[go: up one dir, main page]

CN108264509B - Substituted benzothieno[2,3-c]tetrahydropyridine derivatives and preparation method and use thereof - Google Patents

Substituted benzothieno[2,3-c]tetrahydropyridine derivatives and preparation method and use thereof Download PDF

Info

Publication number
CN108264509B
CN108264509B CN201611258939.3A CN201611258939A CN108264509B CN 108264509 B CN108264509 B CN 108264509B CN 201611258939 A CN201611258939 A CN 201611258939A CN 108264509 B CN108264509 B CN 108264509B
Authority
CN
China
Prior art keywords
compound
mmol
substituted
preparation
nmr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201611258939.3A
Other languages
Chinese (zh)
Other versions
CN108264509A (en
Inventor
孙逊
王明亮
朱继东
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.)
Fudan University
Original Assignee
Fudan University
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 Fudan University filed Critical Fudan University
Priority to CN201611258939.3A priority Critical patent/CN108264509B/en
Publication of CN108264509A publication Critical patent/CN108264509A/en
Application granted granted Critical
Publication of CN108264509B publication Critical patent/CN108264509B/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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic Table
    • C07F5/02Boron compounds
    • C07F5/04Esters of boric acids

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明属药物化学和医药技术领域,涉及式(I)所示的取代的苯并噻吩并[2,3‑c]四氢吡啶衍生物及其制备方法和用作CYP17抑制剂的药用用途,同时包含此类化合物的组合物,及它们的使用方法。本发明进一步涉及包含至少一种如所述化合物的药物组合物,其用于治疗CYP17酶相关病症诸如癌症和其他增殖性疾病。式I中,X是S,NH以及N的取代物,R1包含但不限于:H,卤素,‑OH,‑CN,‑0Ra,‑NRaRb,‑NHCORa或‑C(O)ORa,被0至4个Ra取代的C1‑6烷基;被0至4个Ra取代的C3‑6环烷基;被0至6 Rb取代的芳基以及C1‑6氟代烷基或C3‑6氟代环烷基;R2包含但不限于:被取代的咪唑基以及被取代的吡啶基和嘧啶基;且R1、R2、Ra、Rb和Rc在申请中的定义。

Figure 100004_DEST_PATH_IMAGE002
The invention belongs to the technical field of medicinal chemistry and medicine, and relates to a substituted benzothieno[2,3-c]tetrahydropyridine derivative represented by formula (I), a preparation method thereof, and a medicinal use as a CYP17 inhibitor , and compositions containing such compounds, and methods of their use. The present invention further relates to pharmaceutical compositions comprising at least one compound as described for use in the treatment of CYP17 enzyme related disorders such as cancer and other proliferative diseases. In formula I, X is the substitute of S, NH and N, R 1 includes but is not limited to: H, halogen, -OH, -CN, -OR a , -NR a R b , -NHCOR a or -C(O ) OR a , C 1-6 alkyl substituted by 0 to 4 R a ; C 3-6 cycloalkyl substituted by 0 to 4 R ; aryl and C 1 substituted by 0 to 6 R b -6 fluoroalkyl or C 3-6 fluorocycloalkyl ; R includes but is not limited to: substituted imidazolyl and substituted pyridyl and pyrimidinyl; and R1, R2, Ra, Rb and Rc are in Definitions in the application.
Figure 100004_DEST_PATH_IMAGE002

Description

Substituted benzothieno [2,3-c ] tetrahydropyridine derivatives, preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicinal chemistry and medicine, and relates to substituted benzothieno [2,3-c ] tetrahydropyridine derivatives, a preparation method thereof, medicinal application of the derivatives as CYP17 inhibitors, a composition containing the compounds and a using method of the compounds. The invention further relates to pharmaceutical compositions comprising at least one such compound for use in the treatment of CYP17 enzyme-related disorders such as cancer and other proliferative diseases.
Background
The data show that prostate cancer is one of the most common malignancies threatening the health of men worldwide, with the incidence second among all malignancies in men. According to the statistics of cancer in the united states in 2014, the incidence rate of prostate cancer in the united states has been reported to exceed that of lung cancer, and the cancer becomes the first fatal killer endangering the health of men. Although the incidence rate of prostate cancer in China is lower than that of European and American countries, the increasing rate of the prostate cancer in China is far more rapid than that of European and American developed countries in recent years, and according to the latest statistical data of the cancer center in the country in 2012, the incidence rate of prostate cancer in China ranks six in male malignant tumors and reaches 9.92/10 ten thousand population. In clinical practice, open or laparoscopic prostatectomy is mostly adopted for early and non-metastatic prostate cancer patients, and radical treatment can be generally achieved; however, for patients in middle and late stages, no matter surgical resection or radiotherapy and chemotherapy, the postoperative recurrence rate is high, prostate cancer bone metastasis is easy to generate, the pain and the side effect of the patients are great, especially, a plurality of patients cannot be treated by the operation in the middle and late stages when diagnosis is confirmed, and clinical drug treatment is the best choice. The current primary therapeutic agents are primarily endocrine-related antagonists that block androgen binding to receptors and inhibitors of androgen biosynthesis, where androgens such as testosterone (T) and Dihydrotestosterone (DHT) promote prostate growth at the androgen receptor level and also drive the development of prostate cancer. Clinical practice shows that medical and surgical use of orchiectomy reduces or eliminates androgen production in the testes, but does not affect androgen synthesis in the adrenal gland; in addition, it has been demonstrated that testosterone and dihydrotestosterone produced in the adrenal glands are sufficient to activate the androgen receptor and thereby cause recurrence of prostate cancer; in addition, once a patient has progressed to castration resistant prostate cancer, androgens are also produced in tumor cells, making treatment with antiandrogens more difficult.
The CYP17 enzyme, which is a member of the large family of cytochrome P450 enzymes distributed in tissues such as adrenal gland, testis, prostate, etc., is a membrane-bound mono-oxidase with dual functions, and it comprises 17-alpha hydroxylase and C17, 20-lyase, and Pregnenolone (Pregnenolone) and Progesterone (Progesterone) are first converted into their 17 a-hydroxy derivatives by 17 alpha-hydroxylase in CYP17 enzyme, and then activated by C17, 20-lyase in CYP17 enzyme, respectively, to form corresponding androgens, namely Dehydroepiandrosterone (DHEA) and Androstenedione (androsteenedione), which are precursors of testosterone (T). Testosterone is further converted into Dihydrotestosterone (DHT) with stronger activity under the action of reductase, and trace testosterone and dihydrotestosterone in vivo can stimulate the growth of prostate cancer, so that CYP17 enzyme plays a very critical role in the biosynthesis of testosterone and dihydrotestosterone, and the inhibition of the biological activity of the enzyme can effectively block the biosynthesis of androgen, thereby inhibiting the growth of prostate cancer. In addition, the CYP17 enzyme inhibitor can inhibit the biosynthesis of androgen in testis, can also inhibit the biosynthesis of androgen in adrenal gland and other tissues such as prostate tumor tissue, and has better therapeutic action compared with the current clinical castration therapy which can only inhibit the biosynthesis of androgen in testis.
The clinical concept of CYP17 as a target for prostate cancer has been validated by the antifungal ketoconazole, and in recent years, abiraterone has been approved as an inhibitor of CYP17 for clinically superior therapeutic results. In addition to the use of CYP17 inhibitors for the treatment of prostate cancer, CYP17 inhibitors would be indicated for breast cancer signs, particularly estrogen dependent breast cancer. Treatment with high doses of ketoconazole inhibited testosterone and estradiol levels in postmenopausal patients with advanced breast Cancer, suggesting that CYP17 may be a potential target for hormonal therapy (Harris, a. l. et al, Br. j. Cancer l988, 58, 493). Abiraterone, while effective, has a number of disadvantages: for example, the bioavailability is low, the dosage is up to 1000 mg per day, prednisone is taken twice (5 mg each), especially the selectivity is poor, the prednisone not only is a substrate of CYP17, but also has an effect on other substrates of a plurality of cytochrome enzymes, for example, the prednisone is a strong inhibitor of CYP1A2 and CYP2D6 and is a moderate inhibitor of CYP2C9, CYP2C19 and CYP3A4/5, so that the hepatotoxicity is generated, for example, the increase of aspartate and alanine aspartate is caused; are also substrates of CYP11B1 and CYP19, thereby causing hypertension, hypokalemia, and adverse side effects associated with adrenal cortex dysfunction.
Therefore, new CYP17 inhibitors are needed. The invention provides a substituted benzothieno [2,3-c ] tetrahydropyridine derivative, a preparation method and application thereof in order to meet practical requirements.
Disclosure of Invention
The present invention aims to provide substituted benzothieno [2,3-c ] tetrahydropyridine derivatives (including salts and prodrugs thereof) useful as inhibitors of the CYP17 enzyme, in view of the current state of the art.
The invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and at least one compound of formula (I) or a salt or prodrug thereof.
The invention also provides a method of treating a disease or disorder associated with CYP17 enzyme activity, the method comprising administering a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a mammalian patient.
The invention also provides processes and intermediates for preparing compounds of formula (I) or salts or prodrugs thereof.
The invention also provides a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof, for use in therapy.
The invention also provides the use of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment of cancer.
The compounds of formula (I) and compositions comprising the compounds are inhibitors of the CYP17 enzyme and are useful in the treatment, prevention or cure of a variety of CYP17 enzyme-related disorders. Pharmaceutical compositions comprising these compounds are useful in the treatment, prevention, or slowing the progression of diseases or disorders, such as cancer, in a variety of therapeutic areas.
These and other features of the present invention will be described in expanded form as the disclosure continues.
In particular, the present invention provides in a first aspect a compound of formula (I):
Figure DEST_PATH_IMAGE002
in formula I:
x is S, NH and a substituent of N;
R1including but not limited to:
i. h, halogen, -OH, -CN, -0Ra,-NRaRb, -NHCORaOR-C (O) ORa;
By 0 to 4RaSubstituted C1-6An alkyl group;
is substituted by 0 to 4RaSubstituted C3-6A cycloalkyl group;
is substituted by 0 to 6RbA substituted aryl group;
v. C1-6fluoroalkyl or C3-6A fluorocycloalkyl group;
R2including but not limited to:
i. by RcA substituted imidazolyl group which is a substituted imidazolyl group,
Figure DEST_PATH_IMAGE004
;
a substituted pyridyl group,
Figure DEST_PATH_IMAGE006
wherein ring A is substituted with 0 to 2RbA substituted fused 5-to 6-membered aryl or heteroaryl ring;
each RaIndependently halogen, -OH-, CN, C3-6Cycloalkyl radical, C1-4Fluoroalkyl, C1-6Alkoxy radical, C1-4Fluoroalkoxy, phenyl substituted with 0 to 4, heterocyclyl substituted with 0 to 4, or heteroaryl substituted with 0 to 4;
each RbIndependently halogen, -OH-, CN, C3-6Cycloalkyl radical, C1-4Fluoroalkyl, C1-6Alkoxy radical, C1-4Fluoroalkoxy, phenyl substituted with 0 to 4, heterocyclyl substituted with 0 to 4, or heteroaryl substituted with 0 to 4;
each RcIndependently halogen, -OH-, CN, C3-6Cycloalkyl radical, C1-4Fluoroalkyl, C1-6An alkoxy group;
the features and advantages of the present invention will be more readily understood by those of ordinary skill in the art after reading the following detailed description. It is to be understood 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 in combination to form a subcombination thereof. The embodiments considered herein as exemplary or preferred are intended to be illustrative and not limiting.
Unless otherwise indicated, any heteroatom having unsatisfied valences is assumed to have a hydrogen atom sufficient to satisfy the valences.
The terms "halo" and "halogen" as used herein denote F, Cl, Br and I.
The term "alkyl" as used herein denotes a branched and straight chain saturated aliphatic hydrocarbon group containing, for example, from 1 to 12 carbon atoms, from 1 to 6 carbon atoms, and from 1 to 4 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, sec-butyl and tert-butyl) and pentyl (e.g., n-pentyl, isopentyl, neopentyl), n-hexyl, 2-methylpentyl, 2-ethylbutyl, 3-methylpentyl and 4-methylpentyl. When a number appears as a subscript following the symbol "C," the subscript more specifically defines the number of carbon atoms that a particular group may contain. For example, "C1-6Alkyl "denotes straight and branched alkyl groups having 1 to 6 carbon atoms.
The term "haloalkyl" as used herein, means an alkyl group having one or more hydrogen atoms replaced with halogen atom(s), in an amount ranging from one to up to the total number of hydrogen atoms present in the parent alkyl group when not halogenated. Representative examples of haloalkyl groups include, but are not limited to, chloromethyl (-CH)2C1) Trifluoromethyl (-CF)3) And 2, 2, 2-trifluoroethyl (-CH)2CF3). When a number appears as a subscript following the symbol "C," the subscript more specifically defines the number of carbon atoms that a particular haloalkyl group can contain. For example, "C1-4Haloalkyl "denotes straight and branched haloalkyl groups having 1 to 4 carbon atoms.
The term "cyano" denotes the group-CN.
The term "cycloalkyl" as used herein refers to a group derived from a non-aromatic monocyclic or polycyclic hydrocarbon molecule by the removal of one hydrogen atom from a saturated ring carbon atom. Representative examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclopentyl, and cyclohexyl. When a number appears as a subscript following the symbol "C," the subscript more specifically defines the number of carbon atoms that a particular cycloalkyl group may contain. For example, "C3-6Cycloalkyl "denotes a cycloalkyl group having 3 to 6 carbon atoms.
The term "alkoxy," as used herein, refers to an alkyl group attached to the parent molecular moiety through an oxygen atom, e.g., a methoxy group (-OCH)3)。
The term "aryl" as used herein refers to an atomic group derived from a molecule containing aromatic ring(s) by removal of one hydrogen bonded to the aromatic ring. Representative examples of aryl groups include, but are not limited to, phenyl, naphthyl, indanyl, indenyl, and 1,2,3, 4-tetrahydronaphthalen-5-yl.
The term "heteroatom" refers to oxygen (O), sulfur (S) and nitrogen (N).
The terms "heterocycle" or "heterocyclyl" are used interchangeably and refer to non-aromatic 3-to 7-membered monocyclic groups and 6-to 11-membered bicyclic groups, wherein at least one of the rings has at least one heteroatom (0, S or N), preferably from 1 to 3 heteroatoms independently selected from 0, S and/or N. The heteroatom containing rings of such groups may contain one or two oxygen or sulfur atoms and/or 1 to 4 nitrogen atoms per ring, provided that the total number of heteroatoms on each ring is four or less, and further provided that the ring contains at least one carbon atom. The nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized. The fused rings completing the bicyclic group may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. The heterocyclic group may be attached at any available nitrogen or carbon atom. The heterocyclic ring may be unsubstituted or, where valency permits, may contain one or more substituents.
The term "heteroaryl" refers to substituted or unsubstituted aromatic five-or six-membered monocyclic groups and 9-or 10-membered bicyclic groups having at least one heteroatom (O, S or N) in at least one ring, said heteroatom-containing ring preferably containing 1,2 or 3 heteroatoms independently selected from O, S and/or N. The heteroatom-containing heteroaryl group can contain one or two oxygen or sulfur atoms per ring and/or from 1 to 4 nitrogen atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom. The fused rings completing the bicyclic group may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. The nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized. The bicyclic or tricyclic heteroaryl group must include at least one fully aromatic ring but the other fused ring(s) may be aromatic or non-aromatic. The heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. The heteroaryl ring system may be unsubstituted or may contain one or more substituents. Exemplary monocyclic heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, and triazolyl. Exemplary bicyclic heteroaryl groups include indolyl, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, pentoxazolyl, pyrrolopyridinyl.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the organs of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the compound is modified by making acid or base salts of the parent compound. Examples of pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of basic residues such as amines; and basic or organic salts of acidic residues such as carboxylic acids. For example, the pharmaceutically acceptable salts include the conventional non-toxic salts or quaternary ammonium salts of the parent compound formed from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent, or a mixture of the two; in general, nonaqueous media such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile are preferred. For example, salts of the compounds of formula (I) may be prepared by reacting a compound of formula (I) with, for example, an equivalent amount of an acid or base in a medium which allows the newly formed salt to precipitate out or be isolated via freeze-drying. Exemplary acid salt(s) that the compound of formula (I) may form with inorganic and/or organic acids include, but are not limited to, for example, acetate, ascorbate, benzoate, benzenesulfonate, bisulfate, bitartrate, acid citrate (acid citrate), citrate, ethanesulfonate, formate, fumarate, gentisate, gluconate, glucuronate (glucaronate), glutamate, hydrochloride, hydrobromide, hydroiodide, isonicotinate, maleate, methanesulfonate (mesylate), methotaurate (methanesulfonate), nitrate, pantothenate, phosphate, acid phosphate, saccharinate (saccharate), salicylate, succinate, sulfate, tartrate, p-toluenesulfonate, trifluoroacetate, and lactate. These salts may be formed according to methods known to those of ordinary skill in the art.
