 8 carbon atoms, e.g., 1 to 6 carbon atoms, e.g., 1 to 4 carbon atoms, e.g., 1 to 2 carbon atoms. The alkenyl group may be substituted or unsubstituted.
"haloalkyl" refers to an alkyl group wherein one or more hydrogen atoms are each replaced by a halogen. Examples include, but are not limited to-CH2Cl、CH2F、-CH2Br、CHF2、CF3、-CFClBr、CH2CH2F、-CH2CH2Cl、CH2CHF2、CH2CF3、-CH2CCl3、CH2CH2CH2F、CH2CH2CHF2、CH2CH2CF3And the like, and alkyl groups such as perfluoroalkyl groups in which all hydrogen atoms are substituted with fluorine atoms.
"alkoxy" refers to a moiety of the formula-O-alkyl, wherein the alkyl moiety is as defined above. E.g. C1-C2Alkoxy refers to an alkyl moiety having 1-2 carbon atoms attached to an oxygen. "haloalkoxy" refers to a moiety of the formula-O-haloalkyl, wherein the haloalkyl moiety is as defined above. For example, (C)1-C2) Alkoxy refers to a moiety having 1-2 carbon haloalkyl groups attached to oxygen.
"plurality" independently means 1,2,3, 4, 5, 6, 7, 8,9, or 10.
"optional" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not occur. For example, "optionally substituted with one or more substituents" means that the substituents may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with a substituent and the case where the heterocyclic group is not substituted with a substituent.
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort. For example: amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds.
It will be appreciated by those skilled in the art that salts, including pharmaceutically acceptable salts, of the compounds of formula (I) may be prepared. These salts may be prepared in situ during the final isolation and purification of the compound or by separately reacting the purified compound in its free acid or free base form with a suitable base or acid, respectively.
Pharmaceutically acceptable acid addition salts may be formed with inorganic and organic acids, for example, acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, citrate, edisylate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, stearate, oleate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, dihydrogenphosphate, dihydrogensulfonate, and the like, Polygalacturonate, propionate, stearate, succinate, sulfosalicylate, tartrate, tosylate, or trifluoroacetate.
Inorganic acids that can form salts include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be formed include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like. Pharmaceutically acceptable base addition salts may be formed with inorganic or organic bases.
Inorganic bases which can form salts include, for example, ammonium salts and metals of groups I to XII of the periodic Table of the elements. In certain embodiments, the salt is derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, or copper; particularly suitable salts include ammonium, potassium, sodium, calcium or magnesium salts.
Organic bases from which salts can be formed include, for example, primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Certain organic amines include isopropylamine, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from basic or acidic moieties by conventional chemical methods. In general, these salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (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. These reactions are generally carried out in water or in an organic solvent, or in a mixture of the two. Generally, where appropriate, it is desirable to use a non-aqueous medium, such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. Other suitable salts may be listed in "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "Handbook of Pharmaceutical Salts" by Stahl and Wermuth: properties, Selection, and Use "(Wiley-VCH, Weinheim, Germany, 2002).
Solvates of the compounds of formula (I), including pharmaceutically acceptable solvates, may also be prepared. "solvate" refers to a complex of variable chemical amounts formed by a solute and a solvent. Such solvents for the purposes of the present invention do not affect the biological activity of the solute. Examples of suitable solvents include, but are not limited to, water, MeOH, EtOH, or AcOH. Solvates wherein water is the solvent molecule are generally referred to as hydrates. Hydrates include compositions that include a stoichiometric amount of water, as well as compositions that include variable amounts of water.
As used herein, the term "pharmaceutically acceptable" means a compound suitable for pharmaceutical use. Salts and solvates (e.g., hydrates and hydrates of salts) of the compounds of the invention suitable for use in medicine are those in which the counterion or bound solvent is pharmaceutically acceptable. However, salts and solvates with non-pharmaceutically acceptable counterions or bound solvents are also included within the scope of the invention, e.g., as intermediates in the preparation of other compounds of the invention and pharmaceutically acceptable salts and solvates thereof.
The compounds of formula (I), including salts and solvates thereof, may be present in crystalline form, non-crystalline form or mixtures thereof. The compounds, or salts or solvates thereof, may also exhibit polymorphism, i.e. the ability to occur in different crystalline forms. These different crystalline forms are generally known as "polymorphs". Polymorphs have the same chemical composition, but differ in the packing, geometric arrangement and other descriptive properties of the crystalline solid state. Thus, polymorphs can have different physical properties, such as shape, density, hardness, deformability, stability, and solubility properties. Polymorphs typically exhibit different melting points, IR spectra and X-ray powder diffraction patterns, all of which can be used for identification. It will be appreciated by those skilled in the art that different polymorphs may be produced, for example, by varying or adjusting the conditions used in the crystallization/recrystallization of the compound of formula (I).
The invention also encompasses different isomers of the compounds of formula (I). "isomers" refer to compounds having the same composition and molecular weight, but differing in physical and/or chemical properties. The structural differences can be in the structure (geometric isomers) or in the ability to rotate plane polarized light (stereoisomers). With respect to stereoisomers, the compounds of formula (I) may have one or more asymmetric carbon atoms and may occur as racemates, racemic mixtures and as single enantiomers or diastereomers. All such isomeric forms are included within the scope of the present invention, including mixtures thereof. If the compound contains a double bond, the substituent may be in the E or Z configuration. If the compound comprises a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may have a cis-or trans-configuration. It is also intended to include all tautomeric forms.
Any asymmetric atom (e.g., carbon, etc.) of the compounds of formula (I) can exist in racemic or enantiomerically enriched form, e.g., in 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. If possible, the substituents on the atoms having an unsaturated double bond are present in cis- (Z) -or trans- (E) -form.
Thus, as used herein, a compound of general formula (I) can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example as substantially pure geometric isomers (cis or trans), diastereomers, optical isomers (enantiomers), racemates or mixtures thereof.
Any resulting mixture of isomers can be separated into pure or substantially pure geometric or optical isomers, diastereomers, racemates based on the physicochemical differences 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, for example by separation of diastereomeric salts thereof, which are obtained with an optically active acid or base and release the optically active acidic or basic compound. In particular, basic moieties may therefore be used to resolve the compounds of the invention into their optical enantiomers, for example by fractional crystallisation of a salt with an optically active acid (e.g. tartaric acid, dibenzoyltartaric acid, diacetyltartaric acid, di-O, O' -p-toluoyltartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid). The racemic product can also be resolved by chiral chromatography, such as High Performance Liquid Chromatography (HPLC) using a chiral adsorbent.
The invention includes unlabeled as well as isotopically labeled forms of the compounds of formula (I). Isotopically labeled 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. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as2H、3H、11C、13C、14C、15N、18F、31P、32P、35S、36Cl、125I. The invention includes various isotopically-labeled compounds as defined herein, for example, in which a radioisotope is present (e.g., in which a radioisotope is present3H and14C) or in which non-radioactive isotopes occur (e.g. in2H and13C) those of (a). These isotopically-labeled compounds are useful in metabolic studies (e.g., using14C) Reaction kinetics study (e.g. with2H or3H) Detection or imaging techniques, such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT), including drug substrate tissue distribution analysis, or for radiotherapy of a patient. In particular, it may be particularly desirable for PET or SPECT studies18F or a labeled compound. Isotopically-labelled compounds of general formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying examples and preparations using a suitable isotopically-labelled reagent in place of the unlabelled reagent previously used.
In addition, heavier isotopes, in particular deuterium (i.e. deuterium)2H or D)) may lead to certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements or improvement in therapeutic index. It is understood that deuterium is considered herein as a substituent of the compound of formula (I). The concentration of this heavier isotope, in particular deuterium, may be determined by the isotopic enrichment factor. As herein describedThe term "isotopic enrichment factor" is used to refer to the ratio between the isotopic and natural abundance of a particular isotope. If a substituent in a compound of the invention is designated as deuterium, then for each designated deuterium atom the compound has an isotopic enrichment factor 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).
The person skilled in the art will be able to identify the presence or absence of a stereocenter in the compound of formula (I). Thus, the present invention includes possible stereoisomers, and includes both racemic compounds and individual enantiomers. When the desired compound is a single enantiomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate. Resolution of the final product, intermediate or starting material may be achieved by any suitable method known in the art. See, e.g., E.L.Eliel, S.H.Wilen and L.N.Mander, "Stereochemistry of Organic Compounds" (Wiley-Intererscience, 1994).
Pharmaceutical compositions
The present invention provides a pharmaceutical composition comprising a compound of general formula (I) as described above, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier.
The pharmaceutical compositions can be formulated for a particular route of administration, such as oral, parenteral, and rectal administration. Furthermore, the pharmaceutical compositions of the present invention can be formulated in solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories) or in liquid form (including without limitation solutions, suspensions or emulsions). The pharmaceutical compositions can be subjected to conventional pharmaceutical operations (e.g., sterilization) and/or can contain conventional inert diluents, lubricating agents or buffers, as well as adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers, and the like.
Typically, the pharmaceutical composition is a tablet or capsule comprising the active ingredient and:
a) diluents, such as lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, glycine and the like;
b) lubricants, for example silica, talc, stearic acid, its magnesium or calcium salts and/or polyethylene glycol;
also included for the tablets are:
c) binders, such as magnesium aluminum silicate, starch paste, gelatin, gum tragacanth, methyl cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone; if desired, also
d) Disintegrating agents, such as starch, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or
e) Absorbents, coloring agents, flavoring agents, and sweeteners.
The tablets may be film-coated or enteric-coated according to methods known in the art.
Suitable compositions for oral administration include an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof, in the form of tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions for oral use are prepared according to any method known to the art for the manufacture of pharmaceutical compositions and can contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide a fine and palatable preparation. Tablets may contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. Such excipients are, for example, inert diluents (e.g. calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate); granulating and disintegrating agents (e.g., corn starch, or alginic acid); binding agents (e.g., starch, gelatin, or acacia); and lubricating agents (e.g., magnesium stearate, stearic acid, or talc). Tablets are uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. Formulations for oral use may be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
Certain injectable compositions are isotonic aqueous solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, dissolution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, it may also contain other therapeutically valuable substances. The compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1-75% or about 1-50% of the active ingredient.
Since water may promote the degradation of certain compounds, the present invention also provides anhydrous pharmaceutical compositions and dosage forms comprising a compound of the present invention as an active ingredient.
Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low water content ingredients and low water content or low humidity conditions. Anhydrous pharmaceutical compositions can be prepared and stored so as to retain their anhydrous nature. Thus, anhydrous compositions are packaged using materials known to prevent contact with water so that they can be included in suitable kits of formulations. Examples of suitable packaging include, without limitation, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
The invention further provides pharmaceutical compositions and dosage forms comprising 1 or more agents that reduce the rate of decomposition of a compound of the invention as an active ingredient. Such agents (which are referred to herein as "stabilizers") include, without limitation, antioxidants (e.g., ascorbic acid), pH buffers or salt buffers, and the like.
The pharmaceutical composition or combination product of the invention can be a unit dose of about 1-1000mg of active ingredient, or about 1-500mg or about 1-250mg or about 1-150mg or about 0.5-100mg, or about 1-50mg of active ingredient for about 50-70kg of an individual. The therapeutically effective dose of the compound, pharmaceutical composition or combination thereof will depend on the species, weight, age and condition of the individual, the condition or 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 required to prevent, treat or inhibit the development of the condition or disease.
Therapeutic use
In some embodiments, there is provided the use of a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, in the preparation of an hdac inhibitor.
In some embodiments, there is provided the use of a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease associated with aberrant expression of HDAC activity.
In some embodiments, the disease associated with aberrant expression of HDAC activity comprises dysplasia, tumors, neurodegenerative diseases, metabolic diseases, inflammatory reactions, or heart diseases, among others, wherein the tumors comprise various leukemias, lymphomas, myelomas, triple negative breast cancer, lung cancer, melanoma, esophageal cancer, prostate cancer, breast cancer, and others.
In some embodiments, there is provided a method of treating or preventing a disease associated with aberrant expression of HDACs activity, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
In some embodiments, the method comprises administering a compound of formula (I), or a pharmaceutically acceptable salt thereof, in combination with one, two, three, or four additional therapeutic agents.
Herein, if the term "combination" is used to describe a combined administration, it is to be understood that this may mean simultaneous administration, separate administration or sequential administration. In one aspect of the invention, "combined administration" refers to simultaneous administration. In another aspect of the invention, "combined administration" refers to independent administration. In another aspect of the invention, "combined administration" refers to sequential administration. The delay in administering the second component should not, for example, lose the benefit of the effect produced by the use of the combination when administered sequentially or separately.
The invention is further illustrated by the following specific preparation examples and application examples.
The numbers, names and structures of the compounds in the examples are shown in the following table.
Figure BDA0003271346360000131
Figure BDA0003271346360000141
Example 1: n- (4- (2-propylhydrazine-1-carbonyl) benzyl) -2-naphthamide (I-01)
Figure BDA0003271346360000142
The specific synthesis steps are as follows:
a. synthesis of compound 1[4- ((2-naphthamide) methyl benzoate ]
2-naphthoic acid (345mg,2mmol) was placed in a 100mL eggplant-shaped bottle at room temperature, and dissolved by adding 30mL of methylene chloride. 1-Ethyl- (3-dimethylaminopropyl) carbonyldiimine hydrochloride (EDC. HCl) (460mg,2.4mmol), 1-hydroxybenzotriazole (HOBt) (325mg,2.4mmol) and triethylamine (506mg,5mmol) were added under ice-bath and reacted for 0.5 hour. After addition of methyl 4-aminomethylbenzoate hydrochloride (483mg,2.4mmol), the reaction was allowed to proceed overnight at room temperature. TLC detection of the starting material reaction completion, subsequent washing of the reaction with water (20 mL. times.3) and application of anhydrous Na to the organic phase2SO4And (5) drying. Column chromatography gave compound 1, (415mg, 65% yield) as a white solid, which was used directly in the next synthesis. ESI-MS M/z 319.92[ M + H ]]+
b. Synthesis of compound 2[ N- (4- (hydrazinecarbonyl) benzyl) -2-naphthamide ]
At room temperature, reacting 4- ((2-naphthamide)) Methyl) benzoate (319mg,1mmol) was placed in a 100mL eggplant-shaped flask and dissolved by adding 30mL of methanol. Hydrazine hydrate (840mg,15mmol) was added and refluxed for 24 h. TLC detection of the starting material reaction was complete and after evaporation of the methanol crude 2(305mg, 97% yield) was obtained and used directly in the next reaction. ESI-MS M/z 319.94[ M + H ]]+
c. EXAMPLE 1 Synthesis of [ N- (4- (2-propylhydrazine-1-carbonyl) benzyl) -2-naphthamide ]
N- (4- (hydrazinecarbonyl) benzyl) -2-naphthamide (250mg,0.8mmol) was placed in a 100mL eggplant-shaped flask at room temperature, dissolved by adding 30mL of methanol, and propionaldehyde (58mg,1mmol) was added. After 2 hours of reaction, NaBH was added3CN (250mg,4mmol) and acidified with HCl in methanol and reacted for 6 hours. TLC detection of the reaction completion of the starting material, addition of 20mL of saturated sodium bicarbonate solution, reaction for 0.5 hour, evaporation of methanol, extraction with dichloromethane (20 mL. times.3), and combination of the organic phases with anhydrous Na2SO4And (5) drying. Separation by column chromatography gave compound I-01 (185mg, 64% yield).
