KR20210118297A - p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same - Google Patents

p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same Download PDF

Info

Publication number
KR20210118297A
KR20210118297A KR1020200034179A KR20200034179A KR20210118297A KR 20210118297 A KR20210118297 A KR 20210118297A KR 1020200034179 A KR1020200034179 A KR 1020200034179A KR 20200034179 A KR20200034179 A KR 20200034179A KR 20210118297 A KR20210118297 A KR 20210118297A
Authority
KR
South Korea
Prior art keywords
compound
disease
ethoxy
integer
organic solvent
Prior art date
Application number
KR1020200034179A
Other languages
Korean (ko)
Other versions
KR102624481B1 (en
Inventor
김남중
인경수
이종길
손승환
이나래
송채원
김동환
도지민
Original Assignee
(주)프레이저테라퓨틱스
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 (주)프레이저테라퓨틱스 filed Critical (주)프레이저테라퓨틱스
Priority to KR1020200034179A priority Critical patent/KR102624481B1/en
Priority to PCT/KR2021/002361 priority patent/WO2021187766A1/en
Publication of KR20210118297A publication Critical patent/KR20210118297A/en
Application granted granted Critical
Publication of KR102624481B1 publication Critical patent/KR102624481B1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • 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
    • A23V2200/322Foods, ingredients or supplements having a functional effect on health having an effect on the health of the nervous system or on mental function
    • 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
    • A23V2200/324Foods, ingredients or supplements having a functional effect on health having an effect on the immune system
    • 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
    • A23V2250/00Food ingredients
    • A23V2250/30Other Organic compounds

Abstract

The present invention relates to a novel compound having p38 removal capability, a manufacturing method thereof, and a composition for treating chronic inflammatory disease, comprising the same. The compound represented by chemical formula A, according to the present invention, has an effect of having notably excellent p38 MAPK removal capability, and thus can be useful as a therapeutic agent for chronic inflammatory disease.

Description

p38 제거능을 갖는 화합물, 이의 제조방법 및 이를 포함하는 만성 염증성 질환 치료용 조성물 {p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same}A compound having p38 removal ability, a method for preparing the same, and a composition for treating chronic inflammatory diseases comprising the same {p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same}

본 발명은 p38 제거능을 갖는 신규한 화합물, 이의 제조방법 및 이를 포함하는 만성 염증성 질환 치료용 조성물에 관한 것이다.The present invention relates to a novel compound having p38 scavenging ability, a method for preparing the same, and a composition for treating chronic inflammatory diseases comprising the same.

염증은 여러 가지 원인에 의해서 발생하는 것으로 알려져 있으나, 최근 여러 연구에 의하면 다양한 스트레스 자극에 의해 MAPKs (mitogen-activated protein kinases)가 활성화될 수 있고, 특히 p38 MAPK는 TNF-α, IL-1β, IL-6 등과 같은 염증전구물질(pro-inflammatory 사이토카인s)의 생성에 결정적인 역할을 하여, 염증성장질환(inflammatory bowel disease), 크론병(Crohn's disease), 류마티스 관절염(rheumatoid arthritis), 다발성경화증(multiple sclerosis), 건선(psoriasis), 신경병증 통증(neuropathic pain), 알츠하이머 병(Alzheimer's disease) 등을 유발하는 것으로 알려져 있다(NATURE REVIEWS, Drug discovery (2003), Vol.2, 717-726).Inflammation is known to be caused by various causes, but according to recent studies, mitogen-activated protein kinases (MAPKs) can be activated by various stress stimuli. It plays a crucial role in the production of pro-inflammatory cytokines such as -6, inflammatory bowel disease, Crohn's disease, rheumatoid arthritis, and multiple sclerosis. sclerosis), psoriasis, neuropathic pain, Alzheimer's disease, etc. are known to cause ( NATURE REVIEWS, Drug discovery (2003), Vol.2, 717-726).

만성 염증은 지속적인 염증이 일어나는 상태로, 면역체계가 특정한 노출에 대하여 지속적으로 부적절한 반응을 나타내는 것을 의미하는데, 특히 자가면역 질환은 면역 체계의 혼란으로 인해 장기를 공격하면서 발생한다. 만성 염증은 단일 또는 복합적인 유전 요소 또는 음식이나 매연같은 환경적 요인으로 발생하기도 한다. 염증이 발생했다 진정되기도 하나, 지속적으로 이어지면서 때로는 치료가 큰 효과가 없게 되기도 한다. 만성 염증성 질환은 체내 조직에 상당한 손상을 주게 되고 위치에 따라서는 여러 문제가 발생한다. 예를 들어, 간이나 소화기관에 만성염증이 올 경우 뇌에 변화를 일으켜 피로함이나 성격변화, 장기의 기능장애를 일으키며 전신으로 염증을 확산시키기도 한다.Chronic inflammation is a condition in which persistent inflammation occurs, which means that the immune system continuously responds inappropriately to a specific exposure. Chronic inflammation may be caused by a single or multiple genetic factors or environmental factors such as food or fumes. Sometimes inflammation occurs and then subsides, but as it continues, sometimes the treatment becomes ineffective. Chronic inflammatory disease causes significant damage to body tissues, and various problems occur depending on the location. For example, when chronic inflammation occurs in the liver or digestive system, it causes changes in the brain, causing fatigue, personality changes, organ dysfunction, and spreading inflammation throughout the body.

만성 염증성 질환 중 대표적인 건선(psoriasis)은 피부각질형성세포(keratinocyte)가 빠르게 증식하고 피부염증을 동반함을 특징으로 하는 원인 미상의 만성 재발성 피부질환으로서, 홍반, 인설, 구진 또는 농포 등의 병변이 피부면 어디에서든 나타날 수 있다. 임상적으로 판상 건선, 물방울양 건선, 농포성 건선, 역위 건선, 홍피성 건선 등으로 분류할 수 있다. 건선은 유전적인 원인과 환경적인 요인이 복합적으로 작용하여 발생하는데, 그 원인 이 아직 명확히 밝혀지지 않았으나, 호르몬의 변화, 스트레스와 같은 심리적 측면, 외상, 편도선염과 같은 감염, 면역, 내분비, 염증, 대사능력, 기후, 건조한 피부, 약물, 유전 등으로 그 발병소인이 다양하다. 또한, 만성 염증성 질환으로 면역계의 과민화 또는 환자의 자가면역 이상에 의해 분화된 T 면역세포가 분비하는 IL23, IL-17, IL-6. TNF-α 등의 사이토카인들이 과도하게 발현되면 피부의 염증반응을 일으키고, 각질형성세포의 과다증식 및 분열을 촉진시키고 분화는 억제시킨다. 이러한 요인들로 인하여 피부 각질층의 세포가 정상세포 보다 6~7배 빠르게 증식하기 때문에 발생하며, 이렇게 과다하면서도 불완전하게 증식된 각질 세포가 하얀 인설의 비늘로 겹겹이 쌓여 떨어져 나가게 된다. 건선은 다른 질환을 동반하는 경우가 많아 이를 동반 질환이라 지칭하며 건선성 관절염, 심혈관 질환, 우울증, 크론병 등이 건선 환자에게서 빈번하게 발견되는 질환이다. 건선성 관절염은 통증을 동반한 만성 염증질환으로 건선 환자 중 최대 30%가 관절염 증상을 보이며, 그 중 건선이 관절염에 선행하는 경우는 85%이며 최근의 연구에서는 관절염과 건선의 동시 발병 사례도 보고되어 아직 선후 관계의 원인은 뚜렷하지 않다. 또한 중증의 건선은 비만, 고혈압, 고지혈증, 그리고 제 2형 당뇨병 등의 대사증후군과 사망률 증가와도 연관이 있고, 심혈관 질환 발생 위험도 증가하는데, 이는 만성 염증 반응이 내분비계에 미치는 영향 때문일 가능성이 있다. 그 밖에도 건선피부는 미관상 좋지 않아 손이나 얼굴 등 눈에 띄는 부위에 발생할 경우 대인관계에도 위축을 가져와 우울증이나 불안증을 앓는 이들의 비율이 일반인보다 40% 가량 높다는 보고가 있다.Psoriasis, a representative chronic inflammatory disease, is a chronic recurrent skin disease of unknown cause, characterized by rapid proliferation of keratinocytes and accompanying skin inflammation. Lesions such as erythema, scale, papule or pustule It can appear anywhere on this skin surface. Clinically, it can be classified into plaque psoriasis, drip psoriasis, pustular psoriasis, inverse psoriasis, and erythrodermic psoriasis. Psoriasis is caused by a combination of genetic and environmental factors. The cause is still unknown, but hormonal changes, psychological aspects such as stress, trauma, infections such as tonsillitis, immunity, endocrine, inflammation, metabolism There are various causes of the disease, such as ability, climate, dry skin, drugs, and heredity. In addition, IL23, IL-17, IL-6 secreted by T immune cells differentiated by autoimmune abnormalities or hypersensitivity of the immune system due to chronic inflammatory diseases. When cytokines such as TNF-α are excessively expressed, it causes an inflammatory response in the skin, promotes hyperproliferation and division of keratinocytes, and inhibits differentiation. It occurs because the cells of the stratum corneum of the skin proliferate 6 to 7 times faster than normal cells due to these factors, and these excessive and incompletely proliferated keratinocytes are piled up in layers of white scales and fall off. Psoriasis is often referred to as a comorbidity because it is accompanied by other diseases. Psoriatic arthritis, cardiovascular disease, depression, and Crohn's disease are frequently found in psoriasis patients. Psoriatic arthritis is a chronic inflammatory disease accompanied by pain, and up to 30% of psoriasis patients show arthritis symptoms, and among them, psoriasis precedes arthritis in 85% of cases. The cause of the antecedent relationship is not yet clear. Severe psoriasis is also associated with increased mortality and metabolic syndrome such as obesity, hypertension, hyperlipidemia, and type 2 diabetes, and an increased risk of cardiovascular disease, possibly due to the effects of chronic inflammatory responses on the endocrine system. . In addition, there is a report that psoriasis skin is not aesthetically pleasing, and when it occurs on conspicuous areas such as hands or face, it causes atrophy in interpersonal relationships, and the rate of people suffering from depression or anxiety is 40% higher than that of the general public.

또한, 만성 염증은 각종 다양한 질환의 원인으로 주목받고 있다. 실제로 동맥경화증을 앓고 있는 환자 중 30% 이상은 정상적인 콜레스테롤 수치를 유지하고 있으나, 과도한 염증반응의 활성화로 인해 백혈구를 비롯한 각종 염증세포들이 건강하지 않은 혈관 내 세포벽에 붙어 이상 증식함으로서 혈전 및 동맥경화를 야기하는 것으로 알려져 있다. 또한 만성염증으로 인해 면역세포들이 뇌 세포 손상으로 알츠하이머가 발생할 수 있으며, 심근 세포의 손상으로 울혈성 심부전이 발생하는 등 광범위한 질환의 유병에 만성 염증이 관여한다.In addition, chronic inflammation is attracting attention as a cause of various diseases. In fact, more than 30% of patients suffering from arteriosclerosis maintain normal cholesterol levels, but due to excessive activation of the inflammatory reaction, various inflammatory cells, including white blood cells, attach to unhealthy intravascular cell walls and proliferate abnormally, thereby preventing thrombosis and arteriosclerosis. known to cause In addition, chronic inflammation is involved in the prevalence of a wide range of diseases, such as Alzheimer's disease due to brain cell damage to immune cells due to chronic inflammation, and congestive heart failure due to cardiomyocyte damage.

만성 염증(chronic inflammation)의 치료를 위해 TNF-α 또는 IL-1β를 직접적으로 무효화(neutralize)시키는 모노클론항체(monoclonal antibodies)가 크게 성공하면서, 지난 수십년간 p38 MAPK의 억제제로서 몇몇 저분자 의약품(small molecule)이 개발되어왔다.With the great success of monoclonal antibodies that directly neutralize TNF-α or IL-1β for the treatment of chronic inflammation, several small molecule drugs (small molecule) as inhibitors of p38 MAPK over the past few decades have been molecules) have been developed.

그러나, 현재까지 전임상 및 임상 실험결과에서 치료효과가 낮고, 간독성 및 신경 부작용과 관련된 안전성 이슈 때문에 치료제로 상용화되지 못한 상태이다. 상기 안전성 이슈는 부분적으로는 키나아제 선택성이 부족하기 때문이고, 이는 부정확한 치료적 영향을 야기한다.However, to date, the therapeutic effect is low in preclinical and clinical test results, and it has not been commercialized as a therapeutic agent due to safety issues related to hepatotoxicity and neurological side effects. This safety issue is due in part to a lack of kinase selectivity, which leads to inaccurate therapeutic effects.

따라서, P38 MAPK에 선택적인 저해능을 나타내고, 안전성 및 효능이 우수한 만성 염증 치료제의 개발이 필요한 실정이다.Therefore, there is a need for the development of a therapeutic agent for chronic inflammation that selectively inhibits P38 MAPK and has excellent safety and efficacy.

이에, 본 발명자는 P38 MAPK에 선택적인 저해능을 나타내고, 안전성 및 효능이 우수한 화합물을 연구하던 중, 화학식 A로 표시되는 화합물의 효능이 매우 우수함을 확인하고 본 발명을 완성하였다.Accordingly, the present inventors have completed the present invention by confirming that the efficacy of the compound represented by Formula A is very excellent while researching a compound that exhibits selective inhibition of P38 MAPK and has excellent safety and efficacy.

NATURE REVIEWS, Drug discovery (2003), Vol.2, 717-726NATURE REVIEWS, Drug discovery (2003), Vol.2, 717-726

본 발명의 목적은 신규한 화학식 A로 표시되는 화합물을 제공하는 것이다.An object of the present invention is to provide a novel compound represented by the formula (A).

본 발명의 다른 목적은 상기 화학식 A로 표시되는 화합물의 제조방법을 제공하는 것이다.Another object of the present invention is to provide a method for preparing a compound represented by the above formula (A).

본 발명의 또 다른 목적은 상기 화학식 A로 표시되는 화합물을 포함하는 만성 염증성 질환 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for preventing or treating chronic inflammatory diseases comprising the compound represented by Formula A.

본 발명의 다른 목적은 상기 화학식 A로 표시되는 화합물을 포함하는 만성 염증성 질환 예방 또는 개선용 건강기능식품 조성물을 제공하는 것이다.Another object of the present invention is to provide a health functional food composition for preventing or improving chronic inflammatory disease comprising the compound represented by Formula A.

본 발명의 또 다른 목적은 상기 화학식 A로 표시되는 화합물을 포함하는 만성 염증성 질환 예방 또는 개선용 건강식품 조성물을 제공하는 것이다.Another object of the present invention is to provide a health food composition for preventing or improving chronic inflammatory diseases comprising the compound represented by Formula A.

상기 목적을 달성하기 위하여,In order to achieve the above object,

본 발명은 하기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 제공한다.The present invention provides a compound represented by the following formula (A) or a pharmaceutically acceptable salt thereof.

[화학식 A][Formula A]

Figure pat00001
Figure pat00001

(상기 화학식 A에 있어서,(In the formula A,

상기 p38 Ligand Moiety는

Figure pat00002
이고;The p38 Ligand Moiety is
Figure pat00002
ego;

상기 E3 Ligand Moiety는

Figure pat00003
이고;The E3 Ligand Moiety is
Figure pat00003
ego;

상기 Linker는

Figure pat00004
,
Figure pat00005
,
Figure pat00006
,
Figure pat00007
,
Figure pat00008
또는
Figure pat00009
이고,The Linker is
Figure pat00004
,
Figure pat00005
,
Figure pat00006
,
Figure pat00007
,
Figure pat00008
or
Figure pat00009
ego,

상기 n 및 m은 독립적으로 1 내지 10의 정수이고,Wherein n and m are independently integers from 1 to 10,

상기 a 및 b는 독립적으로 1 내지 10의 정수이다). wherein a and b are independently integers from 1 to 10).

또한, 본 발명은 하기 반응식 1에 나타난 바와 같이,In addition, the present invention, as shown in Scheme 1 below,

화합물 8A와 화합물 13을 유기용매에서 반응시켜 화합물 14A를 합성하는 단계(단계 1);synthesizing compound 14A by reacting compound 8A with compound 13 in an organic solvent (step 1);

화합물 14A와 4-니트로페닐클로로포르메이트를 유기용매에서 반응시켜 화합물 19A를 합성하는 단계(단계 2); 및synthesizing compound 19A by reacting compound 14A with 4-nitrophenylchloroformate in an organic solvent (step 2); and

화합물 19A와 화합물 5를 유기용매에서 반응시켜 화합물 A1을 합성하는 단계(단계 3);를 포함하는 화합물 A1의 제조방법을 제공한다.It provides a method for preparing compound A1, comprising the step of reacting compound 19A and compound 5 in an organic solvent to synthesize compound A1 (step 3).

[반응식 1][Scheme 1]

Figure pat00010
Figure pat00010

(상기 반응식 1에서,(In Scheme 1,

L1은 -(CH2)n-OH 또는 -(CH2CH2-O)n-CH2CH2OH이고, 여기서 상기 n은 1 내지 10의 정수이고;L 1 is —(CH 2 ) n —OH or —(CH 2 CH 2 —O) n —CH 2 CH 2 OH, wherein n is an integer from 1 to 10;

L2는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).L 2 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).

나아가, 본 발명은 하기 반응식 2에 나타난 바와 같이,Furthermore, the present invention, as shown in Scheme 2 below,

화합물 8A와 화합물 13을 유기용매에서 반응시켜 화합물 14A를 합성하는 단계(단계 1);synthesizing compound 14A by reacting compound 8A with compound 13 in an organic solvent (step 1);

화합물 14A와 p-TsCl(p-Toluenesulfonyl chloride)를 유기용매에서 반응시켜 화합물 27A를 합성하는 단계(단계 2); 및synthesizing compound 27A by reacting compound 14A with p-TsCl (p-Toluenesulfonyl chloride) in an organic solvent (step 2); and

화합물 27A와 화합물 6을 유기용매에서 반응시켜 화합물 A2를 합성하는 단계(단계 3);를 포함하는 화합물 A2의 제조방법을 제공한다.It provides a method for preparing compound A2, comprising: synthesizing compound A2 by reacting compound 27A with compound 6 in an organic solvent (step 3).

[반응식 2][Scheme 2]

Figure pat00011
Figure pat00011

(상기 반응식 2에서,(In Scheme 2,

L1은 -(CH2)n-OH 또는 -(CH2CH2-O)n-CH2CH2OH이고, 여기서 상기 n은 1 내지 10의 정수이고;L 1 is —(CH 2 ) n —OH or —(CH 2 CH 2 —O) n —CH 2 CH 2 OH, wherein n is an integer from 1 to 10;

L2는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).L 2 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).

또한, 본 발명은 하기 반응식 3에 나타난 바와 같이,In addition, the present invention, as shown in Scheme 3 below,

화합물 14와 화합물 36A를 유기용매에서 반응시켜 화합물 37A를 합성하는 단계(단계 1);synthesizing compound 37A by reacting compound 14 with compound 36A in an organic solvent (step 1);

화합물 37A와 염산을 유기용매에서 반응시켜 화합물 38A를 합성하는 단계(단계 2); 및synthesizing compound 38A by reacting compound 37A with hydrochloric acid in an organic solvent (step 2); and

화합물 38A와 화합물 40을 유기용매에서 반응시켜 화합물 A3을 합성하는 단계(단계 3);를 포함하는 화합물 A3의 제조방법을 제공한다.It provides a method for preparing compound A3, including a step of reacting compound 38A and compound 40 in an organic solvent to synthesize compound A3 (step 3).

[반응식 3][Scheme 3]

Figure pat00012
Figure pat00012

(상기 반응식 3에서,(In Scheme 3,

L3은 -(CH2)n-OMs 또는 -(CH2CH2-O)n-CH2CH2OMs이고, 여기서 상기 n은 1 내지 10의 정수이고;L 3 is -(CH 2 ) n -OMs or -(CH 2 CH 2 -O) n -CH 2 CH 2 OMs, wherein n is an integer from 1 to 10;

L4는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).L 4 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).

나아가, 본 발명은 상기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 치료용 약학적 조성물을 제공한다.Furthermore, the present invention provides a pharmaceutical composition for preventing or treating chronic inflammatory diseases, comprising the compound represented by Formula A or a pharmaceutically acceptable salt thereof.

또한, 본 발명은 상기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 개선용 건강기능식품 조성물을 제공한다.In addition, the present invention provides a health functional food composition for preventing or improving chronic inflammatory diseases comprising the compound represented by Formula A or a pharmaceutically acceptable salt thereof.

나아가, 본 발명은 상기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 개선용 건강식품 조성물을 제공한다.Furthermore, the present invention provides a health food composition for preventing or improving chronic inflammatory diseases comprising the compound represented by Formula A or a pharmaceutically acceptable salt thereof.

본 발명에 따른 화학식 A로 표시되는 화합물은 p38 MAPK 제거능이 현저히 우수한 효과가 있어, 만성 염증성 질환 치료제로 유용할 수 있다.The compound represented by Formula A according to the present invention has a remarkably excellent effect of removing p38 MAPK, and may be useful as a therapeutic agent for chronic inflammatory diseases.

도 1은 본 발명에 따른 실시예 1 내지 9의 화합물과 비교예 1 내지 3의 화합물의 화학구조식을 나타낸 도면이다.
도 2는 본 발명의 실시예에 따른 화합물 7종의 p38 MAPK 제거능을 면역블롯팅 방법으로 확인한 결과이다.
도 3은 본 발명의 실시예 2에 따른 화합물 24의 처리 농도에 따른 p38(Flag) 및 P-p38의 제거능을 면역블롯팅 방법으로 확인한 결과이다.
도 4는 본 발명의 실시예 2(화합물24)에서 [E3 Ligand Moiety(탈리도미드 유도체)] 대신에 비오틴을 결합한 비교예 1(화합물 43)과 실시예 2(화합물24)에서 [p38 Ligand Moiety]를 제외한 Linker에 Biotin을 결합한 비교예 2(화합물 45)를 각각 p38 MAPK 결합능을 평가한 결과이다.
도 5는 본 발명에 따른 실시예 화합물이 세포 내의 Proteasome system을 사용하여 p38 MAPK를 제거시키는 것을 면역블롯팅 방법으로 확인한 결과이다.
도 6은 본 발명에 따른 실시예 화합물이 세포 내의 E3 ligase complex 단백질 중 하나인 cereblon (CRBN)을 사용하여 p38 MAPK를 제거시키는 것을 면역블롯팅 방법으로 확인한 결과이다.
도 7은 본 발명에 따른 실시예 화합물이 유비퀴틴화 시스템을 사용하여 p38 MAPK를 제거시키는 것을 SDS-PAGE 및 면역블롯팅 방법으로 확인한 결과이다.
도 8은 본 발명에 따른 실시예 화합물이 신경세포에서 농도 의존적으로 p38 MAPK 및 P-p38 MAPK 감소를 유도함을 면역블롯팅 방법으로 확인한 결과이다.
도 8(A): 신경아교세포(Astrocyte)
도 8(B): 신경모세포(Neuroblast)
도 8(C): 미세아교세포(Microglia)
도 8(D): 해마신경세포(Hippocampal Neuronal cell)
도 8(E): 미세아교세포(Microglia)에서 3회 이상 반복 실험한 후 상대적인 값을 수치화하여 나타냄.
도 9는 본 발명의 실시예 화합물 처리로 인한 p38 MAPK 관련 사이토카인 생성량이 감소됨을 ELISA 방법으로 확인한 결과이다.
도 10은 본 발명의 실시예 화합물이 p38 MAPK 관련 사이토카인의 mRNA 발현량 감소시킴을 qRT-PCR 방법으로 확인한 결과이다.
1 is a view showing the chemical structural formulas of the compounds of Examples 1 to 9 and Comparative Examples 1 to 3 according to the present invention.
2 is a result of confirming the p38 MAPK removal ability of 7 types of compounds according to an embodiment of the present invention by immunoblotting method.
3 is a result confirming the removal ability of p38 (Flag) and P-p38 according to the treatment concentration of Compound 24 according to Example 2 of the present invention by immunoblotting method.
Figure 4 shows [p38 Ligand Moiety in Comparative Example 1 (Compound 43) and Example 2 (Compound 24) in which biotin was conjugated instead of [E3 Ligand Moiety (thalidomide derivative)] in Example 2 (Compound 24) of the present invention. ] is the result of evaluating the p38 MAPK binding ability of Comparative Example 2 (Compound 45) in which Biotin was bound to the Linker, respectively.
5 is a result confirming that the Example compound according to the present invention removes p38 MAPK using the intracellular proteasome system by immunoblotting method.
6 is a result of confirming that the Example compound according to the present invention removes p38 MAPK using cereblon (CRBN), one of the E3 ligase complex proteins in cells, by immunoblotting method.
7 is a result confirming that the Example compound according to the present invention removes p38 MAPK using the ubiquitination system by SDS-PAGE and immunoblotting methods.
8 is a result confirming that the compound of Example according to the present invention induces a concentration-dependent decrease in p38 MAPK and P-p38 MAPK in neurons by an immunoblotting method.
Figure 8(A): Astrocytes
Figure 8(B): Neuroblasts
Figure 8(C): Microglia
Figure 8(D): Hippocampal Neuronal cells
Figure 8 (E): After repeated experiments three or more times in microglia (Microglia), the relative values are shown numerically.
9 is a result of confirming by the ELISA method that the amount of p38 MAPK-related cytokine production due to treatment with the compound of Example of the present invention is reduced.
10 is a result confirming that the Example compound of the present invention reduces the mRNA expression level of p38 MAPK-related cytokines by qRT-PCR method.

이하, 본 발명을 상세히 설명한다.Hereinafter, the present invention will be described in detail.

화학식 A로 표시되는 화합물A compound represented by the formula (A)

본 발명은 하기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 제공한다.The present invention provides a compound represented by the following formula (A) or a pharmaceutically acceptable salt thereof.

[화학식 A][Formula A]

Figure pat00013
Figure pat00013

(상기 화학식 A에 있어서,(In the formula A,

상기 p38 Ligand Moiety는

Figure pat00014
이고;The p38 Ligand Moiety is
Figure pat00014
ego;

상기 E3 Ligand Moiety는

Figure pat00015
이고;The E3 Ligand Moiety is
Figure pat00015
ego;

상기 Linker는

Figure pat00016
,
Figure pat00017
,
Figure pat00018
,
Figure pat00019
,
Figure pat00020
또는
Figure pat00021
이고,The Linker is
Figure pat00016
,
Figure pat00017
,
Figure pat00018
,
Figure pat00019
,
Figure pat00020
or
Figure pat00021
ego,

상기 n 및 m은 독립적으로 1 내지 10의 정수이고,Wherein n and m are independently integers from 1 to 10,

상기 a 및 b는 독립적으로 1 내지 10의 정수이다).wherein a and b are independently integers from 1 to 10).

바람직하게는,Preferably,

상기 n은 1 내지 5의 정수이고,Wherein n is an integer from 1 to 5,

상기 m은 3 내지 7의 정수이며,Wherein m is an integer of 3 to 7,

상기 a는 1 내지 5의 정수이고,wherein a is an integer from 1 to 5,

상기 b는 1 내지 7의 정수일 수 있다.The b may be an integer of 1 to 7.

더욱 바람직하게는,More preferably,

상기 n은 1 내지 3의 정수이고,Wherein n is an integer of 1 to 3,

상기 m은 5이며,wherein m is 5,

상기 a는 1 내지 2의 정수이고,wherein a is an integer of 1 to 2,

상기 b는 2 또는 5일 수 있다.b may be 2 or 5.

특히 바람직하게는,Particularly preferably,

상기 화학식 A로 표시되는 화합물은 하기 화합물군으로부터 선택되는 1종일 수 있다.The compound represented by Formula A may be one selected from the following compound group.

1) 2-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)에틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (23);1) 2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl (2-(2,6- dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (23);

2) 2-(2-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)에톡시)에틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (24);2) 2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl (2 -(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (24);

3) 2-(2-(2-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)에톡시)에톡시)에틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (25);3) 2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy) ethoxy)ethyl(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (25);

4) 5-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)펜틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (26);4) 5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl(2-(2,6- dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (26);

5) N-사이클로프로필-4'-(4-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (31);5) N-cyclopropyl-4'-(4-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy) ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (31);

6) N-사이클로프로필-4'-(4-(2-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)에톡시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (32);6) N-cyclopropyl-4'-(4-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl) )oxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (32);

7) N-사이클로프로필-4'-(4-(2-(2-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)에톡시)에톡시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (33);7) N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-) 4-yl)oxy)ethoxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (33);

8) N-사이클로프로필-4'-(4-((5-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)펜틸)옥시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (34); 및8) N-Cyclopropyl-4'-(4-((5-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy )pentyl)oxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (34); and

9) (S)-N-사이클로프로필-4'-(4-(2-(2-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)아세트아미도)에톡시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카르복스아미드 (41).9) (S)-N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-diox) Soisoindolin-4-yl)oxy)acetamido)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (41).

본 발명에 따른 화학식 A로 표시되는 화합물에 있어서, In the compound represented by Formula A according to the present invention,

상기 [p38 Ligand Moiety]는 p38 MAPK를 선택적으로 타겟팅하여 결합하는 역할을 하고,The [p38 Ligand Moiety] serves to selectively target and bind p38 MAPK,

상기 [E3 Ligand Moiety]는 E3(Ubiquitin Ligase)를 선택적으로 타겟팅하여 결합하는 역할을 한다.The [E3 Ligand Moiety] serves to selectively target and bind E3 (Ubiquitin Ligase).

본 발명에 따른 화학식 A로 표시되는 화합물의 p38 MAPK 제거 원리는 다음과 같다. 하기 단계 1 및 단계 2는 순서가 바뀔수도 있다.The principle of p38 MAPK removal of the compound represented by Formula A according to the present invention is as follows. The order of steps 1 and 2 below may be reversed.

(단계 1) [p38 Ligand Moiety]가 p38 MAPK에 결합(Step 1) [p38 Ligand Moiety] binds to p38 MAPK

(단계 2) [E3 Ligand Moiety]에 E3 결합(Step 2) E3 binding to [E3 Ligand Moiety]

(단계 3) 상기 결합된 E3가 p38 MAPK를 제거(Step 3) The bound E3 removes p38 MAPK

약학적으로 허용가능한 염pharmaceutically acceptable salts

본 발명의 유효물질은 약학적으로 허용 가능한 염의 형태로 사용할 수 있으며, 염으로는 약학적으로 허용가능한 유리산(free acid)에 의해 형성된 산부가염이 유용하다. 약학적으로 허용가능한 염이란 표현은 환자에게 비교적 비독성이고 무해한 유효작용을 갖는 농도로서 이 염에 기인한 부작용이 유효물질의 염기 화합물의 이로운 효능을 떨어뜨리지 않는 유효물질의 염기 화합물의 어떠한 유기 또는 무기 부가염을 의미한다. 이들 염은 유리산으로는 무기산과 유기산을 사용할 수 있으며, 무기산으로는 염산, 브롬산, 질산, 황산, 과염소산, 인산 등을 사용할 수 있고, 유기산으로는 구연산, 초산, 젖산, 말레산, 푸마린산, 글루콘산, 메탄설폰산, 글리콘산, 숙신산, 타타르산, 갈룩투론산, 엠본산, 글루탐산, 아스파르트산, 옥살산, (D) 또는 (L) 말산, 말레산, 메테인설폰산, 에테인설폰산, 4-톨루엔술폰산, 살리실산, 시트르산, 벤조산 또는 말론산 등을 사용할 수 있다. 또한, 이들 염은 알칼리 금속염(나트륨염, 칼륨염 등) 및 알칼리 토금속염(칼슘염, 마그네슘염 등) 등을 포함한다. 예를 들면, 산부가염으로는 아세테이트, 아스파테이트, 벤즈에이트, 베실레이트, 바이카보네이트/카보네이트, 바이설페이트/설페이트, 보레이트, 캄실레이트, 시트레이트, 에디실레이트, 에실레이트, 포메이트, 퓨마레이트, 글루셉테이트, 글루코네이트, 글루큐로네이트, 헥사플루오로포스페이트, 하이벤제이트, 하이드로클로라이드/클로라이드, 하이드로브로마이드/브로마이드, 하이드로요오디드/요오디드, 이세티오네이트, 락테이트, 말레이트, 말리에이트, 말로네이트, 메실레이트, 메틸설페이트, 나프틸레이트, 2-나프실레이트, 니코티네이트, 나이트레이트, 오로테이트, 옥살레이트, 팔미테이트, 파모에이트, 포스페이트/수소 포스페이트/이수소 포스페이트, 사카레이트, 스테아레이트, 석시네이트, 타르트레이트, 토실레이트, 트리플루오로아세테이트, 알루미늄, 알기닌, 벤자틴, 칼슘, 콜린, 디에틸아민, 디올아민, 글라이신, 라이신, 마그네슘, 메글루민, 올아민, 칼륨, 나트륨, 트로메타민, 아연염 등이 포함될 수 있으며, 이들 중 하이드로클로라이드 또는 트리플루오로아세테이트가 바람직하다.The active substance of the present invention can be used in the form of a pharmaceutically acceptable salt, and as the salt, an acid addition salt formed by a pharmaceutically acceptable free acid is useful. The expression pharmaceutically acceptable salt is a concentration having an effective action that is relatively non-toxic and harmless to a patient, and any organic or means inorganic addition salts. For these salts, inorganic acids and organic acids can be used as free acids, and hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, perchloric acid, phosphoric acid, etc. can be used as inorganic acids, and citric acid, acetic acid, lactic acid, maleic acid, and fumarin can be used as organic acids. Acid, gluconic acid, methanesulfonic acid, glycolic acid, succinic acid, tartaric acid, galacturonic acid, embonic acid, glutamic acid, aspartic acid, oxalic acid, (D) or (L) malic acid, maleic acid, methanesulfonic acid, ethanesulfonic acid Phonic acid, 4-toluenesulfonic acid, salicylic acid, citric acid, benzoic acid or malonic acid may be used. Further, these salts include alkali metal salts (sodium salt, potassium salt, etc.) and alkaline earth metal salt (calcium salt, magnesium salt, etc.) and the like. For example, acid addition salts include acetate, aspartate, benzate, besylate, bicarbonate/carbonate, bisulfate/sulfate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, Gluceptate, gluconate, glucuronate, hexafluorophosphate, hebenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, malate ate, malonate, mesylate, methylsulfate, naphthylate, 2-naphsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate Late, stearate, succinate, tartrate, tosylate, trifluoroacetate, aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, Potassium, sodium, tromethamine, zinc salt, etc. may be included, and among these, hydrochloride or trifluoroacetate is preferable.

본 발명에 따른 산 부가염은 통상의 방법, 예를 들면, 유효물질을 유기용매, 예를 들면 메탄올, 에탄올, 아세톤, 메틸렌클로라이드, 아세토니트릴 등에 녹이고 유기산 또는 무기산을 가하여 생성된 침전물을 여과, 건조하여 제조되거나, 용매와 과량의 산을 감압 증류한 후 건조하거나 유기용매 하에서 결정화시켜셔 제조할 수 있다.The acid addition salt according to the present invention is prepared by a conventional method, for example, by dissolving an active substance in an organic solvent such as methanol, ethanol, acetone, methylene chloride, acetonitrile, etc. and adding an organic or inorganic acid to filter and dry the resulting precipitate. or by distilling the solvent and excess acid under reduced pressure and then drying or crystallizing in an organic solvent.

또한, 염기를 사용하여 약학적으로 허용 가능한 금속염을 만들 수 있다. 알칼리 금속 또는 알칼리 토금속 염은 예를 들면 화합물을 과량의 알칼리 금속 수산화물 또는 알칼리 토금속 수산화물 용액 중에 용해하고, 비용해 화합물 염을 여과하고, 여액을 증발, 건조시켜 얻는다. 이때, 금속염으로는 나트륨, 칼륨 또는 칼슘염을 제조하는 것이 제약상 적합하다. 또한, 이에 대응하는 은 염은 알칼리 금속 또는 알칼리 토금속 염을 적당한 은 염(예, 질산은)과 반응시켜 얻는다.In addition, a pharmaceutically acceptable metal salt may be prepared using a base. The alkali metal or alkaline earth metal salt is obtained, for example, by dissolving the compound in an excess alkali metal hydroxide or alkaline earth metal hydroxide solution, filtering the undissolved compound salt, and evaporating and drying the filtrate. In this case, it is pharmaceutically suitable to prepare a sodium, potassium or calcium salt as the metal salt. The corresponding silver salt is also obtained by reacting an alkali metal or alkaline earth metal salt with a suitable silver salt (eg silver nitrate).

나아가, 본 발명은 유효물질 및 이의 약학적으로 허용되는 염뿐만 아니라, 이로부터 제조될 수 있는 가능한 용매화물, 수화물, 이성질체, 광학 이성질체 등을 모두 포함한다.Furthermore, the present invention includes all possible solvates, hydrates, isomers, optical isomers and the like that can be prepared therefrom, as well as active substances and pharmaceutically acceptable salts thereof.

제조방법 1Manufacturing method 1

본 발명은 하기 반응식 1에 나타난 바와 같이,The present invention, as shown in Scheme 1 below,

화합물 8A와 화합물 13을 유기용매에서 반응시켜 화합물 14A를 합성하는 단계(단계 1);synthesizing compound 14A by reacting compound 8A with compound 13 in an organic solvent (step 1);

화합물 14A와 4-니트로페닐클로로포르메이트를 유기용매에서 반응시켜 화합물 19A를 합성하는 단계(단계 2); 및synthesizing compound 19A by reacting compound 14A with 4-nitrophenylchloroformate in an organic solvent (step 2); and

화합물 19A와 화합물 5를 유기용매에서 반응시켜 화합물 A1을 합성하는 단계(단계 3);를 포함하는 화합물 A1의 제조방법을 제공한다.It provides a method for preparing compound A1, comprising the step of reacting compound 19A and compound 5 in an organic solvent to synthesize compound A1 (step 3).

[반응식 1][Scheme 1]

Figure pat00022
Figure pat00022

(상기 반응식 1에서,(In Scheme 1,

L1은 -(CH2)n-OH 또는 -(CH2CH2-O)n-CH2CH2OH이고, 여기서 상기 n은 1 내지 10의 정수이고;L 1 is —(CH 2 ) n —OH or —(CH 2 CH 2 —O) n —CH 2 CH 2 OH, wherein n is an integer from 1 to 10;

L2는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).L 2 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).

상기 단계 1 내지 단계 3의 유기용매는 독립적으로 DMF(dimethylformamide), DCM(dichloromethane), 에탄올, 테트라하이드로퓨란(THF), 벤젠, KOH/MeOH, MeOH, 톨루엔, 디이소프로필에테르, 디에틸에테르, 디옥산, 아세토니트릴, 디메틸아세트아미드(DMA), 디메틸설폭사이드(DMSO), 아세톤, 클로로벤젠 등을 단독 또는 혼합하여 사용할 수 있다.The organic solvent of steps 1 to 3 is independently DMF (dimethylformamide), DCM (dichloromethane), ethanol, tetrahydrofuran (THF), benzene, KOH / MeOH, MeOH, toluene, diisopropyl ether, diethyl ether, Dioxane, acetonitrile, dimethylacetamide (DMA), dimethyl sulfoxide (DMSO), acetone, chlorobenzene, etc. may be used alone or in combination.

제조방법 2Manufacturing method 2

본 발명은 하기 반응식 2에 나타난 바와 같이,The present invention is as shown in Scheme 2 below,

화합물 8A와 화합물 13을 유기용매에서 반응시켜 화합물 14A를 합성하는 단계(단계 1);synthesizing compound 14A by reacting compound 8A with compound 13 in an organic solvent (step 1);

화합물 14A와 p-TsCl(p-Toluenesulfonyl chloride)를 유기용매에서 반응시켜 화합물 27A를 합성하는 단계(단계 2); 및synthesizing compound 27A by reacting compound 14A with p-TsCl (p-Toluenesulfonyl chloride) in an organic solvent (step 2); and

화합물 27A와 화합물 6을 유기용매에서 반응시켜 화합물 A2를 합성하는 단계(단계 3);를 포함하는 화합물 A2의 제조방법을 제공한다.It provides a method for preparing compound A2, comprising the step of reacting compound 27A and compound 6 in an organic solvent to synthesize compound A2 (step 3).

[반응식 2][Scheme 2]

Figure pat00023
Figure pat00023

(상기 반응식 2에서,(In Scheme 2,

L1은 -(CH2)n-OH 또는 -(CH2CH2-O)n-CH2CH2OH이고, 여기서 상기 n은 1 내지 10의 정수이고;L 1 is —(CH 2 ) n —OH or —(CH 2 CH 2 —O) n —CH 2 CH 2 OH, wherein n is an integer from 1 to 10;

L2는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).L 2 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).

상기 단계 1 내지 단계 3의 유기용매는 독립적으로 DMF(dimethylformamide), DCM(dichloromethane), 에탄올, 테트라하이드로퓨란(THF), 벤젠, KOH/MeOH, MeOH, 톨루엔, 디이소프로필에테르, 디에틸에테르, 디옥산, 아세토니트릴, 디메틸아세트아미드(DMA), 디메틸설폭사이드(DMSO), 아세톤, 클로로벤젠 등을 단독 또는 혼합하여 사용할 수 있다.The organic solvent of steps 1 to 3 is independently DMF (dimethylformamide), DCM (dichloromethane), ethanol, tetrahydrofuran (THF), benzene, KOH / MeOH, MeOH, toluene, diisopropyl ether, diethyl ether, Dioxane, acetonitrile, dimethylacetamide (DMA), dimethyl sulfoxide (DMSO), acetone, chlorobenzene, etc. may be used alone or in combination.

제조방법 3Manufacturing method 3

본 발명은 하기 반응식 3에 나타난 바와 같이,The present invention, as shown in Scheme 3 below,

화합물 14와 화합물 36A를 유기용매에서 반응시켜 화합물 37A를 합성하는 단계(단계 1);synthesizing compound 37A by reacting compound 14 with compound 36A in an organic solvent (step 1);

화합물 37A와 염산을 유기용매에서 반응시켜 화합물 38A를 합성하는 단계(단계 2); 및synthesizing compound 38A by reacting compound 37A with hydrochloric acid in an organic solvent (step 2); and

화합물 38A와 화합물 40을 유기용매에서 반응시켜 화합물 A3을 합성하는 단계(단계 3);를 포함하는 화합물 A3의 제조방법을 제공한다.It provides a method for preparing compound A3, comprising the step of reacting compound 38A and compound 40 in an organic solvent to synthesize compound A3 (step 3).

[반응식 3][Scheme 3]

Figure pat00024
Figure pat00024

(상기 반응식 3에서,(In Scheme 3,

L3은 -(CH2)n-OMs 또는 -(CH2CH2-O)n-CH2CH2OMs이고, 여기서 상기 n은 1 내지 10의 정수이고;L 3 is -(CH 2 ) n -OMs or -(CH 2 CH 2 -O) n -CH 2 CH 2 OMs, wherein n is an integer from 1 to 10;

L4는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).L 4 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).

상기 단계 1 내지 단계 3의 유기용매는 독립적으로 DMF(dimethylformamide), DCM(dichloromethane), 에탄올, 테트라하이드로퓨란(THF), 벤젠, KOH/MeOH, MeOH, 톨루엔, 디이소프로필에테르, 디에틸에테르, 디옥산, 아세토니트릴, 디메틸아세트아미드(DMA), 디메틸설폭사이드(DMSO), 아세톤, 클로로벤젠 등을 단독 또는 혼합하여 사용할 수 있다.The organic solvent of steps 1 to 3 is independently DMF (dimethylformamide), DCM (dichloromethane), ethanol, tetrahydrofuran (THF), benzene, KOH / MeOH, MeOH, toluene, diisopropyl ether, diethyl ether, Dioxane, acetonitrile, dimethylacetamide (DMA), dimethyl sulfoxide (DMSO), acetone, chlorobenzene, etc. may be used alone or in combination.

만성 염증성 질환 예방 또는 치료용 약학적 조성물Pharmaceutical composition for preventing or treating chronic inflammatory disease

본 발명은 상기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 치료용 약학적 조성물을 제공한다.The present invention provides a pharmaceutical composition for the prevention or treatment of chronic inflammatory diseases comprising the compound represented by Formula A or a pharmaceutically acceptable salt thereof.

상기 만성 염증성 질환은 알츠하이머, 파킨슨 병, 염증성 장질환, 비알콜성 만성 간염, 만성 간염, 크론병, 췌장염, 식도염, 위염, 대장염, 궤양성 대장염, 폐렴, 기관지염, 인후염, 심근경색, 심부전, 관절염, 건선성 관절염, 류마티스 관절염, 신부전, 건선, 빈혈, 당뇨, 섬유화증, 다발성 경화증, 전신 홍반 루프스, 강직성 척추염, 천식, 만성폐쇄성폐질환, 치주염, 신경병증 통증, 특발성 염증성 근육병증 등일 수 있다.The chronic inflammatory disease is Alzheimer's, Parkinson's disease, inflammatory bowel disease, non-alcoholic chronic hepatitis, chronic hepatitis, Crohn's disease, pancreatitis, esophagitis, gastritis, colitis, ulcerative colitis, pneumonia, bronchitis, sore throat, myocardial infarction, heart failure, arthritis , psoriatic arthritis, rheumatoid arthritis, renal failure, psoriasis, anemia, diabetes, fibrosis, multiple sclerosis, systemic lupus erythematosus, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, periodontitis, neuropathic pain, idiopathic inflammatory myopathy, and the like.

본 발명의 유효물질은 임상 투여시에 경구 및 비경구의 여러 가지 제형으로 투여될 수 있으며, 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 제조된다.The active substance of the present invention can be administered in various oral and parenteral dosage forms during clinical administration, and when formulating, commonly used diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, surfactants, etc. are used. is manufactured by

경구투여를 위한 고형 제제에는 정제, 환자, 산제, 과립제, 캡슐제, 트로키제 등이 포함되며, 이러한 고형 제제는 하나 이상의 본 발명의 유효물질에 적어도 하나 이상의 부형제 예를 들면, 전분, 탄산칼슘, 수크로스(sucrose), 락토오스(lactose) 또는 젤라틴 등을 섞어 조제된다. 또한, 단순한 부형제 외에 마그네슘 스티레이트 탈크 같은 윤활제들도 사용된다. 경구 투여를 위한 액상 제제로는 현탁제, 내용액제, 유제 또는 시럽제 등이 해당되는데, 흔히 사용되는 단순 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다.Solid preparations for oral administration include tablets, patients, powders, granules, capsules, troches, etc., and such solid preparations include at least one or more excipients, for example, starch, calcium carbonate, It is prepared by mixing sucrose, lactose, or gelatin. In addition to simple excipients, lubricants such as magnesium stearate talc are also used. Liquid formulations for oral administration include suspensions, solutions, emulsions, or syrups. In addition to commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included. can

비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁용제, 유제, 동결건조제제, 좌제 등이 포함된다. 비수성용제, 현탁용제로는 프로필렌글리콜, 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 글리세롤, 젤라틴 등이 사용될 수 있다.Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspension solutions, emulsions, lyophilized formulations, suppositories, and the like. Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate. As the base of the suppository, witepsol, macrogol, tween 61, cacao butter, laurin, glycerol, gelatin, etc. may be used.

또한, 본 발명의 유효물질의 인체에 대한 효과적인 투여량은 환자의 나이, 몸무게, 성별, 투여형태, 건강상태 및 질환 정도에 따라 달라질 수 있으며, 일반적으로 약 0.001-100 mg/kg/일이며, 바람직하게는 0.01-35 mg/kg/일이다. 몸무게가 70㎏인 성인 환자를 기준으로 할 때, 일반적으로 0.07-7000 mg/일이며, 바람직하게는 0.7-2500 ㎎/일이며, 의사 또는 약사의 판단에 따라 일정시간 간격으로 1일 1회 내지 수회로 분할 투여할 수도 있다.In addition, the effective dosage of the active substance of the present invention to the human body may vary depending on the patient's age, weight, sex, dosage form, health status and disease degree, and is generally about 0.001-100 mg/kg/day, Preferably it is 0.01-35 mg/kg/day. Based on an adult patient weighing 70 kg, it is generally 0.07-7000 mg/day, preferably 0.7-2500 mg/day, and once a day at regular time intervals according to the judgment of a doctor or pharmacist It may be administered in several divided doses.

만성 염증성 질환 예방 또는 개선용 건강기능식품/건강식품 조성물Health functional food/health food composition for preventing or improving chronic inflammatory disease

본 발명은 상기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 개선용 건강기능식품 조성물을 제공한다.The present invention provides a health functional food composition for preventing or improving chronic inflammatory diseases comprising the compound represented by Formula A or a pharmaceutically acceptable salt thereof.

또한, 본 발명은 상기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 개선용 건강식품 조성물을 제공한다.In addition, the present invention provides a health food composition for preventing or improving chronic inflammatory diseases comprising the compound represented by Formula A or a pharmaceutically acceptable salt thereof.

상기 만성 염증성 질환은 알츠하이머, 파킨슨 병, 염증성 장질환, 비알콜성 만성 간염, 만성 간염, 크론병, 췌장염, 식도염, 위염, 대장염, 궤양성 대장염, 폐렴, 기관지염, 인후염, 심근경색, 심부전, 관절염, 건선성 관절염, 류마티스 관절염, 신부전, 건선, 빈혈, 당뇨, 섬유화증, 다발성 경화증, 전신 홍반 루프스, 강직성 척추염, 천식, 만성폐쇄성폐질환, 치주염, 신경병증 통증, 특발성 염증성 근육병증 등일 수 있다.The chronic inflammatory disease is Alzheimer's, Parkinson's disease, inflammatory bowel disease, non-alcoholic chronic hepatitis, chronic hepatitis, Crohn's disease, pancreatitis, esophagitis, gastritis, colitis, ulcerative colitis, pneumonia, bronchitis, sore throat, myocardial infarction, heart failure, arthritis , psoriatic arthritis, rheumatoid arthritis, renal failure, psoriasis, anemia, diabetes, fibrosis, multiple sclerosis, systemic lupus erythematosus, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, periodontitis, neuropathic pain, idiopathic inflammatory myopathy, and the like.

식품의 종류에는 특별한 제한은 없다. 본 발명의 유효물질을 첨가할 수 있는 식품의 예로는 드링크제, 육류, 소시지, 빵, 비스킷, 떡, 초콜릿, 캔디류, 스낵류, 과자류, 피자, 라면, 기타 면류, 껌류, 아이스크림류를 포함한 낙농제품, 각종 스프, 음료수, 알코올 음료 및 비타민 복합제, 유제품 및 유가공 제품 등이 있으며, 통상적인 의미에서의 건강식품 및 건강기능성식품을 모두 포함한다.There are no special restrictions on the type of food. Examples of foods to which the active substance of the present invention can be added include drinks, meat, sausage, bread, biscuits, rice cakes, chocolate, candy, snacks, confectionery, pizza, ramen, other noodles, gums, dairy products including ice cream, There are various soups, beverages, alcoholic beverages and vitamin complexes, dairy products, and dairy products, and includes all health foods and health functional foods in the ordinary sense.

본 발명에 따른 유효물질을 함유하는 건강식품 및 건강기능성식품 조성물은 식품에 그대로 첨가하거나 다른 식품 또는 식품 성분과 함께 사용될 수 있고, 통상적인 방법에 따라 적절하게 사용될 수 있다. 유효물질의 혼합량은 그의 사용 목적(예방 또는 개선용)에 따라 적합하게 결정될 수 있다. 일반적으로, 건강식품 및 건강기능성식품 중의 상기 조성물의 양은 전체 식품 중량의 0.1 내지 90 중량부로 가할 수 있다. 그러나 건강 유지를 목적으로 하거나 또는 건강 조절을 목적으로 하는 장기간의 섭취의 경우에는 상기 양은 상기 범위 이하일 수 있으며, 안전성 면에서 아무런 문제가 없기 때문에 유효성분은 상기 범위 이상의 양으로도 사용될 수 있다.The health food and health functional food composition containing the active substance according to the present invention may be added to food as it is or used together with other food or food ingredients, and may be appropriately used according to a conventional method. The mixing amount of the active substance may be appropriately determined depending on the purpose of its use (for prevention or improvement). In general, the amount of the composition in health food and health functional food may be added in an amount of 0.1 to 90 parts by weight based on the total weight of the food. However, in the case of long-term intake for health maintenance or health control, the amount may be less than the above range, and since there is no problem in terms of safety, the active ingredient may be used in an amount greater than or equal to the above range.

본 발명의 건강식품 및 건강기능성식품 조성물은 지시된 비율로 필수 성분으로서 본 발명 유효물질을 함유하는 외에는 다른 성분에는 특별한 제한이 없으며 통상의 음료와 같이 여러 가지 향미제 또는 천연 탄수화물 등을 추가 성분으로서 함유할 수 있다. 상술한 천연 탄수화물의 예는 모노사카라이드, 예를 들어, 포도당, 과당 등; 디사카라이드, 예를 들어 말토스, 슈크로스 등; 및 폴리사카라이드, 예를 들어 덱스트린, 시클로덱스트린 등과 같은 통상적인 당, 및 자일리톨, 소르비톨, 에리트라이톨 등의 당알코올이다. 상술한 것 이외의 향미제로서 천연 향미제(타우마틴, 스테비아 추출물(예를 들어 레바우디오시드 A, 글리시르히진등) 및 합성 향미제(사카린, 아스파르탐 등)를 유리하게 사용할 수 있다. 상기 천연 탄수화물의 비율은 본 발명의 건강기능성 식품 조성물 100 당 일반적으로 약 1 내지 20 g, 바람직하게는 약 5 내지 12 g이다.The health food and health functional food composition of the present invention is not particularly limited in other ingredients other than containing the active substance of the present invention as an essential ingredient in the indicated ratio, and various flavoring agents or natural carbohydrates are added as additional ingredients like conventional beverages. may contain. Examples of the above-mentioned natural carbohydrates include monosaccharides such as glucose, fructose and the like; disaccharides such as maltose, sucrose and the like; and polysaccharides such as conventional sugars such as dextrin and cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol. As flavoring agents other than those described above, natural flavoring agents (taumatine, stevia extract (eg, rebaudioside A, glycyrrhizin, etc.) and synthetic flavoring agents (saccharin, aspartame, etc.) can be advantageously used. The proportion of the natural carbohydrate is generally about 1 to 20 g, preferably about 5 to 12 g per 100 nutraceuticals of the present invention.

상기 외에 본 발명의 유효물질을 함유하는 건강식품 및 건강기능성식품 조성물은 여러 가지 영양제, 비타민, 광물(전해질), 합성 풍미제 및 천연 풍미제 등의 풍미제, 착색제 및 중진제(치즈, 초콜릿 등), 펙트산 및 그의 염, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 글리세린, 알코올, 탄산음료에 사용되는 탄산화제 등을 함유할 수 있다. 그 밖에 본 발명의 건강식품 및 건강기능성식품 조성물은 천연 과일쥬스 및 과일쥬스 음료 및 야채 음료의 제조를 위한 과육을 함유할 수 있다.In addition to the above, health food and health functional food composition containing the active substance of the present invention are various nutrients, vitamins, minerals (electrolytes), synthetic flavoring agents and flavoring agents such as natural flavoring agents, coloring agents and thickening agents (cheese, chocolate, etc.) ), pectic acid and its salts, alginic acid and its salts, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, glycerin, alcohols, carbonation agents used in carbonated beverages, and the like. In addition, the health food and health functional food composition of the present invention may contain fruit for the production of natural fruit juice, fruit juice beverage, and vegetable beverage.

이러한 성분은 독립적으로 또는 조합하여 사용할 수 있다. 이러한 첨가제의 비율은 그렇게 중요하진 않지만 본 발명의 유효물질을 함유하는 건강식품 및 건강기능성식품 조성물 100 중량부 당 0.1 내지 약 20 중량부의 범위에서 선택되는 것이 일반적이다.These components may be used independently or in combination. The proportion of these additives is not so important, but is generally selected in the range of 0.1 to about 20 parts by weight per 100 parts by weight of the health food and health functional food composition containing the active substance of the present invention.

이하, 본 발명을 하기의 실시예에 의하여 더욱 상세하게 설명한다. 단, 하기의 실시예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 하기의 실시예에 의해 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail with reference to the following examples. However, the following examples are merely illustrative of the present invention, and the content of the present invention is not limited by the following examples.

<제조예 1> 탈리도미도 유도체(화합물 5 및 화합물 6)의 제조<Preparation Example 1> Preparation of thalidomido derivatives (Compound 5 and Compound 6)

Figure pat00025
Figure pat00025

tert-butyl (2,6-dioxopiperidin-3-yl)carbamate(2)의 제조Preparation of tert-butyl (2,6-dioxopiperidin-3-yl)carbamate (2)

알려진 화합물 (tert-butoxycarbonyl)glutamine (100mg, 0.406mmol)와 N-Hydroxysuccinimide (52mg, 0.447mmol), DCC(92mg, 0.447mmol)를 Ar gas로 충진 후 DMF 3mL에 녹였다. 반응은 80℃에서 진행했다. TLC로 출발 물질이 없어진 것이 확인되면 evaporator와 high vacuum 장치를 이용해 DMF를 제거하였다. 이후 반응 중 생긴 urea를 EtOAc로 filtering 해주고, 차가운 ethyl ether를 이용하여 결정화하여 화합물 2를 얻어냈다(92mg. 99%).Known compounds (tert-butoxycarbonyl)glutamine (100mg, 0.406mmol), N-Hydroxysuccinimide (52mg, 0.447mmol), and DCC (92mg, 0.447mmol) were filled with Ar gas and dissolved in 3mL of DMF. The reaction was carried out at 80 °C. When it was confirmed that the starting material disappeared by TLC, DMF was removed using an evaporator and a high vacuum device. After that, the urea generated during the reaction was filtered with EtOAc and crystallized using cold ethyl ether to obtain compound 2 (92 mg. 99%).

1H-NMR (400 MHz, DMSO-d6) δ 10.72 (s, 1H), 7.10 (d, J = 7.6 Hz, 1H), 7.92 (d, J = 8.1 Hz, 2H), 4.17 (m, 1H), 2.67 (m, 1H), 2.41 (m, 1H), 1.86 (m, 2H), 1.35 (s, 9H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 10.72 (s, 1H), 7.10 (d, J = 7.6 Hz, 1H), 7.92 (d, J = 8.1 Hz, 2H), 4.17 (m, 1H) ), 2.67 (m, 1H), 2.41 (m, 1H), 1.86 (m, 2H), 1.35 (s, 9H).

2,2,2-trifluoroacetaldehyde, 2,6-dioxopiperidin-3-aminium salt(3)의 제조Preparation of 2,2,2-trifluoroacetaldehyde, 2,6-dioxopiperidin-3-aminium salt (3)

합성된 화합물 2 (110.7mg, 0.485mmol)에 Ar gas 충진 후 CH2Cl2와 TFA를 각각 2mL씩 적가하여 상온(25℃)에서 반응을 진행했다. TLC로 출발 물질이 없어진 것이 확인되면 evaporator와 high vacuum 장치를 이용해 CH2Cl2를 제거하여 화합물 3을 얻어냈다(119.2mg, 99%). After filling Ar gas to the synthesized compound 2 (110.7 mg, 0.485 mmol), 2 mL of CH 2 Cl 2 and 2 mL of TFA were added dropwise, respectively, and the reaction was carried out at room temperature (25° C.). When it was confirmed that the starting material disappeared by TLC, CH 2 Cl 2 was removed using an evaporator and a high vacuum device to obtain compound 3 (119.2 mg, 99%).

1H-NMR (400 MHz, DMSO-d6) δ 11.31 (s, 1H), 8.51 (s, 3H), 4.21 (dd, J = 13.3, 5.5 Hz, 1H), 2.70 (m, 1H), 2.59 (m, 1H), 2.13 (m, 1H), 2.04 (m, 1H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.31 (s, 1H), 8.51 (s, 3H), 4.21 (dd, J = 13.3, 5.5 Hz, 1H), 2.70 (m, 1H), 2.59 (m, 1H), 2.13 (m, 1H), 2.04 (m, 1H).

2-(2,6-dioxopiperidin-3-yl)-4-nitroisoindoline-1,3-dione(4)의 제조Preparation of 2-(2,6-dioxopiperidin-3-yl)-4-nitroisoindoline-1,3-dione (4)

합성된 화합물 2 (100mg, 0.438mmol)에 Ar gas 충진 후 CH2Cl2와 TFA를 각각 2mL씩 적가하여 상온(25℃)에서 반응을 진행했다. TLC로 출발 물질이 없어진 것이 확인되면 evaporator와 high vacuum 장치를 이용해 CH2Cl2를 제거하여 화합물 3을 얻어내었다. 정제 없이 바로 화합물 3과 4-nitroisobenzofuran-1,3-dione(127mg, 0.657mmol)와 NaOAc(44mg, 0.526mmol)를 AcOH 3mL에 녹이고 reflux 반응을 진행했다. 출발 물질이 없어진 것이 TLC로 확인되면 evaporator와 high vacuum 장치를 이용해 AcOH를 제거하고 H2O를 이용하여 결정화하여 화합물 4를 얻어냈다(105mg. 79%).After filling Ar gas to the synthesized compound 2 (100mg, 0.438mmol), 2mL of CH 2 Cl 2 and TFA were added dropwise each, and the reaction was carried out at room temperature (25℃). When it was confirmed by TLC that the starting material disappeared, using an evaporator and a high vacuum device, CH 2 Cl 2 was removed to obtain compound 3 . Immediately without purification, compound 3 , 4-nitroisobenzofuran-1,3-dione (127 mg, 0.657 mmol) and NaOAc (44 mg, 0.526 mmol) were dissolved in 3 mL of AcOH, followed by reflux reaction. When it was confirmed by TLC that the starting material disappeared, AcOH was removed using an evaporator and a high vacuum device and crystallized using H 2 O to obtain compound 4 (105 mg. 79%).

1H-NMR (400 MHz, DMSO-d6) δ 11.18 (s, 1H), 8.51 (s, 3H), 8.35 (dd, J = 8.1, 0.8 Hz, 1H), 8.24 (dd, J = 7.5, 0.7 Hz, 1H), 8.12 (t, J = 7.8 Hz, 1H), 5.20 (dd, J = 13.4, 5.3 Hz, 1H), 2.89 (m, 1H), 2.61 (m, 1H), 2.52 (m, 1H), 2.07 (m, 1H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.18 (s, 1H), 8.51 (s, 3H), 8.35 (dd, J = 8.1, 0.8 Hz, 1H), 8.24 (dd, J = 7.5, 0.7 Hz, 1H), 8.12 (t, J = 7.8 Hz, 1H), 5.20 (dd, J = 13.4, 5.3 Hz, 1H), 2.89 (m, 1H), 2.61 (m, 1H), 2.52 (m, 1H), 2.07 (m, 1H).

4-amino-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione(5)의 제조Preparation of 4-amino-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (5)

합성한 화합물 4(610mg, 2.012mmol)를 DMF 5mL에 녹인다. 이후 10% Pd/C (60mg, 0.056mmol)을 추가하고 H2 gas 충진 후 상온(25℃)에서 반응을 진행했다.Dissolve the synthesized compound 4 (610mg, 2.012mmol) in 5mL of DMF. After that, 10% Pd/C (60mg, 0.056mmol) was added, and the reaction was carried out at room temperature (25℃) after filling with H 2 gas.

TLC로 출발 물질이 없어진 것이 확인되면 반응 후 남은 Pd을 제거하기 위해 DMF로 filtering 해주고, H2O를 이용하여 결정화하여 화합물 5를 얻어냈다(512mg, 93%). When it was confirmed by TLC that the starting material disappeared, it was filtered with DMF to remove Pd remaining after the reaction, and crystallized using H 2 O to obtain compound 5 (512 mg, 93%).

1H-NMR (400 MHz, DMSO-d6) δ 11.10 (s, 1H), 7.47 (t, J = 8.2 Hz, 1H), 7.01 (t, J = 6.8 Hz, 2H), 6.53 (s, 2H), 5.05 (dd, J = 13.1, 5.3 Hz, 1H), 2.89 (m, 1H), 2.59 (m, 1H), 2.02 (m, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.10 (s, 1H), 7.47 (t, J = 8.2 Hz, 1H), 7.01 (t, J = 6.8 Hz, 2H), 6.53 (s, 2H) ), 5.05 (dd, J = 13.1, 5.3 Hz, 1H), 2.89 (m, 1H), 2.59 (m, 1H), 2.02 (m, 2H).

2-(2,6-dioxopiperidin-3-yl)-4-hydroxyisoindoline-1,3-dione(6)의 제조Preparation of 2-(2,6-dioxopiperidin-3-yl)-4-hydroxyisoindoline-1,3-dione (6)

합성된 화합물 3 (94.5mg, 0.39mmol)와 4-hydroxyisobenzofuran-1,3-dione(57.64mg, 0.351mmol)와 KOAc(107.95mg, 1.1mmol)를 AcOH 3mL에 녹이고 reflux 반응을 진행했다. TLC로 출발 물질이 없어진 것이 확인되면 evaporator와 high vacuum 장치를 이용해 AcOH를 제거하고 H2O를 이용하여 결정화하여 화합물 6을 얻어냈다(54mg. 51%).The synthesized compound 3 (94.5 mg, 0.39 mmol), 4-hydroxyisobenzofuran-1,3-dione (57.64 mg, 0.351 mmol) and KOAc (107.95 mg, 1.1 mmol) were dissolved in 3 mL of AcOH, followed by reflux reaction. When it was confirmed by TLC that the starting material disappeared, AcOH was removed using an evaporator and a high vacuum device, and crystallized using H 2 O to obtain compound 6 (54 mg. 51%).

1H-NMR (400 MHz, CDCl3) δ 11.20 (s, 1H), 11.11 (s, 1H), 7.65 (t, J = 7.7 Hz, 2H), 5.07 (m, 1H), 2.89 (m, 1H), 2.59 (m, 2H), 2.03 (m,1H). 1 H-NMR (400 MHz, CDCl 3 ) δ 11.20 (s, 1H), 11.11 (s, 1H), 7.65 (t, J = 7.7 Hz, 2H), 5.07 (m, 1H), 2.89 (m, 1H) ), 2.59 (m, 2H), 2.03 (m, 1H).

<제조예 2> 화합물 9 내지 12의 제조<Preparation Example 2> Preparation of compounds 9 to 12

Figure pat00026
Figure pat00026

단계 1: 화합물 8의 제조Step 1: Preparation of compound 8

화합물 7과 피리디늄 하이드로클로라이드를 180℃에서 반응시켜, 화합물 8을 제조하였다.Compound 7 and pyridinium hydrochloride were reacted at 180° C. to prepare compound 8.

단계 2: 화합물 9-12의 제조Step 2: Preparation of compound 9-12

(4-bromophenyl)(4-(2-hydroxyethoxy)phenyl)methanone (9)의 제조Preparation of (4-bromophenyl)(4-(2-hydroxyethoxy)phenyl)methanone (9)

합성된 화합물 8(220.2mg, 0.794mmol)과 K2CO3 (274.6mg, 1.98mmol)를 Acetone 5mL와 DMF 3mL에 녹여 30분 reflux 반응을 진행했다. 이후 2-bromoethanol (0.155mL, 2.185mmol)을 적가하여 overnight 반응을 진행했다. TLC로 출발 물질이 없어진 것이 확인되면 2N-HCl로 반응을 중지시키고 EtOAc 용매로 희석시켰다. 유기층은 EtOAc과 H2O로 씻어 추출했다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거했다.The synthesized compound 8 (220.2 mg, 0.794 mmol) and K 2 CO 3 (274.6 mg, 1.98 mmol) were dissolved in 5 mL of Acetone and 3 mL of DMF, followed by a reflux reaction for 30 minutes. Thereafter, 2-bromoethanol (0.155 mL, 2.185 mmol) was added dropwise, and the reaction was carried out overnight. When it was confirmed by TLC that the starting material disappeared, the reaction was stopped with 2N-HCl and diluted with EtOAc solvent. The organic layer was extracted by washing with EtOAc and H 2 O. The extracted organic layer was dried over MgSO 4 and the solvent was removed using an evaporator.

Column chromatography를 통해 반응 혼합물을 정제하여 화합물 9를 얻어냈다(223.4mg, 87%). The reaction mixture was purified through column chromatography to obtain compound 9 (223.4 mg, 87%) .

1H-NMR (400 MHz, CDCl3) δ 7.80 (d, J = 8.8 Hz, 2H), 7.63 (s, 4H), 4.18 (t, J = 4.4 Hz, 2H), 7.80 (d, J = 8.8 Hz, 2H), 4.02 (d, J = 3.8 Hz, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.80 (d, J = 8.8 Hz, 2H), 7.63 (s, 4H), 4.18 (t, J = 4.4 Hz, 2H), 7.80 (d, J = 8.8) Hz, 2H), 4.02 (d, J = 3.8 Hz, 2H).

(4-bromophenyl)(4-(2-(2-hydroxyethoxy)ethoxy)phenyl)methanone (10)의 제조Preparation of (4-bromophenyl)(4-(2-(2-hydroxyethoxy)ethoxy)phenyl)methanone (10)

합성된 화합물 8(313.5mg, 1.13mmol)과 K2CO3 (468.53mg, 3.39mmol)를 Acetone 5mL와 DMF 3mL에 녹여 1시간 reflux 반응을 진행했다. 이후 2-(2-chloroethoxy)ethan-1-ol (0.24mL, 2.26mmol)와 NaI(169.37mg, 1.13mmol)을 적가 하여 overnight 반응을 진행했다. TLC로 출발 물질이 없어진 것이 확인되면 2N-HCl로 반응을 중지시키고 EtOAc 용매로 희석시켰다. 유기층은 EtOAc과 H2O로 씻어 추출했다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거했다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 10를 얻어냈다(364.2mg, 88%). The synthesized compound 8 (313.5 mg, 1.13 mmol) and K 2 CO 3 (468.53 mg, 3.39 mmol) were dissolved in 5 mL of Acetone and 3 mL of DMF, followed by a reflux reaction for 1 hour. Thereafter, 2-(2-chloroethoxy)ethan-1-ol (0.24 mL, 2.26 mmol) and NaI (169.37 mg, 1.13 mmol) were added dropwise, followed by overnight reaction. When it was confirmed by TLC that the starting material disappeared, the reaction was stopped with 2N-HCl and diluted with EtOAc solvent. The organic layer was extracted by washing with EtOAc and H 2 O. The extracted organic layer was dried over MgSO 4 and the solvent was removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 10 (364.2 mg, 88%).

1H-NMR (400 MHz, CDCl3) δ 7.80 (d, J = 8.8 Hz, 2H), 7.63 (s, 1H), 7.00 (d, J = 8.7 Hz, 2H), 4.24 (t, J = 4.6 Hz, 2H), 3.92 (t, J = 4.6 Hz, 2H), 3.76 (m, 2H) 3.70 (t, J = 4.4 Hz, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.80 (d, J = 8.8 Hz, 2H), 7.63 (s, 1H), 7.00 (d, J = 8.7 Hz, 2H), 4.24 (t, J = 4.6) Hz, 2H), 3.92 (t, J = 4.6 Hz, 2H), 3.76 (m, 2H) 3.70 (t, J = 4.4 Hz, 2H).

(4-bromophenyl)(4-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)phenyl)methanone (11)의 제조Preparation of (4-bromophenyl)(4-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)phenyl)methanone (11)

합성한 화합물 8(319.2mg, 1.15mmol)와 2-(2-(2-chloroethoxy)ethoxy)ethan-1-ol (0.17mL, 1.15mmol)출발 물질로 하여 상기 유도체 10와 같은 방법을 통해 화합물 11를 얻어냈다(447mg, 95%). Using the synthesized compound 8 (319.2mg, 1.15mmol) and 2-(2-(2-chloroethoxy)ethoxy)ethan-1-ol (0.17mL, 1.15mmol) as starting materials, use the same method as for the derivative 10 to compound 11 was obtained (447 mg, 95%).

1H-NMR (400 MHz, CDCl3) δ 7.78 (d, J = 8.6 Hz, 2H), 7.62 (s, 4H), 6.98 (d, J = 8.6 Hz, 2H), 4.22 (t, J = 4.6 Hz, 2H), 3.90(t, J = 4.5 Hz, 2H), 3.77-3.68 (m, 6H), 3.62 (t, J = 4.3 Hz, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.78 (d, J = 8.6 Hz, 2H), 7.62 (s, 4H), 6.98 (d, J = 8.6 Hz, 2H), 4.22 (t, J = 4.6) Hz, 2H), 3.90 (t, J = 4.5 Hz, 2H), 3.77-3.68 (m, 6H), 3.62 (t, J = 4.3 Hz, 2H).

(4-bromophenyl)(4-((5-hydroxypentyl)oxy)phenyl)methanone (12)의 제조Preparation of (4-bromophenyl)(4-((5-hydroxypentyl)oxy)phenyl)methanone (12)

합성한 화합물 8(141.5mg, 0.511mmol)와 5-bromopentan-1-ol(0.62mL, 0.511mmol)를 출발 물질로 하여 상기 유도체 10와 같은 방법을 통해 화합물 12를 얻어냈다(72.1mg, 39%). Using the synthesized compound 8 (141.5 mg, 0.511 mmol) and 5-bromopentan-1-ol (0.62 mL, 0.511 mmol) as starting materials, compound 12 was obtained in the same manner as in derivative 10 (72.1 mg, 39%). ).

1H-NMR (400 MHz, CDCl3) δ 7.75 (dd, J = 7.1,5.7 Hz, 2H), 7.59 (d, J = 5.8 Hz 4H), 6.92 (t, J = 7.4 Hz, 2H), 4.02 (d, J = 6 Hz, 2H), 3.65 (t, J = 6.1 Hz, 2H), 2.90 (m, dd, J = 31.7, 5.8 Hz, 2H), 1.86 (d, J = 5.9 Hz, 2H), 1.62 (t, J = 6.2 Hz, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.75 (dd, J = 7.1,5.7 Hz, 2H), 7.59 (d, J = 5.8 Hz 4H), 6.92 (t, J = 7.4 Hz, 2H), 4.02 (d, J = 6 Hz, 2H), 3.65 (t, J = 6.1 Hz, 2H), 2.90 (m, dd, J = 31.7, 5.8 Hz, 2H), 1.86 (d, J = 5.9 Hz, 2H) , 1.62 (t, J = 6.2 Hz, 2H).

<제조예 3> 화합물 36의 제조<Preparation Example 3> Preparation of compound 36

Figure pat00027
Figure pat00027

2-(2-((tert-butoxycarbonyl)amino)ethoxy)ethyl methanesulfonate (36)의 제조Preparation of 2-(2-((tert-butoxycarbonyl)amino)ethoxy)ethyl methanesulfonate (36)

상업적으로 구매 가능한 tert-butyl (2-(2-hydroxyethoxy)ethyl)carbamate (220 mg, 1.072 mmol)을 CH2Cl2 3ml에 녹이고 0℃에서 Et3N (2.358 mmol) 넣은 후 MsCl (1.179 mmol)을 천천히 dropwise하여 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인되면 NH4Cl을 이용하여 반응을 중지시킨다. 유기층은 CH2Cl2과 H2O로 씻어 추출한다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거한다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 36 을 얻어냈다(297 mg, 98%).Dissolve commercially available tert-butyl (2-(2-hydroxyethoxy)ethyl)carbamate (220 mg, 1.072 mmol) in 3 ml of CH 2 Cl 2 , add Et3N (2.358 mmol) at 0° C., and slowly add MsCl (1.179 mmol) The reaction proceeds dropwise. When it is confirmed by TLC that the reaction does not proceed any further, the reaction is stopped using NH 4 Cl. The organic layer was extracted by washing with CH 2 Cl 2 and H 2 O. The extracted organic layer is dried with MgSO 4 and the solvent is removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 36 (297 mg, 98%).

1H-NMR (400 MHz, CDCl3) δ 4.89(bs, 1H), 4.37(m, 2H), 3.73(m, 2H), 3.56(m, 2H), 3.33(m, 2H), 3.07(s, 3H), 1.45(s, 9H). 1 H-NMR (400 MHz, CDCl 3 ) δ 4.89(bs, 1H), 4.37(m, 2H), 3.73(m, 2H), 3.56(m, 2H), 3.33(m, 2H), 3.07(s) , 3H), 1.45 (s, 9H).

<제조예 4> 화합물 40의 제조<Preparation Example 4> Preparation of compound 40

Figure pat00028
Figure pat00028

단계 1: 화합물 39의 제조Step 1: Preparation of compound 39

tert-butyl (S)-2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)acetate (39)의 제조Preparation of tert-butyl (S)-2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)acetate (39)

상업적으로 구매 가능한 (S)-2-(2,6-dioxopiperidin-3-yl)-4-hydroxyisoindoline-1,3-dione (100 mg, 0.365 mmol), tert-butyl 2-bromoacetate (0.365 mmol), K2CO3 (76mg, 0.548 mmol)을 DMF 2ml에 녹이고 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인되면 EtOAc용매로 희석시켰다. 유기층은 EtOAc, brine 과 H2O로 씻어 추출한다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거한다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 39 을 얻어냈다(118 mg, 83%).commercially available (S)-2-(2,6-dioxopiperidin-3-yl)-4-hydroxyisoindoline-1,3-dione (100 mg, 0.365 mmol), tert-butyl 2-bromoacetate (0.365 mmol), K 2 CO 3 (76mg, 0.548 mmol) was dissolved in 2ml of DMF to proceed with the reaction. When it was confirmed by TLC that the reaction did not proceed any further, it was diluted with EtOAc solvent. The organic layer was extracted by washing with EtOAc, brine and H 2 O. The extracted organic layer is dried with MgSO 4 and the solvent is removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 39 (118 mg, 83%).

1H-NMR (400 MHz, DMSO-d6) δ 11.11(s, 1H), 7.80(t, J = 8.0 Hz, 1H), 7.48(d, J = 7.3 Hz, 1H), 7.38(d, J = 8.4 Hz, 1H), 5.10(m, 1H), 4.97(s, 2H), 2.95-2.83, 2.64-2.52, 2.08-1.99(m, 4H), 1.43(s, 9H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.11 (s, 1H), 7.80 (t, J = 8.0 Hz, 1H), 7.48 (d, J = 7.3 Hz, 1H), 7.38 (d, J) = 8.4 Hz, 1H), 5.10(m, 1H), 4.97(s, 2H), 2.95-2.83, 2.64-2.52, 2.08-1.99(m, 4H), 1.43(s, 9H).

단계 2: 화합물 40의 제조Step 2: Preparation of compound 40

(S)-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)acetyl) oxonium 2,2,2-trifluoroacetate (40)의 제조Preparation of (S)-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)acetyl)oxonium 2,2,2-trifluoroacetate (40)

합성한 화합물 39 (100 mg, 0.257 mmol)을 CH2Cl2 1ml에 녹인 후0℃에서 TFA (0.308 mmol)을 넣고 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인되면 용매를 날리고 ether로 여러 번 씻어서 화합물 40 를 얻어 정재 없이 다음반응에 사용했다(114 mg, 99%).The synthesized compound 39 (100 mg, 0.257 mmol) was dissolved in 1 ml of CH 2 Cl 2 , and TFA (0.308 mmol) was added at 0° C. and the reaction proceeded. When it was confirmed by TLC that the reaction did not proceed any further, the solvent was evaporated and washed with ether several times to obtain compound 40, which was used in the next reaction without purification (114 mg, 99%).

<실시예 1 내지 4> 화합물 23 내지 화합물 26의 제조<Examples 1 to 4> Preparation of compounds 23 to 26

단계 1: 화합물 14 내지 18의 제조Step 1: Preparation of compounds 14 to 18

Figure pat00029
Figure pat00029

N-cyclopropyl-4'-(4-hydroxybenzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (14)의 제조Preparation of N-cyclopropyl-4'-(4-hydroxybenzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (14)

합성한 화합물8 (200mg, 0.722mmol)와 알려진 boronic ester 13 (283mg, 1.083mmol)와 K2CO3 (200mg, 1.444mmol)와 Pd(pph3)4 (75mg, 0.065mmol)을 Ar gas 충진후 anhydrous DMF 6mL에 녹였다. 90℃에서 overnight 반응을 진행했다. TLC로 출발 물질이 없어진 것이 확인되면 2N-HCl로 반응을 중지시킨 뒤 celite로 여과하고 EtOAc용매로 희석시켰다. 유기층은 EtOAc과 H2O로 씻어 추출했다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거했다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 14를 얻어냈다(135mg, 50%).After the synthesis of compound 8 (200mg, 0.722mmol) and known boronic ester 13 (283mg, 1.083mmol) and K 2 CO 3 (200mg, 1.444mmol) and Pd(pph 3 ) 4 (75mg, 0.065mmol) were charged with Ar gas It was dissolved in 6 mL of anhydrous DMF. The reaction was carried out overnight at 90°C. When it was confirmed by TLC that the starting material disappeared, the reaction was stopped with 2N-HCl, filtered through celite, and diluted with EtOAc solvent. The organic layer was extracted by washing with EtOAc and H 2 O. The extracted organic layer was dried over MgSO 4 and the solvent was removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 14 (135 mg, 50%).

1H-NMR (400 MHz, DMSO-d6) δ 10.46 (s, 1H), 8.44 (s, 1H), 7.77-7.71 (m, 6H ), 7.56-7.54 (m, 2H), 7.42 (m, 1H), 6.93 (m, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 0.67 (m, 2H), 0.56 (m, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 10.46 (s, 1H), 8.44 (s, 1H), 7.77-7.71 (m, 6H ), 7.56-7.54 (m, 2H), 7.42 (m, 1H), 6.93 (m, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 0.67 (m, 2H), 0.56 (m, 2H).

N-cyclopropyl-4'-(4-(2-hydroxyethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (15)의 제조Preparation of N-cyclopropyl-4'-(4-(2-hydroxyethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (15)

합성한 화합물 9(223.4mg, 0.696mmol)와 알려진 boronic ester 13 (181.63mg, 0.696mmol)를 출발 물질로 하여 상기 유도체 14와 같은 방법을 통해 화합물 15를 얻어냈다(190mg, 66%).Using the synthesized compound 9 (223.4 mg, 0.696 mmol) and known boronic ester 13 (181.63 mg, 0.696 mmol) as starting materials, compound 15 was obtained through the same method as the derivative 14 (190 mg, 66%).

1H-NMR (400 MHz, CDCl3) δ 7.83 (d, J = 8.6 Hz, 2H), 7.76 (d, J = 8.0 Hz, 2H), 7.67 (d, J = 7.8 Hz, 1H ), 7.63 (s, 1H), 7.38 (d, J = 7.9 Hz, 2H), 7.31 (d, J = 7.9 Hz, 1H), 6.98 (d, J = 8.6 Hz, 2H), 6.60 (s, 1H), 4.16 (d, J = 4.4 Hz, 2H), 4.01(t, J = 4.1 Hz, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 0.83 (d, J = 5.9 Hz, 2H), 0.61 (s, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.83 (d, J = 8.6 Hz, 2H), 7.76 (d, J = 8.0 Hz, 2H), 7.67 (d, J = 7.8 Hz, 1H), 7.63 ( s, 1H), 7.38 (d, J = 7.9 Hz, 2H), 7.31 (d, J = 7.9 Hz, 1H), 6.98 (d, J = 8.6 Hz, 2H), 6.60 (s, 1H), 4.16 ( d, J = 4.4 Hz, 2H), 4.01 (t, J = 4.1 Hz, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 0.83 (d, J = 5.9 Hz, 2H), 0.61 ( s, 2H).

N-cyclopropyl-4'-(4-(2-(2-hydroxyethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (16)의 제조Preparation of N-cyclopropyl-4'-(4-(2-(2-hydroxyethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (16)

합성한 화합물 10 (364.2mg, 0.997mmol)와 알려진 boronic ester 13 (260mg, 0.997mmol)를 출발 물질로 하여 상기 유도체 14와 같은 방법을 통해 화합물 16를 얻어냈다(300mg, 66%).Using the synthesized compound 10 (364.2 mg, 0.997 mmol) and the known boronic ester 13 (260 mg, 0.997 mmol) as starting materials, compound 16 was obtained through the same method as the derivative 14 (300 mg, 66%).

1H-NMR (400 MHz, CDCl3) δ 7.86 (d, J = 8.8 Hz, 2H), 7.81 (d, J = 8.2 Hz, 2H), 7.67 (dd, J = 7.9,1.8 Hz, 1H), 7.62 (d, J = 1.7 Hz, 1H), 7.41 (d, J = 8.2 Hz, 2H), 7.34 (d, J = 7.9 Hz, 1H), 7.01 (d, J = 8.8 Hz, 2H), 6.35 (s, 1H), 4.24 (t, J = 4.6 Hz, 2H), 4.24(t, J = 4.6 Hz, 2H), 3.92 (t, J = 4.6 Hz, 2H) 3.79 (t, J = 4.5 Hz, 2H), 2.90 (m, 1H), 2.32 (s, 3H), 0.86 (d, J = 5.6 Hz, 2H), 0.62 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.86 (d, J = 8.8 Hz, 2H), 7.81 (d, J = 8.2 Hz, 2H), 7.67 (dd, J = 7.9,1.8 Hz, 1H), 7.62 (d, J = 1.7 Hz, 1H), 7.41 (d, J = 8.2 Hz, 2H), 7.34 (d, J = 7.9 Hz, 1H), 7.01 (d, J = 8.8 Hz, 2H), 6.35 ( s, 1H), 4.24 (t, J = 4.6 Hz, 2H), 4.24 (t, J = 4.6 Hz, 2H), 3.92 (t, J = 4.6 Hz, 2H) 3.79 (t, J = 4.5 Hz, 2H) ), 2.90 (m, 1H), 2.32 (s, 3H), 0.86 (d, J = 5.6 Hz, 2H), 0.62 (m, 2H).

N-cyclopropyl-4'-(4-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (17)의 제조Preparation of N-cyclopropyl-4'-(4-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (17)

합성한 화합물 11 (457mg, 1.106mmol)와 알려진 boronic ester 13 (288.82mg, 1.106mmol)를 출발 물질로 하여 상기 유도체 14와 같은 방법을 통해 화합물 17을 얻어냈다(271.5mg, 49%).Using the synthesized compound 11 (457 mg, 1.106 mmol) and the known boronic ester 13 (288.82 mg, 1.106 mmol) as starting materials, compound 17 was obtained through the same method as the derivative 14 (271.5 mg, 49%).

1H-NMR (400 MHz, CDCl3) δ 7.11 (dd, J = 8.7 Hz, 4H), 7.67 (d, J = 7.8 Hz, 1H), 7.62 (s, 1H), 7.38 (d, J = 7.9 Hz, 2H), 7.31 (d, J = 7.8 Hz, 1H), 6.99 (d, J = 8.6 Hz, 2H), 6.54 (s, 1H), 4.21 (t, J = 4.4 Hz, 2H), 3.90(t, J = 4.3 Hz, 2H), 3.71 (t, J = 6.1 Hz, 6H), 3.61 (t, J = 4.4 Hz, 2H), 2.87 (m, 1H), 2.30 (s, 3H), 0.84 (m, 2H), 0.57 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.11 (dd, J = 8.7 Hz, 4H), 7.67 (d, J = 7.8 Hz, 1H), 7.62 (s, 1H), 7.38 (d, J = 7.9) Hz, 2H), 7.31 (d, J = 7.8 Hz, 1H), 6.99 (d, J = 8.6 Hz, 2H), 6.54 (s, 1H), 4.21 (t, J = 4.4 Hz, 2H), 3.90 ( t, J = 4.3 Hz, 2H), 3.71 (t, J = 6.1 Hz, 6H), 3.61 (t, J = 4.4 Hz, 2H), 2.87 (m, 1H), 2.30 (s, 3H), 0.84 ( m, 2H), 0.57 (m, 2H).

N-cyclopropyl-4'-(4-((5-hydroxypentyl)oxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (18)의 제조Preparation of N-cyclopropyl-4'-(4-((5-hydroxypentyl)oxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (18)

합성한 화합물 12 (63.2mg, 0.174mmol)와 알려진 boronic ester 13 (45.43mg, 0.174mmol)를 출발 물질로 하여 상기 유도체 14와 같은 방법을 통해 화합물 18를 얻어냈다(300mg, 66%).Using the synthesized compound 12 (63.2 mg, 0.174 mmol) and known boronic ester 13 (45.43 mg, 0.174 mmol) as starting materials, compound 18 was obtained in the same manner as in the derivative 14 (300 mg, 66%).

1H-NMR (400 MHz, CDCl3) δ 7.80 (dd, J = 21.1,8.3 Hz, 3H), 7.70-7.60 (m, 3H), 7.53 (m, 1H), 7.44 (td, J = 7.4,2.5 Hz, 1H), 7.38 (d, J = 8 Hz, 1H), 7.30 (d, J = 7.9 Hz, 1H), 6.95 (d, J = 8.6 Hz, 1H), 6.65 (s, 1H), 4.04 (t, J = 6.3 Hz, 2H), 3.67 (t, J = 6.3 Hz, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 1.89-7.79 (m, 3H), 1.69-1.61 (m, 3H), 1.60-1.50 (m, 3H), 0.82 (d, J = 5.7 Hz, 2H), 0.63-0.58 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.80 (dd, J = 21.1,8.3 Hz, 3H), 7.70-7.60 (m, 3H), 7.53 (m, 1H), 7.44 (td, J = 7.4, 2.5 Hz, 1H), 7.38 (d, J = 8 Hz, 1H), 7.30 (d, J = 7.9 Hz, 1H), 6.95 (d, J = 8.6 Hz, 1H), 6.65 (s, 1H), 4.04 (t, J = 6.3 Hz, 2H), 3.67 (t, J = 6.3 Hz, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 1.89-7.79 (m, 3H), 1.69-1.61 ( m, 3H), 1.60-1.50 (m, 3H), 0.82 (d, J = 5.7 Hz, 2H), 0.63-0.58 (m, 2H).

단계 2: 화합물 19 내지 22의 제조Step 2: Preparation of compounds 19 to 22

Figure pat00030
Figure pat00030

2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl (4-nitrophenyl) carbonate (19)의 제조Preparation of 2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl (4-nitrophenyl) carbonate (19)

둥근 바닥 플라스크에 4-nitro phenyl chloroformate(225.6mg, 1.12mmol)를 anhydrous CH2Cl2 3mL 녹인다. 그리고 vial에 합성한 화합물 15 (93mg, 0.224mmol)와 anhydrous pyridine(0.1mL, 1.12mmol)를 anhydrous CH2Cl2 2mL에 녹이고 이를 천천히 0℃에서 적가하고, 상온(25℃)에서 반응했다. TLC로 출발 물질이 없어진 것이 확인되면 2N-HCl로 반응을 중지시키고 EtOAc 용매로 희석시켰다. 유기층은 EtOAc과 H2O로 씻어 추출했다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거했다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 19를 얻어냈다(105mg, 81%). Dissolve 4-nitro phenyl chloroformate (225.6 mg, 1.12 mmol) in anhydrous CH 2 Cl 2 3 mL in a round bottom flask. And compound 15 (93mg, 0.224mmol) and anhydrous pyridine (0.1mL, 1.12mmol) synthesized in a vial were dissolved in anhydrous CH 2 Cl 2 2mL, and this was slowly added dropwise at 0℃, and reacted at room temperature (25℃). When it was confirmed by TLC that the starting material disappeared, the reaction was stopped with 2N-HCl and diluted with EtOAc solvent. The organic layer was extracted by washing with EtOAc and H 2 O. The extracted organic layer was dried over MgSO 4 and the solvent was removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 19 (105 mg, 81%).

1H-NMR (400 MHz, CDCl3) δ 8.27 (d, J = 9.0 Hz, 2H), 7.82 (dd, J = 31.5, 8.3 Hz, 4H), 7.66 (t, J = 7.3 Hz, 2H), 7.39 (d, J = 8.6 Hz, 4H ), 7.31 (d, J = 7.8 Hz, 1H), 7.02 (d, J = 8.6 Hz, 2H), 6.59 (s, 1H), 4.68 (d, J = 3.1 Hz, 2H), 4.37(d, J = 3.4 Hz, 2H), 2.88 (m, 1H), 2.30 (s, 3H), 0.83 (d, J = 6.7 Hz, 2H), 0.61 (s, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 8.27 (d, J = 9.0 Hz, 2H), 7.82 (dd, J = 31.5, 8.3 Hz, 4H), 7.66 (t, J = 7.3 Hz, 2H), 7.39 (d, J = 8.6 Hz, 4H ), 7.31 (d, J = 7.8 Hz, 1H), 7.02 (d, J = 8.6 Hz, 2H), 6.59 (s, 1H), 4.68 (d, J = 3.1) Hz, 2H), 4.37 (d, J = 3.4 Hz, 2H), 2.88 (m, 1H), 2.30 (s, 3H), 0.83 (d, J = 6.7 Hz, 2H), 0.61 (s, 2H).

2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl (4-nitrophenyl) carbonate (20)의 제조Preparation of 2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl (4-nitrophenyl) carbonate (20)

합성한 화합물 16(101mg, 0.22mmol)을 출발 물질로 하여 상기 유도체 19와 같은 방법을 통해 최종 화합물 20를 얻어냈다(123mg, 89%). Using the synthesized compound 16 (101 mg, 0.22 mmol) as a starting material, the final compound 20 was obtained in the same manner as in the derivative 19 (123 mg, 89%).

1H-NMR (400 MHz, CDCl3) δ 8.26 (d, J = 9.0 Hz, 2H), 7.84 (dd, J = 20.6, 8.2 Hz, 4H), 7.67 (d, J = 7.7 Hz, 1H), 7.62 (s, 1H), 7.42 (d, J = 7.8 Hz, 2H), 7.36 (t, J = 8.8 Hz, 3H), 7.02 (d, J = 8.6 Hz, 2H), 6.27 (s, 1H), 4.48 (d, J = 4.2 Hz, 2H), 4.26(d, J = 4.0 Hz, 2H), 3.93 (dd, J = 17.8,3.9 Hz, 4H), 2.90 (m, 1H), 2.32 (s, 3H), 0.87 (d, J = 6.4 Hz, 2H), 0.62 (s, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 8.26 (d, J = 9.0 Hz, 2H), 7.84 (dd, J = 20.6, 8.2 Hz, 4H), 7.67 (d, J = 7.7 Hz, 1H), 7.62 (s, 1H), 7.42 (d, J = 7.8 Hz, 2H), 7.36 (t, J = 8.8 Hz, 3H), 7.02 (d, J = 8.6 Hz, 2H), 6.27 (s, 1H), 4.48 (d, J = 4.2 Hz, 2H), 4.26 (d, J = 4.0 Hz, 2H), 3.93 (dd, J = 17.8,3.9 Hz, 4H), 2.90 (m, 1H), 2.32 (s, 3H) ), 0.87 (d, J = 6.4 Hz, 2H), 0.62 (s, 2H).

2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethoxy)ethyl (4-nitrophenyl) carbonate (21)의 제조2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethoxy)ethyl (4-nitrophenyl) carbonate Preparation of (21)

합성한 화합물 17 (150.5mg, 0.299mmol)을 출발 물질로 하여 상기 유도체 19 와 같은 방법을 통해 최종 화합물 21를 얻어냈다(131.8mg, 66%). Using the synthesized compound 17 (150.5 mg, 0.299 mmol) as a starting material, the final compound 21 was obtained in the same manner as in the derivative 19 (131.8 mg, 66%).

1H-NMR (400 MHz, CDCl3) δ 8.24 (d, J = 9.1 Hz, 2H), 7.81(dd, J = 20.4, 8.4 Hz, 4H), 7.67 (dd, J = 7.8, 1.6 Hz, 1H), 7.63 (d, J = 1.4 Hz, 1H), 7.42-7.30 (m, 4H), 6.98 (d, J = 8.8 Hz, 1H), 6.49 (s, 1H), 4.46-4.41 (m, 2H), 4.22 (t, J = 4.6 Hz, 2H), 3.91 (t, J = 4.6 Hz, 2H), 3.82 (quint, J = 2.3 Hz, 2H), 3.79-3.70 (m, 4H), 2.89 (m, 1H), 0.84 (dd, J = 12.6, 6.8 Hz, 2H), 0.63-0.58 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 8.24 (d, J = 9.1 Hz, 2H), 7.81 (dd, J = 20.4, 8.4 Hz, 4H), 7.67 (dd, J = 7.8, 1.6 Hz, 1H) ), 7.63 (d, J = 1.4 Hz, 1H), 7.42-7.30 (m, 4H), 6.98 (d, J = 8.8 Hz, 1H), 6.49 (s, 1H), 4.46-4.41 (m, 2H) , 4.22 (t, J = 4.6 Hz, 2H), 3.91 (t, J = 4.6 Hz, 2H), 3.82 (quint, J = 2.3 Hz, 2H), 3.79-3.70 (m, 4H), 2.89 (m, 1H), 0.84 (dd, J = 12.6, 6.8 Hz, 2H), 0.63-0.58 (m, 2H).

5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl (4-nitrophenyl) carbonate (22)의 제조Preparation of 5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl (4-nitrophenyl) carbonate (22)

합성한 화합물 18 (61.2mg, 0.13mmol)을 출발 물질로 하여 상기 유도체 19와 같은 방법을 통해 최종 화합물 22 를 얻어냈다(77.3mg, 93%). Using the synthesized compound 18 (61.2 mg, 0.13 mmol) as a starting material, the final compound 22 was obtained in the same manner as in the derivative 19 (77.3 mg, 93%).

1H-NMR (400 MHz, CDCl3) δ 8.25 (d, J = 8.8 Hz, 2H), 7.84 (d, J = 8.4 Hz, 2H), 7.78 (d, J = 7.7 Hz, 2H), 7.66 (t, J = 7.8 Hz, 2H), 7.37 (t, J = 7.3 Hz, 3H), 7.30 (d, J = 7.7 Hz, 1H), 6.96 (d, J = 8.4 Hz, 2H), 6.58 (s, 1H), 4.33 (t, J = 6.3 Hz, 2H), 4.08 (t, J = 5.9 Hz, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 1.93-1.81 (m, 4H), 1.69-1.61 (m, 2H), 0.82 (d, J = 6.4 Hz, 2H), 0.63-0.57 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 8.25 (d, J = 8.8 Hz, 2H), 7.84 (d, J = 8.4 Hz, 2H), 7.78 (d, J = 7.7 Hz, 2H), 7.66 ( t, J = 7.8 Hz, 2H), 7.37 (t, J = 7.3 Hz, 3H), 7.30 (d, J = 7.7 Hz, 1H), 6.96 (d, J = 8.4 Hz, 2H), 6.58 (s, 1H), 4.33 (t, J = 6.3 Hz, 2H), 4.08 (t, J = 5.9 Hz, 2H), 2.88 (m, 1H), 2.29 (s, 3H), 1.93-1.81 (m, 4H), 1.69-1.61 (m, 2H), 0.82 (d, J = 6.4 Hz, 2H), 0.63-0.57 (m, 2H).

단계 3: 최종 화합물 23 내지 26의 제조Step 3: Preparation of final compounds 23 to 26

Figure pat00031
Figure pat00031

2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (23)의 제조2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl(2-(2,6-dioxopiperidin-3-yl)-1 Preparation of ,3-dioxoisoindolin-4-yl)carbamate (23)

합성한 화합물 19 (50mg, 0.086mmol)와 합성한 화합물 5 (23.53mg, 0.086mmol)에 Ar gas를 충진후 anhydrous DMF 3mL로 녹였다. 이후 NaH(3.1mg, 0.13mmol)를 0℃에서 천천히 적가하고 상온(25℃)에서 반응했다. TLC로 출발 물질이 없어진 것이 확인되면 aq. sat. NH4Cl을 이용하여 반응을 중지시킨 뒤 EtOAc 용매로 희석시켰다. 유기층은 EtOAc과 H2O로 씻어 추출했다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거했다. Column chromatography를 통해 반응 혼합물을 정제하여 최종 화합물 23 를 얻어냈다(24.4mg, 40%).The synthesized compound 19 (50mg, 0.086mmol) and the synthesized compound 5 (23.53mg, 0.086mmol) were filled with Ar gas and dissolved with 3mL of anhydrous DMF. Then, NaH (3.1mg, 0.13mmol) was slowly added dropwise at 0 ℃ and reacted at room temperature (25 ℃). When it was confirmed by TLC that the starting material was gone, aq. sat. The reaction was stopped using NH 4 Cl and then diluted with EtOAc solvent. The organic layer was extracted by washing with EtOAc and H 2 O. The extracted organic layer was dried over MgSO 4 and the solvent was removed using an evaporator. The reaction mixture was purified through column chromatography to obtain the final compound 23 (24.4 mg, 40%).

1H-NMR (400 MHz, DMSO-d6) δ 11.16 (s, 1H), 9.22 (s, 1H), 8.46 (s, 1H), 8.24 (d, J = 8.3 Hz, 1H), 7.86 (t, J = 7.8 Hz, 1H), 7.78 (q, J = 7.9 Hz, 5H), 7.60 (d, J = 7.2 Hz, 1H), 7.57 (d, J = 7.9 Hz, 2H), 7.42 (d, J = 7.8 Hz, 1H), 7.18 (d, J = 8.6 Hz, 2H), 5.13 (dd, J = 12.5, 5.2 Hz, 1H), 4.54 (s, 1H), 4.40 (s, 1H), 2.86 (m, 2H), 2.58 (d, J = 17.8 Hz, 2H), 2.31 (s, 3H), 2.04 (m, 1H), 1.99 (s, 1H), 0.68 (d, J = 6.7 Hz, 2H), 0.56 (s, 2H); 13C-NMR (500 MHz, CDCL3) δ 195.3, 170.8, 168.9, 168.6, 167.9, 162.1, 152.8, 145.0, 141.2, 139.3, 137.8, 137.0, 136.6, 132.7(2C), 132.2, 131.5, 130.9, 130.8, 130.0(2C), 129.2(2C), 128.2, 126.3, 123.9, 118.1, 115.1, 114.4(2C), 66.2, 64.1 49.4, 31.5, 29.8, 23.3, 22.7, 20.6, 6.9 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.16 (s, 1H), 9.22 (s, 1H), 8.46 (s, 1H), 8.24 (d, J = 8.3 Hz, 1H), 7.86 (t) , J = 7.8 Hz, 1H), 7.78 (q, J = 7.9 Hz, 5H), 7.60 (d, J = 7.2 Hz, 1H), 7.57 (d, J = 7.9 Hz, 2H), 7.42 (d, J) = 7.8 Hz, 1H), 7.18 (d, J = 8.6 Hz, 2H), 5.13 (dd, J = 12.5, 5.2 Hz, 1H), 4.54 (s, 1H), 4.40 (s, 1H), 2.86 (m) , 2H), 2.58 (d, J = 17.8 Hz, 2H), 2.31 (s, 3H), 2.04 (m, 1H), 1.99 (s, 1H), 0.68 (d, J = 6.7 Hz, 2H), 0.56 (s, 2H); 13 C-NMR (500 MHz, CDCL3) δ 195.3, 170.8, 168.9, 168.6, 167.9, 162.1, 152.8, 145.0, 141.2, 139.3, 137.8, 137.0, 136.6, 132.7(2C), 132.2, 131.5, 130.9, 130.8, 130.0(2C), 129.2(2C), 128.2, 126.3, 123.9, 118.1, 115.1, 114.4(2C), 66.2, 64.1 49.4, 31.5, 29.8, 23.3, 22.7, 20.6, 6.9

2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (24)의 제조2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl(2-(2,6-dioxopiperidin-3 Preparation of -yl)-1,3-dioxoisoindolin-4-yl)carbamate (24)

합성한 화합물 20 (70mg, 0.112mmol)을 출발 물질로 하여 상기 유도체 23와 같은 방법을 통해 최종 화합물 24 를 얻어냈다(25.8mg, 30.4%).Using the synthesized compound 20 (70 mg, 0.112 mmol) as a starting material, the final compound 24 was obtained in the same manner as in the derivative 23 (25.8 mg, 30.4%).

1H-NMR (400 MHz, DMSO-d6) δ 11.12 (s, 1H), 9.04 (s, 1H), 8.41 (s, 1H), 8.20 (d, J = 8.3 Hz, 1H), 7.71 (d, J = 10.3 Hz, 6H), 7.51 (d, J = 7.8 Hz, 3H), 7.37 (d, J = 7.7 Hz, 1H), 7.06 (d, J = 8.5 Hz, 2H), 5.08 (m, 1H), 4.22 (d, J = 33.8 Hz, 4H), 3.76 (d, J = 27 Hz, 4H), 2.86-2.74 (m, 2H), 2.54 (d, J = 19.5 Hz, 1H), 2.26 (s, 3H), 2.03-1.91 (m, 2H), 0.63 (d, J = 6.9 Hz, 2H), 0.51 (d, J = 1.3 Hz, 2H); 13C-NMR (500 MHz, CDCL3) δ 195.2, 170.9, 168.8, 168.6, 168.0, 166.7, 162.6, 153.1, 144.9, 141.2, 138.3, 137.9, 137.1, 136.5, 132.7(2C), 132.3, 131.4, 130.9, 130.3(2C), 129.9(2C), 129.1, 128.2, 126.3, 123.8, 118.0, 115.1, 114.4(2C), 69.8(2C), 67.5, 65.0, 49.3, 31.5, 29.7, 23.5, 22.3, 20.8, 6.6. 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.12 (s, 1H), 9.04 (s, 1H), 8.41 (s, 1H), 8.20 (d, J = 8.3 Hz, 1H), 7.71 (d , J = 10.3 Hz, 6H), 7.51 (d, J = 7.8 Hz, 3H), 7.37 (d, J = 7.7 Hz, 1H), 7.06 (d, J = 8.5 Hz, 2H), 5.08 (m, 1H) ), 4.22 (d, J = 33.8 Hz, 4H), 3.76 (d, J = 27 Hz, 4H), 2.86-2.74 (m, 2H), 2.54 (d, J = 19.5 Hz, 1H), 2.26 (s) , 3H), 2.03-1.91 (m, 2H), 0.63 (d, J = 6.9 Hz, 2H), 0.51 (d, J = 1.3 Hz, 2H); 13 C-NMR (500 MHz, CDCL 3 ) δ 195.2, 170.9, 168.8, 168.6, 168.0, 166.7, 162.6, 153.1, 144.9, 141.2, 138.3, 137.9, 137.1, 136.5, 132.7(2C), 132.3, 131.4, 130.9 , 130.3(2C), 129.9(2C), 129.1, 128.2, 126.3, 123.8, 118.0, 115.1, 114.4(2C), 69.8(2C), 67.5, 65.0, 49.3, 31.5, 29.7, 23.5, 22.3, 20.8, 6.6 .

2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethoxy)ethyl(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (25)의 제조2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethoxy)ethyl(2-(2, Preparation of 6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (25)

합성한 화합물 21 (74.1mg, 0.11mmol)을 출발 물질로 하여 상기 유도체 23와 같은 방법을 통해 최종 화합물 25 를 얻어냈다(48.6mg, 54.6%). Using the synthesized compound 21 (74.1 mg, 0.11 mmol) as a starting material, the final compound 25 was obtained in the same manner as in the derivative 23 (48.6 mg, 54.6%).

1H-NMR (400 MHz, DMSO-d6) δ 11.12 (s, 1H), 9.04 (s, 1H), 8.40 (s, 1H), 8.20 (d, J = 8.3 Hz, 1H), 7.80-7.64 (m, 6H), 7.51 (d, J = 7.2 Hz, 3H), 7.37 (d, J = 7.5 Hz, 1H), 7.04 (d, J = 7.5 Hz, 2H), 5.07 (m, 1H), 4.18 (d, J = 32.2 Hz, 4H), 3.69 (d, J = 41 Hz, 4H), 3.57 (s, 4H), 2.88-2.73 (m, 2H), 2.53 (d, J = 19.4 Hz, 1H), 2.26 (s, 3H), 2.07-1.87(m, 2H), 0.63 (d, J = 6.3 Hz, 2H), 0.50 (s, 2H); 13C-NMR (500 MHz, CDCL3) δ 195.3, 170.8, 168.8, 168.6, 168.0, 166.8, 162.6, 153.0, 141.2, 137.9, 137.0, 136.5, 132.6(2C), 132.2, 130.8(2C), 129.9(2C), 129.1, 128.2, 126.4, 123.8, 117.9, 115.0, 114.3(2C), 70.9, 70.6, 69.7, 69.2, 67.7, 65.1, 60.5, 49.3, 31.5, 29.6, 23.3, 22.7, 20.6, 6.8(2C). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.12 (s, 1H), 9.04 (s, 1H), 8.40 (s, 1H), 8.20 (d, J = 8.3 Hz, 1H), 7.80-7.64 (m, 6H), 7.51 (d, J = 7.2 Hz, 3H), 7.37 (d, J = 7.5 Hz, 1H), 7.04 (d, J = 7.5 Hz, 2H), 5.07 (m, 1H), 4.18 (d, J = 32.2 Hz, 4H), 3.69 (d, J = 41 Hz, 4H), 3.57 (s, 4H), 2.88-2.73 (m, 2H), 2.53 (d, J = 19.4 Hz, 1H) , 2.26 (s, 3H), 2.07-1.87 (m, 2H), 0.63 (d, J = 6.3 Hz, 2H), 0.50 (s, 2H); 13 C-NMR (500 MHz, CDCL 3 ) δ 195.3, 170.8, 168.8, 168.6, 168.0, 166.8, 162.6, 153.0, 141.2, 137.9, 137.0, 136.5, 132.6(2C), 132.2, 130.8(2C), 129.9( 2C), 129.1, 128.2, 126.4, 123.8, 117.9, 115.0, 114.3 (2C), 70.9, 70.6, 69.7, 69.2, 67.7, 65.1, 60.5, 49.3, 31.5, 29.6, 23.3, 22.7, 20.6, 6.8 (2C) .

5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (26)의 제조5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl(2-(2,6-dioxopiperidin-3-yl)-1 Preparation of ,3-dioxoisoindolin-4-yl)carbamate (26)

합성한 화합물 22 (41.4mg, 0.066mmol)을 출발 물질로 하여 상기 유도체 23와 같은 방법을 통해 최종 화합물 26 를 얻어냈다(16mg, 32%).Using the synthesized compound 22 (41.4 mg, 0.066 mmol) as a starting material, the final compound 26 was obtained in the same manner as in the derivative 23 (16 mg, 32%).

1H-NMR (400 MHz, DMSO-d6) δ 11.15 (s, 1H), 9.04 (s, 1H), 8.44 (d, J = 3.8 Hz. 1H), 8.28 (d, J = 8.4 Hz, 1H), 7.87-7.73 (m, 7H), 7.57 (t, J = 8.1 Hz, 3H), 7.42 (d, J = 7.9 Hz, 1H), 7.11 (d, J = 8.7 Hz, 2H), 5.13 (dd, J = 12.7,5.4 Hz 1H), 4.20 (t, J = 5.7 Hz, 2H), 4.14-4.06 (m, 2H), 2.93-2.80 (m, 2H), 2.60 (m, 1H), 2.54 (s, 1H) , 2.31 (s, 3H), 2.04 (m, 1H), 1.86-1.68 (m, 4H), 1.59-1.49 (m, 2H), 0.71-0.65 (m, 2H), 0.59-0.53 (m, 2H); 13C-NMR (500 MHz, DMSO-d6) δ 194.0, 172.8, 169.7, 168.0, 166.5, 162.5, 152.8, 144.4, 140.0, 138.2, 136.8, 136.5, 136.4, 132.2(2C), 132.1, 131.5, 130.5, 129.4(2C), 129.3(2C), 129.2, 128.1, 126.7, 124.5, 117.7, 116.3, 114.2(2C), 79.2, 67.7, 65.4, 48.9, 30.9, 28.2, 27.9, 23.1, 21.9, 20.1, 5.7(2C). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.15 (s, 1H), 9.04 (s, 1H), 8.44 (d, J = 3.8 Hz. 1H), 8.28 (d, J = 8.4 Hz, 1H) ), 7.87-7.73 (m, 7H), 7.57 (t, J = 8.1 Hz, 3H), 7.42 (d, J = 7.9 Hz, 1H), 7.11 (d, J = 8.7 Hz, 2H), 5.13 (dd) , J = 12.7,5.4 Hz 1H), 4.20 (t, J = 5.7 Hz, 2H), 4.14-4.06 (m, 2H), 2.93-2.80 (m, 2H), 2.60 (m, 1H), 2.54 (s) , 1H) , 2.31 (s, 3H), 2.04 (m, 1H), 1.86-1.68 (m, 4H), 1.59-1.49 (m, 2H), 0.71-0.65 (m, 2H), 0.59-0.53 (m) , 2H); 13 C-NMR (500 MHz, DMSO-d 6 ) δ 194.0, 172.8, 169.7, 168.0, 166.5, 162.5, 152.8, 144.4, 140.0, 138.2, 136.8, 136.5, 136.4, 132.2(2C), 132.1, 131.5, 130.5 , 129.4(2C), 129.3(2C), 129.2, 128.1, 126.7, 124.5, 117.7, 116.3, 114.2(2C), 79.2, 67.7, 65.4, 48.9, 30.9, 28.2, 27.9, 23.1, 21.9, 20.1, 5.7( 2C).

<실시예 5 내지 8> 화합물 31 내지 34의 제조<Examples 5 to 8> Preparation of compounds 31 to 34

단계 1: 화합물 15 내지 18의 제조Step 1: Preparation of compounds 15 to 18

상기 실시예 1 내지 4와 단계 1은 동일하다.Step 1 and Examples 1 to 4 are the same.

단계 2: 화합물 27 내지 30의 제조Step 2: Preparation of compounds 27 to 30

Figure pat00032
Figure pat00032

2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl 4-methylbenzenesulfonate (27)의 제조Preparation of 2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl 4-methylbenzenesulfonate (27)

합성한 화합물 15 (50.1mg, 0.109mmol)를 CH2Cl2 3mL에 녹인 후 0℃에서 NEt3 0.05mL를 천천히 적가한다. 이후 상온(25℃)에서 p-TsCl(25mg, 0.131mmol)를 적가하여 반응한다. TLC로 출발 물질이 없어진 것이 확인되면 H2O로 반응을 중지시키고 EtOAc 용매로 희석시킨다. 유기층은 EtOAc과 H2O로 씻어 추출한다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거한다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 27를 얻어냈다(74.8mg, 99%).After dissolving the synthesized compound 15 (50.1 mg, 0.109 mmol) in 3 mL of CH 2 Cl 2 , 0.05 mL of NEt 3 is slowly added dropwise at 0°C. Thereafter, p-TsCl (25 mg, 0.131 mmol) is added dropwise at room temperature (25° C.) to react. When it is confirmed by TLC that the starting material is gone , the reaction is stopped with H 2 O and diluted with EtOAc solvent. The organic layer was washed with EtOAc and H 2 O and extracted. The extracted organic layer is dried with MgSO 4 and the solvent is removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 27 (74.8 mg, 99%).

1H-NMR (400 MHz, CDCl3) δ 7.91-7.78 (m, 6H), 7.67 (dd, J = 7.9, 1.3 Hz, 1H), 7.63 (d, J = 1.4 Hz, 1H ), 7.42 (d, J = 8.1 Hz, 2H), 7.35 (t, J = 7.1 Hz, 3H), 7.01 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.8 Hz, 2H), 6.29 (s, 1H), 4.51-4.37 (m, 2H), 4.30-4.22 (m, 2H), 2.91 (m, 1H), 2.46 (s, 3H), 2.32 (s, 3H), 0.87 (d, J = 6.3 Hz, 2H), 0.65-0.58 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.91-7.78 (m, 6H), 7.67 (dd, J = 7.9, 1.3 Hz, 1H), 7.63 (d, J = 1.4 Hz, 1H), 7.42 (d , J = 8.1 Hz, 2H), 7.35 (t, J = 7.1 Hz, 3H), 7.01 (d, J = 8.8 Hz, 1H), 6.88 (d, J = 8.8 Hz, 2H), 6.29 (s, 1H) ), 4.51-4.37 (m, 2H), 4.30-4.22 (m, 2H), 2.91 (m, 1H), 2.46 (s, 3H), 2.32 (s, 3H), 0.87 (d, J = 6.3 Hz, 2H), 0.65-0.58 (m, 2H).

2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl 4-methylbenzenesulfonate (28)의 제조Preparation of 2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl 4-methylbenzenesulfonate (28)

합성한 화합물 16 (50.1mg, 0.11mmol)을 출발 물질로 하여 상기 유도체 27와 같은 방법을 통해 최종 화합물 28 를 얻어냈다(47.4mg, 71%). Using the synthesized compound 16 (50.1 mg, 0.11 mmol) as a starting material, the final compound 28 was obtained in the same manner as in the derivative 27 (47.4 mg, 71%).

1H-NMR (400 MHz, CDCl3) δ 7.89-7.75 (m, 6H), 7.67 (d, J = 8.0 Hz, 1H), 7.62 (s, 1H), 7.42 (d, J = 7.8 Hz, 2H), 7.38-7.29 (m, 3H), 6.99 (d, J = 8.6 Hz, 2H), 6.27 (s, 1H), 4.17 (dd, J = 19.4, 4.1 Hz, 4H), 3.87-3.75 (m, 4H), 2.90 (m, 1H), 2.42 (s, 3H), 2.32 (s, 3H), 0.87 (d, J = 6.2 Hz, 2H), 0.62 (s, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.89-7.75 (m, 6H), 7.67 (d, J = 8.0 Hz, 1H), 7.62 (s, 1H), 7.42 (d, J = 7.8 Hz, 2H) ), 7.38-7.29 (m, 3H), 6.99 (d, J = 8.6 Hz, 2H), 6.27 (s, 1H), 4.17 (dd, J = 19.4, 4.1 Hz, 4H), 3.87-3.75 (m, 4H), 2.90 (m, 1H), 2.42 (s, 3H), 2.32 (s, 3H), 0.87 (d, J = 6.2 Hz, 2H), 0.62 (s, 2H).

2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (29)의 제조2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (29) manufacture of

합성한 화합물 17 (49.9mg, 0.099mmol)을 출발 물질로 하여 상기 유도체 27와 같은 방법을 통해 최종 화합물 29를 얻어냈다(48mg, 74%). Using the synthesized compound 17 (49.9 mg, 0.099 mmol) as a starting material, the final compound 29 was obtained in the same manner as in the derivative 27 (48 mg, 74%).

1H-NMR (400 MHz, CDCl3) δ 7.83 (d, J = 8.6 Hz, 2H), 7.77 (dd, J = 8.1,2.1 Hz, 4H), 7.68 (d, J = 7.9 Hz, 1H ), 7.63 (s, 1H), 7.38 (d, J = 7.9 Hz, 2H), 7.31 (d, J = 7.9 Hz, 3H), 6.98 (d, J = 8.6 Hz, 2H), 6.58 (s, 1H), 4.19 (t, J = 4.3 Hz, 2H), 4.14 (t, J = 4.7 Hz, 2H), 3.85 (t, J = 4.3 Hz, 2H), 3.71-3.64 (m, 4H), 3.63-3.59 (m, 2H), 2.87 (m, 1H), 2.41 (s, 3H), 2.29 (s, 3H), 0.82 (d, J = 6.0 Hz, 2H), 0.63-0.57 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.83 (d, J = 8.6 Hz, 2H), 7.77 (dd, J = 8.1,2.1 Hz, 4H), 7.68 (d, J = 7.9 Hz, 1H), 7.63 (s, 1H), 7.38 (d, J = 7.9 Hz, 2H), 7.31 (d, J = 7.9 Hz, 3H), 6.98 (d, J = 8.6 Hz, 2H), 6.58 (s, 1H), 4.19 (t, J = 4.3 Hz, 2H), 4.14 (t, J = 4.7 Hz, 2H), 3.85 (t, J = 4.3 Hz, 2H), 3.71-3.64 (m, 4H), 3.63-3.59 (m) , 2H), 2.87 (m, 1H), 2.41 (s, 3H), 2.29 (s, 3H), 0.82 (d, J = 6.0 Hz, 2H), 0.63-0.57 (m, 2H).

5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl 4-methylbenzenesulfonate (30)의 제조Preparation of 5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl 4-methylbenzenesulfonate (30)

합성한 화합물 18 (46.4mg, 0.101mmol)을 출발 물질로 하여 상기 유도체 27와 같은 방법을 통해 최종 화합물 30 를 얻어냈다(25.7mg, 59%).Using the synthesized compound 18 (46.4 mg, 0.101 mmol) as a starting material, the final compound 30 was obtained in the same manner as in the derivative 27 (25.7 mg, 59%).

1H-NMR (400 MHz, CDCl3) δ 7.83 (d, J = 8.7 Hz, 2H), 7.78 (q, J = 3.8 Hz, 4H), 7.67 (dd, J = 7.9, 1.6 Hz, 1H), 7.63 (d, J = 1.5 Hz, 1H), 7.39 (d, J = 8.1 Hz, 2H), 7.32 (t, J = 7.0 Hz, 3H), 6.93 (d, J = 8.7 Hz, 2H), 6.49 (s, 1H), 4.06 (t, J = 6.3 Hz, 2H), 4.00 (t, J = 6.2 Hz, 2H), 2.88 (m, 1H), 2.43 (s, 3H), 2.30 (s, 3H), 1.81-1.67 (m, 4H), 1.57-1.46 (m, 2H), 0.87-0.80 (m, 2H), 0.64-0.58 (m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.83 (d, J = 8.7 Hz, 2H), 7.78 (q, J = 3.8 Hz, 4H), 7.67 (dd, J = 7.9, 1.6 Hz, 1H), 7.63 (d, J = 1.5 Hz, 1H), 7.39 (d, J = 8.1 Hz, 2H), 7.32 (t, J = 7.0 Hz, 3H), 6.93 (d, J = 8.7 Hz, 2H), 6.49 ( s, 1H), 4.06 (t, J = 6.3 Hz, 2H), 4.00 (t, J = 6.2 Hz, 2H), 2.88 (m, 1H), 2.43 (s, 3H), 2.30 (s, 3H), 1.81-1.67 (m, 4H), 1.57-1.46 (m, 2H), 0.87-0.80 (m, 2H), 0.64-0.58 (m, 2H).

단계 3: 최종 화합물 31 내지 34의 제조Step 3: Preparation of final compounds 31 to 34

Figure pat00033
Figure pat00033

N-cyclopropyl-4'-(4-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (31)의 제조N-cyclopropyl-4'-(4-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)ethoxy)benzoyl)-6-methyl- Preparation of [1,1'-biphenyl]-3-carboxamide (31)

합성한 화합물 27 (63.4 mg, 0.11 mmol)와 합성한 화합물 6(15.3 mg, 0.05 mmol)와 K2CO3(23.1 mg, 0.16 mmol)을 Acetonitrile 3mL에 녹이고 reflux 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인하면 evaporator장치를 이용해 Acetonitrile를 제거한다. 이후 혼합물을 CH2Cl2 용매로 희석시킨다. 유기층은 CH2Cl2과 H2O로 씻어 추출한다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거한다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 31를 얻어냈다(10.3mg, 28%). Dissolve the synthesized compound 27 (63.4 mg, 0.11 mmol) and the synthesized compound 6 (15.3 mg, 0.05 mmol) and K 2 CO 3 (23.1 mg, 0.16 mmol) in 3 mL of Acetonitrile and proceed with a reflux reaction. When it is confirmed by TLC that the reaction does not proceed any further, the acetonitrile is removed using an evaporator device. The mixture is then diluted with CH 2 Cl 2 solvent. The organic layer was extracted by washing with CH 2 Cl 2 and H 2 O. The extracted organic layer is dried with MgSO 4 and the solvent is removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 31 (10.3 mg, 28%).

1H-NMR (400 MHz, DMSO-d6) δ 11.99(s, 1H), 10.25(s, 1H), 8.60(s, 1H), 7.99(d, J = 8.4 Hz, 1H), 7.78 (dd, J = 8.5, 1.6 Hz, 1H), 7.46 (s, 1H), 6.84 (dd, J = 6.4, 2 Hz, 4H), 6.20 (s, 1H), 5.32(m, 1H), 4.28 (s, 1H), 4.13 (t, J = 10.4 Hz, 2H), 3.90 (d, J = 6.3 Hz, 1H), 3.67 (d, J = 5.4 Hz, 1H), 2.67 (m, 1H), 2.05-1.93 (m, 3H) 1.40(s, 1H); 13C-NMR (500 MHz, CDCL3) δ 195.4, 170.9, 168.8, 168.6, 167.0, 165.7, 162.3, 162.1, 156.1, 145.1, 144.9, 141.2, 139.3, 137.1, 136.7, 132.7(2C), 132.2, 130.9(2C), 129.9(2C), 129.1(2C), 128.0, 126.5, 119.9, 117.8, 116.7, 114.4, 68.4, 66.8, 64.3, 31.8, 22.9, 20.4, 14.2, 6.6(2C). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.99(s, 1H), 10.25(s, 1H), 8.60(s, 1H), 7.99 (d, J = 8.4 Hz, 1H), 7.78 (dd, J = 8.5, 1.6 Hz, 1H), 7.46 (s, 1H), 6.84 (dd, J = 6.4, 2 Hz, 4H), 6.20 (s) , 1H), 5.32 (m, 1H), 4.28 (s, 1H), 4.13 (t, J = 10.4 Hz, 2H), 3.90 (d, J = 6.3 Hz, 1H), 3.67 (d, J = 5.4 Hz, 1H), 2.67 (m, 1H), 2.05-1.93 (m, 3H) 1.40 (s, 1H); 13 C-NMR (500 MHz, CDCL 3 ) δ 195.4, 170.9, 168.8, 168.6, 167.0, 165.7, 162.3, 162.1, 156.1, 145.1, 144.9, 141.2, 139.3, 137.1, 136.7, 132.7(2C), 132.2, 130.9 (2C), 129.9(2C), 129.1(2C), 128.0, 126.5, 119.9, 117.8, 116.7, 114.4, 68.4, 66.8, 64.3, 31.8, 22.9, 20.4, 14.2, 6.6(2C).

N-cyclopropyl-4'-(4-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (32)의 제조N-cyclopropyl-4'-(4-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)ethoxy)ethoxy)benzoyl) Preparation of -6-methyl-[1,1'-biphenyl]-3-carboxamide (32)

합성한 화합물 28 (47.4mg, 0.077mmol)을 출발 물질로 하여 상기 유도체 31과 같은 방법을 통해 최종 화합물 32 를 얻어냈다(16.9mg, 31%). Using the synthesized compound 28 (47.4 mg, 0.077 mmol) as a starting material, the final compound 32 was obtained in the same manner as in the derivative 31 (16.9 mg, 31%).

1H-NMR (400 MHz, DMSO-d6) δ 11.10 (s, 1H), 8.41 (s, 1H), 8.29 (s, 1H), 7.73 (dd, J = 7.5,2.1 Hz, 5H), 7.52 (t, J = 6.8 Hz, 3H), 7.44-7.34 (m, 2H), 7.58-7.50 (m, 2H), 7.07 (t, J = 8.4 Hz, 2H), 5.11(m, 1H), 4.35 (s, 1H), 4.17 (dd, J = 17.9, 1.1 Hz, 2H), 3.92-3.79 (m, 3H), 3.73 (s, 1H), 3.48 (m, 1H), 3.53-3.43 (m, 1H), 2.95 (m, 1H), 2.83-2.49 (m, 2H), 2.27 (s, 1H), 2.16-1.87 (m, 2H), 0.65 (d, J = 4.8 Hz, 2H), 0.53 (s, 2H); 13C-NMR (500 MHz, CDCL3) δ 195.5, 171.1, 168.7, 168.2, 167.2, 167.0, 165.8, 162.6, 156.4, 145.1, 141.2, 139.2, 137.1, 136.4, 133.8, 132.6(2C), 132.1, 130.9(2C), 130.3, 129.8(2C), 128.9(2C), 128.2, 119.4, 116.3, 114.1, 70.5, 69.8, 69.6, 68.8, 67.5, 29.7, 23.5, 20.6, 14.2, 6.8(2C). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.10 (s, 1H), 8.41 (s, 1H), 8.29 (s, 1H), 7.73 (dd, J = 7.5,2.1 Hz, 5H), 7.52 (t, J = 6.8 Hz, 3H), 7.44-7.34 (m, 2H), 7.58-7.50 (m, 2H), 7.07 (t, J = 8.4 Hz, 2H), 5.11 (m, 1H), 4.35 (s, 1H), 4.17 (dd, J = 17.9, 1.1 Hz, 2H), 3.92-3.79 (m, 3H), 3.73 (s, 1H), 3.48 (m, 1H), 3.53-3.43 (m, 1H), 2.95 (m, 1H), 2.83-2.49 (m, 2H), 2.27 (s, 1H), 2.16-1.87 (m, 2H), 0.65 (d, J = 4.8 Hz, 2H), 0.53 (s, 2H); 13 C-NMR (500 MHz, CDCL 3 ) δ 195.5, 171.1, 168.7, 168.2, 167.2, 167.0, 165.8, 162.6, 156.4, 145.1, 141.2, 139.2, 137.1, 136.4, 133.8, 132.6(2C), 132.1, 130.9 (2C), 130.3, 129.8 (2C), 128.9 (2C), 128.2, 119.4, 116.3, 114.1, 70.5, 69.8, 69.6, 68.8, 67.5, 29.7, 23.5, 20.6, 14.2, 6.8 (2C).

N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)ethoxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (33)의 제조N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)ethoxy)ethoxy Preparation of )ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (33)

합성한 화합물 29 (38.9mg, 0.059mmol)을 출발 물질로 하여 상기 유도체 31과 같은 방법을 통해 최종 화합물 33 를 얻어냈다(7.5mg, 34%). 1H-NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 7.76 (d, J = 9.4 Hz, 7H), 7.55 (d, J = 7.3 Hz, 3H), 7.41 (m, 1H), 7.15-7.06 (m, 2H), 5.17(m, 1H), 4.33 (s, 1H), 4.19 (s, 2H), 3.77 (s, 2H), 3.70-3.48 (m, 5H), 3.42 (m, 1H), 2.97 (m, 1H), 2.88-2.66 (m, 1H), 2.54 (s, 1H), 2.30 (s, 1H), 2.00 (m, 1H), 0.68 (d, J = 5.2 Hz, 2H), 0.56 (s, 2H); 13C-NMR (500 MHz, CDCL3) δ 195.5, 171.1, 168.4, 167.2, 165.8, 162.7, 156.4, 144.9, 141.3, 139.3, 136.9, 136.4, 133.9, 132.6(2C), 132.2, 130.9(2C), 129.8(2C), 129.0(2C), 128.2, 126.4, 119.6, 117.5, 116.2, 114.3(2C), 72.7, 70.8, 70.5, 69.6, 69.2, 67.8, 61.8, 29.8, 23.4, 21.9, 20.6, 6.9(2C).Using the synthesized compound 29 (38.9 mg, 0.059 mmol) as a starting material, the final compound 33 was obtained in the same manner as in the derivative 31 (7.5 mg, 34%). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 8.47 (s, 1H), 7.76 (d, J = 9.4 Hz, 7H), 7.55 (d, J = 7.3 Hz, 3H), 7.41 (m, 1H) ), 7.15-7.06 (m, 2H), 5.17 (m, 1H), 4.33 (s, 1H), 4.19 (s, 2H), 3.77 (s, 2H), 3.70-3.48 (m, 5H), 3.42 (m, 1H), 2.97 (m, 1H), 2.88-2.66 (m, 1H), 2.54 (s, 1H), 2.30 (s, 1H), 2.00 (m, 1H), 0.68 (d, J = 5.2 Hz, 2H), 0.56 (s, 2H); 13 C-NMR (500 MHz, CDCL 3 ) δ 195.5, 171.1, 168.4, 167.2, 165.8, 162.7, 156.4, 144.9, 141.3, 139.3, 136.9, 136.4, 133.9, 132.6(2C), 132.2, 130.9(2C), 129.8 (2C), 129.0 (2C), 128.2, 126.4, 119.6, 117.5, 116.2, 114.3 (2C), 72.7, 70.8, 70.5, 69.6, 69.2, 67.8, 61.8, 29.8, 23.4, 21.9, 20.6, 6.9 (2C) ).

N-cyclopropyl-4'-(4-((5-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)pentyl)oxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (34)의 제조N-cyclopropyl-4'-(4-((5-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)pentyl)oxy)benzoyl)-6 Preparation of -methyl-[1,1'-biphenyl]-3-carboxamide (34)

합성한 화합물 30 (25.7mg, 0.042mmol)을 출발 물질로 하여 상기 유도체 31과 같은 방법을 통해 최종 화합물 34 를 얻어냈다(4.1mg, 53%). Using the synthesized compound 30 (25.7 mg, 0.042 mmol) as a starting material, the final compound 34 was obtained in the same manner as in the derivative 31 (4.1 mg, 53%).

1H-NMR (400 MHz, DMSO-d6) δ 8.45 (m, 1H), 7.80-7.73 (m, 4H), 7.65-7.59 (m, 4H), 7.57-7.51 (m, 3H), 7.41 (d, J = 8.9 Hz, 1H), 7.10 (dd, J = 8.7, 2.8 Hz, 1H), 5.24 (m, 1H), 4.24 (t, J = 5.8 Hz, 1H), 4.14-4.00 (m, 2H), 3.67 (m, 1H), 2.85 (m, 1H), 2.68 (m, 1H), 2.31 (s, 3H), 2.19-1.96 (m, 3H), 1.96-1.84 (m, 2H), 1.75 (m, 1H), 1.63 (m, 1H), 1.56-1.40 (m, 4H), 0.70-0.64 (m, 2H), 0.59-0.53 (m, 2H); 13C-NMR (500 MHz, CDCL3) δ 195.3, 171.2, 168.8, 168.6, 167.2, 165.8, 162.9, 156.6, 144.7, 141.2, 139.4, 136.6, 134.0, 132.7(2C), 132.3, 130.8(2C), 129.9(2C), 129.0(2C), 128.6, 128.1, 126.3, 119.1, 117.3, 115.9, 114.1, 69.3, 68.2, 62.9, 32.4, 31.0, 29.8, 28.9, 23.2, 22.5, 20.5, 6.9(2C). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 8.45 (m, 1H), 7.80-7.73 (m, 4H), 7.65-7.59 (m, 4H), 7.57-7.51 (m, 3H), 7.41 ( d, J = 8.9 Hz, 1H), 7.10 (dd, J = 8.7, 2.8 Hz, 1H), 5.24 (m, 1H), 4.24 (t, J = 5.8 Hz, 1H), 4.14-4.00 (m, 2H), 3.67 (m, 1H), 2.85 (m, 1H), 2.68 (m, 1H), 2.31 (s, 3H) ), 2.19-1.96 (m, 3H), 1.96-1.84 (m, 2H), 1.75 (m, 1H), 1.63 (m, 1H), 1.56-1.40 (m, 4H), 0.70-0.64 (m, 2H) ), 0.59-0.53 (m, 2H); 13 C-NMR (500 MHz, CDCL 3 ) δ 195.3, 171.2, 168.8, 168.6, 167.2, 165.8, 162.9, 156.6, 144.7, 141.2, 139.4, 136.6, 134.0, 132.7(2C), 132.3, 130.8(2C), 129.9(2C), 129.0(2C), 128.6, 128.1, 126.3, 119.1, 117.3, 115.9, 114.1, 69.3, 68.2, 62.9, 32.4, 31.0, 29.8, 28.9, 23.2, 22.5, 20.5, 6.9(2C).

<실시예 9> 화합물 41의 제조<Example 9> Preparation of compound 41

단계 1: 화합물 37의 제조Step 1: Preparation of compound 37

Figure pat00034
Figure pat00034

tert-butyl (2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl)carbamate (37)의 제조Preparation of tert-butyl (2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl)carbamate (37)

합성한 화합물 14 (23 mg, 0.062 mmol)와 36 (35 mg, 0.124 mmol)와 K2CO3(26 mg, 0.186 mmol)을DMF 1mL에 녹이고 90 ℃에서 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인되면 NH4Cl을 이용하여 반응을 중지시킨다. 유기층은 EtOAc 와 H2O로 씻어 추출한다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거한다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 37 를 얻어냈다(14.4 mg, 42%).The synthesized compounds 14 (23 mg, 0.062 mmol) and 36 (35 mg, 0.124 mmol) and K 2 CO 3 (26 mg, 0.186 mmol) were dissolved in 1 mL of DMF and the reaction was carried out at 90 °C. When it is confirmed by TLC that the reaction does not proceed any further, the reaction is stopped using NH 4 Cl. The organic layer was washed with EtOAc and H 2 O and extracted. The extracted organic layer is dried with MgSO 4 and the solvent is removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 37 (14.4 mg, 42%).

1H-NMR (400 MHz, CDCl3) δ 7.88(d, J = 8.8 Hz, 2H), 7.83(d, J = 8.2 Hz, 2H), 7.69-7.61(m, 2H), 7.42(d, J = 8.8 Hz, 2H), 7.35(d, J = 8.4 Hz, 1H), 7.02(d, J = 9.0 Hz, 2H), 6.22(s, 1H), 4.95(s, 1H), 4.24-4.20(m, 2H), 3.89-3.85(m, 2H), 3.65-3.61(m, 2H), 3.40-3.32(m, 2H), 2.92(m, 1H), 2.33(s, 3H), 1.45(s, 9H), 0.91-0.85(m, 2H), 0.65-0.60(m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.88 (d, J = 8.8 Hz, 2H), 7.83 (d, J = 8.2 Hz, 2H), 7.69-7.61 (m, 2H), 7.42 (d, J) = 8.8 Hz, 2H), 7.35 (d, J = 8.4 Hz, 1H), 7.02 (d, J = 9.0 Hz, 2H), 6.22 (s, 1H), 4.95 (s, 1H), 4.24-4.20 (m) , 2H), 3.89-3.85(m, 2H), 3.65-3.61(m, 2H), 3.40-3.32(m, 2H), 2.92(m, 1H), 2.33(s, 3H), 1.45(s, 9H) ), 0.91-0.85 (m, 2H), 0.65-0.60 (m, 2H).

단계 2: 화합물 38의 제조Step 2: Preparation of compound 38

Figure pat00035
Figure pat00035

2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethan-1-aminium chloride (38)의 제조Preparation of 2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethan-1-aminium chloride (38)

합성한 화합물 37 (13.5 mg, 0.024 mmol)을 CH2Cl2 0.8 m 에 녹인 후 HCl 4M in Dioxane (0. 2ml)을 넣고 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인되면 용매를 날리고 ether로 여러 번 씻어서 화합물 38 을 얻어냈다(8.1 mg, 68%).After dissolving the synthesized compound 37 (13.5 mg, 0.024 mmol) in CH 2 Cl 2 0.8 m, HCl 4M in Dioxane (0.2 ml) is added and the reaction proceeds. When it was confirmed by TLC that the reaction did not proceed any further, the solvent was evaporated and washed several times with ether to obtain compound 38 (8.1 mg, 68%).

1H-NMR (400 MHz, DMSO-d6) δ 8.48(d, J = 0,3 Hz, 1H), 7.93(bs, 3H), 7.83-7.74(m, 6H), 7.57(d, J = 7.9 Hz, 2H), 7.42(d, J = 8.2 Hz, 1H), 7.14(d, J = 8.9 Hz, 2H), 4.29-4.34(m, 2H), 3.87-3.82(m, 2H), 3.04-2.98(m, 2H), 2.85(m, 1H), 2.32(s, 3H), 0.71-0.66(m, 2H), 0.59-0.54(m, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 8.48 (d, J = 0,3 Hz, 1H), 7.93 (bs, 3H), 7.83-7.74 (m, 6H), 7.57 (d, J = 7.9 Hz, 2H), 7.42 (d, J = 8.2 Hz, 1H), 7.14 (d, J = 8.9 Hz, 2H), 4.29-4.34 (m, 2H), 3.87-3.82 (m, 2H), 3.04- 2.98 (m, 2H), 2.85 (m, 1H), 2.32 (s, 3H), 0.71-0.66 (m, 2H), 0.59-0.54 (m, 2H).

단계 3: 최종 화합물 41의 제조Step 3: Preparation of final compound 41

Figure pat00036
Figure pat00036

(S)-N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy)acetamido)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (41)의 제조(S)-N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy Preparation of )acetamido)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1'-biphenyl]-3-carboxamide (41)

합성한 화합물 38 (9 mg, 0.018 mmol), 40 (7 mg, 0.015 mmol), HATU (6 mg, 0.015 mmol)을 DMF 3mL 녹인 후 DIPEA (0.045 mmol)를 넣고 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인하면 brine 넣고 Citiric acid로 pH 1로 만들어 준다. 다시 NaHCO3 중화하고 유기층은 CH2Cl2과 H2O로 씻어 추출한다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거한다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 41을 얻어냈다(12mg, 89%).The synthesized compound 38 (9 mg, 0.018 mmol), 40 (7 mg, 0.015 mmol), and HATU (6 mg, 0.015 mmol) were dissolved in 3 mL of DMF, and then DIPEA (0.045 mmol) was added thereto and the reaction proceeded. When it is confirmed by TLC that the reaction does not proceed any further, add brine and make pH 1 with citric acid. Again, NaHCO 3 is neutralized, and the organic layer is washed with CH 2 Cl 2 and H 2 O and extracted. The extracted organic layer is dried with MgSO 4 and the solvent is removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 41 (12 mg, 89%).

1H-NMR (400 MHz, DMSO-d6) δ 11.12(s, 1H), 8.44(d, J = 4.2 Hz, 1H), 8.03(t, J = 5.76 Hz, 1H), 7.81-7.73(m, 6H), 7.56(d, J = 8.4 Hz, 2H), 7.47(d, J = 7.3 Hz, 1H), 7.43-7.37(m, 2H), 7.10(d, J = 8.9 Hz, 2H), 5.14-5.08(m, 1H), 4.23-4.19(m, 2H), 3.82-3.77(m, 2H), 3.59-3.54(m, 2H), 3.39-3.35(m, 2H), 2.90-2.82(m, 2H), 2.68-2.54(m, 1H), 2.31(s, 3H), 2.07-1.97(m, 2H), 0.71-0.68(m, 2H), 0.58-0.53(m, 2H). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 11.12 (s, 1H), 8.44 (d, J = 4.2 Hz, 1H), 8.03 (t, J = 5.76 Hz, 1H), 7.81-7.73 (m) , 6H), 7.56 (d, J = 8.4 Hz, 2H), 7.47 (d, J = 7.3 Hz, 1H), 7.43-7.37 (m, 2H), 7.10 (d, J = 8.9 Hz, 2H), 5.14-5.08 (m, 1H), 4.23-4.19 (m, 2H), 3.82-3.77 (m, 2H), 3.59-3.54 (m, 2H), 3.39-3.35 (m, 2H), 2.90-2.82 (m) , 2H), 2.68-2.54 (m, 1H), 2.31 (s, 3H), 2.07-1.97 (m, 2H), 0.71-0.68 (m, 2H), 0.58-0.53 (m, 2H).

<비교예 1> 화합물 43의 제조<Comparative Example 1> Preparation of compound 43

Figure pat00037
Figure pat00037

합성한 화합물 38 (28 mg, 0.061 mmol), 상업적으로 구매 가능한 42 (15 mg, 0.061 mmol)를 DMF 0.4mL 녹였다. 0℃에서 EDC (0.073 mmol), DMAP(0.9mg, 0.007mmol)을 넣고 반응을 진행한다. TLC로 더 이상 반응이 진행되지 않음을 확인하면 solvent를 날리고 유기층은 CH2Cl2과 H2O로 씻어 추출한다. 추출한 유기층은 MgSO4로 건조시키고 evaporator를 이용해 용매를 제거한다. Column chromatography를 통해 반응 혼합물을 정제하여 화합물 43을 얻어냈다(17.2mg, 42%).The synthesized compound 38 (28 mg, 0.061 mmol) and commercially available 42 (15 mg, 0.061 mmol) were dissolved in 0.4 mL of DMF. EDC (0.073 mmol) and DMAP (0.9 mg, 0.007 mmol) were added at 0° C. and the reaction was carried out. When it is confirmed by TLC that the reaction does not proceed any further, blow off the solvent and wash the organic layer with CH 2 Cl 2 and H 2 O for extraction. The extracted organic layer is dried with MgSO 4 and the solvent is removed using an evaporator. The reaction mixture was purified through column chromatography to obtain compound 43 (17.2 mg, 42%).

1H-NMR (400 MHz, CDCl3) δ 7.87-7.80(m, 4H), 7.70-7.68(m, 2H), 7.42(d, J = 6.9 Hz, 2H), 7.42 7.34(d, J = 7.3 Hz, 1H), 7.01(d, J = 7.6 Hz, 2H), 6.50(s, 1H), 5.72(s, 1H), 5.17(s, 1H), 4.46(m, 1H), 4.25(d, J = 10.1 Hz, 5H), 3.91(m, 1H), 3.79(m, 1H), 3.12(s, 1H), 2.90(m, 1H), 2.87(m, 1H), 2.71(d, J = 12.6 Hz, 1H), 2.35(s, 2H), 2.32(s, 3H), 1.66(m, 4H), 1.42(s, 2H), 0.86-0.85(m, 2H), 0.62-0.61(m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.87-7.80 (m, 4H), 7.70-7.68 (m, 2H), 7.42 (d, J = 6.9 Hz, 2H), 7.42 7.34 (d, J = 7.3) Hz, 1H), 7.01(d, J = 7.6 Hz, 2H), 6.50(s, 1H), 5.72(s, 1H), 5.17(s, 1H), 4.46(m, 1H), 4.25(d, J) = 10.1 Hz, 5H), 3.91 (m, 1H), 3.79 (m, 1H), 3.12 (s, 1H), 2.90 (m, 1H), 2.87 (m, 1H), 2.71 (d, J = 12.6 Hz) , 1H), 2.35 (s, 2H), 2.32 (s, 3H), 1.66 (m, 4H), 1.42 (s, 2H), 0.86-0.85 (m, 2H), 0.62-0.61 (m, 2H).

<비교예 2> 화합물 45의 제조<Comparative Example 2> Preparation of compound 45

Figure pat00038
Figure pat00038

상업적으로 구매 가능한 44(33.6 mg, 0.184 mmol), 42 (38 mg, 0.221 mmol)를 출발 물질로 하여 상기 유도체 43과 같은 방법을 통해 최종 화합물 45 를 얻어냈다(33.8 mg, 45 %).Using commercially available 44 (33.6 mg, 0.184 mmol) and 42 (38 mg, 0.221 mmol) as starting materials, the final compound 45 was obtained in the same manner as in the derivative 43 (33.8 mg, 45%).

1H-NMR (400 MHz, CDCl3) δ 7.30-7.28(m, 2H), 6.98-6.88(m, 3H), 5.59(s, 1H), 5.02(s, 1H), 4.48(m, 1H), 4.31-4.21(m, 3H), 4.14-4.12(m, 2H), 3.88-3.83(m, 2H), 3.80-3.75(m, 2H), 3.12(m, 1H), 2.88(m, 1H), 2.72(m, 1H), 2.37(t, J = 7.3 Hz, 2H), 1.75-1.57(m, 4H), 1.48-1.38(m, 2H). 1 H-NMR (400 MHz, CDCl 3 ) δ 7.30-7.28(m, 2H), 6.98-6.88(m, 3H), 5.59(s, 1H), 5.02(s, 1H), 4.48(m, 1H) , 4.31-4.21(m, 3H), 4.14-4.12(m, 2H), 3.88-3.83(m, 2H), 3.80-3.75(m, 2H), 3.12(m, 1H), 2.88(m, 1H) , 2.72 (m, 1H), 2.37 (t, J = 7.3 Hz, 2H), 1.75-1.57 (m, 4H), 1.48-1.38 (m, 2H).

<비교예 3> 화합물 16의 제조<Comparative Example 3> Preparation of compound 16

Figure pat00039
Figure pat00039

상기 실시예 1의 단계 1과 동일하게 수행하여, 화합물 16을 제조하였다.In the same manner as in Step 1 of Example 1, compound 16 was prepared.

<실험예 1> 실시예 화합물의 p38 MAPK(mitogen-activated protein kinases) 제거능 평가<Experimental Example 1> Evaluation of p38 MAPK (mitogen-activated protein kinases) removal ability of the compound of Examples

p38 MAPK의 제거를 확인하기 위해 p38 MAPK 가 일정하게 생성되는 간세포 유래 암종 세포주인 Huh7 세포에 각 화합물을 처리 후 p38 MAPK의 감소를 확인하였다.In order to confirm the removal of p38 MAPK, the reduction of p38 MAPK was confirmed after treatment with each compound in Huh7 cells, a hepatocellular carcinoma cell line in which p38 MAPK is constantly generated.

구체적으로, Huh7 세포를 10% 소태아혈청(fetal bovine serum; FBS) 및 페니실린/스트렙토마이신(100U/ml)이 보충된 DMEM(Dulbecco's modified Eagle Medium)에서 배양하였다. 12 웰 플레이트에서 Huh7 세포에 본 발명에 따른 실시예 1 내지 3(화합물 23-25) 및 실시예 5 내지 8(화합물 31-34)을 20μM씩 처리하여 CO2 배양기에서 24시간 배양하였다. 면역블로팅을 통해 각 화합물이 세포의 p38 MAPK의 양의 감소에 어떤 영향을 미쳤는지 확인하였다.Specifically, Huh7 cells were cultured in Dulbecco's modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and penicillin/streptomycin (100 U/ml). In a 12-well plate, Huh7 cells were treated with 20 μM each of Examples 1 to 3 (Compounds 23-25) and Examples 5 to 8 (Compounds 31-34) according to the present invention, and incubated in a CO 2 incubator for 24 hours. It was confirmed how each compound had an effect on the decrease in the amount of p38 MAPK in cells through immunoblotting.

실험 조건에 따라 세포의 배양이 끝난 후, 차가운 PBS로 세척한다. 이후 세포를 수거하여 12000 rpm에서 3분동안 분리한다. 펠렛을 2X sample buffer (1M Tris-HCl pH 6.8, 50 % 글리세롤, 10 % SDS, 2-머캅토 에탄올, 1% 브로모페놀블루)으로 세포를 용해하고, 100 ℃에서 10분 동안 끓였다. 제조된 단백질 샘플을 전기영동(Sodium Dodecyl sulfate poly acrylamide gel electrophoresis, SDS-PAGE)하고, PVDF membrane(제조사: Millipore)으로 전사하였다. PVDF membrane에 비특이적으로 항체가 결합하는 것을 막기위하여 5 % bovine serum albumin (BSA, 제조사: RMbio) 용액으로 실온에서 1시간 blocking을 수행하였다. 5 % bovine serum albumin (BSA, 제조사: RMbio) 용액에 각각 타겟 항체를 희석하여 PVDF membrane을 4 ℃에서 밤새 반응시켰다. 이어서 항체가 희석된 BSA 용액을 폐기하고 PVDF membrane을 PBST buffer (PBS, 0.5 % tweene 20)로 5회 세척하였다. 마지막으로, PVDF membrane을 Horse radish peroxidase(HRP)-접합된 이차 항체 (제조사: Cell signaling Technology)가 희석된 PBST buffer를 넣고 실온에서 2 시간 동안 반응시켰다. 5회 PBST buffer로 세척한 후 HRP substrate (제품명: Luminata Forte, 제조사: Millipore)를 사용하여 membrane의 단백질 양을 확인하였다.After culturing the cells according to the experimental conditions, they are washed with cold PBS. Thereafter, the cells were harvested and separated at 12000 rpm for 3 minutes. The pellet was lysed with 2X sample buffer (1M Tris-HCl pH 6.8, 50% glycerol, 10% SDS, 2-mercaptoethanol, 1% bromophenol blue) and boiled at 100°C for 10 minutes. The prepared protein sample was electrophoresed (Sodium Dodecyl sulfate poly acrylamide gel electrophoresis, SDS-PAGE) and transferred to a PVDF membrane (manufacturer: Millipore). In order to prevent non-specific binding of the antibody to the PVDF membrane, blocking was performed for 1 hour at room temperature with a 5% bovine serum albumin (BSA, manufacturer: RMbio) solution. Each target antibody was diluted in 5% bovine serum albumin (BSA, manufacturer: RMbio) solution, and the PVDF membrane was reacted overnight at 4°C. Then, the antibody-diluted BSA solution was discarded and the PVDF membrane was washed 5 times with PBST buffer (PBS, 0.5% tweene 20). Finally, PBST buffer diluted with Horse radish peroxidase (HRP)-conjugated secondary antibody (manufacturer: Cell signaling Technology) was added to the PVDF membrane and reacted at room temperature for 2 hours. After washing with PBST buffer 5 times, the amount of protein in the membrane was confirmed using HRP substrate (product name: Luminata Forte, manufacturer: Millipore).

도 2는 본 발명의 실시예에 따른 화합물 7종의 p38 MAPK 제거능을 면역블롯팅 방법으로 확인한 결과이다.2 is a result of confirming the p38 MAPK removal ability of 7 types of compounds according to an embodiment of the present invention by immunoblotting method.

도 2에 나타난 바와 같이, 각 화합물의 처리는 총 단백질 수준(β-actin; 세포유지유전자)에 영향을 주지 않으면서, p38 MAPK의 제거를 유도하였다. 그 중 화합물 24(실시예 2)가 가장 효과적으로 p38 MAPK의 감소를 유도하는 것을 확인할 수 있었다. 3회 반복 실험을 통해 화합물 24(실시예 2)가 가장 효과가 좋은 물질임을 확인하였다.As shown in FIG. 2 , treatment with each compound induced the removal of p38 MAPK without affecting the total protein level (β-actin; cytostatic gene). Among them, it was confirmed that compound 24 (Example 2) most effectively induced a decrease in p38 MAPK. It was confirmed that compound 24 (Example 2) was the most effective substance through three repeated experiments.

<실험예 2> 화합물 24(실시예 2)의 농도 의존적 p38 MAPK 제거능 평가<Experimental Example 2> Concentration-dependent evaluation of p38 MAPK removal ability of compound 24 (Example 2)

본 발명에 따른 화합물 24(실시예 2)가 낮은 농도에서도 p38 MAPK의 제거를 유도하는지 확인하기 위해 HEK 293T(Human Embryonic Kidney 293T) 세포에 p38 MAPK-Flag를 과다발현시킨 후 화합물 24를 처리하여 면역블로팅을 진행하였다.In order to determine whether compound 24 according to the present invention (Example 2) induces the removal of p38 MAPK even at a low concentration, p38 MAPK-Flag was overexpressed in HEK 293T (Human Embryonic Kidney 293T) cells and then treated with compound 24 for immunity Blotting was performed.

구체적으로, HEK 293T 세포를 10% 소태아혈청(fetal bovine serum; FBS) 및 페니실린/스트렙토마이신(100U/ml)이 보충된 DMEM(Dulbecco's modified Eagle Medium)에서 배양하였다. 이후 Transfection을 통해 HEK 293T 세포에 p38 MAPK-Flag의 과다발현을 유도하였다. 12 웰 플레이트에서 HEK 293T 세포에 본 발명에 따른 화합물 24(실시예 2)를 농도별로 처리하여 24시간 배양하였다. Specifically, HEK 293T cells were cultured in Dulbecco's modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and penicillin/streptomycin (100U/ml). Then, overexpression of p38 MAPK-Flag was induced in HEK 293T cells through transfection. In a 12-well plate, HEK 293T cells were treated with compound 24 (Example 2) according to the present invention by concentration and cultured for 24 hours.

도 3은 본 발명의 실시예 2에 따른 화합물 24의 처리 농도에 따른 p38(Flag) 및 P-p38의 제거능을 면역블롯팅 방법으로 확인한 결과이다.3 is a result of confirming the removal ability of p38 (Flag) and P-p38 according to the treatment concentration of Compound 24 according to Example 2 of the present invention by immunoblotting method.

도 3에 나타난 바와 같이, 화합물 24의 처리는 총 단백질 수준에 영향을 주지 않으면서(β-actin), 농도 의존적으로 p38 MAPK-Flag를 제거하였으며, p38 MAPK가 인산화된 P-p38이 더 낮은 농도에서도 효과적으로 제거되는 것을 확인하였다. 이는 화합물 24(실시예 2)가 P-p38 선택적으로 유비퀴틴화를 유도하여 제거함을 나타낸다.As shown in FIG. 3 , treatment with compound 24 removed p38 MAPK-Flag in a concentration-dependent manner without affecting the total protein level (β-actin), and p38 MAPK phosphorylated P-p38 had a lower concentration. It was also confirmed that it was effectively removed. This indicates that compound 24 (Example 2) selectively induces and eliminates P-p38 ubiquitination.

<실험예 3> 실시예 화합물에서 p38 MAPK에 선택적으로 결합하는 Moiety의 확인<Experimental Example 3> Identification of a moiety that selectively binds to p38 MAPK in the example compound

본 발명의 실시예 화합물은 [p38 Ligand Moiety]-[Linker]-[E3 Ligand Moiety]으로 구성되며, 이 중에서 어느 부위가 p38 MAPK에 선택적으로 결합하는 Moiety인지 알아보기 위하여 다음과 같은 실험을 하였다.The example compound of the present invention is composed of [p38 Ligand Moiety]-[Linker]-[E3 Ligand Moiety], and in order to find out which moiety is a moiety that selectively binds to p38 MAPK, the following experiment was performed.

구체적으로, 화합물 24(실시예 2)의 E3 Ligase Ligand Moiety (탈리도미드 유도체) 대신 Biotin을 결합한 화합물 43(비교예 1)을 합성하였다. 이 후 Biotin을 이용하여 pull-down assay를 진행하였다. 화합물 24의 [p38 Ligand Moiety]와 p38 MAPK의 결합으로 인해 pull-down assay에서 p38 MAPK 가 검출되는 것인지에 대한 정확한 비교를 위해, 화합물 24에서 [p38 Ligand Moiety]를 제외한 Linker에 Biotin을 결합한 화합물 45를 대조군으로 사용하였다.Specifically, Compound 43 (Comparative Example 1) in which Biotin was bound instead of E3 Ligase Ligand Moiety (thalidomide derivative) of Compound 24 (Example 2) was synthesized. Thereafter, a pull-down assay was performed using Biotin. For an accurate comparison of whether p38 MAPK is detected in the pull-down assay due to the binding of [p38 Ligand Moiety] and p38 MAPK of Compound 24, Compound 45 that binds Biotin to Linker except for [p38 Ligand Moiety] in Compound 24 was used. It was used as a control.

Biotin-pull-down asaay는 다음과 같이 진행하였다. BV-2 세포를 0.1 % TritonX-100을 사용하여 용해시켰다. Biotin과 합성된 화합물 43, Linker와 합성된 화합물 45를 용해물에 20μM 농도로 처리하고 37 ℃에서 밤새 반응시켰다. 이어서, Biotin과 결합하는 것으로 잘 알려진 Streptavidin과 연결된 agarose bead를 용해물에 40μL 첨가하고 37 ℃에서 4 시간 동안 반응시켰다. 이후 4000rpm 1분 동안 원심분리를 통해 Streptavidin을 가라앉히고 상층액을 제거하였다. 0.1 % Triton X-100 Lysis buffer로 세척하고 다시 4000rpm 1분 원심분리를 통해 Streptavidin을 가라앉혔다. 이를 5회 반복하여 Streptavidin과 결합한 Biotin 화합물, 그 화합물과 결합한 단백질을 제거한 다른 불순물을 제거하는 과정을 거쳤다. 2X sample buffer를 첨가한 후 100℃에서 10 분 동안 끓여 샘플을 준비하였다. 이후 단백질 샘플을 SDS-PAGE 및 면역블롯팅으로 분석하였다.Biotin-pull-down asaay was performed as follows. BV-2 cells were lysed using 0.1% TritonX-100. Compound 43 synthesized with Biotin and Compound 45 synthesized with Linker were treated at a concentration of 20 μM in the lysate and reacted at 37° C. overnight. Then, 40 μL of agarose beads linked to Streptavidin, which is well known for binding to biotin, were added to the lysate and reacted at 37° C. for 4 hours. Then, Streptavidin was settled by centrifugation at 4000 rpm for 1 minute and the supernatant was removed. After washing with 0.1 % Triton X-100 Lysis buffer, Streptavidin was submerged again by centrifugation at 4000 rpm for 1 minute. This process was repeated 5 times to remove the Biotin compound bound to Streptavidin and other impurities from which the protein bound to the compound was removed. After adding 2X sample buffer, the sample was prepared by boiling at 100° C. for 10 minutes. Protein samples were then analyzed by SDS-PAGE and immunoblotting.

도 4는 본 발명의 실시예 2(화합물24)에서 [E3 Ligand Moiety(탈리도미드 유도체)] 대신에 비오틴을 결합한 비교예 1(화합물 43)과 실시예 2(화합물24)에서 [p38 Ligand Moiety]를 제외한 Linker에 Biotin을 결합한 비교예 2(화합물 45)를 각각 p38 MAPK 결합능을 평가한 결과이다.Figure 4 shows [p38 Ligand Moiety in Comparative Example 1 (Compound 43) and Example 2 (Compound 24) in which biotin was conjugated instead of [E3 Ligand Moiety (thalidomide derivative)] in Example 2 (Compound 24) of the present invention. ] is the result of evaluating the p38 MAPK binding ability of Comparative Example 2 (Compound 45) in which Biotin was bound to the Linker, respectively.

도 4에 나타난 바와 같이, [p38 Ligand Moiety]를 포함하고 있는 비교예 1(화합물 43)을 처리한 군에서 p38 MAPK의 검출이 확인된 반면에, [p38 Ligand Moiety]를 포함하지 않는 비교예 2(화합물 45)를 처리한 군에서는 p38 MAPK이 미검출을 확인하였다(Pull-down assay 참조). 이 결과는 본 발명의 실시예 화합물에서 [p38 Ligand Moiety] 부위가 p38 MAPK와 선택적으로 결합하는 것을 증명하는 결과이다.As shown in FIG. 4 , the detection of p38 MAPK was confirmed in the group treated with Comparative Example 1 (Compound 43) containing [p38 Ligand Moiety], whereas Comparative Example 2 without [p38 Ligand Moiety] In the group treated with (Compound 45), it was confirmed that p38 MAPK was not detected (see Pull-down assay). This result proves that the [p38 Ligand Moiety] site selectively binds to p38 MAPK in the Example compound of the present invention.

<실험예 4> 실시예 화합물의 Proteasome 의존적 p38 MAPK 제거 확인<Experimental Example 4> Proteasome-dependent p38 MAPK removal confirmation of Example compound

본 발명의 실시예 화합물이 세포 내의 proteasome system을 사용하여 p38 MAPK의 제거를 유도함을 확인하기 위하여 실험을 하였다. An experiment was conducted to confirm that the Example compound of the present invention induces the removal of p38 MAPK using the intracellular proteasome system.

구체적으로, 12 웰 플레이트에서 Huh7 세포에 화합물 24(실시예 2) 20μM을 24시간 처리하고 마지막 3시간에 Lysosome Inhibitor인 Chloroquine(CQ), Proteasome Inhibitor인 MG132를 각각 100μM, 10μM 처리하였다. 마지막 3시간을 통해 세포 내의 Lysosome과 Proteasome의 기능을 차단하였다. 다음으로, 실험예 1의 면역블롯팅 방법과 동일하게 수행하여 평가하였고, 3회 이상 반복을 통하여 유의성을 확인하였다.Specifically, in a 12-well plate, Huh7 cells were treated with 20 μM of Compound 24 (Example 2) for 24 hours, and in the last 3 hours, Lysosome Inhibitor Chloroquine (CQ) and Proteasome Inhibitor MG132 were treated with 100 μM and 10 μM, respectively. Lysosome and proteasome functions in cells were blocked through the last 3 hours. Next, evaluation was performed in the same manner as in the immunoblotting method of Experimental Example 1, and significance was confirmed through repetition of three or more times.

도 5는 본 발명에 따른 실시예 화합물이 세포 내의 Proteasome system을 사용하여 p38 MAPK를 제거시키는 것을 면역블롯팅 방법으로 확인한 결과이다.5 is a result confirming that the Example compound according to the present invention removes p38 MAPK using the intracellular proteasome system by immunoblotting method.

도 5에 나타난 바와 같이, Proteasome의 기능이 감소한 실험군(MG132 + 화합물 24 처리군)에서만 p38 MAPK 단백질 양의 복구를 확인하였다. 이는 실시예 화합물이 세포 내의 Proteasome system을 사용하여 p38 MAPK를 감소시킨다는 것을 확인할 수 있는 결과이다.As shown in FIG. 5 , recovery of the amount of p38 MAPK protein was confirmed only in the experimental group in which the function of the proteasome was reduced (MG132 + compound 24 treatment group). This is a result confirming that the example compound reduces p38 MAPK using the intracellular proteasome system.

<실험예 5> 실시예 화합물의 CRBN 의존적 p38 MAPK 제거 확인<Experimental Example 5> Confirmation of CRBN-dependent p38 MAPK removal of Example compounds

본 발명의 실시예 화합물이 세포 내의 E3 ligase complex 단백질 중 하나인 cereblon (CRBN)을 사용하여 p38 MAPK의 제거를 유도함을 확인하기 위하여 실험하였다. An experiment was conducted to confirm that the Example compound of the present invention induces the removal of p38 MAPK using cereblon (CRBN), one of the E3 ligase complex proteins in the cell.

구체적으로, 12 웰 플레이트에서 BV2 세포에 CRBN Inhibitor인 Pomalidomide를 농도별로 6시간 처리하여 CRBN을 저해하였다. 이후 화합물 24를 24시간 처리한 후 세포를 분리하여 용해하였다. 다음으로, 실험예 1의 면역블롯팅 방법과 동일하게 수행하여 평가하였다.Specifically, in a 12-well plate, BV2 cells were treated with Pomalidomide, a CRBN inhibitor, for 6 hours at each concentration to inhibit CRBN. Thereafter, the cells were separated and lysed after treatment with Compound 24 for 24 hours. Next, evaluation was performed in the same manner as in the immunoblotting method of Experimental Example 1.

도 6은 본 발명에 따른 실시예 화합물이 세포 내의 E3 ligase complex 단백질 중 하나인 cereblon (CRBN)을 사용하여 p38 MAPK를 제거시키는 것을 면역블롯팅 방법으로 확인한 결과이다.6 is a result of confirming that the Example compound according to the present invention removes p38 MAPK using cereblon (CRBN), one of the E3 ligase complex proteins in cells, by immunoblotting method.

도 6에 나타난 바와 같이, Pomalidomide를 처리하여 CRBN를 저해한 후 화합물 24(실시예 2)를 처리한 군에서 P-p38 MAPK 단백질 양이 복구되는 것을 확인하였다. 이는 화합물 24가 CRBN를 매개로 P-p38 MAPK의 감소를 유도시킨다는 것을 확인할 수 있는 결과이다.As shown in FIG. 6 , it was confirmed that the amount of P-p38 MAPK protein was recovered in the group treated with Compound 24 (Example 2) after CRBN was inhibited by treatment with Pomalidomide. This is a result confirming that Compound 24 induces a decrease in P-p38 MAPK mediated by CRBN.

<실험예 6> 실시예 화합물의 유비퀴틴화(Ubiquitination) 의존적 p38 MAPK 제거 확인<Experimental Example 6> Confirmation of ubiquitination-dependent p38 MAPK removal of Example compounds

실시예 화합물이 유비퀴틴화를 사용하여 p38 MAPK의 제거를 유도함을 확인하기 위하여 실험하였다.An experiment was conducted to confirm that the Example compound induces the removal of p38 MAPK using ubiquitination.

구체적으로, P38 MAPK이 유비퀴틴화를 통한 proteasome 의존적 단백질 감소가 발생하는지 확인하기 위해 면역침강법을 사용하여 P38 MAPK 단백질을 분리하였다. 면역 침강법은 각 샘플 당 2 X 107개의 HEK293T 세포를 준비하여 화합물 24(실시예 2)와 화합물 16(비교예 3)을 10μM 로 처리하고 24시간 배양하였다. 이후 세포를 0.5% Triton X-100 lysis buffer (50mM Tris, 150mM NaCl, 1mM EDTA, 0.5% triton X-100)에 용해시켰다. 이어서, 소량의 용해물을, 5X sample buffer를 통해 샘플링하고, Anti-p38 rabbit antibody (제조사: Cell Signaling Technology)를 용해물에 첨가하고 4 ℃에서 밤새 반응시켰다. 이어서, Protein A / G resins(제조사: Sigma-Aldrich)를 첨가하고 4 ℃에서 4 시간 동안 반응시켰다. 0.5 % Triton X-100 lysis buffer으로 세척하여 A/G resins과 결합한 단백질 외의 불순물을 제거하였다. 이후 단백질 샘플을 SDS-PAGE 및 면역블롯팅으로 분석하였다.Specifically, P38 MAPK protein was isolated by immunoprecipitation to determine whether proteasome-dependent protein reduction through ubiquitination of P38 MAPK occurred. For the immunoprecipitation method, 2 X 10 7 HEK293T cells were prepared for each sample, treated with Compound 24 (Example 2) and Compound 16 (Comparative Example 3) with 10 μM and cultured for 24 hours. The cells were then lysed in 0.5% Triton X-100 lysis buffer (50 mM Tris, 150 mM NaCl, 1 mM EDTA, 0.5% triton X-100). Then, a small amount of the lysate was sampled through 5X sample buffer, and Anti-p38 rabbit antibody (manufacturer: Cell Signaling Technology) was added to the lysate and reacted at 4°C overnight. Then, Protein A / G resins (manufacturer: Sigma-Aldrich) were added and reacted at 4 °C for 4 hours. Impurities other than proteins bound to A/G resins were removed by washing with 0.5 % Triton X-100 lysis buffer. Protein samples were then analyzed by SDS-PAGE and immunoblotting.

도 7은 본 발명에 따른 실시예 화합물이 유비퀴틴화 시스템을 사용하여 p38 MAPK를 제거시키는 것을 SDS-PAGE 및 면역블롯팅 방법으로 확인한 결과이다.7 is a result confirming that the Example compound according to the present invention removes p38 MAPK using the ubiquitination system by SDS-PAGE and immunoblotting methods.

도 7에 나타난 바와 같이, 화합물 24(실시예 2)를 처리한 군에서 p38 MAPK 단백질의 유비퀴틴화가 발생함을 확인하였고, 반면 화합물 24에서 [E3 ligase Ligand Moiety]를 붙이기 전단계의 화합물인 화합물 16(비교예 3)을 처리한 군에서는 p38 MAPK 단백질의 유비퀴틴화가 증가되지 않음을 확인하였다.As shown in Figure 7, it was confirmed that ubiquitination of p38 MAPK protein occurred in the group treated with Compound 24 (Example 2), whereas Compound 16, the compound of the step before attaching [E3 ligase Ligand Moiety] in Compound 24 ( In the group treated with Comparative Example 3), it was confirmed that ubiquitination of the p38 MAPK protein was not increased.

<실험예 7> 신경세포에서 실시예 화합물의 p38 MAPK, P-p38 MAPK 감소 유도 확인<Experimental Example 7> Confirmation of p38 MAPK, P-p38 MAPK reduction induction of the compound of Example in neurons

신경세포에서 실시예 화합물이 p38 MAPK의 감소를 유도하는지 확인하기 위해 네 종류의 다른 마우스 세포주에서 실험을 진행하였다.In order to confirm whether the Example compound induces a decrease in p38 MAPK in neurons, an experiment was conducted in four different mouse cell lines.

구체적으로, 각 실험은 도 8에 표기된 바와 같이 화합물 24를 0.001~10μM의 농도로 처리한 후 24시간 동안 배양하였다. 배양 후 실험예 1과 동일하게 면역블로팅을 진행하였다.Specifically, in each experiment, as indicated in FIG. 8, compound 24 was treated at a concentration of 0.001 to 10 μM and then cultured for 24 hours. After culture, immunoblotting was performed in the same manner as in Experimental Example 1.

도 8은 본 발명에 따른 실시예 화합물이 신경세포에서 농도 의존적으로 p38 MAPK 및 P-p38 MAPK 감소를 유도함을 면역블롯팅 방법으로 확인한 결과이다. 8 is a result confirming that the compound of Example according to the present invention induces a concentration-dependent decrease in p38 MAPK and P-p38 MAPK in neurons by immunoblotting method.

도 8(A): 신경아교세포(Astrocyte)Figure 8(A): Astrocytes

도 8(B): 신경모세포(Neuroblast)Figure 8(B): Neuroblasts

도 8(C): 미세아교세포(Microglia)Figure 8(C): Microglia

도 8(D): 해마신경세포(Hippocampal Neuronal cell)Figure 8(D): Hippocampal Neuronal cells

도 8(E): 미세아교세포(Microglia)에서 3회 이상 반복 실험한 후 상대적인 값을 수치화하여 나타냄.Figure 8 (E): After repeated experiments three or more times in microglia (Microglia), the relative values are shown numerically.

도 8에 나타난 바와 같이, 신경아교세포(astrocyte, 도 8A)와 신경모세포(Neuroblast, 도 8B)에 화합물 24(실시예 2)를 처리한 결과 농도 의존적으로 감소하였고 P-p38 MAPK는 0.01μM, 0.1μM에서도 감소하는 것을 확인하였다. 미세아교세포(Microglia, 도 8C)와 해마신경세포(Hippocampal Neuronal cell, 도 8D)에서도 같은 실험을 진행한 결과 화합물 24 농도 의존적으로 p38이 감소하였고, 0.1μM에서도 P-p38이 감소하는 것을 확인하였다. 도 8A ~ 도 8D의 결과로 보았을 때 신경세포에서 화합물 24이 P-p38 MAPK를 더 효과적으로 감소시키는 것을 알 수 있었다.As shown in FIG. 8, as a result of treatment with Compound 24 (Example 2) in glial cells (astrocyte, FIG. 8A) and neuroblastic cells (Neuroblast, FIG. 8B) concentration-dependently decreased, P-p38 MAPK was 0.01 μM, 0.1 It was confirmed that it also decreased in μM. As a result of the same experiment in microglia (Microglia, FIG. 8C) and hippocampal neuronal cells (FIG. 8D), it was confirmed that p38 was decreased in a compound 24 concentration-dependent manner, and that P-p38 was decreased even at 0.1 μM. . 8A to 8D, it was found that Compound 24 more effectively reduced P-p38 MAPK in neurons.

<실험예 8> 실시예 화합물 처리로 인한 p38 MAPK 관련 사이토카인 생성 감소 평가<Experimental Example 8> Evaluation of reduction of p38 MAPK-related cytokine production due to treatment with Example compound

실시예 화합물 처리로 인하여 p38 MAPK가 제거되면, p38 MAPK에 의해 생성되는 사이토카인의 생성량이 감소할 것으로 예상되어, 이를 검증하기 위하여 실험하였다.When p38 MAPK is removed by treatment with the compound of Example, it is expected that the amount of cytokine produced by p38 MAPK will decrease, and an experiment was conducted to verify this.

구체적으로, 24 웰 플레이트에 미세아교세포인 BV2 세포를 배양하였다. 화합물 24(실시예 2)가 세포 내에서 p38 MAPK를 유비퀴틴화하여 프로테아좀 의존적으로 p38을 제거함에 따라서, p38 관련 사이토카인의 생성도 감소되는지 비교하기 위하여, 화합물 16(비교예 3)을 대조군으로 처리하여 비교하였다. 실험 조건은 다음과 같다. 각 화합물을 10μM 또는 20μM로 24시간 선 처리한 후, 사이토카인의 생성을 유도하기 위해 LPS(lipopolysaccharide)를 1μg/mL로 새 세포배양액에 희석하여 처리한 후 배양하였다. 24시간 후에 세포배양액의 상층액을 모아 사이토카인 생산량을 효소면역흡착법(enzyme-linked immunosorbent assay; ELISA, 제조사: Bio-kit)으로 분석하였다. 분석방법은 Bio-kit에 제시된 프로토콜을 따랐다. 실험은 3회 이상 반복하여 도 9의 그래프로 나타냈다.Specifically, microglia, BV2 cells, were cultured in a 24-well plate. In order to compare whether compound 24 (Example 2) ubiquitinates p38 MAPK in cells and removes p38 in a proteasome-dependent manner, the production of p38-related cytokines is also reduced, compound 16 (Comparative Example 3) was used as a control was treated and compared. The experimental conditions are as follows. Each compound was pre-treated with 10 μM or 20 μM for 24 hours, and then diluted with LPS (lipopolysaccharide) at 1 μg/mL in a fresh cell culture medium to induce cytokine production, and then cultured. After 24 hours, the supernatant of the cell culture medium was collected and the cytokine production was analyzed by enzyme-linked immunosorbent assay (ELISA, manufacturer: Bio-kit). The analysis method followed the protocol presented in the Bio-kit. The experiment was repeated three or more times and is shown in the graph of FIG. 9 .

도 9는 본 발명의 실시예 화합물 처리로 인한 p38 MAPK 관련 사이토카인 생성량이 감소됨을 ELISA 방법으로 확인한 결과이다.9 is a result of confirming by the ELISA method that the amount of p38 MAPK-related cytokine production due to treatment with the compound of Example of the present invention is reduced.

도 9에 나타난 바와 같이, 화합물 24(실시예 2)를 처리한 군에서 p38 MAPK 관련 사이토카인인 인터루킨6 (IL-6), 종양괴사인자 알파 (TNF-α)의 생성량이 감소하는 것을 확인하였다. 화합물 16(비교예 3)과 비교시 더 효과적으로 감소한 것을 확인하였다.As shown in FIG. 9 , it was confirmed that the production amount of p38 MAPK-related cytokines interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) decreased in the group treated with Compound 24 (Example 2). . It was confirmed that the decrease was more effectively compared with compound 16 (Comparative Example 3).

<실험예 9> 실시예 화합물 처리로 인한 p38 MAPK 관련 사이토카인의 mRNA 발현량 감소 평가<Experimental Example 9> Evaluation of decrease in mRNA expression level of p38 MAPK-related cytokines due to treatment with Example compounds

실시예 화합물 처리로 인하여 p38 MAPK가 제거되면, p38 MAPK 관련 Cytokine의 mRNA 발현량도 감소할 것으로 예상되어, 이를 검증하기 위하여 실험하였다.When p38 MAPK is removed by treatment with the compound of Example, it is expected that the mRNA expression level of p38 MAPK-related cytokine will also decrease, and an experiment was conducted to verify this.

구체적으로, 6 웰 플레이트에 미세아교세포인 BV2 세포를 배양하였다. 이후 화합물 24(실시예 2)을 10μM, 20μM 농도로 24시간 처리하였다. P38 MAPK 관련 사이토카인의 발현을 증가시키기 위해 화합물의 24시간 처리 중 마지막 6시간은 LPS 1μg/mL을 함께 처리하였다. 이후 사이토카인의 mRNA 전사를 qRT-PCR (Real-Time Quantitative Reverse Transcription PCR) 로 분석하였다. RNA 분리는 RNA 정제 키트를 이용하여 순수한 RNA만 추출하였다. 분리된 RNA의 농도와 순도는 분광 광도계 (spectrophotometer) 로 측정된다. 각 샘플의 RNA는 cDNA 합성 키트를 이용하여 cDNA (complementary DNA)를 합성하고, SYBR Green을 이용한 qRT-PCR 분석법을 통해 염증성 사이토카인인 인터루킨6 (IL-6)와 인터루킨1 베타 (IL-1β) 및 인터루킨12 (IL-12)의 mRNA의 발현량을 측정하였다.Specifically, microglia, BV2 cells, were cultured in a 6-well plate. Thereafter, compound 24 (Example 2) was treated at a concentration of 10 μM and 20 μM for 24 hours. In order to increase the expression of P38 MAPK-related cytokines, the last 6 hours of the 24 hour treatment of the compound was treated with LPS 1 μg/mL. Then, mRNA transcription of cytokines was analyzed by qRT-PCR (Real-Time Quantitative Reverse Transcription   PCR). For RNA isolation, only pure RNA was extracted using an RNA purification kit. The concentration and purity of the isolated RNA were measured with a spectrophotometer. For RNA of each sample, cDNA (complementary DNA) was synthesized using a cDNA synthesis kit, and inflammatory cytokines interleukin 6 (IL-6) and interleukin 1 beta (IL-1β) were synthesized by qRT-PCR analysis using SYBR Green. and interleukin 12 (IL-12) mRNA expression levels were measured.

도 10은 본 발명의 실시예 화합물이 p38 MAPK 관련 사이토카인의 mRNA 발현량 감소시킴을 qRT-PCR 방법으로 확인한 결과이다.10 is a result confirming that the Example compound of the present invention reduces the mRNA expression level of p38 MAPK-related cytokines by qRT-PCR method.

도 10에 나타난 바와 같이, 인터루킨6 (IL-6)와 인터루킨1 베타 (IL-1β) 및 인터루킨12 (IL-12)의 mRNA양이 화합물 24(실시예 2)를 처리한 군에서 감소하는 것을 확인하였다. 이 결과로 보았을 때 화합물 24(실시예 2)는 p38 MAPK의 제거를 유도하고 이로 인하여 p38 MAPK 관련 염증성 사이토카인의 mRNA 발현량 또한 감소하는 것을 알 수 있다.As shown in FIG. 10 , the mRNA levels of interleukin 6 (IL-6), interleukin 1 beta (IL-1β) and interleukin 12 (IL-12) decreased in the group treated with compound 24 (Example 2). Confirmed. From these results, it can be seen that Compound 24 (Example 2) induces the removal of p38 MAPK, thereby also reducing the mRNA expression level of p38 MAPK-related inflammatory cytokines.

약제의 제조예Preparation example of drug

본 발명에 따른 유효물질은 목적에 따라 여러 형태로 제제화가 가능하다. 하기는 본 발명에 따른 유효물질을 활성성분으로 함유시킨 몇몇 제제화 방법을 예시한 것으로 본 발명이 이에 한정되는 것은 아니다.The active substance according to the present invention can be formulated in various forms depending on the purpose. The following exemplifies some formulation methods containing the active substance according to the present invention as an active ingredient, but the present invention is not limited thereto.

<약제 제조예 1> 산제의 제조<Pharmaceutical Preparation Example 1> Preparation of powder

유효물질 2 gactive substance 2 g

유당 1 glactose 1 g

상기의 성분을 혼합한 후, 기밀포에 충진하여 산제를 제조하였다.After mixing the above ingredients, the powder was prepared by filling in an airtight cloth.

<약제 제조예 2> 정제의 제조<Pharmaceutical Preparation Example 2> Preparation of tablets

유효물질 100 ㎎active substance 100 mg

옥수수전분 100 ㎎corn starch 100 mg

유 당 100 ㎎lactose 100 mg

스테아린산 마그네슘 2 ㎎magnesium stearate 2 mg

상기의 성분을 혼합한 후, 통상의 정제의 제조방법에 따라서 타정하여 정제를 제조하였다.After mixing the above ingredients, tablets were prepared by tableting according to a conventional method for manufacturing tablets.

<약제 제조예 3> 캡슐제의 제조<Pharmaceutical Preparation Example 3> Preparation of capsules

유효물질 100 ㎎active substance 100 mg

옥수수전분 100 ㎎corn starch 100 mg

유 당 100 ㎎lactose 100 mg

스테아린산 마그네슘 2 ㎎magnesium stearate 2 mg

상기의 성분을 혼합한 후, 통상의 캡슐제의 제조방법에 따라서 젤라틴 캡슐에 충전하여 캡슐제를 제조하였다.After mixing the above ingredients, the capsules were prepared by filling in gelatin capsules according to a conventional manufacturing method of capsules.

<약제 제조예 4> 주사제의 제조<Pharmaceutical Preparation Example 4> Preparation of injection

유효물질 10 ㎍/㎖active substance 10 μg/ml

묽은 염산 BP pH 3.5로 될 때까지dilute hydrochloric acid BP until pH 3.5

주사용 염화나트륨 BP 최대 1 ㎖Sodium Chloride BP for Injection up to 1 ml

적당한 용적의 주사용 염화나트륨 BP 중에 본 발명에 따른 유효물질을 용해시키고, 생성된 용액의 pH를 묽은 염산 BP를 사용하여 pH 3.5로 조절하고, 주사용 염화나트륨 BP를 사용하여 용적을 조절하고 충분히 혼합하였다. 용액을 투명 유리로 된 5 ㎖ 타입 I 앰플 중에 충전시키고, 유리를 용해시킴으로써 공기의 상부 격자하에 봉입시키고, 120 ℃에서 15 분 이상 오토클래이브시켜 살균하여 주사액제를 제조하였다.The active substance according to the present invention was dissolved in an appropriate volume of sodium chloride BP for injection, the pH of the resulting solution was adjusted to pH 3.5 using dilute hydrochloric acid BP, the volume was adjusted using sodium chloride BP for injection, and the mixture was sufficiently mixed. . The solution was filled in a 5 ml Type I ampoule made of clear glass, sealed under an upper grid of air by dissolving the glass, and sterilized by autoclaving at 120° C. for 15 minutes or more to prepare an injection solution.

<약제 제조예 5> 경비흡수제 (Nasal spray)의 제조<Pharmaceutical Preparation Example 5> Preparation of nasal absorbent (Nasal spray)

유효물질 1.0 gactive substance 1.0 g

아세트산나트륨 0.3 gsodium acetate 0.3 g

메틸파라벤 0.1 gmethylparaben 0.1 g

프로필파라벤 0.02 gPropylparaben 0.02 g

염화나트륨 적량sodium chloride appropriate amount

HCl 또는 NaOH pH 조정 적량HCl or NaOH pH adjustment appropriate amount

정제수 적량Purified water appropriate amount

통상의 경비흡수제의 제조방법에 따라, 염수 (0.9% NaCl, w/v, 용매는 정제수) 1 mL당 유효물질 3 mg이 포함되도록 제조하고, 이를 불투명한 스프레이 용기에 충진하고 멸균시켜 경비흡수제를 제조하였다.According to a conventional method for preparing nasal absorbents, prepare to contain 3 mg of active substance per 1 mL of saline (0.9% NaCl, w/v, solvent is purified water), fill it in an opaque spray container, and sterilize to obtain nasal absorbent. prepared.

<약제 제조예 6> 액제의 제조<Pharmaceutical Preparation Example 6> Preparation of liquid preparation

유효물질 100 mgactive substance 100 mg

이성화당 10 gLee Seonghwadang 10 g

만니톨 5 gmannitol 5 g

정제수 적량Purified water appropriate amount

통상의 액제의 제조방법에 따라, 정제수에 각각의 성분을 가하여 용해시키고 레몬 향을 가한 다음 상기의 성분을 혼합한 다음 정제수를 가하여 전체 100 mL로 조절한 후 갈색 병에 충진하고 멸균시켜 액제를 제조하였다.According to a conventional liquid preparation method, each component is added and dissolved in purified water, lemon flavor is added, the above components are mixed, and purified water is added to adjust the total to 100 mL, then filled in a brown bottle and sterilized to prepare a liquid preparation did.

건강기능식품의 제조예Manufacturing example of health functional food

본 발명에 따른 유효물질은 목적에 따라 여러 형태의 건강기능식품으로 제조 가능하다. 하기는 본 발명에 따른 유효물질을 활성성분으로 함유시킨 몇몇 건강기능식품의 제조방법을 예시한 것으로 본 발명이 이에 한정되는 것은 아니다.The active substance according to the present invention can be manufactured into various types of health functional food depending on the purpose. The following exemplifies the manufacturing method of several health functional foods containing the active ingredient according to the present invention as an active ingredient, but the present invention is not limited thereto.

<건강기능식품 제조예 1> 건강기능식품의 제조<Health functional food manufacturing example 1> Manufacture of health functional food

유효물질 100 mgactive substance 100 mg

비타민 혼합물 적량vitamin mixture appropriate amount

비타민 A 아세테이트 70 μgvitamin A acetate 70 μg

비타민 E 1.0 mgvitamin E 1.0 mg

비타민 B1 0.13 mgvitamin B1 0.13 mg

비타민 B2 0.15 mgvitamin B2 0.15 mg

비타민 B6 0.5 mgvitamin B6 0.5 mg

비타민 B12 0.2 μgvitamin B12 0.2 μg

비타민 C 10 mgvitamin C 10 mg

비오틴 10 μgbiotin 10 μg

니코틴산아미드 1.7 mgNicotinamide 1.7 mg

엽산 50 μgfolic acid 50 μg

판토텐산 칼슘 0.5 mgCalcium Pantothenate 0.5 mg

무기질 혼합물 적량mineral mixture appropriate amount

황산제1철 1.75 mgferrous sulfate 1.75 mg

산화아연 0.82 mgzinc oxide 0.82 mg

탄산마그네슘 25.3 mgmagnesium carbonate 25.3 mg

제1인산칼륨 15 mgmonobasic potassium phosphate 15 mg

제2인산칼슘 55 mgDibasic Calcium Phosphate 55 mg

구연산칼륨 90 mgPotassium Citrate 90 mg

탄산칼슘 100 mgcalcium carbonate 100 mg

염화마그네슘 24.8 mgmagnesium chloride 24.8 mg

상기의 비타민 및 미네랄 혼합물의 조성비는 비교적 건강기능성 식품에 적합한 성분을 바람직한 실시예로 혼합 조성하였지만, 그 배합비를 임의로 변형 실시하여도 무방하며, 통상의 건강기능성 식품 제조방법에 따라 상기의 성분을 혼합한 다음, 과립을 제조하고, 통상의 방법에 따라 건강기능성 식품 조성물 제조에 사용할 수 있다.The composition ratio of the vitamin and mineral mixture is relatively suitable for health functional food, but the composition is mixed in a preferred embodiment, but the mixing ratio may be arbitrarily modified, and the above ingredients are mixed according to a conventional health functional food manufacturing method. Then, the granules can be prepared and used in the preparation of a health functional food composition according to a conventional method.

<건강기능식품 제조예 2> 건강 기능 음료의 제조<Health functional food manufacturing example 2> Manufacture of health functional beverage

유효물질 100 mgactive substance 100 mg

구연산 100 mgcitric acid 100 mg

올리고당 100 mgoligosaccharide 100 mg

매실농축액 2 mgPlum Concentrate 2 mg

타우린 100 mgTaurine 100 mg

정제수를 가하여 전체 500 mLPurified water is added to 500 mL

통상의 건강음료 제조방법에 따라 상기의 성분을 혼합한 다음, 약 1시간 동안 85℃에서 교반 가열한 후, 만들어진 용액을 여과하여 멸균된 1 용기에 취득하여 밀봉 멸균한 뒤 냉장 보관한 다음 본 발명의 건강음료 조성물 제조에 사용한다. 상기 조성비는 비교적 기호 음료에 적합한 성분을 바람직한 실시예로 혼합 조성하였지만, 수요계층, 수요국가, 사용 용도 등 지역적, 민족적 기호도에 따라서 그 배합비를 임의로 변형 실시하여도 무방하다.After mixing the above ingredients according to a conventional health drink manufacturing method, after stirring and heating at 85° C. for about 1 hour, the resulting solution is filtered and obtained in one sterilized container, sealed and sterilized, and then refrigerated. used in the manufacture of health beverage compositions of Although the composition ratio is a composition that is relatively suitable for a beverage of preference in a preferred embodiment, the mixing ratio may be arbitrarily modified according to regional and ethnic preferences such as demand class, demand country, and use purpose.

건강식품의 제조예Manufacturing example of health food

본 발명에 따른 유효물질은 목적에 따라 여러 형태의 건강식품으로 제조 가능하다. 하기는 본 발명에 따른 유효물질을 활성성분으로 함유시킨 몇몇 건강식품의 제조방법을 예시한 것으로 본 발명이 이에 한정되는 것은 아니다.The active substance according to the present invention can be manufactured into various types of health food depending on the purpose. The following exemplifies the manufacturing method of several health foods containing the active ingredient according to the present invention as an active ingredient, but the present invention is not limited thereto.

<건강식품 제조예 1> 유제품(dairy products)의 제조<Health food production example 1> Production of dairy products

본 발명의 유효물질 0.01-1 중량부를 우유에 첨가하고, 상기 우유를 이용하여 버터 및 아이스크림과 같은 다양한 유제품을 제조하였다.0.01-1 parts by weight of the active substance of the present invention was added to milk, and various dairy products such as butter and ice cream were prepared using the milk.

<건강식품 제조예 2> 선식의 제조<Health food production example 2> Preparation of wire

현미, 보리, 찹쌀, 율무를 공지의 방법으로 알파화시켜 건조시킨 것을 배전한 후 분쇄기로 입도 60 메쉬의 분말로 제조하였다. 검정콩, 검정깨, 들깨도 공지의 방법으로 쪄서 건조시킨 것을 배전한 후 분쇄기로 입도 60 메쉬의 분말로 제조하였다. 본 발명의 유효물질을 진공 농축기에서 감압농축하고 건조분말을 얻었다. 상기에서 제조한 곡물류, 종실류 및 유효물질의 건조분말을 다음의 비율로 배합하여 제조하였다.Brown rice, barley, glutinous rice, and barley radish were pregelatinized by a known method and dried, and then roasted and prepared as a powder having a particle size of 60 mesh with a grinder. Black soybeans, black sesame, and perilla were also steamed and dried by a known method, and then roasted and prepared into powder having a particle size of 60 mesh with a grinder. The active material of the present invention was concentrated under reduced pressure in a vacuum concentrator to obtain a dry powder. The above-prepared grains, seeds, and dry powders of active substances were prepared by blending them in the following ratios.

곡물류(현미 34 중량부, 율무 19 중량부, 보리 20 중량부),Grains (34 parts by weight of brown rice, 19 parts by weight of barley, 20 parts by weight of barley),

종실류(들깨 7 중량부, 검정콩 8 중량부, 검정깨 7 중량부),Seeds (7 parts by weight of perilla, 8 parts by weight of black beans, 7 parts by weight of black sesame),

유효물질 (2 중량부),active substance (2 parts by weight),

영지(1.5 중량부), 및Reishi (1.5 parts by weight), and

지황(1.5 중량부).Rehmannia (1.5 parts by weight).

Claims (16)

하기 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염:
[화학식 A]
Figure pat00040

(상기 화학식 A에 있어서,
상기 p38 Ligand Moiety는
Figure pat00041
이고;
상기 E3 Ligand Moiety는
Figure pat00042
이고;
상기 Linker는
Figure pat00043
,
Figure pat00044
,
Figure pat00045
,
Figure pat00046
,
Figure pat00047
또는
Figure pat00048
이고,
상기 n 및 m은 독립적으로 1 내지 10의 정수이고,
상기 a 및 b는 독립적으로 1 내지 10의 정수이다).
A compound represented by the following formula (A) or a pharmaceutically acceptable salt thereof:
[Formula A]
Figure pat00040

(In the formula A,
The p38 Ligand Moiety is
Figure pat00041
ego;
The E3 Ligand Moiety is
Figure pat00042
ego;
The Linker is
Figure pat00043
,
Figure pat00044
,
Figure pat00045
,
Figure pat00046
,
Figure pat00047
or
Figure pat00048
ego,
Wherein n and m are independently integers from 1 to 10,
wherein a and b are independently integers from 1 to 10).
제1항에 있어서,
상기 n은 1 내지 5의 정수이고,
상기 m은 3 내지 7의 정수이며,
상기 a는 1 내지 5의 정수이고,
상기 b는 1 내지 7의 정수인 것을 특징으로 하는 화합물 또는 이의 약학적으로 허용가능한 염.
According to claim 1,
Wherein n is an integer from 1 to 5,
Wherein m is an integer of 3 to 7,
wherein a is an integer from 1 to 5,
wherein b is an integer of 1 to 7, or a pharmaceutically acceptable salt thereof.
제2항에 있어서,
상기 n은 1 내지 3의 정수이고,
상기 m은 5이며,
상기 a는 1 내지 2의 정수이고,
상기 b는 2 또는 5인 것을 특징으로 하는 화합물 또는 이의 약학적으로 허용가능한 염.
3. The method of claim 2,
Wherein n is an integer of 1 to 3,
wherein m is 5,
wherein a is an integer of 1 to 2,
wherein b is 2 or 5, or a pharmaceutically acceptable salt thereof.
제1항에 있어서,
상기 화학식 A로 표시되는 화합물은 하기 화합물군으로부터 선택되는 1종인 것을 특징으로 하는 화합물 또는 이의 약학적으로 허용가능한 염.
1) 2-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)에틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (23);
2) 2-(2-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)에톡시)에틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (24);
3) 2-(2-(2-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)에톡시)에톡시)에틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (25);
4) 5-(4-(5'-(사이클로프로필카바모일)-2'-메틸-[1,1'-바이페닐]-4-카보닐)페녹시)펜틸(2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)카바메이트 (26);
5) N-사이클로프로필-4'-(4-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (31);
6) N-사이클로프로필-4'-(4-(2-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)에톡시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (32);
7) N-사이클로프로필-4'-(4-(2-(2-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)에톡시)에톡시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (33);
8) N-사이클로프로필-4'-(4-((5-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)펜틸)옥시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카복스아미드 (34); 및
9) (S)-N-사이클로프로필-4'-(4-(2-(2-(2-((2-(2,6-다이옥소피페리딘-3-일)-1,3-다이옥소이소인돌린-4-일)옥시)아세트아미도)에톡시)에톡시)벤조일)-6-메틸-[1,1'-바이페닐]-3-카르복스아미드 (41).
According to claim 1,
The compound represented by Formula A is a compound or a pharmaceutically acceptable salt thereof, characterized in that one selected from the following compound group.
1) 2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethyl (2-(2,6- dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (23);
2) 2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy)ethyl (2 -(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (24);
3) 2-(2-(2-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)ethoxy) ethoxy)ethyl(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (25);
4) 5-(4-(5'-(cyclopropylcarbamoyl)-2'-methyl-[1,1'-biphenyl]-4-carbonyl)phenoxy)pentyl(2-(2,6- dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)carbamate (26);
5) N-cyclopropyl-4'-(4-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy) ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (31);
6) N-cyclopropyl-4'-(4-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl) )oxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (32);
7) N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-) 4-yl)oxy)ethoxy)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (33);
8) N-cyclopropyl-4'-(4-((5-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)oxy )pentyl)oxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (34); and
9) (S)-N-cyclopropyl-4'-(4-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-diox) Soisoindolin-4-yl)oxy)acetamido)ethoxy)ethoxy)benzoyl)-6-methyl-[1,1′-biphenyl]-3-carboxamide (41).
하기 반응식 1에 나타난 바와 같이,
화합물 8A와 화합물 13을 유기용매에서 반응시켜 화합물 14A를 합성하는 단계(단계 1);
화합물 14A와 4-니트로페닐클로로포르메이트를 유기용매에서 반응시켜 화합물 19A를 합성하는 단계(단계 2); 및
화합물 19A와 화합물 5를 유기용매에서 반응시켜 화합물 A1을 합성하는 단계(단계 3);를 포함하는 화합물 A1의 제조방법:
[반응식 1]
Figure pat00049

(상기 반응식 1에서,
L1은 -(CH2)n-OH 또는 -(CH2CH2-O)n-CH2CH2OH이고, 여기서 상기 n은 1 내지 10의 정수이고;
L2는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).
As shown in Scheme 1 below,
synthesizing compound 14A by reacting compound 8A with compound 13 in an organic solvent (step 1);
synthesizing compound 19A by reacting compound 14A with 4-nitrophenylchloroformate in an organic solvent (step 2); and
A method of preparing compound A1 comprising: synthesizing compound A1 by reacting compound 19A with compound 5 in an organic solvent (step 3):
[Scheme 1]
Figure pat00049

(In Scheme 1,
L 1 is —(CH 2 ) n —OH or —(CH 2 CH 2 —O) n —CH 2 CH 2 OH, wherein n is an integer from 1 to 10;
L 2 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).
제5항에 있어서,
상기 단계 1 내지 단계 3의 유기용매는 독립적으로 DMF(dimethylformamide), DCM(dichloromethane), 에탄올, 테트라하이드로퓨란(THF), 벤젠, KOH/MeOH, MeOH, 톨루엔, 디이소프로필에테르, 디에틸에테르, 디옥산, 아세토니트릴, 디메틸아세트아미드(DMA), 디메틸설폭사이드(DMSO), 아세톤 및 클로로벤젠으로 이루어지는 군으로부터 선택되는 1종 이상인 것을 특징으로 하는 제조방법.
6. The method of claim 5,
The organic solvent of steps 1 to 3 is independently DMF (dimethylformamide), DCM (dichloromethane), ethanol, tetrahydrofuran (THF), benzene, KOH / MeOH, MeOH, toluene, diisopropyl ether, diethyl ether, Dioxane, acetonitrile, dimethyl acetamide (DMA), dimethyl sulfoxide (DMSO), acetone, and a manufacturing method, characterized in that at least one selected from the group consisting of chlorobenzene.
하기 반응식 2에 나타난 바와 같이,
화합물 8A와 화합물 13을 유기용매에서 반응시켜 화합물 14A를 합성하는 단계(단계 1);
화합물 14A와 p-TsCl(p-Toluenesulfonyl chloride)를 유기용매에서 반응시켜 화합물 27A를 합성하는 단계(단계 2); 및
화합물 27A와 화합물 6을 유기용매에서 반응시켜 화합물 A2를 합성하는 단계(단계 3);를 포함하는 화합물 A2의 제조방법:
[반응식 2]
Figure pat00050

(상기 반응식 2에서,
L1은 -(CH2)n-OH 또는 -(CH2CH2-O)n-CH2CH2OH이고, 여기서 상기 n은 1 내지 10의 정수이고;
L2는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).
As shown in Scheme 2 below,
synthesizing compound 14A by reacting compound 8A with compound 13 in an organic solvent (step 1);
synthesizing compound 27A by reacting compound 14A with p-TsCl (p-Toluenesulfonyl chloride) in an organic solvent (step 2); and
A method of preparing compound A2 comprising: synthesizing compound A2 by reacting compound 27A with compound 6 in an organic solvent (step 3):
[Scheme 2]
Figure pat00050

(In Scheme 2,
L 1 is —(CH 2 ) n —OH or —(CH 2 CH 2 —O) n —CH 2 CH 2 OH, wherein n is an integer from 1 to 10;
L 2 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).
제 7항에 있어서,
상기 단계 1 내지 단계 3의 유기용매는 독립적으로 DMF(dimethylformamide), DCM(dichloromethane), 에탄올, 테트라하이드로퓨란(THF), 벤젠, KOH/MeOH, MeOH, 톨루엔, 디이소프로필에테르, 디에틸에테르, 디옥산, 아세토니트릴, 디메틸아세트아미드(DMA), 디메틸설폭사이드(DMSO), 아세톤 및 클로로벤젠으로 이루어지는 군으로부터 선택되는 1종 이상인 것을 특징으로 하는 제조방법.
8. The method of claim 7,
The organic solvent of steps 1 to 3 is independently DMF (dimethylformamide), DCM (dichloromethane), ethanol, tetrahydrofuran (THF), benzene, KOH / MeOH, MeOH, toluene, diisopropyl ether, diethyl ether, Dioxane, acetonitrile, dimethyl acetamide (DMA), dimethyl sulfoxide (DMSO), acetone, and a manufacturing method, characterized in that at least one selected from the group consisting of chlorobenzene.
하기 반응식 3에 나타난 바와 같이,
화합물 14와 화합물 36A를 유기용매에서 반응시켜 화합물 37A를 합성하는 단계(단계 1);
화합물 37A와 염산을 유기용매에서 반응시켜 화합물 38A를 합성하는 단계(단계 2); 및
화합물 38A와 화합물 40을 유기용매에서 반응시켜 화합물 A3을 합성하는 단계(단계 3);를 포함하는 화합물 A3의 제조방법:
[반응식 3]
Figure pat00051

(상기 반응식 3에서,
L3은 -(CH2)n-OMs 또는 -(CH2CH2-O)n-CH2CH2OMs이고, 여기서 상기 n은 1 내지 10의 정수이고;
L4는 -(CH2)m-O- 또는 -(CH2CH2-O)m-이고, 여기서 상기 m은 1 내지 10의 정수이다).
As shown in Scheme 3 below,
synthesizing compound 37A by reacting compound 14 with compound 36A in an organic solvent (step 1);
synthesizing compound 38A by reacting compound 37A with hydrochloric acid in an organic solvent (step 2); and
A method of preparing compound A3 comprising: synthesizing compound A3 by reacting compound 38A with compound 40 in an organic solvent (step 3):
[Scheme 3]
Figure pat00051

(In Scheme 3,
L 3 is -(CH 2 ) n -OMs or -(CH 2 CH 2 -O) n -CH 2 CH 2 OMs, wherein n is an integer from 1 to 10;
L 4 is -(CH 2 ) m -O- or -(CH 2 CH 2 -O) m -, wherein m is an integer from 1 to 10).
제9항에 있어서,
상기 단계 1 내지 단계 3의 유기용매는 독립적으로 DMF(dimethylformamide), DCM(dichloromethane), 에탄올, 테트라하이드로퓨란(THF), 벤젠, KOH/MeOH, MeOH, 톨루엔, 디이소프로필에테르, 디에틸에테르, 디옥산, 아세토니트릴, 디메틸아세트아미드(DMA), 디메틸설폭사이드(DMSO), 아세톤 및 클로로벤젠으로 이루어지는 군으로부터 선택되는 1종 이상인 것을 특징으로 하는 제조방법.
10. The method of claim 9,
The organic solvent of steps 1 to 3 is independently DMF (dimethylformamide), DCM (dichloromethane), ethanol, tetrahydrofuran (THF), benzene, KOH / MeOH, MeOH, toluene, diisopropyl ether, diethyl ether, Dioxane, acetonitrile, dimethyl acetamide (DMA), dimethyl sulfoxide (DMSO), acetone, and a manufacturing method, characterized in that at least one selected from the group consisting of chlorobenzene.
제1항의 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 치료용 약학적 조성물.
A pharmaceutical composition for the prevention or treatment of chronic inflammatory diseases, comprising the compound represented by Formula A of claim 1 or a pharmaceutically acceptable salt thereof.
제11항에 있어서,
상기 만성 염증성 질환은 알츠하이머, 파킨슨 병, 염증성 장질환, 비알콜성 만성 간염, 만성 간염, 크론병, 췌장염, 식도염, 위염, 대장염, 궤양성 대장염, 폐렴, 기관지염, 인후염, 심근경색, 심부전, 관절염, 건선성 관절염, 류마티스 관절염, 신부전, 건선, 빈혈, 당뇨, 섬유화증, 다발성 경화증, 전신 홍반 루프스, 강직성 척추염, 천식, 만성폐쇄성폐질환, 치주염, 신경병증 통증 및 특발성 염증성 근육병증으로 이루어지는 군으로부터 선택되는 질환인 것을 특징으로 하는 약학적 조성물.
12. The method of claim 11,
The chronic inflammatory disease is Alzheimer's, Parkinson's disease, inflammatory bowel disease, non-alcoholic chronic hepatitis, chronic hepatitis, Crohn's disease, pancreatitis, esophagitis, gastritis, colitis, ulcerative colitis, pneumonia, bronchitis, sore throat, myocardial infarction, heart failure, arthritis , psoriatic arthritis, rheumatoid arthritis, renal failure, psoriasis, anemia, diabetes, fibrosis, multiple sclerosis, systemic lupus erythematosus, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, periodontitis, neuropathic pain and idiopathic inflammatory myopathy. A pharmaceutical composition, characterized in that the selected disease.
제1항의 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 개선용 건강기능식품 조성물.
A health functional food composition for the prevention or improvement of chronic inflammatory diseases comprising the compound represented by Formula A of claim 1 or a pharmaceutically acceptable salt thereof.
제13항에 있어서,
상기 만성 염증성 질환은 알츠하이머, 파킨슨 병, 염증성 장질환, 비알콜성 만성 간염, 만성 간염, 크론병, 췌장염, 식도염, 위염, 대장염, 궤양성 대장염, 폐렴, 기관지염, 인후염, 심근경색, 심부전, 관절염, 건선성 관절염, 류마티스 관절염, 신부전, 건선, 빈혈, 당뇨, 섬유화증, 다발성 경화증, 전신 홍반 루프스, 강직성 척추염, 천식, 만성폐쇄성폐질환, 치주염, 신경병증 통증 및 특발성 염증성 근육병증으로 이루어지는 군으로부터 선택되는 질환인 것을 특징으로 하는 건강기능식품 조성물.
14. The method of claim 13,
The chronic inflammatory disease is Alzheimer's, Parkinson's disease, inflammatory bowel disease, non-alcoholic chronic hepatitis, chronic hepatitis, Crohn's disease, pancreatitis, esophagitis, gastritis, colitis, ulcerative colitis, pneumonia, bronchitis, sore throat, myocardial infarction, heart failure, arthritis , psoriatic arthritis, rheumatoid arthritis, renal failure, psoriasis, anemia, diabetes, fibrosis, multiple sclerosis, systemic lupus erythematosus, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, periodontitis, neuropathic pain and idiopathic inflammatory myopathy. Health functional food composition, characterized in that the selected disease.
제1항의 화학식 A로 표시되는 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 만성 염증성 질환의 예방 또는 개선용 건강식품 조성물.
A health food composition for the prevention or improvement of chronic inflammatory diseases comprising the compound represented by Formula A of claim 1 or a pharmaceutically acceptable salt thereof.
제15항에 있어서,
상기 만성 염증성 질환은 알츠하이머, 파킨슨 병, 염증성 장질환, 비알콜성 만성 간염, 만성 간염, 크론병, 췌장염, 식도염, 위염, 대장염, 궤양성 대장염, 폐렴, 기관지염, 인후염, 심근경색, 심부전, 관절염, 건선성 관절염, 류마티스 관절염, 신부전, 건선, 빈혈, 당뇨, 섬유화증, 다발성 경화증, 전신 홍반 루프스, 강직성 척추염, 천식, 만성폐쇄성폐질환, 치주염, 신경병증 통증 및 특발성 염증성 근육병증으로 이루어지는 군으로부터 선택되는 질환인 것을 특징으로 하는 건강식품 조성물.
16. The method of claim 15,
The chronic inflammatory disease is Alzheimer's, Parkinson's disease, inflammatory bowel disease, non-alcoholic chronic hepatitis, chronic hepatitis, Crohn's disease, pancreatitis, esophagitis, gastritis, colitis, ulcerative colitis, pneumonia, bronchitis, sore throat, myocardial infarction, heart failure, arthritis , psoriatic arthritis, rheumatoid arthritis, renal failure, psoriasis, anemia, diabetes, fibrosis, multiple sclerosis, systemic lupus erythematosus, ankylosing spondylitis, asthma, chronic obstructive pulmonary disease, periodontitis, neuropathic pain and idiopathic inflammatory myopathy. A health food composition, characterized in that the disease is selected.
KR1020200034179A 2020-03-20 2020-03-20 p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same KR102624481B1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
KR1020200034179A KR102624481B1 (en) 2020-03-20 2020-03-20 p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same
PCT/KR2021/002361 WO2021187766A1 (en) 2020-03-20 2021-02-25 Compound having p38 removal capability, preparation method therefor and composition for treating chronic inflammatory disease, comprising same

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020200034179A KR102624481B1 (en) 2020-03-20 2020-03-20 p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same

Publications (2)

Publication Number Publication Date
KR20210118297A true KR20210118297A (en) 2021-09-30
KR102624481B1 KR102624481B1 (en) 2024-01-16

Family

ID=77771327

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020200034179A KR102624481B1 (en) 2020-03-20 2020-03-20 p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same

Country Status (2)

Country Link
KR (1) KR102624481B1 (en)
WO (1) WO2021187766A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023113457A1 (en) * 2021-12-14 2023-06-22 (주)프레이저테라퓨틱스 Novel compound for degrading target protein or polypeptide by using polyubiquitination

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150291562A1 (en) * 2014-04-14 2015-10-15 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
KR20170025603A (en) * 2015-08-31 2017-03-08 경희대학교 산학협력단 Benzophenone derivatives, preparation method thereof and pharmaceutical composition for anti-inflammatory comprising the same

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL1687284T3 (en) * 2003-07-25 2009-04-30 Novartis Ag p-38 KINASE INHIBITORS
EP2846784A4 (en) * 2012-05-11 2016-03-09 Univ Yale Compounds useful for promoting protein degradation and methods using same
AU2016301195B2 (en) * 2015-08-06 2022-09-01 Dana-Farber Cancer Institute, Inc. Targeted protein degradation to attenuate adoptive T-cell therapy associated adverse inflammatory responses

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150291562A1 (en) * 2014-04-14 2015-10-15 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
KR20170025603A (en) * 2015-08-31 2017-03-08 경희대학교 산학협력단 Benzophenone derivatives, preparation method thereof and pharmaceutical composition for anti-inflammatory comprising the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NATURE REVIEWS, Drug discovery (2003), Vol.2, 717-726

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023113457A1 (en) * 2021-12-14 2023-06-22 (주)프레이저테라퓨틱스 Novel compound for degrading target protein or polypeptide by using polyubiquitination

Also Published As

Publication number Publication date
WO2021187766A1 (en) 2021-09-23
KR102624481B1 (en) 2024-01-16

Similar Documents

Publication Publication Date Title
JP6944462B2 (en) Solid form of thienopyrimidinedione ACC inhibitor and methods for its production
JP3794618B2 (en) Aryl-substituted propanolamine derivative, process for producing the same, medicament containing the compound and use thereof
CN105198804B (en) Compound as hiv integrase inhibitor
CN104230865B (en) 4 amino butyric acid derivatives of biaryl substituted and its production and use
CN112672995A (en) Novel sulfonamide urea compounds
JP2022505525A (en) Compounds and compositions for treating conditions associated with NLRP activity
CN113286794A (en) KRAS mutein inhibitors
CN102712628A (en) Hepatitis c virus inhibitors
AU2013272429B2 (en) Pharmaceutical composition containing verbenone derivative for treating or preventing neurodegenerative disease
KR102624481B1 (en) p38 degrader compounds, method for preparing the same, and composition for treating chronic inflammatory diseases including the same
KR20230021083A (en) immunomodulator
KR101845203B1 (en) Lauric acid derivatives, preparation method thereof and anticancer agent comprising the same
KR102022279B1 (en) A composition comprising an extract of Angelica keiskei for treating and preventing muscle-related disorder
JP2013053115A (en) METHOD OF PRODUCING α CYCLIC DIPEPTIDE
JP2013542183A (en) Sesterterpene compounds and uses of these substances
WO2022138735A1 (en) Pharmaceutical composition for ameliorating malignant diseases
WO2022271878A1 (en) 4-ethynyl-3-hydroxy-tetrahydrofuranyl-adenine phosphoramidates and related compounds and their use in treating medical conditions
KR20190112492A (en) 1,2-diacylglycerol compound, method for preparing the same and immunomodulating agent including the same as active ingredient
KR102240948B1 (en) Amino acid derivatives, preparation method thereof and pharmaceutical composition for treatment of hepatitis B virus comprising the same
EP4026562A1 (en) Chronic kidney disease treatment or prevention method
EP3880668B1 (en) Pyridin-sulfonamide compounds for the treatment of conditions related to interleukin 1 beta
JP2022529855A (en) Derivatives of glycero-mannno-heptose phosphate and their use in modulating the immune response
KR101628933B1 (en) Benzidine derivative, preparation method thereof and pharmaceutical composition for treating liver disease related with Hepatitis C virus containing the same
KR101493882B1 (en) Novel heteroarylcarboxamide derivative or pharmacutically acceptable salt thereof, process for the preparation thereof and pharmaceutical composition for prevention or treatment of RAGE receptor related diseases containing the same as an active ingredient
KR102040119B1 (en) A composition comprising the isolated compounds from an extract of Angelica keiskei for treating and preventing muscle-related disorder

Legal Events

Date Code Title Description
E902 Notification of reason for refusal
E701 Decision to grant or registration of patent right
GRNT Written decision to grant