NITROGEN-CONTAINING AROMATIC HETEROCYCLIC COMPOUND

- Astellas Pharma Inc.

As a result of studies on compounds having FGFR inhibitory action, the present inventors have found that the nitrogen-containing aromatic heterocyclic compounds of the present invention have inhibitory action on FGFR1, FGFR2, and/or FGFR3, particularly, mutant FGFR3, and thus, the present invention has been accomplished. The nitrogen-containing aromatic heterocyclic compound of the present invention can be used as a therapeutic agent for various cancers related to FGFR1, FGFR2, and/or FGFR3, such as lung cancer and hormone therapy-resistant breast cancer, stomach cancer, triple negative breast cancer, endometrial cancer, bladder cancer, and glioblastoma, particularly as a prophylactic and/or therapeutic agent for mutant FGFR3-positive bladder cancer.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
TECHNICAL FIELD

The present invention relates to compounds useful as active ingredients in pharmaceutical compositions, particularly in pharmaceutical compositions for the treatment of mutant FGFR3-positive bladder cancer.

BACKGROUND ART

The signaling pathway induced by fibroblast growth factors (FGFs) and their receptors, fibroblast growth factor receptors (FGFRs), is one of signaling pathways having the most important functions in the course of development from early embryogenesis to the formation of various organs. There are 18 genes of FGF ligands and four FGFR genes (FGFR1 to FGFR4), which are expressed in various cells and involved in cell growth, differentiation, and survival. In recent years, the importance of FGF signaling in the pathogenesis of diverse tumor types has been reported, and clinical reagents that specifically target the FGFs or FGF receptors are being developed (Nature Reviews Cancer 2010; 10, 116-129, J. Med. Chem. 2011; 54, 7066-7083, AACR 2011, No. 1643 AstraZeneca).

As for FGFR1, it is reported that FGFR1 gene is amplified in lung cancer (in particular, squamous cell cancer) and hormone therapy-resistant breast cancer, and it is also reported that these cell lines exhibit FGFR1-dependent cell growth (Sci. Transl. Med. 2010; 2(62): 62ra93, Breast Cancer Res. 2007; 9(2): R23, Cancer Res. 2010, 70 (5), 2085-2094).

As for FGFR2, the gene amplification in stomach cancer and triple negative breast cancer and the activating mutation in endometrial cancer are reported (Laboratory Investigation 1998, 78(9); 1143-1153, Virchows Arch. 1997, 431; 383-389, J. Cancer Res. Clin. Oncol., 1993, 119, 265-272, AACR 2011, No. 1643 AstraZeneca, Oncogene 2010; 29, 2013-2023). These cancer cells have been also confirmed to exhibit FGFR2-dependent growth.

Further, FGFR3 exhibits activating gene mutation in about 50% of cases of bladder cancer. Bladder cancer is largely divided into three types: non-invasive, invasive, and metastatic types. There have been issues on them that although non-invasive bladder cancer has a high 5-year survival rate of 70% or above, it frequently recurs or partly progresses to invasive cancer, and that invasive or metastatic bladder cancer has a low 5-year survival rate of 50% or below. Current therapies for non-invasive bladder cancer with FGFR3 mutation are transurethral resection of bladder tumor (TUR-BT) and postoperative BCG therapy or intravesical instillation of chemotherapeutic agents. However, their recurrence-preventing effect remains unsatisfactory, and their adverse effects such as hematuria and irritable bladder have been at issue. Meanwhile, total cystectomy and the systemic administration of chemotherapeutic agents have been used for the treatment of invasive or metastatic bladder cancer. However, there are issues on their effectiveness, and adverse effects. Bladder cancer is known to be characterized in that part of the cancer cells sloughs off from bladder tissues into urine, and, based on this characteristic, urine cytology is used for the diagnostic of bladder cancer. It was recently reported that FGFR3 mutation can be detected using the sediments in urine (Biochem. Biophys. Res. Commun. Nov. 3, 2007; 362(4): 865-71). Based on the presence or absence of this FGFR3 mutation, patients with FGFR3 mutation-positive bladder cancer can be selected, and the creation of an FGFR3 selective inhibitor has been demanded.

It is also reported that fusion genes combining FGFR genes and TACC (Transforming Acidic Coiled-coil) genes (FGFR3-TACC3 and FGFR1-TACC1) are expressed in the tumor of some glioblastoma patients (Science, Sep. 7, 2012; 337(6099): 1231-5). According to this report, the forced expression of FGFR3-TACC3 and FGFR1-TACC1 in astrocytes led to transformation and this result showed the oncogenicity of these fusion genes. It was also shown that FGFR3-TACC3 is localized in mitotic spindle poles and induces kinase activity-dependent chromosomal aneuploidy. Further, treatment of FGFR3-TACC3-expressing cells with an FGFR inhibitor suppressed chromosomal aneuploidy, thereby suppressing the growth of the cells. Thus, it is suggested that FGFR inhibitors might be effective for the treatment of glioblastoma patients with FGFR-TACC fusion genes.

It is also reported that human bladder cancer cell lines RT112, RT4, and LUCC2 express FGFR3-TACC3 fusion gene and that human bladder cancer cell line SW780 also expresses FGFR3-BAIAP2L1 fusion gene (Hum Mol Genet., 2013 Feb. 15, 22(4), 795-803). According to this report, the anchorage-independent growth of these fusion genes has been confirmed as a result of their introduction into NIH3T3 cells. Given that the growth of the foregoing bladder cancer cell lines expressing these FGFR3 fusion genes is inhibited by FGFR inhibitors, the detection of the presence of the fusion genes can be useful to select patients who can be treated effectively with FGFR inhibitors.

It is reported that the compounds of formula (A) shown below exhibit inhibition of various kinases and are useful as therapeutic agents for cancer and vascular disorders including myocardial infarction (Patent Document 1). Table 2 of the document discloses the test results of inhibition of kinases Yes, VEGFR, EphB4, PDGFRβ, and FGFR1 by some of the compounds, which discloses that IC50 values for the FGFR1 inhibitory activity were higher than 1000 nM, showing that the activity was also lower than in the case of inhibition of the activity of the other kinases. Further, in the document, there is no specific disclosure about the compounds of formula (I) of the present invention described below.

(In this formula, each of A is CH, N, or the like; each of B is CH or the like; A1 is O, CR2, or the like; R0 is H or the like; A2 is NR, O, or the like; L1 is a bond, O, or the like; L2 is a bond, C1-C6 alkyl, or the like; R1 is a 3- to 6-membered heterocyclic ring or the like; and each of Re and Rf is H, C1-C6 alkyl, hydroxyalkyl, or the like. For the other symbols, refer to the publication.)

It is reported that the compounds of formula (B) shown below exhibit Abl inhibitory action and are useful against various cancers (Patent Document 2). However, in the document, there is no specific description about FGFR inhibitory action. Further, the compounds of formula (I) of the present invention described below have group (R1)p which differentiate the compounds in structure from the compounds of formula (B).

(In this formula, G is CH or the like; A is 3-hydroxyphenyl or the like; and Y is vinyl or ethylene. For the other symbols, refer to the publication.)

It is reported that the compounds of formula (C) shown below have inhibitory action on various kinases including Src, VEGFR2, Yes, Fyn, Lck, Abl, PDGFR, EGFR, and RET and are usable for the treatment of cancer, vascular disorders, and the like (Patent Document 3). However, there is no disclosure about FGFR inhibitory action in the document. In the document, there is also no specific disclosure about the compounds of formula (I) of the present invention described below.

(In this formula, G1 is aryl optionally having a substituent, heteroaryl optionally having a substituent, or the like; L1 is O, SO, SO2, optionally substituted alkyl, or the like; L2 is optionally substituted alkyl, heterocyclic ring, or the like; A1 is a bond, O, C(Ra)2, or the like; and A2 is NRa, O, or the like. For the other symbols, refer to the publication.)

It is reported that the compounds of formula (D) shown below have TIE-2 and/or VEGFR-2 kinase inhibitory action and are useful in treatment of angiogenesis-related diseases including cancer (Patent Document 4). However, there is no specific description about FGFR inhibition in the document. Further, the compounds of formula (I) of the present invention described below differ in structure from the compounds of formula (D) in that the compounds of formula (I) have a group L1 having no amino group and that the compounds also have two bonds positioned para to each other on a ring comprising X and Y.

(In this formula, W is N or CR; R is H or the like. For the other symbols, refer to the publication.)

It is reported that the compounds of formula (E) shown below exhibit inhibitory action on the activity of many receptor protein tyrosine kinases, particularly, FGFRs, and can be used for the treatment of various diseases related to aberrant or excessive activity of these enzymes (Patent Document 5). However, the compounds of formula (I) of the present invention described below differ in structure from the compounds of formula (E) in that the compounds of formula (I) have a group L1 which does not represent a N atom and that the compounds also have two bonds positioned para to each other on a ring comprising X and Y.

(In this formula, two of X, Y, and Z are N and the third is CH or N. For the other symbols, refer to the publication.)

It is reported that the compounds of formula (F) shown below exhibit inhibitory action on various kinases and are useful against inflammation and autoimmune diseases (Patent Document 6). On the other hand, the compounds of formula (I) of the present invention described below differ in structure from the compounds of formula (F) in that the compounds of formula (I) have a group L1 which is not amide and that the compounds also have two bonds positioned para to each other on a ring comprising X and Y.

(In this formula, A1, A2, A3, and A4 are CR4, CR5, CR6, and CR7, respectively, or are N; L is —C(O)NR7—, —NR7C(O)—, or the like. For the other symbols, refer to the publication.)

It is reported that the compounds of formula (G) and those of formula (H) shown below exhibit FGFR inhibitory action and can be used for the treatment of various cancers (Patent Documents 7 and 8).

(In formula (G), ring B represents a 5- or 6-membered aromatic group that may comprise at least one heteroatom selected from O, S, and N. For the other symbols, refer to the publication.)

It is reported that the compounds of formula (J) shown below exhibit glucokinase activating effects and can be used for the treatment of diseases related to diabetes mellitus (Patent Document 9), and the structural feature is substitution with amino at the 2 position of the pyridine.

(For the symbols in this formula, refer to the publication.)

Also, the known compounds having the structures shown below are registered on the database as 1371065-79-0 and 1317903-92-6 in CAS registry number, respectively.

CITATION LIST Patent Documents

Patent Document 1: International Publication No. WO 2006/101977

Patent Document 2: International Publication No. WO 2007/056075

Patent Document 3: International Publication No. WO 2008/008234

Patent Document 4: International Publication No. WO 2003/066601

Patent Document 5: International Publication No. WO 2007/071752

Patent Document 6: International Publication No. WO 2007/022380

Patent Document 7: International Publication No. WO 2008/075068

Patent Document 8: International Publication No. WO 2009/153592

Patent Document 9: International Publication No. WO 2009/046784

SUMMARY OF INVENTION Technical Problem

The present invention provides compounds useful as active ingredients in pharmaceutical compositions, particularly in pharmaceutical compositions for the treatment of mutant FGFR3-positive bladder cancer.

Solution to Problem

As a result of intensive and extensive studies on compounds having FGFR inhibitory action, the present inventors have found that the nitrogen-containing aromatic heterocyclic compound of the present invention has inhibitory action on FGFR1, FGFR2, and FGFR3, particularly, good inhibitory action on mutant FGFR3. The present invention has been thus accomplished.

More specifically, the present invention relates to a compound of formula (I) or a salt thereof as well as to a pharmaceutical composition comprising a compound of formula (I) or a salt thereof and a pharmaceutically acceptable excipient.

(wherein
X and Y, the same or different from each other, are CH or N, with the proviso that X and Y are not N simultaneously;
L1 is -lower alkylene-, -lower alkylene-O—, —O-lower alkylene-, or -lower alkynylene-;

Z is N or CH;

R1, the same or different from one another, are lower alkyl optionally substituted with halogen, —O-(lower alkyl optionally substituted with halogen), halogen, cyano, or —N(lower alkyl)2;
p is an integer of 2 to 4;
ring W is an optionally substituted aromatic carbocyclic ring, an optionally substituted aromatic heterocyclic ring, or an optionally substituted non-aromatic heterocyclic ring;
Q is -L2-R2 or R3;
L2 is an optionally substituted aromatic heterocyclic ring or an optionally substituted non-aromatic heterocyclic ring;
R2 is a non-aromatic heterocyclic group optionally substituted with lower alkyl, optionally substituted cycloalkyl, lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH and —O-lower alkyl, —C(O)—R0, —C(O)-optionally substituted cycloalkyl, —NH—R0, —N(lower alkyl)-R0, -L3-optionally substituted non-aromatic heterocyclic group, or H;
R0 is lower alkyl optionally substituted with —OH;

R3 is

(1) lower alkyl optionally substituted with one or more groups selected from the group consisting of —C(O)OH, —OH, —O—R0, amino optionally substituted with one or two R0, carbamoyl optionally substituted with one or two R0, an optionally substituted aromatic heterocyclic group, an optionally substituted non-aromatic heterocyclic group, and a —C(O)-optionally substituted non-aromatic heterocyclic group;
(2) —O-(lower alkyl optionally substituted with one or more groups selected from the group consisting of —C(O)OH, —OH, —O—R0, carbamoyl optionally substituted with one or two R0, an optionally substituted non-aromatic heterocyclic group, and a —C(O)-optionally substituted non-aromatic heterocyclic group);
(3) —NH-(lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with lower alkyl, and carbamoyl optionally substituted with one or two R0);
(4) —N(lower alkyl)-(lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with lower alkyl, and carbamoyl optionally substituted with one or two R0);

(5) —C(O)OH;

(6) —C(O)-optionally substituted non-aromatic heterocyclic group;
(7) —O-(a non-aromatic heterocyclic group optionally substituted with lower alkyl); or
(8) carbamoyl optionally substituted with one or two R0; and
L3 is a bond, —NH—, —N(lower alkyl)-, or lower alkylene.)

Unless otherwise specified, when symbols used in one chemical formula herein are also used in another chemical formula, the same symbols have identical meanings.

The present invention also relates to a pharmaceutical composition that comprises a compound of formula (I) or a salt thereof and a pharmaceutically acceptable excipient and which is available for the treatment of various cancers related to FGFR1, FGFR2, and/or FGFR3, such as FGFR-related lung cancer and hormone therapy-resistant breast cancer, FGFR2-related stomach cancer, triple negative breast cancer, and endometrial cancer, and FGFR3-related bladder cancer and glioblastoma. It is to be noted that the pharmaceutical composition includes therapeutic agents for various cancers related to FGFR1, FGFR2, and/or FGFR3. One embodiment is a pharmaceutical composition for the treatment of FGFR3-related bladder cancer, which comprises a compound of formula (I) or a salt thereof and a pharmaceutically acceptable excipient. Another embodiment is a pharmaceutical composition for the treatment of mutant FGFR3-positive bladder cancer, which comprises a compound of formula (I) or a salt thereof and a pharmaceutically acceptable excipient. In the present specification, “mutant” includes point mutation, fusion mutation, deletion mutation and insertion mutation, and in an embodiment, “mutant” means general idea including point mutation and fusion mutation. In another embodiment, “mutant” means point mutation, and in yet another embodiment, “mutant” means fusion mutation.

Further, the present invention relates to: use of a compound of formula (I) or a salt thereof, for the manufacture of a pharmaceutical composition for the treatment of various cancers related to FGFR1, FGFR2, and/or FGFR3; use of a compound of formula (I) or a salt thereof, for the treatment of various cancers related to FGFR1, FGFR2, and/or FGFR3; a compound of formula (I) or a salt thereof, for the treatment of various cancers related to FGFR1, FGFR2, and/or FGFR3; and a method for treating various cancers related to FGFR1, FGFR2, and/or FGFR3, which comprises administering an effective amount of a compound of formula (I) or a salt thereof to a subject. The present invention also relates to: use of a compound of formula (I) or a salt thereof, for the manufacture of a pharmaceutical composition for the treatment of mutant FGFR3-positive bladder cancer; use of a compound of formula (I) or a salt thereof, for the treatment of mutant FGFR3-positive bladder cancer; a compound of formula (I) or a salt thereof, for the treatment of mutant FGFR3-positive bladder cancer; and a method for treating mutant FGFR3-positive bladder cancer, which comprises administering an effective amount of a compound of formula (I) or a salt thereof to a subject. It is to be noted that the “subject” referred to above is a human or another animal in need of the treatment, and is a human in need of the treatment in one embodiment.

Advantageous Effects of Invention

A compound of formula (I) or a salt thereof has inhibitory action on FGFR1, FGFR2, and/or FGFR3, particularly, mutant FGFR3, and can be used as a therapeutic agent for various cancers related to FGFR1, FGFR2, and/or FGFR3, such as lung cancer and hormone therapy-resistant breast cancer, stomach cancer, triple negative breast cancer, endometrial cancer, bladder cancer, and globlastoma, particularly as a therapeutic agent for mutant FGFR3-positive bladder cancer.

DESCRIPTION OF EMBODIMENTS

The present invention is described in detail below.

As used herein, the term “lower alkyl” refers to linear or branched alkyl having 1 to 8 carbon atoms (hereinafter abbreviated as C1-8) including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. Another embodiment is C1-4 alkyl, and yet another embodiment is methyl. Yet another embodiment is ethyl.

The term “lower alkylene” refers to linear or branched C1-8 alkylene including methylene, ethylene, trimethylene, tetramethylene, pentamethylene, hexamethylene, heptamethylene, octamethylene, propylene, methylmethylene, ethylethylene, 1,2-dimethylethylene, 1,1,2,2-tetramethylethylene, and the like. Another embodiment is C1-4 alkylene, and yet another embodiment is methylene. Yet another embodiment is ethylene.

The term “lower alkynylene” refers to linear or branched C2-6 alkynylene including ethynylene, propynylene, butynylene, pentinylene, hexynylene, 1,3-butadiynylene, 1,3-pentadiynylene, and the like. Another embodiment is C2-4 alkynylene, and yet another embodiment is ethynylene.

The term “cycloalkyl” refers to a C3-10 saturated hydrocarbon ring group and it may be bridged. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantyl, and the like. Another embodiment is C3-8 cycloalkyl, and yet another embodiment is C3-6 cycloalkyl. Yet another embodiment is cyclopropyl.

The term “aromatic carbocyclic ring” refers to a C6-14 monocyclic to tricyclic aromatic hydrocarbon ring. Examples include benzene, naphthalene, and anthracene, and another embodiment is benzene.

The term “aromatic heterocyclic ring” refers to a 5- to 10-membered aromatic heterocyclic ring which has 1 to 4 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur. Examples include pyridine, pyrrole, pyrazine, pyrimidine, pyridazine, imidazole, pyrazole, thiazole, oxazole, isoxazole, thiophene, isothiazole, furan, oxadiazole, thiadiazole, indole, isoindole, indazole, benzofuran, benzothiophene, benzimidazole, benzoxazole, benzothiazole, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, thienopyridine, thienopyrimidine, thienopyrazine, and the like. Another embodiment is pyridine, pyrrole, pyrazine, pyrimidine, pyridazine, imidazole, pyrazole, thiazole, oxazole, thiophene, furan, oxadiazole, and indazole. Yet another embodiment is pyridine, pyrimidine, imidazole, pyrazole, thiazole, and indazole. Yet another embodiment is pyridine, imidazole, and pyrazole. Yet another embodiment is pyridine. Yet another embodiment is pyrazole. Yet another embodiment is imidazole.

The term “aromatic heterocyclic group” refers to a monovalent group of the “aromatic heterocyclic ring” described above. Examples include pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, thienyl, furyl, 1,2,4-oxadiazolyl, and the like. Another embodiment is a 5- or 6-membered aromatic heterocyclic group which has 1 or 2 nitrogen atoms, and yet another embodiment is pyridyl.

The term “non-aromatic heterocyclic ring” refers to a 3- to 10-membered non-aromatic heterocyclic ring (or a 4- to 8-membered non-aromatic heterocyclic ring in one embodiment) having 1 to 4 heteroatoms which are selected from the group consisting of nitrogen, oxygen, and sulfur and which are the same or different. The non-aromatic heterocyclic ring may be fused to a benzene ring or a thiophene ring, be bridged by lower alkylene, be combined with another non-aromatic heterocyclic ring to form a spiro ring, or have an unsaturated bond on part of the own ring. The sulfur atom or nitrogen atom which is a ring-forming atom may be oxidized. Examples include aziridine, oxetane, azetidine, pyrrolidine, piperidine, azepane, diazepane, azocane, piperazine, 4-oxidopiperazine, homopiperazine, morpholine, oxazepane, thiomorpholine, 1,1-dioxidothiomorpholine, 1,1-dioxidotetrahydrothiopyran, 1,1-dioxidothiazolidine, thiazepane, 1-azabicyclo[2,2,2]octane, 7-oxabicyclo[2.2.1]heptane, 2,5-diazabicyclo[2.2.1]heptane, 3-azabicyclo[3.2.1]octane, 8-azabicyclo[3.2.1]octane, 9-azabicyclo[3.3.1]nonane, 3,9-diazabicyclo[3.3.1]nonane, 3,9-diazaspiro[5.5]undecane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2-oxa-7-azaspiro[3.5]nonane, tetrahydropyran, tetrahydrofuran, dioxane, dioxolan, tetrahydrothiophene, tetrahydrothiopyran, tetrahydrothienopyridine, tetrahydrobenzoazepine, tetrahydrobenzodiazepine, dihydrobenzofuran, dihydrobenzothiophene, dihydrobenzopyran, dihydrobenzodioxane, benzodioxane, dihydropyran, dihydropyrrole, dihydropyridine, tetrahydropyridine, tetrahydropyrazine, and the like. Another embodiment is a 5- to 7-membered non-aromatic heterocyclic ring having 1 or 2 heteroatoms which are selected from the group consisting of nitrogen, oxygen, and sulfur and which are the same or different. Yet another embodiment is a 5- to 7-membered nitrogen-containing non-aromatic heterocyclic ring which may have at least one nitrogen atom and have one additional heteroatom selected from the group consisting of nitrogen, oxygen, and sulfur. Yet another embodiment is a 6-membered nitrogen-containing non-aromatic heterocyclic ring. Examples include piperazine, piperidine, morpholine, thiomorpholine, 1,1-dioxidothiomorpholine, and the like. Yet another embodiment is oxetane, piperidine, piperazine, morpholine, thiomorpholine, 4-oxidopiperazine, 1,1-dioxidothiomorpholine, tetrahydropyran, tetrahydrofuran, tetrahydrothiophene, tetrahydropyridine, 1-azabicyclo[2.2.2]octane, 8-azabicyclo[3.2.1]octane, 3,9-diazaspiro[5.5]undecane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, or 2-oxa-7-azaspiro[3.5]nonane. Yet another embodiment is morpholine, piperidine, piperazine, 4-oxidopiperazine, 3,9-diazaspiro[5.5]undecane, or 2,6-diazaspiro[3.3]heptane. Yet another embodiment is piperidine. Yet another embodiment is piperazine.

The term “non-aromatic heterocyclic group” refers to a monovalent group of a non-aromatic heterocyclic ring. The non-aromatic heterocyclic group is a 3- to 10-membered non-aromatic heterocyclic group having 1 to 4 heteroatoms which are selected from the group consisting of nitrogen, oxygen, and sulfur and which are the same or different. The non-aromatic heterocyclic group may be bridged by lower alkylene, have an unsaturated bond on part of the ring, or be combined with another non-aromatic heterocyclic ring to form a spiro ring. The sulfur atom or nitrogen atom which is a ring-forming atom may be oxidized. Examples include aziridinyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, azepanyl, diazepanyl, azocanyl, piperazinyl, homopiperazinyl, morpholinyl, oxazepanyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl, thiazepanyl, tetrahydropyranyl, tetrahydrofuryl, dioxanyl, dioxolanyl, tetrahydrothienyl, tetrahydrothiopyranyl, 7-oxabicyclo[2.2.1]heptyl, 2,5-diazabicyclo[2.2.1]heptyl, 3-azabicyclo[3.2.1]octyl, 8-azabicyclo[3.2.1]octyl, 9-azabicyclo[3.3.1]nonyl, 3,9-diazabicyclo[3.3.1]nonyl, dihydropyranyl, dihydropyrrolyl, dihydropyridyl, tetrahydropyridyl, tetrahydropyrazyl, 9-diazaspiro[5.5]undec-3-yl, 1,9-diazaspiro[5.5]undec-9-yl, 2,8-diazaspiro[4.5]dec-8-yl, 1,4-dioxa-8-azaspiro[4.5]dec-8-yl, and the like. Another embodiment is a 5- to 7-membered non-aromatic heterocyclic group having 1 or 2 heteroatoms which are selected from the group consisting of nitrogen, oxygen, and sulfur and which are the same or different. Yet another embodiment is a 5- to 7-membered non-aromatic heterocyclic group having at least one nitrogen atom. Yet another embodiment is a 6-membered nitrogen-containing non-aromatic heterocyclic group. Examples include piperazinyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl, and the like. Yet another embodiment is oxetanyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, 4-oxidopiperazinyl, 1,1-dioxidothiomorpholinyl, tetrahydropyranyl, tetrahydrofuryl, tetrahydrothienyl, tetrahydropyridyl, 1-azabicyclo[2.2.2]octyl, 8-azabicyclo[3.2.1]octyl, 3,9-diazaspiro[5.5]undec-3-yl, 2,6-diazaspiro[3.3]hept-2-yl, or 2-oxa-6-azaspiro[3.3]hept-6-yl. Yet another embodiment is piperidinyl or piperazinyl. Yet another embodiment is piperidinyl. Yet another embodiment is piperazinyl.

The term “halogen” refers to —F, —Cl, —Br, or —I. Another embodiment is —F, and yet another embodiment is —Cl.

A compound of formula (I) or a salt thereof, wherein L1 in formula (I) is -lower alkylene-O—, means a compound of formula (II) or a salt thereof.

(In this formula, L4 represents lower alkylene. The same applies hereinafter.)

Further, two to four R1 in (R1)p may be the same or different from one another.

The phrase “optionally substituted” as used herein means “unsubstituted” or “having 1 to 5 substituents”. When a plurality of substituents are contained, these substituents may be the same or different from one another. Further, for example, two R0 on the nitrogen in the “carbamoyl optionally substituted with one or two R0” may be the same lower alkyl or different lower alkyl from each other. Each R0 may be substituted with —OH, or alternatively, either one may be substituted or neither one may be substituted.

As referred to herein, a substituent in “an optionally substituted aromatic carbocyclic ring”, “an optionally substituted aromatic heterocyclic ring”, or “an optionally substituted non-aromatic heterocyclic ring” as ring W in formula (I) is, for example, a group shown in group D1 described below.

Group D1 is a group consisting of:

  • (1) an aromatic heterocyclic group optionally substituted with one or more substituents selected from —OH and lower alkyl;
  • (2) a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from —OH and lower alkyl;
  • (3) halogens;
  • (4) —O-lower alkyl, —S-lower alkyl, —OH, and —SH;
  • (5) —CN and —NO2;
  • (6) —CO2H and —CO2-lower alkyl; and
  • (7) lower alkyl or —O-lower alkyl, each of which is optionally substituted with one or more groups selected from the group consisting of the groups shown in (1) to (6) above.

Another embodiment of group D1 is a group consisting of:

  • (1) an aromatic heterocyclic group optionally substituted with —OH;
  • (2) halogens;
  • (3) —OH;
  • (4) —CN;
  • (5) —CO2H; and
  • (6) lower alkyl or —O-lower alkyl, each of which is optionally substituted with one or more substituents selected from the group consisting of the substituents shown in (1) to (5) above.

Yet another embodiment of group D1 is a group consisting of:

  • (1) lower alkyl optionally substituted with halogen;
  • (2) —O-lower alkyl optionally substituted with an aromatic heterocyclic group optionally substituted with —OH;
  • (3) halogens; and
  • (4) —CN

Yet another embodiment of group D1 is a group consisting of lower alkyl optionally substituted with halogen; —O-(lower alkyl optionally substituted with one or more substituents selected from the group consisting of a non-aromatic heterocyclic group optionally substituted with oxo, an aromatic heterocyclic group optionally substituted with —OH, and halogens), halogens, cyano, and oxo.

A substituent acceptable in “an optionally substituted aromatic heterocyclic ring” or “an optionally substituted non-aromatic heterocyclic ring” referred to as L2 in formula (I), “optionally substituted cycloalkyl” or “an optionally substituted non-aromatic heterocyclic group” referred to as R2 in formula (I), and “an optionally substituted aromatic heterocyclic group” or “an optionally substituted non-aromatic heterocyclic group” referred to in R3 in formula (I) is, for example, a substituent selected from group D2.

Group D2 is a group consisting of:

  • (1) halogens;
  • (2) —OH and —SH;
  • (3) —CN; and
  • (4) lower alkyl optionally substituted with one or more substituents selected from the group consisting of the substituents shown in (1) to (3) above.

Another embodiment of group D2 is a group consisting of:

  • (1) lower alkyl optionally substituted with —OH; and
  • (2) —OH

Some embodiments of the compounds of formula (I) or salts thereof are given below.

(1) A compound or a salt thereof, wherein X is N and Y is CH. Another embodiment is a compound or a salt thereof, wherein X is CH and Y is N. Yet another embodiment is a compound or a salt thereof, wherein X is CH and Y is CH.

(2) A compound or a salt thereof, wherein L1 is lower alkylene or -lower alkylene-O—. Another embodiment is a compound or a salt thereof, wherein L1 is -lower alkylene-. Yet another embodiment is a compound or a salt thereof, wherein L1 is -lower alkylene-O—. Yet another embodiment is a compound or a salt thereof, wherein L1 is ethylene or -methylene-O—. Yet another embodiment is a compound or a salt thereof, wherein L1 is ethylene. Yet another embodiment is a compound or a salt thereof, wherein L1 is -methylene-O—. Yet another embodiment is a compound or a salt thereof, wherein L1 is ethynylene.

(3) A compound or a salt thereof, wherein Z is CH. Another embodiment is a compound or a salt thereof, wherein Z is N.

(4-1) A compound or a salt thereof, wherein p is 2 or 4. Another embodiment is a compound or a salt thereof, wherein p is 2. Yet another embodiment is a compound or a salt thereof, wherein p is 4.

(4-2) A compound or a salt thereof, wherein R1, the same or different from one another, are —O-lower alkyl or halogen. Another embodiment is a compound or a salt thereof, wherein R1, the same or different from one another, are —O-lower alkyl. Yet another embodiment is a compound or a salt thereof, wherein R1, the same or different from one another, are halogen. Yet another embodiment is a compound or a salt thereof, wherein R1, the same or different from one another, are —O-methyl or F. Yet another embodiment is a compound or a salt thereof, wherein R1, the same or different from one another, are —O-methyl or Cl. Yet another embodiment is a compound or a salt thereof, wherein all of R1 are F.

(5) A compound or a salt thereof, wherein the 6-membered aromatic ring in formula (I) which is substituted with (R1), and which has Z as a ring-forming atom is 2,6-dichloro-3,5-dimethoxyphenyl or 2,6-difluoro-3,5-dimethoxyphenyl. Another embodiment is a compound or a salt thereof, wherein the 6-membered aromatic ring in formula (I) which is substituted with (R1)p and which has Z as a ring-forming atom is 2,6-dichloro-3,5-dimethoxyphenyl. Another embodiment is a compound or a salt thereof, wherein the 6-membered aromatic ring in formula (I) which is substituted with (R1)p and which has Z as a ring-forming atom is 2,6-difluoro-3,5-dimethoxyphenyl.

(6) A compound or a salt thereof, wherein ring W is an aromatic carbocyclic ring optionally substituted with one or more substituents selected from group D1 or is an aromatic heterocyclic ring optionally substituted with one or more substituents selected from group D1. Another embodiment is a compound or a salt thereof, wherein ring W is a benzene ring substituted with one or more substituents selected from group D1 or is pyrazole, pyridine, pyrimidine, thiazole, indazole, or imidazole which in each case is optionally substituted with one or more substituents selected from group D1. Yet another embodiment is a compound or a salt thereof, wherein ring W is a benzene ring optionally substituted with one or more substituents selected from group D1 or is pyrazole optionally substituted with one or more substituents selected from group D1. Yet another embodiment is a compound or a salt thereof, wherein ring W is a benzene ring optionally substituted with one or more substituents selected from group D1. Yet another embodiment is a compound or a salt thereof, wherein ring W is a benzene ring optionally substituted with one or more substituents selected from the group consisting of lower alkyl, —O-lower alkyl, and halogens. Yet another embodiment is a compound or a salt thereof, wherein ring W is a benzene ring optionally substituted with one or more substituents selected from the group consisting of methyl, —O-methyl, and halogens. Yet another embodiment is a compound or a salt thereof, wherein ring W is a benzene ring optionally substituted with —O-methyl. Yet another embodiment is a compound or a salt thereof, wherein ring W is pyrazole optionally substituted with one or more substituents selected from group D1. Yet another embodiment is a compound or a salt thereof, wherein ring W is pyrazole optionally substituted with lower alkyl. Yet another embodiment is a compound or a salt thereof, wherein ring W is pyrazole optionally substituted with methyl. Yet another embodiment is a compound or a salt thereof, wherein ring W is pyrazole substituted with methyl. Yet another embodiment is a compound or a salt thereof, wherein ring W is pyrazole.

(7) A compound or a salt thereof, wherein Q is -L2-R2. Another embodiment is a compound or a salt thereof, wherein Q is R3.

(8) A compound or a salt thereof, wherein L2 is a non-aromatic heterocyclic ring optionally substituted with one or more substituents selected from group D2. Another embodiment is a compound or a salt thereof, wherein L2 is a nitrogen-containing non-aromatic heterocyclic ring optionally substituted with one or more substituents selected from group D2. Yet another embodiment is a compound or a salt thereof, wherein L2 is piperazine, 4-oxidopiperazine, piperidine, morpholine, azetidine, 3,9-diazaspiro[5.5]undecane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2-oxa-7-azaspiro[3.5]nonane, 8-azabicyclo[3.2.1]octane, or 1-azabicyclo[2.2.2]octane which in each case is optionally substituted with one or more substituents selected from group D2. Yet another embodiment is a compound or a salt thereof, wherein L2 is piperazine optionally substituted with one or more methyl, piperidine optionally substituted with one or more methyl, or 3,9-diazaspiro[5.5]undecane. Yet another embodiment is a compound or a salt thereof, wherein L2 is piperidine or 4-methylpyperazine.

(9) A compound or a salt thereof, wherein R2 is lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH and —O-lower alkyl, —NH-(lower alkyl optionally substituted with —OH), a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from group D2, -lower alkylene-(a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from the group D2), or H. Another embodiment is a compound or a salt thereof, wherein R2 is lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH and —O-lower alkyl, —NH-(lower alkyl optionally substituted with —OH), a non-aromatic heterocyclic group optionally substituted with lower alkyl (the lower alkyl is optionally substituted with —OH), or H. Yet another embodiment is a compound or a salt thereof, wherein R2 is piperazine optionally substituted with methyl, piperidine optionally substituted with methyl, 2-hydroxyethylamino, or H. Yet another embodiment is a compound or a salt thereof, wherein R2 is 4-methylpiperazine, 2-hydroxyethylamino, or H. Yet another embodiment is a compound or a salt thereof, wherein R2 is 4-methylpiperazine. Yet another embodiment is a compound or a salt thereof, wherein R2 is 2-hydroxyethylamino. Yet another embodiment is a compound or a salt thereof, wherein R2 is H.

(10) A compound or a salt thereof, wherein R3 is lower alkyl optionally substituted with one or more groups selected from the group consisting of —C(O)OH, carbamoyl optionally substituted with one or two R0, —OH, a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from group D2, and —C(O)-(a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from group D2) or wherein R3 is —O-(lower alkyl optionally substituted with one or more groups selected from the group consisting of —C(O)OH, carbamoyl optionally substituted with one or two R0, —OH, a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from group D2, and —C(O)-(a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from group D2)). Another embodiment is a compound or a salt thereof, wherein R3 is lower alkyl substituted with one or more groups selected from the group consisting of —C(O)OH, carbamoyl optionally substituted with one or two R0, —OH, a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from group D2, and —C(O)-(a non-aromatic heterocylic group optionally substituted with one or more substituents selected from group D2). Yet another embodiment is a compound or a salt thereof, wherein R3 is lower alkyl substituted with one or more substituents selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from —OH and lower alkyl, and —C(O)-(a non-aromatic heterocyclic group optionally substituted with one or more substituents selected from the group consisting of —OH and lower alkyl). Yet another embodiment is a compound or a salt thereof, wherein R3 is lower alkyl substituted with one or more substituents selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with lower alkyl, and —C(O)-(a non-aromatic heterocyclic group optionally substituted with —OH). Yet another embodiment is a compound or a salt thereof, wherein R3 is lower alkyl substituted with one or more groups selected from the group consisting of —OH, piperazinyl optionally substituted with methyl, and —C(O)-(azetidinyl optionally substituted with —OH). Yet another embodiment is a compound or a salt thereof, wherein R3 is 2-hydroxyethyl, 2,3-dihydroxypropyl, or 4-methylpiperazin-1-ylmethyl. Yet another embodiment is a compound or a salt thereof, wherein R3 is 4-methylpiperazin-1-ylmethyl. Yet another embodiment is a compound or a salt thereof, wherein R3 is lower alkyl optionally substituted with one or more —OH. Yet another embodiment is a compound or a salt thereof, wherein R3 is 2-hydroxyethyl or 2,3-dihydroxypropyl.

(11) A compound or a salt thereof, which is a consistent combination of any two or more of the embodiments described in (1) to (10) above.

The present invention encompasses a compound or a salt thereof, which is a combination of any two or more of the embodiments described in (1) to (10) above, as described in (11) above. Specific examples include the embodiments described below.

(12) A compound or a salt thereof, wherein X is N; Y is CH; and L1 is lower alkylene or -lower alkylene-O—.

(13) The compound according to (12) or a salt thereof, wherein Z is CH; R1, the same or different from one another, are —O-lower alkyl or halogen; p is 2 or 4; ring W is an optionally substituted aromatic carbocyclic ring or an optionally substituted aromatic heterocyclic ring.

(14) The compound according to (13) or a salt thereof, wherein L1 is ethylene or -methylene-O—; p is 4; ring W is an optionally substituted benzene ring or optionally substituted pyrazole.

(15) The compound according to any one of (12) to (14) or a salt thereof, wherein Q is -L2-R2; L2 is an optionally substituted non-aromatic heterocyclic ring; R2 is lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH and —O-lower alkyl, —NH-(lower alkyl optionally substituted with —OH), an optionally substituted non-aromatic heterocyclic group, -lower alkylene-(an optionally substituted non-aromatic heterocyclic group), or H.

(16) The compound according to (15) or a salt thereof, wherein p is 4; L2 is piperazine optionally substituted with one or more methyl, piperidine optionally substituted with one or more methyl, or 3,9-diazaspiro[5.5]undecane; R2 is piperazine optionally substituted with methyl, piperidine optionally substituted with methyl, 2-hydroxyethylamino, or H.

(17) The compound according to (16) or a salt thereof, wherein R1, the same or different from one another, are —O-methyl or F; L1 is -methylene-O—; ring W is a benzene ring optionally substituted with —O-methyl; L2 is piperidine or 4-methylpiperazine; R2 is 4-methylpiperazine, 2-hydroxyethylamino, or H.

(18) The compound according to any one of (12) to (14) or a salt thereof, wherein ring W is optionally substituted pyrazole; Q is R3; R3 is lower alkyl substituted with one or more groups selected from the group consisting of —C(O)OH, carbamoyl optionally substituted with one or two R0, —OH, an optionally substituted non-aromatic heterocyclic group, and —C(O)-(an optionally substituted non-aromatic heterocyclic group).

(19) The compound according to (18) or a salt thereof, wherein p is 4 and R3 is lower alkyl substituted with one or more substituents selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with lower alkyl, and —C(O)-(a non-aromatic heterocyclic group optionally substituted with —OH).

(20) The compound according to (19) or a salt thereof, wherein R1, the same or different from one another, are —O-methyl or F; L1 is -methylene-O—; ring W is pyrazole optionally substituted with methyl; R3 is 2-hydroxyethyl, 2,3-dihydroxypropyl, or 4-methylpiperazin-1-ylmethyl.

Another embodiment of the compound of formula (I) or salt thereof is, for example, a compound or a salt thereof, wherein

X and Y, the same or different from each other, are CH or N, with the proviso that X and Y are not N simultaneously;
L1 is -lower alkylene-, -lower alkylene-O—, —O-lower alkylene-, or lower alkynylene;

Z is N or CH;

R1, the same or different from one another, are lower alkyl optionally substituted with halogen, —O-(lower alkyl optionally substituted with halogen), halogen, cyano, or —N(lower alkyl)2;
p is an integer of 2 to 4;
ring W is an optionally substituted aromatic carbocyclic ring, an optionally substituted aromatic heterocyclic ring, or an optionally substituted non-aromatic heterocyclic ring;
Q is -L2-R2 or R3;
L2 is an optionally substituted aromatic heterocyclic ring or an optionally substituted non-aromatic heterocyclic ring;
R2 is lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH and —O-lower alkyl, —C(O)-optionally substituted cycloalkyl, —NH-(lower alkyl optionally substituted with —OH), an -L3-optionally substituted non-aromatic heterocyclic group, or H;
R3 is lower alkyl optionally substituted with one or more groups selected from the group consisting of —C(O)OH, —OH, —NH-lower alkyl, —N(lower alkyl)2, —C(O)—NH-lower alkyl, —C(O)—N(lower alkyl)2, an optionally substituted aromatic heterocyclic group, an optionally substituted non-aromatic heterocyclic group, and a —C(O)-optionally substituted non-aromatic heterocyclic group, —O-(lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH, —C(O)—NH-lower alkyl, and —C(O)—N(lower alkyl)2), —NH-(lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH, —C(O)—NH-lower alkyl, and —C(O)—N(lower alkyl)2), —N(lower alkyl)-(lower alkyl optionally substituted with one or more groups selected from the group consisting of —OH, —C(O)—NH-lower alkyl, and —C(O)—N(lower alkyl)2), —C(O)OH, or a —C(O)-optionally substituted non-aromatic heterocyclic group; and
L3 is a bond or lower alkylene.

Examples of specific compounds falling within the scope of the compound of formula (I) or a salt thereof include the following compounds:

  • 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine,
  • (2S)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol,
  • 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-{3-fluoro-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine,
  • 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(1-methylpiperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine,
  • 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(1-methylpiperidin-4-yl)piperazin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methyl-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{4-[(3R,5S)-3,5-dimethylpiperazin-1-yl]-3-methoxyphenyl}pyrimidin-2-amine,
  • N-[4-(3,9-diazaspiro[5.5]undec-3-yl)-3-methoxyphenyl]-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-amine,
  • 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethanol,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-[2-(4-methylpiperazin-1-yl)ethyl]-1H-pyrazol-4-yl}pyrimidin-2-amine,
  • 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]-1-(3-hydroxyazetidin-1-yl)ethanone,
  • (2R)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol,
  • 2-({1-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperidin-4-yl}amino)ethanol,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-methyl-5-[(4-methylpiperazin-1-yl)methyl]-1H-pyrazol-3-yl}pyrimidin-2-amine, and
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine, and salts thereof.

In another embodiment, examples of specific compounds falling within the scope of the compound of formula (I) or a salt thereof include the following compounds:

  • 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine,
  • (2S)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol,
  • 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-{3-fluoro-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine,
  • 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(1-methylpiperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine,
  • 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(1-methylpiperidin-4-yl)piperazin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methyl-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{4-[(3R,5S)-3,5-dimethylpiperazin-1-yl]-3-methoxyphenyl}pyrimidin-2-amine,
  • N-[4-(3,9-diazaspiro[5.5]undec-3-yl)-3-methoxyphenyl]-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-amine,
  • 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethanol,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-[2-(4-methylpiperazin-1-yl)ethyl]-1H-pyrazol-4-yl}pyrimidin-2-amine,
  • 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]-1-(3-hydroxyazetidin-1-yl)ethanone, and
  • (2R)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol, and salts thereof.

In yet another embodiment, examples of specific compounds falling within the scope of the compound of formula (I) or a salt thereof include the following compounds:

  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethanol,
  • (2R)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol,
  • 2-({1-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperidin-4-yl}amino)ethanol,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-methyl-5-[(4-methylpiperazin-1-yl)methyl]-1H-pyrazol-3-yl}pyrimidin-2-amine, and
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine, and salts thereof.

In yet another embodiment, examples of specific compounds falling within the scope of the compound of formula (I) or a salt thereof include the following compounds:

  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
  • 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethanol, and (2R)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol, and salts thereof.

In yet another embodiment, examples of specific compounds falling within the scope of the compound of formula (I) or a salt thereof include the following compounds:

  • 2-({1-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperidin-4-yl}amino)ethanol,
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-methyl-5-[(4-methylpiperazin-1-yl)methyl]-1H-pyrazol-3-yl}pyrimidin-2-amine, and
  • 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine, and salts thereof.

The compounds of formula (I) may have tautomers and/or geometrical isomers, depending on the type of substituents. Even when the compound of formula (I) appear herein only in one isomer form, the present invention encompasses the other isomers, and also encompasses separated isomers or mixtures thereof.

Further, since some compounds of formula (I) have an asymmetric carbon atom or axial asymmetry, optical isomers based on this asymmetry may also exist. The present invention also encompasses separated optical isomers of the compounds of formula (I) or mixtures thereof.

Furthermore, the present invention encompasses pharmaceutically acceptable prodrugs of the compounds represented by formula (I). The term “pharmaceutically acceptable prodrug” refers to a compound having a group that can be converted into an amino group, a hydroxyl group, a carboxyl group, or the like by solvolysis or under physiological conditions. Examples of a prodrug-forming group include those described in Prog. Med., 5, 2157-2161 (1985) and those described in “Development of Pharmaceuticals” (Hirokawa Publishing, 1990) vol. 7, Molecular Design, 163-198.

Likewise, salts of the compounds of formula (I) are pharmaceutically acceptable salts of the compounds of formula (I). The compounds of formula (I) may form acid addition salts or salts with bases, depending on the type of substituents. Specific examples include acid addition salts with inorganic acids (e.g., hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, phosphoric acid) or with organic acids (e.g., formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, mandelic acid, tartaric acid, dibenzoyltartaric acid, ditoluoyltartaric acid, citric acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, aspartic acid, glutamic acid), salts with inorganic bases (e.g., sodium, potassium, magnesium, calcium, aluminum) or with organic bases (e.g., methylamine, ethylamine, ethanolamine, lysine, ornithine), salts with various amino acids and amino acid derivatives (e.g., acetylleucine), ammonium salts, and the like.

Moreover, the present invention encompasses the compounds of formula (I) and salts thereof in the form of various hydrates, solvates, and crystalline polymorphic substances. The present invention also encompasses the compounds labeled with various radioactive or non-radioactive isotopes.

(Preparation Processes)

The compounds of formula (I) and salts thereof can be prepared by applying various known synthesis methods on the basis of characteristics derived from their basic structure or the type of their substituents. In some cases, depending on the type of functional group, it is technically effective to replace such a functional group with an appropriate protecting group (a group that can be easily converted into the original functional group) between the starting material stage and the intermediate stage. Examples of the protecting group include those described in Wuts (P. G. M. Wuts) and Greene (T. W. Greene), “Greene's Protective Groups in Organic Synthesis (fourth edition, 2006)”, and the like, which may be selected and used as appropriate, depending on reaction conditions. In such a method, after introduction of the protecting group and subsequent reaction, the protecting group may be removed, if needed, to obtain a desired compound.

Likewise, a prodrug of the compound of formula (I) can be prepared by introducing a specific group between the starting material stage and the intermediate stage, as in the case of the above protecting group, or by subjecting the obtained compound of formula (I) to further reaction. The reaction may be accomplished by applying esterification, amidation, dehydration, or the like, which is a method that is common and known to those skilled in the art.

Described below are typical processes for preparing the compounds of formula (I). Each process may also be accomplished by reference to the documents cited in this description. It should be noted that the preparation processes of the present invention are not limited to the examples illustrated below.

(Preparation Process 1)

(In this formula, L5 represents halogen, methylsulfinyl, or methylsulfonyl. The same applies hereinafter.)

The compound (I) of the present invention can be obtained by coupling reaction of compound (1a) and compound (2a).

In this reaction, compounds (1a) and (2a) are used in equal amounts or one of them is used in an excessive amount. A mixture of these compounds is stirred in the presence of a predetermined catalyst, in a solvent inert to the reaction or in the absence of a solvent, generally for 0.1 hour to 5 days under conditions between room temperature and heating to reflux. This reaction is preferably performed under an inert gas atmosphere. Examples of the solvent used in this process include, but are not particularly limited to, aromatic hydrocarbons (e.g., benzene, toluene, xylene), ethers (e.g., diethyl ether, tetrahydrofuran, dioxane, dimethoxyethane), halogenated hydrocarbons (e.g., dichloromethane, 1,2-dichloroethane, chloroform), N-methylpyrrolidone, N,N-dimethylformamide, N,N-dimethylacetamide, dimethyl sulfoxide, ethyl acetate, acetonitrile, tert-butanol, and mixtures thereof. Examples of the predetermined catalyst include palladium acetate, tris(dibenzylideneacetone)dipalladium, and the like. Further, when a palladium catalyst is used, a ligand used for the catalyst may be triphenylphosphine, 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphine), 2-(dicyclohexylphosphino)-2′,4′,6′-triisopropyl-1,1′-biphenyl, or 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene. The reaction may be performed in the presence of an organic base (e.g., triethylamine, N,N-diisopropylethylamine, or N-methylmorpholine) or an inorganic base (e.g., sodium tert-butoxide, potassium carbonate, sodium carbonate, cesium carbonate, or potassium hydroxide), because it is advantageous for smooth reaction in some cases. Heating the reaction mixture by microwave irradiation is advantageous for smooth reaction in some cases.

DOCUMENTS

  • S. R. Sandler and W. Karo, “Organic Functional Group Preparations”, second edition, vol. 1, Academic Press Inc., 1991
  • The Chemical Society of Japan (ed.), “The Fifth Series of Experimental Chemistry”, vol. 14, MARUZEN Co., Ltd., 2005

(Preparation Process 2)

(In this formula, L6 represents lower alkynylene. The same applies hereinafter.)

(Step 1)

This process is intended to prepare compound (I-1) of the present invention by Sonogashira coupling reaction of compound (1b) and a terminal alkyne derivative.

In this process, compound (1b) and a terminal alkyne derivative are used in equal amounts or one of them is used in an excessive amount. A mixture of these is stirred in the presence of a base, a palladium catalyst, and copper iodide, in a solvent inert to the reaction, generally for 0.1 hour to 5 days under conditions between room temperature and heating to reflux. This reaction is preferably performed under an inert gas atmosphere. Examples of the solvent used in this process include, but are not particularly limited to, aromatic hydrocarbons (e.g., benzene, toluene, xylene), ethers (e.g., diethyl ether, tetrahydrofuran, dioxane, dimethoxyethane), halogenated hydrocarbons (e.g., dichloromethane, 1,2-dichloroethane, or chloroform), alcohols (e.g., methanol, ethanol, 2-propanol, butanol), N,N-dimethylformamide, dimethyl sulfoxide, and mixtures thereof. The base is preferably an organic base (e.g., triethylamine, N,N-diisopropylethylamine, or N-methylmorpholine) or an inorganic base (e.g., potassium carbonate, sodium carbonate, cesium carbonate, or potassium hydroxide). The palladium catalyst is preferably tetrakis(triphenylphosphine)palladium, dichlorobis(triphenylphosphine)palladium, palladium chloride-1,1′-bis(diphenylphosphino)ferrocene, or the like. Heating the reaction mixture by microwave irradiation is advantageous for smooth reaction in some cases.

DOCUMENTS

  • A. d. Meijere and F. Diederich (ed.), “Metal-Catalyzed Cross-Coupling Reactions”, first edition, VCH Publishers Inc., 1997
  • The Chemical Society of Japan (ed.), “The Fifth Series of Experimental Chemistry”, vol. 13, MARUZEN Co., Ltd., 2005

(Step 2)

This process is intended to prepare compound (1-2) of the present invention by reducing the alkyne moiety of compound (I-1) of the present invention to alkylene by hydrogenation or diimide reduction.

In this process, compound (I-1) of the present invention and palladium carbon are used in equal amounts or one of them is used in an excessive amount. A mixture of these is stirred in a solvent inert to the reaction, under a hydrogen atmosphere, generally for 0.1 hour to 5 days under conditions between room temperature and heating to reflux. Examples of the solvent used in this process include, but are not particularly limited to, ethers (e.g., diethyl ether, tetrahydrofuran, dioxane, dimethoxyethane), alcohols (e.g., methanol, ethanol, 2-propanol, butanol), and mixtures thereof.

Other than the hydrogenation reaction, compound (I-1) of the present invention and predetermined diimide are used in equal amounts or one of them is used in an excessive amount. A mixture of these is stirred in a solvent inert to the reaction, generally for 0.1 hour to 5 days under conditions between room temperature and heating to reflux. Examples of the solvent used in this process are the same as shown above. The predetermined diimide is, for example, 4-methylbenzenesulfonyl hydrazide.

The substituent(s) on ring W in the compound of formula (I) can be easily converted into other functional groups by the reaction described below in the Examples, reaction obvious to those skilled in the art, or a modified process thereof, using a compound of formula (I) as a starting material. For example, the conversion can be achieved by combining any processes that can be applied generally by those skilled in the art, such as reduction, halogenation, deprotection, hydrolysis, amidation, amination, oxidation, reductive amination, acylation, O-alkylation, N-alkylation, reductive alkylation, and epoxidation.

(Preparation of Starting Compound)

The starting compound used in the preparation process described above can be prepared, for example, by a process described below, the process in the Preparation Examples described later, a known process, or a modified process thereof.

(Starting Material Synthesis 1)

(In this formula, R4 represents —OH or -lower alkylene-OH; L7 represents halogen, —OH, -lower alkylene-OH, -lower alkylene-OMs, -lower alkylene-OTs, -lower alkylene-OTf, or -lower alkylene-halogen; L8 represents -lower alkylene-O— or —O-lower alkylene-. The same applies hereinafter.)

This preparation process is intended to prepare compound (3c) which is starting compound (1a) of the Preparation Process 1 wherein L1 is —O-lower alkylene- or -lower alkylene-O—.

In the case of compound (3a) wherein L7 is halogen, -lower alkylene-OMs, -lower alkylene-OTs, -lower alkylene-OTf, or -lower alkylene-halogen, compounds (3a) and (3b) are used in equal amounts or one of them is used in an excessive amount. A mixture of these is stirred in the presence of a base in a solvent inert to the reaction, generally for 0.1 hour to 5 days under conditions between room temperature and heating to reflux. Examples of the solvent used in this process include, but are not particularly limited to, N-methylpyrrolidone, N,N-dimethylformamide, dimethyl sulfoxide, and the like. The base is preferably an inorganic base such as potassium carbonate, sodium carbonate, cesium carbonate, or potassium hydroxide.

In the case of compound (3a) wherein L7 is —OH or -lower alkylene-OH, compounds (3a) and (3b) are used in equal amounts or one of them is used in an excessive amount. A mixture of these is stirred in the presence of a predetermined phosphine reagent and a predetermined condensing agent in a solvent inert to the reaction, generally for 0.1 hour to 5 days under conditions between room temperature and heating to reflux. Examples of the solvent used in this process include, but are not particularly limited to, ethers such as diethyl ether, tetrahydrofuran, dioxane, and dimethoxyethane. Examples of the predetermined phosphine reagent include tributylphosphine, triphenylphosphine, and the like. Examples of the predetermined condensing agent include diethyl azodicarboxylate, 1,1′-(azodicarbonyl)dipiperidine, and the like. Use of (cyanomethylene)trimethylphosphorane, instead of the predetermined phosphine and the predetermined condensing agent, is advantageous for smooth reaction in some cases.

(Starting Material Synthesis 2)

(In this formula, L9 represents halogen. The same applies hereinafter.)

This preparation process is intended to prepare compound (4d) which is starting compound (1a) of the Preparation Process 1 wherein L1 is lower alkylene.

(Step 1)

This process is intended to prepare compound (4b) by Sonogashira coupling reaction of compound (4a) and a terminal alkyne derivative.

The reaction conditions are the same as in Step 1 of the Preparation Process 2.

(Step 2)

This process is intended to prepare compound (4c) by reducing the alkyne moiety of compound (4b) to lower alkylene by hydrogenation.

The reaction conditions are the same as in Step 2 of the Preparation Process 2.

(Step 3)

This process is intended to prepare compound (4d) by converting the amino group of compound (4c) into halogen.

In this process, compound (4c) and a combination of copper chloride (II) and n-pentyl nitrite are used in equal amounts or one of them is used in an excessive amount. A mixture of these is stirred in a solvent inert to the reaction, generally for 0.1 hour to 5 days under conditions between ice cooling and heating to reflux. Examples of the solvent used in this process include, but are not particularly limited to, halogenated hydrocarbons such as dichloromethane, 1,2-dichloroethane, and chloroform.

(Starting Material Synthesis 3)

This preparation process is intended to prepare compound (5c) which is starting compound (1b) of the Preparation Process 2 wherein X is N.

This reaction is intended to prepare compound (5c) by ipso-substitution reaction of compounds (5a) and (5b).

Compounds (5a) and (5b) are used in equal amounts or one of them is used in an excessive amount. A mixture of these is stirred in a solvent inert to the reaction under a hydrogen atmosphere, generally for 0.1 hour to 5 days under conditions between ice cooling and heating to reflux. Examples of the solvent used in this process include, but are not particularly limited to, alcohols (e.g., methanol, ethanol, 2-propanol, butanol), N-methylpyrrolidone, N,N-dimethylformamide, dimethyl sulfoxide, and mixtures thereof. Use of an acid such as methanesulfonic acid, acetic acid, trifluoroacetic acid, hydrogen chloride, or sulfuric acid is advantageous for smooth reaction in some cases.

The pharmacological activity of the compounds of formula (I) was confirmed in the tests described below.

Test Example 1: FGFR1, FGFR2, and FGFR3 Enzyme Assay

In the enzyme assay, human recombinant FGFR1, FGFR2, and FGFR3 (Carna Biosciences; Catalog Nos. 08-133, 08-134, and 08-135) were used, and reactions were performed at room temperature (FGFR1 and FGFR2) or 30° C. (FGFR3). The measurement method is outlined below.

The compound was diluted with a solution of dimethyl sulfoxide (DMSO) (10-fold common ratio, 4 portions) before dilution with a reaction buffer (100 mM HEPES (pH7.5), 0.003% Brij-35, 0.004% Tween 20, 0.5 mM DTT, and 10 mM MgCl2) so that the final DMSO concentration was 2%. To 4 μL of the compound solution in a 384-well plate, 2 μL each of FGFR1 enzyme (2 or 3 ng/μL), FGFR2 enzyme (2 ng/μL), or FGFR3 enzyme (6 ng/μL) which were diluted with the reaction buffer was added. In 20 minutes, 4 μL of a substrate-ATP solution (100 mM HEPES (pH7.5), 0.003% Brij-35, 0.004% Tween 20, 0.5 mM DTT, 10 mM MgCl2, 3.75 μM substrate-FL-peptide 22+500 μM (FGFR1) ATP, 188 μM (FGFR2) ATP, or 250 μM (FGFR3) ATP) was added before subsequent 30-minute reaction. After the reaction was stopped, the reaction mixture was measured with a LabChip EZ Reader. The IC50 values were calculated by non-linear regression based on the inhibition rates obtained. The results of some compounds are shown in Table 1. The term “Ex” in the table denotes compound No. in the Examples described later.

TABLE 1 FGFR1 FGFR2 FGFR3 Ex IC50 (nM) IC50 (nM) IC50 (nM) 7 2 3 1 11 1 2 27 2 1 <1 33 2 3 2 56 2 2 1 57 1 2 2 71 2 3 2 72 2 2 84 2 2 1 87 3 2 92 2 1 95 3 2 113 1 2 114 2 1 115 2 2 116 1 2 122 2 2 248 4 5 299 <1   2

Test Example 2: Growth Assay of Cells with Forced Expression of Mutant FGFR3 (FGFR3_S249C/NIH3T3)

FGFR3_S249C/NIH3T3 cells were added to a 96-well spheroid plate (U bottom) at a concentration of 3000 cells/well/90 μL, and the compound solution (10 μL) was added thereto on the next day (final DMSO concentration: 0.1%). The compound solution was prepared by serially diluting the compound with DMSO at a 3-fold common ratio (9 portions and DMSO only) from the maximum concentration of 10 mM and then diluted 100-fold with a culture medium (D-MEM, 10% FBS). 5 days after the addition of the compound, the growth inhibition caused by the compound was evaluated by Promega (G7573) CellTiter-Glo™ Luminescent Cell Viability Assay. The IC50 value was calculated by non-linear regression, using DMSO-added wells as control and assuming count 0 to be 100% inhibition. The results of some compounds are shown in Table 2.

TABLE 2 Ex IC50 (nM) 7 18 11 13 27 13 33 26 56 13 57 5 71 10 72 24 84 16 87 20 92 7 95 57 113 11 114 7 115 36 116 8 122 7 248 50 299 10

Test Example 3: Antitumor Test on UM-UC-14 (FGFR3_S249C-Positive Cells, Bladder Cancer)

3×106 UM-UC-14 cells per 0.1 mL (PBS+matrigel, 1:1) were inoculated subcutaneously into the right flank of nude mice (CAnN, Cg-Foxn1nu/CrlCrlj (nu/nu), male, 4- to 5-week-old), and when their tumor size reached about 250 mm3, drug administration was started (Day 1). The drug was administered once a day and the tumor size was measured with a caliper and the body weight was also measured every two or three days. The antitumor effect was finally determined based on the tumor volume (mm3; minor axis (mm)× minor axis (mm)×major axis (mm)/2) at Day 11 (n=3-5). To the control group, 0.5% MC (methyl cellulose) was administered. For “% inhibition” in the table, for example, 100% inhibition indicates that the tumor growth of the control was inhibited to the level of the tumor volume at Day 1. “% regression” indicates what percentage of regression could be achieved compared with the tumor volume at Day 1. Here, the tumor volume at Day 1 means tumor volume immediately before drug administration. The results of some compounds administered orally (1 mg/kg/day for other than Ex 95 and 3 mg/kg/day for Ex 95) are shown in Table 3.

TABLE 3 Ex Antitumor Activity 7 33% inhibition 11 53% regression 27 62% inhibition 33 70% inhibition 56 4% regression 57 77% inhibition 71 50% inhibition 72 30% inhibition 84 34% regression 87 72% inhibition 92 49% inhibition 95 97% inhibition 113 4% regression 114 33% regression 115 70% inhibition 116 54% regression 122 15% regression 248 95% inhibition 299 15% regression

The test described above confirmed that the plural compounds of the Examples included in formula (I) of the present invention had inhibitory action on FGFR1, FGFR2, and/or FGFR3. It was also confirmed that the plural compounds of the Examples included in formula (I) inhibited the growth of the cells with forced expression of mutant FGFR3 and that the compounds also inhibited the growth of bladder cancer or made bladder cancer itself regress, in the animal model bearing mutant FGFR3-positive bladder cancer. In light of the foregoing, the compound of formula (I) or a salt thereof can be used as a therapeutic agent for various cancers related to FGFR1, FGFR2, and/or FGFR3, particularly, mutant FGFR3-positive bladder cancer.

Test Example 4: Isolation of FGFR3-TACC3_v1

cDNA was synthesized by reverse transcription reaction in 200 clinical specimens of lung cancer (Asterand plc.; US) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and random primers (random primers, Life Technologies Corp.) in accordance with the protocol of the kit.

Next, PCR (30 cycles of 98° C. for 10 seconds, 55° C. for 15 seconds, 68° C. for 1.5 minutes) was carried out using primers FGFR3_TACC3_RT_F represented by SEQ ID No: 1 and FGFR3_TACC3_RT_R represented by SEQ ID No: 2, the cDNA obtained above as a template, and DNA polymerase (TaKaRa Ex Taq; Takara Bio Inc.). Additional PCR (30 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 1 minute) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_nested_F represented by SEQ ID No: 3 and FGFR3_TACC3_nested_R represented by SEQ ID No: 4, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 500 bases was obtained from only sample Lg344 specimen.

After that, the PCR product was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies Corp.). As a result, the PCR product of about 500 bases was found to be a sequence obtained by fusion of the 3′ end of exon 18 in the coding sequence (hereinafter, CDS) of FGFR3 (NM_001163213.1) registered in the NCBI to the 5′ end of exon 11 in the CDS of TACC3 (NM_006342.1).

cDNA was synthesized by reverse transcription reaction in the Lg344 specimen RNA which is the lung cancer tissue-derived RNA of a squamous cell lung cancer patient (Asterand plc.; US) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and oligo(dT) primers (oligo(dT)20 primers, Life Technologies, Corp.) in accordance with the protocol of the kit.

Next, PCR (25 cycles of 98° C. for 15 seconds, 60° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using primers FGFR3-TACC3_cloning_F represented by SEQ ID No: 5 and FGFR3-TACC3_cloning_R represented by SEQ ID No: 6, the cDNA obtained above as a template, and DNA polymerase (KOD-plus-Ver. 2; Toyobo Co., Ltd.). Additional PCR (25 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_cloning_BamHI_F represented by SEQ ID No: 7 and FGFR3_TACC3_cloning_EcoRI_R represented by SEQ ID No: 8, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 2.9 k bases was obtained. The PCR product was cloned into a cloning vector (TOPO XL PCR Cloning Kit; Life Technologies, Corp.). The insert was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies, Corp.). As a result, it was found in the PCR product of about 2.9 k bases that there was a transcript obtained by fusion of the region between the 5′-terminus of the CDS of FGFR3 (NM_001163213.1) registered in the NCBI and the 3′ end of exon 18 to the region between the 5′ end of exon 11 in the CDS of TACC3 (NM_006342.1) and the 3′-terminus of the CDS (FGFR3-TACC3_v1) (SEQ ID No: 9). The polypeptide coded by SEQ ID No: 9 (FGFR3-TACC3_v1 fusion polypeptide) is shown in SEQ ID No: 10.

Test Example 5: Isolation of FGFR3-TACC3_v2

cDNA was synthesized by reverse transcription reaction in 59 specimens of bladder cancer (Asterand plc.; US) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and random primers (random primers, Life Technologies Corp.) in accordance with the protocol of the kit.

Next, PCR (30 cycles of 98° C. for 10 seconds, 55° C. for 15 seconds, 68° C. for 1.5 minutes) was carried out using primers FGFR3_TACC3_RT_F represented by SEQ ID No: 1 and FGFR3_TACC3_RT_R represented by SEQ ID No: 2, the cDNA obtained above as a template, and DNA polymerase (TaKaRa Ex Taq; Takara Bio Inc.). Additional PCR (30 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 1 minute) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_nested_F represented by SEQ ID No: 3 and FGFR3_TACC3_nested_R represented by SEQ ID No: 4, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 600 bases was obtained from sample Bd106 specimen.

After that, the PCR product was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies Corp.). As a result, the PCR product of about 600 bases was found to be a sequence obtained by fusion of the 3′ end of exon 18 in the CDS of FGFR3 (NM_001163213.1) registered in the NCBI to the 5′ end of exon 10 in the CDS of TACC3 (NM_006342.1). cDNA was synthesized by reverse transcription reaction in the Bd106 specimen RNA which is the bladder cancer tissue-derived RNA of a bladder cancer patient (Asterand plc.; US) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and oligo(dT) primers (oligo(dT)20 primers, Life Technologies, Corp.) in accordance with the protocol of the kit.

Next, PCR (25 cycles of 98° C. for 15 seconds, 60° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using primers FGFR3-TACC3_cloning_F represented by SEQ ID No: 5 and FGFR3-TACC3_cloning_R represented by SEQ ID No: 6, the cDNA obtained above as a template, and DNA polymerase (KOD-plus-Ver. 2; Toyobo Co., Ltd.). Additional PCR (25 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_cloning_BamHI_F represented by SEQ ID No: 7 and FGFR3_TACC3_cloning_EcoRI_R represented by SEQ ID No: 8, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 3.0 k bases was obtained. The PCR product was cloned into a cloning vector (TOPO XL PCR Cloning Kit; Life Technologies, Corp.). The insert was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies, Corp.). As a result, it was found in the PCR product of about 3.0 k bases that there was a transcript obtained by fusion of the region between the 5′-terminus of the CDS of FGFR3 (NM_001163213.1) registered in the NCBI and the 3′ end of exon 18 to the region between the 5′ end of exon 10 in the CDS of TACC3 (NM_006342.1) and the 3′-terminus of the CDS (FGFR3-TACC3_v2) (SEQ ID No: 11). The polypeptide coded by SEQ ID No: 11 (FGFR3-TACC3 v2 fusion polypeptide) is shown in SEQ ID No: 12.

Test Example 6: Isolation of FGFR3-TACC3_v3

cDNA was synthesized by reverse transcription reaction in 59 specimens of bladder cancer (Asterand plc.; US) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and random primers (random primers, Life Technologies Corp.) in accordance with the protocol of the kit.

Next, PCR (30 cycles of 98° C. for 10 seconds, 55° C. for 15 seconds, 68° C. for 1.5 minutes) was carried out using primers FGFR3_TACC3_RT_F represented by SEQ ID No: 1 and FGFR3 TACC3_RT_R represented by SEQ ID No: 2, the cDNA obtained above as a template, and DNA polymerase (TaKaRa Ex Taq; Takara Bio Inc.). Additional PCR (30 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 1 minute) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_nested_F represented by SEQ ID No: 3 and FGFR3_TACC3_nested_R represented by SEQ ID No: 4, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 650 bases was obtained from sample Bd021 specimen.

After that, the PCR product was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies Corp.). As a result, the PCR product of about 650 bases was found to be a sequence obtained by fusion of a certain sequence of exon 19 in the CDS of FGFR3 (NM_001163213.1) registered in the NCBI to a part of intron 10-11 of TACC3 (NM_006342.1) and to the 5′ end of exon 11 in the CDS of TACC3.

cDNA was synthesized by reverse transcription reaction in the Bd021 specimen RNA which is the bladder cancer tissue-derived RNA of a bladder cancer patient (Asterand plc.; US) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and oligo(dT) primers (oligo(dT)20 primers, Life Technologies, Corp.) in accordance with the protocol of the kit.

Next, PCR (25 cycles of 98° C. for 15 seconds, 60° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using primers FGFR3-TACC3_cloning_F represented by SEQ ID No: 5 and FGFR3-TACC3_cloning_R represented by SEQ ID No: 6, the cDNA obtained above as a template, and DNA polymerase (KOD-plus-Ver. 2; Toyobo Co., Ltd.). Additional PCR (25 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_cloning_BamHI_F represented by SEQ ID No: 7 and FGFR3_TACC3_cloning_EcoRI_R represented by SEQ ID No: 8, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 3.0 k bases was obtained. The PCR product was cloned into a cloning vector (TOPO XL PCR Cloning Kit; Life Technologies, Corp.). The insert was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies, Corp.). As a result, it was found in the PCR product of about 3.0 k bases that there was a transcript obtained by fusion of the region between the 5′-terminus of the CDS of FGFR3 (NM_001163213.1) registered in the NCBI and a certain sequence of exon 19 to part of intron 10-11 of TACC3 (NM_006342.1) and further to the region between the 5′ end of exon 11 in the CDS of TACC3 and the 3′-terminus of the CDS (FGFR3-TACC3_v3) (SEQ ID No: 13). The polypeptide coded by SEQ ID No: 13 (FGFR3-TACC3_v3 fusion polypeptide) is shown in SEQ ID No: 14.

Test Example 7: Isolation of FGFR3-TACC3_v1 from Bladder Cancer Patient-Derived Cell Line RT-112

cDNA was synthesized by reverse transcription reaction in RNA purified from bladder cancer patient-derived cell line RT-112 (purchased from Leibniz-Institut DSMZ-Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and oligo(dT) primers (oligo(dT)20 primers, Life Technologies, Corp.) in accordance with the protocol of the kit.

Next, PCR (25 cycles of 98° C. for 15 seconds, 60° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using primers FGFR3-TACC3_cloning_F represented by SEQ ID No: 5 and FGFR3-TACC3_cloning_R represented by SEQ ID No: 6, the cDNA obtained above as a template, and DNA polymerase (KOD-plus-Ver. 2; Toyobo Co., Ltd.). Additional PCR (25 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_cloning_BamHI_F represented by SEQ ID No: 7 and FGFR3_TACC3_cloning_EcoRI_R represented by SEQ ID No: 8, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 2.9 k bases was obtained. The PCR product was cloned into a cloning vector (TOPO XL PCR Cloning Kit; Life Technologies, Corp.), and the insert was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies, Corp.). As a result, it was found that a transcript obtained was the same as the transcript obtained by fusion of the region between the N-terminus of the CDS of FGFR3 (NM_001163213.1) registered in the NCBI and the 3′ end of exon 18 to the region between the 5′ end of exon 11 in the CDS of TACC3 (NM 006342.1) and the C-terminus of the CDS (FGFR3-TACC3_v1) (SEQ ID No: 9).

Test Example 8: Isolation of FGFR3-TACC3_v4 from Bladder Cancer Patient-Derived Cell Line RT4

cDNA was synthesized by reverse transcription reaction in RNA purified from bladder cancer patient-derived cell line RT4 (purchased from ECACC (European Collection of Cell Cultures)) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and oligo(dT) primers (oligo(dT)20 primers, Life Technologies, Corp.) in accordance with the protocol of the kit. Next, PCR (30 cycles of 98° C. for 15 seconds, 60° C. for 15 seconds, 68° C. for 5.5 minutes) was carried out using primers FGFR3-TACC3_cloning_F represented by SEQ ID No: 5 and FGFR3-TACC3_cloning_R represented by SEQ ID No: 6, the cDNA obtained above as a template, and DNA polymerase (KOD-plus-Ver. 2; Toyobo Co., Ltd.). Additional PCR (30 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 5 minutes) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_TACC3_cloning_BamHI_F represented by SEQ ID No: 7 and FGFR3_TACC3_cloning_EcoRI_R represented by SEQ ID No: 8, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 4.5 k bases was obtained. The PCR product was cloned into a cloning vector (TOPO XL PCR Cloning Kit; Life Technologies, Corp.), and the insert was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies, Corp.). As a result, it was found that there was a transcript obtained by fusion of part of intron 18-19 sequence of FGFR3 (NM_001163213.1) registered in the NCBI to the region between the 5′-terminus of the CDS of the FGFR3 and the 3′ end of exon 18 and further to the region between a certain sequence of exon 4 of TACC3 (NM_006342.1) and the 3′-terminus of the CDS of the TACC3 (FGFR3-TACC3 v4). In the confirmed sequence, T at base position 882 was replaced by C (SNPs registration No.; rs2234909), C at base position 2484 by T, and G at base position 2663 by A (SEQ ID No: 15). The polypeptide coded by SEQ ID No: 15 (FGFR3-TACC3_v4 fusion polypeptide) is shown in SEQ ID No: 16.

Test Example 9: Preparation of Retrovirus Solutions of FGFR3-TACC3_v1, FGFR3-TACC3_v2, FGFR3-TACC3_v3, and FGFR3-TACC3_v4

To express, as proteins, the ORF full lengths of FGFR3-TACC3_v1, FGFR3-TACC3_v2, FGFR3-TACC3_v3, and FGFR3-TACC3_v4, enzyme reaction was performed at 37° C. for 3 hours using the cloning vectors prepared in Test Examples 4, 5, 6, and 8 and restriction enzyme BamHI, and restriction enzyme digested DNA fragments were obtained and purified. Another enzyme reaction was performed at 37° C. for 3 hours using EcoRI and the DNA fragments, and restriction enzyme digested DNA fragments were obtained and purified. The ORF-containing DNA fragments were cloned into BamHI and EcoRI sites in the multicloning site of an expression vector (pMXs-puro; Cosmo Bio) to construct expression plasmids (FGFR3-TACC3_v/pMXs-puro, FGFR3-TACC3_v2/pMXs-puro, FGFR3-TACC3_v3/pMXs-puro, and FGFR3-TACC3_v4/pMXs-puro).

9 μg each of FGFR3-TACC3_v1/pMXs-puro, FGFR3-TACC3_v2/pMXs-puro, FGFR3-TACC3_v3/pMXs-puro, and FGFR3-TACC3_v4/pMXs-puro was transfected into Platinum-E cells, using a transfection reagent (FUGENE® HD, Roche). At 24 hours after the transfection, D-MEM media (Dulbecco's Modified Eagle Medium; Invitrogen) containing 10% bovine serum (Nichirei Biosciences) were replaced, and the culture supernatants were collected after 24 hours to prepare retrovirus solutions.

Test Example 10: Investigation of Anchorage-Independent Growth-Promoting Action of FGFR3-TACC3_v1, FGFR3-TACC3_v2, FGFR3-TACC3_v3, and FGFR3-TACC3_v4

To the virus solutions prepared using FGFR3-TACC3_v1/pMXs-puro, FGFR3-TACC3_v2/pMXs-puro, FGFR3-TACC3_v3/pMXs-puro, and FGFR3-TACC3_v4/pMXs-puro in Test Example 9, 4 μg/mL of polybrene (Polybrene; Sigma) was added followed by addition of the resulting mixtures to NIH3T3 cells for infection. At 6 hours after the addition, the media used were replaced by D-MEM media containing 10% bovine serum (Nichirei Biosciences), and, on the day after the infection, the media were replaced by D-MEM media (Invitrogen) containing 10% bovine serum (Nichirei Biosciences) and 1 μg/mL of puromycin (Sigma). The culture was continued in the presence of 5% CO2 at 37° C. for 4 weeks to obtain NIH3T3 cells stably expressing each of FGFR3-TACC3_v1, FGFR3-TACC3_v2, FGFR3-TACC3_v3, and FGFR3-TACC3_v4 (these cells were designated as FGFR3-TACC3_v1-expressing NIH3T3 cells, FGFR3-TACC3_v2-expressing NIH3T3 cells, FGFR3-TACC3_v3-expressing NIH3T3 cells, and FGFR3-TACC3_v4-expressing NIH3T3 cells, respectively.)

To investigate the anchorage-independent growth-promoting ability of FGFR3-TACC3_v1-expressing NIH3T3 cells, FGFR3-TACC3_v2-expressing NIH3T3 cells, FGFR3-TACC3_v3-expressing NIH3T3 cells, and FGFR3-TACC3_v4-expressing NIH3T3 cells, these cells and NIH3T3 cells infected with a blank vector pMXs-puro (Mock/NIH3T3 cells) were each seeded at 1×103 cells per well in D-MEM media (Invitrogen) containing 10% bovine serum (Nichirei Biosciences) in a 96-well spheroid plate (Sumilon Celltight Spheroid 96U; Sumitomo Bakelite). The cells were cultured in the presence of 5% CO2 at 37° C. and were counted on the next day (Day 1) and 4 days later (Day 4), using a cell counting reagent (CELLTITER-Glo™ Luminescent Cell Viability Assay; Promega) in accordance with the method described in the manual. A luminometer was used for detection. It was confirmed that the count of Mock/NIH3T3 cells did not increase between Day 1 and Day 4, while the counts of FGFR3-TACC3_v1-expressing NIH3T3 cells, FGFR3-TACC3_v2-expressing NIH3T3 cells, FGFR3-TACC3_v3-expressing NIH3T3 cells, and FGFR3-TACC3_v4-expressing NIH3T3 cells increased about 3.1-fold, about 2.8-fold, about 2.3-fold, and about 2.5-fold, respectively, between Day 1 and Day 4.

In light of the foregoing, it was found that FGFR3-TACC3_v1-expressing NIH3T3 cells, FGFR3-TACC3_v2-expressing NIH3T3 cells, FGFR3-TACC3_v3-expressing NIH3T3 cells, and FGFR3-TACC3_v4-expressing NIH3T3 cells exhibit anchorage-independent cell growth.

Test Example 11: Anchorage-Independent Cell Growth-Inhibitory Activity of Compounds on FGFR3-TACC3_v1-Expressing NIH3T3 Cells, FGFR3-TACC3_v2-Expressing NIH3T3 Cells, FGFR3-TACC3_v3-Expressing NIH3T3 Cells, FGFR3-TACC3_v4-Expressing NIH3T3 Cells, and Bladder Cancer Patient-Derived Cell Lines RT-112 and RT4

Measurement for anchorage-independent cell growth (colony method, etc.) is known to be a system for investigating the anticancer action (pharmacological effect) of compounds (Clinical Oncology, second edition, Cancer and Chemotherapy Publishers Inc.). As a method for measuring the non-adhesive growth of cells, there is the following method using a spheroid plate as referred to above in place of the colony method.

In a 96-well spheroid plate (Sumilon Celltight Spheroid 96U; Sumitomo Bakelite), FGFR3-TACC3_v1-expressing NIH3T3 cells, FGFR3-TACC3_v2-expressing NIH3T3 cells, FGFR3-TACC3_v3-expressing NIH3T3 cells, and FGFR3-TACC3_v4-expressing NIH3T3 cells were each seeded at 1×103 cells per well in D-MEM media (Invitrogen) containing 10% fetal bovine serum. Likewise, bladder cancer patient-derived cell line RT-112 was seeded at 1×103 cells per well in RPMI1640 medium containing 10% fetal bovine serum and 2 mM L-glutamine, and bladder cancer patient-derived cell line RT4 was seeded at 1×103 cells per well in RPMI1640 medium containing 10% fetal bovine serum. A well supplemented with only medium was also prepared for a positive control. Culturing was performed overnight in the presence of 5% CO2 at 37° C. followed by addition of test compounds (final concentrations: 100 nM, 10 nM, and 1 nM). As a negative control, DMSO used as a solvent for the compounds was added at the same concentration (0.1%) as in the case of addition of the compounds. Then, culturing was performed in the presence of 5% CO2 at 37° C. for 4 days, and a cell counting reagent (CellTiter-Glo™ Luminescent Cell Viability Assay; Promega) was added and the resulting mixture was stirred for 20 minutes followed by measurement with a luminometer. Assuming that the values of the positive control and the negative control were 100% inhibition and 0% inhibition, respectively, the growth inhibition rate (%) was calculated for each compound. As shown in Table 4, it was found out that some compounds of the present invention inhibited the anchorage-independent growth of FGFR3-TACC3_v1-expressing NIH3T3 cells, FGFR3-TACC3_v2-expressing NIH3T3 cells, FGFR3-TACC3_v3-expressing NIH3T3 cells, FGFR3-TACC3_v4-expressing NIH3T3 cells, and bladder cancer patient-derived cell lines RT-112 and RT4.

The results described above showed that the growth of cancer cells and tumors that express FGFR3-TACC3_v1, FGFR3-TACC3_v2, FGFR3-TACC3_v3, and FGFR3-TACC3_v4 can be inhibited by the compounds of the present invention.

TABLE 4 Ex v1 v2 v3 v4 RT-112 RT4 56 100 nM 92 91 91 90 90 64 10 nM 84 79 78 82 83 42 1 nM 22 21 20 22 29 5 113 100 nM 91 91 87 88 89 64 10 nM 53 42 32 73 77 39 1 nM 4 2 3 13 23 8 116 100 nM 91 90 86 89 89 63 10 nM 44 31 24 70 72 39 1 nM 5 0 3 11 21 8 122 100 nM 90 88 89 89 89 63 10 nM 84 79 79 82 80 43 1 nM 26 23 25 19 23 6 248 100 nM 84 79 81 83 81 43 10 nM 28 29 20 26 33 10 1 nM 7 11 6 −5 5 3 299 100 nM 92 91 89 90 89 63 10 nM 77 63 51 82 84 45 1 nM 9 4 5 16 31 8

Test Example 12: Inhibitory Activity of Compounds on the In Vitro Kinase Activity of FGFR3-TACC3 Fusion Polypeptide

(1) Construction of FLAG-Tag Fusion Expression Plasmids (FGFR3-TACC3_v1 (N-FLAG)/pcDNA3.1/Zeo(+), FGFR3-TACC3_v2 (N-FLAG)/pcDNA3.1/Zeo(+), and FGFR3-TACC3_v3 (N-FLAG)/pcDNA3.1/Zeo(+))

To obtain 5′-terminally FLAG-tagged FGFR3-TACC3 fusion polynucleotide, PCR was carried out for 5′-terminal FLAG tagging using the vectors cloned in Test Examples 4, 5, and 6 as templates. PCR (12 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 3.5 minutes) was carried out using primers FGFR3_N_FLAG_BamHI represented by SEQ ID No: 17 and FGFR3_TACC3_cloning_EcoRI_R represented by SEQ ID No: 8 and DNA polymerase (KOD-plus-Ver. 2; Toyobo Co., Ltd.). PCR products obtained were cloned into cloning vectors (TOPO XL PCR Cloning Kit; Life Technologies, Corp.). The inserts were sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies, Corp.). As a result, it was confirmed that the PCR products were nucleic acid sequences of SEQ ID Nos: 9, 11, and 13 in which the three bases coding for the first methionine (ATG) were deleted and start codon and a nucleic acid sequence coding for FLAG tag (SEQ ID No: 24) were added to the 5′-terminus. Polypeptides coded by the above are referred to FGFR3-TACC3_v1 (N-FLAG) fusion polypeptide, FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide and FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide, respectively, and these polypeptides are collectively referred to FGFR3-TACC3 (N-FLAG) fusion polypeptide. Further, to construct an expression vector expressing, as a protein, each of the ORF full lengths of FGFR3-TACC3_v (N-FLAG), FGFR3-TACC3_v2 (N-FLAG), and FGFR3-TACC3_v3 (N-FLAG) which contained these FLAG sequences added, enzyme reaction was performed at 37° C. for 3 hours using the cloning vectors described above and restriction enzyme BamHI, and restriction enzyme digested DNA fragments were obtained and purified. Further, enzyme reaction was performed at 37° C. for 3 hours using EcoRI and the DNA fragments, and restriction enzyme digested DNA fragments were obtained and purified. These ORF-containing DNA fragments were cloned into BamHI and EcoRI sites in the multicloning site of an expression vector (pcDNA3.1/Zeo(+); Life Technologies, Corp.) to construct expression plasmids (FGFR3-TACC3_v1 (N-FLAG)/pcDNA3.1/Zeo(+), FGFR3-TACC3_v2 (N-FLAG)/pcDNA3.1/Zeo(+), and FGFR3-TACC3_v3 (N-FLAG)/pcDNA3.1/Zeo(+)).

(2) Preparation of FGFR3-TACC3 (N-FLAG) Fusion Polypeptide

On the day before transfection, 0.5×107 HEK293 cells per collagen-coated 15-cm dish were cultured in D-MEM medium containing 10% fetal bovine serum to prepare 10 dishes. On the day of transfection, 27 μg each of FGFR3-TACC3_v1 (N-FLAG)/pcDNA3.1/Zeo(+), FGFR3-TACC3 v2 (N-FLAG)/pcDNA3.1/Zeo(+), and FGFR3-TACC3_v3 (N-FLAG)/pcDNA3.1/Zeo(+) (Test Example 12) per dish was transfected into HEK293 cells, using 81 μL of a transfection reagent (FUGENE® HD, Roche). At 24 hours after the transfection, the media were removed, and after washing three times with PBS, 1 mL of PBS was added. The cells were scraped with a cell scraper (Corning Inc.) and then recovered in polypropylene tubes. After centrifugation at 1200 rpm for 5 minutes, the supernatant was removed, 150 μL of a cell lysate (50 mM Tris-HCl (pH8.0), 150 mM NaCl, 1% NP-40, 1 mM EDTA, and protease inhibitor cocktail complete) was added, and the cells were incubated on ice for 30 minutes and lysed. Each of the FGFR3-TACC3_v1 (N-FLAG) fusion polypeptide, FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, and FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide which were present in the supernatant obtained after the centrifugation was purified using M2 antibody affinity gel (ANTI-FLAG M2 Affinity Gel; Sigma-Aldrich) in accordance with the method described in the product information document. A wash liquid (50 mM Tris-HCl (pH8.0), 150 mM NaCl, 1% NP-40, 1 mM EDTA, and protease inhibitor cocktail complete) and an eluate (20 mM Tris-HCl (pH7.4), 10 mM MgCl2, 10 mM MnCl2, and 0.5 mg/mL of FLAG peptide) were used for washing and elution, respectively, to give 100 μL of eluates. The eluates were subjected to immunoblotting using an anti-FGFR3 antibody (Cell Signaling Technology) and an anti-FLAG M2 antiboty (Sigma-Aldrich) and silver staining, and then confirmed that FGFR3-TACC3_v1 (N-FLAG) fusion polypeptide, FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, and FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide were obtained.

(3) Detection of the In Vitro Kinase Activity of FGFR3-TACC3 (N-FLAG) Fusion Polypeptide

FGFR3-TACC3_v (N-FLAG) fusion polypeptide, FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, and FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide, which were purified as described above were used to investigate their phosphorylating activity against a peptide substrate by using a kinase activity detection kit (HTRF KinEASE-TK; Cisbio). The reaction buffer was prepared by adding 1 mM (final concentration) of DTT and 5 mM (final concentration) of Mg to 5× kinase buffer enclosed in the kit using 1 μL of 1-fold, 3-fold and 10-fold diluted solutions of the above prepared elutates as enzyme solutions, respectively, in 384-well, low-volume black plate (Corning). Using 2.0 μM (final concentration) of TK Substrate enclosed in the kit as a substrate, the reaction was performed in a final volume of 5.0 μL at room temperature for 1 hour in each case of adding no ATP and adding 100 μM ATP (final concentration). After the reaction, Sa-XL665 solution and TK Antibldy-Eu(K) solution were prepared in accordance with kit-recommended method and added each of 2.5 μL of the solutions. After the reaction was performed at room temperature for 1 hour, the HTRF counts (i.e., phosphorylation of the peptide substrate) were detected. As the results, it was showed that compared with ATP-free ones, the HTRF counts in ATP-added ones had increased about 38-fold, about 40-fold, and about 38-fold, respectively, in the case of adding 1 μL of 1-fold diluted solutions of the eluates described above including FGFR3-TACC3_v1 (N-FLAG) fusion polypeptide, FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, and FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide, had increased about 27-fold, 34-fold, and 31-fold, respectively, in the case of adding 1 μL of 3-fold diluted solutions of the eluates, and had increased 5-fold, 18-fold, and 11-fold, respectively, in the case of adding 1 μL of 10-fold diluted solutions of the eluates.

As described above, the in vitro kinase activity of the respective fusion polypeptides could be detected by use of a kinase activity detection kit.

(4) Inhibitory Action of Compounds on the In Vitro Kinase Activity of FGFR3-TACC3 (N-FLAG) Fusion Polypeptide

The inhibitory activity of the test compounds on the in vitro kinase activity of FGFR3-TACC3_v1 (N-FLAG) fusion polypeptide, FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, and FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide was investigated using the kinase activity detection kit described above and 384-well plate of the same sort. The compounds were added so that the final concentrations were 100 nM, 10 nM, and 1 nM, and DMSO was added as a control so that the concentration was 0.1%. For FGFR3-TACC3 v1 (N-FLAG) fusion polypeptide, 1 μL of a 2-fold diluted solution of the eluate described above was added; for FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, 1 μL of a 3-fold diluted solution of the eluate described above was added; and for FGFR3-TACC3 v3 (N-FLAG) fusion polypeptide, 1 μL of a 3-fold diluted solution of the eluate described above was added. TK Substrate enclosed in the kit as a substrate was added in a final concentration of 2.0 μM, the reaction was performed at room temperature for 15 minutes. Then the reaction was performed in a final volume of 5.0 μL at room temperature for 60 minutes in each case of adding no ATP and adding 100 μM ATP (final concentration). After the other processes were performed by addition of each of 2.5 μL of Sa-XL665 solution and TK Antibody-Eu(K) solution prepared by using similar method to that described in (3) above, and the reaction was performed at room temperature for 1 hour, the HTRF counts were detected. Assuming that the phosphorylation counts with adding no ATP and adding ATP in the absence of the compounds (DMSO was added in a concentration of 0.1%, the concentration equal to the compounds) were 100% inhibition and 0% inhibition, respectively, the inhibition rates (%) of the kinase activity of FGFR3-TACC3_v (N-FLAG) fusion polypeptide, FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, and FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide were calculated for the compounds, using the following formula:


[rate (%) of inhibiting kinase activity by compound]=(1−[phosphorylation count with adding compound and adding ATP−phosphorylation count with adding no compound and adding no ATP]/[phosphorylation count with adding no compound and adding ATP−phosphorylation count with adding no compound and adding no ATP])×100

As a result, as shown in Table 5, it was found out that some compounds of the present invention inhibit the phosphorylating activity of purified FGFR3-TACC3_v1 (N-FLAG) fusion polypeptide, purified FGFR3-TACC3_v2 (N-FLAG) fusion polypeptide, and purified FGFR3-TACC3_v3 (N-FLAG) fusion polypeptide against the peptide substrate.

TABLE 5 Ex v1 v2 v3 56 100 nM 92 94 93 10 nM 77 86 85 1 nM 49 33 47 113 100 nM 92 94 96 10 nM 79 74 81 1 nM 28 24 35 116 100 nM 95 95 96 10 nM 79 73 86 1 nM 31 22 41 122 100 nM 94 95 97 10 nM 80 80 85 1 nM 34 27 45 248 100 nM 86 78 91 10 nM 40 25 55 1 nM 7 6 30 299 100 nM 94 95 96 10 nM 84 77 88 1 nM 35 20 47

Test Example 13: Isolation of FGFR3-BAIAP2L1 from Bladder Cancer Patient-Derived Cell Line SW780

cDNA was synthesized by reverse transcription reaction in RNA purified from bladder cancer patient-derived cell line SW780 (purchased from ATCC) using reverse transciptase (SuperScriptIII, Life Technologies, Corp.) and oligo(dT) primers (oligo(dT)20 primers, Life Technologies, Corp.) in accordance with the protocol of the kit.

Next, PCR (30 cycles of 98° C. for 15 seconds, 60° C. for 15 seconds, 68° C. for 5 minutes) was carried out using primers FGFR3-BAIAP2L1_cloning_F represented by SEQ ID No: 18 and FGFR3-BAIAP2L1_cloning_R represented by SEQ ID No: 19, the cDNA obtained above as a template, and DNA polymerase (KOD-plus-Ver. 2; Toyobo Co., Ltd.). Additional PCR (30 cycles of 98° C. for 15 seconds, 55° C. for 15 seconds, 68° C. for 4 minutes) was carried out using the PCR product described above which was diluted 10-fold as a template, primers FGFR3_BAIAP2L1_cloning_BamHI_F represented by SEQ ID No: 20 and FGFR3_BAIAP2L1_cloning_NotI_R represented by SEQ ID No: 21, and the same DNA polymerase as shown above. Electrophoresis performed after the PCR reaction showed that a PCR product of about 3.8 k bases was obtained. The PCR product was cloned into a cloning vector (TOPO XL PCR Cloning Kit; Life Technologies, Corp.), and the insert was sequenced by dideoxy sequencing (BigDye Terminator v3.1 Cycle Sequencing Kit; Life Technologies, Corp.). As a result, the product was found to be a transcript obtained by fusion of the region between the 5′-terminus of the CDS of FGFR3 (NM_001163213.1) registered in the NCBI and the 3′ end of exon 18 to the region between the 5′ end of exon 2 in the CDS of BAIAP2L1 (NM_018842.4) and the 3′-terminus of the CDS (FGFR3-BAIAP2L1). In the confirmed sequence, G at base position 3558 was replaced by A (SNPs registration No.: rs1045916), C at base position 3723 by T, and G at base position 3747 by A (SEQ ID No: 22). The polypeptide coded by SEQ ID No: 22 is shown in SEQ ID No: 23.

Test Example 14: Preparation of Retrovirus Solution of FGFR3-BAIAP2L1

To construct an expression plasmid expressing, as a protein, the ORF full length of FGFR3-BAIAP2L1, enzyme reaction was performed at 37° C. for 3 hours using the cloning vector described above and restriction enzyme BamHI, and restriction enzyme digested DNA fragments were obtained and purified. Further, enzyme reaction was performed at 37° C. for 3 hours using NotI and the DNA fragments, and restriction enzyme digested DNA fragments were obtained and purified. This ORF-containing DNA fragment was cloned into BamHI and NotI sites in the multicloning site of an expression vector (pMXs-puro; Cosmo Bio) to construct an expression plasmid (FGFR3-BAIAP2L1/pMXs-puro). The prepared FGFR3-BAIAP2L1/pMXs-puro was used to prepare a retrovirus solution in accordance with the method used in Test Example 9.

Test Example 15: Investigation of Anchorage-Independent Growth of FGFR3-BAIAP2L1

The virus solution prepared using FGFR3-BAIAP2L1/pMXs-puro in Test Example 14 was used to obtain NIH3T3 cells expressing FGFR3-BAIAP2L1 stably in accordance with the method used in Test Example 10 (designated as FGFR3-BAIAP2L1-expressing NIH3T3 cells).

To investigate the anchorage-independent growth-promoting ability of FGFR3-BAIAP2L1-expressing NIH3T3 cells, the same method as in Test Example 10 was applied. It was confirmed that the count of Mock/NIH3T3 cells did not increase between Day 1 and Day 4, while the count of FGFR3-BAIAP2L1-expressing NIH3T3 cells increased about 2.5-fold between Day 1 and Day 4. In light of the foregoing, it was shown that FGFR3-BAIAP2L1-expressing NIH3T3 cells exhibit anchorage-independent cell growth.

Test Example 16: Inhibitory Activity on Anchorage-Independent Cell Growth of FGFR3-BAIAP2L1-Expressing NIH3T3 Cells

In a 96-well spheroid plate (Sumilon Celltight Spheroid 96U; Sumitomo Bakelite), FGFR3-BAIAP2L1-expressing NIH3T3 cells were seeded at 1×103 cells per well in D-MEM medium containing 10% fetal bovine serum. A well supplemented with only medium was also prepared for a positive control. Culturing was performed overnight in the presence of 5% CO2 at 37° C. followed by addition of test compounds (final concentrations: 100 nM, 10 nM, and 1 nM). As a negative control, DMSO used as a solvent for the compounds was added at the same concentration (0.1%) as in the case of addition of the compounds. Then, culturing was performed in the presence of 5% CO2 at 37° C. for 4 days, and a cell counting reagent (CellTiter-Glo™ Luminescent Cell Viability Assay; Promega) was added and the resulting mixture was stirred for 20 minutes followed by measurement with a luminometer. Assuming that the values of the positive control and the negative control were 100% inhibition and 0% inhibition, respectively, the growth inhibition rate (%) was calculated for each compound. As shown in Table 6, it was found out that some compounds of the present invention inhibit the anchorage-independent growth of FGFR3-BAIAP2L1-expressing NIH3T3 cells.

The results described above showed that the growth of cancer cells and tumors that express FGFR3-BAIAP2L1 can be inhibited by the compounds of the present invention.

TABLE 6 FGFR3-BAIAP2L1- Ex expressing NIH3T3 Cells 56 100 nM 90 10 nM 80 1 nM 24 113 100 nM 89 10 nM 70 1 nM 14 116 100 nM 87 10 nM 74 1 nM 15 122 100 nM 90 10 nM 83 1 nM 17 248 100 nM 82 10 nM 19 1 nM −3 299 100 nM 91 10 nM 81 1 nM 15

A pharmaceutical composition which comprises one or more of the compounds of formula (I) or salts thereof, as active ingredient, can be prepared in a conventional manner by using an excipient commonly used in the art, more specifically, a pharmaceutical excipient, pharmaceutical carrier, or another additive.

Any mode of administration may be used: namely, either oral administration in the form of tablets, pills, capsules, granules, powders, solutions or the like, or parenteral administration in the form of injections (e.g., intraarticular, intravenous, or intramuscular injection), suppositories, eye drops, eye ointments, percutaneous solutions, ointments, percutaneous patches, transmucosal solutions, transmucosal patches, inhalants, intravesical instillation or the like.

Solid compositions used for oral administration include tablets, powders, granules, and the like. In these solid compositions, one or more active ingredients are mixed with at least one inert excipient. The compositions may also comprise inert additives such as lubricants, disintegrating agents, stabilizers, and/or solubilizers, as in usual cases. Tablets or pills may be coated with sugar or a gastrosoluble or enteric film, if needed.

Liquid compositions for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, elixirs, and the like, and comprise a commonly-used inert diluent such as purified water or ethanol. These liquid compositions may comprise auxiliaries (e.g., solubilizers, wetting agents, suspending agents), sweeteners, flavors, aromatics, and/or antiseptics, in addition to such an inert diluent.

Injections for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions. Examples of aqueous solvents include injectable distilled water and physiological saline. Examples of non-aqueous solvents include alcohols such as ethanol. These compositions may further comprise isotonizing agents, antiseptics, wetting agents, emulsifiers, dispersants, stabilizers or solubilizers. They are sterilized, for example, by filtration through a bacteria-retaining filter, by incorporation of disinfectants, or by irradiation. Alternatively, they may be formulated into a sterile solid composition and reconstituted for use by being dissolved or suspended in sterile water or a sterile injectable solvent before use.

Formulations for external use include ointments, plasters, creams, jellies, cataplasms, sprays, lotions, eye drops, eye ointments, and the like. They include commonly-used ointment bases, lotion bases, aqueous or non-aqueous solutions, suspensions, emulsions, or the like.

Transmucosal formulations such as inhalants or transnasal formulations are used in solid, liquid, or semi-solid form and can be prepared in a conventionally known manner. For example, such formulations may be supplemented as appropriate with known excipients, and further with pH adjusters, antiseptics, surfactants, lubricants, stabilizers, thickeners, or the like. For their administration, an appropriate device for inhalation or insufflation may be used. For example, using a known device (e.g., a metered-dose inhalation device) or a nebulizer, the compound(s) may be administered alone or as a powder of a formulated mixture or as a solution or suspension in combination with a pharmaceutically acceptable carrier. Dry powder inhalators and the like may be for single or multiple administration use, and dry powders or powder-containing capsules may be used in such devices. Alternatively, they may be in the form of pressurized aerosol sprays or the like which use an appropriate propellant, for example, a preferred gas such as chlorofluoroalkane or carbon dioxide.

In general, for oral administration, the daily dosage is desirably about 0.001 to 100 mg/kg body weight, preferably 0.1 to 30 mg/kg body weight, more preferably 0.1 to 10 mg/kg body weight, given as a single dose or in 2 to 4 divided doses. For intravenous administration, the daily dosage is desirably about 0.0001 to 10 mg/kg body weight, given in one or several doses per day. Likewise, for transmucosal formulations, the daily dosage is about 0.001 to 100 mg/kg body weight, given in one or several doses per day. The dosage may be determined as appropriate for each case in consideration of symptom, age, sex, and the like.

The pharmaceutical composition of the present invention comprises one or more of the compounds of formula (I) or salts thereof, as active ingredients in an amount of 0.01 to 100 wt. % (0.01 to 50 wt. % in one embodiment), which varies depending on administration route, dosage form, administration site, or the types of excipients and additives.

The compounds of formula (I) can be used in combination with various therapeutic or prophylactic agents for diseases against which the compounds of formula (I) would be effective. In such combination therapy, drugs may be administered simultaneously or separately in succession or at desired time intervals. Formulations for simultaneous administration may be in either mixed form or separate form.

EXAMPLES

The processes for preparing the compounds of formula (I) are described in more detail with reference to the examples shown below. It should be noted that the present invention is not limited to the compounds described in the examples shown below. In addition, the processes for preparing the starting compounds are shown in preparation examples. Processes for preparing the compounds of formula (I) are not limited only to those actually described in the examples shown below, and the compounds of formula (I) may also be prepared by any combination of these processes or by any processes obvious to those skilled in the art.

In the examples, preparation examples and tables shown below, the following abbreviations are used as needed.

PEx: Preparation Example No., Ex: Example No., PSyn: Preparation Example No. of compound prepared in the same manner, Syn: Example No. of compound prepared in the same manner, Str: chemical structural formula (Me: methyl, Et: ethyl, iPr: isopropyl, tBu: tert-butyl, Boc: tert-butoxycarbonyl, Bn: benzyl, THP: tetrahydropyranyl), DAT: physical and chemical data, ESI+: m/z value in mass analysis (ionization method ESI, (M+H)+ unless otherwise specified), ESI−: m/z value (ionization method ESI, (M−H) unless otherwise specified), EI: m/z value in mass analysis (ionization method EI, (M)+ unless otherwise specified), APCI/ESI+: m/z value in mass analysis (simultaneous measurement by ionization methods APCI and ESI, (M+H)+ unless otherwise specified), NMR1: δ (ppm) in 1H-NMR in dimethyl sulfoxide-d6, NMR2: δ (ppm) in 1H-NMR in CDCl3, NMR3: δ (ppm) in 1H-NMR in CD3OD, “M” in Preparation Example and Example: which indicates mol/L. “HCl” in a structural formula indicates hydrochloride and the number in front of the term “HCl” indicates molar ratio. For example, 2HCl means a dihydrochloride salt. The symbol “*” in the tables in Preparation Examples and Examples indicates that the compounds given the symbol are optically active substances.

Preparation Example 1

Under an argon atmosphere, to a mixture of 3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]aniline (300 mg) and ethanol (6 mL), methanesulfonic acid (128 μL) was added followed by stirring at room temperature for 30 minutes. Subsequently, 5-bromo-2-chloropyrimidine (229 mg) was added thereto and the resulting mixture was stirred at 100° C. for 4 hours. Additional 5-bromo-2-chloropyrimidine (95 mg) was added thereto and the resulting mixture was stirred at 100° C. for 12 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was dried over anhydrous sodium sulfate and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/methanol) to give 5-bromo-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (352 mg).

Preparation Example 2

To a mixture of 3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]aniline (253 mg) and isopropanol (6 mL), methanesulfonic acid (162 μL) was added followed by stirring at room temperature for 30 minutes. After that, 2-chloro-5-iodopyrimidine (200 mg) was added thereto, and the resulting mixture was stirred at 90° C. for 12 hours and further stirred at 130° C. for 2 hours under microwave irradiation. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was dried over anhydrous sodium sulfate and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 5-iodo-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (282 mg).

Preparation Example 3

Under an argon atmosphere, a mixture of 1-ethynyl-3,5-dimethoxybenzene (3 g) and acetonitrile (30 mL) was ice cooled, and then sulfuryl chloride (3.15 mL) was added thereto followed by stirring at room temperature for 4 hours. Additional sulfuryl chloride (449 μL) was added thereto followed by stirring at room temperature for 12 hours. After the reaction mixture was concentrated under reduced pressure, ethyl acetate and a saturated aqueous sodium hydrogen carbonate solution were added to the resulting residue followed by stirring at room temperature for 30 minutes. The resulting solid was collected by filtration, washed with ethyl acetate, and then dried under reduced pressure to give 2,4-dichloro-3-ethynyl-1,5-dimethoxybenzene (1.99 g).

Preparation Example 4

A mixture of 1-ethynyl-3,5-dimethoxybenzene (4 g) and acetonitrile (80 mL) was ice cooled, and N-fluoro-N′-(chloromethyl)triethylenediamine bis(tetrafluoroborate) (19.4 g) was added thereto. The resulting mixture was gradually warmed and stirred at room temperature for 12 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, and then dried over anhydrous sodium sulfate and filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) and subsequently purified by silica gel column chromatography (chloroform/hexane) to give 3-ethynyl-2,4-difluoro-1,5-dimethoxybenzene (798 mg, Preparation Example No. PEx. 4-1, which is described later) and 1-ethynyl-2-fluoro-3,5-dimethoxybenzene (375 mg, Preparation Example No. PEx. 4-2, which is described later).

Preparation Example 5

Under an argon atmosphere, a mixture of 1-ethynyl-2-fluoro-3,5-dimethoxybenzene (800 mg) and acetonitrile (8 mL) was ice cooled, and sulfuryl chloride (378 μL) was added thereto followed by stirring at room temperature for 12 hours. To the reaction mixture, ethyl acetate and a saturated aqueous sodium hydrogen carbonate solution were added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was solidified with ethyl acetate/diisopropyl ether to give 2-chloro-3-ethynyl-4-fluoro-1,5-dimethoxybenzene (787 mg).

Preparation Example 6

To a mixture of 2,6-difluoro-3-methoxybenzaldehyde (500 mg), potassium carbonate (803 mg), and methanol (10 mL), dimethyl (1-diazo-2-oxopropyl)phosphonate (523 μL) was added at room temperature under an argon atmosphere followed by stirring for 5 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 2-ethynyl-1,3-difluoro-4-methoxybenzene (452 mg).

Preparation Example 7

To a mixture of 2-amino-5-iodopyrimidine (1 g), 3-ethynyl-2,4-difluoro-1,5-dimethoxybenzene (897 mg), tetrakistriphenylphosphine palladium (261 mg), copper iodide (43 mg), and N,N-dimethylformamide (20 mL), N,N-diisopropylethylamine (1.55 mL) was added under an argon atmosphere followed by stirring at 80° C. for 1 hour. The reaction mixture was concentrated under reduced pressure, and to the obtained residue were added chloroform and water, and insoluble materials were removed by filtration through celite. After the filtrate was extracted with chloroform, the organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 5-[(2,6-difluoro-3,5-dimethoxyphenyl)ethynyl]pyrimidin-2-amine (1.07 g).

Preparation Example 8

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxyphenyl)ethynyl]pyrimidin-2-amine (400 mg), methanol (4 mL), and tetrahydrofuran (4 mL), 10% palladium-carbon (73 mg) was added under an argon atmosphere. After the resulting mixture was stirred at 60° C. for 8 hours under a hydrogen atmosphere, insoluble materials were removed by filtration through celite. The filtrate was concentrated under reduced pressure to give 5-[(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]pyrimidin-2-amine (402 mg).

Preparation Example 9

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]pyrimidin-2-amine (100 mg) and acetonitrile (2 mL) under an argon atmosphere were added copper chloride (II) (68 mg) and n-pentyl nitrite (69 μL), followed by stirring at 60° C. for 4 hours. To the reaction mixture, ethyl acetate was added and insoluble materials were removed by filtration. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 2-chloro-5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]pyrimidine (20 mg).

Preparation Example 10

A mixture of (2-chloropyrimidin-5-yl)methanol (120 mg), 3,5-dimethoxyphenol (186 mg), tributylphosphine (297 μL), and tetrahydrofuran (2.4 mL) was ice cooled, and 1,1′-(azodicarbonyl)dipiperidine (305 mg) was added thereto followed by stirring at room temperature for 12 hours. Insoluble materials were removed by filtration and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 2-chloro-5-[(3,5-dimethoxyphenoxy)methyl]pyrimidine (119 mg).

Preparation Example 13

A mixture of 2-chloro-5-hydroxypyrimidine (278 mg), potassium carbonate (453 mg), and N,N-dimethylformamide (3 mL) was ice cooled, and 3,5-dimethoxybenzyl bromide (541 mg) was added thereto followed by stirring at room temperature for 7 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. The resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 2-chloro-5-[(3,5-dimethoxybenzyl)oxy]pyrimidine (360 mg).

Preparation Example 14

To a mixture of 2-chloro-5-[(3,5-dimethoxybenzyl)oxy]pyrimidine (4.17 g) and N,N-dimethylformamide (40 mL), N-chlorosuccinimide (4.05 g) was added followed by stirring at room temperature for 2 hours and stirring at 60° C. for 2 hours. To the reaction mixture, water was added, and the resulting solid was collected by filtration, washed with water, and then dried under reduced pressure. The obtained solid was suspended in ethyl acetate (40 mL) and heated to 80° C. The solid was collected by filtration, and then dried under reduced pressure to give 2-chloro-5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyrimidine (3.99 g).

Preparation Example 15

A mixture of 2-chloro-5-hydroxypyrimidine (487 mg) and 1-(3,5-dimethoxyphenyl)ethanol (680 mg), tributylphosphine (1.37 mL), and tetrahydrofuran (14 mL) was ice cooled, and 1,1′-(azodicarbonyl)dipiperidine (1.4 g) was added thereto followed by stirring at room temperature for 12 hours and stirring at 50° C. for 3 hours. Insoluble materials were removed by filtration and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 2-chloro-5-[1-(3,5-dimethoxyphenyl)ethoxy]pyrimidine (415 mg).

Preparation Example 16

A mixture of methyl 3,5-dimethoxybenzoate (1 g) and acetonitrile (20 mL) was ice cooled, and N-fluoro-N′-(chloromethyl)triethylenediamine bis(tetrafluoroborate) (4.09 g) was added thereto followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, added anhydrous sodium sulfate and basic silica gel followed by stirring for 30 minutes, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give methyl 2,6-difluoro-3,5-dimethoxybenzoate (292 mg: Preparation Example 16-1) and methyl 2-fluoro-3,5-dimethoxybenzoate (232 mg: Preparation Example 16-2).

Preparation Example 17

A mixture of methyl 2,6-difluoro-3,5-dimethoxybenzoate (10 g) and tetrahydrofuran (50 mL) was ice cooled, and lithium borohydride (3.0M tetrahydrofuran solution, 43 mL) was added thereto followed by stirring at room temperature for 65 hours. The reaction mixture was ice cooled again, and additional lithium borohydride (3.0M tetrahydrofuran solution, 14 mL) was added thereto followed by stirring at room temperature for 22 hours. The reaction mixture was ice cooled and slowly added into ice water (300 mL). Further, concentrated hydrochloric acid (25 mL) was slowly added thereto, and the resulting mixture was stirred at room temperature for 1 hour and extracted with toluene/ethyl acetate (1:1). An organic layer obtained was washed with a saturated aqueous sodium hydrogen carbonate solution and saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure to give (2,6-difluoro-3,5-dimethoxyphenyl)methanol (8.67 g).

Preparation Example 18

A mixture of (2,6-difluoro-3,5-dimethoxyphenyl)methanol (1.71 g), triethylamine (2.57 mL), and tetrahydrofuran (34 mL) was ice cooled, and methanesulfonyl chloride (716 μL) was added thereto followed by stirring for 1 hour. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure to give 2,6-difluoro-3,5-dimethoxybenzyl methanesulfonate (2.32 g).

Preparation Example 19

To a mixture of 2-chloro-5-hydroxypyrimidine (4.38 g), potassium carbonate (9.27 g), and N,N-dimethylformamide (79 mL), 2,6-difluoro-3,5-dimethoxybenzyl methanesulfonate (7.89 g) was added followed by stirring at 60° C. for 1 hour. To the reaction mixture, water was added, and the resulting solid was collected by filtration, washed with water, and then dried under reduced pressure to give 2-chloro-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidine (8.53 g).

Preparation Example 20

A mixture of 2,3,5,6-tetrafluoropyridine (1.5 g) and methanol (15 mL) was ice cooled, and sodium methoxide (4.03 g) was added thereto followed by stirring at room temperature for 2 hours and stirring at 50° C. overnight. To the reaction mixture, water was added followed by extraction with diethyl ether. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure to give 3,5-difluoro-2,6-dimethoxypyridine (1.47 g).

Preparation Example 21

A mixture of diisopropylamine (745 μL) and tetrahydrofuran (5 mL) was cooled to −78° C., and n-butyl lithium (1.6M hexane solution, 3.02 mL) was added thereto followed by stirring at 0° C. for 30 minutes. The reaction mixture was cooled to −78° C., and a mixture of 3,5-difluoro-2,6-dimethoxypyridine (770 mg) and tetrahydrofuran (5 mL) was added thereto dropwise followed by stirring for 1 hour. After N,N-dimethylformamide (440 μL) was added thereto, the resulting mixture was warmed to room temperature and stirred for 1 hour. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 3,5-difluoro-2,6-dimethoxyisonicotinaldehyde (406 mg).

Preparation Example 22

A mixture of 3,5-difluoro-2,6-dimethoxyisonicotinaldehyde (400 mg) and methanol (4 mL) was ice cooled, and sodium borohydride (82 mg) was added thereto followed by stirring for 1 hour. To the reaction mixture, 1M hydrochloric acid was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated to give (3,5-difluoro-2,6-dimethoxypyridin-4-yl)methanol (403 mg).

Preparation Example 23

To a mixture of 2-chloro-5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyrimidine (235 mg), tert-butyl 4-(4-amino-3-methoxyphenyl)piperidine-1-carboxylate (306 mg), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphine) (138 mg), cesium carbonate (660 mg), and dioxane (10 mL), palladium acetate (30 mg) was added at room temperature under an argon atmosphere. The resulting mixture was stirred at 100° C. for 3 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give tert-butyl 4-[4-({5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-3-methoxyphenyl]piperidine-1-carboxylate (298 mg).

Preparation Example 24

To a mixture of 2-fluoro-5-nitrotoluene (500 mg), potassium carbonate (2.0 g), and N,N-dimethylformamide (15 mL), 4-piperidin-4-ylthiomorpholine 1,1-dioxide bistrifluoroacetate (2.16 g) was added followed by stirring at 80° C. for 20 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 4-[1-(2-methyl-4-nitrophenyl)piperidin-4-yl]thiomorpholine 1,1-dioxide (870 mg).

Preparation Example 25

To a mixture of 4-[1-(2-methyl-4-nitrophenyl)piperidin-4-yl]thiomorpholine 1,1-dioxide (1.5 g) and acetic acid (30 mL), 10% palladium-carbon (452 mg) was added under an argon atmosphere. After stirring for 13 hours under a hydrogen atmosphere, insoluble materials were removed by filtration through celite. The filtrate was concentrated under reduced pressure, and then a saturated aqueous sodium hydrogen carbonate solution was added to the resulting residue. The resulted solid was collected by filtration, washed with water, and then dried under reduced pressure to give 4-[4-(1,1-dioxidothiomorpholin-4-yl)piperidin-1-yl]-3-methylaniline (1.26 g).

Preparation Example 26

To a mixture of 1-chloro-2-(difluoromethoxy)-4-nitrobenzene (920 mg), potassium carbonate (1.7 g), and N,N-dimethylformamide (10 mL), 1-methyl-4-piperidin-4-ylpiperazine (1.13 g) was added followed by stirring at 100° C. overnight. The reaction mixture was concentrated under reduced pressure, and water was added to the resulting residue followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol/conc. aqueous ammonia solution) to give 1-{1-[2-(difluoromethoxy)-4-nitrophenyl]piperidin-4-yl}-4-methylpiperazine (1.38 g).

Preparation Example 27

To a mixture of 1-{1-[2-(difluoromethoxy)-4-nitrophenyl]piperidin-4-yl}-4-methylpiperazine (1.38 g) and ethanol (54 mL), 10% palladium-carbon (397 mg) was added under an argon atmosphere. After stirring for 1 hour under a hydrogen atmosphere, insoluble materials were removed by filtration through celite. The filtrate was concentrated under reduced pressure to give 3-(difluoromethoxy)-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]aniline (1.25 g).

Preparation Example 28

A mixture of benzyl piperazine-1-carboxylate (10 g), 2,2,6,6-tetramethylpiperidin-4-one (7.05 g), and dichloromethane (100 mL) was ice cooled, and sodium triacetoxy borohydride (11.5 g) was added thereto followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/hexane) to give benzyl 4-(2,2,6,6-tetramethylpiperidin-4-yl)piperazine-1-carboxylate (7.18 g).

Preparation Example 29

To a mixture of benzyl 4-(2,2,6,6-tetramethylpiperidin-4-yl)piperazine-1-carboxylate (7.18 g) and ethanol (60 mL), 10% palladium-carbon (2.0 g) was added under an argon atmosphere. After stirring for 7 hours under a hydrogen atmosphere, insoluble materials were removed by filtration through celite. The filtrate was concentrated under reduced pressure to give 1-(2,2,6,6-tetramethylpiperidin-4-yl)piperazine (4.35 g).

Preparation Example 30

To a mixture of 2-chloro-5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidine (202 mg), tert-butyl 9-(4-amino-2-methoxyphenyl)-3,9-diazaspiro[5,5]undecane-3-carboxylate (311 mg), 2-(dicyclohexylphosphino)-2′,4′,6′-triisopropyl-1,1′-biphenyl (30 mg), potassium carbonate (134 mg), and tert-butanol (10 mL), tris(dibenzylideneacetone)dipalladium (19 mg) was added at room temperature under an argon atmosphere. The resulting mixture was stirred at 100° C. for 4 hours. Insoluble materials were removed by filtration and washed with ethyl acetate. The filtrate was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give tert-butyl 4-[4-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-2-methoxyphenyl]-3,9-diazaspiro[5,5]undecane-3-carboxylate (259 mg).

Preparation Example 31

To a mixture of N-[3-(1,4-dioxa-8-azaspiro[4,5]dec-8-yl)phenyl]-5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-amine (596 mg), acetic acid (9 mL), and water (9 mL), concentrated hydrochloric acid (0.5 mL) was added followed by stirring at 80° C. for 7 hours. The reaction mixture was ice cooled, and a 1M aqueous sodium hydroxide solution (155 mL) and a saturated aqueous sodium hydrogen carbonate solution were added thereto, and then the resulting solid was collected by filtration. Chloroform was added thereto, and the resulting mixture was dried over anhydrous sodium sulfate and then filtered. The filtrate was concentrated under reduced pressure to give 1-[3-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperidin-4-one (512 mg).

Preparation Example 32

A mixture of 2-[3-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethanol (116 mg), triethylamine (84 μL), and tetrahydrofuran (4 mL) was ice cooled, and methanesulfonyl chloride (47 μL) was added thereto followed by stirring for 3 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. The filtrate was concentrated under reduced pressure to give 2-[3-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethyl methanesulfonate (129 mg).

Preparation Example 33

A mixture of 4-(4-nitro-1H-pyrazol-1-yl)piperidine (250 mg), 1-methylpiperidin-4-one (220 μL), and dichloromethane (5 mL) was ice cooled, and sodium triacetoxy borohydride (810 mg) was added thereto followed by stirring at room temperature for 4 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol/conc. aqueous ammonia solution) to give 1′-methyl-4-(4-nitro-1H-pyrazol-1-yl)-1,4′-bipiperidine (342 mg).

Preparation Example 34

To a mixture of 1-(2-chloro-4-nitrophenyl)-4-(1-methylpiperidin-4-yl)piperazine (3.7 g), ammonium chloride (352 mg), ethanol (94 mL), tetrahydrofuran (47 mL), and water (47 mL), iron powder (3.06 g) was added followed by stirring at 70° C. for 4 hours. After insoluble materials were removed by filtration, the filtrate was concentrated under reduced pressure. To the resulting residue, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was dried over anhydrous magnesium sulfate and then filtered. The filtrate was concentrated under reduced pressure to give 3-chloro-4-[4-(1-methylpiperidin-4-yl)piperazin-1-yl]aniline (1.03 g).

Preparation Example 35

To a mixture of (3R,5S)-1-(2-methoxy-4-nitrophenyl)-3,5,-dimethylpiperazine (3.0 g), N,N-diisopropylethylamine (2.32 mL), di-tert-butyldicarbonate (2.71 g), and dioxane (20 mL), 4-dimethylaminopyridine (69 mg) was added followed by stirring at 80° C. overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give tert-butyl (2R,6S)-4-(2-methoxy-4-nitrophenyl)-2,6-dimethylpiperazine-1-carboxylate (1.73 g).

Preparation Example 36

To a mixture of 2-(2-bromoethoxy)-1-chloro-4-nitrobenzene (3.0 g), cesium carbonate (5.23 g), N-methylpyrrolidone (30 mL), 1H-pyrazole (874 mg) was added followed by stirring at 60° C. for 6 hours. To the reaction mixture, water was added, and the resulting solid was collected by filtration. The solid was washed with water and dried under reduced pressure to give 1-[2-(2-chloro-5-nitrophenoxy)ethyl]-1H-pyrazole (2.57 g).

Preparation Example 37

To a mixture of 1-[2-(2-chloro-5-nitrophenoxy)ethyl]-1H-pyrazole (1.3 g), cesium carbonate (1.0 g), N-methylpyrrolidone (8 mL), cis-2,6-dimethylpiperazine (832 mg) was added followed by stirring at 130° C. overnight. To the reaction mixture, water was added, and the resulting solid was collected by filtration. The solid was washed with water and dried under reduced pressure to give (3R,5S)-3,5-dimethyl-1-{4-nitro-2-[2-(1H-pyrazol-1-yl)ethoxy]phenyl}piperazine (1.15 g).

Preparation Example 38

A mixture of 2-chloro-5-[(3,5-dimethoxybenzyl)oxy]pyridine (500 mg) and acetonitrile (10 mL) was ice cooled, and sulfuryl chloride (297 μL) was added thereto followed by stirring at room temperature for three days. After the reaction mixture was concentrated under reduced pressure, a saturated aqueous sodium hydrogen carbonate solution was added to the residue obtained. The resulting solid was collected by filtration, washed with water, and then dried under reduced pressure to give 2-chloro-5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyridine (596 mg).

Preparation Example 39

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine (3.6 g) and metanol (20 mL), a 4M hydrogen chloride/dioxane solution (40 mL) was added followed by stirring at room temperature for 6 hours. After the reaction mixture was concentrated under reduced pressure, a saturated aqueous sodium hydrogen carbonate solution was added to the residue obtained. The resulting solid was collected by filtration, washed with diethyl ether, and then dried under reduced pressure to give 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-(1 H-pyrazol-4-yl)pyrimidin-2-amine (2.9 g).

Preparation Example 40

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-(1H-pyrazol-4-yl)pyrimidin-2-amine (4.0 g), potassium carbonate (4.6 g), and N,N-dimethylformamide (80 mL), ethyl bromoacetate (2.4 mL) was added followed by stirring at 80° C. for 3 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give ethyl [4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]acetate (4.2 g).

Preparation Example 41

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-(1H-pyrazol-4-yl)pyrimidin-2-amine (50 mg), potassium carbonate (57 mg), and N,N-dimethylformamide (1 mL), [(4R)-2,2-dimethyl-1,3-dioxolan-4-yl]methyl 4-methylbenzenesulfonate (98 μL) was added followed by stirring at 60° C. for 1 hour and stirring at 110° C. for 4 days. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-(1-{[(4S)-2,2-dimethyl-1,3-dioxolan-4-yl]methyl}-1H-pyrazol-4-yl)pyrimidin-2-amine (45 mg).

Preparation Example 111

To a mixture of 4-nitro-1H-pyrazole (500 mg), tert-butyl (3-endo)-3-[(methylsulfonyl)oxy]-8-azabicyclo[3,2,1]octane-8-carboxylate (1.35 g) and N-methylpyrrolidone (6 mL), cesium carbonate (2.16 g) was added followed by stirring at 100° C. for 6 hours. To the reaction mixture, water was added, and the resulting solid was collected by filtration, washed with water, and then dried under reduced pressure to give tert-butyl (3-exo)-3-(4-nitro-1H-pyrazol-1-yl)-8-azabicyclo[3,2,1]octane-8-carboxylate (1.07 g).

Preparation Example 118

A mixture of 4-nitro-1H-pyrazol (3 g), quinuclidin-3-ol (4.05 g), triphenylphosphine (9.05 g), and tetrahydrofuran (60 mL) was ice cooled, and diisopropyl azodicarboxylate (6.84 mL) was added thereto followed by stirring at room temperature overnight. After the reaction mixture was concentrated under reduced pressure, 1M hydrochloric acid (50 mL) was added to the resulting residue. The aqueous layer obtained was washed with ethyl acetate, and then a 1M aqueous sodium hydroxide solution (60 mL) was added for basification. After extraction with chloroform, an organic layer obtained was dried over anhydrous sodium sulfate and then filtered. The filtrate was concentrated under reduced pressure and then the resulting residue was purified by basic silica gel column chromatography (ethyl acetate) to give 3-(4-nitro-1H-pyrazol-1-yl)quinuclidine (5.15 g).

Preparation Example 133

A mixture of tert-butyl (4-amino-2-methoxyphenyl)[2-(4-methylpiperazin-1-yl)ethyl]carbamate (1.21 g) and tetrahydrofuran (24 mL) was ice cooled, and lithium aluminum hydride (629 mg) was added thereto followed by stirring for 1 hour under heating to reflux. To the reaction mixture, water (0.63 mL), a 1M aqueous sodium hydroxide solution (0.63 mL), and water (1.89 mL) in that order were added. After insoluble materials were removed by filtration through celite, the filtrate was extracted with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (chloroform/methanol) to give 2-methoxy-N1-methyl-N1-[2-(4-methylpiperazin-1-yl)ethyl]benzene-1,4-diamine (922 mg).

Preparation Example 138

To a mixture of 2-chloro-5-nitropyrimidine (798 mg), potassium carbonate (1.04 g), and N,N-dimethylformamide (16 mL), 1-methyl-4-(piperidin-4-yl)piperazine (1.1 g) was added followed by stirring at room temperature for 3 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 2-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]-5-nitropyrimidine (542 mg).

Preparation Example 143

To a mixture of tert-butyl 4-(2-amino-1,3-thiazol-5-yl)piperidine-1-carboxylate (1.13 g) and ethyl acetate (8 mL), 4M hydrogen chloride/ethyl acetate solution (8 mL) was added followed by stirring at room temperature for 3 hours. The solvent was concentrated under reduced pressure to give 5-(piperidin-4-yl)-1,3-thiazol-2-amine hydrochloride (877 mg).

Preparation Example 144

To a mixture of 5-(piperidin-4-yl)-1,3-thiazol-2-amine hydrochloride (519 mg), dichloromethane (5 mL), and methanol (5 mL), 1H-benzotriazol-1-ylmethanol (423 mg), sodium acetate (388 mg), and sodium triacetoxy borohydride (1.0 g) in that order were added followed by stirring at room temperature for 2 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution and basic silica gel were added followed by concentration of the solvent under reduced pressure. The resulting residue was purified by basic silica gel column chromatography (chloroform/methanol) to give 5-(1-methylpiperidin-4-yl)-1,3-thiazol-2-amine (411 mg).

Preparation Example 145

A mixture of 5-nitropyridin-2(1H)-one (700 mg), (R)-2,2-dimethyl-1,3-dioxolane-4-methanol (661 mg), triphenylphosphine (1.97 g), and tetrahydrofuran (20 mL) was ice cooled, diisopropyl azodicarboxylate (1.49 mL) was added followed by stirring at room temperature for 5 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (R)-2-[(2,2-dimethyl-1,3-dioxolan-4-yl)methoxy]-5-nitropyridine (541 mg).

Preparation Example 152

To a mixture of (S)-2,2-dimethyl-1,3-dioxolane-4-methanol (661 mg) and N,N-dimethylformamide (23 mL), sodium hydride (218 mg) was added followed by stirring at room temperature for 10 minutes. To the reaction mixture, 2-chloro-5-nitropyridine (793 mg) was added followed by stirring at room temperature for 2 hours. After water was added to the reaction mixture, extraction with ethyl acetate was performed. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (S)-2-[(2,2-dimethyl-1,3-dioxolan-4-yl)methoxy]-5-nitropyridine (810 mg).

Preparation Example 162

To a mixture of 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethyl methanesulfonate (320 mg) and N-methylpyrrolidone (6 mL), tert-butyl piperazine-1-carboxylate (1.31 g) was added followed by stirring at 80° C. overnight and additional stirring at 120° C. overnight. To the reaction mixture, water and a saturated aqueous sodium hydrogen carbonate solution were added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate/diisopropyl ether to give tert-butyl 4-{2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethyl}piperazine-1-carboxylate (202 mg).

Preparation Example 175

A mixture of 5-methyl-1H-pyrazol-3-amine (522 mg) and N,N-dimethylformamide (10 mL) was ice cooled, and sodium hydride (473 mg) was added thereto followed by stirring for 30 minutes. To the reaction mixture, 2-(2-bromoethoxy)tetrahydro-2H-pyran (893 μL) was added followed by stirring at room temperature for 12 hours. After saturated aqueous ammonium chloride solution was added to the reaction mixture, extraction with ethyl acetate was performed. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 5-methyl-1-[2-(tetrahydro-2H-pyran-2-yloxy)ethyl]-1H-pyrazol-3-amine (427 mg: Preparation Example 175-1) and 3-methyl-1-[2-(tetrahydro-2H-pyran-2-yloxy)ethyl]-1H-pyrazol-5-amine (199 mg: Preparation Example 175-2).

Preparation Example 176

To a mixture of 5-[2-(benzyloxy)ethyl]-3-(2,5-dimethyl-1H-pyrrol-1-yl)-1-methyl-1H-pyrazole (640 mg) and ethanol (9.7 mL), hydroxylamine (1.37 mL) and p-toluenesulfonic acid monohydrate (1.95 g) in that order were added followed by stirring at 95° C. overnight. The reaction mixture was concentrated under reduced pressure, and then water was added to the resulting residue followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 5-[2-(benzyloxy)ethyl]-1-methyl-1H-pyrazol-3-amine (470 mg).

Preparation Example 183

To a mixture of (1-methyl-3-nitro-1H-pyrazol-5-yl)methanol (398 mg), 3,4-dihydro-2H-pyran (459 μL), and ethyl acetate (8 mL), p-toluenesulfonic acid monohydrate (96 mg) was added followed by stirring at room temperature for 1.5 hours. Additional 3,4-dihydro-2H-pyran (459 μL) and p-toluenesulfonic acid monohydrate (96 mg) were added thereto followed by stirring at room temperature for 1.5 hours. After water was added to the reaction mixture, extraction with ethyl acetate was performed. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 1-methyl-3-nitro-5-[(tetrahydro-2H-pyran-2-yloxy)methyl]-1H-pyrazole (487 mg).

Preparation Example 186

A mixture of 4-nitro-1H-pyrazole (300 mg), 2-phenyl-1,3-dioxan-5-ol (717 mg), triphenylphosphine (1.11 g) and tetrahydrofuran (4.5 mL) was ice cooled, and then diisopropyl azodicarboxylate (842 μL) was added thereto followed by stirring at room temperature for 12 hours. After the reaction mixture was concentrated under reduced pressure, the resulting residue was purified by silica gel chromatography (ethyl acetate/hexane) to give 4-nitro-1-(2-phenyl-1,3-dioxan-5-yl)-1H-pyrazol (121 mg).

Preparation Example 189

To a mixture of 5-nitropyridine-2-carbaldehyde (761 mg), 2-(piperazin-1-yl)ethanol (1.23 mL), acetic acid (570 μL), and dichloromethane (20 mL), sodium triacetoxy borohydride (2.23 g) was added followed by stirring at room temperature for 16 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform/2-propanol. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 2-{4-[(5-nitropyridin-2-yl)methyl]piperazin-1-yl}ethanol (726 mg).

Preparation Example 191

To a mixture of methyl 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylate (871 mg), ethanol (8.7 mL), and tetrahydrofuran (8.7 mL), a 1M aqueous sodium hydroxide solution (3.45 mL) was added followed by stirring at 60° C. for 2 hours. To the reaction mixture, 1M hydrochloric acid was added, and the resulting solid was collected by filtration, washed with water, and then dried under reduced pressure to give 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylic acid (846 mg).

Preparation Example 193

A mixture of 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carboxylic acid (300 mg) and dioxane (8.5 mL) was ice cooled, and 1,1′-carbonyldiimidazole (99 mg) was added thereto followed by stirring at room temperature for 2 hours and stirring at 60° C. for 2 hours. Additional 1,1′-carbonyldiimidazole (99 mg) was added thereto followed by stirring at 60° C. for 2 hours. Further, 1,1′-carbonyldiimidazole (297 mg) was added thereto followed by stirring at room temperature for 1 hour. The reaction mixture was ice cooled and sodium borohydride (230 mg) was added thereto followed by stirring at room temperature for 12 hours. Water was added to the reaction mixture and extraction with ethyl acetate was performed. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give [5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl]methanol (126 mg).

Preparation Example 201

To a mixture of 1-methyl-3-nitro-1H-pyrazole-5-carbaldehyde (850 mg) and tetrahydrofuran (50 mL), methyl (triphenylphosphoranylidene)acetate (3.66 g) was added followed by stirring at 60° C. for 3 hours. After the reaction mixture was concentrated under reduced pressure, water was added to the resulting residue followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the residue obtained was washed with chloroform and the resulting solid was collected by filtration. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then combined with the solid obtained earlier to give methyl (E)-3-(1-methyl-3-nitro-1H-pyrazol-5-yl)acrylate (1.15 g).

Preparation Example 202

Under an argon atmosphere, to a mixture of methyl (E)-3-(1-methyl-3-nitro-1H-pyrazol-5-yl)acrylate (1.15 g) and ethanol (50 mL) was added 10% palladium-carbon (580 mg). After stirring under a hydrogen atmosphere in 1 atm for 12 hours and in 2.7 atm for 4 hours, insoluble materials were removed by filtration through celite. The resulting filtrate was concentrated under reduced pressure to give methyl 3-(3-amino-1-methyl-1H-pyrazol-5-yl)propanoate (955 mg).

Preparation Example 204

To a mixture of 2-[(tert-butoxycarybonyl)amino]-1,3-thiazole-5-carboxylic acid (500 mg), N-[3-(diethylamino)propyl]-N′-ethylcarbodiimide hydrochloride (589 mg), 1H-benzotriazol-1-ol (415 mg), and N,N-dimethylformamide (10 mL), 1-methylpiperazine (451 μL) was added followed by stirring at room temperature for 3 days. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give tert-butyl {5-[(4-methylpiperazin-1-yl)carbonyl]-1,3-thiazol-2-yl}carbamate (560 mg).

Preparation Example 205

A mixture of 4-aminopyridin-2(1H)-one (400 mg) and N-methylpyrrolidone (15 mL) was ice cooled, and sodium hydride (218 mg) was added thereto followed by stirring at room temperature for 30 minutes. To the reaction mixture, (S)-2,2-dimethyl-1,3-dioxolan-4-ylmethyl p-toluenesulfonate (1.14 g) and sodium iodide (109 mg) in that order were added followed by stirring at room temperature for 4 hours. After sodium hydride (218 mg) was added to the reaction mixture followed by stirring at 80° C. overnight. To the reaction mixture, a saturated aqueous ammonium chloride solution was added, and then the resulting mixture was saturated with sodium chloride, and extraction with methanol/chloroform was performed. An organic layer obtained was dried over anhydrous sodium sulfate and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/methanol) to give (R)-4-amino-1-[(2,2-dimethyl-1,3-dioxolan-4-yl)methyl]pyridin-2(1H)-one (136 mg).

Preparation Example 209

A mixture of [5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl]methanol (126 mg), triethylamine (147 μL), dichloromethane (6 mL), and tetrahydrofuran (6 mL) was ice cooled, and then methanesulfonyl chloride (82 μL) was added thereto followed by stirring at room temperature for 3 hours. To the reaction mixture, N,N-dimethylformamide (6 mL) was added followed by stirring at room temperature for 12 hours. Water was added to the reaction mixture and extraction with chloroform was performed. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure to give N-[3-(chloromethyl)-1H-pyrazol-5-yl]-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-amine (109 mg).

Preparation Example 210

To a mixture of tert-butyl {5-[(4-methylpiperazin-1-yl)carbonyl]-1,3-thiazol-2-yl}carbamate (560 mg) and ethyl acetate (8 mL) was added 4M hydrogen chloride/ethyl acetate solution (8 mL) followed by stirring at room temperature for 3 hours. After the reaction mixture was concentrated under reduced pressure, the resulted residue was purified by basic silica gel chromatography (methanol/chloroform) to give (2-amino-1,3-thiazol-5-yl)(4-methylpiperazin-1-yl)methanone (357 mg).

Preparation Example 211

To a mixture of (5-nitro-1H-pyrazol-3-yl)methanol (1.86 g), 3,4-dihydro-2H-pyran (4.7 mL), and acetonitrile (28 mL), trifluoroacetic acid (40 μL) was added followed by stirring at 70° C. for 3 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 5-nitro-1-(tetrahydro-2H-pyran-2-yl)-3-[(tetrahydro-2H-pyran-2-yloxy)methyl]-1H-pyrazole (3.98 g).

Preparation Example 214

A mixture of [5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-3-yl]methanol (200 mg) and 1,2-dichloroethane (12 mL) was ice cooled, and manganese dioxide (442 mg) was added thereto followed by stirring at room temperature for 30 minutes and then stirring at 90° C. for 2 hours. After insoluble materials were removed by filtration, the filtrate was concentrated under reduced pressure to give 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazole-3-carbaldehyde (142 mg).

Preparation Example 229

A mixture of methyl 2-chloro-6-fluoro-3,5-dimethoxybenzoate (682 mg) and tetrahydrofuran (25 mL) was ice cooled, and lithium aluminum hydride (104 mg) was added thereto followed by stirring at room temperature for 3 hours. To the reaction mixture, diethylether was added for dilution under ice cooling, and then a saturated aqueous sodium sulfate solution was added thereto. Insoluble materials were separated by filtration and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (2-chloro-6-fluoro-3,5-dimethoxyphenyl)methanol (363 mg).

Preparation Example 232

To a mixture of 2-bromo-5-nitroanisole (3.15 g), tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-8-azabicyclo[3.2.1]oct-2-ene-8-carboxylate (5.00 g), and dioxane (40 mL), [1,1′-bis(diphenylphosphino)ferrocene]palladium dichloride dichloromethane complex (554 mg) and potassium carbonate (2.81 g) in that order were added under an argon atmosphere followed by stirring at 80° C. for 21 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give tert-butyl 3-(2-methoxy-4-nitrophenyl)-8-azabicyclo[3.2.1]oct-2-ene-8-carboxylate (2.78 g).

Preparation Example 252

A mixture of tert-butyl 4,4-bis(acetoxymethyl)-1,4′-bipiperidin-1′-carboxylate (712 mg) and dichloromethane (6 mL) was ice cooled, and then trifluoroacetic acid (3 mL) was added thereto followed by stirring at room temperature for 3 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with dichloromethane. An organic layer obtained was washed with brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure to give 1,4′-bipiperidin-4,4-diylbis(methylene) diacetate (529 mg).

Preparation Example 255

A mixture of tert-butyl 4,4-bis(hydroxymethyl)piperidine-1-carboxylate (1.01 g), triethylamine (861 μL), and dichloromethane (10 mL) was ice cooled, and acetic anhydride (950 μL) was added thereto followed by stirring for 2 hours. To the reaction mixture, water was added followed by extraction with dichloromethane. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give tert-butyl 4,4-bis(acetoxymethyl)piperidine-1-carboxylate (1.38 g).

Preparation Example 295

After a mixture of 2-[3-(2,5-dimethyl-1H-pyrrol-1-yl)-1-methyl-1H-pyrazol-5-yl]ethanol (630 mg), benzyl bromide (376 μL), and tetrahydrofuran (8 mL) was ice cooled, sodium hydride (173 mg) was added thereto followed by stirring at room temperature for 6 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 5-[2-(benzyloxy)ethyl]-3-(2,5-dimethyl-1H-pyrrol-1-yl)-1-methyl-1H-pyrazole (640 mg).

Preparation Example 296

After a mixture of 3-(2,5-dimethyl-1H-pyrrol-1-yl)-1-methyl-1H-pyrazole (2 g) and tetrahydrofuran (60 mL) was cooled to −78° C., n-butyl lithium (1.6M hexane solution, 8.56 mL) was added thereto followed by stirring for 2 hours. To the reaction mixture, oxirane (1.1M tetrahydrofuran solution, 15.6 mL) and borontrifluoride tetrahydrofuran complex (1.51 mL) were added followed by stirring for 30 minutes. After that, the mixture obtained was warmed to room temperature and stirred for 6 hours. To the reaction mixture, a saturated aqueous ammonium chloride solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) to give 2-[3-(2,5-dimethyl-1H-pyrrol-1-yl)-1-methyl-1H-pyrazol-5-yl]ethanol (630 mg).

The compounds shown in Tables 7 to 62 below were prepared in the same manner as in the preparation examples described above. Tables 7 to 62 also show the processes for preparing the compounds of the preparation examples and the structures and physical and chemical data of the compounds.

Example 1

To a mixture of 5-bromo-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (104 mg), 1-ethynyl-3,5-dimethoxybenzene (37 mg), tetrakistriphenylphosphine palladium (13 mg), copper iodide (4 mg), and N,N-dimethylformamide (2 mL), triethylamine (157 μL) was added under an argon atmosphere followed by stirring at 120° C. for 30 minutes. Further, a mixture of 1-ethynyl-3,5-dimethoxybenzene (146 mg) and N,N-dimethylformamide (1 mL) was added thereto followed by stirring at 120° C. for 2 hours. The reaction mixture was diluted with ethyl acetate, and insoluble materials were removed by filtration through celite. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and subsequently purified by basic silica gel column chromatography (ethyl acetate/methanol), and then solidified with ethyl acetate to give 5-[(3,5-dimethoxyphenyl)ethynyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (23 mg).

Example 2

To a mixture of 5-[(3,5-dimethoxyphenyl)ethynyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (72 mg), methanol (2 mL), and tetrahydrofuran (2 mL), 10% palladium-carbon (25 mg) was added under an argon atmosphere. After stirring for 4 hours under a hydrogen atmosphere (3 atm), insoluble materials were removed by filtration through celite. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with diethyl ether to give 5-[2-(3,5-dimethoxyphenyl)ethyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (17 mg).

Example 3

To a mixture of 5-iodo-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (100 mg), 2,4-dichloro-3-ethynyl-1,5-dimethoxybenzene (55 mg), tetrakistriphenylphosphine palladium (23 mg), copper iodide (2 mg), and N,N-dimethylformamide (2 mL), N,N-diisopropylethylamine (67 μL) was added under an argon atmosphere followed by stirring at 100° C. for 4 hours. The reaction mixture was diluted with ethyl acetate, and insoluble materials were removed by filtration through celite. To the filtrate, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and subsequently purified by basic silica gel column chromatography (ethyl acetate/methanol), and then solidified with ethyl acetate to give 5-[(2,6-dichloro-3,5-dimethoxyphenyl)ethynyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (56 mg).

Example 4

To a mixture of 5-[(2,6-dichloro-3,5-dimethoxyphenyl)ethynyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (92 mg) and ethyl acetate (6 mL), a 4M hydrogen chloride/ethyl acetate solution (1 mL) was added followed by stirring at room temperature for 4 hours. The resulting solid was collected by filtration and dried under reduced pressure to give 5-[(2,6-dichloro-3,5-dimethoxyphenyl)ethynyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine trihydrochloride (101 mg).

Example 5

A mixture of 5-[2-(3,5-dimethoxyphenyl)ethyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (131 mg) and acetonitrile (1.3 mL) was ice cooled, and then sulfuryl chloride (41 μL) was added thereto followed by stirring at room temperature for 12 hours. After the reaction mixture was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then washed with diisopropyl ether to give N-{2-chloro-5-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}-5-[2-(2,6-dichloro-3,5-dimethoxyphenyl)ethyl]pyrimidin-2-amine (29 mg).

Example 6

Under an argon atmosphere, a mixture of 5-[(2,6-difluoro-3,5-dimethoxyphenyl)ethynyl]-N-{3-methoxy-4-[4-(1-methylpiperidin-4-yl)piperazin-1-yl]phenyl}pyrimidin-2-amine (164 mg), 4-methylbenzenesulfonyl hydrazide (2.63 g), and 1,2-dimethoxyethane (3 mL) was stirred at 110° C., and a mixture of sodium acetate (1.16 g) and water (1 mL) was added thereto. After 2 hours, 4-methylbenzenesulfonyl hydrazide (1.32 g) was added thereto, and then an additional mixture of sodium acetate (581 mg) and water (1 mL) was added thereto followed by stirring at 110° C. for 2 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was dried over anhydrous magnesium sulfate and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/methanol/conc. aqueous ammonia solution) and then solidified with ethyl acetate/diisopropyl ether to give 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-{3-methoxy-4-[4-(1-methylpiperidin-4-yl)piperazin-1-yl]phenyl}pyrimidin-2-amine (114 mg).

Example 7

A mixture of 5-[(2,6-difluoro-3,5-dimethoxyphenyl)ethynyl]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine (100 mg), tetrahydrofuran (5 mL), and methanol (5 mL) was reacted using H-Cube (trademark) (10% palladium-carbon, 0.5 mL/min, 50° C., 1 atm). The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate/diisopropyl ether to give 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine (29 mg).

Example 8

To a mixture of ethyl [4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]acetate (292 mg), tetrahydrofuran (6 mL), and ethanol (6 mL), a 1M aqueous sodium hydroxide solution (1.3 mL) was added at room temperature followed by stirring for 5 hours. The reaction mixture was neutralized with 1M hydrochloric acid and the resulting solid was collected by filtration. The solid was washed with water and dried under reduced pressure to give [4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]acetic acid (267 mg).

Example 9

To a mixture of 2-chloro-5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]pyrimidine (56 mg), 1-(1-methylpiperidin-4-yl)-1H-pyrazol-4-amine (48 mg), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphine) (33 mg), cesium carbonate (174 mg), and dioxane (2.2 mL), palladium acetate (8 mg) was added at room temperature under an argon atmosphere followed by stirring at 100° C. for 4 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate/diisopropyl ether to give 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-[1-(1-methylpiperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine (43 mg).

Example 10

To a mixture of 1-(bromomethyl)-2,6-difluorobenzene (14 mg), 2-chloro-5-hydroxypyrimidine (9.1 mg), and N,N-dimethylformamide (1 mL), potassium carbonate (16 mg) was added followed by stirring at room temperature overnight. To the reaction mixture, water was added followed by extraction with chloroform. An organic layer obtained was concentrated under reduced pressure. To the resulting residue, a mixture of 3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]aniline (30 mg), cesium carbonate (65 mg), palladium acetate-X-Phos (Pd:P=1:2) ChemDose (trademark) tablet, and tert-butyl alcohol (0.5 mL) was added followed by stirring at 120° C. overnight under a nitrogen atmosphere. To the reaction mixture, water was added followed by extraction with chloroform. An organic layer obtained was concentrated under reduced pressure. The resulting residue was purified by HPLC (0.1% aqueous formic acid solution/methanol) to give 5-[(2,6-difluorobenzyl)oxy]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (17 mg).

Example 11

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-(1-{[(4S)-2,2-dimethyl-1,3-dioxolan-4-yl]methyl}-1H-pyrazol-4-yl)pyrimidin-2-amine (45 mg) and tetrahydrofuran (2 mL), 1M hydrochloric acid (1 mL) was added followed by stirring at 50° C. for 3 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate to give (2S)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol (27 mg).

Example 12

A mixture of tert-butyl 4-[4-({5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-3-methoxyphenyl]piperidin-1-carboxylate (298 mg) and chloroform (6 mL) was ice cooled, and trifluoroacetic acid (1 mL) was added thereto followed by stirring at room temperature for 4 hours. After the reaction mixture was ice cooled, a 1M aqueous sodium hydroxide solution (10 mL) and a saturated aqueous sodium hydrogen carbonate solution were added thereto for basification followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. The filtrate was concentrated under reduced pressure to give a crude product (273 mg). Further, the crude product (60 mg) was purified by silica gel chromatography (chloroform/methanol/conc. aqueous ammonia solution), and then solidified with ethyl acetate/diisopropyl ether to give 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[2-methoxy-4-(piperidin-4-yl)phenyl]pyrimidin-2-amine (23 mg).

Example 13

To a mixture of 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[2-methoxy-4-(piperidin-4-yl)phenyl]pyrimidin-2-amine (63 mg), dichloromethane (2 mL), and methanol (1 mL), 1H-benzotriazol-1-ylmethanol (20 mg) was added followed by stirring at room temperature for 1 hour. Subsequently, sodium triacetoxy borohydride (51 mg) was added thereto followed by stirring at room temperature for 2 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added and extraction with chloroform was performed. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (chloroform/methanol/conc. aqueous ammonia solution) and then solidified with ethyl acetate/diisopropyl ether to give 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[2-methoxy-4-(1-methylpiperidin-4-yl)phenyl]pyrimidin-2-amine (28 mg).

Example 14

To a mixture of 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine (200 mg), ethanol (3 mL), and N,N-dimethylformamide (3 mL), 2,2-dimethyloxyrane (112 μL) was added followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by basic silica gel column chromatography (ethyl acetate) and then solidified with ethyl acetate to give 1-{4-[4-({5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]piperidin-1-yl}-2-methylpropan-2-ol (93 mg).

Example 15

A mixture of 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine (150 mg), triethylamine (131 μL), and dichloromethane (4 mL) was ice cooled, and cyclopropanecarbonyl chloride (29 μL) was added thereto followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was solidified with ethyl acetate to give cyclopropyl {4-[4-({5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]piperidin-1-yl}methanone (159 mg).

Example 16

To a mixture of 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine (150 mg), potassium carbonate (130 mg), and N,N-dimethylformamide (4 mL), 2-bromoethyl methyl ether (32 μL) was added followed by stirring at room temperature overnight and stirring at 60° C. for 3 hours. Additional 2-bromoethyl methyl ether (12 μL) was added thereto followed by stirring at 60° C. for 4 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/hexane) and then solidified with ethyl acetate to give 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-{1-[1-(2-methoxyethyl)piperidin-4-yl]-1H-pyrazol-4-yl}pyrimidin-2-amine (41 mg).

Example 17

To a mixture of ethyl 1-methyl-5-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-3-carboxylate (663 mg), ethanol (6.6 mL), and tetrahydrofuran (6.6 mL), a 1M aqueous sodium hydroxide solution (3.2 mL) was added followed by stirring at room temperature for 4 hours. To the reaction mixture, 1M hydrochloric acid (3.2 mL) was added, and the resulting solid was collected by filtration, washed with water, and dried under reduced pressure to give 1-methyl-5-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-3-carboxylic acid (464 mg).

Example 18

To a mixture of 1-methyl-5-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-3-carboxylic acid (100 mg), 1-methylpiperazine (83 μL), 1H-benzotriazol-1-ol (68 mg), and N,N-dimethylformamide (2 mL), N-[3-(diethylamino)propyl]-N′-ethylcarbodiimide hydrochloride (97 mg) was added followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. The filtrate was concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (chloroform/methanol/conc. aqueous ammonia solution) to give (4-methylpiperazin-1-yl)[1-methyl-5-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-3-yl]methanone (79 mg).

Example 19

A mixture of tert-butyl 4-[4-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-2-methoxyphenyl]-3,9-diazaspiro[5,5]undecane-3-carboxylate (232 mg) and dichloromethane (3 mL) was ice cooled, and trifluoroacetic acid (0.5 mL) was added thereto followed by stirring at room temperature for 1 hour. After the reaction mixture was concentrated under reduced pressure, ethyl acetate and a saturated aqueous sodium hydrogen carbonate solution were added to the resulting residue. The resulting solid was collected by filtration, washed with ethyl acetate, and then dried under reduced pressure to give N-[4-(3,9-diazaspiro[5,5]undec-3-yl)-3-methoxyphenyl]-5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-amine (167 mg).

Example 20

To a mixture of 2-[3-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethyl methanesulfonate (160 mg) and N-methylpyrrolidone (6 mL), 1-methylpiperazine (382 μL) was added followed by stirring at 80° C. for 2 hours. To the reaction mixture, water and a saturated aqueous sodium hydrogen carbonate solution were added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (chloroform/methanol/conc. aqueous ammonia solution) and then solidified with ethyl acetate/diisopropyl ether to give N-{1-[2-(4-methylpiperazin-1-yl)ethyl]-1H-pyrazol-3-yl}-5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-amine (64 mg).

Example 21

A mixture of N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]-5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-amine (100 mg) and chloroform (4 mL) was ice cooled, and m-chloroperbenzoic acid (43 mg) was added thereto followed by stirring at 4 to 10° C. for 3 hours and stirring at room temperature for 2 hours. To the reaction mixture, an aqueous sodium thiosulfate solution was added, and the resulting mixture was stirred at room temperature for 1 hour followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by basic silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate/diisopropyl ether to give N-[3-methoxy-4-(4-methyl-4-oxidopiperazin-1-yl)phenyl]-5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-amine (16 mg).

Example 22

To a mixture of 2-chloro-5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyridine (100 mg), 3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]aniline (87 mg), 2-(dicyclohexylphosphino)-2′,4′,6′-triisopropyl-1,1′-biphenyl (27 mg), sodium tert-butoxide (41 mg), and N-methylpyrrolidone (3 mL), palladium acetate (6.4 mg) was added under an argon atmosphere followed by stirring at 160° C. for 2 hours under microwave irradiation. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by basic silica gel column chromatography (chloroform/methanol) to give 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyridin-2-amine (27 mg).

Example 23

A mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-(1H-pyrazol-4-yl)pyrimidin-2-amine (200 mg), cesium carbonate (215 mg), (2S)-2-methyloxylane (128 mg), and N-methylpyrrolidone (4 mL) was stirred at 130° C. for 30 minutes under microwave irradiation. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnisium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (ethyl acetate/hexane) and then washed with ethyl acetate/diisopropyl ether to give (2S)-1-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propan-2-ol (171 mg).

Example 24

To a mixture of [4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]acetic acid (50 mg), ammonium chloride (25 mg), triethylamine (66 μL), 1H-benzotriazol-1-ol (32 mg) and N,N-dimethylformamide (1 mL) was added N-[3-(diethylamino)propyl]-N′-ethylcarbodiimide hydrochloride (45 mg) followed by stirring at room temperature for 12 hours. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was concentrated under reduced pressure, and the resulted residue was solidified with diisopropyl ether to give 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]acetamide (48 mg).

Example 64

To a mixture of 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[2-methoxy-4-(piperidin-4-yl)phenyl]pyrimidin-2-amine (62 mg), acetone (118 μL), and dichloromethane (3 mL), sodium triacetoxy borohydride (51 mg) was added followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol/conc. aqueous ammonia solution) and then solidified with ethyl acetate/diisopropyl ether to give 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[4-(1-isopropylpiperidin-4-yl)-2-methoxyphenyl]pyrimidin-2-amine (14 mg).

Example 106

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{4-(piperazin-1-yl)-3-[2-(1H-pyrazol-1-yl)ethoxy]phenyl}pyrimidin-2-amine (229 mg), formaldehyde (37% aqueous solution, 164 μL), acetic acid (231 μL), and dichloromethane (6 mL), sodium triacetoxy borohydride (257 mg) was added followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/methanol) and then solidified with ethyl acetate/diisopropyl ether to give 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{4-(4-methylpiperazin-1-yl)-3-[2-(1H-pyrazol-1-yl)ethoxy]phenyl}pyrimidin-2-amine (72 mg).

Example 120

To a mixture of 1-[3-({5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperidin-4-one (209 mg), 1-methylpiperazine (103 μL), and dichloromethane (4 mL), sodium triacetoxy borohydride (298 mg) was added followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol/conc. aqueous ammonia solution) and then solidified with ethyl acetate/diisopropyl ether to give N-{3-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}-5-[(2,3,5,6-tetrafluorobenzyl)oxy]pyrimidin-2-amine (98 mg).

Example 161

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[4-(piperidin-4-yl)phenyl]pyrimidin-2-amine (52 mg), glycolic acid (26 mg), 1H-benzotriazol-1-ol (31 mg), and N,N-dimethylformamide (1 mL), N-[3-(diethylamino)propyl]-N′-ethylcarbodiimide hydrochloride (44 mg) was added followed by stirring at room temperature for 2 days. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate/diisopropyl ether to give 1-{4-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperidin-1-yl}-2-hydroxyethanone (10 mg).

Example 162

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine (123 mg) and ethanol (3 ml), fumaric acid (26 mg) was added followed by heating to reflux. To the reaction mixture, water was added followed by stirring at room temperature overnight, and the resulting solid was collected by filtration. The solid was washed with ethanol and then dried under reduced pressure to give 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine hemifumarate (82 mg).

Example 166

To a mixture of tert-butyl 4-[4-({5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]piperidine-1-carboxylate (276 mg) and ethyl acetate (2 mL), a 4M hydrogen chloride/ethyl acetate solution (2 mL) was added followed by stirring at room temperature for 3 hours. After the reaction mixture was concentrated under reduced pressure, a saturated aqueous sodium hydrogen carbonate solution was added to the resulting residue followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate/diisopropyl ether to give 5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine (119 mg).

Example 190

To a mixture of tert-butyl {2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethyl}carbamate (101 mg) and ethyl acetate (2 mL), a 4M hydrogen chloride/ethyl acetate solution (2 mL) was added followed by stirring at room temperature for 3 hours. The resulting solid was collected by filtration and then dried under reduced pressure to give N-[1-(2-aminoethyl)-1H-pyrazol-4-yl]-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-amine trihydrochloride (100 mg).

Example 212

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-[3-(tetrahydro-2H-pyran-2-yloxy)propyl]-1H-pyrazol-4-yl}pyrimidin-2-amine (1.6 g), tetrahydrofuran (6.9 mL), and water (3.4 mL), acetic acid (13.8 mL) was added followed by stirring at 70° C. for 2 days. After the reaction mixture was concentrated under reduced pressure, a saturated aqueous sodium hydrogen carbonate solution was added to the resulting residue followed by extraction with chloroform. An organic layer obtained was dried over anhydrous magnesium sulfate and then filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was dissolved in methanol (30 mL). Potassium carbonate (656 mg) was added thereto followed by stirring at 60° C. for 5 hours. To the reaction mixture, water was added and extraction with chloroform was performed. An organic layer obtained was dried over anhydrous magnesium sulfate and then filtered. The filtrate was concentrated under reduced pressure to give 3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propan-1-ol (510 mg).

Example 213

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{6-[(2-phenyl-1,3-dioxan-5-yl)oxy]pyridin-3-yl}pyrimidin-2-amine (335 mg) and acetic acid (10 mL), water (2 mL) was added followed by stirring at 60° C. for 16 hours. After the solvent was concentrated under reduced pressure, a saturated aqueous sodium hydrogen carbonate solution was added to the resulting residue followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate to give 2-{[5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)pyridin-2-yl]oxy}propane-1,3-diol (92 mg).

Example 214

To a mixture of 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{6-[2-(tetrahydro-2H-pyran-2-yloxy)ethoxy]pyridin-3-yl}pyrimidin-2-amine (1.49 g) and methanol (5 mL), a 4M hydrogen chloride/dioxane solution (5 mL) was added followed by stirring at room temperature for 2 hours. After the reaction mixture was concentrated under reduced pressure, a saturated aqueous sodium hydrogen carbonate solution was added to the resulting residue followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. The filtrate was concentrated under reduced pressure, and the resulted residue was solidified with ethyl acetate. The solid was collected by filtration to give 2-{[5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)pyridin-2-yl]oxy}ethanol (452 mg). Further the filtrate was purified by silica gel column chromatography (chloroform/methanol) to give the product (701 mg).

Example 217

To a mixture of 1-[5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)pyridin-2-yl]piperidin-4-one (256 mg), 2-aminoethanol (131 μL), acetic acid (200 μL), and dichloromethane (9.3 mL), sodium triacetoxy borohydride (243 mg) was added followed by stirring at room temperature overnight. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform/2-propanol. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (chloroform/methanol) and then solidified with ethyl acetate to give 2-({1-[5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)pyridin-2-yl]piperidin-4-yl}amino)ethanol (137 mg).

Example 239

A mixture of N-{5-[2-(benzyloxy)ethyl]-1-methyl-1H-pyrazol-3-yl}-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-amine (283 mg) and dichloromethane (47 mL) was cooled to −78° C., and boron tribromide (1.0M dichloromethane solution, 830 μL) was added thereto followed by stirring at −78° C. for 1 hour and stirring at 0° C. for 1 hour. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 2-[3-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-methyl-1H-pyrazol-5-yl]ethanol (38 mg).

Example 246

A mixture of 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-methyl-1H-pyrazol-3-carboxylic acid (320 mg) and dioxane (6 mL) was ice cooled, and 1,1′-carbonyldiimidazole (616 mg) was added thereto followed by stirring at room temperature for 2 hours. To the reaction mixture, sodium borohydride (287 mg) was added followed by stirring at room temperature for 12 hours. To the reaction mixture, water and chloroform were added, and insoluble materials were removed by filtration through celite, and then the filtrate was extracted with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give [5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-methyl-1H-pyrazol-3-yl]methanol (125 mg).

Example 253

To a mixture of 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-(2-hydroxyethyl)pyridin-2(1H)-one (90 mg), triethylamine (50 μL), and dichloromethane (3 mL), methanesulfonyl chloride (20 μL) was added followed by stirring at room temperature for 1 hour. To the reaction mixture, 1-methylpiperazine (50 μL) and N,N-dimethylformamide (3 mL) were added followed by stirring at 50° C. for 20 hours. To the reaction mixture, water was added and extraction with ethyl acetate was performed. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/chloroform) and then solidified with diethyl ether to give 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1-[2-(4-methylpiperazin-1-yl)ethyl]pyridin-2(1H)-one (28 mg).

Example 254

To a mixture of 2-chloro-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidine (270 mg), 1-(1-methylpiperidin-4-yl)-1H-imidazol-4-amine (231 mg), 1,1′-binaphthalene-2,2′-diylbis(diphenylphosphine) (80 mg), cesium carbonate (556 mg), and dioxane (5.4 mL), palladium acetate (19 mg) was added under an argon atmosphere followed by stirring at 150° C. for 30 minutes under microwave irradiation. To the reaction mixture, water was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethanol/diethyl ether to give 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[1-(1-methylpiperidin-4-yl)-1H-imidazol-4-yl]pyrimidin-2-amine (241 mg).

Example 278

To a mixture of 2-chloro-5-[(2-fluoro-3,5-dimethoxybenzyl)oxy]pyrimidine (200 mg), 2-(4-amino-1H-pyrazol-1-yl)ethanol (170 mg), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (39 mg), cesium carbonate (655 mg), and dioxane (4 mL), tris(dibenzylideneacetone)dipalladium (31 mg) was added under an argon atmosphere followed by stirring at 80° C. overnight. To the reaction mixture, water was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (ethyl acetate/methanol) and then solidified with ethanol to give 2-[4-({5-[(2-fluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethanol (58 mg).

Example 282

To a mixture of 5-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-3-carbaldehyde (100 mg), morpholine (67 μL), and N,N-dimethylformamide (2 mL), sodium triacetoxy borohydride (243 mg) was added followed by stirring at room temperature for 12 hours. To the reaction mixture, a saturated aqueous sodium hydrogen carbonate solution was added followed by extraction with chloroform. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by basic silica gel column chromatography (chloroform/methanol) and purified by silica gel column chromatography (chloroform/methanol) and then solidified with ethanol/diisopropyl ether to give 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[3-(morpholin-4-ylmethyl)-1H-pyrazol-5-yl]pyrimidin-2-amine (42 mg).

Example 286

To a mixture of 2-chloro-5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]pyrimidine (159 mg), 3-methoxy-4-(1-methylpiperidin-4-yl)aniline (100 mg), and tert-butanol (5 mL), tris(dibenzylideneacetone)dipalladium (13 mg), 2-(dicyclohexylphosphino)-2′,4′,6′-triisopropyl-1,1′-biphenyl (20 mg), and potassium carbonate (88 mg) were added under an argon atmosphere followed by stirring at 100° C. for 8 hours. Insoluble materials were removed by filtration, washed with ethyl acetate, and then the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[3-methoxy-4-(1-methylpiperidin-4-yl)phenyl]pyrimidin-2-amine (35 mg).

Example 302

To a mixture of 2-{4-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperazin-1-yl}ethyl methanesulfonate (100 mg) and methanol (3 mL), sodium methoxide (25% methanol solution, 3 mL) was added followed by stirring at 90° C. for 15 minutes under microwave irradiation. After the reaction mixture was concentrated under reduced pressure, water was added to the resulting residue followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{4-[4-(2-methoxyethyl)piperazin-1-yl]phenyl}pyrimidin-2-amine (46 mg).

Example 315

To a mixture of {1′-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-2-methoxyphenyl]-1,4′-bipiperidine-4,4-diyl}bis(methylene)diacetate (46 mg) and methanol (3 mL), sodium methoxide (25% methanol solution, 0.2 mL) was added followed by stirring at room temperature for 14 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give {1′-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-2-methoxyphenyl]-1,4′-bipiperidine-4,4-diyl}dimethanol (34 mg).

Example 336

A mixture of ethyl 4-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-2-fluorophenyl]butanoate (150 mg) and tetrahydrofuran (3 mL) was ice cooled, and lithium aluminum hydride (11 mg) was added thereto followed by stirring at room temperature for 3 hours. The reaction mixture was diluted with diethyl ether under ice cooling and then a saturated aqueous sodium sulfate solution was added thereto. Insoluble materials were separated by filtration and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 4-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-2-fluorophenyl]butan-1-ol (70 mg).

Example 349

A mixture of ethyl 4-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-2-fluorophenyl]butanoate (150 mg) and tetrahydrofuran (3 mL) was ice cooled, and then methyl magnesium bromide (1.0M tetrahydrofuran solution, 1.2 mL) was added thereto followed by stirring for 3 hours. To the reaction mixture, a saturated aqueous ammonium chloride solution was added followed by extraction with ethyl acetate. An organic layer obtained was washed with saturated brine, dried over anhydrous sodium sulfate, and then filtered. After the filtrate was concentrated under reduced pressure, the resulting residue was purified by silica gel column chromatography (chloroform/methanol) to give 5-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-2-fluorophenoxy]-2-methylpentan-2-ol (104 mg).

Example 356

To a mixture of 2-(4-aminophenoxy)-2-methylpropionic acid (14.6 mg), cesium carbonate (49 mg), 2-chloro-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidine (15.8 mg) and tert-butanol (0.5 mL) was added palladium(II) acetate-2-(dicyclohexylphosphino)-2′,4′,6′-triisopropyl-1,1′-biphenyl (Pd:P 1:2) ChemDose (trademark) tablet under a nitrogen atmosphere followed by stirring at 120° C. overnight. To the reaction mixture, water was added followed by extraction with chloroform (2 mL), and then the solvent was concentrated under reduced pressure. The resulting residue was purified by HPLC (0.1% aqueous formic acid solution/methanol) to give 2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)phenoxy]-2-methylpropionic acid (0.7 mg).

Example 375

To a mixture of 4-amino-1-(1-tert-butoxycarbonyl-azetidin-3-yl)-1H-pyrazole (17.9 mg), cesium carbonate (49 mg), 2-chloro-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidine (15.8 mg), tert-butanol (0.34 mL) and N,N-dimethylformamide (0.16 mL) was added palladium(II) acetate-2-(dicyclohexylphosphino)-2′,4′,6′-triisopropyl-1,1′-biphenyl (Pd:P 1:2) ChemDose (trademark) tablet under a nitrogen atmosphere followed by stirring at 120° C. overnight. To the reaction mixture, water was added followed by extraction with chloroform (2 mL), and then the solvent was concentrated under reduced pressure. To the resulting residue, ethanol (1 mL) and a 4M hydrogen chloride/ethyl acetate solution (0.5 mL) were added followed by stirring at room temperature overnight. After that, the solvent was concentrated under reduced pressure. The resulting residue was purified by HPLC (0.1% aqueous formic acid solution/methanol) to give N-[1-(azetidin-3-yl)-1H-pyrazol-4-yl]-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-amine (1.7 mg).

In the same manner as in the examples shown above, the compounds shown in Tables 63 to 137 below were prepared. Tables 63 to 137 show the structures of the compounds of the examples and Tables 138 to 156 show the preparation processes and physical and chemical data of the compounds of the examples.

TABLE 7 PEx PSyn Str DAT 1 1 APCI/ESI+: 461,463 2 2 APCI/ESI+: 509 3 3 NMR2: 3.68(1H, s), 3.92(6H, s), 6.58 (1H, s) 4-1 4 APCI/ESI+: 199 4-2 4 APCI/ESI+: 181 5 5 NMR2: 3.60(1H, S), 3.89(3H, S), 3.91 (3H, S), 6.61(1H, d, J = 7.5 Hz)

TABLE 8 PEx PSyn Str DAT 6 6 EI: 168 7 7 APCI/ ESI+: 292 8 8 APCI/ ESI+: 296 9 9 APCI/ ESI+: 315 10 10 APCI/ ESI+: 281

TABLE 9 PEx PSyn Str DAT 13 13 APCI/ ESI+: 281 14 14 APCI/ ESI+: 349 15 15 ESI+: 295 16-1 16 ESI+: 233 16-2 16 ESI+: 215

TABLE 10 PEx PSyn Str DAT 17 17 ESI+: 205 18 18 NMR2: 3.04(3H, s), 3.88(6H, s), 5.34(2H, s), 6.72 (1H, t, J = 8.2 Hz) 19 19 APCI/ESI+: 317 20 20 APCI/ESI+: 176 21 21 NMR2: 4.03(6H, s), 10.3(1H, s) 22 22 APCI/ESI+: 206 23 23 APCI/ESI+: 619

TABLE 11 PEx PSyn Str DAT 24 24 ESI+: 354 25 25 ESI+: 324 26 26 ESI+: 371 27 27 APCI/ESI+: 341 28 28 NMR2: 1.10-1.19(2H, m), 1.12(6H, s), 1.19(6H, s), 1.69- 1.76(2H, m), 2.27(1H, brs), 2.55-2.59(4H, m), 2.76-2.87 (1H, m), 3.48-3.55(4H, m), 5.13(2H, s), 7.23-7.40(5H, m) 29 29 APCI/ESI+: 226

TABLE 12 PEx PSyn Str DAT 30 30 ESI+: 632 31 31 ESI+: 447 32 32 APCI/ESI+: 462 33 33 APCI/ESI+: 294 34 34 ESI+: 309 35 35 APCI/ESI+: 366

TABLE 13 PEx PSyn Str DAT 36 36 APCI/ESI+: 268 37 37 APCI/ESI+: 346 38 38 APCI/ESI+: 348 39 39 APCI/ESI+: 364 40 40 APCI/ESI+: 450 41 41 APCI/ESI+: 478

TABLE 14 PEx PSyn Str DAT 42 2 APCI/ESI+: 497 43 2 APCI/ESI+: 509 44 6 EI: 138 45 6 EI: 174 46 14 APCI/ESI+: 363 47 18 APCI/ESI+: 284

TABLE 15 PEx PSyn Str DAT 48 19 APCI/ESI+: 318 49 13 APCI/ESI+: 293 50 24 NMR2: 1.06-1.26 (14H, m), 1.77-1.84(2H, m), 2.74-2.91(6H, m), 3.22-3.31(4H, m), 3.95 (3H, s), 6.88(1H, d, J = 8.8 Hz), 7.70(1H, s), 7.86(1H, d, J = 8.8 Hz) 51 27 APCI/ESI+: 347 52 23 APCI/ESI+: 579 53 23 APCI/ESI+: 426

TABLE 16 PEx PSyn Str DAT 54 24 NMR2: 1.47- 1.71(17H, m), 3.17-3.20(4H, m), 3.40-3.43(4H, m), 3.95(3H, s), 6.89(1H, d, J = 8.8 Hz), 7.69(1H, d, J = 2.7 Hz), 7.85 (1H, dd, J = 8.8, 2.4 Hz) 55 27 NMR2: 1.46- 1.87(13H, m), 2.88-2.90(4H, m), 3.38-3.41(4H, m), 3.73-3.81(6H, m), 6.23-6.26 (2H, m), 6.78(1H, d, J = 8.1 Hz) 56 24 ESI+: 320 57 27 ESI+: 290 58 23 ESI+: 491

TABLE 17 PEx PSyn Str DAT 59 23 ESI+: 549 60 24 NMR2: 1.45 (9H, s), 3.84(3H, s), 4.10(4H, s), 4.26 (4H, s), 6.22 (1H, d, J = 9.0 Hz), 7.60(1H, d, J = 2.4 Hz), 7.82(1H, dd, J = 8.8, 2.4 Hz) 61 27 NMR2: 1.44(9H, s), 3.40(2H, brs), 3.74(3H, s), 3.87 (4H, s), 4.05(4H, s), 6.21-6.31(3H, m) 62 30 ESI+: 576 63 27 APCI/ESI+: 264 64 33 APCI/ESI+: 281 65 27 ESI+: 251

TABLE 18 PEx PSyn Str DAT 66 24 ESI+: 330 67 27 ESI+: 300 68 27 ESI+: 311 69 24 ESI+: 339 70 24 APCI/ESI+: 266 71 27 APCI/ESI+: 336

TABLE 19 PEx PSyn Str DAT 72 23 APCI/ESI+: 616 73 28 APCI/ESI+: 332 74 35 APCI/ESI+: 432 75 29 APCI/ESI+: 298 76 24 APCI/ESI+: 449 77 27 APCI/ESI+: 419

TABLE 20 PEx PSyn Str DAT 78 23 APCI/ESI+: 699 79 24 NMR2: 1.23 (6H, d, J = 6.3 Hz), 2.44-2.50 (2H, m), 3.50- 3.52(2H, m), 3.85-3.90(2H, m), 3.95(3H, s), 6.85(1H, d, J = 9.0 Hz), 7.71 (1H, d, J = 2.4 Hz), 7.85(1H, dd, J = 8.8, 2.7 Hz) 80 27 NMR2: 1.20(6H, d, J = 6.3 Hz), 2.27-2.32(2H, m), 3.15-3.18 (2H, m), 3.49 (2H, brs), 3.82- 3.93(5H, m), 6.23-6.27(2H, m), 6.74(1H, d, J = 8.1 Hz) 81 30 ESI+: 656 82 30 ESI+: 600

TABLE 21 PEx PSyn Str DAT 83 24 ESI+: 349 84 27 ESI+: 319 85 28 ESI+: 338 86 27 ESI+: 308 87 35 APCI/ESI+: 446 88 27 APCI/ESI+: 416

TABLE 22 PEx PSyn Str DAT 89 23 APCI/ESI+: 696 90 37 APCI/ESI+: 418 91 27 APCI/ESI+: 388 92 23 APCI/ESI+: 668 93 36 APCI/ESI+: 295

TABLE 23 PEx PSyn Str DAT 94 37 APCI/ESI+: 373 95 35 APCI/ESI+: 473 96 27 APCI/ESI+: 443 97 23 ESI+: 723

TABLE 24 PEx PSyn Str DAT 98 30 ESI+: 688 99 30 ESI+: 632 100 13 APCI/ESI+: 280 101 2 APCI/ESI+: 509 102 2 APCI/ESI+: 426 103 2 APCI/ESI+: 456

TABLE 25 PEx PSyn Str DAT 104 32 APCI/ESI+: 486 105 23 APCI/ESI+: 448 106 41 APCI/ESI+: 478 107 19 ESI+: 317 108 32 APCI/ESI+: 462

TABLE 26 PEx PSyn Str DAT 109 23 APCI/ESI+: 545 110 31 APCI/ESI+: 501 111 111 NMR2: 1.50 (9H, s), 1.72-1.81 (2H, m), 1.95-2.24 (6H, m), 4.32- 4.49(2H, m), 4.61- 4.72(1H, m), 8.06(1H, s), 8.12 (1H, s) 112 27 NMR2: 1.49 (9H, s), 1.71-1.81 (2H, m), 1.90-2.12 (6H, m), 2.87 (2H, brs), 4.20-4.45 (2H, m), 4.50- 4.61(1H, m), 6.98 (1H, s), 7.12 (1H, s)

TABLE 27 PEx PSyn Str DAT 113 23 APCI/ESI+: 605 114 23 APCI/ESI+: 523 115 23 APCI/ESI+: 547 116 23 ESI+: 557 117 23 APCI/ESI+: 515

TABLE 28 PEx PSyn Str DAT 118 118 APCI/ESI+: 223 119 27 APCI/ESI+: 193 120 23 APCI/ESI+: 558 121 23 APCI/ESI+: 576 122 23 APCI/ESI+: 614 123 23 APCI/ESI+: 543

TABLE 29 PEx PSyn Str DAT 124 31 APCI/ESI+: 499 125 24 APCI/ESI+: 295 126 17 NMR2: 1.74- 1.80(1H, m), 3.79 (3H, s), 3.86 (3H, s), 4.74(2H, dd, J = 6.2, 1.2 Hz), 6.47(1H, dd, J = 7.0, 3.0 Hz), 6.51 (1H, dd, J = 4.8, 3.0 Hz) 127 23 APCI/ESI+: 586 128 35 APCI/ESI+: 395 129 27 APCI/ESI+: 365

TABLE 30 PEx PSyn Str DAT 130 27 APCI/ESI+: 265 131 18 NMR2: 3.01(3H, s), 3.79(3H, s), 3.87(3H, s), 5.26 (2H, d, J = 1.6 Hz), 6.47(1H, dd, J = 4.7, 3.0 Hz), 6.57 (1H, dd, J = 7.0, 3.0 Hz) 132 19 APCI/ESI+: 299 133 133 APCI/ESI+: 279 134 31 APCI/ESI+: 471

TABLE 31 PEx PSyn Str DAT 135 23 APCI/ESI+: 450 136 23 APCI/ESI+: 573 137 23 APCI/ESI+: 545 138 138 APCI/ESI+: 307 139 27 APCI/ESI+: 277 140 27 APCI/ESI+: 208

TABLE 32 PEx PSyn Str DAT 141 24 APCI/ESI+: 320 142 27 APCI/ESI+: 290 143 143 ESI+: 184 144 144 ESI+: 198 145 145 ESI+: 255 146 189 ESI+: 671 147 40 APCI/ESI+: 507

TABLE 33 PEx PSyn Str DAT 148  27 APCI/ESI+: 225 149 143 APCI/ESI+: 197 150  23 APCI/ESI+: 505 151  40 ESI+: 575 152 152 ESI+: 255 153  27 ESI+: 225 154 152 ESI+: 303

TABLE 34 PEx PSyn Str DAT 155 27 ESI+: 273 156 24 APCI/ESI+: 336 157 27 APCI/ESI+: 306 158 40 APCI/ESI+: 464 159 40 APCI/ESI+: 464 160 40 APCI/ESI+: 478

TABLE 35 PEx PSyn Str DAT 161  40 APCI/ESI+: 464 162 162 ESI+: 576 163  23 APCI/ESI+: 505 164 111 ESI+: 256 165 152 ESI+: 269 166  27 ESI+: 226

TABLE 36 PEx PSyn Str DAT 167 27 APCI/ESI+: 155(−THP) 168 23 APCI/ESI+: 519 169 23 ESI+: 506 170 23 APCI/ESI+: 553 171 23 APCI/ESI+: 516

TABLE 37 PEx PSyn Str DAT 172    32 ESI+: 500 173    31 APCI/ESI+: 472 174    40 ESI+: 561 175-1 175 APCI/ESI+: 226 175-2 175 APCI/ESI+: 226 176   176 ESI+: 232

TABLE 38 PEx PSyn Str DAT 177 23 ESI+: 512 178 23 APCI/ESI+: 506 179 23 APCI/ESI+: 506 180 23 APCI/ESI+: 506

TABLE 39 PEx PSyn Str DAT 181  23 APCI/ESI+: 450 182  27 ESI+: 226 183 183 APCI/ESI+: 242 184  27 APCI/ESI+: 212 185  23 APCI/ESI+: 492 186 186 APCI/ESI+: 276 187  27 APCI/ESI+: 246

TABLE 40 PEx PSyn Str DAT 188  23 APCI/ESI+: 526 189 189 APCI/ESI+: 267 190  27 APCI/ESI+: 237 191 191 APCI/ESI−: 490 192 27+23 APCI/ESI+: 435(−THP)

TABLE 41 PEx PSyn Str DAT 193 193 APCI/ESI+: 478 194 175 APCI/ESI+: 239 195  32 NMR2:2.96 (3H, s), 3.85(3H, s), 3.89(6H, s), 5.16 (2H, s), 5.25(2H, s), 6.68(1H, t, J = 8.0 Hz), 6.91 (1H, s), 7.63(1H, brs), 8.24(2H, s) 196 193 APCI/ESI+: 158 197 214 NMR1:4.22 (3H, s), 7.77(1H, s), 9.91(1H, s) 198 144 APCI/ESI+: 211

TABLE 42 PEx PSyn Str DAT 199  27 APCI/ESI+: 181 200  23 ESI+: 519 201 201 ESI+: 212 202 202 ESI+: 184 203  23 ESI+: 464 204 204 APCI/ESI+: 327 205 205 ESI+: 225

TABLE 43 PEx PSyn Str DAT 206 205 ESI+: 225 207  23 ESI+: 505 208  23 ESI+: 505 209 209 APCI/ESI+: 412 210 210 APCI/ESI+: 227

TABLE 44 PEx PSyn Str DAT 211 211 NMR2: 1.36-1.94(9H, m), 1.99- 2.07(1H, m), 2.09-2.20(1H, m), 2.35- 2.54(1H, m), 3.44-3.75(2H, m), 3.78- 4.04(2H, m), 4.51-4.72(2H, m), 4.78- 4.90(1H, m), 5.52-5.65(1H, m), 6.87- 6.94(1H, m) 212  27 NMR2: 1.31-1.94(10H, m),2.29- 2.44(1H, m), 3.27-3.77(4H, m), 3.81- 3.97(1H, m), 4.00-4.11(1H, m), 4.47- 4.59(1H, m), 4.61-4.73(2H, m), 5.20- 5.33(1H, m), 5.66(1H, s) 213  23 APCI/ESI+: 562 214 214 APCI/ESI+: 392

TABLE 45 PEx PSyn Str DAT 215 214 APCI/ESI+: 406 216 204 APCI/ESI+: 324 217  27 APCI/ESI+: 294 218  23 APCI/ESI+: 574

TABLE 46 PEx PSyn Str DAT 210 32 ESI+: 500 220 27 ESI+: 221 221 24 ESI+: 251 222 27 ESI+: 294 223 28 ESI+: 324 224 24 NMR2: 2.64(4H, t, J = 6.0 Hz), 3.54(4H, t, J = 6.0 Hz), 3.99(3H, s), 6.93(1H, d, J = 8.8 Hz), 7.75(1H, d, J = 2.4 Hz), 7.88(1H, dd, J = 8.8, 2.4 Hz)

TABLE 47 PEx PSyn Str DAT 225  27 ESI+: 322 226  28 ESI+: 352 227  13 EI: 332 228  18 EI: 298 229 229 EI: 220 230  30 ESI+: 613

TABLE 48 PEx PSyn Str DAT 231  27 ESI+: 333 232 232 ESI+: 361 233  27 ESI+: 294 234  28 ESI+: 324 235  30 ESI+: 616 236  27 ESI+: 294 237  28 ESI+: 324

TABLE 49 PEx PSyn Str DAT 238 32 ESI+: 580 239 27 ESI+: 336 240 28 ESI+: 366 241 27 EI: 248 242 24 ESI+: 279 243 30 ESI+: 588

TABLE 50 PEx PSyn Str DAT 244 19 NMR2: 3.94 (6H, s), 5.35(2H, s), 6.62(1H, s), 7.24 (1H, d, J = 8.8 Hz), 7.31(1H, dd, J = 8.8, 3.2 Hz), 8.16 (1H, d, J = 3.2 Hz) 245 30 ESI+: 680 246 27 ESI+: 400 247 30 ESI+: 430 248 19 NMR2: 3.94 (6H, s), 5.63(2H, s), 6.63(1H, s), 8.02 (1H, s), 8.15(1H, s)

TABLE 51 PEx PSyn Str DAT 249  30 ESI+: 714 250  27 NMR2: 1.93- 1.96(6H, m), 2.06- 2.09(2H, m), 2.08 (6H, s), 2.54- 2.60(3H, m), 2.95- 2.97(4H, m), 3.42- 3.45(2H, m), 3.81(3H, s), 4.06 (4H, s), 6.22- 6.26(2H, m), 6.74 (1H, d, J = 8.0 Hz) 251  24 ESI+: 464 252 252 ESI+: 313

TABLE 52 PEx PSyn Str DAT 253  28 ESI+: 413 254 252 ESI+: 230 255 255 NMR2: 1.46-1.52(13H, m), 2.07(6H, s), 3.40-3.43(4H, m), 4.03 (4H, s) 256  27 NMR3: 3.97(2H, t, J = 5.2 Hz), 4.42(2H, t, J = 5.2 Hz), 6.85(1H, dd, J = 2.0, 0.8 Hz), 6.92(1H, dd, J = 8.8, 2.0 Hz), 7.41(1H, dd, J = 8.8, 0.8 Hz), 7.89(1H, d, J = 0.8 Hz)

TABLE 53 PEx PSyn Str DAT 257 40 NMR2: 2.91(1H, t, J = 6.0 Hz), 4.15-4.19(2H, m), 4.61(2H, t, J = 4.8 Hz), 7.74(1H, d, J = 9.2 Hz), 8.11 (1H, dd, J = 9.2, 2.0 Hz), 8.29(1H, s), 8.73(1H, d, J = 2.4 Hz) 258 27 NMR3: 3.95(2H, t, J = 5.6 Hz), 4.37(2H, t, J = 5.6 Hz), 6.65(1H, dd, J = 8.8, 2.0 Hz), 6.72(1H, m), 7.44(1H, dd, J = 8.8, 0.8 Hz), 7.99(1H, s) 259 40 NMR2 :2.87(1H, t, J = 6.0 Hz), 4.17-4.20(2H, m), 4.62(2H, t, J = 4.8 Hz), 7.77(1H, dd, J = 9.2, 0.8 Hz), 7.91(1H, dd, J = 9.2, 2.0 Hz), 8.13 (1H, s), 8.67-8.68(1H, m)

TABLE 54 PEx PSyn Str DAT 260 30 ESI+: 631 261 27 ESI+: 351 262 24 ESI+: 381 263 30 ESI+: 482

TABLE 55 PEx PSyn Str DAT 264 30 ESI+: 522 265 27 ESI+: 264 [M + Na]+ 266 19 EI: 271 267 30 ESI+: 522 268 27 ESI+: 242 269 30 ESI+: 490

TABLE 56 PEx PSyn Str DAT 270 27 NMR2: 2.09(3H, s), 2.29(3H, s), 2.48-2.65(8H, m), 2.83(2H, t, J = 5.6 Hz), 3.54(2H, brs), 4.06(2H, t, J = 5.6 Hz), 6.20-6.22(2H, m), 6.88(1H, d, J = 8.4 Hz) 271 36 NMR2: 2.30-2.66(14H, m), 2.89(2H, t, J = 5.4 Hz), 4.19(2H, t, J = 5.6 Hz), 7.24(1H, d, J = 8.0 Hz), 7.66 (1H, d, J = 2.0 Hz), 7.75(1H, dd, J = 8.0, 2.0 Hz) 272 30 ESI+: 490

TABLE 57 PEx PSyn Str DAT 273  27 NMR3: 1.75-1.83(2H, m), 1.95- 2.01(6H, m), 2.53-2.59(2H, m), 2.86-2.97(7H, m), 3.81(3H, s), 4.45(4H, s), 6.27(1H, dd, J = 8.4, 2.4 Hz), 6.42(1H, d, J = 2.4 Hz), 6.78 (1H, d, J = 8.0 Hz) 274  24 NMR2: 1.69-1.79(2H, m), 1.85- 1.90(6H, m), 2.43-2.49(5H, m), 2.67-2.73(2H, m), 3.73-3.76(2H, m), 3.94(3H, s), 4.41(4H, s), 6.87(1H, d, J = 8.8 Hz), 7.69(1H, d, J = 2.4 Hz), 7.84(1H, dd, J = 8.8, 2.4 Hz). 275 252 ESI+: 211 276  28 ESI+: 311

TABLE 58 PEx PSyn Str DAT 277 30 EI: 497 278 27 EI: 217 279 36 ESI+: 648 280 27 ESI+: 250 281 28 ESI+: 280 282 30 ESI+: 518

TABLE 59 PEx PSyn Str DAT 283 27 ESI+: 238 284 19 EI: 267 285 27 ESI+: 197 286 30 ESI+: 506 287 27 ESI+: 264 288 28 ESI+: 294 289 30 ESI+: 631

TABLE 60 PEx PSyn Str DAT 290 27 NMR2: 1.14-1.16(2H, m), 1.45(9H, s), 1.63-1.87(5H, m), 2.70-2.71(2H, m), 3.48-3.49(2H, m), 3.81(3H, s), 3.95- 4.07(4H, m), 6.21(1H, dd, J = 8.0, 2.4 Hz), 6.30(1H, d, J = 2.4 Hz), 6.71(1H, d, J = 8.4 Hz) 291 13 NMR2: 1.18-1.22(2H, m), 1.48(9H, s), 1.59(2H, br-s), 1.69-1.75(3H, m), 1.82-1.87(2H, m), 2.71-2.73(2H, m), 3.95(3H, s), 4.16(2H, t, J = 6.4 Hz), 6.89(1H, d, J = 8.8Hz), 7.73(1H, d, J = 2.4 Hz), 7.89(1H, d, J = 8.8, 2.4 Hz) 292 19 NMR2: 2.22(3H, s), 3.89(3H, s), 3.93(3H, s), 5.33(2H, s), 6.57(1H, s), 8.37(2H, s) 293 18 EI: 294

TABLE 61 PEx PSyn Str DAT 294 229 NMR2: 1.85 (1H, t, J = 6.4 Hz), 2.26(3H, s), 3.84 (3H, s), 3.90(3H, s), 4.86(2H, d, J = 6.4 Hz), 6.49(1H, s) 295 295 ESI+: 310 296 296 ESI+: 220 297  27 ESI+: 336 298  35 ESI+: 366

TABLE 62 PEx PSyn Str DAT 299  28 ESI+: 266 300  23 APCI/ESI+: 558 301 175 APCI/ESI−: 268 302  23 ESI+: 476 303  24 ESI+: 267 304  27 ESI+: 237

TABLE 63 Ex Str 1 2 3 4 5

TABLE 64 Ex Str  6  7  8  9 10 11

TABLE 65 Ex Str 12 13 14 15 16

TABLE 66 Ex Str 17 18 19 20 21

TABLE 67 Ex Str 22 23 24 25 26

TABLE 68 Ex Str 27 28 29 30 31

TABLE 69 Ex Str 32 33 34 35 36

TABLE 70 Ex Str 37 38 39 40 41

TABLE 71 Ex Str 42 43 44 45 46

TABLE 72 Ex Str 47 48 49 50 51

TABLE 73 Ex Str 52 53 54 55 56

TABLE 74 Ex Str 57 58 59 60 61

TABLE 75 Ex Str 62 63 64 65 66

TABLE 76 Ex Str 67 68 69 70 71

TABLE 77 Ex Str 72 73 74 75 76

TABLE 78 Ex Str 77 78 79 80 81

TABLE 79 Ex Str 82 83 84 85

TABLE 80 Ex Str 86 87 88 89 90

TABLE 81 Ex Str 91 92 93 94 95

TABLE 82 Ex Str  96  97  98  99 100

TABLE 83 Ex Str 101 102 103 104 105

TABLE 84 Ex Str 106 107 108 109 110

TABLE 85 Ex Str 111 112 113 114 115

TABLE 86 Ex Str 116 117 118 119 120

TABLE 87 Ex Str 121 122 123 124 125

TABLE 88 Ex Str 126 127 128 129 130

TABLE 89 Ex Str 131 132 133 134 135

TABLE 90 Ex Str 136 137 138 139 140

TABLE 91 Ex Str 141 142 143 144 145

TABLE 92 Ex Str 146 147 148 149 150 151

TABLE 93 Ex Str 152 153 154 155 157

TABLE 94 Ex Str 158 159 160 161 162

TABLE 95 Ex Str 165 166 167 168 169

TABLE 96 Ex Str 170 171 172 173 174

TABLE 97 Ex Str 175 176 177 178 179

TABLE 98 Ex Str 180 181 182 183 184

TABLE 99 Ex Str 185 186 187 188 189

TABLE 100 Ex Str 190 191 192 193 194

TABLE 101 Ex Str 195 196 197 198 199

TABLE 102 Ex Str 200 201 202 203 204

TABLE 103 Ex Str 205 206 207 208 209

TABLE 104 Ex Str 210 211 212 213 214

TABLE 105 Ex Str 215 216 217 218 219

TABLE 106 Ex Str 220 221 222 223 224

TABLE 107 Ex Str 225 226 227 228 229

TABLE 108 Ex Str 230 231 232 233 234

TABLE 109 Ex Str 235 236 237 238 239

TABLE 110 Ex Str 240 241 242 243 244

TABLE 111 Ex Str 245 246 247 248 249

TABLE 112 Ex Str 250 251 252 253 254

TABLE 113 Ex Str 255 256 257 258 259

TABLE 114 Ex Str 260 261 262 263 264

TABLE 115 Ex Str 265 266 267 268 269

TABLE 116 Ex Str 270 271 272 273 274

TABLE 117 Ex Str 275 276 277 278 279

TABLE 118 Ex Str 280 281 282 283 284

TABLE 119 Ex Str 285 286 287 288 289

TABLE 120 Ex Str 290 291 292 293 294

TABLE 121 Ex Str 295 296 297 298 299 300

TABLE 122 Ex Str 301 302 303 304 305

TABLE 123 Ex Str 306 307 308 309 310

TABLE 124 Ex Str 311 312 313 314 315

TABLE 125 Ex Str 316 317 318 319 320

TABLE 126 Ex Str 321 322 323 324 325

TABLE 127 Ex Str 326 327 328 329 330

TABLE 128 Ex Str 331 332 333 334 335 336

TABLE 129 Ex Str 337 338 339 340 341 342

TABLE 130 Ex Str 343 344 345 346 347 348

TABLE 131 Ex Str 349 350 351 352 353

TABLE 132 Ex Str 354 355 356 357 358 359

TABLE 133 Ex Str 360 361 362 363 364 365

TABLE 134 Ex Str 366 367 368 369 370 371

TABLE 135 Ex Str 372 373 374 375 376 377

TABLE 136 Ex Str 378 379 380 381 382 383

TABLE 137 Ex Str 384 385 386 387 388

TABLE 138 Ex Syn DAT 1 1 ESI+: 543 NMR1: 1.49-1.61(2H, m), 1.77-1.85(2H, m), 2.19(3H, s), 2.22-2.71(11H, m), 3.28-3.43(2H, m), 3.76(3H, s), 3.77(6H, s), 6.55(1H, t, J = 2.3 Hz), 6.69(2H, d, J = 2.3 Hz), 6.83(1H, d, J = 8.6 Hz), 7.27(1H, dd, J = 8.6, 2.3 Hz), 7.31(1H, d, J = 2.3 Hz), 8.61(2H, s), 9.79(1H, s) 2 2 ESI+: 547 NMR1: 1.46-1.59(2H, m), 1.74-1.83(2H, m), 2.14(3H, s), 2.19-2.54(11H, m), 2.71-2.82(4H, m), 3.26-3.36(2H, m), 3.70(6H, s), 3.74(3H, s), 6.30(1H, t, J = 2.3 Hz), 6.38(2H, d, J = 2.3 Hz), 6.79(1H, d, J = 8.6 Hz), 7.24(1H, dd, J = 8.6, 2.3 Hz), 7.35(1H, d, J = 2.3 Hz), 8.26(2H, s), 9.22(1H, s) 3 3 ESI+: 611 4 4 ESI+: 611 5 5 ESI+: 649, 651 6 6 ESI+: 583 7 7 ESI+: 500 NMR1: 2.21(3H, s), 2.35-2.59(4H, m), 2.66-2.76(2H, m), 2.81-3.00(6H, m), 3.74(3H, s), 3.81(6H, s), 6.78(1H, d, J = 8.4 Hz), 6.85(1H, t, J = 8.4 Hz), 7.24(1H, dd, J = 8.4, 2.4 Hz), 7.36(1H, d, J = 2.4 Hz), 8.18(2H, s), 9.26(1H, s) 8 8 APCI/ESI+: 422 9 9 APCI/ESI+: 459 10 10 ESI+: 525 11 11 ESI+: 438 NMR1: 3.28-3.37(2H, m), 3.72-3.80(1H, m), 3.83-3.91(7H, m), 4.15(1H, dd, J = 13.8, 4.1 Hz), 4.67(1H, t, J = 5.6 Hz), 4.91(1H, d, J = 5.3 Hz), 5.14(2H, s), 7.06(1H, t, J = 8.4 Hz), 7.45(1H, d, J = 0.6 Hz), 7.87(1Hd, J = 0.6 Hz), 8.26(2H, s), 9.21(1H, s) 12 12 ESI+: 519

TABLE 139 Ex Syn DAT 13 13 ESI+: 533 14 14 ESI+: 551 15 15 ESI+: 547 16 16 ESI+: 537 17 17 APCI/ESI+: 398 18 18 ESI+: 480 19 19 ESI+: 532 NMR1: 1.59-1.61(8H, m), 2.84-2.86(4H, m), 3.04-3.05(4H, m), 3.74(3H, s), 5.26(2H, s), 6.82(1H, d, J = 8.4 Hz), 7.24(1H, d, J = 8.4 Hz), 7.32(1H, s), 7.97-8.03(1H, m), 8.34(2H, s), 8.53(1H, brs), 9.27(1H, s) 20 20 ESI+: 466 21 21 ESI+: 494 22 22 ESI+: 616 23 23 ESI+: 422 NMR1: 0.97-1.05(3H, m), 3.87(6H, s), 3.92-3.96(3H, m), 4.82-4.86(1H, m), 5.14(2H, s), 7.06(1H, t, J = 8.4 Hz), 7.44(1H, d, J = 0.6 Hz), 7.86(1H, d, J = 0.6 Hz), 8.26(2H, s), 9.21(1H, s) 24 24 ESI+: 421 NMR1: 3.87(6H, s), 4.69(2H, s), 5.15(2H, s), 7.06(1H, t, J = 8.4 Hz), 7.19(1H, brs), 7.34(1H, brs), 7.46(1H, s), 7.89(1H, s), 8.26(2H, s), 9.26(1H, s) 25 3 ESI+: 579 26 4 ESI+: 579

TABLE 140 Ex Syn DAT 27 2 ESI+: 583 NMR1: 1.46-1.59(2H, m), 1.74-1.82(2H, m), 2.14(3H, s), 2.18-2.58(11H, m), 2.65-2.75(2H, m), 2.84-2.93(2H, m), 3.24-3.38(2H, m), 3.74(3H, s), 3.81(6H, s), 6.79(1H, d, J = 8.8 Hz), 6.85(1H, t, J = 8.4 Hz), 7.23(1H, dd, J = 8.8, 2.4 Hz), 7.34(1H, d, J = 2.4 Hz), 8.17(2H, s), 9.25(1H, s) 28 7 + 4 ESI+: 530 29 3 ESI+: 579 30 3 ESI+: 496 31 3 ESI+: 526 32 3 ESI+: 567 33 6 APCI/ESI+: 571 NMR1: 1.48-1.61(2H, m), 1.77-1.87(2H, m), 2.13(3H, s), 2.20-2.64(11H, m), 2.69-2.76(2H, m), 2.85-2.93(2H, m), 3.22-3.34(2H, m), 3.81(6H, s), 6.85(1H, t, J = 8.4 Hz), 6.95(1H, dd, J = 9.9, 9.0 Hz), 7.32(1H, dd, J = 8.8, 1.8 Hz), 7.65(1H, dd, J = 15.2, 2.4 Hz), 8.21(2H, s), 9.50(1H, s) 34 3 ESI+: 579 35 4 ESI+: 579 36 7 ESI+: 583 37 3 ESI+: 561 38 6 ESI+: 565

TABLE 141 Ex Syn DAT 39 3 ESI+: 595 40 4 ESI+: 595 41 6 ESI+: 599 42 6 ESI+: 615 43 3 ESI+: 549 44 6 ESI+: 553 45 3 ESI+: 519 46 6 ESI+: 523 47 3 ESI+: 555 48 6 ESI+: 559 49 9 ESI+: 549 50 9 ESI+: 549 NMR1: 1.43-1.62(2H, m), 1.68-1.87(2H, m), 2.14(3H, s), 2.17-2.70(11H, m), 3.23-3.35(2H, m), 3.74(6H, s), 3.74(3H, s), 5.07(2H, s), 6.46(1H, t, J = 2.4 Hz), 6.60(2H, d, J = 2.4 Hz), 6.78(1H, d, J = 8.6 Hz), 7.23(1H, dd, J = 8.6, 2.2 Hz), 7.32(1H, d, J = 2.2 Hz), 8.29(2H, s), 9.15(1H, s) 51 9 ESI+: 466 52 9 ESI+: 617 NMR1: 1.40-1.60(2H, m), 1.73-1.84(2H, m), 2.14(3H, s), 2.17-2.70(11H, m), 3.24-3.36(2H, m), 3.75(3H, s), 3.94(6H, s), 5.29(2H, s), 6.79(1H, d, J = 8.6 Hz), 7.00(1H, s), 7.24(1H, dd, J = 8.6, 2.3 Hz), 7.33(1H, d, J = 2.3 Hz), 8.32(2H, s), 9.21(1H, s) 53 9 ESI+: 534

TABLE 142 Ex Syn DAT 54 9 ESI+: 631 55 9 ESI+: 548 56 9 ESI+: 585 NMR1: 1.45-1.60(2H, m), 1.73-1.84(2H, m), 2.14(3H, s), 2.17-2.58(11H, m), 3.24-3.36(2H, m), 3.75(3H, s), 3.87(6H, s), 5.16(2H, s), 6.79(1H, d, J = 8.8 Hz), 7.07(1H, t, J = 8.4 Hz), 7.24(1H, dd, J = 8.8, 2.4 Hz), 7.32(1H, d, J = 2.4 Hz), 8.29(2H, s), 9.21(1H, s) 57 9 ESI+: 502 NMR1: 2.21(3H, s), 2.37-2.53(4H, m), 2.83-2.94(4H, m), 3.75(3H, s), 3.87(6H, s), 5.16(2H, s), 6.79(1H, d, J = 8.4 Hz), 7.07(1H, t, J = 8.4 Hz), 7.25(1H, dd, J = 8.4, 2.4 Hz), 7.34(1H, d, J = 2.4 Hz), 8.30(2H, s), 9.23(1H, s) 58 9 ESI+: 561 59 9 ESI+: 478 60 9 ESI+: 586 61 9 ESI+: 522 62 9 ESI+: 601 63 9 ESI+: 601 64 64 ESI+: 561 65 64 ESI+: 616 66 9 ESI+: 572

TABLE 143 Ex Syn DAT 67 9 ESI+: 636 68 9 ESI+: 653 69 9 ESI+: 605 70 9 APCI/ESI+: 659 71 9 ESI+: 493 NMR1: 1.80-2.12(6H, m), 2.19(3H, s), 2.77-2.89(2H, m), 3.94(6H, s), 3.98-4.10(1H, m), 5.27(2H, s), 7.00(1H, s), 7.46(1H, s), 7.88(1H, s), 8.28(2H, s), 9.19(1H, s) 72 12 ESI+: 479 NMR1: 1.66-1.78(2H, m), 1.85-1.95(2H, m), 2.50-2.61(2H, m), 2.98-3.06(2H, m), 3.94(6H, s), 4.06-4.16(1H, m), 5.27(2H, s), 7.00(1H, s), 7.45(1H, s), 7.87(1H, s), 8.29(2H, s), 9.19(1H, s) 73 64 ESI+: 535 74 9 ESI+: 437 75 9 ESI+: 549 76 64 ESI+: 546 77 64 ESI+: 574 NMR2: 1.07(6H, d, J = 6.8 Hz), 1.51-1.72(8H, m), 2.51(4H, t, J = 5.2 Hz), 2.69-2.73(1H, m), 2.95(4H, t, J = 5.2 Hz), 3.87(3H, s), 5.14(2H, d, J = 1.2 Hz), 6.91-7.03(3H, m), 7.10-7.15(1H, m), 7.20(1H, d, J = 2.4 Hz), 8.20(2H, s) 78 286 ESI+: 546 NMR2: 1.67-1.71(4H, m), 1.76-1.78(4H, m), 2.31(3H, s), 2.54(2H, s), 2.71-2.72(4H, m), 2.85-3.00(4H, m), 5.14(2H, s), 6.84(1H, s), 7.05-7.15(2H, m), 7.30(1H, brs), 7.34-7.60 (1H, m), 8.19(2H, s) 79 64 ESI+: 490 NMR2: 2.43(3H, s), 3.56(4H, brs), 3.81(3H, s), 3.96(4H, s), 5.14(2H, s), 6.41(1H, d, J = 8.3 Hz), 6.81(1H, brs), 6.93(1H, dd, J = 8.3, 2.4 Hz), 7.10-7.12(2H, m), 8.17(2H, s) 80 64 ESI+: 518 NMR2: 0.95(6H, d, J = 5.4 Hz), 2.30(1H, brs), 3.37(4H, brs), 3.80-3.94(7H, m), 5.14(2H, s), 6.41(1H, d, J = 8.1 Hz), 6.81(1H, brs), 6.93(1 H, d, J = 8.5 Hz), 7.11-7.14(2H, m), 8.17(2H, s). 81 9 ESI+: 461 NMR1: 1.83-2.07(6H, m), 2.19(3H, s), 2.78-2.88(2H, m), 3.87(6H, s), 3.98-4.09(1H, m), 5.14(2H, s), 7.07(1H, t, J = 8.4 Hz), 7.45(1H, s), 7.88(1H, s), 8.25(2H, s), 9.19(1H, s)

TABLE 144 Ex Syn DAT 82 9 ESI+: 544 83 9 ESI+: 531 84 9 ESI+: 585 NMR1: 1.34-1.50(2H, m), 1.68-1.89(4H, m), 2.07-2.19(4H, m), 2.47-2.64(4H, m), 2.73-2.95(6H, m), 3.74(3H, s), 3.87(6H, s), 5.16(2H, s), 6.78(1H, d, J = 8.6 Hz), 7.07(1H, t, J = 8.4 Hz), 7.25(1H, dd, J = 8.6, 2.2 Hz), 7.33(1H, d, J = 2.2 Hz), 8.29(2H, s), 9.22(1H, s) 85 19 ESI+: 476 86 9 ESI+: 384 87 9 ESI+: 569 NMR1: 1.45-1.63(2H, m), 1.75-1.88(2H, m), 2.14(3H, s), 2.16-2.70(14H, m), 2.92-3.08(2H, m), 3.87(6H, s), 5.16(2H, s), 6.92(1H, d, J = 8.4 Hz), 7.07(1H, t, J = 8.4 Hz), 7.40-7.50(2H, m), 8.28(2H, s), 9.19(1H, s) 88 9 ESI+: 501 89 9 ESI+: 580 90 9 ESI+: 591 91 9 ESI+: 589 92 12 ESI+: 516 NMR1: 0.96(6H, d, J = 6.4 Hz), 1.98-2.10(2H, m), 2.82-2.95(2H, m), 3.06-3.15(2H, m), 3.74(3H, s), 3.87(6H, s), 5.16(2H, s), 6.76(1H, d, J = 8.8 Hz), 7.07(1H, t, J = 8.4 Hz), 7.24(1H, dd, J = 8.8, 2.4 Hz), 7.32(1H, d, J = 2.4 Hz), 8.29(2H, s), 9.21(1H, s)

TABLE 145 Ex Syn DAT 93 12 ESI+: 599 94 286 ESI+: 517 95 12 ESI+: 556 NMR2: 1.76-1.81(8H, m), 2.97(4H, brs), 3.15(4H, brs), 3.88(9H, s), 5.14(2H, s), 6.67(1H, t, J = 8.2 Hz), 6.88-6.90(2H, m), 7.01(1H, dd, J = 8.4, 2.4 Hz), 8.20(2H, s), 9.26(1H, brs) 96 64 ESI+: 570 NMR2: 1.62-1.68(8H, m), 2.32(3H, s), 2.43-2.45(4H, m), 2.94- 2.97(4H, m), 3.88(9H, s), 5.14(2H, s), 6.67(1H, t, J = 8.2 Hz), 6.88-7.00(3H, m), 7.21-7.22(1H, m), 8.19(2H, t, J = 2.2 Hz) 97 12 ESI+: 500 NMR2: 3.81-4.01(17H, m), 5.13(2H, s), 6.40(1H, d, J = 8.3 Hz), 6.64-6.68(1H, m), 6.77(1H, d, J = 7.1 Hz), 6.92(1H, d, J-8.5 Hz), 7.12(1H, brs), 8.17(2H, s) 98 64 ESI+: 514 NMR2: 2.38(3H, s), 3.47-3.58(4H, m), 3.81-3.94(13H, m), 5.13(2H, s), 6.40(1H, d, J = 8.5 Hz), 6.64-6.68(1H, m), 6.78(1H, brs), 6.92(1H, dd, J = 8.5, 2.0 Hz), 7.12(1H, d, J = 2.7 Hz), 8.17(2H, s) 99 64 ESI+: 542 NMR2: 0.90-0.94(6H, m), 2.10(1H, brs), 3.40-3.96(17H, m), 5.13(2H, s), 6.41(1H, d, J = 8.3 Hz), 6.66(1H, d, J = 8.0 Hz), 6.76(1H, brs), 6.91-6.93(1H, m), 7.13(1H, s), 8.17(2H, s) 100 286 ESI+: 599 NMR2: 1.66-1.69(4H, m), 2.29(3H, s), 2.30(2H, s), 2.40(3H, s), 2.45(4H, brs), 2.70(4H, brs), 2.84-2.87(2H, m), 2.96-3.01(2H, m), 3.88(6H, s), 5.13(2H, s), 6.67(1H, t, J = 8.2 Hz), 6.87(1H, s), 7.04(1H, d, J = 8.8 Hz), 7.29(1H, d, J = 2.8 Hz), 7.34(1H, d, J = 8.4 Hz), 8.18(2H, s) 101 286 ESI+: 588 NMR3: 1.08(3H, d, J = 6.4 Hz), 1.12(3H, s), 1.20(3H, s), 1.49-1.52(1H, m), 1.64-1.66(1H, m), 1.94-1.96(1H, m), 2.03-2.04(1H, m), 2.59-2.68(4H, m), 3.29-3.34(2H, m), 3.87(9H, s), 5.15(2H, d, J = 2.0 Hz), 6.89-6.93(2H, m), 7.10-7.13(1H, m), 7.37(1H, d, J = 2.0 Hz), 8.19(2H, d, J = 2.0 Hz)

TABLE 146 Ex Syn DAT 102 286 ESI+: 470 103 9 ESI+: 502 104 19 ESI+: 596 105 12 ESI+: 568 106 106 ESI+: 582 107 12 ESI+: 623 108 19 ESI+: 588 NMR2: 1.76-1.81(8H, m), 2.96-2.97(4H, m), 3.15-3.16(4H, m), 3.88(3H, s), 3.94(6H, s), 5.33(2H, s), 6.61(1H, s), 6.89-6.91(2H, m), 7.01(1H, dd, J = 8.0, 2.4 Hz), 7.22-7.26(1H, m), 8.23(2H, s) 109 19 ESI+: 532

TABLE 147 Ex Syn DAT 110 64 ESI+: 546 NMR2: 2.36(3H, brs), 3.45-3.50(4H, m), 3.81(3H, s), 3.93(10H, s), 5.32(2H, s), 6.40(1H, d, J = 8.5 Hz), 6.60(1H, s), 6.76(1H, brs), 6.92-6.95(1H, m), 7.14(1H, s), 8.20(2H, s) 111 4 ESI+: 649 112 10 ESI+: 543 113 9 ESI+: 408 NMR1: 3.69(2H, dd, J = 11.0, 5.6 Hz), 3.87(6H, s), 4.07(2H, t, J = 5.6 Hz), 4.83(1H, t, J = 5.4 Hz), 5.14(2H, s), 7.07(1H, t, J = 8.4 Hz), 7.45(1H, d, J = 0.6 Hz), 7.88(1H, d, J = 0.6 Hz), 8.26(2H s), 9.20(1H, s) 114 20 ESI+: 490 NMR1: 2.13(3H, s), 2.17-2.54(8H, m), 2.66(2H, t, J = 6.8 Hz), 3.87(6H, s), 4.14(2H, t, J = 6.8 Hz), 5.14(2H, s), 7.06(1H, t, J = 8.4 Hz), 7.43(1H, d, J = 0.6 Hz), 7.89(1H, d, J = 0.6 Hz), 8.25(2H, s), 9.20(1H, s) 115 18 ESI+: 477 NMR1: 3.58-3.65(1H, m), 3.75-3.84(1H, m), 3.87(6H, s), 4.04-4.10 (1H, m), 4.17-4.24(1H, m), 4.41-4.49(1H, m), 4.78(2H, s), 5.15(2H, s), 5.72(1H, s), 7.07(1H, t, J = 8.4 Hz), 7.46(1H, d, J = 0.4 Hz), 7.88(1H, d, J = 0.4 Hz), 8.26(2H, s), 9.27(1H, s) 116 11 ESI+: 438 NMR1: 3.23-3.38(2H, m), 3.72-3.80(1H, m), 3.84-3.96(7H, m), 4.15(1H, dd, J = 13.8, 4.1 Hz), 4.67(1H, t, J = 5.6 Hz), 4.91(1H, d, J = 5.3 Hz), 5.14(2H, s), 7.06(1H, t, J = 8.4 Hz), 7.45(1H, d, J = 0.6 Hz), 7.87(1H, d, J = 0.6 Hz), 8.26(2H, s), 9.21(1H, s) 117 286 ESI+: 502 118 9 ESI+: 463 119 12 APCI/ESI+: 449 120 120 ESI+: 531 121 13 ESI+: 463 122 9 ESI+: 472 NMR1: 2.21(3H, s), 2.41-2.48(4H, m), 2.98-3.08(4H, m), 3.87(6H, s), 5.15(2H, s), 6.81-6.90(2H, m), 7.07(1H, t, J = 8.4 Hz), 7.47-7.55(2H, m), 8.26(2H, s), 9.15(1H, s)

TABLE 148 Ex Syn DAT 123 9 ESI+: 555 124 9 ESI+: 531 125 20 ESI+: 466 126 20 ESI+: 453 127 9 APCI/ESI+: 384 128 20 ESI+: 453 129 9 ESI+: 599 130 9 ESI+: 603 131 120 ESI+: 599 132 120 ESI+: 613 133 120 ESI+: 572 134 12 ESI+: 505 135 9 ESI+: 499 136 12 ESI+: 423 137 12 ESI+: 447 138 9 ESI+: 573 139 9 ESI+: 470 140 9 ESI+: 505 141 120 ESI+: 597 142 12 ESI+: 458 143 12 ESI+: 476 144 166 + 4  ESI+: 514 145 12 ESI+: 486 146 12 ESI+: 457 147 13 + 4  ESI+: 611 148 9 + 4 ESI+: 583 149 9 + 4 ESI+: 543 150 9 + 4 ESI+: 557 151 13 ESI+: 471 152 64 ESI+: 554 153 9 ESI+: 567

TABLE 149 Ex Syn DAT 154 9 ESI+: 474 155 9 ESI+: 487 157 15 ESI+: 499 158 16 ESI+: 501 159 64 ESI+: 541 160 17 ESI+: 422 161 161 ESI+: 515 162 120 + 162  ESI+: 555 165 9 ESI+: 487 166 166 ESI+: 445 167 18 ESI+: 504 168 12 + 162 ESI+: 473 169 18 ESI+: 465 170 13 + 162 ESI+: 487 171 9 ESI+: 557 172 18 ESI+: 491 173 20 ESI+: 477 174 20 ESI+: 504 175 23 ESI+: 436 176 9 ESI+: 461 177 9 ESI+: 556 178 9 ESI+: 488

TABLE 150 Ex Syn DAT 179 9 APCI/ESI+: 487 180 18 ESI+: 465 181 18 ESI+: 509 182 9 ESI+: 483 183 9 ESI+: 570 184 23 ESI+: 422 185 9 ESI+: 556 186 17 ESI+: 448 187 166 ESI+: 458 188 16 ESI+: 422 189 16 ESI+: 378 190 190 APCI/ESI+: 407 191 64 ESI+: 555 192 106 ESI+: 502 193 19 ESI+: 571 194 12 ESI+: 475 195 9 ESI+: 586 196 18 ESI+: 491 197 18 ESI+: 491 198 18 ESI+: 505

TABLE 151 Ex Syn DAT 199 18 ESI+: 504 200 18 ESI+: 518 201 11 ESI+: 465 202 13 ESI+: 489 203 20 ESI+: 504 204 20 ESI+: 435 205 11 ESI+: 465 206 20 + 4 ESI+: 463 207 12 + 4 ESI+: 476 208 9 ESI+: 461 209 20 ESI+: 504 210  20 + 162 ESI+: 518 211 9 ESI+: 448 212 212 ESI+: 422 213 213 ESI+: 465 214 214 ESI+: 435 215 12 ESI+: 461 216 120 ESI+: 531 217 217 ESI+: 517 218 17 ESI+: 436 219 17 ESI+: 450 220 17 ESI+: 436 221 17 ESI+: 450 222 18 ESI+: 491 223 18 ESI+: 505 224 18 ESI+: 491 225 18 ESI+: 505 226 13 ESI+: 475

TABLE 152 Ex Syn DAT 227 18 ESI+: 495 228 18 ESI+: 479 229 9 ESI+: 503 230 214 ESI+: 422 231 20 + 162 ESI+: 504 232 217 ESI+: 531 233 214 ESI+: 422 234 217 ESI+: 547 235 213 ESI+: 438 236 9 + 4  ESI+: 517 237 9 ESI+: 517 238 17 APCI/ESI+: 422 239 239 ESI+: 422 240 214 ESI+: 408 241 20 + 162 ESI+: 504 242 18 ESI+: 504 243 18 ESI+: 505 244 24 ESI+: 421 245 214 ESI+: 408 246 246 APCI/ESI+: 408 247 214 ESI+: 435 248 20 ESI+: 490 NMR1: 2.14(3H, s), 2.18-2.53(8H, m), 3.45(2H, s), 3.67(3H, s), 3.87(6H, s), 5.15(2H, s), 6.46(1H, s), 7.06(1H, t, J = 8.4 Hz), 8.26(2H, s), 9.42(1H, s) 249 20 + 4  ESI+: 491 250 64 ESI+: 514 251 214 ESI+: 435 252 9 ESI+: 478 253 253 ESI+: 517 254 254 ESI+: 461 255 253 ESI+: 517 256 17 ESI+: 450

TABLE 153 Ex Syn DAT 257 246 ESI+: 436 258 24 ESI+: 449 259 11 ESI+: 465 260 11 ESI+: 465 261 18 ESI+: 505 262 18 ESI+: 532 263 20 ESI+: 476 264 254 ESI+: 507 265 254 ESI+: 507 266 18 ESI+: 491 267 18 ESI+: 463 268 18 ESI+: 504 269 24 ESI+: 407 270 214 APCI/ESI+: 394 271 20 ESI+: 504 272 20 ESI+: 477 273 20 ESI+: 461 274 20 ESI+: 504 275 20 ESI+: 504 276 20 ESI+: 520 277 20 ESI+: 477 278 278 ESI+: 390 279 20 ESI+: 490 280 282 ESI+: 447 281 282 + 4 ESI+: 490 282 282 ESI+: 463 283 282 ESI+: 490 284 282 ESI+: 506 285 214 ESI+: 490 286 286 ESI+: 533

TABLE 154 Ex Syn DAT 287 286 ESI+: 501 288 286 ESI+: 530 289 286 ESI+: 574 290 286 ESI+: 602 291 286 ESI+: 601 292 12 ESI+: 513 293 64 ESI+: 527 294 64 ESI+: 555 295 286 ESI+: 574 NMR3: 1.23(6H, s), 1.61-1.64(2H, m), 1.97-1.99(2H, m), 2.57-2.62(5H, m), 3.31-3.34(2H, m), 3.87(9H, s), 5.16(2H, t, J = 1.6 Hz), 6.89-6.94(2H, m), 7.11(1H, dd, J = 8.8, 2.4 Hz), 7.36(1H, d, J = 2.4 Hz), 8.20(2H, s) 296 16 ESI+: 544 297 286 ESI+: 532 298 286 ESI+: 502 NMR2: 2.61(2H, t, J = 5.6 Hz), 2.69(4H, t, J = 4.8 Hz), 3.16(4H, t, J = 4.8 Hz), 3.66(2H, t, J = 5.6 Hz), 3.88(6H, s), 5.13(2H, s), 6.66(1H, t, J = 8.0 Hz), 6.80(1H, br-s), 6.92(2H, d, J = 9.2 Hz), 7.43(2H, d, J = 9.2 Hz), 8.18(2H, s) 299 12 ESI+: 516 NMR2: 1.67-1.69(2H, m), 2.06-2.09(2H, m), 2.70-2.81(3H, m), 2.98(2H, t, J = 5.2 Hz), 3.60(2H, d, J = 12.4 Hz), 3.76-3.77(2H, m), 3.88(6H, s), 5.13(2H, s), 6.66(1H, t, J = 8.0 Hz), 6.82(1H, s), 6.92(2H, d, J = 8.8 Hz), 7.41(2H, d, J = 8.8 Hz), 8.18(2H, s) 300 286 ESI+: 574 301 16 ESI+: 571 302 302 ESI+: 516 303 64 ESI+: 540 304 286 ESI+: 616 305 4 ESI+: 529 306 286 ESI+: 529 307 16 ESI+: 546 308 12 ESI+: 488 309 286 ESI+: 533 310 336 ESI+: 560 311 12 ESI+: 580 312 64 ESI+: 594 313 64 ESI+: 622 314 286 ESI+: 534

TABLE 155 Ex Syn DAT 315 315 ESI+: 630 316 286 ESI+: 585 317 286 ESI+: 458 318 286 ESI+: 458 319 286 ESI+: 458 320 286 ESI+: 458 321 315 ESI+: 547 322 16 ESI+: 533 323 16 ESI+: 546 324 11 ESI+: 482 325 17 ESI+: 494 326 17 ESI−: 460 327 286 ESI+: 448 328 18 ESI+: 531 329 286 ESI+: 530 330 286 ESI+: 517 331 17 ESI−: 460 332 18 ESI+: 544 333 286 ESI+: 516 334 286 ESI+: 612 335 16 ESI+: 549 336 336 ESI+: 480 337 12 ESI+: 548 338 286 ESI+: 530 339 11 ESI+: 478 340 286 ESI+: 408 341 286 ESI+: 490 342 286 ESI+: 477 343 17 ESI−: 476 344 286 ESI+: 438 NMR1: 3.26-3.40(2H, m), 3.76-3.78(1H, m), 3.87(6H, s), 3.89-3.94(1H, m), 4.15(1H, dd, J = 14.0, 4.0 Hz), 4.71(1H, t, J = 5.6 Hz), 4.95(1H, d, J = 5.2 Hz), 5.27(2H, s), 7.05(1H, t, J = 8.4 Hz), 7.41(1H, s), 7.74(1H, d, J = 1.6 Hz), 7.84(1H, d, J = 1.6 Hz), 7.88(1H, s), 8.95(1H, brs) 345 64 ESI+: 562 346 286 ESI+: 544 347 18 ESI+: 563 348 18 ESI+: 521 349 349 ESI+: 508

TABLE 156 Ex Syn DAT 350 18 ESI+: 576 351 286 ESI+: 473 352 286 ESI+: 422 353 12 ESI+: 531 354 64 ESI+: 545 355 286 ESI+: 597 356 356 ESI+: 476 357 356 ESI+: 446 358 356 ESI+: 446 359 356 ESI+: 460 360 356 ESI+: 473 361 356 ESI+: 486 362 356 ESI+: 432 363 356 ESI+: 418 364 356 ESI+: 432 365 356 ESI+: 418 366 356 ESI+: 466 367 356 ESI+: 478 368 356 ESI+: 460 369 356 ESI+: 466 370 356 ESI+: 378 371 356 ESI+: 444 372 356 ESI+: 473 373 375 ESI+: 459 374 375 ESI+: 433 375 375 ESI+: 419 376 356 ESI+: 458 377 356 ESI+: 447 378 356 ESI+: 471 379 356 ESI+: 490 380 356 ESI+: 417 381 375 ESI+: 403 382 356 ESI+: 459 383 356 ESI+: 501 384 356 ESI+: 519 385 356 ESI+: 477 386 356 ESI+: 459 387 356 ESI+: 476 388 286 ESI+: 517

INDUSTRIAL APPLICABILITY

The compound of formula (I) or a salt thereof according to the present invention has inhibitory action on FGFR1, FGFR2, and/or FGFR3, particularly, mutant FGFR3, and can be used as a therapeutic agent for various cancers related to FGFR1, FGFR2, and/or FGFR3, such as lung cancer and hormone therapy-resistant breast cancer, stomach cancer, triple negative breast cancer, endometrial cancer, and bladder cancer, particularly as a therapeutic agent for mutant FGFR3-positive bladder cancer.

SEQUENCE LISTING FREE TEXT

The numerical heading <223> in the Sequence Listing shown below contains an explanation of “Artificial Sequence”. More specifically, the base sequences represented by SEQ ID NOs: 7, 8, 17, 20, and 21 in the Sequence Listing are artificially synthesized primer sequences. The base sequence represented by SEQ ID NO: 24 in the Sequence Listing is an artificially synthesized FLAG tag sequence.

Claims

1: A compound of formula (I) or a salt thereof:

wherein X and Y are each independently CH or N, with the proviso that X and Y are not N simultaneously;
L1 is -lower alkylene-, -lower alkylene-O—, —O-lower alkylene-, or -lower alkynylene-;
Z is N or CH;
each R1 is independently lower alkyl optionally substituted with halogen, —O-(lower alkyl optionally substituted with halogen), halogen, cyano, or —N(lower alkyl)2;
p is an integer of from 2 to 4;
ring W is an optionally substituted aromatic carbocyclic ring, an optionally substituted aromatic heterocyclic ring, or an optionally substituted non-aromatic heterocyclic ring;
Q is -L2-R2 or R3;
L2 is an optionally substituted aromatic heterocyclic ring or an optionally substituted non-aromatic heterocyclic ring;
R2 is a non-aromatic heterocyclic group optionally substituted with lower alkyl, optionally substituted cycloalkyl, lower alkyl optionally substituted with at least one selected from the group consisting of —OH and —O-lower alkyl, —C(O)—R0, —C(O)-optionally substituted cycloalkyl, —NH—R0, —N(lower alkyl)-R0, an -L3-optionally substituted non-aromatic heterocyclic group, or H;
R0 is lower alkyl optionally substituted with —OH;
L3 is a bond, —NH—, —N(lower alkyl)-, or lower alkylene; and
R3 is:
a lower alkyl optionally substituted with at least one selected from the group consisting of —C(O)OH, —OH, —O—R0, amino optionally substituted with one or two R0, carbamoyl optionally substituted with one or two R0, an optionally substituted aromatic heterocyclic group, an optionally substituted non-aromatic heterocyclic group, and a —C(O)-optionally substituted non-aromatic heterocyclic group,
—O-(lower alkyl optionally substituted with at least one selected from the group consisting of —C(O)OH, —OH, —O—R0, carbamoyl optionally substituted with one or two R0, an optionally substituted non-aromatic heterocyclic group, and a —C(O)-optionally substituted non-aromatic heterocyclic group),
—NH-(lower alkyl optionally substituted with at least one selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with lower alkyl, and carbamoyl optionally substituted with one or two R0),
—N(lower alkyl)-(lower alkyl optionally substituted with at least one selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with lower alkyl, and carbamoyl optionally substituted with one or two R0),
—C(O)OH,
—C(O)-optionally substituted non-aromatic heterocyclic group,
—O-(a non-aromatic heterocyclic group optionally substituted with lower alkyl), or
carbamoyl optionally substituted with one or two R0.

2: The compound or salt thereof according to claim 1,

wherein X is N;
Y is CH; and
L1 is lower alkylene or -lower alkylene-O—.

3: The compound or salt thereof according to claim 2,

wherein Z is CH;
each R1 is independently —O-lower alkyl or halogen;
p is 2 or 4; and
ring W is an optionally substituted aromatic carbocyclic ring or an optionally substituted aromatic heterocyclic ring.

4: The compound or salt thereof according to claim 3,

wherein L1 is ethylene or -methylene-O—;
p is 4; and
ring W is an optionally substituted benzene ring or optionally substituted pyrazole.

5: The compound or salt thereof according to claim 2,

wherein Q is -L2-R2;
L2 is an optionally substituted non-aromatic heterocyclic ring; and
R2 is lower alkyl optionally substituted with at least one selected from the group consisting of —OH and —O-lower alkyl, —NH-(lower alkyl optionally substituted with —OH), an optionally substituted non-aromatic heterocyclic group, -lower alkylene-(an optionally substituted non-aromatic heterocyclic group), or H.

6: The compound or salt thereof according to claim 5,

wherein p is 4;
L2 is piperazine optionally substituted with one or more methyl, piperidine optionally substituted with one or more methyl, or 3,9-diazaspiro[5.5]undecane; and
R2 is piperazine optionally substituted with methyl, piperidine optionally substituted with methyl, 2-hydroxyethylamino, or H.

7: The compound or salt thereof according to claim 6,

wherein each R1 is independently —O-methyl or F;
L1 is -methylene-O—;
ring W is a benzene ring optionally substituted with —O-methyl;
L2 is piperidine or 4-methylpiperazine; and
R2 is 4-methylpiperazine, 2-hydroxyethylamino, or H.

8: The compound or salt thereof according to claim 2,

wherein ring W is optionally substituted pyrazole;
Q is R3; and
R3 is lower alkyl substituted with at least one selected from the group consisting of —C(O)OH, carbamoyl optionally substituted with one or two R0, —OH, an optionally substituted non-aromatic heterocyclic group, and —C(O)-(an optionally substituted non-aromatic heterocyclic group).

9: The compound or salt thereof according to claim 8,

wherein p is 4; and
R3 is lower alkyl substituted with at least one selected from the group consisting of —OH, a non-aromatic heterocyclic group optionally substituted with lower alkyl, and —C(O)-(a non-aromatic heterocyclic group optionally substituted with —OH).

10: The compound or salt thereof according to claim 9,

wherein each R1 is independently —O-methyl or F;
L1 is -methylene-O—;
ring W is pyrazole optionally substituted with methyl; and
R3 is 2-hydroxyethyl, 2,3-dihydroxypropyl, or 4-methylpiperazin-1-ylmethyl.

11: The compound or salt thereof according to claim 1, wherein the compound is selected from the group consisting of:

5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine,
(2S)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol,
5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
5-[2-(2,6-difluoro-3,5-dimethoxyphenyl)ethyl]-N-{3-fluoro-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[3-methoxy-4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine,
5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(1-methylpiperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine,
5-[(2,6-dichloro-3,5-dimethoxybenzyl)oxy]-N-[1-(piperidin-4-yl)-1H-pyrazol-4-yl]pyrimidin-2-amine,
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methoxy-4-[4-(1-methylpiperidin-4-yl)piperazin-1-yl]phenyl}pyrimidin-2-amine,
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{3-methyl-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidin-2-amine,
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{4-[(3R,5S)-3,5-dimethylpiperazin-1-yl]-3-methoxyphenyl}pyrimidin-2-amine,
N-[4-(3,9-diazaspiro[5.5]undec-3-yl)-3-methoxyphenyl]-5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-amine,
2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]ethanol,
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-[2-(4-methylpiperazin-1-yl)ethyl]-1 H-pyrazol-4-yl}pyrimidin-2-amine,
2-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]-1-(3-hydroxyazetidin-1-yl)ethanone,
(2R)-3-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)-1H-pyrazol-1-yl]propane-1,2-diol,
2-({1-[4-({5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]pyrimidin-2-yl}amino)phenyl]piperidin-4-yl}amino)ethanol,
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-{1-methyl-5-[(4-methylpiperazin-1-yl)methyl]-1H-pyrazol-3-yl}pyrimidin-2-amine, and
5-[(2,6-difluoro-3,5-dimethoxybenzyl)oxy]-N-[4-(4-methylpiperazin-1-yl)phenyl]pyrimidin-2-amine.

12: A pharmaceutical composition, comprising:

the compound or salt thereof according to claim 1 and
a pharmaceutically acceptable excipient.

13: The pharmaceutical composition according to claim 12, wherein the compound is suitable for treatment of mutant FGFR3-positive cancer.

14: A method of manufacturing a pharmaceutical composition, the method comprising:

manufacturing the pharmaceutical composition with the compound or salt thereof according to claim 1,
wherein the pharmaceutical composition is suitable for treatment of mutant FGFR3-positive cancer.

15. (canceled)

16: The compound or salt thereof according to claim 1, wherein the compound is suitable for treatment of mutant FGFR3-positive cancer.

17: A method of treating mutant FGFR3-positive cancer, the method comprising:

administering an effective amount of the compound or salt thereof according to claim 1 to a subject in need thereof.
Patent History
Publication number: 20170152232
Type: Application
Filed: Sep 27, 2016
Publication Date: Jun 1, 2017
Applicants: Astellas Pharma Inc. (Tokyo), Kotobuki Pharmaceutical Co., Ltd. (Nagano)
Inventors: Minoru KAMEDA (Tokyo), Ikumi KURIWAKI (Tokyo), Kazuhiko IIKUBO (Tokyo), Hiroyuki HISAMICHI (Tokyo), Yuichiro KAWAMOTO (Tokyo), Hiroyuki MORITOMO (Tokyo), Tomoyuki SUZUKI (Tokyo), Takashi FUTAMI (Tokyo), Atsushi SUZUKI (Tokyo), Kazuhisa TSUNOYAMA (Tokyo), Makoto ASAUMI (Tokyo), Hiroshi TOMIYAMA (Nagano), Atsushi NODA (Nagano), Yoshinori IWAI (Nagano), Kazuo TOKUZAKI (Nagano), Haruki OKADA (Nagano), Kozo MIYASAKA (Nagano)
Application Number: 15/277,484
Classifications
International Classification: C07D 239/47 (20060101); C07D 239/42 (20060101); C07D 403/12 (20060101); C07D 401/12 (20060101); C07D 491/107 (20060101); C07D 405/14 (20060101); C07D 487/10 (20060101); C07D 413/12 (20060101); C07D 471/08 (20060101); C07D 417/12 (20060101); C07D 401/14 (20060101); C07D 471/10 (20060101);