CN106008503B - Spirocyclic aryl sulfones as protein kinase inhibitors - Google Patents

Spirocyclic aryl sulfones as protein kinase inhibitors Download PDF

Info

Publication number
CN106008503B
CN106008503B CN201610179546.7A CN201610179546A CN106008503B CN 106008503 B CN106008503 B CN 106008503B CN 201610179546 A CN201610179546 A CN 201610179546A CN 106008503 B CN106008503 B CN 106008503B
Authority
CN
China
Prior art keywords
compound
acid
pharmaceutically acceptable
reaction
acceptable salt
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
CN201610179546.7A
Other languages
Chinese (zh)
Other versions
CN106008503A (en
Inventor
丁照中
陈曙辉
赵保平
刘希乐
鄢笑非
张路
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.)
Qilu Pharmaceutical Co Ltd
Original Assignee
Qilu Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Qilu Pharmaceutical Co Ltd filed Critical Qilu Pharmaceutical Co Ltd
Publication of CN106008503A publication Critical patent/CN106008503A/en
Application granted granted Critical
Publication of CN106008503B publication Critical patent/CN106008503B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

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
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses a spiro aryl sulfone compound, in particular a compound shown as a formula (I) as an ALK inhibitor.

Description

Spirocyclic aryl sulfones as protein kinase inhibitors
Technical Field
The present invention relates to spirocyclic aryl sulfone compounds, particularly compounds of formula (I) as ALK inhibitors.
Background
Protein kinases can regulate various links of the cell cycle; the abnormal expression of protein kinase can cause diseases, and the diseases also can cause the abnormal expression of protein kinase, and the diseases comprise cancer, inflammation, diabetes and the like, so the protein kinase is gradually becoming a more popular drug target.
Since the time of its introduction into the market of gleevec 2001, many protein kinases have become very attractive targets for cancer therapy. Anaplastic Lymphoma Kinase (ALK) has attracted a great deal of attention because of its potential oncogenes in many human cancers and its important role in the pathogenesis of cancers, such as ALCLS, non-small cell lung cancer, breast cancer, colorectal cancer, neuroblastoma, ovarian cancer, etc.
Anaplastic Lymphoma Kinase (ALK) is a Receptor Tyrosine Kinase (RTK), a member of the Insulin Receptor (IR) superfamily. ALK was first discovered in 1994 in the 60-80% Anaplastic Large Cell Lymphoma (ALCLS) cell line as a fusion protein NPM (nucleophosmin) -ALK, which is caused by a t (2; 5) chromosomal translocation. Although the physiological function of ALK in cancer is not yet known, ALK fusion proteins have been found in ALCLs as well as in various human cancers, such as breast cancer, colorectal cancer, Inflammatory Myofibroblastic Tumors (IMT), diffuse large B-cell lymphoma (DLBCL), the most significant of which is in non-small cell lung cancer (NSCLC). Therefore, the kinase activity associated with ALK fusion proteins is considered to play a very important role in human cancer cell survival and proliferation.
The ALK gene provides an instruction for signal transduction, allowing receptor tyrosine kinases to transmit signals from the cell surface into the cell. This process starts when the kinase is stimulated on the cell surface and then attaches to a similar kinase (dimerisation). After dimerization, the kinase is tagged with a phosphate group, a process called phosphorylation. Phosphorylation activates kinases, another protein capable of transferring phosphate groups into cells, and activation continues through a series of proteins in signaling pathways. These signaling pathways are important for many cellular processes, such as cell growth and division (proliferation) or maturation (differentiation).
Often the ALK chromosome rearranges such that the tyrosine kinase domain of ALK fuses with domains at the 5' -end of other proteins, such as NSCLC or Nucleophosmin (NPM) in echinoderm microtubule-associated protein-like 4(EML4) in anaplastic large cell lymphoma. The breakpoint for all ALK fusion genes is well conserved, located in the upstream kinase domain of the exon-encoded intragenic region. Since the rearranged portion involved in ALK does not include a transmembrane domain, the fusion protein formed migrates back from the cell membrane to the cytoplasm. The 5' -end of the fusion protein typically contains a coiled coil or leucine zipper domain, thereby oligomerizing the fusion protein and resulting in ligand-dependent activation of the ALK tyrosine kinase. This, in turn, constitutively activates downstream signaling, such as the Ras/MAPK, PI3K/AKT, and JAK/STAT pathways. ALK-driven lung cancer responds and regresses upon treatment with ALK small molecule tyrosine kinase inhibitors. This finding suggests "tumor gene dependence", i.e., cancer cells become dependent on oncogenes and are therefore highly sensitive to inhibiting oncogenes.
Crizotinib is the first ALK inhibitor approved by the FDA for the treatment of ALK-positive lung cancer. Although very effective initially in response to treatment with Crizotinib, most patients relapse in the first year of treatment due to development of resistance. The mechanism of resistance to criptiotinib is generally divided into two major classes, the first being genetic alteration of the target, that is to say alteration of ALK by mutation or gene amplification. A number of ALK kinase domain resistance mutations have been identified from patient samples, including L1196M and C1156Y G1269A, 1151Tins, L1152R, G1202R and S1206Y. Approximately one third of patients relapse during treatment with criptotritinib due to mutation and/or amplification of the ALK fusion gene. The second type of resistance mechanism is that other signaling pathways can be activated, bypassing ALK. For example, upregulation of epidermal growth factor receptor signaling has been observed in vitro ALK-positive crizotinib-resistant cell lines, consistent with preclinical hypertonic results, with nearly half of ALK-positive tumors showing EGFR activation when crizotinib is resistant. Clinically, recurrence of treatment with Crizotinib is often associated with metastasis of the Central Nervous System (CNS). This finding also raises a pharmacokinetic problem, insufficient drug exposure, which may be responsible for the high CNS recurrence rate in ALK-positive patients.
On 29 months 4 2014, FDA approved certinib for the treatment of Anaplastic Lymphoma Kinase (ALK) positive metastatic non-small cell lung cancer (NSCLC), including patients who are effective as well as resistant to Crizotinib. Still other compounds are in clinical development for the treatment of cancer, such as Alectinib, AP-26113, X-396, PF-06463992, LDK378, and others. Some heterocyclic compounds have also been disclosed for use in the treatment of various cancers, including WO2014033136, WO2014025128, WO2014006554, WO2014002922, WO2013192512, WO2013177092, WO2013148857, WO2013138210, WO2012139499, WO2012140114, WO 2012048259.
Figure BDA0000950558680000021
However, although more than half of NSCLC patients have good therapeutic effects on Crizotinib, resistance always develops with the treatment time, and thus the drug loses its effectiveness. Although ALK inhibitors for treating non-small cell lung cancer have been vigorously developed at home and abroad in recent years, their therapeutic effects are not satisfactory. Therefore, there is a great need to develop new, more effective and safer ALK inhibitors.
Disclosure of Invention
The invention aims to provide a compound shown as a formula (I) or a pharmaceutically acceptable salt thereof
Figure BDA0000950558680000022
Wherein the content of the first and second substances,
T1selected from N or C (R)01);
T2Is selected from-N (R)01)-、O、S(=O)2、-CH(NR01R02) -or-C (═ O) N (R)01)-;
R01、R02Each independently selected from H, F, Cl, Br, I, CN, OH, SH, NH2Or optionally substituted with 1, 2 or 3 halogen, hydroxy, amino and/or cyano groups: c1-6Alkyl radical, C1-6Heteroalkyl group, C3-6Cycloalkyl- (CH)2)0-3-and C3-6Heterocycloalkyl- (CH)2)0-3-, wherein said "hetero" represents 1, 2 or 3 heteroatoms or heteroatom groups selected from O, S, N, S (═ O)2Or S (═ O);
optionally, T2R of (A) to01And R02Are mutually connected to the same N to form 1-6 membered ring containing 1, 2 or 3A heteroatom selected from O, S and N;
D1~D4each independently selected from- (CR)1R2)1-3-、O、S、C(=O)、S(=O)2And S (═ O);
R3is selected from R03、OR03And SR03
R03Is selected from C1-4Alkyl radical, C1-4Haloalkyl and C3-5Cycloalkyl- (CH)2)0-3-;
Z is selected from N and C (R)4);
R5Is selected from C1-4An alkyl group;
R1、R2and R4Each independently selected from H, F, Cl, Br, I, CN, OH, SH, NH2Or optionally substituted with 1, 2 or 3 halogen, hydroxy and/or cyano groups: c1-6Alkyl radical, C1-6Heteroalkyl group, C3-6Cycloalkyl- (CH)2)0-3-and C3-6Heterocycloalkyl- (CH)2)0-3-, wherein said "hetero" represents 1, 2, or 3 heteroatoms selected from O, S and N.
In one embodiment of the present invention, R is01And R02Each independently selected from H, -CH3、-CD3、-CF3、-CHF2、-CH2CH3、CH(CH3)2、-CH2CF3、-CH2CH2CF3、-CH2CH2F、-CH2CH2S(=O)2CH3、-CH2CH2CN、
Figure BDA0000950558680000031
Figure BDA0000950558680000032
-CH2CH(OH)(CH3)2、-CH2CH(F)(CH3)2、-CH2CH2F。
In one embodiment of the present invention, NR01R02Selected from NHCH3、N(CH3)2、N(CH2CH3)2
Figure BDA0000950558680000033
In one embodiment of the present invention, R is03Is selected from-CH3、-CD3、-CF3、-CHF2、-CH2CH3、-CH(CH3)2、-CH2CF3、-CH2CH2CF3、-CH2CH2F and
Figure BDA0000950558680000034
in one embodiment of the present invention, R is5Is selected from-CH (CH)3)2
In one embodiment of the present invention, R is1、R2And R4Are respectively and independently selected from H, F, Cl, Br, I and-CH3、-CD3、-CF3、-CHF2、-CH2CH3、-CH(CH3)2、-CH2CF3、-CH2CH2CF3、-CH2CH2F、-CH2CH2S(=O)2CH3、-CH2CH2CN、
Figure BDA0000950558680000035
Figure BDA0000950558680000036
-CH2CH(OH)(CH3)2、-CH2CH(F)(CH3)2and-CH2CH2F。
In an embodiment of the present invention, the above-mentioned T2Selected from-NH-, -N (Me) -, -C (═ O) NH-, -C (═ O) N (CH)3) -, -O-and-CH (NCH)3CH3)-。
One of the present inventionIn the above-mentioned aspect, the spiro structural unit
Figure BDA0000950558680000037
Selected from:
Figure BDA0000950558680000038
Figure BDA0000950558680000041
and
Figure BDA0000950558680000042
in one embodiment of the present invention, the above compound or a pharmaceutically acceptable salt thereof is selected from:
Figure BDA0000950558680000043
it is another object of the present invention to provide a process for preparing the above compound, wherein T1Represents N or CH, T2Represents NH, the preparation route of which is shown in scheme A or C:
scheme A
Figure BDA0000950558680000044
Scheme C
Figure BDA0000950558680000051
Wherein PG is an amino protecting group, preferably selected from BOC, Bn and Cbz.
In one embodiment of the present invention, the preparation of A5 is as shown in scheme B:
scheme B
Figure BDA0000950558680000052
The invention aims to treat non-small cell lung cancer and other cancers caused by ALK and EGFR and mutation of ALK and EGFR by using the compounds.
It is also an object of the present invention to use the above compounds in combination with inhibitors of ROS1, BRAF, c-MET, HER2, KRAS/MEK, PIK3CA, FDFR, DDR2, VEGFR, and the like, for the treatment of cancer, as well as in combination with cytotoxins, such as docetaxel, carboplatin, and the like.
Definitions and explanations
C1-6Is selected from C1,C2,C3,C4,C5And C6Constituent groups, the number representing the number of carbon atoms; c3-6Is selected from C3,C4,C5And C and6the constituent groups.
C1-6Alkyl radical, C1-6Heteroalkyl group, C3-6Cycloalkyl radical, C3-6Heterocycloalkyl radical, C1-6Alkyl radical C3-6Cycloalkyl or C3-6Heterocycloalkyl substitution, and C1-6Heteroalkyl group with C3-6Cycloalkyl or C3-6Heterocycloalkyl substitution, including but not limited to: methyl, ethyl, n-propyl, isopropyl, -CH2C(CH3)(CH3) (OH), cyclopropyl, cyclobutyl, propylmethylene, cyclopropylacyl, benzyloxy, cyclopropylalkenyl, trifluoromethyl, aminomethyl, hydroxymethyl, methoxy, methylacyl, methoxyacyl, methylsulfonyl, methylsulfinyl, ethoxy, acetyl, ethylsulfonyl, ethoxyacyl, dimethylamino, diethylamino, dimethylamino, and diethylamino; n (CH)3)2,NH(CH3),-CH2CF3,-CH2CH2CF3,-CH2CH2F,-CH2CH2S(=O)2CH3,-CH2CH2CN,
Figure BDA0000950558680000061
Figure BDA0000950558680000062
-CH2CH(OH)(CH3)2,-CH2CH(F)(CH3)2,-CH2CH2F,-CH2CF3,-CH2CH2CF3,-CH2CH2NH2,-CH2CH2OH,-CH2CH2OCH3,-CH2CH2CH2OCH3,-CH2CH2N(CH3)2,-S(=O)2CH3,-CH2CH2S(=O)2CH3
Figure BDA0000950558680000063
Figure BDA0000950558680000064
The term "pharmaceutically acceptable" as used herein is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salts" refers to salts of the compounds of the present invention, prepared from the compounds of the present invention found to have particular substituents, with relatively nontoxic acids or bases. When compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of a base in neat solution or in a suitable inert solvent. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of acid in neat solution or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, hydrogen sulfate, hydroiodic acid, phosphorous acid, and the like; and salts of organic acids including acids such as acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic, and the like; also included are salts of amino acids (e.g., arginine, etc.), and salts of organic acids such as glucuronic acid (see Berge et al, "Pharmaceutical salts", Journal of Pharmaceutical Science 66: 1-19 (1977)). Certain specific compounds of the invention contain both basic and acidic functionalities and can thus be converted to any base or acid addition salt.
Preferably, the neutral form of the compound is regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of the compound differs from the various salt forms by certain physical properties, such as solubility in polar solvents.
As used herein, "pharmaceutically acceptable salts" belong to derivatives of the compounds of the present invention, wherein the parent compound is modified by forming a salt with an acid or a salt with a base. Examples of pharmaceutically acceptable salts include, but are not limited to: inorganic or organic acid salts of bases such as amines, alkali metal or organic salts of acid groups such as carboxylic acids, and the like. Pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound, for example, salts formed with non-toxic inorganic or organic acids. Conventional non-toxic salts include, but are not limited to, those derived from inorganic or organic acids selected from the group consisting of 2-acetoxybenzoic acid, 2-hydroxyethanesulfonic acid, acetic acid, ascorbic acid, benzenesulfonic acid, benzoic acid, bicarbonate, carbonic acid, citric acid, edetic acid, ethanedisulfonic acid, ethanesulfonic acid, fumaric acid, glucoheptose, gluconic acid, glutamic acid, glycolic acid, hydrobromic acid, hydrochloric acid, hydroiodide, hydroxyl, hydroxynaphthalene, isethionic acid, lactic acid, lactose, dodecylsulfonic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, nitric acid, oxalic acid, pamoic acid, pantothenic acid, phenylacetic acid, phosphoric acid, polygalacturonic acid, propionic acid, salicylic acid, stearic acid, glycolic acid, succinic acid, sulfamic acid, sulfanilic acid, sulfuric acid, tannin, tartaric acid, and p-toluenesulfonic acid.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, which contains an acid or base, by conventional chemical methods. In general, such salts are prepared by the following method: prepared by reacting these compounds in free acid or base form with a stoichiometric amount of the appropriate base or acid, in water or an organic solvent or a mixture of the two. Generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
In addition to salt forms, the compounds provided herein also exist in prodrug forms. Prodrugs of the compounds described herein readily undergo chemical changes under physiological conditions to convert to the compounds of the present invention. In addition, prodrugs can be converted to the compounds of the present invention in an in vivo environment by chemical or biochemical means.
Certain compounds of the present invention may exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may have asymmetric carbon atoms (optical centers) or double bonds. Racemates, diastereomers, geometric isomers and individual isomers are all included within the scope of the present invention.
The illustrations of mesomeric, ambiscientific and scientific, or enantiomerically pure compounds herein are from Maehr, j.chem.ed.1985, 62: 114-120. In 1985, 62: 114-120. Unless otherwise indicated, the absolute configuration of a stereocenter is indicated by wedge bonds and dashed bonds. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, they include the E, Z geometric isomer unless otherwise specified. Likewise, all tautomeric forms are included within the scope of the invention.
The compounds of the present invention may exist in specific geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, as well as racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the present invention. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the present invention.
Optically active (R) -and (S) -isomers as well as D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one of the enantiomers of a compound of the invention is desired, it can be prepared by asymmetric synthesis or derivatization with a chiral auxiliary, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide the pure desired enantiomer. Alternatively, when the molecule contains a basic functional group (e.g., amino) or an acidic functional group (e.g., carboxyl), diastereomeric salts are formed with an appropriate optically active acid or base, followed by diastereomeric resolution by art-recognized resolution methods, and the pure enantiomers are recovered. Furthermore, separation of enantiomers and diastereomers is typically accomplished by using chromatography employing a chiral stationary phase, optionally in combination with chemical derivatization (e.g., carbamate formation from amines).
The compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the compounds may be labelled with radioactive isotopes, such as tritium (A), (B), (C3H) Iodine-125 (125I) Or C-14(14C) In that respect All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
The term "pharmaceutically acceptable carrier" refers to any formulation or carrier medium capable of delivering an effective amount of an active agent of the present invention, without interfering with the biological activity of the active agent, and without toxic side effects to the host or patient, and representative carriers include water, oils, vegetables and minerals, cream bases, lotion bases, ointment bases, and the like. These include suspending agents, viscosity enhancers, skin penetration enhancers, and the like. Their preparation is known to those skilled in the cosmetic or topical pharmaceutical field. For additional information on the carrier, reference may be made to Remington: the Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams & Wilkins (2005), The contents of which are incorporated herein by reference.
The term "excipient" generally refers to a carrier, diluent, and/or vehicle necessary to formulate an effective pharmaceutical composition.
The term "effective amount" or "therapeutically effective amount" with respect to a drug or pharmacologically active agent refers to a sufficient amount of the drug or agent that is non-toxic but achieves the desired effect. For oral dosage forms of the invention, an "effective amount" of one active agent in a composition is the amount required to achieve the desired effect when combined with another active agent in the composition. The determination of an effective amount varies from person to person, depending on the age and general condition of the recipient and also on the particular active substance, and an appropriate effective amount in an individual case can be determined by a person skilled in the art according to routine tests.
The terms "active ingredient," "therapeutic agent," "active substance," or "active agent" refer to a chemical entity that is effective in treating a target disorder, disease, or condition.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is replaced with a substituent, including deuterium and hydrogen variants, so long as the valency of the particular atom is normal and the substituted compound is stable. When the substituent is a keto group (i.e., ═ O), it means that two hydrogen atoms are substituted. The keto substitution does not occur on the aromatic group. The term "optionally substituted" means that it may or may not be substituted, and unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemical realizability.
When any variable (e.g., R) occurs more than one time in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 0-2R, the group may optionally be substituted with up to two R, and there are separate options for R in each case. Furthermore, combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
When a substituent's bond can cross-link two atoms on a ring, such substituent can be bonded to any atom on the ring. When no atom is indicated in the listed substituents for connecting to a compound included in the general chemical structure but not specifically mentioned, such substituent may be bonded through any atom thereof. Combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
Substituents for alkyl and heteroalkyl radicals (including those groups commonly referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) are generally referred to as "alkyl substituents" and may be selected from, but are not limited to, one or more of the following groups: -R ', -OR', -O, ═ NR ', -N-OR', -NR 'R ", -SR', halogen, -SiR 'R" R' ", oc (O) R ', -c (O) R', -CO2R’、-CONR’R”、-OC(O)NR’R”、-NR”C(O)R’、NR’C(O)NR”R”’、-NR”C(O)2R’、-NR””’-C(NR’R”R”’)=NR””、NR””C(NR’R”)=NR”’、-S(O)R’、-S(O)2R’、-S(O)2NR’R”、NR”SO2R’、-CN、-NO2、-N3、-CH(Ph)2And fluoro (C)1-C4) Alkyl, the number of substituents being 0 to (2m '+ 1), where m' is the total number of carbon atoms in such radicals. R ', R ", R ' and R ' are each independently preferably hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl (e.g. aryl substituted with 1 to 3 halogens), substituted or unsubstituted alkyl, alkoxy, thioalkoxy groups or aralkyl. When a compound of the invention includes more than one R group, for example, each R group is independently selected, as is each of the groups when more than one R ', R ", R'", R "" and R "" group is present. When R 'and R' are attached to the same nitrogenWhen atomic, they may be combined with the nitrogen atom to form a 5-, 6-or 7-membered ring. For example, -NR' R "is intended to include, but not limited to, 1-pyrrolidinyl and 4-morpholinyl. From the above discussion of substituents, those skilled in the art will appreciate that the term "alkyl" is intended to include groups consisting of carbon atoms bonded to non-hydrogen groups, such as haloalkyl (e.g., -CF)3、-CH2CF3) And acyl (e.g., -C (O) CH)3、-C(O)CF3、-C(O)CH2OCH3Etc.). Similar to the substituents described for the alkyl radicals, aryl and heteroaryl substituents are generally collectively referred to as "aryl substituents" and are selected, for example, from the group consisting of-R ', -OR ', -NR ' R ", -SR ', -halogen, -SiR ' R" R ' ", OC (O) R ', -C (O) R ', -CO2R ', -CONR ' R", -OC (O) NR ' R ", -NR" C (O) R ', NR ' C (O) NR "R '", -NR "C (O)2R ', -NR" "' -C (NR ' R" R ' ")" "", NR "" C (NR ' R ") -NR '", -S (O) R ', -S (O) ")2R’、-S(O)2NR’R”、NR”SO2R’、-CN、-NO2、-N3、-CH(Ph)2Fluorine (C)1-C4) Alkoxy and fluorine (C)1-C4) Alkyl, etc., the number of substituents being between 0 and the total number of open valences on the aromatic ring; wherein R ', R ", R'", R "" and R "" are independently preferably selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl. When a compound of the invention includes more than one R group, for example, each R group is independently selected, as are each of these groups when more than one R ', R ", R'", R "" and R "" groups are present.
Two substituents on adjacent atoms of the aryl or heteroaryl ring may be optionally substituted by a substituent of the formula-T-C (O) - (CRR ') q-U-, wherein T and U are independently selected from-NR-, -O-, CRR' -or a single bond, and q is an integer from 0 to 3. Alternatively, two substituents on adjacent atoms of the aryl or heteroaryl ring may be optionally substituted with a substituent of the formula-A (CH2) r B-, wherein A and B are independently selected from-CRR' -, -O-, and,-NR-、-S-、-S(O)-、S(O)2-、-S(O)2NR' -or a single bond, and r is an integer of 1 to 4. Optionally, one single bond on the new ring thus formed may be replaced by a double bond. Alternatively, two substituents on adjacent atoms of the aryl or heteroaryl ring may be optionally substituted with a substituent of the formula-A (CH2) r B-, wherein S and d are each independently an integer selected from 0 to 3, and X is-O-, -NR', -S-, -S (O)2-or-S (O)2NR' -. The substituents R, R ', R "and R'" are each independently preferably selected from hydrogen and substituted or unsubstituted (C)1-C6) An alkyl group.
Unless otherwise specified, the term "halogen" or "halogen" by itself or as part of another substituent means a fluorine, chlorine, bromine or iodine atom. Furthermore, the term "haloalkyl" is intended to include monohaloalkyl and polyhaloalkyl. For example, the term "halo (C)1-C4) Alkyl "is intended to include, but not be limited to, trifluoromethyl, 2, 2, 2-trifluoroethyl, 4-chlorobutyl, and 3-bromopropyl, and the like.
Examples of haloalkyl groups include, but are not limited to: trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl. "alkoxy" represents the above alkyl group having the specified number of carbon atoms attached through an oxygen bridge. C1-6Alkoxy radicals comprising C1、C2、C3、C4、C5And C6Alkoxy group of (2). Examples of alkoxy groups include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy and S-pentoxy. "cycloalkyl" includes saturated cyclic groups such as cyclopropyl, cyclobutyl, or cyclopentyl. 3-7 cycloalkyl radicals including C3、C4、C5、C6And C7A cycloalkyl group. "alkenyl" includes hydrocarbon chains in either a straight or branched configuration, wherein one or more carbon-carbon double bonds, such as ethenyl and propenyl, are present at any stable site along the chain.
The term "halo" or "halogen" refers to fluorine, chlorine, bromine and iodine.
The term "hetero atom" as used herein includes atoms other than carbon (C) and hydrogen (H), and includes, for example, oxygen (O), nitrogen (N), sulfur (S), silicon (Si), germanium (Ge), aluminum (Al), boron (B), and the like.
Unless otherwise indicated, the terms "hetero", "heteroatom" or "heteroradical" (i.e., a radical containing a heteroatom) include atoms other than carbon (C) and hydrogen (H), radicals also containing heteroatoms as described above. Examples of the organic compound include oxygen (O), nitrogen (N), sulfur (S), silicon (Si), germanium (Ge), aluminum (Al), boron (B), and the like, and optionally substituted compounds such as-C (═ O) N (h) -, -C (═ NH) -, -S (═ O)2N (h) -, or-S (═ O) N (h) -.
"Ring" means substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. The so-called ring includes fused rings. The number of atoms in the ring is generally defined as the number of ring members, for example, "5 to 7 membered ring" means 5 to 7 atoms arranged around the ring. Unless otherwise specified, the ring optionally contains 1-3 heteroatoms. Thus, "5 to 7 membered ring" includes, for example, phenylpyridine and piperidinyl; in another aspect, the term "5-to 7-membered heterocycloalkyl ring" includes pyridyl and piperidyl, but does not include phenyl. The term "ring" also includes ring systems containing at least one ring, each of which "ring" independently conforms to the above definition.
The term "heterocycle" or "heterocyclyl" means a stable monocyclic or bicyclic heterocyclic ring which may be saturated, partially unsaturated or unsaturated (aromatic), which contains carbon atoms and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S, wherein any of the above heterocyclic rings may be fused to a benzene ring to form a bicyclic ring. The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., NO and S (O) p). The nitrogen atom may be substituted or unsubstituted (i.e. N or NR, wherein R is H or other substituents already defined herein). The heterocyclic ring may be attached to any heteroatom or carbon pendant group to form a stable structure. The heterocyclic rings described herein may be substituted at the carbon or nitrogen position if the resulting compound is stable. The nitrogen atoms in the heterocycle are optionally quaternized. In a preferred embodiment, when the total number of S and O atoms in the heterocycle exceeds 1, these heteroatoms are not adjacent to each other. In another preferred embodiment, the total number of S and O atoms in the heterocycle does not exceed 1. As used herein, the term "aromatic heterocyclic group" or "heteroaryl" means a stable 5, 6, 7 membered monocyclic or bicyclic or 7, 8, 9 or 10 membered bicyclic heterocyclic group aromatic ring comprising carbon atoms and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S. The nitrogen atom may be substituted or unsubstituted (i.e. N or NR, wherein R is H or other substituents already defined herein). The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., NO and S (O) p). It is noted that the total number of S and O atoms on the heteroaromatic ring does not exceed 1. Bridged rings are also included in the definition of heterocyclic. Bridged rings are formed when one or more atoms (i.e., C, O, N or S) connect two non-adjacent carbon or nitrogen atoms. Preferred bridged rings include, but are not limited to: one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms and one carbon-nitrogen group. It is worth noting that a bridge always converts a single ring into a three ring. In bridged rings, ring substituents may also be present on the bridge.
Examples of heterocyclic compounds include, but are not limited to: acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzomercaptofuranyl, benzomercaptophenyl, benzoxazolyl, benzoxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4 aH-carbazolyl, carbolinyl, chromanyl, chromene, cinnolinyl decahydroquinolinyl, 2H, 6H-1, 5, 2-dithiazinyl, dihydrofuro [2, 3-b ] tetrahydrofuranyl, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatino, isobenzofuranyl, pyran, isoindolyl, indolyl, etc, Isoquinolyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolyl, oxadiazolyl, 1, 2, 3-oxadiazolyl, 1, 2, 4-oxadiazolyl, 1, 2, 5-oxadiazolyl, 1, 3, 4-oxadiazolyl, oxazolidinyl, oxazolyl, isoxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazine, phenothiazine, benzoxanthine, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, Pyrazolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1, 2, 5-thiadiazinyl, 1, 2, 3-thiadiazolyl, 1, 2, 4-thiadiazolyl, 1, 2, 5-thiadiazolyl, 1, 3, 4-thiadiazolyl, thianthrenyl, thiazolyl, isothiazolylthiothienyl, thienyl, thienooxazolyl, thienothiazolyl, thienoimidazolyl, thienyl, triazinyl, 1, 2, 3-triazolyl, 1, 2, 4-triazolyl, 1, 2, 5-triazolyl, 1, 3, 4-triazolyl, and xanthenyl. Fused ring and spiro compounds are also included.
Unless otherwise specified, the term "hydrocarbyl" or a subset thereof (e.g., alkyl, alkenyl, alkynyl, phenyl, etc.) by itself or as part of another substituent means a straight, branched, or cyclic hydrocarbon radical, or combination thereof, that may be fully saturated, mono-, di-, or poly-unsaturated, that may be mono-, di-, or poly-substituted, that may include divalent or polyvalent radicals, that has the specified number of carbon atoms (e.g., C)1-C10Representing 1 to 10 carbons). "hydrocarbyl" includes, but is not limited to, aliphatic hydrocarbyl including linear and cyclic, specifically including, but not limited to, alkyl, alkenyl, alkynyl, and aromatic hydrocarbyl including, but not limited to, 6-12 membered aromatic hydrocarbyl such as benzene, naphthalene, and the like. In some embodiments, the term "alkyl" denotes a straight or branched chain radical or a combination thereof, which may be fully saturated, mono or polyunsaturated, and may include divalent and polyvalent radicals. Examples of saturated hydrocarbon radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, isobutyl, cyclohexyl, (cyclohexyl) methyl, cyclopropylmethyl, and n-propylPentyl, n-hexyl, n-heptyl, n-octyl, etc. homologues or isomers of the radicals. Unsaturated alkyl groups have one or more double or triple bonds, examples of which include, but are not limited to, ethenyl, 2-propenyl, butenyl, crotyl, 2-isopentenyl, 2- (butadienyl), 2, 4-pentadienyl, 3- (1, 4-pentadienyl), ethynyl, 1-and 3-propynyl, 3-butynyl, and higher homologs and isomers.
Unless otherwise specified, the term "heterohydrocarbyl" or a subset thereof (such as heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, and the like) by itself or in combination with another term means a stable straight-chain, branched, or cyclic hydrocarbon radical, or combination thereof, consisting of a number of carbon atoms and at least one heteroatom. In some embodiments, the term "heteroalkyl," by itself or in combination with another term, means a stable straight-chain, branched-chain hydrocarbon radical, or combination thereof, having a number of carbon atoms and at least one heteroatom constituent. In one exemplary embodiment, the heteroatoms are selected from B, O, N and S, wherein the nitrogen and sulfur atoms are optionally oxidized and the nitrogen heteroatom is optionally quaternized. The heteroatoms B, O, N and S can be located at any internal position of the heterohydrocarbyl group (except where the hydrocarbyl group is attached to the rest of the molecule). Examples include, but are not limited to-CH2-CH2-O-CH3、-CH2-CH2-NH-CH3、-CH2-CH2-N(CH3)-CH3、-CH2-S-CH2-CH3、-CH2-CH2、-S(O)-CH3、-CH2-CH2-S(O)2-CH3、-CH=CH-O-CH3、-CH2-CH=N-OCH3and-CH ═ CH-N (CH)3)-CH3. Up to two heteroatoms may be consecutive, e.g. -CH2-NH-OCH3
The terms "alkoxy", "alkylamino" and "alkylthio" (or thioalkoxy) are used in the conventional sense to refer to those alkyl groups attached to the rest of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
Unless otherwise specified, the terms "cycloalkyl", "heterocycloalkyl", "cyclohydrocarbacyl" or their derivatives (such as aryl, heteroaryl, heteroaromato, cycloalkyl, heterocycloalkyl, cycloheteroalkenyl, cycloalkenyl, heterocycloalkenyl, cycloheteroalkenyl, cycloalkynyl, heterocycloalkynyl, cycloheteroalkynyl, and the like) by themselves or in combination with other terms mean cyclized "alkyl", "heteroalkyl" or "hydrocarbacyl", respectively. Furthermore, in the case of a heterohydrocarbyl or heterocycloalkyi (e.g., heteroalkyl, heterocycloalkyl), a heteroatom may occupy the position of the heterocycle attached to the rest of the molecule. Examples of cycloalkyl groups include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Non-limiting examples of heterocyclyl groups include 1- (1, 2, 5, 6-tetrahydropyridinyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran indol-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 1-piperazinyl, and 2-piperazinyl.
Unless otherwise specified, the term "aryl" means a polyunsaturated aromatic hydrocarbon substituent, which may be mono-, di-or poly-substituted, which may be monocyclic or polycyclic (preferably 1 to 3 rings), fused together or covalently linked. The term "heteroaryl" refers to an aryl (or ring) containing one to four heteroatoms. In one illustrative example, the heteroatom is selected from B, N, O and S, wherein the nitrogen and sulfur atoms are optionally oxidized and the nitrogen atom is optionally quaternized. The heteroaryl group may be attached to the rest of the molecule through a heteroatom. Non-limiting examples of aryl or heteroaryl include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 2-oxazolyl, 2-thiazolyl, 2-pyridyl, 4-pyridyl, and the like, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalyl, 5-quinoxalyl, 3-quinolyl, and 6-quinolyl. The substituents for any of the above aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below.
For simplicity, aryl when used in combination with other terms (e.g., aryloxy, arylthio, aralkyl) includes aryl and heteroaryl rings as defined above. Thus, the term "aralkyl" is intended to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl and the like), including those alkyl groups in which a carbon atom (e.g., methylene) has been replaced by, for example, an oxygen atom, such as phenoxymethyl, 2-pyridyloxymethyl 3- (1-naphthyloxy) propyl and the like.
The term "leaving group" refers to a functional group or atom that can be substituted by another functional group or atom through a substitution reaction (e.g., an affinity substitution reaction). For example, representative leaving groups include triflate; chlorine, bromine, iodine; sulfonate groups such as methanesulfonate, toluenesulfonate, p-bromobenzenesulfonate, p-toluenesulfonate and the like; acyloxy groups such as acetoxy, trifluoroacetyloxy, and the like.
The term "protecting group" includes, but is not limited to, "amino protecting group," hydroxyl protecting group, "or" thiol protecting group. The term "amino protecting group" refers to a protecting group suitable for use in preventing side reactions at the amino nitrogen position. Representative amino protecting groups include, but are not limited to: a formyl group; acyl, for example alkanoyl (such as acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl such as tert-butoxycarbonyl (Boc); arylmethoxycarbonyl groups such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl groups such as benzyl (Bn), trityl (Tr), 1-bis- (4' -methoxyphenyl) methyl; silyl groups, such as Trimethylsilyl (TMS) and t-butyldimethylsilyl (TBS), and the like. The term "hydroxy protecting group" refers to a protecting group suitable for use in preventing side reactions of a hydroxy group. Representative hydroxy protecting groups include, but are not limited to: alkyl groups such as methyl, ethyl and tert-butyl; acyl groups, such as alkanoyl (e.g., acetyl); arylmethyl groups such as benzyl (Bn), p-methoxybenzyl (PMB), 9-fluorenylmethyl (Fm) and diphenylmethyl (benzhydryl, DPM); silyl groups, such as Trimethylsilyl (TMS) and t-butyldimethylsilyl (TBS), and the like.
All solvents used in the present invention are commercially available and can be used without further purification. The reaction is generally carried out under inert nitrogen in an anhydrous solvent. Proton NMR data were recorded on a Bruker Avance III 400(400MHz) spectrometer with chemical shifts expressed as (ppm) at the low field of tetramethylsilane. Mass spectra were measured on an agilent 1200 series plus 6110(& 1956A). LC/MS or Shimadzu MS contain a DAD: SPD-M20A (LC) and Shimadzu Micromass2020 detector. The mass spectrometer was equipped with an electrospray ion source (ESI) operating in either positive or negative mode.
The invention employs the following abbreviations: aq represents water; DCM represents dichloromethane; PE represents petroleum ether; DMF represents N, N-dimethylformamide; DMSO represents dimethyl sulfoxide; EtOAc for ethyl acetate; EtOH stands for ethanol; MeOH represents methanol; cbz represents benzyloxycarbonyl, an amine protecting group; boc represents tert-butyloxycarbonyl, an amine protecting group; HOAc represents acetic acid; NaBH (OAc)3Represents sodium triacetoxyborohydride; r.t represents room temperature; THF represents tetrahydrofuran; boc2O represents di-tert-butyl dicarbonate; TFA represents trifluoroacetic acid; DIPEA for diisopropylethylamine; pd (dppf) Cl2Represents [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride; POCl3Represents phosphorus oxychloride; NaH represents sodium hydride; LAH represents lithium aluminum hydride; pd (OAc)2Represents palladium (II) acetate; pd2(dba)3Represents tris (dibenzylideneacetone) dipalladium; pd (PPh)3)4Represents tetrakis (triphenylphosphine) palladium; et (Et)3SiH represents triethylsilane; PPh3Represents triphenylphosphine; xantphos represents 4, 5-bis (diphenylphosphino) -9, 9-dimethyl; MeSO3H represents methanesulfonic acid; xphos represents 2-dicyclohexylphosphino-2 ', 4 ', 6 ' -triisopropylbiphenyl; lawson's reagent represents 2, 4-bis (4-methoxyphenyl) -1, 3-dithia-2, 4-diphosphane-2, 4-disulfide; NBS represents N-bromosuccinimide; t-BuOK represents potassium tert-butoxide.
The compound is made by hand or
Figure BDA0000950558680000131
Software commandThe name, commercial compound, is the supplier catalog name.
Using a sample prepared with a Shimadzu SIL-20A autosampler and a Japanese Shimadzu DAD: HPLC analysis was performed using an Shimadzu LC20AB system from SPD-M20A detector using an Xtimate C18(3M packing, 2.1X 300mm specification) column. Method 0-60AB — 6 min: the elution was started with 100% a (a is 0.0675% TFA in water) and ended with 60% B (B is 0.0625% TFA in MeCN) using a linear gradient, with the entire procedure being 4.2 min followed by 1 min of 60% B. The column was equilibrated to 100: 0 for an additional 0.8 min for a total run time of 6 min. Method for 10-80 AB-6 minutes: elution was started with 90% a (a is 0.0675% TFA in water) and ended with 80% B (B is 0.0625% TFA in acetonitrile) using a linear gradient, with the entire procedure being 4.2 min followed by 80% B for 1 min. The column was equilibrated to 90: 10 for an additional 0.8 minutes for a total run time of 6 minutes. The column temperature was 50 ℃ and the flow rate was 0.8 mL/min. The scanning wavelength of the diode array detector is 200-400 nm.
Thin Layer Chromatography (TLC) was performed on a Sanpont-group silica gel GF254, spots were detected by irradiation with a UV light lamp, and in some cases by other methods, in these cases iodine (about 1g iodine was added to 10g silica gel and mixed thoroughly), vanillin (about 1g vanillin dissolved in 100mL 10% H)2SO4Prepared in (r)), ninhydrin (available from Aldrich) or special color developer (mixed thoroughly (NH)4)6Mo7O24·4H2O、5g(NH4)2Ce(IV)(NO3)6、450mLH2O and 50mL concentrated H2SO4Prepared) thin layer plates were spread and the compounds were examined. Still, w.c. was used; kahn, m.; and Mitra, M.journal of Organic Chemistry, 1978, 43, 2923-. Common solvents for flash or thin layer chromatography are mixtures of dichloromethane/methanol, ethyl acetate/methanol and hexane/ethyl acetate.
On a Gilson-281Prep LC 322 system using a Gilson UV/VIS-156 detectorFor chromatographic analysis, the chromatographic column used was Agella Venusil ASB Prep C18, 5m, 150X 21.2 mm; phenomenex Gemini C18, 5m, 150x 30 mm; boston Symmetrix C18, 5m, 150x 30 mm; or Phenomenex Synergi C18, 4m, 150X 30 mm. Eluting the compound with a low gradient of acetonitrile/water containing 0.05% HCl, 0.25% HCOOH or 0.5% NH at a flow rate of about 25mL/min3·H2O, total run time 8-15 minutes.
The invention relates to a novel spiro aryl sulfone compound which is an inhibitor of ALK and mutants thereof and can be used for treating cancers and other diseases. The novel spiroarylsulfone compound shows better inhibitory activity to ALK, ALK mutant and EGFR mutant enzyme; meanwhile, in terms of in vivo efficacy, the activity of the compound is better than that of a reference compound LDK378 in an ALK-driven patient-derived non-small cell lung cancer cell line PDX model, so that the invention can provide more effective treatment for patients with diseases caused by ALK enzyme abnormality.
Detailed Description
In some embodiments, compounds having formula (I) may be prepared according to the synthetic methods described in scheme a.
Scheme A
Figure BDA0000950558680000141
And (3) treating the aryl sulfone compound (A1) and 2, 4, 5-trichloropyrimidine (A2) in a solvent such as THF (tetrahydrofuran) and DMF (DMF), and reacting in the presence of a base such as sodium hydride and potassium carbonate to obtain the pyrimidine aryl sulfone compound (A3). Compound (A3) can be reacted with arylamine a6 catalyzed by an acid (e.g., methanesulfonic acid) in a solvent (e.g., t-butanol) to provide compound a 7. A7 can be further reacted, e.g. by reductive amination, to give compounds of formula (I). The spirocyclic amine compound a4 can be purchased or can be prepared by scheme B. A4 can be prepared into compound A5 by substitution reaction in a solvent such as dimethylformamide, and A5 is reduced by hydrogenation to obtain arylamine compound A6.
Scheme B
Figure BDA0000950558680000142
Scheme B is a general synthetic method for spiro amine compound B5(a 4). Ketone B1 reacts with cyanoacetate to give spiro compound B2, which is hydrolyzed to give B3, then B4, which is reduced with a reducing agent such as lithium aluminum hydride to give B5 by reducing B4; b5 can be coupled with aryl chloride in scheme a to afford a 5.
Scheme C
Figure BDA0000950558680000151
Scheme C is a general synthetic method for preparing compounds of formula (I) wherein T1 is CH. Reacting C1 compound of aldehyde with methyl vinyl ketone under the action of alkali such as KOH to obtain ketone C2, hydrogenating to obtain C3, and esterifying with trifluoromethanesulfonic acid to obtain C4; then coupling with amino aryl or nitro aryl boric acid to obtain C5, and reducing to obtain a compound C6; then coupled with compound A4 to give compound C7(I) of formula I, or after removal of the protecting group, further reductively aminated to give compound C7(I) of formula I.
The invention will now be further described by way of example. The following examples are given for illustrative purposes only and are not intended to limit the invention thereto. The compounds of the present invention can be prepared by a number of methods known in the art of organic synthesis. Embodiments of the invention may be synthesized using the methods described below, as well as synthetic methods known in the art of organic synthetic chemistry, or by modified methods based thereon. Preferred methods include, but are not limited to, the methods described below.
Detailed Description
In order to illustrate the present invention in more detail, the following examples are given, but the scope of the present invention is not limited thereto.
Example 1
Compound 1: 5-chloro N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy-4- (9-methyl-3, 9-diazaspiro [5.5] undec-3-yl) phenyl) pyrimidine-2, 4-diamine
Figure BDA0000950558680000152
Example 1A
9-benzyl-2, 4-dioxo-3, 9-diazaspiro [5.5] undecane-1, 5-dinitrile
Figure BDA0000950558680000161
Ammonium acetate (2.04 g, 26.42 mmol, 0.10 equiv.) was added to a solution of cyanoacetate (90 g, 796 mmol, 3.00 equiv.) in methanol (100mL) at 5-8 ℃; 1-benzylpiperidine 4-one (50 g, 0.264 mol) was then added to the reaction mixture; then, aqueous ammonia (46.3 g, 370 mmol, 1.40 eq) was added to the reaction mixture at below 10 ℃ and the mixture was stirred at 0-5 ℃ for 1 hour. The reaction mixture was then warmed to 20 ℃ (room temperature) and stirred for 20 hours. LCMS showed product formation. Water (100mL) was added to the mixture and heated to 55 ℃. The pH was adjusted to 4 by the addition of concentrated HCl (12M) and the temperature was kept at no more than 70 ℃. Then, the reaction solution was cooled to 10 ℃, stirred for 30 minutes, and then filtered. The filter cake was washed with water and left to dry in air to give the title compound (66 g, 77% yield) as a white solid. LCMS (ESI) (0-30 AB): m/z: 323.0[ M +1].
Example 1B
1 '-benzyl-3, 7-diazaspiro [ bicyclo [3.3.1] nonane-9, 4' -piperidine ] -2, 4, 6, 8-tetraone
Figure BDA0000950558680000162
A mixture of example 1A (1.00g, 3.10 mmol, 1.00eq) in sulfuric acid (88%, 4mL) was stirred at 60 ℃ for 4 hours. Then, water (1.4mL) was added to the reaction solution, and the mixture was heated to 100 ℃ and stirred for 1 hour. Water (5mL) was added to the reaction mixture, cooled to 10 ℃ and stirred at 10 ℃ for 30 minutes, then filtered. The filter cake was washed with cold water (5mL) and dried to give the title compound (1.11 g, crude) as a white solid.1H NMR(400MHz,CDCl3):11.87(s,2H),9.56(br.s.,1H),7.47(s,5H),4.34(d,J=4.4Hz,2H),3.75(br.s.,1H),3.22(br.s.,4H),1.88(br.s.,4H).
Example 1C
9-benzyl-3, 9-diazaspiro [5.5] undecane-2, 4-dione
Figure BDA0000950558680000163
To a flask of example 1B (1.10 g, 3.22 mmol, 1.00eq) was added aqueous NaOH (5N, 5mL) at 15 ℃, and then the mixture was warmed to 70 ℃ and stirred for 4 hours. The mixture was cooled to 45 ℃ and then concentrated hydrochloric acid (12N, 1.5mL) was added slowly until the pH of the solution was around 7. The mixture was then heated to 70-75 deg.C, and concentrated HCl (12N, 1mL) was added dropwise to control the rate of carbon dioxide release until the pH was adjusted to about 3-4. The mixture was heated to 70-75 ℃ and reacted further for 1 hour. The resulting suspension was cooled to 10 ℃ and stirred for 0.5 h. The solid was filtered and washed with water (25 mL). The solid was dried to give the title compound (380 mg, 1.40 mmol, 45% over 2 steps) as a white solid.1H NMR(400MHz,CDCl3):10.89(s,1H),10.60(br.s,1H),7.57(br.s.,2H),7.44(br.s.,3H),4.28(br.s.,2H),3.11(br.s.,4H),2.76(br.s.,2H),2.42(br.s.,2H),2.00-1.48(m,4H).
Example 1D
3-benzyl-3, 9-diazaspiro [5.5] undecane
Figure BDA0000950558680000171
To a solution of example 1C (10.6 g, 38.92 mmol, 1.00eq) in tetrahydrofuran (120mL) was added lithium aluminium hydride (5.17 g, 136.22 mmol, 3.50 eq) at 0-10 ℃ and the mixture was stirred at 65 ℃ for 3 h. TLC showed the reaction was complete. The mixture was cooled to 10 ℃ and quenched by the addition of water (5.2mL), followed by the addition of aqueous sodium hydroxide (1N, 5.2 mL). The mixture was filtered and the filtrate was concentrated to give the title compoundThe material (7.40 g, 30.28 mmol, 77.81% yield) was a light yellow oil.1H NMR(400MHz,CDCl3):7.34-7.27(m,5H),3.52(s,2H),2.88-2.73(m,4H),2.46-2.35(m,4H),1.58-1.51(m,4H),1.48-1.39(m,4H).
Example 1E
Tert-butyl ester 3, 9-diazaspiro [5.5] undecane-3-carboxylic acid methyl ester
Figure BDA0000950558680000172
Example 1D (500mg, 2.05 mmol) and Boc2O (450mg, 2.06 mmol) in methanol was added triethylamine (311 mg, 3.08 mmol) and the mixture was stirred at 20-30 ℃ for 16 h, the reaction mixture was concentrated, the residue was diluted with ethyl acetate (20mL) and washed with water (15mL × 2) and brine (20mL), the organic layer was dried over anhydrous sodium sulfate, concentrated, the crude intermediate was dissolved in ethanol (15mL) and acetic acid (2mL), then palladium hydroxide/carbon (0.1g) was added and the mixture was reacted under hydrogen (50Psi) for 20 h, the mixture was filtered and the filtrate was concentrated to give the acetate salt of the title compound (320 mg, 1.26 mmol, 61.37% yield).
Example 1F
Tert-butyl ester 9- (3-methoxy-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylic acid methyl ester
Figure BDA0000950558680000173
Example 1D (150 mg, 0.589 mmol) was dissolved in 4.0mL DMSO and 4-fluoro-2-methoxy-1-nitrobenzene (131 mg, 0.766 mmol) and potassium carbonate (244 mg, 1.77 mmol) were added under nitrogen and the reaction stirred at 90 ℃ for 16 h, cooled to room temperature, diluted with 60mL dichloromethane and the organic phase washed with water (15mL × 3), saturated brine (15mL), dried over anhydrous sodium sulfate and concentrated to give the title compound (250 mg, yield: 62%) as a yellow oil.1H NMR(400MHz,CD3OD):,7.95(d,J=9.2Hz,1H),6.55(dd,J=9.6,2.4Hz,1H),6.48(d,J=2.4Hz,1H),3.95(s,3H),3.56-3.41(m,8H),1.72-1.65(m,4H),1.58-1.50(m,4H),1.48(s,9H).
Example 1G
3- (3-methoxy-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane
Figure BDA0000950558680000181
Example 1F (0.7g, 1.73mmol) was dissolved in a mixed solution of 4mL of trifluoroacetic acid and 4mL of dichloromethane and stirred at 16 ℃ for 1 hour. LCMS showed reaction completion. The reaction mixture was extracted with 50mL of a saturated sodium carbonate solution and dichloromethane (50 mL. times.2). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound (0.7g, crude) as a yellow solid lcms (esi) (0-60 AB): m/z: 306.0[ M +1].
Example 1H
3- (3-methoxy-4-nitrophenyl) -9-methyl-3, 9-diazaspiro [5.5] undecane
Figure BDA0000950558680000182
Example 1G (0.7G, 2.3mmol) was dissolved in 10ml of tetrahydrofuran solution, 37% aqueous formaldehyde (207mg, 6.9mmol) was added at 16 ℃ and stirred for 0.5 h. Sodium borohydride (1.5g, 6.9mmol) was then added and stirring continued for 12 hours. LCMS showed reaction completion. The reaction was diluted with 60ml DCM, filtered and concentrated to give the title compound (0.6g, yield: 82%) as a yellow oil, LCMS (ESI) (0-60 AB): m/z: 320.2[ M +1].
Example 1I
2-methoxy-4- (9-methyl-3, 9-diazaspiro [5.5] undec-3-yl) aniline
Figure BDA0000950558680000183
To a solution of example 1H (0.6g, 1.9 mmol) in ethanol-water (12 ml) were added iron powder (1.1 g, 18.8 mmol) and ammonium chloride (1.1 g, 18.8 mmol). The mixture was stirred at 80 ℃ for 2 hours. TLC (dichloromethane: methanol 6: 1) showed the reaction was complete. The mixture was filtered and concentrated. The resulting residue was purified by preparative TLC (dichloromethane: methanol ═ 6: 1) to give the title compound (400mg, 74% yield) as a green solid 1H NMR (400MHz, CD3 OD): 6.72(d, J ═ 8.4Hz, 1H), 6.64(d, J ═ 2.0Hz, 1H), 6.51(d, J ═ 8.4Hz, 1H), 3.86(s, 3H), 3.08-2.96(m, 4H), 2.58-2.45(m, 4H), 2.32(s, 3H), 1.75-1.52(m, 8H).
Compound 1
5-chloro N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy-4- (9-methyl-3, 9-diazaspiro [5.5] undec-3-yl) phenyl) pyrimidine-2, 4-diamine
Figure BDA0000950558680000191
Example 1I (200mg, 0.69mmol) and 2, 5-dichloro-N- (2- (isopropylsulfonyl) phenyl) pyrimidin-4-amine (263mg, 0.76mmol) were added to 5ml of tert-butanol, and methanesulfonic acid (399mg, 4.15mmol) was added with stirring. The reaction mixture was stirred at 100 ℃ for 12 hours. LCMS showed reaction completion. The reaction mixture was purified by preparative HPLC (acidic method) to give the title compound (221mg, yield: 53%) as a colorless oil. 1H NMR (400MHz, CD3 OD): 8.35(s, 1H), 8.11(m, 1H), 8.07(d, J ═ 8.0Hz, 1H), 7.89(t, J ═ 8.0Hz, 1H), 7.73(d, J ═ 8.8Hz, 1H), 7.68(t, J ═ 8.0Hz, 1H), 7.60(m, 1H), 7.28(m, 1H), 4.03(s, 3H), 3.66-3.82(m, 4H), 3.38-3.52(m, 3H), 3.20-3.28(m, 2H), 2.94(s, 3H), 1.95-2.46(m, 6H), 1.78-1.89(m, 2H), 1.26(d, J ═ 6.8, 6H), 5-95 (AB), (5-95 Hz): m/z: 599.2[ M +1].
Example 2
Compound 2: 5-chloro-N2- (2- (difluoromethoxy) -4- (9-methyl-3, 9-diazaspiro [5.5] undec-3-yl) phenyl) -N4- (2- (isopropylsulfonyl) phenyl) pyrimidine-2-, 4-diamine
Figure BDA0000950558680000192
Example 2A
5-fluoro-2-nitrophenol
Figure BDA0000950558680000193
To a stirred solution of 4-fluoro-2-methoxynitrobenzene (3 g, 17.53 mmol) in dichloromethane (30mL) at 0 ℃ was added boron tribromide dropwise the reaction was stirred at 0 ℃ for 1.5 h TLC (petroleum ether: ethyl acetate 10: 1) showed the disappearance of 4-fluoro-2-methoxynitrobenzene, the solution was slowly added to ice water (100mL) and extracted with dichloromethane (50mL × 3), the organic phase was dried and concentrated to give the title compound (2.5g, yield 90.8%) as a yellow oil.1H NMR(400MHz,CDCl3):,10.80(s,1H),8.16(dd,J=9.6,5.6Hz,1H),6.84(dd,J=9.6,2.4Hz,1H),6.77-6.67(m,1H).
Example 2B
2- (difluoromethoxy) -4-fluoro-1-nitrobenzene
Figure BDA0000950558680000201
To a solution of example 11A (2.0 g, 12.73 mmol) in N, N-dimethylformamide (20mL) was added ClCF with constant stirring2COONa (6.9 g, 44.56 mmol) and sodium carbonate (1.62 g, 15.28 mmol), the reaction mixture was heated to 90 ℃ and stirred for 16 hours TLC (petroleum ether: ethyl acetate: 10: 1) showed 5-fluoro-2-nitrophenol to disappear, the reaction was diluted with ethyl acetate (100mL) and washed with water (20mL × 2), the organic layer was dried and concentrated to give the crude product, which was purified by silica gel column (petroleum ether: ethyl acetate: 10: 1) to give the title compound (1.4g, yield 53.1%) as a yellow oil.1H NMR(400MHz,CDCl3):,8.07-7.98(m,1H),7.17-7.05(m,2H),6.65(t,J=72.0Hz,1H).
Example 2C
3- (3- (difluoromethoxy) -4-nitrophenyl) -9-methyl-3, 9-diazaspiro [5.5] undecane
Figure BDA0000950558680000202
3-methyl-3, 9-diaza [5.5] undecane (1g, 5.94mmol), example 2B (1.48g, 7.13mmol) and potassium carbonate (2.46g, 17.82mmol) were added to 20mL of acetonitrile and the mixture was stirred at 90 ℃ for 16 h. TLC (dichloromethane: methanol 10: 1) showed the starting material was reacted. The mixture was filtered and concentrated, and the resulting crude product was separated by column chromatography (eluent: dichloromethane/methanol 50: 1, 20: 1) to give the title compound (1.9g, yield: 90%) as a yellow oil. 1H NMR (400MHz, CD3 OD): 8.03-8.06(m, 1H), 6.44-6.82(m, 3H), 3.39-3.43(m, 3H), 3.03-3.07(m, 1H), 2.43-2.49(m, 4H), 2.33(s, 3H), 1.61-1.65(m, 8H).
Example 2D
2- (difluoromethoxy) -4- (9-methyl-3, 9-diazaspiro [5.5] undec-3-yl) aniline
Figure BDA0000950558680000203
To a solution of example 2C (1.9g, 5.35mmol) in methanol (30mL) were added iron powder (2.1g, 32.08mmol) and ammonium chloride (1.72g, 32.08mmol), and the reaction mixture was stirred at 20 ℃ for 0.5 h. TLC (dichloromethane: methanol 10: 1) showed the reaction was complete. The mixture was filtered and concentrated, 30mL of saturated sodium carbonate solution were added and the aqueous phase was extracted with dichloromethane (50mL × 3). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound (1.5g, crude) as a brown oil. 1H NMR (400MHz, CD3 OD): 6.28-6.76(m, 4H), 3.00-3.03(m, 3H), 2.82-2.85(m, 1H), 2.43-2.46(m, 4H), 2.33(s, 3H), 1.57-1.62(m, 8H).
Example 2E
5-chloro-N2- (2- (difluoromethoxy) -4- (9-methyl-3, 9-diazaspiro [5.5] undec-3-yl) phenyl) -N4- (2- (isopropylsulfonyl) phenyl) pyrimidine-2-, 4-diamine
Figure BDA0000950558680000211
To a solution of example 2D (80mg, 0.25mmol) and 2, 5-dichloro-N- (2- (isopropylsulfonyl) phenyl) pyrimidin-4-amine (94mg, 0.27mmol) in tert-butanol (5mL) was added methanesulfonic acid (71mg, 0.74mmol) slowly and the reaction mixture was stirred at 100 ℃ for 12 h. LCMS showed reaction completion. The reaction mixture was purified by Pre-HPLC to give the title compound (71.01mg, yield: 11%) as a colorless oil. LCMS (ESI) (5-95 AB): m/z: 635.2[ M +1] RT: 1.774min/4min.
Example 3
Compound 3: 5-chloro N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy-4- (3-methyl-3-azaspiro [5.5] undec-9-yl) phenyl) pyrimidine-2, 4-diamine
Figure BDA0000950558680000212
Example 3A
4-Formylpiperidine-1-carboxylic acid tert-butyl ester
Figure BDA0000950558680000213
To a solution of dimethyl sulfoxide (4.37 g, 46.52 mmol) in dichloromethane (25mL) was added oxalyl chloride (5.9 g, 46.52 mmol) in dichloromethane (75mL) dropwise at-70 ℃ under nitrogen; a solution of tert-butyl 4- (hydroxymethyl) piperidine-1-carboxylate (5 g, 23.26 mmol) in dichloromethane (40mL) was then added dropwise to the mixture at-70 ℃. The reaction mixture was stirred at-70 ℃ for 15 minutes and triethylamine (11.76 g, 116.3 mmol) was added dropwise to the reaction mixture. After the addition was complete, the reaction mixture was stirred at-70 ℃ for 1 hour and warmed to 15 ℃. The reaction mixture was poured into water and extracted with dichloromethane (200 mL). The organic layer was washed with sodium bicarbonate solution (100mL), dried over anhydrous sodium sulfate, filtered and concentrated to dryness. The resulting oil is purified by column chromatography (petroleum ether: ethyl acetate 10: 1 to 1%3: 1) to give the title compound (2.4 g, 48% yield) as a yellow oil.1H NMR(400MHz,CDCl3):,9.68(s,1H),3.98-3.92(m,2H),2.96-2.91(m,2H),2.45-2.42(m,1H),1.89-1.60(m,2H),1.60-1.54(m,2H),1.47(s,9H).
Example 3B
9-oxo-3-azaspiro [5.5] undec-7-ene-3-carboxylic acid tert-butyl ester
Figure BDA0000950558680000221
But-3-en-2-one (0.658 g, 9.39 mmol) was added to a solution of example 3A (2 g, 9.39 mmol) in tetrahydrofuran (100 mL). The reaction mixture was cooled to-5 ℃ and a solution of potassium hydroxide-ethanol (3 mol/l, 1.57mL, 4.7 mmol) was added dropwise to the reaction mixture over 5 minutes. The reaction mixture was warmed to 15 ℃ and stirred for 16 hours. Petroleum ether (10mL) was added to the reaction mixture and the mixture was washed with brine (100 mL). The organic layer was concentrated to give the crude product which was purified by column chromatography on silica gel (petroleum ether: ethyl acetate ═ 10: 1 to 2: 1) to give the title compound (1.12 g, 45% yield) as a yellow oil.1H NMR(400MHz,CDCl3):,6.82(d,J=10Hz,1H),5.97(d,J=10Hz,1H),3.57-3.56(m,2H),2.50-2.47(m,2H),2.01-1.97(m,2H),1.67-1.65(m,2H),1.61-1.59(m,2H),1.49(s,9H).
Example 3C
9-oxo-3-azaspiro [5.5] undecane-3-carboxylic acid tert-butyl ester
Figure BDA0000950558680000222
To a solution of example 3B (5.00g, 18.84 mmol) in methanol (100mL) was added palladium on carbon (200mg, 1.88 mmol). The suspension was evacuated and replaced several times with hydrogen. The reaction mixture was maintained at 10-25 ℃ under hydrogen (18psi) and stirred for 5 hours. The reaction mixture was filtered and the filtrate was concentrated. The crude product was chromatographed on silica gel columnPurification (petroleum ether: ethyl acetate 2: 1) gave the title compound (4.78 g, 17.88 mmol, 94.9% yield) as a white solid.1H NMR(400MHz,CDCl3):,3.47-3.44(m,4H),2.38-2.35(m,4H),1.81-1.77(m,4H),1.58-1.56(m,4H),1.49(s,9H).
Example 3D
9- (((trifluoromethyl) sulfonyl) oxy) -3-azaspiro [5.5] undec-8-ene-3-carboxylic acid tert-butyl ester
Figure BDA0000950558680000223
Lithium diisopropylamide (2.5M, 1.22mL, 9mmol) was added dropwise to a solution of example 3C (2 g, 7.5 mmol) in tetrahydrofuran (20mL) at-78 deg.C under nitrogen, and after addition, the reaction mixture was stirred for 2 hours, then 1, 1, 1-trifluoro-N-phenyl-N- ((trifluoromethyl) sulfonyl) methanesulfonamide (2.67 g, 7.48 mmol, dissolved in 5mL of THF) was added dropwise to the reaction and stirred at-78 deg.C for 1.5 hours, the mixture was warmed to 10 deg.C and stirred for 2.5 hours, the reaction mixture was quenched with ammonium chloride solution (30mL) and extracted with ethyl acetate (50mL × 2. the organic layer was washed with brine (50mL), washed with Na (50mL), and extracted with Na (× mL)2SO4Drying and concentration gave a residue which was purified by silica gel column chromatography (petroleum ether: ethyl acetate 5: 1) to give the title compound (2.3g, 77% yield) as a brown oil.1H NMR(400MHz,CDCl3):,5.71(t,J=4.0Hz,1H),3.51-3.45(m,2H),3.39-3.32(m,2H),2.37-2.35(m,2H),2.16-2.09(m,2H),1.71-1.67(m,2H),1.48(s,9H).1.45-1.42(m,4H).
Example 3E
4-bromo-2-methoxyaniline
Figure BDA0000950558680000231
To a solution of 2-methoxyaniline (3 g, 24.36 mmol) in acetonitrile (30mL) was added N-bromosuccinimide (4.34 g, 24.36 mmol) and the reaction was mixedThe mixture was stirred at 15 ℃ for 15 minutes. The reaction mixture was quenched by adding sodium sulfite solution (40mL), and the mixture was extracted with ethyl acetate (50 mL); the organic layer was dried over sodium sulfate and concentrated to give the crude product which was purified by silica gel column chromatography (petroleum ether: ethyl acetate ═ 10: 1) to give the title compound (2.7 g, 55% yield) as a yellow solid.1H NMR(400MHz,CDCl3):,6.93-6.91(m,2H),6.62-6.59(dd,J=1.6,2.0Hz,1H),3.86(s,1H).
Example 3F
2-methoxy-4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) aniline
Figure BDA0000950558680000232
To a solution of example 3E (500mg, 2.47 mmol) in DMSO (5mL) was added bis pinacolato borate (628.4 mg, 2.47 mmol), tetrakistriphenylphosphine palladium (150 mg, 0.13 mmol) and potassium acetate (485 mg, 4.95 mmol), and the reaction mixture was stirred at 150 ℃ for 40 minutes under microwave. The reaction mixture was diluted with ethyl acetate (50mL) and water (30 mL). The organic layer was separated, dried and concentrated to give the crude product which was purified by silica gel column chromatography (petroleum ether: ethyl acetate ═ 10: 1) to give the title compound (160 mg, 26% yield) as a yellow oil.1H NMR(400MHz,CDCl3):,7.31(d,J=8.0Hz,1H),7.22(s,1H),6.71(d,J=8.0Hz,1H),3.91(s,3H),1.35(s,12H).
Example 3G
9- (4-amino-3-methoxyphenyl) -3-azaspiro [5.5] undec-8-ene-3-carboxylic acid tert-butyl ester
Figure BDA0000950558680000233
To a solution of example 27D (300mg, 0.75 mmol), example 3F (187 mg, 0.75 mmol) in dioxane (10mL) was added Pd (dppf) Cl2(15 mg, 0.075 mmol) and potassium carbonate (204 mg, 1.5 mmol),the reaction mixture was stirred at 110 ℃ for 16 hours. The reaction mixture was filtered through celite, and the filtrate was concentrated to give a crude product, which was purified by silica gel column chromatography (petroleum ether: ethyl acetate ═ 3: 1) to give the title compound (110mg, yield 39%) as a yellow oil. LCMS (ESI) (5-95 AB): m/z: 373.2[ M +1]].
Example 3H
9- (4-amino-3-methoxyphenyl) -3-azaspiro [5.5] undecane-3-carboxylic acid tert-butyl ester
Figure BDA0000950558680000241
In N2To a solution of example 3G (110mg, 0.3 mmol) in methanol (5mL) was added palladium on carbon (10 mg, 10%) under protection, and the reaction mixture was stirred under hydrogen (15PSI) at 16 ℃ for 5 h. The reaction mixture was filtered through celite and concentrated in vacuo to give the crude title compound (130 mg) as a yellow oil. LCMS (ESI) (5-95 AB): m/z: 319.1[ M-56+1].
Example 3I
5-chloro N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy-4- (3-azaspiro [5.5] undec-9-yl) phenyl) pyrimidine-2, 4-diamine
Figure BDA0000950558680000242
Example 3H (1.20g, 3.20mmol) and 2, 5-dichloro-N- (2-isopropylphenylsulfone) pyrimidin-4-amine (1.22g, 3.52mmol) were added to 20mL of tert-butanol under protection of N2, and methanesulfonic acid (1.54g, 16.02mmol) was slowly added dropwise. The reaction solution was stirred at 90 ℃ for 16 hours. LCMS showed reaction completion. The reaction mixture was poured into 10mL of saturated sodium carbonate solution and stirred for 10 minutes. Washing the obtained solid with water; the filter cake was collected and dried in vacuo to give the title compound (580mg, 0.95mmol, yield: 29.79%, purity: 96%) as a white solid. LCMS (ESI) (5-95 AB): m/z: 584.2[ M +1].
Compound 3
5-chloro N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy-4- (3-methyl-3-azaspiro [5.5] undec-9-yl) phenyl) pyrimidine-2, 4-diamine
Figure BDA0000950558680000243
Example 3I (180mg, 0.308mmol) was dissolved in 6mL MeOH, HCHO solution (46.27mg, 1.54mmol) was added in one portion, the reaction mixture was stirred at 25 ℃ for 0.5h, then NaBH3CN (97mg, 1.54mmol) was added. The reaction mixture was stirred for an additional 2 hours. The reaction mixture was diluted with water (15mL) and extracted with DCM (20mL × 2); the organic layer was dried and concentrated, and the resulting crude product was purified by Pre-HPLC (acidic method) to give the title compound (162mg, 0.25mmol, yield: 82.84%) as a yellow solid.
Example 4
Compound 4: 5-chloro-N2- (2-isopropoxy-5-methyl-4- (3-methyl-3-azaspiro [5.5] undec-9-yl) phenyl) -N4- (2- (isopropylsulfonyl) phenyl) pyrimidine-2-, 4-diamine
Figure BDA0000950558680000251
Example 4A
1-chloro-5-isopropoxy-2-methyl-4-nitrobenzene
2-
Figure BDA0000950558680000252
1-chloro-5-fluoro-2-methyl-4-nitrobenzene (5.40g, 28.49mmol) was dissolved in 60ml isopropanol and cesium carbonate (46.41g, 142.45mol) was added. The reaction solution was stirred at 60 ℃ for 16 hours. TLC (20: 1 petroleum ether: ethyl acetate) showed the reaction was complete. The mixture was concentrated, and ethyl acetate (100mL) was added to the residue, which was washed with water (100mL) and saturated brine (100mL), respectively. The organic phase was dried over anhydrous sodium sulfate and concentrated to give 1-chloro-5-isopropoxy-2-methyl-4-nitrobenzene (6.50g, 28.30mmol, yield: 99.34%) as a yellow solid. 1H NMR (400MHz, CDCl 3): 7.71(s, 1H), 7.08(s, 1H), 4.66-4.57(m, 1H), 2.35(s, 3H), 1.40(d, J ═ 6.0Hz, 6H).
Example 4B
2- (5-isopropoxy-2-methyl-4-nitrophenyl) -4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolane
Figure BDA0000950558680000253
Example 4A (1.00g, 4.35mmol) was dissolved in 20ml of 1, 4-dioxane and bis-pinacolato borate (1.22g, 4.79mmol), PCy3(122.11mg, 0.44mmol), Pd2(dba)2(199.36mg, 0.22mmol) and potassium acetate (619.62mg, 6.31mmol) were added. The reaction mixture was stirred at 100 ℃ for 2.5 hours under protection of N2. TLC (petroleum ether: ethyl acetate 10: 1) showed the reaction was complete. The reaction solution was filtered and concentrated, and the resulting residue was separated by column chromatography (eluent: petroleum ether: ethyl acetate: 50: 1) to give the title compound (2.00g, crude) as a yellow oil. 1H NMR (400MHz, CDCl 3): 7.52(s, 1H), 7.45(s, 1H), 4.79-4.64(m, 1H), 2.49(s, 3H), 1.40-1.37(m, 6H), 1.36(s, 12H).
Example 4C
Tert-butyl-9- (5-isopropoxy-2-methyl-4-nitrophenyl) -3-azaspiro [5.5] undec-8-ene-3-carboxylic acid
Figure BDA0000950558680000261
To a solution of example 4B (600mg, 1.87mmol) in 1, 4-dioxane (10mL) were added example 3D (746.16mg, 1.87mmol), Pd (dppf) Cl2(136.69mg, 0.18mmol) and sodium carbonate (594.00mg, 5.60 mmol). The reaction was stirred at 100 ℃ for 16 h under the protection of N2. TLC (petroleum ether: ethyl acetate 10: 1) showed the reaction was complete. The mixture was filtered and concentrated. The residue was purified by column chromatography (eluent: petroleum ether: ethyl acetate ═ 20: 1) to give the title compound (580mg, yield: 69.77%) as a yellow oil. 1H NMR (400MHz, CDCl 3): 7.62(s, 1H), 6.75(s, 1H), 5.53(br.s., 1H), 4.67-4.59(m, 1H), 3.59-3.48(m, 2H), 3.43-3.33(m, 2H), 2.23-2.17(m, 5H), 2.10(br.s., 2H), 1.65(t, J ═ 6.4Hz, 2H), 1.49-1.46(m, 13H), 1.38(d, J ═ 6.0Hz, 6H).
Example 4D
9- (5-Isopropoxy-2-methyl-4-nitrophenyl) -3-methyl-3-azaspiro [5.5] undec-8-ene
Figure BDA0000950558680000262
The solution in example 4C (590 mg, 1.33 mmol) in a mixture of DCM (2ml) and TFA (2ml) was stirred at 25 ℃ for 1 h. The reaction mixture was concentrated to give a yellow oil. The oil was dissolved in MeOH (5mL) and aq. hcho (538.41 mg, 6.63 mmol) was added. The mixture was stirred at 25 ℃ for 1 hour. To the mixture was added sodium triacetoxyborohydride (843.58 mg, 3.98 mmol). The mixture was stirred at 25 ℃ for 16 hours. LCMS showed reaction completion. The mixture was concentrated, and water (10mL) was added to the residue and extracted with dichloromethane (10X 3). The organic layer was dried over sodium sulfate and concentrated. The residue was purified by column chromatography (DCM: methanol 50: 1) to give the title compound (369.00mg, 77.40% yield) as a yellow oil. 1H NMR (400MHz, CDCl 3): 7.62(s, 1H), 6.75(s, 1H), 5.53(br.s., 1H), 4.69-4.58(m, 1H), 2.79-2.55(m, 4H), 2.48(s, 3H), 2.28-2.16(m, 5H), 2.13-2.07(m, 2H), 1.73-1.61(m, 6H), 1.37-1.41(m, 6H).
Example 4E
2-isopropoxy-5-methyl-4- (3-methyl-3-azaspiro [5.5] undec-9-yl) aniline
Figure BDA0000950558680000271
Example 4D (369.00mg, 1.03mmol) was dissolved in 10mL of methanol and Pd/C (50.00mg) was added under protection of N2. The reaction mixture was evacuated and replaced with hydrogen several times. The reaction mixture was stirred under 50psi of hydrogen at 50 ℃ for 16 hours. LCMS showed the starting material was reacted. The reaction mixture was filtered and concentrated to give the title compound (300mg, 0.91mmol, yield: 88.13%) as a yellow solid. LCMS (ESI) (30-90 CD): m/z: 331.2[ M +1].
Example 4F
5-chloro-N2- (2-isopropoxy-5-methyl-4- (3-methyl-3-azaspiro [5.5] undec-9-yl) phenyl) -N4- (2- (isopropylsulfonyl) phenyl) pyrimidine-2-, 4-diamine
Figure BDA0000950558680000272
To a solution of example 4D (500.00mg, 1.51mmol) and 2, 5-dichloro-N- (2-isopropylphenylsulfone) pyrimidin-4-amine (523.78mg, 1.51mmol) in tert-butanol (10ml) was added dropwise methanesulfonic acid (872.38mg, 9.08mmol), and the reaction was stirred at 90 ℃ for 16 hours. LCMS showed reaction completion. The mixture was concentrated, and the resulting residue was isolated and purified by Pre-HPLC (acidic method) to give the title compound (271mg, yield 28.03%) as a brown solid. 1H NMR (400MHz, MeOD): 8.23(br.s., 2H), 8.01(dd, J ═ 8.0, 1.2Hz, 1H), 7.74(br.s., 1H), 7.56(t, J ═ 7.2Hz, 1H), 7.23(br.s., 1H), 6.98(s, 1H), 4.74-4.62(m, 1H), 3.46-3.32(m, 3H), 3.25-3.11(m, 2H), 2.89(s, 3H), 2.82-2.69(m, 1H), 2.45(d, J ═ 14.4Hz, 1H), 2.31(d, J ═ 13.6Hz, 1H), 2.18(br.s., 3H), 1.87-1.50(m, 10H), 1.31 (s., 1.5H) (lcm, 5-5): m/z: 640.3[ M +1].
Example 5
Compound 5: 9- (4- ((5-chloro-4- ((2- (isopropylsulfonyl) phenyl) amino) pyrimidin-2-yl) amino) -3-methoxyphenyl) -3, 9-diazaspiro [5.5] undec-2-one
Figure BDA0000950558680000281
Example 5A
9-benzyl-3, 9-diazaspiro [5.5] undec-2-ones
Figure BDA0000950558680000282
9-benzyl-3, 9-diazaspiro [5.5] undecane-2-, 4-dione (5.00g, 18.36mmol) was dissolved in tetrahydrofuran (80mL), lithium aluminum hydride (1.74g, 45.90mmol) was added slowly under protection of N2 and the temperature of the system was controlled below 55 ℃. The reaction solution was stirred at 55 ℃ for 1 hour. TLC showed the reaction was complete. The mixture was cooled to 25 ℃ and quenched by the addition of 2ml of water and sodium hydroxide solution (5mol/L, 2ml) slowly in that order, the solid was filtered off and the filtrate was concentrated in vacuo. The residue was purified by column chromatography (eluent: dichloromethane/methanol ═ 20/1) to give the title compound (450mg, yield: 7.8%, purity: 78%) as a white solid. 1HNMR (400MHz, DMSO-d 6): 1.31-1.47(m, 4H), 1.55(t, J ═ 6.0Hz, 2H), 2.00(s, 1H), 2.26-2.40(m, 4H), 3.11(t, J ═ 6Hz, 1H), 3.45(s, 2H), 7.21-7.34(m, 5H), 7.37(br.s., 1H), lcms (esi) (10-80 CD): m/z: 259.1[ M +1].
Example 5B
3, 9-diazaspiro [5.5] undec-2-ones
Figure BDA0000950558680000283
To a solution of example 5A (500mg, 1.94mmol) in ethanol (10mL) was added Pd-C (100mg) and AcOH (0.1mL) with the protection of N2. The reaction mixture was evacuated and replaced several times with H2. The reaction mixture was stirred under 50psi of hydrogen at 50 ℃ for 12 hours. The reaction mixture was filtered and concentrated to give the title compound (400mg, crude) as a white solid, which was used directly in the next reaction.
Example 5C
9- (3-methoxy-4-nitrophenyl) -3, 9-diazaspiro [5.5] undec-2-one
Figure BDA0000950558680000284
To a solution of example 5B (220mg, 1.31mmol) and 4-fluoro-2-methoxy-1-nitro-benzene (223.78mg, 1.31mmol) in DMA (10mL) was added N, N-diisopropylethylamine (169.31mg, 1.31mmol) and the reaction mixture was stirred at 90 ℃ for 6 h. TLC showed the reaction was complete. The reaction liquid was concentrated under reduced pressure, and the residue was separated by column chromatography (eluent: dichloromethane: methanol 50: 1, 10: 1) to give the title compound (280mg, yield: 63.58%, purity: 95%) as a yellow solid. 1HNMR (400MHz, CDCl 3): 1.69(t, J ═ 6.0Hz, 4H)1.78(t, J ═ 6.0Hz, 2H), 2.36(s, 2H), 3.34-3.45(m, 4H), 3.46-3.55(m, 2H), 3.96(s, 3H), 5.82(br.s., 1H), 6.32(d, J ═ 4.0Hz, 1H), 6.43(dd, J ═ 8.4Hz, 1H), 8.02(d, J ═ 8.0Hz, 1H)
Example 5D
9- (4-amino-3-methoxyphenyl) -3, 9-diazaspiro [5.5] undec-2-one
Figure BDA0000950558680000291
Example 5C (100mg, 0.31mmol) was dissolved in 5ml tetrahydrofuran solution and Pd-C (0.1g) was added under protection of N2. The reaction mixture was evacuated and replaced several times with hydrogen. The mixture was stirred under hydrogen (50psi) for 3 hours at 25 ℃. LCMS showed reaction complete. The reaction mixture was filtered, and the filtrate was concentrated to give the title compound (110mg, crude) as a white solid. LCMS (ESI) (0-60 CD): m/z: 290.1[ M +1].
Example 5E
9- (4- ((5-chloro-4- ((2- (isopropylsulfonyl) phenyl) amino) pyrimidin-2-yl) amino) -3-methoxyphenyl) -3, 9-diazaspiro [5.5] undec-2-one
Figure BDA0000950558680000292
To a solution of example 5D (120.00mg, 0.41mmol) and 2, 5-dichloro-N- (2-isopropylphenylsulfone) pyrimidin-4-amine (172.30mg, 0.5mmol) in 1, 4-dioxane (3mL) was added MeSO3H (119.57mg, 1.24 mmol). The reaction solution was stirred at 50 ℃ for 12 hours. LCMS showed reaction completion. The mixture was purified by Pre-HPLC (acidic method) to give the title compound (33.00mg, 0.05mmol, yield: 13.15%, purity: 99%) as a yellow solid. 1H NMR (400MHz, METHANOL-d 4): 1.24(d, J ═ 8.0Hz, 6H)1.90-2.16(m, 6H)2.47(br.s., 2H)3.37-3.45(m, 3H)3.71(br.s., 4H)4.01(s, 3H)7.21(d, J ═ 8.0Hz, 1H)7.53(br.s., 1H)7.62-7.75(m, 2H)7.86(t, J ═ 8.0Hz, 1H)8.04-8.12(m, 2H)8.34(s, 1H). lcms (esi) (0-60 AB): m/z: 599.2[ M +1].
Example 6
Compound 6: 9- (4- ((5-chloro-4- ((2- (isopropylsulfonyl) phenyl) amino) pyrimidin-2-yl) amino) -3-methoxyphenyl) -3-methyl-3, 9-diazaspiro [5.5] undecan-2-one
Figure BDA0000950558680000301
Example 6A
9- (3-methoxy-4-nitrophenyl) -3-methyl-3, 9-diazaspiro [5.5] undec-2-one
Figure BDA0000950558680000302
To a solution of example 5C (150.00mg, 0.47mmol) in DMF (5mL) was added NaH (15.03mg, 0.38mmol, 60%) and stirred at 0 ℃ for 0.5h, MeI (53.34mg, 0.38mmol, 1.20eq) was added at 0 ℃ and the reaction was warmed to 25 ℃ and stirred for 3 h. LCMS showed the starting material reacted completely. The reaction was quenched by slow addition of ice water and then extracted with DCM (15 ml. times.3). The combined organic phases were washed with saturated brine (10 ml. times.2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the title compound (110mg, yield: 61.82%, purity: 88%) as a yellow solid. LCMS (ESI) (5-95 AB): m/z: 356.1[ M +23] RT: 1.090min/2min.
Example 6B
9- (4-amino-3-methoxyphenyl) -3-methyl-3, 9-diazaspiro [5.5] undec-2-one
Figure BDA0000950558680000303
Example 6A (110.00mg, 0.33mmol, 1.00eq) was dissolved in 5.00ml tetrahydrofuran and Pd-C (100mg) was added under protection of N2. The reaction mixture was evacuated and replaced several times with hydrogen. The reaction was stirred at 25 ℃ under 15psi hydrogen pressure for 3 hours. LCMS showed complete reaction of starting material. The reaction mixture was filtered and concentrated to give the title compound (100mg, 0.29mmol, yield: 87.91%, purity: 88%) as a white solid, which was used directly in the next step.
Example 6C
9- (4- ((5-chloro-4- ((2- (isopropylsulfonyl) phenyl) amino) pyrimidin-2-yl) amino) -3-methoxyphenyl) -3-methyl-3, 9-diazaspiro [5.5] undecan-2-one
Figure BDA0000950558680000304
To a solution of example 6B (100mg, 0.33mmol) and 2, 5-dichloro-N- (2-isopropylphenylsulfone) -pyrimidin-4-amine (114.12mg, 0.33mmol) in 1, 4-dioxane (3.00ml) was added methanesulfonic acid (95.03mg, 0.99mmol) slowly. The mixture was stirred at 60 ℃ for 12 hours. LCMS showed reaction completion. The reaction solution was purified by Pre-HPLC (acidic method) to obtain the title compound (45mg, yield: 22.10%, purity: 97%) as a yellow solid. 1H NMR (400MHz, METHANOL-d 4): 1.23(s, 3H)1.24(s, 3H)2.05(m, 6H)2.53(br.s., 2H)2.95-3.05(m, 3H)3.41(dt, J ═ 12.0, 6.0Hz, 1H)3.50(t, J ═ 6.0Hz, 2H)3.72(br.s., 4H)4.01(s, 3H)7.28(d, J ═ 12Hz, 1H)7.60(s, 1H)7.65-7.71(m, 2H)7.87(t, J ═ 6.0Hz, 1H)8.05(d, J ═ 8.0Hz, 2H)8.36(s, 1H), (s esi) (0-60): m/z: 613.2[ M +1].
Biochemical experiments
Experimental Material
Enzyme: ALK wild type, ALK C1156Y and ALK L1196M were all purchased from Cama Biosciences (Japan), and EGFRT790M/L858R were purchased from Life technology (Madison, Wis.).
HTRF kit: substrates purchased from Cis-Bio International, including Eu-labeled TK1 antibody, XL665 and biotin-labeled TK1 polypeptide.
A detection instrument: envision (perkinelmer).
Experimental methods
Test compounds were diluted in 3-fold gradient to obtain 11 doses at final concentrations from 1uM to 0.017 nM.
10ul wild type ALK enzyme reaction mixture system: 0.5nM wild type ALK, 1uM biotin-TK1peptide, 30uM ATP. Reaction buffer: 50mM Hepes (pH7.5), 10mM MgCl2, 0.01mM NaV3VO4. the reaction plate is a whitet Proxiplate 384-Plus plate (Perkinelmer), and the reaction is carried out at room temperature for 90 minutes.
10ul ALK C1156Y enzyme reaction mixture system: 0.15nM ALK C1156Y, 1uM biotin-TK1peptide, 30uM ATP. Reaction buffer: 50mM Hepes (pH7.5), 10mM MgCl2, 0.01mM NaV3VO4. the reaction plate is white Proxiplate 384-Plus plate (Perkinelmer). The reaction was carried out at room temperature for 60 minutes.
10ul ALK L1196M enzyme reaction mixture system: 0.15nM ALK L1196M, 1uM biotin-TK1peptide, 30uM ATP. Reaction buffer: 50mM Hepes (pH7.5), 10mM MgCl2, 0.01mM NaV3VO4. the reaction plate is white Proxiplate 384-Plus plate (Perkinelmer), and the reaction is carried out at room temperature for 60 minutes.
10ul EGFR T790M/L858R enzyme reaction mixture System: 0.08nM EGFR T790M/L858R, 1 uMbitin-TK 1peptide, 20uM ATP. Reaction buffer: 50mM Hepes (pH7.5), 10mM MgCl2, 0.01mM NaV3VO4. the reaction plate is white Proxiplate 384-Plus plate (Perkinelmer). The reaction was carried out at room temperature for 60 minutes.
Detection reaction: 10ul of detection reagent was added to the reaction plate at a final concentration of 2nM for antibodies and 62.5nM for XL 665. Incubate at room temperature for 60 minutes. Envision read plate.
Data analysis
The readings were converted to inhibition (%) (Min-Ratio)/(Max-Min) × 100% by the following formula. IC50 data were measured by 4-parameter curve fitting (Model 205in XLFIT5, iDBS).
Cell experiments
Experimental Material
RPMI1640, fetal bovine serum, penicillin/streptomycin solution, all purchased from Life Technology (Madison, Wis.). Cell Titer-Glo luminescence Cell viability reagents were purchased from Promega (Madison, Wis.). Karpas299Cell line was purchased from European Collection of Cell Cultures (ECACC). Plate reading instrument: envision (perkinelmer).
Experimental methods
384 well plates, each of 2500 Karpas-299 cells, 45ul volume. The cells were incubated overnight at 37 ℃ in a CO2 incubator. Test compounds were diluted in 3-fold gradients to obtain 10 dose concentrations from 2.5mM to 0.127uM, in duplicate wells. The middle plate was filled with 49ul of medium per well. Transfer 1ul of compound from the gradient dilution compound plate to the middle plate and mix well. Then 5ul of liquid was taken from the middle plate and transferred to the cell plate. The cells were further cultured in a CO2 incubator for 72 hours. After 72 hours, 25ul of detection reagent was added. Incubate for 10 min at room temperature and Envision read the plate.
Data analysis
The reading was converted into an inhibition ratio (%)(Max-Sample)/(Max-Min)*100%. A parametric curve fit (Model 205in XLFIT5, iDBS) measured the IC50 data.
Data for ALK enzyme inhibition IC50, ALK L1196M enzyme inhibition IC50, ALK C1156Y enzyme inhibition IC50, EGFR T790M/L858R enzyme inhibition IC50, and ALK IC50 of Karpas-299 cells for the compounds of the invention are shown in table 1 below.
TABLE 1
Figure BDA0000950558680000321
In vivo efficacy study
The following in vivo pharmacodynamic data show that the compound of the invention exhibits slightly superior or comparable antitumor activity to that of the reference compound LDK378 in both the wild-type LU-01-0015 lung cancer patient-derived xenograft (PDX) model (BALB/c nude mice) and the wild-type LU-01-0319 lung cancer patient-derived xenograft (PDX) model (BALB/c nude mice). For example, representative Compound 2 in a wild-type LU-01-0319 Lung cancer patient-derived xenograft (PDX) model (BALB/c nude mice) was administered (10 mg/kg) 21 days later, with a tumor volume of about 370mm from the very beginning3Reduced to 222mm3While LDK378 is reduced to 268mm3
1. Antitumor in vivo drug effect experiment is carried out on xenograft tumor (PDX) from LU-01-0319 lung cancer patient and BALB/c nude mouse
Initially, a LU-01-0319 xenograft tumor model obtained from surgically excised clinical specimens and implanted in nude mice, defined as lot P0 (LU-01-0319-P0)). The next batch of tumor implantations from P0 was defined as batch P1 (LU-01-0319-P1). FP3 was recovered from P2 and the next batch was defined as batch FP4 from FP3 tumor implantation. About 2-3 weeks after tumor implantation, the tumor size reaches about 200-400mm3At that time, administration is started. Test compounds were administered orally once daily. Tumor size was measured twice weekly, in two dimensions with calipers, and volume was calculated using the following formula: v ═ 0.5a x b2, where a and b are the major and minor diameters of the tumor, respectively. The anti-tumor efficacy was determined by dividing the mean tumor gain volume of animals treated with the compound by the mean tumor gain volume of untreated animals. The results are shown in Table 2.
TABLE 2
Figure BDA0000950558680000331
2. In vivo drug efficacy experiments were performed on xenograft (PDX) BALB/c nude mice from patients with LU-01-0015 lung cancer implanted subcutaneously.
BALB/c nude mice, female, 6-8 weeks, weighing about 18-22 grams, were kept in a special pathogen-free environment in a single ventilated cage (5 mice per cage). All cages, bedding and water were sterilized prior to use. All animals were free to obtain a standard certified commercial laboratory diet. A total of 80 mice purchased from BK laboratory animal co, LTD, shanghai were used for hypertonicity. Each mouse was implanted subcutaneously in tumor tissue (20-30 cubic millimeters) in the right flank for tumor growth. The experiment was started when the average tumor volume reached about-160 cubic millimeters. The test compounds were administered orally, 10mg/kg or 20mg/kg daily. Tumor volume was measured every 3 days with a two-dimensional caliper, the volume being measured in cubic millimeters and calculated by the following formula: v ═ 0.5a × b2, where a and b are the major and minor diameters of the tumor, respectively. The anti-tumor efficacy was determined by dividing the mean tumor gain volume of animals treated with the compound by the mean tumor gain volume of untreated animals. The results are shown in Table 3.
TABLE 3
Figure BDA0000950558680000332
Note: 10mg/kg QD X1W, 5mg/kg QD X2W, PO, 10 μ l/g; 20mg/kg PO, 10. mu.l/g QD X21
The ALK inhibitor reported by the invention can be used for treating various cancers, including anaplastic large-cell lymphoma, non-small-cell lung cancer, diffuse large B-cell lymphoma, inflammatory myofibroblast tumors, neuroblastoma, thyroid undifferentiated carcinoma and rhabdomyosarcoma. An ALK inhibitor, either as a sole therapy or in combination with other chemotherapeutic agents.

Claims (12)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0002521253220000011
wherein the content of the first and second substances,
t1 is selected from N or C (R)01);
T2 is selected from-N (R)01)-;
R01Selected from H, F, Cl, Br, I, CN, OH, SH, NH2, or optionally substituted with 1, 2 or 3 halogen, hydroxy, amino and/or cyano groups: c1-6Alkyl radical, C1-6Heteroalkyl group, C3-6Cycloalkyl- (CH)2)0-3-and C3-6Heterocycloalkyl- (CH)2)0-3-, wherein said "hetero" represents 1, 2 or 3 heteroatoms or heteroatom groups selected from O, S, N, S (═ O)2Or S (═ O);
D1-D4 are independently selected from- (CR)1R2)1-3-;
R3Is selected from R03、OR03And SR03
R03Is selected from C1-4Alkyl radical, C1-4Haloalkyl and C3-5Cycloalkyl- (CH)2)0-3-;
Z is selected from N and C (R)4);
R5Is selected from C1-4An alkyl group;
R1、R2and R4Each independently selected from H, F, Cl, Br, I, CN, OH, SH, NH2, or optionally substituted with 1, 2 or 3 halogen, hydroxy and/or cyano groups: c1-6Alkyl radical, C1-6Heteroalkyl group, C3-6Cycloalkyl- (CH)2)0-3-and C3-6Heterocycloalkyl- (CH)2)0-3-, wherein said "hetero" represents 1, 2, or 3 heteroatoms selected from O, S and N.
2. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein said R01Selected from H, -CH3、-CF3、-CHF2、-CH2CH3、CH(CH3)2、-CH2CF3、-CH2CH2CF3、-CH2CH2F、-CH2CH2S(=O)2CH3、-CH2CH2CN、
Figure FDA0002521253220000012
Figure FDA0002521253220000013
-CH2CH(OH)(CH3)2、-CH2CH(F)(CH3)2
3. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein said R03Is selected from-CH3、-CF3、-CHF2、-CH2CH3、-CH(CH3)2、-CH2CF3、-CH2CH2CF3、-CH2CH2F and
Figure FDA0002521253220000014
4. a compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein R5Is selected from-CH (CH)3)2
5. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein said R1、R2And R4Are respectively and independently selected from H, F, Cl, Br, I and-CH3、-CF3、-CHF2、-CH2CH3、-CH(CH3)2、-CH2CF3、-CH2CH2CF3、-CH2CH2F、-CH2CH2CN、
Figure FDA0002521253220000021
-CH2CH(OH)(CH3)2、-CH2CH(F)(CH3)2and-CH2CH2F。
6. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein T is2Selected from-NH-, -N (Me) -.
7. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the spiro structural unit
Figure FDA0002521253220000022
Selected from:
Figure FDA0002521253220000023
Figure FDA0002521253220000024
8. the compound according to claim 1, or a pharmaceutically acceptable salt thereof, selected from:
Figure FDA0002521253220000025
9. a process for the preparation of a compound of formula (I) according to claim 1, wherein T is1Represents N or CH, T2Represents NH, the preparation route of which is shown in scheme A or C:
scheme A
Figure FDA0002521253220000031
Scheme C
Figure FDA0002521253220000032
Wherein PG is an amino protecting group and the other variables are as defined in the preceding claims.
10. The process for producing a compound according to claim 9, wherein PG as an amino-protecting group is selected from the group consisting of BOC, Bn and Cbz.
11. A process for the preparation of a compound according to claim 9, wherein a5 is prepared as shown in scheme B:
scheme B
Figure FDA0002521253220000041
12. Use of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of cancer associated with ALK and/or EGFR and mutations thereof, cancer treated in combination with ROS1, BRAF, c-MET, HER2, KRAS/MEK, PIK3CA, FDFR, DDR2 and/or VEGFR inhibitors, or cancer treated in combination with a cytotoxin.
CN201610179546.7A 2015-03-31 2016-03-25 Spirocyclic aryl sulfones as protein kinase inhibitors Active CN106008503B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2015101506763 2015-03-31
CN201510150676 2015-03-31

Publications (2)

Publication Number Publication Date
CN106008503A CN106008503A (en) 2016-10-12
CN106008503B true CN106008503B (en) 2020-09-01

Family

ID=57081958

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201610179546.7A Active CN106008503B (en) 2015-03-31 2016-03-25 Spirocyclic aryl sulfones as protein kinase inhibitors

Country Status (1)

Country Link
CN (1) CN106008503B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106699743B (en) 2015-11-05 2020-06-12 湖北生物医药产业技术研究院有限公司 Pyrimidine derivative and application thereof
WO2020259573A1 (en) * 2019-06-25 2020-12-30 南京明德新药研发有限公司 Seven-membered heterocyclic derivative acting as kras g12c mutant protein inhibitor
CN115521309A (en) * 2022-09-16 2022-12-27 思科(深圳)药物研发有限公司 Iruaoke impurity and preparation method thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1832929A (en) * 2003-08-15 2006-09-13 诺瓦提斯公司 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
CN101466375A (en) * 2006-05-08 2009-06-24 神经原公司 Substituted azaspiro derivatives
WO2009097405A2 (en) * 2008-01-30 2009-08-06 Neurogen Corporation Compounds and methods for preparing diazaspiro derivatives
CN101687822A (en) * 2007-07-06 2010-03-31 安斯泰来制药株式会社 Di(arylamino)aryl compound
CN102365276A (en) * 2009-03-25 2012-02-29 格吕伦塔尔有限公司 Substituted spiro-amide compounds

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1832929A (en) * 2003-08-15 2006-09-13 诺瓦提斯公司 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
CN101466375A (en) * 2006-05-08 2009-06-24 神经原公司 Substituted azaspiro derivatives
CN101687822A (en) * 2007-07-06 2010-03-31 安斯泰来制药株式会社 Di(arylamino)aryl compound
WO2009097405A2 (en) * 2008-01-30 2009-08-06 Neurogen Corporation Compounds and methods for preparing diazaspiro derivatives
CN102365276A (en) * 2009-03-25 2012-02-29 格吕伦塔尔有限公司 Substituted spiro-amide compounds

Also Published As

Publication number Publication date
CN106008503A (en) 2016-10-12

Similar Documents

Publication Publication Date Title
US10093656B2 (en) Fused-ring or tricyclic aryl pyrimidine compound used as kinase inhibitor
JP6726677B2 (en) Substituted 2-H-pyrazole derivatives as anticancer agents
TWI571471B (en) Spire aryl oxides and aryl phosphorous sulfides
KR101718645B1 (en) Cdc7 inhibitors
CN105330698B (en) Loop coil aryl phosphorous oxides and sulfide
CN109415341B (en) Benzotriazole-derived alpha, beta unsaturated amide compounds as TGF-beta R1 inhibitor
WO2018108064A1 (en) Spiro-aryl-phosphorus-oxygen compound as fourth generation of egfr kinase inhibitor
CN110382495B (en) CDK4/6 inhibitors
JP2020511524A (en) Quinazoline compounds for EGFR inhibition
EP3694330A1 (en) Indazolyl-spiro[2.2]pentane-carbonitrile derivatives as lrrk2 inhibitors, pharmaceutical compositions, and uses thereof
CN106008503B (en) Spirocyclic aryl sulfones as protein kinase inhibitors
JP7329510B2 (en) pyrazolopyridinone compound
JP2021504334A (en) Pyrazolopyridinone compound
CN113087671A (en) Cyano-substituted pyridine and cyano-substituted pyrimidine compounds, and preparation methods and applications thereof
TWI685494B (en) Fused-ring or tricyclic aryl pyridine compuond used as kinase inhibitor
JP2022515622A (en) Thienopyridinone compound
RU2806625C2 (en) Compounds based on pyrazolopyridinone
RU2806751C2 (en) Compounds based on pyrazolopyridinone
CN117402164A (en) Aryl-substituted nitrogen-containing heterocyclic compounds

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20161222

Address after: 250100 Ji'nan Industrial Road, Shandong, No. 243

Applicant after: Qilu Pharmaceutical Co., Ltd.

Address before: 210032 Jiangsu city of Nanjing province Nanjing high tech Development Zone of High Road No. 9 business office building room 218

Applicant before: NANJING MINGDE NEW DRUG RESEARCH AND DEVELOPMENT CO., LTD.

SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant