[go: up one dir, main page]

CN115490640B - Substituted benzimidazole compounds, compositions containing the same and uses thereof - Google Patents

Substituted benzimidazole compounds, compositions containing the same and uses thereof Download PDF

Info

Publication number
CN115490640B
CN115490640B CN202210665562.2A CN202210665562A CN115490640B CN 115490640 B CN115490640 B CN 115490640B CN 202210665562 A CN202210665562 A CN 202210665562A CN 115490640 B CN115490640 B CN 115490640B
Authority
CN
China
Prior art keywords
compound
pharmaceutically acceptable
deuterium
acceptable salt
propan
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202210665562.2A
Other languages
Chinese (zh)
Other versions
CN115490640A (en
Inventor
王义汉
赵九洋
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Publication of CN115490640A publication Critical patent/CN115490640A/en
Application granted granted Critical
Publication of CN115490640B publication Critical patent/CN115490640B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明提供了一种取代的苯并咪唑类化合物及包含该化合物的组合物及其用途,所述的取代的苯并咪唑类化合物如式(I)所示化合物或其互变异构体、立体异构体、前药、晶型、药学上可接受的盐、水合物或溶剂化合物。式(I)化合物可作为有效的MEK1/2抑制剂,其可通过抑制活化的MEK抑制ERK磷酸化,对多种癌症模型表现出广泛的抗肿瘤活性,同时,本发明化合物联合其他抗肿瘤治疗剂可以更好地治疗多种癌症。除了抑制作用和效力外,本发明化合物还显示出更好地代谢稳定性和/或药代动力学性能。 The present invention provides a substituted benzimidazole compound and a composition comprising the compound and its use. The substituted benzimidazole compound is a compound shown in formula (I) or its tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate or solvent compound. The compound of formula (I) can be used as an effective MEK1/2 inhibitor, which can inhibit ERK phosphorylation by inhibiting activated MEK, and exhibits a wide range of anti-tumor activity in various cancer models. At the same time, the compound of the present invention can better treat various cancers in combination with other anti-tumor therapeutic agents. In addition to the inhibitory effect and efficacy, the compound of the present invention also shows better metabolic stability and/or pharmacokinetic performance.

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 thereof. More particularly, the present invention relates to certain deuterium-substituted compounds of 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-N- (2-hydroxyethoxy) -1-methyl-1H-benzimidazole-6-carboxamide and its derivatives, and tautomers, stereoisomers, prodrugs, crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof. These deuterium-substituted compounds and compositions thereof are useful as potent and selective inhibitors of MEK1 and MEK2 proteins, and are useful in the treatment of diseases caused by MEK kinase, and these deuterium-substituted compounds 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 RAS GTPASE (gtpase) proteins in response to growth factors, hormones, cytokines, etc., stimulates phosphorylation and activation of RAF kinase. These kinases then phosphorylate and activate the intracellular protein kinases MEK1 and MEK2, which in turn 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 essential function of this pathway is to link receptor activity at the cell membrane to modifications of cytoplasmic or nuclear targeting that control cell proliferation, differentiation and survival.
The structural activation of this pathway is sufficient to induce cellular transformation. Deregulated activation of the MAPK pathway by 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. In human malignancies, RAS mutations are common and have been identified in approximately 30% of cancers. The RAS family of gtpase proteins (proteins that convert guanosine triphosphate to guanosine diphosphate) transmits signals from activated growth factor receptors to downstream intracellular counterparts. Among the targets complemented by active membrane-bound RAS, an important target is the RAF family of serine/threonine protein kinases. The RAF family consists of three related kinases (A-, B-and C-RAF) that act as downstream effectors of the RAS. RAS mediated RAF activation also initiates 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 position 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 cancers, in cancer and other proliferative diseases, the kinase components of the signaling cascade are incorporated as potentially important targets for regulating disease progression.
MEK1 and MKE2 are members of a larger family of dual-specific kinases that phosphorylate threonine and tyrosine disabilities of various MAPKs. MEK1 and MEK2 have unique gene codes, but they share a high degree of homology (80%) within the C-terminal catalytic kinase domain and most of the N-terminal regulatory region. Oncogene 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. In addition to RAF, MEK may also be activated by other oncogenes. 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 aberrant matrix specificity places MEK1 and MEK2 at critical points in the signaling cascade, which would enable it to integrate many extracellular signals into the MAPK pathway.
The fundamental role and role 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 for model organisms. RAF may have a prominent role in the formation of certain tumors, for example, the activating allele of BRAF has been identified in-70% melanoma, 40% papillary thyroid carcinoma, 30% low grade ovarian carcinoma, and 10% colorectal carcinoma. Most BRAF mutations are found in the kinase domain, with a single substitution (V600E) of at least 80%. The 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, having the following structural formula) is a potent, non-ATP-competitive, highly selective MEK1/2 inhibitor developed by Array BioPharma that can inhibit MEK, ERK phosphorylation and the growth of BRAF or KRAS mutant cancer cells at nanomolar concentrations. The U.S. Food and Drug Administration (FDA) awards Binimetinib orphan status in 11 months 2013 and approves its combination with BRAF inhibitor Encorafenib in 6 months 2018 for the treatment of metastatic or unresectable melanoma patients with the BRAF V600E/K mutation. It has now been shown that the use of BRAF inhibitors in combination with MEK inhibitors can increase efficacy and potentially reduce toxic effects. In the united states national integrated cancer network (National Comprehensive Cancer Network, NCCN) guidelines, BRAF inhibitor/MEK inhibitor combination immunotherapy is recommended as a first line therapy for metastatic or unresectable melanoma.
Poor absorption, distribution, metabolism and/or excretion (ADME) properties are known to be the leading cause of failure in many candidate drug clinical trials. Many drugs currently on the market also limit their range of application due to poor ADME properties. Rapid metabolism of drugs can result in many drugs that would otherwise be effective in treating the disease being difficult to formulate due to too rapid clearance from the body's metabolism. Frequent or high dose administration, while potentially solving the problem of rapid drug clearance, can lead to problems such as poor patient compliance, side effects caused by high dose administration, and increased cost of treatment. In addition, rapidly metabolized drugs may also expose the patient to poorly toxic or reactive metabolites.
The discovery of novel potent highly selective MEK1/2 inhibitors that have good oral bioavailability and are pharmaceutically acceptable is also a challenging task. 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 for various cancer models, including melanoma, acute myeloid leukemia, glioma, neurofibromatosis, 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 treat a variety of cancers in combination with other anti-tumor therapeutic agents. In addition to inhibition and potency, the compounds of the invention also show good solubility and better metabolic stability and/or pharmacokinetic properties.
In this regard, the present invention adopts the following technical scheme:
In a first aspect of the invention, there is provided a compound of formula (I):
Wherein,
Y 1、Y2、Y3、Y4 and Y 5 are each independently selected from hydrogen, deuterium or halogen;
R 1、R2、R3 and R 4 are each independently selected from hydrogen or deuterium;
Each X is independently selected from CH 3、CD3、CHD2 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, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, and a pharmaceutically acceptable excipient. In a specific embodiment, the compounds of the present invention are provided in the pharmaceutical composition in an effective amount. 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 specific 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 particular embodiments, the BRAF inhibitor is selected from the group consisting of vitamin Mo Feini (vemurafenib), dabrafenib (dabrafenib), kang Naifei ni (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) 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 specific embodiments, the EGFR inhibitor is selected from gefitinib (gefitinib), erlotinib (erlotinib), afatinib (afatinib), dacatinib (dacomitinib), lapatinib (lapatinib), octtinib (osimertinib), ametinib, fu Mei tinib, CO-1686, wz4002, pd153035, pf00299804. In specific embodiments, the EGFR antibody is selected from cetuximab (cetuximab), panitumumab (panitumumab), and cetuximab ((Necitumumab). In specific embodiments, the immune checkpoint inhibitor is selected from the group consisting of pamglizumab (pembrolizumab), ipilimumab mother (ipilimumab) and nivolumab (nivolumab), alemtuzumab (atezolizumab), avermectin (avelumab), dewaruzumab (durvalumab), and plalimumab (pidilzumab). In a specific embodiment, the CDK4/6 inhibitor is selected from the group consisting of palbociclib (palbociclib), rebabociclib (ribociclib), abbe cilib (abemaciclib).
In another aspect, the present invention provides a method of preparing a pharmaceutical composition as described above, comprising the steps of: a pharmaceutically acceptable excipient is admixed with a compound of the invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, thereby forming a pharmaceutical composition.
In another aspect, the invention 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 invention, or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention.
In specific embodiments, the MEK kinase mediated disease is melanoma, acute myeloid leukemia, glioma, neurofibromatosis, 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 mutated melanoma. In specific embodiments, the colorectal cancer is selected from the group consisting of BRAF V600 mutated colorectal cancers. In specific embodiments, the BRAF V600 mutation is selected from a BRAF V600E mutation or a BRAF V600K mutation. In specific embodiments, the neurofibromatosis 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 detailed description, examples and claims that follow.
Definition of the definition
Herein, "deuterated" refers to a compound or group in which one or more hydrogens are replaced with deuterium, unless otherwise indicated; deuteration may be mono-, di-, poly-or full-substituted. The term "one or more deuterated" is used interchangeably with "one or more deuterated".
Herein, unless otherwise specified, "non-deuterated compound" refers to a compound having a deuterium atom content of 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., male or female of any age group, e.g., pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young adults, middle aged adults, or senior adults)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., cynomolgus monkey, rhesus monkey), cow, pig, horse, sheep, goat, rodent, cat, and/or 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 indicated, the term "treating" includes an effect that occurs when a subject suffers from 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"), as well as an effect that occurs before the subject begins to suffer from the particular disease, disorder or condition ("prophylactic treatment").
In general, an "effective amount" of a compound refers to 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 the compounds of the present invention may vary depending on the following factors: for example, biological targets, pharmacokinetics of the compound, the disease being treated, the mode of administration, and the age health and symptoms of the subject. Effective amounts include therapeutically and prophylactically therapeutically effective amounts.
As used herein, unless otherwise indicated, 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" may include an amount that improves 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 indicated, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or to prevent 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 used alone or in combination with other agents, which provides a prophylactic benefit in preventing a disease, disorder or condition. The term "prophylactically effective amount" may 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, the compounds of the invention may be administered simultaneously or sequentially in separate unit dosage forms with another therapeutic agent, or simultaneously in a single unit dosage form with another therapeutic agent.
Detailed Description
Compounds of formula (I)
Herein, "the compound of the present invention" refers to the compounds of the following formulas (I) and (II) or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvent compound thereof.
In one embodiment, the present invention relates to compounds of formula (I):
Wherein,
Y 1、Y2、Y3、Y4 and Y 5 are each independently selected from hydrogen, deuterium or halogen;
R 1、R2、R3 and R 4 are each independently selected from hydrogen or deuterium;
X is selected from CH 3、CD3、CHD2 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 specific embodiment, the deuterium isotope content of deuterium at the deuterated position is at least greater than 0.015%, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99% of the natural deuterium isotope content.
Specifically, the deuterium isotope content of each deuterated position of Y 1、Y2、Y3、Y4、Y5、R1、R2、R3、R4 and X in the present invention is at least greater than 0.015%, more 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, more preferably two deuterium atoms, more preferably three deuterium atoms, more preferably four deuterium atoms, more preferably five deuterium atoms, more preferably six deuterium atoms, more preferably seven deuterium atoms, more preferably eight deuterium atoms, more preferably nine deuterium atoms, more preferably ten deuterium atoms, more preferably eleven deuterium atoms, more preferably twelve deuterium atoms.
In another specific embodiment, "Y 1、Y2、Y3、Y4 and Y 5 are each independently selected from hydrogen, deuterium, or halogen" includes schemes where Y 1 is selected from hydrogen, deuterium, or halogen, Y 2 is selected from hydrogen, deuterium, or halogen, Y 3 is selected from hydrogen, deuterium, or halogen, and so on, until Y 5 is selected from hydrogen, deuterium, or halogen. More specifically, include the technical schemes that 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 until Y 5 is hydrogen, Y 5 is deuterium or Y 5 is halogen (F, cl, br or I).
In another specific embodiment, "R 1、R2、R3 and R 4 are each independently selected from hydrogen or deuterium" includes embodiments in which R 1 is selected from hydrogen or deuterium, R 2 is selected from hydrogen or deuterium, R 3 is selected from hydrogen or deuterium, and R 4 is selected from hydrogen or deuterium. More specifically, the technical scheme that 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 deuterium is included.
In another specific embodiment, "X is selected from CH 3、CD3、CHD2 or CH 2 D" includes the technical scheme that X is CH 3, X is CD 3, X is CHD 2, or X is CH 2 D.
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 solvent compound thereof, wherein X is CD 3,Y1、Y2、Y3、Y4、Y5、R1、R2、R3 and R 4 are 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 solvent compound thereof, wherein R 1 and R 2 are deuterium, Y 1、Y2、Y3、Y4、Y5、R3、R4 and X are 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 solvent compound thereof, wherein R 3 and R 4 are deuterium, Y 1、Y2、Y3、Y4、Y5、R1、R2 and X are 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 solvent compound thereof, wherein R 1、R2、R3 and R 4 are deuterium, Y 1、Y2、Y3、Y4、Y5 and X are 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 solvent compound thereof, wherein Y 1 is deuterium, Y 2、Y3、Y4、Y5、R1、R2、R3、R4 and X are 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 solvent compound thereof, wherein X is CD 3,Y1 is deuterium, Y 2、Y3、Y4、Y5、R1、R2、R3 and R 4 are 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 solvent compound thereof, wherein X is CD 3,R1 and R 2 are deuterium, and Y 1、Y2、Y3、Y4、Y5、R3 and R 4 are 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 solvent compound thereof, wherein X is CD 3,R3 and R 4 are deuterium, and Y 1、Y2、Y3、Y4、Y5、R1 and R 2 are 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 solvent compound thereof, wherein X is CD 3,R1、R2、R3 and R 4 are deuterium, and Y 1、Y2、Y3、Y4 and Y 5 are 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 solvent compound thereof, wherein Y 1、R1 and R 2 are deuterium, and Y 2、Y3、Y4、Y5、R3、R4 and X are 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 solvent compound thereof, wherein Y 1、R3 and R 4 are deuterium, and Y 2、Y3、Y4、Y5、R1、R2 and X are 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 solvent compound thereof, wherein Y 1、R1、R2、R3 and R 4 are deuterium, and Y 2、Y3、Y4、Y5 and X are 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 solvent compound thereof, wherein X is CD 3,Y1、R1 and R 2 are deuterium, and Y 2、Y3、Y4、Y5、R3 and R 4 are 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 solvent compound thereof, wherein X is CD 3,Y1、R3 and R 4 are deuterium, and Y 2、Y3、Y4、Y5、R1 and R 2 are 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 solvent compound thereof, wherein X is CD 3,Y1、R1、R2、R3 and R 4 are deuterium, and Y 2、Y3、Y4 and Y 5 are as defined above.
In another embodiment, the invention relates to a compound of formula (II):
Wherein,
Y 1 is selected from hydrogen, deuterium, or halogen;
R 1、R2、R3 and R 4 are each independently selected from hydrogen or deuterium;
X is selected from CH 3、CD3、CHD2 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 compound, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvent compound thereof, wherein X is CD 3,Y1、R1、R2、R3 and R 4 are 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 solvent compound thereof, wherein R 1 and R 2 are deuterium, Y 1、R3、R4 and X are 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 solvent compound thereof, wherein R 3 and R 4 are deuterium, Y 1、R1、R2 and X are 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 solvent compound thereof, wherein R 1、R2、R3 and R 4 are deuterium, Y 1 and X are 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 solvent compound thereof, wherein Y 1 is deuterium, R 1、R2、R3、R4 and X are 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 solvent compound thereof, wherein X is CD 3,Y1 is deuterium, and R 1、R2、R3 and R 4 are 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 solvent compound thereof, wherein X is CD 3,R1 and R 2 are deuterium, and Y 1、R3、R4 and Y 5 are 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 solvent compound thereof, wherein X is CD 3,R3 and R 4 are deuterium, and Y 1、R1 and R 2 are 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 solvent compound thereof, wherein X is CD 3,R1、R2、R3 and R 4 is deuterium, and Y 1 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 solvent compound thereof, wherein Y 1、R1 and R 2 are deuterium, and R 3、R4 and X are 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 solvent compound thereof, wherein Y 1、R3 and R 4 are deuterium, and R 1、R2 and X are 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 solvent compound thereof, wherein Y 1、R1、R2、R3 and R 4 are deuterium, 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 solvent compound thereof, wherein X is CD 3,Y1、R1 and R 2 are deuterium, and R 3 and R 4 are 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 solvent compound thereof, wherein X is CD 3,Y1、R3 and R 4 are deuterium, and R 1 and R 2 are 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 solvent compound thereof, wherein X is CD 3,Y1、R1、R2、R3 and R 4 is deuterium.
As a preferred embodiment of the present invention, the compound, or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate, or solvate thereof, is selected from any one of the following compounds:
The compounds of the invention may include one or more asymmetric centers and thus may exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be individual enantiomers, diastereomers, or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. The isomers may be separated from the mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC), formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
Those skilled in the art will appreciate that the organic compound may form a complex with a solvent in or from 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 bound to 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, diethyl ether, and the like. The compounds described herein may be prepared, for example, in crystalline form, and may be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates 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 both solvates in solution and separable solvates. Representative solvates include hydrates, ethanolates and methanolates.
The term "hydrate" refers to a compound that binds to water. Generally, 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, the hydrates of the compounds may be represented by, for example, the general formula R.xH 2 O, where 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 hydrate (x is a number greater than 0 and less than 1, e.g., hemihydrate (r.0.5H 2 O)), and polyhydrate (x is a number greater than 1, e.g., dihydrate (r.2h 2 O) and hexahydrate (r.6h 2 O)).
The compounds of the present 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 (or salt, hydrate or solvate thereof) of a compound of a particular crystal stacking 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 shapes, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors can lead to a crystalline form predominating. Various polymorphs of a compound can be prepared by crystallization under different conditions.
The invention also includes isotopically-labeled compounds, which are identical to those of the compounds of 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 compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F and 36 Cl, respectively. The compounds of the invention, prodrugs thereof, and pharmaceutically acceptable salts of the compounds or prodrugs thereof, which contain the isotopes described above and/or other isotopes of other atoms, are within the scope of this invention. Certain isotopically-labeled compounds of the present invention, for example those into which radioactive isotopes (e.g., 3 H and 14 C) are introduced, are useful 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 they are easy to prepare and detect. Further, substitution with heavier isotopes, such as deuterium, i.e., 2 H, may be preferred in some circumstances because greater metabolic stability may afford therapeutic benefits such as increased in vivo half-life or reduced dosage requirements. Isotopically-labeled compounds of formula (I) of the present invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes and/or 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, e.g. in blood, into its active form having a medical effect. Pharmaceutically acceptable prodrugs are described in t.higuchi and v.stilla, prodrugs as Novel DELIVERY SYSTEMS, A.C.S.SYMPOSIUM Series Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987, and d.fleisher, s.ramon and H.Barbra"Improved oral drug delivery:solubility limitations overcome by the use of prodrugs",Advanced Drug Delivery Reviews(1996)19(2)115-130, each of which are incorporated herein by reference.
Prodrugs are any covalently bonded compounds of the invention which, when administered to a patient, release the parent compound in vivo. Prodrugs are typically prepared by modifying functional groups in such a way that the modification may be performed by conventional procedures or cleavage in vivo to yield the parent compound. Prodrugs include, for example, compounds of the invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, may cleave to form the hydroxy, amino, or sulfhydryl group. Representative examples of prodrugs therefore include, but are not limited to, acetate, formate and benzoate/amide derivatives of hydroxy, mercapto and amino functional groups of compounds of formula (I). In addition, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like can be used. The esters themselves may be active and/or may be hydrolysed under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those groups which readily decompose in the human body to release the parent acid or salt thereof.
Process for preparing compounds of the invention
The compounds of the present 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 those skilled in the art of organic synthesis. Suitable solvents may be substantially unreactive with the starting materials (reactants), intermediates, or products at the temperature at which the reaction is carried out (e.g., at a temperature in the range of the solvent freezing temperature to the solvent boiling temperature). The given reaction may be carried out in one solvent or a mixture of more than one solvent. The skilled artisan 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 protection and deprotection of different chemical groups. One skilled in the art can readily determine whether protection is desired and removal of the protection and selection of the appropriate protecting group. The chemical nature of the 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 individual stereoisomers thereof by reacting a racemic mixture of the compounds with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereoisomers and recovering the optically pure enantiomer. Enantiomeric resolution may be carried out using diastereomeric derivatives of the compounds of the invention, preferably dissociable complexes (e.g., crystalline diastereomeric salts). Diastereomers have significantly different physical properties (e.g., melting point, boiling point, solubility, reactivity, etc.), and can be readily separated by the advantages of these dissimilarities. Diastereomers may be separated by chromatography, preferably by separation/resolution techniques based on differences in solubility. The optically pure enantiomer is then recovered by any practical means that does not racemize, along with the resolving agent. A more detailed description of techniques suitable for resolution of stereoisomers of compounds starting from racemic mixtures can be found in Jean Jacques, andre Collet, samue h.wilen, "enantiomers, racemates and resolution" ("Enantiomers, RACEMATES AND resolution"), john Wiley And Sons, inc.
The reaction may be monitored according to any suitable method known in the art. For example, product formation may be monitored by spectroscopic means, such as Nuclear Magnetic Resonance (NMR) spectroscopy (e.g., 1 H or 13 C), infrared (IR) spectroscopy, spectrophotometry (e.g., UV-visible), 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 invention provides a pharmaceutical composition comprising a compound of the invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of a compound of the present 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 co-formulated compounds. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and lanolin.
The invention also includes kits (e.g., pharmaceutical packages). Kits provided can include a compound of the invention, other therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispersible packages or other suitable containers) containing a compound of the invention, other therapeutic agent. In some embodiments, the provided kits may also optionally include a third container containing pharmaceutically acceptable excipients for diluting or suspending the compounds of the invention and/or other therapeutic agents. In some embodiments, the compounds of the invention and other therapeutic agents provided in the first and second containers are combined to form one unit dosage form.
The following formulation examples illustrate representative pharmaceutical compositions that may be prepared according to the present invention. However, the present invention is not limited to the following pharmaceutical compositions.
Exemplary formulation 1-tablet: the compounds of the present invention in dry powder form may be mixed with a dry gel binder in a weight ratio of about 1:2. A lesser amount of magnesium stearate was added as a lubricant. The mixture is formed into tablets of 0.3-30mg (each tablet containing 0.1-10mg of active compound) in a tablet press.
Exemplary formulation 2-tablet: the compounds of the present invention in dry powder form may be mixed with a dry gel binder in a weight ratio of about 1:2. A lesser amount of magnesium stearate was added as a lubricant. The mixture is formed into tablets of 30-90mg (each tablet containing 10-30mg of active compound) in a tablet press.
Exemplary formulation 3-tablet: the compounds of the present invention in dry powder form may be mixed with a dry gel binder in a weight ratio of about 1:2. A lesser amount of magnesium stearate was added as a lubricant. The mixture is formed into tablets of 90-150mg (each tablet containing 30-50mg of active compound) in a tablet press.
Exemplary formulation 4-tablet: the compounds of the present invention in dry powder form may be mixed with a dry gel binder in a weight ratio of about 1:2. A lesser amount of magnesium stearate was added as a lubricant. The mixture is formed into 150-240mg tablets (each tablet containing 50-80mg of active compound) in a tablet press.
Exemplary formulation 5-tablet: the compounds of the present invention in dry powder form may be mixed with a dry gel binder in a weight ratio of about 1:2. A lesser amount of magnesium stearate was added as a lubricant. The mixture is formed into 240-270mg tablets (each tablet containing 80-90mg of active compound) in a tablet press.
Exemplary formulation 6-tablet: the compounds of the present invention in dry powder form may be mixed with a dry gel binder in a weight ratio of about 1:2. A lesser amount of magnesium stearate was added as a lubricant. The mixture is formed into 270-450mg tablets (each tablet containing 90-150mg of active compound) in a tablet press.
Exemplary formulation 7-tablet: the compounds of the present invention in dry powder form may be mixed with a dry gel binder in a weight ratio of about 1:2. A lesser amount of magnesium stearate was added as a lubricant. The mixture is formed into 450-900mg tablets (each tablet containing 150-300mg of active compound) in a tablet press.
Exemplary formulation 8-capsule: the compounds of the present invention in dry powder form may be mixed with the starch diluent in a weight ratio of about 1:1. The mixture was filled into 250mg capsules (each capsule containing 125mg of active compound).
Exemplary formulation 9-liquid: the compound of the present invention (125 mg) may be mixed with sucrose (1.75 g) and xanthan gum (4 mg), and the resulting mixture may be blended, passed through a No.10 mesh U.S. sieve, and then mixed with an aqueous solution of microcrystalline cellulose and sodium carboxymethylcellulose (11:89, 50 mg) prepared in advance. 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 buffered sterile saline injectable aqueous medium to a concentration of about 5 mg/mL.
Administration of drugs
The pharmaceutical compositions provided herein may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implantation or other means of administration. For example, the number of the cells to be processed, parenteral administration as used herein includes subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intraarterial, and the like synovial cavity administration, sternal administration, cerebrospinal membrane administration, intralesional administration, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of the compound actually administered may be determined by a physician, according to the circumstances involved, including the condition being treated, the route of administration selected, 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 disorder of the present invention, a subject at risk of developing the disorder is administered a compound provided herein, typically based on physician recommendations and administered under the supervision of a physician, at a dosage level as described above. Subjects at risk for developing a particular disorder generally include subjects having a family history of the disorder, or those subjects determined by genetic testing or screening to be particularly susceptible to developing the disorder.
The pharmaceutical compositions provided herein may also be administered chronically ("chronically"). Chronic administration refers to administration of a compound or pharmaceutical composition thereof over a prolonged period of time, e.g., 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue administration 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 a 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. Bolus doses depend on the targeted systemic level of active ingredient through the body, e.g., intramuscular or subcutaneous bolus doses cause slow release of the active ingredient, whereas bolus injections delivered directly to veins (e.g., by IV intravenous drip) can be delivered more rapidly, causing the concentration of the active ingredient in the blood to rise rapidly to effective levels. In other embodiments, the pharmaceutical composition may be administered in the form of a continuous infusion, for example, by IV intravenous drip, thereby providing a steady state concentration of the active ingredient in the subject's body. 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 in order to facilitate 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 suitable for producing the desired therapeutic effect in association with a suitable pharmaceutical excipient. Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of liquid compositions, or in the case of solid compositions, pills, tablets, capsules and the like. In such compositions, the compound is typically a minor component (about 0.1 to about 50 wt.%, or preferably about 1 to about 40 wt.%) with the remainder being various carriers or excipients and processing aids useful for forming the desired administration form.
For oral doses, a typical regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses. Using these modes of dosing, each dose provides from about 0.01 to about 20mg/kg of a compound of the invention, with preferred doses each providing from about 0.1 to about 10mg/kg, especially from about 1 to about 5mg/kg.
In order to provide similar blood levels to, or lower than, the use of an injected dose, a transdermal dose is typically selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 10% by weight, and more preferably 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 achieve adequate steady state levels, a preloaded 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, buffers, suspending and dispersing agents, colorants, flavors, and the like. Solid forms may include, for example, any of the following components, or compounds having similar properties: binders, 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, for example, colloidal silicon dioxide; sweeteners, 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 injectable use, 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, the remainder being an injectable excipient or the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as ointments, the active ingredients are typically combined with a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with, for example, an oil-in-water cream base. 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 provided by the present invention.
The compounds of the invention may also be administered via a transdermal device. Transdermal administration may thus be achieved using a reservoir (reservoir) or porous membrane type, or a variety of solid matrix patches.
The above components of the compositions for oral administration, injection or topical administration are merely representative. Other materials and processing techniques, etc. are set forth in Remington's Pharmaceutical Sciences, part 8 of 17th edition,1985,Mack Publishing Company,Easton,Pennsylvania, 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, optionally including one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated and sulfoalkyl ether substitutions. In some embodiments, the cyclodextrin is a sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, for example, U.S.5,376,645. In some embodiments, the formulation comprises hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Indication of disease
The present invention provides a method of treating a disease, such as MEK kinase mediated disease, in a subject comprising administering to the subject a compound of the invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention. The methods of treatment may also be combined with other therapies, such as radiation therapy, chemotherapy.
In specific embodiments, MEK kinase mediated diseases include inflammatory diseases, infections, autoimmune disorders, stroke, ischemia, heart disorders, neurological disorders, fibrotic disorders, proliferative disorders, hyperproliferative disorders, tumors, leukemias, neoplasms, cancers, malignant tumors, metabolic diseases, and malignant diseases.
The invention also provides a method of treating a MEK kinase mediated inflammatory disease comprising administering to the subject a compound of the invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention.
In specific embodiments, the MEK kinase mediated inflammatory disease comprises rheumatoid arthritis or multiple sclerosis.
The invention also provides a method of treating a MEK kinase-mediated proliferative disease comprising administering to said subject a compound of the invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention.
In specific embodiments, MEK kinase mediated proliferative diseases include cancer, psoriasis, restenosis, autoimmune diseases or atherosclerosis.
The invention also provides a method of treating a MEK kinase mediated cancer comprising administering to the subject a compound of the invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention.
In specific embodiments, MEK kinase mediated cancers include melanoma (e.g., BRAF V600 mutated melanoma), acute myeloid leukemia, glial flow, neurofibromatosis (e.g., neurofibromatosis type 1 (NF 1) or plexiform neurofibromatosis), non-small cell lung cancer, breast cancer, serous cancer, gastrointestinal stromal tumor, lung non-squamous carcinoma, colorectal cancer (e.g., BRAF V600 mutated colorectal cancer), biliary tract cancer, myeloma.
Inhibitors of MEK kinases are described for the treatment of diseases driven by over-activation, aberrant activation, constitutive activation, mutations that gain function of MEK kinase and/or substrate kinase including but not limited to ERK. Such diseases encompass hyperproliferative diseases and, including but not limited to psoriasis, keloids, hyperplasia of the skin, benign Prostatic Hyperplasia (BPH), solid tumors such as the respiratory tract (including but not limited to small cell and non-small cell lung cancer), brain (including but not limited to glioma, neurofibroma, plexiform neurofibromas, medulloblastoma, ependymoma, neuroectodermal and pineal tumor), breast (including but not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma), reproductive organs (including but not limited to prostate carcinoma, testicular carcinoma, ovarian carcinoma, endometrial carcinoma, cervical carcinoma, vaginal carcinoma, vulval carcinoma, and uterine sarcoma), digestive tract (including but not limited to esophagus colon, colorectal, stomach, gall bladder, pancreas, rectum, anus, small intestine, and salivary gland cancers), urinary tract (including but not limited to bladder, ureter, kidney, urinary tract, and renal papillary cancers), eye (including but not limited to intraocular melanoma and retinoblastoma), liver (including but not limited to hepatocellular carcinoma and cholangiocarcinoma), skin (including but not limited to melanoma, squamous cell carcinoma, kaposi's sarcoma, merkel cell skin carcinoma, non-melanoma skin carcinoma), head and neck (including but not limited to laryngeal, nasopharyngeal, hypopharynx, oropharyngeal cancer, lip and oral cancer, and squamous cell carcinoma), thyroid, parathyroid cancer, and metastatic tumors thereof. Hyperproliferative diseases also include leukemias (including but not limited to acute lymphoblastic leukemia, acute follow-up leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, and hairy cell leukemia), sarcomas (including but not limited to soft tissue sarcomas, osteosarcomas, lymphosarcomas, rhabdomyosarcomas), and lymphomas (including but not limited to non-hodgkin's lymphoma, AIDS-related lymphomas, cutaneous T-cell lymphomas, 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 aberrant regulation of mitogenic 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 the treatment of diseases and disorders associated with aberrant, abnormal and/or excessive angiogenesis. Such angiogenesis-related disorders 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 invention also provides a method of treating acute myeloid leukemia comprising administering to said subject a compound of the invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention. In specific embodiments, the acute myeloid leukemia is relapsed and/or refractory acute myeloid leukemia.
The invention also provides a method of treating glioma comprising administering to said subject a compound of the invention or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention.
The invention also provides a method of treating neurofibromatosis comprising administering to the subject a compound of the invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention. In specific embodiments, the neurofibromatosis is selected from neurofibromatosis type 1 (NF 1) or plexiform neurofibromas.
The invention also provides a method of treating serous cancer comprising administering to the subject a compound of the invention, or a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate, or solvate thereof, or a pharmaceutical composition of the invention. In specific embodiments, the serous cancer is selected from recurrent or persistent low grade ovarian, fallopian tube, or primary peritoneal serous cancer.
Combination therapy
The compounds of the present invention may be used alone or in combination with other therapeutic agents. Combination therapy according to the invention thus comprises administration of at least one compound of the invention and use of at least one other pharmaceutically active agent. One or more of the 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 performed simultaneously or sequentially in any order. The amounts and relative timing of administration of one or more compounds of the invention and one or more other pharmaceutically active agents will be selected to achieve the desired combined therapeutic effect. Specifically:
The present invention provides a method of treating BRAF kinase mediated cancer comprising administering to said subject a compound of the invention and a BRAF inhibitor (each optionally in a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate or solvate), and optionally a third therapeutic agent, in combination.
In specific embodiments, the BRAF inhibitor is selected from vitamin Mo Feini (vemurafenib), dabrafenib (dabrafenib), kang Naifei ni (encorafenib) or the following compounds disclosed in WO 2020/01141 A1:
Compounds disclosed in WO 2020/011111 A1
Preferably, the BRAF inhibitor is selected from the group consisting of vitamin Mo Feini (vemurafenib), dabrafenib (dabrafenib), kang Naifei ni (encorafenib), and the following compounds disclosed in WO 2020/011115 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) carbamic acid methyl ester,
(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) carbamic acid methyl ester,
(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) carbamic acid methyl ester,
(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) carbamic acid methyl ester,
(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 the group consisting of vitamin Mo Feini (vemurafenib), dabrafenib (dabrafenib), kang Naifei ni (encorafenib), and the following compounds disclosed in WO 2020/011115 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- (propan-2-yl) -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) carbamic acid methyl ester.
In particular embodiments, the method of treating BRAF kinase mediated cancer does not comprise a third therapeutic agent.
In particular embodiments, the method of treating BRAF kinase mediated cancer comprises a third therapeutic agent. In specific embodiments, the third therapeutic agent is selected from immune checkpoint inhibitors, e.g., pamglizumab (pembrolizumab), ipili mother (ipilimumab) and nano-arm mab (nivolumab), alemtuzumab (atezolizumab), avilamab (avelumab), dewaruzumab (durvalumab), plamizumab (pidilzumab), PDR-001 (BAP 049-clone-E, disclosed and used in WO 2017/019896); preferably, for example, palbociclizumab (pembrolizumab), iprolipram mother (ipilimumab) and a device. In specific embodiments, the third therapeutic agent is selected from EGFR antibodies, e.g., cetuximab (cetuximab), panitumumab (panitumumab), cetuximab ((Necitumumab)), preferably, e.g., cetuximab (cetuximab). In specific embodiments, the third therapeutic agent is a mitotic inhibitor, e.g., a CDK4/6 inhibitor, preferably, e.g., palbociclib (palbociclib), rebabociclib (ribociclib), abb (abemaciclib), preferably, e.g., palbociclib (palbociclib).
In particular embodiments, the BRAF kinase mediated cancer is melanoma, brain tumor 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 a specific embodiment, the BRAF V600 mutation is BRAF V600E and BRAF V600K. In a specific embodiment, the BRAF kinase mediated cancer is metastatic or unresectable melanoma of the BRAF V600 mutation. In a specific embodiment, the BRAF kinase mediated cancer is metastatic or unresectable melanoma of a BRAF V600E or BRAF V600K mutation. In a specific embodiment, the BRAF kinase mediated cancer is colorectal cancer with a BRAF V600 mutation. In a specific embodiment, the BRAF kinase mediated cancer is colorectal cancer of the BRAF V600E or BRAF V600K mutation.
The invention also provides a method of treating cancer with NRAS or KRAS or EGFR mutations comprising administering to the subject a compound of the invention and an EGFR inhibitor (each optionally in the form of a tautomer, stereoisomer, prodrug, crystal, pharmaceutically acceptable salt, hydrate, or solvate) in combination. In specific embodiments, the EGFR inhibitor is selected from gefitinib (gefitinib), erlotinib (erlotinib), afatinib (afatinib), dactinib (dacomitinib), lapatinib (lapatinib), octtinib (osimertinib), ametinib, vomitinib, CO-1686, WZ4002, PD153035, PF00299804, cetuximab, panitumumab, rituximab. 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 E63K, G12V, G12R, G12A, G12D, G S and G12C, or an increase in NRAS gene copy number.
The 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 mFOLFIRI (each optionally in a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate or solvate).
The invention also provides a method of treating gastrointestinal stromal tumors comprising administering to said 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 invention also provides a method of treating gastrointestinal stromal tumors comprising administering to said subject a compound of the invention and imatinib (imatinib) (each optionally in the form of a tautomer, stereoisomer, prodrug, crystalline form, pharmaceutically acceptable salt, hydrate or solvate).
The invention also provides a method of treating non-squamous cancers of the lung comprising administering to said subject a compound of the invention in combination with carboplatin and pemetrexed (each optionally in a tautomer, stereoisomer, prodrug, crystal, pharmaceutically acceptable salt, hydrate, or solvate).
The invention also provides a method of treating biliary tract cancer comprising administering to said subject a compound of the invention in combination with capecitabine (capecitabine) (each optionally in the form of a tautomer, stereoisomer, prodrug, crystal, pharmaceutically acceptable salt, hydrate, or solvate).
Examples
The invention will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention. The experimental methods, in which specific conditions are not noted in the following examples, are generally conducted under conventional conditions or under conditions recommended by the manufacturer. Parts and percentages are parts by weight 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: para-toluene sulfonic acid
EDCI: 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride
HOBT: 1-hydroxybenzotriazole
DMAP: dimethylaminopyridine
TEA: triethylamine
DIEA: n, N-diisopropylethylamine
TFAA: trifluoroacetic anhydride
Na 2CO3: sodium carbonate
K 2CO3: potassium carbonate
Cs 2CO3: cesium carbonate
EtOH: ethanol
BnOH: benzyl alcohol
DCM: dichloromethane (dichloromethane)
THF: tetrahydrofuran (THF)
ACN: acetonitrile
DME: ethylene glycol dimethyl ether
DMF: n, N-dimethylformamide
DMSO: dimethyl sulfoxide
TMSCL: trimethylchlorosilane
Preparation of intermediate A-15- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d 3) -1H-benzimidazole-6-carboxylic acid
The following synthetic route was adopted
Step 1: synthesis of 2,3, 4-trifluoro-5-nitrobenzoic acid compound
2,3, 4-Trifluorobenzoic acid (20 g,113.6 mmol) was dissolved in 60ml of concentrated sulfuric acid, the reaction solution was heated to 90 ℃, then a mixed solution of concentrated sulfuric acid (12 g,122.4 mmol) and concentrated nitric acid (12.8 g,132.1 mmol) was added dropwise, the reaction was stirred for 5 hours, the TLC monitored the reaction was completed, cooled to room temperature, the reaction solution was slowly added dropwise to ice water, extraction was performed 3-4 times with ethyl acetate, the organic phase was combined, the saturated brine was washed 2-3 times, dried over anhydrous sodium sulfate, and after filtration concentration, a white solid was obtained, 24.0g, the yield was 95.6%. LC-MS (APCI) M/z=220.1 (M-1) -.
Step 2: synthesis of 2, 3-difluoro-4-amino-5-nitrobenzoic acid compound
2,3, 4-Trifluoro-5-nitrobenzoic acid (5.81 g,26.3 mmol) obtained in the previous step is placed in a 100ml flask, 30ml deionized water is added, the temperature of the mixture is reduced to 0 ℃ in an ice bath, concentrated ammonia water (18.4 g,131.5 mmol) is slowly added dropwise, the mixture is heated to room temperature and stirred for reaction overnight after the addition, TLC monitors that the reaction is finished, the pH value is regulated to be less than 2 in the ice bath by 1N dilute hydrochloric acid, light yellow solid is separated out, 4.99g of product is obtained after filtration and vacuum drying is carried out, and the yield is 87.1%. LC-MS (APCI): M/z= 219.4 (m+1) +.
Step 3: synthesis of 2, 3-difluoro-4-amino-5-nitrobenzoic acid methyl ester
2, 3-Difluoro-4-amino-5-nitrobenzoic acid (4.99 g,22.9 mmol) and trimethylchlorosilane (4.97 g,45.8 mmol) were added to the reaction flask, 100ml methanol was added to dissolve, the reaction mixture was heated to 65℃under nitrogen protection and stirred overnight, TLC monitoring was completed, cooled to room temperature, concentrated and separated by a silica gel column to give 4.51g of yellow solid with a yield of 84.9%. LC-MS (APCI): M/z= 233.4 (m+1) +.
Step 4: synthesis of 2, 4-diamino-3-fluoro-5-nitrobenzoic acid methyl ester
Methyl 2, 3-difluoro-4-amino-5-nitrobenzoate (1.0 g,4.3 mmol) obtained in the previous step was dissolved in 10ml dioxane, ammonia (1.6 ml,21.4 mmol) was added, and the reaction was carried out at 90℃for 2-4 hours under nitrogen protection, and TLC monitoring the reaction was completed. The reaction solution was cooled to room temperature, diluted with 30ml of water, extracted with ethyl acetate 3-4 times, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and dried to give 1.03g of a yellow solid, which was directly put into the next reaction. LC-MS (APCI): M/z=230.8 (m+1) +.
Step 5: synthesis of compound methyl 2,4, 5-triamino-3-fluorobenzoate
Methyl 2, 4-diamino-3-fluoro-5-nitrobenzoate (1.03 g,4.5 mmol), reduced iron powder (2.5 g,45 mmol) and anhydrous ammonium chloride (1.44 g,26.9 mmol) obtained in the previous step were placed in a 100ml flask, 15ml ethanol and 5ml water were added, and the mixture was heated to 70℃and stirred for 1-2 hours, after which the reaction was completed by TLC monitoring. The catalyst was removed by celite assist filtration and the filtrate concentrated to give 860mg of grey solid which was directly put into the next reaction without purification. LC-MS (APCI): M/z=200.1 (m+1) +.
Step 6: synthesis of methyl 4-fluoro-5-amino-1H-benzimidazole-6-carboxylate
Methyl 2,4, 5-triamino-3-fluorobenzoate (860 mg,4.32 mmol) and formamidine acetate (552 mg,5.21 mmol) obtained in the previous step were added to 10ml of absolute ethanol, and the temperature was raised to 80℃for reflux reaction for 4 hours, and TLC was monitored to complete the reaction. The reaction solution was cooled to room temperature, concentrated to remove the solvent, diluted with 20ml of water, extracted 3-4 times with dichloromethane, the organic phases were combined, concentrated and purified by silica gel column chromatography to give 486mg of pale yellow solid, yield: 53.8%. LC-MS (APCI): M/z=210.5 (m+1) +.
Step 7: synthesis of Compound 4-fluoro-5-amino-1- (methyl-d 3) -1H-benzimidazole-6-carboxylic acid methyl ester
Methyl 4-fluoro-5-amino-1H-benzimidazole-6-carboxylate (600 mg,2.86 mmol), deuterated iodomethane (458 mg,3.16 mmol) and potassium carbonate (794 mg,5.76 mmol) obtained in the previous step are added into a 50ml reaction bottle, 10ml anhydrous DMF is added, the temperature is raised to 70 ℃ under the protection of nitrogen, the mixture is stirred for 2 hours, and TLC monitors the completion of the reaction. After cooling to room temperature, 30ml of water was added for dilution, extraction was performed 3 to 4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 201mg of brown solid in 31.5% yield. LC-MS (APCI): M/z= 227.5 (m+1) +.
Step 8: synthesis of Compound 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d 3) -1H-benzimidazole-6-carboxylic acid methyl ester
Methyl 4-fluoro-5-amino-1- (methyl-d 3) -1H-benzimidazole-6-carboxylate (200 mg,0.9 mmol), 2-fluoro-4-bromo-1-iodobenzene (300 mg,0.99 mmol), pd 2(dba)3 (16 mg,0.017 mmol), xantphos (26 mg,0.044 mmol) and cesium carbonate (284 mg,1.79 mmol) obtained in the above step were added to a 20ml microwave tube, 8ml ethylene glycol dimethyl ether was added under nitrogen protection, the reaction was carried out by microwave heating to 90℃after sealing for 1 hour, TLC monitoring was completed, the solvent was concentrated and removed after cooling to room temperature, and silica gel column chromatography was purified to obtain 229mg of a earthy yellow solid, yield: 64.0%. LC-MS (APCI): M/z= 399.2 (m+1) +.
Step 9: synthesis of intermediate A-1
5- ((4-Bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d 3) -1H-benzimidazole-6-carboxylic acid methyl ester (1.63 g,4.09 mmol) is added into a 100ml reaction bottle, 30ml tetrahydrofuran and 10ml water are added for dissolution, sodium hydroxide (0.68 g,17.0 mmol) is added, the mixture is heated to 45 ℃ for stirring reaction for 3-5 hours, after TLC monitoring reaction is finished, tetrahydrofuran is concentrated and removed, 10ml water is added for dilution, 1N diluted hydrochloric acid is used for regulating pH to acidity, off-white solid is separated out, 1.3g crude product is obtained after filtration and vacuum drying, and the crude product is directly put into the next reaction. LC-MS (APCI): M/z=385.2 (m+1) +.
Intermediate A-25- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-carboxylic acid methyl ester preparation
The following synthetic route was adopted
Step 1: synthesis of Compound methyl 4-fluoro-5-amino-1-methyl-1H-benzimidazole-6-carboxylate
4-Fluoro-5-amino-1H-benzimidazole-6-carboxylic acid methyl ester (600 mg,2.86 mmol), methyl iodide (457 mg,3.16 mmol) and potassium carbonate (794 mg,5.76 mmol) were added to a 50ml reaction flask, 10ml anhydrous DMF was added, and the temperature was raised to 70℃under nitrogen protection, stirring was carried out for 2 hours, and TLC was monitored to complete the reaction. After cooling to room temperature, 30ml of water was added for dilution, extraction was performed 3 to 4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 203mg of brown solid in 31.9% yield. LC-MS (APCI): M/z=224.5 (m+1) +.
Step 2: synthesis of Compound 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-carboxylic acid methyl ester
Methyl 4-fluoro-5-amino-1-methyl-1H-benzimidazole-6-carboxylate (200 mg,0.9 mmol), 2-fluoro-4-bromo-1-iodobenzene (300 mg,0.99 mmol), pd2 (dba) 3 (16 mg,0.017 mmol), xantphos (26 mg,0.044 mmol) and cesium carbonate (284 mg,1.79 mmol) obtained in the above steps were added to a20 ml microwave tube, 8ml ethylene glycol dimethyl ether was added under nitrogen protection, the reaction was carried out for 1 hour by heating to 90℃with microwaves after sealing, after TLC monitoring, the solvent was removed by concentration after cooling to room temperature, and silica gel column chromatography was purified to obtain 208mg of a yellowish brown solid, yield: 58.1%. LC-MS (APCI): M/z=396.2 (m+1) +.
Step 3: synthesis of intermediate A-2
5- ((4-Bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-methyl formate (1.63 g,4.09 mmol) was added into a 100ml reaction flask, 30ml tetrahydrofuran and 10ml water were added for dissolution, sodium hydroxide (0.68 g,17.0 mmol) was further added, the mixture was heated to 45 ℃ and stirred for 3-5 hours, after the completion of the reaction by TLC monitoring, tetrahydrofuran was concentrated and removed, 10ml water was added for dilution, pH was adjusted to acidity with 1N diluted hydrochloric acid, off-white solid was precipitated, 1.3g crude product was obtained by vacuum drying after filtration, and was directly put into the next reaction. LC-MS (APCI): M/z= 382.1 (m+1) +.
Intermediate B-12- (aminooxy) -2, 2-dideutero benzyl acetate preparation
The following synthetic route was adopted
Step 1: synthesis of 2-bromo-2, 2-dideuteroacetic acid
Deuterated acetic acid (10 g,156 mmol) was added to 80ml trifluoroacetic anhydride, bromine dioxane complex (37 g,149 mmol) was added at room temperature, the reaction was stirred overnight under nitrogen protection, the reaction was monitored by GC, 10.8g oily liquid was obtained after concentrating under reduced pressure, and the oily liquid was directly put into the next reaction without purification.
Step 2: synthesis of 2-bromo-2, 2-dideutero benzyl acetate
2-Bromo-2, 2-dideuteroacetic acid (10.8 g,78.55 mmol), benzyl alcohol (8.5 g,78.6 mmol), EDCI (16.6 g,86.6 mmol) and DMAP (1.06 g,8.7 mmol) were dissolved in 80ml anhydrous DMF and stirred at room temperature under nitrogen for 3-5 hours, and TLC monitoring was complete. 200ml of water was added for dilution, extraction was performed 3 to 4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to obtain 7.5g of an anhydrous oily liquid in 21% yield in two steps. LC-MS (APCI): M/z= 280.3 (m+1) +.
Step 3: synthesis of Compound 2- ((1, 3-dioxoisoindolin-2-yl) oxy) -2, 2-dideuteroacetic acid benzyl ester
Benzyl 2-bromo-2, 2-dideuteroacetate (3.94 g,17.3 mmol) obtained in the above step, N-hydroxyphthalimide (2.51 g,15.38 mmol) and triethylamine (2.36 g,23.3 mmol) were added to 40ml of anhydrous DMF and stirred at room temperature under nitrogen atmosphere overnight. 150ml of water was added for dilution, extraction was performed 3 to 4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to give 4.36g of a white solid in 81% yield. LC-MS (APCI): M/z=314.4 (m+1) +.
Step 4: synthesis of intermediate B-1
Benzyl 2- ((1, 3-dioxoisoindolin-2-yl) oxy) -2, 2-dideuteroacetate (4.36 g,14.0 mmol) and hydrazine hydrate (1.0 g,20.0 mmol) obtained in the previous step were dissolved in 45ml dichloromethane, stirred at room temperature and reacted overnight, and TLC monitoring was completed. Insoluble matter was removed by filtration, and the filtrate was concentrated and separated by a silica gel column to give 1.99g of a yellow oily liquid, yield 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-benzimidazole-6-carboxamide (compound T-1) preparation
The following synthetic route was adopted
Step 1: synthesis of Compound 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d 3) -N- (2- (ethyleneoxy) ethoxy) -1H-benzimidazole-6-carboxamide
Intermediate A-1 (1.3 g,3.4 mmol), O- (2- (ethyleneoxy) ethyl) hydroxylamine (0.41 g,4.02 mmol), EDCI (0.78 g,4.06 mmol), HOBT (0.55 g,4.08 mmol) and triethylamine (0.41 g,4.06 mmol) were added to a 100ml reaction flask, 15ml anhydrous DMF was added under nitrogen protection, the reaction was stirred overnight at room temperature, TLC was monitored, 50ml water was added to dilute the mixture, extraction 3-4 times with ethyl acetate, the organic phase was combined, washed with saturated brine, and column chromatography on silica gel was performed after concentration to give 0.88g of off-white solid, yield :55.3%.LC-MS(APCI):m/z=470.5(M+1)+.1H NMR(400MHz,DMSO-d6):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).
Step 2: synthesis of Compound T-1
Dissolving 5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1- (methyl-d 3) -N- (2- (ethyleneoxy) ethoxy) -1H-benzimidazole-6-carboxamide (0.88 g,1.88 mmol) obtained in the previous step in 15ml ethanol, cooling to 0 ℃ in an ice bath, slowly dropwise adding 2N dilute hydrochloric acid (9.4 ml,18.8 mmol), heating to room temperature, stirring for 2-4 hours, monitoring the reaction completion by TLC, adjusting pH to alkalescence by using 1N sodium hydroxide aqueous solution, concentrating to remove the solvent, purifying by silica gel column chromatography to obtain 0.42g white solid, obtaining the yield :50.5%.LC-MS(APCI):m/z=444.7(M+1)+.1H NMR(400MHz,CD3OD):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-methyl-1H-benzimidazole-6-carboxamide (Compound T-2)
The following synthetic route was adopted
Step 1: synthesis of benzyl 2- ((5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzoimidazole-6-formylamino) oxy) -2, 2-dideuterio acetate
Intermediate A-2 (1.3 g,3.4 mmol), intermediate B-1 (0.74 g,4.02 mmol), EDCI (0.78 g,4.06 mmol), HOBT (0.55 g,4.08 mmol) and triethylamine (0.41 g,4.06 mmol) were added to a 100ml reaction flask, 15ml anhydrous DMF was added under nitrogen protection, the reaction was stirred overnight at room temperature, TLC monitored for completion of the reaction, 50ml water was added for dilution, extraction 3-4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, and silica gel column chromatography purification after concentration gave 1.78g of off-white solid, yield :95.7%.LC-MS(APCI):m/z=547.3(M+1)+.1H NMR(400MHz,DMSO-d6):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).
Step 2: synthesis of Compound T-2
Dissolving 2- ((5- ((4-bromo-2-fluorophenyl) amino) -4-fluoro-1-methyl-1H-benzimidazole-6-formylamino) oxy) -2, 2-dideuteric benzyl acetate (1.78 g,3.25 mmol) obtained in the previous step in 40ml anhydrous THF, cooling to-10 ℃ under nitrogen protection, adding deuterated lithium aluminum hydride (273 mg,6.5 mmol) in batches, continuing stirring at low temperature for 3-4 hours, monitoring the reaction by TLC, adding 5ml water for quenching reaction at low temperature, adding 10ml of 15% sodium hydroxide aqueous solution, diatomite for assisting filtration, concentrating the filtrate, and purifying by silica gel column chromatography to obtain off-white solid 0.47g, yield :32.6%.LC-MS(APCI):m/z=445.6(M+1)+.1H NMR(400MHz,CD3OD):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).
Biological activity testing.
(1) Cytotoxicity test
The inhibitory effect of the compounds of the examples on HT-29 cell activity was examined.
Cell line: HT-29 (cell type: adherent; cell number/well: 3000; culture medium: RPMI-1640+10% FBS; culture) was carried out at 37℃under 5% CO2 and 95% humidity.
Consumable and reagent: fetal bovine serum FBS (GBICO, cat#10099-141),Luminescent Cell Viability Assay (Promega, cat#G7572), 96-well transparent flat bottom black wall plate @Cat#3603)。
Instrument: spectraMax multi-label microplate detector, MD,2104-0010A; CO2 incubator Thermo Scientific, model 3100Series; a biosafety cabinet Thermo Scientific, model 1300series A2; inverted microscope, olympus, CKX41SF; refrigerator, SIEMENS, KK25E76TI.
The experimental steps are as follows:
1) Cell culture and seeding: i) Cells in the logarithmic growth phase were harvested and counted using a platelet counter. Detecting the cell activity by trypan blue exclusion method, and ensuring the cell activity to be more than 90%; ii) adjusting the cell concentration; add 90. Mu.L of cell suspension to 96-well plates, respectively; iii) Cells in 96-well plates were incubated overnight at 37 ℃, 5% co2, 95% humidity.
2) Drug dilution and dosing: i) Preparing 10 times of medicine solution, wherein the highest concentration is 100 mu M,9 concentrations and 3.16 times of dilution, adding 10 mu L of medicine solution into each well of a 96-well plate inoculated with cells, and setting three compound wells for each medicine concentration; ii) cells in the dosed 96-well plates were incubated at 37℃under 5% CO2 at 95% humidity for a further 72 hours before CTG analysis.
3) End point reading plate: i) Thawing CTG reagent and equilibration of cell plates to room temperature for 30 min; ii) adding an equal volume of CTG solution per well; iii) Vibrating on an orbital shaker for 5 minutes to lyse cells; iv) the cell plates were left at room temperature for 20 minutes to stabilize the luminescence signal; v) reading the luminescence value.
And (3) data processing: the data were analyzed using GRAPHPAD PRISM 5.0.0 software, and a non-linear S-curve regression was used to fit the data to a dose-response curve, and IC50 values were calculated therefrom. Cell viability (%) = (Lum test drug-Lum broth control)/(Lum cell control-Lum broth control) ×100%.
The compounds of the present invention were tested in the cytotoxicity experiments described above, and the results showed that: the compounds of the invention have a more potent activity on HT-29 cells than non-deuterated compounds Binimetinib.
(2) Evaluation of Metabolic stability
Metabolic stability is generally used to describe the rate and extent to which a compound is metabolized and is one of the primary factors affecting pharmacokinetic properties. Many compounds are substrates for CYP450 enzymes and other drug metabolizing enzymes, and liver microsomes are CYP 450-rich systems, and the aim of this experiment was to conduct in vitro stability studies by incubating the compounds of the invention with human and/or mouse liver microsomes, respectively, and detecting the remaining proportion of the compounds using LC-MS/MS.
① Preparation of the solution
Phosphate Buffer (PBS): 150mL of a pre-prepared KH 2PO4 (0.5M) solution and 700mL of a K 2HPO4 (0.5M) solution were mixed, the pH of the mixture was adjusted to 7.4 with a K 2HPO4 (0.5M) solution, and the mixture was stored at 4℃as 5-fold concentration PBS for use. Before use, the solution was diluted 5-fold with ultrapure water, and 3.3mM magnesium chloride was added to obtain phosphate buffer PBS (100 mM).
NADPH regeneration system solution: NADPH solution containing 6.5mM NADP,16.5mM G-6-P,3U/mL G-6-P D was formulated with 5mL PBS.
Internal standard stop solution: propranolol hydrochloride (50 ng/mL) and tolbutamide (200 ng/mL) were prepared with acetonitrile as internal standard working solutions.
Human liver microsomal solution: 0.31mL of human liver microsome (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 microsome (25 mg/mL) was added to 0.961mL of PBS (pH 7.4) and mixed to obtain a dilution of mouse liver microsome with a protein concentration of 0.625 mg/mL.
Sample working solution: the compound of the present invention and non-deuterated compound powder, positive control dextromethorphan powder and omeprazole powder were formulated with DMSO to 10mM as sample stock solutions. Then, the mixture was diluted with 70% acetonitrile-water to obtain a 0.25mM sample working solution.
② Sample incubation
398 Μl of human liver microsome dilution was added to 96 Kong Fuyo plates (n=2), and 2 μl of 0.25mM test compound and dextromethorphan were added, respectively, and mixed well.
Mu.L of mouse liver microsome dilution was added to 96 Kong Fuyo plates (N=2), and 2. Mu.L of 0.25mM test compound and dextromethorphan were added, respectively, and mixed well.
Each well was filled with 300. Mu.L of pre-chilled stop solution into a 96-well deep well plate and placed on ice as a stop plate.
The 96 Kong Fuyo plates and NADPH regeneration system were placed in a 37℃water bath, shaken at 100 revolutions per minute, and pre-incubated for 5min. 80. Mu.L of the incubation solution was removed from each well of the incubation plate, added to the termination plate, mixed well, and supplemented with 20. Mu.L of NADPH regeneration system solution as a 0min sample. Then 80. Mu.L of NADPH regeneration system solution was added to each well of the incubation plate, the reaction was started and timing was started. The reaction concentration of the compound to be tested is 1 mu M, and the protein concentration is 0.5mg/mL.
100. Mu.L of each reaction solution was added to the termination plate at 10, 30 and 90min, and the reaction was terminated by vortexing for 3 min.
The final plates were centrifuged at 5000rpm at 4℃for 15min. 200. Mu.L of the supernatant was mixed with a 96-well plate to which 200. Mu.L of ultrapure water had been added in advance, and the mixture was analyzed by LC-MS/MS to obtain a sample of 10. Mu.L.
③ Sample analysis method
The LC-MS/MS system is used in the experiment to detect the peak areas of the compound to be detected, dextromethorphan, omeprazole and an internal standard, and the ratio of the peak areas 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 single-exponential degradation model of GRAPHPAD PRISM 7.0.0 software is used for plotting the residual amount (R%) of the compound with time to obtain a substrate elimination rate constant K
Ct/C0=exp(-K*t)
And half-life T 1/2 and intrinsic clearance CL int were calculated according to the following formulas, where V/M is equal to 1/C (protein).
T1/2(min);CLint(μL/min/mg)。
Experimental results: the compounds of the present invention and their non-deuterated compounds were simultaneously tested and compared to evaluate their metabolic stability in human and/or mouse liver microsomes. Compared with the non-deuterated compound Binimetinib, the compound provided by the invention has longer half-life T 1/2 and lower clearance CL int, and can obviously improve metabolic stability. The results of the compounds of the tabular examples are summarized in tables 1 and 2.
Table 1:
Table 2:
(3) Rat pharmacokinetic experiments
6 Male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 animals each, and their pharmacokinetic differences were compared by intravenous or oral administration of a single dose of the compound (oral administration 10 mg/kg).
Rats were fed with standard feed and given water. Fasted food was started 16 hours prior to the trial. The drug was dissolved with PEG400 and dimethylsulfoxide. The eyebox was sampled at 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours and 24 hours post-administration.
Rats were briefly anesthetized after inhalation of diethyl ether and 300 μl of blood was collected from the orbit in a tube. There was 30. Mu.L of 1% heparin salt solution in the tube. Before use, the tube was baked overnight at 60 ℃. After blood collection was completed at the last time point, rats were sacrificed after ether anesthesia.
Immediately after blood sample collection, the tube was gently inverted at least 5 times, ensuring that the mix was well placed on ice. The blood sample was centrifuged at 5000rpm at 4℃for 5 minutes to separate the plasma from the erythrocytes. 100. Mu.L of plasma was aspirated with a pipette into a clean plastic centrifuge tube, indicating the name and time point of the compound. Plasma was stored at-80 ℃ prior to analysis. The concentration of the compounds of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the blood concentration of each animal at different time points.
Experiments show that compared with the non-deuterated compound Binimetinib, the compound provided by the invention has better pharmacokinetic properties in animals, thus having better pharmacodynamics and treatment effects.
The foregoing is a further detailed description of the invention in connection with the preferred embodiments, and it is not intended that the invention be limited to the specific embodiments described. It will be apparent to those skilled in the art that several simple deductions or substitutions may be made without departing from the spirit of the invention, and these should be considered to be within the scope of the invention.

Claims (20)

1. A compound of formula (II), or a pharmaceutically acceptable salt thereof:
Wherein,
Y 1 is hydrogen;
R 1、R2、R3 and R 4 are each independently selected from hydrogen or deuterium;
X is selected from CH 3 or CD 3;
With the proviso that X is CD 3 when R 1 and R 2 are simultaneously deuterium, and/or R 3 and R 4 are simultaneously deuterium, or R 1、R2、R3 and R 4 are simultaneously hydrogen.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 1 and R 2 are deuterium.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R 3 and R 4 are deuterium.
4. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R 3 and R 4 are deuterium.
5. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein X is CD 3.
6. The compound of claim 1, selected from the group consisting of compounds of the formula:
Or a pharmaceutically acceptable salt thereof.
7. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof.
8. The pharmaceutical composition of claim 7, further comprising an additional therapeutic agent.
9. The pharmaceutical composition of claim 8, wherein 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.
10. The pharmaceutical composition of claim 9, wherein the BRAF inhibitor is selected from the group consisting of vitamin Mo Feini, dabrafenib, kang Naifei ni, (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) 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) carbamic acid methyl ester.
11. The pharmaceutical composition of claim 9, wherein the EGFR inhibitor is selected from gefitinib, erlotinib, afatinib, dacatinib, lapatinib, octyitinib, ametinib, vomertinib, CO-1686, WZ4002, PD153035, PF00299804.
12. The pharmaceutical composition of claim 9, wherein the EGFR antibody is selected from cetuximab, panitumumab, and rituximab-resistant.
13. The pharmaceutical composition of claim 9, wherein the immune checkpoint inhibitor is selected from the group consisting of palbociclib, mopril mother, warrior, alemtuzumab, avermectin, dewaruzumab, and pioglitazone.
14. The pharmaceutical composition according to claim 9, wherein the CDK4/6 inhibitor is selected from the group consisting of palbociclib, rebabociclib, abbe-cilib.
15. Use of a compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 7-14, in the manufacture of a medicament for treating a MEK kinase-mediated disease.
16. The use of claim 15, wherein the MEK kinase-mediated disease is selected from melanoma, acute myeloid leukemia, glioma, neurofibromatosis, non-small cell lung cancer, breast cancer, serous cancer, gastrointestinal stromal tumor, lung non-squamous carcinoma, colorectal cancer, biliary tract cancer, myeloma.
17. The use of claim 16, wherein the melanoma is selected from BRAF V600 mutated melanoma.
18. The use according to claim 16, wherein colorectal cancer is selected from BRAF V600 mutant colorectal cancers.
19. The use according to claim 16, wherein the neurofibromatosis is selected from neurofibromatosis type 1 or plexiform neurofibromas.
20. The use of claim 17 or 18, wherein the BRAF V600 mutation is selected from a BRAF V600E or BRAF V600K mutation.
CN202210665562.2A 2021-06-17 2022-06-14 Substituted benzimidazole compounds, compositions containing the same and uses thereof Active CN115490640B (en)

Applications Claiming Priority (2)

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

Publications (2)

Publication Number Publication Date
CN115490640A CN115490640A (en) 2022-12-20
CN115490640B true CN115490640B (en) 2024-11-26

Family

ID=84464364

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210665562.2A Active CN115490640B (en) 2021-06-17 2022-06-14 Substituted benzimidazole compounds, compositions containing the same and uses thereof

Country Status (2)

Country Link
CN (1) CN115490640B (en)
WO (1) WO2022262699A1 (en)

Citations (2)

* 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

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104447701B (en) * 2013-09-17 2019-03-22 广东东阳光药业有限公司 Pyrazole derivatives and application thereof
WO2015087279A1 (en) * 2013-12-12 2015-06-18 Glaxosmithkline Intellectual Property (No.2) Limited Combinations of trametinib, panitumumab and dabrafenib for the treatment of cancer
CN111620821B (en) * 2019-02-27 2022-02-11 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 (2)

* 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

Also Published As

Publication number Publication date
WO2022262699A1 (en) 2022-12-22
CN115490640A (en) 2022-12-20

Similar Documents

Publication Publication Date Title
ES2833576T3 (en) Novel glutaminase inhibitors
US9604935B2 (en) Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing the same
CN115836070B (en) Fused ring compounds as EGFR inhibitors, preparation method and application thereof
EP3878852A1 (en) Substituted pyrazolo[1,5-a]pyridine compound, composition containing the same and use thereof
CN109970745A (en) Substituted pyrrolo-triazine class compound and its medical composition and its use
CN112789279B (en) Inhibitor containing tricyclic derivatives, preparation method and application thereof
US11254696B2 (en) Dianilinopyrimidine compound for inhibiting kinase activity
EP3845538B1 (en) Dihydroimidazopyrazinone compound, composition including same, and use thereof
KR20200096261A (en) Sulfonamide compounds and uses thereof
CN111518082B (en) Aminopyrimidine compound, composition containing aminopyrimidine compound and application of aminopyrimidine compound
CN115490640B (en) Substituted benzimidazole compounds, compositions containing the same and uses thereof
US12116348B2 (en) Substituted benzimidazole compound and composition comprising same
WO2020168963A1 (en) Substituted fused aromatic ring derivative, composition and use thereof
WO2023078267A1 (en) Amino-containing macrocyclic compound as protein kinase modulator
JP2022521964A (en) New pan-RAF kinase inhibitor and its use
WO2023109751A1 (en) Pyrimidine or pyridine derivative and medicinal use thereof
US12076320B2 (en) Diarylpyrazole compound, composition comprising same, and use thereof
CN116102545B (en) A diaryl urea PI3K/mTOR/HDAC multi-target inhibitor and its pharmaceutical composition and application
JP7710223B2 (en) Substituted pyrrolotriazine compounds and pharmaceutical compositions thereof and their uses
WO2025093058A1 (en) 2-oxoindoline derivatives, preparation method therefor and use thereof
JP2024531717A (en) Polymorphs of pyrimidine derivatives and pharma-ceutically acceptable salts thereof and uses 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
GR01 Patent grant
GR01 Patent grant