"therapeutically effective amount" is intended to include an amount of a compound of the invention alone or in combination with the claimed compounds or in combination with other active ingredients effective to exert an antagonist effect on the CYP17 enzyme, or effective to treat cancer.
The compounds of the present invention are intended to include all isotopes of atoms occurring in the compounds of the present invention. Isotopes include those atoms having the same number of atoms but different mass numbers. By way of general example, but not limitation, isotopes of hydrogen include deuterium and tritium. Isotopes of carbon include13C and14C. isotopically-labeled compounds of the present invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein by substituting an appropriate isotopically-labeled reagent for an otherwise non-labeled reagent.
The invention also includes a class of pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, together with one or more non-toxic, pharmaceutically acceptable carriers and/or diluents and/or adjuvants (collectively referred to herein as "carrier" materials) and, if desired, other active ingredients. The compounds of formula (I) may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to that route, and in dosages effective for the intended treatment. For example, the compounds and compositions of the present invention may be administered orally, mucosally, or parenterally (including intravascular, intravenous, intraperitoneal, subcutaneous, intramuscular, intrasternal, and infusion techniques) in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles. For example, the pharmaceutically acceptable carrier may comprise a mixture of mannitol or lactose and microcrystalline cellulose. The mixture may contain additional components such as lubricants, e.g. magnesium stearate, and disintegrants such as crospovidone. The carrier mixture may be enclosed in gelatin capsules or compressed into tablets.
The pharmaceutically active compounds of the present invention can be processed according to conventional methods of pharmacy to produce medicaments for administration to patients, including humans and other mammals.
For oral administration, the pharmaceutical composition may be, for example, in the form of a tablet, capsule, suspension or liquid. The pharmaceutical composition is preferably prepared in the form of a dosage unit containing a specific amount of the active ingredient. Examples of such dosage units are tablets or capsules. The daily dosage suitable for humans or other mammals may vary widely depending on the condition of the patient and other factors, but may equally be determined using conventional methods.
For therapeutic purposes, the active compounds of the invention will generally be combined with one or more adjuvants appropriate to the indicated route of administration. If administered orally, the compounds may be mixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, gum arabic, sodium alginate, polyvinyl alcohol and/or polyvinyl pyrrolidone, and then tableted or encapsulated for convenient administration. The capsules or tablets may contain a controlled release formulation which may be provided in a dispersion of the active compound in hydroxypropylmethylcellulose.
The oily phase of the emulsion containing the compound of formula (I) may be constituted by known ingredients in a known manner. Although the phase may contain only one emulsifier, it may also contain a mixture of at least one emulsifier with fats and/or oils. Preferably, both hydrophilic and lipophilic emulsifiers are included as stabilizers. It is also preferred to include both oil and fat. In general, the emulsifier(s), with or without stabilizer(s), constitute the so-called emulsifying wax, which together with the oil and fat constitutes the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulation. Emulsifiers and emulsion stabilizers suitable for use in the formulations of the present invention include tween 60, span 80, cetostearyl alcohol, carob fringed pink alcohol, glyceryl monostearate, sodium lauryl sulfate/glyceryl distearate alone or with a wax, or other materials well known in the art.
The choice of oil or fat suitable for use in the formulation is based on achieving the desired cosmetic properties, since the solubility of the active compound in most oils that may be used in a pharmaceutical emulsion formulation is low. Thus, the cream should preferably be a non-greasy, non-staining and washable product having a suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain alkyl mono-or dibasic acids such as diisoadipate, isocetyl stearate, propylene glycol diester of coconut oil fatty acids, isopropyl carob fringed pink, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or mixtures of branched chain esters may be used. These may be used alone or in combination depending on the desired properties. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils may be used.
Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in orally administered formulations or using other suitable dispersing or wetting agents and suspending agents. The compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum and/or various buffers. Other adjuvants and modes of administration are well known in the pharmaceutical art. The active ingredient may also be administered by injection in the form of a composition with: suitable carriers include physiological saline, dextrose or water, or cyclodextrins, solubilizing co-solvents (i.e., propylene glycol), or solubilizing micelles (i.e., tween 80).
In the present invention, the compounds of formula (I) are useful in the treatment of cancer, for example, cancers that rely on androgen receptor signaling. These compounds inhibit the activity of CYP17 enzymes involved in androgen and estrogen biosynthesis. Blocking this enzyme can inhibit androgen production by gonads, adrenals and tumors and provide new treatment options for patients with cancers that depend on androgen receptor and estrogen receptor signaling, such as prostate cancer and estrogen receptor-positive breast cancer. Thus, the treatment comprises administering to the patient a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
In one embodiment, it provides a method of treating cancer comprising administering to a mammal in need thereof a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof. The methods of this embodiment may be used to treat a variety of cancers, including, but not limited to, breast, ovarian, and prostate cancers. Preferably, the method of this embodiment is used to treat prostate or breast cancer. In one method of this embodiment, a therapeutically effective amount of a compound of formula (I) is administered.
In one embodiment, it provides a method of treating a cancer that is dependent on CYP17 activation, the method comprising administering to a patient in need thereof a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof. In one method of this embodiment, a compound of formula (I) is administered to treat prostate cancer. In another method of this embodiment, a compound of formula (I) is administered to treat breast cancer. Preferably, a therapeutically effective amount of compound (I) is administered.
In cancer treatment, a combination of chemotherapeutic agents and/or other therapies (e.g., radiation therapy) is often advantageous. The mechanism of action of the second (or third) agent may be the same or different from that of the first therapeutic agent. It may be particularly effective to use a combination of cytotoxic drugs, wherein the two or more drugs administered act in different ways or in different phases of the cell cycle, and/or wherein the two or more drugs have overlapping toxicities or side effects, and/or wherein each of the drugs combined has a proven efficacy in treating the particular condition exhibited by the patient.
Thus, the compounds of formula (I) may be administered in combination with other anti-cancer treatments for the treatment of cancer or other proliferative diseases. The invention further includes the use of a compound of formula (I) in the manufacture of a medicament for the treatment of cancer, and/or the invention includes a package containing a compound of formula (I) as herein described together with instructions for use of the compound in combination with other anti-cancer or cytotoxic drugs and treatments for the treatment of cancer. The invention further includes combinations of a compound of formula (I) and one or more additional drugs in kit form, e.g., they are packaged together or in separate packages for sale together as a kit, or they are packaged so as to be formulated together.
The additional anti-cancer drug may be selected from any one or more of: alkylating agents (including nitrogen mustards, alkyl sulfonates, nitrosoureas, aziridine derivatives, and triazenes); anti-angiogenic agents (including matrix metalloproteinase inhibitors); antimetabolites (including adenosine deaminase inhibitors, folic acid antagonists, purine analogs, and pyrimidine analogs); antibiotics or antibodies (including monoclonal antibodies, CTLA-4 antibodies, anthracyclines); an aromatase inhibitor; hormonal and anti-hormonal agents and steroids (including synthetic analogs, glucocorticoids, estrogens/anti-estrogens, androgens/anti-androgens, progestins, progestin receptor agonists, and agonists and antagonists of luteinizing hormone-releasing hormone); insulin-like growth factor/insulin-like growth factor receptor (IGFR) systemic modulators; an integrin signaling inhibitor; kinase inhibitors (including multi-kinase inhibitors and/or inhibitors of Src kinase or Src/abl, cyclin-dependent kinase inhibitors), VEGF inhibitors, EGFR inhibitors, MEK inhibitors, Aurora kinase inhibitors, PDGF inhibitors, and other tyrosine kinase inhibitors or serine/threonine kinase inhibitors; microtubule stabilizing agents, such as taxanes, naturally occurring epothilones and synthetic and semi-synthetic analogs thereof; and so on.
In the present invention, the compounds described may be prepared by synthetic routes including methods analogous to those well known in the chemical arts, especially with reference to the description provided herein. For illustrative purposes, the following general schemes 1-4 show general methods of preparing the compounds of the present invention, as well as key intermediates. For a more detailed description of the individual reaction steps, see the examples section below. One skilled in the art will appreciate that other synthetic routes may also be used to synthesize the compounds of the present invention. Although specific starting materials and reagents are depicted in the schemes and discussed below, other starting materials and reagents can readily be substituted to provide a variety of compounds of the invention. In addition, many of the compounds prepared by the following methods may be further modified in accordance with the present disclosure using conventional chemical methods generally known to those skilled in the art.
For example, as shown in scheme 1, the product B can be obtained by the reduction of the commercially available reagent cyano compound a to an amino group while Boc protection is performed. A is reacted with formaldehyde in the presence of catalytic amounts of p-toluenesulfonic acid to form a six-membered ring compound C by the Pictet-Spengler reaction, further Boc removal in the presence of an acid such as trifluoroacetic acid to form a compound D, and D is reacted with a halide in the presence of a base or with an aldehyde by reductive amination to form a compound of general structure (I).
Scheme 1
Figure DEST_PATH_IMAGE008
Alternatively, as shown in scheme 2, commercially available reagent thiophenol compound E can be reacted with reagent F compound under the action of base to form intermediate G, G can be dehydrated to ring under acidic conditions to form H, H can be exchanged with ethyl ester in ammonia gas to form formamide intermediate I, formamide can be further reduced to amine to form intermediate J, and amine can be further protected to form product K. K reacts with formaldehyde under the action of catalytic amount of p-toluenesulfonic acid to form a six-membered ring compound L through a Pictet-Spengler reaction, ethyl formate is further removed under the action of a base such as sodium hydroxide to form a compound D, and the compound D reacts with a halide under the condition of existence of the base or reacts with aldehyde through a reductive amination reaction to form a compound with a general structure (I).
Scheme 2
Figure DEST_PATH_IMAGE010
Alternatively, as shown in scheme 3, the compound of general structure (I) is formed from a halogenated compound such as a brominated product as a starting material by a Suzuki reaction with boric acid or a borate ester under the action of a metal catalyst such as a palladium catalyst. Alternatively, a halogenated compound such as bromo product M may be initially formed as intermediate N in the boronic acid or boronic ester class, and intermediate N may then be reacted with the halogenated compound to form the compound of general structure (I). Or boric acid ester intermediate N is hydrolyzed into hydroxyl compound O, and the intermediate O reacts with halogenated compound under the action of alkali to form the compound with the general structure (I).
Scheme 3
Figure DEST_PATH_IMAGE012
Alternatively, as shown in scheme 4, starting from a halogenated compound such as a brominated product, a donor such as Zn (CN) with a cyano group under the action of a metal catalyst such as a palladium catalyst2Forming a cyano compound P, further hydrolyzing the cyano compound P into a carboxylic acid compound Q, and forming a compound with a general formula structure (I) by the Q and an amino compound. Or a halogenated compound such as a brominated product is used as a raw material and reacts with the amino compound to form the compound with the general formula structure (I).
Scheme 4
Figure DEST_PATH_IMAGE014
Alternatively, as shown in scheme 5, compounds of general structure (I) can also be formed on the N-substituted branch by derivatization. Such as starting from halogenated compounds, e.g., brominated products, forming a series of compounds by Suzuki reaction with halogenated compounds, reacting with amines by Buchward-Hartwig reaction to form a series of compounds, and the like.
Scheme 5
Figure DEST_PATH_IMAGE016
The present invention provides substituted benzothieno [2,3-c ] tetrahydropyridine derivatives, methods for their preparation and their use in medicine as inhibitors of CYP17, compositions containing such compounds, and methods for their use. The compounds of the present invention, and pharmaceutical compositions thereof, are useful for treating CYP17 enzyme-related disorders such as cancer and other proliferative diseases.
Detailed Description
The present invention is further illustrated below with reference to examples, which are by no means intended to limit the scope of the invention. It should be understood that these examples are given by way of illustration only. One skilled in the art can ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt the invention to various usages and conditions. Accordingly, the present invention is not limited to the following exemplary embodiments set forth herein, but is defined by the following claims.
All temperatures are degrees Celsius (. degree.C.) unless otherwise noted. Commercial reagents were used as received without additional purification. The starting materials may be varied using routine modifications, and additional steps may be employed to produce other compounds provided herein. All final products were characterized by NMR, HPLC, electrospray ionization mass spectrometry (ESI MS) or atmospheric pressure ionization (API MS) mass spectrometry.1H NMR spectra were obtained on a 400MHz Bruker instrument.13The C NMR spectrum was recorded at 100 MHz. The magnetic field strength is expressed in units of solvent peaks (parts per million, ppm), and the multiplicity of peaks is specified as follows: s, singlet; d, doublet; dd, doublet of doublets; t, triplet; q, quartet; brs, broad singlet; m, multiplet.
Abbreviations
PPA polyphosphoric acid
mL of
mg of
NMR nuclear magnetic resonance
ESI-MS electrospray ionization mass spectrometry
PdCl2 (dppf)CH2Cl2 Dichloro [2 ]1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) methylene chloride adduct
HATU 2- (7-benzotriazol oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate
Pd2(dba)3 Tris (dibenzylideneacetone) dipalladium
Xanthphos 4, 5-bis diphenylphosphine-9, 9-dimethylxanthene.
Example 1
2- (pyridin-3-ylmethyl) -2,3,4, 9-tetrahydro-1H-pyrido [3,4-b ] indole (1)
Figure DEST_PATH_IMAGE018
A50 mL dry round bottom flask was charged with 2,3,4, 9-tetrahydro-1H-pyrido [3,4-b ]]Indole (258 mg, 1.5 mmol) and 3-pyridinecarboxaldehyde (193 mg, 169 μ L, 1.8 mmol) were dissolved with 15mL of 1, 2-dichloroethane, stirred for 10 minutes in an ice bath, then 0.5mL of acetic acid was added, the reaction mixture was stirred for 10 minutes at room temperature, then sodium triacetoxyborohydride (636 mg, 3.0 mmol, 2 equiv) was added. The reaction was stirred at room temperature for 18 h under nitrogen, then 30mL of saturated aqueous sodium bicarbonate was added, dichloromethane was added for extraction (25 mL. times.3), the combined organic phases were washed with water, brine, respectively, the organic phases were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography (methanol/dichloromethane = 0 to 10%) to obtain 280 mg of a white solid with a yield of 71%. Melting point 171.2-173.0oC; 1H NMR (400 MHz, DMSO-d6) δ 10.85 (s, 1H), 8.57 (s, 1H), 8.50 (d, J = 4.8 Hz, 1H), 7.79 (d, J = 7.6 Hz, 1H), 7.40 – 7.34 (m, 2H), 7.26 (d, J = 8.0 Hz, 1H), 7.01 (t, J = 7.4 Hz, 1H), 6.93 (t, J = 7.4 Hz, 1H), 3.76 (s, 2H), 3.58 (s, 2H), 2.80 (t, J = 5.6 Hz, 2H), 2.69 (t, J = 5.2 Hz, 2H); 。
Example 2
2- (pyridin-3-ylmethyl) -2,3,4, 9-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (2)
Figure DEST_PATH_IMAGE020
Step 1: intermediate tert-butyl (2- (benzo [ b ] thiophen-3-yl) ethyl) carbamate (2c)
Figure DEST_PATH_IMAGE022
A50 mL dry round bottom flask was charged with benzo [ b ]]Thien-3-yl-acetonitrile (1.50 g, 8.66 mmol) and nickel (II) chloride hexahydrate (2.06 g, 8.66 mmol), 80mL of methanol was added, di-tert-butyl dicarbonate (3.78 g, 17.32 mmol) was gradually added dropwise, and then sodium borohydride (1.63 g, 43.3 mmol) was added to the reaction solution in portions, keeping the temperature below 45oC. The reaction was stirred at room temperature for 16 h under nitrogen, diluted with ethyl acetate (200 mL) and carefully added water (300 mL). The mixture was filtered through celite and the filter cake was washed with ethyl acetate (200 mL). The combined organic phases were washed with water, brine, respectively, the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the crude product, which was purified by chromatography on silica gel column (n-hexane/ethyl acetate = 4: 1) to give product 2c as a white solid (1.15 g, 47.9%). Melting point 71-72oC; 1H NMR (400 MHz, DMSO-d6): δ 7.96 (d, J = 8.8 Hz, 1H), 7.84 (d, J= 7.6 Hz, 1H), 7.44 – 7.35 (m, 3H), 6.99 (t, J = 5.6 Hz, 1H), 3.26 (q, J = 6.8 Hz, 2H), 2.94 (t, J = 7.2 Hz, 1H), 1.38 (s, 9H).
Step 2: intermediate 3, 4-dihydrobenzo [4,5] thieno [2,3-c ] pyridine-2 (1H) -carboxylic acid tert-butyl ester (2b)
Figure DEST_PATH_IMAGE024
A50 mL dry three-necked flask was charged with Compound 2c (1.08 g, 3.89 mmol), paraformaldehyde (234 mg, 7.78 mmol) and p-toluenesulfonylAcid monohydrate (37.1 mg, 0.195 mmol), then 80mL of dry toluene was added at 110 oAnd C, reacting for 2 hours. The reaction mixture was cooled to room temperature, the solvent was evaporated under reduced pressure, and then extracted by adding ethyl acetate (80 mL. times.2) and water (60 mL). The combined organic layers were washed with saturated aqueous sodium bicarbonate (50 mL) and brine (50 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure. The resulting residue was purified by silica gel column chromatography (n-hexane/ethyl acetate = 30:1 to 10: 1) to give product 2b (0.76 g, 67.5%) as a white solid. Mp is 88-89 ℃;1H NMR (400 MHz, DMSO-d6) δ 7.93 (d, J = 7.6 Hz, 1H), 7.67 (d, J= 8.0 Hz, 1H), 7.41 – 7.32 (m, 2H), 4.66 (s, 2H), 3.71 (t, J = 5.6 Hz, 2H), 2.80 (t, J = 5.6 Hz, 2H), 1.44 (s, 9H).
and step 3: intermediate 1,2,3, 4-tetrahydro-benzo [4,5] thieno [2,3-c ] pyridine (2a)
Figure DEST_PATH_IMAGE026
A50 mL dry round bottom flask was taken, compound 2b (700 mg, 2.42 mmol) was added, dissolved in 80mL dry dichloromethane and 2 mL trifluoroacetic acid was added dropwise. After stirring at room temperature for 2 hours, the solvent was evaporated under reduced pressure. The residue was diluted with water (30 mL), 20 mL of saturated sodium bicarbonate solution was added, and extracted with ethyl acetate (50 mL. times.2). The combined extracts were washed with brine (30 mL × 1), dried over anhydrous sodium sulfate, then filtered and the solvent was evaporated under reduced pressure to give compound 2a as a white solid (390 mg, 85.2%).1H NMR (400 MHz, DMSO-d6) δ 7.89 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.37 – 7.28 (m, 2H), 3.94 (s, 2H), 3.03 (t, J = 5.6 Hz, 2H), 2.68 (t, J = 5.6 Hz, 2H).
And 4, step 4:
in a similar manner to the preparation of compound 1, starting from compound 2a (190 mg, 1.0 mmol) and 3-pyridinecarboxaldehyde (129 mg, 1.2 mmol) 280 mg of white are obtainedCompound 2 as solid in 71% yield. Melting point of 61.2-62.9oC;1 H NMR (400 MHz, DMSO-d6) δ 8.83 (s, 1H), 8.76 (d, J = 4.4 Hz, 1H), 8.15 (d, J = 7.6 Hz, 1H), 7.99 (d, J = 7.6 Hz, 1H), 7.76 (d, J = 7.6 Hz, 1H),7.68 – 7.65 (m, 1H), 7.47 – 7.39 (m, 2H), 4.62 (s, 2H), 4.56 (s, 2H), 3.68 (s, 2H), 3.15 (s, 2H).。
Example 3
6-fluoro-2- (pyridin-3-ylmethyl) -2,3,4, 9-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (3)
Figure DEST_PATH_IMAGE028
Step 1: intermediate ethyl 4- ((4-fluorophenyl) thio) -3-oxobutanoate (3 g)
Figure DEST_PATH_IMAGE030
A500 mL dry round bottom flask was charged with 4-fluorobenzenethiol (12.8 g, 100.0 mmol), dissolved in 300 mL dry dichloromethane, and ethyl 4-chloroacetoacetate (13.5 mL, 100.0 mmol) was added dropwise over ice, followed by triethylamine (15.2 mL, 150.0 mmol). After the resulting suspension was stirred for another 2 hours under ice bath, the reaction mixture was poured into water (500 mL), and the aqueous layer was extracted with dichloromethane (200 mL. times.2). The combined organic layers were washed with saturated sodium bicarbonate solution, dilute hydrochloric acid (0.25N), brine, the organic layer was dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure. The resulting residue was purified by silica gel column chromatography (n-hexane/ethyl acetate = 9/1) to obtain 3g (23.8 g, 92.9%) as a pale yellow oil, which was used directly in the next step.1H NMR (400 MHz, CDCl3) δ 7.39 – 7.34 (m, 2H), 7.03 – 6.97 (m, 2H), 4.17 (q, J = 7.2 Hz, 2H), 3.74 (s, 4H), 3.62 (s, 2H), 1.26 (t, J = 7.2 Hz, 3H).
Step 2: intermediate ethyl 2- (5-fluorobenzo [ b ] thiophen-3-yl) acetate (3 f)
Figure DEST_PATH_IMAGE032
A500 mL dry round bottom flask was taken, 3g (23.6 g, 92.1 mmol) was dissolved in 20 mL toluene, and the solution was stirred under a stirring rod at 100oC was added to a mixed solution of PPA (100 g) and toluene (150 mL), and the reaction solution was then made 100 deg.f under nitrogenoStir overnight at C. Cooling the reaction mixture to room temperature, quenching in ice water, basification with potassium carbonate (pH-8), extraction with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure. Purification by silica gel column chromatography (petrol/ethyl acetate = 100/1 to 10/1) gave 3f as a white solid (12.2 g, 55.6%).1H NMR (400 MHz, CDCl3) δ 7.76 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.46 – 7.43 (m, 2H), 7.14 – 7.09 (m, 1H), 4.19 (q, J = 6.8 Hz, 2H), 3.80 (s, 2H), 1.27 (t, J = 6.8 Hz, 3H); 19F-NMR: -118.42 to -118.49.
And step 3: intermediate 2- (5-Fluorobenzo [ b ] thiophen-3-yl) acetamide (3 e)
Figure DEST_PATH_IMAGE034
A250 mL dry round bottom flask was taken, compound 3f (10.2 g, 42.8 mmol) was added, followed by a solution of ammonia in methanol (7M, 183mL, 1284 mmol), and the reaction mixture was sealed at room temperature (about 100 @)oC) Stirring was continued for 3 days to predict a large amount of solid, TLC showed the reaction was complete, then the solvent was removed and the residue was washed with cold water and slurried with PE/EA (10/1) mixed solution to give 3e (7.8 g, 87.6%) as a white solid.1H NMR (400 MHz, CDCl3) δ 8.01 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.66 – 7.59 (m, 3H), 7.28 – 7.23 (m, 1H), 7.01 (s, 1H), 3.63 (s, 2H); ESI-MS m/z: 210.0 [M+H]+.
And 4, step 4: intermediate 2- (5-Fluorobenzo [ b ] thiophen-3-yl) ethan-1-amine (3 d)
Figure DEST_PATH_IMAGE036
A500 mL dry three-necked flask was taken, compound 3e (7.2 g, 34.45 mmol) was added, dissolved in 150 mL dry tetrahydrofuran and heated to 80 under nitrogenoC, 1M borane/tetrahydrofuran (86.1 ml, 86.1 mmol) was added dropwise with heating and stirred at reflux overnight, then cooled to room temperature at 0oThe reaction was quenched by dropwise addition of 6N hydrochloric acid (60 mL) under C, then stirred at room temperature for 3 hours, the bulk of tetrahydrofuran was evaporated under reduced pressure, then basified with 2M sodium hydroxide solution, extracted with diethyl ether (300 mL. times.3), the combined diethyl ether layers were washed with water (2X 50 mL), dried over anhydrous sodium sulfate and concentrated in vacuo to give product 3d (5.83 g, 86.7%) which was used in the next step without further purification.
And 5: intermediate (2- (5-fluorobenzo [ b ] thiophen-3-yl) ethyl) carbamic acid ethyl ester (3 c)
Figure DEST_PATH_IMAGE038
A 500 mL dry round bottom flask was taken, 3d (6.72 g, 29.86 mmol) was added, dissolved with dichloromethane (200 mL), triethylamine (12.5 mL, 89.58 mmol) was added, then ethyl chloroformate (4.26 mL, 44.79 mmol) was added dropwise, after the addition was complete the reaction was allowed to react at room temperature for 2 hours, TLC showed completion of the reaction, then quenched with cold water, followed by saturated ammonium chloride solution, brine, drying over anhydrous sodium sulfate, removal of solvent under reduced pressure, and column chromatography of the residue (n-hexane/ethyl acetate = 100/1 to 10/1) gave 3c as a pale yellow oil (7.1 g, 88.9%).
1H NMR (400 MHz, CDCl3) δ 7.77 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.42 (dd, J = 9.6 Hz, J = 2.0 Hz, 1H), 7.24 (s, 1H), 7.13 – 7.08 (m, 1H), 4.84 (s, 1H), 4.15 – 4.09 (m, 2H), 3.52 (q, J = 6.4 Hz, 2H), 3.01 (t, J = 6.8 Hz, 2H), 1.23 (t, J = 6.8 Hz, 3H); ESI-MS m/z: 268.1 [M+H]+.
And 5: intermediate 6-fluoro-3, 4-dihydrobenzo [4,5] thieno [2,3-c ] pyridine-2 (1H) -carboxylic acid ethyl ester (3 b)
Figure DEST_PATH_IMAGE040
A500 mL dry round bottom flask was charged with Compound 3c (7.1 g, 26.56 mmole), dissolved in 200 mL toluene, charged with powdered paraformaldehyde (1.19 g, 39.84 mmole) and p-toluenesulfonic acid monohydrate (252 mg, 1.33 mmol), and refluxed under dehydrated conditions for 2 hours. The reaction mixture was cooled to room temperature, the solvent was evaporated under reduced pressure, and then extracted with ethyl acetate (200 mL. times.2) and water (100 mL). The combined organic layers were washed with aqueous sodium bicarbonate (100 mL) and brine (100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and the solvent was evaporated under reduced pressure. The residue was purified by silica gel column chromatography (petrol/ethyl acetate = 30/1 to 10/1) to give 3b (6.5 g, 87.8%) as a pale yellow oil.1H NMR (400 MHz, CDCl3) δ 7.70 (dd, J = 8.4 Hz, J = 4.8 Hz, 1H), 7.26 – 7.23 (m, 1H),7.08 – 7.03 (m, 1H), 4.76 (s, 2H), 4.21 (q, J = 6.8 Hz, 2H), 3.85 (t, J = 5.6 Hz, 2H), 2.81 (t, J = 5.6 Hz, 2H),1.23 (t, J = 6.8 Hz, 3H); ESI-MS m/z: 280.1 [M+H]+.
Step 6: intermediate 6-fluoro-1, 2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (3 a)
Figure DEST_PATH_IMAGE042
At 0oCompound 3b (6.5 g, 23.27 mmol) and NaOH (4.65 g, 116.35 mmol) were suspended in EtOH (100 mL) and water (30 mL) under CThen the reaction was stirred under reflux overnight, TLC followed the end of the reaction, then cooled, the solvent was removed under reduced pressure, water (100 mL) was added, extracted with ethyl acetate (100 mL × 3), dried over anhydrous sodium sulfate, the solvent was removed in vacuo, and the residue was slurried with a mixed solution of n-hexane and ethyl acetate (PE/EA = 6/1) to give 3a (3.8 g, 79.2%) as a white solid.
1H NMR (400 MHz, CDCl3) δ 7.69 (dd, J = 8.4 Hz, J = 4.8 Hz, 1H), 7.24 (dd, J = 8.8 Hz, J = 2.4 Hz, 1H),7.06 – 7.01 (m, 1H), 4.11 (t, J = 1.6 Hz, 2H), 3.23 (t, J = 5.8 Hz, 2H), 2.75 – 2.71 (m, 2H), 1.80 (s, 1H); ESI-MS m/z: 208.1 [M+H]+.
And 7:
in a similar manner to preparation 1, from compound 3a (103.5 mg, 0.5 mmol) and 3-pyridinecarboxaldehyde (64.3 mg, 0.6 mmol), compound 3 (109.5 mg, 73.4%) was obtained as a white solid. Melting point of 57.3-59.1oC; 1H NMR (400 MHz, CDCl3) δ 8.61 (d, J = 1.6 Hz, 1H), 8.59 – 8.57 (m, 2H), 7.89 (d, J= 8.0 Hz, 1H), 7.70 – 7.67 (m, 1H),7.35 – 7.32 (m, 1H), 7.26 – 7.23 (m, 1H), 7.08 – 7.03 (m, 1H), 3.87 (s, 2H), 3.85 (s, 2H), 3.03 (s, 2H), 2.88 (s, 2H); ESI-MS m/z: 299.0.。
Example 4: 6-chloro-2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (4)
Figure DEST_PATH_IMAGE044
Step 1: intermediate ethyl 4- ((4-chlorophenyl) thio) -3-oxobutanoate (4 g)
Figure DEST_PATH_IMAGE046
4g of compound was obtained as a pale yellow solid (4.69 g, 86%) from 4-chlorobenzenethiol (2.89 g, 20.0 mmol) in a similar manner as described for preparation 3 g. This compound was used directly in the next step immediately after purification by flash column chromatography.
Step 2: intermediate ethyl 2- (5-chlorobenzo [ b ] thiophen-3-yl) acetate (4 f)
Figure DEST_PATH_IMAGE048
Compound 4f was obtained as a white solid (2.02 g, 47%) from 3g (4.60 g, 16.9 mmol) in a similar manner as described for preparation 3 f.
And step 3: intermediate 2- (5-chlorobenzo [ b ] thiophen-3-yl) acetamide (4 e)
Figure DEST_PATH_IMAGE050
Compound 4e was obtained as a white solid (1.32 g, 76.2%) from 4f (1.95 g, 7.65 mmol) in a similar manner as described for preparation 3 e.1H NMR (400 MHz, DMSO) δ 8.01 (d, J = 8.4 Hz, 1H), 7.90 (d, J = 1.6 Hz, 1H), 7.62 (brs, 2H), 7.39 (dd, J = 8.4 Hz, J = 1.6 Hz, 1H), 7.02 (s, 1H), 3.66 (s, 2H); ESI-MS m/z: 226.0 [M+H]+.
And 4, step 4: intermediate 2- (5-chlorobenzo [ b ] thiophen-3-yl) ethan-1-amine (4 d)
Figure DEST_PATH_IMAGE052
Compound 4d was obtained as a pale yellow solid (1.18 g, 91.2%) from 4e (1.30 g, 6.14 mmol) in a manner similar to that described for preparation 3 d. This compound was used in the next step without further purification.
And 5: intermediate (2- (5-chlorobenzo [ b ] thiophen-3-yl) ethyl) carbamic acid ethyl ester (4 c)
Figure DEST_PATH_IMAGE054
Compound 4c was obtained as a pale yellow solid (1.38 g, 86.7%) from 4d (1.18 g, 5.57 mmol) in a similar manner as described for preparation 3 c.1H NMR (400 MHz, CDCl3) δ 7.76 (d, J = 8.8 Hz, 1H), 7.73 (d, J = 2.0 Hz, 1H), 7.31 (dd, J = 8.4 Hz, J = 2.0 Hz, 1H), 7.22 (s, 1H), 4.78 (s, 1H), 4.13 (q, J = 7.2 Hz, 2H), 3.53 (q, J = 6.0 Hz, 2H), 3.03 (t, J = 6.8 Hz, 2H), 1.24 (t, J = 6.8 Hz, 3H); ESI-MS m/z: 284.0 [M+H]+.
Step 6: intermediate 6-chloro-3, 4-dihydrobenzo [4,5] thieno [2,3-c ] pyridine-2 (1H) -carboxylic acid ethyl ester (4 b)
Figure DEST_PATH_IMAGE056
Compound 4b (1.04 g, 76.5%) was obtained as a pale yellow solid from 4c (1.30 g, 4.58 mmol) in a similar manner as described for preparation 3 b.1H NMR (400 MHz, CDCl3) δ 7.68 (d, J = 8.4 Hz, 1H), 7.55 (s, 1H), 7.26 (dd, J = 8.4 Hz, J = 2.0 Hz, 1H), 4.75 (s, 2H), 4.21 (q, J = 7.2 Hz, 2H), 3.84 (s, 2H), 2.81 (s, 2H), 1.30 (t, J = 7.2 Hz, 3H); ESI-MS m/z: 296.0 [M+H]+.
And 7: 6-chloro-1, 2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (4 a)
Figure DEST_PATH_IMAGE058
Compound 4a (570 mg, 72.5%) was obtained as a pale yellow solid from 4b (1.04 g, 3.52 mmol) in a similar manner as described for preparation 3 a.1H NMR (400 MHz, CDCl3) δ 7.75 (d, J = 2.0 Hz, 1H), 7.69 (d, J= 8.4 Hz, 1H), 7.37(dd, J = 8.4 Hz, J = 2.0 Hz, 1H), 4.02 (t, J = 2.0 Hz, 2H), 3.14 (t, J = 5.8 Hz, 2H), 2.77 – 2.73 (m, 2H); ESI-MS m/z: 268.0 [M+H]+.
And 8:
compound 4 was obtained as a white solid (63.1 mg, 66.8%) from compound 4a (67.2 mg, 0.3 mmol) and 3-pyridinecarbaldehyde (38.5 mg, 0.36 mmol) in a similar manner to preparation 1. Melting point 230.2-231.9oC; 1H NMR (400 MHz, DMSO-d6) δ 9.18 (s, 1H), 8.96 (d, J = 5.2 Hz, 1H), 8.81 (d, J = 8.0 Hz, 1H), 8.05 – 8.02 (m, 2H),7.87 (d, J = 7.6 Hz, 1H), 7.44 – 7.42 (m, 1H), 4.76 (s, 2H), 4.55 (s, 2H), 3.70 (s, 2H), 3.21 (s, 2H); ESI-MS m/z: 315.0[M+H]+.。
Example 5: 6-bromo-2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (5)
Figure DEST_PATH_IMAGE060
Step 1: intermediate ethyl 4- ((4-bromophenyl) thio) -3-oxobutanoate (5 g)
Figure DEST_PATH_IMAGE062
Compound 5g was obtained as a pale yellow solid (29.3 g, 92.4%) from 4-chlorobenzenethiol (18.9 g, 100.0 mmol) in a similar manner as described for preparation 3 g. This compound was used directly in the next step immediately after purification by flash column chromatography.
Step 2: intermediate ethyl 2- (5-bromobenzo [ b ] thiophen-3-yl) acetate (5 f)
Figure DEST_PATH_IMAGE064
Compound 5f was obtained as a white solid (7.7 g, 61.8%) from 5g (13.2 g, 41.6 mmol) in a similar manner as described for preparation 3 f.
And step 3: intermediate 2- (5-bromobenzo [ b ] thiophen-3-yl) acetamide (5 e)
Figure DEST_PATH_IMAGE066
Compound 5e (5.58 g, 82.3%) was obtained as a white solid from 5f (7.52 g, 25.13 mmol) in a similar manner as described for preparation 3 e.1H NMR (400 MHz, CDCl3) δ 7.88 (d, J = 2.0 Hz, 1H), 7.70 (d, J= 8.8 Hz, 1H), 7.43 (dd, J = 8.4 Hz, J = 5.6 Hz, 1H), 7.19 (s, 1H), 3.06 (t, J = 7.2 Hz, 2H), 2.95 (t, J = 7.2 Hz, 2H), 1.50 (brs, 2H); ESI-MS m/z: 270.0 [M+H]+.
And 4, step 4: 2- (5-Bromobenzo [ b ] thiophen-3-yl) eth-1-amine (5 d)
Figure DEST_PATH_IMAGE068
Compound 5d was obtained as a pale yellow solid (4.25 g, 86.3%) from 5e (5.20 g, 19.25 mmol) in a similar manner as described for preparation 3 d. This compound was used in the next step without further purification.
And 5: (2- (5-Bromobenzo [ b ] thiophen-3-yl) ethyl) carbamic acid ethyl ester (5 c)
Figure DEST_PATH_IMAGE070
Compound 5c (4.88 g, 90.8%) was obtained as a pale yellow solid from 5d (4.20 g, 16.39 mmol) in a similar manner as described for preparation 3 c.1H NMR (400 MHz, CDCl3) δ 7.89 (d, J = 2.0 Hz, 1H), 7.71 (d, J = 8.8 Hz, 1H), 7.44 (dd, J = 8.4 Hz, J = 5.6 Hz, 1H), 7.20 (s, 1H), 4.76 (s, 1H), 4.13 (q, J = 7.2 Hz, 2H), 3.53 (q, J = 6.4 Hz, 2H), 3.03 (t, J = 7.2 Hz, 2H), 1.24 (t, J = 7.2 Hz, 3H); ESI-MS m/z: 328.0 [M+H]+.
Step 6: 6-bromo-3, 4-dihydrobenzo [4,5] thieno [2,3-c ] pyridine-2 (1H) -carboxylic acid ethyl ester (5 b)
Figure DEST_PATH_IMAGE072
Compound 5b (3.34 g, 79.6%) was obtained as a pale yellow solid from 5c (4.05 g, 12.34 mmol) in a similar manner as described for preparation 3 b.1H NMR (400 MHz, CDCl3) δ 7.71 (s, 1H), 7.63 (d, J = 8.4 Hz, 1H), 7.39 (dd, J = 8.4 Hz, J = 2.0 Hz, 1H), 4.75 (s, 2H), 4.21 (q, J = 7.2 Hz, 2H), 3.84 (s, 2H), 2.81 (s, 2H), 1.30 (t, J = 7.2 Hz, 3H); ESI-MS m/z: 340.0 [M+H]+.
And 7: 6-bromo-1, 2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (5 a)
Figure DEST_PATH_IMAGE074
Compound 5a (1.86 g, 71.5%) was obtained as a white solid from 5b (3.30 g, 9.70 mmol) in a similar manner as described for preparation 3 a.
And 8:
in a similar manner to preparation 1, compound 5 was obtained as a white solid (523.8 mg, 72.9%) from compound 5a (536.3 mg, 2.0 mmol) and 3-pyridinecarboxaldehyde (257 mg, 2.4 mmol). Melting point of 113.2-114.9oC; 1H NMR (400 MHz, CDCl3) δ 8.61 (s, 1H), 8.57 – 8.56 (m, 1H), 7.83 (d, J = 8.0 Hz, 1H), 7.71 (d, J = 1.6 Hz, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.40 – 7.30 (m, 2H), 3.83 (s, 2H), 3.81 (s, 2H), 2.98 (t, J = 5.8 Hz, 2H), 2.86 (t, J = 5.6 Hz, 2H); ESI-MS m/z: 358.9/ 360.9[M+H]+.。
Example 6
6-methoxy-2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (6)
Figure DEST_PATH_IMAGE076
Step 1: intermediate ethyl 4- ((4-methoxyphenyl) thio) -3-oxobutanoate (6 g)
Figure DEST_PATH_IMAGE078
Compound 6g was obtained as a pale yellow solid (4.6 g, 85.8%) from 4-methoxythiophenol (2.8 g, 20.0 mmol) in a manner similar to that described for preparation 3 g. This compound was used directly in the next step immediately after purification by flash column chromatography.
Step 2: intermediate ethyl 2- (5-methoxybenzo [ b ] thiophen-3-yl) acetate (6 f)
Figure DEST_PATH_IMAGE080
Compound 6f was obtained as a pale yellow solid (2.14 g, 53%) from 6g (4.32 g, 16.1 mmol) in a similar manner as described for preparation 3 f.
And step 3: intermediate 2- (5-methoxybenzo [ b ] thiophen-3-yl) acetamide (6 e)
Figure DEST_PATH_IMAGE082
Compound 6e was obtained as a white solid (1.25 g, 72.1%) from 6f (1.96 g, 7.83 mmol) in a similar manner as described for preparation 3 e.1H NMR (400 MHz, DMSO-d6) δ 7.83 (d, J = 8.8 Hz, 1H), 7.59 (s, 1H), 7.49 (s, 1H), 7.01 (dd, J = 8.8 Hz, J = 2.4 Hz, 2H), 3.82 (s, 3H), 3.62 (s, 2H);
And 4, step 4: intermediate 2- (5-methoxybenzo [ b ] thiophen-3-yl) ethan-1-amine (6 d)
Figure DEST_PATH_IMAGE084
Compound 6d was obtained from 6e (1.20 g, 5.42 mmol) in a similar manner as described for preparation 3d as a pale yellow solid (0.93 g, 82.5%) which was used in the next step without further purification.
And 5: intermediate (2- (5-methoxybenzo [ b ] thiophen-3-yl) ethyl) carbamic acid ethyl ester (6 c)
Figure DEST_PATH_IMAGE086
Compound 6c (1.04 g, 82.7%) was obtained as a pale yellow oil from 6d (0.93 g, 4.48 mmol) in a similar manner as described for preparation 3 c.1H NMR (400 MHz, CDCl3) δ 7.72 (d, J = 8.8 Hz, 1H), 7.22 (s, 1H), 7.17 (s, 1H), 7.01 (dd, J = 8.8 Hz, J = 2.4 Hz, 1H), 4.78 (brs, 1H), 4.12 (q, J = 6.8 Hz, 2H), 3.89 (s, 3H), 3.53 (q, J = 6.8 Hz, 2H), 3.03 (t, J= 6.8 Hz, 2H), 1.23 (t, J = 6.8 Hz, 3H); ESI-MS m/z: 280.1 [M+H]+.
Step 6: intermediate 6-methoxy-3, 4-dihydrobenzo [4,5] thieno [2,3-c ] pyridine-2 (1H) -carboxylic acid ethyl ester (6 b)
Figure DEST_PATH_IMAGE088
Compound 6b was obtained as a pale yellow solid (0.81 g, 81.6%) from 6c (0.95 g, 3.40 mmol) in a similar manner as described for preparation 3 b.1H NMR (400 MHz, CDCl3) δ 7.65 (d, J = 8.8 Hz, 1H), 7.03 (d, J = 2.4 Hz, 1H), 6.95 (dd, J = 8.8 Hz, J = 2.4 Hz, 1H), 4.75 (s, 2H), 4.20 (q, J = 7.2 Hz, 2H), 3.87 (s, 2H), 3.85 (s, 2H), 2.82 (s, 2H), 1.30 (t, J = 7.2 Hz, 3H); ESI-MS m/z: 292.1 [M+H]+.
And 7: intermediate 6-methoxy-1, 2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (6 a)
Figure DEST_PATH_IMAGE090
Compound 6a was obtained as a pale yellow solid (417 mg, 73.8%) from 6b (750 mg, 2.58 mmol) in a similar manner as described for preparation 3 a.
And 8:
compound 6 was obtained as a white solid (64.5 mg, 69.3%) from compound 6a (65.8 mg, 0.3 mmol) and 3-pyridinecarboxaldehyde (38.5 mg, 0.36 mmol) in a similar manner to preparation 1. Melting point of 211.7-212.3oC; 1H NMR (400 MHz, CDCl3) δ 9.17 (s, 1H), 8.94 (d, J = 5.2 Hz, 1H), 8.77 (d, J = 7.6 Hz, 1H), 8.02 – 7.99 (m, 1H), 7.85 (d, J = 8.8 Hz, 1H), 7.26 (d, J = 2.0 Hz, 1H), 7.04 – 7.01 (m, 1H), 4.75 (s, 2H), 4.51 (s, 2H), 3.84 (s, 3 H), 3.53 (s, 2H), 3.18 (s, 2H); ESI-MS m/z: 311.0 [M+H]+.。
Example 7
6-methyl-2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (7)
Figure DEST_PATH_IMAGE092
Compound 7 was obtained as a pale yellow oil (50.2 mg, 56.8%) from 5 (107.8 mg, 0.3 mmol) and methylboronic acid (27 mg, 0.45 mmol) following the general Suzuki reaction experimental procedure.1H NMR (400 MHz, CDCl3) δ 8.61 (d, J = 1.2 Hz, 1H), 8.56 – 8.54 (m, 1H), 8.61 (d, J = 1.2 Hz, 1H), 7.77 (d, J = 7.6 Hz, 1H), 7.65 (d, J = 8.0 Hz, 1H), 7.37 – 7.28 (m, 2H), 7.12 (d, J = 8.0 Hz, 1H), 3.78 (s, 2H), 3.77 (s, 2H), 2.93 (t, J = 5.6 Hz, 2 H), 2.84 (t, J = 5.6 Hz, 2 H), 2.46 (s, 2H); ESI-MS m/z: 295.0[M+H]+.。
Example 8
2- (pyridin-3-ylmethyl) -6- (4, 4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (8)
Figure DEST_PATH_IMAGE094
To a suspension of 5 (359.3 mg, 1.0 mmol), bis (pinacol) diboron (380.9 mg, 1.5 mmol) and KOAc (294.0 mg, 3.0 mmol) in DMF (4.0 mL) was added Pd Cl2 (dppf)CH2Cl2 (36.5 mg, 0.05 mmol) and the reaction mixture was stirred at 100 deg.CoStirring for 3 hours under C. After cooling to room temperature, the mixture was diluted with ethyl acetate (50 mL) and water (20 mL), the organic layer was separated and the aqueous layer was extracted three times with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was then purified by silica gel column chromatography (DCM/MeOH = 100/1 to 100/5) to give 8 (257.8 mg, 63.5%) as a white solid. Melting point of 128.7-130.2oC; 1H NMR (400 MHz, CDCl3) δ 8.60 (d, J = 1.6 Hz, 1H), 8.55 – 8.54 (m, 1H), 8.04 (s, 1H), 7.77 – 7.70 (m, 3H), 7.31 – 7.28 (m, 1H), 3.78 (s, 2H), 3.77 (s, 2H), 2.94 – 2.87 (m, 4H), 1.37 (s, 12H); ESI-MS m/z: 407.1[M+H]+.。
Example 9
2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridin-6-ol (9)
Figure DEST_PATH_IMAGE096
At 0oTo a solution of 8 (203.5 mg, 0.5 mmol) and NaOH (40 mg, 1.0 mmol) in tetrahydrofuran and methanol (1: 1, 8 mL) was added 30% hydrogen peroxide (0.2 mL) at C. The reaction mixture was allowed to warm to room temperature and stirred for 2 hours. After quenching with water, the resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate, and then concentrated under reduced pressure. The residue was then purified by silica gel column chromatography (DCM/MeOH = 100/1 to 100/5) to give 9 (79.5 mg, 53.6%) as a white solid. Melting point 207.5-208.3oC; 1H NMR (400 MHz, CDCl3) δ 8.55 (d, J = 1.2 Hz, 1H), 8.49 – 8.48 (m, 1H), 7.78 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.4 Hz, 1H), 7.38 (t, J = 5.6 Hz, 1H), 6.94 (d, J = 2.0 Hz, 1H), 6.81 – 6.79 (m, 1H), 3.75 (s, 2H), 3.66 (s, 2H), 2.80 (t, J = 5.6 Hz, 2H), 2.69 (t, J = 5.2 Hz, 2H); ESI-MS m/z: 297.0[M+H]+.。
Example 10
2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine-6-carbonitrile (10)
Figure DEST_PATH_IMAGE098
A mixture of compound 5 (450 mg, 1.25 mmol), zinc cyanide (176 mg, 1.5 mmol) and tetrakis (triphenylphosphine) palladium (145 mg, 0.125 mmol) in DMF (10 mL) was taken at 100oC was stirred under nitrogen for 6 hours. After cooling, it was poured into saturated sodium bicarbonate solution, then extracted with ethyl acetate, the combined organic phases were washed with water, brine, dried over anhydrous sodium sulfate, the solvent was distilled under pressure, and the residue was purified by silica gel column chromatography (DCM/MeOH = 100/1 to 100/5) to give 10 (274.2 mg, 71.8%) as a white solid. Melting point of 131.7-132.9oC; 1H NMR (400 MHz, CDCl3) δ 8.61(s, 1H), 8.56 (d, J = 3.6 Hz, 1H), 7.88 – 7.78 (m, 3H), 7.51 (dd, J = 8.4 Hz, J = 1.6 Hz, 1H), 7.33 – 7.30 (m, 1H), 3.82 (s, 2H), 3.80 (s, 2H), 2.98 (t, J = 5.6 Hz, 2H), 2.90 (t, J = 5.2 Hz, 2H); ESI-MS m/z: 306.0[M+H]+.。
Example 11
N-methyl-2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine-6-carboxamide (11)
Figure DEST_PATH_IMAGE100
To a solution of 10 (210 mg, 0.68 mmol) in ethanol (5 mL) and water (5 mL) was added sodium potassium hydroxide (220 mg, 5.51 mmol). The resulting mixture was stirred under nitrogen at reflux for 12 hours and then evaporated in vacuo. The residue was dissolved in ethyl acetate (30 mL) and washed with 2M dilute hydrochloric acid (10 mL). The organic extracts were dried over anhydrous sodium sulfate and evaporated in vacuo to give crude 11a, which was used in the next step without further purification.
Compound 11a from the above step was dissolved in DMF (6 mL) then at 0oMethylamine hydrochloride (210 mg, 1.36 mmol), HATU (775.6 mg, 2.04 mmol) and DIPEA (0.71 mL, 4.08 mmol) were added under C. The reaction mixture was stirred at room temperature for 12 hours. After quenching with water, the resulting mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over anhydrous sodium sulfate, and then concentrated under reduced pressure. The residue was then purified by silica gel column chromatography (DCM/MeOH = 100/1 to 10/1) to give 11 (83.7 mg, 36.5%, two steps) as a white solid. Melting point 175.7-177.2oC; 1H NMR (400 MHz, DMSO-d6) δ 8.58(s, 1H), 8.52 (d, J = 3.6 Hz, 1H), 8.02 (s, 1H), 7.75 – 7.61 (m, 3H), 7.29 – 7.26 (m, 1H), 6.65 (d, J = 3.6 Hz, 1H), 3.75 (s, 2H), 3.73 (s, 2H), 3.01(d, J = 4.8 Hz, 3H), 2.88 (t, J = 5.6 Hz, 2H), 2.81 (t, J = 5.2 Hz, 2H); ESI-MS m/z: 338.0[M+H]+.。
Example 12
(2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridin-6-yl) carbamic acid tert-butyl ester (12)
Figure DEST_PATH_IMAGE102
To a round bottom flask under dry nitrogen was added Pd2(dba)3(91.5 mg, 0.1 mmol), Xantphos (173.6 mg, 0.3 mmol), 5 (359.3 mg, 1.0 mmol), tert-butyl carbamate (175.8 mg, 1.5 mmol) and cesium carbonate (651.6 mg, 2.0 mmol). Dry 1, 4-dioxane (15 mL) was added via syringe and the mixture was added at 100 deg.CoStirring under dry nitrogen for 6 hours under C. The reaction mixture was cooled to room temperature, diluted with water and extracted three times with ethyl acetate. The combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude mixture was purified by silica gel chromatography (DCM/MeOH = 100/1 to 10/1) to give 12 (247.2 mg, 62.5%) as a white solid. Melting point of 181.7-182.5oC; 1H NMR (400 MHz, CDCl3) δ 8.61(s, 1H), 8.55 (d, J = 4.4 Hz, 1H), 7.80 – 7.58 (m, 2H), 7.32 – 7.28 (m, 1H), 6.60 (s, 1H), 3.79 (s, 2H), 3.77 (s, 2H), 2.95 (t, J = 5.6 Hz, 2H), 2.87 (t, J = 5.2 Hz, 2H), 1.53 (s, 9H); ESI-MS m/z: 396.1[M+H]+.。
Example 13: n- (2- (pyridin-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridin-6-yl) acetamide (13)
Figure DEST_PATH_IMAGE104
At 0oTrifluoroacetic acid (1 mL) was added to a solution of 12 (138.5 mg, 0.35 mmol) in DCM (5 mL) under C. The reaction mixture was warmed to room temperature and stirred for 2 hours, the solvent was removed under reduced pressure, and the residue was dissolved in DCM (5 mL) at 0oDIPEA (0.24 mL, 1.38 mmol) followed by acetic anhydride (0.13 mL, 1.38 mmol) was added under C. The reaction mixture was warmed to room temperature, stirred for 12 hours, saturated sodium bicarbonate solution was added, dichloromethane was added and extracted, the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo, and the residue was purified by column chromatography on silica eluting with 5% MeOH in DCM to give 13 as a white solid (81.2 mg, 68.7%). Melting point of 168.6-169.9oC; 1H NMR (400 MHz, CDCl3) δ 8.61(s, 1H), 8.55 (d, J = 3.6 Hz, 1H), 7.90 (s, 1H), 7.76 (d, J = 7.6 Hz, 1H), 7.66 (d, J = 8.4 Hz, 1H), 7.48 (s, 1H), 7.31 – 7.28 (m, 2H), 3.77 (s, 2H), 3.75 (s, 2H), 2.91 (t, J = 5.6 Hz, 2H), 2.82 (t, J = 5.2 Hz, 2H), 2.20 (s, 9H); ESI-MS m/z: 338.0[M+H]+.。
Example 14
(6-fluoro-3, 4-dihydrobenzo [4,5] thieno [2,3-c ] pyridin-2 (1H) -yl) (pyridin-3-yl) methanone (14)
Figure DEST_PATH_IMAGE106
To a solution of 3a (62.1 mg, 0.3 mmol) and nicotinic acid (44.5 mg, 0.36 mmol) in DMF (5 mL) was added HATU (228.5 mg, 0.6 mmol) and DIPEA (0.16 mL, 0.9 mmol). The reaction mixture was then warmed to room temperature and stirred for 12 hours, after quenching with water, the resulting mixture was extracted with ethyl acetate, the organic layer was washed with brine, dried over anhydrous sodium sulfate, and then concentrated and the residue was purified by silica gel column chromatography (DCM/MeOH = 100/1 to 10/1) to give 14 as a white solid (53.2 mg, 56.7%). Melting point: 159.7 to 161.1 ℃;1H NMR (400 MHz, CDCl3) δ 8.76 – 8.72 (m, 2H), 7.83 (d, J = 6.8 Hz, 1H), 7.72 (s, 1H), 7.46 – 7.20 (m, 2H), 7.11 – 7.07 (m, 1H), 5.04 – 3.80 (m, 4H), 2.95 – 2.83 (m, 2H); ESI-MS m/z: 313.0[M+H]+.。
example 15
2- ((4-Bromopyridin-3-yl) methyl) -6-fluoro-1, 2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (15)
Figure DEST_PATH_IMAGE108
Compound 15 (516.8 mg, 68.5%) was obtained as a white solid from 3a (414.5 mg, 2.0 mmol) and 4-bromonicotinaldehyde (446.5 mg, 2.4 mmol) in a similar manner to preparation 1. Melting point of 121.8-123.1oC; 1H NMR (400 MHz, CDCl3) δ 8.66(s, 1H), 8.33 (d, J = 5.2 Hz, 1H), 7.70 – 7.67 (m, 1H), 7.53 (d, J = 5.2 Hz, 1H), 7.24 (d, J = 7.6 Hz, 1H), 7.07 – 7.02 (m, 1H), 3.88 (s, 2H), 3.86 (s, 2H), 3.00(t, J = 5.6 Hz, 2H), 2.84 (t, J = 5.8 Hz, 2H); ESI-MS m/z: 376.9/378.9[M+H]+.。
Example 16
6-fluoro-2- ((4-methylpyridin-3-yl) methyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (16)
Figure DEST_PATH_IMAGE110
Compound 16 (40.8 mg, 43.5%) was obtained as a white solid from 15 (113.2 mg, 0.3 mmol) and methylboronic acid (27 mg, 0.45 mmol) in a similar manner to preparation 7. Melting point of 113.6-115.1oC; 1H NMR (400 MHz, CDCl3) δ 8.67 – 8.34 (m, 2H), 7.68 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.25 – 7.22 (m, 2H), 7.07 – 7.02 (m, 1H), 3.80 (s, 2H), 3.78 (s, 2H), 2.97 (t, J = 5.6 Hz, 2H), 2.82 (t, J = 5.6 Hz, 2H), 2.48 (s, 3H); ESI-MS m/z: 313.0[M+H]+.。
Example 17
6-fluoro-2- ((4-phenylpyridin-3-yl) methyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (17)
Figure DEST_PATH_IMAGE112
Compound 17 (62.8 mg, 55.8%) was obtained as a white solid from 15 (113.2 mg, 0.3 mmol) and phenylboronic acid (54.8 mg, 0.45 mmol) in a similar manner to preparation 7. Melting point of 141.3-142.8oC; 1H NMR (400 MHz, CDCl3) δ 8.85 (s, 1H), 8.61 (d, J = 4.4 Hz, 1H), 7.67 (dd, J = 8.8 Hz, J= 4.8 Hz, 1H), 7.46 – 7.40 (m, 5H), 7.30 (d, J = 5.2 Hz, 1H), 7.21 (dd, J = 9.2 Hz, J = 2.4 Hz, 1H), 7.06 – 7.01 (m, 1H), 3.76 (s, 2H), 3.74 (s, 2H), 2.88 (t, J = 5.6 Hz, 2H), 2.77 (t, J = 5.2 Hz, 2H); ESI-MS m/z: 375.0[M+H]+.。
Example 18
2- ((4-Cyclopropylpyridin-3-yl) methyl) -6-fluoro-1, 2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (18)
Figure DEST_PATH_IMAGE114
Compound 18 (64.5 mg, 63.5%) was obtained as a white solid from 15 (113.2 mg, 0.3 mmol) and cyclopropylboronic acid (38.8 mg, 0.45 mmol) in a similar manner to preparation 7. Melting point 138.1-140.3oC; 1H NMR (400 MHz, CDCl3) δ 8.60 – 8.34 (m, 2H), 7.68 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.24 (dd, J = 9.2 Hz, J = 2.4 Hz, 1H), 7.07 – 7.02 (m, 1H), 6.90 – 6.86 (m, 1H), 3.95 (s, 2H), 3.83 (s, 2H), 2.99 (t, J = 5.6 Hz, 2H), 2.83 (t, J = 5.6 Hz, 2H), 2.39 – 2.32 (m, 1H), 1.19 – 1.14 (m, 1H), 0.86 – 0.82 (m, 1H); ESI-MS m/z: 339.1[M+H]+.。
Example 19
6-fluoro-2- ((4- (prop-1-en-2-yl) pyridin-3-yl) methyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (19)
Figure DEST_PATH_IMAGE116
Compound 19 was obtained as a pale yellow oil (90.5 mg, 53.5%) from 15 (188.5 mg, 0.5 mmol) and isopropenylboronic acid (64.5 mg, 0.75 mmol) in a similar manner to preparation 7.1H NMR (400 MHz, CDCl3) δ 8.67 (s, 1H), 8.46 (d, J = 5.2 Hz, 1H), 7.67 (dd, J = 8.4 Hz, J = 4.8 Hz, 1H), 7.22 (dd, J = 8.8 Hz, J = 2.4 Hz, 1H), 7.07 (d, J = 5.2 Hz, 1H), 7.05 – 7.00 (m, 1H), 5.25 (t, J = 1.6 Hz, 1H), 4.98 (d, J = 0.4 Hz, 1H), 3.77 (s, 2H), 3.74 (s, 2H), 2.89 (t, J = 5.8 Hz, 2H), 2.77 (t, J = 5.6 Hz, 2H), 2.05 (s, 1H); ESI-MS m/z: 339.1[M+H]+.。
Example 20
6-fluoro-2- ((4-isopropylpyridin-3-yl) methyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (20)
Figure DEST_PATH_IMAGE118
Mixing 19 (60 mg, 0.18 mmol) and 20% Pd (OH)2After stirring (30 mg) in a mixed solvent of MeOH (3 mL) and EtOAc (3 mL) under a balloon of hydrogen at room temperature for 6 hours, the mixture was filtered through celite, filtered, and concentrated. The residue was then purified by silica gel column chromatography (DCM/MeOH = 100/1 to 10/1) to give 20 (41.9 mg, 68.5%) as a light yellow oil.1H NMR (400 MHz, CDCl3) δ 8.55 – 8.45 (m, 2H), 7.68 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.27 (d, J = 5.3 Hz, 1H), 7.22 (dd, J = 9.6 Hz, J = 2.4 Hz, 1H), 7.05 – 7.00 (m, 1H), 3.78 (s, 2H), 3.75 (s, 2H), 3.44 – 3.37 (m, 1H), 2.90 (t, J = 5.6 Hz, 2H), 2.78 (t, J = 5.2 Hz, 2H), 1.22 (d, J= 6.8 Hz, 6H); ESI-MS m/z: 341.1[M+H]+.。
Example 21
6-fluoro-2- ((5-fluoropyridin-3-yl) methyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (21)
Figure DEST_PATH_IMAGE120
Compound 21 (55.6 mg, 58.5%) was obtained as a white solid from 3a (62.5 mg, 0.3 mmol) and 5-fluoronicotinaldehyde (45.5 mg, 0.36 mmol) in a similar manner to preparation 1. Melting Point:60.8-62.6℃; 1H NMR (400 MHz, CDCl3) δ 8.42 – 8.41 (m, 2H), 7.68 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.54 – 7.51 (m, 1H), 7.24 (dd, J = 9.2 Hz, J = 2.4 Hz, 1H), 7.07 – 7.02 (m, 1H), 3.80 (s, 2H), 3.78 (s, 2H), 2.94 (t, J = 5.8 Hz, 2H), 2.83 (t, J = 5.6 Hz, 2H); ESI-MS m/z: 317.0[M+H]+.。
Example 22
6-fluoro-2- ((5-methoxypyridin-3-yl) methyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (22)
Figure DEST_PATH_IMAGE122
Compound 22 (67.2 mg, 68.3%) was obtained as a white solid from 3a (62.5 mg, 0.3 mmol) and 5-methoxynicotinaldehyde (49.5 mg, 0.36 mmol) in a similar manner to preparation 1. Melting point of 81.3-83.1oC; 1H NMR (400 MHz, CDCl3) δ 8.25 (d, J = 2.8 Hz, 1H), 8.20 (d, J = 1.2 Hz, 1H),7.67 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.30 (t, J = 2.4 Hz, 1H), 7.24 (dd, J = 9.6 Hz, J = 2.4 Hz, 1H), 7.06 – 7.01 (m, 1H), 3.86 (s, 3H), 3.77 (s, 2H), 3.76 (s, 2H), 2.93 (t, J = 5.6 Hz, 2H), 2.82 (t, J = 5.6 Hz, 2H); ESI-MS m/z: 329.0[M+H]+.。
Example 23
6-fluoro-2- ((5-methylpyridin-3-yl) methyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (23)
Figure DEST_PATH_IMAGE124
Compound 23 (63.5 mg, 67.8%) was obtained as a white solid from 3a (62.5 mg, 0.3 mmol) and 5-methyl nicotinaldehyde (43.6 mg, 0.36 mmol) in a similar manner to preparation 1. Melting point of 120.8-122.3oC; 1H NMR (400 MHz, CDCl3) δ 8.40 – 8.38 (m, 2H), 7.66 (dd, J = 8.4 Hz, J = 4.8 Hz, 1H), 7.60 (s, 1H), 7.22 (dd, J = 9.6 Hz, J = 2.4 Hz, 1H), 7.05 – 7.00 (m, 1H), 3.76 (s, 2H), 3.75 (s, 2H), 2.94 (t, J = 5.8 Hz, 2H), 2.82 (t, J = 5.6 Hz, 2H); 2.34 (s, 3H); ESI-MS m/z: 313.0[M+H]+.。
Example 24
6-fluoro-2- (pyrimidin-5-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (24)
Figure DEST_PATH_IMAGE126
Compound 24 (64.2 mg, 71.5%) was obtained as a white solid from 3a (62.5 mg, 0.3 mmol) and pyrimidine-5-carbaldehyde (38.9 mg, 0.36 mmol) in a similar manner to preparation 1. Melting point of 109.8-111.2oC; 1H NMR (400 MHz, CDCl3) δ 9.17 (s, 1H), 8.78 (s, 1H), 7.69 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.24 (dd, J = 9.2 Hz, J = 2.4 Hz, 1H), 7.07 – 7.02 (m, 1H), 3.80 (s, 2H), 3.79 (s, 2H), 2.95 (t, J = 5.8 Hz, 2H), 2.83 (t, J = 5.6 Hz, 2H); ESI-MS m/z: 300.0[M+H]+.。
Example 25
6-fluoro-2- (pyridin-4-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (25)
Figure DEST_PATH_IMAGE128
Compound 25 was obtained in a similar manner to preparation 1 from 3a (62.5 mg, 0.3 mmol) and isonicotinal (38.6 mg, 0.36 mmol) as a light yellow oil (64.5 mg, 71.8%).1H NMR (400 MHz, CDCl3) δ 8.60 (d, J = 5.6 Hz, 2H), 7.71 (dd, J = 8.4 Hz, J = 4.8 Hz, 1H), 7.37 (d, J = 6.0 Hz, 2H), 7.27 (dd, J = 9.6 Hz, J = 2.4 Hz, 1H), 7.08 – 7.03 (m, 1H), 3.79 (s, 4H), 2.95 (t, J = 5.8 Hz, 2H), 2.85 (t, J = 5.6 Hz, 2H); ESI-MS m/z: 299.0[M+H]+.。
Example 26
2- ((1H-imidazol-4-yl) methyl) -6-fluoro-1, 2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (26)
Figure DEST_PATH_IMAGE130
Compound 26 (37.8 mg, 43.8%) was obtained as a white solid from 3a (62.5 mg, 0.3 mmol) and 1H-imidazole-4-carbaldehyde (34.6 mg, 0.36 mmol) in a similar manner to preparation 1. Melting point: 173.6-175.7 oC; 1H NMR (400 MHz, CDCl3) δ 7.66 (dd, J = 8.8 Hz, J = 4.8 Hz, 1H), 7.62 (s, 1H), 7.22 (dd, J = 9.6 Hz, J = 2.4 Hz, 1H), 7.05 – 7.00 (m, 2H), 3.80 (s, 2H), 3.79 (s, 2H), 2.96 (t, J = 5.6 Hz, 2H), 2.81 (t, J = 5.6 Hz, 2H); ESI-MS m/z: 288.0[M+H]+.。
Example 27
6-fluoro-2- (isoquinolin-4-ylmethyl) -1,2,3, 4-tetrahydrobenzo [4,5] thieno [2,3-c ] pyridine (27)
Figure DEST_PATH_IMAGE132
Compound 27 (66.5 mg, 63.6%) was obtained as a white solid from 3a (62.5 mg, 0.3 mmol) and isoquinoline-4-carbaldehyde (56.6 mg, 0.36 mmol) in a similar manner to preparation 1. Melting point of 160.8-162.1oC; 1H NMR (400 MHz, CDCl3) δ 9.25 (s, 1H), 8.51 (s, 1H), 8.35 (d, J = 8.4 Hz, 1H), 8.02 (d, J = 8.0 Hz, 1H), 7.77 – 7.62 (m, 1H), 7.22 (dd, J = 9.2 Hz, J = 2.4 Hz, 1H), 7.05 – 7.00 (m, 1H), 4.16 (s, 2H), 3.84 (s, 2H), 3.03 (t, J = 5.8 Hz, 2H), 2.82 (t, J = 5.8 Hz, 2H); ESI-MS m/z: 349.0[M+H]+.。
EXAMPLE 28 pharmacological test
Preparation of mouse/human CYP17 protein
Such as previously reported [ J. Steroid biochem. mol. biol. 42 (1992) 313-320.]The preparation of rat testis microsome can be specifically referred to the following operations: at 4oTestis from Sprague Dawley male rats weighing 220-250g (60 days old) was minced and placed in STKM buffer (0.25M sucrose, 50mM Tris-HCl, 2.5mM KCl, 50mM MgCl)2) Then, the mixture was centrifuged at 9000g for 30 minutes, the resulting supernatant was further centrifuged at 105,000g for 1 hour, and the pellet was resuspended in STKM buffer and then stored at-80oC. Prior to use, the protein concentration of microsomes can be determined using an enhanced BCA protein assay kit (P0010; Beyotime Biotechnology, Shanghai, China) according to the manufacturer's protocol. The human Cyp17 gene was cloned and expressed in the adenovirus expression system of the a549 cell line. The purified cell membrane preparation was used as a source of human Cyp17 enzyme. Then stored at-80oC, prior to use, membrane protein concentrations can be determined using an enhanced BCA protein assay kit (P0010; Beyotime Biotechnology, Shanghai, China) according to the manufacturer's protocol.
Determination of human and rat-cytochrome P450, 17-20 lyase activity:
the enzymatic activity of CYP17 was determined by measuring the conversion of 17 alpha-hydroxyprogesterone to androstenedione. The assay was performed as follows: mu.g of microsome/membrane protein in 25. mu.L of phosphate buffer (0.05M, pH 7.5) and inhibitor in 25oPre-incubation for 5 min under C. Then 25. mu.L of substrate-containing phosphate buffer (10. mu.M 17. alpha. -hydroxyprogesterone, 4.2 mM NADPH) was added to the reaction mixture at 25oIncubate for 60 min under C. Subsequently, the reaction was stopped by adding methanol (250 uL). Quantifying the concentration of the reaction product androstenedione by LC/MS;
Figure DEST_PATH_IMAGE134
sample preparation: an enzymatic reaction in the presence of an inhibitor;
positive control: enzyme reactions without inhibitors but containing 1% final concentration of DMSO;
blank: contains all reagents except enzyme;
100% -% inhibition of lyase Activity
Dose-response studies were performed with the standard compound abiraterone as part of experimental optimisation;
to measure IC50Plot% inhibition as a function of inhibitor concentration; fitting the data into an S-shaped equation by using Graphpad software IV to obtain IC50The value is obtained.
The results of the above example compounds using the above test are shown in table 1.
TABLE 1
Figure DEST_PATH_IMAGE136

Claims (6)

1.式(I)化合物或其药学上可接受的盐:1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure FDA0002985140190000011
Figure FDA0002985140190000011
在式(I)中:In formula (I): R1选自:氢、卤素、-OH、-CN;R 1 is selected from: hydrogen, halogen, -OH, -CN; -ORa,Ra独立地为C1-4氟代烷基;-ORa, Ra is independently C1-4 fluoroalkyl; 被0至4个Ra取代的C1-C6烷基,被0至4个Ra取代的C3-6环烷基,其中每个Ra独立地为卤素、-OH、CN;C1-C6 alkyl substituted by 0 to 4 Ra, C3-6 cycloalkyl substituted by 0 to 4 Ra, wherein each Ra is independently halogen, -OH, CN; R2选自:
Figure FDA0002985140190000012
其中Ra独立地为卤素、-OH、CN、C3-6环烷基、C1-4氟代烷基,Rc为卤素,A为苯基。
R 2 is selected from:
Figure FDA0002985140190000012
wherein Ra is independently halogen, -OH, CN, C3-6 cycloalkyl, C1-4 fluoroalkyl, Rc is halogen, and A is phenyl.
2.如权利要求1所述的化合物或其药学上可接受的盐,其中R1为氢、甲基、F、Cl、Br、-OH、-CN。2. The compound of claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl, F, Cl, Br, -OH, -CN. 3.一种具有如下结构的化合物或其药学上可接受的盐,其中所述化合物选自:3. A compound having the following structure or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
Figure FDA0002985140190000013
Figure FDA0002985140190000013
Figure FDA0002985140190000021
Figure FDA0002985140190000021
4.一种如权利要求1所述的式(I)化合物的制备方法,其特征在于按下述方案:4. a preparation method of formula (I) compound as claimed in claim 1 is characterized in that by following scheme: 方案1plan 1
Figure FDA0002985140190000022
其中X1-R2为卤代物;
Figure FDA0002985140190000022
Wherein X 1 -R 2 is halide;
或方案2or option 2
Figure FDA0002985140190000031
其中X1-R2为卤代物;
Figure FDA0002985140190000031
Wherein X 1 -R 2 is halide;
或方案3or option 3
Figure FDA0002985140190000032
其中R1-X2和R1-X1为卤代化合物,R1-B(OR3)2为硼酸酯,N为硼酸或者硼酸酯类中间体;
Figure FDA0002985140190000032
wherein R 1 -X 2 and R 1 -X 1 are halogenated compounds, R 1 -B(OR 3 ) 2 is boronic acid ester, and N is boronic acid or boronic ester intermediate;
或方案4or option 4
Figure FDA0002985140190000033
Figure FDA0002985140190000033
式中R1、R2的定义同权利要求1所述。The definitions of R 1 and R 2 in the formula are the same as those described in claim 1 .
5.一种药物组合物,其特征在于,其包含:药学上可接受的载体和权利要求1-3中任一项的化合物或其药学上可接受的盐。5. A pharmaceutical composition, characterized in that it comprises: a pharmaceutically acceptable carrier and the compound of any one of claims 1-3 or a pharmaceutically acceptable salt thereof. 6.权利要求1-3中任一项的化合物或其药学上可接受盐在用于制备治疗癌症的药物中的用途,所述癌症是前列腺癌、乳腺癌或卵巢癌。6. Use of a compound of any one of claims 1-3, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of cancer, the cancer being prostate cancer, breast cancer or ovarian cancer.
CN201611258939.3A 2016-12-30 2016-12-30 Substituted benzothieno[2,3-c]tetrahydropyridine derivatives and preparation method and use thereof Active CN108264509B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201611258939.3A CN108264509B (en) 2016-12-30 2016-12-30 Substituted benzothieno[2,3-c]tetrahydropyridine derivatives and preparation method and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201611258939.3A CN108264509B (en) 2016-12-30 2016-12-30 Substituted benzothieno[2,3-c]tetrahydropyridine derivatives and preparation method and use thereof

Publications (2)

Publication Number Publication Date
CN108264509A CN108264509A (en) 2018-07-10
CN108264509B true CN108264509B (en) 2021-05-04

Family

ID=62754752

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201611258939.3A Active CN108264509B (en) 2016-12-30 2016-12-30 Substituted benzothieno[2,3-c]tetrahydropyridine derivatives and preparation method and use thereof

Country Status (1)

Country Link
CN (1) CN108264509B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101965186A (en) * 2007-11-28 2011-02-02 塞阔伊亚药品公司 Compositions and methods for inhibiting cytochrome p450 2d6
CN102803250A (en) * 2009-06-26 2012-11-28 诺瓦提斯公司 1, 3-disubstituted imidazolidin-2-one derivatives as inhibitors of CYP 17
CN103108871A (en) * 2010-09-16 2013-05-15 诺瓦提斯公司 17a-hydroxylase/c17,20-lyase inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101965186A (en) * 2007-11-28 2011-02-02 塞阔伊亚药品公司 Compositions and methods for inhibiting cytochrome p450 2d6
CN102803250A (en) * 2009-06-26 2012-11-28 诺瓦提斯公司 1, 3-disubstituted imidazolidin-2-one derivatives as inhibitors of CYP 17
CN103108871A (en) * 2010-09-16 2013-05-15 诺瓦提斯公司 17a-hydroxylase/c17,20-lyase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
"STN检索报告";来源于ChemBridge Corporation提供的产品目录;《数据库REGISTRY(在线)》;20150618;CAS号:1783166-73-3等 *

Also Published As

Publication number Publication date
CN108264509A (en) 2018-07-10

Similar Documents

Publication Publication Date Title
TWI530499B (en) Benzimidazolone derivatives as bromodomain inhibitors
DK3157915T3 (en) Substituted dihydroisoquinolinone compounds
JP6254169B2 (en) Benzamide and heterobenzamide compounds
TW202122389A (en) Heterocyclic rip1 kinase inhibitors
BR112021008991A2 (en) 2,3-dihydro-1h-pyrrolo[3,4c]pyridin-1-one derivatives as hpk1 inhibitors for cancer treatment
AU2016217874A1 (en) Substituted mono- and polyazanaphthalene derivatives and their use
CA2596015A1 (en) Fused heterocyclic compounds useful as inhibitors of histone deacetylase
AU2018380132A1 (en) Tubulin inhibitors
CN103313970B (en) New bicyclo formula compound or its salt
WO2008109613A1 (en) Benzo[c][2,7]naphthyridine derivatives, and their use as kinase inhibitors
WO2014079364A1 (en) Imidazolone derivatives, pharmaceutical compositions and uses thereof
JP2022521537A (en) Imidazopyridinyl compounds and their use for the treatment of proliferative disorders
MX2013006284A (en) Substituted imidazoquinoline derivatives.
RU2579513C2 (en) Inhibitors of akt activity
US9266880B2 (en) Substituted azaindazole compounds
US9145419B2 (en) Imidazopyridazinyl compounds
CN103833756B (en) One-class pyridazinone compounds and its production and use
JP7357146B2 (en) Azaheteroaryl compounds and their uses
JP2010505922A (en) Novel imidazolone derivatives, their production as pharmaceuticals, pharmaceutical compositions and their use as protein kinase inhibitors, in particular as CDC7 inhibitors
US20200055847A1 (en) Modulators of hedgehog (hh) signalling pathway
WO2019043372A1 (en) Substitutes sulfonyl azacycles as modulators of hedgehog (hh) signalling pathway
CA3078942A1 (en) Pyrrolotriazine compounds and methods of inhibiting tam kinases
CA3021185A1 (en) Fused heterocyclic compound
CN108264509B (en) Substituted benzothieno[2,3-c]tetrahydropyridine derivatives and preparation method and use thereof
EP2714667A2 (en) Aminooxazole inhibitors of cyclin dependent kinases

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