CN109651359B - Substituted nicotinamide compound, pharmaceutical composition and application thereof - Google Patents

Substituted nicotinamide compound, pharmaceutical composition and application thereof Download PDF

Info

Publication number
CN109651359B
CN109651359B CN201910096790.0A CN201910096790A CN109651359B CN 109651359 B CN109651359 B CN 109651359B CN 201910096790 A CN201910096790 A CN 201910096790A CN 109651359 B CN109651359 B CN 109651359B
Authority
CN
China
Prior art keywords
compound
deuterium
hydrogen
another embodiment
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201910096790.0A
Other languages
Chinese (zh)
Other versions
CN109651359A (en
Inventor
王义汉
赵九洋
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Publication of CN109651359A publication Critical patent/CN109651359A/en
Application granted granted Critical
Publication of CN109651359B publication Critical patent/CN109651359B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Abstract

The invention provides a substituted nicotinamide compound, a composition containing the compound and application of the compound, wherein the substituted nicotinamide compound is a compound shown as a formula (I), or pharmaceutically acceptable salt, a prodrug, a hydrate or a solvate, a crystal form, a stereoisomer or an isotopic variant thereof. The compounds of the invention inhibit the Abelson protein (ABL1), Abelson-related protein (ABL2) and related chimeric proteinsParticularly, the tyrosine kinase activity of Bcr-Abl1 has better pharmacokinetic property.

