CN116969920A - Fused pyridazine derivative, preparation method and medical application thereof - Google Patents

Fused pyridazine derivative, preparation method and medical application thereof Download PDF

Info

Publication number
CN116969920A
CN116969920A CN202310479336.XA CN202310479336A CN116969920A CN 116969920 A CN116969920 A CN 116969920A CN 202310479336 A CN202310479336 A CN 202310479336A CN 116969920 A CN116969920 A CN 116969920A
Authority
CN
China
Prior art keywords
alkyl
halogen
mmol
compound
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202310479336.XA
Other languages
Chinese (zh)
Inventor
程耀邦
余发志
王永辉
陈伟
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.)
Shanghai Huiqi Biomedical Technology Co ltd
Original Assignee
Shanghai Huiqi Biomedical Technology Co ltd
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 Shanghai Huiqi Biomedical Technology Co ltd filed Critical Shanghai Huiqi Biomedical Technology Co ltd
Publication of CN116969920A publication Critical patent/CN116969920A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • C07D237/30Phthalazines
    • C07D237/34Phthalazines with nitrogen atoms directly attached to carbon atoms of the nitrogen-containing ring, e.g. hydrazine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D453/00Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids
    • C07D453/02Heterocyclic compounds containing quinuclidine or iso-quinuclidine ring systems, e.g. quinine alkaloids containing not further condensed quinuclidine ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Abstract

The invention relates to the field of pharmaceutical chemistry, in particular to a group of condensed pyridazine derivatives shown in a formula (I),Processes for their preparation, pharmaceutical compositions containing them and their use for the treatment or prophylaxis of NLRP3 inflammatory smallscale associated diseases.

Description

Fused pyridazine derivative, preparation method and medical application thereof
Technical Field
The present invention relates to the field of pharmaceutical chemistry, in particular to a group of fused pyridazine derivative chemical entities (e.g. compounds or isomers thereof, stable isotopic variants, pharmaceutically acceptable salts or solvates), pharmaceutical compositions comprising the same, methods for their preparation, methods for the treatment or prophylaxis of related diseases using the same, uses thereof for the treatment or prophylaxis of related diseases, and uses thereof for the manufacture of a medicament for the treatment or prophylaxis of related diseases.
Background
Inflammatory corpuscles are a macromolecular complex composed of a number of proteins that mediate activation of the effector protein caspase-1, part of innate immunity. Different pattern recognition receptors (pattern recognition receptor, PRR) recognize pathogen-associated molecular patterns (PAMP) or lesion-associated molecular patterns (damage associated molecular patterns, DAMP), thereby forming a variety of different inflammatory corpuscles. To date, a number of PRRs have been proposed to form inflammatory corpuscles, including the nucleotide-binding oligomerization domain (NOD) -like receptor (NLR). Among the inflammatory corpuscles formed from NOD-like receptors, the NOD-like receptor (NOD like receptor containing pyrin domain, nlrp 3) containing the protein 3 related to the thermal protein structure is most typical, and the studies are most mature and extensive.
NLRP3 (NOD-like receptor protein3, NOD-like receptor protein3, or pyrin domain-containing protein 3) belongs to the NLR protein family. Most NLRs consist of 3 parts, including an amino-terminal Caspase recruitment region (CARD), a PYRIN (PYD) domain, an intermediate nucleotide binding NACHT domain, and a carboxy-terminal Leucine Rich Repeat (LRR) domain (Inoue et al, immunology,2013,139,11-18). NLRP3 is an intracellular signaling molecule capable of sensing exogenous infectious stimuli (viruses, bacteria, etc.), endogenous dangerous stimuli and particles in the environment, and is a key signaling node controlling maturation and secretion of two pro-inflammatory cytokines IL-1 beta and IL-18. Specifically, upon activation, NLRP3 binds to apoptosis-related spot-like proteins (ASCs) containing a caspase activation and recruitment domain; the ASCs then polymerize to form large aggregates, known as ASC specks; the polymerized ASC in turn interacts with caspase-1 to form a complex known as an inflammatory body; this causes activation of caspase-1, cleaving precursor forms of the pro-inflammatory cytokines IL-1β (interleukin-1β) and IL-18 (interleukin-18), referred to as pre-IL-1β and pre-IL-18, respectively, to convert to mature and bioactive IL-1β and IL-18, and induce apoptosis of the cell.
As a result of involvement in the maturation and secretion of IL-1 beta and IL-18, NLRP3 inflammatory bodies can contribute to the occurrence and progression of various related diseases such as neuroinflammation-related disorders, cardiovascular/metabolic disorders/diseases, wound healing and scar formation, inflammatory skin diseases, cancer-related diseases/disorders, and the like. Several genetic and pharmacological evidence have also shown the importance of NLRP3 inflammatory bodies in human diseases, which are associated with the pathogenesis of a number of complex diseases or disorders. Such diseases and disorders and related documents are detailed in CN112424207A, CN113784957a, for example, auto-inflammatory fever syndrome, liver-related diseases/disorders, inflammatory arthritis-related disorders, kidney-related diseases, and the like. Thus, NLRP3 inflammatory corpuscles can be an important target for the treatment of the above diseases.
Several small molecule compounds have been shown to inhibit NLRP3 inflammatory bodies. Glibenclamide (Glyburide) inhibits IL-1β at micromolar concentrations in response to activation of NLRP 3. Other previously characterized weak NLRP3 inhibitors include parthenolide, 3, 4-methylenedioxy-beta-nitrostyrene, and Dimethylsulfoxide (DMSO), but these agents have limited efficacy and are non-specific. Mangan et al (Nat Rev Drug discovery.2018 Aug;17 (8): 588-606) reviewed several small molecule NLRP3 inflammatory corpuscle inhibitors.
However, most of these compounds have been reported to have mixed modes of action and limited efficacy. Thus, there is a need to study and develop NLRP3 inflammasome inhibitors with potent NLRP3 inflammasome inhibitory activity, with improved patentability such as improved metabolic stability and/or pharmacokinetic properties and/or showing minimized hERG inhibition.
Brief description of the invention
It is an object of the present invention to provide compounds that can be used to treat, alleviate or prevent a group of diseases or disorders that are NLRP3 inflammasome-related diseases or disorders. The inventors have surprisingly found that these objects can be achieved by a compound of formula (I) as described below.
The present inventors have identified by research that the compounds of the present invention show satisfactory NLRP3 inflammatory body inhibitory activity and also good performance in metabolic stability and pharmacokinetic experiments, suggesting improved drug formation and improved bioavailability. Thus, the compounds of the present invention may not only achieve the objective of preventing or treating diseases associated with small NLRP3 inflammatory diseases, but also produce drugs expected to have improved absorption, increased efficacy at equivalent doses, or provide the same efficacy at lower doses and/or reduce possible side effects. Thus, the invention also provides the use of a compound of the invention in the manufacture of a medicament for the prevention or treatment of diseases associated with small NLRP3 inflammatory diseases, pharmaceutical compositions comprising said compound and methods of preventing and/or treating small NLRP3 inflammatory diseases associated by administering said compound.
Thus, in one aspect of the invention, there is provided a compound of formula (I), stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof:
wherein each group is as defined in the detailed description.
In another aspect of the invention, there is provided a compound of formula (I), a stereoisomer, a tautomer, a stable isotopic variant, a pharmaceutically acceptable salt or a solvate thereof, of the invention having NLRP3 inflammatory small body inhibitory activity, for use as a medicament, in particular as an inhibitor of NLRP3 inflammatory small bodies, for the treatment or prophylaxis of diseases in which NLRP3 inflammatory small bodies are involved.
In another aspect of the invention, there is provided a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient. In a particular aspect, the pharmaceutical composition may additionally comprise additional therapeutically active ingredients suitable for use in combination with the compounds of the present invention. In a particular aspect, pharmaceutical combination products, such as kits, comprising a compound of the invention and an additional active agent are provided.
In another aspect of the invention there is provided the use of a compound of the invention or a pharmaceutical composition comprising the same for the prevention or treatment of diseases associated with NLRP3 inflammatory minibodies in a mammal, particularly a human.
In another aspect of the invention, there is provided a method of preventing or treating a disease associated with a small amount of NLRP3 inflammation in a subject, such as a mammal, particularly a human, comprising administering an effective amount of a compound of the invention described herein or a pharmaceutical composition comprising the same.
In another aspect of the invention there is provided the use of a compound of the invention as described above, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for the prevention or treatment of a disease associated with NLRP3 inflammatory minibody.
In a further aspect, methods for synthesizing the compounds of the invention are provided, with representative synthetic schemes and pathways described below.
Other objects and advantages of the present invention will become apparent to those skilled in the art from a reading of the following detailed description.
Detailed Description
Definition of the definition
Unless otherwise indicated, each term used in the specification and claims has the meaning indicated below. Where a particular term or phrase is not specifically defined, it should not be construed as being ambiguous or otherwise clear, but rather should be construed in accordance with the ordinary meaning of the art in conjunction with the context herein.
Unless explicitly stated otherwise herein, singular references may also include plural references. For example, "a" and "an" may refer to one, or one or more.
Unless otherwise indicated, any heteroatom having an unsatisfied valence is considered to have a hydrogen atom sufficient to satisfy the valence.
The term "NLRP3" as used herein is meant to include, but is not limited to, nucleic acids, polynucleotides, oligonucleotides, sense and antisense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous NLRP3 molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.
The terms "inhibit" and "reduce" or any variant of these terms, as used herein, refer to the ability of a bioactive agent to reduce signaling activity of a target of interest, reduce or inhibit a given condition, symptom or disorder, or significantly reduce baseline activity of a biological activity or process by directly or indirectly interacting with the target. In particular, the term "inhibiting NLRP3" or "inhibiting NLRP3 inflammatory small body pathway" encompasses the ability to reduce IL-1 beta and/or IL-18 production by NLRP3 or NLRP3 inflammatory small body pathway. This may be accomplished by mechanisms including, but not limited to, inactivating, destabilizing NLRP3, and/or altering NLRP3 distribution. For the purposes of the present application, the term "NLRP3 inflammatory corpuscle inhibitor" refers to a compound that inhibits NLRP3 inflammatory corpuscle activity (e.g., as characterized by the inhibition of IL-1β release by the biological example 1 of the present application) with an IC50 value of 500-1000nM, 200-500nM, 100-200nM, 50-100nM, 20-50nM, <20nM or any range derivable therein.
The term "NLRP3 inflammatory-small-body-related disease" as used herein refers to a disease in which NLRP3 signaling contributes to the pathology, and/or symptoms, and/or progression of the disease, including inflammatory diseases, immune diseases, cancer, infections (including viral and bacterial infections), central nervous system diseases, metabolic diseases, cardiovascular diseases, respiratory diseases, liver diseases, kidney diseases, eye diseases, skin diseases, lymphoid disorders, psychological disorders, graft versus host diseases, bone diseases, hematological diseases, allodynia, such as, but not limited to: autoinflammatory fever syndrome (e.g., leng Yan element-related periodic syndrome), liver-related diseases/disorders (e.g., chronic liver disease, viral hepatitis, nonalcoholic steatohepatitis (NASH), alcoholic steatohepatitis, and alcoholic liver disease), inflammatory arthritis-related disorders (e.g., gout, pseudogout (chondrocalcanesis), osteoarthritis, rheumatoid arthritis, joint diseases such as acute, chronic), kidney-related diseases (e.g., hyperoxalic urine syndrome, lupus nephritis, type I/type II diabetes and related complications (e.g., nephropathy, retinopathy), hypertensive nephropathy, hemodialysis-related inflammation), neuroinflammation-related diseases (e.g., multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, alzheimer's disease), cardiovascular/metabolic diseases/disorders (e.g., reduced cardiovascular risk (CvRR), hypertension, atherosclerosis, type I and type II diabetes and related complications, peripheral Arterial Disease (PAD), acute heart failure), inflammatory skin diseases (e.g., suppurative gland inflammation, acne), wound and formation, aging, cancer, myelodysplasia, myelosis, cancer, myelosis, and myelosis-related diseases (e.g., myelosis).
The term "hERG" as used herein refers to the human Ether-a-go-go related gene which encodes potassium ion channels (sometimes simply denoted by "hERG") most well known for their potential activity on the heart, hERG channels mediating delayed rectifier potassium currents- -I in cardiac action potentials Kr When the ability of this channel to mediate electrical flow through the cell membrane is inhibited or compromised by drug action or by certain genetic mutations, this can lead to potentially fatal diseases-QT interval prolongation syndrome and arrhythmia. When there is a trend in drugs to inhibit hERG, there is a risk of adverse drug reactions that may lead to sudden death. Therefore, there is a need to avoid as much as possible the hERG inhibition of the drug against the target, and minimize the hERG inhibition of the drug during drug developmentIt would be beneficial to make. In some embodiments of the invention, the compounds of the invention (or pharmaceutically acceptable salts thereof) exhibit minimal hERG inhibition in the assays described in the examples, as compared to prior art active agents.
The term "treating" as used herein refers to administering one or more compounds of the invention described herein to a subject, e.g., mammal, e.g., human, suffering from or having associated symptoms, to cure, alleviate or affect the disease or associated symptoms. In a specific embodiment of the invention, the disease is referred to as NLRP3 inflammatory body-related disease as defined above.
The term "preventing" as used herein refers to administering one or more compounds of the invention described herein to a subject, e.g., mammal, e.g., human, suspected of suffering from or susceptible to an NLRP3 inflammatory small-scale related disease as defined herein, such that the risk of suffering from the defined disease is reduced. The term "preventing" encompasses the use of the compounds of the present invention prior to diagnosing or determining any clinical and/or pathological symptoms.
The term "therapeutically effective amount" as used herein means an amount sufficient to reduce or completely alleviate symptoms or other deleterious effects of the disease when administered to a subject to treat the disease; reversing, completely stopping or slowing the progression of the disorder; or reducing the risk of exacerbation of the disease, "effective amount" may vary depending on the compound, the disease and its severity, the age, weight, etc., of the individual to be treated.
The term "individual" or "subject" as used herein includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) or normal individuals suffering from a disease (e.g., a disease described herein). "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, domestic animals and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
The term "pharmaceutical composition" or "pharmaceutical formulation" as used herein refers to a composition comprising one or more compounds of the present invention or stereoisomers, tautomers, stable isotopic derivatives, pharmaceutically acceptable salts or solvates thereof, and pharmaceutically acceptable excipients or carriers generally accepted in the art, and may be in solid, semi-solid, liquid or gaseous form.
The term "pharmaceutical combination" as used herein means that the compounds of the present invention may be combined with other active agents for the purpose of the present invention. The other active agent may be one or more additional compounds of the present invention, or may be a second or additional (e.g., third) compound that is compatible with, i.e., does not adversely affect each other, or has complementary activity to, the compounds of the present invention. Such agents are suitably present in combination in an amount effective to achieve the intended purpose. The additional active agents may be co-administered with the compounds of the present invention in a single pharmaceutical composition or administered separately from the compounds of the present invention in separate discrete units, e.g., in kit form, which may be simultaneous or sequential when administered separately. The successive administrations may be close or distant in time.
The term "pharmaceutically acceptable" as used herein means molecular entities and compositions approved by or by the corresponding agency of the respective country or listed in the generally recognized pharmacopoeia for animals, and more particularly humans, or which do not produce adverse, allergic or other untoward reactions when administered in appropriate amounts to animals, such as humans.
The term "pharmaceutically acceptable excipient or carrier" as used herein refers to one or more compatible solid or liquid filler or gel materials which are pharmacologically inactive, compatible with the other ingredients of the composition, and which are intended to be acceptable for administration to warm-blooded animals such as humans, for use as carriers or vehicles in the form of administration of the compounds of the invention, examples of which include, but are not limited to, cellulose and its derivatives (e.g., sodium carboxymethyl cellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g., magnesium stearate), calcium sulfate, vegetable oils, polyols (e.g., propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifying agents (e.g., tweens), wetting agents (e.g., sodium lauryl sulfate), colorants, flavoring agents, stabilizers, antioxidants, preservatives, and the like.
The term "pharmaceutically acceptable salt" as used herein means a salt of a compound of the invention which is pharmaceutically acceptable and which has the desired pharmacological activity of the parent compound. In particular, such salts are non-toxic and may be inorganic acid addition salts or organic acid addition salts and base addition salts. In particular, such salts include: (1) Acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or acid addition salts with organic acids such as acetic acid, propionic acid, caproic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptanoic acid, 3-phenylpropionic acid, trimethylacetic acid, t-butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) a salt formed when an acidic proton present in the parent compound is replaced with a metal ion such as an alkali metal ion, alkaline earth metal ion or aluminum ion, or coordinated with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, or the like. Those skilled in the art will be aware of the general principles and techniques for preparing pharmaceutically acceptable salts, such as those described in Berge et al, pharm ScL,66,1-19 (1977).
The term "stereoisomer" as used herein refers to an isomer formed from at least one asymmetric center. In compounds having one or more, e.g., 1, 2, 3 or 4 asymmetric centers, racemic mixtures, single enantiomers, diastereomers and individual diastereomers may be produced. Specific molecules may also exist as geometric isomers (cis/trans). The compounds of the present invention may also exist in a mixture of two or more structurally distinct forms in rapid equilibrium, commonly referred to as tautomers, representative examples include keto-enol, phenol-keto, nitroso-oxime tautomers, and the like. It is to be understood that the scope of the present invention encompasses all such isomers and mixtures thereof in any proportion, e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%.
The term "solvate" as used herein refers to a solvent addition form comprising a stoichiometric or non-stoichiometric solvent, including any solvated form of the compounds of the present invention, such as a solvate with water, such as a hydrate, or with an organic solvent, such as methanol, ethanol or acetonitrile, i.e. as a methanolate, ethanolate or acetonitrile, respectively; or in the form of any polymorph. It is to be understood that such solvates of the compounds of the present invention also include solvates of pharmaceutically acceptable salts of the compounds of the present invention.
The term "isotopic variant" as used herein refers to a compound comprising a non-natural proportion of isotopes on one or more atoms constituting the compound. The compounds of the present invention may contain an unnatural proportion of an atomic isotope on one or more of the atoms comprising the compound, thereby forming isotopic variations. It will be appreciated that such isotopically-labelled compounds are useful in metabolic studies (with 14 C) Kinetic studies of the reaction (e.g. using 2 H or 3 H) Detection or imaging techniques (e.g., positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT), including drug or substrate tissue distribution assays), or for radiation therapy of a patient. Furthermore, by isotopes, e.g. deuterium 2 H substitution may provide certain therapeutic advantages because of higher metabolic stability, e.g., increased in vivo half-life or reduced dosage requirements.
Examples of isotopes that can be incorporated into compounds of the invention include, but are not limited to 2 H or D, 3 H、 11 C、 13 C、 14 C、 15 N、 18 F 31 P、 32 P、 35 S、 36 Cl、 123 I、 124 I、 125 I. In particular, the present invention relates to deuterated compounds of formula (I), stereoisomers, tautomers, pharmaceutically acceptable salts or solvates thereof. It will be appreciated that isotopically-labelled compounds of the present invention can generally be employed by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying examples and preparations using the appropriate isotopes Labeled reagent is prepared in place of unlabeled previously used reagent.
It is to be understood that deuterium in deuterated compounds of the present invention is considered a substituent for the compounds of formula (I). The concentration of deuterium may be defined by an isotopic enrichment factor. The term "isotopically enriched factor" as used herein means the ratio between the abundance of an isotope and the natural abundance of a particular isotope. The deuterated compounds of the present invention have an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation on each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). It should be understood that the term "isotopically enriched factor" can be applied to any isotope in the same manner as described for deuterium.
As used in the structural formulae or structural fragments of the compounds hereinOr->Indicating the presence of stereoisomers and the absolute configuration of the asymmetric centers, are generally denoted by R or S in the nomenclature of the compounds or intermediates provided herein. When present in the racemic mixture, the real and imaginary wedge symbols define the relative stereochemistry, not the absolute stereochemistry.
As used in the structural fragments referred to hereinThe bond indicating the intersection therewith is the bond of the structural fragment to the rest of the molecule.
The term "halo" or "halogen" as used herein refers to fluorine (F), chlorine (Cl), bromine (Br) and iodine (I). Preferred halo is fluoro or chloro.
The term "halogen-substituted" as used herein to define groups is intended to include monohalogenated or polyhalogenated groups in which one or more (e.g., 2, 3, 4, 5, or 6) hydrogens in the group are replaced with one or more (e.g., 2, 3, 4, 5, or 6) same or different halogens.
The term "hydroxy" as used herein refers to-OH.
The term "oxo" as used herein refers to = O.
The term "alkyl" as used herein refers to a straight or branched saturated hydrocarbon group consisting of carbon and hydrogen atoms. In particular, the alkyl groups have 1-10, for example 1 to 6, 1 to 5, 1 to 4, 1 to 3 or 1 to 2 carbon atoms. For example, the term "C" as used herein 1 -C 6 Alkyl "refers to a straight or branched saturated hydrocarbon group having 1 to 6 carbon atoms, examples of which are methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl or tert-butyl), pentyl (including n-pentyl, isopentyl, neopentyl), n-hexyl, 2-methylpentyl, and the like. Specific alkyl groups have 1 to 3 carbon atoms.
The term "halogen-substituted alkyl" as used herein refers to an alkyl group as defined herein wherein one or more hydrogen atoms are replaced by an independently selected halogen, such as, but not limited to, -CH 2 F、-CHF 2 、-CF 3 、-CH 2 Br、-CHBr 2 、-CH 2 Cl、-CHCl 2 、-CCl 3 、-CH 2 CH 2 F、-CH 2 CHF 2 、-CH 2 CF 3 、-CH 2 CH 2 CH 2 F、-CH 2 CH 2 CHF 2 、-CH 2 CH 2 CF 3 、-C 2 F 5 、-C 2 Cl 5 、-CH(CF 3 ) 2
The terms "alkoxy" and "alkylthio" as used herein refer to-O-alkyl and-S-alkyl, respectively, wherein the alkyl is as defined herein.
The terms "halogen-substituted alkoxy" and "halogen-substituted alkylthio" as used herein refer to halogen-substituted alkyl as defined above, which is attached through-O-or-S-.
The term "alkylene" as used herein, alone or in combination with other groups, refers to saturated straight or branched chain divalent hydrocarbon radicals, such as "- (CH" as used in the definition of the compounds of the invention 2 ) n "refers to a straight or branched chain alkylene group having n carbon atoms, representative examples being methylene, ethylene, and the like.
The term "cycloalkyl" as used herein refers to a monocyclic, fused, bridged polycyclic, or spiro non-aromatic saturated monovalent hydrocarbon ring structure having the indicated number of ring atoms. Cycloalkyl groups can have 3 to 12 carbon atoms (i.e., C 3 -C 12 Cycloalkyl), e.g. C 3-10 Cycloalkyl, C 3-8 Cycloalkyl, C 3-6 Cycloalkyl, C 5-6 Cycloalkyl groups. Examples of suitable cycloalkyl groups include, but are not limited to, monocyclic structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like; or polycyclic (e.g. bicyclic) structures, including spiro, fused or bridged systems, e.g. bicyclo [1.1.1 ]Amyl, bicyclo [2.2.1]Heptyl, spiro [3.4 ]]Octyl and bicyclo [3.1.1]Hexalkyl and bicyclo [3.1.1]Heptyl or bicyclo [3.2.1]Octyl, and the like.
The term "cycloalkenyl" as used herein refers to cycloalkyl groups as defined above which contain one or more carbon-carbon double bonds and which are non-aromatic, such as cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl or cycloheptatrienyl and the like.
The term "heterocycloalkyl" as used herein is meant to include one or more (e.g., 1, 2, 3, or 4) heteroatoms independently selected from O, N and S, a monocyclic, fused polycyclic, spiro, or bridged polycyclic non-aromatic saturated ring structure having the specified number of ring atoms, or an N-oxide thereof, or an S-oxide or S-dioxide thereof. Heterocycloalkyl groups can have 3 to 12 ring members (which can be referred to as 3-12 membered heterocycloalkyl groups), for example 3 to 10 ring members, 3 to 8 ring members, 3 to 7 ring members, 4 to 10 ring members, 4 to 7 ring members, etc., and can be, for example, a 5-6 membered monocyclic ring, or a fused bicyclic ring structure formed from two fused 6-membered rings, two fused 5-membered rings, fused 6-membered rings and 5-membered rings, or fused 5-membered rings and 4-membered rings. Specifically, examples of suitable heterocycloalkyl groups include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl (e.g., 1-pyrrolidinyl, 2-pyrrolidinyl, and 3-pyrrolidinyl), tetrahydrofuranyl (e.g., 1-tetrahydrofuranyl, 2-tetrahydrofuranyl, and 3-tetrahydrofuranyl), tetrahydrothienyl (e.g., 1-tetrahydrothienyl, 2-tetrahydrothienyl, and 3-tetrahydrothienyl), piperidinyl (e.g., 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, and 4-piperidinyl), tetrahydropyranyl (e.g., 4-tetrahydropyranyl), tetrahydrothiopyranyl (e.g., 4-tetrahydrothiopyranyl), morpholinyl (e.g., morpholino), thiomorpholino, dioxanyl, piperazinyl, or azepanyl, diazacycloheptyl such as 1, 4-diazacycloheptyl, 3, 6-diaza-bicyclo [3.1.1] heptyl, or 3-aza-bicyclo [3.2.1] octyl. The atom of the heterocycloalkyl group to which the remainder of the compound is attached may be a carbon atom or a heteroatom, as long as it is chemically feasible. Exemplary heterocycloalkyl groups for the compounds of the present invention include, but are not limited to:
And the corresponding groups in which the points of attachment to the remainder of the molecule are at other available positions in the heterocycloalkyl group shown. It is to be understood that structures with asymmetric centers encompass racemic and/or single enantiomeric forms thereof, e.g.>Can represent->And/or +.>
The term "heterocycloalkenyl" as used herein refers to a "heterocyclyl" as defined herein that contains at least one (e.g., 1, 2, or 3) double bond. Examples of suitable heterocycloalkenyl groups include, but are not limited to:
wherein each W is selected from CH 2 NH, O and S, each Y is selected from NH, O, C (=O), SO 2 And S, provided that each ring contains at least one atom selected from N, O or S; for example pyrrolinyl, dihydrofuryl, dihydrothienyl, tetrahydropyridinyl, tetrahydropyranyl or tetrahydrothiopyranyl. It is understood that heterocycloalkenyl in the definition of compounds herein refers to heterocycloalkenyl fused to an aromatic ring containing at least one heteroatom selected from N, O, S, for example heterocycloalkenyl fused to a phenyl group, for example phenyl-azacyclopentenyl, benzoazacyclohexenyl, benzoxazolenyl, benzoxazetenyl, and the like.
The term "aryl" as used herein refers to a monovalent aromatic hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom in an aromatic ring system. Specifically, aryl refers to a monocyclic or fused polycyclic aromatic ring structure having the specified number of ring atoms. In particular, the term includes groups comprising 6 to 14, for example 6 to 10, preferably 6 ring members. Specific aryl groups include phenyl and naphthyl, most particularly phenyl.
The term "heteroaryl" as used herein is meant to include one or more (e.g., 1,2, 3, or 4) heteroatoms independently selected from O, N and S, a monocyclic or fused polycyclic aromatic ring structure having the specified number of ring atoms, or an N-oxide thereof, or an S-oxide or S-dioxide thereof, wherein at least one ring is an aromatic ring containing heteroatoms. In particular, the heteroaryl structure may have 5 to 10 ring members and may be, for example, a 5-6 membered monocyclic ring, or a fused bicyclic ring structure formed from two fused 6-membered rings, two fused 5-membered rings, a fused 6-membered ring and a 5-membered ring, or a fused 5-membered ring and a 4-membered ring. Heteroaryl rings will typically contain up to 4 heteroatoms, more typically up to 3 heteroatoms, more typically up to 2, e.g., a single heteroatom independently selected from O, N and S. In one embodiment, the heteroaryl ring contains at least one ring N atom, at least one ring S atom, and/or at least one ring O atom. For example, the heteroaryl group may be a 5-6 membered heteroaryl group comprising 1-3 heteroatoms independently selected from N, O or S. Examples of suitable 5-membered monocyclic heteroaryl groups include, but are not limited to, pyrrolyl, furanyl, thienyl, imidazolyl, furazanyl, oxazolyl, oxadiazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrazolyl, triazolyl, and tetrazolyl; examples of suitable 6-membered monocyclic heteroaryl groups include, but are not limited to, pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, and triazinyl. Heteroaryl may also be a fused ring comprising 1,2, 3 or 4 heteroatoms independently selected from N, O or S, such as benzofuran, benzothiophene, indole, benzimidazole, indazole, benzotriazole, pyrrolo [2,3-b ] pyridine, pyrrolo [2,3-c ] pyridine, pyrrolo [3,2-b ] pyridine, imidazo [4,5-c ] pyridine, pyrazolo [4,3-d ] pyridine, pyrazolo [4,3-c ] pyridine, pyrazolo [3,4-b ] pyridine, isoindole, purine, indolizine, imidazo [1,2-a ] pyridine, imidazo [1,5-a ] pyridine, pyrazolo [1,5-a ] pyridazine, pyrrolo [1,2-b ] pyrimidine imidazo [1,2-c ] pyrimidine, 5H-pyrrolo [3,2-b ] pyrazine, 1H-pyrazolo [4,3-b ] pyrazine, 1H-pyrazolo [3,4-d ] pyrimidine, 7H-pyrrolo [2,3-d ] pyrimidine, quinoline, isoquinoline, cinnoline, quinazoline, quinoxaline, phthalazine, 1, 6-naphthyridine, 1, 7-naphthyridine, 1, 8-naphthyridine, 1, 5-naphthyridine, 2, 6-naphthyridine, 2, 7-naphthyridine, pyrido [3,2-d ] pyrimidine, pyrido [4,3-d ] pyrimidine, pyrido [2,3-b ] pyrazine, pyrido [3,4-b ] pyrazine, pyrimido [5,4-d ] pyrimidine, pyrazino [2,3-b ] pyrazines and pyrimido [4,5-d ] pyrimidines. The atom in the heteroaryl group that is attached to the remainder of the compound may be a carbon atom or a heteroatom, as long as it is chemically feasible.
Those of ordinary skill in the art of organic synthesis understand that stable, chemically feasible heterocycles, whether aromatic or non-aromatic, wherein the maximum number of heteroatoms or the type of heteroatoms contained is determined by the ring size, unsaturation, and valence of the heteroatoms. In general, a heterocycle may have from 1 to 4 heteroatoms, provided that the heterocycle or heteroaryl ring is chemically feasible and stable.
It will be appreciated by those of ordinary skill in the art of organic synthesis that the cyclic groups containing heteroatoms, whether aromatic or non-aromatic, attached in the structure of the compounds of the present invention, wherein the ring atoms attached to the remainder of the molecule may be carbon atoms or heteroatoms, provided that they are chemically feasible and stable.
The term "substituted" or "substituted" as used herein means that one or more (e.g., 1, 2, 3, or 4) hydrogens on the designated atom are replaced with a reference to the designated group, provided that the designated atom's normal valence is not exceeded and a stable compound is formed, and that the combination of substituents and variables is permitted only when such combination forms a stable compound.
The term "optionally substituted" as used herein, unless otherwise indicated, means that a group may be unsubstituted or substituted with one or more (e.g., 0, 1, 2, 3, 4, or 5 or more) substituents listed for that group, wherein the substituents may be the same or different.
Unless otherwise specified, C in the definition of the compounds of the application n-n+m Cases involving n to n+m carbons, e.g. C 1-6 Comprises C 1 、C 2 、C 3 、C 4 、C 5 And C 6 Also included is any one of the ranges n to n+m, e.g. C 0-6 Comprises C 1 、C 2 、C 3 、C 4 、C 5 、C 6 、C 0-1 、C 0-2 、C 0-3 、C 0-4 、C 0-5 、C 1-2 、C 1-3 、C 1-4 、C 2-3 Etc. Similarly, n-to n+m-members in the definition of the compounds of the present application means that the number of ring atoms may be any one of n to n+m, and any range of n to n+m-members is also included.
As used in this specification and the claims that follow, the words "comprise", "comprising" and "include" mean "including but not limited to", and do not exclude, for example, other additives, ingredients, integers or steps. It is to be understood that the term includes "consisting of, or" consisting essentially of, the recited components, steps, or conditions.
It is to be understood that the dosages referred to when describing the compounds of the application, pharmaceutical compositions, combinations of drugs and related uses and methods thereof are based on the weight of the free form and not on any salts, hydrates or solvates thereof, etc., unless otherwise defined in the specification.
Compounds of the application
The terms "inventive compounds" and the like as used throughout the present application, unless otherwise indicated, encompass compounds of formula (I), stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof as defined in the various embodiments herein and in particular or preferred embodiments thereof. The stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates and prodrugs are as described in the definition section above. Preferably, the compounds of the present application are free forms of the compounds of formula (I) or a pharmaceutically acceptable salt or solvate thereof; most preferred are the free forms of the compounds of formula (I) or pharmaceutically acceptable salts thereof.
Certain compounds of the invention may exist in polymorphic or amorphous forms and are also within the scope of the present invention. When in solid crystalline form, the compound of formula (I) may be in the form of a co-crystal with another chemical entity, and this specification includes all such co-crystals.
In the presence of chiral centers, the compounds of the present invention may exist as individual enantiomers or as mixtures of enantiomers, and one skilled in the art will be able to determine the isomeric forms of the compounds of the present invention that are stable and viable. According to one embodiment, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof, which is a single enantiomer having an enantiomeric excess (% ee) > 95, > 98% or > 99%. Preferably, a single enantiomer is present in > 99% enantiomeric excess (% ee).
The compounds of the invention also encompass the N-oxides which may be present, and the person skilled in the art is able to determine the stable and viable N-oxides of the compounds of the invention. The compounds of the present invention also encompass metabolites of the compounds of the present invention, i.e., substances formed in vivo by oxidation, reduction, hydrolysis, amidation, esterification, etc. upon administration of the compounds of the present invention, which can be identified by techniques well known in the art.
Various embodiments of the invention are described below. It will be appreciated that features from each embodiment may be combined with features from one or more other embodiments to provide further embodiments of the invention.
Embodiment 1: the present invention provides compounds of formula (I), stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof:
wherein, the liquid crystal display device comprises a liquid crystal display device,
x is-NH- (CH) 2 ) n -or-O-;
ring A is selected from phenyl, 5-6 membered heteroaryl ring, 5-7 membered cycloalkenyl, and 5-7 membered heterocycloalkenyl, each optionally substituted with oxo, halogen, CN, C 1-6 Alkyl-or halogen-substituted C 1-6 Alkyl substitution;
R 1 selected from halogen, CN, C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, -SO 3 H、COOH-、-S(O) 2 -C 1-6 Alkyl, -S (O) -C 1-6 Alkyl, -CO-C 1-6 Alkyl and-CO-OC 1-6 Alkyl group, wherein C 1-6 Alkyl, C 1-6 Alkoxy or C 1-6 Alkylthio groups are each independently at each occurrence optionally substituted with halogen;
R 2 selected from H, halogen, CN and C optionally substituted by halogen 1-3 An alkyl group;
R 3 selected from-OH, -SH, -NHSO 2 -R 3a 、-NH-SO-R 3a 、-NH-CO-R 3a 、-CH(R 3b )(SO 2 -R 3a )、-CH(R 3b )(SO-R 3a )、-CH(R 3b )(CO-R 3a ) and-CH (R) 3c ) 2
R 3a Each at each occurrence is independently selected from-C 1-6 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl, - (CH) 2 ) m -3-8 membered heterocycloalkyl, - (CH) 2 ) m -3-8 membered heterocycloalkenyl, - (CH) 2 ) m -C 6-10 Aryl and- (CH) 2 ) m -a 5-10 membered heteroaryl, each optionally substituted with a group selected from: halogen, CN, C optionally substituted by halogen 1-6 Alkyl, optionally halogen-substituted C 1-6 Alkoxy and optionally halogen-substituted C 3-6 Cycloalkyl;
R 3b independently at each occurrence selected from H, halogen and C optionally substituted with halogen 1-6 An alkyl group;
R 3c independently at each occurrence selected from halogen, CN and optionally halogen substituted-C 1-6 An alkyl group;
ra, rb are each independently selected from H, halogen, CN, C optionally substituted with halogen 1-6 Alkyl and optionally halogen-substituted C 1-6 An alkoxy group;
R 4 selected from the group consisting of-3-10 membered heterocycloalkyl and-C 3-6 Cycloalkyl groups, each optionally substituted with a group selected from: -OH, halogen, oxo, -CO 2 H、-SO 2 NH 2 C optionally substituted by halogen 1-6 Alkyl, C 1-6 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -3-8 membered heterocycloalkyl, and C in the substituents 3-6 Cycloalkyl and-3-8 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 NH 2 、-SO 2 (C 1-3 Alkyl group) or-NH-SO 2 (C 1-3 Alkyl) substitution; and is also provided with
Wherein n is selected from 0, 1 and 2;
m is independently selected from 0, 1 or 2 at each occurrence.
Embodiment 2: according to embodiment 1, wherein X is-NH- (CH) 2 ) n -。
Embodiment 2a: according to embodiment 2, wherein X is-NH- (CH) 2 ) n -, and n is 0 or 1, preferably n is 0.
Embodiment 3: according to embodiment 1, wherein X is-O-.
Embodiment 4: according to any of embodiments 1-3, wherein ring A is a benzene ring, optionally substituted with oxo, halogen, CN, C 1-6 Alkyl-or halogen-substituted C 1-6 Alkyl substitution; preferably A is a benzene ring.
Embodiment 5: according to any of embodiments 1-3, wherein ring A is a 5-6 membered heteroaromatic ring containing 1 or 2 heteroatoms selected from O, N and S, and optionally oxo, halogen, CN, C 1-6 Alkyl-or halogen-substituted C 1-6 Alkyl substitution;
for example, ring A is a 6 membered heteroaryl ring containing 1 or 2N heteroatoms, a 5 membered heteroaryl ring containing 1 or 2N atoms, a 5 membered heteroaryl ring containing 1O or S atoms, or a 5 membered heteroaryl ring containing 2 heteroatoms selected from N, O and S, each optionally substituted with oxo, halogen, C 1-3 Alkyl-or halogen-substituted C 1-3 Alkyl substitution;
in particularExamples of (a) include, but are not limited to:
/>
embodiment 5a: according to embodiment 5, whereinSelected from:
embodiment 6: according to any of embodiments 1-3, wherein ring A is a 5-7 membered cycloalkenyl ring, and is optionally substituted with oxo, halogen, CN, C 1-6 Alkyl-or halogen-substituted C 1-6 Alkyl substitution; for example cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl or cycloheptatrienyl, optionally substituted by said substituents; preferably A is a cyclohexenyl ring.
Embodiment 7: according to any of embodiments 1-3, wherein ring A is a 5-7 membered heterocycloalkenyl ring containing 1 or 2 heteroatoms selected from O, N and S, and optionally substituted with oxo, halogen, CN, C 1-6 Alkyl-or halogen-substituted C 1-6 Alkyl substitution; for example, ring A is a 5-, 6-or 7-membered heterocycloalkenyl group containing 1O, N or S heteroatom, a 5-, 6-or 7-membered heterocycloalkenyl group containing 2 heteroatoms selected from O, N and S, each optionally substituted with oxo, halogen, C 1-3 Alkyl-or halogen-substituted C 1-3 Alkyl substitution;
in particularExamples include, but are not limited to:
embodiment 7a: according to embodiment 7, whereinSelected from->
Embodiment 8: according to any of embodiments 1-7a, wherein R 1 Selected from halogen, for example F, cl, br, I, preferably F or Cl.
Embodiment 9: according to any of embodiments 1-7a, wherein R 1 Is CN.
Embodiment 10: according to any of embodiments 1-7a, wherein R 1 For C optionally substituted by halogen 1-6 Alkyl, preferably C substituted by halogen 1-3 Alkyl groups, specific examples include, but are not limited to, -CH 3 、-CH 2 CH 3 、-CH 2 CH 2 CH 3 、-CH(CH 3 )(CH 3 )、-CH 2 F、-CHF 2 、-CF 3 、-CH 2 Br、-CHBr 2 、-CH 2 Cl、-CHCl 2 、-CCl 3 、-CH 2 CH 2 F、-CH 2 CHF 2 、-CH 2 CF 3 、-CH 2 CH 2 CH 2 F、-CH 2 CH 2 CHF 2 、-CH 2 CH 2 CF 3 、-C 2 F 5 、-C 2 Cl 5 、-CH(CF 3 ) 2 The method comprises the steps of carrying out a first treatment on the surface of the preferably-CH 3 、-CH 2 CH 3 、-CH 2 F、-CHF 2 、-CF 3 、-CH 2 Br、-CHBr 2 、-CH 2 Cl、-CHCl 2
Embodiment 11: according to any of embodiments 1-7a, wherein R 1 For C optionally substituted by halogen 1-6 Alkoxy, preferably C substituted by halogen 1-3 Alkoxy groups, specific examples include, but are not limited to, -OCH 3 、-OCH 2 CH 3 、-OCH 2 CH 2 CH 3 、-OCH(CH 3 )(CH 3 )、-OCH 2 F、-OCHF 2 、-OCF 3 、-OCH 2 Br、-OCHBr 2 、-OCH 2 Cl、-OCHCl 2 、-OCCl 3 、-OCH 2 CH 2 F、-OCH 2 CHF 2 、-OCH 2 CF 3 、-OCH 2 CH 2 CH 2 F、-OCH 2 CH 2 CHF 2 、-OCH 2 CH 2 CF 3 、-OC 2 F 5 、-OC 2 Cl 5 、-OCH(CF 3 ) 2 The method comprises the steps of carrying out a first treatment on the surface of the preferably-OCH 3 、-OCH 2 CH 3 、-OCH 2 F、-OCHF 2 、-OCF 3 、-OCH 2 Br、-OCHBr 2 、-OCH 2 Cl、-OCHCl 2
Embodiment 12: according to any of embodiments 1-7a, wherein R 1 For C optionally substituted by halogen 1-6 Alkylthio, preferably C substituted by halogen 1-3 Alkylthio groups, specific examples include, but are not limited to, -SCH 3 、-SCH 2 CH 3 、-SCH 2 CH 2 CH 3 、-SCH(CH 3 )(CH 3 )、-SCH 2 F、-SCHF 2 、-SCF 3 、-SCH 2 Br、-SCHBr 2 、-SCH 2 Cl、-SCHCl 2 、-SCCl 3 、-SCH 2 CH 2 F、-SCH 2 CHF 2 、-SCH 2 CF 3 、-SCH 2 CH 2 CH 2 F、-SCH 2 CH 2 CHF 2 、-SCH 2 CH 2 CF 3 、-SC 2 F 5 、-SC 2 Cl 5 、-SCH(CF 3 ) 2 The method comprises the steps of carrying out a first treatment on the surface of the preferred-SCH 3 、-SCH 2 CH 3 、-SCH 2 F、-SCHF 2 、-SCF 3 、-SCH 2 Br、-SCHBr 2 、-SCH 2 Cl、-SCHCl 2
Embodiment 13: according to any of embodiments 1-7a, wherein R 1 is-SO 3 H。
Embodiment 14: according to any of embodiments 1-7a, wherein R 1 COOH.
Embodiment 15: according to any embodiment1-7a, wherein R 1 Selected from-S (O) 2 -C 1-6 Alkyl and-S (O) -C 1-6 Alkyl, preferably-S (O) 2 -C 1-3 Alkyl and-S (O) -C 1-3 Alkyl, wherein alkyl is optionally substituted with halogen; specific examples of alkyl groups wherein the alkyl group is optionally substituted with halogen are as in embodiment 10 for R 1 Defined as follows.
Embodiment 16: according to any of embodiments 1-7a, wherein R 1 Selected from-CO-C 1-6 Alkyl and-CO-OC 1-6 Alkyl, preferably-CO-C 1-3 Alkyl and-CO-OC 1-3 Alkyl, wherein alkyl is optionally substituted with halogen, e.g., specific examples of alkyl wherein alkyl is optionally substituted with halogen are as in embodiment 10 for R 1 Defined as follows.
Embodiment 17: according to any of embodiments 1-16, wherein R 2 H.
Embodiment 18: according to any of embodiments 1-16, wherein R 2 Halogen, for example F, cl, br, I.
Embodiment 19: according to any of embodiments 1-16, wherein R 2 Is CN.
Embodiment 20: according to any of embodiments 1-16, wherein R 2 For C optionally substituted by halogen 1-3 Specific examples of alkyl groups, e.g., wherein the alkyl group is optionally substituted with halogen, are as for R in embodiment 10 1 Defined as follows.
Embodiment 21: according to any of embodiments 1-20, wherein R 3 is-OH.
Embodiment 22: according to any of embodiments 1-20, wherein R 3 is-SH.
Embodiment 23: according to any of embodiments 1-20, wherein R 3 Selected from-NHSO 2 -R 3a 、-NH-SO-R 3a and-NH-CO-R 3a preferably-NHSO 2 -R 3a
Embodiment 23a; according to embodiment 23, wherein R 3a Selected from-C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl, - (CH) 2 ) m -4-6 membered heterocycloalkyl, - (CH) 2 ) m -4-6 membered heterocycloalkenyl、-(CH 2 ) m -C 6 Aryl and- (CH) 2 ) m -a 5-6 membered heteroaryl, wherein m is independently at each occurrence 0 or 1, each optionally substituted with a group selected from: halogen, CN, C optionally substituted by halogen 1-6 Alkyl and optionally halogen-substituted C 1-6 An alkoxy group; preferably R 3a Selected from-C optionally substituted by halogen 1-3 Alkyl, specific examples are as in embodiment 10 for R 1 Defined as follows.
Embodiment 24: according to any of embodiments 1-20, wherein R 3 Selected from-CH (R) 3b )(SO 2 -R 3a )、-CH(R 3b )(SO-R 3a ) and-CH (R) 3b )(CO-R 3a )。
Embodiment 24a: according to embodiment 24, wherein R 3a Selected from-C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl, - (CH) 2 ) m -4-6 membered heterocycloalkyl, - (CH) 2 ) m -4-6 membered heterocycloalkenyl, - (CH) 2 ) m -C 6 Aryl and- (CH) 2 ) m -a 5-6 membered heteroaryl, wherein m is independently at each occurrence 0 or 1, each optionally substituted with a group selected from: halogen, CN, C optionally substituted by halogen 1-6 Alkyl and optionally halogen-substituted C 1-6 An alkoxy group; preferably R 3a Selected from-C optionally substituted by halogen 1-3 Alkyl, specific examples are as in embodiment 10 for R 1 Defined as follows.
Embodiment 24b: according to embodiment 24 or 24a, wherein R 3b Independently at each occurrence selected from H, halogen and C optionally substituted with halogen 1-3 Alkyl, specific examples are shown in embodiments 8 and 10 for R, respectively 1 Defined as follows.
