CN111072652B - Compounds for the treatment of diabetes and/or related disorders - Google Patents

Compounds for the treatment of diabetes and/or related disorders Download PDF

Info

Publication number
CN111072652B
CN111072652B CN201811226113.8A CN201811226113A CN111072652B CN 111072652 B CN111072652 B CN 111072652B CN 201811226113 A CN201811226113 A CN 201811226113A CN 111072652 B CN111072652 B CN 111072652B
Authority
CN
China
Prior art keywords
optionally substituted
compound
amylin
compounds
amyloid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201811226113.8A
Other languages
Chinese (zh)
Other versions
CN111072652A (en
Inventor
G·J·S·库伯尔
S·张
J·艾特肯
李正球
郑彬彬
丁克
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jinan University
Auckland Uniservices Ltd
Original Assignee
Jinan University
Auckland Uniservices Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jinan University, Auckland Uniservices Ltd filed Critical Jinan University
Priority to CN201811226113.8A priority Critical patent/CN111072652B/en
Priority to PCT/NZ2019/050138 priority patent/WO2020080960A1/en
Priority to US17/286,794 priority patent/US20220041591A1/en
Priority to EP19872857.8A priority patent/EP3867227A4/en
Publication of CN111072652A publication Critical patent/CN111072652A/en
Application granted granted Critical
Publication of CN111072652B publication Critical patent/CN111072652B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates

Abstract

Disclosed herein are compounds for use in the treatment of diabetes and/or related disorders. In particular, methods for preventing, inhibiting and/or reversing amyloid fibril formation of an amylin, preventing beta-cell death of langerhans' islets, preventing and/or reversing conversion of soluble human amylin to insoluble human amylin, preventing and inhibiting and/or reversing cytotoxic amyloid fibril formation are disclosed. The invention also relates to compounds useful in the manufacture of medicaments, for example, for the prevention and/or treatment of amylin-related diseases.

Description

Compounds for the treatment of diabetes and/or related disorders
Technical Field
The present invention relates to compounds useful in and methods of treating or preventing diabetes and/or related conditions, and/or for preventing, inhibiting and/or reversing amyloid fibril formation of amylin (amylin), preventing langerhans' island (islets of Langerhans) (β) -cell death, preventing and/or reversing the conversion of soluble human amylin (human amylin) to insoluble human amylin, preventing and inhibiting and/or reversing cytotoxic amylin fibril formation. The invention also relates to compounds useful in the manufacture of medicaments, for example, for the prevention and/or treatment of amylin-related diseases (such as type II diabetes).
Background
Misfolded protein aggregates, known as amyloid proteins, have been reported to play a critical role in the pathology of a variety of diseases such as rheumatoid arthritis, atherosclerosis, alzheimer's disease, parkinson's disease, huntington's disease, and diabetes.
Misfolded human amylin (hA) is the major component of amylin amyloid in diabetics and is thought to contribute, at least in part, to the development and progression of type II diabetes (type II diabetes mellitus/type II diabetes). Human amylin thus represents a potential target for the development of drugs that can prevent or slow the progression of type II diabetes.
Certain molecules capable of altering hA misfolding and aggregation are known in the art and include broad spectrum antibiotics, tetracyclines. However, many of these molecules suffer from drawbacks, such as side effect profiles or off-target activities, which make them unsuitable for long-term use. There remains a need for compounds capable of preventing, inhibiting and/or reversing amyloid fibril formation of an amylin, and/or preventing beta-cell death of langerhans' islets, and/or preventing and/or reversing the conversion of soluble human amylin to insoluble human amylin, and/or preventing and inhibiting and/or reversing cytotoxic amyloid fibril formation.
It is an object of the present invention to provide compounds which overcome or at least partially ameliorate some of the above disadvantages and/or at least provide the public with a useful choice.
Other objects of the invention will become apparent from the following description, given by way of example only.
In this specification, where reference has been made to external sources of information including patent specifications and other documents, this is generally for the purpose of providing a context for discussing the features of the invention. Unless otherwise specified, references to such sources of information are not to be construed in any jurisdiction as an admission that such sources of information are prior art or form part of the common general knowledge in the art.
Disclosure of Invention
In one aspect, the invention provides a compound of formula I:
Figure BDA0001835577520000021
wherein:
R 1 selected from hydrogen, halo (halide), optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
when present, the or each R 2 Independently selected from hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 3 Selected from halogen radicals,Optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl;
n is selected from 0, 1, 2, 3, 4;
or a pharmaceutically acceptable salt thereof.
In one aspect, the invention provides a compound of formula II:
Figure BDA0001835577520000022
wherein:
R 8 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 9 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 10 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 11 selected from optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;
R 12 selected from optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;
or a pharmaceutically acceptable salt thereof.
In another aspect, the invention relates to a composition comprising a compound of formula I or a compound of formula II together with a carrier, excipient or diluent.
In another aspect, the invention relates to a method of synthesizing a compound of formula I or a compound of formula II as depicted or described herein, e.g., in an example as depicted or described herein.
In another aspect, the invention relates to a process for synthesizing a compound of formula I, the process comprising refluxing a mixture comprising an acetophenone analogue, sodium hydride, and diethyl carbonate to form a second mixture comprising compound S5;
combining a second mixture comprising S5 with an aminopyridine analog and BiCl 2 Blending to form a third mixture, and maintaining the third mixture at about 120 ℃ for greater than about 6 hours to form compound S6:
optionally purifying compound S6, for example by chromatography;
blending S6 or a mixture comprising S6 with anhydrous DCM and BBr3 to form a fourth mixture, and maintaining the fourth mixture until completed to form the compound of formula I.
In another aspect, the invention relates to a method of synthesizing a compound of formula II, the method comprising refluxing a mixture comprising compound S1, sodium hydride, and diethyl carbonate to form a second mixture comprising compound S2;
blending the second mixture comprising S2 with an aminopyridine analog to form a third mixture, and maintaining the third mixture at about 120 ℃ for greater than about 6 hours:
optionally purifying the third mixture, for example by chromatography;
the third mixture is admixed with ethyl acetate and Pd/C to form a fourth mixture, and the fourth mixture is maintained for at least about 1 to 3 hours to form the compound of formula I.
In another aspect, the invention relates to a method of treating or preventing an amylin amyloid-related disease comprising administering to a subject in need thereof an effective amount of a compound of formula I or a compound of formula II or a pharmaceutically acceptable salt thereof.
In another aspect, the invention relates to a method of inhibiting, preventing or reversing an amyloidosis or formation of amyloid fibrils or amyloid plaques in a subject in need thereof, comprising administering to the subject in need thereof an effective amount of a compound of formula I or a compound of formula II or a pharmaceutically acceptable salt thereof.
In another aspect, the invention relates to a method of inhibiting, preventing or reversing the formation of an amylin amyloidosis disorder or one or more amylin amyloid fibrils or amylin amyloid plaques, the method comprising contacting the amylin amyloidosis disorder or one or more amylin amyloid fibrils or amylin plaques with an effective amount of formula I or formula II or a pharmaceutically acceptable salt thereof.
In one embodiment, the method is a method of inhibiting, preventing or reversing the formation of amylin amyloid fibrils.
In another aspect, the invention relates to a method of treating or preventing diabetes in a subject in need thereof, the method comprising administering to the subject in need thereof an effective amount of formula I or formula II or a pharmaceutically acceptable salt thereof.
In another aspect, the present invention relates to a method of treating or preventing langerhans' islet β -cell death in a subject in need thereof, the method comprising administering to a subject in need thereof an effective amount of a compound of formula I or a compound of formula II or a pharmaceutically acceptable salt thereof.
In another aspect, the invention relates to a method of treating or preventing langerhans 'islet B-cell death, the method comprising contacting one or more langerhans' islet β -cells with an effective amount of a compound of formula I or a compound of formula II, or a pharmaceutically acceptable salt thereof.
In another aspect, the invention provides a pharmaceutical composition comprising one or more compounds of formula I or formula II, optionally together with a pharmaceutically acceptable carrier or excipient.
The invention further relates to a compound of formula I or formula II for use in, e.g., treating or preventing an amylin amyloid-related disease in a subject in need thereof; inhibiting, preventing or reversing an amyloidosis or formation of amyloid fibrils or amyloid plaques; inhibiting, preventing or reversing the formation of islet amyloid fibrils; treating or preventing diabetes; or to treat or prevent one or more of langerhans' islet β -cell death.
The invention further relates to the use of a compound of formula I or a compound of formula II for the preparation of a medicament for the treatment or prevention of an amyloid-related disease, e.g., in a subject in need thereof; inhibiting, preventing or reversing an amyloidosis or formation of amyloid fibrils or amyloid plaques; inhibiting, preventing or reversing the formation of islet amyloid fibrils; treating or preventing diabetes, or treating or preventing one or more of langerhans' islet β -cell death.
Embodiments set forth herein may relate to any of the above aspects.
Other aspects of the invention, not limited to or by the information in this summary, may be apparent from the following description, which is given by way of example only, and with reference to the accompanying drawings.
Broadly speaking, the invention may also consist in the parts, elements and features referred to or indicated in the specification of the application, individually or collectively, and any or all combinations of any two or more of said parts, elements or features, and where specific integers (integers) having known equivalents in the art to which the invention relates are herein referred to, such known equivalents are deemed to be incorporated herein as if individually set forth.
Drawings
The invention will now be described, by way of example only, with reference to the accompanying drawings, in which:
figure 1 is three graphs showing the results of a thioflavin-T assay that investigated the ability of compounds described herein to inhibit amylin fibril formation, as discussed in example 2 herein. FIG. 1A shows the result of the hA to compound molar ratio of 1:1, FIG. 1B shows the result of the hA to compound molar ratio of 1:0.1, and FIG. 1C shows the result of the hA to compound molar ratio of 1:0.01.
Fig. 2 is a graph showing the results of a cell death assay that studies the ability of compounds described herein to inhibit amylin-induced cell death, as discussed in example 3 herein.
Figure 3 is a graph showing the results of a thioflavin-T assay that studies the ability of compounds described herein to inhibit amylin fibril formation, as discussed in example 4 herein.
Fig. 4 is three graphs showing the results of a thioflavin-T assay, as discussed in example 4 herein, that investigated the ability of compounds described herein to inhibit amylin fibril formation. FIG. 4A shows the result of the hA to compound molar ratio of 1:1, FIG. 4B shows the result of the hA to compound molar ratio of 1:0.1, and FIG. 4C shows the result of the hA to compound molar ratio of 1:0.01.
Fig. 5 is two graphs showing the results of a cell death assay, as discussed in example 5 herein, that studies the ability of compounds described herein to inhibit amylin-induced cell death. FIG. 5A shows the data observed in CM cells, while FIG. 5B shows the data observed in RINm5F cells.
Fig. 6 is two graphs showing the results of a cell death assay, as discussed in example 6 herein, that studies the ability of compounds described herein to inhibit amylin-induced cell death. FIG. 6A shows the data observed in CM cells, while FIG. 6B shows the data observed in RINm5F cells.
Detailed Description
The present invention provides various compounds useful in the treatment of amyloidosis including, for example, diabetes. The compounds useful herein have advantageous physicochemical and/or therapeutic properties that make them particularly useful in the treatment of amylin-related diseases, such as diabetes.
In a preferred embodiment, the compound of formula I is a compound of formula Ia:
Figure BDA0001835577520000051
wherein the method comprises the steps of
R 1 Selected from: hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
when present, the or each R 2 Independently selected from hydroxy, halo, optionally substituted amino, optionally taken Substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
when present, the or each R 4 Independently selected from hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
n is selected from 0, 1, 2, 3, 4;
m is selected from 0, 1, 2, 3, 4, 5;
or a pharmaceutically acceptable salt thereof.
In a preferred embodiment, the compound of formula I is a compound of formula Ib:
Figure BDA0001835577520000061
Wherein:
R 1 selected from: hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
when present, the or each R 2 Independently selected from hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted haloalkoxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroarylOptionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 5 selected from the group consisting of hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 6 Selected from the group consisting of hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
n is selected from 0, 1, 2, 3, 4
Or a pharmaceutically acceptable salt thereof.
In a preferred embodiment, the compound of formula I is a compound of formula Ic:
Figure BDA0001835577520000071
wherein the method comprises the steps of
R 1 Selected from: hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
when present, the or each R 2 Independently selected from hydroxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkenyl, optionally substituted alkyloxy, optionallyOptionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 6 Selected from the group consisting of hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
n is selected from 0, 1, 2, 3, 4;
or a pharmaceutically acceptable salt thereof.
In a preferred embodiment, the compound of formula I is a compound of formula Id:
Figure BDA0001835577520000072
wherein the method comprises the steps of
R 1 Selected from: hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 2 selected from the group consisting of hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 6 Selected from hydroxy, halo, optionally substitutedAmino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
or a pharmaceutically acceptable salt thereof.
In any of the compounds of formula I, formula Ia, formula Ib, formula Ic, or formula Id:
R 1 preferably hydrogen;
preferably when present, the or each R 2 Independently selected from hydroxy and alkyloxy, such as methyloxy (also known as methoxy);
R 3 preferably selected from optionally substituted aryl;
preferably when present, the or each R 4 Independently selected from hydroxy and alkyloxy, such as methyloxy (also known as methoxy);
R 5 preferably selected from hydroxy and alkyloxy groups, such as methyloxy (also known as methoxy);
R 6 preferably selected from hydroxy and alkyloxy groups, such as methyloxy (also known as methoxy);
n is preferably 1; and/or
m is preferably 1 or 2.
Preferred compounds of formula I are selected from Zbbp39, zbb03-41-2, zbb03-40-2, zbbp144 (described herein) or pharmaceutically acceptable salts thereof.
In a preferred embodiment, the compound of formula II is a compound of formula IIa:
Figure BDA0001835577520000081
wherein:
R 8 selected from hydrogen, optionally substitutedC 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 9 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 10 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
when present, the or each R 13 Independently selected from hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
when present, the or each R 14 Independently selected from
When present, A is selected from
Figure BDA0001835577520000091
Wherein the method comprises the steps of
When present, the or each R 19 Independently selected from hydroxy, halo, carboxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
u is selected from 0, 1, 2, 3, 4;
x is selected from C or N;
y is selected from C or N;
z is selected from C or N;
p is selected from 0, 1, 2, 3, 4, 5;
q is selected from 0, 1, 2, 3, 4;
r is selected from 0 or 1;
or a pharmaceutically acceptable salt thereof.
In a preferred embodiment, the compound of formula II is a compound of formula IIb:
Figure BDA0001835577520000092
wherein:
R 8 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 9 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 10 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
when present, the or each R 13 Independently selected from hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
when present, the or each R 14 Independently selected from hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxyl, optionally substituted cycloalkylOptionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
When present, A is selected from
Figure BDA0001835577520000101
Wherein the method comprises the steps of
When present, the or each R 19 Independently selected from hydroxy, halo, carboxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
u is selected from 0, 1, 2, 3, 4;
p is selected from 0, 1, 2, 3, 4, 5;
q is selected from 0, 1, 2, 3, 4;
r is selected from 0 or 1;
or a pharmaceutically acceptable salt thereof.
In a preferred embodiment, the compound of formula II is a compound of formula IIc:
Figure BDA0001835577520000102
wherein:
R 8 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 9 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 10 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
when present, the or each R 13 Independently selected from hydroxy, halo, and optionally Optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 15 selected from the group consisting of hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 16 selected from the group consisting of hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
When present, A is selected from
Figure BDA0001835577520000111
Wherein the method comprises the steps of
When present, the or each R 19 Independently selected from hydroxy, halo, carboxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted haloalkoxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenylSubstituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
u is selected from 0, 1, 2, 3, 4;
p is selected from 0, 1, 2, 3, 4, 5;
r is selected from 0 or 1;
or a pharmaceutically acceptable salt thereof.
In a preferred embodiment, the compound of formula II is a compound of formula IId:
Figure BDA0001835577520000121
wherein:
R 8 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 9 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 10 selected from hydrogen, optionally substituted C 1-4 Alkyl, optionally substituted C 2-4 Alkenyl groups;
R 15 selected from the group consisting of hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 16 Selected from hydroxy, halo, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl,Optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxyl, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 17 selected from the group consisting of hydroxy, halo, carboxy, optionally substituted amino, optionally substituted aminoalkyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
R 18 selected from the group consisting of hydroxy, halo, carboxy, optionally substituted amino, optionally substituted aminoalkyl, amino alkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkyl, optionally substituted carboxyalkenyl;
When present, A is selected from
Figure BDA0001835577520000131
Wherein the method comprises the steps of
When present, the or each R 19 Independently selected from hydroxy, carboxy, optionally substituted amino, optionally substituted aminoalkyl, amino alkenyl, optionally substituted alkyloxy, optionally substituted alkenyloxy, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, optionally substituted cycloalkyl, carboxy, optionally substituted carboxyalkylA group, optionally substituted carboxyalkenyl;
u is selected from 0, 1, 2, 3, 4;
r is selected from 0 or 1;
or a pharmaceutically acceptable salt thereof.
In any of the compounds of formula II, formula IIa, formula IIb, formula IIc, or formula IId:
R 8 preferably hydrogen;
R 9 preferably hydrogen;
R 10 preferably hydrogen;
R 11 preferably an optionally substituted aryl group, such as an optionally substituted phenyl (known as biphenyl);
when present, R 12 Preferably an optionally substituted heteroaryl, such as an optionally substituted pyridinyl;
when present, R 13 Preferably carboxyl, alkyloxy (such as methoxy), or hydroxy or halo;
When present, R 14 Preferably an alkyloxy (e.g., methoxy) or halo group;
a is preferably benzene-1, 4-yl;
u is preferably 0;
preferably, X is N and Y and Z are C;
R 15 preferably an alkyloxy (e.g., methoxy) or halo group;
R 16 preferably an alkyloxy (e.g., methoxy) or halo group;
R 17 preferably carboxyl, alkyloxy (such as methoxy), or hydroxy or halo; and/or
R 18 Preferably a carboxyl group, an alkyloxy group (such as methoxy), or a hydroxyl group or a halogen group.
Preferred compounds of formula II are selected from Zbbp92, zbb03-41-1, zbb03-44-2, zbb03-44-1, zbb03-32 (described herein) or pharmaceutically acceptable salts thereof.
As used herein, the term "and/or" means "and" or both.
As used herein, "s" following a noun means the plural and/or singular forms of the noun.
It is intended that references (e.g., 1 to 10) to the number of ranges disclosed herein also include all reasonable numbers of references (e.g., 1, 1.1, 2, 3, 3.9, 4, 5, 6, 6.5, 7, 8, 9, and 10) within the ranges as well as any reasonable numbers of any ranges (e.g., 2 to 8, 1.5 to 5.5, and 3.1 to 4.7) within the ranges, and therefore all subranges of the ranges disclosed herein are explicitly disclosed herein. These are only examples of what is specifically intended, and all possible combinations of numerical values between the lowest value and the highest value enumerated are to be considered to be expressly stated in this application in a similar manner.
The term "comprising" as used in this specification means "consisting at least in part of … …". When interpreting statements in this specification which include said terms, the features recited in each statement or claim by said terms need to be present, but other features can also be present. Related terms such as "comprising" and "comprises" should be interpreted in the same manner.
Human amylin (hA)
The present invention relates, in certain embodiments, to compounds capable of preventing or reversing misfolding, aggregation, and/or amyloid plaque formation in human amylin (hA). In certain embodiments, the present invention relates to compounds capable of preventing and/or reversing the conversion from soluble human amylin to insoluble human amylin, and/or preventing and inhibiting and/or reversing the formation of cytotoxic amylin fibrils
Human amylin, also known as islet amylin amyloid polypeptide (islet amylin amyloid polypeptide, IAPP), is a 37 residue peptide hormone produced in and secreted by beta cells of langerhans' islets in the human pancreas. The production of hA in humans involves the conversion of a 89 residue coding sequence to a 67 amino acid pre-amylin sequence (proamylin sequence, proIAPP). The present pre-amylin sequence is then subjected to post-translational modification to produce hA. Human amylin is produced in the human body under normal conditions and plays a role in glycemic control along with insulin.
Amyloidosis of amylin
The term "amyloidosis" as used herein refers to the deposition of amyloid proteins in humans. It will be apparent to those skilled in the art that there is generally a relationship between protein identity and where amyloidosis appears in the human body. Thus, several types of amyloid are present, each of which is classified according to the proteins it contains, and a particular type of amyloidosis is classified based on the site of amyloid accumulation.
The deposition of amyloid in humans typically involves the accumulation of insoluble amyloid fibrils comprising misfolded proteins. The terms "amyloid", "amyloid fibrils" and "amyloid plaques" as used herein refer to protein aggregates produced by misfolding of a protein into a form that aids and/or promotes aggregation, and/or may result in cytotoxicity.
Human amylin is a protein that may misfold resulting in the conversion of soluble amylin monomers and soluble amylin oligomers to insoluble amylin amyloid fibrils. The terms "amyloid" or "amyloid fibrils" or "amyloid aggregates" or "amyloid plaques" are used herein to refer to an amyloid protein comprising human amylin as a protein component in an insoluble state. The terms "islet amyloid protein", "islet amyloid fibril", "islet aggregate" or "islet amyloid plaque" are used herein to refer to an amyloid protein comprising human amylin as a protein component in an insoluble state and typically found in Yu Langge h islets. While present in islets, these insoluble aggregates can lead to β -cell death and other cytotoxic effects, the mechanisms of which are poorly understood. Without wishing to be bound by theory, the inventors believe that the aggregation of the islet amyloid to the islet amyloid fibrils and/or hA misfolding at least in part results in toxicity observed in the pathology of the amyloid-related disease (e.g., type II diabetes) and believed to play a role in the pathology of the amyloid-related disease.
Interference of amylin amyloid
The inventors have found that the compounds described herein are capable of interfering with amylin amyloid formation, such as islet amylin amyloid formation.
The term "interference" as used herein refers to both a priori and posterior interference of amyloidosis of amylin. A priori interference refers to the interference of compounds described herein with any portion of the process that involves the conversion of soluble monomers and oligomers (e.g., amylin monomers and oligomers) to insoluble amylin amyloid. As used herein, a posterior interference refers to the deaggregation of insoluble amylin amyloid proteins (e.g., islet amylin amyloid proteins) that have formed, which may include resolubilization of constituent proteins.
Interference of insoluble amylin amyloid can occur directly or indirectly. Direct interference involves one or more of the compounds described herein or their metabolites that bind to a monomeric or oligomeric portion of a protein, such as a human amylin monomer or oligomer, or a portion of a preformed aggregate, and physically prevents further aggregation and/or reverses aggregation, such as by attenuating interactions between monomers and/or oligomers in the aggregate. In contrast, indirect interference involves preventing fibril or aggregate formation by mechanisms that do not require direct contact between the compound and the protein, e.g., by altering local conditions that favor soluble forms of the protein over insoluble forms, or by favoring the non-aggregated conformation of the monomer or oligomer.
The term "amylin monomer" as used herein refers to a 37 residue peptide hormone produced in and secreted by the beta cells of the islets of langerhans. The amino acid sequence for human preproteins, also known as islet amyloid polypeptide preproteins, is present in RefSeq np_000406.1 and UniProtKB/SwissProt A0a024RAU1.
The term "oligomer" as used herein refers to a molecule having two or more monomer units. The amylin oligomer may comprise from about 2, 3, 4, 5, 6, 7, 8, 10, 15, 16, 20, or 25 monomer units to about 100 monomer units, or from about 100 to about 1000, 2000, 3000, or more monomer units. The term "amylin oligomer" refers to an oligomer in which the monomeric units are human amylin.
The term "interference" as used herein in the context of protein aggregation refers to interference by the methods of aggregation of the compounds described herein such that aggregation is slowed or prevented.
In some embodiments, direct interference of aggregates (e.g., islet aggregates) by a compound described herein involves binding by covalent interactions, and in other embodiments involves binding by non-covalent interactions. In various embodiments, the compounds described herein interact with monomers or oligomers of proteins comprising amyloid, or with amyloid itself, through a combination of one or more covalent interactions and/or one or more non-covalent interactions, such as van der waals interactions (van der Waals interaction).
For example, without wishing to be bound by theory, the inventors believe that in various embodiments the compounds described herein are covalently bound to a monomeric form of a protein (e.g., hA). Again without wishing to be bound by any theory, it is believed that this stabilizes the conformation of the monomeric and/or soluble oligomers, non-toxic forms, directly inhibiting amyloid formation.
For example, if a compound does not bind directly to a monomer or oligomer of a protein that constitutes an amyloid protein or to the amyloid protein itself, but affects another portion of the amyloid amyloidosis process that prevents or inhibits the formation of other aggregates or causes deagglomeration of preformed aggregates, then interference of the amyloid protein may also occur indirectly.
Without wishing to be bound by theory, the inventors believe that one mechanism for forming insoluble islet amyloid protein begins with aggregation of pre-amylin, which then serves as a starting site for deposition of insoluble islet amyloid fibrils. In some embodiments, the interference of the amylin amyloid protein thus occurs by directly or indirectly interfering with the aggregation of pre-amylin or thereon.
In the case of islet amyloid, experiments report that hA can be reasonably unstructured under conventional conditions and is typically composed of random coil structures. Under certain conditions normally associated with pathogenesis, it is desirable that hA misfolded to produce Beta sheets stacked to form a highly structured aggregate comprising good few layers of Beta sheets (Beta-sheet). In some embodiments, the interference with islet amylin amyloid comprises prevention or reversal of hA misfolding. Misfolding of the protein component of amylin is also believed to play a role in other diseases associated with amylin amyloidosis.
In various embodiments, the compounds described herein interfere with amyloid aggregates by one or more mechanisms selected from the group consisting of: preventing, inhibiting and/or reversing amyloid fibril formation of an amylin, preventing and/or reversing conversion of soluble human amylin to insoluble human amylin, and preventing, inhibiting and/or reversing cytotoxic amyloid fibril formation. In some embodiments, the compounds described herein interfere with amyloid fibril formation by preventing, inhibiting, and/or reversing protein misfolding. For example, the compounds described herein interfere with islet amyloid fibril formation by preventing, reversing, and/or reversing hA misfolding.
In various embodiments, the compounds described herein prevent, inhibit, and/or reverse protein misfolding and/or prevent, inhibit, and/or reverse protein aggregation.
The term "aggregate" or "aggregate" as used herein refers to the accumulation of proteins (such as human amylin or its precursor proIAPP), particularly in insoluble form. Without wishing to be bound by theory, the inventors believe that misfolding of the amylin increases the likelihood of hA aggregating into structured β sheets.
Effects on disease states
Amylin is thought to play a role in the pathogenesis of a variety of diseases. The term "amylin amyloid-related disease" as used herein refers to diseases including, but not limited to, diabetes mellitus, including type II diabetes (T2D), metabolic syndrome, syndrome X, dysregulation of blood glucose, insulin resistance, and the like.
Amylin-related diseases exist in a variety of animals (including mammals, e.g., humans).
The inventors have found that the compounds described herein are capable of preventing, inhibiting and/or reversing, for example, amyloid fibril formation, langerhans' islet β -cell death, conversion of soluble human amylin to insoluble human amylin, and cytotoxic oligomer formation. Without wishing to be bound by theory, the inventors believe that these methods are associated with the development and/or progression of one or more amylin-related diseases (e.g., type II diabetes).
The term "preventing" as used herein refers to stopping the progression of, for example, unoccupied amyloid fibril formation, langerhans' islet β -cell death, conversion of soluble human amylin to insoluble human amylin, and formation of cytotoxic oligomers. In certain embodiments, such prevention continues for a certain period of time-e.g., as long as the concentration of one or more compounds as described herein (e.g., local concentration of hA) remains above a certain threshold. It should be appreciated that in such embodiments, the term "preventing" does not encompass permanent prevention.
The term "inhibit" as used herein is used in a similar manner to "prevent," but refers to stopping the progression of, for example, already initiated amyloid fibril formation, langerhan's islet β -cell death, conversion of soluble human amylin to insoluble human amylin, and formation of cytotoxic oligomers.
Prevention, inhibition, or reversal of progression associated with the development of one or more amylin-related diseases may manifest in a variety of ways. For example, in some embodiments, preventing, inhibiting, or reversing the progression of processes such as amyloid fibril formation of amylin, langerhans' islet β -cell death, conversion of soluble human amylin to insoluble human amylin, and formation of cytotoxic oligomers results in slowing disease progression and improving the quality of life of individuals with disease.
In various embodiments, prevention, inhibition, or reversal of progression associated with the development of one or more amylin-related diseases appears to be increased survival. For example, prevention, inhibition, or reversal of the progression associated with hA amylin amyloid formation will in certain embodiments manifest as, for example, increased survival of islet β -cells.
The term "treating" as used herein refers to inhibiting or arresting the development of an amylin-related disease and/or reducing, ameliorating or regressing an amylin-related disease or one or more side effects thereof. Methods of assessing treatment, including methods of assessing inhibition, suppression, reduction, remission, and/or regression of a disease state, are known and will be apparent to those of skill in the art.
The term "reverse" as used herein refers to the return of an amyloid-related disease to a previous state or a state of lower progression. The terms "alleviating" and "fading" are interpreted in a similar manner.
Administration and formulation
The term "administering" as used herein refers to providing a therapeutically effective amount of a compound to an individual using one or more methods of administering a compound known in the art. These methods include the use of oral, sublingual, intravenous, subcutaneous, transdermal, intramuscular, intradermal, intrathecal, epidural, intraocular, intracranial, inhalation, rectal, vaginal and the like administration of the compounds.
In exemplary embodiments, one or more active agents may be administered orally.
It will be apparent to those skilled in the art that the formulation of the compounds described herein will depend on the method of administration. For example, in certain embodiments, the compounds described herein are formulated as creams, lotions, tablets, capsules, pills, dispersible powders, granules, suppositories, syrups, elixirs, troches, injectable solutions, sterile aqueous or non-aqueous solutions, suspensions or emulsions, patches, and the like.
For example, in various embodiments, the compounds described herein are formulated as tablets, capsules, pills, dispersible powders, granules, syrups, suspensions, or emulsions.
In exemplary embodiments, the compounds described herein are formulated in a solid dosage form, such as a tablet, capsule, or pill.
Formulations suitable for particular methods of administration will be apparent to those skilled in the art.
As will be readily appreciated by those skilled in the art, the route of administration and the nature of the pharmaceutically acceptable carrier will depend on the nature of the disorder and the mammal to be treated. It is believed that the choice of the particular carrier or delivery system and route of administration can be readily determined by one skilled in the art. In preparing any formulation containing a compound active agent, care should be taken to ensure that the activity of the compound is not destroyed during the process and that the compound is able to reach its site of action without being destroyed. In some cases, it may be necessary to protect the compound by means known in the art (e.g., microcapsules). Similarly, the route of administration is chosen such that the compound reaches its site of action.
The person skilled in the art can easily determine the appropriate formulation for the compounds of the invention using conventional methods. Determination of the preferred pH range and suitable excipients (e.g., antioxidants) is conventional in the art. Buffer systems are conventionally used to provide a desired range of pH values and include carboxylic acid buffers such as acetate, citrate, lactate and succinate. Various antioxidants are available for use in such formulations, including: phenolic compounds such as BHT or vitamin E; reducing agents, such as methionine (methionine) or sulfite; and metal chelators such as EDTA.
The compounds as described above, or pharmaceutically acceptable salts thereof, may be prepared in parenteral dosage forms, including dosage forms suitable for intravenous, intrathecal and intracerebral or epidural delivery. Pharmaceutical forms suitable for injectable use include sterile injectable solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions. It should be stable under the conditions of manufacture and storage and protect it from reduction or oxidation and contamination by microorganisms such as bacteria and fungi.
The solvent or dispersion medium for the injectable solution or dispersion may contain any of the conventional solvents or carrier systems for the compound active agent and may contain, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycols, and the like), suitable mixtures thereof, and vegetable oils. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. Prevention of the action of microorganisms can be achieved by including various antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like), if necessary. In many cases, it will be preferable to include an agent that regulates osmotic pressure, such as sugar or sodium chloride. Preferably, the formulation for injection will be isotonic with blood. Long-term absorption of injectable compositions can be achieved by the use of delayed absorbers in the composition, such as aluminum monostearate and gelatin. Pharmaceutical forms suitable for injectable use may be delivered by any suitable route, including intravenous, intramuscular, intracerebral, intrathecal, epidural injection or infusion.
The sterile injectable solution is prepared by the following procedure: the desired amount of active compound is optionally incorporated into the appropriate solvent along with various other ingredients of those listed above, followed by filter sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains an alkaline dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-dried or lyophilized from a pre-sterile-filtered solution of the active ingredient plus any additional desired ingredient.
Other pharmaceutical forms include oral and enteral formulations according to the invention, in which the active compound(s) may be formulated with inert diluents or with assimilable edible carriers, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compounds may be combined with excipients and used in the form of ingestible tablets, buccal or sublingual tablets, dragees, capsules, elixirs, suspensions, syrups, wafers, and the like. The amount of active compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
Tablets, dragees, pills, capsules and the like may also contain the components as set forth below: binders, such as gums, acacia, corn starch or gelatin; excipients, such as dicalcium phosphate; disintegrants such as corn starch, potato starch, alginic acid and the like; lubricants, such as magnesium stearate; and sweeteners such as sucrose, lactose or saccharin which may be added; or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring. Where the unit dosage form (dosage unit) is a capsule, it may contain a liquid carrier in addition to materials of the type described above. Various other materials may be present in the form of coatings or used to otherwise adjust the physical form of the dosage unit. For example, a tablet, pill, or capsule may be coated with shellac, sugar, or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically pure and substantially non-toxic in terms of amount used. In addition, the active compounds can be incorporated into sustained release formulations and preparations, including those that provide for the specific delivery of the active compound to specific areas of the intestinal tract.
Liquid formulations may also be administered enterally through gastric or esophageal tubes.
Enteral formulations may be prepared in the form of suppositories by mixing with suitable bases, such as emulsifying bases or water-soluble bases. Topical, intranasal, intravaginal, intraocular, etc. administration of the compounds of the invention is also possible, but not necessary.
The invention also extends to any other form suitable for administration, for example topical applications, such as creams, lotions and gels, or compositions suitable for inhalation or intranasal delivery, such as solutions, dry powders, suspensions or emulsions.
The compounds of the present invention may be administered by inhalation from a pressurized dispenser or container containing a propellant such as carbon dioxide gas, dichlorodifluoromethane, nitrogen, propane or other suitable gas or combination of gases in the form of a mist spray. The compounds may also be applied using a nebulizer.
Pharmaceutically acceptable vehicles and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic agents, absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional medium or agent is incompatible with the active ingredient, use thereof in therapeutic compositions is contemplated. Supplementary active ingredients may also be incorporated into the composition.
It is particularly advantageous to formulate the composition in unit dosage form for ease of administration and uniformity of dosage. A unit dosage form as used herein refers to physically discrete units suitable as unitary dosages for the mammalian subject to be treated; each unit contains a predetermined amount of active material calculated to produce the desired therapeutic effect in combination with the desired pharmaceutically acceptable vehicle. The present specification for the novel unit dosage forms of the invention is defined by and directly depends on the following: (a) Unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) limitations inherent in the art of compounding active materials for the treatment of diseases in living individuals suffering from pathological conditions, wherein physical health is impaired as disclosed in detail herein.
As mentioned above, for convenient and effective administration, the primary active ingredient may be admixed in a therapeutically effective amount with a suitable pharmaceutically acceptable vehicle in unit dosage form. Unit dosage forms (unit dose) may contain the primary active compound, for example, in an amount ranging from 0.25 μg to about 2000 mg. Expressed in proportions, the active compound may be present in a carrier of about 0.25 μg to about 2000 mg/mL. Where the composition contains a supplemental active ingredient, the dosage is determined with reference to the usual dosages and manner of administration of the ingredient.
The term "therapeutically effective amount" as used herein refers to a dose of a compound sufficient to provide a concentration high enough to affect the desired result. For example, in certain embodiments, a therapeutically effective amount is a dose of a compound described herein sufficient to result in one or more of the following: preventing, inhibiting or reversing amyloid fibril formation of amylin, beta-cell death of langerhans' islets, conversion of soluble human amylin to insoluble human amylin, and cytotoxic oligomer formation.
A therapeutically effective amount of a compound will in certain embodiments be affected by, and may be adjusted based on, a variety of factors. For example, a therapeutically effective dose may be affected by the synergy between the weight, metabolic capacity, and active agent administration combination of the individual. The dosage that can be administered to an individual can also be affected by other factors, such as interactions with other drugs taken by the individual and the severity/ability to tolerate the side effects of any administered compound.
In some embodiments, the desired outcome achieved by the therapeutically effective amount comprises slowing disease progression, improving the quality of life of the individual, and/or improving survival.
In various embodiments, the desired result achieved by the therapeutically effective amount is an improvement in one or more symptoms associated with an amyloid-related disease, such as a reduction in loss of kidney function, heart failure, obstructive sleep apnea, dysphagia, or synovitis.
In various embodiments, the desired result achieved by the therapeutically effective amount is an improvement in one or more symptoms associated with type II diabetes, such as weight loss, polyurea, polydipsia, polyphagia, blurred vision, headache, fatigue, or diabetic skin disease (diabetic dermadrome), and signs such as reduced blood glucose levels and HbA 1C.
Where the compound comprises one or more functional groups that are protonatable or deprotonated (e.g., at physiological pH), the compound may be prepared and/or isolated as a pharmaceutically acceptable salt. It will be appreciated that the compound may be zwitterionic at a given pH. As used herein, the expression "pharmaceutically acceptable salt" refers to a salt of a given compound, wherein the salt is suitable for administration as a medicament. For example, such salts may be formed by reacting an acid or base with an amine or carboxylic acid group, respectively. Acid/base addition salts tend to be more soluble in aqueous solvents than the corresponding free acid/base forms.
In some embodiments, one or more pharmaceutically acceptable salts of the compounds described herein are administered to a subject in a therapeutically effective amount. Such salts may be prepared by methods known in the art involving reacting the compound with a suitable organic or inorganic acid or base. Representative organic salts include methane sulfonate, acetate, oxalate, adipate, alginate, aspartate, valerate, oleate, laurate, borate, benzoate, lactate, phosphate, tosylate (tosylate/tosylate), citrate, malate, maleate, fumarate (fumarate), succinate, tartrate, naphthalene sulfonate, methane sulfonate, 2-naphthalene sulfonate, nicotinate, benzene sulfonate, butyrate, camphorate, camphorsulfonate, cyclopentane propionate, digluconate, dodecyl sulfate, glucoheptonate, glycerophosphate, heptanoate, hexanoate, undecanoate, 2-hydroxyethane sulfonate, ethane sulfonate, and the like. Representative inorganic salts may be formed from inorganic acids such as sulfate, bisulfate, hemisulfate, hydrochloride, chlorate, perchlorate, hydrobromide, hydroiodide, and the like. Examples of the alkali salt include ammonium salts; alkali metal salts such as sodium salt, potassium salt, etc.; alkaline earth metal salts such as calcium salts, magnesium salts, and the like; salts with organic bases such as dicyclohexylamine salts, N-methyl-D-glucamine, phenethylamine, and the like; and salts with amino acids (e.g., arginine, lysine), and the like. Such salts can be readily prepared using methods well known in the art.
In some embodiments, one or more compounds described herein are administered to an individual as prodrugs. Prodrugs are well known in the art and are compounds that are administered in a subsequently metabolised pharmaceutically active pharmaceutical form. The term "prodrug" is used in its broadest sense and encompasses those derivatives which are converted in vivo to the compounds of the present invention. Such derivatives will readily occur to those skilled in the art and include, for example, compounds in which the free hydroxyl groups are converted to ester derivatives or the ring nitrogen atoms are converted to N-oxides. Examples of the ester derivatives include alkyl esters (e.g., acetate, lactate, and glutamine), phosphate esters, and those formed from amino acids (e.g., valine). Any compound that is a prodrug of the compounds of the present invention is within the scope and spirit of the present invention. Conventional procedures for preparing suitable prodrugs according to the present invention are described in textbooks, e.g. "prodrug design (Design of Prodrugs)" ed.h. bundegaad, elsevier, 1985-incorporated herein by reference in its entirety.
The compounds of the invention may be in crystalline form or in a solvate (e.g., hydrate), and both forms are intended to be within the scope of the invention. The term "solvate" is a complex of variable stoichiometry formed by a solute (in the present case, a compound of the present invention) and a solvent. Such solvents should not interfere with the biological activity of the solute. By way of example, the solvent may be water, ethanol, or acetic acid. The fusion method is generally known in the art.
Unless otherwise specified, structures depicted herein are also intended to include all stereochemical forms of the structures; i.e., R and S configurations for each asymmetric center. Thus, single stereochemical isomers, as well as mixtures of enantiomers and diastereomers, of the compounds of the invention are within the scope of the invention. Thus, the present invention encompasses each diastereomer or enantiomer substantially free of the other isomer (by mole, > 90%, and preferably > 95%, free of the other stereoisomer) as well as mixtures of such isomers.
The particular optical isomer may be obtained by resolution of the racemic mixture according to conventional methods, for example, by formation of diastereomeric salts, by treatment with optically active acids or bases. Examples of suitable acids are tartaric acid, diacetyl tartaric acid, dibenzoyl tartaric acid, ditoluoyl tartaric acid and camphorsulfonic acid, and the separation of mixtures of diastereomers from these salts followed by dissociation (separation) of the optically active base after crystallization. The different methods of separating optical isomers involve the use of chiral chromatographic columns that are optimally selected to maximize the separation of enantiomers. Still another method involves synthesizing covalent diastereomeric molecules by reacting a compound of the invention with an optically pure acid or an optically pure isocyanate in an activated form. The synthetic diastereomers may be separated by conventional means, such as chromatography, distillation, crystallization or sublimation, and subsequently hydrolyzed to deliver the enantiomerically pure compounds.
The optically active compounds of the present invention can be obtained by using an active starting material. These isomers may be in the form of free acids, free bases, esters or salts.
Where purification of the compounds of the invention is desired, chromatographic techniques such as high-performance liquid chromatography (HPLC) and reverse phase HPLC can be used. The compounds may be characterized by mass spectrometry and/or other suitable methods.
The invention also provides a pharmaceutical composition comprising a therapeutically effective amount of a compound as defined above, or a pharmaceutically acceptable salt thereof, together with at least one pharmaceutically acceptable carrier or diluent.
The term "composition" is intended to include a formulation of an active ingredient with an encapsulating material as a carrier to obtain a capsule, wherein the active ingredient (with or without other carriers) is surrounded by the carrier.
The compounds described herein are administered alone in certain embodiments, and in other embodiments in combination with other compounds described herein or with other therapeutic agents, or both. The combination may allow for the administration of the compounds as described above with other active ingredients alone, sequentially or simultaneously. The combination may be provided in the form of a pharmaceutical composition.
For example, in some embodiments, two or more compounds described herein are administered to an individual together. When a combination of compounds is administered to an individual, the dose of each individual compound may be less than the therapeutically effective amount such that the combined dose of two or more compounds is equal to or greater than the therapeutically effective amount.
Furthermore, in some embodiments, one or more compounds described herein are administered to an individual along with one or more additional agents. The additional agent may be, for example, an agent that treats, inhibits, and/or reverses amyloid fibril formation, langerhans' islet β -cell death, conversion of soluble human amylin to insoluble human amylin, and formation of cytotoxic oligomers. In some embodiments, the one or more additional agents may be compounds that prevent or treat an amyloid-related disease of amylin (e.g., type II diabetes).
For example, the one or more additional agents will in certain embodiments be selected from the group comprising blood glucose regulating agents, such as insulin, insulin analogs or derivatives, symlin, GLP agonists, metformin, sulfonylurea, thiazolidinediones, SGLT2 inhibitors, and selective dipeptidyl peptidase (DPP-IV) inhibitors.
In various examples, the GLP-1 agonist is exenatide (exenatide), liraglutide (liraglutide), liraglutide (lixisenatide), abiratide (albiglutide), or dulragide (dulragide), or any combination of two or more thereof.
In various examples, the selective dipeptidyl peptidase (DPP-IV) inhibitor is selected from the group consisting of Sitagliptin (Sitagliptin), vildagliptin (Vildagliptin), saxagliptin (Saxagliptin), linagliptin (Linagliptin), alagliptin (Anagliptin), tergliptin (teneliptin), alogliptin (Alogliptin), trelagliptin (Trelagliptin), gemigliptin (Gemigliptin), dulgliptin (Dutogliptin), and aogliptin (omaigliptin).
In one example, the selective dipeptidyl peptidase (DPP-IV) inhibitor is selected from the group comprising: alogliptin, linagliptin, saxagliptin, sitagliptin, nesina, tradjenta, an Lize (Onglyza) and henovine (Januvia).
In some embodiments, the one or more additional agents may be, for example, agents that reduce side effects associated with the compounds described herein.
In various embodiments, one or more compounds described herein can be administered to an individual with one or more additional agents selected from quinacrine (quinacrine), tetracycline, doxycycline.
In various embodiments employed ex vivo, one or more of the compounds described herein may be administered to or contacted with a sample ex vivo with one or more additional agents selected from the group comprising: anthracene, phenanthrene, quinacrine, neutral red, chlorpromazine, acridine orange, methylene blue, phenyldiazine, phenothiazine, tetracycline, doxycycline, congo red, pyrene,
Figure BDA0001835577520000241
Benzo [ a ]]Anthracene, benzo [ m ]]Anthracene, benzo [ c ]]Phenanthrene and tetracene.
When one or more compounds described herein are administered in combination with one or more additional agents, the dose of each compound administered may be less than the dose that would be administered when the compounds were administered alone.
In some embodiments, when one or more compounds described herein are administered in combination with one or more additional agents, the dose of each compound administered may be greater than the dose that would be administered when the compounds were administered alone. For example, this may be the case when one or more additional agents produce reduced side effects that render larger doses tolerable.
The structural elements (e.g. groups, substituents, heterocyclic members, numbers or other features, e.g. alkyl, groups such as R1, R2, R3, etc., which may occur several times in the compounds of formula I, II or III) may all have, independently of one another, at each occurrence, any indicated meaning and may in each case be identical to or different from one another. For example, the alkyl groups in the dialkylamino groups can be the same or different.
As discussed above, the terms "include" and "comprise" are used herein in their open, non-limiting sense. As used herein, the term "(C 1 -C 6 ) "equivalent" means a moiety having 1 to 6 carbon atoms, etc., respectively. In compositional terms such as "hydroxy- (C) 0 -C 4 ) -alkyl'In, options "(C) 0 ) Alkyl "means a bond (i.e. a hydroxyl group directly bonded in this case), or in unsubstituted" (C) 0 ) In the case of alkyl ", it refers to hydrogen.
As used herein, the term "alkyl" refers to a saturated monovalent hydrocarbon group. As used herein, the term "alkenyl" refers to a monovalent hydrocarbon radical containing at least one carbon-carbon double bond, wherein each double bond may have an E or Z configuration. As used herein, the term "alkynyl" refers to a monovalent hydrocarbon radical containing at least one carbon-carbon triple bond. Alkyl, alkenyl and alkynyl groups may be linear, i.e., straight-chain, or branched. This also applies when it is part of another group, for example an alkyloxy (=alkoxy, O-alkyl), alkyloxycarbonyl or alkyl-substituted amino group, or when it is substituted. The number of carbon atoms in the alkyl group may be 1-22, such as 1 to 12, such as 1, 2, 3, 4, 5 or 6, or 1, 2, 3 or 4, depending on the respective definition. Examples of alkyl groups are methyl, ethyl, propyl including n-propyl and isopropyl, butyl including n-butyl, sec-butyl, isobutyl and tert-butyl, pentyl including n-pentyl, 1-methylbutyl, isopentyl, neopentyl and tert-pentyl, hexyl including n-hexyl, 3-dimethylbutyl and isohexyl. Double and triple bonds in alkenyl and alkynyl groups, respectively, may be present in any position. Examples of alkenyl and alkynyl are vinyl, prop-1-enyl, prop-2-enyl (=allyl), but-2-enyl, 2-methylprop-2-enyl, 3-methylbut-2-enyl, hex-3-enyl, hex-4-enyl, prop-2-ynyl (=propargyl), but-2-ynyl, but-3-ynyl, hex-4-ynyl or hex-5-ynyl. Substituted alkyl, alkenyl and alkynyl groups may be substituted in any position, provided that the corresponding compounds are sufficiently stable and suitable for the desired purpose, such as use as therapeutic substances. The proviso that a particular group and a compound of formula I, a compound of formula II or a compound of formula III are sufficiently stable and suitable for the desired purpose (e.g. use as a therapeutic substance) is generally applied with respect to the definition of all groups in a compound of formula I, a compound of formula II or a compound of formula III.
The term "alkenyloxy" as used herein correspondingly denotes alkenyl-O-groups, wherein alkenyl is the aforementioned alkenyl group; for example, 2-propenyloxy, 2-butenyloxy, 1-methyl-2-propenyloxy, 2-methyl-2-propenyloxy and the like.
As used herein, the term "alkanediyl" or "alkylene" refers to a saturated divalent hydrocarbon group. As used herein, the term "alkenediyl" refers to a divalent hydrocarbon group containing at least one carbon-carbon double bond, wherein each double bond may have an E or Z configuration. As used herein, the term "alkynediyl" refers to a divalent hydrocarbon radical containing at least one carbon-carbon triple bond. The foregoing explanations regarding alkyl, alkenyl and alkynyl apply correspondingly to alkanediyl, alkenediyl and alkynediyl, as far as applicable, which may thus likewise be linear and branched. Examples of divalent alkyl groups are-CH 2 - (=methylene) -CH 2 -CH 2 -、-CH 2 -CH 2 -CH 2 -、-CH 2 -CH 2 -CH 2 -CH 2 -、-CH(CH 3 )-、-C(CH 3 ) 2 -、-CH(CH 3 )-CH 2 -、-CH 2 -CH(CH 3 )-、-C(CH 3 ) 2 -CH 2 -and-CH 2 -C(CH 3 ) 2 -。
As used herein, unless otherwise indicated, the term "cycloalkyl" refers to a monovalent group of a saturated hydrocarbon ring system that is a single ring. In a monocyclic cycloalkyl group, the number of ring carbon atoms may be, for example, 3, 4, 5, 6, 7 or 8. In one embodiment of the invention, the number of ring carbon atoms in the cycloalkyl group, independent of the number of ring carbon atoms in any other cycloalkyl group, is 3, 4, 5 or 6, in another embodiment 3 or 4, in another embodiment 3, in another embodiment 5 or 6, in another embodiment 5, in another embodiment 6. Examples of cycloalkyl groups are cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
As used herein, unless otherwise indicated, the term "heterocycle" refers to cycloalkyl as defined above wherein 1, 2, 3 or 4 carbon atoms are replaced by sulfur, nitrogen or oxygen atoms, provided that the heterocycloalkyl system is stable and suitable as a subunit for the desired purpose of the compound of formula I (e.g., use as a drug substance). Depending on the definition of the corresponding heterocyclyl, in one embodiment of the invention the number of ring heteroatoms present in the heterocyclyl, which may be independent of the number of ring heteroatoms in any other heterocyclyl, is 1 or 2, in another embodiment 1, wherein the ring heteroatoms may be the same or different. The heterocycloalkyl group may be attached by any ring carbon atom or saturated ring nitrogen or oxygen atom.
As used herein, unless otherwise indicated, the term "alkyloxy" (also referred to as "alkoxy") refers to the group-OR a Wherein R is a Are alkyl groups as defined above, e.g., methoxy, ethoxy, propoxy, butoxy, and the like.
As used herein, unless otherwise indicated, the term "halo" refers to fluorine, chlorine, bromine or iodine, preferably fluorine and chlorine. In some embodiments, halo is F. It will be appreciated that in some cases the fluorine atoms may act as isosteres (isosteres) for hydrogen, and accordingly one skilled in the art may in some cases replace, for example, one or more fluorine atoms with one or more hydrogen atoms in the alkyl, alkenyl, aryl and/or cycloalkyl groups.
As used herein, unless otherwise indicated, the term "haloalkyl" refers to an alkyl group substituted with one or more, preferably one, two or three, same or different halogen atoms, e.g., -CH 2 Cl、-CF 3 、-CH 2 CF 3 、-CH 2 CCl 3 Etc.
As used herein, unless otherwise indicated, the term "haloalkoxy" refers to the group-OR b Wherein R is b Is a haloalkyl group as defined above, for example, trifluoromethoxy, trichloroethoxy, 2-dichloropropoxy, and the like.
As used herein, unless otherwise indicated, the term "acyl" refers to the group-C (O) R c Wherein R is c Is hydrogen, alkyl or haloalkyl as defined herein, e.g., formyl, acetyl, trifluoroacetyl, butyryl, and the like.
As used herein, unless otherwise indicated, the term "aryl" refers to all carbon monocyclic or fused ring polycyclic (i.e., rings sharing an adjacent pair of carbon atoms) groups of 6 to 12 carbon atoms having a fully conjugated pi-electron system. Examples of aryl groups, without limitation, are phenyl, naphthyl and anthracenyl. Aryl groups may be substituted or unsubstituted. Unless specifically stated otherwise, "substituted aryl" refers to aryl substituted with one or more, more preferably one, two or three, even more preferably one or two substituents independently selected from the group consisting of: alkyl (where alkyl may be optionally substituted with one or two substituents), haloalkyl, halo, hydroxy, alkoxy, alkylthio, cyano, acyl, nitro, phenoxy, heteroaryl, heteroaryloxy, haloalkyl, haloalkoxy, carboxy, alkoxycarbonyl, amino, alkylamino, dialkylamino, aryl, heteroaryl, carbocycle or heterocycle (where aryl, heteroaryl, carbocycle or heterocycle may be optionally substituted).
As used herein, the prefix "hetero" as part of a group refers to the presence of at least one heteroatom in at least one carbon atom-containing ring of the group. Each ring containing a heteroatom may have 1,2, 3 or 4 heteroatoms selected from N, O and/or S, where the N and S heteroatoms may optionally be oxidized and the N heteroatom may optionally be quaternized. Preferably, the heteroatom will be N or O.
As used herein, unless otherwise indicated, the term "heteroaryl" refers to a monocyclic or fused ring (i.e., rings sharing adjacent pairs of atoms) group of 5 to 12 ring atoms containing one, two, three, or four ring heteroatoms selected from N, O or S, the remaining ring atoms being C, and additionally having a fully conjugated pi-electron system. Examples of typical heteroaryl rings include: a 5-membered monocyclic group such as thienyl, pyrrolyl, imidazolyl, pyrazolyl, furyl, isothiazolyl, furoxanyl (furazanyl), isoxazolyl, thiazolyl, etc.; a 6-membered monocyclic group such as pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, and the like; and polycyclic heterocyclic groups such as benzo [ b ] thienyl, naphtho [2,3-b ] thienyl, thianthrenyl, isobenzofuranyl, benzopyranyl, xanthenyl, oxathianthenyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolinyl, phthalazinyl, naphthyridinyl, quinolyl, quinazolinyl, benzothiazole, benzimidazole, tetrahydroquinolinyl, pteridinyl, carbazolyl, β -carbolinyl, phenanthridinyl, acridinyl, pyridyl, phenanthrinyl, oxazinyl, isothiazolyl, oxazinyl, and the like (see, for example, katritzky, handbook of heterocyclic chemistry (Handbook of Heterocyclic Chemistry)). Other specific examples of heteroaryl rings include 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxadiazolyl, 5-oxadiazolyl, 2-oxazolyl, 4-oxazolyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 3-pyridazinyl, 2-thiazolyl, 4-thiazolyl, 5-tetrazolyl, 2-triazolyl, 5-triazolyl, 2-thienyl, 3-thienyl, carbazolyl, benzimidazolyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, benzotriazole, benzoxazolyl, benzimidazolyl, isoquinolyl, indolyl, isoindolyl, or benzooxazolyl. Heteroaryl further includes groups in which the heteroaryl ring is fused to one or more aromatic or non-aromatic rings in which the linking group or point is on the heteroaryl ring. Examples include tetrahydroquinoline, tetrahydroisoquinoline, and pyrido [3,4-d ] pyrimidinyl, imidazo [1,2-a ] pyrazinyl, imidazo [1,2-a ] pyridinyl, imidazo [1,2-c ] pyrimidinyl, pyrazolo [1,5-a ] [1,3,5] triazinyl, pyrazolo [1,5c ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, imidazo [1,5-a ] pyrimidinyl, pyrazolo [1,5-b ] [1,2,4] triazinyl, quinolinyl, isoquinolinyl, quinoxalinyl, imidazotriazinyl, pyrrolo [2,3-d ] pyrimidinyl, triazolopyrimidinyl, pyridopyrazinyl. Unless specifically stated otherwise, "substituted heteroaryl" refers to heteroaryl substituted with one or more, more preferably one, two or three, even more preferably one or two substituents independently selected from the group consisting of: alkyl (where alkyl may be optionally substituted with one or two substituents), haloalkyl, halo, hydroxy, alkoxy, alkylthio, cyano, acyl, nitro, haloalkyl, haloalkoxy, carboxy, alkoxycarbonyl, amino, alkylamino, dialkylamino, aryl, heteroaryl, carbocycle or heterocycle (where aryl, heteroaryl, carbocycle or heterocycle may be optionally substituted).
As used herein, unless otherwise indicated, the term "carbocycle" refers to a saturated, unsaturated, or aromatic monocyclic or polycyclic ring system having 3 to 14 ring carbon atoms. The term "carbocycle", whether saturated or partially unsaturated, also refers to an optionally substituted ring. The term "carbocycle" includes aryl groups. The term "carbocycle" also includes aliphatic rings fused to one or more aromatic or non-aromatic rings, such as decalin or tetrahydronaphthalin, wherein the attached group or point is on the aliphatic ring. The carbocyclic group may be substituted or unsubstituted. Unless specifically stated otherwise, "substituted carbocycle" refers to a carbocycle group substituted with one or more, more preferably one, two or three, even more preferably one or two substituents independently selected from the group consisting of: alkyl (where alkyl may be optionally substituted with one or two substituents), haloalkyl, halo, hydroxy, alkoxy, alkylthio, cyano, acyl, nitro, haloalkyl, haloalkoxy, carboxy, alkoxycarbonyl, amino, alkylamino, dialkylamino, aryl, heteroaryl, carbocycle or heterocycle (where aryl, heteroaryl, carbocycle or heterocycle may be optionally substituted).
The term "heterocycle" refers to a saturated, unsaturated or aromatic cyclic ring system (cyclic ring system) having 3 to 14 ring atoms, wherein one, two or three ring atoms are selected from N, O or S (O) m Wherein m is an integer from 0 to 2) and the remaining ring atoms are C, wherein one or two C atoms may optionally be replaced by a carbonyl group. The term "heterocycle" includes heteroaryl. Unless specifically stated otherwise, "substituted heterocyclyl" refers to heterocyclyl rings independently substituted with one or more, preferably one, two or three substituents selected from the group consisting of: alkyl (wherein alkyl may be optionally substituted with one or two substituents), haloalkyl, cycloalkylamino, cycloalkylalkylCycloalkyl aminoalkyl, cycloalkylalkylaminoalkyl, cyanoalkyl, halo, nitro, cyano, hydroxy, alkoxy, amino, alkylamino, dialkylamino, hydroxyalkyl, carboxyalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, aralkyl, heteroaralkyl, aryl, heteroaryl, carbocycle, heterocycle (wherein aryl, heteroaryl, carbocycle or heterocycle may be optionally substituted), aralkyl, heteroaralkyl, saturated or unsaturated heterocyclic amino, saturated or unsaturated heterocyclic aminoalkyl, and-COR d (wherein R is d Is alkyl). More specifically, the term heterocyclyl includes, but is not limited to, tetrahydropyranyl, 2-dimethyl-1, 3-dioxolan, piperidinyl, N-methylpiperidin-3-yl, piperazinyl, N-methylpyrrolidin-3-yl, pyrrolidinyl, morpholinyl, 4-cyclopropylmethylpiperazinyl, thiomorpholinyl-1-oxide, thiomorpholinyl-1, 1-dioxide, 4-ethoxycarbonylpiperazinyl, 3-oxopiperazinyl, 2-imidazolidinone, 2-pyrrolidone, 2-oxo homopiperazinyl, tetrahydropyrimidin-2-one and derivatives thereof, including 2-methyl-4, 5,6, 7-tetrahydro-1H-pyrrolo [2,3-c ]]A pyridyl group. In certain embodiments, the heterocyclyl is optionally substituted with one or two substituents independently selected from the group consisting of: halo, alkyl, carboxy-substituted alkyl, ester, hydroxy, alkylamino, saturated or unsaturated heterocycloaminoalkyl or dialkylamino.
As used herein, "carboxy" refers to the group-CO 2 H. Similarly, "alkylcarboxy" refers to the group-CO 2 (alkyl) and "carboxyalkyl" means the group-alkyl-CO 2 H。
As used herein, "optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally alkyl-substituted heterocyclyl" means that an alkyl group may be present but need not be present, and the description includes cases where the heterocyclyl group is substituted with an alkyl group, and cases where the heterocyclyl group is not substituted with an alkyl group.
The term "substituted" as used herein means any of the above groups (e.g., alkyl, aryl, heteroaryl, carbocyclic, heterocyclic, etc.), wherein at least one hydrogen atom is replaced with a substituent, unless otherwise specifically stated. "substituents", as contemplated herein, unless otherwise specified, include halogen, hydroxy, pendant oxy (=o), thio (=s), cyano, nitro, amino, aminoalkyl, alkylamino, dialkylamino, alkyl, cycloalkyl, alkenyl, alkyloxy, sulfanyl, haloalkyl, (e.g., -CF) 3 ) Hydroxyalkyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heteroaryl, substituted heteroaryl, heteroaralkyl, substituted heteroaralkyl, heterocycle, substituted heterocycle, heterocycloalkyl, substituted heterocycloalkyl, -NR e R f 、-NR e C(=O)R f 、-NR e C(=O)NR e R f 、-NR e C(=O)OR f 、-NReSO 2 R f 、-OR e 、-C(=O)R e 、-C(=O)OR e 、-OC(=O)R e 、-C(=O)NR e R f 、-OC(=O)NR e R f 、-SH、-SR e 、-SOR e 、-S(=O) 2 R e 、-OS(=O) 2 R e 、-S(=O) 2 OR e Wherein R is e And R is f The same or different and are independently hydrogen, alkyl, haloalkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heteroaryl, substituted heteroaryl, heteroaralkyl, substituted heteroaralkyl, heterocycle, substituted heterocycle, heterocycloalkyl, or substituted heterocycloalkyl.
Preferred substituents are selected from: halo, C 1-22 Alkyl, C 2-22 Alkenyl, C 6-16 Aryl, C 5-14 Heteroaryl, C 3-12 Cycloalkyl, C 3-12 Heterocycloalkyl, hydroxy C 1-22 Alkyl, amino C 1-22 Alkyl, C 1-22 Alkyloxy, C 1-22 Alkylamino, (C) 1-22 Alkyl) (C) 1-22 Alkyl) amino, C 1-22 Alkylcarbonyloxy, C 1-22 Alkyloxycarbonyl, C 2-22 Alkenylcarbonyloxy, C 2-22 Alkenyloxycarbonyl, C 6-16 Arylcarbonyloxy, C 6-16 Aryloxycarbonyl, C 3-12 Cycloalkyl carbonyl oxy, C 3-12 Cycloalkyl-oxycarbonyl, C 1-22 Alkylcarbonylamino, C 2-22 Alkenyl carbonylamino, C 6-16 Aryl amido, C 3-12 Cycloalkyl carbonylamino, C 1-22 Alkylaminocarbonyl, C 2-22 Alkenylaminocarbonyl, C 6-16 Aryl aminoacyl, C 3-12 Cycloalkyl aminocarbonyl, carboxyl, carboxyalkyl, carboxyalkenyl, =o, =s, =nh, =nnr 32 R 33 、=NNHC(O)R 32 、=NNHCO 2 R 32 Or=nnhso 2 R 32 Wherein R is 32 And R is 33 Independently at each occurrence is hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted cycloalkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl.
General synthetic method
Those skilled in the art will appreciate that the synthetic methods described below are readily adapted to prepare compounds of formula I or formula II, as appropriate.
Chemical synthesis of zbbp92
Figure BDA0001835577520000291
(S1) to a solution of dihydroxyacetophenone in acetone, 2eq of K was added 2 CO 3 And the mixture was stirred at room temperature for 30min. Then 2.2eq of benzyl bromide was added and refluxed overnight. After the reaction was completed, the mixture was concentrated in vacuo and suspended in ethyl acetate. The suspension was washed with water and brine. The organic layer is treated by Na 2 SO 4 Drying and decompressingConcentrating under the condition. The crude product was then purified by column chromatography.
(S2) to a suspension of 2.5eq sodium hydride (60%) in anhydrous toluene was added 2.5eq diethyl carbonate and the mixture was heated to reflux. The solution of S1 in toluene was then added dropwise and the resulting mixture was refluxed for an additional 30min until the hydrogen production ceased. After cooling, a mixture of acetic acid and water (1:1) was added. Then 20mL of ice water was added, then the organic layer was separated, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined and washed several times with brine, over Na 2 SO 4 Dried and concentrated under reduced pressure. The residue was dried to give S2, and it was used directly in the next step.
Figure BDA0001835577520000301
(S3) stirring the mixture of S2 and 2-amino-6-methoxypyridine at 120℃for 6-8 hours without any solvent. After cooling, ethyl acetate was added, and the organic layer was concentrated under reduced pressure. The crude product was then purified by column chromatography.
(Zbbp 92) to a solution of S3 in ethyl acetate was added Pd/C, and stirred at room temperature under an atmosphere of hydrogen for 2.5 hours. After completion, pd/C was filtered with DCM/MeOD and the solvent was concentrated under reduced pressure to give Zbbp92. 1 H NMR(300MHz,DMSO)δ10.44(s,1H),9.98(s,1H),9.42(s,1H),7.67(d,J=5.6Hz,2H),7.38(dt,J=6.6,2.1Hz,2H),6.84(d,J=8.1Hz,1H),6.56-6.49(m,1H),4.09(s,2H),3.84(d,J=7.6Hz,3H)。[M+H] + LC-MS (ESI) calculated 303.1; experimental value 303.1.
Figure BDA0001835577520000303
Chemical synthesis of pyrimidine analogs
Figure BDA0001835577520000302
(S5) to a suspension of 2.5eq sodium hydride (60%) in anhydrous toluene was added 2.5eq diethyl carbonate and the mixture was heated to reflux. A solution of acetophenone analog in toluene was then added dropwise and the resulting mixture was refluxed for another 30min until the hydrogen generation ceased. After cooling, a mixture of acetic acid and water (1:1) was added. Subsequently, 20mL of ice water was added, the organic layer was separated, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined and washed several times with brine, over Na 2 SO 4 Dried and concentrated under reduced pressure. The residue was dried to give S5, and it was used directly in the next step.
(S6) adding 10 mol% BiCl to the mixture of S5 and aminopyridine analogues 3 The mixture was then stirred at 120℃for 6-8 hours. After cooling, ethyl acetate was added, and the organic layer was concentrated under reduced pressure. The crude product was then purified by column chromatography.
(S7) dropwise adding BBr to a solution of S6 in anhydrous DCM at 0deg.C 3 . After the reaction was completed, water was added to quench the reaction and filtered. The filter cake is collected and subsequently purified by column chromatography to give S7 analogues such as zbbp39. 1 H NMR(300MHz,DMSO)δ9.47(s,1H),9.32(s,1H),7.62(d,J=8.7Hz,1H),7.12(s,2H),6.87(dd,J=7.1,5.1Hz,2H),6.78(dd,J=8.1,2.1Hz,1H),6.44(d,J=8.7Hz,1H),5.89(s,1H)。 13 C NMR(101MHz,DMSO)δ161.16(d,J=5.3Hz),147.66(s),146.00(s),137.37(s),126.36(s),120.30(s),116.27(d,J=19.0Hz),106.87(s),106.63(s),102.64(s),40.60(s),40.39(s),40.18(s),39.97(s),39.76(s),39.55(s),39.35(s)。[M+H] + 271.0 calculated for LC-MS (ESI); experimental 271.0.
Figure BDA0001835577520000311
The experiments set forth in the following examples illustrate the invention and are not intended to limit the invention described herein.
Examples
Example 1 verification of Compounds
This example describes the synthesis and analysis of representative compounds as described herein using HNMR, CNMR and MS techniques.
Method
Synthesis and structural analysis
The compounds zbbp144, zbbp148, zbb03-41-2, zbb03-40-2, zbb03-44-1, zbb03-44-2, zbb03-32, and zbb03-41-1 were synthesized according to the modes above and as described below. Structural analysis using LC-MS (ESI) through 1 H NMR、 13 C NMR and MS.
Zbb03-44-2
Figure BDA0001835577520000312
(S1) 1eq of K was added to a solution of 4-hydroxyacetophenone in acetone 2 CO 3 And the mixture was stirred at room temperature for 30min. 1.1eq of benzyl bromide was then added and refluxed overnight. After the reaction was completed, the mixture was concentrated in vacuo and suspended in ethyl acetate. The suspension was washed with water and brine. The organic layer is treated by Na 2 SO 4 Dried, and concentrated under reduced pressure. The crude product was then purified by column chromatography.
S1 is synthesized based on a previous procedure.
(S2) to a suspension of 2.5eq sodium hydride (60%) in anhydrous toluene was added 2.5eq diethyl carbonate and the mixture was heated to reflux. The solution of S1 in toluene was then added dropwise and the resulting mixture was refluxed for an additional 30min until the hydrogen production ceased. After cooling, a mixture of acetic acid and water (1:1) was added. Then 20mL of ice water was added, then the organic layer was separated and the aqueous layer was extracted with ethyl acetate, the organic layers were combined and washed several times with brine, over Na 2 SO 4 Dried, and concentrated under reduced pressure. The residue was dried to give S2, and it was used directly in the next step.
Figure BDA0001835577520000321
S2 is synthesized based on the previous procedure.
(S3) stirring the mixture of S2 and 2-amino-6-methoxypyridine at 120℃for 6-8 hours without any solvent. After cooling, ethyl acetate was added, and the organic layer was concentrated under reduced pressure. The crude product was then purified by column chromatography.
(Zbb 03-44-2) Pd/C was added to a solution of S3 in ethyl acetate and stirred at room temperature under an atmosphere of hydrogen for 2.5 hours. After completion, pd/C was filtered with DCM/MeOD and the solvent was concentrated under reduced pressure to give Zbb03-44-2. 1 H NMR(400MHz,DMSO)δ11.88(s,1H),10.55(s,1H),8.26(dd,J=6.8,4.0Hz,1H),7.90(d,J=8.7Hz,2H),7.25-7.02(m,2H),6.91(dd,J=8.6,6.7Hz,2H),4.29(s,2H),3.99(d,J=5.0Hz,3H)。[M+H] + LC-MS (ESI) calculated 287.0; experimental value 287.1
Figure BDA0001835577520000322
Zbb03-41-1
Figure BDA0001835577520000331
(S4) to a suspension of 2.5eq sodium hydride (60%) in anhydrous toluene was added 2.5eq diethyl carbonate and the mixture was heated to reflux. A solution of 4' -bromoacetophenone in toluene was then added dropwise and the resulting mixture was refluxed for another 30min until the generation of hydrogen was stopped. After cooling, a mixture of acetic acid and water (1:1) was added. Subsequently 20mL of ice water was added, the organic layer was separated and the aqueous layer was extracted with ethyl acetate, the organic layers were combined and washed several times with brine, over Na 2 SO 4 Dried, and concentrated under reduced pressure. The residue was dried to give S4, and it was used directly in the next step.
(S5) stirring the mixture of S4 and 2-amino-6-methoxypyridine at 120℃for 6-8 hours without any solvent. After cooling, ethyl acetate was added, and the organic layer was concentrated under reduced pressure. The crude product was then purified by column chromatography.
(S6) toluene, ethanol, H 2 O=3:2:1 with Pd (pph) 3 ) 4 Fill the tube, then add S5 and K 2 CO 3 3-carboxy-4-methoxyphenylboronic acid. The reaction was carried out under nitrogen at 80℃for 6h. The mixture was diluted with DCM and washed three times with brine. The organic layer was taken up with Na 2 SO 4 And (5) drying. The crude product was then purified by column chromatography.
Figure BDA0001835577520000332
(S7) dropwise adding BBr to a solution of S6 in anhydrous DCM at 0deg.C 3 . After the reaction was completed, water was added to quench the reaction and filtered. The filter cake was collected and subsequently purified by column chromatography to give S7. 1 H NMR(400MHz,DMSO)δ11.73(s,1H),10.53(s,1H),8.18-8.10(m,1H),8.06(d,J=8.4Hz,1H),7.95(dd,J=8.6,2.5Hz,1H),7.89-7.78(m,2H),7.69(d,J=4.8Hz,2H),7.11(d,J=8.7Hz,1H),6.69-6.37(m,1H),4.28(s,2H),3.86(d,J=11.6Hz,3H)。[M+H] + LC-MS (ESI) calculated 287.0; experimental value 407.1.
Figure BDA0001835577520000341
Zbb03-41-2
Figure BDA0001835577520000342
(S8) to a suspension of 2.5eq sodium hydride (60%) in anhydrous toluene was added 2.5eq diethyl carbonate and the mixture was heated to reflux. Subsequently, a solution of 3, 4-dimethoxyacetophenone in toluene was added dropwise and the resulting mixture was refluxed for an additional 30min untilUntil the generation of hydrogen is stopped. After cooling, a mixture of acetic acid and water (1:1) was added. Subsequently 20mL of ice water was added, the organic layer was separated and the aqueous layer was extracted with ethyl acetate, the organic layers were combined and washed several times with brine, over Na 2 SO 4 Dried, and concentrated under reduced pressure. The residue was dried to give S8, and it was used directly in the next step.
(S9) adding 10 mol% BiCl to the mixture of S8 and 6-aminopyridin-2-ol 3 The mixture was then stirred at 120℃for 6-8 hours. After cooling, ethyl acetate was added, and the organic layer was concentrated under reduced pressure. The crude product was then purified by column chromatography.
(S10) dropwise adding BBr to a solution of S9 in anhydrous DCM at 0deg.C 3 . After the reaction was completed, water was added to quench the reaction and filtered. The filter cake is collected and subsequently purified by column chromatography to give S10. 1 H NMR(400MHz,DMSO)δ10.05(s,2H),8.25(s,1H),7.62(d,J=8.6Hz,1H),7.13(s,2H),6.87(dd,J=8.5,5.1Hz,2H),6.78(dd,J=8.1,2.0Hz,1H),6.44(d,J=8.7Hz,1H),5.89(s,1H)。[M+H] + LC-MS (ESI) calculated 287.0; experimental value 271.0
Figure BDA0001835577520000351
Zbbp144
Figure BDA0001835577520000352
(S11) to a suspension of 2.5eq sodium hydride (60%) in anhydrous toluene was added 2.5eq diethyl carbonate and the mixture was heated to reflux. A solution of 4-hydroxyacetophenone in toluene was then added dropwise and the resulting mixture was refluxed for a further 30min until the hydrogen generation ceased. After cooling, a mixture of acetic acid and water (1:1) was added. Subsequently 20mL of ice water was added, the organic layer was separated and the aqueous layer was extracted with ethyl acetate, the organic layers were combined and washed several times with brine, over Na 2 SO 4 Dried, and concentrated under reduced pressure. The residue was dried to give S11, and it was directly used in the next step.
(S12) adding 10 mol% BiCl to the mixture of S11 and 6-aminopyridin-2-ol 3 The mixture was then stirred at 120℃for 6-8 hours. After cooling, ethyl acetate was added, and the organic layer was concentrated under reduced pressure. The crude product was then purified by column chromatography.
(S13) dropwise adding BBr to a solution of S12 in anhydrous DCM at 0deg.C 3 . After the reaction was completed, water was added to quench the reaction and filtered. The filter cake is collected and subsequently purified by column chromatography to give S13. 1 H NMR(400MHz,DMSO)δ9.95(s,1H),7.58(d,J=8.7Hz,1H),7.34(d,J=8.6Hz,2H),7.14(s,2H),6.92(d,J=8.6Hz,2H),6.44(d,J=8.7Hz,1H),5.93(s,1H)。[M+H] + LC-MS (ESI) calculated 287.0; experimental value 255.0
Figure BDA0001835577520000361
Results
Synthesis and structural analysis
Compound zbbp148 is a golden solid, zbb-40-2 and zbb-03-44-2 are yellow solids, zbb03-41-1 is a yellow-white solid, zbb03-32 is a white solid, zbb03-44-1 is a yellow solid, zbbp144 is a yellow solid, and zbb03-41-2 is a green solid. Each compound of the formula 1 H NMR 13 The C NMR data are presented below and further confirmed by MS results (data not shown schematically).
zbbp148
Figure BDA0001835577520000362
1 H NMR(400MHz,DMSO)δ9.79(s,1H),7.50(d,J=8.6Hz,1H),7.34(t,J=7.8Hz,1H),7.18(s,2H),6.95-6.72(m,3H),6.44(d,J=8.6Hz,1H),5.96(s,1H); 13 C NMR(101MHz,DMSO)δ161.33(s),161.02(s),160.30(s),158.02(s),156.19(s),137.11(s),136.75(s),130.49(s),119.32(s),116.97(s),115.41(s),107.73(s),106.80(s),102.54(s),40.60(s),40.39(s),40.18(s),39.97(s),39.76(s),39.55(s),39.34(s);[M+H]LC-MS (ESI) calculated for+ 255.1; experimental value 255.1.
zbb03-40-2
Figure BDA0001835577520000363
1 H NMR(400MHz,DMSO)δ7.62(d,J=8.7Hz,1H),7.14(d,J=13.0Hz,2H),7.10(s,1H),7.08(d,J=1.9Hz,1H),7.05(dd,J=8.2,2.0Hz,1H),6.44(d,J=8.7Hz,1H),6.03(s,1H),3.83(d,J=6.1Hz,6H); 13 C NMR(101MHz,DMSO)δ161.17(d,J=13.0Hz),160.36(s),155.95(s),150.38(s),149.26(s),137.32(s),127.78(s),121.51(s),112.40(s),112.18(s),107.55(s),106.79(s),102.64(s),56.08(d,J=1.7Hz),40.49(d,J=21.0Hz),40.18(s),40.12-40.08(m),39.97(s),39.76(s);[M+H]LC-MS (ESI) calculated for+ 299.1; experimental value 299.1.
zbb03-44-2
Figure BDA0001835577520000371
1 H NMR(400MHz,DMSO)δ11.88(s,1H),10.55(s,1H),8.26(dd,J=6.8,4.0Hz,1H),7.90(d,J=8.7Hz,2H),7.68(d,J=8.8Hz,1H),7.21-7.01(m,2H),7.04-6.74(m,2H),6.10(d,J=27.7Hz,1H),4.29(s,2H),3.99(d,J=5.0Hz,3H); 13 C NMR(101MHz,DMSO)δ192.15(s),170.93(s),169.31(s),163.22(s),161.82(s),149.99(s),142.61(s),131.59(s),128.48(s),127.87(s),123.43(s),116.31(s),115.88(s),114.56(s),109.32(s),98.86(s),87.02(s),57.49(s),48.11(s);[M+H]LC-MS (ESI) calculated 287.1; experimental value 287.1.
zbb03-41-1
Figure BDA0001835577520000372
1 H NMR(400MHz,DMSO)δ10.53(s,1H),8.14(dd,J=4.3,2.5Hz,1H),8.06(d,J=8.4Hz,2H),7.95(dt,J=8.3,4.1Hz,1H),7.89-7.79(m,3H),7.70(dd,J=11.7,4.9Hz,2H),7.10(dd,J=8.7,3.1Hz,1H),6.61-6.47(m,1H),6.36(s,1H),4.28(s,2H),3.85(s,3H);[M+H]LC-MS (ESI) calculated for +407.1; experimental value 407.1.
zbb03-32
Figure BDA0001835577520000381
1 H NMR(400MHz,DMSO)δ12.86(s,1H),10.53(s,1H),8.06(d,J=8.0Hz,1H),8.00(d,J=2.2Hz,1H),7.93(d,J=6.9Hz,1H),7.85(d,J=8.4Hz,2H),7.69(d,J=4.3Hz,2H),7.26(d,J=8.8Hz,1H),6.67-6.43(m,1H),5.76(s,1H),4.28(s,1H),3.87(d,J=14.9Hz,6H); 13 C NMR(101MHz,DMSO)δ167.74(s),162.88(s),158.69(s),150.19(s),144.12(s),141.48(s),135.16(s),131.70(s),130.87(s),129.44(d,J=25.2Hz),126.85(s),122.81(s),113.61(s),105.74(s),56.44(s),53.59(s),40.59(s),40.39(s),40.18(s),39.97(s),39.76(s),39.57(s);[M+H]LC-MS (ESI) calculated for+ 421.1; experimental value 421.1.
zbb03-44-1
Figure BDA0001835577520000382
1 H NMR(400MHz,DMSO)δ10.63(s,1H),9.97(s,1H),9.40(s,1H),8.14(t,J=5.8Hz,1H),7.72(s,1H),7.50-7.23(m,2H),6.84(dd,J=8.2,3.2Hz,1H),6.72(dd,J=5.8,2.3Hz,1H),4.08(s,2H),3.83(d,J=11.5Hz,3H); 13 C NMR(101MHz,DMSO)δ193.04(s),167.19(d,J=15.5Hz),163.51(s),153.85(s),151.56(s),149.45(s),145.74(s),128.71(s),122.35(s),115.53(d,J=6.2Hz),106.87(s),99.01(s),55.75(s),47.90(s),40.60(s),40.39(s),40.18(s),39.97(s),39.76(s),39.55(s),39.34(s);[M+H]LC-MS (ESI) calculated for +303.1; experimental value 303.1.
zbbp144
Figure BDA0001835577520000383
1 H NMR(300MHz,DMSO)δ10.44(s,1H),9.95(s,1H),9.38(s,1H),8.01(d,J=5.7Hz,1H),7.36(dt,J=4.9,2.1Hz,2H),7.09-6.93(m,2H),6.82(d,J=8.2Hz,1H),4.01(s,2H),3.80(d,J=3.4Hz,4H); 13 C NMR(101 MHz,DMSO)δ161.17(d,J=9.1Hz),160.37(s),159.35(s),156.08(s),137.27(s),130.43(s),125.97(s),116.09(s),107.09(s),106.70(s),102.63(s),40.59(s),40.38(s),40.18(s),39.97(s),39.76(s),39.55(s),39.34(s);[M+H]LC-MS (ESI) calculated for+ 255.1; experimental value 255.1.
zbb03-41-2
Figure BDA0001835577520000391
1 H NMR(400MHz,DMSO)δ8.25(s,1H),7.62(d,J=8.6Hz,1H),7.13(s,2H),6.87(dd,J=8.5,5.1Hz,2H),6.78(dd,J=8.1,2.0Hz,1H),6.44(d,J=8.7Hz,1H),5.89(s,1H); 13 C NMR(101MHz,DMSO)δ161.16(d,J=5.3Hz),147.66(s),146.00(s),137.37(s),126.36(s),120.30(s),116.27(d,J=19.0Hz),106.87(s),106.63(s),102.64(s),40.60(s),40.39(s),40.18(s),39.97(s),39.76(s),39.55(s),39.35(s);[M+H]LC-MS (ESI) calculated 271.1 of +; experimental 271.1.
EXAMPLE 2 inhibition of amylin fibril formation
This example describes an assay for representative compounds of formula III using a thioflavin-T assay to assess compound-mediated inhibition of amylin fibril formation.
Method
Solubility and spectroscopic analysis
The test compounds (zbbp 92, zbb03-3P5, LZQ0316 and zbb 03-44-1) dissolved with the addition of 1M NaOH and were all 100% soluble in water at pH 11 or higher and were pale yellow (pale yellow) when dissolved. The compounds were titrated to pH 8-9 with 1M HCl and the solubility was measured immediately and then stored overnight and for a longer period of time.
The absorbance of all compounds at pH 10.8 was measured using a UV-Vid spectrophotometer (Molecular Devices SpectraMax, not shown).
thioflavin-T analysis
The ability of compounds to inhibit human amylin (hA) fibril formation was assessed using thioflavin-T assays as described in j.f. aitken et al, inhibition of polycyclic compounds by conversion of human amylin to insoluble amylin amyloid (Suppression by polycyclic compounds of the conversion of human amylin into insoluble amylin amyloid.), biochem J374 (2003) 779-784. Inhibition was assessed at three hA: compound ratios (1:1, 1:0.1, 1:0.01) for each compound.
Results
Solubility and spectroscopic analysis
Compound zbbp92, zbb03-3P5, LZQ0316 remained 100% soluble during initial suspension and for titration and pH adjustment, while the solubility of compound zbb03-44-1 (95%) could be slightly lower at pH 8-9. The compounds remained soluble overnight and stored for long periods of time. The compound is sensitive to light upon dissolution and becomes progressively darker in color over time.
All 4 compounds underwent discoloration with the addition of 1M HCl, becoming either very pale yellow (zbbp 92, zbb03-3P5, zbb-44-1) or colorless (LZQ 0316).
The absorbance of each compound was measured over a spectrum from 400nm to 550nm at pH 10.8 (data not shown).
thioflavin-T analysis
Inhibition of hA fibril formation was observed for each compound, and fibril inhibition was dependent on the hA: compound ratio and initial concentration of hA. As can be seen in FIG. 1A, complete inhibition of fibril formation was observed for each compound at a hA: compound ratio of 1:1. Inhibition of hA fibril formation was observed at an hA: compound ratio of 1:0.1 as shown in FIG. 1B, especially for compound zbbp92, and to a lesser extent at an hA: compound ratio of 1:0.01 as shown in FIG. 1C.
The time course depicted in fig. 1A-1C shows that even at lower concentrations of the compound, if the compound prevents an initial nucleation event, the compound has greater inhibition of fibril formation over time.
Example 3-inhibition of amylin-induced cell death
This example describes an assay for representative compounds of formula III using a cell death assay to assess compound-mediated inhibition of amylin-induced cell death.
Method
Test compound zbbp92; LZQ0316; ZBB03-3P5; and ZBB03 44-1.
CM cells were cultured overnight in RPMI medium. A stock solution of human amylin (hA) (500 μm) was freshly prepared in water and diluted in medium to a final concentration of 10 μm. Aliquots of hA were then premixed with the various compounds in a molar ratio of 1:1 and 1:10 and incubated for 2hr at room temperature. The mixture was then added to the cells and incubated overnight for 16-20 hours. Untreated control cells were similarly treated, and treatment with Kp7-6 peptide served as a positive treatment control group.
Apoptotic beta cell death was measured using cell death detection Elisa, such as Zhang S diabetes 57, 348-356, 2008; described in journal of biochemistry (J Biol Chem) 278, 52810-52819, 2003, as in the ROCHE cell death detection ELISA plus catalog number: 11774425001.
Results
The results shown in fig. 2 represent the enrichment of nucleosomes in the sample (cell lysate) and appear to be related to the control group (Co) set to one. Values are the mean ± SE of four independent experiments, each in duplicate. * P is less than 0.05; * P < 0.01 vs control group; * P < 0.001 vs control group; the hA-treated cells were subjected to #P < 0.05.
As can be clearly seen in fig. 2, at hA: compound molar ratios of 1:1 and 1:10, statistically significant inhibition of amylin-induced cell death was observed for each compound. Little or no compound-mediated cell death (i.e., in the absence of amylin) was observed.
Example 4 inhibition of formation of amylin fibrils
This example describes an analysis of representative compounds of formula II and III using a thioflavin-T analysis to assess compound-mediated inhibition of amylin fibril formation.
Method
Solubility and physical characteristics
Solutions of test compounds (zbbp 144, zbbp148, zbbo3 40-2, zbb03-44-2, zbb03-41-2, zbb03-44-1, zbbo3 41-1, and zbb 03-32) were prepared as follows.
Zbbp144: light yellow solid. Mass 254.1
1M NaOH was added. The partial solubility was about 70%. Pale yellow, ph=11.3. Hold for 30min, completely soluble. 1M HCl, pH 8.9, slightly cloudy, and some precipitate was added. 1M NaOH- -was added- -the fully soluble pale yellow solution pH 10.4, but less than pH 10 was insoluble.
Zbbp148: golden solid. Mass 254.1. Completely insoluble pH 11.3 and pH 2.1.
zbbp03-41-2: green solid. Mass 270.1
1M NaOH- -gold/orange was added to be partially soluble, maintaining the 10min- -100% soluble pH 11.2.
1M HCl,pH 9.2,1M HCl,pH 8.3, red, was added, still 100% soluble. A loss of solubility, about 70% solubility, was achieved overnight.
zbb03-40-2: an off-white solid. Mass 298.1. Completely insoluble pH 11.1 and pH 1.8.
zbb03-44-1: as described in example 2 above.
zbb03-44-2: pale yellow solid. Mass 286.1
1M NaOH was added- -only partially soluble about 50%. The solution was kept clear at 100% solubility for 30min, pH 10.4. 1M HCl, pH9.4, was added, still 100% soluble clear solution. 1M HCl, pH 8.7, was added, still 100% soluble clear solution. Still 100% soluble overnight.
zbb03-32: white solid. Mass 420.1
1M NaOH was added- -partially soluble about 50%. The solution was clarified and maintained at 100% soluble pH 10.6 for 30 min. 1M HCl, pH 8.4, was added, still 100% soluble clear solution. Still 100% soluble overnight.
zbb03-41-1: mass 406.1. Light yellow (Light yellow) solid. 1M NaOH- -off-white/pale yellow solution 100% soluble pH 10.6 was added. 1M HCl, pH 9.3, was added and the color slightly changed to a clear solution, still 100% soluble. Adding
1M HCl, pH 8.4, clarifies the solution, and remains 100% soluble. Still 100% soluble overnight.
thioflavin-T analysis
The ability of compounds to inhibit human amylin (hA) fibril formation was assessed using thioflavin-T assays as described in j.f. aitken et al, inhibition of polycyclic compounds by conversion of human amylin to insoluble amylin amyloid, biochem J374 (2003) 779-784. Inhibition was assessed at three hA: compound ratios (1:1, 1:0.1, 1:0.01) for each compound.
Results
Thioflavin T analysis
Inhibition of hA fibril formation was evaluated for each compound, and at least some inhibition was observed for each compound except zbb 03-32. Fibril inhibition depends on the ratio of hA to compound and the initial concentration of hA.
As can be seen in FIG. 3, complete inhibition of fibril formation of compound zbb03-41-2 was observed at a hA: compound ratio of 1:1 for the duration of the analysis. Strong inhibition of fibril formation of zbb03-41-1 was also observed:
compounds zbb-44-2, zbbp144 and zbb03-32 exhibited little inhibition of fibril formation. For zbb03-32 and zbbp144, it is believed that the lack of observed fibril inhibition was due to lack of solubility at pH 7.4 at which the assay was performed.
The time course depicted in FIGS. 4A through 4C shows that even at lower concentrations of compounds, if the selected compounds prevent initial nucleation events, the selected compounds zbb-41-1 and zbb-03-41-2 are able to inhibit fibril formation at higher hA: compound ratios and have greater inhibition of fibril formation over time. Both compounds still inhibited amylin fibril formation at a ratio of hA to compound of 1:0.01.
Example 5-inhibition of amylin-induced cell death
This example describes an analysis of representative compounds of formula II and III using a cell death assay to assess compound-mediated inhibition of amylin-induced cell death.
Method
Compounds zbbp90 and zbbp92 were tested.
RINm5F and CM cells were incubated and pre-incubated with test compounds at various concentrations of 1h (1:1 and 1:10 molar ratio of human amylin) prior to the addition of amylin. A stock solution of amylin was freshly prepared by dissolving lyophilized human amylin in 500 μm water. Aliquots were then added to 10 μm culture (final) followed by incubation overnight for 16-20 hours. Untreated control cells were similarly treated, and treatment with Kp7-6 served as a positive treatment control group.
Apoptotic beta cell death was measured using cell death detection Elisa as described above.
Results
The results shown in fig. 5 represent the enrichment of nucleosomes in the sample (cell lysate) and appear to be related to the control group (Co) set to one. Values are the mean ± SE of four independent experiments, each in duplicate. * P is less than 0.05; * P < 0.01 vs control group; * P < 0.001 vs control group; the hA-treated cells were subjected to #P < 0.05.
As can be clearly seen in fig. 5A (CM cells) and fig. 5B (RINm 5F cells), a statistically significant inhibition of amylin-induced cell death was observed for each compound at a molar ratio of hA: compound of 1:1 and 1:10. Little or no compound-mediated cell death (i.e., in the absence of amylin) was observed.
Example 6-inhibition of amylin-induced cell death
This example describes an assay of representative compounds described herein, using a cell death assay to evaluate compound-mediated inhibition of amylin-induced cell death.
Method
Test compound zbbp92; LZQ0316; ZBB03-3P5; ZBB03 41-1 and ZBB03 41-2.
RINm5F and CM cells were incubated and pre-incubated with test compounds at various concentrations of 1h (1:10 molar ratio of human amylin) prior to the addition of amylin. A stock solution of amylin was freshly prepared by dissolving lyophilized human amylin in 500 μm water. Aliquots were then added to 10 μm culture (final) followed by incubation overnight for 16-20 hours. Untreated control cells were similarly treated, and treatment with Kp7-6 served as a positive treatment control group.
Apoptotic beta cell death was measured using cell death detection Elisa as described above.
Results
The results shown in fig. 6 represent the enrichment of nucleosomes in the sample (cell lysate) and appear to be related to the control group (Co) set to one. Values are the mean ± SE of four independent experiments, each in duplicate. * P is less than 0.05; * P < 0.001 vs control group; the hA-treated cells were subjected to #P < 0.05.
As can be clearly seen in fig. 6A (CM cells) and 6B (RINm 5F cells), statistically significant inhibition of amylin-induced cell death was observed for these compounds at a molar ratio of hA: compound of 1:10. Little or no compound-mediated cell death (i.e., in the absence of amylin) was observed for all compounds except ZBB03 41-2.
Conclusion(s)
The goal of these experiments was to test the effect of the compounds on the misfolding of human amylin and to test their in vitro efficacy in human amylin transgenic mouse lines, which overexpress human amylin in pancreatic β -cells.
Thioflavin-t fluorescence has been shown to inhibit misfolding and aggregation of hA in vitro by treatment with representative compounds (fig. 1, 3, 4). Cell death assays (fig. 2, 5, 6) showed that representative compounds can suppress amylin-induced apoptotic cell death.
These data provide evidence for the deagglomeration of hA oligomers in the presence of compounds as described herein. Without wishing to be bound by any theory, these results are consistent with the manipulation of compound-mediated interactions with soluble hA oligomers, which are thought to prevent their conversion to cytotoxic structures and thus protect islet β -cells from cell death.

Claims (11)

1. A compound of formula IIb:
Figure FDA0004088020120000011
IIb
Wherein:
R 8 is hydrogen;
R 9 is hydrogen;
R 10 is hydrogen;
each R 13 Independently selected from hydroxy, carboxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
each R 14 Independently selected from hydroxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
a is selected from
Figure FDA0004088020120000012
p is 1 or 2;
q is 1 or 2;
r is 0 or 1;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, which is a compound of formula IIc:
Figure FDA0004088020120000013
IIc
Wherein:
R 8 is hydrogen;
R 9 is hydrogen;
R 10 is hydrogen;
each R 13 Independently selected from hydroxy, carboxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
R 15 selected from hydroxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
R 16 selected from hydroxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
A is selected from
Figure FDA0004088020120000021
p is 1 or 2;
r is 0 or 1;
or a pharmaceutically acceptable salt thereof.
3. A compound of formula IId:
Figure FDA0004088020120000022
IId (IId)
Wherein:
R 8 is hydrogen;
R 9 is hydrogen;
R 10 is hydrogen;
R 15 selected from hydroxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
R 16 selected from hydroxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
R 17 selected from hydroxy, carboxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
R 18 selected from hydroxy, carboxy, halo, and C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
a is selected from
Figure FDA0004088020120000023
r is 0 or 1;
or a pharmaceutically acceptable salt thereof.
4. The compound according to claim 1, wherein
R 8 Is hydrogen;
R 9 is hydrogen;
R 10 is hydrogen;
R 13 a hydroxy, carboxyl, halo or C1-C6 alkyloxy optionally substituted with one or more halogen atoms;
R 14 a C1-C6 alkyloxy group which is halo or optionally substituted with one or more halogen atoms;
a is benzene-1, 4-yl;
or a pharmaceutically acceptable salt thereof.
5. A compound selected from:
Figure FDA0004088020120000031
/>
Figure FDA0004088020120000041
6. a pharmaceutical composition comprising a compound according to any one of claims 1 to 5.
7. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of an amyloid-related disease of amylin.
8. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inhibiting, preventing or reversing an amyloidosis or an amyloid fibril or an amyloid plaque formation in a subject in need thereof.
9. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inhibiting, preventing or reversing an amyloidosis of an amylin or the formation of one or more amyloid fibrils or amyloid plaques.
10. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of diabetes.
11. Use of a compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prevention of langerhans' islet β -cell death in a subject in need thereof.
CN201811226113.8A 2018-10-19 2018-10-19 Compounds for the treatment of diabetes and/or related disorders Active CN111072652B (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN201811226113.8A CN111072652B (en) 2018-10-19 2018-10-19 Compounds for the treatment of diabetes and/or related disorders
PCT/NZ2019/050138 WO2020080960A1 (en) 2018-10-19 2019-10-18 Compounds for treating diabetes and/or related conditions
US17/286,794 US20220041591A1 (en) 2018-10-19 2019-10-18 Compounds for treating diabetes and/or related conditions
EP19872857.8A EP3867227A4 (en) 2018-10-19 2019-10-18 Compounds for treating diabetes and/or related conditions

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811226113.8A CN111072652B (en) 2018-10-19 2018-10-19 Compounds for the treatment of diabetes and/or related disorders

Publications (2)

Publication Number Publication Date
CN111072652A CN111072652A (en) 2020-04-28
CN111072652B true CN111072652B (en) 2023-05-23

Family

ID=70308251

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811226113.8A Active CN111072652B (en) 2018-10-19 2018-10-19 Compounds for the treatment of diabetes and/or related disorders

Country Status (1)

Country Link
CN (1) CN111072652B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005009435A1 (en) * 2003-07-25 2005-02-03 Pfizer Inc. Aminopyrazole compounds and use as chk1 inhibitors
WO2011014299A2 (en) * 2009-06-05 2011-02-03 Beth Israel Deaconess Medical Center Identification and use of small molecules to modulate transcription factor function and to treat transcription factor associated diseases
WO2014207213A1 (en) * 2013-06-28 2014-12-31 Medizinische Universität Innsbruck Novel inhibitors of protein kinase c epsilon signaling

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CL2007003841A1 (en) * 2006-12-28 2008-03-14 Metabolex Inc COMPOUNDS DERIVED FROM 4 (5H-1 LAMBDA-TIAZOL-2-IL) PIPERIDINE; PHARMACEUTICAL COMPOSITION CONTAINING THESE COMPOUNDS; AND ITS USE TO TREAT DIABETES.

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005009435A1 (en) * 2003-07-25 2005-02-03 Pfizer Inc. Aminopyrazole compounds and use as chk1 inhibitors
WO2011014299A2 (en) * 2009-06-05 2011-02-03 Beth Israel Deaconess Medical Center Identification and use of small molecules to modulate transcription factor function and to treat transcription factor associated diseases
WO2014207213A1 (en) * 2013-06-28 2014-12-31 Medizinische Universität Innsbruck Novel inhibitors of protein kinase c epsilon signaling

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
《Certain Azomethines in the Pyrimidazolone Series》;C. F. H. Allen等;《Communication》;19441108;第66卷;第1805-1810页 *
《Novel lanthanide hybrid functional materials for high performance luminescence application: The relationship between structures and photophysical behaviors》;Jun Xu等;《Materials Chemistry and Physics》;20121231;第136卷;第112-119页 *
《Synthesis and Anti-inflammatory Activity of N-(4,6-Dimethylpyridin-2-yl) β-Ketoamides and Derivatives》;S. ROBERT-PIESSARD等;《Pharmaceutical Sciences》;19971231;第3卷;第295-299页 *
《SYNTHESIS AND EVALUATION OF 4-ARYL-2(1H)-QUINOLINONES AS POTENT AMYLOID β FIBRILLOGENESIS INHIBITORS》;Yoko Shimokawa等;《HETEROCYCLES》;20120615;第85卷(第8期);第1933-1940页 *
《Synthesis of N-Pyridinyl(methyl)-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carboxamides and analogues and their anti-infammatory activity in mice and rats》;X Collin等;《Pharmacy and Pharmacology》;20011231;第53卷;第417-423页 *

Also Published As

Publication number Publication date
CN111072652A (en) 2020-04-28

Similar Documents

Publication Publication Date Title
CN108430994B (en) Compounds, compositions and methods for modulating CFTR
CA2787360C (en) Pyrrol0[3,2-c]pyridinyl-4-benzamide compounds and their use as apoptosis signal-regulating kinase 1 inhibitors
JP6498705B2 (en) Dihydropyridinone MGAT2 inhibitor for use in the treatment of metabolic disorders
JP6522602B2 (en) Tricyclic pyrido-carboxamide derivatives as ROCK inhibitors
JP2019038839A (en) Novel bicyclic bromodomain inhibitors
EP2861590B1 (en) Compound as wnt signaling inhibitor, composition, and use thereof
JP6449919B2 (en) Tetrazolone-substituted dihydropyridinone MGAT2 inhibitors
CN105682655B (en) Antimicrobial compounds
CA3122105A1 (en) Bicyclic derivatives
JP6417405B2 (en) Dihydropyridinone MGAT2 inhibitor
JP2019512474A (en) Cyano substituted indole compounds and their use as LSD1 inhibitors
EP3068389B1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
KR20140099519A (en) Aryl dihydropyridinone and piperidinone as mgat2 inhibitors
US20160009681A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
WO2012130322A1 (en) Imidazo [1,2-a]pyridine compounds for use in therapy
JP2020506878A (en) Activator of TREK (TWIK related K channel) channel
US9895358B2 (en) Combination therapies for treatment of spinal muscular atrophy
CN111072652B (en) Compounds for the treatment of diabetes and/or related disorders
JP2015531382A (en) Pyrrolidine
US10723716B2 (en) Alpha-helix mimetics as modulators of Abeta self-assembly
NZ241140A (en) Pharmaceutical compositions comprising condensed dihydropyridines and tautomers, and selected compounds
CA2504820C (en) Pyrazolonaphthyridine derivatives
US20220041591A1 (en) Compounds for treating diabetes and/or related conditions
KR102623218B1 (en) Phenylacetic acid derivatives and composition for preventing or treating autoimmune diseases comprising the same
JP2019519576A (en) Macrocycle inhibitors of myeloperoxidase

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant