CN116262752A - Pyrazolopyrimidine compound and application thereof - Google Patents

Pyrazolopyrimidine compound and application thereof Download PDF

Info

Publication number
CN116262752A
CN116262752A CN202111536751.1A CN202111536751A CN116262752A CN 116262752 A CN116262752 A CN 116262752A CN 202111536751 A CN202111536751 A CN 202111536751A CN 116262752 A CN116262752 A CN 116262752A
Authority
CN
China
Prior art keywords
alkyl
group
ring
heteroaryl
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
CN202111536751.1A
Other languages
Chinese (zh)
Inventor
叶斌
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Yidi Biotechnology Co ltd
Original Assignee
Shanghai Yidi Biotechnology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Yidi Biotechnology Co ltd filed Critical Shanghai Yidi Biotechnology Co ltd
Priority to CN202111536751.1A priority Critical patent/CN116262752A/en
Priority to PCT/CN2022/139044 priority patent/WO2023109870A1/en
Publication of CN116262752A publication Critical patent/CN116262752A/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
    • A61K31/495Heterocyclic 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
    • A61K31/495Heterocyclic 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
    • A61K31/52Purines, e.g. adenine
    • 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
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • 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

Abstract

The invention belongs to the field of medicine synthesis, and relates to pyrazolopyrimidine compound shown as a formula (I) and/or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the compound and/or the pharmaceutically acceptable salt thereof, a method for preparing the compound, and application of the compound in preparing medicines for treating diseases mediated by PI 3K.

Description

Pyrazolopyrimidine compound and application thereof
Technical Field
The invention belongs to the field of medicine synthesis, and relates to pyrazolopyrimidine compound shown as a formula (I) and/or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the compound and/or the pharmaceutically acceptable salt thereof, a method for preparing the compound, and application of the compound in preparing medicines for treating diseases mediated by PI 3K.
Figure BDA0003412931580000011
Background
Inositol Phosphates (PI) play a key role as second messengers in a variety of cellular functions including cell survival, signal transduction, and regulation of membrane transport and transport (Di Paolo, g.et al Nature,2006,443,651-657;Schaeffer,E.M.et al Curr.Opin.Immunol, 2000,12,282-288). Various phosphoinositides are involved in various diseases including cancer, autoimmune diseases, inflammation, and the like. Phosphoinositides are lipids comprising two layers of fatty acids, the fatty acids of which can be linked to cytosolic 1-inositol by glycerophosphate linkages. Lipid kinase PI3K converts PI phosphatidylinositol 4, 5-biphosphate (PIP 2) to phosphatidylinositol 3,4, 5-triphosphate (PIP 3) by ATP phosphorylating the 3-OH of the inositol ring moiety (Parker, p.j. Biochem. Soc. Trans.,2004,32,893-898). PIP3 acts as an anchor site for protein kinases containing highly conserved Pleckstrin Homology (PH) domains (Hawkins, p.t.et al biochem. Soc. Trans.,2006,34,647-662) and binds to intracellular PH domain-containing signaling proteins such as serine/threonine kinase Akt and PDK1 (3-phosphoinositide-dependent protein kinase), promoting phosphorylation of Akt protein by PDK1, leading to Akt activation. These kinases, when activated (phosphorylated or dephosphorylated), can regulate a number of important cellular functions, such as most of the growth, survival and proliferation functions of cells, which can be controlled by modulating downstream proteins such as mTOR, gsk3β, foxo3a, p70S6K and NF- κb. Furthermore, calcium mobilization and gene transcription are also controlled by the Tec family of tyrosine kinases BTK, ETK and ITK of the PH domain (Fruman, d.a. et al immunol. Rev.2009,228, 253-272.). Thus, phosphoinositide metabolizing enzymes such as PI3K have become attractive targets for modulating these second messenger functions.
The most studied class I in PI3K is that which can be divided into two subclasses (class 1A and class 1B). Of these, class 1A (pi3kα, pi3kβ, and pi3kδ) is downstream of Receptor Tyrosine Kinase (RTK), while class 1B (pi3kγ) is downstream of G Protein Coupled Receptors (GPCRs). These four homologous kinases exist as heterodimers of the regulatory and catalytic subunits. The regulatory subunits contain domains that allow anchoring to cell surface receptors and other regulatory proteins. The catalytic subunits (p110α, p110β, p110γ, p110δ) contain ATP binding domains. Catalytic subunits, particularly their ATP binding sites, are currently the focus of PI3K small molecule inhibitor research. Small molecule compounds block the phosphorylation of PIP2 and block the formation of PIP3 by competitive inhibition of the ATP binding site. Important regulatory proteins such as AKT will therefore not be anchored to the cell membrane and their function will be inhibited.
Pi3kα and pi3kβ are ubiquitously expressed in vivo, pi3kγ and pi3kδ are mainly present in leukocytes, and pi3kδ is almost limited to spleen, thymus and peripheral blood leukocytes (Eicholt, b.j.et al PLoS One,2007,9, e 869). Deregulation of pi3kα and pi3kβ is associated with the occurrence and progression of solid tumor diseases; whereas dysregulation of pi3kγ and pi3kδ is associated with inflammatory and immune system diseases such as Rheumatoid Arthritis (RA), systemic Lupus Erythematosus (SLE), pulmonary diseases such as chronic obstructive pulmonary disease, COPD, and asthma, hematological malignancies. The data obtained from transgenic mice and two successful B cell targeted biologicals demonstrate a strict expression pattern of pi3kδ, driving researchers in the field to develop new pi3kδ small molecule inhibitors. These biologicals, such as rituximab and belimumab, have been demonstrated to be effective against RA and SLE, respectively. Since pi3kδ has a unique role in B cell maturation and function (Vallerskog, t.et al clin.immunol.,2007,122,62-74), inhibition of PI3K should have a role similar to both biologicals. In addition to the regulatory effects of B cells, PI3K delta plays a part in T cell activation (Soond, R.et al; blood,2010,115,2203-3313) and neutrophil trafficking (Ali, K.et al Nature,2004,431,1007-1011), and also helps to activate other leukocytes associated with autoimmune diseases such as macrophages, dendritic cells and NK T cells (Fung-Leung, W.P.cell.signaling 2011,23,603-608). The role of pi3kδ in leukocyte function is supported from the last two documents. These two documents describe several major functionally acquired pi3kδ mutations from several different families in europe. PI3K delta mutants manifest as lymphoproliferative and immunodeficiency, which predisposes them to lung and ear infections (Angulo, i.et al science2013,342, 866-871). The mechanism of immunodeficiency associated with this mutation is not well understood, and it is speculated in the literature that the overactive p110δ -AKT-mTOR pathway promotes aerobic glycolysis, limiting the function and survival of lymphocytes and contributing to their premature aging, compromising the appropriate immune response to pathogens (Lucas, c.l.et al, nat.immunol.2014,15,88-97). Chronic inflammation is not unique to autoimmune diseases, but is also associated with prolonged exposure to stress or damage to the body. These all have the potential to lead to sustained signaling in response to the PI3K delta pathway in leukocytes. For example, in Chronic Obstructive Pulmonary Disease (COPD), sustained contact with cigarette smoke is associated with high levels of PI3K delta and phosphorylated AKT in the lung (To, y.et al, am.j. Repir.crit. Care med.2010,182, 897-904). As with functionally acquired PI3K delta mutant individuals, COPD patients are more susceptible to recurrent bacterial respiratory infections, including haemophilus influenzae and streptococcus pneumoniae. Like patients with functional gain-of-function mutations, similar immunodeficiency in such patients may be associated with continuously activated PI3K delta signaling in COPD. Leukocyte deficiency in functionally acquired mutant patients can be ameliorated by rapamycin treatment. Rapamycin is an inhibitor of the PI3K delta downstream protein mTOR. Thus, inhibition of pi3kδ is also a potential therapeutic option for PI3kδ signaling pathway-related diseases, presumably not only for APDS (activated PI3K syndrome), complex autoimmune diseases such as SLE, RA, etc., but also for pathological diseases caused by PI3kδ -driven B-cell and other leukocyte subpopulations (Palanki, m.et al, j.med.chem.2007,50,4279-4294;Pei,Z.et al,ACS Med.Chem.Lett.2013,4,103-107;Cushing,T.D.et al,J.Med.Chem.2012,55,8559-8581;Shukla,MR.et al J.Med.Chem.2020,63,14700-14723, method, j.l., et al, j.med.chem.2021,64, 5137-5156), as well as for chronic non-autoimmune diseases such as respiratory disease COPD.
Oral pi3kδ inhibitors, such as Idelalisib, duvelisib and recently available batches of ubralinib, have been used to treat B cell lymphomas, but have limited clinical use due to dose-limiting toxicity, for use in two-line and later treatment of patients. Compared with the other two medicines on the market, the Chinese medicament has relatively good safety, and has relatively small proctitis and immune hepatotoxicity. One of the reasons for this is based on the summary of the latest Nature journal, which is that the Drug has the highest selectivity for PI3K subtypes other than pi3kδ (Nat Rev Drug discovery.2021 oct;20 (10): 741-769.).
In addition, the use of PI3K delta inhibitors in the cancer field is currently limited to hematological cancers, which have limited research and study results in solid tumors. Whereas studies have shown that PI3K delta can also be used in combination with anti-PD 1 antibodies for thermal tumors (J Mol Med (Berl). 2016Jan;94 (1): 5-11.). Therefore, although more than 5 PI3K delta inhibitors are currently marketed or developed in advanced clinical stages, they are all studied for hematological cancer and have not yet been marketed in China, and thus, there is still a need for a PI3K delta inhibitor with higher selectivity and lower toxicity to be applied in clinic, especially, to be expanded for first-line lymphoma or to be expanded for solid tumor, and to be applied in non-cancer fields such as inflammatory diseases.
Notably, none of the drugs currently marketed are approved for use in inflammatory diseases. Apart from the two small studies, duvelisib did not study inflammatory disease further. Inhaled administration has been explored to target pulmonary treatment, as systemic administration may lead to intolerable toxicity. In the prior art, patent documents WO2005113556, US20130071212, US20140179718, WO2016045591, CN108017641 and US2021046075 disclose a series of compounds of selective PI3K delta inhibitors and medical application thereof in the fields of PI3K delta gene function acquired mutation, autoimmune diseases, chronic inflammatory diseases, cancers and the like.
Considering that there are mainly two types of PI3K delta inhibitor structures, flat type and propeller type with chiral groups (propeller-type). The flat structure has a plurality of aromatic groups which are generally conjugated with each other, the structural planarity is high, and the solubility and the selectivity are poor.
Based on solving the defects and shortcomings in the prior art, the inventor and team of the application expect to select a propeller type framework, and design and synthesize a new compound with pyrazolopyrimidine as a parent nucleus so as to obtain a PI3K delta inhibitor with high solubility and better selectivity.
Although the development of compounds into salts is an effective method for improving solubility, the compounds of the present invention do not form salts with all organic or inorganic acids, and the salification is unpredictable, and the solubility, stability, crystallinity, and other physicochemical properties of different salts are more unpredictable.
Disclosure of Invention
In a first aspect of the present invention there is provided a compound of formula (I) and/or a pharmaceutically acceptable salt thereof,
Figure BDA0003412931580000041
wherein:
R 1 independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, mercapto, amino, -C 1-6 Alkyl, -C 1-6 Alkoxy, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocyclylalkyl, -C 3-6 Cycloalkoxy, -C 3-6 Heterocyclyloxy, -C 1-6 Alkyl (O) C 1-6 Alkyl, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR 1a Aryl, aralkyl, aralkoxy, heteroaryl, heteroaralkyl, heteroaralkoxy; wherein said mercapto, amino, alkyl, alkoxy, alkenyl, alkyneRadicals, cycloalkyl, cycloalkoxy, heterocycloalkyl, heterocycloalkoxy, aryl, aralkyl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Optionally substituted by one or more R 1a And (3) substitution.
R 1a Selected from hydrogen, deuterium, halogen, cyano, hydroxy, mercapto, amino, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkoxy, heterocycloalkyl, heterocycloalkoxy, aryl, heteroaryl, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, heteroaryl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Selectively by halogen, cyano, -OR a 、-SR a 、NR b R c Any one or more of alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocyclyloxy, substituted or unsubstituted;
R 2 selected from deuterium, hydroxy, -C 1-6 Alkyl, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocyclylalkyl, -C 3-6 Cycloalkoxy, -C 3-6 Heterocyclyloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) NH (C) 1-6 Alkyl group 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 、-C(O)OR 2a 、-C(O)NH 2 Aryl, heteroaryl; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, heteroaryl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Can be optionally substituted with one or more R 2a Substitution;
R 2a selected from hydrogen, deuterium, halogen, cyano, hydroxy, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroAryl, heteroaryloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Is optionally selected from halogen, cyano, -OR a 、-SR a 、NR b R c One or more of alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocyclyloxy, substituted or unsubstituted;
and/or R 1 And R is R 2 Optionally forming 3-6 membered cycloalkyl, 3-6 membered heterocycloalkyl, 3-6 membered cycloalkenyl, 3-6 membered heterocycloalkenyl, aryl, heteroaryl with the carbon atom to which each is attached, and optionally substituted with one or more R 1a Or R is 2a Substitution; and the carbon atom or sulfur atom in the cycloalkyl group, heterocycloalkyl group, cycloalkenyl group, heterocycloalkenyl group may be selectively oxo-to form a part of a cyclic carbonyl group, an amide group, an ester group, a sulfone group (-S (O) 2-), a sulfonamide group (-S (O) 2 NH-).
R 3 Selected from hydrogen, deuterium, C 1-6 Alkyl, deuterated alkyl, halogenated C 1-6 Alkyl, hydroxy substituted C 1-6 Alkyl, cyanoalkyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocycloalkyl, alkoxyalkyl, cycloalkylalkyl, heterocyclylalkyl, cycloalkoxyalkyl, heterocycloalkoxyalkyl;
R 4 independently selected from heteroaryl groups, said heteroaryl groups being monocyclic aromatic hydrocarbon groups having 5, 6 or 7 ring atoms which contain 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or bicyclic aromatic hydrocarbon groups having 9 or 10 ring atoms which contain 1, 2, 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring, and said heteroaryl groups include those heteroaryl groups in which the N ring heteroatoms are in the form of N-oxides, and said heteroaryl groups are optionally substituted with One or more substituents selected from the group consisting of: halogen, deuterium, cyano, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, cycloalkoxy, heterocyclyl, heterocyclyloxy, -NH 2 、-NH(C 1-6 Alkyl), -N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -COOH, -C (O) NH 2 -C (O) NH (C1-6 alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a 、-NHC(O)(C 1-6 Alkyl), -NHC (O) (C) 1-6 Cycloalkyl), -NHC (O) O (C) 1-6 Alkyl), -NHC (O) O (C) 1-6 Cycloalkyl), phenyl, and 5-or 6-membered heteroaryl; wherein C is 1-6 Alkyl, haloalkyl, -NH 2 、-COOH、-C(O)NH 2 Cycloalkyl, heterocycloalkyl, optionally substituted with one or more R 1a Optionally substituted; wherein R is as R 4 The 5 or 6 membered heteroaryl group of the substituents of (a) is a monocyclic aromatic hydrocarbon group having 5 or 6 ring atoms which contains 1, 2 or 3 ring heteroatoms independently selected from N, O or S in the ring, the remaining ring atoms being carbon atoms, the S and O heteroatoms being non-adjacent to each other when the total number of S and O atoms in the heteroaryl group exceeds 1, wherein the phenyl or 5 or 6 membered heteroaryl group as substituents of the heteroaryl group is optionally substituted with one or more substituents selected from: halogen, amino, hydroxy, mercapto, C1-6 alkyl and-O (C1-6 alkyl), haloalkyl;
R 5 independently selected from C 1-6 Alkyl, hydroxy-substituted alkyl, deuterated alkyl, hydroxyalkyl, C 3-6 Cycloalkyl, C 3-6 Heterocycloalkyl, cycloalkylalkyl or cycloalkoxyalkyl, wherein said alkyl is optionally substituted with one or more R 5a Substitution; r is R 5a Selected from deuterium, halogen, CN, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl; wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl groups may be substituted with halogen, CN, -OR a 、-SR a 、-NR b R c Alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl substitution;
and/or R 4 And R is 5 Selectivity ofAnd the N atom to which the two are attached together form a nitrogen-containing heterocyclic group or heteroaryl group, wherein the nitrogen-containing heterocyclic group and heteroaryl group may be optionally substituted with one or more substituents including, but not limited to, halogen, deuterium, amino, hydroxy, mercapto, cyano, alkyl, deuteroalkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a 、-OR a 、-SR a 、NR b R c
Wherein R is a 、R b And R is c Independently selected from hydrogen, deuterium, halogen, C 1-6 Alkyl, deuterated C 1-6 Alkyl, halogenated C 1-6 Alkyl, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocycloalkyl, hydroxy-substituted C 1-6 Alkyl, -C 1-6 Alkyl (O) C 1-6 Alkyl, aryl, heteroaryl;
and/or R b And R is c Together with the nitrogen atom to which they are both attached form a 4-to 7-membered heterocycloalkyl or are substituted with 1 to 3R a And (3) substitution.
Further, the aryl group is a carbocyclic hydrocarbon group containing 6 to 12 ring carbon atoms consisting of one ring or a plurality of rings condensed, wherein at least one ring is an aromatic ring and the other rings are not heteroaryl groups;
the heteroaryl is a monocyclic aromatic hydrocarbon group having 5, 6 or 7 ring atoms which contains 1 or 2 ring heteroatoms independently selected from N, O and S in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 or 10 ring atoms which contains 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring, when the total number of S and O atoms on the heterocycle exceeds 1, the S and O atoms are not adjacent to each other, and the heteroaryl includes those heteroaryl groups in which the N heteroatom is in the form of an N-oxide;
the aryl and heteroaryl groups are optionally substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, hydroxy, mercapto, -C 1-6 Alkyl, haloalkyl, alkoxy, haloalkoxy, haloalkoxyalkyl, cycloalkyl, halocycloalkyl, - (C) 1-6 Alkyl) OH and-SO 2 (C 1-6 Alkyl), alkenyl, alkynyl.
Further, the aryl or heteroaryl is selected from aryl and heteroaryl groups having 6 ring atoms, heteroaryl groups having 5 ring atoms, bicyclic heteroaryl groups having 9 or 10 ring atoms, or aryl groups;
wherein the aryl and/or heteroaryl group comprises phenyl, deuterated phenyl, naphthyl, pyridinyl, pyrimidine, pyridazine, piperazine, pyrazolyl, imidazolyl, quinolinyl, thienyl, benzothienyl, indolyl, optionally substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, hydroxy, -C 1-6 Alkyl, - (C) 1-6 Alkyl) OH, C 1-6 Haloalkyl and-SO 2 (C) 1-6 Alkyl).
Further, R4 is selected from heteroaryl, or R4 and R5 form heteroaryl;
wherein the heteroaryl group is a monocyclic aromatic hydrocarbon group having 6 ring atoms which contains 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 ring atoms which contains 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring and the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of: halogen, cyano, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl) -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-C(O)NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), phenyl, and 5-or 6-membered heteroaryl.
Further, R 4 Selected from heteroaryl, or R 4 And R is R 5 Forming a heteroaryl group;
wherein the heteroaryl group is a monocyclic aromatic hydrocarbon group having 6 ring atoms which contains 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 ring atoms which contains 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring and the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, hydroxy, mercapto, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-C(O)NH 2 、-C(O)NH(C 1-6 Alkyl), phenyl, pyridyl, pyrimidinyl, piperazinyl, pyridazinyl, pyrazolyl, imidazolyl, triazolyl, and tetrazolyl.
Further, the R 4 Optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, mercapto, amino, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl).
Further, the aryl group in the heteroaryl group is selected from pyrimidine, purinyl, pyrazolopyrimidine, pyrrolopyrimidine.
Further, R 4 Or R is 4 And R is R 5 The heteroaryl group formed is selected from the substituents shown below
Figure BDA0003412931580000071
Wherein the substituents are optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, hydroxy, mercapto, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-CONH 2 、-C(O)NH(C 1-6 Alkyl), phenyl, pyridyl, pyrazolylImidazolyl, oxazolyl, triazolyl and tetrazolyl, wherein R is 4 Phenyl, pyridinyl, pyrimidinyl, piperazinyl, pyridazinyl, pyrazolyl, imidazolyl, triazolyl and tetrazolyl of the substituents are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl).
Further, R4 is selected from
Figure BDA0003412931580000072
Optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), -CO2H, -CONH 2 Phenyl, pyridyl, pyrazolyl, triazolyl and tetrazolyl,
wherein R is as R 4 Phenyl, pyridinyl, pyrazolyl, imidazolyl, triazolyl and tetrazolyl of the substituents are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, C 1-6 Alkyl and-O (C) 1-6 Alkyl).
Further, R 3 Is C 1-4 Alkyl and C 1-4 A haloalkyl group.
Further, R 3 Methyl, ethyl and trifluoromethyl.
Further, the compound of formula (I) is selected from:
Figure BDA0003412931580000081
in a second aspect of the invention, the use of a compound of formula (I) as defined above, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease associated with PI 3K.
Further, diseases mediated by PI3K include diseases responsive to inhibition of PI3K, including auto-inflammatory diseases, respiratory diseases, infectious diseases, autoimmune diseases, and cancers.
Further, the use in a medicament for the treatment of a disease mediated by PI3K, which PI3K is pi3kδ.
Further, the disease is selected from the group consisting of cancer, inflammation, respiratory disease, infectious disease, bone marrow related disease, and autoimmune disease.
Further, cancers are solid tumors, leukemias, lymphomas, and multiple myelomas. Leukemia can be selected from Acute Lymphoblastic Leukemia (ALL), acute Myelogenous Leukemia (AML), chronic Lymphoblastic Leukemia (CLL) and Chronic Myelogenous Leukemia (CML); the lymphoma is selected from hodgkin's lymphoma, non-hodgkin's lymphoma (NHL), mantle Cell Lymphoma (MCL), follicular lymphoma, B-cell lymphoma, and diffuse large B-cell lymphoma; the autoimmune, inflammatory, respiratory, bone marrow related and infectious diseases may be selected from skin inflammation, rheumatoid arthritis, allergic rhinitis, asthma, crohn's disease, chronic Obstructive Pulmonary Disease (COPD), systemic lupus erythematosus, psoriasis, multiple sclerosis, activated PI3K syndrome, sjogren's syndrome, myelofibrosis and/or leishmaniasis.
Further, the cancer is a solid tumor including breast cancer, melanoma, head and neck cancer, lung cancer, thymus cancer, and hematological malignancy.
In a third aspect of the present invention there is provided a pharmaceutical composition comprising a compound of formula (I) as defined above, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient.
Further, the pharmaceutical compositions also include use in combination with small molecule compounds and/or large molecule antibodies to treat cancer, inflammation, bone marrow related diseases, and autoimmune diseases.
Further, the small molecule compounds include BTK inhibitors, bcl2 inhibitors.
Further, the macromolecular antibodies include anti-CD 20, anti-CTLA 4, anti-PD-1, anti-PD-L1 antibodies.
In a fourth aspect of the invention, there is provided the use of any of the above pharmaceutical compositions in the treatment of a disease mediated by PI3K, wherein the disease is selected from cancer, inflammation, respiratory disease, infectious disease, bone marrow related disease and autoimmune disease.
In a fifth aspect the invention provides a solvate, racemic mixture, enantiomer, diastereomer, tautomer, or mixture comprising any proportion of a racemic mixture of a compound of any one of formula (I) above, wherein R 1 、R 2 、R 3 、R 4 And R is 5 Each substituent is as defined above.
Figure BDA0003412931580000091
In a sixth aspect of the invention there is provided a prodrug of a compound of any one of the formulae (I) above.
In a seventh aspect of the invention there is provided a method of inhibiting PI3K activity in vivo or in vitro by a compound of any one of the above formulas (I), comprising contacting an effective dose of a compound of any one of the above formulas (I) and/or a pharmaceutically acceptable salt thereof with PI3K kinase.
Further, the PI3K kinase is any one or more of pi3kα, pi3kβ, pi3kγ, pi3kδ. Further, the PI3K kinase is pi3kδ.
In an eighth aspect of the invention there is provided a combination therapy for the treatment of cancer, inflammation and autoimmune diseases comprising at least a compound of any one of formula (I) or a pharmaceutically acceptable salt thereof, as described above, and a small molecule compound or a large molecule antibody.
The compounds of the invention may be asymmetric, for example having one or more stereocenters. Unless otherwise defined, all stereoisomers, such as enantiomers and diastereomers. Containing asymmetrically substituted carbon atoms. The compounds of the invention may be isolated in optically pure or racemic forms. Optically pure forms can be prepared by resolution of racemates or by the use of chiral synthons or chiral reagents.
The compounds of the invention may also include tautomeric forms. The new form of the tautomer results from the exchange of single bonds and adjacent double bonds together with the migration of protons.
The compounds of the present invention may also include all isotopic forms of the atoms present in the intermediate or final compound. Isotopes include those atoms having the same atomic number but different mass numbers. Isotopes of hydrogen include deuterium and tritium, for example.
The invention also includes pharmaceutically acceptable salts of the compounds of formula (I). Pharmaceutically acceptable salts refer to derivatives of the compounds of formula (I) wherein the parent compound is modified by conversion of the existing base moiety to its salt form, or wherein the parent compound is modified by conversion of the existing acid moiety to its salt form.
Specifically, examples of pharmaceutically acceptable salts include, but are not limited to: salts of inorganic or organic acids of basic groups (e.g., amines) or salts of inorganic or organic bases of acidic groups (e.g., carboxylic acids). Pharmaceutically acceptable salts of the invention can be synthesized from the parent compound of formula (I) by reacting the free base form of these compounds with 1 to 4 equivalents of the appropriate acid in a solvent system. Suitable salts are listed in Remington's PharmaceuticalSciences,17th ed., mack Publishing Company, easton, pa.,1985, p.1418 and Journal of Pharmaceutical Science,66,2 (1977).
The compounds of the present invention, and pharmaceutically acceptable salts thereof, also include solvate forms or hydrate forms. In general, solvate forms or hydrated forms are equivalent to non-solvate forms or non-hydrated forms and are included within the scope of the present invention. Some compounds of the invention may exist in a variety of crystalline or amorphous forms. In general, all physical forms of the compounds are included within the scope of the present invention.
The invention also includes prodrugs of the compounds of formula (I). A prodrug is a pharmacological substance (i.e., drug) derived from a parent drug that, once administered, is metabolized in the body to the parent drug. Prodrugs can be prepared by substituting one or more functional groups present in the compound, wherein the substituents of the prodrug are removed in vivo in such a way as to be converted to the parent compound. The preparation and use of prodrugs is described, for example, in T.Higuchi and V.stella, "Pro-drugs asNovel Delivery Systems" Vol.14of the A.C.S. symposium Series and Bioreversible.
Detailed Description
The invention is further illustrated by the following examples, which are only intended to more particularly illustrate preferred embodiments of the invention and are not intended to limit the technical solutions of the invention. The above-mentioned solutions of the present invention are all technical solutions for achieving the objects of the present invention. The temperatures and reagents employed in the examples below may be replaced with the corresponding temperatures and reagents described above to achieve the objects of the present invention.
Definition of the definition
The following terms, phrases and symbols used in this application have the meanings as described below, unless the context indicates otherwise.
The short dash ("-") between two letters or symbols represents the attachment site for a substituent. For example, -O (C) 1-4 Alkyl) means C linked to the remainder of the molecule through an oxygen atom 1-4 An alkyl group. However, "-" may be omitted when the attachment site for the substituent is apparent to those skilled in the art, for example, a halogen substituent.
Unless explicitly stated otherwise, the use of terms such as "a" or "1" refer to one or more.
The term "alkyl" as used herein refers to a straight or branched chain saturated hydrocarbon group containing from 1 to 18 carbon atoms, for example from 1 to 12 carbon atoms, for example from 1 to 6 carbon atoms, for example from 1 to 4 carbon atoms. For example, "C1-6 alkyl" is within the scope of "alkyl" and refers to alkyl groups having 1 to 6 carbon atoms as described. Examples of alkyl groups include, but are not limited to, methyl ("Me"), ethyl ("Et"), n-propyl ("n-Pr"), isopropyl ("i-Pr"), n-butyl ("n-Bu"), isobutyl ("i-Bu"), sec-butyl ("sBu"), and tert-butyl ("t-Bu").
The term "alkenyl" as used herein refers to a straight or branched chain hydrocarbon radical containing one or more, for example 1, 2 or 3, carbon-carbon double bonds (c=c), containing 2 to 10 carbon atoms, for example 2 to 6 carbon atoms, further for example 2 to 4 carbon atoms. For example, "C2-6 alkenyl" is within the scope of "alkenyl" and means that the alkenyl group has 2 to 6 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl, 2-propenyl, and 2-butenyl.
The term "alkynyl" as used herein refers to a straight or branched chain hydrocarbon radical containing 2 to 10 carbon atoms, for example 2 to 6 carbon atoms, and still more for example 2 to 4 carbon atoms, containing one or more, for example 1, 2 or 3 carbon-carbon triple bonds (c≡c). For example, "C 2-6 Alkynyl "means the alkynyl group described comprising 1 carbon-carbon triple bond (c≡c) and having 2 to 6 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl, 2-propynyl, and 2-butynyl.
The term "halo" as used herein refers to fluoro, chloro, bromo and iodo, and "halogen" refers to fluoro, chloro, bromo and iodo.
The term "haloalkyl" as used herein refers to an alkyl group as defined herein wherein one or more hydrogen atoms, for example 1, 2, 3, 4 or 5 hydrogen atoms, are replaced by halogen atoms, and when more than one hydrogen atom is replaced by a halogen atom, the halogen atoms may be the same or different from each other. In one embodiment, the term "haloalkyl" as used herein refers to an alkyl group as defined herein wherein two or more hydrogen atoms, for example 2, 3, 4 or 5 hydrogen atoms, are replaced by halogen atoms, wherein the halogen atoms are identical to each other. In another embodiment, the term "haloalkyl" as used herein refers to an alkyl group as defined herein wherein two or more hydrogen atoms, for example 2, 3, 4 or 5 hydrogen atoms, are replaced by halogen atoms, wherein the halogen atoms are different from each other. Examples of haloalkyl groups include, but are not limited to, -CF3, -CHF2, -CH2CF3, and the like.
The term "alkoxy" as used herein refers to the group-O-alkyl, wherein alkyl is as defined above. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy and hexoxy, including isomers thereof.
The term "cycloalkyl" as used herein refers to a saturated or partially unsaturated cyclic hydrocarbon group containing 3 to 12 ring carbon atoms, for example 3 to 8 ring carbon atoms, and still more for example 3 to 6 ring carbon atoms, which may have one or more rings, for example 1 or 2 rings. For example, "C3-8 cycloalkyl" means cycloalkyl having 3 to 8 ring carbon atoms as described. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantyl, and the like.
The term "heterocyclyl" or "heterocyclic" as used herein refers to a ring selected from 4 to 12 membered monocyclic, bicyclic and tricyclic, saturated and partially unsaturated, which contains at least one carbon atom in addition to at least one, such as 1-4, still more such as 1-3 or still more such as 1 or 2 heteroatoms selected from O, S and N. The point of attachment of the heterocyclyl may be on a heteroatom or on a carbon. "heterocyclyl" or "heterocyclic" also refers to a single ring comprising at least one heteroatom selected from O, S and N; or fused rings wherein at least one ring contains at least one heteroatom selected from O, S and N and the other rings are not heteroaryl or aryl, the point of attachment of which may be on a heterocycle or on other rings.
The term "cycloalkoxy" as used herein refers to the group-O-cycloalkyl, wherein cycloalkyl is as defined above. Examples of cycloalkoxy groups include, but are not limited to, cyclopropyloxy, cyclobutyloxy, including their isomers.
The term "heterocycloalkoxy" as used herein refers to the group-O-heterocycloalkyl, where heterocycloalkyl is as defined above. Examples of cycloalkoxy groups include, but are not limited to, aziridioxy, glycidoxyoxy, azetidinyloxy, oxetyloxy, including isomers thereof.
"cycloalkenyl" as used herein refers to a non-aromatic cyclic hydroxyl group containing one or more, e.g., 1, 2 or 3 carbon-carbon double bonds, containing 3 to 12 ring carbon atoms, preferably 3 to 8 ring carbon atoms, more preferably 3 to 6 ring carbon atoms, which may have one or more rings, preferably 1 or 2 rings. For example, C 3-8 Having ring carbon atoms and cycloalkenyl groups, preferably "C 3-6 Cycloalkenyl ", i.e. the cycloalkenyl group having 3 to 6 ring carbon atoms. Examples of cycloalkenyl groups include, but are not limited to, cyclopropene and, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl.
"heterocycloalkenyl" as used herein means that one or more carbon atoms in the above "cycloalkenyl" are replaced by N, O, S. For example, C 3-8 Heterocycloalkenyl of ring atoms, preferably "C 3-6 Heterocycloalkenyl ", i.e., the heterocycloalkenyl group having 3-6 ring atoms. Examples of heterocycloalkenyl groups include, but are not limited to, azetidine, oxetenyl, azapentenyl, oxapentenyl, and oxahexenyl.
The term "aryl" as used herein, unless explicitly defined, refers to a carbocyclic hydrocarbon group containing 6 to 14 ring carbon atoms, e.g., 6 to 12 ring carbon atoms, consisting of one ring or multiple rings fused together, wherein at least one ring is an aromatic ring and the other rings are not heteroaryl groups as defined below, the points of attachment of which may be on an aromatic ring or on other rings. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, 1,2,3, 4-tetrahydronaphthyl, indenyl, indanyl, azulenyl, preferably phenyl and naphthyl. As used herein, "aryl" or "aromatic" follow the huckel's rule, where pi electrons number is equal to 4n+2, n is zero or any positive integer up to 6.
The term "heterocyclyl" or "heterocyclic" as used herein refers to a ring selected from 4 to 12 membered monocyclic, bicyclic and tricyclic, saturated and partially unsaturated, which contains at least one carbon atom in addition to at least one, such as 1-4, still more such as 1-3 or still more such as 1 or 2 heteroatoms selected from O, S and N. The point of attachment of the heterocyclyl may be on a heteroatom or on a carbon. "heterocyclyl" or "heterocyclic" also refers to a single ring comprising at least one heteroatom selected from O, S and N; or fused rings wherein at least one ring contains at least one heteroatom selected from O, S and N and the other rings are not heteroaryl or aryl, the point of attachment of which may be on a heterocycle or on other rings.
The term "heteroaryl" as used herein, unless explicitly defined, refers to: a monocyclic aromatic hydrocarbon group having 5, 6 or 7 ring atoms, for example having 6 ring atoms, which contains one or more, for example 1, 2 or 3, for example 1 or 2, ring heteroatoms independently selected from N, O and S (for example N) in the ring, the remaining ring atoms being carbon atoms; and bicyclic aromatic hydrocarbon groups having 8-12 ring atoms, for example 9 or 10 ring atoms, comprising one or more, for example 1, 2, 3 or 4, for example 1 or 2, ring heteroatoms independently selected from N, O and S (for example N) in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring. For example, bicyclic heteroaryl groups include 5-6 membered heteroaryl rings fused to a 5-6 membered cycloalkyl, heterocyclyl or aryl ring, where the point of attachment may be on the heteroaryl ring or on the cycloalkyl/heterocyclyl/aryl ring. When the total number of S and O atoms in the heteroaryl group exceeds 1, these S and O heteroatoms are not adjacent to each other. Heteroaryl groups also include those in which the N-ring heteroatom is in the form of an N-oxide, such as an N-oxidized pyrimidinyl group. In some embodiments, the ring heteroatoms in the heteroaryl groups described above are N atoms, and such heteroaryl groups are referred to as "nitrogen-containing heteroaryl groups". Nitrogen-containing heteroaryl groups also include those heteroaryl groups in which the N-ring heteroatom is in the form of an N-oxide, such as a pyridyl N-oxide.
Examples of heteroaryl groups include, but are not limited to: pyridyl, N-pyridyl oxide; pyrazinyl; pyrimidinyl; pyrazolyl; imidazolyl; oxazolyl; isoxazolyl; thiazolyl; isothiazolyl; thiadiazolyl; tetrazolyl; triazolyl; thienyl; a furyl group; pyranyl; a pyrrole group; a pyridazinyl group; benzo [ d ] thiazolyl; benzodioxolyl groups, such as benzo [ d ] [1,3] dioxolyl; benzoxazolyl groups, such as benzo [ d ] oxazolyl; imidazopyridinyl, such as imidazo [1,2-a ] pyridinyl; triazolopyridinyl groups such as [1,2,4] triazolo [4,3-a ] pyridinyl and [1,2,4] triazolo [1,5-a ] pyridinyl; indazolyl; 2H-indazolyl; pyrrolo pyrimidinyl groups, such as pyrrolo [3,4-d ] pyrimidinyl, 7H-pyrrolo [2,3-d ] pyrimidinyl; pyrazolopyrimidinyl, such as pyrazolo [1,5-a ] pyrimidinyl; tetrazolopyridinyl, such as tetrazolo [1,5-a ] pyridinyl; benzothienyl; benzofuranyl; a benzimidazolinyl group; indolyl; indolinyl; purinyl groups, e.g. 9H-purinyl and 7H-purinyl
Examples of nitrogen-containing heteroaryl groups include, but are not limited to: a pyrrole group; pyrazolyl; imidazolyl; a pyridyl group; pyrazinyl; pyrimidinyl, N-oxidized pyrimidinyl; a pyridazinyl group; pyrrolo pyrimidinyl groups, such as pyrrolo [3,4-d ] pyrimidinyl, 7H-pyrrolo [2,3-d ] pyrimidinyl; purinyl groups such as 9H-purinyl and 7H-purinyl; quinolinyl; indolyl; indazolyl.
The term "hydroxy" as used herein refers to an-OH group.
The term "mercapto" as used herein refers to a-SH group.
The term "oxo" as used herein refers to an =o group.
The term "carboxy" as used herein refers to a-C (O) -OH group.
The term "cyano" as used herein refers to a-CN group.
The term "amino" as used herein refers to a-NH 2 group.
As used herein, "group" and "group" are synonymous and are used to denote a functional group or molecular fragment that can be attached to other molecular fragments.
If a formula herein comprises an asterisk "," the compound represented by the formula is a chiral compound, i.e., the compound is in the R-configuration or S-configuration. The configuration of the compounds can be determined by one skilled in the art using a variety of analytical techniques, such as single crystal X-ray crystallography and/or optical polarimetry, and according to conventional protocols.
The terms "selective," "selectively," "optional," or "optionally" as used herein mean that the subsequently described substitution pattern, event, or circumstance may or may not occur, and that the description includes instances where said substitution pattern occurs and instances where said substitution pattern does not occur. For example, "optionally substituted alkyl" includes "unsubstituted alkyl" and "substituted alkyl" as defined herein. It will be appreciated by those skilled in the art that for any group containing one or more substituents, the group does not include any sterically impractical, chemically incorrect, synthetically infeasible and/or inherently unstable substitution patterns.
The term "including but not limited to" as used herein means that the selected range is preferably followed by the recited group, but that groups not recited may also be used.
The term "substituted" or "substituted with … …" as used herein means that one or more hydrogen atoms on a given atom or group is replaced with one or more substituents selected from a given set of substituents, provided that the normal valence of the given atom is not exceeded. When the substituent is oxo (i.e., =o), then two hydrogen atoms on a single atom are replaced with oxygen. Such combinations are permissible only if the combination of substituents and/or variables result in chemically correct and stable compounds. Chemically correct and stable compounds means that the compounds are stable enough to be separated from the reaction mixture and to determine the chemical structure of the compounds and can then be formulated into a formulation having at least practical utility.
Unless otherwise indicated, substituents are named into the core structure. For example, it is understood that when a (cycloalkyl) alkyl group is listed as one possible substituent, it means that the point of attachment of the substituent to the core structure is in the alkyl moiety.
The term "substituted with one or more substituents" as used herein means that one or more hydrogen atoms on a given atom or group are independently replaced with one or more substituents selected from the given group. In some embodiments, "substituted with one or more substituents" means that a given atom or group is substituted with 1, 2, 3, or 4 substituents independently selected from a given group.
It will be appreciated by those skilled in the art that some compounds of formula (I) may contain one or more chiral centers and thus two or more stereoisomers may be present. Racemic mixtures of these isomers, single isomers and one enantiomerically enriched mixture, as well as diastereomeric and particular diastereomeric partially enriched mixtures when there are two chiral centers are all within the scope of the invention. It will also be understood by those skilled in the art that the present invention includes all individual stereoisomers (e.g. enantiomers), racemic mixtures or partially resolved mixtures of compounds of formula (I) and, where appropriate, individual tautomers thereof.
In other words, in some embodiments, the present invention provides compounds containing multiple stereoisomers in purity, i.e., diastereomeric or enantiomeric purity, expressed in terms of different "ee" or "de" values. In some embodiments, a compound of formula (I) (e.g., as described herein) has an enantiomeric purity of at least 60% ee (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9% ee, or any value between these recited values). In some embodiments, the compound of formula (I) (e.g., as described herein) has an enantiomeric purity of greater than 99.9% ee, up to 100% ee. In some embodiments, a compound of formula (I) (e.g., as described herein) has a diastereomeric purity of at least 60% de (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9% de, or any value between these recited values). In some embodiments, the compound of formula (I) (e.g., as described herein) has a diastereomeric purity of greater than 99.9% de.
The term "enantiomeric excess" or "ee" refers to how much one enantiomer is relative to another enantiomer. For mixtures of R and S enantiomers, the percent enantiomeric excess is defined as |r-s| 100, where R and S are the molar or weight fractions of the respective enantiomers in the mixture, r+s=1. If the optical rotation of a chiral species is known, the percent enantiomeric excess is defined as ([ a ] obs/[ a ] max) ×100, where [ a ] obs is the optical rotation of the enantiomeric mixture and [ a ] max is the optical rotation of the pure enantiomer.
The term "diastereomeric excess" or "de" refers to how much one diastereomer is relative to another diastereomer, and is defined by analogy in terms of enantiomeric excess. Thus, for a mixture of diastereomers D1 and D2, the percent diastereomeric excess is defined as |d1-d2| 100, where D1 and D2 are the molar or weight fraction of each diastereomer in the mixture, d1+d2=1.
Determination of diastereomer and/or enantiomeric excess can be accomplished using a variety of analytical techniques, including nuclear magnetic resonance spectroscopy, chiral column chromatography, and/or optical polarimetry, and according to conventional protocols familiar to those skilled in the art.
The racemic mixture may be used as such or resolved into individual isomers. The resolution can be carried out to obtain a stereochemically pure compound or a mixture enriched in one or more isomers. Methods for separating isomers are well known and include physical methods such as chromatography using chiral adsorbents. Individual isomers of chiral form can be prepared from chiral precursors. Alternatively, one or both of the isomers substantially free of the other isomer, i.e., the desired stereoisomer having an optical purity of, for example, at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 99.5% by weight, may be obtained by chemical separation of the individual isomers from the mixture by formation of diastereomeric salts with chiral acids (e.g., individual enantiomers of 10-camphorsulfonic acid, camphoric acid, α -bromocamphoric acid, tartaric acid, diacetyltartaric acid, malic acid, pyrrolidone-5-carboxylic acid, etc.), fractional crystallization of said salts, and then liberating one or both of the resolved bases, optionally repeating the process. Alternatively, as is well known to those skilled in the art, the racemate may be covalently attached to a chiral compound (adjunct) to provide diastereomers, which may be separated by chromatography or fractional crystallization, followed by chemical removal of the chiral adjunct to provide the pure enantiomer.
The term "pharmaceutically acceptable salt" as used herein refers to salts of the free acid or base of a compound of formula (I) that are non-toxic, biologically tolerable or otherwise biologically suitable for administration to a subject being treated. .
"pharmaceutically acceptable salts" include, but are not limited to: acid addition salts of the compounds of formula (I) with inorganic acids, such as hydrochloride, hydrobromide, carbonate, bicarbonate, phosphate, sulfate, sulfite, nitrate, etc.; and acid addition salts of compounds of formula (I) with organic acids, such as formate, acetate, malate, maleate, fumarate, tartrate, succinate, citrate, lactate, methanesulfonate, p-toluenesulfonate, 2-hydroxyethanesulfonate, benzoate, salicylate, stearate and salts with alkanedicarboxylic acids of formula HOOC- (CH 2) n-COOH (where n is 0-4), and the like. "pharmaceutically acceptable salts" also include base addition salts of compounds of formula (I) bearing an acidic group with pharmaceutically acceptable cations such as sodium, potassium, calcium, aluminum, lithium and ammonium. The molar ratio of the compound of formula (I) to the acid or cation in the resulting pharmaceutically acceptable salt includes, but is not limited to, 1:1, 1:2, 1:3, and 1:4.
The term "prodrug" as used herein refers to a pharmacological substance derived from a parent drug (i.e., drug) that, once administered, is metabolized in vivo to the parent drug. Prodrugs can be prepared by substituting one or more functional groups present in the compound, wherein the substituents of the prodrug are removed in vivo in such a way as to be converted to the parent compound. Preparation and use of prodrugs can be found in T.Higuchi and V.stilla, "Pro-drugs asNovel Delivery Systems," Vol.14of the A.C.S. symposium Series and Bioreversible. "prodrugs" include, but are not limited to: esters of the compounds of formula (I) such as phosphates, formates, carbamates; amides such as formamide, acetamide.
Furthermore, if the compounds described herein are obtained in the form of an acid addition salt, the free base form thereof may be obtained by basifying a solution of the acid addition salt. Conversely, if the product is in the form of the free base, its acid addition salts, particularly the pharmaceutically acceptable acid addition salts, can be obtained by dissolving the free base in a suitable solvent and treating the solution with an acid according to conventional procedures for preparing acid addition salts from basic compounds. One skilled in the art can determine, without undue experimentation, various synthetic methods that can be used to prepare non-toxic pharmaceutically acceptable acid addition salts.
The term "solvate" means a solvent addition form comprising a stoichiometric or non-stoichiometric amount of solvent. Some compounds have a tendency to cross a fixed molar ratio of solvent molecules in the solid state, thereby forming solvates. If the solvent is water, the solvate formed is a hydrate, and when the solvent is ethanol, the solvate formed is an ethanolate. Hydrates are formed by one or more molecules of water with a molecule of the substance, wherein the water retains its molecular state of H2O, and such a combination is capable of forming one or more hydrates, such as hemihydrate, monohydrate, and dihydrate, as well as variable hydrates.
The terms "group" and "radical" as used herein are synonymous and are used to denote a functional group or molecular fragment that can be attached to other molecular fragments.
The term "active ingredient" is used to denote a chemical substance having biological activity. In some embodiments, an "active ingredient" is a chemical substance that has pharmaceutical use. In the united states, the actual pharmaceutical activity can be determined by appropriate preclinical testing, whether in vitro or in vivo. But is sufficiently pharmaceutically active to be accepted by regulatory authorities (e.g., FDA in the united states) to have a higher standard than preclinical testing. Whether such a higher standard of pharmaceutical activity is successful or not is generally reasonably unexpected from preclinical test results, can be established by appropriate and effective randomized, double-blind, controlled clinical trials in humans.
The term "treating" or "treatment" of a disease or disorder, where a beneficial therapeutic effect is achieved, refers to administering one or more pharmaceutical substances, in particular a compound of formula (I) as described herein and/or a pharmaceutically acceptable salt thereof, to an individual, e.g. a human, suffering from, or having symptoms of, or predisposed to, the disease or disorder, for curing, healing, alleviating, altering, treating, ameliorating, improving or affecting the disease or disorder, the symptoms of the disease or disorder, or the predisposition to the disease or disorder. In some embodiments, the disease or disorder is cancer.
When chemical reactions are involved, the terms "treat," "contacting," and "reacting" mean adding or mixing two or more reagents under appropriate conditions to produce the indicated and/or desired product. It will be appreciated that the reaction to produce the indicated and/or desired product may not necessarily result directly from the combination of the two reagents initially added, i.e., there may be one or more intermediates formed in the mixture that ultimately lead to the formation of the indicated and/or desired product.
The term "effective amount" as used herein refers to an amount or dose of a PI3K inhibitor that is generally sufficient to produce a beneficial therapeutic effect in a patient in need of treatment for a disease or disorder mediated by PI3K activity. The effective amount or dosage of an active ingredient of the present invention can be determined by conventional means (e.g., modeling, dose escalation studies or clinical trials) in combination with conventional influencing factors (e.g., the manner or route of administration or administration, the pharmacokinetics of the pharmaceutical ingredient, the severity and course of the disease or disorder, the previous or ongoing treatment of the individual, the health condition and response of the individual to the drug, and the judgment of the attending physician). In the united states, determination of effective doses is generally difficult to predict from preclinical trials. In fact, dosages are completely unpredictable, and new unpredictable dosage regimens may develop after the initial use in randomized, double-blind, controlled clinical trials.
Typical dosages range from about 0.0001 to about 200 milligrams of active ingredient per kilogram of body weight of the subject per day, for example from about 0.001 to 100 milligrams per kilogram of body weight per day, or from about 0.01 to 35 milligrams per kilogram per day, or from about 0.1 to 10 milligrams per kilogram of body weight per day, once daily or in divided dosage units (e.g., twice daily, three times daily, four times daily). For a 70 kg person, a suitable dosage range may be about 0.05 to about 7 grams per day, or about 0.2 to about 5 grams per day. Once the patient's disease or disorder has improved, the dosage may be adjusted to maintain the treatment. For example, the amount or number of administrations, or the amount and number of administrations, may be reduced to a level that maintains the desired therapeutic effect, depending on the change in symptoms. Of course, if symptoms are reduced to appropriate levels, treatment may be stopped. However, for recurrence of symptoms, the patient may require intermittent long-term treatment.
The term "inhibition" refers to a decrease in baseline activity of a biological activity or process. The term "inhibiting PI3K activity" is the actual pharmaceutical activity for the purposes of the present invention and refers to a decrease in PI3K activity in the presence of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof as described herein, relative to PI3K activity in the absence of the compound of formula (I) and/or a pharmaceutically acceptable salt thereof. The reduction in activity may be caused by direct interaction of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof as described herein with PI3K, or by interaction of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof as described herein with one or more other factors which in turn affect PI3K activity. For example, the presence of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof as described herein may reduce PI3K activity by binding directly to PI3K, may reduce PI3K activity by affecting another factor directly or indirectly, or may reduce PI3K activity by reducing the amount of PI3K present in a cell or organism directly or indirectly.
The term "individual" as used herein refers to both mammalian and non-mammalian animals. By mammal is meant any member of the mammal, including but not limited to: a person; non-human primates such as chimpanzees and other apes and monkey species; farm animals such as cattle, horses, sheep, goats and pigs; domestic animals such as rabbits, dogs, and cats; laboratory animals, including rodents, such as rats, mice and guinea pigs; etc. Examples of non-mammals include, but are not limited to, birds and the like. The term "individual" is not limited to a particular age or sex. In some embodiments, the individual is a human.
Generally, the term "about" is used herein to adjust a given value to be above or below 20% of that value.
In one embodiment, the compound of formula (I) and/or a pharmaceutically acceptable salt thereof,
Figure BDA0003412931580000191
wherein:
R 1 independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, mercapto, amino, -C 1-6 Alkyl, -C 1-6 Alkoxy, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocyclylalkyl, -C 3-6 Cycloalkoxy, -C 3-6 Heterocyclyloxy, -C 1-6 Alkyl (O) C 1-6 Alkyl, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR 1a Aryl, aralkyl, aralkoxy, heteroaryl, heteroaralkyl, heteroaralkoxy; wherein said mercapto, amino, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkoxy, heterocycloalkyl, heterocycloalkoxy, aryl, aralkyl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Optionally substituted by one or more R 1a And (3) substitution.
R 1a Selected from hydrogen, deuterium, halogen, cyano, hydroxy, mercapto, amino, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkoxy, heterocycloalkyl, heterocycloalkoxy, aryl, heteroaryl, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, heteroaryl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Selectively by halogen, cyano, -OR a 、-SR a 、NR b R c Alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycleAny one or more of the oxy groups are substituted or unsubstituted;
R 2 selected from deuterium, hydroxy, -C 1-6 Alkyl, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocyclylalkyl, -C 3-6 Cycloalkoxy, -C 3-6 Heterocyclyloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) NH (C) 1-6 Alkyl group 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 、-C(O)OR 2a 、-C(O)NH 2 Aryl, heteroaryl; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, heteroaryl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Can be optionally substituted with one or more R 2a Substitution;
R 2a selected from the group consisting of hydrogen, deuterium, halogen, cyano, hydroxy, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Is optionally selected from halogen, cyano, -OR a 、-SR a 、NR b R c One or more of alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocyclyloxy, substituted or unsubstituted;
and/or R 1 And R is R 2 Optionally forming 3-6 membered cycloalkyl, 3-6 membered heterocycloalkyl, 3-6 membered cycloalkenyl, 3-6 membered heterocycloalkenyl, aryl, heteroaryl with the carbon atom to which each is attached, and optionally substituted with one or more R 1a Or R is 2a Substitution; and the carbon atom in the cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl group orThe sulfur atom may be selectively oxo to form part of a cyclic carbonyl group, an amide group, an ester group, a sulfone group (-S (O) 2-), a sulfonamide group (-S (O) 2 NH-).
R 3 Selected from hydrogen, deuterium, C 1-6 Alkyl, deuterated alkyl, halogenated C 1-6 Alkyl, hydroxy substituted C 1-6 Alkyl, cyanoalkyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocycloalkyl, alkoxyalkyl, cycloalkylalkyl, heterocyclylalkyl, cycloalkoxyalkyl, heterocycloalkoxyalkyl;
R 4 independently selected from heteroaryl groups, said heteroaryl groups being monocyclic aromatic hydrocarbon groups having 5, 6 or 7 ring atoms which contain 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or bicyclic aromatic hydrocarbon groups having 9 or 10 ring atoms which contain 1, 2, 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring, and said heteroaryl groups include those heteroaryl groups in which the N ring heteroatoms are in the form of an N-oxide, and said heteroaryl groups are optionally substituted with one or more substituents selected from the group consisting of: halogen, deuterium, cyano, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, cycloalkoxy, heterocyclyl, heterocyclyloxy, -NH 2 、-NH(C 1-6 Alkyl), -N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -COOH, -C (O) NH 2 -C (O) NH (C1-6 alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a 、-NHC(O)(C 1-6 Alkyl), -NHC (O) (C) 1-6 Cycloalkyl), -NHC (O) O (C) 1-6 Alkyl), -NHC (O) O (C) 1-6 Cycloalkyl), phenyl, and 5-or 6-membered heteroaryl; wherein C is 1-6 Alkyl, haloalkyl, -NH 2 、-COOH、-C(O)NH 2 Cycloalkyl, heterocycloalkyl, optionally substituted with one or more R 1a Optionally substituted; wherein R is as R 4 The 5 or 6 membered heteroaryl group of the substituents of (a) is a monocyclic aromatic hydrocarbon group having 5 or 6 ring atoms which contains 1, 2 or 3 ring heteroatoms independently selected from N, O or S in the ring, the remaining ring atoms being carbon atoms, when the total of S and O atoms in the heteroaryl group exceeds1, wherein the S and O heteroatoms are not adjacent to each other, and wherein the phenyl or 5 or 6 membered heteroaryl as substituents for the heteroaryl is optionally substituted with one or more substituents selected from the group consisting of: halogen, amino, hydroxy, mercapto, C1-6 alkyl and-O (C1-6 alkyl), haloalkyl;
R 5 independently selected from C 1-6 Alkyl, hydroxy-substituted alkyl, deuterated alkyl, hydroxyalkyl, C 3-6 Cycloalkyl, C 3-6 Heterocycloalkyl, cycloalkylalkyl or cycloalkoxyalkyl, wherein said alkyl is optionally substituted with one or more R 5a Substitution; r is R 5a Selected from deuterium, halogen, CN, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl; wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl groups may be substituted with halogen, CN, -OR a 、-SR a 、-NR b R c Alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl substitution;
and/or R 4 And R is 5 Optionally together with the N atom to which they are attached, form a nitrogen-containing heterocyclic group or heteroaryl group, wherein the nitrogen-containing heterocyclic group and heteroaryl group may be optionally substituted with one or more substituents including, but not limited to, halogen, deuterium, amino, hydroxy, mercapto, cyano, alkyl, deuteroalkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a 、-OR a 、-SR a 、NR b R c
Wherein R is a 、R b And R is c Independently selected from hydrogen, deuterium, halogen, C 1-6 Alkyl, deuterated C 1-6 Alkyl, halogenated C 1-6 Alkyl, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocycloalkyl, hydroxy-substituted C 1-6 Alkyl, -C 1-6 Alkyl (O) C 1-6 Alkyl, aryl, heteroaryl;
and/or R b And R is c Together with the nitrogen atom to which they are both attached form a 4-to 7-membered heterocycloalkyl or are substituted with 1 to 3R a And (3) substitution.
Further, the aryl group is a carbocyclic hydrocarbon group containing 6 to 12 ring carbon atoms consisting of one ring or a plurality of rings condensed, wherein at least one ring is an aromatic ring and the other rings are not heteroaryl groups;
the heteroaryl is a monocyclic aromatic hydrocarbon group having 5, 6 or 7 ring atoms which contains 1 or 2 ring heteroatoms independently selected from N, O and S in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 or 10 ring atoms which contains 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring, when the total number of S and O atoms on the heterocycle exceeds 1, the S and O atoms are not adjacent to each other, and the heteroaryl includes those heteroaryl groups in which the N heteroatom is in the form of an N-oxide;
the aryl and heteroaryl groups are optionally substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, hydroxy, mercapto, -C 1-6 Alkyl, haloalkyl, alkoxy, haloalkoxy, haloalkoxyalkyl, cycloalkyl, halocycloalkyl, - (C) 1-6 Alkyl) OH and-SO 2 (C 1-6 Alkyl), alkenyl, alkynyl.
Further, the aryl or heteroaryl is selected from aryl and heteroaryl groups having 6 ring atoms, heteroaryl groups having 5 ring atoms, bicyclic heteroaryl groups having 9 or 10 ring atoms, or aryl groups;
wherein the aryl and/or heteroaryl group comprises phenyl, deuterated phenyl, naphthyl, pyridinyl, pyrimidine, pyridazine, piperazine, pyrazolyl, imidazolyl, quinolinyl, thienyl, benzothienyl, indolyl, optionally substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, hydroxy, -C 1-6 Alkyl, - (C) 1-6 Alkyl) OH, C 1-6 Haloalkyl and-SO 2 (C) 1-6 Alkyl).
Further, R4 is selected from heteroaryl, or R4 and R5 form heteroaryl;
wherein the heteroaryl group is a monocyclic aromatic hydrocarbon group having 6 ring atoms which contains 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 ring atoms which contains 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring and the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of: halogen, cyano, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl) -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-C(O)NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), phenyl, and 5-or 6-membered heteroaryl.
Further, R 4 Selected from heteroaryl, or R 4 And R is R 5 Forming a heteroaryl group;
wherein the heteroaryl group is a monocyclic aromatic hydrocarbon group having 6 ring atoms which contains 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 ring atoms which contains 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring and the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, hydroxy, mercapto, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-C(O)NH 2 、-C(O)NH(C 1-6 Alkyl), phenyl, pyridyl, pyrimidinyl, piperazinyl, pyridazinyl, pyrazolyl, imidazolyl, triazolyl, and tetrazolyl.
Further, the R 4 Optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, mercapto, amino, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl).
Further, the aryl group in the heteroaryl group is selected from pyrimidine, purinyl, pyrazolopyrimidine, pyrrolopyrimidine.
Further, R 4 Or R is 4 And R is R 5 The heteroaryl group formed is selected from the substituents shown below
Figure BDA0003412931580000221
Wherein the substituents are optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, hydroxy, mercapto, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-CONH 2 、-C(O)NH(C 1-6 Alkyl), phenyl, pyridyl, pyrazolyl, imidazolyl, oxazolyl, triazolyl and tetrazolyl, wherein R is 4 Phenyl, pyridinyl, pyrimidinyl, piperazinyl, pyridazinyl, pyrazolyl, imidazolyl, triazolyl and tetrazolyl of the substituents are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl).
Further, R4 is selected from
Figure BDA0003412931580000222
Optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), -CO2H, -CONH 2 Phenyl, pyridyl, pyrazolyl, triazolyl and tetrazolyl,
wherein R is as R 4 Phenyl, pyridyl, pyrazolyl, imidazolyl, triazolyl and tetrazolyl of the substituents are optionally substituted with one or more substituents selected from the group consisting ofGroup substitution: halogen, hydroxy, C 1-6 Alkyl and-O (C) 1-6 Alkyl).
Further, R 3 Is C 1-4 Alkyl and C 1-4 A haloalkyl group.
Further, R 3 Methyl, ethyl and trifluoromethyl.
General synthetic method of the above compound of formula (I)
Scheme 1
Figure BDA0003412931580000231
As shown in scheme 1, the compound of formula i-1 is reacted with diethyl ethoxymethylene malonate under basic conditions to provide the pyrazolopyrimidine compound of formula i-2. The compound of formula i-2 is reacted with phosphorus oxychloride to give a compound of formula i-3. Compounds of the formula i-3 with ArB (OH) 2 The compounds of formula (Ar) are as defined herein are prepared by Suzuki coupling reactions under catalysis of a suitable palladium reagent. The palladium-catalyzed carbon-carbon coupling reaction may be carried out under suitable conditions: such as in a suitable polar solvent such as DMF, ACN, THF or DMSO, etc.; in a suitable base such as TEA, DIEPA, cs 2 CO 3 KOAc, etc.; using catalysts such as Pd (OAc) 2 ,Pd(dppf) 2 Cl 2 ,Pd(PPh 3 ) 4 Or Pd (or) 2 (dba) 3 Etc. The following reaction may proceed as follows:
the compound of formula i-4 is subjected to an ester hydrolysis reaction to obtain a compound acid of formula i-5. The compound acid of formula i-5 is reacted with N, O-dimethylhydroxylamine to give an amide compound of formula i-6. The resulting compound of formula i-6 is reacted under appropriate conditions with a grignard reagent (alkyl magnesium halide) to produce a compound of formula i-7. Introducing halogen into the compound of formula i-7 under proper conditions to obtain a compound of formula i-8, or further modifying the obtained halogenide to obtain a compound of formula i-8. The compound of formula i-8 is condensed with (R) -2-methylpropane-2-sulfinamide and then reduced in the presence of a suitable reducing agent to give the compound of formula i-10. Removing the protecting group from the compound of formula i-10 to give a compound of formula i- 11. The compound of formula i-11 is reacted with R in the presence of a base (e.g., DIEPA, etc.), under appropriate conditions 4 -Cl reaction, and/or further derivatization (R 5 -x) to give the compound of formula (I).
Figure BDA0003412931580000241
As shown in scheme 2, compounds of formula i-4 may be synthesized from alternative routes. The compound of formula i-12 is reacted with ethyl acetate under basic conditions to give the compound of formula i-13. Refluxing the compound of formula i-13 with DMF-DMA in toluene, then reacting the concentrated reaction with further compound of formula i-1 under re-acidic conditions to yield compound of formula i-4.
Pharmaceutical composition and use thereof
The compounds of formula (I) described herein (e.g., any of the compounds herein) and/or pharmaceutically acceptable salts thereof, may be formulated into pharmaceutical compositions alone or in combination with one or more additional active ingredients. The pharmaceutical composition comprises: (a) An effective amount of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof as described herein; and (b) a pharmaceutically acceptable excipient (e.g., a pharmaceutically acceptable carrier).
Pharmaceutically acceptable carrier refers to a carrier that is compatible with the active ingredient of the composition (and, in some embodiments, stabilizes the active ingredient) and is not deleterious to the subject to be treated. For example, solubilizing agents such as cyclodextrins (which are capable of forming specific, more soluble complexes with the compounds of formula (I) and/or pharmaceutically acceptable salts thereof described herein) can be used as pharmaceutical excipients to deliver active ingredients. Examples of other carriers include colloidal silica, magnesium stearate, cellulose, sodium lauryl sulfate, and pigments such as D & C Yellow No. 10 (D & C Yellow # 10). Suitable pharmaceutically acceptable carriers are disclosed in a reference book (Remington's Pharmaceutical Sciences, a.osol) standard in the art.
Pharmaceutical compositions comprising a compound of formula (I) described herein (e.g., any of the compounds herein) and/or a pharmaceutically acceptable salt thereof may be administered in a variety of known ways, such as orally, topically, rectally, parenterally, inhaled or implanted, among others. The term "parenteral" as used herein includes subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intraspinal, intralesional and intracranial injection or infusion.
The pharmaceutical composition described herein may be prepared in the form of tablets, capsules, sachets, dragees, powders, granules, lozenges, powder injection, liquid preparations or suppositories. In some embodiments, pharmaceutical compositions comprising a compound of formula (I) and/or a pharmaceutically acceptable salt thereof may be formulated for intravenous drip, topical administration, or oral administration.
The orally administered composition may be any orally acceptable dosage form including, but not limited to: tablets, capsules, emulsions, and aqueous suspensions, dispersions, and solutions. Common tablet carriers include lactose and corn starch. Lubricants such as magnesium stearate are also commonly added to tablets. Useful diluents include lactose and dried cornstarch when administered orally in the form of capsules. When administered orally in the form of an aqueous suspension or emulsion, the active ingredient may be suspended or dissolved in the oil phase with emulsifying or suspending agents. If desired, certain sweeteners, flavoring agents or pigments may also be added.
In some embodiments, the amount of the compound of formula (I) and/or a pharmaceutically acceptable salt thereof in the tablet may be 1, 5, 10, 15, 20, 25, 50, 75, 80, 85, 90, 95, 100, 125, 150, 200, 250, 300, 400, and 500 milligrams. In some embodiments, the amount of the compound of formula (I) and/or a pharmaceutically acceptable salt thereof in the capsule may be 1, 5, 10, 15, 20, 25, 50, 75, 80, 85, 90, 95, 100, 125, 150, 200, 250, 300, 400, and 500 milligrams.
Sterile injectable compositions (e.g., aqueous or oleaginous suspensions) may be formulated according to the art-known techniques using suitable dispersing or wetting agents (e.g., tween 80) and suspending agents. The sterile injectable intermediate medium may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Among the pharmaceutically acceptable carriers and solvents that may be used are mannitol, water, ringer's solution and physiological saline. In addition, sterile, nonvolatile oils, such as synthetic mono-or diglycerides, are conventionally employed as a solvent or suspending medium. Fatty acids such as oleic acid and its glyceride derivatives, as well as natural pharmaceutically-acceptable oils such as olive oil or castor oil (especially in their polyoxyethylated versions) are commonly used as injectable intermediate media. These oil solutions or suspensions may also contain a long-chain alcoholic diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents.
Inhalation compositions may be prepared according to techniques well known in the art of pharmaceutical formulation, or may be prepared as solutions in saline, using benzyl alcohol or other suitable preservatives, using absorption promoters to enhance bioavailability, using fluorocarbons and/or other solubilizing or dispersing agents known in the art.
Topical compositions may be formulated as oils, creams, lotions, ointments and the like. Suitable carriers for the compositions include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohols (i.e., alcohols having more than 12 carbon atoms). In some embodiments, the pharmaceutically acceptable carrier is a carrier in which the active ingredient is soluble. The composition may further comprise emulsifiers, stabilizers, wetting agents and antioxidants, as well as substances imparting color or fragrance thereto, if desired. In addition, transdermal penetration enhancers may be added to the topical formulation. Examples of such accelerators can be found in U.S. Pat. Nos. 3,989,816 and 4,444,762.
The cream may be formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which the active ingredient dissolved in a small amount of oil, such as almond oil, is mixed. One example of a cream comprises about 40 parts by weight water, about 20 parts beeswax, about 40 parts mineral oil, and about 1 part almond oil. Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil, such as almond oil, with warm soft paraffin and cooling the mixture. One example of an ointment comprises about 30% almond oil and about 70% white soft paraffin by weight.
Suitable in vitro assays may be used to evaluate the practical use of the compounds of formula (I) and/or pharmaceutically acceptable salts thereof described herein in inhibiting PI3K activity. Further practical use of the compounds of formula (I) and/or pharmaceutically acceptable salts thereof described herein in the treatment of cancer or autoimmune diseases may be detected by in vivo assays. For example, a compound of formula (I) as described herein and/or a pharmaceutically acceptable salt thereof may be administered to an animal (e.g., a mouse model) suffering from cancer or an autoimmune disease, and then its therapeutic effect assessed and if the result of a preclinical test is successful, its dosage range and route of administration to the animal, e.g., a human, may also be predicted.
The compounds of formula (I) and/or pharmaceutically acceptable salts thereof described herein may exhibit sufficient preclinical practical use to be worth conducting clinical trials and are expected to exhibit beneficial therapeutic or prophylactic effects, for example, in individuals suffering from cancer.
The term "cancer" as used herein refers to a cellular disorder characterized by uncontrolled or deregulated cell proliferation, reduced cell differentiation, improper ability to invade surrounding tissue, and/or the ability to establish new foci at other sites. The term "cancer" includes, but is not limited to: solid tumors and hematological malignancies. The term "cancer" includes cancers of the skin, tissues, organs, bones, cartilage, blood and blood vessels. The term "cancer" includes both primary and metastatic cancers.
Non-limiting examples of solid tumors include pancreatic cancer; bladder cancer; colorectal cancer; breast cancer, including metastatic breast cancer; prostate cancer, including androgen-dependent and non-androgen-dependent prostate cancer; renal cancers, including, for example, metastatic renal cell carcinoma; hepatocellular carcinoma; lung cancer, including, for example, non-small cell lung cancer (NSCLC), bronchioloalveolar cancer (BAC), and lung adenocarcinoma; ovarian cancer, including, for example, progressive epithelial cancer or primary peritoneal cancer; cervical cancer; stomach cancer; esophageal cancer; head and neck cancers, including, for example, head and neck squamous cell carcinoma; skin cancers, including, for example, malignant melanoma; neuroendocrine cancers, including metastatic neuroendocrine tumors; brain tumors, including, for example, glioma, anaplastic oligodendroglioma (anaplastic oligodendroglioma), adult glioblastoma multiforme, and adult anaplastic astrocytoma; bone cancer; soft tissue sarcoma; and thyroid cancer.
Non-limiting examples of hematological malignancies include Acute Myelogenous Leukemia (AML); chronic Myelogenous Leukemia (CML), including acceleration phase CML and CML acute phase (CML-BP); acute Lymphoblastic Leukemia (ALL); chronic Lymphocytic Leukemia (CLL); hodgkin lymphoma; non-hodgkin lymphomas (NHL), including follicular lymphomas and mantle cell lymphomas; b cell lymphoma; t cell lymphomas; multiple Myeloma (MM); waldenstrom macroglobulinemia (Waldenstrom' smacroglobinemia); myelodysplastic syndrome (myelodysplastic syndrome, MDS), including refractory anemia (refractory anemia, RA), refractory iron granulomatous anemia (refractory anemia with ringed siderblast, RARS), refractory anemia of excessive bud cells (refractory anemia with excess blast, RAEB) and refractory anemia of excessive bud cells combined with acute transformation (refractory anemia with excess blast in transformation, RAEB-T); myeloproliferative syndrome (myeloproliferative syndrome).
In some embodiments, typical hematological malignancies include leukemias, such as Acute Lymphoblastic Leukemia (ALL), acute Myelogenous Leukemia (AML), chronic Lymphoblastic Leukemia (CLL), and Chronic Myelogenous Leukemia (CML); multiple Myeloma (MM); and lymphomas such as hodgkin's lymphoma, non-hodgkin's lymphoma (NHL), mantle Cell Lymphoma (MCL), follicular lymphoma, B-cell lymphoma, T-cell lymphoma, and diffuse large B-cell lymphoma (DLBCL).
The term "inflammatory disease" refers to a pathological condition that leads to an inflammatory response, particularly due to neutrophil chemotaxis. Examples of such diseases include inflammatory skin diseases (including psoriasis and atopic dermatitis); systemic scleroderma and scleroderma; responses associated with Inflammatory Bowel Disease (IBD) (e.g., crohn's disease and ulcerative colitis); ischemia reperfusion injury, including surgically induced tissue reperfusion injury, myocardial ischemia such as myocardial infarction, cardiac arrest, post-cardiac reperfusion and abnormal contractile response of coronary vessels following percutaneous transluminal coronary angioplasty, stroke and abdominal aortic aneurysm surgical tissue reperfusion injury; cerebral edema secondary to stroke; cranium trauma; hemorrhagic shock; suffocation; adult respiratory distress syndrome; acute lung injury; behcet's disease; dermatomyositis; polymyositis; multiple Sclerosis (MS); dermatitis is treated; meningitis; encephalitis; uveitis; osteoarthritis; lupus nephritis; autoimmune diseases such as Rheumatoid Arthritis (RA); sjogren's syndrome; vasculitis; diseases involving leukocyte exudation; inflammatory diseases of the Central Nervous System (CNS) secondary to sepsis or trauma, multiple organ injury syndrome; alcoholic hepatitis; bacterial pneumonia; antigen-antibody complex mediated diseases, including glomerulonephritis; sepsis; sarcoidosis; immunopathogenic response by tissue/organ transplantation; pulmonary inflammation, including pleurisy, alveolitis, vasculitis, pneumonia, chronic bronchitis, bronchiectasis, diffuse panbronchiolitis, allergic pneumonia, idiopathic Pulmonary Fibrosis (IPF), cystic fibrosis, and the like. Preferred indications include, but are not limited to, chronic inflammation, autoimmune diabetes, rheumatoid Arthritis (RA), rheumatoid spondylitis, gouty arthritis and other arthritic conditions, multiple Sclerosis (MS), asthma, systemic lupus erythematosus, adult respiratory distress syndrome, behcet's disease, psoriasis, chronic pulmonary inflammatory diseases, graft versus host reaction, crohn's disease, ulcerative colitis, inflammatory Bowel Disease (IBD), alzheimer's disease and poliomyelitis.
The term "infectious disease" refers to a pathological condition caused by infection with a virus, bacterium, parasite, etc. Non-limiting examples of such diseases include leishmaniasis, novel coronavirus infection pneumonia, sepsis.
The term "pulmonary disease" refers to a pathological condition associated with a pulmonary organ. Non-limiting examples of such diseases include bronchiectasis, idiopathic pulmonary fibrosis, asthma, chronic obstructive pulmonary disease, pneumonia, acute lung injury, acute respiratory distress syndrome.
The term "bone marrow related disease" refers to a pathological condition associated with bone marrow organs. Non-limiting examples of such diseases include myelofibrosis, polycythemia vera.
The compounds of formula (I) and/or pharmaceutically acceptable salts thereof described herein may be used to achieve beneficial therapeutic or prophylactic effects, for example, in individuals suffering from autoimmune diseases.
The term "autoimmune disease" refers to a disease or condition caused by an immune response of an organism to an autoantigen resulting in damage to self tissues or organs. Examples of autoimmune diseases include, but are not limited to: chronic Obstructive Pulmonary Disease (COPD), allergic rhinitis, lupus erythematosus, myasthenia gravis, multiple Sclerosis (MS), rheumatoid Arthritis (RA), psoriasis, inflammatory Bowel Disease (IBD), asthma and idiopathic thrombocytopenic purpura (idiopathic thrombocytopenicpurpura) and myeloproliferative diseases (myeloid proliferative disorder), such as myelofibrosis (myelofilrosis), polycythemia vera/primary thrombocythemia myelofibrosis (postpolycythemia vera/essential thrombocytosis myelofibrosis, PV/ET).
In some embodiments, the inflammatory and autoimmune diseases include rheumatoid arthritis, chronic Obstructive Pulmonary Disease (COPD), allergic rhinitis, asthma, lupus erythematosus, systemic lupus erythematosus, psoriasis, and multiple sclerosis.
Furthermore, the compounds of formula (I) described herein (e.g., any of the compounds herein) and/or pharmaceutically acceptable salts thereof can be used in combination with additional active ingredients for the treatment of cancer, inflammatory or autoimmune diseases. The additional active ingredient may be administered separately from the compound of formula (I) and/or pharmaceutically acceptable salts thereof described herein, or it may be included in a pharmaceutical composition according to the present disclosure, e.g., a fixed dose combination. In a typical embodiment, the additional active ingredient is one that is known or has been found to be effective in treating a disease mediated by PI3K activity, such as another PI3K modulator or a compound that is effective in antagonizing another target associated with the particular disease. The combination may be used to increase the efficacy (e.g., by including in the combination a compound that enhances the efficacy or effectiveness of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof as described herein), reduce one or more side effects, or reduce the required dose of a compound of formula (I) and/or a pharmaceutically acceptable salt thereof as described herein.
