The present application relates to the field of medicinal chemistry, in particular to a pyridine derivative used as a lysine-specific demethylase 1 (LSD1) inhibitor, or a pharmaceutically acceptable salt thereof, a preparation method therefor and a use thereof.
Histone lysine-specific demethylase 1 (LSD1) is the first reported histone demethylase that, with the assistance of flavin adenine dinucleotide (FAD), can specifically recognize H3K4 and H3K9 substrates and remove their single-methyl or double-methyl modifications. At the same time, the LSD1 can form transcriptional regulatory complexes with multiple factors, and under the recruitment of transcription factors, it can reach a promoter region of a designated gene, thereby regulating gene expression.
Researches have shown that the LSD1 is related to various diseases, is highly expressed in various tumor cells, and is closely related to poor tumor prognosis. Downregulation of LSD1 expression or inhibition of its activity can significantly inhibit tumor cell growth. Therefore, developing an efficient and specific LSD1 inhibitor is of great significance for the treatment of diseases such as tumors.
The present application relates to a pyridine derivative as an LSD1 inhibitor, and particularly relates to a pyridine derivative as well as a preparation method therefor and an application thereof in medicines, in particular, a pyridine derivative as shown in formula I below and a use thereof in preparation of drugs for treating LSD1-mediated diseases, more specifically, a use in preparation of drugs suitable for tumors.
One aspect of the present application provides a pyridine derivative with a structure as shown in formula I below, and a stereoisomer or a pharmaceutically acceptable salt thereof:
and R1 and R2 are not hydrogen or fluorine at the same time, and when R1 is hydrogen, R2 is not fluorine, and when R1 is fluorine, R2 is not hydrogen; or R1 and R2 together with a carbon atom to which they are connected form a ring structure selected from
In some implementations, the ring A is selected from the group consisting of C4-C13 cycloalkyl and 3- to 13-membered heterocycloalkyl, and the heterocycloalkyl contains 1-3 heteroatoms selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom;
In some implementations, the ring B is selected from the group consisting of C5-C10 cycloalkenyl, 5- to 10-membered heterocycloalkenyl, C5-C10 aryl and 5- to 10-membered heteroaryl, and the heterocycloalkenyl or the heteroaryl contain 1-3 heteroatoms selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom;
In some implementations, R1 and R2 are each independently selected from the group consisting of hydrogen, cyano,
or R1 and R2 together with a carbon atom to which they are connected form a ring structure selected from
and R6 and R7 are each independently selected from the group consisting of hydrogen, C1-C6 alkyl and C3-C6 cycloalkyl;
(e.g., hydrogen, cyano or
R2 is selected from the group consisting of hydrogen cyano,
(e.g., hydrogen, cyano,
or R and R2 together with a carbon atom to which they are connected form a ring structure selected from
and R6 and R7 are each independently selected from the group consisting of hydrogen, C1-C5 alkyl (e.g., C1-C4 alkyl, and C1-C3 alkyl) and C3-C8 cycloalkyl (e.g., C3-C4 cycloalkyl and cyclopropyl).
In some implementations, m is 0, 1, 2, 3, 4 or 5, and R3 is selected from the group consisting of halogen (including fluorine, chlorine, bromine or iodine), oxo, hydroxyl, substituted or unsubstituted amino, C1-C6 alkyl and C1-C6 alkoxy, wherein the C1-C6 alkyl or the C1-C6 alkoxy is unsubstituted or is substituted with one or more substituents selected from the group consisting of halogen, cyano, hydroxyl and amino, and preferably, when m is 2 to 5, a plurality of R3 may be selected from different groups;
In some implementations, n is selected from the group consisting of 0, 1, 2, 3, 4 and 5, and R4 is selected from the group consisting of halogen (including fluorine, chlorine, bromine or iodine), oxo, hydroxyl, C1-C6 alkyl, C1-C6 alkoxy, C3-C6 cycloalkyl and C3-C6 cycloalkyloxy, wherein the C1-C6 alkyl, the C1-C6 alkoxy, the C3-C6 cycloalkyl or the C3-C6 cycloalkyloxy is unsubstituted or is substituted with one or more substituents selected from the group consisting of halogen, hydroxyl and amino; and preferably, when n is 2 to 5, a plurality of R4 may be selected from different groups;
In some implementations, q is selected from the group consisting of 0, 1 and 2, and R5 is selected from the group consisting of halogen, hydroxyl, amino, C1-C6 alkyl and C1-C6 alkoxy, wherein the C1-C6 alkyl or the C1-C6 alkoxy is unsubstituted or is substituted with one or more substituents selected from the group consisting of halogen and hydroxyl;
Preferably, in the compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof,
R1 and R2 are not hydrogen or fluorine at the same time, when R1 is hydrogen, R2 is not fluorine, and when R1 is fluorine, R2 is not hydrogen; or R1 and R2 together with a carbon atom to which they are connected form a ring structure selected from
Preferably, in the compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof,
R1 and R2 are not hydrogen or fluorine at the same time, when R1 is hydrogen, R2 is not fluorine, and when R1 is fluorine, R2 is not hydrogen; or R1 and R2 together with a carbon atom to which they are connected form a ring structure selected from
preferably, R1 and R2 are each independently selected from the group consisting of hydrogen, fluorine, cyano,
(e.g., R1 and R2 are each independently selected from the group consisting of hydrogen, fluorine, cyano,
R1 and R2 are not hydrogen or fluorine at the same time, when R1 is hydrogen, R2 is not fluorine, and when R1 is fluorine, R2 is not hydrogen; or R1 and R2 together with a carbon atom to which they are connected form a ring structure selected from
(e.g., R4 is selected from the group consisting of fluorine, chlorine, oxo, methyl, methoxy,
In some implementations, in the compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof,
or R1 and R2 together with a carbon atom to which they are connected form a ring structure selected from
Preferably, the compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof further have a structure shown in formula II,
R2 is selected from the group consisting of hydrogen, cyano,
and R6 and R7 are each independently selected from the group consisting of hydrogen, C1-C5 alkyl and C3-C8 cycloalkyl;
More preferably, a heterocyclic derivative with a structure shown in formula I, and a stereoisomer or a pharmaceutically acceptable salt thereof are selected from the group consisting of following compounds, and stereoisomers or pharmaceutically acceptable salts thereof:
The present application also covers solutions obtained through any combination, deletion, or exchange of the above implementations and preferred solutions.
Another aspect of the present application provides a preparation method for preparing the pyridine derivative with the structure shown in formula I above, and the method includes the following steps: method I:
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, N,N-dimethylformamide, N,N-dimethylacetamide, 1,4-dioxane and any combination thereof, preferably N,N-dimethylformamide. The reaction is preferably carried out with the presence of a suitable base and/or carried out with the presence of a transition metal catalyst and a ligand. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate, sodium carbonate and potassium tert-butoxide, preferably potassium carbonate. The transition metal catalyst may be selected from the group consisting of Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3, Pd(PPh3)4 and RuPhos-Pd-G3. The reaction is preferably carried out under a suitable temperature which is preferably 50° C. to 80° C. The reaction is preferably carried out for suitable time such as 2 to 8 hours.
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of acetonitrile, toluene, chloroform, tetrahydrofuran and any combination thereof, preferably acetonitrile. The reaction is preferably carried out with the presence of a suitable bromo reagent. The bromo reagent may be selected from the group consisting of N-bromosuccinimide, liquid bromine and pyridine bromine onium salt, preferably N-bromosuccinimide. The reaction is preferably carried out under a suitable temperature which is preferably 20° C. to 50° C. The reaction is preferably carried out for suitable time such as 2 to 6 hours.
(3) The Compound I-4 is Subjected to a Coupling Reaction with Boric Acid or Borate of a Compound I-5 to Obtain a Compound I-6
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1,4-dioxane, toluene, acetonitrile, ethanol, water and any combination thereof, preferably a combination of 1,4-dioxane and water. The reaction is preferably carried out with the presence of a suitable catalyst. The catalyst may be selected from the group consisting of Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3 and Pd(PPh3)4, preferably Pd(dppf)Cl2. The reaction is preferably carried out with the presence of a suitable base. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate and sodium carbonate, preferably potassium carbonate. The reaction is preferably carried out under a suitable temperature which is preferably 80° C. to 120° C. The reaction is preferably carried out for suitable time such as 8 to 12 hours.
(4) The Compound I-6 is Subjected to a Coupling Reaction with a Compound I-7 to Obtain the Compound with the Structure Shown in Formula I
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1,4-dioxane, toluene, acetonitrile, ethanol, water and any combination thereof, preferably a combination of 1,4-dioxane and water. The reaction is preferably carried out with the presence of a suitable catalyst. The catalyst may be selected from the group consisting of Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3 and Pd(PPh3)4, preferably Pd(dppf)Cl2. The reaction is preferably carried out with the presence of a suitable base. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate and sodium carbonate, preferably potassium carbonate. The reaction is preferably carried out under a suitable temperature which is preferably 80° C. to 120° C. The reaction is preferably carried out for suitable time such as 8 to 12 hours.
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of acetonitrile, toluene, chloroform, tetrahydrofuran and any combination thereof, preferably acetonitrile. The reaction is preferably carried out with the presence of a suitable bromo reagent. The bromo reagent may be selected from the group consisting of N-bromosuccinimide, liquid bromine and pyridine bromine onium salt, preferably N-bromosuccinimide. The reaction is preferably carried out under a suitable temperature which is preferably 20° C. to 50° C. The reaction is preferably carried out for suitable time such as 2 to 6 hours.
(2) The Compound I-8 is Subjected to a Coupling Reaction with Boric Acid or Borate of a Compound I-5 to Obtain a Compound I-9
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1,4-dioxane, toluene, acetonitrile, ethanol, water and any combination thereof, preferably a combination of 1,4-dioxane and water. The reaction is preferably carried out with the presence of a suitable catalyst. The catalyst may be selected from the group consisting of Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3 and Pd(PPh3)4, preferably Pd(dppf)Cl2. The reaction is preferably carried out with the presence of a suitable base. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate and sodium carbonate, preferably potassium carbonate. The reaction is preferably carried out under a suitable temperature which is preferably 80° C. to 120° C. The reaction is preferably carried out for suitable time such as 8 to 12 hours.
(3) The Compound I-9 is Subjected to a Coupling Reaction with a Compound I-7 to Obtain a Compound I-10
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1,4-dioxane, toluene, acetonitrile, ethanol, water and any combination thereof, preferably a combination of 1,4-dioxane and water. The reaction is preferably carried out with the presence of a suitable catalyst. The catalyst may be selected from the group consisting of Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3 and Pd(PPh3)4, preferably Pd(dppf)Cl2. The reaction is preferably carried out with the presence of a suitable base. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate and sodium carbonate, preferably potassium carbonate. The reaction is preferably carried out under a suitable temperature which is preferably 80° C. to 120° C. The reaction is preferably carried out for suitable time such as 8 to 12 hours.
(4) The Compound I-10 Reacts with a Compound I-2 to Obtain the Compound with the Structure Shown in Formula I
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, N,N-dimethylformamide, N,N-dimethylacetamide, 1,4-dioxane and any combination thereof, preferably N,N-dimethylformamide. The reaction is preferably carried out with the presence of a suitable base and/or carried out with the presence of a transition metal catalyst and a ligand. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate, sodium carbonate and potassium tert-butoxide, preferably potassium carbonate. The transition metal catalyst may be selected from the group consisting of Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3, Pd(PPh3)4 and RuPhos-Pd-G3. The reaction is preferably carried out under a suitable temperature which is preferably 50° C. to 80° C. The reaction is preferably carried out for suitable time such as 2 to 8 hours.
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1,4-dioxane, toluene, acetonitrile, ethanol, water and any combination thereof, preferably a combination of 1,4-dioxane and water. The reaction is preferably carried out with the presence of a suitable catalyst. The catalyst may be selected from the group consisting of Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3 and Pd(PPh3)4, preferably Pd(dppf)Cl2. The reaction is preferably carried out with the presence of a suitable base. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate and sodium carbonate, preferably potassium carbonate. The reaction is preferably carried out under a suitable temperature which is preferably 80° C. to 120° C. The reaction is preferably carried out for suitable time such as 8 to 12 hours.
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of acetonitrile, toluene, chloroform, tetrahydrofuran and any combination thereof, preferably acetonitrile. The reaction is preferably carried out with the presence of a suitable bromo reagent. The bromo reagent may be selected from the group consisting of N-bromosuccinimide, liquid bromine and pyridine bromine onium salt, preferably N-bromosuccinimide. The reaction is preferably carried out under a suitable temperature which is preferably 20° C. to 50° C. The reaction is preferably carried out for suitable time such as 2 to 6 hours.
(3) The Compound I-12 is Subjected to a Coupling Reaction with Boric Acid or Borate of a Compound I-5 to Obtain the Compound of Formula I
The reaction is preferably carried out in a suitable organic solvent. The organic solvent may be selected from the group consisting of tetrahydrofuran, 1,4-dioxane, toluene, acetonitrile, ethanol, water and any combination thereof, preferably a combination of 1,4-dioxane and water. The reaction is preferably carried out with the presence of a suitable catalyst. The catalyst may be selected from Pd(dppf)Cl2, Pd(OAc)2, Pd2(dba)3, Pd(PPh3)4, preferably Pd(dppf)Cl2. The reaction is preferably carried out with the presence of a suitable base. The base may be selected from the group consisting of triethylamine, pyridine, 4-dimethylaminopyridine, diisopropylethylamine, potassium carbonate, cesium carbonate and sodium carbonate, preferably potassium carbonate. The reaction is preferably carried out under a suitable temperature which is preferably 80° C. to 120° C. The reaction is preferably carried out for suitable time such as 8 to 12 hours.
The specific conditions for the above reaction steps are well-known in the art and are not specifically limited in the present application. As the teachings of the present application are combined with common knowledge in the art, those skilled in the art can select and replace each substituent in the general formula to prepare different compounds, all of which are within the scope of protection of the present application.
The present application further relates to a pharmaceutical composition, including the above compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof as well as a pharmaceutically acceptable excipient.
The present application further relates to a use of the above compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof, or the above pharmaceutical composition in preparation of drugs for preventing or treating a lysine-specific demethylase 1-related disease. Or, the present application further relates to a compound of formula I, and a stereoisomer or a pharmaceutically acceptable salt thereof, or the above pharmaceutical composition used for preventing or treating an LSD1-related disease. Or, the present application further relates to a method for preventing or treating an LSD1-related disease, including administrating to a subject in need the above compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof, or the above pharmaceutical composition.
In some embodiments, the LSD1-related disease is a tumor or a cancer, such as acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), lymphoma, malignant sarcoma, breast cancer, cervical cancer, colon cancer, lung cancer, oral cancer, brain cancer, stomach cancer, liver cancer, colorectal cancer, pancreatic cancer, skin cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, fallopian tube tumor, peritoneal tumor, melanoma, glioma, neuroblastoma, mastoid malignant tumor, head and neck tumor or myeloma.
The present application further relates to a use of the above compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof, or the above pharmaceutical composition in preparation of an LSD1 inhibitor. Or, the present application further relates to the above compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof, or the above pharmaceutical composition for use as an LSD1 inhibitor. Or, the present application further relates to a method for inhibiting LSD1, including administrating to a subject in need the above compound of formula I, and the stereoisomer or the pharmaceutically acceptable salt thereof, or the above pharmaceutical composition.
The present application discovers a type of brand-new-structure LSD1 inhibitor having a structure as shown in formula I, and the inhibitor has good inhibiting activity.
In order to make the aspects and technical solutions of the present application clearer, the following will further elaborate on the present application in conjunction with specific examples. It should be understood that these examples are merely used for illustrating the present application, instead of limiting the scope of the present application. Moreover, specific experimental methods not mentioned in the following examples are all carried out according to conventional experimental methods.
Unless otherwise stated, the terms used in the present application have the following meanings. A specific term should not be considered uncertain or unclear without a specific definition, but should be understood according to the common meaning in the art.
When covalent bonds in certain structural units or groups in the present application are not connected to specific atoms, it indicates that the covalent bonds may be connected to any atom in the structural units or groups, as long as they do not violate the rules of valent bond connection.
Herein, unless otherwise stated, “alkoxy” refers to —O-alkyl.
Herein, unless otherwise stated, “halogen” refers to fluorine, chlorine, bromine or iodine.
Herein, unless otherwise stated, “cycloalkyloxy” refers to —O-cycloalkyl.
Herein, unless otherwise stated, “saturated cycloalkyl” covers bridged rings in monocyclic and dicyclic forms, spiro in a dicyclic form, etc., and refers to fully saturated alicyclic hydrocarbon. For example, cycloalkyl herein may be C3-C10 monocyclic cycloalkyl, C5-C15 spiro cycloalkyl or C4-C12 bridged-ring cycloalkyl. Non-limiting instances of cycloalkyl include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, spiropentyl, spirohexyl, spiroheptyl, spiroheptyl, spirooctyl, spirononyl, spirodecyl, spiroundecyl, etc.
Herein, unless otherwise stated, “saturated heterocycloalkyl” refers to cycloalkyl in which one or more (such as 1-6, 1-5, 1-3, 1 or 2) carbon atoms are substituted with heteroatoms selected from the group consisting of N, O and S.
Herein, unless otherwise stated, “cyclohydrocarbyl” refers to hydrocarbyl with carbon atoms forming a ring, including saturated cyclohydrocarbyl and unsaturated cyclohydrocarbyl (such as aryl). Herein, unsaturated cyclohydrocarbyl is also sometimes referred to as unsaturated cycloalkyl. Herein, cyclohydrocarbyl may be a bridged ring in a monocyclic or dicyclic form, spiro in a dicyclic form, and a fused ring in a dicyclic form, such as C3-C10 monocyclic cyclohydrocarbyl, C5-C15 spiro cyclohydrocarbyl and C4-C12 bridged-ring cyclohydrocarbyl.
Herein, unless otherwise stated, “substituted or unsubstituted amino” covers unsubstituted amino or amino substituted with groups selected from the group consisting of C1-C6 alkyl, C1-C6 alkoxy, C3-C8 cycloalkyl and halogen.
The term “subject” is equivalent to “patient” and “individual” and represents humans or non-human animals (mammals, such as primates and rodents). “Mammals” include humans and livestock (such as laboratory mammals and household pets, such as cats, dogs, pigs, sheep, cows, sheep, goats, horses and rabbits), as well as non-domesticated mammals, such as wild mammals.
The term “treatment” means administering the compound or preparation described in the present application to alleviate or eliminate diseases or one or more symptoms related to the diseases, and includes inhibiting the progression of the diseases or symptoms, and alleviating the diseases or symptoms.
The term “pharmaceutically acceptable” refers to that a carrier, a vehicle, a diluent, an excipient, and/or a formed salt are/is typically chemically or physically compatible with other compositions that make up a drug formulation, and physiologically compatible with a receptor, without excessive toxicity, irritation, allergic reaction, or other issues or complications, commensurate with a reasonable benefit/risk ratio.
In the present application, the terms “including”, “comprising”, “containing” and their equivalents shall be understood as open and non-exclusive, meaning “including but not limited to”, which means that in addition to the listed elements, components, and steps, other unspecified elements, components, and steps may also be covered. Herein, unless the context otherwise specifies clearly, singular terms cover plural referents, and vice versa. Similarly, unless the context otherwise indicates clearly, the word “or” is intended to include “and”.
Unless otherwise stated, herein, parameter values representing the quantity, physicochemical properties, or reaction conditions of compositions should be understood to be modified by the term “about” in all cases. When the term “about” is used to describe the present application, the term “about” indicates the presence of error values, such as variations within a range of ±5%, such as ±1% or ±0.1%, of a certain specific value.
In the present application, when chemical names and structural formulas are inconsistent, the one shown in the structural formulas shall prevail, unless it can be inferred from the context that the chemical names rather than the structural formulas are correct.
Abbreviations Herein have the Following Meanings:
1H NMR
Structures of compounds are determined through mass spectrometry (MS) or proton nuclear magnetic resonance (1H NMR). Proton nuclear magnetic resonance (1H NMR) displacement (δ) is given with a unit of parts per million (ppm). The determination of proton nuclear magnetic resonance (1H NMR) adopts a BrukerAVANCE-400 NMR spectrometer, a determination solvent is deuterodimethyl sulfoxide (DMSO-d6), an internal standard is tetramethyl silane (TMS), and chemical displacement is given with a unit of 10−6 (ppm). The determination of mass spectrometry (MS) is performed using a FINNIGAN LCQAd (ESI) mass spectrometer (Manufacturer: Therm, Model: Finnigan LCQ advantage MAX). Thin layer silica gel uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plates. Column chromatography generally uses silica gel of 200-300 meshes from Yantai Huanghai silica gel as a carrier.
Unless otherwise stated in the present application, all reactions mentioned in the present application are carried out under a nitrogen atmosphere. The term “nitrogen atmosphere” in the present application refers to, for example, connecting a reaction flask to a 1 L volume nitrogen balloon.
The term “hydrogen atmosphere” in the present application refers to, for example, connecting a reaction flask to a 1 L volume hydrogen balloon.
Unless otherwise stated in the present application, solutions mentioned in reactions of the present application are aqueous solutions.
The term “room temperature” in the present application refers to a temperature between 10° C. and 25° C.
For the purpose of description and disclosure, all patents, patent applications, and other established publications are hereby expressly incorporated by reference. These publications are provided only because their publication occurred earlier than the filing date of the present application. All statements regarding the dates of these documents or expression of the contents of these documents is based on information available to the applicant and does not constitute any recognition of the correctness of the dates or contents of these documents. Moreover, in any country, any reference to these publications herein does not constitute recognition that the publication becomes part of common knowledge in the art.
The present application will be explained in detail below through examples, but those skilled in the art can understand that the scope of protection of the present application is not limited to this. Those skilled in the art can make various modifications, changes, combinations, etc. to the implementations and examples of the present application without departing from the spirit or scope of the present application, and the adjusted solution obtained from this is also within the scope of protection of the present application.
A compound 1a (550 mg, 3.18 mmol), 4-tert-butoxycarbonyl aminopiperidine (643 mg, 3.21 mmol), and K2CO3 (658 mg, 4.76 mmol) were dissolved in N,N-dimethylformamide (10 mL), and this system was heated to 80° C. and stirred to react overnight. A reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 856 mg of a title compound, a yield being 80%.
The compound 1b (538 mg, 1.60 mmol) was dissolved in acetonitrile (15 mL), NBS (288 mg, 1.62 mmol) was slowly added into a system under an ice bath, and a reaction system was placed at the room temperature and stirred to react for 2 hours. The reaction system was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 542 mg of a title compound, a yield being 82%.
The compound 1c (231 mg, 0.56 mmol), 3-fluoro-4-methoxyphenylboronic acid (97 mg, 0.57 mmol) and K2CO3 (116 mg, 0.84 mmol) were dissolved in 1,4-dioxane (5 mL) and water (1 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (44 mg, 0.06 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 126 mg of a title compound, a yield being 49%.
The compound 1d (47 mg, 0.10 mmol), 3-fluoro-4-cyanophenylboronic acid (25 mg, 0.15 mmol) and K2CO3 (41 mg, 0.30 mmol) were dissolved in 1,4-dioxane (1 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (7 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 27 mg of a title compound, a yield being 49%.
The compound 1e (27 mg, 0.05 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 16 mg of a title compound, a yield being 73%.
LC-MS (ESI) m/z (M+H)+: 446.2
1H NMR (400 MHz, DMSO-d6) δ 7.83-7.77 (m, 1H), 7.53 (s, 1H), 7.37 (dd, J=10.8, 1.2 Hz, 1H), 7.21-7.10 (m, 3H), 6.98-6.93 (m, 1H), 4.34-4.25 (m, 2H), 3.84 (s, 3H), 3.08-2.98 (m, 2H), 2.89-2.79 (m, 1H), 1.82-1.73 (m, 2H), 1.25-1.14 (m, 2H).
A compound 1d (125 mg, 0.27 mmol) was dissolved in a methanol solution (1 mL), then dimethyl sulfoxide (1 mL) and NaOH (32 mg, 0.80 mmol) were added in sequence, H2O2 (153 mg, 1.35 mmol, 30% aqueous solution) was slowly added dropwise into a reaction system under stirring, and the reaction system was placed at 50° C. and stirred to react for 2 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 90 mg of a title compound, a yield being 69%.
The compound 2a (55 mg, 0.11 mmol), 3-fluoro-4-cyanophenylboronic acid (28 mg, 0.17 mmol) and K2CO3 (46 mg, 0.33 mmol) were dissolved in 1,4-dioxane (1 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (8 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 48 mg of a title compound, a yield being 77%.
The compound 2b (48 mg, 0.09 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 31 mg of a title compound, a yield being 79%.
LC-MS (ESI) m/z (M+H)+: 464.2
1H NMR (400 MHz, DMSO-d6) δ 7.80-7.72 (m, 2H), 7.45-7.40 (m, 1H), 7.31 (dd, J=10.8, 1.2 Hz, 1H), 7.14 (dd, J=8.0, 1.2 Hz, 1H), 7.02 (t, J=8.4 Hz, 1H), 6.95 (dd, J=12.4, 2.4 Hz, 1H), 6.88 (s, 1H), 6.88-6.72 (m, 1H), 4.32-4.23 (m, 2H), 3.80 (s, 3H), 3.02-2.92 (m, 2H), 2.86-2.77 (m, 1H), 1.85-1.73 (m, 2H), 1.26-1.13 (m, 2H).
A compound 3a (10.0 g, 36.51 mmol) was dissolved in dry tetrahydrofuran (110 mL), replacement with nitrogen was carried out, a system was placed at −50° C., LDA (20 mL, 40.00 mmol, 2 mol/L in THF/Heptane/Ethylbenzene) was slowly added dropwise into the system, and after dropwise adding was finished, the system was maintained at −50° C. and continued to be stirred to react for 2 hours. A reaction liquid was quickly poured into a measuring cup containing smashed dry ice and stirred, the reaction liquid slowly restored to the room temperature, the reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was directly used for the next step of reaction without further purification.
The compound 3b (12.0 g, 37.75 mmol) was dissolved in N,N-dimethylformamide (130 mL), then K2CO3 (7.9 g, 57.16 mmol) and iodomethane (8.1 g, 57.04 mmol) were added in sequence, and a reaction system was stirred at the room temperature to react for 1 hour. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 8.3 g of a title compound, a total yield in the two steps being 68%.
The compound 3c (4.3 g, 12.95 mmol) and Zn(CN)2 (4.56 g, 38.85 mmol) were dissolved in N,N-dimethylformamide (40 mL), replacement with nitrogen was carried out, Pd(PPh3)4 (750 mg, 0.65 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.0 g of a title compound, a yield being 33%.
The compound 3d (1.0 g, 4.33 mmol), 4-tert-butoxycarbonyl aminopiperidine (1.3 g, 6.49 mmol) and triethylamine (900 mg, 8.89 mmol) were dissolved in tetrahydrofuran (10 mL). A system was heated to 50° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 647 mg of a title compound, a yield being 38%.
The compound 3e (647 mg, 1.64 mmol) was added into a reaction flask and dissolved in methanol (10 mL), then ammonium hydroxide (10 drops) and Raney-Ni (267 mg) were added in sequence, replacement with nitrogen was carried out, and a reaction system was placed at the room temperature under a hydrogen atmosphere to react overnight. A reaction liquid was filtered, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 218 mg of a title compound, a yield being 36%.
The compound 3f (175 mg, 0.48 mmol) was dissolved in acetonitrile (4 mL), NBS (128 mg, 0.72 mmol) was slowly added into a system under an ice bath condition, and a reaction system was stirred to react for 1 hour with the ice bath condition maintained. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 210 mg of a title compound, a yield being 99%.
The compound 3g (120 mg, 0.27 mmol), 3-fluoro-4-methoxyphenylboronic acid (46 mg, 0.27 mmol) and K2CO3 (112 mg, 0.81 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (36 mg, 0.05 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 70° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 33 mg of a title compound, a yield being 25%.
The compound 3h (33 mg, 0.07 mmol), 3-fluoro-4-cyanophenylboronic acid (16 mg, 0.01 mmol) and K2CO3 (56 mg, 0.40 mmol) were dissolved in 1,4-dioxane (1 mL) and water (0.1 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (22 mg, 0.03 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 34 mg of a title compound, a yield being 88%.
The compound 3i (34 mg, 0.06 mmol) was dissolved in 1,4-dioxane (2 mL), then a hydrochloric acid-1,4-dioxane solution (2 mL, 4 M in 1,4-dioxane) was added, and a reaction system was stirred at the room temperature to react for 2 hours. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 12 mg of a title compound, a yield being 43%.
LC-MS (ESI) m/z (M+H)+: 476.2
1H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 7.84-7.78 (m, 1H), 7.45 (dd, J=10.8, 1.2 Hz, 1H), 7.24 (dd, J=8.0, 1.6 Hz, 1H), 7.18 (dd, J=12.0, 2.0 Hz, 1H), 7.12 (t, J=8.8 Hz, 1H), 6.95-6.90 (m, 1H), 4.49-4.37 (m, 2H), 4.15 (s, 2H), 3.83 (s, 3H), 3.06-2.95 (m, 2H), 2.87-2.76 (m, 1H), 2.03-1.93 (m, 1H), 1.86-1.74 (m, 2H), 1.39-1.33 (m, 1H).
A compound 1c (130 mg, 0.31 mmol), 1-methylindazole-5-boric acid (56 mg, 0.32 mmol) and K2CO3 (64 mg, 0.46 mmol) were dissolved in 1,4-dioxane (3 mL) and water (0.6 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (22 mg, 0.03 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 120 mg of a title compound, a yield being 82%.
The compound 4a (50 mg, 0.11 mmol), 3-fluoro-4-cyanophenylboronic acid (28 mg, 0.17 mmol) and K2CO3 (46 mg, 0.33 mmol) were added into a reaction flask and dissolved in 1,4-dioxane (1 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (8 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 20 mg of a title compound, a yield being 34%.
The compound 4b (20 mg, 0.04 mmol) was dissolved in ethyl acetate (0.5 mL), then a hydrochloric acid-ethyl acetate solution (0.5 mL, 3 M) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 15 mg of a title compound, a yield being 92%.
LC-MS (ESI) m/z (M+H)+: 452.2
1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J=0.8 Hz, 1H), 7.75-7.70 (m, 1H), 7.69-7.67 (m, 1H), 7.63-7.59 (m, 1H), 7.57 (s, 1H), 7.38 (dd, J=10.8, 1.2 Hz, 1H), 7.18-7.12 (m, 2H), 4.38-4.28 (m, 2H), 4.05 (s, 3H), 3.10-3.00 (m, 2H), 2.91-2.81 (m, 1H), 1.85-1.74 (m, 2H), 1.26-1.16 (m, 2H).
A compound 5a (1.0 g, 4.29 mmol) was dissolved in ethanol (5 mL), then ammonium hydroxide (5 mL) was added, and a reaction system was sealed and then heated to 70° C. and stirred to react overnight. The reaction system was cooled to the room temperature, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was directly used for the next step of reaction without purification.
The compound 5b (700 mg, 2.80 mmol) was dissolved in dichloromethane (10 mL), triethylamine (424 mg, 4.19 mmol) and di-tert-butyl dicarbonate (610 mg, 2.80 mmol) were added into a system in sequence under an ice bath, and a reaction system was placed at the room temperature and stirred to react overnight. Water was added into the system to quench the reaction, extraction was carried out 3 times with dichloromethane, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 500 mg of a title compound, a total yield in the two steps being 33%.
The compound 5c (500 mg, 1.43 mmol) was dissolved in isopropanol (5 mL), then Pd/C (50 mg, 10%) was added, air in a system was replaced with hydrogen, and then the system was stirred at the room temperature under a hydrogen atmosphere to react overnight for 4 hours. A reaction system was filtered and subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 200 mg of a title compound, a yield being 65%.
A compound 1a (7.0 g, 40.46 mmol) was dissolved in DMSO (70 mL), then DIEA (5.3 g, 41.09 mmol) and p-methoxybenzylamine (5.7 g, 41.55 mmol) were added in sequence, and a reaction system was placed at 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was directly used for the next step without purification.
The compound 5e (9.0 g, 32.88 mmol) was dissolved in dichloromethane (50 mL), then trifluoroacetic acid (50 mL) was added, and a reaction system was placed at the room temperature and stirred to react for 6 hours. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and an obtained crude product was purified using a thin layer silica gel plate to obtain 5.0 g of a title compound, a total yield in the two steps being 81%.
The compound 5f (5.0 g, 32.56 mmol) was dissolved in acetonitrile (50 mL), bromine (5.3 g, 33.13 mmol) was slowly added dropwise into a system under an ice bath, and a reaction system was placed at the room temperature and stirred to react for 6 hours. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 4.0 g of a title compound, a yield being 53%.
The compound 5g (2.0 g, 8.60 mmol) was dissolved in 1,4-dioxane (20 mL) and water (4 mL), then 3-fluoro-4-methoxyphenylboronic acid (1.5 g, 8.83 mmol) and K2CO3 (3.6 g, 26.05 mmol) were added in sequence, replacement with nitrogen was carried out, Pd(dppf)Cl2 (628 mg, 0.86 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react for 6 hours. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.5 g of a title compound, a yield being 63%.
The compound 5h (1.5 g, 5.40 mmol) was dissolved in 1,4-dioxane (15 mL) and water (3 mL), then 3-fluoro-4-cyanophenylboronic acid (2.7 g, 16.37 mmol) and K2CO3 (2.3 g, 16.64 mmol) were added in sequence, replacement with nitrogen was carried out, Pd(dppf)Cl2 (395 mg, 0.54 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react for 9 hours. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.5 g of a title compound, a yield being 77%.
The compound 5i (1.5 g, 4.14 mmol) was dissolved in acetonitrile (15 mL), then potassium iodide (4.1 g, 24.70 mmol) and isoamyl nitrite (2.9 g, 24.75 mmol) were added in sequence, and a reaction system was placed at 75° C. and stirred to react for 36 hours. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 700 mg of a title compound, a yield being 36%.
The compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then the compound 5d (48 mg, 0.22 mmol) and Cs2CO3 (108 mg, 0.33 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 80° C. and stirred to react for 3 hours. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 20 mg of a title compound, a yield being 34%.
The compound 5k (20 mg, 0.04 mmol) was dissolved in a 1,4-dioxane solution (2 mL), then a hydrochloric acid-1,4-dioxane solution (2 mL, 4 M in 1,4-dioxane) was added, and a reaction system was placed at the room temperature and stirred to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 8 mg of a title compound, a yield being 49%.
LC-MS (ESI) m/z (M+H)+: 462.2
A compound 6-1 (500 mg, 2.46 mmol) was dissolved in methanol (2 mL), then ammonium hydroxide (5 mL) was added, and a system was sealed and then placed at the room temperature and stirred to react for 4 hours. A reaction system was cooled to the room temperature, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was directly used for the next step of reaction without purification.
The compound 6b (500 mg, 2.27 mmol) was dissolved in dichloromethane (10 mL), di-tert-butyl dicarbonate (495 mg, 2.27 mmol) was added into a system under an ice bath condition, and a reaction system was placed at the room temperature and stirred to react overnight. Water was added into the system to quench the reaction, extraction was carried out 3 times with dichloromethane, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 600 mg of a title compound, a total yield in the two steps being 76%.
The compound 6c (300 mg, 0.94 mmol) was dissolved in isopropanol (5 mL), then palladium hydroxide (30 mg, 10%) was added at the room temperature, air in a system was replaced with hydrogen, and then the system was placed at the room temperature under a hydrogen atmosphere and stirred overnight. A reaction system was filtered, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 100 mg of a title compound, a yield being 46%.
The compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then the compound 6d (51 mg, 0.22 mmol) and Cs2CO3 (108 mg, 0.33 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react for 3 hours. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 22 mg of a title compound, a yield being 36%.
The compound 6e (22 mg, 0.04 mmol) was dissolved in a 1,4-dioxane solution (2 mL), then a hydrochloric acid-1,4-dioxane solution (2 mL, 4 M in 1,4-dioxane) was added, and a reaction system was placed at the room temperature and stirred to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 10 mg of a title compound, a yield being 55%.
LC-MS (ESI) m/z (M+H)+: 476.2
1H NMR (400 MHz, DMSO-d6) δ 7.86-7.77 (m, 1H), 7.60-7.53 (m, 1H), 7.43-7.36 (m, 1H), 7.23-7.10 (m, 3H), 7.00-6.92 (m, 1H), 4.60 (brs, 1H), 4.21-4.07 (m, 2H), 3.85 (s, 3H), 3.52-3.36 (m, 2H), 2.54 (s, 2H), 1.58-1.39 (m, 4H).
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then tert-butyl azetidin-3-ylcarbamate (38 mg, 0.22 mmol) and Cs2CO3 (108 mg, 0.33 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react for 3 hours. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 22 mg of a title compound, a yield being 40%.
The compound 7a (22 mg, 0.04 mmol) was dissolved in dichloromethane (2 mL), then trifluoroacetic acid (1 mL) was added, and a reaction system was placed at the room temperature and stirred to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 15 mg of a title compound, a yield being 85%.
LC-MS (ESI) m/z (M+H)+: 418.2
1H NMR (400 MHz, DMSO-d6) δ 7.81 (dd, J=8.0, 6.8 Hz, 1H), 7.37 (dd, J=10.4, 1.2 Hz, 1H), 7.19-7.10 (m, 3H), 7.09 (s, 1H), 6.97-6.91 (m, 1H), 4.27-4.19 (m, 2H), 3.89-3.81 (m, 4H), 3.70-3.63 (m, 2H).
A compound 3f (287 mg, 0.78 mmol), 3-fluoro-4-cyanophenylboronic acid (193 mg, 1.17 mmol) and K2CO3 (647 mg, 4.68 mmol) were dissolved in 1,4-dioxane (4 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (29 mg, 0.04 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 100° C. and stirred to react for 2 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 315 mg of a title compound, a yield being 89%.
The compound 8a (310 mg, 0.69 mmol) was dissolved in acetonitrile (12 mL), NBS (246 mg, 1.38 mmol) was slowly added into a system under an ice bath condition, and a reaction system was stirred to react for 1 hour with the ice bath condition maintained. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 364 mg of a title compound, a yield being 100%.
The compound 8b (70 mg, 0.13 mmol), 1-methylindazole-5-boric acid (35 mg, 0.20 mmol) and K2CO3 (108 mg, 0.78 mmol) were dissolved in 1,4-dioxane (4 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 100° C. and stirred to react for 1 hour. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 57 mg of a title compound, a yield being 74%.
The compound 8c (57 mg, 0.10 mmol) was dissolved in dichloromethane (3 mL), then trifluoroacetic acid (3 mL) was added at 0° C., and a reaction system was maintained at 0° C. to be stirred to react for 2 hours. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 15 mg of a title compound, a yield being 32%.
LC-MS (ESI) m/z (M+H)+: 482.2
1H NMR (400 MHz, DMSO-d6) δ 8.60 (s, 1H), 8.02 (s, 1H), 7.77-7.70 (m, 1H), 7.68 (s, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.49 (d, J=10.4 Hz, 1H), 7.20 (d, J=8.0 Hz, 1H), 7.16 (d, J=8.4 Hz, 1H), 4.61-4.51 (m, 2H), 4.15 (s, 2H), 4.06 (s, 3H), 3.30-3.17 (m, 1H), 3.07-2.95 (m, 2H), 2.04-1.92 (m, 2H), 1.72-1.58 (m, 2H).
A compound 9a (430 mg, 2.00 mmol) was dissolved in N,N-dimethylformamide (10 mL), then Cs2CO3 (2.0 g, 6.14 mmol) and 1,1-dimethyl ethylene oxide (216 mg, 3.00 mmol) were added in sequence, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 488 mg of a title compound, a yield being 85%.
The compound 9b (286 mg, 1.00 mmol), bis(pinacolato)diboron (381 mg, 1.50 mmol) and potassium acetate (196 mg, 0.10 mmol) were dissolved in N,N-dimethylformamide (5 mL), replacement with nitrogen was carried out, triphenylphosphine (52 mg, 0.20 mmol) and Pd(AcO)2 (23 mg, 0.10 mmol) were added in sequence, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 296 mg of a title compound, a yield being 89%.
A compound 8b (110 mg, 0.21 mmol), the compound 9c (107 mg, 0.32 mmol) and K2CO3 (175 mg, 1.27 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react for 5 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 12 mg of a title compound, a yield being 9%.
The compound 9d (12 mg, 0.02 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 6 mg of a title compound, a yield being 59%.
LC-MS (ESI) m/z (M+H)+: 558.2
1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 1H), 8.08 (s, 1H), 7.86 (d, J=7.2 Hz, 1H), 7.80-7.72 (m, 1H), 7.54 (d, J=10.8 Hz, 1H), 7.44 (d, J=10.4 Hz, 1H), 7.27 (d, J=8.4 Hz, 1H), 4.65 (s, 1H), 4.55-4.42 (m, 2H), 4.40-4.22 (m, 3H), 3.96-3.87 (m, 1H), 3.11-2.98 (m, 2H), 2.87-2.75 (m, 1H), 1.89-1.75 (m, 2H), 1.44-1.32 (m, 2H), 1.16 (s, 3H), 1.10 (s, 3H).
A compound 4a (190 mg, 0.41 mmol) was dissolved in a methanol solution (4 mL), then dimethyl sulfoxide (2 mL), NaOH (49 mg, 1.23 mmol) and H2O2 (232 mg, 2.05 mmol, 30% aqueous solution) were added in sequence, and a reaction system was placed at 50° C. and stirred to react for 2 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 126 mg of a title compound, a yield being 64%.
The compound 10a (100 mg, 0.21 mmol), 3-fluoro-4-cyanophenylboronic acid (53 mg, 0.32 mmol) and K2CO3 (87 mg, 0.63 mmol) were dissolved in 1,4-dioxane (4 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 95 mg of a title compound, a yield being 81%.
The compound 10b (95 mg, 0.17 mmol) was dissolved in ethyl acetate (2 mL), then a hydrochloric acid-ethyl acetate solution (2 mL, 3 M in EA) was added, and a reaction system was placed at the room temperature and stirred to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 31 mg of a title compound, a yield being 40%.
LC-MS (ESI) m/z (M+H)+: 470.2
1H NMR (400 MHz, DMSO-d6) δ 7.98 (d, J=0.4 Hz, 1H), 7.73-7.64 (m, 2H), 7.51-7.46 (m, 2H), 7.34-7.31 (m, 1H), 7.31 (dd, J=9.2, 1.2 Hz, 1H), 7.10 (dd, J=8.0, 1.2 Hz, 1H), 7.10 (dd, J=8.4, 1.6 Hz, 1H), 6.91 (s, 1H), 4.35-4.25 (m, 2H), 4.02 (s, 3H), 3.03-2.93 (m, 2H), 2.89-2.79 (m, 1H), 1.85-1.74 (m, 2H), 1.29-1.16 (m, 2H).
A compound 11a (3.0 g, 15.63 mmol) was dissolved in chloroform (30 mL), thionyl chloride (18.6 g, 156.30 mmol) was slowly added into a system under an ice bath, and the system was heated to 70° C. and stirred to react for 3 hours. A reaction system was subjected to vacuum concentration and dissolved by tetrahydrofuran (10 mL), then was added dropwise into an ice-bath-cooled solution of ammonium hydroxide (30 mL) in tetrahydrofuran (10 mL), and was stirred to react for 30 minutes. Water was added into the reaction system, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 2.8 g of a title compound, a yield being 94%.
The compound 11b (2.8 g, 14.66 mmol), 4-tert-butoxycarbonyl aminopiperidine (4.9 g, 24.47 mmol) and triethylamine (3.0 g, 29.65 mmol) were dissolved in acetonitrile (30 mL), and a system was heated to 70° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 4.4 g of a title compound, a yield being 88%.
The compound 11c (400 mg, 1.13 mmol), 3-fluoro-4-cyanophenylboronic acid (280 mg, 1.70 mmol) and K2CO3 (937 mg, 6.78 mmol) were dissolved in 1,4-dioxane (5 mL) and water (0.5 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (41 mg, 0.06 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react for 4 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 448 mg of a title compound, a yield being 90%.
The compound 11d (448 mg, 1.02 mmol) was dissolved in N,N-dimethylformamide (5 mL), NBS (200 mg, 1.12 mmol) was slowly added into a system under an ice bath condition, and a reaction system was maintained at 0° C. to be stirred to react for 2 hours. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 528 mg of a title compound, a yield being 100%.
The compound 11e (518 mg, 1.00 mmol), 1-methylindazole-5-boric acid (263 mg, 1.49 mmol) and K2CO3 (829 mg, 6.00 mmol) were dissolved in 1,4-dioxane (6 mL) and water (0.6 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (37 mg, 0.05 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react for 3 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a silica gel column to obtain 423 mg of a title compound, a yield being 74%.
The compound 11f (30 mg, 0.05 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 6 mg of a title compound, a yield being 24%.
LC-MS (ESI) m/z (M+H)+: 470.2
1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1H), 8.00-7.95 (m, 1H), 7.79 (s, 1H), 7.79-7.73 (m, 1H), 7.68-7.62 (m, 2H), 7.58-7.54 (m, 1H), 7.50-7.45 (m, 1H), 7.22 (dd, J=8.4, 1.6 Hz, 1H), 7.22 (dd, J=8.8, 1.6 Hz, 1H), 4.04 (s, 3H), 3.91-3.80 (m, 2H), 3.02-2.90 (m, 2H), 2.84-2.73 (m, 1H), 1.86-1.74 (m, 2H), 1.43-1.32 (m, 2H).
A compound 11f (200 mg, 0.35 mmol) was dissolved in tetrahydrofuran (6 mL), replacement with nitrogen was carried out, pyridine (194 mg, 2.45 mmol) and a solution of trifluoroacetic anhydride (147 mg, 0.70 mmol) in dichloromethane (1 mL) were added in sequence under an ice bath, and a reaction system slowly restored to the room temperature and was stirred to react overnight. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 10 mg of a title compound, a yield being 5%.
The compound 12a (10 mg, 0.02 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 1 mg of a title compound, a yield being 12%.
LC-MS (ESI) m/z (M+H)+: 452.2
A compound 13-1 (1.0 g, 4.60 mmol) was dissolved in methanol (10 mL), ammonium acetate (2.5 g, 32.43 mmol) and sodium cyanoborohydride (375 mg, 5.97 mmol) were added in sequence, and a reaction system was placed at the room temperature and stirred to react overnight. Water was added into the reaction system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 500 mg of a title compound, a yield being 50%.
The compound 13b (500 mg, 2.29 mmol) was dissolved in ethyl acetate (5 mL), then a saturated sodium bicarbonate solution (10 mL) was added, benzyl chloroformate (391 mg, 2.29 mmol) was added into a system under an ice bath condition, and a reaction system was placed at the room temperature and stirred to react overnight. A reaction liquid was extracted 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 420 mg of a title compound, a yield being 52%.
The compound 13c (200 mg, 0.57 mmol) was dissolved in ethyl acetate (10 mL), then a hydrochloric acid-ethyl acetate solution (4 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration, and a crude product was directly used for the next step without purification.
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then the compound 13d (64 mg, 0.22 mmol) and t-BuONa (53 mg, 0.55 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react for 3 hours. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 21 mg of a title compound, a yield being 33%.
The compound 13e (21 mg, 0.04 mmol) was dissolved in ethyl acetate (2 mL), then Pd/C (2 mg, 10%) was added at the room temperature, air in a system was replaced with hydrogen, and then the system was heated to 35° C. under a hydrogen atmosphere and stirred to react for 4 hours. A reaction system was filtered and subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 10 mg of a title compound, a yield being 61%.
LC-MS (ESI) m/z (M+H)+: 464.2
1H NMR (400 MHz, DMSO-d6) δ 7.95-7.78 (m, 1H), 7.64 (s, 1H), 7.41 (dd, J=10.8, 1.6 Hz, 1H), 7.23-7.11 (m, 3H), 7.00-6.94 (m, 1H), 4.22-4.15 (m, 2H), 4.10-4.00 (m, 1H), 3.85 (s, 3H), 3.45-3.33 (m, 2H), 3.03-2.92 (m, 1H), 1.93-1.82 (m, 1H), 1.41-1.30 (m, 1H).
A compound 5j (80 mg, 0.17 mmol) was dissolved in 1,4-dioxane (3 mL) and water (0.5 mL), tert-butyl (4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)cyclohexyl-3-alken-1-yl)carbamate (165 mg, 0.51 mmol) and K2CO3 (70 mg, 0.51 mmol) were added in sequence, replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a system was heated to 80° C. and stirred to react for 2 hours. A reaction system was cooled to the room temperature, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 79 mg of a title compound, a yield being 86%.
The compound 14a (40 mg, 0.07 mmol) was dissolved in ethyl acetate (2 mL), then Pd/C (4 mg, 10%) was added at the room temperature, air in a system was replaced with hydrogen, and then the system was stirred at the room temperature under a hydrogen atmosphere to react overnight. A reaction system was filtered, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 23 mg of a title compound, a yield being 57%.
The compound 14b (23 mg, 0.04 mmol) was dissolved in ethyl acetate (5 mL), then a hydrochloric acid-ethyl acetate solution (4 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 11 mg of a title compound, a yield being 59%.
LC-MS (ESI) m/z (M+H)+: 445.2
1H NMR (400 MHz, DMSO-d6) δ 8.08 (s, 0.48H), 8.01 (s, 0.47H), 7.89-7.82 (m, 1H), 7.48-7.41 (m, 1H), 7.55-7.28 (m, 1H), 7.25-7.16 (m, 2H), 7.11-7.05 (m, 1H), 3.88-3.84 (m, 3H), 3.16-3.10 (m, 0.5H), 2.95-2.86 (m, 0.5H), 2.86-2.68 (m, 1H), 2.15-1.89 (m, 3H), 1.75-1.60 (m, 4H), 1.33-1.21 (m, 1H).
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), tert-butyl 1,4-diazacycloheptane-1-carboxylate (44 mg, 0.22 mmol) and Cs2CO3 (108 mg, 0.33 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a system was heated to 80° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, vacuum concentration was carried out to remove a solvent, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 24 mg of a title compound, a yield being 42%.
The compound 15a (24 mg, 0.04 mmol) was dissolved in ethyl acetate (5 mL), then a hydrochloric acid-ethyl acetate solution (4 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 10 mg of a title compound, a yield being 51%.
LC-MS (ESI) m/z (M+H)+: 446.2
1H NMR (400 MHz, DMSO-d6) δ 7.85-7.79 (m, 1H), 7.42-7.34 (m, 2H), 7.23-7.11 (m, 3H), 7.00-6.93 (m, 1H), 3.85 (s, 3H), 3.80-3.68 (m, 4H), 2.93-2.86 (m, 2H), 2.76-2.69 (m, 2H), 1.84-1.73 (m, 2H).
A compound 1b (1.7 g, 5.05 mmol), 3-fluoro-4-cyanophenylboronic acid (1.2 g, 7.28 mmol) and K2CO3 (2.1 g, 15.20 mmol) were dissolved in acetonitrile (20 mL) and water (10 mL), replacement with nitrogen was carried out, Pd(PPh3)4 (289 mg, 0.25 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.8 g of a title compound, a yield being 85%.
The compound 16a (421 mg, 1.00 mmol) was dissolved in acetonitrile (15 mL), NBS (182 mg, 1.02 mmol) was slowly added into a system under an ice bath, and a reaction system was placed at the room temperature and stirred to react for 3 hours. The reaction system was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 450 mg of a title compound, a yield being 90%.
The compound 16b (100 mg, 0.20 mmol), a compound 9c (100 mg, 0.30 mmol) and K2CO3 (83 mg, 0.60 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 30 mg of a title compound, a yield being 24%.
The compound 16c (30 mg, 0.05 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 24 mg of a title compound, a yield being 95%.
LC-MS (ESI) m/z (M+H)+: 528.2
1H NMR (400 MHz, DMSO-d6) δ 8.11 (s, 1H), 7.80-7.72 (m, 2H), 7.61 (s, 1H), 7.57 (d, J=10.8 Hz, 1H), 7.40-7.34 (m, 1H), 7.18 (dd, J=8.0, 1.2 Hz, 1H), 4.68 (s, 1H), 4.40-4.24 (m, 4H), 3.12-3.01 (m, 2H), 2.92-2.82 (m, 1H), 1.85-1.75 (m, 2H), 1.27-1.19 (m, 2H), 1.14 (s, 3H), 1.10 (s, 3H).
A compound 1c (229 mg, 0.55 mmol), a compound 9c (368 mg, 1.10 mmol) and K2CO3 (228 mg, 1.65 mmol) were dissolved in 1,4-dioxane (5 mL) and water (1 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (44 mg, 0.06 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 138 mg of a title compound, a yield being 46%.
The compound 17a (138 mg, 0.25 mmol) was dissolved in a methanol solution (1 mL), then dimethyl sulfoxide (1 mL), NaOH (30 mg, 0.75 mmol) and H2O2 (170 mg, 1.50 mmol, 30% aqueous solution) were added in sequence, and a reaction system was placed at 50° C. and stirred to react for 2 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 126 mg of a title compound, a yield being 88%.
The compound 17b (126 mg, 0.22 mmol), 3-fluoro-4-cyanophenylboronic acid (54 mg, 0.33 mmol) and K2CO3 (91 mg, 0.66 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 74 mg of a title compound, a yield being 51%.
The compound 17c (74 mg, 0.11 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 24 mg of a title compound, a yield being 38%.
LC-MS (ESI) m/z (M+H)+: 546.2
1H NMR (400 MHz, DMSO-d6) δ 8.03 (d, J=0.8, 1H), 7.85 (s, 1H), 7.25-7.67 (m, 1H), 7.59 (d, J=7.2, 1H), 7.42-7.34 (m, 2H), 7.27 (dd, J=10.4, 1.2 Hz, 1H), 7.16 (dd, J=8.0, 1.6 Hz, 1H), 6.96 (s, 1H), 4.66 (s, 1H), 4.37-4.19 (m, 4H), 3.04-2.94 (m, 2H), 2.88-2.48 (m, 1H), 1.85-1.75 (m, 2H), 1.27-1.19 (m, 2H), 1.13 (s, 3H), 1.07 (s, 3H).