Description

Substituted nicotinamide compound, pharmaceutical composition and application thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a substituted nicotinamide compound, a composition containing the compound and application of the compound. More particularly, the present invention relates to certain deuterium substituted nicotinamide compounds that inhibit the tyrosine kinase activity of Abelson protein (ABL1), Abelson-related protein (ABL2) and related chimeric proteins, particularly Bcr-ABL 1. The invention also provides methods of making, pharmaceutical compositions containing and methods of using the compounds in the treatment of cancer, and the compounds of the invention have superior pharmacokinetic properties.
Background
The tyrosine kinase activity of the ABL1 protein is usually tightly regulated, where the N-terminal cap region of the SH3 domain plays an important role. One regulatory mechanism involves myristoylation of the N-terminal cap glycine-2-residue, followed by interaction with the myristate binding site in the SH1 catalytic domain. One hallmark of Chronic Myelogenous Leukemia (CML) is philadelphia chromosome (Ph), formed by the reciprocal translocation of the t (9, 22) chromosomes in hematopoietic stem cells. The chromosome carries the Bcr-Abl oncogene, which encodes a chimeric Bcr-Abl1 protein lacking the N-terminal cap and having a constitutively activated tyrosine kinase domain.
Although drugs that inhibit the tyrosine kinase activity of Bcr-Abl1 via an ATP-competitive mechanism (such as imatinib) are effective in treating CML, some patients relapse due to the emergence of drug-resistant clones, where mutations in the SH1 domain attenuate inhibitor binding. While dasatinib, nilotinib, and bosutinib remain efficacious against the drug-resistant mutant forms of various imatinib of Bcr-Abl1, the mutation in which threonine-315 residue is replaced with isoleucine (T315I) remains insensitive to all three drugs and can lead to the development of resistance to treatment in CML patients. Thus, there remains an unmet medical need for inhibiting Bcr-Abl1 mutations, such as T315I.
Active agents that target the myristoyl binding site (known as allosteric isomers) have potential for the treatment of Bcr-Abl1 disorders (Targeting BCR-ABL by combining allosteric with ATP-binding-site inhibitors. Nature 2010; 463: 501-6). To prevent the emergence of resistance to the use of ATP inhibitors and/or allosteric inhibitors, combination therapies using both types of inhibitors may be developed for the treatment of Bcr-Abl 1-related disorders. In particular, there is a need for active small molecule classes or combinations thereof that inhibit the Bcr-Abl1 and Bcr-Abl1 mutations via the ATP binding site, the myristoyl binding site, or a combination of these two sites.
ABL-001 (also known as Asciminib, chemical name (R) -N- (4- (chlorodifluoromethoxy) phenyl) -6- (3-hydroxypyrrolidin-1-yl) -5- (1H-pyrazol-5-yl) nicotinamide, which has the following structural formula) is an allosteric ABL1 kinase inhibitor developed by nova pharmaceutical company, targets the myristoyl pocket of ABL1 to inactivate it, and is combined with an ATP-competitive inhibitor of BCR-ABL tyrosine kinase to effectively prevent the emergence of resistance to ATP inhibitors and/or allosteric inhibitor applications. It has been demonstrated that ABL-001, in combination with the BCR-ABL inhibitor nilotinib, can act as a curative effect on CML in a mouse model (Andrew A. Wylie et al (2017) Nature 543, 733-737). Norwalk is developing clinical treatment regimens for ABL-001 in combination with multiple ATP-competitive BCR-ABL inhibitors, including imatinib, nilotinib, and dasatinib.
Figure BDA0001964663670000021
Poor absorption, distribution, metabolism and/or excretion (ADME) properties are known to be the major cause of failure in many drug candidate clinical trials. Many drugs currently on the market also have limited their range of application due to poor ADME properties. The rapid metabolism of drugs can result in the difficulty of obtaining many drugs that are otherwise effective in treating disease due to their rapid metabolic clearance from the body. Although frequent or high dose administration may solve the problem of rapid clearance of the drug, this method may cause problems such as poor patient compliance, side effects caused by high dose administration, and increased treatment costs. In addition, rapidly metabolizing drugs may also expose patients to undesirable toxic or reactive metabolites.
Although ABL-001 is effective as an allosteric inhibitor of ABL1 kinase in the treatment of CML and other disorders, it is a challenging task to find novel compounds that have good oral bioavailability and druggability in the treatment of CML and other disorders. Accordingly, there remains a need in the art to develop compounds having selective inhibitory activity or better pharmacodynamics/pharmacokinetics for Bcr-Abl1 kinase mediated diseases useful as therapeutic agents, and the present invention provides such compounds.
Summary of The Invention
Aiming at the technical problems, the invention discloses a novel deuterium-substituted nicotinamide compound, a composition and application thereof, wherein the novel deuterium-substituted nicotinamide compound has better BCR-ABLT315IThe inhibitor has the advantages of inhibiting activity, lower side effect and better pharmacodynamics/pharmacokinetics performance, and can be used for treating Bcr-Abl1 kinase mediated diseases.
As used herein, the term "compounds of the invention" refers to compounds of formulae (I) - (IV) (including subsets of compounds of formula (IIIa)). The term also includes pharmaceutically acceptable salts, prodrugs, hydrates or solvate, polymorphs, stereoisomers or isotopic variations of the compounds of formulae (I) - (IV).
In contrast, the invention adopts the following technical scheme:
in a first aspect of the invention, there is provided a compound of formula (I):
Figure BDA0001964663670000031
wherein the content of the first and second substances,
x is selected from-OH or halogen;
y is selected from
Figure BDA0001964663670000032
R1、R2、R3、R4、R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R12、R13、R14And R15Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, polymorph, stereoisomer, or isotopic variant thereof.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient. In a specific embodiment, the compounds of the present invention are provided in an effective amount in the pharmaceutical composition. In particular embodiments, the compounds of the present invention are provided in a therapeutically effective amount. In particular embodiments, the compounds of the present invention are provided in a prophylactically effective amount. In particular embodiments, the compositions of the present invention comprise other therapeutic agents. In particular embodiments, the other therapeutic agent is a different inhibitor of Bcr-Abl 1.
In another aspect, the present invention provides a method for preparing the pharmaceutical composition as described above, comprising the steps of: pharmaceutically acceptable excipients are mixed with the compounds of the present invention to form pharmaceutical compositions.
In another aspect, the invention also relates to providing a method of treating a condition mediated by Bcr-Abl1 in a subject. The method comprises administering to the subject a therapeutically effective amount of a compound of the invention. In particular embodiments, the disorder is mediated by Bcr-Abl 1. In particular embodiments, the compound is administered orally, subcutaneously, intravenously, or intramuscularly. In particular embodiments, the compound is administered chronically. In particular embodiments, the condition is selected from the group consisting of a solid tumor, sarcoma, acute lymphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, gastrointestinal stromal tumor, thyroid cancer, gastric cancer, rectal cancer, multiple myeloma, neoplasia and other proliferative or proliferative diseases, viral infections or CNS disorders.
Other objects and advantages of the present invention will be apparent to those skilled in the art from the following detailed description, examples and claims.
Detailed Description
Definition of
Herein, "deuterated", unless otherwise specified, means that one or more hydrogens of a compound or group are replaced with deuterium; deuterium can be mono-, di-, poly-, or fully substituted. The terms "deuterated one or more" and "deuterated one or more" are used interchangeably.
Herein, unless otherwise specified, "non-deuterated compound" means a compound containing deuterium at an atomic ratio of deuterium not higher than the natural deuterium isotope content (0.015%).
"halogen" or "halo" refers to fluorine (F), chlorine (Cl), bromine (Br), and iodine (I). In some embodiments, the halogen group is F, Cl or Br. In some embodiments, the halogen group is F. In some embodiments, the halogen group is Cl. In some embodiments, the halogen group is Br.
The term "pharmaceutically acceptable salts" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without excessive toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, the pharmaceutically acceptable salts are described in detail by Berge et al in J.pharmaceutical Sciences (1977)66: 1-19. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases.
The invention also includes isotopically-labeled compounds, equivalent to those disclosed herein as the original compound. Examples of isotopes that can be listed as compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine,respectively as2H,3H,13C,14C,15N,17O,18O,31P,32P,35S,18F and36and (4) Cl. The compounds of the present invention, or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates thereof, wherein isotopes or other isotopic atoms containing such compounds are within the scope of the present invention. Certain isotopically-labelled compounds of the invention, e.g.3H and14among these, the radioactive isotope of C is useful in tissue distribution experiments of drugs and substrates. Tritium, i.e.3H and carbon 14, i.e.14C, their preparation and detection are relatively easy, and are the first choice among isotopes. Isotopically labeled compounds can be prepared by conventional methods by substituting readily available isotopically labeled reagents for non-isotopically labeled reagents using the protocols set forth in the examples.
The compounds of the invention may include one or more asymmetric centers, and thus may exist in a variety of "stereoisomeric" forms, e.g., enantiomeric and/or diastereomeric forms. For example, the compounds of the present invention may be individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. Isomers may be separated from mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
The compounds of the present invention may be in amorphous or crystalline form. Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "crystalline form" refers to the different arrangements of chemical drug molecules, typically expressed as the presence of the drug substance in the solid state. One drug can exist in a plurality of crystal form substances, and different crystal forms of the same drug can be dissolved and absorbed in vivo differently, so that the dissolution and release of the preparation can be influenced.
The term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio. "hydrate" refers to a complex formed by coordination of a compound of the present invention with water.
The term "prodrug" refers to a compound that is converted in vivo by hydrolysis, for example in the blood, to its active form with a medicinal effect. Pharmaceutically acceptable Prodrugs are described in t.higuchi and v.stella, Prodrugs as Novel Delivery Systems, vol.14 of a.c.s.symposium Series, Edward b.roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and d.fleisher, s.ramon and h.bara "Improved oral Delivery: solubility limits overview by the use of drivers, Advanced Drug Delivery Reviews (1996)19(2)115-130, each of which is incorporated herein by reference.
A prodrug is any covalently bonded compound of the present invention that releases the parent compound in vivo when such prodrug is administered to a patient. Prodrugs are generally prepared by modifying functional groups in a manner such that the modification is effected by routine manipulation or in vivo cleavage to produce the parent compound. Prodrugs include, for example, compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, cleaves to form a hydroxy, amino, or sulfhydryl group. Thus, representative examples of prodrugs include, but are not limited to, acetate/amide, formate/amide, and benzoate/amide derivatives of hydroxy, mercapto, and amino functional groups of the compounds of formula (I). In addition, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like may be used. The ester itself may be active and/or may hydrolyze under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those which readily break down in the human body to release the parent acid or salt thereof.
The term "crystalline form" refers to the different arrangements of chemical drug molecules, typically expressed as the presence of the drug substance in the solid state. One drug can exist in a plurality of crystal form substances, and different crystal forms of the same drug can be dissolved and absorbed in vivo differently, so that the dissolution and release of the preparation can be influenced.
As used herein, the term "subject" includes, but is not limited to: a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., an infant, a child, an adolescent) or an adult subject (e.g., a young adult, a middle-aged adult, or an older adult)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog. In some embodiments, the subject is a human. In other embodiments, the subject is a non-human animal.
"disease," "disorder," and "condition" are used interchangeably herein.
As used herein, unless otherwise specified, the term "treatment" includes the effect that occurs when a subject has a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition ("therapeutic treatment"), and also includes the effect that occurs before the subject begins to have the particular disease, disorder or condition ("prophylactic treatment").
Generally, an "effective amount" of a compound is an amount sufficient to elicit a biological response of interest. As will be appreciated by those of ordinary skill in the art, the effective amount of a compound of the present invention may vary depending on the following factors: for example, biological goals, pharmacokinetics of the compound, the disease being treated, mode of administration, and the age, health, and condition of the subject. An effective amount includes both therapeutically and prophylactically therapeutically effective amounts.
As used herein, unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder, or condition, or to delay or minimize one or more symptoms associated with a disease, disorder, or condition. A therapeutically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder, or condition. The term "therapeutically effective amount" can include an amount that improves the overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic efficacy of other therapeutic agents.
As used herein, unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or one or more symptoms associated with a disease, disorder, or condition, or to prevent recurrence of a disease, disorder, or condition. A prophylactically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in preventing a disease, disorder, or condition. The term "prophylactically effective amount" can include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic efficacy of other prophylactic agents.
"combination" and related terms refer to the simultaneous or sequential administration of the therapeutic agents of the present invention. For example, a compound of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or simultaneously with another therapeutic agent in a single unit dosage form.
Detailed Description
Compound (I)
In one embodiment, the present invention provides a compound of formula (I), or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, polymorph, stereoisomer, or isotopic variant thereof:
Figure BDA0001964663670000071
wherein the content of the first and second substances,
x is selected from-OH or halogen;
y is selected from
Figure BDA0001964663670000072
R1、R2、R3、R4、R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R12、R13、R14And R15Each independently selected from hydrogen or deuterium;
with the proviso that the above compound contains at least one deuterium atom.
In another embodiment, the present invention provides a compound of formula (II), or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, polymorph, stereoisomer, or isotopic variant thereof:
Figure BDA0001964663670000073
wherein the content of the first and second substances,
x is selected from-OH or halogen;
y is selected from
Figure BDA0001964663670000081
R1、R2、R3、R4、R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R12、R13、R14And R15Each independently selected from hydrogen or deuterium;
with the proviso that the above compound contains at least one deuterium atom.
In one embodiment, the compounds of the present invention contain at least one deuterium atom, preferably one deuterium atom, more preferably two deuterium atoms, more preferably three deuterium atoms, more preferably four deuterium atoms, more preferably five deuterium atoms, more preferably six deuterium atoms, more preferably seven deuterium atoms, more preferably eight deuterium atoms, more preferably nine deuterium atoms, more preferably ten deuterium atoms, more preferably eleven deuterium atoms, more preferably twelve deuterium atoms, more preferably thirteen deuterium atoms, more preferably fourteen deuterium atoms, more preferably fifteen deuterium atoms.
In another embodiment, the deuterium isotope content of deuterium at the deuterated position is at least 0.015% greater than the natural deuterium isotope content, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
Specifically, in the present invention R1、R2、R3、R4、R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R12、R13、R14And R15The deuterium isotope content in each deuterated position is at least 5%, preferably greater than 10%, more preferably greater than 15%, more preferably greater than 20%, more preferably greater than 25%, more preferably greater than 30%, more preferably greater than 35%, more preferably greater than 40%, more preferably greater than 45%, more preferably greater than 50%, more preferably greater than 55%, more preferably greater than 60%, more preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, more preferably greater than 99%.
In another embodiment, R of a compound of the invention1、R2、R3、R4、R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R12、R13、R14And R15Preferably, at least one of the deuterium containing groups comprises deuterium, more preferably two deuterium containing groups, more preferably three deuterium containing groups, more preferably four deuterium containing groups, more preferably five deuterium containing groups, more preferably six deuterium containing groups, more preferably seven deuterium containing groups, more preferably eight deuterium containing groups, more preferably nine deuterium containing groups, more preferably ten deuterium containing groups, more preferably eleven deuterium containing groups, more preferably twelve deuterium containing groups, more preferably thirteen deuterium containing groups, more preferably fourteen deuterium containing groups, more preferably fifteen deuterium containing groups. In particular, the compounds of the present invention contain at least one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen deuterium atoms.
In another embodiment, R1、R2、R3、R4、R5、R6And R7Each independently selected from hydrogen or deuterium. In another embodiment, R1Is hydrogen; in another embodiment, R1Is deuterium; in another embodiment, R2Is hydrogen; in another embodiment, R2Is deuterium; in another embodiment, R3Is hydrogen; in another embodiment, R3Is deuterium; in another embodiment, R4Is hydrogen; in another embodiment, R4Is deuterium; in another embodiment, R5Is hydrogen; in another embodiment, R5Is deuterium; in another embodiment, R6Is hydrogen; in another embodiment, R6Is deuterium; in another embodiment, R7Is hydrogen; in another embodiment, R7Is deuterium; in another embodiment, R1And R2Are the same; in another embodiment, R3And R4Are the same; in another embodiment, R5And R6Are the same; in another embodiment, R1、R2、R3And R4Is hydrogen; in another embodiment, R1、R2、R3And R4Is deuterium.
In another embodiment, R8And R9Each independently selected from hydrogen or deuterium. In another embodiment, R8Is hydrogen; in another embodiment, R8Is deuterium; in another embodiment, R9Is hydrogen; in another embodiment, R9Is deuterium; in another embodiment, R8And R9Is hydrogen; in another embodiment, R8And R9Is deuterium.
In another embodiment, R8’、R9’And R10’Are independently selected fromFrom hydrogen or deuterium. In another embodiment, R8’Is hydrogen; in another embodiment, R8’Is deuterium; in another embodiment, R9’Is hydrogen; in another embodiment, R9’Is deuterium; in another embodiment, R10’Is hydrogen; in another embodiment, R10’Is deuterium; in another embodiment, R8’、R9’And R10’Is hydrogen; in another embodiment, R8’、R9’And R10’Is deuterium.
In another embodiment, R10And R11Each independently selected from hydrogen or deuterium. In another embodiment, R10Is hydrogen; in another embodiment, R10Is deuterium; in another embodiment, R11Is hydrogen; in another embodiment, R11Is deuterium; in another embodiment, R10And R11Is hydrogen; in another embodiment, R10And R11Is deuterium.
In another embodiment, R12、R13、R14And R15Each independently selected from hydrogen or deuterium. In another embodiment, R12Is hydrogen; in another embodiment, R12Is deuterium; in another embodiment, R13Is hydrogen; in another embodiment, R13Is deuterium; in another embodiment, R14Is hydrogen; in another embodiment, R14Is deuterium; in another embodiment, R15Is hydrogen; in another embodiment, R15Is deuterium; in another embodiment, R12And R13Is hydrogen; in another embodiment, R12And R13Is deuterium; in another embodiment, R14And R15Is hydrogen; in another embodiment, R14And R15Is deuterium; in another embodiment, R12、R13、R14And R15Is hydrogen; in another embodiment, R12、R13、R14And R15Is deuterium.
In another embodiment, R12And R13Is deuterium, R1、R2、R3、R4、R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R14And R15Each independently selected from hydrogen or deuterium. In another embodiment, R12And R13Is deuterium, R1、R2、R3And R4Is deuterium, R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R14And R15Is hydrogen.
In another embodiment, the invention relates to compounds of formula (III):
Figure BDA0001964663670000101
wherein the content of the first and second substances,
x is selected from-OH or halogen;
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14and R15Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, crystal form, stereoisomer, or isotopic variant thereof.
In another embodiment, the invention relates to compounds of formula (IIIa):
Figure BDA0001964663670000102
wherein the content of the first and second substances,
x is selected from-OH or halogen;
R1、R2、R3、R4、R5、R6、R7、R12、R13、R14and R15Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
preferably, X is selected from-OH or F;
preferably, R14And R15Is hydrogen;
preferably, R5、R6And R7Is hydrogen;
preferably, R12And R13Is deuterium;
preferably, R1、R2、R3And R4Is deuterium;
or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, crystal form, stereoisomer, or isotopic variant thereof.
In one embodiment, X is selected from-OH, F or Cl. In another embodiment, X is selected from-OH. In another embodiment, X is selected from F.
In one embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R14And R15Is hydrogen, R1、R2、R3、R4、R5、R6、R7、R12And R13Each independently selected from hydrogen or deuterium, provided that the above compounds contain at least one deuterium atom.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R5、R6And R7Is hydrogen, R1、R2、R3、R4、R12、R13、R14And R15Each independently selected from hydrogen or deuterium, provided thatThe above compound contains at least one deuterium atom.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R5、R6、R7、R14And R15Is hydrogen, R1、R2、R3、R4、R12And R13Each independently selected from hydrogen or deuterium, provided that the above compounds contain at least one deuterium atom.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R12And R13Is deuterium, R1、R2、R3、R4、R5、R6、R7、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R12And R13Is deuterium, R14And R15Is hydrogen, R1、R2、R3、R4、R5、R6And R7Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R12And R13Is deuterium, R5、R6And R7Is hydrogen, R1、R2、R3、R4、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R12And R13Is deuterium, R5、R6、R7、R14And R15Is hydrogen, R1、R2、R3And R4Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R5、R6、R7、R12、R13、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R14And R15Is hydrogen, R5、R6、R7、R12And R13Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R5、R6And R7Is hydrogen, R12、R13、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R5、R6、R7、R14And R15Is hydrogen, R12And R13Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R5、R6、R7、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R14And R15Is hydrogen, R5、R6And R7Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R5、R6And R7Is hydrogen, R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IIIa) wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R5、R6、R7、R14And R15Is hydrogen.
In another embodiment, the present invention relates to a compound of formula (a), or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, crystal form, stereoisomer, or isotopic variant thereof:
Figure BDA0001964663670000121
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14and R15Each independently selected from hydrogen or deuterium;
provided that R is1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14And R15At least one of which is deuterated or deuterium.
Furthermore, R1-R15As in the compound of formula (III) for R1-R15As defined.
In another embodiment, the invention relates to compounds of formula (IV):
Figure BDA0001964663670000131
wherein the content of the first and second substances,
x is selected from-OH or halogen;
R1、R2、R3、R4、R5、R6、R7、R8’、R9’、R10’、R10、R11、R12、R13、R14and R15Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, crystal form, stereoisomer, or isotopic variant thereof.
In another embodiment, the invention relates to compounds of formula (IVa):
Figure BDA0001964663670000132
wherein the content of the first and second substances,
x is selected from-OH or halogen;
R1、R2、R3、R4、R5、R6、R7、R12、R13、R14and R15Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
preferably, X is selected from-OH or F;
preferably, R14And R15Is hydrogen;
preferably, R5、R6And R7Is hydrogen;
preferably, R12And R13Is deuterium;
preferably, R1、R2、R3And R4Is deuterium;
or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, crystal form, stereoisomer, or isotopic variant thereof.
In one embodiment, X is selected from-OH, F or Cl. In another embodiment, X is selected from-OH. In another embodiment, X is selected from F.
In one embodiment, the invention relates to compounds of formula (IVa) wherein X is selected from OH, R14And R15Is hydrogen, R1、R2、R3、R4、R5、R6、R7、R12And R13Each independently selected from hydrogen or deuterium, provided that the above compounds contain at least one deuterium atom.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R5、R6And R7Is hydrogen, R1、R2、R3、R4、R12、R13、R14And R15Each independently selected from hydrogen or deuterium, provided that the above compounds contain at least one deuterium atom.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R5、R6、R7、R14And R15Is hydrogen, R1、R2、R3、R4、R12And R13Each independently selected from hydrogen or deuterium, provided that the above compounds contain at least one deuterium atom.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R12And R13Is deuterium, R1、R2、R3、R4、R5、R6、R7、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R12And R13Is deuterium, R14And R15Is hydrogen, R1、R2、R3、R4、R5、R6And R7Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R12And R13Is a deuterium compound, and is a hydrogen atom,R5、R6and R7Is hydrogen, R1、R2、R3、R4、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R12And R13Is deuterium, R5、R6、R7、R14And R15Is hydrogen, R1、R2、R3And R4Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R5、R6、R7、R12、R13、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R14And R15Is hydrogen, R5、R6、R7、R12And R13Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R5、R6And R7Is hydrogen, R12、R13、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3And R4Is deuterium, R5、R6、R7、R14And R15Is hydrogen, R12And R13Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R5、R6、R7、R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R14And R15Is hydrogen, R5、R6And R7Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R5、R6And R7Is hydrogen, R14And R15Each independently selected from hydrogen or deuterium.
In another embodiment, the invention relates to compounds of formula (IVa), wherein X is selected from OH, R1、R2、R3、R4、R12And R13Is deuterium, R5、R6、R7、R14And R15Is hydrogen.
In another embodiment, the present invention relates to a compound of formula (B), or a pharmaceutically acceptable salt, prodrug, hydrate, or solvate, crystal form, stereoisomer, or isotopic variant thereof:
Figure BDA0001964663670000151
wherein the content of the first and second substances,
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15and R16Each independently selected from hydrogen or deuterium;
provided that R is1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15And R16At least one of which is deuterated or deuterium.
Furthermore, R1-R6、R10-R15、R7-R9And R16As in the compound of formula (IV) for R1-R6、R10-R15、R8’-R10’And R7As defined.
As a preferred embodiment of the present invention, the compound is selected from the group consisting of:
Figure BDA0001964663670000152
Figure BDA0001964663670000161
Figure BDA0001964663670000171
Figure BDA0001964663670000181
as a preferred embodiment of the present invention, the compounds do not include non-deuterated compounds.
Pharmaceutical compositions and methods of administration
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of an active ingredient.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 0.5-2000mg of a compound of the invention per dose, more preferably, 1-500mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable excipient" refers to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compounds formulated together. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
The pharmaceutical composition of the present invention can be prepared by combining the compound of the present invention with a suitable pharmaceutically acceptable excipient, and can be formulated, for example, into solid, semi-solid, liquid or gaseous preparations such as tablets, pills, capsules, powders, granules, pastes, emulsions, suspensions, solutions, suppositories, injections, inhalants, gels, microspheres, aerosols, and the like.
Typical routes of administration of the compounds of the present invention or pharmaceutical compositions thereof include, but are not limited to, oral, rectal, transmucosal, enteral, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intrapleural, intramuscular, subcutaneous, intravenous administration.
The pharmaceutical compositions of the present invention may be manufactured by methods well known in the art, such as conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, lyophilizing, and the like.
For oral administration, the pharmaceutical compositions may be formulated by mixing the active compound with pharmaceutically acceptable excipients well known in the art. These excipients enable the compounds of the present invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, slurries, suspensions and the like, for oral administration to a patient.
Solid oral compositions may be prepared by conventional mixing, filling or tableting methods. For example, it can be obtained by the following method: the active compounds are mixed with solid excipients, the resulting mixture is optionally milled, if desired with further suitable auxiliaries, and the mixture is then processed to granules, to give tablets or dragee cores. Suitable excipients include, but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like. Such as microcrystalline cellulose, glucose solutions, gum arabic syrups, gelatin solutions, sucrose and starch pastes; talc, starch, calcium stearate or stearic acid; lactose, sucrose, starch, mannitol, sorbitol, or dicalcium phosphate; silicon dioxide; croscarmellose sodium, pregelatinized starch, sodium starch glycolate, alginic acid, corn starch, potato starch, methylcellulose, agar, hydroxymethylcellulose, cross-linked polyvinylpyrrolidone, and the like. The dragee cores may optionally be coated, in particular with enteric coatings, according to methods well known in normal pharmaceutical practice.
The pharmaceutical compositions may also be adapted for parenteral administration, as sterile solutions, suspensions or lyophilized products in suitable unit dosage forms. Suitable excipients, such as fillers, buffers or surfactants can be used.
The compounds of the invention may be administered by any route and method of use, for example, by oral or parenteral (e.g., intravenous) administration. A therapeutically effective amount of a compound of the invention is from about 0.0001 to 20mg/kg body weight/day, for example from 0.001 to 10mg/kg body weight/day.
The frequency of dosage of the compounds of the invention is determined by the individual requirements of the patient, for example 1 or 2 times per day, or more times per day. Administration may be intermittent, for example, wherein a patient receives a daily dose of a compound of the invention over a period of several days, followed by a period of several days or more in which the patient does not receive a daily dose of a compound of the invention.
Treatment of
The compounds of the invention are also useful for treating the following diseases, disorders or conditions mediated by Bcr-Abl1 kinase: respiratory diseases, allergies, rheumatoid arthritis, osteoarthritis, rheumatic disorders, psoriasis, ulcerative colitis, crohn's disease, septic shock, proliferative disorders, atherosclerosis, allograft rejection following transplantation, diabetes, stroke, obesity or restenosis, leukemia, interstitial tumors, thyroid cancer, systemic mastocytosis, hypereosinophilic syndrome, fibrosis, polyarthritis, scleroderma, lupus erythematosus, graft versus host disease, neurofibromatosis, pulmonary hypertension, alzheimer's disease, seminoma, dysgerminoma, mast cell tumors, lung cancer, bronchial carcinoma, dysgerminoma, intraepithelial neoplasia, melanoma, breast cancer, neuroblastoma, papillary/follicular parathyroid hyperplasia/adenoma, colon cancer, and colon cancer, Colorectal adenomas, ovarian cancer, prostate cancer, glioblastoma, brain tumors, malignant gliomas, pancreatic cancer, malignant pleural mesothelioma, hemangioblastoma, hemangioma, kidney cancer, liver cancer, adrenal cancer, bladder cancer, stomach cancer, rectal cancer, vaginal cancer, cervical cancer, endometrial cancer, multiple myeloma, neck and head tumors, neoplasia and other proliferative or proliferative diseases, or a combination thereof.
The present invention thus provides compounds of the invention for use in therapy, particularly in the treatment of diseases and conditions mediated by inappropriate Bcr-Abl1 activity.
Inappropriate Bcr-Abl1 activity as referred to herein is any Bcr-Abl1 activity that deviates from the expected normal Bcr-Abl1 activity in a particular mammalian subject. Inappropriate Bcr-Abl1 activity can be in the form of, for example: abnormal increase in activity, or a disruption in the timing and or control of Bcr-Abl1 activity. Such inappropriate activity may then result, for example, from overexpression or mutation of the protein kinase leading to inappropriate or uncontrolled activation.
In another embodiment, the invention relates to methods of modulating, modulating or inhibiting Bcr-Abl1 for the purpose of preventing and/or treating conditions associated with dysregulated or inappropriate Bcr-Abl1 activity.
In another embodiment, the disorder mediated by Bcr-Abl1 activity is a respiratory disease. In another embodiment, the disorder is a proliferative disorder. In yet another embodiment, the disorder is cancer. In another embodiment, the disorder is leukemia.
In another embodiment, the compounds of the present invention may also be used for the treatment of neurodegeneration. While the native C-ABL tyrosine kinase remains relatively quiescent in healthy adult brain, it can be activated in the brain of patients with CNS diseases including neurodegenerative diseases such as Alzheimer's Disease (AD), parkinson's disease (AD), frontotemporal dementia (FTD), pick's disease, niemann-pick disease type C (NPC) and other degenerative, inflammatory and autoimmune diseases and aging.
The compounds of the invention may be used in combination with one or more existing therapies for the above diseases: for example, the compounds of the present invention may be used in combination with levodopa or other L-DOPA containing drugs or dopamine receptor agonists for the treatment of parkinson's disease or in combination with cholinesterase inhibitors such as esnerat capsules or transdermal patches for the treatment of alzheimer's disease.
An effective amount of a compound of the invention is generally in a mean daily dose of from 0.01mg to 50mg of compound per kilogram of body weight of the patient, preferably from 0.1mg to 25mg of compound per kilogram of body weight of the patient, in single or multiple administrations. Generally, the compounds of the present invention may be administered to such patients in need of such treatment in a daily dosage range of from about 1mg to about 3500mg, preferably from 10mg to 1000mg per patient. For example, the daily dose per patient may be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000 mg. Administration may be once or more daily, weekly (or at intervals of several days), or on an intermittent schedule. For example, the compound may be administered one or more times per day on a weekly basis (e.g., monday), indefinitely or for several weeks, e.g., 4-10 weeks. Alternatively, the administration may be continued daily for several days (e.g., 2-10 days), followed by several days (e.g., 1-30 days) without administration of the compound, with the cycle repeated indefinitely or for a given number of times, e.g., 4-10 cycles. For example, the compounds of the invention may be administered daily for 5 days, followed by 9 days, followed by 5 days, followed by 9 days, and so on, with the cycle repeated indefinitely or 4-10 times in total.
Combination therapy
The compounds of the present invention may be used alone or in combination with other therapeutic agents for the treatment of Bcr-Abl1 mediated diseases associated with inappropriate Bcr-Abl1 activity. The combination therapy according to the invention therefore comprises the administration of at least one compound according to the invention and the use of at least one further pharmaceutically active agent. One or more compounds of the present invention and one or more other pharmaceutically active agents may be administered together or separately, and when administered separately, may be administered simultaneously or sequentially in any order. The amounts and relative timing of administration of the one or more compounds of the present invention and the one or more other pharmaceutically active agents will be selected to achieve the desired combined therapeutic effect.
The compounds of the present invention have a number of advantages over the non-deuterated compounds known in the prior art. The advantages of the invention include: first, the compounds and compositions of the present invention provide a more advantageous therapeutic tool for the treatment of Bcr-Abl 1-mediated diseases. Second, the metabolism of the compound in the organism is improved, giving the compound better pharmacokinetic parameters. In this case, the dosage can be varied and a long acting formulation formed, improving the applicability. Thirdly, the medicine concentration of the compound in the animal body is improved, and the medicine curative effect is improved. Fourth, certain metabolites are inhibited, increasing the safety of the compounds.
Examples
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Parts and percentages are parts and percentages by weight unless otherwise indicated.
In general, in the preparative schemes, each reaction is usually carried out in an inert solvent at a temperature ranging from room temperature to reflux temperature (e.g., from 0 ℃ to 100 ℃, preferably from 0 ℃ to 80 ℃). The reaction time is usually 0.1 to 60 hours, preferably 0.5 to 24 hours.
Abbreviations
EtOD: ethanol-d
PtO2: platinum dioxide
HATU: 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate
DIPEA: n, N-diisopropylethylamine
DMF: n, N-dimethylformamide
BH (pin): pinacol borane
Pd(PPh3)4: tetrakis (triphenylphosphine) palladium
K3PO4: potassium phosphate
A Dioxane: dioxane
Na2CO3: sodium carbonate
DME: ethylene glycol dimethyl ether
xylene: xylene
LiAlD4: lithium aluminum hydride-d4
MeOD: methanol-d
DAST: diethylaminosulfur trifluoride
2Example 1(R) -N- (4- (chlorodifluoromethoxy) phenyl-2, 6-d) -6- (3-hydroxypyrrolidin-1-yl) -5- Preparation of (isothiazol-4-yl) nicotinamide (Compound A-1).
Figure BDA0001964663670000221
The synthesis was carried out using the following route:
Figure BDA0001964663670000231
step 1: synthesis of Compound 1
4- (chlorodifluoromethoxy) aniline (6.0g, 31mmol) is dissolved in 30mL ethanol-d, platinum dioxide (60mg,0.26mmol) is added to be sealed and dispersed evenly, and the mixture is heated by microwave for 120 minutes at 160 ℃. After the temperature is reduced to room temperature, the mixture is filtered, concentrated and subjected to column chromatography to obtain 2.86g of brown oily liquid, namely the compound 1. The yield thereof was found to be 47.1%. LC-MS (APCI): M/z 196.1, 198.1(M +1)+
Step 2: synthesis of Compound 2
Compound 1(2.86g,14.6mmol) was dissolved in 20mL of DMF, and 5-bromo-6-chloronicotinic acid (3.45g,14.6mmol), HATU (6.66g,17.52mmol) and DIPEA (3.77g,29.2mmol) were added at room temperature, followed by stirring at room temperature for 2 hours. The reaction solution was poured into 100mL of water, extracted three times with 20mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 3/1) to obtain 3.65g of a white solid powder, which was the compound 2. The yield thereof was found to be 60.3%.1H NMR(500MHz,DMSO-d6)δ10.69(s,1H),8.91(d,J=2.2Hz,1H),8.77–8.69(m,1H),7.42–7.33(m,2H)。
And step 3: synthesis of Compound 3
Compound 2(1.5g,3.62mmol), (R) -3-hydroxypyrrolidine (0.38g,4.35mmol) and DIPEA (0.94g,7.25mmol) were dissolved in 20mL DMF and heated to 130 deg.C and stirred for 2 hours. The reaction solution was poured into 100mL of water, extracted three times with 20mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 1.41g of a white solid powder. The yield thereof was found to be 83.8%. LC-MS (APCI): M/z 464.0, 466.0(M +1)+
And 4, step 4: synthesis of Compound A-1
Dissolving compound 3(650mg,1.40mmol) in 15mL dioxane, adding Pd (PPh) at room temperature under nitrogen protection3)4(48mg,0.042mmol)、K3PO4(891mg,4.20mmol) was stirred and dispersed uniformly, and pinacolborane (1.07g, 8.39mmol) was added slowly and stirred at 110 ℃ for 10 hours under nitrogen. Cooled to room temperature, filtered, the filter cake washed with dichloromethane, the filtrate concentrated and the brown oily liquid used directly in the next step. LC-MS (APCI): M/z 512.1, 514.1(M +1)+
The oily liquid was dissolved in 10mL DME and 2mL water, and 4-bromoisothiazole (275mg, 1.68mmol), Pd (PPh) and nitrogen were added3)4(48mg, 0.042mmol) and Na2CO3(296mg, 2.80mmol), the reaction was stirred at 100 ℃ for 2 hours under nitrogen. After cooling to room temperature, the reaction mixture was poured into 60mL of water, extracted three times with 20mL of ethyl acetate, and the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 72mg of a white solid powder. LC-MS (APCI) M/z 469.1(M +1)+1H NMR(300MHz,DMSO-d6)δ10.19(s,1H),9.04(s,1H),8.75(d,J=2.4Hz,1H),8.70(s,1H),8.05(d,J=2.4Hz,1H),7.33(dq,J=7.7,1.1Hz,2H),4.89(d,J=3.5Hz,1H),4.20(d,J=4.3Hz,1H),4.12(q,J=5.2Hz,1H),3.40(t,J=5.1Hz,1H),3.30–3.18(m,2H),,2.93–2.82(m,1H)。
4Example 2(R) -N- (4- (chlorodifluoromethoxy) phenyl) -6- (3-hydroxypyrrolidin-1-yl-2, 2,5,5-d) - Preparation of 5- (isothiazol-4-yl) nicotinamide (Compound A-2).
Figure BDA0001964663670000241
The synthesis was carried out using the following route:
Figure BDA0001964663670000242
step 1: synthesis of Compound 4
D-malic acid (10.0g, 74.6mmol) and benzylamine (9.59g, 89.5mmol) were dispersed in 250mL xylene and heated for 8 hours to dehydrate. After cooling to room temperature, cooling and stirring for 2 hours in an ice bath, filtering, washing a filter cake with petroleum ether, taking the filter cake, and performing column chromatography (petroleum ether/ethyl acetate, 1/2) to obtain 9.91g of white solid powder, namely the compound 4. Yield: 64.8 percent. LC-MS (APCI): M/z 206.1(M +1)+
Step 2: synthesis of Compound 5
Taking LiAlD4(2.45g, 58.3mmol) in 70mL of anhydrous tetrahydrofuran, and a solution of Compound 4(3.0g, 14.6mmol) in tetrahydrofuran (70mL) was added dropwise slowly at-15 deg.C, and the mixture was stirred overnight at room temperature after dropping. Adding a small amount of sodium sulfate decahydrate for quenching, filtering the reaction solution, washing filter residues by using 100mL of ethyl acetate, concentrating the filtrate, and obtaining 1.26g of colorless oily liquid, namely the compound 5 which is directly used in the next step.
And step 3: synthesis of Compound 6
Compound 5(526mg, 2.9mmol) was dissolved in 10mL of MeOD and hydrogenated at 50 ℃ overnight. Filtration was carried out, and the residue was washed with 30mL of ethyl acetate, and the filtrate was concentrated to obtain 255mg of a brown oily liquid, which was Compound 6 and was used in the next step.
And 4, step 4: synthesis of Compound 7
The compound 4- (chlorodifluoromethoxy) aniline (2.86g,14.6mmol) was dissolved in 20mL DMF, and 5-bromo-6-chloronicotinic acid (3.45g,14.6mmol), HATU (6.66g,17.52mmol) and DIPEA (3.77g,29.2mmol) were added at room temperature and stirred at room temperature for 2 hours. The reaction solution was poured into 100mL of water, extracted three times with 20mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 3/1) to obtain 3.65g of a white solid powder. The yield thereof was found to be 60.3%.
And 5: synthesis of Compound 8
Compound 7(1.0g, 2.43mmol) and compound 6(266mg, 2.91mol) were dissolved in 5mL of DMF, DIPEA (628mg, 2.0mmol) was added at room temperature, and the reaction was stirred at 130 ℃ for 2 hours. The reaction mixture was poured into 50mL of water, extracted three times with 10mL of ethyl acetate, and the ethyl acetate was combined and washed with saturated brineWashing, drying over anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 1.03g of white solid powder. The yield thereof was found to be 90.9%. LC-MS (APCI): M/z-466.0, 468.0(M +1)+
Step 6: synthesis of Compound A-2
Dissolve compound 8(511mg, 1.09mmol) in 10mL dioxane, add Pd (PPh) at room temperature under nitrogen protection3)4(38mg,0.033mmol)、K3PO4(698mg,3.28mmol) was stirred and dispersed uniformly, and pinacolborane (841mg, 6.57mmol) was added slowly and stirred at 110 ℃ for 10 hours under nitrogen protection. Cooled to room temperature, filtered, the filter cake washed with dichloromethane, the filtrate concentrated and the brown oily liquid used directly in the next step. LC-MS (APCI): M/z 514.2, 516.2(M +1)+
Dissolving the oily liquid in 10mL of ethylene glycol dimethyl ether and 2mL of water, and adding 4-bromoisothiazole (215mg, 1.31mmol) and Pd (PPh) under the protection of nitrogen3)4(38mg,0.033mmol) and Na2CO3(348mg,3.28mmol), the reaction was stirred at 100 ℃ for 2 hours under nitrogen. After cooling to room temperature, the reaction mixture was poured into 60mL of water, extracted three times with 20mL of ethyl acetate, and the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 42mg of a white solid powder. LC-MS (APCI): M/z 471.2(M +1)+1H NMR(300MHz,DMSO-d6)δ10.19(s,1H),9.05(s,1H),8.75(d,J=2.4Hz,1H),8.70(s,1H),8.05(d,J=2.4Hz,1H),7.33(d,J=8.7Hz,3H),4.87(d,J=3.5Hz,1H),4.19(q,J=3.7Hz,1H),4.02(q,J=7.2Hz,1H)。
2Example 3(R) -N- (4- (chlorodifluoromethoxy) phenyl-2, 6-d) -6- (3-hydroxypyrrolidin-1-yl-2, 4preparation of 5,5-d) -5- (isothiazol-4-yl) nicotinamide (Compound A-3).
Figure BDA0001964663670000261
The synthesis was carried out using the following route:
Figure BDA0001964663670000262
step 1: synthesis of Compound 10
Compound 2(1.5g,3.62mmol), compound 6(0.38g,4.35mmol) and DIPEA (0.94g,7.25mmol) were dissolved in 20mL of DMF and stirred at 130 ℃ for 2 hours. The reaction solution was poured into 100mL of water, extracted three times with 20mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 1.47g of a white solid powder. The yield thereof was found to be 86.6%. LC-MS (APCI): M/z-468.0, 470.0(M +1)+
Step 2: synthesis of Compound A-3
Dissolve compound 10(387mg,0.83mmol) in 5mL dioxane, add Pd (PPh) at room temperature under nitrogen protection3)4(29mg,0.025mmol)、K3PO4(526mg,2.48mmol) was stirred and dispersed uniformly, and pinacolborane (634mg, 4.95mmol) was added slowly and stirred at 110 ℃ for 10 hours under nitrogen. Cooled to room temperature, filtered, the filter cake washed with dichloromethane, the filtrate concentrated and the brown oily liquid used directly in the next step. LC-MS (APCI): M/z ═ 516.2, 518.2(M +1)+
Dissolving the oily liquid in 10mL of ethylene glycol dimethyl ether and 2mL of water, and adding 4-bromoisothiazole (163mg, 0.99mmol) and Pd (PPh) under the protection of nitrogen3)4(29mg,0.025mmol) and Na2CO3(263mg,2.48mmol), the reaction was stirred at 100 ℃ for 2 hours under nitrogen. After cooling to room temperature, the reaction mixture was poured into 60mL of water, extracted three times with 20mL of ethyl acetate, and the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 43mg of a white solid powder. LC-MS (APCI) M/z 473.3(M +1)+1H NMR(300MHz,DMSO-d6)δ10.19(s,1H),9.04(s,1H),8.75(d,J=2.4Hz,1H),8.70(s,1H),8.05(d,J=2.4Hz,1H),7.33(dq,J=7.7,1.1Hz,2H),4.89(d,J=3.5Hz,1H),4.20(d,J=4.3Hz,1H),4.12(q,J=5.2Hz,1H)。
2Example 4(R) -N- (4- (chlorodifluoromethoxy) phenyl-2, 6-d)) -6- (3-fluoropyrrolidin-1-yl) -5- (pir-rolidin-1-yl) Preparation of oxazin-2-yl) nicotinamide (Compound A-1 a).
Figure BDA0001964663670000271
The synthesis was carried out using the following route:
Figure BDA0001964663670000272
step 1: synthesis of Compound 1
4- (chlorodifluoromethoxy) aniline (6.0g, 31mmol) was dissolved in 30mL EtOD, platinum dioxide (60mg,0.26mmol) was added, the mixture was sealed and dispersed uniformly, and the mixture was reacted at 160 ℃ for 120 minutes by microwave heating. After the temperature is reduced to room temperature, the mixture is filtered, concentrated and subjected to column chromatography to obtain 2.86g of brown oily liquid, namely the compound 1. The yield thereof was found to be 47.1%. LC-MS (APCI): M/z 196.1, 198.1(M +1)+
Step 2: synthesis of Compound 2
Compound 1(2.86g,14.6mmol) was dissolved in 20mL of DMF, and 5-bromo-6-chloronicotinic acid (3.45g,14.6mmol), HATU (6.66g,17.52mmol) and DIPEA (3.77g,29.2mmol) were added at room temperature, followed by stirring at room temperature for 2 hours. The reaction solution was poured into 100mL of water, extracted three times with 20mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 3/1) to obtain 3.65g of a white solid powder, which was the compound 2. The yield thereof was found to be 60.3%.1H NMR(500MHz,DMSO-d6)δ10.69(s,1H),8.91(d,J=2.2Hz,1H),8.77–8.69(m,1H),7.42–7.33(m,2H)。
And step 3: synthesis of Compound 3a
Compound 2(2.0g,4.83mmol), (R) -3-fluoropyrrolidine (0.52g,5.80mmol) and DIPEA (1.87g,14.49mmol) were dissolved in 30mL DMF and heated to 130 deg.C and stirred for 2 hours. The reaction solution was poured into 150mL of water, extracted three times with 40mL of ethyl acetate, and ethyl acetate was combinedThe ester was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 1.87g of a white solid powder as compound 3 a. The yield thereof was found to be 83.0%. LC-MS (APCI): M/z-466.0, 468.0(M +1)+
And 4, step 4: synthesis of Compound A-1a
Compound 3a (675mg,1.45mmol) was dissolved in 15mL dioxane, Pd (PPh) was added under nitrogen protection at room temperature3)4(50mg,0.043mmol)、K3PO4(921mg,4.34mmol) was stirred and dispersed uniformly, and pinacolborane (1.11g, 8.68mmol) was added slowly and stirred at 110 ℃ for 10 hours under nitrogen. Cooled to room temperature, filtered, the filter cake washed with dichloromethane, the filtrate concentrated and the brown oily liquid used directly in the next step. LC-MS (APCI): M/z 514.2(M +1)+
Dissolving the oily liquid in 10mL of ethylene glycol dimethyl ether and 2mL of water, and adding 2-bromopyrazine (276mg, 1.74mmol) and Pd (PPh) under the protection of nitrogen3)4(50mg,0.043mmol) and Na2CO3(460mg,4.34mmol), the reaction was stirred at 100 ℃ for 2 hours under nitrogen. Cooling to room temperature, pouring the reaction solution into 60mL of water, extracting with 20mL of ethyl acetate for three times, combining ethyl acetate, washing with saturated saline, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 65mg of white solid powder, namely the compound A-1 a. LC-MS (APCI): M/z 466.2(M +1)+1H NMR(300MHz,DMSO-d6)δ10.27(s,1H),8.87(d,J=19.0Hz,2H),8.70(d,J=29.7Hz,2H),8.24(s,1H),7.35(s,2H),5.29(d,J=53.3Hz,1H),3.66–3.11(m,5H),2.09(d,J=14.0Hz,1H).
4Example 5(R) -N- (4- (chlorodifluoromethoxy) phenyl) -6- (3-fluoropyrrolidin-1-yl-2, 2,5,5-d) -5- Preparation of (pyrazin-2-yl) nicotinamide (compound A-2 a).
Figure BDA0001964663670000291
The synthesis was carried out using the following route:
Figure BDA0001964663670000292
step 1: synthesis of Compound 4
D-malic acid (10.0g, 74.6mmol) and benzylamine (9.59g, 89.5mmol) were dispersed in 250mL xylene and heated for 8 hours to dehydrate. After cooling to room temperature, cooling and stirring for 2 hours in an ice bath, filtering, washing a filter cake with petroleum ether, taking the filter cake, and performing column chromatography (petroleum ether/ethyl acetate, 1/2) to obtain 9.91g of white solid powder, namely the compound 4. Yield: 64.8 percent. LC-MS (APCI): M/z 206.1(M +1)+
Step 2: synthesis of Compound 5
Taking LiAlD4(2.45g, 58.3mmol) in 70mL of anhydrous tetrahydrofuran, and a solution of Compound 4(3.0g, 14.6mmol) in tetrahydrofuran (70mL) was added dropwise slowly at-15 deg.C, and the mixture was stirred overnight at room temperature after dropping. Adding a small amount of sodium sulfate decahydrate for quenching, filtering the reaction solution, washing filter residues by using 100mL of ethyl acetate, concentrating the filtrate, and obtaining 1.26g of colorless oily liquid, namely the compound 5 which is directly used in the next step.
And step 3: synthesis of Compound 6
Compound 5(526mg, 2.9mmol) was dissolved in 10 mM LEOD and hydrogenated at 50 ℃ overnight. Filtration was carried out, and the residue was washed with 30mL of ethyl acetate, and the filtrate was concentrated to obtain 255mg of a brown oily liquid, which was Compound 6 and was used in the next step.
And 4, step 4: synthesis of Compound 7
The compound 4- (chlorodifluoromethoxy) aniline (2.86g,14.6mmol) was dissolved in 20mL DMF, and 5-bromo-6-chloronicotinic acid (3.45g,14.6mmol), HATU (6.66g,17.52mmol) and DIPEA (3.77g,29.2mmol) were added at room temperature and stirred at room temperature for 2 hours. The reaction solution was poured into 100mL of water, extracted three times with 20mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 3/1) to obtain 3.65g of a white solid powder. The yield thereof was found to be 60.3%.
And 5: synthesis of Compound 8
Compound 7(1.0g, 2.43mmol) and compound 6(266mg, 2.91mol) were dissolved in 5mL of DMF, DIPEA (628mg, 2.0mmol) was added at room temperature, and the reaction was stirred at 130 ℃ for 2 hours. The reaction solution was poured into 50mL of water, extracted three times with 10mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 1.03g of a white solid powder. The yield thereof was found to be 90.9%. LC-MS (APCI): M/z-466.0, 468.0(M +1)+
Step 6: synthesis of Compound 9a
Dissolving the compound 8(1.0g,2.14mmol) in a mixed solvent of 10mL of anhydrous dichloromethane and 5mL of anhydrous THF, slowly adding DAST dropwise at-78 ℃ under the protection of nitrogen, stirring at-78 ℃ for 2 hours after adding, and naturally heating to room temperature and stirring overnight. 10mL of saturated NaHCO was added3The reaction was quenched with shaking, the aqueous phase was extracted three times with 10mL of dichloromethane, the organic phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 3/1) to obtain 792mg of a brown oily liquid. Yield: 78.9 percent. LC-MS (APCI) M/z 468.0,470.0(M +1)+
And 7: synthesis of Compound A-2
Dissolve compound 9a (317mg,0.68mmol) in 10mL dioxane, add Pd (PPh) at room temperature under nitrogen protection3)4(24mg,0.020mmol)、K3PO4(431mg,2.03mmol) was stirred and dispersed uniformly, and pinacolborane (520mg, 4.06mmol) was added slowly and stirred at 110 ℃ for 10 hours under nitrogen. Cooled to room temperature, filtered, the filter cake washed with dichloromethane, the filtrate concentrated and the brown oily liquid used directly in the next step. LC-MS (APCI): M/z ═ 516.2, 518.2(M +1)+
Dissolving the oily liquid in 10mL of ethylene glycol dimethyl ether and 2mL of water, and adding 2-bromopyrazine (129mg, 0.81mmol) and Pd (PPh) under the protection of nitrogen3)4(24mg,0.020mmol) and Na2CO3(215mg,2.03mmol) and the reaction stirred at 100 ℃ for 2 hours under nitrogen. Cooled to room temperature, the reaction mixture was poured into 60mL of water, extracted three times with 20mL of ethyl acetate, and the ethyl acetate was combined and added with saturated brineWashing, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 36mg white solid powder as compound A-2. LC-MS (APCI) M/z 468.7(M +1)+1H NMR(300MHz,DMSO-d6)δ10.27(s,1H),8.87(d,J=19.0Hz,2H),8.70(d,J=29.7Hz,2H),8.24(s,1H),7.87(d,J=8.5Hz,2H),7.35(d,J=8.4Hz,2H),5.41(dd,J=53.1,2.9Hz,1H),2.32–1.96(m,2H).
2Example 6(R) -N- (4- (chlorodifluoromethoxy) phenyl-2, 6-d) -6- (3-fluoropyrrolidin-1-yl-2, 2,5, 4preparation of 5-d) -5- (pyrazin-2-yl) nicotinamide (compound A-3 a).
Figure BDA0001964663670000311
The synthesis was carried out using the following route:
Figure BDA0001964663670000312
step 1: synthesis of Compound 10
Compound 2(1.5g,3.62mmol), compound 6(0.38g,4.35mmol) and DIPEA (0.94g,7.25mmol) were dissolved in 20mL of DMF and stirred at 130 ℃ for 2 hours. The reaction solution was poured into 100mL of water, extracted three times with 20mL of ethyl acetate, the ethyl acetate was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 1.47g of a white solid powder. The yield thereof was found to be 86.6%. LC-MS (APCI): M/z-468.0, 470.0(M +1)+
Step 2: synthesis of Compound 11a
Dissolving compound 10(384mg,0.82mmol) in a mixed solvent of 10mL of anhydrous dichloromethane and 5mL of anhydrous THF, slowly adding DAST dropwise at-78 ℃ under the protection of nitrogen, stirring at-78 ℃ for 2 hours after adding, and naturally heating to room temperature and stirring overnight. 10mL of saturated NaHCO was added3The reaction was quenched with water, separated by shaking, the aqueous phase was extracted three times with 10mL of dichloromethane, the organic phases were combined and washed with saturated brineWashing, drying over anhydrous sodium sulfate, filtering, concentrating, and column chromatography (petroleum ether/ethyl acetate, 3/1) to obtain 215mg of brown oily liquid. Yield: 56.0 percent. LC-MS (APCI) M/z 470.0,472.0(M +1)+
And step 3: synthesis of Compound A-3a
Dissolve compound 11a (215mg,0.46mmol) in 10mL dioxane, add Pd (PPh) at room temperature under nitrogen protection3)4(16mg,0.014mmol)、K3PO4(291mg,1.37mmol) was stirred and dispersed uniformly, and pinacolborane (351mg, 2.74mmol) was added slowly and stirred at 110 ℃ for 10 hours under nitrogen. Cooled to room temperature, filtered, the filter cake washed with dichloromethane, the filtrate concentrated and the brown oily liquid used directly in the next step. LC-MS (APCI): M/z ═ 516.2, 518.2(M +1)+
Dissolving the oily liquid in 10mL of ethylene glycol dimethyl ether and 2mL of water, and adding 2-bromopyrazine (88mg, 0.55mmol) and Pd (PPh) under the protection of nitrogen3)4(16mg,0.014mmol) and Na2CO3(145mg,1.37mmol), the reaction was stirred at 100 ℃ for 2 hours under nitrogen. Cooling to room temperature, pouring the reaction solution into 60mL of water, extracting with 20mL of ethyl acetate for three times, combining ethyl acetate, washing with saturated saline, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography (petroleum ether/ethyl acetate, 1/1) to obtain 30mg of white solid powder, namely the compound A-3 a. LC-MS (APCI): M/z 470.5(M +1)+1H NMR(300MHz,DMSO-d6)δ10.27(s,1H),8.87(d,J=19.0Hz,2H),8.70(d,J=29.7Hz,2H),8.24(s,1H),7.35(t,J=1.1Hz,1H),5.56–5.09(m,1H),2.33–2.06(m,2H)。
And (4) testing the biological activity.
(1) Metabolic stability evaluation
Microsome experiment: human liver microsomes: 0.5mg/mL, Xenotech; rat liver microsomes: 0.5mg/mL, Xenotech; coenzyme (NADPH/NADH): 1mM, Sigma Life Science; magnesium chloride: 5mM, 100mM phosphate buffer (pH 7.4).
Preparing a stock solution: an amount of the compound of example was weighed out finely and dissolved in DMSO to 5mM each.
Preparation of phosphate buffer (100mM, pH 7.4): 150mL of 0.5M potassium dihydrogenphosphate and 700mL of 0.5M dipotassium hydrogenphosphate solution prepared in advance were mixed, the pH of the mixture was adjusted to 7.4 with the 0.5M dipotassium hydrogenphosphate solution, diluted 5-fold with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer solution (100mM) containing 100mM potassium phosphate and 3.3mM magnesium chloride at a pH of 7.4.
NADPH regenerating system solution (containing 6.5mM NADP, 16.5mM G-6-P, 3U/mL G-6-P D, 3.3mM magnesium chloride) was prepared and placed on wet ice before use.
Preparing a stop solution: acetonitrile solution containing 50ng/mL propranolol hydrochloride and 200ng/mL tolbutamide (internal standard). 25057.5 mu L of phosphate buffer solution (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of human liver microsome is respectively added and mixed evenly, and liver microsome dilution liquid with the protein concentration of 0.625mg/mL is obtained. 25057.5 mu L of phosphate buffer (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of SD rat liver microsome is respectively added, and the mixture is mixed evenly to obtain liver microsome dilution with the protein concentration of 0.625 mg/mL.
Incubation of the samples: the stock solutions of the corresponding compounds were diluted to 0.25mM each with an aqueous solution containing 70% acetonitrile, and used as working solutions. 398. mu.L of human liver microsome or rat liver microsome dilutions were added to a 96-well plate (N-2), and 2. mu.L of 0.25mM working solution, respectively, and mixed well.
Determination of metabolic stability: 300. mu.L of pre-cooled stop solution was added to each well of a 96-well deep-well plate and placed on ice as a stop plate. The 96-well incubation plate and the NADPH regeneration system are placed in a 37 ℃ water bath box, shaken at 100 rpm and pre-incubated for 5 min. 80. mu.L of the incubation solution was taken out of each well of the incubation plate, added to the stop plate, mixed well, and supplemented with 20. mu.L of NADPH regenerating system solution as a 0min sample. Then 80. mu.L of NADPH regenerating system solution was added to each well of the incubation plate, the reaction was started, and the timer was started. The reaction concentration of the corresponding compound was 1. mu.M, and the protein concentration was 0.5 mg/mL. When the reaction was carried out for 10min, 30 min and 90min, 100. mu.L of each reaction solution was added to the stop plate and vortexed for 3min to terminate the reaction. The stop plates were centrifuged at 5000 Xg for 10min at 4 ℃. And (3) taking 100 mu L of supernatant to a 96-well plate in which 100 mu L of distilled water is added in advance, mixing uniformly, and performing sample analysis by adopting LC-MS/MS.
And (3) data analysis: and detecting peak areas of the corresponding compound and the internal standard through an LC-MS/MS system, and calculating the peak area ratio of the compound to the internal standard. The slope is determined by plotting the natural logarithm of the percentage of compound remaining against time and calculating t according to the following formula1/2And CLintWhere V/M is equal to 1/protein concentration.
Figure BDA0001964663670000331
The compounds of the invention and compounds without deuteration were tested simultaneously and compared to evaluate their metabolic stability in human and rat liver microsomes. The compound of the invention can obviously improve the metabolic stability.
(2) Pharmacokinetic experiment of rat
6 male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 per group and compared for pharmacokinetic differences by intravenous or oral administration of a single dose of compound (10 mg/kg oral).
Rats were fed with standard feed and given water. Fasting began 16 hours prior to the experiment. The drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the orbit at 0.083 hr, 0.25 hr, 0.5 hr, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post-dose.
The rats were briefly anesthetized after ether inhalation and 300 μ L of blood was collected from the orbit into a test tube. In the test tube there was 30. mu.L of 1% heparin salt solution. Before use, the tubes were dried overnight at 60 ℃. After completion of blood collection at the last time point, rats were sacrificed after ether anesthesia.
Immediately after blood collection, the tubes were gently inverted at least 5 times to ensure mixing and then placed on ice. The blood samples were centrifuged at 5000rpm for 5 minutes at 4 ℃ to separate the plasma from the erythrocytes. Pipette 100 μ L of plasma into a clean plastic centrifuge tube, designating the name of the compound and the time point. Plasma was stored at-80 ℃ before analysis. The concentration of the compounds of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.
Experiments show that the compound has better pharmacokinetic property in animals, thereby having better pharmacodynamics and treatment effects.
The foregoing is a more detailed description of the invention in connection with specific preferred embodiments and it is not intended that the invention be limited to these specific details. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.

Claims (13)

1. A compound of formula (II):
Figure FDA0002968707050000011
wherein the content of the first and second substances,
y is selected from
Figure FDA0002968707050000012
When Y is selected from
Figure FDA0002968707050000013
When X is selected from-OH;
when Y is selected from
Figure FDA0002968707050000014
When X is selected from-F;
R5、R6、R7、R8、R9、R8’、R9’、R10’、R10、R11、R14and R15Is hydrogen;
R1、R2、R3、R4、R12and R13Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, having formula (IIIa):
Figure FDA0002968707050000021
wherein the content of the first and second substances,
x is selected from-OH;
R5、R6、R7、R14and R15Is hydrogen;
R1、R2、R3、R4、R12and R13Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
or a pharmaceutically acceptable salt thereof.
3. The compound of claim 2, wherein R12And R13Is deuterium.
4. A compound according to claim 2 or 3, wherein R1、R2、R3And R4Is deuterium.
5. The compound of claim 1, having formula (IVa):
Figure FDA0002968707050000022
wherein the content of the first and second substances,
x is selected from-F;
R5、R6、R7、R14and R15Is hydrogen;
R1、R2、R3、R4、R12and R13Each independently selected from hydrogen or deuterium;
with the proviso that said compound contains at least one deuterium atom;
or a pharmaceutically acceptable salt thereof.
6. The compound of claim 5, wherein R12And R13Is deuterium.
7. A compound according to claim 5 or 6, wherein R1、R2、R3And R4Is deuterium.
8. The compound of claim 1, wherein the compound is selected from the following compounds, or pharmaceutically acceptable salts thereof:
Figure FDA0002968707050000031
9. a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof.
10. The pharmaceutical composition of claim 9, further comprising an additional therapeutic agent.
11. The pharmaceutical composition of claim 10, wherein the additional therapeutic agent is a different inhibitor of Bcr-Abl 1.
12. Use of a compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 9-11, in the manufacture of a medicament for treating a condition mediated by Bcr-Abl 1.
13. The use according to claim 12, wherein the condition is selected from acute lymphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, gastrointestinal stromal tumors, thyroid cancer, gastric cancer, rectal cancer, multiple myeloma, viral infection or CNS disorders.
CN201910096790.0A 2018-02-07 2019-01-31 Substituted nicotinamide compound, pharmaceutical composition and application thereof Active CN109651359B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2018101222074 2018-02-07
CN2018101215719 2018-02-07
CN201810122207 2018-02-07
CN201810121571 2018-02-07

Publications (2)

Publication Number Publication Date
CN109651359A CN109651359A (en) 2019-04-19
CN109651359B true CN109651359B (en) 2021-06-22

Family

ID=66122018

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910096790.0A Active CN109651359B (en) 2018-02-07 2019-01-31 Substituted nicotinamide compound, pharmaceutical composition and application thereof

Country Status (2)

Country Link
CN (1) CN109651359B (en)
WO (1) WO2019154252A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021213380A1 (en) * 2020-04-20 2021-10-28 深圳市塔吉瑞生物医药有限公司 Solid form of pyrazine substituted nicotinamide, and preparation and use thereof
TW202306567A (en) * 2021-05-28 2023-02-16 大陸商正大天晴藥業集團股份有限公司 Compound used as BCR-ABL inhibitor
CN114149409B (en) * 2021-11-16 2024-03-22 中国药科大学 (hetero) aryl amide compound with protein kinase inhibition activity
CN116135851A (en) * 2021-11-17 2023-05-19 武汉众诚康健生物医药科技有限公司 Aromatic amine compound and application thereof
CN114369085B (en) * 2021-12-27 2023-01-03 武汉九州钰民医药科技有限公司 Preparation method of Asciminib hydrochloride
CN115109048B (en) * 2022-08-10 2023-12-08 中国药科大学 (hetero) aryl amide compound

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104302634A (en) * 2012-05-15 2015-01-21 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
CN104379574A (en) * 2012-05-15 2015-02-25 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of ABL1, ABL 2 and BCR-ABL 1
WO2018133826A1 (en) * 2017-01-20 2018-07-26 深圳市塔吉瑞生物医药有限公司 (hetero)arylamide compound for inhibiting protein kinase activity
WO2018133827A1 (en) * 2017-01-20 2018-07-26 深圳市塔吉瑞生物医药有限公司 (hetero)arylamide compound for inhibiting protein kinase activity

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104302634A (en) * 2012-05-15 2015-01-21 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of ABL1, ABL2 and BCR-ABL1
CN104379574A (en) * 2012-05-15 2015-02-25 诺华股份有限公司 Benzamide derivatives for inhibiting the activity of ABL1, ABL 2 and BCR-ABL 1
WO2018133826A1 (en) * 2017-01-20 2018-07-26 深圳市塔吉瑞生物医药有限公司 (hetero)arylamide compound for inhibiting protein kinase activity
WO2018133827A1 (en) * 2017-01-20 2018-07-26 深圳市塔吉瑞生物医药有限公司 (hetero)arylamide compound for inhibiting protein kinase activity

Also Published As

Publication number Publication date
CN109651359A (en) 2019-04-19
WO2019154252A1 (en) 2019-08-15

Similar Documents

Publication Publication Date Title
CN109651359B (en) Substituted nicotinamide compound, pharmaceutical composition and application thereof
AU2018386298B2 (en) Inhibitors of fibroblast activation protein
AU2013230146B2 (en) 2-amino, 6-phenyl substituted pyrido [2, 3 - d] pyrimidine derivatives useful as Raf kinase inhibitors
US9809610B2 (en) Compounds and compositions as kinase inhibitors
AU2022203071A1 (en) Amino pyrimidine SSAO inhibitors
WO2019144835A1 (en) Substituted pyridazinone compound
JP2024037954A (en) RIP1 inhibitory compounds and methods for making and using the same
CN110092745B (en) Compound containing aromatic ring and application thereof
TW202229274A (en) Novel salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
CN104302292A (en) Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activities
EP3878852A1 (en) Substituted pyrazolo[1,5-a]pyridine compound, composition containing the same and use thereof
CN110759908A (en) N-benzenesulfonyl benzamide compound for inhibiting Bcl-2 protein and composition and application thereof
JP2015180679A (en) Crystalline forms of sodium 4-{[9-chloro-7-(2-fluoro-6-methoxyphenyl)-5h-pyrimido[5,4-d][2]benzazepin-2-yl]amino}-2-methoxybenzoate
KR20200096261A (en) Sulfonamide compounds and uses thereof
WO2021098850A1 (en) Crystal form of nucleoprotein inhibitor and use thereof
TWI771280B (en) Solid forms of substituted 5,6-dihydro-6-phenylbenzo[f]isoquinolin-2-amine compounds
CN109535164A (en) Jak kinase inhibitor and preparation method thereof and application in field of medicaments
KR20140040671A (en) Aprepitant l-proline composition and cocrystal
CA3028824A1 (en) Substituted pyrrolo[2,3-d]pyridazin-4-ones and pyrazolo[3,4-d]pyridazin-4-ones as protein kinase inhibitors
CN112457296A (en) Pyrimidine compound and preparation method thereof
WO2019241496A1 (en) Peptidomimetic inhibitors of the peptidyl-prolyl cis/trans isomerase (pin1)
WO2019096112A1 (en) Substituted benzimidazole compound and composition comprising same
TWI828489B (en) Pyrimidine-2(1H)-ketobicyclic compounds with MAT2A inhibitory activity and their uses
WO2024067744A1 (en) Heterocyclic substituted quinazoline, preparation method therefor, and use thereof
US20240002365A1 (en) Pyridazine and 1,2,4-triazine derivatives as fgfr kinase inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant