KR20230139388A - Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same - Google Patents

Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same Download PDF

Info

Publication number
KR20230139388A
KR20230139388A KR1020220036547A KR20220036547A KR20230139388A KR 20230139388 A KR20230139388 A KR 20230139388A KR 1020220036547 A KR1020220036547 A KR 1020220036547A KR 20220036547 A KR20220036547 A KR 20220036547A KR 20230139388 A KR20230139388 A KR 20230139388A
Authority
KR
South Korea
Prior art keywords
cancer
liver
compound
group
pyrimidin
Prior art date
Application number
KR1020220036547A
Other languages
Korean (ko)
Inventor
박병현
배은주
전라옥
이은
Original Assignee
전북대학교산학협력단
숙명여자대학교산학협력단
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 전북대학교산학협력단, 숙명여자대학교산학협력단 filed Critical 전북대학교산학협력단
Priority to KR1020220036547A priority Critical patent/KR20230139388A/en
Publication of KR20230139388A publication Critical patent/KR20230139388A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Hospice & Palliative Care (AREA)
  • Polymers & Plastics (AREA)
  • Psychiatry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

본 발명은 피라졸로[3,4-d]피리미딘 계열 p21-activated kinase 4 (PAK4) 저해제 및 이를 포함하는 약학 조성물에 관한 것으로, PAK4 억제 활성이 우수할 뿐만 아니라, 간 세포의 산화적 스트레스 및 염증 억제 효과가 우수하므로, 허혈성 재관류에 의한 손상 등 다양한 간 손상 또는 간 염증을 예방 또는 치료하거나, 간 기능을 보호하기 위한 용도로 유용하게 활용될 수 있으며, 또한, 암, 퇴행성뇌질환 등 PAK4가 과발현하는 질환에 효과적으로 적용될 수 있다.The present invention relates to a pyrazolo[3,4-d]pyrimidine series p21-activated kinase 4 (PAK4) inhibitor and a pharmaceutical composition containing the same, which not only has excellent PAK4 inhibitory activity but also reduces oxidative stress and oxidative stress in liver cells. Because it has an excellent anti-inflammatory effect, it can be useful for preventing or treating various liver damage or liver inflammation, such as damage caused by ischemia-reperfusion, or to protect liver function. In addition, PAK4 can be used to protect liver function, such as cancer and degenerative brain disease. It can be effectively applied to diseases that overexpress it.

Description

피라졸로[3,4-d]피리미딘 계열 p21-activated kinase 4 (PAK4) 저해제 및 이를 포함하는 약학 조성물{Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same}Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition containing the same {Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same}

본 발명은 피라졸로[3,4-d]피리미딘 계열 p21-activated kinase 4 (PAK4) 저해제 및 이를 포함하는 약학 조성물에 관한 것이다.The present invention relates to a pyrazolo[3,4-d]pyrimidine series p21-activated kinase 4 (PAK4) inhibitor and a pharmaceutical composition containing the same.

간 종양 절제 및 간 이식과 같은 외과적 절차에 따른 간 허혈성 재관류(Hepatic ischemia/reperfusion, I/R) 손상은 기능 장애 및 간 기능 부전의 주요 원인이다. 간 I/R 손상의 주요 병원성 기전은 세포 사멸 및 염증으로, 혈류 차단으로 인한 초기 저산소 손상은 실질적인 세포 사멸을 유발한다. 재관류 기간 동안 혈류의 회복은 대식세포 및 호중구 침윤 및 사이토카인/케모카인 생성을 포함한 염증 반응을 동반함으로써 간 손상을 더욱 악화시키며, 이러한 현상은 재산소화와 함께 활성산소종(reactive oxygen species, ROS)을 생성하여 추가적인 세포 기능 장애 및 손상을 유발한다.Hepatic ischemia/reperfusion (I/R) injury following surgical procedures such as liver tumor resection and liver transplantation is a major cause of functional impairment and liver failure. The main pathogenic mechanisms of liver I/R injury are apoptosis and inflammation, with initial hypoxic injury due to blood flow obstruction leading to substantial cell death. Restoration of blood flow during the reperfusion period further aggravates liver damage by being accompanied by inflammatory responses including macrophage and neutrophil infiltration and cytokine/chemokine production, which together with reoxygenation produce reactive oxygen species (ROS). This causes further cellular dysfunction and damage.

간세포는 다양한 항산화 단백질의 유도를 포함하는 방어 메커니즘을 통하여 ROS에 의한 조직 손상을 억제하며, 대표적으로 NF-E2 관련 인자 2(NF-E2-related factor 2, Nrf2)가 있다. 기본 조건에서 Nrf2는 억제 인자인 Keap1(Kelch-like ECH-associated protein 1)에 결합하여 세포질에서 격리되어 있는데, 산화 스트레스는 Keap1의 시스테인 잔기를 수정하여 Keap1에서 Nrf2가 해리되고 이후 세포질에서 핵으로 이행한다. 핵 Nrf2는 헴 옥시게나제-1(heme oxygenase-1, HO-1), NAD(P)H 퀴논 산화환원효소 1(NAD(P)H quinone oxidoreductase 1, NQO1) 및 글루타티온 퍼옥시다제(glutathione peroxidase, GPx)와 같은 항산화 효소의 프로모터 영역에 존재하는 항산화 반응 요소(antioxidant response element, ARE)에 결합한다. 이 과정에서 여러 단백질 키나아제가 Nrf2의 세포내 위치와 단백질 안정성에 참여한다. 예를 들어, 단백질 키나아제 C, 카제인 키나아제 2 및 AMP 활성화 키나아제는 Nrf2의 다른 영역을 인산화하고 Nrf2의 핵 전위를 촉진하는 반면, 글리코겐 합성효소 키나제 3β 및 Fyn 매개 인산화는 Nrf2의 세포내 분포를 세포질로 이동시켜 유비퀴틴-프로테아좀 경로를 통해 분해를 유도한다. Hepatocytes suppress tissue damage caused by ROS through a defense mechanism that includes the induction of various antioxidant proteins, most notably NF-E2-related factor 2 (Nrf2). Under basic conditions, Nrf2 binds to the repressor Keap1 (Kelch-like ECH-associated protein 1) and is sequestered in the cytoplasm. Oxidative stress modifies the cysteine residue of Keap1, causing Nrf2 to dissociate from Keap1 and subsequently move from the cytoplasm to the nucleus. do. Nuclear Nrf2 encodes heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase 1 (NQO1), and glutathione peroxidase. , GPx) and binds to the antioxidant response element (ARE) present in the promoter region of antioxidant enzymes. In this process, several protein kinases participate in the intracellular localization and protein stability of Nrf2. For example, protein kinase C, casein kinase 2, and AMP-activated kinase phosphorylate different regions of Nrf2 and promote nuclear translocation of Nrf2, whereas glycogen synthase kinase 3β and Fyn-mediated phosphorylation direct the intracellular distribution of Nrf2 to the cytoplasm. It moves and induces decomposition through the ubiquitin-proteasome pathway.

최근에 발암성 단백질인 p21-activated kinase 4(PAK4)가 T369에서 Nrf2를 인산화하고 Nrf2를 핵에서 세포질로 내보내는 것으로 보고되었으며, 여러 PAK4 억제제가 보고되었다. 고처리량 스크리닝과 결합된 구조 기반 설계를 통해 PF-3758309는 Pfizer에서 처음으로 pan-PAK 억제제로 개발되었으며, 여러 유형의 암에 대해 항종양 효과가 확인되었다. 또한, 최근 Guo et al.은 전구약물 형태로 PAK4 억제제(CZh-226)를 개발하고 암의 쥐 모델에서 개선된 약동학 및 조직 분포 및 내약성이 실험되었다. 다만, 앞서 언급한 PAK4 억제제의 사용은 약한 효능 및/또는 PAK4 선택성이 낮은 것으로 나타나 임상에 부적합한 것으로 보고되었다.Recently, the oncogenic protein p21-activated kinase 4 (PAK4) was reported to phosphorylate Nrf2 at T369 and export Nrf2 from the nucleus to the cytoplasm, and several PAK4 inhibitors have been reported. Through structure-based design combined with high-throughput screening, PF-3758309 was developed by Pfizer as the first pan-PAK inhibitor, and its antitumor effects were confirmed against several types of cancer. Additionally, Guo et al. recently developed a PAK4 inhibitor (CZh-226) in prodrug form and tested its improved pharmacokinetics, tissue distribution, and tolerability in a rat model of cancer. However, the use of the aforementioned PAK4 inhibitors was reported to be unsuitable for clinical use due to weak efficacy and/or low PAK4 selectivity.

이에, 본 발명자들은 간 손상 치료 및 간 보호에 적용할 수 있는 신규 화합물을 발굴하기 위한 연구를 수행하여 본 발명을 완성하였다.Accordingly, the present inventors conducted research to discover new compounds applicable to liver damage treatment and liver protection and completed the present invention.

대한민국 특허등록 제10-1706017호Republic of Korea Patent Registration No. 10-1706017

본 발명의 하나의 목적은 하기 화학식 1로 표시되는 화합물, 이의 약학적으로 허용되는 염, 이의 수화물, 이의 용매화물 및 이의 이성질체로 이루어진 군으로부터 선택되는 화합물을 제공하는 것이다:One object of the present invention is to provide a compound selected from the group consisting of a compound represented by the following formula (1), a pharmaceutically acceptable salt thereof, a hydrate thereof, a solvate thereof, and an isomer thereof:

상기 화학식 1에서,In Formula 1,

R1 및 R2는 서로 같거나, 서로 다르거나, 또는 동일한 고리(A)상에 위치하는 것으로써, C1-6알킬, C1-6할로알킬, -(CH2)m-C1-6알콕시(이때, m은 1 내지 6의 정수), C3-8사이클로알킬, C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고, R 1 and R 2 are the same, different from each other, or are located on the same ring (A), and are C 1-6 alkyl, C 1-6 haloalkyl, -(CH 2 ) m -C 1- 6 is a substituent selected from the group consisting of alkoxy (where m is an integer of 1 to 6), C 3-8 cycloalkyl, and C 6-12 aryl,

상기 고리(A)는 C3-8사이클로알킬, C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고,The ring (A) is a substituent selected from the group consisting of C 3-8 cycloalkyl and C 6-12 aryl,

R3는 -CH2-R7R8 이거나, 또는 R3는 R4 및 R5와 동일한 방향족 고리(B)에 위치할 수 있고,R 3 may be -CH 2 -R 7 R 8 , or R 3 may be located in the same aromatic ring (B) as R 4 and R 5 ,

상기 방향족 고리(B)는 C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고,The aromatic ring (B) is a substituent selected from the group consisting of C 6-12 aryl,

R5 및 R6는 서로 같거나 또는 서로 다른 것으로써, C 또는 N이고,R 5 and R 6 are the same or different from each other and are C or N,

상기 R7은 H, C1-6알킬, C2-6알케닐 또는 C2-6알키닐로 이루어진 군으로부터 선택되는 치환기이고,R 7 is a substituent selected from the group consisting of H, C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl,

상기 R8은 H, O 또는 N으로부터 선택된 헤테로원자가 1 내지 2개 포함된 C3-8헤테로고리 치환 C1-6 알킬로 이루어진 군으로부터 선택되는 치환기이다.The R 8 is a substituent selected from the group consisting of C 3-8 heterocyclic substituted C 1-6 alkyl containing 1 to 2 heteroatoms selected from H, O or N.

본 발명의 다른 목적은 4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-메틸부틴-2-올; 3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐에틴일사이클로헥산-1-올; 4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-메틸부틴일-1,2-다이올; 4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-페닐부틴-2-올; 3-[4-아미노-3-(3-몰포린-1-일프로필)피라졸로[3,4-d]피리미딘-1-일]페닐에틴일)사이클로헥산올; 3-[4-아미노-3-(3-피페리딘-1-일프로필)피라졸로[3,4-d]피리미딘-1-일]페닐에틴일)사이클로헥산올; 4-(3-(1-아미노피리도[4,3-b]인돌-5-일)페닐)-2-메틸부틴-2-올; 3-(1-아미노피리도[4,3-b]인돌-5-일)페닐에틴일사이클로엑산-1-올; 및 4-(3-(1-아미노피리도[4,3-b]인돌-5-일)페닐)-2-메틸부틴-1,2-다이올로 이루어진 군으로부터 선택되는 화합물을 제공하는 것이다.Another object of the present invention is 4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutyn-2-ol; 3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol; 4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutynyl-1,2-diol; 4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-phenylbutyn-2-ol; 3-[4-amino-3-(3-morpholin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol; 3-[4-amino-3-(3-piperidin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol; 4-(3-(1-aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyn-2-ol; 3-(1-aminopyrido[4,3- b ]indol-5-yl)phenylethynylcycloexan-1-ol; and 4-(3-(1-aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyne-1,2-diol. .

본 발명의 또 다른 목적은 3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐에틴일사이클로헥산-1-올인 것인 화합물을 제공하는 것이다.Another object of the present invention is to provide a compound that is 3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol.

본 발명의 또 다른 목적은 상기 화합물을 포함하는 PAK4 발현 증가 질환 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition containing the above compound for preventing or treating diseases with increased PAK4 expression.

본 발명의 또 다른 목적은 상기 화합물을 포함하는 간 기능 보호용 조성물을 제공하는 것이다.Another object of the present invention is to provide a composition for protecting liver function containing the above compound.

본 발명의 또 다른 목적은 상기 화합물을 포함하는 간 기능 보호용 건강기능식품을 제공하는 것이다.Another object of the present invention is to provide a health functional food for protecting liver function containing the above compound.

본 발명의 일 양상은 하기 화학식 1로 표시되는 화합물, 이의 약학적으로 허용되는 염, 이의 수화물, 이의 용매화물 및 이의 이성질체로 이루어진 군으로부터 선택되는 화합물을 제공한다:One aspect of the present invention provides a compound selected from the group consisting of a compound represented by the following formula (1), a pharmaceutically acceptable salt thereof, a hydrate thereof, a solvate thereof, and an isomer thereof:

[화학식 1][Formula 1]

상기 화학식 1에서,In Formula 1,

R1 및 R2는 서로 같거나, 서로 다르거나, 또는 동일한 고리(A)상에 위치하는 것으로써, C1-6알킬, C1-6할로알킬, -(CH2)m-C1-6알콕시(이때, m은 1 내지 6의 정수), C3-8사이클로알킬, C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고, R 1 and R 2 are the same, different from each other, or are located on the same ring (A), and are C 1-6 alkyl, C 1-6 haloalkyl, -(CH 2 ) m -C 1- 6 is a substituent selected from the group consisting of alkoxy (where m is an integer of 1 to 6), C 3-8 cycloalkyl, and C 6-12 aryl,

상기 고리(A)는 C3-8사이클로알킬, C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고,The ring (A) is a substituent selected from the group consisting of C 3-8 cycloalkyl and C 6-12 aryl,

R3는 -CH2-R7R8 이거나, 또는 R3는 R4 및 R5와 동일한 방향족 고리(B)에 위치할 수 있고,R 3 may be -CH 2 -R 7 R 8 , or R 3 may be located in the same aromatic ring (B) as R 4 and R 5 ,

상기 방향족 고리(B)는 C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고,The aromatic ring (B) is a substituent selected from the group consisting of C 6-12 aryl,

R5 및 R6는 서로 같거나 또는 서로 다른 것으로써, C 또는 N이고,R 5 and R 6 are the same or different from each other and are C or N,

상기 R7은 H, C1-6알킬, C2-6알케닐 또는 C2-6알키닐로 이루어진 군으로부터 선택되는 치환기이고,R 7 is a substituent selected from the group consisting of H, C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl,

상기 R8은 H, O 또는 N으로부터 선택된 헤테로원자가 1 내지 2개 포함된 C3-8헤테로고리 치환 C1-6 알킬로 이루어진 군으로부터 선택되는 치환기이다.The R 8 is a substituent selected from the group consisting of C 3-8 heterocyclic substituted C 1-6 alkyl containing 1 to 2 heteroatoms selected from H, O or N.

본 발명의 화학식 1로 표시되는 화합물은 PAK4 억제제로 작용하며, 허혈성 재관류 손상이 있는 마우스에서 아미노트랜스퍼라제 및 전염증성 사이토카인, 간세포 괴사와 세포자멸사, 염증 세포 침윤의 혈청 수준을 현저히 감소시킬 수 있을 뿐만 아니라, Nrf2를 안정화할 수 있고, 저산소증-재산소화로 인한 간 세포의 세포 사멸 세포사 및 염증을 감소시킬 수 있으므로, 간 손상, 특히 허혈성 재관류에 의한 간 손상의 완화에 효과적으로 적용될 수 있다. 또한, 암, 퇴행성뇌질환 등 PAK4가 과발현하는 질환에 효과적으로 적용될 수 있다.The compound represented by Formula 1 of the present invention acts as a PAK4 inhibitor and can significantly reduce serum levels of aminotransferases and pro-inflammatory cytokines, hepatocyte necrosis and apoptosis, and inflammatory cell infiltration in mice with ischemia-reperfusion injury. In addition, it can stabilize Nrf2 and reduce apoptotic cell death and inflammation of liver cells caused by hypoxia-reoxygenation, so it can be effectively applied to alleviating liver damage, especially liver damage caused by ischemic reperfusion. In addition, it can be effectively applied to diseases in which PAK4 is overexpressed, such as cancer and degenerative brain disease.

본 발명의 화합물은 다음과 같은 방법으로 합성될 수 있다(도 2). 말로노니트릴로부터 합성된(a) 엔올에터(1)를 히드라진으로 처리(b)하여 아미노피라졸 고리(11)를 얻은 다음, 포름아미드로 고리화하여(c) 4-아미노피라졸로[3,4-d]피리미딘 스캐폴드(12)를 제공하고, 후속적인 Ullmann 반응(d) 및 Sonogashira 커플링(e) 반응을 통하여 본 발명의 화학식 1로 표시되는 다양한 화합물(14)이 합성될 수 있다. 추가적으로(도 3), 용매에 접근할 수 있는 부분을 4-아미노피라졸로[3,4-d]피리미딘 골격에 도입하고(a), 3-요오도피라졸로[3,4-d]피리미딘과 치환된 프로핀의 Sonogashira 커플링을 통하여 용매 접근 가능한 모이어티를 도입하고 수소화한 다음(b), Ullmann-형 커플링(c) 및 Sonogashira 커플링(d) 반응을 순차적으로 수행하여 화학식 1로 표시되는 다양한 화합물(18)을 더 합성할 수 있다.The compounds of the present invention can be synthesized by the following method (Figure 2). The enolether (1) synthesized from malononitrile (a) is treated with hydrazine (b) to obtain an aminopyrazole ring (11), and then cyclized with formamide (c) to form 4-aminopyrazolo [3 ,4-d]pyrimidine scaffold (12) is provided, and various compounds (14) represented by Formula 1 of the present invention can be synthesized through the subsequent Ullmann reaction (d) and Sonogashira coupling (e) reaction. there is. Additionally (Figure 3), a solvent-accessible moiety was introduced into the 4-aminopyrazolo[3,4-d]pyrimidine skeleton (a), and 3-iodopyrazolo[3,4-d]pyrimidine A solvent-accessible moiety is introduced and hydrogenated through Sonogashira coupling of mydine and substituted propyne (b), and then Ullmann-type coupling (c) and Sonogashira coupling (d) reactions are sequentially performed to obtain Formula 1 Various compounds (18) represented by can be further synthesized.

본 발명의 다른 양상은 4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-메틸부틴-2-올; 3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐에틴일사이클로헥산-1-올; 4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-메틸부틴일-1,2-다이올; 4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-페닐부틴-2-올; 3-[4-아미노-3-(3-몰포린-1-일프로필)피라졸로[3,4-d]피리미딘-1-일]페닐에틴일)사이클로헥산올; 및 3-[4-아미노-3-(3-피페리딘-1-일프로필)피라졸로[3,4-d]피리미딘-1-일]페닐에틴일)사이클로헥산올; 4-(3-(1-아미노피리도[4,3-b]인돌-5-일)페닐)-2-메틸부틴-2-올; 3-(1-아미노피리도[4,3-b]인돌-5-일)페닐에틴일사이클로엑산-1-올; 및 4-(3-(1-아미노피리도[4,3-b]인돌-5-일)페닐)-2-메틸부틴-1,2-다이올로 이루어진 군으로부터 선택되는 화합물을 제공한다.Another aspect of the invention is 4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutyn-2-ol; 3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol; 4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutynyl-1,2-diol; 4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-phenylbutyn-2-ol; 3-[4-amino-3-(3-morpholin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol; and 3-[4-amino-3-(3-piperidin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol; 4-(3-(1-aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyn-2-ol; 3-(1-aminopyrido[4,3- b ]indol-5-yl)phenylethynylcycloexan-1-ol; and 4-(3-(1-aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyn-1,2-diol.

본 발명의 또 다른 양상은 3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐에틴일사이클로헥산-1-올인 것인 화합물을 제공한다.Another aspect of the invention provides a compound that is 3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol.

본 발명의 또 다른 양상은 상기 화합물을 포함하는 PAK4 발현 증가 질환 예방 또는 치료용 약학적 조성물을 제공한다.Another aspect of the present invention provides a pharmaceutical composition for preventing or treating diseases with increased PAK4 expression, comprising the above compound.

본 발명의 약학적 조성물은 약학적으로 허용되는 담체를 포함할 수 있다. 본 발명의 약학적 조성물에 포함되는 약학적으로 허용되는 담체는 약제의 제조에 통상적으로 이용되는 것으로써, 락토오스, 덱스트로스, 수크로오스, 솔비톨, 만니톨, 전분, 아카시아 고무, 인산칼슘, 알기네이트, 젤라틴, 규산칼슘, 미세결정성 셀룰로오스, 폴리비닐피롤리돈, 셀룰로오스, 물, 시럽, 메틸 셀룰로오스, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 활석, 스테아르산 마그네슘 및 미네랄 오일 등을 포함하나, 이에 한정되는 것은 아니다. 본 발명의 약학적 조성물은 상기 성분들 이외에 윤활제, 습윤제, 감미제, 향미제, 유화제, 현탁제, 보존제 등을 추가로 포함할 수 있다. 적합한 약학적으로 허용되는 담체 및 제제는 Remington's Pharmaceutical Sciences (22th ed., 2013)에 상세히 기재되어 있다.The pharmaceutical composition of the present invention may include a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers included in the pharmaceutical composition of the present invention are those commonly used in the manufacture of drugs, and include lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum acacia, calcium phosphate, alginate, and gelatin. , calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil, etc. It is not limited. In addition to the above ingredients, the pharmaceutical composition of the present invention may further include lubricants, wetting agents, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, etc. Suitable pharmaceutically acceptable carriers and formulations are described in detail in Remington's Pharmaceutical Sciences (22nd ed., 2013).

본 발명의 일 구체예에 따른 약학적 조성물은 하나 이상의 PAK4 억제 활성을 나타내는 물질과 함께 투여될 수 있다.The pharmaceutical composition according to one embodiment of the present invention may be administered together with one or more substances exhibiting PAK4 inhibitory activity.

또한, 본 발명의 일 구체예에 따른 약학적 조성물은 PAK4 발현 증가 질환의 예방 또는 치료를 위하여 단독으로, 또는 수술, 호르몬 치료, 약물치료 및/또는 생물학적 반응 조절제를 사용하는 방법들과 병용하여 사용될 수 있다.In addition, the pharmaceutical composition according to one embodiment of the present invention may be used alone or in combination with methods using surgery, hormone therapy, drug therapy, and/or biological response regulators for the prevention or treatment of diseases with increased PAK4 expression. You can.

본 발명의 약학적 조성물은 그 제형의 제제화에 필요하고 적절한 각종 기제 및/또는 첨가물을 포함할 수 있으며, 그 효과를 감소시키지 않는 범위 내에서 비이온 계면활성제, 실리콘 폴리머, 체질안료, 향료, 방부제, 살균제, 산화 안정화제, 유기 용매, 이온성 또는 비이온성 증점제, 유연화제, 산화방지제, 자유 라디칼 파괴제, 불투명화제, 안정화제, 에몰리언트(emollient), 실리콘, α-히드록시산, 소포제, 보습제, 비타민, 곤충 기피제, 향료, 보존제, 계면활성제, 소염제, 물질 P 길항제, 충전제, 중합체, 추진제, 염기성화 또는 산성화제, 또는 착색제 등 공지의 화합물을 더 포함하여 제조될 수 있다.The pharmaceutical composition of the present invention may contain various bases and/or additives necessary and appropriate for the formulation of the dosage form, and may include nonionic surfactants, silicone polymers, extenders, fragrances, and preservatives to the extent that they do not reduce the effectiveness. , disinfectants, oxidation stabilizers, organic solvents, ionic or non-ionic thickeners, softeners, antioxidants, free radical destroyers, opacifiers, stabilizers, emollients, silicones, α-hydroxy acids, anti-foaming agents, humectants. , vitamins, insect repellent, fragrance, preservative, surfactant, anti-inflammatory agent, substance P antagonist, filler, polymer, propellant, alkalinizing or acidifying agent, or colorant.

본 발명의 약학적 조성물의 적합한 투여량은 제제화 방법, 투여 방식, 환자의 연령, 체중, 성, 병적 상태, 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감응성과 같은 요인들에 의해 다양하게 처방될 수 있다. 본 발명의 약학적 조성물의 투여량은 성인 기준으로 0.001~1000㎎/kg일 수 있다.The appropriate dosage of the pharmaceutical composition of the present invention varies depending on factors such as formulation method, administration method, patient's age, weight, sex, pathological condition, food, administration time, administration route, excretion rate, and reaction sensitivity. It can be. The dosage of the pharmaceutical composition of the present invention may be 0.001 to 1000 mg/kg for adults.

본 발명의 약학적 조성물은 경구 또는 비경구로 투여될 수 있다.The pharmaceutical composition of the present invention can be administered orally or parenterally.

본 발명의 약학적 조성물은 경구 투여 시 다양한 제형으로 투여될 수 있는데, 환제, 분말제, 과립제, 정제 또는 캡슐제 등의 고형제제 형태로 투여될 수 있으며, 여러 가지 부형제, 예를 들어, 습윤제, 감미제, 방향제, 보존제 등을 더 포함할 수 있다. 구체적으로, 본 발명의 조성물을 분말, 과립, 정제 또는 캅셀 형태로 제형화할 경우, 이의 제조에 통상적으로 사용하는 적절한 담체, 부형제 및 희석제를 더 포함할 수 있다. 상기 담체, 부형제 및 희석제로는 예를 들어, 락토오스, 덱스트로스, 수크로오스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알기네이트, 젤라틴, 인산칼슘, 규산칼슘, 셀룰로오스, 메틸 셀룰로오스, 미정질 셀룰로오스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및/또는 광물유가 사용될 수 있으나, 이에 한정되는 것은 아니다. 또한, 제제화에 일반적으로 사용되는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 포함하여 조제될 수 있으며, 상기 부형제 이외에 마그네슘 스테아레이트 또는 탈크 같은 윤활제를 더 포함할 수 있다.The pharmaceutical composition of the present invention can be administered in various dosage forms when administered orally. It may be administered in the form of solid preparations such as pills, powders, granules, tablets, or capsules, and may be administered in the form of various excipients, such as wetting agents, It may further include sweeteners, aromatics, preservatives, etc. Specifically, when the composition of the present invention is formulated in the form of powder, granule, tablet or capsule, it may further include appropriate carriers, excipients and diluents commonly used in its production. The carriers, excipients and diluents include, for example, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, Microcrystalline cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and/or mineral oil may be used, but are not limited thereto. In addition, it may be prepared including diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants commonly used in formulation, and may further include lubricants such as magnesium stearate or talc in addition to the above excipients. .

본 발명의 약학적 조성물은 비경구 투여시 다양한 제형으로 투여될 수 있는데, 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 액상제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순희석제인 물, 리퀴드, 파라핀 이외에 여러 가지 부형제, 예를 들어, 습윤제 감미제, 방향제, 보존제 등이 포함될 수 있다. 구체적으로, 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제 및 동결건조제제가 포함될 수 있다. 비수성용제, 현탁용제로는 프로필렌글리콜(Propylene glycol), 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 또한, 치료제의 효능 증진을 위해 칼슘이나 비타민 D3를 첨가할 수 있다. The pharmaceutical composition of the present invention can be administered in various dosage forms when administered parenterally. Solid preparations include tablets, pills, powders, granules, capsules, etc., and liquid preparations include suspensions, oral solutions, emulsions, and syrups. These include, in addition to the commonly used simple diluents such as water, liquid, and paraffin, various excipients such as humectants, sweeteners, fragrances, and preservatives may be included. Specifically, preparations for parenteral administration may include sterilized aqueous solutions, non-aqueous solvents, suspensions, emulsions, and freeze-dried preparations. Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate. Additionally, calcium or vitamin D3 can be added to improve the efficacy of the treatment.

이러한 조성물은 단위-용량(1회분) 또는 다중-용량(수 회분) 용기, 예를 들어, 밀봉된 앰플 및 바이알에 제시될 수 있고, 사용 직전에 멸균성 액상 담체, 예를 들어, 주사용 수의 부가만을 요구하는 동결-건조 조건하에 저장될 수 있다. 즉석의 사용제 및 현탁제는 멸균성 산제, 과립제 및 정제로부터 제조할 수 있다.These compositions may be presented in unit-dose (single-dose) or multi-dose (several-dose) containers, such as sealed ampoules and vials, and may be immersed in a sterile liquid carrier, e.g., water for injection, immediately before use. Can be stored under freeze-drying conditions requiring only the addition of. Ready-to-use preparations and suspensions can be prepared from sterile powders, granules and tablets.

본 발명의 일 구체예에 따르면, 상기 PAK4 발현 증가 질환은 암, 신경계 질환, 간 손상, 간 염증, 멜라닌 색소 침착으로 이루어진 군으로부터 선택되는 하나 이상의 질환일 수 있다.According to one embodiment of the present invention, the disease with increased PAK4 expression may be one or more diseases selected from the group consisting of cancer, neurological disease, liver damage, liver inflammation, and melanin pigmentation.

본 발명의 일 구체예에 따르면, 상기 암은 위암, 폐암, 간암, 대장암, 소장암, 췌장암, 뇌암, 뼈암, 흑색종, 유방암, 경화성선증, 자궁암, 자궁경부암, 두경부암, 식도암, 갑상선암, 부갑상선암, 신장암, 육종, 전립선암, 요도암, 방광암, 혈액암, 림프 종, 건선, 또는 섬유선종으로 이루어진 군으로부터 선택되는 하나 이상의 암일 수 있다.According to one embodiment of the present invention, the cancer includes stomach cancer, lung cancer, liver cancer, colon cancer, small intestine cancer, pancreatic cancer, brain cancer, bone cancer, melanoma, breast cancer, sclerosing gonadotropin, uterine cancer, cervical cancer, head and neck cancer, esophageal cancer, thyroid cancer, It may be one or more cancers selected from the group consisting of parathyroid cancer, kidney cancer, sarcoma, prostate cancer, urethral cancer, bladder cancer, hematological cancer, lymphoma, psoriasis, or fibroadenoma.

본 발명의 일 구체예에 따르면, 상기 신경계 질환은 퇴행성뇌질환일 수 있다.According to one embodiment of the present invention, the neurological disease may be a degenerative brain disease.

본 발명의 일 구체예에 따르면, 상기 퇴행성뇌질환은 알츠하이머병, 파킨슨병, 진행성 핵상마비, 다계통 위축증, 감람핵-뇌교-소뇌 위축증 (OPCA), 샤이-드래거 증후군, 선조체-흑질 퇴행증, 헌팅톤병, 근위축성 측색 경화증 (ALS), 본태성 진전증, 피질-기저핵 퇴행증, 미만성 루이 소체 질환), 파킨스-ALS-치매 복합증, 픽병, 뇌허혈 및 뇌경색으로 이루어진 군으로부터 선택되는 하나 이상의 질환일 수 있다.According to one embodiment of the present invention, the degenerative brain diseases include Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, multiple system atrophy, olivary nucleus-pontine-cerebellar atrophy (OPCA), Shay-Drager syndrome, and striatal-substantia nigra degeneration. , Huntington's disease, amyotrophic lateral sclerosis (ALS), essential tremor, cortico-basal gangrene, diffuse Lewy body disease), Parkinson-ALS-dementia complex, Pick's disease, cerebral ischemia, and cerebral infarction. It could be a disease.

본 발명의 일 구체예에 따르면, 상기 간 손상은 염증에 의한 손상, 산화적 손상, 알코올성 손상 및 허혈성 재관류 손상으로 이루어진 군으로부터 선택되는 하나 이상의 손상일 수 있다..According to one embodiment of the present invention, the liver damage may be one or more damage selected from the group consisting of inflammatory damage, oxidative damage, alcoholic damage, and ischemia-reperfusion damage.

본 발명의 또 다른 양상은 상기 화합물을 포함하는 간 기능 보호용 조성물을 제공한다.Another aspect of the present invention provides a composition for protecting liver function containing the above compound.

본 발명의 간 기능 보호용 조성물은 화학식 1로 표시되는 화합물을 유효성분으로 함유하므로, 다양한 산화적 스트레스, 염증 반응, 알코올성 손상, 저산소 및 재산소화 등 다양한 간 손상 원인으로부터 간 기능을 효과적으로 보호할 수 있다.Since the composition for protecting liver function of the present invention contains the compound represented by Formula 1 as an active ingredient, it can effectively protect liver function from various causes of liver damage such as various oxidative stress, inflammatory reaction, alcoholic damage, hypoxia and reoxygenation. .

본 발명의 또 다른 양상은 상기 화합물을 포함하는 간 기능 보호용 건강기능식품을 제공한다.Another aspect of the present invention provides a health functional food for protecting liver function containing the above compound.

본 발명의 건강기능식품 조성물은, 유효성분으로써 화학식 1로 표시되는 화합물 외에, 건강기능식품 제조 시에 통상적으로 첨가되는 성분을 포함할 수 있으며, 예를 들어, 단백질, 탄수화물, 지방, 영양소, 조미제 및 향미제를 포함할 수 있다. 탄수화물의 예는 모노사카라이드, 예를 들어, 포도당, 과당 등; 디사카라이드, 예를 들어 말토스, 슈크로스, 올리고당 등; 및 폴리사카라이드, 예를 들어 덱스트린, 사이클로덱스트린 등과 같은 통상적인 당 및 자일리톨, 소르비톨, 에리트리톨 등의 당알콜일 수 있다. 향미제로서 천연 향미제[타우마틴, 스테비아 추출물(예를 들어 레바우디오시드 A, 글리시르히진 등)] 및 합성 향미제(사카린, 아스파르탐 등)를 사용할 수 있다.The health functional food composition of the present invention may contain, in addition to the compound represented by Formula 1 as an active ingredient, ingredients commonly added when manufacturing health functional foods, such as proteins, carbohydrates, fats, nutrients, and seasonings. It may include agents and flavoring agents. Examples of carbohydrates include monosaccharides such as glucose, fructose, etc.; Disaccharides such as maltose, sucrose, oligosaccharides, etc.; and polysaccharides, for example, common sugars such as dextrin and cyclodextrin, and sugar alcohols such as xylitol, sorbitol, and erythritol. As a flavoring agent, natural flavoring agents (thaumatin, stevia extract (e.g., rebaudioside A, glycyrrhizin, etc.)) and synthetic flavoring agents (saccharin, aspartame, etc.) can be used.

예를 들어, 본 발명의 건강기능식품이 드링크제로 제조되는 경우에는 본 발명의 화학식 1로 표시되는 화합물 외에 구연산, 액상과당, 설탕, 포도당, 초산, 사과산, 과즙, 두충 추출액, 대추 추출액 및/또는 감초 추출액 등이 추가로 포함될 수 있다. For example, when the health functional food of the present invention is manufactured as a drink, in addition to the compound represented by Formula 1 of the present invention, citric acid, high fructose corn syrup, sugar, glucose, acetic acid, malic acid, fruit juice, eucommia extract, jujube extract and/or Licorice extract, etc. may be additionally included.

또한, 본 발명의 건강기능식품은 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, 알콜, 탄산음료에 사용되는 탄산화제 등을 함유할 수 있다. In addition, the health functional food of the present invention contains various nutrients, vitamins, minerals (electrolytes), flavoring agents such as synthetic and natural flavors, colorants and thickening agents (cheese, chocolate, etc.), pectic acid and its salts, and alginic acid. and its salts, organic acids, protective colloidal thickeners, pH adjusters, stabilizers, preservatives, glycerin, alcohol, carbonating agents used in carbonated beverages, etc.

이러한 성분은 독립적으로 또는 조합하여 사용할 수 있으며, 이러한 첨가제의 비율은 본 발명의 건강기능식품 100 중량부 당 0 내지 약 20 중량부의 범위에서 선택될 수 있으나, 이에 한정되는 것은 아니다.These ingredients can be used independently or in combination, and the ratio of these additives can be selected in the range of 0 to about 20 parts by weight per 100 parts by weight of the health functional food of the present invention, but is not limited thereto.

피라졸로[3,4-d]피리미딘 계열 p21-activated kinase 4 (PAK4) 저해제 및 이를 포함하는 약학 조성물에 따르면, PAK4 억제 활성이 우수할 뿐만 아니라, 간 세포의 산화적 스트레스 및 염증 억제 효과가 우수하므로, 허혈성 재관류에 의한 손상 등 다양한 간 손상 또는 간 염증을 예방 또는 치료하거나, 간 기능을 보호하기 위한 용도로 유용하게 활용될 수 있으며, 또한, 암, 퇴행성뇌질환 등 PAK4가 과발현하는 질환에 효과적으로 적용될 수 있다.According to the pyrazolo[3,4-d]pyrimidine series p21-activated kinase 4 (PAK4) inhibitor and a pharmaceutical composition containing the same, it not only has excellent PAK4 inhibitory activity, but also has an effect of suppressing oxidative stress and inflammation in liver cells. Because it is excellent, it can be usefully used to prevent or treat various liver damage or liver inflammation, such as damage caused by ischemia-reperfusion, or to protect liver function. It can also be used to treat diseases in which PAK4 is overexpressed, such as cancer and degenerative brain disease. It can be applied effectively.

도 1은 신규 PAK4 선택적 억제제에 대한 설계 전략을 나타낸 그림이다.
도 2는 1-아미노피리도[4,3-b]인돌 유도체의 합성을 나타낸 그림이다: (a) 트리메틸 오르토아세테이트(trimethyl orthoacetate), 100℃, (b) DMF-DMA, MeOH, 환류, (c) 48% HBr, AcOH, 실온, (d) 농축 HCl, 160℃, (e) 페닐 히드라진(phenyl hydrazine), Ph2O, 240℃, (f) POCl3, 환류, (g) p-메톡시벤질아민(p-methoxybenzylamine), 200℃, (h) 1-브로모-3-요오도벤젠(1-bromo-3-iodobenzene), Cu, K2CO3, 18-크라운-6, 1,2-디클로로벤젠(18-crown-6, 1,2-dichlorobenzene), 180℃, (i) TFA, 60℃, (j) R-prop-2-yn-1-ol, Pd(PPh3)2Cl2, TEA/DMSO(2:1), 70℃.
도 3은 4-아미노피라졸로[3,4-d]피리미딘 유도체의 합성 과정을 나타낸 그림이다: (a) 트리메틸 오르토아세테이트(trimethyl orthoacetate), 100℃, (b) 히드라진일수화물(hydrazine monohydrate), EtOH, 80℃, (c) 포름아마이드(formamide), 180℃, (d) 1-브로모-3-요오도벤젠(1-bromo-3-iodobenzene), CuI, K2CO3, DMEDA, DMF, 110℃, (e) R-prop-2-yn-1-ol, Pd(PPh3)2Cl2, TEA/DMSO(2:1), 70℃.
도 4는 용매 접근기를 도입한 4-아미노피라졸로[3,4-d]피리미딘 유도체의 합성과정을 나타낸 그림이다: (a) 트리메틸 오르토아세테이트(trimethyl orthoacetate), 100℃, (b) DMF-DMA, MeOH, 환류, (c) 48% HBr, AcOH, 실온, (d) 농축 HCl, 160℃, (e) 페닐 히드라진(phenyl hydrazine), Ph2O, 240℃, (f) POCl3, 환류, (g) p-메톡시벤질아민(p-methoxybenzylamine), 200℃, (h) 1-브로모-3-요오도벤젠(1-bromo-3-iodobenzene), Cu, K2CO3, 18-크라운-6, 1,2-디클로로벤젠(18-crown-6, 1,2-dichlorobenzene), 180℃, (i) TFA, 60℃, (j) R-prop-2-yn-1-ol, Pd(PPh3)2Cl2, TEA/DMSO(2:1), 70℃.
도 5는 SPA7012, SPA7016 및 SPA7017에 대한 효소 IC50 곡선을 나타낸 그래프이다.
도 6은 C57BL/6 마우스에 대한 SPA7012 처리 방법을 나타낸 개략도이다.
도 7은 AST 및 ALT의 혈청 수준(n=5)을 나타낸 그래프이다.
도 8 및 도 9는 간의 전체 형태, 간 단면의 현미경 사진 및 괴사 면적 측정(n=5) 결과를 나타낸 사진 및 그래프이다.
도 10은 간 조직에서 TUNEL 양성 세포 사멸 세포의 (a) 면역형광 염색 사진 및 (b) 정량화(n=5) 결과를 나타낸 그래프이다.
도 11은 간 조직에서 세포 사멸 관련 단백질의 (a) 웨스턴 블로팅 사진 및 (b) 분석(n=5) 결과를 나타낸 그래프이다: **, p<0.01. I/R(허혈-재관류), HPF(high power field).
도 12는 말론디알데히드(MDA)의 간 수치(n=5)를 나타낸 그래프이다.
도 13은 글루타티온(GSH)의 간 수치(n=5)를 나타낸 그래프이다.
도 14는 간 조직의 4-하이드록시노넨알(4-HNE)에 대한 (a) 면역조직화학 염색(n=5) 사진 및 (b) 결과를 나타낸 그래프이다.
도 15는 24시간 형질감염 후 HEK293T 세포의 co-IP 분석 결과를 나타낸 그림이다.
도 16은 I/R 손상된 간 조직의 핵(NE) 및 세포질 추출물(CE)의 Nrf2 단백질 수준을 나타낸 사진이다.
도 17은 ARE 24시간 형질감염 후 HEK293T 세포에서의 루시퍼라제 활성(n=5)을 나타낸 그래프이다.
도 18 및 도 19는 Nrf2 및 간 조직의 표적 유전자 수준(n=5)을 나타낸 결과이다: **, p<0.01. I/R(허혈-재관류), HPF(high power field), V(vehicle), S(SPA7012 50 mg/kg).
도 20은 간 조직에서 F4/80-양성 대식세포 및 Ly6G-양성 호중구의 (a) 면역형광 염색 사진 및 (b)(c) 이를 각각 정량화(n=5)한 결과를 나타낸 그래프이다.
도 21은 혈청 및 간 조직에서 전염증성 사이토카인/케모카인의 단백질 수준을 나타낸 그래프이다.
도 22는 혈청 및 간 조직에서 전염증성 사이토카인/케모카인의 mRNA 수준을 나타낸 그래프이다.
도 23은 NF-κB 신호 전달 경로의 단백질 수준을 나타낸 결과이다: **, p<0.01. HPF(high power field).
도 24는 1차 간세포에 대한 SPA7012 처리 방법을 나타낸 개략도이다.
도 25는 세포 생존력에 대한 MTT 분석(n=5) 결과를 나타낸 그래프이다.
도 26은 12시간 재산소화 후 1차 간세포로부터의 젖산 탈수소효소(LDH) 방출(n=5) 결과를 나타낸 그래프이다.
도 27은 Annexin-V 염색에 의한 세포 사멸 분석(n=5) 결과이다.
도 28은 웨스턴 블롯팅에 의한 세포 사멸 분석(n=5) 결과이다.
도 29는 배양 배지 및 간세포에서 전염증성 사이토카인/케모카인의 단백질 수준(n=5)을 나타낸 그래프이다: *, p<0.05 및 **, p<0.01. H/R(저산소증-재산소화).
도 30은 배양 배지 및 간세포에서 전염증성 사이토카인/케모카인의 mRNA 수준(n=5)을 나타낸 그래프이다: *, p<0.05 및 **, p<0.01. H/R(저산소증-재산소화).
Figure 1 is a diagram showing the design strategy for a new PAK4 selective inhibitor.
Figure 2 is a diagram showing the synthesis of 1-aminopyrido[4,3-b]indole derivatives: (a) trimethyl orthoacetate, 100°C, (b) DMF-DMA, MeOH, reflux, ( c) 48% HBr, AcOH, room temperature, (d) concentrated HCl, 160°C, (e) phenyl hydrazine, Ph 2 O, 240°C, (f) POCl 3 , reflux, (g) p-meth Toxybenzylamine (p-methoxybenzylamine), 200°C, (h) 1-bromo-3-iodobenzene, Cu, K 2 CO 3 , 18-Crown-6, 1, 2-dichlorobenzene (18-crown-6, 1,2-dichlorobenzene), 180℃, (i) TFA, 60℃, (j) R-prop-2-yn-1-ol, Pd(PPh 3 ) 2 Cl 2 , TEA/DMSO (2:1), 70°C.
Figure 3 is a diagram showing the synthesis process of 4-aminopyrazolo[3,4-d]pyrimidine derivatives: (a) trimethyl orthoacetate, 100°C, (b) hydrazine monohydrate. , EtOH, 80℃, (c) formamide, 180℃, (d) 1-bromo-3-iodobenzene, CuI, K 2 CO 3 , DMEDA, DMF, 110°C, (e) R-prop-2-yn-1-ol, Pd(PPh 3 ) 2 Cl 2 , TEA/DMSO (2:1), 70°C.
Figure 4 is a diagram showing the synthesis process of 4-aminopyrazolo[3,4-d]pyrimidine derivative introduced with a solvent-accessible group: (a) trimethyl orthoacetate, 100°C, (b) DMF- DMA, MeOH, reflux, (c) 48% HBr, AcOH, room temperature, (d) concentrated HCl, 160°C, (e) phenyl hydrazine, Ph 2 O, 240°C, (f) POCl 3 , reflux. , (g) p-methoxybenzylamine, 200°C, (h) 1-bromo-3-iodobenzene, Cu, K 2 CO 3 , 18 -Crown-6, 1,2-dichlorobenzene (18-crown-6, 1,2-dichlorobenzene), 180℃, (i) TFA, 60℃, (j) R-prop-2-yn-1-ol , Pd(PPh 3 ) 2 Cl 2 , TEA/DMSO (2:1), 70°C.
Figure 5 is a graph showing enzyme IC 50 curves for SPA7012, SPA7016, and SPA7017.
Figure 6 is a schematic diagram showing the SPA7012 treatment method for C57BL/6 mice.
Figure 7 is a graph showing serum levels of AST and ALT (n=5).
Figures 8 and 9 are photographs and graphs showing the overall shape of the liver, micrographs of liver cross-sections, and necrosis area measurements (n=5).
Figure 10 is a graph showing (a) immunofluorescence staining photos and (b) quantification (n=5) results of TUNEL-positive apoptotic cells in liver tissue.
Figure 11 is a graph showing the results of (a) Western blotting and (b) analysis (n=5) of apoptosis-related proteins in liver tissue: **, p<0.01. I/R (ischemia-reperfusion), HPF (high power field).
Figure 12 is a graph showing liver levels of malondialdehyde (MDA) (n=5).
Figure 13 is a graph showing liver levels of glutathione (GSH) (n=5).
Figure 14 is a graph showing (a) a photograph of immunohistochemical staining (n=5) and (b) the results for 4-hydroxynonenal (4-HNE) in liver tissue.
Figure 15 is a diagram showing the results of co-IP analysis of HEK293T cells after 24 hours of transfection.
Figure 16 is a photograph showing Nrf2 protein levels in nuclear (NE) and cytoplasmic extracts (CE) of I/R damaged liver tissue.
Figure 17 is a graph showing luciferase activity (n=5) in HEK293T cells after ARE transfection for 24 hours.
Figures 18 and 19 show the results showing the levels of Nrf2 and target genes in liver tissue (n=5): **, p<0.01. I/R (ischemia-reperfusion), HPF (high power field), V (vehicle), S (SPA7012 50 mg/kg).
Figure 20 is a graph showing (a) immunofluorescence staining photos of F4/80-positive macrophages and Ly6G-positive neutrophils in liver tissue and (b) (c) the results of quantifying them (n=5).
Figure 21 is a graph showing protein levels of pro-inflammatory cytokines/chemokines in serum and liver tissue.
Figure 22 is a graph showing mRNA levels of pro-inflammatory cytokines/chemokines in serum and liver tissue.
Figure 23 shows the results showing the protein level of the NF-κB signaling pathway: **, p<0.01. HPF (high power field).
Figure 24 is a schematic diagram showing the SPA7012 treatment method for primary hepatocytes.
Figure 25 is a graph showing the results of MTT analysis (n=5) for cell viability.
Figure 26 is a graph showing the results of lactate dehydrogenase (LDH) release (n=5) from primary hepatocytes after 12 hours of reoxygenation.
Figure 27 shows the results of cell death analysis (n=5) by Annexin-V staining.
Figure 28 shows the results of cell death analysis (n=5) by Western blotting.
Figure 29 is a graph showing protein levels of pro-inflammatory cytokines/chemokines (n=5) in culture medium and hepatocytes: *, p<0.05 and **, p<0.01. H/R (hypoxia-reoxygenation).
Figure 30 is a graph showing mRNA levels (n=5) of pro-inflammatory cytokines/chemokines in culture medium and hepatocytes: *, p<0.05 and **, p<0.01. H/R (hypoxia-reoxygenation).

이하 본 발명을 하나 이상의 실시예를 통하여 보다 상세하게 설명한다. 그러나, 이들 실시예는 본 발명을 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail through one or more examples. However, these examples are for illustrative purposes only and the scope of the present invention is not limited to these examples.

실시예 1. PAK4 억제제의 합성 및 SPA7012의 선별Example 1. Synthesis of PAK4 inhibitors and selection of SPA7012

<1-1> 시약 및 분석 방법 <1-1> Reagents and analysis methods

대부분의 시약과 용매는 상업적 공급처에서 구입하여 추가 정제 없이 사용하였다. 모든 반응은 무수 용매를 사용하여 질소 또는 아르곤 하에서 수행되었다. 반응 완료를 모니터링하기 위해 Kieselgel 60 F254 플레이트(Merk Millipore, MA, USA)를 사용하여 박층 크로마토그래피(TLC)를 수행하였다. 플래시 컬럼 크로마토그래피는 지시된 용매와 함께 실리카겔 60(230-400 메쉬, Merck Millipore)을 사용하여 수행하였다. 중압 액체 크로마토그래피(MPLC)는 RediSep Rf 컬럼(Teledyne ISO, Hunt Valley, MD, USA)이 있는 Combiflash®Rf 200으로 수행되었다. NMR 스펙트럼은 Varian YH 400 분광기(1H 400 MHz, 13C 100 MHz, Agilent, Palo Alto, CA, USA) 및/또는 Avance III HD 500 분광기(1H 500 MHz, 13C 125 MHz, Bruker, Billerica, MA, USA)를 사용하여 기록하였다. 화학적 이동은 테트라메틸실란(tetramethylsilane)을 외부 표준(external standard)으로 사용하여 δ단위로 표시하였다. Most reagents and solvents were purchased from commercial sources and used without further purification. All reactions were performed under nitrogen or argon using anhydrous solvents. To monitor reaction completion, thin layer chromatography (TLC) was performed using Kieselgel 60 F254 plates (Merk Millipore, MA, USA). Flash column chromatography was performed using silica gel 60 (230-400 mesh, Merck Millipore) with the indicated solvents. Medium pressure liquid chromatography (MPLC) was performed with a Combiflash®Rf 200 with a RediSep Rf column (Teledyne ISO, Hunt Valley, MD, USA). NMR spectra were obtained using a Varian YH 400 spectrometer (1H 400 MHz, 13C 100 MHz, Agilent, Palo Alto, CA, USA) and/or Avance III HD 500 spectrometer (1H 500 MHz, 13C 125 MHz, Bruker, Billerica, MA, USA). It was recorded using . Chemical shifts were expressed in δ units using tetramethylsilane as an external standard.

<1-2> SPA7001-7003, SPA7011-1017의 합성<1-2> Synthesis of SPA7001-7003, SPA7011-1017

신규 PAK4 선택적 억제제를 선별하기 위하여, 구조 기반 약물 설계가 수행되었다. 즉, 선택적 억제제인 GNE-2861과 공결정화된 PAK4의 결정 구조는 비정상적인 R-spine rearrangement로 야기된 PAK4의 특정 back pocket을 대상으로 정의되었다.To screen novel PAK4 selective inhibitors, structure-based drug design was performed. In other words, the crystal structure of PAK4 co-crystallized with the selective inhibitor GNE-2861 was defined targeting the specific back pocket of PAK4 caused by abnormal R-spine rearrangement.

약동학 특성을 개선하기 위하여, 아데닌 포켓-결합 그룹을 변경하였다(도 1). 즉, 개선된 logP 값 및 추가가 용이한 용매 접근 가능 모이터디를 형성하기 위하여, 아데닌 모방 일원식을 이원식 또는 삼원식화하기 위한 스캐폴드 호핑을 수행하였다.To improve the pharmacokinetic properties, the adenine pocket-binding group was changed (Figure 1). That is, in order to form an improved logP value and a solvent-accessible moiety that is easy to add, scaffold hopping was performed to convert the adenine mimetic monomer into a binary or ternary form.

이와 같은 아데닌 포켓-결합 그룹의 변경 후, 이환식 링커는 전체 화합물의 구조적 강성을 줄이기 위해 일환식으로 변경하였다. In silico 단편 스크리닝을 통하여 헤테로사이클릭 스캐폴드에 구조적 신규성을 제공하였다. 2개의 구조, 즉 피리도[4,3-b]인돌(pyrido[4,3-b]indole, γ카르볼린) 및 피라졸로[3,4-d]피리미딘(pyrazolo[3,4-d]pyrimidine)이 I형 1/2 억제제로 선택 및 설계되었다. 그 후, 이들의 특성을 개선하기 위해 용매에 접근할 수 있는 부분이 도입되었다. After changing the adenine pocket-binding group, the bicyclic linker was changed to a monocyclic linker to reduce the structural rigidity of the entire compound. In silico fragment screening provided structural novelty to the heterocyclic scaffold. Two structures, pyrido[4,3-b]indole (γcarboline) and pyrazolo[3,4-d]pyrimidine (pyrazolo[3,4-d) ]pyrimidine) was selected and designed as a type I 1/2 inhibitor. Afterwards, solvent-accessible parts were introduced to improve their properties.

즉, PAK4 억제제의 합성을 위하여, 말로노니트릴(malononitrile) 및 트리메틸 오르토아세테이트(trimethyl orthoacetate)로부터 시작하여 엔올에터(enol ether)를 합성하고 엔아민을 통해 아미노피라졸로(aminopyrazol) 변형시킨 다음 이를 고리화하여 4-아미노피라졸로[3,4-d]피리미딘(4-aminopyrazolo[3,4-d]pyrimidine) 유도체를 합성하였다. 그 후, 울만 타입 반응 및 소노가시라 커플링을 통하여 최종적으로 PAK4 억제제 화합물 SPA7001 내지 SPA7003 및 SPA7011 내지 SPA1017을 수득하였다(도 2 내지 도 4).That is, for the synthesis of a PAK4 inhibitor, starting from malononitrile and trimethyl orthoacetate, enol ether is synthesized and transformed into aminopyrazol through enamine. By cyclization, a 4-aminopyrazolo[3,4-d]pyrimidine derivative was synthesized. Thereafter, PAK4 inhibitor compounds SPA7001 to SPA7003 and SPA7011 to SPA1017 were finally obtained through Ullman-type reaction and Sonogashira coupling (FIGS. 2 to 4).

[반응식 1][Scheme 1]

Scheme 1. Synthesis of 1-aminopyrido[4,3-b]indole derivatives: Reagents and conditions; (a) trimethyl orthoacetate, 100℃; (b) DMF-DMA, MeOH, reflux; (c) 48% HBr, AcOH, rt; (d) conc. HCl, 160℃; (e) phenyl hydrazine, Ph2O, 240℃; (f) POCl3, reflux; (g) p-methoxybenzylamine, 200℃; (h) 1-bromo-3-iodobenzene, Cu, K2CO3, 18-crown-6, 1,2-dichlorobenzene, 180℃; (i) TFA, 60℃; (j) R-prop-2-yn-1-ol, Pd(PPh3)2Cl2, TEA/DMSO (2:1), 70℃.Scheme 1. Synthesis of 1-aminopyrido[4,3-b]indole derivatives: Reagents and conditions; (a) trimethyl orthoacetate, 100℃; (b) DMF-DMA, MeOH, reflux; (c) 48% HBr, AcOH, rt; (d) conc. HCl, 160°C; (e) phenyl hydrazine, Ph2O, 240℃; (f) POCl3, reflux; (g) p-methoxybenzylamine, 200℃; (h) 1-bromo-3-iodobenzene, Cu, K2CO3, 18-crown-6, 1,2-dichlorobenzene, 180°C; (i) TFA, 60°C; (j) R-prop-2-yn-1-ol, Pd(PPh3)2Cl2, TEA/DMSO (2:1), 70°C.

[반응식 2][Scheme 2]

Scheme 2. Synthesis of the 4-aminopyrazolo[3,4-d]pyrimidine derivatives: Reagents and conditions; (a) trimethyl orthoacetate, 100℃; (b) hydrazine monohydrate, EtOH, 80℃; (c) formamide, 180℃; (d) 1-bromo-3-iodobenzene, CuI, K2CO3, DMEDA, DMF, 110℃; (e) R-prop-2-yn-1-ol, Pd(PPh3)2Cl2, TEA/DMSO (2:1), 70℃.Scheme 2. Synthesis of the 4-aminopyrazolo[3,4-d]pyrimidine derivatives: Reagents and conditions; (a) trimethyl orthoacetate, 100℃; (b) hydrazine monohydrate, EtOH, 80°C; (c) formamide, 180℃; (d) 1-bromo-3-iodobenzene, CuI, K2CO3, DMEDA, DMF, 110°C; (e) R-prop-2-yn-1-ol, Pd(PPh3)2Cl2, TEA/DMSO (2:1), 70°C.

[반응식 3][Scheme 3]

Scheme 3. Synthesis of the 4-aminopyrazolo[3,4-d]pyrimidine derivatives with a solvent-accessible moiety: Reagents and conditions; (a) CuI, Pd(PPh3)4, TEA, DMF, 70℃; (b) Pd/C, H2, MeOH, rt; (c) 1-bromo-3-iodobenzene, CuI, K2CO3, DMEDA, DMF, 110℃; (d) 1-Ethynylcyclohexanol, Pd(PPh3)2Cl2, TEA/DMSO (2:1), 70℃.Scheme 3. Synthesis of the 4-aminopyrazolo[3,4-d]pyrimidine derivatives with a solvent-accessible moiety: Reagents and conditions; (a) CuI, Pd(PPh 3 ) 4 , TEA, DMF, 70°C; (b) Pd/C, H 2 , MeOH, rt; (c) 1-bromo-3-iodobenzene, CuI, K 2 CO 3 , DMEDA, DMF, 110°C; (d) 1-Ethynylcyclohexanol, Pd(PPh 3 ) 2 Cl 2 , TEA/DMSO (2:1), 70°C.

화합물compound SPA No.SPA No. 구조식
[화합물명]
constitutional formula
[Compound name]
화합물 1Compound 1 SPA7001SPA7001
[4-(3-(1-Aminopyrido[4,3-b]indol-5-yl)phenyl)-2-methylbutyn-2-ol]

[4-(3-(1-Aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyn-2-ol]
화합물 2compound 2 SPA7002SPA7002
[3-(1-Aminopyrido[4,3-b]indol-5-yl)phenylethynylcyclohexan-1-ol]

[3-(1-Aminopyrido[4,3- b ]indol-5-yl)phenylethynylcyclohexan-1-ol]
화합물 3Compound 3 SPA7003SPA7003
[4-(3-(1-Aminopyrido[4,3-b]indol-5-yl)phenyl)-2-methylbutyn-1,2-diol]

[4-(3-(1-Aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyn-1,2-diol]

화합물
compound
SPA No.SPA No. 구조식
[화합물명]
constitutional formula
[Compound name]
화합물 4Compound 4 SPA7011SPA7011
[4-(3-(4-Amino-3-methylpyrazolo[3,4-d]pyrimidin-1-yl)phenyl)-2-methylbutyn-2-ol]

[4-(3-(4-Amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutyn-2-ol]
화합물 5Compound 5 SPA7012SPA7012
[3-(4-Amino-3-methylpyrazolo[3,4-d]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol]

[3-(4-Amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol]
화합물 6Compound 6 SPA7013SPA7013
[4-(3-(4-Amino-3-methylpyrazolo[3,4-d]pyrimidin-1-yl)phenyl)-2-methylbutyn-1,2-diol]

[4-(3-(4-Amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutyn-1,2-diol]
화합물 7Compound 7 SPA7014SPA7014
[4-(3-(4-Amino-3-methylpyrazolo[3,4-d]pyrimidin-1-yl)phenyl)-2-phenylbutyn-2-ol]

[4-(3-(4-Amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-phenylbutyn-2-ol]
화합물 8Compound 8 SPA7016SPA7016
[3-[4-Amino-3-(3-morpholin-1-ylpropyl)pyrazolo[3,4-d]pyrimidin-1-yl]phenylethynyl)cyclohexanol]

[3-[4-Amino-3-(3-morpholin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol]
화합물 9Compound 9 SPA7017SPA7017
[3-[4-Amino-3-(3-piperidin-1-ylpropyl)pyrazolo[3,4-d]pyrimidin-1-yl]phenylethynyl)cyclohexanol]

[3-[4-Amino-3-(3-piperidin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol]

<화합물 1-18의 합성><Synthesis of Compound 1-18>

2-(1-Methoxyethylidene)malononitrile (1)2-(1-Methoxyethylidene)malononitrile (1)

말로노니트릴(5.0ml, 78.60mmol) 및 트리메틸 오르토아세테이트(11.0ml, 86.46mmol)의 혼합물을 4시간 동안 환류 가열하였다. 실온으로 냉각시킨 후, 반응 혼합물을 n-헥산 중 0-30% EtOAc의 구배로 MPLC로 정제하여 2를 밝은 황색 액체로서 제공하였다(9.51 g, 99 %): R f = 0.30 (n-hexane:EtOAc = 3:2); 1H NMR (400 MHz, CDCl3) δ 4.08 (s, 3H), 2.41 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 185.7, 113.2, 111.1, 66.3, 58.6, 17.4.A mixture of malononitrile (5.0 ml, 78.60 mmol) and trimethyl orthoacetate (11.0 ml, 86.46 mmol) was heated to reflux for 4 hours. After cooling to room temperature, the reaction mixture was purified by MPLC with a gradient of 0-30% EtOAc in n-hexane to give 2 as a light yellow liquid (9.51 g, 99%): R f = 0.30 ( n -hexane: EtOAc = 3:2); 1 H NMR (400 MHz, CDCl 3 ) δ 4.08 (s, 3H), 2.41 (s, 3H); 13 C NMR (100 MHz, CDCl 3 ) δ 185.7, 113.2, 111.1, 66.3, 58.6, 17.4.

2-(3-Dimethylamino-1-methoxyallylidene)malononitrile (2)2-(3-Dimethylamino-1-methoxyallylidene)malononitrile (2)

MeOH(120ml) 중의 1(9.51g, 77.86mmol)의 혼합물에 DMF-DMA(11.7ml, 85.65mmol)를 첨가하였다. 반응 혼합물을 1시간 동안 환류 가열하였다. 실온으로 냉각한 후, 반응 혼합물을 농축하고 물로 희석하였다. 생성된 혼합물을 DCM으로 추출하였다. 합한 유기층을 염수로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 재결정화하고(EtOH 100%) 플래시 컬럼 크로마토그래피로 정제하여 3(14.60g, 67%)을 갈색 고체로서 제공하였다: R f = 0.19 (n-hexane:EtOAc = 1:1); 1H NMR (400 MHz, CDCl3) δ 7.55 (d, J = 12.3 Hz, 1H), 5.09 (d, J = 12.3 Hz, 1H), 4.10 (s, 3H), 3.21 (s, 3H), 2.95 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 181.7, 152.8, 117.4, 116.1, 87.4, 60.7, 45.8, 37.4.To a mixture of 1 (9.51 g, 77.86 mmol) in MeOH (120 ml) was added DMF-DMA (11.7 ml, 85.65 mmol). The reaction mixture was heated to reflux for 1 hour. After cooling to room temperature, the reaction mixture was concentrated and diluted with water. The resulting mixture was extracted with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The residue was recrystallized (EtOH 100%) and purified by flash column chromatography to give 3 (14.60 g, 67%) as a brown solid: R f = 0.19 ( n -hexane:EtOAc = 1:1); 1H NMR (400 MHz, CDCl 3 ) δ 7.55 (d, J = 12.3 Hz, 1H), 5.09 (d, J = 12.3 Hz, 1H), 4.10 (s, 3H), 3.21 (s, 3H), 2.95 (s, 3H); 13 C NMR (100 MHz, CDCl 3 ) δ 181.7, 152.8, 117.4, 116.1, 87.4, 60.7, 45.8, 37.4.

2-Bromo-4-methoxynicotinonitrile (3)2-Bromo-4-methoxynicotinonitrile (3)

48% HBr(26ml) 중의 2(2.30g, 12.96mmol)의 혼합물에 아세트산 빙수(13ml)를 첨가하였다. 실온에서 14시간 동안 교반한 후, 반응 혼합물을 NaHCO3(수성)로 중화하고 EtOAc로 추출하였다. 합한 유기층을 염수로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 n-헥산 중 0-40% EtOAc의 구배로 MPLC로 정제하여 3(1.81 g, 66%)을 황색 고체로서 제공하였다: R f = 0.30 (n-hexane:EtOAc = 1:1); 1H NMR (400 MHz, CDCl3) δ 8.40 (d, J = 5.9 Hz, 1H), 6.92 (d, J = 5.9 Hz, 1H), 4.03 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 168.7, 153.5, 145.3, 113.3, 106.1, 103.8, 57.0.To a mixture of 2 (2.30 g, 12.96 mmol) in 48% HBr (26 ml) was added acetic acid ice water (13 ml). After stirring at room temperature for 14 hours, the reaction mixture was neutralized with NaHCO3 (aq) and extracted with EtOAc. The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The residue was purified by MPLC with a gradient of 0-40% EtOAc in n-hexane to give 3 (1.81 g, 66%) as a yellow solid: R f = 0.30 ( n -hexane:EtOAc = 1:1); 1 H NMR (400 MHz, CDCl 3 ) δ 8.40 (d, J = 5.9 Hz, 1H), 6.92 (d, J = 5.9 Hz, 1H), 4.03 (s, 3H); 13 C NMR (100 MHz, CDCl 3 ) δ 168.7, 153.5, 145.3, 113.3, 106.1, 103.8, 57.0.

4-Hydroxy-14-Hydroxy-1 HH -pyridin-2-one (4)-pyridin-2-one (4)

반응관에 농축액을 채웠다. HCl(6ml) 용액에 이어서 3(1.38g, 6.48mmol)을 첨가하였다. 튜브를 밀봉하고 혼합물을 160℃에서 15시간 동안 가열하였다. 실온으로 냉각한 후, 반응 혼합물을 물로 희석하고 2 N NaOH(aq)로 중화시켰다. 생성된 혼합물을 감압 건조하고 MeOH에 용해시켰다. 생성된 현탁액을 여과하고, 여액을 CHCl3 중 0-20% MeOH의 구배로 MPLC로 정제하여 4(676 mg, 94%)를 백색 고체로서 제공하였다: R f = 0.22 (CHCl3:MeOH = 8:1); 1H NMR (400 MHz, DMSO-d 6) δ 11.07 (s, 1H), 7.26 (d, J = 7.2 Hz, 1H), 5.88 (d, J = 7.2 Hz, 1H), 5.60 (s, 1H), 3.12 (s, 1H); 13C NMR (100 MHz, DMSO-d 6 ) δ 168.5, 164.9, 136.2, 100.5, 99.1.The reaction tube was filled with the concentrate. To the HCl (6ml) solution was added 3 (1.38g, 6.48mmol). The tube was sealed and the mixture was heated at 160°C for 15 hours. After cooling to room temperature, the reaction mixture was diluted with water and neutralized with 2 N NaOH (aq). The resulting mixture was dried under reduced pressure and dissolved in MeOH. The resulting suspension was filtered and the filtrate was purified by MPLC with a gradient of 0-20% MeOH in CHCl 3 to give 4 (676 mg, 94%) as a white solid: R f = 0.22 (CHCl 3 :MeOH = 8 :One); 1H NMR (400 MHz, DMSO- d6 ) δ 11.07 (s, 1H), 7.26 (d, J = 7.2 Hz, 1H), 5.88 (d, J = 7.2 Hz, 1H), 5.60 (s, 1H) , 3.12 (s, 1H); 13 C NMR (100 MHz, DMSO- d 6 ) δ 168.5, 164.9, 136.2, 100.5, 99.1.

2,5-Dihydropyrido[4,3-2,5-Dihydropyrid[4,3- bb ]indolone (5)]indolone (5)

디페닐에터(28ml) 중 4(312mg, 2.81mmol)의 혼합물에 페닐히드라진(0.34ml, 3.37mmol)을 첨가하였다. 반응 혼합물을 예열된 모래 욕에서 환류 가열하고 18시간 동안 교반하였다. 실온으로 냉각시킨 후, 반응 혼합물을 n-헥산(300ml)에 부었다. 침전물을 여과하고 THF로 헹구었다. 합한 여액을 플래시 컬럼 크로마토그래피로 정제하여 5(101mg, 20%)를 갈색 고체로서 제공하였다: R f = 0.33 (CHCl3:MeOH = 8:1); 1H NMR (400 MHz, CD3OD) δ 8.22 (d, J = 7.8 Hz, 1H), 7.47 (d, J = 8.1 Hz, 1H), 7.36 (d, J = 7.1 Hz), 7.34 (t, J = 8.1 Hz, 1H), 7.24 (t, J = 7.8 Hz, 1H), 6.68 (d, J = 7.1 Hz, 1H).To a mixture of 4 (312 mg, 2.81 mmol) in diphenyl ether (28 ml) was added phenylhydrazine (0.34 ml, 3.37 mmol). The reaction mixture was heated to reflux in a preheated sand bath and stirred for 18 hours. After cooling to room temperature, the reaction mixture was poured into n-hexane (300 ml). The precipitate was filtered and rinsed with THF. The combined filtrates were purified by flash column chromatography to provide 5 (101 mg, 20%) as a brown solid: R f = 0.33 (CHCl 3 :MeOH = 8:1); 1 H NMR (400 MHz, CD 3 OD) δ 8.22 (d, J = 7.8 Hz, 1H), 7.47 (d, J = 8.1 Hz, 1H), 7.36 (d, J = 7.1 Hz), 7.34 (t, J = 8.1 Hz, 1H), 7.24 (t, J = 7.8 Hz, 1H), 6.68 (d, J = 7.1 Hz, 1H).

1-Chloro-51-Chloro-5 HH -pyrido[4,3--pyrido[4,3- bb ]indole (6)]indole (6)

5(648 mg, 3.52 mmol) 및 옥시염화인(10 ml)의 혼합물을 14시간 동안 가열 환류하였다. 실온으로 냉각시킨 후, 반응 혼합물을 농축하고, NaHCO3(수성)로 중화하고, DCM으로 추출하였다. 합한 유기층을 염수로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피로 정제하여 6 (376 mg, 53%)을 옅은 노란색 고체로 제공하였다: R f = 0.51 (CHCl3:MeOH = 10:1); 1H NMR (500 MHz, DMSO-d 6) δ 12.15 (s, 1H), 8.38 (d, J = 7.9 Hz, 1H), 8.24 (d, J = 5.6 Hz, 1H), 7.65 (d, J = 7.5 Hz, 1H), 7.57 (t, J = 7.5 Hz, 1H), 7.54 (d, J = 5.6 Hz, 1H), 7.37 (t, J = 7.9 Hz, 1H); 13C NMR (125 MHz, DMSO-d 6 ) δ 145.9, 144.2, 144.1, 140.1, 127.7, 122.4, 121.1, 120.0, 116.7, 112.3, 107.0.A mixture of 5 (648 mg, 3.52 mmol) and phosphorus oxychloride (10 ml) was heated to reflux for 14 hours. After cooling to room temperature, the reaction mixture was concentrated, neutralized with NaHCO3 (aq) and extracted with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography to give 6 (376 mg, 53%) as a pale yellow solid: R f = 0.51 (CHCl 3 :MeOH = 10:1); 1H NMR (500 MHz, DMSO- d6 ) δ 12.15 (s, 1H), 8.38 ( d , J = 7.9 Hz, 1H), 8.24 (d, J = 5.6 Hz, 1H), 7.65 (d, J = 7.5 Hz, 1H), 7.57 (t, J = 7.5 Hz, 1H), 7.54 (d, J = 5.6 Hz, 1H), 7.37 (t, J = 7.9 Hz, 1H); 13 C NMR (125 MHz, DMSO- d 6 ) δ 145.9, 144.2, 144.1, 140.1, 127.7, 122.4, 121.1, 120.0, 116.7, 112.3, 107.0.

4-Methoxybenzyl(54-Methoxybenzyl(5 HH -pyrido[4,3--pyrido[4,3- bb ]indol-1-yl)amine (7)]indol-1-yl)amine (7)

반응 튜브에 6(407mg, 2.01mmol) 및 4-메톡시벤질아민(5.3ml, 40.12mmol)을 채웠다. 튜브를 밀봉하고, 혼합물을 200℃에서 4시간 동안 가열하였다. 실온으로 냉각시킨 후, 반응 혼합물을 물로 희석하고 EtOAc로 추출하였다. 합한 유기층을 염수로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피로 정제하여 7(760 mg, 정량적)을 옅은 노란색 고체로 제공하였다: R f = 0.32 (n-hexane:EtOAc = 1:1); 1H NMR (400 MHz, CDCl3) δ 8.76 (s, 1H), 8.12 (dd, J = 5.6, 2.0 Hz, 1H), 7.76 (d, J = 8.0 Hz, 1H) 7.49-7.33 (m, 3H), 7.30-7.20 (m, 2H), 6.96-6.84 (m, 2H), 6.78 (dd, J = 5.6, 2.0 Hz, 1H), 5.13 (s, 1H), 4.87(d, J = 3.2 Hz, 2H), 3.80 (s, 3H).6 (407 mg, 2.01 mmol) and 4-methoxybenzylamine (5.3 ml, 40.12 mmol) were charged into the reaction tube. The tube was sealed and the mixture was heated at 200° C. for 4 hours. After cooling to room temperature, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography to give 7 (760 mg, quantitative) as a pale yellow solid: R f = 0.32 ( n -hexane:EtOAc = 1:1); 1H NMR (400 MHz, CDCl 3 ) δ 8.76 (s, 1H), 8.12 (dd, J = 5.6, 2.0 Hz, 1H), 7.76 (d, J = 8.0 Hz, 1H) 7.49-7.33 (m, 3H) ), 7.30-7.20 (m, 2H), 6.96-6.84 (m, 2H), 6.78 (dd, J = 5.6, 2.0 Hz, 1H), 5.13 (s, 1H), 4.87(d, J = 3.2 Hz, 2H), 3.80 (s, 3H).

(5-(3-Bromophenyl)-5(5-(3-Bromophenyl)-5 HH -pyrido[4,3--pyrido[4,3- bb ]indolyl)(4-methoxybenzyl)amine (8)]indolyl)(4-methoxybenzyl)amine (8)

7(674mg, 2.22mmol), 구리 분말(283mg, 4.45mmol), 탄산칼륨(922 mg, 6.67 mmol), 18-크라운-6(178mg, 0.67mmol)의 혼합물에 1,2- 디클로로벤젠(10ml)을 3-브로모-1-요오도벤젠(434㎕, 3.33mmol)에 첨가하였다. 반응 혼합물을 180℃에서 5시간 동안 가열하였다. 뜨거운 반응 혼합물을 셀라이트 패드를 통해 여과하고 뜨거운 EtOAc로 헹구었다. 합한 여액을 염수로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피로 정제하여 8(436mg, 43%)을 황색 고체로서 제공하였다: R f = 0.63 (n-hexane:EtOAc = 2:1); 1H NMR (400 MHz, CDCl3) δ 8.12 (s, 1H), 7.81 (s, 1H) 7.72-7.58 (m, 2H), 7.52-7.27 (m, 7H), 6.92 (d, J = 6.8 Hz, 2H), 6.70 (s, 1H), 5.19 (s, 1H), 4.87(s, 2H), 3.81 (s, 3H).1,2-dichlorobenzene (10 ml) in a mixture of 7 (674 mg, 2.22 mmol), copper powder (283 mg, 4.45 mmol), potassium carbonate (922 mg, 6.67 mmol), and 18-crown-6 (178 mg, 0.67 mmol). was added to 3-bromo-1-iodobenzene (434㎕, 3.33mmol). The reaction mixture was heated at 180°C for 5 hours. The hot reaction mixture was filtered through a pad of Celite and rinsed with hot EtOAc. The combined filtrates were washed with brine, dried over MgSO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography to give 8 (436 mg, 43%) as a yellow solid: R f = 0.63 ( n -hexane:EtOAc = 2:1); 1H NMR (400 MHz, CDCl 3 ) δ 8.12 (s, 1H), 7.81 (s, 1H) 7.72-7.58 (m, 2H), 7.52-7.27 (m, 7H), 6.92 (d, J = 6.8 Hz , 2H), 6.70 (s, 1H), 5.19 (s, 1H), 4.87(s, 2H), 3.81 (s, 3H).

5-(3-Bromophenyl)-55-(3-Bromophenyl)-5 HH -pyrido[4,3--pyrido[4,3- bb ]indolylamine (9)]indolylamine (9)

8(391mg, 0.85mmol) 및 트리플루오로아세트산(3.3ml)의 혼합물을 30분 동안 60℃로 가열하였다. 실온으로 냉각한 후, 반응 혼합물을 얼음물에 붓고, NaHCO3(수성)로 중화하고, EtOAc로 추출하였다. 합한 유기층을 염수로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피로 정제하여 9(336mg, 정량적)를 옅은 노란색 고체로 제공하였다: R f = 0.11 (n-hexane:EtOAc = 1:1); 1H NMR (400 MHz, CDCl3) δ 8.00 (d, J = 5.6 Hz, 1H), 7.92 (d, J = 7.2 Hz, 1H), 7.67 (s, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.53-7.28 (m, 5H), 6.72 (t, J = 5.2 Hz, 1H), 5.47 (s, 2H).A mixture of 8 (391 mg, 0.85 mmol) and trifluoroacetic acid (3.3 ml) was heated to 60° C. for 30 min. After cooling to room temperature, the reaction mixture was poured into ice-water, neutralized with NaHCO 3 (aq.) and extracted with EtOAc. The combined organic layers were washed with brine, dried over MgSO 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography to give 9 (336 mg, quantitative) as a pale yellow solid: R f = 0.11 ( n -hexane:EtOAc = 1:1); 1H NMR (400 MHz, CDCl 3 ) δ 8.00 (d, J = 5.6 Hz, 1H), 7.92 (d, J = 7.2 Hz, 1H), 7.67 (s, 1H), 7.63 (d, J = 7.2 Hz) , 1H), 7.53-7.28 (m, 5H), 6.72 (t, J = 5.2 Hz, 1H), 5.47 (s, 2H).

화합물 10a-c, 14a-d 및 18a-b의 합성을 위한 일반 절차General procedure for synthesis of compounds 10a-c, 14a-d and 18a-b

TEA/DMSO(2:1) 중 화합물 9, 13 또는 17(1.0 당량)의 혼합물에 질소 분위기 하에 비스(트리페닐포스핀)팔라듐(II) 디클로라이드(1.0 당량)를 첨가하였다. 실온에서 10분 동안 교반한 후, 알킬 아세틸렌(10.0 당량)을 첨가하였다. 반응 혼합물을 70℃에서 1시간 동안 가열하였다. 실온으로 냉각한 후, 혼합물을 셀라이트 패드를 통해 여과하고 EtOAc로 헹구었다. 물을 여액에 첨가하고 혼합물을 EtOAc로 추출하였다. 합한 유기층을 MgSO4상에서 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피(CHCl3:EtOAc 또는 EtOAc:MeOH)로 정제하여 10, 14 또는 18을 제공하였다.To a mixture of compounds 9, 13 or 17 (1.0 equiv) in TEA/DMSO (2:1) was added bis(triphenylphosphine)palladium(II) dichloride (1.0 equiv) under nitrogen atmosphere. After stirring at room temperature for 10 minutes, alkyl acetylene (10.0 equiv) was added. The reaction mixture was heated at 70° C. for 1 hour. After cooling to room temperature, the mixture was filtered through a pad of Celite and rinsed with EtOAc. Water was added to the filtrate and the mixture was extracted with EtOAc. The combined organic layers were dried over MgSO 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography (CHCl3:EtOAc or EtOAc:MeOH) to give 10, 14 or 18.

4-(3-(1-Aminopyrido[4,3-4-(3-(1-Aminopyrido[4,3- bb ]indol-5-yl)phenyl)-2-methylbutyn-2-ol (10a)]indol-5-yl)phenyl)-2-methylbutyn-2-ol (10a)

미색 고체(71%): R f = 0.31 (EtOAc 100%); 1H NMR (400 MHz, CDCl3) δ 7.97 (d, J = 5.8 Hz, 1H), 7.87 (d, J = 7.9 Hz, 1H), 7.54-7.48 (m, 3H), 7.40-7.31 (m, 4H), 6.67 (d, J = 5.8 Hz, 1H), 5.35 (s, 2H), 3.56 (s, 1H), 1.65 (s, 6H); 13C NMR (100 MHz, CDCl3) δ 153.9, 146.4, 143.5, 139.8, 136.6, 131.3, 130.0, 129.9, 126.8, 125.2, 125.0, 121.8, 121.4, 120.3, 110.0, 104.2, 97.8, 96.0, 80.6, 65.2, 31.4.Off-white solid (71%): R f = 0.31 (EtOAc 100%); 1H NMR (400 MHz, CDCl 3 ) δ 7.97 (d, J = 5.8 Hz, 1H), 7.87 (d, J = 7.9 Hz, 1H), 7.54-7.48 (m, 3H), 7.40-7.31 (m, 4H), 6.67 (d, J = 5.8 Hz, 1H), 5.35 (s, 2H), 3.56 (s, 1H), 1.65 (s, 6H); 13 C NMR (100 MHz, CDCl 3 ) δ 153.9, 146.4, 143.5, 139.8, 136.6, 131.3, 130.0, 129.9, 126.8, 125.2, 125.0, 121.8, 121.4, 120.3, 110 .0, 104.2, 97.8, 96.0, 80.6, 65.2 , 31.4.

3-(1-Aminopyrido[4,3-3-(1-Aminopyrido[4,3- bb ]indol-5-yl)phenylethynylcyclohexan-1-ol (10b) ]indol-5-yl)phenylethynylcyclohexan-1-ol (10b)

황색 고체 (49%): R f = 0.38 (EtOAc 100%); 1H NMR (500 MHz, CDCl3) δ 7.99 (d, J = 5.9 Hz, 1H), 7.90 (d, J = 7.6 Hz, 1H), 7.60-7.51 (m, 3H), 7.46-7.33 (m, 4H), 6.71 (d, J = 5.9 Hz, 1H), 5.42 (s, 2H), 4.01 (s, 1H), 2.07-1.99 (m, 2H), 1.80-1.53 (m, 7H), 1.35-1.24 (m, 1H); 13C NMR (125 MHz, CDCl3) δ 153.9, 146.5, 143.3, 139.9, 136.7, 131.5, 130.1, 130.0, 127.0, 125.3, 125.2, 121.8, 121.6, 120.4, 110.1, 104.3, 98.0, 94.9, 82.9, 68.9, 40.0, 25.2, 23.4.Yellow solid (49%): R f = 0.38 (EtOAc 100%); 1H NMR (500 MHz, CDCl 3 ) δ 7.99 (d, J = 5.9 Hz, 1H), 7.90 (d, J = 7.6 Hz, 1H), 7.60-7.51 (m, 3H), 7.46-7.33 (m, 4H), 6.71 (d, J = 5.9 Hz, 1H), 5.42 (s, 2H), 4.01 (s, 1H), 2.07-1.99 (m, 2H), 1.80-1.53 (m, 7H), 1.35-1.24 (m, 1H); 13 C NMR (125 MHz, CDCl 3 ) δ 153.9, 146.5, 143.3, 139.9, 136.7, 131.5, 130.1, 130.0, 127.0, 125.3, 125.2, 121.8, 121.6, 120.4, 110 .1, 104.3, 98.0, 94.9, 82.9, 68.9 , 40.0, 25.2, 23.4.

4-(3-(1-Aminopyrido[4,3-4-(3-(1-Aminopyrido[4,3- bb ]indol-5-yl)phenyl)-2-methylbutyn-1,2-diol (10c)]indol-5-yl)phenyl)-2-methylbutyn-1,2-diol (10c)

미색 고체 (10%): R f = 0.16 (EtOAc 100%); 1H NMR (500 MHz, CD3OD) δ 8.24 (d, J = 7.5 Hz, 1H), 7.88 (d, J = 6.1 Hz, 1H), 7.70-7.63 (m, 3H), 7.55 (dt, J = 7.0, 2.1 Hz, 1H), 7.49-7.36 (m, 3H), 6.73 (d, J = 6.1 Hz, 1H), 3.61 (s, 2H); 13C NMR (125 MHz, CD3OD) δ 154.1, 146.5, 141.9, 139.8, 136.6, 131.3, 130.1, 129.8, 126.9, 125.2, 125.1, 121.8, 121.5, 120.6, 109.5, 103.8, 97.0, 93.3, 81.7, 69.6, 68.2, 24.7.Off-white solid (10%): R f = 0.16 (EtOAc 100%); 1H NMR (500 MHz, CD 3 OD) δ 8.24 (d, J = 7.5 Hz, 1H), 7.88 (d, J = 6.1 Hz, 1H), 7.70-7.63 (m, 3H), 7.55 (dt, J = 7.0, 2.1 Hz, 1H), 7.49-7.36 (m, 3H), 6.73 (d, J = 6.1 Hz, 1H), 3.61 (s, 2H); 13 C NMR (125 MHz, CD 3 OD) δ 154.1, 146.5, 141.9, 139.8, 136.6, 131.3, 130.1, 129.8, 126.9, 125.2, 125.1, 121.8, 121.5, 120.6, 10 9.5, 103.8, 97.0, 93.3, 81.7, 69.6, 68.2, 24.7.

5-Amino-3-methyl-15-Amino-3-methyl-1 HH -pyrazole-4-carbonitrile (11)-pyrazole-4-carbonitrile (11)

EtOH(25ml) 중의 1(1.64g, 13.42mmol)의 용액에 히드라진 일수화물(1.3ml, 26.84mmol)을 첨가하였다. 반응 혼합물을 80℃에서 1시간 동안 가열하였다. 실온으로 냉각시킨 후, 반응 혼합물을 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피로 정제하여 11(1.60 g, 98%)을 회백색 고체로서 제공하였다: R f = 0.31 (n-hexane:EtOAc = 1:2); 1H NMR (500 MHz, CD3OD) δ 2.25 (s, 3H).To a solution of 1 (1.64 g, 13.42 mmol) in EtOH (25 ml) was added hydrazine monohydrate (1.3 ml, 26.84 mmol). The reaction mixture was heated at 80° C. for 1 hour. After cooling to room temperature, the reaction mixture was concentrated. The residue was purified by flash column chromatography to provide 11 (1.60 g, 98%) as an off-white solid: R f = 0.31 ( n -hexane:EtOAc = 1:2); 1 H NMR (500 MHz, CD 3 OD) δ 2.25 (s, 3H).

3-Methyl-13-Methyl-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (12)]pyrimidin-4-ylamine (12)

11(1.37g, 11.22mmol) 및 포름아미드(9ml)의 혼합물을 18시간 동안 180℃로 가열하였다. 실온으로 냉각시킨 후, 반응 혼합물을 물에 붓고 빙욕으로 냉각시켰다. 고체를 여과하고 물 및 MeOH로 헹구어 12(1.10g, 65%)를 추가 정제 없이 황색 고체로서 제공하였다: R f = 0.26 (EtOAc:MeOH = 10:1); 1H NMR (500 MHz, CD3OD) δ 8.15 (s, 1H), 2.62 (s, 3H); 13C NMR (125 MHz, CD3OD) δ 164.7, 159.0, 155.5, 152.0, 98.6, 12.9.A mixture of 11 (1.37 g, 11.22 mmol) and formamide (9 ml) was heated to 180° C. for 18 hours. After cooling to room temperature, the reaction mixture was poured into water and cooled in an ice bath. The solid was filtered and rinsed with water and MeOH to give 12 (1.10 g, 65%) as a yellow solid without further purification: R f = 0.26 (EtOAc:MeOH = 10:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.15 (s, 1H), 2.62 (s, 3H); 13 C NMR (125 MHz, CD 3 OD) δ 164.7, 159.0, 155.5, 152.0, 98.6, 12.9.

화합물 13 및 17a-b의 합성을 위한 일반 절차General procedure for synthesis of compounds 13 and 17a-b

DMF 중 12 또는 16(1.0당량), 요오드화구리(0.2당량), 탄산칼륨(2.0당량)의 혼합물에 DMEDA(0.4당량) 및 3-브로모-1-요오도벤젠( 1.2 등가)을 첨가하고, 반응 혼합물을 110℃에서 3-23시간 동안 가열하였다. 실온으로 냉각한 후, 반응 혼합물을 셀라이트 패드를 통해 여과하고 EtOAc로 헹구었다. 합한 여액을 염수로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피(EtOAc:MeOH)로 정제하여 13 또는 17을 제공하였다.To a mixture of 12 or 16 (1.0 equivalents), copper iodide (0.2 equivalents), and potassium carbonate (2.0 equivalents) in DMF, add DMEDA (0.4 equivalents) and 3-bromo-1-iodobenzene (1.2 equivalents); The reaction mixture was heated at 110°C for 3-23 hours. After cooling to room temperature, the reaction mixture was filtered through a pad of Celite and rinsed with EtOAc. The combined filtrates were washed with brine, dried over MgSO 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography (EtOAc:MeOH) to provide 13 or 17.

(3-Bromophenyl)-3-methyl-1(3-Bromophenyl)-3-methyl-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (13)]pyrimidin-4-ylamine (13)

미색 고체 (25%): R f = 0.67 (EtOAc:MeOH = 10:1); 1H NMR (500 MHz, CD3OD) δ 8.40 (s, 1H), 8.28 (s, 1H), 8.18 (d, J = 8.0 Hz, 1H), 7.49-7.40 (m, 2H), 2.69 (s, 3H); 13C NMR (125 MHz, CD3OD) δ 158.9, 156.2, 154.4, 143.8, 142.2, 140.0, 130.2, 128.6, 123.5, 122.0, 119.4, 13.1.Off-white solid (25%): R f = 0.67 (EtOAc:MeOH = 10:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.40 (s, 1H), 8.28 (s, 1H), 8.18 (d, J = 8.0 Hz, 1H), 7.49-7.40 (m, 2H), 2.69 (s) , 3H); 13 C NMR (125 MHz, CD 3 OD) δ 158.9, 156.2, 154.4, 143.8, 142.2, 140.0, 130.2, 128.6, 123.5, 122.0, 119.4, 13.1.

4-(3-(4-Amino-3-methylpyrazolo[3,4-4-(3-(4-Amino-3-methylpyrazolo[3,4- dd ]pyrimidin-1-yl)phenyl)-2-methylbutyn-2-ol (14a).]pyrimidin-1-yl)phenyl)-2-methylbutyn-2-ol (14a).

옅은 황색 고체(58%): R f = 0.29 (CHCl3:EtOAc = 1:3); 1H NMR (500 MHz, CD3OD) δ 8.26 (s, 1H), 8.18 (s, 1H), 8.08 (d, J = 8.2 Hz, 1H), 7.48 (t, J = 8.0 Hz, 1H), 7.36 (d, J = 7.7 Hz, 1H), 2.69 (s, 3H), 1.61 (s, 6H); 13C NMR (125 MHz, CD3OD) δ 158.9, 156.1, 154.1, 143.5, 138.8, 128.8, 123.9, 123.8, 120.9, 100.5, 94.5, 80.5, 64.5, 30.3, 13.1.Pale yellow solid (58%): R f = 0.29 (CHCl 3 :EtOAc = 1:3); 1 H NMR (500 MHz, CD 3 OD) δ 8.26 (s, 1H), 8.18 (s, 1H), 8.08 (d, J = 8.2 Hz, 1H), 7.48 (t, J = 8.0 Hz, 1H), 7.36 (d, J = 7.7 Hz, 1H), 2.69 (s, 3H), 1.61 (s, 6H); 13 C NMR (125 MHz, CD 3 OD) δ 158.9, 156.1, 154.1, 143.5, 138.8, 128.8, 123.9, 123.8, 120.9, 100.5, 94.5, 80.5, 64.5, 30.3, 13.1.

3-(4-Amino-3-methylpyrazolo[3,4-3-(4-Amino-3-methylpyrazolo[3,4- dd ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol (14b)]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol (14b)

옅은 황색 고체(85%): R f = 0.33 (CHCl3:EtOAc = 1:3); 1H NMR (500 MHz, CD3OD) δ 8.14 (s, 1H), 8.06 (s, 1H), 7.98 (d, J = 8.0 Hz, 1H), 7.37 (dd, J = 8.0, 7.7 Hz, 1H), 7.26 (d, J = 7.7 Hz, 1H), 2.57 (s, 3H), 1.96-1.84 (m, 2H), 1.71-1.45 (m, 7H), 1.29-1.16 (m, 1H); 13C NMR (125 MHz, CD3OD) δ 158.9, 156.1, 154.2, 143.5, 138.8, 128.9, 128.8, 124.0, 123.8, 120.9, 100.5, 93.6, 68.0, 39.5, 25.0, 23.0, 13.1.Pale yellow solid (85%): R f = 0.33 (CHCl 3 :EtOAc = 1:3); 1H NMR (500 MHz, CD 3 OD) δ 8.14 (s, 1H), 8.06 (s, 1H), 7.98 (d, J = 8.0 Hz, 1H), 7.37 (dd, J = 8.0, 7.7 Hz, 1H ), 7.26 (d, J = 7.7 Hz, 1H), 2.57 (s, 3H), 1.96-1.84 (m, 2H), 1.71-1.45 (m, 7H), 1.29-1.16 (m, 1H); 13 C NMR (125 MHz, CD 3 OD) δ 158.9, 156.1, 154.2, 143.5, 138.8, 128.9, 128.8, 124.0, 123.8, 120.9, 100.5, 93.6, 68.0, 39.5, 25.0, 23.0, 13.1.

4-(3-(4-Amino-3-methylpyrazolo[3,4-4-(3-(4-Amino-3-methylpyrazolo[3,4- dd ]pyrimidin-1-yl)phenyl)-2-methylbutyn-1,2-diol (14c)]pyrimidin-1-yl)phenyl)-2-methylbutyn-1,2-diol (14c)

황색 고체(82%): R f = 0.13 (CHCl3:EtOAc = 1:3); 1H NMR (500 MHz, CD3OD) δ 8.26 (s, 1H), 8.21 (s, 1H), 8.09 (d, J = 8.0 Hz, 1H), 7.48 (dd, J = 8.0, 7.8 Hz, 1H), 7.40 (d, J = 7.8 Hz, 1H), 3.63 (s, 2H), 2.69 (s, 3H), 1.56 (s, 3H); 13C NMR (125 MHz, CD3OD) δ 156.1, 143.5, 138.8, 129.0, 128.8, 123.9, 123.8, 121.0, 92.2, 82.4, 69.7, 68.2, 60.1, 24.8, 19.4, 13.1.Yellow solid (82%): R f = 0.13 (CHCl 3 :EtOAc = 1:3); 1H NMR (500 MHz, CD 3 OD) δ 8.26 (s, 1H), 8.21 (s, 1H), 8.09 (d, J = 8.0 Hz, 1H), 7.48 (dd, J = 8.0, 7.8 Hz, 1H ), 7.40 (d, J = 7.8 Hz, 1H), 3.63 (s, 2H), 2.69 (s, 3H), 1.56 (s, 3H); 13 C NMR (125 MHz, CD 3 OD) δ 156.1, 143.5, 138.8, 129.0, 128.8, 123.9, 123.8, 121.0, 92.2, 82.4, 69.7, 68.2, 60.1, 24.8, 19.4, 13 .1.

4-(3-(4-Amino-3-methylpyrazolo[3,4-4-(3-(4-Amino-3-methylpyrazolo[3,4- dd ]pyrimidin-1-yl)phenyl)-2-phenylbutyn-2-ol (14d)]pyrimidin-1-yl)phenyl)-2-phenylbutyn-2-ol (14d)

옅은 황색 고체(46%): R f = 0.33 (CHCl3:EtOAc = 1:5); 1H NMR (500 MHz, CD3OD) δ 8.26 (s, 1H), 8.24 (t, J = 1.6 Hz, 1H), 8.12 (dd, J = 8.2, 1.0 Hz, 1H), 7.76-7.71 (m, 2H), 7.51 (t, J = 7.9 Hz, 1H), 7.46-7.37 (m, 3H), 7.31 (d, J = 7.4 Hz, 1H), 2.70 (s, 3H), 1.85 (s, 3H); 13C NMR (125 MHz, CD3OD) δ 158.9, 156.1, 154.1, 146.0, 143.6, 138.9, 128.9, 128.8, 127.8, 127.1, 124.7, 123.8, 123.7, 121.1, 100.5, 93.6, 83.0, 69.3, 32.5, 13.1.Pale yellow solid (46%): R f = 0.33 (CHCl 3 :EtOAc = 1:5); 1H NMR (500 MHz, CD 3 OD) δ 8.26 (s, 1H), 8.24 (t, J = 1.6 Hz, 1H), 8.12 (dd, J = 8.2, 1.0 Hz, 1H), 7.76-7.71 (m , 2H), 7.51 (t, J = 7.9 Hz, 1H), 7.46-7.37 (m, 3H), 7.31 (d, J = 7.4 Hz, 1H), 2.70 (s, 3H), 1.85 (s, 3H) ; 13 C NMR (125 MHz, CD 3 OD) δ 158.9, 156.1, 154.1, 146.0, 143.6, 138.9, 128.9, 128.8, 127.8, 127.1, 124.7, 123.8, 123.7, 121.1, 10 0.5, 93.6, 83.0, 69.3, 32.5, 13.1.

화합물 15a-b의 합성을 위한 일반 절차General procedure for synthesis of compounds 15a-b

단계 1. DCM 중 메탄설포닐 클로라이드(1.2당량)의 혼합물에 프로파길 알코올(1.0당량) 및 트리에틸아민(1.2당량)을 동시에 천천히 첨가하였다. 반응 혼합물을 실온에서 1시간 동안 교반하였다. 반응 혼합물을 물로 세척하고 MgSO4로 건조하고 여과하고 농축하여 추가 정제 없이 메탄술폰산 프로프-2-이닐 에스테르를 황색 액체로서 제공하였다.Step 1. Propargyl alcohol (1.0 equiv) and triethylamine (1.2 equiv) were slowly added simultaneously to a mixture of methanesulfonyl chloride (1.2 equiv) in DCM. The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was washed with water, dried over MgSO 4 , filtered and concentrated to provide methanesulfonic acid prop-2-ynyl ester as a yellow liquid without further purification.

단계 2. DCM 중 메탄설폰산 프로프-2-이닐 에스테르의 혼합물에 모르폴린 또는 피페리딘(2.0 당량)을 첨가하였다. 실온에서 4-12시간 동안 교반한 후, 반응 혼합물을 여과하였다. 합한 여액을 NaHCO3(aq)로 세척하고, MgSO4로 건조시키고, 여과하고, 진공에서 농축시켰다. 조 중간체를 추가 정제 없이 다음 단계에 사용하였다.Step 2. Morpholine or piperidine (2.0 equiv) was added to the mixture of methanesulfonic acid prop-2-ynyl ester in DCM. After stirring at room temperature for 4-12 hours, the reaction mixture was filtered. The combined filtrates were washed with NaHCO 3 (aq), dried over MgSO 4 , filtered and concentrated in vacuo. The crude intermediate was used in the next step without further purification.

단계 3. 질소 분위기에서 DMF 중 3-요오도-1H-피라졸로[3,4-d]피리미딘-4-아민(1.0 당량) 및 구리(I) 요오다이드(0.2 당량)의 혼합물에 테트라키스(트리페닐포스핀)팔라듐(0)(0.1 당량)을 첨가하였다. 실온에서 10분 동안 교반한 후, 트리에틸아민(2.0 당량) 및 프로파길 모르폴린 또는 프로파길 피페리딘(10.0 당량)을 첨가하였다. 반응 혼합물을 70℃에서 1.5시간 동안 가열하였다. 실온으로 냉각한 후, 혼합물을 셀라이트 패드를 통해 여과하고 EtOAc로 헹구었다. 물을 여액에 첨가하고 혼합물을 EtOAc로 추출하였다. 합한 유기층을 MgSO4상에서 건조시키고, 여과하고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피(EtOAc: MeOH)로 정제하여 15를 제공하였다.Step 3. Add tetramethylamine to a mixture of 3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1.0 equiv) and copper(I) iodide (0.2 equiv) in DMF under nitrogen atmosphere. Kiss(triphenylphosphine)palladium(0) (0.1 equivalent) was added. After stirring at room temperature for 10 minutes, triethylamine (2.0 equiv) and propargyl morpholine or propargyl piperidine (10.0 equiv) were added. The reaction mixture was heated at 70° C. for 1.5 hours. After cooling to room temperature, the mixture was filtered through a pad of Celite and rinsed with EtOAc. Water was added to the filtrate and the mixture was extracted with EtOAc. The combined organic layers were dried over MgSO 4 , filtered and concentrated in vacuo. The residue was purified by flash column chromatography (EtOAc:MeOH) to provide 15.

3-(3-Morpholin-4-ylprop-1-ynyl)-13-(3-Morpholin-4-ylprop-1-ynyl)-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (15a).]pyrimidin-4-ylamine (15a).

옅은 황색 고체(47%): R f = 0.22 (DCM:MeOH = 6:1); 1H NMR (500 MHz, CD3OD) δ 8.10 (s, 1H), 3.72-3.60 (m, 4H), 3.54 (s, 2H), 2.59 (br s, 4H).Pale yellow solid (47%): R f = 0.22 (DCM:MeOH = 6:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.10 (s, 1H), 3.72-3.60 (m, 4H), 3.54 (s, 2H), 2.59 (br s, 4H).

3-(3-Piperidin-1-ylprop-1-ynyl)-13-(3-Piperidin-1-ylprop-1-ynyl)-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (15b)]pyrimidin-4-ylamine (15b)

옅은 황색 고체(47%): R f = 0.22 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD3OD) δ 8.21 (s, 1H), 3.61 (s, 2H), 2.68 (br s, 4H), 1.74-1.65 (m, 4H), 1.53 (br s, 2H).Pale yellow solid (47%): R f = 0.22 (EtOAc:MeOH = 4:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.21 (s, 1H), 3.61 (s, 2H), 2.68 (br s, 4H), 1.74-1.65 (m, 4H), 1.53 (br s, 2H) .

화합물 16a-b의 합성을 위한 일반 절차General procedure for synthesis of compounds 16a-b

MeOH 중 15(1.0 당량)의 혼합물에 Pd/C(50 중량%)를 첨가하고 반응 혼합물을 수소 대기 하에 1.5시간 동안 교반하였다. 혼합물을 셀라이트 패드를 통해 여과하고, MeOH로 헹구고, 진공에서 농축시켰다. 잔류물을 플래시 컬럼 크로마토그래피(EtOAc:MeOH)로 정제하여 16을 제공하였다.To a mixture of 15 (1.0 equiv) in MeOH was added Pd/C (50% by weight) and the reaction mixture was stirred under hydrogen atmosphere for 1.5 hours. The mixture was filtered through a pad of Celite, rinsed with MeOH and concentrated in vacuo. The residue was purified by flash column chromatography (EtOAc:MeOH) to provide 16.

3-(3-Morpholin-4-ylpropyl)-13-(3-Morpholin-4-ylpropyl)-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (16a)]pyrimidin-4-ylamine (16a)

백색 고체(61%): R f = 0.15 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD3OD) δ 8.18 (s, 1H), 3.70 (t, J = 4.6 Hz, 4H), 3.03 (t, J = 7.1 Hz, 2H), 2.53-2.40 (m, 6H), 1.98 (quint., J = 7.1 Hz, 2H).White solid (61%): R f = 0.15 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD 3 OD) δ 8.18 (s, 1H), 3.70 (t, J = 4.6 Hz, 4H), 3.03 (t, J = 7.1 Hz, 2H), 2.53-2.40 (m, 6H) ), 1.98 (quint., J = 7.1 Hz, 2H).

3-(3-Piperidin-1-ylpropyl)-13-(3-Piperidin-1-ylpropyl)-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (16b)]pyrimidin-4-ylamine (16b)

황색 고체(97%): R f = 0.04 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD3OD) δ 8.16 (s, 1H), 3.02 (t, J = 7.2 Hz, 2H), 2.58-2.27 (m, 6H), 1.98 (quint., J = 7.2 Hz, 2H), 1.68-1.59 (m, 4H), 1.50 (br s, 2H).Yellow solid (97%): R f = 0.04 (EtOAc:MeOH = 4:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.16 (s, 1H), 3.02 (t, J = 7.2 Hz, 2H), 2.58-2.27 (m, 6H), 1.98 (quint., J = 7.2 Hz, 2H), 1.68-1.59 (m, 4H), 1.50 (br s, 2H).

1-(3-Bromophenyl)-3-(3-morpholin-4-ylpropyl)-11-(3-Bromophenyl)-3-(3-morpholin-4-ylpropyl)-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (17a)]pyrimidin-4-ylamine (17a)

백색 고체(57%): R f = 0.30 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD3OD) δ 8.30 (t, J = 1.9 Hz, 1H), 8.18 (s, 1H), 8.09 (dt, J = 8.1, 1.9 Hz, 1H), 7.37-7.29 (m, 2H), 3.61 (t, J = 5.0 Hz, 4H), 3.00 (t, J = 7.1 Hz, 2H), 2.50-2.38 (m, 6H), 1.98 (quint., J = 7.1 Hz, 2H).White solid (57%): R f = 0.30 (EtOAc:MeOH = 4:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.30 (t, J = 1.9 Hz, 1H), 8.18 (s, 1H), 8.09 (dt, J = 8.1, 1.9 Hz, 1H), 7.37-7.29 (m , 2H), 3.61 (t, J = 5.0 Hz, 4H), 3.00 (t, J = 7.1 Hz, 2H), 2.50-2.38 (m, 6H), 1.98 (quint., J = 7.1 Hz, 2H).

1-(3-Bromophenyl)-3-(3-piperidin-1-ylpropyl)-11-(3-Bromophenyl)-3-(3-piperidin-1-ylpropyl)-1 HH -pyrazolo[3,4--pyrazolo[3,4- dd ]pyrimidin-4-ylamine (17b)]pyrimidin-4-ylamine (17b)

(52%): R f = 0.14 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD3OD) δ 8.43 (t, J = 1.9 Hz, 1H), 8.30 (s, 1H), 8.24-8.20 (m, 1H), 7.51-7.42 (m, 2H), 3.10 (t, J = 7.2 Hz, 2H), 2.59-2.44 (m, 6H), 2.09 (quint., J = 7.2 Hz, 2H), 1.70-1.61 (m, 4H), 1.52 (br s, 2H).(52%): R f = 0.14 (EtOAc:MeOH = 4:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.43 (t, J = 1.9 Hz, 1H), 8.30 (s, 1H), 8.24-8.20 (m, 1H), 7.51-7.42 (m, 2H), 3.10 (t, J = 7.2 Hz, 2H), 2.59-2.44 (m, 6H), 2.09 (quint., J = 7.2 Hz, 2H), 1.70-1.61 (m, 4H), 1.52 (br s, 2H).

(3-(4-Amino-3-(3-morpholin-4-ylpropyl)pyrazolo[3,4-(3-(4-Amino-3-(3-morpholin-4-ylpropyl)pyrazolo[3,4- dd ]pyrimidin-1-yl)phenylethynyl)cyclohexanol (18a)]pyrimidin-1-yl)phenylethynyl)cyclohexanol (18a)

미색 고체(66%): R f = 0.33 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD3OD) δ 8.28 (s, 1H), 8.20 (t, J = 2.0 Hz, 1H), 8.14 (ddd, J = 8.0, 2.0, 1.0 Hz, 1H), 7.50 (t, J = 8.0 Hz, 1H), 7.38 (dt, J = 8.0, 1.0 Hz, 1H), 3.72 (t, J = 4.6 Hz, 4H), 3.12 (t, J = 7.1 Hz, 2H), 2.58-2.43 (m, 6H), 2.13-1.98 (m, 4H), 1.83-1.59 (m, 7H), 1.35 (br s, 1H); 13C NMR (125 MHz, CD3OD) δ 156.0, 154.1, 146.9, 138.9, 128.8, 124.0, 123.7, 120.8, 100.4, 93.7, 82.9, 68.0, 66.2, 57.0, 53.3, 39.5, 39.0, 25.2, 25.0, 24.9, 23.0, 13.1.Off-white solid (66%): R f = 0.33 (EtOAc:MeOH = 4:1); 1 H NMR (500 MHz, CD 3 OD) δ 8.28 (s, 1H), 8.20 (t, J = 2.0 Hz, 1H), 8.14 (ddd, J = 8.0, 2.0, 1.0 Hz, 1H), 7.50 (t , J = 8.0 Hz, 1H), 7.38 (dt, J = 8.0, 1.0 Hz, 1H), 3.72 (t, J = 4.6 Hz, 4H), 3.12 (t, J = 7.1 Hz, 2H), 2.58-2.43 (m, 6H), 2.13-1.98 (m, 4H), 1.83-1.59 (m, 7H), 1.35 (br s, 1H); 13 C NMR (125 MHz, CD 3 OD) δ 156.0, 154.1, 146.9, 138.9, 128.8, 124.0, 123.7, 120.8, 100.4, 93.7, 82.9, 68.0, 66.2, 57.0, 53.3, 3 9.5, 39.0, 25.2, 25.0, 24.9, 23.0, 13.1.

(3-[4-Amino-3-(3-piperidin-1-ylpropyl)pyrazolo[3,4-(3-[4-Amino-3-(3-piperidin-1-ylpropyl)pyrazolo[3,4- dd ]pyrimidin-1-yl]phenylethynyl)cyclohexanol (18b)]pyrimidin-1-yl]phenylethynyl)cyclohexanol (18b)

미색 고체 (40%): R f = 0.36 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD3OD) δ 8.22 (s, 1H), 8.17 (s, 1H), 8.10 (d, J = 8.0 Hz, 1H), 7.43 (dd, J = 8.0, 7.6 Hz, 1H), 7.32 (d, J = 7.6 Hz, 1H), 3.00 (t, J = 7.0 Hz, 2H), 2.48-2.30 (m, 5H), 2.04-1.93 (m, 4H), 1.80-1.37 (m, 14H), 1.28 (s, 1H).Off-white solid (40%): R f = 0.36 (EtOAc:MeOH = 4:1); 1H NMR (500 MHz, CD 3 OD) δ 8.22 (s, 1H), 8.17 (s, 1H), 8.10 (d, J = 8.0 Hz, 1H), 7.43 (dd, J = 8.0, 7.6 Hz, 1H ), 7.32 (d, J = 7.6 Hz, 1H), 3.00 (t, J = 7.0 Hz, 2H), 2.48-2.30 (m, 5H), 2.04-1.93 (m, 4H), 1.80-1.37 (m, 14H), 1.28 (s, 1H).

<1-3> 키나아제 활성 분석에 따른 SPA7012의 우수한 PAK4 효소 억제 활성 확인<1-3> Confirmation of excellent PAK4 enzyme inhibitory activity of SPA7012 according to kinase activity analysis

일련의 피리도[4,3-b]인돌 유도체 및 피라졸로[3,4-d]피리미딘 유도체를 합성하고, 합성된 화합물에 대해 시험관 내 PAK4 효소 억제 활성을 평가하기 위하여, 단일 농도(20μM)에서 상대 효소 억제 활성에 대한 HotSpot 키나아제 분석, 및 10μM의 ATP 농도에서 IC50 평가를 통해 In vitro 키나아제 분석을 수행하였다.To synthesize a series of pyrido[4,3-b]indole derivatives and pyrazolo[3,4-d]pyrimidine derivatives and to evaluate the in vitro PAK4 enzyme inhibitory activity of the synthesized compounds at a single concentration (20 μM), ), an in vitro kinase assay was performed using the HotSpot kinase assay for relative enzyme inhibitory activity, and IC 50 evaluation at an ATP concentration of 10 μM.

구체적으로, 효소 활성을 조사하기 위해 Reaction Biology Corporation(Malvern, PA, USA)에서 HotSpot kinase assay를 수행하였다. 키나아제 및 기질 쌍은 반응 완충액에서 준비되었다: pH 7.5의 20mM HEPES, 10mM MgCl2, 1mM EGTA, 0.02% Brij 35, 0.02mg/mL BSA, 0.1mM Na3VO4, 2mM DTT 및 1% DMSO. 실시예 1에서 제조한 각 화합물을 키나아제 반응 혼합물에 첨가하고 20분 동안 인큐베이션하였다. 그 후, 33P-ATP를 반응 혼합물에 전달하여 10μM의 ATP 농도에서 반응을 개시하였다. 실온에서 2시간 후, 반응 혼합물을 P81 이온 교환 종이에 스폿팅한 후, 필터-바인딩(filter-binding) 방법으로 키나아제 활성을 검출하였다. 20μM의 농도의 화합물에 대해 단일 용량 효소 억제(single dose enzyme inhibition) 테스트를 수행하였다. 대조 화합물인 스타우로스포린(staurosporine)은 10회 용량 IC50 모드에서 테스트되었다. IC50 값을 결정하기 위해 화합물을 100μM에서 시작하여 3배 연속 희석으로 10회 투여량에 대해 분석하였다.Specifically, to investigate enzyme activity, HotSpot kinase assay was performed by Reaction Biology Corporation (Malvern, PA, USA). Kinase and substrate pairs were prepared in reaction buffer: 20mM HEPES, 10mM MgCl2, 1mM EGTA, 0.02% Brij 35, 0.02mg/mL BSA, 0.1mM Na3VO4, 2mM DTT and 1% DMSO at pH 7.5. Each compound prepared in Example 1 was added to the kinase reaction mixture and incubated for 20 minutes. Afterwards, 33P-ATP was transferred to the reaction mixture to initiate the reaction at an ATP concentration of 10 μM. After 2 hours at room temperature, the reaction mixture was spotted on P81 ion exchange paper, and the kinase activity was detected by filter-binding method. A single dose enzyme inhibition test was performed on the compound at a concentration of 20 μM. The control compound, staurosporine, was tested in 10 dose IC 50 mode. To determine IC 50 values, compounds were analyzed for 10 doses in 3-fold serial dilutions starting at 100 μM.

단일 농도(20μM)에서 상대 효소 억제 활성에 대한 HotSpot 키나아제 분석, 및 10μM의 ATP 농도에서 IC50 평가를 수행한 결과 결과, 이환식 피라졸로[3,4-d]피리미딘 유도체(화합물 4 내지 9)는 삼환식 피리도[4,3-b]인돌 유도체(화합물 1 내지 3)보다 더 나은 PAK4 효소 억제 활성을 나타내는 것으로 확인되었다(표 3). Results of a HotSpot Kinase Assay for relative enzyme inhibitory activity at a single concentration (20 μM), and IC 50 evaluation at an ATP concentration of 10 μM for the bicyclic pyrazolo[3,4-d]pyrimidine derivatives (compounds 4 to 9). was confirmed to exhibit better PAK4 enzyme inhibitory activity than tricyclic pyrido[4,3-b]indole derivatives (compounds 1 to 3) (Table 3).

화합물compound PAK4 효소 억제PAK4 enzyme inhibition clogPclogP 억제 활성(%)Inhibitory activity (%) IC50 (μM)IC 50 (μM) 화합물 1Compound 1 3232 NDN.D. 3.793.79 화합물 2compound 2 5454 34.034.0 5.105.10 화합물 3Compound 3 2929 NDN.D. 2.662.66 화합물 4Compound 4 6060 NDN.D. 1.251.25 화합물 5Compound 5 9797 0.770.77 2.572.57 화합물 6Compound 6 3636 NDN.D. 0.120.12 화합물 7Compound 7 6565 NDN.D. 2.592.59 화합물 8Compound 8 8888 9.929.92 2.572.57 화합물 9Compound 9 9090 4.414.41 3.783.78

또한, 알로스테릭 포켓 바인더의 경우, 사이클로헥실기(SPA7012)가 더 작은 디메틸(SPA7011) 또는 하이드록시메틸기(SPA7013)보다 더 선호된 것으로 나타났으며, 방향족 고리(SPA7014)는 포화 고리(SPA7012)에 비해 활성 감소시키는 것으로 확인되었다. In vitro 효소 IC50 값은 SPA7012, SPA7017, SPA7016의 순서로 나타났다(도 5). Additionally, for the allosteric pocket binder, the cyclohexyl group (SPA7012) was found to be preferred over the smaller dimethyl (SPA7011) or hydroxymethyl group (SPA7013), and the aromatic ring (SPA7014) was found to be preferred over the saturated ring (SPA7012). It was confirmed that activity was reduced compared to . In vitro enzyme IC 50 values appeared in the following order: SPA7012, SPA7017, and SPA7016 (Figure 5).

서브마이크로몰 IC50 값을 갖는 화합물 SPA7012의 서브타입 선택성을 평가하기 위해 선택되었고(표 S3), PAK1(16% 억제) 및 PAK2(36% 억제, > 100μM of IC50) 대비 PAK4(81% 억제, 0.77 μM의 IC50)에 대해 우수한 선택성을 나타내는 것으로 확인되었다(표 4).Compound SPA7012 was selected to evaluate the subtype selectivity with submicromolar IC 50 values (Table S3) and PAK4 (81% inhibition) compared to PAK1 (16% inhibition) and PAK2 (36% inhibition, > 100 μM of IC 50 ). , IC 50 of 0.77 μM) was confirmed to exhibit excellent selectivity (Table 4).

SubtypeSubtype 억제 활성(%)Inhibitory activity (%) IC50 (μM)IC 50 (μM) PAK1PAK1 1616 NDN.D. PAK2PAK2 3636 -- PAK3PAK3 55 NDN.D. PAK4PAK4 8181 0.770.77 PAK5PAK5 5252 >100>100 PAK6PAK6 3636 --

<1-4> 분자 도킹 분석에 따른 SPA7012의 우수한 PAK4 선택성 확인<1-4> Confirmation of excellent PAK4 selectivity of SPA7012 according to molecular docking analysis

GNE-2861(PDB 코드: 4O0V)과 콤플렉스를 형성한 인간 PAK4 촉매 도메인 구조에 대해 SPA7012의 분자 도킹 분석을 수행하였다.Molecular docking analysis of SPA7012 was performed on the structure of the human PAK4 catalytic domain in complex with GNE-2861 (PDB code: 4O0V).

구체적으로, 분자 도킹은 Cresset Flare V.5.0(Litlington, Cambridgeshire, UK)을 사용하여 수행되었다. 활성 부위의 3D 좌표는 RCSB Protein Data Bank의 리간드(PDB 코드 4O0V)와 복합체에서 인간 PAK4의 보고된 결정 구조에서 가져왔다. 단백질 구조를 준비하여 최소화한 후, 클러스터링된 리간드에 따라 결합 부위의 그리드 박스를 정의하였다. 합성된 화합물의 도킹 계산은 일반 도킹 모드에서 Flare V.5.0을 사용하여 수행되었다.Specifically, molecular docking was performed using Cresset Flare V.5.0 (Litlington, Cambridgeshire, UK). The 3D coordinates of the active site were taken from the reported crystal structure of human PAK4 in complex with its ligand (PDB code 4O0V) from the RCSB Protein Data Bank. After preparing and minimizing the protein structure, a grid box of the binding site was defined according to the clustered ligands. Docking calculations of the synthesized compounds were performed using Flare V.5.0 in normal docking mode.

그 결과, SPA7012의 결합 자세는 특정 백 포켓에 알로스테릭 결합제가 적절하게 위치하여 GNE-2861의 결정 구조와 오버랩되는 것으로 나타났다. 소수성 알로스테릭 바인더의 3차 알코올(tertiary alcohol)의 산소 원자는 Phe459의 백본 및 Glu366의 측쇄와 수소 결합을 형성하였다. 피라졸로[3,4-d]피리미딘 코어 스캐폴드의 아민 그룹과 질소원자 역시 힌지영역에서 Lue398과 수소결합을 형성하였다. 따라서, 주요 화합물로써 SPA7012를 선정하여 이후의 실험을 수행하였다.As a result, the binding pose of SPA7012 was shown to overlap with the crystal structure of GNE-2861 with the allosteric binder appropriately located in a specific back pocket. The oxygen atom of the tertiary alcohol of the hydrophobic allosteric binder formed a hydrogen bond with the backbone of Phe459 and the side chain of Glu366. The amine group and nitrogen atom of the pyrazolo[3,4-d]pyrimidine core scaffold also formed hydrogen bonds with Lue398 in the hinge region. Therefore, SPA7012 was selected as the main compound and subsequent experiments were performed.

실시예 2. SPA7012의 PAK4 표적화에 따른 간 허혈 재관류 손상 개선 효과 확인을 위한 실험 방법Example 2. Experimental method to confirm the effect of SPA7012 on improving liver ischemia-reperfusion damage by targeting PAK4

<2-1> 부분 간 I/R 손상 모델<2-1> Inter-part I/R damage model

병원균이 없는 8-10주령 C57BL/6 수컷 마우스(Orient Bio, Seoul, Korea)는 22℃에서 습도 50%로 조절된 12시간 명암 주기 환경에서 음식과 물에 자유롭게 접근할 수 있도록 유지되었다. 간 I/R 손상은 간문맥, 간동맥 및 우측 분지 바로 위의 담관을 폐쇄하여 생성되었다.Pathogen-free 8- to 10-week-old C57BL/6 male mice (Orient Bio, Seoul, Korea) were maintained in a 12-h light/dark environment controlled at 22°C and 50% humidity with free access to food and water. Hepatic I/R injury was created by occlusion of the portal vein, hepatic artery, and bile duct immediately above the right branch.

구체적으로, 마우스를 케타민(100mg/kg) 및 자일라진(10mg/kg)으로 복강내 주사로 마취시킨후 절개하고 간문맥, 간동맥 및 우측 분지 바로 위의 담관을 가로질러 비외상성 클립을 삽입하여 간으로 가는 전체 혈액 공급의 약 70%를 차지하는 좌측엽과 정중엽으로의 혈류를 차단하였다. 간은 식염수를 적신 거즈로 촉촉하게 유지하고 체온은 허혈 기간 동안 따뜻한 담요로 37℃로 유지하였다. 부분 간 허혈 60분 후 클립을 제거하여 재관류를 시작하였다. 대조군 마우스에 대해서는 혈관 폐색 없이 동일한 수술을 수행하였다. 충분한 재관류 기간 후, 마우스를 마취하에 방혈(exsanguination)하여 희생시키고 혈청 샘플을 수집하였다. 간의 우측 및 정중엽을 수집하여 추가 분석이 있을 때까지 -80℃에 보관하거나 즉시 10% 포르말린에 고정시켰다. 모든 동물 실험은 미국 국립 보건원(NIH Publication No. 85-23, 개정 2011)에서 발행한 실험 동물의 관리 및 사용에 대한 지침에 따라 수행되었습니다. 연구 프로토콜은 전북대학교 동물보호 및 이용위원회(허가번호: JBNU-2019-00122)의 승인을 받았다.Specifically, mice were anesthetized by intraperitoneal injection with ketamine (100 mg/kg) and xylazine (10 mg/kg), then dissected, and atraumatic clips were inserted across the portal vein, hepatic artery, and bile duct just above the right branch into the liver. Blood flow to the left and median lobes, which account for about 70% of the total blood supply, was blocked. The liver was kept moist with saline-soaked gauze, and body temperature was maintained at 37°C with a warm blanket during the ischemic period. After 60 minutes of partial liver ischemia, the clip was removed and reperfusion was initiated. The same surgery was performed on control mice without vascular occlusion. After a sufficient reperfusion period, mice were sacrificed by exsanguination under anesthesia and serum samples were collected. The right and median lobes of the liver were collected and stored at -80°C until further analysis or immediately fixed in 10% formalin. All animal experiments were performed in accordance with the Guide for the Care and Use of Laboratory Animals published by the National Institutes of Health (NIH Publication No. 85-23, revised 2011). The research protocol was approved by the Chonbuk National University Animal Care and Use Committee (permit number: JBNU-2019-00122).

<2-2> 1차 간세포의 분리<2-2> Isolation of primary hepatocytes

8-10주령 C57BL/6 수컷 마우스로부터 1차 간세포를 관류에 의해 제조하였다. 하대정맥에 삽관한 후, 칼슘이 없는 HEPES 완충액(10mM, pH7.4)을 0.35ml/min의 유속으로 5분 동안 간을 관류시키고, 5mM 염화칼슘과 함께 콜라게나제 IV(Sigma-Aldrich, St. Louis, MO, USA) 5㎍을 함유하는 HEPES 완충액을 5분 동안 관류하였다. 간세포를 42% Percoll과 혼합된 10% FBS, 10units/ml 페니실린, 10㎍/ml 스트렙토마이신 및 10nM 덱사메타손이 보충된 DMEM/F12에 재현탁하고 1,300rpm에서 5분 동안 원심분리하였다. 세포를 6웰 배양 접시에 1×106세포/웰로 플레이팅하였다.Primary hepatocytes were prepared by perfusion from 8-10 week old C57BL/6 male mice. After cannulation of the inferior vena cava, the liver was perfused with calcium-free HEPES buffer (10mM, pH7.4) for 5 minutes at a flow rate of 0.35ml/min, and collagenase IV (Sigma-Aldrich, St. Louis, MO, USA) perfused with HEPES buffer containing 5 μg for 5 minutes. Hepatocytes were resuspended in DMEM/F12 supplemented with 10% FBS mixed with 42% Percoll, 10 units/ml penicillin, 10 μg/ml streptomycin, and 10 nM dexamethasone and centrifuged at 1,300 rpm for 5 min. Cells were plated at 1×106 cells/well in a 6-well culture dish.

<2-3> 저산소증-재산소화 프로토콜<2-3> Hypoxia-reoxygenation protocol

1차 간세포는 산소 흡수 팩(AnaeroGen, Oxoid)과 함께 37℃의 혐기성 병(Oxoid, Basingstoke, Hampshire, UK)에서 배양되었다. 이 방법은 병의 산소 수준을 1% 미만으로 달성하는 것으로 나타났다. 다양한 기간의 저산소증 후, 챔버를 열고 저산소 배지를 산소가 함유된 배지로 교체하여 세포의 재산소화를 개시하였다Primary hepatocytes were cultured in anaerobic bottles (Oxoid, Basingstoke, Hampshire, UK) at 37°C with oxygen absorption packs (AnaeroGen, Oxoid). This method has been shown to achieve bottle oxygen levels of less than 1%. After various periods of hypoxia, the chamber was opened and the hypoxic medium was replaced with oxygenated medium to initiate reoxygenation of the cells.

<2-4> 세포 배양 및 일시적 형질감염<2-4> Cell culture and transient transfection

인간 배아 신장 세포주 HEK293T는 American Type Culture Collection(Manassas, VA, USA)에서 입수하였다. Nrf2 리포터 유전자 분석을 위해, ARE 유도 루시페라제 발현을 갖는 프로모터를 함유하는 플라스미드 1㎍을 사용하였다. HEK293T 세포에 1㎍의 Nrf2 및 PAK4를 Lipofectamine 3000(Invitrogen, Carlsbad, CA, USA)으로 형질도입하여 외인성 단백질을 발현시켰다. Lumat LB 9507(Berthold, Bad Wildbad, Germany)의 Dual-Luciferase Reporter Assay(Promega, Madison, WI, USA)를 사용하여 루시퍼라제 활성을 측정하였다.The human embryonic kidney cell line HEK293T was obtained from the American Type Culture Collection (Manassas, VA, USA). For Nrf2 reporter gene analysis, 1 μg of plasmid containing a promoter with ARE-driven luciferase expression was used. HEK293T cells were transduced with 1 μg of Nrf2 and PAK4 with Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA) to express exogenous proteins. Luciferase activity was measured using the Dual-Luciferase Reporter Assay (Promega, Madison, WI, USA) on Lumat LB 9507 (Berthold, Bad Wildbad, Germany).

<2-5> 세포 생존력에 대한 MTT 분석<2-5> MTT assay for cell viability

세포 생존율은 3-(4,5-디메틸티아졸-2-일)-2,5-디페닐테트라졸륨 브로마이드(MTT)의 포르마잔으로의 환원을 분석하여 결정되었다.Cell viability was determined by analyzing the reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to formazan.

<2-6> 세포사멸의 유세포 분석<2-6> Flow cytometry analysis of apoptosis

저산소증 재산소화(Hypoxia-reoxygenation, H/R)에 의해 유도된 세포 사멸은 제조사의 지시에 따라 Annexin V-propidium iodide(PI) 이중 염색 키트(BD Biosciences, San Jose, CA, USA)를 사용하여 모니터링되었으며, 유세포 분석(BD Biosciences)으로 정량화되었다. 세포사멸된 세포의 비율은 PI 양성 세포에 대한 Annexin V 양성 세포의 백분율로 정의되었다.Cell death induced by hypoxia-reoxygenation (H/R) was monitored using Annexin V-propidium iodide (PI) double staining kit (BD Biosciences, San Jose, CA, USA) according to the manufacturer's instructions. and quantified by flow cytometry (BD Biosciences). The proportion of apoptotic cells was defined as the percentage of Annexin V-positive cells to PI-positive cells.

<2-7> 생화학적 분석<2-7> Biochemical analysis

간 조직내 혈청(Asan Pharm, Seoul, Korea), 글루타티온(GSH, Arbor Assays, Ann Arbor, MI, USA) 및 말론알데하이드(malondialdehyde)(MDA, ab118970, Abcam, Cambridge, UK) 중 알라닌 아미노전이효소(Alanine aminotransferase, ALT) 및 아스파르테이트 아미노전이효소(aspartate aminotransferase, AST), 및 배양 배지 중 젖산 탈수소효소(lactate dehydrogenase, LDH, Biovision, Milpitas, CA, USA)를 이의 분석에 사용되는 키트를 사용하여 분석하였다. IL-1βIL-6, TNF-α 및 CCL-2의 혈청 수준은 ELISA 키트(모두 Invitrogen에서 제공)를 사용하여 결정되었다.Alanine aminotransferase (aminotransferase) in liver tissue serum (Asan Pharm, Seoul, Korea), glutathione (GSH, Arbor Assays, Ann Arbor, MI, USA) and malondialdehyde (MDA, ab118970, Abcam, Cambridge, UK). Alanine aminotransferase (ALT) and aspartate aminotransferase (AST), and lactate dehydrogenase (LDH, Biovision, Milpitas, CA, USA) in the culture medium were analyzed using kits used for their analysis. analyzed. Serum levels of IL-1βIL-6, TNF-α, and CCL-2 were determined using ELISA kits (all from Invitrogen).

<2-8> RNA 분리 및 qPCR<2-8> RNA isolation and qPCR

동결된 간조직 또는 TRIzol 시약(Invitrogen)이 처리된 1차 간세포에서 총 RNA를 추출하였다. RNA를 이소프로판올(isopropanol)로 침전시키고 70% 에탄올로 건조시킨 후 디에틸 피로카보네이트(diethyl pyrocarbonate) 처리된 증류수에 녹였다. first-strand cDNA 합성 키트(Applied Biosystems, Foster City, CA, USA)에서 제공된 무작위 6량체 프라이머를 사용하여 첫 번째 가닥 cDNA를 생성하였다. 프라이머는 PrimerBank(https://pga.mgh.harvard.edu/primerbank, 표 5)를 사용하여 설계되었다. qPCR 반응은 역전사된 총 RNA 10ng, 정방향 및 역방향 프라이머 200nM 및 PCR 마스터 혼합물을 포함하는 최종부피 10ml 시약으로 수행되었다.Total RNA was extracted from frozen liver tissue or primary hepatocytes treated with TRIzol reagent (Invitrogen). RNA was precipitated with isopropanol, dried with 70% ethanol, and then dissolved in distilled water treated with diethyl pyrocarbonate. First-strand cDNA was generated using random hexamer primers provided in a first-strand cDNA synthesis kit (Applied Biosystems, Foster City, CA, USA). Primers were designed using PrimerBank (https://pga.mgh.harvard.edu/primerbank, Table 5). qPCR reactions were performed with a final volume of 10 ml reagent containing 10 ng of reverse transcribed total RNA, 200 nM forward and reverse primers, and PCR master mixture.

유전자gene 핵산서열(5'→3')Nucleic acid sequence (5'→3') Accession No.Accession No. 서열번호sequence number Tnfatnfa forwardforward AGGGTCTGGGCCATAGAACTAGGGTCTGGGCCATAGAACT NM_013693NM_013693 서열번호 1SEQ ID NO: 1 reversereverse CCACCACGCTCTTCTGTCTACCCACCACGCTCTTCTGTCTAC 서열번호 2SEQ ID NO: 2 Il1bIl1b forwardforward GGTCAAAGGTTTGGAAGCAGGGTCAAAGGTTTGGAAGCAG NM_008361NM_008361 서열번호 3SEQ ID NO: 3 reversereverse TGTGAAATGCCACCTTTTGATGTGAAATGCCACCTTTTGA 서열번호 4SEQ ID NO: 4 Il6IL6 forwardforward AGGGTCTGGGCCATAGAACTAGGGTCTGGGCCATAGAACT NM_013693NM_013693 서열번호 5SEQ ID NO: 5 reversereverse CCACCACGCTCTTCTGTCTACCCACCACGCTCTTCTGTCTAC 서열번호 6SEQ ID NO: 6 Icam1Icam1 forwardforward AACAGTTCACCTGCACGGACAACAGTTCACCTGCACGGAC NM_010493NM_010493 서열번호 7SEQ ID NO: 7 reversereverse GTCACCGTTGTGATCCCTGGTCACCGTTTGTGATCCCTG 서열번호 8SEQ ID NO: 8 Cxcl1Cxcl1 forwardforward AATGAGCTGCGCTGTCAGTGAATGAGCTGCGCTGTCAGTG NM_008176NM_008176 서열번호 9SEQ ID NO: 9 reversereverse TGAGGGCAACACCTTCAAGCTGAGGGCAACACCTTCAAGC 서열번호 10SEQ ID NO: 10 Ccl2Ccl2 forwardforward ACCGACAACAGGAAGTGGAGACCGACAACAGGAAGTGGAG NM_009140NM_009140 서열번호 11SEQ ID NO: 11 reversereverse TGGACGTTTCACACAGTGGTTGGACGTTTCACACAGTGGT 서열번호 12SEQ ID NO: 12 Ho1Ho1 forwardforward AGGTACACATCCAAGCCGAGAAGGTACACATCCAAGCCGAGA NM_010442NM_010442 서열번호 13SEQ ID NO: 13 reversereverse CATCACCAGCTTAAAGCCTTCTCATCACCAGCTTAAAGCCTTCT 서열번호 14SEQ ID NO: 14 Nqo1Nqo1 forwardforward AGGATGGGAGGTACTCGAATCAGGATGGGAGGTACTCGAATC NM_008706NM_008706 서열번호 15SEQ ID NO: 15 reversereverse TGCTAGAGATGACTCGGAAGGTGCTAGAGATGACTCGGAAGG 서열번호 16SEQ ID NO: 16 Nrf2Nrf2 forwardforward CTTTAGTCAGCGACAGAAGGACCTTTAGTCAGCGACAGAAGGAC NM_010902NM_010902 서열번호 17SEQ ID NO: 17 reversereverse AGGCATCTTGTTTGGGAATGTGAGGCATCTTGTTTGGGAATGTG 서열번호 18SEQ ID NO: 18 GapdhGapdh forwardforward CGTCCCGTAGACAAAATGGTCGTCCCGTAGACAAAATGGT NM_008084NM_008084 서열번호 19SEQ ID NO: 19 reversereverse TTGATGGCAACAATCTCCACTTGATGGCAACAATCTCCAC 서열번호 20SEQ ID NO: 20

<2-9> 세포하 분획화, 웨스턴 블로팅 및 공동 면역 침전(Co-IP)<2-9> Subcellular fractionation, Western blotting and co-immunoprecipitation (Co-IP)

핵 및 세포질 분획은 NE-PER 핵 및 세포질 추출 키트(Thermo Fisher Scientific, Waltham, MA, USA)를 사용하여 준비하였다. 조직 균질 현탁액 또는 세포 용해물(15㎍)을 6 내지 14% SDS-PAGE로 분리하고 PVDF 막으로 옮겼다. 5% 스킴 밀크(skim milk)로 블로킹한 후, PAK4 (G222), p-PAK4 (S474), p-IKKβ(S176/180), p-IKBα (S32), p-p65 (S536), cleaved caspase-3 (Asp175), Bax (Cell Signaling Technology, Beverly, MA, USA), GAPDH (A531), lamin B1(L75), Bcl2(P65), NQO1 (F252) (Bioworld Technology, St Louis Park, MN, USA), NF-κp65 (C-20), IKBα (H-4), IKKβ(10A9B6) (Santa Cruz Biotechnology, Dallas, TX, USA), Nrf2 (Proteintech, Rosemont, IL, USA), HO-1 (Enzo Life Sciences, Farmingdale, New York, USA), p-Thr (Merck KGaA, Darmstadt, Germany)에 대한 1차 항체를 사용하여 블롯을 탐침하였다. 공동 면역침강반응을 위하여, 600㎍ 단백질을 4℃에서 밤새 항-Nrf2 항체(Proteintech, Sankt Leon-Rot, Germany)와 함께 배양한 다음, 4℃에서 2시간 동안 단백질 G 아가로스를 배양하고, PAK4, Nrf2 또는 p-Thr에 대한 항체로 블롯을 탐침하였다. 면역 반응 밴드는 Las-4000 이미저(GE Healthcare Life Science, Pittsburgh, PA, USA)로 검출하였다.Nuclear and cytoplasmic fractions were prepared using the NE-PER nuclear and cytoplasmic extraction kit (Thermo Fisher Scientific, Waltham, MA, USA). Tissue homogenate suspensions or cell lysates (15 μg) were separated by 6 to 14% SDS-PAGE and transferred to PVDF membranes. After blocking with 5% skim milk, PAK4 (G222), p-PAK4 (S474), p-IKKβ (S176/180), p-IKBα (S32), p-p65 (S536), cleaved caspase -3 (Asp175), Bax (Cell Signaling Technology, Beverly, MA, USA), GAPDH (A531), lamin B1 (L75), Bcl2 (P65), NQO1 (F252) (Bioworld Technology, St Louis Park, MN, USA ), NF-κp65 (C-20), IKBα (H-4), IKKβ(10A9B6) (Santa Cruz Biotechnology, Dallas, TX, USA), Nrf2 (Proteintech, Rosemont, IL, USA), HO-1 (Enzo Blots were probed using primary antibodies against p-Thr (Merck KGaA, Darmstadt, Germany) (Life Sciences, Farmingdale, New York, USA). For co-immunoprecipitation reactions, 600 μg protein was incubated with anti-Nrf2 antibody (Proteintech, Sankt Leon-Rot, Germany) overnight at 4°C, then incubated with protein G agarose for 2 h at 4°C, and PAK4 , blots were probed with antibodies against Nrf2 or p-Thr. Immunoreactive bands were detected with a Las-4000 imager (GE Healthcare Life Science, Pittsburgh, PA, USA).

<2-10> 조직학<2-10> Histology

간 조직(5μm)의 파라핀 절편을 헤마톡실린과 에오신(H&E)으로 염색한 후 광학 현미경으로 검출하였다. 슬라이드당 5개의 무작위 섹션을 조사하여 괴사 영역을 측정하고 세포 사멸 세포의 백분율을 결정하였다. 괴사 영역을 측정하기 위하여, Axiovert 40 CFL 현미경(Carl Zeiss, Oberkochen, Germany)으로 절편을 관찰하고, iSolution DT 36 소프트웨어(Carl Zeiss)를 사용하여 측정하였다. 면역형광염색을 위하여, 탈파라핀화 후 섹션을 F4/80(ab6640), Ly6G(ab25377), 4-하이드록시노넨알(4-HNE, ab46545)(모두 Abcam에서 제공)에 대한 항체로 면역염색하였다. TUNEL 염색은 상용 키트(Promega)로 수행되었다. PBS로 세척한 후 2차 항체(Alexa Fluor 488-conjugated goat anti-mouse IgG1 and Alexa Fluor 594-conjugated goat anti-rabbit IgM, Thermo Fisher Scientific)와 함께 37℃에서 1시간 동안 배양하였으며, 섹션은 DAPI로 대조 염색되었다.Paraffin sections of liver tissue (5 μm) were stained with hematoxylin and eosin (H&E) and detected by light microscopy. Five random sections per slide were examined to measure necrotic areas and determine the percentage of apoptotic cells. To measure the necrotic area, sections were observed with an Axiovert 40 CFL microscope (Carl Zeiss, Oberkochen, Germany) and measured using iSolution DT 36 software (Carl Zeiss). For immunofluorescence staining, after deparaffinization, sections were immunostained with antibodies against F4/80 (ab6640), Ly6G (ab25377), and 4-hydroxynonenal (4-HNE, ab46545) (all provided by Abcam). . TUNEL staining was performed with a commercial kit (Promega). After washing with PBS, the sections were incubated with secondary antibodies (Alexa Fluor 488-conjugated goat anti-mouse IgG1 and Alexa Fluor 594-conjugated goat anti-rabbit IgM, Thermo Fisher Scientific) at 37°C for 1 hour, and the sections were incubated with DAPI. were counterstained.

<2-11> 통계분석<2-11> Statistical analysis

데이터는 평균(SD)의 평균±표준 편차로 표시되었다. 통계적 비교는 일원 분산 분석에 이어 Tukey의 사후 검정을 사용하여 이루어졌고, 두 그룹 간의 차이의 유의성은 Student's unpaired t-test를 사용하여 결정되었으며, 0.05 미만의 p-값은 유의한 것으로 간주되었다.Data were expressed as mean ± standard deviation of the mean (SD). Statistical comparisons were made using one-way analysis of variance followed by Tukey's post hoc test, the significance of differences between two groups was determined using Student's unpaired t-test, and a p-value of less than 0.05 was considered significant.

실시예 3. SPA7012의 PAK4 표적화에 따른 간 허혈 재관류 손상 보호 효과 확인Example 3. Confirmation of the protective effect of SPA7012 on liver ischemia-reperfusion damage by targeting PAK4

<3-1> SPA7012의 복강 내 투여에 따른 마우스에서 간 I/R 손상 보호 효과 확인<3-1> Confirmation of liver I/R damage protection effect in mice following intraperitoneal administration of SPA7012

SPA7012가 1시간의 허혈과 6시간 또는 24시간의 재관류를 받은 마우스에서 간 손상에 영향을 미치는지 여부를 조사하였다(도 6). We investigated whether SPA7012 affected liver damage in mice subjected to 1 hour of ischemia and 6 or 24 hours of reperfusion (Figure 6).

그 결과, AST 및 ALT의 혈청 수준에 의해 평가된 간 손상은 대조군 마우스 대비 I/R을 받은 마우스에서 명백한 것으로 확인되었다(도 7). 그러나 I/R 손상 전 및 손상 동안 50mg/kg의 용량으로 SPA7012가 투여될 경우, I/R 손상 마우스에서 AST 및 ALT 수준이 모두 유의하게 감소한 것으로 나타났으며, 간의 종합 소견은 AST 및 ALT 수준과 일치한 것으로 확인되었다(도 8). 또한, H&E 염색을 통하여 대조군과 비교하여 I/R 손상된 간 조직에서 광범위한 간세포 괴사가 확인되었으나(도 8 및 도 9), SPA7012 처리된 마우스에서 괴사 영역은 I/R 그룹에 비해 유의하게 작았고, 상당한 영역이 정상적인 간 구조를 갖는 것으로 확인되었다.As a result, liver damage assessed by serum levels of AST and ALT was confirmed to be evident in mice receiving I/R compared to control mice (FIG. 7). However, when SPA7012 was administered at a dose of 50 mg/kg before and during I/R injury, both AST and ALT levels were found to be significantly reduced in I/R injured mice, and the liver composite findings were significantly different from those of AST and ALT levels. It was confirmed that they matched (Figure 8). In addition, extensive hepatocyte necrosis was confirmed in the I/R damaged liver tissue compared to the control group through H&E staining (Figures 8 and 9), but the necrotic area in SPA7012-treated mice was significantly smaller than that in the I/R group, and there was significant hepatocyte necrosis in the I/R damaged liver tissue. The area was confirmed to have normal liver structure.

또한, 간 I/R 손상 중 세포 사멸의 주요 원인은 괴사이지만, 손상 과정에서 세포 사멸에 따른 세포사가 관찰된다. TUNEL 양성 세포 사멸 세포의 수는 I/R 손상된 간 조직에서 현저하게 증가한 반면 TUNEL 양성 세포는 대조군에서 거의 관찰되지 않았다(도 10). 웨스턴 블롯팅 분석에 따르면, I/R 손상된 간에서 pro-apoptotic Bax와 cleaved caspase-3의 증가와 anti-apoptotic Bcl-2의 감소가 나타난 것으로 확인되었다(도 11). 그러나 마우스에 SPA7012를 처리했을 때 TUNEL 양성 세포의 현저한 감소와 세포 사멸 단백질의 발현이 변화된 것으로 확인되었다. In addition, the main cause of cell death during liver I/R injury is necrosis, but cell death due to apoptosis is observed during the injury process. The number of TUNEL-positive apoptotic cells was significantly increased in I/R damaged liver tissue, whereas TUNEL-positive cells were rarely observed in the control group (Figure 10). According to Western blotting analysis, it was confirmed that the I/R damaged liver showed an increase in pro-apoptotic Bax and cleaved caspase-3 and a decrease in anti-apoptotic Bcl-2 (Figure 11). However, when mice were treated with SPA7012, a significant decrease in TUNEL-positive cells and changes in the expression of apoptotic proteins were confirmed.

이와 같은 결과를 통하여, I/R 손상 시 SPA7012는 간세포 손상에 대한 보호 효과를 나타낼 수 있음이 확인되었다.Through these results, it was confirmed that SPA7012 can exert a protective effect against hepatocyte damage during I/R injury.

<3-2> I/R 손상이 있는 마우스에서 SPA7012의 산화 스트레스 감소 효과 확인<3-2> Confirmation of oxidative stress reduction effect of SPA7012 in mice with I/R injury

SPA7012가 산화 스트레스 감소를 통해 I/R 손상에 대한 보호 효과를 발휘하는지 여부를 확인하기 위하여, 간 조직에서 산화 스트레스 마커를 측정하였다.To determine whether SPA7012 exerts a protective effect against I/R damage through reducing oxidative stress, oxidative stress markers were measured in liver tissue.

그 결과, MDA(지질 과산화의 지표) 수준은 대조군과 비교하여 I/R 손상된 간 조직에서 현저하게 증가한 것으로 나타났다(도 12). 반면, I/R은 간 조직에서 항산화제 GSH 수준의 상당한 억제를 야기하였다(도 13). 4-HNE를 사용한 간 조직의 면역염색을 통하여 I/R 손상에 의한 지질 과산화의 증가를 확인하였다(도 14). 그러나 SPA7012로 마우스를 처리하면 I/R로 인한 이러한 변화가 역전된 것으로 나타났으며, MDA의 혈청 수준은 유의하게 낮았고, 간 GSH 수준은 유의하게 높았으며, 4-HNE-양성 세포는 I/R 군보다 SPA7012 군에서 유의하게 낮은 것으로 확인되었다.As a result, the level of MDA (an indicator of lipid peroxidation) was found to be significantly increased in I/R damaged liver tissue compared to the control group (FIG. 12). On the other hand, I/R caused significant inhibition of antioxidant GSH levels in liver tissue (Figure 13). An increase in lipid peroxidation due to I/R injury was confirmed through immunostaining of liver tissue using 4-HNE (Figure 14). However, treatment of mice with SPA7012 appeared to reverse these I/R-induced changes, with serum levels of MDA being significantly lower, liver GSH levels being significantly higher, and 4-HNE-positive cells increasing significantly after I/R. It was confirmed to be significantly lower in the SPA7012 group than in the group.

한편, 최근 PAK4가 Thr369에서 Nrf2를 인산화하여 세포질에서 핵 방출(nuclear export) 및 후속 프로테아좀 분해를 유발한다고 보고되었다. 따라서, SPA7012가 Nrf2의 세포내 국소화, 단백질 안정성 및 전사 활성에 영향을 미칠 수 있는지 여부를 조사하였다. Meanwhile, it was recently reported that PAK4 phosphorylates Nrf2 at Thr369, causing nuclear export from the cytoplasm and subsequent proteasome degradation. Therefore, we investigated whether SPA7012 could affect the subcellular localization, protein stability, and transcriptional activity of Nrf2.

그 결과, HEK293T 세포에서 Nrf2 및 PAK4의 과발현 후 Co-IP 분석에 따르면, SPA7012가 트레오닌 잔기에서 Nrf2의 PAK4 매개 인산화를 효과적으로 억제함하는 것으로 나타났다(도 15). 일관되게, SPA7012 처리에 의하여 I/R 손상된 간 조직에서 Nrf2의 핵 수준이 증가되었고(도 16), HEK293T 세포에서 Nrf2 전사 활성이 증가된 것으로 나타났다(도 17). qPCR 및 웨스턴 블롯팅 분석에 따르면, I/R 손상된 간 조직에서 SPA7012 처리에 의해 Nrf2의 향상된 전사 활성이 추가로 확인되었다(도 18 및 도 19).As a result, Co-IP analysis after overexpression of Nrf2 and PAK4 in HEK293T cells showed that SPA7012 effectively inhibited PAK4-mediated phosphorylation of Nrf2 at the threonine residue (FIG. 15). Consistently, the nuclear level of Nrf2 was increased in I/R injured liver tissue by SPA7012 treatment (Figure 16), and Nrf2 transcriptional activity was increased in HEK293T cells (Figure 17). According to qPCR and Western blotting analysis, enhanced transcriptional activity of Nrf2 was further confirmed by SPA7012 treatment in I/R damaged liver tissue (Figures 18 and 19).

이와 같은 결과를 통하여, SPA7012가 Nrf2 단백질을 안정화시키고 항산화 단백질 발현을 향상시켜 I/R 손상에 대한 간 보호 효과를 나타낸다는 것을 확인하였다.Through these results, it was confirmed that SPA7012 exhibits a hepatoprotective effect against I/R damage by stabilizing Nrf2 protein and improving antioxidant protein expression.

<3-3> I/R 손상이 있는 마우스에서 SPA7012의 염증 억제 효과 확인<3-3> Confirmation of the inflammation-inhibiting effect of SPA7012 in mice with I/R injury

간 I/R은 또한 간세포 사멸에 대한 반응으로, 간내 쿠퍼(Kupffer) 세포가 활성화되거나 면역 세포(단핵구 및 호중구)가 간 조직으로 동원되는 멸균 염증을 특징으로 한다. 따라서, 재관류 후 24시간 후 염증 정도를 평가하기 위해 간 조직을 면역염색하였다.Hepatic I/R is also characterized by sterile inflammation with activation of intrahepatic Kupffer cells or recruitment of immune cells (monocytes and neutrophils) into liver tissue in response to hepatocyte death. Therefore, liver tissue was immunostained to evaluate the degree of inflammation 24 hours after reperfusion.

그 결과, F4/80-양성 대식세포와 Ly6G-양성 호중구의 수는 모두 간 I/R 동안 현저하게 증가하여(도 20) 간에 명백한 염증이 확인되었다. 혈청 및 간 내 상응하는 mRNA의 전염증성 사이토카인/케모카인(TNF-α, IL-1βIL-6 및 CCL-2)의 수준도 I/R 손상에 의해 유의하게 증가하였다(도 21 및 도 22). 그러나, SPA7012로 마우스를 치료하면 마우스에서 염증 세포 침윤과 사이토카인 생성이 유의하게 감소한 것으로 나타났다. As a result, the numbers of both F4/80-positive macrophages and Ly6G-positive neutrophils increased significantly during liver I/R (FIG. 20), confirming obvious inflammation in the liver. The levels of pro-inflammatory cytokines/chemokines (TNF-α, IL-1βIL-6, and CCL-2) of the corresponding mRNAs in serum and liver were also significantly increased by I/R injury (Figures 21 and 22). However, treatment of mice with SPA7012 showed a significant reduction in inflammatory cell infiltration and cytokine production in mice.

또한, NF-κ는 염증성 사이토카인 생성과 밀접한 관련이 있기 때문에 Western blotting을 통해 NF-κ신호 전달 경로를 분석한 결과, SPA7012 처리가 대조군에 비해 IKKβ및 p65의 인산화 수준을 유의하게 감소시켰고 Iκα의 단백질 수준을 증가시킨 것으로 나타났다(도 23).In addition, because NF-κ is closely related to the production of inflammatory cytokines, we analyzed the NF-κ signaling pathway through Western blotting and found that SPA7012 treatment significantly reduced the phosphorylation levels of IKKβ and p65 compared to the control group and increased the level of Iκα. It was found that the protein level was increased (Figure 23).

이와 같은 결과를 통하여, SPA7012에 의한 염증 억제가 I/R 손상에 대한 보호에 추가적으로 기여함을 확인하였다.Through these results, it was confirmed that inhibition of inflammation by SPA7012 additionally contributes to protection against I/R damage.

<3-4> 간세포에서 SPA7012의 H/R로 인한 세포 사멸 및 염증 억제 효과 확인<3-4> Confirmation of H/R-induced cell death and inflammation inhibition effects of SPA7012 in hepatocytes

간 I/R 손상을 시뮬레이션하기 위하여, 시험관 내 H/R 유도 간세포 손상 모델을 추가로 확립하였으며(도 24), 세포 사멸 및 염증 억제 효과를 분석하였다.To simulate liver I/R injury, an in vitro H/R-induced hepatocyte injury model was further established (Figure 24), and the cell death and inflammation inhibition effects were analyzed.

그 결과, 1μM 미만 농도의 SPA7012는 세포 생존율에 거의 영향을 미치지 않았다(도 25). 한편, H/R에 대한 노출은 정상산소 그룹과 비교하여 1차 간세포에서 LDH 방출을 유의하게 증가시켰다. 그러나, 세포가 재산소화 손상에 노출되기 전 0.5μM가 아닌 1μM에서 SPA7012와 함께 배양되었을 때, 세포 손상은 H/R 단독에 비해 유의하게 감소한 것으로 나타났다(도 26). As a result, SPA7012 at a concentration of less than 1 μM had little effect on cell viability (Figure 25). Meanwhile, exposure to H/R significantly increased LDH release from primary hepatocytes compared to the normoxic group. However, when cells were incubated with SPA7012 at 1 μM rather than 0.5 μM before exposure to reoxygenation damage, cell damage appeared to be significantly reduced compared to H/R alone (Figure 26).

이러한 결과를 바탕으로 1μM의 SPA7012를 선택하여 실험을 수행한 결과, I/R 손상과 유사하게, H/R 손상은 또한 1차 간세포에서 세포 사멸 세포사(도 27 및 도 28) 및 사이토카인 방출이 증가되었으나(도 29 및 도 30), SPA7012의 처리에 따라 이러한 반응이 감소된 것으로 확인되었다.Based on these results, 1 μM of SPA7012 was selected and an experiment was performed. As a result, similar to I/R injury, H/R injury also resulted in apoptotic cell death (Figures 27 and 28) and cytokine release in primary hepatocytes. However, it was confirmed that this response was decreased upon treatment with SPA7012 (Figures 29 and 30).

실시예 4. 항증식 실험에 따른 암 세포주 억제 효과 확인Example 4. Confirmation of cancer cell line inhibition effect according to anti-proliferation experiment

PAk4 억제 활성을 나타내는 화합물 1 내지 9가 암 세포주 억제 효과를 나타내는지 확인하였다.It was confirmed whether compounds 1 to 9, which exhibit PAk4 inhibitory activity, exhibit an inhibitory effect on cancer cell lines.

구체적으로, 항증식 활성 실험을 통하여, MDA-MB-231, U-87MG, PC-3, BxPC-3, NCI-H23 및 HCT-15 암 세포주에 대한 각 화합물의 GI50 값을 평가하였다.Specifically, through antiproliferative activity experiments, the GI 50 value of each compound was evaluated against MDA-MB-231, U-87MG, PC-3, BxPC-3, NCI-H23, and HCT-15 cancer cell lines.

화합물compound MDA-MB-231MDA-MB-231 U-87MGU-87MG PC-3PC-3 BxPC-3BxPC-3 NCI-H23NCI-H23 HCT-15HCT-15 화합물 1Compound 1 18.618.6 16.816.8 16.316.3 14.714.7 19.119.1 17.017.0 화합물 2compound 2 13.513.5 10.610.6 13.413.4 13.013.0 11.711.7 15.215.2 화합물 3Compound 3 >50>50 >50>50 >50>50 >50>50 >50>50 >50>50 화합물 4Compound 4 >50>50 >50>50 >50>50 >50>50 >50>50 >50>50 화합물 5Compound 5 >50>50 >50>50 >50>50 >50>50 >50>50 >50>50 화합물 6Compound 6 >50>50 >50>50 >50>50 >50>50 >50>50 >50>50 화합물 7Compound 7 19.619.6 18.418.4 20.720.7 17.317.3 15.515.5 17.117.1 화합물 8Compound 8 41.641.6 38.138.1 38.838.8 40.440.4 37.237.2 42.442.4 화합물 9Compound 9 20.320.3 14.914.9 19.219.2 19.019.0 13.613.6 17.517.5 GNE-2861GNE-2861 18.018.0 17.617.6 19.419.4 15.515.5 16.916.9 18.718.7

그 결과, 화합물 1 내지 9의 암세포 성장 억제 활성은 모든 암세포주에서 확인되었으며(표 6), 억제 활성은 화합물의 logP 값과 밀접한 관련이 있는 것으로 나타났다. 특히, 화합물 2 및 화합물 7이 GNE-2861 대비 유사하거나 개선된 GI50 값을 나타내는 것으로 확인되었다.As a result, the cancer cell growth inhibitory activity of compounds 1 to 9 was confirmed in all cancer cell lines (Table 6), and the inhibitory activity appeared to be closely related to the logP value of the compounds. In particular, Compound 2 and Compound 7 were confirmed to have similar or improved GI 50 values compared to GNE-2861.

이제까지 본 발명에 대하여 그 실시예들을 중심으로 살펴보았다. 본 발명이 속하는 기술 분야에서 통상의 지식을 가진 자는 본 발명이 본 발명의 본질적인 특성에서 벗어나지 않는 범위에서 변형된 형태로 구현될 수 있음을 이해할 수 있을 것이다. 그러므로 개시된 실시예들은 한정적인 관점이 아니라 설명적인 관점에서 고려되어야 한다. 본 발명의 범위는 전술한 설명이 아니라 청구범위에 나타나 있으며, 그와 동등한 범위 내에 있는 모든 차이점은 본 발명에 포함된 것으로 해석되어야 할 것이다.So far, the present invention has been examined focusing on its embodiments. A person skilled in the art to which the present invention pertains will understand that the present invention may be implemented in a modified form without departing from the essential characteristics of the present invention. Therefore, the disclosed embodiments should be considered from an illustrative rather than a restrictive perspective. The scope of the present invention is indicated in the claims rather than the foregoing description, and all differences within the equivalent scope should be construed as being included in the present invention.

<110> INDUSTRIAL COOPERATION FOUNDATION JEONBUK NATIONAL UNIVERSITY <120> Pyrazolo[3,4-d]pyrimidine Derivative, and Composition for Preventing or Treating Liver Injury and Composition for Protecting Liver comprising the same <130> DHP22-132 <160> 20 <170> KoPatentIn 3.0 <210> 1 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Tnfa_forward <400> 1 agggtctggg ccatagaact 20 <210> 2 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Tnfa_reverse <400> 2 ccaccacgct cttctgtcta c 21 <210> 3 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Il1b_forward <400> 3 ggtcaaaggt ttggaagcag 20 <210> 4 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Il1b_reverse <400> 4 tgtgaaatgc caccttttga 20 <210> 5 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Il6_forward <400> 5 agggtctggg ccatagaact 20 <210> 6 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Il6_reverse <400> 6 ccaccacgct cttctgtcta c 21 <210> 7 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Icam1_forward <400> 7 aacagttcac ctgcacggac 20 <210> 8 <211> 19 <212> DNA <213> Artificial Sequence <220> <223> Icam1_reverse <400> 8 gtcaccgttg tgatccctg 19 <210> 9 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Cxcl1_forward <400> 9 aatgagctgc gctgtcagtg 20 <210> 10 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Cxcl1_reverse <400> 10 tgagggcaac accttcaagc 20 <210> 11 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Ccl2_forward <400> 11 accgacaaca ggaagtggag 20 <210> 12 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Ccl2_reverse <400> 12 tggacgtttc acacagtggt 20 <210> 13 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Ho1_forward <400> 13 aggtacacat ccaagccgag a 21 <210> 14 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Ho1_reverse <400> 14 catcaccagc ttaaagcctt ct 22 <210> 15 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Nqo1_forward <400> 15 aggatgggag gtactcgaat c 21 <210> 16 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Nqo1_reverse <400> 16 tgctagagat gactcggaag g 21 <210> 17 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nrf2_forward <400> 17 ctttagtcag cgacagaagg ac 22 <210> 18 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nrf2_reverse <400> 18 aggcatcttg tttgggaatg tg 22 <210> 19 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Gapdh_forward <400> 19 cgtcccgtag acaaaatggt 20 <210> 20 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Gapdh_reverse <400> 20 ttgatggcaa caatctccac 20 <110> INDUSTRIAL COOPERATION FOUNDATION JEONBUK NATIONAL UNIVERSITY <120> Pyrazolo[3,4-d]pyrimidine Derivative, and Composition for Preventing or Treating Liver Injury and Composition for Protecting Liver comprising the same <130>DHP22-132 <160> 20 <170> KoPatentIn 3.0 <210> 1 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Tnfa_forward <400> 1 agggtctggg ccatagaact 20 <210> 2 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Tnfa_reverse <400> 2 ccaccacgct cttctgtcta c 21 <210> 3 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Il1b_forward <400> 3 ggtcaaaggt ttggaagcag 20 <210> 4 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Il1b_reverse <400> 4 tgtgaaatgc caccttttga 20 <210> 5 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Il6_forward <400> 5 agggtctggg ccatagaact 20 <210> 6 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Il6_reverse <400> 6 ccaccacgct cttctgtcta c 21 <210> 7 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Icam1_forward <400> 7 aacagttcac ctgcacggac 20 <210> 8 <211> 19 <212> DNA <213> Artificial Sequence <220> <223> Icam1_reverse <400> 8 gtcaccgttg tgatccctg 19 <210> 9 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Cxcl1_forward <400> 9 aatgagctgc gctgtcagtg 20 <210> 10 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Cxcl1_reverse <400> 10 tgagggcaac accttcaagc 20 <210> 11 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Ccl2_forward <400> 11 accgacaaca ggaagtggag 20 <210> 12 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Ccl2_reverse <400> 12 tggacgtttc acacagtggt 20 <210> 13 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Ho1_forward <400> 13 aggtacacat ccaagccgag a 21 <210> 14 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Ho1_reverse <400> 14 catcaccagc ttaaagcctt ct 22 <210> 15 <211> 21 <212> DNA <213> Artificial Sequence <220> <223>Nqo1_forward <400> 15 aggatggggag gtactcgaat c 21 <210> 16 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Nqo1_reverse <400> 16 tgctagagat gactcggaag g 21 <210> 17 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nrf2_forward <400> 17 ctttagtcag cgacagaagg ac 22 <210> 18 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nrf2_reverse <400> 18 aggcatcttg tttgggaatg tg 22 <210> 19 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Gapdh_forward <400> 19 cgtcccgtag acaaaatggt 20 <210> 20 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> Gapdh_reverse <400> 20 ttgatggcaa caatctccac 20

Claims (11)

하기 화학식 1로 표시되는 화합물, 이의 약학적으로 허용되는 염, 이의 수화물, 이의 용매화물 및 이의 이성질체로 이루어진 군으로부터 선택되는 화합물:
[화학식 1]

상기 화학식 1에서,
R1 및 R2는 서로 같거나, 서로 다르거나, 또는 동일한 고리(A)상에 위치하는 것으로써, C1-6알킬, C1-6할로알킬, -(CH2)m-C1-6알콕시(이때, m은 1 내지 6의 정수), C3-8사이클로알킬, C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고,
상기 고리(A)는 C3-8사이클로알킬, C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고,
R3는 -CH2-R7R8 이거나, 또는 R3는 R4 및 R5와 동일한 방향족 고리(B)에 위치할 수 있고,
상기 방향족 고리(B)는 C6-12아릴로 이루어진 군으로부터 선택되는 치환기이고,
R5 및 R6는 서로 같거나 또는 서로 다른 것으로써, C 또는 N이고,
상기 R7은 H, C1-6알킬, C2-6알케닐 또는 C2-6알키닐로 이루어진 군으로부터 선택되는 치환기이고,
상기 R8은 H, O 또는 N으로부터 선택된 헤테로원자가 1 내지 2개 포함된 C3-8헤테로고리 치환 C1-6 알킬로 이루어진 군으로부터 선택되는 치환기이다.
A compound selected from the group consisting of a compound represented by the following formula (1), a pharmaceutically acceptable salt thereof, a hydrate thereof, a solvate thereof, and an isomer thereof:
[Formula 1]

In Formula 1,
R 1 and R 2 are the same, different from each other, or are located on the same ring (A), and are C 1-6 alkyl, C 1-6 haloalkyl, -(CH 2 ) m -C 1- 6 is a substituent selected from the group consisting of alkoxy (where m is an integer of 1 to 6), C 3-8 cycloalkyl, and C 6-12 aryl,
The ring (A) is a substituent selected from the group consisting of C 3-8 cycloalkyl and C 6-12 aryl,
R 3 may be -CH 2 -R 7 R 8 , or R 3 may be located in the same aromatic ring (B) as R 4 and R 5 ,
The aromatic ring (B) is a substituent selected from the group consisting of C 6-12 aryl,
R 5 and R 6 are the same or different from each other and are C or N,
R 7 is a substituent selected from the group consisting of H, C 1-6 alkyl, C 2-6 alkenyl, or C 2-6 alkynyl,
The R 8 is a substituent selected from the group consisting of C 3-8 heterocyclic substituted C 1-6 alkyl containing 1 to 2 heteroatoms selected from H, O or N.
제 1 항에 있어서,
4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-메틸부틴-2-올;
3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐에틴일사이클로헥산-1-올;
4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-메틸부틴일-1,2-다이올;
4-(3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐)-2-페닐부틴-2-올;
3-[4-아미노-3-(3-몰포린-1-일프로필)피라졸로[3,4-d]피리미딘-1-일]페닐에틴일)사이클로헥산올;
3-[4-아미노-3-(3-피페리딘-1-일프로필)피라졸로[3,4-d]피리미딘-1-일]페닐에틴일)사이클로헥산올;
4-(3-(1-아미노피리도[4,3-b]인돌-5-일)페닐)-2-메틸부틴-2-올;
3-(1-아미노피리도[4,3-b]인돌-5-일)페닐에틴일사이클로엑산-1-올; 및
4-(3-(1-아미노피리도[4,3-b]인돌-5-일)페닐)-2-메틸부틴-1,2-다이올
로 이루어진 군으로부터 선택되는 화합물.
According to claim 1,
4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutyn-2-ol;
3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol;
4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-methylbutynyl-1,2-diol;
4-(3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenyl)-2-phenylbutyn-2-ol;
3-[4-amino-3-(3-morpholin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol;
3-[4-amino-3-(3-piperidin-1-ylpropyl)pyrazolo[3,4- d ]pyrimidin-1-yl]phenylethynyl)cyclohexanol;
4-(3-(1-aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyn-2-ol;
3-(1-aminopyrido[4,3- b ]indol-5-yl)phenylethynylcycloexan-1-ol; and
4-(3-(1-aminopyrido[4,3- b ]indol-5-yl)phenyl)-2-methylbutyne-1,2-diol
A compound selected from the group consisting of.
제 1 항에 있어서,
3-(4-아미노-3-메틸피라졸로[3,4-d]피리미딘-1-일)페닐에틴일사이클로헥산-1-올인 것인 화합물.
According to claim 1,
A compound that is 3-(4-amino-3-methylpyrazolo[3,4- d ]pyrimidin-1-yl)phenylethynylcyclohexan-1-ol.
제 1 항의 화합물을 포함하는 PAK4 발현 증가 질환 예방 또는 치료용 약학적 조성물.
A pharmaceutical composition for preventing or treating diseases with increased PAK4 expression, comprising the compound of claim 1.
제 4 항에 있어서, 상기 PAK4 발현 증가 질환은 암, 신경계 질환, 간 손상, 간 염증, 멜라닌 색소 침착으로 이루어진 군으로부터 선택되는 하나 이상의 질환인 것인 약학적 조성물.
The pharmaceutical composition according to claim 4, wherein the disease with increased PAK4 expression is one or more diseases selected from the group consisting of cancer, neurological disease, liver damage, liver inflammation, and melanin pigmentation.
제 5 항에 있어서, 상기 암은 위암, 폐암, 간암, 대장암, 소장암, 췌장암, 뇌암, 뼈암, 흑색종, 유방암, 경화성선증, 자궁암, 자궁경부암, 두경부암, 식도암, 갑상선암, 부갑상선암, 신장암, 육종, 전립선암, 요도암, 방광암, 혈액암, 림프 종, 건선, 또는 섬유선종으로 이루어진 군으로부터 선택되는 하나 이상의 암인 것인 약학적 조성물.
The method of claim 5, wherein the cancer is stomach cancer, lung cancer, liver cancer, colon cancer, small intestine cancer, pancreatic cancer, brain cancer, bone cancer, melanoma, breast cancer, sclerosing gonadosis, uterine cancer, cervical cancer, head and neck cancer, esophageal cancer, thyroid cancer, parathyroid cancer, A pharmaceutical composition, wherein the pharmaceutical composition is one or more cancers selected from the group consisting of kidney cancer, sarcoma, prostate cancer, urethral cancer, bladder cancer, hematological cancer, lymphoma, psoriasis, or fibroadenoma.
제 5 항에 있어서, 상기 신경계 질환은 퇴행성뇌질환인 것인 약학적 조성물.
The pharmaceutical composition according to claim 5, wherein the neurological disease is a degenerative brain disease.
제 7 항에 있어서, 상기 퇴행성뇌질환은 알츠하이머병, 파킨슨병, 진행성 핵상마비, 다계통 위축증, 감람핵-뇌교-소뇌 위축증 (OPCA), 샤이-드래거 증후군, 선조체-흑질 퇴행증, 헌팅톤병, 근위축성 측색 경화증 (ALS), 본태성 진전증, 피질-기저핵 퇴행증, 미만성 루이 소체 질환), 파킨스-ALS-치매 복합증, 픽병, 뇌허혈 및 뇌경색으로 이루어진 군으로부터 선택되는 하나 이상의 질환인 것인 약학적 조성물.
The method of claim 7, wherein the degenerative brain disease is Alzheimer's disease, Parkinson's disease, progressive supranuclear palsy, multiple system atrophy, olive nucleus-pontine-cerebellar atrophy (OPCA), Shay-Drager syndrome, striatal-substantia nigra degeneration, and Huntington's disease. , amyotrophic lateral sclerosis (ALS), essential tremor, cortico-basal gangrene, diffuse Lewy body disease), Parkinson-ALS-dementia complex, Pick's disease, cerebral ischemia, and cerebral infarction. Phosphorus pharmaceutical composition.
제 5 항에 있어서, 상기 간 손상은 염증에 의한 손상, 산화적 손상, 알코올성 손상 및 허혈성 재관류 손상으로 이루어진 군으로부터 선택되는 하나 이상의 손상인 것인 약학적 조성물.
The pharmaceutical composition according to claim 5, wherein the liver damage is at least one damage selected from the group consisting of inflammatory damage, oxidative damage, alcoholic damage, and ischemia-reperfusion damage.
제 1 항의 화합물을 포함하는 간 기능 보호용 조성물.
A composition for protecting liver function comprising the compound of claim 1.
제 1 항의 화합물을 포함하는 간 기능 보호용 건강기능식품.A health functional food for protecting liver function containing the compound of claim 1.
KR1020220036547A 2022-03-24 2022-03-24 Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same KR20230139388A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020220036547A KR20230139388A (en) 2022-03-24 2022-03-24 Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020220036547A KR20230139388A (en) 2022-03-24 2022-03-24 Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same

Publications (1)

Publication Number Publication Date
KR20230139388A true KR20230139388A (en) 2023-10-05

Family

ID=88294180

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020220036547A KR20230139388A (en) 2022-03-24 2022-03-24 Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same

Country Status (1)

Country Link
KR (1) KR20230139388A (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101706017B1 (en) 2015-07-31 2017-02-13 재단법인 전남생물산업진흥원 Hepatoprotective composition comprising extracts of fruits of stauntonia hexaphylla for preventing alcoholic liver injury

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101706017B1 (en) 2015-07-31 2017-02-13 재단법인 전남생물산업진흥원 Hepatoprotective composition comprising extracts of fruits of stauntonia hexaphylla for preventing alcoholic liver injury

Similar Documents

Publication Publication Date Title
US8309550B2 (en) Kinase inhibitors and their use as pharmaceutical agents
KR100820543B1 (en) 2-Amino-3-Alkyl-Pyrimidone Derivatives as GSK3.Beta. Inhibitors
JP5357763B2 (en) Imidazo [1,2-b] pyridazine derivatives and pyrazolo [1,5-a] pyridazine derivatives and their use as protein kinase inhibitors
US8518953B2 (en) Aminopyrimidines useful as inhibitors of protein kinases
WO2020156242A1 (en) Shp2 inhibitor and application thereof
RU2578608C2 (en) Novel imidazooxazine compound or salt thereof
US20100184790A1 (en) Pyrrolo[2,3-b]pyridine compounds, azaindole compounds used for synthesizing said pyrrolo[2,3-b]pyridine compounds, methods for the production thereof, and uses thereof
US20090325968A1 (en) Compositions Useful as Inhibitors of Protein Kinases
EA018964B1 (en) PYRIDO[2,3-d]PYRIMIDIN-7-ONE COMPOUNDS AS INHIBITORS OF PI3K-ALPHA FOR THE TREATMENT OF CANCER
TW200825079A (en) Antidepressant, neuroprotectant, amyloid β deposition inhibitor or age retardant containing heterocyclic compound having specific structure
TW201908317A (en) Treating cancer with a combination of PARP inhibitors, temozolomide and/or radiation therapy
HUE027577T2 (en) Morpholinylbenzotriazines for use in cancer therapy
US8304421B2 (en) Indole compounds and their use as radiation sensitizing agents and chemotherapeutic agents
US11858938B2 (en) Imidazo-fused heterocycles and uses thereof
US20230122909A1 (en) Compounds, pharmaceutical compositions, and methods of preparing compounds and of their use
US20230142913A1 (en) Tricyclic heteroarenes, pharmaceutical compositions containing the same, and methods of using the same
Dou et al. Rational modification, synthesis and biological evaluation of 3, 4-dihydroquinoxalin-2 (1H)-one derivatives as potent and selective c-Jun N-terminal kinase 3 (JNK3) inhibitors
TW200409775A (en) Novel pyrazolopyridines as cyclin dependent kinase inhibitors
KR102468670B1 (en) Inhibition of OLIG2 activity
US20150306070A1 (en) Use of maleimide derivatives for preventing and treating leukemia
US20180312502A1 (en) N1-And N7-Substituted Sibiriline Derivatives And Their Use As Inhibitor Of Cellular Necroptosis
KR20230139388A (en) Pyrazolo[3,4-d]pyrimidine-based p21-activated kinase 4 (PAK4) inhibitor and pharmaceutical composition comprising the same
CA3214900A1 (en) Carboxamide pyrolopyrazine and pyridine compounds useful as inhibitors of myt1 and use thereof in the treatment of cancer
Mao et al. Targeting p21-activated kinase 4 (PAK4) with pyrazolo [3, 4-d] pyrimidine derivative SPA7012 attenuates hepatic ischaemia-reperfusion injury in mice
JP2002526450A (en) CHK1 kinase inhibitor

Legal Events

Date Code Title Description
E902 Notification of reason for refusal