A compound 18b (75 mg, 0.15 mmol), 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)indoline-2-one (63 mg, 0.23 mmol) and K2CO3 (62 mg, 0.46 mmol) were dissolved in 1,4-dioxane (1 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 36 mg of a title compound, a yield being 42%.
The compound 18a (36 mg, 0.06 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 18 mg of a title compound, a yield being 61%.
LC-MS (ESI) m/z (M+H)+: 467.2
1H NMR (400 MHz, DMSO-d6) δ 7.77 (dd, J=8.0, 7.2 Hz, 1H), 7.53 (s, 1H), 7.39 (dd, J=10.8, 1.2 Hz, 1H), 7.19-7.12 (m, 2H), 7.09-7.07 (m, 1H), 7.00-6.96 (m, 1H), 4.34-4.26 (m, 2H), 3.51 (s, 2H), 3.12 (s, 3H), 3.08-2.98 (m, 2H), 2.89-2.80 (m, 1H), 1.82-1.74 (m, 2H), 1.26-1.17 (m, 2H).
A compound 19a (1.0 g, 4.29 mmol) was dissolved in acetonitrile (40 mL), then K2CO3 (1.8 g, 13.02 mmol) and iodomethane (1.5 g, 10.57 mmol) were added in sequence, and a reaction system was heated to 60° C. and stirred to react for 3 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.0 g of a title compound, a yield being 94%.
The compound 19b (815 mg, 3.30 mmol) was dissolved in dry tetrahydrofuran (30 mL), then methyl magnesium bromide (5.5 mL, 3 M in THF) was slowly added dropwise under an ice bath, and after dropwise adding was finished, the temperature of a reaction system slowly returned to the room temperature, and the reaction system was stirred to react overnight. The reaction system was cooled to 0° C., an appropriate amount of water was slowly added to quench the reaction, filtering was carried out, a filtrate was extracted 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 366 mg of a title compound, a yield being 45%.
The compound 19c (360 mg, 1.46 mmol), bis(pinacolato)diboron (741 mg, 2.92 mmol) and potassium acetate (429 mg, 4.37 mmol) were dissolved in 1,4-dioxane (15 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (107 mg, 0.15 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react for 24 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 177 mg of a title compound, a yield being 41%.
A compound 16b (100 mg, 0.20 mmol), the compound 19d (118 mg, 0.40 mmol) and K2CO3 (83 mg, 0.60 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 50 mg of a title compound, a yield being 43%.
The compound 19e (50 mg, 0.09 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 36 mg of a title compound, which was a yellow solid, a yield being 87%.
LC-MS (ESI) m/z (M+H)+: 488.2
1H NMR (400 MHz, DMSO-d6) δ 7.82-7.76 (m, 1H), 7.59 (s, 1H), 7.27-7.17 (m, 3H), 7.08 (s, 1H), 7.07-7.04 (m, 1H), 4.56 (s, 1H), 4.37-4.27 (m, 2H), 3.10-3.00 (m, 2H), 2.91-2.80 (m, 1H), 2.68 (s, 2H), 1.85-1.74 (m, 2H), 1.27-1.17 (m, 2H), 1.04 (s, 3H), 1.03 (s, 3H).
A compound 1a (1.5 g, 8.67 mmol), tert-butyl 1,4-diazacycloheptane-1-formate (1.7 g, 8.49 mmol), and K2CO3 (2.4 g, 17.36 mmol) were dissolved in acetonitrile (20 mL), and this system was heated to 80° C. and stirred to react overnight. A reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 2.1 g of a title compound, a yield being 72%.
The compound 20a (1.0 g, 2.97 mmol), 3-fluoro-4-cyanophenylboronic acid (735 mg, 4.46 mmol) and Na2CO3 (630 mg, 5.94 mmol) were dissolved in acetonitrile (10 mL) and water (5 mL), replacement with nitrogen was carried out, Pd(PPh3)4 (173 mg, 0.15 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 85° C. and stirred to react for 6 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 800 mg of a title compound, a yield being 64%.
The compound 20b (500 mg, 1.19 mmol) was dissolved in acetonitrile (5 mL), NBS (211 mg, 1.19 mmol) was slowly added into a system under an ice bath, and a reaction system was placed at the room temperature and stirred to react for 3 hours. The reaction system was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 420 mg of a title compound, a yield being 71%.
The compound 20c (50 mg, 0.10 mmol), 1-methylindazole-5-boric acid (53 mg, 0.30 mmol) and K2CO3 (41 mg, 0.30 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (7 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 80° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 30 mg of a title compound, a yield being 54%.
The compound 20d (30 mg, 0.05 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (2 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 20 mg of a title compound, a yield being 82%.
LC-MS (ESI) m/z (M+H)+: 452.2
1H NMR (400 MHz, DMSO-d6) δ 8.07-8.05 (m, 1H), 7.75-7.70 (m, 1H), 7.69-7.66 (m, 1H), 7.63-7.59 (m, 1H), 7.40-7.35 (m, 2H), 7.18-7.13 (m, 2H), 4.06 (s, 3H), 3.83-3.67 (m, 4H), 2.93-2.87 (m, 2H), 2.76-2.69 (m, 2H), 1.85-1.74 (m, 2H).
A compound 20c (50 mg, 0.10 mmol), a compound 9c (100 mg, 0.30 mmol) and K2CO3 (41 mg, 0.30 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (7 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 80° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 10 mg of a title compound, a yield being 16%.
The compound 21a (10 mg, 0.02 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (2 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 4 mg of a title compound, a yield being 48%.
LC-MS (ESI) m/z (M+H)+: 528.2
1H NMR (400 MHz, DMSO-d6) δ 8.11 (s, 1H), 7.79-7.73 (m, 2H), 7.58 (d, J=10.4 Hz, 1H), 7.41 (s, 1H), 7.39-7.33 (m, 1H), 7.21-7.16 (m, 1H), 4.67 (s, 1H), 4.32-4.23 (m, 2H), 3.88-3.67 (m, 4H), 2.94-2.85 (m, 2H), 2.76-2.68 (m, 2H), 1.86-1.72 (m, 2H), 1.15 (s, 3H), 1.10 (s, 3H).
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then 2-tert-butoxycarbonyl-2,7-diazaspiro[3.5]nonane (50 mg, 0.22 mmol) and Cs2CO3 (107 mg, 0.33 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and this system was heated to 80° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 30 mg of a title compound, a yield being 50%.
The compound 22a (30 mg, 0.05 mmol) was dissolved in dichloromethane (2 mL), then trifluoroacetic acid (1 mL) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 15 mg of a title compound, a yield being 61%.
LC-MS (ESI) m/z (M+H)+: 472.2
1H NMR (400 MHz, DMSO-d6) δ 7.85-7.78 (m, 1H), 7.59 (s, 1H), 7.44-7.36 (m, 1H), 7.22-7.11 (m, 3H), 6.99-6.93 (m, 1H), 3.85 (s, 3H), 3.72-3.56 (m, 8H), 1.85-1.67 (m, 4H).
A compound 5g (1.5 g, 6.45 mmol) was dissolved in 1,4-dioxane (20 mL) and water (3 mL), then 1-methylindazole-5-boric acid (1.1 g, 6.25 mmol) and K2CO3 (1.8 g, 13.02 mmol) were added in sequence, replacement with nitrogen was carried out, Pd(dppf)Cl2 (472 mg, 0.64 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react for 3 hours. The reaction system was cooled to the room temperature, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.1 g of a title compound, a yield being 60%.
The compound 23a (1.1 g, 3.88 mmol) was dissolved in 1,4-dioxane (30 mL) and water (3 mL), then 3-fluoro-4-cyanophenylboronic acid (1.9 g, 11.52 mmol) and K2CO3 (1.6 g, 11.58 mmol) were added in sequence, replacement with nitrogen was carried out, Pd(dppf)Cl2 (278 mg, 0.38 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react overnight. The reaction system was cooled to the room temperature, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.1 g of a title compound, a yield being 77%.
The compound 23b (500 mg, 1.36 mmol) was dissolved in acetonitrile (10 mL), then potassium iodide (451 mg, 2.72 mmol) and isoamyl nitrite (319 mg, 2.72 mmol) were added in sequence, and a reaction system was placed at 80° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 90 mg of a title compound, a yield being 14%.
The compound 23c (90 mg, 0.19 mmol) was dissolved in 1,4-dioxane (3 mL) and water (0.6 mL), then tert-butyl (4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolane-2-yl)cyclohexyl-3-alken-1-yl)carbamate (184 mg, 0.57 mmol) and K2CO3 (79 mg, 0.57 mmol) were added in sequence, replacement with nitrogen was carried out, Pd(dppf)Cl2 (14 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was placed at 80° C. and stirred to react for 2 hours. The reaction system was cooled to the room temperature, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 80 mg of a title compound, a yield being 78%.
The compound 23d (80 mg, 0.14 mmol) was dissolved in ethyl acetate (3 mL), then Pd/C (8 mg, 10%) was added at the room temperature, air in a system was replaced with hydrogen, and then the system was stirred at the room temperature under a hydrogen atmosphere to react overnight. A reaction system was filtered, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 67 mg of a title compound, a yield being 83%.
The compound 23e (67 mg, 0.12 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (3 mL, 3 M in EA) was added, and a reaction system was placed at the room temperature and stirred to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate (DCM/MeOH=10:1) to obtain 10 mg of a title compound 23-1 (upper point: Rf=0.3), a yield being 18%, and 10 mg of a title compound 23-2 (lower point: Rf=0.25), a yield being 18%.
LC-MS (ESI) m/z (M+H)+: 451.2
1H NMR (400 MHz, DMSO-d6) δ 8.13-8.08 (m, 2H), 7.83-7.80 (m, 1H), 7.79-7.73 (m, 1H), 7.69-7.64 (m, 1H), 7.49-7.44 (m, 1H), 7.26-7.22 (m, 1H), 7.21-7.16 (m, 1H), 4.07 (s, 3H), 3.24-3.18 (m, 1H), 3.03-2.92 (m, 1H), 2.15-2.02 (m, 2H), 1.80-1.60 (m, 4H), 1.25-1.10 (m, 2H).
LC-MS (ESI) m/z (M+H)+: 451.2
1H NMR (400 MHz, DMSO-d6) δ 8.12-8.10 (m, 1H), 8.03 (s, 1H), 7.82-7.80 (m, 1H), 7.79-7.73 (m, 1H), 7.68-7.64 (m, 1H), 7.46-7.41 (m, 1H), 7.26-7.22 (m, 1H), 7.20-7.16 (m, 1H), 4.07 (s, 3H), 2.88-2.72 (m, 2H), 2.03-1.91 (m, 4H), 1.75-1.62 (m, 2H), 1.36-1.25 (m, 2H).
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then 1-tert-butoxycarbonyl-1,4-diaza-5-cycloheptanone (47 mg, 0.22 mmol) and Cs2CO3 (107 mg, 0.33 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and this system was heated to 80° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 40 mg of a title compound, a yield being 68%.
The compound 24a (40 mg, 0.07 mmol) was dissolved in dichloromethane (2 mL), then trifluoroacetic acid (1 mL) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 16 mg of a title compound, a yield being 49%.
LC-MS (ESI) m/z (M+H)+: 460.2
1H NMR (400 MHz, DMSO-d6) δ 8.29 (s, 1H), 7.89-7.82 (m, 1H), 7.49 (dd, J=10.4, 1.2 Hz, 1H), 7.31 (dd, J=12.0, 2.0 Hz, 1H), 7.25-7.18 (m, 2H), 7.11-7.05 (m, 1H), 4.29-4.19 (m, 2H), 3.85 (s, 3H), 3.03-2.94 (m, 4H), 2.91-2.84 (m, 2H).
A compound 16b (75 mg, 0.15 mmol), (2,2-difluorobenzo[1,3-d]dioxol-5-yl)boric acid (46 mg, 0.23 mmol) and K2CO3 (62 mg, 0.45 mmol) were dissolved in 1,4-dioxane (1.5 mL) and water (0.3 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 53 mg of a title compound, a yield being 61%.
The compound 25a (53 mg, 0.09 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 34 mg of a title compound, a yield being 78%.
LC-MS (ESI) m/z (M+H)+: 478.2
1H NMR (400 MHz, DMSO-d6) δ 7.80 (dd, J=8.0, 6.8 Hz, 1H), 7.57 (s, 1H), 7.44 (d, J=1.6 Hz, 1H), 7.43-7.38 (m, 2H), 7.15 (dd, J=8.0, 1.2 Hz, 1H), 7.04 (dd, J=8.0, 1.2 Hz, 1H), 4.36-4.26 (m, 2H), 3.10-3.00 (m, 2H), 2.90-2.80 (m, 1H), 1.83-1.73 (m, 2H), 1.26-1.14 (m, 2H).
A compound 11b (550 mg, 2.88 mmol), tert-butyl 1,4-diazacycloheptane-1-formate (572 mg, 2.86 mmol), and K2CO3 (792 mg, 5.73 mmol) were dissolved in acetonitrile (7 mL), and this system was heated to 80° C. and stirred to react overnight. A reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 620 mg of a title compound, a yield being 61%.
The compound 26a (620 mg, 1.75 mmol), 3-fluoro-4-cyanophenylboronic acid (433 mg, 2.63 mmol) and K2CO3 (723 mg, 5.23 mmol) were dissolved in 1,4-dioxane (7 mL) and water (0.7 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (64 mg, 0.09 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 754 mg of a title compound, a yield being 98%.
The compound 26b (754 mg, 1.72 mmol) was dissolved in N,N-dimethylformamide (9 mL), NBS (609 mg, 3.42 mmol) was slowly added into a system under an ice bath, and a reaction system was stirred at the room temperature to react for 2 hours. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 315 mg of a title compound, a yield being 35%.
The compound 26c (160 mg, 0.31 mmol), 1-methylindazole-5-boric acid (82 mg, 0.47 mmol) and K2CO3 (128 mg, 0.93 mmol) were dissolved in 1,4-dioxane (3 mL) and water (0.3 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (12 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a silica gel column to obtain 145 mg of a title compound, a yield being 82%.
The compound 26d (72 mg, 0.13 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 2 hours. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 49 mg of a title compound, a yield being 83%.
LC-MS (ESI) m/z (M+H)+: 470.2
1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1H), 8.02-7.98 (m, 1H), 7.79-7.74 (m, 1H), 7.73-7.69 (m, 1H), 7.68-7.64 (m, 1H), 7.58-7.53 (m, 1H), 7.51-7.41 (m, 2H), 7.26-7.21 (m, 1H), 7.14-7.08 (m, 1H), 4.04 (s, 3H), 3.76-3.66 (m, 2H), 3.56-3.49 (m, 2H), 2.98-2.91 (m, 2H), 2.77-2.70 (m, 2H), 1.91-1.78 (m, 2H).
A compound 11a (500 mg, 2.60 mmol) was dissolved in tetrahydrofuran (5 mL), and then dimethylamine hydrochloride (636 mg, 7.80 mmol), 2-(7-azabenzotriazole)-N,N,N′,N′-tetramethylurea hexafluorophosphate (1.2 g, 3.16 mmol) and N,N-diisopropylethylamine (1.0 g, 7.74 mmol) were added and stirred at the room temperature to react overnight. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and 570 mg of a title compound was obtained and was directly used for the next step of reaction without purification.
The compound 27a (570 mg, 2.60 mmol), tert-butyl 1,4-diazacycloheptane-1-formate (518 mg, 2.59 mmol), and K2CO3 (708 mg, 5.12 mmol) were dissolved in acetonitrile (7 mL), and this system was heated to 80° C. and stirred to react overnight. A reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 653 mg of a title compound, a total yield in the two steps being 65%.
The compound 27b (520 mg, 1.36 mmol), 3-fluoro-4-cyanophenylboronic acid (335 mg, 2.03 mmol) and K2CO3 (559 mg, 4.04 mmol) were dissolved in 1,4-dioxane (7 mL) and water (0.7 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (50 mg, 0.07 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 399 mg of a title compound, a yield being 63%.
The compound 27c (389 mg, 0.83 mmol) was dissolved in N,N-dimethylformamide (4 mL), NBS (295 mg, 1.66 mmol) was slowly added into a system under an ice bath condition, and a reaction system was stirred at the room temperature to react for 2 hours. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 311 mg of a title compound, a yield being 68%.
The compound 27d (160 mg, 0.29 mmol), 1-methylindazole-5-boric acid (76 mg, 0.43 mmol) and K2CO3 (120 mg, 0.87 mmol) were dissolved in 1,4-dioxane (3 mL) and water (0.3 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (11 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a silica gel column to obtain 53 mg of a title compound, a yield being 30%.
The compound 27e (53 mg, 0.09 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 3 hours. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 31 mg of a title compound, a yield being 70%.
LC-MS (ESI) m/z (M+H)+: 498.3
1H NMR (400 MHz, DMSO-d6) δ 8.01 (s, 1H), 7.79-7.73 (m, 1H), 7.65 (s, 1H), 7.57-7.52 (m, 2H), 7.49-7.43 (m, 1H), 7.26-7.21 (m, 1H), 7.12-7.06 (m, 1H), 4.03 (s, 3H), 3.73-3.40 (m, 4H), 3.00 (s, 3H), 2.95 (s, 3H), 2.94-2.89 (m, 2H), 2.79-2.71 (m, 2H), 1.90-1.74 (m, 2H).
A compound 11a (500 mg, 2.60 mmol) was dissolved in chloroform (10 mL), replacement with nitrogen was carried out, thionyl chloride (3.1 g, 26.00 mmol) was slowly added into a system under an ice bath, replacement with nitrogen was carried out again, and a system was heated to 70° C. and stirred to react for 2 hours. A reaction system was subjected to vacuum concentration and dissolved by dichloromethane (1 mL), then was added dropwise into an ice-bath-cooled solution of cyclopropanamine (327 mg, 5.73 mmol) in dichloromethane (1 mL), and was stirred to react for 1 hour. Cold water was added into the reaction system to precipitate a solid, and after filtering, 600 mg of a title compound was obtained and directly used for the next step of reaction without purification.
The compound 28a (600 mg, 2.60 mmol), tert-butyl 1,4-diazacycloheptane-1-formate (520 mg, 2.60 mmol), and K2CO3 (718 mg, 5.20 mmol) were dissolved in acetonitrile (8 mL), and this system was heated to 80° C. and stirred to react overnight. A reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 913 mg of a title compound, a total yield in the two steps being 89%.
The compound 28b (520 mg, 1.32 mmol), 3-fluoro-4-cyanophenylboronic acid (327 mg, 1.98 mmol) and K2CO3 (547 mg, 3.96 mmol) were dissolved in 1,4-dioxane (5 mL) and water (0.5 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (48 mg, 0.07 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 630 mg of a title compound, a yield being 100%.
The compound 28c (630 mg, 1.31 mmol) was dissolved in N,N-dimethylformamide (8 mL), NBS (480 mg, 2.70 mmol) was slowly added into a system under an ice bath condition, and a reaction system was stirred at the room temperature to react for 2 hours. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 315 mg of a title compound, a yield being 43%.
The compound 28d (160 mg, 0.29 mmol), 1-methylindazole-5-boric acid (76 mg, 0.43 mmol) and K2CO3 (120 mg, 0.87 mmol) were dissolved in 1,4-dioxane (3 mL) and water (0.3 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (11 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a silica gel column to obtain 52 mg of a title compound, a yield being 30%.
The compound 28e (52 mg, 0.09 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 5 hours. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 20 mg of a title compound, a yield being 46%.
LC-MS (ESI) m/z (M+H)+: 510.3
1H NMR (400 MHz, DMSO-d6) δ 8.60-8.57 (m, 1H), 8.01 (s, 1H), 7.79-7.73 (m, 1H), 7.66 (s, 1H), 7.60 (s, 1H), 7.57-7.52 (m, 1H), 7.47-7.41 (m, 1H), 7.25-7.20 (m, 1H), 7.11-7.06 (m, 1H), 4.01 (s, 3H), 3.71-3.64 (m, 2H), 3.54-3.47 (m, 2H), 2.96-2.88 (m, 2H), 2.84-2.76 (m, 1H), 2.75-2.68 (m, 2H), 1.89-1.77 (m, 2H), 0.72-0.65 (m, 2H), 0.54-0.48 (m, 2H).
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then tert-butyl 2,8-diazaspiro[4.5]decane-2-carboxylate (53 mg, 0.22 mmol) and Cs2CO3 (107 mg, 0.33 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and this system was heated to 80° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 32 mg of a title compound, a yield being 52%.
The compound 29a (32 mg, 0.05 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (2 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 16 mg of a title compound, a yield being 60%.
LC-MS (ESI) m/z (M+H)+: 486.3
1H NMR (400 MHz, DMSO-d6) δ 7.85-7.77 (m, 1H), 7.57 (s, 1H), 7.46-7.36 (m, 1H), 7.24-7.11 (m, 3H), 7.01-6.92 (m, 1H), 3.85 (s, 3H), 3.81-3.57 (m, 4H), 3.50-3.24 (m, 3H), 2.96-2.88 (m, 1H), 1.81-1.72 (m, 1H), 1.65-1.58 (m, 1H), 1.58-1.48 (m, 4H).
A compound 30a (3.0 g, 14.07 mmol) was dissolved in tetrahydrofuran (50 mL), replacement with nitrogen was carried out, the temperature was reduced to −78° C., lithium bis(trimethylsilyl)amide (19 mL, 1 M in THF) was added dropwise, this system reacted at −78° C. for 1 hour, then N-phenylbis(trifluoromethanesulfon)imide (6.5 g, 18.19 mmol) was added to continue the reaction for 0.5 hour, the system restored to the room temperature to react for 14 hours, then water was added for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 2.0 g of a title compound, a yield being 41%.
The compound 30b (500 mg, 1.45 mmol), bis(pinacolato)diboron (1.1 g, 4.33 mmol) and potassium acetate (426 mg, 4.34 mmol) were dissolved in 1,4-dioxane (5 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (106 mg, 0.15 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 90° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 300 mg of a title compound, a yield being 64%.
A compound 5j (50 mg, 0.11 mmol), the compound 30c (177 mg, 0.55 mmol) and K2CO3 (45 mg, 0.33 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (7 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 80° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 20 mg of a title compound, a yield being 35%.
The compound 30d (20 mg, 0.04 mmol) was dissolved in ethyl acetate (2 mL), then Pd/C (2 mg, 10%) was added at the room temperature, air in a system was replaced with hydrogen, and then the system was stirred at the room temperature under a hydrogen atmosphere to react overnight. A reaction system was filtered, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 10 mg of a title compound, a yield being 50%.
The compound 30e (10 mg, 0.02 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (2 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 6 mg of a title compound, a yield being 74%.
LC-MS (ESI) m/z (M+H)+: 445.2
1H NMR (400 MHz, DMSO-d6) δ 8.03-7.98 (m, 1H), 7.88-7.82 (m, 1H), 7.46-7.41 (m, 1H), 7.33-7.28 (m, 1H), 7.24-7.16 (m, 2H), 7.10-7.04 (m, 1H), 3.86 (s, 3H), 3.66-3.45 (m, 1H), 3.22-3.10 (m, 1H), 3.08-2.88 (m, 2H), 2.87-2.74 (m, 1H), 2.05-1.58 (m, 6H).
A compound 31a (10.0 g, 75.18 mmol) was dissolved in pyridine (200 mL), then tosyl chloride (28.0 g, 146.87 mmol) was added, and this system was heated to 100° C. and stirred to react for 16 hours. A reaction system was cooled to the room temperature and then poured into water, a solid was precipitated, and filtering and vacuum drying were carried out to obtain 18.0 g of a title compound, a yield being 65%.
The compound 31b (5.8 g, 15.74 mmol) was dissolved in acetonitrile (100 mL), potassium hydroxide (2.2 g, 39.21 mmol) was added, a reaction system was heated to 80° C. to react for 0.5 hour, the system was cooled to the room temperature, then 1,3-dibromo-2-propanol (4.1 g, 18.82 mmol) was added, and the reaction system was heated to 80° C. again to react for 16 hours. The reaction system was cooled to the room temperature and then subjected to vacuum concentration, then water was added for dilution, extraction was carried out 3 times with dichloromethane, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 5.3 g of a title compound, a yield being 79%.
The compound 31c (2.0 g, 4.71 mmol) was dissolved in a hydrogen bromide aqueous solution (20 mL, 40%), and a reaction system was heated to 100° C. to react for 22 hours. The reaction system was cooled to the room temperature and then subjected to vacuum concentration, and then beating was carried out using methyl tert-butyl ester to obtain 600 mg of a crude product, a yield being 46%.
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then the compound 31d (214 mg, 0.77 mmol) and Cs2CO3 (358 mg, 1.10 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and this system was heated to 80° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 5 mg of a title compound, a yield being 10%.
LC-MS (ESI) m/z (M+H)+: 462.2
A compound 31c (2.5 g, 5.89 mmol) was dissolved in dichloromethane (30 mL) replacement with nitrogen was carried out, a system was cooled to 0° C., then (diethylamino)sulfur trifluoride (1.9 g, 11.79 mmol) was added, and a reaction system was heated to the room temperature to react for 16 hours. Water was then added to quench the reaction, extraction was carried out 3 times with dichloromethane, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.6 g of a title compound, a yield being 64%.
The compound 32a (1.6 g, 3.75 mmol) was dissolved in a hydrogen bromide aqueous solution (20 mL, 40%), and a reaction system was heated to 120° C. to react for 22 hours. The reaction system was cooled to the room temperature and then subjected to vacuum concentration, and then beating was carried out using methyl tert-butyl ester to obtain 700 mg of a crude product, a yield being 67%.
A compound 5j (50 mg, 0.11 mmol) was dissolved in 1,4-dioxane (3 mL), then the compound 32b (216 mg, 0.77 mmol) and Cs2CO3 (358 mg, 1.10 mmol) were added in sequence, replacement with nitrogen was carried out, RuPhos-Pd-G3 (9 mg, 0.01 mmol) was added, replacement with nitrogen was carried out again, and this system was heated to 80° C. and stirred to react for 3 hours. A reaction system was cooled to the room temperature, a reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain title compounds 32-1 (3 mg, yield: 6%) and 32-2 (4 mg, yield: 8%).
32-1: LC-MS (ESI) m/z (M+H)+: 464.2
1H NMR (400 MHz, DMSO-d6) δ 7.85-7.78 (m, 1H), 7.56 (s, 1H), 7.43-7.38 (m, 1H), 7.24-7.12 (m, 3H), 7.00-6.94 (m, 1H), 4.53-4.44 (m, 1H), 4.41-4.28 (m, 2H), 4.26-4.18 (m, 1H), 3.85 (s, 3H), 3.03-2.88 (m, 2H), 2.78-2.65 (m, 2H), 2.03-1.95 (m, 1H).
32-2: LC-MS (ESI) m/z (M+H)+: 464.2
1H NMR (400 MHz, DMSO-d6) δ 7.85-7.79 (m, 1H), 7.47 (s, 1H), 7.40 (dd, J=10.8, 1.2 Hz, 1H), 7.23-7.12 (m, 3H), 6.99-6.93 (m, 1H), 4.20-3.91 (m, 2H), 3.85 (s, 3H), 3.80-3.69 (m, 2H), 3.00-2.75 (m, 4H), 2.03-1.94 (m, 1H).
