WO2022028507A1 - 一种杂环类化合物、其制备方法及用途 - Google Patents

一种杂环类化合物、其制备方法及用途 Download PDF

Info

Publication number
WO2022028507A1
WO2022028507A1 PCT/CN2021/110751 CN2021110751W WO2022028507A1 WO 2022028507 A1 WO2022028507 A1 WO 2022028507A1 CN 2021110751 W CN2021110751 W CN 2021110751W WO 2022028507 A1 WO2022028507 A1 WO 2022028507A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
membered
group
reaction
ring
Prior art date
Application number
PCT/CN2021/110751
Other languages
English (en)
French (fr)
Inventor
栾林波
陈永凯
王朝东
Original Assignee
上海美悦生物科技发展有限公司
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 上海美悦生物科技发展有限公司 filed Critical 上海美悦生物科技发展有限公司
Priority to US18/040,966 priority Critical patent/US20230331710A1/en
Priority to EP21853576.3A priority patent/EP4194449A4/en
Publication of WO2022028507A1 publication Critical patent/WO2022028507A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • C07D211/44Oxygen atoms attached in position 4
    • C07D211/46Oxygen atoms attached in position 4 having a hydrogen atom as the second substituent in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/06Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present invention relates to a heterocyclic compound, its preparation method and use.
  • Complement is a class of soluble pattern recognition molecules in the immune system that can perform a variety of effector functions.
  • complement components exist in the form of inactive zymogens, and a variety of specific and non-specific immunological mechanisms decompose these inactive zymogens to generate active large and small fragments.
  • large fragments usually stay on the surface of pathogens or cells, lysing the latter or accelerating their clearance; small fragments leave the cell surface and mediate various inflammatory responses.
  • Activation of complement consists of two closely following processes, which form a cascade of complement activation. There are currently three known complement activation pathways: the classical pathway, the lectin pathway, and the alternative pathway.
  • the initiation mechanisms and activation sequences of the three complement activation pathways are different, they share a common terminal pathway.
  • the activation of the alternative pathway does not depend on the antigen-antibody complex.
  • C3b deposited on the cell surface binds to factor B and becomes a state that is easily decomposed by factor D in serum.
  • factor B is decomposed into Ba and Bb.
  • C3b and Bb then form a complex to become the C3 convertase C3bBb in the alternative pathway; in this process, complement factor B plays an early and central role in the alternative pathway activation of the complement cascade.
  • C3b is not only the product after C3 convertase decomposes C3, but also a component of the alternative pathway C3 convertase, thus forming a feedback amplification mechanism of the interaction between the classical pathway and the alternative pathway.
  • Current studies have found that a variety of diseases such as hematological, autoimmune, inflammatory and neurodegeneration are associated with abnormal complement system function.
  • Paroxysmal nocturnal hemoglobinuria is a chronic disease with persistent hemolysis caused by a non-malignant clonal disease caused by acquired somatic PIG-A gene mutation in one or several hematopoietic stem cells. Ultra-rare blood disorders (Medicine (Baltimore) 1997, 76(2):63-93). The disease course can be manifested by varying degrees of exacerbation of hemolysis (paroxysmal), chronic or recurrent acute intravascular hemolysis or subsequent venous/arterial thrombosis that can lead to progressive end-organ damage and death, and is a classic PNH with chronic vascular disease. Internal hemolysis, hemoglobinuria, and hemosiderinuria are the main manifestations, but most patients are often atypical, with insidious onset, protracted course, and varying severity of disease.
  • GPI-anchored proteins There are more than ten proteins on the surface of red blood cells that inhibit the activation of the complement pathway, all of which are anchored to their cell membranes by glycosylated phosphatidylinositol (GPI), collectively referred to as GPI-anchored proteins (AP). Under certain conditions, it mutates and produces glycosylphosphatidylinositol (GPI)-deficient PNH clones; then, due to some factors (now mostly considered to be immune factors), hematopoietic damage or hematopoietic failure occurs, and the PNH clones proliferate advantage over normal clones.
  • GPI glycosylated phosphatidylinositol
  • CD59 membrane attack complex (MAC) inhibitor CD59, which inhibit complement pathway activation, are involved in the pathogenesis of The mechanism, clinical manifestations, diagnosis and treatment are closely followed with PNH (Frontiers in Immunology 2019, 10, 1157).
  • CD59 can prevent the incorporation of C9 into the C5b-8 complex, and prevent the formation of membrane attack units, thereby inhibiting the terminal attack response of complement. It is currently believed that the typical manifestations of PNH-intravascular hemolysis and thrombosis are caused by CD59 deficiency.
  • IgAN is the most common form of primary glomerulonephritis. The disease is characterized by IgA deposition in the mesangial area on immunofluorescence; its clinical manifestations are variable, usually manifesting as recurrent microscopic or gross hematuria.
  • IgAN is related to congenital or acquired immune dysregulation. Due to the stimulatory effects of viruses, bacteria and food proteins on the respiratory tract or digestive tract, mucosal IgA1 synthesis increases, or immune complexes containing IgA1 are deposited in the mesangial area, and activate the alternative pathway of complement, causing glomerular damage. Human IgA molecules are divided into two subtypes, IgA1 and IgA2.
  • IgA1 is the main form of blood circulation in healthy individuals (about 85%), and it is also the main component of mesangial deposition in patients with IgAN.
  • IgA molecules can exist in both monomeric and multimeric forms.
  • the IgA1 molecule has a unique heavy chain hinge region between the first and second constant regions that serves as the domain of the attachment site for O-linked glycan groups.
  • IgA molecules deposited in the serum and mesangial region of IgAN patients are mainly glycosylation-deficient IgA1 (gd-IgA1). At present, it is believed that the initiation link of the pathogenesis of IgAN is the abnormal increase in the production of gd-IgA1.
  • IgAN patients More than 90% of IgAN patients are accompanied by deposition of complement C3 in the mesangial region. 75%-100% of IgAN patients have co-deposition of properdin, IgA and C3 in renal tissue, and 30%-90% of IgAN patients have co-deposition of complement factor H, IgA and C3 in renal tissue. In addition to deposition in renal tissue, some studies have also found that the levels of markers of the alternative complement pathway in the plasma of IgAN patients are also associated with IgAN activity (J Nephrol 2013, 26(4):708-715). One study confirmed that C3a in renal tissue and urine and C3a receptors in renal tissue were significantly associated with the activity and severity of renal damage (J clin Immunol 2014, 34(2):224-232).
  • IgA can activate the alternative complement pathway under in vitro conditions.
  • the abnormality of the IgA hinge region does not play a decisive role, and the formation of IgA multimers is the key link (Eur J Immunol 1987,17(3):321-326).
  • the deposition of complement C3 in the mesangial region of the glomerulus has become an auxiliary diagnostic marker for IgAN.
  • MN membranous nephropathy
  • C3G C3 glomerulonephritis
  • AMD age-related macular degeneration
  • GA geographic atrophy
  • aHUS atypical hemolytic uremic syndrome
  • HUS hemolytic uremic syndrome
  • NMO neuromyelitis
  • arthritis rheumatoid arthritis
  • hepatic inflammation hepatic inflammation
  • dermatomyositis hepatic inflammation
  • muscular atrophy Lateral sclerosis myasthenia gravis
  • MG myasthenia gravis
  • oligonucleotide drugs developed by IONIS Pharmaceuticals Inc. are specific for complement factor B (CFB). Sex inhibitors to treat, prevent or alleviate diseases associated with dysregulation of the alternative complement pathway (WO2015038939).
  • the small molecule complement factor B inhibitor developed by Novartis AG is used for the treatment of diseases such as age-related macular degeneration (AMD) (WO2013164802, WO2013192345, WO2014143638, WO2015009616, WO2015066241), and for the treatment of diseases such as C3G and IgAN (WO2019043609A1).
  • Small molecule complement factor B inhibitors developed by Achillion Pharmaceuticals Inc. are used to treat diseases such as age-related macular degeneration (AMD) (WO2018005552).
  • Inflammation and immune-related diseases are characterized by diversity and incurability; only eculizumab is marketed as a drug for PNH disease, but due to its price, it brings a heavy burden to patients; at the same time, many patients have used eculizumab.
  • Anemia persists after treatment with eculizumab, and many patients still require continuous blood transfusions; in addition, eculizumab is administered intravenously.
  • IgAN there is no specific treatment drug so far, such as IgAN.
  • IgAN there are unmet clinical needs in these areas that require the development of new small molecule drugs for medical treatment.
  • the technical problem to be solved by the present invention is to provide a heterocyclic compound, its preparation method and use in order to overcome the deficiency of the lack of small molecule compounds as complement factor B inhibitors in the prior art.
  • the heterocyclic compounds provided by the present invention can be used as complement factor B inhibitors, and can be used to prepare medicines for treating diseases related to the abnormal activation of the complement system or the normal function of the complement system; Therapeutic agents for the disease are expected to meet clinical needs.
  • the present invention solves the above technical problems through the following technical solutions.
  • the present invention provides a heterocyclic compound as shown in formula I or a pharmaceutically acceptable salt, isotope analog, and prodrug thereof, optionally in a pharmaceutically acceptable carrier;
  • W is O or C (R 7' R 7" );
  • R 7 , R 7′ and R 7′′ are independently hydrogen, hydroxy, halogen, C 1 -C 4 alkyl or C 1 -C 4 alkyl-O-;
  • R 6 is hydrogen, C 1 -C 4 alkyl or hydroxy C 1 -C 4 alkyl
  • R 4' and R 4 are independently hydrogen
  • n 0, 1 or 2;
  • R5 is Ring B is a phenyl group or a 6-membered heteroaryl group; in the 6-membered heteroaryl group, the number of heteroatoms is 1, 2 or 3, and the heteroatoms are selected from one or more of N, O and S ;
  • Z 1 is C(R 21 ) or N;
  • Z 2 is C(R 51 ) or N;
  • Ring A 1 is pyridyl; wherein, Z 3 is C(R 22 ) or N; Z 4 and Z 5 are independently C or N; (the rest are C as shown;)
  • R 21 , R 22 and R 51 are independently hydrogen
  • Ring A 2 is 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl or 5-6-membered heteroaryl, or, substituted by one or more R a1 : 5-6-membered heterocycloalkyl , 5-6 membered heterocycloalkenyl or 5-6 membered heteroaryl; the 5-6 membered heterocycloalkyl and the 5-6 membered heterocycloalkyl substituted by one or more R a1
  • the number of heteroatoms is 1, 2 or 3, the heteroatom is selected from
  • Ring A 3 is 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl, 6-membered heteroaryl or (the a-terminus represents a cyclic connection to a benzene ring), or, substituted by one or more R a2 : 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl, 6-membered heteroaryl or
  • a 3' is a 5-membered heteroaryl group; in the 5-membered heteroaryl group, the number of heteroatoms is 1 or 2, and the heteroatoms are selected from one or more of N, O and S; and Z 7 is N, O or S, and/or Z is CH, O or S ;
  • R 11 , R 31 , R 12 , R 32 , R 13 and R 33 are independently C 1 -C 4 alkyl, C 1 -C 4 alkyl-O- or 3-6 membered cycloalkyl;
  • Z 8 is CH or N;
  • R 14 is C 1 -C 4 alkyl-O-;
  • R 23 and R 24 are H;
  • R 34 is C 1 -C 4 alkyl or 3-6 membered cycloalkyl;
  • a carbon atom with "*" means that when it is a chiral carbon atom, it is in S configuration, R configuration or a mixture thereof.
  • W is C(R 7' R 7" ).
  • R 7' and R 7" are independently hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 alkyl-O-; for example, R 7 " is independently hydrogen, C 1 -C 4 alkyl -O- or halogen; R 7' is independently hydrogen or C 1 -C 4 alkyl.
  • R 7 is hydrogen
  • R b is H.
  • Z 1 is CH and Z 2 is N, or Z 1 is CH and Z 2 is CH, or Z 1 is N and Z 2 is CH.
  • Z 4 is N, or Z 5 is N.
  • Ring A 2 is a 5- to 6-membered heteroaryl group, eg, a 5-membered heteroaryl group.
  • Ring A 2 is 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl or 6-membered heteroaryl, or, substituted by one or more R a1 : 5-6-membered cycloalkenyl, 5- 6-membered heterocycloalkyl, 5-6 membered heterocycloalkenyl or 6-membered heteroaryl.
  • Ring A 3 is 5-6 membered heterocycloalkyl, 5-6 membered heterocycloalkenyl or Or, substituted by one or more R a2 : 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl, 6-membered heteroaryl or
  • 5-membered heterocycloalkyl For example 5-membered heterocycloalkyl, 5-membered heterocycloalkenyl or Or, substituted by one or more R a2 : 5-membered heterocycloalkyl, 5-membered heterocycloalkenyl or
  • R 11 , R 12 and R 13 are independently C 1 -C 4 alkyl or C 1 -C 4 alkyl-O-, preferably C 1 -C 4 alkyl-O-.
  • R 31 , R 32 and R 33 are independently C 1 -C 4 alkyl.
  • R 14 is C 1 -C 4 alkyl-O-.
  • Z 8 is N and R 34 is C 1 -C 4 alkyl; or Z 8 is CH and R 34 is 3-6 membered cycloalkyl.
  • heterocyclic compounds represented by formula I are defined as follows (unmentioned groups are the same as those described in any scheme of this application), the The heterocyclic compound shown in formula I is shown in the following formula Ia, Ib or Ic;
  • R 7" is independently hydrogen, C 1 -C 4 alkyl-O- or halogen;
  • R 7' is independently hydrogen or C 1 -C 4 alkyl;
  • W is C(R 7' R 7" ); R 7 is hydrogen;
  • R 7' and R 7" are independently hydrogen, halogen, C 1 -C 4 alkyl or C 1 -C 4 alkyl-O-; for example, R 7 " is independently hydrogen, C 1 -C 4 alkyl -O- or halogen; R 7' is independently hydrogen or C 1 -C 4 alkyl;
  • R 6 is hydrogen
  • R 4' and R 4 are independently hydrogen
  • n 1;
  • R5 is Ring B is phenyl or 6-membered heteroaryl
  • Z 1 is CH or N;
  • Z 2 is CH or N;
  • Ring A 1 is pyridyl;
  • Z 3 is CH or N;
  • Z 4 and Z 5 are independently C or N;
  • Ring A 2 is 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl or 5-6-membered heteroaryl, or, substituted by one or more R a1 : 5-6-membered heterocycloalkyl , 5-6 membered heterocyclenyl or 5-6 membered heteroaryl;
  • Ring A 3 is 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl, 6-membered heteroaryl or Alternatively, substituted with one or more R a2 : 5-6 membered heterocycloalkyl, 5-6 membered heterocycloalkenyl, 6 membered heteroaryl or
  • R 11 , R 31 , R 12 , R 32 , R 13 , R 23 and R 33 are independently C 1 -C 4 alkyl, C 1 -C 4 alkyl-O- or 3-6 membered cycloalkyl;
  • Z 8 is CH or N;
  • R 14 is C 1 -C 4 alkyl-O-;
  • R 23 and R 24 are H;
  • R 34 is C 1 -C 4 alkyl or 3-6 membered cycloalkyl;
  • a carbon atom with "*" means that when it is a chiral carbon atom, it is in S configuration, R configuration or a mixture thereof.
  • W is C(R 7' R 7" ); R 7 is hydrogen;
  • R 7' and R 7" are independently hydrogen or C 1 -C 4 alkyl-O-;
  • R 6 is hydrogen
  • R 4' and R 4 are independently hydrogen
  • n 1;
  • Ring A 1 is pyridyl; Z 3 is N; Z 4 and Z 5 are C;
  • Ring A 2 is a 5-heteroaryl group or a 5-heteroaryl group substituted by one or more R a1 ; for example, a 5-heteroaryl group;
  • Ring A 3 is 5-6-membered heterocycloalkyl, 5-6-membered heterocycloalkenyl, 6-membered heteroaryl or Alternatively, substituted with one or more R a2 : 5-6 membered heterocycloalkyl, 5-6 membered heterocycloalkenyl, 6 membered heteroaryl or such as 5-membered heterocycloalkyl, 5-membered heterocycloalkenyl or
  • R 12 and R 13 are independently C 1 -C 4 alkyl-O-;
  • R 32 and R 33 are independently C 1 -C 4 alkyl
  • Z 8 is CH or N;
  • R 14 is C 1 -C 4 alkyl-O-;
  • R 23 and R 24 are H;
  • R 34 is C 1 -C 4 alkyl or 3-6 membered cycloalkyl;
  • a carbon atom with "*" means that when it is a chiral carbon atom, it is in S configuration, R configuration or a mixture thereof.
  • W is C(R 7' R 7" ); R 7 is hydrogen;
  • R 7' and R 7" are independently hydrogen or C 1 -C 4 alkyl-O-;
  • R 6 is hydrogen
  • R 4' and R 4 are independently hydrogen
  • n 1;
  • Ring A 3 is 5-membered heterocycloalkyl, 5-membered heterocycloalkenyl or
  • R 13 is C 1 -C 4 alkyl-O-;
  • R 33 is C 1 -C 4 alkyl
  • Z 8 is CH or N;
  • R 14 is C 1 -C 4 alkyl-O-;
  • R 23 and R 24 are H;
  • R 34 is C 1 -C 4 alkyl or 3-6 membered cycloalkyl;
  • a carbon atom with "*" means that when it is a chiral carbon atom, it is in S configuration, R configuration or a mixture thereof.
  • R 7 , R 7' and R 7" are independently C 1 -C 4 alkyl or C 1 -C 4 alkyl-O-
  • the C 1 -C 4 alkyl or C 1 -C 4 alkyl C1 - C4 alkyl in alkyl-O- is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl; for example methyl.
  • R 6 is a C 1 -C 4 alkyl group or a hydroxy C 1 -C 4 alkyl group
  • the C 1 -C 4 alkyl group in the C 1 -C 4 alkyl group and the hydroxy C 1 -C 4 alkyl group is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl; eg methyl.
  • ring B is a 6-membered heteroaryl group
  • the 6-membered heteroaryl group is pyridyl; for example (The b-terminal represents connection with COOH).
  • R b is a 3-6 membered cycloalkyl group
  • the 3-6 membered cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl; for example, cyclopentyl.
  • R b is a 5-6 membered heterocycloalkyl
  • the 5-6 membered heterocycloalkyl is tetrahydrofuranyl; for example (The c-terminal represents a parallel connection to ring B).
  • R b is a 5-6-membered heteroaryl group
  • the 5-6-membered heteroaryl group is a pyridyl group; for example (c-terminal represents a cyclic connection to ring B) or imidazolyl (e.g. The c-terminus represents a cyclic connection to ring B).
  • Ring A 2 is a 5-6 membered heterocycloalkyl
  • the 5-6 membered heterocycloalkyl is ( Indicates the position where the ring is connected to ring A 1 ).
  • Ring A 2 is a 5-6 membered heterocyclenyl
  • the 5-6 membered heterocyclenyl is ( represents the position where the ring is connected in parallel with ring A 1 , means single or double bonds).
  • Ring A 2 is a 5-6 membered heteroaryl
  • the 5-6 membered heteroaryl is
  • Ring A 3 is a 5-6 membered heterocycloalkyl
  • the 5-6 membered heterocycloalkyl is ( Indicates the position where the ring is connected to ring A 1 ).
  • Ring A 3 is a 5-6 membered heterocyclenyl
  • the 5-6 membered heterocyclenyl is ( represents the position where the ring is connected in parallel with ring A 1 , means single or double bonds).
  • Ring A 3 is a 6-membered heteroaryl group
  • the 6-membered heteroaryl group is
  • R a1 and R a2 are independently C 1 -C 4 alkyl or C 1 -C 4 alkyl-O-, said C 1 -C 4 alkyl or C 1 -C 4 alkyl -O- where C 1 -C 4 alkyl is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl; for example methyl.
  • R 11 , R 31 , R 12 , R 32 , R 13 , R 23 and R 33 are independently C 1 -C 4 alkyl, C 1 -C 4 alkyl-O-, the C 1 - C 1 -C 4 alkyl in C 4 alkyl and C 1 -C 4 alkyl-O- is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl; eg methyl.
  • R 11 , R 31 , R 12 , R 32 , R 13 , R 23 and R 33 are independently 3-6 membered cycloalkyl
  • the 3-6 membered cycloalkyl is cyclopropyl, cyclobutyl cyclopentyl, cyclopentyl and cyclohexyl; eg cyclopropyl.
  • R 14 is C 1 -C 4 alkyl-O-
  • the C 1 -C 4 alkyl in the C 1 -C 4 alkyl-O- is methyl, ethyl, n-propyl, isopropyl propyl, n-butyl, isobutyl, sec-butyl or tert-butyl; eg methyl.
  • R 34 is a C 1 -C 4 alkyl group
  • the C 1 -C 4 alkyl group is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-Butyl; eg methyl.
  • R 34 is a 3-6 membered cycloalkyl group
  • the 3-6 membered cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl; for example, cyclopropyl.
  • R 7' and R 7" are independently hydrogen, F, methyl, ethyl-O-.
  • R5 is For example
  • R 11 , R 31 , R 12 , R 32 , R 13 and R 33 are independently methyl, methyl-O- or cyclopropyl
  • R 11 , R 12 and R 13 are independently methyl, methyl-O- or cyclopropyl
  • R 31 , R 32 and R 33 are independently methyl.
  • R 14 is methyl-O-.
  • R 34 is methyl or cyclopropyl.
  • W is methylene; e.g.
  • heterocyclic compound represented by formula I is any of the following structures,
  • the pharmaceutically acceptable salt of the heterocyclic compound represented by formula I is any of the following structures,
  • the heterocyclic compounds represented by formula I or their pharmaceutically acceptable salts, isotopic analogs, and prodrugs have one or more chiral carbon atoms, so they can be separated to obtain optically pure isomers isomers, such as pure enantiomers, or racemates, or mixed isomers. Pure single isomers can be obtained by separation methods in the art, such as chiral crystallization into salts, or chiral preparative column separation.
  • the heterocyclic compounds shown in formula I or their pharmaceutically acceptable salts, isotopic analogs and prodrugs, if there is a stereoisomer can be a single stereoisomer or their in the form of a mixture (eg, a racemate).
  • stereoisomer refers to a cis-trans isomer or an optical isomer.
  • stereoisomers can be separated, purified and enriched by asymmetric synthesis methods or chiral separation methods (including but not limited to thin layer chromatography, spin chromatography, column chromatography, gas chromatography, high pressure liquid chromatography, etc.) It can be obtained by chiral resolution by forming bonds with other chiral compounds (chemical bonding, etc.) or forming salts (physical bonding, etc.).
  • single stereoisomer means that the mass content of one stereoisomer of the compound of the present invention relative to all stereoisomers of the compound is not less than 95%.
  • heterocyclic compound represented by formula I or its pharmaceutically acceptable salts, isotopic analogs, and prodrugs has a tautomer
  • it can be a single tautomer. or their mixtures, preferably in the form of more stable tautomers.
  • structural fragments are included:
  • heterocyclic compounds shown in formula I or their pharmaceutically acceptable salts, isotopic analogs and prodrugs can be synthesized by methods similar to those known in the chemical field, and the steps and conditions can be Reference is made to the procedures and conditions for similar reactions in the art, particularly syntheses as described herein.
  • Starting materials are typically from commercial sources such as Aldrich or can be readily prepared using methods well known to those skilled in the art (obtained via SciFinder, Reaxys online database).
  • heterocyclic compounds shown in formula I or their pharmaceutically acceptable salts, isotopic analogs, and prodrugs can also be prepared by the prepared heterocyclic compounds shown in formula I.
  • Cyclic compounds or their pharmaceutically acceptable salts, isotopic analogs, and prodrugs using conventional methods in the art, through peripheral modification to obtain other described heterocyclic compounds as shown in formula I or their pharmaceutically acceptable compounds of salt.
  • Necessary raw materials or reagents for preparing the heterocyclic compounds represented by formula I or their pharmaceutically acceptable salts, isotopic analogs and prodrugs can be obtained commercially, or prepared by synthetic methods known in the art.
  • Compounds of the invention can be prepared as free bases or as acid salts thereof as described in the experimental section below.
  • the term pharmaceutically acceptable salt refers to a pharmaceutically acceptable salt as defined herein, and has all the effects of the parent compound.
  • Pharmaceutically acceptable salts The pharmaceutically acceptable salts can be prepared by treating the organic base in a suitable organic solvent with the corresponding acid according to conventional methods.
  • the present invention also provides a method for preparing the above-mentioned heterocyclic compound shown in formula I, which comprises the following steps:
  • R 8 is a C 1 -C 4 alkyl group (such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl; such as methyl or ethyl base);
  • R b , R 4 , R 4′ , R 6 , R 7 , A, W, m and * are all as defined above.
  • the conditions and operations of the de-esterification reaction can be conventional conditions and operations in this type of reaction in the field; in the present invention, the following are preferred:
  • Said solvent can be water and/or alcoholic solvent (eg, methanol and/or ethanol), such as water and alcoholic solvent.
  • the consumption of the described solvent can not affect the reaction. ).
  • the base may be an alkali metal hydroxide, such as sodium hydroxide and/or potassium hydroxide.
  • the temperature of the deesterification reaction may be 0-100°C (eg, 70-80°C).
  • the progress of the shown deesterification reaction can be detected by conventional monitoring methods in the art (such as TLC, HPLC or NMR), and generally the reaction end point is when the compound shown in formula II disappears or no longer reacts .
  • the present invention also provides the above-mentioned heterocyclic compound represented by formula II,
  • R 8 is a C 1 -C 4 alkyl group (such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl; such as methyl or ethyl base);
  • R b , R 4 , R 4′ , R 6 , R 7 , A, W, m and * are all as defined above.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the above-mentioned heterocyclic compound represented by formula I or a pharmaceutically acceptable salt, isotopic analog, prodrug thereof, and one or more pharmaceutically acceptable acceptable carrier.
  • the amount of the heterocyclic compound represented by formula I or a pharmaceutically acceptable salt, isotope analog, or prodrug thereof can be a therapeutically effective amount.
  • the pharmaceutically acceptable carriers can be those excipients widely used in the field of pharmaceutical production. Excipients are mainly used to provide a safe, stable and functional pharmaceutical composition, and can also provide a method to enable the subject to dissolve the active ingredient at a desired rate after administration, or to promote the activity of the subject after the composition is administered. The ingredients are effectively absorbed.
  • the pharmaceutical excipients can be inert fillers, or provide some function, such as stabilizing the overall pH of the composition or preventing degradation of the active ingredients of the composition.
  • Described pharmaceutical adjuvants may include one or more of the following adjuvants: binders, suspending agents, emulsifiers, diluents, fillers, granulating agents, adhesives, disintegrating agents, lubricants, anti-sticking agents Agents, glidants, wetting agents, gelling agents, absorption delaying agents, dissolution inhibitors, enhancers, adsorbents, buffers, chelating agents, preservatives, colorants, flavors and sweeteners.
  • adjuvants may include one or more of the following adjuvants: binders, suspending agents, emulsifiers, diluents, fillers, granulating agents, adhesives, disintegrating agents, lubricants, anti-sticking agents Agents, glidants, wetting agents, gelling agents, absorption delaying agents, dissolution inhibitors, enhancers, adsorbents, buffers, chelating agents, preservatives, colorants, flavors and sweeten
  • compositions of the present invention can be prepared in light of the disclosure using any method known to those skilled in the art. For example, conventional mixing, dissolving, granulating, emulsifying, attenuating, encapsulating, entrapping or lyophilizing processes.
  • compositions of the present invention may be administered in any form, including injection (intravenous), mucosal, oral (solid and liquid formulations), inhalation, ophthalmic, rectal, topical or parenteral (infusion, injection, implant Intradermal, subcutaneous, intravenous, intraarterial, intramuscular) administration.
  • the pharmaceutical compositions of the present invention may also be in controlled release or delayed release dosage forms (eg, liposomes or microspheres).