1H NMR(400MHz,DMSO-d6)δ9.94(d,J=5.4Hz,1H),9.24(t,J=6.0Hz,1H),8.47(s,1H),8.02-7.92(m,4H),7.76(d,J=8.3Hz,2H),7.62-7.53(m,2H),7.39(d,J=8.3Hz,2H),5.04(d,J=6.4Hz,1H),4.54(d,J=5.9Hz,2H),2.70(q,J=6.9,6.4Hz,2H),1.48-1.39(m,2H),0.86(t,J=7.4Hz,3H);13C NMR(101MHz,DMSO-d6)δ167.74,167.01,142.51,134.92,132.67,131.33,129.03,128.68,127.99,127.92,127.87,127.70,127.41,126.96,123.63,54.21,43.76,21.29,11.64;HRMS(ESI):m/z calcd for C22H22N3O2:360.17175[M-H]-;found:360.17194。
Example 2: n- (4- (2-propylhydrazine-1-carbonyl) benzyl) quinoline-3-carboxamide (I-02)
Figure BDA0003271346360000151
The specific synthesis steps are as follows:
the same as in example 1 except that 2-naphthoic acid in example 1 was replaced with quinoline-3-carboxylic acid. The total yield was 37%.
1H NMR(400MHz,DMSO-d6)δ9.96(s,1H),9.43(t,J=5.9Hz,1H),9.29(d,J=2.3Hz,1H),8.85(d,J=1.4Hz,1H),8.11-8.02(m,2H),7.89-7.79(m,1H),7.81-7.72(m,2H),7.72-7.62(m,1H),7.41(d,J=8.4Hz,2H),5.05(s,1H),4.56(d,J=5.9Hz,2H),2.70(t,J=7.1Hz,2H),1.47-1.41(m,2H),0.86(t,J=7.4Hz,3H);13C NMR(101MHz,DMSO-d6)δ165.65,165.50,149.45,149.05,143.18,136.20,132.39,131.79,129.70,129.31,127.99,127.69,127.38,127.08,53.61,43.04,21.38,12.21;HRMS(ESI):m/z calcd for C21H21N4O2:361.16700[M-H]-;found:361.16714。
Example 3: n- (4- (2-propylhydrazine-1-carbonyl) benzyl) isoquinoline-3-carboxamide (I-03)
Figure BDA0003271346360000161
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 3-isoquinolinecarboxylic acid. The total yield was 35%.
1H NMR(400MHz,DMSO-d6)δ9.97(s,1H),9.56(t,J=6.5Hz,1H),9.40(s,1H),8.58(s,1H),8.26(d,J=8.1Hz,1H),8.20(d,J=8.3Hz,1H),7.91-7.85(m,1H),7.84-7.79(m,1H),7.77(d,J=8.3Hz,2H),7.44-7.39(m,2H),5.11(s,1H),4.60(d,J=6.4Hz,2H),2.73(t,J=7.1Hz,2H),1.49-1.39(m,2H),0.89(t,J=7.4Hz,3H);13C NMR(101MHz,DMSO-d6)δ165.70,164.89,152.16,144.25,143.57,135.94,132.27,131.93,129.81,129.70,128.55,128.37,127.69,127.61,120.45,53.61,42.79,21.38,12.21;HRMS(ESI):m/z calcd for C21H21N4O2:361.16700[M-H]-;found:361.16696。
Example 4: n- (4- (2-propylhydrazine-1-carbonyl) benzyl) quinoline-2-carboxamide (I-04)
Figure BDA0003271346360000162
The specific synthesis steps are as follows:
the same as in example 1 except that 2-naphthoic acid in example 1 was replaced with 2-quinolinecarboxylic acid. The total yield was 41%.
1H NMR(400MHz,DMSO-d6)δ9.94(s,1H),9.53(t,J=6.4Hz,1H),8.54(d,J=8.6Hz,1H),8.13(d,J=8.6Hz,1H),8.11(d,J=8.4Hz,1H),8.06(d,J=8.1Hz,1H),7.87-7.82(m,1H),7.74(d,J=8.3Hz,2H),7.72-7.66(m,1H),7.39(d,J=8.3Hz,2H),5.04(s,1H),4.57(d,J=6.4Hz,2H),2.69(t,J=7.1Hz,2H),1.46-1.37(m,2H),0.86(t,J=7.5Hz,3H);13C NMR(101MHz,DMSO-d6)δ165.68,164.80,150.60,146.58,143.37,138.48,132.34,131.12,129.72,129.40,128.68,127.76,127.64,119.29,53.61,42.87,21.38,12.20;HRMS(ESI):m/z calcd for C21H21N4O2:361.16700[M-H]-;found:361.16684。
Example 5: n- (4- (2-propylhydrazine-1-carbonyl) benzyl) isoquinoline-6-carboxamide (I-05)
Figure BDA0003271346360000163
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 6-isoquinolinecarboxylic acid. The total yield was 37%.
1H NMR(400MHz,DMSO-d6)δ10.03-9.92(m,1H),9.40(d,J=5.1Hz,2H),8.59(d,J=5.7Hz,1H),8.52(d,J=1.6Hz,1H),8.23(d,J=8.6Hz,1H),8.12(dd,J=8.6,1.7Hz,1H),7.95(d,J=5.7Hz,1H),7.85-7.76(m,2H),7.44(d,J=8.2Hz,2H),5.08(s,1H),4.59(d,J=5.9Hz,2H),2.75(t,J=7.3,2H),1.52-1.44(m,2H),0.91(t,J=7.4Hz,3H);13C NMR(101MHz,DMSO-d6)δ166.41,165.61,152.80,144.09,143.19,136.29,135.09,132.35,129.41,128.41,127.61,126.55,126.20,121.61,53.58,43.10,21.32,12.13;HRMS(ESI):m/z calcd for C21H21N4O2:361.16700[M-H]-;found:361.16711。
Example 6: n- (4- (2-propylhydrazine-1-carbonyl) benzyl) isoquinoline-7-carboxamide (I-06)
Figure BDA0003271346360000171
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 7-isoquinolinecarboxylic acid. The total yield was 38%.
1H NMR(400MHz,DMSO-d6)δ9.91(s,1H),9.35(s,1H),9.30(t,J=6.0Hz,1H),8.66-8.60(m,1H),8.52(d,J=5.7Hz,1H),8.16(dd,J=8.6,1.8Hz,1H),8.00(d,J=8.6Hz,1H),7.82(d,J=5.7Hz,1H),7.77-7.68(m,2H),7.42-7.32(m,2H),5.01(s,1H),4.53(d,J=5.9Hz,2H),2.68(t,J=7.1Hz,2H),1.44-1.37(m,2H),0.83(t,J=7.4Hz,3H);13C NMR(101MHz,DMSO-d6)δ166.23,165.62,153.85,144.77,143.27,136.88,133.53,132.34,129.15,127.97,127.88,127.61,127.26,120.68,53.58,43.09,21.32,12.13;HRMS(ESI):m/z calcd for C21H21N4O2:361.16700[M-H]-;found:361.16705。
Example 7: n- (4- (2-propylhydrazine-1-carbonyl) benzyl) -1, 6-naphthyridine-2-carboxamide (I-07)
Figure BDA0003271346360000172
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 1, 6-naphthyridine-2-carboxylic acid. The total yield was 35%.
1H NMR(400MHz,DMSO-d6)δ9.96(s,1H),9.67(t,J=6.4Hz,1H),9.49(s,1H),8.78(dd,J=19.2,7.2Hz,2H),8.27(d,J=8.5Hz,1H),7.97(d,J=6.0Hz,1H),7.74(d,J=8.1Hz,2H),7.39(d,J=8.1Hz,2H),4.57(d,J=6.4Hz,2H),2.69(t,J=7.1Hz,2H),1.48-1.37(m,2H),0.85(t,J=7.4Hz,3H);13C NMR(101MHz,DMSO-d6)δ165.68,164.23,154.64,154.12,148.82,147.99,143.13,138.86,132.37,127.78,127.65,124.49,122.13,120.97,53.60,42.97,21.36,12.20;HRMS(ESI):m/z calcd for C20H20N5O2:362.16225[M-H]-;found:362.16214。
Example 8: 6-chloro-N- (4- (2-propylhydrazine-1-carbonyl) benzyl) -2-naphthamide (I-08)
Figure BDA0003271346360000181
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 6-chloro-2-naphthoic acid. The total yield was 41%.
1H NMR(500MHz,DMSO-d6)δ9.98(s,1H),9.31(t,J=6.0Hz,1H),8.54(d,J=1.6Hz,1H),8.12(d,J=2.2Hz,1H),8.07(d,J=8.8Hz,1H),8.06–7.97(m,2H),7.82-7.77(m,2H),7.60(dd,J=8.7,2.2Hz,1H),7.42(d,J=8.3Hz,2H),5.10(s,1H),4.57(d,J=5.9Hz,2H),2.74(t,J=7.1Hz,2H),1.51-1.40(m,2H),0.89(t,J=7.4Hz,4H);13C NMR(126MHz,DMSO-d6)δ166.53,165.59,143.36,135.29,132.66,132.55,132.26,131.97,131.52,131.03,129.12,128.04,127.76,127.57,127.55,126.78,125.85,53.56,43.01,21.31,12.12;HRMS(ESI):m/z calcd for C22H21N3O2Cl:394.12641[M-H]-;found:394.12633。
Example 9: 6-bromo-N- (4- (2-propylhydrazine-1-carbonyl) benzyl) -2-naphthamide (I-09)
Figure BDA0003271346360000182
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 6-bromo-2-naphthoic acid. The total yield thereof was found to be 39%.
1H NMR(500MHz,DMSO-d6)δ9.98(s,1H),9.35(t,J=6.0Hz,1H),8.54(d,J=1.7Hz,1H),8.28(d,J=2.0Hz,1H),8.03(dd,J=8.7,1.7Hz,1H),8.02-7.96(m,2H),7.79(d,J=8.4Hz,2H),7.74-7.67(m,1H),7.42(d,J=8.4Hz,2H),5.08(s,1H),4.56(d,J=5.9Hz,2H),2.73(t,J=7.1Hz,2H),1.511.38(m,2H),0.89(t,J=7.4Hz,4H);13C NMR(126MHz,DMSO-d6)δ166.53,165.58,143.37,135.73,132.62,132.25,131.97,131.53,131.18,130.23,130.05,129.12,128.12,127.67,127.57,127.55,125.82,121.44,53.56,43.01,21.30,12.12;HRMS(ESI):m/z calcd for C22H21N3O2Br:438.15674[M-H]-;found:438.15667。
Example 10: 6-methoxy-N- (4- (2-propylhydrazine-1-carbonyl) benzyl) -2-naphthamide (I-10)
Figure BDA0003271346360000191
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 6-methoxy-2-naphthoic acid. The total yield was 42%.
1H NMR(500MHz,DMSO-d6)δ9.97(s,1H),9.16(t,J=6.0Hz,1H),8.43(d,J=1.8Hz,1H),7.97-7.90(m,2H),7.88(d,J=8.6Hz,1H),7.79(d,J=8.4Hz,2H),7.41(d,J=8.2Hz,2H),7.38(d,J=2.6Hz,1H),7.23(dd,J=9.0,2.6Hz,1H),5.08(s,1H),4.56(d,J=5.9Hz,2H),3.90(s,3H),2.74(t,J=7.1Hz,2H),1.51-1.40(m,2H),0.90(t,J=7.4Hz,3H);13C NMR(126MHz,DMSO-d6)δ166.86,165.61,159.02,143.58,136.28,132.21,130.90,129.75,127.96,127.90,127.55,127.52,127.16,125.15,119.84,106.33,55.78,53.57,42.95,21.31,12.12;HRMS(ESI):m/z calcd for C23H24N3O3:390.16959[M-H]-;found:390.16952。
Example 11: 6-hydroxy-N- (4- (2-propylhydrazine-1-carbonyl) benzyl) -2-naphthamide (I-11)
Figure BDA0003271346360000192
The specific synthesis steps are as follows:
the same procedure as in example 1 was repeated except that 2-naphthoic acid in example 1 was replaced with 6-hydroxy-2-naphthoic acid. The total yield was 34%.
1H NMR(500MHz,DMSO-d6)δ9.97(s,1H),9.12(t,J=5.9Hz,1H),8.38(d,J=1.8Hz,1H),7.90-7.83(m,2H),7.78(d,J=8.3Hz,2H),7.74(d,J=8.7Hz,1H),7.41(d,J=8.1Hz,2H),7.19-7.11(m,2H),4.55(d,J=6.0Hz,2H),2.74(t,J=7.1Hz,2H),1.50-1.38(m,2H),0.89(t,J=7.4Hz,3H);13C NMR(126MHz,DMSO-d6)δ166.93,157.39,143.66,136.55,131.07,128.81,128.79,128.04,128.01,127.55,127.49,127.11,126.45,124.84,119.91,109.07,53.56,42.91,21.31,12.12;HRMS(ESI):m/z calcd for C22H22N3O3:376.18652[M-H]-;found:376.18643。
Example 12: HDAC1,2,3,6 inhibitory Activity in vitro and cell proliferation inhibitory Activity
1. HDAC1,2,3,6 in vitro inhibitory activity assay methods:
50 μ L of HDAC buffer containing the drug was mixed with 10 μ L of the enzyme solution and incubated for 5min in advance, 40 μ L of the substrate was added, and then the reaction was carried out at 37 ℃ for 30min, and then 100 μ L of trypsin stop buffer was added to stop the reaction, and the reaction was carried out at 37 ℃ for 20min, and the fluorescence intensity was measured at 390nm/460 nm.
Figure BDA0003271346360000201
Finally, the inhibition rate (%) of the compound and the corresponding concentration are subjected to S-curve fitting, and IC is calculated50The value is obtained.
2. Cell proliferation inhibitory activity assay method:
the test of the in vitro cell proliferation inhibiting activity of the target compound adopts a resazurin method. The human leukemia cell MV4-11 cell line is adopted and all the cells are cultured conventionally. Cells in logarithmic growth phase were used for all experiments. The cell suspension of the above cells was counted under an inverted microscope to count the number of cells, and the number of cells was adjusted to 1X 105/mL by adding a medium. Taking 96-well cellsThe culture plate is used for cell inoculation and drug experiments, the peripheral holes are not used (filled with sterile PBS), a blank control group, a negative control group, a positive control group and a drug experiment group are set, wherein the blank control group is only added with 150 mu L/hole of cell culture solution, the negative control group is only added with 100 mu L/hole of cell suspension and is added with 50 mu L/hole of cell culture solution, the positive control group is only added with 100 mu L/hole of cell suspension and is added with 50 mu L/hole of positive control drug solution, the drug experiment group is added with 100 mu L/hole of cell suspension and is added with 50 mu L/hole of test compound solution, the positive control group and the drug experiment group are respectively set with 8 different drug final concentrations of 0.03, 0.1, 0.3, 1, 3, 10, 30, 100 mu mol.L-1Each drug concentration is provided with 3 parallel multiple holes. After the drug addition was complete, 96-well cell culture plates were incubated at 37 ℃ with 5% CO2And incubated under saturated humidity for 72h, 10. mu.L of Resazurin (1mg/mL) was added to each well, and after further incubation for 3h, the fluorescence was measured at ex.560/em.590nm using a microplate reader.
3. The experimental results are as follows:
the results of the inhibitory activity are shown in Table 1 below, and the experimental results show that the compounds have a low nanomolar inhibitory IC on both HDAC1 and 250The inhibition effect on HDAC3 is significant, and is about 1nM, wherein the compound with the best inhibition activity is I-01, and the IC on HDAC1,2 and 3507.98, 30.51 and 0.89nM, respectively; and all compounds have no obvious activity on HDAC6, which indicates that the compounds have HDAC subtype selectivity.
IC of Compound I-01 having the best inhibitory Activity in cell line MV4-11 of AML5043.59nM, the remaining compounds showed low nanomolar inhibitory IC50Values (40-75 nM). Therefore, the compound provided by the invention has remarkable class I HDACs inhibitory activity and AML (AML resistant activity).
TABLE 1 IC of examples 1-11 on HDAC1,2,3,6 and MV4-11 cells50Value of
Figure BDA0003271346360000202
Figure BDA0003271346360000211
It should be noted that the above-mentioned embodiments are only preferred embodiments of the present invention, and it should be noted that, for those skilled in the art, several modifications and decorations can be made without departing from the inventive concept, and these modifications and decorations should not be excluded from the scope of the present invention.

Claims (10)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure FDA0003271346350000011
wherein:
A. b and E are independently selected from CR2Or N; wherein R is2Selected from H, halogen, C1-C2Alkane, C1-C2Alkyl halide, amino, nitro, hydroxyl or cyano; said amino or hydroxy group being unsubstituted or optionally substituted by 1-2C1-C2Alkoxy radical, C2-C3Alkynyloxy substitution;
the D ring is selected from:
Figure FDA0003271346350000012
Figure FDA0003271346350000013
wherein R is3The substituents are one or more and have an indefinite position on the ring, and are selected from H, halogen, C1-C2Alkane, C1-C2Alkyl halide, amino, nitro, hydroxyl or cyano; said amino or hydroxy group being optionally substituted by 1-2C1-C2Alkyl radical, C2-C3Alkynyl substitution;
l is selected from
Figure FDA0003271346350000014
Figure FDA0003271346350000015
Wherein R is4Selected from H, optionally substituted C1-C12An alkyl group;
R1is selected from C1-C6Alkane, C3-C6A cycloalkane.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) has a structure according to formula (ii):
Figure FDA0003271346350000016
wherein L is attached to the para or meta position of the benzamide group.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) has a structure of formula (iii):
Figure FDA0003271346350000021
wherein L is attached to the para-or meta-position of the benzamide group and E is a carbon atom.
4. A process for the preparation of a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure FDA0003271346350000022
taking a compound 1 as a raw material, and reacting the compound with substituted or unsubstituted 4-aminomethyl methyl benzoate or 4-aminobenzoic acid methyl ester under the action of 1-ethyl- (3-dimethylaminopropyl) carbodiimide and 1-hydroxybenzotriazole to obtain an intermediate 2; the intermediate 2 reacts with hydrazine hydrate to obtain an intermediate 3; the intermediate 3 is further reacted with a substituted aldehyde to obtain the final target compound.
5. A pharmaceutical composition comprising a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable diluent or carrier.
6. The pharmaceutical composition of claim 5, wherein the compound or pharmaceutically acceptable salt thereof is present in an amount of 0.1 to 99.9 wt%.
7. Use of a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, in the manufacture of a histone deacetylase inhibitor.
8. Use of a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease associated with aberrant expression of histone deacetylase activity.
9. Use according to claim 8, characterized in that the diseases associated with an abnormal expression of histone deacetylase activity comprise tumors, neurodegenerative diseases, metabolic diseases, inflammatory reactions or heart diseases.
10. Use according to claim 9, characterized in that said tumors comprise various leukemias, lymphomas, myelomas, triple negative breast cancer, lung cancer, melanoma, liver cancer, esophageal cancer, kidney cancer, oral cancer, tongue cancer, prostate cancer, breast cancer, cervical cancer, ovarian cancer, stomach cancer, pancreatic cancer, bladder cancer, colorectal cancer, nasopharyngeal cancer, brain tumors, gliomas, anaplastic oligodendrogliomas, adult glioblastomas, adult anaplastic astrocytomas, bone cancer or soft tissue sarcomas.
CN202111103830.3A 2021-09-20 2021-09-20 Histone deacetylase inhibitor and preparation method and application thereof Active CN113831338B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111103830.3A CN113831338B (en) 2021-09-20 2021-09-20 Histone deacetylase inhibitor and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111103830.3A CN113831338B (en) 2021-09-20 2021-09-20 Histone deacetylase inhibitor and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN113831338A true CN113831338A (en) 2021-12-24
CN113831338B CN113831338B (en) 2022-12-20

Family

ID=78960082

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111103830.3A Active CN113831338B (en) 2021-09-20 2021-09-20 Histone deacetylase inhibitor and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113831338B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023011416A1 (en) * 2021-08-04 2023-02-09 中国海洋大学 Multi-target inhibitor targeting hdac and nad synthesis and use of multi-target inhibitor
CN115806503A (en) * 2022-12-02 2023-03-17 中国海洋大学 Selective histone deacetylase inhibitor as well as preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104592114A (en) * 2015-02-15 2015-05-06 山东大学 Quinolone substituted histone deacetylase inhibitor, preparation method and application thereof
CN106977425A (en) * 2017-04-01 2017-07-25 清华大学深圳研究生院 Hydroxamic acid derivs with dnmt rna and histon deacetylase (HDAC) inhibitory activity and preparation method and application
CN113754591A (en) * 2020-06-05 2021-12-07 山东大学 HDAC, JAK and BET three-target inhibitor and preparation method and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104592114A (en) * 2015-02-15 2015-05-06 山东大学 Quinolone substituted histone deacetylase inhibitor, preparation method and application thereof
CN106977425A (en) * 2017-04-01 2017-07-25 清华大学深圳研究生院 Hydroxamic acid derivs with dnmt rna and histon deacetylase (HDAC) inhibitory activity and preparation method and application
CN113754591A (en) * 2020-06-05 2021-12-07 山东大学 HDAC, JAK and BET three-target inhibitor and preparation method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
XIAOYANG LI: "《Design of Hydrazide-Bearing HDACIs Based on Panobinostat and Their p53 and FLT3-ITD Dependency in Antileukemia Activity》", 《JOURNAL OF MEDICINAL CHEMISTRY》, vol. 63, no. 10, 22 April 2020 (2020-04-22) *
YUHONG DONG: "《Design, synthesis and biological evaluation of novel c-Met/HDAC dual inhibitors》", 《BIOORGANIC & MEDICINAL CHEMISTRY LETTERS》, vol. 30, no. 20, 9 October 2020 (2020-10-09) *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023011416A1 (en) * 2021-08-04 2023-02-09 中国海洋大学 Multi-target inhibitor targeting hdac and nad synthesis and use of multi-target inhibitor
CN115806503A (en) * 2022-12-02 2023-03-17 中国海洋大学 Selective histone deacetylase inhibitor as well as preparation method and application thereof

Also Published As

Publication number Publication date
CN113831338B (en) 2022-12-20

Similar Documents

Publication Publication Date Title
RU2425041C2 (en) Crystalline and other forms of salts of lactic acid and 4-amino-5-fluorine-3-[6-(4-methylpiperazine-1-yl)-1h-benzimidazole-2-yl]-1h-quinoline-2-one
CN113831338B (en) Histone deacetylase inhibitor and preparation method and application thereof
TWI630194B (en) Bicyclic analgesic compounds
JP5897566B2 (en) Cyclic N, N'-diarylthiourea and N, N'-diarylurea-androgen receptor antagonists, anticancer agents, methods for their preparation and uses
CN104470911B (en) Urethane/urea derivative
JP2009242437A (en) Sulfonamide derivative
JP4660045B2 (en) Prophylactic or therapeutic agent for myocarditis, dilated cardiomyopathy and heart failure comprising NF-κB inhibitor as an active ingredient
US9790158B2 (en) Substituted tropolone derivatives and methods of use
CN103917534A (en) Carbamate/ urea derivatives containing piperidin and piperazin rings as h3 receptor inhibitors
CA2701638A1 (en) Pharmaceutical calcimimetics
BR112019015191A2 (en) STARCH COMPOUNDS AND USE OF THE SAME
JP5486008B2 (en) 1-Butyl-2-hydroxyaralkylpiperazine derivatives and their use as antidepressants
JPWO2014034626A1 (en) N- [2-({2-[(2S) -2-cyanopyrrolidin-1-yl] -2-oxoethyl} amino) -2-methylpropyl] -2-methylpyrazolo [1,5-a] pyrimidine-6 -Carboxamide crystals
CN107043373B (en) Oxotriazole NO donor statin derivative and preparation method and application thereof
WO2014159501A2 (en) Processes for preparing tetrahydroisoquinolines
WO2019232662A1 (en) Indolizine compound having anticancer activity and derivative thereof
WO2017175185A1 (en) Heteroaryl butanoic acid derivatives as lta4h inhibitors
WO2008080290A1 (en) Selective m4 receptor antagonist and its medical use
JP7186874B2 (en) Pyrazolyl compound and method of use thereof
WO2021062168A1 (en) Synthetic sphingolipid inspired molecules with heteroaromatic appendages, methods of their synthesis and methods of treatment
TW201206910A (en) Substituted N-heterocycloalkyl bipyrrolidinylphenyl amide derivatives, preparation and therapeutic use thereof
CN115806503A (en) Selective histone deacetylase inhibitor as well as preparation method and application thereof
ES2672326T3 (en) Deuterated thiazolidinone analogues as agonists for the follicle stimulating hormone receptor
JP2019089822A (en) New crystal form of topiroxostat, and preparation method therefor
JP3112356B2 (en) Cyclopentenone compound and cerebral function improving agent containing the compound as active ingredient

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