A compound 33a (2.0 g, 10.47 mmol) was dissolved in dichloromethane (20 mL), triethylamine (1.6 g, 15.81 mmol) was slowly added into a system under an ice bath, then acetyl chloride (1.2 g, 15.29 mmol) was slowly added, and a reaction system restored to the room temperature and was stirred to react for 1 hour. A reaction liquid was poured into water, extraction was carried out 3 times with dichloromethane, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 2.4 g of a title compound, a yield being 98%.
The compound 33b (2.4 g, 10.30 mmol) was dissolved in a boron trifluoride-acetic acid solution (26 mL, 33% BF3), replacement with nitrogen was carried out, and a reaction system was heated to 150° C. and stirred to react for 8 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water to precipitate a solid, and after filtering, 1.6 g of a title compound crude product was obtained and directly used for the next step without purification.
The compound 33c (1.6 g, 6.87 mmol) was dissolved in methanol (16 mL), then hydroxylamine hydrochloride (963 mg, 13.86 mmol) and sodium acetate (853 mg, 10.40 mmol) were added, and a reaction system was heated to 60° C. and stirred to react for 1 hour. The reaction system was cooled to the room temperature, a reaction liquid was poured into cold water to precipitate a solid, filtering was carried out, and a filter cake was dried to obtain a brown solid. The obtained solid was dissolved in acetic anhydride (6 mL), a reaction system was heated to 60° C. and stirred to react for 2 hours, the reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, and a filtrate was subjected to vacuum concentration to obtain a crude product. The obtained crude product was dissolved in N,N-dimethylformamide (10 mL), then K2CO3 (1.9 g, 13.75 mmol) was added, and a reaction system was stirred at the room temperature to react overnight. A reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 693 mg of a title compound, a total yield in two steps being 29%.
The compound 33d (693 mg, 3.01 mmol), bis(pinacolato)diboron (914 mg, 3.60 mmol) and potassium acetate (588 mg, 5.99 mmol) were dissolved in 1,4-dioxane (7 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (66 mg, 0.09 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 757 mg of a title compound, a yield being 91%.
A compound 16b (150 mg, 0.30 mmol), the compound 33e (166 mg, 0.60 mmol) and K2CO3 (124 mg, 0.90 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 41 mg of a title compound, a yield being 24%.
The compound 33f (41 mg, 0.07 mmol) was dissolved in ethyl acetate (1 mL), then an ethyl hydrochloride solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 12 mg of a title compound, which was a yellow solid, a yield being 35%.
LC-MS (ESI) m/z (M+H)+: 471.2
1H NMR (400 MHz, DMSO-d6) δ 7.90-7.87 (m, 1H), 7.82-7.74 (m, 2H), 7.66 (s, 1H), 7.47-7.42 (m, 1H), 7.16 (dd, J=8.0, 1.6 Hz, 1H), 4.42-4.26 (m, 2H), 3.14-3.03 (m, 2H), 2.92-2.82 (m, 1H), 2.55 (s, 3H), 1.84-1.74 (m, 2H), 1.26-1.16 (m, 2H).
A compound 34a (2.0 g, 8.85 mmol) was dissolved in dichloromethane (30 mL), replacement with nitrogen was carried out, a system was cooled to 0° C., then (diethylamino)sulfur trifluoride (4.3 g, 26.68 mmol) was added, and a reaction system was placed at the room temperature to react for 16 hours. Water was then added into the system to quench the reaction, extraction was carried out 3 times with dichloromethane, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 1.8 g of a title compound, a yield being 82%.
The compound 34b (1.0 g, 4.03 mmol) was dissolved in acetonitrile (15 mL), then iodomethane (1.1 g, 7.75 mmol) and Cs2CO3 (2.0 g, 6.14 mmol) were added, and a reaction system was placed at the room temperature and stirred to react for 16 hours. A reaction liquid was subjected to vacuum concentration, water was added into the system for dilution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 900 mg of a title compound, a yield being 85%.
The compound 34c (500 mg, 1.91 mmol), bis(pinacolato)diboron (1.5 g, 5.91 mmol) and potassium acetate (562 mg, 5.73 mmol) were dissolved in 1,4-dioxane (5 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (139 mg, 0.19 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 90° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 420 mg of a title compound, a yield being 71%.
A compound 16b (80 mg, 0.16 mmol), the compound 34d (247 mg, 0.80 mmol) and K2CO3 (66 mg, 0.48 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 80° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 30 mg of a title compound, a yield being 31%.
The compound 34e (30 mg, 0.05 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (2 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 22 mg of a title compound, a yield being 88%.
LC-MS (ESI) m/z (M+H)+: 503.2
1H NMR (400 MHz, DMSO-d6) δ 7.85-7.78 (m, 1H), 7.63-7.54 (m, 2H), 7.51-7.45 (m, 1H), 7.44-7.38 (m, 1H), 7.26-7.20 (m, 1H), 7.19-7.13 (m, 1H), 4.40-4.26 (m, 2H), 3.18 (s, 3H), 3.11-2.98 (m, 2H), 2.95-2.82 (m, 1H), 1.86-1.73 (m, 2H), 1.27-1.15 (m, 2H).
A compound 35a (1.3 g, 4.32 mmol) was dissolved in dimethylsulfoxide (20 mL), replacement with nitrogen was carried out, copper powder (707 mg, 11.13 mmol) was added and stirred at the room temperature for 1 hour, then 2-bromo-2,2-difluoroethyl acetate (2.3 g, 11.33 mmol) was added, and a reaction system was heated to 80° C. and stirred to react for 2 hours. The reaction system was cooled to the room temperature, filtering was carried out, a filtrate was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 719 mg of a title compound, a yield being 56%.
The compound 35b (719 mg, 2.42 mmol) was dissolved in dry tetrahydrofuran (20 mL), then a methyl magnesium bromide reagent (4.1 mL, 3 M in THF) was slowly added dropwise under an ice bath, and after dropwise adding was finished, a reaction system slowly restored to the room temperature and was stirred to react overnight. The reaction system was cooled to 0° C., an appropriate amount of water was slowly added to quench the reaction, filtering was carried out, a filtrate was extracted 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 577 mg of a title compound, a yield being 84%.
The compound 35c (577 mg, 2.04 mmol), bis(pinacolato)diboron (1.0 g, 3.94 mmol) and potassium acetate (600 mg, 6.12 mmol) were dissolved in 1,4-dioxane (20 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (146 mg, 0.20 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react for 24 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 364 mg of a title compound, a yield being 54%.
A compound 16b (100 mg, 0.20 mmol), the compound 35d (132 mg, 0.40 mmol) and K2CO3 (83 mg, 0.60 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.2 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 100° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 57 mg of a title compound, a yield being 46%.
The compound 35e (57 mg, 0.09 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (1 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 37 mg of a title compound, which was a yellow solid, a yield being 77%.
LC-MS (ESI) m/z (M+H)+: 524.2
1H NMR (400 MHz, DMSO-d6) δ 7.84-7.78 (m, 1H), 7.64 (s, 1H), 7.50-7.43 (m, 1H), 7.39-7.28 (m, 3H), 7.24-7.19 (m, 1H), 5.49 (s, 1H), 4.40-4.28 (m, 2H), 3.14-3.03 (m, 2H), 2.91-2.82 (m, 1H), 1.85-1.75 (m, 2H), 1.28-1.18 (m, 2H), 1.15 (s, 6H).
A compound 9a (500 mg, 2.33 mmol) was dissolved in acetonitrile (7.5 mL), glacial acetic acid (0.75 mL) and 1-chloromethyl-4-fluoro-1,4-diazabicyclo[2.2.2]octane bis(tetrafluoroborate) (1.6 g, 4.66 mmol) were added in sequence, and a reaction system was heated to 80° C. and stirred to react for 5 hours. The reaction system was cooled to the room temperature, water was added into the system to quench the reaction, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 300 mg of a title compound, a yield being 55%.
The compound 36a (300 mg, 1.29 mmol) was dissolved in acetonitrile (10 mL), then Cs2CO3 (630 mg, 1.94 mmol) and 1,1-dimethyl ethylene oxide (185 mg, 2.57 mmol) were added in sequence, and a reaction system was heated to 80° C. and stirred to react for 16 hours. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 150 mg of a title compound, a yield being 38%.
The compound 36b (150 mg, 0.49 mmol), bis(pinacolato)diboron (373 mg, 1.47 mmol) and potassium acetate (144 mg, 1.47 mmol) were dissolved in 1,4-dioxane (5 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (37 mg, 0.05 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 90° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using silica gel column chromatography to obtain 76 mg of a title compound, a yield being 44%.
A compound 16b (107 mg, 0.21 mmol), the compound 36c (74 mg, 0.21 mmol) and K2CO3 (87 mg, 0.63 mmol) were dissolved in 1,4-dioxane (2 mL) and water (0.4 mL), replacement with nitrogen was carried out, Pd(dppf)Cl2 (15 mg, 0.02 mmol) was added, replacement with nitrogen was carried out again, and a reaction system was heated to 80° C. and stirred to react overnight. The reaction system was cooled to the room temperature, a reaction liquid was poured into water, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 35 mg of a title compound, a yield being 26%.
The compound 36d (35 mg, 0.05 mmol) was dissolved in ethyl acetate (1 mL), then a hydrochloric acid-ethyl acetate solution (2 mL, 3 M in EA) was added, and a reaction system was stirred at the room temperature to react for 1 hour. The reaction system was subjected to vacuum concentration and then diluted by adding water, a pH value of the system was adjusted to about 8 using a saturated sodium bicarbonate solution, extraction was carried out 3 times with ethyl acetate, an organic phase was combined and then was washed with a saturated saline solution, drying was carried out with anhydrous sodium sulfate, filtering was carried out, a filtrate was subjected to vacuum concentration, and a crude product was purified using a thin layer silica gel plate to obtain 23 mg of a title compound, a yield being 78%.
LC-MS (ESI) m/z (M+H)+: 546.3
1H NMR (400 MHz, DMSO-d6) δ 7.86-7.81 (m, 1H), 7.80-7.74 (m, 1H), 7.62 (s, 1H), 7.60-7.54 (m, 1H), 7.44-7.38 (m, 1H), 7.19 (dd, J=8.0, 1.2 Hz, 1H), 4.67 (brs, 1H), 4.40-4.28 (m, 2H), 4.20-4.07 (m, 2H), 3.13-3.01 (m, 2H), 2.94-2.82 (m, 1H), 1.86-1.75 (m, 2H), 1.30-1.18 (m, 2H), 1.14 (s, 3H), 1.10 (s, 3H).
Compounds 1-36 used in following test examples are prepared by synthesis methods of Examples 1-36 respectively.
In this test, a homogeneous time resolved fluorescence (HTRF) technology was utilized, and the inhibitory activity of compounds on LSD1 was detected by adopting a two-step method. The first step was to incubate LSD1, compounds of different concentrations, and a biotin-labeled methylated polypeptide substrate (H3(1-21)K4 me1-biotin) at the room temperature for fixed time. The second step was to add an antibody (Anti-H3K4 me0-Eu(K)) to a demethylated polypeptide substrate and an XL-665-labeled streptomycin (SA-XL665) detection reagent, and continue to incubate at the room temperature for fixed time. A signal value Intensity (665 nM)/Intensity (615 nM) was read using EnVision to determine the content of the demethylated polypeptide substrate so as to evaluate the effects of the tested compounds on LSD1 activity, and meanwhile IC50 values of the tested compounds on the LSD1 were calculated using an inhibition rate.
Preparation of a 10 mM compound stock solution: compound powder was dissolved in 100% DMSO to prepare the 10 mM compound stock solution respectively.
An inhibition percentage was calculated using a maximum value, a minimum value, and a signal value (maximum value: fluorescence intensity of 5 μL of 2× enzyme solution+5 μL of 2× substrate mixed solution+10 nL of 100% DMSO+10 μL of detection solution; minimum value: fluorescence intensity of 5 μL of 1× reaction solution+5 μL of 2× substrate mixed solution+10 nL of 100% DMSO+10 μL of detection solution; and signal value: fluorescence intensity of 5 μL of 2× enzyme solution+5 μL of 2× substrate mixed solution+10 nL of diluted compound to be tested+10 μL of detection solution), and a calculation formula was as follows:
Using a logarithm of the concentration as an X axis and the percentage inhibition rate as a Y axis, a dose-response curve was fitted using a formula log(inhibitor) vs. response-Variable slope (Four parameters) of analysis software Graphpad Prism 5 to obtain the IC50 value of each compound on the enzyme activity.
See Table 1 for inhibitory activities of the tested compounds in the present application and positive controls on the LSD1.
It can be known from the test data of the inhibitory activity of each compound on the LSD1 shown in Table 1 that, the compounds of the present application may have obvious LSD1 inhibitory activity.
Research had confirmed that LSD1 played a crucial role in the occurrence process of various types of cancers, including small cell lung cancer (SCLC) and acute myeloid leukemia (AML). Therefore, a human acute myeloid leukemia (Kasumi-1) cell line was used to determine the ATP content in cells at different drug concentrations using a Celltiter GLO assay kit, the cell activity was reflected by luminescence intensity, and IC50 values of different compounds on Kasumi-1 cells were calculated using inhibition rates. The inhibitory effects of the compounds in the present application on the proliferation of Kasumi-1 cells were studied to evaluate the anti-tumor efficacy of the tested compounds at the cellular level.
An inhibition rate was calculated by utilizing luminescence intensity of the above DMSO group (with DMSO added), test group (with the tested compound or a positive control solution added) and blank group (not treating cells), and a calculation formula was as follows:
Using a logarithm of the concentration as an X axis and the percentage inhibition rate as a Y axis, a dose-response curve was fitted using a formula log(inhibitor) vs. response-Variable slope (Four parameters) of analysis software Graphpad Prism 8 to obtain the IC50 value of each compound on Kasumi-1 proliferation inhibition activity.
See Table 2 for Kasumi-1 cell growth inhibition activities of the tested compounds in the present application and positive controls.
It can be known from the test data of the Kasumi-1 cell inhibition activity of each compound shown in Table 2 that, the compounds of the present application may have a high inhibition effect on Kasumi-1 cell growth.
Research had confirmed that LSD1 played a crucial role in the occurrence process of various types of cancers, including small cell lung cancer (SCLC) and acute myeloid leukemia (AML). Therefore, a human small cell lung cancer (NCI-H1417) cell line was used to determine the ATP content in cells at different drug concentrations using a Celltiter GLO assay kit, and the cell activity was reflected by luminescence intensity. IC50 values of different compounds on the NCI-H1417 cells were calculated using inhibition rates. The inhibitory effects of the compounds in the present application on the proliferation of the NCI-H1417 cells were studied to evaluate the anti-tumor efficacy of the tested compounds at the cellular level.
An inhibition percentage was calculated by utilizing luminescence intensity of the above DMSO group (with DMSO added), test group (with the tested compound and a positive control solution added) and blank group (not treating cells), a calculation formula being
Using a logarithm of the concentration as an X axis and the percentage inhibition rate as a Y axis, a dose-response curve was fitted using a formula log(inhibitor) vs. response-Variable slope (Four parameters) of analysis software Graphpad Prism 8 to obtain the IC50 value of each compound on NCI-H1417 proliferation inhibition activity.
See Table 3 for NCI-H1417 cell growth inhibition activities of the tested compounds in the present application and positive controls.
It can be known from the test data of the NCI-H1417 cell inhibition activity of each compound shown in Table 3 that, the compounds of the present application may have a high inhibition effect on NCI-H1417 cell growth.
With SD rats as tested animals, blood drug concentrations in plasma at different moments after oral administration of the compounds of the present application by the rats in a clarification kit mode were determined using an LC-MS/MS method. Pharmacokinetic parameters of the compounds of the present application in the rats were obtained, and pharmacokinetic characteristics of the compounds were studied.
3.1. Grouping: Random Grouping was Carried Out According to Table 4, and after Grouping, there was No Statistical Difference in Weight of the SD Rats Between Groups.
According to Table 4, each group of rats was given the corresponding tested drugs or positive drugs by gavage, before administration, and 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, and 24 h after administration, a fixed volume of blood was collected through eye sockets and placed in an EDTA-K2 anticoagulant tube, centrifuging was carried out at 8000 rpm for 1 min to separate plasma into a centrifuge tube, and the plasma was frozen in a −20° C. refrigerator.
The plasma at each time point stored at −20° C. was taken out, then acetonitrile was added, after whirling at 1500 revolutions for 2 min, centrifuging was carried out for 15 min (3500 r/min), and a fixed volume of solution supernatant was taken for LC-MS/MS analysis.
Non-compartment model fitting was carried out on pharmacokinetic behaviors of the tested compounds, and DAS3.31 software was adopted to calculate main pharmacokinetic parameters (T1/2, Tmax, Cmax, AUClast, etc.).
It can be seen from the test results in Table 5 that compared to the positive group (CC-90011), the compound 1, compound 4, compound 14-16, compound 18, compound 20-21, and compound 36 administration groups are superior to CC-90011 in terms of plasma exposure and the like, indicating that the pharmacokinetic properties of the compounds prepared in Examples 1, 4, 14, 15, 16, 18, 20, 21 and 36 of the present application are significantly improved compared to CC-90011. Therefore, the compounds of the present application can exhibit good pharmacokinetic properties.
With SD rats as tested animals, blood drug concentrations of the compounds of the present application in plasma at different moments after oral administration and intravenous administration to the rats were determined using an LC-MS/MS method, pharmacokinetic parameters and bioavailabilities of the compounds of the present application in the rats were obtained, and pharmacokinetic characteristics of the compounds were studied.
3.1. Grouping: Random Grouping was Carried Out According to Table 6, and after Grouping, there was No Statistical Difference in Weight of the SD Rats Between Groups.
According to Table 6, each group of rats was given the corresponding tested drugs or positive drugs by gavage, before administration, and 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, 10 h and 24 h after administration, a fixed volume of blood was collected through eye sockets and placed in an EDTA-K2 anticoagulant tube, centrifuging was carried out at 8000 rpm for 1 min to separate plasma into a centrifuge tube, and the plasma was frozen in a −20° C. refrigerator.
The plasma at each time point stored at −20° C. was taken out, then acetonitrile was added, after whirling at 1500 revolutions for 2 min, centrifuging was carried out for 15 min (3500 r/min), and a fixed volume of solution supernatant was taken for LC-MS/MS analysis.
Non-compartment model fitting was carried out on pharmacokinetic behaviors of the tested compounds, and DAS3.31 software was adopted to calculate main pharmacokinetic parameters (T1/2, Tmax, Cmax, AUClast, etc.).
It can be seen from the test results in Table 7 that, under the same dosage, the compound 16 is superior to CC-90011 in terms of plasma exposure, maximum blood drug concentration, bioavailability and the like of oral administration and intravenous administration, indicating that pharmacokinetic properties of the compound prepared in Example 16 of the present application are obviously improved compared to CC-90011.
With Balb/c mice as tested animals, blood drug concentrations of the compounds of the present application in plasma at different moments after administration of the mice under different solvent conditions were determined using an LC-MS/MS method. Pharmacokinetic parameters of the compounds of the present application in the mice under the different solvent conditions were obtained, and pharmacokinetic characteristics of the compounds were studied.
3.1. Grouping: Random Grouping was Carried Out According to Table 8, and after Grouping, there was No Statistical Difference in Weight of the Balb/c Mice Between Groups.
According to Table 8, each group of mice was given the corresponding tested drugs or positive drugs by gavage, before administration, and 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, 10 h and 24 h after administration, a fixed volume of blood was collected through eye sockets and placed in an EDTA-K2 anticoagulant tube, centrifuging was carried out at 8000 rpm for 1 min to separate plasma into a centrifuge tube, and the plasma was frozen in a −20° C. refrigerator.
The plasma at each time point stored at −20° C. was taken out, then acetonitrile was added, after whirling at 1500 revolutions for 2 min, centrifuging was carried out for 15 min (3500 r/min), and a fixed volume of solution supernatant was taken for LC-MS/MS analysis.
Non-compartment model fitting was carried out on pharmacokinetic behaviors of the tested compounds, and DAS3.31 software was adopted to calculate main pharmacokinetic parameters (T1/2, Tmax, Cmax, AUClast, etc.).
It can be seen from the test results in Table 9 that, compared to the positive group (CC-90011), the compound 16 is superior to CC-90011 in terms of plasma exposure, maximum blood drug concentration and the like under different solvent conditions, indicating that pharmacokinetic properties of the compound prepared in Example 16 of the present application are obviously improved compared to CC-90011, and the compound has pharmacokinetic advantages under the different solvent conditions.
A liver microsome in-vitro warm incubation method used liver microsomes, supplemented by an NADPH regeneration system, to simulate physiological environmental conditions in vitro for metabolic reaction, after a certain period of reaction, LC-MS/MS was used to determine prototype drugs and metabolites in a warm incubation solution, and the content was preliminarily analyzed. Studying the liver microsome stability of drugs in different species was crucial for understanding a change process of the drugs in vivo.
According to the requirements of the instructions of a Phase I Metabolic Stability Kit, a liver microsome incubation system was prepared, and according to a sequence, a mixed system of a PBS, liver microsomes, an NADPH regeneration system solution A and an NADPH regeneration system solution B, as well as substances to be tested were added in sequence respectively, and were incubated at 37° C. Each sample was parallelized 3 times, with a sample without an NADPH generation system as a negative control, after 0, 5, 15, 30, 45, and 60 min, equal volumes of pre-cooled acetonitrile were added to terminate the reaction respectively, and the prototype drug content in a warm incubation solution was determined by LC-MS/MS.
The above experiment results indicate that the compounds 16 and 36 have high metabolic stability in liver microsomes of humans, rats, mice, dogs and monkeys. Therefore, the compounds of the present application can exhibit good druggability.
A rapidly-activated human delay rectifier outward potassium current was mainly mediated by an hERG ion channel and participated in human myocardial cell repolarization. Drug blocking of this current was the main cause of prolonged QT interval syndrome, even acute arrhythmia and sudden death in clinical practice. A whole cell sheet technology was used to detect the blocking effects of compounds on hERG channels on HEK293 cells stably expressing hERG channels, and a median inhibitory concentration IC50 of the compound was determined. As part of cardiac safety evaluation, its safety in-vitro screening for cardiac toxicity was preliminarily evaluated.
Cells were transferred into a perfusion trough, 137 mM NaCl, 4 mM KCl, 1.8 mM CaCl2, 1 mM MgCl2, 10 mM HEPES and 10 mM glucose were adopted to prepare an extracellular fluid, and NaOH was used to adjust to 7.4 (pH) for perfusion. Electrodes were drawn using PC-10 (Narishige, Japan). The whole cell patch clamp was used for recording, and noise was filtered using one fifth of a sampling frequency. The cells were clamped at −80 mV, then depolarized to 40 mV using square waves lasting for 4 seconds, and then hyperpolarized to −40 mV using square waves lasting for 2 seconds to obtain an hERG tail current. This procedure was repeated every 20 seconds. The hERG tail current was a pure hERG current. A maximum current triggered by the second square wave was detected, after the maximum current was stable, the compounds were tested through perfusion, and after reaction was stable, a blocking intensity was calculated.
The above experiment results indicate that compared to a positive phase (CC-90011), an hERG inhibition IC50 value of the compound 16 is greater than 10 μM, indicating that the compound of the present application has the lower cardiac toxicity risk than a positive control.
CB17 SCID mice, female, 6-8 weeks, weight about 18-22 grams, and each mouse was subcutaneously inoculated with 0.1 mL (1×107 cells+matrigel) of Kasumi-1 cells on the right side. When an average volume of tumors reached 100-200 cubic millimeters, group administration was performed, see the table below (Table 12) for administration dosages and methods. The tumor volume was measured twice a week, the weights of the mice as well as lengths and widths of tumors were recorded in each time of measurement, and the tumor volume=length×width×width/2 mm3. When the average tumor volume of a solvent group grew to 3000 cubic millimeters or above, administration was ended, each group of mice was killed and dissected to take out tumor lumps, the tumor weight of each mouse was weighed and the tumor volume was measured, and an average tumor volume difference between a tested compound group and the solvent group was obtained via comparison. The tumor suppression effects of the compounds were evaluated with TGI (%), which could reflect a tumor growth inhibition rate.
Calculation of TGI (%):TGI (%)=[1−(current average tumor volume of compounds−average tumor volume at the beginning of compounds)/(current average tumor volume of solvent group−average tumor volume at the beginning of solvent group)]×100%
The changes in tumor volume and tumor weight of the mice in each group were shown in
The above test results indicate that the compounds 16 and 36 of the present application exhibit good in-vivo efficacy in a human acute myeloid leukemia Kasumi-1 cell subcutaneous heterograft tumor model, with a significant tumor suppression effect (TGI>60%), the tested animals have good tolerance to the compounds 16 and 36, and after administration, the weight of the mice does not decrease (as shown in
Balb/C nu mice, female, 6-8 weeks, weight about 18-22 grams, and each mouse was subcutaneously inoculated with 0.1 mL (1×107 cells+matrigel) of HL-60 cells on the right side. When an average volume of tumors reached 100-200 cubic millimeters, group administration was performed, see the table below (Table 13) for administration dosages and methods. The tumor volume was measured twice a week, the weights of the mice as well as lengths and widths of tumors were recorded in each time of measurement, and the tumor volume=length×width×width/2 mm3. When the average tumor volume of a solvent group grew to 3000 cubic millimeters or above, administration was ended, and an average tumor volume difference between a tested compound group and the solvent group was obtained via comparison. The tumor suppression effects of the compounds were evaluated with TGI (%), which could reflect a tumor growth inhibition rate.
The changes in tumor volume of the mice in each group were shown in
The above test results indicate that, compared to the positive compound CC-90011, the compounds of the present application exhibit good in-vivo efficacy in a human myeloid leukemia HL-60 cell subcutaneous heterograft tumor model, with a significant tumor suppression effect (TGI>60%), the tested animals have good tolerance to the compounds of the present application, and after administration, the weight of the mice does not decrease (as shown in
It should be understood that the above detailed description and accompanying examples are only illustrative and should not be considered as limiting the scope of the present application, which is solely defined by the accompanying claims and their equivalents. Those skilled in the art can easily see various changes and modifications to the disclosed embodiments. Such changes and modifications may be made without departing from its spirit and scope, including but not limited to those related to the chemical structures, substituents, derivatives, intermediates, synthesis methods, formulations, and/or methods of use of the present application. All publications, patents, and patent applications referred to herein are hereby incorporated for various purposes.
Number | Date | Country | Kind |
---|---|---|---|
202210085981.9 | Jan 2022 | CN | national |
202211383830.8 | Nov 2022 | CN | national |
This application is the U.S. National Stage of PCT/CN2022/134330 filed on Nov. 25, 2022, which claims priority to Chinese Patent Application 202210085981.9 filed on Jan. 25, 2022, and Chinese Patent Application 202211383830.8 filed on Nov. 7, 2022, the entire content of both are incorporated herein by reference in their entirety.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/CN2022/134330 | 11/25/2022 | WO |