CN115873000A - Isoquinolone and quinazolinone compounds, and composition and application thereof - Google Patents

Isoquinolone and quinazolinone compounds, and composition and application thereof Download PDF

Info

Publication number
CN115873000A
CN115873000A CN202211183447.8A CN202211183447A CN115873000A CN 115873000 A CN115873000 A CN 115873000A CN 202211183447 A CN202211183447 A CN 202211183447A CN 115873000 A CN115873000 A CN 115873000A
Authority
CN
China
Prior art keywords
radical
alkyl
haloalkyl
deuterated
cycloalkyl
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
CN202211183447.8A
Other languages
Chinese (zh)
Inventor
吴双
孙明明
许世民
习宁
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.)
Zhongshan Medical Novishen New Drug R & D Co ltd
Original Assignee
Zhongshan Medical Novishen New Drug R & D 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 Zhongshan Medical Novishen New Drug R & D Co ltd filed Critical Zhongshan Medical Novishen New Drug R & D Co ltd
Publication of CN115873000A publication Critical patent/CN115873000A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • 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
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention belongs to the field of medicaments, and particularly relates to isoquinolone and quinazolinone compounds shown in formula (I) or (II), or stereoisomers, tautomers, deuterions, nitrogen oxides and solvates thereofOr a pharmaceutically acceptable salt thereof, and pharmaceutical compositions comprising the compounds and uses of the compounds and pharmaceutical compositions thereof in the preparation of medicaments for the prevention, treatment, and/or alleviation of PI 3-kinase mediated diseases, disorders, and/or conditions, or inhibition of PI 3-kinase activity. The compound provided by the invention has excellent inhibitory activity and kinase selectivity on target kinases, good pharmacokinetic properties and higher bioavailability.

Description

Isoquinolone and quinazolinone compounds, and composition and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to novel isoquinolinone and quinazolinone compounds, pharmaceutically acceptable salts thereof, pharmaceutical compositions containing the compounds, and application of the compounds and the pharmaceutical compositions in preparing medicines for preventing, treating and/or relieving diseases, disorders and/or conditions related to PI 3-kinase abnormality.
Background
The phosphoinositide 3-kinase (PI 3K) pathway is an intracellular signaling pathway that has a regulatory role in cell survival, proliferation and differentiation. The phosphoinositide 3-kinase (PI 3K) enzyme family is a central regulator of growth, proliferation, migration and metabolism in many cells and tissues. PI3K is a lipid kinase that produces lipid second messenger phosphatidylinositol-3, 4, 5-triphosphate (PIP 3), which is used downstream of cell surface receptors to regulate growth, metabolism, survival and differentiation. PIP3 is produced by four different class I PI3K catalytic isomers, divided into two groups: class IA (p 110 α, p110 β, and p110 δ) and class IB (p 110 γ). All class I PI3ks are constitutively associated with regulatory subunits, with the main difference between class IA and class IB PI3ks being their association with unique regulatory subunits. Class IA PI3Ks (PI 3K α, PI3K β and PI3K δ) are heterodimeric complexes consisting of a catalytic subunit p110 (p 110 α, p110 β and p110 δ, respectively) and a regulatory subunit p85 (e.g., p85 α, p85 β, p55 δ, p55 α and p50 α). These signal responses are typically transmitted via Receptor Tyrosine Kinases (RTKs). The signal of class IB PI3K γ is transmitted via G Protein Coupled Receptors (GPCRs) and consists of the catalytic subunit p110 γ. In normal cells, the PI3K/mTOR pathway has a regulatory role in cell survival, proliferation, and differentiation. However, aberrant activation of this pathway has been implicated in a variety of human diseases, including cancer, immunodeficiency, inflammation, and developmental disorders. Multiple inhibitors directed against key nodes within the PI3K pathway are in different stages of clinical development for the treatment of a variety of human diseases. ("Small-molecular inhibitors of the PI3K signaling network," Future MedChem.2011,3 (5), 549-565).
The expression patterns of the two subtypes, PI3K α and PI3K β, are ubiquitous, whereas the two subtypes, PI3K δ and PI3K γ, are mainly expressed in leukocytes. The relatively restricted expression patterns of PI3K δ and PI3K γ suggest an important role for these two subtypes in the adaptive and innate immune systems (j.med. Chem.2012,55 (20), 8559-8581).
The PI3K α subtype is associated with angiogenesis and glucose homeostasis. The PI3K/mTOR pathway is often deregulated in cancer, usually due to activating mutations or amplification of PIK3 CA. Gain-of-function mutations in PIK3CA, a gene encoding the PI3K p110 alpha catalytic subunit, are among the most common somatic alterations in solid tumors.
PI3K δ is expressed primarily in cells of the hematopoietic lineage and is activated by cytokine receptors, antigen receptors, growth factor receptors, and co-stimulatory receptors. PI3K δ is important in the development and activation of T and B cells. Blockade of the PI3K δ signaling pathway can increase gene instability. Gain of function (GOF) mutations in PI3K δ result in a range of developmental and functional defects in B and T cells, compromising host defense. Loss of function (LOF) mutations result in more severe B cell lymphopenia and agammaglobulinemia, but do not lead to T cell senescence.
The IB class PI3K catalytic subunit p110 γ is a major regulator of immune cell function, and p110 γ plays a key role in immune signaling. p110 γ is a key factor in inflammatory diseases and has been identified as a therapeutic target for cancer due to its immunomodulatory effects. PI3K γ plays an important role in the regulation of bone marrow (macrophages, mast cells, neutrophils) and lymphocyte (T cells, B cells and natural killer cells) derived immune cells. It modulates immune cell chemotaxis, cytokine release and the production of reactive oxygen species. The ability of PI3K γ to mediate a variety of immune cell functions is controlled by its activation downstream of numerous cell surface receptors, including G protein-coupled receptors (GPCRs), igE/antigen receptors, receptor Tyrosine Kinases (RTKs), and Toll-like receptors (TLRs). Genetic or pharmacological loss of PI3K γ has protective effects in mouse models against a variety of inflammatory diseases, including cardiovascular disease, arthritis, lupus, asthma, pulmonary inflammation and fibrosis, and metabolic syndrome. PI3K γ is also a driver of progression of pancreatic ductal adenocarcinoma through immunomodulatory effects. Targeting PI3K γ in combination with checkpoint inhibitors in the immune system has shown promise in clinical cancer therapy studies (Eganelisib, innovative Pharmaceuticals News Release, 2021). ( Henau OD, rausch M, winkler D et al Nature 539 (7629), 443-447 (2016); kaneda MM, messer KS, ralaiinirina N et al. Nature 539 (7629), 437-442 (2016). )
In view of its key role in cancer and immunity, the PI3K pathway has been the focus of drug development research for the last two decades. In 2014, the PI3K delta inhibitor idelalisib (Zyderig; gilead Sciences) was the first PI3K inhibitor approved for use in specific B cell malignant tumors. Subsequently, the pan-I class PI3K inhibitor copanlisib (Aliqopa; bayer) was approved in 2017, and the PI3K delta/PI 3K gamma dual inhibitor duvelisib (Copiktra; verastem, now Secura Bio) was approved for the same indication in 2018. The PI3K alpha inhibitor, alpelisib (Piqray; nowa), was the first and only PI3K alpha inhibitor approved by the FDA at 2019 at 5 months for use in combination with the Estrogen Receptor (ER) down-regulator fulvestrant in the treatment of PIK3CA mutant HER2/ER + metastatic advanced breast cancer.
There is a need to provide new, good candidate drug PI3K inhibitors. In particular, preferred compounds should bind strongly to the PI3K receptor while showing little affinity for other receptors and show functional activity as agonists. The compound should be well absorbed from the gastrointestinal tract, be metabolically stable and have good pharmacokinetic properties. When targeting receptors in the central nervous system, they are free to cross the blood-brain barrier, and when selectively targeting receptors in the peripheral nervous system, they should not cross the blood-brain barrier. They should be non-toxic and exhibit few side effects. In addition, the ideal drug candidate should exist in a physical form that is stable, non-hygroscopic, and easily formulated. The compounds of the invention show specific levels of selectivity for PI3K α, β, γ and δ against different paralogs. In particular, a certain level of selectivity against Ρ Ι 3K γ is shown.
The compounds, compositions and methods described herein directly address these needs and other objects. In particular, the invention provides a class of compounds that inhibit, modulate and/or modulate PI 3-kinase activity for use in the treatment and/or prevention of diseases, disorders, and/or conditions associated with PI 3-kinase abnormalities. Compared with the existing similar compounds, the compound has better pharmacological activity, particularly, the compound shows excellent inhibitory activity on PI 3-kinase, has high selectivity on PI3K gamma, has obvious advantage on the stability of liver microsome, has good pharmacokinetic property and higher bioavailability. Therefore, the compound has a very good development prospect.
Disclosure of Invention
Definition of terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. One skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, such as compounds of the general formula of the invention, or as specified in the examples, subclasses, and classes encompassed by the invention.
It is understood that the term "optionally substituted" may be used interchangeably with the term "substituted or unsubstituted". In general, the term "substituted" means that one or more hydrogen atoms in a given structure is replaced with a particular substituent, whether the hydrogen atom is attached to a C or N atom or other atom. "optionally" unless otherwise indicated, an optional substituent group may be substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently.
The term "optionally substituted with (8230) \8230; substituted" may be used interchangeably with the term "unsubstituted or substituted with (8230) \8230; substituted with (i.e. the structure is unsubstituted or substituted with one or more substituents described herein, including, but not limited to, H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、-OC(=O)R 7 、-C(=O)OR 7a 、-S(=O) 0-2 R 7 、-OS(=O) 1-2 R 7 、-S(=O) 1-2 OR 7a 、-N(R 8a )C(=O)R 8 、-C(=O)NR 8a R 8 、-OC(=O)NR 8a R 8 、-N(R 8a )S(=O) 1-2 R 8 、-S(=O) 1-2 NR 8a R 8 、-N(R 8a )C(=O)NR 8a R 8 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 1-6 Alkoxy radical, C 1-6 Alkylamino radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-7 Heterocyclic group, C 2-7 Heterocyclyl radical C 1-6 Alkyl radical, C 6-12 Aryl radical, C 6-12 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each substituent is independently optionally substituted with 0, 1, 2, 3 or 4 substituents independently selected from H, D, oxo (= O), F, cl, br, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Deuterated alkyl, C 1-6 Alkyl and C 1-6 Alkoxy, and the like. Wherein R is e 、R f 、R 9 、R 9a 、R 10 And R 10a Have the definitions as described in the present invention.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C 1 -C 6 Alkyl "means in particular independently disclosed methyl, ethyl, C 3 Alkyl radical, C 4 Alkyl radical, C 5 Alkyl and C 6 An alkyl group.
The term "alkyl" or "alkyl group" as used herein, denotes a saturated, straight or branched chain monovalent hydrocarbon radical containing from 1 to 20 carbon atoms, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1-20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 3 carbon atoms. The alkyl group may be optionally substituted with one or more substituents described herein.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH) 3 ) Ethyl group (Et, -CH) 2 CH 3 ) N-propyl (n-Pr, -CH) 2 CH 2 CH 3 ) Isopropyl group (i-Pr, -CH (CH) 3 ) 2 ) N-butyl (n-Bu, -CH) 2 CH 2 CH 2 CH 3 ) Isobutyl (i-Bu, -CH) 2 CH(CH 3 ) 2 ) Sec-butyl (s-Bu, -CH (CH) 3 )CH 2 CH 3 ) Tert-butyl (t-Bu, -C (CH) 3 ) 3 ) N-pentyl (-CH) 2 CH 2 CH 2 CH 2 CH 3 ) 2-pentyl (-CH (CH) 3 )CH 2 CH 2 CH 3 ) 3-pentyl (-CH (CH) 2 CH 3 ) 2 ) 2-methyl-2-butyl (-C (CH) 3 ) 2 CH 2 CH 3 ) 3-methyl-2-butyl (-CH (CH) 3 )CH(CH 3 ) 2 ) 3-methyl-1-butyl (-CH) 2 CH 2 CH(CH 3 ) 2 ) 2-methyl-1-butyl (-CH) 2 CH(CH 3 )CH 2 CH 3 ) N-hexyl (-CH) 2 CH 2 CH 2 CH 2 CH 2 CH 3 ) 2-hexyl (-CH (CH) 3 )CH 2 CH 2 CH 2 CH 3 ) 3-hexyl (-CH (CH) 2 CH 3 )(CH 2 CH 2 CH 3 ) 2-methyl-2-pentyl (-C (CH)) 3 ) 2 CH 2 CH 2 CH 3 ) 3-methyl-2-pentyl (-CH (CH) 3 )CH(CH 3 )CH 2 CH 3 ) 4-methyl-2-pentyl (-CH (CH) 3 )CH 2 CH(CH 3 ) 2 ) 3-methyl-3-pentyl (-C (CH) 3 )(CH 2 CH 3 ) 2 ) 2-methyl-3-pentyl (-CH (CH) 2 CH 3 )CH(CH 3 ) 2 ) 2, 3-dimethyl-2-butyl (-C (CH) 3 ) 2 CH(CH 3 ) 2 ) 3, 3-dimethyl-2-butyl (-CH (CH) 3 )C(CH 3 ) 3 ) N-heptyl, n-octyl, and the like.
Term "Alkenyl "denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp 2 Double bonds, which include both "cis" and "trans" orientations, or "E" and "Z" orientations. In one embodiment, the alkenyl group contains 2 to 8 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH = CH) 2 ) Allyl (-CH) 2 CH=CH 2 ) And so on. The alkenyl group may be optionally substituted with one or more substituents described herein.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp triple bond. In one embodiment, alkynyl groups contain 2-8 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, an alkynyl group contains 2-4 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C ≡ CH), propargyl (-CH) 2 C [ identical to ] CH), 1-propynyl (-C [ identical to ] C-CH) 3 ) And so on. The alkynyl group may be optionally substituted with one or more substituents described herein.
The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the definition as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein.
Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH) 3 ) Ethoxy (EtO, -OCH) 2 CH 3 ) 1-propoxy (n-PrO, n-propoxy, -OCH) 2 CH 2 CH 3 ) 2-propoxy group(i-PrO, i-propoxy, -OCH (CH) 3 ) 2 ) 1-butoxy (n-BuO, n-butoxy, -OCH) 2 CH 2 CH 2 CH 3 ) 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH) 2 CH(CH 3 ) 2 ) 2-butoxy (s-BuO, s-butoxy, -OCH (CH) 3 )CH 2 CH 3 ) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH) 3 ) 3 ) 1-pentyloxy (n-pentyloxy, -OCH) 2 CH 2 CH 2 CH 2 CH 3 ) 2-pentyloxy (-OCH (CH) 3 )CH 2 CH 2 CH 3 ) 3-Pentoxy (-OCH (CH)) 2 CH 3 ) 2 ) 2-methyl-2-butoxy (-OC (CH)) 3 ) 2 CH 2 CH 3 ) 3-methyl-2-butoxy (-OCH (CH) 3 )CH(CH 3 ) 2 ) 3-methyl-l-butoxy (-OCH) 2 CH 2 CH(CH 3 ) 2 ) 2-methyl-l-butoxy (-OCH) 2 CH(CH 3 )CH 2 CH 3 ) And so on.
The term "haloalkyl" or "haloalkoxy" means an alkyl or alkoxy group substituted with one or more halogen atoms, examples of which include, but are not limited to, trifluoromethyl (-CF) 3 ) Trifluoromethoxy (-OCF) 3 ) Difluoroethyl (-CH) 2 CHF 2 ,-CF 2 CH 3 ,-CHFCH 2 F) Trifluoroethyl (-CH) 2 CF 3 ,-CF 2 CH 2 F,-CFHCHF 2 )、-CF(CH 3 ) 2 And so on.
The terms "deuterated alkyl," "deuterated alkoxy," "deuterated cycloalkyl," "deuterated heterocyclyl," "deuterated aryl," or "deuterated heteroaryl" mean that the alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, or heteroaryl group is substituted with one or more D atoms, examples of which include, but are not limited to, a deuterated methyl group (-CH) 2 D) Dideuteromethyl (-CHD) 2 ) Tri-deuterated methyl (-CD) 3 ) And deuterated methoxy (-OCH) 2 D) Dideuteromethoxy (-OCHD) 2 ) Trideuteromethoxy (-OCD) 3 ) Two, twoDeuterated ethyl (-CH) 2 CHD 2 ,-CD 2 CH 3 ,-CHDCH 2 D) Pentadeuteroethyl (-CD) 2 CD 3 ) Deuterated cyclopropyl, deuterated cyclohexyl, pentadeuterated phenyl, and the like.
The term "deuterium (D) -containing group" means that one or more H atoms on a group or part of a group described herein are replaced by D, but does not contain a separate D group, such groups including, but not limited to, C 1-6 Deuterated alkyl, C 3-8 Deuterated cycloalkyl, C 2-9 Deuterated heterocyclic radical, C 6-10 Deuterated aryl, C 1-9 Deuterated heteroaryl, each having the definitions described herein, wherein each of said groups is independently optionally substituted with one or more of H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl and C 1-6 Substituted by deuterated alkyl radicals, wherein R e And R f All have the definitions as set forth herein.
The terms "hydroxyalkyl" or "hydroxy-substituted alkyl" and "hydroxyalkoxy" or "hydroxy-substituted alkoxy" respectively denote an alkyl or alkoxy group, optionally substituted with one or more hydroxy groups, wherein "hydroxyalkyl" and "hydroxyalkyl" may be used interchangeably, and such examples include, but are not limited to, hydroxymethyl (-CH) 2 OH), 2-hydroxyethyl (-CH) 2 CH 2 OH), 1-hydroxyethyl (-CH (OH) CH 3 ) 2-hydroxypropan-2-yl (-COH (CH) 3 ) 2 ) 2-hydroxy-2-methylpropyl (-CH) 2 COH(CH 3 ) 2 ) 3-hydroxypropyl (-CH) 2 CH 2 CH 2 OH), 2-hydroxypropyl (-CH) 2 CH(OH)CH 3 ) 2-hydroxy-2-methylpropyl (-CH) 2 CH(OH)(CH 3 )CH 3 ) Hydroxy methoxy (-OCH) 2 OH), and the like.
The term "cyano-substituted alkyl" or "cyanoalkyl" includes C substituted with one or more cyano groups 1-10 A straight or branched alkyl group. Wherein some embodiments are cyanoalkyl is C substituted with one or more cyano groups 1-6 "lower cyanoalkyl", othersFor example, cyanoalkyl is C substituted by one or more cyano groups 1-4 "lower cyanoalkyl", examples of such include, but are not limited to, CNCH 2 -、CNCH 2 CH 2 -、CNCH 2 CH 2 CH 2 -、CNCH 2 CHCNCH 2 -and the like.
The term "alkylamino" includes "N-alkylamino" and "N, N-dialkylamino" in which the amino groups are each independently substituted with one or two alkyl groups. In some of these embodiments, the alkylamino group is one or two C 1-6 Lower alkylamino groups in which the alkyl group is attached to the nitrogen atom. In other embodiments, the alkylamino group is C 1-3 Lower alkylamino groups. Suitable alkylamino groups can be monoalkylamino or dialkylamino, and such examples include, but are not limited to, N-methylamino, N-ethylamino, N-dimethylamino, N-diethylamino, and the like.
The term "aminoalkyl" includes C substituted with one or more amino groups 1-10 A straight or branched alkyl group. In some of these embodiments, aminoalkyl is C substituted with one or more amino groups 1-6 "lower aminoalkyl" and other examples are aminoalkyl which is C substituted with one or more amino groups 1-4 "lower aminoalkyl" such examples include, but are not limited to, aminomethyl, aminoethyl, aminopropyl, aminobutyl, and aminohexyl.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 carbon atoms. Bicyclic cycloalkyl includes spirobicycloalkyl, fused bicycloalkyl and bridged bicycloalkyl. In some embodiments, cycloalkyl groups contain 3 to 12 carbon atoms; in other embodiments, cycloalkyl groups contain 3 to 10 carbon atoms; in other embodiments, cycloalkyl groups contain 3 to 8 carbon atoms; in other embodiments, cycloalkyl groups contain 3 to 7 carbon atoms; in other embodiments, cycloalkyl groups contain 3 to 6 carbon atoms; in still other embodiments, cycloalkyl is C 7 -C 12 Cycloalkyl group containing C 7 -C 12 Monocyclic alkyl, C 7 -C 12 Bicycloalkyl (e.g. C) 7 -C 12 Spirobicycloalkyl radical, C 7 -C 12 Fused bicycloalkyl and C 7 -C 12 Bridged bicycloalkyl) or C 7 -C 12 A tricyclic alkyl group. The cycloalkyl group may be independently unsubstituted or substituted with one or more substituents described herein. The term "monocyclic cycloalkyl" or "monocycloalkyl" denotes a cycloalkyl group of a monocyclic ring system, wherein the cycloalkyl group has the definitions as described above, and the monocyclic cycloalkyl group can independently be unsubstituted or substituted by one or more substituents described herein. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-1-alkenyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
The term "cycloalkylalkyl" includes cycloalkyl-substituted alkyl groups. In some embodiments, cycloalkylalkyl groups refer to "lower cycloalkylalkyl" groups, i.e., the cycloalkyl group is attached to C 1-6 On the alkyl group of (a). In other embodiments, cycloalkylalkyl groups refer to C-containing groups 1-3 The "phenylalkylene" of an alkyl group of (a). Specific examples thereof include, but are not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, cyclopentylethyl, cyclohexylethyl, and the like. The cycloalkyl group on the cycloalkylalkyl group may be further substituted with one or more substituents described herein.
The terms "heterocyclyl" and "heterocycle" are used interchangeably herein and refer to a mono-, bi-or tricyclic ring system containing 3 to 12 ring atoms which is mono-or polyvalent, saturated or partially unsaturated, and which is not aromatic, wherein at least one ring atom is selected from nitrogen, oxygen and sulfur atoms. In some embodiments, heterocyclyl or heterocyclic contains 4-12 ring atoms. In some embodiments, heterocyclyl or heterocyclic ring contains 5-12 ring atoms. In some embodiments, heterocyclyl or heterocyclic ring includes4-8 ring atoms. In some embodiments, heterocyclyl or heterocyclic contains 3-10 ring atoms. In some embodiments, heterocyclyl or heterocyclic ring contains 3-8 ring atoms. In some embodiments, heterocyclyl or heterocyclic contains 3-6 ring atoms. In some embodiments, heterocyclyl or heterocyclic contains 4-7 ring atoms. Unless otherwise specified, a heterocyclic group may be carbon-or nitrogen-based, and-CH 2 The group can optionally be replaced by-C (= O) -the sulphur atom of the ring can optionally be oxidized to S-oxide and the nitrogen atom of the ring can optionally be oxidized to N-oxygen compound. The heterocyclic group includes a saturated heterocyclic group (heterocycloalkyl group) and a partially unsaturated heterocyclic group. The heterocyclic group has one or more attachment points to the rest of the molecule. Examples of heterocyclyl groups include, but are not limited to: <xnotran> , , , , , , , , , , , , , ,1,3- , , , , 2H- , 4H- , , , , , , , , , , , , , , </xnotran>
Figure BDA0003866228850000041
Yl (e.g., 1, 4-oxa->
Figure BDA0003866228850000042
Base, 1, 2-oxa->
Figure BDA0003866228850000051
Base), diaza->
Figure BDA0003866228850000052
Based on (e.g., 1, 4-diaza->
Figure BDA0003866228850000053
Basic, 1, 2-dinitrogen Is mixed and/or screened>
Figure BDA0003866228850000054
Base), dioxan->
Figure BDA0003866228850000055
Base (e.g., 1, 4-dioxa->
Figure BDA0003866228850000056
Base, 1, 2-dioxa->
Figure BDA0003866228850000057
Yl), thiamethoxam->
Figure BDA0003866228850000058
Yl (e.g. 1, 4-thiaza ™ is present>
Figure BDA0003866228850000059
Base, 1, 2-thiaza->
Figure BDA00038662288500000510
Yl), indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1 ] yl]Hept-5-yl, 2-azaspiro [4.4]Nonyl, 1, 6-dioxaspiro [4.4 ]]Nonyl, 2-azaspiro [4.5 ]]Decyl, 8-azaspiro [4.5 ]]Decyl, 7-azaspiro [4.5 ]]Decyl, 3-azaspiro [5.5 ]]Undecyl, 2-azaspiro [5.5]Undecyl, octahydro-1H-isoindolyl, octahydrocyclopenta [ c]Pyrrolyl, indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, hexahydrofuro [3,2-b ]]Furyl and dodecahydroisoquinolinyl, and the like. Examples of substitutions of the-CH 2-group in the heterocyclyl by-C (= O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl and 3, 5-dioxopiperidinyl. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, sulfolane group, 1-dioxothiomorpholinyl group. The heterocyclyl group may be optionally substituted with one or more substituents as described herein.
In yet another embodiment, heterocyclyl is a heterocyclic group of 4 to 7 atoms, meaning that A mono-or polyvalent, saturated or partially unsaturated, non-aromatic mono-or bicyclic ring comprising 4 to 7 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur and oxygen atoms. Unless otherwise specified, a heterocyclic group of 4 to 7 atoms may be carbon-based or nitrogen-based, and-CH 2 The-group may optionally be replaced by-C (= O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. The 4-7 atom heterocyclyl group has one or more attachment points to the rest of the molecule. Among them, examples of the monocyclic heterocyclic group consisting of 4 to 7 atoms include, but are not limited to: azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxazinyl, 1, 2-oxazinyl, 1, 2-thiazinyl, hexahydropyridazinyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepanyl, oxazepinyl
Figure BDA00038662288500000511
Yl (1, 4-oxazepine ™)>
Figure BDA00038662288500000512
Base, 1, 2-oxazepine>
Figure BDA00038662288500000513
Basyl), diaza->
Figure BDA00038662288500000514
Base (1, 4-diaza->
Figure BDA00038662288500000515
Base, 1, 2-diaza->
Figure BDA00038662288500000516
Radical) and sulfur nitrogenIs mixed and/or screened>
Figure BDA00038662288500000517
Yl (1, 4-thiaza ™ is present>
Figure BDA00038662288500000518
Base, 1, 2-thiaza->
Figure BDA00038662288500000519
Base), etc.; examples of 4-7 atom bicyclic heterocyclic groups include, but are not limited to: 3-azabicyclo [3,2,0]Heptane, 3-oxobicyclo [3,2,0 ]]Heptane and the like; 4-7 atoms in heterocyclic radical 2 Examples of the substitution of the-group by-C (= O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl and 3, 5-dioxopiperidinyl; examples of the sulfur atom in the heterocyclic group consisting of 4 to 7 atoms being oxidized include, but are not limited to, sulfolane group, 1-dioxotetrahydrothiophene, 1-dioxotetrahydrothiopyran, 1-dioxothiomorpholinyl. Said heterocyclyl group of 4 to 7 atoms may be optionally substituted by one or more substituents as described herein.
The term "heterocyclylalkyl" includes heterocyclyl-substituted alkyl groups in which both heterocyclyl and alkyl have the meanings as described herein, and such examples include, but are not limited to, tetrahydrofuryl methyl, pyrrol-2-ylmethyl, morpholin-4-ylethyl, piperazin-4-ylethyl, piperidin-4-ylethyl, and the like.
The term "aryl" denotes monocyclic, bicyclic and tricyclic carbon ring systems containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring system is aromatic, wherein each ring system contains 3 to 7 atoms in the ring and one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring". Examples of the aryl group may include phenyl, naphthyl and anthracenyl. The aryl group may independently be optionally substituted with one or more substituents described herein.
The term "arylalkyl" or "aralkyl" includes arylAn alkyl group substituted with a substituent. In some embodiments, an arylalkyl group refers to a "lower arylalkyl" group, i.e., the aryl group is attached to C 1-6 On the alkyl group of (a). In other embodiments, arylalkyl group refers to a C-containing radical 1-3 The "phenylalkylene" of an alkyl group of (a). Specific examples thereof include, but are not limited to, benzyl, diphenylmethyl, phenylethyl, and the like. The aryl group on the arylalkyl group can be further substituted with one or more substituents described herein.
The term "heteroaryl" denotes monocyclic, bicyclic, and tricyclic ring systems containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein all rings are aromatic and at least one aromatic ring contains one or more heteroatoms, wherein each ring system contains a ring of 5 to 7 atoms with one or more attachment points to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". In one embodiment, heteroaryl is a 5-12 atom heteroaryl comprising 1,2,3, or 4 heteroatoms independently selected from O, S, and N. In another embodiment, heteroaryl is a 5-10 atom composed heteroaryl containing 1,2,3, or 4 heteroatoms independently selected from O, S, and N. In another embodiment, heteroaryl is 5-6 atom composed of 1,2,3, or 4 heteroatoms independently selected from O, S, and N. The heteroaryl group is optionally substituted with one or more substituents described herein.
Examples of heteroaryl groups include, but are not limited to, 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 3-triazolyl, 1,2, 3-thiadiazolyl, 1,3, thiadiazolyl, 1, 3-thiadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 5-thiadiazolyl, 1, 3-triazinyl; the following bicyclic rings are also included, but are in no way limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl), imidazo [1,2-a ] pyridyl, pyrazolo [1,5-a ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, [1,2,4] triazolo [4,3-b ] pyridazinyl, [1,2,4] triazolo [1,5-a ] pyrimidinyl, [1,2,4] triazolo [1,5-a ] pyridyl, and the like.
The term "heteroarylalkyl" denotes an alkyl group substituted with one or more heteroaryl groups, wherein both the heteroaryl and alkyl groups have the meaning described herein, and such examples include, but are not limited to, pyridine-2-methyl, imidazole-2-methyl, furan-2-ethyl, indole-3-methyl, and the like.
The term "halogen" refers to F, cl, br or I.
As described herein, there are two linking points in the ring system that are linked to the rest of the molecule, as shown in formula (a 1), meaning that either the E or E' end is linked to the rest of the molecule, i.e., the linking modes at the two ends can be interchanged.
E-(CH 2 ) t1 -L-E′ (a1)
As described herein, the ring system formed by the substituents on the ring with a bond to the center (as shown below) represents that the substituents may be substituted at any substitutable position on either ring. For example, formula B represents that any possible substituted position on the A or B ring may be substituted as shown in formulas c, d, e, f, g, h, i, j, k, l, m, n, o, p, q, etc.
Figure BDA0003866228850000061
As used herein, "pharmaceutically acceptable salts" refers to both organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptableAcceptable salts are well known in the art, as in the literature: berge et al, description of the descriptive pharmaceutical acceptable salts in detail in J. Pharmaceutical Sciences,1977, 66. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids formed by reaction with amino groups such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or those obtained by other methods described in the literature above, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, lauryl sulfates, malates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, thiocyanates, p-toluenesulfonates, undecanoates, valeric acid salts, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl radical) 4 A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C 1-8 Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
Description of the Compounds of the invention
The present invention discloses a novel class of pyrimidine amine compounds, which are useful as inhibitors of PI 3-kinase activity, in particular PI 3K-gamma activity, for the prevention, treatment, and/or amelioration of diseases, disorders, and/or conditions associated with PI 3-kinase abnormalities, such as respiratory diseases, viral infections, non-viral respiratory infections, allergic diseases, autoimmune diseases, inflammatory diseases, cardiovascular diseases, hematological malignancies, neurodegenerative diseases, pancreatitis, multi-organ failure, kidney diseases, platelet aggregation, cancer, sperm motility, transplant rejection, lung injury or pain, and the like. Compared with the existing similar compounds, the compound has better pharmacological activity, particularly, the compound shows excellent inhibitory activity on PI 3-kinase, has high selectivity on PI3K gamma, and shows obvious advantages in the aspect of pharmacokinetics. Therefore, the compound has very good development prospect.
The compound disclosed by the invention can show stronger inhibitory activity on PI 3-kinase, particularly PI 3K-gamma. On one hand, the invention provides isoquinolone and quinazolinone compounds, which have structures shown in formula (I):
Figure BDA0003866228850000071
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof;
wherein, the first and the second end of the pipe are connected with each other,
x is-C (R) c ) -, or N;
Z 1 and Z 2 Each independentlyis-C (R) 4d ) -or N;
R a 、R b and R c Each independently is H, D, F, cl, C 1-3 Alkyl radical, C 1-6 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl radical, C 1-3 Aminoalkyl radical, C 1-3 Cyanoalkyl, C 1-6 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-6 Haloalkyl, C substituted by 1 to 5 deuterium 1-6 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-8 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group;
R 1 is H, D, -C 1-6 alkyl-NR e R f 、-C(=O)R 7 、-C(=O)OR 7a 、-C(=O)NR 7 R 7a 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl radical, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
R 2 at each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 2 At each occurrence, independently is optionally substituted with 0, 1, 2, 3, 4, or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution;
R 3 is H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-8 Cycloalkyl, C 3-8 Deuterated cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 3 Optionally substituted by 0, 1, 2, 3, 4 or 5R 6 Substitution;
R 4a 、R 4b 、R 4c and R 4d Each independently of the other is H, D, F, cl, -CN, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 4a 、R 4b 、R 4c And R 4d Each independently is optionally substituted by 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy radical, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl and C 1-6 Substituted with a group of halogenated deuterated alkyl;
R 5 And R 6 Each occurrence is independently H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of said-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution;
R e 、R f 、R 7 and R 7a At each occurrence, independently H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-12 Aryl radical, C 6-12 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group; and
n is 0, 1, 2, or 3;
with the following conditions: 1) When R is 1 Is pyrazolyl, wherein R 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substituted, then each R 2 、R 4a 、R 4b And R 4c At least one of is independentGround is F, cl, -CN, C 1-6 Alkoxy radical, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-6 Haloalkyl, C substituted by 1 to 5 deuterium 1-6 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-8 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group; or 2) when R is 1 Is H, D, -C 1-6 alkyl-NR e R f 、-C(=O)R 7 、-C(=O)OR 7a 、-C(=O)NR 7 R 7a 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl radical, C 2-9 Heterocyclic group, C 6-10 Aryl, or C other than pyrazolyl 1-9 In the case of heteroaryl, wherein R 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substituted, then each R a 、R b 、R c 、R 2 、R 3 、R 4a 、R 4b 、R 4c And R 4d Is independently D (deuterium), C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-6 Haloalkyl, C substituted by 1 to 5 deuterium 1-6 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-8 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group.
On the other hand, the invention provides isoquinolinone and quinazolinone compounds, which have the structures shown in the formula (Ib):
Figure BDA0003866228850000081
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof;
wherein the content of the first and second substances,
x is-C (R) c ) -, or N;
Z 1 and Z 2 Each independently is-C (R) 4d ) -or N;
R a 、R b and R c Each of which isIndependently H, D, F, cl, C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl radical, C 1-3 Aminoalkyl, or C 1-3 Cyanoalkyl;
R 1 is H, D, -C 1-6 alkyl-NR e R f 、-C(=O)R 7 、-C(=O)OR 7a 、-C(=O)NR 7 R 7a 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
R 2 at each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each R 2 Independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group;
R 3 is H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-8 Cycloalkyl radical, C 3-8 Deuterated cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 3 Optionally substituted by 0, 1, 2, 3, 4 or 5R 6 Substitution;
R 4a 、R 4b 、R 4c and R 4d Each independently is H, D, F, cl, a deuterium-containing group, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 4a 、R 4b 、R 4c And R 4d Each independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy radical, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl and C 1-6 Substituted with a group of halogenated deuterated alkyl;
R 5 and R 6 At each occurrence, independently is H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of said-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution;
R e 、R f 、R 7 and R 7a At each occurrence, independently H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-12 Aryl radical, C 6-12 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group; and
n is 0, 1, 2, or 3;
with the following conditions: 1) When R is 1 When is pyrazolyl, then R 2 、R 4a 、R 4b And R 4c Is F, cl, or a deuterium containing group; or 2) when R is 1 Is H, D, -C 1-6 alkyl-NR e R f 、-C(=O)R 7 、-C(=O)OR 7a 、-C(=O)NR 7 R 7a 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl radical, C 2-9 Heterocyclic group, C 6-10 Aryl, or C other than pyrazolyl 1-9 When it is heteroaryl, R a 、R b 、R c 、R 2 、R 3 、R 4a 、R 4b 、R 4c And R 4d Is independently D (deuterium), or a deuterium-containing group.
In some embodiments, the compound of formula (I) has the structure of formula (Ia):
Figure BDA0003866228850000091
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof.
In some embodiments of formula (I) or (Ia), R 1 Is phenyl, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, thiazolyl, imidazolyl, oxazolyl, thiadiazolyl,
Figure BDA0003866228850000101
Figure BDA0003866228850000102
Wherein Y is 1 Is O, S, or-NH-;
Y 6 is O, or-NH-;
t3 and t4 are each independently 1, 2, 3 or 4; and
wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
the conditions are as follows: 1) R 1 Is composed of
Figure BDA0003866228850000103
In which R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substituted, then each R 2 、R 4a 、R 4b And R 4c At least one of which is independently F, cl, -CN, C 1-3 Alkoxy radical, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C substituted by 1 to 5 deuterons 1-3 Haloalkyl, C substituted by 1 to 5 deuterium 1-3 Hydroxyalkyl quilt1 to 5 deuterium substituted C 3-6 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group; 2) When R is 1 Is phenyl, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, thiazolyl, imidazolyl, oxazolyl, thiadiazolyl, or>
Figure BDA0003866228850000104
In which R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substituted, then each R a 、R b 、R c 、R 2 、R 3 、R 4a 、R 4b 、R 4c And R 4d Is independently D (deuterium), C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-3 Haloalkyl, C substituted by 1 to 5 deuterium 1-3 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-6 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group.
In some embodiments of formula (I) or (Ib), R 1 Is phenyl, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, thiazolyl, imidazolyl, oxazolyl, thiadiazolyl,
Figure BDA0003866228850000105
Figure BDA0003866228850000106
Wherein Y is 1 Is O, S, or-NH-;
Y 6 is O, or-NH-;
t3 and t4 are each independently 1, 2, 3 or 4; and
wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
with the following conditions: 1) R is 1 Is composed of
Figure BDA0003866228850000107
When then R is 2 、R 4a 、R 4b And R 4c Is independently F, cl, or contain A radical of deuterium; (ii) a 2) When R is 1 Is phenyl, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, thiazolyl, imidazolyl, oxazolyl, thiadiazolyl, or>
Figure BDA0003866228850000108
Figure BDA0003866228850000111
When it is, then R a 、R b 、R c 、R 2 、R 3 、R 4a 、R 4b 、R 4c And R 4d Is independently D (deuterium), or a deuterium-containing group.
In some embodiments of formula (I) or (Ia), R 4a 、R 4b 、R 4c And R 4d Each independently is H, D, F, cl, C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, C 1-3 Hydroxyalkyl radical, C 1-3 Aminoalkyl radical, C 1-3 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Deuterated heterocyclic group, C 3-6 Cycloalkyl radical, C 3-6 Deuterated cycloalkyl, phenyl, or heteroaryl of 5-6 atoms; wherein R is 4a 、R 4b 、R 4c And R 4d Each independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl and C 1-3 Substituted with a group of halogenated deuterated alkyl.
In some embodiments of formula (I), (Ia) or (Ib), R 4a 、R 4b 、R 4c And R 4d Each independently of the other is H, F, cl, D, -CN, -OCH 3 ,-OCH 2 CH 3
Figure BDA0003866228850000112
/>
Figure BDA0003866228850000113
In another aspect, the present invention also provides a compound of formula (II):
Figure BDA0003866228850000114
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof;
Wherein the content of the first and second substances,
x is-C (R) c ) -, or N;
w is C 3-10 Cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein W is optionally substituted with 0, 1, 2, 3 or 4R 4 Substitution;
R a 、R b and R c Each independently is H, D, F, cl, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl, or C 1-6 A cyanoalkyl group;
R 1 is-C 1-3 hydroxyalkyl-C (= O) OR 7a ,-C(=O)OR 7a ,-C(=O)NR 7 R 7a
Figure BDA0003866228850000115
Figure BDA0003866228850000116
Wherein Y is 2 And Y 3 Each independently is- (CH) 2 ) t1 -、-(CH 2 ) t1 -L-、-(CH 2 ) t1 -L-(CH 2 ) t2 -, O, or-NH-;
Y 4 is- (CH) 2 ) t1 -、-(CH 2 ) t1 -L-、-(CH 2 ) t1 -L-(CH 2 ) t2 -、-C (= O) -, O, S, or-NH-;
Y 5 is-CH-, or N;
Y 6 is O, or-NH-;
Y 7 is O, or-NH-;
l is-C (= O) -, O, S, or-NH-;
t1, t2 and t3 are each independently at each occurrence 1, 2, 3 or 4;
t4 is 1; t5 is 1 or 2; and
Figure BDA0003866228850000121
represents a single or double bond; and
wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
R 2 at each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 2 At each occurrence, is independently optionally substituted with 0, 1, 2, 3, 4, or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group;
R 3 is H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-8 Cycloalkyl, C 3-8 Deuterated cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 3 Optionally substituted by 0, 1, 2, 3, 4 or 5R 6 Substitution;
R 4 at each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 4 At each occurrence, is independently optionally substituted with 0, 1, 2, 3, 4, or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution;
R 5 and R 6 At each occurrence, independently is H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of said-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted by 0, 1, 2, 3 4 or 5 independently selected from H, D, oxo (= O), F, cl,Br、I、-OH、-NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group;
R 5a is-C (= O) R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of said-C (= O) R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution;
R e 、R f 、R 7 And R 7a At each occurrence, independently H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each C is 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-12 Aryl radical, C 6-12 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group; and
n is 0, 1, 2, or 3.
In some embodiments of formula (II), W is
C 3-8 Cycloalkyl radical, C 3-8 A heterocyclic group,
Figure BDA0003866228850000122
Figure BDA0003866228850000131
Wherein Z 1 、Z 2 And Z 3 Each independently is-CH-, or N; and
wherein W is optionally substituted with 0, 1, 2, 3 or 4R 4 And (4) substitution.
In some embodiments, the compound of formula (II) has the structure of formula (IIa):
Figure BDA0003866228850000132
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof;
wherein m is 0, 1, 2 or 3.
In some embodiments of formula (II) or (IIa), R 1 Is that
Figure BDA0003866228850000133
Figure BDA0003866228850000134
Figure BDA0003866228850000135
Wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 And (4) substitution.
In some embodiments of formula (II) or (IIa), R 4 At each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NH 2 、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, C 1-3 Hydroxyalkyl radical, C 1-3 Aminoalkyl radical, C 1-3 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Cycloalkyl, phenyl, or heteroaryl of 5 to 6 atoms; wherein R is 4 Each occurrence is independently optionally substituted with 0, 1, 2, 3, 4 or 5 independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy and C 1-3 Haloalkyl groups.
In some embodiments of formula (II) or (IIa), R 4 At each occurrence, is independently R 4 At each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NH 2 Methyl, ethyl, isopropyl, methoxy, ethoxy, fluoromethyl, -CD 3 、-CHD 2 、-CH 2 D. Cyclopropyl, phenyl, or 5-6 atom heteroaryl; wherein each R 4 Independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy and C 1-3 Haloalkyl groups.
In some embodiments of formula (II) or (IIa), R 5a is-C (= O) R 7 、C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl radical、C 1-4 Alkoxy radical, C 1-4 Haloalkyl, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl radical, C 1-4 Aminoalkyl radical, C 1-4 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of the-C (= O) R 7 、C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, C 1-4 Alkoxy radical, C 1-4 Haloalkyl, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl radical, C 1-4 Aminoalkyl radical, C 1-4 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Cycloalkyl radical, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Alkoxy and C 1-4 Haloalkyl groups.
In some embodiments of formula (II) or (IIa), R 5a Is methyl, ethyl, isopropyl, cyclopropyl, -CF 3 、-CH 2 CH 2 OH、-C(=O)CH 3 、-C(=O)CH 2 CH 3 or-CH 2 CH 2 OCH 3
In some embodiments of formula (I), (Ia), (Ib), (II) or (IIa), R 3 Is cyclopropyl, pyridyl, or phenyl; wherein R is 3 Optionally substituted by 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Deuterated alkyl and C 1-3 Haloalkyl groups.
In some embodiments of formula (I), (Ia), (Ib), (II) or (IIa), R 5 And R 6 At each occurrence, independently is H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、C 1-3 Alkyl radical, C 1-6 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl radical, C 3-6 Cycloalkyl, phenyl, or heteroaryl of 5 to 6 atoms; wherein each of said-NR e R f 、-C(=O)R 7 、C 1-3 Alkyl radical, C 1-6 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl radical, C 3-6 Cycloalkyl, phenyl and 5-6 atom consisting of heteroaryl are independently optionally substituted by 0, 1, 2, 3, 4 or 5 atoms independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy and C 1-3 Haloalkyl groups.
In some embodiments of formula (I), (Ia), (Ib), (II) or (IIa), R 5 And R 6 At each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f Methyl, ethyl, isopropyl, cyclopropyl, -CD 3 、-OCD 3 、-CF 3 、-CH 2 CH 2 OH、-C(=O)CH 3 、-C(=O)CH 2 CH 3 or-CH 2 CH 2 OCH 3
In some embodiments of formula (I), (Ia), (II), or (IIa), R a 、R b And R c Each independently is H, D, F, -CN, -NO 2 、-NR e R f Methyl, ethyl, isopropyl, -CD 3 、-CHD 2 、-CH 2 D. Methoxy, ethoxy, halomethyl, haloethyl, or-CH 2 CH 2 OH。
In some embodiments of formula (I), (Ia), (Ib), (II) or (IIa), R e 、R f 、R 7 And R 7a At each occurrence, independently H, D, C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl radical, C 3-6 Cycloalkyl radical, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 3-6 Heterocyclic group, C 3-6 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-4 An alkyl group; wherein each C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl radical, C 3-6 Cycloalkyl, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 3-6 Heterocyclic group, C 3-6 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-4 Alkyl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Alkoxy and C 1-4 Haloalkyl groups.
In some embodiments of formula (I), (Ia), (Ib), (II) or (IIa), R e 、R f 、R 7 And R 7a At each occurrence, independently is H, D, C 1-2 Alkyl radical, C 1-2 Deuterated alkyl, C 1-2 Haloalkyl, phenyl, or cyclopropyl.
In some embodiments of formula (I), (Ia) or (Ib), the compound is a compound having one of the following structures:
Figure BDA0003866228850000141
/>
Figure BDA0003866228850000151
/>
Figure BDA0003866228850000161
Or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof.
In some embodiments of formula (II) or (IIa), the compound is a compound having one of the following structures:
Figure BDA0003866228850000162
/>
Figure BDA0003866228850000171
or a stereoisomer, tautomer, nitroxide, solvate, or pharmaceutically acceptable salt thereof.
Unless otherwise indicated, stereoisomers, tautomers, solvates, metabolites or pharmaceutically acceptable salts of the compounds of formula (I), (Ia), (Ib), (II) or (IIa) are included within the scope of the present invention.
The compounds of the present disclosure may contain asymmetric or chiral centers and thus may exist in different stereoisomeric forms. The present invention contemplates that all stereoisomeric forms of the compounds of formula (I), (Ia), (Ib), (II) or (IIa), including but not limited to diastereomers, enantiomers, atropisomers and geometric (or conformational) isomers, and mixtures thereof, such as racemic mixtures, are integral to the present invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not specified, then all stereoisomers of that structure are contemplated as within this invention and are included as disclosed compounds in this invention. When stereochemistry is indicated by a solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of the structure are so well-defined and defined.
The compounds of formula (I), (Ia), (Ib), (II) or (IIa) may be present in the form of salts. In one embodiment, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith. In another embodiment, the salt need not be a pharmaceutically acceptable salt, and may be an intermediate useful in the preparation and/or purification of a compound of formula (I), (Ia), (Ib), (II) or (IIa) and/or in the isolation of the enantiomers of a compound of formula (I), (Ia), (II) or (IIa).
In another aspect, the invention relates to intermediates for the preparation of compounds of formula (I), (Ia), (Ib), (II) or (IIa).
In another aspect, the invention relates to a process for the preparation, isolation and purification of a compound of formula (I), (Ia), (Ib), (II) or (IIa).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention. In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable excipient, diluent or carrier, or a combination thereof. In another embodiment, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.
In some embodiments, the pharmaceutical compositions of the present invention further comprise an additional therapeutic agent.
In another aspect, the invention relates to the use of a compound of the invention or a pharmaceutical composition of the invention in the manufacture of a medicament for preventing, treating, and/or alleviating a PI 3-kinase mediated disease, disorder, and/or condition, or inhibiting PI 3-kinase activity.
In some embodiments, the PI 3-kinase mediated disease, disorder, and/or condition is selected from a respiratory disease, viral infection, non-viral respiratory infection, allergic disease, autoimmune disease, inflammatory disease, cardiovascular disease, hematologic malignancy, neurodegenerative disease, pancreatitis, multi-organ failure, kidney disease, platelet aggregation, cancer, sperm motility, transplant rejection, graft rejection, lung injury, or pain.
In some embodiments, the PI 3-kinase mediated disease, disorder, and/or condition is selected from asthma, chronic Obstructive Pulmonary Disease (COPD), viral respiratory infection, viral respiratory disease exacerbations, aspergillosis, leishmaniasis, allergic rhinitis, allergic dermatitis, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, thrombosis, atherosclerosis, hematological malignancies, neurodegenerative disease, pancreatitis, multiorgan failure, renal disease, platelet aggregation, cancer, sperm motility, transplant rejection, graft rejection, lung injury, pain associated with rheumatoid arthritis or osteoarthritis, back pain, systemic inflammatory pain, posthepatic neuralgia, diabetic neuropathy, inflammatory neuropathic pain (trauma), trigeminal neuralgia or central pain.
In some embodiments, the cancer is selected from acute myelogenous leukemia, myelodysplastic syndrome, myeloproliferative disorders, chronic myelogenous leukemia, T-cell acute lymphocytic leukemia, B-cell acute lymphocytic leukemia, non-hodgkin's lymphoma, B-cell lymphoma, solid tumors, or breast cancer.
In some embodiments, wherein the PI 3-kinase is PI3K- γ.
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The present invention provides a pharmaceutical composition comprising a compound disclosed herein, or a compound listed in the examples, or a stereoisomer, tautomer, nitroxide, solvate, metabolite or pharmaceutically acceptable salt thereof; and pharmaceutically acceptable adjuvants, diluents, carriers, vehicles or combinations thereof. The amount of compound in the pharmaceutical compositions disclosed herein is an amount effective to detectably inhibit a protein kinase in a biological sample or patient.
It will also be appreciated that certain compounds of the invention may be present in free form for use in therapy or, if appropriate, in the form of a pharmaceutically acceptable derivative thereof. Some non-limiting embodiments of pharmaceutically acceptable derivatives include pharmaceutically acceptable salts, esters, salts of such esters, or any additional adduct or derivative that upon administration to a patient in need thereof provides, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
Various carriers for formulating pharmaceutically acceptable compositions, and well known techniques for their preparation, are disclosed in, for example, remington, the Science and Practice of Pharmacy,21st edition,2005, ed.D.B.Troy, lippincott Williams and Wilkins, philadelphia, and Encyclopedia of Pharmaceutical technology, eds.J.Swarbrick and J.C.Boylan,1988-1999, marcel Dekker, new York, the contents of each of which are incorporated herein by reference. Except insofar as any conventional carrier is incompatible with the disclosed compounds, such as by producing any undesirable biological effect or interacting in a deleterious manner with any other ingredient in a pharmaceutically acceptable composition, its use is contemplated as falling within the scope of the present invention.
The pharmaceutical compositions provided by the present invention may be co-formulated with other active ingredients that do not impair the intended therapeutic effect, or with substances that supplement the intended effect.
Use of the Compounds and compositions of the invention
The compounds of the invention are inhibitors of kinase activity, particularly PI 3-kinase activity. Compounds that are inhibitors of PI 3-kinase may be useful in the treatment of disorders in which the underlying pathology is (at least in part) due to inappropriate PI 3-kinase activity, such as asthma, chronic Obstructive Pulmonary Disease (COPD), viral infections, non-viral respiratory infections, allergic diseases, autoimmune diseases, inflammatory diseases, cardiovascular diseases, hematological malignancies, neurodegenerative diseases, pancreatitis, multi-organ failure, kidney disease, platelet aggregation, cancer, sperm motility, transplant rejection, graft rejection, lung injury or pain, and the like. By "inappropriate PI 3-kinase activity" is meant any PI 3-kinase activity that deviates from the normal PI 3-kinase activity expected in a particular patient. Inappropriate PI 3-kinases may take the form of, for example, an abnormal increase in activity, or an aberration in PI 3-kinase or dysregulation. These inappropriate activities may result, for example, from overexpression or mutation of protein kinases that result in inappropriate or uncontrolled activation. Thus, in another aspect, the invention relates to a method of treating said disease or disorder.
Such diseases or conditions include, but are not limited to, respiratory diseases including asthma, chronic obstructive pulmonary disease, and Idiopathic Pulmonary Fibrosis (IPF); viral infections, including viral respiratory infections and viral exacerbations of respiratory diseases, such as asthma and COPD; non-viral respiratory infections including aspergillosis and leishmaniasis; allergic diseases including allergic rhinitis and atopic dermatitis; autoimmune diseases, including rheumatoid arthritis and multiple sclerosis; inflammatory diseases, including inflammatory bowel disease; cardiovascular diseases including thrombosis and atherosclerosis; hematological malignancies; neurodegenerative diseases; pancreatitis; multiple organ failure; kidney disease; platelet aggregation; cancer; sperm motility; transplant rejection; graft rejection; lung injury; and pain, including pain associated with rheumatoid arthritis or osteoarthritis, back pain, systemic inflammatory pain, posthepatic neuralgia, diabetic neuropathy, inflammatory neuropathic pain (trauma), trigeminal neuralgia and central pain. In one embodiment, such disorders include, respiratory diseases including asthma and Chronic Obstructive Pulmonary Disease (COPD); allergic diseases including allergic rhinitis and atopic dermatitis; autoimmune diseases, including rheumatoid arthritis and multiple sclerosis; inflammatory diseases, including inflammatory bowel disease; cardiovascular diseases including thrombosis and atherosclerosis; hematological malignancies; neurodegenerative diseases; pancreatitis; multiple organ failure; kidney disease; platelet aggregation; cancer; sperm motility; transplant rejection; graft rejection; lung injury; and pain, including pain associated with rheumatoid arthritis or osteoarthritis, back pain, systemic inflammatory pain, posthepatic neuralgia, diabetic neuropathy, inflammatory neuropathic pain (trauma), trigeminal neuralgia, and central pain.
In such diseases or conditions, the cancer is selected from acute myelogenous leukemia, myelodysplastic syndrome, myeloproliferative disorders, chronic myelogenous leukemia, T-cell acute lymphocytic leukemia, B-cell acute lymphocytic leukemia, non-hodgkin's lymphoma, B-cell lymphoma, solid tumors, or breast cancer.
The treatment methods of the present invention comprise administering to a patient in need thereof a safe and effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. Various embodiments of the present invention include methods of treating any one of the disorders or diseases mentioned herein by administering to a patient in need thereof a safe and effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
Combination therapy
The compounds of the present invention may be administered as the sole active agent or may be administered in combination with other therapeutic agents, including other compounds that have the same or similar therapeutic activity and are identified as safe and effective for such combination administration.
In one aspect, the invention provides a method of treating, preventing or ameliorating a disease or disorder, comprising administering a safe and effective amount of a combination comprising a compound disclosed herein and one or more therapeutically active agents. In some embodiments, the combination comprises one or two additional therapeutic agents.
Examples of other therapeutic agents include, but are not limited to: anti-cancer agents, including chemotherapeutic agents and antiproliferative agents; an anti-inflammatory agent; and an immunomodulator or immunosuppressant.
In another aspect, the invention provides a product comprising a compound of the invention and at least one other therapeutic agent, formulated for simultaneous, separate or sequential administration in therapy. In some embodiments, the treatment is directed to treatment of a disease or condition mediated by one or more protein kinase, such as PI3 k-kinase, activity. The combined preparation provides products including compositions comprising the disclosed compounds and other therapeutic agents in the same pharmaceutical composition, or in different forms, e.g., kits.
In another aspect, the invention provides a pharmaceutical composition comprising a compound disclosed herein and one or more additional therapeutic agents. In some embodiments, the pharmaceutical composition may comprise a pharmaceutically acceptable excipient as described above.
In another aspect, the invention provides a kit comprising two or more separate pharmaceutical compositions, wherein at least one pharmaceutical composition comprises a compound disclosed herein. In some embodiments, the kit comprises means for separately holding the compositions, such as a container, a separate bottle, or a separate foil box. An example of such a kit is a blister pack, which is commonly used for packaging tablets, capsules and the like.
The compounds disclosed herein may be administered as a single active ingredient or as, for example, an adjuvant, co-administered with other therapeutic agents.
In some embodiments, the additional therapeutic agent includes, for example, an immunosuppressive, immunomodulatory or other anti-inflammatory agent, a drug for treating or preventing allo-or xenotransplant acute or chronic rejection, or inflammation, or an autoimmune disease, or a chemotherapeutic agent, such as a malignant cell antiproliferative agent.
The compounds of formula (I) of the present invention are co-administered with other immunosuppressive/immunomodulatory, anti-inflammatory, chemotherapeutic or anti-infective agents, wherein the dosage of the immunosuppressive/immunomodulatory, anti-inflammatory, chemotherapeutic or anti-infective agent co-administered depends on the type of co-administration, whether it is a steroid or a calcineurin inhibitor, and the particular drug being used for treatment and the condition being treated, etc.
Examples of anti-inflammatory agents include non-steroidal anti-inflammatory drugs (NSAIDs). Examples of NSAIDs include cromolyn sodium, nedocromil sodium (sodium), phosphodiesterase (PDE) inhibitors (such as theophylline, PDE4 inhibitors, or mixed PDE3/PDE4 inhibitors), leukotriene antagonists, leukotriene synthesis inhibitors (such as montelukast), iNOS inhibitors, trypsin and elastase inhibitors, beta-2 integrin antagonists and adenosine receptor agonists or antagonists (such as adenosine 2 alpha receptor agonists), cytokine antagonists (such as chemokine receptor antagonists, including CCR3 antagonists), cytokine synthesis inhibitors, or 5-lipoxygenase inhibitors.
The compounds of formula (I) may also be advantageously employed in combination with other compounds, or in combination with other therapeutic agents, especially antiproliferative agents. Such antiproliferative agents include, but are not limited to, aromatase inhibitors; an antiestrogen; a topoisomerase I inhibitor; a topoisomerase II inhibitor; a microtubule active agent; an alkylating agent; (ii) histone deacetylase inhibitors; compounds that induce a cellular differentiation process; a cyclooxygenase inhibitor; an MMP inhibitor; an mTOR inhibitor; an antineoplastic antimetabolite; a platinum compound; compounds that target/reduce protein or lipid kinase activity and other anti-angiogenic compounds; a compound that targets, reduces or inhibits protein or lipid phosphatase activity; gonadorelin agonists; an antiandrogen; methionine aminopeptidase inhibitors; a bisphosphonate; a biological response modifier; an anti-proliferative antibody; heparanase inhibitors; ras oncogenic subtype inhibitors; a telomerase inhibitor; a proteasome inhibitor; agents for treating hematological tumors; compounds that target, decrease or inhibit Flt-3 activity; an Hsp90 inhibitor; temozolomide and calcium folinate.
"combination" means a fixed combination or a kit of parts for the combined administration in the form of a single dosage unit, in which a compound disclosed in the invention and a combination partner may be administered separately at the same time or may be administered separately at certain time intervals, in particular such that the combination partners show a cooperative, e.g. synergistic, effect. The terms "co-administration" or "co-administration" and the like as used herein are intended to encompass administration of the selected combination partners to a single individual in need thereof (e.g., a patient), and are intended to encompass treatment regimens in which the substances are not necessarily administered by the same route of administration or simultaneously.
Method of treatment
In some embodiments, the presently disclosed methods of treatment comprise administering to a patient in need thereof a safe and effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the present invention. Various embodiments disclosed herein include methods of treating a disease or condition described herein by administering to a patient in need thereof a safe and effective amount of a disclosed compound or a pharmaceutical composition comprising a disclosed compound.
In some embodiments, a disclosed compound or pharmaceutical composition comprising a disclosed compound may be administered once or several times at different time intervals over a specified period of time according to a dosing regimen. For example, once, twice, three times or four times daily. In one embodiment, the administration is once daily. In yet another embodiment, the administration is twice daily. The administration may be carried out until the desired therapeutic effect is achieved or the desired therapeutic effect is maintained indefinitely. Suitable dosing regimens for the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds depend on the pharmacokinetic properties of the compounds, such as dilution, distribution and half-life, which can be determined by the skilled person. In addition, suitable dosing regimens for a compound or pharmaceutical composition comprising a compound disclosed herein, including the duration of administration of the regimen, will depend upon the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient being treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and experience of the skilled artisan. It will also be appreciated by those skilled in the art that adjustment of an appropriate dosage regimen may be required for the individual patient's response to the dosage regimen, or as the individual patient needs to change over time.
The pharmaceutical composition or combination/association of the invention may be a unit dose of about 1-1000mg of the active ingredient, or about 1-500mg or about 1-250mg or about 1-150mg or about 0.5-100mg, or about 1-50mg of the active ingredient for about 50-70kg of an individual. The therapeutically effective dose of the compound, pharmaceutical composition, or combination thereof, depends on the species, weight, age of the individual, and the severity of the individual's disease, disorder, or condition or the treatment to be treated. A physician, clinician or veterinarian of ordinary skill in the art can readily determine the effective amount of each active ingredient to prevent, treat or inhibit the progression of the disease or condition. The above cited dose profiles have been demonstrated in vitro and in vivo experiments with beneficial mammals, such as mice, rats, dogs, monkeys, or isolated organs, tissues and specimens thereof. The compounds of the invention can be used in vitro in the form of solutions, for example aqueous solutions, and in vivo in the form of suspensions or aqueous solutions, enterally, parenterally and intravenously, where appropriate. The therapeutically effective amount in vivo ranges from about 0.01 to about 500mg/kg, or from about 1 to about 100mg/kg, depending on the route of administration.
The presently disclosed compounds may be administered simultaneously, or before or after, one or more other therapeutic agents. The compounds of the invention may be administered separately from the other therapeutic agents, by the same or different routes of administration, or in the same pharmaceutical composition.
General synthetic schemes
To describe the invention, examples are set forth below. It is to be understood that the invention is not limited to these embodiments, but is provided as a means of practicing the invention.
In general, the compounds of the invention can be prepared by the processes described in the present invention, where the substituents are as defined for formula (I), (Ia), (Ib), (II) or (IIa), unless otherwise specified. The following reaction schemes and examples serve to further illustrate the context of the invention.
The examples described below, unless otherwise indicated, all temperatures are set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, alfa Chemical Company, shanghai Shao Yuan reagents, inc., SAIN Chemical technology (Shanghai) Inc., shanghai Bye pharmaceutical technology Inc., shanghai Macro biomedical technology Inc., unless otherwise indicated, the reagents used in the present invention were used without further purification. The solvent is commercially available from commercial suppliers such as VISCO technologies, inc., of Beijing Hai.
Anhydrous THF, dioxane, DCM, toluene and DMF were all purchased from commercial suppliers such as the Energy chemical company and Aldrich chemical company. EtOAc, PE, CH 3 CN, NMP and DMSO were treated with anhydrous Na before use 2 SO 4 And (6) processing.
The following reactions are generally carried out under a positive pressure of nitrogen or argon or by placing a drying tube over an anhydrous solvent (unless otherwise indicated), the reaction vial is stoppered with a suitable rubber stopper and the substrate is driven in by syringe. The glassware was dried.
The column chromatography is performed using a silica gel column. Silica gel (300-400 mesh) was purchased from Qingdao oceanic plants.
1 H NMR spectrum of 13 The C/2D data are at BrukerCollected at 400MHz on Avance III. 1 H NMR spectrum with CDC1 3 、DMSO-d 6 、CD 3 OD or acetone-d 6 TMS (0 ppm) or chloroform (7.26 ppm) was used as a reference standard for the solvent (in ppm). When multiple peaks occur, the following abbreviations will be used: s (singlets), d (doublets), t (triplets), m (multiplets), br (broad singlets), dd (doublets of doublets), dt (doublets of triplets). Coupling constants are expressed in hertz (Hz).
The LC/MS was performed on an Agilent 1260 (binary Pump/DAD detector) coupled to an Agilent 6120/6125 mass spectrometer.
The method comprises the following steps:
column: HALO C18.7 μm,4.6mm × 30mm, mobile phase: meCN (0.05% hcooh) -Water (0.05% hcooh); gradient: eluting with 5-95% MeCN for 0.8min, and maintaining for 0.8min, wherein the total running time is 2.0min; flow rate: 1.8mL/min; column temperature: 45 ℃;
The method 2 comprises the following steps:
column: HALO C18.7 μm,4.6mm × 50mm, mobile phase: meCN (0.025% trifluoroacetic acid) -water (0.025% trifluoroacetic acid); gradient: eluting from 5% to 95% MeCN for 1.0min, keeping for 1.0min, and keeping the total running time for 2.5min; flow rate: 1.8mL/min; column temperature: at 45 ℃.
Purity testing by RP-HPLC:
compound purity testing on RP-HPLC (Shimadzu 2010/2030)
The method comprises the following steps:
column: gemini 4.6 multiplied by 150mm 5um; mobile phase: h 2 O (0.05% trifluoroacetic acid) -MeCN (0.05% trifluoroacetic acid). Gradient: from 10% to 100% MeCN, elute 8 minutes, hold 2 minutes. Flow rate: 1.2mL/min, column temperature: 35 ℃/40 ℃. The method 2 comprises the following steps:
column: XBRIDGE 2.1X 50mm,3.5um; mobile phase: h 2 O (0.05% trifluoroacetic acid) -MeCN (0.05% trifluoroacetic acid). Gradient: from 10% to 100% MeCN, elute 7 min, hold 1 min. Flow rate: 0.8mL/min, column temperature: at 35 deg.C/40 deg.C. Conversion by SFCAnd (3) purifying the compound:
SFC purification was performed on a THar P80 equipped with a UV detector.
The method comprises the following steps: column CHIRALPAK AD-H250mm, 20mm,5 μm, modifier: 30% EtOH (0.2% NH) 4 OH)。
Compound purification by RP-HPLC:
RP-HPLC purification was performed on a Gilson purification system (322 or 306 pump and GX-281 fraction collector), a Shimadzu LC20Ap and Waters MS trigger purification system;
The method comprises the following steps:
column Gemini C18 21x150 mm,5 μm Xbridge C18 19x150 mm,5 μm, spolar C18 20x150 mm and Ultimate AQ-C18 30x250 mm,10 μm
Mobile phase:
aqueous solution of mecn (0.1% hcooh), flow rate: 20ml/min,50ml/min, column 30x250 mm,10 μm; wavelength: 210-400nm. Samples were injected in DMSO (+ optionally formic acid and water) and eluted for 10 minutes from 10% to 95% linear gradient of mecn.
Aqueous solution of mecn (0.1% trifluoroacetic acid), flow rate: 20ml/min,50ml/min, column 30x250 mm,10 μm; wavelength: 210-400nm. Samples were injected in DMSO (+ optionally formic acid and water) and eluted for 10 minutes from 10% to 95% linear gradient of mecn.
Aqueous solution of MeCN (0.1% NH) 3 -H 2 O/10mM NH 4 AC), flow rate: 20ml/min,50ml/min, column 30x250 mm,10 μm; wavelength: 210-400nm. Samples were injected in DMSO (+ optionally formic acid and water) and eluted for 10 minutes from 10% to 95% linear gradient of mecn.
Typical synthetic procedures for preparing the disclosed compounds are shown in the following synthetic schemes. Unless otherwise indicated, R 1 、R 2 、R 3 、R 4a 、R 4b 、R 4c 、R a 、R b 、X、Z 1 、Z 2 W and n have the definitions as defined in the present invention.
Synthesis scheme 1:
Figure BDA0003866228850000221
the compounds of the present invention having the structure represented by the formula (6) can be prepared by a general synthetic method described in the synthetic scheme 1, and specific steps can be referred to examples. In FIG. 1, compound (1) can be obtained by commercially available reagents or by synthesis from the reference (WO 2016149160A 1). Compound (1) and compound having R 1 Coupling of the substituted terminal alkynes under catalysis of appropriate palladium and copper salts affords compound (3). The Boc protecting group is removed under acidic conditions to give a derivative of the compound (4). The compound (4) and the compound (5) react under appropriate basic conditions or in the presence of a condensing agent to obtain the target kinase inhibitor (6).
Synthesis scheme 2:
Figure BDA0003866228850000222
the compounds of the present invention having the structure represented by the formula (8) can be prepared by a general synthetic method described in the synthesis scheme 2, and specific procedures can be referred to examples. In scheme 2, amine derivative (4) and carboxylic acid derivative (7) are condensed in the presence of a suitable condensing agent to give the target kinase inhibitor (8).
Fragment synthesis scheme 1:
Figure BDA0003866228850000231
the alkyne derivative having the structure shown in formula (2) can be prepared by the general synthetic method described in the fragment synthesis scheme 1, and the specific steps can be referred to in the examples. In FIG. 2, compound (2-a) is coupled with silicon trimethylacetylene under suitable basic conditions (e.g., potassium carbonate, cesium carbonate, potassium phosphate, etc.) under catalysis of a suitable palladium salt and a copper salt to give compound (2-b). And removing the TMS protecting group by TBAF to obtain the alkyne derivative (2). Or using R 1 Substituted aldehyde derivative (2-c) under suitable basic conditions, and (1-diazo-2-oxopropyl) phosphonic acid dimethyl ester The acetylene derivative (2) is obtained by reaction at low temperature.
Fragment synthesis scheme 2:
Figure BDA0003866228850000232
the carboxylic acid derivatives having the structure shown in formula (5) can be prepared by the general synthetic method described in the fragment synthesis scheme 2, and the specific steps can be referred to the examples. In FIG. 3, the compound (5-a) is subjected to deethylation under suitable basic conditions to give a carboxylic acid derivative (5-b). The compound (5-b) and the compound (4) are condensed in the presence of a condensing agent (such as EDCI or HATU) to obtain the target kinase inhibitor (6). The compound (5-b) and N-hydroxysuccinimide are condensed in the presence of a condensing agent to obtain a compound (5-c). Under appropriate alkaline conditions, the compound (5-c) and the compound (4) react to obtain the target kinase inhibitor (6).
Deuterated fragment example 1:
the building blocks with deuterium used for synthesizing the compounds of the present invention include commercial reagents and are synthesized by reference (e.g. WO2017161116 A1), including but not limited to the following structures:
Figure BDA0003866228850000233
examples
Intermediate 1, 5-dioxopyrrol-1-yl 2-aminopyrazolo [1,5-a ] pyrimidine-3-carboxylic acid ester-5, 7-D2
Figure BDA0003866228850000241
Step 1) 2-Aminopyrazolo [1,5-a ] pyrimidine-3-carboxylic acid ethyl ester-5, 7-D2
To a solution of ethyl 3, 5-diamino-1H-pyrazole-4-carboxylate (1g, 5.9mmol) in acetic acid (30 mL) was added 1, 3-tetraethoxypropane-1, 3-D 2 (1313.3mg, 5.9mmol). The mixture is warmed to 120 ℃ and stirred for 16h, cooled to room temperature, concentrated under reduced pressure, and the residueDiluting with acetone (30 mL), and filtering with suction to obtain 2-aminopyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester-5, 7-D2 (1.2g, 93.21% yield). MS (ESI) 209.1[ 2 ] M + H] +
Step 2) 2-Aminopyrazolo [1,5-a ] pyrimidine-3-carboxylic acid-5, 7-D2
To 2-aminopyrazolo [1,5-a ]]Ethyl pyrimidine-3-carboxylate (1.2g, 5.8mmol) in MeOH/H 2 To the O (50 mL/10 mL) solution was added LiOH (1.22g, 29mmol). The mixture was stirred at 60 ℃ for 4h, the solvent was removed under reduced pressure and the residue was adjusted to pH 5 with 1N HCl. Filtering, drying the filter residue to obtain 2-amino pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid-5,7-D2 (785mg, 72.41% yield) was a yellow solid. MS (ESI) 181.0[ m ] +H] +
Step 3) 2, 5-dioxopyrrol-1-yl 2-aminopyrazolo [1,5-a ] pyrimidine-3-carboxylic acid ester-5, 7-D2
EDCI (279.8mg, 1.46mmol) and 1-hydroxypyrrolidine-2, 5-dione (168mg, 1.46mmol) were added to 2-aminopyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (200mg, 1.12mmol) in DMF (10 mL) and the mixture was stirred at room temperature for 16h. After completion of the reaction, it was diluted with water (30 ml) and extracted three times with ethyl acetate (30mL × 3). The combined organic phases were washed with brine (40mL. Times.2), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give 2, 5-dioxopyrrolidin-1-yl 2-aminopyrazolo [1,5-a ] ]Pyrimidine-3-carboxylate-5, 7-D2 (238mg, 69.32% yield) was a yellow solid. MS (ESI) 277.9[ deg. ] M + H ]] +
Intermediate 2 (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinoline-1 (2H) -one
Figure BDA0003866228850000242
Step 1) (S) - (1- (methoxy (methyl) amino) -1-oxopropan-2-yl) carbamic acid tert-butyl ester
To a solution of (tert-butoxycarbonyl) -L-alanine (8.0 g, 0.042mol) in DCM (200 mL) was added CDI (8.92g, 0.055mmol). The mixture was stirred at room temperature for 1h, then N, O-dimethylhydroxylamine (4.95g, 0.051mol) and TEA (17.1 g, 0.17mol) were added and the mixture stirred at 25 ℃ for 16h. From the mixture with H 2 O(20mL) and then extracted with DCM (50 mL × 3). The combined organic layers were washed with brine (40mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by flash chromatography (DCM/MeOH = 98/2) to give tert-butyl (S) - (1- (methoxy (methyl) amino) -1-oxopropan-2-yl) carbamate (6.8g, 62.17% yield) as a white solid. MS (ESI) 254.9[ 2 ], [ M ] +Na] +
Step 2) 2-chloro-6-methyl-N-phenylbenzamide
Oxalyl chloride (4.83g, 0.038mol) was added dropwise to a solution of 2-chloro-6-methylbenzoic acid (5.0 g, 0.029mol) and DMF (210mg, 0.299mol) in DCM (50 mL) at 0 deg.C, and the mixture was stirred at 0 deg.C for 2h. The reaction mixture was added to a solution of aniline (2.73g, 0.029 mol) and TEA (8.88g, 0.088mol) in DCM (50 mL) at 0 deg.C. The mixture was stirred at room temperature for 5 hours. For mixtures H 2 O (30 mL) was diluted and extracted with DCM (50 mL. Times.3). The combined organic layers were washed with brine (40mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by flash chromatography (PE/EA = 3/1) to give 2-chloro-6-methyl-N-phenylbenzamide (5.66g, 70.65% yield) as a yellow solid. MS (ESI) 245.9[ 2 ] M + H] +
Step 3) (S) - (4- (3-chloro-2- (phenylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamic acid tert-butyl ester
To a stirred solution of 2-chloro-6-methyl-N-phenylbenzamide (1g, 4.1mmol) in THF (20 mL) at-30 deg.C under nitrogen was added dropwise a solution of N-BuLi (4.3mL, 10.2mmol) over 10 minutes, and the reaction mixture was stirred at-30 deg.C for 1 hour. To a stirred solution of tert-butyl (S) - (1- (methoxy (methyl) amino) -1-oxopropan-2-yl) carbamate (1.43g, 6.15mmol) in THF (20 mL) at-30 ℃ under nitrogen was added dropwise a solution of i-PrMgCl (9.5mL, 12.3mmol) over 10min and the reaction mixture was stirred at-30 ℃ for 1h. The resulting solution was then slowly added to the reaction mixture above. The mixture was stirred at room temperature for 3 hours. NH for the mixture 4 Cl (30 mL) was quenched and then extracted with EtOAc (30mL. Times.3). The combined organic layers were washed with brine (10mL. Times.2), and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by flash chromatography (PE/EA = 3/1) Tert-butyl (S) - (4- (3-chloro-2- (phenylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamate (1.45g, 75.61% yield) was obtained as a yellow solid. MS (ESI) 438.8[ m ] +Na] +
Step 4) (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinoline-1 (2H) -one
To a solution of (S) - (4- (3-chloro-2- (phenylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamic acid tert-butyl ester (1.4 g,3.4 mmol) in MeOH (30 mL) at room temperature was added concentrated hydrochloric acid (2.8 mL) and the mixture was stirred at 80 ℃ for 2 h. The mixture is concentrated under reduced pressure and the residue is taken up in H 2 O (10 mL) diluted and NaHCO 3 The pH of the solution is adjusted to 7-8. The mixture was extracted with DCM (30mL. Times.3), and the combined organic layers were washed with brine (20mL. Times.2), over Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by flash chromatography (DCM/MeOH = 95/5) to give (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinoline-1 (2H) -one (830mg, 73.53% yield) as a yellow solid. MS (ESI) 298.9[ 2 ] M + H] +
Intermediate 3-amino-5-methylpyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
Figure BDA0003866228850000251
/>
Step 1) Ethyl 2-amino-5-methylpyrazolo [1,5-a ] pyrimidine-3-carboxylate
To a solution of ethyl 3, 5-diamino-1H-pyrazole-4-carboxylate (1g, 6.0 mmol) in DMF (10 mL) was added 4, 4-dimethoxy-2-butanone (1.6 g,12.0 mmol), and the reaction was heated at 110 ℃ for 1 hour, followed by addition of CH 3 COOH (361mg, 6.0 mmol), and the reaction was heated for 2 hours. After cooling to room temperature, the mixture was neutralized with saturated sodium bicarbonate and diluted with water (30 mL). The mixture was extracted with EtOAc (40mL. Times.3). The combined organic layers were washed with brine (60mL. Times.2) and over anhydrous Na 2 SO 4 Drying, filtering and concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 2-amino-5-methylpyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (940 mg, 71%) was a white solid. MS (ESI) m/z 224.1[ 2 ] M + H] +
Step 2) 2-amino-5-methylpyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
To 2-amino-5-methylpyrazolo [1,5-a ]]Ethyl pyrimidine-3-carboxylate (940mg, 4.3mmol) in MeOH/H 2 NaOH (854 mg,21.4 mmol) was added to the O (20 mL/4 mL) solution, and the mixture was stirred at 60 ℃ for 16 hours. The mixture was concentrated under reduced pressure and the residue was diluted with water (20 mL). The mixture was acidified with 1N HCl to pH =3-4, and then the mixture was extracted with EtOAc (200 mL). The separated organic layer was washed with brine (100 mL) and Na 2 SO 4 Drying and vacuum concentrating to obtain 2-amino-5-methylpyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (450mg, 53%) as a yellow solid. MS (ESI) m/z 196.0[ m + H ]] +
Example 1 (S) -2-amino-N- (1- (8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-5, 7-D2-3-carboxamide
Figure BDA0003866228850000252
Step 1) Nitroproline
To pyrrolidine-2-carboxylic acid and NaNO at 0 deg.C 2 (4.19g, 0.061mmol) in water (10 mL) was added concentrated hydrochloric acid (5 mL). After the mixture was stirred at room temperature for 16h, it was diluted with water (20 mL) and extracted three times with ethyl acetate (30mL. Times.3). The combined organic phases were washed with brine (40mL. Times.2), dried over anhydrous sodium sulfate, and concentrated to remove the solvent to give nitrosoproline (5.5g, 83% yield) as a white solid. MS (ESI) 145.1[ m ] +H] +1 H NMR(400MHz,DMSO)δ6.50(d,J=8.6Hz,1H),5.81-5.67(m,1H),5.07-4.90(m,2H),4.64-4.54(m,1H),3.44-3.37(m,1H),2.31-2.19(m,1H),2.07-1.96(m,1H),1.41-1.33(m,9H)。
Step 2) 5, 6-dihydro-4H-pyrrolo [1,2-c ] [1,2,3] oxadiazol-7-azonia-3-inner salt
TFAA (3.8g, 0.018mol) was added to a solution of nitrosoproline (2g, 0.014mol) in acetonitrile (6 mL) at 0 ℃ and stirring was continued for 3h. The mixture was quenched with potassium carbonate (3.1g, 0.022mol), diluted with water: (20 mL). The mixture was extracted three times with ethyl acetate (20mL. Times.3). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (DCM/MeOH = 20/1) to give 5, 6-dihydro-4H-pyrrole [1,2-c ]][1,2,3]Oxadiazole-7-azonia-3-inner salt (1.5g, 77% yield) was a brown oil. MS (ESI) 252.8[2M ] +H] +1 H NMR(400MHz,CDCl 3 )δ4.44-4.39(m,2H),2.94-2.90(m,2H),2.81-2.74(m,2H)。
Step 3) Ethyl 5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole-2-carboxylate
Reacting 5, 6-dihydro-4H-pyrrole [1,2-c ] ][1,2,3]A xylene solution (10 mL) of oxadiazole-7-azonia-3-inner salt (0.5g, 4mmol) and methyl prop-2-ynoate (1.18g, 12mmol) was warmed to 140 ℃ and stirred for 16h. Cooled to room temperature and concentrated. The residue was diluted with EtOAc (30 mL) and washed with saturated brine (20 mL), and the organic phase was dried over saturated sodium sulfate and concentrated under reduced pressure. Subjecting the residue to silica gel column chromatography to obtain 5, 6-dihydro-4H-pyrrole [1,2-b ]]Pyrazole-2-carboxylic acid ethyl ester (0.23g, 30% yield) was a brown oil. MS (ESI) 181.1[ 2 ] M + H] +1 H NMR(400MHz,CDCl 3 )δ6.55(s,1H),4.39(q,J=7.2Hz,2H),4.25-4.16(m,2H),2.99-2.87(m,2H),2.69-2.56(m,2H),1.40(t,J=7.2Hz,3H)。
Step 4) (5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-2-yl) methanol
At 0 deg.C, to 5, 6-dihydro-4H-pyrrole [1,2-b ]]To a solution of pyrazole-2-carboxylic acid ethyl ester (230mg, 1.27mmol) in tetrahydrofuran (10 mL) was added LiAlH 4 (48mg, 1.27mmol) and the mixture was kept stirring at 0 ℃ for 2h. The mixture was quenched with 10% aqueous NaOH solution (2 mL) and filtered. Extracting the filtrate with ethyl acetate (20mL x 3) for three times, combining organic phases, drying over anhydrous sodium sulfate, and concentrating under reduced pressure to remove the solvent to obtain (5, 6-dihydro-4H-pyrrole [1,2-b ]]Pyrazol-2-yl) methanol (200mg, 90% yield) was a colorless oil. MS (ESI) 139.1[ 2 ] M + H] +1 H NMR(400MHz,CDCl 3 )δ5.99(s,1H),5.09(s,1H),4.65(d,J=1.6Hz,2H),4.16-4.12(m,2H),2.88(t,J=7.2Hz,2H),2.62–2.58(m,2H)。
Step 5) 5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole-2-carbaldehyde
To (5, 6-dihydro-4H-pyrrolo [1, 2-b) ]MnO was added to a solution of pyrazol-2-yl) methanol (50mg, 0.36mmol) in chloroform (20 mL) 2 (157mg, 1.8 mmol), the mixture was warmed to 60 ℃ and stirred for 16h. Filtering the mixture, and concentrating the filtrate to obtain 5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole-2-carbaldehyde (30mg, 58% yield) was a colorless oil. MS (ESI) 137.1[ deg. ] M + H ]] +1 H NMR(400MHz,CDCl 3 )δ9.92(s,1H),6.53(s,1H),4.23(t,J=7.2Hz,2H),2.99-2.95(m,2H),2.72-2.62(m,2H)。
Step 6) 2-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole
To 5, 6-dihydro-4H-pyrrolo [1,2-b ]]To a solution of pyrazole-2-carbaldehyde (30mg, 0.22mmol) and potassium carbonate (61mg, 0.44mmol) in methanol (5 mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (63mg, 0.33mmol), and the mixture was stirred at room temperature for 16h. The solvent was removed by concentration and the residue was diluted with ethyl acetate (30 mL). The mixture was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, and subjected to silica gel column chromatography to give 2-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole (22mg, 61.8% yield) was a colorless oil. MS (ESI) 133.2[ m ] +H] +1 H NMR(400MHz,CDCl 3 )δ6.19(s,1H),4.28-4.17(m,2H),3.13(s,1H),2.96-2.87(m,2H),2.67-2.60(m,2H)。
Step 7) (S) -3- (1-aminoethyl) -8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-2-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (40mg, 0.13mmol) and 2-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ] under a nitrogen atmosphere ]Pyrazole (21.2mg, 0.16mmol) in acetonitrile (30 mL) was added with X-Phos (31.9mg, 0.067mmol) and Pd 2 (dba) 3 (30.6mg, 0.033mmol) and K 3 PO 4 (85.3mg, 0.40mmol), the gas is displaced three times by evacuation, and the mixture is warmed to 80 ℃ and stirred for 5h. Cooled to room temperature and concentrated under reduced pressure to remove the solvent. The residue was diluted with ethyl acetate (30 mL). The mixture was washed with saturated brine (20 mL), concentrated under reduced pressure to remove the solvent, and the residue was subjected to flash silica gel column chromatography (DCM/MeOH = 91/9) to give (S) -3- (1-aminoethyl) -8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazoles-2-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (60mg, 90.9% yield) is a yellow oil. MS (ESI) 395.1[ 2 ] M + H] +
Step 8) (S) -2-amino-N- (1- (8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-5, 7-D2-3-carboxamide
DIEA (63.8mg, 0.49mmol) was added to (S) -3- (1-aminoethyl) -8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b) at room temperature]Pyrazol-2-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (60mg, 0.16mmol) and 2, 5-dioxopyrrolidin-1-yl 2-aminopyrazolo [1,5-a ]]Pyrimidine-3-carboxylate-5, 7-D2 (59mg, 0.21mmol) in acetonitrile (10 mL), the mixture was warmed to 80 ℃ and stirred for 16h. The mixture was cooled to room temperature, concentrated under reduced pressure to remove the solvent, and diluted with dichloromethane (30 mL). The mixture was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure. The residue was purified by preparative HPLC (Gemini-C18 150x21.2mm,5um ACN- -H2O (0.1% FA) 45-50) to give (S) -2-amino-N- (1- (8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ])) ]Pyrazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-5, 7-D2-3-carboxamide (5.3 mg,5.8% yield) was a white solid. MS (ESI) 556.8[ 2 ] M + H] +1 H NMR(400MHz,DMSO)δ8.93(dd,J=6.7,1.6Hz,1H),8.56(dd,J=4.5,1.6Hz,1H),8.01(d,J=6.7Hz,1H),7.66(d,J=4.5Hz,2H),7.63-7.55(m,2H),7.53-7.46(m,3H),7.41-7.36(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.76(s,1H),6.44(s,2H),6.17(s,1H),4.56(t,J=6.8Hz,1H),4.09-4.02(m,2H),2.82(t,J=7.3Hz,2H),2.54(d,J=7.7Hz,1H),1.35(d,J=6.8Hz,3H)。
Example 2 (S) -2-amino-5-methyl-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000271
Step 1) 1-methyl-4- ((trimethylsilyl) ethynyl) -1H-pyrazole
Trimethylacetylenyl silicon (2.8g, 28.8mmol), cuI (366.24mg, 1.923mmol) and bis (triphenylphosphine) palladium (II) dichloride (1.3g, 1.9mmol) were added to a solution of 4-iodo-1-methylpyrazole (4 g, 19.2mmol) in triethylamine (30 mL), and the mixture was stirred at 90 ℃ for 16h. After cooling to room temperature, the solvent was removed by concentration under reduced pressure. The residue was dissolved in DCM (300 mL), diluted with water (150 mL) and washed with saturated brine (150mL. Times.2). The organic phase was dried over anhydrous sodium sulfate, concentrated and the residue was column chromatographed on silica gel (DCM/MEOH = 10/1) to give 1-methyl-4- ((trimethylsilanyl) ethynyl) -1H-pyrazole (2.5g, 72.9% yield) as a brown oil. MS (ESI) 179.1[ deg. ] M + H] +
Step 2) 4-ethynyl-1-methyl-1H-pyrazole
To a solution of 1-methyl-4- ((trimethylsilyl) ethynyl) -1H-pyrazole (2.3g, 7.291mmol) in THF (20 mL) was added tetrabutylammonium fluoride (14.6 mL,14.6mmol,1N in THF) at room temperature. The reaction was stirred at room temperature for 1 hour. The reaction was diluted with EtOAc (300 mL) and washed with brine (150 mL. Times.2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by chromatography (PE: EA = 10) to give 4-ethynyl-1-methyl-1H-pyrazole (0.9 g,65% yield) as a clear oil. MS (ESI) 107.0[ m ] +H] +1 HNMR(400MHz,CDCl 3 )δ7.60(s,1H),7.52(s,1H),3.88(s,3H),3.01(s,1H)。
Step 3) (S) -3- (1-aminoethyl) -8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (400mg, 1.34mmol) and 4-ethynyl-1-methyl-1H-pyrazole (213.1mg, 2.00mmol) in CH at room temperature under a nitrogen atmosphere 3 CN (30 mL) mixture was added with X-Phos (319.1mg, 0.67mmol), pd 2 (dba) 3 (306.5mg, 0.33mmol) and K 3 PO 4 (852.5mg, 4.02mmol) and the mixture was stirred at 80 ℃ for 5h. After cooling to room temperature, the mixture was concentrated in vacuo and the residue was diluted with EtOAc (130 mL). The mixture was washed with brine (50 mL) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by flash chromatography (DCM/MeOH = 91/9)To give (S) -3- (1-aminoethyl) -8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (460mg, 83.93% yield) as a yellow oil. MS (ESI) 369.1[ 2 ] M + H] +
Step 4) (S) -2-amino-5-methyl-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (60mg, 0.16mmol) and 2-amino-5-methylpyrazolo [1,5-a ]]DIEA (84.1mg, 0.65mmol), EDCI (46.8mg, 0.24mmol) and HOAT (39.4mg, 0.24mmol) were added to a solution of pyrimidine-3-carboxylic acid (31.31mg, 0.16mmol) in DCM (20 mL). The mixture was stirred at 40 ℃ for 16 hours. The mixture was diluted with DCM (30 mL) and washed with brine (20 mL) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by preparative HPLC (Gemini-C18 150x21.2mm,5um ACN- -H2O (0.1% FA) 30-70) to give (S) -2-amino-5-methyl-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (28.8mg, 32.47% yield) was a white solid. MS (ESI) 545.9[ m ] +H] +1 H NMR(400MHz,DMSO)δ8.78(d,J=6.9Hz,1H),8.17(d,J=6.6Hz,1H),8.01(s,1H),7.66-7.45(m,8H),7.40(d,J=7.6Hz,1H),6.91(d,J=6.9Hz,1H),6.73(s,1H),6.32(s,2H),4.526-4.49(m,1H),3.82(s,3H),2.57(s,3H),1.35(d,J=6.8Hz,3H)。
Example 3-amino-N- ((1S) -1- (1-oxo-8- (2- (5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000281
Step 1) 1- (2, 4-dimethoxybenzyl) -5-oxopyrrolidine-3-carboxylic acid
A solution of (2, 4-dimethoxyphenyl) methylamine (2.0 g,11.9 mmol) and 2-methylenesuccinic acid (1.55g, 11.9 mmol) in toluene (50 mL) was warmed to 100 ℃ and stirred for 16h. The mixture is added Water (20 mL) was diluted and extracted with EtOAc (150 mL). The combined organic phases were washed with saturated brine (40mL × 2), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (DCM/MeOH = 20/1) to give 1- (2, 4-dimethoxybenzyl) -5-oxopyrrolidine-3-carboxylic acid (3.2g, 91% yield) as a colorless oily liquid. MS (ESI) 280.1[ deg. ] M + H ]] +
Step 2) 1- [ (2, 4-dimethoxyphenyl) methyl ] -4- (hydroxymethyl) pyrrolidin-2-one
BH3.THF (13mL, 1mol/L) was added to a solution of 1- (2, 4-dimethoxybenzyl) -5-oxopyrrolidine-3-carboxylic acid (3.2g, 11.4 mmol) in THF (50 mL) at 0 deg.C, then warmed to room temperature and stirred for 16h. The mixture was quenched with MeOH (20 mL) and concentrated under reduced pressure. The residue was dissolved in EtOAc (200 mL) and washed with saturated brine (40mL. Times.2). The organic phase is passed through anhydrous Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH = 20/1) to give 1- [ (2, 4-dimethoxyphenyl) methyl group]4- (hydroxymethyl) pyrrolidin-2-one (2.0 g,59% yield) as a colorless oily liquid. MS (ESI) 266.1[ deg. ] M + H ]] +
Step 3) 1- [ (2, 4-dimethoxyphenyl) methyl ] -5-oxopyrrolidine-3-carbaldehyde
At 20 deg.C, to 1- [ (2, 4-dimethoxyphenyl) methyl group ]A solution of-4- (hydroxymethyl) pyrrolidin-2-one (1.6 g, 6.03mmol) in DCM (50 mL) was added Dess-Martin periodinane (3.1 g,6 mmol) and stirred for 16h with incubation. DCM (250 mL) was added to the mixture and filtered. The filtrate is taken with Na 2 SO 3 Aqueous solution (50 mL) and NaHCO 3 Aqueous (50 mL). The organic phase is passed through anhydrous Na 2 SO 4 Drying, concentrating under reduced pressure to obtain 1- [ (2, 4-dimethoxyphenyl) methyl]-5-oxopyrrolidine-3-carbaldehyde (1.6 g,80% yield) was a colorless oily liquid. MS (ESI) 264.1[ 2 ] M + H] +
Step 4) 1- [ (2, 4-dimethoxyphenyl) methyl ] -4-ethynylpyrrolidin-2-one
To 1- [ (2, 4-dimethoxyphenyl) methyl group at 20 DEG C]-5-oxopyrrolidine-3-carbaldehyde (1.6 g, 6.1mmol) and K 2 CO 3 (2.5g, 18mmol) in MeOH (100 mL) was added (1-diazo-2)-oxopropyl) phosphonic acid dimethyl ester (1.38g, 7.3 mmol), stirring was continued for 4h at RT. The mixture was diluted with EtOAc (250 mL) and washed with saturated brine (50mL. Times.2). The organic phase is treated with anhydrous Na 2 SO 4 Drying, concentrating under reduced pressure to obtain 1- [ (2, 4-dimethoxyphenyl) methyl]-4-ethynylpyrrolidin-2-one (1.1g, 62% yield) was a colorless oily liquid. MS (ESI) 260.1[ 2 ] M + H] +
Step 5) 3- ((1S) -1-aminoethyl) -8- (2- (1- ((2, 4-dimethoxyphenyl) methyl) -5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-1-one
Under nitrogen atmosphere, to 1- [ (2, 4-dimethoxyphenyl) methyl group](iii) -4-ethynylpyrrolidin-2-one (208mg, 0.8 mmol) and (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinoline-1 (2H) -one (160mg, 0.53mmol) in acetonitrile (50 mL) was added K 3 PO 4 (341mg, 1.6 mmol), x-Phos (2-dicyclohexyl-phosphorus-2 ',4',6' -triisopropyl-biphenyl, 102mg, 0.21mmol) and Pd 2 (dba) 3 (98mg, 0.1mmol). The mixture was warmed to 80 ℃ and stirred for 16h. After the reaction was completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was diluted with EtOAc (200 mL), washed with saturated brine (100 mL), and the organic phase was taken up over anhydrous Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH = 20/1) to give 3- ((1S) -1-aminoethyl) -8- (2- (1- ((2, 4-dimethoxyphenyl) methyl) -5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-1-one (280mg, 89% yield) as a brown solid. MS (ESI) 522.2[ m ] +H] +
Step 6) 2-amino-N- ((1S) -1- (8- (2- (1- ((2, 4-dimethoxyphenyl) methyl) -5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 2-aminopyrazolo [1,5-a ] at room temperature]To a solution of pyrimidine-3-carboxylic acid (95mg, 0.53mmol), HOAt (109mg, 0.8mmol), 3- ((1S) -1-aminoethyl) -8- (2- (1- ((2, 4-dimethoxyphenyl) methyl) -5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-1-one (280mg, 0.53mmol) and EDCI (154mg, 0.80mmol) in DCM (10 mL) was added DIEA (208mg, 1.6mmol). The mixture was warmed to 40 ℃ and stirring was continued for 16h. Adding into the mixture DCM (60 mL) was diluted and washed with brine (50 mL). The organic phase is passed through anhydrous Na 2 SO 4 Drying, concentrating under reduced pressure, and purifying the residue by silica gel column chromatography (DCM/MeOH = 20/1) to obtain 2-amino-N- ((1S) -1- (8- (2- (1- ((2, 4-dimethoxyphenyl) methyl) -5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (320mg, 78% yield) was a brown solid. MS (ESI) 682.3[ 2 ] M + H] +
Step 7) 2-amino-N- ((1S) -1- (1-oxo-8- (2- (5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Reacting 2-amino-N- ((1S) -1- (8- (2- (1- ((2, 4-dimethoxyphenyl) methyl) -5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinoline-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (150mg, 0.22mmol) was added to 6N HCl (10 mL) and stirred at 80 ℃ for 1h. Concentrated under reduced pressure and the residue subjected to preparative HPLC (CAN-H) 2 O,0.1% by weight of FA, gradient 20% to 50%) to obtain 2-amino-N- ((1S) -1- (1-oxo-8- (2- (5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (3.7 mg,3.1% yield) was a white solid. MS (ESI) 532.2[ 2 ], [ M ] +H ] +1 H NMR(400MHz,CD 3 OD)δ8.72(d,J=6.8Hz,1H),8.52(s,1H),8.43(t,J=6.4Hz,1H),8.26(t,J=7.2Hz,1H),8.12(d,J=7.2Hz,1H),8.04(d,J=8.0Hz,1H),8.00(s,1H),7.93(d,J=8.0Hz,1H),7.80-7.71(m,3H),7.02-6.98(m,1H),6.72(d,J=8.8Hz,1H),5.09-5.05(m,1H),3.66-3.52(m,3H),2.56-2.48(m,1H),2.35-2.30(m,1H),1.65-1.57(m,3H)。
Example 4-amino-N- ((1S) -1- (8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000291
Step 1) 4- (hydroxymethyl) -1-methylpyrrolidin-2-one
At 0 deg.C, reacting NaBH 4 (481mg, 12.7 mmol) was added to a solution of methyl 1-methyl-5-oxopyrrolidine-3-carboxylate (1g, 6.3 mmol) in MeOH (50 mL). The mixture was warmed to room temperature, stirred for 16h, concentrated under reduced pressure, and the residue was dissolved with MeOH/DCM =1/20 (200 mL), and filtered with suction. The filtrate is treated with anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the residue was separated by column chromatography (MeOH/DCM = 1/20) to give 4- (hydroxymethyl) -1-methylpyrrolidin-2-one (900mg, 98.57% yield) as a colorless oily liquid. MS (ESI) 130.1[ 2 ] M + H] +1 H NMR(400MHz,CDCl 3 )δ3.62-3.57(m,2H),3.50(dd,J=10.0,8.0Hz,1H),3.26(dd,J=10.0,5.0Hz,1H),2.84(s,3H),2.63-2.55(m,1H),2.55-2.49(m,1H),2.22(dd,J=16.4,5.2Hz,1H)。
Step 2) 1-methyl-5-oxopyrrolidine-3-carbaldehyde
To a solution of 4- (hydroxymethyl) -1-methylpyrrolidin-2-one (420mg, 3.2mmol) in DCM (50 mL) at 20 ℃ was added Dess-Martin periodinane (Dess-Martin periodinane,2.7g,6.5 mmol) and stirred at RT for 16h. DCM (100 mL) was added to the mixture and filtered. The filtrate is taken with Na 2 SO 3 Aqueous solution (50 mL) and NaHCO 3 Aqueous (50 mL). The organic phase is treated with anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the residue was isolated by column chromatography (MeOH/DCM = 1/20) to give 1-methyl-5-oxopyrrolidine-3-carbaldehyde (280mg, 67.72% yield) as a colorless oily liquid. MS (ESI) 128.1[ 2 ] M + H ] +
Step 3) 4-ethynyl-1-methylpyrrolidin-2-one
To 1-methyl-5-oxopyrrolidine-3-carbaldehyde (400mg, 3.1mmol) and K at 20 deg.C 2 CO 3 (1.3g, 9.4 mmol) in MeOH (30 mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (718mg, 3.78mmol) and stirring was continued for 4h with incubation. The mixture is passed through H 2 After dilution with O (10 mL), the mixture was washed with saturated brine (40mL. Times.2). The organic phase is passed through anhydrous Na 2 SO 4 Dried, concentrated under reduced pressure, and the residue was isolated by column chromatography (MeOH/DCM = 1/30) to give 4-ethynyl-1-methylpyrrolidin-2-one (180mg, 37.17% yield) as a colorless oily liquid. MS (ESI) 124.1[ deg. ] M + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ3.70-3.55(m,2H),3.27(dd,J=9.1,6.3Hz,1H),2.71(s,3H),2.56(dd,J=16.3,8.6Hz,1H),2.22(dd,J=16.3,7.1Hz,1H)。
Step 4) 3- ((1S) -1-aminoethyl) -8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinoline-1-one
Under a nitrogen atmosphere, to a solution of 4-ethynyl-1-methylpyrrolidin-2-one (37.1mg, 0.3mmol) and (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinoline-1 (2H) -one (60mg, 0.2mmol) in acetonitrile (20 mL) was added K 3 PO 4 (128mg, 0.6 mmol), x-Phos (38mg, 0.08mmol) and Pd 2 (dba) 3 (37mg, 0.04mmol). The mixture was warmed to 80 ℃ and stirred for 4h. After the reaction was completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was diluted with EtOAc (60 mL), washed with saturated brine (30mL X2), and the organic phase was taken up over anhydrous Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH = 20/1) to give 3- ((1S) -1-aminoethyl) -8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-1-one (30mg, 35.0% yield) as a yellow solid. MS (ESI) 473.0[ 2 ] M + H] +
Step 5) 2-amino-N- ((1S) -1- (8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 2-aminopyrazolo [1,5-a ] at room temperature]To a solution of pyrimidine-3-carboxylic acid (15mg, 0.08mmol), HOAt (16mg, 0.12mmol), 3- ((1S) -1-aminoethyl) -8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinoline-1-one (30mg, 0.08mmol) and EDCI (22mg, 0.12mmol) in DCM (10 mL) was added DIEA (30mg, 0.23mmol). The mixture was warmed to 40 ℃ and stirring was continued for 16h. DCM (60 mL) was added to the mixture to dilute it, followed by washing with saturated brine (40 mL). The organic phase is passed through anhydrous Na 2 SO 4 Drying, concentrating under reduced pressure, and subjecting the residue to preparative HPLC (CAN-H) 2 O0.1 FA, gradient 30% to 60%) to give 2-amino-N- ((1S) -1- (8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a) ]Pyrimidine-3-carboxamide (18.0 mg,42.42% yield) was a white solid. MS (ESI) 546.2[ alpha ], [ M ] +H] +1 H NMR(400MHz,CDCl 3 )δ8.47(d,J=6.8Hz,1H),8.42(d,J=4.5Hz,1H),7.94-7.89(m,1H),7.53(d,J=1.7Hz,1H),7.51(d,J=7.5Hz,1H),7.47(d,J=1.3Hz,3H),7.42(s,1H),7.35(d,J=8.0Hz,1H),6.82(dd,J=6.6,4.5Hz,1H),6.62(s,1H),4.84-4.78(m,1H),3.63(dd,J=12.8,5.0Hz,1H),3.57-3.51(m,1H),3.51-3.43(m,1H),2.84(s,3H),2.78-2.71(m,1H),2.63(s,1H),1.40(d,J=6.8Hz,3H)。
Example 5 (S) -2-amino-N- (1- (8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-ylethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000301
Step 1) 5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole-3-carbaldehyde
To 3-bromo-4H, 5H, 6H-pyrrolo [1,2-b ] at-78 DEG C]To a solution of pyrazole (200mg, 1.07mmol) in tetrahydrofuran (10 mL) was slowly added N-butyllithium (2.4M in N-hexane, 0.49mL, 1.18mmol), and the mixture was stirred at-70 ℃ for 1 hour, then a solution of N, N-dimethylformamide (156mg, 2.1mmol) in tetrahydrofuran (1 mL) was added, and the mixture was stirred at 0 ℃ for 2 hours. The mixture was quenched with aqueous ammonium chloride (10 mL) and extracted with ethyl acetate (20mL × 3). The combined organic layers were washed with saturated brine (10mL × 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel chromatography (EA/PE = 1/3) to give 5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole-3-carbaldehyde (100mg, 65% yield) was a white solid. MS (ESI) 137.1[ 2 ] M + H] +1 H NMR(400MHz,CDCl 3 )δ9.81(s,1H),7.95(s,1H),4.25-4.16(m,2H),3.18-3.10(m,2H),2.75-2.63(m,2H)。
Step 2) 3-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole
To 5, 6-dihydro-4H-pyrrole [1,2-b ] ]Pyrazole-3-carbaldehyde (100mg, 0.73mmol) and potassium carbonate (203mg, 1.47mmol) in methanol (10 mL) were added dimethyl (1-diazo-2-oxopropyl) phosphonate (212mg, 1.1mmol), the mixture was stirred at 25 ℃ for 16 h, concentrated under reduced pressure, and the residue was taken up in vacuoDilute with ethyl acetate (40 mL). The organic layer was washed with saturated brine (20mL. Times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give 3-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole (80mg, 70% yield) was a colorless oil. MS (ESI) 133.1[ mu ] M + H] +1 H NMR(400MHz,CDCl 3 )δ7.61(s,1H),4.16-4.11(m,2H),3.03(s,1H),3.00-2.92(m,2H),2.67-2.60(m,2H)。
Step 3) (S) -3- (1-aminoethyl) -8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (60mg, 0.20mmol) and 3-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ] under a nitrogen atmosphere]Pyrazole (40mg, 0.30mmol) in acetonitrile (30 mL) was added with X-Phos (48mg, 0.10 mmol) and Pd 2 (dba) 3 (46mg, 0.05mmol) and potassium phosphate (128mmol, 0.60mmol). The mixture was heated at 80 ℃ for 5 hours, cooled to room temperature, and then the mixture was concentrated under reduced pressure, the residue was diluted with ethyl acetate (30 mL), the mixture was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel chromatography (DCM/MeOH = 91/9) to give (S) -3- (1-aminoethyl) -8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b) ]Pyrazol-3-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (35mg, 39.8% yield) is a yellow oil. MS (ESI) 394.9[ deg. ] M + H ]] +
Step 4) (S) -2-amino-N- (1- (8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-ylethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b) at room temperature]Pyrazol-3-pyrazolyl-3-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (37mg, 0.09mmol) and 2-amino-5-methylpyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid (18mg, 0.10mmol) in dichloromethane (20 mL) were added N, N' -diisopropylethylamine (36mg, 0.28mmol), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (27mg, 0.14mmol), and 1-hydroxy-7-azabenzotriazole (19mg, 0.14mmol). The mixture was stirred at 40 ℃ for 16 hours and then with dichloromethaneAfter dilution (30 mL), the mixture was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product was prepared in liquid phase by high efficiency (Gemini-C18 150x21.2mm,5um ACN-H 2 O (0.1% FA) 30-70) to give (S) -2-amino-N- (1- (8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b)) ]Pyrazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinoline-3-ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (9.3mg, 17.70% yield) was a white solid. MS (ESI) 554.8[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),8.00(d,J=6.7Hz,1H),7.66-7.53(m,5H),7.52-7.46(m,3H),7.42-7.34(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.73(s,1H),6.44(s,2H),4.55(q,J=6.7Hz,1H),4.07(t,J=7.2Hz,2H),2.93-2.80(m,2H),2.58-2.53(m,1H),1.35(d,J=6.8Hz,3H)。
Example 6-amino-N- ((1S) -1- (1-oxo-8- ((5-oxotetrahydrofuran-3-yl) ethynyl) -2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000311
/>
Step 1) 4- (1-bromovinyl) dihydrofuran-2 (3H) -one
To a solution of 4-vinyloxy-2-one (500mg, 4.46mmol) in dichloromethane (40 mL) at 0 ℃ was added liquid bromine (784 mg, 4.9mmol), and after the mixture was stirred at 0 ℃ for 2 hours, the mixture was concentrated under reduced pressure and the residue was dissolved in acetonitrile (20 mL). 1, 8-diazabicycloundec-7-ene (1.35g, 8.9 mmol) was added to the mixture at 0 ℃ and stirred for 16 h at 25 ℃. The mixture was diluted with ethyl acetate (50 mL) and washed with saturated brine (30mL x 2). The separated organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (EA/PE = 1/4) to give 4- (1-bromovinyl) dihydrofuran-2 (3H) -one (600mg, 31.7% yield) as a brown oil. MS (ESI) 191.0,193.0[ 2 ], [ M ] +H] +1 H NMR(400MHz,CDCl3)δ5.80(dd,J=2.4,0.8Hz,1H),5.58(d,J=2.4Hz,1H),4.45-4.39(m,1H),4.25-4.20(m,1H),3.55-3.46(m,1H),2.71-2.66(m,2H)。
Step 2) 4-Ethyldihydrofuran-2 (3H) -one
To a solution of diisopropylamine (1.38g, 13.6 mmol) in tetrahydrofuran (30 mL) was slowly dropped N-butyllithium (2.4N N-hexane solution, 5.9mL,14.3 mmol) at-70 ℃ and stirred at-30 ℃ for 1 hour. The mixture was then cooled to-70 ℃ and a solution of 4- (1-bromovinyl) dihydrofuran-2 (3H) -one (650mg, 3.4 mmol) in tetrahydrofuran (3 mL) was added and stirred at-10 ℃ for 2 hours. The mixture was quenched with aqueous ammonium chloride (30 mL) and extracted with ethyl acetate (30mL × 3). The combined organic layers were washed with saturated brine (50 mL), and the separated organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (EA/PE = 1/4) to give 4-ethynyldihydrofuran-2 (3H) -one (250mg, 43.4% yield) as a colorless oil. 1 H NMR(400MHz,CDCl 3 )δ4.51(dd,J=8.8,7.6Hz,1H),4.22(dd,J=8.8,7.6Hz,1H),3.45-3.33(m,1H),2.81(dd,J=17.4,8.8Hz,1H),2.63(dd,J=17.4,8.8Hz,1H),2.25(d,J=2.4Hz,1H)。
Step 3) 3- ((S) -1-aminoethyl) -8- ((5-oxotetrahydrofuran-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To a solution of (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinoline-1 (2H) -one (50mg, 0.17mmol) and 4-ethynyldihydrofuran-2 (3H) -one (37mg, 0.33mmol) in acetonitrile (30 mL) under a nitrogen atmosphere was added X-Phos (40mg, 0.083mmol), pd 2 (dba) 3 (38mg, 0.042mmol) and potassium phosphate (107mg, 0.50mmol), and the mixture was heated at 80 ℃ for 5 hours. After cooling to room temperature, the mixture was concentrated under reduced pressure and the residue was diluted with ethyl acetate (30 mL). The mixture was washed with saturated brine (20 mL), and the separated organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (DCM/MeOH = 91/9) to give 3- ((S) -1-aminoethyl) -8- ((5-oxotetrahydrofuran-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (48 mg, yield 69.30%) as a yellow oil. MS (ESI) 373.0[ M ] +H ] +
Step 4) 2-amino-N- ((1S) -1- (1-oxo-8- ((5-oxotetrahydrofuran-3-yl) ethynyl) -2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To a mixture of 3- ((S) -1-aminoethyl) -8- ((5-oxotetrahydrofuran-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (48mg, 0.13mmol) and 2, 5-dioxopyrrolidin-1-yl 2-aminopyrazolo [1.5-a ]]N, N' -diisopropylethylamine (50mg, 0.39mmol) was added to a mixture of pyrimidine-3-carboxylic acid ester (35.5mg, 0.13mmol) in acetonitrile (10 mL), and the mixture was stirred at 80 ℃ for 16 hours. After cooling to room temperature, the mixture was concentrated under reduced pressure, the residue was diluted with dichloromethane (30 mL), washed with saturated brine (20 mL), the separated organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a residue (Gemini-C18 150x21.2mm.2mm.5um ACN-H) 2 O (0.1% by weight FA) 40-55) to give 2-amino-N- ((1S) -1- (1-oxo-8- ((5-oxotetrahydrofuran-3-yl) ethynyl) -2-phenyl-1, 2-dihydroisoquinoline-3-ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (14.8mg, 21.1% yield) was a white solid. MS (ESI) 532.8[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),7.99(d,J=6.7Hz,1H),7.67-7.42(m,7H),7.37(dd,J=5.7,3.5Hz,1H),7.01(dd,J=6.7,4.5Hz,1H),6.74(s,1H),6.43(s,2H),4.59-4.47(m,2H),4.20(dd,J=8.3,6.9Hz,1H),3.77-3.68(m,1H),2.86(dd,J=17.0,8.6Hz,1H),2.61(dd,J=17.0,7.8Hz,1H),1.34(d,J=6.8Hz,3H)。
Example 7 (S) -2-amino-N- (1- (8- ((3-hydroxyoxetan-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000321
Step 1) 3-acetylenyloxyoxetan-3-ol
Ethynylmagnesium bromide (25mL, 12.50mmol,0.5mmol in THF) was slowly added dropwise to a solution of 3-oxetanone (750mg, 10.41mmol) in tetrahydrofuran (20 mL) at-78 ℃ under nitrogen protection, and the mixture was stirred at room temperature for 2 hours. The reaction was quenched with aqueous ammonium chloride (20 mL) and extracted with ethyl acetate (50mL. Times.3). The combined organic layers were saturated with saturated foodWashed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by flash silica gel chromatography (DCM/MeOH = 30/1) to give 3-acetylenyloxyoxetan-3-ol (485mg, 42.75% yield) as a yellow oil. 1 H NMR(400MHz,CDCl 3 )δ4.83(dd,J=6.7,0.9Hz,2H),4.68(dd,J=6.6,0.9Hz,2H),3.52(s,1H),2.71(s,1H)。
Step 2) 3- ((S) -1-aminoethyl) -8- ((3-hydroxyoxetan-3-yl) ethynyl) -2-phenylnaphthalen-1 (2H) -one
To a solution of (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (60mg, 0.20 mmol) and 3-ethynyl-3-ol (29mg, 0.30mmol) in acetonitrile (10 mL) was added X-Phos (48mg, 0.10 mmol), pd 2 (dba) 3 (37mg, 0.04mmol) and potassium phosphate (128mg, 0.60mmol), and the mixture was heated at 80 ℃ for 5 hours. After cooling to room temperature, the mixture was concentrated under reduced pressure and the residue was diluted with ethyl acetate (60 mL). The mixture was washed with saturated brine (40 mL), and the separated organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (DCM/MeOH = 91/9) to give 3- ((S) -1-aminoethyl) -8- ((3-hydroxyoxetan-3-yl) ethynyl) -2-phenylnaphthalen-1 (2H) -one (50 mg, yield 62.20%) as a yellow solid. MS (ESI) 361.0[ m ] +H ] +
Step 3) (S) -2-amino-N- (1- (8- ((3-hydroxyoxetan-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 3- ((S) -1-aminoethyl) -8- ((3-hydroxyoxetan-3-yl) ethynyl) -2-phenylnaphthalen-1 (2H) -one (25mg, 0.07mmol) and 2-aminopyrazolo [1.5-a ]]To a solution of pyrimidine-3-carboxylic acid (12mg, 0.07mmol) in dichloromethane (20 mL) were added N, N' -diisopropylethylamine (27mg, 0.21mmol), 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (20mg, 0.10mmol), and 1-hydroxy-7-azabenzotriazole (14mg, 0.10mmol). The mixture was stirred at 40 ℃ for 16 hours, and then the mixture was diluted with dichloromethane (60 mL), washed with saturated brine (40 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The crude product is prepared into liquid phase (-Gemini-C18 150x21.2mm,5um 2 O (0.1% by weight of FA), 40-60) to give (S) -2-amino-N- (1- (8- ((3-hydroxyoxetan-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinoline-3-ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (4.1mg, 11.24%) was a yellow solid. MS (ESI) 520.9[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),8.00(d,J=6.7Hz,1H),7.70-7.63(m,2H),7.60-7.54(m,2H),7.53-7.45(m,3H),7.41-7.35(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.75(s,1H),6.58-6.35(m,3H),4.73(d,J=6.3Hz,2H),4.55(t,J=6.1Hz,3H),1.35(d,J=6.8Hz,3H)。
Example 8-amino-N- ((1S) -1- (8- (2- (1, 2-dimethyl-5-oxopyrazol-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000331
Step 1) 1, 2-dimethyl-5-oxopyrazole-3-carboxylic acid methyl ester
To a solution of methyl 2-methyl-5-oxo-1H-pyrazole-3-carboxylate (1g, 6.4 mmol) in DMF (20 mL) at 0 ℃ was added NaH (184mg, 7.7 mmol) and the mixture was stirred for an additional 0.5H. MeI (1.3g, 9.6 mmol) was then added, followed by stirring for 4h. The mixture is quenched with water, NH 4 The Cl solution (50 mL) was diluted and extracted with EtOAc (50mL x 3). The combined organic layers were washed with brine (100mL. Times.2) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by silica gel chromatography (EA: PE = 1) to give methyl 1, 2-dimethyl-5-oxopyrazole-3-carboxylate (1.05g, 91.5% yield) as a brown solid. MS (ESI) 171.1[ deg. ] M + H] +
Step 2) 5- (hydroxymethyl) -1, 2-dimethylpyrazol-3-one
To a solution of methyl 1, 2-dimethyl-5-oxopyrazole-3-carboxylate (1g, 5.9 mmol) in THF (50 mL) at 0 deg.C was added LiAlH 4 (446mg, 11.7mmol) and stirred for 2h with heat preservation. The mixture was quenched by adding 10% NaOH (0.5 mL) and water (1 mL), and filtered. The filter cake was washed with EtOAc (40 mL) and the filtrate was taken over Na 2 SO 4 Drying and vacuum concentrating to obtainTo crude 5- (hydroxymethyl) -1, 2-dimethylpyrazol-3-one (740 mg, yield 88%) as a colorless oil. MS (ESI) 143.1[ 2 ] M + H] +
Step 3) 1, 2-dimethyl-5-oxopyrazole-3-carbaldehyde
To a solution of 5- (hydroxymethyl) -1, 2-dimethylpyrazol-3-one (740mg, 5.2mmol) in DCM (100 mL) at 20 ℃ was added dess-martin reagent (4.4g, 10.4 mmol), and the mixture was stirred for 16 hours. The mixture was diluted with DCM (100 mL) and filtered. The filtrate was washed with water (50 mL) and brine (50 mL), over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by silica gel chromatography (EA/PE = 1/1) to give 1, 2-dimethyl-5-oxopyrazole-3-carbaldehyde (710mg, 87.5% yield) as a yellow oil. MS (ESI) 141.1[ 2 ] M + H] +
Step 4) 5-ethynyl-1, 2-dimethylpyrazol-3-one
To 1, 2-dimethyl-5-oxopyrazole-3-carbaldehyde (710mg, 5.1mmol) and K 2 CO 3 To a solution of (2.1g, 15mmol) in MeOH (50 mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (1.7g, 6.1mmol), and the mixture was stirred at room temperature for 16 hours. The mixture was concentrated in vacuo and diluted with EtOAc (100 mL); then washed with brine (100 mL) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by silica gel chromatography (EA: PE = 1) to give 5-ethynyl-1, 2-dimethylpyrazol-3-one (540 mg,70.4% yield) as a colorless oil. MS (ESI) 137.1[ deg. ] M + H ]] +
Step 5) 3- ((1S) -1-aminoethyl) -8- (2- (1, 2-dimethyl-5-oxopyrazol-3-yl) ethynyl) -2-phenylisoquinoline-1-one
5-ethynyl-1, 2-dimethylpyrazol-3-one (9.6mg, 0.07mmol), 3- [ (1S) -1-aminoethyl]-8-chloro-2-phenylisoquinoline-1-one (13.1mg, 0.044 mmol), K 3 PO 4 (28mg, 0.13mmol), X-phos (8.4mg, 0.02mmol) and Pd 2 (dba) 3 (8.1mg, 0.01mmol) of CH 3 The CN (10 mL) solution was warmed to 80 ℃ and stirred for 16 hours. Cooled to room temperature, concentrated in vacuo, and the residue diluted with EtOAc (50 mL). The mixture was washed with brine (20 mL) and Na 2 SO 4 Drying and vacuum concentrating. The residue was chromatographed on silica gel (MeOH/DCM =)1/20) to give 3- ((1S) -1-aminoethyl) -8- (2- (1, 2-dimethyl-5-oxopyrazol-3-yl) ethynyl) -2-phenylisoquinolin-1-one (120mg, 95% yield) as a brown solid. MS (ESI) 399.2[ m ] +H] +
Step 6) 2-amino-N- ((1S) -1- (8- (2- (1, 2-dimethyl-5-oxopyrazol-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 2-aminopyrazolo [1,5-a ]]3- ((1S) -1-aminoethyl) -8- (2- (1, 2-dimethyl-5-oxopyrazol-3-yl) ethynyl) -2-phenylisoquinolin-1-one (120mg, 0.30mmol) was added to a solution of pyrimidine-3-carboxylic acid (54mg, 0.3mmol) and HOAT (61.5mg, 0.45mmol), EDCI (86.6mg, 0.45mmol), DIEA (117mg, 0.9mmol) in DCM (20 mL), and the mixture was heated to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (60 mL). The mixture was washed with brine (50 mL) and Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (ACN-H) 2 O0.1% FA, gradient 30% to 50%) to give 2-amino-N- ((1S) -1- (8- (2- (1, 2-dimethyl) -5-oxopyrazol-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (56mg, 32.6% yield) was a white solid. MS (ESI): 559.2[ M ] +H] +1 H NMR(400MHz,CDCl 3 )δ8.49(dd,J=6.8,1.6Hz,1H),8.44(dd,J=4.4,1.6Hz,1H),7.97(d,J=6.8Hz,1H),7.66(dd,J=7.6,1.2Hz,1H),7.60-7.56(m,1H),7.55-7.48(m,4H),7.38-7.33(m,1H),6.86-6.82(m,1H),6.67(s,1H),5.88(s,1H),4.86-4.81(m,1H),3.93(s,3H),3.91(s,3H),1.42(d,J=6.8Hz,3H)。
Example 9 (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000341
Step 1) 2- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole
To 2-bromo-5H,6H,7H-pyrrolo[1,2-a]To a solution of imidazole (300mg, 1.6 mmol), ethynyltriisopropylsilane (439mg, 2.4mmol) and palladium acetate (36mg, 0.16mmol) in DMF (10 mL) was added K 2 CO 3 (443mg, 3.2mmol) and X-phos (76mg, 0.16mmol), the mixture was warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with EtOAc (50 mL) and washed with brine (30 mL. Times.2). The organic phase is treated with Na 2 SO 4 Drying and vacuum concentrating. The residue was isolated and purified by column chromatography (DCM/MeOH = 20/1) to give the product (200mg, 41% yield) as a yellow solid. MS (ESI) 289.2[ M ] +H ] +
Step 2) 2- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole
To 2- (2- (triisopropylsilyl) ethynyl) -5H,6H, 7H-pyrrolo [1,2-a ]]Tetrabutylammonium fluoride (0.8mL, 0.8mmol,1M in THF) was added to a solution of imidazole (150mg, 0.52mmol) in THF (5 mL), and the mixture was stirred at 25 ℃ for 2 hours. The mixture was diluted with EtOAc (50 mL) and washed with brine (30 mL). The organic phase is passed through Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to column chromatography (DCM/MeOH = 15/1) to give 2-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ]]Imidazole (60 mg, yield 78%) was a white solid. MS (ESI) 133.0[ m ] +H] +
Step 3) (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazol-2-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (60mg, 0.20mmol) and 2-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ] under a nitrogen atmosphere]To a solution of imidazole (34.5mg, 0.26mmol) in MeCN (50 mL) were added X-Phos (38mg, 0.08mmol), pd 2 (dba) 3 (37mg, 0.04mmol) and K 3 PO 4 (128mg, 0.60mmol). The mixture was warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, the mixture was concentrated in vacuo and the residue was diluted with EtOAc (130 mL) and washed with brine (50 mL. Times.2). The organic phase is passed through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 91/9) to give (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-2-yl) ethynyl) -2-phenylIsoquinolin-1 (2H) -one (50mg, 57% yield) was a yellow oil. MS (ESI) 394.9[ deg. ] M + H ]] +
Step 4) (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-2-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (50mg, 0.13mmol) and 2-aminopyrazolo [1,5-a]A solution of pyrimidine-3-carboxylic acid (25mg, 0.14mmol) in DCM (20 mL) was added DIEA (65mg, 0.51mmol), EDCI (36mg, 0.19mmol) and HOAT (26mg, 0.19mmol). The mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (100 mL) and washed with brine (50mL × 2). The organic phase is treated with Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (-Xbridge-C18 150x 19mm,5um, ACN-H 2 O(0.05%NH 4 OH), 15% -40%) to obtain (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1, 2-a))]Imidazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ]Pyrimidine-3-carboxamide (13.4 mg,19.01% yield) was a white solid. MS (ESI) 554.8[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.94(dd,J=6.7,1.6Hz,1H),8.56(dd,J=4.5,1.6Hz,1H),8.02(d,J=6.6Hz,1H),7.89(s,1H),7.79-7.65(m,3H),7.61-7.46(m,4H),7.43-7.38(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.81(s,1H),6.43(s,2H),4.60-4.52(m,1H),4.17-4.09(m,2H),3.04(t,J=7.6Hz,2H),2.66-2.55(m,2H),1.36(d,J=6.8Hz,3H)。
Example 10-amino-5- (fluoromethyl-d) -N- ((S) -1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000351
Step 1) Ethyl 2-amino-5-methylpyrazolo [1,5-a ] pyrimidine-3-carboxylate
To 3, 5-diamino-1H-pyrazole-4-carboxylic acidTo a solution of ethyl ester (2g, 11.7 mmol) in DMF (20 mL) was added 4,4-dimethoxybutan-2-one (3.1g, 23.5mmol). The mixture was warmed to 110 ℃ and stirred for 1h, then acetic acid (2.8g, 47.0mmol) was added and stirring was continued for 16h. Cooled to room temperature and concentrated in vacuo. The residue was dissolved in EtOAc (200 mL) and brine (100 mL. Times.3), separated, and the organic phase was Na-free 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (PE: EA = 3) to give 2-amino-5-methylpyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (2.2g, 85% yield) was a yellow solid. LCMS (ESI) m/z 221.2[ 2 ], [ M + H ]] +
Step 2) Ethyl 5-methyl-2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
To 2-amino-5-methylpyrazolo [1,5-a ] at 0 deg.C]Trifluoroacetic anhydride (3.8g, 18mmol) was added to a solution of pyrimidine-3-carboxylic acid ethyl ester (2g, 9.0mmol) and DIEA (2.3g, 18mmol) in DCM (30 mL). The mixture was transferred to room temperature and stirred for 16 hours. The mixture was diluted with DCM (100 mL) and washed with brine (50mL × 2). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give 5-methyl-2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (1.5g, 47% yield) was a yellow solid. LCMS MS (ESI) m/z 316.9, [ M ] +H] +
Step 3) Ethyl 5-formyl-2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
To 5-methyl-2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a]Selenium dioxide (421mg, 3.8mmol) was added to a solution of pyrimidine-3-carboxylic acid ethyl ester (600mg, 1.9mmol) in 1, 4-dioxane (30 mL). The mixture was warmed to 100 ℃ and stirred for 36 hours. After cooling to room temperature, filtration and concentration of the filtrate in vacuo. The residue was dissolved in EtOAc (200 mL) and washed with brine (100mL. Times.2). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 2/1) to give 5-formyl-2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (270mg, 43% yield) was a yellow solid. LCMS (ESI) m/z 349.0[ 2 ], [ M + H ]] +
Step 4) Ethyl 5- (hydroxymethyl-d) -2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
To 5-formyl-2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a ] at 0 deg.C ]To a solution of pyrimidine-3-carboxylic acid ethyl ester (200mg, 0.6 mmol) in THF (3 mL) was added deuterated sodium borohydride (11mg, 0.3 mmol). The mixture was transferred to room temperature and stirred for 16 hours. The mixture was quenched with 10% citric acid (30 mL) and extracted with EtOAc (50 mL. Times.2). The combined organic layers were washed with brine (30mL. Times.2) and dried over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 5/1) to give 5- (hydroxymethyl-d) -2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (60mg, 29% yield) was a yellow solid. LCMS (ESI) m/z 334.0[ 2 ], [ M + H ]] +
Step 5) Ethyl 5- (fluoromethyl-d) -2- (2, 2-trifluoroacetamido) pyrazolo [1,5-a ] pyrimidine-3-carboxylate
To 5- (hydroxymethyl) -2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a ] at 0 deg.C]To a solution of pyrimidine-3-carboxylic acid ethyl ester (50mg, 0.15mmol) in DCM (10 mL) was added DAST (48mg, 0.3mmol). The mixture was transferred to room temperature and stirred for 1 hour. The mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain 5- (fluoromethyl-d) -2- (2, 2-trifluoroacetamido) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (40mg, 80% yield) was a yellow oil. LCMS (ESI) m/z 336.1[ m + H ] ] +
Step 6) 2-amino-5- (fluoromethyl-d) pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
To 5- (fluoromethyl) -2- (2, 2-trifluoroacetamide) pyrazolo [1,5-a ]]Ethyl pyrimidine-3-carboxylate (40mg, 0.12mmol) in EtOH (4 mL) and H 2 To the O (1 mL) solution was added sodium hydroxide (10mg, 0.24mmol). The mixture was transferred to room temperature and stirred for 16h. The mixture was adjusted to pH =6-7 with 1M HCl and extracted with EtOAc (30mL x 3). The combined organic phases were passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give 2-amino-5- (fluoromethyl-d) pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (20 mg, yield 80%) wasYellow solid. LCMS MS (ESI) m/z 212.1[ 2 ] M + H] +
Step 7) 2-amino-5- (fluoromethyl-d) -N- ((S) -1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To a solution of (S) -3- (1-aminoethyl) -8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (20mg, 0.05mmol) in DCM (15 mL) was added 2-amino-5- (fluoromethyl-d) pyrazolo [1, 5-a)]Pyrimidine-3-carboxylic acid (14mg, 0.06mmol), HOAt (113mg, 0.08mmol), EDCI (13mg, 0.08mmol), and DIEA (21mg, 0.16mmol). The mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 15/1) to give 2-amino-5- (fluoromethyl-d) -N- ((S) -1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (0.8mg, 2.6% yield) was a white solid. LCMS (ESI) m/z 562.0[ M + H ]] +1 H NMR(400MHz,CD 3 OD)δ8.67(d,J=7.0Hz,1H),7.77(s,1H),7.59(s,1H),7.58-7.48(m,4H),7.47-7.39(m,3H),7.35(d,J=8.8Hz,1H),7.01(d,J=7.0Hz,1H),6.80(s,1H),5.39(d,J=2.3Hz,1H),4.67(d,J=6.9Hz,1H),3.79(s,3H),1.38(d,J=6.8Hz,3H)。
Example 11 (S) -2-amino-N- (1- (2-cyclopropyl-8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-2-yl) ethynyl) -1-oxo-1, 2-dihydroisoquinolin-3-yl) ethyl) -5-methylpyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000361
Step 1) 2-chloro-N-cyclopropyl-6-methylbenzamide
Oxalyl chloride (970mg, 7.67mmol) was added dropwise to a solution of 2-chloro-6-methylbenzoic acid (1.0g, 5.9mmol) in DCM (50 mL) and DMF (40 mg) at 0 ℃. The mixture was stirred at 0 ℃ for a further 2h and concentrated under reduced pressure. To cyclopropylA solution of amine (340mg, 5.9 mmol) and TEA (1.79g, 17.7 mmol) in DCM (50 mL) was added to a solution of the above acid chloride in DCM (10 mL), and the mixture was stirred at room temperature for 5 hours. Subjecting the mixture to hydrogenation with H 2 Dilute O (30 mL) and extract with DCM (50mL × 3). The combined organic phases were washed with brine (40mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (PE/EA = 3/1) to give 2-chloro-N-cyclopropyl-6-methylbenzamide (1.2g, 88% yield) as a yellow solid. MS (ESI) 210.01[ 2 ] M + H ] +
Step 2) (S) - (4- (3-chloro-2- (cyclopropylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamic acid tert-butyl ester
To a solution of 2-chloro-N-cyclopropyl-6-methylbenzamide (300mg, 1.43mmol) in THF (20 mL) at-30 ℃ under a nitrogen atmosphere was added N-BuLi (1.6 mL, 3.72mmol). The mixture was stirred for 1 hour with heat preservation. To a solution of tert-butyl (S) - (1- (methoxy (methyl) amino) -1-oxopropan-2-yl) carbamate (498mg, 2.15mmol) in THF (20 mL) at-30 ℃ under a nitrogen atmosphere was slowly added dropwise a solution of i-PrMgCl (3.5mL, 4.58mmol), and the mixture was stirred for 1 hour. The solution was then added slowly to the above mixture, transferred to room temperature and stirred for 3h. NH for the mixture 4 Aqueous Cl (30 mL) was quenched and extracted with EtOAc (50mL × 3). The combined organic phases were washed with brine (10mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (PE/EA = 3/1) to give tert-butyl (S) - (4- (3-chloro-2- (cyclopropylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamate (463 mg, yield 81%) as a yellow solid. MS (ESI) 402.9[ 2 ], [ M ] +Na] +
Step 3) (S) -3- (1-aminoethyl) -8-chloro-2-cyclopropylisoquinolin-1 (2H) -one
To a solution of (S) - (4- (3-chloro-2- (cyclopropylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamic acid tert-butyl ester (200mg, 0.53mmol) in MeOH (10 mL) was added HCl (2.0 mL) and the mixture was warmed to 80 deg.C and stirred for 2h. After cooling to room temperature, the mixture was concentrated under reduced pressure. The residue is substituted by H 2 O (10 mL) dilution with NaHCO 3 The pH was adjusted to 7-8 with aqueous solution, then extracted with DCM (30mL. Times.3), and the combined organic phases were washed with brine (30 m. Times.3)L x 2) over Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (DCM/MeOH = 95/5) to give (S) -3- (1-aminoethyl) -8-chloro-2-cyclopropylisoquinolin-1 (2H) -one (130mg, 85% yield) as a yellow solid. MS (ESI) 263.0[ deg. ] M + H] +
Step 4) (S) -3- (1-aminoethyl) -2-cyclopropyl-8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazol-2-yl) ethynyl) isoquinolin-1 (2H) -one to (S) -3- (1-aminoethyl) -8-chloro-2-cyclopropylisoquinolin-1 (2H) -one (50mg, 0.19mmol) and 2-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ] under a nitrogen atmosphere]Pyrazole (33mg, 0.25mmol) in MeCN (20 mL) was added X-Phos (45mg, 0.09mmol), pd 2 (dba) 3 (43mg, 0.05mmol) and K 3 PO 4 (121mg, 0.57mmol), the mixture was warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (100 mL) and washed with brine (50 mL. Times.2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 91/9) to give (S) -3- (1-aminoethyl) -2-cyclopropyl-8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b) ]Pyrazol-2-yl) ethynyl) isoquinolin-1 (2H) -one (33mg, 45.3% yield) was a yellow oil. MS (ESI) 359.0[ m ] +H] +
Step 5) (S) -2-amino-N- (1- (2-cyclopropyl-8- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-2-yl) ethynyl) -1-oxo-1, 2-dihydroisoquinolin-3-yl) ethyl) -5-methylpyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -2-cyclopropyl-8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazol-2-yl) ethynyl) isoquinolin-1 (2H) -one (33mg, 0.09mmol) and 2-amino-5-methylpyrazolo [1,5-a ]]A solution of pyrimidine-3-carboxylic acid (18mg, 0.09mmol) in DCM (20 mL) was added DIEA (36mg, 0.28mmol), EDCI (26mg, 0.14mmol) and HOAT (19mg, 0.14mmol), and the mixture was warmed to 40 ℃ and stirred for 16 hours. After cooling to room temperature, the mixture was diluted with DCM (100 mL) and washed with brine (40mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by preparative HPLC (-Gemini-C18 150x 21.2mm,5um 2 O (0.1% FA), 40% -60%) to obtain (S)) -2-amino-N- (1- (2-cyclopropyl-8- ((5, 6-dihydro-4H-pyrrolo [1, 2-b))]Pyrazol-2-yl) ethynyl) -1-oxo-1, 2-dihydroisoquinolin-3-yl) ethyl) -5-methylpyrazolo [1,5-a]Pyrimidine-3-carboxamide (5.5mg, 11% yield) was a white solid. MS (ESI) 532.9[ 2 ], [ M ] +H ] +1 H NMR(400MHz,DMSO-d 6 )δ8.79(d,J=6.9Hz,1H),8.22(d,J=7.2Hz,1H),7.60-7.53(m,3H),6.91(d,J=6.9Hz,1H),6.62(s,1H),6.38(s,2H),6.26(s,1H),5.84-5.78(m,1H),4.22(t,J=6.6Hz,1H),4.14-4.08(m,2H),3.01(s,1H),2.90-2.84(m,2H),2.56(s,3H),2.54(s,1H),1.58(d,J=6.7Hz,3H),1.11-1.03(m,2H),0.91(t,J=7.4Hz,1H),0.86-0.76(m,2H)。
Example 12 (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1,2-b ] [1,2,4] triazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000371
Step 1) 4-chloro-N' - (diphenylmethylene) butyryl hydrazine
To a solution of (diphenylmethylene) hydrazine (3.9g, 20mmol) and pyridine (1.58g, 20mmol) in DCM (60 mL) at 0 ℃ was added dropwise 4-chlorobutyryl chloride (2.8g, 20mmol), and the mixture was stirred at room temperature for 16 hours. The mixture was diluted with DCM (200 mL) and washed with brine (100mL. Times.2), the layers were separated, and the organic phase was purified over anhydrous Na 2 SO 4 Drying, filtration and concentration gave 4-chloro-N' - (diphenylmethylene) hydrazonobutyryl (6.8 g, crude) as a white solid. MS (ESI) 301.0[ 2 ] M + H] +
Step 2) 1- ((diphenylmethylene) amino) pyrrolidin-2-one
To a solution of 4-chloro-N' - (diphenylmethylene) butyrohydrazide (905mg, 3.0 mmol) in THF (20 mL) at 0 ℃ was slowly added NaH (73mg, 3.0 mmol), and the mixture was stirred at room temperature for 16 hours. The mixture was diluted with EtOAc (60 mL), washed with brine (30 mL. Times.2), the layers were separated, and the organic layer was Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 2/1) to give 1- ((diphenylmethylene) amino) pyrrolidin-2-one (768mg, 92% yield) as a white solid. MS (ESI) 265.0[ 2 ], [ M ] +H] +
Step 3) 1-Aminopyrrolidin-2-one hydrochloride
To 1- ((diphenylmethylene) amino) pyrrolidin-2-one (503mg, 1.9mmol) in H 2 To the O (10 mL) solution was added 2N HCl (5 mL), transferred to room temperature, and stirred for 16h. The mixture was extracted with DCM (30 mL), the layers separated and the aqueous layer concentrated in vacuo to give 1-aminopyrrolidin-2-one hydrochloride (330 mg, crude) as a white solid. MS (ESI) 101.1[ deg. ] M + H ]] +
Step 4) Ethyl 2-imino-2- ((2-oxopyrrolidin-1-yl) amino) acetate
To a solution of 1-aminopyrrolidin-2-one hydrochloride (330mg, 2.4 mmol) in EtOH (20 mL) was added ethyl 2-ethoxy-2-iminoacetate (351mg, 2.4 mmol) and the mixture was stirred at 40 ℃ for 16h. The mixture was concentrated in vacuo to give ethyl 2-imino-2- ((2-oxopyrrolidin-1-yl) amino) acetate (450 mg, crude) as a white solid. MS (ESI) 200.0[ m ] +H] +
Step 5) Ethyl 6, 7-dihydro-5H-pyrrolo [1,2-b ] [1,2,4] triazole-2-carboxylate
Ethyl 2-imino-2- ((2-oxopyrrolidin-1-yl) amino) acetate (450mg, 2.3mmol) and POCl 3 The mixture (10 mL) was stirred at 110 ℃ for 16h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with EtOAc (50 mL) and aqueous sodium bicarbonate (30 mL). The layers were separated and the organic phase was washed with brine (30mL. Times.2) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 6, 7-dihydro-5H-pyrrolo [1,2-b ]][1,2,4]Triazole-2-carboxylic acid ethyl ester (100mg, 25% yield) was a white solid. MS (ESI) 182.0[ m ] +H] +
Step 6) (6, 7-dihydro-5H-pyrrolo [1,2-b ] [1,2,4] triazol-2-yl) methanol
At 0 deg.C, liAlH 4 (21mg, 0.55mmol) was slowly added to 6, 7-dihydro-5H-pyrrolo [1,2-b ]][1,2,4]Triazole-2-carboxylic acid ethyl ester (100mg, 0.55mmol) in THF (10 mL), the mixture was stirred at room temperature for 16h, quenched with 10% NaOH (0.1 mL) and water (5 mL), then extracted with EtOAc (30mL x 2), and the combined organic phasesWashed with brine (50 mL) over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain (6, 7-dihydro-5H-pyrrolo [1, 2-b)][1,2,4]Triazol-2-yl) methanol (80 mg, crude) was a white solid. MS (ESI) 140.1[ 2 ] M + H] +
Step 7) 6, 7-dihydro-5H-pyrrolo [1,2-b ] [1,2,4] triazole-2-carbaldehyde
To (6, 7-dihydro-5H-pyrrolo [1, 2-b)][1,2,4]Triazol-2-yl) methanol (50mg, 0.36mmol) in CHCl 3 (20 mL) solution added MnO 2 (157mg, 1.8 mmol), the mixture was warmed to 70 ℃ and stirred for 16h. Cooling to room temperature, filtering, and vacuum concentrating the filtrate to obtain 6, 7-dihydro-5H-pyrrolo [1,2-b ]][1,2,4]Triazole-2-carbaldehyde (40 mg, crude) was a white solid. MS (ESI) 138.0[ m ] +H ] +
Step 8) 2-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-b ] [1,2,4] triazole
To 6, 7-dihydro-5H-pyrrolo [1,2-b ]][1,2,4]Triazole-2-carboxaldehyde (40mg, 0.30mmol) in MeOH (10 mL) was added to dimethyl (1-diazo-2-oxopropyl) phosphonate (85mg, 0.44mmol) and K 2 CO 3 (81mg, 0.58mmol), the mixture is stirred at room temperature for 16h and concentrated in vacuo. The residue was diluted with EtOAc (50 mL) and washed with brine (30 mL. Times.2). The organic phase is passed through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 2-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-b ]][1,2,4]Triazole (18mg, 45% yield) was a white solid. MS (ESI) 156.1[ 2 ] M + Na] +
Step 9) (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1,2-b ] [1,2,4] triazol-2-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (40mg, 0.13mmol) in CH under a nitrogen atmosphere 3 CN (20 mL) solution was added with 2-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-b ]][1,2,4]Triazole (27mg, 0.20mmol), K 3 PO 4 (57mg,0.27mmol)、Pd 2 (dba) 3 (31mg, 0.03mmol) and X-Phos (32mg, 0.07mmol), the mixture was warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with EtOAc (50 mL)Released, washed with brine (30mL x 3). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1, 2-b)][1,2,4]Triazol-2-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (25mg, 45% yield) was a white solid. MS (ESI) 395.9[ m ] +H] +
Step 10) (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1,2-b ] [1,2,4] triazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1, 2-b)][1,2,4]Triazol-2-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (25mg, 0.06mmol) in DCM (20 mL) was added 2-aminopyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (12mg, 0.06mmol), EDCI (19mg, 0.09mmol), HOAT (13mg, 0.09mmol) and DIEA (25mg, 0.19mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (40 mL) and washed with brine (30mL × 2). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by preparative HPLC (Gemini-C18 150x 21.2mm,5um, ACN- -H2O (0.1% FA) 20% -30%) to give (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1,2-b ] -a) ][1,2,4]Triazol-2-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (16.1 mg, yield 45%) was a white solid. MS (ESI) 555.9[ 2 ] M +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.56(dd,J=4.5,1.6Hz,1H),8.02(d,J=6.7Hz,1H),7.75-7.66(m,3H),7.58-7.46(m,4H),7.41-7.37(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.78(s,1H),6.44(s,2H),4.60-4.55(m,1H),4.11-4.06(m,2H),2.85-2.80(m,2H),2.65-2.58(m,2H),1.36(d,J=6.8Hz,3H)。
Example 13 (S) -2-amino-N- (1- (8- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-6-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000391
Step 1) Ethyl 2, 3-dihydropyrazolo [5,1-b ] oxazole-6-carboxylate
To ethyl 5-hydroxy-1H-pyrazole-3-carboxylate (2g, 12.8 mmol) in CH 3 CN (10 mL) solution was added 1, 2-dibromoethane (6 g, 32mmol) and potassium carbonate (2.5g, 17.68mmol), and the mixture was stirred at 65 ℃ for 36h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (200 mL) and washed with water (100 mL) and brine (100 mL), respectively. Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give 2, 3-dihydropyrazolo [5, 1-b)]Oxazole-6-carboxylic acid ethyl ester (1.8g, 77% yield) was a white solid. LCMS (ESI) m/z 183.1[ 2 ] M + H] +
Step 2) (2, 3-dihydropyrazolo [5,1-b ] oxazol-6-yl) methanol
At 0 ℃ to 2, 3-dihydropyrazolo [5,1-b ]]To a solution of oxazole-6-carboxylic acid ethyl ester (100mg, 0.55mmol) in THF (10 mL) was added LiAlH 4 (31mg, 0.82mmol) and the mixture was transferred to room temperature and stirred for 2h. The mixture was quenched with 10% NaOH (0.3 mL) and water (1 mL), then diluted with ethyl acetate (100 mL), the layers were separated, washed with brine (50 mL), and the organic phase was over anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain (2, 3-dihydropyrazolo [5, 1-b)]Oxazol-6-yl) methanol (60 mg, yield 78%) was a white solid. LCMS MS (ESI) M/z141.2[ M + H ]] +
Step 3) 2, 3-dihydropyrazolo [5,1-b ] oxazole-6-carbaldehyde
To (2, 3-dihydropyrazolo [5, 1-b)]To a solution of oxazol-6-yl) methanol (50mg, 0.36mmol) in chloroform (10 mL) was added MnO 2 (155mg, 1.78mmol), the mixture was warmed to 62 ℃ and stirred for 36h. After cooling to room temperature, it was filtered. The filter cake was washed twice with DCM (10 mL) and the filtrate was concentrated in vacuo to give 2, 3-dihydropyrazolo [5,1-b]Oxazole-6-carbaldehyde (40mg, 81% yield) was a white solid. LCMS MS (ESI) m/z 139.1[ 2 ] M + H] +
Step 4) 6-ethynyl-2, 3-dihydropyrazolo [5,1-b ] oxazole
To 2, 3-dihydropyrazolo[5,1-b]To a solution of oxazole-6-carbaldehyde (40mg, 0.29mmol) in MeOH (10 mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (83mg, 0.43mmol) and potassium carbonate (81mg, 0.58mmol), and the mixture was stirred at room temperature for 16h. The mixture was concentrated under reduced pressure. The residue was dissolved in EtOAc (100 mL) and washed with brine (50 mL). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain 6-ethynyl-2, 3-dihydropyrazolo [5, 1-b)]Oxazole (20 mg, yield 52%) was a yellow solid. LCMS (ESI) m/z 135.2[ 2 ], [ M + H ]] +
Step 5) (S) -3- (1-aminoethyl) -8- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-6-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (30mg, 0.1mmol) in CH 3 CN (10 mL) solution was added with 6-ethynyl-2, 3-dihydropyrazolo [5,1-b ]]Oxazole (20mg, 0.15mmol), pd 2 (dba) 3 (9mg, 0.01mmol), X-phos (5mg, 0.01mmol) and K 3 PO 4 (32mg, 0.15mmol), the mixture was warmed to 80 ℃ and stirred for 4h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was dissolved in DCM (50 mL) and washed with brine (30 mL). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -3- (1-aminoethyl) -8- ((2, 3-dihydropyrazolo [5, 1-b)]Oxazol-6-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (20mg, 50% yield) as a yellow solid. LCMS MS (ESI) m/z 396.8[ m + H ]] +
Step 6) (S) -2-amino-N- (1- (8- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-6-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((2, 3-dihydropyrazolo [5, 1-b)]Oxazol-6-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (20mg, 0.05mmol) and 2-aminopyrazolo [1,5-a ]]HOAt (10mg, 0.07mmol), EDCI (15mg, 0.07mmol) and DIEA (20mg, 0.15mmol) were added to a solution of pyrimidine-3-carboxylic acid (111mg, 0.06mmol) in DCM (20 mL), and the mixture was warmed to 40 ℃ and stirred for 16 hours. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and brine (30)mL) was washed. Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM: meOH = 10) to give (S) -2-amino-N- (1- (8- ((2, 3-dihydropyrazolo [5, 1-b)]Oxazol-6-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (9.5mg, 33% yield) was a white solid. LCMS (ESI) m/z 556.9[ 2 ], [ M + H ]] +1 H NMR(400MHz,CD 3 OD)δ8.69(dd,J=6.8,1.6Hz,1H),8.50(dd,J=4.5,1.6Hz,1H),8.21(d,J=7.0Hz,1H),7.63(dd,J=14.4,8.2,6.9,2.0Hz,4H),7.53-7.38(m,4H),6.96(dd,J=6.8,4.5Hz,1H),6.88(s,1H),5.68(s,1H),5.10-5.02(m,2H),4.76(q,J=6.8Hz,1H),4.32-4.24(m,2H),1.46(d,J=6.8Hz,3H)。
Example 14 (S) -2-amino-N- (1- (8- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-7-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000401
Step 1) 2, 3-dihydropyrazolo [5,1-b ] oxazole
To 1, 2-dihydropyrazol-3-one (2.0 g, 0.02mol) in CH 3 CN (20 mL) solution was added with 1, 2-dibromoethane (19g, 0.09mol) and K 2 CO 3 (6.58g, 0.047mol), the mixture is warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, filtration and concentration of the filtrate in vacuo. The residue was diluted with DCM (100 mL) and washed with brine (50 mL). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 2, 3-dihydropyrazolo [5,1-b ]]Oxazole (1.0 g,34.45% yield) was a yellow oil. MS (ESI) 111.2[ 2 ], [ M ] +H] +
Step 2) 7-bromo-2, 3-dihydropyrazolo [5,1-b ] oxazole
To 2, 3-dihydropyrazolo [5,1-b ]]Oxazole (450mg, 4.09mmol) CH 3 NBS (873 mg, 4.9mmol) was added to a CN (10 mL) solution, and the mixture was stirred for 5 hours. The mixture was diluted with water (20 mL) and then extracted with DCM (30 mL. Times.3).The combined organic phases were washed with brine (30mL. Times.2) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (PE: etOAc = 2)]Oxazole (500mg, 51.79% yield) was a white solid. MS (ESI) 189.1[ alpha ], [ M ] +H] +
Step 3) 7- ((triisopropylsilyl) ethynyl) -2, 3-dihydropyrazolo [5,1-b ] oxazole
To 7-bromo-2, 3-dihydropyrazolo [5,1-b ] under a nitrogen atmosphere]Oxazole (600mg, 3.17mmol) in CH 3 Ethynyltriisopropylsilane (695mg, 3.8mmol), tripotassium phosphate (2g, 9.5mmol), tris (dibenzylideneacetone) dipalladium (581mg, 0.64mmol) and X-phos (302mg, 0.64mmol) were added to a CN (20 mL) solution, and the mixture was heated to 80 ℃ and stirred for 16 hours. After cooling to room temperature, concentrate in vacuo. The residue was diluted with DCM (60 mL) and washed with brine (30mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (PE: etOAc =2]Oxazole (300 mg, yield 26.03%) was a yellow oil. MS (ESI) 291.1[ m ] +H] +
Step 4) 7-ethynyl-2, 3-dihydropyrazolo [5,1-b ] oxazole
To 7- ((triisopropylsilyl) ethynyl) -2, 3-dihydropyrazolo [5,1-b]TBAF (1mL, 1M in THF) was added to a solution of oxazole (300mg, 1.03mmol) in THF (20 mL) and the mixture was stirred for 2h. The mixture was diluted with EtOAc (30 mL) and washed with brine (20 mL. Times.2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM: meOH = 10) to give 7-ethynyl-2, 3-dihydropyrazolo [5,1-b]Oxazole (60mg, 34.64% yield) was a colorless oil. MS (ESI) 135.1[ deg. ] M + H] +
Step 5) (S) -3- (1-aminoethyl) -8- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-7-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (30mg, 0.1mmol) and 7-ethynyl-2, 3-dihydropyrazolo [5,1-b ] under a nitrogen atmosphere]Oxazole (20mg, 0.15mmol)) To a solution of acetonitrile (5 mL) of (b) was added tris (dibenzylideneacetone) dipalladium (9 mg, 0.01mmol), X-phos (5 mg, 0.01mmol) and K 3 PO 4 (43mg, 0.2mmol), the mixture was warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with DCM (30 mL) and washed with brine (30 mL). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10) to give (S) -3- (1-aminoethyl) -8- ((2, 3-dihydropyrazolo [5,1-b]Oxazol-7-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (23mg, 60.0% yield) is a yellow solid. MS (ESI) 398.0[ m ] +H] +
Step 6) (S) -2-amino-N- (1- (8- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-7-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((2, 3-dihydropyrazolo [5, 1-b)]Oxazol-7-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (20mg, 0.05mmol) and 2-aminopyrazolo [1,5-a ]]HOAT (10mg, 0.075mmol), EDCI (14mg, 0.075mmol) and DIEA (13mg, 0.1mmol) were added to a solution of pyrimidine-3-carboxylic acid (111mg, 0.06mmol) in DCM (5 mL), and the mixture was heated to 40 ℃ and stirred for 6 hours. After cooling to room temperature, the mixture was diluted with DCM (40 mL) and washed with brine (20 mL). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -2-amino-N- (1- (8- ((2, 3-dihydropyrazolo [5, 1-b) ]Oxazol-7-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (5.3mg, 18.49% yield) was a yellow solid. MS (ESI) 557.0[ M ] +H] +1 H NMR(400MHz,)δ8.89(dd,J=6.7,1.6Hz,1H),8.51(dd,J=4.5,1.6Hz,1H),7.96(d,J=6.7Hz,1H),7.61-7.51(m,3H),7.49-7.43(m,3H),7.43-7.39(m,2H),7.35-7.30(m,1H),6.98(dd,J=6.7,4.5Hz,1H),6.68(s,1H),6.40(s,2H),5.14-5.07(m,2H),4.53-4.48(m,1H),4.29-4.22(m,2H),1.30(t,J=3.4Hz,3H)。
Example 15 (S) -2-amino-N- (1- (5- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000411
/>
Step 1) (S) -2- (1-aminoethyl) -5- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one
To (S) -2- (1-aminoethyl) -5-bromo-3-phenylquinazolin-4 (3H) -one (50mg, 0.15mmol) and 3-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ] under a nitrogen atmosphere]Pyrazole (29mg, 0.22mmol) in CH 3 CN (20 mL) solution was added with K 3 PO 4 (62mg,0.3mmol)、Pd 2 (dba) 3 (34mg, 0.03mmol) and X-Phos (35mg, 0.08mmol), the mixture was warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, concentrate in vacuo. The residue was diluted with EtOAc (50 mL) and washed with brine (30mL. Times.3). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -2- (1-aminoethyl) -5- ((5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazol-3-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one (20mg, 35% yield) was a white solid. MS (ESI) 395.9[ m ] +H ] +
Step 2) (S) -2-amino-N- (1- (5- ((5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -2- (1-aminoethyl) -5- ((5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazol-3-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one (20mg, 0.05mmol) and 2-aminopyrazolo [1,5-a ]]EDCI (15mg, 0.07mmol)), HOAT (111mg, 0.07mmol) and DIEA (20mg, 0.15mmol) were added to a solution of pyrimidine-3-carboxylic acid (10mg, 0.05mmol) in DCM (20 mL), and the mixture was warmed to 40 ℃ and stirred for 16 hours. The mixture was diluted with DCM (50 mL) and washed with brine (30mL. Times.2), the layers were separated and the organic phase was purified over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was subjected to preparative-HPLC (Gemini-C18 150x 21.2mm,5um, ACN- -H 2 O (0.1% FA), gradient 20% -30%) to give (S) -2-amino-N- (1- (5- ((5, 6-dihydro-4H-pyrrolo)[1,2-b]Pyrazol-3-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (6.2 mg, yield 23%) was a white solid. MS (ESI) 555.8[ 2 ] M +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.92(dd,J=6.7,1.7Hz,1H),8.72(d,J=7.4Hz,1H),8.65(dd,J=4.5,1.6Hz,1H),7.79(d,J=7.6Hz,1H),7.69(dd,J=8.1,1.2Hz,1H),7.65-7.58(m,6H),7.53(d,J=7.5Hz,1H),7.04-7.01(m,1H),6.49-6.42(m,2H),4.74(t,J=7.0Hz,1H),4.08(t,J=7.2Hz,2H),2.88(t,J=7.2Hz,2H),2.57-2.54(m,2H),1.32(d,J=6.6Hz,3H)。
Example 16 (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl)) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000421
Step 1) (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one
To a solution of (S) -3- (1-aminoethyl) -8-chloro-7-fluoro-2-phenylisoquinolin-1 (2H) -one (304mg, 0.96mmol) and 4-ethynyl-1-methyl-1H-pyrazole (153mg, 1.44mmol) in MeCN (50 mL) under a nitrogen atmosphere were added X-Phos (183mg, 0.38mmol), pd 2 (dba) 3 (176mg, 0.20mmol) and K 3 PO 4 (611mg, 2.88mmol), the mixture was warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with EtOAc (130 mL), washed with brine (60mL. Times.2), separated, and the organic phase was taken over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 91/9) to give (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (210mg, 53.8% yield) as a yellow oil. MS (ESI) 386.9[ 2 ], [ M ] +H] +
Step 2) (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl)) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To a solution of (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (120mg, 0.31mmol) and 2-amino-5-methylpyrazolo [1,5-a ] - ]To a solution of pyrimidine-3-carboxylic acid (61mg, 0.34mmol) in DCM (20 mL) were added DIEA (160mg, 1.24mmol), EDCI (89mg, 0.47mmol) and HOAT (63mg, 0.47mmol), and the mixture was warmed to 40 deg.C and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (100 mL) and washed with brine (60mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (-Xbridge-C18 150x 19mm,5um, ACN-H 2 O(0.05%NH 4 OH), 40% -45%) to obtain (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinoline-3-yl)) ethyl) pyrazolo [1, 5-a) and a]Pyrimidine-3-carboxamide (55mg, 32.4% yield) was a white solid. MS (ESI) 546.8[ deg. ] M + H] +1 H NMR(400MHz,DMSO)δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),8.05(s,1H),7.99(d,J=6.6Hz,1H),7.75-7.65(m,2H),7.63-7.55(m,2H),7.53-7.46(m,3H),7.41-7.36(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.79(s,1H),6.43(s,2H),4.54(t,J=6.7Hz,1H),3.82(s,3H),1.35(d,J=6.8Hz,3H)。
Example 17 (S) -2-amino-6-fluoro-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000431
Step 1) (E) -2-fluoro-3-hydroxyacrylaldehyde
PTSA (2.1 g, 0.01mol) was added to a solution of tetramethoxypropane (2.0 g, 0.01mol) in water (10 mL), and the mixture was stirred at room temperature for 2h. Selective fluorine reagent (Selectflur, 5.9g, 0.02mol) was added to the mixture and stirring was continued for 16h. The resulting solution was used directly in the next step. MS (ESI) 91.1[ 2 ] M + H ] +
Step 2) Ethyl 2-amino-6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylate
To (E) -2-fluoro-3-To a solution of hydroxyacrylene (2 mL) was added DMSO (10 mL) slowly. After the mixture was stirred for 10 minutes, ethyl 3, 5-diamino-1H-pyrazole-4-carboxylate (283mg, 1.67mmol) was added. The mixture was warmed to 85 ℃ and stirred for 4h. After cooling to room temperature, the mixture was extracted with EtOAc (30mL × 3). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20) to give 2-amino-6-fluoropyrazolo [1,5-a]Pyrimidine-3-carboxylic acid ethyl ester (100mg, 40.3% yield) was a yellow solid. MS (ESI) 225.0[ m ] +H]+。
Step 3) 2-amino-6-fluoropyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
To 2-amino-6-fluoropyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid ethyl ester (100mg, 0.45mmol) in MeOH (5 mL) was added NaOH (89mg, 2.23mmol) in H 2 O (2 mL) solution, the mixture was warmed to 40 ℃ and stirred for 8h. After cooling to room temperature, concentrate in vacuo. The mixture was acidified with 1N HCl to pH =5-6, filtered, and the filter cake was washed with water (5 mL). The filtrate is concentrated in vacuum to obtain 2-amino-6-fluoropyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (60mg, 54.67% yield) was a yellow solid. MS (ESI) 197.1[ deg. ] M + H] +
Step 4) (S) -2-amino-6-fluoro-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 2-amino-6-fluoropyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid (42mg, 0.21mmol) and (S) -3- (1-aminoethyl) -8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (95mg, 0.26mmol) in DCM (5 mL) was added HOAT (5mg, 0.32mmol), EDCI (62mg, 0.32mmol) and DIEA (55mg, 0.43mmol), and the mixture was warmed to 40 ℃ and stirred for 16H. The mixture was diluted with DCM (20 mL) and washed with brine (20 mL). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (ACN- -H) 2 O (0.1% FA) gradient: 40% -70%) to obtain (S) -2-amino-6-fluoro-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (1695 mg,14.11% yield) was a yellow solid. MS (ESI): 547.0[ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ9.41(dd,J=4.8,2.6Hz,1H),8.78(d,J=2.4Hz,1H),8.00(s,1H),7.84(d,J=6.8Hz,1H),7.68-7.55(m,5H),7.53-7.44(m,3H),7.39-7.35(m,1H),6.75(s,1H),6.50(s,2H),4.59-4.52(m,1H),3.82(s,3H),1.35(d,J=6.8Hz,3H)。
Example 18 (S) -N- (1- (8- ((1-acetoacet-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) -2-aminopyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000432
Step 1) 3-ethynylazetidine-1-carboxylic acid tert-butyl ester
To a solution of 3-formylazetidine-1-carboxylic acid tert-butyl ester (200mg, 1.07mmol) in MeOH (10 mL) was added K 2 CO 3 (370mg, 2.69mmol) and dimethyl (1-diazo-2-oxopropyl) phosphonate (309mg, 1.61mmol), the mixture was transferred to room temperature and stirred for 16h. The mixture was concentrated in vacuo, diluted with EtOAc (100 mL), and washed with brine (40 mL. Times.2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MEOH = 20/1) to give 3-ethynylazetidine-1-carboxylic acid tert-butyl ester (180mg, 83% yield) as a brown oil. MS (ESI) 126.0[ M-55 ]] +
Step 2) 3-ethynylazetidine
To a solution of tert-butyl 3-ethynylazetidine-1-carboxylate (180mg, 0.99mmol) in DCM (10 mL) was added TFA (5 mL) and the mixture was stirred at room temperature for 2h. The mixture was concentrated in vacuo to give 3-ethynylazetidine (100mg, 99.8% yield) as a brown oil. MS (ESI) 82.1[ deg. ] M + H ]] +
Step 3) 1- (3-ethynylazetidin-1-yl) ethan-1-one
Acetyl chloride (228mg, 2.91mmol) was added to a solution of 3-ethynylazetidine (118mg, 1.45mmol) and DIEA (1.8g, 14.5 mmol) in DCM (20 mL) at 0 deg.C, and the mixture was stirred at room temperature for 3 hours. The mixture was diluted with DCM (50 mL) and washed with brine (20mL. Times.2).Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 1- (3-ethynylazetidin-1-yl) ethan-1-one (150mg, 75.36% yield) as a yellow oil. MS (ESI) 124.1[ deg. ] M + H ]] +
Step 4) (S) -8- ((1-Acetazetidin-3-yl) ethynyl) -3- (1-aminoethyl) -2-phenylisoquinolin-1 (2H) -one
To a solution of (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (60mg, 0.20mmol) and 1- (3-ethynylazetidin-1-yl) ethan-1-one (37mg, 0.30mmol) in MeCN (30 mL) was added X-Phos (38mg, 0.08mmol), pd 2 (dba) 3 (37mg, 0.04mmol) and K 3 PO 4 (128mg, 0.60mmol)), the mixture was warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (100 mL) and washed with brine (50 mL. Times.2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 91/9) to give (S) -8- ((1-acetoacet-3-yl) ethynyl) -3- (1-aminoethyl) -2-phenylisoquinoline-1 (2H) -one (47mg, 54.6% yield) as a yellow oil. MS (ESI) 386.0[ m ] +H] +
Step 5) (S) -N- (1- (8- ((1-acetoacet-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) -2-aminopyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -8- ((1-acetoacet-3-yl) ethynyl) -3- (1-aminoethyl) -2-phenylisoquinoline-1 (2H) -one and 2-aminopyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid (22mg, 0.12mmol) in DCM (20 mL) were added DIEA (47mg, 0.37mmol), EDCI (35mg, 0.18mmol) and HOAT (25mg, 0.18mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (30 mL) and washed with brine (20mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by preparative HPLC (Gemini-C18 150x21.2mm,5um ACN- -H 2 O (0.1% by weight FA) 35% to 50%) to give (S) -N- (1- (8- ((1-acetoacet-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroiso-phenyl)Quinolin-3-yl) ethyl) -2-aminopyrazolo [1,5-a]Pyrimidine-3-carboxamide (13.1mg, 19.7% yield) was a white solid. MS (ESI) 546.0[ alpha ], [ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),8.00(d,J=6.6Hz,1H),7.64(dd,J=7.0,4.3Hz,2H),7.60-7.53(m,2H),7.53-7.48(m,2H),7.45(d,J=7.8Hz,1H),7.37(dd,J=5.7,3.5Hz,1H),7.02(dd,J=6.7,4.5Hz,1H),6.74(s,1H),6.43(s,2H),4.54(q,J=6.6Hz,1H),4.38(t,J=8.4Hz,1H),4.15-4.08(m,2H),3.79(dd,J=8.8,6.3Hz,1H),3.68(dd,J=8.9,6.2Hz,1H),1.73(s,3H),1.34(d,J=6.8Hz,3H)。
Example 19-amino-N- ((1S) -1- (2- (2-methyl-5-oxooxooxooxooxooxopyrrolidin-2-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000441
Step 1) methyl 4-oxo-6- (trimethylsilyl) hex-5-ynoate
To a solution of methyl 4-chloro-4-oxobutanoate (100mg, 0.66mmol) and trimethyl (2- (trimethylsilyl) ethynyl) silane (113mg, 0.66mmol) in DCM (10 mL) at 0 deg.C was added AlCl 3 (133mg, 0.99mmol) and the mixture was stirred for 1 hour. The mixture was quenched with 1N HCl (4 mL) and diluted with DCM (50 mL). The mixture was washed with brine (30mL. Times.2) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/3) to give methyl 4-oxo-6- (trimethylsilyl) hex-5-ynoate (96.0 mg,68.1% yield) as a colorless oil. MS (ESI) 213.1[ 2 ] M + H ] +
Step 2) 5-methyl-5- (2- (trimethylsilyl) ethynyl) oxolane-2-one
To a solution of methyl 4-oxo-6- (trimethylsilyl) hex-5-ynoate (120mg, 0.56mmol) in THF (10 mL) at-20 deg.C was added MeMgCl (0.18mL, 0.56mmol, 3mol/L) and the mixture was stirred at room temperature for 16h. NH for the mixture 4 The Cl solution (5 mL) was quenched and extracted with EtOAc (50 mL). Separating, brine for organic phase (3)0 mL) over Na 2 SO 4 Drying and vacuum concentrating to obtain crude 5-methyl-5- [2- (trimethylsilyl) ethynyl]Oxetan-2-one (105mg, 94.62% yield) was a colorless oil. MS (ESI) 143.1[ 2 ] M + H] +
Step 3) 5-ethynyl-5-methyloxolane-2-one
TBAF (0.25mL, 1M) was added to a solution of 5-methyl-5- (2- (trimethylsilyl) ethynyl) oxolane-2-one (92mg, 0.47mmol) in THF (10 mL) at-10 ℃ and the mixture was stirred at room temperature for 1 hour. The mixture was diluted with EtOAc (50 mL) and washed with brine (30 mL). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/1) to give 5-ethynyl-5-methyloxolan-2-one (25mg, 41.25% yield) as a brown oil. 1 H NMR(400MHz,CDCl 3 )δ2.85-2.75(m,1H),2.63-2.49(m,2H),2.59(s,1H),2.23-2.15(m,1H),1.72(s,3H)。
Step 4) 3- ((1S) -1-aminoethyl) -8- (2- (2-methyl-5-oxooxooxooxooxooxooxooxolan-2-yl) ethynyl) -2-phenylisoquinoline-1-one
To 3- ((1S) -1-aminoethyl) -8-chloro-2-phenylisoquinolin-1-one (60mg, 0.2mmol) and 5-ethynyl-5-methyloxolan-2-one (30mg, 0.24mmol) in CH under a nitrogen atmosphere 3 Adding K into CN solution 3 PO 4 (128mg, 0.6 mmol), X-phos (38mg, 0.08mmol) and Pd 2 (dba) 3 (37mg, 0.04mmol), the mixture was warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (100 mL) and washed with brine (30mL x 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (MeOH/DCM = 1/20) to give 3- ((1S) -1-aminoethyl) -8- (2- (2-methyl-5-oxooxooxooxooxooxooxooxolan-2-yl) ethynyl) -2-phenylisoquinolin-1-one (37mg, 74% yield) as a yellow solid. MS (ESI) 387.1[ M ] +H] +
Step 5) 2-amino-N- ((1S) -1- (2- (2-methyl-5-oxooxooxooxooxocyclopent-2-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 2-aminopyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid (25mg, 0.14mmol), HOAt (26mg, 0.19mmol), EDCI (37mg, 0.19mmol) and DIEA (50mg, 0.39mmol) in DCM (10 mL) was added 3- ((1S) -1-aminoethyl) -8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -2-phenylisoquinolin-1-one (50mg, 0.13mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (ACN-H) 2 O0.1% FA, gradient 30% to 60%) to yield 2-amino-N- ((1S) -1- (2- (2-methyl-5-oxooxooxooxooxoheterocyclopentan-2-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] f]Pyrimidine-3-carboxamide (3.1mg, 4.25% yield) was a white solid. MS (ESI) 547.2[ 2 ] M + H] +1 H NMR(400MHz,CD 3 OD)δ8.72(d,J=6.8Hz,1H),8.53(d,J=4.1Hz,1H),8.25(s,1H),7.66(d,J=4.4Hz,2H),7.64(d,J=1.7Hz,1H),7.61(d,J=8.6Hz,1H),7.51(d,J=7.1Hz,1H),7.48(s,1H),7.43(d,J=8.0Hz,1H),6.99(dd,J=6.7,4.9Hz,1H),6.90(s,1H),4.79-4.74(m,1H),2.86(d,J=5.1Hz,1H),2.66-2.56(m,2H),2.28(dd,J=8.8,2.8Hz,1H),1.77(s,3H),1.48(dd,J=6.8,1.5Hz,3H)。
Example 20 (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000461
Step 1) 6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole-3-carbaldehyde
To 6, 7-dihydro-5H-pyrrolo [1,2-a ] at-78 deg.C]To a solution of imidazole (200mg, 1.85mmol) in THF (10 mL) was added n-BuLi (0.88mL, 2.22mmol), and the mixture was warmed to-5 deg.C and stirred for 1 hour. The mixture was cooled to-75 deg.C, N-dimethylformamide (202mg, 2.77mmol) was added, the temperature was raised to 20 deg.C, and stirring was carried out for 2 hours. Cooling to 0-5 deg.C, reacting with saturated NH 4 Aqueous Cl solution (40 mL)Ethance, extract with EtOAc (60 mL. Times.2). The combined organic layers were washed with brine (50mL. Times.2) and dried over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 6, 7-dihydro-5H-pyrrolo [1,2-a ] ]Imidazole-3-carbaldehyde (200 mg, yield 79%) was a yellow oil. LCMS (ESI) m/z 137.0[ m + H ]] +
Step 2) 3-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole
To 6, 7-dihydro-5H-pyrrolo [1,2-a ]]To a solution of imidazole-3-carbaldehyde (200mg, 1.47mmol) in MeOH (10 mL) were added dimethyl (1-diazo-2-oxopropyl) phosphonate (423mg, 2.2mmol) and potassium carbonate (412mg, 2.94mmol), and the mixture was stirred at room temperature for 16h. The mixture was concentrated in vacuo, and the residue was diluted with EtOAc (100 mL) and washed with brine (50 mL). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain 3-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ]]Imidazole (130mg, 67% yield) was a yellow solid. LCMS (ESI) m/z 133.1[ 2 ], [ M + H ]] +
Step 3) (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one to (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (30mg, 0.1mmol) and 3-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ] -c]Imidazole (20mg, 0.15mmol) in CH 3 Adding Pd into CN (10 mL) solution 2 (dba) 3 (9mg, 0.01mmol), X-phos (5mg, 0.01mmol) and K 3 PO 4 (32mg, 0.15mmol), the mixture was warmed to 80 ℃ and stirred for 4h. After cooling to room temperature, it was concentrated in vacuo. The residue was dissolved in DCM (50 mL) and washed with brine (30 mL). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] -c]Imidazol-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (40mg, 99% yield) as a yellow solid. LCMS (ESI) m/z 395.2[ 2 ], [ M + H ]] +
Step 4) (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (40mg, 0.1mmol) and 2-aminopyrazolo [1,5-a ]]HOAt (21mg, 0.15mmol), EDCI (24mg, 0.15mmol) and DIEA (39mg, 0.3mmol) were added to a solution of pyrimidine-3-carboxylic acid (18mg, 0.1mmol) in DCM (20 mL), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ]Pyrimidine-3-carboxamide (10.1mg, 18% yield) was a white solid. LCMS (ESI) m/z 555.0[ 2 ], [ M + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),8.01(d,J=6.7Hz,1H),7.70-7.64(m,2H),7.64-7.55(m,2H),7.54-7.46(m,3H),7.43-7.37(m,1H),7.21(s,1H),7.02(dd,J=6.7,4.5Hz,1H),6.76(s,1H),6.43(s,2H),4.56(t,J=6.7Hz,1H),3.97(t,J=7.0Hz,2H),2.79(t,J=7.4Hz,2H),2.55(d,J=7.5Hz,2H),1.35(d,J=6.8Hz,3H)。
Example 21 (S) -2-amino-5- (methoxy-d 3) -N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000471
To (S) -3- (1-aminoethyl) -8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (30mg, 0.08mmol) and 2-amino-5- (methoxy-d 3) pyrazolo [1,5-a]To a solution of pyrimidine-3-carboxylic acid (17mg, 0.08mmol) in DCM (20 mL) were added DIEA (31mg, 0.24mmol), EDCI (23mg, 0.12mmol) and HOAT (17mg, 0.12mmol), and the mixture was stirred at 40 ℃ for 24 h. After cooling to room temperature, the mixture was diluted with DCM (100 mL) and washed with brine (40mL × 2). Is divided intoLiquid, organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (-Xbridge-C18 150x 19mm,5um, ACN-H 2 O (0.1% TFA) 40% -60%) to give (S) -2-amino-5- (methoxy-d 3) -N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (19.1mg, 41.3% yield) was a white solid. MS (ESI) 561.8[ deg. ] M + H] +1 H NMR(400MHz,DMSO)δ8.74-8.64(m,1H),8.01(s,1H),7.88(d,J=6.4Hz,1H),7.68-7.44(m,8H),7.39(d,J=6.9Hz,1H),6.79(s,1H),6.47(d,J=7.3Hz,1H),6.14(s,2H),4.52-4.41(m,1H),3.82(s,3H),1.36(d,J=6.6Hz,3H)。
Example 22 (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrazolo [5,1-b ] [1,3] oxazin-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000472
Step 1) 6, 7-dihydro-5H-pyrazolo [5,1-b ] [1,3] oxazine
To a solution of 2, 4-dihydro-3H-pyrazol-3-one (2.5g, 30mmol) in DMF (50 mL) was added 1, 3-dibromopropane (9.1g, 45mmol) and K 2 CO 3 (6.2g, 45mmol), the mixture was warmed to 130 ℃ and stirred for 16h. After cooling to room temperature, the mixture was washed with H 2 O (30 mL) was diluted and extracted with EtOAc (50mL. Times.3). The combined organic phases were washed with brine (30mL. Times.2) and over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain 6, 7-dihydro-5H-pyrazolo [5, 1-b)][1,3]Oxazine (1.9 g, crude) was a white solid. MS (ESI) 125.1[ deg. ] M + H ]] +
Step 2) 3-bromo-6, 7-dihydro-5H-pyrazolo [5,1-b ] [1,3] oxazine
To 6, 7-dihydro-5H-pyrazolo [5,1-b ] at 20 DEG C][1,3]To a solution of oxazine (1.9g, 15.3mmol) in DCM (40 mL) was added NBS (2.7g, 15.3mmol) and the mixture was stirred for 16h. The mixture was diluted with DCM (50 mL) and washed with brine (20mL. Times.2). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 1/1) to give 3-bromo-6,7-dihydro-5H-pyrazolo [5, 1-b)][1,3]Oxazine (1.7g, 53% yield) was a white solid. MS (ESI) 202.9[ 2 ] M + H] +
Step 3) 3- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-5H-pyrazolo [5,1-b ] [1,3] oxazine
To 3-bromo-6, 7-dihydro-5H-pyrazolo [5,1-b ] under a nitrogen atmosphere][1,3]Oxazine (780 mg, 3.84mmol) CH 3 To a CN (20 mL) solution were added ethynyl triisopropylsilane (1.05g, 5.76mmol), K 3 PO 4 (1.6g,7.68mmol)、Pd 2 (dba) 3 (880mg, 0.96mmol) and X-Phos (916mg, 1.92mmol), the mixture was warmed to 80 ℃ and stirred for 16h. Cooled to room temperature and concentrated in vacuo. The residue was diluted with EtOAc (50 mL) and washed with brine (30mL. Times.3). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 3- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-5H-pyrazolo [5, 1-b)][1,3]Oxazine (450mg, 38% yield) was a white solid. MS (ESI) 305.0[ m ] +H] +
Step 4) 3-ethynyl-6, 7-dihydro-5H-pyrazolo [5,1-b ] [1,3] oxazine
To 3- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-5H-pyrazolo [5,1-b ]][1,3]To a solution of oxazine (450mg, 1.48mmol) in THF (10 mL) was added TBAF (3.0 mL,1M in THF), and the mixture was stirred at room temperature for 1 hour. The mixture was extracted with EtOAc (60 mL) and washed with brine (40 mL. Times.2). Separating, passing organic phase through anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 1/1) to give 3-ethynyl-6,7-dihydro-5H-pyrazolo [5, 1-b) ][1,3]Oxazine (200mg, 88% yield) was a brown solid. MS (ESI) 149.0[ m ] +H] +
Step 5) (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrazolo [5,1-b ] [1,3] oxazin-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To 3-ethynyl-6, 7-dihydro-5H-pyrazolo [5,1-b ] under a nitrogen atmosphere][1,3]Oxazine (37.2mg, 0.25mmol) and (S) -3- (1-aminoEthyl) -8-chloro-2-phenylisoquinolin-1 (2H) -one (50mg, 0.16mmol) in CH 3 CN (20 mL) solution was added with K 3 PO 4 (72mg,0.34mmol)、Pd 2 (dba) 3 (16mg, 0.01mmol) and X-Phos (16mg, 0.03mmol), the mixture was warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (50 mL) and washed with brine (30mL. Times.3). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrazolo [5, 1-b)][1,3]Oxazin-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (30mg, 43% yield) was a white solid. MS (ESI) 410.9[ 2 ] M + H] +
Step 6) (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrazolo [5,1-b ] [1,3] oxazin-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-5H-pyrazolo [5, 1-b) ][1,3]Oxazin-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (30mg, 0.07mmol) and 2-aminopyrazolo [1,5-a]EDCI (22mg, 0.10 mmol), HOAT (15mg, 0.10 mmol) and DIEA (29mg, 0.21mmol) were added to a solution of pyrimidine-3-carboxylic acid (14mg, 0.07mmol) in DCM (20 mL), and the mixture was warmed to 40 ℃ and stirred for 16h. The mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). The organic phase was washed with brine (50 mL) and anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -2-amino-N- (1- (8- ((6, 7-dihydro-5H-pyrazolo [5, 1-b)][1,3]Oxazin-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (27.8mg, 66% yield) was a white solid. MS (ESI) 570.8[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),8.00(d,J=6.8Hz,1H),7.63-7.55(m,3H),7.52-7.45(m,4H),7.41(s,1H),7.39-7.35(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.72(s,1H),6.44(s,2H),4.57-4.52(m,1H),4.38-4.34(m,2H),4.07(t,J=6.1Hz,2H),2.21-2.13(m,2H),1.35(d,J=6.8Hz,3H)。
Example 23 (S) -2-amino-5- (methoxy-d 3) -N- (1- (5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000481
Step 1) (S) -2- (1-aminoethyl) -5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one
In the CH of 2- ((1S) -1-aminoethyl) -5-bromo-3-phenylquinazolin-4-one (200mg, 0.58mmol) and 4-ethynyl-1-methylpyrazole (93mg, 0.87mmol) 3 CN (30 mL) solution was added with K 3 PO 4 (247mg,1.2mmol)、Pd 2 (dba) 3 (53mg, 0.058 mmol) and X-phos (55mg, 0.12mmol), the mixture was heated to 80 ℃ for 5h. The mixture was cooled to room temperature and concentrated in vacuo. The residue was diluted with EtOAc (60 mL) and washed with brine (40mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -2- (1-aminoethyl) -5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one (130mg, 54.5% yield) as a yellow solid. MS (ESI) 370.1[ m ] +H] +
Step 2) 2-amino-5- (methoxy-d 3) pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
Na (29mg, 1.24mmol) was added to CD 3 OD (3 mL) and the mixture was stirred at 20 ℃ for 1 hour. Then 2-amino-5-chloropyrazolo [1,5-a ] is added]Pyrimidine-3-carboxylic acid ethyl ester (100mg, 0.41mmol). The mixture was stirred for 2h. From the mixture with H 2 Quenching with O (1 mL), heating to 40 ℃ and heating for 16h. The mixture was concentrated in vacuo and the residue was diluted with water (5 mL) and then acidified with 1N HCl to pH =4-5. Filtering, washing filter cake with water (2 mL), decompressing and concentrating filtrate to obtain 2-amino-5- (methoxyl-d 3) pyrazolo [1, 5-a)]Pyrimidine-3-carboxylic acid (90mg, 92.3% yield) was a light colored solid. MS (ESI) 237.1[ deg. ] M + H ] ] +1 H NMR(400MHz,DMSO-d 6 )δ8.66-8.56(m,1H),6.49-6.37(m,1H),6.15(s,2H)。
Step 3) (S) -2-amino-5- (methoxy-d 3) -N- (1- (5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 2- ((1S) -1-aminoethyl) -5- (2- (1-methylpyrazol-4-yl) ethynyl) -3-phenylquinazolin-4-one (30mg, 0.08mmol) and 2-amino-5-methoxypyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid (17mg, 0.08mmol) in DCM (20 mL) were added HOAt (13mg, 0.097mmol), DIEA (32mg, 0.24mmol) and EDCI (23mg, 0.12mmol), and the mixture was warmed to 40 ℃ and heated for 16h. After cooling to room temperature, the mixture was diluted with DCM (30 mL) and washed with brine (30 mL). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 15/1) to give (S) -2-amino-5- (methoxy-d 3) -N- (1- (5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] f]Pyrimidine-3-carboxamide (14.1mg, 28.7% yield) was a white solid. MS (ESI) 563.2[ M ] +H] +1 H NMR(400MHz,CD 3 OD)δ8.44-8.41(m,1H),7.88(s,1H),7.79(t,J=8.0Hz,1H),7.69-7.61(m,5H),7.59-7.56(m,1H),7.53-7.50(m,1H),6.45(d,J=7.2Hz,1H),4.93-4.91(m,1H),3.89(s,3H),1.51(d,J=6.8Hz,3H)。
Example 24 (S) -2-amino-N- (1- (6-fluoro-5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000491
Step 1) 2-bromo-3-fluoro-6-nitrobenzoic acid
To 2-bromo-3-fluorobenzoic acid (3.0g, 13.7mmol) in H at 0 ℃ was added 2 SO 4 (15 mL) of the solution was added dropwise with HNO 3 (3.54g, 20.55mmol), the mixture was stirred at room temperature for 3 hours. The mixture was diluted with ice water (100 mL) and extracted with DCM (100mL × 3). The combined organic phases are passed over Na 2 SO 4 Drying and vacuum concentrating to obtain 2-bromo-3-fluoro6-Nitrobenzoic acid (3.35 g, 83.21% yield) was a yellow solid. MS (ESI) 263.9[ deg. ] M + H] +
Step 2) 2-bromo-3-fluoro-6-nitro-N-phenylbenzamide
2-bromo-3-fluoro-6-nitrobenzoic acid (3.35g, 12.7 mmol) was added to SOCl 2 (30 mL), the mixture was warmed to 80 ℃ and stirred for 16 h. The mixture was then concentrated under reduced pressure to give the acid chloride. To a solution of aniline (1.16g, 12.5 mmol) and TEA (8.1g, 62.5 mol) in DCM (50 mL) at 0 deg.C was added the above acid chloride. The mixture was stirred at room temperature for 5 hours and then with H 2 O (100 mL) was diluted and extracted with DCM (150mL x 3). The combined organic layers were washed with brine (70mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (PE/EA = 3/1) to give 2-bromo-3-fluoro-6-nitro-N-phenylbenzamide (2.0 g,43.2% yield) as a yellow solid. MS (ESI) 338.8[ 2 ] M + H ] +
Step 3) 6-amino-2-bromo-3-fluoro-N-phenylbenzamide
To 2-bromo-3-fluoro-6-nitro-N-phenylbenzamide (400mg, 1.2mmol) and a saturated aqueous ammonium chloride solution (10 mL), fe (660.0 mg,12.0 mmol) was added, and the mixture was stirred at room temperature for 16h. Filtering, and using H to filtrate 2 O (100 mL) was diluted and extracted with EtOAc (50 mL. Times.3). The combined organic layers were washed with brine (40mL. Times.2) and Na 2 SO 4 Drying and concentration under reduced pressure gave 6-amino-2-bromo-3-fluoro-N-phenylbenzamide (366mg, 90.34% yield) as a yellow solid. MS (ESI) 310.9[ 2 ] M + H] +
Step 4) (S) - (1- ((3-bromo-4-fluoro-2- (phenylcarbamoyl) phenyl) amino) -1-oxopropan-2-yl) carbamic acid tert-butyl ester
To a solution of 6-amino-2-bromo-3-fluoro-N-phenylbenzamide (366mg, 1.18mmol) in DCM (50 mL) was added (tert-butoxycarbonyl) -L-alanine (224mg, 1.18mmol), DIEA (611mg, 4.74mmol), and HATU (675mg, 1.78mmol), and the mixture was stirred at room temperature for 16h. Subjecting the mixture to hydrogenation with H 2 O (30 mL) was diluted and extracted with DCM (50mL × 3). The combined organic layers were washed with brine (30mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (PE/EA =)3/1) to give tert-butyl (S) - (1- ((3-bromo-4-fluoro-2- (phenylcarbamoyl) phenyl) amino) -1-oxopropan-2-yl) carbamate (448 mg, yield 70.75%) as a yellow solid. MS (ESI) 501.8[ 2 ] M + Na ] +
Step 5) (S) -2- (1-aminoethyl) -5-bromo-6-fluoro-3-phenylquinazolin-4 (3H) -one
To a solution of tert-butyl (S) - (1- ((3-bromo-4-fluoro-2- (phenylcarbamoyl) phenyl) amino) -1-oxopropan-2-yl) carbamate (448mg, 0.93mmol) in DCM (30 mL) was added HMDS (451mg, 2.79mmol) and I 2 (472mg, 1.86mmol), the mixture was warmed to 45 ℃ and stirred for 16h. After cooling to room temperature, the mixture was taken over Na 2 SO 3 Diluted (20 mL) with aqueous solution and extracted with DCM (30mL. Times.3). The combined organic layers were washed with brine (50mL. Times.2) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 95/5) to give (S) -2- (1-aminoethyl) -5-bromo-6-fluoro-3-phenylquinazolin-4 (3H) -one (267mg, 71.3% yield) as a yellow solid. MS (ESI) 361.8[ M ] +H] +
Step 6) (S) -2- (1-aminoethyl) -6-fluoro-5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one
To a mixture of (S) -2- (1-aminoethyl) -5-bromo-6-fluoro-3-phenylquinazolin-4 (3H) -one (60mg, 0.17mmol) and 4-ethynyl-1-methyl-1H-pyrazole (23mg, 0.22mmol) in CH 3 CN (20 mL) solution was added with X-Phos (39mg, 0.083mmol), pd 2 (dba) 3 (38mg, 0.041mmol) and K 3 PO 4 (105mg, 0.50mmol), the mixture was warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (60 mL) and washed with brine (30mL X2) over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 91/9) to give (S) -2- (1-aminoethyl) -6-fluoro-5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one (49mg, 69% yield) as a yellow oil. MS (ESI) 388.0[ m ] +H] +
Step 7) (S) -2-amino-N- (1- (6-fluoro-5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin) -2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -2- (1-aminoethyl) -6-fluoro-5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one (49mg, 0.13mmol) and 2-amino-5-methylpyrazolo [1,5-a ]]DIEA (65mg, 0.51mmol), EDCI (36mg, 0.19mmol) and HOAT (26mg, 0.19mmol) were added to a solution of pyrimidine-3-carboxylic acid (25mg, 0.14mmol) in DCM (20 mL), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (70 mL) and washed with brine (40mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by preparative HPLC (-Gemini-C18 150x 21.2mm,5um 2 O (0.1% by weight of FA), 30% to 60%) to give (S) -2-amino-N- (1- (6-fluoro-5- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] or ]Pyrimidine-3-carboxamide (2.95mg, 4.27% yield) was a white solid. MS (ESI): 547.8[ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.92(dd,J=6.7,1.6Hz,1H),8.70(d,J=7.4Hz,1H),8.64(dd,J=4.5,1.6Hz,1H),8.09(s,1H),7.85-7.74(m,2H),7.67-7.57(m,5H),7.54(d,J=7.9Hz,1H),7.03(dd,J=6.7,4.5Hz,1H),6.45(s,2H),4.79-4.69(m,1H),3.83(s,3H),1.32(d,J=6.7Hz,3H)。
Example 25 (S) -2-amino-N- (1- (5- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazol-3-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000501
Step 1) 6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole-3-carbaldehyde
To 6, 7-dihydro-5H-pyrrolo [1,2-a ] at-78 deg.C]To a solution of imidazole (200mg, 1.85mmol) in THF (10 mL) was added n-BuLi (0.9mL, 2.22mmol,2.4M in THF), and the mixture was warmed to-10 deg.C and stirred for 1 hour. The mixture was then cooled to-75 deg.C, N-dimethylformamide (203mg, 2.8 mmol) was added, transferred to room temperature, and stirred for 2 hours. Adding saturated NH at 0-5 deg.C 4 Aqueous Cl solution (20 mL)) The reaction was quenched. The mixture was extracted with EtOAc (50mL x 2), and the combined organic layers were washed with brine (50mL x 2) over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give 6, 7-dihydro-5H-pyrrolo [1,2-a ]]Imidazole-3-carbaldehyde (200 mg, yield 79%) was a yellow oil. LCMS (ESI) m/z 137.0[ m + H ]] +
Step 2) 3-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole
To 6, 7-dihydro-5H-pyrrolo [1,2-a ] ]imidazole-3-Formaldehyde (200mg, 1.47mmol) in MeOH (10 mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (423mg, 2.2mmol) and K 2 CO 3 (412mg, 2.94mmol), and the mixture was stirred at room temperature for 16 hours. The mixture was concentrated in vacuo, dissolved with EtOAc (100 mL), and washed with brine (50mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating to obtain 3-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ]]Imidazole (130mg, 67% yield) was a yellow solid. LCMS (ESI) m/z 133.1[ 2 ] M + H] +
Step 3) (S) -2- (1-aminoethyl) -5- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-3-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one to (S) -2- (1-aminoethyl) -5-bromo-3-phenylquinazolin-4 (3H) -one (50mg, 0.15mmol) in CH 3 CN (10 mL) solution was added with 3-ethynyl-6, 7-dihydro-5H-pyrrolo [1,2-a ]]Imidazole (38mg, 0.29mmol), pd 2 (dba) 3 (13mg, 0.015mmol), X-phos (7mg, 0.015mmol) and K 3 PO 4 (46mg, 0.22mmol), the mixture was warmed to 70 ℃ and stirred for 3h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was dissolved in DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -2- (1-aminoethyl) -5- ((6, 7-dihydro-5H-pyrrolo [1, 2-a) ]Imidazol-3-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one (30mg, 52% yield) was a yellow solid. LCMS MS (ESI) m/z 396.2[ m + H ]] +
Step 4) (S) -2-amino-N- (1- (5- ((6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazol-3-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -2- (1-aminoethyl) -5- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-3-yl) ethynyl) -3-phenylquinazolin-4 (3H) -one (30mg, 0.08mmol) in DCM (15 mL) was added 2-aminopyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (14mg, 0.08mmol), [1,2,3]Triazolo [4,5-b]Pyridin-3-ol (116mg, 0.11mmol), EDCI (18mg, 0.11mmol) and DIEA (30mg, 0.23mmol), the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -2-amino-N- (1- (5- ((6, 7-dihydro-5H-pyrrolo [1, 2-a)]Imidazol-3-yl) ethynyl) -4-oxo-3-phenyl-3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (6.5mg, 15% yield) was a white solid. LCMS (ESI) m/z 556.2[ 2 ], [ M + H ] ] +1 H NMR(400MHz,DMSO-d 6 )δ8.92(dd,J=6.7,1.6Hz,1H),8.72(d,J=7.4Hz,1H),8.65(dd,J=4.5,1.6Hz,1H),7.82(d,J=7.7Hz,1H),7.74(d,J=1.2Hz,1H),7.67(dd,J=7.5,1.2Hz,1H),7.63(s,2H),7.61(s,3H),7.25(s,1H),7.03(dd,J=6.7,4.5Hz,1H),6.45(s,2H),4.78-4.71(m,1H),3.96(s,2H),2.76(d,J=7.6Hz,2H),2.54(s,2H),1.32(d,J=6.7Hz,3H)。
Example 26 (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) -5- (methoxy-d 3) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000511
To (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (49mg, 0.38mmol) and 2-amino-5- (methoxy-d 3) pyrazolo [1,5-a ]]A solution of pyrimidine-3-carboxylic acid (20mg, 0.10mmol) in DCM (20 mL) was added DIEA (49mg, 0.38mmol), EDCI (27mg, 0.14mmol) and HOAT (20mg, 0.14mmol), and the mixture was heated at 40 deg.CStirring for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (-Xbridge-C18 150x 19mm,5um, ACN-H 2 O (0.1 The. TFA)) to obtain (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin) -3-yl) ethyl) -5- (methoxy-d 3) pyrazolo [1,5-a ] ethynyl)]Pyrimidine-3-carboxamide (27.3mg, 50.0% yield) was a white solid. MS (ESI) 579.8[ 2 ], [ M ] +H] +1 H NMR(400MHz,DMSO-d 6 )δ8.74-8.67(m,1H),8.05(s,1H),7.88(d,J=6.5Hz,1H),7.75-7.65(m,2H),7.61-7.46(m,5H),7.39(d,J=7.5Hz,1H),6.83(s,1H),6.51-6.45(m,1H),6.13(s,2H),4.52-4.41(m,1H),3.83(s,3H),1.35(d,J=6.7Hz,3H)。
Example 27 (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-5, 7-d 2-3-carboxamide
Figure BDA0003866228850000521
Step 1) N 1 ,N 3 -dimethoxy-N 1 ,N 3 -dimethylmalonamide
To a solution of methoxy (methyl) amine (1.08g, 17.7 mmol) and DIEA (4.59g, 35.5 mmol) in DCM (20 mL) at 0 deg.C was added malonyl chloride (1g, 7.1mmol) and the mixture was stirred at room temperature for 16h. The mixture was diluted with DCM (50 mL) and washed with brine (30mL × 3). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/8) to give N 1 ,N 3 -dimethoxy-N 1 ,N 3 Dimethylmalonamide (700 mg, yield 31%) as a colorless oil. MS (ESI) 191.1[ mu ] M + H] +
Step 2) malondialdehyde-1, 3-d2
At 0 ℃ to N 1 ,N 3 -dimethoxy-N 1 ,N 3 Dimethylmalonamide (500mg, 2.63mmol) in THF (THF: (TM)) (15 mL) was added portionwise LiAlD4 (220mg, 5.24mmol) and the mixture was stirred for 2h. The mixture was then purified with 1N deuterated hydrochloric acid (5 mmol, dissolved in D) 2 O (5 mL)), extracted with EtOAc (10 mL) and the aqueous phase was used in the next step without further purification. MS (ESI) 75.2[ deg. ] M + H] +
Step 3) Ethyl 2-amino-5, 7-d 2-pyrazolo [1,5-a ] pyrimidine-3-carboxylate
To a solution of ethyl 3, 5-diamino-1H-pyrazole-4-carboxylate (250mg, 0.29mmol) in HOAc (20 mL) was added the aqueous solution of malondialdehyde-1, 3-d2 from the previous step, the mixture was warmed to 90 ℃ and stirred for 2H. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (60 mL) and NaHCO respectively 3 Aqueous (40 mL) and brine (40 mL). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/1) to give 2-amino-5, 7-d 2-pyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid ethyl ester (110mg, 34% yield) was a yellow solid. MS (ESI) 210.2[ 2 ] M + H] +
Step 4) 2-amino-5, 7-d 2-pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid
To 2-amino-5, 7-d 2-pyrazolo [1,5-a ]]Ethyl pyrimidine-3-carboxylate (120mg, 0.58mmol) in MeOH (20 mL) was added NaOH (115mg, 2.9 mmol) and H 2 O (5 mL), the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with water (10 mL) and adjusted to pH 5-6 with 1N FA. Filtering, washing the filter cake with water, and drying in vacuum to obtain 2-amino-5, 7-d 2-pyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (60mg, 54.9% yield) was a yellow solid. MS (ESI) 180.9[ deg. ] M + H] +
Step 5) (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin) -3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-5, 7-d 2-3-carboxamide
To a mixture of 3- ((1 s) -1-aminoethyl) -7-fluoro-8- (2- (1-methylpyrazol-4-yl) ethyl) -2-phenylmethanquinolin-1-one (43mg, 0.11mmol) and 2-amino-5, 7-d 2-pyrazolo [1,5-a ] ]To a solution of pyrimidine-3-carboxylic acid (20mg, 0.11mmol) in DCM (30 mL) were added HOAt (18mg, 0.13mmol), EDCI (32mg, 0.17mmol) and DIEA (43mg, 0.33mmol), and the mixture was mixedThe mixture was warmed to 40 ℃ and stirred for 15h. After cooling to room temperature, the mixture was diluted with DCM (40 mL) and washed with brine (30mL × 2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 15/1) to give (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-5, 7-d 2-3-carboxamide (26.9mg, 44% yield) was a light yellow solid. MS (ESI) 549.1[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.95-8.91(m,1H),8.04(s,1H),7.99(d,J=6.4Hz,1H),7.74-7.64(m,2H),7.61(s,1H),7.59-7.55(m,1H),7.53-7.45(m,3H),7.40-7.36(m,1H),6.79(s,1H),6.42(s,2H),4.59-4.50(m,1H),3.82(s,1H),1.35(d,J=6.8Hz,3H)。
Example 28 (S) -2-amino-N- (1- (7-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000531
Step 1) 4-iodo-1- (methyl-d 3) -1H-pyrazole
To a solution of 4-iodo-1H-pyrazole (2g, 10mmol) in DMF (10 mL) at 0 ℃ was added sodium hydride (297mg, 12.3mmol), and the mixture was stirred for 0.5H. Methyl iodide-d 3 (1.8g, 12.3mmol) was then added, the mixture was transferred to room temperature, and the mixture was stirred for 16 hours. The mixture was quenched with ice water (20 mL) and extracted with EtOAc (50 mL. Times.3). The combined organic layers were washed with brine (50mL. Times.3) and over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 2/1) to give 4-iodo-1- (methyl-d 3) -1H-pyrazole (2g, 91% yield) as a yellow oil. LCMS (ESI) m/z 212.0[ m + H ]] +
Step 2) 1- (methyl-d 3) -4- ((trimethylsilyl) ethynyl) -1H-pyrazole
To a solution of 4-iodo-1- (methyl-d 3) -1H-pyrazole (2g, 9.6 mmol) in TEA (30 mL) was added ethynyltrimethylsilane (1.4g, 14.4mmol), cuI (183mg, 0.96mmol), bis (tris (4-methyl-3-yl-trimethyl-phenyl) and the mixture was addedPhenylphosphine) Palladium (II) chloride (675mg, 0.96mmol) and triethylamine (9.7g, 96mmol), the mixture was warmed to 90 ℃ and stirred for 4h. After cooling to room temperature, concentrate in vacuo. The residue was dissolved in DCM (50 mL) and washed with water (30 mL) and brine (30 mL). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 3/1) to give 1- (methyl-d 3) -4- ((trimethylsilyl) ethynyl) -1H-pyrazole (1.2 g, yield 70%) as a brown oil. LCMS (ESI) M/z182.1[ M + H ] M] +
Step 3) 4-ethynyl-1- (methyl-d 3) -1H-pyrazole
To a solution of 1- (methyl-d 3) -4- ((trimethylsilyl) ethynyl) -1H-pyrazole (2.3g, 12.7mmol) in THF (20 mL) was added TBAF (13mL, 12.7mmol,1N in THF), and the mixture was stirred at room temperature for 0.5H. The reaction mixture was diluted with EtOAc (100 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 10/1) to give 4-ethynyl-1- (methyl-d 3) -1H-pyrazole (800 mg, yield 87%) as a yellow solid. LCMS MS (ESI) M/z110.1[ M + H ]] +
Step 4) (S) -3- (1-aminoethyl) -7-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one
To a mixture of (S) -3- (1-aminoethyl) -7-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (150mg, 0.47mmol) and 4-ethynyl-1- (methyl-d 3) -1H-pyrazole (77mg, 0.71mmol) in CH 3 Adding Pd into CN (20 mL) solution 2 (dba) 3 (87mg, 0.095mmol), X-phos (90mg, 0.19mmol) and K 3 PO 4 (150mg, 0.71mmol) and the mixture was warmed to 80 ℃ and stirred for 4h. After cooling to room temperature, concentrate in vacuo. The residue was dissolved in DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -3- (1-aminoethyl) -7-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (120mg, 65% yield) as a brown oil.LCMS:MS(ESI)m/z 390.1[M+H] +
Step 5) (S) -2-amino-N- (1- (7-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To a solution of (S) -3- (1-aminoethyl) -7-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (90mg, 0.23mmol) and 2-aminopyrazolo [1,5-a ]]Pyrimidine-3-carboxylic acid (41mg, 0.23mmol) in DCM (15 mL) was added [1,2,3]Triazole [4,5-b ]]Pyridin-3-ol (47mg, 0.34mmol), EDCI (54mg, 0.34mmol) and DIEA (90mg, 0.69mmol) and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). The combined organic layers were passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by preparative HPLC (DCM/MeOH = 10/1) to give (S) -2-amino-N- (1- (7-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (48.5mg, 38% yield) was a white solid. LCMS MS (ESI) m/z 550.0[ m + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.7,1.6Hz,1H),8.55(dd,J=4.5,1.6Hz,1H),8.05(d,J=0.6Hz,1H),7.99(d,J=6.6Hz,1H),7.72-7.68(m,2H),7.64-7.55(m,2H),7.55-7.45(m,3H),7.41-7.36(m,1H),7.02(dd,J=6.7,4.5Hz,1H),6.79(s,1H),6.43(s,2H),4.54(q,J=6.8Hz,1H),1.35(d,J=6.8Hz,3H)。
Example 29: 2-amino-N- ((1S) -1- (8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide; and example 30:6- (3- ((S) -1- (2-Aminopyrazolo [1,5-a ] pyrimidine-3-carboxamido) ethyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-8-yl) -4-hydroxy-hex-5-ynoic acid methyl ester
Figure BDA0003866228850000541
Step 1) methyl 4-hydroxy-6- (trimethylsilyl) hex-5-ynoate
To a solution of methyl 4-oxo-6- (trimethylsilyl) hex-5-ynoate (300mg, 1.4 mmol) in MeOH (10 mL) at-20 deg.C was added NaBH 4 (59mg, 1.55mmol) and the mixture was stirred at room temperature for 16h. Then with NH 4 The reaction was quenched with Cl solution (5 mL) and concentrated in vacuo. The residue was diluted with EtOAc (50 mL) and washed with brine (30mL. Times.2). The separated organic layer was passed over Na 2 SO 4 Dried and concentrated in vacuo to give methyl 4-hydroxy-6- (trimethylsilyl) hex-5-ynoate (270 mg, 89% yield) as a colorless oil. MS (ESI) 215.2[ deg. ] M + H] +
Step 2) 4-hydroxy-hex-5-ynoic acid methyl ester
TBAF (1.2 mL, 1M/L) was added to a solution of methyl 4-hydroxy-6- (trimethylsilyl) hex-5-nonanoate (250mg, 1.17mmol) in THF (10 mL) at-10 deg.C, and the mixture was stirred at room temperature for 1h. The mixture was diluted with EtOAc (50 mL) and washed with brine (30 mL. Times.2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA: PE = 1) to give methyl 4-hydroxy-hex-5-ynoate (125mg, 60% yield) as a colorless oil. 1 H NMR(400MHz,CDCl3)δ4.50(td,J=6.1,2.1Hz,1H),3.70(s,3H),2.65-2.53(m,2H),2.49(d,J=2.1Hz,1H),2.09-2.04(m,2H)。
Step 3) 3- ((1S) -1-aminoethyl) -8- (2- (5-oxooxooxooxooxocyclopent-2-yl) ethynyl) -2-phenylisoquinolin-1-one; 6- (3- ((S) -1-aminoethyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-8-yl) -4-hydroxy-hex-5-ynoic acid methyl ester
To a solution of 3- ((1S) -1-aminoethyl) -8-chloro-2-phenylisoquinolin-1-one (60mg, 0.2mmol) and 4-hydroxy-hex-5-ynoic acid methyl ester (28mg, 0.2mmol) in CH 3 CN (10 mL) solution was added with K 3 PO 4 (128mg, 0.6 mmol), X-phos (38mg, 0.08mmol) and Pd 2 (dba) 3 (37mg, 0.04mmol), the mixture was warmed to 80 ℃ and stirred for 16h. After cooling to room temperature, concentrate in vacuo. The residue was diluted with DCM (100 mL) and washed with brine (30 mL). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (MeOH/DCM = 1/20) to give 3- ((1S) -1-aminoethyl) -8- (2- (5-oxooxooxooxooxooxolane-2-yl)Ethynyl) -2-phenylisoquinoline-1-one, MS (ESI): 373.1[ 2 ], [ M + H ]] + (ii) a And 6- (3- ((S) -1-aminoethyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-8-yl) -4-hydroxy-hex-5-ynoic acid methyl ester (60mg, 60.16% yield) as a yellow solid, MS (ESI): 405.2[ M ] +H +] +
Step 4) 2-amino-N- ((1S) -1- (8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide; 6- (3- ((S) -1- (2-Aminopyrazolo [1,5-a ] pyrimidine-3-carboxamido) ethyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-8-yl) -4-hydroxy-hex-5-ynoic acid methyl ester
To a mixture of 3- ((1S) -1-aminoethyl) -8- (2- (5-oxooxooxooxooxooxooxooxooxolan-2-yl) ethynyl) -2-phenylisoquinolin-1-one and methyl 6- (3- ((S) -1-aminoethyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-8-yl) -4-hydroxy-hex-5-ynoate (60mg, 0.169mol) and 2-aminopyrazolo [1,5-a ] was added]To a solution of pyrimidine-3-carboxylic acid (29mg, 0.16mmol) in DCM (10 mL) was added HOAT (33mg, 0.24mmol), EDCI (46mg, 0.24mmol) and DIEA (62mg, 0.48mmol), and the mixture was warmed to 40 deg.C and stirred for 16h. The mixture was diluted with DCM (150 mL) and washed with brine (50mL × 2). Separating, passing organic phase through Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (ACN-H) 2 O0.1% FA, gradient 30% to 60%) to give 2-amino-N- ((1S) -1- (8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (8.0mg, 11.30% yield) as a white solid, and 6- (3- ((S) -1- (2-aminopyrazolo [1, 5-a)]Pyrimidine-3-carboxamido) ethyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-8-yl) -4-hydroxy-hex-5-ynoic acid methyl ester (3.4 mg,3.74% yield) as a white solid.
2-amino-N- ((1S) -1- (8- (2- (1-methyl-5-oxopyrrolidin-3-yl) ethynyl) -1-oxo-2-phenylisoquinoline-3-yl) ethyl) pyrazolo [1,5-a ]Pyrimidine-3-carboxamide: MS (ESI) 533.2[ deg. ] M + H] +1 H NMR(400MHz,CD 3 OD)δ8.71(dd,J=6.8,1.6Hz,1H),8.54-8.51(m,1H),7.67-7.35(m,8H),6.98(dd,J=6.8,4.5Hz,1H),6.90(d,J=4.1Hz,1H),5.47(t,J=6.4Hz,1H),4.80-4.74(m,1H),4.56(s,1H),2.66-2.41(m,4H),1.48(d,J=6.7Hz,3H)。
6- (3- ((S) -1- (2-aminopyrazolo [1, 5-a)]Pyrimidine-3-carboxamido) ethyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-8-yl) -4-hydroxy-hex-5-ynoic acid methyl ester: MS (ESI) 565.2[ deg. ] M + H] +1 H NMR(400MHz,CD 3 OD) δ 8.72 (dd, J =6.8,1.6hz, 1h), 8.53 (dd, J =4.5,1.6hz, 1h), 7.63-7.61 (m, 4H), 7.54-7.47 (m, 4H), 6.99 (dd, J =6.8,4.5hz, 1h), 6.89 (s, 1H), 4.83-4.77 (m, 1H), 4.63-4.60 (m, 1H), 3.64 (s, 3H), 2.56 (t, J =7.7hz, 2h), 2.06-2.02 (m, 2H), 1.48 (d, J =6.8hz, 3h). Example 31 (S) -2-amino-N- (1- (8- ((6, 7-dihydro-4H-pyrazolo [5, 1-c)][1,4]Oxazin-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamides
Figure BDA0003866228850000551
Step 1) 4-bromo-1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-pyrazole
To a solution of 4-bromo-1H-pyrazole (2.0 g,13.6 mmol) and (2-bromoethoxy) (tert-butyl) dimethylsilane (3.58g, 149.6 mmol) in MeCN (100 mL) was added cesium carbonate (8.86g, 27.2mmol), and the mixture was stirred at room temperature for 16H. The mixture was concentrated in vacuo, the residue diluted with EtOAc (130 mL), washed with brine (50mL x 2), and the separated organic layer was Na filtered 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 50/1) to give 4-bromo-1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-pyrazole (2.5g, 70% yield) as a yellow oil. MS (ESI) 304.9[ 2 ] M + H ] +
Step 2) 4-bromo-1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-pyrazole-5-carbaldehyde
LDA (0.6 mL, 1.18mmol) was added to a solution of 4-bromo-1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-pyrazole (300mg, 0.98mmol) in THF (30 mL) at-78 deg.C, and the mixture was stirred for 1H. DMF (144mg, 1.96mmol) was then added and the mixture was transferred to room temperature and stirred for 1h. NH for the mixture 4 Quenched with aqueous Cl (20 mL) and EtOAcExtract (50mL x 2) and wash with brine (40mL x 2). The combined organic layers were washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 50/1) to give 4-bromo-1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-pyrazole-5-carbaldehyde (200mg, 66% yield) as a yellow oil. MS (ESI) 333.1[ 2 ] M + H] +
Step 3) 3-bromo-6, 7-dihydro-4H-pyrazolo [5,1-c ] [1,4] oxazine
Et is added dropwise to a solution of 4-bromo-1- (2- ((tert-butyldimethylsilyl) oxy) ethyl) -1H-pyrazole-5-carbaldehyde (200mg, 0.60mmol) in DCM (20 mL) at-78 ℃ under a nitrogen atmosphere 3 SiH (209mg, 1.80mmol) and TMSOTf (800mg, 3.60mmol), the mixture was transferred to room temperature and stirred for 15h. The mixture is washed with NaHCO 3 Aqueous solution (20 mL) was quenched and extracted with DCM (50mL × 3). The combined organic layers were washed with brine (50mL. Times.2) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 40/1) to give 3-bromo-6, 7-dihydro-4H-pyrazolo [5, 1-c)][1,4]Oxazine (100mg, 74% yield) was a yellow oil. MS (ESI) 203.1[ m ] +H] +
Step 4) 3- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-4H-pyrazolo [5,1-c ] [1,4] oxazine
To 3-bromo-6, 7-dihydro-4H-pyrazolo [5,1-c ] under a nitrogen atmosphere][1,4]Oxazine (100mg, 0.49mmol) in MeCN (20 mL) was added X-Phos (117mg, 0.25mmol), pd 2 (dba) 3 (71mg, 0.12mmol) and K 3 PO 4 (313mg, 1.48mmol), the mixture is warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with EtOAc (60 mL) and washed with brine (40mL × 2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 50/1) to give 3- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-4H-pyrazolo [5, 1-c)][1,4]Oxazine (150mg, 90% yield) was a yellow oil. MS (ESI) 305.2[ deg. ] M + H] +
Step 5) 3-ethynyl-6, 7-dihydro-4H-pyrazolo [5,1-c ] [1,4] oxazine
At the temperature of 0 ℃, the temperature of the mixture is controlled,to 3- ((triisopropylsilyl) ethynyl) -6, 7-dihydro-4H-pyrazolo [5,1-c][1,4]To a solution of oxazine (150mg, 0.49mmol) in THF (10 mL) was added TBAF (1mL, 1mmol) and the mixture was stirred for 2h. Subjecting the mixture to hydrogenation with H 2 O (30 mL) was diluted and extracted with EtOAc (30mL x 3). The combined organic layers were washed with brine (20 mL) and Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 30/1) to give 3-ethynyl-6, 7-dihydro-4H-pyrazolo [5,1-c][1,4]Oxazine (114mg, 78% yield) was a yellow oil. MS (ESI) 149.0[ m ] +H] +
Step 6) (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-4H-pyrazolo [5,1-c ] [1,4] oxazin-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To (S) -3- (1-aminoethyl) -8-chloro-2-phenylisoquinoline-1 (2H) -one (65mg, 0.22mmol) and 3-ethynyl-6, 7-dihydro-4H-pyrazolo [5, 1-c)][1,4]Oxazine (113mg, 0.76mmol) in MeCN (20 mL) was added X-Phos (52mg, 0.11mmol), pd 2 (dba) 3 (31mg, 0.05mmol) and K 3 PO 4 (138mg, 0.65mmol), the mixture was warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with EtOAc (70 mL) and washed with brine (50mL. Times.2) over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-4H-pyrazolo [5, 1-c)][1,4]Oxazin-3-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (20mg, 21% yield) as a yellow oil. MS (ESI) 411.1[ deg. ] M + H ] ] +
Step 7) (S) -2-amino-N- (1- (8- ((6, 7-dihydro-4H-pyrazolo [5,1-c ] [1,4] oxazin-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((6, 7-dihydro-4H-pyrazolo [5, 1-c)][1,4]Oxazin-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (20mg, 0.05mmol) and 2-amino-5-methylpyrazolo [1,5-a ]]DIEA (17mg, 0.15mmol), EDCI (14mg, 0.07mmol) and HOAt (10mg, 0.07mmol) were added to a solution of pyrimidine-3-carboxylic acid (7mg, 0.05mmol) in DCM (10 mL), and the mixture was warmed to 40 ℃ and stirred for 16 hours. Mixing ofThe material was diluted with DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by preparative HPLC (-Gemini-C18 150x 19mm,5um 2 O (NH3 in 0.05%), 20-50) to obtain (S) -2-amino-N- (1- (8- ((6, 7-dihydro-4H-pyrazolo [5, 1-c)][1,4]Oxazin-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (6.59mg, 23% yield) was a yellow solid. MS (ESI) 570.8[ 2 ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.92(dd,J=6.8,1.6Hz,1H),8.55(dd,J=4.4,1.6Hz,1H),8.00(d,J=6.8Hz,1H),7.67-7.55(m,5H),7.55-7.45(m,3H),7.41-7.36(m,1H),7.01(dd,J=6.8,4.4Hz,1H),6.74(s,1H),6.42(s,2H),4.84(s,2H),4.56(q,J=6.4Hz,1H),4.13-4.02(m,4H),1.35(d,J=6.8Hz,3H)。
Example 32 (S) -2-amino-N- (1- (5- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-6-yl) ethynyl) -6-fluoro-4-oxo-3-phenyl) -3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000561
Step 1) (S) -2- (1-aminoethyl) -5- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-6-yl) ethynyl) -6-fluoro-3-phenylquinazolin-4 (3H) -one
Under nitrogen atmosphere, 6-ethynyl-2, 3-dihydropyrazolo [5,1-b ] is reacted with a catalyst]Oxazole (50mg, 0.14mmol) and 2-ethynyl-5H, 6H-pyrazolo [3, 2-b)][1,3]Pd was added to a solution of oxazole (18.5mg, 0.14mmol) in acetonitrile (20 mL) 2 (dba) 3 (32mg, 0.034mmol), X-phos (33mg, 0.07mmol) and K 3 PO 4 (88mg, 0.41mmol), the mixture was warmed to 80 ℃ and stirred for 4h. After cooling to room temperature, it was concentrated in vacuo. The residue was dissolved in DCM (50 mL), washed with water (30 mL) and brine (30 mL), and concentrated in vacuo. The residue was purified by column chromatography (DCM/MEOH = 10/1) to give (S) -2- (1-aminoethyl) -5- ((2, 3-dihydropyrazolo [5, 1-b)]Oxazol-6-yl) ethynyl) -6-fluoro-3-phenylquinazolin-4 (3H) -one (20mg, 30% yield) was a white solid. LCMS (ESI) m/z 416.1[ 2 ] M + H] +
Step 2) (S) -2-amino-N- (1- (5- ((2, 3-dihydropyrazolo [5,1-b ] oxazol-6-yl) ethynyl) -6-fluoro-4-oxo-3-phenyl) -3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -2- (1-aminoethyl) -5- ((2, 3-dihydropyrazolo [5, 1-b)]Oxazol-6-yl) ethynyl) -6-fluoro-3-phenylquinazolin-4 (3H) -one (20mg, 0.048mmol) and 2-aminopyrazolo [1,5-a ] ]To a solution of pyrimidine-3-carboxylic acid (8.6mg, 0.048mmol) in DCM (15 mL) were added HOAt (10mg, 0.072mmol), EDCI (11mg, 0.072mmol) and DIEA (18.6mg, 0.14mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with water (30 mL) and extracted with DCM (30mL × 2). The combined organic layers were washed with brine (30mL. Times.2) and dried over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MEOH = 10/1) to give (S) -2-amino-N- (1- (5- ((2, 3-dihydropyrazolo [5, 1-b)]Oxazol-6-yl) ethynyl) -6-fluoro-4-oxo-3-phenyl) -3, 4-dihydroquinazolin-2-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (4.14mg, 14% yield) was a white solid. LCMS MS (ESI) m/z 576.0[ m + H ]] +1 H NMR(400MHz,CDCl 3 )δ8.78(d,J=6.2Hz,1H),8.54(dd,J=4.4,1.7Hz,1H),8.42(dd,J=6.8,1.7Hz,1H),8.29(s,
1H),7.72(d,J=7.4Hz,1H),7.68-7.63(m,1H),7.60-7.58(m,1H),7.56-7.51(m,1H),7.35-7.28(m,1H),6.81(dd,J=6.8,4.4Hz,1H),5.65(s,1H),5.06-4.99(m,2H),4.88-4.82(m,1H),4.34-4.27(m,2H),1.71(d,J=6.9Hz,3H)。
Example 33-amino-N- ((S) -1- (8- (((R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000571
Step 1) (2S,4R) -4- (benzyloxy) pyrrolidine-2-carboxylic acid
To (2S,4R) -4- (benzyloxy) -1- (tert-butoxycarbonyl) pyrrolidine-2-carboxylic acid (4 g,12.4 mmo)l) in DCM (20 mL) was added TFA (4 mL) and the mixture was stirred for 2h. The mixture was concentrated in vacuo to give (2s,4r) -4- (benzyloxy) pyrrolidine-2-carboxylic acid (3g, 98.4% yield) as a yellow oil. MS (ESI) 222.1[ deg. ] M + H ] ] +
Step 2) (2S, 4R) -4- (benzyloxy) -1-nitrosopyrrolidine-2-carboxylic acid
To (2S,4R) -4- (benzyloxy) pyrrolidine-2-carboxylic acid (3g, 13.6 mmol) in H at 0 deg.C 2 Adding NaNO into O (10 mL) solution 2 (1.41g, 20.4mmol) of H 2 O (5 mL) solution, then HOAc (3 mL) was added and the mixture was stirred at room temperature for 16h. Filtration and washing of the filter cake with water (10 mL) and drying in vacuo gave the product (3.5g, 98% yield) as a white solid. MS (ESI) 251.1[ 2 ] M + H] +
Step 3) (R) -5- (benzyloxy) -3-oxo-5, 6-dihydro-3H-pyrrolo [1,2-c ] [1,2,3] oxadiazol-7 (4H) -ammonium-3 a-ylide
To a solution of (2S, 4R) -4- (benzyloxy) -1-nitrosopyrrolidine-2-carboxylic acid (3.5g, 14mmol) in THF (50 mL) at 0 deg.C was added TFAA (4.41g, 21mmol), and the mixture was stirred at room temperature for 16h. The mixture was diluted with EtOAc (80 mL) and washed with brine (50 mL. Times.2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/2) to give (R) -5- (benzyloxy) -3-oxo-5, 6-dihydro-3H-pyrrolo [1, 2-c)][1,2,3]Oxadiazole-7 (4H) -ammonium-3 a-ylide (1.5g, 41.4% yield) was a yellow oil. MS (ESI) 233.1[ deg. ] M + H ]] +
Step 4) (R) -5- (benzyloxy) -2- (trimethylsilyl) -5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole
Reacting (R) -5- (benzyloxy) -3-oxo-5, 6-dihydro-3H-pyrrolo [1, 2-c)][1,2,3]A mixed solution of oxadiazole-7 (4H) -ammonium-3 a-ylide (1g, 4.3 mmol) and ethynyltrimethylsilane (0.84g, 8.6 mmol) in xylene (10 mL) was warmed to 130 ℃ and heated for 16H. After cooling to room temperature, concentrate in vacuo. The residue was purified by column chromatography (EA/PE = 1/3) to give (R) -5- (benzyloxy) -2- (trimethylsilyl) -5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole (600mg, 46.5% yield) was a yellow oil. MS (ESI) 287.1[ alpha ], [ M ] +H] +
Step 5) (R) -5- (benzyloxy) -5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole
To (R) -5- (benzyloxy) -2- (trimethylsilyl) -5, 6-dihydro-4H-pyrrolo [1, 2-b)]TBAF (24.4 mL,24.4 mmol) was added to a solution of pyrazole (700mg, 2.44mmol) in THF (5 mL), and the mixture was warmed to 60 ℃ and heated for 48h. After cooling to room temperature, the mixture was diluted with EtOAc (40 mL) and washed with brine (30 mL. Times.2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/2) to give (R) -5- (benzyloxy) -5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazole (330mg, 56.7% yield) was a yellow solid. MS (ESI) 215.1[ 2 ] M + H] +
Step 6) (R) -5- (benzyloxy) -5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazole-3-carbaldehyde
To (R) -5- (benzyloxy) -5, 6-dihydro-4H-pyrrolo [1,2-b ]]To a solution of pyrazole (330mg, 1.54mmol) in DCM (20 mL) was added (chloromethylene) dimethylammonium chloride (296 mg, 2.31mmol), the mixture was heated to 40 ℃ for 48h. To the mixture was added 1N HCl (3 mL), warmed to 40 ℃ and heated for 2h. Then concentrated in vacuo, the residue diluted with water (5 mL) and NaHCO 3 Aqueous (10 mL) wash. The mixture was extracted with EtOAc (30mL. Times.3) and the combined organic layers were washed with brine (20 mL) and Na 2 SO 4 Drying and vacuum concentrating to obtain (R) -5- (benzyloxy) -5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazole-3-carbaldehyde (350 mg, yield 84%) was a yellow oil. MS (ESI) 243.1[ 2 ] M + H] + . Step 7) (R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazole-3-carbaldehyde
Reacting (R) -5- (benzyloxy) -5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole-3-carbaldehyde (200mg, 0.82mmol) was dissolved in concentrated HCl (10 mL), warmed to 70 ℃ and stirred for 16h. After cooling to room temperature, concentrate in vacuo. The residue was diluted with water (3 mL), washed with aqueous sodium bicarbonate (10 mL), then extracted with EtOAc (20mL. Times.3), and the combined organic layers were Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/3) to give (R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1,2-b ] ]Pyrazole-3-carbaldehyde (140mg, 89% yield) was a yellow solid. MS (ESI) 153.1[ deg. ] M + H ]] +1 H NMR(400MHz,CDCl 3 )δ9.82(s,1H),7.97(s,1H),5.21-5.17(m,1H),4.40(dd,J=12.4,6.0Hz,1H),4.16(dd,J=12.4,2.4Hz,1H),3.46(dd,J=17.6,6.4Hz,1H),3.15(dd,J=17.6,2.4Hz,1H)。
Step 8) (R) -3-ethynyl-5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-5-ol
To (R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1,2-b ]]Pyrazole-3-carbaldehyde (50mg, 0.33mmol) and dimethyl (1-diazo-2-oxopropyl) phosphonate (95mg, 0.49mmol) in MeOH (20 mL) were added K 2 CO 3 (91mg, 0.66mmol) and the mixture was stirred at room temperature for 16h. The mixture was concentrated in vacuo, diluted with EtOAc (60 mL), and washed with brine (20 mL. Times.2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating to obtain (R) -3-ethynyl-5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazol-5-ol (45mg, 83% yield) was a yellow oil. MS (ESI) 148.9[ 2 ] M + H] +1 HNMR(400MHz,CDCl 3 )δ7.62(s,1H),5.15-5.07(m,1H),4.35(dd,J=12.0,6.0Hz,1H),4.09(dd,J=12.0,2.8Hz,1H),3.77(d,J=10.8Hz,1H),3.30(dd,J=16.8,6.4Hz,1H),3.02(s,1H),2.93(dd,J=16.8,2.8Hz,1H)。
Step 9) 3- ((S) -1-aminoethyl) -8- (((R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To a solution of 3- ((1S) -1-aminoethyl) -8-chloro-2-phenylisoquinoline-1-one (50mg, 0.17mmol) and 4-ethynyl-1-methylpyrazole (37mg, 0.25mmol) in CH under a nitrogen atmosphere 3 Adding Pd into CN (20 mL) solution 2 (dba) 3 (31mg,0.033mmol)、K 3 PO 4 (71mg, 0.33mmol) and X-phos (32mg, 0.067mmol) and the mixture was warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, the mixture was diluted with EtOAc (60 mL) and washed with brine (40 mL. Times.2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give 3- ((S) -1-aminoethyl) -8- (((R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazol-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (40mg, 61%) was a yellow solid. MS (ESI) 394.0[ m ] +H] +
Step 10) 2-amino-N- ((S) -1- (8- (((R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1,2-b ] pyrazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To 3- ((S) -1-aminoethyl) -8- (((R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1, 2-b)]Pyrazol-3-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (40mg, 0.1mmol) and 2-aminopyrazolo [1,5-a]To a solution of pyrimidine-3-carboxylic acid (19.5mg, 0.1mmol) in DCM (20 mL) were added EDCI (31.5mg, 0.116mmol), HOAt (18mg, 0.13mmol), and DIEA (43mg, 0.32mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (40 mL) and washed with brine (20mL × 2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (ACN-H) 2 O,0.1% FA, gradient 20% to 50%) to give 2-amino-N- ((S) -1- (8- (((R) -5-hydroxy-5, 6-dihydro-4H-pyrrolo [1, 2-b) ]Pyrazol-3-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide (14.6 mg,23% yield) was a pale yellow solid. MS (ESI) 571.2[ m ] +H] +1 H NMR(400MHz,CD 3 OD)δ8.62(dd,J=6.8,1.6Hz,1H),8.43(dd,J=4.4,1.6Hz,1H),8.14(d,J=6.8Hz,1H),7.61(s,1H),7.57-7.54(m,2H),7.52-7.48(m,2H),7.45-7.33(m,4H),6.89(dd,J=6.8,4.4Hz,1H),6.80(s,1H),4.93-4.90(m,1H),4.72-4.65(m,1H),4.24(dd,J=11.6,5.6Hz,1H),3.88(dd,J=11.6,2.4Hz,1H),3.19(d,J=6.4Hz,1H),2.82(dd,J=16.8,2.4Hz,1H),1.39(d,J=6.8Hz,3H)。
Example 34 (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2- (phenyl-d 5) -1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000591
Step 1) 2-chloro-3-fluoro-6-methyl-N- (phenyl-d 5) benzamide
To a solution of 2-chloro-3-fluoro-6-methylbenzoic acid (1.0 g,5.3 mmol) and DMF (40mg, 0.53mmol) in DCM (25 mL) at 0 deg.C was added oxalyl chloride (8 mg, 0.53mmol) dropwise10mg, 6.36mmol), and the mixture was stirred for 2h. The mixture was concentrated under reduced pressure to give a mixture of acid chlorides. To a solution of benzene-d 5-amine (550mg, 5.3mmol) and TEA (1.61g, 15.9mol) in DCM (25 mL) at 0 deg.C was added a solution of the above acid chloride in DCM (10 mL), and the mixture was warmed to room temperature and stirred for 5h. For mixtures H 2 O (30 mL) was diluted and extracted with DCM (30mL × 3). The combined organic layers were washed with brine (50mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (PE/EA = 3/1) to give 2-chloro-3-fluoro-6-methyl-N- (phenyl-d 5) benzamide (1.01g, 64% yield) as a yellow solid. MS (ESI) 269.1[ alpha ] M + H ] +
Step 2) (S) - (4- (3-chloro-4-fluoro-2- ((phenyl-d 5) carbamoyl) phenyl) -3-oxobutan-2-yl) carbamic acid tert-butyl ester
A THF (25 mL) solution of 2-chloro-3-fluoro-6-methyl-N- (phenyl-d 5) benzamide (1.01g, 3.8mmol) was cooled to-30 ℃ under a nitrogen atmosphere, N-BuLi (4.0mL, 9.5mmol) was added thereto, and the mixture was stirred for 1 hour to obtain a yellow liquid. To a solution of (S) - (1- (methoxy (methyl) amino) -1-oxopropan-2-yl) carbamic acid tert-butyl ester (1.32g, 5.7 mmol) in THF (25 mL) at-30 deg.C was added a solution of isopropyl magnesium chloride (8.8mL, 11.4 mmol) and the mixture was stirred for 1h. The mixture was then slowly added to the above yellow mixture, transferred to room temperature, and stirred for 3 hours. The mixture is treated with NH 4 Aqueous Cl (30 mL) was diluted and extracted with EtOAc (50mL × 3). The combined organic layers were washed with brine (40mL. Times.2) and Na 2 SO 4 Drying, and concentrating under reduced pressure. The residue was purified by column chromatography (PE/EA = 3/1) to give tert-butyl (S) - (4- (3-chloro-4-fluoro-2- ((phenyl-d 5) carbamoyl) phenyl) -3-oxobutan-2-yl) carbamate (0.63g, 37% yield) as a yellow solid. MS (ESI) 462.1[ 2 ], [ M ] +Na ]] +
Step 3) (S) -3- (1-aminoethyl) -8-chloro-7-fluoro-2- (phenyl-d 5) isoquinolin-1 (2H) -one
To a solution of tert-butyl (S) - (4- (3-chloro-4-fluoro-2- ((phenyl-d 5) carbamoyl) phenyl) -3-oxobutan-2-yl) carbamate (0.63g, 1.43mmol) in MeOH (30 mL) was added concentrated HCl (15 mL), and the mixture was warmed to 80 deg.C and stirred for 24h. After cooling to room temperature, the mixture was concentrated under reduced pressure. The residue is substituted by H 2 O (10 mL) diluted and NaHCO 3 The pH value of the aqueous solution is adjusted to 7-8. The mixture was extracted with DCM (60mL × 3) and washed with brine (50mL × 2). The separated organic layer was washed with Na 2 SO 4 Dried and concentrated under reduced pressure. The residue was purified by column chromatography (DCM/MeOH = 95/5) to give (S) -3- (1-aminoethyl) -8-chloro-7-fluoro-2- (phenyl-d 5) isoquinolin-1 (2H) -one (160mg, 33% yield) as a yellow solid. MS (ESI) 321.9[ 2 ], [ M ] +H] +
Step 4) (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2- (phenyl-d 5) isoquinolin-1 (2H) -one
To a solution of (S) -3- (1-aminoethyl) -8-chloro-7-fluoro-2- (phenyl-d 5) isoquinolin-1 (2H) -one (160mg, 0.50mmol) and 4-ethynyl-1-methyl-1H-pyrazole (79mg, 0.75mmol) in MeCN (10 mL) was added X-Phos (118mg, 0.25mmol), pd 2 (dba) 3 (72mg, 0.12mmol) and K 3 PO 4 (317mg, 1.50mmol) and the mixture is warmed to 80 ℃ and stirred for 5h. After cooling to room temperature, concentrate in vacuo. The residue was diluted with EtOAc (80 mL) and washed with brine (50mL × 2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2- (phenyl-d 5) isoquinolin-1 (2H) -one (110mg, 53% yield) as a yellow oil. MS (ESI) 391.9[ 2 ] M + H ] +
Step 5) (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2- (phenyl-d 5) -1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To a mixture of (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -2- (phenyl-d 5) isoquinolin-1 (2H) -one (110mg, 0.28mmol) and 2-amino-5-methylpyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid (55mg, 0.31mmol) in DCM (30 mL) were added DIEA (109mg, 0.84mmol), EDCI (81mg, 0.42mmol) and HOAt (57mg, 0.42mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (60 mL) and washed with brine (40mL × 2). The separated organic layer was passed over Na 2 SO 4 Drying and curingConcentrating under vacuum. The residue was purified by preparative HPLC (-Gemini-C18 150x 19mm,5um 3 ) 20-50) to obtain (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2- (phenyl-d 5) -1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (31.8mg, 20% yield) was a yellow solid. MS (ESI) 552.1[ deg. ] M + H ]] +1 H NMR(400MHz,DMSO-d 6 )δ8.93(dd,J=6.8,1.6Hz,1H),8.55(dd,J=4.4,1.6Hz,1H),8.05(s,1H),8.00(d,J=6.8Hz,1H),7.70(ddd,J=21.2,13.2,7.2Hz,2H),7.61(s,1H),7.02(dd,J=6.8,4.4Hz,1H),6.79(s,1H),6.43(s,2H),4.58-4.50(m,1H),3.82(s,3H),1.34(d,J=6.8Hz,3H)。
Example 35 (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl-5-d) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000601
Step 1) 1-methyl-1H-pyrazole-5-d
To a solution of 5-bromo-1-methyl-1H-pyrazole (2g, 12.42mmol) in THF (10 mL) at-15 deg.C was added i-PrMgCl. LiCl (2.16mg, 14.9mmol in 1.3M in THF). The mixture is heated to-10 to 0 ℃ and stirred for 1 hour. Methanol-d 4 (0.5 mL) was added to the reaction mixture at-10 ℃ and stirred at room temperature for 16h. NH for the mixture 4 Aqueous Cl (5 mL) and MTBE (50 mL) quenched; the layers were separated and the organic phase was washed with brine (20mL. Times.2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtration, and concentration in vacuo afforded 1-methyl-1H-pyrazole-5-d (500mg, 48% yield) as a colorless oil. LCMS (ESI) m/z 84.1[ 2 ], [ M + H ]] +
Step 2) 4-iodo-1-methyl-1H-pyrazole-5-d
To a solution of 1-methyl-1H-pyrazole-5-d (500mg, 6mmol) in water (10 mL) was added elemental iodine (763mg, 3mmol), the mixture was stirred at room temperature for 0.5H, then H was added 2 O 2 (3 mL), the temperature was raised to 60 ℃ and the mixture was stirred for 1 hour. After cooling to room temperature, the reaction was quenched with aqueous sodium sulfite (30 mL). The mixture was extracted with EtOAc (50mL x 2), and the combined organic layers were washed with brine (50mL x 2) over anhydrous Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (PE: EA = 3) to give 4-iodo-1-methyl-1H-pyrazole-5-d (1g, 79% yield) as a yellow solid. LCMS MS (ESI) m/z 210.1[ 2 ] M + H ] +
Step 3) 1-methyl-4- ((trimethylsilyl) ethynyl) -1H-pyrazole-5-d
To a solution of 4-iodo-1-methyl-1H-pyrazole-5-d (1g, 4.78mmol) and ethynyltrimethylsilane (705mg, 7.18mmol) in THF (100 mL) at 0 ℃ were added CuI (91mg, 0.48mmol), pd (PPh) 3 ) 2 Cl 2 (168mg, 0.24mmol) and triethylamine (968mg, 9.57mmol), and the mixture was stirred at room temperature for 2 hours. The residue was diluted with EtOAc (300 mL) and washed with brine (200mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE: EA = 3)) to give 1-methyl-4- ((trimethylsilyl) ethynyl) -1H-pyrazole-5-d (700mg, 81% yield) as a yellow oil. LCMS MS (ESI) m/z 180.2[ 2 ], [ M + H ]] +
Step 4) 4-ethynyl-1-methyl-1H-pyrazole-5-d
To a solution of 1-methyl-4- ((trimethylsilyl) ethynyl) -1H-pyrazole-5-d (700mg, 3.9mmol) in THF (20 mL) was added TBAF (4 mL,4mmol,1M in THF), and the mixture was stirred at room temperature for 1H. The mixture was then diluted with EtOAc (50 mL) and washed with brine (30mL x 2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (PE: EA = 3) to give 4-ethynyl-1-methyl-1H-pyrazole-5-d (200mg, 47% yield) as a yellow oil. LCMS MS (ESI) m/z 108.0[ m + H ] ] +
Step 5) (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl-5-d) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To a solution of (S) -3- (1-aminoethyl) -8-chloro-7-fluoro-2-phenylisoquinoline-1 (2H) -one (50mg, 0.11mmol) and 4-ethynyl-1-methyl-1H-pyrazole-5-d (25mg, 0.24mmol) in MeCN (20 mL) was added Pd 2 (dba) 3 (29mg,0.032mmol), X-phos (30mg, 0.063mmol) and K 3 PO 4 (67mg, 0.32mmol), the mixture was warmed to 80 ℃ and stirred for 4h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM: meOH = 10) to give (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl-5-d) ethynyl) -2-phenylisoquinoline-1 (2H) -one (20mg, 32% yield) as a yellow solid. LCMS (ESI) m/z 388.1[ 2 ], [ M + H ]] +
Step 6) (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl-5-d) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To a solution of (S) -3- (1-aminoethyl) -7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl-5-d) ethynyl) -2-phenylisoquinoline-1 (2H) -one (20mg, 0.052mmol) and 2-aminopyrazolo [1,5-a ] ]A solution of pyrimidine-3-carboxylic acid (9mg, 0.052mmol) in DCM (15 mL) was added HOAt (10mg, 0.078mmol), EDCI (12mg, 0.078mmol) and DIEA (20mg, 0.155mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM: meOH = 10) to give (S) -2-amino-N- (1- (7-fluoro-8- ((1-methyl-1H-pyrazol-4-yl-5-d) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (7.3mg, 25% yield) was a white solid. LCMS MS (ESI) m/z 548.0[ 2 ] M + H] +1 H NMR(400MHz,CD 3 OD)δ8.72(dd,J=6.8,1.6Hz,1H),8.53(dd,J=4.4,1.6Hz,1H),7.69(s,1H),7.67-7.60(m,2H),7.59-7.42(m,5H),6.99(dd,J=6.8,4.4Hz,1H),6.91(s,1H),4.77(q,J=6.8Hz,1H),3.89(s,3H),1.48(d,J=6.8Hz,3H)。
Example 36 (S) -2-amino-N- (1- (5-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000611
Step 1) 6-chloro-2-methyl-3-nitrobenzoic acid
Nitric acid (4.1g, 0.064mol,2.6 mL) was added dropwise to a solution of 2-chloro-6-methylbenzoic acid (10g, 0.0586mol) in sulfuric acid (100 mL) at 0 ℃ and the mixture was stirred for 30 minutes, then, the mixture was transferred to room temperature and stirred for 16 hours. The mixture was poured slowly into ice water (260 mL) and stirring was continued for 1h. Filtration and washing of the filter cake with water (50 mL) and vacuum drying gave 6-chloro-2-methyl-3-nitrobenzoic acid (12.5 g, 98% yield) as a white solid. LCMS (ESI) m/z 214.4[ m-H ] ] -
Step 2) 6-chloro-2-methyl-3-nitro-N-phenylbenzamide
To a solution of 6-chloro-2-methyl-3-nitrobenzoic acid (11.5g, 0.053 mol) in DCM (60 mL) at 0 deg.C was added DMF (1 mL) and oxalyl chloride (8.2g, 0.064 mol), the mixture was transferred to room temperature and stirred for 2h; the mixture was concentrated in vacuo to give a white solid (13 g). The above white solid (13 g) was dissolved in DCM (20 mL) at 0 ℃ and aniline (7.5g, 0.08mol), triethylamine (5.4 g, 0.0533mol) and DCM (60 mL) were added and the mixture was stirred at room temperature for 16 hours. The mixture was quenched with water (200 mL) and extracted with DCM (200mL × 2). The combined organic layers were washed with brine (200mL. Times.2) over anhydrous Na 2 SO 4 Drying, filtration, and vacuum concentration gave 6-chloro-2-methyl-3-nitro-N-phenylbenzamide (14g, 89% yield) as a yellow solid. LCMS (ESI) m/z 291.0[ 2 ], [ M + H ]] +
Step 3) 3-amino-6-chloro-2-methyl-N-phenylbenzamide
To a solution of 6-chloro-2-methyl-3-nitro-N-phenylbenzamide (15g, 0.051mol) in MeOH (200 mL) was added SnCl 2 (29.7 g, 0.155mol), the mixture was heated to reflux and stirred for 24h. After cooling to room temperature, it was concentrated in vacuo. The residue is taken up in NaHCO 3 Aqueous (200 mL) was quenched and extracted with EtOAc (300mL. Times.2). The combined organic layers were washed with brine (200mL. Times.2) over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 3/1)) to give 3-amino group-6-chloro-2-methyl-N-phenylbenzamide (7g, 51% yield) as a white solid. LCMS (ESI) m/z 261.1[ 2 ], [ M + H ]] +
Step 4) 6-chloro-3-fluoro-2-methyl-N-phenylbenzamide
To a solution of 3-amino-6-chloro-2-methyl-N-phenylbenzamide (2g, 7.67mmol) in tetrafluoroboric acid (48 mass%,20 mL) at 0 deg.C was slowly added dropwise an aqueous solution (5 mL) of sodium nitrate (1g, 15.34mmol), and after completion of the addition, the mixture was transferred to room temperature and stirred for 1h. Filtration, rinsing of the filter cake with cold water (5mL x 2), and vacuum drying afforded the diazonium salt as a white solid (2 g). The diazonium salt was heated at 120 ℃ for 30 minutes, cooled to room temperature, poured into ice water (50 mL), and then extracted with ethyl acetate (50 mL. Times.3). The combined organic layers were washed with brine (50mL. Times.2) and over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 10/1)) to give 6-chloro-3-fluoro-2-methyl-N-phenylbenzamide (800 mg, yield 35%) as a white solid. LCMS MS (ESI) m/z 264.1[ 2 ] M + H] +
Step 5) (S) - (4- (3-chloro-6-fluoro-2- (phenylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamic acid tert-butyl ester
To a solution of 6-chloro-3-fluoro-2-methyl-N-phenylbenzamide (400mg, 1.52mmol) in THF (5 mL) at-30 ℃ was added N-BuLi (3.8mmol, 2.4m/L =1.58 mL) and the mixture was stirred for 2h to give a yellow liquid. To a solution of N- ((1S) -1- (methoxy (methyl) carbamoyl) ethyl) carbamic acid tert-butyl ester (528mg, 2.27mmol) in THF (10 mL) at-30 ℃ under a nitrogen atmosphere was added i-PrMgCl (4.55mmol, 1.3M/L,3.5 mL) and the mixture was stirred for 2h. To this mixture was added the above yellow liquid at-40 ℃, transferred to room temperature and stirred for 16h. NH for the mixture 4 The Cl solution (50 mL) was quenched and extracted with EtOAc (50 mL. Times.2). The combined organic layers were washed with brine (50mL. Times.2) and over anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (PE/EA = 3/1) to give tert-butyl (S) - (4- (3-chloro-6-fluoro-2- (phenylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamate (160mg, 24% yield) as a yellow solid. LCMS (ESI) M/z457.0[ M + Na ]] +
Step 6) (S) -3- (1-aminoethyl) -8-chloro-5-fluoro-2-phenylisoquinolin-1 (2H) -one
To a solution of tert-butyl (S) - (4- (3-chloro-6-fluoro-2- (phenylcarbamoyl) phenyl) -3-oxobutan-2-yl) carbamate (150mg, 0.345 mmol) in MeOH (6 mL) was added concentrated hydrochloric acid (3 mL) and the mixture was warmed to 70 deg.C and stirred for 3h. After cooling to room temperature, it was concentrated in vacuo. The residue was poured into water (50 mL) and the resulting solution was neutralized with aqueous sodium bicarbonate to pH =8. The mixture was extracted with EtOAc (50mL x 3), and the combined organic layers were washed with brine (50 mL) and Na-free 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 95/5) to give (S) -3- (1-aminoethyl) -8-chloro-5-fluoro-2-phenylisoquinolin-1 (2H) -one (50mg, 45% yield) as a white solid. LCMS (ESI) m/z 317.0[ 2 ], [ M + H ]] +
Step 7) (S) -3- (1-aminoethyl) -5-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one
To a solution of (S) -3- (1-aminoethyl) -8-chloro-5-fluoro-2-phenylisoquinolin-1 (2H) -one (50mg, 0.23mmol) and 4-ethynyl-1-methylpyrazole (26mg, 0.23mmol) in acetonitrile (20 mL) under a nitrogen atmosphere was added Pa 2 (dba) 3 (36mg, 0.039mmol), X-phos (38mg, 0.039mmol) and K 3 PO 4 (67mg, 0.32mmol), the mixture was warmed to 80 ℃ and stirred for 4h. After cooling to room temperature, concentrate in vacuo. The residue was dissolved in DCM (50 mL) and washed with brine (30 mL). The separated organic layer was washed with anhydrous Na 2 SO 4 Drying, filtering and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 10/1) to give (S) -3- (1-aminoethyl) -5-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (50mg, 80% yield) as a yellow solid. LCMS MS (ESI) m/z 390.1[ 2 ] M + H] +
Step 8) (S) -2-amino-N- (1- (5-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -5-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H)-ketone (50mg, 0.13mmol) and 2-aminopyrazolo [1,5-a]To a solution of pyrimidine-3-carboxylic acid (23mg, 0.13mmol) in DCM (15 mL) were added HOAt (26mg, 0.19mmol), EDCI (30mg, 0.19mmol) and DIEA (50mg, 0.39mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL) and washed with brine (30mL × 2). The separated organic layer was passed over anhydrous Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM: meOH = 15) to give (S) -2-amino-N- (1- (5-fluoro-8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (24.5mg, 34% yield) was a white solid. LCMS (ESI) m/z 550.1[ 2 ], [ M + H ]] + . 1 H NMR(400MHz,DMSO-d 6 )δ8.94(dd,J=6.8,1.6Hz,1H),8.56(dd,J=4.4,1.6Hz,1H),8.08(d,J=6.8Hz,1H),8.00(d,J=0.8Hz,1H),7.62-7.49(m,7H),7.43(d,J=7.2Hz,1H),7.03(dd,J=6.8,4.4Hz,1H),6.75(s,1H),6.45(s,2H),4.55(t,J=6.8Hz,1H),1.36(d,J=6.8Hz,3H)。
Example 37 (S) -2-amino-5- (methoxy-d 3) -N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
Figure BDA0003866228850000621
Step 1) (S) -3- (1-aminoethyl) -8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one
To 3- ((1S) -1-aminoethyl) -8-chloro-2-phenylisoquinolin-1-one (100mg, 0.33mmol) and 4-ethynyl-1-methylpyrazole (55mg, 0.5mmol) in CH 3 Adding Pd into CN (20 mL) solution 2 (dba) 3 (61mg,0.067mmol)、K 3 PO 4 (142mg, 0.67mmol) and X-phos (64mg, 0.13mmol), the mixture was warmed to 80 ℃ and stirred for 5 h. After cooling to room temperature, the mixture was diluted with EtOAc (60 mL) and washed with brine (40 mL. Times.2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to giveTo (S) -3- (1-aminoethyl) -8- ((1- (methyl-d 3) -1H-pyrazol-4-yl)) ethynyl) -2-phenylisoquinolin-1 (2H) -one (80mg, 61%) as a yellow solid. MS (ESI) 371.9[ 2 ] M + H] +
Step 2) (S) -2-amino-5- (methoxy-d 3) -N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (40mg, 0.1mmol) and 2-amino-5-methoxypyrazolo [1,5-a ]]To a solution of pyrimidine-3-carboxylic acid (23mg, 0.1mmol) in DCM (20 mL) were added EDCI (31mg, 0.11mmol), HOAt (18mg, 0.13mmol) and DIEA (42mg, 0.32mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (40 mL) and washed with brine (30mL × 2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by preparative HPLC (DCM/MeOH = 15/1) to give (S) -2-amino-5- (methoxy-d 3) -N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1, 5-a)]Pyrimidine-3-carboxamide (19mg, 30% yield) was a white solid. MS (ESI) 564.8[ solution of ] M + H] +1 H NMR(400MHz,DMSO-d 6 )δ8.72-8.66(m,1H),8.01(d,J=0.8Hz,1H),7.88(d,J=6.8Hz,1H),7.68-7.61(m,2H),7.60(s,1H),7.59-7.50(m,4H),7.46(d,J=7.6Hz,1H),7.39(d,J=7.6Hz,1H),6.79(s,1H),6.49-6.44(m,1H),6.14(s,2H),4.49-4.43(m,1H),1.36(d,J=6.8Hz,3H)。
Example 38 (S) -2-amino-5-cyano-N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyridine-3-carboxamide
Figure BDA0003866228850000631
Step 1) 1-amino-4-bromopyridin-1-ium
To a solution of 4-bromopyridine (1g, 6.3mmol) in DCM (10 mL) was added portionwise aminodiphenylphosphinate (4.41g, 18.9)mmol), the mixture was stirred at room temperature for 48h. Hydriodic acid (1mL, 55% H) was then added 2 Solution O), the precipitate was filtered, and the filter cake was washed with DCM (10 mL) and dried in vacuo to give 1-amino-4-bromopyridin-1-ium (1g, 46% yield) as a brown solid. MS (ESI) 173.0,175.0[ 2 ] M + H] +
Step 2) dimethyl 5-bromopyrazolo [1,5-a ] pyridine-2, 3-dicarboxylate
To a solution of 1-amino-4-bromopyridin-1-ium (800mg, 4.6 mmol) in DMF (20 mL) was added 1, 4-dimethylsuccinic acid (784 mg,5.5 mmol) and K 2 CO 3 (953mg, 6.89mmol) and the mixture was stirred at RT for 16h. The mixture was diluted with EtOAc (80 mL) and washed with brine (40 mL. Times.2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/5) to give 5-bromopyrazolo [1,5-a ]]Dimethyl pyridine-2, 3-dicarboxylate (200mg, 13% yield) was a yellow solid. MS (ESI) 313.0,315.0[ m ] +H] +
Step 3) 5-bromo-3- (methoxycarbonyl) pyrazolo [1,5-a ] pyridine-2-carboxylic acid
To 5-bromopyrazolo [1,5-a ]]To a solution of dimethyl pyridine-2, 3-dicarboxylate (200mg, 0.19mmol) in THF (10 mL) was added NaOH (26mg, 0.64mmol) in H 2 O (3 mL) solution, and the mixture was stirred at room temperature for 16h. The mixture was concentrated in vacuo and the residue was diluted with water (10 mL) and acidified to pH 4-5 with 1N HCl. Filtering the precipitate, washing the filter cake with water, and vacuum drying to obtain 5-bromo-3- (methoxycarbonyl) pyrazolo [1,5-a]Pyridine-2-carboxylic acid (180 mg, yield 89%) was a white solid. MS (ESI) 299.0,301.0[ 2 ] M + H] +
Step 4) methyl 5-bromo-2- ((tert-butoxycarbonyl) amino) pyrazolo [1,5-a ] pyridine-3-carboxylate
To 5-bromo-3- (methoxycarbonyl) pyrazolo [1,5-a]To a solution of pyridine-2-carboxylic acid (230mg, 0.77mmol) in dioxane (20 mL) were added triethylamine (233mg, 2.3mmol) and DPPA (254mg, 0.92mmol), and the mixture was stirred at room temperature for 1h. t-BuOH (114mg, 1.53mmol) was then added, the temperature was raised to 100 ℃ and stirred for 16h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with EtOAc (60 mL) and washed with brine (40mL × 2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/5) to give 5-bromo-2- ((tert-butoxycarbonyl) amino) pyrazolo [1,5-a]Pyridine-3-carboxylic acid methyl ester (150mg, 53% yield) and 2-amino-5-bromopyrazolo [1,5-a ]]Methyl pyridine-3-carboxylate (50mg, 17.5% yield) was a white solid. MS (ESI) 392.0,394.0[ 2 ] M + H] + ,270.0,272.0[M+H] + . Step 5) 2-amino-5-bromopyrazolo [1,5-a]Pyridine-3-carboxylic acids
To 2-amino-5-bromopyrazolo [1,5-a ]]To a solution of methyl pyridine-3-carboxylate (50mg, 0.18mmol) in MeOH (10 mL) was added an aqueous solution of NaOH (22mg, 0.55mmol) (2 mL), and the mixture was stirred at 50 ℃ for 3h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was diluted with water (5 mL) and acidified with 1N HCl to pH =4-5. Then extracted with EtOAc (20mL x 3), and the combined organic layers were washed with brine (20mL x 2) and then washed with Na 2 SO 4 Drying and vacuum concentrating to obtain 2-amino-5-bromopyrazolo [1,5-a]Pyridine-3-carboxylic acid (45mg, 90% yield) was a light colored solid. MS (ESI) 255.9,257.9[ 2 ] M + H] +
Step 6) (S) -2-amino-5-bromo-N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyridine-3-carboxamide
To (S) -3- (1-aminoethyl) -8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinolin-1 (2H) -one (65mg, 0.18mmol) and 2-amino-5-bromopyrazolo [1,5-a ] ]To a solution of pyridine-3-carboxylic acid (45mg, 0.18mmol) in DCM (30 mL) were added HOAt (32mg, 0.23mmol), EDCI (56mg, 0.29mmol), and DIEA (75mg, 0.59mmol), and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (30 mL) and washed with brine (30mL × 2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (DCM/MeOH = 20/1) to give (S) -2-amino-5-bromo-N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] as]Pyridine-3-carboxamide (100mg, 76% yield) was a yellow solid. MS (ESI) 609.0[ 2 ] M + H] +
Step 7) (S) -2-amino-5-cyano-N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyridine-3-carboxamide
To (S) -2-amino-5-bromo-N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] under a nitrogen atmosphere]Pyridine-3-carboxamide (50mg, 0.08mmol) in DMF (5 mL) was added Zn (CN) 2 (19mg, 0.116mmol), zn (11mg, 0.116mmol), and Pd (dppf) Cl 2 DCM (7mg, 0.008mmol) and the mixture was heated to 100 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with EtOAc (60 mL) and washed with brine (30 mL. Times.3). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (ACN-H) 2 O,0.1FA, gradient 40% -60%) to give (S) -2-amino-5-cyano-N- (1- (8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ]]Pyridine-3-carboxamide (7.8mg, 17% yield) was a yellow solid. MS (ESI) 556.1[ 2 ] M + H] + . 1 H NMR(400MHz,CD 3 OD)δ8.48(dd,J=6.8,0.8Hz,1H),8.18(d,J=0.8Hz,1H),7.87(s,1H),7.69(s,1H),7.67-7.52(m,7H),7.44(d,J=7.6Hz,1H),7.04(dd,J=7.2,1.6Hz,1H),6.94(s,1H),4.76(q,J=6.8Hz,1H),1.50(d,J=6.8Hz,3H)。
Example 39 (S) -2-amino-5-methoxy-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyridine-3-carboxamide
Figure BDA0003866228850000641
Step 1) methyl 2- ((tert-butoxycarbonyl) amino) -5-methoxypyrazolo [1,5-a ] pyridine-3-carboxylate
To 5-bromo-2- ((tert-butoxycarbonyl) amino) pyrazolo [1,5-a]To a solution of methyl pyridine-3-carboxylate (100mg, 0.27mmol) and sodium methoxide (44mg, 0.81mmol) in dioxane (10 mL) and MeOH (5 mL) was added Pd 2 (dba) 3 (25mg, 0.027mmol) and X-phos (26mg, 0.054 mmol), the mixture was warmed to 70 ℃ and stirred 1And 6h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with EtOAc (50 mL) and washed with brine (30mL x 2). The separated organic layer was washed with Na 2 SO 4 Drying and vacuum concentrating. The residue was purified by column chromatography (EA/PE = 1/2) to give 2- ((tert-butoxycarbonyl) amino) -5-methoxypyrazolo [1, 5-a) ]Pyridine-3-carboxylic acid methyl ester (80mg, 73.7% yield) was a yellow solid. MS (ESI) 344.1[ m ] +Na] +
Step 2) methyl 2-amino-5-methoxypyrazolo [1,5-a ] pyridine-3-carboxylate
To 2- ((tert-butoxycarbonyl) amino) -5-methoxypyrazolo [1,5-a ]]To a solution of pyridine-3-carboxylic acid methyl ester (80mg, 0.25mmol) in DCM (3 mL) was added TFA (1 mL), and the mixture was stirred at RT for 2h. The mixture was concentrated in vacuo to give 2-amino-5-methoxypyrazolo [1,5-a ]]Pyridine-3-carboxylic acid methyl ester (60 mg, yield 87%) was a yellow oil. MS (ESI) 222.1[ 2 ] M + H] +
Step 3) 2-amino-5-methoxypyrazolo [1,5-a ] pyridine-3-carboxylic acid
To 2-amino-5-methoxypyrazolo [1,5-a ]]To a solution of pyridine-3-carboxylic acid methyl ester (60mg, 0.27mmol) in MeOH (10 mL) was added aqueous NaOH (32mg, 0.81mmol) (3 mL), and the mixture was warmed to 65 deg.C and stirred for 48h. After cooling to room temperature, it was concentrated in vacuo. The residue was diluted with water (3 mL) and acidified to pH =4-5 with 1N HCl and the precipitate collected by filtration. The filter cake was washed with water (2 mL) and dried under vacuum to give 2-amino-5-methoxypyrazolo [1,5-a ]]Pyridine-3-carboxylic acid (50 mg, yield 53%) was a yellow solid. MS (ESI) 208.1[ deg. ] M + H ]] +
Step 4) (S) -2-amino-5-methoxy-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ] pyridine-3-carboxamide
To a solution of (S) -3- (1-aminoethyl) -8- ((1- (methyl-d 3) -1H-pyrazol-4-yl) ethynyl) -2-phenylisoquinoline-1 (2H) -one (89mg, 0.24mmol) and 2-amino-5-methoxypyrazolo [1,5-a ]]EDCI (69mg, 0.36mmol), HOAt (49mg, 0.36mmol) and DIEA (93mg, 0.72mmol) were added to a solution of pyridine-3-carboxylic acid (50mg, 0.24mmol) in DCM (40 mL) and the mixture was warmed to 40 ℃ and stirred for 16h. After cooling to room temperature, the mixture was diluted with DCM (50 mL)And washed with brine (30mL x 2). The separated organic layer was passed over Na 2 SO 4 Drying and vacuum concentrating. The residue was subjected to preparative HPLC (ACN-H) 2 O,0.1FA, gradient 40% -60%) to give (S) -2-amino-5-methoxy-N- (1- (8- ((1-methyl-1H-pyrazol-4-yl) ethynyl) -1-oxo-2-phenyl-1, 2-dihydroisoquinolin-3-yl) ethyl) pyrazolo [1,5-a ]]Pyridine-3-carboxamide (6.5mg, 5% yield) was a white solid. MS (ESI) 561.2[ m ] +H] +1 H NMR(400MHz,CD 3 OD)δ8.15(d,J=7.6Hz,1H),7.86(s,1H),7.69(d,J=3.2Hz,1H),7.67–7.53(m,7H),7.43(d,J=7.6Hz,1H),7.10(d,J=2.4Hz,1H),6.94(s,1H),6.55(dd,J=7.6,2.7Hz,1H),4.74(q,J=6.8Hz,1H),3.90(s,3H),1.47(d,J=6.8Hz,3H)。
Biological assay
Kinase activity assay the activity of the compounds of the invention as PI3K and mTOR kinase inhibitors can be assessed by the following assay. General description of kinase assays are performed by detecting Myelin Basic Protein (MBP) that incorporates gamma-33P-ATP. mu.g/mL of MBP (Sigma # M-1891) Tris buffered saline (TBS; 50mM Tris pH 8.0,138mM NaCl,2.7mM KCl) was prepared, coated in high binding white 384 well plates (Greiner) at 60. Mu.L per well. Incubate at 4 ℃ for 24 hours. The plate was then washed 3 times with 100. Mu.L TBS. Kinase reaction kinase buffer (5 mM Hepes pH 7.6,15mM NaCl,0.01% bovine serum albumin (Sigma # I-5506), 10mM MgCl in a total volume of 34. Mu.L 2 1mM DTT, 0.02%; tritonX-100). Compounds were dissolved in DMSO and added to each well at a final DMSO concentration of 1%. Each data was assayed in two passes, with at least two trials for each compound assay. For example, the final concentration of the enzyme is 10nM or 20nM. The reaction was started by adding unlabeled ATP (10. Mu.M) and gamma-33P-labeled ATP (2X 106cpm per well, 3000 Ci/mmole). The reaction was allowed to shake at room temperature for 1 hour. The 384 well plates were washed with 7 XPBS and 50. Mu.L of scintillation fluid per well was added. It can also be measured by a Wallac Trilux meter.
IC inhibition by the test method 50 And/or the inhibition constant Ki. IC (integrated circuit) 50 Defined as the concentration of compound that inhibits 50% of the enzyme activity under the conditions tested. By using1/2log dilution A curve containing 10 concentration points was made and IC was estimated 50 Values (e.g., a typical curve is made by compound concentrations of 10. Mu.M, 3. Mu.M, 1. Mu.M, 0.3. Mu.M, 0.1. Mu.M, 0.03. Mu.M, 0.01. Mu.M, 0.003. Mu.M, 0.001. Mu.M and 0. Mu.M).
General assay protocol for PI3 kinase
PI3K (p 110. Alpha./p 85. Alpha.) (h) [ non-radioactive assay]
PI3K (p 110. Alpha./p 85. Alpha.) (h) was incubated in a buffer solution containing 10. Mu.M phosphoinositide-4, 5-diphosphate and MgATP (concentration determined as required). After addition of ATP solution, the reaction was started. After incubation at room temperature for 30 minutes, a stop solution containing EDTA and biotin phosphatidylinositol-3, 4, 5-triphosphate was added thereto to terminate the reaction. Finally, a detection buffer comprising europium-labeled anti-GST monoclonal antibody, GST-labeled GRP1PH domain and streptavidin-allophycocyanin was added. The plates were read in time resolved fluorescence mode and Homogeneous Time Resolved Fluorescence (HTRF) signals were determined by the equation HTRF =10000 × (Em 665 nm/Em620 nm).
PI3K (p 110. Beta./p 85. Alpha.) (h) [ non-radioactive assay]
PI3K (p 110. Beta./p 85. Alpha.) (h) was incubated in a buffer solution containing 10. Mu.M phosphoinositide-4, 5-diphosphate and MgATP (concentrations determined as required). After addition of ATP solution, the reaction was started. After incubation at room temperature for 30 minutes, a stop solution containing EDTA and biotin phosphatidylinositol-3, 4, 5-triphosphate was added thereto to terminate the reaction. Finally, a detection buffer solution comprising europium-labeled anti-GST monoclonal antibody, GST-labeled GRP1PH domain and streptavidin-allophycocyanin is added. The plates were read in time resolved fluorescence mode and Homogeneous Time Resolved Fluorescence (HTRF) signals were determined by the equation HTRF =10000 × (Em 665 nm/Em620 nm).
PI3K (p 110. Delta./p 85. Alpha.) (h) [ non-Radioactive test]
PI3K (p 110. Delta./p 85. Alpha.) (h) was incubated in a buffer solution containing 10. Mu.M phosphoinositide-4, 5-diphosphate and MgATP (concentration determined as required). After addition of ATP solution, the reaction was started. After incubation at room temperature for 30 minutes, a stop solution containing EDTA and biotin phosphatidylinositol-3, 4, 5-trisphosphate was added thereto to stop the reaction. Finally, a detection buffer solution comprising europium-labeled anti-GST monoclonal antibody, GST-labeled GRP1PH domain and streptavidin-allophycocyanin is added. The plates were read in time-resolved fluorescence mode and homogeneous time-resolved fluorescence (HTRF) signal was determined by the equation HTRF =10000 × (Em 665 nm/Em620 nm).
PI3K (p 120. Gamma.) (h) [ non-radioactive assay]
PI3K (p 120. Gamma.) (h) was incubated in a buffered solution containing 10. Mu.M phosphoinositide-4, 5-diphosphate and MgATP (concentration determined as required). After addition of ATP solution, the reaction was started. After incubation at room temperature for 30 minutes, a stop solution containing EDTA and biotin phosphatidylinositol-3, 4, 5-trisphosphate was added thereto to stop the reaction. Finally, a detection buffer comprising europium-labeled anti-GST monoclonal antibody, GST-labeled GRP1PH domain and streptavidin-allophycocyanin was added. The plates were read in time resolved fluorescence mode and Homogeneous Time Resolved Fluorescence (HTRF) signals were determined by the equation HTRF =10000 × (Em 665 nm/Em620 nm).
The kinase assay in the present invention was carried out by the company Eurofins France (Eurofins Cerep SA, le Bois L 'Ev E que,86600Celle L' Eescault, france) and the results are shown in Table 1, where, +: >100nM; ++:50-100nM; +++:10-50nM; ++++: < 10nM.
TABLE 1 kinase inhibition data for compounds of the invention
Figure BDA0003866228850000661
Figure BDA0003866228850000671
Test results show that the compound has good inhibitory activity on PI3K gamma kinase and high selectivity for other subtypes.
Cell viability assay
The cellular activity of the compounds of the invention as inhibitors of PI3K kinase can be assessed by the following assay.
General description of cell assays:
PI3K- α, γ subtype: first compounds were diluted from stock concentration to 5mM with 100% DMSO. The second step uses 5mM of the compound as the first point and 10 points of dilution with 100% DMSO 4-fold. The third step was a 250-fold dilution with serum-free medium, in which case the concentration of DMSO was 0.4%. Then 50. Mu.L of the compound which had been diluted with the culture medium was transferred to a 50. Mu.L cell plate, where the concentration of DMSO was 0.2% and the final concentration of the compound was 10000nM,2500nM,625nM,156.25nM,39.06nM,9.77nM,2.44nM,0.61nM,0.15nM,0.04nM.
PI3K- β, δ subtype: compounds were first diluted from stock concentrations to 1.25mM with 100% DMSO. Second step 1.25mM of compound as first point was diluted 4-fold with 100% DMSO for 10 points. The third step was 35.714-fold dilution with serum-free medium, then transferred to 2.5. Mu.L of the compound diluted with the medium into a 30. Mu.L cell plate, incubated in an incubator for 1 hour, and then added with 2.5. Mu. Lanti-IgM at a DMSO concentration of 0.2% and at a final concentration of 2500nM,625nM,156.25nM,39.06nM,9.77nM,2.44nM,0.61nM,0.15nM,0.04nM,0.01nM.
PI 3K-alpha inhibitory activity detection method
SKOV-3 cells were seeded into cell culture-grade 96-well plates at a density of 60000 cells/50. Mu.L/well in serum-free RPMI-1640, cultured overnight in a 5% CO2 incubator at 37 ℃. Add 50. Mu.L/well of test compound to the cells and incubate them at 37 ℃ for 60 minutes in a 5% CO2 incubator to a final DMSO concentration of 0.2%, aspirate the medium and add 50. Mu.L of 1 Xlysate per well. Shake for 45 minutes at room temperature. mu.L of lysate was transferred to 384 plates and 4. Mu.L of premixed antibody from the Phospho-AKT (Ser 473) kit from Cisbio was added. Centrifuge at 1000rpm/min for one minute, incubate at 22 ℃ for 4 hours, and read with Spark (665 nm/615 nm).
PI 3K-beta inhibitory activity detection method
786-O cells were seeded at a density of 30000/50. Mu.L/well in 96-well cell culture grade plates in RPMI-1640 medium without serum, cultured overnight in a CO2 incubator at 37 ℃ 5%. Adding 50 μ L/well of test compound to the cells, incubating for 60 minutes in a 5% CO2 incubator at 37 deg.C, to a final DMSO concentration of 0.2%, aspirating the medium and adding 50 μ L of 1 Xlysate per well. Shaking for 45 minutes at room temperature. mu.L of lysate was transferred to 384 plates and 4. Mu.L of premixed antibody from the Phospho-AKT (Ser 473) kit from Cisbio was added. Centrifuge at 1000rpm/min for one minute, incubate at 22 ℃ for 4 hours, and read with Envision (665 nm/615 nm).
PI 3K-delta inhibitory activity detection method
Raji cells were cultured in 96-well plates at 30 μ L per well, 50,000 cell species in cell culture medium RPMI-1640 without serum. Cells were incubated overnight in 5% CO2 and 37 ℃ incubator. After 18 hours of serum-free starvation, 2.5 μ L of compound (14X) was added to the cells and incubated in an incubator for 60 minutes. Then 2.5. Mu.L (14X, diluted with serum medium) of anti-human IgM (Jackson Immuno Research) was added and placed in an incubator for 30 minutes of stimulation (final concentration 10. Mu.g/mL). Add 11.5. Mu.L of 4 Xlysate to each well. Shaking for 45 minutes at room temperature. mu.L of lysate was added to the transfer plates 384, and 4. Mu.L of premixed antibody from the Phospho-AKT (Ser 473) kit from Cisbio was added. Centrifuge at 1000rpm/min for one minute, incubate at 22 ℃ for 4 hours, and read with Spark (665 nm/615 nm).
PI 3K-gamma inhibitory activity detection method
RAW264.7 cells were resuspended in serum-free DMEM medium and 60000/45. Mu.L of cell suspension was added per well in a 96-well plate. The cells were incubated overnight in an incubator at 37 ℃ with 5% CO2. After 18 hours of serum-free starvation, 50 μ L of compound was added and incubated in an incubator for 60min. Then 5. Mu.L of 25nM C5a (R & D Systems, diluted in serum medium) was added for stimulation for 5min. Aspirate medium 50. Mu.L of 1 Xlysate per well. Shaking for 45 minutes at room temperature. mu.L of lysate was transferred to 384 plates and 4. Mu.L of premixed antibody from Phospho-AKT (Ser 473) kit from Cisbio was added. Centrifuge at 1000rpm/min for one minute, 22 ℃ after 4 hours of incubation read with Spark (665 nm/615 nm).
The kinase assay of the present invention was performed by the Biotech company, bonus Biotech (Jiangsu), inc., and the results are shown in Table 2, wherein +: >500nM; ++:100-500nM; +++:50-100nM; ++++: < 50nM.
The synthesis of positive drug IPI-549 is referred to patent WO2015143012A1. The chemical structural formula is as follows:
Figure BDA0003866228850000681
TABLE 2 cellular activity data for compounds of the invention
Figure BDA0003866228850000682
Test results show that the compound has obvious inhibitory activity on PI3K gamma and has very obvious selectivity on other PI3K subtypes. The compound of the invention has higher selectivity compared with positive drug IPI-549.
Metabolic stability Studies of Compounds of the invention in human liver microsomes
The metabolic stability of the compounds of the present invention as PI3K kinase inhibitors can be evaluated by the following assay.
General description of the liver microsome stability assay:
the test compound was co-incubated with human liver microsomes, NADPH was added and the reaction was initiated. At 0,5, 15, 30 and 60 minutes, 30 microliters were removed and transferred to 300 microliters of acetonitrile containing an internal standard to terminate the reaction. Followed by vigorous vortexing for 1 minute and centrifugation at 4000rpm below 4 ℃ for 15 minutes. After the protein was precipitated, 100. Mu.l of the supernatant was taken out, diluted with 100. Mu.l of distilled water, and analyzed by LC-MS/MS method. Intrinsic clearance in vitro was calculated from the clearance half-life of the test compound in the incubation system. Verapamil was used as a control compound, and both were incubated for 5 minutes in parallel.
And (3) data analysis:
the Remaining percentage (% Remaining) is calculated from the ratio of the peak areas of the test compound in the non-zero time point sample and the zero time sample. Half-life (T) of the test compound clearance 1/2 Min) and in vitro intrinsic clearance (C) Lint ,μL.min -1 .mg -1 ) This is given by the following equation:
Figure BDA0003866228850000691
/>
Figure BDA0003866228850000692
Figure BDA0003866228850000693
the relevant parameters of the compounds of the examples of the present invention and the stability of human liver microsomes of IPI-549 are shown in Table 3.
TABLE 3 relevant parameters for human liver microsome stability of the compounds of the invention
Compound (I) Half life (T) 1/2 ,min)
IPI-549 122.6
Example 13 63.7
Example 16 154.7
Example 21 77.2
Example 28 656.9
Example 37 94.7
The test result shows that compared with IPI-549, the half-life period of the invention in the embodiment 16 and the embodiment 28 is obviously prolonged, and the stability of the human liver microsome is obviously improved. Compared with the examples 16 and 28, when the hydrogen of the methyl group on the pyrazole is replaced by deuterium, the stability of the human liver microsome is obviously improved; similarly, the stability of human liver microsomes is significantly improved by replacing the hydrogen of the methyl group on pyrazole with deuterium in examples 21 and 37 of the present invention. Study of the oral pharmacokinetics of the Compound CD-1 of the present invention in mice
The pharmacokinetic studies of the compounds of the invention as PI3K kinase inhibitors can be evaluated by the following assays.
General description of pharmacokinetic experiments:
CD-1 mice are taken as test animals, and the drug concentration in blood plasma at different times is determined by measuring the gavage administration of the compound by adopting an LC-MS/MS method. The intravenous injection dosage and the formula are as follows: 2mg/kg,5% NMP/40% PEG400/55% PBS; oral dosage administration and formulation: 10.0mg/kg,0.5% (w/v) hydroxymethylcellulose and 0.05% Tween 80 in water. Weighing an appropriate amount of test compound, adding an appropriate amount of an aqueous solution of 0.5% (w/v) hydroxymethylcellulose and 0.05% Tween 80, vortexing and sonicating, and finally formulating into a test solution having a concentration of 1 mg/mL. The sampling time of the intravenous injection group is as follows: 0.0833,0.25,0.5,1,2,4,8,24h post-administration. The oral group sampling times were: 0.25,0.5,1,2,4,8,24h. And finally, determining the content of the compound to be tested in the plasma of the CD-1 mice after the different compounds are intravenously injected and orally administered by adopting an LC-MS/MS method.
TABLE 4 pharmacokinetic parameters of the Compounds of the invention
Figure BDA0003866228850000694
The test result shows that the example 16 and the example 37 of the invention have good pharmacokinetic absorption, longer half-life and high bioavailability in CD-1 mice and have good pharmacokinetic properties.
Finally, it should be noted that there are other ways of implementing the invention. Accordingly, the embodiments of the invention are to be considered as illustrative and not restrictive, and modifications within the scope of the invention or equivalents to be added to the claims are possible. All publications or patents cited herein are incorporated by reference.

Claims (25)

1. A compound having the structure shown in formula (I):
Figure FDA0003866228840000011
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof;
wherein the content of the first and second substances,
x is-C (R) c ) -, or N;
Z 1 and Z 2 Each independently is-C (R) 4d ) -or N;
R a 、R b and R c Each independently of the other is H, D, F, cl, C 1-3 Alkyl radical, C 1-6 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl radical, C 1-3 Aminoalkyl radical, C 1-3 Cyanoalkyl, C 1-6 Deuterated alkoxy, C substituted by 1 to 5 deuterons 1-6 Haloalkyl, C substituted by 1 to 5 deuterium 1-6 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-8 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group;
R 1 is H, D, -C 1-6 alkyl-NR e R f 、-C(=O)R 7 、-C(=O)OR 7a 、-C(=O)NR 7 R 7a 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl radical, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
R 2 at each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 2 At each occurrence, is independently optionally substituted with 0, 1, 2, 3, 4, or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group;
R 3 is H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-8 Cycloalkyl radical, C 3-8 Deuterated cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 3 Optionally substituted by 0, 1, 2, 3, 4 or 5R 6 Substitution;
R 4a 、R 4b 、R 4c and R 4d Each independently of the other is H, D, F, cl, -CN, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radicals、C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 4a 、R 4b 、R 4c And R 4d Each independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy radical, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl and C 1-6 Substituted with a group of halogenated deuterated alkyl;
R 5 and R 6 At each occurrence, independently is H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of said-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group;
R e 、R f 、R 7 and R 7a At each occurrence, independently H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 A halogenated alkyl group,C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl radical, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-12 Aryl radical, C 6-12 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group; and
n is 0, 1, 2, or 3;
the conditions are as follows: 1) When R is 1 Is pyrazolyl, wherein R 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substituted, then each R 2 、R 4a 、R 4b And R 4c At least one of which is independently F, cl, -CN, C 1-6 Alkoxy radical, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-6 Haloalkyl, C substituted by 1 to 5 deuterium 1-6 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-8 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group; or 2) when R is 1 Is H, D, -C 1-6 alkyl-NR e R f 、-C(=O)R 7 、-C(=O)OR 7a 、-C(=O)NR 7 R 7a 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 3-8 Cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C other than pyrazolyl 1-9 In the case of heteroaryl, wherein R 1 Optionally substituted by 0,1. 2, 3 or 4R 5 Substituted, then each R a 、R b 、R c 、R 2 、R 3 、R 4a 、R 4b 、R 4c And R 4d At least one of which is independently D (deuterium), C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-6 Haloalkyl, C substituted by 1 to 5 deuterium 1-6 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-8 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group.
2. The compound of claim 1, having a structure according to formula (Ia):
Figure FDA0003866228840000021
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof.
3. The compound of claim 1, wherein R 1 Is phenyl, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, thiazolyl, imidazolyl, oxazolyl, thiadiazolyl,
Figure FDA0003866228840000022
Figure FDA0003866228840000023
Wherein Y is 1 Is O, S, or-NH-;
Y 6 is O, or-NH-;
t3 and t4 are each independently 1, 2, 3 or 4; and
wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
the conditions are as follows: 1) When R is 1 Is composed of
Figure FDA0003866228840000024
In which R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substituted, then each R 2 、R 4a 、R 4b And R 4c Is independently F, cl, -CN, C 1-3 Alkoxy radical, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-3 Haloalkyl, C substituted by 1 to 5 deuterium 1-3 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-6 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group; 2) When R is 1 Is phenyl, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, thiazolyl, imidazolyl, oxazolyl, thiadiazolyl,
Figure FDA0003866228840000031
In which R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substituted, then each R a 、R b 、R c 、R 2 、R 3 、R 4a 、R 4b 、R 4c And R 4d At least one of which is independently D (deuterium), C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C substituted with 1 to 5 deuterium 1-3 Haloalkyl, C substituted by 1 to 5 deuterium 1-3 Hydroxyalkyl, C substituted by 1 to 5 deuterium 3-6 Cycloalkyl, or C substituted by 1 to 5 deuterium 1-9 A heteroaryl group.
4. A compound according to any one of claims 1-3, wherein R 4a 、R 4b 、R 4c And R 4d Each independently of the other is H, D, F, cl, C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, C 1-3 Hydroxyalkyl radical, C 1-3 Aminoalkyl radical, C 1-3 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Deuterated heterocyclic group, C 3-6 Cycloalkyl, C 3-6 Deuterated cycloalkyl, phenyl, or 5-6 atom-containing heteroaryl; it is provided withIn R 4a 、R 4b 、R 4c And R 4d Each independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl and C 1-3 Substituted with a group of halogenated deuterated alkyl.
5. A compound according to any one of claims 1 to 3, wherein R 4a 、R 4b 、R 4c And R 4d Each independently of the others is H, F, cl, D, -CN, -OCH 3 ,-OCH 2 CH 3
Figure FDA0003866228840000032
Figure FDA0003866228840000033
6. A compound having the structure shown in formula (II):
Figure FDA0003866228840000034
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof;
wherein the content of the first and second substances,
x is-C (R) c ) -, or N;
w is C 3-10 Cycloalkyl radical, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein W is optionally substituted by 0, 1, 2, 3 or 4R 4 Substitution;
R a 、R b and R c Each independently is H, D, F, cl, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl, or C 1-6 Cyanoalkyl;
R 1 is-C 1-3 hydroxyalkyl-C (= O) OR 7a ,-C(=O)OR 7a ,-C(=O)NR 7 R 7a
Figure FDA0003866228840000035
Figure FDA0003866228840000041
Wherein Y is 2 And Y 3 Each independently is- (CH) 2 ) t1 -、-(CH 2 ) t1 -L-、-(CH 2 ) t1 -L-(CH 2 ) t2 -, O, or-NH-;
Y 4 is- (CH) 2 ) t1 -、-(CH 2 ) t1 -L-、-(CH 2 ) t1 -L-(CH 2 ) t2 -, -C (= O) -, O, S, or-NH-;
Y 5 is-CH-, or N;
Y 6 Is O, or-NH-;
Y 7 is O, or-NH-;
l is-C (= O) -, O, S, or-NH-;
t1, t2 and t3 are each independently at each occurrence 1, 2, 3 or 4;
t4 is 1; t5 is 1 or 2; and
Figure FDA0003866228840000042
represents a single or double bond; and
wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 Substitution;
R 2 at each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 2 At each occurrence, is independently optionally substituted with 0, 1, 2, 3, 4, or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group;
R 3 is H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 3-8 Cycloalkyl radical, C 3-8 Deuterated cycloalkyl, C 2-9 Heterocyclic group, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 3 Optionally substituted by 0, 1, 2, 3, 4 or 5R 6 Substitution;
R 4 at each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein R is 4 At each occurrence, independently is optionally substituted with 0, 1, 2, 3, 4, or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Substituted with a haloalkyl group;
R 5 and R 6 Each occurrence is independently H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of said-NR e R f 、-C(=O)R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution;
R 5a is-C (= O) R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl radical, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of the-C (= O) R 7 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl radical, C 1-6 Aminoalkyl radical, C 1-6 Cyanoalkyl, C 2-9 Heterocyclic group, C 3-8 Cycloalkyl, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted0. 1, 2, 3, 4 or 5 are independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution;
R e 、R f 、R 7 and R 7a At each occurrence, independently H, D, C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-6 An alkyl group; wherein each C is 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl radical, C 3-8 Cycloalkyl, C 3-8 Cycloalkyl radical C 1-6 Alkyl radical, C 2-9 Heterocyclic group, C 2-9 Heterocyclyl radical C 1-6 Alkyl radical, C 6-12 Aryl radical, C 6-12 Aryl radical C 1-6 Alkyl radical, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-6 Alkyl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-6 Deuterated alkyl, C 1-6 Alkoxy and C 1-6 Haloalkyl group substitution; and
n is 0, 1, 2, or 3.
7. The compound of claim 6, wherein W is
C 3-8 Cycloalkyl radical, C 3-8 A heterocyclic group,
Figure FDA0003866228840000051
Figure FDA0003866228840000052
Wherein Z 1 、Z 2 And Z 3 Each independently is-CH-, or N; and
wherein W is optionally substituted with 0, 1, 2, 3 or 4R 4 And (4) substitution.
8. The compound of claim 6, having a structure represented by formula (IIa):
Figure FDA0003866228840000053
or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof;
wherein m is 0, 1, 2 or 3.
9. A compound according to any one of claims 6-8, wherein R 1 Is that
Figure FDA0003866228840000054
/>
Figure FDA0003866228840000055
Figure FDA0003866228840000056
Wherein R is 1 Optionally substituted by 0, 1, 2, 3 or 4R 5 And (4) substitution.
10. A compound according to any one of claims 6-8, wherein R 4 At each occurrence, independently is H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NH 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, C 1-3 Hydroxyalkyl radical, C 1-3 Aminoalkyl radical, C 1-3 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Cycloalkyl, phenyl,Or a heteroaryl group of 5 to 6 atoms; wherein R is 4 Each occurrence is independently optionally substituted with 0, 1, 2, 3, 4 or 5 independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy and C 1-3 Haloalkyl groups.
11. The compound of any one of claims 6-8, wherein R 5a is-C (= O) R 7 、C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, C 1-4 Alkoxy radical, C 1-4 Haloalkyl, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl radical, C 1-4 Aminoalkyl radical, C 1-4 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Cycloalkyl, C 6-10 Aryl, or C 1-9 A heteroaryl group; wherein each of said-C (= O) R 7 、C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 2-4 Alkenyl radical, C 2-4 Alkynyl, C 1-4 Alkoxy radical, C 1-4 Haloalkyl, C 1-4 Haloalkoxy, C 1-4 Hydroxyalkyl radical, C 1-4 Aminoalkyl radical, C 1-4 Cyanoalkyl, C 3-6 Heterocyclic group, C 3-6 Cycloalkyl, C 6-10 Aryl and C 1-9 Heteroaryl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-6 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Alkoxy and C 1-4 Haloalkyl groups.
12. The compound of any one of claims 1-3 and 6-8, wherein R 3 Is cyclopropyl, pyridyl, or phenyl; wherein R is 3 Optionally substituted by 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Deuterated alkyl and C 1-3 Haloalkyl groups.
13. The compound of any one of claims 1-3 and 6-8, wherein R 5 And R 6 At each occurrence, independently is H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f 、-C(=O)R 7 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl radical, C 3-6 Cycloalkyl, phenyl, or 5-6 atom heteroaryl; wherein each of said-NR e R f 、-C(=O)R 7 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl radical, C 3-6 Cycloalkyl, phenyl and 5-6 atom heteroaryl are independently optionally substituted with 0, 1, 2, 3, 4 or 5 atoms selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-3 Alkyl radical, C 1-3 Deuterated alkyl, C 1-3 Alkoxy and C 1-3 Haloalkyl groups.
14. The compound of any one of claims 1-3 and 6-8, wherein R 5 And R 6 At each occurrence, independently is H, D, oxo (= O), F, cl, br, I, -OH, -CN, -NO 2 、-NR e R f Methyl, ethyl, cyclopropyl, -CD 3 、-OCD 3 、-CF 3 、-CH 2 CH 2 OH、-C(=O)CH 3 、-C(=O)CH 2 CH 3 or-CH 2 CH 2 OCH 3
15. The compound of any one of claims 1-3 and 6-8, wherein R a 、R b And R c Each independently is H, D, F, -CN, -NO 2 、-NR e R f Methyl, ethyl, isopropyl, -CD 3 、-CHD 2 、-CH 2 D. Methoxy, ethoxy, halomethyl, haloethyl, or-CH 2 CH 2 OH。
16. The compound of any one of claims 1-3 and 6-8, wherein R e 、R f 、R 7 And R 7a At each occurrence, independently is H, D, C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl radical, C 3-6 Cycloalkyl, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 3-6 Heterocyclic group, C 3-6 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-9 Heteroaryl, or C 1-9 Heteroaryl C 1-4 An alkyl group; wherein each C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Haloalkyl, C 1-4 Hydroxyalkyl radical, C 3-6 Cycloalkyl radical, C 3-6 Cycloalkyl radical C 1-4 Alkyl radical, C 3-6 Heterocyclic group, C 3-6 Heterocyclyl radical C 1-4 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-4 Alkyl radical, C 1-9 Heteroaryl and C 1-9 Heteroaryl C 1-4 Alkyl is independently optionally substituted with 0, 1, 2, 3, 4 or 5 substituents independently selected from H, D, oxo (= O), F, cl, br, I, -OH, -NH 2 、-CN、-NO 2 、C 1-4 Alkyl radical, C 1-4 Deuterated alkyl, C 1-4 Alkoxy and C 1-4 Haloalkyl groups.
17. The compound of claim 1, which is a compound having one of the following structures:
Figure FDA0003866228840000061
/>
Figure FDA0003866228840000071
/>
Figure FDA0003866228840000081
Or a stereoisomer, tautomer, deuteron, nitroxide, solvate, or pharmaceutically acceptable salt thereof.
18. The compound of claim 6, which is a compound having one of the following structures:
Figure FDA0003866228840000082
/>
Figure FDA0003866228840000091
Figure FDA0003866228840000101
or a stereoisomer, tautomer, nitroxide, solvate, or pharmaceutically acceptable salt thereof.
19. A pharmaceutical composition comprising a compound of any one of claims 1-18, or a stereoisomer, a tautomer, a nitric oxide, a solvate, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable adjuvant, diluent, or carrier, or a combination thereof.
20. The pharmaceutical composition of claim 19, further comprising an additional therapeutic agent.
21. Use of a compound according to any one of claims 1-18 or a pharmaceutical composition according to any one of claims 19-20 in the manufacture of a medicament for preventing, treating, and/or alleviating a PI 3-kinase mediated disease, disorder, and/or condition, or inhibiting PI 3-kinase activity.
22. The use of claim 21, wherein the PI 3-kinase mediated disease, disorder, and/or condition is selected from respiratory diseases, viral infections, non-viral respiratory infections, allergic diseases, autoimmune diseases, inflammatory diseases, cardiovascular diseases, hematologic malignancies, neurodegenerative diseases, pancreatitis, multi-organ failure, renal diseases, platelet aggregation, cancer, sperm motility, transplant rejection, graft rejection, lung injury or pain.
23. The use of claim 21, wherein the PI 3-kinase mediated disease, disorder, and/or condition is selected from asthma, chronic Obstructive Pulmonary Disease (COPD), viral respiratory infections, viral respiratory disease exacerbations, aspergillosis, leishmaniasis, allergic rhinitis, allergic dermatitis, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, thrombosis, atherosclerosis, hematological malignancies, neurodegenerative diseases, pancreatitis, multi-organ failure, kidney disease, platelet aggregation, cancer, sperm motility, transplant rejection, graft rejection, lung injury, pain associated with rheumatoid arthritis or osteoarthritis, back pain, systemic inflammatory pain, posthepatic neuralgia, diabetic neuropathy, inflammatory neuropathic pain (trauma), trigeminal neuralgia or central pain.
24. The use of claim 22 or 23, wherein the cancer is selected from acute myelogenous leukemia, myelodysplastic syndrome, myeloproliferative disorders, chronic myelogenous leukemia, T-cell acute lymphocytic leukemia, B-cell acute lymphocytic leukemia, non-hodgkin's lymphoma, B-cell lymphoma, solid tumors, or breast cancer.
25. The use of claim 21, wherein the PI 3-kinase is PI3K- γ.
CN202211183447.8A 2021-09-28 2022-09-27 Isoquinolone and quinazolinone compounds, and composition and application thereof Pending CN115873000A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021111453594 2021-09-28
CN202111145359 2021-09-28

Publications (1)

Publication Number Publication Date
CN115873000A true CN115873000A (en) 2023-03-31

Family

ID=85770085

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211183447.8A Pending CN115873000A (en) 2021-09-28 2022-09-27 Isoquinolone and quinazolinone compounds, and composition and application thereof

Country Status (2)

Country Link
CN (1) CN115873000A (en)
WO (1) WO2023051495A1 (en)

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20160685A1 (en) * 2013-10-04 2016-07-23 Infinity Pharmaceuticals Inc HETEROCYCLIC COMPOUNDS AND USES OF THEM
US9708348B2 (en) * 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
WO2017161116A1 (en) * 2016-03-17 2017-09-21 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as pi3k kinase inhibitors
WO2017214269A1 (en) * 2016-06-08 2017-12-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2019228404A1 (en) * 2018-05-31 2019-12-05 信达生物制药(苏州)有限公司 Novel phosphoinositide 3-kinase inhibitor and preparation method and use thereof
CN112194659A (en) * 2019-07-08 2021-01-08 浙江海正药业股份有限公司 Alkyne derivative and preparation method and application thereof

Also Published As

Publication number Publication date
WO2023051495A1 (en) 2023-04-06

Similar Documents

Publication Publication Date Title
US10000507B2 (en) Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9896449B2 (en) Ring-fused bicyclic pyridyl derivatives as FGFR4 inhibitors
CN109348715B (en) Pyrrolotriazine compounds as TAM inhibitors
JP6708661B2 (en) 1-Cyano-pyrrolidine compounds as USP30 inhibitors
JP6615752B2 (en) Substituted nicotinimide inhibitors of BTK and their preparation and use in the treatment of cancer, inflammation and autoimmune diseases
TWI623530B (en) Bicyclic-fused heteroaryl or aryl compounds
CN105732636B (en) Heteroaromatic compounds and their use in medicine
CN107011348B (en) Heteroaryl pyridone and aza-pyridone compounds as inhibitors of BTK activity
JP2019081786A (en) N-pyrrolidinyl, n&#39;-pyrazolyl urea, thiourea, guanidine, and cyanoguanidine compound as trka kinase inhibitor
CN113272303A (en) Pyrimido five-membered nitrogen heterocyclic derivative, preparation method and application thereof in medicine
CN104884458A (en) Fused heterocyclic compounds as protein kinase inhibitors
TW201404779A (en) Novel heteroaryl and heterocycle compounds, compositions and methods
TW201512173A (en) Alkynyl alcohols and methods of use
KR20180051624A (en) Pyrazolyl-substituted heteroaryls and their use as medicaments
TWI785474B (en) Novel heterocyclic compounds useful as selective aurora a inhibitors
CN113061132A (en) Condensed ring lactam compound, preparation method and application
CN114340634A (en) Heterocyclic compounds as kinase inhibitors
CN114667287A (en) Adenosine A2A receptor antagonists and uses thereof
KR20150130311A (en) N-(2-cyano heterocyclyl)pyrazolo pyridones as janus kinase inhibitors
CN114075221A (en) Substituted heteroaryl compounds, compositions and uses thereof
JP2023506436A (en) Pyrazole-Containing Polycyclic Derivative Inhibitors, Methods of Preparation Therefor, and Uses Therefor
US20230054411A1 (en) 7-, 8-, and 10-SUBSTITUTED AMINO TRIAZOLO QUINAZOLINE DERIVATIVES AS ADENOSINE RECEPTOR ANTAGONISTS, PHARMACEUTICAL COMPOSITIONS AND THEIR USE
CN112279837B (en) Pyrazine compounds and uses thereof
CN115873000A (en) Isoquinolone and quinazolinone compounds, and composition and application thereof
CN114630822A (en) Biaryl dihydroorotate dehydrogenase inhibitors

Legal Events

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