CN115490640A - Substituted benzimidazoles, compositions containing them and their use - Google Patents

Substituted benzimidazoles, compositions containing them and their use Download PDF

Info

Publication number
CN115490640A
CN115490640A CN202210665562.2A CN202210665562A CN115490640A CN 115490640 A CN115490640 A CN 115490640A CN 202210665562 A CN202210665562 A CN 202210665562A CN 115490640 A CN115490640 A CN 115490640A
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
solvate
deuterium
hydrate
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.)
Pending
Application number
CN202210665562.2A
Other languages
Chinese (zh)
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 CN115490640A publication Critical patent/CN115490640A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D235/10Radicals substituted by halogen atoms or nitro radicals
    • 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 benzimidazole compound, a composition containing the compound and application of the compound, wherein the substituted benzimidazole compound is a compound shown as a formula (I) or a tautomer, a stereoisomer, a prodrug, a crystal form, pharmaceutically acceptable salt, hydrate or a solvate of the compound. The compounds of formula (I) are potent MEK1/2 inhibitors that inhibit ERK phosphorylation by inhibiting activated MEK, exhibit broad antitumor activity in a variety of cancer models, and, at the same time, are better treatable in combination with other antitumor therapeuticsTreating various cancers. In addition to inhibition and potency, the compounds of the invention also exhibit better metabolic stability and/or pharmacokinetic properties.

Description

Substituted benzimidazoles, compositions containing them and their use
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a substituted benzimidazole compound, a composition containing the compound and application of the compound. More particularly, the present invention relates to certain deuterium substituted 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-N- (2-hydroxyethoxy) -1-methyl-1H-benzimidazole-6-carboxamide compounds and derivatives thereof, and tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates, or solvate compounds thereof. These deuterium substituted compounds and compositions thereof are useful as potent and selective inhibitors of MEK1 and MEK2 proteins and for the treatment of diseases caused by MEK kinase, and have superior ADME and pharmacokinetic properties.
Background
The RAS/RAF/MEK/ERK kinase pathway is activated in more than 30% of human cancers. Activation of the RAS GTPase (GTPase) protein in response to growth factors, hormones, cytokines, etc. stimulates phosphorylation and activation of RAF kinases. These kinases then phosphorylate and activate the intracellular protein kinases MEK1 and MEK2, which subsequently phosphorylate and activate the other protein kinases ERK1 and 2. This signaling pathway, also known as the mitogen-activated protein kinase (MAPK) pathway or cytoplasmic cascade, mediates cellular responses to growth signals. The underlying function of this pathway is to link receptor activity at the cell membrane with modifications of cytoplasmic or nuclear targets that control cell proliferation, differentiation and survival.
Structural activation of this pathway is sufficient to induce cellular transformation. Deregulated activation of MAPK pathways due to aberrant receptor tyrosine kinase activation, RAS mutations or RAF mutations is commonly found in human cancers and represents a major factor in determining aberrant growth control. RAS mutations are common in human malignancies and have been identified in about 30% of cancers. The RAS family of gtpase proteins (proteins that convert guanosine triphosphate to guanosine diphosphate) allows signals to be transmitted from activated growth factor receptors to downstream intracellular counterparts. Among the targets complemented by active membrane-bound RAS, the important targets are the RAF family of serine/threonine protein kinases. The RAF family consists of three related kinases (A-, B-, and C-RAF), which act as downstream effectors of the RAS. RAS-mediated RAF activation also triggers activation of MEK1 and MEK2, followed by phosphorylation of ERK1 and ERK2 (extracellular signal-regulated kinases 1 and 2) on tyrosine-185 and threonine-183. Activated ERK1 and ERK2 change positions and accumulate in the nucleus where they can phosphorylate various substrates, including transcription factors that control cell growth and survival. Given the importance of the MAPK pathway in the development of human cancer, in cancer and other proliferative diseases, the kinase components of the signaling cascade are incorporated as potentially important targets for modulating disease progression.
MEK1 and MKE2 are members of a larger family of bispecific kinases that phosphorylate the threonine and tyrosine disabilities of various MAPKs. MEK1 and MEK2 have unique gene codes, but they share a high degree of homology (80%) in the C-terminal catalytic kinase domain and most of the N-terminal regulatory region. The oncogenic forms of MEK1 and MEK2 have not been found in human cancers, but it has been shown that structural activation of MEK leads to cellular transformation. MEK can also be activated by other oncogenes in addition to RAF. To date, the only known substrates for MEK1 and MEK2 are ERK1 and ERK2. In addition to the unique ability to phosphorylate tyrosine and threonine residues, this abnormal substrate specificity places MEK1 and MEK2 at a critical point in the signaling cascade, which allows it to integrate many extracellular signals into the MAPK pathway.
The fundamental role and position of RAF in many signaling cascades has been demonstrated from studies using mammalian cells to regulate and significantly inhibit RAF mutations and studies using biochemical and genetic techniques on model organisms. RAF may have a prominent role in the development of certain tumors, for example, the activating allele of BRAF has been identified in-70% melanoma, 40% thyroid papillary carcinoma, 30% low grade ovarian carcinoma and 10% colorectal carcinoma. Most BRAF mutations are found in the kinase domain, with monosubstitution (V600E) accounting for at least 80%. Mutant BRAF proteins activate the RAS/RAF/MEK/ERK kinase pathway either by MEK-elevated kinase activity or by activating C-RAF.
Binimetinib (chemical name 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-N- (2-hydroxyethoxy) -1-methyl-1H-benzimidazole-6-carboxamide, which has the following structural formula) is a potent, non-ATP competitive, highly selective MEK1/2 inhibitor developed by Array BioPharma that inhibits MEK, ERK phosphorylation and growth of BRAF or KRAS mutant cancer cells at nanomolar concentrations. The U.S. Food and Drug Administration (FDA) awards a Binimetinib orphan drug status in 11 months 2013 and approves it in combination with the BRAF inhibitor Encorafenib for the treatment of metastatic or unresectable melanoma patients with BRAF V600E/K mutations in 6 months 2018. It has now been demonstrated that the use of BRAF inhibitors in combination with MEK inhibitors can improve the therapeutic efficacy and possibly reduce the toxic effects. BRAF inhibitor/MEK inhibitor combination immunotherapy is recommended as a first-line therapy for metastatic or unresectable melanoma in the National Comprehensive Cancer Network (NCCN) guidelines.
Figure BDA0003692806340000021
Figure BDA0003692806340000031
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.
It remains a challenging task to find new potent and highly selective MEK1/2 inhibitors with good oral bioavailability and pharmaceutical properties. Accordingly, there remains a need in the art to develop compounds having selective inhibitory activity and/or better pharmacodynamics/pharmacokinetics for use as MEK1/2 inhibitors, and the present invention provides such compounds.
Disclosure of Invention
In view of the above technical problems, the present invention discloses a novel deuterium-substituted benzimidazole compound as an effective MEK1/2 inhibitor, which can inhibit ERK phosphorylation by inhibiting activated MEK, and exhibit a wide range of antitumor activities against various cancer models, including melanoma, acute myeloid leukemia, glioma, neurofibroma, non-small cell lung cancer, breast cancer, serous cancer, gastrointestinal stromal tumor, lung non-squamous carcinoma, colorectal cancer, biliary tract cancer, myeloma, and the like. At the same time, the compounds of the present invention are used in combination with other anti-tumor therapeutic agents to treat a variety of cancers. In addition to the inhibitory effect and potency, the compounds of the invention also show good solubility and better metabolic stability and/or pharmacokinetic properties.
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 BDA0003692806340000032
wherein the content of the first and second substances,
Y 1 、Y 2 、Y 3 、Y 4 and Y 5 Each independently selected from hydrogen, deuterium or halogen;
R 1 、R 2 、R 3 and R 4 Each independently selected from hydrogen or deuterium;
each X is independently selected from CH 3 、CD 3 、CHD 2 Or CH 2 D;
With the proviso that said compound contains at least one deuterium atom;
or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention or a tautomer thereof,A pharmaceutical composition of a stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate compound 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 pharmaceutical composition further comprises an additional therapeutic agent. In particular embodiments, the additional therapeutic agent is selected from one or more of a BRAF inhibitor, an EGFR antibody, an immune checkpoint inhibitor or a CDK4/6 inhibitor. In specific embodiments, the BRAF inhibitor is selected from vemurafenib (vemurafenib), dabrafenib (dabrafenib), canrafenib (encorafenib), (S) -methyl- (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (prop-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate, (S) - (methyl-d 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1-isopropyl-1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate, (S) - (methyl-d 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate, (S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5 ) Methyl carbamate, (S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5) carbamic acid methyl ester. In particular embodiments, the EGFR inhibitor is selected from gefitinib (gefitinib), erlotinib (erlotinib), afatinib (afatinib), dacomitinib (dacomitinib), lapatinib (lapatinib), oxitinib (osimertinib), amitinib, fomitinib, CO-1686, wz4002, pd153035, pf00299804. In particular embodiments, the EGFR antibody is selected from cetuximab (cetuximab), panitumumab (panitumumab), and rituximab ((Necitumumab)brilizumab), yipriuma (ipilimumab), and nivolumab (nivolumab), atelizumab (atezolizumab), avizumab (avelumab), devolumab (durvalumab), pilizumab (pidilzumab). In particular embodiments, the CDK4/6 inhibitor is selected from pabociclib (palbociclib), ribociclib (ribociclib), abeciclib (abemacciclib).
In another aspect, the present invention provides a method for preparing the pharmaceutical composition as described above, comprising the steps of: a pharmaceutically acceptable excipient is mixed with a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, to form a pharmaceutical composition.
In another aspect, the present invention further provides a method of treating a MEK kinase mediated disease, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In particular embodiments, the MEK kinase mediated disease is melanoma, acute myeloid leukemia, glioma, neurofibroma, non-small cell lung cancer, breast cancer, serous cancer, gastrointestinal stromal tumor, lung non-squamous carcinoma, colorectal cancer, biliary tract cancer, myeloma. In specific embodiments, the melanoma is selected from BRAF V600 mutant melanoma. In a specific embodiment, the colorectal cancer is selected from BRAF V600 mutated colorectal cancer. In specific embodiments, the BRAF V600 mutation is selected from a BRAF V600E mutation or a BRAF V600K mutation. In specific embodiments, the neurofibroma is selected from neurofibromatosis type 1 (NF 1) or plexiform neurofibromas;
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.
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%).
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 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 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)
Herein, "compound of the present invention" refers to the following compounds of formula (I) and formula (II) or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate compounds thereof.
In one embodiment, the invention relates to compounds of formula (I):
Figure BDA0003692806340000061
Figure BDA0003692806340000071
wherein the content of the first and second substances,
Y 1 、Y 2 、Y 3 、Y 4 and Y 5 Each independently selected from hydrogen, deuterium or halogen;
R 1 、R 2 、R 3 and R 4 Each independently selected from hydrogen or deuterium;
x is selected from CH 3 、CD 3 、CHD 2 Or CH 2 D;
With the proviso that said compound contains at least one deuterium atom;
or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof.
In a particular 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, Y in the present invention 1 、Y 2 、Y 3 、Y 4 、Y 5 、R 1 、R 2 、R 3 、R 4 And the deuterium isotope content of each deuterium position of X is at least 0.015% greater than the natural isotope content, preferably greater than 1%, more preferably greater than 5%, more 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, the compounds of the present invention contain at least one deuterium atom, 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.
In another embodiment, "Y" is 1 、Y 2 、Y 3 、Y 4 And Y 5 Each independently selected from hydrogen, deuterium or halogen "including Y 1 Selected from hydrogen, deuterium or halogen, Y 2 Selected from hydrogen, deuterium or halogen, Y 3 Selected from hydrogen, deuterium or halogen, and so on, up to Y 5 Selected from hydrogen, deuterium or halogen. More specifically, including Y 1 Is hydrogen, Y 1 Is deuterium or Y 1 Is halogen (F, cl, br or I), Y 2 Is hydrogen, Y 2 Is deuterium or Y 2 Is halogen (F, cl, br or I), Y 3 Is hydrogen, Y 3 Is deuterium or Y 3 Is halogen (F, cl, br or I), and so on, up to Y 5 Is hydrogen, Y 5 Is deuterium or Y 5 Is a technical proposal of halogen (F, cl, br or I).
In another embodiment, "R" is 1 、R 2 、R 3 And R 4 Each independently selected from hydrogen or deuterium "comprising R 1 Selected from hydrogen or deuterium, R 2 Selected from hydrogen or deuterium, R 3 Selected from hydrogen or deuterium, and R 4 Selected from hydrogen or deuterium. More specifically, includes R 1 Is hydrogen or R 1 Is deuterium, R 2 Is hydrogen or R 2 Is deuterium, R 3 Is hydrogen or R 3 Is deuterium, and R 4 Is hydrogen or R 4 Is a technical scheme of deuterium.
In another embodiment, "X is selected from CH 3 、CD 3 、CHD 2 Or CH 2 D' includes X is CH 3 X is CD 3 X is CHD 2 Or X is CH 2 And D, technical scheme.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates thereofA compound of a solvent or a compound of a solvent, wherein X is CD 3 ,Y 1 、Y 2 、Y 3 、Y 4 、Y 5 、R 1 、R 2 、R 3 And R 4 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein R is 1 And R 2 Is deuterium, Y 1 、Y 2 、Y 3 、Y 4 、Y 5 、R 3 、R 4 And X is as defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein R is 3 And R 4 Is deuterium, Y 1 、Y 2 、Y 3 、Y 4 、Y 5 、R 1 、R 2 And X is as defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein R is 1 、R 2 、R 3 And R 4 Is deuterium, Y 1 、Y 2 、Y 3 、Y 4 、Y 5 And X is as defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compound, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 Is deuterium, Y 2 、Y 3 、Y 4 、Y 5 、R 1 、R 2 、R 3 、R 4 And X is as defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms thereofA pharmaceutically acceptable salt, hydrate or solvate thereof, wherein X is CD 3 ,Y 1 Is deuterium, Y 2 、Y 3 、Y 4 、Y 5 、R 1 、R 2 、R 3 And R 4 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein X is CD 3 ,R 1 And R 2 Is deuterium, Y 1 、Y 2 、Y 3 、Y 4 、Y 5 、R 3 And R 4 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,R 3 And R 4 Is deuterium, Y 1 、Y 2 、Y 3 、Y 4 、Y 5 、R 1 And R 2 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein X is CD 3 ,R 1 、R 2 、R 3 And R 4 Is deuterium, Y 1 、Y 2 、Y 3 、Y 4 And Y 5 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 、R 1 And R 2 Is deuterium, Y 2 、Y 3 、Y 4 、Y 5 、R 3 、R 4 And X is as defined above.
In some embodiments of formula (I), preference is given toThe invention relates to the compound, or tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate or solvent compound thereof, wherein Y is 1 、R 3 And R 4 Is deuterium, Y 2 、Y 3 、Y 4 、Y 5 、R 1 、R 2 And X is as defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 、R 1 、R 2 、R 3 And R 4 Is deuterium, Y 2 、Y 3 、Y 4 、Y 5 And X is as defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,Y 1 、R 1 And R 2 Is deuterium, Y 2 、Y 3 、Y 4 、Y 5 、R 3 And R 4 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,Y 1 、R 3 And R 4 Is deuterium, Y 2 、Y 3 、Y 4 、Y 5 、R 1 And R 2 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein X is CD 3 ,Y 1 、R 1 、R 2 、R 3 And R 4 Is deuterium, Y 2 、Y 3 、Y 4 And Y 5 As defined above.
In another embodiment, the invention relates to compounds of formula (II):
Figure BDA0003692806340000101
wherein, the first and the second end of the pipe are connected with each other,
Y 1 selected from hydrogen, deuterium or halogen;
R 1 、R 2 、R 3 and R 4 Each independently selected from hydrogen or deuterium;
x is selected from CH 3 、CD 3 、CHD 2 Or CH 2 D;
With the proviso that said compound contains at least one deuterium atom;
or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,Y 1 、R 1 、R 2 、R 3 And R 4 As defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein R is 1 And R 2 Is deuterium, Y 1 、R 3 、R 4 And X is as defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein R is 3 And R 4 Is deuterium, Y 1 、R 1 、R 2 And X is as defined above.
In some embodiments of formula (II), preference is given toThe invention also relates to the compound, or tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate or solvent compound thereof, wherein R is 1 、R 2 、R 3 And R 4 Is deuterium, Y 1 And X is as defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compound, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 Is deuterium, R 1 、R 2 、R 3 、R 4 And X is as defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein X is CD 3 ,Y 1 Is deuterium, R 1 、R 2 、R 3 And R 4 As defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,R 1 And R 2 Is deuterium, Y 1 、R 3 、R 4 And Y 5 As defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,R 3 And R 4 Is deuterium, Y 1 、R 1 And R 2 As defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,R 1 、R 2 、R 3 And R 4 Is deuterium, Y 1 As defined above.
In some embodiments of formula (I), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 、R 1 And R 2 Is deuterium, R 3 、R 4 And X is as defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compound, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 、R 3 And R 4 Is deuterium, R 1 、R 2 And X is as defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compound, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 、R 1 、R 2 、R 3 And R 4 Is deuterium and X is as defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvate thereof, wherein X is CD 3 ,Y 1 、R 1 And R 2 Is deuterium, R 3 And R 4 As defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein X is CD 3 ,Y 1 、R 3 And R 4 Is deuterium, R 1 And R 2 As defined above.
In some embodiments of formula (II), preferably, the present invention relates to the above compounds, or tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically thereofAn acceptable salt, hydrate or solvate compound, wherein X is CD 3 ,Y 1 、R 1 、R 2 、R 3 And R 4 Is deuterium.
As a preferred embodiment of the present invention, the compound, or tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate or solvate thereof, is selected from any one of the following compounds:
Figure BDA0003692806340000121
Figure BDA0003692806340000131
Figure BDA0003692806340000141
Figure BDA0003692806340000151
Figure BDA0003692806340000161
the compounds of the invention may include one or more asymmetric centers and may therefore exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be in the form of 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 chiral salt formation and crystallization; alternatively, preferred isomers may be prepared by asymmetric synthesis.
One skilled in the art will appreciate that the organic compound may form a complex with a solvent in which it reacts or from which it precipitates or crystallizes. These complexes are referred to as "solvates". When the solvent is water, the complex is referred to as a "hydrate". The present invention encompasses all solvates of the compounds of the present invention.
The term "solvate" refers to a form of a compound or salt thereof that is combined with a solvent, typically formed by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, and the like. The compounds described herein can be prepared, for example, in crystalline form, and can be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates. In some cases, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. "solvate" includes a solvate in a solution state and a solvate which can be isolated. Representative solvates include hydrates, ethanolates, and methanolates.
The term "hydrate" refers to a compound that is associated with an aqueous phase. In general, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, hydrates of the compounds can be used, for example, with the formula R.xH 2 O represents, wherein R is the compound, and x is a number greater than 0. A given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrates (x is a number greater than 0 and less than 1), e.g., hemihydrate (R0.5H) 2 O)) and polyhydrates (x is a number greater than 1, e.g. dihydrate (R.2H) 2 O) and hexahydrate (R.6H) 2 O))。
The compounds of the invention may be in amorphous or crystalline form (polymorphs). 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 "polymorph" refers to a crystalline form of a compound (or a salt, hydrate, or solvate thereof) in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors may cause a crystalline form to dominate. Various polymorphs of a compound may be prepared by crystallization under different conditions.
The invention also includes isotopically-labeled compounds, which are identical to those recited in the present invention, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H、 3 H、 13 C、 11 C、 14 C、 15 N、 18 O、 17 O、 31 P、 32 P、 35 S、 18 F and 36 and (4) Cl. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labelled compounds of the invention, e.g. by incorporation of radioactive isotopes (e.g. by introducing 3 H and 14 c) Can be used in drug and/or substrate tissue distribution assays. Tritium, i.e. 3 H and carbon-14, i.e. 14 C isotopes are particularly preferred because of their ease of preparation and detection. Further, by heavier isotopes, e.g. deuterium, i.e. 2 H, may be preferred in some cases because higher metabolic stability may provide therapeutic benefits, such as increased in vivo half-life or reduced dosage requirements. Isotopically labeled compounds of formula (I) and prodrugs thereof of the present invention can generally be prepared by carrying out the procedures disclosed in the schemes and/or in the examples and preparations below, by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
In addition, prodrugs are also included within the context of the present invention. The term "prodrug" as used herein 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.bra "Improved oral Delivery: the solubility limits of the overcom by the use of the drugs ", 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 are readily broken down in the human body to release the parent acid or salt thereof.
Process for preparing the Compounds of the invention
The compounds of the invention (including salts thereof) may be prepared using known organic synthesis techniques and may be synthesized according to any of a number of possible synthetic routes, such as those in the schemes below. The reaction for preparing the compounds of the present invention may be carried out in a suitable solvent, which may be readily selected by one skilled in the art of organic synthesis. Suitable solvents may be substantially unreactive with the starting materials (reactants), intermediates, or products at the temperatures at which the reaction is carried out (e.g., temperatures in the range of solvent freezing temperatures to solvent boiling temperatures). A given reaction may be carried out in one solvent or a mixture of more than one solvent. The skilled person can select the solvent for a particular reaction step depending on the particular reaction step.
The preparation of the compounds of the invention may involve the protection and deprotection of different chemical groups. One skilled in the art can readily determine whether protection and deprotection is required and the choice of an appropriate protecting group. The chemistry of protecting Groups can be found, for example, in Wuts and Greene, protective Groups in Organic Synthesis, 4 th edition, john Wiley & Sons: new Jersey, (2006), which is incorporated herein by reference in its entirety.
The compounds of the present invention can be prepared as their individual stereoisomers by reacting a racemic mixture of the compounds with an optically active resolving agent to form a pair of diastereomeric compounds, separating the diastereomers and recovering the optically pure enantiomers. Enantiomeric resolution can be carried out using diastereomeric derivatives of the compounds of the invention, preferentially dissociable complexes (e.g., crystalline diastereomeric salts). Diastereomers have significantly different physical properties (e.g., melting points, boiling points, solubilities, reactivities, etc.) and can be readily separated by virtue of these dissimilarities. Diastereomers can be separated by chromatography, preferably by separation/resolution techniques based on differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that does not racemize. A more detailed description of techniques suitable for resolving stereoisomers of compounds starting from racemic mixtures can be found in Jean Jacques, andre Collet, samue1 h.wilen, "Enantiomers, racemates And Resolutions" ("Enantiomers, racemics And Resolutions"), john Wiley And Sons, inc.,1981.
The reaction may be monitored by any suitable method known in the art. For example, it can be determined by spectroscopic means, such as Nuclear Magnetic Resonance (NMR) spectroscopy (e.g. 1 H or 13 C) Infrared (IR) spectroscopy and spectroscopyProduct formation is monitored photometrically (e.g., UV-visible light), mass Spectrometry (MS), or by chromatographic methods such as High Performance Liquid Chromatography (HPLC) or Thin Layer Chromatography (TLC).
Pharmaceutical compositions, formulations and kits
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 a compound of the invention. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a compound of the invention. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of a compound of the present invention.
Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds formulated therewith. 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 invention also includes kits (e.g., pharmaceutical packages). Kits provided can include a compound of the invention, an additional therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes and/or dispensable packages or other suitable containers) containing the compound of the invention, the additional therapeutic agent. In some embodiments, provided kits may also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent. In some embodiments, the compound of the present invention and the additional therapeutic agent provided in the first container and the second container are combined to form one unit dosage form.
The following formulation examples illustrate representative pharmaceutical compositions that can be prepared according to the present invention. However, the present invention is not limited to the following pharmaceutical compositions.
Exemplary formulation 1-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 0.3-30mg tablets (each containing 0.1-10mg of active compound).
Exemplary formulation 2-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 30-90mg tablets (each tablet containing 10-30mg of active compound).
Exemplary formulation 3-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to form 90-150mg tablets (each containing 30-50mg of active compound).
Exemplary formulation 4-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 150-240mg tablets (each containing 50-80mg of active compound).
Exemplary formulation 5-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to 240-270mg tablets (each containing 80-90mg of active compound).
Exemplary formulation 6-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press to form 270-450mg tablets (each containing 90-150mg of active compound).
Exemplary formulation 7-tablet: the compound of the invention in dry powder form may be mixed with the dry gel binder in a weight ratio of about 1. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped in a tablet press into 450-900mg tablets (each tablet containing 150-300mg of active compound).
Exemplary formulation 8-capsule: the compound of the invention in dry powder form may be mixed with a starch diluent in a weight ratio of about 1. The mixture is filled into 250mg capsules (each containing 125mg of active compound).
Exemplary formulation 9-liquid: the compound of the present invention (125 mg) can be mixed with sucrose (1.75 g) and xanthan gum (4 mg), and the resulting mixture can be blended, passed through a No.10 mesh U.S. sieve, and then mixed with a previously prepared aqueous solution of microcrystalline cellulose and sodium carboxymethylcellulose (11, 89, 50 mg). Sodium benzoate (10 mg), flavouring and colouring agents were diluted with water and added with stirring. Sufficient water may then be added to give a total volume of 5 mL.
Exemplary formulation 10-injection: the compounds of the present invention may be dissolved or suspended in aqueous media, which may be injected in buffered sterile saline, to a concentration of about 5mg/mL.
Administration of drugs
The pharmaceutical compositions provided by the present invention may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implant or other modes of administration. For example, parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, intrasternal administration, intracerebrospinal administration, intralesional administration, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of compound actually administered can be determined by a physician, as the case may be, including the condition to be treated, the chosen route of administration, the compound actually administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
When used to prevent a condition according to the invention, a subject at risk of developing the condition is administered a compound provided herein, typically based on physician's advice and under the supervision of a physician, at a dosage level as described above. Subjects at risk of developing a particular disorder, typically include subjects with a family history of the disorder, or those determined to be particularly susceptible to developing the disorder by genetic testing or screening.
The pharmaceutical compositions provided herein may also be administered chronically ("chronic administration"). By long-term administration is meant administration of the compound or pharmaceutical composition thereof over a long period of time, e.g., 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or administration may continue indefinitely, e.g., for the remainder of the subject's life. In some embodiments, chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, e.g., within the therapeutic window.
Various methods of administration may be used to further deliver the pharmaceutical compositions of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered as a bolus, e.g., in order to increase the concentration of the compound in the blood to an effective level. The bolus dose depends on the targeted systemic level of the active ingredient through the body, e.g., intramuscular or subcutaneous bolus doses result in slow release of the active ingredient, while a bolus delivered directly to the vein (e.g., by IV intravenous drip) can be delivered more rapidly, resulting in a rapid rise in the concentration of the active ingredient in the blood to an effective level. In other embodiments, the pharmaceutical composition may be administered as a continuous infusion, e.g., by IV intravenous drip, to provide a steady state concentration of the active ingredient in the body of the subject. Furthermore, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders. More typically, however, the compositions are provided in unit dosage form for convenient accurate dosing. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material adapted to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of the liquid compositions, or pills, tablets, capsules and the like in the case of solid compositions. In such compositions, the compound is typically a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being various carriers or excipients and processing aids useful in forming the desired form of administration.
For oral dosages, a representative regimen is one to five oral dosages, particularly two to four oral dosages, typically three oral dosages per day. Using these dosing modes, each dose provides about 0.01 to about 20mg/kg of a compound of the invention, with preferred doses each providing about 0.1 to about 10mg/kg, especially about 1 to about 5mg/kg.
In order to provide blood levels similar to, or lower than, those used with the injected dose, transdermal doses are generally selected in amounts of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
From about 1 to about 120 hours, especially 24 to 96 hours, the injection dosage level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour. To obtain sufficient steady state levels, a preload bolus of about 0.1mg/kg to about 10mg/kg or more may also be administered. For human patients of 40 to 80kg, the maximum total dose cannot exceed about 2 g/day.
Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous carriers, as well as buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like. Solid forms may include, for example, any of the following components, or compounds with similar properties: a binder, for example, microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example, starch or lactose, disintegrants, for example, alginic acid, primogel or corn starch; lubricants, for example, magnesium stearate; glidants, e.g., colloidal silicon dioxide; sweetening agents, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based on sterile saline or phosphate buffered saline for injection, or other injectable excipients known in the art. As previously mentioned, in such compositions, the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as an ointment, the active ingredient is typically combined with a paraffinic or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with a cream base, for example of the oil-in-water type. Such transdermal formulations are well known in the art and typically include other components for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and compositions are included within the scope of the present invention.
The compounds of the present invention may also be administered by transdermal means. Thus, transdermal administration can be achieved using a reservoir (reservoir) or porous membrane type, or a patch of various solid matrices.
The above components of the compositions for oral, injectable or topical administration are merely representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences,17th edition,1985, mack Publishing company, easton, pennsylvania, section 8, which is incorporated herein by reference.
The compounds of the present invention may also be administered in sustained release form, or from a sustained release delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6, 7 and 8 α -1, 4-linked glucose units, respectively, which optionally include one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated and sulfoalkyl ether substitution. In some embodiments, the cyclodextrin is sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, e.g., U.S.5,376,645. In some embodiments, the formulation includes hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Indications of
The present invention provides a method of treating a disease, such as a MEK kinase mediated disease, in a subject comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate compound thereof, or a pharmaceutical composition of the present invention. The treatment may also be combined with other therapies such as radiation therapy, chemotherapy.
In particular embodiments, MEK kinase-mediated diseases include inflammatory diseases, infections, autoimmune disorders, stroke, ischemia, cardiac disorders, neurological disorders, fibrotic disorders, proliferative disorders, hyperproliferative disorders, tumors, leukemias, neoplasms, cancers, malignancies, metabolic diseases, and malignant diseases.
The present invention also provides a method of treating an MEK kinase mediated inflammatory disease comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In particular embodiments, the MEK kinase-mediated inflammatory disease comprises rheumatoid arthritis or multiple sclerosis.
The present invention also provides a method of treating a MEK kinase mediated proliferative disease comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In particular embodiments, the MEK kinase-mediated proliferative disease comprises cancer, psoriasis, restenosis, autoimmune disease or atherosclerosis.
The present invention also provides a method of treating MEK kinase mediated cancer comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
In particular embodiments, MEK kinase-mediated cancers include melanoma (e.g., BRAF V600 mutated melanoma), acute myeloid leukemia, glial flow, neurofibroma (e.g., neurofibromatosis type 1 (NF 1) or plexiform neurofibromas), non-small cell lung cancer, breast cancer, serous cancer, gastrointestinal stromal tumor, lung non-squamous cancer, colorectal cancer (e.g., BRAF V600 mutated colorectal cancer), biliary tract cancer, myeloma.
The present invention describes inhibitors of MEK kinase useful in the treatment of diseases driven by over-activation, abnormal activation, constitutive activation, gain-of-function mutations of MEK kinase and/or substrate kinases including but not limited to ERK. Such diseases encompass hyperproliferative diseases including, but not limited to, psoriasis, keloids, hyperplasia of the skin, benign Prostate Hyperplasia (BPH), solid tumors such as respiratory tract (including, but not limited to, small cell and non-small cell lung cancers), brain (including, but not limited to, glioma, neurofibroma, plexiform neurofibroma, medulloblastoma, ependymoma, neuroectodermal and pineal tumors), breast (including, but not limited to, invasive ductal carcinoma, invasive lobular carcinoma, ductal and lobular carcinoma in situ), reproductive organs (including, but not limited to, prostate, testicular, ovarian, endometrial, cervical, vaginal, vulval, and uterine sarcomas), digestive tract (including, but not limited to, esophagus, colon, stomach, gall bladder, pancreas, rectum, anus, small intestine, and salivary gland adenocarcinomas), urinary tract (including, but not limited to, bladder, ureter, kidney, urinary and renal papillary carcinomas), eye (including, but not limited to, melanoma and retinoblastoma), liver (including, but not limited to, hepatocellular carcinoma and bile duct), skin (including, melanoma, squamous cell carcinoma of the nasopharynx, squamous cell carcinoma of the pharynx), squamous cell carcinoma of the mouth, squamous cell carcinoma of the pharynx, thyroid, and thyroid. Hyperproliferative diseases also include leukemias (including, but not limited to, acute lymphoblastic leukemia, acute concomitant leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, and hairy cell leukemia), sarcomas (including, but not limited to, soft tissue sarcoma, osteosarcoma, lymphosarcoma, rhabdomyosarcoma), and lymphomas (including, but not limited to, non-hodgkin's lymphoma, AIDS-related lymphoma, cutaneous T-cell lymphoma, burkitt's lymphoma, hodgkin's disease, and lymphomas of the central nervous system).
Inhibitors of MEK kinase are described for use in certain diseases involving abnormal regulation of mitogen extracellular kinase activity, including but not limited to hepatomegaly, heart failure, cardiac hypertrophy, diabetes, stroke, alzheimer's disease, cystic fibrosis, septic shock or asthma.
Inhibitors of MEK kinase are described for use in the treatment of diseases and disorders associated with aberrant, abnormal and/or excessive angiogenesis. Such disorders associated with angiogenesis include, but are not limited to, tumor growth and metastasis, ischemic retinal vein occlusion, diabetic retinopathy, macular degeneration, neovascular glaucoma, psoriasis, inflammation, rheumatoid arthritis, vascular graft restenosis, and in-stent restenosis.
The present invention also provides a method for treating acute myeloid leukemia, comprising administering to said subject a compound of the present invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention. In particular embodiments, the acute myeloid leukemia is relapsed and/or refractory acute myeloid leukemia.
The present invention also provides a method of treating glioma comprising administering to said subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention.
The present invention also provides a method of treating neurofibroma comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention. In a specific embodiment, the neurofibroma is selected from neurofibromatosis type 1 (NF 1) or plexiform neurofibromas.
The present invention also provides a method of treating serous cancer comprising administering to the subject a compound of the present invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the present invention. In particular embodiments, the serous carcinoma is selected from recurrent or persistent low grade ovarian, fallopian tube, or primary peritoneal serous carcinoma.
Combination therapy
The compounds of the present invention may be used alone or in combination with other therapeutic agents. 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. Specifically, the method comprises the following steps:
the present invention provides a method of treating BRAF kinase-mediated cancer comprising administering to the subject a compound of the present invention in combination with a BRAF inhibitor (each optionally in the form of a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate), and optionally a third therapeutic agent.
In particular embodiments, the BRAF inhibitor is selected from vemurafenib (vemurafenib), dabrafenib (dabrafenib), cannfenib (encorafenib) or the following compounds disclosed in WO 2020/011141 A1:
Figure BDA0003692806340000261
compounds disclosed in WO 2020/011141 A1
Preferably, the BRAF inhibitor is selected from vemurafenib (vemurafenib), dabrafenib (dabrafenib), canofenib (encorafenib), and the following compounds disclosed in WO 2020/011141 A1:
(S) -methyl- (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate,
(S) - (methyl-d) 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1-isopropyl-1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate,
(S) - (methyl-d) 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate,
(1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5 ) A methyl carbamate (methyl carbamate),
(S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5 ) A methyl carbamate (methyl carbamate),
(R) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5 ) The amino-formic acid methyl ester is obtained,
(1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5) carbamic acid methyl ester,
(S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5 ) The amino-formic acid methyl ester is obtained,
(R) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5) carbamic acid methyl ester.
More preferably, the BRAF inhibitor is selected from vemurafenib (vemurafenib), dabrafenib (dabrafenib), canofenib (encorafenib), and the following compounds disclosed in WO 2020/011141 A1:
(S) -methyl- (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate,
(S) - (methyl-d) 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1-isopropyl-1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate,
(S) - (methyl-d) 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate,
(S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (prop-2-yl) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) prop-2-yl-1, 3-d 5 ) The amino-formic acid methyl ester is obtained,
(S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5 ) Methyl carbamate.
In particular embodiments, the method of treating a BRAF kinase-mediated cancer does not comprise a third therapeutic agent.
In particular embodiments, the method of treating a BRAF kinase-mediated cancer comprises a third therapeutic agent. In particular embodiments, the third therapeutic agent is selected from an immune checkpoint inhibitor, e.g., pabulizumab (pembrolizumab), ipilimumab (ipilimumab) and nivolumab (nivolumab), atelizumab (atezolizumab), avilumab (avelumab), devaluzumab (durvalumab), pidilizumab (pidilizumab), PDR-001 (BAP 049-clone-E, disclosed and used in WO 2017/019896); preferably, for example, paclilizumab (pembrolizumab), yipriuma (ipilimumab) and nivolumab (nivolumab). In particular embodiments, the third therapeutic agent is selected from an EGFR antibody, e.g., cetuximab (cetuximab), panitumumab (panitumumab), tolitumumab ((Necitumumab); preferably, e.g., cetuximab (cetitumumab); in particular embodiments, the third therapeutic agent is a mitotic inhibitor, e.g., CDK4/6 inhibitor; preferably, e.g., palbociclib (palbociclib), ribbociclib (ribociclib), abbesib (abemaciclib); preferably, e.g., palbociclib (palbociclib).
In particular embodiments, the BRAF kinase-mediated cancer is melanoma, brain tumors such as glioblastoma multiforme (GBM), acute Myelogenous Leukemia (AML), lung cancer, papillary thyroid cancer, low grade ovarian cancer, colorectal cancer, multiple myeloma, and nervous system cancer. Preferably, the BRAF kinase-mediated cancer is metastatic or unresectable melanoma, papillary thyroid cancer, low grade ovarian cancer, and colorectal cancer. In a specific embodiment, the BRAF kinase is a BARF V600 mutant kinase. In specific embodiments, the BRAF V600 mutation is BRAF V600E, BRAF V600D, BRAF V600R, BRAF V600G, and BRAF V600K. In particular embodiments, the BRAF V600 mutations are BRAF V600E and BRAF V600K. In particular embodiments, the BRAF kinase-mediated cancer is metastatic or unresectable melanoma with a BRAF V600 mutation. In particular embodiments, the BRAF kinase-mediated cancer is metastatic or unresectable melanoma with BRAF V600E or BRAF V600K mutations. In a specific embodiment, the BRAF kinase-mediated cancer is BRAF V600 mutated colorectal cancer. In particular embodiments, the BRAF kinase-mediated cancer is BRAF V600E or BRAF V600K mutated colorectal cancer.
The present invention also provides a method of treating NRAS or KRAS or EGFR mutated cancer comprising administering to the subject a compound of the present invention in combination with an EGFR inhibitor (each optionally in the form of a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate). In specific embodiments, the EGFR inhibitor is selected from gefitinib (gefitinib), erlotinib (erlotinib), afatinib (afatinib), dacomitinib (dacomitinib), lapatinib (lapatinib), oxitinib (osicertinib), amitinib, formetinib, CO-1686, WZ4002, PD153035, PF00299804, cetuximab, panitumumab, tolbizumab. In a specific embodiment, the NRAS-mutated cancer is NRAS-mutated non-small cell lung cancer. In specific embodiments, the NRAS mutation is selected from the group consisting of E63K, G12V, G12R, G12A, G12D, G12S and G12C, or an increase in the copy number of the NRAS gene.
The present invention also provides a method of treating advanced KRAS positive metastatic colorectal cancer comprising administering to the subject a compound of the invention in combination with mflfiri (each optionally in the form of a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate).
The present invention also provides a method of treating gastrointestinal stromal tumors comprising administering to the subject a compound of the invention in combination with pexidartinib (each optionally in the form of a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate).
The present invention also provides a method of treating a gastrointestinal stromal tumor comprising administering to the subject a compound of the invention in combination with imatinib (imatinib) (each optionally in the form of a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate).
The present invention also provides a method of treating non-squamous carcinoma of the lung comprising administering to the subject a compound of the invention in combination with carboplatin and pemetrexed (each optionally in the form of a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate).
The present invention also provides a method of treating biliary tract cancer comprising administering to the subject a compound of the present invention in combination with capecitabine (each optionally in the form of a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate).
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.
Abbreviations:
Pd 2 (dba) 3 : tris (dibenzylideneacetone) dipalladium
Xant-phos 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene
NBS: n-bromosuccinimide
NIS: n-iodosuccinimide
PTSA: p-toluenesulfonic acid
EDCI: 1-Ethyl- (3-dimethylaminopropyl) carbodiimides hydrochloride
HOBT: 1-hydroxybenzotriazole
DMAP: dimethylaminopyridine
TEA: triethylamine
DIEA: n, N-diisopropylethylamine
TFAA: trifluoroacetic anhydride
Na 2 CO 3 : sodium carbonate
K 2 CO 3 : potassium carbonate
Cs 2 CO 3 : cesium carbonate
EtOH: ethanol
BnOH: benzyl alcohol
DCM: methylene dichloride
THF: tetrahydrofuran (THF)
ACN: acetonitrile (ACN)
DME: ethylene glycol dimethyl ether
DMF: n, N-dimethylformamide
DMSO, DMSO: dimethyl sulfoxide
TMSCl: trimethylchlorosilane
3 Intermediate A-15- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d) -1H-benzimidazole-6-carboxylic acid Preparation of
Figure BDA0003692806340000301
The following synthetic route is adopted
Figure BDA0003692806340000302
Step 1: synthesis of Compound 2,3, 4-trifluoro-5-nitrobenzoic acid
2,3, 4-trifluorobenzoic acid (20g, 113.6mmol) is dissolved in 60ml of concentrated sulfuric acid, the reaction liquid is heated to 90 ℃, then a mixed solution of the concentrated sulfuric acid (12g, 122.4mmol) and concentrated nitric acid (12.8g, 132.1mmol) is dropwise added, the stirring reaction is carried out for 5 hours, the TLC monitoring reaction is finished, the reaction liquid is cooled to room temperature, the reaction liquid is slowly dropwise added into ice water, extraction is carried out for 3-4 times by ethyl acetate, organic phases are combined, saturated saline is washed for 2-3 times, anhydrous sodium sulfate is dried, and 24.0g of white solid is obtained after filtration and concentration, and the yield is 95.6%. LC-MS (APCI) M/z =220.1 (M-1) -
Step 2: synthesis of Compound 2, 3-difluoro-4-amino-5-nitrobenzoic acid
Placing the 2,3, 4-trifluoro-5-nitrobenzoic acid (5.81g, 26.3mmol) obtained in the previous step into a 100ml flask, adding 30ml of deionized water, cooling the mixture to 0 ℃ in ice bath, slowly dropwise adding concentrated ammonia water (18.4g, 131.5mmol), heating to room temperature, stirring for reaction overnight, monitoring by TLC, adjusting the pH to be less than 2 by using 1N dilute hydrochloric acid in ice bath, precipitating a light yellow solid, filtering, and drying in vacuum to obtain 4.99g of product with the yield of 87.1%。LC-MS(APCI):m/z=219.4(M+1) +
And 3, step 3: synthesis of compound methyl 2, 3-difluoro-4-amino-5-nitrobenzoate
2, 3-difluoro-4-amino-5-nitrobenzoic acid (4.99g, 22.9mmol) and trimethylchlorosilane (4.97g, 45.8mmol) are added into a reaction bottle, 100ml of methanol is added for dissolution, the reaction liquid is heated to 65 ℃ under the protection of nitrogen and stirred overnight, TLC monitors the reaction to be finished, the reaction liquid is cooled to room temperature, and after concentration, the reaction liquid is separated by a silica gel column to obtain 4.51g of yellow solid, and the yield is 84.9%. LC-MS (APCI) M/z =233.4 (M + 1) +
And 4, step 4: synthesis of compound methyl 2, 4-diamino-3-fluoro-5-nitrobenzoate
Methyl 2, 3-difluoro-4-amino-5-nitrobenzoate (1.0g, 4.3mmol) obtained in the previous step is dissolved in 10ml dioxane, ammonia water (1.6 ml,21.4 mmol) is added, the temperature is raised to 90 ℃ under the protection of nitrogen for reaction for 2-4 hours, and the reaction is monitored by TLC to be finished. The reaction solution is cooled to room temperature, added with 30ml of water for dilution, extracted with ethyl acetate for 3-4 times, the organic phases are combined, washed with saturated saline solution, dried by anhydrous sodium sulfate, filtered, and the filtrate is concentrated and dried to obtain 1.03g of yellow solid which is directly put into the next reaction. LC-MS (APCI) M/z =230.8 (M + 1) + .
And 5: synthesis of compound methyl 2,4, 5-triamino-3-fluorobenzoate
The methyl 2, 4-diamino-3-fluoro-5-nitrobenzoate (1.03g, 4.5 mmol), reduced iron powder (2.5 g, 45mmol) and anhydrous ammonium chloride (1.44g, 26.9 mmol) obtained in the above step were placed in a 100ml flask, 15ml of ethanol and 5ml of water were added, the reaction was stirred at 70 ℃ for 1 to 2 hours, and the completion of the reaction was monitored by TLC. The catalyst was removed by filtration with celite and the filtrate was concentrated to yield 860mg of a gray solid which was used directly in the next step without further purification. LC-MS (APCI) M/z =200.1 (M + 1) +
Step 6: synthesis of compound 4-fluoro-5-amino-1H-benzimidazole-6-methyl formate
The methyl 2,4, 5-triamino-3-fluorobenzoate (860mg, 4.32mmol) and formamidine acetate (542mg, 5.21mmol) obtained in the above step were added to 10ml of anhydrous ethanol, the mixture was heated to 80 ℃ and refluxed for 4 hours, and the reaction was monitored by TLCAnd finishing. Cooling the reaction solution to room temperature, concentrating to remove the solvent, adding 20ml of water for dilution, extracting with dichloromethane for 3-4 times, combining organic phases, concentrating, and purifying by silica gel column chromatography to obtain 486mg of light yellow solid, yield: and (4) 53.8%. LC-MS (APCI) M/z =210.5 (M + 1) +
And 7: synthesis of compound 4-fluoro-5-amino-1- (methyl-d 3) -1H-benzimidazole-6-methyl formate
The 4-fluoro-5-amino-1H-benzimidazole-6-carboxylic acid methyl ester (600mg, 2.86mmol), deuterated iodomethane (458mg, 3.16mmol) and potassium carbonate (794mg, 5.76mmol) obtained in the above step were charged into a 50ml reaction flask, 10ml of anhydrous DMF was added, the temperature was raised to 70 ℃ under nitrogen protection, the reaction was stirred for 2 hours, and the completion of the reaction was monitored by TLC. Cooling to room temperature, diluting with 30ml water, extracting with ethyl acetate for 3-4 times, mixing organic phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain brown solid 201mg with 31.5% yield. LC-MS (APCI) M/z =227.5 (M + 1) +
And step 8: synthesis of compound methyl 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d 3) -1H-benzimidazole-6-carboxylate
4-fluoro-5-amino-1- (methyl-d 3) -1H-benzimidazole-6-methyl formate (200mg, 0.9mmol), 2-fluoro-4-bromo-1-iodobenzene (300mg, 0.99mmol) and Pd obtained in the previous step 2 (dba) 3 Adding (16mg, 0.017mmol), xantphos (26mg, 0.044mmol) and cesium carbonate (584mg, 1.79mmol) into a 20ml microwave tube, adding 8ml ethylene glycol dimethyl ether under the protection of nitrogen, sealing, heating to 90 ℃ by microwave for 1 hour, monitoring by TLC to complete the reaction, cooling to room temperature, concentrating to remove the solvent, and purifying by silica gel column chromatography to obtain 229mg of an earthy yellow solid, wherein the yield is as follows: 64.0 percent. LC-MS (APCI) M/z =399.2 (M + 1) +
And step 9: synthesis of intermediate A-1
Adding methyl 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d 3) -1H-benzimidazole-6-carboxylate (1.63g, 4.09mmol) into a 100ml reaction flask, dissolving with 30ml tetrahydrofuran and 10ml water, adding sodium hydroxide (0.68g, 17.0 mmol), heating to 45 deg.C, stirring for 3-5 hours, TLC monitoring reaction, concentrating to remove tetrahydrofuran, addingDiluting with 10ml of water, adjusting pH to acidity with 1N diluted hydrochloric acid, separating out a white solid, filtering, and vacuum-drying to obtain 1.3g of crude product, which is directly used in the next reaction. LC-MS (APCI) M/z =385.2 (M + 1) +
Preparation of intermediate A-25- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-methyl formate Prepare for
Figure BDA0003692806340000321
The following synthetic route is adopted
Figure BDA0003692806340000331
Step 1: synthesis of compound 4-fluoro-5-amino-1-methyl-1H-benzimidazole-6-methyl formate
4-fluoro-5-amino-1H-benzimidazole-6-carboxylic acid methyl ester (600mg, 2.86mmol), methyl iodide (453mg, 3.16mmol) and potassium carbonate (794mg, 5.76mmol) were added to a 50ml reaction flask, 10ml of anhydrous DMF was added, the temperature was raised to 70 ℃ under nitrogen protection and the reaction was stirred for 2 hours, and the reaction was monitored by TLC. Cooling to room temperature, diluting with 30ml water, extracting with ethyl acetate for 3-4 times, mixing organic phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain 203mg brown solid with 31.9% yield. LC-MS (APCI) M/z =224.5 (M + 1) +
Step 2: synthesis of compound methyl 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-carboxylate
Adding methyl 4-fluoro-5-amino-1-methyl-1H-benzimidazole-6-carboxylate (200mg, 0.9mmol), 2-fluoro-4-bromo-1-iodobenzene (300mg, 0.99mmol), pd2 (dba) 3 (16mg, 0.017mmol), xantphos (26mg, 0.044mmol) and cesium carbonate (584mg, 1.79mmol) into a 20ml microwave tube, adding 8ml ethylene glycol dimethyl ether under the protection of nitrogen, sealing, heating to 90 deg.C with microwave for 1 hr, monitoring by TLC, cooling to room temperature, concentrating, and removing impuritiesThe solvent was removed and purified by silica gel column chromatography to give 208mg of a tan solid, yield: 58.1 percent. LC-MS (APCI) M/z =396.2 (M + 1) +
And step 3: synthesis of intermediate A-2
Adding methyl 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-carboxylate (1.63g, 4.09mmol) into a 100ml reaction bottle, adding 30ml tetrahydrofuran and 10ml water for dissolving, adding sodium hydroxide (0.68g, 17.0 mmol), heating to 45 ℃, stirring for reaction for 3-5 hours, monitoring by TLC (thin layer chromatography), concentrating to remove tetrahydrofuran, adding 10ml water for diluting, adjusting pH to acidity by using 1N diluted hydrochloric acid, precipitating a white-like solid, filtering, drying in vacuum to obtain 1.3g of a crude product, and directly putting the crude product into the next reaction. LC-MS (APCI) M/z =382.1 (M + 1) +
Preparation of intermediate B-benzyl 12- (aminooxy) -2, 2-dideuteroacetate
Figure BDA0003692806340000341
The following synthetic route is adopted
Figure BDA0003692806340000342
Step 1: synthesis of compound 2-bromo-2, 2-dideuteroacetic acid
Deuterated acetic acid (10g, 156mmol) is added into 80ml trifluoroacetic anhydride, a bromine dioxane complex (37g, 149mmol) is added at room temperature, the mixture is stirred and reacted overnight under the protection of nitrogen, the reaction is monitored by GC to be finished, 10.8g of oily liquid is obtained after decompression and concentration, and the oily liquid is directly put into the next reaction without purification.
And 2, step: synthesis of compound 2-bromo-2, 2-dideuterobenzyl acetate
2-bromo-2, 2-dideuteroacetic acid (10.8g, 78.55mmol), benzyl alcohol (8.5g, 78.6 mmol), EDCI (16.6g, 86.6 mmol) and DMAP (1.06g, 8.7mmol) were dissolved in 80ml of anhydrous DMF, and the reaction was stirred at room temperature under nitrogen for 3-5 hours and monitored by TLC for completion. Adding 200ml of water for dilution,extracting with ethyl acetate for 3-4 times, mixing organic phases, washing with saturated saline, drying with anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain anhydrous oily liquid 7.5g with yield of 21% in two steps. LC-MS (APCI) M/z =280.3 (M + 1) +
And 3, step 3: synthesis of Compound 2- ((1, 3-dioxoisoindolin-2-yl) oxy) -2, 2-dideuterobenzyl acetate
Benzyl 2-bromo-2, 2-dideuterio acetate (3.94g, 17.3mmol), N-hydroxyphthalimide (2.51g, 15.38mmol) and triethylamine (2.36g, 23.3mmol) obtained in the previous step were added to 40ml of anhydrous DMF and stirred overnight at room temperature under nitrogen. Diluting with 150ml of water, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated brine, drying with anhydrous sodium sulfate, concentrating, and separating with silica gel column to obtain white solid 4.36g with yield of 81%. LC-MS (APCI) M/z =314.4 (M + 1) +
And 4, step 4: synthesis of intermediate B-1
Benzyl 2- ((1, 3-dioxoisoindolin-2-yl) oxy) -2, 2-dideuteroacetate (4.36g, 14.0 mmol) and hydrazine hydrate (1.0 g,20.0 mmol) obtained in the above step were dissolved in 45ml of dichloromethane, the reaction was stirred overnight at room temperature, and the completion of the reaction was monitored by TLC. Insoluble matter was removed by filtration, and the filtrate was concentrated and then separated by a silica gel column to obtain 1.99g of a yellow oily liquid in a yield of 78.3%. LC-MS (APCI) M/z =184.7 (M + 1) +
Example 15- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-N- (2-hydroxyethoxy) -1- (methyl-d) 3 )-1H- Preparation of benzimidazole-6-carboxamide (Compound T-1)
Figure BDA0003692806340000351
The following synthetic route is adopted
Figure BDA0003692806340000352
Step 1: the compound 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d) 3 ) Synthesis of (E) -N- (2- (ethyleneoxy) ethoxy) -1H-benzimidazole-6-carboxamide
Intermediate A-1 (1.3g, 3.4 mmol), O- (2- (vinyloxy) ethyl) hydroxylamine (0.41g, 4.02mmol), EDCI (0.78g, 4.06mmol), HOBT (0.55g, 4.08mmol) and triethylamine (0.41g, 4.06mmol) were charged into a 100ml reaction flask, 15ml of anhydrous DMF was added under nitrogen protection, the reaction was stirred overnight at room temperature, TLC monitored for completion of the reaction, diluted with 50ml of water, extracted 3-4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, concentrated and purified by silica gel column chromatography to give 0.88g of off-white solid, yield: and (5) 55.3%. LC-MS (APCI) M/z =470.5 (M + 1) +1 H NMR(400MHz,DMSO-d 6 ):δ11.79(s,1H),8.39(s,1H),7.92(s,1H),7.71(d,J=13.2Hz,1H),7.61(t,J=9.4Hz,1H),7.23-7.27(m,1H),6.52-6.43(m,1H),6.35(dd,J=8.7,4.1Hz,1H),4.23-4.10(m,1H),4.06-3.91(m,3H),3.83(s,2H).
And 2, step: synthesis of Compound T-1
The 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d) obtained in the previous step 3 ) -N- (2- (ethyleneoxy) ethoxy) -1H-benzimidazole-6-carboxamide (0.88g, 1.88mmol) is dissolved in 15ml ethanol, cooled to 0 ℃ in ice bath, 2N dilute hydrochloric acid (9.4ml, 18.8mmol) is slowly added dropwise, after addition, the mixture is heated to room temperature and stirred for 2-4 hours, TLC monitors the completion of the reaction, 1N sodium hydroxide aqueous solution is used for adjusting the pH to be alkalescent, the solvent is removed by concentration, and silica gel column chromatography purification is carried out to obtain 0.42g white solid, the yield is: 50.5 percent. LC-MS (APCI) M/z =444.7 (M + 1) +1 H NMR(400MHz,CD 3 OD):δ8.30(s,1H),8.09(s,1H),7.72(s,1H),7.50(d,J=2.0Hz,1H),7.21-7.17(m,1H),6.40(dd,J=8.8,4.8Hz,1H),3.97-3.89(m,2H),3.70-3.66(m,2H).
Example 25- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-N- (2-hydroxyethoxy-1, 2-d) 4 ) -1-A Preparation of 1H-benzimidazole-6-carboxamide (Compound T-2)
Figure BDA0003692806340000361
The following synthetic route is adopted
Figure BDA0003692806340000362
Step 1: synthesis of the compound benzyl 2- ((5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-formylamino) oxy) -2, 2-dideuteroacetate
Adding intermediate A-2 (1.3g, 3.4 mmol), intermediate B-1 (0.74g, 4.02mmol), EDCI (0.78g, 4.06mmol), HOBT (0.55g, 4.08mmol) and triethylamine (0.41g, 4.06mmol) into a 100ml reaction flask, adding 15ml of anhydrous DMF under the protection of nitrogen, stirring at room temperature overnight, monitoring the reaction by TLC, adding 50ml of water for dilution, extracting with ethyl acetate for 3-4 times, combining organic phases, washing with saturated saline, and purifying by silica gel column chromatography after concentration to obtain 1.78g of off-white solid, wherein the yield is as follows: 95.7 percent. LC-MS (APCI) M/z =547.3 (M + 1) +1 H NMR(400MHz,DMSO-d 6 ):δ8.39(s,1H),7.84(s,1H),7.78-7.64(m,1H),7.58(d,J=2.3Hz,1H),7.40-7.28(m,5H),7.23(dd,J=8.8,2.2Hz,1H),6.32(dd,J=8.7,3.6Hz,1H),5.13(s,2H),3.88(s,3H).
And 2, step: synthesis of Compound T-2
Dissolving the benzyl 2- ((5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-formylamino) oxy) -2, 2-dideuteroacetate (1.78g, 3.25mmol) obtained in the previous step in 40ml of anhydrous THF, cooling to-10 ℃ under the protection of nitrogen, adding deuterated lithium aluminum hydride (273mg, 6.5mmol) in batches, continuing to stir for 3-4 hours at low temperature, monitoring by TLC after the reaction is finished, adding 5ml of water for quenching reaction at low temperature, then adding 10ml of 15% sodium hydroxide aqueous solution, assisting filtration by using kieselguhr, concentrating the filtrate, and purifying by using silica gel column chromatography to obtain 0.47g of white-like solid with yield: 32.6 percent. LC-MS (APCI) M/z =445.6 (M + 1) +1 H NMR(400MHz,CD 3 OD):δ11.73-11.64(m,1H),8.40(s,1H),7.93(brs,1H),7.74(s,1H),7.61(s,1H),7.26(dd,J=8.8Hz,2.0Hz,1H),6.36(dd,J=8.7Hz,4.0Hz,1H),4.67(brs,1H),3.91(s,3H).
And (4) testing the biological activity.
(1) Cytotoxicity test
The inhibitory effect of the compounds of the examples on the activity of HT-29 cells was examined.
Cell line: HT-29 (cell type: adherent; cell number/well: 3000; culture medium: RPMI-1640+10% FBS;) was cultured at 37 ℃ under 5% CO2, 95% humidity conditions.
Consumable and reagent: fetal bovine serum FBS (GBICO, cat # 10099-141),
Figure BDA0003692806340000371
Luminecent Cell visual Assay (Promega, cat # G7572), 96-well transparent flat-bottom black-wall plate (r) ((r))
Figure BDA0003692806340000372
Cat#3603)。
The instrument comprises the following steps: spectraMax multi-label micropore plate detector, MD,2104-0010A; CO2 incubator, thermo Scientific, model 3100Series; biological safety cabinet, thermo Scientific, model 1300Series A2; inverted microscope, olympus, CKX41SF; refrigerator, SIEMENS, KK25E76TI.
The experimental steps are as follows:
1) Cell culture and inoculation: i) Cells in the logarithmic growth phase were harvested and counted using a platelet counter. Detecting the cell viability by using a trypan blue exclusion method to ensure that the cell viability is over 90 percent; ii) adjusting the cell concentration; add 90 μ Ι _ of cell suspension to 96-well plates, respectively; iii) The cells in the 96-well plate were incubated overnight at 37 ℃ in 5% CO2 at 95% humidity.
2) Drug dilution and dosing: i) Preparing 10 times of drug solution, wherein the highest concentration is 100 mu M, the concentration is 9, the dilution is 3.16 times, 10 mu L of drug solution is added into each hole of a 96-hole plate inoculated with cells, and three multiple holes are arranged at each drug concentration; ii) the cells in the dosed 96-well plates were placed under conditions of 37 ℃ 5% CO2, 95% humidity for further incubation for 72 hours, after which CTG analysis was performed.
3) Reading the plate at the end: i) Melt CTG reagents and equilibrate cell plates to room temperature for 30 minutes; ii) adding an equal volume of CTG solution per well; iii) Vibrating on an orbital shaker for 5 minutes to lyse the cells; iv) the cell plate is left at room temperature for 20 minutes to stabilize the luminescence signal; v) reading the cold light value.
Data processing: data were analyzed using GraphPad Prism 5.0 software, fitted to data using nonlinear S-curve regression to derive dose-response curves, and IC50 values were calculated therefrom. Cell survival (%) = (Lum test drug-Lum culture medium control)/(Lum cell control-Lum culture medium control) × 100%.
The compounds of the invention were tested in the cytotoxicity assay described above and the results show that: compared with the non-deuterated compound Binimetinib, the compound has stronger activity on HT-29 cells.
(2) Metabolic stability evaluation
Metabolic stability, which is generally used to describe the rate and extent to which a compound is metabolized, is one of the major factors affecting pharmacokinetic properties. Many compounds are substrates for CYP450 enzymes and other drug metabolizing enzymes, and liver microsomes are CYP450 rich systems, and the purpose of this experiment was to study the in vitro metabolic stability by incubating the compounds of the invention with human liver microsomes and/or mouse liver microsomes separately and measuring the remaining proportion of compounds using LC-MS/MS.
(1) Preparation of the solution
Phosphate Buffered Saline (PBS): taking prepared KH 2 PO 4 (0.5M) 150mL of solution and K 2 HPO 4 700mL of the (0.5M) solution was mixed and then K was added 2 HPO 4 (0.5M) solution the pH of the mixture was adjusted to 7.4 and the mixture was stored at 4 ℃ as 5-fold concentration PBS for further use. Before use, the mixture was diluted 5-fold with ultrapure water, and 3.3mM magnesium chloride was added to obtain phosphate buffered saline PBS (100 mM).
NADPH regeneration system solution: an NADPH solution containing 6.5mM NADP,16.5mM G-6-P,3U/mL G-6-P D was prepared in 5mL of PBS.
Internal standard stop solution: and preparing 50ng/mL propranolol hydrochloride and 200ng/mL tolbutamide as internal standard working solution by using acetonitrile.
Human liver microsome solution: 0.31mL of human liver microsomes (25 mg/mL) was added to 0.961mL of PBS (pH 7.4) and mixed to obtain a human liver microsome dilution with a protein concentration of 0.625 mg/mL.
Mouse liver microsome solution: 0.31mL of mouse liver microsomes (25 mg/mL) was added to 0.961mL of PBS (pH 7.4) and mixed to obtain a mouse liver microsome dilution with a protein concentration of 0.625 mg/mL.
Sample working solution: the compounds of the invention and non-deuterated compound powders, positive control dextromethorphan powder and omeprazole powder were formulated in DMSO to 10mM as sample stocks. Then diluted with 70% acetonitrile-water to obtain 0.25mM sample working solution.
(2) Incubation of samples
398. Mu.L of human liver microsome diluent was added to a 96-well incubation plate (N = 2), and 2. Mu.L of 0.25mM test compound and dextromethorphan were added, respectively, and mixed well.
398. Mu.L of mouse liver microsome diluent was added to a 96-well incubation plate (N = 2), and 2. Mu.L of 0.25mM test compound and dextromethorphan were added thereto, respectively, and mixed well.
Add 300. Mu.L of pre-chilled stop solution to each well in a 96-well deep-well plate and place 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 5min. mu.L of the incubation solution was taken out from 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 test compound was 1. Mu.M, and the protein concentration was 0.5mg/mL.
When the reaction was carried out for 10 min, 30 min and 90min, 100. Mu.L of each reaction solution was added to the stop plate and vortexed for 3min to stop the reaction.
The plates were centrifuged at 5000rpm at 4 ℃ for 15min. And (3) taking 200 mu L of supernatant into a 96-well plate which is added with 200 mu L of ultrapure water in advance, mixing uniformly, carrying out sample analysis by adopting LC-MS/MS, and injecting 10uL of sample.
(3) Sample analysis method
In the experiment, an LC-MS/MS system is adopted to detect peak areas of a compound to be detected, dextromethorphan, omeprazole and an internal standard, and the peak area ratio of the compound to the internal standard is calculated.
(4) Data processing
The peak areas of the sample and the internal standard are obtained by a mass spectrometer and analysis software, and a substrate elimination rate constant K can be obtained by using a Graphpad prism7.0 software single exponential degradation model to plot the residual amount (R%) of the compound and time
C t /C 0 =exp(-K*t)
And calculating the half-life period T according to the following formula 1/2 And intrinsic clearance CL int Where V/M is equal to 1/C (protein).
Figure BDA0003692806340000391
T 1/2 (min);CL int (μL/min/mg)。
The experimental results are as follows: the compounds of the invention and non-deuterated compounds thereof are tested simultaneously and compared to evaluate their metabolic stability in human and/or mouse liver microsomes. Compared with the non-deuterated compound Binimetinib, the compound of the invention has longer half-life T 1/2 And lower clearance CL int The metabolic stability can be obviously improved. The results for the compounds of the representative examples are summarized in tables 1 and 2.
Table 1:
Figure BDA0003692806340000392
Figure BDA0003692806340000401
table 2:
Figure BDA0003692806340000402
(3) 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 standard chow 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. The tubes were oven dried at 60 ℃ overnight before use. After completion of blood collection at the last time point, the 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 compared with the non-deuterated compound Binimetinib, the compound has better pharmacokinetic property in animals, thereby having better pharmacodynamics and therapeutic 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, numerous simple deductions or substitutions may be made without departing from the spirit of the invention, which shall be deemed to belong to the scope of the invention.

Claims (11)

1. A compound of formula (I), or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof:
Figure FDA0003692806330000011
wherein the content of the first and second substances,
Y 1 、Y 2 、Y 3 、Y 4 and Y 5 Each independently selected from hydrogen, deuterium or halogen;
R 1 、R 2 、R 3 and R 4 Each independently selected from hydrogen or deuterium;
each X is independently selected from CH 3 、CD 3 、CHD 2 Or CH 2 D;
With the proviso that the above-mentioned compounds contain at least one deuterium atom.
2. The compound of claim 1, which is a compound of formula (II):
Figure FDA0003692806330000012
wherein, Y 1 、R 1 、R 2 、R 3 、R 4 And X is as defined in claim 1;
or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof.
3. The compound of claim 1 or 2, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein Y is 1 Is deuterium.
4. The compound of any one of claims 1-3, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein R is 1 And R 2 Is deuterium.
5. The compound of any one of claims 1-4, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein R is 3 And R 4 Is deuterium.
6. The compound of any one of claims 1-5, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein X is CD 3
7. The compound of claim 1, selected from compounds of the formula:
Figure FDA0003692806330000021
Figure FDA0003692806330000031
Figure FDA0003692806330000041
Figure FDA0003692806330000051
Figure FDA0003692806330000061
or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof.
8. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of any one of claims 1-7, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof.
9. The pharmaceutical composition of claim 8, further comprising an additional therapeutic agent.
10. The pharmaceutical composition according to claim 9, wherein the additional therapeutic agent is selected from one or more of BRAF inhibitors, EGFR antibodies, immune checkpoint inhibitors or CDK4/6 inhibitors;
preferably, the BRAF inhibitor is selected from vemurafenib (vemurafenib), dabrafenib (dabrafenib), canofenib (encorafenib), (S) -methyl- (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate, (S) - (methyl-d 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1-isopropyl-1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate, (S) - (methyl-d 3 ) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl) carbamate, (S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5 ) Methyl carbamate, (S) - (1- ((4- (3- (5-chloro-2-fluoro-3- (methylsulfonylamino) phenyl) -1- (propan-2-yl-d) 7 ) -1H-pyrazol-4-yl) pyrimidin-2-yl) amino) propan-2-yl-1, 3-d 5) methyl carbamate;
preferably, the EGFR inhibitor is selected from gefitinib (gefitinib), erlotinib (erlotinib), afatinib (afatinib), dacomitinib (dacomitinib), lapatinib (lapatinib), oxitinib (osimertinib), amitinib, vementib, CO-1686, wz4002, pd035, pf00299804;
preferably, the EGFR antibody is selected from cetuximab (cetuximab), panitumumab (panitumumab), cetuximab ((Necitumumab);
preferably, the immune checkpoint inhibitor is selected from the group consisting of pertuzumab (pembrolizumab), yiprimumab (ipilimumab), and nivolumab (nivolumab), atezumab (atezolizumab), avizumab (avelumab), delazumab (durvalumab), pidilizumab (pidilizumab);
preferably, the CDK4/6 inhibitor is selected from pabociclib (palbociclib), ribbociclib (ribociclib), abercinib (abemacciclib).
11. Use of a compound of any one of claims 1-7, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of any one of claims 8-10, in the manufacture of a medicament for the treatment of a MEK kinase mediated disease;
preferably, the MEK kinase mediated disease is selected from melanoma, acute myeloid leukemia, glioma, neurofibroma, non-small cell lung cancer, breast cancer, serous cancer, gastrointestinal stromal tumor, lung non-squamous carcinoma, colorectal cancer, biliary tract cancer, myeloma;
preferably, the melanoma is selected from BRAF V600 mutant melanoma;
preferably, the colorectal cancer is selected from BRAF V600 mutant colorectal cancer;
preferably, the neurofibroma is selected from neurofibromatosis type 1 or plexiform neurofibromas;
preferably, the BRAF V600 mutation is selected from BRAF V600E or BRAF V600K mutation.
CN202210665562.2A 2021-06-17 2022-06-14 Substituted benzimidazoles, compositions containing them and their use Pending CN115490640A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110671907 2021-06-17
CN2021106719070 2021-06-17

Publications (1)

Publication Number Publication Date
CN115490640A true CN115490640A (en) 2022-12-20

Family

ID=84464364

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210665562.2A Pending CN115490640A (en) 2021-06-17 2022-06-14 Substituted benzimidazoles, compositions containing them and their use

Country Status (2)

Country Link
CN (1) CN115490640A (en)
WO (1) WO2022262699A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1652776A (en) * 2002-03-13 2005-08-10 阵列生物制药公司 N3 alkylated benzimidazole derivatives as MEK inhibitors
CN1874768A (en) * 2003-08-29 2006-12-06 阵列生物制药公司 N3 alkylated benzimidazole derivatives as MEK inhibitors
CN104447701A (en) * 2013-09-17 2015-03-25 广东东阳光药业有限公司 Pyrazole derivative and application thereof
CN106029074A (en) * 2013-12-12 2016-10-12 诺华股份有限公司 Combinations of trametinib, panitumumab and dabrafenib for the treatment of cancer
CN111620821A (en) * 2019-02-27 2020-09-04 深圳市塔吉瑞生物医药有限公司 Substituted pyrazole compounds, compositions containing the same and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1652776A (en) * 2002-03-13 2005-08-10 阵列生物制药公司 N3 alkylated benzimidazole derivatives as MEK inhibitors
CN1874768A (en) * 2003-08-29 2006-12-06 阵列生物制药公司 N3 alkylated benzimidazole derivatives as MEK inhibitors
CN104447701A (en) * 2013-09-17 2015-03-25 广东东阳光药业有限公司 Pyrazole derivative and application thereof
CN106029074A (en) * 2013-12-12 2016-10-12 诺华股份有限公司 Combinations of trametinib, panitumumab and dabrafenib for the treatment of cancer
CN111620821A (en) * 2019-02-27 2020-09-04 深圳市塔吉瑞生物医药有限公司 Substituted pyrazole compounds, compositions containing the same and uses thereof

Also Published As

Publication number Publication date
WO2022262699A1 (en) 2022-12-22

Similar Documents

Publication Publication Date Title
EP3519398B1 (en) Pyridine compound
JP7347853B2 (en) Substituted pyrazolo[1,5-a]pyridine compounds, compositions containing the compounds and uses thereof
US11459334B2 (en) Substituted pyrrolo[2,1-f][1,2,4]triazines as KIT and/or PDGFR-α inhibitors
BR112015002709B1 (en) COMPOUNDS, PHARMACEUTICAL COMPOSITION, USE THEREOF AND USE OF A COMPOUND COMBINATION OR PHARMACEUTICAL COMPOSITION AND A SECOND PROPHYLATIC OR THERAPEUTIC AGENT
EP2949647A1 (en) Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing same
CN109851638B (en) Substituted diaminopyrimidine compounds
KR20200096261A (en) Sulfonamide compounds and uses thereof
EP3845538B1 (en) Dihydroimidazopyrazinone compound, composition including same, and use thereof
CN108947974B (en) Aminopyrimidine compound, composition containing aminopyrimidine compound and application of aminopyrimidine compound
EP3865488A1 (en) Macrocyclic compound as cdk inhibitor, preparation method therefor, and use thereof in medicine
CN110066272B (en) Substituted benzo [ d ] imidazoles and pharmaceutical compositions thereof
CN109111439B (en) Amide compound, composition containing same and application thereof
JP2022521964A (en) New pan-RAF kinase inhibitor and its use
CN109438362B (en) Substituted benzimidazole compound and composition containing same
WO2022242712A1 (en) Substituted macrocyclic compound, composition comprising compound, and use thereof
CN115490640A (en) Substituted benzimidazoles, compositions containing them and their use
CN110256408B (en) Diaryl pyrazole compound, composition containing compound and application of diaryl pyrazole compound
CN117083265A (en) Kinase inhibitors and uses thereof
CN115701429B (en) 4- (1H-indol-1-yl) pyrimidine-2-amino derivative, and preparation method and application thereof
CN111138426B (en) Indazole kinase inhibitor and application thereof
WO2020168963A1 (en) Substituted fused aromatic ring derivative, composition and use thereof

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