Examples
The following examples are illustrative of the invention and are not intended to limit the invention in any way. All fractions are weight fractions and temperatures are degrees celsius unless otherwise indicated. The pressure is at or near atmospheric pressure. All data were measured by Agilent 6120and/or 1100. All reagents used in the present invention are commercially available except for synthetic intermediates. The names of all compounds except the reagent were generated by ChemDrew 20.0.
ACN acetonitrile
Boc t-Butoxycarbonyl group
(Boc) 2 O-pyrocarbonic acid tert-butyl ester
DAST diethylaminosulfur trifluoride
DCM dichloromethane
DEA diethylamino
DMF N, N-dimethylformamide
DMA dimethylacetamide
DIBAL-H diisobutylaluminum hydride
DIEPA N, N-diisopropylethylamine
EDCI 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride
EtOAc/EA ethyl acetate
Et3N triethylamine
HATU 2- (7-azabenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate
HBTU O-benzotriazol-tetramethylurea hexafluorophosphate
HOAc acetic acid
HOBt 1-hydroxybenzotriazole
ee enantiomer excess
mL of
g
mg
ng Nake
mol
mmol millimoles
h hours
MeOH methanol
NaH sodium hydride
NCS N-chlorosuccinimide
NMP N-methyl-2-pyrrolidone
PE Petroleum ether
Pd(dppf) 2 Cl 2 [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
Pd 2 (dba) 3 Tridibenzylidene acetone dipalladium
Pd(PPh 3 ) 4 Tetrakis (triphenylphosphine) palladium
PMB p-methoxycyanobenzyl group
PPh 3 Triphenylphosphine and process for preparing same
THF tetrahydrofuran
TFA trifluoroacetic acid
TFE trifluoroethanol
TsOH 4-toluene sulfonic acid
Xphos 2-dicyclohexylphosphine-2 ',4',6' -triisopropylbiphenyl
Example 1: synthesis of Compounds 1-7
Compound 4: (S-2, 4-diamino-6- (1- (5-phenylpyrazol [1,5-a ] pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile
Figure BDA0003412931580000301
The method comprises the following specific steps:
a) 3-Aminopyrazole was dissolved (1.43 g, 17.2 mmol) in N, N-dimethylformamide (17 ml), and diethyl ethoxymethylene malonate (4.65 g, 21.5 mmol, 1.25 eq.) K was added sequentially 2 CO 3 (5.94 g, 40.3 mmol, 2.5 eq.). The reaction solution was stirred at 120-125℃for 8 hours under nitrogen protection. Cooling to room temperature, quenching with ice water, regulating pH to 1-2 with 6N hydrochloric acid, separating out solid, filtering, washing with water, and air drying for 2 days to obtain 5-hydroxypyrazole [1,5-a ] ]Pyrimidine-6-carboxylic acid ethyl ester (2.8 g, 78% yield) LC-MS (M/z): 208.1 (M) + +1)。
b) At room temperature, 5-hydroxypyrazole [1,5-a ] is added]Pyrimidine-6-ethyl ester was dissolved (10 g, 48.3 mmol) in acetonitrile (50 ml) followed by the addition of phosphorus oxychloride (30 g, 193.2 mmol, 4 eq.) in sequence, N-diisopropylethylamine (12.5 g, 96.6 mmol, 2 eq.). The reaction solution was heated to 120-125℃and stirred for a further 7 hours. Cooled to room temperature and poured into ice water (300 ml), extracted with ethyl acetate (3×350 ml), the organic phases combined, anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain yellow solid 5-chloropyrazolo [1,5-a ]]Pyrimidine-6-carboxylic acid ethyl ester (6.91 g, 61% yield) LC-MS (M/z): 226.1 (M) + +1). The product was used directly in the next reaction without purification.
c) Sequentially adding 5-chloropyrazolo [1,5-a ] into a tube sealing]Pyrimidine-6-ethyl ester (1.95 g, 8.7 mmol), phenylboronic acid (1.27 g, 10.4 mmol, 1.2 eq.) and Pd (dppf) 2 Cl 2 (318 mg, 0.44 mmol, 0.05 eq.) Na 2 CO 3 (1.1 g, 10.4 mmol, 1.2 eq.) magnetic stirrer, dioxane (29 ml) and water (11 ml). And (3) after the air in the sealing tube is exhausted through three times of vacuum air extraction and nitrogen gas flushing, closing the sealing tube. The tube was sealed and stirred at 90-95℃for 4 hours. Cooled to room temperature, added to water (30 ml), extracted with ethyl acetate (3 x 60 ml), the organic phases combined, dried over anhydrous Na2SO4, filtered and concentrated to give the initial product. Purifying the primary product by flash column chromatography to obtain yellow solid 5-phenylpyrazole [1,5-a ] ]Pyrimidine-6-carboxylic acid ethyl ester (1.61 g, yield)69%)。LC-MS:268.1(M + +1)。
d) 5-phenylpyrazol [1,5-a ] pyrimidine-6-ethyl ester (2.41 g, 9.03 mmol) was dissolved in water/tetrahydrofuran/methanol (20 ml/10 ml) at 0deg.C, then solid LiOH H2O (947 mg, 22.6 mmol, 2.5 eq.) was added in one portion. Stirring overnight at room temperature, then adjusting to pH 2-3 with 1N hydrochloric acid, extracting with ethyl acetate, combining the organic phases, drying over anhydrous Na2SO4, filtering and concentrating to give 5-phenylpyrazol [1,5-a ] pyrimidine-6-carboxylic acid as a white solid (2.51 g, yield > 100%). The product was used directly in the next reaction without purification.
e) At 0 ℃, 5-phenylpyrazole [1,5-a ]]Pyrimidine-6-carboxylic acid (2.16 g, 9.03 mmol) was dissolved in dichloromethane (30 ml) and then N, O-dimethylhydroxylamine hydrochloride (1.77 g, 18 mmol, 2 eq.) EDCI (2.72 g, 14.2 mmol, 1.57 eq.), HOBt (2.1 g, 13.6 mmol, 1.5 eq.) and Et were added sequentially 3 N (2.74 g, 27.1 mmol, 3 eq.). The reaction was stirred overnight at room temperature, quenched with ice water (50 ml), extracted with ethyl acetate (3×60 ml), the organic phases combined, dried over anhydrous Na2SO4, filtered and concentrated to give the initial product. Purifying the primary product by flash column chromatography to obtain yellow solid N-methoxy-N-methyl 5-phenylpyrazole [1,5-a ] ]Pyrimidine-6-carboxamide (960 mg, 38% yield). LC-MS 283.1 (M + +1)。
f) At 0 ℃, N-methoxy-N-methyl 5-phenylpyrazole [1,5-a ]]Pyrimidine-6-carboxamide (730 mg, 2.59 mmol) was dissolved in THF (10 ml) and then methyl grignard reagent MeMgBr (1.9 ml, 3.0m,5.7 mmol, 2.2 eq) was added dropwise. Stirring at room temperature for 4 hours, quenching the reaction with saturated aqueous ammonia chloride (15 ml), extraction with ethyl acetate (3×30 ml), combining the organic phases, anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain a primary product. Purifying the primary product by flash column chromatography to obtain yellow solid 1- (5-phenylpyrazole [1, 5-a)]Pyrimidin-6-yl) -ethyl ketone (390 mg, 63% yield). LC-MS 238.1 (M + +1)。
g) 1- (5-phenylpyrazole [1,5-a ] at room temperature]Pyrimidine-6) -ethyl ketone (350 mg, 1.47 mmol) was dissolved in THF (10 ml), then (R) -2-methylpropane-2-sulfinyl (196 mg, 1.62 mmol, 1.1 eq.) was added sequentially),Ti(OEt) 4 (675 mg, 2.96 mmol, 2 eq.). Reflux for 6 h, cool to room temperature, quench the reaction with saturated aqueous ammonia chloride (10 ml), extract with ethyl acetate (3×20 ml), combine the organic phases, anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain a primary product. Purifying the primary product by flash column chromatography to obtain yellow solid (R, Z) -2-methyl-N- (1- (5-phenylpyrazole [1, 5-a)) ]Pyrimidin-6-yl) vinyl) -2-methylpropan-2-sulfinamide (301 mg, 62% yield). LC-MS 341.1 (M) + +1)。
h) (R, Z) -2-methyl-N- (1- (5-phenylpyrazole [1, 5-a)) at room temperature]Pyrimidin-6-yl) vinyl) -2-methylpropan-2-sulfinamide (340 mg, 1 mmol) was dissolved in THF (8 ml), cooled to-78 ℃, and L-selectride (2.5 ml, 1.0m in THF,2.5 mmol, 2.5 eq.) was added dropwise. After stirring at-78 ℃ for 2 hours, the reaction was quenched with saturated aqueous ammonia chloride (15 ml), extracted with ethyl acetate (2 x20 ml), the organic phases combined, anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain a primary product. Purifying the primary product by flash column chromatography to obtain yellow solid (R) -2-methyl-N- ((S) -1- (5-phenylpyrazole [1, 5-a)]Pyrimidin-6-yl) ethyl) -2-methylpropan-2-sulfinamide (197 mg, 58% yield). LC-MS 343.1 (M) + +1)。
i) (R) -2-methyl-N- ((S) -1- (5-phenylpyrazole [1, 5-a) at 0deg.C]Pyrimidin-6-yl) ethyl) -2-methylpropan-2-sulfinamide (200 mg, 0.58 mmol) was dissolved in dichloromethane (3 ml) and then a solution of 4N HCl in dioxane (0.58 ml, 2.34 mmol, 4 eq.) was added dropwise. Stirring at room temperature for 2 hr, concentrating to obtain (S) -1- (5-phenylpyrazole [1, 5-a)]Pyrimidin-6-yl) -ethylamine hydrochloride. This salt was dissolved in nBuOH (3 ml) and then 2, 4-diamino-6-chloropyrimidine-5-carbonitrile (149 mg, 0.88 mmol, 1.5 eq.) was added followed by DIEPA (302 mg, 2.34 mmol, 4 eq.). Heating and refluxing for 16 hours, cooling to room temperature, and concentrating to obtain a primary product. Purifying the primary product by flash column chromatography to obtain yellow solid (S) -2, 4-diamino-6- (1- (5-phenylpyrazole [1, 5-a) ]Pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile 4 (59 mg, yield 27%). LC-MS 372.2 (M) + +1)。
Compound 1: (S) -2, 4-diamino-6- (1- (2-methyl-5-phenylpyrazol [1,5-a ] pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile
Figure BDA0003412931580000331
a) Ethyl benzoylformate (32.9 g, 171 mmol), N-dimethylformamide dimethyl acetal (24.5 g, 205.2 mmol, 1.2 eq.) and toluene (350 ml) were added to a 1000 ml round bottom flask. Reflux was performed for 2 hours, cooled to room temperature, and toluene was removed by steaming under reduced pressure. The residue was dissolved in 350 ml of ethanol, and 5-methyl-3-aminopyrazole (16.6 g, 171 mmol, 1 eq.) and glacial acetic acid (100 ml) were added sequentially. Stirring at room temperature for 24 hours, removing ethanol by distillation under reduced pressure, adjusting the pH of the residue to 7-8 with saturated NaHCO3, extracting with ethyl acetate (3X 200 ml), combining the organic phases, washing with saturated brine, anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain a primary product. Purifying the primary product by flash column chromatography to obtain white solid 2-methyl-5-phenylpyrazole [1,5-a ]]Pyrimidine-6-carboxylic acid ethyl ester (27.8 g, 58% yield.) LC-MS:282.2 (M) + +1)。
b) Other reaction steps: (S) -2, 4-diamino-6- (1- (2-methyl-5-phenylpyrazol [1,5-a ] pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile 1 may be obtained in a similar manner as compound 4.
The compound 2 (S) -N- (1- (2-methyl-5-phenylpyrazolo [1,5-a ] pyrimidin-6-yl) ethyl) -9H-purin-6-amine and the compound 3 (S) -4-amino-6- (1- (2-methyl-5-phenylpyrazolo [1,5-a ] pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile may be synthesized from (S) -1- (2-methyl-5-phenylpyrazolo [1,5-a ] pyrimidin-6-yl) ethylamino in a manner analogous to that of compound 1.
Figure BDA0003412931580000341
Compound 5: (S) -2, 4-diamino-6- (1- (3-chloro-2-methyl-5-phenylpyrazol [1,5-a ] pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile
Figure BDA0003412931580000342
a) 1- (2-methyl-5-phenylpyrazole [1,5-a ] at room temperature]Pyrimidine-6) -ethyl ketone (3.22 g, 12.83 mmol) was dissolved in CHCl 3 To (40 ml) was then added NCS (2.06 g, 15.4 mmol, 1.2 eq.) and heated to reflux for 7 hours, cooled to room temperature, silica gel was added and the concentrated concentrate was purified by flash column chromatography to give 1- (3-chloro-2-methyl-5-phenylpyrazole [1,5-a ] as a yellow solid]Pyrimidin-6-yl) ethanone (2.41 g, 64% yield).
b) 1- (3-chloro-2-methyl-5-phenylpyrazole [1,5-a ] at room temperature]Pyrimidin-6-yl) ethanone (2.45 g, 9.8 mmol) was dissolved in THF (40 ml) and then (R) -2-methylpropan-2-sulfinyl (1.89 g, 15.6 mmol, 1.6 eq.) Ti (OEt) was added sequentially 4 (4.45 g, 19.5 mmol, 2 eq.) and reflux for a further 20 hours, then cooled to room temperature, quenched with saturated aqueous chlorinated ammonia (50 ml), extracted with ethyl acetate (3×50 ml), the organic phases combined, anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain a primary product. Purifying the primary product by flash column chromatography to obtain yellow solid (R, Z) -N- (1- (3-chloro-2-methyl-5-phenylpyrazole [1,5-a ])]Pyrimidin-6-yl) vinyl) -2-methylpropan-2-sulfinamide (2.2 g, 71% yield). LC-MS 389.1 (M) + +1)。
c) (R, Z) -N- (1- (3-chloro-2-methyl-5-phenylpyrazole [1,5-a ]) was prepared at room temperature]Pyrimidin-6-yl) vinyl) -2-methylpropan-2-sulfinamide (2.04 g, 5.76 mmol) was dissolved in THF (20 ml), cooled to-78 ℃, and L-selectride (14.4 ml, 1.0m in THF,14.4 mmol, 2.5 eq) was added dropwise. After stirring at-78 ℃ for 2 hours, the reaction was quenched with saturated aqueous ammonia chloride (50 ml), extracted with ethyl acetate (3×40 ml), the organic phases combined with anhydrous Na 2 SO 4 Drying, filtering and concentrating to obtain a primary product. Purifying the primary product by flash column chromatography to obtain yellow solid (R) -N- ((S) -1- (3-chloro-2-methyl-5-phenylpyrazole [1, 5-a)]Pyrimidin-6-yl) ethyl) -2-methylpropan-2-sulfinamide (1.97 g, 96% yield). LC-MS 391.1 (M + +1)。
d) (R) -N- ((S) -1- (3-chloro-2-methyl-5-phenylpyrazole [1, 5-a) at 0deg.C]Pyrimidin-6-yl) ethyl) -2-methylpropan-2-sulfinamide (530 mg, 1.41 mmol)Dichloromethane (10 ml) was dissolved and then a solution of 4N HCl in dioxane (1.41 ml, 5.64 mmol, 4 eq.) was added dropwise. Stirring at room temperature for 2 hr, and concentrating to obtain (S) -1- (3-chloro-2-methyl-5-phenylpyrazole [1, 5-a) ]Pyrimidin-6-yl) -ethylamine hydrochloride. The salt was dissolved in nBuOH (10 ml) and 2, 4-diamino-6-chloropyrimidine-5-carbonitrile (359 mg, 2.12 mmol, 1.5 eq.) and DIEPA (729 mg, 5.64 mmol, 4 eq.) were added sequentially. Reflux for 14 h, cool to room temperature, dilute with water, extract with ethyl acetate (3×40 ml), combine the organic phases, dry over anhydrous Na2SO4, filter and concentrate to give the initial product. Purifying the primary product by flash column chromatography to obtain yellow solid (S) -2, 4-diamino-6- (1- (3-chloro-2-methyl-5-phenylpyrazole [1, 5-a)]Pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile (270 mg, 49% yield). LC-MS 420.14 (M) + +1)。
Compound 6 (S) -4-amino-6- (1- (3-chloro-2-methyl-5-phenylpyrazol [1,5-a ] pyrimidin-6-yl) ethylamino) pyrimidine-5-carbonitrile and compound 7
(S) -N- (1- (3-chloro-2-methyl-5-phenylpyrazolo [1,5-a ] pyrimidin-6-yl) ethyl) -9H-purin-6-amine
Can be synthesized from (S) -1- (3-chloro-2-methyl-5-phenylpyrazolo [1,5-a ] pyrimidin-6-yl) ethylamino in a similar manner to the synthesis of compound 5.
TABLE 1 summary of spectral data for the synthesis of example compounds
Figure BDA0003412931580000351
/>
Figure BDA0003412931580000361
Example 2: determination of PI3K enzyme Activity by fluorescence method
PI3K kinases include PIK3CG (p 110 gamma) purchased from Invitrogen, p110α/p85α, p110β/p85α and p110δ/p85α purchased from BPS.
ADP-Glo TM Kinase Assay(ADP-Glo TM Kinase assay kit) (Promega, v 9102) is a luminescent kinase assay kit that detects ADP formed in the kinase reaction;ADP is converted to ATP, which is then subjected to Ultra-Glo TM Luciferase is converted into a light signal. The luminescent signal is positively correlated with kinase activity. The kit is suitable for detecting the influence of compounds on the activities of various purified kinases, and makes the kit an ideal tool for primary screening and researching the selectivity of kinase action. The operation of the kit comprises two steps: first, after the kinase reaction, an equivalent volume of ADP-Glo as the kinase reaction system was added thereto TM Reagents stop the reaction and consume the remaining ATP. In the second step, a kinase detection reagent is added which, while converting ADP to ATP, also uses a coupled luciferase/luciferin reaction to detect newly synthesized ATP.
The detection process is as follows:
the kinase buffer comprises 50mM HEPES,pH 7.5;3mM MgCl2;1mM EGTA;100mM NaCl;0.03% chaps;2mM DTT.
1) To 384 assay plates (Corning, cat# 4512) 1 μl of compound dilution was added. ( The final concentrations of compound detection were 10 concentrations of 1000nM, 333.33nM, 111.11nM, 37.037nM, 12.345nM, 4.115nM, 1.372nM, 0.457nM, 0.1524nM and 0.0508nM, multiplexing wells. DMSO final concentration is 1% )
2) 2. Mu.l of kinase solution (final concentration of PI 3K. Alpha. 0.25. Mu.g/ml; the final concentration of PI3K beta is 0.3 mug/ml; the final concentration of PI3K gamma is 1.2 mug/ml; the final concentration of PI3K delta was 0.3. Mu.g/ml).
3) 2. Mu.l of substrate solution (PI3Kα: PIP2 final concentration 20. Mu.M, ATP final concentration 10. Mu.M) was added to the corresponding position of 384 assay plates; PI3K gamma, beta and delta PIP2 final concentration 30. Mu.M, ATP final concentration 20. Mu.M). The reaction was carried out at room temperature for 120 minutes.
4) The ADP-Glo reagent was equilibrated to room temperature. The assay plate was quenched by adding 5. Mu.l of ADP-Glo reagent per well. Centrifuge 1000 rpm, centrifuge 1 min. Then slowly shake on a vibrating screen and equilibrate for 40 minutes at room temperature.
5) Mu.l of kinase assay reagent was added to each well, centrifuged for 1 min, and after 30 min equilibration an Envision reading was used.
Data analysis: the detection value is copied from Envision and the detection value data is converted into a percentage suppression value. Inhibition ratio = (Max-Sample)/(Max-Min) 100. Fitting the data in XLfit excel plug-in version yields IC50 values. (Max: the reaction system contains kinase and substrate; min: the reaction system contains only substrate; sample: the reaction system contains kinase, substrate and compound).
Table 2 shows the IC50 (nM) values of some of the compounds of the invention measured in this example
Figure BDA0003412931580000371
From the above table it can be seen that some of the compounds of the invention showed activity on the order of nM against PI3K delta kinase in vitro kinase assays, and that some compounds were over 200-fold selective for other subtypes.
Example 3: compounds were evaluated for their inhibitor effects on pi3kδ and pi3kγ, respectively, in human lymphoma cell lines RAJI and in the mouse megaphaga-like cell raw264.7 cell line
This example is an assessment of the effect of compounds on different isoforms of PI3K at the cellular level by detecting AKT phosphorylation. To assess the effect of compounds on the PI 3K-delta subtype, the human lymphoma cell line RAJI needs to bind to anti-IgM antibodies. The inhibitory effect of the compounds on PI 3K-gamma was evaluated and the giant phagocyte raw264.7 of mice required stimulation of the G protein-coupled receptor agonist C5 a.
PI 3K-gamma subtype: RAW264.7 cells were resuspended in serum-free DMEM medium and 60000/45. Mu.L of cell suspension was added to each well in 96-well plates. Cells were exposed to 5% CO 2 Incubate overnight at 37 ℃. After 18 hours of serum-free starvation, 50. Mu.L of the compound was added and incubated in an incubator for 60min. Then 5. Mu.L of 25nM C5a (R&Dssystems, diluted with serum medium) for 5min. mu.L of 1 Xlysate was added to each well of the aspirated medium. Shake for 45 min at room temperature. mu.L of lysate was transferred to 384 plates and 4. Mu.L of premixed antibody from Cisbio's Phospho-AKT (Ser 473) kit was added. Centrifugation at 1000rpm/min for 1 min, incubation at 22℃for 4 hours followed by Envision reading (665 nm/615 nm).
PI 3K-delta subtype: raji cells were cultured in 96-well plates at 30 μl per well, 50,000 cell linesThe cell culture medium was RPMI-1640 without serum. Cells at 5% CO 2 And incubated overnight in an incubator at 37 ℃. After 18 hours of serum-free starvation, 2s.5 μl of compound (14X) was added to the cells and incubated in the 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 stimulation for 30 minutes (final concentration 10. Mu.g/mL). 11.5. Mu.L of 4 Xlysate was added to each well. Shake for 45 min at room temperature. mu.L of lysate was added to 384 plates, and 4. Mu.L of premixed antibody from Cisbio's Phospho-AKT (Ser 473) kit was added. Centrifugation at 1000rpm/min for 1 min was performed and after incubation at 22℃for 4 hours, an Envision reading (665 nm/615 nm) was performed.
Table 3 shows the IC50 (nM) values of some of the compounds of the invention measured in this example
Figure BDA0003412931580000381
From the above table, it can be seen that some of the compounds of the invention showed activity on the order of nM against PI3K delta kinase in vitro cell assays, and that some compounds were over 100-fold selective for other subtypes.
It will be appreciated that, after reading the above disclosure of the present invention, those skilled in the art may make various modifications, adaptations, or combinations of the technical solutions of the present invention without departing from the spirit and scope of the aspects of the present application, and such equivalent modifications or combinations are also within the scope of the present application.

Claims (23)

1. A compound represented by the formula (I) and/or a pharmaceutically acceptable salt thereof, characterized in that,
Figure FDA0003412931570000011
wherein:
R 1 independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, mercapto, amino, -C 1-6 Alkyl, -C 1-6 Alkoxy, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocyclylalkyl, -C 3-6 Cycloalkoxy, -C 3-6 Heterocyclyloxy, -C 1-6 Alkyl (O) C 1-6 Alkyl, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR 1a Aryl, aralkyl, aralkoxy, heteroaryl, heteroaralkyl, heteroaralkoxy; wherein said mercapto, amino, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkoxy, heterocycloalkyl, heterocycloalkoxy, aryl, aralkyl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Optionally substituted by one or more R 1a And (3) substitution.
R 1a Selected from hydrogen, deuterium, halogen, cyano, hydroxy, mercapto, amino, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkoxy, heterocycloalkyl, heterocycloalkoxy, aryl, heteroaryl, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, heteroaryl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Selectively by halogen, cyano, -OR a 、-SR a 、NR b R c Any one or more of alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocyclyloxy, substituted or unsubstituted;
R 2 selected from deuterium, hydroxy, -C 1-6 Alkyl, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocyclylalkyl, -C 3-6 Cycloalkoxy, -C 3-6 Heterocyclyloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) NH (C) 1-6 Alkyl group 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 、-C(O)OR 2a 、-C(O)NH 2 Aryl, heteroaryl; wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, heteroaryl, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Can be optionally substituted with one or more R 2a Substitution;
R 2a selected from the group consisting of hydrogen, deuterium, halogen, cyano, hydroxy, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a The method comprises the steps of carrying out a first treatment on the surface of the Wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -C (O) NH 2 、-S(O)2NH 2 、-S(O)(NH)NH 2 Is optionally selected from halogen, cyano, -OR a 、-SR a 、NR b R c One or more of alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkoxy, heterocyclyloxy, substituted or unsubstituted;
And/or R 1 And R is R 2 Optionally forming 3-6 membered cycloalkyl, 3-6 membered heterocycloalkyl, 3-6 membered cycloalkenyl, 3-6 membered heterocycloalkenyl, aryl, heteroaryl with the carbon atom to which each is attached, and optionally substituted with one or more R 1a Or R is 2a Substitution; and the carbon atom or sulfur atom in the cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl may be selectively oxo to form a cyclic carbonyl group, an amide group, an ester group, a sulfone group (-S (O)) 2 (-) sulfonamide (-S (O)) 2 A portion of NH-).
R 3 Selected from hydrogen, deuterium, C 1-6 Alkyl, deuterated alkyl, halogenated C 1-6 Alkyl, hydroxy substituted C 1-6 Alkyl, cyanoalkyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocycloalkyl, alkoxyalkyl, cycloalkylAlkyl, heterocyclylalkyl, cycloalkoxyalkyl, heterocycloalkoxyalkyl;
R 4 independently selected from heteroaryl groups, said heteroaryl groups being monocyclic aromatic hydrocarbon groups having 5, 6 or 7 ring atoms which contain 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or bicyclic aromatic hydrocarbon groups having 9 or 10 ring atoms which contain 1, 2, 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring, and said heteroaryl groups include those heteroaryl groups in which the N ring heteroatoms are in the form of an N-oxide, and said heteroaryl groups are optionally substituted with one or more substituents selected from the group consisting of: halogen, deuterium, cyano, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, cycloalkoxy, heterocyclyl, heterocyclyloxy, -NH 2 、-NH(C 1-6 Alkyl), -N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -COOH, -C (O) NH 2 -C (O) NH (C1-6 alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a 、-NHC(O)(C 1-6 Alkyl), -NHC (O) (C) 1-6 Cycloalkyl), -NHC (O) O (C) 1-6 Alkyl), -NHC (O) O (C) 1-6 Cycloalkyl), phenyl, and 5-or 6-membered heteroaryl; wherein C is 1-6 Alkyl, haloalkyl, -NH 2 、-COOH、-C(O)NH 2 Cycloalkyl, heterocycloalkyl, optionally substituted with one or more R 1a Optionally substituted; wherein R is as R 4 The 5 or 6 membered heteroaryl group of the substituents of (a) is a monocyclic aromatic hydrocarbon group having 5 or 6 ring atoms which contains 1, 2 or 3 ring heteroatoms independently selected from N, O or S in the ring, the remaining ring atoms being carbon atoms, the S and O heteroatoms being non-adjacent to each other when the total number of S and O atoms in the heteroaryl group exceeds 1, wherein the phenyl or 5 or 6 membered heteroaryl group as substituents of the heteroaryl group is optionally substituted with one or more substituents selected from: halogen, amino, hydroxy, mercapto, C1-6 alkyl and-O (C1-6 alkyl), haloalkyl;
R 5 independently selected from C 1-6 Alkyl, hydroxy-substituted alkyl, deuterated alkyl, hydroxyalkyl, C 3-6 NaphtheneRadical, C 3-6 Heterocycloalkyl, cycloalkylalkyl or cycloalkoxyalkyl, wherein said alkyl is optionally substituted with one or more R 5a Substitution; r is R 5a Selected from deuterium, halogen, CN, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl; wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl groups may be substituted with halogen, CN, -OR a 、-SR a 、-NR b R c Alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl substitution;
and/or R 4 And R is 5 Optionally together with the N atom to which they are attached, form a nitrogen-containing heterocyclic group or heteroaryl group, wherein the nitrogen-containing heterocyclic group and heteroaryl group may be optionally substituted with one or more substituents including, but not limited to, halogen, deuterium, amino, hydroxy, mercapto, cyano, alkyl, deuteroalkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, heterocycloalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, -COOH, -C (O) NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), -S (O) 2NH 2 、-S(O)(NH)NH 2 、-C(O)OR a 、-OR a 、-SR a 、NR b R c
Wherein R is a 、R b And R is c Independently selected from hydrogen, deuterium, halogen, C 1-6 Alkyl, deuterated C 1-6 Alkyl, halogenated C 1-6 Alkyl, -C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 3-6 Cycloalkyl, -C 3-6 Heterocycloalkyl, hydroxy-substituted C 1-6 Alkyl, -C 1-6 Alkyl (O) C 1-6 Alkyl, aryl, heteroaryl;
and/or R b And R is c Together with the nitrogen atom to which they are both attached form a 4-to 7-membered heterocycloalkyl or are substituted with 1 to 3R a And (3) substitution.
2. A compound of formula (I) according to claim 1 and/or a pharmaceutically acceptable salt thereof, wherein the aryl group is a carbocyclic hydrocarbon group containing 6-12 ring carbon atoms consisting of one ring or multiple ring fusions, wherein at least one ring is an aromatic ring and the other rings are not heteroaryl;
the heteroaryl is a monocyclic aromatic hydrocarbon group having 5, 6 or 7 ring atoms which contains 1 or 2 ring heteroatoms independently selected from N, O and S in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 or 10 ring atoms which contains 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring, when the total number of S and O atoms on the heterocycle exceeds 1, the S and O atoms are not adjacent to each other, and the heteroaryl includes those heteroaryl groups in which the N heteroatom is in the form of an N-oxide;
the aryl and heteroaryl groups are optionally substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, hydroxy, mercapto, -C 1-6 Alkyl, haloalkyl, alkoxy, haloalkoxy, haloalkoxyalkyl, cycloalkyl, halocycloalkyl, - (C) 1-6 Alkyl) OH and-SO 2 (C 1-6 Alkyl), alkenyl, alkynyl.
3. A compound of formula (I) according to claim 1 or 2 and/or a pharmaceutically acceptable salt thereof, wherein the aryl or heteroaryl group is selected from aryl and heteroaryl groups having 6 ring atoms, heteroaryl groups having 5 ring atoms, bicyclic heteroaryl groups having 9 or 10 ring atoms or aryl groups;
wherein the aryl and/or heteroaryl group comprises phenyl, deuterated phenyl, naphthyl, pyridinyl, pyrimidine, pyridazine, piperazine, pyrazolyl, imidazolyl, quinolinyl, thienyl, benzothienyl, indolyl, optionally substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, hydroxy, -C 1-6 Alkyl, - (C) 1-6 Alkyl) OH, C 1-6 Haloalkyl and-SO 2 (C) 1-6 Alkyl).
4. A compound of formula (I) according to claim 1 and/or a medicament thereofA pharmaceutically acceptable salt, characterized in that R 4 Selected from heteroaryl, or R 4 And R is R 5 Forming a heteroaryl group;
wherein the heteroaryl group is a monocyclic aromatic hydrocarbon group having 6 ring atoms which contains 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 ring atoms which contains 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring and the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of: halogen, cyano, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl) -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-C(O)NH 2 、-C(O)NH(C 1-6 Alkyl), -C (O) N (C) 1-6 Alkyl) (C) 1-6 Alkyl), phenyl, and 5-or 6-membered heteroaryl.
5. A compound of formula (I) according to claim 1 and/or a pharmaceutically acceptable salt thereof, wherein R 4 Selected from heteroaryl, or R 4 And R is R 5 Forming a heteroaryl group;
wherein the heteroaryl group is a monocyclic aromatic hydrocarbon group having 6 ring atoms which contains 1 or 2N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, or a bicyclic aromatic hydrocarbon group having 9 ring atoms which contains 3 or 4N ring heteroatoms in the ring, the remaining ring atoms being carbon atoms, wherein at least one ring is an aromatic ring and the heteroaryl group is optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, hydroxy, mercapto, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-C(O)NH 2 、-C(O)NH(C 1-6 Alkyl), phenyl, pyridyl, pyrimidinyl, piperazinyl, pyridazinyl, pyrazolyl, imidazolyl, triazolyl, and tetrazolyl.
6. A compound of formula (I) according to claim 5 and/or a pharmaceutically acceptable salt thereof, wherein R 4 Optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, mercapto, amino, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl).
7. The compound of formula (I) according to claim 5 and/or a pharmaceutically acceptable salt thereof, wherein the aryl group in the heteroaryl group is selected from pyrimidine, purinyl, pyrazolopyrimidine, pyrrolopyrimidine.
8. A compound of formula (I) and/or a pharmaceutically acceptable salt thereof according to claim 4 or 5,
R 4 or R is 4 And R is R 5 The heteroaryl group formed is selected from the substituents shown below
Figure FDA0003412931570000041
Wherein the substituents are optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, hydroxy, mercapto, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), N (C) 1-6 Alkyl) (C) 1-6 Alkyl) -CO 2 H、-CONH 2 、-C(O)NH(C 1-6 Alkyl), phenyl, pyridyl, pyrazolyl, imidazolyl, oxazolyl, triazolyl and tetrazolyl, wherein R is 4 Phenyl, pyridinyl, pyrimidinyl, piperazinyl, pyridazinyl, pyrazolyl, imidazolyl, triazolyl and tetrazolyl of the substituents are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, mercapto, C 1-6 Alkyl, haloalkyl, cycloalkyl, -O (C) 1-6 Alkyl) and-O (C) 1-6 Haloalkyl).
9. A compound of formula (I) according to claim 8 and/or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
Figure FDA0003412931570000051
Optionally substituted with one or more substituents selected from the group consisting of: fluorine, chlorine, bromine, cyano, haloalkyl, C 1-6 Alkyl, -NH 2 、NH(C 1-6 Alkyl), -CO2H, -CONH 2 Phenyl, pyridyl, pyrazolyl, triazolyl and tetrazolyl,
wherein R is as R 4 Phenyl, pyridinyl, pyrazolyl, imidazolyl, triazolyl and tetrazolyl of the substituents are optionally substituted with one or more substituents selected from the group consisting of: halogen, hydroxy, C 1-6 Alkyl and-O (C) 1-6 Alkyl).
10. A compound of formula (I) according to any one of claims 1 to 9 and/or a pharmaceutically acceptable salt thereof, wherein R 3 Is C 1-4 Alkyl and C 1-4 A haloalkyl group.
11. A compound of formula (I) according to any one of claim 10 and/or a pharmaceutically acceptable salt thereof, wherein R 3 Methyl, ethyl and trifluoromethyl.
12. A compound of formula (I) according to any one of claims 1 to 10 and/or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) is selected from:
Figure FDA0003412931570000052
Figure FDA0003412931570000061
13. Use of a compound of formula (I), a pharmaceutically acceptable salt thereof, according to any one of claims 1-12, in the manufacture of a medicament for the treatment of a PI3K mediated related disease.
14. Use of a compound of formula (I) according to claim 13, wherein PI3K is pi3kδ, in the manufacture of a medicament for the treatment of a disease mediated by PI 3K.
15. Use of a compound of formula (I), a pharmaceutically acceptable salt thereof, according to claim 13, for the manufacture of a medicament for the treatment of PI3K mediated diseases selected from cancer, inflammation, respiratory diseases, infectious diseases, bone marrow related diseases and autoimmune diseases.
16. Use of a compound of formula (I), a pharmaceutically acceptable salt thereof, according to claim 15, in the manufacture of a medicament for the treatment of PI3K mediated diseases, wherein the cancer is a solid tumor including breast cancer, melanoma, head and neck cancer, lung cancer, thymus cancer and hematological malignancy.
17. A pharmaceutical composition comprising a compound of formula (I) as defined in any one of claims 1 to 12, a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier or excipient.
18. The pharmaceutical composition of claim 17, further comprising use in combination with a small molecule compound and/or a large molecule antibody to treat cancer, inflammation, bone marrow related diseases, and autoimmune diseases.
19. The pharmaceutical composition of claim 18, wherein the small molecule compound comprises a kinase inhibitor and other non-kinase inhibitors.
20. The pharmaceutical composition of claim 18, wherein the small molecule compound comprises a BTK inhibitor, a Bcl2 inhibitor.
21. The pharmaceutical composition of claim 18, wherein the macromolecular antibody comprises an anti-CD 20, anti-CTLA 4, anti-PD-1, anti-PD-L1 antibody.
22. Use of a composition according to claim 17 in the treatment of a PI3K mediated related disease, wherein the disease is selected from cancer, inflammation, respiratory disease, infectious disease, bone marrow related disease and autoimmune disease.
23. A solvate, racemic mixture, enantiomer, diastereomer, tautomer, or mixture comprising a racemic mixture of a compound of formula (I) according to any one of claims 1 to 16, wherein R is as defined in claim 1 1 、R 2 、R 3 、R 4 And R is 5 Each substituent group
Figure FDA0003412931570000071
CN202111536751.1A 2021-12-15 2021-12-15 Pyrazolopyrimidine compound and application thereof Pending CN116262752A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202111536751.1A CN116262752A (en) 2021-12-15 2021-12-15 Pyrazolopyrimidine compound and application thereof
PCT/CN2022/139044 WO2023109870A1 (en) 2021-12-15 2022-12-14 Pyrazolopyrimidine compound and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111536751.1A CN116262752A (en) 2021-12-15 2021-12-15 Pyrazolopyrimidine compound and application thereof

Publications (1)

Publication Number Publication Date
CN116262752A true CN116262752A (en) 2023-06-16

Family

ID=86722450

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111536751.1A Pending CN116262752A (en) 2021-12-15 2021-12-15 Pyrazolopyrimidine compound and application thereof

Country Status (2)

Country Link
CN (1) CN116262752A (en)
WO (1) WO2023109870A1 (en)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7449488B2 (en) * 2002-06-04 2008-11-11 Schering Corporation Pyrazolopyrimidines as protein kinase inhibitors
US7119200B2 (en) * 2002-09-04 2006-10-10 Schering Corporation Pyrazolopyrimidines as cyclin dependent kinase inhibitors
JP2010504933A (en) * 2006-09-29 2010-02-18 ノバルティス アーゲー Pyrazolopyrimidines as PI3K lipid kinase inhibitors
CN108017641B (en) * 2016-11-02 2021-01-05 深圳铂立健医药有限公司 Pyrazolopyrimidine compound as PI3K inhibitor and application thereof
CN113278022B (en) * 2020-07-07 2023-12-12 南京纳丁菲医药科技有限公司 Pyrazolopyrimidine compound, pharmaceutical composition and application thereof

Also Published As

Publication number Publication date
WO2023109870A1 (en) 2023-06-22

Similar Documents

Publication Publication Date Title
JP6427257B2 (en) Heterocyclylamines as PI3K inhibitors
JP6617130B2 (en) 4-Imidazopyridazin-1-yl-benzamide and 4-Imidazotriazin-1-yl-benzamide as BTK inhibitors
AU2014324595B2 (en) Substituted nicotinimide inhibitors of BTK and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
AU2014358792B2 (en) Pyrrolo[2,3-d]pyrimidinyl, pyrrolo[2,3-b]pyrazinyl and pyrrolo[2,3-d]pyridinyl acrylamides
JP5808869B2 (en) Bicyclic piperazine compounds
JP5766820B2 (en) Heterocyclic compounds as PI3 kinase inhibitors
CA2868156C (en) Substituted pyridopyrimidine compounds and their use as flt3 inhibitors
JP2023521321A (en) Allosteric chromenone inhibitors of phosphoinositide 3-kinase (PI3K) for the treatment of disease
TWI707855B (en) Novel imidazopyridazine compounds and their use
CA2796311A1 (en) Fused derivatives as pi3k.delta. inhibitors
WO2015191677A1 (en) Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
AU2013300106A1 (en) Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
CN105732637B (en) Heteroaromatic compounds and their use in medicine
BR112021010358A2 (en) Heteroaromatic derivatives for use as a regulator, method for preparing them and their use
TW201404779A (en) Novel heteroaryl and heterocycle compounds, compositions and methods
MX2012009059A (en) Pyrido[3,2-d]pyrimidine pi3k delta inhibitor compounds and methods of use.
JP7248256B2 (en) JAK Kinase Inhibitors, Preparation Methods Thereof, and Uses Thereof in the Pharmaceutical Field
TW201639845A (en) Novel heteroaryl and heterocycle compounds, compositions and methods
CN111032630B (en) Compound, pharmaceutical composition, application and application thereof
TW202140488A (en) Heteroaryl heterocyclic compounds and uses thereof
CA3173804A1 (en) Fused aza-heterocyclic amide compound and use thereof
WO2017202719A1 (en) New phenylpyrazolylacetamide compounds and derivatives as cdk8/cdk19 inhibitors
CN116262752A (en) Pyrazolopyrimidine compound and application thereof
CN116249696A (en) Pyrimidinone compounds and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication