KR102116166B1 - Nlrp3 inhibitor - Google Patents

Nlrp3 inhibitor Download PDF

Info

Publication number
KR102116166B1
KR102116166B1 KR1020170121040A KR20170121040A KR102116166B1 KR 102116166 B1 KR102116166 B1 KR 102116166B1 KR 1020170121040 A KR1020170121040 A KR 1020170121040A KR 20170121040 A KR20170121040 A KR 20170121040A KR 102116166 B1 KR102116166 B1 KR 102116166B1
Authority
KR
South Korea
Prior art keywords
nlrp3
dihydro
benzo
cyclohexyl
formula
Prior art date
Application number
KR1020170121040A
Other languages
Korean (ko)
Other versions
KR20190032790A (en
Inventor
이광호
심도완
김병학
예상규
강태봉
Original Assignee
건국대학교 글로컬산학협력단
서울대학교산학협력단
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 건국대학교 글로컬산학협력단, 서울대학교산학협력단 filed Critical 건국대학교 글로컬산학협력단
Priority to KR1020170121040A priority Critical patent/KR102116166B1/en
Publication of KR20190032790A publication Critical patent/KR20190032790A/en
Application granted granted Critical
Publication of KR102116166B1 publication Critical patent/KR102116166B1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/39Heterocyclic compounds having sulfur as a ring hetero atom having oxygen in the same ring
    • 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
    • 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
    • A23V2200/324Foods, ingredients or supplements having a functional effect on health having an effect on the immune system

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

본 발명은 NLRP3 억제제로서의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온(2-cyclohexylimino-6-methyl-6,7-dihydro-5H-benzo[1,3]oxathiol-4-one)의 용도에 관한 것으로, 본 발명에 따른 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온은 NLRP3를 알킬화함으로써 이의 ATPase 활성을 억제한 뒤 유비퀴틴화 시키고, 이로 인해 ASC와의 결합과 이동을 억제하여, NLRP3 염증조절복합체(inflammasome)의 활성화를 억제하는 것을 확인하였으며, 마우스에 유발된 복막염에 대한 치료 효과가 우수함을 확인하였으므로, 이를 NLRP3 알킬화제 및, NLRP3 또는 NLRP3 염증조절복합체를 억제하는 용도로 이용할 수 있고, 염증 질환 예방 또는 치료용 약제학적 조성물로도 유용하게 사용될 수 있다.The present invention is 2-cyclohexyl-diamino-6 inhibitor as NLRP3-6,7-dihydro -5 H - benzo [1,3] oxa thiol-4-one (2-cyclohexylimino-6-methyl -6, 7-dihydro-5 H -benzo [1,3] oxathiol-4-one) relates to the use of 2-cyclohexylimino-6-methyl-6,7-dihydro- of Formula 1 according to the present invention 5 H -benzo [1,3] oxathiol-4-one inhibits its ATPase activity by alkylating NLRP3 and then ubiquitinates it, thereby inhibiting binding and migration with ASC, thereby inhibiting the NLRP3 inflammation control complex (inflammasome). Since it was confirmed to suppress activation, and it was confirmed that the therapeutic effect on peritonitis caused by mice was excellent, it can be used for the purpose of suppressing the NLRP3 alkylating agent and the NLRP3 or NLRP3 inflammatory regulatory complex, and preventing or treating inflammatory diseases It can also be useful as a medical composition.

Description

NLRP3 억제제{NLRP3 INHIBITOR}NLRP3 inhibitor {NLRP3 INHIBITOR}

본 발명은 NLRP3 억제제로서의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온(2-cyclohexylimino-6-methyl-6,7-dihydro-5H-benzo[1,3]oxathiol-4-one)의 용도에 관한 것이다.The present invention is 2-cyclohexyl-diamino-6 inhibitor as NLRP3-6,7-dihydro -5 H - benzo [1,3] oxa thiol-4-one (2-cyclohexylimino-6-methyl -6, 7-dihydro-5 H- benzo [1,3] oxathiol-4-one).

선천성 면역 반응은 대식세포와 수지상 세포와 같은 선천성 면역 세포에 의해 조절되는 미생물에 대한 최초 방어선이다. 미생물에 감염되면 이들 선천성 면역 세포들은 감염성 미생물에 저항하기 위하여 면역 반응을 활성화시키는데, 선천성 면역 세포는 톨-유사 수용체(Toll-like receptors), NOD-유사 수용체(NOD-like receptors), RIG-I-유사 수용체(RIG-I-like receptors) 및 C-유형 렉틴 수용체(C-type lectin receptors)와 같은 패턴인식 수용체(pattern recognition receptors, PRRs)를 통하여 미생물을 인식한다. 이러한 패턴인식 수용체들은 병원균-연관 분자 패턴(athogen-associated molecular patterns, PAMPs)과 손상-연관 분자 패턴(damage-associated molecular patterns, DAMPs)과 같이 미생물 종 간에 보존적으로 존재하는 구조를 인식함으로써 미생물을 식별하고, 이에 따라 미생물, 병원균, 감염성 세포들을 제거하기 위한 면역 조절자들을 활성화시킨다. 이 중, NOD-유사 수용체(NOD-like receptors, NLRs)는 인간에서 20개 이상의 패밀리 멤버를 가지고 있는 세포내 면역 수용체 패밀리로, NLR 패밀리 멤버는 leucine-rich repeats (LRRs)와 NATCH nucleotide-binding omain (NBD)과 같은 적어도 2개 이상의 공통된 특징을 가지고 있는 것으로 알려져 있다. 이 중 LRR 도메인은 원균-연관 분자 패턴(PAMPs)를 인식하고, 단백질 간의 상호결합에 중요한 역할을 하는 것으로 알려져 있다. NOD-유사 수용체 패밀리 멤버는 상기와 같은 공통된 특징을 가지고 있으나, N-말단 도메인에는 차이가 있는 것으로 알려져 있으며, 그 차이에 근거하여 NOD-유사 수용체 패밀리 멤버가 세분화된다. 이중 NLRP는 Nterminal pyrin domain (PYD)을 가지고 있는 NLR 패밀리 중 가장 큰 그룹이고, NOD1(nucleotide-binding ligomerization domain-containing 1) (NLR1으로도 알려져 있음), NOD2 및 CARD domain-containing 4 NLRC4 (IPAF로도 알려져 있음)는 모두 N-terminal caspase recruitment domain을 가지고 있다.The innate immune response is the first line of defense against microorganisms that are regulated by innate immune cells, such as macrophages and dendritic cells. When infected with microorganisms, these innate immune cells activate an immune response to resist infectious microorganisms. Innate immune cells include Toll-like receptors, NOD-like receptors, and RIG-I Recognize microorganisms through pattern recognition receptors (PRRs), such as RIG-I-like receptors and C-type lectin receptors. These pattern recognition receptors recognize microorganisms by conserving structures that exist conservatively between microbial species, such as pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Identify and activate immune modulators to eliminate microbes, pathogens, and infectious cells accordingly. Among them, NOD-like receptors (NLRs) are intracellular immune receptor families with more than 20 family members in humans, and NLR family members are leucine-rich repeats (LRRs) and NATCH nucleotide-binding omain. It is known to have at least two or more common features such as (NBD). Among them, the LRR domain is known to recognize proto-associated molecular patterns (PAMPs) and play an important role in protein-to-protein interaction. The NOD-like receptor family members have the same common characteristics as above, but it is known that there is a difference in the N-terminal domain, and based on the difference, the NOD-like receptor family members are subdivided. Of these, NLRP is the largest group of NLR families with Nterminal pyrin domain (PYD), NOD1 (also known as nucleotide-binding ligomerization domain-containing 1), NOD2 and CARD domain-containing 4 NLRC4 (also known as IPAF) All known) have an N-terminal caspase recruitment domain.

한편, NOD-유사 수용체 패밀리 중 NLRP1, NLRP3 및 NLRC4을 가지고 있는 패밀리 멤버는 다양한 활성인자들에 의해 다분자 복합체를 형성할 수 있고, 이에 따라 면역 caspase들을 활성화시킬 수 있다고 알려져 있는데, 이중에서도 활성 caspase-1은 IL-1 패밀리의 사이토카인 분비를 조절한다. NOD-유사 수용체 다분자 복합체는 염증조절복합체(inflammasomes)라고도 불리는데, 이러한 염증조절복합체는 사이토졸 내의 면역 감시 체계를 위한 주요 구성요소에 해당한다. 염증조절복합체는 다단백질 복합체로써, 단백질 분해 효소인 caspase-1을 활성화시키고, IL-1β(interleukin-1β)와 IL-18과 같은 전-염증 사이토카인을 성숙시킨다. 이러한 활성화 반응과 함께, 염증조절복합체는 자가올리고화되고, NOD-유사 수용체와 pro-caspase-1의 CARDs 도메인의 직접적인 상호결합을 통해 pro-caspase-1을 리크루팅(recruiting)한다. 하지만, 많은 NOD-유사 수용체는 CARDs 도메인을 가지고 있지 않기 때문에, CARDs 또는 PYD 도메인을 포함하는 ASC(apoptotic speck protein containing a caspase recruitment domain; also called PYCARD)의 도움을 받게 되는데, 즉, ASC의 PYD 도메인은 NOD-유사 수용체와 상호작용하고, ASC의 CARD 도메인은 pro-caspase-1의 CARD 도메인과 상호작용하게 되는 것이다. Caspase-1은 IL-1-전환 효소로도 알려져 있는데, 이는 염증조절복합체를 형성한후, pro-caspase-1이 자가절단되면서 활성형 caspase-1이 형성되고, 활성형 caspase-1이 pro-IL-1β를 IL-1β로 성숙시켜, 피롭토시스(pyroptosis)라고 알려진 caspase-1 의존적인 세포 사멸을 유도하기 때문이다. caspase-1에 의해 성숙된 활성형 IL-1β는 잠재적인 전-염증 활성을 보유하고 있는데, IL-1β의 억제는 IL-1β에 의해 조절되는 죽상 동맥 경화증, 인슐린 저항성, 제2형 당뇨병, 류마티스성 관절염, 통풍 등의 자가면역질환을 감소시킨다고 알려져 있다. 따라서, IL-1β의 급격한 유출은 상당히 유해한 결과를 야기할 수 있으므로, IL-1β의 분비는 매우 엄격하게 조절되어야 할 것이다.On the other hand, family members having NLRP1, NLRP3 and NLRC4 among NOD-like receptor families are known to be able to form multi-molecular complexes by various active factors, thereby activating immune caspases. -1 regulates the cytokine secretion of the IL-1 family. NOD-like receptor multi-molecular complexes are also called inflammation control complexes (inflammasomes), which are key components for the immune surveillance system in cytosols. Inflammatory regulatory complexes are multiprotein complexes that activate caspase-1, a proteolytic enzyme, and mature pro-inflammatory cytokines such as IL-1β (interleukin-1β) and IL-18. Along with this activation reaction, the inflammatory regulatory complex is self-oligolysed and recruiting pro-caspase-1 through direct cross-linking of the NOD-like receptor and the CARDs domain of pro-caspase-1. However, since many NOD-like receptors do not have a CARDs domain, they are assisted by an apoptotic speck protein containing a caspase recruitment domain (ASC) containing CARDs or PYD domains, that is, the PYD domain of ASC. Interacts with the NOD-like receptor, and the CARD domain of ASC interacts with the CARD domain of pro-caspase-1. Caspase-1 is also known as an IL-1-converting enzyme, which forms an inflammatory regulatory complex, and then pro-caspase-1 self-cuts to form active caspase-1, and active caspase-1 is pro- This is because IL-1β matures into IL-1β and induces caspase-1 dependent cell death, known as pyroptosis. Active IL-1β matured by caspase-1 possesses potential pro-inflammatory activity, and inhibition of IL-1β is atherosclerosis regulated by IL-1β, insulin resistance, type 2 diabetes, rheumatism It is known to reduce autoimmune diseases such as sex arthritis and gout. Therefore, the rapid outflow of IL-1β can lead to fairly detrimental results, so the secretion of IL-1β will have to be very tightly controlled.

염증 질환 치료 관련 선행 특허로는 우산나물 추출물이 염증조절복합체 활성 조절을 통해 염증 매개인자의 생성을 감소시키는 것에 대한 대한민국 등록특허 (제10-1732365호), 청고추 추출물의 인플라마좀 매개 염증성 질환 개선용 조성물에 대한 대한민국 등록특허 (제10-1637476호) 등이 보고되어 있다.As a precedent patent related to the treatment of inflammatory diseases, the U.S. registered extract (No. 10-1732365) for reducing the production of inflammatory mediators by regulating the activity of the inflammatory control complex activity, inflammasome mediated inflammatory disease of green pepper extract Republic of Korea Patent Registration (No. 10-1637476) for the composition for improvement has been reported.

이에 본 발명자들은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온의 NLRP3(NACHT, LRR and PYD domains-containing protein 3) 염증조절복합체(inflammasome)에 대한 억제 효과를 확인하고, 이에 대한 억제 메카니즘을 확인함으로써 본 발명을 완성하였다.The present inventors have found that 2-cyclohexyl-6-a mino-6,7-dihydro -5 H - [1,3] oxazol-4-thiol of NLRP3 (NACHT, LRR and PYD domains -containing protein 3 ) The present invention was completed by confirming the inhibitory effect on the inflammatory regulatory complex (inflammasome) and confirming the inhibitory mechanism therefor.

본 발명에서는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 포함하는 NLRP3 활성 또는 NLRP3 염증조절복합체(inflammasome) 활성 억제용 시약조성물을 제공하는 것을 목적으로 한다.In the present invention, 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - benzo [1,3] NLRP3 active inflammation or NLRP3 control complex (inflammasome) activity comprising a thiol-oxa-4-one It is an object to provide a reagent composition for inhibition.

또한, 본 발명에서는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 포함하는 NLRP3 알킬화제를 제공하는 것을 목적으로 한다.In the present invention, 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - is an object of the present invention to provide a NLRP3 alkylating agent containing the benzo [1,3] oxa thiol-4-one .

또한, 본 발명에서는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 처리하는 단계를 포함하는, 시험관 내(in vitro)에서 NLRP3 활성 또는 NLRP3 염증조절복합체 활성을 억제하는 방법을 제공하는 것을 목적으로 한다.In the present invention, 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - [1,3], including the step of processing the oxa thiol-4-one in vitro (in It is an object to provide a method for inhibiting NLRP3 activity or NLRP3 inflammatory regulatory complex activity in vitro.

또한, 본 발명에서는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 유효성분으로 함유하는 염증 질환 예방 또는 치료용 약제학적 조성물을 제공하는 것을 목적으로 한다.In the present invention, 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - [1,3] for the prevention or treatment of inflammatory diseases containing oxa thiol-4-one as an active ingredient It is an object to provide a pharmaceutical composition.

아울러, 본 발명에서는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 함유하는 염증 질환의 예방 또는 개선용 식품 조성물을 제공하는 것을 목적으로 한다.In addition, in the present invention, 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - benzo [1, 3] a food composition for the prevention or amelioration of inflammatory disorders, containing a thiol-oxa-4-one It aims to provide.

상기 목적을 달성하기 위하여, 본 발명자들은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온 (또는 BOT-4-one으로 표기)의 NLRP3 활성 및 NLRP3 염증조절복합체(inflammasome)에 대한 효과와, 구체적인 작용 기작을 확인하였다.In order to achieve the above object, the present inventors have found that 2-cyclohexyl-6-a mino-6,7-dihydro -5 H - [1,3] oxazol-4-thiol (or BOT-4-one ) And the effect on NLRP3 activity and NLRP3 inflammatory regulatory complex (inflammasome) and specific action mechanism were confirmed.

본 발명에 따른 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온은 NLRP3를 알킬화함으로써 이의 ATPase 활성을 억제 및 유비퀴틴화를 증가시키고, 이로 인해 ASC의 이동과 결합을 억제하여, NLRP3 염증조절복합체(inflammasome)의 활성화를 억제하는 것을 확인하였으며, 마우스에 유발된 복막염에 대한 치료 효과가 우수함을 확인하였으므로, 이를 NLRP3 알킬화제 및 NLRP3 ATPase 활성 억제를 통한 NLRP3 염증조절복합체 억제용으로 이용할 수 있고, 이를 통한 염증 질환 예방 또는 치료용 약제학적 조성물로도 유용하게 사용될 수 있다.2-cyclohexyl-6-a mino-6,7-dihydro -5 H of the formula (I) according to the present invention benzo [1,3] oxa thiol-4-one inhibits the ATPase activity thereof, by alkylating the NLRP3 And increasing ubiquitination, thereby inhibiting the movement and binding of ASC, thereby inhibiting the activation of the NLRP3 inflammatory regulatory complex (inflammasome), and confirming that the therapeutic effect on peritonitis caused by the mouse is excellent. The NLRP3 alkylating agent and the NLRP3 ATPase can be used for suppressing the NLRP3 inflammatory regulatory complex through inhibitory activity, and it can also be usefully used as a pharmaceutical composition for preventing or treating inflammatory diseases.

도 1은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온의 농도별 BMDMs에 대한 세포 독성을 확인한 도이다:
Sup: 세포 상등액.
도 2는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온의 NLRP3 염증조절복합체 활성 억제 효과를 확인한 도이다:
BOT: 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온;
Nig: 니제리신(nigericin); 및
Sil: 실리카 크리스탈.
도 3은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온의 NLRP3 매개 ASC 올리고머화 억제 효과를 확인한 도이다:
Sup: 세포 상등액;
Pel: 펠릿 (가교 결합 된 세포질 펠렛); 및
Lys: 세포 파쇄물.
도 4는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온의 NLRP3 및 ASC의 Triton X-100 불용성 분획으로의 이동 억제 효과를 확인한 도이다:
Sup: 세포 상등액 (Triton X-100의 가용성 분획);
Pel: 펠릿 (Triton X-100의 불용성 분획); 및
Lys: 세포 파쇄물 (Triton X-100의 가용성 분획).
도 5는 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온의 NLRP3 ATPase 활성 억제 및 알킬화를 확인한 도이다:
Cys: L-시스테인; 및
Bay: Bay11-7082.
도 6은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온이 NLRP3 알킬화를 통해 ASC 올리고머화와 NLRP3/ASC의 이동을 억제하는 것을 확인한 도이다:
Cys: L-시스테인;
Sup: 세포 상등액 (Triton X-100의 가용성 분획);
Pel: 펠릿 (Triton X-100의 불용성 분획); 및
Lys: 세포 파쇄물 (Triton X-100의 가용성 분획).
도 7은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온이 NLRP3의 알킬화를 통해 NLRP3의 유비퀴틴화를 유발하고 이어서 NLRP3/ASC의 결합을 억제하는 것을 확인한 도이다.
도 8은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온의 in vivo에서 MSU-유발 복막염 완화 효과를 확인한 도이다.
Figure 1 is 2-cyclohexyl-6-a mino-6,7-dihydro -5 H - is a check the cytotoxicity of the concentrations BMDMs of benzo [1,3] oxa thiol-4-one:
Sup: Cell supernatant.
Figure 2 is a 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - is the benzo [1, 3] oxazol-thiol is also confirming the NLRP3 inflammation control complex activity inhibitory effect of the 4-one:
BOT: 2- cyclohexyl-6-butylimino-6,7-dihydro -5 H - benzo [1,3] oxa thiol-4-one;
Nig: nigericin; And
Sil: Silica crystal.
3 is a 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - is the benzo [1, 3] oxazol-thiol is also confirming the NLRP3 ASC oligomerization mediated inhibitory effect of 4-one:
Sup: cell supernatant;
Pel: pellets (crosslinked cytoplasmic pellets); And
Lys: cell debris.
Figure 4 is 2-cyclohexyl-6-a mino-6,7-dihydro -5 H - movement in the benzo [1,3] oxa thiol-4-one of NLRP3 and ASC of Triton X-100 insoluble fraction This is a diagram confirming the inhibitory effect:
Sup: cell supernatant (soluble fraction of Triton X-100);
Pel: pellet (insoluble fraction of Triton X-100); And
Lys: Cell lysate (soluble fraction of Triton X-100).
5 is a 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - is the benzo [1, 3] oxazol-thiol is also confirming the NLRP3 ATPase activity and inhibit the alkylation of 4-one:
Cys: L-cysteine; And
Bay: Bay11-7082.
Figure 6 is a 2-cyclohexyl-6-diamino-6,7-dihydro -5 H - [1,3] oxazol-thiol movement of ASC NLRP3 oligomerization and / ASC via the NLRP3 alkylated-4-one It is also confirmed to suppress:
Cys: L-cysteine;
Sup: cell supernatant (soluble fraction of Triton X-100);
Pel: pellet (insoluble fraction of Triton X-100); And
Lys: Cell lysate (soluble fraction of Triton X-100).
7 is a 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - induced ubiquitination of NLRP3 through benzo [1, 3] oxazol-4-thiol, followed by alkylation of the NLRP3 It is the figure which confirmed that it suppresses the binding of NLRP3 / ASC.
Figure 8 is 2-cyclohexyl-6-butylimino-6,7-dihydro -5 H - [1,3] is also confirming the MSU- peritonitis induced relaxation effect in the in vivo oxa thiol-4-one .

이하, 본 발명을 상세히 설명하기로 한다. 다만, 본 발명은 다양한 형태로 변경되어 구현될 수 있으며, 여기에서 설명하는 구현예에 한정되는 것은 아니다.Hereinafter, the present invention will be described in detail. However, the present invention can be implemented by being modified in various forms, and is not limited to the embodiments described herein.

일 측면에서, 본 발명은 하기 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온(2-cyclohexylimino-6-methyl-6,7-dihydro-5H-benzo[1,3]oxathiol-4-one, 본 명세서 내에서 BOT-4-one으로도 표기함)을 포함하는 NLRP3 활성 억제용 시약조성물에 관한 것이다:In one aspect, the present invention is 2-cyclohexyl-6,7-dihydro-6-butylimino of the following general formula (1) -5 H - [1,3] oxa thiol-4-one (2-cyclohexylimino-6 -methyl-6,7-dihydro-5 H -benzo [1,3] oxathiol-4-one, also referred to herein as BOT-4-one) relates to a reagent composition for inhibiting NLRP3 activity :

Figure 112017091645861-pat00001
Figure 112017091645861-pat00001

일 구현예에서, 상기 NLRP3 활성은 NLRP3의 ATPase 활성일 수 있으며, NLRP3을 유비퀴틴화(ubiquitination)하여 NLRP3의 활성을 억제할 수 있고, 이어서 NLRP3 및 ASC의 결합도 억제할 수 있다.In one embodiment, the NLRP3 activity may be the ATPase activity of NLRP3, ubiquitination of NLRP3 can inhibit the activity of NLRP3, and then the binding of NLRP3 and ASC can also be inhibited.

일 측면에서, 본 발명은 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 포함하는 NLRP3 염증조절복합체(inflammasome) 활성 억제용 시약조성물에 관한 것이다. In one aspect, the present invention is 2-cyclohexyl-6-a mino-6,7-dihydro -5 H of the formula (1) benzo [1,3] NLRP3 inflammation control complex including a oxazole-4-thiol (inflammasome) Reagent composition for inhibiting activity .

일 구현예에서, 본 발명의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온은 ASC 올리고머화와 NLRP3 및 ASC의 Triton X-100 불용성 분획으로의 이동을 억제하고, NLRP3 및 ASC(apoptotic speck protein containing a caspase recruitment domain)의 결합을 억제함으로써 NLRP3 염증조절복합체의 활성을 억제할 수 있다.In one embodiment, the present invention is 2-cyclohexyl-6-diamino-6,7-dihydro -5 H - benzo [1,3] oxa thiol-4-one is in the ASC and ASC oligomerization and NLRP3 By inhibiting the migration to the Triton X-100 insoluble fraction, and inhibiting the binding of NLRP3 and the ASC (apoptotic speck protein containing a caspase recruitment domain), it is possible to inhibit the activity of the NLRP3 inflammatory regulatory complex.

일 측면에서, 본 발명은 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 포함하는 NLRP3 알킬화제에 관한 것이다.In one aspect, the present invention is a 2-cyclohexyl-6-methyl-6,7-diamino of formula dihydro -5 H - [1,3] on the NLRP3 alkylating agent containing the thiol-oxa-4-one will be.

일 구현예에서, 본 발명의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온은 NLRP3를 알킬화함으로써 NLRP3의 ATPase 활성을 억제하고, 유비퀴틴화(ubiquitination)시켜 NLRP3의 활성을 억제하며, 이를 통해 ASC 올리고머화와 NLRP3/ASC의 결합 및 이동을 억제하여 NLRP3 염증조절복합체를 억제하고, 이로 인한 인터루킨-1베타의 성숙 및 분비를 억제할 수 있다. In one implementation, the 2-cyclohexyl-6-a mino-6,7-dihydro -5 H of the present invention benzo [1,3] oxa thiol-4-one is NLRP3 of ATPase activity by alkylating the NLRP3 And inhibits the activity of NLRP3 by ubiquitination, thereby inhibiting the binding and migration of ASC oligomerization and NLRP3 / ASC, thereby inhibiting the NLRP3 inflammatory regulatory complex, resulting in maturation and interleukin-1 beta maturation and Secretion can be suppressed.

일 실시예에서, 본 발명의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온이 NLRP3의 활성을 조절하는 기작이 알킬화에 의해서임을 확인하였으므로, 이를 NLRP3 알킬화제로 이용할 수 있다.In one embodiment, the 2-cyclohexyl-6 of the present invention diamino-6,7-dihydro -5 H - a mechanism for controlling the benzo [1,3] oxazole The thiol NLRP3 activity of the 4-one Since it was confirmed by alkylation, it can be used as an NLRP3 alkylating agent.

일 측면에서, 본 발명은 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 처리하는 단계를 포함하는 시험관 내(in vitro) NLRP3 염증조절복합체 활성을 억제하는 방법에 관한 것이다.In one aspect, the present invention is a 2-cyclohexyl-6-methyl-6,7-diamino of formula dihydro -5 H - comprising the step of treating the benzo [1,3] oxa thiol-4-one It relates to a method for inhibiting NLRP3 inflammation control complex activity in vitro.

일 실시예에서, 본 발명의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온이 in vitro 상에서 NLRP3 염증조절복합체의 활성화를 억제하는 것을 확인하였으므로, 이를 이용하여 시험관 내에서 NLRP3 염증조절복합체의 활성을 억제할 수 있다.In one embodiment, the present invention is 2-cyclohexyl-6-diamino-6,7-dihydro -5 H - of benzo [1, 3] oxazol-4-thiol NLRP3 inflammation control complex on the in vitro Since it was confirmed to inhibit activation, it can be used to inhibit the activity of the NLRP3 inflammatory regulatory complex in vitro.

일 측면에서, 본 발명은 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 처리하는 단계를 포함하는, 시험관 내에서 NLRP3 활성을 억제하는 방법에 관한 것이다.In one aspect, the present invention is a 2-cyclohexyl-6-methyl-6,7-diamino of formula dihydro -5 H - comprising the step of treating the benzo [1,3] oxa thiol-4-one , It relates to a method for inhibiting NLRP3 activity in vitro.

일 실시예에서, 본 발명의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온이 in vitro 상에서 NLRP3 활성을 억제하는 것을 확인하였으므로, 이를 이용하여 시험관 내에서 NLRP3 활성을 억제할 수 있다.In one embodiment, the 2-cyclohexyl-6 of the present invention diamino-6,7-dihydro -5 H - NLRP3 of inhibiting activity on the benzo [l, 3] The in vitro oxa thiol-4-one Since it was confirmed that it can be used to suppress NLRP3 activity in vitro.

일 측면에서 본 발명은 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온(2-cyclohexylimino-6-methyl-6,7-dihydro-5H-benzo[1,3]oxathiol-4-one)을 유효성분으로 함유하는 염증 질환 예방 또는 치료용 약제학적 조성물에 관한 것이다.In one aspect the present invention is a 2-cyclohexyl-6-methyl-6,7-diamino of formula dihydro -5 H - benzo [1,3] oxa thiol-4-one (2-cyclohexylimino-6-methyl -6,7-dihydro-5 H -benzo [ 1,3] oxathiol-4-one) the present invention relates to inflammatory disease prevention or treatment a pharmaceutical composition containing as an active ingredient.

일 구현예에서, 본 발명의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온은 하기 특성 중 어느 하나 이상을 가질 수 있다:In one embodiment, the 2-cyclohexyl-6 of the present invention diamino-6,7-dihydro -5 H - have any one or more of the following properties [1,3] oxa thiol-4-one Can:

1) NLRP3 활성 억제;1) inhibition of NLRP3 activity;

2) NLRP3 염증조절복합체 활성 억제;2) inhibition of NLRP3 inflammatory regulatory complex activity;

3) NLRP3 및 ASC의 결합 및 이동 억제; 및3) NLRP3 and ASC binding and migration inhibition; And

4) IL-1β 성숙 및 분비 억제.4) IL-1β maturation and secretion inhibition.

일 구현예에서, 상기 염증 질환은 염증조절복합체(inflammasome) 매개 염증성 질환일 수 있다.In one embodiment, the inflammatory disease may be an inflammatory disease-mediated inflammatory disease.

일 구현예에서, 상기 염증 질환은 복막염, 척추염, 염증 통증, 요도염, 방광염, 패혈증, 피부염, 치주염, 치은염, 당뇨병, 천식, 결핵, 기관지염, 알레르기성 및 비-알레르기성 비염, 만성 및 급성 비염, 만성 및 급성 위염 또는 장염, 궤양성 위염, 급성 및 만성 신장염, 급성 및 만성 간염, 만성 폐쇄성 폐질환, 폐섬유증, 과민성 대장 증후군, 염증 통증, 편두통, 두통, 허리 통증, 섬유 근육통, 근막 질환, 바이러스 감염(예컨대, C형 감염), 박테리아 감염, 곰팡이 감염, 화상, 외과적 또는 치과적 수술에 의한 상처, 프로스타글라딘 E 과다 증후군, 아테롬성 동맥 경화증, 통풍, 관절염, 류머티즘성 관절염, 강직성 척추염, 호지킨병, 췌장염, 결막염, 각막염, 공막염, 포도막염, 습진 및 다발성 경화증일 수 있으며, 복막염인 것이 더욱 바람직하다.In one embodiment, the inflammatory disease is peritonitis, spondylitis, inflammatory pain, urethritis, cystitis, sepsis, dermatitis, periodontitis, gingivitis, diabetes, asthma, tuberculosis, bronchitis, allergic and non-allergic rhinitis, chronic and acute rhinitis, Chronic and acute gastritis or enteritis, ulcerative gastritis, acute and chronic nephritis, acute and chronic hepatitis, chronic obstructive pulmonary disease, pulmonary fibrosis, irritable bowel syndrome, inflammatory pain, migraine, headache, back pain, fibromyalgia, fascia disease, virus Infections (e.g., type C infection), bacterial infections, fungal infections, burns, wounds caused by surgical or dental surgery, prostaglandin E hypersyndrome, atherosclerosis, gout, arthritis, rheumatoid arthritis, ankylosing spondylitis, Hodgkin's disease, pancreatitis, conjunctivitis, keratitis, scleritis, uveitis, eczema, and multiple sclerosis, more preferably peritonitis.

일 구현예에서, 본 발명의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온은 3μM 이하로 포함될 수 있다.In one implementation, the 2-cyclohexyl-6-a mino-6,7-dihydro -5 H of the present invention benzo [1,3] oxa thiol-4-one can be included in more than 3μM.

본 발명의 조성물은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온 뿐 아니라 이와 동일 또는 유사한 기능을 지닌 다른 유효성분을 추가로 함유하거나, 또는 상기 성분들과 상이한 기능을 지닌 다른 유효성분을 추가로 함유함으로써, 염증 질환의 예방 또는 치료용 약학 조성물로 제조될 수 있다.The compositions of the present invention is 2-cyclohexyl-6-diamino-6,7-dihydro -5 H - benzo [1,3] oxa thiol-4-one, as well as this same or other active ingredients having a similar function By further containing, or by further containing another active ingredient having a different function from the above components, it can be prepared as a pharmaceutical composition for the prevention or treatment of inflammatory diseases.

본 발명에서, 용어 "예방"이란 본 발명에 따른 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온 또는 이를 포함하는 조성물의 투여에 의해 염증의 발생, 확산 및 재발을 억제 또는 지연시키는 모든 행위를 의미한다.In the present invention, the term "prevention" means the present invention according to the 2-cyclohexyl-6-diamino-6,7-dihydro -5 H -, including benzo [1,3] oxa thiol-4-one, or it Any action that inhibits or delays the development, spread and recurrence of inflammation by administration of the composition.

본 발명에서 사용된 용어 "치료"란 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온 또는 이를 포함하는 조성물의 투여로 염증 질환 또는 이의 합병증의 증세를 호전시키거나 이롭게 변경하는 모든 행위를 의미한다. 본 발명이 속하는 기술분야에서 통상의 지식을 가진 자라면, 대한의학협회 등에서 제시된 자료를 참조하여 본원의 조성물이 효과가 있는 질환의 정확한 기준을 알고, 개선, 향상 및 치료된 정도를 판단할 수 있을 것이다.The term "treatment" is 2-cyclohexyl-6-a mino-6,7-dihydro -5 H used in the present invention benzo [1,3] oxa thiol-4-one or the dose of the composition comprising the same By means all actions to improve or beneficially improve the symptoms of inflammatory diseases or complications. If the person having ordinary knowledge in the technical field to which the present invention belongs, can refer to the data presented by the Korean Medical Association and the like to know the exact criteria of the disease in which the composition of the present invention is effective, and to determine the degree of improvement, improvement and treatment. will be.

본 발명의 조성물의 치료적으로 유효한 양은 여러 요소, 예를 들면 투여방법, 목적부위, 환자의 상태 등에 따라 달라질 수 있다. 따라서, 인체에 사용 시 투여량은 안전성 및 효율성을 함께 고려하여 적정량으로 결정되어야 한다. 동물실험을 통해 결정한 유효량으로부터 인간에 사용되는 양을 추정하는 것도 가능하다. 유효한 양의 결정시 고려할 이러한 사항은, 예를 들면 Hardman and Limbird, eds., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed.(2001), Pergamon Press; 및 E.W. Martin ed., Remington's Pharmaceutical Sciences, 18th ed.(1990), Mack Publishing Co.에 기술되어있다.The therapeutically effective amount of the composition of the present invention may vary depending on several factors, for example, the method of administration, the target site, the condition of the patient, and the like. Therefore, when used in the human body, the dosage should be determined in an appropriate amount in consideration of safety and efficiency. It is also possible to estimate the amount used in humans from the effective amount determined through animal experiments. Such considerations to be taken into account when determining an effective amount include, for example, Hardman and Limbird, eds., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed. (2001), Pergamon Press; And E.W. Martin ed., Remington's Pharmaceutical Sciences, 18th ed. (1990), Mack Publishing Co.

본 발명의 약학조성물은 약학적으로 유효한 양으로 투여한다. 본 발명에서 사용되는 용어, "약학적으로 유효한 양"은 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미하며, 유효용량 수준은 환자의 건강상태, 염증 질환의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료기간, 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 본 발명의 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와 순차적으로 또는 동시에 투여될 수 있으며, 단일 또는 다중 투여될 수 있다. 상기한 요소들을 모두 고려하여, 부작용없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.The pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount. The term “pharmaceutically effective amount” used in the present invention means an amount sufficient to treat a disease at a reasonable benefit / risk ratio applicable to medical treatment and does not cause side effects, and an effective dose level of the patient Well known in the medical field, factors including inflammatory disease type, severity, drug activity, sensitivity to the drug, administration method, administration time, administration route and discharge rate, duration of treatment, combination or concurrent drug use and other medical fields It can be determined by factors. The composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with a conventional therapeutic agent, and may be administered single or multiple. Taking all of the above factors into consideration, it is important to administer an amount that can achieve the maximum effect in a minimal amount without side effects, which can be easily determined by those skilled in the art.

일 구현예에서, 상기 약학 조성물은 경구형 제형, 외용제, 좌제, 멸균 주사용액 및 분무제를 포함하는 군으로부터 선택되는 하나 이상의 제형일 수 있다. In one embodiment, the pharmaceutical composition may be one or more formulations selected from the group comprising oral dosage forms, external preparations, suppositories, sterile injectable solutions and sprays.

본 발명의 조성물은 또한 생물학적 제제에 통상적으로 사용되는 담체, 희석제, 부형제 또는 둘 이상의 이들의 조합을 포함할 수 있다. 약학적으로 허용 가능한 담체는 조성물을 생체 내 전달에 적합한 것이면 특별히 제한되지 않으며, 예를 들면, Merck Index, 13th ed., Merck & Co. Inc. 에 기재된 화합물, 식염수, 멸균수, 링거액, 완충 식염수, 덱스트로스 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 이용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한, 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주이용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. 더 나아가 당 분야의 적정한 방법으로 또는 Remington's Pharmaceutical Science(Mack Publishing Company, Easton PA, 18th, 1990)에 개시되어 있는 방법을 이용하여 각 질환에 따라 또는 성분에 따라 바람직하게 제제화할 수 있다.The compositions of the present invention may also include carriers, diluents, excipients, or combinations of two or more of those commonly used in biological agents. The pharmaceutically acceptable carrier is not particularly limited as long as the composition is suitable for in vivo delivery, for example, Merck Index, 13th ed., Merck & Co. Inc. The compound, saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or more of these components may be used in combination, and other antioxidants, buffers, bacteriostatic agents, etc. may be used as required. Conventional additives can be added. In addition, diluents, dispersants, surfactants, binders, and lubricants may be additionally added to formulate into main-use formulations such as aqueous solutions, suspensions, emulsions, pills, capsules, granules, or tablets. Furthermore, it can be preferably formulated according to each disease or component using methods appropriate in the art or using methods disclosed in Remington's Pharmaceutical Science (Mack Publishing Company, Easton PA, 18th, 1990).

본 발명의 조성물에 추가로 동일 또는 유사한 기능을 나타내는 유효성분을 1종 이상 함유할 수 있다. 본 발명의 조성물은, 조성물 총 중량에 대하여 상기 단백질을 0.0001 내지 10 중량 %로, 바람직하게는 0.001 내지 1 중량 %를 포함한다. The composition of the present invention may further contain one or more active ingredients exhibiting the same or similar functions. The composition of the present invention, based on the total weight of the composition, comprises 0.0001 to 10% by weight of the protein, preferably 0.001 to 1% by weight.

본 발명의 약학 조성물은 약학적으로 허용 가능한 첨가제를 더 포함할 수 있으며, 이때 약학적으로 허용 가능한 첨가제로는 전분, 젤라틴화 전분, 미결정셀룰로오스, 유당, 포비돈, 콜로이달실리콘디옥사이드, 인산수소칼슘, 락토스, 만니톨, 엿, 아라비아고무, 전호화전분, 옥수수전분, 분말셀룰로오스, 히드록시프로필셀룰로오스, 오파드라이, 전분글리콜산나트륨, 카르나우바 납, 합성규산알루미늄, 스테아린산, 스테아린산마그네슘, 스테아린산알루미늄, 스테아린산칼슘, 백당, 덱스트로스, 소르비톨 및 탈크 등이 사용될 수 있다. 본 발명에 따른 약학적으로 허용 가능한 첨가제는 상기 조성물에 대해 0.1 중량부 내지 90 중량부 포함되는 것이 바람직하나, 이에 한정되는 것은 아니다.The pharmaceutical composition of the present invention may further include a pharmaceutically acceptable additive, wherein pharmaceutically acceptable additives include starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, calcium hydrogen phosphate, Lactose, mannitol, syrup, gum arabic, pregelatinized starch, corn starch, powdered cellulose, hydroxypropyl cellulose, opadry, sodium starch glycolate, carnauba lead, synthetic aluminum silicate, stearic acid, magnesium stearate, aluminum stearate, stearic acid Calcium, white sugar, dextrose, sorbitol and talc can be used. The pharmaceutically acceptable additive according to the present invention is preferably included in an amount of 0.1 to 90 parts by weight based on the composition, but is not limited thereto.

본 발명의 조성물은 목적하는 방법에 따라 비 경구 투여(예를 들어 정맥 내, 피하, 복강 내 또는 국소에 적용)하거나 경구 투여할 수 있으며, 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 투여방법, 배설률 및 질환의 중증도 등에 따라 그 범위가 다양하다. 본 발명에 따른 조성물의 일일 투여량은 0.0001 ~ 10 ㎎/㎖이며, 바람직하게는 0.0001 ~ 5 ㎎/㎖이며, 하루 일 회 내지 수회에 나누어 투여하는 것이 더욱 바람직하다. The composition of the present invention may be administered orally (for example, intravenously, subcutaneously, intraperitoneally, or topically) or orally, depending on the desired method, and the dosage is the patient's weight, age, sex, health status, The range varies depending on the diet, administration time, administration method, excretion rate and disease severity. The daily dosage of the composition according to the present invention is 0.0001 to 10 mg / ml, preferably 0.0001 to 5 mg / ml, and more preferably, divided and administered once to several times a day.

본 발명의 조성물의 경구 투여를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데, 통상적으로 사용되는 단순 희석제인 물, 액체 파라핀 이외에 다양한 부형제, 예컨대 습윤제, 감미제, 방향제, 보존제 등이 함께 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성 용제, 현탁제, 유제, 동결건조 제제, 좌제 등이 포함된다.Liquid preparations for oral administration of the compositions of the present invention include suspending agents, intravenous solutions, emulsions, syrups, etc., and various excipients, such as wetting agents, sweeteners, flavoring agents, preservatives, in addition to water and liquid paraffin, which are commonly used simple diluents. Etc. may be included. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, suppositories, and the like.

일 측면에서, 본 발명은 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 함유하는 염증 질환의 예방 또는 개선용 식품 조성물에 관한 것이다.In one aspect, the present invention is 2-cyclohexyl-6-a mino-6,7-dihydro -5 H - [1,3] for the prevention or amelioration of inflammatory disorders, containing a thiol-oxa-4-one It relates to a food composition.

본 발명의 조성물을 식품 조성물로 사용하는 경우, 상기 아출 추출물을 그대로 첨가하거나 다른 식품 또는 식품 성분과 함께 사용할 수 있고, 통상의 방법에 따라 적절하게 사용할 수 있다. 상기 조성물은 유효성분 이외에 식품학적으로 허용가능한 식품보조첨가제를 포함할 수 있으며, 유효성분의 혼합량은 사용 목적(예방, 건강 또는 치료적 처치)에 따라 적합하게 결정될 수 있다.When the composition of the present invention is used as a food composition, the above extract can be added as it is or used with other foods or food ingredients, and can be suitably used according to a conventional method. The composition may include a food additive that is food-acceptable in addition to the active ingredient, and the mixing amount of the active ingredient may be appropriately determined according to the purpose of use (prevention, health, or therapeutic treatment).

본 발명에서 사용되는 용어 "식품보조첨가제"란 식품에 보조적으로 첨가될 수 있는 구성요소를 의미하며, 각 제형의 건강기능식품을 제조하는데 첨가되는 것으로서 당업자가 적절히 선택하여 사용할 수 있다. 식품보조첨가제의 예로는 여러 가지 영양제, 비타민, 광물(전해질), 합성 풍미제 및 천연 풍미제 등의 풍미제, 착색제 및 충진제, 펙트산 및 그의 염, 알긴산 및 그의 염, 유기산, 보호성 콜로이드 증점제, pH 조절제, 안정화제, 방부제, 글리세린, 알콜, 탄산음료에 사용되는 탄산화제 등이 포함되지만, 상기 예들에 의해 본 발명의 식품보조첨가제의 종류가 제한되는 것은 아니다.The term "food supplement additive" used in the present invention means a component that can be supplementally added to food, and is added to prepare a health functional food of each formulation, and can be appropriately selected and used by those skilled in the art. Examples of food supplements include flavors such as various nutrients, vitamins, minerals (electrolytes), synthetic flavors and natural flavors, colorants and fillers, pectic acid and salts thereof, alginic acid and salts thereof, organic acids, protective colloidal thickeners , pH adjusting agents, stabilizers, preservatives, glycerin, alcohol, carbonic acid used in carbonated beverages, and the like, but the types of food supplements of the present invention are not limited by the examples.

본 발명의 식품 조성물에는 건강기능식품이 포함될 수 있다. 본 발명에서 사용되는 용어 "건강기능식품"이란 인체에 유용한 기능성을 가진 원료나 성분을 사용하여 정제, 캅셀, 분말, 과립, 액상 및 환 등의 형태로 제조 및 가공한 식품을 말한다. 여기서 '기능성'이라 함은 인체의 구조 및 기능에 대하여 영양소를 조절하거나 생리학적 작용 등과 같은 보건용도에 유용한 효과를 얻는 것을 의미한다. 본 발명의 건강기능식품은 통상의 기술분야에서 통상적으로 사용되는 방법에 의하여 제조가능하며, 상기 제조시에는 통상의 기술분야에서 통상적으로 첨가하는 원료 및 성분을 첨가하여 제조할 수 있다. 또한 상기 건강기능식품의 제형 또한 건강기능식품으로 인정되는 제형이면 제한없이 제조될 수 있다. 본 발명의 식품용 조성물은 다양한 형태의 제형으로 제조될 수 있으며, 일반 약품과는 달리 식품을 원료로 하여 약품의 장기 복용 시 발생할 수 있는 부작용 등이 없는 장점이 있고, 휴대성이 뛰어나, 본 발명의 건강기능식품은 항암제의 효과를 증진시키기 위한 보조제로 섭취가 가능하다.The food composition of the present invention may include a health functional food. The term "health functional food" used in the present invention refers to food prepared and processed in the form of tablets, capsules, powders, granules, liquids and pills using ingredients or ingredients having useful functional properties for the human body. Here, the term 'functional' refers to obtaining a useful effect for health use, such as regulating nutrients or physiological effects on the structure and function of the human body. The health functional food of the present invention can be manufactured by a method conventionally used in the general technical field, and may be prepared by adding raw materials and components conventionally added in the general technical field. In addition, the formulation of the health functional food can also be prepared without limitation as long as the formulation is recognized as a health functional food. The food composition of the present invention can be prepared in various types of formulations, and has the advantage of not having side effects that may occur when taking the drug for a long time using food as a raw material, unlike general drugs, and has excellent portability, and the present invention Can be consumed as a supplement to enhance the effectiveness of anticancer drugs.

또한, 본 발명의 조성물이 사용될 수 있는 건강식품의 종류에는 제한이 없다. 아울러 본 발명의 아출 추출물을 활성성분으로 포함하는 조성물은 당업자의 선택에 따라 건강기능식품에 함유될 수 있는 적절한 기타 보조 성분과 공지의 첨가제를 혼합하여 제조할 수 있다. 첨가할 수 있는 식품의 예로는 육류, 소세지, 빵, 쵸코렛, 캔디류, 스낵류, 과자류, 피자, 라면, 기타 면류, 껌류, 아이스크림 류를 포함한 낙농제품, 각종 스프, 음료수, 차, 드링크제, 알콜 음료 및 비타민 복합제 등이 있으며, 본 발명에 따른 추출물을 주성분으로 하여 제조한 즙, 차, 젤리 및 주스 등에 첨가하여 제조할 수 있다.In addition, there is no limitation on the type of healthy foods to which the composition of the present invention can be used. In addition, the composition comprising the extract of the extract of the present invention as an active ingredient may be prepared by mixing a known additive with other suitable auxiliary ingredients that may be contained in a health functional food according to the choice of a person skilled in the art. Examples of foods that can be added are meat, sausage, bread, chocolate, candy, snacks, confectionery, pizza, ramen, other noodles, dairy products including gums, ice creams, various soups, beverages, teas, drinks, alcoholic beverages, and There are vitamin complexes and the like, and can be prepared by adding the extract according to the present invention as a main ingredient, and adding it to juice, tea, jelly, and juice.

하기의 실시예를 통하여 본 발명을 보다 상세하게 설명한다. 그러나 하기 실시예는 본 발명의 내용을 구체화하기 위한 것일 뿐 이에 의해 본 발명이 한정되는 것은 아니다.The present invention will be described in more detail through the following examples. However, the following examples are only intended to materialize the contents of the present invention, and the present invention is not limited thereto.

실시예 1. 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5Example 1 2-cyclohexylimino-6-methyl-6,7-dihydro-5 HH -벤조[1,3]옥사티올-4-온-Benzo [1,3] oxathiol-4-one (2-cyclohexylimino-6-methyl-6,7-dihydro-5(2-cyclohexylimino-6-methyl-6,7-dihydro-5 HH -benzo[1,3]oxathiol-4-one, BOT-4-one)의 세포독성 확인-Cytotoxicity confirmation of benzo [1,3] oxathiol-4-one, BOT-4-one)

[화학식 1][Formula 1]

Figure 112017091645861-pat00002
Figure 112017091645861-pat00002

상기 화학식 1의 BOT-4-one(2-cyclohexylimino-6-methyl-6,7-dihydro-5H-benzo[1,3]oxathiol-4-one)의 독성과 유효 농도를 확인하기 위하여, 마우스 골수 유래 대식세포 (bone-marrow derived macrophages, BMDMs)를 이용하여 세포생존 분석을 수행하였다. 구체적으로, 8주령의 C57BL/6 마우스의 대퇴골 및 경골을 살균된 PBS에서 수세하여 골수세포를 수득하였다. 그 후 골수세포들을 10% FBS, 30% LCM(L929 cell-conditioned medium) 및 1% 페니실린-스트렙토마이신이 포함된 RPMI 1640 배지 (Gibco, Grand Island, NY, USA)에서 7일 동안 배양하여 대식세포(BMDM)로 분화시켰다. 분화된 세포들을 비-조직 배양-처리 패트리 디쉬(non-tissue culture-treated Petri dishes) (SPL Life Science Co., Gyeonggi-do, Republic of Korea)에 분주하고 37℃ 및 5% CO2 조건의 인큐베이터에서 배양하였다. 준비된 BMDMs에 BOT-4-one (Seoul National University of College of Medicine, South Korea)을 0, 1.5626, 3.125 및 6.25 mM로 각각 처리하고 MTT 분석을 통해 세포 생존율을 확인하였다.In order to determine the toxicity and the effective concentration of the BOT-4-one (2- cyclohexylimino-6-methyl-6,7-dihydro-5 H -benzo [1,3] oxathiol-4-one) of the formula (1), mouse Cell survival analysis was performed using bone-marrow derived macrophages (BMDMs). Specifically, the femur and tibia of 8-week-old C57BL / 6 mice were washed with sterile PBS to obtain bone marrow cells. Then, bone marrow cells were cultured for 7 days in RPMI 1640 medium (Gibco, Grand Island, NY, USA) containing 10% FBS, 30% L929 cell-conditioned medium (LCM) and 1% penicillin-streptomycin. (BMDM). Differentiated cells are dispensed into non-tissue culture-treated Petri dishes (SPL Life Science Co., Gyeonggi-do, Republic of Korea) and incubated at 37 ° C. and 5% CO 2 conditions. Cultured in Prepared BMDMs were treated with BOT-4-one (Seoul National University of College of Medicine, South Korea) at 0, 1.5626, 3.125 and 6.25 mM, respectively, and cell viability was confirmed by MTT analysis.

그 결과, BOT-4-one 농도 3μM까지는 BMDMs의 세포 생존율에 영향이 없음을 알 수 있었다 (도 1).As a result, it was found that the BOT-4-one concentration up to 3 μM had no effect on the cell viability of BMDMs (FIG. 1).

실시예Example 2. BOT-4-one의 NLRP3 염증조절복합체(inflammasome) 억제 확인2. Confirmation of inhibition of NLRP3 inflammation control complex (inflammasome) of BOT-4-one

BOT-4-one의 NLRP3 염증조절복합체 활성화에 대한 영향을 확인하기 위하여, LPS 전처리된 BMDMs에 정규적인(canonical) 또는 비정규적인(noncanonical) NLRP3 염증조절복합체 활성화제를 처리하고 BOT-4-one의 존재 여부에 따른 영향을 확인하였다. 구체적으로, BMDMs에 정규적인 NLRP3 염증조절복합체의 활성화제로서 LPS 100ng/mL를 4시간 동안 전처리한 뒤, Opti-MEM 배지로 바꿔주고, BOT-4-one 및 zVAD (20 μM)를 처리하여 1시간 동안 인큐베이션하였다. 그 후 ATP (5 mM) 또는 니제리신(nigericin) (10 μM)으로 1시간 동안, 또는 실리카 크리스탈(silica crystals) (150 ㎍/mL)으로 3시간 동안 자극하였다. 또한, 비정규적인 NLRP3 염증조절복합체의 활성화제로서 Pam3CSK4 (200 ng/mL)를 3시간 동안 전처리한 뒤, Opti-MEM 배지로 바꿔주고, BOT-4-one 및 KCl (150 mM)를 처리하여 1시간 동안 인큐베이션하였다. 그 후 LPS (E.coli 011:B4) (70 ㎍/mL)를 리포펙타민(Lipofectamine) 2000 (Invitrogen)으로 트랜스펙션하여 3시간 동안 인큐베이션하여 자극하였다. 그 후, 세포를 파쇄한 파쇄물을 이용하여 웨스턴 블롯 분석 (Western blot chemiluminescence reagent kit, Pierce Chemical, Rockford, IL, USA), ELISA (ELISA kit, R&D Systems, Minneapolis, MN, USA) 및 LDH 분석을 수행하여 BOT-4-one이 NLRP3 염증조절복합체의 활성화에 미치는 영향을 확인하였다. To confirm the effect of BOT-4-one on NLRP3 inflammatory regulatory complex activation, LPS pretreated BMDMs were treated with canonical or noncanonical NLRP3 inflammatory regulatory complex activators and BOT-4-one. The effect of existence was confirmed. Specifically, 100 ml / mL of LPS was pretreated for 4 hours as an activator of the NLRP3 inflammatory regulatory complex normal to BMDMs, replaced with Opti-MEM medium, and treated with BOT-4-one and zVAD (20 μM) 1 Incubated for hours. It was then stimulated with ATP (5 mM) or nigericin (10 μM) for 1 hour, or with silica crystals (150 μg / mL) for 3 hours. In addition, Pam3CSK4 (200 ng / mL) as an activator of the irregular NLRP3 inflammatory regulatory complex was pre-treated for 3 hours, replaced with Opti-MEM medium, and treated with BOT-4-one and KCl (150 mM) 1 Incubated for hours. Thereafter, LPS ( E.coli 011: B4) (70 μg / mL) was transfected with Lipofectamine 2000 (Invitrogen), and stimulated by incubation for 3 hours. Then, Western blot analysis (Western blot chemiluminescence reagent kit, Pierce Chemical, Rockford, IL, USA), ELISA (ELISA kit, R & D Systems, Minneapolis, MN, USA) and LDH analysis were performed using the crushed cells. The effect of BOT-4-one on the activation of the NLRP3 inflammatory regulatory complex was confirmed.

그 결과, BOT-4-one이 농도 의존적으로 NLRP3 염증조절복합체의 활성화에 의해 유발된 IL-1β 분비 및 caspase-1 절단을 억제하는 것을 알 수 있었다 (도 2A-C). 또한, BOT-4-one의 IL-1β 분비 억제는 IC50 값으로 약 1.28 μM (ATP), 0.67 μM (nigericin) 및 0.59 μM (silica crystals)로 나타났다 (도 2D). 또한, BOT-4-one은 NLRP3 염증조절복합체에 의해 유발되는 LDH(lactate dehydrogenase) 방출도 억제하였다 (도 2E). 아울러, BOT-4-one은 비정규적인 NLRP3 염증조절복합체에 의한 IL-1β 분비 및 caspase-1 절단도 농도의존적으로 억제하였다 (도 2F). 따라서, 상기 결과에 따라 본 발명의 BOT-4-one가 NLRP3 염증조절복합체의 활성화를 정규적인 조건과 비정규적인 조건 모두에서 억제하는 것을 알 수 있었다.As a result, it was found that BOT-4-one inhibits IL-1β secretion and caspase-1 cleavage induced by activation of the NLRP3 inflammatory regulatory complex in a concentration-dependent manner (FIGS. 2A-C). In addition, inhibition of IL-1β secretion of BOT-4-one was found to be about 1.28 μM (ATP), 0.67 μM (nigericin) and 0.59 μM (silica crystals) with IC 50 values (FIG. 2D). In addition, BOT-4-one also inhibited the release of lactate dehydrogenase (LDH) caused by the NLRP3 inflammatory regulatory complex (Figure 2E). In addition, BOT-4-one also inhibited IL-1β secretion and caspase-1 cleavage by the non-regular NLRP3 inflammatory regulatory complex (Figure 2F). Accordingly, it was found that the BOT-4-one of the present invention inhibits the activation of the NLRP3 inflammatory regulatory complex in both normal and non-normal conditions according to the above results.

실시예Example 3.3. BOT-4-one의 NLRP3-매개 ASC 올리고머화 차단 확인Confirmation of blocking NLRP3-mediated ASC oligomerization of BOT-4-one

BOT-4-one이 염증조절복합체-매개 ASC 올리고머 형성에 미치는 영향을 확인하기 위해, ASC 올리고머 가교-결합을 면역형광 이미징과 웨스턴 블롯으로 확인하였다. 구체적으로, 상기 실시예 2에서와 같이 LPS 전처리된 BMDMs에 BOT-4-one 또는 KCl을 1시간 동안 처리하고, ATP, 니제리신 또는 실리카 크리스탈로 NLRP3 염증조절복합체를 활성화시킨 후, 세포를 A0 버퍼 (0.5% Triton X100, 20 mM HEPES-KOH, pH 7.5, 150 mM KCl 및 complete protease and phosphatase inhibitor cocktail)와 G26 주사기로 파쇄하였다. 세포 파쇄물을 4℃에서 6000rpm으로 10분 동안 원심분리하여 얻은 펠릿을 PBS에 재부유한 뒤, DSS(disuccinimidyl suberate) (2 mM) (Thermo Scientific-Pierce, Rockford, IL, USA)와 가교시켰다. 가교-결합된 펠릿을 13000rpm으로 15분동안 원심분리하여 SDS 샘플 버퍼와 혼합하여 웨스턴 블롯 시료를 준비하였다. 세포 파쇄물의 상등액에 대해 IL-1β를, 펠릿 파쇄물에 대해 ASC의 올리고머, 다이머 및 모노머의 존재를, 전체 세포 파쇄물에 대해 pro-IL-1β, NLRP3, ASC, pro-caspase-1 및 β-actin의 존재를 확인하였다. 아울러, ASC 반점 염색(speck staining)을 위해, NLRP3 염증조절복합체가 활성화된 BMDMs를 4% 파라포름알데하이드로 고정하였으며, 아세톤으로 투과시키고(permeabilized), 10% 말 혈청(horse serum)으로 블로킹하였다. 그 후, 세포들을 ASC 항체 (Santa Cruz) 및 Cy3-결합된 항-rabbit 항체 (Jackson ImmunoResearch Lab)로 염색하였다. 핵은 DAPI (Sigma-Aldrich)로 염색하여 관찰하였다.To confirm the effect of BOT-4-one on inflammatory regulatory complex-mediated ASC oligomer formation, ASC oligomer cross-linking was confirmed by immunofluorescence imaging and Western blot. Specifically, as in Example 2, BPS-4-one or KCl was treated with LPS pre-treated BMDMs for 1 hour, and after activation of the NLRP3 inflammatory regulatory complex with ATP, nigericin, or silica crystals, cells were A0. It was crushed with a buffer (0.5% Triton X100, 20 mM HEPES-KOH, pH 7.5, 150 mM KCl and complete protease and phosphatase inhibitor cocktail) and a G26 syringe. The pellet obtained by centrifuging the cell lysate at 4 ° C. at 6000 rpm for 10 minutes was resuspended in PBS, and then crosslinked with DSS (disuccinimidyl suberate) (2 mM) (Thermo Scientific-Pierce, Rockford, IL, USA). The cross-linked pellet was centrifuged at 13000 rpm for 15 minutes to mix with the SDS sample buffer to prepare a Western blot sample. IL-1β for supernatant of cell debris, presence of oligomers, dimers and monomers of ASC for pellet debris, pro-IL-1β, NLRP3, ASC, pro-caspase-1 and β-actin for whole cell debris Confirmed the existence of. In addition, for ASC spot staining, NLRP3 inflammatory regulatory complex-activated BMDMs were fixed with 4% paraformaldehyde, permeabilized with acetone, and blocked with 10% horse serum. Cells were then stained with ASC antibody (Santa Cruz) and Cy3-bound anti-rabbit antibody (Jackson ImmunoResearch Lab). Nuclei were observed by staining with DAPI (Sigma-Aldrich).

그 결과, BOT-4-one이 LPS-전처리된 BMDMs에서 니제리신에 의해 자극된 ASC 반점 형성을 현저히 억제하였다 (도 3A). 아울러, BOT-4-one은 니제리신, ATP 및 실리카 크리스탈에 의해 가교-결합된 세포질 펠렛에서 ASC 올리고머화를, NLRP3 염증조절복합체의 발현 변화 없이, BOT-4-one의 농도의존적으로 억제하였다 (도 3B). 즉, 이를 통해 BOT-4-one이 NLRP3-의존적 ASC 반점 형성을 조절하는 것을 알 수 있다.As a result, BOT-4-one significantly inhibited the formation of ASC spots stimulated by nigericin in LPS-pretreated BMDMs (FIG. 3A). In addition, BOT-4-one inhibited ASC oligomerization in cytoplasmic pellets cross-linked by nigericin, ATP, and silica crystals without concentration change of the NLRP3 inflammatory regulatory complex and BOT-4-one concentration-dependently. (Figure 3B). That is, it can be seen that BOT-4-one modulates NLRP3-dependent ASC spot formation.

실시예Example 4. BOT-4-one에 의한 NLRP3 및 ASC의 이동 억제 확인4. Confirmation of movement inhibition of NLRP3 and ASC by BOT-4-one

BOT-4-one가 염증조절복합체(inflammasome)의 이동에 미치는 영향을 확인하기 위하여, Triton X-100 가용성 또는 불용성 분획으로의 재분배 효과를 확인하였다. 구체적으로, 상기 실시예 3에서와 같이 LPS 전처리된 BMDMs에 BOT-4-one 또는 KCl을 1시간 동안 처리하고, ATP, 니제리신 또는 실리카 크리스탈로 NLRP3 염증조절복합체를 활성화시킨 후, 세포를 TTNE 버퍼 (1% Triton X-100, 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 2mM EDTA 및 complete protease and phosphatase inhibitor cocktail)로 파쇄하였다. 세포 파쇄물을 6000rpm으로 15분 동안 4℃에서 원심분리한 뒤, 상등액 및 펠릿을 각각 Triton X-100의 가용성 및 불용성 분획으로서 사용하여 각 분획 (상등액, 펠릿, 세포파쇄물 전체)에서의 IL-1β, caspase-1, pro-IL-1β, NLRP3, ASC, pro-caspase-1 및 β-actin의 존재를 각각 웨스턴 블롯 분석 방법으로 확인하였다. To confirm the effect of BOT-4-one on the migration of the inflammatory regulatory complex (inflammasome), the effect of redistribution into Triton X-100 soluble or insoluble fraction was confirmed. Specifically, as in Example 3, BPS-4-one or KCl was treated with LPS pre-treated BMDMs for 1 hour, and after activation of the NLRP3 inflammatory regulatory complex with ATP, nigericin, or silica crystals, the cells were TTNE. Crushed with buffer (1% Triton X-100, 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 2 mM EDTA and complete protease and phosphatase inhibitor cocktail). The cell lysate was centrifuged at 6000 rpm for 15 minutes at 4 ° C., and then supernatant and pellets were used as soluble and insoluble fractions of Triton X-100, respectively. The presence of caspase-1, pro-IL-1β, NLRP3, ASC, pro-caspase-1 and β-actin were confirmed by Western blot analysis, respectively.

그 결과, BOT-4-one이 NLRP3 염증조절복합체의 활성화 후, 상등액에서 IL-1β 및 caspase-1의 분비뿐만 아니라, NLRP3 및 ASC의 불용성 분획 (펠릿)으로의 이동까지 감소시킨 것을 알 수 있었다 (도 4). 이를 통해, BOT-4-one이 NLRP3 및 ASC의 Triton X-100 불용성 분획으로의 이동을 억제함으로써 NLRP3 염증조절복합체 활성화를 특이적으로 억제하는 것을 알 수 있다.As a result, it was found that after activation of the NLRP3 inflammatory regulatory complex, BOT-4-one decreased not only the secretion of IL-1β and caspase-1 from the supernatant, but also the migration of NLRP3 and ASC to the insoluble fraction (pellet). (Figure 4). Through this, it can be seen that BOT-4-one specifically inhibits NLRP3 inflammatory regulatory complex activation by inhibiting migration of NLRP3 and ASC to the Triton X-100 insoluble fraction.

실시예Example 5. BOT-4-one의 NLRP3 염증조절복합체 활성 억제 메커니즘 확인5. Confirmation of the mechanism of inhibition of BOT-4-one's NLRP3 inflammation control complex activity

5-1. NLRP3의 ATPase 활성 억제 5-1. Inhibition of ATPase activity of NLRP3

BOT-4-one이 NLRP3 염증조절복합체의 활성화 억제 메커니즘을 확인하기 위하여, NLRP3의 ATPase 활성에 미치는 영향을 확인하였다. 구체적으로, 인간 재조합 NLRP3 (BPS Bioscience, San Diego, CA, USA)을 DMSO (대조군) 또는 BOT-4-one과 함께 37℃의 반응 버퍼 (20mM Tris HCl, pH 7.8, 133 mM NaCl, 20 mM MgCl2, 3 mM KCl 및 0.56 mM EDTA)에서 20분 동안 인큐베이션시켰다. 그 후 ATP (250 mM, ultra-pure ATP)를 첨가하여 37℃에서 40분 동안 반응시켰다. NLRP3에 의한 ATP 가수분해는 ADP-Glo Kinase Assay (Promega, Madison, WI, USA)를 이용하여 세포 상등액에서의 발광(luminescent) ADP를 검출함으로써 확인하였다. To confirm the mechanism by which BOT-4-one inhibits the activation of NLRP3 inflammatory regulatory complex, the effect of NLRP3 on ATPase activity was confirmed. Specifically, human recombinant NLRP3 (BPS Bioscience, San Diego, CA, USA) was reacted with DMSO (control) or BOT-4-one at 37 ° C in a reaction buffer (20mM Tris HCl, pH 7.8, 133 mM NaCl, 20 mM MgCl 2 , 3 mM KCl and 0.56 mM EDTA) for 20 minutes. Then, ATP (250 mM, ultra-pure ATP) was added to react at 37 ° C. for 40 minutes. ATP hydrolysis by NLRP3 was confirmed by detecting luminescent ADP in the cell supernatant using the ADP-Glo Kinase Assay (Promega, Madison, WI, USA).

그 결과, BOT-4-one이 NLRP3의 ATPase 활성을 농도 의존적으로 현저하게 억제하는 것을 알 수 있었다 (도 5A).As a result, it was found that BOT-4-one significantly inhibited the NLRP3 ATPase activity in a concentration-dependent manner (FIG. 5A).

5-2. NLRP3 알킬화5-2. NLRP3 alkylation

BOT-4-one의 NLRP3의 활성 억제가 NLRP3 알킬화에 의한 것인지 확인하기 위하여, 상기 실시예들에서와 같이 LPS 전처리된 BMDMs에 BOT-4-one 또는 Bay11-7082 (Bay) (대조군)을 1시간 동안 처리하면서과 알킬화 억제제인 L-시스테인(L-cysteine)을 함께 처리하였다. 그 후, ATP, 니제리신 또는 실리카 크리스탈을 처리하여 NLRP3 염증조절복합체를 활성화시켰다. 그 후, 세포 상등액에서의 NLRP3-매개 LDH 방출을 확인하고, IL-1β 및 caspase-1를 웨스턴 블롯 분석으로 확인하였다.BOT-4-one or Bay11-7082 (Bay) (control) for 1 hour in LPS pretreated BMDMs, as in the above examples, to confirm that the inhibition of NLRP3 activity of BOT-4-one is due to NLRP3 alkylation During the treatment, the alkylation inhibitor L-cysteine was treated together. Thereafter, ATP, nigericin, or silica crystals were treated to activate the NLRP3 inflammatory regulatory complex. Thereafter, NLRP3-mediated LDH release from the cell supernatant was confirmed, and IL-1β and caspase-1 were confirmed by Western blot analysis.

그 결과, BOT-4-one이 NLRP3-매개 LDH 방출과 IL-1β 및 caspase-1의 성숙을 현저히 억제하였으며, L-시스테인 처리시 NLRP3-매개 LDH 방출과 IL-1β 및 caspase-1의 성숙이 L-시스테인의 농도의존적으로 다시 회복되는 것을 확인할 수 있었다 (도 5B 및 C). 이를 통해, BOT-4-one이 NLRP3를 알킬화함으로써 NLRP3의 ATPase 활성을 억제하고, 이를 통해 NLRP3 염증조절복합체 활성을 억제함을 알 수 있었다.As a result, BOT-4-one significantly inhibited NLRP3-mediated LDH release and maturation of IL-1β and caspase-1, and NLRP3-mediated LDH release and IL-1β and caspase-1 maturation during L-cysteine treatment It was confirmed that recovery was again dependent on the concentration of L-cysteine (FIGS. 5B and C). Through this, it was found that BOT-4-one inhibits ATPase activity of NLRP3 by alkylating NLRP3, and thereby inhibits NLRP3 inflammatory regulatory complex activity.

실시예 6. NLRP3 알킬화를 통한 ASC 올리고머화 및 이동 억제Example 6. ASC oligomerization and migration inhibition through NLRP3 alkylation

BOT-4-one에 의한 NLRP3의 알킬화를 통한 NLRP3의 ATPase 활성 억제가 ASC 올리고머화와 NLRP3 및 ASC의 Triton X-100 불용성 분획으로의 이동에 미치는 영향을 확인하고자 하였다. 구체적으로, LPS 전처리된 BMDMs에 BOT-4-one을 1시간 동안 처리하면서 알킬화 억제제인 L-시스테인을 함께 처리하고, ATP, 니제리신 또는 실리카 크리스탈로 NLRP3 염증조절복합체를 활성화시킨 뒤, 실시예 3에서와 동일한 방법으로 ASC의 올리고머화를 확인하였다. 또한, 실시예 4와 동일한 방법으로 NLRP3 및 ASC의 Triton X-100 가용성 또는 불용성 분획으로의 이동을 확인하였다.The effect of inhibiting ATPase activity of NLRP3 through alkylation of NLRP3 with BOT-4-one was to determine the effect of ASC oligomerization and migration of NLRP3 and ASC to Triton X-100 insoluble fraction. Specifically, while treating BOT-4-one with LPS pre-treated BMDMs for 1 hour, the alkylation inhibitor L-cysteine was treated together, and after activating the NLRP3 inflammation-regulating complex with ATP, nigericin, or silica crystals, Examples The oligomerization of ASC was confirmed by the same method as in 3. In addition, the movement of NLRP3 and ASC to Triton X-100 soluble or insoluble fraction was confirmed in the same manner as in Example 4.

그 결과, BOT-4-one 처리에 의해 억제된 NLRP3-매개 IL-1β 및 caspase-1의 성숙과 ASC 올리고머화가 알킬화 억제제에 의해 농도의존적으로 회복되었다 (도 6A). 또한, BOT-4-one은 NLRP3 및 ASC의 불용화 분획으로의 이동을 억제하였으나, 이는 알킬화 억제제에 의해 회복되었으며 (도 6B), 이는 L-시스테인의 농도에 의존적으로 나타났다 (도 6C). 이를 통해, BOT-4-one이 ASC 올리고머화와 NLRP3 및 ASC의 Triton X-100 불용성 분획으로의 이동을 억제하는 것이 NLRP3를 알킬화함으로써 이루어지는 것을 알 수 있었다.As a result, maturation and ASC oligomerization of NLRP3-mediated IL-1β and caspase-1 inhibited by BOT-4-one treatment were recovered in a concentration-dependent manner by an alkylation inhibitor (FIG. 6A). In addition, BOT-4-one inhibited the migration of NLRP3 and ASC to the insoluble fraction, but was recovered by the alkylation inhibitor (Figure 6B), which appeared dependent on the concentration of L-cysteine (Figure 6C). Through this, it was found that BOT-4-one inhibits ASC oligomerization and migration of NLRP3 and ASC to the Triton X-100 insoluble fraction by alkylation of NLRP3.

실시예Example 7.7. NLRP3 유비퀴틴화 확인Confirmation of NLRP3 ubiquitination

BOT-4-one에 의한 NLRP3의 유비퀴틴화를 확인하기 위하여, LPS로 전처리된 BMDMs을 L-시스테인(Cys)으로 15분 동안 처리한 뒤, BOT-4-one를 1시간 동안 처리하였다. 그 후, BMDMs를 TINE 버퍼로 파쇄하고, 세포파쇄물과 비드에 결합된 NLRP3 항체를 4에서 하룻밤동안 인큐베이션하였다. 비드를 세척한 뒤 용출 버퍼 (0.1 M Tris-HCl, pH 6.8, 0.4% sodium dodecyl sulfate, 12% glycerol, 0.286 M β-mercaptoethanol 및 0.32% bromophenol blue)로 용출하여 NLRP3의 유비퀴틴화를 확인하였다. 또한, NLRP3와 ASC의 상호 작용을 확인하기 위하여, LPS로 전처리된 BMDMs에 BOT-4-one를 L-시스테인과 함께 1시간 동안 처리한 뒤, ATP, 니제리신 또는 실리카 크리스탈로 NLRP3 염증조절복합체 활성화를 유도하였다. 그 후, 상기 세포들을 IP 버퍼 (1% NP-40, 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.5% sodium deoxycholate 및 complete protease and phosphatase inhibitor cocktail)로 파쇄하고, 20amp로 5초 동안 초음파 파쇄하였다. 세포 파쇄물을 워싱한 뒤 ASC 항체 (Santa Cruz)와 4℃에서 하룻밤 동안 인큐베이션하고 면역침전물(immunoprecipitates)을 NLRP3 항체로 웨스턴 블롯하여 NLRP3와 ASC의 상호 작용을 확인하였다.To confirm the ubiquitination of NLRP3 by BOT-4-one, BMDMs pretreated with LPS were treated with L-cysteine (Cys) for 15 minutes, and then BOT-4-one was treated for 1 hour. Subsequently, BMDMs were crushed with TINE buffer, and the cell lysate and NLRP3 antibody bound to beads were incubated at 4 overnight. After washing the beads, eluting with an elution buffer (0.1 M Tris-HCl, pH 6.8, 0.4% sodium dodecyl sulfate, 12% glycerol, 0.286 M β-mercaptoethanol and 0.32% bromophenol blue) confirmed ubiquitination of NLRP3. In addition, in order to confirm the interaction between NLRP3 and ASC, BOT-4-one was treated with L-cysteine in BMDMs pre-treated with LPS for 1 hour, and then NLRP3 inflammation control complex with ATP, nigericin, or silica crystals. Activation was induced. Then, the cells were crushed with IP buffer (1% NP-40, 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.5% sodium deoxycholate and complete protease and phosphatase inhibitor cocktail), and ultrasonicated at 20 amp for 5 seconds. Shredded. After washing the cell debris, the ASC antibody (Santa Cruz) was incubated overnight at 4 ° C., and immunoprecipitates were Western-blotted with the NLRP3 antibody to confirm the interaction between NLRP3 and ASC.

그 결과, BOT-4-one 처리에 의해 NLRP3 유비퀴틴화가 유발되었으며, 이는 알킬화 억제제에 의해 저해되는 것으로 나타났다 (도 7A). 이에, BOT-4-one에 의한 NLRP3의 알킬화 및 ATPase 활성 억제가 NLRP3의 유비퀴틴화를 유발하는 것을 알 수 있다. 또한, BOT-4-one이 NLRP4 및 ASC의 결합을 억제하는 것으로 나타났으며, 이는 알킬화 억제제에 의해 저해되었다 (도 7B). 이를 통해, BOT-4-one이 NLRP3의 알킬화를 통해 NLRP3의 유비퀴틴화를 유발하고 이어서 NLRP3/ASC의 결합도 억제하는 것을 알 수 있다.As a result, NLRP3 ubiquitination was induced by BOT-4-one treatment, which was shown to be inhibited by an alkylation inhibitor (FIG. 7A). Thus, it can be seen that the alkylation of NLRP3 by BOT-4-one and the inhibition of ATPase activity induce ubiquitination of NLRP3. In addition, BOT-4-one was shown to inhibit the binding of NLRP4 and ASC, which was inhibited by alkylation inhibitors (FIG. 7B). Through this, it can be seen that BOT-4-one induces ubiquitination of NLRP3 through alkylation of NLRP3 and then also inhibits NLRP3 / ASC binding.

실시예 8. BOT-4-one의 복막염 완화 효과Example 8. Effect of BOT-4-one on peritonitis relief

BOT-4-one의 NLRP3 염증조절복합체 관련 질병에 대한 효과를 확인하기 위하여, MSU(Monosodiumurate)로 마우스에 복막염을 유발하고 이에 대한 효과를 확인하였다. 구체적으로, BOT-4-one (10 mg/kg)를 MSU 처리 2시간 및 12시간 전에 복강투여하였다. 그 후, MSU (50 mg/kg) 또는 PBS (대조군)를 마우스에 복강투여하였다. 6시간 후, 마우스들을 희생시킨 뒤, 이로부터 복막세포(Peritoneal cells)를 수득하였다. 그 후, 세포들을 F4/80 및 Ly-6G 항체 (eBioscience)로 염색하여 유세포분석기(flow cytometry) (BD San Diego, CA, USA)로 계수하고, 하기의 수학식 1을 통해 호중구의 수를 구하였다. 또한, 복강 세척액의 상층액에서의 IL-1β의 농도를 ELISA로 측정 하였다.In order to confirm the effect of BOT-4-one on NLRP3 inflammatory regulatory complex-related diseases, peritoneitis was induced in mice with MSU (Monosodiumurate) and its effect was confirmed. Specifically, BOT-4-one (10 mg / kg) was intraperitoneally administered 2 and 12 hours before MSU treatment. MSU (50 mg / kg) or PBS (control) was then intraperitoneally administered to the mice. After 6 hours, the mice were sacrificed, from which peritoneal cells were obtained. Thereafter, the cells were stained with F4 / 80 and Ly-6G antibody (eBioscience), counted by flow cytometry (BD San Diego, CA, USA), and the number of neutrophils was obtained through Equation 1 below. Did. In addition, the concentration of IL-1β in the supernatant of the abdominal cavity was measured by ELISA.

Figure 112017091645861-pat00003
Figure 112017091645861-pat00003

그 결과, BOT-4-one이 MSU-매개 총 세포수와 Ly-6G+/F480- 호중구의 수를 현저하게 감소시켰다 (도 8A 및 B). 또한, MSU-유발 IL-1β 생성을 감소시켰다 (도 8C). 이를 통해, BOT-4-one이 NLRP3 염증조절복합체-매개 IL-1β 생성을 억제함으로써 MSU-유발 복막염을 완화하는 것을 알 수 있었다.As a result, BOT-4-one significantly reduced the number of MSU-mediated total cells and the number of Ly-6G + / F480 neutrophils (FIGS. 8A and B). In addition, MSU-induced IL-1β production was reduced (Figure 8C). Through this, it was found that BOT-4-one relieves MSU-induced peritonitis by inhibiting NLRP3 inflammatory regulatory complex-mediated IL-1β production.

Claims (12)

하기 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온(2-cyclohexylimino-6-methyl-6,7-dihydro-5H-benzo[1,3]oxathiol-4-one)을 포함하는 NLRP3 활성 억제용 시약조성물:
[화학식 1]
Figure 112017091645861-pat00004
.
To the 2-cyclohexyl-6-methyl-6,7-diamino of formula dihydro -5 H - benzo [1,3] oxa thiol-4-one (2-cyclohexylimino-6-methyl -6,7- dihydro-5 H -benzo [1,3] oxathiol-4-one) a reagent composition for NLRP3 inhibition, including:
[Formula 1]
Figure 112017091645861-pat00004
.
제 1항에 있어서, NLRP3의 ATPase 활성을 억제하는, NLRP3 활성 억제용 시약조성물.The reagent composition for inhibiting NLRP3 activity according to claim 1, which inhibits ATPase activity of NLRP3. 제 1항에 있어서, NLRP3을 유비퀴틴화(ubiquitination)하여 NLRP3의 활성을 억제하는, NLRP3 활성 억제용 시약조성물.The reagent composition for inhibiting NLRP3 activity according to claim 1, wherein NLRP3 is inhibited by ubiquitination. 하기 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 포함하는 NLRP3 염증조절복합체(inflammasome) 활성 억제용 시약조성물:
[화학식 1]
Figure 112017091645861-pat00005
.
To 2-cyclohexyl-6-a mino-6,7-dihydro -5 H of the formula (1) benzo [1,3] for NLRP3 inflammation control complex (inflammasome) inhibition, including oxa thiol-4-one Reagent composition:
[Formula 1]
Figure 112017091645861-pat00005
.
제 4항에 있어서, NLRP3 및 ASC(apoptotic speck protein containing a caspase recruitment domain)의 결합을 억제하는, NLRP3 염증조절복합체 활성 억제용 시약조성물.According to claim 4, NLRP3 and inhibiting the binding of ASC (apoptotic speck protein containing a caspase recruitment domain), NLRP3 reagent composition for inhibiting the activity of the inflammatory inflammatory regulatory complex. 하기 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 포함하는 NLRP3 알킬화용 시약조성물:
[화학식 1]
Figure 112020001519462-pat00006
.
NLRP3 reagent composition for the alkylation, including benzo [1,3] oxa thiol-4-one - To a 2-cyclohexyl-6-methyl-6,7-diamino of formula dihydro -5 H:
[Formula 1]
Figure 112020001519462-pat00006
.
하기 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 처리하는 단계를 포함하는, 시험관 내(in vitro) NLRP3 염증조절복합체 활성을 억제하는 방법:
[화학식 1]
Figure 112017091645861-pat00007
.
To the 2-cyclohexyl-6-methyl-6,7-diamino of formula dihydro -5 H - comprising the step of treating the benzo [1,3] oxa thiol-4-one, in vitro (in vitro ) How to inhibit the NLRP3 inflammation control complex activity:
[Formula 1]
Figure 112017091645861-pat00007
.
하기 화학식 1의 2-사이클로헥실이미노-6-메틸-6,7-디하이드로-5H-벤조[1,3]옥사티올-4-온을 처리하는 단계를 포함하는, 시험관 내에서 NLRP3 활성을 억제하는 방법:
[화학식 1]
Figure 112017091645861-pat00008
.
To formula (I) 2-cyclohexyl-6-a mino-6,7-dihydro -5 H of benzo [1,3] NLRP3 activity in, in vitro comprising the step of processing the oxa thiol-4-one To suppress:
[Formula 1]
Figure 112017091645861-pat00008
.
삭제delete 삭제delete 삭제delete 삭제delete
KR1020170121040A 2017-09-20 2017-09-20 Nlrp3 inhibitor KR102116166B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020170121040A KR102116166B1 (en) 2017-09-20 2017-09-20 Nlrp3 inhibitor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020170121040A KR102116166B1 (en) 2017-09-20 2017-09-20 Nlrp3 inhibitor

Publications (2)

Publication Number Publication Date
KR20190032790A KR20190032790A (en) 2019-03-28
KR102116166B1 true KR102116166B1 (en) 2020-05-27

Family

ID=65908181

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020170121040A KR102116166B1 (en) 2017-09-20 2017-09-20 Nlrp3 inhibitor

Country Status (1)

Country Link
KR (1) KR102116166B1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023066825A1 (en) * 2021-10-19 2023-04-27 F. Hoffmann-La Roche Ag Fused bicyclic heteroaryl compounds useful as nlrp3 inhibitors

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102409345B1 (en) * 2020-09-22 2022-06-16 가톨릭대학교 산학협력단 Nlrp3 inflammasome inhibitor and uses thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20090052439A (en) * 2007-11-21 2009-05-26 충북대학교 산학협력단 Dihydrobenzoxathiolone derivatives, method of the same and pharmaceutical composition comprising same

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023066825A1 (en) * 2021-10-19 2023-04-27 F. Hoffmann-La Roche Ag Fused bicyclic heteroaryl compounds useful as nlrp3 inhibitors

Also Published As

Publication number Publication date
KR20190032790A (en) 2019-03-28

Similar Documents

Publication Publication Date Title
Xiang et al. Protective effect of Andrographolide on 5-Fu induced intestinal mucositis by regulating p38 MAPK signaling pathway
KR102116166B1 (en) Nlrp3 inhibitor
KR101928553B1 (en) A Composition for Preventing or Treating Inflammasome Mediated Inflammatory Disease Containing Ginsenoside Compounds
WO2015068940A1 (en) Pharmaceutical composition for preventing and treating osteoporosis comprising artemisinin as active ingredient
JP6000206B2 (en) Antiallergic oral composition
KR101729078B1 (en) Composition for anti-obesity containing enmides
KR102212997B1 (en) A composition for improving, preventing and treating of chronic inflammation diseases caused by excessive NLRP3 inflammasome activation comprising burdock extract
KR20120056243A (en) Composition for preventing and treating inflammatory disease comprising piperine or pharmaceutically acceptable salt thereof as an active ingredient
KR101473965B1 (en) Composition containing sorafenib and imidazopyridine derivative for preventing or treating pancreatic cancer
KR101200671B1 (en) Preventing and treating composition for obesity comprising polygammaglutamic acid
KR20210043152A (en) Pharmacieutical composition comprising Quercetin-3-O-xyloside for prevention and treatment of Type 2 diabetes
KR101715996B1 (en) Composition for antidiabetic activity comprising dichloromethane or ethyl acetate fraction of Hizikia fusiformis extract as effective component
KR20200129596A (en) Composition for Preventing or Treating of Degenerative Brain Disease Comprising Extract of Zanthoxylum piperitum Fruit
KR20110053639A (en) Composition for anti-cancer or immuno-modulating activity comprising morphinane alkaloids compound
KR20190118286A (en) Composition for preventing or treating cancer comprising diallyl disulfide and trail protein
KR101926021B1 (en) Pharmaceutical composition comprising extract of Telectadium dongnaiense for preventing or treating colon cancer
KR20120067715A (en) Composition for treating and preventing lung cancer comprising narigenin and tumor necrosis factor-ralated apoptosis-inducing ligand
KR102135148B1 (en) Pharmaceutical Composition for Treatment and Inhibiting Metastasis of Brain Tumor Comprising Acteoside
KR102284073B1 (en) An Extract of Umbilicaria antarctica Having Anti-inflammatory and Immuno-modulating Activity and Composition Comprising the Same
KR102418556B1 (en) Pharmaceutical composition for Preventing or Treating Immune Disease Comprising butaclamol
KR102144264B1 (en) Composition for preventing and treating a cancer comprising leonurus sibiricus
KR101907908B1 (en) Composition for inhibiting the differentiation of osteoblast and osteoclast and pharmaceutical compostion for preventing or treating bone diseases
KR102155713B1 (en) Composition for preventing or treating cancer comprising sea cucumber extracts or fraction thereof, and trail protein
KR101770395B1 (en) Composition for preventing or treating of autophagy mediated disease comprising conessine as an active ingredient
KR101511090B1 (en) A composition for prevention and treatment of inflammatory diseases

Legal Events

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