  • solid oral formulations include, but are not limited to, powders, capsules, caplets, softgels, and tablets.
  • liquid formulations for oral or mucosal administration include, but are not limited to, suspensions, emulsions, elixirs, and solutions.
  • topical formulations include, but are not limited to, creams, gels, ointments, creams, patches, pastes, foams, lotions, drops, or serum formulations.
  • formulations for parenteral administration include, but are not limited to, solutions for injection, dry formulations that can be dissolved or suspended in a pharmaceutically acceptable carrier, suspensions for injection, and emulsions for injection.
  • suitable formulations of the pharmaceutical compositions include, but are not limited to, eye drops and other ophthalmic formulations; aerosols: such as nasal sprays or inhalants; liquid dosage forms suitable for parenteral administration; suppositories and lozenges agent.
  • One object of the present invention is to provide the use of the above-mentioned heterocyclic compound represented by formula I or a pharmaceutically acceptable salt, isotope analog or prodrug thereof as a complement factor B inhibitor.
  • the complement factor B inhibitor can be used in mammalian organisms; it can also be used in vitro, mainly for experimental purposes, such as: providing comparison as a standard sample or a control sample, or according to the routine in the art Methods A kit was prepared to provide rapid detection of the inhibitory effect of complement factor B.
  • One object of the present invention is to provide the above-mentioned use of the above-mentioned heterocyclic compound represented by formula I or a pharmaceutically acceptable salt, isotopic analog and prodrug thereof in the preparation of medicine;
  • the medicine can be For the treatment of the abnormal activation of complement factor B, or with complement factor B to perform normal functions in the related diseases; or, the drug can be used for the treatment of hematological, autoimmune, inflammatory and Drugs for neurodegenerative and other diseases.
  • the heterocyclic compound represented by formula I or a pharmaceutically acceptable salt, isotope analog or prodrug thereof can be in a therapeutically effective amount.
  • An object of the present invention is to provide a heterocyclic compound represented by formula I as described above or a pharmaceutically acceptable salt, isotopic analog, prodrug thereof in the treatment of abnormal activation of the complement system, or with complement The application of the system to perform normal functions in related diseases.
  • the heterocyclic compounds or their pharmaceutically acceptable salts, isotopic analogs and prodrugs are particularly useful as pharmaceuticals and pharmaceuticals.
  • the above-mentioned diseases related to the abnormal activation of the complement system or the normal function of the complement system can be hematological, autoimmune, inflammatory and neurodegenerative diseases; the diseases include but are not limited to: Paroxysmal nocturnal hemoglobinuria (PNH), primary glomerulonephritis (IgAN), membranous nephropathy (MN), C3 glomerulonephritis (C3G), age-related macular degeneration (AMD), geographic Atrophy (GA), atypical hemolytic uremic syndrome (aHUS), hemolytic uremic syndrome (HUS), complications of hemodialysis, hemolytic anemia or hemodialysis, neuromyelitis (NMO), arthritis, rheumatoid Arthritis, liver-like inflammations, dermatomyositis and amyotrophic lateral sclerosis, myasthenia gravis (MG), respiratory diseases and cardiovascular diseases.
  • PNH Paroxysmal nocturnal hemoglobinuria
  • Said complement system is preferably a complement system associated or regulated by complement factor B.
  • the heterocyclic compound represented by formula I or a pharmaceutically acceptable salt, isotope analog or prodrug thereof can be administered in the form of a pharmaceutical composition.
  • compositions can be prepared according to methods well known in the art of pharmacy and can be administered by various routes depending on the need for local or systemic treatment and the area to be treated. Administration can be topical (including epidermal and transdermal, ocular and mucosal, including intranasal, vaginal and rectal delivery), pulmonary (eg, by powder or aerosol inhalation or insufflation, including by nebulizer; intratracheal or intranasal) , in the form of oral or parenteral administration.
  • Oral administration may include dosage forms formulated for once-daily or twice-daily (BID) administration.
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial such as intrathecal or intraventricular administration.
  • Parenteral administration can be in the form of a single bolus dose, or it can be by a continuous infusion pump.
  • Pharmaceutical compositions and formulations for topical administration may include transdermal patches, salves, lotions, ointments, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, water, powder or oily bases, thickeners and the like may be necessary or desirable.
  • treatment refers to therapeutic or palliative measures.
  • Beneficial or desired clinical outcomes include, but are not limited to: all or part of the symptoms associated with the disease or disorder or condition, reducing the extent of the disease, stabilizing (ie, not worsening) the disease state, delaying or slowing the progression of the disease, alleviating or alleviating the disease Status (eg, one or more symptoms of a disease), and detectable or undetectable remission (whether partial or total).
  • Treatment can also mean prolonging survival as compared to expected survival without treatment.
  • the heterocyclic compounds represented by formula I or pharmaceutically acceptable salts, isotopic analogs, prodrugs or pharmaceutical compositions as described above can be used for prophylaxis as defined herein diseases and disorders (eg, hematologic, autoimmune, inflammatory, and neurodegenerative diseases).
  • diseases and disorders eg, hematologic, autoimmune, inflammatory, and neurodegenerative diseases.
  • preventing means preventing, in whole or in part, the onset, recurrence or spread of a disease or disorder or symptoms thereof described herein.
  • prodrug represents the conversion of a compound to a compound of formula I in vivo. Such conversion is effected by hydrolysis of the prodrug in blood or enzymatic conversion to the parent structure in blood or tissue.
  • the prodrug compounds of the present invention can be esters.
  • esters that can be used as prodrugs include phenyl esters, aliphatic (C1-24) esters, acyloxymethyl esters, carbonates, Carbamates and amino acid esters.
  • a compound of the present invention contains a hydroxyl group, which can be acylated to give the compound in prodrug form.
  • prodrug forms include phosphates, such as these phosphates are phosphorylated by the hydroxyl group on the parent.
  • phosphates such as these phosphates are phosphorylated by the hydroxyl group on the parent.
  • a complete discussion of prodrugs can be found in the following references: T.Higuchiand V.Stella, Pro-drugsas Novel Delivery Systems, Vol.14 of the ACSSymposium Series, Edward B.Roche, ed., Bioreversible Carriersin Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, J. Rautio et al., Prodrugs: Design and Clinical Applications, Nature Review Drug Discovery, 2008, 7, 255-270, and SJ Hecker et al., Prodrugs of Phosphates and Phosphonates, Journal of Medicinal Chemistry, 2008, 51, 2328-2345 .
  • Compounds of Formula I and salts thereof are intended to encompass any isotopically-labeled (or "radiolabeled") derivatives of compounds of Formula I or salts thereof.
  • Such derivatives are derivatives of a compound of formula I, or a salt thereof, wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from that normally found in nature.
  • the radionuclide used will depend on the specific application of the radiolabeled derivative. For example, for in vitro receptor labeling and competition assays, 3H or14C are often useful. For radiographic applications, 11C or18F are often useful.
  • isotopic variants of the compounds of the present invention may be useful, for example, to investigate the mechanism of action or distribution of the active component in vivo; due to the relatively easy manufacturability and Detectability, compounds labeled with3H or14C isotopes are particularly useful for this purpose.
  • isotopes such as deuterium may yield particular therapeutic benefits due to the compound's greater metabolic stability, such as increased half-life in vivo or reduced effective doses required; thus such modifications of the compounds of the present invention may also be used in some This case constitutes a preferred embodiment of the present invention.
  • Isotopic variants of the compounds of the present invention can be prepared by methods known to those skilled in the art, for example by the methods described below and in the working examples, by using the corresponding isotopically modified specific reagents and/or starting compounds .
  • pharmaceutical excipient refers to a pharmaceutically acceptable chemical substance, such as an agent known to those of ordinary skill in the art of pharmacy to aid in the administration of pharmaceuticals. It is a compound that can be used to prepare a pharmaceutical composition that is generally safe, non-toxic, and biologically or otherwise undesirable, including excipients that are pharmaceutically acceptable for veterinary and human use. Common excipients include binders, surfactants, diluents, disintegrants and lubricants.
  • C1 - C6 alkyl refers to an alkyl group as defined below having a total of 1, 2, 3, 4, 5 or 6 carbon atoms.
  • the total number of carbon atoms in the simplified notation does not include carbons that may be present in the substituents of the group.
  • substituted means that any one or more hydrogen atoms on a specified atom are replaced by a substituent, including deuterium and hydrogen variants, as long as the valence of the specified atom is normal and the substituted compound is stable .
  • substituted means that one or more hydrogen atoms in a given structure have been replaced with a specified substituent.
  • the substituents are independent of each other, that is, the one or more substituents may be different from each other or the same of.
  • a substituent group may be substituted at various substitutable positions of the substituted group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents can be substituted identically or differently at each position.
  • Cx - Cy alkyl refers to a straight or branched chain saturated hydrocarbon containing from x to y carbon atoms.
  • C1 - C6 alkyl or " C1-6 alkyl” specifically refers to the independently disclosed methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl and C6 alkanes
  • C 1-4 alkyl specifically refers to independently disclosed methyl, ethyl, C 3 alkyl (ie propyl, including n-propyl and isopropyl ), C 4 alkyl (ie butyl, including n-butyl, isobutyl, sec-butyl and tert-butyl).
  • halogen is selected from F, Cl, Br or I, especially F or Cl.
  • alkoxy refers to the group -OR X , wherein R X is an alkyl group as defined above.
  • moiety refers to a specific fragment or functional group in a molecule.
  • a chemical moiety is usually thought of as a chemical entity embedded or attached to a molecule.
  • linking substituents are described.
  • the Markush variables listed for that group should be understood to be the linking group.
  • the structure requires a linking group and "alkyl” is listed for the definition of a Markush group for that variable, it should be understood that the "alkyl” represents the attached alkylene group.
  • alkyl group when an alkyl group is clearly represented as a linking group, the alkyl group represents the alkylene group to which it is attached, eg, the group "halo-C 1 -C 6 alkane
  • a C 1 -C 6 alkyl group in "radical” should be understood as a C 1 -C 6 alkylene group.
  • alkylene refers to a saturated divalent hydrocarbyl group obtained by removing two hydrogen atoms from a saturated straight or branched chain hydrocarbyl group.
  • alkylene groups include methylene ( -CH2- ), ethylene ⁇ including -CH2CH2- or -CH( CH3 )- ⁇ , isopropylidene ⁇ including -CH( CH3 ) )CH 2 -or -C(CH 3 ) 2 - ⁇ and so on.
  • alkyl as a group or as part of another group (eg, as used in halogen-substituted alkyl groups, etc.) is meant to include branched and straight chains having the specified number of carbon atoms.
  • a saturated aliphatic hydrocarbon group such as a straight or branched saturated hydrocarbon chain containing 1 to 20 carbon atoms; another example, a C1 - C6 alkyl group.
  • C1 - C6 alkyl is meant to include groups having 1, 2, 3, 4, 5, or 6 carbon atoms in a straight or branched chain structure.
  • propyl is C3 alkyl (including isomers, such as n-propyl or isopropyl); butyl is C4 alkyl (including isomers, such as n-butyl, sec-butyl) , isobutyl or tert-butyl); pentyl is C5 alkyl (including isomers such as n-pentyl, 1-methyl-butyl, 1-ethyl-propyl, 2-methyl) -1-butyl, 3-methyl-1-butyl, isopentyl, tert-amyl or neopentyl); hexyl is C6 alkyl (including isomers such as n-hexyl, 1-ethyl yl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl , 1,3-dimethylbutyl, 2-ethyl
  • cycloalkyl refers to a saturated monocyclic or polycyclic carbocyclic substituent consisting only of carbon and hydrogen atoms and which may be attached to the remainder of the molecule by a single bond through any suitable carbon atom; when When polycyclic, it can be a folded, bridged, or spiro linked (ie, two geminal hydrogens on a carbon atom are replaced by an alkylene group) folded, bridged, or spiro linked.
  • typical monocyclic cycloalkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl.
  • cycloalkenyl refers to a monocyclic, polycyclic or bridged carbocyclic substituent containing a partially unsaturated double bond, and which may be via any suitable carbon Atom is attached to the rest of the molecule by a single bond; when polycyclic, it can be a bridged ring system or spirocyclic linkage, either paracyclic or spiro (i.e., two gem hydrogens on a carbon atom are replaced by an alkylene group) system.
  • cycloalkenyl is a monocyclic, unsaturated carbocycloalkenyl group having 5 to 6 ring atoms ("5-6 membered cycloalkenyl”). This term includes, but is not limited to, cyclopentenyl (eg ), cyclopentadienyl (e.g. ), cyclohexenyl (e.g. ) or cyclohexadienyl, and its stereoisomers.
  • a heterocycloalkyl group may be monocyclic ("monocyclic heterocycloalkyl"), or a bicyclic, tricyclic or more cyclic ring system, which may include and Cyclic (fused), bridged (bridged), or spiro ring systems (eg, bicyclic ring systems ("bicyclic heterocycloalkyl").
  • Heterocycloalkyl bicyclic ring systems may be in a and is saturated.
  • "heterocycloalkyl” is a 5-6 membered heterocycloalkyl.
  • Exemplary 3-membered heterocyclyl groups Including, but not limited to, aziridyl, oxiranyl, and thiirane, or stereoisomers thereof;
  • exemplary 4-membered heterocyclyl groups include, but are not limited to, azetidine group, propylene oxide group, thietane group, or isomers and stereoisomers thereof;
  • exemplary 5-membered heterocyclyl groups include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, thiazolidinyl, isothiazolidinyl, oxazolidinyl, isoxazolidinyl, imidazolidinyl, pyrazolidinyl, dioxo
  • Exemplary 6-membered heterocyclyl groups include, but are not limited to, piperidinyl, tetrahydropyranyl, cyclopentyl sulfide, morpholinyl, thio Morpholinyl, dithianyl, dioxanyl, piperazinyl, triazinyl, or isomers and stereoisomers thereof.
  • heterocycloalkenyl means a cyclic alkenyl group attached through a heteroatom or heteroatom group.
  • heterocycloalkenyl includes the definitions of “hetero” and cycloalkenyl above.
  • a heterocycloalkenyl group may either be monocyclic ("monocyclic heterocycloalkenyl"), or a bicyclic, tricyclic or more cyclic ring system, which may include Fused, bridged, or spiro ring systems (eg, bicyclic ring systems ("bicyclic heterocycloalkenyl").
  • Heterocycloalkenyl bicyclic ring systems may include one or more in one or both rings heteroatom.) and contains unsaturated double bonds.
  • a heterocycloalkenyl group can be attached to the rest of the molecule via a carbon atom and by a single bond; in a heterocycloalkenyl group containing one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom; or, to the rest of the molecule And ring connections; as long as the valence permits.
  • aryl refers to an all-carbon aromatic group with a fully conjugated pi-electron system, which may be monocyclic or fused, usually having 6-14 carbon atoms, preferably 6-12 carbon atoms, and most It preferably has 6 carbon atoms.
  • aryl groups include, but are not limited to, phenyl, naphthyl, and anthracenyl.
  • heteroaryl refers to an aromatic group containing heteroatoms, which may be monocyclic or fused, preferably containing 1-4 5-12 membered heteroaryl groups independently selected from N, O and S, including but not limited to Not limited to pyrrolyl, furanyl, thienyl, indolyl, imidazolyl, oxazolyl, isoxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, quinolyl, iso Quinolinyl, (benzo)oxazolyl, (benzo)furanyl, (benzo)thienyl, (benzo)thiazolyl, triazolyl.
  • a 5-6 membered monocyclic heteroaryl group typically contains 1 or more heteroatoms independently selected from N, O and S.
  • heteroaryl is a 5-6 membered heteroaryl, wherein the heteroatoms are selected from one or more of N, O and S, and the number of heteroatoms is 1, 2 or 3.
  • each step and condition may refer to the conventional operation steps and conditions in the art.
  • the present invention employs standard nomenclature and standard laboratory procedures and techniques of analytical chemistry, synthetic organic chemistry, and optics. In some cases, standard techniques are used for chemical synthesis, chemical analysis, and performance testing of light-emitting devices.
  • the description mode "...independently” used in the present invention should be understood in a broad sense, meaning that the described individuals are independent of each other and may be independent of each other. are the same or different specific groups.
  • the description mode "...independently” can either mean that in different groups, the specific options expressed between the same symbols do not affect each other; it can also mean that in the same group, the same symbols are The specific options expressed between them do not affect each other.
  • the reagents and raw materials used in the present invention are all commercially available.
  • the positive improvement effect of the present invention is that: the compound of the present invention has inhibitory activity on complement factor B (in the complement hemolytic activity test, the IC 50 values of the inhibitory activity against complement factor B are all ⁇ 5 ⁇ M, most IC 50 ⁇ 1.5 ⁇ M, and some IC 50 values. 50 ⁇ 1 ⁇ M).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • MS was performed with Agilent 6110, Agilent 1100, Agilent 6120, and Agilent G6125B LC-MS.
  • HPLC used Shimadzu HPLC-2010C high pressure liquid chromatograph (XBRIDGE 2.1*50mm, 3.5um chromatographic column).
  • the thin layer chromatography silica gel plate uses Yantai Qingdao GF254 silica gel plate, the size of the silica gel plate used for thin layer chromatography (TLC) is 0.15mm-0.2mm, and the size of the thin layer chromatography separation and purification products is 0.4mm-0.5mm .
  • HPLC preparations were performed using Waters 2767, Waters 2545, and innovative Hengtong LC3000 preparative chromatographs.
  • the pressurized hydrogenation reaction used Beijing Jiawei Kechuang Technology GCD-500G hydrogen generator.
  • Microwave reactions were performed using a Biotage initiator+ type microwave reactor.
  • the reactions were carried out in an argon atmosphere or a nitrogen atmosphere.
  • Argon or nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon with a volume of about 1 liter.
  • Hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon with a volume of about 1 liter.
  • reaction temperature is room temperature, and the temperature range is 20°C-30°C.
  • the reaction system was diluted with anhydrous tetrahydrofuran (500 mL), cooled to -5 °C, 4-methoxypyridine (25 mL) was added, and benzyl chloroformate (35 mL) was slowly added dropwise ( The temperature of the system was maintained below 0°C), after the dropwise addition, the reaction was continued at 0°C for 2 hours, and then the temperature was raised to room temperature and reacted at this temperature for 16 hours.
  • anhydrous tetrahydrofuran 500 mL
  • 4-methoxypyridine 25 mL
  • benzyl chloroformate 35 mL
  • tetrabutylammonium fluoride tetrahydrofuran solution (1 M, 30 mL) and Intermediate 4 (5 g) were sequentially added, and the reaction was carried out at room temperature for 2 hours. After the reaction was completed, water (100 mL) was added to dilute, and the mixture was extracted three times with ethyl acetate (50 mL). The extract was washed with saturated brine (100 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column.
  • reaction solution was directly purified by Prep-HPLC (chromatographic column: Gemini-C18, 150 x 21.2 mm, 5um; flow term: acetonitrile-water (0.1% formic acid); gradient: 15-35%), the obtained The solution was concentrated, and the remaining small amount of aqueous solution was lyophilized to obtain the title compound (33.0 mg, yield: 46%, containing 0.5 equivalent of formic acid).
  • Aqueous sodium hydroxide solution (15%, 10 mL) was added to a solution of N-(4-methoxy-2-methyl-6-nitrophenyl)acetamide (500 mg) in ethanol (10 mL) at room temperature, The reaction was heated to 90°C and carried out at this temperature for 16 hours. After the reaction, the reaction system was cooled to room temperature, concentrated under reduced pressure to remove ethanol, diluted with water (20 mL), adjusted to pH 5-6 with 5M hydrochloric acid, and then extracted with ethyl acetate (20 mL) five times.
  • Phosphorus oxychloride/N,N-dimethylformamide mixed solution 9:10, 6mL was added to the 50mL single-neck flask, stirred for 15 minutes under nitrogen atmosphere and room temperature, then intermediate 4 (350mg) was added, and the reaction was heated to 60°C and stirred at this temperature for 1 hour.
  • Lithium hydroxide (100 mg) was added to a methanol/tetrahydrofuran/water mixed solution (3 mL, 1:1:1) of intermediate 7 (100 mg), and the reaction was stirred at room temperature overnight. After the reaction, the reaction solution was directly concentrated, and the residue was purified by Prep-HPLC (chromatographic column: Gemini-C18, 150 x 21.2 mm, 5um; flow term: acetonitrile-water (0.1% formic acid); gradient: 25-40% ), the obtained product solution was concentrated to remove the solvent, and the remaining small amount of aqueous solution was freeze-dried to obtain the target compound (50 mg, yield: 51%, containing 0.5 equivalent of formic acid).
  • Prep-HPLC chromatographic column: Gemini-C18, 150 x 21.2 mm, 5um; flow term: acetonitrile-water (0.1% formic acid); gradient: 25-40%
  • tert-butyllithium (1.68 mL) was slowly added dropwise to a tetrahydrofuran solution (5 mL) of intermediate 3 (400 mg), the reaction was stirred at -78°C for 1.5 hours, and N,N-dimethyl dimethyl was added dropwise.
  • a solution of formamide (110 mg) in tetrahydrofuran (15 mL). After the dropwise addition, the reaction system was gradually warmed to room temperature and stirred at that temperature for 1.5 hours. After the reaction, the reaction solution was quenched by adding saturated aqueous ammonium chloride solution (20 mL), and extracted three times with ethyl acetate (50 mL).
  • reaction solution was directly purified by Prep-HPLC (chromatographic column: Gemini-C18, 150 x 21.2 mm, 5um; flow term: acetonitrile-water (0.1% formic acid); gradient: 15-35%), the obtained solution Concentrated, and the remaining small amount of aqueous solution was lyophilized to obtain the title compound (20.1 mg, yield: 16%, containing 0.8 equiv of formic acid).
  • N,N-dimethylformamide (12mL), intermediate 1 (1.05g), trimethylcyclotriboroxane (3.3g), cesium carbonate (2.87g) and tetrakis were added in sequence (triphenylphosphine)palladium (254 mg), the reaction system was heated to 90°C under nitrogen protection and stirred at this temperature for 16 hours. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (50 mL), extracted with ethyl acetate (100 mL) three times, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filtered, and the filtrate was reduced.
  • n-Butyllithium (0.37 mL) was slowly added dropwise to a solution of Intermediate 1 (160 mg) in tetrahydrofuran (5 mL) at -78°C; after the reaction was stirred at -78°C for 1 hour, N,N-dimethyl Formamide (99 mg) was slowly added dropwise to the reaction solution, and then the reaction mixture was naturally warmed to room temperature and stirred at that temperature for 16 hours. After the reaction, the reaction mixture was poured into water (20 mL), and extracted with ethyl acetate (15 mL) twice.
  • the SPR experiment was performed at 25°C with PBS buffer supplemented with 0.05% (v/v) P20 and 5% DMSO as the running buffer, and the analytical instrument used was Biacore 8K from GE Healthcare.
  • CM7 chips (GE Healthcare) were activated with 400 mM EDC and 100 mM NHS at a flow rate of 30 ⁇ L/min for 420 s.
  • the complement factor B was diluted to 50 ⁇ g/mL with 10 mM sodium acetate (pH 4.0), and then coupled at a flow rate of 10 ⁇ L/min for 1200 s to covalently fix the complement B factor on the detection chip (the protein immobilization level was 25000RU); then the detection chip Chip blocking was performed with 1 M ethanolamine hydrochloride at a flow rate of 10 ⁇ L/min for 300 s.
  • the concentration of the compound to be tested was 500 ⁇ M, the binding time was 120 s, and the dissociation time was 300 s.
  • Data analysis was performed using a 1:1 binding model (Biacore Insight Evaluation Software, Version 2.0.15.12933).
  • the inhibitory activity of the compounds on human complement factor B was screened by competitive binding experiments using Cy5 fluorescently labeled small molecule inhibitors as probes.
  • Complement factor B was incubated with EZ-Link TM Sulfo-NHS-LC-LC-Biotin at a ratio of 1:2 on ice for 1 hour and then 1M Tris (pH 7.5) was added to stop the reaction.
  • Biotin-labeled complement factor B was then purified twice with 2 mL of Zeba TM desalt spin column (EZ-LinkTM Sulfo-NHS-LC-Biotin instructions).
  • biotin-labeled complement factor B with a final concentration of 10 nM was pre-incubated with different concentrations of compounds in buffer for 1 hour at room temperature.
  • the reaction was initiated by addition of Cy5 fluorescently labeled probe and europium chelate-labeled streptavidin (petroleum ether rkin Elmer, #AD0060) at final concentrations of 75 nM and 5 nM, respectively.
  • Kinetic readings were performed on a microplate reader (excitation at 337 nm, emission at 665 nm, 70 ⁇ s time-gated) to read time-dependent fluorescence energy transfer (TR-FRET) data to determine IC50.
  • TR-FRET time-dependent fluorescence energy transfer
  • the test concentration of the test compound is 10 ⁇ M starting, 3-fold dilution, 7 concentration points, single-well detection.
  • the test compounds were diluted to a 1000-fold final concentration with DMSO in a 96-well plate, and then diluted with Diluent (SYSTEM ALTERNATIVE PATHWAY AP330) diluted to a 5-fold final concentration solution. Transfer 30uL to a 96-well plate, add 120 ⁇ L of spare serum, and incubate at room temperature for 15 minutes. Add 30 ⁇ L of 5 ⁇ DMSO and 120 ⁇ L of spare serum to the positive control wells, and add 30 ⁇ L of 5 ⁇ DMSO and 120 ⁇ L of Diluent to the negative control wells.
  • RE from healthy Japanese white rabbits freshly suspended in GVB0 buffer containing 10 mM Mg-EGTA was added to a final concentration of 1 x 108 cells/ml and incubated at 37°C for 30 minutes.
  • Positive control (100% lysis) consisted of GVB0 buffer containing 10 mM Mg-EGTA with NHS and RE but no test compound;
  • negative control (0% lysis) consisted of NHS with inactivated (56°C for 30 min or 65°C for 5 min) and RE but no test compound in GVB0 buffer containing 10 mM Mg-EGTA. The samples were centrifuged at 2000g for 5 minutes and the supernatant was collected.

Abstract

式I所示的杂环类化合物、其制备方法、包含其的药物组合物及其用途。所述杂环类化合物可作为补体因子B抑制剂,并用于制备治疗与补体***的异常激活或者与补体***执行正常功能中出现的相关疾病的药物;可作为炎症和免疫性相关的疾病的治疗剂。

Description

一种杂环类化合物、其制备方法及用途
本申请要求申请日为2020/8/7的中国专利申请2020107875372和申请日为2021/3/18的中国专利申请2021102931493的优先权。本申请引用上述中国专利申请的全文。
技术领域
本发明涉及一种杂环类化合物、其制备方法及用途。
背景技术
补体是免疫***中一类可溶性模式识别分子,可以行使多种效应功能。在自然条件下,补体成分以无活性的酶原形式存在,多种特异性和非特异性免疫学机制使这些无活性的酶原分解,产生有活性的大片段和小片段。其中,大片段通常停留在病原体或细胞表面,使后者裂解或者加速其清除;小片段离开细胞表面、介导多种炎症反应。补体的激活由两个紧密相随的过程组成,并由此形成补体激活的级联反应。目前已知的补体激活途径主要包括3条:经典途径、凝集素途径、旁路途径。虽然3条补体激活途径的启动机制和激活顺序不同,但是它们具有共同的末端通路。其中,旁路途径的激活不依赖抗原抗体复合物,通常是沉积于细胞表面的C3b与B因子结合,成为易于被血清中D因子分解的状态,在这个过程中B因子被分解成Ba和Bb;然后C3b和Bb组成复合物,成为旁路途径中的C3转化酶C3bBb;在该过程中,补体因子B在补体级联的旁路途径激活中起着早期和核心的作用。这里,C3b既是C3转化酶分解C3之后出现的产物,也是旁路途径C3转化酶的组成部分,由此形成了经典途径和旁路途径相互影响的一种反馈放大机制。当前研究发现血液性、自身免疫性、炎症性和神经变性等多种疾病和补体***功能异常相关。
阵发性睡眠性血红蛋白尿症(PNH)是一种持续溶血的慢性疾病,病因是由于1个或几个造血干细胞经获得性体细胞PIG-A基因突变造成的非恶性的克隆性疾病,属于超罕见血液疾病(Medicine(Baltimore)1997,76(2):63-93)。病程可表现为不同程度的溶血加重(阵发性),慢性或反复急性血管内溶血或随后的静脉/动脉血栓形成可导致进展性终末器官损伤和死亡,是一种典型的PNH以慢性血管内溶血,血红蛋白尿,及含铁血黄素尿为主要表现,但大多数患者常不典型,发病隐袭,病程迁延,病情轻重不一。
红细胞表面有十余种抑制补体通路活化的蛋白,均通过糖基化磷脂酰肌醇(GPI)锚定在其细胞膜上,统称为GPI-锚定蛋白(AP),目前认为,首先是造血干细胞在一定条件下 发生突变并产生糖基磷脂酰肌醇(GPI)缺陷的PNH克隆;然后,由于某些因素(现多认为是免疫因素),发生造血功能损伤或造血功能衰竭,PNH克隆获得增殖优势,超过正常克隆。GPI接连的多种抗原,也造成对PNH细胞生物学行为解释的复杂性,其中最重要的抑制补体通路活化的蛋白C3转化酶衰变加速因子CD55和膜攻击复合物(MAC)抑制因子CD59在发病机制、临床表现、诊断和治疗与PNH密切关注(Frontiers in Immunology 2019,10,1157)。CD59可以阻止C9掺入C5b-8复合物中,而阻止膜攻击单位形成,达到抑制补体终末攻击反应的作用。目前认为PNH的典型表现-血管内溶血和血栓是由于CD59缺乏所致。据报道先天性CD59缺乏症患者,其表现出众多PNH的典型表现,如血管内溶血、血红蛋白尿和静脉血栓等。在PNH患者中,由于GPI合成缺陷导致CD59不能结合到红细胞的细胞膜上,导致其抑制补体通路活化的功能丧失;因此补体通路异常活化的发生并对红细胞进行攻击,导致血管内溶血、血红蛋白尿,以及平滑肌功能障碍等多种临床表现。当前,临床除通过造血干细胞移植重建正常造血功能的治疗方案可治愈PNH外,尚无其它有效的治愈手段。由于造血干细胞移植存在一定风险,并且PNH为良性克隆性疾病,因此控制溶血发作仍然是临床治疗该病的主要策略。目前,仅有依库珠单抗(Eculizumab)被批准用于治疗PNH。然而,许多患者经用依库珠单抗治疗后仍然出现贫血现象,并且许多患者依然需要持续的输血。此外,在用药方式上依库珠单抗需要静脉注射。因此,存在着对开发补体途径的新型抑制剂的开发用于PNH的治疗的未满足需要。
IgAN为最常见的原发性肾小球肾炎,该疾病的特点是免疫荧光显示系膜区有IgA沉积;其临床表现多样,通常表现为反复发作的镜下或者肉眼血尿。现有资料表明IgAN的发生与先天或者获得性免疫调节异常有关。由于病毒、细菌和食物蛋白等对呼吸道或消化道的刺激作用,黏膜IgA1合成增多,或含IgA1的免疫复合物沉积于系膜区,并激活补体旁路途径,引起肾小球损伤。人类的IgA分子分为IgA1和IgA2这2种亚型,其中IgA1是健康个体血液循环的主要形式(约占85%),也是IgAN患者肾小球系膜区沉积的主要成分。IgA分子能够以单体和多聚体2种形式存在。IgA1分子在第一和第二恒定区之间具有独特的重链铰链区,可作为O-连接聚糖基团连接位点的结构域。近年研究发现,IgAN患者血清中及肾小球系膜区沉积的IgA分子主要为糖基化缺陷的IgA1(gd-IgA1)。目前认为IgAN发病机制的启动环节为gd-IgA1产生异常增多。
超过90%的IgAN患者肾小球系膜区伴有补体C3的沉积。75%-100%的IgAN患者肾组织内存在备解素和IgA、C3的共同沉积,30%-90%的IgAN患者肾组织存在补体因子H、IgA、C3的共同沉积。除肾组织内的沉积外,一些研究还发现IgAN患者的血浆中补体旁路途径的标志物水平也与IgAN的活动度有关(J Nephrol 2013,26(4):708-715)。一 项研究证实,肾组织和尿液中C3a以及肾组织中C3a受体与肾脏损害的活动性和严重程度显著相关(J clin Immunol 2014,34(2):224-232)。另有研究证实,在体外条件下IgA能够激活补体旁路途径。在这一过程中,IgA铰链区异常并不起到决定性的作用,而IgA多聚体的形成则是其关键环节(Eur J Immunol 1987,17(3):321-326)。当前,补体C3沉积于肾小球系膜区已经成为IgAN的一项辅助诊断标志。有研究对163例IgAN患者肾组织进行C3c和C3d免疫荧光检测,结果显示C3c沉积强度高于C3d沉积强度的IgAN患者表现为肾小球滤过率更低、肾小球毛细血管内增生的发生率更高、血尿也更严重,提示肾小球C3c沉积与IgAN的活动性病变有关(Am J Nephrol.2000,20(2):122-128)。当前临床上并没有特效药物治疗IgAN,主要为通用性药物肾素血管紧张素抑制剂(ACEI或ARB)、糖皮质激素和各种免疫抑制剂等。此外,这类药物带来的安全性也是一个不可忽视的问题,例如虽然糖皮质激素有降蛋白尿的作用,但STOP-IgAN试验和TESTING-I试验清楚地证实了糖皮质激素潜在的副作用,因此有必要和每个病人谈论糖皮质激素的风险和获益(IgA nephropathy 2019,95,4,750-756)。
与补体级联相关的其它疾病还包括膜性肾病(MN)、C3肾小球肾炎(C3G)、年龄相关性黄斑变性(AMD)、地图状萎缩(GA)、非典型溶血***综合征(aHUS)、溶血***综合征(HUS)、血液透析并发症、溶血性贫血或血液透析、神经脊髓炎(NMO)、关节炎、类风湿性关节炎、肝脏类炎症、皮肌炎和肌萎缩性侧索硬化、重症肌无力(MG)、呼吸***疾病和心血管等疾病。
目前,还没有用于临床治疗的补体因子B抑制剂的小分子药物,在发现和开发阶段有以下公开的描述:IONIS Pharmaceuticals Inc.开发的寡核苷酸类药物作为补体因子B(CFB)特异性抑制剂来治疗、预防或缓解与补体旁路途径失调相关的疾病(WO2015038939)。Novartis AG公司开发的小分子补体因子B抑制剂用于治疗年龄相关性黄斑变性(AMD)等疾病(WO2013164802,WO2013192345,WO2014143638,WO2015009616,WO2015066241),用于治疗C3G和IgAN等疾病(WO2019043609A1)。Achillion Pharmaceuticals Inc.开发的小分子补体因子B抑制剂用于治疗年龄相关性黄斑变性(AMD)等疾病(WO2018005552)。
炎症和免疫性相关的疾病具有多样性、难治愈的特点;PNH疾病上市的药物仅有依库珠单抗,但是由于价格,给患者带来了沉重的负担;同时,许多患者经用依库珠单抗治疗后仍然出现贫血现象,并且许多患者依然需要持续的输血;此外,在用药方式上依库珠单抗需要静脉注射。而一些疾病截至目前并没有特效的治疗药物,比如IgAN等。在这些领域有尚未满足的临床需求,需要开发新的小分子药物用于医学治疗。
发明内容
本发明所要解决的技术问题是为了克服现有技术中作为补体因子B抑制剂的小分子化合物缺乏的缺陷,而提供了一种杂环类化合物、其制备方法及用途。本发明提供的杂环类化合物可作为补体因子B抑制剂,并用于制备治疗与补体***的异常激活、或者与补体***执行正常功能的中出现的相关疾病的药物;可作为炎症和免疫性相关的疾病的治疗剂有望满足临床需要。
本发明是通过下述技术方案来解决上述技术问题的。
本发明提供了一种如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,其任选在药学上可接受的载体中;
Figure PCTCN2021110751-appb-000001
其中,
W为O或C(R 7’R 7”);
R 7、R 7’和R 7”独立地为氢、羟基、卤素、C 1-C 4烷基或C 1-C 4烷基-O-;
R 6为氢、C 1-C 4烷基或羟基C 1-C 4烷基;
R 4’和R 4独立地为氢;
m为0、1或2;
R 5
Figure PCTCN2021110751-appb-000002
环B为苯基或6元杂芳基;所述的6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;
R b为H、羟基、=O,或,与环B并环连接的:苯基、3-6元环烷基、5-6元杂环烷基或5-6元杂芳基;所述5-6元杂环烷基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;当取代基为多个时,相同或不同;
A为
Figure PCTCN2021110751-appb-000003
Z 1为C(R 21)或N;Z 2为C(R 51)或N;R 41为NH 2或C(=O)NH 2
环A 1为吡啶基;其中,Z 3为C(R 22)或N;Z 4和Z 5独立地为C或N;(其余为所示的C;)
R 21、R 22和R 51独立地为氢;
环A 2为5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基,或者,被一个或多个R a1取代的:5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基;所述的5-6元杂环烷基和被一个或多个R a1取代的5-6元杂环烷基里的5-6元杂环烷基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂环烯基和被一个或多个R a1取代的5-6元杂环烯基里的5-6元杂环烯基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂芳基和被一个或多个R a1取代的5-6元杂芳基里的5-6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;当取代基为多个时,相同或不同;
环A 3为5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
Figure PCTCN2021110751-appb-000004
(a端表示与苯环并环连接),或者,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
Figure PCTCN2021110751-appb-000005
所述的5-6元杂环烷基和被一个或多个R a2取代的5-6元杂环烷基里的5-6元杂环烷基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂环烯基和被一个或多个R a2取代的5-6元杂环烯基里的5-6元杂环烯基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的6元杂芳基和被一个或多个R a2取代的5-6元杂芳基里的6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;当取代基为多个时,相同或不同;环A 3与苯环并环连接;
A 3’为5元杂芳基;所述的5元杂芳基中,杂原子个数为1或2个,杂原子选自N、O和S中的一种或多种;且Z 7为N、O或S,和/或,Z 6为CH、O或S;
R a1和R a2独立地为羟基、=O、卤素、CN、C 1-C 4烷基或C 1-C 4烷基-O-;
R 11、R 31、R 12、R 32、R 13和R 33独立地为C 1-C 4烷基、C 1-C 4烷基-O-或3-6元环烷基;
Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基;
带“*”碳原子表示当为手性碳原子时,为S构型、R构型或它们的混合物。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如 下定义(未提及的基团同本申请任一方案所述),
W为C(R 7’R 7”)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 7’和R 7”独立地为氢、卤素、C 1-C 4烷基或C 1-C 4烷基-O-;例如,R 7”独立地为氢、C 1-C 4烷基-O-或卤素;R 7’独立地为氢或C 1-C 4烷基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 7为氢。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
m为1。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R b为H。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
Z 1为CH、Z 2为N,或Z 1为CH、Z 2为CH,或Z 1为N、Z 2为CH。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
Z 4为N,或Z 5为N。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
环A 2为5-6元杂芳基,例如5元杂芳基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
环A 2为5-6元杂环烷基、5-6元杂环烯基或6元杂芳基,或,被一个或多个R a1取代的:5-6元环烯基、5-6元杂环烷基、5-6元杂环烯基或6元杂芳基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
环A 3为5-6元杂环烷基、5-6元杂环烯基或
Figure PCTCN2021110751-appb-000006
或,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
Figure PCTCN2021110751-appb-000007
例如5元杂环烷基、5元杂环烯基或
Figure PCTCN2021110751-appb-000008
或,被一个或多个R a2取代的:5元杂环烷基、5元杂环烯基或
Figure PCTCN2021110751-appb-000009
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R a1和R a2独立地为羟基、卤素、=O或C 1-C 4烷基;例如羟基、=O或C 1-C 4烷基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 11、R 12和R 13独立地为C 1-C 4烷基或C 1-C 4烷基-O-,优选C 1-C 4烷基-O-。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 31、R 32和R 33独立地为C 1-C 4烷基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 14为C 1-C 4烷基-O-。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
Z 8为N,R 34为C 1-C 4烷基;或Z 8为CH,R 34为3-6元环烷基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),所述的如式I所示的杂环类化合物为如下式Ia、Ib或Ic所示;
Figure PCTCN2021110751-appb-000010
例如,R 7”独立地为氢、C 1-C 4烷基-O-或卤素;R 7’独立地为氢或C 1-C 4烷基;
又例如
Figure PCTCN2021110751-appb-000011
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
W为C(R 7’R 7”);R 7为氢;
R 7’和R 7”独立地为氢、卤素、C 1-C 4烷基或C 1-C 4烷基-O-;例如,R 7”独立地为氢、C 1-C 4烷基-O-或卤素;R 7’独立地为氢或C 1-C 4烷基;
R 6为氢;
R 4’和R 4独立地为氢;
m为1;
R 5
Figure PCTCN2021110751-appb-000012
环B为苯基或6元杂芳基;
R b为H、羟基或=O,或,与环B并环连接的:苯基、3-6元环烷基、5-6元杂环烷基或5-6元杂芳基;
A为
Figure PCTCN2021110751-appb-000013
Z 1为CH或N;Z 2为CH或N;R 41为NH 2或C(=O)NH 2
环A 1为吡啶基;Z 3为CH或N;Z 4和Z 5独立地为C或N;
环A 2为5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基,或者,被一个或多个R a1取代的:5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基;
环A 3为5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
Figure PCTCN2021110751-appb-000014
或者,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
Figure PCTCN2021110751-appb-000015
R a1和R a2独立地为羟基、卤素、=O或C 1-C 4烷基;例如羟基、=O或C 1-C 4烷基;
R 11、R 31、R 12、R 32、R 13、R 23和R 33独立地为C 1-C 4烷基、C 1-C 4烷基-O-或3-6元环烷基;
Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基;
带“*”碳原子表示当为手性碳原子时,为S构型、R构型或它们的混合物。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
W为C(R 7’R 7”);R 7为氢;
R 7’和R 7”独立地为氢或C 1-C 4烷基-O-;
R 6为氢;
R 4’和R 4独立地为氢;
m为1;
R 5
Figure PCTCN2021110751-appb-000016
A为
Figure PCTCN2021110751-appb-000017
环A 1为吡啶基;Z 3为N;Z 4和Z 5为C;
环A 2为5杂芳基或被一个或多个R a1取代的5杂芳基;例如5杂芳基;
环A 3为5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
Figure PCTCN2021110751-appb-000018
或者,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
Figure PCTCN2021110751-appb-000019
例如5 元杂环烷基、5元杂环烯基或
Figure PCTCN2021110751-appb-000020
R a1和R a2独立地为羟基、=O或C 1-C 4烷基;例如=O或C 1-C 4烷基;
R 12和R 13独立地为C 1-C 4烷基-O-;
R 32和R 33独立地为C 1-C 4烷基;
Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基;
带“*”碳原子表示当为手性碳原子时,为S构型、R构型或它们的混合物。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
W为C(R 7’R 7”);R 7为氢;
R 7’和R 7”独立地为氢或C 1-C 4烷基-O-;
R 6为氢;
R 4’和R 4独立地为氢;
m为1;
R 5
Figure PCTCN2021110751-appb-000021
A为
Figure PCTCN2021110751-appb-000022
环A 3为5元杂环烷基、5元杂环烯基或
Figure PCTCN2021110751-appb-000023
R 13为C 1-C 4烷基-O-;
R 33为C 1-C 4烷基;
Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基;
带“*”碳原子表示当为手性碳原子时,为S构型、R构型或它们的混合物。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R 7、R 7’和R 7”独立地为C 1-C 4烷基或C 1-C 4烷基-O-时,所述的C 1-C 4烷基或C 1-C 4 烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R 6为C 1-C 4烷基或羟基C 1-C 4烷基时,所述的C 1-C 4烷基和羟基C 1-C 4烷基里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当环B为6元杂芳基时,所述的6元杂芳基为吡啶基;例如
Figure PCTCN2021110751-appb-000024
(b端表示与COOH连接)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R b为3-6元环烷基时,所述的3-6元环烷基为环丙基、环丁基、环戊基和环己基;例如环戊基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R b为5-6元杂环烷基时,所述的5-6元杂环烷基为四氢呋喃基;例如
Figure PCTCN2021110751-appb-000025
(c端表示与环B并环连接)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R b为5-6元杂芳基时,所述的5-6元杂芳基为吡啶基;例如
Figure PCTCN2021110751-appb-000026
(c端表示与环B并环连接)或咪唑基(例如
Figure PCTCN2021110751-appb-000027
c端表示与环B并环连接)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当环A 2为5-6元杂环烷基时,所述的5-6元杂环烷基为
Figure PCTCN2021110751-appb-000028
Figure PCTCN2021110751-appb-000029
Figure PCTCN2021110751-appb-000030
(
Figure PCTCN2021110751-appb-000031
表示与环A 1并环连接的位置)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当环A 2为5-6元杂环烯基时,所述的5-6元杂环烯基为
Figure PCTCN2021110751-appb-000032
Figure PCTCN2021110751-appb-000033
Figure PCTCN2021110751-appb-000034
(
Figure PCTCN2021110751-appb-000035
表示与环A 1并环连接的位置,
Figure PCTCN2021110751-appb-000036
表示单键或双键)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当环A 2为5-6元杂芳基时,所述的5-6元杂芳基为
Figure PCTCN2021110751-appb-000037
Figure PCTCN2021110751-appb-000038
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如 下定义(未提及的基团同本申请任一方案所述),
当环A 3为5-6元杂环烷基时,所述的5-6元杂环烷基为
Figure PCTCN2021110751-appb-000039
Figure PCTCN2021110751-appb-000040
Figure PCTCN2021110751-appb-000041
(
Figure PCTCN2021110751-appb-000042
表示与环A 1并环连接的位置)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当环A 3为5-6元杂环烯基时,所述的5-6元杂环烯基为
Figure PCTCN2021110751-appb-000043
Figure PCTCN2021110751-appb-000044
Figure PCTCN2021110751-appb-000045
(
Figure PCTCN2021110751-appb-000046
表示与环A 1并环连接的位置,
Figure PCTCN2021110751-appb-000047
表示单键或双键)。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当环A 3为6元杂芳基时,所述的6元杂芳基为
Figure PCTCN2021110751-appb-000048
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当环A 3
Figure PCTCN2021110751-appb-000049
所述的A 3’
Figure PCTCN2021110751-appb-000050
Figure PCTCN2021110751-appb-000051
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R a1和R a2独立地为C 1-C 4烷基或C 1-C 4烷基-O-时,所述的C 1-C 4烷基或C 1-C 4烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R 11、R 31、R 12、R 32、R 13、R 23和R 33独立地为C 1-C 4烷基、C 1-C 4烷基-O-时,所述的C 1-C 4烷基和C 1-C 4烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R 11、R 31、R 12、R 32、R 13、R 23和R 33独立地为3-6元环烷基时,所述的3-6元环烷基为环丙基、环丁基、环戊基和环己基;例如环丙基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R 14为C 1-C 4烷基-O-时,所述的C 1-C 4烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R 34为C 1-C 4烷基时,所述的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当R 34为3-6元环烷基时,所述的3-6元环烷基为环丙基、环丁基、环戊基和环己基;例如环丙基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 7’和R 7”独立地为氢、F、甲基、乙基-O-。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 5
Figure PCTCN2021110751-appb-000052
Figure PCTCN2021110751-appb-000053
例如
Figure PCTCN2021110751-appb-000054
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R a1和R a2独立地为羟基、=O或甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 11、R 31、R 12、R 32、R 13和R 33独立地为甲基、甲基-O-或环丙基;
例如R 11、R 12和R 13独立地为甲基、甲基-O-或环丙基;
R 31、R 32和R 33独立地为甲基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 14为甲基-O-。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
R 34为甲基或环丙基。
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
W为
Figure PCTCN2021110751-appb-000055
亚甲基;例如
Figure PCTCN2021110751-appb-000056
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
Figure PCTCN2021110751-appb-000057
Figure PCTCN2021110751-appb-000058
Figure PCTCN2021110751-appb-000059
Figure PCTCN2021110751-appb-000060
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当A为
Figure PCTCN2021110751-appb-000061
时,例如,
Figure PCTCN2021110751-appb-000062
Figure PCTCN2021110751-appb-000063
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当A为
Figure PCTCN2021110751-appb-000064
时,例如,
Figure PCTCN2021110751-appb-000065
Figure PCTCN2021110751-appb-000066
例如
Figure PCTCN2021110751-appb-000067
Figure PCTCN2021110751-appb-000068
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当A为
Figure PCTCN2021110751-appb-000069
时,例如,
Figure PCTCN2021110751-appb-000070
Figure PCTCN2021110751-appb-000071
Figure PCTCN2021110751-appb-000072
例如
Figure PCTCN2021110751-appb-000073
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物中的某些基团如下定义(未提及的基团同本申请任一方案所述),
当A为
Figure PCTCN2021110751-appb-000074
时,例如,
Figure PCTCN2021110751-appb-000075
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物为如下任一结构,
Figure PCTCN2021110751-appb-000076
Figure PCTCN2021110751-appb-000077
Figure PCTCN2021110751-appb-000078
Figure PCTCN2021110751-appb-000079
Figure PCTCN2021110751-appb-000080
Figure PCTCN2021110751-appb-000081
在本发明某些优选实施方案中,所述的如式I所示的杂环类化合物的药学上可接受的盐为如下任一结构,
Figure PCTCN2021110751-appb-000082
Figure PCTCN2021110751-appb-000083
本发明中,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药具有一个或多个手性碳原子,因此可以分离得到光学纯度异构体,例如纯的对映异构体,或者外消旋体,或者混合异构体。可以通过本领域的分离方法来获得纯的单一异构体,如手性结晶成盐,或者手性制备柱分离得到。
本发明中,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药如存在立体异构体,则可以以单一的立体异构体或它们的混合物(例如外消旋体)的形式存在。术语“立体异构体”是指顺反异构体或旋光异构体。这些立体异构体可以通过不对称合成方法或手性分离法(包括但不限于薄层色谱、旋转色谱、柱色谱、气相色谱、高压液相色谱等)分离、纯化及富集,还可以通过与其它手性化合物成键(化学结合等)或成盐(物理结合等)等方式进行手性拆分获得。术语“单一的立体异构体”是指本发明化合物的一种立体异构体相对于该化合物的所有立体异构体的质量含量不低于95%。
本发明中,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药如存在互变异构体,则可以以单一的互变异构体或它们的混合物的形式存在,较佳地以较稳定的互变异构体为主的形式存在。例如,当含如下结构片段时:
Figure PCTCN2021110751-appb-000084
Figure PCTCN2021110751-appb-000085
本发明中,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药可通过包括与化学领域公知方法相似的方法合成,其步骤和条件可参考本领域类似反应的步骤和条件,特别是根据本文说明进行合成。起始原料通常是来自商业来源,例如Aldrich或可使用本领域技术人员公知的方法(通过SciFinder、Reaxys联机数据库得到)容易地制备。
本发明中,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似 物、前药,也可以通过已制备得到的所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,采用本领域常规方法,经外周修饰进而得到其他所述的如式I所示的杂环类化合物或其药学上可接受的盐。
用于制备如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药的必要原料或试剂可以商购获得,或者通过本领域已知的合成方法制备。如下实验部分所描述的方法,可以制备游离碱或者其加酸所成盐的本发明的化合物。术语药学上可接受的盐指的是本文所定义的药学上可接受的盐,并且具有母体化合物所有的作用。药学上可接受的盐可以通过在有机碱的合适的有机溶剂中加入相应的酸,根据常规方法处理来制备药学上可接受的盐。
本发明还提供了一种如上所述的如式I所示的杂环类化合物的制备方法,其包括如下步骤:
在溶剂中,在碱存在下,将如式II所示的化合物进行如下所示的脱酯基反应,得到所述的如式I所示的杂环类化合物即可;
Figure PCTCN2021110751-appb-000086
其中,R 8为C 1-C 4的烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基或乙基);R b、R 4、R 4’、R 6、R 7、A、W、m和*的定义均如上所示。
所述的如式I所示的杂环类化合物的制备方法中,所述的脱酯基反应的条件和操作可为本领域该类反应中常规的条件和操作;本发明中优选如下:所述的溶剂可为水和/或醇类溶剂(例如,甲醇和/或乙醇),例如水和醇类溶剂。
所述的溶剂的用量以不影响反应即可,例如所述的如式II所示的化合物与所述的溶剂的质量体积比为1g/L-20g/L(例如15g/L-20g/L)。
所述的碱可为碱金属氢氧化物,例如氢氧化钠和/或氢氧化钾。
所述的脱酯基反应的温度可为0-100℃(例如70-80℃)。
所示的脱酯基反应的进程可采用本领域中的常规监测方法(例如TLC、HPLC或NMR)进行检测,一般以所述的如式II所示的化合物消失或不再反应时作为反应终点。
本发明还提供了一种如上所述的如式II所示的杂环类化合物,
Figure PCTCN2021110751-appb-000087
其中,R 8为C 1-C 4的烷基(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基或乙基);R b、R 4、R 4’、R 6、R 7、A、W、m和*的定义均如上所示。
在本发明的某一方案中,所述的如式I所示的杂环类化合物为如下结构:
Figure PCTCN2021110751-appb-000088
本发明提供了一种药物组合物,其包括如上所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,和一种或多种药学上可接受的载体。在所述的药物组合物中,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药的用量可为治疗有效量。
所述的药学上可接受的载体(药用辅料)可为药物生产领域中广泛采用的那些辅料。辅料主要用于提供一个安全、稳定和功能性的药物组合物,还可以提供方法,使受试者接受给药后活性成分以所期望速率溶出,或促进受试者接受组合物给药后活性成分得到有效吸收。所述的药用辅料可以是惰性填充剂,或者提供某种功能,例如稳定该组合物的整体pH值或防止组合物活性成分的降解。所述的药用辅料可以包括下列辅料中的一种或多种:粘合剂、助悬剂、乳化剂、稀释剂、填充剂、成粒剂、胶粘剂、崩解剂、润滑剂、抗粘着剂、助流剂、润湿剂、胶凝剂、吸收延迟剂、溶解抑制剂、增强剂、吸附剂、缓冲剂、螯合剂、防腐剂、着色剂、矫味剂和甜味剂。
本发明的药物组合物可根据公开的内容使用本领域技术人员已知的任何方法来制备。例如,常规混合、溶解、造粒、乳化、磨细、包封、包埋或冻干工艺。
本发明所述的药物组合物可以任何形式给药,包括注射(静脉内)、粘膜、口服(固体和液体制剂)、吸入、眼部、直肠、局部或胃肠外(输注、注射、植入、皮下、静脉内、 动脉内、肌内)给药。本发明的药物组合物还可以是控释或延迟释放剂型(例如脂质体或微球)。固体口服制剂的实例包括但不限于粉末、胶囊、囊片、软胶囊剂和片剂。口服或粘膜给药的液体制剂实例包括但不限于悬浮液、乳液、酏剂和溶液。局部用制剂的实例包括但不限于乳剂、凝胶剂、软膏剂、乳膏剂、贴剂、糊剂、泡沫剂、洗剂、滴剂或血清制剂。胃肠外给药的制剂实例包括但不限于注射用溶液、可以溶解或悬浮在药学上可接受载体中的干制剂、注射用悬浮液和注射用乳剂。所述的药物组合物的其它合适制剂的实例包括但不限于滴眼液和其他眼科制剂;气雾剂:如鼻腔喷雾剂或吸入剂;适于胃肠外给药的液体剂型;栓剂以及锭剂。
本发明的一个目的是提供一种如上所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药在作为补体因子B抑制剂中的应用。
在所述的应用中,所述的补体因子B抑制剂可用于哺乳动物生物体内;也可用于生物体外,主要作为实验用途,例如:作为标准样或对照样提供比对,或按照本领域常规方法制成试剂盒,为补体因子B的抑制效果提供快速检测。
本发明的一个目的是提供一种如上所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药在制备药物中的应用;所述的药物可为用于治疗与补体因子B的异常激活、或者与补体因子B执行正常功能的中出现的相关疾病的药物;或者,所述的药物可为用于治疗血液性、自身免疫性、炎性和神经变性等疾病的药物。所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药可为治疗有效量的。
本发明的一个目的是提供一种如上所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药在治疗与补体***的异常激活、或者与补体***执行正常功能的中出现的相关疾病中的应用。
本发明的一个目的是提供一种治疗与补体***的异常激活、或者与补体***执行正常功能的中出现的相关疾病的方法;其包括向患者施用有效剂量的如上所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药。
如上所述的与补体***的异常激活、或者与补体***执行正常功能的中出现的相关疾病可为血液性、自身免疫性、炎性和神经变性等疾病;所述的疾病包括但不限于:阵发性睡眠性血红蛋白尿症(PNH)、原发性肾小球肾炎(IgAN)、膜性肾病(MN)、C3肾小球肾炎(C3G)、年龄相关性黄斑变性(AMD)、地图状萎缩(GA)、非典型溶血***综合征(aHUS)、溶血***综合征(HUS)、血液透析并发症、溶血性贫血或血液透析、神经脊髓炎(NMO)、关节炎、类风湿性关节炎、肝脏类炎症、皮肌炎和肌萎缩性侧索硬化、重 症肌无力(MG)、呼吸***疾病和心血管等疾病。
所述的补体***较佳地为由补体因子B相关联或调节的补体***。
当用作药物时,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药可以以药物组合物的形式给药。这些组合物可根据药学领域熟知的方法制备,可以各种途径施用,视需要局部或***性治疗和要治疗的区域而定。给予可以是局部(包括表皮和透皮,眼部和粘膜,包括鼻内,***和直肠递送),肺(例如,通过粉末或气溶胶吸入或吹入,包括通过喷雾器;气管内或鼻内),口服或胃肠外给予形式。口服给药可以包括配制为每日一次或每日两次(BID)给药的剂型。胃肠外给药包括静脉内、动脉内、皮下、腹膜内肌肉内或注射或输液;或颅内如鞘内或心室内给药。胃肠外给药可以单次推注剂量形式,或可以是通过连续灌注泵。外用给药的药物组合物和制剂可包括透皮贴片、油膏剂、乳液、软膏剂、凝胶、滴剂、栓剂、喷剂、液体和粉末。常规药学载体、水、粉末或油状基底、增稠剂等可能是必须或需要的。
如本文所用,术语“治疗”是指治疗性或缓解性措施。有益的或期望的临床结果包括但不限于:全部或部分减轻与疾病或紊乱或病症有关的症状,减小疾病程度,稳定(即不恶化)疾病状态,延迟或减缓疾病进展,缓解或减轻疾病状态(例如,疾病的一种或多种症状),以及可检测或不可检测的缓解(无论是部分还是全部)。“治疗”也可以指与不接受治疗的期望存活相比延长生存期。
在某些实施方式中,所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药或者如上所述的药物组合物可用于预防如本文所限定的疾病和病症(例如,血液性、自身免疫性、炎性和神经变性等疾病)。本文使用的术语“预防”意指全部或部分预防本文所述的疾病或病症或其症状的发作、复发或扩散。
如本文所用,术语“前药”,代表一个化合物在体内转化为式I所示的化合物。这样的转化受前体药物在血液中水解或在血液或组织中经酶转化为母体结构的影响。本发明前体药物类化合物可以是酯,在现有的发明中酯可以作为前体药物的有苯酯类,脂肪族(C1-24)酯类,酰氧基甲基酯类,碳酸酯,氨基甲酸酯类和氨基酸酯类。例如本发明里的一个化合物包含羟基,即可以将其酰化得到前体药物形式的化合物。其他的前体药物形式包括磷酸酯,如这些磷酸酯类化合物是经母体上的羟基磷酸化得到的。关于前体药物完整的讨论可以参考以下文献:T.Higuchiand V.Stella,Pro-drugsas Novel Delivery Systems,Vol.14 of the A.C.S.Symposium Series,Edward B.Roche,ed.,Bioreversible Carriersin Drug Design,American Pharmaceutical Associationand Pergamon Press,1987,J.Rautioetal.,Prodrugs:Designand Clinical Applications,Nature Review Drug Discovery,2008,7,255-270, and S.J.Heckeretal.,Prodrugs of Phosphatesand Phosphonates,Journal of Medicinal Chemistry,2008,51,2328-2345。
式I化合物和其盐意图涵盖式I化合物或其盐的任何同位素标记的(或“放射性标记的”)衍生物。这种衍生物是如下的式I化合物或其盐的衍生物,其中一个或多个原子被原子质量或质量数不同于通常在自然界中所发现的原子质量或质量数的原子置换。所使用的放射性核素将取决于该放射性标记的衍生物的具体应用。举例来说,对于体外受体标记和竞争测定, 3H或 14C常常是有用的。对于放射成像应用, 11C或 18F常常是有用的。
本发明化合物的特定同位素变体,特别是其中已经结合一种或多种放射性同位素的同位素变体,可有益于例如考察作用机制或在体内的活性组分分布;由于相对容易的可制备性和可检测性,标记有 3H或 14C同位素的化合物特别适用于此目的。另外,纳入同位素如氘,由于该化合物具有更好的代谢稳定性,例如延长体内的半衰期或降低所需的有效剂量,可产生特别的治疗益处;因此本发明化合物的这种修饰还可在一些情况下构成本发明的优选实施方案。本发明化合物的同位素变体可通过本领域技术人员已知的方法,例如通过以下描述的方法及操作实施例中描述的方法,通过使用相应的同位素修饰的特定试剂和/或起始化合物来制备。
术语“药用辅料”或“赋形剂”是指药学上可接受的化学物质,例如药学领域的普通技术人员已知的用于帮助给予药用的试剂。它是可以用于制备药物组分的化合物,通常是安全的、无毒的,且是生物学或者其它方面所不可期望的,其包括对于兽用和人用药物可接受的赋形剂。通常的赋形剂包括粘合剂、表面活性剂、稀释剂、崩解剂和润滑剂。
除非另外说明,应当应用本文所使用的下列定义。出于本发明的目的,化学元素与元素周期表CAS版,和《化学和物理手册》,第75版,1994一致。此外,有机化学一般原理可参考"Organic Chemistry",Thomas Sorrell,University Science Books,Sausalito:1999,和"March's Advanced Organic Chemistry”by Michael B.Smith and Jerry March,John Wiley&Sons,New York:2007中的描述,其全部内容通过引用并入本文。
在本说明书中,可由本领域技术人员选择基团及其取代基以提供稳定的结构部分和化合物。当通过从左向右书写的常规化学式描述取代基时,该取代基也同样包括从右向左书写结构式时所得到的在化学上等同的取代基。
在本文中定义的某些化学基团前面通过简化符号来表示该基团中存在的碳原子总数。例如,C 1-C 6烷基是指具有总共1、2、3、4、5或6个碳原子的如下文所定义的烷基。简化符号中的碳原子总数不包括可能存在于所述基团的取代基中的碳。
在本文中,取代基中定义的数值范围如0至4、1-4、1至3等表明该范围内的整数, 如1-6为1、2、3、4、5、6。
除前述以外,当用于本申请的说明书及权利要求书中时,除非另外特别指明,否则以下术语具有如下所示的含义。
术语“一种(个)或多种(个)”或“一种(个)或两种(个)以上”是指即1、2、3、4、5、6、7、8、9或更多。
术语“包括”为开放式表达,即包括本发明所指明的内容,但并不排除其他方面的内容。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。
一般而言,术语“取代的”表示所给结构中的一个或多个氢原子被具体取代基所取代。进一步地,当该基团被1个以上所述取代基取代时,所述取代基之间是相互独立,即,所述的1个以上的取代基可以是互不相同的,也可以是相同的。除非其他方面表明,一个取代基团可以在被取代基团的各个可取代的位置进行取代。当所给出的结构式中不只一个位置能被选自具体基团的一个或多个取代基所取代,那么取代基可以相同或不同地在各个位置取代。
在本说明书的各部分,本发明公开化合物的取代基按照基团种类或范围公开。特别指出,本发明包括这些基团种类和范围的各个成员的每一个独立的次级组合。术语“C x-C y烷基"是指含有x至y个碳原子的直链或支链饱和烃。例如,术语“C 1~C 6烷基”或“C 1- 6烷基”特别指独立公开的甲基、乙基、C 3烷基、C 4烷基、C 5烷基和C 6烷基;“C 1- 4烷基”特指独立公开的甲基、乙基、C 3烷基(即丙基,包括正丙基和异丙基)、C 4烷基(即丁基,包括正丁基、异丁基、仲丁基和叔丁基)。
术语“卤素”选自于F,Cl,Br或I,尤其指F或Cl。
术语“烷氧基”是指基团-O-R X,其中,R X为如上文所定义的烷基。
本文所用术语“部分”、“结构部分”、“化学部分”、“基团”、“化学基团”是指分子中的特定片段或官能团。化学部分通常被认为是嵌入或附加到分子上的化学实体。
当所列举的取代基中没有指明其通过哪一个原子连接到化学结构通式中包括但未具体提及的化合物时,这种取代基可以通过其任何原子相键合。取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当所列举的基团中没有明确指明其具有取代基时,这种基团仅指未被取代。例如当“C 1~C 4烷基”前没有“取代或未取代的”的限定时,仅指“C 1~C 4烷基”本身或“未取代的C 1~C 4烷基”。
在本发明的各部分,描述了连接取代基。当该结构清楚地需要连接基团时,针对该基团所列举的马库什变量应理解为连接基团。例如,如果该结构需要连接基团并且针对该变量的马库什基团定义列举了“烷基”,则应该理解,该“烷基”代表连接的亚烷基基团。
在一些具体的结构中,当烷基基团清楚地表示为连接基团时,则该烷基基团代表连接的亚烷基基团,例如,基团“卤代-C 1~C 6烷基”中的C 1-C 6烷基应当理解为C 1~C 6亚烷基。
术语“亚烷基”表示从饱和的直链或支链烃基中去掉两个氢原子所得到的饱和的二价烃基基团。亚烷基基团的实例包括亚甲基(-CH 2-),亚乙基{包括-CH 2CH 2-或-CH(CH 3)-},亚异丙基{包括-CH(CH 3)CH 2-或-C(CH 3) 2-}等等。
在本申请中,作为基团或是其它基团的一部分(例如用在卤素取代的烷基等基团中),术语“烷基”意指包括具有指定碳原子数目的支链和直链的饱和脂族烃基;比如含有1至20个碳原子的直链或支链饱和烃链;又例如,C 1-C 6的烷基。如在“C 1~C 6烷基”中定义为包括在直链或者支链结构中具有1、2、3、4、5、或者6个碳原子的基团。其中,丙基为C 3烷基(包括同分异构体,例如正丙基或异丙基);丁基为C 4烷基(包括同分异构体,例如正丁基、仲丁基、异丁基或叔丁基);戊基为C 5烷基(包括同分异构体,例如正戊基、1-甲基-丁基、1-乙基-丙基、2-甲基-1-丁基、3-甲基-1-丁基、异戊基、叔戊基或新戊基);己基为C 6烷基(包括同分异构体,例如正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基)。
术语“环烷基”,指仅由碳原子和氢原子组成的饱和的单环或者多环碳环取代基,且其可经由任何适宜的碳原子通过单键与分子的其余部分连接;当为多环时,可为并环连接、桥环连接或螺环连接(即,碳原子上的两个偕氢被亚烷基取代)的并环体系、桥环体系或螺环体系。在某一方案中,典型的单环环烷基,如环丙基、环丁基、环戊基、环己基或环庚基。
在本申请中,本身或者作为另一取代基的一部分,术语“环烯基”是指含有部分不饱和双键的单环、多环或者桥接碳环取代基,且其可经由任何适宜的碳原子通过单键与分子的其余部分连接;当为多环时,可为并环连接或螺环连接(即,碳原子上的两个偕氢被亚烷基取代)的桥环体系或螺环体系。在一些实施例中,“环烯基”是具有5至6个环原子的单环的,不饱和的碳环烯基基团(“5-6元环烯基”)。该术语包括但不限于环戊烯基(例如
Figure PCTCN2021110751-appb-000089
)、环戊二烯基(例如
Figure PCTCN2021110751-appb-000090
)、环己烯基(例如
Figure PCTCN2021110751-appb-000091
)或环己二烯基,以 及其立体异构体。
术语“杂环烷基”,指具有杂原子的饱和的环状基团,含1个或多个独立选自N、O、S、S(=O)和S(=O) 2的杂原子,其余为碳组成的稳定的3-10元饱和杂环***的基团。除非本说明书中另外特别指明,否则杂环烷基基团可以是单环的(“单环的杂环烷基”),或者是双环、三环或更多环的环体系,其可包括并环的(稠合的)、桥联的(桥环的)或螺的环***(例如二环***(“二环的杂环烷基”)。杂环烷基二环的环***可以在一个或两个环中包括一个或多个杂原子;并且是饱和的。在一些实施例中,“杂环烷基”是5-6元杂环烷基。示例性3-元杂环基基团包括但不限于,氮杂环丙基、环氧乙烷基以及硫杂环丙烷基,或者其立体异构体;示例性4-元杂环基基团包括但不限于,氮杂环丁烷基,环氧丙烷基,硫杂环丁烷基,或者其同分异构体和立体异构体;示例性5-元杂环基基团包括但不限于,四氢呋喃基,四氢噻吩基,吡咯烷基,噻唑烷基,异噻唑烷基,噁唑烷基,异噁唑烷基,咪唑烷基,吡唑烷基,二氧戊环基,氧杂硫呋喃基,二硫呋喃基,或者其同分异构体和立体异构体。示例性6-元杂环基基团包括但不限于,哌啶基,四氢吡喃基,硫化环戊烷基,吗啉基,硫代吗啉基,二噻烷基,二噁烷基,哌嗪基,三嗪烷基,或者其同分异构体和立体异构体。
在本申请中,本身或者作为另一取代基的一部分,除非另有规定,术语“杂环稀基”表示通过杂原子或杂原子团连接的环状稀基。由此,“杂环稀基”包含以上“杂”和环稀基的定义。在一些实施例中,在某一实施例方案中,所述的“杂环稀基”为由2-9个碳原子以及含有1、2、3或4个选自N、O、S、S(=O)或S(=O) 2杂原子或含杂原子的基团组成的稳定的3-10元含不饱和双键的杂环***的基团。非本说明书中另外特别指明,否则杂环稀基基团或者可以是单环的(“单环的杂环稀基”),或者是双环、三环或更多环的环体系,其可包括融合的、桥联的或螺的环***(例如二环***(“二环的杂环稀基”)。杂环稀基二环的环***可以在一个或两个环中包括一个或多个杂原子。)并且是含不饱和双键的。杂环稀基可以经由碳原子并通过单键与分子其余部分连接;在包含一个或多个氮原子的杂环稀基基团中,连接点可以是碳或氮原子;或者,与分子其余部分并环连接;只要化合价许可。
术语“芳基”,指具有完全共轭的π电子体系的全碳芳香基团,可为单环或稠合环,通常具有6-14个碳原子,优选具有6-12个碳原子,最优选具有6个碳原子。芳基的实例包括但不限于苯基、萘基和蒽基。
术语“杂芳基”是指含有杂原子的芳香基团,可为单环或稠合环,优选含有1-4个独立选自N、O和S的5-12元杂芳基,包括但不限于吡咯基、呋喃基、噻吩基、吲哚基、咪 唑基、噁唑基、异噁唑基、吡唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、喹啉基、异喹啉基、(苯并)噁唑基、(苯并)呋喃基、(苯并)噻吩基、(苯并)噻唑基、***基。在某一方案中,典型地含1个或多个独立选自N、O和S的杂原子的5-6元单环杂芳基。在某一方案中,“杂芳基”为5-6元杂芳基,其中杂原子选自N、O和S中的一种或多种,杂原子数为1、2或3个。
除非另有规定,本文使用的所有技术术语和科学术语具有要求保护主题所属领域的标准含义。倘若对于某术语存在多个定义,则以本文定义为准。
应该理解,在本发明中使用的单数形式,如“一种”,包括复数指代,除非另有规定。此外,术语“包括”是开放性限定并非封闭式,即包括本发明所指明的内容,但并不排除其他方面的内容。
除非另有说明,本发明采用质谱、元素分析的传统方法,各步骤和条件可参照本领域常规的操作步骤和条件。
除非另有指明,本发明采用分析化学、有机合成化学和光学的标准命名及标准实验室步骤和技术。在某些情况下,标准技术被用于化学合成、化学分析、发光器件性能检测。
另外,需要说明的是,除非以其他方式明确指出,在本发明中所采用的描述方式“…独立地为”应做广义理解,是指所描述的各个个体之间是相互独立的,可以独立地为相同或不同的具体基团。更详细地,描述方式“…独立地为”既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响;也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。
本领域技术人员可以理解,根据本领域中使用的惯例,本申请描述基团的结构式中所使用的
Figure PCTCN2021110751-appb-000092
是指,相应的基团通过该位点与化合物中的其它片段、基团进行连接。
本领域技术人员可以理解,根据本领域中使用的惯例,本申请描述基团的结构式中所使用的
Figure PCTCN2021110751-appb-000093
表示单键或双键。
在不违背本领域常识的基础上,上述各优选条件,可任意组合,即得本发明各较佳实例。
本发明所用试剂和原料均市售可得。
本发明的积极进步效果在于:本发明化合物对补体因子B具有抑制活性(在补体溶血活性实验中,对补体因子B抑制活性IC 50值均<5μM,绝大部分IC 50<1.5μM,部分IC 50<1μM)。
具体实施方式
下面通过实施例的方式进一步说明本发明,但并不因此将本发明限制在所述的实施例范围之中。下列实施例中未注明具体条件的实验方法,按照常规方法和条件,或按照商品说明书选择。
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10 -6(ppm)的单位给出。NMR的测定是用Bruker ASCEND TM-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6)、氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS)。
MS的测定用Agilent 6110,Agilent 1100,Agilent 6120,AgilentG6125B液相质谱联用仪。
HPLC的测定使用岛津HPLC-2010C高压液相色谱仪(XBRIDGE 2.1*50mm,3.5um色谱柱)。
手性HPLC分析测定使用THARSFC X5。
薄层层析硅胶板使用烟台青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm-0.2mm,薄层层析分离纯化产品采用的规格是0.4mm-0.5mm。
柱层析一般使用青岛海洋硅胶200-300目硅胶为载体。
高效液相制备使用Waters 2767、Waters 2545、和创新恒通LC3000制备型色谱仪。
手性制备柱层析使用Shimadzu LC-20AP、THARSFC PREP 80。
加压氢化反应使用北京佳维科创科技GCD-500G型氢气发生器。
微波反应使用Biotage initiator+型微波反应器。
实验例中如无特殊说明,反应均在氩气氛或者氮气氛下进行。
氩气氛或者氮气氛是指反应瓶连接一个约1升容积的氩气或者氮气气球。
氢气氛是指反应瓶连接一个约1升容积的氢气气球。
实验例中如无特殊说明,反应温度均为室温,温度范围是20℃-30℃。
试剂英文缩写对应的试剂名称
Figure PCTCN2021110751-appb-000094
Figure PCTCN2021110751-appb-000095
Figure PCTCN2021110751-appb-000096
Figure PCTCN2021110751-appb-000097
实施例1:
中间体1:
Figure PCTCN2021110751-appb-000098
在3L的三口瓶中,依次加入四氢呋喃(150mL)和4-溴苯腈(50g),在氮气保护下将异丙基氯化镁氯化锂络合物(1.3M,210mL)缓慢加到反应体系中。反应在室温下进行2小时后,反应体系加入无水四氢呋喃(500mL)进行稀释后,降温至-5℃,加入4-甲氧基吡啶(25mL),缓慢滴加氯甲酸苄酯(35mL)(维持体系温度在0℃以下),滴加完毕后反应在0℃继续进行2小时,然后升至室温并在该温度下反应16小时。反应结束后,加入盐酸溶液(6M,150mL),室温搅拌半小时并加水(1000mL)稀释,用乙酸乙酯(500mL)进行萃取两次,萃取相用饱和食盐水(50mL)洗涤,然后用无水硫酸钠干燥并过滤,滤液浓缩后获得的粗品并通过硅胶柱分离纯化(石油醚:乙酸乙酯=3:1-1:1)得到中间体1(23g,收率:23%)。MS m/z(ESI):333.0[M+H].
中间体2:
Figure PCTCN2021110751-appb-000099
在500mL单口瓶中,依次加入中间体1(28g)、锌粉(55g)和乙酸(200mL),反应加热到100℃并在该温度下进行反应16小时。反应结束后过滤,滤液加水(500mL) 稀释,用乙酸乙酯(500mL)进行萃取,萃取相用饱和碳酸氢钠水溶液(500mL)洗涤两次,饱和食盐水(100mL)洗涤一次,无水硫酸钠干燥并过滤,滤液减压浓缩获得中间体2(26g,收率:73%)。MS m/z(ESI):334.8[M+H].
中间体3:
Figure PCTCN2021110751-appb-000100
在1L单口瓶中,依次加入四氢呋喃(100mL)、乙醇(100mL)和中间体2(26g),然后分批加入硼氢化钠(2g),反应在室温下进行2小时。反应结束后,将体系降温至0℃,加入饱和氯化铵水溶液(100mL)至不再升温,加水(300mL)稀释,用乙酸乙酯(200mL)进行萃取两次;萃取相用饱和食盐水(500mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩得到中间体3(25g,收率:76%)。MS m/z(ESI):336.9[M+H].
中间体4:
Figure PCTCN2021110751-appb-000101
将二氯甲烷(200mL)加入到500mL单口瓶中,然后依次加入中间体3(25g)、咪唑(6.6g)和叔丁基二苯基氯硅烷(25g),室温反应2小时。反应结束后,加水(500mL)稀释,用二氯甲烷(200mL)萃取,萃取相用水(50mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩,残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体4(5.7g,收率:13%,R f=0.55;异构体R f=0.50)。MS m/z(ESI):597.0[M+23].
中间体5:
Figure PCTCN2021110751-appb-000102
在250mL单口瓶中,依次加入四丁基氟化铵四氢呋喃溶液(1M,30mL)和中间体4(5g),反应在室温下进行2小时。反应结束后,加水(100mL)稀释,用乙酸乙酯(50mL)萃取三次,萃取相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩,残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=3:1-0:1)得到消旋体中间体,该 中间体经过SFC进行手性分离(Apparatus:SFC Thar prep 80;Column:CHIRALPAK AD-H,250mm x 20mm,5μm;Modifier:35%甲醇(0.2%氨水);柱温:40℃;柱压:60bar;波长:214/254nm;流速:40g/min;Rt=4.78min)得到中间体5(1.2g,收率:41%)。MS m/z(ESI):358.8[M+23].
中间体6:
Figure PCTCN2021110751-appb-000103
在100mL单口瓶中,依次加入溶剂N,N-二甲基甲酰胺(15mL)、中间体5(1.2g)和碘乙烷(1.1g);反应体系降温至0℃后,加入钠氢(60%,243mg),然后体系升温至室温并在该温度下进行2小时。反应结束后,加水(30mL)稀释,乙酸乙酯(50mL)萃取,萃取相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩得到中间体6(1.2g,收率:83%)。MS m/z(ESI):386.9[M+23].
中间体7:
Figure PCTCN2021110751-appb-000104
在100mL单口瓶中,依次加入甲醇(10mL)、水(10mL)、浓硫酸(10mL)和中间体6(1.2g),反应加热到80℃并在该温度下进行48小时。反应结束后,反应液浓缩除去甲醇,残余物用饱和氢氧化钠水溶液调pH至中性,乙酸乙酯(10mL)萃取三次,萃取相用饱和食盐水(5mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩得到中间体7(850mg,收率:81%)。MS m/z(ESI):264.1[M+H]. 1H NMR(400MHz,CDCl 3)δ8.01(d,J=8.3Hz,2H),7.49(d,J=8.3Hz,2H),4.13(dd,J=11.7,2.4Hz,1H),3.92(s,3H),3.82–3.70(m,1H),3.62–3.47(m,2H),3.27–3.10(m,1H),3.02–2.88(m,1H),2.07–1.97(m,1H),1.95–1.85(m,1H),1.82-1.62(m,2H),1.27(t,J=7.0Hz,3H).
中间体8:
Figure PCTCN2021110751-appb-000105
在250mL单口瓶中,依次加入二氯甲烷(50mL)、5-甲氧基-7-甲基-1H-吲哚(3g)、BOC酸酐(5.68g)、4-二甲氨基吡啶(227mg)和三乙胺(2.26g),反应在室温下进行 16小时。反应结束后,反应液加入饱和氯化铵溶液(5mL)淬灭,二氯甲烷萃取(20mL)三次,合并的有机相用水洗涤(5mL),无水硫酸钠干燥并过滤,过滤液浓缩,残余物硅胶经柱层析法(石油醚:乙酸乙酯=10:1)纯化得到中间体8(4.6g,收率:94%)。MS m/z(ESI):262.0[M+H].
中间体9:
Figure PCTCN2021110751-appb-000106
在250mL单口瓶中,依次加入二氯甲烷(80mL)、N-甲基甲酰苯胺(3.8g)和草酰氯(3.6g),反应在室温下搅拌3小时。然后将反应温度降至-14℃,加入中间体8(2.5g),反应体系自然升温到室温并搅拌1小时。反应结束后,反应液倒入冰水中,用二氯甲烷(100mL)萃取三次,合并的萃取相用水(10mL)洗涤两次,无水硫酸钠干燥并过滤,过滤液浓缩,残余物经硅胶柱分离纯化(石油醚:乙酸乙酯=20:1)得到中间体9(1.3g,收率:47%)。MS m/z(ESI):290.0[M+H]. 1H NMR(400MHz,CDCl 3)δ10.65(s,1H),7.65(d,J=3.4Hz,1H),7.49(d,J=3.4Hz,1H),6.76(s,1H),3.98(s,3H),2.70(s,3H),1.65(s,9H).
中间体10:
Figure PCTCN2021110751-appb-000107
在50mL三口瓶中,依次加入1,2-二氯乙烷(5mL)、中间体7(127mg)和中间体9(130mg),反应在室温下进行18小时,然后加入三乙酰氧基硼氢化钠(438.72mg),室温继续进行反应18小时。反应结束后,加入二氯甲烷(10mL)稀释,用10mL水洗,有机相用无水硫酸钠干燥并过滤,过滤液浓缩,残余物硅胶柱分离纯化(甲醇:二氯甲烷=1:10),得到中间体10(50mg,收率:14.58%)。MS m/z(ESI):437.3[M+H],RT=1.142min。
中间体11:
Figure PCTCN2021110751-appb-000108
在50mL三口瓶中,依次加入四氢呋喃(0.5mL)、甲醇(0.5mL)、水(0.5mL)、氢氧化钠(44mg)和中间体10(50mg),反应在室温下进行18小时。反应结束后,反应液直接减压浓缩和冷冻干燥,得到的中间体11(50mg,收率:92%)。MS m/z(ESI):423.1[M+H].
中间体7可以通过另外一个方案获得:
中间体12:
在2L三口瓶中,依次加入四丁基氟化铵四氢呋喃溶液(1M,840mL)和中间体4(140g),反应在室温下进行2小时。反应结束后,加水(600mL)稀释,用乙酸乙酯(700mL)萃取三次,萃取相用饱和食盐水(500mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩,残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=3:1-1:1)得到中间体12(77g,收率95%),MS m/z(ESI):358.8[M+23].
中间体13:
在2L三口瓶中,依次加入溶剂N,N-二甲基甲酰胺(700mL)、中间体12(77g)和碘乙烷(56g);反应体系降温至0℃后,加入钠氢(60%,14.61g),然后体系升温至室温并在该温度下进行2小时。反应结束后,降温至0℃,加氯化铵水溶液至反应不升温,乙酸乙酯(500mL)萃取,萃取相用饱和食盐水(300mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩得到中间体13(75g,收率:89%)。MS m/z(ESI):386.9[M+23].
中间体14:
在2L三口瓶中,依次加入异丙醇(300mL)、水(800mL)、中间体3(75g)、Ba(OH)2·8H2O(233g),反应加热到100℃并在该温度下进行20小时。反应结束后,反应液浓缩除去异丙醇,残余物用饱和氢氧化钠水溶液调pH至2-3,二氯甲烷(300mL)萃取三次,萃取相用饱和食盐水(200mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩得到中间体14(67g,收率:85%)。MS m/z(ESI):384.1[M+H].
中间体15:
在2L三口瓶中,依次加入N,N-二甲基甲酰胺(670mL)、碳酸钾(96.6g)、碘甲烷(37.3g)和中间体14(67g),室温下反应2小时。反应结束后,加入300mL水淬灭 反应,用甲基叔丁基醚萃取(300mL*2)次,萃取相用饱和食盐水(5mL)洗涤,无水硫酸钠干燥并过滤,滤液减压浓缩,柱层析(石油醚:乙酸乙酯=10:1-3:1)得到中间体15(54g,收率:78%)。MS m/z(ESI):394.1[M+H].
中间体7:
在1L单口瓶中,依次加入乙酸乙酯(500mL)、钯碳(5.4g,10%负载量)、中间体15(54g),在一个氢气压力下,反应在室温下进行16小时。反应结束后,反应液加入硅藻土过滤,滤液减压浓缩得消旋中间体,该中间体进行手性分离(Apparatus:Shimadzu LC-20AD;Column:CHIRALPAK AD-H(ADH0CD-SK003),0.46cm I.D.*25cm L;Modifier:(甲醇/二乙胺0.1%)/CO2=25/75(V/V);流速:2.0ml/min;Rt=3.58min)得到中间体7(15.7g,收率:43%)。1H NMR(400MHz,DMSO-d 6)δ7.89(d,J=8.26Hz,2H),7.50(d,J=8.26Hz,2H),3.92(dd,J=11.36,2.32Hz,1H),3.84(s,3H),3.69-3.64(m,1H),3.51-3.42(m,2H),2.94(dt,J=12.15,2.56Hz,1H),2.76(ddd,J=11.62,4.24,2.62Hz,1H),1.85(dd,J=13.23,2.16Hz,1H),1.73(d,J=13.47Hz,1H),1.59-1.41(m,2H),1.16(t,J=6.98Hz,3H),MS m/z(ESI):264.0[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000109
在25mL三口瓶中,依次加入乙酸(1mL)、中间体11(50mg)和氰基硼氢化钠(23mg),反应在室温下进行18小时。反应结束后,反应液加入乙酸乙酯(5mL)稀释,用饱和食盐水洗涤(5mL)一次,无水硫酸钠干燥并过滤,过滤液减压浓缩,残余物经Prep-HPLC(Apparatus:Shimadzu LC-20AD;Column:CHIRALPAK AD-H(ADH0CD-SK003),0.46cm I.D.*25cm L;Modifier:(甲醇/二乙胺0.1%)/CO 2=25/75(V/V);流速:2.0ml/min;Rt=3.58min)纯化得到目标化合物(10mg,收率:18.67%)。MS m/z(ESI):425.1[M+H]. 1H NMR(400MHz,DMSO-d 6)δ8.19(d,J=8.4Hz,2H),7.69(d,J=8.4Hz,2H),6.63(s,1H),4.83-4.68(m,1H),4.15–4.01(m,1H),4.00–3.85(m,2H),3.72(s,3H),3.67-3.57(m,2H),3.55–3.35(m,4H),3.10–2.85(m,2H),2.35–2.22(m,2H),2.18(s,3H),2.17–2.04(m,2H),1.31(t,J=6.8Hz,3H).
实施例2:
中间体1:
Figure PCTCN2021110751-appb-000110
向实施例1的中间体10(50mg)的N,N-二甲基甲酰胺(1mL)反应液中分批加入N-溴代琥珀酰亚胺(22.4mg)。反应在室温下进行18小时后,反应液直接通过Prep-HPLC进行纯化(色谱柱:C18spherical,100A,20g,20-35um;乙腈-水=10-70%,UV:214nm)得到中间体1(120mg,收率:48.38%)MS m/z(ESI):501.1[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000111
在10mL小瓶中依次加入四氢呋喃(1mL)、中间体1(10mg)和50%氢溴酸溶液(0.5mL),反应升温至70℃并在该温度下进行反应5小时。反应结束后,减压浓缩,得到的残余物经Prep-HPLC纯化(色谱柱:C18spherical,100A,20g,20-35um;乙腈-水=10-70%;UV:214nm.)纯化得到目标化合物(51.5mg,收率:55.8%)。MS m/z(ESI):439.3[M+H]. 1H NMR(400MHz,CD 3OD)δ8.20(d,J=8.0Hz,2H),7.72(d,J=8.0Hz,2H),6.76(s,1H),4.77(dd,J=12.0Hz,3.6Hz,1H),4.08–3.97(m,2H),3.89–3.82(m,1H),3.66–3.55(m,4H),3.53–3.39(m,3H),2.36–2.21(m,5H),2.18–2.09(m,2H),1.95–1.85(m,1H),1.70–1.60(m,1H),1.30(t,J=7.0Hz,3H).
实施例3
中间体1:
Figure PCTCN2021110751-appb-000112
在250mL单口瓶中,依次加入甲醇(100mL)和3-溴-2-氟-6-甲基吡啶(10g),然后叔丁醇钾(11g)分批加到反应体系中,反应加热到75℃并在该温度下搅拌3小时。反应结束后,反应液直接减压浓缩并通过硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体1(10g,收率:89%)。MS m/z(ESI):201.9[M+H].
中间体2:
Figure PCTCN2021110751-appb-000113
在250mL单口瓶中,依次加入浓硫酸(50mL)和中间体1(10g),然后缓慢滴加硝酸(20mL),滴加完毕后,反应在室温进行1小时。反应结束后,反应液倒入冰水中,用乙酸乙酯(500mL)萃取两遍,有机相用饱和食盐水(300mL)洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩得到中间体2(7.5g,收率:75%)。 1H NMR(400MHz,DMSO-d 6)δ8.65(s,1H),4.04(s,3H),2.69(s,3H).
中间体3:
Figure PCTCN2021110751-appb-000114
在500mL单口瓶中,依次加入溶剂四氢呋喃(100mL)和中间体2(5g)。在-40℃和氮气保护下缓慢滴加乙烯基氯化镁(1M in THF,100mL)到反应体系中,反应缓慢升温到-20℃并在该温度下进行2小时。反应结束后,加氯化铵水溶液(100mL)淬灭,用乙酸乙酯(200mL)萃取两遍,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥并过滤,过滤液减压浓缩,残余物通过硅胶柱分离纯化(二氯甲烷:甲醇=30:1)得到中间体3(4g,收率:75%)。MS m/z(ESI):240.8[M+H].
中间体4:
Figure PCTCN2021110751-appb-000115
在500mL单口瓶中,依次加入二氯甲烷(100mL)、中间体3(4g)、Boc酸酐(6g)和4-二甲氨基吡啶(3g),反应在室温下进行2小时。反应结束后,加水(100mL)稀释,二氯甲烷(100mL)萃取,有机相用水(30mL)洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩。残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体4(2.7g,收率: 44%)。MS m/z(ESI):340.8[M+H].
中间体5:
Figure PCTCN2021110751-appb-000116
在到50mL三口瓶中,依次加入溶剂四氢呋喃(10mL)和中间体4(500mg),用干冰降温至-78℃,然后在氮气保护下缓慢滴加正丁基锂(0.75mL)。反应在-78℃进行1小时后,无水DMF(131mg)缓慢加到反应体系中并在该温度下继续反应2小时。反应体系自然升温到0℃,加饱和氯化铵(20mL)淬灭反应,用乙酸乙酯(50mL)萃取两遍,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体5(160mg,收率:16%)。MS m/z(ESI):291.1[M+H].
中间体6:
Figure PCTCN2021110751-appb-000117
在100mL单口瓶中,依次加入溶剂1,2二氯乙烷(3mL)、中间体5(60mg)和实施例1中间体7(55mg)。反应在25℃进行8小时后,加入三乙酰氧基硼氢化钠(120mg),25℃继续反应16小时。反应结束后,反应液直接加入硅胶拌样,用硅胶柱分离纯化(甲醇:二氯甲烷=1:20)得到中间体6(75mg,60%)。MS m/z(ESI):537.8[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000118
在25mL单口瓶中,依次加入甲醇(2mL)、水(2mL)、中间体6(75mg)和氢氧化钠(50 mg),反应加热到75℃并在该温度下进行3小时。反应结束后,反应液直接通过Prep-HPLC进行纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:15-35%),得到的溶液浓缩,剩余少量水溶液冷冻干燥得到目标化合物(33.0mg,收率:46%,含0.5当量的甲酸)。MS m/z(ESI):423.9[M+H]. 1H NMR(400MHz,CD 3OD)δ8.43(s,0.5H),8.16(d,J=8.4Hz,2H),7.65(d,J=8.4Hz,2H),7.51(d,J=3.2Hz,1H),6.34(s,1H),4.76–4.66(m,1H),4.33–4.13(m,2H),3.89(s,3H),3.88–3.82(m,1H),3.66–3.58(m,2H),3.57–3.49(m,1H),3.41-3.34(m,1H),2.64(s,3H),2.32–2.22(m,2H),2.14–2.04(m,2H),1.32(t,J=6.8Hz,3H).
实施例4:
中间体1:
Figure PCTCN2021110751-appb-000119
室温下,将氢氧化钠水溶液(15%,10mL)加到N-(4-甲氧基-2-甲基-6-硝基苯基)乙酰胺(500mg)的乙醇(10mL)溶液中,反应加热到90℃并在该温度下进行反应16小时。反应结束后,反应体系冷却至室温,减压浓缩除去乙醇,加入水(20mL)稀释,用5M盐酸调节pH至5-6,然后用乙酸乙酯进行萃取(20mL)五次。合并的萃取相用饱和食盐水洗涤(20mL),无水硫酸钠干燥并过滤,过滤液减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=5:1)得到中间体1(180mg,收率44%)。MS m/z(ESI):183.0[M+H].
中间体2:
Figure PCTCN2021110751-appb-000120
室温下将10%Pd/C(54mg,30%wt/wt)加到中间体1(180mg)的甲醇(15mL)溶液中,室温进行氢化反应16小时。反应结束后,反应液过滤,过滤液直接减压浓缩得到中间体2(120mg,收率80%)。MS m/z(ESI):153.1[M+H].
中间体3:
Figure PCTCN2021110751-appb-000121
室温下将甲酸(681mg)加到中间体2(450mg)的盐酸(4M,20mL)溶液中,反应体系加 热到100℃并在该温度下进行反应3小时。反应结束后,反应液倒入水中(20mL),用5M氢氧化钠溶液调节pH至8-9,用乙酸乙酯萃取(30mL)三次,合并的有机相用饱和食盐水洗涤(20mL)二次,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物用硅胶柱分离纯化(二氯甲烷:甲醇=10:1)得到中间体3(320mg,收率66%)。MS m/z(ESI):163.1[M+H].
中间体4:
Figure PCTCN2021110751-appb-000122
5℃下将乌托洛品(220mg)加到中间体3(170mg)的三氟乙酸(10mL)溶液中,反应加热到80℃并在该温度下进行反应3小时。反应结束后,反应液减压浓缩,用硅胶柱分离纯化(二氯甲烷:甲醇=10:1)得到中间体4(170mg,收率85%)。MS m/z(ESI):191.0[M+H].
中间体5:
Figure PCTCN2021110751-appb-000123
室温下将三乙酰基硼氢化钠(381.60mg)加到中间体4(171mg)和实施例1的中间体7(157mg)的1,2-二氯乙烷(10mL)溶液中,反应在25℃进行16小时。反应结束后,反应液进行减压浓缩,粗品用硅胶柱分离纯化(二氯甲烷:甲醇=20:1)获得到中间体5(47mg,收率18%)。MS m/z(ESI):438.0[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000124
室温下将氢氧化钠(60mg)的水(2mL)溶液加到中间体5(67mg)的甲醇(6mL)溶液中, 反应在室温进行16小时。反应结束后,减压浓缩除去甲醇溶剂,水相用盐酸(5M)溶液调节pH至5-6,然后减压浓缩。粗品用Prep-HPLC进行分离纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:5-30%),再通过制备TLC(二氯甲烷/甲醇=5:1)纯化得到目标化合物(13mg,收率20%,含0.4当量的甲酸)。MS m/z(ESI):423.8[M+H]. 1H NMR(400MHz,CD 3OD)δ8.54(s,0.4H),8.13(s,1H),8.02(d,J=8.2Hz,2H),7.58(d,J=8.2Hz,2H),6.82(s,1H),4.26–4.16(m,1H),4.11(d,J=13.6Hz,1H),3.83(d,J=13.6Hz,1H),3.76(s,4H),3.56(q,J=6.8Hz,2H),3.15–2.91(m,2H),2.55(s,3H),2.17–2.07(m,2H),1.98–1.92(m,2H),1.27(t,J=6.8Hz,3H).
实施例5:
中间体1:
Figure PCTCN2021110751-appb-000125
向2-溴-5-甲氧基-1,3-二甲苯(10g)的四氯化碳(100mL)溶液中加入N-溴代琥珀酰亚胺(8g)和偶氮二异丁腈(AIBN)(1g),反应加热到80℃并在该温度下搅拌过夜。反应结束后,反应液倒入二氯甲烷(300mL)稀释,用碳酸氢钠水溶液洗涤(30mL)两次,再用水洗涤(30mL)一次,无水硫酸钠干燥并过滤,滤液减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到粗品中间体1(11g,收率:64%)。
中间体2:
Figure PCTCN2021110751-appb-000126
向中间体1(15g)的二氧六环(250mL)溶液中加入碳酸铯(30g)和水(250mL),反应体系加热到110℃并在该温度下搅拌16小时。反应结束后,反应液减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体2(6.5g,48%)。MS m/z(ESI):252.9[M+Na].
中间体3:
Figure PCTCN2021110751-appb-000127
向中间体2(1.8g)的二氯甲烷(50mL)溶液中加入二氧化锰(3g),反应在25℃搅拌过夜。反应结束后,反应体系过滤,过滤液进行减压浓缩,残余物通过硅胶柱色谱纯化(石油醚:乙酸乙酯=10:1)得到中间体3(0.72g,收率:44%)。MS m/z(ESI):228.8[M+H].
中间体4:
Figure PCTCN2021110751-appb-000128
将中间体3(680mg)加入到对甲苯磺酰肼(550mg)的甲醇(10mL)溶液中,反应进行加热回流3小时。然后减压浓缩除去溶剂,得到的产物加入到氧化亚铜(2g)的均三甲苯(10mL)混合物中,反应加热到130℃并在该温度下继续进行反应16小时。反应结束后,反应液直接浓缩除去溶剂,残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体4(400mg,收率:42%)。MS m/z(ESI):317.0[M+H].
中间体5:
Figure PCTCN2021110751-appb-000129
将中间体4(800mg)加入到二氯甲烷溶液(6mL)中,然后降温至-78℃,在该温度下缓慢滴加四氯化钛(1.8g),反应搅拌五分钟后加入二氯甲基甲醚(1.2g),得到的反应液慢慢升温至0℃并进行反应6小时。反应结束后,反应体系冷却至-40℃,缓慢加入水(10mL)淬灭反应并用乙酸乙酯(30mL)和水(10mL)稀释,有机相分离后用碳酸氢钠水溶液洗涤(5mL)一次,饱和食盐水洗涤(5mL),无水硫酸钠干燥并过滤。过滤液进行减压浓缩,残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体5(400mg,收率:38%)。 1H NMR(400MHz,CDCl 3)δ10.53(s,1H),8.94(s,1H),7.71(d,J=8.4Hz,2H),7.24(d,J=8.4Hz,2H),7.04(s,1H),4.00(s,3H),2.93(s,3H),2.38(s,3H).
中间体6:
Figure PCTCN2021110751-appb-000130
中间体5(214mg)和实施例1的中间体7(140mg)的1,2-二氯乙烷(2mL)溶液在室温 和氮气保护下搅拌6小时,再加入三醋酸硼氢化钠(340mg),反应在室温和氮气保护下继续搅拌过夜。反应结束后,加入二氯乙烷(10mL)和水(10mL)稀释反应,有机相分离后用无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物通过硅胶柱分离纯化(石油醚:乙酸乙酯=5:1)得到中间体6(170mg,收率:54%)。MS m/z(ESI):592.0[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000131
将氢氧化锂(90mg)的水溶液(1mL)加入到中间体6(170mg)的四氢呋喃(2mL)和甲醇(2mL)溶液中,反应在室温下搅拌过夜。反应结束后,反应液用1M盐酸调节pH值到3-4,减压浓缩除去溶剂,残余物通过Prep-HPLC进行分离纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:10-40%),得到的溶液浓缩,剩余少量水溶液冷冻干燥得到目标化合物(49mg,收率:38%,含0.5当量的对甲苯磺酸)。MS m/z(ESI):424.3[M+H]. 1H NMR(400MHz,CD 3OD)δ8.18(d,J=8.0Hz,2H),7.95(s,1H),7.75–7.65(m,3H),7.21(d,J=8.0Hz,1H),7.10(s,1H),4.80–4.65(m,1H),4.40-4.20(m,2H),3.90–3.77(m,4H),3.65–3.50(m,3H),3.58(s,3H),2.36(s,1.5H),2.32–2.20(m,2H),2.15–1.95(m,2H),1.30(t,J=6.8Hz,3H).
实施例6:
中间体1:
Figure PCTCN2021110751-appb-000132
在室温下,将亚硝酸叔丁酯(499mg)添加5-甲氧基-3-甲基苯-1,2-二胺(500mg)的乙腈(20mL)溶液中,反应混合物在室温下搅拌16小时。反应结束后,反应液减压浓缩,残余物用硅胶柱分离纯化(二氯甲烷:甲醇=20:1)得到中间体1(360mg,收率67%)。MS m/z(ESI):164.1[M+H].
中间体2:
Figure PCTCN2021110751-appb-000133
室温下将乌托洛品(582mg)加到中间体1(340mg)的三氟乙酸(10mL)溶液中,反应加热到80℃并在该温度下搅拌3小时。反应结束后,反应体系冷却至室温,反应液直接进行减压浓缩,所得残余物用硅胶柱分离纯化(二氯甲烷:甲醇=10:1)得到中间体2(200mg,收率50%)。MS m/z(ESI):192.0[M+H].
中间体3:
Figure PCTCN2021110751-appb-000134
室温下将三乙酰硼氢化钠(419.76mg)加到中间体2(189.27mg)和实施例1的中间体7(174mg)的1,2-二氯乙烷(20mL)溶液中,反应在室温搅拌16小时。反应结束后,反应液直接进行减压浓缩,所得残余物用硅胶柱分离纯化(二氯甲烷:甲醇=50:1)得到中间体3(180mg,收率62%)。MS m/z(ESI):438.8[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000135
室温下将氢氧化钠(164mg)的水溶液(2mL)加到中间体3(180mg)的甲醇(6mL)溶液中,反应在室温下搅拌16小时。反应结束后,减压浓缩除去甲醇,水相用5M盐酸溶液调节pH至5-6,直接进行减压浓缩,所得残余物用Prep-HPLC进行分离纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:10-30%)得到目标化合物(65mg,收率37%,含0.5当量的甲酸)。MS m/z(ESI):424.8[M+H]. 1H NMR(400MHz,CD 3OD)δ8.37(s,0.5H),8.11(d,J=8.0Hz,2H),7.68(d,J=8.0Hz,2H),7.10(s, 1H),4.54–4.44(m,1H),4.34–4.26(d,J=13.2Hz,1H),4.14–4.04(m,1H),3.87-3.80(m,4H),3.65–3.55(m,2H),3.43-3.32(m,1H),3.17–3.08(m,1H),2.69(s,3H),2.28-2.15(m,2H),2.08-1.99(m,2H),1.30(t,J=7.0Hz,3H).
实施例7:
目标化合物:
Figure PCTCN2021110751-appb-000136
在50mL单口瓶中依次加入乙腈(20mL)、水(2mL)、实施例1的中间体11(150mg)、七水合氯化铈(8.1mg)和2-碘酰基苯甲酸(252mg),反应体系加热到80℃并在该温度下搅拌3.5小时。反应结束后,反应液直接进行减压浓缩,所得残余物经Prep-HPLC进行纯化(色谱柱:C18spherical,100A,20g,20-35um;乙腈-水=10-70%,UV:214nm.)得到目标化合物(5.9mg;收率:55.8%,含0.8当量的甲酸)。MS m/z(ESI):453.1[M+H]. 1H NMR(400MHz,CD 3OD)δ8.43(s,0.8H),8.04(d,J=8.0Hz,2H),7.58(d,J=8.0Hz,2H),6.72(s,1H),3.76-3.71(m,4H),3.60-3.40(m,4H),3.20–3.00(m,1H),2.55-2.35(m,2H),2.20(s,3H),2.10-1.98(m,2H),1.93–1.83(m,1H),1.70-1.56(m,1H),1.23(t,J=6.8Hz,3H).
实施例8:
中间体1:
Figure PCTCN2021110751-appb-000137
向250mL单口瓶中加入吡啶(70mL)、4-甲氧基-2-甲基苯甲醛(10g)、丙二酸(13.87g)和哌啶(568mg),反应体系加热到90℃并在该温度下搅拌16小时。反应结束后,反应液倒入冰水中,用稀盐酸调节pH=3-4有固体析出,过滤并收集固体、干燥得到中间体1(12g,收率:86%)。MS m/z(ESI):193.1[M+H].
中间体2:
Figure PCTCN2021110751-appb-000138
向250mL的单口瓶中加入甲醇(100mL)、中间体1(5g)和Pd/C(1g),在氢气气氛和室温下进行催化氢化反应16小时。反应结束后,反应液过滤,滤液减压除去溶剂(水浴:40℃)得到中间体2(4.9g,收率:97%)。MS m/z(ESI):195.0[M+H].
中间体3:
Figure PCTCN2021110751-appb-000139
向闷罐中加入二氯甲烷(20mL)、中间体2(2g)和三氟甲磺酸(4.6g),反应加热到80℃并在该温度下进行搅拌16小时。反应结束后,反应液倒入冰水中,用二氯甲烷萃取(30mL)三次,合并的有机相用无水硫酸钠干燥并过滤,过滤液减压浓缩(水浴:40℃),所得残余物用制备TLC进行分离纯化(石油醚:乙酸乙酯=10:1)得到中间体3(300mg,收率:16%)。MS m/z(ESI):177.0[M+H].
中间体4:
Figure PCTCN2021110751-appb-000140
向50mL单口瓶中依次加入***(30mL)、氯化铝(1.7g)和氢化铝锂(494mg),降温至0℃,加入中间体3(130mg),反应自然升温至室温并在该温度下搅拌16小时。反应结束后,反应液缓慢加入5M氢氧化钠溶液进行淬灭,用乙酸乙酯萃取(50mL)三次,合并的有机相用饱和食盐水洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,得到中间体4(收率:47%)。 1H NMR(400MHz,CDCl 3)δ6.67–6.57(m,2H),3.85(s,3H),2.92–2.77(m,4H),2.29(s,3H),2.11-2.06(m,2H).
中间体5:
Figure PCTCN2021110751-appb-000141
向50mL单口瓶中加入三氯氧磷/N,N-二甲基甲酰胺混合溶液(9:10,6mL),氮气气氛和室温下搅拌15分钟,然后加入中间体4(350mg),反应加热到60℃并在该温度下搅拌1小时。反应结束后,反应液倒入冰水中,乙酸乙酯萃取(50mL)三次,合并的有机相用无水硫酸钠干燥并过滤,减压浓缩除去溶剂(水浴:40℃),残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=10:1),过滤液进行减压浓缩,通过Prep-HPLC进行分离纯化(色谱柱:Gemini-C18 150 x 21.2mm,5um,流动项:乙腈-水(0.1%甲酸),梯度:60-65%)得到中间 体5(20mg,收率:5%)。MS m/z(ESI):191.1[M+H].
中间体6:
Figure PCTCN2021110751-appb-000142
向25mL的微波管中加入四氢呋喃(4mL)、实施例1的中间体7(20mg)、中间体5(20mg)和钛酸四乙酯(16mg)。反应体系在氮气气氛下加热到70℃并在该温度下反应16小时,然后反应体系降温至室温,加入三乙酰氧基硼氢化钠(44mg)并继续反应1小时。反应结束后,加入甲醇至溶液澄清,经制备TLC(石油醚:乙酸乙酯=2:1)纯化得到中间体6(30mg,收率:85%)。MS m/z(ESI):438.1[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000143
向50mL单口瓶中依次加入甲醇/水混合溶液(3:1,4mL)、中间体6(40mg)和氢氧化钠(73mg),反应在室温下进行16个小时。反应结束后,反应液倒入水中,稀盐酸调节反应液pH=7-8,减压浓缩(45℃)除去有机溶剂;剩余水相用二氯甲烷/甲醇(10:1)混合溶液洗涤(20mL)五次,然后水相残余物通过Prep-HPLC进行分离纯化(色谱柱:Gemini-C18 150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:20-50%,UV:214nm)得到目标化合物(5.3mg,收率:14%)。MS m/z(ESI):424.1[M+H]. 1H NMR(400MHz,CD 3OD)δ8.10(d,J=8.0Hz,2H),7.57(d,J=8.0Hz,2H),6.65(s,1H),4.70–4.55(m,1H),4.12–3.98(m,1H),3.94–3.80(m,2H),3.74(s,3H),3.63–3.54(m,2H),3.46–3.38(m,1H),2.94–2.65(m,5H),2.30–1.95(m,9H),1.29(t,J=7.0Hz,3H).
实施例9:
中间体1:
Figure PCTCN2021110751-appb-000144
0℃下将乙酸酐(8.9g)加到4-甲氧基-2-甲基苯胺(10g)的二氯甲烷(200mL)溶液中,在氮气气氛和室温下反应3小时。反应结束后,反应液分别用饱和碳酸氢钠溶液(100mL)和饱和食盐水(100mL)洗涤,有机相用无水硫酸钠干燥并过滤,过滤液通过减压浓缩得到中间体1(13.3g,收率91%)。MS m/z(ESI):180.1[M+H].
中间体2:
Figure PCTCN2021110751-appb-000145
0℃下将浓硝酸(8.2g,0.14mol)滴加到中间体1(11.7g)的醋酸(300mL)溶液中,反应在20℃搅拌3小时。反应结束后,反应混合液倒入水(500mL)中,用乙酸乙酯萃取(300mL)三次。合并的有机相用饱和食盐水洗涤(300mL)两次,无水硫酸钠干燥并过滤,过滤液减压浓缩,得到的粗品用硅胶柱分离纯化(石油醚:乙酸乙酯=1:1)得到中间体2(5.2g,收率33%)。MS m/z(ESI):225.0[M+H].
中间体3:
Figure PCTCN2021110751-appb-000146
室温下将10%Pd/C(150mg,30%WT/WT)加到中间体2(480mg)的甲醇(15mL)溶液中,反应在氢气气氛和室温条件下进行催化氢化16小时。反应结束后,反应液过滤,过滤液进行减压浓缩得到中间体3(430mg,纯度:80%,收率83%)。MS m/z(ESI):195.1[M+H].
中间体4:
Figure PCTCN2021110751-appb-000147
室温下将氢氧化钠水溶液(15%,10mL)加到中间体3(430mg)的乙醇(10mL)溶液中, 反应体系在90℃进行搅拌16小时。反应结束后,反应液直接进行减压浓缩除去乙醇,残余相加入水(20mL),用5M盐酸调节pH至5-6,然后用乙酸乙酯萃取(20mL)五次。合并的有机相用饱和食盐水洗涤(20mL),无水硫酸钠干燥并过滤,过滤液进行减压浓缩得到中间体4(370mg,收率94%)。MS m/z(ESI):177.1[M+H].
中间体5:
Figure PCTCN2021110751-appb-000148
室温下将乌托洛品(271mg)加到中间体4(310mg)的三氟乙酸(10mL)溶液中,反应体系在80℃搅拌3小时。反应结束后,反应液直接进行减压浓缩,粗品用硅胶柱分离纯化(二氯甲烷:甲醇=20:1)得到中间体5(170mg,收率47%)。MS m/z(ESI):205.1[M+H].
中间体6:
Figure PCTCN2021110751-appb-000149
室温下将三乙酰硼氢化钠(330mg)加到中间体5(159.30mg)和实施例1的中间体7(137mg)的1,2-二氯乙烷(20mL)溶液中,反应在室温下搅拌16小时。反应结束后,反应液直接进行减压浓缩,残余物用硅胶柱分离纯化(二氯甲烷:甲醇=20:1)得到中间体6(200mg,收率85%)。MS m/z(ESI):451.8[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000150
室温下将氢氧化钠(176mg)的水溶液(2mL)加到中间体6(200mg)的甲醇(6mL)溶液中,反应体系在室温下进行16小时。反应结束后,减压浓缩除去甲醇,水相用5M盐酸 溶液调节pH至5-6,然后减压浓缩,残余物用Prep-HPLC进行分离纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:0-30%)得到目标化合物(32mg,收率17%,含0.7当量的甲酸)。MS m/z(ESI):437.8[M+H]. 1H NMR(400MHz,CD 3OD)δ8.35(bs,0.7H),8.15(d,J=8.0Hz,2H),7.69(d,J=8.0Hz,2H),6.80(d,J=4.2Hz,1H),4.78–4.65(m,1H),4.38–4.32(m,1H),4.21(d,J=12.8Hz,1H),3.89–3.82(m,1H),3.77(s,3H),3.67–3.55(m,3H),3.32–3.22(m,1H),2.62(s,3H),2.52(s,3H),2.29-2.27(m,2H),2.08-2.00(m,2H),1.32(t,J=7.0Hz,3H).
实施例10:
中间体1:
Figure PCTCN2021110751-appb-000151
室温下向2-甲基-4-甲氧基苯甲醛(12g)的N,N-二甲基甲酰胺(200mL)溶液中分批次加入N-溴代琥珀酰亚胺(15.6g)(0.5小时),反应体系在室温下搅拌过夜。反应结束后,反应液加入乙酸乙酯(120mL)和碳酸氢钠水溶液(120mL)稀释,有机相分离,水相再用乙酸乙酯(120mL)萃取一次,合并的有机相用饱和食饱和食盐水洗涤(30mL),用无水硫酸钠干燥并过滤,过滤液进行减压浓缩。残余物用硅胶柱色谱进行分离纯化(石油醚/乙酸乙酯=20:1–5:1)得到中间体1(13g,收率:71%)。MS m/z(ESI):228.9[M+H].
中间体2:
Figure PCTCN2021110751-appb-000152
室温下向中间体1(10.7g)的二氯甲烷(150mL)溶液中分批加入间氯过氧苯甲酸(16g),反应混合物加热到40℃并在该温度下搅拌反应过夜。反应结束后,反应液加入二氯甲烷(100mL)稀释,用碳酸氢钠水溶液(30mL)洗涤三次,用水洗涤(30mL)一次,有机相用无水硫酸钠干燥并过滤,过滤液进行减压浓缩得到中间体2(10g,收率:8%)。MS m/z(ESI):244.9[M+H].
中间体3:
Figure PCTCN2021110751-appb-000153
向中间体2(10g)的乙腈溶液(150mL)中加入三乙胺(10g),反应体系加热到50℃并在该温度下搅拌6小时。反应结束后,反应液直接浓缩,残余物通过硅胶柱色谱进行分离纯化(石油醚:乙酸乙酯=10:1)得到中间体3(6g,收率:80%)。MS m/z(ESI):216.9[M+H].
中间体4:
Figure PCTCN2021110751-appb-000154
向中间体3(6g)和溴代乙醛缩二乙醇(9.5g)的N,N-二甲基甲酰胺(80mL)溶液中加入碘化钠(0.5g)和碳酸钾(15g),反应加热到100℃并在该温度下搅拌10小时。反应结束后,反应液加入乙酸乙酯(200mL)和水(200mL)稀释,水相用乙酸乙酯(100mL)萃取两次,合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物通过硅胶柱色谱进行分离纯化(石油醚:乙酸乙酯=10:1)得到中间体4(2.8g,收率:30%)。MS m/z(ESI):354.9[M+Na].
中间体5:
Figure PCTCN2021110751-appb-000155
向中间体4(2.8g)的甲苯溶液(50mL)中加入多聚磷酸(4g),反应液加热到100℃并在该温度下进行搅拌6小时。反应结束后,反应液加乙酸乙酯(100mL)和水(50mL)稀释,水相用乙酸乙酯(50mL)萃取一次,合并有机相用碳酸钠水溶液调节pH值为中性,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物通过硅胶柱色谱进行分离纯化(石油醚:乙酸乙酯=15:1)得到中间体5(1.5g,收率:74%)。 1H NMR(400MHz,CDCl 3)δ7.64(d,J=2.0Hz,1H),6.75–6.78(m,2H),3.92(s,3H),3.49(s,3H).
中间体6:
Figure PCTCN2021110751-appb-000156
中间体5(1.07g)的四氢呋喃(10mL)溶液在氮气保护下冷却到-78℃,缓慢滴加叔丁基锂溶液(1.3M,7.5mL),滴加完毕后反应在-78℃搅拌1小时,加入N,N-二甲基甲酰胺(1 g),然后反应混合物自然升温并在室温下搅拌过夜。反应结束后,反应液加入氯化铵水溶液(10mL)和乙酸乙酯(20mL)稀释并分相,有机相用无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物通过硅胶柱色谱进行分离纯化(石油醚:乙酸乙酯=10:1)得到中间体6(300mg,收率:36%)。 1H NMR(400MHz,CDCl 3)δ10.58(s,1H),7.72(d,J=2.0Hz,1H),7.53(d,J=2.0Hz,1H),6.75(s,1H),3.95(s,3H),2.58(s,3H).
中间体7:
Figure PCTCN2021110751-appb-000157
向1,2-二氯乙烷溶液(2mL)依次加入中间体6(150mg)和实施例1的中间体7(150mg),反应体系在室温和氮气保护下搅拌8小时,然后加入三乙酰基硼氢化钠(360mg),室温下继续反应过夜。反应结束后,反应液加入水(10mL)淬灭,加入乙酸乙酯(20mL)稀释,有机相用无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余相通过硅胶柱色谱进行分离纯化(石油醚:乙酸乙酯=5:1)得到中间体7(100mg,收率:40%)。MS m/z(ESI):438[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000158
向中间体7(100mg)的甲醇/四氢呋喃/水混合溶液(3mL,1:1:1)中加入氢氧化锂(100mg),反应室温搅拌过夜。反应结束后,反应液直接浓缩,残余物通过Prep-HPLC制备纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:25-40%),得到的产物溶液浓缩除去溶剂,剩余少量水溶液冷冻干燥得到目标化合物(50mg,收率:51%,含0.5当量的甲酸)。MS m/z(ESI):424.1[M+H]. 1H NMR(400MHz,CD 3OD)δ8.41(s,0.5H),8.16(d,J=8.4Hz,2H),7.82(d,J=2.0Hz,1H),7.67(d,J= 8.4Hz,2H),6.89(s,1H),6.79(s,1H),4.60–4.72(m,1H),4.26(d,J=12.8Hz,1H),4.10(d,J=12.8Hz,1H),3.87–3.82(m,1H),3.79(s,3H),3.66–3.56(m,2H),3.54–3.42(m,1H),3.31–3.22(m,1H),2.51(s,3H),2.32–2.20(m,2H),2.15–1.97(m,2H),1.32(t,J=6.8Hz,3H).
实施例11:
中间体1:
Figure PCTCN2021110751-appb-000159
0℃下将硝酸钾(4.35g)加到1-溴-2-甲氧基-4-(三氟甲基)苯(10g)的硫酸(40mL)溶液中,然后反应体系自然升温到室温并在该温度下搅拌1小时。反应结束后,反应液倒入冰水(200mL)中,用乙酸乙酯(200mL)萃取三次。合并的有机相用饱和食盐水(50mL)洗涤三次,饱和碳酸氢钠水溶液(50mL)洗涤一次,无水硫酸钠干燥并过滤,过滤液进行减压浓缩得到中间体1(10g,收率90%),粗品直接进行下一步。
中间体2:
Figure PCTCN2021110751-appb-000160
-78℃下将乙烯基溴化镁(150mL)缓慢滴加到中间体1(10g)的四氢呋喃(400mL)溶液中,反应-78℃进行2小时。反应结束后,向反应体系加入饱和氯化铵水溶液(100mL)淬灭,用乙酸乙酯(800mL)萃取三次。合并的有机相用饱和食饱和食盐水(200mL)洗涤一次,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)纯化得到中间体2(1.5g,收率15%)。MS m/z(ESI):293.9[M+H].
中间体3:
Figure PCTCN2021110751-appb-000161
将4-二甲氨基吡啶(372mg)加到中间体2(600mg)和Boc 2O酸酐(664mg)的二氯甲烷(20mL)溶液中,反应在室温下搅拌2小时。反应结束后,反应液直接进行减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体3(700mg,收率85%)。 1H  NMR(400MHz,DMSO-d 6)δ7.86(d,J=3.8Hz,1H),7.40(s,1H),6.73(d,J=3.8Hz,1H),3.97(s,3H),1.58(s,9H).
中间体4:
Figure PCTCN2021110751-appb-000162
-78℃下将叔丁基锂(1.68mL)缓慢滴加到中间体3(400mg)的四氢呋喃溶液(5mL)中,反应在-78℃搅拌1.5小时后,滴加N,N-二甲基甲酰胺(110mg)的四氢呋喃(15mL)溶液。滴加结束后,反应体系慢慢升温到室温并在该温度下搅拌1.5小时。反应结束后,反应液加入饱和氯化铵水溶液(20mL)淬灭,用乙酸乙酯(50mL)萃取三次。合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=5:1)得到中间体4(30mg,收率8%)。 1H NMR(400MHz,CDCl 3)δ10.71(s,1H),7.66(d,J=3.6Hz,1H),7.51(d,J=3.6Hz,1H),7.24(s,1H),4.02(s,3H),1.63(s,9H).
中间体5:
Figure PCTCN2021110751-appb-000163
在室温下,向1,2-二氯乙烷(5mL)溶液依次加入实施例1的中间体7(100mg)、中间体4(131.21mg),反应在室温下进行16小时,然后再加入三醋酸硼氢化钠(43.12mg),室温继续搅拌16小时。反应结束后,反应液直接进行减压浓缩并通过制备TLC进行分离纯化(二氯甲烷:甲醇=5:1)得到中间体5(30mg,收率12.8%)。MS m/z(ESI):591.1[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000164
将氢氧化钠(11.99mg)加到中间体5(60mg)的甲醇(1mL)和水(1mL)溶液中,反应体系加热到75℃并在该温度下搅拌3小时。反应结束后,反应液直接进行减压浓缩,残余物通过Prep-HPLC进行分离纯化(色谱柱:Xbridge-C18 150 x 19mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:20-40%)得到目标化合物(18.8mg,收率31.3%)。MS m/z(ESI):477.1[M+H]. 1H NMR(400MHz,CD 3OD)δ8.15(d,J=8.0Hz,2H),7.66(d,J=8.0Hz,2H),7.44(d,J=3.2Hz,1H),7.17(s,1H),6.52(s,1H),4.67–4.56(m,1H),4.36–4.15(m,2H),3.87–3.77(m,4H),3.64–3.55(m,2H),3.50–3.40(m,1H),3.24–3.17(m,1H),2.28–2.19(m,2H),2.04–2.00(m,2H),1.30(t,J=6.8Hz,3H).
实施例12:
中间体1:
Figure PCTCN2021110751-appb-000165
将4-溴-2-甲氧基-苯甲醛(11g)加入到250mL单口瓶中,加入硫酸(100mL),冰浴下缓慢加硝酸钾(5.5g)到反应体系中,撤掉冰浴继续反应1小时。反应结束后将反应液缓慢倒入冰水中,用乙酸乙酯(200mL)萃取两遍,有机相用饱和碳酸氢钠水溶液(100mL)洗涤,饱和食盐水(50mL)洗涤三遍,无水硫酸钠干燥并过滤,过滤液减压浓缩得到中间体1(12g,收率:86%)。 1H NMR(400MHz,CD 3OD)δ10.24(s,1H),8.29(s,1H),7.78(s,1H),4.07(s,3H).
中间体2:
Figure PCTCN2021110751-appb-000166
在500mL单口瓶中,依次加入甲苯(200mL)、乙二醇(30g)、中间体1(10g)和对甲苯磺酸(0.76g),反应体系加热到120℃并在该温度下进行反应4小时。反应结束后,浓缩除去溶剂,加入水(500mL)和乙酸乙酯(500mL)稀释并分相,有机相用无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余相用硅胶柱分离纯化(石油醚:乙酸乙酯=3:1)得到中间体2(10g,81%)。MS m/z(ESI):304.0[M+H].
中间体3:
Figure PCTCN2021110751-appb-000167
将中间体2(3g)的四氢呋喃(100mL)溶液降温至-30℃,缓慢滴加乙烯基溴化镁(1M,60mL)到反应体系中,反应在该温度下搅拌3小时,然后自然升温到0℃。反应结束后,向反应体系中加入饱和氯化铵水溶液(200mL)淬灭反应,反应液用乙酸乙酯(500mL)萃取两遍,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥并过滤,过滤液浓缩,残余相用硅胶柱分离纯化(二氯甲烷)得到中间体3(1.2g,收率:45%)。MS m/z(ESI):253.8[M+H].
中间体4:
Figure PCTCN2021110751-appb-000168
向10mL微波管中,依次加入N,N-二甲基甲酰胺(4mL)、中间体3(340mg)、氰化锌(230mg)和四三苯基膦钯(115mg),反应体系在氮气保护下加热到130℃进行微波反应1.5小时。反应结束后,反应液直接用硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体4(220mg,收率:78%)。MS m/z(ESI):201.1[M+H].
中间体5:
Figure PCTCN2021110751-appb-000169
依次将中间体4(130mg)和实施例1的中间体7(110mg)加入到向1,2二氯乙烷(3mL)中,反应在室温下搅拌8小时,然后三乙酰氧基硼氢化钠(440mg)加到反应体系中,在室温下继续反应16小时。反应结束后,反应液直接用硅胶柱分离纯化(甲醇:二氯甲烷=1:20)得到中间体5(200mg,41%)。MS m/z(ESI):447.9[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000170
向25mL单口瓶中,依次加入甲醇(3mL)、水(3mL)、中间体5(100mg)和氢氧化钠(40mg),反应体系加热到75℃并在该温度下进行反应3小时。反应结束后,反应液直接通过Prep-HPLC进行分离纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:15-45%),得到的溶液浓缩,剩余少量水溶液冷冻干燥得到目标化合物(60.2mg,收率:62.5%,含0.3当量的甲酸)。MS m/z(ESI):434.1[M+H]. 1H NMR(400MHz,CD 3OD)δ8.42(s,0.3H),8.11(d,J=8.0Hz,2H),7.63(d,J=8.0Hz,2H),7.46(d,J=3.2Hz,1H),7.28(s,1H),6.57(s,1H),4.53-4.38(m,1H),4.25–4.02(m,2H),3.82–3.76(m,4H),3.62-3.54(m,2H),3.30–3.23(m,1H),3.14–3.04(m,1H),2.23–1.87(m,4H),1.29(t,J=7.0Hz,3H).
实施例13:
中间体1:
Figure PCTCN2021110751-appb-000171
向实施例13的中间体3(500mg)的二氧六环(9mL)溶液中依次加入水(1mL)、环丙基硼酸(200mg)、碳酸钠(500mg)和Pd(dppf)Cl 2二氯甲烷(80mg)。混合物加热到90℃并在该温度下搅拌16小时。反应结束后直接加入乙酸乙酯(20mL)稀释,加入硅胶进行拌样,然后用硅胶柱分离纯化(乙酸乙酯:石油醚=1:10)得到中间体1(340mg,收率:74%)。MS m/z(ESI):215.9[M+H].
中间体2:
Figure PCTCN2021110751-appb-000172
室温下,向25mL单口瓶中依次加入溶剂1,2二氯乙烷(3mL)、中间体1(150mg)和实施例1的中间体7(130mg)。反应在室温进行8小时后,加入三乙酰氧基硼氢化钠(450mg),在室温下继续反应16小时。反应结束后,反应液直接加入硅胶拌样,用硅胶柱分离纯化(甲醇:二氯甲烷=1:20)得到中间体2(100mg,28%)。MS m/z(ESI):462.8[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000173
室温下,向25mL单口瓶中,依次加入甲醇(2mL)、水(2mL)、中间体2(100mg)和氢氧化钠(50mg),反应体系加热到40℃并在该温度下搅拌3小时。反应结束后,反应液直接通过Prep-HPLC纯化(色谱柱:Gemini-C18,150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:15-35%),得到的溶液浓缩,剩余少量水溶液冷冻干燥得到目标化合物(20.1mg,收率:16%,含0.8当量的甲酸)。MS m/z(ESI):448.9[M+H]. 1H NMR(400MHz,CD 3OD)δ8.43(bs,0.8H),8.17(d,J=8.0Hz,2H),7.67(d,J=8.0Hz,2H),7.36(d,J=2.8Hz,1H),6.54(s,1H),6.37(d,J=2.8Hz,1H),4.80-4.71(m,1H),4.37–4.19(m,2H),3.87–3.81(m,1H),3.76(s,3H),3.65–3.48(m,3H),3.42–3.36(m,1H),2.30–1.94(m,5H),1.33(t,J=7.2Hz,3H),1.11–1.03(m,2H),0.82–0.75(m,2H).
实施例14:
中间体1:
Figure PCTCN2021110751-appb-000174
室温下,向4-溴萘-2-醇(970mg)和碘甲烷(1.23g)的N,N-二甲基甲酰胺(10mL)溶液中缓慢加入氢化钠(349mg),反应体系在氮气气氛和室温条件下搅拌2个小时。反应结束后,反应液倒入水(5mL)中淬灭,用乙酸乙酯萃取(100ml)三次,合并的有机相用饱和食盐水(20mL)洗涤一次,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物经柱层析法(石油醚:乙酸乙酯=100:1)得到中间体1(920mg,收率:89%)。MS m/z(ESI):236.9[M+H]
中间体2:
Figure PCTCN2021110751-appb-000175
在100mL单口瓶中,依次加入N,N-二甲基甲酰胺(12mL)、中间体1(1.05g)、三甲基环三硼氧烷(3.3g)、碳酸铯(2.87g)和四(三苯基膦)钯(254mg),反应体系在氮气保护下加热到90℃并在该温度下进行搅拌16个小时。反应结束后,反应液降温至室温,倒入水(50mL)中,用乙酸乙酯萃取(100ml)三次,合并的有机相用饱和食盐水洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=5:1)得到中间体2(680mg,收率:80%)。MS m/z(ESI):173.1[M+H].
中间体3:
Figure PCTCN2021110751-appb-000176
在100mL单口瓶中,依次加入N,N-二甲基甲酰胺(4.2mL)和三氯氧磷(3.8mL),反应体系在氮气保护和室温条件下搅拌15分钟,然后加入中间体2(660mg),反应加热到60℃并在该温度下继续搅拌1小时。反应结束后,反应液倒入冰水(30mL)中,用乙酸乙酯萃取(100mL)三次,合并的有机相用饱和食盐水洗涤,无水硫酸钠干燥并过滤,过滤液进行减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)纯化得到中间体3(430mg,收率:56%)。MS m/z(ESI):201.1[M+H].
中间体4:
Figure PCTCN2021110751-appb-000177
在50mL的单口瓶中,依次加入1,2-二氯乙烷(4mL)、中间体3(100mg)、实施例1的中间体7(197mg)和三乙酰氧基硼氢化钠(318mg)。反应体系在氮气保护和室温下搅拌16小时。反应结束后,反应液直接进行减压浓缩,残余物经硅胶柱分离纯化(石油醚:乙酸乙酯=10:1)得到中间体4(120mg,收率:53%)。MS m/z(ESI):448.1[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000178
在50mL单口瓶中,依次加入甲醇(3mL)、水(1mL)、中间体4(120mg)和氢氧化钠(214mg)。反应体系在室温下搅拌16小时。反应结束后,反应液加入水(5mL)稀释,1M盐酸调节pH到7-8,然后混合物直接进行减压浓缩,残余物经Prep-HPLC进行分离纯化(色谱柱:Gemini-C18 150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:20-40%;UV:214nm)纯化得到目标化合物(93.5mg,收率:79%)。MS m/z(ESI):434.0[M+H]. 1H NMR(400MHz,DMSO-d 6)δ12.89(s,1H),8.00(d,J=8.0Hz,2H),7.96(d,J=8.4Hz,1H),7.89(d,J=8.4Hz,1H),7.69(d,J=8.0Hz,2H),7.45(t,J=7.2Hz,1H),7.35(t,J=7.2Hz,1H),7.26(s,1H),3.87(s,3H),3.63–3.50(m,4H),3.43(q,J=7.2Hz,2H),2.63(s,3H),2.44–2.30(m,2H),1.87–1.77(m,2H),1.70-1.62(m,1H),1.47–1.37(m,1H),1.68(t,J=7.0Hz,3H).
实施例15:
中间体1:
Figure PCTCN2021110751-appb-000179
将N-溴代丁二酰亚胺(125mg)加入到到1,3-二甲基萘(100mg)的乙腈(6mL)溶液中,反应体系在室温下搅拌16小时。反应结束后,反应液直接进行减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=20:1)得到中间体1(160mg,收率85%)。GC-MS:234.0,236.0(MS)。
中间体2:
Figure PCTCN2021110751-appb-000180
-78℃下将正丁基锂(0.37mL)缓慢滴加到中间体1(160mg)的四氢呋喃(5mL)溶液中;该反应在-78℃搅拌1小时后,将N,N-二甲基甲酰胺(99mg)缓慢滴加到反应液中,然后反应混合物自然升温至室温并在该温度下搅拌16小时。反应结束后,反应混合液倒入水中(20mL),用乙酸乙酯萃取(15mL)两次。合并的有机相用饱和食盐水洗涤(15mL)三次,无水硫酸钠干燥并过滤,过滤液减压浓缩得到中间体2(120mg,收率95%)。MS m/z(ESI):185.1[M+H].
中间体3:
Figure PCTCN2021110751-appb-000181
室温下,将钛酸四乙酯(118mg)缓慢加到中间体2(100mg)和实施例1的中间体7(142mg)的四氢呋喃(10mL)溶液中,反应加热到70℃并在该温度下搅拌16小时。反应自然冷却至室温后,加入三乙酰硼氢化钠(343mg),反应加热到70℃并在该温度下继续搅拌1小时。反应结束后,反应液直接进行减压浓缩,残余物用硅胶柱分离纯化(石油醚:乙酸乙酯=2:1)得到中间体3(30mg,收率13%)。MS m/z(ESI):431.9[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000182
在中间体3(30mg)的甲醇(4mL)溶液中加入氢氧化钠(32mg)的水(1mL)溶液,反应体系在室温下搅拌16小时。反应结束后,减压浓缩除去甲醇,水相用5M的盐酸溶液调节pH至5-6,然后将混合物进行减压浓缩,残余物用Prep-HPLC进行分离纯化(色谱柱:Gemini-C18 150 x 21.2mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:5-30%)得到目标化合物(15mg,收率50%)。MS m/z(ESI):417.8[M+H]. 1H NMR(400MHz,CD 3OD)δ8.27(d,J=7.8Hz,2H),8.08–8.02(m,1H),7.87–7.80(m,2H),7.58–7.50(m,3H),7.29(s,1H),4.80-4.65(m,1H),4.55-4.38(m,1H),3.84(s,1H),3.71–3.49(m,3H),3.17–3.09(s,1H),2.65(s,3H),2.54–2.22(m,6H),2.10–1.85(m,2H),1.30(t,J=7.0Hz,3H).
实施例16:
中间体1:
Figure PCTCN2021110751-appb-000183
室温下,向3,3-二乙氧基丙酸乙酯(20g)的水(50mL)溶液中加入氢氧化钠(5g),反应加热到110℃并在该温度下搅拌1小时。然后反应体系自然降温到室温,加入水(2L)进行稀释,用乙酸乙酯(1L)萃取,萃取相用饱和食盐水(1L)洗涤,无水硫酸钠干燥并过滤;向滤液直接浓缩得到的粗品中加入氯化亚砜(30mL),反应加热到70℃并在该温度下继续搅拌一小时。反应结束后,反应液直接减压浓缩得到棕色固体中间体1(14g,收率90%)。
中间体2:
Figure PCTCN2021110751-appb-000184
在冰浴条件和氮气保护下,向2-甲基-4-甲氧基苯胺(14g)和吡啶(16g)的二氯甲烷(200mL)溶液中慢慢加入中间体1(14g)的二氯甲烷(200mL)溶液,反应在室温下搅拌16小时。反应结束后,加水(500mL)淬灭,用二氯甲烷(500mL)萃取, 萃取相用水(500mL)洗涤,无水硫酸钠干燥并过滤,过滤液浓缩并通过硅胶柱(二氯甲烷:甲醇=10:1)纯化得到黄色油状中间体2(12g,收率50%)。MS m/z(ESI):236.1[M+H]。
中间体3:
Figure PCTCN2021110751-appb-000185
在冰浴下,将中间体2(12g)加到硫酸(50mL)溶液中,反应在室温搅拌1小时。反应结束后,反应液倒入冰水中(200mL),析出的固体进行过滤,用水洗涤并干燥得到棕色固体中间体3(3g,收率30%)。MS m/z(ESI):190.1[M+H]。
中间体4:
Figure PCTCN2021110751-appb-000186
向中间体3(600mg)的醋酸(10mL)溶液中加入Pd/C(34mg),反应在90℃和氢气气球下进行催化氢化反应32小时。反应结束并自然降温到室温后,反应液过滤,过滤液减压浓缩,残余物通过硅胶柱(石油醚:乙酸乙酯=5:1)纯化得到黄色固体中间体4(100mg,收率16%)。MS m/z(ESI):192.0[M+H]。
中间体5:
Figure PCTCN2021110751-appb-000187
在-78℃和氮气保护下,向中间体4(500mg)的二氯甲烷(3mL)溶液中加入四氯化钛(1.5g),反应液搅拌5分钟后,再慢慢加入二氯甲基甲醚(900mg,7.80mmol)。反应混合物自然升温至室温,并在室温下继续搅拌16小时。反应结束后,反应液二氯甲烷(20mL)稀释,用(5mL)洗涤,Na 2SO 4干燥并过滤,滤液浓缩,得到的残余物通过硅胶柱(二氯甲烷:甲醇=10:1)纯化得到黄色固体中间体5(35mg,收率7%)。MS m/z(ESI):220.0[M+H]。
中间体6:
Figure PCTCN2021110751-appb-000188
在氮气保护下,向中间体5(35mg)的1,2二氯乙烷(5mL)溶液中加入实施例1的中间体7(42mg),反应液在室温搅拌16小时后,然后加入醋酸硼氢化钠(18.15mg),反应在室温下继续搅拌16小时。反应结束后,反应液直接浓缩,残余物通过硅胶柱(二氯甲烷:甲醇=10:1)纯化得到黄色固体中间体6(30mg,收率86%)。MS m/z(ESI):467.2[M+H].
目标化合物:
Figure PCTCN2021110751-appb-000189
在50mL单口瓶中,依次加入甲醇(1mL)、水(1mL)、中间体6(30mg)和氢氧化钠(7.7mg),反应混合物室温搅拌3小时。反应结束后,在冰浴下,将稀盐酸(1M,0.5mL)加入到反应液中调节pH至中性,然后直接减压浓缩,得到的残余物通过制备高压液相色谱纯化(色谱柱:-Xbridge-C18 150 x 19mm,5um;流动项:乙腈-水(0.1%甲酸);梯度:20-40%)得到白色固体目标化合物(1.1mg,收率3.66%,含0.4当量的甲酸)。 1H NMR(400MHz,CD 3OD)δ8.48(bs,1H),8.07(d,J=8.0Hz,2H),7.56(d,J=8.0Hz,2H),6.74(s,1H),4.40–4.30(m,1H),3.92–3.83(m,2H),3.82–3.76(m,1H),3.72(s,3H),3.60–3.52(m,2H),3.15-2.87(m,3H),2.81–2.70(m,1H),2.50-2.41(m,2H),2.25(s,3H),2.17-2.11(m,2H),2.06–1.96(m,2H),1.27(t,J=7.0Hz,3H).MS m/z(ESI):453.1[M+H].
下述实施例参照上述方法制备得到
Figure PCTCN2021110751-appb-000190
Figure PCTCN2021110751-appb-000191
Figure PCTCN2021110751-appb-000192
Figure PCTCN2021110751-appb-000193
Figure PCTCN2021110751-appb-000194
Figure PCTCN2021110751-appb-000195
生物学试验:
1.光学表面等离子共振(SPR)结合力检测
SPR实验在25℃条件下,以补充有0.05%(v/v)P20和5%DMSO的PBS缓冲液作为运行缓冲液,采用的分析仪器为GE Healthcare的Biacore 8K。400mM EDC和100mM NHS以30μL/min的流速活化CM7芯片(GE Healthcare)420s。补体B因子用10mM醋酸钠(pH 4.0)稀释到50μg/mL,然后以10μL/min的流速偶联1200s,使补体B因子共价固定到检测芯片上(蛋白固化水平为25000RU);然后检测芯片用1M盐酸乙醇胺以10μL/min的流速作用300s进行芯片封闭。待测化合物浓度为500μM,结合时间120s,解离时间300s。数据分析采用的是1:1binding结合模型进行分析(Biacore Insight Evalution Software,Version2.0.15.12933)。
2.TR-FRET结合力检测
以Cy5荧光标记的小分子抑制剂作为探针的竞争性结合实验来筛选化合物对人补体因子B的抑制活性。补体因子B与EZ-Link TM Sulfo-NHS-LC-LC-Biotin以1:2比例在冰上孵育1小时后加入1M Tris(pH7.5)终止反应。随后用2mL Zeba TM desalt spin column纯化两次获得生物素标记的补体因子B(EZ-LinkTM Sulfo-NHS-LC-Biotin说明书)。实验时,取终浓度为10nM生物素标记的补体因子B与不同浓度的化合物在缓冲液中室温预孵育1小时。加入终浓度分别为75nM和5nM的Cy5荧光标记的探针和铕螯合物标记的链霉亲和素(石油醚rkin Elmer,#AD0060),启动反应。在酶标仪上(337nm激发光,665nm发射光,70μs time-gated)进行动力学读数,读取时间依赖的荧光能量转移(TR-FRET)的数据,确定IC50。
3.补体***水解C3活性检测
受试化合物测试浓度为10μM起始,3倍稀释,7个浓度点,单孔检测。在96孔板用DMSO将受试化合物稀释成1000倍终浓度的溶液,再用Diluent(
Figure PCTCN2021110751-appb-000196
SYSTEM ALTERNATIVE PATHWAY AP330)稀释成5倍终浓度的溶液。转移30uL到96孔板中,加入120μL的备用血清,室温孵育15分钟。阳性对照孔加30μL的5‰DMSO和120μL的备用血清,阴性对照孔加30μL的5‰DMSO和120μL的Diluent。(3)取100μL加入反应板中,37℃孵育60分钟。弃去孔中液体,每孔用300μL洗涤液洗涤3次。每孔加入100μL Conjugate(
Figure PCTCN2021110751-appb-000197
SYSTEM ALTERNATIVE PATHWAY AP330),室温孵育30分钟。去孔中液体,每孔用300μL洗涤液洗涤3次。然后每孔加入100μL底物,室温孵育30分钟。使用酶标仪(Perkin Elmer,EnSight)检测,读取OD405值。
4.补体溶血活性检测
溶血实验参考Xuan Yuan等,Haematologica.(2017)102:466-475描述,在实验之前, 实现兔红细胞(RE)的100%裂解所需的正常人血清(NHS)的最佳浓度通过滴定测试获得。在该实验中,NHS(取自健康志愿者)在含10mM Mg-EGTA的GVB0缓冲液(0.1%明胶,5mM Veronal,145mM NaCl,0.025%NaN 3,pH 7.3,Complement technology)中稀释并与各种浓度梯度的测试化合物在37℃孵育15分钟。新悬浮在含10mM Mg-EGTA的GVB0缓冲液中的RE(取自健康日本大耳白兔)添加达到1×108细胞/ml的终浓度且在37℃下孵育30分钟。阳性对照组(100%裂解)由具有NHS和RE但没有测试化合物的含10mM Mg-EGTA的GVB0缓冲液组成;阴性对照组(0%裂解)由具有灭活的NHS(56℃加热30分钟或65℃加热5分钟)和RE但没有测试化合物的含10mM Mg-EGTA的GVB0缓冲液组成。样品以2000g离心5分钟后收集上清液。415nm的吸光度(A415)使用酶标仪(Molecular Devices,SpectraMax i3X)检测。IC 50值通过非线性回归从作为测试化合物浓度的函数的溶血百分比计算。测试结果见表1。
表1:
Figure PCTCN2021110751-appb-000198
虽然以上描述了本发明的具体实施方式,但是本领域的技术人员应当理解,这些仅是举例说明,在不背离本发明的原理和实质的前提下,可以对这些实施方式做出多种变更或修改。因此,本发明的保护范围由所附权利要求书限定。

Claims (10)

  1. 一种如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,其任选在药学上可接受的载体中;
    Figure PCTCN2021110751-appb-100001
    其中,
    W为O或C(R 7’R 7”);
    R 7、R 7’和R 7”独立地为氢、羟基、卤素、C 1-C 4烷基或C 1-C 4烷基-O-;
    R 6为氢、C 1-C 4烷基或羟基C 1-C 4烷基;
    R 4’和R 4独立地为氢;
    m为0、1或2;
    R 5
    Figure PCTCN2021110751-appb-100002
    环B为苯基或6元杂芳基;所述的6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;
    R b为H、羟基、=O,或,与环B并环连接的:苯基、3-6元环烷基、5-6元杂环烷基或5-6元杂芳基;所述5-6元杂环烷基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;当取代基为多个时,相同或不同;
    A为
    Figure PCTCN2021110751-appb-100003
    Z 1为C(R 21)或N;Z 2为C(R 51)或N;R 41为NH 2或C(=O)NH 2
    环A 1为吡啶基;其中,Z 3为C(R 22)或N;Z 4和Z 5独立地为C或N;
    R 21、R 22和R 51独立地为氢;
    环A 2为5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基,或者,被一个或多个R a1取代的:5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基;所述的5-6元杂环烷基和被一个或多个R a1取代的5-6元杂环烷基里的5-6元杂环烷基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂环烯基和 被一个或多个R a1取代的5-6元杂环烯基里的5-6元杂环烯基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂芳基和被一个或多个R a1取代的5-6元杂芳基里的5-6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;当取代基为多个时,相同或不同;
    环A 3为5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
    Figure PCTCN2021110751-appb-100004
    或者,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
    Figure PCTCN2021110751-appb-100005
    所述的5-6元杂环烷基和被一个或多个R a2取代的5-6元杂环烷基里的5-6元杂环烷基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的5-6元杂环烯基和被一个或多个R a2取代的5-6元杂环烯基里的5-6元杂环烯基中,杂原子个数为1、2或3个,杂原子选自N、O、S、S(=O)和S(=O) 2中的一种或多种;所述的6元杂芳基和被一个或多个R a2取代的5-6元杂芳基里的6元杂芳基中,杂原子个数为1、2或3个,杂原子选自N、O和S中的一种或多种;当取代基为多个时,相同或不同;环A 3与苯环并环连接;
    A 3’为5元杂芳基;所述的5元杂芳基中,杂原子个数为1或2个,杂原子选自N、O和S中的一种或多种;且Z 7为N、O或S,和/或,Z 6为CH、O或S;
    R a1和R a2独立地为羟基、=O、卤素、CN、C 1-C 4烷基或C 1-C 4烷基-O-;
    R 11、R 31、R 12、R 32、R 13和R 33独立地为C 1-C 4烷基、C 1-C 4烷基-O-或3-6元环烷基;
    Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基;
    带“*”碳原子表示当为手性碳原子时,为S构型、R构型或它们的混合物。
  2. 如权利要求1所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,其任选在药学上可接受的载体中;其特征在于,其中,
    W为C(R 7’R 7”);
    和/或,R 7’和R 7”独立地为氢、卤素、C 1-C 4烷基或C 1-C 4烷基-O-;
    和/或,R 7为氢;
    和/或,m为1;
    和/或,R b为H;
    和/或,Z 1为CH、Z 2为N,或Z 1为CH、Z 2为CH,或Z 1为N、Z 2为CH;
    和/或,Z 4为N,或Z 5为N;
    和/或,环A 2为5-6元杂芳基;或者,环A 2为5-6元杂环烷基、5-6元杂环烯基或6元杂芳基,或,被一个或多个R a1取代的:5-6元环烯基、5-6元杂环烷基、5-6元杂环烯基或6元杂芳基;
    和/或,环A 3为5-6元杂环烷基、5-6元杂环烯基或
    Figure PCTCN2021110751-appb-100006
    或,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
    Figure PCTCN2021110751-appb-100007
    和/或,R a1和R a2独立地为羟基、卤素、=O或C 1-C 4烷基;
    和/或,R 11、R 12和R 13独立地为C 1-C 4烷基或C 1-C 4烷基-O-;
    和/或,R 31、R 32和R 33独立地为C 1-C 4烷基;
    和/或,R 14为C 1-C 4烷基-O-;
    和/或,Z 8为N,R 34为C 1-C 4烷基;或Z 8为CH,R 34为3-6元环烷基。
  3. 如权利要求1所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,其任选在药学上可接受的载体中;其特征在于,其中,所述的如式I所示的杂环类化合物为如下任一方案:
    方案1、
    所述的如式I所示的杂环类化合物为如下式Ia、Ib或Ic所示;
    Figure PCTCN2021110751-appb-100008
    方案2、
    W为C(R 7’R 7”);R 7为氢;
    R 7’和R 7”独立地为氢、卤素、C 1-C 4烷基或C 1-C 4烷基-O-;
    R 6为氢;
    R 4’和R 4独立地为氢;
    m为1;
    R 5
    Figure PCTCN2021110751-appb-100009
    环B为苯基或6元杂芳基;
    R b为H、羟基或=O,或,与环B并环连接的:苯基、3-6元环烷基、5-6元杂环烷基或5-6元杂芳基;
    A为
    Figure PCTCN2021110751-appb-100010
    Z 1为CH或N;Z 2为CH或N;R 41为NH 2或C(=O)NH 2
    环A 1为吡啶基;Z 3为CH或N;Z 4和Z 5独立地为C或N;
    环A 2为5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基,或者,被一个或多个R a1取代的:5-6元杂环烷基、5-6元杂环烯基或5-6元杂芳基;
    环A 3为5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
    Figure PCTCN2021110751-appb-100011
    或者,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
    Figure PCTCN2021110751-appb-100012
    R a1和R a2独立地为羟基、卤素、=O或C 1-C 4烷基;
    R 11、R 31、R 12、R 32、R 13、R 23和R 33独立地为C 1-C 4烷基、C 1-C 4烷基-O-或3-6元环烷基;
    Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基;
    方案3、
    W为C(R 7’R 7”);R 7为氢;
    R 7’和R 7”独立地为氢或C 1-C 4烷基-O-;
    R 6为氢;
    R 4’和R 4独立地为氢;
    m为1;
    R 5
    Figure PCTCN2021110751-appb-100013
    A为
    Figure PCTCN2021110751-appb-100014
    环A 1为吡啶基;Z 3为N;Z 4和Z 5为C;
    环A 2为5元杂芳基或被一个或多个R a1取代的5元杂芳基;
    环A 3为5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
    Figure PCTCN2021110751-appb-100015
    或者,被一个或多个R a2取代的:5-6元杂环烷基、5-6元杂环烯基、6元杂芳基或
    Figure PCTCN2021110751-appb-100016
    R a1和R a2独立地为羟基、=O或C 1-C 4烷基;
    R 12和R 13独立地为C 1-C 4烷基-O-;
    R 32和R 33独立地为C 1-C 4烷基;
    Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基;
    方案4、
    W为C(R 7’R 7”);R 7为氢;
    R 7’和R 7”独立地为氢或C 1-C 4烷基-O-;
    R 6为氢;
    R 4’和R 4独立地为氢;
    m为1;
    R 5
    Figure PCTCN2021110751-appb-100017
    A为
    Figure PCTCN2021110751-appb-100018
    环A 3为5元杂环烷基、5元杂环烯基或
    Figure PCTCN2021110751-appb-100019
    R 13为C 1-C 4烷基-O-;
    R 33为C 1-C 4烷基;
    Z 8为CH或N;R 14为C 1-C 4烷基-O-;R 23和R 24为H;R 34为C 1-C 4烷基或3-6元环烷基。
  4. 如权利要求1-3中任一项所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,其任选在药学上可接受的载体中;其特征在于,其中,
    当R 7、R 7’和R 7”独立地为C 1-C 4烷基或C 1-C 4烷基-O-时,所述的C 1-C 4烷基或C 1-C 4烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基;
    和/或,当R 6为C 1-C 4烷基或羟基C 1-C 4烷基时,所述的C 1-C 4烷基和羟基C 1-C 4烷基里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基;
    和/或,当环B为6元杂芳基时,所述的6元杂芳基为吡啶基;例如
    Figure PCTCN2021110751-appb-100020
    Figure PCTCN2021110751-appb-100021
    和/或,当R b为3-6元环烷基时,所述的3-6元环烷基为环丙基、环丁基、环戊基和环己基;例如环戊基;
    和/或,当R b为5-6元杂环烷基时,所述的5-6元杂环烷基为四氢呋喃基;例如
    Figure PCTCN2021110751-appb-100022
    和/或,当R b为5-6元杂芳基时,所述的5-6元杂芳基为吡啶基或咪唑基;例如
    Figure PCTCN2021110751-appb-100023
    Figure PCTCN2021110751-appb-100024
    和/或,当环A 2为5-6元杂环烷基时,所述的5-6元杂环烷基为
    Figure PCTCN2021110751-appb-100025
    Figure PCTCN2021110751-appb-100026
    和/或,当环A 2为5-6元杂环烯基时,所述的5-6元杂环烯基为
    Figure PCTCN2021110751-appb-100027
    Figure PCTCN2021110751-appb-100028
    和/或,当环A 2为5-6元杂芳基时,所述的5-6元杂芳基为
    Figure PCTCN2021110751-appb-100029
    Figure PCTCN2021110751-appb-100030
    和/或,当环A 3为5-6元杂环烷基时,所述的5-6元杂环烷基为
    Figure PCTCN2021110751-appb-100031
    Figure PCTCN2021110751-appb-100032
    和/或,当环A 3为5-6元杂环烯基时,所述的5-6元杂环烯基为
    Figure PCTCN2021110751-appb-100033
    Figure PCTCN2021110751-appb-100034
    Figure PCTCN2021110751-appb-100035
    和/或,当环A 3为6元杂芳基时,所述的6元杂芳基为
    Figure PCTCN2021110751-appb-100036
    和/或,当环A 3
    Figure PCTCN2021110751-appb-100037
    所述的A 3’
    Figure PCTCN2021110751-appb-100038
    Figure PCTCN2021110751-appb-100039
    和/或,当R a1和R a2独立地为C 1-C 4烷基或C 1-C 4烷基-O-时,所述的C 1-C 4烷基或C 1-C 4烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基;
    和/或,当R 11、R 31、R 12、R 32、R 13、R 23和R 33独立地为C 1-C 4烷基、C 1-C 4烷基-O-时,所述的C 1-C 4烷基和C 1-C 4烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基;
    和/或,当R 11、R 31、R 12、R 32、R 13、R 23和R 33独立地为3-6元环烷基时,所述的3-6元环烷基为环丙基、环丁基、环戊基和环己基;例如环丙基;
    和/或,当R 14为C 1-C 4烷基-O-时,所述的C 1-C 4烷基-O-里的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基;
    和/或,当R 34为C 1-C 4烷基时,所述的C 1-C 4烷基为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基;例如甲基;
    和/或,当R 34为3-6元环烷基时,所述的3-6元环烷基为环丙基、环丁基、环戊基和环己基;例如环丙基。
  5. 如权利要求4所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,其任选在药学上可接受的载体中;其特征在于,其中,
    R 7’和R 7”独立地为氢、F、甲基、乙基-O-;例如,W为
    Figure PCTCN2021110751-appb-100040
    Figure PCTCN2021110751-appb-100041
    亚甲基;
    和/或,R 5
    Figure PCTCN2021110751-appb-100042
    Figure PCTCN2021110751-appb-100043
    例如,
    Figure PCTCN2021110751-appb-100044
    Figure PCTCN2021110751-appb-100045
    Figure PCTCN2021110751-appb-100046
    Figure PCTCN2021110751-appb-100047
    和/或,R a1和R a2独立地为羟基、=O或甲基;
    和/或,R 11、R 12和R 13独立地为甲基、甲基-O-或环丙基;
    和/或,R 31、R 32和R 33独立地为甲基;
    和/或,R 14为甲基-O-;
    和/或,R 34为甲基或环丙基;
    和/或,当A为
    Figure PCTCN2021110751-appb-100048
    时,为
    Figure PCTCN2021110751-appb-100049
    Figure PCTCN2021110751-appb-100050
    和/或,当A为
    Figure PCTCN2021110751-appb-100051
    时,为
    Figure PCTCN2021110751-appb-100052
    Figure PCTCN2021110751-appb-100053
    和/或,当A为
    Figure PCTCN2021110751-appb-100054
    时,为
    Figure PCTCN2021110751-appb-100055
    Figure PCTCN2021110751-appb-100056
    Figure PCTCN2021110751-appb-100057
    和/或,当A为
    Figure PCTCN2021110751-appb-100058
    时,为
    Figure PCTCN2021110751-appb-100059
  6. 如权利要求1所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,其任选在药学上可接受的载体中;其特征在于,其中,所述的如式I所示的杂环类化合物为如下任一结构,
    Figure PCTCN2021110751-appb-100060
    Figure PCTCN2021110751-appb-100061
    Figure PCTCN2021110751-appb-100062
    Figure PCTCN2021110751-appb-100063
    Figure PCTCN2021110751-appb-100064
    Figure PCTCN2021110751-appb-100065
    和/或,所述的如式I所示的杂环类化合物的药学上可接受的盐为如下任一结构,
    Figure PCTCN2021110751-appb-100066
    Figure PCTCN2021110751-appb-100067
  7. 一种如权利要求1-6中任一项所述的如式I所示的杂环类化合物的制备方法,其特征在于,其包括如下步骤:
    在溶剂中,在碱存在下,将如式II所示的化合物进行如下所示的脱酯基反应,得到所述的如式I所示的杂环类化合物即可;
    Figure PCTCN2021110751-appb-100068
    其中,R 8为C 1-C 4的烷基;R b、R 4、R 4’、R 6、R 7、A、W、m和*的定义均如权利要求1-6中任一项所示。
  8. 一种如式II所示的杂环类化合物,
    Figure PCTCN2021110751-appb-100069
    其中,R 8为C 1-C 4的烷基;R b、R 4、R 4’、R 6、R 7、A、W、m和*的定义均如权利要求1-6中任一项所示;
    例如,
    Figure PCTCN2021110751-appb-100070
    Figure PCTCN2021110751-appb-100071
  9. 一种药物组合物,其包括如权利要求1-6中任一项所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药,和一种或多种药学上可接受的载体。
  10. 一种如权利要求1-6中任一项所述的如式I所示的杂环类化合物或其药学上可接受的盐、同位素类似物、前药在作为补体因子B抑制剂或在制备药物中的应用;其中,所述的药物可为用于治疗与补体因子B的异常激活、或者与补体因子B执行正常功能的中出现的相关疾病的药物;或者,所述的药物可为用于治疗血液性、自身免疫性、炎性和神经变性等疾病的药物。
PCT/CN2021/110751 2020-08-07 2021-08-05 一种杂环类化合物、其制备方法及用途 WO2022028507A1 (zh)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US18/040,966 US20230331710A1 (en) 2020-08-07 2021-08-05 Heterocyclic compound, preparation method and use thereof
EP21853576.3A EP4194449A4 (en) 2020-08-07 2021-08-05 HETEROCYCLIC COMPOUND, PREPARATION METHOD AND USE THEREOF

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202010787537 2020-08-07
CN202010787537.2 2020-08-07
CN202110293149 2021-03-18
CN202110293149.3 2021-03-18

Publications (1)

Publication Number Publication Date
WO2022028507A1 true WO2022028507A1 (zh) 2022-02-10

Family

ID=80120027

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/110751 WO2022028507A1 (zh) 2020-08-07 2021-08-05 一种杂环类化合物、其制备方法及用途

Country Status (4)

Country Link
US (1) US20230331710A1 (zh)
EP (1) EP4194449A4 (zh)
CN (1) CN114057692B (zh)
WO (1) WO2022028507A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023020566A1 (zh) 2021-08-18 2023-02-23 四川海思科制药有限公司 一种苯并氮杂芳环衍生物及其在医药上的应用
WO2023072197A1 (en) * 2021-10-27 2023-05-04 Hansoh Bio Llc Piperidinyl indole derivatives, preparation methods and medicinal uses thereof
WO2023246677A1 (zh) * 2022-06-20 2023-12-28 深圳信立泰药业股份有限公司 一种吲哚-苯基哌啶化合物及其制备方法与应用
WO2024051849A1 (en) * 2022-09-10 2024-03-14 Jiangsu Hansoh Pharmaceutical Group Co., Ltd. 2-substituted piperidine derivatives, preparation methods and medicinal uses thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023237012A1 (zh) * 2022-06-07 2023-12-14 正大天晴药业集团股份有限公司 双环取代的芳香羧酸类氘代化合物

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013164802A1 (en) 2012-05-04 2013-11-07 Novartis Ag Complement pathway modulators and uses thereof
WO2013192345A1 (en) 2012-06-20 2013-12-27 Novartis Ag Complement pathway modulators and uses thereof
WO2014143638A1 (en) 2013-03-14 2014-09-18 Novartis Ag 2-(1h-indol-4-ylmethyl)-3h-imidazo[4,5-b]pyridine-6-carbonitrile derivatives as complement factor b inhibitors useful for the treatment of ophthalmic diseases
WO2015009616A1 (en) 2013-07-15 2015-01-22 Novartis Ag Piperidinyl indole derivatives and their use as complement factor b inhibitors
WO2015038939A2 (en) 2013-09-13 2015-03-19 Isis Pharmaceuticals, Inc. Modulators of complement factor b
WO2015066241A1 (en) 2013-10-30 2015-05-07 Novartis Ag 2-benzyl-benzimidazole complement factor b inhibitors and uses thereof
WO2018005552A1 (en) 2016-06-27 2018-01-04 Achillion Pharmaceuticals, Inc. Quinazoline and indole compounds to treat medical disorders
WO2019043609A1 (en) 2017-08-31 2019-03-07 Novartis Ag NEW USES OF PIPERIDINYL-INDOLE DERIVATIVES

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013164802A1 (en) 2012-05-04 2013-11-07 Novartis Ag Complement pathway modulators and uses thereof
WO2013192345A1 (en) 2012-06-20 2013-12-27 Novartis Ag Complement pathway modulators and uses thereof
WO2014143638A1 (en) 2013-03-14 2014-09-18 Novartis Ag 2-(1h-indol-4-ylmethyl)-3h-imidazo[4,5-b]pyridine-6-carbonitrile derivatives as complement factor b inhibitors useful for the treatment of ophthalmic diseases
WO2015009616A1 (en) 2013-07-15 2015-01-22 Novartis Ag Piperidinyl indole derivatives and their use as complement factor b inhibitors
CN105579444A (zh) * 2013-07-15 2016-05-11 诺华股份有限公司 哌啶基吲哚衍生物和它们作为补体因子b抑制剂的用途
WO2015038939A2 (en) 2013-09-13 2015-03-19 Isis Pharmaceuticals, Inc. Modulators of complement factor b
WO2015066241A1 (en) 2013-10-30 2015-05-07 Novartis Ag 2-benzyl-benzimidazole complement factor b inhibitors and uses thereof
WO2018005552A1 (en) 2016-06-27 2018-01-04 Achillion Pharmaceuticals, Inc. Quinazoline and indole compounds to treat medical disorders
CN109414441A (zh) * 2016-06-27 2019-03-01 艾其林医药公司 治疗医学障碍的喹唑啉和吲哚化合物
WO2019043609A1 (en) 2017-08-31 2019-03-07 Novartis Ag NEW USES OF PIPERIDINYL-INDOLE DERIVATIVES
CN111032042A (zh) * 2017-08-31 2020-04-17 诺华股份有限公司 哌啶基-吲哚衍生物的新用途

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"Handbook of Chemistry and Physics", 1994
AM J NEPHROL, vol. 20, no. 2, 2000, pages 122 - 128
EDWARD B. ROCHE: "Bioreversible Carriersin Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATIONAND PERGAMON PRESS
EUR J IMMUNOL, vol. 17, no. 3, 1987, pages 321 - 326
FRONTIERS IN IMMUNOLOGY, vol. 10, 2019, pages 1157
IGA NEPHROPATHY, vol. 95, no. 4, 2019, pages 750 - 756
J CLIN IMMUNOL, vol. 34, no. 2, 2014, pages 224 - 232
J NEPHROL, vol. 26, no. 4, 2013, pages 708 - 715
J. RAUTIOETAL.: "Prodrugs: Designand Clinical Applications", NATURE REVIEW DRUG DISCOVERY, vol. 7, 2008, pages 255 - 270
MAINOLFI NELLO, EHARA TAKERU, KARKI RAJESHRI G., ANDERSON KAREN, MAC SWEENEY AENGUS, LIAO SHA-MEI, ARGIKAR UPENDRA A., JENDZA KEIT: "Discovery of 4-((2 S ,4 S )-4-Ethoxy-1-((5-methoxy-7-methyl-1 H -indol-4-yl)methyl)piperidin-2-yl)benzoic Acid (LNP023), a Factor B Inhibitor Specifically Designed To Be Applicable to Treating a Diverse Array of Complement Mediated Diseases", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 63, no. 11, 11 June 2020 (2020-06-11), US , pages 5697 - 5722, XP055791648, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.9b01870 *
MEDICINE, vol. 76, no. 2, 1997, pages 63 - 93
MICHAEL B. SMITHJERRY MARCH: "March's Advanced Organic Chemistry", 2007, JOHN WILEY & SONS
S. J. HECKERETAL.: "Prodrugs of Phosphate sand Phosphonates", JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, 2008, pages 2328 - 2345
See also references of EP4194449A4
T. HIGUCHIAND V. STELLA: "Pro-drugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14
THOMAS SORRELL: "Organic Chemistry", 1999, UNIVERSITY SCIENCE BOOKS
XUAN YUAN ET AL., HAEMATOLOGICA, vol. 102, 2017, pages 466 - 475

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023020566A1 (zh) 2021-08-18 2023-02-23 四川海思科制药有限公司 一种苯并氮杂芳环衍生物及其在医药上的应用
WO2023072197A1 (en) * 2021-10-27 2023-05-04 Hansoh Bio Llc Piperidinyl indole derivatives, preparation methods and medicinal uses thereof
WO2023246677A1 (zh) * 2022-06-20 2023-12-28 深圳信立泰药业股份有限公司 一种吲哚-苯基哌啶化合物及其制备方法与应用
WO2024051849A1 (en) * 2022-09-10 2024-03-14 Jiangsu Hansoh Pharmaceutical Group Co., Ltd. 2-substituted piperidine derivatives, preparation methods and medicinal uses thereof

Also Published As

Publication number Publication date
EP4194449A1 (en) 2023-06-14
CN114057692A (zh) 2022-02-18
CN114057692B (zh) 2023-07-21
US20230331710A1 (en) 2023-10-19
EP4194449A4 (en) 2024-03-20

Similar Documents

Publication Publication Date Title
WO2022028507A1 (zh) 一种杂环类化合物、其制备方法及用途
WO2022028527A1 (zh) 补体因子b抑制剂及其药物组合物、制备方法和用途
JP5913631B2 (ja) Parp阻害剤としての縮合四環式または縮合五環式ピリドフタラジノン
CN112752756A (zh) 作为PD-L1免疫调节剂的四氢-咪唑并[4,5-c]吡啶衍生物
CN113784963B (zh) 用作ret激酶抑制剂的化合物及其应用
WO2019154257A1 (zh) 一种甾族类衍生物调节剂及其制备方法和应用
CN106536503A (zh) 一种酪氨酸激酶抑制剂及其用途
CN103038233A (zh) 吡啶酮和氮杂吡啶酮化合物及使用方法
WO2022143845A1 (zh) 含氮桥杂环化合物、其制备方法及其在医药上的应用
CN115298165A (zh) 作为HIF-2α抑制剂的四氢化萘和四氢喹啉化合物
TWI452044B (zh) 嗎啉衍生物
CN105263924B (zh) Cxcr7受体调节剂
WO2021093795A1 (zh) 一种rock抑制剂及其制备方法和用途
WO2022247816A1 (zh) 含氮杂环类化合物、其制备方法及其在医药上的应用
WO2020224652A1 (zh) 一类泛素化特异性蛋白酶抑制剂及其制备方法与应用
EP3368023B1 (en) Derivatives of porphyrins, their process of preparation and their use for treating viral infections
TW201408672A (zh) □烷化合物
WO2015021894A1 (zh) 新型羟肟酸衍生物及其医疗应用
WO2024008129A1 (zh) 作为kat6抑制剂的化合物
WO2021104413A1 (zh) 稠合吡啶环衍生物、其制备方法及其在医药上的应用
WO2022007966A1 (zh) Pb2抑制剂及其制备方法和用途
KR20220000854A (ko) 신규한 화합물 및 이를 포함하는 내성암 예방 또는 치료용 약학 조성물
WO2020233645A1 (zh) 大环类衍生物、及其制备方法和用途
CN115427407B (zh) 一种新型n-杂环bet溴结构域抑制剂、其制备方法及医药用途
WO2023274280A1 (zh) 一种联苯类衍生物抑制剂的晶型及其制备方法

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21853576

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021853576

Country of ref document: EP

Effective date: 20230307