KR102129367B1 - Compound for inducing the degradation of cereblon protein, preparation method thereof and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient - Google Patents

Compound for inducing the degradation of cereblon protein, preparation method thereof and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient Download PDF

Info

Publication number
KR102129367B1
KR102129367B1 KR1020180119161A KR20180119161A KR102129367B1 KR 102129367 B1 KR102129367 B1 KR 102129367B1 KR 1020180119161 A KR1020180119161 A KR 1020180119161A KR 20180119161 A KR20180119161 A KR 20180119161A KR 102129367 B1 KR102129367 B1 KR 102129367B1
Authority
KR
South Korea
Prior art keywords
dioxopiperidin
hydroxy
linker
methylthiazol
benzyl
Prior art date
Application number
KR1020180119161A
Other languages
Korean (ko)
Other versions
KR20190044499A (en
Inventor
황종연
하재두
조성윤
김필호
박병철
김선홍
김정훈
박성구
Original Assignee
한국화학연구원
한국생명공학연구원
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 한국화학연구원, 한국생명공학연구원 filed Critical 한국화학연구원
Publication of KR20190044499A publication Critical patent/KR20190044499A/en
Application granted granted Critical
Publication of KR102129367B1 publication Critical patent/KR102129367B1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/4035Isoindoles, e.g. phthalimide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

본 발명은 세레브론 단백질의 분해 유도 화합물, 이의 제조방법 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물에 관한 것으로, 본 발명에 따른 A-Linker-A 또는 A-Linker-B로 표시되는 신규 화합물은 세레브론을 우수하게 저해하거나 분해시킬 수 있는 바, 이를 유효성분으로 함유하여 세레브론 관련 질환, 바람직하게 세레브론을 저해 또는 분해하는 것으로부터 치료, 예방 또는 개선과 같은 효과가 달성될 수 있는 질환, 예를 들어, 암, 자가면역질환, 또는 대사질환의 예방 또는 치료용 약학적 조성물로서 유용한 효과가 있다.The present invention relates to a pharmaceutical composition for the prevention or treatment of cancer containing a compound for inducing the decomposition of a cerebron protein, a preparation method thereof, and an active ingredient thereof, to A-Linker-A or A-Linker-B according to the present invention The new compounds displayed can inhibit or decompose the cerebrone excellently, and contain it as an active ingredient to achieve an effect such as treatment, prevention or improvement from inhibiting or decomposing a cerebrone-related disease, preferably cerebrone. It has a useful effect as a pharmaceutical composition for the prevention or treatment of diseases that can be, for example, cancer, autoimmune diseases, or metabolic diseases.

Description

세레브론 단백질의 분해 유도 화합물, 이의 제조방법 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물{Compound for inducing the degradation of cereblon protein, preparation method thereof and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient}Compound for inducing the degradation of cereblon protein, preparation method thereof and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient}

본 발명은 세레브론 단백질의 분해 유도 화합물, 이의 제조방법 및 이를 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing or treating cancer, which is a compound for inducing the degradation of a celebrative protein, a method for preparing the same, and an active ingredient thereof.

세포는 단백질-항상성(protein homeostasis)을 유지하기 위해 유비퀴틴-프로테아좀 시스템(Ubiquitin-proteasome system, UPS)을 이용하여 표적 단백질을 분해한다. 이는 E1, E2, E3 리가제라 불리는 단백질이 76개의 아미노산으로 이루어진 유비퀴틴(Ub)을, 분해하고자 하는 표적단백질(substrate)에 전달하여 다중유비퀴틴화(polyubiquitination) 시키면, 26S 프로테아좀(proteasome)이 이를 인지하여 표적 단백질을 분해하는 과정을 통해서 이루어진다. 리가제 중 E3 유비퀴틴 리가제는 600여종 이상 알려져 있으며, 리가제 종류에 따라 분해되는 단백질이 다르다고 알려져 있다.Cells degrade target proteins using the Ubiquitin-proteasome system (UPS) to maintain protein homeostasis. This is when the protein called E1, E2, E3 ligase transfers ubiquitin (Ub) composed of 76 amino acids to the target protein to be degraded, and polyubiquitination results in 26S proteasome. It is achieved through the process of cognizing and degrading target proteins. Among the ligase, more than 600 kinds of E3 ubiquitin ligase are known, and it is known that different proteins are decomposed according to the type of ligase.

한편, E3 유비퀴틴 리가제 중 세레브론(CRBN, Cereblon)은 면역조절약물(Immunomodulatory drug, IMiD)로 알려진 탈리도마이드, 레날리도마이드, 포말리도마이드 등이 결합하면, 전사요인(transcription factor)인 이카로스(ikaros), 에이오로스(aiolos)를 분해 시켜 우수한 항암 효과를 보일 뿐만 아니라, 루푸스를 포함하는 자가면역질환 질환에 효과가 있다. 이에, 세레브론 자체를 표적하는 면역조절약물 개발이 활발히 이루어지고 있다.On the other hand, of E3 ubiquitin ligase, cereblon (CRBN, Cereblon), when thalidomide, lenalidomide, pomalidomide, etc., known as immunomodulatory drugs (IMiD), are combined, Ikaros (transcription factor) ikaros), Aioros (aiolos) decomposes to show excellent anti-cancer effects, as well as effective in autoimmune disease diseases including lupus. Accordingly, the development of an immunomodulatory drug targeting Celebron itself has been actively conducted.

또 다른 한편, 세레브론은 특정 단백질의 분해에 관여하는 E3 유비퀴틴 리가제 역할외에도, 광주과학기술원 박철승 교수연구팀은 CRBN이 AMPK의 활성을 억제함을 확인하였고, CRBN을 억제하였을 경우에는, 에너지센서인 AMPK가 활성화됨을 확인한 바, 이러한 CRBN 억제 유도로부터 비만, 지방간, 당뇨병 등 대사질환의 예방 또는 치료 가능성을 제시하였다.On the other hand, in addition to the role of E3 ubiquitin ligase, which is involved in the decomposition of specific proteins, Celebron, Professor Chul-Seung Park of Gwangju Institute of Science and Technology confirmed that CRBN inhibits AMPK activity, and when CRBN is inhibited, it is an energy sensor. When it was confirmed that AMPK was activated, the possibility of preventing or treating metabolic diseases such as obesity, fatty liver and diabetes was suggested from the induction of CRBN inhibition.

나아가, 광주과학기술원 박성규 교수 연구팀은 면역세포(T-세포)에 CRBN이 많이 발현되어 있음을 확인하였고, CRBN이 결핍되면 T 세포에서 칼륨 이온채널의 발현이 증가하고, 이를 통해 T 세포가 활성화되는 것을 확인하였으며, 이와 같은 후성적 유전자 발현 조절을 통해 항원에 대한 T세포의 민감도를 조절 할 수 있다는 연구 결과를 보고하였다. 이와 같은 연구 결과로부터, CRBN 결핍의 유도는 T세포에 과발현 되어 있는 CRBN의 양을 낮추어 T세포의 활성을 증가시키는 효과를 유도됨을 알 수 있고, 이러한 효과를 야기할 수 있는 CRBN 결핍 유도로부터 항암면역치료제 개발 가능성을 제시하였다.Furthermore, the research team of Prof. Seong-Kyu Park of Gwangju Institute of Science and Technology confirmed that CRBN is highly expressed in immune cells (T-cells), and when CRBN is deficient, the expression of potassium ion channels in T cells increases, thereby activating T cells. The results of this study were reported to control the sensitivity of T cells to antigens by regulating epigenetic gene expression. From the results of this study, it can be seen that induction of CRBN deficiency leads to an effect of increasing the activity of T cells by lowering the amount of CRBN overexpressed in T cells, and anti-cancer immunity from induction of CRBN deficiency that can cause such effects. It suggested the possibility of developing a therapeutic agent.

상술된 E3 유비퀴틴 리가제인 CRBN과 관련한 배경기술로부터 알 수 있듯이, 세레브론의 억제 또는 결핍을 유도한다면, 암, 자가면역질환, 대사질환, 등에 유용한 효과가 달성될 수 있다.As can be seen from the background related to the above-mentioned E3 ubiquitin ligase CRBN, if induction or inhibition of cerebron is induced, useful effects can be achieved in cancer, autoimmune diseases, metabolic diseases, and the like.

그러나, 아직까지 CRBN을 표적한 저분자 약물 개발이 전무하고, 또한 기존의 CRBN 억제제는 목적하는 수준의 약리 효과가 달성되지 못하고 있다. 특히, 기존의 항암 표적 치료제는 표적의 신호 전달을 저해하기 위해서는 표적에 대한 높은 저해력이 요구되며, 치료효과를 보이기 위해서는 많은 양을 오랜 기간 투여해야 하는 단점을 가지고 있고, 또한 약물에 대한 내성이 빠르게 나타나, 투여 후 1년 이내에 약물 내성이 발생하여 약물의 효과를 낮추게 되는 문제점이 있다. 또한, 세포독성 치료제는 암이 발생한 표적에 직접 작용할 수 없어, 상기 항암 표적 치료제와 비교하여 부작용 발생이 크고, 치료효가 적은 문제가 있다.However, there has been no development of a low-molecular drug targeting CRBN, and the conventional CRBN inhibitor has not achieved the desired pharmacological effect. In particular, the existing anti-cancer targeted therapeutics require a high inhibitory power to the target in order to inhibit the signal transmission of the target, and have a disadvantage in that a large amount must be administered for a long period of time in order to show a therapeutic effect, and also have resistance to drugs. It appears quickly, and there is a problem in that drug resistance occurs within a year after administration, thereby lowering the effect of the drug. In addition, since the cytotoxic therapeutic agent cannot directly act on the target in which cancer has occurred, there is a problem in that side effects are large and the therapeutic effect is small compared to the anticancer target therapeutic agent.

따라서, 항암 표적 치료제, 세포독성 치료제 보다 우수한 치료효과가 달성되고, 약물 내성 부작용 등의 문제점을 극복할 수 있는 새로운 기술 개발이 요구되며, 세레브론을 목적하는 수준으로 저해 또는 결핍시켜, 목적하는 수준으로 약리효과가 달성될 수 있는 치료제 개발이 요구된다.Therefore, a better therapeutic effect is achieved than an anti-cancer targeted therapeutic agent and a cytotoxic therapeutic agent, and new technology development is needed to overcome the problems such as drug resistance side effects. Therefore, it is required to develop a therapeutic agent that can achieve a pharmacological effect.

상술된 배경에서, 최근 예일대학교의 연구팀(Craig Crews)이 단백질 분해 유도제(Proteolysis Targeting Chimera, PROTAC)기술을 개발하였다.Against the background described above, a research team at Yale University (Craig Crews) recently developed a proteolysis targeting chimera (PROTAC) technology.

PROTAC은 특정 단백질에 결합하는 저해제(inhibitor) 또는 결합인자(binder) 화합물과 E3 유비퀴틴 리가아제에 특징적으로 결합하는 리간드 화합물을 다양한 종류의 링커로 연결하여 제조되는 신규 치료제로서, PROTAC 물질은 질병의 원인이 되는 표적 단백질만을 타깃하여 분해하기 때문에 약물에 대한 내성발생이 낮고, 표적 단백질에 결합만 가능하다면, PROTAC 자체의 대상 단백질에 대한 높은 저해활성 없이도, 활성표적 단백질 분해로부터 충분한 치료효과가 달성되는 바, 기존 약물로는 저해효과가 달성이 어려웠던 표적(undruggable target)에도 적용이 가능한 바, 신규한 메카니즘의 질환 치료기술로 각광받고 있다(특허문헌 1).PROTAC is a novel therapeutic agent that is produced by linking an inhibitor or binder compound that binds to a specific protein and a ligand compound that specifically binds E3 ubiquitin ligase with various types of linkers, and PROTAC substances cause disease Since the target protein is degraded by targeting only, the occurrence of resistance to the drug is low, and if binding to the target protein is possible, sufficient therapeutic effect is achieved from the degradation of the active target protein, without high inhibitory activity on the target protein of PROTAC itself , As it can be applied to targets that have been difficult to achieve with inhibitory effects (undruggable target) with existing drugs, it has been spotlighted as a novel mechanism of disease treatment technology (Patent Document 1).

이에, 본 발명자들은 CRBN에 결합하는 결합 화합물(binder)인 탈리도마이드 및 이의 유도체와 E3 리가제, 특히 VHL 리간드 또는 CRBN 리간드(탈리도마이드 유도체)를 다양한 링커를 통해 연결하여 신규 CRBN-PROTAC 물질을 개발하였고, 이로부터 성공적으로 CRBN이 분해되는 것을 확인 하였으며, 향후 CRBN 분해제로의 활용 뿐만 아니라, CRBN 관련 질환, 바람직하게 대사질환, 자가면역질환 또는 암의 예방 또는 치료용 약학적 조성물 또는 자가면역질환 예방 또는 치료용 약학적 조성물로 유용할 수 있음을 규명하여 본 발명을 완성하였다.Accordingly, the present inventors have developed a new CRBN-PROTAC material by connecting thalidomide, a binding compound that binds to CRBN, and its derivatives and E3 ligase, in particular VHL ligand or CRBN ligand (thalidomide derivative), through various linkers, From this, it was confirmed that CRBN is successfully degraded, and in the future, it is not only utilized as a CRBN decomposition agent, but also a pharmaceutical composition or autoimmune disease prevention or treatment for the prevention or treatment of CRBN-related diseases, preferably metabolic diseases, autoimmune diseases or cancer. It was found that it can be useful as a pharmaceutical composition to complete the present invention.

WO 2013/106643 A2WO 2013/106643 A2

본 발명의 목적은 세레브론(CRBN) 분해하는 화합물을 제공하는 것이다.An object of the present invention is to provide a compound that degrades cerebrone (CRBN).

본 발명의 다른 목적은 상기 화합물의 제조방법을 제공하는 것이다.Another object of the present invention is to provide a method for preparing the compound.

본 발명의 또 다른 목적은 상기 화합물을 유효성분으로 함유하는 세레브론 관련 질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of cerebron related diseases containing the compound as an active ingredient.

본 발명의 다른 목적은 상기 화합물을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for preventing or treating cancer containing the compound as an active ingredient.

본 발명의 또 다른 목적은 상기 화합물을 유효성분으로 함유하는 자가면역질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of autoimmune diseases containing the compound as an active ingredient.

본 발명의 다른 목적은 상기 화합물을 유효성분으로 함유하는 대사질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for the prevention or treatment of metabolic diseases containing the compound as an active ingredient.

본 발명의 또 다른 목적은 상기 화합물을 유효성분으로 함유하는 세레브론 관련 질환의 예방 또는 개선용 건강기능식품을 제공하는 것이다.Another object of the present invention is to provide a health functional food for preventing or improving cerebron-related diseases containing the compound as an active ingredient.

상기 목적을 해결하기 위해,To solve the above object,

본 발명은 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염을 제공한다:The present invention provides a compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:

상기 A는 세레브론(Cerebron) E3 유비퀴틴 리가아제 결합 모이어티(moiety)이고;A is Cerebron E3 ubiquitin ligase binding moiety;

상기 Linker는 -(CH2)-, -(C=O)-, -NH- 및 -O-로 이루어진 군으로부터 선택되는 1종 이상이 3-20개 연결된 연결기(linker)이되,The linker is -(CH 2 )-, -(C=O)-, -NH- and -O- is one or more selected from the group consisting of 3-20 connected linkers (linker),

상기 연결기는 연속하여 -O-로 연결될 수 없고; 및The linking groups cannot be continuously connected to -O-; And

상기 B는 E3 유비퀴틴 리가아제 결합 모이어티(moiety)이고,B is an E3 ubiquitin ligase binding moiety,

여기서 Linker와 A 또는 B는 화학적으로 연결된다.Here, Linker and A or B are chemically connected.

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

U-Linker-V로 표시되는 화합물로부터 A-Linker-V로 표시되는 화합물을 제조하는 단계(단계 1);Preparing a compound represented by A-Linker-V from the compound represented by U-Linker-V (step 1);

상기 단계 1에서 제조한 A-Linker-V로 표시되는 화합물로부터 A-Linker-A 또는 A-Linker-B로 표시되는 화합물을 제조하는 단계(단계 2);를 포함하는 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 제조방법을 제공한다:Preparing a compound represented by A-Linker-A or A-Linker-B from the compound represented by A-Linker-V prepared in step 1 (step 2); the A-Linker-A comprising or A method for preparing a compound represented by A-Linker-B is provided:

[반응식 1][Scheme 1]

Figure 112018098559920-pat00001
Figure 112018098559920-pat00001

(상기 반응식 1에 있어서,(In Scheme 1,

A, Linker 및 B는 상기에서 정의한 바와 같고; 및A, Linker and B are as defined above; And

U 또는 V는 각각 독립적으로 NH2 또는 OH이다).U or V are each independently NH 2 or OH).

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 세레브론(Cereblon) 관련 질환의 예방 또는 치료용 약학적 조성물을 제공한다.Furthermore, the present invention is to prevent or treat cereblon-related diseases containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient. It provides a pharmaceutical composition.

또한, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating cancer containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient. do.

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 자가면역질환의 예방 또는 치료용 약학적 조성물을 제공한다.Furthermore, the present invention is a pharmaceutical composition for the prevention or treatment of autoimmune diseases containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient. Gives

또한, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 대사질환의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for the prevention or treatment of metabolic diseases containing the compound represented by the A-Linker-A or A-Linker-B, a stereoisomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient. to provide.

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 세레브론(Cereblon) 관련 질환의 예방 또는 개선용 건강기능식품 조성물을 제공한다.Furthermore, the present invention prevents or improves cereblon-related diseases containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient. It provides a health functional food composition.

본 발명에 따른 A-Linker-A 또는 A-Linker-B로 표시되는 신규 화합물은 세레브론을 우수하게 저해하거나 분해시킬 수 있는 바, 이를 유효성분으로 함유하여 세레브론 관련 질환, 바람직하게 세레브론을 저해 또는 분해하는 것으로부터 치료, 예방 또는 개선과 같은 효과가 달성될 수 있는 질환, 예를 들어, 암, 자가면역질환, 또는 대사질환의 예방 또는 치료용 약학적 조성물로서 유용한 효과가 있다.The novel compound represented by A-Linker-A or A-Linker-B according to the present invention can excellently inhibit or decompose celebrity, and contains it as an active ingredient to treat cerebron-related diseases, preferably celebrone. It has a useful effect as a pharmaceutical composition for the prevention or treatment of diseases, for example, cancer, autoimmune diseases, or metabolic diseases, in which effects such as treatment, prevention or improvement can be achieved from inhibition or degradation.

도 1은 본 발명 실시예 1, 3, 및 8 화합물을 0 내지 10 μM로 하여, α-CRBN 및 α-Tubulin에서 웨스턴 블롯하여 나타낸 단백질 분석 결과이다.
도 2는 본 발명 실시예 1, 3, 및 8 화합물의 농도에 따른 단백질 분해력을 도시한 DC50 그래프이다.
1 is a protein analysis result shown by Western blot in α-CRBN and α-Tubulin, using the inventive examples 1, 3, and 8 compounds as 0 to 10 μM.
2 is a DC 50 graph showing proteolytic power according to the concentration of the compounds of Examples 1, 3, and 8 of the present invention.

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

이하의 설명은 본 발명의 이해를 돕기 위한 예시로서 이해되어야 하며, 본 발명의 사상 또는 범주가 하기의 설명으로부터 제한되는 것은 아니다.The following description should be understood as an example to aid the understanding of the present invention, and the spirit or scope of the present invention is not limited from the following description.

본 발명은 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염을 제공한다:The present invention provides a compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:

상기 A는 세레브론(Cerebron) E3 유비퀴틴 리가아제 결합 모이어티(moiety)이고;A is Cerebron E3 ubiquitin ligase binding moiety;

상기 Linker는 -(CH2)-, -(C=O)-, -NH- 및 -O-로 이루어진 군으로부터 선택되는 1종 이상이 3-20개 연결된 연결기(linker)이되,The linker is -(CH 2 )-, -(C=O)-, -NH- and -O- is one or more selected from the group consisting of 3-20 connected linkers (linker),

상기 연결기는 연속하여 -O-로 연결될 수 없고; 및The linking groups cannot be continuously connected to -O-; And

상기 B는 E3 유비퀴틴 리가아제 결합 모이어티(moiety)이고,B is an E3 ubiquitin ligase binding moiety,

여기서 Linker와 A 또는 B는 화학적으로 연결된다.Here, Linker and A or B are chemically connected.

이때, 상기의 화학적인 연결은 예를 들어 단일결합, 이중결합, 또는 삼중결합과 같은 공유결합일 수 있다.In this case, the chemical linkage may be, for example, a single bond, a double bond, or a covalent bond such as a triple bond.

한편,Meanwhile,

바람직하게, 상기 A는 탈리도마이드(Thalidomide), 레날리도마이드(Lenalidomide), 포말리도마이드(Pormalidomide), 이들의 유사체, 이들의 동배체, 또는 이들의 유도체이다.Preferably, the A is thalidomide, lenalidomide, pomalidomide, analogs thereof, isomers thereof, or derivatives thereof.

더욱 바람직하게,More preferably,

상기 A는 하기 (a) 내지 (g)로 표시되는 화학식 중 하나이고,A is one of the formulas represented by the following (a) to (g),

Figure 112018098559920-pat00002
,
Figure 112018098559920-pat00003
,
Figure 112018098559920-pat00004
,
Figure 112018098559920-pat00005
,
Figure 112018098559920-pat00006
,
Figure 112018098559920-pat00007
, 또는
Figure 112018098559920-pat00008
Figure 112018098559920-pat00002
,
Figure 112018098559920-pat00003
,
Figure 112018098559920-pat00004
,
Figure 112018098559920-pat00005
,
Figure 112018098559920-pat00006
,
Figure 112018098559920-pat00007
, or
Figure 112018098559920-pat00008

상기 화학식 (a), (b), (c), (d), (e), (f), 및 (g)에 있어서,In the formulas (a), (b), (c), (d), (e), (f), and (g),

W는 CH2, CHR, C=O, SO2, NH, 또는 N-C1-10의 직쇄 또는 분지쇄의 알킬이고,W is CH 2 , CHR, C=O, SO 2 , NH, or straight or branched alkyl of NC 1-10 ,

각각의 X는 독립적으로 O, S, 또는 H2이고,Each X is independently O, S, or H 2 ,

Y는 NH, N-C1-10의 직쇄 또는 분지쇄 알킬, N-C6-10 아릴, N-헤테로아릴(N, O, 및 S로 이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 5-10각환의 헤테로아릴), N-C3-10 사이클로알킬, N-헤테로사이클로알킬(N, O, 및 S로 이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 3-10각환의 헤테로사이클로알킬), O, 또는 S이고,Y is NH, NC 1-10 straight or branched chain alkyl, NC 6-10 aryl, N-heteroaryl (N, O, and 5-10 angles containing one or more heteroatoms selected from the group consisting of S) Ring heteroaryl), NC 3-10 cycloalkyl, N-heterocycloalkyl (3-10 heterocyclic alkyl containing 1 or more hetero atoms selected from the group consisting of N, O, and S), O, Or S,

Z는 O, S, 또는 H2이고,Z is O, S, or H 2 ,

G 및 G'는 각각 독립적으로 H, C1-10의 직쇄 또는 분지쇄 알킬, OH, R'로 치환 또는 비치환된 CH2-헤테로사이클릴(N, O, 및 S로 이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 3-10각환의 헤테로사이클릴), 또는 R'로 치환 또는 비치환된 벤질이고,G and G'are each independently selected from the group consisting of CH 2 -heterocyclyl (N, O, and S) substituted or unsubstituted with H, C 1-10 linear or branched alkyl, OH, R' 3-10 heterocyclic heterocyclyl containing at least one hetero atom), or benzyl substituted or unsubstituted with R',

Q1, Q2, Q3, 및 Q4는 각각 독립적으로 R'로 치환된 탄소, 또는 N 이고,Q 1 , Q 2 , Q 3 , and Q 4 are each independently carbon substituted with R', or N,

A는 C1-10의 직쇄 또는 분지쇄 알킬, C3-10 사이클로알킬, 또는 할로젠이고,A is C 1-10 straight or branched chain alkyl, C 3-10 cycloalkyl, or halogen

R은 -CONR'R", -OR', -NR'R", -SR', -SO2R', -SO2NR'R", -CR'R"-, -CR'NR'R"-, -C6-10아릴, -헤테로아릴(N, O, 및 S로 이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 5-10각환의 헤테로아릴), -C1-10의 직쇄 또는 분지쇄 알킬, -C3-10사이클로알킬, -헤테로사이클릴(N, O, 및 S로 이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 3-10각환의 헤테로사이클릴), -P(O)(OR')R", -P(O)R'R", -OP(O)(OR')R", -OP(O)R'R", -Cl, -F, -Br, -I, -CF3, -CN, -NR'SO2NR'R", -NR'CONR'R", -CONR'COR", -NR'C(=N-CN)NR'R", -C(=N-CN)NR'R", -NR'C(=N-CN)R", -NR'C(=CNO2)NR'R", -SO2NR'COR", -NO2, -CO2R', -C(C=N-OR')R", -CR'=CR'R", -CCR', -S(C=O)(C=N-R')R", -SF5 또는 -OCF3이고,R is -CONR'R", -OR', -NR'R", -SR', -SO 2 R', -SO 2 NR'R", -CR'R"-, -CR'NR'R" -, -C 6-10 aryl, -heteroaryl (a 5- to 10-membered heteroaryl containing one or more hetero atoms selected from the group consisting of N, O, and S), -C 1-10 straight chain, or Branched chain alkyl, -C 3-10 cycloalkyl, -heterocyclyl ( 3-10 heterocyclic heterocyclyl containing at least one hetero atom selected from the group consisting of N, O, and S), -P( O)(OR')R", -P(O)R'R", -OP(O)(OR')R", -OP(O)R'R", -Cl, -F, -Br, -I, -CF 3 , -CN, -NR'SO 2 NR'R", -NR'CONR'R", -CONR'COR", -NR'C(=N-CN)NR'R",- C(=N-CN)NR'R", -NR'C(=N-CN)R", -NR'C(=CNO 2 )NR'R", -SO 2 NR'COR", -NO 2 , -CO 2 R', -C(C=N-OR')R", -CR'=CR'R", -CCR', -S(C=O)(C=N-R')R" , -SF 5 or -OCF 3 ,

R' 및 R"는 H, C1-10의 직쇄 또는 분지쇄 알킬, C3-10사이클로알킬, C6-10아릴, 헤테로아릴(N, O, 및 S로 이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 5-10각환의 헤테로아릴), 헤테로사이클릴(N, O, 및 S로 이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 3-10각환의 헤테로사이클릴)로 구성된 군에서 각각 독립적으로 선택되고,R'and R" are one or more selected from the group consisting of H, C 1-10 linear or branched alkyl, C 3-10 cycloalkyl, C 6-10 aryl, heteroaryl (N, O, and S) 5-10-membered ring heteroaryl containing hetero atoms), heterocyclyl (3-10-membered ring heterocyclyl containing one or more hetero atoms selected from the group consisting of N, O, and S) Each independently selected from,

Figure 112018098559920-pat00009
는 입체특이적(R 또는 S) 또는 비-입체특이적인 결합을 나타내고; 및
Figure 112018098559920-pat00009
Represents stereospecific (R or S) or non-stereospecific binding; And

Rn은 -(CH2)m-NR3-, -(CH2)m-O-, -O-(CH2)m-(C=O)-NR3- 또는 -NR3-(CH2)m-(C=O)-NR3-이고,R n is -(CH 2 ) m -NR 3 -, -(CH 2 ) m -O-, -O-(CH 2 ) m -(C=O)-NR 3 -or -NR 3 -(CH 2 ) m -(C=O)-NR 3 -,

다시 여기서, m은 0 내지 5의 정수이고,Here again, m is an integer from 0 to 5,

상기 R3는 각각 H, 또는 비치환 또는 치환된 C1-10의 직쇄 또는 분지쇄의 알킬이고,R 3 is H, or unsubstituted or substituted C 1-10 linear or branched alkyl,

다시 여기서, 상기 치환된 알킬은 할로겐, 니트로, 또는 시아노기로 치환되고, Rn은 Linker와 공유결합된다.Here again, the substituted alkyl is substituted with a halogen, nitro, or cyano group, and R n is covalently linked with a Linker.

보다 바람직하게,More preferably,

상기 A는

Figure 112018098559920-pat00010
,
Figure 112018098559920-pat00011
,
Figure 112018098559920-pat00012
, 또는
Figure 112018098559920-pat00013
이되,A is
Figure 112018098559920-pat00010
,
Figure 112018098559920-pat00011
,
Figure 112018098559920-pat00012
, or
Figure 112018098559920-pat00013
This,

여기서 상기 R1은 -(CH2)n-NR3-, -(CH2)n-O-, -O-(CH2)n-(C=O)-NR3- 또는 -NR3-(CH2)n-(C=O)-NR3-이고,Wherein R 1 is -(CH 2 ) n -NR 3 -, -(CH 2 ) n -O-, -O-(CH 2 ) n -(C=O)-NR 3 -or -NR 3 -( CH 2 ) n -(C=O)-NR 3 -,

다시 여기서, n은 0 내지 5의 정수이고,Here again, n is an integer from 0 to 5,

상기 R2 및 R3는 각각 독립적으로 H, 또는 비치환 또는 치환된 C1-10의 직쇄 또는 분지쇄의 알킬이고,R 2 and R 3 are each independently H or unsubstituted or substituted C 1-10 linear or branched alkyl;

다시 여기서, 상기 치환된 알킬은 할로겐, 니트로, 또는 시아노기로 치환된다.Here again, the substituted alkyl is substituted with a halogen, nitro, or cyano group.

더욱 바람직하게,More preferably,

상기 A는

Figure 112018098559920-pat00014
,
Figure 112018098559920-pat00015
,
Figure 112018098559920-pat00016
,
Figure 112018098559920-pat00017
,
Figure 112018098559920-pat00018
,
Figure 112018098559920-pat00019
,
Figure 112018098559920-pat00020
, 또는
Figure 112018098559920-pat00021
이되,A is
Figure 112018098559920-pat00014
,
Figure 112018098559920-pat00015
,
Figure 112018098559920-pat00016
,
Figure 112018098559920-pat00017
,
Figure 112018098559920-pat00018
,
Figure 112018098559920-pat00019
,
Figure 112018098559920-pat00020
, or
Figure 112018098559920-pat00021
This,

여기서 상기 R1은 -(CH2)n-NR3-, -(CH2)n-O-, -O-(CH2)n-(C=O)-NR3- 또는 -NR3-(CH2)n-(C=O)-NR3-이고,Wherein R 1 is -(CH 2 ) n -NR 3 -, -(CH 2 ) n -O-, -O-(CH 2 ) n -(C=O)-NR 3 -or -NR 3 -( CH 2 ) n -(C=O)-NR 3 -,

다시 여기서, n은 0 내지 5의 정수이고,Here again, n is an integer from 0 to 5,

상기 R2 및 R3는 각각 독립적으로 H, 또는 비치환 또는 치환된 C1-10의 직쇄 또는 분지쇄의 알킬이고,R 2 and R 3 are each independently H or unsubstituted or substituted C 1-10 linear or branched alkyl;

다시 여기서, 상기 치환된 알킬은 할로겐, 니트로, 또는 시아노기로 치환된다.Here again, the substituted alkyl is substituted with a halogen, nitro, or cyano group.

한편, 본 발명에 따른 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염에 있어서,Meanwhile, in the compound represented by A-Linker-A or A-Linker-B according to the present invention, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof,

바람직하게, 상기 Linker는,Preferably, the Linker,

Figure 112018098559920-pat00022
이되,
Figure 112018098559920-pat00022
This,

여기서, 상기 a 및 i는 독립적으로 0 또는 1이고;Where a and i are independently 0 or 1;

상기 b는 0-20의 정수이고;B is an integer of 0-20;

상기 c는 0 또는 1이고;C is 0 or 1;

상기 d는 0-3의 정수이고;D is an integer of 0-3;

상기 e는 0 또는 1이고;E is 0 or 1;

상기 f는 1-10의 정수이고;F is an integer of 1-10;

상기 g는 0 또는 1이고;G is 0 or 1;

상기 h는 0-3의 정수이고;H is an integer of 0-3;

상기 j 및 k는 독립적으로 0-5의 정수이다.J and k are independently integers of 0-5.

한편,Meanwhile,

보다 바람직하게,More preferably,

상기 Linker는,The Linker,

Figure 112018098559920-pat00023
,
Figure 112018098559920-pat00024
,
Figure 112018098559920-pat00025
,
Figure 112018098559920-pat00026
,
Figure 112018098559920-pat00027
,
Figure 112018098559920-pat00028
,
Figure 112018098559920-pat00029
,
Figure 112018098559920-pat00030
,
Figure 112018098559920-pat00031
,
Figure 112018098559920-pat00032
,
Figure 112018098559920-pat00033
,
Figure 112018098559920-pat00034
,
Figure 112018098559920-pat00035
,
Figure 112018098559920-pat00036
,
Figure 112018098559920-pat00037
,
Figure 112018098559920-pat00038
,
Figure 112018098559920-pat00039
,
Figure 112018098559920-pat00040
,
Figure 112018098559920-pat00041
, 또는
Figure 112018098559920-pat00042
이다.
Figure 112018098559920-pat00023
,
Figure 112018098559920-pat00024
,
Figure 112018098559920-pat00025
,
Figure 112018098559920-pat00026
,
Figure 112018098559920-pat00027
,
Figure 112018098559920-pat00028
,
Figure 112018098559920-pat00029
,
Figure 112018098559920-pat00030
,
Figure 112018098559920-pat00031
,
Figure 112018098559920-pat00032
,
Figure 112018098559920-pat00033
,
Figure 112018098559920-pat00034
,
Figure 112018098559920-pat00035
,
Figure 112018098559920-pat00036
,
Figure 112018098559920-pat00037
,
Figure 112018098559920-pat00038
,
Figure 112018098559920-pat00039
,
Figure 112018098559920-pat00040
,
Figure 112018098559920-pat00041
, or
Figure 112018098559920-pat00042
to be.

또 다른 한편, 본 발명에 따른 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염에 있어서,On the other hand, in the compound represented by A-Linker-A or A-Linker-B according to the present invention, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof,

바람직하게,Preferably,

상기 B는 E3 유비퀴틴 리가제 결합 모이어티(리간드)라면 제한없이 사용될 수있고, 여기서, 상기 E3 유비퀴틴 리가제로의 일예로, AMFR, APC/Cdc20, APC/Cdh1, C6orf157, Cbl, CBLL1, CHFR, CHIP, DTL, E6-AP, HACE1, HECW1, HECW2, HERC2, HUWE1, HYD, ITCH, LNX1, MARCH-I, MARCH-IV, MARCH-VII, MARCH-VIII, MDM2, MEKK1, MIB1, MIB2, MycBP2, NEDD4L, PELI1, Pirh2, PJA1, RFFL, RFWD2, Rictor, RNF5, RNF8, RNF19, RNF20, RNF34, RNF40, RNF138, RNF168, SCF/β-TrCP, SCF/FBW7, SCF/Skp2, SHPRH, SIAH1, SIAH2, SMURF1, SMURF2, TOPORS, TRAF6, TRIM63, UBR2, UHRF2, VHL, WWP1, WWP2, 또는 CRBN일 수 있고, 이의 결합 뫼어티라면 제한없이 본 발명에 적용하여 사용할 수 있다.The B can be used without limitation as long as it is an E3 ubiquitin ligase binding moiety (ligand), wherein as an example of the E3 ubiquitin ligase, AMFR, APC/Cdc20, APC/Cdh1, C6orf157, Cbl, CBLL1, CHFR, CHIP , DTL, E6-AP, HACE1, HECW1, HECW2, HERC2, HUWE1, HYD, ITCH, LNX1, MARCH-I, MARCH-IV, MARCH-VII, MARCH-VIII, MDM2, MEKK1, MIB1, MIB2, MycBP2, NEDD4L , PELI1, Pirh2, PJA1, RFFL, RFWD2, Rictor, RNF5, RNF8, RNF19, RNF20, RNF34, RNF40, RNF138, RNF168, SCF/β-TrCP, SCF/FBW7, SCF/Skp2, SHPRH, SIAH1, SIAH2, SMURF1 , SMURF2, TOPORS, TRAF6, TRIM63, UBR2, UHRF2, VHL, WWP1, WWP2, or CRBN, and any combination moieties thereof can be applied to the present invention without limitation.

본 발명의 일 구체예에서는, 상기 B는 CRBN(cereblon) 또는 VHL(von Hippel-Lindau) E3 리가제 결합 모이어티일 수 있다.In one embodiment of the invention, the B may be a CRBN (cereblon) or VHL (von Hippel-Lindau) E3 ligase binding moiety.

본 발명의 또 다른 구체예에서는,In another embodiment of the invention,

상기 B는 하기 화학식 1로 표시되는 화합물이되,B is a compound represented by Formula 1 below,

[화학식 1][Formula 1]

Figure 112018098559920-pat00043
Figure 112018098559920-pat00043

상기 화학식 1에 있어서,In Chemical Formula 1,

R4은 OR6이고,R 4 is OR 6 ,

R5는 선택적으로 치환 또는 비치환된 -NR6-(CH2)o-C6-10아릴-N, O, 및 S로이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 5-10원자 헤테로아릴이되,R 5 is an optionally substituted or unsubstituted -NR 6 -(CH 2 )oC 6-10 aryl-N, O, and 5-10 atom heteroaryl comprising one or more hetero atoms selected from the group consisting of S This,

상기 치환된 -NR6-(CH2)o-C6-10아릴-N, O, 및 S로이루어진 군으로부터 선택되는 1종 이상의 헤테로 원자를 포함하는 5-10원자 헤테로아릴은 C1-10의 직쇄 또는 분지쇄의 알킬로 치환되고,The substituted -NR 6 -(CH 2 )oC 6-10 aryl-N, O, and 5-10 atom heteroaryl including at least one hetero atom selected from the group consisting of S is a straight chain of C 1-10 Or substituted with branched alkyl,

여기서, 상기 R6는 H 또는 C1-5의 직쇄 또는 분지쇄의 알킬이고, 상기 o는 0-5이고;Wherein R 6 is H or C 1-5 straight or branched chain alkyl, and o is 0-5;

R7는 -CR8-NR9-이되,R 7 is -CR 8 -NR 9-

여기서, R8는 C1-10의 직쇄 또는 분지쇄의 알킬이고, R9는 H 또는 C1-5의 직쇄 또는 분지쇄의 알킬이다.Here, R 8 is C 1-10 straight or branched chain alkyl, and R 9 is H or C 1-5 straight or branched chain alkyl.

더욱 바람직하게,More preferably,

상기 B는

Figure 112018098559920-pat00044
이되,B is
Figure 112018098559920-pat00044
This,

여기서 상기 R2 및 R3는 각각 독립적으로 H, 또는 비치환 또는 치환된 C1-10의 직쇄 또는 분지쇄의 알킬이고,Wherein R 2 and R 3 are each independently H, or unsubstituted or substituted C 1-10 linear or branched alkyl;

다시 여기서, 상기 치환된 알킬은 할로겐, 니트로, 또는 시아노기로 치환된다.Here again, the substituted alkyl is substituted with a halogen, nitro, or cyano group.

본 발명에 따른 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 바람직한 예로는 하기 화합물을 들 수 있다.Preferred examples of the compound represented by A-Linker-A or A-Linker-B according to the present invention include the following compounds.

(1) (2S,4R)-1-((S)-2-(7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헵탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(1) (2S,4R)-1-((S)-2-(7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)heptanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;

(2) (2S,4R)-1-((S)-2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥산아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(2) (2S,4R)-1-((S)-2-(6-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Ylamino)hexaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;

(3) (2S,4R)-1-((S)-2-(9-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)노난아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(3) (2S,4R)-1-((S)-2-(9-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)nonanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;

(4) (2S,4R)-1-((S)-2-tert-부틸-17-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-4-옥소-6,9,12,15-테트라옥사-3-아자헵타데카-1-노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(4) (2S,4R)-1-((S)-2-tert-butyl-17-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)-4-oxo-6,9,12,15-tetraoxa-3-azaheptadeca-1-noyl)-4-hydroxy-N-(4-(4-methylthiazole- 5-yl)benzyl)pyrrolidine-2-carboxamide;

(5) (2S,4R)-1-((S)-2-(2-(3-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-2-옥소에톡시)프로폭시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(5) (2S,4R)-1-((S)-2-(2-(3-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-di Oxoisoindoline-4-ylamino)-2-oxoethoxy)propoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazole- 5-yl)benzyl)pyrrolidine-2-carboxamide;

(6) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N9-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)노난디아미드;(6) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 9 -((S)-1-(( 2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutane- 2-yl)nonanediamide;

(7) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N8-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)옥탄디아미드;(7) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)-N 8 -((S)-1-(( 2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutane- 2-yl)octanediamide;

(8) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N7-((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)헵탄디아미드;(8) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 7 -((S)-1-(( 2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxo Butan-2-yl)heptanediamide;

(9) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N14-((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)-3,6,9,12-테트라옥사테트라데칸-1,14-디아미드;(9) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 14 -((S)-1-(( 2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxo Butan-2-yl)-3,6,9,12-tetraoxatetradecane-1,14-diamide;

(10) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N5-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)글루타르아미드;(10) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 5 -((S)-1-(( 2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutane- 2-yl)glutaramide;

(11) N1-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)메틸)-N7-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)헵탄디아미드;(11) N 1 -((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)methyl)-N 7 -((S)-1 -((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1- Oxobutan-2-yl)heptanediamide;

(12) (2S,4R)-1-((S)-2-(11-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)운데칸아미도)-3.3-디메틸부탄오일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(12) (2S,4R)-1-((S)-2-(11-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)undecanamido)-3.3-dimethylbutanyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;

(13) (2S,4R)-1-((S)-2-(8-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)옥탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(13) (2S,4R)-1-((S)-2-(8-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Ylamino)octaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;

(14) (2S,4R)-1-((S)-2-(12-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)도데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(14) (2S,4R)-1-((S)-2-(12-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Ilamino)dodecaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide ;

(15) (2S,4R)-1-((S)-2-(2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥실옥시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(15) (2S,4R)-1-((S)-2-(2-(6-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)hexyloxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine -2-carboxamide;

(16) (2S,4R)-1-((S)-2-(10-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(16) (2S,4R)-1-((S)-2-(10-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)decanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;

(17) (2S,4R)-1-((S)-2-tert-부틸-14-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-4-옥소-6,9,12-트리옥사-3-아자테트라데카-1-노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(17) (2S,4R)-1-((S)-2-tert-butyl-14-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)-4-oxo-6,9,12-trioxa-3-azatetradeca-1-noyl)-4-hydroxy-N-(4-(4-methylthiazole-5- 1)benzyl)pyrrolidine-2-carboxamide;

(18) (2S,4R)-1-((S)-2-(11-((3-(2,6- 디옥소피페리딘-3-일)-2-메틸-4-옥소-3,4-디히드로퀴나졸린-5-일)아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카르복스아미드;(18) (2S,4R)-1-((S)-2-(11-((3-(2,6-dioxopiperidin-3-yl)-2-methyl-4-oxo-3, 4-dihydroquinazolin-5-yl)amino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl )Pyrrolidine-2-carboxamide;

(19) 4,4'-(노난-1,9-디일비스(아자네디일))비스(2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온);(19) 4,4'-(nonane-1,9-diylbis(azanediyl))bis(2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione );

(20) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(20) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolid Dean-2-carboxamide;

(21) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일옥시)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(21) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-yloxy)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolid Dean-2-carboxamide;

(22) (2S,4R)-1-((S)-2-(11-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-5-일아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;(22) (2S,4R)-1-((S)-2-(11-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5- Ilamino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide ;

(23) 3,3'-(5,5'-(노난-1,9-디일비스(아잔디일))비스(4-옥소벤조[d][1,2,3]트리아진-5,3(4H)-디일))디피페리딘-2,6-디온;(23) 3,3'-(5,5'-(nonane-1,9-diylbis(azanediyl))bis(4-oxobenzo[d][1,2,3]triazine-5, 3(4H)-diyl))dipiperidine-2,6-dione;

(24) 2-(2,6-디옥소피페리딘-3-일)-4-(9-(3-(2,6-디옥소피페리딘-3-일)-4-옥소-3,4-디히드로벤조[d][1,2,3]트리아진-5-일아미노)노닐아미노)이소인돌린-1,3-디온; 및(24) 2-(2,6-dioxopiperidin-3-yl)-4-(9-(3-(2,6-dioxopiperidin-3-yl)-4-oxo-3,4 -Dihydrobenzo[d][1,2,3]triazin-5-ylamino)nonylamino)isoindoline-1,3-dione; And

(25) (2S,4R)-1-((S)-2-(11-(3-(2,6-디옥소피페리딘-3-일)-4-옥소-3,4-디히드로벤조[d][1,2,3]트리아진-5-일아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드.(25) (2S,4R)-1-((S)-2-(11-(3-(2,6-dioxopiperidin-3-yl)-4-oxo-3,4-dihydrobenzo [d][1,2,3]triazin-5-ylamino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazole- 5-yl)benzyl)pyrrolidine-2-carboxamide.

본 발명의 A-Linker-A 또는 A-Linker-B로 표시되는 화합물은 약학적으로 허용 가능한 염의 형태로 사용할 수 있으며, 염으로는 약학적으로 허용 가능한 유리산(free acid)에 의해 형성된 산 부가염이 유용하다. 산 부가염은 염산, 질산, 인산, 황산, 브롬화수소산, 요드화수소산, 아질산, 아인산 등과 같은 무기산류, 지방족 모노 및 디카르복실레이트, 페닐-치환된 알카노에이트, 하이드록시 알카노에이트 및 알칸디오에이트, 방향족 산류, 지방족 및 방향족 설폰산류 등과 같은 무독성 유기산, 아세트산, 안식향산, 구연산, 젖산, 말레인산, 글루콘산, 메탄설폰산, 4-톨루엔설폰산, 주석산, 푸마르산 등과 같은 유기산으로부터 얻는다. 이러한 약학적으로 무독한 염의 종류로는 설페이트, 피로설페이트, 바이설페이트, 설파이트, 바이설파이트, 니트레이트, 포스페이트, 모노하이드로겐 포스페이트, 다이하이드로겐 포스페이트, 메타포스페이트, 피로포스페이트 클로라이드, 브로마이드, 아이오다이드, 플루오라이드, 아세테이트, 프로피오네이트, 데카노에이트, 카프릴레이트, 아크릴레이트, 포메이트, 이소부티레이트, 카프레이트, 헵타노에이트, 프로피올레이트, 옥살레이트, 말로네이트, 석시네이트, 수베레이트, 세바케이트, 푸마레이트, 말리에이트, 부틴-1,4-디오에이트, 헥산-1,6-디오에이트, 벤조에이트, 클로로벤조에이트, 메틸벤조에이트, 디니트로 벤조에이트, 하이드록시벤조에이트, 메톡시벤조에이트, 프탈레이트, 테레프탈레이트, 벤젠설포네이트, 톨루엔설포네이트, 클로로벤젠설포네이트, 크실렌설포네이트, 페닐아세테이트, 페닐프로피오네이트, 페닐부티레이트, 시트레이트, 락테이트, β-하이드록시부티레이트, 글리콜레이트, 말레이트, 타트레이트, 메탄설포네이트, 프로판설포네이트, 나프탈렌-1-설포네이트, 나프탈렌-2-설포네이트, 만델레이트 등을 포함한다.The compound represented by A-Linker-A or A-Linker-B of the present invention can be used in the form of a pharmaceutically acceptable salt, and as the salt, an acid formed by a pharmaceutically acceptable free acid is added. Salt is useful. Acid addition salts include inorganic acids such as hydrochloric acid, nitric acid, phosphoric acid, sulfuric acid, hydrobromic acid, hydroiodic acid, nitrous acid and phosphorous acid, aliphatic mono and dicarboxylates, phenyl-substituted alkanoates, hydroxy alkanoates and alkanes It is obtained from non-toxic organic acids such as dioate, aromatic acids, aliphatic and aromatic sulfonic acids, acetic acid, benzoic acid, citric acid, lactic acid, maleic acid, gluconic acid, methanesulfonic acid, 4-toluenesulfonic acid, tartaric acid, fumaric acid and the like. The types of pharmaceutically non-toxic salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate chloride, bromide, and eye Odide, fluoride, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, sube Late, sebacate, fumarate, maleate, butyne-1,4-dioate, hexane-1,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitro benzoate, hydroxybenzoate, Methoxybenzoate, phthalate, terephthalate, benzenesulfonate, toluenesulfonate, chlorobenzenesulfonate, xylenesulfonate, phenylacetate, phenylpropionate, phenylbutyrate, citrate, lactate, β-hydroxybutyrate, Glycolate, malate, tartrate, methanesulfonate, propanesulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, mandelate and the like.

본 발명에 따른 산 부가염은 통상의 방법으로 제조할 수 있으며, 예를 들면 A-Linker-A 또는 A-Linker-B의 유도체를 메탄올, 에탄올, 아세톤, 디클로로메탄, 아세토니트릴 등과 같은 유기용매에 녹이고 유기산 또는 무기산을 가하여 생성된 침전물을 여과, 건조시켜 제조하거나, 용매와 과량의 산을 감압 증류한 후 건조시켜 유기용매 하에서 결정화시켜서 제조할 수 있다.The acid addition salt according to the present invention can be prepared by a conventional method, for example, a derivative of A-Linker-A or A-Linker-B in an organic solvent such as methanol, ethanol, acetone, dichloromethane, acetonitrile, etc. The precipitate produced by dissolving and adding an organic acid or an inorganic acid can be produced by filtration and drying, or by distilling the solvent and excess acid under reduced pressure and drying to crystallize under an organic solvent.

또한, 염기를 사용하여 약학적으로 허용가능한 금속염을 만들 수 있다. 알칼리 금속 또는 알칼리 토금속 염은 예를 들면 화합물을 과량의 알칼리 금속 수산화물 또는 알칼리 토금속 수산화물 용액 중에 용해하고, 비용해 화합물 염을 여과하고, 여액을 증발, 건조시켜 얻는다. 이때, 금속염으로는 나트륨, 칼륨 또는 칼슘염을 제조하는 것이 제약상 적합하다. 또한, 이에 대응하는 염은 알칼리 금속 또는 알칼리 토금속 염을 적당한 음염(예, 질산은)과 반응시켜 얻는다.In addition, bases can be used to make pharmaceutically acceptable metal salts. The alkali metal or alkaline earth metal salt is obtained, for example, by dissolving the compound in an excess of an alkali metal hydroxide or alkaline earth metal hydroxide solution, filtering the inexpensive compound salt, and evaporating and drying the filtrate. At this time, it is suitable to manufacture sodium, potassium or calcium salts as metal salts. Further, the corresponding salt is obtained by reacting an alkali metal or alkaline earth metal salt with a suitable negative salt (eg, silver nitrate).

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물 및 이의 약학적으로 허용가능한 염뿐만 아니라, 이로부터 제조될 수 있는 용매화물, 광학 이성질체, 수화물 등을 모두 포함한다.Furthermore, the present invention includes all of the compounds represented by A-Linker-A or A-Linker-B and pharmaceutically acceptable salts thereof, as well as solvates, optical isomers, hydrates, etc., which can be prepared therefrom. .

또한, 하기 반응식 1에 나타난 바와 같이,In addition, as shown in Scheme 1 below,

U-Linker-V로 표시되는 화합물로부터 A-Linker-V로 표시되는 화합물을 제조하는 단계(단계 1);Preparing a compound represented by A-Linker-V from the compound represented by U-Linker-V (step 1);

상기 단계 1에서 제조한 A-Linker-V로 표시되는 화합물로부터 A-Linker-A 또는 A-Linker-B로 표시되는 화합물을 제조하는 단계(단계 2);를 포함하는 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 제조방법을 제공한다:Preparing a compound represented by A-Linker-A or A-Linker-B from the compound represented by A-Linker-V prepared in step 1 (step 2); the A-Linker-A comprising or A method for preparing a compound represented by A-Linker-B is provided:

[반응식 1][Scheme 1]

Figure 112018098559920-pat00045
Figure 112018098559920-pat00045

(상기 반응식 1에 있어서,(In Scheme 1,

A, Linker 및 B는 상기에서 정의한 바와 같고; 및A, Linker and B are as defined above; And

U 또는 V는 각각 독립적으로 NH2 또는 OH이다).U or V are each independently NH 2 or OH).

이하, 본 발명에 따른 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 제조방법을 단계별로 상세히 설명한다.Hereinafter, a method for preparing a compound represented by A-Linker-A or A-Linker-B according to the present invention will be described in detail step by step.

본 발명에 따른 화학식 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 제조방법에 있어서, 상기 단계 1은 U-Linker-V로 표시되는 화합물로부터 A-Linker-V로 표시되는 화합물을 제조하는 단계이다.In the method for preparing a compound represented by the formula A-Linker-A or A-Linker-B according to the present invention, the step 1 is a compound represented by A-Linker-V from the compound represented by U-Linker-V It is a manufacturing step.

이때, 상기 단계는 본 발명 화합물의 저해 또는 분해하고자 하는 표적 단백질인 세레브론에 결합하기 위한 리간드 모이어티를 도입하는 단계로, 예를 들어 연결기(Linker)에 A로 표시되는 세레브론(Cereblon) 리가제 리간드 화합물의 도입 단계로 생각될 수 있다.At this time, the step is a step of introducing a ligand moiety for binding to the target protein to be inhibited or degraded of the compound of the present invention, cerebron, for example, a cereblon ligase represented by A in the linker It can be considered as the step of introducing the first ligand compound.

이때, Linker와 A가 연결되는 결합은 화학적인 결합으로, 예를 들어, 공유결합, 바람직하게 단일결합, 이중결합 또는 삼중결합일 수 있다. 따라서, 상기 Linker와 A를 연결하는 단계인 단계 1은 해당 분야에 사용될 수 있는 연결기 도입 방법이라면 특별히 제한되지 않고 사용될 수 있으며, U-Linker-V에 있어서, U가 NH2인 경우는 A-할로젠, 바람직하게 A-F로 표시되는 화합물, 일 실시예로 하기 본 발명의 실시예에서, A-F로 표시되는 화합물의 하나의 예시 화합물은 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온이고, U-Linker-V 및 A-할로젠을 반응시켜 단계 1의 목적 화합물인 A-Linker-V로 표시되는 화합물을 제조할 수 있는 방법이라면, 제한없이 본 발명의 제조방법에 포함된다. 한편, U가 OH인 경우, A-NH2로 표시되는 화합물, 일 실시예로 하기 본 발명의 실시예에서, 포말리도마이드(Pomalidomide)를 사용하였고, U-Linker-V 및 A-NH2을 반응시켜 단계 1의 목적 화합물인 A-Linker-V로 표시되는 화합물을 제조할 수 있는 방법이라면, 제한없이 본 발명의 제조방법에 포함된다.At this time, the linker and the linker A is a chemical bond, for example, may be a covalent bond, preferably a single bond, a double bond or a triple bond. Thus, the step of coupling the Linker and if A 1 is a linking group introducing method that can be used in the art may be used is not particularly limited, in the U-Linker-V, U is the case of the NH 2 to A- Rosen, preferably a compound represented by AF, in one embodiment, in the Examples of the present invention, one exemplary compound of the compound represented by AF is 2-(2,6-dioxopiperidin-3-yl)- If 4-fluoroisoindoline-1,3-dione, U-Linker-V and A-halogen is reacted to produce a compound represented by A-Linker-V, the target compound of step 1, It is included in the manufacturing method of the present invention without limitation. On the other hand, when U is OH, a compound represented by A-NH 2 , in one embodiment of the present invention as below, was used pomalidomide (Pomalidomide), U-Linker-V and A-NH 2 Any method capable of producing a compound represented by A-Linker-V, the target compound of step 1 by reaction, is included in the production method of the present invention without limitation.

또한, 상기 단계 1에서 사용 가능한 용매로는 H2O, 에탄올, 테트라하이드로퓨란(THF), 디클로로메탄, 톨루엔, 아세토니트릴, 디메틸포름아미드, 이의 혼합물 등을 사용할 수 있고, 바람직하게는 디메틸포름아미드(DMF)를 사용할 수 있다.In addition, as the solvent usable in step 1, H 2 O, ethanol, tetrahydrofuran (THF), dichloromethane, toluene, acetonitrile, dimethylformamide, mixtures thereof, and the like can be used, and preferably dimethylformamide (DMF) can be used.

또한, 상기 단계에서 반응온도는 특별히 제한되지 않으나, 바람직하게 80-100℃에서 용매의 비등점 사이에서 수행하는 것이 바람직하고, 반응시간은 특별한 제약은 없으나, 5시간 이상, 10시간 이상, 20시간 이상, 밤새 동안 반응하는 것이 바람직하다.In addition, the reaction temperature in the above step is not particularly limited, but preferably, it is preferably performed between the boiling points of the solvent at 80-100°C, and the reaction time is not particularly limited, but 5 hours or more, 10 hours or more, 20 hours or more It is preferred to react overnight.

본 발명에 따른 화학식 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 제조방법에 있어서, 상기 단계 2는 상기 단계 1에서 제조한 A-Linker-V로 표시되는 화합물로부터 A-Linker-A 또는 A-Linker-B로 표시되는 화합물을 제조하는 단계이다.In the method for preparing a compound represented by the formula A-Linker-A or A-Linker-B according to the present invention, step 2 is A-Linker- from the compound represented by A-Linker-V prepared in step 1 above This is a step for preparing a compound represented by A or A-Linker-B.

이때, 상기 단계는 A-Linker로 표시되는 화합물에 A 또는 B를 도입하는 단계로, 리간드 A 또는 B는 세레브론 또는 VHL E3 유비퀴틴 리가제와 결합할 수 있고, 표적 단백질(세레브론)에 다중유비퀴틴화(Polyubiquitination)를 유도한다.At this time, the step is a step of introducing A or B into the compound represented by A-Linker, ligand A or B can bind to celebrity or VHL E3 ubiquitin ligase, and multi-ubiquitin to target protein (cerebron) Induces polyubiquitination.

이때, A-Linker와 A 또는 B가 연결되는 결합은 화학적인 결합으로, 예를 들어, 공유결합, 바람직하게 단일결합, 이중결합 또는 삼중결합일 수 있다. 따라서, 상기 A-Linker와 A 또는 B를 연결하는 단계인 단계 2는 비제한적으로, A-Linker-V에 있어서, V가 NH2인 경우는 A-할로젠 또는 B-할로젠, 바람직하게 A-F 또는 B-F로 표시되는 화합물, 일 실시예로 하기 본 발명의 실시예에서, A-F로 표시되는 화합물의 하나의 예시 화합물은 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온이고, A-Linker-V 및 A-할로젠 또는 B-할로젠을 반응시켜 단계 1의 목적 화합물인 A-Linker-A 또는 A-Linker-B로 표시되는 화합물을 제조할 수 있는 방법이라면, 제한없이 본 발명의 제조방법에 포함된다. 한편, V가 OH인 경우, A-NH2 또는 B-NH2를 사용할 수 있고, 일 실시예로 B-NH2는 (2R,4R)-1-((R)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드이고, A-Linker-V 및 A-NH2 또는 B-NH2을 반응시켜 단계 1의 목적 화합물인 A-Linker-A 또는 A-Linker-B로 표시되는 화합물을 제조할 수 있는 방법이라면, 제한없이 본 발명의 제조방법에 포함된다.At this time, the A-Linker and A or B are connected to a chemical bond, for example, a covalent bond, preferably a single bond, a double bond or a triple bond. Therefore, step 2, which is a step of connecting A or B with A-Linker, is not limited, and in A-Linker-V, when V is NH 2 , A-halogen or B-halogen, preferably AF Or a compound represented by BF, in one embodiment, in the Examples of the present invention, one exemplary compound of the compound represented by AF is 2-(2,6-dioxopiperidin-3-yl)-4-fluoro A compound represented by A-Linker-A or A-Linker-B, which is the desired compound of Step 1 by reacting A-Linker-V and A-halogen or B-halogen, which is a leusoindole-1,3-dione If it can be produced, it is included in the manufacturing method of the present invention without limitation. Meanwhile, when V is OH, A-NH 2 or B-NH 2 may be used, and in one embodiment, B-NH 2 is (2R, 4R)-1-((R)-2-amino-3, 3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide, A-Linker-V and A-NH Any method capable of producing a compound represented by A-Linker-A or A-Linker-B, which is the target compound of Step 1 by reacting 2 or B-NH 2 , is included in the production method of the present invention without limitation.

또한, 상기 단계 1에서 사용 가능한 용매로는 H2O, 에탄올, 테트라하이드로퓨란(THF), 디클로로메탄, 톨루엔, 아세토니트릴, 디메틸포름아미드, 이의 혼합물 등을 사용할 수 있고, 바람직하게는 디메틸포름아미드(DMF)를 사용할 수 있다.In addition, as the solvent usable in step 1, H 2 O, ethanol, tetrahydrofuran (THF), dichloromethane, toluene, acetonitrile, dimethylformamide, mixtures thereof, and the like can be used, and preferably dimethylformamide (DMF) can be used.

또한, 상기 단계에서 반응온도는 특별히 제한되지 않으나, 바람직하게 10-40℃에서 용매의 비등점 사이에서 수행하는 것이 바람직하고, 반응시간은 특별한 제약은 없으나, 5시간 이상, 10시간 이상, 20시간 이상, 밤새 동안 반응하는 것이 바람직하다.In addition, the reaction temperature in the above step is not particularly limited, but preferably is performed between the boiling point of the solvent at 10-40 ℃, the reaction time is not particularly limited, 5 hours or more, 10 hours or more, 20 hours or more It is preferred to react overnight.

상기의 제조방법은 보다 바람직하게 하기 본 발명의 실시예 화합물과 같이 제조할 수 있으나, 본 발명이 이에 제한되는 것은 아니다.The above production method may be more preferably prepared as in Example compound of the present invention, but the present invention is not limited thereto.

한편, 본 명세서에 기재된 제조방법은 당업자가 제조되는 화합물의 수율 등을 고려하여, 반응 조건, 예를 들어 반응 온도, 반응시간, 반응 단계를 수정하거나 변경할 수 있고, 반응 단계는 반복되거나 순서가 바뀌어 수행될 수 있다.On the other hand, the manufacturing method described in this specification can be modified or changed reaction conditions, for example, reaction temperature, reaction time, and reaction step, in consideration of the yield of the compound prepared by those skilled in the art, and the reaction step is repeated or the order is changed. Can be performed.

또한, 종래 알려진 유기합성 분야에서의 통상적인 합성방법을 사용하여, 본 발명에서 제공하고자 하는 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 제조방법은 제한없이 본 발명의 범주로 포함되는 것으로 이해되어야 한다.In addition, using a conventional synthetic method known in the field of organic synthesis, the method for preparing a compound represented by A-Linker-A or A-Linker-B to be provided in the present invention is included in the scope of the present invention without limitation. It should be understood to be.

나아가, 각각의 연결기(Linker)와 본 발명에서 제공하고자 하는 화합물의 양단의 모이어티를 설계할 수 있는 방법이라면, 예를들어 한쪽으로 세레브론 단백질 결합 리간드 모이어티를, 다른 한편으로 E3 유비퀴틴 리가제 리간드 모이어티를 구성하고, 이를 용이하게 연결하여 설계하는 제조 방법이라면 본 발명에 포함되는 것으로 이해되어야 한다.Furthermore, if a method capable of designing the moieties at both ends of each linker and the compound to be provided in the present invention is, for example, a cerebron protein binding ligand moiety on one side and an E3 ubiquitin ligase on the other side It should be understood that any manufacturing method of constructing and designing a ligand moiety and easily connecting it is included in the present invention.

예를 들어, 본 발명 화합물을 2 단위(unit) 이상으로 직렬 또는 병렬로 구성하는 정도의 변경은 본 발명이 달성하고자 하는 바가, 한편으로 세로브론 단백질과 결합하는 모이어티를 제공하고, 다른 한편으로 E3 유비퀴틴 리가제와 결합하는 모이어티를 제공하여, 세레브론 단백질로 다중유비퀴틴화를 유도하고자 하는 것을 목적하는 바, 상기 변경과 같이 화합물의 설계를 직렬 또는 병렬 반복 또는 연결기를 변형 또는 수정하여, 하나의 세레브론 결합 모이어티에, 두개 이상의 E3 유비퀴틴 리가제 모이어티로 구성하거나, 반대로 하나의 E3 유비퀴틴 리가제 모이어티에 두개 이상의 세레브론 결합 모이어티를 구성하는 것과 같은 간단한 설계 변형으로 도출될 수 있는 화합물 및 이로부터 본 발명과 같이, 세레브론의 우수한 저해 또는 분해를 달성하고자 한다면, 본 발명은 이러한 변경 또는 수정을 포함하는 것으로 이해되어야 한다. 즉, 이건 발명에서 용이하게 당업자가 도출해내는 정도의 변경과 수정은, 본 발명에 포함되는 것으로 이해되어야 한다.For example, a change in the degree to which the compound of the present invention is composed of two or more units in series or in parallel is what the present invention seeks to achieve, on the one hand providing a moiety that binds to the serobron protein, and on the other hand By providing a moiety that binds to E3 ubiquitin ligase, it is intended to induce multiple ubiquitination with a celebrity protein. Compounds that can be derived from simple design modifications such as constituting two or more E3 ubiquitin ligase moieties, or conversely, one or more E3 ubiquitin ligase moieties to a celebrity-binding moiety of From this, if it is desired to achieve good inhibition or degradation of cerebron, as in the present invention, it should be understood that the present invention includes such alterations or modifications. That is, it should be understood that this is easily included in the present invention, changes and modifications to the extent that a person skilled in the art derives from the present invention.

한편, 본 발명의 A-Linker-A 또는 A-Linker-B로 표시되는 화합물은 한 편의 A 리간드 모이어티로는 표적 단백질인 세레브론과 결합하고, 다른 한 편의 A 또는 B 리간드 모이어티로는 E3 유비퀴틴 리가제와 결합하여, 최종적으로 E3 유비퀴틴 리가제가 표적 단백질(세레브론)로 다중유비퀴틴화를 유도시키고, 이 후 단백질 분해 효소, 예를 들어 26S 프로테아좀(proteasome)에 다중유비퀴틴화된 단백질(세레브론)을 인식시켜, 분해시키는 효과를 달성하는데, 이러한 단백질 분해를 통한 약리 효과는, 세레브론으로 기인하는 모든 관련 질병, 예를 들어, 세레브론의 이상, 과활성, 등과 같은 작용으로부터 야기되는 질환, 예를 들어, 암, 예를 들어, 자가면역질환, 예를 들어 대사질환과 같은, 종래 세레브론과 관련된 질환으로 규명된 모든 질환에서 유용한 효과를 나타낼 수 있다.On the other hand, the compound represented by A-Linker-A or A-Linker-B of the present invention binds to the target protein cerebrone as one A ligand moiety, and E3 as the other A or B ligand moiety. In combination with ubiquitin ligase, finally E3 ubiquitin ligase induces multiple ubiquitination to the target protein (cerebron), which is then probisome, e.g., 26S proteasome, multiubiquitinated protein ( Celebron) to achieve the effect of decomposing, and the pharmacological effect through the proteolysis is caused by actions related to all related diseases caused by Celebron, for example, abnormalities, hyperactivity, and the like of Celebron. Diseases, for example, cancer, for example, autoimmune diseases, for example, metabolic diseases, such as the conventional cerebron-related diseases, it can exhibit useful effects in all diseases identified.

특히, 본 발명의 화합물이 세포저해제, 표적저해제와 같은 종래 약의 부작용 문제, 내성 문제, 미비한 약리활성과 같은 문제점을 극복할 수 있는, 단백질 분해제(PROTAC) 약물이라는 점에서, 유용하다.In particular, the compound of the present invention is useful in that it is a protease drug (PROTAC) drug that can overcome problems such as side effects, resistance problems, and poor pharmacological activity of conventional drugs such as cell inhibitors and target inhibitors.

또한, 본 발명 화합물은 A로 표시되는 세레브론 결합 모이어티 자체가 면역조절약물(Immunomodulatory drug, IMiD)로 알려진 탈리도마이드, 레날리도마이드, 포말리도마이드, 이의 유사체, 이의 동배체, 이의 유도체인 바, 본 발명 화합물이 세레브론 단백질에 분해를 유도하기 전 단계로, 단순히 세레브론에 결합되는 것만으로도 면역조절약물로서의 약리효과를 나타낸다.In addition, the compound of the present invention is a thredomide, lenalidomide, pomalidomide, an analog thereof, an isomer thereof, a derivative thereof, the cerebrone-binding moiety represented by A itself known as an immunomodulatory drug (IMiD) , As a step before the compound of the present invention induces decomposition to a celebrative protein, it exhibits a pharmacological effect as an immunomodulatory drug simply by being bound to celebrative.

따라서, 본 발명 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염은, 종래 알려진 면역조절약물로서의 세레브론 저해 효과와, 종래 약물의 문제점을 극복할 수 있는 단백질 분해제로서의 약리 효과를 동시에 달성할 수 있는 바, 약리 활성에서 우수한 시너지 효과를 나타내고, 동시에 약의 내성, 부작용과 같은 문제점을 현저히 개선시킨다.Therefore, the compound represented by A-Linker-A or A-Linker-B of the present invention, its stereoisomer, or a pharmaceutically acceptable salt thereof, has a problem of inhibiting cerebrone as a known immunomodulatory drug and problems of conventional drugs. At the same time, the pharmacological effect as a proteolytic agent that can be overcome can be simultaneously achieved, exhibiting excellent synergistic effect in pharmacological activity, and at the same time, significantly improve the problems such as drug resistance and side effects.

본 발명에서는, 암, 자가면역질환 또는 대사질환에 있어서, 본 발명 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 약리활성은 매우 우수하며, 또한 본 발명 화합물은 세레브론 단백질 자체를 분해시키는 바, 기존의 표적치료제, 세포치료제 등의 단점, 내성문제 등을 효과적으로 해결할 수 있음을 실험적으로 확인하였다.In the present invention, in cancer, autoimmune disease or metabolic disease, the pharmacological activity of the compound represented by A-Linker-A or A-Linker-B of the present invention is very excellent, and the compound of the present invention is also used for the celebrity protein itself. As a result of decomposition, it has been experimentally confirmed that it can effectively solve disadvantages, resistance problems, etc. of existing targeted therapies and cell therapies.

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 세레브론(Cereblon) 관련 질환의 예방 또는 치료용 약학적 조성물을 제공한다.Furthermore, the present invention is to prevent or treat cereblon-related diseases containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient. It provides a pharmaceutical composition.

본 발명의 A-Linker-A 또는 A-Linker-B로 표시되는 화합물은 세레브론 단백질의 저해 활성을 나타냄과 동시에, 세레브론을 분해하는 바, 세레브론에 기인한 질환(세레브론 관련 질환), 세레브론 이상활성, 세레브론 과활성 또는 세레브론의 기작에 의해 유도되는 활성으로부터 야기되는 질환에 우수한 약리효과를 달성할 수 있다. 구체적인 예시의 질환으로, 암, 자가면역 질환, 대사질환의 치료에서, 본 발명 화합물은 예방, 개선을 위한 약학적 조성물의 유효성분으로 사용될 수 있다.The compound represented by A-Linker-A or A-Linker-B of the present invention exhibits inhibitory activity of a celebrative protein, and at the same time decomposes celebrative bar, a disease caused by celebratory (cerebron-related disease), It is possible to achieve excellent pharmacological effects on diseases resulting from celebrity abnormal activity, celebrative hyperactivity or activity induced by the mechanism of celebrity. As a specific exemplary disease, in the treatment of cancer, autoimmune disease, and metabolic disease, the compounds of the present invention can be used as an active ingredient of a pharmaceutical composition for prevention and improvement.

상술된 배경기술의 기재에서 확인되듯이, E3 유비퀴틴 리가제 중 세레브론(CRBN, Cereblon)은 면역조절약물(Immunomodulatory drug, IMiD)로 알려진 탈리도마이드, 레날리도마이드, 포말리도마이드 등이 결합하면, 전사요인(transcription factor)인 이카로스(ikaros), 에이오로스(aiolos)를 분해 시켜 우수한 항암 효과를 보일 뿐만 아니라, 루푸스를 포함하는 자가면역질환 질환에 효과가 있다. 본 발명 A-Linker-A 또는 A-Linker-B로 표시되는 화합물에서 A로 표시되는 부분은 탈리도마이드, 레날리도마이드, 포말리도마이드, 이의 유사체, 이으 동배체, 이의 유도체인 바, 동일하게 암 또는 자가면역질환에 우수한 효과를 나타내고, 또 다른 한편, 세레브론은 특정 단백질의 분해에 관여하는 E3 유비퀴틴 리가제 역할외에도, CRBN을 억제하였을 경우, 에너지센서인 AMPK가 활성화되어 비만, 지방간, 당뇨병 등 대사질환의 예방 또는 치료가 가능하다.As can be seen from the description of the background art described above, cereblon (CRBN, Cereblon) among E3 ubiquitin ligase is combined with thalidomide, lenalidomide, pomalidomide, etc., also known as an immunomodulatory drug (IMiD), It decomposes transcription factors (ikaros) and ikaros (aiolos), which not only show excellent anticancer effects, but are also effective in autoimmune disease diseases including lupus. In the compound represented by A-Linker-A or A-Linker-B of the present invention, the part represented by A is thalidomide, lenalidomide, pomalidomide, analogs thereof, homoploids, derivatives thereof, and the same cancer Or, it has an excellent effect on autoimmune diseases, and on the other hand, in addition to the role of E3 ubiquitin ligase, which is involved in the decomposition of specific proteins, when the CRBN is suppressed, the energy sensor AMPK is activated, resulting in obesity, fatty liver, diabetes, etc. Prevention or treatment of metabolic diseases is possible.

나아가, 면역세포(T-세포)에 CRBN을 억제 또는 분해하면 T세포에 과발현 되어 있는 CRBN의 양을 낮추어 T세포의 활성을 증가시키는 효과를 유도하여 항암면역치료제로 사용 가능하다.Furthermore, when CRBN is suppressed or degraded to immune cells (T-cells), the amount of CRBN overexpressed in T cells is lowered to induce the effect of increasing the activity of T cells, and thus it can be used as an anticancer immunotherapy agent.

또한, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating cancer containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient. do.

이때, 상기 암은 세레브론 활성을 억제 또는 세레브론 단백질을 분해하는 것으로부터 호전될 수 있는 모든 암 질환을 말하며, 비제한적인 예로, 상기 암은 가성점액종, 간내 담도암, 간모세포종, 간암, 갑상선암, 결장암, 고환암, 골수이형성증후군, 교모세포종, 구강암, 구순암, 균상식육종, 급성골수성백혈병, 급성림프구성백혈병, 기저세포암, 난소상피암, 난소생식세포암, 남성유방암, 뇌암, 뇌하수체선종, 다발성골수종, 담낭암, 담도암, 대장암, 만성골수성백혈병, 만성림프구백혈병, 망막모세포종, 맥락막흑색종, 미만성거대B세포림프종, 바터팽대부암, 방광암, 복막암, 부갑상선암, 부신암, 비부비동암, 비소세포폐암, 비호지킨림프종, 설암, 성상세포종, 소세포폐암, 소아뇌암, 소아림프종, 소아백혈병, 소장암, 수막종, 식도암, 신경교종, 신경모세포종, 신우암, 신장암, 심장암, 십이지장암, 악성 연부조직 암, 악성골암, 악성림프종, 악성중피종, 악성흑색종, 안암, 외음부암, 요관암, 요도암, 원발부위불명암, 위림프종, 위암, 위유암종, 위장관간질암, 윌름스암, 유방암, 육종, 음경암, 인두암, 임신융모질환, 자궁경부암, 자궁내막암, 자궁육종, 전립선암, 전이성 골암, 전이성뇌암, 종격동암, 직장암, 직장유암종, 질암, 척수암, 청신경초종, 췌장암, 침샘암, 카포시 육종, 파제트병, 편도암, 편평상피세포암, 폐선암, 폐암, 폐편평상피세포암, 피부암, 항문암, 횡문근육종, 후두암, 흉막암, 및 흉선암으로 이루어진 군으로부터 선택되는 1종 이상일 수 있다.At this time, the cancer refers to all cancer diseases that can be improved from inhibiting cerebron activity or decomposing the cerebron protein, and for non-limiting examples, the cancer is pseudomyxoma, intrahepatic biliary cancer, hepatoblastoma, liver cancer, thyroid cancer , Colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, cleft carcinoma, mycosis mycosis, acute myelogenous leukemia, acute lymphocytic leukemia, basal cell carcinoma, ovarian epithelial cancer, ovarian reproductive cell cancer, male breast cancer, brain cancer, pituitary adenoma, Multiple myeloma, gallbladder cancer, biliary tract cancer, colorectal cancer, chronic myelogenous leukemia, chronic lymphocytic leukemia, retinoblastoma, choroidal melanoma, diffuse giant B cell lymphoma, barter swelling cancer, bladder cancer, peritoneal cancer, parathyroid cancer, adrenal cancer, non-sinusoidal cancer , Non-small cell lung cancer, non-Hodgkin's lymphoma, snow cancer, astrocytoma, small cell lung cancer, pediatric brain cancer, pediatric lymphoma, pediatric leukemia, small intestine cancer, meningioma, esophageal cancer, glioma, neuroblastoma, renal cancer, kidney cancer, heart cancer, duodenal cancer, Malignant soft tissue cancer, malignant bone cancer, malignant lymphoma, malignant mesothelioma, malignant melanoma, eye cancer, vulvar cancer, ureteral cancer, urethral cancer, primary cancer, gastric lymphoma, gastric cancer, gastric carcinoma, gastrointestinal interstitial cancer, Wilms' cancer, breast cancer , Sarcoma, penis cancer, pharyngeal cancer, chorionic disease, cervical cancer, endometrial cancer, uterine sarcoma, prostate cancer, metastatic bone cancer, metastatic brain cancer, mediastinal cancer, rectal cancer, rectal carcinoma, vaginal cancer, spinal cord cancer, auditory nerve sheath, pancreatic cancer, Selected from the group consisting of salivary gland cancer, Kaposi's sarcoma, Paget's disease, tonsil cancer, squamous cell carcinoma, lung adenocarcinoma, lung cancer, pulmonary squamous cell carcinoma, skin cancer, anal cancer, rhabdomyosarcoma, larynx cancer, pleural cancer, and thymic cancer It may be one or more.

나아가, 바람직하게 상기 암은 급성 골수 백혈병(AML); 만성적 골수성 백혈병(CML); 급성 림프아구성 백혈병(ALL); 만성적 림프구성 백혈병(CLL); 호지킨 질환(HD); 비-호지킨 림프종(NHL); B-세포 림프종; T-세포 림프종; 다발성 골수종(MM); 아밀로이드증; 발덴스트롬 거대글로불린혈증; 골수이형성 증후군(MDS); 작은 림프구 림프종(SLL); 변연부 림프종; 무증상 다발성 골수종; 및 골수증식성 증후군으로 이루어진 군으로부터 선택되는 1종 이상일 수 있다.Furthermore, preferably the cancer is acute myeloid leukemia (AML); Chronic myelogenous leukemia (CML); Acute lymphoblastic leukemia (ALL); Chronic lymphocytic leukemia (CLL); Hodgkin's disease (HD); Non-Hodgkin's lymphoma (NHL); B-cell lymphoma; T-cell lymphoma; Multiple myeloma (MM); Amyloidosis; Waldenstrom megaglobulinemia; Myelodysplastic syndrome (MDS); Small lymphocyte lymphoma (SLL); Marginal zone lymphoma; Asymptomatic multiple myeloma; And it may be one or more selected from the group consisting of myeloproliferative syndrome.

본 명세서에서 사용된 바와 같이, 용어 "암"은 조절되지 않는 또는 이상조절된 세포 증식, 감소된 세포성 분화, 주위의 조직에 침입하는 부절적한 능력, 및/또는 이소성 부위에서 신규 성장을 확립하는 능력을 특징으로 하는 세포성 장애를 의미한다. 용어 "암"은 비제한적으로, 고형 종양 및 혈액매개 종양 (혈액성 악성종양)을 포함한다. 용어 "암"은 피부, 조직, 기관, 골, 연골, 혈액, 및 혈관의 질환을 포함한다. 용어 "암"은 추가로, 1차 및 전이암을 포함한다.As used herein, the term “cancer” establishes unregulated or dysregulated cell proliferation, reduced cellular differentiation, inadequate ability to invade surrounding tissue, and/or establishes new growth at ectopic sites. Refers to a cellular disorder characterized by the ability to. The term "cancer" includes, but is not limited to, solid tumors and blood-borne tumors (hematologic malignancy). The term "cancer" includes diseases of the skin, tissues, organs, bone, cartilage, blood, and blood vessels. The term "cancer" further includes primary and metastatic cancer.

상기 고형 종양은 췌장암; 침습성 방광암 포함하는 방광암; 결장직장암; 갑상선암, 위암, 전이성 유방암을 포함하는 유방암; 안드로겐-의존적 및 안드로겐-독립적인 전립선암을 포함하는 전립선암; 예를 들면, 전이성 신장 세포 암종을 포함하는 신장암; 예를 들면 간세포 암 및 간내 담관을 포함하는 간암; 비-소세포 폐암 (NSCLC), 편평상피 폐암, 세기관지폐포 암종 (BAC), 폐의 선암종, 및 소세포 폐암 (SCLC)을 포함하는 폐 및 기관지 암; 예를 들면, 진행성 상피성 또는 1차 복막 암을 포함하는 난소암; 자궁경부암; 예를 들면 자궁 체부 및 자궁 경부를 포함하는 자궁암; 자궁내막 암; 위암; 식도암; 예를 들면, 두경부의 편평상피 세포 암종, 비인두 암, 구강 및 인두를 포함하는 두경부 암; 흑색종; 전이성 신경내분비 종양을 포함하는 신경내분비 암; 예를 들면, 신경아교종/교모세포종, 역형성 희소돌기아교세포종, 성인 교모세포종 다형성, 및 성인 역형성 별아교세포종을 포함하는 뇌암; 전이성 신경내분비 종양; 골 암이고; 그리고 연조직 육종을 포함하는 신경내분비를 포함한다.The solid tumor is pancreatic cancer; Bladder cancer, including invasive bladder cancer; Colorectal cancer; Breast cancer, including thyroid cancer, stomach cancer, and metastatic breast cancer; Prostate cancer, including androgen-dependent and androgen-independent prostate cancer; Kidney cancer, including, for example, metastatic kidney cell carcinoma; Liver cancer, including, for example, hepatocellular carcinoma and intrahepatic bile ducts; Lung and bronchial cancers including non-small cell lung cancer (NSCLC), squamous epithelial lung cancer, bronchoalveolar carcinoma (BAC), adenocarcinoma of the lung, and small cell lung cancer (SCLC); Ovarian cancer, including, for example, advanced epithelial or primary peritoneal cancer; Cervical cancer; Uterine cancer including, for example, the uterine body and cervix; Endometrial cancer; Stomach cancer; Esophageal cancer; Head and neck cancers including, for example, squamous cell carcinoma of the head and neck, nasopharyngeal cancer, oral cavity and pharynx; Melanoma; Neuroendocrine cancer, including metastatic neuroendocrine tumors; Brain cancers, including, for example, glioma/glioblastoma, anaplastic oligodendrocyte glioma, adult glioblastoma polymorphism, and adult anaplastic astrocytoma; Metastatic neuroendocrine tumor; Bone cancer; And neuroendocrine, including soft tissue sarcoma.

상기 혈액성 악성종양은 급성 골수 백혈병 (AML); 만성적 골수성 백혈병 (CML) (가속화된 CML 및 CML 아세포기 (CML-BP) 포함); 급성 림프아구성 백혈병 (ALL); 만성적 림프구성 백혈병 (CLL); 호지킨 질환 (HD); 비-호지킨 림프종 (NHL) (여포성 림프종 및 외투 세포 림프종 포함); B-세포 림프종 (미만성 큰 B-세포 림프종 (DLBCL) 포함); T-세포 림프종; 다발성 골수종 (MM); 아밀로이드증; 발덴스트롬 거대글로불린혈증; 골수이형성 증후군 (MDS) (난치의 빈혈 (RA), 관상 철아구 (RARS)를 갖는 난치의 빈혈 (과다 모세포 (RAEB), 및 형질전환 동반 RAEB (RAEB-T)를 갖는 난치의 빈혈) 포함); 작은 림프구 림프종 (SLL); 변연부 림프종; 무증상 다발성 골수종; 및 골수증식성 증후군을 포함한다.The hematologic malignancy is acute myeloid leukemia (AML); Chronic myelogenous leukemia (CML) (including accelerated CML and CML subcellular phase (CML-BP)); Acute lymphoblastic leukemia (ALL); Chronic lymphocytic leukemia (CLL); Hodgkin's disease (HD); Non-Hodgkin's lymphoma (NHL) (including follicular lymphoma and mantle cell lymphoma); B-cell lymphoma (including diffuse large B-cell lymphoma (DLBCL)); T-cell lymphoma; Multiple myeloma (MM); Amyloidosis; Waldenstrom megaglobulinemia; Myelodysplastic syndrome (MDS) (including refractory anemia (RA), refractory anemia with coronary blasts (RARS) (excessive blast cells (RAEB), and refractory anemia with RAEB (RAEB-T) with transformation) ; Small lymphocyte lymphoma (SLL); Marginal zone lymphoma; Asymptomatic multiple myeloma; And myeloproliferative syndrome.

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 자가면역질환의 예방 또는 치료용 약학적 조성물을 제공한다.Furthermore, the present invention is a pharmaceutical composition for the prevention or treatment of autoimmune diseases containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient. Gives

이때, 상기 자가면역질환은 세레브론 활성을 억제 또는 세레브론 단백질을 분해하는 것으로부터 호전될 수 있는 모든 자가면역질환을 말하며, 비제한적인 예로, 상기 자가면역질환은 면역성 호중구감소증, 길랑바레 신드롬, 간질, 자가면역 뇌염, 아이삭 신드롬, 모반 신드롬, 심상성천포창, 낙엽성천포창, 수포성류천포창, 후천성표피수포증, 임신성 유사수포창, 뮤코스 막 유사수포창, 항인지질 신드롬, 자가면역 빈혈, 자가면역 그래이브 병, 굿파스튜어 증후군, 중증근무력증, 다발성경화증, 류마티스성 관절염, 루프스, 특발성 혈소판 감소성 자반증(idiopathic thrombocytopenic purpura, ITP), 루프스 신염 및 막성 신병증으로 구성된 군에서 선택되는 하나 이상일 수 있다.At this time, the autoimmune disease refers to all autoimmune diseases that can be improved from inhibiting cerebron activity or decomposing the cerebron protein, and for non-limiting examples, the autoimmune disease is immune neutropenia, Guillain-Barre syndrome, Epilepsy, autoimmune encephalitis, Isaac syndrome, birthmark syndrome, vulgar vesicular vesicle, deciduous celestial vesicle, vesicular vesicular vesicles, acquired epidermal vesicles, gestational pseudovesicles, mucose membrane-like vesicles, antiphospholipid syndrome, autoimmune anemia, autologous It may be one or more selected from the group consisting of immune grave disease, Goodpasture's syndrome, myasthenia gravis, multiple sclerosis, rheumatoid arthritis, lupus, idiopathic thrombocytopenic purpura (ITP), lupus nephritis and membranous nephropathy .

또한, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 대사질환의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for the prevention or treatment of metabolic diseases containing the compound represented by the A-Linker-A or A-Linker-B, a stereoisomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient. to provide.

상기 대사질환은, 세레브론을 억제 또는 세레브론 단백질의 분해로부터 치료 또는 예방의 효과가 달성될 수 있는 모든 대사질환을 말하며, 비제한적인 예로, 비만, Ⅰ형 당뇨병, Ⅱ형 당뇨병, 부적합한 내당력, 인슐린 내성, 고인슐린혈증, 고혈당증, 이상지질혈증, 고지질혈증, 고중성지방혈증, 고콜레스테롤혈증, 이상지질혈증, 대사 증후군 X, 혈관 질환, 죽상경화증, 관상심장질환, 대뇌혈관질환, 심부전증, 말초혈관질환, 및 반흔에서 선택되는 1종 이상일 수 있다.The metabolic disease refers to all metabolic diseases in which the effect of treatment or prevention can be achieved by inhibiting cerebron or decomposing cerebron protein, and non-limiting examples include obesity, type I diabetes, type II diabetes, and inappropriate history , Insulin resistance, hyperinsulinemia, hyperglycemia, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, metabolic syndrome X, vascular disease, atherosclerosis, coronary heart disease, cerebrovascular disease, heart failure , Peripheral vascular disease, and scars.

상술된 본 발명에 따른 약학적 조성물에 있어서, 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염은 임상 투여시에 경구 및 비경구의 여러 가지 제형으로 투여될 수 있는데, 제제화할 경우에는 보통 사용하는 충전제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 제조될 수 있다.In the pharmaceutical composition according to the present invention described above, the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof may be administered in various oral and parenteral forms during clinical administration. It can be administered in the form of an eggplant, and when formulated, it can be prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrating agents, and surfactants.

경구 투여용 제형으로는 예를 들면 정제, 환제, 경/연질 캅셀제, 액제, 현탁제, 유화제, 시럽제, 과립제, 엘릭시르제, 트로키제 등이 있는데, 이들 제형은 유효성분 이외에 희석제(예: 락토즈, 덱스트로즈, 수크로즈, 만니톨, 솔비톨, 셀룰로즈 및/또는 글리신), 활택제(예: 실리카, 탈크, 스테아르산 및 그의 마그네슘 또는 칼슘염 및/또는 폴리에틸렌 글리콜)를 함유하고 있다. 정제는 마그네슘 알루미늄 실리케이트, 전분 페이스트, 젤라틴, 메틸셀룰로즈, 나트륨 카복시메틸셀룰로즈 및/또는 폴리비닐피롤리딘 등과 같은 결합제를 함유할 수 있으며, 경우에 따라 전분, 한천, 알긴산 또는 그의 나트륨 염 등과 같은 붕해제 또는 비등 혼합물 및/또는 흡수제, 착색제, 향미제, 및 감미제를 함유할 수 있다.Formulations for oral administration include, for example, tablets, pills, hard/soft capsules, liquids, suspensions, emulsifiers, syrups, granules, elixirs, troches, etc. , Dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine), lubricants (eg silica, talc, stearic acid and its magnesium or calcium salts and/or polyethylene glycols). Tablets may contain a binder such as magnesium aluminum silicate, starch paste, gelatin, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidine, and, if desired, a boron such as starch, agar, alginic acid or its sodium salt, etc. It can contain an releasing or boiling mixture and/or absorbent, colorant, flavoring agent, and sweetening agent.

상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물을 유효 성분으로 하는 약학적 조성물은 비경구 투여할 수 있으며, 비경구 투여는 피하주사, 정맥주사, 근육 내 주사 또는 흉부 내 주사를 주입하는 방법에 의하였다. The pharmaceutical composition containing the compound represented by A-Linker-A or A-Linker-B as an active ingredient may be administered parenterally, and parenteral administration may include subcutaneous injection, intravenous injection, intramuscular injection, or intrathoracic injection. By injection method.

이때, 비경구 투여용 제형으로 제제화하기 위하여 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물 또는 이의 약학적으로 허용 가능한 염을 안정제 또는 완충제와 함께 물에 혼합하여 용액 또는 현탁액으로 제조하고, 이를 앰플 또는 바이알 단위 투여형으로 제조할 수 있다. 상기 조성물은 멸균되고/되거나 방부제, 안정화제, 수화제 또는 유화 촉진제, 삼투압 조절을 위한 염 및/또는 완충제 등의 보조제, 및 기타 치료적으로 유용한 물질을 함유할 수 있으며, 통상적인 방법인 혼합, 과립화 또는 코팅 방법에 따라 제제화할 수 있다.At this time, the compound represented by the A-Linker-A or A-Linker-B or a pharmaceutically acceptable salt thereof is mixed with water together with a stabilizer or buffer to prepare a solution or suspension for formulation into a formulation for parenteral administration. And it can be prepared in ampule or vial unit dosage form. The composition may be sterile and/or contain preservatives, stabilizers, hydrating or emulsifying accelerators, adjuvants such as salts and/or buffers for osmotic pressure control, and other therapeutically useful substances, conventional methods of mixing, granulation It can be formulated according to the chemical or coating method.

상기 제제화의 예시는 통상적인 제제예에 관한 것일 뿐, 본 발명의 제제화가 이에 제한되지는 않음을 통상의 기술지식을 가진 자라면 용이하게 이해할 수 있다.The example of the formulation is related to the conventional formulation example, it can be easily understood by those of ordinary skill in the art that the formulation of the present invention is not limited thereto.

나아가, 본 발명의 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물 또는 이의 약학적으로 허용 가능한 염의 인체에 대한 투여량은 대상의 나이, 몸무게, 성별, 투여형태, 건강상태 및 질환 정도에 따라 달라질 수 있으며, 몸무게가 70Kg인 성인 대상를 기준으로 할 때, 일반적으로 0.1-1000 mg/일이며, 바람직하게는 1-500 mg/일이며, 또한 의사 또는 약사의 판단에 따라 일정시간 간격으로 1일 1회 내지 수회로 분할 투여할 수도 있다.Furthermore, the dosage of the compound represented by the A-Linker-A or A-Linker-B of the present invention or a pharmaceutically acceptable salt thereof to the human body is the age, weight, sex, dosage form, health condition and disease of the subject. It may vary depending on the degree, and based on an adult subject having a weight of 70 Kg, it is generally 0.1-1000 mg/day, preferably 1-500 mg/day, and also a predetermined time interval according to the judgment of a doctor or pharmacist It can also be administered in divided doses once to several times a day.

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 세레브론(Cereblon) 관련 질환의 예방 또는 개선용 건강기능식품 조성물을 제공한다.Furthermore, the present invention prevents or improves cereblon-related diseases containing the compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof as an active ingredient. It provides a health functional food composition.

이때, 상기 세레브론 관련 질환은 세레브론에 기인한 질환(세레브론 관련 질환), 세레브론 이상활성, 세레브론 과활성 또는 세레브론의 기작에 의해 유도되는 활성으로부터 야기되는 질환으로, 비제한적인 예로, 암, 자가면역질환, 및 대사질환이다.At this time, the cerebron-related disease is a disease caused by cerebron-related disease (cerebron-related disease), cerebron abnormal activity, cerebron hyperactivity or activity induced by the mechanism of cerebrone, and is not limited to , Cancer, autoimmune diseases, and metabolic diseases.

상기 암은 세레브론 활성을 억제 또는 세레브론 단백질을 분해하는 것으로부터 호전될 수 있는 모든 암 질환을 말하며, 비제한적인 예로, 상기 암은 가성점액종, 간내 담도암, 간모세포종, 간암, 갑상선암, 결장암, 고환암, 골수이형성증후군, 교모세포종, 구강암, 구순암, 균상식육종, 급성골수성백혈병, 급성림프구성백혈병, 기저세포암, 난소상피암, 난소생식세포암, 남성유방암, 뇌암, 뇌하수체선종, 다발성골수종, 담낭암, 담도암, 대장암, 만성골수성백혈병, 만성림프구백혈병, 망막모세포종, 맥락막흑색종, 미만성거대B세포림프종, 바터팽대부암, 방광암, 복막암, 부갑상선암, 부신암, 비부비동암, 비소세포폐암, 비호지킨림프종, 설암, 성상세포종, 소세포폐암, 소아뇌암, 소아림프종, 소아백혈병, 소장암, 수막종, 식도암, 신경교종, 신경모세포종, 신우암, 신장암, 심장암, 십이지장암, 악성 연부조직 암, 악성골암, 악성림프종, 악성중피종, 악성흑색종, 안암, 외음부암, 요관암, 요도암, 원발부위불명암, 위림프종, 위암, 위유암종, 위장관간질암, 윌름스암, 유방암, 육종, 음경암, 인두암, 임신융모질환, 자궁경부암, 자궁내막암, 자궁육종, 전립선암, 전이성 골암, 전이성뇌암, 종격동암, 직장암, 직장유암종, 질암, 척수암, 청신경초종, 췌장암, 침샘암, 카포시 육종, 파제트병, 편도암, 편평상피세포암, 폐선암, 폐암, 폐편평상피세포암, 피부암, 항문암, 횡문근육종, 후두암, 흉막암, 및 흉선암으로 이루어진 군으로부터 선택되는 1종 이상일 수 있다.The cancer refers to all cancer diseases that can be improved by inhibiting cerebron activity or decomposing the cerebron protein, and for non-limiting examples, the cancer is pseudomyxoma, intrahepatic biliary cancer, hepatoblastoma, liver cancer, thyroid cancer, colon cancer , Testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, celiac cancer, mycosis, acute myeloid leukemia, acute lymphocytic leukemia, basal cell carcinoma, ovarian epithelial cancer, ovarian reproductive cell cancer, male breast cancer, brain cancer, pituitary adenoma, multiple myeloma , Gallbladder cancer, Biliary tract cancer, Colon cancer, Chronic myelogenous leukemia, Chronic lymphocytic leukemia, Retinoblastoma, Choroid melanoma, Diffuse giant B cell lymphoma, Barter swelling cancer, Bladder cancer, Peritoneal cancer, Parathyroid cancer, Adrenal cancer, Non sinus cancer, Arsenic Cell lung cancer, non-Hodgkin's lymphoma, snow cancer, astrocytoma, small cell lung cancer, pediatric brain cancer, pediatric lymphoma, pediatric leukemia, small intestine cancer, meningioma, esophageal cancer, glioma, neuroblastoma, renal cancer, kidney cancer, heart cancer, duodenal cancer, malignant softening Tissue cancer, malignant bone cancer, malignant lymphoma, malignant mesothelioma, malignant melanoma, eye cancer, vulvar cancer, ureteral cancer, urethral cancer, primary site unknown cancer, gastric lymphoma, gastric cancer, gastric carcinoma, gastrointestinal stromal cancer, Wilms' cancer, breast cancer, sarcoma , Penile cancer, pharyngeal cancer, chorionic villus disease, cervical cancer, endometrial cancer, uterine sarcoma, prostate cancer, metastatic bone cancer, metastatic brain cancer, mediastinal cancer, rectal cancer, rectal carcinoma, vaginal cancer, spinal cord cancer, auditory choroidoma, pancreatic cancer, salivary gland cancer , Kaposi's sarcoma, Paget's disease, tonsil cancer, squamous cell carcinoma, lung adenocarcinoma, lung cancer, lung squamous cell carcinoma, skin cancer, anal cancer, rhabdomyosarcoma, laryngeal cancer, pleural cancer, and thymic cancer 1 It may be more than a species.

상기 자가면역질환은 세레브론 활성을 억제 또는 세레브론 단백질을 분해하는 것으로부터 호전될 수 있는 모든 자가면역질환을 말하며, 비제한적인 예로, 상기 자가면역질환은 면역성 호중구감소증, 길랑바레 신드롬, 간질, 자가면역 뇌염, 아이삭 신드롬, 모반 신드롬, 심상성천포창, 낙엽성천포창, 수포성류천포창, 후천성표피수포증, 임신성 유사수포창, 뮤코스 막 유사수포창, 항인지질 신드롬, 자가면역 빈혈, 자가면역 그래이브 병, 굿파스튜어 증후군, 중증근무력증, 다발성경화증, 류마티스성 관절염, 루프스, 특발성 혈소판 감소성 자반증(idiopathic thrombocytopenic purpura, ITP), 루프스 신염 및 막성 신병증으로 구성된 군에서 선택되는 하나 이상일 수 있다. The autoimmune disease refers to all autoimmune diseases that can be improved from inhibiting cerebron activity or decomposing the cerebron protein, and for non-limiting examples, the autoimmune disease is immune neutropenia, Guillain Barre syndrome, epilepsy, Autoimmune encephalitis, Isaac syndrome, birthmark syndrome, vulgar vesicular vesicles, deciduous celestial vesicles, vesicular vesicular vesicles, acquired epithelial vesicles, gestational pseudo blisters, mucos membrane pseudo blisters, antiphospholipid syndromes, autoimmune anemia, autoimmune yeah It may be one or more selected from the group consisting of Eve's disease, Goodpasture's syndrome, myasthenia gravis, multiple sclerosis, rheumatoid arthritis, lupus, idiopathic thrombocytopenic purpura (ITP), lupus nephritis and membranous nephropathy.

상기 대사질환은, 세레브론을 억제 또는 세레브론 단백질의 분해로부터 치료 또는 예방의 효과가 달성될 수 있는 모든 대사질환을 말하며, 비제한적인 예로, 비만, Ⅰ형 당뇨병, Ⅱ형 당뇨병, 부적합한 내당력, 인슐린 내성, 고인슐린혈증, 고혈당증, 이상지질혈증, 고지질혈증, 고중성지방혈증, 고콜레스테롤혈증, 이상지질혈증, 대사 증후군 X, 혈관 질환, 죽상경화증, 관상심장질환, 대뇌혈관질환, 심부전증, 말초혈관질환, 및 반흔에서 선택되는 1종 이상일 수 있다.The metabolic disease refers to all metabolic diseases in which the effect of treatment or prevention can be achieved by inhibiting cerebron or decomposing cerebron protein, and non-limiting examples include obesity, type I diabetes, type II diabetes, and inappropriate history , Insulin resistance, hyperinsulinemia, hyperglycemia, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, metabolic syndrome X, vascular disease, atherosclerosis, coronary heart disease, cerebrovascular disease, heart failure , Peripheral vascular disease, and scars.

나아가, 본 발명은 상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 치료학적으로 유효한 양으로 대상(subject)에게 투여하는 단계를 포함하는 세레브론(Cereblon) 관련 질환의 치료 방법을 제공한다.Furthermore, the present invention includes the step of administering a compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof or a pharmaceutically acceptable salt thereof to a subject in a therapeutically effective amount. It provides a method for treating cereblon-related diseases.

이때, 상기 세레브론(Cereblon) 관련 질환은 본 명세서에서 설명된 바와 같이, 세레브론(Cereblon) 활성을 저해 또는 세레브론(Cereblon) 단백질을 분해하는 것으로부터 예방 또는 치료할 수 있는 질환을 말하고, 바람직하게 암, 자가면역질환 또는 대사질환이다.At this time, the cereblon-related disease refers to a disease that can be prevented or treated from inhibiting cereblon activity or decomposing a cereblon protein, as described herein, preferably Cancer, autoimmune disease or metabolic disease.

상기 치료학적 유효량은 투여 방법에 따라, 체내로 투여시 대상(subject)의 증상 또는 상태를 치료, 예방 또는 개선시킬 수 있을 정도의 양을 말한다. 또한 상기 양은 투여하는 대상의 체중, 나이, 성별, 상태, 가족력에 따라 상이할 수 있고, 본 발명에서 상기 치료 방법은 이처럼 대상별로 상이한 조건에 따라 다른 양의 투여량을 정할 수 있다.The therapeutically effective amount refers to an amount sufficient to treat, prevent or improve the symptoms or condition of a subject when administered into the body, depending on the administration method. In addition, the amount may vary depending on the weight, age, sex, condition, and family history of the subject to be administered, and in the present invention, the treatment method may determine a different amount of dose according to different conditions for each subject.

상기 "유효한 양"은 세레브론(Cereblon) 관련 질환, 예를 들어 암, 자가면역질환 또는 대사질환을 치료하는데 유효한 양이다. 다른 구체예에서, 화합물의 "유효한 양"은 세레브론(Cereblon) 활성을 억제 또는 세레브론 단백질을 분해시킬수 있는 양이다.The "effective amount" is an amount effective to treat cereblon-related diseases, such as cancer, autoimmune diseases or metabolic diseases. In another embodiment, the “effective amount” of the compound is an amount that can inhibit Cereblon activity or degrade the celebrity protein.

본 발명의 방법에 따른 화합물 및 조성물은 질환을 치료하는데 유효한 임의의 양 및 임의의 투여 경로를 사용하여 투여될 수 있다. 필요한 정확한 양은 대상(subject)의 종, 연령, 및 일반적인 병태, 감염의 중증도, 특정한 제제, 그것의 투여 방식, 등에 따라 대상별로 변할 것이다. 본 발명의 화합물은 복용량의 투여 용이성 및 균일성에 대해 투약량 단위 형태로 빈번하게 제형화된다. 표현 "투약량 단위 형태"는, 본 명세서에서 사용된 바와 같이 치료될 대상에 적절한 제제의 물리적으로 별개의 단위를 의미한다. 또한, 본 발명의 화합물 및 조성물의 총 일일 사용량이 건전한 의료 판단의 범위 내에 주치의에 의해 결정될 것으로 이해될 것이다. 임의의 특정한 대상 또는 유기체에 대한 특정 유효한 투여 수준은 하기를 포함하는 다양한 인자에 의존된다: 치료될 질환 및 질환의 중증도; 이용된 특정 화합물의 활성; 특정 조성물 이용된; 연령, 체중, 일반적인 건강, 대상의 성별 및 다이어트; 투여 시간, 투여 경로, 및 이용된 특정 화합물의 배출 속도; 치료의 지속시간; 이용된 특정 화합물 단독 또는 공동투여된 약물, 및 의료 기술에서 잘 알려진 이 외의 요인.The compounds and compositions according to the methods of the present invention can be administered using any amount and any route of administration effective for treating a disease. The exact amount required will vary from subject to subject, depending on the subject's species, age, and general condition, severity of infection, specific agent, mode of administration, and the like. The compounds of the present invention are frequently formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein means a physically discrete unit of agent suitable for the subject to be treated. It will also be understood that the total daily use of the compounds and compositions of the present invention will be determined by the attending physician within the scope of sound medical judgment. The specific effective dosage level for any particular subject or organism depends on various factors, including: the disease to be treated and the severity of the disease; The activity of the specific compound employed; Specific composition used; Age, weight, general health, subject's gender and diet; The time of administration, route of administration, and rate of excretion of the specific compound employed; Duration of treatment; The specific compound employed alone or co-administered drugs, and other factors well known in medical technology.

용어 "대상(subject)"은, 본 명세서에서 사용된 바와 같이, 동물, 예를 들면 포유동물, 예컨대 인간을 의미한다.The term "subject" as used herein means an animal, for example a mammal, such as a human.

이하, 본 발명을 실시예 및 실험예에 의하여 상세히 설명한다.Hereinafter, the present invention will be described in detail by examples and experimental examples.

단, 하기 실시예 및 실험예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 이에 한정되는 것은 아니다.However, the following examples and experimental examples are merely illustrative of the present invention, and the contents of the present invention are not limited thereto.

<실시예 1> (2S,4R)-1-((S)-2-(7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헵탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 1> (2S,4R)-1-((S)-2-(7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-ylamino)heptanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carbox Preparation of amide

Figure 112018098559920-pat00046
Figure 112018098559920-pat00046

단계 1: 7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헵탄산의 제조Step 1: Preparation of 7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-ylamino)heptanoic acid

DMF에 7-아미노헵탄산(158 mg, 1.09 mmol)과 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온(300 mg, 1.09 mmol)을 녹인 후, DIPEA(0.285 mL, 1.64 mmol)를 넣고 실온에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 10% 28 분)를 이용하여 분리, 정제하여 7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헵탄산(118 mg, 27%, 황색 고체)을 얻었다.7-aminoheptanoic acid (158 mg, 1.09 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-1,3-dione (300 mg, 1.09 mmol) in DMF ) Was dissolved, DIPEA (0.285 mL, 1.64 mmol) was added and stirred at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 10% 28 min) to 7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)heptanoic acid (118 mg, 27%, yellow solid) was obtained.

LC/MS (ESI) m/z [M+H]+ : 401.9, [M-H]- : 399.9LC/MS (ESI) m/z [M+H] + : 401.9, [MH] - : 399.9

1H NMR (CDCl3, 300MHz) δ 8.34 (s, 1H), 7.49 (dd, J = 8.5, 7.2 Hz, 1H), 7.09 (d, J = 7.1 Hz, 1H), 6.88 (d, J = 8.6 Hz, 1H), 6.29-6.18 (m, 1H), 4.96-4.87 (m, 1H), 3.32-3.22 (m, 2H), 2.94-2.65 (m, 3H), 2.36 (t, J = 7.3 Hz, 2H), 2.19-2.09 (m, 1H), 1.75-1.58 (m, 4H), 1.43 (t, J = 3.5 Hz, 4H). 1 H NMR (CDCl 3 , 300 MHz) δ 8.34 (s, 1H), 7.49 (dd, J = 8.5, 7.2 Hz, 1H), 7.09 (d, J = 7.1 Hz, 1H), 6.88 (d, J = 8.6 Hz, 1H), 6.29-6.18 (m, 1H), 4.96-4.87 (m, 1H), 3.32-3.22 (m, 2H), 2.94-2.65 (m, 3H), 2.36 (t, J = 7.3 Hz, 2H), 2.19-2.09 (m, 1H), 1.75-1.58 (m, 4H), 1.43 (t, J = 3.5 Hz, 4H).

단계 2: (2S,4R)-1-((S)-2-(7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헵탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 2: (2S,4R)-1-((S)-2-(7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Of 1-amino)heptanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide Produce

DMF에 상기 단계 1에서 제조한 화합물(30 mg, 0.074 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(35 mg, 0.074 mmol), HATU (43 mg, 0.112 mmol)을 녹이고 DIPEA(0.052 mL, 0.299 mmol)를 가한 뒤 실온에서 교반한다. 반응 종결 후, EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6% 30 min)를 이용하여 분리, 정제하여 (2S,4R)-1-((S)-2-(7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헵탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드(10 mg, 17%, 황색 고체)를 얻었다.Compound prepared in step 1 above in DMF (30 mg, 0.074 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N- (4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (35 mg, 0.074 mmol), HATU (43 mg, 0.112 mmol) dissolved and DIPEA (0.052 mL, 0.299) mmol) and stirred at room temperature. After completion of the reaction, dilute with EA and water, extract the organic layer, wash with brine, remove water with MgSO 4 and concentrate under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 6% 30 min) (2S,4R)-1-((S)-2-(7-(2-(2,6-dioxophyte) Peridin-3-yl)-1,3-dioxoisoindolin-4-ylamino)heptanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methyl Thiazole-5-yl)benzyl)pyrrolidine-2-carboxamide (10 mg, 17%, yellow solid) was obtained.

LC/MS (ESI) m/z [M+H]+ : 814.6, [M-H]- : 812.7LC/MS (ESI) m/z [M+H] + : 814.6, [MH] - : 812.7

1H NMR (CDCl3, 300MHz) δ 10.00 (s, 1H), 8.68 (s, 1H), 7.61-7.53 (m, 1H), 7.47 (dd, J = 8.5, 7.1 Hz, 1H), 7.35 (S,4H), 7.08 (d, J = 7.1 Hz, 1H), 6.87 (d, J = 8.5 Hz, 1H), 6.72 (d, J = 9.1 Hz, 1H), 6.24 (t, J = 5.7 Hz, 1H), 4.89-4.81 (m, 1H), 4.65-4.54 (m, 4H), 4.32-4.23 (m, 1H), 4.06 (d, J = 11.3 Hz, 1H), 3.70-3.62 (m, 1H), 3.55 (s, 1H), 3.26 (q, J = 6.3 Hz, 2H), 2.87-2.75 (m, 2H), 2.74-2.62 (m, 1H), 2.52 (s, 3H), 2.50-2.42 (m, 1H), 2.23-2.06 (m, 4H), 1.84 (s, 1H), 1.68-1.52 (m, 4H), 1.44-1.28 (m, 5H), 1.25 (s, 3H), 0.93 (s, 9H). 1 H NMR (CDCl 3 , 300 MHz) δ 10.00 (s, 1H), 8.68 (s, 1H), 7.61-7.53 (m, 1H), 7.47 (dd, J = 8.5, 7.1 Hz, 1H), 7.35 (S ,4H), 7.08 (d, J = 7.1 Hz, 1H), 6.87 (d, J = 8.5 Hz, 1H), 6.72 (d, J = 9.1 Hz, 1H), 6.24 (t, J = 5.7 Hz, 1H ), 4.89-4.81 (m, 1H), 4.65-4.54 (m, 4H), 4.32-4.23 (m, 1H), 4.06 (d, J = 11.3 Hz, 1H), 3.70-3.62 (m, 1H), 3.55 (s, 1H), 3.26 (q, J = 6.3 Hz, 2H), 2.87-2.75 (m, 2H), 2.74-2.62 (m, 1H), 2.52 (s, 3H), 2.50-2.42 (m, 1H), 2.23-2.06 (m, 4H), 1.84 (s, 1H), 1.68-1.52 (m, 4H), 1.44-1.28 (m, 5H), 1.25 (s, 3H), 0.93 (s, 9H) .

<실시예 2> (2S,4R)-1-((S)-2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥산아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 2> (2S,4R)-1-((S)-2-(6-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-ylamino)hexaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carbox Preparation of amide

Figure 112018098559920-pat00047
Figure 112018098559920-pat00047

단계 1: 6-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)헥산산의 제조Step 1: Preparation of 6-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)hexanoic acid

DMF에 6-아미노헥산산(119 mg, 0.905 mmol)과 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온(250 mg, 0.905 mmol)을 녹인 후, DIPEA(0.236 mL, 1.36 mmol)를 넣고 실온에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻고, MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 10% 28 min)를 이용하여 분리, 정제하여 6-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)헥산산(60 mg, 17%, 황색 고체)을 얻었다.6-aminohexanoic acid (119 mg, 0.905 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-1,3-dione (250 mg, 0.905 mmol) in DMF ) Was dissolved, DIPEA (0.236 mL, 1.36 mmol) was added and stirred at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 10% 28 min) to 6-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoin Doolin-4-yl)amino)hexanoic acid (60 mg, 17%, yellow solid) was obtained.

LC/MS (ESI) m/z [M+H]+ : 388.6, [M-H]- : 399.9LC/MS (ESI) m/z [M+H] + : 388.6, [MH] - : 399.9

1H NMR (CDCl3, 300MHz) δ 8.39 (s, 1H), 7.50 (dd, J = 8.5, 7.2 Hz, 1H), 7.09 (d, J = 7.1 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.29-6.19 (m, 1H), 4.97-4.87 (m, 1H), 3.32-3.29 (m, 2H), 2.95-2.64 (m, 3H), 2.39 (t, J = 7.3 Hz, 2H), 2.19-2.08 (m, 1H), 1.76-1.61 (m, 4H), 1.55-1.41 (m, 2H). 1 H NMR (CDCl 3 , 300 MHz) δ 8.39 (s, 1H), 7.50 (dd, J = 8.5, 7.2 Hz, 1H), 7.09 (d, J = 7.1 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.29-6.19 (m, 1H), 4.97-4.87 (m, 1H), 3.32-3.29 (m, 2H), 2.95-2.64 (m, 3H), 2.39 (t, J = 7.3 Hz, 2H), 2.19-2.08 (m, 1H), 1.76-1.61 (m, 4H), 1.55-1.41 (m, 2H).

단계 2: (2S,4R)-1-((S)-2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥산아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 2: (2S,4R)-1-((S)-2-(6-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Of monoamino)hexanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide Produce

DMF에 상기 단계 1에서 제조한 화합물(35 mg, 0.090 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(42 mg, 0.090 mmol), HATU(52 mg, 0.136 mmol) 을 녹이고 DIPEA(0.063 mL, 0.360 mmol)을 가한 뒤 실온에서 교반한다. 반응 종결 후 EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6% 30 min)를 이용하여 분리, 정제하여 (2S,4R)-1-((S)-2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥산아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드(43 mg, 73%, 황색 고체)를 얻었다.Compound prepared in step 1 above in DMF (35 mg, 0.090 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N- (4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (42 mg, 0.090 mmol), HATU (52 mg, 0.136 mmol) dissolved and DIPEA (0.063 mL, 0.360) mmol) and stirred at room temperature. After completion of the reaction, the organic layer was extracted by diluting with EA and water, washed with brine, water was removed with MgSO 4 and concentrated under reduced pressure. Separated and purified using silica gel column chromatography (MPLC, MeOH/DCM 6% 30 min) to (2S,4R)-1-((S)-2-(6-(2-(2,6-dioxophyte) Peridin-3-yl)-1,3-dioxoisoindolin-4-ylamino)hexanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methyl Thiazole-5-yl)benzyl)pyrrolidine-2-carboxamide (43 mg, 73%, yellow solid) was obtained.

LC/MS (ESI) m/z [M+H]+ : 800.7, [M-H]- : 798.7LC/MS (ESI) m/z [M+H] + : 800.7, [MH] - : 798.7

1H NMR (300 MHz, CDCl3) δ 10.35 (s, 0.5H), 9.67 (s, 0.5H), 8.68 (s, 1H), 7.60 (t, J = 6.2 Hz, 1H), 7.54-7.40 (m, 2H), 7.40-7.30 (m, 4H), 7.07 (t, J = 6.6 Hz, 1H), 6.86 (t, J = 7.8 Hz, 1H), 6.23-6.08 (m, 1H), 4.95-4.81 (m, 1H), 4.71-4.49 (m, 4H), 4.36-4.23 (m, 1H), 4.05 (d, J = 11.5 Hz, 1H), 3.64 (d, J = 11.1 Hz, 1H), 3.27-3.14 (m, 2H), 2.86-2.62 (m, 3H), 2.51 (S,4H), 2.28-2.00 (m, 5H), 1.83 (s, 1H), 1.67-1.47 (m, 4H), 1.34 (d, J = 9.4 Hz, 2H), 0.99-0.77 (m, 9H). 1 H NMR (300 MHz, CDCl 3 ) δ 10.35 (s, 0.5H), 9.67 (s, 0.5H), 8.68 (s, 1H), 7.60 (t, J = 6.2 Hz, 1H), 7.54-7.40 ( m, 2H), 7.40-7.30 (m, 4H), 7.07 (t, J = 6.6 Hz, 1H), 6.86 (t, J = 7.8 Hz, 1H), 6.23-6.08 (m, 1H), 4.95-4.81 (m, 1H), 4.71-4.49 (m, 4H), 4.36-4.23 (m, 1H), 4.05 (d, J = 11.5 Hz, 1H), 3.64 (d, J = 11.1 Hz, 1H), 3.27- 3.14 (m, 2H), 2.86-2.62 (m, 3H), 2.51 (S,4H), 2.28-2.00 (m, 5H), 1.83 (s, 1H), 1.67-1.47 (m, 4H), 1.34 ( d, J = 9.4 Hz, 2H), 0.99-0.77 (m, 9H).

<실시예 3> (2S,4R)-1-((S)-2-(9-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)노난아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 3> (2S,4R)-1-((S)-2-(9-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-ylamino)nonanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carbox Preparation of amide

Figure 112018098559920-pat00048
Figure 112018098559920-pat00048

단계 1: 9-브로모노난 산의 제조Step 1: Preparation of 9-bromononanoic acid

농축된 질산(30 mL, 258 mmol)에 9-브로모노난-1-올(2 g, 8.96 mmol)을 드로핑 펀넬을 이용하여 30분간 가한 후, 4시간 동안 실온에서 교반한다. 이 후 80℃로 가열하여 1시간 동안 교반한다. 반응 종결 후 실온으로 온도를 낮추고, 3차 증류수로 희석한다. 디에틸 에테르로 유기층을 추출하고 MgSO4로 건조하고 농축하여 9-브로모노난 산(1.57 g, 74%, 갈색 오일)을 얻는다.9-bromononan-1-ol (2 g, 8.96 mmol) was added to the concentrated nitric acid (30 mL, 258 mmol) using a dropping funnel for 30 minutes, followed by stirring at room temperature for 4 hours. Thereafter, the mixture was heated to 80° C. and stirred for 1 hour. After completion of the reaction, the temperature was lowered to room temperature and diluted with tertiary distilled water. The organic layer was extracted with diethyl ether, dried over MgSO 4 and concentrated to give 9-bromononanoic acid (1.57 g, 74%, brown oil).

1H NMR (DMSO, 300MHz) δ 9.64 (s, 1H), 3.52 (t, J = 6.4 Hz, 2H), 2.19 (t, J = 6.8 Hz, 2H), 1.83 - 1.77 (m, 2H), 1.62 - 1.17 (m, 10H). 1 H NMR (DMSO, 300 MHz) δ 9.64 (s, 1H), 3.52 (t, J = 6.4 Hz, 2H), 2.19 (t, J = 6.8 Hz, 2H), 1.83-1.77 (m, 2H), 1.62 -1.17 (m, 10H).

단계 2: 9-아미노노난산의 제조Step 2: Preparation of 9-aminononanoic acid

상기 단계 1에서 제조한 화합물(1 g, 4.22 mmol)을 수산화 암모늄 수용액(25% NH3, 40 mL)에 가하고 실온에서 교반한다. 반응 종결 후, 농축하여 9-아미노노난산(529 mg, 72%, 갈색 오일)을 얻는다.The compound (1 g, 4.22 mmol) prepared in step 1 was added to an aqueous ammonium hydroxide solution (25% NH 3 , 40 mL) and stirred at room temperature. After completion of the reaction, it was concentrated to obtain 9-aminononanoic acid (529 mg, 72%, brown oil).

단계3: 9-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)노난산의 제조Step 3: Preparation of 9-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)nonanoic acid

DMF에 상기 단계 2에서 제조한 화합물(200 mg, 1.15 mmol), 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온(318 mg, 1.15 mmol)을 넣고 DIPEA(0.301 mL, 1.73 mmol)를 가한 뒤, 90℃에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 건조하여 농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, EA/Hx 60%, 35 min)를 이용하여 분리, 정제하여 9-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)노난산(25 mg, 5%, 황색 고체)을 얻는다.Compound prepared in Step 2 above in DMF (200 mg, 1.15 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindole-1,3-dione (318 mg, 1.15 mmol) was added and DIPEA (0.301 mL, 1.73 mmol) was added, followed by stirring at 90°C. After completion of the reaction, it was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, dried over MgSO 4 and concentrated. Separation and purification using silica gel column chromatography (MPLC, EA/Hx 60%, 35 min) to 9-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-yl)amino)nonanoic acid (25 mg, 5%, yellow solid) is obtained.

LC/MS (ESI) m/z [M+H]+ : 429.9 , [M-H]- : 427.9LC/MS (ESI) m/z [M+H] + : 429.9, [MH] - : 427.9

1H NMR (CDCl3, 300MHz) δ 8.79 (s, 1H), 7.52 - 7.43 (m, 1H), 7.08 (d, J = 7.1 Hz, 1H), 6.87 (d, J = 8.5 Hz, 1H), 6.24 (s, 1H), 4.96 - 4.90 (m,, 1H), 3.26 - 3.23 (m, 2H), 2.94 - 2.65 (m, 3H), 2.34 (t, J = 7.4 Hz, 2H), 2.17 - 2.05 (m, 1H), 1.64 (d, J = 6.6 Hz, 5H), 1.34 (s, 9H). 1 H NMR (CDCl 3 , 300 MHz) δ 8.79 (s, 1H), 7.52-7.43 (m, 1H), 7.08 (d, J = 7.1 Hz, 1H), 6.87 (d, J = 8.5 Hz, 1H), 6.24 (s, 1H), 4.96-4.90 (m,, 1H), 3.26-3.23 (m, 2H), 2.94-2.65 (m, 3H), 2.34 (t, J = 7.4 Hz, 2H), 2.17-2.05 (m, 1H), 1.64 (d, J = 6.6 Hz, 5H), 1.34 (s, 9H).

단계 4: (2S,4R)-1-((S)-2-(9-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)노난아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 4: (2S,4R)-1-((S)-2-(9-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Of monoamino)nonanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide Produce

DMF에 상기 단계 3에서 제조한 화합물(25 mg, 0.058 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(27 mg, 0.058 mmol), HATU(33 mg, 0.087 mmol)를 넣고 DIPEA(0.041 mL, 0.233 mmol)를 가한 뒤, 90℃에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 건조하여 농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 (2S,4R)-1-((S)-2-(9-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)노난아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드(17 mg, 35%, 황색 고체)를 얻는다.Compound prepared in Step 3 above in DMF (25 mg, 0.058 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N- (4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (27 mg, 0.058 mmol), HATU (33 mg, 0.087 mmol) and DIPEA (0.041 mL, 0.233) mmol), and stirred at 90°C. After completion of the reaction, it was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, dried over MgSO 4 and concentrated. Separated and purified using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to (2S,4R)-1-((S)-2-(9-(2-(2,6-diox) Sopiperidin-3-yl)-1,3-dioxoisoindolin-4-ylamino)nonaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4- Methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide (17 mg, 35%, yellow solid).

LC/MS (ESI) m/z [M+H]+ : 842.6 , [M-H]- : 840.7LC/MS (ESI) m/z [M+H] + : 842.6, [MH] - : 840.7

1H NMR (300 MHz, CDCl3) δ 10.63 (s, 0.5H), 10.37 (s, 0.5H), 10.32 (s, 0.5H), 10.29 (s, 0.5H), 8.67 (s, 1H), 7.68-7.53 (m, 1H), 7.44 (t, J = 7.8 Hz, 1H), 7.38-7.30 (m, 3H), 7.03 (d, J = 7.1 Hz, 1H), 6.94-6.76 (m, 2H), 6.22 (s, 1H), 4.98-4.78 (m, 1H), 4.69-4.44 (m, 4H), 4.34-4.09 (m, 2H), 4.05-3.90 (m, 1H), 3.69-3.56 (m, 1H), 3.28-3.11 (m, 2H), 2.83-2.56 (m, 3H), 2.52-2.32 (m, 4H), 2.22-1.96 (m, 4H), 1.66-1.44 (m, 4H), 1.40-1.09 (m, 7H), 0.91 (s, 9H). LC/MS (ESI) m/z 842.6[M+H]+, 840.7[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 10.63 (s, 0.5H), 10.37 (s, 0.5H), 10.32 (s, 0.5H), 10.29 (s, 0.5H), 8.67 (s, 1H), 7.68-7.53 (m, 1H), 7.44 (t, J = 7.8 Hz, 1H), 7.38-7.30 (m, 3H), 7.03 (d, J = 7.1 Hz, 1H), 6.94-6.76 (m, 2H) , 6.22 (s, 1H), 4.98-4.78 (m, 1H), 4.69-4.44 (m, 4H), 4.34-4.09 (m, 2H), 4.05-3.90 (m, 1H), 3.69-3.56 (m, 1H), 3.28-3.11 (m, 2H), 2.83-2.56 (m, 3H), 2.52-2.32 (m, 4H), 2.22-1.96 (m, 4H), 1.66-1.44 (m, 4H), 1.40- 1.09 (m, 7H), 0.91 (s, 9H). LC/MS (ESI) m/z 842.6 [M+H] + , 840.7 [MH] -

<실시예 4> (2S,4R)-1-((S)-2-tert-부틸-17-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-4-옥소-6,9,12,15-테트라옥사-3-아자헵타데카-1-노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 4> (2S,4R)-1-((S)-2-tert-butyl-17-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-ylamino)-4-oxo-6,9,12,15-tetraoxa-3-azaheptadeca-1-noyl)-4-hydroxy-N-(4-(4-methylthia Preparation of zol-5-yl)benzyl)pyrrolidine-2-carboxamide

Figure 112018098559920-pat00049
Figure 112018098559920-pat00049

단계 1: tert-부틸 14-아미노-3,6,9,12-테트라옥사테트라데카-1-노에이트의 제조Step 1: Preparation of tert-butyl 14-amino-3,6,9,12-tetraoxatetradeca-1-noate

tert-부틸 14-요오도-3,6,9,12-테트라옥사테트라데카-1-노에이트(315 mg, 0.753 mmol)를 DMF에 녹이고 소듐 아자이드(147 mg, 2.26 mmol)를 가하여 50℃에서 교반한다. 반응 종결 후, 농축하여 조 생성물 상태의 아자이드 화합물을 얻은 후, 에탄올에 녹이고 Pd(OH)2(6 mg)를 가하여 수소 기체 하에서 교반한다. 반응 종결 후, 메탄올을 이용하여 셀라이트 여과한 후, 농축하여 조 생성물 상태의 tert-부틸 14-아미노-3,6,9,12-테트라옥사테트라데카-1-노에이트(240 mg, 정량. 황색 고체)를 얻는다.tert-butyl 14-iodo-3,6,9,12-tetraoxatetradeca-1-noate (315 mg, 0.753 mmol) was dissolved in DMF and sodium azide (147 mg, 2.26 mmol) was added to add 50°C. Stir in. After completion of the reaction, it was concentrated to obtain an azide compound in the crude product state, dissolved in ethanol, Pd(OH) 2 (6 mg) was added, and the mixture was stirred under hydrogen gas. After completion of the reaction, celite was filtered using methanol, and then concentrated to give crude tert-butyl 14-amino-3,6,9,12-tetraoxatetradeca-1-noate (240 mg, quantitative). Yellow solid).

1H NMR (DMSO-d6, 300MHz) δ 3.99 (s, 2H), 3.62 - 3.46 (m, 13H), 3.46 - 3.40 (m, 2H), 3.35 (t, J = 5.8 Hz, 7H), 2.63 (t, J = 5.8 Hz, 2H), 1.42 (s, 9H). 1 H NMR (DMSO-d 6 , 300 MHz) δ 3.99 (s, 2H), 3.62-3.46 (m, 13H), 3.46-3.40 (m, 2H), 3.35 (t, J = 5.8 Hz, 7H), 2.63 (t, J = 5.8 Hz, 2H), 1.42 (s, 9H).

단계 2: tert-부틸 14-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-3,6,9,12-테트라옥사테트라데카노에이트의 제조Step 2: tert-Butyl 14-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-3,6,9,12 -Preparation of tetraoxatetradecanoate

DMSO에 상기 단계 1에서 제조한 화합물(100 mg, 0.325 mmol), 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온(90 mg, 0.325 mmol)을 첨가하고, DIPEA(0.085 mL, 0.488 mmol)를 가한 후, 90℃에서 교반한다. 반응 종결 후, EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 물을 제거하여 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 5% 28 min)를 이용하여 분리, 정제하여 tert-부틸 14-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-3,6,9,12-테트라옥사테트라데카-1-노에이트(7 mg, 4%, 황색 고체)를 얻는다.Compound prepared in step 1 above in DMSO (100 mg, 0.325 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindole-1,3-dione (90 mg, 0.325 mmol) was added, DIPEA (0.085 mL, 0.488 mmol) was added, followed by stirring at 90°C. After completion of the reaction, dilute with EA and water, extract the organic layer, wash with brine, remove water with MgSO 4 and concentrate under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 5% 28 min) to tert-butyl 14-(2-(2,6-dioxopiperidin-3-yl)-1,3-di Oxoisoindoline-4-ylamino)-3,6,9,12-tetraoxatetradeca-1-noate (7 mg, 4%, yellow solid) is obtained.

LC/MS (ESI) m/z [M+H]+ = 564.9 [M-H]- = 562.9LC/MS (ESI) m/z [M+H] + = 564.9 [MH] - = 562.9

단계 3: 14-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-3,6,9,12-테트라옥사테트라데칸산의 제조Step 3: 14-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)amino)-3,6,9,12-tetraoxa Preparation of tetradecanoic acid

40% TFA/DCM 용액에 tert-부틸 14-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-3,6,9,12-테트라옥사테트라데카-1-노에이트(7 mg, 0.012 mmol)를 가하고 실온에서 교반한다. 반응 종결 후, 농축하여 14-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-3,6,9,12-테트라옥사테트라데칸산(10 mg, 조 생성물, 갈색 오일)을 조 생성물 상태로 얻는다.Tert-butyl 14-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-ylamino)-3,6,9 in 40% TFA/DCM solution ,12-tetraoxatetradeca-1-noate (7 mg, 0.012 mmol) was added and stirred at room temperature. After completion of the reaction, it was concentrated to give 14-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-3,6,9,12 -Tetraoxatetradecanoic acid (10 mg, crude product, brown oil) is obtained as crude product.

LC/MS (ESI) m/z [M+H]+ = 508.8 [M-H]- = 506.8LC/MS (ESI) m/z [M+H] + = 508.8 [MH] - = 506.8

단계 4: (2S,4R)-1-((S)-2-(tert-부틸)-17-((2-(2,6-디옥사피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-4-옥소-6,9,12,15-테트라옥사-3-아자헵타데카노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 4: (2S,4R)-1-((S)-2-(tert-butyl)-17-((2-(2,6-dioxapiperidin-3-yl)-1,3- Dioxoisoindolin-4-yl)amino)-4-oxo-6,9,12,15-tetraoxa-3-azaheptadecanoyl)-4-hydroxy-N-(4-(4-methylthia Preparation of zol-5-yl)benzyl)pyrrolidine-2-carboxamide

상기 단계 3에서 제조한 화합물(6 mg, 0.012 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(6 mg, 0.012 mmol), HATU(7 mg, 0.018 mmol)를 DMF 에 녹인 후, DIPEA(0.008 mL, 0.048 mmol)를 가한 후, 실온에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 (2S,4R)-1-((S)-2-(tert-부틸)-17-((2-(2,6-디옥사피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-4-옥소-6,9,12,15-테트라옥사-3-아자헵타데카노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드(3 mg, 27%, 황색 고체)를 얻는다.Compound prepared in Step 3 (6 mg, 0.012 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4 -(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (6 mg, 0.012 mmol) and HATU (7 mg, 0.018 mmol) were dissolved in DMF, followed by DIPEA (0.008 mL) , 0.048 mmol), and stirred at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separated and purified using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to (2S,4R)-1-((S)-2-(tert-butyl)-17-((2- (2,6-Dioxapiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)amino)-4-oxo-6,9,12,15-tetraoxa-3-aza Heptadecanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide (3 mg, 27%, yellow solid) is obtained.

LC/MS (ESI) m/z [M+H]+ : 921.8 , [M-H]- : 919.8LC/MS (ESI) m/z [M+H] + : 921.8, [MH] - : 919.8

1H NMR (300 MHz, CDCl3) δ 9.29 (s, 1H), 8.68 (s, 1H), 7.63-7.43 (m, 2H), 7.42-7.32 (m, 3H), 7.32-7.27 (m, 1H), 7.10 (d, J = 7.1 Hz, 1H), 6.95-6.86 (m, 1H), 6.52 (s, 1H), 4.93-4.79 (m, 1H), 4.72 (t, J = 8.0 Hz, 1H), 4.64-4.45 (m, 2H), 4.37-4.20 (m, 1H), 4.17-4.06 (m, 1H), 4.04-3.97 (m, 1H), 3.79-3.54 (m, 12H), 3.52-3.38 (m, 2H), 2.94-2.62 (m, 3H), 2.52 (s, 3H), 2.22-1.96 (m, 3H), 1.26 (s, 9H), 0.99-0.78 (m, 10H). 1 H NMR (300 MHz, CDCl 3 ) δ 9.29 (s, 1H), 8.68 (s, 1H), 7.63-7.43 (m, 2H), 7.42-7.32 (m, 3H), 7.32-7.27 (m, 1H ), 7.10 (d, J = 7.1 Hz, 1H), 6.95-6.86 (m, 1H), 6.52 (s, 1H), 4.93-4.79 (m, 1H), 4.72 (t, J = 8.0 Hz, 1H) , 4.64-4.45 (m, 2H), 4.37-4.20 (m, 1H), 4.17-4.06 (m, 1H), 4.04-3.97 (m, 1H), 3.79-3.54 (m, 12H), 3.52-3.38 ( m, 2H), 2.94-2.62 (m, 3H), 2.52 (s, 3H), 2.22-1.96 (m, 3H), 1.26 (s, 9H), 0.99-0.78 (m, 10H).

<실시예 5> (2S,4R)-1-((S)-2-(2-(3-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-2-옥소에톡시)프로폭시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 5> (2S,4R)-1-((S)-2-(2-(3-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3) -Dioxoisoindolin-4-ylamino)-2-oxoethoxy)propoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthia Preparation of zol-5-yl)benzyl)pyrrolidine-2-carboxamide

Figure 112018098559920-pat00050
Figure 112018098559920-pat00050

단계 1: 디-tert-부틸 2,2'-(프로판-1,3-디일비스(옥시))디아세테이트의 제조Step 1: Preparation of di-tert-butyl 2,2'-(propane-1,3-diylbis(oxy))diacetate

톨루엔과 NaOH 수용액(50 wt%)에 프로판-1,3-디올(1 g, 13.1 mmol), tert-부틸 2-브로모아세테이트(21.5 g, 110.4 mmol), 테트라부틸 암모늄 황산 수소염(445 mg, 1.31 mmol)을 가하고 실온에서 16시간 동안 교반한다. 반응 종결 후, 빙수로 희석하고 EA로 추출한 후, 염수로 씻고 MgSO4로 물을 제거하고 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, EA/Hx 30% 40 min)를 이용하여 분리, 정제하여 디-tert-부틸 2,2'-(프로판-1,3-디일비스(옥시))디아세테이트(580 mg, 15%, 무색 오일)를 7얻는다.Propane-1,3-diol (1 g, 13.1 mmol), tert-butyl 2-bromoacetate (21.5 g, 110.4 mmol), tetrabutyl ammonium hydrogen sulfate (445 mg) in toluene and NaOH aqueous solution (50 wt%) , 1.31 mmol) and stirred at room temperature for 16 hours. After completion of the reaction, diluted with ice water, extracted with EA, washed with brine, water was removed with MgSO 4 and concentrated under reduced pressure. Separated and purified using silica gel column chromatography (MPLC, EA/Hx 30% 40 min) to di-tert-butyl 2,2'-(propane-1,3-diylbis(oxy)) diacetate (580 mg , 15%, colorless oil).

1H NMR (CDCl3, 300MHz) δ 3.96 (S,4H), 3.63 (t, J = 6.3 Hz, 4H), 1.94 (p, J = 6.3 Hz, 2H), 1.48 (s, 18H). 1 H NMR (CDCl 3 , 300 MHz) δ 3.96 (S,4H), 3.63 (t, J = 6.3 Hz, 4H), 1.94 (p, J = 6.3 Hz, 2H), 1.48 (s, 18H).

단계 2: 2,2'-(프로판-1,3-디일비스(옥시))디아세트산의 제조Step 2: Preparation of 2,2'-(propane-1,3-diylbis(oxy))diacetic acid

40% TFA/DCM 용액에 디-tert-부틸 2,2'-(프로판-1,3-디일비스(옥시))디아세테이트(530 mg, 1.74 mmol)를 가하고 실온에서 교반한다. 반응 종결 후, 농축하여 2,2'-(프로판-1,3-디일비스(옥시))디아세트산(631 mg, 조 생성물, 갈색 오일) 을 조 생성물 상태로 얻는다.Di-tert-butyl 2,2'-(propane-1,3-diylbis(oxy)) diacetate (530 mg, 1.74 mmol) was added to a 40% TFA/DCM solution and stirred at room temperature. After completion of the reaction, it was concentrated to obtain 2,2'-(propane-1,3-diylbis(oxy))diacetic acid (631 mg, crude product, brown oil) as a crude product.

1H NMR (CDCl3, 300MHz) δ 10.12 (s, 2H), 4.21 (S,4H), 3.75 (t, J = 5.9 Hz, 4H), 2.08 - 1.90 (m, 2H). 1 H NMR (CDCl 3 , 300 MHz) δ 10.12 (s, 2H), 4.21 (S,4H), 3.75 (t, J = 5.9 Hz, 4H), 2.08-1.90 (m, 2H).

단계 3: 2-(3-(2-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-2-옥소에톡시)프로폭시)아세트산의 제조Step 3: 2-(3-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-2-oxo Preparation of ethoxy)propoxy)acetic acid

상기 단계 2에서 제조한 화합물(167 mg, 0.869 mmol)을 DCM에 녹이고 염화 옥살일(0.373 mL, 4.35 mmol)을 가하고, DMF를 한 방울 가한다. 산염화물이 만들어진 것을 확인한 후, 농축한다. THF에 녹이고 포말리도마이드(Pomalidomide)를 가한 후, 환류한다. 반응 종결 후, 0.1 N HCl(aq.)용액으로 반응을 종결시키고 EA와 물로 희석하여 유기층을 추출한 후, 염수로 씻고, MgSO4로 물을 제거한 후, 감압농축한다. 생성된 고체를 EA로 여과하여 2-(3-(2-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-2-옥소에톡시)프로폭시)아세트산(54 mg, 28%, 회색 고체)를 얻는다. LC/MS (ESI) m/z [M+H]+ = 447.8 [M-H]- = 445.8The compound (167 mg, 0.869 mmol) prepared in step 2 was dissolved in DCM, oxalyl chloride (0.373 mL, 4.35 mmol) was added, and DMF was added dropwise. After confirming that the acid chloride has been formed, it is concentrated. Dissolve in THF, add pomalidomide, and reflux. After completion of the reaction, the reaction was terminated with a 0.1 N HCl (aq.) solution, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. The resulting solid was filtered with EA to give 2-(3-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino) 2-Oxoethoxy)propoxy)acetic acid (54 mg, 28%, gray solid). LC/MS (ESI) m/z [M+H] + = 447.8 [MH] - = 445.8

1H NMR (CDCl3, 300MHz) δ 10.41 (s, 1H), 8.96 - 8.78 (m, 1H), 8.65 (s, 1H), 7.81 - 7.66 (m, 1H), 7.62 - 7.50 (m, 1H), 5.04 - 4.86 (m, 1H), 4.27 - 4.04 (m, 4H), 3.86 (t, J = 6.7 Hz, 2H), 3.74 (t, J = 6.0 Hz, 2H), 3.01 - 2.64 (m, 3H), 2.15 - 1.99 (m, 3H). 1 H NMR (CDCl 3 , 300MHz) δ 10.41 (s, 1H), 8.96-8.78 (m, 1H), 8.65 (s, 1H), 7.81-7.66 (m, 1H), 7.62-7.50 (m, 1H) , 5.04-4.86 (m, 1H), 4.27-4.04 (m, 4H), 3.86 (t, J = 6.7 Hz, 2H), 3.74 (t, J = 6.0 Hz, 2H), 3.01-2.64 (m, 3H) ), 2.15-1.99 (m, 3H).

단계 4: (2S,4R)-1-((S)-2-(2-(3-(2-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-2-옥소에톡시)프로폭시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 4: (2S,4R)-1-((S)-2-(2-(3-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3- Dioxoisoindoline-4-yl)amino)-2-oxoethoxy)propoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthia Preparation of zol-5-yl)benzyl)pyrrolidine-2-carboxamide

상기 단계 3에서 제조한 화합물(30 mg, 0.067 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(31 mg, 0.067 mmol), HATU(38 mg, 0.101 mmol)를 DMF에 녹인 후, DIPEA(0.047 mL, 0.268 mmol)를 가한 후, 실온에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고, MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 (2S,4R)-1-((S)-2-(2-(3-(2-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-2-옥소에톡시)프로폭시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드(5 mg, 9%, 흰색 고체)를 얻는다.Compound prepared in step 3 (30 mg, 0.067 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4 -(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (31 mg, 0.067 mmol) and HATU (38 mg, 0.101 mmol) were dissolved in DMF, followed by DIPEA (0.047 mL) , 0.268 mmol), and stirred at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separated and purified using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to (2S,4R)-1-((S)-2-(2-(3-(2-((2 -(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)amino)-2-oxoethoxy)propoxy)acetamido)-3,3- Dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide (5 mg, 9%, white solid) is obtained.

LC/MS (ESI) m/z [M+H]+ : 860.5 , [M-H]- : 858.7LC/MS (ESI) m/z [M+H] + : 860.5, [MH] - : 858.7

1H NMR (CDCl3, 500MHz) δ 10.35 (s, 1H), 8.81 (d, J = 8.4 Hz, 1H), 8.68 (s, 1H), 7.76-7.68 (m, 1H), 7.62-7.53 (m, 1H), 7.40-7.35 (m, 4H), 7.32 (d, J = 9.5 Hz, 1H), 7.19 (d, J = 6.2 Hz, 1H), 4.95-4.84 (m, 1H), 4.67-4.59 (m, 2H), 4.50 (s, 2H), 4.33-4.25 (m, 1H), 4.23-4.14 (m, 1H), 4.12-4.01 (m, 3H), 4.01-3.97 (m, 1H), 3.93-3.85 (m, 1H), 3.82-3.73 (m, 2H), 3.65-3.56 (m, 2H), 2.88-2.57 (m, 3H), 2.53 (s, 3H), 2.40 (s, 1H), 2.19-1.90 (m, 6H), 1.71-1.60 (m, 2H), 1.59-1.38 (m, 2H), 0.95 (s, 9H). 1 H NMR (CDCl 3 , 500 MHz) δ 10.35 (s, 1H), 8.81 (d, J = 8.4 Hz, 1H), 8.68 (s, 1H), 7.76-7.68 (m, 1H), 7.62-7.53 (m , 1H), 7.40-7.35 (m, 4H), 7.32 (d, J = 9.5 Hz, 1H), 7.19 (d, J = 6.2 Hz, 1H), 4.95-4.84 (m, 1H), 4.67-4.59 ( m, 2H), 4.50 (s, 2H), 4.33-4.25 (m, 1H), 4.23-4.14 (m, 1H), 4.12-4.01 (m, 3H), 4.01-3.97 (m, 1H), 3.93- 3.85 (m, 1H), 3.82-3.73 (m, 2H), 3.65-3.56 (m, 2H), 2.88-2.57 (m, 3H), 2.53 (s, 3H), 2.40 (s, 1H), 2.19- 1.90 (m, 6H), 1.71-1.60 (m, 2H), 1.59-1.38 (m, 2H), 0.95 (s, 9H).

<실시예 6> N<Example 6> N 1One -(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 99 -((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)노난디아미드의 제조-((S)-1-((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3 Preparation of ,3-dimethyl-1-oxobutan-2-yl)nonanediamide

Figure 112018098559920-pat00051
Figure 112018098559920-pat00051

9-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-9-옥소노난산(30 mg, 0.068 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(32 mg, 0.068 mmol), HATU(39 mg, 0.101 mmol)를 DMF 에 녹인 후, DIPEA(0.047 mL, 0.272 mmol)를 가한 후, 실온에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻고 MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N9-((R)-1-((2R,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)노난디아미드(21 mg, 36%, 흰색 고체)를 얻는다.9-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-ylamino)-9-oxononanoic acid (30 mg, 0.068 mmol), (2S ,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine After -2-carboxamide hydrochloride (32 mg, 0.068 mmol) and HATU (39 mg, 0.101 mmol) were dissolved in DMF, DIPEA (0.047 mL, 0.272 mmol) was added, followed by stirring at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-yl)-N 9 -((R)-1-((2R,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl) Pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)nonanediamide (21 mg, 36%, white solid) is obtained.

LC/MS (ESI) m/z [M+H]+ : 856.6 , [M-H]- : 854.7LC/MS (ESI) m/z [M+H] + : 856.6, [MH] - : 854.7

1H NMR (300 MHz, CDCl3) δ 10.92 (s, 0.5H), 10.35 (s, 0.5H), 9.53 (s, 0.5H), 9.50 (s, 0.5H), 8.84 (d, J = 8.4 Hz, 0.5H), 8.79 (d, J = 8.4 Hz, 0.5H), 8.69 (s, 1H), 7.79-7.65 (m, 1H), 7.59-7.48 (m, 1H), 7.40-7.30 (m, 4H), 6.83 (d, J = 9.0 Hz, 0.5H), 6.63 (d, J = 9.0 Hz, 0.5H), 5.01-4.83 (m, 1H), 4.73-4.48 (m, 4H), 4.35-4.21 (m, 1H), 4.09 (t, J = 10.5 Hz, 1H), 3.70-3.59 (m, 1H), 2.89-2.60 (m, 3H), 2.57-2.36 (m, 6H), 2.23-2.06 (m, 4H), 1.91-1.65 (m, 2H), 1.63-1.51 (m, 2H), 1.51-1.41 (m, 2H), 1.40-1.16 (m, 7H), 1.00-0.81 (m, 9H). LC/MS (ESI) m/z 856.6[M+H]+, 854.7[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 10.92 (s, 0.5H), 10.35 (s, 0.5H), 9.53 (s, 0.5H), 9.50 (s, 0.5H), 8.84 (d, J = 8.4 Hz, 0.5H), 8.79 (d, J = 8.4 Hz, 0.5H), 8.69 (s, 1H), 7.79-7.65 (m, 1H), 7.59-7.48 (m, 1H), 7.40-7.30 (m, 4H), 6.83 (d, J = 9.0 Hz, 0.5H), 6.63 (d, J = 9.0 Hz, 0.5H), 5.01-4.83 (m, 1H), 4.73-4.48 (m, 4H), 4.35-4.21 (m, 1H), 4.09 (t, J = 10.5 Hz, 1H), 3.70-3.59 (m, 1H), 2.89-2.60 (m, 3H), 2.57-2.36 (m, 6H), 2.23-2.06 (m , 4H), 1.91-1.65 (m, 2H), 1.63-1.51 (m, 2H), 1.51-1.41 (m, 2H), 1.40-1.16 (m, 7H), 1.00-0.81 (m, 9H). LC/MS (ESI) m/z 856.6 [M+H] + , 854.7 [MH] -

<실시예 7> N<Example 7> N 1One -(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 88 -((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)옥탄디아미드의 제조-((S)-1-((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3 Preparation of ,3-dimethyl-1-oxobutan-2-yl)octanediamide

Figure 112018098559920-pat00052
Figure 112018098559920-pat00052

8-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-8-옥소옥탄산(30 mg, 0.070 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(33 mg, 0.070 mmol), HATU(40 mg, 0.105 mmol)를 DMF에 녹인 후, DIPEA(0.049 mL, 0.279 mmol)를 가한 후, 실온에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻어주고 MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N8-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)옥탄디아미드(17 mg, 29%, 흰색 고체)를 얻는다.8-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-ylamino)-8-oxooctanoic acid (30 mg, 0.070 mmol), (2S ,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine After -2-carboxamide hydrochloride (33 mg, 0.070 mmol) and HATU (40 mg, 0.105 mmol) were dissolved in DMF, DIPEA (0.049 mL, 0.279 mmol) was added, followed by stirring at room temperature. After completion of the reaction, it was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-yl)-N 8 -((S)-1-((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl) Pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)octanediamide (17 mg, 29%, white solid) is obtained.

LC/MS (ESI) m/z [M+H]+ : 842.6 , [M-H]- : 840.7LC/MS (ESI) m/z [M+H] + : 842.6, [MH] - : 840.7

1H NMR (300 MHz, CDCl3) δ 11.23 (s, 0.5H), 10.21 (s, 0.5H), 9.48 (d, J = 10.3 Hz, 1H), 8.77 (t, J = 9.0 Hz, 1H), 8.72-8.65 (m, 1H), 7.77-7.66 (m, 1H), 7.61-7.47 (m, 1.5H), 7.40-7.29 (m, 4H), 7.03 (d, J = 9.0 Hz, 0.5H), 6.59 (d, J = 9.0 Hz, 0.5H), 6.26-6.19 (m, 0.5H), 4.97-4.84 (m, 1H), 4.75-4.48 (m, 4H), 4.37-4.25 (m, 1H), 4.18-4.04 (m, 1H), 3.69-3.57 (m, 2H), 2.91-2.61 (m, 3H), 2.57-2.40 (m, 5H), 2.32-2.06 (m, 4H), 2.06-1.97 (m, 1H), 1.85 (t, J = 7.5 Hz, 1H), 1.79-1.68 (m, 1H), 1.68-1.52 (m, 3H), 1.44-1.32 (m, 3H), 0.97-0.83 (m, 9H). LC/MS (ESI) m/z 842.6[M+H]+, 840.7[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 11.23 (s, 0.5H), 10.21 (s, 0.5H), 9.48 (d, J = 10.3 Hz, 1H), 8.77 (t, J = 9.0 Hz, 1H) , 8.72-8.65 (m, 1H), 7.77-7.66 (m, 1H), 7.61-7.47 (m, 1.5H), 7.40-7.29 (m, 4H), 7.03 (d, J = 9.0 Hz, 0.5H) , 6.59 (d, J = 9.0 Hz, 0.5H), 6.26-6.19 (m, 0.5H), 4.97-4.84 (m, 1H), 4.75-4.48 (m, 4H), 4.37-4.25 (m, 1H) , 4.18-4.04 (m, 1H), 3.69-3.57 (m, 2H), 2.91-2.61 (m, 3H), 2.57-2.40 (m, 5H), 2.32-2.06 (m, 4H), 2.06-1.97 ( m, 1H), 1.85 (t, J = 7.5 Hz, 1H), 1.79-1.68 (m, 1H), 1.68-1.52 (m, 3H), 1.44-1.32 (m, 3H), 0.97-0.83 (m, 9H). LC/MS (ESI) m/z 842.6 [M+H] + , 840.7 [MH] -

<실시예 8> N<Example 8> N 1One -(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 77 -((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)헵탄디아미드의 제조-((S)-1-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pyrrolidin-1-yl) Preparation of -3,3-dimethyl-1-oxobutan-2-yl)heptanediamide

Figure 112018098559920-pat00053
Figure 112018098559920-pat00053

7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-7-옥소헵탄산(50 mg, 0.120 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(56 mg, 0.120 mmol), HATU(68 mg, 0.180 mmol)를 DMF에 녹인 후 DIPEA(0.084 mL, 0.480 mmol)를 가한 후, 실온에서 교반한다. 반응 종결 후, 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻고 MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N7-((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)헵탄디아미드(33 mg, 33%, 흰색 고체)을 얻는다.7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-ylamino)-7-oxoheptanoic acid (50 mg, 0.120 mmol), (2S ,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine After dissolving -2-carboxamide hydrochloride (56 mg, 0.120 mmol) and HATU (68 mg, 0.180 mmol) in DMF, DIPEA (0.084 mL, 0.480 mmol) was added, followed by stirring at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, and the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-yl)-N 7 -((S)-1-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)cover Moyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)heptanediamide (33 mg, 33%, white solid) is obtained.

LC/MS (ESI) m/z [M+H]+ : 828.6 , [M-H]- : 826.7LC/MS (ESI) m/z [M+H] + : 828.6, [MH] - : 826.7

1H NMR (CDCl3, 300MHz) δ 10.23 (s, 1H), 9.44 (s, 1H), 8.75 (d, J = 8.5 Hz, 1H), 8.68 (s, 1H), 7.68 (t, J = 7.9 Hz, 1H), 7.54 (dd, J = 10.5, 6.7 Hz, 2H), 7.35-7.32 (m, 4H), 6.76 (d, J = 9.1 Hz, 1H), 4.92-4.82 (m, 1H), 4.68-4.53 (m, 4H), 4.91-4.84 (m, 1H), 4.07 (d, J = 11.3 Hz, 2H), 3.74-3.63 (m, 1H), 2.88-2.61 (m, 3H), 2.56-2.37 (m, 6H), 2.30-1.99 (m, 5H), 1.81-1.54 (m, 4H), 1.47-1.30 (m, 2H), 0.95 (s, 9H). 1 H NMR (CDCl 3 , 300 MHz) δ 10.23 (s, 1H), 9.44 (s, 1H), 8.75 (d, J = 8.5 Hz, 1H), 8.68 (s, 1H), 7.68 (t, J = 7.9 Hz, 1H), 7.54 (dd, J = 10.5, 6.7 Hz, 2H), 7.35-7.32 (m, 4H), 6.76 (d, J = 9.1 Hz, 1H), 4.92-4.82 (m, 1H), 4.68 -4.53 (m, 4H), 4.91-4.84 (m, 1H), 4.07 (d, J = 11.3 Hz, 2H), 3.74-3.63 (m, 1H), 2.88-2.61 (m, 3H), 2.56-2.37 (m, 6H), 2.30-1.99 (m, 5H), 1.81-1.54 (m, 4H), 1.47-1.30 (m, 2H), 0.95 (s, 9H).

<실시예 9> N<Example 9> N 1One -(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 1414 -((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)-3,6,9,12-테트라옥사테트라데칸-1,14-디아미드의 제조-((S)-1-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pyrrolidin-1-yl) Preparation of -3,3-dimethyl-1-oxobutan-2-yl)-3,6,9,12-tetraoxatetradecane-1,14-diamide

Figure 112018098559920-pat00054
Figure 112018098559920-pat00054

단계 1: 디-tert-부틸 3,6,9,12-테트라옥사테트라데칸디오에이트의 제조Step 1: Preparation of di-tert-butyl 3,6,9,12-tetraoxatetradecanedioate

톨루엔과 NaOH 수용액(50 wt%)에 2-(2-(히드록시메톡시)에톡시)에탄올(1 g, 7.34 mmol), tert-부틸 2-브로모아세테이트(9.12 mL, 61.7 mmol), 테트라부틸 암모늄 황산 수소염(249 mg, 0.734 mmol)을 가하고 실온에서 16시간 동안 교반한다. 반응 종결 후, 빙수로 희석하고 EA로 추출한 후, 염수로 씻어주고, MgSO4로 물을 제거하고 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, EA/Hx 50% 40 min)를 이용하여 분리, 정제하여 디-tert-부틸 3,6,9,12-테트라옥사테트라데칸디오에이트(1.24 g, 31%, 무색의 오일)를 얻는다.2-(2-(hydroxymethoxy)ethoxy)ethanol (1 g, 7.34 mmol), tert-butyl 2-bromoacetate (9.12 mL, 61.7 mmol), tetra in toluene and NaOH aqueous solution (50 wt%) Butyl ammonium hydrogen sulfate (249 mg, 0.734 mmol) was added and stirred at room temperature for 16 hours. After completion of the reaction, diluted with ice water, extracted with EA, washed with brine, water was removed with MgSO 4 and concentrated under reduced pressure. Separated and purified using silica gel column chromatography (MPLC, EA/Hx 50% 40 min) to di-tert-butyl 3,6,9,12-tetraoxatetradecanedioate (1.24 g, 31%, colorless Oil).

1H NMR (CDCl3, 300MHz) δ 4.02 (S,4H), 3.75 - 3.62 (m, 12H), 1.47 (s, 18H). 1 H NMR (CDCl 3 , 300 MHz) δ 4.02 (S, 4H), 3.75-3.62 (m, 12H), 1.47 (s, 18H).

단계 2: 3,6,9,12-테트라옥사테트라데칸-1,14-디온산의 제조Step 2: Preparation of 3,6,9,12-tetraoxatetradecane-1,14-diionic acid

40% TFA/DCM 용액에 상기 단계 1에서제조한 화합물(450 mg, 1.48 mmol)을 가하고 실온에서 교반한다. 반응 종결 후, 농축하여 3,6,9,12-테트라옥사테트라데칸-1,14-디온산33 (502 mg, 조 생성물, 갈색 오일) 을 조 생성물 상태로 얻는다.To the 40% TFA/DCM solution, the compound prepared in step 1 (450 mg, 1.48 mmol) was added and stirred at room temperature. After completion of the reaction, it is concentrated to obtain 3,6,9,12-tetraoxatetradecane-1,14-diionic acid 33 (502 mg, crude product, brown oil) as a crude product.

1H NMR (CDCl3, 300MHz) δ 10.14 (s, 1H), 4.23 (S,4H), 3.98 - 3.40 (m, 12H). 1 H NMR (CDCl 3 , 300 MHz) δ 10.14 (s, 1H), 4.23 (S,4H), 3.98-3.40 (m, 12H).

단계 3: 14-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-14-옥소-3,6,9,12-테트라옥사테트라데칸산의 제조Step 3: 14-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)amino)-14-oxo-3,6,9, Preparation of 12-tetraoxatetradecanoic acid

상기 단계 2에서 제조한 화합물(394 mg, 1.48 mmol)을 DCM에 녹이고 염화 옥살일(0.635 mL, 7.40 mmol)을 가하고, DMF를 한 방울 가한다. 산염화물이 만들어진 것을 확인한 후, 농축한다. THF에 녹이고 포말리도마이드(202 mg, 0.740 mmol)를 가한 후, 환류한다. 반응 종결 후 0.1 N HCl(aq.) 용액으로 반응을 종결 시키고 EA와 물로 희석하여 유기층을 추출한 후, 염수로 씻고, MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 20% 40 min)로 분리, 정제하여 14-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-14-옥소-3,6,9,12-테트라옥사테트라데칸산(78 mg, 20%, 갈색 고체)을 얻는다.The compound (394 mg, 1.48 mmol) prepared in Step 2 was dissolved in DCM, oxalyl chloride (0.635 mL, 7.40 mmol) was added, and DMF was added dropwise. After confirming that the acid chloride has been formed, it is concentrated. Dissolve in THF, add pomalidomide (202 mg, 0.740 mmol), and reflux. After completion of the reaction, the reaction was terminated with a 0.1 N HCl (aq.) solution, diluted with EA and water, and the organic layer was extracted, washed with brine, removed with MgSO 4 and concentrated under reduced pressure. Separated and purified by silica gel column chromatography (MPLC, MeOH/DCM 20% 40 min) to 14-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-yl)amino)-14-oxo-3,6,9,12-tetraoxatetradecanoic acid (78 mg, 20%, brown solid).

LC/MS (ESI) m/z [M+H]+ = 522.8 [M-H]- = 520.8LC/MS (ESI) m/z [M+H] + = 522.8 [MH] - = 520.8

1H NMR (CDCl3, 300MHz) δ 10.47 (s, 1H), 9.18 (s, 1H), 8.84 (d, J = 8.4 Hz, 1H), 7.72 (t, J = 7.9 Hz, 1H), 7.57 (d, J = 7.3 Hz, 1H), 5.07-4.92 (m, 1H), 4.29-4.05 (m, 8H), 3.87-3.78 (m, 4H), 3.78-3.61 (m, 17H), 3.57 (t, J = 5.7 Hz, 2H), 3.01-2.68 (m, 3H), 2.26-2.08 (m, 1H) 1 H NMR (CDCl 3 , 300 MHz) δ 10.47 (s, 1H), 9.18 (s, 1H), 8.84 (d, J = 8.4 Hz, 1H), 7.72 (t, J = 7.9 Hz, 1H), 7.57 ( d, J = 7.3 Hz, 1H), 5.07-4.92 (m, 1H), 4.29-4.05 (m, 8H), 3.87-3.78 (m, 4H), 3.78-3.61 (m, 17H), 3.57 (t, J = 5.7 Hz, 2H), 3.01-2.68 (m, 3H), 2.26-2.08 (m, 1H)

단계 4: NStep 4: N 1One -(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 1414 -((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)-3,6,9,12-테트라옥사테트라데칸-1,14-디아미드의 제조-((S)-1-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pyrrolidin-1-yl) Preparation of -3,3-dimethyl-1-oxobutan-2-yl)-3,6,9,12-tetraoxatetradecane-1,14-diamide

상기 단계 3에서 제조한 화합물(25 mg, 0.048 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(22 mg, 0.048 mmol), HATU(27 mg, 0.072 mmol)를 DMF 에 녹인 후, DIPEA(0.033 mL, 0.192 mmol)를 가한 후, 실온에서 교반한다. 반응 종결 후 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻고 MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N14-((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)-3,6,9,12-테트라옥사테트라데칸-1,14-디아미드(11 mg, 24%, 아이보리색 고체)를 얻는다.Compound prepared in step 3 (25 mg, 0.048 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4 -(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (22 mg, 0.048 mmol) and HATU (27 mg, 0.072 mmol) were dissolved in DMF, followed by DIPEA (0.033 mL) , 0.192 mmol), and stirred at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, the organic layer was extracted, washed with brine, and water was removed with MgSO 4 , and then concentrated under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-yl)-N 14 -((S)-1-((2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)cover Moyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)-3,6,9,12-tetraoxatetradecane-1,14-diamide (11 mg, 24%, ivory solid).

LC/MS (ESI) m/z [M+H]+ : 935.5 , [M-H]- : 933.6LC/MS (ESI) m/z [M+H] + : 935.5, [MH] - : 933.6

1H NMR (CDCl3, 300MHz) δ 10.50 (s, 1H), 8.87-8.79 (m, 1H), 8.68 (s, 1H), 7.76-7.67 (m, 1H), 7.59-7.52 (m, 1H), 7.51-7.42 (m, 1H), 7.41-7.31 (m, 4H), 4.99-4.88 (m, 1H), 4.81-4.47 (m, 4H), 4.40-4.27 (m, 1H), 4.27-4.06 (m, 4H), 4.06-3.95 (m, 2H), 3.79 (s, 3H), 3.75-3.52 (m, 12H), 2.90-2.59 (m, 3H), 2.56-2.38 (m, 4H), 2.22-2.06 (m, 2H), 2.06-1.94 (m, 1H), 1.88-1.77 (m, 1H), 1.02-0.88 (m, 9H). 1 H NMR (CDCl 3 , 300 MHz) δ 10.50 (s, 1H), 8.87-8.79 (m, 1H), 8.68 (s, 1H), 7.76-7.67 (m, 1H), 7.59-7.52 (m, 1H) , 7.51-7.42 (m, 1H), 7.41-7.31 (m, 4H), 4.99-4.88 (m, 1H), 4.81-4.47 (m, 4H), 4.40-4.27 (m, 1H), 4.27-4.06 ( m, 4H), 4.06-3.95 (m, 2H), 3.79 (s, 3H), 3.75-3.52 (m, 12H), 2.90-2.59 (m, 3H), 2.56-2.38 (m, 4H), 2.22- 2.06 (m, 2H), 2.06-1.94 (m, 1H), 1.88-1.77 (m, 1H), 1.02-0.88 (m, 9H).

<실시예 10> N<Example 10> N 1One -(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 55 -((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)글루타르아미드의 제조-((S)-1-((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3 Preparation of ,3-dimethyl-1-oxobutan-2-yl)glutaramide

Figure 112018098559920-pat00055
Figure 112018098559920-pat00055

단계 1: 5-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)-5-옥소펜탄산의 제조Step 1: Preparation of 5-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)amino)-5-oxopentanoic acid

아미노-2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온(0.1 g, 0.37 mmol)을 아세트산(2 mL)에 녹이고 글루타릭 무수물 (0.046 g, 0.403 mmol), 칼륨 아세테이트(0.108 g, 1.1 mmol)를 첨가한 후, 90℃에서 2시간 동안 교반하였다. 반응이 종결되고, 반응물은 에틸아세테이트로 희석하고 물과 소금물로 씻어주었다. 유기층은 무수 황산마그네슘으로 건조하고, 감압 농축, 분리정제하여 목적 화합물을 제조하였다.Dissolve amino-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (0.1 g, 0.37 mmol) in acetic acid (2 mL) and glutaric anhydride (0.046 g, 0.403 mmol) and potassium acetate (0.108 g, 1.1 mmol) were added, followed by stirring at 90°C for 2 hours. The reaction was terminated, and the reaction was diluted with ethyl acetate and washed with water and brine. The organic layer was dried over anhydrous magnesium sulfate, concentrated under reduced pressure, and purified to prepare a target compound.

1H NMR (300 MHz, DMSO) δ 11.14 (s, 1H), 9.72 (s, 1H), 8.44 (d, J = 8.4 Hz, 1H), 7.90 - 7.78 (m, 1H), 7.62 (d, J = 7.2 Hz, 1H), 5.15 (dd, J = 12.8, 5.4 Hz, 1H), 2.98 - 2.83 (m, 1H), 2.67 - 2.53 (m, 2H), 2.31 (t, J = 7.4 Hz, 2H), 2.24 (t, J = 7.4 Hz, 2H), 2.13 - 2.00 (m, 1H), 1.92 - 1.76 (m, 2H), 1.76 - 1.63 (m, 1H).1H NMR (300 MHz, DMSO) δ 11.14 (s, 1H), 9.72 (s, 1H), 8.44 (d, J = 8.4 Hz, 1H), 7.90-7.78 (m, 1H), 7.62 (d, J = 7.2 Hz, 1H), 5.15 (dd, J = 12.8, 5.4 Hz, 1H), 2.98-2.83 (m, 1H), 2.67-2.53 (m, 2H), 2.31 (t, J = 7.4 Hz, 2H), 2.24 (t, J = 7.4 Hz, 2H), 2.13-2.00 (m, 1H), 1.92-1.76 (m, 2H), 1.76-1.63 (m, 1H).

단계2: NStep 2: N 1One -(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 55 -((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)글루타르아미드의 제조 -((S)-1-((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3 Preparation of ,3-dimethyl-1-oxobutan-2-yl)glutaramide

상기 단계 1에서 제조한 화합물(25 mg, 0.048 mmol), (2S,4R)-1-((S)-2-아미노-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드 염산염(22 mg, 0.048 mmol), HATU(27 mg, 0.072 mmol)를 DMF 에 녹인 후, DIPEA(0.033 mL, 0.192 mmol)를 가한 후, 실온에서 교반한다. 반응 종결 후 농축하여 EA와 물로 희석하여 유기층을 추출하고 염수로 씻고 MgSO4로 물을 제거한 후, 감압농축한다. 실리카겔 컬럼 크로마토그래피(MPLC, MeOH/DCM 6%, 30 min)를 이용하여 분리, 정제하여 N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N5-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)글루타르아미드를 얻는다.Compound prepared in Step 1 (25 mg, 0.048 mmol), (2S,4R)-1-((S)-2-amino-3,3-dimethylbutanoyl)-4-hydroxy-N-(4 -(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide hydrochloride (22 mg, 0.048 mmol) and HATU (27 mg, 0.072 mmol) were dissolved in DMF, followed by DIPEA (0.033 mL) , 0.192 mmol), and stirred at room temperature. After completion of the reaction, the mixture was concentrated, diluted with EA and water, the organic layer was extracted, washed with brine, and water was removed with MgSO4, and then concentrated under reduced pressure. Separation and purification using silica gel column chromatography (MPLC, MeOH/DCM 6%, 30 min) to N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-yl)-N 5 -((S)-1-((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl) Pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutan-2-yl)glutaramide is obtained.

1H NMR (300 MHz, CDCl3) δ 10.25 (s, 0.5H), 10.03 (s, 0.5H), 8.77-8.63 (m, 2H), 7.72-7.61 (m, 1H), 7.61-7.48 (m, 2H), 7.36-7.29 (m, 4H), 6.85 (d, J = 9.0 Hz, 0.5H), 6.77 (d, J = 9.0 Hz, 0.5H), 4.95-4.84 (m, 1H), 4.69-4.51 (m, 4H), 4.31-4.21 (m, 1H), 4.08-3.99 (m, 1H), 3.75-3.63 (m, 2H), 2.87-2.62 (m, 3H), 2.50 (s, 3H), 2.48-2.38 (m, 3H), 2.37-2.23 (m, 2H), 2.19-1.94 (m, 4H), 1.50-1.38 (m, 4H), 0.94 (s, 9H). LC/MS (ESI) m/z 800.3[M+H]+, 798.2[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 10.25 (s, 0.5H), 10.03 (s, 0.5H), 8.77-8.63 (m, 2H), 7.72-7.61 (m, 1H), 7.61-7.48 (m , 2H), 7.36-7.29 (m, 4H), 6.85 (d, J = 9.0 Hz, 0.5H), 6.77 (d, J = 9.0 Hz, 0.5H), 4.95-4.84 (m, 1H), 4.69- 4.51 (m, 4H), 4.31-4.21 (m, 1H), 4.08-3.99 (m, 1H), 3.75-3.63 (m, 2H), 2.87-2.62 (m, 3H), 2.50 (s, 3H), 2.48-2.38 (m, 3H), 2.37-2.23 (m, 2H), 2.19-1.94 (m, 4H), 1.50-1.38 (m, 4H), 0.94 (s, 9H). LC/MS (ESI) m/z 800.3 [M+H] + , 798.2 [MH] -

<실시예 11> N<Example 11> N 1One -((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)메틸)-N-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)methyl)-N 77 -((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)헵탄디아미드의 제조-((S)-1-((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3 Preparation of ,3-dimethyl-1-oxobutan-2-yl)heptanediamide

Figure 112018098559920-pat00056
Figure 112018098559920-pat00056

상기 실시예 10의 단계 1에서 사용한 글루타릭 무수물을 대신하여, 옥소칸-2,8-디온을 사용한 점을 제외하고, 상기 실시예 10과 유사한 방법으로 목적 화합물을 제조하였다.In place of the glutaric anhydride used in Step 1 of Example 10, the target compound was prepared in a similar manner to Example 10, except that oxocan-2,8-dione was used.

1H NMR (500 MHz, CDCl3) δ 8.70 (d, J = 4.7 Hz, 1H), 7.81 (dd, J = 13.9, 6.2 Hz, 3H), 7.72 (t, J = 7.6 Hz, 1H), 7.44 - 7.34 (m, 4H), 7.24 - 7.06 (m, 2H), 5.04 - 4.92 (m, 1H), 4.81 - 4.55 (m, 4H), 4.50 (m, 2H), 4.33 (td, J = 14.8, 4.9 Hz, 1H), 3.98 (t, J = 10.2 Hz, 1H), 3.62 (m, 1H), 2.85 (m, 2H), 2.80 - 2.58 (m, 1H), 2.52 (d, J = 5.7 Hz, 3H), 2.49 - 2.35 (m, 1H), 2.30 - 2.02 (m, 6H), 1.59 - 1.46 (m, 2H), 1.42 (m, 1H), 1.34 - 1.02 (m, 3H), 0.89 (d, J = 8.8 Hz, 9H); LC/MS (ESI) m/z 842 [M+H]+ 1 H NMR (500 MHz, CDCl 3 ) δ 8.70 (d, J = 4.7 Hz, 1H), 7.81 (dd, J = 13.9, 6.2 Hz, 3H), 7.72 (t, J = 7.6 Hz, 1H), 7.44 -7.34 (m, 4H), 7.24-7.06 (m, 2H), 5.04-4.92 (m, 1H), 4.81-4.55 (m, 4H), 4.50 (m, 2H), 4.33 (td, J = 14.8, 4.9 Hz, 1H), 3.98 (t, J = 10.2 Hz, 1H), 3.62 (m, 1H), 2.85 (m, 2H), 2.80-2.58 (m, 1H), 2.52 (d, J = 5.7 Hz, 3H), 2.49-2.35 (m, 1H), 2.30-2.02 (m, 6H), 1.59-1.46 (m, 2H), 1.42 (m, 1H), 1.34-1.02 (m, 3H), 0.89 (d, J = 8.8 Hz, 9H); LC/MS (ESI) m/z 842 [M+H]+

<실시예 12> (2S,4R)-1-((S)-2-(11-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)운데칸아미도)-3.3-디메틸부탄오일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 12> (2S,4R)-1-((S)-2-(11-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-ylamino)undecanamido)-3.3-dimethylbutanyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide Manufacture of

Figure 112018098559920-pat00057
Figure 112018098559920-pat00057

상기 실시예 1의 단계 1에서 사용한 7-아미노헵탄산을 대신하여, 11-아미노운데칸산을 사용한 점을 제외하고, 상기 실시예 1과 같이 수행하여 목적 화합물을 제조하였다.The target compound was prepared in the same manner as in Example 1, except that 11-aminoundecanoic acid was used instead of 7-aminoheptanoic acid used in Step 1 of Example 1.

1H NMR (300 MHz, CDCl3) δ 9.92 (s, 0.5H), 9.86 (s, 0.5H), 8.69 (s, 1H), 7.56-7.44 (m, 2H), 7.40-7.30 (m, 4H), 7.08 (d, J = 7.1 Hz, 1H), 6.92-6.84 (m, 1H), 6.67-6.55 (m, 1H), 6.30-6.20 (m, 1H), 4.96-4.79 (m, 1H), 4.73-4.47 (m, 4H), 4.36-4.23 (m, 1H), 4.15-4.04 (m, 1H), 3.67-3.57 (m, 1H), 3.33-3.19 (m, 2H), 2.89-2.59 (m, 3H), 2.57-2.46 (m, 4H), 2.22-1.98 (m, 4H), 1.70-1.59 (m, 2H), 1.59-1.47 (m, 2H), 1.47-1.36 (s, 2H), 1.36-1.14 (m, 12H), 0.98-0.80 (m, 9H). LC/MS (ESI) m/z 871.7[M+H]+, 869.8[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 9.92 (s, 0.5H), 9.86 (s, 0.5H), 8.69 (s, 1H), 7.56-7.44 (m, 2H), 7.40-7.30 (m, 4H ), 7.08 (d, J = 7.1 Hz, 1H), 6.92-6.84 (m, 1H), 6.67-6.55 (m, 1H), 6.30-6.20 (m, 1H), 4.96-4.79 (m, 1H), 4.73-4.47 (m, 4H), 4.36-4.23 (m, 1H), 4.15-4.04 (m, 1H), 3.67-3.57 (m, 1H), 3.33-3.19 (m, 2H), 2.89-2.59 (m , 3H), 2.57-2.46 (m, 4H), 2.22-1.98 (m, 4H), 1.70-1.59 (m, 2H), 1.59-1.47 (m, 2H), 1.47-1.36 (s, 2H), 1.36 -1.14 (m, 12H), 0.98-0.80 (m, 9H). LC/MS (ESI) m/z 871.7 [M+H] + , 869.8 [MH] -

<실시예 13> (2S,4R)-1-((S)-2-(8-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)옥탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 13> (2S,4R)-1-((S)-2-(8-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-ylamino)octanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carbox Preparation of amide

Figure 112018098559920-pat00058
Figure 112018098559920-pat00058

상기 실시예 1의 단계 1에서 사용한 7-아미노헵탄산을 대신하여, 8-아미노옥탄산을 사용한 점을 제외하고, 상기 실시예 1과 같이 수행하여 목적 화합물을 제조하였다.The target compound was prepared in the same manner as in Example 1, except that 8-aminooctanoic acid was used instead of 7-aminoheptanoic acid used in Step 1 of Example 1.

1H NMR (300 MHz, CDCl3) δ 10.63 (s, 0.5H), 9.63 (s, 0.5H), 8.68 (s, 1H), 7.76-7.67 (m, 0.5H), 7.55-7.44 (m, 1.5H), 7.41-7.29 (m, 4H), 7.12-7.04 (m, 1H), 6.95-6.84 (m, 1.5H), 6.57 (d, J = 9.1 Hz, 0.5H), 6.32-6.21 (m, 1H), 4.96-4.82 (m, 1H), 4.72-4.50 (m, 4H), 4.36-4.22 (m, 1H), 4.17-4.03 (m, 1H), 3.63 (dd, J = 11.4, 3.5 Hz, 1H), 3.40-3.18 (m, 2H), 2.88-2.60 (m, 3H), 2.58-2.44 (m, 4H), 2.25-1.98 (m, 4H), 1.84-1.70 (m, 2H), 1.70-1.50 (m, 4H), 1.50-1.19 (m, 5H), 0.99-0.80 (m, 9H). LC/MS (ESI) m/z 829.7[M+H]+, 827.8[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 10.63 (s, 0.5H), 9.63 (s, 0.5H), 8.68 (s, 1H), 7.76-7.67 (m, 0.5H), 7.55-7.44 (m, 1.5H), 7.41-7.29 (m, 4H), 7.12-7.04 (m, 1H), 6.95-6.84 (m, 1.5H), 6.57 (d, J = 9.1 Hz, 0.5H), 6.32-6.21 (m , 1H), 4.96-4.82 (m, 1H), 4.72-4.50 (m, 4H), 4.36-4.22 (m, 1H), 4.17-4.03 (m, 1H), 3.63 (dd, J = 11.4, 3.5 Hz , 1H), 3.40-3.18 (m, 2H), 2.88-2.60 (m, 3H), 2.58-2.44 (m, 4H), 2.25-1.98 (m, 4H), 1.84-1.70 (m, 2H), 1.70 -1.50 (m, 4H), 1.50-1.19 (m, 5H), 0.99-0.80 (m, 9H). LC/MS (ESI) m/z 829.7 [M+H] + , 827.8 [MH] -

<실시예 14> (2S,4R)-1-((S)-2-(12-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)도데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 14> (2S,4R)-1-((S)-2-(12-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-ylamino)dodecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-car Preparation of Voxamide

Figure 112018098559920-pat00059
Figure 112018098559920-pat00059

상기 실시예 1의 단계 1에서 사용한 7-아미노헵탄산을 대신하여, 12-아미노도데칸산을 사용한 점을 제외하고, 상기 실시예 1과 같이 수행하여 목적 화합물을 제조하였다.The target compound was prepared in the same manner as in Example 1, except that 12-aminododecanoic acid was used instead of the 7-aminoheptanoic acid used in Step 1 of Example 1.

1H NMR (300 MHz, CDCl3) δ 10.14 (s, 0.5H), 9.96 (s, 0.5H), 8.69 (s, 1H), 7.64-7.42 (m, 2H), 7.42-7.30 (m, 4H), 7.13-7.03 (m, 1H), 6.87 (d, J = 8.6 Hz, 1H), 6.71 (d, J = 9.0 Hz, 0.5H), 6.62 (d, J = 9.0 Hz, 0.5H), 6.24 (s, 1H), 4.96-4.76 (m, 1H), 4.72-4.48 (m, 3H), 4.36-4.21 (m, 1H), 4.12-4.00 (m, 1H), 3.70-3.58 (m, 1H), 3.34-3.19 (m, 2H), 2.88-2.58 (m, 3H), 2.58-2.40 (m, 4H), 2.22-1.94 (m, 4H), 1.72-1.47 (m, 4H), 1.47-1.08 (m, 14H), 0.99-0.77 (m, 9H). LC/MS (ESI) m/z 885.7[M+H]+, 883.7[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 10.14 (s, 0.5H), 9.96 (s, 0.5H), 8.69 (s, 1H), 7.64-7.42 (m, 2H), 7.42-7.30 (m, 4H ), 7.13-7.03 (m, 1H), 6.87 (d, J = 8.6 Hz, 1H), 6.71 (d, J = 9.0 Hz, 0.5H), 6.62 (d, J = 9.0 Hz, 0.5H), 6.24 (s, 1H), 4.96-4.76 (m, 1H), 4.72-4.48 (m, 3H), 4.36-4.21 (m, 1H), 4.12-4.00 (m, 1H), 3.70-3.58 (m, 1H) , 3.34-3.19 (m, 2H), 2.88-2.58 (m, 3H), 2.58-2.40 (m, 4H), 2.22-1.94 (m, 4H), 1.72-1.47 (m, 4H), 1.47-1.08 ( m, 14H), 0.99-0.77 (m, 9H). LC/MS (ESI) m/z 885.7 [M+H] + , 883.7 [MH] -

<실시예 15> (2S,4R)-1-((S)-2-(2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥실옥시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 15> (2S,4R)-1-((S)-2-(2-(6-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-ylamino)hexyloxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pi Preparation of lollidine-2-carboxamide

Figure 112018098559920-pat00060
Figure 112018098559920-pat00060

상기 실시예 1의 단계 1에서 사용한 7-아미노헵탄산을 대신하여, 2-(6-아미노헥실옥시)아세트산을 사용한 점을 제외하고, 상기 실시예 1과 같이 수행하여 목적 화합물을 제조하였다.The target compound was prepared in the same manner as in Example 1, except that 2-(6-aminohexyloxy)acetic acid was used instead of 7-aminoheptanoic acid used in Step 1 of Example 1.

1H NMR (300 MHz, CDCl3) δ 9.42 (s, 0.5H), 9.19 (s, 0.5H), 8.69 (s, 1H), 7.54 - 7.38 (m, 2H), 7.38 - 7.30 (m, 4H), 7.25 - 7.17 (m, 1H), 7.07 (d, J = 7.1 Hz, 1H), 6.86 (d, J = 8.5 Hz, 1H), 6.32 - 6.19 (m, 1H), 4.97 - 4.84 (m, 1H), 4.76 - 4.63 (m, 1H), 4.61 - 4.44 (m, 3H), 4.38 - 4.24 (m, 1H), 4.07 (d, J = 10.9 Hz, 1H), 3.91 (s, 2H), 3.69 - 3.59 (m, 1H), 3.56 - 3.37 (m, 3H), 3.33 - 3.18 (m, 2H), 2.89 - 2.62 (m, 3H), 2.56 - 2.39 (m, 4H), 2.19 - 2.01 (m, 2H), 1.93 (s, 1H), 1.76 - 1.53 (m, 4H), 1.53 - 1.34 (m, 4H), 0.95 (s, 9H). LC/MS (ESI) m/z 844.8[M+H]+, 842.7[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 9.42 (s, 0.5H), 9.19 (s, 0.5H), 8.69 (s, 1H), 7.54-7.38 (m, 2H), 7.38-7.30 (m, 4H ), 7.25-7.17 (m, 1H), 7.07 (d, J = 7.1 Hz, 1H), 6.86 (d, J = 8.5 Hz, 1H), 6.32-6.19 (m, 1H), 4.97-4.84 (m, 1H), 4.76-4.63 (m, 1H), 4.61-4.44 (m, 3H), 4.38-4.24 (m, 1H), 4.07 (d, J = 10.9 Hz, 1H), 3.91 (s, 2H), 3.69 -3.59 (m, 1H), 3.56-3.37 (m, 3H), 3.33-3.18 (m, 2H), 2.89-2.62 (m, 3H), 2.56-2.39 (m, 4H), 2.19-2.01 (m, 2H), 1.93 (s, 1H), 1.76-1.53 (m, 4H), 1.53-1.34 (m, 4H), 0.95 (s, 9H). LC/MS (ESI) m/z 844.8 [M+H] + , 842.7 [MH] -

<실시예 16> (2S,4R)-1-((S)-2-(10-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 16> (2S,4R)-1-((S)-2-(10-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 4-ylamino)decanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carbox Preparation of amide

Figure 112018098559920-pat00061
Figure 112018098559920-pat00061

상기 실시예 1의 단계 1에서 사용한 7-아미노헵탄산을 대신하여, 10-아미노데칸산을 사용한 점을 제외하고, 상기 실시예 1과 같이 수행하여 목적 화합물을 제조하였다.The target compound was prepared in the same manner as in Example 1, except that 10-aminodecanoic acid was used instead of the 7-aminoheptanoic acid used in Step 1 of Example 1.

1H NMR (300 MHz, CDCl3) δ 10.24 (s, 0.5H), 10.16 (s, 0.5H), 7.65-7.53 (m, 1H), 7.48 (t, J = 7.8 Hz, 1H), 7.42-7.30 (m, 4H), 7.07 (d, J = 7.1 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.71 (dd, J = 9.1, 6.7 Hz, 1H), 6.31-6.18 (m, 1H), 4.97-4.81 (m, 1H), 4.73-4.49 (m, 4H), 4.36-4.24 (m, 1H), 4.11-4.03 (m, 1H), 3.68-3.57 (m, 1H), 3.35-3.21 (m, 2H), 2.88-2.58 (m, 3H), 2.56-2.43 (m, 4H), 2.22-2.08 (m, 3H), 1.99-1.81 (m, 2H), 1.73-1.60 (m, 2H), 1.60-1.45 (m, 2H), 1.45-1.37 (m, 2H), 1.37-1.12 (m, 10H), 0.99-0.77 (m, 9H). LC/MS (ESI) m/z 857.1[M+H]+, 855.0[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 10.24 (s, 0.5H), 10.16 (s, 0.5H), 7.65-7.53 (m, 1H), 7.48 (t, J = 7.8 Hz, 1H), 7.42- 7.30 (m, 4H), 7.07 (d, J = 7.1 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.71 (dd, J = 9.1, 6.7 Hz, 1H), 6.31-6.18 (m , 1H), 4.97-4.81 (m, 1H), 4.73-4.49 (m, 4H), 4.36-4.24 (m, 1H), 4.11-4.03 (m, 1H), 3.68-3.57 (m, 1H), 3.35 -3.21 (m, 2H), 2.88-2.58 (m, 3H), 2.56-2.43 (m, 4H), 2.22-2.08 (m, 3H), 1.99-1.81 (m, 2H), 1.73-1.60 (m, 2H), 1.60-1.45 (m, 2H), 1.45-1.37 (m, 2H), 1.37-1.12 (m, 10H), 0.99-0.77 (m, 9H). LC/MS (ESI) m/z 857.1 [M+H] + , 855.0 [MH] -

<실시예 17> (2S,4R)-1-((S)-2-tert-부틸-14-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-4-옥소-6,9,12-트리옥사-3-아자테트라데카-1-노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 17> (2S,4R)-1-((S)-2-tert-butyl-14-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindoline-4-ylamino)-4-oxo-6,9,12-trioxa-3-azatetradeca-1-noyl)-4-hydroxy-N-(4-(4-methylthiazole- Preparation of 5-yl)benzyl)pyrrolidine-2-carboxamide

Figure 112018098559920-pat00062
Figure 112018098559920-pat00062

상기 실시예 4의 단계 1에서 사용한, tert-부틸 14-요오도-3,6,9,12-테트라옥사테트라데카-1-노에이트를 대신하여, tert-부틸 2-(2-(2-(2-요오도에톡시)에톡시)에톡시)아세테이트를 사용한 점을 제외하고, 상기 실시예 4와 같이 수행하여 목적 화합물을 제조하였다.In place of tert-butyl 14-iodo-3,6,9,12-tetraoxatetradeca-1-noate, used in step 1 of Example 4 above, tert-butyl 2-(2-(2- Except that (2-iodoethoxy)ethoxy)ethoxy)acetate was used, the same procedure as in Example 4 was carried out to prepare the target compound.

1H NMR (300 MHz, CDCl3) δ 9.81-9.72 (m, 1H), 8.70 (s, 1H), 7.57-7.46 (m, 2H), 7.41-7.31 (m, 5H), 7.11 (d, J = 7.1 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 6.54 (d, J = 4.1 Hz, 1H), 4.95-4.82 (m, 1H), 4.76-4.66 (m, 1H), 4.66-4.50 (m, 3H), 4.38-4.27 (m, 1H), 4.19-3.92 (m, 3H), 3.78-3.59 (m, 11H), 3.51-3.37 (m, 2H), 2.90-2.62 (m, 3H), 2.53 (d, J = 1.2 Hz, 3H), 2.52-2.41 (m, 1H), 2.22-2.09 (m, 1H), 1.91 (brs, merged with H2O, 1H), 0.97 (s, 9H). LC/MS (ESI) m/z 877.1[M+H]+, 875.0[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 9.81-9.72 (m, 1H), 8.70 (s, 1H), 7.57-7.46 (m, 2H), 7.41-7.31 (m, 5H), 7.11 (d, J = 7.1 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 6.54 (d, J = 4.1 Hz, 1H), 4.95-4.82 (m, 1H), 4.76-4.66 (m, 1H), 4.66 -4.50 (m, 3H), 4.38-4.27 (m, 1H), 4.19-3.92 (m, 3H), 3.78-3.59 (m, 11H), 3.51-3.37 (m, 2H), 2.90-2.62 (m, 3H), 2.53 (d, J = 1.2 Hz, 3H), 2.52-2.41 (m, 1H), 2.22-2.09 (m, 1H), 1.91 (brs, merged with H2O, 1H), 0.97 (s, 9H) . LC/MS (ESI) m/z 877.1 [M+H] + , 875.0 [MH] -

<실시예 18> (2S,4R)-1-((S)-2-(11-((3-(2,6- 디옥소피페리딘-3-일)-2-메틸-4-옥소-3,4-디히드로퀴나졸린-5-일)아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카르복스아미드의 제조<Example 18> (2S,4R)-1-((S)-2-(11-((3-(2,6-dioxopiperidin-3-yl)-2-methyl-4-oxo- 3,4-dihydroquinazolin-5-yl)amino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl )Benzyl) Preparation of pyrrolidine-2-carboxamide

Figure 112018098559920-pat00063
Figure 112018098559920-pat00063

상기 실시예 1의 단계 1에서 사용한 7-아미노헵탄산을 대신하여, 11-아미노운데칸산을 사용사고, 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온을 대신하여, 3-(5-플루오로-2-메틸-4-옥소퀴나졸린-3(4H)-yl)피페리딘-2,6-디온을 사용한 점을 제외하고, 상기 실시예 1과 같이 수행하여 목적 화합물을 제조하였다.Instead of the 7-aminoheptanoic acid used in Step 1 of Example 1, 11-aminoundecanoic acid was used, and 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindole Except that 3-(5-fluoro-2-methyl-4-oxoquinazoline-3(4H)-yl)piperidine-2,6-dione was used instead of -1,3-dione , Was carried out as in Example 1 to prepare a target compound.

1H NMR (300 MHz, CD3OD) δ 8.89 (s, 1H), 7.85-7.77 (m, 1H), 7.54-7.40 (m, 4H), 6.69 (d, J = 7.9 Hz, 1H), 6.55 (d, J = 8.5 Hz, 1H), 5.21-5.13 (m, 1H), 4.69-4.63 (m, 1H), 4.63-4.48 (m, 3H), 4.42-4.32 (m, 1H), 3.92 (d, J = 11.1 Hz, 1H), 3.82 (dd, J = 11.0, 3.9 Hz, 1H), 3.20 (t, J = 6.9 Hz, 2H), 2.92-2.70 (m, 4H), 2.63 (s, 3H), 2.49 (s, 3H), 2.34-2.16 (m, 5H), 2.16-2.06 (m, 1H), 1.76-1.55 (m, 5H), 1.50-1.41 (m, 3H), 1.41-1.31 (m, 12H), 1.05 (s, 9H), 0.95-0.89 (m, 1H). LC/MS (ESI) m/z 884.1[M+H]+, 882.1[M-H]- 1 H NMR (300 MHz, CD 3 OD) δ 8.89 (s, 1H), 7.85-7.77 (m, 1H), 7.54-7.40 (m, 4H), 6.69 (d, J = 7.9 Hz, 1H), 6.55 (d, J = 8.5 Hz, 1H), 5.21-5.13 (m, 1H), 4.69-4.63 (m, 1H), 4.63-4.48 (m, 3H), 4.42-4.32 (m, 1H), 3.92 (d , J = 11.1 Hz, 1H), 3.82 (dd, J = 11.0, 3.9 Hz, 1H), 3.20 (t, J = 6.9 Hz, 2H), 2.92-2.70 (m, 4H), 2.63 (s, 3H) , 2.49 (s, 3H), 2.34-2.16 (m, 5H), 2.16-2.06 (m, 1H), 1.76-1.55 (m, 5H), 1.50-1.41 (m, 3H), 1.41-1.31 (m, 12H), 1.05 (s, 9H), 0.95-0.89 (m, 1H). LC/MS (ESI) m/z 884.1 [M+H] + , 882.1 [MH] -

<실시예 19> 4,4'-(노난-1,9-디일비스(아자네디일))비스(2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온)의 제조<Example 19> 4,4'-(nonane-1,9-diylbis(azanediyl))bis(2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3 -Dion)

Figure 112018098559920-pat00064
Figure 112018098559920-pat00064

단계 1: tert-부틸 (9-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)노닐)카바메이트의 제조Step 1: Preparation of tert-butyl (9-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)amino)nonyl)carbamate

tert-부틸 (9-아미노노닐)카바메이트(212 mg, 0.82 mmol), 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온(227 mg, 0.82 mmol), DIPEA(0.429 mL, 2.46 mmol)을 DMF(3 mL)에 녹인 후, 90℃에서 가열하였다. 반응물은 에틸아세테이트 용액으로 희석한 후, 물로 씻어준다. 유기층은 소금물로 씻은 후, 무수황산마그네슘으로 건조하고 농축하고, 컬럼크로마토그래피법을 이용하여 정제하여 목적 화합물을 얻었다.tert-butyl (9-aminononyl)carbamate (212 mg, 0.82 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindole-1,3-dione ( 227 mg, 0.82 mmol) and DIPEA (0.429 mL, 2.46 mmol) were dissolved in DMF (3 mL) and heated at 90°C. The reaction was diluted with ethyl acetate solution, and then washed with water. The organic layer was washed with brine, dried over anhydrous magnesium sulfate, concentrated, and purified by column chromatography to obtain the target compound.

1H NMR (300 MHz, CDCl3)δ8.14(s,1H),7.49(dd,J = 8.5, 7.1 Hz, 1H), 7.09 (d, J = 7.0 Hz, 1H), 6.88 (d, J = 8.6 Hz, 1H), 6.23 (t, J = 5.5 Hz, 1H), 4.97-4.86 (m, 1H), 4.52 (s, 1H), 3.26 (q, J = 6.6 Hz, 2H), 3.10 (q, J = 6.5 Hz, 2H), 2.94-2.65 (m, 3H), 2.19-2.09 (m, 1H), 1.72-1.63 (m, 2H), 1.55-1.38 (m, 13H), 1.38-1.23 (m, 8H). 1 H NMR (300 MHz, CDCl 3 )δ8.14(s,1H),7.49(dd,J = 8.5, 7.1 Hz, 1H), 7.09 (d, J = 7.0 Hz, 1H), 6.88 (d, J = 8.6 Hz, 1H), 6.23 (t, J = 5.5 Hz, 1H), 4.97-4.86 (m, 1H), 4.52 (s, 1H), 3.26 (q, J = 6.6 Hz, 2H), 3.10 (q , J = 6.5 Hz, 2H), 2.94-2.65 (m, 3H), 2.19-2.09 (m, 1H), 1.72-1.63 (m, 2H), 1.55-1.38 (m, 13H), 1.38-1.23 (m , 8H).

단계 2: 4-((9-아미노노닐)아미노)-2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온 염산염의 제조Step 2: Preparation of 4-((9-aminononyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione hydrochloride

tert-부틸 (9-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)아미노)노닐)카바메이트(0.112 g, 0.218 mmol), 4 N HCl/1,4-디옥산 용액(2 mL), 디클로로메탄(1 mL) 혼합용액을 상온에서 교반한 후, 농축하여 4-((9-아미노노닐)아미노)-2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온 염산염을 얻었다.tert-butyl (9-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)nonyl)carbamate (0.112 g, 0.218 mmol) ), 4 N HCl/1,4-dioxane solution (2 mL), dichloromethane (1 mL) The mixed solution was stirred at room temperature, and then concentrated to 4-((9-aminononyl)amino)-2- (2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione hydrochloride was obtained.

단계 3: 4,4'-(노난-1,9-디일비스(아자네디일))비스(2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온)의 제조Step 3: 4,4'-(nonane-1,9-diylbis(azanediyl))bis(2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione )

4-((9-아미노노닐)아미노)-2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온 염산염(63 mg, 0.14 mmol), 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온(39 mg, 0.14 mmol), DIPEA(0.073 mL, 0.42 mmol)을 DMF(1 mL)에 녹인 후, 90℃에서 가열한다. 반응물은 물에 희석한 후, 에틸아세테이트로 추출하고 소금물로 씻어주었다. 유기층은 무수황산마그네슘으로 건조, 농축한 후, 컬럼크로마토그래피로 분리정제하여 4,4'-(노난-1,9-디일비스(아자네디일))비스(2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온)을 얻었다.4-((9-aminononyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione hydrochloride (63 mg, 0.14 mmol), 2-( 2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-1,3-dione (39 mg, 0.14 mmol), DIPEA (0.073 mL, 0.42 mmol) dissolved in DMF (1 mL) Then, it is heated at 90°C. The reaction was diluted with water, extracted with ethyl acetate and washed with brine. The organic layer was dried and concentrated with anhydrous magnesium sulfate, and then separated and purified by column chromatography to obtain 4,4'-(nonane-1,9-diylbis(azanediyl))bis(2-(2,6-dioxophyte) Peridin-3-yl)isoindoline-1,3-dione) was obtained.

1H NMR (300 MHz, CDCl3) δ 8.42 (s, 1H), 8.29 (s, 1H), 7.48 (dd, J = 8.5, 7.1 Hz, 2H), 7.08 (d, J = 7.1 Hz, 2H), 6.87 (d, J = 8.5 Hz, 2H), 6.24 (t, J = 5.5 Hz, 2H), 4.97-4.86 (m, 2H), 3.26 (q, J = 6.6 Hz, 4H), 2.92-2.66 (m, 6H), 2.18-2.08 (m, 2H), 1.72-1.59 (m, 4H), 1.49-1.30 (m, 10H). LC/MS (ESI) m/z 671.8[M+H]+, 669.0[M-H]- 1 H NMR (300 MHz, CDCl 3 ) δ 8.42 (s, 1H), 8.29 (s, 1H), 7.48 (dd, J = 8.5, 7.1 Hz, 2H), 7.08 (d, J = 7.1 Hz, 2H) , 6.87 (d, J = 8.5 Hz, 2H), 6.24 (t, J = 5.5 Hz, 2H), 4.97-4.86 (m, 2H), 3.26 (q, J = 6.6 Hz, 4H), 2.92-2.66 ( m, 6H), 2.18-2.08 (m, 2H), 1.72-1.59 (m, 4H), 1.49-1.30 (m, 10H). LC/MS (ESI) m/z 671.8 [M+H] + , 669.0 [MH] -

<실시예 20> (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 20> (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindolin-4-ylamino)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl) Preparation of pyrrolidine-2-carboxamide

Figure 112018098559920-pat00065
Figure 112018098559920-pat00065

단계 1: tert-부틸 11-아미노운데카노에이트의 제조Step 1: Preparation of tert-butyl 11-aminoundecanoate

티오닐 클로라이드 (24.80 ml, 24.8 mmol) 중 11-아미노운데칸산 (500 mg, 2.48 mmol)의 용액을 실온에서 1.5 시간 동안 교반하고, 그 후 과량의 티오닐 클로라이드를 진공하에 제거하였다. 이 잔류물에 tBuOH (6 ml) 중 NaHCO3 (458 mg, 5.45 mmol)의 용액을 교반하에 첨가하고, 반응 혼합물을 밤새 실온에서 교반하였다. 휘발성 물질을 진공하에 제거하고, 잔류물을 EtOAc (30 ml) 및 NaOH (1M, 수성) (20 ml)에 취하였다. 합한 유기층을 물 (30 ml) 및 염수 (20 ml ×3)로 세척하고, MgSO4상에서 건조시키고, 용매를 진공하에 제거하여 단계 1의 목적 화합물 (521 mg, 2.02 mmol, 81 %)을 황색 투명 오일로서 수득하였다.A solution of 11-aminoundecanoic acid (500 mg, 2.48 mmol) in thionyl chloride (24.80 ml, 24.8 mmol) was stirred at room temperature for 1.5 hours, after which excess thionyl chloride was removed under vacuum. To this residue was added a solution of NaHCO 3 (458 mg, 5.45 mmol) in tBuOH (6 ml) under stirring, and the reaction mixture was stirred overnight at room temperature. The volatiles were removed under vacuum, and the residue was taken into EtOAc (30 ml) and NaOH (1M, aqueous) (20 ml). The combined organic layer was washed with water (30 ml) and brine (20 ml × 3), dried over MgSO 4 and the solvent removed in vacuo to give the desired compound of step 1 (521 mg, 2.02 mmol, 81%) as a yellow transparent. Obtained as an oil.

단계 2: tert-부틸 11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)아세트아미도)운데카노에이트의 제조Step 2: tert-Butyl 11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-ylamino)acetamido)undecanoate Manufacture of

DMF 중 상기 단계 1에서 제조한 화합물 (50 ㎎, 0.194 mmol)의 용액에, 4- 글리신 탈리도마이드 (64 ㎎, 0.194 mmol), HATU (110 ㎎, 0.291 mmol), DIPEA (0.135 mL, 0.776 mmol)을 첨가하고, 실온에서 교반 2 시간 동안 교반하였다. 반응 혼합물을 물로 희석하고 EtOAc로 추출하였다. 합쳐진 유기층을 염수로 세척하고 MgSO4상에서 건조시키고 용매를 진공하에 제거하여 오일을 수득하였다. 조질의 혼합물을 EtOAc / Hex = 55 %를 사용하여 실리카겔 컬럼 크로마토그래피로 정제하여 단계 2의 목적 화합물 (71mg, 혼합물)을 황색 고체로서 수득하였다.To a solution of the compound (50 mg, 0.194 mmol) prepared in step 1 above in DMF, 4-glycine thalidomide (64 mg, 0.194 mmol), HATU (110 mg, 0.291 mmol), DIPEA (0.135 mL, 0.776 mmol) It was added and stirred at room temperature for 2 hours. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO 4 and the solvent was removed in vacuo to give an oil. The crude mixture was purified by silica gel column chromatography using EtOAc/Hex=55% to give the desired compound of step 2 (71 mg, mixture) as a yellow solid.

단계 3: 11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)아세트아미도)운데칸산의 제조Step 3: Preparation of 11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-ylamino)acetamido)undecanoic acid

DCM (2 ml) 중 상기 단계 2에서 제조한 화합물 (71 mg, 0.124 mmol)의 용액에, TFA (2 ml)를 첨가하고, 생성된 혼합물을 실온에서 3 시간 동안 교반하였다. 반응 혼합물을 진공 농축하여 오일을 수득하였다. 조 혼합물을 DCM / MeOH = 7 %를 사용하여 실리카겔 컬럼 크로마토그래피로 정제하여 단계 3의 목적 화합물 (44mg, 0.085mmol, 단계 2-3 동안 44 %)을 황색 고체로서 수득하였다.To a solution of the compound (71 mg, 0.124 mmol) prepared in step 2 above in DCM (2 ml), TFA (2 ml) was added and the resulting mixture was stirred at room temperature for 3 hours. The reaction mixture was concentrated in vacuo to an oil. The crude mixture was purified by silica gel column chromatography using DCM/MeOH=7% to give the desired compound of step 3 (44mg, 0.085mmol, 44% during steps 2-3) as a yellow solid.

단계 4: (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 4: (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolid Preparation of Dean-2-carboxamide

DMF 중 상기 단계 3에서 제조한 화합물 (30mg, 0.058mmol)의 용액에, VHL 리간드 (27mg, 0.058mmol), HATU (33mg, 0.087mmol), DIPEA (0.040mL, 0.232mmol)를 첨가하고 실온에서 밤새 교반하였다. 반응 혼합물을 물로 희석하고 EtOAc로 추출하였다. 합쳐진 유기층을 염수로 세척하고 MgSO4상에서 건조시키고 용매를 진공하에 제거하여 오일을 수득하였다. 조 혼합물을 MeOH / DCM = 5 %를 사용하여 PREP TLC상에서 정제하여 최종 목적 화합물 (10mg, 0.011mmol, 18 %)을 황색 고체로서 수득하였다.To the solution of the compound (30mg, 0.058mmol) prepared in step 3 above in DMF, VHL ligand (27mg, 0.058mmol), HATU (33mg, 0.087mmol), DIPEA (0.040mL, 0.232mmol) were added overnight at room temperature. It was stirred. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO 4 and the solvent was removed in vacuo to give an oil. The crude mixture was purified on PREP TLC using MeOH / DCM = 5% to give the final target compound (10mg, 0.011mmol, 18%) as a yellow solid.

1H NMR (500 MHz, Chloroform-d) δ 8.68 (d, J = 6.2 Hz, 1H), 7.62-7.52 (m, 2H), 7.40-7.32 (m, 4H), 7.22 (t, J = 7.8 Hz, 1H), 6.84 (t, J = 7.8 Hz, 1H), 6.70-6.56 (m, 2H), 4.93-4.84 (m, 1H), 4.72-4.53 (m, 4H), 4.31-4.22 (m, 1H), 4.08 (d, J = 11.5 Hz, 1H), 4.05-3.87 (m, 2H), 3.70-3.62 (m, 1H), 2.86-2.60 (m, 3H), 2.50 (s, 3H), 2.50-2.44 (m, 1H), 2.22-2.01 (m, 4H), 1.60-1.48 (m, 2H), 1.49-1.38 (m, 2H), 1.34-1.23 (m, 3H), 1.23-0.97 (m, 13H), 0.93 (s, 9H). , LC/MS (ESI) m/z 927.2 [M + H]+, 925.1 [M - H]- 1 H NMR (500 MHz, Chloroform- d ) δ 8.68 (d, J = 6.2 Hz, 1H), 7.62-7.52 (m, 2H), 7.40-7.32 (m, 4H), 7.22 (t, J = 7.8 Hz , 1H), 6.84 (t, J = 7.8 Hz, 1H), 6.70-6.56 (m, 2H), 4.93-4.84 (m, 1H), 4.72-4.53 (m, 4H), 4.31-4.22 (m, 1H) ), 4.08 (d, J = 11.5 Hz, 1H), 4.05-3.87 (m, 2H), 3.70-3.62 (m, 1H), 2.86-2.60 (m, 3H), 2.50 (s, 3H), 2.50- 2.44 (m, 1H), 2.22-2.01 (m, 4H), 1.60-1.48 (m, 2H), 1.49-1.38 (m, 2H), 1.34-1.23 (m, 3H), 1.23-0.97 (m, 13H) ), 0.93 (s, 9H). , LC/MS (ESI) m/z 927.2 [M + H] + , 925.1 [M-H] -

<실시예 21> (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일옥시)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 21> (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoyi Soindolin-4-yloxy)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl) Preparation of pyrrolidine-2-carboxamide

Figure 112018098559920-pat00066
Figure 112018098559920-pat00066

단계 1: tert-부틸 11-아미노운데카노에이트의 제조Step 1: Preparation of tert-butyl 11-aminoundecanoate

티오닐 클로라이드 (24.80 ml, 24.8 mmol) 중 11-아미노운데칸산 (500 mg, 2.48 mmol)의 용액을 실온에서 1.5 시간 동안 교반하고, 그 후 과량의 티오닐 클로라이드를 진공하에 제거하였다. 이 잔류물에 tBuOH (6 ml) 중 NaHCO3 (458 mg, 5.45 mmol)의 용액을 교반하에 첨가하고, 반응 혼합물을 밤새 실온에서 교반하였다. 휘발성 물질을 진공하에 제거하고, 잔류물을 EtOAc (30 ml) 및 NaOH (1M, 수성) (20 ml)에 취하였다. 합한 유기층을 물 (30 ml) 및 염수 (20 ml ×3)로 세척하고, MgSO4상에서 건조시키고, 용매를 진공하에 제거하여 단계 1의 목적 화합물 (521 mg, 2.02 mmol, 81 %)을 황색 투명 오일로서 수득하였다.A solution of 11-aminoundecanoic acid (500 mg, 2.48 mmol) in thionyl chloride (24.80 ml, 24.8 mmol) was stirred at room temperature for 1.5 hours, after which excess thionyl chloride was removed under vacuum. To this residue was added a solution of NaHCO 3 (458 mg, 5.45 mmol) in tBuOH (6 ml) under stirring, and the reaction mixture was stirred overnight at room temperature. The volatiles were removed under vacuum, and the residue was taken into EtOAc (30 ml) and NaOH (1M, aqueous) (20 ml). The combined organic layer was washed with water (30 ml) and brine (20 ml × 3), dried over MgSO 4 and the solvent removed in vacuo to give the desired compound of step 1 (521 mg, 2.02 mmol, 81%) as a yellow transparent. Obtained as an oil.

단계 2: tert-부틸 11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일옥시)아세트아미도)운데카노에이트의 제조Step 2: tert-Butyl 11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yloxy)acetamido)undecanoate Manufacture of

DMF 중 상기 단계 1에서 제조한 화합물 (50 ㎎, 0.194 mmol)의 용액에, 4- 글리콜산 탈리도마이드 (64 ㎎, 0.194 mmol), HATU (110 ㎎, 0.291 mmol), DIPEA (0.135 mL, 0.776 mmol)을 첨가하고, 실온에서 교반 2 시간 동안 교반하였다. 반응 혼합물을 물로 희석하고 EtOAc로 추출하였다. 합쳐진 유기층을 염수로 세척하고 MgSO4상에서 건조시키고 용매를 진공하에 제거하여 오일을 수득하였다. 조질의 혼합물을 EtOAc / Hex = 55 %를 사용하여 실리카겔 컬럼 크로마토그래피로 정제하여 단계 2의 목적 화합물 (48mg, 혼합물)을 베이지색 고체로서 수득하였다.To a solution of the compound (50 mg, 0.194 mmol) prepared in step 1 above in DMF, 4-glycolic acid thalidomide (64 mg, 0.194 mmol), HATU (110 mg, 0.291 mmol), DIPEA (0.135 mL, 0.776 mmol) Was added and stirred at room temperature for 2 hours. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO 4 and the solvent was removed in vacuo to give an oil. The crude mixture was purified by silica gel column chromatography using EtOAc/Hex=55% to give the desired compound of step 2 (48 mg, mixture) as a beige solid.

단계 3: 11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일옥시)아세트아미도)운데카논산의 제조Step 3: Preparation of 11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yloxy)acetamido)undecanoic acid

DCM (1 ml) 중 상기 단계 2에서 제조한 화합물 (48 mg, 0.084 mmol)의 용액에, TFA (1 ml)를 첨가하고, 생성된 혼합물을 실온에서 1.5 시간 동안 교반하였다. 반응 혼합물을 진공 농축하여 오일을 수득하였다. 조 혼합물을 DCM / MeOH = 7 %를 사용하여 실리카겔 컬럼 크로마토그래피로 정제하여 단계 3의 목적 화합물 (29mg, 0.056 mmol, 단계 2-3 동안 29 %)을 백색 고체로서 수득하였다.To a solution of the compound (48 mg, 0.084 mmol) prepared in step 2 above in DCM (1 ml), TFA (1 ml) was added and the resulting mixture was stirred at room temperature for 1.5 hours. The reaction mixture was concentrated in vacuo to an oil. The crude mixture was purified by silica gel column chromatography using DCM/MeOH = 7% to give the desired compound of step 3 (29mg, 0.056mmol, 29% during steps 2-3) as a white solid.

단계 4: (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일옥시)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 4: (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-yloxy)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolid Preparation of Dean-2-carboxamide

DMF 중 상기 단계 3에서 제조한 화합물 (30mg, 0.058mmol)의 용액에, VHL 리간드 (27mg, 0.058mmol), HATU (33mg, 0.087mmol), DIPEA (0.040mL, 0.232mmol)를 첨가하고 실온에서 밤새 교반하였다. 반응 혼합물을 물로 희석하고 EtOAc로 추출하였다. 합쳐진 유기층을 염수로 세척하고 MgSO4상에서 건조시키고 용매를 진공하에 제거하여 오일을 수득하였다. 조 혼합물을 MeOH / DCM = 5 %를 사용하여 PREP TLC상에서 정제하여 최종 목적 화합물 (13mg, 0.014mmol, 24 %)을 백색 고체로서 수득하였다.To the solution of the compound (30mg, 0.058mmol) prepared in step 3 above in DMF, VHL ligand (27mg, 0.058mmol), HATU (33mg, 0.087mmol), DIPEA (0.040mL, 0.232mmol) were added overnight at room temperature. It was stirred. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO 4 and the solvent was removed in vacuo to give an oil. The crude mixture was purified on PREP TLC using MeOH / DCM = 5% to give the final target compound (13mg, 0.014mmol, 24%) as a white solid.

1H NMR (500 MHz, Chloroform-d) δ8.71-8.66 (m, 1H), 7.73 (q, J = 7.4 Hz, 1H), 7.63-7.60 (m, 0.5 H), 7.55-7.50 (m, 1.5H), 7.45 (t, J = 5.2 Hz, 1H), 7.38-7.30 (m, 4H), 7.23-7.17 (m, 0.5H), 4.97-4.90 (m, 0.5H), 4.87-4.80 (m, 1H), 4.70-4.51 (m, 6H), 4.32-4.18 (m, 1H), 4.12 (d, J = 11.6 Hz, 1H), 3.67-3.58 (m, 1H), 3.54-3.43 (m, 1H), 3.35-3.24 (m, 1H), 2.93-2.63 (m, 3H), 2.56-2.45 (m, 4H), 2.24-2.05 (m, 4H), 1.63-1.56 (m, 2H), 1.56-1.47 (m, 2H), 1.46-1.13 (m, 14H), 0.93 (s, 9H). , LC/MS (ESI) m/z 928.1 [M + H]+, 926.1 [M - H]- 1 H NMR (500 MHz, Chloroform- d ) δ8.71-8.66 (m, 1H), 7.73 (q, J = 7.4 Hz, 1H), 7.63-7.60 (m, 0.5 H), 7.55-7.50 (m, 1.5H), 7.45 (t, J = 5.2 Hz, 1H), 7.38-7.30 (m, 4H), 7.23-7.17 (m, 0.5H), 4.97-4.90 (m, 0.5H), 4.87-4.80 (m , 1H), 4.70-4.51 (m, 6H), 4.32-4.18 (m, 1H), 4.12 (d, J = 11.6 Hz, 1H), 3.67-3.58 (m, 1H), 3.54-3.43 (m, 1H) ), 3.35-3.24 (m, 1H), 2.93-2.63 (m, 3H), 2.56-2.45 (m, 4H), 2.24-2.05 (m, 4H), 1.63-1.56 (m, 2H), 1.56-1.47 (m, 2H), 1.46-1.13 (m, 14H), 0.93 (s, 9H). , LC/MS (ESI) m/z 928.1 [M + H] + , 926.1 [M-H] -

<실시예 22> (2S,4R)-1-((S)-2-(11-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-5-일아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 22> (2S,4R)-1-((S)-2-(11-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole- 5-ylamino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-car Preparation of Voxamide

Figure 112018098559920-pat00067
Figure 112018098559920-pat00067

단계 1: tert-부틸 11-아미노운데카노에이트의 제조Step 1: Preparation of tert-butyl 11-aminoundecanoate

티오닐 클로라이드 (24.80 ml, 24.8 mmol) 중 11-아미노운데칸산 (500 mg, 2.48 mmol)의 용액을 실온에서 1.5 시간 동안 교반하고, 그 후 과량의 티오닐 클로라이드를 진공하에 제거하였다. 이 잔류물에 tBuOH (6 ml) 중 NaHCO3 (458 mg, 5.45 mmol)의 용액을 교반하에 첨가하고, 반응 혼합물을 밤새 실온에서 교반하였다. 휘발성 물질을 진공하에 제거하고, 잔류물을 EtOAc (30 ml) 및 NaOH (1M, 수성) (20 ml)에 취하였다. 합한 유기층을 물 (30 ml) 및 염수 (20 ml ×3)로 세척하고, MgSO4상에서 건조시키고, 용매를 진공하에 제거하여 단계 1의 목적 화합물 (521 mg, 2.02 mmol, 81 %)을 황색 투명 오일로서 수득하였다.A solution of 11-aminoundecanoic acid (500 mg, 2.48 mmol) in thionyl chloride (24.80 ml, 24.8 mmol) was stirred at room temperature for 1.5 hours, after which excess thionyl chloride was removed under vacuum. To this residue was added a solution of NaHCO 3 (458 mg, 5.45 mmol) in tBuOH (6 ml) under stirring, and the reaction mixture was stirred overnight at room temperature. The volatiles were removed under vacuum, and the residue was taken into EtOAc (30 ml) and NaOH (1M, aqueous) (20 ml). The combined organic layer was washed with water (30 ml) and brine (20 ml × 3), dried over MgSO 4 and the solvent removed in vacuo to give the desired compound of step 1 (521 mg, 2.02 mmol, 81%) as a yellow transparent. Obtained as an oil.

단계 2: tert-부틸 11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-5-일아미노)운데카노에이트의 제조Step 2: Preparation of tert-butyl 11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-ylamino)undecanoate

DMSO 중 상기 단계 1에서 제조한 화합물 (86 ㎎, 0.334 mmol)의 용액에, 5-F-탈리도마이드 (61 mg, 0.223 mmol), DIPEA (0.058 mL, 0.334 mmol)를 첨가하고, 90 ℃에서 밤새 교반하였다. 반응 혼합물을 물로 희석하고 EtOAc로 추출하였다. 합쳐진 유기층을 염수로 세척하고 MgSO4상에서 건조시키고 용매를 진공하에 제거하여 오일을 수득하였다. 조질의 혼합물을 EtOAc / Hx = 37 %를 사용하여 실리카겔 컬럼 크로마토그래피로 정제하여 단계 2의 목적 화합물 (38 mg, 혼합물)을 황색 고체로서 수득하였다.To a solution of the compound (86 mg, 0.334 mmol) prepared in step 1 above in DMSO, 5-F-thalidomide (61 mg, 0.223 mmol) and DIPEA (0.058 mL, 0.334 mmol) were added and stirred at 90°C overnight. Did. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO 4 and the solvent was removed in vacuo to give an oil. The crude mixture was purified by silica gel column chromatography using EtOAc/Hx=37% to give the desired compound of step 2 (38 mg, mixture) as a yellow solid.

단계 3: 11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-5-일아미노)운데카논산의 제조Step 3: Preparation of 11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-ylamino)undecanoic acid

DCM (1 ml) 중 상기 단계 2에서 제조한 화합물 (38 mg, 0.074 mmol)의 용액에, TFA (1 ml)를 첨가하고, 생성된 혼합물을 실온에서 3 시간 동안 교반하였다. 반응 혼합물을 진공 농축하여 오일을 수득하였다. 조 혼합물을 DCM / MeOH = 7 %를 사용하여 실리카겔 컬럼 크로마토그래피로 정제하여 단계 3의 목적 화합물 (12mg, 0.026 mmol, 단계 2-3 동안 11 %)을 황색 고체로서 수득하였다.To a solution of the compound (38 mg, 0.074 mmol) prepared in step 2 above in DCM (1 ml), TFA (1 ml) was added and the resulting mixture was stirred at room temperature for 3 hours. The reaction mixture was concentrated in vacuo to an oil. The crude mixture was purified by silica gel column chromatography using DCM/MeOH=7% to give the desired compound of step 3 (12mg, 0.026mmol, 11% during steps 2-3) as a yellow solid.

단계 4: (2S,4R)-1-((S)-2-(11-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-5-일아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조Step 4: (2S,4R)-1-((S)-2-(11-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5- Ilamino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide Manufacture of

DMF 중 상기 단계 3에서 제조한 화합물 (12mg, 0.026mmol)의 용액에, VHL- 리간드 (12mg, 0.026mmol), HATU (15mg, 0.039mmol), DIPEA (0.018mL, 0.104mmol)를 첨가하고, 실온에서 밤새 교반하였다. 반응 혼합물을 물로 희석하고 EtOAc로 추출하였다. 합쳐진 유기층을 염수로 세척하고 MgSO4상에서 건조시키고 용매를 진공하에 제거하여 오일을 수득하였다. 조 혼합물을 MeOH / DCM = 5 %를 사용하여 PREP TLC상에서 정제하여 최종 목적 화합물 (12mg, 0.014mmol, 53 %)을 황색 고체로서 수득하였다.To a solution of the compound (12mg, 0.026mmol) prepared in step 3 above in DMF, VHL-ligand (12mg, 0.026mmol), HATU (15mg, 0.039mmol), DIPEA (0.018mL, 0.104mmol) was added, and at room temperature Stir overnight. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO 4 and the solvent was removed in vacuo to give an oil. The crude mixture was purified on PREP TLC using MeOH / DCM = 5% to give the final target compound (12mg, 0.014mmol, 53%) as a yellow solid.

1H NMR (500 MHz, Chloroform-d) δ8.68 (s, 1H), 7.66-7.61 (m, 0.5H), 7.59 (d, J = 8.2 Hz, 1H), 7.52 (t, J = 6.3 Hz, 0.5H), 7.39-7.32 (m, 4H), 6.98-6.93 (m, 1H), 6.79-6.77 (m, 0.5H), 6.73 (d, J = 8.4 Hz, 1H), 6.56-6.54 (m, 0.5H), 4.96-4.87 (m, 1H), 4.72 (t, J = 5.0 Hz, 1H), 4.70-4.65 (m, 1H), 4.65-4.57 (m, 1H), 4.57-4.50 (m, 2H), 4.12-4.04 (m, 1H), 3.68-3.60 (m, 1H), 2.91-2.61 (m, 3H), 2.56-2.46 (m, 4H), 2.21-2.05 (m, 4H), 1.62 (p, J = 7.2 Hz, 2H), 1.58-1.48 (m, 2H), 1.43-1.29 (m, 5H), 1.30-1.11 (m, 11H), 0.91 (s, 9H). 1 H NMR (500 MHz, Chloroform- d ) δ8.68 (s, 1H), 7.66-7.61 (m, 0.5H), 7.59 (d, J = 8.2 Hz, 1H), 7.52 (t, J = 6.3 Hz , 0.5H), 7.39-7.32 (m, 4H), 6.98-6.93 (m, 1H), 6.79-6.77 (m, 0.5H), 6.73 (d, J = 8.4 Hz, 1H), 6.56-6.54 (m , 0.5H), 4.96-4.87 (m, 1H), 4.72 (t, J = 5.0 Hz, 1H), 4.70-4.65 (m, 1H), 4.65-4.57 (m, 1H), 4.57-4.50 (m, 2H), 4.12-4.04 (m, 1H), 3.68-3.60 (m, 1H), 2.91-2.61 (m, 3H), 2.56-2.46 (m, 4H), 2.21-2.05 (m, 4H), 1.62 ( p, J = 7.2 Hz, 2H), 1.58-1.48 (m, 2H), 1.43-1.29 (m, 5H), 1.30-1.11 (m, 11H), 0.91 (s, 9H).

<실시예 23> 3,3'-(5,5'-(노난-1,9-디일비스(아잔디일))비스(4-옥소벤조[d][1,2,3]트리아진-5,3(4H)-디일))디피페리딘-2,6-디온의 제조<Example 23> 3,3'-(5,5'-(nonane-1,9-diylbis(azandiyl))bis(4-oxobenzo[d][1,2,3]triazine- Preparation of 5,3(4H)-diyl))dipiperidine-2,6-dione

Figure 112018098559920-pat00068
Figure 112018098559920-pat00068

상기 실시예 19의 단계 1과 단계 4에서 사용한, 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온을 대신하여, 출원번호 10-2017-0184761호에 공지된 바와 같이 제조되는 3-(5-아미노-4-옥소벤조[d][1,2,3]트리아진-3(4H)-일)피페리딘-2,6-디온을 사용한 점을 제외하고, 상기 실시예 19와 같이 수행하여 목적 화합물을 제조하였다.In place of 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-1,3-dione, used in steps 1 and 4 of Example 19 above, application number 10- 3-(5-amino-4-oxobenzo[d][1,2,3]triazine-3(4H)-yl)piperidine-2,6-prepared as known from 2017-0184761 Except that dione was used, the same procedure as in Example 19 was carried out to prepare a target compound.

1H NMR (300 MHz, CDCl3)δ8.74(s,1H),8.68(s,1H),8.15(t,J = 5.2 Hz, 2H), 7.67 (t, J = 8.2 Hz, 2H), 7.24 (d, J = 7.7 Hz, 2H), 6.77 (d, J = 8.5 Hz, 2H), 5.74-.64 (m, 2H), 3.25-.13 (m, 4H), 3.00-.84 (m, 6H), 2.41-.28 (m, 2H), 1.76-.62 (m, 6H), 1.49-.29 (m, 11H), 0.95-.79 (m, 1H). 1 H NMR (300 MHz, CDCl 3 )δ8.74(s,1H),8.68(s,1H),8.15(t,J = 5.2 Hz, 2H), 7.67 (t, J = 8.2 Hz, 2H), 7.24 (d, J = 7.7 Hz, 2H), 6.77 (d, J = 8.5 Hz, 2H), 5.74-.64 (m, 2H), 3.25-.13 (m, 4H), 3.00-.84 (m , 6H), 2.41-.28 (m, 2H), 1.76-.62 (m, 6H), 1.49-.29 (m, 11H), 0.95-.79 (m, 1H).

<실시예 24> 2-(2,6-디옥소피페리딘-3-일)-4-(9-(3-(2,6-디옥소피페리딘-3-일)-4-옥소-3,4-디히드로벤조[d][1,2,3]트리아진-5-일아미노)노닐아미노)이소인돌린-1,3-디온의 제조<Example 24> 2-(2,6-dioxopiperidin-3-yl)-4-(9-(3-(2,6-dioxopiperidin-3-yl)-4-oxo-3 Preparation of ,4-dihydrobenzo[d][1,2,3]triazin-5-ylamino)nonylamino)isoindoline-1,3-dione

Figure 112018098559920-pat00069
Figure 112018098559920-pat00069

상기 실시예 19의 단계 1에서 사용한, 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온을 대신하여, 출원번호 10-2017-0184761호에 공지된 바와 같이 제조되는 3-(5-아미노-4-옥소벤조[d][1,2,3]트리아진-3(4H)-일)피페리딘-2,6-디온을 사용한 점을 제외하고, 상기 실시예 19와 같이 수행하여 목적 화합물을 제조하였다.In place of 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-1,3-dione, used in step 1 of Example 19 above, application number 10-2017-0184761 3-(5-amino-4-oxobenzo[d][1,2,3]triazine-3(4H)-yl)piperidine-2,6-dione, prepared as known in the heading, Except for the point, it was carried out as in Example 19 above to prepare the target compound.

1H NMR (300 MHz, CDCl3)δ8.452(d,J = 6.1 Hz, 2H), 8.15 (s, 1H), 7.68 (t, J = 8.1 Hz, 1H), 7.48 (dd, J = 8.5, 7.1 Hz, 1H), 7.24 (dd, J = 7.9, 0.8 Hz, 2H), 7.08 (d, J = 7.0 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.78 (d, J = 8.5 Hz, 1H), 6.24 (t, J = 5.6 Hz, 1H), 5.73-5.62 (m, 1H), 4.96-4.86 (m, 1H), 3.30-3.15 (m, 4H), 3.01-2.69 (m, 6H), 2.39-2.29 (m, 1H), 2.17-2.08 (m, 1H), 1.75-1.61 (m, 4H), 1.49-1.30 (m, 10H). 1 H NMR (300 MHz, CDCl 3 )δ8.452 (d,J = 6.1 Hz, 2H), 8.15 (s, 1H), 7.68 (t, J = 8.1 Hz, 1H), 7.48 (dd, J = 8.5 , 7.1 Hz, 1H), 7.24 (dd, J = 7.9, 0.8 Hz, 2H), 7.08 (d, J = 7.0 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.78 (d, J = 8.5 Hz, 1H), 6.24 (t, J = 5.6 Hz, 1H), 5.73-5.62 (m, 1H), 4.96-4.86 (m, 1H), 3.30-3.15 (m, 4H), 3.01-2.69 ( m, 6H), 2.39-2.29 (m, 1H), 2.17-2.08 (m, 1H), 1.75-1.61 (m, 4H), 1.49-1.30 (m, 10H).

<실시예 25> (2S,4R)-1-((S)-2-(11-(3-(2,6-디옥소피페리딘-3-일)-4-옥소-3,4-디히드로벤조[d][1,2,3]트리아진-5-일아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드의 제조<Example 25> (2S,4R)-1-((S)-2-(11-(3-(2,6-dioxopiperidin-3-yl)-4-oxo-3,4-di Hydrobenzo[d][1,2,3]triazine-5-ylamino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthia Preparation of zol-5-yl)benzyl)pyrrolidine-2-carboxamide

Figure 112018098559920-pat00070
Figure 112018098559920-pat00070

상기 실시예 1의 단계 1에서 사용한 7-아미노헵탄산을 대신하여, 11-아미노운데칸산을 사용사고, 2-(2,6-디옥소피페리딘-3-일)-4-플루오로이소인돌린-1,3-디온을 대신하여, 출원번호 10-2017-0184761호에 공지된 바와 같이 제조되는 3-(5-아미노-4-옥소벤조[d][1,2,3]트리아진-3(4H)-일)피페리딘-2,6-디온을 사용한 점을 제외하고, 상기 실시예 1과 같이 수행하여 목적 화합물을 제조하였다.Instead of the 7-aminoheptanoic acid used in Step 1 of Example 1, 11-aminoundecanoic acid was used, and 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindole In place of -1,3-dione, 3-(5-amino-4-oxobenzo[d][1,2,3]triazine-3, prepared as known from application number 10-2017-0184761 (4H)-yl) was carried out as in Example 1, except that the piperidine-2,6-dione was used to prepare the target compound.

1H NMR (300 MHz, CDCl3)δ10.17(s,0.5H),9.74(s,0.5H),8.68(s,1H),8.25-8.13(m,1H),7.67(t,J = 8.1 Hz, 1H), 7.58-7.50 (m, 0.5H), 7.46 (t, J = 5.5 Hz, 0.5H), 7.40-7.30 (m, 4H), 7.23 (d, J = 7.7 Hz, 1H), 6.80-6.70 (m, 1.5H), 6.50 (d, J = 8.7 Hz, 1H), 5.72-5.62 (m, 0.5H), 5.56-5.46 (m, 0.5H), 4.72-4.50 (m, 4H), 4.36-4.20 (m, 1H), 4.18-4.06 (m, 1H), 3.68-3.58 (m, 1H), 3.29 (s, 0.5H), 3.26-3.14 (m, 2.5H), 2.99-2.67 (m, 3H), 2.58-2.45 (m, 4H), 2.37-2.23 (m, 1H), 2.23-2.07 (m, 3H), 1.61-1.49 (m, 2H), 1.49-1.38 (m, 2H), 1.38-1.16 (m, 12H), 0.98-0.87 (m, 9H). 1 H NMR (300 MHz, CDCl 3 )δ10.17(s,0.5H),9.74(s,0.5H),8.68(s,1H),8.25-8.13(m,1H),7.67(t,J = 8.1 Hz, 1H), 7.58-7.50 (m, 0.5H), 7.46 (t, J = 5.5 Hz, 0.5H), 7.40-7.30 (m, 4H), 7.23 (d, J = 7.7 Hz, 1H), 6.80-6.70 (m, 1.5H), 6.50 (d, J = 8.7 Hz, 1H), 5.72-5.62 (m, 0.5H), 5.56-5.46 (m, 0.5H), 4.72-4.50 (m, 4H) , 4.36-4.20 (m, 1H), 4.18-4.06 (m, 1H), 3.68-3.58 (m, 1H), 3.29 (s, 0.5H), 3.26-3.14 (m, 2.5H), 2.99-2.67 ( m, 3H), 2.58-2.45 (m, 4H), 2.37-2.23 (m, 1H), 2.23-2.07 (m, 3H), 1.61-1.49 (m, 2H), 1.49-1.38 (m, 2H), 1.38-1.16 (m, 12H), 0.98-0.87 (m, 9H).

상기 실시예 1 - 25에서 제조한 화합물의 화학 구조식을 하기 표 1에 나타내었다.The chemical structural formulas of the compounds prepared in Examples 1-25 are shown in Table 1 below.

실시예Example 구조식constitutional formula 실시예Example 구조식constitutional formula 1One

Figure 112018098559920-pat00071
Figure 112018098559920-pat00071
1414
Figure 112018098559920-pat00072
Figure 112018098559920-pat00072
22
Figure 112018098559920-pat00073
Figure 112018098559920-pat00073
1515
Figure 112018098559920-pat00074
Figure 112018098559920-pat00074
33
Figure 112018098559920-pat00075
Figure 112018098559920-pat00075
1616
Figure 112018098559920-pat00076
Figure 112018098559920-pat00076
44
Figure 112018098559920-pat00077
Figure 112018098559920-pat00077
1717
Figure 112018098559920-pat00078
Figure 112018098559920-pat00078
55
Figure 112018098559920-pat00079
Figure 112018098559920-pat00079
1818
Figure 112018098559920-pat00080
Figure 112018098559920-pat00080
66
Figure 112018098559920-pat00081
Figure 112018098559920-pat00081
1919
Figure 112018098559920-pat00082
Figure 112018098559920-pat00082
77
Figure 112018098559920-pat00083
Figure 112018098559920-pat00083
2020
Figure 112018098559920-pat00084
Figure 112018098559920-pat00084
88
Figure 112018098559920-pat00085
Figure 112018098559920-pat00085
2121
Figure 112018098559920-pat00086
Figure 112018098559920-pat00086
99
Figure 112018098559920-pat00087
Figure 112018098559920-pat00087
2222
Figure 112018098559920-pat00088
Figure 112018098559920-pat00088
1010
Figure 112018098559920-pat00089
Figure 112018098559920-pat00089
2323
Figure 112018098559920-pat00090
Figure 112018098559920-pat00090
1111
Figure 112018098559920-pat00091
Figure 112018098559920-pat00091
2424
Figure 112018098559920-pat00092
Figure 112018098559920-pat00092
1212
Figure 112018098559920-pat00093
Figure 112018098559920-pat00093
2525
Figure 112018098559920-pat00094
Figure 112018098559920-pat00094
1313
Figure 112018098559920-pat00095
Figure 112018098559920-pat00095

<실험예 1> 세레브론(CRBN) 단백질 분해활성 평가<Experimental Example 1> Evaluation of cerebron (CRBN) proteolytic activity

본 발명에 따른 화합물의 세레브론(CRBN) 단백질 분해활성을 평가하기 위해, 다음과 같이 실험하였다.In order to evaluate the cerebron (CRBN) proteolytic activity of the compound according to the present invention, experiments were conducted as follows.

구체적으로, HEK293T 세포를 12 웰 플레이트의 각 웰에 5 X 105개를 주입하였다. 다음날 각 웰에 실시예 화합물 1-22를 최종농도 10nM, 30 nM, 100 nM, 300 nM, 1 μM, 3 μM, 10 μM이 되도록 처리해 주었다. 한 개의 웰에는 DMSO를 동일한 백분율로 처리하였다. 처리 24시간 후, 세포를 모아 NP40 Lysis buffer(20mM Tris, pH7.5, 150mM NaCl, 1% NP-40, 1mM EDTA, and protease inhibitor cocktail)를 이용하여 세포 용해물(cell lysate)을 만들어 웨스턴블롯을 수행하였고, 그 결과를 하기 표 2에 나타내었다.Specifically, 5 x 10 5 HEK293T cells were injected into each well of a 12-well plate. The next day, each well was treated with Example Compounds 1-22 to a final concentration of 10 nM, 30 nM, 100 nM, 300 nM, 1 μM, 3 μM, 10 μM. One well was treated with the same percentage of DMSO. After 24 hours of treatment, cells were collected and Western blotting was performed using NP40 Lysis buffer (20mM Tris, pH7.5, 150mM NaCl, 1% NP-40, 1mM EDTA, and protease inhibitor cocktail) to make cell lysates. And the results are shown in Table 2 below.

또한, 실시예 1, 3, 8에 대한 세레브론 단백질 분석 결과를 도 1에 나타내었고, 실시예 1, 3, 8 화합물 처리 농도에 따른 세레브론 단백질 분해력을 평가(DC50)하여 도 2에 도시하였다.In addition, the results of analysis of cerebron protein for Examples 1, 3, and 8 are shown in FIG. 1, and the results of evaluating the decomposition ability of cerebron protein according to the concentrations of compounds treated in Examples 1, 3, and 8 (DC 50 ) are shown in FIG. 2. Did.

실시예Example 세레브론(CRBN) 분해활성Cerebron (CRBN) decomposition activity 실시예Example 세레브론(CRBN) 분해활성Cerebron (CRBN) decomposition activity 1One ++++ 1414 ++++ 22 ++++ 1515 ++ 33 ++++ 1616 ++++ 44 ++++ 1717 ++ 55 ++ 1818 ++ 66 ++ 1919 ++++ 77 ++ 2020 ++++ 88 ++ 2121 ++ 99 ++ 2222 ++++ 1010 ++ 2323 ++++ 1111 ++ 2424 ++++ 1212 ++++ 2525 ++++ 1313 ++++

++ : 300 nM 농도에서 50% 초과로 분해(> 50%)+ : 300 nM 농도에서 50% 미만으로 분해(< 50%)++: decomposition to more than 50% at 300 nM concentration (> 50%) +: decomposition to less than 50% at 300 nM concentration (< 50%)

표 2를 살펴보면, 본 발명에 따른 실시예 화합물은 세레브론(CRBN) 단백질을 300 nM의 농도에서 우수하게 분해할 수 있으며, 특히 실시예 1, 2, 3, 4, 12, 13, 14, 16, 19, 20, 22, 23, 24, 및 25 화합물은 300 nM의 농도에서 50 % 초과의 분해활성을 나타내는 것을 확인할 수 있다.Looking at Table 2, the example compound according to the present invention can excellently decompose Celebron (CRBN) protein at a concentration of 300 nM, in particular Examples 1, 2, 3, 4, 12, 13, 14, 16 , 19, 20, 22, 23, 24, and 25 compounds can be seen that exhibit a decomposition activity of more than 50% at a concentration of 300 nM.

따라서, 본 발명에 따른 화합물은 세레브론(CRBN) 단백질을 나노몰 단위의 농도로 우수하게 분해할 수 있으며, 이로부터 세레브론(CRBN) 관련 질환의 예방 및 치료에 효과적으로 사용될 수 있다.Therefore, the compound according to the present invention can excellently decompose celebrity (CRBN) protein to a concentration of nanomolar units, from which it can be effectively used for the prevention and treatment of cerebron (CRBN) related diseases.

Claims (10)

A-Linker-A 또는 A-Linker-B로 표시되는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염:
상기 A는 세레브론(cereblon) E3 유비퀴틴 리가아제 결합 모이어티(moiety)이고; 및
상기 B는 E3 유비퀴틴 리가아제 결합 모이어티(moiety)이되,
상기 세레브론(cereblon) E3 유비퀴틴 리가아제 결합 모이어티는
Figure 112020029592003-pat00134
,
Figure 112020029592003-pat00135
,
Figure 112020029592003-pat00136
, 또는
Figure 112020029592003-pat00137
이고,
여기서 상기 R1은 -(CH2)n-NR3-, -(CH2)n-O-, -O-(CH2)n-(C=O)-NR3- 또는 -NR3-(CH2)n-(C=O)-NR3-이고,
다시 여기서, n은 0 내지 2의 정수이고,
상기 R2 및 R3는 각각 H이고;

상기 E3 유비퀴틴 리가아제 결합 모이어티는
Figure 112020029592003-pat00138
이되,
여기서 상기 R2 및 R3는 각각 H이고; 및

상기 Linker는,
Figure 112020029592003-pat00139
이되,
여기서, 상기 a 및 i는 독립적으로 0 또는 1이고;
상기 b는 0-3의 정수이고;
상기 c는 0 또는 1이고;
상기 d는 0-3의 정수이고;
상기 e는 0 또는 1이고;
상기 f는 1-5의 정수이고;
상기 g는 0 또는 1이고;
상기 h는 0-3의 정수이고;
상기 j 및 k는 독립적으로 0-5의 정수인 것을 특징으로 하는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염.
A compound represented by A-Linker-A or A-Linker-B, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
A is a cereblon E3 ubiquitin ligase binding moiety; And
The B is an E3 ubiquitin ligase binding moiety,
The cereblon E3 ubiquitin ligase binding moiety is
Figure 112020029592003-pat00134
,
Figure 112020029592003-pat00135
,
Figure 112020029592003-pat00136
, or
Figure 112020029592003-pat00137
ego,
Wherein R 1 is -(CH 2 ) n -NR 3 -, -(CH 2 ) n -O-, -O-(CH 2 ) n -(C=O)-NR 3 -or -NR 3 -( CH 2 ) n -(C=O)-NR 3 -,
Here again, n is an integer from 0 to 2,
R 2 and R 3 are each H;

The E3 ubiquitin ligase binding moiety
Figure 112020029592003-pat00138
This,
Wherein R 2 and R 3 are each H; And

The Linker,
Figure 112020029592003-pat00139
This,
Where a and i are independently 0 or 1;
B is an integer of 0-3;
C is 0 or 1;
D is an integer of 0-3;
E is 0 or 1;
F is an integer of 1-5;
G is 0 or 1;
H is an integer of 0-3;
J and k are independently an integer of 0-5, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
삭제delete 제1항에 있어서,
상기 Linker는,
Figure 112020029592003-pat00106
,
Figure 112020029592003-pat00107
,
Figure 112020029592003-pat00108
,
Figure 112020029592003-pat00109
,
Figure 112020029592003-pat00110
,
Figure 112020029592003-pat00111
,
Figure 112020029592003-pat00112
,
Figure 112020029592003-pat00113
,
Figure 112020029592003-pat00114
,
Figure 112020029592003-pat00115
,
Figure 112020029592003-pat00116
,
Figure 112020029592003-pat00117
,
Figure 112020029592003-pat00118
,
Figure 112020029592003-pat00119
,
Figure 112020029592003-pat00120
,
Figure 112020029592003-pat00121
,
Figure 112020029592003-pat00122
,
Figure 112020029592003-pat00123
, 또는
Figure 112020029592003-pat00125
인 것을 특징으로 하는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염.
According to claim 1,
The Linker,
Figure 112020029592003-pat00106
,
Figure 112020029592003-pat00107
,
Figure 112020029592003-pat00108
,
Figure 112020029592003-pat00109
,
Figure 112020029592003-pat00110
,
Figure 112020029592003-pat00111
,
Figure 112020029592003-pat00112
,
Figure 112020029592003-pat00113
,
Figure 112020029592003-pat00114
,
Figure 112020029592003-pat00115
,
Figure 112020029592003-pat00116
,
Figure 112020029592003-pat00117
,
Figure 112020029592003-pat00118
,
Figure 112020029592003-pat00119
,
Figure 112020029592003-pat00120
,
Figure 112020029592003-pat00121
,
Figure 112020029592003-pat00122
,
Figure 112020029592003-pat00123
, or
Figure 112020029592003-pat00125
A compound, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
삭제delete 제1항에 있어서,
상기 A-Linker-A 또는 A-Linker-B로 표시되는 화합물은 하기 화합물로 이루어진 군으로부터 선택되는 화합물, 이의 입체 이성질체, 또는 이의 약학적으로 허용 가능한 염:
(1) (2S,4R)-1-((S)-2-(7-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헵탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(2) (2S,4R)-1-((S)-2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥산아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(3) (2S,4R)-1-((S)-2-(9-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)노난아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(4) (2S,4R)-1-((S)-2-tert-부틸-17-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-4-옥소-6,9,12,15-테트라옥사-3-아자헵타데카-1-노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(5) (2S,4R)-1-((S)-2-(2-(3-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-2-옥소에톡시)프로폭시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(6) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N9-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)노난디아미드;
(7) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N8-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)옥탄디아미드;
(8) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N7-((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)헵탄디아미드;
(9) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N14-((S)-1-((2S,4R)-4-히드록시-2-((4-(4-메틸티아졸-5-일)벤질)카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)-3,6,9,12-테트라옥사테트라데칸-1,14-디아미드;
(10) N1-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)-N5-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)글루타르아미드;
(11) N1-((2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일)메틸)-N7-((S)-1-((2S,4R)-4-히드록시-2-(4-(4-메틸티아졸-5-일)벤질카바모일)피롤리딘-1-일)-3,3-디메틸-1-옥소부탄-2-일)헵탄디아미드;
(12) (2S,4R)-1-((S)-2-(11-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)운데칸아미도)-3.3-디메틸부탄오일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(13) (2S,4R)-1-((S)-2-(8-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)옥탄아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(14) (2S,4R)-1-((S)-2-(12-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)도데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(15) (2S,4R)-1-((S)-2-(2-(6-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)헥실옥시)아세트아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(16) (2S,4R)-1-((S)-2-(10-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(17) (2S,4R)-1-((S)-2-tert-부틸-14-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)-4-옥소-6,9,12-트리옥사-3-아자테트라데카-1-노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(18) (2S,4R)-1-((S)-2-(11-((3-(2,6- 디옥소피페리딘-3-일)-2-메틸-4-옥소-3,4-디히드로퀴나졸린-5-일)아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카르복스아미드;
(19) 4,4'-(노난-1,9-디일비스(아자네디일))비스(2-(2,6-디옥소피페리딘-3-일)이소인돌린-1,3-디온);
(20) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일아미노)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(21) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-4-일옥시)아세트아미도)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(22) (2S,4R)-1-((S)-2-(11-(2-(2,6-디옥소피페리딘-3-일)-1,3-디옥소이소인돌린-5-일아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드;
(23) 3,3'-(5,5'-(노난-1,9-디일비스(아잔디일))비스(4-옥소벤조[d][1,2,3]트리아진-5,3(4H)-디일))디피페리딘-2,6-디온;
(24) 2-(2,6-디옥소피페리딘-3-일)-4-(9-(3-(2,6-디옥소피페리딘-3-일)-4-옥소-3,4-디히드로벤조[d][1,2,3]트리아진-5-일아미노)노닐아미노)이소인돌린-1,3-디온; 및
(25) (2S,4R)-1-((S)-2-(11-(3-(2,6-디옥소피페리딘-3-일)-4-옥소-3,4-디히드로벤조[d][1,2,3]트리아진-5-일아미노)운데칸아미도)-3,3-디메틸부타노일)-4-히드록시-N-(4-(4-메틸티아졸-5-일)벤질)피롤리딘-2-카복스아미드.
According to claim 1,
The compound represented by A-Linker-A or A-Linker-B is a compound selected from the group consisting of the following compounds, stereoisomers thereof, or pharmaceutically acceptable salts thereof:
(1) (2S,4R)-1-((S)-2-(7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)heptanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(2) (2S,4R)-1-((S)-2-(6-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Ylamino)hexaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(3) (2S,4R)-1-((S)-2-(9-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)nonanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(4) (2S,4R)-1-((S)-2-tert-butyl-17-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)-4-oxo-6,9,12,15-tetraoxa-3-azaheptadeca-1-noyl)-4-hydroxy-N-(4-(4-methylthiazole- 5-yl)benzyl)pyrrolidine-2-carboxamide;
(5) (2S,4R)-1-((S)-2-(2-(3-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-di Oxoisoindoline-4-ylamino)-2-oxoethoxy)propoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazole- 5-yl)benzyl)pyrrolidine-2-carboxamide;
(6) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 9 -((S)-1-(( 2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutane- 2-yl)nonanediamide;
(7) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindoline-4-yl)-N 8 -((S)-1-(( 2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutane- 2-yl)octanediamide;
(8) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 7 -((S)-1-(( 2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxo Butan-2-yl)heptanediamide;
(9) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 14 -((S)-1-(( 2S,4R)-4-hydroxy-2-((4-(4-methylthiazol-5-yl)benzyl)carbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxo Butan-2-yl)-3,6,9,12-tetraoxatetradecane-1,14-diamide;
(10) N 1 -(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-N 5 -((S)-1-(( 2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1-oxobutane- 2-yl)glutaramide;
(11) N 1 -((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)methyl)-N 7 -((S)-1 -((2S,4R)-4-hydroxy-2-(4-(4-methylthiazol-5-yl)benzylcarbamoyl)pyrrolidin-1-yl)-3,3-dimethyl-1- Oxobutan-2-yl)heptanediamide;
(12) (2S,4R)-1-((S)-2-(11-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)undecanamido)-3.3-dimethylbutanyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(13) (2S,4R)-1-((S)-2-(8-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Ylamino)octaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(14) (2S,4R)-1-((S)-2-(12-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- Ilamino)dodecaneamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide ;
(15) (2S,4R)-1-((S)-2-(2-(6-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)hexyloxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine -2-carboxamide;
(16) (2S,4R)-1-((S)-2-(10-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4- 1amino)decanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide;
(17) (2S,4R)-1-((S)-2-tert-butyl-14-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)-4-oxo-6,9,12-trioxa-3-azatetradeca-1-noyl)-4-hydroxy-N-(4-(4-methylthiazole-5- 1)benzyl)pyrrolidine-2-carboxamide;
(18) (2S,4R)-1-((S)-2-(11-((3-(2,6-dioxopiperidin-3-yl)-2-methyl-4-oxo-3, 4-dihydroquinazolin-5-yl)amino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl )Pyrrolidine-2-carboxamide;
(19) 4,4'-(nonane-1,9-diylbis(azanediyl))bis(2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione );
(20) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-ylamino)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolid Dean-2-carboxamide;
(21) (2S,4R)-1-((S)-2-(11-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindole -4-yloxy)acetamido)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolid Dean-2-carboxamide;
(22) (2S,4R)-1-((S)-2-(11-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5- Ilamino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide ;
(23) 3,3'-(5,5'-(nonane-1,9-diylbis(azanediyl))bis(4-oxobenzo[d][1,2,3]triazine-5, 3(4H)-diyl))dipiperidine-2,6-dione;
(24) 2-(2,6-dioxopiperidin-3-yl)-4-(9-(3-(2,6-dioxopiperidin-3-yl)-4-oxo-3,4 -Dihydrobenzo[d][1,2,3]triazin-5-ylamino)nonylamino)isoindoline-1,3-dione; And
(25) (2S,4R)-1-((S)-2-(11-(3-(2,6-dioxopiperidin-3-yl)-4-oxo-3,4-dihydrobenzo [d][1,2,3]triazin-5-ylamino)undecanamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazole- 5-yl)benzyl)pyrrolidine-2-carboxamide.
하기 반응식 1에 나타난 바와 같이,
U-Linker-V로 표시되는 화합물로부터 A-Linker-V로 표시되는 화합물을 제조하는 단계(단계 1);
상기 단계 1에서 제조한 A-Linker-V로 표시되는 화합물로부터 A-Linker-A 또는 A-Linker-B로 표시되는 화합물을 제조하는 단계(단계 2);를 포함하는 제1항의 A-Linker-A 또는 A-Linker-B로 표시되는 화합물의 제조방법:
[반응식 1]
Figure 112018098559920-pat00131

(상기 반응식 1에 있어서,
A, Linker 및 B는 제1항에서 정의한 바와 같고; 및
U 또는 V는 각각 독립적으로 NH2 또는 OH이다).
As shown in Scheme 1 below,
Preparing a compound represented by A-Linker-V from the compound represented by U-Linker-V (step 1);
Preparing a compound represented by A-Linker-A or A-Linker-B from the compound represented by A-Linker-V prepared in step 1 (step 2); A-Linker of claim 1 comprising a Method for preparing a compound represented by A or A-Linker-B:
[Scheme 1]
Figure 112018098559920-pat00131

(In Scheme 1,
A, Linker and B are as defined in claim 1; And
U or V are each independently NH 2 or OH).
삭제delete 제1항의 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 암의 예방 또는 치료용 약학적 조성물.
A pharmaceutical composition for preventing or treating cancer comprising the compound of claim 1, a stereoisomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient.
제1항의 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 자가면역질환의 예방 또는 치료용 약학적 조성물.
A pharmaceutical composition for the prevention or treatment of an autoimmune disease comprising the compound of claim 1, a stereoisomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient.
제1항의 화합물, 이의 입체 이성질체 또는 이의 약학적으로 허용 가능한 염을 유효성분으로 함유하는 대사질환의 예방 또는 치료용 약학적 조성물.
A pharmaceutical composition for the prevention or treatment of metabolic diseases comprising the compound of claim 1, a stereoisomer thereof or a pharmaceutically acceptable salt thereof as an active ingredient.
KR1020180119161A 2017-10-20 2018-10-05 Compound for inducing the degradation of cereblon protein, preparation method thereof and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient KR102129367B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20170136771 2017-10-20
KR1020170136771 2017-10-20

Publications (2)

Publication Number Publication Date
KR20190044499A KR20190044499A (en) 2019-04-30
KR102129367B1 true KR102129367B1 (en) 2020-07-03

Family

ID=66174056

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020180119161A KR102129367B1 (en) 2017-10-20 2018-10-05 Compound for inducing the degradation of cereblon protein, preparation method thereof and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient

Country Status (2)

Country Link
KR (1) KR102129367B1 (en)
WO (1) WO2019078522A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3429996A4 (en) 2016-03-16 2020-02-12 H. Lee Moffitt Cancer Center And Research Institute, Inc. Small molecules against cereblon to enhance effector t cell function
AU2019301679A1 (en) * 2018-07-11 2021-01-28 H. Lee Moffitt Cancer Center And Research Institute, Inc. Dimeric immuno-modulatory compounds against cereblon-based mechanisms
JPWO2020027225A1 (en) 2018-07-31 2021-11-11 ファイメクス株式会社 Heterocyclic compound
AU2020404956A1 (en) * 2019-12-17 2022-07-07 Orionis Biosciences, Inc. Compounds modulating protein recruitment and/or degradation
WO2021152113A1 (en) * 2020-01-31 2021-08-05 Bayer Aktiengesellschaft Substituted 2,3-benzodiazepines derivatives
WO2021168314A1 (en) * 2020-02-21 2021-08-26 Plexium, Inc. Quinazolinone compounds and related compounds
WO2021188537A1 (en) * 2020-03-17 2021-09-23 Dana-Farber Cancer Institute, Inc. Selective small molecule degraders of cereblon
BR112023005344A2 (en) * 2020-09-23 2023-05-09 St Jude Childrens Res Hospital Inc SUBSTITUTED N-(2-(2,6-DIOXOPIPERIDIN-3-IL)-1,3-DIOXOISOINDOLIN-5-IL)ARYLSULFONAMIDE ANALOGS AS CEREBLON PROTEIN MODULATORS
CN112094307A (en) * 2020-09-28 2020-12-18 深圳市术理科技有限公司 Compound for target ubiquitination degradation of ER alpha protein and application thereof
US20240158370A1 (en) 2022-09-09 2024-05-16 Innovo Therapeutics, Inc. CK1 alpha AND DUAL CK1 alpha / GSPT1 DEGRADING COMPOUNDS

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016149668A1 (en) 2015-03-18 2016-09-22 Arvinas, Inc. Compounds and methods for the enhanced degradation of targeted proteins

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2702410A2 (en) * 2011-04-29 2014-03-05 Celgene Corporation Methods for the treatment of cancer and inflammatory diseases using cereblon as a predictor
EP3608317A1 (en) 2012-01-12 2020-02-12 Yale University Compounds & methods for the enhanced degradation of targeted proteins & other polypeptides by an e3 ubiquitin ligase
US9694084B2 (en) * 2014-12-23 2017-07-04 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
GB201504314D0 (en) * 2015-03-13 2015-04-29 Univ Dundee Small molecules
EP3609889B1 (en) * 2017-04-14 2023-07-05 University Of Dundee Small molecules

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016149668A1 (en) 2015-03-18 2016-09-22 Arvinas, Inc. Compounds and methods for the enhanced degradation of targeted proteins

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Bioorganic & Medicinal Chemistry Letters, Vol.26(21), pp.5260-5262 (2016)*
Nature Communications, 8:830 (2017.10.10.)*

Also Published As

Publication number Publication date
WO2019078522A1 (en) 2019-04-25
KR20190044499A (en) 2019-04-30

Similar Documents

Publication Publication Date Title
KR102129367B1 (en) Compound for inducing the degradation of cereblon protein, preparation method thereof and pharmaceutical composition for use in preventing or treating cancer containing the same as an active ingredient
ES2968669T3 (en) Selective NLRP3 inflammasome inhibitors
KR102090780B1 (en) Substituted pyrrolidines as factor xia inhibitors for the treatment thromboembolic diseases
AU2009331179B2 (en) Novel bicyclic heterocyclic compound
EP4122925A1 (en) Proteolysis regulator and method for using same
EP3919483A1 (en) Benzopyridone heterocyclic compound and use thereof
JP2019521082A (en) Cyclopropyl-amide compounds as LSD1 / HDAC dual inhibitors
CN112566916B (en) Substituted Thienopyrroles as PAD4 Inhibitors
KR20160094980A (en) (6s,9as)-n-benzyl-6-[(4-hydroxyphenyl)methyl]-4,7-dioxo-8-({6-[3-(piperazine-1-yl)azetidine-1-yl]pyridine-2-yl}methyl)-2-(prop-2-en-1-yl)-octahydro-1h-pyrazino[2,1-c][1,2,4]triazine-1-carboxamide compound
JP2011148811A (en) Biarylurea derivative
JPWO2004096806A1 (en) Condensed imidazole derivatives
CA2971114A1 (en) Pyrrolopyrimidine compound
BR112020013759A2 (en) heterocyclylamino-substituted triazoles as modulators of rho-associated protein kinase
JP7260718B2 (en) Diazaindole derivatives and their use as Chk1 inhibitors
US9000182B2 (en) 2H-imidazol-4-amine compounds and their use as BACE inhibitors
KR20210120070A (en) immunomodulators
CN112300153B (en) Heterocyclic compound, pharmaceutical composition and application
AU2021339298A1 (en) Compounds for suppressing egfr mutant cancer and pharmaceutical use thereof
JP2010523530A (en) [2,6] Naphthyridine useful as a protein kinase inhibitor
JP2021513984A (en) TRPC6 inhibitor
WO2005021550A1 (en) Bicyclic pyrazole derivative
JP2022526295A (en) Quinoline and quinazoline compounds and how to use them
TW202115035A (en) Heterocyclic amide compounds and preparation method and application thereof
WO2020208002A1 (en) Inhibitors of trpc6
KR102505664B1 (en) A compound containing EZH2 inhibitor and E3 ligase binder, and pharmaceutical composition for use in preventing or treating EZH2 related diseases containing the same as an active ingredient

Legal Events

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