KR101937529B1 - Novel pyrimidine-2,4-diamine derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient - Google Patents

Novel pyrimidine-2,4-diamine derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient Download PDF

Info

Publication number
KR101937529B1
KR101937529B1 KR1020160094560A KR20160094560A KR101937529B1 KR 101937529 B1 KR101937529 B1 KR 101937529B1 KR 1020160094560 A KR1020160094560 A KR 1020160094560A KR 20160094560 A KR20160094560 A KR 20160094560A KR 101937529 B1 KR101937529 B1 KR 101937529B1
Authority
KR
South Korea
Prior art keywords
phenyl
chloro
isopropylsulfonyl
pyrimidine
diamine
Prior art date
Application number
KR1020160094560A
Other languages
Korean (ko)
Other versions
KR20180012352A (en
Inventor
윤창수
김형래
김필호
박지훈
안선주
이정옥
정희정
조성윤
하재두
황종연
박종배
이승훈
Original Assignee
한국화학연구원
국립암센터
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 한국화학연구원, 국립암센터 filed Critical 한국화학연구원
Priority to KR1020160094560A priority Critical patent/KR101937529B1/en
Priority to PCT/KR2017/008052 priority patent/WO2018021826A1/en
Publication of KR20180012352A publication Critical patent/KR20180012352A/en
Application granted granted Critical
Publication of KR101937529B1 publication Critical patent/KR101937529B1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines having two or more nitrogen atoms in the same ring, e.g. oxadiazines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/308Foods, ingredients or supplements having a functional effect on health having an effect on cancer prevention

Abstract

The present invention relates to a novel pyrimidine-2,4-diamine derivative or a pharmaceutically acceptable salt thereof and a pharmaceutical composition for the prevention or treatment of cancer containing the same as an active ingredient, (ALK) activity, and thus can improve the therapeutic effect on cancer cells having an inverted lymphoma kinase (ALK) fusion protein such as EML4-ALK and NPM-ALK, and the cancer recurrence It is expected to be effective in preventing cancer, and thus it can be usefully used as a pharmaceutical composition for preventing or treating cancer.

Description

[0001] The present invention relates to a novel pyrimidine-2,4-diamine derivative and a pharmaceutical composition for preventing or treating cancer containing the same as an active ingredient. as an active ingredient}

The present invention relates to a novel pharmaceutical composition for preventing or treating a pyrimidine-2,4-diamine derivative or a pharmaceutically acceptable salt thereof and a cancer containing the same as an active ingredient.

Cancer is a mass of cells composed of undifferentiated cells which, unlike normal cells, can regulate and inhibit proliferation and suppression according to the individual's needs, ignoring the necessary conditions in the tissues and is called a tumor. These unlimited, proliferating cancer cells penetrate into surrounding tissues and, in the worse case, become metastasized to other organs of the body, resulting in severe pain and ultimately death.

According to the American Cancer Society data, there are more than 12 million new cases of cancer diagnosed globally in 2007, with 7.6 million deaths and about 20,000 deaths every day. In Korea, according to 2006 National Statistical Office, death from cancer was the leading cause of death. Therefore, the development of a tumor treatment agent having excellent therapeutic effect for the reduction of mental and physical pain caused by cancer and bruising and improvement of the quality of life is urgently required.

However, in many efforts, it has not yet been accurately determined how normal cells are transformed into cancer cells. However, the internal factors such as environmental factors, chemical substances, radiation, and viruses, and external factors such as genetic factors and immunological factors Factors are complexly entangled, resulting in cancer. The genes involved in the development of cancer include oncogenes and tumor suppressor genes. Cancer occurs when the balance between them is destroyed by the internal or external factors described above.

Cancer is largely classified into blood cancer and solid cancer and occurs in almost all parts of the body such as lung cancer, stomach cancer, breast cancer, oral cancer, liver cancer, uterine cancer, esophageal cancer and skin cancer. Recently, a few targets such as Gleevec or Herceptin Therapeutics are currently being used to treat certain cancers, but until now, chemotherapy with chemotherapeutic agents that inhibit surgery, radiation therapy and cell proliferation is the main method. However, since it is not a target drug, the biggest problem of conventional chemotherapy is cytotoxic side effects and drug resistance, which are the main factors that eventually fail treatment despite successful initial response by anticancer drugs. Therefore, in order to overcome the limitations of these chemotherapeutic agents, it is necessary to develop a targeted therapeutic agent with clear mechanism of action.

Therefore, many studies on specific molecular biologic factors involved in tumorigenesis for developing a target therapeutic agent are under way, and in particular, molecular biological factors are widely used to determine the prognosis of cancer, chemotherapy, and radiation therapy .

Gleevec is one of the most representative drugs that inhibit tyrosine kinase receptors, one of the specific molecular biological factors. The Gleevec inhibits the action of the Bcr-Abl fusion gene, which is formed by chromosomal translocation in the Philadelphia chromosome observed in chronic myelogenous leukemia, and thus acts as an anticancer agent. As a tyrosine kinase inhibitor, Gleevec exhibits satisfactory results when administered to patients suffering from chronic myelogenous leukemia have. Thereafter, the drugs that exhibit anticancer effects with tyrosine kinase inhibitors include gefitinib and erlotinib, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, which are used as therapeutic agents for non-small cell lung cancer, and kidney cells Sorafenib and sunitinib have been used to treat cancer, but they have been known to have side effects such as bleeding, heart attack, heart failure, and liver failure.

Recently, anaplastic lymphoma kinase (ALK) has been found in many human tumors and has been studied as a target of target treatment.

The carcinogenic process of inverse lymphoma kinase (ALK) is known to be a fusion gene of ALK-NPM (nucleophosmin, nucleophosphin), which is mainly observed in inverse large cell lymphoma. When the inverse lymphoma kinase (ALK) is activated by gene fusion, the tyrosine kinase of the inverse lymphoma kinase (ALK) acts abnormally and causes cancer. In other words, abnormally activated degenerative lymphoma kinase (ALK) induces cell proliferation, prevents apoptosis, prevents cell death, rearranges the cellular skeleton, and modifies cell morphology. Cancer geneation of inverted lymphoma kinase (ALK) is mediated by interaction with a downstream molecule, the target of reverse forming lymphoma kinase (ALK), which mediates intracellular signaling. Inverse lymphoma kinase (ALK) is linked to other tyrosine kinases that are normal or cancer-genetically modified to interact or activate a variety of other pathways.

In particular, the inverse Lymphoma kinase (ALK) gene in lung cancer cells is fused with EML4 (Echinoderm Microtubule-Associated Protein-Like 4) gene to produce active tyrosine kinase, EML4-ALK, In addition, Mosse et al. Reported that 26% of the 491 neuroblastoma specimens were amplified with the reverse priming lymphoma kinase (ALK) gene amplification, and that EML4-reverse forming lymphoma kinase (ALK) There is one. In addition, the inverse lymphoma kinase (ALK) gene may be useful for the treatment of numerous B-cell lymphomas, systemic breakfast necrosis, inflammatory myoblastic fibrosarcoma, esophageal squamous cell carcinoma, non-small cell lung carcinoma, rhabdomyosarcoma, myoblastoma, melanoma and melanoma cell lines In the rare disease of inflammatory bone marrow fibroblastoma tumor, several types of reverse priming lymphoma kinase (ALK) fusion proteins have been found to be expressed in non-hematopoietic cell tumors, and these fusion proteins are thought to be deeply involved in tumor development have.

Thus, therapeutic agents targeting ALK-NPM for the treatment of cancer are being developed by blocking the activation pathway of inverse lymphoma kinase (ALK). Recently, Crytotinib (PF-02341066), a small molecule tyrosine phosphorylase inhibitor, has been developed as a selective inhibitor of tumorigenic mutation in Pfizer. It has been shown that ATP competitiveness c-Met / HGFR and inverse lymphoma kinase ) Inhibitor, which is known to be effective for the treatment of non-small cell lung cancer, and was approved as a new drug in FDA 2011 (Patent Document 1).

In addition, NVP-TAE684, LDK-378 (Patent Document 2) of Novartis, and CH5424802 of Chugai Co. are known to have an effect of reducing tumor size in neuroblastoma cell lines in addition to inverse large cell lymphoma cell lines have.

On the other hand, receptor tyrosine kinases (RTKs), which are one of protein kinases, are located on the surface of the cell membrane and serve to transmit signals received from growth factors and hormones, which are signal substances transmitted from the central nervous system, into cells. These tyrosine kinases ) Is overexpressed, various cancers occur. In particular, among the receptor tyrosine kinases (RTKs), ROS kinase has been shown to inhibit various types of CNS cancers, including meningiomas, astrocytomas, and glioblastoma multiforme, And recent studies have shown that overexpression of ROS kinase due to mutations in ROS1 receptor kinase is a cause of non-small cell lung cancer, which accounts for more than 85% of all lung cancers It is revealed. This mutation of ROS receptor kinase has been shown to occur at a very high frequency in a variety of cancer cells, suggesting that ROS1 kinase is a useful target for cancer therapy.

However, to date, no drug has been marketed as a ROS kinase inhibitor, and staurosporine as a compound exhibiting ROS kinase inhibitory activity, but development of a compound exhibiting a suitable inhibitory activity for the treatment of cancer is continuously required.

Accordingly, the present inventors have made efforts to develop compounds exhibiting an inhibitory activity of ALK (Anaplastic lymphoma kinase) and ROS1 kinase inhibitory activity, and have found that pyrimidine-2,4-diamine derivatives of a specific structure (ALK) and ROS1 kinase, and it can be useful as a prophylactic or therapeutic agent for cancer as a pharmaceutical composition containing the same. The present invention has been completed based on this finding.

WO 2004080980 A1 WO 2005016894 A1

It is an object of the present invention to provide a pyrimidine-2,4-diamine derivative, an optical isomer thereof, or a pharmaceutically acceptable salt thereof.

Another object of the present invention is to provide a process for preparing the pyrimidine-2,4-diamine derivative.

Yet another object of the present invention is to provide a pharmaceutical composition for preventing or treating cancer comprising the pyrimidine-2,4-diamine derivative, an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

It is another object of the present invention to provide a pharmaceutical composition for preventing or treating osteoporosis due to activity of anaplastic lymphoma kinase (ALK) comprising the pyrimidine-2,4-diamine derivative, an optical isomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient And to provide a pharmaceutical composition for the prevention or treatment of the diseases caused.

Another object of the present invention is to provide a pharmaceutical composition for preventing or treating diseases caused by overexpression of ROS1 kinase comprising the pyrimidine-2,4-diamine derivative, an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient To provide a composition.

Yet another object of the present invention is to provide an inhibitor of reversed-type lymphoma kinase (ALK) which comprises as an active ingredient a pyrimidine-2,4-diamine derivative, an optical isomer thereof, or a pharmaceutically acceptable salt thereof.

Another object of the present invention is to provide an inhibitor of ROS1 containing as an active ingredient a pyrimidine-2,4-diamine derivative, an optical isomer thereof, or a pharmaceutically acceptable salt thereof.

In order to achieve the above object,

The present invention provides a compound represented by the following general formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof.

[Chemical Formula 1]

Figure 112016072414212-pat00001

(In the formula 1,

X a and X b are independently hydrogen or halogen;

R 1 is alkylene of C 1-5 which is connected to a hydrogen, a linear or branched C 1-5 alkyl, or Y 2;

R 2 is straight or branched C 1-5 alkyl unsubstituted or substituted with one or more halogens;

Figure 112016072414212-pat00002
Is a single bond or a double bond;

Y 1 is nitrogen (N), Y 2 is CR 6 , Y 3 is nitrogen (N), or Y 1 is carbon (C), Y 2 is nitrogen (N), Y 3 is oxygen R < 6 > is hydrogen or a bond linked to Y < 2 >;

Y 4 is CH, oxygen (O), NR 3 or N + R 4 R 5 ,

Wherein R 3 is hydrogen, an unsubstituted or at least one hydroxyl group substituted linear or branched C 1-5 alkyl, unsubstituted C 1-5 alkyl or one or more hydroxy groups substituted with linear or branched carbonyl, or dimethyl Aminomethylcarbonyl, R 4 and R 5 are independently straight or branched C 1-5 alkyl;

Y < 5 > is CH or NH; And

When Y 1 is carbon (C), Y 2 is nitrogen (N), and Y 3 is oxygen (O), the ratio between Y 1 and Y 2

Figure 112016072414212-pat00003
Is a double bond and between Y 2 and Y 3
Figure 112016072414212-pat00004
Is a single bond,

When Y 1 is nitrogen (N), Y 2 is CR 6 , and Y 3 is nitrogen (N), the relationship between Y 1 and Y 2

Figure 112016072414212-pat00005
Is a single bond and between Y 2 and Y 3
Figure 112016072414212-pat00006
Is a double bond.

Also, as shown in the following Reaction Scheme 1,

Reacting a compound represented by the formula (2) with a compound represented by the formula (3) to prepare a compound represented by the formula (1) (step 1):

[Reaction Scheme 1]

Figure 112016072414212-pat00007

(In the above Reaction Scheme 1,

X a , X b , R 1 , R 2 , Y 1 , Y 2 , Y 3 , Y 4 , Y 5 ,

Figure 112016072414212-pat00008
Is as defined in the above formula (1)).

Further, the present invention provides a pharmaceutical composition for preventing or treating cancer comprising the compound represented by the formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

The present invention also provides an inhibitor of Anaplastic Lymphoma Kinase (ALK) comprising the compound represented by Formula 1, an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

Furthermore, the present invention relates to a pharmaceutical composition for the treatment of diseases caused by an activity of anaplastic lymphoma kinase (ALK) comprising the compound represented by the formula (1), an optical isomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient Or a pharmaceutically acceptable salt thereof.

The present invention also provides a pro-oncogene tyrosine-protein kinase (ROS) inhibitor comprising the compound of Formula 1, an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

Furthermore, the present invention relates to the use of a compound represented by the above formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient and a prophylactic or therapeutic agent for a disease caused by the activity of ROS kinase (Proto-oncogene tyrosine-protein kinase) Or a pharmaceutically acceptable salt thereof.

The compounds according to the present invention are remarkably effective in inhibiting reverse transcription-forming lymphoma kinase (ALK) activity. Therefore, the therapeutic effect of the compounds according to the present invention on cancer cells having an inverted lymphoma kinase (ALK) fusion protein such as EML4-ALK and NPM-ALK And can be effectively used as a pharmaceutical composition for prevention or treatment of cancer since it is expected to be effective in preventing recurrence of cancer.

FIG. 1 is a graph showing the results of observation of cancer size in nude mice injected with a CD74-ROS1 (WT) cell line over time in the drug administration control group, LDK378 treatment group and Example 13 treatment group.
FIG. 2 is a graph showing the results of observing the size of cancer in nude mice injected with the CD74-ROS1 G2032R cell line over time in the drug-treated control group, LDK378 treated group, and Example 13 treated group.
FIG. 3 is a graph showing the results of observing the size of cancer in the drug-treated control group, the LDK378 treated group, and the compound treated group of Example 1 with time.
FIG. 4 is a graph showing the results of observing the size of cancer in the drug-treated control group (control) LDK378 treated group, the treated group of Example 13 and the compound treated group of Example 14 over time.

Hereinafter, the present invention will be described in detail.

The present invention provides a compound represented by the following general formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof.

[Chemical Formula 1]

Figure 112016072414212-pat00009

In Formula 1,

X a and X b are independently hydrogen or halogen;

R 1 is alkylene of C 1-5 which is connected to a hydrogen, a linear or branched C 1-5 alkyl, or Y 2;

R 2 is straight or branched C 1-5 alkyl unsubstituted or substituted with one or more halogens;

Figure 112016072414212-pat00010
Is a single bond or a double bond;

Y 1 is nitrogen (N), Y 2 is CR 6 , Y 3 is nitrogen (N), or Y 1 is carbon (C), Y 2 is nitrogen (N), Y 3 is oxygen R < 6 > is hydrogen or a bond linked to Y < 2 >;

Y 4 is CH, oxygen (O), NR 3 or N + R 4 R 5 ,

Wherein R 3 is hydrogen, an unsubstituted or at least one hydroxyl group substituted linear or branched C 1-5 alkyl, unsubstituted C 1-5 alkyl or one or more hydroxy groups substituted with linear or branched carbonyl, or dimethyl Aminomethylcarbonyl, R 4 and R 5 are independently straight or branched C 1-5 alkyl;

Y < 5 > is CH or NH; And

When Y 1 is carbon (C), Y 2 is nitrogen (N), and Y 3 is oxygen (O), the ratio between Y 1 and Y 2

Figure 112016072414212-pat00011
Is a double bond and between Y 2 and Y 3
Figure 112016072414212-pat00012
Is a single bond,

When Y 1 is nitrogen (N), Y 2 is CR 6 , and Y 3 is nitrogen (N), the relationship between Y 1 and Y 2

Figure 112016072414212-pat00013
Is a single bond and between Y 2 and Y 3
Figure 112016072414212-pat00014
Is a double bond.

In this case, N + R 4 R 5 may exist in the form of a salt or may exist in the form of an anion such as I - , preferably a halide, a tosylate, and the like, Methane sulfonic acid, sulfuric acid and the like, and an anion capable of forming a common amine salt is included in the present invention.

Preferably,

X a and X b are independently hydrogen, fluoro or chloro;

R 1 is alkylene of C 1-3 which is connected to a hydrogen, a linear or branched C 1-3 alkyl, or Y 2;

R 2 is linear or branched C 1-3 alkyl unsubstituted or substituted with one or more halogens;

Figure 112016072414212-pat00015
Is a single bond or a double bond;

Y 1 is nitrogen (N), Y 2 is CR 6 , Y 3 is nitrogen (N), or Y 1 is carbon (C), Y 2 is nitrogen (N), Y 3 is oxygen R < 6 > is hydrogen or a bond linked to Y < 2 >;

Y 4 is CH, oxygen (O), NR 3 or N + R 4 R 5 ,

R 3 is hydrogen, straight or branched C 1-3 alkyl unsubstituted or substituted with at least one hydroxy group, straight or branched C 1-3 alkylcarbonyl which is unsubstituted or substituted by at least one hydroxy group, or dimethyl Aminomethylcarbonyl, R 4 and R 5 are independently straight or branched C 1-3 alkyl;

Y < 5 > is CH or NH; And

When Y 1 is carbon (C), Y 2 is nitrogen (N), and Y 3 is oxygen (O), the ratio between Y 1 and Y 2

Figure 112016072414212-pat00016
Is a double bond and between Y 2 and Y 3
Figure 112016072414212-pat00017
Is a single bond,

When Y 1 is nitrogen (N), Y 2 is CR 6 , and Y 3 is nitrogen (N), the relationship between Y 1 and Y 2

Figure 112016072414212-pat00018
Is a single bond and between Y 2 and Y 3
Figure 112016072414212-pat00019
Is a double bond.

More preferably,

X a is hydrogen or chloro;

X b is fluoro or chloro;

R 1 is hydrogen, methyl or C 2 alkylene linked to Y 2 ;

R 2 is methyl, ethyl, isopropyl or -CHF 2 ;

Figure 112016072414212-pat00020
Is a single bond or a double bond;

Y 1 is nitrogen (N), Y 2 is CR 6 , Y 3 is nitrogen (N), or Y 1 is carbon (C), Y 2 is nitrogen (N), Y 3 is oxygen R < 6 > is hydrogen or a bond linked to Y < 2 >;

Y 4 is carbon (C), oxygen (O), NR 3 or N + R 4 R 5 ,

Wherein R 3 is hydrogen, methyl, ethyl, propyl, isopropyl, methylcarbonyl, hydroxymethylcarbonyl, dimethylaminomethylcarbonyl or hydroxyethyl, and R 4 and R 5 are independently ethyl;

Y < 5 > is CH or NH; And

When Y 1 is carbon (C), Y 2 is nitrogen (N), and Y 3 is oxygen (O), the ratio between Y 1 and Y 2

Figure 112016072414212-pat00021
Is a double bond and between Y 2 and Y 3
Figure 112016072414212-pat00022
Is a single bond,

When Y 1 is nitrogen (N), Y 2 is CR 6 , and Y 3 is nitrogen (N), the relationship between Y 1 and Y 2

Figure 112016072414212-pat00023
Is a single bond and between Y 2 and Y 3
Figure 112016072414212-pat00024
Is a double bond.

Preferable examples of the compound represented by the formula (1) according to the present invention include the following compounds.

(1) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(2) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropoxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;

(3) Synthesis of 5-chloro-N4- (5-chloro-2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(4) Synthesis of 5-chloro-N4- (4-chloro-2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(5) Synthesis of 5-chloro-N4- (3-chloro-2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(6) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- 4- (5,6-dihydro- 1H) -yl) phenyl) pyrimidine-2,4-diamine;

(7) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropoxy-4- (5,6-dihydro- (1H) -yl) phenyl) pyrimidine-2,4-diamine;

(8) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(9) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(10) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;

(11) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (9-methoxy-2,3,5,6-tetrahydro- Azino [4,3-a] quinolin-8-yl) pyrimidine-2,4-diamine;

(12) 5-Chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (9-methoxy-3-methyl-2,3,5,6-tetrahydro- , 4] triazino [4,3-a] quinolin-8-yl) pyrimidine-2,4-diamine;

(13) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(14) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;

(15) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazine-l (4H) -yl) -2- (dimethylamino) ethan-1-one;

(16) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazin-l (4H) -yl) -2-hydroxyethan-l-one;

(17) Synthesis of 2- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazin-1 (4H) -yl) ethan-l-ol;

(18) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine hydrochloride;

(19) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-ethoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(20) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-difluoromethoxy- , 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;

(21) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-ethoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(22) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-difluoromethoxy- -Triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(23) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(24) 5- fluoro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;

(25) 5-Fluoro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropoxy- -Triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(26) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- (2,4-triazine-4 (1H) -yl) phenyl) pyrimidine-2,4-diamine iodide;

(27) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;

(28) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;

(29) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- ) -5,6-dihydro-l, 2,4-triazine-l (4H) -yl-2-hydroxyethan-l-one;

(30) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazine-l (4H) -yl-2-hydroxyethan-l-one;

(31) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazine-l (4H) -yl-2-dimethylaminoethan-l-one;

(32) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;

(33) 5-Chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;

(34) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (4- (5,6-dihydro- -2-methoxyphenyl) pyrimidine-2,4-diamine;

(35) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (4- (5,6-dihydro- -2-isopropoxyphenyl) pyrimidine-2,4-diamine; And

(36) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (4- (5,6-dihydro- -2-methoxyphenyl) pyrimidine-2,4-diamine.

The pyrimidine-2,4-diamine derivative represented by the above formula (1) of the present invention can be used in the form of a pharmaceutically acceptable salt. Examples of the salt include pharmaceutically acceptable acid addition salts formed by free acid Salts are useful. Acid addition salts include those derived from inorganic acids such as hydrochloric acid, nitric acid, phosphoric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, nitrous acid or phosphorous acid, and aliphatic mono- and dicarboxylates, phenyl-substituted alkanoates, hydroxyalkanoates, Derived from organic acids such as acetic acid, benzoic acid, citric acid, lactic acid, maleic acid, gluconic acid, methanesulfonic acid, 4-toluenesulfonic acid, tartaric acid, fumaric acid, and the like. Such pharmaceutically innocuous salts include, but are not limited to, sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate chloride, bromide, Butyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, succinate, maleic anhydride, maleic anhydride, , Sebacate, fumarate, maleate, butyne-1,4-dioate, hexane-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, Methoxybenzoate, phthalate, terephthalate, benzene sulfonate, toluene sulfonate, chlorobenzene sulfide Propyl sulphonate, naphthalene-1-yne, xylenesulfonate, phenylsulfate, phenylbutyrate, citrate, lactate,? -Hydroxybutyrate, glycolate, maleate, Sulfonate, naphthalene-2-sulfonate or mandelate.

The acid addition salt according to the present invention may be prepared by dissolving the pyrimidine-2,4-diamine derivative in an organic solvent, for example, methanol, ethanol, acetone, methylene chloride, acetonitrile, etc., Filtration and drying of the resulting precipitate, or by distillation of the solvent and excess acid under reduced pressure, followed by drying or crystallization in an organic solvent.

In addition, bases can be used to make pharmaceutically acceptable metal salts. The alkali metal or alkaline earth metal salt is obtained, for example, by dissolving the compound in an excess amount of an alkali metal hydroxide or an alkaline earth metal hydroxide solution, filtering the insoluble compound salt, and evaporating and drying the filtrate. At this time, it is preferable for the metal salt to produce sodium, potassium or calcium salt. In addition, the corresponding salt is obtained by reacting an alkali metal or alkaline earth metal salt with a suitable salt (such as silver nitrate).

Furthermore, the present invention encompasses the compounds represented by the formula (1) and pharmaceutically acceptable salts thereof as well as solvates, optical isomers and hydrates thereof which can be prepared therefrom.

Also, as shown in the following Reaction Scheme 1,

Reacting a compound represented by the formula (2) with a compound represented by the formula (3) to prepare a compound represented by the formula (1) (step 1):

[Reaction Scheme 1]

Figure 112016072414212-pat00025

In the above Reaction Scheme 1,

X a , X b , R 1 , R 2 , Y 1 , Y 2 , Y 3 , Y 4 , Y 5 ,

Figure 112016072414212-pat00026
Is as defined in the above formula (1).

Hereinafter, a method for preparing the compound represented by Formula 1 according to the present invention will be described in detail.

In the process for preparing a compound represented by the following formula 1 according to the present invention, the above step 1 is a step for preparing a compound represented by the formula 1 by reacting a compound represented by the formula 2 with a compound represented by the formula 3, Specifically, the compound represented by the formula (2) is subjected to an alkylation reaction with a compound represented by the formula (3) under an organic solvent and a base to prepare a compound represented by the formula (1).

Wherein the solvent is selected from the group consisting of tetrahydrofuran; Dioxane; Ether solvents including ethyl ether, 1,2-dimethoxyethane and the like; Lower alcohols including methanol, ethanol, propanol and butanol; Dimethylformamide (DMF), dimethylsulfoxide (DMSO); Butyrate, glycolate, maleate, tartrate, glutarate, glutarate, glutarate, glutarate, glutaraldehyde, glutaraldehyde, glutaraldehyde, glutaraldehyde, Methane sulfonate, propane sulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, mandelate and the like.

The base may be an organic base such as pyridine, triethylamine, N, N-diisopropylethylamine (DIPEA), or 1,8-diazabicyclo [5.4.0] -7-anthesene (DBU); Or inorganic bases such as sodium hydroxide, sodium carbonate, potassium carbonate, cesium carbonate, sodium hydride and the like may be used in an equivalent amount or in an excess amount.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing the compound represented by the general formula (1) according to the present invention, the process for producing the compound represented by the general formula (3) in the step 1 can be produced through the examples of the following Schemes 2 to 4, It is not.

[Reaction Scheme 2]

Figure 112016072414212-pat00027
;

[Reaction Scheme 3]

Figure 112016072414212-pat00028
; And

[Reaction Scheme 4]

Figure 112016072414212-pat00029

In the above Reaction Schemes 2 to 4,

R 1 and R 2 and R 3 are the same as defined in the above formula (1).

In the process for preparing the compound of Formula 3 according to the present invention, Reaction Schemes 2 to 4 are an example of preparing the compound of Formula 3 required for the above Step 1.

Wherein the solvent is selected from the group consisting of tetrahydrofuran; Dioxane; Ether solvents including ethyl ether, 1,2-dimethoxyethane and the like; Lower alcohols including methanol, ethanol, propanol and butanol; Dichloromethane, dimethylformamide (DMF), dimethylsulfoxide (DMSO); Butyrate, glycolate, maleate, tartrate, glutarate, glutarate, glutarate, glutarate, glutaraldehyde, glutaraldehyde, glutaraldehyde, glutaraldehyde, Methane sulfonate, propane sulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, mandelate and the like.

In addition, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing the compound represented by the general formula (1) according to the present invention, the step (1) may be carried out by dissolving the compound represented by the general formula (2) in an organic solvent, adding the compound represented by the general formula Followed by stirring to obtain a compound represented by the formula (1).

Wherein the solvent is selected from the group consisting of tetrahydrofuran; Dioxane; Ether solvents including ethyl ether, 1,2-dimethoxyethane and the like; Lower alcohols including methanol, ethanol, propanol and butanol; Dichloromethane, dimethylformamide (DMF), dimethylsulfoxide (DMSO); Butyrate, glycolate, maleate, tartrate, glutarate, glutarate, glutarate, glutarate, glutaraldehyde, glutaraldehyde, glutaraldehyde, glutaraldehyde, Methane sulfonate, propane sulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, mandelate and the like.

In addition, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

Production method 1 of starting material (compound represented by formula (3)) 1

The compound represented by Formula 3, which is the starting material of Reaction Scheme 1, can be prepared and used as shown in Reaction Scheme 2 below.

Reacting a compound represented by the general formula (4) with aminoethanol and protecting the alcohol group with a methanesulfonyl group to obtain a compound represented by the general formula (5) (step 1);

(Step 2) of reacting methanesulfonyl with hydrazine to the compound of formula 5 obtained in step 1 to obtain the compound of formula 6;

A step (3) of obtaining a compound represented by the formula (7) by reductive alkylation of the compound represented by the formula (6) obtained in the step 2 in the presence of an acid catalyst;

(Step 4) of reducing the nitro group to an amino group by successive hydrogenation reactions in the presence of an acid in the presence of an acid in the presence of an acid to obtain a compound represented by the formula 8 ≪ / RTI >

[Reaction Scheme 2]

Figure 112016072414212-pat00030

In the above Reaction Scheme 2,

R 1 , R 2 , and R 3 are the same as defined in Formula 1 above.

Hereinafter, the method for preparing the compound represented by the above formula (3) will be described step by step.

In the process for preparing a compound represented by the general formula (3) according to the present invention, the step (1) is a step of reacting a compound represented by the general formula (4) with amino ethanol and then protecting the alcohol group with a methanesulfonyl group to obtain a compound represented by the general formula to be.

Examples of the reaction solvent include ether solvents such as tetrahydrofuran, dioxane, dichloromethane (DCM) and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing a compound represented by the general formula (3) according to the present invention, the above step (2) is a step for obtaining a compound represented by the general formula (6) by reacting methanesulfonyl with hydrazine to the compound represented by the general formula (5)

The acid used herein may be hydrochloric acid, sulfuric acid, methanesulfonic acid, polyphosphoric acid, nitric acid, or the like, preferably nitric acid. .

The reaction solvent may be an ether-based solvent such as acetic anhydride, nitromethane, tetrahydrofuran, dioxane, dichloromethane (DCM), or 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination, and acetic anhydride and nitromethane may be preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing a compound represented by the general formula (3) according to the present invention, the step (3) is a step of subjecting the compound represented by the general formula (6) obtained in the step 2 to a reduction alkylation reaction in the presence of an acid catalyst to obtain a compound represented by the general formula .

As the acid usable herein, hydrochloric acid, sulfuric acid, methanesulfonic acid, acetic acid, polyphosphoric acid, nitric acid and the like can be used, and nitric acid can be preferably used .

The reaction solvent may be an ether-based solvent such as acetic anhydride, nitromethane, tetrahydrofuran, dioxane, dichloromethane (DCM), or 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination, and acetic anhydride and nitromethane may be preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing the compound represented by the general formula (3) according to the present invention, the step (4) is a step of reacting the compound represented by the general formula (7) obtained in the step 3 with a nitro group as an amino group To obtain a compound represented by the formula (8).

Examples of the base usable herein include organic bases such as pyridine, triethylamine, N, N-diisopropylethylamine and DBU; It may be equivalent or an excess amount of; NaOH, Na 2 CO 3, K 2 CO 3, an inorganic base such as Cs 2 CO 3.

At this time, a palladium catalyst (Pd / C) can be used as the catalyst that can be used.

The reaction solvent may be methanol; Ether solvents such as tetrahydrofuran, dioxane, dichloromethane (DCM) and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile and the like can be used, and methanol is preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

Production method 2 (starting material) of the starting material

The compound represented by Formula 3, which is the starting material of Reaction Scheme 1, can be prepared and used as shown in Reaction Scheme 3 below.

Reacting a compound represented by the formula (9) with aminoethanol to obtain a compound represented by the formula (10) (step 1);

Reacting a compound represented by the formula (10) obtained in the step 1 with phthalimide to obtain a compound represented by the formula (11) (step 2);

Reacting the compound of Formula 11 obtained in Step 2 with a hydrazine solution to obtain an amine compound of Formula 12 (Step 3);

(Step 4) of reducing the nitro group to an amino group by successive hydrogenation reactions in the presence of an acid in the presence of an acid in the presence of an acid to obtain a compound represented by the formula 13 ≪ / RTI >

[Reaction Scheme 3]

Figure 112016072414212-pat00031

In Scheme 3,

R 1 and R 2 are the same as defined in the above formula (1).

Hereinafter, the method for preparing the compound represented by the above formula (3) will be described step by step.

In the process for preparing the compound represented by the general formula (3) according to the present invention, the step (1) is a step of reacting the compound represented by the general formula (4) with amino ethanol to obtain the compound represented by the general formula (10).

Examples of the reaction solvent include ether solvents such as tetrahydrofuran, dioxane, dichloromethane (DCM) and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing the compound represented by the general formula (3) according to the present invention, the step (2) is a step of reacting the compound represented by the general formula (10) obtained in the above step 1 with phthalimide to obtain the compound represented by the general formula (11).

The acid used herein may be hydrochloric acid, sulfuric acid, methanesulfonic acid, polyphosphoric acid, nitric acid, or the like, preferably nitric acid. .

The reaction solvent may be an ether-based solvent such as acetic anhydride, nitromethane, tetrahydrofuran, dioxane, dichloromethane (DCM), or 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination, and acetic anhydride and nitromethane may be preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing the compound represented by the general formula (3) according to the present invention, the step (3) is a step for obtaining the amine compound represented by the general formula (12) by reacting the compound represented by the general formula (11) obtained in the step 2 with hydrazine.

As the acid usable herein, hydrochloric acid, sulfuric acid, methanesulfonic acid, acetic acid, polyphosphoric acid, nitric acid and the like can be used, and nitric acid can be preferably used .

The reaction solvent may be an ether-based solvent such as acetic anhydride, nitromethane, tetrahydrofuran, dioxane, dichloromethane (DCM), or 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination, and acetic anhydride and nitromethane may be preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing the compound represented by the general formula (3) according to the present invention, the amine compound represented by the general formula (12) obtained in the step 3 is subjected to intramolecular cyclization in the presence of an acid, To obtain a compound represented by the formula (13).

Examples of the base usable herein include organic bases such as pyridine, triethylamine, N, N-diisopropylethylamine and DBU; It may be equivalent or an excess amount of; NaOH, Na 2 CO 3, K 2 CO 3, an inorganic base such as Cs 2 CO 3.

At this time, a palladium catalyst (Pd / C) can be used as the catalyst that can be used.

The reaction solvent may be methanol; Ether solvents such as tetrahydrofuran, dioxane, dichloromethane (DCM) and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile and the like can be used, and methanol is preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

Preparation method 3 of the starting material (compound represented by formula (3)) 3

The compound represented by Formula 3, which is the starting material of Reaction Scheme 1, can be prepared and used as shown in Reaction Scheme 3 below.

Reacting a compound represented by the formula (14) with phthalimide to obtain a compound represented by the formula (15) (step 1);

A step (step 2) of reacting an imide group of the compound represented by the formula (15) obtained in the step 1 with a hydrazine solution to obtain an amine compound represented by the formula (16);

Cyclizing the compound of formula 16 and the nitrile compound 17 obtained in step 2 in the presence of an acid catalyst to obtain a compound of formula 18;

(Step 4) of reducing the nitro group to an amino group by hydrogenation to obtain the compound represented by the formula 19 (step 4).

[Reaction Scheme 4]

Figure 112016072414212-pat00032

In Scheme 4,

R 1 and R 2 are the same as defined in the above formula (1).

Hereinafter, the method for preparing the compound represented by the above formula (3) will be described step by step.

In the process for preparing a compound represented by the general formula (3) according to the present invention, the step (1) is a step of reacting a compound represented by the general formula (14) with phthalimide to obtain a compound represented by the general formula (15).

Examples of the reaction solvent include ether solvents such as tetrahydrofuran, dioxane, dichloromethane (DCM) and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the method for preparing the compound represented by the general formula (3) according to the present invention, the step (2) is a step of reacting the imide group of the compound represented by the formula (15) obtained in the step 1 with the hydrazine solution to obtain the compound represented by the formula .

The acid used herein may be hydrochloric acid, sulfuric acid, methanesulfonic acid, polyphosphoric acid, nitric acid, or the like, preferably nitric acid. .

The reaction solvent may be an ether-based solvent such as acetic anhydride, nitromethane, tetrahydrofuran, dioxane, dichloromethane (DCM), or 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination, and acetic anhydride and nitromethane may be preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for producing the compound represented by the general formula (3) according to the present invention, in the step (3), the compound represented by the formula (16) obtained in the above step 2 and the nitrile compound represented by the compound (17) Is obtained.

As the acid usable herein, hydrochloric acid, sulfuric acid, methanesulfonic acid, acetic acid, polyphosphoric acid, nitric acid and the like can be used, and nitric acid can be preferably used .

The reaction solvent may be an ether-based solvent such as acetic anhydride, nitromethane, tetrahydrofuran, dioxane, dichloromethane (DCM), or 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile, etc. may be used alone or in combination, and acetic anhydride and nitromethane may be preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

In the process for preparing the compound represented by the general formula (3) according to the present invention, the step (4) is a step of reacting the compound represented by the general formula (12) obtained in the step 3 with an amino group To obtain a compound represented by the formula (13).

Examples of the base usable herein include organic bases such as pyridine, triethylamine, N, N-diisopropylethylamine and DBU; It may be equivalent or an excess amount of; NaOH, Na 2 CO 3, K 2 CO 3, an inorganic base such as Cs 2 CO 3.

At this time, a palladium catalyst (Pd / C) can be used as the catalyst that can be used.

The reaction solvent may be methanol; Ether solvents such as tetrahydrofuran, dioxane, dichloromethane (DCM) and 1,2-dimethoxyethane; Dimethylformamide (DMF), dimethylsulfoxide, acetonitrile and the like can be used, and methanol is preferably used.

Further, the reaction is preferably carried out at a temperature of 0 ° C between the boiling point of the solvent, and the reaction time is not particularly limited, but preferably 0.5-10 hours.

The present invention also provides a pharmaceutical composition for preventing or treating cancer comprising the compound represented by the formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

Further, the present invention provides a health functional food for preventing or ameliorating a cancer containing the compound represented by the above-mentioned formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

At this time, the health functional food for preventing or ameliorating cancer is prepared as a normal health functional food containing the compound represented by the formula 1 of the present invention, its optical isomer, or a pharmaceutically acceptable salt thereof as an active ingredient And forms, forms or administration forms known to those skilled in the art are included in the scope of the present invention and are included in the health functional foods of the present invention as long as they can be recognized as health functional foods.

The present invention also relates to the use of an anaplastic lymphoma kinase (ALK) comprising an active ingredient of the compound represented by the formula (1), an optical isomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient, Or a pharmaceutically acceptable salt thereof.

Furthermore, the present invention relates to the use of ROS1 (Proto-oncogene tyrosine-protein kinase ROS) containing the compound represented by the formula (1), an optical isomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient, Or a pharmaceutically acceptable salt thereof.

The present invention also provides an inhibitor of Anaplastic Lymphoma Kinase (ALK) comprising the compound represented by Formula 1, an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

Further, the present invention provides a ROS1 (proto-oncogene tyrosine-protein kinase ROS) inhibitor containing the compound represented by the above-mentioned formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.

The pharmaceutical composition and the inhibitor according to the present invention are characterized by inhibiting the activity of anaplastic lymphoma kinase (ALK) and inhibiting the expression and growth of cancer cells.

Also, the pharmaceutical composition and the inhibitor according to the present invention are characterized by inhibiting the activity of ROS1 (Proto-oncogene tyrosine-protein kinase ROS) and inhibiting the expression and growth of cancer cells.

The inverse Lymphoma kinase (ALK) induces cell proliferation in cancer cells present in cancer cells, and the gene fusion process activates inverse lymphoma kinase (ALK), and in this case, inverse lymphoma kinase (ALK) (ALK) is not normal, nor is it caused by cancer. In addition, it has been reported that the tyrosine kinase, which acts as an antagonist of lymphocytic leukemia, induces proliferation of cells, inhibits apoptosis to prevent cell death, rearranges the cell skeleton, Interacts with other tyrosine kinases that have been genetically engineered, or activates a variety of other pathways.

As a result of experiments conducted to measure the inhibitory activity of the compound of formula (I) according to the present invention on the proliferation inhibitory activity of lymphoma kinase (ALK), most of the compounds of the examples according to the present invention were found to be inactivated by lymphocyte kinase (ALK) L1196M, an non-small cell lung cancer cell containing an enzyme activity and a reverse forming lymphoma kinase (ALK) enzyme, was superior in the ability to inhibit the cell activity (see Table 2 in Experimental Example 1).

In addition, experiments were performed to determine the cytotoxicity of BaF3 EML4-ALK L1196M and BaF3 EML4-ALK WT cells of the compound of formula (1) according to the present invention. As a result, most of the compounds according to the present invention were BaF3 The cytotoxic IC 50 values were low in BaF3 EML4-ALK L1196M cells resistant to EML4-ALK WT (wild-type) cells and crizotinib (see Table 3 of Experimental Example 2).

Therefore, the compound represented by the formula (1) according to the present invention can be used for preventing or treating cancer by inhibiting the activity of the reverse forming lymphoma kinase (ALK). For example, there is provided a method of treating a disease selected from the group consisting of non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, endometrioid sarcoma, myofiber neoplasia, breast cancer, gastric cancer, lung cancer, melanoma, large B-cell lymphoma, Squamous cell cancer, uterine cancer, prostate cancer, and the like.

On the other hand, ROS1 is a cause of various CNS cancers such as meningiomas, astrocytomas, and glioblastoma multiforme, and overexpression due to mutations of ROS1 receptor kinase occurs in all lung cancers The non-small cell lung cancer is a cause of refractory non-small cell lung cancer which accounts for 85% or more. The compound represented by the formula (1) or the pharmaceutical composition containing the compound of the present invention can inhibit ROS1 activity, Can be useful for preventing or treating the diseases caused. For example, there is provided a method of treating a disease selected from the group consisting of non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, endometrioid sarcoma, myofiber neoplasia, breast cancer, gastric cancer, lung cancer, melanoma, large B-cell lymphoma, Squamous cell carcinoma, uterine cancer, prostate cancer and the like can be confirmed by experiments of the present invention.

In order to measure the inhibitory activity of ROS1 on the proliferation of the compound of formula 1 according to the present invention in the enzyme step and to examine the inhibitory effect on cancer cell proliferation, this embodiment the compounds according to the invention can be confirmed to exhibit a markedly lower concentration than the IC 50 (LDK-378) it can be seen that excellent inhibitory activity against ROS1 (see experimental example 3-1).

In addition, the compound represented by formula (I) according to the present invention is excellent in the effect of inhibiting ROS1 activity in nude mice to which CD74-ROS1 is applied, and can be used for the treatment of non-small cell lung cancer, neuroblastoma, inflammatory myeloblastic fibroblast , Rhabdomyosarcoma, myofiber oblastoma, breast cancer, stomach cancer, lung cancer, melanoma and the like (see Experimental Example 3-2).

Further, in order to evaluate the inhibitory effect on the cancer cell proliferation of the non-small cell lung cancer cells H2228 and H3122 of the compound represented by the formula (1) according to the present invention, the following compounds according to the present invention were compared with non- It is confirmed that the cell proliferation activity of H2228 and H3122 is effectively reduced (see Experimental Example 4).

In order to evaluate the inhibition of cancer cell proliferation of H3122 phosphorylated lung cancer of the compound represented by the formula (1) according to the present invention, the compound represented by the formula (1) according to the present invention was tested for H3122 phosphorylated lung cancer It is confirmed that the effect of suppressing cancer cell proliferation is excellent (see Experimental Example 5).

Accordingly, the compound represented by formula (I) according to the present invention is useful as a preventive or therapeutic agent for various cancers including non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, endometrial sarcoma, myofiber blastoma, breast cancer, gastric cancer, lung cancer, And the like.

When the composition of the present invention is used as a medicine, the pharmaceutical composition containing the compound represented by the formula (1), its optical isomer, or a pharmaceutically acceptable salt thereof as an active ingredient may be administered orally, The composition may be formulated and administered in a parenteral administration form, but is not limited thereto.

Examples of formulations for oral administration include tablets, pills, light / soft capsules, liquids, suspensions, emulsions, syrups, granules, elixirs and troches, (E.g., silica, talc, stearic acid and its magnesium or calcium salts and / or polyethylene glycols), such as, for example, dextrose, sucrose, mannitol, sorbitol, cellulose and / or glycine. The tablets may also contain binders such as magnesium aluminum silicate, starch paste, gelatin, methylcellulose, sodium carboxymethylcellulose and / or polyvinylpyrrolidine and may optionally contain additives such as starch, agar, alginic acid or its sodium salt A disintegrating or boiling mixture and / or an absorbent, a colorant, a flavoring agent and a sweetening agent.

The pharmaceutical composition containing the compound represented by the formula (1) as an active ingredient may be administered parenterally, and the parenteral administration may be by subcutaneous injection, intravenous injection, intramuscular injection, or intrathoracic injection.

In order to formulate the composition for parenteral administration, the compound of Formula 1, its optical isomer, or a pharmaceutically acceptable salt thereof is mixed with water or a stabilizer or a buffer to prepare a solution or suspension, Or vial unit dosage forms. The composition may be sterilized and / or contain adjuvants such as preservatives, stabilizers, wettable or emulsifying accelerators, salts for the control of osmotic pressure and / or buffers, and other therapeutically useful substances, Or a coating method.

The dosage of the pharmaceutical composition containing the compound represented by the formula (1) as an active ingredient may vary depending on the patient's age, weight, sex, dosage form, health condition and disease severity, preferably 0.01 To 1000 mg / kg / day, depending on the judgment of a physician or pharmacist, by administering an oral or parenteral route by dividing the period of time by several times a day, preferably once or three times a day.

Hereinafter, a method for preparing the compound represented by the formula (1) according to the present invention will be described in detail with reference to the following Examples or Examples. The following examples are merely illustrative of the method of preparing the compound represented by the above formula (1), but the present invention is not limited thereto. The preparation methods described by the following examples can be obtained by using synthetic conditions well-known in the art of organic synthesis, suitable reagents and the like.

< Example  1 > 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (1- methyl -5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00033

Amine (314 mg, 0.91 mmol) was dissolved in THF (15 mL), and 2-methoxy (2-methoxyphenyl) (200 mg, 0.91 mmol) was added, and the azeotrope (52.5 mg, 0.09 &lt; RTI ID = 0.0 & mmol), Cs 2 CO 3 ( 888 mg, 2.73 mmol), was Pd (OAc) 2 (10 mg , 0.0454 mmol was added) the sequence, after degassing at 100 to charge the nitrogen stirred for 18 hours. After completion of the reaction, the reaction mixture was extracted with EA / H 2 O, and the organic layer was dried over MgSO 4 , filtered and concentrated. Purification by column chromatography (IPA: MC) gave the desired compound (163 mg, 31%).

(M, 2H), 7.92 (d, J = 7.9 Hz, 1 H), 8.56 (d, J = 2H), 3.90 (s, 3H), 3.84 (m, 2H), 7.65-7.58 (m, t, J = 4.8 Hz, 2H), 3.25-3.19 (m, 1H), 3.01 (t, t = 4.8 Hz, 2H), 2.80 (s, 3H), 1.31 (d, J = 6.9 Hz, 6H);

LC / MS 530.51 (M &lt; + & gt ; + H).

< Example  2> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Isopropoxy -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00034

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- (1-methyl-5,6-dihydro-1,2,4-triazin-4 (1H) -yl) aniline was used as the starting material to obtain the target compound .

1H NMR (300 MHz, CD 3 OD) δ 9.56 (s, 1H), 8.56 (d, J = 8.4 Hz, 1H), 8.22-8.15 (m, 2H), 7.92 (d, J = 7.9 Hz, 1H) , 7.65-7.58 (m, 1 H), 7.44 (s, 1 H), 7.31-7.22 (m, , 3.87-3.78 (m, 2H), 3.31-3.20 (m, 1H), 3.01 (t, J = 4.8 Hz, 2H) 1.31 (d, J = 6.9 Hz, 6 H);

LC / MS 557.79 (M &lt; + & gt ; + H).

< Example  3> 5- Chloro - N4 - (5- Chloro -2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00035

Instead of using 2,5-dichloro-N- (2- (isopropylsulfonyl) phenyl) pyrimidin-4-amine, 2,5-dichloro- Trifluoromethyl-phenyl) pyrimidin-4-amine was used in place of the compound obtained in Example 1, the target compound was obtained.

8.16 (s, 1 H), 8.09 (d, J = 8.7 Hz, 1 H), 7.83 (d, J = 1.1 Hz). 1H NMR (300 MHz, CDCl 3 )? J = 8.6 Hz, 1H), 7.44 (s, 1H), 7.25-7.17 (m, 2H), 6.68 (dd, J = J = 6.8 Hz, 1H), 3.91 (s, 3H), 3.84 (t, J = 4.8 Hz, 2H), 3.23 ), 1.32 (d, J = 6.8 Hz, 6H);

LC / MS 564.1 (M &lt; + & gt ; ).

< Example  4> 5- Chloro - N4 -(4- Chloro -2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00036

Instead of using 2,5-dichloro-N- (2- (isopropylsulfonyl) phenyl) pyrimidin-4-amine, 2,5-dichloro- Trifluoromethyl-phenyl) pyrimidin-4-amine was used in place of the compound obtained in Example 1, the target compound was obtained.

1H NMR (300 MHz, CDCl 3 ) δ 9.50 (s, 1H), 8.56 (d, J = 8.9 Hz, 1H), 8.15-8.09 (m, 2H), 7.89 (d, J = 2.5 Hz, 1H), 2H), 3.90 (s, 3H), 3.84 (t, J = 4.8 Hz, 2H), 7.58-7.53 (m, , 3.27-3.23 (m, 1H), 3.02 (t, J = 4.9 Hz, 2H), 2.81 (s, 3H), 1.35 (d, J = 8.9 Hz, 6H);

LC / MS 564.1 (M &lt; + & gt ; + H).

< Example  5> 5- Chloro - N4 - (3- Chloro -2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00037

Instead of using 2,5-dichloro-N- (2- (isopropylsulfonyl) phenyl) pyrimidin-4-amine, 2,5-dichloro- Trifluoromethyl-phenyl) pyrimidin-4-amine was used in place of the compound obtained in Example 1, the target compound was obtained.

1H NMR (300 MHz, CDCl 3 ) δ 10.53 (s, 1H), 8.58 (d, J = 8.2 Hz, 1H), 8.57-8.12 (m, 2H), 8.09-7.36 (m, 3H), 7.02 (s (M, 2H), 3.88 (s, 3H), 3.87-3.74 (m, 3H), 3.03-2.99 , &Lt; / RTI &gt; J = 6.8 Hz, 6H);

LC / MS 564.1 (M &lt; + & gt ; ).

< Example  6> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (5,6-dihydro-1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00038

Instead of using 2,5-dichloro-N- (2- (isopropylsulfonyl) phenyl) pyrimidin-4-amine, tert- Synthesis of tert-butyl 4- (4 - (((4-fluoropyridin-2-yl) Amino) -3-methoxyphenyl) -5,6-dihydro-1,2,4-triazolo [3,3-d] pyrimidin- Azine-1 (4H) -carboxylate (40 mg, 0.064 mmol) was dissolved in dichloromethane (0.5 mL), 1.5 mL of 4M HCl-dissolved dioxane was added, and the mixture was stirred at room temperature for 5 hours. , The HCl solution was removed under reduced pressure to obtain the desired compound (22 mg, 63%).

1H NMR (300 MHz, CD 3 OD) δ 8.56 (s, 1H), 8.27-8.21 (m, 2H), 8.02-7.98 (m, 1H), 7.80-7.74 (m, 3H), 7.57-7.55 (m 2H), 7.16 (s, 1H), 6.89-6.80 (m, IH), 4.19-4.10 (m, IH), 3.92 (s, 3H), 3.64-3.55 , &Lt; / RTI &gt; 1H), 1.31-1.12 (m, 6H);

LC / MS 517.2 (M &lt; + & gt ; + H).

< Example  7> 5- Chloro -N4- (2- ( Isopropylsulfonyl ) Phenyl) -N2- (2- Isopropoxy (5,6-dihydro-1,2,4-triazin-4 (1H) -yl) phenyl) pyrimidine-2,4-diamine

Figure 112016072414212-pat00039

butyl 4 - ((4-amino-3-methoxyphenyl) -5,6-dihydro-1,2,4-triazine- Dihydro-l, 2,4-triazine-1 (4H) -carboxylate was used as the starting material, To obtain the target compound.

1H NMR (300 MHz, CDCl 3 ) δ 8.57 (s, 1H), 8.34 (s, 1H), 8.17 (d, J = 9 Hz, 2H), 8.02 (d, J = 9 Hz, 1H), 7.8 ( (s, 1H), 7.20 (s, 1H), 6.84 (d, J = 3 Hz, 2H), 4.18 3.91 (s, 2H), 3.41 (s, 3H), 1.35 (d, J = 6 Hz, 6H), 1.22 (d, J = 6 Hz, 6H);

LC / MS 544.1 (M &lt; + & gt ; + H).

< Example  8> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (1-ethyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00040

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- 1-ethyl-5,6-dihydro-1,2,4-triazin-4 (1H) -yl) aniline was used as the starting material.

1H NMR (300 MHz, CDCl 3 ) δ 9.56 (s, 1H), 8.55 (d, J = 8.5 Hz, 1H), 8.17-8.14 (m, 2H), 7.92 (d, J = 7.8 Hz, 1H), 2H), 3.89 (s, 3H), 3.86-3.83 (m, 2H), 7.62 (t, J = 7.8 Hz, 1H) 2H), 3.26-3.21 (m, 1H), 3.06-2.94 (m, 4H), 2.04 (m, 1H), 1.32-1.24 (m, 12H);

LC / MS 544.5 (M &lt; + & gt ; + H).

< Example  9> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (1-propyl-5, 6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00041

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- 1-propyl-5,6-dihydro-1,2,4-triazin-4 (1H) -yl) aniline was used as the starting material.

1H NMR (300 MHz, CDCl 3 ) δ 9.58 (s, 1H), 8.53 (d, J = 8.3 Hz, 1H), 8.33-8.22 (m, 2H), 8.16 (s, 1H), 7.92 (d, J 1H), 7.70-7.63 (m, 2H), 7.52 (s, 1H), 7.35-7.28 2H), 1.86-1. 26 (m, 2H), 1.36-1.24 (m, 8H (m, 2H) ), 1.05-0.98 (m, 3H);

LC / MS 558 (M &lt; + & gt ; + H).

< Example  10> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (l-isopropyl-5, 6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00042

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- 1-isopropyl-5,6-dihydro-1,2,4-triazin-4 (1H) -yl) aniline was used as the starting material to obtain the target compound .

1H NMR (300 MHz, CDCl 3 ) δ 9.56 (s, 1H), 8.55 (d, J = 8.4 Hz, 1H), 8.15 (s, 2H), 7.92 (d, J = 7.8 Hz, 1H), 7.61 ( 1H), 7.37 (s, 1H), 7.29-7.26 (m, 1H), 6.57-6.54 (m, 2H), 3.89-3.85 m, 2H), 1.31 (d, J = 6.6 Hz, 6H), 1.21 (d, J = 6.2 Hz, 6H);

LC / MS 558 (M &lt; + & gt ; + H).

< Example  11> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 - (9- Methoxy -2,3,5,6-te TRA Hydro-lH- [l, 2,4] Triazino [4,3-a] quinolin-8-yl) pyrimidine-2,4-diamine

Figure 112016072414212-pat00043

Instead of using 2-methoxy-4- (l -methyl-5,6-dihydro-l, 2,4-triazin-4 (lH) -yl) aniline, 9- , 5,6-tetrahydro-1H-pyrazin o [1, 2-a] quinolin-8-amine was used as the starting material.

(D, J = 6.9 Hz, 6H), 2.60-2.51 (m, 2H), 2.73-2.62 (m, 2H), 3.32-3.18 (m, (s, 3H), 6.38 (s, IH), 7.29-7.22 (m, IH), 7.33 (s, 2H) 1H), 7.63-7.55 (m, IH), 7.89 (s, IH), 7.92 (dd, J = 8.0, 1.5 Hz, ), 9.53 (s, 1 H);

LC / MS 542.17 (M &lt; + &gt; + H).

< Example  12> 5-Chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (9-methoxy- 4] triazino [4,3-a] quinolin-8-yl) pyrimidine-2,4-diamine

Figure 112016072414212-pat00044

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazine-4 (lH) -yl) aniline, 9- -2,3,5,6-tetrahydro-1H-pyrazin o [1, 2-a] quinolin-8-amine was used in place of 2-chloro-

(M, 2H), 2.79 (s, 3H), 3.00 (t, 3H) 1H, J = 5.1 Hz, 2H), 3.31-3.17 (m, 1H), 3.81-3.72 (m, 2H), 3.89 (s, (D, J = 8.0, 1.4 Hz, 1H), 8.13 (s, 1H), 8.55 (d, J = 7.2 Hz, 1H) , J = 8.4 Hz, 1 H), 9.53 (s, 1 H);

LC / MS 556.18 (M &lt; + &gt; + H).

< Example  13> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -5- methyl -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00045

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazine-4 (lH) -yl) aniline 2-methoxy- (1H) -yl) -4- (l-methyl-5,6-dihydro-l, 2,4-triazin- Compound.

1H-NMR (300 MHz, CDCl 3) δ 9.54 (s, 1H), 8.53 (d, J = 8.3 Hz, 1H), 8.17 (s, 1H), 8.12 (s, 1H), 7.96-7.91 (m, (S, 3H), 3.69 (s, 3H), 3.69 (s, 3H) (t, J = 4.8 Hz, 2H), 3.30-3.21 (m, 1H), 2.98 (t, J = 4.8 Hz, 2H) J = 6.8 Hz, 6H);

LC / MS 544.07 (M &lt; + & gt ; + H).

<Example 14> 5-chloro-N4 - (2- (isopropyl) phenyl) - N2 - (2- isopropyl proxy Foxy-5-methyl-4- (1-methyl-5,6-dihydro- -1,2,4 -triazin- 4 (1H) -yl ) phenyl ) pyrimidine-2,4-diamine

Figure 112016072414212-pat00046

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- The procedure of Example 1 was repeated except that methyl-4- (1 -methyl-5,6-dihydro-1,2,4-triazin-4 (1H) To obtain the target compound.

1H-NMR (300 MHz, CDCl 3) δ 9.56 (s, 1H), 8.49 (d, J = 7.8 Hz, 1H), 8.23-8.19 (m, 2H), 7.94 (d, J = 7 Hz, 1H) , 7.65 (m, 2H), 7.47 (s, IH), 7.32-7.27 (m, IH), 6.72 (m, 3H), 3.02 (s, 3H), 2.10 (s, 3H), 1.41 - 1.31 (m, 14H);

LC / MS 572.1 (M &lt; + & gt ; ).

< Example  15 >. 1- (4- (4 - ((5- Chloro -4 - ((2- ( Isopropylsulfonyl ) Phenylamino ) Pyrimidin-2-yl) amino-3- Methoxyphenyl ) -5,6- Dihydro -1,2,4- Triazine -1 (4H) -yl) -2- (dimethylamino) ethan-1-one

Figure 112016072414212-pat00047

(2-methoxy-4- (5,6-dihydro-1, 2,4-tri (30 mg, 0.058 mmol) was dissolved in THF (1 mL), diisopropylamine (0.026 mL, 0.14 mmol), HATU (33.2 mg, 0.087 mmol) and dimethylglycine (9 mg, 0.087 mmol) at 0 ° C, and the mixture was stirred at room temperature for 10 hours. The reaction solution was diluted with water, extracted three times with dichloromethane, and washed with a saturated aqueous sodium chloride solution. The organic layer was dried over anhydrous magnesium sulfate, concentrated under reduced pressure, and purified by column chromatography to obtain the desired compound (4.88 mg, 13%).

1H-NMR (500 MHz, CDCl 3) δ 9.61 (s, 1H), 8.62-8.56 (m, 1H), 8.29 (d, J = 9.3Hz, 1H), 8.19 (s, 1H), 7.96 (d, 1H, J = 7.9 Hz, 1H), 7.67 (t, J = 7.8 Hz, 1H), 7.47 (s, 1H), 7.34-7.31 2H), 3.59 (s, 2H), 3.26-3.22 (m, 1H), 2.44 (s, 3H) (s, 6H), 1.35 (d, J = 6.9 Hz, 6H);

LC / MS 600.9 (M &lt; + & gt ; ).

< Example  16> 1- (4- (4 - ((5- Chloro -4 - ((2- ( Isopropylsulfonyl ) Phenylamino ) Pyrimidin-2-yl) amino-3- Methoxyphenyl ) -5,6- Dihydro -1,2,4- Triazine -1 (4H) -yl) -2- Heide Preparation of &lt; RTI ID = 0.0 &gt;

Figure 112016072414212-pat00048

The procedure of Example 15 was repeated, except that glycolic acid was used instead of dimethylglycine to obtain the desired compound.

1H-NMR (300 MHz, CDCl 3) δ 8.43 (d, J = 8.3Hz, 1H), 8.16 (s, 1H), 7.93 (d, J = 6.8Hz, 1H), 7.86-7.69 (m, 1H) , 7.75-7.67 (m, IH), 7.46-7.37 (m, IH), 7.29 (s, IH), 6.87 (d, J = 2.3Hz, 1H), 6.73-6.67 , 2H), 4.03 (t, J = 4.9 Hz, 2H), 3.92 (s, 3H), 3.83 (t, J = 4.9 Hz, 2H), 1.25 (d, J = 6.8 Hz, 6H);

LC / MS 574.1 (M &lt; + & gt ; ).

< Example  17 2- (4- (4 - ((5- Chloro -4 - ((2- ( Isopropylsulfonyl ) Phenylamino ) Pyrimidin-2-yl) amino-3- Methoxyphenyl ) -5,6- Dihydro -1,2,4- Triazine -1 (4H) -yl) ethan-1-ol

Figure 112016072414212-pat00049

The objective compound was obtained in the same manner as in Example 15, except that 2-iodo-alcohol was used instead of dimethyl glycine.

1H-NMR (300 MHz, CDCl 3) δ 9.57 (s, 1H), 8.56 (d, J = 9.0Hz, 1H), 8.21 (d, J = 9.0Hz, 1H), 8.16 (s, 1H), 7.93 (d, J = 1.3, 7.9 Hz, IH), 7.64 (t, J = 7.9 Hz, IH), 7.41 (s, IH), 7.31-7.28 J = 4.9 Hz, 2H), 3.29-3.18 (m, 1H), 3.10 (t, J = 4.9 Hz, 2H), 3.02 (t, J = 4.5 Hz, 2H), 1.32 (d, J = 6.9 Hz, 6H);

LC / MS 560.7 (M &lt; + & gt ; ).

< Example  18 > 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (1- Me Dihydro-1,2,4-triazin-4 (1H) -yl) phenyl) pyrimidine-2,4-diamine Of hydrochloride  Produce

Figure 112016072414212-pat00050

N2- (2-methoxy-4- (1-methyl-5,6-dihydro-1,2 Pyridin-2,4-diamine (200 mg, 0.37 mmol) was dissolved in dichloromethane (2 mL), and 0.19 mL of 4M HCl-dissolved dioxane was added And the mixture was stirred at room temperature for 5 hours. After the reaction was completed, the HCl solution was removed under reduced pressure to obtain the desired compound (170 mg, 81%).

1H-NMR (300 MHz, DMSO -d 6) δ 9.81 (s, 1H), 8.88 (s, 1H), 8.65 (s, 1H), 8.40 (d, J = 7.8 Hz, 1H), 8.36 (s, J = 7.5 Hz, 1H), 7.21 (m, 1H), 6.92 (dd, J = 8.8, 2.1 Hz), 7.85-7.86 (m, 2H), 2.83 (s, 3H), 1.15 (d, 7 Hz, 1H), 4.00-3.97 (m, 2H), 3.88 6H);

LC / MS 530.1 (M &lt; + & gt ; ).

< Example  19> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Ethoxy -5- methyl -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00051

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazine-4 (lH) -yl) aniline, (1H) -yl) -4- (l-methyl-5,6-dihydro-l, 2,4-triazin- Compound.

1H-NMR (300 MHz, CDCl 3) δ 9.53 (s, 1H), 8.53 (d, J = 8.31 Hz, 1H), 8.18 (s, 1H), 8.10 (s, 1H), 7.94 (dd J = 7.86 (S, 1H), 6.65 (s, 1H), 4.08 (q, J = 7.00 Hz, 1H), 7.62 (t, J = 7.88 Hz, 2H), 3.68 (t, J = 4.85 Hz, 2H), 3.30-3.21 (m, 1.46 (t, J = 6.96 Hz, 3H), 1.32 (d, J = 6.87 Hz, 6H);

LC / MS 558.2 (M &lt; + & gt ; + H).

< Example  20> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Difluoromethoxy -5- methyl -4- (1- methyl -5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) &Lt; / RTI &gt; pyrimidine-2,4-diamine

Figure 112016072414212-pat00052

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin-4 (lH) -yl) aniline, 2-difluoromethoxy- (1H) -yl) aniline was used as the starting material in place of (1-methyl-5,6-dihydro-1,2,4-triazine-4 To obtain the target compound.

1H-NMR (300 MHz, CDCl 3) δ 9.61 (s, 1H), 8.49 (d, J = 8.46 Hz, 1H), 8.19 (s, 2H), 7.93 (dd, J = 7.89 Hz, 1.41 Hz, 1H J = 7.9 Hz, 2H), 3.72 (s, 1H), 6.69 (s, 3,15 (m, 1H), 2.97 (t, J = 4.78 Hz, 2H), 2.80 (s, 3H), 2.19 (s, 3H) 1.32 (dd, J = 6.87 Hz, 3.03 Hz, 6H);

LC / MS 580.2 (M &lt; + & gt ; + H).

< Example  21> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Ethoxy -4- (1- Me Yl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00053

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- 1-methyl-5,6-dihydro-1,2,4-triazin-4 (1H) -yl) aniline was used as a starting material.

1H-NMR (300 MHz, CDCl 3) δ 9.56 (s, 1H), δ 8.57 (d, 1H, J = 9 Hz), 8.17 ~ 8.55 (m, 2H), 7.93 (d, 1H, J = 9 Hz 1H), 7.41 (s, 1H), 7.30-7.26 (m, 2H), 7.17 (s, 2H, J = 6.9 Hz), 3.25 (t, 2H, J = 4.5 Hz), 3.24 (m, 3H, J = 6 Hz), 1.32 (d, 6H, J = 6 Hz);

LC / MS 544.2 (M &lt; + & gt ; + H).

< Example  22> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Difluoromethoxy -4- (1- methyl -5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine Manufacturing

Figure 112016072414212-pat00054

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin-4 (lH) -yl) aniline, 2-difluoromethoxy- - (1 -methyl-5,6-dihydro-1,2,4-triazin-4 (1H) -yl) aniline was used in place of .

1H-NMR (300 MHz, CDCl 3) δ 8.51 (d, 1H, J = 9 Hz), 8.19 (d, 1H, J = 9 Hz), 8.15 (s, 1H), 7.92 (d, 1H, J = 1H, J = 6 Hz), 7.60 (t, 1H, J = 6 Hz), 7.60 1H), 6.82 (s, 1H), 6.84 (t, 1H, J = 75 Hz), 3.81 (m, d, 6H, J = 6 Hz);

LC / MS 566.1 (M &lt; + & gt ; + H).

< Example  23> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -5- methyl -4- (5,6-dihydro-1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00055

5-methyl-4- (1-acetyl-5, 6-dihydro-1, 2, 4-dihydroxy- (2- (isopropylsulfonyl) phenyl) -N2- (2- (4-fluorophenyl) pyridin- Dihydro-l, 2,4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine was used in place of methoxy- , The target compound was obtained by carrying out the same processes as in the Example 27 below.

1H NMR (300 MHz, CDCl 3 ) δ 9.63 (s, 1H), 8.51 (d, 1H, J = 9 Hz), 8.20 ~ 8.15 (m, 2H), 7.92 (d, 1H, J = 6 Hz), 1H, J = 9 Hz), 7.17 (s, 1H), 7.12 (s, 1H), 6.85-6.82 (m, 2H), 6.54 2H, J = 4.5 Hz), 3.28-3.19 (m, 2H), 3.02 (t, 2H, J = 6 Hz), 2.81 (s, 3H), 1.32 (d, 6H, J = 6 Hz);

LC / MS 530.1 (M &lt; + & gt ; + H).

< Example  24> 5- Fluoro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00056

Instead of using 2, 5-dichloro-N- (2- (isopropylsulfonyl) phenyl) pyrimidin- Phenyl) pyrimidin-4-amine was used as the starting material, the target compound was obtained.

1H NMR (300 MHz, CDCl 3 ) δ 9.05 (s, 1H), 8.65 (d, J = 8.4 Hz, 1H), 8.20 (d, J = 9.3 Hz, 1H), 8.05 (d, J = 2.8 Hz) , 7.89 (d, J = 7.9 Hz, 1 H), 7.64 (t, J = 8.5 Hz, 1 H), 7.33 , 6.60 (t, J = 2.4 Hz, IH), 6.58 (s, IH), 3.91 (s, 3H), 3.84 (t, J = 4.8 Hz, 2H), 3.28-3.19 t, J = 5.0 Hz, 2H), 2.81 (s, 3H), 1.32 (d, J = 6.8 Hz, 6H);

LC / MS 514.2 (M &lt; + & gt ; + H).

< Example  25> 5- Fluoro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Isopropoxy -4- (l-methyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00057

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- The procedure of Example 24 was repeated except for using (1-methyl-5,6-dihydro-1,2,4-triazin-4 (1H) Compound.

1H-NMR (300 MHz, CDCl 3) δ 9.43 (s, 1H), 8.44 (d, J = 8.4 Hz, 1H), 8.24-8.17 (m, 2H), 7.95 (m 1H), 7.82-7.66 (m , 7.36 (s, IH), 7.17-7.12 (m, IH), 7.10 (s, IH), 6.61-6.52 (m, 2H), 4.78-4.62 , 2H), 3.31-3.20 (m, 1H), 3.22 (m, 2H), 2.82 (s, 3H), 1.40 (m, 6H), 1.35 (d, J = 6.9 Hz, 6H);

LC / MS 542.2 (M &lt; + & gt ; + H).

< Example  26> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Isoproxoxy -5-methyl-4- (1,1'- Diethyl -5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4-diamine Iodide  Produce

Figure 112016072414212-pat00058

5-methyl-4- (5,6-dihydro-1,2 (isopropylsulfonyl) phenyl) -N2- (49.2 mg, 0.09 mmol) was dissolved in ethanol (1 mL), diisopropylamine (0.024 mL, 0.14 mmol) and iodine Ethane (0.014 mL, 0.14 mmol) was added thereto, followed by stirring at room temperature for 18 hours. The reaction solution was diluted with water, extracted three times with dichloromethane, and washed with a saturated aqueous sodium chloride solution. The organic layer was dried over anhydrous magnesium sulfate, concentrated under reduced pressure, and purified by column chromatography to obtain the desired compound (26 mg, 48%).

1H NMR (300 MHz, CDCl 3 ) δ 9.54 (s, 1H), 8.50 (d, J = 8.4 Hz, 1H), 8.20 (d, J = 6.0 Hz, 1H), 7.94 (d, J = 7.8 Hz, J = 6.0 Hz, 1H), 7.74-7.67 (m, 2H), 7.35-7. 20 (m, 2H), 7.14 ), 4.23-4.09 (m, 2H), 3.88-3.63 (m, 4H), 3.34-3.13 (m, = 6.0 Hz, 6H), 1.32 (d, J = 6.9 Hz, 6H);

LC / MS 614.2 (M &lt; + & gt ; ).

< Example  27> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Isoproxoxy -5-methyl-4- (5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00059

(2-isopropoxy-5-methyl-4- (1-acetyl-5,6-dihydro-naphthalen- Yl) phenyl) pyrimidine-2,4-diamine (331 mg, 0.55 mmol) was dissolved in methanol / water (3 ml), KOH (47 mg, 0.83 mmol) And the mixture was stirred at 80 ° C for 3 days. The reaction solution was diluted with water, extracted three times with dichloromethane, and washed with a saturated aqueous sodium chloride solution. The organic layer was dried over anhydrous magnesium sulfate, concentrated under reduced pressure, and then purified by column chromatography to obtain the desired compound (114 mg, 39%).

1H-NMR (500 MHz, CDCl 3) δ 9.53 (s, 1H), 8.53 (d, J = 8.4 Hz, 1H), 8.18 (s, 1H), 8.12 (s, 1H), 7.94 (dd, J = 1H, J = 8.7 Hz, 1H), 7.58 (s, 1H), 7.35-7.72 (m, 2H), 3.37-3.16 (m, 3H), 2.12 (s, 3H), 1.39 (d, J = 6.0 Hz, 6H), 4.55 (q, J = 6.0 Hz, ), 1.32 (d, J = 6.9 Hz, 6H);

LC / MS 558.2 (M &lt; + & gt ; + H).

< Example  28> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Isoproxoxy -5-methyl-4- (1-acetyl-5, 6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) &Lt; / RTI &gt; pyrimidine-2,4-diamine

Figure 112016072414212-pat00060

Instead of using 2-methoxy-4- (l-methyl-5,6-dihydro-l, 2,4-triazin- Methyl-4- (1-acetyl-5,6-dihydro-1,2,4-triazin-4 (1H) -yl) aniline was used in place of To obtain the target compound.

1H-NMR (500 MHz, CDCl 3) δ 9.55 (s, 1H), 8.53 (d, J = 8.4 Hz, 1H), 8.18 (d, J = 3.6 Hz, 2H), 7.94 (d, J = 7.8 Hz 1H), 7.70-7.53 (m, 2H), 7.36-7.20 (m, IH), 6.71 (s, IH), 6.64 (s, IH), 4.57 (quint, (t, J = 4.8 Hz, 2H), 3.59 (t, J = 4.8 Hz, 2H), 3.26 d, J = 6.0 Hz, 6H), 1.30 (d, J = 8.7 Hz, 6H);

LC / MS 600.2 (M &lt; + & gt ; + H).

< Example  29> 1- (4- (4 - ((5- Chloro -4 - ((2- ( Isopropylsulfonyl ) Phenylamino ) Pyrimidin-2-yl) amino-5- Isopropoxy -2- Methylphenyl ) -5,6- Dihydro -1,2,4- Triazine -1 (4H) -yl-2- Hydroxyethane -1-one

Figure 112016072414212-pat00061

The objective compound was obtained in the same manner as in Example 26 except that glycolic acid was used instead of iodinated ethane.

1H-NMR (300 MHz, CDCl 3) δ 9.56 (s, 1H), 8.53 (d, J = 8.4 Hz, 1H), 8.19 (s, 2H), 7.94 (d, J = 6.9 Hz, 1H), 7.70 2H), 7.35-7.20 (m, 1H), 6.74 (d, J = 9.6 Hz, 1H), 6.63 2H), 3.70-3.53 (m, 2H), 3.26 (quint, J = 6.9 Hz, 1H), 2.11 (s, 3H), 1.39 6.6 Hz, 6H);

LC / MS 616.2 (M &lt; + & gt ; + H).

< Example  30> 1- (4- (4 - ((5- Chloro -4 - ((2- ( Isopropylsulfonyl ) Phenylamino ) Pyrimidin-2-yl) amino-5- Methoxy -2- Methylphenyl ) -5,6- Dihydro -1,2,4- Triazine -1 (4H) -yl-2-h Droxie Carbonitrile

Figure 112016072414212-pat00062

Dihydro-l, 2,4-triazin-4 (lH) - (2-methoxy- Phenyl) -N2- (2-methoxy-5-methyl-4- ((2-methoxyphenyl) Dihydro-l, 2,4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine was used in place of To obtain the target compound.

1H-NMR (300 MHz, CDCl 3) δ 9.57 (s, 1H), 8.54 (d, 1H, J = 9 Hz), 8.19 (s, 2H), 7.95 (d, 6H, J = 9 Hz), 7.65 (t, 1H, J = 4.5 Hz), 7.55 (s, IH), 7.31 ~ 7.26 (m, 3H), 6.73 (s, IH), 6.62 1H), 3.40 (s, 1H), 3.64 (t, 1H, J = 4.5 Hz), 3.36 (s, ), 1.32 (d, 6H, J = 6 Hz);

LC / MS 588.1 (M &lt; + & gt ; + H).

< Example  31> 1- (4- (4 - ((5- Chloro -4 - ((2- ( Isopropylsulfonyl ) Phenylamino ) Pyrimidin-2-yl) amino-5- Methoxy -2- Methylphenyl ) -5,6- Dihydro -1,2,4- Triazine -1 (4H) -yl-2-di Methyl amino Carbonitrile

Figure 112016072414212-pat00063

Dihydro-l, 2,4-triazin-4 (lH) - (2-methoxy- Phenyl) -N2- (2-methoxy-5-methyl-4- ((2-methoxyphenyl) Dihydro-l, 2,4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine was used in place of To obtain the target compound.

1H-NMR (300 MHz, CDCl 3) δ 9.56 (s, 1H), 8.54 (d, 1H, J = 9 Hz), 8.19 (s, 2H), 7.95 (d, 1H, J = 9 Hz), 7.31 2H), 6.62 (s, 1H), 6.62 (s, 1H), 4.06 (t, 3.30 to 3.21 (m, 1H), 2.45 (s, 6H), 2.14 (s, 3H), 1.32 (d, 6H, J = 6 Hz);

LC / MS 615.2 (M &lt; + & gt ; + H).

< Example  32> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Isoproxoxy -5-methyl-4- (1-ethyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) &Lt; / RTI &gt; pyrimidine-2,4-diamine

Figure 112016072414212-pat00064

(2-isopropoxy-5-methyl-4- (5,6-dihydro-1, 2-isopropylphenyl) Pyridin-2,4-diamine (331 mg, 0.55 mmol) was dissolved in methanol (1 mL), and acetaldehyde (0.01 mL, 0.14 mmol) and acetic acid ) And sodium hydride (9 mg, 0.14 mmol) as a cyan coating, and the mixture was stirred at room temperature for 5 hours. The reaction solution was diluted with water, extracted three times with dichloromethane, and washed with a saturated aqueous sodium chloride solution. The organic layer was dried over anhydrous magnesium sulfate, concentrated under reduced pressure, and then purified by column chromatography to obtain the desired compound (24 mg, 45%).

8.13 (d, J = 8.1 Hz, 1H), 8.17 (s, 1H), 8.11 (s, 1H), 7.93 J = 6.0 Hz, 1H), 3.78-3.53 (m, 2H), 7.70-7.50 (m, (m, 4H), 3.26 (quint, J = 6.9 Hz, 1H), 3.04-2.89 (m, 2H), 1.45-1.18 (m, 15H);

LC / MS 586.2 (M &lt; + & gt ; + H).

< Example  33> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 -(2- Methoxy -5- methyl -4- (1-ethyl-5,6- Dihydro -1,2,4- Triazine -4 (1H) -yl) Phenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00065

5-methyl-4- (5,6-dihydro-l, 2,4-triazine-benzoimidazol- 4-yl) phenyl) pyrimidine-2,4-diamine instead of 5-chloro-4- (lH- (4-fluorophenyl) pyrimidine-2,4-diamine was used in place of the compound obtained in Example 32 To give the title compound.

1H, J = 9 Hz), 8.17 (s, 1H), 8.11 (d, 1H, J = 3 Hz), 7.94 (s, 1H, J = 9 Hz), 7.64 (t, IH, J = 7.5 Hz), 7.50 (s, IH), 7.30-7.26 (M, 3H), 2.15 (s, 3H), 1.32 (d, 2H), 3.67 (s, , 6H, J = 6 Hz), 1.26-1.20 (m, 1H);

LC / MS 557.1 (M &lt; + & gt ; + H).

< Example  34> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 - (4- (5,6- Dihydro -41H-l, 2,4-oxadiazin-4-yl) -2- Methoxyphenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00066

Amine (334 mg, 0.96 mmol) was dissolved in THF (15 mL), and then 4- (5-dichlorobenzyl) 4-yl) -2-methoxyaniline (200 mg, 0.96 mmol) was added and the mixture was treated with AztFor (56 mg, 0.096 mmol), Cs 2 CO 3 (944 mg, 2.89 mmol) and Pd (OAc) 2 (10 mg, 0.048 mmol) were successively added thereto, followed by degassing with nitrogen and stirring at 110 ° C for 18 hours. After completion of the reaction, the mixture was extracted with EA / H2O, and the organic layer was dried over MgSO4, filtered and concentrated. Purification by column chromatography (IPA: MC) gave the desired compound (10 mg, 2%).

1H NMR (300 MHz, CD 3 OD) δ 9.65 (s, 1H), 8.78 (d, J = 9.1 Hz, 1H), 8.29 (s, 1H), 7.89-7.85 (m, 1H), 7.61-7.56 ( 1H), 7.40-7.18 (m, 1H), 6.64-6.81 (m, 1H), 6.71-6.67 4.55 (m, 2H), 3.87-3.78 (m, 5H), 3.31-3.20 (m, 1H), 1.34-1.27 (m, 6H);

LC / MS 517.1 (M &lt; + & gt ; + H).

< Example  35> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 - (4- (5,6- Dihydro -41H-l, 2,4-oxadiazin-4-yl) -2- Isopropoxyphenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00067

Instead of using 4- (5,6-dihydro-4H-1,2,4-oxadiazin-4-yl) -2- methoxyaniline, 4- 1,2,4-oxadiazin-4-yl) -2-isopropoxyaniline was used in place of the compound obtained in Example 34.

1H NMR (300 MHz, CDCl 3 ) δ 8.38 (d, J = 8.2Hz, 1H), 8.02 (s, 1H), 7.94 (dd, J = 7.9, 1.4Hz, 1H), 7.81 (s, 1H), 2H), 4.63-4.53 (m, 1H), 4.28 (m, 2H), 7.71 (d, J = 8.5 Hz, 1H), 7.52-7.44 t, J = 4.5 Hz, 2H), 3.83 (t, J = 4.5 Hz, 2H), 3.27-3.17 (m, 1H), 1.40-1.29 (m, 12H);

LC / MS 544.9 (M &lt; + & gt ; ).

< Example  36> 5- Chloro - N4 -(2-( Isopropylsulfonyl ) Phenyl ) - N2 - (4- (5,6- Dihydro -41H-l, 2,4-oxadiazin-3-yl) -2- Methoxyphenyl ) Pyrimidine-2,4- Diamine  Produce

Figure 112016072414212-pat00068

Amine (167 mg, 0.48 mmol) was dissolved in THF (9 mL), and then 4- (5-dichlorobenzyl) 4-yl) -2-methoxyaniline (100 mg, 0.48 mmol) was added and the mixture was treated with AztFor (28 mg, 0.0483 mmol), Cs2CO3 (472 mg, 1.45 mmol) and Pd (OAc) 2 (5 mg, 0.024 mmol) were successively added thereto, followed by degassing with nitrogen, followed by stirring at 100 ° C for 18 hours. After completion of the reaction, the mixture was extracted with EA / H2O, and the organic layer was dried over MgSO4, filtered and concentrated. Purification by column chromatography (IPA: MC) gave the desired compound (48 mg, 19%).

1H NMR (300 MHz, CD 3 OD) δ 9.53 (s, 1H), 8.51 (d, J = 8.4Hz, 1H), 8.32 (d, J = 8.4Hz, 1H), 8.16 (s, 1H), 7.91 (d, J = 7.9 Hz, 1H), 7.72-7.64 (m, 2H), 7.34-7.25 3.99 (m, 2H), 3.91 (s, 3H), 3.65-3.57 (m, 2H), 3.31-3. 18 (m, 1H), 1.30 (d, J = 6.8 Hz, 6H);

LC / MS 517.4 (M &lt; + & gt ; + H).

< Comparative Example  1> Crizotinib ( Crizotinib )

Figure 112016072414212-pat00069

Crizotinib was prepared by a known method.

< Comparative Example  2> LDK Manufacture of -378

Figure 112016072414212-pat00070

LDK-378 was prepared by a known method.

The chemical structures of the compounds prepared in Examples 1-36 are summarized in Table 1 below.

Example rescue Example rescue One

Figure 112016072414212-pat00071
19
Figure 112016072414212-pat00072
2
Figure 112016072414212-pat00073
20
Figure 112016072414212-pat00074
3
Figure 112016072414212-pat00075
21
Figure 112016072414212-pat00076
4
Figure 112016072414212-pat00077
22
Figure 112016072414212-pat00078
5
Figure 112016072414212-pat00079
23
Figure 112016072414212-pat00080
6
Figure 112016072414212-pat00081
24
Figure 112016072414212-pat00082
7
Figure 112016072414212-pat00083
25
Figure 112016072414212-pat00084
8
Figure 112016072414212-pat00085
26
Figure 112016072414212-pat00086
9
Figure 112016072414212-pat00087
27
Figure 112016072414212-pat00088
10
Figure 112016072414212-pat00089
28
Figure 112016072414212-pat00090
11
Figure 112016072414212-pat00091
29
Figure 112016072414212-pat00092
12
Figure 112016072414212-pat00093
30
Figure 112016072414212-pat00094
13
Figure 112016072414212-pat00095
31
Figure 112016072414212-pat00096
14
Figure 112016072414212-pat00097
32
Figure 112016072414212-pat00098
15
Figure 112016072414212-pat00099
33
Figure 112016072414212-pat00100
16
Figure 112016072414212-pat00101
34
Figure 112016072414212-pat00102
17
Figure 112016072414212-pat00103
35
Figure 112016072414212-pat00104
18
Figure 112016072414212-pat00105
36
Figure 112016072414212-pat00106

< Experimental Example  1> Inverse formation  Lymphoma Kinase ( ALK ) Inhibitory activity evaluation

In order to measure the proliferation inhibitory activity of lymphokine kinase (ALK) of the compound represented by formula (1) according to the present invention in the enzyme step, the following experiment was conducted.

In order to measure the inhibitory activity against inverse lymphoma kinase (ALK), the compounds (2 μL) prepared in Examples 1 to 36 were added to a Grayer 96 well round bottom plate, and the reverse forming lymphoma kinase (ALK) enzyme μL) and biotin-adhered peptide substrate (2 μL) were mixed for 15 minutes and cultured. To this was added ATP solution (5 μL) and the kinase reaction was carried out at room temperature for 30 minutes. (XL665) (5 μL) with streptavidin dissolved in ethylenediaminetetraacetic acid solution and 5 μL of anti-phosphotyrosine antibody (5 μL) with europium (Eu 3+ ) (HTRF, Cisbio), and analyzed using a Wallac Envision 2103 instrument in a wavelength range of 615/665 nm. The fluorescence intensity was measured using a fluorescence microscope . The IC 50 of the test compound subjected to the above experiment was shown using a prism (version 5.01, GraphPad) software, and the results are shown in Table 2 below.

Example ALK wt.
IC 50 ([mu] M)
ALK L1196M
IC 50 ([mu] M)
One 0.006 0.04 2 0.003 0.008 3 0.007 <0.01 4 0.05 0.05 5 0.03 0.02 6 0.0048 <0.01 7 0.022 0.05 8 0.008 0.014 9 0.013 0.015 10 0.043 0.012 11 0.033 0.0083 12 0.0017 0.0081 13 0.00095 0.0035 14 0.0049 0.021 15 0.0022 0.001 16 0.0044 0.0048 17 0.0019 0.0039 18 0.0037 0.058 19 <0.01 0.014 20 0.023 0.029 21 0.016 0.036 22 0.021 0.031 23 0.011 0.021 24 0.018 0.093 25 0.073 0.36 26 0.0076 0.0086 27 0.029 0.12 28 0.039 0.12 29 0.018 0.041 30 0.0049 0.0098 31 0.0038 0.0058 32 0.099 0.1 33 0.018 0.042 34 0.006 0.043 35 0.005 0.01 36 0.013 0.05 Comparative Example 1
(crizotinib)
0.036 0.22
Comparative Example 2
(LDK-378)
0.001 0.005

Table 2 shows that the compounds prepared in Examples 1 to 36 according to the present invention all had IC 50 values of 0.01 μM or less and had IC 50 concentrations that were equal or significantly lower than those of Comparative Example 2 (LDK-378) .

Therefore, as shown in Experimental Example 1, the compound represented by Formula 1 according to the present invention is superior as an inhibitor of reversed-type lymphoma kinase (ALK) activity, And is useful as a composition for the prophylaxis or treatment of cancers such as non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, rhabdomyosarcoma, myofiber oblastoma, breast cancer, gastric cancer, lung cancer, melanoma and the like. Lt; / RTI &gt;

< Experimental Example  2> EML4 - By ALK  Transfected BaF3  Cell cytotoxicity assessment

In order to measure the cytotoxicity of BaF3 EML4-ALK L1196M and BaF3 EML4-ALK WT cells of the compound of formula (1) according to the present invention, the following experiment was carried out.

Specifically, a BaF3 EML4-ALK wt cell line stably expressing EML4-ALK wt was prepared by infecting BaF3 cells with EML4-ALK wt (wild-type) gene as a lentivirus. In addition, a BaF3 EML4-ALK L1196M cell line stably expressing EML4-ALK L1196M was prepared by infecting BaF3 cells with EML4-ALK L1196M gene as a lentivirus. The cell numbers of the two cell lines were measured and then injected into each well of a 96-well plate at 4,000 in a volume of 90 μL. The concentration of the compound of Example of the present invention was adjusted to 10 μM, 2 mu] M, 0.4 [mu] M, 0.08 [mu] M, 0.016 [mu] M, 0.0032 [mu] M, 0.00064 [mu] M and 0 [mu] M in each well and incubated for 3 days in a 37 [deg.] C cell incubator. After 3 days, 10 μL of the WST-1 solution was added to each well. When the color of the solution of each well changed, an ELISA (manufacturer: Molecular Devices, Model: EMax Endpoint ELISA Microplate reader) Lt; / RTI &gt; Using the measured values, the amount of cells was calculated to calculate the IC 50 value in the cytotoxicity of each compound. The results are shown in Table 3 below.

Example BaF3
EML4-ALK
L1196M ([mu] M)
BaF3
EML4-ALK
WT ([mu] M)
One 0.02 0.009 2 0.059 0.031 3 0.29 0.05 4 0.92 0.05 5 1.4 0.5 6 0.4 0.13 7 1.7 0.36 8 0.07 0.018 9 0.1 0.058 10 0.15 0.063 11 0.26 0.069 12 0.12 0.047 13 0.027 0.013 14 0.25 0.048 15 0.074 0.021 16 0.058 0.013 17 0.076 0.021 18 0.28 0.036 19 0.062 0.016 20 0.11 0.06 21 0.098 0.057 22 0.3 0.073 23 0.081 0.031 24 0.18 0.037 25 1.6 0.099 26 1.4 0.48 27 0.3 0.084 28 0.11 0.057 29 0.26 0.064 30 0.027 0.012 31 0.057 0.016 32 0.33 0.095 33 0.063 0.019 34 0.47 0.12 35 0.082 0.07 36 0.05 0.012 Comparative Example 1
(crizotinib)
0.88 0.06
Comparative Example 2
(LDK-378)
0.041 0.019

Table 3 shows that most of the compounds according to the present invention have cytotoxic IC 50 values comparable in BaF3 EML4-ALK L1196M cells resistant to BaF3 EML4-ALK WT (wild-type) cells and crizotinib Which is significantly lower than that of Example 1 and Comparative Example 2.

Therefore, as shown in Experimental Example 2, the compound represented by Formula 1 according to the present invention is superior in the effect of inhibiting reverse priming lymphoma kinase activity in BaF3 cells transfected with EML4-ALK, ALK) activity inhibitor as well as the prevention of cancers such as non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, endometrial sarcoma, myoblastoma, breast cancer, gastric cancer, lung cancer and melanoma Or therapeutic compositions of the present invention.

< Experimental Example  3> CD74- ROS1 (wt) and CD74- ROS1 G2032R (mt)  Evaluation of inhibitory activity

In order to measure the inhibitory activity of the compound represented by the formula (1) according to the present invention on the proliferation inhibitory activity of ROS1 at the enzyme level and to examine the inhibitory effect of the cancer cell proliferation, the following experiment was conducted.

&Lt; 3-1 > CD74- By ROS1  Transfected BaF3  Assessment of cytotoxicity against

BaF3 CD74-ROS1 wt cell line stably expressing CD74-ROS1 wt was prepared by infecting BaF3 cells with a CD74-ROS1 wt (wild-type) gene as a lentivirus. In addition, CD74-ROS1 G2032R cell line stably expressing CD74-ROS1 G2032R was prepared by infecting BaF3 cells with a lentivirus of CD74-ROS1 G2032R gene. Cell numbers of the two cell lines were measured using tryphan blue, and the cell numbers of 5 (5 &lt; th &gt; and 5 &lt; th &gt; The cells were divided into 5 x 10 &lt; 5 &gt; cells / 0.1 ml in wells. The concentration of the drug was 0.1 μM, 0.01 μM, and 0.001 μM, and the same concentration of drug was tested in each well for 3 days. Cell viability was then measured using Promega's CellTiter-Glo Luminescent Cell Viability Assay. The mean values of the three experimental values except for the minimum value and the maximum value of the measured values and the IC 50 were obtained. The results are shown in Table 4 below.

Example ROS1 wt.
IC 50 ([mu] M)
ROS1 G2032R
IC 50 ([mu] M)
One 0.002 0.033 13 0.0007 0.005 14 0.003 0.010 16 0.001 0.030 17 0.005 0.050 18 0.0048 0.010 19 0.001 0.03 21 0.004 0.06 22 0.006 0.05 23 0.0009 0.005 24 0.005 0.05 25 0.014 0.03 27 0.024 0.05 28 0.006 0.01 29 0.016 0.05 30 0.0008 0.005 31 0.004 0.01 32 0.048 0.05 33 0.002 0.01 Comparative Example 2
(LDK-378)
0.027 > 0.050

Table 4 shows that the compound of Example according to the present invention exhibits a significantly lower IC 50 concentration than that of Comparative Example 2 (LDK-378), indicating that the compound according to the present invention has excellent inhibitory activity against ROS1 Able to know.

Therefore, as shown in Experimental Example 3-1, the compound of Formula 1 according to the present invention is excellent in the effect of inhibiting ROS1 activity in BaF3 transfected with CD74-ROS1, and thus can be usefully used as an inhibitor of ROS1 activity As well as compositions for the prophylaxis or treatment of cancers such as non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, somatic rhabdomyosarcoma, myofiber blastoma, breast cancer, gastric cancer, lung cancer and melanoma have.

<3-2> CD74- ROS1  applied Balb / C Evaluation of drug efficacy against Nude mice

Five 5-week-old Balb / C mice were divided into two groups. One group was injected subcutaneously with CD74-ROS1 WT 5 × 106 cells and the remaining 15 mice were subcutaneously injected with CD74-ROS1 G2032R 5 × 106 cells. Then, each of 15 animals having similar tumor size mean values was divided into three groups of control (control), LDK378, and Example 13, and each drug was orally administered. The concentration of the drug is 50 mg / kg. The size of cancer was positive by applying the formula: (major axis x speed 2/2) and then using a caliper measurement of long and short axes, the results are shown in Figs.

FIG. 1 is a graph showing the results of observation of cancer size in nude mice injected with a CD74-ROS1 (WT) cell line over time in the drug administration control group, LDK378 treatment group and Example 13 treatment group.

FIG. 2 is a graph showing the results of observing the size of cancer in nude mice injected with the CD74-ROS1 G2032R cell line over time in the drug-treated control group, LDK378 treated group, and Example 13 treated group.

1, the size of the cancer significantly increased with the lapse of time in the control group, and the compound treated group according to Example 13 and the group treated with LDK378 according to the present invention showed a decrease in cancer size, It can be confirmed that the treatment group suppresses the cancer size better than the LDK378 treatment group.

2, the size of the cancer significantly increased with the lapse of time in the control group, and the compound treated group according to Example 13 and the LDK378 treated group according to the present invention showed a decrease in cancer size. In particular, It can be confirmed that the treatment group suppresses the cancer size better than the LDK378 treatment group.

Therefore, as shown in Experimental Example 3-2, the compound represented by Chemical Formula 1 according to the present invention is excellent in the effect of inhibiting ROS1 activity in nude mice to which CD74-ROS1 is applied, and thus can be effectively used as an inhibitor of ROS1 activity And is useful as a composition for preventing or treating cancer such as non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma tumor, rhabdomyosarcoma sarcoma, myofiber blastoma, breast cancer, gastric cancer, lung cancer and melanoma which can be induced therefrom .

< Experimental Example  4> Non-small cell lung cancer cell  Evaluation of cancer cell proliferation inhibition against H2228, H3122

The following experiment was conducted to evaluate the inhibitory effect of the compound represented by the formula (1) according to the present invention on the cancer cell proliferation of non-small cell lung cancer cells H2228 and H3122.

&Lt; 4-1 >

Cell culture medium, RPMI 1640 medium, fetal bovine serum (FBS) and creosin were purchased from Gibco (Grand Island, NY). Sodium bicarbonate, amphotericin B and gentamycin were purchased from Sigma Chemical.

In addition, reagents such as SRB (sulforhodamine) B, trisma base, and trichloroacetic acid (TCA), which were used in the cytotoxicity measurement experiment, were purchased from Sigma Chemical Company. For MTS analysis, the CellTiter 96 R Non-Radioactive Cell Proliferation Assay product was purchased from Promega.

In addition, T-25 culture containers, 96-well plates used for cell culture, and disposable chrysanthemums used for other cell cultures were manufactured by Lincoln Park, NJ.

<4-2> Used equipment

An E-max or SpectraMax 250 instrument from Molecular Devices (Sunnyvale, Calif.) Was used as a microplate reader for cytotoxicity measurements.

<4-3> Experimental method

Step 1: Cell culture

The final dimethyl sulfoxide concentration was adjusted to be 0.5% or less.

The cancer cell lines used in the experiment were all human cancer cell lines, specifically H2228 and H3122, which are non - small cell lung cancer cell lines.

Cultures were incubated in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS) in a 37 and 5% CO2 incubator, and maintained in a passage every 3 to 4 days.

Step 2: Evaluation of proliferation inhibitory activity by compound treatment

1 × 10 4 cells were dispensed into each well of a 96-well flat-bottom microplate and cultured for 24 hours to allow the cells to adhere to the bottom surface, and then the culture medium was removed. The culture medium in which each of the compounds of Examples 1 to 36 was diluted was added thereto and cultured for 72 hours. After completion of the culture with the compound, cytotoxicity was measured using SRB, a protein staining reagent, or MTS assay. After completion of the cultivation with the compounds of Examples 1 to 36, the culture medium was removed, the cold TCA solution was treated in each well, and the cells were allowed to stand at 4 캜 for 1 hour. After removing the TCA solution and drying at room temperature, a staining solution in which 0.4% SRB was dissolved in 1% acetic acid solution was added and left at room temperature for 30 minutes to stain the cells. The excess SRB not bound to the cells was washed away with 1% acetic acid solution, and 10 mM Tris buffer (unbuffered) of pH 10.3 to 10.5 was added to the stained cells to elute the SRB. The absorbance of each well was measured in a wavelength range of 520 mM using a microplate reader.

From the OD value of the well (C) in which the drug is not added, the well (T) in which the drug is added, and the well (Tz)

(T-Tz) / (C-Tz) 100 when Tz = T; or

When Tz> T, the cytotoxicity of the drug was calculated by the formula [(T-Tz) / (Tz)] 100.

The cancer cell proliferation inhibition assay using the MTS assay was performed as follows. Specifically, after the culture with the compounds prepared in Examples 1 to 36 was completed, the PMS solution and the MTS solution constituting the CellTiter 96 R AQueous Non-Radioactive Cell Proliferation Assay product of Promega Co. were mixed with the MTS solution, Was added to each well. Placed in an incubator for 4 hours, removed, and left at room temperature for 10 minutes. The absorbance at 490 nM was measured using SpectraMax250 from Molecular Device, and the growth inhibition effect, GI 50 (growth inhibition 50%) was calculated. The results are shown in Table 5 below.

Example H2228
GI 50 ([mu] M)
H3122
GI 50 ([mu] M)
One 0.007 0.017 2 0.03 0.038 3 0.018 0.03 4 0.31 0.22 5 0.43 0.31 6 0.1 0.1 7 0.2 0.28 8 0.03 0.024 9 0.04 0.037 10 0.08 0.06 11 0.08 0.038 12 0.05 0.036 13 0.05 0.0074 14 0.01 0.0066 15 0.09 0.055 16 0.05 0.024 17 0.08 0.034 18 0.08 0.061 19 0.05 0.036 20 0.12 0.078 21 0.12 0.076 22 0.2 0.11 23 0.09 0.055 24 0.08 0.077 25 0.5 0.28 26 0.5 0.63 27 0.4 0.18 28 0.08 0.08 29 0.2 0.111 30 0.05 0.02 31 0.08 0.039 32 0.5 0.26 33 0.05 0.038 34 0.09 0.16 35 0.039 0.067 36 0.01 0.03 Comparative Example 1
(crizotinib)
0.085 0.28
Comparative Example 2
(LDK-378)
0.025 0.023

As shown in Table 5, the compound of Example according to the present invention exhibits a significantly lower GI 50 than that of Comparative Example 2 (LDK-378), and it is confirmed that the proliferative activity of H2228 and H3122, which are non-small cell lung cancer cell lines, is effectively reduced.

Therefore, the compound represented by formula (I) according to the present invention is excellent in inhibiting the proliferative activity of H2228 and H3122. Therefore, the compound represented by formula (I) according to the present invention is excellent in the inhibitory effect on proliferation of H2228 and H3122, and thus can be used for the prophylaxis and treatment of non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, endometrioid sarcoma, , Melanoma and the like can be effectively used as a composition for preventing or treating cancer.

< Experimental Example  5> H3122 Cancer cell proliferation inhibition against lung cancer  evaluation

The following experiment was conducted to evaluate the inhibition of cancer cell proliferation of H3122 phosphorylated lung cancer of the compound represented by Formula 1 according to the present invention.

<5-1> Preparing the experiment

Experimental nude mice (BALB / c nu / nu, female) were purchased from Charles River Japan, Inc. and fed and tested under SPF (Specific Pathogen Free) control. The H3122 cell line, a human non-small cell lung cancer cell line, was used by the Korean Chemical Research Institute.

<5-2> Experimental method

After the purchase, the female was transplanted into female nude mice adapted to the laboratory. H3122 grown to an appropriate size for transferring was placed in a 3 × 3 × 3 mm 3 (Sc) subcutaneously transplanted into the right lateral side of the nude mouse, and when the size of the implanted canine reached about 200 mm 3 , administration of the compounds of Examples 1, 13, and 14 as a drug was started, (1 day). In the control group, 20% PEG400 + 3% Tween 80 in DDW was orally administered and the drug test group (6 mice / group) dissolved in the same solvent as the control group was administered orally 14 times (qd × 14) orally. The size of the cancer is measured by caliper (long diameter (a), short diameter (b)) every 2-3 days after administration and the size of the cancer is calculated by the following equation Volume, V) were measured.

[Equation 1]

Volume (mm 3) = a × b 2/2

In Equation (1), a represents a large transverse length of the arm, and b represents a transverse short length of the arm. The results of the above experiment are shown in Fig. 1 and Fig.

FIG. 3 is a graph showing the results of observing the size of cancer in the drug-treated control group, the LDK378 treated group, and the compound treated group of Example 1 with time.

FIG. 4 is a graph showing the results of observing the size of cancer in the drug-treated control group (control) LDK378 treated group, the treated group of Example 13 and the compound treated group of Example 14 over time.

3, it can be seen that the size of the cancer was significantly increased with the lapse of time in the control group, and it was confirmed that the group treated with the compound of Example 1 according to the present invention suppressed the size of the cancer to a greater extent than the group treated with LDK378 .

4, it can be seen that the size of the cancer significantly increases with time in the control group, and that the compound treated group according to Example 13 or Example 14 according to the present invention is superior to the group treated with LDK378 .

Therefore, the compound represented by formula (I) according to the present invention is excellent in inhibiting cancer cell proliferation against H3122 lung cancer. Therefore, the compound represented by formula (I) according to the present invention is useful as a prophylactic treatment against non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, endometrioid sarcoma, , Lung cancer, melanoma, and the like.

Meanwhile, the compound represented by Formula 1 according to the present invention can be formulated into various forms according to the purpose. Hereinafter, some formulation methods in which the compound represented by Formula 1 according to the present invention is contained as an active ingredient are exemplified, and the present invention is not limited thereto.

< Formulation example  1> Preparation of pharmaceutical preparations

1-1. Sanje  Produce

500 mg of the derivative of formula (1);

Lactose 100 mg; And

Talc 10 mg.

The above components are mixed and filled in airtight bags to prepare powders.

1-2. Manufacture of tablets

500 mg of the derivative of formula (1);

100 mg of corn starch;

Lactose 100 mg; And

Magnesium stearate 2 mg.

After mixing the above components, tablets are prepared by tableting according to the usual preparation method of tablets.

1-3. Manufacture of capsules

500 mg of the derivative of formula (1);

100 mg of corn starch;

Lactose 100 mg; And

Magnesium stearate 2 mg.

The above components are mixed according to a conventional capsule preparation method and filled in gelatin capsules to prepare capsules.

1-4. Injection preparation

500 mg of the derivative of formula (1);

Sterile distilled water suitable for injection; And

pH adjuster.

(2 ml) per ampoule in accordance with the usual injection method.

1-5. Liquid  Produce

100 mg of the derivative of formula (1);

10 g per isomerization;

5 g mannitol; And

Purified water.

Each component was added to purified water in accordance with the usual liquid preparation method and dissolved, and the lemon flavor was added in an appropriate amount. Then, the above components were mixed, and purified water was added thereto. The whole was adjusted to 100 ml with purified water, The liquid is prepared by sterilization.

Claims (10)

Claims 1. A compound represented by the following formula (1), an optical isomer thereof, or a pharmaceutically acceptable salt thereof:
[Chemical Formula 1]
Figure 112018120300010-pat00107

(In the formula 1,
X a is independently hydrogen or chloro;
X b is fluoro or chloro;

R &lt; 1 &gt; is hydrogen, straight or branched C1-3 alkyl or C1-3 alkylene linked to Y &lt; 2 &gt;;
R 2 is unsubstituted or one or more C1-3 alkyl, and halogen-substituted straight-chain or branched; And

Figure 112018120300010-pat00129
Quot; is a single bond or a double bond,

Between Y 1 and Y 2
Figure 112018120300010-pat00130
Is a single bond, and the bond between Y 2 and Y 3
Figure 112018120300010-pat00131
Y 1 is nitrogen (N), Y 2 is CR 6 (wherein R 6 is hydrogen or a bond connected to R 1 ), Y 3 is nitrogen (N), Y 4 is oxygen O), NR 3 or N + R 4 R 5 , and Y 5 is CH, or

Between Y 1 and Y 2
Figure 112018120300010-pat00132
Is a double bond, and the bond between Y 2 and Y 3
Figure 112018120300010-pat00133
If this is a single bond, Y 1 is a carbon (C), Y 2 is nitrogen (N), Y 3 is oxygen (O), Y 4 are CH, and Y 5 is NH, and

Wherein R 3 is hydrogen, C1-3 alkyl carbonyl unsubstituted or at least one hydroxy group is substituted by straight-chain or C1-3 alkyl, unsubstituted or at least one of the side-chain hydroxy group is substituted straight or branched chain carbonyl, or dimethylaminomethyl Carbonyl, and R 4 and R 5 are independently straight or branched C 1-3 alkyl.
delete The method according to claim 1,
R 1 is hydrogen, methyl or C 2 alkylene linked to Y 2 ; And
R 2 is methyl, ethyl, isopropyl or -CHF 2 , an optical isomer thereof, or a pharmaceutically acceptable salt thereof.
The method according to claim 1,
The compound represented by the formula (1) is any one selected from the group consisting of the following compounds, an optical isomer thereof, or a pharmaceutically acceptable salt thereof:
(1) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(2) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropoxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;
(3) Synthesis of 5-chloro-N4- (5-chloro-2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(4) Synthesis of 5-chloro-N4- (4-chloro-2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(5) Synthesis of 5-chloro-N4- (3-chloro-2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(6) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- 4- (5,6-dihydro- 1H) -yl) phenyl) pyrimidine-2,4-diamine;
(7) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropoxy-4- (5,6-dihydro- (1H) -yl) phenyl) pyrimidine-2,4-diamine;
(8) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(9) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(10) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;
(11) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (9-methoxy-2,3,5,6-tetrahydro- Azino [4,3-a] quinolin-8-yl) pyrimidine-2,4-diamine;
(12) 5-Chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (9-methoxy-3-methyl-2,3,5,6-tetrahydro- , 4] triazino [4,3-a] quinolin-8-yl) pyrimidine-2,4-diamine;
(13) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(14) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;
(15) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazine-l (4H) -yl) -2- (dimethylamino) ethan-1-one;
(16) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazin-l (4H) -yl) -2-hydroxyethan-l-one;
(17) Synthesis of 2- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazin-1 (4H) -yl) ethan-l-ol;
(18) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine hydrochloride;
(19) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-ethoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(20) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-difluoromethoxy- , 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;
(21) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-ethoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(22) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-difluoromethoxy- -Triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(23) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- Azin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(24) 5- fluoro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;
(25) 5-Fluoro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropoxy- -Triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(26) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- (2,4-triazine-4 (1H) -yl) phenyl) pyrimidine-2,4-diamine iodide;
(27) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 4 (1H) -yl) phenyl) pyrimidine-2,4-diamine;
(28) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;
(29) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- ) -5,6-dihydro-l, 2,4-triazine-l (4H) -yl-2-hydroxyethan-l-one;
(30) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazine-l (4H) -yl-2-hydroxyethan-l-one;
(31) Synthesis of 1- (4- (4 - ((5-chloro-4 - ((2- (isopropylsulfonyl) phenylamino) pyrimidin- Dihydro-l, 2,4-triazine-l (4H) -yl-2-dimethylaminoethan-l-one;
(32) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-isopropyloxy- 2,4-triazin-4 (1 H) -yl) phenyl) pyrimidine-2,4-diamine;
(33) 5-Chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (2-methoxy- , 4-triazin-4 (lH) -yl) phenyl) pyrimidine-2,4-diamine;
(34) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (4- (5,6-dihydro- -2-methoxyphenyl) pyrimidine-2,4-diamine;
(35) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (4- (5,6-dihydro- -2-isopropoxyphenyl) pyrimidine-2,4-diamine; And
(36) Synthesis of 5-chloro-N4- (2- (isopropylsulfonyl) phenyl) -N2- (4- (5,6-dihydro- -2-methoxyphenyl) pyrimidine-2,4-diamine.
As shown in Scheme 1 below,
A process for preparing a compound represented by the general formula (1) as set forth in claim 1, comprising the step of reacting a compound represented by the general formula (2) with a compound represented by the general formula (3) to prepare a compound represented by the general formula (1)
[Reaction Scheme 1]
Figure 112016072414212-pat00123

(In the above Reaction Scheme 1,
X a , X b , R 1 , R 2 , Y 1 , Y 2 , Y 3 , Y 4 , Y 5 ,
Figure 112016072414212-pat00124
Are as defined in formula (1) of claim 1).
The present invention relates to a pharmaceutical composition for the treatment of non-small cell lung cancer, neuroblastoma, inflammatory myelofibroblastoma, endometrial sarcoma, myoblastoma, breast cancer, A pharmaceutical composition for the prophylaxis or treatment of gastric cancer, lung cancer, melanoma, large B-cell lymphoma, systemic atrophy, inflammatory myofibroblastic sarcoma, or esophageal squamous cell cancer.
The method according to claim 6,
Wherein said pharmaceutical composition inhibits the activity of ALK (Anaplastic Lymphoma Kinase) to inhibit the expression and growth of cancer cells.
The method according to claim 6,
Wherein said pharmaceutical composition inhibits the activity of ROS kinase (pro-oncogene tyrosine-protein kinase) to prevent or treat non-small cell lung cancer.
A pharmaceutical composition for preventing or treating CNS cancer, which comprises the compound represented by the general formula (1) of claim 1, an optical isomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient.
10. The method of claim 9,
Wherein the CNS cancer is brain cancer, meningiomas, astrocytomas, or glioblastoma multiforme.
KR1020160094560A 2016-07-26 2016-07-26 Novel pyrimidine-2,4-diamine derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient KR101937529B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
KR1020160094560A KR101937529B1 (en) 2016-07-26 2016-07-26 Novel pyrimidine-2,4-diamine derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient
PCT/KR2017/008052 WO2018021826A1 (en) 2016-07-26 2017-07-26 Novel pyrimidine-2,4-diamine derivative and pharmaceutical composition for prevention or treatment of cancer containing same as active ingredient

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020160094560A KR101937529B1 (en) 2016-07-26 2016-07-26 Novel pyrimidine-2,4-diamine derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient

Publications (2)

Publication Number Publication Date
KR20180012352A KR20180012352A (en) 2018-02-06
KR101937529B1 true KR101937529B1 (en) 2019-01-14

Family

ID=61016427

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020160094560A KR101937529B1 (en) 2016-07-26 2016-07-26 Novel pyrimidine-2,4-diamine derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient

Country Status (2)

Country Link
KR (1) KR101937529B1 (en)
WO (1) WO2018021826A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111170996B (en) * 2018-11-09 2021-12-28 天津大学 Pyrimidine derivative with ALK inhibitory activity and synthetic method and application thereof
CN109593065A (en) * 2018-12-03 2019-04-09 重庆医科大学 A kind of bis- target spot inhibitor of HDAC/ALK and the preparation method and application thereof
KR20230076504A (en) 2021-11-24 2023-05-31 성균관대학교산학협력단 Apparatus, method, computer-readable storage medium and computer program for analyzing performance of next-generation memory and storage based on full system simulator

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008073687A2 (en) * 2006-12-08 2008-06-19 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2016130920A2 (en) 2015-02-13 2016-08-18 Dana-Farber Cancer Institute, Inc. Lrrk2 inhibitors and methods of making and using the same

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0305929D0 (en) * 2003-03-14 2003-04-23 Novartis Ag Organic compounds
WO2009032694A1 (en) * 2007-08-28 2009-03-12 Dana Farber Cancer Institute Amino substituted pyrimidine, pyrollopyridine and pyrazolopyrimidine derivatives useful as kinase inhibitors and in treating proliferative disorders and diseases associated with angiogenesis
US8592432B2 (en) * 2008-04-07 2013-11-26 Bei Chen Compounds and compositions as protein kinase inhibitors
CA2952787A1 (en) * 2014-06-17 2015-12-23 Korea Research Institute Of Chemical Technology Pyrimidine-2,4-diamine derivative and anticancer pharmaceutical composition comprising same as effective ingredient

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008073687A2 (en) * 2006-12-08 2008-06-19 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2016130920A2 (en) 2015-02-13 2016-08-18 Dana-Farber Cancer Institute, Inc. Lrrk2 inhibitors and methods of making and using the same

Also Published As

Publication number Publication date
KR20180012352A (en) 2018-02-06
WO2018021826A1 (en) 2018-02-01

Similar Documents

Publication Publication Date Title
KR101656382B1 (en) pyrimidine-2,4-diamine derivatives and pharmaceutical composition for anti cancer containing the same as an active ingredient
US11958855B2 (en) Aurora kinase inhibitors for inhibiting mitotic progression
EP2498607B1 (en) Kinase inhibitors
KR101446742B1 (en) N2,N4-bis(4-(piperazin-1-yl)phenyl)pyrimidine-2,4-diamine derivatives or pharmaceutically acceptable salt thereof, and pharmaceutical composition for the prevention or treatment of cancer containing the same as an active ingredient
KR101323566B1 (en) Di(arylamino)aryl compound
US7541367B2 (en) 3-benzoimidazolyl-pyrazolopyridines useful in treating kinase disorders
EA019966B1 (en) Compounds and compositions as protein kinase inhibitors
US9512121B2 (en) [1,2,4] triazol [4,3-A] pyridine derivative, preparation method therefor or medical application thereof
US20130303518A1 (en) Methods of Inhibiting the Catalytic Activity of a Protein Kinase and of Treating a Protein Kinase Related Disorder
KR20090129434A (en) Pyrimidine-2,4-diamine derivatives and their use as jak2 kinase inhibitors
KR20200115583A (en) 2H-indazole derivatives as CDK4 and CDK6 inhibitors and their therapeutic use
CA3124678A1 (en) Aza-heterobicyclic inhibitors of mat2a and methods of use for treating cancer
CA2987466A1 (en) Use of pteridinone derivative serving as egfr inhibitor
KR101854512B1 (en) Novel chemical compounds (variants) and the use thereof to treat oncological diseases
KR101937529B1 (en) Novel pyrimidine-2,4-diamine derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient
US9321762B2 (en) Quinazoline compounds, method for preparing the same and use thereof
US10745360B2 (en) Quinoxaline compounds, method for preparing the same and use thereof
CN114763360A (en) Chiral macrocyclic compounds as protein kinase inhibitors and uses thereof
JP2024503273A (en) Heteroaryl derivative compounds and their uses
KR20200134179A (en) Heteroaryl comprising N derivatives and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient
JP2021500350A (en) Inhibitor of mutant EGFR family tyrosine kinase
KR101633722B1 (en) 4-(1-pyrrole-3,4-dicarboxamide)pyrimidine derivatives, preparation method thereof, and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient
CN116323566A (en) Sulfanone compound and preparation method and medical application thereof

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
E701 Decision to grant or registration of patent right