Embodiment 25: according to any of embodiments 1-20, wherein R 3 is-CH (R) 3c ) 2
Embodiment 25a; according to embodiment 25, wherein R 3c Each independently is halogen, preferably F; or two R 3c All halogen, preferably all F.
Description of the embodiments25b: according to embodiment 25, wherein R 3c Each independently selected from halogen, CN and optionally halogen substituted-C 1-3 An alkyl group; or one of R 3c Is halogen or CN, the other being-C optionally substituted by halogen 1-3 An alkyl group; or two R 3c Are all optionally halogen-substituted-C 1-3 An alkyl group; wherein-C is optionally substituted by halogen 1-3 Specific examples of alkyl groups are as in embodiment 10 for R 1 Defined as follows.
Embodiment 26: the method according to any of embodiments 1-25b, wherein Ra and Rb are both H.
Embodiment 27: according to any of embodiments 1-25b, wherein one of Ra and Rb is H and the other is selected from halogen, CN, C optionally substituted with halogen 1-3 Alkyl and optionally halogen-substituted C 1-3 Alkoxy, wherein C is optionally substituted by halogen 1-3 Alkyl and optionally halogen-substituted C 1-3 Specific examples of alkoxy groups are shown in embodiments 10 and 11 for R, respectively 1 Defined as follows.
Embodiment 28: according to any of embodiments 1-27, wherein R 4 Is a-3-10 membered heterocycloalkyl, preferably a 3-8 membered heterocycloalkyl, optionally substituted with 1 to 2 groups selected from: -OH, halogen, oxo, -CO 2 H. C optionally substituted by halogen 1-6 Alkyl, C 1-6 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, and C in the substituents 3-6 Cycloalkyl and-4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl), -NH-SO 2 (C 1-3 Alkyl) substitution, wherein m is independently selected from 0, 1 and 2 for each occurrence.
Embodiment 28a: according to embodiment 28, wherein R 4 4-10 membered heterocycloalkyl containing 1 or 2 heteroatoms selected from N and O, for example 4-6 membered monocyclic heterocycloalkyl containing 1 or 2 heteroatoms selected from N and O, 9-10 membered and bicyclic heterocycloalkyl containing 1N heteroatom, 7-8 membered bridged heterocycloalkyl containing 1N heteroatom, including the following groups:
In which case X is-O-or-NH- (CH) 2 ) n -, and n is 0;
in which case X is-NH- (CH) 2 ) n -, and n is 1;
wherein Y is selected from O or CH 2
R 4a Independently selected from H, C 1-3 Alkyl, C 1-3 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, wherein C 3-6 Cycloalkyl and 4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl), -NH-SO 2 (C 1-3 Alkyl) substitution, and R 4b Independently selected from H, -OH, halogen, oxo, -CO 2 H. C optionally substituted by halogen 1-3 Alkyl and C 1-3 Hydroxyalkyl, wherein m is selected from 0 and 1;
preferably R 4a Independently selected from H, C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, and R 4b Independently selected from H, wherein m is 0;
more preferably R 4a Selected from H and C 1-3 Alkyl, and R 4b H.
Embodiment 28b: according to embodiment 28 or 28a, wherein R 4 The stereochemistry of the carbon atoms as described above and labeled "+" (R) or (S); preferably, when Z is-O-or-NH- (CH) 2 ) n-and n=0, the stereochemistry of the carbon atom labeled "×" is (R).
Embodiment 28c: according to embodiment 28, 28a or 28b, whichR in (B) 4 Has a structure selected from the group consisting of:
/>
in which case X is-O-or-NH- (CH) 2 ) n -, and n is 0; or (b)
When X is-NH- (CH) 2 ) n -and n is 1, R 4 -CH in X to which it is attached 2 -together have a structure selected from:
Wherein Y is selected from O or CH 2
R 4a Independently selected from H, C 1-3 Alkyl, C 1-3 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, wherein C 3-6 Cycloalkyl and 4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl), -NH-SO 2 (C 1-3 Alkyl) substitution, and R 4b Independently selected from H, -OH, halogen, oxo, -CO 2 H. C optionally substituted by halogen 1-3 Alkyl and C 1-3 Hydroxyalkyl, wherein m is selected from 0 and 1;
preferably R 4a Selected from H, C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, and R 4b Is H, wherein m is 0;
more preferably R 4a Independently selected from H and C 1-3 Alkyl, and R 4b H.
Embodiment 28d: according to embodiments 28, 28a, 28b or 28c, wherein R 4 Has a structure selected from the group consisting of:where n=0, r 4a Selected from H and C 1-3 Alkyl, and R 4b H.
Embodiment 29: according to any of embodiments 1-27, wherein R 4 is-C 3-6 Cycloalkyl optionally substituted with 1 to 3 groups selected from: -OH, halogen, oxo, -CO 2 H. C optionally substituted by halogen 1-6 Alkyl, C 1-6 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, and C in the substituents 3-6 Cycloalkyl and-4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl), -NH-SO 2 (C 1-3 Alkyl) substitution, wherein m is selected from 0, 1 and 2.
Embodiment 29a: according to embodiment 29, wherein R 4 Is cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, optionally substituted by 1 to 3 groups selected from: -OH, halogen and optionally halogen-substituted C 1-3 An alkyl group.
Embodiment 29b: according to embodiment 29 or 29a, wherein R 4 Selected from the following structures:
/>
wherein R is 4c 、R 4c’ And R is 4d Each independently selected from H, -OH, halogen and C optionally substituted with halogen 1-3 An alkyl group;
preferably R's attached to the same carbon atom 4c And R is 4c’ One is OH and the other is selected from H, halogen and C optionally substituted by halogen 1-3 Alkyl, R 4d Selected from H, halogen and C optionally substituted by halogen 1-3 An alkyl group;
more preferably, R's bound to the same carbon atom 4c And R is 4c’ One of OH and the other H or C 1-3 Alkyl, R 4d Is H; most preferably, R's attached to the same carbon atom 4c And R is 4c’ One of OH and the other C 1-3 Alkyl, R 4d H.
Embodiment 29c: according to embodiments 29, 29a or 29b, wherein R 4 Selected from the following structures:
wherein R is 4c Selected from H, halogen and C optionally substituted by halogen 1-3 Alkyl, preferably C 1-3 An alkyl group.
Embodiment 29d: according to embodiments 29, 29a, 29b or 29c, wherein R 4 Selected from the following structures:
preferably
Wherein R is 4c Selected from H, halogen and C optionally substituted by halogen 1-3 Alkyl, preferably C 1-3 An alkyl group.
Embodiment 29e: according to any embodiment 29-29d, wherein X is-O-or-NH- (CH) 2 ) n -, and n is 0.
Embodiment 30: according to any of embodiments 1-3 and 8-27, wherein A is selected from phenyl and a 5-6 membered heteroaryl ring, and R 4 is-C 3-6 Cycloalkyl, wherein A is phenyl, as defined in embodiment 4, A is a 5-6 membered heteroaromatic ring, as defined in embodiment 5 or 5a, as R 4 C of (2) 3-6 Cycloalkyl is as defined in any one of embodiments 29 to 29 e.
Embodiment 31: according to embodiment 1, the present invention provides a compound of formula (I), a stable isotopic variant, a pharmaceutically acceptable salt or solvate:
wherein, the liquid crystal display device comprises a liquid crystal display device,
x is-NH- (CH) 2 ) n -or-O-;
wherein the method comprises the steps ofSelected from: />
Optionally oxo, halogen, C 1-3 Alkyl-or halogen-substituted C 1-3 Alkyl substitution;
R 1 selected from halogen, C 1-6 Alkyl, C 1-6 Alkoxy and C 1-6 Alkylthio group, wherein C 1-6 Alkyl, C 1-6 Alkoxy or C 1-6 Alkylthio groups are each independently at each occurrence optionally substituted with halogen;
R 2 selected from H and C optionally substituted by halogen 1-3 An alkyl group;
R 3 selected from-OH, -NHSO 2 -R 3a 、-NH-SO-R 3a 、-NH-CO-R 3a 、-CH(R 3b )(SO 2 -R 3a )、-CH(R 3b )(SO-R 3a )、-CH(R 3b )(CO-R 3a ) and-CH (R) 3c ) 2
R 3a At each occurrence is-C optionally substituted with halogen 1-6 An alkyl group;
R 3b independently at each occurrence selected from H, halogen and optionally halogenSubstituted C 1-6 An alkyl group;
R 3c independently at each occurrence selected from the group consisting of halogen and optionally halogen-substituted-C 1-6 An alkyl group;
ra and Rb are each H;
R 4 selected from the group consisting of-3-8 membered heterocycloalkyl and-C 3-6 Cycloalkyl groups, each optionally substituted with a group selected from: -OH, C optionally substituted by halogen 1-6 Alkyl, C 1-6 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, wherein C 3-6 Cycloalkyl and 4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl), -NH-SO 2 (C 1-3 Alkyl) substitution; and is also provided with
Wherein n is selected from 0, 1 or 2;
m is independently selected from 0, 1 or 2 at each occurrence.
Embodiment 32: according to embodiment 31, wherein:
x is-NH- (CH) 2 ) n -or-O-, wherein n is selected from 0 and 1;
wherein the method comprises the steps ofSelected from: />
R 1 Selected from halogen, C 1-6 Alkyl and C 1-6 Alkoxy group, wherein C 1-6 Alkyl and C 1-6 Alkoxy groups are each independently optionally substituted with halogen;
R 2 is H;
R 3 selected from-OH, -NHSO 2 -R 3a and-CH (R) 3c ) 2
R 3a is-C optionally substituted by halogen 1-6 An alkyl group;
R 3c independently at each occurrence selected from halogen;
ra and Rb are each H;
R 4 selected from the group consisting of
In which case X is-O-or-NH- (CH) 2 ) n -, and n is 0;
in which case X is-NH- (CH) 2 ) n -, and n is 1; and
in which case X is O or-NH- (CH) 2 ) n -, and n is 0;
wherein Y is selected from O or CH 2
R 4a Independently selected from H, C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, wherein C 3-6 Cycloalkyl and 4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl) or-NH-SO 2 (C 1-3 Alkyl) substitution, wherein m is 0 or 1, preferably R 4a Independently selected from H and C 1-3 An alkyl group, a hydroxyl group,
R 4b and R is 4d In the presence of a hydrogen atom, which is H,
R 4c and R is 4c’ Each independently selected from H, -OH, halogen and optionally halogenSubstituted C 1-3 An alkyl group; preferably R 4c And R is 4c’ One of OH and the other is selected from C 1-3 An alkyl group.
Embodiment 33: the method according to any of embodiments 31 and 32, wherein:selected from: /> Preferably->
Embodiment 34: according to any of embodiments 31-33, wherein R 1 Selected from halogen, C 1-3 Alkyl and C 1-3 Alkoxy group, wherein C 1-3 Alkyl and C 1-3 The alkoxy groups are each independently substituted with halogen.
Embodiment 35: according to any of embodiments 31-34, wherein R 3 is-OH.
Embodiment 36: according to any of embodiments 31-35, wherein R 4 Selected from:
in which case X is-O-or-NH- (CH) 2 ) n -, and n is 0; or (b)
When X is-NH- (CH) 2 ) n -and n is 1, R 4 -CH in X to which it is attached 2 -together formOr R is 4 Selected from the group consisting of
In which case X is O or-NH- (CH) 2 ) n -and n is 0;
wherein, the liquid crystal display device comprises a liquid crystal display device,
R 4a independently selected from H, C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, m is 0 or 1; preferably R 4a Independently selected from H and C 1-3 An alkyl group, a hydroxyl group,
R 4b in the presence of a hydrogen atom, which is H,
R 4c selected from H and C 1-3 Alkyl, preferably C 1-3 An alkyl group.
Embodiment 37: according to any of embodiments 31-36, wherein R 4 Selected from:
in which case X is O or-NH- (CH) 2 ) n -and n is 0,
wherein R is 4a Selected from H and C 1-3 An alkyl group, a hydroxyl group,
R 4b is H, and
R 4c selected from H and C 1-3 Alkyl, preferably C 1-3 An alkyl group.
Embodiment 38: a compound, stable isotopic variant, pharmaceutically acceptable salt or solvate thereof:
/>
/>
embodiment 39: according to any of the preceding embodimentsA compound of the scheme wherein one or more H atoms present may be replaced by deuterium atoms; for example, in one embodiment, a hydrogen atom carried on a ring atom of the a ring and/or a hydrogen atom on an alkyl substituent of the a ring when it carries an alkyl substituent may be replaced with a deuterium atom; in another embodiment, R 4 Hydrogen atoms carried on ring atoms of substituents and/or when R 4 The hydrogen atoms on the alkyl substituents of the alkyl substituents may be replaced by deuterium atoms when carrying the alkyl substituents.
It is to be noted that the compounds of the present invention encompass each individual embodiment or each specific embodiment described above, as well as any combination or sub-combination of the individual embodiments or specific embodiments described above, as well as any combination of any of the preferred or exemplary embodiments described above.
Advantageous effects of the invention
As previously described, NLRP3 inflammatory bodies are known to play a role in autoimmune diseases, inflammatory diseases, and a variety of other diseases. We have surprisingly found that the compounds of formula (I) as defined herein are capable of potently inhibiting NLRP3 inflammatory bodies, and thus have prophylactic and therapeutic medicinal value for diseases in which abnormal activation of NLRP3 inflammatory bodies contributes to the pathology and/or symptoms and/or progression of the disease. In particular, the compounds of the invention are expected to be useful in the prevention or treatment of diseases that are susceptible to inhibition by NLRP3 inflammatory bodies. In addition, the compounds of the invention are expected to be useful in the prevention or treatment of those diseases mediated alone or in part by NLRP3 inflammatory bodies.
Specifically, the compound of the invention can effectively inhibit the activity of NLRP3 inflammatory corpuscles, and can realize one or more of the following technical effects:
strong inhibitory activity on NLRP3 inflammatory bodies: IC50 in NLRP3 inflammasome inhibition assay experiments showed an IC50 in the range of 0.1nM to 0.5 μm, preferably in the range of 0.5nM to 0.2 μm, more preferably in the range of 1nM to 50nM, most preferably less than 20nM, as verified in biological example 1;
have good metabolic stability of the drug For example with longer t 1/2 Lower purge rates, e.g. to increase
The dosing interval, reduced dosing, resulted in better patient compliance as demonstrated in biological example 2;
have improved pharmacokinetic forms, have better drug formation, higher bioavailability, as implemented by the following biology
Verification of example 3;
excellent physicochemical properties such as solubility, physical and/or chemical stability;
significantly improved safety, significantly reduced toxicity and/or side effects, such as minimized or absent hERG inhibition, as follows
As verified in biological example 4.
Based on the beneficial effects of the compound, the invention also provides the following technical schemes.
Compounds of the invention for use in therapy or as medicaments
As mentioned above, a number of genetic and pharmacological evidence have shown the importance of NLRP3 inflammatory bodies in human diseases, which are associated with the pathogenesis of a number of complex diseases or disorders.
For example, evidence is enumerated in WO2020021447A1, the entire contents of which are incorporated herein by reference, to demonstrate that: the role of NLRP3 inflammatory body-induced IL-1 and IL-18 in inflammatory responses associated with, or arising from, a number of different disorders, and indicate that NLRP3 mutations have been found to cause a rare group of auto-inflammatory diseases known as CAPS including: familial Cold Autoinflammatory Syndrome (FCAS), mu Kele-Weirles (Mucke-Wells) syndrome (MWS), and chronic infant cutaneous neurogenic joint syndrome (CINCA; also known as neonatal onset multisystem inflammatory disease NOMID), and all have been shown to result from functionally acquired mutations in the NLRP3 gene that lead to increased secretion of IL-1β. NLRP3 is also implicated in a number of auto-inflammatory diseases including suppurative arthritis, pyodermia gangrenosum and acne (PAPA), shewlet's syndrome, chronic non-bacterial osteomyelitis (CNO) and acne vulgaris;
The literature cited in WO2020021447A1 also shows that many autoimmune diseases are involved in NLRP3, including in particular multiple sclerosis, type 1 diabetes (T1D), psoriasis, rheumatoid Arthritis (RA), behcet's disease, schner's syndrome (Schnitzler syndrome), macrophage activation syndrome, systemic lupus erythematosus and its complications such as lupus nephritis, and systemic sclerosis; also included are a number of lung diseases including Chronic Obstructive Pulmonary Disorder (COPD), asthma (including steroid resistant asthma), asbestosis, and silicosis; also included are a variety of central nervous system disorders including multiple sclerosis (M S), parkinson's disease (P D), alzheimer's disease (A D), dementia, huntington's disease, cerebral malaria, brain damage caused by pneumococcal meningitis, intracranial aneurysms, and traumatic brain injury; also included are various metabolic disorders, including type 2 diabetes (T2D) and its organ specific complications, atherosclerosis, obesity, gout, pseudogout, metabolic syndrome, and nonalcoholic steatohepatitis. There is also evidence that NLRP3 plays a role in the production of IL-1β in: atherosclerosis, myocardial infarction, heart failure, aortic aneurysm, dissection and other cardiovascular events.
Other diseases that have been shown to be involved in NLRP3 include: ocular diseases such as wet and dry age-related macular degeneration, diabetic retinopathy, noninfectious uveitis, and optic nerve damage; liver diseases including nonalcoholic steatohepatitis (NASH) and acute alcoholic hepatitis; inflammatory reactions in the lungs and skin, including contact hypersensitivity, atopic dermatitis, hidradenitis suppurativa, and sarcoidosis; inflammatory response in the joint; amyotrophic lateral sclerosis; cystic fibrosis; chronic kidney disease; and inflammatory bowel disease, including ulcerative colitis and Crohn's disease. NLRP3 inflammatory corpuscles have been found to be activated in response to oxidative stress. NLRP3 has also been shown to be involved in inflammatory hyperalgesia.
Activation of NLRP3 inflammatory bodies has been shown to enhance certain pathogenic infections such as influenza and leishmaniasis.
Thus, in connection with the research work that the present invention has completed, another aspect of the present invention provides a compound of the present invention for use in therapy.
In a specific embodiment, the invention provides a compound of the invention for use as a medicament, in particular as an inhibitor of NLRP3 inflammatory bodies, more particularly as a therapeutic or prophylactic medicament for NLRP3 inflammatory body-related diseases.
In particular embodiments, the NLRP3 inflammatory body-related disease is a disease in which NLRP3 signaling contributes to the pathology and/or symptoms and/or progression of the disease, i.e., the NLRP3 inflammatory body contributes to the occurrence and progression of the disease or inhibits the occurrence of the disease, reduces or eliminates the incidence of the disease, e.g., a disease mediated substantially or entirely by NLRP3 inflammatory body activity, and/or NKRP 3-induced IL-1 β and/or NLRP 3-induced IL-18, such as, but not limited to: immune diseases, inflammatory diseases, such as, for example, auto-inflammatory fever syndrome (e.g., leng Yan element-related periodic syndrome), sickle cell disease, systemic Lupus Erythematosus (SLE), liver-related diseases/disorders (e.g., chronic liver disease, viral hepatitis, nonalcoholic steatohepatitis (NASH), alcoholic steatohepatitis, and alcoholic liver disease), inflammatory arthritis-related disorders (e.g., gout, pseudogout (chorea) osteoarthritis, rheumatoid arthritis, joint diseases such as acute, chronic joint disease), kidney-related diseases (e.g., hyperoxalic acid urine disease, lupus nephritis, type I/II diabetes and related complications (e.g., nephropathy, retinopathy), hypertensive nephropathy, hemodialysis-related inflammation), neuroinflammation-related diseases (e.g., multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, alzheimer's disease), cardiovascular/metabolic diseases/disorders (e.g., reduced cardiovascular risk (CvRR), hypertension, atherosclerosis, type I and II diabetes and related complications (padd), acute aging), atherosclerosis, acute aging, skin diseases (e.g., aging), atherosclerosis, scar formation, acne-related conditions, and conditions. In particular, auto-inflammatory fever syndrome (e.g., CAPS), sickle cell disease, type I/II diabetes and related complications (e.g., nephropathy, retinopathy), gout, pseudogout (chondrocalcareous pigmentation disease), chronic liver disease, NASH, neuroinflammation related disorders) such as multiple sclerosis, brain infections, acute injury, neurodegenerative diseases, alzheimer's disease), atherosclerosis and cardiovascular risk (e.g., reduced cardiovascular risk (CvRR), hypertension), hidradenitis suppurativa, wound healing and scarring, and cancer related diseases/disorders (e.g., colon cancer, lung cancer, myeloproliferative neoplasms, leukemia, myelodysplastic syndrome (MDS), myelofibrosis).
In particular, the present invention provides compounds of the invention for use as a therapeutic or prophylactic agent for the following NLRP3 inflammatory smallbody related diseases: autoinflammatory fever syndrome (e.g., CAPS), sickle cell disease, type I/II diabetes and related complications (e.g., nephropathy, retinopathy), hyperoxaluria, gout, pseudogout (chondrocalcareous pigmentation), chronic liver disease, NASH, neuroinflammation related disorders (e.g., multiple sclerosis, brain infection, acute injury, neurodegenerative disease, alzheimer's disease), atherosclerosis and cardiovascular risk (e.g., reduced cardiovascular risk (CvRR), hypertension), suppurative sweat gland, wound healing and scarring, and cancer (e.g., colon cancer, lung cancer, myeloproliferative neoplasms, leukemia, myelodysplastic syndrome (MDS), myelofibrosis).
Pharmaceutical composition and administration thereof
In another aspect, the compounds of the present invention may be formulated as pharmaceutical compositions according to standard pharmaceutical practice for therapeutic or prophylactic purposes. Meanwhile, based on the good pharmacokinetic property, the improved AUC0-last and the good drug formation of the compound, the compound can be used for preparing the drug with better pharmacokinetic property and higher bioavailability.
Accordingly, the present invention provides a pharmaceutical composition comprising an effective amount of a compound of the present invention as described above and a pharmaceutically acceptable excipient.
The choice of excipients included in a particular composition will depend on a variety of factors, such as the mode of administration and the form of the composition provided. Suitable pharmaceutically acceptable excipients are well known to those skilled in the art and are described, for example, in Ansel, howard C., et al, ansel's Pharmaceutical Dosage Forms and Drug Delivery systems, philadelphia: lippincott, williams & Wilkins,2004, including, for example, adjuvants, diluents (e.g., glucose, lactose or mannitol), carriers, pH adjusting agents, buffers, sweeteners, fillers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, flavoring agents, other known additives.
The pharmaceutical compositions of the invention may be formulated by techniques known to those skilled in the art, such as those disclosed in Remington' sPharmaceutical Sciences, 20 th edition.
The pharmaceutical compositions of the present invention may be administered in a standard manner. For example, suitable modes of administration include oral, intravenous, rectal, parenteral, topical, transdermal, ocular, nasal, buccal, or pulmonary (inhalation), wherein parenteral infusion includes intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. For these purposes, the compounds of the present invention may be formulated by methods known in the art into the form of, for example, tablets, capsules, syrups, powders, granules, aqueous or oily solutions or suspensions, (lipid) emulsions, dispersible powders, suppositories, ointments, creams, drops, aerosols, dry powder formulations and sterile injectable aqueous or oily solutions or suspensions.
The size of the prophylactic or therapeutic dose of a compound of the invention will vary depending on a number of factors, including the severity of the individual, disorder or condition being treated, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. For the treatment of a particular disease, an effective amount is an amount of drug sufficient to ameliorate or alleviate symptoms associated with the disease. Such amounts may be administered as a single dose or may be administered according to an effective treatment regimen. Generally, an effective dose is about 0.0001 to about 5000mg, e.g., about 0.01 to about 1000 mg/kg/day per kg body weight per day (single or divided administration). For a 70kg person, this amounts to about 0.007 mg/day to about 7000 mg/day, for example about 0.7 mg/day to about 1500 mg/day. Depending on the mode of administration, the compounds of the invention may be present in the pharmaceutical composition in an amount of about 0.01mg to about 1000mg, suitably 0.1 to 500mg, preferably 0.5 to 300mg, more preferably 1 to 150mg, particularly preferably 1 to 50mg, for example 1.5mg, 2mg, 4mg, 10mg, 25mg, etc.; accordingly, the pharmaceutical composition of the invention will comprise from 0.05 to 99% w/w (weight percent), such as from 0.05 to 80% w/w, such as from 0.10 to 70% w/w, such as from 0.10 to 50% w/w of the compound of the invention, all weight percentages being based on the total composition. It will be appreciated that it may be necessary in some circumstances to use doses beyond these limits.
In one embodiment, the present invention provides a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients, the composition being formulated for oral administration. The composition may be provided in unit dosage form, for example in the form of a tablet, capsule or oral liquid formulation. Such unit dosage forms may contain from 0.1mg to 1g, for example from 5mg to 250mg, of a compound of the invention as active ingredient.
In one embodiment, the present invention provides a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients, the composition being formulated for topical administration. Topical administration may be in the form of, for example, a cream, lotion, ointment or transdermal patch, where the concentration of the compound of the present invention may be from about 0.01 to 100mg per gram of carrier.
In one embodiment, the present invention provides a pharmaceutical composition comprising a compound of the present invention and one or more pharmaceutically acceptable excipients, the composition being formulated for administration by inhalation. Inhaled administration may be by oral inhalation or by intranasal administration. When administered by oral inhalation, the compounds of the present invention may be effectively used in the present invention in daily doses, for example up to 500 μg, such as 0.1-50 μg, 0.1-40 μg, 0.1-30 μg, 0.1-20 μg or 0.1-10 μg of the compounds of the present invention. The pharmaceutical compositions of the invention for oral inhalation may be formulated as a dry powder, suspension (in liquid or gas) or solution (in liquid) and may be administered in any suitable form and using any suitable inhaler device known in the art including, for example, metered Dose Inhalers (MDI), dry Powder Inhalers (DPI), nebulizers and soft mist inhalers. Multi-chamber devices can be used to deliver the compounds of the present description and one or more other active ingredients (when present).
Pharmaceutical combination
The term "pharmaceutical combination" or "combination therapy" as used herein means that the compounds of the present invention may be combined with other active agents for the purposes of the present invention. The other active agent may be one or more additional compounds of the present invention, or may be a second or additional (e.g., third) compound that is compatible with, i.e., does not adversely affect each other, or has complementary or enhanced activity. Such agents are suitably present in combination in an amount effective to achieve the intended purpose. The other active agents may be co-administered with the compounds of the present invention in a single pharmaceutical composition or may be administered separately in separate discrete units from the compounds of the present invention, either simultaneously or sequentially when administered separately. The successive administrations may be close or distant in time.
Thus, in one aspect, the present invention provides a product comprising a compound as defined herein and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy. In one embodiment, the therapy is the treatment or prevention of NLRP3 inflammatory minibody related diseases. The products provided as a combined preparation include compositions comprising a compound of the invention and one or more other therapeutic agents in the same pharmaceutical composition, or comprising a compound of the invention and one or more other therapeutic agents in separate forms.
In one embodiment, the present invention provides a pharmaceutical combination comprising a compound of the present invention and one or more other therapeutic agents, and optionally a pharmaceutically acceptable carrier as described above.
In one embodiment, the invention provides a kit comprising two or more separate pharmaceutical compositions (e.g., compositions of different dosage forms, such as oral and parenteral), at least one of which comprises a compound of the invention. In one embodiment, the kit comprises means for separately retaining the composition, such as a container, a separate bottle, or a separate foil packet. Instructions for use. Examples of such a kit are blister packs, as are typically used for packaging tablets, capsules and the like.
In the combination therapies of the invention, the compounds of the invention and the other therapeutic agent may be produced and/or formulated by the same or different manufacturers.
The invention thus also provides the use of a compound of the invention in the treatment or prevention of a disease associated with a small inflammatory group of NLRP3, wherein the compound of the invention is administered in combination with one or more other therapeutic agents; and the use of a pharmaceutical combination comprising a compound of the invention and one or more other active agents in the treatment or prevention of NLRP3 inflammatory smalls associated diseases.
The invention thus also provides the use of a compound of the invention in the manufacture of a medicament for the treatment or prophylaxis of a disease associated with NLRP3 inflammatory small body disorders, wherein the compound of the invention is administered in combination with one or more other therapeutic agents; and the use of a pharmaceutical combination comprising a compound of the invention and one or more other active agents in the manufacture of a medicament for the treatment or prophylaxis of a disease in which NLRP3 inflammation is small.
Other active agents that may be used in combination with the compounds of the present invention are therapeutic agents useful in the treatment of inflammatory small-related diseases/disorders, immune diseases, inflammatory diseases, autoimmune diseases, or auto-inflammatory diseases as described herein.
Therapeutic methods and uses
Based on the beneficial effects of the compounds of the invention described above, the compounds of the invention are useful for inhibiting NLRP3 inflammatory body activity, and for treating, preventing or alleviating NLRP3 inflammatory body-related diseases in animals, particularly mammals such as humans, i.e., diseases in which NLRP3 inflammatory bodies contribute to the occurrence and progression of the disease and/or inhibiting NLRP3 inflammatory bodies would reduce the incidence of the disease, reduce or eliminate disease conditions, e.g., diseases mediated by IL-1β and/or NLRP3 induced IL-18, substantially or entirely by NLRP3 inflammatory body activity, and/or NLRP3 induced IL-1β and/or IL-18, preferably diseases responsive to reduced IL-1β and/or IL-18 levels.
Thus, in another aspect, the invention provides a method of modulating, especially inhibiting, the activity of an NLRP3 inflammatory body in a cell, the method comprising contacting the cell with a compound of the invention described herein to modulate, especially inhibit, the activity of an NLRP3 inflammatory body in a cell.
Based on the same properties, the present invention accordingly provides a method of inhibiting IL-1. Beta. And/or IL-18 levels in a mammal comprising administering to said mammal a therapeutically effective amount of a compound of the invention described herein, or a pharmaceutical composition comprising the same.
In another aspect, the invention provides a method of preventing or treating a disease associated with a small inflammatory NLRP3 disorder, comprising administering to a subject in need thereof an effective amount of a compound of the invention described herein or a pharmaceutical composition of the invention comprising the same.
In another aspect, the invention provides the use of a compound of the invention described herein, or a pharmaceutical composition comprising the same, for inhibiting NLRP3 inflammatory body activity. In a specific embodiment, the invention provides the use of a compound of the invention or a pharmaceutical composition comprising the same as described herein for reducing IL-1 β and/or IL-18 levels.
In another aspect, the invention provides the use of a compound of the invention described herein, or a pharmaceutical composition comprising the same, for the treatment and/or prevention of diseases associated with NLRP3 inflammatory bodies.
In another aspect, the invention also provides the use of a compound of the invention described herein or a pharmaceutical composition comprising the same in the manufacture of a medicament for inhibiting NLRP3 inflammatory small body activity, preferably for reducing IL-1 β and/or IL-18 levels.
In another aspect, the invention provides the use of a compound of the invention described herein, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for the treatment or prevention of a disease associated with NLRP3 inflammation. In particular embodiments, the compound or pharmaceutical composition is optionally used in combination with one or more chemotherapy or immunotherapy.
For the methods of treatment and uses provided by the invention described above, wherein the NLRP3 inflammatory small-scale associated disease is such as, but not limited to: immune diseases, inflammatory diseases, e.g., auto-inflammatory fever syndrome (e.g., leng Yan element-related periodic syndrome), sickle cell disease, systemic Lupus Erythematosus (SLE), liver-related diseases/disorders (e.g., chronic liver disease, viral hepatitis, non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis, and alcoholic liver disease), inflammatory arthritis-related disorders (e.g., gout, pseudogout (chocolatopsis), osteoarthritis, rheumatoid arthritis, joint diseases such as acute, chronic joint disease), kidney-related diseases (e.g., hyperoxalic urine disease, lupus nephritis, type I/II diabetes mellitus and related complications (e.g., nephropathy, retinopathy), hypertensive nephropathy, hemodialysis-related inflammation), neuroinflammation-related diseases (e.g., multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, alzheimer's disease), cardiovascular/metabolic diseases/disorders (e.g., cardiovascular risk reduction (CvRR), hypertension, atherosclerosis, type I and II diabetes and related complications (padd), acute and peripheral), aging, atherosclerosis, skin diseases (e.g., aging), atherosclerosis, macular degeneration, scar formation, acne-related conditions, and scar formation of the like). In particular, auto-inflammatory fever syndrome (e.g., CAPS), sickle cell disease, type I/II diabetes and related complications (e.g., nephropathy, retinopathy), gout, pseudogout (chondrocalcareous pigmentation disease), chronic liver disease, NASH, neuroinflammation related disorders) such as multiple sclerosis, brain infections, acute injury, neurodegenerative diseases, alzheimer's disease), atherosclerosis and cardiovascular risk (e.g., reduced cardiovascular risk (CvRR), hypertension), hidradenitis suppurativa, wound healing and scarring, and cancer related diseases/disorders (e.g., colon cancer, lung cancer, myeloproliferative neoplasms, leukemia, myelodysplastic syndrome (MDS), myelofibrosis).
In particular, the NLRP3 inflammatory small-body related disease is selected from the group consisting of: autoinflammatory fever syndrome (e.g., CAPS), sickle cell disease, type I/II diabetes and related complications (e.g., nephropathy, retinopathy), hyperoxaluria, gout, pseudogout (chondrocalcareous pigmentation), chronic liver disease, NASH, neuroinflammation related disorders (e.g., multiple sclerosis, brain infection, acute injury, neurodegenerative disease, alzheimer's disease), atherosclerosis and cardiovascular risk (e.g., reduced cardiovascular risk (CvRR), hypertension), suppurative sweat gland and wound healing and scarring, and cancer (e.g., colon cancer, lung cancer, myeloproliferative neoplasms, leukemia, myelodysplastic syndrome (MDS), myelofibrosis).
Process for the preparation of the compounds of the invention
The compounds of the invention may be prepared by a variety of methods including the methods set forth in the schemes below, the methods set forth in the examples, or the like. Suitable reaction conditions for the individual reaction steps are known to the person skilled in the art or can be readily determined. The starting materials are generally commercially available or can be readily prepared using methods well known in the art or as described herein. The variables in the formula have the meanings defined herein unless otherwise indicated.
For illustrative purposes only, the following schemes provide exemplary routes for synthesizing the compounds of the present application. Those skilled in the art will appreciate that other synthetic pathways are available and that compounds prepared by the methods described below may be further modified in accordance with the teachings of the present application using conventional compounds well known to those skilled in the art.
In the preparation of the compounds of the application, protection of the groups (e.g. amino protecting groups, hydroxyl protecting groups) may be desired, which can be readily determined by a person skilled in the art. For a general description of protecting groups and their use, see T.W. Greene, protective Groups in Organic Synthesis, john Wiley & Sons, new York, 1991. The starting materials and reagents used in the preparation of these compounds are generally commercially available, if not specified, or may be prepared by the methods set forth below, analogous to the methods set forth below, or known in the art.
If desired, the starting materials and intermediates in the synthetic reaction scheme may be isolated and purified using conventional techniques including, but not limited to, filtration, distillation, crystallization, chromatography, and the like. The materials may be characterized using conventional methods including physical constants and spectroscopic data.
Synthesis scheme 1:
synthesis scheme 1 of the present invention provides an exemplary method of synthesizing a compound of formula (I) wherein R 1 、R 2 、R 3 、R 4 Variables X and fused ring a are as defined herein for compounds of formula (I), comprising the steps of:
step a: allowing a compound of formula (I-1) to react with a functional group X-R 4 Ligation provides a compound of formula (I-2). Such reactions include, but are not limited to, nucleophilic substitution reactions. Nucleophilic substitution reactions can be performed in the presence or absence of bases including, but not limited to, na 2 CO 3 、K 2 CO 3 CsF, DIPEA. In particular, these reactions are preferably carried out in a suitable organic solvent, which may be selected from NMP, DMF, DMSO, etOH and any combination thereof. These reactions are preferably carried out at a suitable temperature, for example 0-200 ℃, 10-100 ℃, 20-50 ℃ or room temperature (about 20-25 ℃).
Step b: linking the compound of formula (I-2) with a substituted phenyl group gives the compound of formula (I). Such reactions include, but are not limited to, suzuki coupling reactions. Suzuki coupling Using PdCl 2 (dppf)、Pd(dtbpf)Cl 2 RuPhos Pd G3, XPhos Pd G3, etc. as catalysts. The Suzuki coupling reaction can be carried out in the presence of alkaliIn the following, the base includes but is not limited to Na 2 CO 3 、K 2 CO 3 、K 3 PO 4 DIPEA. In particular, these reactions are preferably carried out in a suitable organic solvent, which may be selected from the group consisting of a mixture of DCE and water, a mixture of dioxane and water, and any combination thereof. These reactions are preferably carried out at a suitable temperature, for example 0-200 ℃, 10-100 ℃, 20-50 ℃ or room temperature (about 20-25 ℃).
Detailed Description
The following examples are provided to further illustrate the application. It should be understood that this is merely to enable a better understanding of the application and is not intended to limit the scope of the application in any way.
In the present application, when chemical names and structural formulas are not identical, the structural formulas should be shown unless it can be inferred that the chemical names are correct instead of the structural formulas according to the context. For simplicity, not all hydrogen atoms are explicitly shown in some of the compound formulae presented herein. When free valences are present in a compound, the presence of an unidentified hydrogen atom is indicated.
Abbreviations used herein have the meanings commonly understood in the art unless clearly defined otherwise in the specification. The meanings of abbreviations used in the specification are listed below:
in the preparation method of the target compound, column chromatography adopts silica gel (100-200 meshes and 200-300 meshes) produced by Shanghai Taitan technology Co., ltd; thin layer chromatography using GF254 (0.25 mm); nuclear magnetic resonance chromatography (NMR) was performed using a Bruker Avance III 400MHz NMR; LC/MS using an Agilent 1200 LC-20AP for preparative HPLC.
LC-MS
LC-MS data provided in the examples are given in terms of retention time, purity and/or mass (m/z), obtained using any of the following methods:
method A: agilent 1200HPLC,6110single quadrupole MSD; column: luna C18 (2.0 x 50mm,5 μm); mobile phase a:0.04% tfa in water; mobile phase B, 0.02% tfa in acetonitrile; flow rate: 1.0ml/min; column temperature: 40 ℃; detection wavelength: 214nm,254nm; gradient elution was performed according to table 1:
TABLE 1
Time(min) A% B%
0.00 95 5
0.40 95 5
3.00 5 95
4.00 5 95
4.01 95 5
4.50 95 5
Method B: agilent 1200HPLC,6120single quadrupole MSD; column: XBridge C18 (2.1 x 50mm,5 μm); mobile phase a: h 2 O+10mM NH 4 HCO 3 The method comprises the steps of carrying out a first treatment on the surface of the Mobile phase B: acetonitrile; flow rate: 0.8ml/min; column temperature: 40 ℃; detection wavelength: 214nm,254nm; gradient elution was performed according to table 2:
TABLE 2
Time(min) A% B%
0.00 95 5
3.40 5 95
3.85 5 95
3.86 95 5
4.50 95 5
In addition, all operations involving readily oxidizable or hydrolyzable feedstocks are performed under nitrogen protection. Unless otherwise indicated, the raw materials used in the present invention are all commercially available raw materials, and can be used directly without further purification, and the temperatures used in the present invention are all degrees celsius (deg.c). The proportion of liquid is the volume ratio, for example in PE/EA.
Examples
Example 1:2- (4- (((R) -1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -3, 5-dimethylphenol
Step 1: synthesis of (R) -3- ((4-bromophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
1, 4-dibromophthalazine (765 mg,2.66 mmol), (R) -3-aminopiperidine-1-carboxylic acid tert-butyl ester (532 mg,2.66 mmol) and K 2 CO 3 (441 mg,3.19 mmol) was added to NMP (7.50 mL) and the resulting reaction mixture was stirred at 80℃for 12 hours. The reaction mixture was cooled to room temperature, diluted with EA (20.0 mL), washed successively with water (5.00 mL. Times.3), saturated brine (5.00 mL), and anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification of the residue by Prep-TLC (PE/ea=1/1) gave the title compound (450 mg, yield 41.5%, yellow solid). LC-MS (ESI) m/z 409.2[ M+H ]] +
Step 2: synthesis of tert-butyl (3R) -3- ((4- (2- (methoxymethoxy) -4, 6-dimethylphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate
(R) -3- ((4-bromophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (165 mg,405 mmol), 2- (2- (methoxymethoxy) -4, 6-dimethylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (178 mg,608 mmol), pd (PPh) 3 ) 4 (93.6 mg,81.0 umol) and K 2 CO 3 (168 mg,1.22 mmol) was added to a mixed solvent of ethylene glycol dimethyl ether (2.00 mL) and water (1.00 mL), and the resulting reaction mixture was stirred at 90℃under nitrogen protectionMix for 12 hours. The reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by Prep-TLC (PE/ea=1/1) to give the title compound (190 mg, yield 95.2%, yellow solid). LC-MS (ESI) m/z 493.4[ M+H ] ] +
Step 3: synthesis of 3, 5-dimethyl-2- (4- ((R) -piperidin-3-ylamino) phthalazin-1-yl) phenol
To a solution of tert-butyl (3R) -3- ((4- (2- (methoxymethoxy) -4, 6-dimethylphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate (80.0 mg, 365. Mu. Mol) in DCM (10.0 mL) at 25℃was added dropwise a solution of HCl-dioxane (4M, 2.00 mL) and the resulting reaction was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure to give the objective compound (150 mg, crude product, yellow solid) which was directly used for the next reaction. LC-MS (ESI) m/z 349.2[ M+H ]] +
Step 4: synthesis of 2- (4- (((R) -1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -3, 5-dimethylphenol
To a solution of 3, 5-dimethyl-2- (4- ((R) -piperidin-3-ylamino) phthalazin-1-yl) phenol (100 mg,287 mmol) in EtOH (2.00 mL) at 0deg.C was added NaBH in sequence 3 CN (90.2 mg,1.43 mmol) and ice AcOH (1.72 mg,28.7 mmol), and the resulting mixture was stirred at 0℃for 5 minutes, followed by dropwise addition of acetaldehyde (37.9 mg,861 mmol) at 0℃and the resulting reaction mixture was stirred at 0℃for 1 hour. The reaction was quenched with water (5.00 mL), the mixture extracted with EA (5.00 mL. Times.3), the organic phases combined, washed with saturated brine (5.00 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column Waters Xbridge BEH C18 100X 30mm X10 um; mobile phase [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -50%; 10 min) to give the title compound (4.76 mg, yield 4.41%, white solid). LC-MS (ESI) m/z 377.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.03(br s,1H),9.41-10.83(m,1H),8.64-9.03(m,1H),8.19(br d,J=6.6Hz,1H),8.08(br t,J=7.4Hz,1H),7.78(br d,J=8.0Hz,1H),7.29(d,J=7.6Hz,1H),6.92(s,1H),6.87(br d,J=7.8Hz,1H),4.45-4.68(m,1H),3.79(br d,J=9.9Hz,1H),3.55(br d,J=11.3Hz,1H),3.14-3.33(m,2H),2.94(br s,2H),2.36(s,3H),2.15-2.25(m,1H),2.15-2.25(m,1H),2.04(br d,J=12.9Hz,1H),1.67-1.97(m,2H),1.26(br t,J=7.2Hz,3H)。
Example 2:(R) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -5-methylphenol
Step 1: synthesis of (R) -3- ((4- (2- (methoxymethoxy) -4-methylphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
(R) -3- ((4-bromophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (500 mg,1.23 mmol), 2- (2- (methoxymethoxy) -4-methylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (376mg, 1.35 mmol), pd (dtbpf) Cl 2 (80.0 mg,123 mmol) and K 2 CO 3 (399 mg,2.46 mmol) was successively added to a mixed solvent of dioxane (5.00 mL) and water (1.00 mL), nitrogen was replaced 3 times, and the resulting reaction mixture was stirred at 90℃for 12 hours under nitrogen atmosphere. The reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by Prep-TLC (PE/ea=1/1) to give the title compound (400 mg, yield 68.1%, yellow solid). LC-MS (ESI) m/z 479.3[ M+H ]] +
Step 2: synthesis of (R) -5-methyl-2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenol
To a solution of tert-butyl (R) -3- ((4- (2- (methoxymethoxy) -4-methylphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate (300 mg,627 mmol) in DCM (5.00 mL) at 25deg.C was added dropwise a solution of HCl-dioxane (4M, 1.00 mL) and the resulting reaction was stirred at 25deg.C for 12 hours. The reaction solution was concentrated under reduced pressure to give the objective compound (200 mg, crude product, yellow solid) which was directly used for the next reaction. LC-MS (ESI) m/z 335.1[ M+H ] ] +
Step 3: synthesis of (R) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -5-methylphenol
To a solution of (R) -5-methyl-2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenol (200 mg,598 mmol) in EtOH (2.00 mL) at 0deg.C, naBH was added sequentially 3 CN (188 mg,2.99 mmol) and ice AcOH (3.59 mg,59.8 mmol), the reaction was obtainedThe solution was stirred at 0deg.C for 5 minutes, then acetaldehyde (79.0 mg,1.79 mmol) was added dropwise at 0deg.C, and the resulting reaction solution was stirred at 0deg.C for 1 hour. The reaction mixture was quenched with water (5.00 mL), extracted with EA (5.00 mL. Times.3), and the organic phases were combined, washed with saturated brine (5.00 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column: phenomnex Luna 80 x 30mm 3um; mobile phase: [ A-water (TFA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 1% -35%;8 min) to give the title compound (39.5 mg, yield 18.24%, white solid). LC-MS (ESI) m/z 363.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm10.03(br s,1H),9.41-10.83(m,1H),8.64-9.03(m,1H),8.19(br d,J=6.6Hz,1H),8.08(br t,J=7.4Hz,1H),7.78(br d,J=8.0Hz,1H),7.29(d,J=7.6Hz,1H),6.92(s,1H),6.87(br d,J=7.8Hz,1H),4.45-4.68(m,1H),3.79(br d,J=9.9Hz,1H),3.55(br d,J=11.3Hz,1H),3.14-3.33(m,2H),2.94(br s,2H),2.36(s,3H),2.15-2.25(m,1H),2.15-2.25(m,1H),2.04(br d,J=12.9Hz,1H),1.67-1.97(m,2H),1.26(br t,J=7.2Hz,3H)。
Example 3:(R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (R) -3- ((4-chlorophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
1, 4-dichlorophthalazine (100 mg,0.502 mmol) was added to a single vial containing NMP (1.00 mL), followed by tert-butyl (R) -3-aminopiperidine-1-carboxylate (101 mg,0.502 mmol) and DIPEA (97 mg,0.754 mmol), and the resulting reaction mixture was stirred overnight under nitrogen at 80 ℃. The reaction mixture was cooled to room temperature, poured into water (10.0 mL), extracted with EA (10.0 mL. Times.2), the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and isolation and purification of the residue by silica gel chromatography (DCM/meoh=10/1) gave the title compound (45 mg, yield 24.7%, yellow solid). LC-MS (ESI) m/z 363.1[ M+H ]] +
Step 2: synthesis of (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
To a three-necked flask containing tert-butyl (R) -3- ((4-chlorophthalazin-1-yl) amino) piperidine-1-carboxylate (35.0 mg,0.0965 mmol) was added dioxane (1.00 mL) and water (0.200 mL), followed by (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (19.9 mg,0.0965 mmol), K 2 CO 3 (26.7 mg,0.193 mmol) and Pd (dtbpf) Cl 2 (6.00 mg, 9.70. Mu. Mol) and the resulting reaction mixture was stirred overnight at 100℃under nitrogen. The reaction mixture was cooled to room temperature, poured into water (5.00 mL), extracted with EA (5.00 mL. Times.2), the organic phases were combined, washed with saturated brine, and the organic phase was washed with anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing prep-HPLC (column: YMC, C18X 20mm,5 μm; mobile phase: [ A-water (0.1% NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 20% -80%;20 min) to give the objective compound (144 mg, yield 35.7%, white solid). LC-MS (ESI) m/z 489.2[ M+H ]] +
Step 3: synthesis of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate (17.0 mg,0.0348 mmol) in DCM (1.00 mL) at room temperature under nitrogen was added dropwise HCl-dioxane (0.300 mL) and the resulting reaction mixture stirred at room temperature under nitrogen for 1 hour. The reaction was dried by spin-drying and the residue was purified by prep-HPLC (Shimadzu LC-20AP; YMC-acts Triart C18X 20.0mm,5um; mobile phase A: water/0.1% FA, mobile phase B: ACN; gradient: 2min 20% B, 20% B to 50% B, holding 2min 50% B, then 1min 5% B, detection wavelength 220nm,254 nm) to give the title compound (7.00 mg, yield 51.7% white solid). LC-MS (ESI) m/z 389.1[ M+H ]] +1 H NMR(400MHz,DMSO-d6)δ10.77(s,1H),8.96–8.83(m,1H),8.81–8.69(m,1H),8.65–8.54(m,1H),8.14–8.04(m,1H),8.04–7.93(m,1H),7.63(d,J=7.9Hz,1H),7.59(d,J=8.1Hz,1H),7.37(d,J=7.9Hz,1H),7.34(s,1H),4.63–4.32(m,1H),3.12–2.91(m,2H),2.46–2.29(m,2H),2.23–2.10(m,1H),2.07–1.94(m,1H),1.84–1.73(m,2H)。
Example 4:(R) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (R) -3- ((4-chlorophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
1, 4-dichlorophthalazine (2.00 g,10.1 mmol) was added to a single vial containing NMP (20.0 mL), and tert-butyl (R) -3-aminopiperidine-1-carboxylate (2.01 g,10.1 mmol) and DIPEA (1.95 g,15.1 mmol) were added sequentially, and the resulting reaction mixture was warmed to 80℃and stirred for 12 hours. The reaction mixture was cooled to room temperature, poured into water (50.0 mL), extracted with EA (50.0 mL. Times.2), the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave crude product which was purified by silica gel chromatography (DCM/meoh=10:1) to give the title compound (1.44 g, 39.5% yield as yellow solid). LC-MS (ESI) m/z 363.1[ M+H ]] +
Step 2: synthesis of (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
To a three-necked flask containing tert-butyl (R) -3- ((4-chlorophthalazin-1-yl) amino) piperidine-1-carboxylate (300 mg,0.827 mmol) was added dioxane (3.00 mL) and water (0.600 mL), followed by (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (170 mg,0.827 mmol), DIPEA (214 mg,1.65 mmol) and Pd (dtbpf) Cl 2 (53.4 mg,0.0827 mmol) and the resulting reaction mixture was stirred overnight at 90℃under nitrogen. The reaction mixture was cooled to room temperature, poured into water (20.0 mL), extracted with EA (20.0 mL. Times.2), the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and the residue was purified by Pre-HPLC (column: YMC, C18X 20mm,5 μm; mobile phase: [ A-water (0.1% NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 20% -80%;20 min) to give the objective compound (144 mg, yield 35.7%, white solid). LC-MS (ESI) m/z 489.1[ M+H ]] +
Step 3: synthesis of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate (144 mg, 0.025 mmol) in DCM (2.00 mL) at room temperature was added dropwise HCl-dioxane (0.600 mL) and the resulting reaction mixture stirred at room temperature under nitrogen for 1 hour and LC-MS monitored the reaction to completion. The reaction solution was concentrated under reduced pressure to give the objective compound (142 mg, crude product, yellow solid) which was directly used for the next reaction. LC-MS (ESI) m/z 389.1[ M+H ]] +
Step 4: synthesis of (R) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol (92.0 mg,0.237 mmol) in 1, 2-dichloroethane (2.00 mL) was added acetaldehyde (15.7 mg,0.355 mmol) and the resulting mixture stirred at room temperature for 2min and NaBH (OAc) was added 3 (99.9 mg,0.474 mmol) and the resulting reaction mixture was stirred at room temperature under nitrogen for 1 hour. The reaction mixture was poured into water (20.0 mL), extracted with DCM (20.0 ml×2), and the organic phases were combined, washed with saturated brine, and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure and isolation of the residue by prep-HPLC (Shimadzu LC-20AP; YMC-acts Triart C18X 20.0mm,5um; mobile phase A: water/0.1% FA, mobile phase B: ACN; gradient: 2min 20% B, 20% B to 50% B, holding 2min 50% B, then 1min 5% B, detection wavelength 220nm,254 nm) gives the title compound (19.0 mg, two step yield 19.3%, white solid). LC-MS (ESI) m/z 417.3[ M+H ] ] +1 H NMR(400MHz,DMSO-d6)δ8.40(d,J=8.2Hz,1H),8.21(s,1H),7.86(t,J=7.5Hz,1H),7.78(t,J=7.6Hz,1H),7.51(d,J=7.7Hz,1H),7.44(d,J=8.2Hz,1H),7.33–7.24(m,2H),7.22–7.10(m,1H),4.51–4.38(m,1H),3.28–3.17(m,1H),2.93–2.80(m,1H),2.50–2.41(m,2H),2.09–1.96(m,3H),1.85–1.72(m,1H),1.70–1.43(m,2H),1.04(t,J=7.1Hz,3H)。
Example 5:2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) phthalazin-1-yl) -5- (trifluormethyl) Radical) phenol
Step 1: synthesis of (1S, 3S) -3- ((4-chlorophthalazin-1-yl) amino) -1-methylcyclobutane-1-ol
1, 4-dichlorophthalazine (500 mg,2.51 mmol) was added to a single-necked flask containing NMP (5.00 mL), and (1S, 3S) -3-amino-1-methylcyclobutane-1-ol (254 mg,2.51 mmol) and DIPEA (974 mg,7.54 mmol) were further added, and the resultant reaction solution was stirred under nitrogen at 80℃for 3 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (30.0 mL), extracted with EA (30.0 mL. Times.2), and the organic phases were combined, washed with saturated brine, and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and isolation and purification of the residue by silica gel chromatography (DCM/meoh=10/1) gave the title compound (500 mg, yield 75.5%, white solid). LC-MS (ESI) m/z 264.1[ M+H ]] +
Step 2: synthesis of 2- (4- (((1S, 3S) -3-hydroxy-3-methylcyclobutyl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a three-necked flask containing (1S, 3S) -3- ((4-chlorophthalazin-1-yl) amino) -1-methylcyclobutan-1-ol (200 mg,0.758 mmol) was added dioxane (2.00 mL) and water (0.400 mL), followed by (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (156 mg,0.758 mmol), DIPEA (196 mg,1.52 mmol) and Pd (dtbpf) Cl 2 (48.9 mg,0.0758 mmol) and the resulting reaction was stirred overnight at 90℃under nitrogen. After the reaction mixture was cooled to room temperature, it was diluted with water (10.0 mL), extracted with EA (10.0 mL. Times.2), and the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and isolation of the residue by prep-HPLC (Shimadzu LC-20AP; YMC-acts Triart C18X 20.0mm,5um; mobile phase A: water/0.1% FA, mobile phase B: ACN; gradient: 2min 20% B, 20% B to 50% B, holding 2min50% B, then 1min 5% B, detection wavelength 220nm,254 nm) gave the title compound (20.3 mg, yield 6.87% white solid). LC-MS (ESI) m/z 390.2[ M+H ]] +1 H NMR(400MHz,DMSO-d6)δ10.34(s,1H),8.43(d,J=8.3Hz,1H),7.86(t,J=7.6Hz,1H),7.78(t,J=7.6Hz,1H),7.69–7.56(m,1H),7.51(d,J=7.8Hz,1H),7.44(d,J=8.2Hz,1H),7.33–7.23(m,2H),5.01(s,1H),4.39–4.11(m,1H),2.49–2.45(m,2H),2.25–2.06(m,2H),1.34(s,3H)。
Example 6:(R)-N- (2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -5-methylphenyl) methanesulfonate Amides and their use
Step 1: synthesis of 5-methyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) aniline
2-bromo-5-methylaniline (10.0 g,53.7 mmol), triethylamine (21.7 g,215 mmol) and Pd (dppf) Cl 2 ·CH 2 Cl 2 (8.78 g,10.7 mmol) was added to dioxane (150 mL), and 4, 5-tetramethyl-1, 3, 2-dioxaborolan (20.6 g,161 mmol) was added dropwise, and the resulting reaction solution was stirred under nitrogen at 120℃for 12 hours. The reaction was cooled to room temperature, diluted with saturated aqueous ammonium chloride (200 mL), extracted with DCM (100 mL. Times.3), the organic phases combined, washed with saturated brine (50.0 mL), anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification of the residue by Prep-TLC (PE/ea=5/1) gave the title compound (5.00 g, yield 39.9%, yellow solid). LC-MS (ESI) m/z 234.3[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 7.26(d,J=7.58Hz,1H),6.39(s,1H),6.31(d,J=7.58Hz,1H),5.41(s,2H),2.15(s,3H),1.21-1.34(m,12H)。
Step 2: synthesis of N- (5-methyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) methanesulfonamide
To a solution of 5-methyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) aniline (2.00 g,8.58 mmol) and triethylamine (4.34 g,42.9 mmol) in DCM (20.0 mL) at 0deg.C under nitrogen was slowly added dropwise methanesulfonyl chloride (1.53 g,13.4 mmol), and the resulting reaction was stirred at 0deg.C for 2 hours. Saturated NaHCO with ice 3 The reaction was quenched with aqueous (50.0 mL), the mixture extracted with DCM (10.0 mL. Times.3), the organic phases combined and washed with saturated brine (10.0 mL)Anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification of the residue by Prep-TLC (PE/ea=5/1) gave the title compound (1.00 g, yield 37.5%, white solid). 1 H NMR(400MHz,DMSO-d 6 )δppm 8.45(br s,1H),7.54(br d,J=7.51Hz,1H),7.18-7.24(m,1H),6.99(br d,J=7.27Hz,1H),3.02(s,3H),2.32(s,3H),1.31(s,12H)。
Step 3: synthesis of (R) -3- ((4- (4-methyl-2- (methylsulfonylamino) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
(R) -3- ((4-bromophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (600 mg,1.47 mmol), N- (5-methyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) methanesulfonamide (917 mg,2.95 mmol), K 2 CO 3 (407 mg,2.95 mmol) and Pd (dtbpf) Cl 2 (192 mg, 294.62. Mu. Mol) was added to dioxane (10.0 mL) and water (2.00 mL), and the resulting reaction mixture was stirred under nitrogen at 100deg.C for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (5.00 mL), extracted with EA (5.00 mL. Times.3), and the organic phases were combined, washed with saturated brine (10.0 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification of the residue by Prep-TLC (PE/ea=1/1) gave the title compound (350 mg, yield 46.4%, yellow solid). LC-MS (ESI) m/z 512.2[ M+H ]] +
Step 4: synthesis of (R) -N- (5-methyl-2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenyl) methanesulfonamide
To a solution of tert-butyl (R) -3- ((4- (4-methyl-2- (methylsulfonylamino) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate (300 mg,586 mmol) in DCM (10.0 mL) at 25deg.C was slowly added dropwise HCl-dioxane (4M, 2.00 mL) and the resulting reaction stirred at 25deg.C for 12 hours. The reaction solution was concentrated under reduced pressure to give the objective compound (250 mg, crude product, yellow solid) which was directly used for the next reaction. LC-MS (ESI) m/z 412.3[ M+H ]] +
Step 5: synthesis of (R) -N- (2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) -5-methylphenyl) methanesulfonamide
To (R) -N- (5-methyl-2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenyl) methanesulfonamide (2) at 0 ℃ 00mg, 4816 mmol) was dissolved in EtOH (5.00 mL) and NaBH was added in sequence 3 CN (91.6 mg,1.46 mmol) and ice AcOH (292 mg,4.86 mmol), the resulting mixture was stirred at 0℃for 5 minutes, then acetaldehyde (5M, 4816 mL) was added dropwise at 0℃and the resulting reaction solution was stirred at 0℃for 1 hour. The reaction mixture was quenched with water (5.00 mL), extracted with EA (5.00 mL. Times.3), and the organic phases were combined, washed with saturated brine (5.00 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column Waters Xbridge BEH C18 100X 30mm X10 um; mobile phase [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -60%;10 min) to give the title compound (23.6 mg, yield 11.0% as yellow solid). LC-MS (ESI) m/z 440.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 9.09(br s,1H),8.40(br d,J=8.2Hz,1H),7.83-7.90(m,1H),7.77(t,J=7.6Hz,1H),7.74-7.82(m,1H),7.52(d,J=8.1Hz,1H),7.43(s,1H),7.27(d,J=7.7Hz,1H),7.10-7.20(m,2H),4.44(br s,1H),3.20(br d,J=8.7Hz,1H),2.90(s,3H),2.85(br d,J=10.4Hz,1H),2.42-2.46(m,1H),2.42-2.46(m,2H),2.42(s,3H),1.90-2.06(m,3H),1.77(br d,J=12.5Hz,1H),1.44-1.68(m,2H),1.03(br t,J=7.0Hz,3H)。
Example 7:(R) 2- (4- ((1-ethylpiperidin-3-yl) amino) pyrido [3,4- ]d]Pyridazin-1-yl) -5- (tris Fluoromethyl) phenol
Step 1: synthesis of (R) -3- ((1-chloropyrido [3,4-d ] pyridazin-4-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
1, 4-dichloropyrido [3,4-d ]]Pyridazine (500 mg,2.50 mmol) was added to a single-necked flask equipped with NMP (5.00 mL), and tert-butyl (R) -3-aminopiperidine-1-carboxylate (501 mg,2.50 mol) and DIPEA (284 mg,3.75 mmol) were further added, and the resulting reaction solution was stirred at 80℃for 4 hours. The reaction solution was cooled to room temperature, poured into water (50.0 mL), extracted with EA (50.0 mL. Times.2), and the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtering, concentrating under reduced pressure, and passing the residue through silica gelChromatography (DCM/meoh=10/1) afforded the title compound (781 mg, yield 85.9%, yellow solid). LC-MS (ESI) m/z 364.1[ M+H ]] +
Step 2: synthesis of tert-butyl (R) -3- ((1- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [3,4-d ] pyridazin-4-yl) amino) piperidine-1-carboxylate
To be filled with (R) -3- ((1-chloropyrido [3, 4-d)]To a three-necked flask of t-butyl pyridazin-4-yl) amino-piperidine-1-carboxylate (340 mg,0.936 mmol) was added dioxane (4.00 mL) and water (0.800 mL), followed by (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (192.8 mg,0.936 mmol), K 3 PO 4 (397 mg,1.872 mmol) and Pd (dtbpf) Cl 2 (60.0 mg,0.0935 mmol) and the resulting reaction mixture was stirred overnight under nitrogen at 90 ℃. The reaction mixture was cooled to room temperature, poured into water (20.0 mL), extracted with EA (20.0 mL. Times.2), the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue on Prep-HPLC (column: YMC, C18X 20mm,5 μm; mobile phase: [ A-water (0.1% NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 20% -80%;20 min) to give the objective compound (298 mg, yield 65.1%, white solid). LC-MS (ESI) m/z 490.4[ M+H ]] +
Step 3: (R) -2- (4- (piperidin-3-ylamino) pyrido [3, 4-d) ]Synthesis of pyridazin-1-yl) -5- (trifluoromethyl) phenol to (R) -3- ((1- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [3,4-d ] at room temperature]To a solution of t-butyl pyridazin-4-yl) amino-piperidine-1-carboxylate (181 g,688 mmol) in DCM (4.00 mL) was added dropwise a solution of HCl-dioxane (4.00 mL), and the resulting reaction mixture was stirred at room temperature under nitrogen for 1 hour, and LC-MS monitored for the end of the reaction. The reaction solution was concentrated under reduced pressure to give the objective compound (230 mg, crude product, yellow solid). LC-MS (ESI) m/z 390.2[ M+H ]] +
Step 4: synthesis of (R) -2- (4- ((1-ethylpiperidin-3-yl) amino) pyrido [3,4-d ] pyridazin-1-yl) -5- (trifluoromethyl) phenol
To (R) -2- (4- (piperidin-3-ylamino) pyrido [3, 4-d)]To a solution of pyridazin-1-yl) -5- (trifluoromethyl) phenol (N218122-038-A1) (200 mg,389 mmol) in 1, 2-dichloroethane (2 mL) was added acetaldehyde (33.9 mg,0.770 mmol),the resulting mixture was stirred at room temperature for 2 minutes and NaBH (OAc) was added 3 (217 mg,1.03 mmol) and the resulting reaction solution was stirred at room temperature under nitrogen for 1 hour. Water (20.0 mL) was added to the reaction mixture, the mixture was extracted with DCM (20.0 mL. Times.2), and the organic phases were combined, washed with saturated brine and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and isolation of the residue by prep-HPLC (Shimadzu LC-20AP; YMC-acts Triart C18X 20.0mm,5um; mobile phase A: water/0.1% FA, mobile phase B: ACN; gradient: 2min 20% B, 20% B to 50% B, holding 2min 50% B, then 1min 5% B, detection wavelength 220nm,254 nm) gives the title compound (52.0 mg, two step yield 19.7%, white solid). LC-MS (ESI) m/z 418.2[ M+H ] ] +1 H NMR(400MHz,DMSO)δ10.75–10.45(m,1H),9.87–9.73(m,1H),9.62–9.37(m,1H),8.90–8.81(m,1H),8.33–8.06(m,1H),7.53–7.43(m,1H),7.35–7.25(m,1H),7.28–7.21(m,1H),3.83–3.70(m,1H),3.52–3.42(m,1H),3.42–3.22(m,1H),3.20–3.07(m,2H),2.94–2.81(m,1H),2.79–2.67(m,1H),2.14–2.06(m,1H),2.04–1.94(m,1H),1.83–1.60(m,2H),1.27–1.07(m,3H)。
Example 9:2- (4- (quinin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of N- (4-chlorophthalazin-1-yl) quinine-3-amine
To a solution of 1, 4-dichlorophthalazine (300 mg,1.51 mmol) in EtOH (3.00 mL) was added quinine-3-amine (190 mg,1.51 mmol), na 2 CO 3 (240 mg,2.26 mmol) and the resulting reaction mixture was stirred overnight at 80℃under nitrogen. The reaction mixture was cooled to room temperature, water (30.0 mL) was added, extracted with EA (30.0 mL. Times.2), the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (DCM/meoh=10/1) gave the title compound (70.0 mg, yield 16.1%, white solid). LC-MS (ESI) m/z 289.2[ M+H ]] +
Step 2: synthesis of 2- (4- (quinin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a three-necked flask containing N- (4-chlorophthalazin-1-yl) quinine-3-amine (N218122-057-A1) (70.0 mg,0.242 mmol) was added dioxane (2.00 mL) and water (0.400 mL), followed by (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (59.9 mg,0.29 mmol), DIPEA (62.7 mg,1.872 mmol) and Pd (dtbpf) Cl 2 (15.8 mg,0.0242 mmol) and the resulting reaction mixture was stirred overnight at 90℃under nitrogen. The reaction solution was cooled to room temperature, poured into water (5.00 mL), extracted with EA (5.00 mL. Times.2), and the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and isolation of the residue by prep-HPLC (Shimadzu LC-20AP; YMC-acts Triart C18X 20.0mm,5um; mobile phase A: water/0.1% FA, mobile phase B: ACN; gradient: 2min 20% B, 20% B to 50% B, holding 2min50% B, then 1min 5% B, detection wavelength 220nm,254 nm) gave the title compound (6.4 mg, yield 6.37% white solid). LC-MS (ESI) m/z 415.3[ M+H ]] +1 H NMR(400MHz,DMSO)δ8.52(d,J=8.3Hz,1H),8.26(s,1H),7.90(t,J=7.6Hz,1H),7.80(t,J=7.6Hz,1H),7.51(d,J=8.0Hz,1H),7.46(d,J=8.1Hz,1H),7.34–7.24(m,3H),4.49–4.35(m,1H),3.14–3.07(m,1H),3.05–2.99(m,1H),2.97–2.87(m,3H),2.47–2.43(m,1H),2.37–2.22(m,1H),2.10–1.92(m,1H),1.85–1.69(m,2H),1.61–1.41(m,1H)。
Example 10:compound 2- (4- ((2-hydroxycyclohexyl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of 2- ((4-bromophthalazin-1-yl) amino) cyclohexanol
A solution of 1, 4-dibromophthalazine (100 mg, 277 mmol) and 2-aminocyclohexanol (40.0 mg, 277 mmol) in EtOH (3.00 mL) was stirred at 100deg.C for 24 hours. The reaction solution was cooled to room temperature, and a solid was precipitated, filtered, and the cake was dried under reduced pressure to give the objective compound (50.0 mg, yield 44.7%, white solid) which was directly used as followsOne step. LC-MS (ESI) m/z 322.1324.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.64(br d,J=6.97Hz,1H),8.27-8.26(m,2H),8.06-8.16(m,2H),7.73-7.84(m,1H),3.54-3.66(m,1H),3.23-3.39(m,1H),1.82-2.11(m,2H),1.58-1.78(m,2H),1.15-1.44(m,4H)。
Step 2: synthesis of 2- (4- ((2-hydroxycyclohexyl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a mixed solution of 2- (4- ((2-hydroxycyclohexyl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol (50.0 mg,155 mmol) and (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (63.9 mg,310 mmol) in dioxane (2.00 mL) and water (0.40 mL) under nitrogen protection at 25℃was added Pd (dtbpf) Cl in sequence 2 (20.2 mg,31.0 mmol) and K 2 CO 3 (42.9 mg,310 mmol) and the resulting reaction mixture was stirred under nitrogen at 90℃for 12 hours. The reaction mixture was cooled to room temperature, filtered through celite, the filtrate was concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge Prep OBD C18150 x 40mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 25% -55%;8 min) to give the title compound (8.65 mg, yield 13.1%, light grey solid). LC-MS (ESI) m/z 404.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.41(br d,J=7.88Hz,1H),7.83-7.90(m,1H),7.74-7.81(m,1H),7.51(br d,J=7.50Hz,1H),7.44(br d,J=7.50Hz,1H),7.23-7.36(m,2H),7.13(br d,J=5.00Hz,1H),4.83(br d,J=1.13Hz,0.5H),4.55(br s,0.5H),4.39-4.39(m,0.5H),4.27(s,1.5H),4.12-4.18(m,0.5H),3.58-3.64(m,0.5H),2.14-2.18(m,0.5H),1.96-2.01(m,0.5H),1.69(br dd,J=3.00,2.13Hz,21H),1.23–1.31(m,5H)。
Example 11:(R) 2- (8- ((1-ethylpiperidin-3-yl) amino) pyrido [2,3- ]d]Pyridazin-5-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (R) -3- ((5-chloropyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
5, 8-dichloropyrido [2,3-d ]]A solution of pyridazine (3.00 g,15.0 mmol) and tert-butyl (R) -3-aminopiperidine-1-carboxylate (3.15 g,15.7 mmol) in EtOH (30.0 mL) was stirred at 70℃for 12 h. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by Prep-TLC (PE/ea=1/1) to give the objective compound (1.20 g, yield 13.2%, yellow solid). LC-MS (ESI) m/z 364.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 9.15(br d,J=3.51Hz,1H),8.43(br d,J=7.89Hz,1H),7.94-8.07(m,1H),7.26-7.52(m,1H),4.06-4.19(m,1H),3.45-4.00(m,2H),2.92-3.15(m,1H),1.64-1.97(m,3H),1.39-1.54(m,2H),1.06-1.39(m,9H)。
Step 2: synthesis of tert-butyl (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylate
(R) -3- ((5-chloropyrido [2, 3-d)]Pyridazin-8-yl) amino piperidine-1-carboxylic acid tert-butyl ester (960 mg,2.64 mmol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (815 mg,3.96 mmol), K 2 CO 3 (729 mg,5.28 mmol) and Pd (dtbpf) Cl 2 (344 mg,528 umol) was added to a mixed solution of dioxane (10.0 mL) and water (2.00 mL), and the resulting reaction mixture was stirred under nitrogen at 90℃for 12 hours. The reaction mixture was cooled to room temperature, diluted with water (5.00 mL), extracted with EA (5.00 mL. Times.3), the organic phases were combined, washed with saturated brine (10.0 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification of the residue by Prep-TLC (PE/ea=1/1) gave the title compound (350 mg, yield 27.1%, yellow solid). LC-MS (ESI) m/z 490.3[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.44-10.52(m,1H),9.11(dd,J=4.25,1.50Hz,1H),7.92-7.97(m,1H),7.86(dd,J=8.38,4.38Hz,1H),7.54-7.62(m,1H),7.27-7.38(m,3H),4.55(qd,J=7.02,3.69Hz,1H),4.28(br dd,J=7.57,3.81Hz,1H),4.03(q,J=7.09Hz,2H),1.75(br dd,J=9.76,3.50Hz,1H),1.43(br s,13H)。
Step 3: synthesis of (R) -2- (8- (piperidin-3-ylamino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
At 25 ℃, to (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2, 3-d)]To a solution of t-butyl pyridazin-8-yl) amino-piperidine-1-carboxylate (300 mg, 313 mmol) in DCM (10.0 mL) was slowly added dropwise a solution of HCl-dioxane (4M, 2.00 mL) and the resulting reaction was stirred at 25℃for 12 h. The reaction solution was concentrated under reduced pressure to give the objective compound (250 mg, crude product, yellow solid) which was directly used for the next reaction. LC-MS (ESI) m/z 390.1[ M+H ] ] +
Step 4: synthesis of (R) -2- (8- ((1-ethylpiperidin-3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
At 0 ℃, to (R) -2- (8- (piperidin-3-ylamino) pyrido [2, 3-d)]To a solution of pyridazin-5-yl) -5- (trifluoromethyl) phenol (150 mg,385 mmol) in EtOH (5.00 mL) was added NaBH successively 3 CN (72.6 mg,1.16 mmol) and glacial AcOH (231 mg,3.85 mmol), the resulting mixture was stirred at 0deg.C for 5 min, then acetaldehyde (5M, 385 mL) was added dropwise at 0deg.C, and the resulting reaction was stirred at 0deg.C for 1 hr. The reaction mixture was quenched with water (5.00 mL), extracted with EA (5.00 mL. Times.3), the organic phases were combined, washed with saturated brine (5.00 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column Waters Xbridge BEH C18 100X 30mm X10 um; mobile phase [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -60%;10 min) to give the title compound (24.6 mg, yield 15.3%, white solid). LC-MS (ESI) m/z 418.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm10.56(br s,1H),9.07-9.15(m,1H),7.79-7.98(m,2H),7.58(d,J=7.8Hz,1H),7.24-7.40(m,3H),4.43(br s,1H),2.80(br d,J=8.6Hz,1H),2.28-2.45(m,5H),1.66-1.86(m,3H),1.57(br d,J=4.5Hz,1H),1.04(t,J=7.1Hz,3H)。
Example 12:(R) -4- (2- (difluoromethyl) -4-methylphenyl) schemeN- (1-ethylpiperidin-3-yl) phthalazin-1-amine
Step 1: synthesis of 1-bromo-2- (difluoromethyl) -4-methylbenzene
At 25 ℃, diethylamino is trifluorideSulfur (10.8 g,60.3 mmol) is added dropwise to a solution of 2-bromo-5-methylbenzaldehyde (6.00 g,30.2 mmol) in DCM (48.0 mL) and the resulting reaction stirred at 25℃for 3 h. Saturated Na was used as the reaction solution 2 CO 3 Aqueous (50.0 mL) dilution, DCM (50.0 mL. Times.3) extraction, combining the organic phases, and drying with anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification of the residue by silica gel chromatography (pe=100%) gave the title compound (4.70 g, yield 70.5%, transparent oil). 1 H NMR(400MHz,DMSO-d 6 )δppm 7.62(d,J=8.11Hz,1H),7.49(s,1H),7.31(br d,J=8.11Hz,1H),7.09(t,J=54.4Hz,1H),2.33(s,3H)。
Step 2: synthesis of 2- (2- (difluoromethyl) -4-methylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan
1-bromo-2- (difluoromethyl) -4-methylbenzene (4.60 g,20.8 mmol), bis-pinacolato borate (10.6 g,41.6 mmol), potassium acetate (6.13 g,62.4 mmol) and PdCl were reacted at 25℃ 2 (dppf) (3.05 mg,4.16 mmol) was added to dioxane (230 mL) and the resulting reaction mixture was stirred under nitrogen at 100deg.C for 12 hr. The reaction mixture was cooled to room temperature, diluted with EA (300 mL), filtered, and the filtrate concentrated under reduced pressure. The residue was purified by silica gel chromatography (PE/ea=10/1 to 5/1 to 3/1) to give the objective compound (2.10 g, yield 37.6%, transparent oil). 1 H NMR(400MHz,DMSO-d 6 )δppm7.68(d,J=7.65Hz,1H),7.49(s,1H),7.15-7.46(m,2H),2.38(s,3H),1.30(s,12H)。
Step 3: synthesis of (R) -3- ((4- (2- (difluoromethyl) -4-methylphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
(R) -3- ((4-bromophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (600 mg,1.47 mmol), 2- (2- (difluoromethyl) -4-methylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (79mg, 2.95 mmol), K at 25 ℃C 2 CO 3 (407 mg,2.95 mmol) and Pd (dtbpf) Cl 2 (192 mg, 295. Mu. Mol) was added to a mixed solution of dioxane (10.0 mL)/water (2.00 mL). The resulting reaction mixture was stirred under nitrogen at 90℃for 12 hours. The reaction mixture was cooled to room temperature, diluted with saturated brine (10.0 mL), extracted with DCM (10.0 mL. Times.3), and the organic phases combined with anhydrous Na 2 SO 4 Drying, filtration, concentration by distillation under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=2/1 to 1/1) gave the objective compound (680 mg, yield 98.5%, black oil). LC-MS (ESI) m/z 469.2[ M+H ]] +
Step 4: synthesis of (R) -4- (2- (difluoromethyl) -4-methylphenyl) -N- (piperidin-3-yl) phthalazin-1-amine
To a solution of tert-butyl (R) -3- ((4- (2- (difluoromethyl) -4-methylphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate (680 mg,1.45 mmol) in DCM (10.0 mL) at 25℃was added dropwise a solution of HCl-dioxane (2.00 mL). The resulting reaction mixture was stirred at 25℃for 12 hours. The reaction mixture was diluted with saturated aqueous NaCl (10.0 mL), extracted with DCM (10.0 mL. Times.3), and the organic phases combined with anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (730 mg, trude, yellow solid) which was used directly in the next reaction. LC-MS (ESI) m/z 369.1[ M+H ] ] +
Step 5: synthesis of (R) -4- (2- (difluoromethyl) -4-methylphenyl) -N- (1-ethylpiperidin-3-yl) phthalazin-1-amine
To a solution of (R) -4- (2- (difluoromethyl) -4-methylphenyl) -N- (piperidin-3-yl) phthalazin-1-amine (660 mg,1.79 mmol) in EtOH (10.0 mL) at 0deg.C was added NaBH successively 3 CN (338 mg,5.37 mmol) and AcOH (1.08 mg, 17.9. Mu. Mol, 1.02. Mu.L), after stirring for 5 min, acetaldehyde (5M, 1.79 mL) was added dropwise. The resulting reaction mixture was stirred at 0℃for 1 hour. The reaction mixture was diluted with saturated aqueous NaCl (10.0 mL), extracted with DCM (10.0 mL. Times.3), and the organic phases combined with anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column: phenomenex C1880. Times.40 mm. Times.3 um; mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -60%;8 min) to give the title compound (20.0 mg, yield 2.86%, pale yellow solid). LC-MS (ESI) m/z 397.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.43(d,J=8.6Hz,1H),7.88(t,J=7.6Hz,1H),7.76-7.82(m,1H),7.63(s,1H),7.49(d,J=7.7Hz,1H),7.33-7.40(m,2H),7.18(d,J=7.7Hz,1H),6.63-6.93(m,1H),4.41(br s,1H),3.16(br d,J=8.3Hz,1H),2.82(br d,J=11.6Hz,1H),2.48(br s,3H),2.38(q,J=6.8Hz,2H),2.01(br d,J=7.2Hz,1H),1.85-1.97(m,2H),1.75(br d,J=12.9Hz,1H),1.42-1.65(m,2H),1.01(t,J=7.1Hz,3H)。
Example 13:(R) 2- (1- ((1-ethylpiperidin-3-yl) amino) pyrido [3,4- ]d]Pyridazin-4-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (R) -2- (1- (piperidin-3-ylamino) pyrido [3,4-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
At room temperature, to (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [3, 4-d)]To a solution of t-butyl pyridazin-1-yl) amino-piperidine-1-carboxylate (69.0 mg,0.140 mmol) in DCM (1.00 mL) was added dropwise a solution of HCl-dioxane (1.00 mL) and the resulting reaction was stirred at room temperature under nitrogen for 1 hour. The reaction solution was concentrated under reduced pressure to remove the solvent to give the objective compound (51.0 mg, crude product, yellow solid) which was directly used for the next reaction. LC-MS (ESI) m/z 390.2[ M+H ] ] +
Step 2: synthesis of (R) -2- (1- ((1-ethylpiperidin-3-yl) amino) pyrido [3,4-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
To (R) -2- (1- (piperidin-3-ylamino) pyrido [3, 4-d)]To a solution of pyridazin-4-yl) -5- (trifluoromethyl) phenol (51.0 mg,0.122 mmol) in 1, 2-dichloroethane (1 mL) was added acetaldehyde (33.9 mg,0.130 mmol), and the resulting mixture was stirred at room temperature for 2min and then NaBH (OAc) was added 3 (84.0 mg,0.260 mmol) and the resulting reaction mixture was stirred at room temperature under nitrogen for 1 hour. Water (20.0 mL) was added to the reaction mixture, extracted with DCM (20.0 mL. Times.3), and the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and isolation and purification of the residue by prep-HPLC (Shimadzu LC-20AP; YMC-acts Triart C18X 20.0mm,5um; mobile phase A: water/0.1% FA, mobile phase B: ACN; gradient: 2min 20% B, 20% B to 50% B, holding 2min 50% B, then 1min 5% B, detection wavelength 220nm,254 nm) to give the title compound (1.7 mg, two-step yield 3.1)% white solid). LC-MS (ESI) m/z 418.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 9.94–9.66(m,1H),9.56–9.21(m,1H),9.14–8.85(m,1H),8.46–8.12(m,1H),8.02–7.80(m,1H),7.70–7.49(m,1H),7.31–7.01(m,1H),6.80–6.42(m,1H),4.31–4.17(m,1H),3.93–3.76(m,1H),3.74–3.60(m,1H),3.58–3.48(m,1H),3.11–2.99(m,1H),2.95–2.78(m,2H),2.69–2.62(m,1H),2.08–1.86(m,1H),1.78–1.56(m,1H),1.48–1.26(m,1H),1.21–1.13(m,3H)。
Example 16:(R) -5- (dibromomethyl) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) phenol
Step 1: synthesis of 1-bromo-4- (difluoromethyl) -2-methoxybenzene
DAST (8.33 g,46.50 mmol) was added to a solution of 4-bromo-3-methoxybenzaldehyde (5.00 g,23.2 mmol) in DCM (40.0 mL) at 25℃and the resulting reaction mixture was stirred at 25℃for 3 h. The reaction mixture was taken up in NaHCO 3 Aqueous (50.0 mL) was diluted, extracted with anhydrous DCM (50.0 mL. Times.3), the organic phases combined, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (pe=100%) gave the title compound (4.30 g, yield 78.0%, transparent oil).
Step 2: synthesis of 2- (4- (difluoromethyl) -2-methoxyphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan
1-bromo-4- (difluoromethyl) -2-methoxybenzene (1.00 g,4.22 mmol), bis-pinacolatoborate (5.36 g,21.1 mmol), KOAc (1.24 g,12.7 mmol) and PdCl were reacted at 25℃ 2 (dppf). DCM (689 mg, 844. Mu. Mol) was added to a mixture of dioxane (9.00 mL) and DMSO (1.00 mL), and the resulting reaction mixture was stirred at 80℃under nitrogen for 12 hours. The reaction mixture was cooled to room temperature, then aqueous NaCl (10.0 mL) was added, extracted with EA (10.0 mL. Times.3), and the organic phases were combined with anhydrous Na 2 SO 4 Dried, filtered, concentrated under reduced pressure and the residue was purified by silica gel chromatography (PE/ea=5/1About 3/1 to 1/1) to obtain the target compound (920 mg, yield 76.7%, yellow solid). 1 H NMR(400MHz,DMSO-d 6 )δppm 7.64(d,J=7.75Hz,1H),7.12(s,2H),7.11-6.87(m,1H),3.79(s,3H),1.28-1.27(m,3H),1.28(s,12H)。
Step 3: synthesis of (R) -3- ((4- (4- (difluoromethyl) -2-methoxyphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
(R) -3- ((4-bromophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (400 mg, 982. Mu. Mol), 2- (4- (difluoromethyl) -2-methoxyphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (503 mg,1.77 mmol), K at 25 ℃C 2 CO 3 (272 mg,1.96 mmol) and Pd (dtbpf) Cl 2 (128 mg, 197. Mu. Mol) was added to a mixture of dioxane (10.0 mL) and water (2.00 mL), and the resulting reaction mixture was stirred at 90℃for 12 hours under nitrogen. The reaction mixture was cooled to room temperature, then aqueous NaCl (10.0 mL) was added, and the mixture was taken up with EA (10.0 mL. Times.3), and the organic phases were combined with anhydrous Na 2 SO 4 Dried, filtered, concentrated under reduced pressure, and the residue was purified by silica gel chromatography (ea=100%) to give the target compound (260 mg, yield 54.6%, pale yellow solid). LC-MS (ESI) m/z 485.3[ M+H ]] +
Step 4: synthesis of (R) -4- (4- (difluoromethyl) -2-methoxyphenyl) -N- (piperidin-3-yl) phthalazin-1-amine
A solution of HCl-dioxane (2.00 mL) was added dropwise to (R) -3- ((4- (4- (difluoromethyl) -2-methoxyphenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (210 mg, 433.40. Mu. Mol) in DCM (10.0 mL) at 0deg.C, and the resulting reaction mixture was stirred at 25deg.C for 12 hours. The reaction mixture was diluted with anhydrous EtOH (10.0 mL), filtered, and the filtrate concentrated under reduced pressure to give the title compound (200 mg, eude, yellow solid). LC-MS (ESI) m/z 385.1[ M+H ] ] +
Step 5: synthesis of (R) -4- (4- (difluoromethyl) -2-methoxyphenyl) -N- (1-ethylpiperidin-3-yl) phthalazin-1-amine
At 0 ℃, naBH is sequentially added 3 CN (73.6 mg,1.17 mmol) and AcOH (235. Mu.g, 3.90. Mu. Mol) were added to EtOH (5.00 mL) of (R) -4- (4- (difluoromethyl) -2-methoxyphenyl) -N- (piperidin-3-yl) phthalazin-1-amine (150 mg, 390. Mu. Mol), stirredAfter stirring for 5 minutes, acetaldehyde (86.0 mg,1.95 mmol) was added dropwise and the resulting reaction mixture was stirred at 0deg.C for 1 hour. The reaction mixture was taken up in aqueous NaCl (10.0 mL), extracted with DCM (10.0 mL. Times.3), and the organic phases combined with anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (150 mg, yield 93.2%, yellow solid). LC-MS (ESI) m/z 413.2[ M+H ]] +
Step 6: synthesis of (R) -5- (dibromomethyl) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) phenol
BBr was subjected to nitrogen protection at-60 ℃ 3 (1M, 969.76. Mu.L) was added dropwise to (R) -4- (4- (difluoromethyl) -2-methoxyphenyl) -N- (1-ethylpiperidin-3-yl) phthalazin-1-amine (80.0 mg, 194. Mu. Mol) in DCM (10.0 mL) and the resulting reaction mixture stirred at 25℃for 12 hours. The reaction mixture was concentrated directly under reduced pressure and the residue was purified by Prep-HPLC (column Phenomenex Luna C18 75 x 30mm x 3um; mobile phase: [ a-water (FA)/B-ACN) ]The method comprises the steps of carrying out a first treatment on the surface of the B% = 1% -35%;8 min) to give the title compound (17.5 mg,17.4% as a yellow solid). LC-MS (ESI) m/z 521.0[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.38(br d,J=7.75Hz,1H),8.13(s,1H),7.90(m,1H),7.83-7.77(m,1H),7.54-7.44(m,3H),7.32(d,J=7.88Hz,1H),7.29-7.27(m,1H),7.18(dd,J=7.88,1.75Hz,1H),4.43(m,1H),3.24(br s,1H),3.01-3.12(m,1H),2.47-2.41(m,2H),2.13-1.98(m,2H),1.92-1.83(m,1H),1.79-1.53(m,3H),1.11(br s,3H)。
Example 17:(R) -2- (4- ((1-methylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (R) -2- (4- ((1-methylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol (60.0 mg,0.155 mmol) in 1, 2-dichloroethane (2.00 mL) was added paraformaldehyde (9.00 mg,0.309 mmol) and the resulting mixture stirred at room temperature for 2 min and NaBH (OAc) was added 3 (81.5 mg, 0.383 mmol) and the resulting reaction mixture was stirred at room temperature under nitrogen for 1 hour. To the reaction mixture was added water (5.00 mL), extracted with DCM (5.00 mL. Times.3), and the organic phases were combined, washed with saturated aqueous NaCl solution (20.0 mL), and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column: YMC, C18 (250X 20mm,5 μm); mobile phase: [ water (0.1% NH) 4 HCO 3 )-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B%:20% -50% ACN,20 min) to give the title compound (15.7 mg, yield 25.3%, white solid). LC-MS (ESI) m/z 403.3[ M+H ]] +1 H-NMR(400MHz,DMSO-d 6 )δppm 8.40(d,J=8.2Hz,1H),7.86(t,J=7.7Hz,1H),7.78(t,J=7.6Hz,1H),7.52(d,J=7.8Hz,1H),7.43(d,J=8.1Hz,1H),7.31(d,J=8.4Hz,1H),7.27(s,1H),7.16(d,J=6.9Hz,1H),4.50–4.37(m,1H),3.20–3.14(m,1H),3.13–3.03(m,1H),2.78–2.68(m,1H),2.23(s,3H),1.98(d,J=11.6Hz,3H),1.83–1.71(m,1H),1.68–1.54(m,1H),1.55–1.38(m,1H)。
Example 18:(R) -5- (difluoromethyl) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) phenol
Step 1: synthesis of (R) -5- (difluoromethyl) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) phenol
At 0 ℃, naBH is added 3 CN (35.6 mg, 566. Mu. Mol) and a drop of ice AcOH were added to a solution of (R) -5- (difluoromethyl) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenol (70.0 mg, 188. Mu. Mol) in dry EtOH (1.00 mL), and then a solution of acetaldehyde THF (5M, 188. Mu.L) was added dropwise to the above mixture, and the resulting reaction mixture was stirred at 0deg.C for 1 hour. The reaction mixture was filtered and the filtrate was purified by Pre-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 15% -45%;10 min) to give the title compound (R) -5- (difluoromethyl) -2- (4- ((1-ethylpiperidin-3-yl) amino) phthalazin-1-yl) phenol (10.1 mg, yield 13.4% as a yellow solid). LC-MS (ESI) m/z 399.3[ M+H ]] +1 H NMR(400MHz,DMSO-d6)δppm 10.07(br s,1H),8.38(br d,J=8.13Hz,1H),7.82-7.90(m,1H),7.74-7.81(m,1H),7.37-7.48(m,2H),6.93-7.20(m,4H),4.43(br s,1H),3.20(br s,1H),2.83(br s,1H),2.40-2.50(m,2H),1.89-2.13(m,2H),1.40-1.78(m,2H),1.35-1.60(m,2H),0.93-1.10(m,3H)。
Example 19:(R) -5- (difluoromethyl) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenol
Step 1: synthesis of 1-bromo-4- (difluoromethyl) -2- (methoxymethoxy) benzene
Sodium hydrogen (358 mg,8.97mmol,60.0% purity) was added in portions to a solution of 2-bromo-5- (difluoromethyl) phenol (1.00 g,4.48 mmol) in THF (10.0 mL) at 0 ℃, the resulting reaction mixture was stirred at 0 ℃ for half an hour, chloromethyl methyl ether (470 mg,5.84 mmol) was then added dropwise at 0 ℃ and the resulting reaction mixture was stirred for 12 hours after returning to 25 ℃. To the reaction solution, 10.0mL of saturated aqueous ammonium chloride solution was slowly added dropwise, extracted with EA (10.0 mL. Times.3), and the organic phases were combined, followed by anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (1.10 g, yield 91.8%, colorless oil) which was used directly in the next reaction.
Step 2: synthesis of 2- (4- (difluoromethyl) -2- (methoxymethoxy) phenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan
Under the protection of nitrogen, pdCl is added 2 (dppf) (547 mg, 748. Mu. Mol) was added to a solution of 1-bromo-4- (difluoromethyl) -2- (methoxymethoxy) benzene (1.00 g,3.74 mmol), potassium acetate (1.84 g,18.7 mmol) and duplex pinacol borate (1.90 g,7.49 mmol) in dioxane (5.00 mL). The reaction mixture was stirred under nitrogen at 100℃for 12 hours. The reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=10/1 to 1/1) to give the objective compound (500 mg, yield 42.5%, yellow solid). 1 H NMR(400MHz,CHCl 3 -d)δppm 7.75(d,J=7.45Hz,1H),7.09-7.19(m,2H),6.39-6.79(m,1H),5.22(s,2H),3.52(s,3H),1.36(s,12H)。
Step 3: synthesis of tert-butyl (R) -3- ((4- (4- (difluoromethyl) -2- (methoxymethoxy) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate
Pd (dtbpf) Cl under nitrogen protection 2 (103 mg, 159. Mu. Mol) was added to tert-butyl 2- (4- (difluoromethyl) -2- (methoxymethoxy) phenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (250 mg, 795. Mu. Mol), (R) -3- ((4-bromophthalazin-1-yl) amino) piperidine-1-carboxylate (324 mg, 795. Mu. Mol) and anhydrous K 2 CO 3 (219 mg,1.59 mmol) of dioxane (3.00 mL)/water (0.30 mL) was added and the mixture was stirred at 90℃for 12 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=10/1 to 1/1) to give the objective compound (180 mg, yield 43.9%, white solid). LC-MS (ESI) m/z 515.3[ M+H ]] +
Step 4: synthesis of (R) -5- (difluoromethyl) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenol
A solution of HCl-dioxane (4M, 100. Mu.L) was added dropwise to a solution of (R) -3- ((4- (4- (difluoromethyl) -2- (methoxymethoxy) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (180 mg, 349. Mu. Mol) in DCM (1.00 mL) at 0deg.C, and the resulting reaction was stirred at 25deg.C for 2 hours. The reaction mixture was concentrated under reduced pressure and the crude product obtained was purified by Pre-HPLC (column: phenomenex Luna 80X 30mm 3um; mobile phase: [ A-water (HCl)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 10% -45%;8 min) to give the title compound (R) -5- (difluoromethyl) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) phenol (140 mg, yield 98.3%, white solid). LC-MS (ESI) m/z 371.1[ M+H ]] +1 H NMR(400MHz,MeOH-d 4 )δppm 8.79(br d,J=8.34Hz,1H),8.28(br t,J=7.51Hz,1H),8.13(t,J=7.45Hz,1H),7.96(br d,J=7.99Hz,1H),7.61(br d,J=7.63Hz,1H),7.25-7.35(m,2H),6.87(t,J=38Hz,1H),4.57-4.71(m,1H),3.74(br dd,J=12.40,3.58Hz,1H),3.42(br d,J=12.64Hz,1H),3.25(br t,J=10.91Hz,1H),3.07-3.18(m,1H),2.26-2.39(m,1H),2.19(br dd,J=10.67,3.76Hz,1H),1.90-2.09(m,2H)。
Example 20:compounds [ (]R) -2- (4- ((1-methylpiperidin-3-yl) oxy) phthalazin-1-yl) -5- (trifluormethyl) Radical) phenol
Step 1: synthesis of tert-butyl (R) -3- ((4-chlorophthalazin-1-yl) oxy) piperidine-1-carboxylate
Anhydrous K at 25deg.C 2 CO 3 (5.21 g,37.7 mmol) was added to a stirred solution of 1, 4-dichlorophthalazine (5.00 g,25.1 mmol) and tert-butyl (3R) -3-hydroxypiperidine-1-carboxylate (6.07 g,30.2 mmol) in DMF (100 mL). The resulting reaction mixture was stirred at 100℃for 12 hours. The reaction mixture was cooled to room temperature, diluted with EA (100 mL), washed successively with water (30.0 mL. Times.3), saturated brine (30.0 mL), and the organic phase was taken up in anhydrous Na 2 SO 4 Drying, filtration, concentration of the filtrate under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=5/1 to 2/1 to 1/1) gave the objective compound (3.00 g, yield 32.8%, pale yellow solid). LC-MS (ESI) m/z 364.2,366.1[ M+H ]] +1 H NMR(400MHz,CHCl 3 -d)δppm8.16(m,2H),7.78-7.97(m,2H),5.49(br s,1H),4.17-4.31(m,1H),3.83-3.96(m,1H),3.30-3.44(m,1H),3.02-3.14(m,1H),1.99-2.09(m,2H),1.60(br s,1H),1.42(br,s,1H),1.04(br s,9H)。
Step 2: synthesis of tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) oxy) piperidine-1-carboxylate
To a mixed solution of tert-butyl (R) -3- ((4-chlorophthalazin-1-yl) oxy) piperidine-1-carboxylate (1.00 g,2.75 mmol) of dioxane (20.0 mL)/water (2.00 mL) at 25℃was added successively (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (679 mg,3.30 mmol), pdCl 2 (dppf) (201 mg, 275. Mu. Mol) and anhydrous K 3 PO 4 (1.75 g,8.25 mmol). After the addition was complete, the reaction mixture was stirred at 100℃for 12 hours under nitrogen. The reaction mixture was cooled to room temperature, filtered, concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=10/1 to 5/1 to 2/1) to give the objective compound (500 mg, yield 34.0%, pale yellow solid). LC-MS (ESI) m/z 490.2[ M+H ] ] +1 H NMR(400MHz,DMSO-d6)δppm 10.46(br s,1H),8.21(br d,J=7.88Hz,1H),7.95-8.01(m,1H),7.90-7.95(m,1H),7.59(br d,J=8.00Hz,1H),7.48(br s,1H),7.35(br d,J=8.13Hz,1H),7.32(s,1H),5.45(br s,1H),4.34-4.50(m,1H),3.91-3.98(m,1H),3.28-3.37(m,1H),2.86-3.09(m,1H),2.02(br s,2H),1.60(br s,1H),1.40-1.45(m,1H),0.85(br s,9H)。
Step 3: synthesis of (R) -2- (4- (piperidin-3-yloxy) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) oxy) piperidine-1-carboxylate (200 mg, 409. Mu. Mol) in dioxane (2.00 mL) at 25℃was added dropwise a solution of HCl-dioxane (4M, 2.00 mL), and the resulting reaction mixture was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure at 25℃to give the title compound (173 mg, yield 99.4%, white solid). LC-MS (ESI) m/z 390.3[ M+H ]] +
Step 4: synthesis of (R) -2- (4- ((1-methylpiperidin-3-yl) oxy) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of (R) -2- (4- (piperidin-3-yloxy) phthalazin-1-yl) -5- (trifluoromethyl) phenol (80.0 mg, 205. Mu. Mol) in MeOH (2.00 mL) at 25℃was added paraformaldehyde (30.9 mg,1.03 mmol) and glacial AcOH (1.23 mg, 20.6. Mu. Mol) in sequence. Stirring at room temperature for 30 min, and adding NaBH 3 CN (38.7 mg, 616. Mu. Mol). The reaction mixture was stirred at room temperature for 12 hours. The reaction solution was filtered, the filtrate was concentrated under reduced pressure at 25℃and the residue was purified by Prep-HPLC (column: phenomenex luna C, 100X 40mm X3 um; mobile phase: [ A-water (FA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 15% -45%;8 min) to give the title compound (1.83 mg, yield 2.15%, white solid). LC-MS (ESI) m/z 404.1[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.44(s,1H),8.33(br s,1H),7.98-8.04(m,1H),7.91-7.97(m,1H),7.56(t,J=8.05Hz,2H),7.29-7.38(m,2H),5.64(br,s,1H),3.24-3.30(m,2H),2.53(br s,3H),2.45-2.49(m,2H),1.92-2.07(m,2H),1.64-1.84(m,2H)。
Example 22:(R) -2- (4- ((1-methylpiperidin-3-yl) amino) -5,6,7, 8-tetrahydrophthalazin-1-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (R) -3- ((4-chloro-5, 6,7, 8-tetrahydrophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
CsF (747 mg,4.92 mmol) and tert-butyl (R) -3-aminopiperidine-1-carboxylate (591 mg,2.95 mmol) were added to 1, 4-dichloro-5, 6,7, 8-tetrahydrophthalazine (500 mg,2.46 mmol) in DMSO (1.00 mL) at 0deg.C and the resulting reaction mixture stirred at 100deg.C for 2 hours. The reaction mixture was cooled to room temperature, diluted with water (2.00 mL), extracted with EA (2.00 mL. Times.3), the organic phases combined, washed successively with saturated NaCl solution, anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=10/1 to 0/1) gave the objective compound (500 mg, yield 55.4%, yellow solid). LC-MS (ESI) m/z 367.3[ M+H ]] +
Step 2: synthesis of (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) -5,6,7, 8-tetrahydrophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
Tert-butyl (R) -3- ((4-chloro-5, 6,7, 8-tetrahydrophthalazin-1-yl) amino) piperidine-1-carboxylate (150 mg, 409. Mu. Mol) and (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (101 mg, 491. Mu. Mol) are added to a mixed solution of dioxane (2.00 mL) in water (0.20 mL) at 25℃and PdCl is then added under nitrogen protection 2 (dppf) (29.9 mg, 40.9. Mu. Mol) and anhydrous K 3 PO 4 (260 mg,1.23 mmol) and the resulting reaction mixture was stirred at 110℃for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (2.00 mL), extracted with EA (2.00 mL. Times.3), and the organic phases were combined, washed with saturated brine, and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=10/1-0/1) gave the objective compound (100 mg, yield 49.7%, brown solid). LC-MS (ESI) m/z 493.4[ M+H ]] +
Step 3: synthesis of (R) -2- (4- (piperidin-3-ylamino) -5,6,7, 8-tetrahydrophthalazin-1-yl) -5- (trifluoromethyl) phenol
A solution of HCl-dioxane (4M, 2 mL) was added dropwise to (R) -3- ((4-chloro-5, 6,7, 8-tetrahydrophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (100 mg, 203. Mu. Mol) of dioxane (2.00 mL) at 0deg.C, and the resulting reaction mixture was purified in a solventStirred at 25℃for 12 hours. The reaction mixture was concentrated directly under reduced pressure to give the objective compound (70.0 mg, yield 87.9%, pale yellow solid) which was used directly in the next reaction. LC-MS (ESI) m/z 393.3[ M+H ]] +
Step 4: synthesis of (R) -2- (4- ((1-methylpiperidin-3-yl) amino) -5,6,7, 8-tetrahydrophthalazin-1-yl) -5- (trifluoromethyl) phenol
At 0 ℃, naBH is added 3 CN (48.0 mg, 764. Mu. Mol) and AcOH (153. Mu.g, 2.55. Mu. Mol) were added to (R) -2- (4- (piperidin-3-ylamino) -5,6,7, 8-tetrahydrophthalazin-1-yl) -5- (trifluoromethyl) phenol (100 mg, 255. Mu. Mol) in dry MeOH (1.00 mL), stirred for 5 min, formaldehyde (38.3 mg,1.27 mmol) was added and the resulting reaction mixture stirred at 25℃for 12 h. The reaction mixture was filtered, the filter cake was washed with MeOH, the filtrate was concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C100 x 30mm x 10um; mobile phase: [ a-water (NH 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the 30% -60% of B%; 10 min) to give the title compound (5.06 mg, yield 4.89%, white solid). LC-MS (ESI) m/z 407.3[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.31(br,s,1H),7.34(d,J=7.63Hz,1H),7.24-7.13(m,2H),5.57(br d,J=8.00Hz,1H),4.31-4.18(m,1H),2.93-2.84(m,1H),2.63-2.58(m,1H),2.40-2.28(m,4H),2.18(s,3H),2.03-1.89(m,2H),1.86-1.66(m,4H),1.64-1.39(m,4H)。
Example 23:(R) -5-chloro-2- (4- ((1-methylpiperidin-3-yl) oxy) phthalazin-1-yl) phenol
Step 1: synthesis of (R) -3- ((4- (4-chloro-2-hydroxyphenyl) phthalazin-1-yl) oxy) piperidine-1-carboxylic acid tert-butyl ester
Under the protection of nitrogen, pdCl is added 2 (dppf) (101 mg, 138. Mu. Mol) was added to (R) -3- ((4-chlorophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (500 mg,1.38 mmol), (4-chloro-2-hydroxyphenyl) boronic acid (284 mg,1.65 mmol) and anhydrous K 3 PO 4 (877 mg,4.13 mmol) of dioxane (1.00 mL)/water (0.10 mL) and the resulting reaction mixtureThe mixture was stirred at 100℃for 12 hours. After the reaction mixture was cooled to room temperature, it was filtered, and the cake was washed with EA (2.00 mL. Times.3), the organic phases were combined, washed with saturated brine (2.00 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=5/1-1/1) gave the objective compound (400 mg, yield 63.8%, light brown solid). LC-MS (ESI) m/z 455.2[ M+H ]] +
Step 2: synthesis of (R) -5-chloro-2- (4- (piperidin-3-yloxy) phthalazin-1-yl) phenol
HCl-dioxane (4.00M, 2.00 mL) was added dropwise to (R) -3- ((4- (4-chloro-2-hydroxyphenyl) phthalazin-1-yl) oxo) piperidine-1-carboxylic acid tert-butyl ester (180 mg, 396. Mu. Mol) of dioxane (2.00 mL) at 0deg.C and the resulting reaction mixture was stirred at 25deg.C for 12 hours. The reaction mixture was concentrated under reduced pressure to give the objective compound (100 mg, yield 71.1% as a white solid) which was directly used in the next reaction. LC-MS (ESI) m/z 355.2[ M+H ] ] +
Step 3: synthesis of (R) -5-chloro-2- (4- ((1-methylpiperidin-3-yl) oxo) phthalazin-1-yl) phenol
At 0 ℃, naBH is added 3 CN (106 mg,1.69 mmol) was added to (R) -5-chloro-2- (4- (piperidin-3-yloxy) phthalazin-1-yl) phenol (200 mg, 564. Mu. Mol) and HOAc (338. Mu.g, 5.64. Mu. Mol) in anhydrous MeOH (2.00 mL), stirred for 5 min, then paraformaldehyde (84.6 mg,2.82 mmol) was added and the reaction mixture stirred at 25℃for 12 h. The reaction mixture was filtered, the filter cake was washed with MeOH (2.00 mL. Times.3), the filtrates were combined, concentrated under reduced pressure, and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100X 30mm X10 um; mobile phase: A- [ water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the 25% -55% of B%; 10 min) to give the title compound (20.5 mg, yield 9.86%, white solid). LC-MS (ESI) m/z 369.1[ M+H ]] +1 HNMR(400MHz,DMSO-d 6 )δppm 10.20(br s,1H),8.38(d,J=8.13Hz,1H),7.87-7.81(m,1H),7.80-7.73(m,1H),7.49-7.43(m,1H),7.33-7.26(m,1H),7.06(d,J=7.63Hz,1H),7.03-6.98(m,2H),4.48-4.34(m,1H),3.07(br d,J=7.63Hz,1H),2.70(br d,J=10.88Hz,1H),2.21(s,3H),2.03-1.87(m,3H),1.81-1.70(m,1H),1.67-1.54(m,1H),1.53-1.39(m,1H)。
Example 24:(R) -2- (4- ((1-methylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethoxy) benzene Phenol
Step 1: synthesis of (R) -3- ((4- (2-methoxy-4- (trifluoromethoxy) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
(R) -3- ((4-Chlorephthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (500 mg,1.38 mmol) was dissolved in a mixed solution of water (0.10 mL) and dioxane (1.00 mL) at 25℃followed by sequential addition of [ (2-methoxy-4- (trifluoromethoxy) phenyl) boronic acid (399mg, 1.66 mmol), pdCl 2 (dppf) (101 mg, 138. Mu. Mol) and anhydrous K 3 PO 4 (879 mg,4.14 mmol). The resulting reaction mixture was stirred under nitrogen at 100℃for 12 hours. After the reaction mixture was cooled to room temperature, water (2.00 mL) was added, extracted with EA (2.00 mL. Times.3), the organic phases were combined, washed with saturated brine (2.00 mL), and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=5/1 to 1/1) gave the objective compound (400 mg, yield 55.9%, white solid). LC-MS (ESI) m/z 519.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.40(br d,J=8.07Hz,1H),7.82-7.92(m,1H),7.74-7.81(m,1H),7.43(br d,J=8.31Hz,1H),7.34(d,J=7.58Hz,1H),7.05-7.23(m,3H),4.26(br s,1H),3.76-3.96(m,1H),3.69(s,3H),3.59-3.67(m,1H),2.59-3.20(m,2H),2.09(br d,J=8.80Hz,1H),1.59-1.92(m,2H),1.16-1.58(m,10H)。
Step 2: synthesis of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethoxy) phenol
To a solution of (R) -3- ((4- (2-methoxy-4- (trifluoromethoxy) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (200 mg, 386. Mu. Mol) in DCM (5.00 mL) at 25℃under nitrogen, BBr was added dropwise 3 (483 mg,1.93 mmol). The resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure to give the title compound (100 mg, yield 64.1% as a white solid). LC-MS (ESI) m/z 405.2[M+H] +
Step 3: synthesis of (R) -2- (4- ((1-methylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethoxy) phenol
To a solution of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethoxy) phenol (200 mg, 495. Mu. Mol) in anhydrous MeOH (2.00 mL) at 0deg.C was added NaBH 3 CN (93.2 mg,1.48 mmol) and ice AcOH (297. Mu.g, 4.95. Mu. Mol), after stirring for 5 minutes, formaldehyde (74.3 mg,2.47 mmol) was added dropwise and the resulting reaction was stirred at 25℃for 12 hours. The reaction solution was filtered, the filter cake was washed with MeOH (2.00 mL), the organic phases were combined, concentrated under reduced pressure, and the residue was purified by Prep-HPLC (column: waters Xbridge BEH C, 100X 30mm X10 um; mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -60%;10 min) to give the title compound (11.7 mg, yield 5.67%, white solid). LC-MS (ESI) m/z 419.2[ M+H ]] +1 HNMR(400MHz,DMSO-d 6 )δppm 10.38(br,s,1H),8.39(d,J=8.25Hz,1H),7.83-7.89(m,1H),7.75-7.82(m,1H),7.46(d,J=8.13Hz,1H),7.37-7.43(m,1H),7.08(br d,J=7.75Hz,1H),6.93(br s,2H),4.36-4.49(m,1H),3.01-3.13(m,1H),2.71(br d,J=10.63Hz,1H),2.22(s,3H),1.86-2.04(m,3H),1.71-1.81(m,1H),1.55-1.68(m,1H),1.46(qd,J=11.59,3.88Hz,1H)。
Examples 25 and 26:(R) -2- (4- ((1-methylpiperidin-3-yl) amino) thieno [2, 3-d)]Pyridazin-7-yl) 5- (trifluoromethyl) phenol and (R) -2- (7- ((1-methylpiperidin-3-yl) amino) thieno [2,3 ]d]Pyridazin-4-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of tert-butyl (R) -3- ((7-chlorothieno [2,3-d ] pyridazin-4-yl) amino) piperidine-1-carboxylate and tert-butyl (R) -3- ((4-chlorothieno [2,3-d ] pyridazin-7-yl) amino) piperidine-1-carboxylate
(3R) -3-aminopiperidine-1-carboxylic acid tert-butyl ester (1.17 g,5.85 mmol) and K at 25 ℃ 2 CO 3 (1.01 g,7.31 mmol) y was added sequentially to 4, 7-dichlorothieno [2,3-d ]]Pyridazine (PYRIZE)(1.00 g,4.88 mmol) in anhydrous DMF (30.0 mL) and the resulting reaction mixture was stirred at 100deg.C for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with EA (50.0 mL), washed with water (15.0 mL. Times.3), and the organic phase was dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and purification of the residue by silica gel chromatography (PE/ea=5/1 to 2/1) gave a stereoisomeric mixture of the target compound (350 mg, crude, yellow solid) which was used directly in the next step. LC-MS (ESI) m/z 369.1 371.1[ M+H ]] +1 H NMR(400MHz,DMSO-d6)δppm 8.15-8.27(m,1H),7.81-7.90(m,1H),7.52(d,J=5.25Hz,1H),7.72(d,J=5.25Hz,1H),7.16-7.32(m,1H),4.10(br d,J=2.88Hz,1H),3.46-3.98(m,2H),2.96-3.18(m,1H),2.61-2.72(m,1H),2.03(br s,1H),1.54-1.90(m,3H),1.37(s,9H)。
Step 2: synthesis of tert-butyl (R) -3- ((7- (2-hydroxy-4- (trifluoromethyl) phenyl) thieno [2,3-d ] pyridazin-4-yl) amino) piperidine-1-carboxylate and tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) thieno [2,3-d ] pyridazin-7-yl) amino) piperidine-1-carboxylate
The mixture obtained in the last step (350 mg,949 mmol) is H at 25℃under nitrogen protection 2 To a mixed solution of O (1.00 mL) and dioxane (10.0 mL), successively added (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (469 mg,2.28 mmol) and Pd (dppf) 2 Cl 2 (69.4 mg,94.9 mmol) and K 3 PO 4 (604 mg,2.85 mmol) and the resulting reaction mixture was stirred at 100deg.C for 12 hours. The reaction mixture was cooled to 25 ℃, filtered, the filtrate concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 50% -80%;10 min), separating and purifying to obtain the following components:
(R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) thieno [2, 3-d)]Pyridazin-7-yl) amino piperidine-1-carboxylic acid tert-butyl ester (50.0 mg, yield 10.6%, white solid), [ LC-MS (ESI) m/z:495.2[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δppm 11.52(br,s,1H),8.12(d,J=5.26Hz,1H),7.74(d,J=8.55Hz,1H),7.44(br d,J=4.38Hz,1H),7.27-7.33(m,2H),7.24(d,J=6.80Hz,1H),4.23(dt,J=5.92,2.96Hz,1H),3.58-4.05(m,1H),2.71-3.28(m,1H),2.00-2.13(m,1H),1.57-1.89(m,2H),1.14-1.55(m,12H)]A kind of electronic device
(R) -3- ((7- (2-hydroxy-4- (trifluoromethyl) phenyl) thieno [2, 3-d)]Pyridazin-4-yl) amino piperidine-1-carboxylic acid tert-butyl ester (100 mg, yield 21.3%, white solid). LC-MS (ESI) m/z 495.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 13.26(br,s,1H),8.23(d,J=5.48Hz,1H),8.01(br dd,J=4.38,2.85Hz,1H),7.95(d,J=8.55Hz,1H),7.30-7.39(m,2H),7.28(d,J=1.10Hz,1H),4.17(td,J=6.30,4.93Hz,1H),3.53-4.05(m,1H),2.72-3.24(m,1H),2.07(br d,J=7.89Hz,1H),1.56-1.93(m,2H),1.07-1.52(m,12H)。
Step 3: synthesis of (R) -2- (4- (piperidin-3-ylamino) thieno [2,3-d ] pyridazin-7-yl) -5- (trifluoromethyl) phenol
A solution of HCl/dioxane (4M, 0.500 mL) was added dropwise to (R) -3- ((7- (2-hydroxy-4- (trifluoromethyl) phenyl) thieno [2, 3-d) at 25 ℃]In a solution of t-butyl pyridazin-4-yl) amino-piperidine-1-carboxylate (100 mg,202 mmol) in dioxane (0.500 mL), the resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure to give the objective compound (100 mg, crude product, white solid) which was directly used for the next reaction. LC-MS (ESI) m/z 395.2[ M+H ]] +
Step 4: synthesis of (R) -2- (7- (piperidin-3-ylamino) thieno [2,3-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
At 25 ℃, to (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) thieno [2, 3-d)]To a solution of t-butyl pyridazin-7-yl) amino-piperidine-1-carboxylate (50.0 mg,101 mmol) in dioxane (1.00 mL) was added dropwise HCl/dioxane solution (4M, 25.3 mL) and the resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure to give the objective compound (50 mg, crude product, pale yellow solid) which was directly used for the next reaction. LC-MS (ESI) m/z 395.2[ M+H ] ] +
Step 5: synthesis of (R) -2- (4- ((1-methylpiperidin-3-yl) amino) thieno [2,3-d ] pyridazin-7-yl) -5- (trifluoromethyl) phenol
To (R) -2- (4- (piperidin-3-ylamino) thieno [2, 3-d) at 25 ℃C]To a solution of pyridazin-7-yl) -5- (trifluoromethyl) phenol (100 mg,254 mmol) in MeOH (3.00 mL) was added successively paraformaldehyde(76.1 mg,2.54 mmol) and AcOH (1.52 mg,25.4 mmol), the resulting mixture was stirred at 25℃for 30 minutes, and then NaBH was added 3 CN (79.7 mg,1.27 mmol) and the resulting reaction mixture was stirred at 25℃for 12 hours. The reaction mixture was filtered and the filtrate was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -80%;10 min) to give the title compound (20.9 mg, yield 20.1% as a white solid). LC-MS (ESI) m/z 409.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 13.20(br s,1H),8.24(d,J=5.26Hz,1H),8.03(d,J=5.26Hz,1H),7.98(d,J=8.11Hz,1H),7.35(br dd,J=7.78,4.28Hz,2H),7.31(s,1H),4.32-4.45(m,1H),3.06(br d,J=9.87Hz,1H),2.71(br d,J=10.74Hz,1H),2.21(s,3H),1.96-2.05(m,1H),1.85-1.96(m,2H),1.71-1.81(m,1H),1.54-1.68(m,1H),1.34-1.47(m,1H)。
Step 6: synthesis of (R) -2- (7- ((1-methylpiperidin-3-yl) amino) thieno [2,3-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
HCHO (34.9 mg,1.16 mmol) was added to (R) -2- (7- (piperidin-3-ylamino) thieno [2, 3-d) at 25 ℃]To a solution of pyridazin-4-yl) -5- (trifluoromethyl) phenol (50.0 mg,116 mmol) in MeOH (3.00 mL) was then added dropwise AcOH (6967 mg,11.6 mmol) and the resulting mixture was stirred for 30 min before NaBH was added 3 CN (36.46 mg,580.22 mmol) and the resulting reaction mixture was stirred at 25℃for 12 hours. The reaction mixture was filtered and the filtrate was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 20% -50%;10 min) to give the title compound (12.7 mg, yield 24.7%, white solid). LC-MS (ESI) m/z 409.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 11.43(br,s,1H),8.10(d,J=5.48Hz,1H),7.74(d,J=8.11Hz,1H),7.42(d,J=5.26Hz,1H),7.26-7.32(m,2H),7.14(br d,J=7.89Hz,1H),4.33-4.45(m,1H),3.00-3.08(m,1H),2.68~2.73(m,1H),2.20(s,3H),1.83-2.01(m,3H),1.68-1.79(m,1H),1.54-1.65(m,1H),1.36-1.50(m,1H)。
Example 28:5- (difluoromethyl) -2- (4- (quinuclidin-3-ylamino) phthalazin-1-yl) phenol
Step 1: synthesis of N- (4-chlorophthalazin-1-yl) quinuclidin-3-amine
CsF (2.29 g,15.0 mmol) was added to a solution of 1, 4-dichlorophthalazine (1.00 g,5.02 mmol) and quinuclidin-3-amine (817 mg,5.02 mmol) in DMSO (20.0 mL) and the resulting reaction mixture stirred at 100deg.C for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (20.0 mL), extracted with EA (10.0 mL. Times.3), and the aqueous phase was then extracted with saturated Na 2 CO 3 The solution was adjusted ph=8, extracted with EA (10.0 mL x 2), the final aqueous phase was washed with DCM (20.0 mL) and the aqueous phase concentrated under reduced pressure to give the title compound (500 mg, yield 34.4%, yellow oil). LC-MS (ESI) m/z 289.1[ M+H ]] +
Step 2: synthesis of N- (4- (4- (difluoromethyl) -2- (methoxymethoxy) phenyl) phthalazin-1-yl) quinuclidin-3-amine
RuPhos Pd G3 (74.5 mg, 89.1. Mu. Mol) was added to 2- (4- (difluoromethyl) -2- (methoxymethoxy) phenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (280 mg, 891. Mu. Mol), N- (4-chlorophthalazin-1-yl) quinuclidin-3-amine (3836 mg,1.34 mmol) and K under nitrogen 2 CO 3 (369 mg,2.67 mmol) of dioxane (3.00 mL)/water (0.30 mL) was added and the resulting reaction mixture was stirred under nitrogen at 100deg.C for 12 hours. The reaction mixture was cooled to room temperature and filtered, the filtrate was concentrated under reduced pressure and the residue was purified by Pre-HPLC (column: phenomnex C18X 30mm X3 um; mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 20% -55%;8 min) to give the objective compound (100 mg, yield 25.4%, white solid). LC-MS (ESI) m/z 441.2[ M+H ]] +
Step 3: synthesis of 5- (difluoromethyl) -2- (4- (quinuclidin-3-ylamino) phthalazin-1-yl) phenol
A solution of HCl-dioxane (4M, 0.30 mL) was added to N- (4- (4- (difluoromethyl) -2- (methoxymethoxy) phenyl) phthalazin-1-yl) quinuclidin-3-amine (100 mg, 227. Mu. Mol) in DCM (1.00 mL) at 0deg.C, followed by stirring at 25deg.C for 12 hours.The solvent was removed from the reaction solution under reduced pressure to give the title compound (22.8 mg, yield 25.4% as a white solid). LC-MS (ESI) m/z 397.1[ M+H ] ] +1 H NMR(400MHz,MeOH-d 4 )δppm 8.82(br d,J=7.67Hz,1H),8.17(br d,J=7.67Hz,1H),8.04(br t,J=7.23Hz,1H),7.79-7.93(m,1H),7.56(br d,J=7.45Hz,1H),7.20-7.31(m,2H),6.62-7.04(m,1H),4.64-4.76(m,1H),3.91-4.08(m,1H),3.56-3.75(m,2H),3.35-3.51(m,3H),2.61-2.74(m,1H),2.37-2.51(m,1H),2.11-2.31(m,2H),1.95-2.07(m,1H)。
Example 30:(R) -2- (4- ((1-methylpyrrolidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) benzene Phenol
Step 1: synthesis of (R) -3- ((4-chlorophthalazin-1-yl) amino) pyrrolidine-1-carboxylic acid tert-butyl ester
To a solution of 1, 4-dichlorophthalazine (2.14 g,10.7 mmol) and tert-butyl (R) -3-aminopyrrolidine-1-carboxylate (2.00 g,10.7 mmol) in DMSO (50.0 ml) at 25℃was added CsF (4.89 g,32.2 mmol) and the resulting reaction mixture was stirred at 100℃for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (40.0 mL) and EA (40.0 mL) and separated, then the aqueous phase was extracted with EA (30.0 mL. Times.3), the organic phases were combined, and the mixture was extracted with anhydrous Na 2 SO 4 Drying, filtration and removal of the solvent under reduced pressure gave the title compound (3.62 g, 96.6% yield as yellow solid) which was isolated without further purification and used in the next step. LC-MS (ESI) m/z 349.1[ M+H ]] +
Step 2: synthesis of (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) pyrrolidine-1-carboxylic acid tert-butyl ester
(R) -3- ((4-Chlorephthalazin-1-yl) amino) pyrrolidine-1-carboxylic acid tert-butyl ester (3.60 g,10.3 mmol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (2.13 g,10.3 mmol) and K at 25 ℃ 2 CO 3 (4.28 g,30.9 mmol) was dissolved in a mixed solution of dioxane (60.0 ml) and water (6.00 ml). RuPhos Pd G3 (863 mg,1.03 mmol) was added to the above mixture under nitrogen protection The resulting reaction mixture was stirred under nitrogen at 100℃for 12 hours. After the reaction mixture was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=1/1) to give the objective compound (2.00 g, yield 40.8%, pale yellow solid). LC-MS (ESI) m/z 475.2[ M+H ]] +
Step 3: synthesis of (R) -2- (4- (pyrrolidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) pyrrolidine-1-carboxylate (1.00 g,2.11 mmol) in anhydrous DCM (30.0 mL) at 0deg.C was added dropwise a solution of HCl-dioxane (4M, 2.63 mL) and after the addition was completed the reaction was stirred at 25deg.C for 12 hours. The reaction solution was concentrated under reduced pressure to give the objective compound (750 mg, yield 95.1%, yellow solid) which was used directly in the next step without further purification. LC-MS (ESI) m/z 375.1[ M+H ]] +
Step 4: synthesis of (R) -2- (4- ((1-methylpyrrolidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of (R) -2- (4- (pyrrolidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol (300 mg, 801. Mu. Mol) and formaldehyde (241 mg,8.01 mmol) in dry MeOH (20.0 mL) was added dropwise ice AcOH (4.81 mg, 80.1. Mu. Mol) and the resulting mixture was stirred for 30 min, then NaBH was added 3 CN (252 mg,4.01 mmol). The resulting reaction mixture was stirred at 25℃for 12 hours. The reaction mixture was concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 10% -50%;8 min) to give the title compound (28.0 mg, yield 8.99% as a yellow solid). LC-MS (ESI) m/z 389.1[ M+H ]] +1 H NMR(400MHz,DMSO-d6)δppm 10.36(br s,1H),8.46(d,J=8.23Hz,1H),7.82-7.89(m,1H),7.74-7.81(m,1H),7.52(d,J=7.63Hz,1H),7.44(d,J=7.51Hz,2H),7.30(d,J=7.99Hz,1H),7.27(s,1H),4.67-4.80(m,1H),2.87(dd,J=9.06,7.27Hz,1H),2.65-2.73(m,1H),2.59(br dd,J=9.30,4.65Hz,1H),2.41-2.48(m,1H),2.32-2.37(m,1H),2.30(s,3H),1.86-2.01(m,1H)。
Example 31:(S) -2- (4- (((1-methylpyrrolidin-2-yl) methyl)Group) amino) phthalazin-1-yl) -5- (trifluoromethyl Radical) phenol
Step 1: synthesis of (S) -2- (((4-chlorophthalazin-1-yl) amino) methyl) pyrrolidine-1-carboxylic acid tert-butyl ester
To a solution of 1, 4-dichlorophthalazine (2.00 g,10.1 mmol) in dimethyl sulfoxide (20.0 mL) at 25℃were added tert-butyl (S) -2- (aminomethyl) pyrrolidine-1-carboxylate (3.01 g,10.1 mmol) and CsF (4.55 g,30.0 mmol), and the resulting reaction mixture was stirred at 100℃for 12 hours. The reaction mixture was cooled to room temperature, water (20.0 mL) was added to the reaction mixture, extracted with EA (20.0 mL. Times.3), and the organic phases were combined with anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (3.60 g, yield 98.7%, yellow solid). LC-MS (ESI) m/z 363.3[ M+H ]] +
Step 2: synthesis of (S) -2- (((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) methyl) pyrrolidine-1-carboxylic acid tert-butyl ester
To a mixed solution of (S) -tert-butyl 2- (((4-chlorophthalazin-1-yl) amino) methyl) pyrrolidine-1-carboxylate (2.00 g,5.51 mmol) of dioxane (20.0 mL) and water (2.00 mL) at 25℃were added successively (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (1.25 g,6.06 mmol) and K 2 CO 3 (2.29G, 16.5 mmol) and RuPhos Pd G3 (4631 mg, 551. Mu. Mol) were added under nitrogen after the addition, and the resulting reaction mixture was stirred at 100℃for 12 hours. The reaction mixture was cooled to room temperature, water (20.0 mL) was added to the reaction mixture, extracted with EA (20.0 mL. Times.3), and the organic phases were combined with anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=1/1) gave the objective compound (1.90 g, yield 70.56%, yellow solid). LC-MS (ESI) m/z 489.3[ M+H ]] +
Step 3: synthesis of (S) -2- (4- ((pyrrolidin-2-ylmethyl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
At 0deg.C, to (S) -2- (((4- (2-hydroxy)To a solution of tert-butyl 4- (trifluoromethyl) phenyl) phthalazin-1-yl amino) methyl pyrrolidine-1-carboxylate (1.03 g,2.12 mmol) in DCM (10.0 mL) was added dropwise a solution of HCl-dioxane (4M, 2.65 mL) and the reaction was stirred at 25℃for 12 h after the addition. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure to give the objective compound (800 mg, yield 97.3%, yellow solid). LC-MS (ESI) m/z 389.3[ M+H ] ] +
Step 4: synthesis of (S) -2- (4- (((1-methylpyrrolidin-2-yl) methyl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of (S) -2- (4- ((pyrrolidin-2-ylmethyl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol (500 mg,1.29 mmol) in MeOH (5.00 mL) at 0deg.C. Sequentially adding NaBH 3 CN (405 mg,6.44 mmol) and ice AcOH (7.73 mg, 129. Mu. Mol), and after stirring the resultant reaction solution at 0℃for 5 minutes, formaldehyde (387 mg,12.8 mmol) was added dropwise, and the resultant reaction mixture was stirred at 25℃for 12 hours. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure at 25℃and the residue was purified by Prep-HPLC (column: waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 25% -55%;8 min) to give the title compound (41.3 mg, yield 7.98%, yellow solid). LC-MS (ESI) m/z 403.0[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.37(br,s,1H),8.33(d,J=8.11Hz,1H),7.83-7.93(m,1H),7.75-7.82(m,1H),7.52(d,J=7.89Hz,1H),7.45(d,J=8.11Hz,1H),7.30(d,J=8.77Hz,1H),7.27(s,1H),3.84(br d,J=13.15Hz,1H),3.42-3.49(m,1H),2.99-3.05(m,1H),2.59-2.74(m,1H),2.42(s,3H),2.34-2.48(m,1H),1.89-1.99(m,1H),1.64-1.74(m,3H)。
Example 32:(R) 2- (7- ((1-ethylpiperidin-3-yl) amino) furo [2,3- ]d]Pyridazin-4-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of methyl furan-2, 3-dicarboxylate
To MeOH in furan-2, 3-dicarboxylic acid (5.00 g,32.0 mmol) at 25 ℃To the solution (50.0 mL) was added thionyl chloride (19.1 g,160 mmol) dropwise, and the resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure, and the residue was taken up in saturated NaHCO 3 In aqueous solution (20.0 mL), extracted with EA (20.0 mL. Times.3), the organic phases were combined, and dried over anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (5.58 g, yield 99.1% as a clear oil). LC-MS (ESI) m/z 185.2[ M+H ]] +
Step 2: synthesis of furo [2,3-d ] pyridazine-4, 7-diol
To a solution of methyl furan-2, 3-dicarboxylate (4.00 g,21.7 mmol) in MeOH (50.0 mL) at 25℃was added dropwise hydrazine hydrate (5.55 g,108 mmol). The resulting reaction solution was stirred at 80℃for 12 hours. The reaction solution was cooled and concentrated under reduced pressure. The residue was stirred with 2.00mol/L hydrochloric acid solution at 100℃for 12 hours, filtered and dried to give the objective compound (2.67 g, yield 80.8%, white solid). LC-MS (ESI) m/z 153.2[ M+H ]] +
Step 3: synthesis of 4, 7-dichlorofuro [2,3-d ] pyridazine
At 25 ℃, furo [2,3-d ]]To a solution of pyridazine-4, 7-diol (1.00 g,6.57 mmol) in ACN (10.0 mL) were added pyridine (1.04 g,13.15 mmol) and phosphorus oxychloride (5.04 g,32.9 mmol) dropwise, and the resulting reaction solution was stirred at 80℃for 12 hours. The reaction mixture was diluted with ice water (10.0 mL), extracted with EA (20.0 mL. Times.3), and the organic phases combined with anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (1.20 g, yield 96.5%, white solid). LC-MS (ESI) m/z 189.1[ M+H ] ] +
Step 4: synthesis of tert-butyl (R) -3- ((4-chlorofuro [2,3-d ] pyridazin-7-yl) amino) piperidine-1-carboxylate and tert-butyl (R) -3- ((7-chlorofuro [2,3-d ] pyridazin-4-yl) amino) piperidine-1-carboxylate
At 25 ℃,4, 7-dichloro-furo [2,3-d]To a solution of pyridazine (1.00 g,5.29 mmol) in DMSO (10.0 mL) were added tert-butyl (R) -3-aminopiperidine-1-carboxylate (2.12 g,10.6 mmol) and cesium fluoride (2.41 g,15.9 mmol), and the resulting reaction mixture was stirred at 100deg.C for 12 hours. The reaction mixture was cooled to room temperature, diluted with water (5.00 mL), extracted with EA (5.00 mL. Times.3), the organic phases combined, and dried over anhydrous Na 2 SO 4 Drying, filtering, concentrating under reduced pressure, and separating and purifying the residue by silica gel chromatography (PE/EA=1/1) to obtain the target compound (R) -3- ((4-chlorofuro [2, 3-d) respectively]Pyridazin-7-yl) amino piperidine-1-carboxylic acid tert-butyl ester (300 mg, yield 16.0%, yellow colour oil), LC-MS (ESI) m/z 353.3[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 ) Delta ppm 8.35-8.44 (m, 1H), 7.41 (br d, j=6.50 hz, 1H), 7.07-7.13 (m, 1H), 4.04-4.13 (m, 1H), 3.42-3.87 (m, 2H), 1.47-1.89 (m, 3H), 1.07-1.46 (m, 12H); target compound (R) -3- ((7-chlorofuro [2, 3-d)]Pyridazin-4-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (400 mg, yield 21.4%, yellow oil). LC-MS (ESI) m/z 353.3[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.24(d,J=2.13Hz,1H),7.24-7.34(m,2H),3.99-4.03(m,1H),3.64-3.85(m,2H),2.03(br d,J=3.13Hz,1H),1.77-1.85(m,1H),1.43-1.63(m,4H),1.25-1.41(m,9H)。
Step 5: synthesis of tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) furo [2,3-d ] pyridazin-7-yl) amino) piperidine-1-carboxylate
At 25 ℃, to (R) -3- ((4-chlorofuro [2, 3-d)]To a mixed solution of t-butyl pyridazin-7-yl) amino-piperidine-1-carboxylate (300 mg, 850. Mu. Mol) in dioxane (10.0 mL)/water (1.00 mL), K was added 2 CO 3 (353 mg,2.55 mmol) and (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (263 mg,1.28 mmol) were added under nitrogen and RuPhos Pd G3 (142 mg, 170. Mu. Mol) was added. The resulting reaction mixture was stirred at 90℃for 12 hours. After the reaction solution was cooled to room temperature, it was then filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=1/2) to give the objective compound (260 mg, yield 63.9%, yellow solid). LC-MS (ESI) m/z 479.3[ M+H ]] +
Step 6: synthesis of (R) -2- (7- (piperidin-3-ylamino) furo [2,3-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
At 0deg.C, to (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) furo [2, 3-d)]To a solution of t-butyl pyridazin-7-yl) amino-piperidine-1-carboxylate (120 mg, 251. Mu. Mol) in DCM (5.00 mL) was added dropwise a solution of HCl-dioxane (4M, 2.00 mL), and the resulting reaction solution was stirred at 25℃for 12 hours. Filtering the reaction liquid, and filtering The solution was concentrated under reduced pressure to give the title compound (90.0 mg, yield 94.8%, yellow solid). LC-MS (ESI) m/z 379.2[ M+H ]] +
Step 7: synthesis of (R) -2- (7- ((1-ethylpiperidin-3-yl) amino) furo [2,3-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
At 0℃to (R) -2- (7- (piperidin-3-ylamino) furo [2, 3-d)]To a solution of pyridazin-4-yl) -5- (trifluoromethyl) phenol (90 mg, 237.88. Mu. Mol) in EtOH (2.00 mL) was added NaBH successively 3 CN (44.85 mg, 713.63. Mu. Mol) and AcOH (142.85 ug, 2.38. Mu. Mol), after stirring for 5 minutes, acetaldehyde (5M, 237.88. Mu.L) was added dropwise and the resulting reaction mixture was stirred at 0℃for 1 hour. The reaction mixture was filtered, the filtrate concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 5% -75%;8 min) to give the title compound (62.7 mg, yield 64.8%, yellow solid). LC-MS (ESI) m/z 407.2[ M+H ]] +11 HNMR(400MHz,DMSO-d 6 )δppm 14.25(s,1H),8.49(d,J=2.15Hz,1H),8.16(d,J=8.23Hz,1H),7.60(d,J=2.15Hz,1H),7.52(br d,J=7.99Hz,1H),7.28(s,1H),7.26(br,s,1H),4.27-4.38(m,1H),3.01-3.07(m,1H),2.72-2.78(m,1H),2.37(q,J=7.15Hz,2H),1.90-2.03(m,3H),1.70-1.76(m,1H),1.46-1.62(m,2H),1.01(t,J=7.15Hz,3H)。
Example 33:(R) 2- (4- ((1-ethylpiperidin-3-yl) amino) furo [2,3 ]d]Pyridazin-7-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of tert-butyl (R) -3- ((7- (2-hydroxy-4- (trifluoromethyl) phenyl) furo [2,3-d ] pyridazin-4-yl) amino) piperidine-1-carboxylate
At 25 ℃, to (R) -3- ((7-chlorofuro [2, 3-d) ]To a mixed solution of t-butyl pyridazin-4-yl) amino-piperidine-1-carboxylate (400 mg,1.13 mmol) of dioxane (10.0 mL)/water (1.00 mL) was added K 2 CO 3 (469 mg,3.39 mmol) and (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid(233 mg,1.13 mmol) and RuPhos Pd G3 (189 mg, 226. Mu. Mol) were added under nitrogen and the resulting reaction mixture was stirred at 90℃for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (10.0 mL), extracted with EA (10.0 mL. Times.3), the organic phases were combined, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=1/2) gave the objective compound (300 mg, yield 55.5%, yellow solid). LC-MS (ESI) m/z 479.3[ M+H ]] +
Step 2: synthesis of (R) -2- (4- (piperidin-3-ylamino) furo [2,3-d ] pyridazin-7-yl) -5- (trifluoromethyl) phenol
At 0deg.C, to (R) -3- ((7- (2-hydroxy-4- (trifluoromethyl) phenyl) furo [2, 3-d)]To t-butyl pyridazin-4-yl) amino-piperidine-1-carboxylate (150 mg, 314. Mu. Mol) in DCM (5.00 mL) was added dropwise a solution of HCl-dioxane (4M, 2.00 mL), and the resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure to give the objective compound (110 mg, yield 92.7%, yellow solid). LC-MS (ESI) m/z 379.3[ M+H ] ] +
Step 3: synthesis of (R) -2- (4- ((1-ethylpiperidin-3-yl) amino) furo [2,3-d ] pyridazin-7-yl) -5- (trifluoromethyl) phenol
At 0℃to (R) -2- (4- (piperidin-3-ylamino) furo [2, 3-d)]Pyridazin-7-yl) -5- (trifluoromethyl) phenol was dissolved in EtOH (2.00 mL) and NaBH was added sequentially 3 CN (54.8 mg, 872. Mu. Mol) and AcOH (175. Mu.g, 2.91. Mu. Mol), after stirring for 5 minutes, acetaldehyde (64.0 mg,1.45 mmol) was added dropwise, and the resulting reaction solution was stirred at 0℃for 1 hour. The reaction solution was concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 45% -70%;8 min) to give the title compound (56.71 mg, yield 48.0%, yellow solid). LC-MS (ESI) m/z 470.0[ M+H ]] +1 HNMR(400MHz,DMSO-d 6 )δppm 14.65(s,1H),8.49(d,J=7.88Hz,1H),8.40(d,J=2.00Hz,1H),7.55(br d,J=8.00Hz,1H),7.44(d,J=2.00Hz,1H),7.36(br d,J=8.00Hz,1H),7.29(s,1H),4.31(br dd,J=5.13,2.88Hz,1H),3.08-3.16(m,1H),2.76-2.84(m,1H),2.38(q,J=7.13Hz,2H),1.99-2.08(m,1H),1.87-1.97(m,2H),1.72-1.80(m,1H),1.53-1.64(m,1H),1.32-1.45(m,1H),1.01(t,J=7.19Hz,3H)。
Example 34: (R) -N- (2- (8- ((1-ethylpiperidin-3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5-methylphenyl) methanesulfonamide
/>
Step 1: synthesis of (R) -3- ((5-chloropyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
At 25 ℃, to 5, 8-dichloropyrido [2,3-d ]]A solution of pyridazine (3.00 g,15.0 mmol) in dry EtOH (50.0 mL) was added tert-butyl (R) -3-aminopiperidine-1-carboxylate (3.30 g,16.5 mmol) and the resulting reaction mixture was stirred at 70℃for 12 h. After the reaction mixture was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=1/1) to give the objective compound (900 mg, yield 16.5%, yellow solid). LC-MS (ESI) m/z 364.3[ M+H ] ] +1 HNMR(400MHz,DMSO-d 6 )δppm 9.23(dd,J=1.3,4.3Hz,1H),9.01-8.81(m,1H),8.10-7.93(m,1H),7.50(br s,1H),4.20-4.08(m,1H),3.94-3.45(m,2H),3.19-2.80(m,1H),2.01-1.54(m,3H),1.47(br dd,J=3.6,9.9Hz,2H),1.41-1.15(m,9H)。
Step 2: synthesis of tert-butyl (R) -3- ((5- (4-methyl-2- (methylsulfonylamino) phenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylate
XPhos Pd G3 (83.8 mg, 99.0. Mu. Mol) was added to (R) -3- ((5-chloropyrido [2, 3-d) under nitrogen]Pyridazin-8-yl) amino piperidine-1-carboxylic acid tert-butyl ester (180 mg, 495. Mu. Mol), N- (5-methyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) methanesulfonamide (154 mg, 495. Mu. Mol) and K 2 CO 3 (205 mg,1.48 mmol) of dioxane (10.0 mL)/water (1.00 mL) and the resulting reaction mixture was stirred at 90℃for 12 hours. After the reaction mixture was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=1/1) to give the objective compound (180 mg, yield 71.0%, yellow solid). LC-MS (ESI) m/z 513.3[ M+H] +
Step 3: synthesis of (R) -N- (5-methyl-2- (8- (piperidin-3-ylamino) pyrido [2,3-d ] pyridazin-5-yl) phenyl) methanesulfonamide
A solution of HCl-dioxane (4M, 4.00 mL) was added dropwise to (R) -3- ((5- (4-methyl-2- (methylsulfonylamino) phenyl) pyrido [2, 3-d) at 0deg.C]Pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (90.0 mg, 176. Mu. Mol) in DCM (20.0 mL) and the reaction mixture was stirred at 25℃for 12 h. The reaction mixture was concentrated under reduced pressure to give the objective compound (70.0 mg, yield 96.7%, yellow solid) which was directly used in the next reaction. LC-MS (ESI) m/z 413.3[ M+H ] ] +
Step 4: synthesis of (R) -N- (2- (8- ((1-ethylpiperidin-3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5-methylphenyl) methanesulfonamide
At 0 ℃, naBH is added 3 CN (32.0 mg, 509. Mu. Mol) was added to (R) -N- (5-methyl-2- (8- (piperidin-3-ylamino) pyrido [2, 3-d)]Pyridazin-5-yl) phenyl methanesulfonamide (70.0 mg, 170. Mu. Mol) and HOAc (1.02 mg, 17.0. Mu. Mol) in anhydrous EtOH (20.0 mL) were stirred for 10 minutes, then acetaldehyde (5M, 170. Mu.L) was added and the reaction mixture was stirred at 0℃for 0.5 hours. The reaction mixture was filtered and concentrated under reduced pressure, and the residue was purified by Prep-HPLC (column: waters Xbridge BEH C, 18, 100*30mm*10um;mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 15% -55%;8 min) to give the title compound (24.3 mg, yield 32.4% as a yellow solid). LC-MS (ESI) m/z 441.2[ M+H ]] +1 HNMR(400MHz,DMSO-d 6 )δppm 9.11(dd,J=4.28,1.64Hz,1H),7.92-7.87(m,1H),7.86-7.81(m,1H),7.43(s,1H),7.39(br d,J=8.11Hz,1H),7.30(d,J=7.67Hz,1H),7.15(d,J=7.89Hz,1H),4.46(br s,1H),3.34(s,2H),3.31(s,1H),2.95(s,3H),2.89-2.76(m,1H),2.48-2.43(m,2H),2.42(s,3H),1.82-1.67(m,3H),1.66-1.51(m,1H),1.04(t,J=7.13Hz,3H)。
Example 35:(R) -5- (2- (difluoromethyl) -4-methylphenyl) schemeN- (1-ethylpiperidin-3-yl) pyrido [2, 3-d]pyridazin-8-amines
Step 1 Synthesis of 1-bromo-2- (difluoromethyl) -4-methylbenzene
DAST (10.8 g,60.3 mmol) was added dropwise to a solution of 2-bromo-5-methylbenzaldehyde (6.00 g,30.2 mmol) in DCM (48.0 mL) at 25℃and the resulting reaction mixture stirred at 25℃for 3 h. Adding Na into the reaction mixture 2 CO 3 Aqueous (50.0 mL), extracted with DCM (50.0 mL. Times.3), the organic phases combined, dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (pe=100%) gave the title compound (4.70 g, yield 70.5%, transparent oil). 1 H NMR(400MHz,DMSO-d 6 ),δppm 7.62(d,J=8.11Hz,1H),7.49(s,1H),7.31(br d,J=8.11Hz,1H),7.24-6.95(m,1H),2.33(s,3H)。
Step 2 Synthesis of 2- (2- (difluoromethyl) -4-methylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolane
1-bromo-2- (difluoromethyl) -4-methylbenzene (4.60 g,20.8 mmol), pinacol borate (10.6 g,41.6 mmol), KOAc (6.13 g,62.4 mmol) and PdCl were successively reacted under nitrogen 2 (dppf) (3.05 mg,4.16 mmol) was added to dioxane (230 mL) and the resulting reaction mixture was stirred at 100deg.C for 12 hours. The reaction mixture was cooled to room temperature, filtered, the filter cake was washed with EA (50.0 ml×2), the filtrates were combined, concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=10/1 to 5/1 to 3/1) to give the title compound (2.10 g, yield 37.6%, transparent oil).
Step 3 Synthesis of tert-butyl (R) -3- ((5- (2- (difluoromethyl) -4-methylphenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylate
2- (2- (difluoromethyl) -4-methylphenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (210 mg, 783. Mu. Mol) and K were reacted at 25 ℃C 2 CO 3 (271mg, 1.96 mmol) to (R) -3- ((5-chloropyrido [2, 3-d)]To a mixture of t-butyl pyridazin-8-yl) amino piperidine-1-carboxylate (238 mg, 653. Mu. Mol) and dioxane (10.0 mL)/water (2.00 mL) was added Pd (dtbpf) Cl under nitrogen protection 2 (85.1 mg, 131. Mu. Mol) and the resulting reaction mixture was stirred at 90℃for 12 hours. The mixture to be reacted is cooledAfter cooling to room temperature, filtration, washing of the filter cake with EA (10.0 ml×2), combining the filtrates, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=1/2) gave the title compound (250 mg, yield 81.5%, transparent oil). LC-MS (ESI) m/z 470.3[ M+H ]] +
Step 4 Synthesis of (R) -5- (2- (difluoromethyl) -4-methylphenyl) -N- (piperidin-3-yl) pyrido [2,3-d ] pyridazin-8-amine
HCl-dioxane (4M, 2.00 mL) was added dropwise to (R) -3- ((5- (2- (difluoromethyl) -4-methylphenyl) pyrido [2, 3-d) at 0deg.C]Pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (130 mg, 277. Mu. Mol) in DCM (5.00 mL) and the resulting reaction mixture was stirred at 25℃for 5 h. The reaction mixture was concentrated directly under reduced pressure to give the title compound (100 mg, yield 97.7%, yellow solid). LC-MS (ESI) m/z 370.3[ M+H ]] +
Step 5 Synthesis of (R) -5- (2- (difluoromethyl) -4-methylphenyl) -N- (1-ethylpiperidin-3-yl) pyrido [2,3-d ] pyridazin-8-amine
At 0 ℃, naBH is added 3 CN (51.03 mg, 812. Mu. Mol) was added to (R) -5- (2- (difluoromethyl) -4-methylphenyl) -N- (piperidin-3-yl) pyrido [2, 3-d)]To a solution of pyridazin-8-amine (100 mg, 271. Mu. Mol) and AcOH (163. Mu.g, 2.71. Mu. Mol) in ethanol (3.00 mL) was added acetaldehyde (5M, 271. Mu.L) after stirring for 5 minutes, and the reaction mixture was stirred at 0℃for 1 hour. The reaction mixture was concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -60%;8 min) to give the title compound (33.2 mg, yield 30.8%, off-white solid). LC-MS (ESI) m/z 398.3[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm9.13(dd,J=3.75,2.25Hz,1H),7.90-7.81(m,2H),7.65(s,1H),7.52(br d,J=7.88Hz,1H),7.44-7.34(m,2H),6.87(t,J=54.97Hz,1H),4.45(br s,1H),2.80(br d,J=9.76Hz,1H),2.57-2.51(m,2H),2.49(br s,3H),2.45-2.29(m,4H),1.85-1.67(m,3H),1.63-1.51(m,1H),1.04(t,J=7.13Hz,3H)。
Example 36:(R) -2- (4- ((1-cyclobutylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) benzene Phenol
Step 1: synthesis of (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
To a mixed solution of (R) -3- ((4-chlorophthalazin-1-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (500 mg,1.38 mmol) of dioxane (1.00 mL)/water (0.10 mL) at 25℃was added successively (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (3411 mg,1.65 mmol), pdCl 2 (dppf) (101 mg, 138. Mu. Mol) and anhydrous K 3 PO 4 (877 mg,4.13 mmol). The resulting reaction mixture was stirred at 100℃for 12 hours. The reaction mixture was cooled to room temperature, diluted with water (2.00 mL), extracted with EA (2.00 mL. Times.3), the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=10:1/1:1) gave the title compound (400 mg, yield 59.4%, brown solid). LC-MS (ESI) m/z 489.3[ M+H ]] +
Step 2: synthesis of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of tert-butyl (R) -3- ((4- (2-hydroxy-4- (trifluoromethyl) phenyl) phthalazin-1-yl) amino) piperidine-1-carboxylate (400 mg, 818. Mu. Mol) in dioxane (4.00 mL) at 25℃was added dropwise a solution of HCl-dioxane (4M, 4.00 mL). The resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure at 25℃to give the title compound (300 mg, yield 94.3%, brown solid). LC-MS (ESI) m/z 389.2[ M+H ]] +
Step 3: synthesis of (R) -2- (4- ((1-cyclobutylpiperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol (50.0 mg, 129. Mu. Mol) in dry EtOH (3.00 mL) at 25℃was added NaBH successively 3 CN (24.3 mg, 386. Mu. Mol) and glacial AcOH (77.3. Mu.g, 1.29. Mu. Mol) were stirred for 5 minutes, after which cyclobutanone (9.93 mg, 142. Mu. Mol) was added dropwise. The resulting reaction solution was stirred at 25℃for a further 12 hours. Reaction solution subtractionThe residue was concentrated by pressure and purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 40% -60%;8 min) to give the title compound (2.24 mg, yield 3.93%, yellow solid). LC-MS (ESI) m/z 443.3[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.38(br d,J=8.25Hz,1H),7.80-7.88(m,1H),7.72-7.80(m,1H),7.50(br d,J=7.50Hz,1H),7.43(d,J=8.25Hz,1H),7.24-7.30(m,1H),7.11(br d,J=7.50Hz,1H),4.35-4.48(m,1H),4.03-4.14(m,1H),2.69-2.79(m,2H),1.91-2.02(m,3H),1.69-1.86(m,5H),1.45-1.67(m,4H)。
Example 37:(R) -2- (4- ((1- (oxa Ding Huanwan-3-yl) piperidin-3-yl) amino) phthalazin-1-yl) -5- (tri) Fluoromethyl) phenol
Step 1: synthesis of (R) -2- (4- ((1- (oxa Ding Huanwan-3-yl) piperidin-3-yl) amino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
To a solution of (R) -2- (4- (piperidin-3-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol (50.0 mg, 129. Mu. Mol) in dry EtOH (3.00 mL) at 25℃was added NaBH 3 CN (24.3 mg, 386. Mu. Mol) and ice AcOH (77.3. Mu.g, 1.29. Mu. Mol) were added, after stirring for 5 minutes, to oxa Ding Huanwan-3-one (10.2 mg, 142. Mu. Mol). The resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 25% -55%;8 min) to give the title compound (2.70 mg, yield 4.72%, yellow solid). LC-MS (ESI) m/z 445.2[ M+H ]] +1 H NMR(400MHz,METHANOL-d 4 )δppm 8.30(d,J=8.19Hz,1H),7.89(t,J=7.70Hz,1H),7.80(t,J=7.46Hz,1H),7.60(d,J=7.95Hz,1H),7.53(d,J=7.70Hz,1H),7.28-7.33(m,1H),7.24(s,1H),4.67-4.74(m,3H),4.65(t,J=6.00Hz,1H),4.54-4.61(m,1H),3.57(quin,J=6.66Hz,1H),3.03-3.11(m,1H),2.62(dt,J=7.55,1.54Hz,1H),2.02-2.21(m,3H),1.84-1.92(m,1H),1.66-1.82(m,2H)。
Example 38:2- (8- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) pyrido [2, 3-d)]Pyridazin-5-yl) -5- (trifluoromethyl) benzenePhenol
Step 1: synthesis of (1 s,3 s) -3- ((5-chloropyrido [2,3-d ] pyridazin-8-yl) amino) -1-methylcyclobutanol and (1 s,3 s) -3- ((8-chloropyrido [2,3-d ] pyridazin-5-yl) amino) -1-methylcyclobutanol
At 25 ℃, to 5, 8-dichloropyrido [2,3-d ]]To a solution of pyridazine (3.00 g,15.0 mmol) and (1 s,3 s) -3-amino-1-methylcyclobutanol hydrochloride (2.06 g,15.0 mmol) in DMSO (50.0 mL) was added CsF (6.83 g,45.0 mmol) and the resulting reaction mixture was stirred at 135℃for 12 hours. The reaction mixture was cooled to room temperature, diluted with water (40.0 mL), extracted with EA (30.0 mL. Times.3), the organic phases combined, and dried over anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave a mixture of the target compound (1.35 g, yield 34.0%, brown solid) which was used directly in the next reaction without further purification and isolation and specific position isomerism identification. LC-MS (ESI) m/z 265.2[ M+H ]] +
Step 2: synthesis of 2- (8- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
The mixture obtained in the previous step (200 mg,756 mmol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (77.8 mg, 378. Mu. Mol) and anhydrous K at 25 ℃ 2 CO 3 (157 mg,1.13 mmol) was dissolved in a mixture of dioxane (10.0 mL)/water (1.00 mL) and under nitrogen protection, (2-dicyclohexylphosphine-2, 4, 6-triisopropyl-1, 1-biphenyl) [2- (2-amino-1, 1-biphenyl) was added]Palladium mesylate (32.0 mg, 37.8. Mu. Mol). The resulting reaction solution was stirred at 90℃for 12 hours under nitrogen protection. The reaction solution was cooled to room temperature and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 25% -55%;8 min) to give the title compound 2- (8- (((1)s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) pyrido [2,3-d]Pyridazin-5-yl) -5- (trifluoromethyl) phenol (20.6 mg, yield 4.00%, white solid). LC-MS (ESI) m/z 391.0[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.45(br s,1H),9.11(dd,J=4.25,1.50Hz,1H),7.89-7.93(m,1H),7.82-7.87(m,1H),7.59(t,J=7.25Hz,2H),7.27-7.35(m,2H),4.98(s,1H),4.25-4.37(m,1H),2.38-2.48(m,2H),2.18-2.25(m,2H),1.33(s,3H)。
Example 39:5-chloro-2- (8- (((1)s,3s) -3-hydroxy-3-methylcyclobutyl) amino) pyrido [2,3- ]d]Pyridazine (Da) Oxazin-5-yl) phenols
Step 1: synthesis of 5-chloro-2- (8- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) pyrido [2,3-d ] pyridazin-5-yl) phenol
At 25 ℃, to (1 s,3 s) -3- ((5-chloropyrido [2, 3-d)]To a mixed solution of pyridazin-8-yl) amino) -1-methylcyclobutanol (100 mg, 378. Mu. Mol) in dioxane (1.00 mL)/water (0.10 mL), was added (4-chloro-2-hydroxy-phenyl) boronic acid (78.1 mg, 453. Mu. Mol), pdCl 2 (dppf) (27.6 mg, 37.8. Mu. Mol) and K 3 PO 4 (241 mg,1.13 mmol). The resulting reaction mixture was stirred under nitrogen at 100 ℃ for 12 hours. After the reaction mixture was cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 15% -45%,8 min) to give the title compound (17.3 mg, 12.8% yield, grey solid). LC-MS (ESI) m/z 357.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.27(s,1H),9.10(dd,J=4.22,1.53Hz,1H),7.87-7.94(m,1H),7.82-7.86(m,1H),7.56(br d,J=6.60Hz,1H),7.36(d,J=8.56Hz,1H),7.00-7.08(m,2H),5.00(s,1H),4.22-4.36(m,1H),2.45-2.48(m,2H),2.15-2.26(m,2H),1.32(s,3H)。
Example 40:2- (8- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethoxy) amino)
Phenol (P)
Step 1: synthesis of 2- (8- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethoxy) phenol
At 25 ℃, to (1 s,3 s) -3- ((5-chloropyrido [2, 3-d)]Pyridazin-8-yl) amino) -1-methylcyclobutanol and (1 s,3 s) -3- ((8-chloropyrido [2, 3-d)]To a mixed solution of dioxane (5.00 mL) and water (1.00 mL) of a mixture (100 mg, 378. Mu. Mol) of pyridazin-5-yl) amino) -1-methylcyclobutanol was added [ (2-hydroxy-4- (trifluoromethoxy) phenyl) boronic acid (126 mg, 567. Mu. Mol) and K in this order 2 CO 3 (157 mg,1.13 mmol) under nitrogen, pd (dtbpf) Cl was added 2 (49.2 mg, 75.6. Mu. Mol). The resulting reaction mixture was stirred at 90℃for 12 hours. The reaction solution was cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by Prep-HPLC (column: phenomenex Luna C, 75 x 30mm x 3um; mobile phase: [ a-water (FA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 1% -35%,8 min) to give the title compound (10.6 mg, yield 6.92%, grey solid). LC-MS (ESI) m/z 407.0[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.42(br s,1H),9.10(br d,J=3.95Hz,1H),7.87-7.92(m,1H),7.82-7.87(m,1H),7.57(br d,J=7.23Hz,1H),7.47(d,J=7.45Hz,1H),6.92-6.99(m,2H),4.99(s,1H),4.22-4.38(m,1H),2.40-2.44(m,2H),2.17-2.24(m,2H),1.32(s,3H)。
Example 41:5-chloro-2- (4- (((1)s,3s) -3-hydroxy-3-methylcyclobutyl) amino) phthalazin-1-yl) phenol
Step 1: synthesis of 5-chloro-2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) phthalazin-1-yl) phenol
At 25℃to 3- [ (4-chlorophthalazin-1-yl) amino group]-1-methyl-cyclobutanol(100 mg, 379. Mu. Mol) was dissolved in a mixed solution of dioxane (1.00 mL) and water (0.10 mL), and (4-chloro-2-hydroxy-phenyl) boric acid (78.4 mg, 455. Mu. Mol), pdCl were added in this order 2 (dppf) (27.8 mg, 37.9. Mu. Mol) and anhydrous K 3 PO 4 (241 mg,1.14 mmol). The resulting reaction solution was stirred at 100℃for 12 hours under nitrogen protection. The reaction solution was cooled to room temperature, filtered, the filtrate was diluted with EA (10.0 mL) and then washed with water (5.00 mL. Times.3), and the organic phase was taken up in anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column: phenomenex Luna C18, 75X 30mm X3 um; mobile phase: [ A-Water (FA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 1% -50%;8 min) to give the title compound (9.18 mg, yield 6.39%, yellow solid, 0.5FA salt). LC-MS (ESI) m/z 356.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.27(s,1H),9.10(dd,J=4.22,1.53Hz,1H),7.87-7.94(m,1H),7.82-7.86(m,1H),7.56(br d,J=6.60Hz,1H),7.36(d,J=8.56Hz,1H),7.00-7.08(m,2H),5.00(s,1H),4.22-4.36(m,1H),2.45-2.48(m,2H),2.15-2.26(m,2H),1.32(s,3H)。
Example 42:2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) phthalazin-1-yl) -5- (trifluormethyl) Oxy) phenol
Step 1: synthesis of 2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) phthalazin-1-yl) -5- (trifluoromethoxy) phenol
To a mixed solution of (1 s,3 s) -3- ((4-chlorophthalazin-1-yl) amino) -1-methylcyclobutanol (100 mg, 379. Mu. Mol) of dioxane (5.00 mL) and water (1.00 mL) at 25℃were added [ (2-hydroxy-4- (trifluoromethoxy) phenyl) boronic acid (126 mg, 568. Mu. Mol) and K 2 CO 3 (157 mg,1.14 mmol) under nitrogen, pd (dtbpf) Cl was added 2 (49.2 mg, 75.6. Mu. Mol). The resulting reaction mixture was stirred at 90℃for 12 hours. The reaction mixture was cooled to room temperature, filtered, the filtrate concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C, 100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN];B%=32% -62%;8 min) to give the title compound (15.2 mg, yield 9.90%, brown solid). LC-MS (ESI) m/z 406.0[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.31(s,1H),8.41(d,J=8.11Hz,1H),7.82-7.88(m,1H),7.75-7.80(m,1H),7.58(d,J=5.70Hz,1H),7.45(d,J=8.11Hz,1H),7.40(d,J=8.99Hz,1H),6.89-6.98(m,2H),5.01(s,1H),4.18-4.29(m,1H),2.45-2.49(m,2H),2.12-2.21(m,2H),1.34(s,3H)。
Example 43:2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) -5,6,7, 8-tetrahydrophthalazine-1- Phenyl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (1 s,3 s) -3- ((4-chloro-5, 6,7, 8-tetrahydrophthalazin-1-yl) amino) -1-methylcyclobutanol
To a solution of 1, 4-dichloro-5, 6,7, 8-tetrahydrophthalazine (50.0 mg, 246. Mu. Mol) in DMSO (1.00 mL) at 0deg.C was added cesium fluoride (74.8 mg, 493. Mu. Mol) and 3-amino-1-methyl-cyclobutanol (29.9 mg, 217. Mu. Mol, HCl) in this order, and the resulting reaction solution was stirred at 100deg.C for 2 hours. The reaction mixture was cooled to room temperature, diluted with water (5.00 mL), extracted with EA (5.00 mL. Times.3), and the organic phases were combined, washed with saturated brine, and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=0/1) gave the objective compound (200 mg, yield 30.3%, pale yellow solid). LC-MS (ESI) m/z 268.2[ M+H ]] +
Step 2: synthesis of 2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) -5,6,7, 8-tetrahydrophthalazin-1-yl) -5- (trifluoromethyl) phenol
To a mixed solution of (1 s,3 s) -3- ((4-chloro-5, 6,7, 8-tetrahydrophthalazin-1-yl) amino) -1-methylcyclobutanol (100 mg, 373. Mu. Mol) dioxane (3 mL) and water (0.3 mL) at 25℃was added successively [ 2-hydroxy-4- (trifluoromethyl) phenyl ] ]Boric acid (92.3 mg, 448. Mu. Mol) PdCl 2 (dppf) (27.3 mg, 37.4. Mu. Mol) and anhydrous K 3 PO 4 (238 mg,1.12 mmol) and the resulting reaction mixture was stirred at 100℃for 12 hours.The reaction mixture was cooled to room temperature, filtered, and the filtrate was diluted with EA (5.00 mL) and then washed with water (5.00 mL. Times.3) and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column: watersXbridge BEH C18100. Times.30 mm. Times.10 um; mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -55%;8 min) to give the title compound (43.8 mg, yield 29.8% as yellow solid). LC-MS (ESI) m/z 394.2[ M+H] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.38(s,1H),7.34(d,J=7.70Hz,1H),7.21(br d,J=8.07Hz,1H),7.18(s,1H),6.15(br d,J=6.23Hz,1H),4.93(s,1H),4.03-4.16(m,1H),2.36-2.44(m,4H),2.31(br t,J=6.36Hz,2H),2.03-2.12(m,2H),1.70-1.79(m,2H),1.60(br d,J=5.38Hz,2H),1.30(s,3H)。
Example 44:2- (8- ((1-ethylazetidin-3-yl) amino) pyrido [2, 3-d)]Pyridazin-5-yl) -5- (trifluoromethyl) phenol
Step 1 Synthesis of 3- ((5-chloropyrido [2,3-d ] pyridazin-8-yl) amino) azetidine-1-carboxylic acid tert-butyl ester
CsF (2.28 g,15.0 mmol) was added to 5, 8-dichloropyrido [2,3-d ]]Pyridazine (1.00 g,5.00 mmol) and tert-butyl 3-aminoazetidine-1-carboxylate (861 mg,5.00 mmol) in DMSO (10.0 mL) and the reaction mixture stirred at 130℃for 12 h. After the reaction mixture was cooled to room temperature, it was diluted with water (20.0 mL), extracted with EA (10.0 mL. Times.3), the organic phases were combined and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=20/1 to 1/1) gave the objective compound (0.350 g, yield 20.8%, yellow solid). LC-MS (ESI) m/z 280.0[ M-56+H ] +1 HNMR(400MHz,CDCl 3 -d)δppm 9.13-9.01(m,1H),8.46-8.26(m,1H),7.83(dd,J=4.5,8.2Hz,1H),6.91-6.64(m,1H),5.01-4.87(m,1H),4.43(t,J=8.3Hz,2H),3.92(td,J=4.6,9.2Hz,2H),1.43(s,9H)。
Step 2 Synthesis of 3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) azetidine-1-carboxylic acid tert-butyl ester
XPhos Pd G3 (75.6 mg, 89.34. Mu. Mol) was added to 3- ((5-chloropyrido [2, 3-d) under nitrogen]Pyridazin-8-yl) amino-azetidine-1-carboxylic acid tert-butyl ester (300 mg, 893. Mu. Mol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (275 mg,1.34 mmol) and K 2 CO 3 In a mixture of (370 mg,2.68 mmol) of dioxane (3.00 mL)/water (0.300 mL), the reaction mixture was stirred at 90℃for 12 hours. After the reaction mixture was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=20/1 to 1/1) to give the objective compound (100 mg, yield 24.2%, yellow solid). LC-MS (ESI) m/z 462.2[ M+H ]] +
Step 3 Synthesis of 2- (8- (azetidin-3-ylamino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol hydrochloride
HCl-dioxane (4.00M, 0.600 mL) was added dropwise to 3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2, 3-d) at 0deg.C]Pyridazin-8-yl) amino) azetidine-1-carboxylic acid tert-butyl ester (120 mg, 260. Mu. Mol) in DCM (1.00 mL) and the reaction mixture was stirred at 20℃for 12 h. The reaction mixture was concentrated under reduced pressure to give the title compound (90.0 mg, yield 95.7% as a yellow solid). LC-MS (ESI) m/z 362.1[ M+H ] ] +
Step 4 Synthesis of 2- (8- ((1-ethylazetidin-3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
At 0 ℃, naBH is added 3 CN (13.9 mg, 221. Mu. Mol) and glacial AcOH (66.4. Mu.g, 1.11. Mu. Mol) were added to 2- (8- (azetidin-3-ylamino) pyrido [2, 3-d)]Pyridazin-5-yl) -5- (trifluoromethyl) phenol hydrochloride (40.0 mg, 110. Mu. Mol) in EtOH (1.00 mL) and then a solution of formaldehyde in THF (5.00M, 22.1. Mu.L) was added dropwise and the reaction mixture stirred at 0deg.C for 1 hour. The reaction mixture was filtered and purified by Pre-HPLC (column: phenomenex Luna 80X 30mm 3um; mobile phase: [ A-water (TFA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 10% -40%;8 min) separating the target component, and subjecting to Pre-HPLC (column Waters Xbridge BEH C18 100×30mm×10um; mobile phase [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 15% -40%;8 min) purification to give the title compound(1.87 mg, yield 4.34%, yellow solid). LC-MS (ESI) m/z 390.0[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.89(dd,J=4.38,1.38Hz,1H),7.69(dd,J=8.25,4.50Hz,1H),7.57(dd,J=8.25,1.25Hz,1H),7.51(d,J=8.00Hz,1H),7.32-7.26(m,2H),4.55-4.44(m,1H),4.40(br t,J=10.8Hz,1H),4.10(br t,J=9.57Hz,1H),3.15(br d,J=4.75Hz,1H),3.09(br d,J=7.25Hz,1H),2.96(q,J=6.92Hz,2H),1.20(t,J=7.19Hz,3H)。
Example 45: (R) -2- (8- ((1- (tetrahydro-2H-pyran-4-yl) piperidin-3-yl) amino) pyrido [2,3-d]Pyridazin-5-yl) -5- (trifluoroMethyl) phenol
Step 1: synthesis of (R) -3- ((5-chloropyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
5, 8-dichloropyrido [2,3-d ]]A solution of pyridazine (1.80 g,9.00 mmol) and tert-butyl (R) -3-aminopiperidine-1-carboxylate (1.98 g,9.90 mmol) in dry EtOH (50.0 mL) was stirred at 70℃for 12 h. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=1/1) to give the objective compound (540 mg, yield 16.5%, yellow solid). LC-MS (ESI) m/z 364.3[ M+H ] ] +
Step 2: synthesis of tert-butyl (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylate
(R) -3- ((5-chloropyrido [2, 3-d) at 25 DEG C]Pyridazin-8-yl) amino piperidine-1-carboxylic acid tert-butyl ester (470 mg,1.29 mol), 2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (399 mg,1.94 mmol) and K 2 CO 3 (535 mg,3.88 mmol) was dissolved in a mixed solution of dioxane (50.0 ml)/water (0.50 ml), and under nitrogen protection, (2-dicyclohexylphosphine-2, 4, 6-triisopropyl-1, 1-biphenyl) [2- (2-amino-1, 1-biphenyl) was added]Palladium methanesulfonate (109 mg,129 μmol) and the resulting reaction mixture was stirred under nitrogen at 90 ℃ for 12 hours. The reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel chromatography (DCM/meoh=20/1) to give the title compoundThe title compound (330 mg, yield 52.1% as yellow solid). LC-MS (ESI) m/z 490.3[ M+H ]] +
Step 3: synthesis of (R) -2- (8- (piperidin-3-ylamino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
At 0deg.C, to (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2, 3-d)]To a solution of t-butyl pyridazin-8-yl) amino-piperidine-1-carboxylate (310 mg, 633. Mu. Mol) in anhydrous DCM (15.0 mL) was added dropwise a solution of HCl-dioxane (4M, 3.00 mL), and the resulting reaction solution was stirred at 25℃for 12 hours. The reaction solution was concentrated under reduced pressure to give the objective compound (220 mg, yield 89.2%, yellow solid) which was used directly in the next reaction without further purification. LC-MS (ESI) m/z 390.2[ M+H ] ] +
Step 4: synthesis of (R) -2- (8- ((1- (tetrahydro-2H-pyran-4-yl) piperidin-3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
At 0 ℃, to (R) -2- (8- (piperidin-3-ylamino) pyrido [2, 3-d)]To a solution of pyridazin-5-yl) -5- (trifluoromethyl) phenol (150 mg, 385. Mu. Mol) in anhydrous MeOH (10.0 mL) was added NaBH successively 3 CN (72.6 mg,1.16 mmol) and ice AcOH (2.31 mg, 38.5. Mu. Mol), after stirring for 10 minutes, tetrahydropyran-4-one (192 mg,1.93 mmol) was added dropwise and the resulting reaction mixture was stirred at 0℃for 2 hours. The reaction mixture was filtered, the filtrate concentrated under reduced pressure and the residue was purified by Prep-HPLC (column: phenomenex Luna C, 75X 30mm X3 um; mobile phase: [ A-water (FA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 1% -50%;8 min) to give the title compound (2.62 mg, yield 1.44%, white solid). LC-MS (ESI) m/z 474.2[ M+H ]] +1 HNMR(400MHz,METHANOL-d 4 )δppm 9.11(dd,J=4.38,1.50Hz,1H),8.03(dd,J=8.32,1.56Hz,1H),7.83(dd,J=8.38,4.38Hz,1H),7.61(d,J=7.75Hz,1H),7.34(d,J=8.13Hz,1H),7.28(s,1H),4.48-4.59(m,1H),3.97-4.12(m,2H),3.45(br t,J=10.94Hz,3H),2.62-2.98(m,4H),1.61-2.13(m,8H)。
Example 46: (R) -2- (8- ((1- ((tetrahydro-2H-pyran-4-yl) methyl) piperidin-3-yl) amino) pyrido [2,3-d]Pyridazin-5-yl) -5-(trifluoromethyl) phenol
Step 1: synthesis of (R) -2- (8- ((1- ((tetrahydro-2H-pyran-4-yl) methyl) piperidin-3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
(R) -2- (8- (piperidin-3-ylamino) pyrido [2, 3-d) ]Pyridazin-5-yl) -5- (trifluoromethyl) phenol (100 mg, 257. Mu. Mol), tetrahydro-2H-pyran-4-carbaldehyde (147 mg,1.28 mmol) and a solution of ice AcOH (1.54 mg, 25.7. Mu. Mol) in dry EtOH (10.0 mL) were stirred at room temperature for 10 minutes, then NaBH was added at 0deg.C 3 The resulting reaction mixture of CN (48.4 mg, 770. Mu. Mol) was stirred at 0℃for 2 hours. The reaction mixture was filtered, the filtrate concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C100 x 30mm x 10um; mobile phase: [ a-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 40% -70%;8 min) to give the title compound (3.72 mg, yield 2.97%, yellow solid). LC-MS (ESI) m/z 488.3[ M+H ]] +1 HNMR(400MHz,METHANOL-d 4 )δppm 9.06(dd,J=4.49,1.64Hz,1H),8.00(dd,J=8.22,1.64Hz,1H),7.80(dd,J=8.33,4.17Hz,1H),7.58(d,J=8.11Hz,1H),7.30(d,J=8.55Hz,1H),7.25(s,1H),4.47(dd,J=5.48,3.51Hz,1H),3.89-3.98(m,3H),3.38-3.52(m,3H),3.34-3.38(m,1H),2.44-2.89(m,1H),2.25-2.32(m,2H),1.76-1.92(m,6H),1.63-1.73(m,1H),1.23-1.34(m,2H)。
Examples 47 and 48:2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2, 3-d)]Pyridazin-7-yl) -5- (trifluormethyl)Phenyl) phenols and 2- (7- (((1)s,3s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2, 3-d]pyridazin-4-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (1 s,3 s) -3- ((7-chlorothieno [2,3-d ] pyridazin-4-yl) amino) -1-methylcyclobutanol and (1 s,3 s) -3- ((4-chlorothieno [2,3-d ] pyridazin-7-yl) amino) -1-methylcyclobutanol
At 25 ℃,4, 7-dichloro-thieno [2,3-d ]]To a mixture of pyridazine (1.00 g,4.88 mmol) and cis-3-amino-1-methyl-cyclobutanol (672 mg,4.88mmol, HCl) in DMSO (20.0 mL) was added CsF (1.48 g,9.76 mmol). The resulting reaction mixture was stirred at 100℃for 12 hours. Dilute with EA (100 mL), then wash with water (30.0 mL x 3), then extract the aqueous phase with EA (30.0 mL x 6), combine the organic phases with anhydrous Na 2 SO 4 Drying, filtering, concentrating the filtrate under reduced pressure to obtain (1 s,3 s) -3- ((7-chlorothiophene [2, 3-d)]Pyridazin-4-yl) amino) -1-methylcyclobutanol and (1 s,3 s) -3- ((4-chlorothieno [2, 3-d)]Pyridazin-7-yl) amino) -1-methylcyclobutanol (1.40 g, trude, yellow solid) was used directly in the next reaction. LC-MS (ESI) m/z 270.1[ M+H ]] +1 HNMR(400MHz,DMSO-d 6 )δppm 8.21(d,J=5.25Hz,1H),8.17(d,J=5.25Hz,1H),7.95(d,J=5.38Hz,1H),7.70-7.81(m,2H),7.49(d,J=5.25Hz,1H),5.02(br,s,2H),4.11(dq,J=14.76,7.55Hz,2H),2.38-2.48(m,4H),2.05-2.17(m,4H),1.30(d,J=2.88Hz,6H)。
Step 2: synthesis of 2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2,3-d ] pyridazin-7-yl) -5- (trifluoromethyl) phenol and 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2,3-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
To a mixed solution of the mixture (300 mg,1.11 mmol) obtained in the previous step and (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (343 mg,1.67 mmol) of dioxane (20.0 mL)/water (2.00 mL) under the protection of nitrogen at 25℃were added PdCl in sequence 2 (dppf) (81.4 mg, 111. Mu. Mol) and Anhydrous K 3 PO 4 (708 mg,3.34 mmol) was replaced with nitrogen three times and the resulting reaction mixture was stirred at 100deg.C for 12 hours. The reaction mixture was cooled to 25 ℃, filtered, the filtrate diluted with EA (50.0 mL), then washed with water (15.0 ml×3), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column: phenomenex Luna C18, 75X 30mm X3 um; mobile phase: [ A-Water (FA)/B-ACN) ]The method comprises the steps of carrying out a first treatment on the surface of the B% = 10% -40%;8 min) to give the title compound 2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2, 3-d) in this order]Pyridazin-7-yl) -5- (trifluoromethyl) phenol (108 mg, yield 8.95%, yellow solid), LC-MS (ESI) m/z 396.0[ M+H ]] +1 HNMR(400MHz,DMSO-d 6 ) Delta ppm 13.33 (br s, 1H), 8.25 (d, j=5.36 hz, 1H), 8.03 (d, j=5.36 hz, 1H), 7.99 (d, j=8.23 hz, 1H), 7.81 (br d, j=6.20 hz, 1H), 7.35 (br d, j=8.11 hz, 1H), 7.30 (s, 1H), 5.04 (br s, 1H), 4.19-4.31 (m, 1H), 2.44-2.49 (m, 2H), 2.11-2.20 (m, 2H), 1.34 (s, 3H); and the title compound 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2, 3-d)]Pyridazin-4-yl) -5- (trifluoromethyl) phenol (61.9 mg, yield 27.2%, yellow solid). LC-MS (ESI) m/z 396.0[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 11.62(br s,1H),8.10(br d,J=5.25Hz,1H),7.76(br d,J=8.23Hz,1H),7.64(br d,J=5.48Hz,1H),7.44(br d,J=5.13Hz,1H),7.29(br s,2H),4.99(br s,1H),4.17-4.33(m,1H),2.45-2.48(m,2H),2.17(br t,J=9.12Hz,2H),1.33(s,3H)。
Examples 49 and 50:2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2, 3-d)]Pyridazine (Da) Oxazin-7-yl) -5- (trifluoromethyl) phenol and 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) thieno [2, 3-d]pyridazin-4-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of 4, 7-dichlorofuro [2,3-d ] pyridazine
At 25 ℃, furo [2,3-d ]]To a solution of pyridazine-4, 7-diol (1.00 g,6.57 mmol) in ACN (10.0 mL) was added dropwise pyridine (1.04 g,13.2 mmol) and POCl in this order 3 (5.04 g,32.9 mmol) and the reaction mixture was stirred at 80℃for 12 hours. The reaction mixture was cooled to room temperature, poured into ice water (10.0 mL), extracted with EA (30.0 mL. Times.3), the organic phases combined, and dried over anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (1.18 g, yield 94.9%, light brown solid) which was used directly in the next reaction. LC-MS (ESI) m/z 189.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 8.66(d,J=2.08Hz,1H),7.42(d,J=2.08Hz,1H)。
Step 2: synthesis of (1 s,3 s) -3- ((7-chlorofuro [2,3-d ] pyridazin-4-yl) amino) -1-methylcyclobutanol and (1 s,3 s) -3- ((4-chlorofuro [2,3-d ] pyridazin-7-yl) amino) -1-methylcyclobutanol
At 25 ℃, 4, 7-dichloro-furo [2,3-d]To a solution of pyridazine (500 mg,2.65 mmol) in DMSO (5.00 mL) was added cesium fluoride (1.21 g,7.95 mmol) and 3-amino-1-methyl-cyclobutanol (322 mg,3.18 mmol). The resulting reaction mixture was stirred at 100℃for 12 hours. The reaction mixture was cooled to room temperature, diluted with water (5.00 mL), extracted with EA (5.00 mL. Times.3), the organic phases combined, and dried over anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave a mixture of the target compound (350 mg, crude, dark brown solid) which was used directly in the next reaction. LC-MS (ESI) m/z 254.1[ M+H ]] +
Step 3: synthesis of 2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) furo [2,3-d ] pyridazin-7-yl) -5- (trifluoromethyl) phenol and 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) furo [2,3-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
To a mixed solution of dioxane (3.00 mL) and water (0.300 mL) of the mixture (150 mg, 591. Mu. Mol) obtained in step 2 was added [ 2-hydroxy-4- (trifluoromethyl) phenyl ] at 25 ℃]Boric acid (146 mg, 710. Mu. Mol), XPhos Pd G3 (50.1 mg, 59.1. Mu. Mol) and anhydrous K 2 CO 3 (248 mg,1.77 mmol). The resulting reaction mixture was stirred at 100℃for 12 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was diluted with EA (10.0 mL) and then washed with water (5.00 mL. Times.3) and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure and residue passing through Prep-HPLC (column: phenomenex Luna C18, 75X 30mm X3 um; mobile phase: [ A-Water (FA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -60%;8 min), and collecting the desired components, and subjecting to Prep-HPLC (column Waters Xbridge BEH C, 100×30mm×10um; mobile phase [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 35% -65%;8 min) to give the title compound 2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) furo [2, 3-d) in sequence]Pyridazin-7-yl) -5- (trifluoromethyl) phenol (14.5 mg, yield 6.46%, pale yellow solid), [ LC-MS (ESI) m/z:380.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 14.70(s,1H),8.49(d,J=8.23Hz,1H),8.40(d,J=2.03Hz,1H),7.97(d,J=6.20Hz,1H),7.40(d,J=2.03Hz,1H),7.34(d,J=8.34Hz,1H),7.28(s,1H),5.06(s,1H),4.16-4.29(m,1H),2.46-2.48(m,2H),2.08-2.16(m,2H),1.33(s,3H)。]And the title compound 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) furo [2, 3-d)]Pyridazin-4-yl) -5- (trifluoromethyl) phenol (22.0 mg,9.81%, pale yellow solid). LC-MS (ESI) m/z 380.1[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δppm 14.27(br s,1H),8.49(br s,1H),8.15(br d,J=7.39Hz,1H),8.09(br s,1H),7.59(br s,1H),7.22-7.31(m,2H),5.00(br s,1H),4.14-4.29(m,1H),2.43-2.47(m,2H),2.16-2.18(m,2H),1.32(br s,3H)。
Example 51:2-(4-([1,2,4]triazolo [4,3-a ]]Pyridin-6-ylamino) phthalazin-1-yl) -5- (trifluormethyl alcohol Radical) phenol
Step 1 Synthesis of N- ([ 1,2,4] triazolo [4,3-a ] pyridin-6-yl) -4-chlorophthalazin-1-amine
NaH (80.3 mg,2.01mmol,60.0% purity) was added to [1,2,4] at 0deg.C]Triazolo [4,3-a ]]To a solution of pyridin-6-amine (134 mg,1.00 mmol) in DMF (1.00 mL) was added dropwise a solution of 1, 4-dichlorophthalazine (200 mg,1.00 mmol) in DMF (0.500 mL) after stirring for 1 hour, and the reaction mixture was stirred at 60℃for 11 hours. After the reaction mixture was cooled to room temperature, ice water (10.0 mL) was added thereto, followed by stirring, filtration and drying of the cake under reduced pressure to give the objective compound (220 mg, yield 73.7%, yellow solid). LC-MS (ESI) m/z 297.0[ M+H ]] +
Step 2 Synthesis of 2- (4- ([ 1,2,4] triazolo [4,3-a ] pyridin-6-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
XPhos Pd G3 (34.2 mg, 40.4. Mu. Mol) was added to N- ([ 1,2, 4) under nitrogen]Triazolo [4,3-a ]]Pyridin-6-yl) -4-chlorophthalazin-1-amine (120 mg, 404. Mu. Mol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (124 mg, 606. Mu. Mol) and K 2 CO 3 (167 mg,1.21 mmol) of dioxane (1.00 mL)/water (0.100 mL) and the resulting reaction mixture was stirred at 90℃for 12 hours. After the reaction mixture had cooled to room temperature Filtration and filtration of the filtrate by Pre-HPLC (column: phenomnex C1880. Times.40 mm. Times.3 um; mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 25% -55%;8 min) to give the title compound (22.1 mg, yield 12.9%, yellow solid). LC-MS (ESI) m/z 423.1[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 10.42(s,1H),10.06(d,J=1.38Hz,1H),9.63(s,1H),8.71(d,J=8.38Hz,1H),8.45(s,1H),8.14-8.02(m,2H),7.96-7.87(m,2H),7.59(d,J=8.00Hz,2H),7.37-7.30(m,2H)。
Example 54:2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) -2, 3-dihydrofuro [2, 3-d)]Pyridazin-4-yl) -5- (trifluoroMethyl) phenol
Step 1 Synthesis of 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) -2, 3-dihydrofuro [2,3-d ] pyridazin-4-yl) -5- (trifluoromethyl) phenol
Palladium hydroxide (0.05 g,20% purity) was added to 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) furo [2, 3-d) under argon]Pyridazin-4-yl) -5- (trifluoromethyl) phenol (12.0 mg, 31.6. Mu. Mol) in MeOH (10.0 mL), the resulting reaction mixture was reacted with H 2 (15 Psi) and stirring at 25℃for 3 hours. The reaction mixture was filtered through celite, washed with MeOH (10.0 mL), the filtrate concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C100 x 30mm x 10um; mobile phase: [ a-water (NH) 3 H 2 O+NH 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 40% -70%;8 min) to give the title compound (1.31 mg, yield 10.8% as a pale yellow solid). LC-MS (ESI) m/z 382.2[ M+H ] ] +1 HNMR(400MHz,MeOH-d 4 )δppm 8.39(d,J=8.11Hz,1H),7.16(s,1H),7.13(d,J=8.55Hz,1H),4.93-4.89(m,2H),4.21-4.06(m,1H),3.18(t,J=9.21Hz,2H),2.63-2.55(m,2H),2.14(td,J=9.04,2.52Hz,2H),1.42(s,3H)。
Example 55:2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) -2, 3-dihydrofuro [2, 3-d)]Pyridazin-7-yl) -5- (trifluoroMethyl group)Phenol (P)
Step 1 Synthesis of 2- (4- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) -2, 3-dihydrofuro [2,3-d ] pyridazin-7-yl) -5- (trifluoromethyl)
Palladium hydroxide (0.05 g) was added to 2- (7- (((1 s,3 s) -3-hydroxy-3-methylcyclobutyl) amino) furo [2, 3-d) under argon]Pyridazin-4-yl) -5- (trifluoromethyl) phenol (14.4 mg, 37.96. Mu. Mol) in MeOH (10.0 mL), the resulting reaction mixture was reacted with H 2 (15 Psi) and stirring at 25℃for 3 hours. The reaction mixture was filtered through celite, washed with MeOH (10.0 mL), the filtrate concentrated under reduced pressure and the residue was purified by Prep-HPLC (column Waters Xbridge BEH C100 x 30mm x 10um; mobile phase: [ a-water (NH) 3 H 2 O+NH 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 35% -65%;8 min) to give the title compound (7.50 mg, yield 51.8%, pale yellow solid). LC-MS (ESI) m/z 382.2[ M+H ]] +1 HNMR(400MHz,MeOH-d 4 )δppm 8.40(d,J=8.55Hz,1H),7.16(s,1H),7.11-7.15(m,1H),4.93-4.89(m,2H),4.15(t,J=7.78Hz,1H),3.19(t,J=9.32Hz,2H),2.62-2.55(m,2H),2.18-2.08(m,2H),1.42(s,3H)。
Example 56:2- (4- (pyrimidin-4-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of 4-chloro-N- (pyrimidin-4-yl) phthalazin-1-amines
Sodium hydride (301 mg,7.54mmol,60% purity) was added to pyrimidine-4-amine (356 mg,3.77 mmol) in DMSO (5.00 mL) at 0deg.C, and after stirring at 25deg.C for 1 hour, 1, 4-dichlorophthalazine (500 mg,2.51 mmol) was added and the reaction mixture stirred at 60deg.C for 12 hours. After the reaction mixture had cooled to room temperature, it was quenched with saturated NH 4 Cl solution (20.0 mL) was quenched, EA (5.00 mL. Times.3) extracted, and the organic phases were combined and quenched with saturated brine (5.00)mL) washing, anhydrous Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (450 mg, yield 69.5% as pale yellow solid) which was used directly in the next reaction. LC-MS (ESI) m/z 258.1[ M+H ]] +
Step 2: synthesis of 2- (4- (pyrimidin-4-ylamino) phthalazin-1-yl) -5- (trifluoromethyl) phenol
XPhos Pd G3 (49.3 mg, 58.2. Mu. Mol) was added to 4-chloro-N- (pyrimidin-4-yl) phthalazin-1-amine (150 mg, 582. Mu. Mol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (144 mg, 699. Mu. Mol) and anhydrous K under nitrogen 2 CO 3 (241 mg,1.75 mmol) of dioxane (2.00 mL)/water (0.20 mL), and the resulting reaction mixture was stirred at 100deg.C for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (5.00 mL), extracted with EA (5.00 mL. Times.3), the organic phases were combined, washed with saturated brine (5.00 mL), and dried over Na 2 SO 4 Drying, filtration, concentration under reduced pressure and the residue was purified by two Prep-HPLC (column: phenomnex C18 75X 30mm X3 um; mobile phase: [ A-water (FA)/B-ACN)];B%=20%-50%;8min)&(column: waters Xbridge BEH C18 100 x 30mm x 10um; mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 40% -70%;8 min) to give the title compound (19.3 mg, yield 8.66%, yellow solid). LC-MS (ESI) m/z 384.1[ M+H ] ] +1 H NMR(400MHz,MeOH-d 4 )δppm 8.70(br s,1H),8.52-8.37(m,2H),8.11(br s,2H),7.94(br s,1H),7.74-7.65(m,2H),7.65-7.60(m,2H)。
Example 61:(R) -2- (8- ([ 1,4' -bipiperidine)]3-ylamino) pyrido [2,3 ]d]Pyridazin-5-yl) -5- (trifluoromethyl) phenol
Reference example 62 was synthesized. LC-MS (ESI) m/z 390.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm9.17-9.07(m,1H),8.89(br d,J=8.1Hz,1H),8.35(br s,1H),8.05(d,J=8.2Hz,1H),7.90(dd,J=4.3,8.3Hz,1H),7.55(br d,J=5.2Hz,1H),7.26-7.20(m,2H),4.42-4.30(m,1H),3.31-3.16(m,3H),2.86-2.72(m,3H),2.69-2.55(m,1H),2.24-2.10(m,2H),2.09-1.97(m,1H),1.89-1.75(m,3H),1.72-1.44(m,4H)。
Example 62: (R) -2- (8- ((1 '-methyl- [1,4' -bipiperidine)]-3-yl) amino) pyrido [2,3-d]Pyridazin-5-yl) -5- (trifluoromethyl) benzenePhenol
Step 1: synthesis of (R) -3- ((5-chloropyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
(R) -3-aminopiperidine-1-carboxylic acid tert-butyl ester (751 mg,3.75 mmol) and CsF (1.14 g,7.50 mmol) were added sequentially to 5, 8-dichloropyrido [2,3-d ] at room temperature]Pyridazine (500 mg,2.50 mmol) in DMSO (10.0 mL) and the resulting reaction mixture was stirred at 100deg.C for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (20.0 mL), extracted with EA (10.0 mL. Times.3), the organic phases were combined, washed with saturated brine (5.00 mL), and dried over Na 2 SO 4 Drying, filtering, concentrating under reduced pressure to obtain the target compound (800 mg,87.9%, yellow solid) LC-MS (ESI) m/z 364.1[ M+H)] +
Step 2: synthesis of tert-butyl (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylate
RuPhos Pd G3 (115 mg, 137. Mu. Mol) was added to (R) -3- ((5-chloropyrido [2, 3-d) under nitrogen ]Pyridazin-8-yl) amino piperidine-1-carboxylic acid tert-butyl ester (500 mg,1.37 mmol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (283 mg,1.37 mmol) and K 2 CO 3 (570 mg,4.12 mmol) of dioxane (10.0 mL)/water (1.00 mL) and the reaction mixture was stirred at 90℃for 12 hours. After the reaction mixture was cooled to room temperature, it was diluted with water (10.0 mL), extracted with EA (10.0 mL. Times.3), the organic phases were combined, washed with saturated brine (5.00 mL), and dried over Na 2 SO 4 Drying, filtration and concentration under reduced pressure gave the title compound (300 mg, yield 44.6%, orange solid). LC-MS (ESI) m/z 490.2[ M+H ]] +
Step 3: synthesis of (R) -2- (8- (piperidin-3-ylamino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
A solution of HCl-dioxane (2.00 mL) was added dropwise to (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2, 3-d) at room temperature]Pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (200 mg, 409. Mu. Mol) in DCM (10.0 mL) and the reaction mixture was stirred at room temperature for 12 h. The reaction mixture was concentrated directly under reduced pressure to give the target compound (260 mg, eude, pale yellow solid) which was used directly in the next reaction. LC-MS (ESI) m/z 390.2[ M+H ]] +
Step 4: synthesis of (R) -2- (8- ((1 '-methyl- [1,4' -bipiperidin ] -3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
At 0 ℃, naBH is added 3 CN (48.4 mg, 770. Mu. Mol) was added to AcOH (154 mg,2.57 mmol) and (R) -2- (8- (piperidin-3-ylamino) pyrido [2, 3-d)]To a solution of pyridazin-5-yl) -5- (trifluoromethyl) phenol (100 mg, 256.82. Mu. Mol) in MeOH (3.00 mL) was added 1-methylpiperidin-4-one (145.3 mg,1.28 mmol) after stirring for 10 min, and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated directly under reduced pressure and the residue was purified by Prep-HPLC (column: column: waters Xbridge BEH C, 18: 18 100*30mm*10um;mobile phase: [ A-water (NH) 4 HCO 3 )/B-ACN]The method comprises the steps of carrying out a first treatment on the surface of the B% = 30% -60%;8 min) to give the title compound (21.4 mg, yield 17.2%, yellow solid). LC-MS (ESI) m/z 487.3[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 11.65(br s,1H),9.13(dd,J=1.4,4.3Hz,1H),8.88(dd,J=1.4,8.5Hz,1H),8.14(d,J=8.0Hz,1H),7.91(dd,J=4.3,8.4Hz,1H),7.49(d,J=7.6Hz,1H),7.25(d,J=8.3Hz,1H),7.21(s,1H),4.45-4.31(m,1H),3.24-3.15(m,1H),2.87-2.73(m,3H),2.31-2.21(m,1H),2.17(br t,J=10.0Hz,2H),2.11(s,3H),2.06-1.98(m,1H),1.89-1.73(m,3H),1.67(br d,J=10.6Hz,2H),1.60-1.41(m,4H)。
Example 63:(R) -2- (8- ((1 '- (methylsulfonyl) - [1,4' -bipiperidine)]-3-yl) amino) pyrido [2,3- ] d]Pyridazin-5-yl) -5- (trifluoromethyl) phenol
Step 1: synthesis of (R) -3- ((5-chloropyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester
At room temperature, 5, 8-dichloropyrido [2,3-d ]]Pyridazine (2.00 g,10.0 mmol) was added to a solution of tert-butyl (R) -3-aminopiperidine-1-carboxylate (2.20 g,11.0 mmol) and CsF (4.56 g,30.0 mmol) in DMSO (20.0 mL) and the resulting reaction mixture was stirred at 135℃for 12 hours. After the reaction mixture was cooled to room temperature, water (20.0 mL) was added and stirred, extracted with EA (20.0 mL. Times.3), the organic phases were combined and dried over anhydrous Na 2 SO 4 Drying, filtration, concentration under reduced pressure, and separation and purification of the residue by silica gel chromatography (PE/ea=1/1) gave the objective compound (1.95 g, yield 53.6%, brown solid). LC-MS (ESI) m/z 364.2[ M+H ]] +
Step 2: synthesis of tert-butyl (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) piperidine-1-carboxylate
XPhos Pd G3 (209 mg,247 mmol) was added to (R) -3- ((5-chloropyrido [2, 3-d) under nitrogen]Pyridazin-8-yl) amino piperidine-1-carboxylic acid tert-butyl ester (900 mg,2.47 mmol), (2-hydroxy-4- (trifluoromethyl) phenyl) boronic acid (764 mg,3.71 mmol) and K 2 CO 3 (1.03 g,7.42 mmol) of dioxane (20.0 mL)/water (2.00 mL), and the reaction mixture was stirred at 90℃for 12 hours. After the reaction mixture was cooled to room temperature, it was concentrated under reduced pressure, and the residue was purified by silica gel chromatography (PE/ea=1/1) to give the objective compound (1.00 g, yield 82.6%, brown solid). LC-MS (ESI) m/z 490.2[ M+H ]] +
Step 3: synthesis of (R) -2- (8- (piperidin-3-ylamino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol hydrochloride
A solution of HCl-dioxane (4M, 356 mL) was added dropwise to (R) -3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2, 3-d) at 0deg.C]Pyridazin-8-yl) amino) piperidine-1-carboxylic acid tert-butyl ester (700 mg,1.43 mmol) in DCM (10.0 mL) and the reaction mixture stirred at 25℃for 5 h. The reaction mixture was concentrated directly under reduced pressure to give the title compound (500 mg, yield 89.8%, yellow solid) which was used directly for the next reaction And (3) performing a step reaction. LC-MS (ESI) m/z 390.1[ M+H ]] +
Step 4: synthesis of (R) -2- (8- ((1 '- (methylsulfonyl) - [1,4' -bipiperidin ] -3-yl) amino) pyrido [2,3-d ] pyridazin-5-yl) -5- (trifluoromethyl) phenol
AcOH (1.54 mg,25.68mmol,1.47 mL) and NaBH (OAc) were combined at 0deg.C 3 (163 mg,770 mmol) added to (R) -2- (8- (piperidin-3-ylamino) pyrido [2, 3-d)]To a solution of pyridazin-5-yl) -5- (trifluoromethyl) phenol hydrochloride (100 mg,257 mmol) in DCE (5.00 mL) was added 1- (methylsulfonyl) piperidin-4-one (137 mg,770 mmol) after stirring for 10 min, and the reaction mixture was stirred at 25℃for 12 h. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by Prep-HPLC (column: phenomenex Luna C18:18: 18 75*30mm*3um;mobile phase: [ A-water (FA)/B-ACN)]The method comprises the steps of carrying out a first treatment on the surface of the B% = 15% -45%;8 min) to give the title compound (21.1 mg, yield 15.0% as a yellow solid). LC-MS (ESI) m/z 551.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm 9.12(dd,J=1.5,4.4Hz,1H),8.88(dd,J=1.5,8.5Hz,1H),8.33(s,1H),8.11(d,J=7.9Hz,1H),7.91(dd,J=4.4,8.4Hz,1H),7.51(d,J=7.6Hz,1H),7.30-7.14(m,2H),3.24-2.78(m,8H),2.77-2.61(m,3H),2.21(br t,J=10.0Hz,2H),2.08-1.96(m,1H),1.90-1.69(m,3H),1.63-1.40(m,4H)。
Example 64: (R) -N- (4- (3- ((5- (2-hydroxy-4- (trifluoromethyl) phenyl) pyrido [2,3-d ] pyridazin-8-yl) amino) piperidin-1-yl)
Cyclohexyl) methanesulfonamide
Reference example 62 was synthesized. LC-MS (ESI) m/z 565.2[ M+H ]] +1 H NMR(400MHz,DMSO-d 6 )δppm9.13(dd,J=1.5,4.4Hz,1H),8.92-8.85(m,1H),8.18(s,1H),8.12(d,J=8.0Hz,1H),7.92(dd,J=4.4,8.4Hz,1H),7.57-7.48(m,1H),7.25(d,J=8.1Hz,1H),7.22(s,1H),7.04-6.94(m,1H),4.49-4.31(m,1H),3.47-3.40(m,1H),3.28-3.15(m,2H),3.10-2.96(m,1H),2.87(d,J=1.1Hz,3H),2.41-2.30(m,1H),2.28-2.14(m,2H),2.08-1.88(m,2H),1.82-1.18(m,9H)。
Comparative example 1: inzomelid
The MCC7840 is obtained by the method described in WO2016131098A 1.
Biological examples
Unless otherwise indicated, the experimental materials, reagents, procedures and methods used in the following examples of in vitro biological experiments are all available from commercial sources or are readily known or prepared based on the prior art.
Biological example 1: IL-1 beta release inhibition assay of human acute mononuclear leukemia cell THP-1
The activity of the compounds of the invention can be assessed by the following in vitro method. The compounds having formula (I) or pharmaceutically acceptable salts thereof exhibit valuable pharmacological activity, e.g. inhibitory properties against NLRP3 activity as shown in the following test, and are therefore useful in the treatment and prophylaxis associated with NLRP3 inflammatory body activity.
Human acute mononuclear leukemia cells THP-1 (purchased from Nanjac, bai Biotechnology Co., ltd. (ATCC, CBP 60518)) were used to evaluate the activity of NLRP3 inhibitors in inhibiting secretion of IL-1 beta by cells. Inducing THP-1 cells to differentiate into mature macrophages by using PMA (crotyl alcohol-12-tetradecanoate-13 acetate, sigma, P8139-25MG, molecular weight 616.83), then stimulating the differentiated cells by using LPS (lipopolysaccharide, sigma, L4524-10 MG), promoting the transcriptional expression of NLRP3 and IL-1 beta and IL-18, and finally stimulating and activating NLRP3 inflammatory corpuscles by using Nigericin sodium salt (Absin, abs819747-25MG, molecular weight 747), and activating caspase-1. Activated caspase-1 cleaves precursors of IL-1 beta, IL-18 to form mature IL-1 beta, IL-18 and is secreted extracellularly. NLRP3 inhibitor can effectively inhibit the activation of NLRP3 inflammatory corpuscles induced by Nigericin, thereby inhibiting the maturation and secretion of IL-1 beta and IL-18.
Specifically, a 10mM DMSO (Solarbio D8371) solution of the test compound was diluted in a DMSO gradient and moderately diluted with a medium, and the final concentration of DMSO in the test compound solution was <0.2%. At the time of reaction, specific volumes of each concentration (50, 16.7, 5.6, 1.85, 0.62, 0.21, 0.07, 0.02. Mu.M) of the test compound solution were added to the microplate as described below.
To a suspension of 10ml THP-1 cells (cell density 1.5X10) in medium (99% RPMI 1640 (Gibco 11875093) +10% FBS (Gibco 10099141C) +0.05mM beta-mercaptoethanol (Solarbio M8210)) 6 cells/mL) was added with 10 μl of 20 μm PMA solution (prepared by dilution of 20mM PMA in DMSO medium), mixed well, and plated in 96-well plates at 0.1 mL/well; at 37℃with 5% CO 2 Incubate in incubator for 24h. All medium in the wells was discarded, taking care not to interfere with cells adhering to the bottom of the wells. Mu.l of LPS at a concentration of 100ng/ml was added to each well (from 2mg/ml LPS at 1xPBS (137mM NaCl,2.7mM KCl,10mM Na) 2 HPO 4 ,1.76mM K 2 HPO 4 The solution in pH 7.4) was diluted with medium) and incubated at 37℃for 3h. All medium in the wells was discarded, taking care not to interfere with cells adhering to the bottom of the wells. Mu.l of serum-free medium and 1. Mu.l of test compound solutions of different concentrations (50, 16.7, 5.6, 1.85, 0.62, 0.21, 0.07, 0.02. Mu.M) were added to each well at 37℃with 5% CO 2 Incubate in incubator for 0.5h. Then 99. Mu.l of niger of 20. Mu.M concentration (prepared by dilution of 10mM-niger stock solution (DMSO) in culture medium) was added to each well, followed by 1. Mu.l of test compound solutions of 50, 16.7, 5.6, 1.85, 0.62, 0.21, 0.07, 0.02. Mu.M concentration, respectively, at 37℃in 5% CO 2 Incubate in incubator for 1h.
The 96-well plate thus cultured was centrifuged at 1000rpm for 5 minutes at room temperature, 4. Mu.l of the supernatant was diluted 25-fold with a diluent (human IL-1β ELISA kit, absin, abs 510002-96T) in an IL-1β ELISA kit, and then ELISA of IL-1β (human IL-1β ELISA kit, absin, abs 510002-96T) was performed on the 96-well plate, and the IL-1β content was detected by the procedure of ELISA kit instructions.
Statistical data were used with GraphPad software to derive IC50 values for inhibition of IL-1β (%) release.
Table 1 shows that the compounds of the examples of the present invention inhibit IL-1β release with an IC50<500nM, preferably <200nM, more preferably <100nM, even more preferably <50nM, most preferably <20nM. The compounds of most of the examples show a significantly better ability to inhibit the release of IL-1 beta than the comparative compound 1. "+++" means test compound inhibition "+++" "indicates test compound inhibition the IC50 of IL-1 beta release is 20-50 nM," ++ "indicates that the test compound has an IC50 of 50 to 100nM for inhibiting IL-1β release," ++ "indicates that the test compound has an IC50 of 100 to 200nM for inhibiting IL-1β release, and" + "indicates that the test compound has an IC50 of 200 to 500nM for inhibiting IL-1β release.
Table 1: representative Activity of the Compounds of the invention to inhibit IL-1 beta Release
/>
Biological example 2 experiments on the metabolic stability of liver microsomes
Human or CD-1 mouse liver microsome mixtures were purchased from Corning (# 452117) and Xenotech (#M1000), respectively. Liver microsomes were diluted to the appropriate concentration with PBS buffer. mu.L of the test compound (final concentration 1. Mu.M) dissolved in 90% 100mM potassium phosphate buffer (pH 7.4) and 9.9% acetonitrile, 80. Mu.L of liver microsomes (final concentration 0.5 mg/mL) and 10. Mu.L of 100mM potassium phosphate buffer were added to a 96-well plate, and the mixture was placed in a 37℃water bath to pre-incubate for 10 minutes, followed by adding 10. Mu.L of NADPH (Vendor: BONTAC, # BT 04) regeneration system to each well, continuing the incubation at 37℃and starting the timer. When the reaction was completed for 5, 10, 20, 30, and 60 minutes, 300. Mu.L of a reaction termination solution (acetonitrile containing 200ng/mL of tolbutamide and 200ng/mL of labetalol) was added to the reaction system to terminate the reaction, and the mixture was homogenized and centrifuged. Transferring 100 μl of the reaction solution to a new 96-well plate containing 300 μl of ultrapure water, mixing thoroughly, and LC-MS/MS analyzing the residual compound amount in the reaction solution to fit the residual compoundThe Slope (Slope) was calculated by linear regression of the natural logarithm of the compound (%) with time. Calculation of T using first order kinetic equation 1/2 And CLint (mic) (μL/min/mg), where Ke is the Slope (Slope).
The results show that: representative compounds of the invention have good metabolic stability of liver microsomes and longer t 1/2 And lower clearance rates, thereby allowing the drug to exert pharmacological activity in vivo for an extended period of time, thereby allowing for increased dosing intervals, reduced dosing, better patient compliance and reduced potential for toxic side effects.
TABLE 2 results of human and mouse liver microsome metabolic stability experiments
Bioactivity example 3: pharmacokinetic (PK) assays of the compounds of the invention in mice
The PK assay method for the compounds is as follows: 6 CD-1 mice (Shanghai Ling Biotechnology Co., ltd.) were divided into two groups of 3. One group was given Intravenously (IV) at a dose of 1mg/kg with vehicle 5% DMSO/95% (20% Captisol); the other group was administered by oral (Po) gavage at a dose of 5mg/kg with vehicle 1% HPMC. Each group was bled through the saphenous vein of the lower leg at 0, 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, 24 hours after dosing. About 40. Mu.L of blood was collected into an anticoagulant tube containing EDTA-K2. The blood collection tube was inverted at least 5 times immediately after collection was completed to ensure uniform mixing and then placed on ice. Blood was centrifuged at 4 ℃/8000rpm for 5 minutes at each time point collected to obtain plasma. Another 1.5mL centrifuge tube, labeled compound name, animal number, time point, was taken and plasma was transferred to the tube. Plasma was stored at-80 ℃ until analysis.
The concentration of the compound in the plasma was determined by UPLC-MS/MS method, and the pharmacokinetic parameters were calculated for the obtained data using Phoenix WinNolin 6.4 pharmacokinetic software.
The specific experimental results are shown below, and the results show that the representative compound provided by the invention has better pharmacokinetic absorption and pharmacokinetic advantage.
TABLE 3 in vivo PK results for representative compounds of the invention
The results show that the compounds of the present invention have improved pharmacokinetic properties, with better potency and higher bioavailability being expected.
Bioactivity example 4: hERG current inhibition assay
Culturing Chinese hamster ovary cells (CHO cells, built in Shanghai pharmaceutical institute of China academy of sciences) stably expressing hERG channel in 35mm diameter cell culture dish, placing at 37deg.C, 5% CO 2 Is cultured in an incubator. Passaging was performed every 48 hours at a ratio of 1:5, medium formulation: 90% F12 (Invitrogen, 11765054), 10% fetal bovine serum (Gibco, 10099141C), 100G/mL G418 (Invivogen, NC 9107150) and 100G/mL Hygromycin B (Invitrogen, 10687010).
ExperimentOn the day, the cell culture solution was aspirated, and extracellular fluid (140mM NaCl,5mM KCl,1mM CaCl) 2 ,1.25mM MgCl 2 10mM HEPES and 10mM Glucose,pH7.4) was rinsed once, 0.25% Trypsin-EDTA (Invitrogen, 25200056) solution was added and digested at room temperature for 3-5 minutes. The digestate was aspirated, resuspended in extracellular fluid and the cells were transferred to a dish for electrophysiological recording for later use.
On the day of the experiment, the compounds of the examples were prepared as 10mM stock solution in DMSO, and then 5. Mu.L was added to 4995. Mu.L of extracellular fluid to give the final concentration of 10. Mu.M to be tested. The positive control compound cisapride (Sigma, C4740-10mg, MW 483.96) was first prepared as a 150. Mu.M stock solution in DMSO and 10. Mu.L of the stock solution was added to 4990. Mu.L of extracellular fluid to give the final concentration to be tested of 300nM.
Whole-cell electrophysiological recording process: the prepared suspension of hERG potassium channel expressing CHO cells in a laboratory dish was placed under an inverted microscope and the hERG potassium channel current was recorded using whole cell patch clamp technique. The glass microelectrode is formed by drawing a glass electrode blank (BF 150-86-10, sutter) by a drawing instrument, and pouring an electrode inner liquid (140mM KCl,1mM MgCl) 2 ,1mM CaCl 2 10mM EGTA and 10mM HEPES,pH 7.2) is about 2-5MΩ, and the glass microelectrode can be connected to the patch clamp amplifier by inserting the amplifier probe. The clamp voltage and data recording are controlled and recorded by pClamp 10 software through a computer, the sampling frequency is 10kHz, and the filtering frequency is 2kHz. After whole cell recordings were obtained, cells were clamped at-80 mV, and hERG potassium current was induced (I hERG ) From-80 mV gives a depolarization voltage of 2s to +20mV and repolarization to-50 mV for 1s and back to-80 mV. This voltage stimulus was administered every 10s, and the course of administration was started (DMSO content in final concentration of administration was not more than 0.2%) after the hERG potassium current had stabilized (1 minute), and the effect of the drug was observed after at least 1 minute of administration of the compound. At least 3 cells (n.gtoreq.3) were tested per compound. The recorded data were processed using pClamp 10,GraphPad Prism 8 and Excel software. The extent of inhibition of hERG potassium current (-peak hERG tail current induced at 50 mV) by the 10 μm example compounds was calculated using the following formula:
Inhibition%=[1–(I/Io)]×100%
Wherein Inhibition% represents the percent Inhibition of hERG potassium current by the compound of the example, I and Io represent the magnitude of hERG potassium current after and before dosing, respectively.
TABLE 4 inhibition of hERG potassium channel currents by representative compounds of the invention (%)
Examples Testing concentration Inhibition rate Determination of cell number
5 10μM 21.5% 3
9 10μM 44.9% 3
It will be appreciated by those skilled in the art that the foregoing description is exemplary and illustrative in nature and is intended to illustrate the invention and its preferred embodiments. Through routine experimentation, those skilled in the art will appreciate that obvious modifications and variations can be made without departing from the spirit of the invention. All such modifications are intended to be included within the scope of the following claims. Accordingly, it is intended that the invention be defined not by the above description but by the scope of the following claims and their equivalents.
All publications cited in this specification are herein incorporated by reference.

Claims (21)

1. A compound of formula (I), stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof:
wherein, the liquid crystal display device comprises a liquid crystal display device,
x is-NH- (CH) 2 ) n -or-O-;
ring A is selected from phenyl, 5-6 membered heteroaryl ring, 5-7 membered cycloalkenyl, and 5-7 membered heterocycloalkenyl, each optionally substituted with oxo, halogen, CN, C 1-6 Alkyl-or halogen-substituted C 1-6 Alkyl substitution;
R 1 selected from halogen, CN, C 1-6 Alkyl, C 1-6 Alkoxy, C 1-6 Alkylthio, -SO 3 H、COOH-、-S(O) 2 -C 1-6 Alkyl, -S (O) -C 1-6 Alkyl, -CO-C 1-6 Alkyl and-CO-OC 1-6 Alkyl group, wherein C 1-6 Alkyl, C 1-6 Alkoxy or C 1-6 Alkylthio groups are each independently at each occurrence optionally substituted with halogen;
R 2 selected from H, halogen, CN and C optionally substituted by halogen 1-3 An alkyl group;
R 3 selected from-OH, -SH, -NHSO 2 -R 3a 、-NH-SO-R 3a 、-NH-CO-R 3a 、-CH(R 3b )(SO 2 -R 3a )、-CH(R 3b )(SO-R 3a )、-CH(R 3b )(CO-R 3a ) and-CH (R) 3c ) 2
R 3a Each at each occurrence is independently selected from-C 1-6 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl, - (CH) 2 ) m -3-8 membered heterocycloalkyl, - (CH) 2 ) m -3-8 membered heterocycloalkenyl,-(CH 2 ) m -C 6-10 Aryl and- (CH) 2 ) m -a 5-10 membered heteroaryl, each optionally substituted with a group selected from: halogen, CN, C optionally substituted by halogen 1-6 Alkyl, optionally halogen-substituted C 1-6 Alkoxy and optionally halogen-substituted C 3-6 Cycloalkyl;
R 3b independently at each occurrence selected from H, halogen and C optionally substituted with halogen 1-6 An alkyl group;
R 3c independently at each occurrence selected from halogen, CN and optionally halogen substituted-C 1-6 An alkyl group;
ra, rb are each independently selected from H, halogen, CN, C optionally substituted with halogen 1-6 Alkyl and optionally halogen-substituted C 1-6 An alkoxy group;
R 4 selected from the group consisting of-3-10 membered heterocycloalkyl and-C 3-6 Cycloalkyl groups, each optionally substituted with a group selected from: -OH, halogen, oxo, -CO 2 H、-SO 2 NH 2 C optionally substituted by halogen 1-6 Alkyl, C 1-6 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -3-8 membered heterocycloalkyl, and C in the substituents 3-6 Cycloalkyl and-3-8 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 NH 2 、-SO 2 (C 1-3 Alkyl) or-NH-SO 2 (C 1-3 Alkyl) substitution; and is also provided with
Wherein n is selected from 0, 1 and 2;
m is independently selected from 0, 1 or 2 at each occurrence.
2. The compound of claim 1, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, wherein X is-NH- (CH) 2 ) n -, and n is 0 or 1.
3. The compound of claim 1, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, wherein a is a benzene ring or cyclohexenyl.
4. The compound of claim 1, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, wherein ring a is a 5-6 membered heteroaryl ring containing 1 or 2 heteroatoms selected from O, N and S, preferably
5. The compound of any one of claims 1-4, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, wherein R 1 Selected from halogen, optionally halogen-substituted C 1-3 Alkyl and optionally halogen-substituted C 1-3 An alkoxy group; preferably R 1 Selected from halogen, C substituted by halogen 1-3 Alkyl and C substituted by halogen 1-3 An alkoxy group.
6. The compound of any one of claims 1-5, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, wherein R 2 H.
7. The compound of any one of claims 1-6, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, wherein R 3 Selected from-OH, -NHSO 2 -R 3a and-CH (R) 3c ) 2 Wherein R is 3a Selected from-C optionally substituted by halogen 1-3 Alkyl, R 3c Each independently selected from halogen, CN and optionally halogen substituted-C 1-3 An alkyl group; preferably R 3 is-OH.
8. The compound of any one of claims 1-7, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, wherein Ra and Rb are both H.
9. The compound of any one of claims 1-8, stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, wherein R 4 Selected from the group consisting of-3-8 membered heterocycloalkyl and-C 3-6 Cycloalkyl optionally substituted with a group selected from: -OH, halogen, oxo, -CO 2 H. C optionally substituted by halogen 1-6 Alkyl, C 1-6 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, and C in the substituents 3-6 Cycloalkyl and-4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl) or-NH-SO 2 (C 1-3 Alkyl) substitution, wherein m is independently selected at each occurrence from 0 and 1.
10. The compound of claim 9, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, wherein R 4 Selected from the following groups:
in which case X is-O-or-NH- (CH) 2 ) n -, and n is 0;
in which case X is-NH- (CH) 2 ) n -, and n is 1;
preferably R 4 Has a structure selected from the group consisting of:
in which case X is-O-or-NH- (CH) 2 ) n -, and n is 0; and when X is-NH- (CH) 2 ) n -and n is 1, R 4 -CH in X to which it is attached 2 -together have a structure selected from:
more preferably R 4 Selected from the group consisting ofIn which case X is-O-or-NH- (CH) 2 ) n -, and n is 0, wherein Y is selected from O or CH 2
R 4a Independently selected from H, C 1-3 Alkyl, C 1-3 Hydroxyalkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, wherein C 3-6 Cycloalkyl and 4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl), -NH-SO 2 (C 1-3 Alkyl) substitution, and R 4b Independently selected from H, -OH, halogen, oxo, -CO 2 H. C optionally substituted by halogen 1-3 Alkyl and C 1-3 Hydroxyalkyl, wherein m is selected from 0 and 1;
preferably R 4a Independently selected from H, C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, and R 4b Is H, wherein m is 0;
more preferably R 4a Selected from H and C 1-3 Alkyl, and R 4b H.
11. The compound of claim 9, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, wherein R 4 Selected from the following groups:
in which case X is O or-NH- (CH) 2 ) n -, and n is preferably 0,
wherein R is 4c 、R 4c’ And R is 4d Each independently selected from H, -OH, halogen and C optionally substituted with halogen 1-3 Alkyl is preferably R 4 Selected from the following structures:
more preferably R 4 Is that
Wherein R is 4c Selected from H, halogen and C optionally substituted by halogen 1-3 Alkyl, preferably C 1-3 An alkyl group.
12. The compound of claim 1, stereoisomers, tautomers, stable isotopic variants, pharmaceutically acceptable salts or solvates thereof, wherein
X is-NH- (CH) 2 ) n -or-O-, wherein n is selected from 0 or 1;
wherein the method comprises the steps ofSelected from: />
R 1 Selected from halogen, C 1-6 Alkyl and C 1-6 Alkoxy group, wherein C 1-6 Alkyl and C 1-6 Alkoxy groups are each independently optionally substituted with halogen;
R 2 is H;
R 3 selected from-OH, -NHSO 2 -R 3a and-CH (R) 3c ) 2
R 3a is-C optionally substituted by halogen 1-6 An alkyl group;
R 3c independently at each occurrence selected from halogen;
Ra and Rb are each H;
R 4 selected from the group consisting of In which case X is-O-or-NH- (CH) 2 ) n -, and n is 0;
in which case X is-NH- (CH) 2 ) n -, and n is 1; and in which case X is O or-NH- (CH) 2 ) n -, and n is preferably 0;
wherein Y is selected from O or CH 2
R 4a Independently selected from H, C 1-3 Alkyl, - (CH) 2 ) m -C 3-6 Cycloalkyl and- (CH) 2 ) m -4-6 membered heterocycloalkyl, wherein C 3-6 Cycloalkyl and-4-6 membered heterocycloalkyl are each optionally C-substituted 1-3 Alkyl, -SO 2 (C 1-3 Alkyl) or-NH-SO 2 (C 1-3 Alkyl) substitution, wherein m is perEach occurrence is independently selected from 0 and 1; preferably R 4a Independently selected from H and C 1-3 An alkyl group;
R 4b and R is 4d Is H;
R 4c and R is 4c’ Each independently selected from H, -OH, halogen and C optionally substituted with halogen 1-3 An alkyl group; preferably R 4c And R is 4c’ One of OH and the other is selected from C 1-3 An alkyl group.
13. The compound of claim 1, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, selected from the group consisting of:
14. the compound of any one of claims 1 to 13, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, wherein one or more H atoms present are replaced by deuterium atoms; preferably, the hydrogen atom carried on the ring atom of the A ring and/or the hydrogen atom carried on the alkyl substituent of the A ring when carrying the alkyl substituent is replaced by a deuterium atom, and/or R 4 Hydrogen atom carried on substituent and/or when R 4 The hydrogen atoms on the alkyl substituents are replaced by deuterium atoms when carrying the alkyl substituents.
15. A compound according to any one of claims 1 to 14, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, for use in therapy, preferably as an NLRP3 inflammatory small inhibitor.
16. A pharmaceutical composition comprising a compound according to any one of claims 1 to 14, a stereoisomer, a tautomer, a stable isotope variant, a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.
17. A method of preventing or treating a disease associated with a small body of NLRP3 inflammation comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1 to 14, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition of claim 16.
18. Use of a compound according to any one of claims 1 to 14, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof, for inhibiting NLRP3 inflammatory body activity, in particular for treating and/or preventing diseases associated with NLRP3 inflammatory bodies.
19. Use of a compound according to any one of claims 1 to 14, a stereoisomer, tautomer, stable isotope variant, pharmaceutically acceptable salt or solvate thereof in the manufacture of a medicament for inhibiting NLRP3 inflammatory body activity, preferably for treating or preventing diseases associated with NLRP3 inflammatory body.
20. The method of claim 17 or use of any one of claims 18-19, wherein the NLRP3 inflammatory small-scale related disease is selected from the group consisting of auto-inflammatory febrile syndrome (e.g., leng Yan element-related periodic syndrome), sickle cell disease, systemic Lupus Erythematosus (SLE), liver-related diseases/disorders (e.g., chronic liver disease, viral hepatitis, non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis, and alcoholic liver disease), inflammatory arthritis-related disorders (e.g., gout, pseudogout (chondrocalpain), osteoarthritis, rheumatoid arthritis, joint diseases such as acute, chronic joint diseases), kidney-related diseases (e.g., hyperoxaluria, lupus nephritis, type I/II diabetes and related complications (e.g., kidney disease, retinopathy), hypertensive nephropathy, hemodialysis-related inflammation), neuroinflammation-related diseases (e.g., multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, alzheimer's disease), cardiovascular/metabolic diseases/disorders (e.g., cardiovascular risk reduction (crr), hypertension, atherosclerosis, type I and diabetes mellitus, and related conditions such as atherosclerosis, and vascular disease, acute atherosclerosis, aging-related conditions (e.g., atherosclerosis, acute atherosclerosis, and associated conditions), atherosclerosis, aging, and wound healing, and acne-related conditions (e.g., acne-related conditions). In particular, auto-inflammatory fever syndrome (e.g., CAPS), sickle cell disease, type I/II diabetes and related complications (e.g., nephropathy, retinopathy), gout, pseudogout (chondrocalcareous pigmentation disease), chronic liver disease, NASH, neuroinflammation related disorders) such as multiple sclerosis, brain infections, acute injury, neurodegenerative diseases, alzheimer's disease), atherosclerosis and cardiovascular risk (e.g., reduced cardiovascular risk (CvRR), hypertension), hidradenitis suppurativa, wound healing and scarring, and cancer related diseases/disorders (e.g., colon cancer, lung cancer, myeloproliferative neoplasms, leukemia, myelodysplastic syndrome (MDS), myelofibrosis).
21. The method or use of claim 20, wherein the NLRP3 inflammatory small-scale related disease is selected from the group consisting of: autoinflammatory fever syndrome (e.g., CAPS), sickle cell disease, type I/II diabetes and related complications (e.g., nephropathy, retinopathy), hyperoxaluria, gout, pseudogout (chondrocalcareous pigmentation), chronic liver disease, NASH, neuroinflammation related disorders (e.g., multiple sclerosis, brain infection, acute injury, neurodegenerative disease, alzheimer's disease), atherosclerosis and cardiovascular risk (e.g., reduced cardiovascular risk (CvRR), hypertension), suppurative sweat gland, wound healing and scarring, and cancer (e.g., colon cancer, lung cancer, myeloproliferative neoplasms, leukemia, myelodysplastic syndrome (MDS), myelofibrosis).
CN202310479336.XA 2022-04-29 2023-04-28 Fused pyridazine derivative, preparation method and medical application thereof Pending CN116969920A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210493977.6 2022-04-29
CN202210493977 2022-04-29

Publications (1)

Publication Number Publication Date
CN116969920A true CN116969920A (en) 2023-10-31

Family

ID=88473779

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310479336.XA Pending CN116969920A (en) 2022-04-29 2023-04-28 Fused pyridazine derivative, preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN116969920A (en)

Similar Documents

Publication Publication Date Title
AU2017323584C1 (en) Compounds and compositions as inhibitors of endosomal toll-like receptors
CN115448923B (en) Pyrimidine-fused ring compound, preparation method and application thereof
AU2006289281B2 (en) Pyridine derivatives and their use in the treatment of psychotic disorders
TW202144349A (en) Preparation and application method of heterocyclic compound as KRAS inhibitor
JP7381485B2 (en) Quinazolinone as a PARP14 inhibitor
EP3112366B1 (en) P2x7 modulators
PT2989106T (en) Fused heterocyclic compounds as protein kinase inhibitors
CN111050765B (en) Spiro compounds and methods of making and using the same
CN108003161B (en) Neurotrophic factor tyrosine kinase receptor inhibitors
JP2009507801A5 (en)
TW201422624A (en) Tricyclic fused thiophene derivatives as JAK inhibitors
AU2022334474A1 (en) Inhibitors of nlrp3
KR20220061958A (en) Heterobicyclic amides as inhibitors of CD38
EP3936509A1 (en) Fgfr4 kinase inhibitor, preparation method therefor and use thereof
EP4079734A1 (en) Triazolopyridazine derivative, preparation method therefor, pharmaceutical composition thereof, and use thereof
CA3182105A1 (en) Heterocyclic compounds as triggering receptor expressed on myeloid cells 2 agonists and methods of use
WO2023003002A1 (en) Annulated pyridazine compound
CN113527299A (en) Nitrogen-containing condensed ring compounds, preparation method and application
EP4322945A2 (en) Kras g12c inhibitors
CN113045569B (en) Compounds useful as RET kinase inhibitors and uses thereof
JP2023552838A (en) N-(imidazo[1,2-B]pyridazin-3-yl)-1-cyclohexyl-2H-indazole-5-carboxamide and N-(pyrazolo[1,5- A] Pyrimidin-3-yl)-1-cyclohexyl-2H-indazole-5-carboxamide derivative
JP2023536891A (en) JAK inhibitor compounds for treating severe pneumonia
TW202045501A (en) Bicyclic ether o-glycoprotein-2-acetamido-2-de oxy-3-d-glucopyranosidase inhibitors
CN116969920A (en) Fused pyridazine derivative, preparation method and medical application thereof
IL305203A (en) New thiazolopyrimidinone derivatives

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication