KR20220117805A - Composition for stimulating interferon genes comprising a benzimidazole derivative as an active ingredient - Google Patents

Composition for stimulating interferon genes comprising a benzimidazole derivative as an active ingredient Download PDF

Info

Publication number
KR20220117805A
KR20220117805A KR1020220006893A KR20220006893A KR20220117805A KR 20220117805 A KR20220117805 A KR 20220117805A KR 1020220006893 A KR1020220006893 A KR 1020220006893A KR 20220006893 A KR20220006893 A KR 20220006893A KR 20220117805 A KR20220117805 A KR 20220117805A
Authority
KR
South Korea
Prior art keywords
compound
mmol
pyrazole
methyl
butyl
Prior art date
Application number
KR1020220006893A
Other languages
Korean (ko)
Inventor
김혜진
이상희
전민재
이혜림
Original Assignee
한국화학연구원
한국과학기술연구원
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, 한국과학기술연구원 filed Critical 한국화학연구원
Priority to PCT/KR2022/002319 priority Critical patent/WO2022177307A1/en
Publication of KR20220117805A publication Critical patent/KR20220117805A/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants

Abstract

The present disclosure provides a compound useful as a STING agonist and pharmaceutically acceptable salts thereof. The present disclosure provides a pharmaceutical composition comprising the compound or pharmaceutically acceptable salts thereof. In addition, the present disclosure provides a pharmaceutical use for treatment or prevention of STING-mediated diseases using the compound or pharmaceutically acceptable salts thereof as an active ingredient. Moreover, the present disclosure provides a method for treating or preventing STING-mediated diseases including administration of an effective amount of the compound, salts thereof or the composition comprising the same to a subject in need of treatment for STING-mediated diseases.

Description

벤즈이미다졸 유도체를 유효 성분으로 포함하는 인터페론 유전자 자극제 조성물{Composition for stimulating interferon genes comprising a benzimidazole derivative as an active ingredient}TECHNICAL FIELD [0002] Composition for stimulating interferon genes comprising a benzimidazole derivative as an active ingredient

본 개시는 다양한 종류의 암, 전암성 증후군, 감염성 질환 등과 같은 STING (Stimulator of Interferon genes) 매개 질환의 치료 또는 예방에 유용한 화합물 및 이의 용도에 관한 것이다.The present disclosure relates to compounds useful for the treatment or prevention of STING (Stimulator of Interferon genes) mediated diseases such as various types of cancer, precancerous syndromes, infectious diseases, and the like, and uses thereof.

암세포가 면역반응을 회피하는 면역회피기전을 발견한 이래로 인체의 면역 시스템을 이용하여 암을 치료하는 면역항암제가 차세대 항암제로 각광을 받고 있다. 종양 환경 하에서는 주로 암세포의 신호전달 기능이 활성화됨에 따라 면역세포들의 기능이 억제되거나, 항암 활성을 유도하는 면역 과정을 회피하여 종양이 형성되도록 한다. 면역 항암제는 세포 내에 문제가 생긴 부분에 대한 비정상적인 기능을 인식하고 이를 공격하여 효과적으로 제거할 수 있도록 면역세포를 활성화 시키는 체내의 다양한 단백질들을 표적으로 한다. 특히 PD-1, PD-L1, CTLA-4 등 면역관문을 표적으로 하는 면역관문억제제는 암세포가 면역세포를 회피하는 기전을 저해하고, 결과적으로 T세포 면역 반응성에 의하여 암세포 사멸을 유도하는 대표적인 면역항암제이다.Since the discovery of the immune evasion mechanism by which cancer cells evade immune responses, immuno-oncology drugs that use the body's immune system to treat cancer have been in the spotlight as next-generation anticancer agents. In a tumor environment, as the signaling function of cancer cells is mainly activated, the function of immune cells is suppressed or an immune process that induces anticancer activity is avoided to form a tumor. Immune anticancer drugs target various proteins in the body that activate immune cells so that they can recognize abnormal functions of problematic areas within cells and attack and effectively remove them. In particular, immune checkpoint inhibitors that target immune checkpoints, such as PD-1, PD-L1, and CTLA-4, inhibit the mechanism by which cancer cells evade immune cells and, as a result, induce cancer cell death by T cell immune reactivity. It is an anticancer drug.

이 때, 면역 세포, 기질 세포, 세포외기질 등으로 구성된 종양 미세 환경 (tumor microenvironment, TME)는 암의 형성 및 면역항암제의 반응성에 주요한 영향을 미친다. 따라서 TME는 면역반응을 이용한 항암제의 효능에도 영향을 주게 된다. 종양의 면역표현형은 T세포 반응성에 따라 크게 'hot tumor'와 'cold tumor'로 구분된다. 'Hot tumor'는 면역반응이 존재했으나 TME 내 면역 억제 작용에 의해 저해된 경우를 의미한다. 이들 대부분이 면역관문억제제에 대해 반응성을 보인다. 하지만 'cold tumor'의 경우 T cell이 침투하지 않는 면역 결핍 표현형으로, 면역관문억제제에 반응을 보이지 않는 환자가 존재한다. 뿐만 아니라 면역관문억제제에 반응을 보이는 경우에도 단독 치료 시에는 항암 활성이 크지 않다는 한계점을 갖는다.At this time, the tumor microenvironment (TME) composed of immune cells, stromal cells, extracellular matrix, etc. has a major influence on the formation of cancer and the reactivity of immuno-cancer drugs. Therefore, TME also affects the efficacy of anticancer drugs using the immune response. The immunophenotype of tumor is largely divided into 'hot tumor' and 'cold tumor' according to T cell reactivity. 'Hot tumor' refers to a case in which an immune response was present but was inhibited by the immunosuppressive action in TME. Most of them are responsive to immune checkpoint inhibitors. However, 'cold tumor' is an immune deficiency phenotype in which T cells do not penetrate, and there are patients who do not respond to immune checkpoint inhibitors. In addition, even when responding to immune checkpoint inhibitors, there is a limitation in that the anticancer activity is not great when treated alone.

최근 들어 면역 반응이 존재하지 않는 'cold tumor'의 한계점을 극복하기 위한 새로운 약물 표적으로 STING (Stimulator of Interferon Genes) 단백질이 각광받고 있다. STING 활성화에 의한 선천 면역 과정의 활성화는 T cell의 priming을 유도하고, 그 결과 후천 면역의 활성화를 통해 cytotoxic T cell에 의한 면역반응을 일으킬 수 있다는 장점이 있다. 이는 그 동안 면역관문억제제에 대한 반응성이 거의 없었던 'cold tumor'를 'hot tumor'로 변형시킬 수 있다는 점에서 새로운 치료 전략으로서 중요한 의미를 갖는다. Recently, STING (Stimulator of Interferon Genes) protein has been in the spotlight as a new drug target to overcome the limitation of 'cold tumor' in which no immune response exists. Activation of the innate immune process by STING activation induces priming of T cells, and as a result, it has the advantage of being able to induce an immune response by cytotoxic T cells through activation of acquired immunity. This has important significance as a new treatment strategy in that it can transform a 'cold tumor' that has had little reactivity to an immune checkpoint inhibitor into a 'hot tumor'.

포유류 세포의 세포질에서 cGAS-STING 신호 전달 체계는 외부 인자로부터 자극을 받아 시작되는 숙주의 첫 번째 방어 기전중의 하나이다. cGAS (Cyclic GMP-AMP synthase)가 외부 바이러스나 암에서 비롯된 dsDNA에 의해 자극받게 되면, cGAS에 의해 합성된 cGAMP가 STING의 기질로서 작용한다. cGAMP가 STING에 결합하면 TBK1(TANK binding kinase 1)를 리쿠르팅 및 인산화 시키고, 이것이 IRF3 (Interferon Regulatory transcription Factor 3)의 인산화를 유도한다. 그 결과 인산화된 IRF3가 전사인자로 작용하여 세포질에서 핵 안으로 이동하여 특정 프로모터에 결합하고 이를 활성화 시키면 type-1 Interferon과 여러 염증성 사이토카인을 생산한다. Interferon 및 관련 인자는 JAK-STAT pathway를 활성화 시켜 ISG (Interferon Stimulated Genes)들의 전사를 유도하고, 이 단백질들이 외부의 감염으로부터 숙주를 보호하기 위한 항바이러스성 기전을 제어한다. 다음과 같은 선천면역과정이 활성화 되면 APC (antigen presenting cell)에 의해 T cell이 priming되어 tumor에 특정적으로 작용하는 cytotoxic T cell이 활성화된다. In the cytoplasm of mammalian cells, the cGAS-STING signaling system is one of the first defense mechanisms of the host, which is initiated by stimuli from external factors. When cGAS (Cyclic GMP-AMP synthase) is stimulated by dsDNA derived from an external virus or cancer, cGAMP synthesized by cGAS acts as a substrate for STING. When cGAMP binds to STING, it recruits and phosphorylates TBK1 (TANK binding kinase 1), which induces phosphorylation of IRF3 (Interferon Regulatory transcription factor 3). As a result, phosphorylated IRF3 acts as a transcription factor, moves from the cytoplasm to the nucleus, binds to a specific promoter, and activates it to produce type-1 interferon and several inflammatory cytokines. Interferon and related factors activate the JAK-STAT pathway to induce transcription of ISGs (Interferon Stimulated Genes), and these proteins control the antiviral mechanism to protect the host from external infection. When the following innate immune processes are activated, T cells are primed by APC (antigen presenting cells), and cytotoxic T cells that specifically act on tumors are activated.

위의 면역항암제 작용 기전의 장점을 바탕으로 다양한 STING 작용제(agonist)가 보고되었다. 첫 번째로 알려진 CDN 계열의 작용제는 electronegative 하고 hydrophilic 성질을 갖는다는 한계가 있었다. 또, CDN 계열의 ADU-S100같은 경우 종양내(intratumoral) 투여 방법의 한계점으로 인하여 용량최적화가 어려울 뿐만 아니라 특정 단계를 넘어서면 면역반응이 과하게 활성화되어 사이토카인 폭풍에 의한 부작용이 있을 수 있다. Various STING agonists have been reported based on the advantages of the above mechanism of action of immuno-oncology. The first known CDN-type agonist has limitations in that it has electronegative and hydrophilic properties. In addition, in the case of CDN-type ADU-S100, it is difficult to optimize the dose due to the limitations of intratumoral administration methods, and if a certain stage is exceeded, the immune response is excessively activated, and there may be side effects due to a cytokine storm.

한편, 국제특허출원 공개번호 WO 2017/175147는 이러한 STING 작용제 화합물들을 개시하고 있으며, 이러한 화합물들이 암, 전암성 증후군, 감염성 질환 (예를 들어, 인플루엔자, HIV, HCV, HPV 또는 HBV 감염증 질환) 등을 포함하는 다양한 질환의 치료 또는 개선에 유용함을 개시하고 있다. 또한, STING 작용제가 면역원성 조성물 또는 백신 애주번트로 유용할 수 있음을 개시하고 있다.On the other hand, International Patent Application Publication No. WO 2017/175147 discloses such STING agonist compounds, such compounds are cancer, precancerous syndrome, infectious diseases (eg, influenza, HIV, HCV, HPV or HBV infectious disease), etc. It is disclosed that it is useful for the treatment or improvement of various diseases, including. It is also disclosed that STING agonists may be useful as immunogenic compositions or vaccine adjuvants.

국제특허출원 공개번호 WO 2017/175147International Patent Application Publication No. WO 2017/175147

따라서 본 발명이 해결하고자 하는 과제는 STING 작용제로서 유용한 신규 화합물 및 이의 의약 용도를 제공하는 것이다.Accordingly, the problem to be solved by the present invention is to provide a novel compound useful as a STING agonist and a pharmaceutical use thereof.

상기 과제를 해결하기 위하여, 본 개시의 일 양태는 하기 화학식 1의 화합물 또는 이의 약학적으로 허용 가능한 염을 제공한다. In order to solve the above problems, one aspect of the present disclosure provides a compound of Formula 1 or a pharmaceutically acceptable salt thereof.

[화학식 1][Formula 1]

Figure pat00001
Figure pat00001

상기 화학식 1에서, R5

Figure pat00002
(화합물 37),
Figure pat00003
(화합물 38), 또는
Figure pat00004
(화합물 39)임.In Formula 1, R 5 is
Figure pat00002
(Compound 37),
Figure pat00003
(Compound 38), or
Figure pat00004
(Compound 39).

바람직하게, 상기 화학식 1에서, R5

Figure pat00005
, 또는
Figure pat00006
이며, 더욱 바람직하게, R5
Figure pat00007
임.Preferably, in Formula 1, R 5 is
Figure pat00005
, or
Figure pat00006
and more preferably, R 5 is
Figure pat00007
lim.

본 발명의 다른 양태는 또한 2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1-(4-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)부틸)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 26) 또는 이의 약학적으로 허용 가능한 염을 제공한다.Another aspect of the present invention also relates to 2-(1-ethyl-3-methyl-1H-pyrazole-5-carboxamido)-1-(4-(1-ethyl-3-methyl-1H-pyrazole- 5-carboxamido)butyl)-1H-benzo[d]imidazole-5-carboxamide (Compound 26) or a pharmaceutically acceptable salt thereof.

본 개시에 따른 화합물들은 STING 단백질의 저분자 화합물 기반 약효 성분으로서, STING 관련 경로 활성화를 통해 선천면역반응이 일어날 수 있도록 자극시켜, 이에 의존적인 항암 활성 등 다양한 약리학적 활성을 나타내는 특성이 있다. 이는 저분자 합성 화합물로서, 정맥(intravenous) 투여가 가능하여 CDN 계열 약물의 한계점을 효과적으로 극복할 수 있으며, 기존 보고된 같은 계열의 화합물과 비교하였을 때, 매우 우수한 활성을 보여주고 있다. 이는 STING 작용제에 의한 선천 면역 반응과 그로 인한 후천 면역 활성화를 통해 항암 효과를 증진시켜 면역 항암 치료의 효율을 높일 수 있다는 점에서 그 중요성을 시사한다.The compounds according to the present disclosure are small-molecular compound-based pharmaceutical ingredients of the STING protein, which stimulate the innate immune response to occur through the activation of the STING-related pathway, thereby exhibiting various pharmacological activities such as anticancer activity dependent on it. This is a low-molecular synthetic compound, which can be administered intravenously, effectively overcoming the limitations of CDN-based drugs, and shows very good activity when compared to previously reported compounds of the same class. This suggests its importance in that it can enhance the effectiveness of immunotherapy by enhancing the anticancer effect through the innate immune response by the STING agonist and the resulting acquired immune activation.

본 개시에 따른 화합물들은 구조가 유사한 화합물들과 비교하여 활성, 물리화학적 안정성, 체내 안정성, 약물동력학적 특성, 안전성 등의 다양한 측면에서 큰 장점을 가진다. 구체적으로, 본 개시에 따른 화합물들은 국제특허출원 공개번호 WO 2017/175147에 개시된 화합물과 비교하여 훨씬 높은 활성을 나타낸다. The compounds according to the present disclosure have great advantages in various aspects, such as activity, physicochemical stability, in vivo stability, pharmacokinetic properties, and safety, compared to compounds having similar structures. Specifically, the compounds according to the present disclosure show much higher activity compared to the compounds disclosed in International Patent Application Publication No. WO 2017/175147.

또한, 본 개시에 따른 화합물들의 우수한 활성은 STING 경로 활성화의 대표적인 마커인 IFN-beta와 같은 사이토카인 분비에 대한 활성 검증에서도 그대로 나타나서, IFN-beta 분비에 대한 EC50 값을 측정하였을 때, 국제특허출원 공개번호 WO 2017/175147에 개시된 화합물 중 대표적인 화합물(예를 들어, 실시예 14 화합물)은 대략 130 nM - 200 nM 값을 보였지만 본 개시에 따른 화합물들의 경우 이보다 훨씬 낮은 4 pM 내지 1.6 nM값을 나타내었다.In addition, the excellent activity of the compounds according to the present disclosure is shown as it is in the verification of the activity for the secretion of cytokines such as IFN-beta, which is a representative marker of STING pathway activation, and when measuring the EC 50 value for IFN-beta secretion, international patent Among the compounds disclosed in Application Publication No. WO 2017/175147, a representative compound (eg, Example 14 compound) showed approximately 130 nM - 200 nM values, but the compounds according to the present disclosure showed much lower values of 4 pM to 1.6 nM. indicated.

또한, 본 개시에 따른 화합물들은 구조가 유사한 다른 화합물들 대비 반감기, AUC, clearance 등 약동학적 특성에서도 큰 차이(장점)를 나타내었으며, 체내대사안정성이 월등히 뛰어나다. 예를 들어, 국제특허출원 공개번호 WO 2017/175147에 개시된 화합물 중 대표적인 화합물(예를 들어, 실시예 14 화합물)은 반감기가 1.4 시간, AUC가 3 μg·h-1·ml-1, volume of distribution이 0.4 L·kg-1, clearance가 17 ml·min-1·kg-1인데, 같은 조건(정맥주사, 3 mpk)에서 본 발명 화합물의 일 실시예 화합물인 화합물 37은 반감기가 반감기가 10.5 시간, AUC가 4.2 μg·h-1·ml-1, volume of distribution이 17.7 L·kg-1, clearance가 2.1 ml·min-1·kg-1로 월등히 뛰어나다.In addition, the compounds according to the present disclosure exhibited a large difference (advantage) in pharmacokinetic properties such as half-life, AUC, and clearance compared to other compounds having similar structures, and their metabolic stability in the body is remarkably excellent. For example, a representative compound (eg, Example 14 compound) among the compounds disclosed in International Patent Application Publication No. WO 2017/175147 has a half-life of 1.4 hours and an AUC of 3 μg·h -1 ·ml -1 , volume of The distribution is 0.4 L·kg -1 , and the clearance is 17 ml·min -1 ·kg -1 . Under the same conditions (intravenous injection, 3 mpk), Compound 37, an exemplary compound of the present invention, has a half-life of 10.5 Time, AUC of 4.2 μg·h -1 ·ml -1 , volume of distribution 17.7 L·kg -1 , and clearance of 2.1 ml·min -1 ·kg -1 are exceptionally excellent.

본 발명에 있어 "약학적으로 허용 가능한 염"은 여기서 언급한 화합물에서 발견되는 특정 치환체에 의존하는 비교적 비독성 산으로 제조된 활성 화합물의 염들을 포함한다. 본 발명의 화합물의 산성 부가 염들은 충분한 양의 원하는 산, 순수한 또는 적당한 비활성(inert) 용매로 본 발명 화합물의 중성 형태를 접촉하여 얻을 수 있다. 약학적으로 허용 가능한 산성 부가 염의 예들은 초산, 프로피온산, 이소부틸산, 옥살릭산(oxalic), 마레익(maleic), 말로닉(malonic), 안식향성, 숙신산, 수버릭(suberic), 푸마릭(fumaric), 만데릭(mandelic), 프탈릭(phthalic), 벤젠설포닉(benzenesulfonic), p-토릴설포닉(tolylsulfonic), 구연산, 주석산, 메탄솔포닉(methanesulfonic), 및 그 유사체를 포함하는 상대적으로 비독성 유기산에서 유래한 염들 뿐만 아니라, 염화수소, 브롬화 수소, 질산, 탄산, 일수소탄산(monohydrogencarbonic), 인산(phosphoric), 일수소인산, 이수소인산, 황산, 일수소황산, 요오드화수소 또는 아인산(phosphorous acid) 및 그 유사체를 포함한다. 또한 알긴네이트(arginate)와 그 유사체와 같은 아미노산의 염 및 글루쿠로닉(glucuronic) 또는 갈락투노릭(galactunoric) 산들과 그 유사체와 같은 유기산의 유사체를 포함한다. "Pharmaceutically acceptable salts" in the present invention include salts of the active compounds prepared with relatively non-toxic acids which depend on the particular substituents found on the compounds mentioned herein. Acid addition salts of a compound of the present invention may be obtained by contacting the neutral form of a compound of the present invention with a sufficient amount of the desired acid, neat or with a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts are acetic acid, propionic acid, isobutyric acid, oxalic acid, maleic, malonic, benzoic, succinic, suberic, fumaric ( fumaric), mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric acid, tartaric acid, methanesulfonic, and the like. As well as salts derived from non-toxic organic acids, hydrogen chloride, hydrogen bromide, nitric acid, carbonic acid, monohydrogencarbonic, phosphoric, monohydrogenphosphate, dihydrogenphosphate, sulfuric acid, monohydrogensulfuric acid, hydrogen iodide or phosphorous acid ( phosphorous acid) and its analogues. Also included are salts of amino acids such as arginate and analogues thereof and analogues of organic acids such as glucuronic or galactunoric acids and analogues thereof.

본 명세서에서 사용된 용어인 "본 발명의 화합물"은 화학식 1의 화합물(들)뿐만 아니라, 이의 클라드레이트(clathrates), 수화물, 용매화물, 또는 다형체를 포함하는 의미이다. 또한 용어 "본 발명의 화합물"은 이의 약학적으로 허용 가능한 염이 언급되지 않을 경우 본 발명 화합물의 약학적으로 허용 가능한 염도 포함하는 의미이다. As used herein, the term “compound of the present invention” is meant to include the compound(s) of Formula 1 as well as clathrates, hydrates, solvates, or polymorphs thereof. In addition, the term "compound of the present invention" is meant to include pharmaceutically acceptable salts of the compounds of the present invention unless a pharmaceutically acceptable salt thereof is mentioned.

일 실시예에 본 발명의 화합물은 입체이성질체적으로 순수한 화합물들(예를 들어, 다른 입체이성질체가 실질적으로 없는(예를 들어, 85% ee 이상, 90% ee 이상, 95% ee 이상, 97% ee 이상, 또는 99% ee 이상))로 존재할 수 있다. 즉, 본 발명에 따른 화학식 1의 화합물 또는 그의 염은 아민-이민 호변이성질체들이 존재할 수 있으며 이들 또한 본 발명의 화합물 범주에 포함된다.In one embodiment, a compound of the invention is a stereoisomerically pure compound (e.g., substantially free of other stereoisomers (e.g., at least 85% ee, at least 90% ee, at least 95% ee, 97% ee or more, or 99% ee or more)). That is, in the compound of Formula 1 or a salt thereof according to the present invention, amine-imine tautomers may exist, and these are also included in the scope of the compound of the present invention.

본 명세서에서 사용될 경우, 용어 "결정다형(polymorph)"은 본 발명의 화합물의 고체 결정 형태 또는 그것의 복합체를 의미한다. 같은 화합물의 다른 결정다형은 다른 물리적, 화학적 그리고/또는 스펙트럼적 특성을 보인다. 물리적 특성 측면의 차이점으로는 안정성(예를 들어, 열 또는 빛 안정성), 압축성과 밀도(제제화 및 생산물 제조에 중요함), 그리고 용해율(생물학적 이용률에 영향을 줄 수 있음)을 포함하나, 이에 한정되지 아니한다. 안정성에서 차이는 화학반응성 변화들(예를 들어, 또 다른 다형으로 구성되었을 때보다 하나의 다형으로 구성되었을 때 더 빠르게 변색이 되는 것 같은 차별적 산화) 또는 기계적인 특징들(예를 들어 동역학적으로 선호된 다형체로서 저장된 정제 파편들이 열역학 적으로 더 안정된 다형으로 변환) 또는 둘 다(하나의 다형의 정제는 높은 습도에서 더 분해에 예민)를 야기한다. 결정다형의 다른 물리적 성질들은 그들의 가공에 영향을 줄 수 있다. 예를 들어, 한 결정다형은 또 다른 결정다형에 비하여, 예를 들어, 그것의 형태 또는 입자의 크기 분포에 기인하여 용매화합물을 형성할 가능성이 많을 수 있거나, 여과 또는 세척이 더 어려울 수 있다.As used herein, the term “polymorph” refers to a solid crystalline form of a compound of the present invention or a complex thereof. Different polymorphs of the same compound exhibit different physical, chemical and/or spectral properties. Differences in physical properties include, but are not limited to, stability (eg, thermal or light stability), compressibility and density (important for formulation and product manufacturing), and dissolution rate (which may affect bioavailability). doesn't happen Differences in stability may be due to changes in chemical reactivity (e.g., differential oxidation, such as a faster discoloration when composed of one polymorph than when composed of another polymorph) or mechanical properties (e.g., kinetically Tablet fragments stored as the preferred polymorph are converted to the thermodynamically more stable polymorph) or both (tablets of one polymorph are more susceptible to degradation at high humidity). Other physical properties of polymorphs can affect their processing. For example, one polymorph may be more likely to form a solvate than another polymorph, for example due to its shape or particle size distribution, or it may be more difficult to filter or wash.

본 명세서에서 사용된 용어 "용매 화합물"은 비공유 분자간의 힘에 의해 결합된 화학량론적 또는 비-화학량론적인 양의 용매를 포함하는 본 발명의 화합물 또는 이의 약학적으로 허용 가능한 염을 의미한다. 바람직한 용매들은 휘발성이고, 비독성이며, 인간에게 극소량 투여될 수 있다.As used herein, the term "solvent compound" refers to a compound of the present invention, or a pharmaceutically acceptable salt thereof, comprising a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces. Preferred solvents are volatile, non-toxic, and can be administered in trace amounts to humans.

본 명세서에서 사용된 용어 "수화물(hydrate)"은 비공유 분자간의 힘에 의해 결합된 화학량론적 또는 비-화학량론적인 양의 물을 포함하는 본 발명의 화합물 또는 이의 약학적으로 허용 가능한 염을 의미한다. As used herein, the term "hydrate" refers to a compound of the present invention, or a pharmaceutically acceptable salt thereof, comprising a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces. .

본 명세서에서 사용된 용어 "클라드레이트(clathrate)"은 게스트 분자(예를 들어, 용매 또는 물)를 가두어 놓은 공간(예를 들어, 채널(channel))을 포함한 결정 격자의 형태의 본 발명의 화합물 또는 그것의 염을 의미한다.As used herein, the term "clathrate" refers to a compound of the present invention in the form of a crystal lattice containing spaces (eg, channels) that confine guest molecules (eg, solvent or water). or salts thereof.

본 명세서에서 사용된 용어 "정제된(purified)"은 분리될 때, 분리체는 90% 이상 순수한 것을 의미하며, 일 실시예에서는 95% 이상 순수하고, 다른 실시 예에서는 99% 이상 순수하고, 또 다른 실시예에서는 99.9% 이상 순수한 것을 의미한다.The term "purified" as used herein, when isolated, means that the isolate is at least 90% pure, in one embodiment at least 95% pure, in another embodiment at least 99% pure, and In another embodiment, it means at least 99.9% pure.

용어 "약학적으로 허용 가능한"은 약학적 제제로 사용하기에 적합한 것을 의미하며, 일반적으로 이러한 사용을 위하여 안전한 것으로 간주되며, 이러한 사용을 위하여 국가의 관리 기관에 의하여 공식적으로 승인되거나 한국 약전 또는 미국 약전의 명단에 있는 것을 의미한다. The term "pharmaceutically acceptable" means suitable for use as a pharmaceutical preparation, which is generally considered safe for such use, and is officially approved for such use by a national regulatory agency or It means being on the list of pharmacopeias.

본 개시의 일 양태는 본 발명 화합물 또는 이의 약학적으로 허용 가능한 염의, STING 매개 질환의 치료 또는 예방을 위한 의약 용도를 제공한다. One aspect of the present disclosure provides a pharmaceutical use of the compound of the present invention or a pharmaceutically acceptable salt thereof for the treatment or prevention of STING-mediated diseases.

본 명세서에 있어, "STING 매개 질환"은 STING의 활성화로 인해 치료될 수 있는 질환을 의미하며, 장애(disorder)를 포함하는 의미이다. STING 매개 질환으로는 다양한 종류의 암 (예를 들어, 고형암, 비 T세포 반응성 암); 전암성 증후군; 감염성 질환 (예를 들어, 바이러스 또는 박테리아 매개 감염성 질환) 등이 있다. 본 발명의 일 양태에 있어, 상기 감염성 질환은 인플루엔자, HIV, HCV, HPV, HBV, PIV, HRV (rhinovirus), 엔테로바이러스, 폴리오바이러스, 지카바이러스, 뎅기바이러스, 코로나바이러스 등으로 인해 야기된 감염성 질환이다. 바람직하게, 상기 STING 매개 질환은 암 또는 전암성 증후군이다.As used herein, "STING-mediated disease" refers to a disease that can be treated due to activation of STING, and is meant to include disorders. STING-mediated diseases include various types of cancer (eg, solid cancer, non-T cell responsive cancer); precancerous syndrome; infectious diseases (eg, viral or bacterial mediated infectious diseases); In one aspect of the present invention, the infectious disease is an infectious disease caused by influenza, HIV, HCV, HPV, HBV, PIV, HRV (rhinovirus), enterovirus, poliovirus, Zika virus, dengue virus, coronavirus, etc. to be. Preferably, the STING mediated disease is cancer or a precancerous syndrome.

따라서, 본 개시는 또한 본 발명 화합물 또는 이의 약학적으로 허용 가능한 염을 유효 성분으로 포함하는 STING 매개 질환, 예를 들어, 다양한 종류의 암(예를 들어, 고형암, 비 T세포 반응성 암), 전암성 증후군, 감염성 질환(예를 들어, 바이러스 또는 박테리아 매개 감염성 질환) 등과 같은 질환의 치료 또는 예방용 약학 조성물을 제공한다. 본 개시의 다른 양태는 또한 본 발명의 화합물 또는 이의 약학적으로 허용 가능한 염의 치료학적으로 유효한 양을 STING-매개 질환의 치료 또는 예방이 필요한 개체에게 투여하는 것을 특징으로 하는, STING-매개 질환의 치료 또는 예방 방법을 제공한다. 또 다른 양태에서, 상기 개체는 동물이다. 다른 바람직한 양태에서, 상기 개체는 인간이다.Accordingly, the present disclosure also provides a STING-mediated disease comprising the compound of the present invention or a pharmaceutically acceptable salt thereof as an active ingredient, for example, various types of cancer (eg, solid cancer, non-T-cell reactive cancer), Provided is a pharmaceutical composition for treating or preventing diseases such as cancerous syndromes and infectious diseases (eg, viral or bacterial mediated infectious diseases). Another aspect of the present disclosure is also characterized in that a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof is administered to an individual in need of treatment or prevention of a STING-mediated disease, treatment of a STING-mediated disease or a preventive method. In another embodiment, the subject is an animal. In another preferred embodiment, the subject is a human.

본 명세서에서 사용된 "유효량" 또는 "유효한 양"은 본 발명 화합물의 의약 용도가 암 또는 전암성 증후군인 경우 원발, 국소성 또는 전이성(metastatic) 암세포 또는 암조직을 파괴, 변형, 통제 또는 제거하거나; 암의 확장을 늦추거나 또는 최소화하거나; 또는 암, 신생물 질환, 또는 종양의 치료 또는 관리에서 치료상 이점을 제공하기에 충분한 본 발명의 화합물의 양을 말한다. "유효량" 은 또한 암 또는 신생물 세포 사멸을 야기하기에 충분한 본 발명의 화합물의 양을 말한다. "유효량"은 또한 생체외(in vitro) 또는 생체내(in vivo) 어떤 쪽이든 STING을 활성화하기에 충분한 양을 말한다. As used herein, "effective amount" or "effective amount" means destroying, modifying, controlling or eliminating primary, local or metastatic cancer cells or cancer tissue when the pharmaceutical use of the compounds of the present invention is cancer or a precancerous syndrome; slowing or minimizing the spread of cancer; or an amount of a compound of the invention sufficient to provide a therapeutic benefit in the treatment or management of cancer, neoplastic disease, or tumor. “Effective amount” also refers to an amount of a compound of the invention sufficient to cause cancer or neoplastic cell death. "Effective amount" also refers to an amount sufficient to activate STING, either in vitro or in vivo.

본 명세서에서 사용된 용어 "신생물(neoplastic)"은 양성이거나 암성일지 모르는 세포 또는 조직(예를 들어, 종기)의 비정상적 성장을 의미한다.As used herein, the term “neoplastic” refers to an abnormal growth of cells or tissues (eg, boils) that may be benign or cancerous.

본 명세서에서 사용된 "치료"는 본 발명 화합물의 의약 용도가 암 또는 전암성 증후군인 경우 원발, 국소성 또는 전이성 암조직의 근절, 제거, 변형, 또는 통제를 포함하고; 암의 확장을 최소화하거나 지연시키는 것이다. "Treatment" as used herein includes eradication, removal, modification, or control of primary, focal or metastatic cancer tissue when the medicinal use of the compound of the present invention is cancer or a precancerous syndrome; To minimize or delay the expansion of cancer.

본 명세서에서 사용된 "유효량" 또는 "유효한 양"은 본 발명 화합물의 의약 용도가 감염성 질환인 경우 감염원으로 인해 야기된 증상을 완화, 통제 또는 제거하거나; 감염원 자체를 통제 또는 제거하거나 이의 전파를 늦추거나 또는 최소화하거나; 또는 감염성 질환의 치료 또는 관리에서 치료상 이점을 제공하기에 충분한 본 발명의 화합물의 양을 말한다. "유효량"은 또한 생체외(in vitro) 또는 생체내(in vivo) 어떤 쪽이든 STING을 활성화하기에 충분한 양을 말한다. As used herein, "effective amount" or "effective amount" means alleviating, controlling or eliminating symptoms caused by an infectious agent when the pharmaceutical use of the compound of the present invention is an infectious disease; control or eliminate the source of infection itself or slow or minimize its spread; or an amount of a compound of the invention sufficient to provide a therapeutic benefit in the treatment or management of an infectious disease. "Effective amount" also refers to an amount sufficient to activate STING, either in vitro or in vivo.

본 명세서에서 사용된 "치료"는 본 발명 화합물의 의약 용도가 감염성 질환인 경우 감염원의 근절, 제거, 또는 통제를 포함하고; 감염성 질환 증상의 확장을 최소화하거나 지연시키는 것이다. "Treatment" as used herein includes eradication, elimination, or control of an infectious agent when the medicinal use of a compound of the present invention is an infectious disease; Minimizing or delaying the spread of infectious disease symptoms.

본 명세서에서 사용된 "예방(prevention)"은 환자에서 상기 STING 매개 질환의 재발의 방지를 의미하며, 확장 또는 발병의 예방을 포함한다.As used herein, “prevention” refers to the prevention of recurrence of the STING-mediated disease in a patient, and includes prevention of expansion or onset.

본 발명의 화합물 또는 이의 약학적으로 허용 가능한 염은 일반적으로 치료적으로 유효한 양이 투여된다. 본 발명의 화합물은 임의의 적합한 경로에 의하여 이러한 경로에 적당한 약학 조성물의 형태, 그리고 의도된 치료를 위하여 효과적인 투여량으로 투여될 수 있다. 효과적인 투여량은 단일 또는 분할 투여로 일반적으로 약 0.0001 내지 약 200 mg/체중kg/일이고, 바람직하게는 약 0.001 내지 약 100 mg/kg/일이고, 더욱 바람직하게는 약 0.015 내지 15 mg/체중kg/일이다. 나이, 종, 및 치료될 질병 또는 상태(condition)에 따라 이 범위의 하한 미만의 투여량 수준이 적합할 수 있다. 다른 경우에는, 여전히 더 큰 투여량이 해로운 부작용없이 사용될 수 있다. 더 큰 투여량은 하루 동안 투여를 위하여, 여러 작은 투여량으로 분할될 수 있다. 적절한 투여량을 결정하기 위한 방법들이 본 발명이 속한 분야에 잘 알려져 있다. The compound of the present invention or a pharmaceutically acceptable salt thereof is generally administered in a therapeutically effective amount. The compounds of the present invention may be administered by any suitable route, in the form of a pharmaceutical composition suitable for such route, and in an effective dosage for the intended treatment. An effective dosage is generally from about 0.0001 to about 200 mg/kg body weight/day, preferably from about 0.001 to about 100 mg/kg/day, more preferably from about 0.015 to 15 mg/body weight, in single or divided doses kg/day. Dosage levels below the lower limit of this range may be suitable depending on the age, species, and disease or condition being treated. In other cases, still larger doses can be used without deleterious side effects. The larger dose may be divided into several smaller doses for administration throughout the day. Methods for determining the appropriate dosage are well known in the art.

본 개시의 일 양태는 본 발명 화합물 또는 이의 약학적으로 허용 가능한 염의, 면역원성 조성물 또는 백신 애주번트로서의 의약 용도를 제공한다. 따라서, 본 개시는 또한 본 발명 화합물 또는 이의 약학적으로 허용 가능한 염을 애주번트로 포함하는 면역원성 또는 백신 조성물을 제공한다. One aspect of the present disclosure provides a pharmaceutical use of a compound of the present invention or a pharmaceutically acceptable salt thereof as an immunogenic composition or vaccine adjuvant. Accordingly, the present disclosure also provides an immunogenic or vaccine composition comprising a compound of the present invention or a pharmaceutically acceptable salt thereof as an adjuvant.

인터페론은 바이러스 감염으로부터 세포를 보호할 수 있는 물질이다. 예를 들어, 재조합 IFNα는 최초 승인된 생물학적 치료제이며, 바이러스 감염 및 암에서 중요한 요법이 되었다. 인터페론은 세포에 대한 직접적인 항바이러스 활성뿐만 아니라, 면역계의 세포에 작용하는 면역 반응의 강력한 조정제인 것으로 공지되어 있다. 본 발명의 화합물은 STING 작용제이며, IFN-베타 분비를 촉진할 뿐만 아니라, Type 1 IFN 매개 다양한 ISG 유전자 발현을 촉진한다.Interferon is a substance that can protect cells from viral infection. For example, recombinant IFNα was the first approved biotherapeutic agent and has become an important therapy in viral infections and cancer. Interferons are known to be potent modulators of immune responses acting on cells of the immune system, as well as direct antiviral activity against cells. The compounds of the present invention are STING agonists, and not only promote IFN-beta secretion, but also promote Type 1 IFN-mediated expression of various ISG genes.

STING 작용제가 바이러스 감염 및 자가면역 질환을 포함한 다양한 질환(예를 들어, 감염성 질환, 암, 알레르기성 질환, 신경병성 질환(예를 들어, 근위축성 측삭 경화증, 다발성 경화증 등), 염증성 질환, 과민성 장 질환)의 치료 또는 예방을 위한 중요한 전략이 될 수 있으며, 광범위한 DNA 및 RNA 바이러스 및 박테리아에 대한 보호를 포함한 항미생물 숙주 방어에 있어서 본질인 역할을 하고, 백신 애주번트로 유용할 수 있음은 국제특허출원 공개번호 WO 2017/175147에 공개되어 있으며, 상기 국제특허출원 공개번호 WO 2017/175147에 개시된 내용은 본 인용에 의해 그 전체가 본 명세서 내에 포함된다. STING agonists may be used in a variety of diseases including viral infections and autoimmune diseases (eg, infectious diseases, cancer, allergic diseases, neuropathic diseases (eg, amyotrophic lateral sclerosis, multiple sclerosis, etc.), inflammatory diseases, irritable bowel disease), plays an essential role in antimicrobial host defenses, including protection against a wide range of DNA and RNA viruses and bacteria, and may be useful as a vaccine adjuvant. The content disclosed in Application Publication No. WO 2017/175147 and disclosed in International Patent Application Publication No. WO 2017/175147 is incorporated herein by reference in its entirety.

본 개시의 다른 양태는 또한 본 발명의 화합물 또는 이의 약학적으로 허용 가능한 염, 및 약학적으로 허용 가능한 첨가제를 포함하는 조성물을 제공한다. 이러한 조성물은 다음과 같이 다양한 방법의 투여를 위한 형태로 제조될 수 있다.Another aspect of the present disclosure also provides a composition comprising a compound of the present invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable additive. Such a composition may be prepared in a form for administration by various methods as follows.

구강 투여(Oral administration)Oral administration

본 발명의 화합물은 구강으로 투여될 수 있으며, 구강은 연하(swallowing)를 포함하는 개념이다. 구강 투여에 의하여 본 발명의 화합물이 위장관(gastrointestinal tract)에 들어가거나, 예를 들어, 구강(buccal) 또는 설하(sublingual) 투여와 같이, 입으로부터 혈류로 직접적으로 흡수될 수 있다. The compound of the present invention may be administered orally, and the oral cavity is a concept including swallowing. By oral administration, the compound of the present invention may enter the gastrointestinal tract or may be absorbed directly into the bloodstream from the mouth, such as, for example, buccal or sublingual administration.

구강 투여를 위한 적합한 조성물은 고형상, 액상, 겔(gel), 또는 파우더 형상일 수 있으며, 정제(tablet), 로젠지(lozenge), 캡슐(capsule), 과립제, 산제 등의 제형을 가질 수 있다. Suitable compositions for oral administration may be in solid, liquid, gel, or powder form, and may have formulations such as tablets, lozenges, capsules, granules, and powders. .

구강 투여를 위한 조성물은 선택적으로 장용 코팅(enteric coating)될 수 있으며, 장용 코팅을 통하여 지연된(delayed) 또는 지속된(sustained) 방출을 나타낼 수 있다. 즉, 본 발명에 따른 구강 투여를 위한 조성물은 즉시 또는 변형된(modified) 방출 패턴을 가진 제형일 수 있다. Compositions for oral administration may optionally be enteric coated and may exhibit delayed or sustained release through the enteric coating. That is, the composition for oral administration according to the present invention may be a formulation having an immediate or modified release pattern.

액체 제형은 용액, 시럽 및 현탁액을 포함할 수 있으며, 이러한 액상 조성물은 연질 또는 경질 캡슐 내에 함유된 형태일 수 있다. 이러한 제형은 약학적으로 허용 가능한 담체, 예를 들어, 물, 에탄올, 폴리에틸렌글리콜, 셀룰로오스, 또는 오일(oil)을 포함할 수 있다. 상기 제형은 또한 하나 이상의 유화제 및/또는 현탁제를 포함할 수 있다.Liquid formulations may include solutions, syrups and suspensions, and such liquid compositions may be in the form contained within soft or hard capsules. Such formulations may contain a pharmaceutically acceptable carrier, for example, water, ethanol, polyethylene glycol, cellulose, or oil. The formulation may also contain one or more emulsifying and/or suspending agents.

정제(tablet) 제형에서, 활성 성분인 약물의 양은 정제 총 중량 대비 약 0.05 중량% 내지 약 95 중량%, 더욱 일반적으로 제형의 약 2 중량% 내지 약 50 중량%로 존재할 수 있다. 또한, 정제는 약 0.5 중량% 내지 약 35 중량%, 더욱 일반적으로 제형의 약 2 중량% 내지 약 25 중량%를 포함하는 붕해제를 함유할 수 있다. 붕해제의 예로는 유당, 전분, 소디움스타치글리콜레이트, 크로스포비돈, 크로스카멜로스소디움(croscarmellose sodium), 말토덱스트린 또는 이들의 혼합물이 사용될 수 있으나 이에 한정되는 것은 아니다.In tablet formulations, the amount of drug as the active ingredient may be present in an amount of from about 0.05% to about 95% by weight relative to the total weight of the tablet, more typically from about 2% to about 50% by weight of the dosage form. Tablets may also contain from about 0.5% to about 35% by weight of a disintegrant, more typically from about 2% to about 25% by weight of the dosage form. Examples of the disintegrant include, but are not limited to, lactose, starch, sodium starch glycolate, crospovidone, croscarmellose sodium, maltodextrin, or mixtures thereof.

정제로 제조하기 위해 포함되는 적합한 활택제는 약 0.1 중량% 내지 약 5 중량% 양으로 존재할 수 있고, 탈크(talc), 이산화규소, 스테아린산, 칼슘, 아연 또는 마그네슘 스테아레이트, 소듐 스테아릴 푸마레이트 등이 활택제로 사용될 수 있으나, 본 발명은 이러한 첨가제들의 종류에 한정되는 것은 아니다. Suitable glidants included for the preparation of tablets may be present in an amount from about 0.1% to about 5% by weight, and include talc, silicon dioxide, stearic acid, calcium, zinc or magnesium stearate, sodium stearyl fumarate, and the like. This lubricant may be used, but the present invention is not limited to the types of these additives.

정제로 제조하기 위한 결합제(binder)로는 젤라틴, 폴리에틸렌글리콜, 당(sugar), 검(gum), 녹말(starch), 폴리비닐피롤리돈, 하이드록시프로필셀룰로오스, 하이드록시프로필메틸셀룰로오스 등이 사용될 수 있으며, 정제로 제조하기 위한 적합한 희석제로는 만니톨, 자일리톨, 락토오스, 덱스트로오스, 수크로오스, 솔비톨, 녹말(starch), 미결정셀룰로오스 등이 사용될 수 있으나, 본 발명은 이러한 첨가제들의 종류에 한정되는 것은 아니다. Gelatin, polyethylene glycol, sugar, gum, starch, polyvinyl pyrrolidone, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, etc. may be used as a binder for manufacturing tablets. In addition, suitable diluents for manufacturing tablets include mannitol, xylitol, lactose, dextrose, sucrose, sorbitol, starch, microcrystalline cellulose, etc., but the present invention is not limited to the types of these additives. .

선택적으로 정제에 포함될 수 있는 가용화제는 정제 총 중량 대비 약 0.1 중량% 내지 약 3 중량% 양이 사용될 수 있고, 예를 들어, 폴리소르베이트, 소디움 라우릴설페이트, 소디움 도데실설페이트, 프로필렌 카보네이트, 디에틸렌글리콜모노에틸에테르, 디메틸이소소르비드, 폴리옥시에틸렌글리콜화된 천연 또는 수소화 피마자유, HCORTM(Nikkol), 올레일에스테르, 젤루시어(GelucireTM), 카프릴릭/카프릴산 모노/디글리세리드, 소르비탄지방산에스테르, 솔루톨HSTM 등이 본 발명에 따른 약학 조성물에 사용될 수 있으나, 본 발명은 이러한 가용화제의 구체적 종류에 한정되는 것은 아니다.Optionally, the solubilizer that may be included in the tablet may be used in an amount of about 0.1% to about 3% by weight based on the total weight of the tablet, for example, polysorbate, sodium lauryl sulfate, sodium dodecyl sulfate, propylene carbonate, Diethylene glycol monoethyl ether, dimethylisosorbide, polyoxyethylene glycolated natural or hydrogenated castor oil, HCOR (Nikkol), oleyl ester, Gelucire , caprylic/caprylic acid mono/ Diglyceride, sorbitan fatty acid ester, Solutol HS TM , etc. may be used in the pharmaceutical composition according to the present invention, but the present invention is not limited to the specific type of the solubilizer.

비경구 투여(Parenteral Administration)Parenteral Administration

본 발명의 화합물은 혈류, 근육, 또는 내장 내로 직접 투여될 수 있다. 비경구 투여를 위한 적합한 방법은 정맥내(intravenous), 근육내(intra-muscular), 피하 동맥내(subcutaneous intraarterial), 복강내(intraperitoneal), 척추강내(intrathecal), 두개내(intracranial) 주사 등을 포함한다. 비경구 투여를 위한 적합한 장치는 (바늘 및 바늘 없는 주사기를 포함하는) 주사기(injector) 및 주입 방법(infusion method)을 포함한다.The compounds of the present invention may be administered directly into the bloodstream, muscle, or intestine. Suitable methods for parenteral administration include intravenous, intra-muscular, subcutaneous intraarterial, intraperitoneal, intrathecal, intracranial injection, and the like. include Suitable devices for parenteral administration include injectors (including needle and needleless syringes) and infusion methods.

비경구 투여를 위한 조성물은 즉시 또는 변형된 방출 패턴을 가진 제형일 수 있으며, 변형된 방출 패턴은 지연된(delayed) 또는 지속된(sustained) 방출 패턴일 수 있다. Compositions for parenteral administration may be formulations with an immediate or modified release pattern, and the modified release pattern may be a delayed or sustained release pattern.

대부분의 비경구 제형은 액상 조성물이며, 이러한 액상 조성물은 본 발명에 따른 약효 성분, 염, 완충제, 등장화제 등을 포함하는 수용액이다.Most parenteral formulations are liquid compositions, and the liquid composition is an aqueous solution containing the active ingredient according to the present invention, a salt, a buffer, an isotonic agent, and the like.

비경구 제형은 또한 건조된 형태(예를 들어, 동결 건조) 또는 멸균 비-수용액으로서 제조될 수 있다. 이들 제형은 멸균수(sterile water)와 같은 적합한 비히클(vehicle)과 함께 사용될 수 있다. 용해도 증강제(solubility-enhancing agents) 또한 비경구 용액의 제조에 사용될 수 있다.Parenteral formulations may also be prepared in dried form (eg, lyophilized) or as sterile non-aqueous solutions. These formulations may be used with a suitable vehicle such as sterile water. Solubility-enhancing agents may also be used in the preparation of parenteral solutions.

국소 투여(Topical Administration)Topical Administration

본 발명의 화합물은 피부 또는 경피로 국소적으로 투여될 수 있다. 이 국소 투여를 위한 제형은 로션, 용액, 크림, 젤, 하이드로젤, 연고, 폼(foam), 임플란트(implant), 패치 등을 포함한다. 국소 투여 제형을 위한 약학적으로 허용 가능한 담체는 물, 알코올, 미네랄 오일, 글리세린, 폴리에틸렌글리콜 등을 포함할 수 있다. 국소 투여는 또한 전기천공법(electroporation), 이온도입법(iontophoresis), 음파영동(phonophoresis) 등에 의하여 수행될 수 있다.The compounds of the present invention may be administered topically dermally or transdermally. Formulations for topical administration include lotions, solutions, creams, gels, hydrogels, ointments, foams, implants, patches, and the like. Pharmaceutically acceptable carriers for topical dosage forms may include water, alcohol, mineral oil, glycerin, polyethylene glycol, and the like. Topical administration may also be performed by electroporation, iontophoresis, phonophoresis, and the like.

국소 투여를 위한 조성물은 즉시 또는 변형된 방출 패턴을 가진 제형일 수 있으며, 변형된 방출 패턴은 지연된(delayed) 또는 지속된(sustained) 방출 패턴일 수 있다. Compositions for topical administration may be formulations with an immediate or modified release pattern, and the modified release pattern may be a delayed or sustained release pattern.

한편, 본 개시의 다른 양태는 또한 본 발명의 화합물 또는 이의 약학적으로 허용 가능한 염을 애주번트로 포함하는 면역원성 조성물 또는 백신 조성물을 제공한다. 이러한 조성물은 단백질, DNA, RNA, 살아있는 또는 죽은 박테리아 및/또는 바이러스 또는 바이러스-유사 입자를 포함하나, 이에 제한되지는 않는 항체(들) 또는 항체 단편(들) 또는 항원 성분을 포함할 수 있다. 본 개시의 면역원성 또는 백신 조성물은 본 개시의 애주번트 이외에도 알루미늄 염, 오일, 열 쇼크 단백질, 지질 A 제제 및 유도체, 당지질, 다른 TLR (톨-유사 수용체) 효능제 (예컨대 CpG DNA 또는 유사한 작용제), 시토카인 (예컨대 GM-CSF, IL-12)을 포함하나, 이에 제한되지는 않는 애주번트 활성을 갖는 다른 1종 이상의 성분과 함께 함유할 수 있다.On the other hand, another aspect of the present disclosure also provides an immunogenic composition or vaccine composition comprising the compound of the present invention or a pharmaceutically acceptable salt thereof as an adjuvant. Such compositions may include antibody(s) or antibody fragment(s) or antigenic components including, but not limited to, proteins, DNA, RNA, live or dead bacteria and/or viruses or virus-like particles. The immunogenic or vaccine compositions of the present disclosure may contain, in addition to the adjuvants of the present disclosure, aluminum salts, oils, heat shock proteins, lipid A agents and derivatives, glycolipids, other TLR (Toll-Like Receptor) agonists (such as CpG DNA or similar agonists) , cytokines (such as GM-CSF, IL-12), including, but not limited to, one or more other ingredients having adjuvant activity.

본 개시는 다양한 종류의 암(예를 들어, 고형암, 비 T세포 반응성 암), 전암성 증후군, 감염성 질환(예를 들어, 바이러스 또는 박테리아 매개 감염성 질환) 등과 같은 STING 작용제에 의해 치료 또는 예방될 수 있는 STING 매개 질환의 치료 또는 예방에 유용한 화합물 및 이의 의약 용도를 제공한다. 본 개시에 따른 화합물은 의약품의 유효 성분으로 다양한 장점을 가진다.The present disclosure discloses that various types of cancers (eg, solid cancers, non-T-cell reactive cancers), precancerous syndromes, infectious diseases (eg, viral or bacterial mediated infectious diseases) can be treated or prevented by STING agonists, etc. Provided are compounds useful for the treatment or prevention of STING-mediated diseases and their medicinal uses. The compound according to the present disclosure has various advantages as an active ingredient of a pharmaceutical.

본 명세서에 첨부되는 다음의 도면들은 본 발명의 바람직한 실시예를 예시하는 것이며, 전술한 발명의 내용과 함께 본 발명의 기술사상을 더욱 이해시키는 역할을 하는 것이므로, 본 발명은 그러한 도면에 기재된 사항에만 한정되어 해석되어서는 아니 된다.
도 1은 본 발명에 따른 일 실시예 화합물들의 ISRE 리포터 어세이 결과이다. 이를 통해 본 발명 화합물의 농도 의존성 및 STING 의존성을 확인하였다.
도 2는 본 발명에 따른 일 실시예 화합물들의 MTS 세포생존률 평가 결과이다.
도 3은 본 발명에 따른 일 실시예 화합물의 시험관 내 리간드-경쟁 결합 실험 결과이다.
도 4는 CETSA(cellular thermal shift assay)를 이용한 본 발명 일 실시예 화합물의 STING 결합 여부 평가 결과이다. 도 4에서 위쪽 결과가 인간 면역세포 (THP-1)를 이용한 결과이며, 아래쪽 결과가 마우스 면역세포 (RAW264.7)를 이용한 결과이다.
도 5는 본 발명 일 실시예 화합물과 비교예인 화합물 31의 IFN-β 및 IP-10 사이토카인 분비에 대한 ELISA 결과이다.
도 6은 본 발명 일 실시예 화합물과 비교예인 화합물 diABZI-3의 IFN-β 및 IP-10 사이토카인 분비에 대한 ELISA 결과이다.
도 7은 본 발명 일 실시예 화합물에 의한 Type 1 IFN 매개 다양한 ISG 유전자 발현 검증 결과이다.
도 8은 STING 신호 전달 체계에 따른 하위 인자 활성화 확인을 위한 웨스턴 블랏팅 실험 결과이다.
도 9는 In vivo에서 본 발명 일 실시예 화합물의 면역항암효능을 평가한 결과이다 (Data represents as the mean volume of xenograft tumors ± SEM (N=6). Statistical difference was analyzed by Student's t test. *P > 0.05. Arrow indicated the day of compound injection)
도 10은 CT26 보유 마우스를 이용한 본 발명 일 실시예 화합물의 항암 효과 평가 결과이다 (Graphs are depicted by mean and SD. *: P<0.05. **: P<0.01 by two-way ANOVA.)
도 11은 본 발명 일 실시예 화합물(화합물 39)을 투여하였던 마우스에 화합물의 추가 투여 없이 CT26을 다시 심어서 종양 재발을 모니터링한 결과이다.
The following drawings attached to the present specification illustrate preferred embodiments of the present invention, and serve to further understand the technical idea of the present invention together with the above-described contents of the present invention, so the present invention is limited to the matters described in such drawings It should not be construed as being limited.
1 is an ISRE reporter assay result of the compounds of one embodiment according to the present invention. Through this, the concentration dependence and STING dependence of the compound of the present invention were confirmed.
2 is a result of evaluating the MTS cell viability of the compounds of one embodiment according to the present invention.
3 is an in vitro ligand-competitive binding experiment result of the compound of one embodiment according to the present invention.
4 is a result of evaluating whether the STING binding of the compound of one embodiment of the present invention using CETSA (cellular thermal shift assay). In FIG. 4 , the upper result is a result using human immune cells (THP-1), and the lower result is a result using mouse immune cells (RAW264.7).
5 is an ELISA result for the secretion of IFN-β and IP-10 cytokines of the compound of one embodiment of the present invention and the compound 31 of a comparative example.
6 is an ELISA result for IFN-β and IP-10 cytokine secretion of the compound of one embodiment of the present invention and the compound diABZI-3, which is a comparative example.
7 is a result of verifying the expression of various ISG genes mediated by Type 1 IFN by the compound of an embodiment of the present invention.
8 is a Western blotting experiment result for confirming the activation of a sub-factor according to the STING signal transduction system.
9 is a result of evaluating the immuno-anticancer efficacy of the compound of one embodiment of the present invention in vivo (Data represents as the mean volume of xenograft tumors ± SEM (N=6). Statistical difference was analyzed by Student's t test. *P > 0.05. Arrow indicated the day of compound injection)
10 is an anticancer effect evaluation result of the compound of one embodiment of the present invention using a CT26-bearing mouse (Graphs are depicted by mean and SD. *: P <0.05. **: P <0.01 by two-way ANOVA.)
11 is a result of monitoring tumor recurrence by re-implanting CT26 in mice administered with the compound (Compound 39) in Example 1 of the present invention without additional administration of the compound.

이하, 본 발명의 이해를 돕기 위하여 실시예 등을 들어 상세하게 설명하기로 한다. 그러나, 본 발명에 따른 실시예들은 여러 가지 다른 형태로 변형될 수 있으며, 본 발명의 범위가 하기 실시예들에 한정되는 것으로 해석되어서는 안 된다. 본 발명의 실시예들은 본 발명이 속한 분야에서 평균적인 지식을 가진 자에게 본 발명을 보다 완전하게 설명하기 위해 제공되는 것이다.Hereinafter, examples and the like will be described in detail to help the understanding of the present invention. However, the embodiments according to the present invention may be modified in various other forms, and the scope of the present invention should not be construed as being limited to the following examples. The embodiments of the present invention are provided to more completely explain the present invention to those of ordinary skill in the art to which the present invention pertains.

하기 표 3의 화합물(단량체)들의 합성Synthesis of compounds (monomers) of Table 3 below

Figure pat00008
Figure pat00008

단계 1: (Step 1: ( SS )-4-((2-히드록시-2-페닐에틸)아미노)-3-니트로벤즈아미드)-4-((2-hydroxy-2-phenylethyl)amino)-3-nitrobenzamide

Figure pat00009
Figure pat00009

4-플루오로-3-메톡시-5-니트로벤즈아미드 (1.50 g, 8.15 mmol)을 DMSO (10 mL)에 용해시킨 뒤 (S)-2-아미노-1-페닐에탄-1-올 (1.50 g, 8.15 mmol) 및 K2CO3 (1.69 g, 12.2 mmol)를 첨가하였다. 반응물을 60 ℃에서 16시간 동안 교반하고, 실온으로 냉각하였다. 반응 혼합물을 물로 희석하고 냉수로 여과한 뒤 고체를 진공 하에 건조시켜 노란색 고체상의 (S)-4-((2-히드록시-2-페닐에틸)아미노)-3-니트로벤즈아미드 (1.76 g, 5.79 mmol, 72% 수율)를 수득하였다. 4-Fluoro-3-methoxy-5-nitrobenzamide (1.50 g, 8.15 mmol) was dissolved in DMSO (10 mL) followed by ( S )-2-amino-1-phenylethan-1-ol (1.50 g, 8.15 mmol) and K 2 CO 3 (1.69 g, 12.2 mmol) were added. The reaction was stirred at 60 °C for 16 h and cooled to room temperature. The reaction mixture was diluted with water, filtered with cold water, and the solid dried under vacuum to obtain ( S )-4-((2-hydroxy-2-phenylethyl)amino)-3-nitrobenzamide (1.76 g, 5.79 mmol, 72% yield).

1 H NMR (300 MHz, methanol-d 4) δ 8.74 (d, J = 2.2 Hz, 1H), 7.94 (dd, J = 9.1, 2.2 Hz, 1H), 7.52-7.43 (m, 2H), 7.41-7.25 (m, 3H), 7.05 (d, J = 9.1 Hz, 1H), 4.98 (dd, J = 7.7, 4.4 Hz, 1H), 3.76-3.49 (m, 2H). 1 H NMR (300 MHz, methanol- d 4 ) δ 8.74 (d, J = 2.2 Hz, 1H), 7.94 (dd, J = 9.1, 2.2 Hz, 1H), 7.52-7.43 (m, 2H), 7.41 7.25 (m, 3H), 7.05 (d, J = 9.1 Hz, 1H), 4.98 (dd, J = 7.7, 4.4 Hz, 1H), 3.76-3.49 (m, 2H).

단계 2: (Step 2: ( SS )-3-아미노-4-((2-히드록시-2-페닐에틸)아미노)벤즈아미드)-3-amino-4-((2-hydroxy-2-phenylethyl)amino)benzamide

Figure pat00010
Figure pat00010

(S)-4-((2-히드록시-2-페닐에틸)아미노)-3-니트로벤즈아미드 (3.00 g, 9.96 mmol)을 MeOH (50 mL)에 용해 시킨 뒤 10% 습윤 Pd/C (106 mg, 0.996 mmol)을 첨가한 후 수소 풍선 하에 실온에서 18시간 동안 교반 시켰다. 이 후 MeOH 100 mL로 세척하면서 셀라이트®를 통해 여과하였다. 생성물을 함유하는 여과물을 진공하에 건조시켜 황갈색 고체상의 (S)-3-아미노-4-((2-히드록시-2-페닐에틸)아미노)벤즈아미드 (1.51 g, 5.57 mmol, 56% 수율)를 수득하였다. ( S )-4-((2-hydroxy-2-phenylethyl)amino)-3-nitrobenzamide (3.00 g, 9.96 mmol) was dissolved in MeOH (50 mL) followed by 10% wet Pd/C ( 106 mg, 0.996 mmol) was added and stirred for 18 hours at room temperature under a hydrogen balloon. It was then filtered through Celite®, washing with 100 mL of MeOH. The filtrate containing the product was dried under vacuum to yield ( S )-3-amino-4-((2-hydroxy-2-phenylethyl)amino)benzamide (1.51 g, 5.57 mmol, 56% yield) as a tan solid. ) was obtained.

1 H NMR (300 MHz, DMSO-d 6) δ 7.46-7.39 (m, 2H), 7.35 (ddd, J = 7.8, 6.7, 1.2 Hz, 2H), 7.30-7.23 (m, 1H), 7.12 (h, J = 2.1 Hz, 2H), 6.75 (s, 1H), 6.45 (d, J = 8.8 Hz, 1H), 5.51 (d, J = 4.4 Hz, 1H), 4.98 (dd, J = 6.9, 4.6 Hz, 1H), 4.80 (dt, J = 8.5, 4.4 Hz, 1H), 4.57 (s, 2H), 3.31-3.11 (m, 2H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 7.46-7.39 (m, 2H), 7.35 (ddd, J = 7.8, 6.7, 1.2 Hz, 2H), 7.30-7.23 (m, 1H), 7.12 (h) , J = 2.1 Hz, 2H), 6.75 (s, 1H), 6.45 (d, J = 8.8 Hz, 1H), 5.51 (d, J = 4.4 Hz, 1H), 4.98 (dd, J = 6.9, 4.6 Hz) , 1H), 4.80 (dt, J = 8.5, 4.4 Hz, 1H), 4.57 (s, 2H), 3.31-3.11 (m, 2H).

단계 3: (Step 3: ( SS )-2-아미노-1-(2-히드록시-2-페닐에틸)-1)-2-amino-1-(2-hydroxy-2-phenylethyl)-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복스아미드]Imidazole-5-carboxamide

Figure pat00011
Figure pat00011

(S)-3-아미노-4-((2-히드록시-2-페닐에틸)아미노)벤즈아미드 (1.00 g, 3.69 mmol)를 MeOH (6.15 mL)에 용해시킨 후 시아노겐 브로마이드 (0.977 g, 9.23 mmol) 첨가하고, 반응물을 60 ℃에서 3시간 동안 교반하였다. 이어서 생성물을 함유하는 잔여물을 진공하에 MeOH을 제거시켜 황갈색 고체상의 시아노겐 브로마이드를 함유한 (S)-2-아미노-1-(2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-5-카르복스아미드를 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. ( S )-3-amino-4-((2-hydroxy-2-phenylethyl)amino)benzamide (1.00 g, 3.69 mmol) was dissolved in MeOH (6.15 mL) followed by cyanogen bromide (0.977 g, 9.23 mmol) and the reaction was stirred at 60 °C for 3 h. The residue containing the product was then removed in vacuo by removal of MeOH to ( S )-2-amino-1-(2-hydroxy-2-phenylethyl) -1H -benzo[ d ] The imidazole-5-carboxamide was obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6 ) δ 7.77 (s, 1H), 7.69 (d, J = 1.6 Hz, 1H), 7.51-7.41 (m, 3H), 7.36-7.23 (m, 3H), 7.12-7.00 (m, 2H), 6.44 (s, 2H), 4.94 (q, J = 5.7 Hz, 1H), 4.10 (d, J = 5.7 Hz, 2H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 7.77 (s, 1H), 7.69 (d, J = 1.6 Hz, 1H), 7.51-7.41 (m, 3H), 7.36-7.23 (m, 3H), 7.12-7.00 (m, 2H), 6.44 (s, 2H), 4.94 (q, J = 5.7 Hz, 1H), 4.10 (d, J = 5.7 Hz, 2H).

단계 4: (Step 4: ( SS )-2-(산 치환기)아미도-1-(2-히드록시-2-페닐에틸)-1)-2-(acid substituent)amido-1-(2-hydroxy-2-phenylethyl)-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복스아미드]Imidazole-5-carboxamide

Figure pat00012
Figure pat00012

(S)-2-아미노-1-(2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-5-카르복스아미드 (100 mg, 0.338 mmol), 치환기 카르복실산 (1.2 eq, 0.406 mmol), HATU (643 mg, 1.69 mmol), 및 트리에틸아민 (0.565 mL, 4.06 mmol)을 NMP (1.13 mL)에 용해 시킨 후, 80 ℃에서 48시간 동안 가열하였다. 반응물에 물 (30 mL)을 첨가하고 EtOAc (4 X 40 mL)로 추출하였다. 유기층을 무수 MgSO4를 첨가하여 건조시키고 진공하에 용매를 제거하였다. 생성된 잔류물을 실리카겔 상에서 CH2Cl2:MeOH=9:1로 정제하였다. 그 후 목적화합물과 불순물이 포함된 분획을 합하고, 진공 하에 농축시키고, CH2Cl2 (10 mL)을 첨가하여 맑게 용해시킨 뒤 상온에서 10분간 두어 침전시켰다. 생성된 침전물을 여과하여 CH2Cl2 (4 X 10 mL)으로 헹구었다. 고체를 진공하에 건조시켜 표제 화합물들을 수득하였다.( S )-2-amino-1-(2-hydroxy-2-phenylethyl) -1H -benzo[ d ]imidazole-5-carboxamide (100 mg, 0.338 mmol), substituted carboxylic acid ( 1.2 eq, 0.406 mmol), HATU (643 mg, 1.69 mmol), and triethylamine (0.565 mL, 4.06 mmol) were dissolved in NMP (1.13 mL) and heated at 80 °C for 48 hours. To the reaction was added water (30 mL) and extracted with EtOAc (4 X 40 mL). The organic layer was dried by the addition of anhydrous MgSO 4 and the solvent was removed in vacuo. The resulting residue was purified on silica gel with CH 2 Cl 2: MeOH=9:1. After that, the fractions containing the target compound and impurities were combined, concentrated in vacuo, and cleared by adding CH 2 Cl 2 (10 mL), followed by precipitation at room temperature for 10 minutes. The resulting precipitate was filtered and rinsed with CH 2 Cl 2 (4 X 10 mL). The solid was dried under vacuum to afford the title compounds.

Figure pat00013
Figure pat00013

단계 1: 4-클로로-3-니트로벤즈아미드 Step 1: 4-Chloro-3-nitrobenzamide

Figure pat00014
Figure pat00014

메틸 4-클로로-3-니트로벤조에이트 (5.00 g, 23.2 mmol)를 NH4OH (28% 수용액, 100 mL)에 용해시킨 뒤 실온에서 16시간 동안 교반하였다. 16시간 동안 교반 후, 냉수로 고체를 여과한 뒤 건조하여 흰색 고체상의 4-클로로-3-니트로벤즈아미드 (3.43 g, 17.1 mmol, 74% 수율)를 수득하였다. Methyl 4-chloro-3-nitrobenzoate (5.00 g, 23.2 mmol) was dissolved in NH 4 OH (28% aqueous solution, 100 mL) and stirred at room temperature for 16 hours. After stirring for 16 hours, the solid was filtered with cold water and dried to obtain 4-chloro-3-nitrobenzamide (3.43 g, 17.1 mmol, 74% yield) as a white solid.

1 H NMR (300 MHz, methanol-d 4 ) δ 8.43 (d, J = 2.1 Hz, 1H), 8.11 (dd, J = 8.4, 2.1 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H). 1 H NMR (300 MHz, methanol- d 4 ) δ 8.43 (d, J = 2.1 Hz, 1H), 8.11 (dd, J = 8.4, 2.1 Hz, 1H), 7.78 (d, J = 8.4 Hz, 1H) .

단계 2: Step 2: terttert -부틸(4-((4-카르바모일-2-니트로페닐)아미노)부틸)카르바메이트-Butyl(4-((4-carbamoyl-2-nitrophenyl)amino)butyl)carbamate

Figure pat00015
Figure pat00015

N-boc-1,4-부탄디아민 (9.38 g, 49.8 mmol) 및 4-클로로-3-니트로벤즈아미드 (5.00 g, 24.9 mmol)을 DMSO (40 mL)에 용해시킨 뒤 K2CO3 (17.3 g, 125 mmol)를 첨가하였다. 반응물을 실온에서 16시간동안 교반하였다. 교반 후 냉수로 고체를 여과하고 진공 하에 건조시켜 노란색 고체상의 tert-부틸(4-((4-카르바모일-2-니트로페닐)아미노)부틸)카르바메이트 (5.73 g, 16.3 mmol, 65% 수율)를 수득하였다. N -boc-1,4-butanediamine (9.38 g, 49.8 mmol) and 4-chloro-3-nitrobenzamide (5.00 g, 24.9 mmol) were dissolved in DMSO (40 mL) followed by K 2 CO 3 (17.3) g, 125 mmol) was added. The reaction was stirred at room temperature for 16 h. After stirring, the solid was filtered with cold water and dried under vacuum to form a yellow solid tert- butyl(4-((4-carbamoyl-2-nitrophenyl)amino)butyl)carbamate (5.73 g, 16.3 mmol, 65%) yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6) δ 8.65 (d, J = 2.2 Hz, 1H), 8.37 (t, J = 5.8 Hz, 1H), 7.99 (dd, J = 8.9, 2.3 Hz, 2H), 7.31-7.22 (m, 1H), 7.10 (d, J = 9.1 Hz, 1H), 6.83 (t, J = 5.8 Hz, 1H), 3.40 (q, J = 6.7 Hz, 2H), 2.95 (q, J = 6.6 Hz, 2H), 1.60 (p, J = 7.3 Hz, 2H), 1.46 (p, J = 6.7 Hz, 2H), 1.36 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.65 (d, J = 2.2 Hz, 1H), 8.37 (t, J = 5.8 Hz, 1H), 7.99 (dd, J = 8.9, 2.3 Hz, 2H) , 7.31-7.22 (m, 1H), 7.10 (d, J = 9.1 Hz, 1H), 6.83 (t, J = 5.8 Hz, 1H), 3.40 (q, J = 6.7 Hz, 2H), 2.95 (q, J = 6.6 Hz, 2H), 1.60 (p, J = 7.3 Hz, 2H), 1.46 (p, J = 6.7 Hz, 2H), 1.36 (s, 9H).

단계 3: Step 3: terttert -부틸(4-((2-아미노-4-카르바모일페닐)아미노)부틸)카르바메이트-Butyl(4-((2-amino-4-carbamoylphenyl)amino)butyl)carbamate

Figure pat00016
Figure pat00016

tert-부틸(4-((4-카르바모일-2-니트로페닐)아미노)부틸)카르바메이트 (5.00 g, 14.2 mmol)를 MeOH (284 mL)에 용해시킨 뒤 10% 습윤 Pd/C (151 mg, 1.42 mmol)을 첨가하고 수소 풍선 하에 실온에서 18시간 동안 교반 시켰다. 이 후 MeOH 400 mL로 세척하면서 셀라이트®를 통해 여과하였다. 생성물을 함유하는 여과물을 진공하에 농축시켜 회색 고체로서 tert-부틸(4-((2-아미노-4-카르바모일페닐)아미노)부틸)카르바메이트을 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl(4-((4-carbamoyl-2-nitrophenyl)amino)butyl)carbamate (5.00 g, 14.2 mmol) was dissolved in MeOH (284 mL) followed by 10% wet Pd/C ( 151 mg, 1.42 mmol) was added and stirred under a hydrogen balloon at room temperature for 18 hours. It was then filtered through Celite®, washing with 400 mL of MeOH. The filtrate containing the product was concentrated in vacuo to give tert -butyl(4-((2-amino-4-carbamoylphenyl)amino)butyl)carbamate as a gray solid which was used in the next reaction without further purification. was used.

1 H NMR (500 MHz, methanol-d 4) δ 7.29 (dd, J = 8.3, 2.1 Hz, 1H), 7.22 (d, J = 2.1 Hz, 1H), 6.56 (d, J = 8.4 Hz, 1H), 3.19 (t, J = 6.9 Hz, 2H), 3.09 (t, J = 6.9 Hz, 2H), 1.68 (dq, J = 11.4, 6.7 Hz, 2H), 1.65-1.56 (m, 2H), 1.43 (s, 9H). 1 H NMR (500 MHz, methanol- d 4 ) δ 7.29 (dd, J = 8.3, 2.1 Hz, 1H), 7.22 (d, J = 2.1 Hz, 1H), 6.56 (d, J = 8.4 Hz, 1H) , 3.19 (t, J = 6.9 Hz, 2H), 3.09 (t, J = 6.9 Hz, 2H), 1.68 (dq, J = 11.4, 6.7 Hz, 2H), 1.65-1.56 (m, 2H), 1.43 ( s, 9H).

단계 4: Step 4: terttert -부틸(4-(2-아미노-5-카르바모일-1-Butyl(4-(2-amino-5-carbamoyl-1) HH -벤조[-benzo[ dd ]이미다졸-1-일)부틸)카르바메이트]Imidazol-1-yl)butyl)carbamate

Figure pat00017
Figure pat00017

tert-부틸(4-((2-아미노-4-카르바모일페닐)아미노)부틸)카르바메이트 (3.33 g, 10.3 mmol)를 MeOH (17.2 mL)에 용해시킨 뒤 시아노겐 브로마이드 (2.73 g, 25.8 mmol)를 첨가한 후, 60 ℃에서 3시간 동안 교반하였다. 이어서 생성물을 함유하는 잔여물을 진공하에 MeOH을 제거시켜 황갈색 고체상의 시아노겐 브로마이드를 함유한 tert-부틸(4-(2-아미노-5-카르바모일-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트를 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl(4-((2-amino-4-carbamoylphenyl)amino)butyl)carbamate (3.33 g, 10.3 mmol) was dissolved in MeOH (17.2 mL) followed by cyanogen bromide (2.73 g, 25.8 mmol) was added, and the mixture was stirred at 60 °C for 3 hours. The residue containing the product was then removed in vacuo with MeOH removed tert -butyl(4-(2-amino-5-carbamoyl- 1H -benzo[ d ]imidazole- containing cyanogen bromide as a tan solid) 1-yl)butyl)carbamate was obtained and used in the next reaction without further purification.

1 H NMR (500 MHz, DMSO-d 6) δ 8.61 (s, 2H), 8.05 (s, 1H), 7.86 (d, J = 1.5 Hz, 1H), 7.80 (dd, J = 8.4, 1.6 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.37 (s, 1H), 6.85 (t, J = 5.8 Hz, 1H), 4.15 (t, J = 7.4 Hz, 2H), 2.94 (q, J = 6.6 Hz, 2H), 1.64 (q, J = 7.7 Hz, 2H), 1.44 (p, J = 7.2 Hz, 2H), 1.35 (s, 9H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 8.61 (s, 2H), 8.05 (s, 1H), 7.86 (d, J = 1.5 Hz, 1H), 7.80 (dd, J = 8.4, 1.6 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.37 (s, 1H), 6.85 (t, J = 5.8 Hz, 1H), 4.15 (t, J = 7.4 Hz, 2H), 2.94 (q, J = 6.6 Hz, 2H), 1.64 (q, J = 7.7 Hz, 2H), 1.44 (p, J = 7.2 Hz, 2H), 1.35 (s, 9H).

단계 5: Step 5: terttert -부틸(4-(2-산치환기아미도-5-카르바모일-1-Butyl (4-(2-acid substituted groupamido-5-carbamoyl-1) HH -벤조[-benzo[ dd ]이미다졸-1-일)부틸)카르바메이트]Imidazol-1-yl)butyl)carbamate

Figure pat00018
Figure pat00018

tert-부틸(4-(2-아미노-5-카르바모일-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 (200 mg, 0.576 mmol), 치환기카르복실산 (1.2 eq, 0.691 mmol), HATU (1.10 g, 2.88 mmol), 및 트리에틸아민 (0.963 mL, 6.91 mmol)를 NMP (1.00 mL)에 용해시킨 뒤 80 ℃에서 48시간 동안 가열하였다. 반응물에 물 (30 mL)을 첨가하고 EtOAc (4 X 10 mL)로 추출하였다. 유기층을 무수 MgSO4을 첨가하여 건조시키고 진공하에 용매를 제거시켰다. 생성된 잔류물을 실리카 겔 상에서 CH2Cl2:MeOH=9:1로 정제하여 목적화합물을 수득하였다. tert -Butyl(4-(2-amino-5-carbamoyl- 1H -benzo[ d ]imidazol-1-yl)butyl)carbamate (200 mg, 0.576 mmol), substituted carboxylic acid (1.2 eq, 0.691 mmol), HATU (1.10 g, 2.88 mmol), and triethylamine (0.963 mL, 6.91 mmol) were dissolved in NMP (1.00 mL) and heated at 80 °C for 48 hours. To the reaction was added water (30 mL) and extracted with EtOAc (4 X 10 mL). The organic layer was dried by the addition of anhydrous MgSO 4 and the solvent was removed in vacuo. The resulting residue was purified on silica gel with CH 2 Cl 2 :MeOH=9:1 to obtain the target compound.

화합물 1 내지 16의 NMR 측정 결과를 하기 표 1에 종합하여 나타내었다.The NMR measurement results of compounds 1 to 16 are summarized in Table 1 below.

Compound
No.
Compound
No.
IUPAC nameIUPAC name 1H NMR 1 H NMR
1One (S)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1-(2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-5-카르복사미드( S )-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1-(2-hydroxy-2-phenylethyl) -1H -benzo[ d ]imi Dazole-5-carboxamide 1H NMR (400 MHz, DMSO-d 6 ) δ 7.78 (s, 1H), 7.70-7.58 (m, 3H), 7.52-7.28 (m, 5H), 7.04 (s, 1H), 6.75 (s, 2H), 6.61 (s, 1H), 6.19 (dd, J = 9.7, 3.7 Hz, 1H), 4.62 (dd, J = 15.4, 9.7 Hz, 1H), 4.42 (dd, J = 15.4, 3.8 Hz, 1H), 4.19 (qd, J = 6.9, 3.5 Hz, 2H), 2.14 (s, 3H), 1.08 (t, J = 7.1 Hz, 3H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 7.78 (s, 1H), 7.70-7.58 (m, 3H), 7.52-7.28 (m, 5H), 7.04 (s, 1H), 6.75 (s, 2H) ), 6.61 (s, 1H), 6.19 (dd, J = 9.7, 3.7 Hz, 1H), 4.62 (dd, J = 15.4, 9.7 Hz, 1H), 4.42 (dd, J = 15.4, 3.8 Hz, 1H) , 4.19 (qd, J = 6.9, 3.5 Hz, 2H), 2.14 (s, 3H), 1.08 (t, J = 7.1 Hz, 3H). 22 (S)-2-(1-에틸-3-(트리플루오로메틸)-1H-피라졸-5-카르복사미도)-1-(2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-5-카르복사미드( S )-2-(1-ethyl-3-(trifluoromethyl) -1H -pyrazole-5-carboxamido)-1-(2-hydroxy-2-phenylethyl) -1H- Benzo[ d ]imidazole-5-carboxamide 1H NMR (300 MHz, DMSO-d 6) δ 7.78 (s, 1H), 7.68-7.61 (m, 3H), 7.49 (dd, J = 8.3, 1.6 Hz, 1H), 7.45-7.33 (m, 4H), 7.27 (s, 1H), 7.05 (s, 1H), 6.76 (s, 2H), 6.24 (dd, J = 9.6, 3.6 Hz, 1H), 4.62 (dd, J = 15.3, 9.7 Hz, 1H), 4.44 (dd, J = 15.4, 3.7 Hz, 1H), 4.28 (q, J = 7.2 Hz, 2H), 1.35 (t, J = 7.2 Hz, 3H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 7.78 (s, 1H), 7.68-7.61 (m, 3H), 7.49 (dd, J = 8.3, 1.6 Hz, 1H), 7.45-7.33 (m, 4H) ), 7.27 (s, 1H), 7.05 (s, 1H), 6.76 (s, 2H), 6.24 (dd, J = 9.6, 3.6 Hz, 1H), 4.62 (dd, J = 15.3, 9.7 Hz, 1H) , 4.44 (dd, J = 15.4, 3.7 Hz, 1H), 4.28 (q, J = 7.2 Hz, 2H), 1.35 (t, J = 7.2 Hz, 3H). 33 (S)-1-(2-히드록시-2-페닐에틸)-2-(1-메틸-3-(트리플루오로메틸)-1H-피라졸-4-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드( S )-1-(2-hydroxy-2-phenylethyl)-2-(1-methyl-3-(trifluoromethyl) -1H -pyrazole-4-carboxamido) -1H- Benzo[ d ]imidazole-5-carboxamide 1H NMR (300 MHz, DMSO-d 6 ) δ 12.80 (s, 1H), 8.37 (s, 1H), 7.95 (d, J = 1.6 Hz, 2H), 7.72 (dd, J = 8.4, 1.7 Hz, 1H), 7.52-7.43 (m, 3H), 7.38-7.24 (m, 4H), 5.66 (d, J = 4.5 Hz, 1H), 5.08 (dt, J = 9.5, 5.1 Hz, 1H), 4.39-4.27 (m, 2H), 3.98 (s, 3H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.80 (s, 1H), 8.37 (s, 1H), 7.95 (d, J = 1.6 Hz, 2H), 7.72 (dd, J = 8.4, 1.7 Hz, 1H), 7.52-7.43 (m, 3H), 7.38-7.24 (m, 4H), 5.66 (d, J = 4.5 Hz, 1H), 5.08 (dt, J = 9.5, 5.1 Hz, 1H), 4.39-4.27 (m, 2H), 3.98 (s, 3H). 44 (S)-2-(푸란-3-카르복사미도)-1-(2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-5-카르복사미드( S )-2-(furan-3-carboxamido)-1-(2-hydroxy-2-phenylethyl) -1H -benzo[ d ]imidazole-5-carboxamide 1H NMR (500 MHz, methanol-d 4) δ 8.11 (d, J = 1.5 Hz, 1H), 7.73 (d, J = 1.7 Hz, 1H), 7.56 (dd, J = 8.3, 1.7 Hz, 1H), 7.52 (t, J = 1.8 Hz, 1H), 7.51-7.47 (m, 2H), 7.40-7.32 (m, 4H), 7.22 (d, J = 8.3 Hz, 1H), 6.67 (d, J = 1.8 Hz, 1H), 6.29 (dd, J = 8.6, 4.2 Hz, 1H), 4.60 (dd, J = 15.5, 8.6 Hz, 1H), 4.46 (dd, J = 15.5, 4.2 Hz, 1H). 1 H NMR (500 MHz, methanol- d 4 ) δ 8.11 (d, J = 1.5 Hz, 1H), 7.73 (d, J = 1.7 Hz, 1H), 7.56 (dd, J = 8.3, 1.7 Hz, 1H) , 7.52 (t, J = 1.8 Hz, 1H), 7.51-7.47 (m, 2H), 7.40-7.32 (m, 4H), 7.22 (d, J = 8.3 Hz, 1H), 6.67 (d, J = 1.8) Hz, 1H), 6.29 (dd, J = 8.6, 4.2 Hz, 1H), 4.60 (dd, J = 15.5, 8.6 Hz, 1H), 4.46 (dd, J = 15.5, 4.2 Hz, 1H). 55 (S)-2-(벤조푸란-2-카르복사미도)-1-(2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-5-카르복사미드( S )-2-(benzofuran-2-carboxamido)-1-(2-hydroxy-2-phenylethyl) -1H -benzo[ d ]imidazole-5-carboxamide 1H NMR (500 MHz, DMSO-d 6) δ 12.82 (s, 1H), 7.98 (d, J = 19.5 Hz, 2H), 7.83 (d, J = 7.8 Hz, 1H), 7.78-7.70 (m, 2H), 7.63 (s, 1H), 7.53 (dd, J = 13.2, 8.0 Hz, 3H), 7.46 (t, J = 7.7 Hz, 1H), 7.36 (dt, J = 21.6, 7.8 Hz, 4H), 7.27 (t, J = 7.4 Hz, 1H), 5.77 (d, J = 4.7 Hz, 1H), 5.20 (dt, J = 9.1, 4.5 Hz, 1H), 4.44-4.33 (m, 2H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 12.82 (s, 1H), 7.98 (d, J = 19.5 Hz, 2H), 7.83 (d, J = 7.8 Hz, 1H), 7.78-7.70 (m, 2H), 7.63 (s, 1H), 7.53 (dd, J = 13.2, 8.0 Hz, 3H), 7.46 (t, J = 7.7 Hz, 1H), 7.36 (dt, J = 21.6, 7.8 Hz, 4H), 7.27 (t, J = 7.4 Hz, 1H), 5.77 (d, J = 4.7 Hz, 1H), 5.20 (dt, J = 9.1, 4.5 Hz, 1H), 4.44-4.33 (m, 2H). 66 (S)-N-(5-카르바모일-1-(2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-2-일)-1H-인다졸-3-카르복사미드( S ) -N-(5-carbamoyl-1-(2-hydroxy-2-phenylethyl) -1H -benzo[ d ]imidazol-2-yl) -1H -indazole-3- carboxamide 1H NMR (400 MHz, DMSO-d 6 ) δ 8.17 (d, J = 8.2 Hz, 1H), 7.74 (s, 1H), 7.67-7.60 (m, 4H), 7.38 (ddt, J = 35.7, 19.9, 7.7 Hz, 8H), 7.00 (s, 1H), 6.66 (s, 2H), 6.47 (dd, J = 9.0, 4.5 Hz, 1H), 4.81 (dd, J = 15.3, 9.0 Hz, 1H), 4.52 (dd, J = 15.3, 4.5 Hz, 1H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.17 (d, J = 8.2 Hz, 1H), 7.74 (s, 1H), 7.67-7.60 (m, 4H), 7.38 (ddt, J = 35.7, 19.9) , 7.7 Hz, 8H), 7.00 (s, 1H), 6.66 (s, 2H), 6.47 (dd, J = 9.0, 4.5 Hz, 1H), 4.81 (dd, J = 15.3, 9.0 Hz, 1H), 4.52 (dd, J = 15.3, 4.5 Hz, 1H). 77 N-(5-카르바모일-1-((S)-2-히드록시-2-페닐에틸)-1H-벤조[d]이미다졸-2-일)-1-(테트라히드로-2H-피란-2-일)- 1H-인다졸-3-카르복사미드 N- (5-carbamoyl-1-(( S )-2-hydroxy-2-phenylethyl)-1H-benzo[ d ]imidazol-2-yl) -1- (tetrahydro- 2H -pyran-2-yl)- 1 H -indazole-3-carboxamide 1H NMR (400 MHz, DMSO-d 6 ) δ 8.17 (ddd, J = 16.2, 8.2, 3.1 Hz, 1H), 7.84 (dd, J = 8.5, 3.1 Hz, 1H), 7.74 (s, 1H), 7.64 (d, J = 8.8 Hz, 3H), 7.51 (t, J = 8.2 Hz, 1H), 7.37 (ddd, J = 19.5, 7.8, 3.3 Hz, 6H), 7.01 (s, 1H), 6.68 (s, 2H), 6.46 (tt, J = 7.9, 3.7 Hz, 1H), 5.96 (d, J = 9.3 Hz, 1H), 4.81 (dq, J = 13.6, 4.2 Hz, 1H), 4.53 (dt, J = 15.4, 3.8 Hz, 1H), 3.91-3.82 (m, 1H), 3.79-3.70 (m, 1H), 2.43-2.28 (m, 1H), 2.11-1.92 (m, 2H), 1.74 (d, J = 12.0 Hz, 1H), 1.62 (d, J = 17.3 Hz, 2H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.17 (ddd, J = 16.2, 8.2, 3.1 Hz, 1H), 7.84 (dd, J = 8.5, 3.1 Hz, 1H), 7.74 (s, 1H), 7.64 (d, J = 8.8 Hz, 3H), 7.51 (t, J = 8.2 Hz, 1H), 7.37 (ddd, J = 19.5, 7.8, 3.3 Hz, 6H), 7.01 (s, 1H), 6.68 (s) , 2H), 6.46 (tt, J = 7.9, 3.7 Hz, 1H), 5.96 (d, J = 9.3 Hz, 1H), 4.81 (dq, J = 13.6, 4.2 Hz, 1H), 4.53 (dt, J = 15.4, 3.8 Hz, 1H), 3.91-3.82 (m, 1H), 3.79-3.70 (m, 1H), 2.43-2.28 (m, 1H), 2.11-1.92 (m, 2H), 1.74 (d, J = 12.0 Hz, 1H), 1.62 (d, J = 17.3 Hz, 2H). 88 tert-부틸 (4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-1- yl) butyl) carbamate 1H NMR (300 MHz, DMSO-d 6) δ 12.82 (s, 1H), 8.03-7.93 (m, 2H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.32 (s, 1H), 6.79 (d, J = 5.9 Hz, 1H), 6.66 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 7.0 Hz, 2H), 2.96 (d, J = 6.6 Hz, 2H), 2.17 (s, 3H), 1.81-1.69 (m, 2H), 1.48-1.27 (m, 14H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.82 (s, 1H), 8.03-7.93 (m, 2H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.56 (d, J = 8.4) Hz, 1H), 7.32 (s, 1H), 6.79 (d, J = 5.9 Hz, 1H), 6.66 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 7.0) Hz, 2H), 2.96 (d, J = 6.6 Hz, 2H), 2.17 (s, 3H), 1.81-1.69 (m, 2H), 1.48-1.27 (m, 14H). 99 tert-부틸 (4-(5-카르바모일-2-(1-에틸-3-(트리플루오로메틸)-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(5-carbamoyl-2-(1-ethyl-3-(trifluoromethyl) -1H -pyrazole-5-carboxamido) -1H -benzo[ d ] imidazol-1-yl)butyl)carbamate 1H NMR (500 MHz, DMSO-d 6 ) δ 12.77 (s, 1H), 8.02 (s, 1H), 7.97 (s, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.31 (d, J = 12.9 Hz, 2H), 6.77 (t, J = 5.9 Hz, 1H), 4.34 (q, J = 7.2 Hz, 2H), 4.26 (d, J = 8.3 Hz, 2H), 2.99 (d, J = 6.4 Hz, 2H), 1.80-1.71 (m, 2H), 1.41 (dt, J = 23.7, 8.5 Hz, 5H), 1.33 (s, 9H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 12.77 (s, 1H), 8.02 (s, 1H), 7.97 (s, 1H), 7.79 (d, J = 8.4 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.31 (d, J = 12.9 Hz, 2H), 6.77 (t, J = 5.9 Hz, 1H), 4.34 (q, J = 7.2 Hz, 2H), 4.26 (d, J = 8.3 Hz, 2H), 2.99 (d, J = 6.4 Hz, 2H), 1.80-1.71 (m, 2H), 1.41 (dt, J = 23.7, 8.5 Hz, 5H), 1.33 (s, 9H). 1010 tert-부틸 (4-(5-카르바모일-2-(1-메틸-3-(트리플루오로메틸)-1H-피라졸-4-카르복스아미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(5-carbamoyl-2-(1-methyl-3-(trifluoromethyl) -1H -pyrazole-4-carboxamido) -1H -benzo[ d ] imidazol-1-yl)butyl)carbamate 1H NMR (500 MHz, DMSO-d 6 ) δ 12.78 (s, 1H), 8.44 (s, 1H), 8.01-7.92 (m, 2H), 7.77 (dd, J = 8.4, 1.7 Hz, 1H), 7.53 (d, J = 8.4 Hz, 1H), 7.31 (s, 1H), 6.80 (t, J = 5.6 Hz, 1H), 4.22 (t, J = 7.0 Hz, 2H), 3.95 (s, 3H), 2.98 (d, J = 6.4 Hz, 2H), 1.73 (p, J = 7.2 Hz, 2H), 1.42 (t, J = 7.5 Hz, 2H), 1.34 (s, 9H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 12.78 (s, 1H), 8.44 (s, 1H), 8.01-7.92 (m, 2H), 7.77 (dd, J = 8.4, 1.7 Hz, 1H), 7.53 (d, J = 8.4 Hz, 1H), 7.31 (s, 1H), 6.80 (t, J = 5.6 Hz, 1H), 4.22 (t, J = 7.0 Hz, 2H), 3.95 (s, 3H), 2.98 (d, J = 6.4 Hz, 2H), 1.73 (p, J = 7.2 Hz, 2H), 1.42 (t, J = 7.5 Hz, 2H), 1.34 (s, 9H). 1111 tert-부틸 (4-(2-(1-알릴-5-메틸-1H-피라졸-3-카르복스아미도)-5-카르바모일-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(2-(1-allyl-5-methyl- 1H -pyrazole-3-carboxamido)-5-carbamoyl- 1H -benzo[ d ]imidazole-1- yl) butyl) carbamate 1H NMR (500 MHz, DMSO-d 6 ) δ 8.03 (s, 1H), 7.95 (s, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.54 (s, 1H), 7.28 (s, 1H), 6.80 (d, J = 5.8 Hz, 1H), 6.63 (s, 1H), 6.00 (td, J = 12.2, 11.5, 5.9 Hz, 1H), 5.19 (d, J = 10.3 Hz, 1H), 4.94 (d, J = 17.1 Hz, 1H), 4.80 (s, 2H), 4.18 (s, 2H), 2.95 (s, 2H), 2.27 (s, 3H), 1.73 (s, 2H), 1.34 (s, 11H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 8.03 (s, 1H), 7.95 (s, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.54 (s, 1H), 7.28 (s, 1H), 6.80 (d, J = 5.8 Hz, 1H), 6.63 (s, 1H), 6.00 (td, J = 12.2, 11.5, 5.9 Hz, 1H), 5.19 (d, J = 10.3 Hz, 1H), 4.94 (d, J = 17.1 Hz, 1H), 4.80 (s, 2H), 4.18 (s, 2H), 2.95 (s, 2H), 2.27 (s, 3H), 1.73 (s, 2H), 1.34 (s) , 11H). 1212 tert-부틸 (4-(5-카르바모일-2-(푸란-3-카르복사미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(5-carbamoyl-2-(furan-3-carboxamido) -1H -benzo[ d ]imidazol-1-yl)butyl)carbamate 1H NMR (400 MHz, DMSO-d 6 ) δ 12.64 (s, 1H), 8.24 (s, 1H), 7.97 (d, J = 7.6 Hz, 2H), 7.77 (d, J = 8.3 Hz, 1H), 7.70 (s, 1H), 7.52 (d, J = 8.4 Hz, 1H), 7.30 (s, 1H), 6.81 (d, J = 5.7 Hz, 2H), 4.21 (d, J = 7.1 Hz, 2H), 2.99 (d, J = 6.2 Hz, 2H), 1.75 (t, J = 7.6 Hz, 2H), 1.43 (d, J = 7.6 Hz, 2H), 1.34 (s, 9H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.64 (s, 1H), 8.24 (s, 1H), 7.97 (d, J = 7.6 Hz, 2H), 7.77 (d, J = 8.3 Hz, 1H) , 7.70 (s, 1H), 7.52 (d, J = 8.4 Hz, 1H), 7.30 (s, 1H), 6.81 (d, J = 5.7 Hz, 2H), 4.21 (d, J = 7.1 Hz, 2H) , 2.99 (d, J = 6.2 Hz, 2H), 1.75 (t, J = 7.6 Hz, 2H), 1.43 (d, J = 7.6 Hz, 2H), 1.34 (s, 9H). 1313 tert-부틸 (4-(5-카르바모일-2-(푸란-3-카르복사미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(5-carbamoyl-2-(furan-3-carboxamido) -1H -benzo[ d ]imidazol-1-yl)butyl)carbamate 1H NMR (400 MHz, DMSO-d 6 ) δ 12.81 (s, 1H), 8.04 (s, 1H), 7.98 (s, 1H), 7.79 (dd, J = 18.9, 8.0 Hz, 2H), 7.72-7.55 (m, 3H), 7.43 (t, J = 7.7 Hz, 1H), 7.36-7.27 (m, 2H), 6.80 (t, J = 5.8 Hz, 1H), 4.30 (t, J = 6.9 Hz, 2H), 3.01 (q, J = 6.6 Hz, 2H), 1.81 (p, J = 7.6, 7.0 Hz, 2H), 1.46 (p, J = 7.0 Hz, 2H), 1.32 (s, 9H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.81 (s, 1H), 8.04 (s, 1H), 7.98 (s, 1H), 7.79 (dd, J = 18.9, 8.0 Hz, 2H), 7.72 7.55 (m, 3H), 7.43 (t, J = 7.7 Hz, 1H), 7.36-7.27 (m, 2H), 6.80 (t, J = 5.8 Hz, 1H), 4.30 (t, J = 6.9 Hz, 2H) ), 3.01 (q, J = 6.6 Hz, 2H), 1.81 (p, J = 7.6, 7.0 Hz, 2H), 1.46 (p, J = 7.0 Hz, 2H), 1.32 (s, 9H). 1414 tert-부틸 (4-(5-카르바모일-2-(1,3-디메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(5-carbamoyl-2-(1,3-dimethyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazol-1-yl) butyl) carbamate 1H NMR (400 MHz, DMSO-d 6 ) δ 12.80 (s, 1H), 7.98 (d, J = 12.1 Hz, 2H), 7.79 (d, J = 8.3 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.31 (s, 1H), 6.79 (t, J = 5.6 Hz, 1H), 6.66 (s, 1H), 4.22 (t, J = 6.9 Hz, 2H), 4.14 (s, 3H), 2.96 (q, J = 6.8 Hz, 2H), 2.17 (s, 3H), 1.75 (t, J = 7.7 Hz, 2H), 1.41 (t, J = 7.7 Hz, 2H), 1.34 (s, 9H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.80 (s, 1H), 7.98 (d, J = 12.1 Hz, 2H), 7.79 (d, J = 8.3 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.31 (s, 1H), 6.79 (t, J = 5.6 Hz, 1H), 6.66 (s, 1H), 4.22 (t, J = 6.9 Hz, 2H), 4.14 (s, 3H) , 2.96 (q, J = 6.8 Hz, 2H), 2.17 (s, 3H), 1.75 (t, J = 7.7 Hz, 2H), 1.41 (t, J = 7.7 Hz, 2H), 1.34 (s, 9H) . 1515 tert-부틸 (4-(5-카르바모일-2-(1-이소프로필-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -Butyl (4-(5-carbamoyl-2-(1-isopropyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-1 -yl)butyl)carbamate 1H NMR (500 MHz, DMSO-d 6 ) δ 12.81 (s, 1H), 7.98 (d, J = 9.2 Hz, 2H), 7.81-7.77 (m, 1H), 7.57 (d, J = 8.4 Hz, 1H), 7.32 (s, 1H), 6.80 (t, J = 5.9 Hz, 1H), 6.63 (s, 1H), 5.86 (p, J = 6.7 Hz, 1H), 4.20 (t, J = 7.0 Hz, 2H), 2.96 (q, J = 6.6 Hz, 2H), 2.19 (s, 3H), 1.75 (t, J = 7.7 Hz, 2H), 1.41 (d, J = 6.6 Hz, 8H), 1.34 (s, 9H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 12.81 (s, 1H), 7.98 (d, J = 9.2 Hz, 2H), 7.81-7.77 (m, 1H), 7.57 (d, J = 8.4 Hz, 1H), 7.32 (s, 1H), 6.80 (t, J = 5.9 Hz, 1H), 6.63 (s, 1H), 5.86 (p, J = 6.7 Hz, 1H), 4.20 (t, J = 7.0 Hz, 2H), 2.96 (q, J = 6.6 Hz, 2H), 2.19 (s, 3H), 1.75 (t, J = 7.7 Hz, 2H), 1.41 (d, J = 6.6 Hz, 8H), 1.34 (s, 9H). 1616 tert-부틸 (4-(2-(1-(tert-부틸)-3-메틸-1H-피라졸-5-카르복스아미도)-5-카르바모일-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 tert -butyl (4-(2-(1-( tert -butyl)-3-methyl- 1H -pyrazole-5-carboxamido)-5-carbamoyl- 1H -benzo[ d ]imi Dazol-1-yl)butyl)carbamate 1H NMR (400 MHz, DMSO-d 6 ) δ 12.78 (s, 1H), 8.05-7.99 (m, 1H), 7.96 (s, 1H), 7.80 (dd, J = 8.4, 1.6 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.32 (s, 1H), 6.78 (t, J = 5.8 Hz, 1H), 6.61 (s, 1H), 4.19 (t, J = 7.0 Hz, 2H), 2.95 (q, J = 6.6 Hz, 2H), 2.15 (s, 3H), 1.69 (s, 11H), 1.40 (t, J = 7.7 Hz, 2H), 1.34 (s, 9H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.78 (s, 1H), 8.05-7.99 (m, 1H), 7.96 (s, 1H), 7.80 (dd, J = 8.4, 1.6 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.32 (s, 1H), 6.78 (t, J = 5.8 Hz, 1H), 6.61 (s, 1H), 4.19 (t, J = 7.0 Hz, 2H), 2.95 (q, J = 6.6 Hz, 2H), 2.15 (s, 3H), 1.69 (s, 11H), 1.40 (t, J = 7.7 Hz, 2H), 1.34 (s, 9H).

하기 표 4 화합물의 합성Synthesis of the compounds of Table 4 below

tert -부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1 H -벤조[ d ]이미다졸-1-일)프로필)카르바메이트 (화합물 17), 및 tert -Butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H - benzo[ d ]imidazole-1- yl)propyl)carbamate (Compound 17), and

2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1-(3-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)프로필)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 27) 2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-1-(3-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido ) do) propyl) -1H - benzo[ d ]imidazole-5-carboxamide (Compound 27)

Figure pat00019
Figure pat00019

단계 1: Step 1: terttert -부틸(4-((4-카바모일-2-니트로페닐)아미노)프로필)카바메이트-Butyl(4-((4-carbamoyl-2-nitrophenyl)amino)propyl)carbamate

Figure pat00020
Figure pat00020

N-boc-1,3-프로판디아민 (9.38 g, 49.8 mmol) 및 4-클로로-3-니트로벤즈아미드 (3.00 g, 15.0 mmol)을 DMSO (25 mL)에 용해시킨 뒤 K2CO3 (3.14 g, 18.0 mmol)를 첨가하였다. 반응물을 70 ℃에서 16시간동안 교반하였다. 교반 후 냉수로 고체를 여과하고 진공 하에 건조시켜 노란색 고체상의 tert-부틸(4-((4-카바모일-2-니트로페닐)아미노)프로필)카바메이트 (4.15 g, 12.3 mmol, 82% 수율)를 수득하였다. N -boc-1,3-propanediamine (9.38 g, 49.8 mmol) and 4-chloro-3-nitrobenzamide (3.00 g, 15.0 mmol) were dissolved in DMSO (25 mL) followed by K 2 CO 3 (3.14) g, 18.0 mmol) was added. The reaction was stirred at 70 °C for 16 h. After stirring, the solid was filtered with cold water and dried under vacuum. tert -butyl(4-((4-carbamoyl-2-nitrophenyl)amino)propyl)carbamate as a yellow solid (4.15 g, 12.3 mmol, 82% yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6) δ 8.65 (d, J = 2.2 Hz, 1H), 8.45 (t, J = 5.9 Hz, 1H), 7.99 (dd, J = 8.8, 2.3 Hz, 2H), 7.27 (s, 1H), 7.08 (d, J = 9.1 Hz, 1H), 6.92 (d, J = 6.1 Hz, 1H), 3.42 (q, J = 6.5 Hz, 2H), 3.01 (q, J = 6.4 Hz, 2H), 1.71 (t, J = 6.6 Hz, 2H), 1.37 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.65 (d, J = 2.2 Hz, 1H), 8.45 (t, J = 5.9 Hz, 1H), 7.99 (dd, J = 8.8, 2.3 Hz, 2H) , 7.27 (s, 1H), 7.08 (d, J = 9.1 Hz, 1H), 6.92 (d, J = 6.1 Hz, 1H), 3.42 (q, J = 6.5 Hz, 2H), 3.01 (q, J = 6.4 Hz, 2H), 1.71 (t, J = 6.6 Hz, 2H), 1.37 (s, 9H).

단계 2: Step 2: terttert -부틸(4-((2-아미노-4-카바모일페닐)아미노)프로필)카바메이트-Butyl(4-((2-amino-4-carbamoylphenyl)amino)propyl)carbamate

Figure pat00021
Figure pat00021

tert-부틸(4-((4-카바모일-2-니트로페닐)아미노)프로필)카바메이트 (2.00 g, 5.91 mmol)를 MeOH (118 mL)에 용해시킨 뒤 10% 습윤 Pd/C (63 mg, 0.59 mmol)을 첨가하고 수소 풍선 하에 실온에서 18시간 동안 교반 시켰다. 이 후 MeOH 400 mL로 세척하면서 셀라이트®를 통해 여과하였다. 생성물을 함유하는 여과물을 진공하에 농축시켜 보라색 액체로서 tert-부틸(4-((2-아미노-4-카바모일페닐)아미노)프로필)카바메이트(1.79 g, 5.80 mmol, 98% 수율)를 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl(4-((4-carbamoyl-2-nitrophenyl)amino)propyl)carbamate (2.00 g, 5.91 mmol) was dissolved in MeOH (118 mL) followed by 10% wet Pd/C (63 mg) , 0.59 mmol) and stirred for 18 h at room temperature under a hydrogen balloon. It was then filtered through Celite®, washing with 400 mL of MeOH. The filtrate containing the product was concentrated in vacuo to give tert -butyl(4-((2-amino-4-carbamoylphenyl)amino)propyl)carbamate (1.79 g, 5.80 mmol, 98% yield) as a purple liquid. obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6) δ 7.41 (s, 1H), 7.10 (d, J = 8.2 Hz, 2H), 6.88 (t, J = 5.7 Hz, 1H), 6.72 (s, 1H), 6.36 (d, J = 8.0 Hz, 1H), 4.88 (t, J = 5.4 Hz, 1H), 4.56 (s, 2H), 3.05 (dq, J = 12.9, 6.6 Hz, 4H), 1.70 (p, J = 6.9 Hz, 2H), 1.38 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 7.41 (s, 1H), 7.10 (d, J = 8.2 Hz, 2H), 6.88 (t, J = 5.7 Hz, 1H), 6.72 (s, 1H) , 6.36 (d, J = 8.0 Hz, 1H), 4.88 (t, J = 5.4 Hz, 1H), 4.56 (s, 2H), 3.05 (dq, J = 12.9, 6.6 Hz, 4H), 1.70 (p, J = 6.9 Hz, 2H), 1.38 (s, 9H).

단계 3: Step 3: terttert -부틸 (4-(2-아미노-5-카르바모일-1-Butyl (4-(2-amino-5-carbamoyl-1) HH -벤조[-benzo[ dd ]이미다졸-1-일)프로필)카르바메이트]Imidazol-1-yl)propyl)carbamate

Figure pat00022
Figure pat00022

tert-부틸(4-((2-아미노-4-카바모일페닐)아미노)프로필)카바메이트 (1.79 g, 5.80 mmol)를 MeOH (9.67 mL)에 용해시킨 뒤 시아노겐 브로마이드 (1.54 g, 14.5 mmol)를 첨가한 후, 60 ℃에서 3시간 동안 교반하였다. 이어서 생성물을 함유하는 잔여물을 진공하에 MeOH을 제거시켜 검은색 고체상의 시아노겐 브로마이드를 함유한 tert-부틸 (4-(2-아미노-5-카르바모일-1H-벤조[d]이미다졸-1-일)프로필)카르바메이트를 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl(4-((2-amino-4-carbamoylphenyl)amino)propyl)carbamate (1.79 g, 5.80 mmol) was dissolved in MeOH (9.67 mL) followed by cyanogen bromide (1.54 g, 14.5 mmol) ), and stirred at 60 °C for 3 hours. The residue containing the product was then removed from MeOH in vacuo to tert -butyl (4-(2-amino-5-carbamoyl- 1H -benzo[ d ]imidazole) containing cyanogen bromide as a black solid. -1-yl)propyl)carbamate was obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6) δ 12.85 (s, 1H), 8.76 (s, 2H), 8.06 (s, 1H), 7.89-7.81 (m, 2H), 7.58 (d, J = 8.3 Hz, 1H), 7.41 (s, 1H), 6.94 (t, J = 5.4 Hz, 1H), 4.14 (t, J = 7.2 Hz, 2H), 3.02 (q, J = 6.5 Hz, 2H), 1.83 (p, J = 7.3 Hz, 2H), 1.37 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.85 (s, 1H), 8.76 (s, 2H), 8.06 (s, 1H), 7.89-7.81 (m, 2H), 7.58 (d, J = 8.3) Hz, 1H), 7.41 (s, 1H), 6.94 (t, J = 5.4 Hz, 1H), 4.14 (t, J = 7.2 Hz, 2H), 3.02 (q, J = 6.5 Hz, 2H), 1.83 ( p, J = 7.3 Hz, 2H), 1.37 (s, 9H).

단계 4: tert -부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1 H -벤조[ d ]이미다졸-1-일)프로필)카르바메이트 (화합물 17) Step 4: tert -Butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H - benzo[ d ]imidazole ) -1-yl)propyl)carbamate (compound 17)

Figure pat00023
Figure pat00023

1-에틸-3-메틸-1H-피라졸-5-카복실산 (463 mg, 3.00 mmol), HATU (1.14 g, 3.00 mmol), HOBt (203 mg, 1.50 mmol) 및 TEA (1.05 mL, 7.50 mmol)를 DMF (38 mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, tert-부틸 (4-(2-아미노-5-카르바모일-1H-벤조[d]이미다졸-1-일)프로필)카르바메이트 (500 mg, 1.50 mmol)를 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 밝은 노란색 고체상의 tert-부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)프로필)카르바메이트 (456 mg, 0.975 mmol, 65% 수율)를 수득하였다.1-Ethyl-3-methyl-1 H -pyrazole-5-carboxylic acid (463 mg, 3.00 mmol), HATU (1.14 g, 3.00 mmol), HOBt (203 mg, 1.50 mmol) and TEA (1.05 mL, 7.50 mmol) ) was dissolved in DMF (38 mL). After the reaction was stirred at room temperature for 5 min, tert -butyl (4-(2-amino-5-carbamoyl-1 H -benzo[ d ]imidazol-1-yl)propyl)carbamate (500 mg , 1.50 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) as a light yellow solid tert -butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl)) -1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazol-1-yl)propyl)carbamate (456 mg, 0.975 mmol, 65% yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6) δ 12.83 (s, 1H), 8.02-7.93 (m, 2H), 7.80 (dd, J = 8.4, 1.7 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.33 (s, 1H), 6.91 (t, J = 5.6 Hz, 1H), 6.68 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 7.1 Hz, 2H), 3.00 (q, J = 6.6 Hz, 2H), 2.18 (s, 3H), 1.90 (q, J = 7.0 Hz, 2H), 1.36 (s, 12H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.83 (s, 1H), 8.02-7.93 (m, 2H), 7.80 (dd, J = 8.4, 1.7 Hz, 1H), 7.56 (d, J = 8.4) Hz, 1H), 7.33 (s, 1H), 6.91 (t, J = 5.6 Hz, 1H), 6.68 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 7.1) Hz, 2H), 3.00 (q, J = 6.6 Hz, 2H), 2.18 (s, 3H), 1.90 (q, J = 7.0 Hz, 2H), 1.36 (s, 12H).

단계 5: 1-(3-아미노프로필)-2-(1-에틸-3-메틸-1Step 5: 1-(3-Aminopropyl)-2-(1-ethyl-3-methyl-1 HH -피라졸-5-카르복스아미도)-1-pyrazole-5-carboxamido)-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복스아미드 염산염)]Imidazole-5-carboxamide hydrochloride)

Figure pat00024
Figure pat00024

tert-부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)프로필)카르바메이트 (화합물 17, 200 mg, 0.426 mmol)에 MeOH (0.71 mL)을 용해 시킨 뒤 디옥산 (2.00 mL) 중 4 M HCl을 천천히 첨가하였다. 혼합물을 실온에서 1시간 동안 교반한 후, 생성된 고체를 여과 시켜준 뒤, Et2O로 3회 세척하고, 진공 하에 건조시켜 백색 고체상의 1-(3-아미노프로필)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복스아미드 염산염)을 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-1- MeOH (0.71 mL) was dissolved in yl)propyl)carbamate (compound 17, 200 mg, 0.426 mmol) and then 4M HCl in dioxane (2.00 mL) was slowly added. After the mixture was stirred at room temperature for 1 hour, the resulting solid was filtered, washed 3 times with Et 2 O, and dried under vacuum to form 1-(3-aminopropyl)-2-(1-(3-aminopropyl)-2-(1-) as a white solid. Ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide hydrochloride) was obtained and used in the next reaction without further purification. .

1 H NMR (500 MHz, DMSO-d 6) δ 7.87 (d, J = 1.7 Hz, 1H), 7.72 (dd, J = 8.5, 1.7 Hz, 1H), 7.50 (d, J = 8.5 Hz, 1H), 6.65 (s, 1H), 4.49 (q, J = 7.4 Hz, 2H), 4.22 (t, J = 6.8 Hz, 2H), 2.86 (t, J = 7.7 Hz, 2H), 2.12 (s, 3H), 2.05 (p, J = 7.1 Hz, 2H), 1.28 (t, J = 7.2 Hz, 3H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 7.87 (d, J = 1.7 Hz, 1H), 7.72 (dd, J = 8.5, 1.7 Hz, 1H), 7.50 (d, J = 8.5 Hz, 1H) , 6.65 (s, 1H), 4.49 (q, J = 7.4 Hz, 2H), 4.22 (t, J = 6.8 Hz, 2H), 2.86 (t, J = 7.7 Hz, 2H), 2.12 (s, 3H) , 2.05 (p, J = 7.1 Hz, 2H), 1.28 (t, J = 7.2 Hz, 3H).

단계 6: 2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1-(3-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)프로필)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 27) Step 6: 2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-1-(3-(1-ethyl-3-methyl-1 H -pyrazole-5- Carboxamido)propyl)-1H - benzo[ d ]imidazole-5-carboxamide (Compound 27)

Figure pat00025
Figure pat00025

1-에틸-3-메틸-1H-피라졸-5-카복실산 (37 mg, 0.243 mmol), HATU (123 mg, 0.243 mmol), HOBt (22 mg, 0.162 mmol) 및 TEA (0.226 mL, 1.62 mmol)를 DMF (4.1 mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, 1-(3-아미노프로필)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복스아미드 염산염) (60 mg, 0.162 mmol)을 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 흰색 고체상의 2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1-(3-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)프로필)-1H-벤조[d]이미다졸-5-카르복사미드 (62.5 mg, 0.123 mmol, 76% 수율)를 수득하였다.1-Ethyl-3-methyl-1 H -pyrazole-5-carboxylic acid (37 mg, 0.243 mmol), HATU (123 mg, 0.243 mmol), HOBt (22 mg, 0.162 mmol) and TEA (0.226 mL, 1.62 mmol) ) was dissolved in DMF (4.1 mL). The reaction was stirred at room temperature for 5 min, then 1-(3-aminopropyl)-2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-1 H -benzo[ d ]imidazole-5-carboxamide hydrochloride) (60 mg, 0.162 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) to 2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) as a white solid. -1-(3-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)propyl) -1H -benzo[ d ]imidazole-5-carboxamide (62.5 mg, 0.123 mmol, 76% yield).

1 H NMR (500 MHz, DMSO-d 6) δ 8.45 (t, J = 5.7 Hz, 1H), 8.01 (s, 2H), 7.81 (d, J = 8.4 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.35 (s, 1H), 6.58 (s, 1H), 6.55 (s, 1H), 4.58 (q, J = 7.1 Hz, 2H), 4.35 (q, J = 7.2 Hz, 2H), 4.27 (t, J = 7.2 Hz, 2H), 3.31-3.27 (m, 2H), 2.14 (s, 3H), 2.10 (s, 3H), 2.00 (q, J = 7.2 Hz, 2H), 1.33 (t, J = 7.1 Hz, 3H), 1.22 (t, J = 7.1 Hz, 3H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 8.45 (t, J = 5.7 Hz, 1H), 8.01 (s, 2H), 7.81 (d, J = 8.4 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.35 (s, 1H), 6.58 (s, 1H), 6.55 (s, 1H), 4.58 (q, J = 7.1 Hz, 2H), 4.35 (q, J = 7.2 Hz, 2H) , 4.27 (t, J = 7.2 Hz, 2H), 3.31-3.27 (m, 2H), 2.14 (s, 3H), 2.10 (s, 3H), 2.00 (q, J = 7.2 Hz, 2H), 1.33 ( t, J = 7.1 Hz, 3H), 1.22 (t, J = 7.1 Hz, 3H).

tert -부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1 H -벤조[ d ]이미다졸-1-일)펜틸)카르바메이트 (화합물 18), 및 tert -Butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H - benzo[ d ]imidazole-1- yl)pentyl)carbamate (Compound 18), and

2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1-(3-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)펜틸)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 28) 2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-1-(3-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido ) do) pentyl)-1H - benzo[ d ]imidazole-5-carboxamide (Compound 28)

Figure pat00026
Figure pat00026

단계 1: Step 1: terttert -부틸(4-((4-카바모일-2-니트로페닐)아미노)펜틸)카바메이트-Butyl(4-((4-carbamoyl-2-nitrophenyl)amino)pentyl)carbamate

Figure pat00027
Figure pat00027

N-boc-1,3-펜탄디아민 (3.64 g, 18.0 mmol) 및 4-클로로-3-니트로벤즈아미드 (6, 3.00 g, 15.0 mmol)을 DMSO (25 mL)에 용해시킨 뒤 K2CO3 (3.14 g, 18.0 mmol)를 첨가하였다. 반응물을 70 ℃에서 16시간동안 교반하였다. 교반 후 냉수로 고체를 여과하고 진공 하에 건조시켜 노란색 고체상의 tert-부틸(4-((4-카바모일-2-니트로페닐)아미노)펜틸)카바메이트 (3.20 g, 8.70 mmol, 58% 수율)를 수득하였다. Dissolve N -boc-1,3-pentanediamine (3.64 g, 18.0 mmol) and 4-chloro-3-nitrobenzamide (6, 3.00 g, 15.0 mmol) in DMSO (25 mL) followed by K 2 CO 3 (3.14 g, 18.0 mmol) was added. The reaction was stirred at 70 °C for 16 h. After stirring, the solid was filtered with cold water and dried under vacuum. tert -butyl(4-((4-carbamoyl-2-nitrophenyl)amino)pentyl)carbamate as a yellow solid (3.20 g, 8.70 mmol, 58% yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6) δ 8.65 (d, J = 2.1 Hz, 1H), 8.36 (t, J = 5.7 Hz, 1H), 8.00 (dd, J = 9.1, 2.2 Hz, 2H), 7.26 (s, 1H), 7.09 (d, J = 9.1 Hz, 1H), 6.77 (d, J = 6.0 Hz, 1H), 3.44-3.36 (m, 2H), 2.91 (q, J = 6.4 Hz, 2H), 1.62 (p, J = 7.5 Hz, 2H), 1.36 (s, 13H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.65 (d, J = 2.1 Hz, 1H), 8.36 (t, J = 5.7 Hz, 1H), 8.00 (dd, J = 9.1, 2.2 Hz, 2H) , 7.26 (s, 1H), 7.09 (d, J = 9.1 Hz, 1H), 6.77 (d, J = 6.0 Hz, 1H), 3.44-3.36 (m, 2H), 2.91 (q, J = 6.4 Hz, 2H), 1.62 (p, J = 7.5 Hz, 2H), 1.36 (s, 13H).

단계 2: Step 2: terttert -부틸(4-((2-아미노-4-카바모일페닐)아미노)펜틸)카바메이트-Butyl(4-((2-amino-4-carbamoylphenyl)amino)pentyl)carbamate

Figure pat00028
Figure pat00028

tert-부틸(4-((4-카바모일-2-니트로페닐)아미노)펜틸)카바메이트 (2.00 g, 5.46 mmol)를 MeOH (109 mL)에 용해시킨 뒤 10% 습윤 Pd/C (59 mg, 0.55 mmol)을 첨가하고 수소 풍선 하에 실온에서 18시간 동안 교반 시켰다. 이 후 MeOH 400 mL로 세척하면서 셀라이트®를 통해 여과하였다. 생성물을 함유하는 여과물을 진공하에 농축시켜 보라색 액체로서 tert-부틸(4-((2-아미노-4-카바모일페닐)아미노)펜틸)카바메이트 (1.70 g, 5.08 mmol, 93% 수율)를 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl(4-((4-carbamoyl-2-nitrophenyl)amino)pentyl)carbamate (2.00 g, 5.46 mmol) was dissolved in MeOH (109 mL) followed by 10% wet Pd/C (59 mg , 0.55 mmol) and stirred for 18 h at room temperature under a hydrogen balloon. It was then filtered through Celite®, washing with 400 mL of MeOH. The filtrate containing the product was concentrated in vacuo to give tert -butyl(4-((2-amino-4-carbamoylphenyl)amino)pentyl)carbamate (1.70 g, 5.08 mmol, 93% yield) as a purple liquid. obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6) δ 7.40 (s, 1H), 7.13-7.04 (m, 2H), 6.77 (d, J = 6.1 Hz, 1H), 6.71 (s, 1H), 6.35 (d, J = 8.1 Hz, 1H), 4.84 (t, J = 5.3 Hz, 1H), 4.58 (s, 2H), 3.04 (q, J = 6.5 Hz, 2H), 2.92 (q, J = 6.4 Hz, 2H), 1.58 (p, J = 7.3 Hz, 2H), 1.37 (s, 14H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 7.40 (s, 1H), 7.13-7.04 (m, 2H), 6.77 (d, J = 6.1 Hz, 1H), 6.71 (s, 1H), 6.35 ( d, J = 8.1 Hz, 1H), 4.84 (t, J = 5.3 Hz, 1H), 4.58 (s, 2H), 3.04 (q, J = 6.5 Hz, 2H), 2.92 (q, J = 6.4 Hz, 2H), 1.58 (p, J = 7.3 Hz, 2H), 1.37 (s, 14H).

단계 3: tert-부틸 (4-(2-아미노-5-카르바모일-1H-벤조[d]이미다졸-1-일)펜틸)카르바메이트 Step 3: tert -Butyl (4-(2-amino-5-carbamoyl- 1H -benzo[ d ]imidazol-1-yl)pentyl)carbamate

Figure pat00029
Figure pat00029

tert-부틸(4-((2-아미노-4-카바모일페닐)아미노)펜틸)카바메이트 (1.70 g, 5.05 mmol)를 MeOH (8.42 mL)에 용해시킨 뒤 시아노겐 브로마이드 (1.34 g, 12.0 mmol)를 첨가한 후, 60 ℃에서 3시간 동안 교반하였다. 이어서 생성물을 함유하는 잔여물을 진공하에 MeOH을 제거시켜 검은색 고체상의 시아노겐 브로마이드를 함유한 tert-부틸 (4-(2-아미노-5-카르바모일-1H-벤조[d]이미다졸-1-일)펜틸)카르바메이트를 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl(4-((2-amino-4-carbamoylphenyl)amino)pentyl)carbamate (1.70 g, 5.05 mmol) was dissolved in MeOH (8.42 mL) followed by cyanogen bromide (1.34 g, 12.0 mmol) ), and stirred at 60 °C for 3 hours. The residue containing the product was then removed from MeOH in vacuo to tert -butyl (4-(2-amino-5-carbamoyl- 1H -benzo[ d ]imidazole) containing cyanogen bromide as a black solid. -1-yl)pentyl)carbamate was obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6) δ 12.80 (s, 1H), 8.73 (s, 2H), 8.06 (s, 1H), 7.87-7.80 (m, 2H), 7.61 (d, J = 8.4 Hz, 1H), 7.40 (s, 1H), 6.77 (t, J = 5.7 Hz, 1H), 4.12 (t, J = 7.4 Hz, 2H), 2.89 (q, J = 6.4 Hz, 2H), 1.67 (t, J = 7.5 Hz, 2H), 1.35 (s, 13H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.80 (s, 1H), 8.73 (s, 2H), 8.06 (s, 1H), 7.87-7.80 (m, 2H), 7.61 (d, J = 8.4) Hz, 1H), 7.40 (s, 1H), 6.77 (t, J = 5.7 Hz, 1H), 4.12 (t, J = 7.4 Hz, 2H), 2.89 (q, J = 6.4 Hz, 2H), 1.67 ( t, J = 7.5 Hz, 2H), 1.35 (s, 13H).

단계 4: tert -부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1 H -벤조[ d ]이미다졸-1-일)펜틸)카르바메이트 (화합물 18) Step 4: tert -Butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H - benzo[ d ]imidazole ) -1-yl)pentyl)carbamate (Compound 18)

Figure pat00030
Figure pat00030

1-에틸-3-메틸-1H-피라졸-5-카복실산 (426 mg, 2.76 mmol), HATU (1.05 g, 2.76 mmol), HOBt (186 mg, 1.38 mmol) 및 TEA (0.962 mL, 6.90 mmol)를 DMF (34.5 mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, tert-부틸 (4-(2-아미노-5-카르바모일-1H-벤조[d]이미다졸-1-일)펜틸)카르바메이트 (500 mg, 1.50 mmol)를 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 밝은 노란색 고체상의 tert-부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)펜틸)카르바메이트 (423 mg, 0.856 mmol, 62% 수율)를 수득하였다.1-Ethyl-3-methyl-1 H -pyrazole-5-carboxylic acid (426 mg, 2.76 mmol), HATU (1.05 g, 2.76 mmol), HOBt (186 mg, 1.38 mmol) and TEA (0.962 mL, 6.90 mmol) ) was dissolved in DMF (34.5 mL). After the reaction was stirred at room temperature for 5 min, tert -butyl (4-(2-amino-5-carbamoyl-1 H -benzo[ d ]imidazol-1-yl)pentyl)carbamate (500 mg , 1.50 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) as a light yellow solid tert -butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl)) -1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazol-1-yl)pentyl)carbamate (423 mg, 0.856 mmol, 62% yield) was obtained.

1 H NMR (400 MHz, DMSO-d 6 ) δ 12.81 (s, 1H), 7.99 (d, J = 8.1 Hz, 2H), 7.80 (d, J = 8.2 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.32 (s, 1H), 6.73 (d, J = 5.8 Hz, 1H), 6.64 (s, 1H), 4.63 (q, J = 7.2 Hz, 2H), 4.19 (t, J = 7.2 Hz, 2H), 2.89 (q, J = 6.5 Hz, 2H), 2.18 (s, 3H), 1.76 (p, J = 7.4 Hz, 2H), 1.46-1.27 (m, 16H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.81 (s, 1H), 7.99 (d, J = 8.1 Hz, 2H), 7.80 (d, J = 8.2 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.32 (s, 1H), 6.73 (d, J = 5.8 Hz, 1H), 6.64 (s, 1H), 4.63 (q, J = 7.2 Hz, 2H), 4.19 (t, J = 7.2 Hz, 2H), 2.89 (q, J = 6.5 Hz, 2H), 2.18 (s, 3H), 1.76 (p, J = 7.4 Hz, 2H), 1.46-1.27 (m, 16H).

단계 5: 1-(3-아미노펜틸)-2-(1-에틸-3-메틸-1Step 5: 1-(3-Aminopentyl)-2-(1-ethyl-3-methyl-1 HH -피라졸-5-카르복스아미도)-1-pyrazole-5-carboxamido)-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복스아미드 염산염)]Imidazole-5-carboxamide hydrochloride)

Figure pat00031
Figure pat00031

tert-부틸 (3-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)펜틸)카르바메이트 (화합물 18, 200 mg, 0.402 mmol)에 MeOH (0.67 mL)을 용해 시킨 뒤 디옥산 (2.00 mL) 중 4 M HCl을 천천히 첨가하였다. 혼합물을 실온에서 1시간 동안 교반한 후, 생성된 고체를 여과 시켜준 뒤, Et2O로 3회 세척하고, 진공 하에 건조시켜 백색 고체상의 1-(3-아미노펜틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복스아미드 염산염)을 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl (3-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-1- MeOH (0.67 mL) was dissolved in yl)pentyl)carbamate (compound 18, 200 mg, 0.402 mmol) and then 4M HCl in dioxane (2.00 mL) was slowly added. After the mixture was stirred at room temperature for 1 hour, the resulting solid was filtered, washed 3 times with Et 2 O, and dried under vacuum to form 1-(3-aminopentyl)-2-(1-(3-aminopentyl)-2-(1-) as a white solid. Ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide hydrochloride) was obtained and used in the next reaction without further purification. .

1 H NMR (500 MHz, D2O) δ 7.52 (s, 1H), 7.44 (d, J = 8.4 Hz, 1H), 7.23 (d, J = 8.5 Hz, 1H), 6.42 (s, 1H), 4.36 (q, J = 7.5 Hz, 2H), 3.86 (t, J = 7.8 Hz, 2H), 2.92 (t, J = 7.7 Hz, 2H), 2.13 (s, 3H), 1.62 (p, J = 8.0 Hz, 2H), 1.54 (q, J = 7.8 Hz, 2H), 1.33 (p, J = 7.9 Hz, 2H), 1.22 (t, J = 7.3 Hz, 3H). 1 H NMR (500 MHz, D 2 O) δ 7.52 (s, 1H), 7.44 (d, J = 8.4 Hz, 1H), 7.23 (d, J = 8.5 Hz, 1H), 6.42 (s, 1H), 4.36 (q, J = 7.5 Hz, 2H), 3.86 (t, J = 7.8 Hz, 2H), 2.92 (t, J = 7.7 Hz, 2H), 2.13 (s, 3H), 1.62 (p, J = 8.0) Hz, 2H), 1.54 (q, J = 7.8 Hz, 2H), 1.33 (p, J = 7.9 Hz, 2H), 1.22 (t, J = 7.3 Hz, 3H).

단계 6: 2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1-(3-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)펜틸)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 28) Step 6: 2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-1-(3-(1-ethyl-3-methyl-1 H -pyrazole-5- Carboxamido)pentyl)-1H - benzo[ d ]imidazole-5-carboxamide (Compound 28)

Figure pat00032
Figure pat00032

1-에틸-3-메틸-1H-피라졸-5-카복실산 (35 mg, 0.227 mmol), HATU (115 mg, 0.302 mmol), HOBt (20 mg, 0.151 mmol) 및 TEA (0.210 mL, 1.51 mmol)를 DMF (3.8 mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, 1-(3-아미노펜틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복스아미드 염산염) (60 mg, 0.151 mmol)을 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 흰색 고체상의 2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1-(3-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)펜틸)-1H-벤조[d]이미다졸-5-카르복사미드 (46.5 mg, 0.087 mmol, 58% 수율)를 수득하였다.1-Ethyl-3-methyl-1 H -pyrazole-5-carboxylic acid (35 mg, 0.227 mmol), HATU (115 mg, 0.302 mmol), HOBt (20 mg, 0.151 mmol) and TEA (0.210 mL, 1.51 mmol) ) was dissolved in DMF (3.8 mL). After the reaction was stirred at room temperature for 5 min, 1-(3-aminopentyl)-2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-1 H -benzo[ d ]imidazole-5-carboxamide hydrochloride) (60 mg, 0.151 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) to 2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) as a white solid. -1-(3-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)pentyl) -1H -benzo[ d ]imidazole-5-carboxamide (46.5 mg, 0.087 mmol, 58% yield).

1 H NMR (400 MHz, DMSO-d 6) δ 12.82 (s, 1H), 8.30 (t, J = 5.7 Hz, 1H), 8.03-7.95 (m, 2H), 7.80 (dd, J = 8.4, 1.6 Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.33 (s, 1H), 6.63 (s, 1H), 6.48 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.35 (q, J = 7.1 Hz, 2H), 4.20 (t, J = 7.2 Hz, 2H), 3.18 (q, J = 6.6 Hz, 2H), 2.14 (d, J = 3.7 Hz, 6H), 1.80 (p, J = 7.3 Hz, 2H), 1.56 (p, J = 7.1 Hz, 2H), 1.35 (q, J = 7.1, 5.7 Hz, 5H), 1.22 (t, J = 7.1 Hz, 3H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.82 (s, 1H), 8.30 (t, J = 5.7 Hz, 1H), 8.03-7.95 (m, 2H), 7.80 (dd, J = 8.4, 1.6) Hz, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.33 (s, 1H), 6.63 (s, 1H), 6.48 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.35 (q, J = 7.1 Hz, 2H), 4.20 (t, J = 7.2 Hz, 2H), 3.18 (q, J = 6.6 Hz, 2H), 2.14 (d, J = 3.7 Hz, 6H), 1.80 ( p, J = 7.3 Hz, 2H), 1.56 (p, J = 7.1 Hz, 2H), 1.35 (q, J = 7.1, 5.7 Hz, 5H), 1.22 (t, J = 7.1 Hz, 3H).

1-(4-아미노부틸)-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복사미도)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 19) 1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H - benzo[ d ]imidazole-5-carboxamide (compound 19)

Figure pat00033
Figure pat00033

tert-부틸 (4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 (화합물8, 200 mg, 0.414 mmol)을 MeOH(0.69 mL)에 용해 시킨 후 디옥산 중 4 M HCl (4.00 mL)를 천천히 첨가하였다. 혼합물을 실온에서 1시간 동안 교반한 후, Et2O를 첨가하고 생성된 침전물을 Et2O로 여과하였다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH:aq.NH4OH(28%) = 9:1:0.1)로 정제하여 1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (55.3 mg, 0.144 mmol, 35%)를 분홍빛 고체로서 수득하였다. tert -Butyl (4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-1- yl)butyl)carbamate (compound 8, 200 mg, 0.414 mmol) was dissolved in MeOH (0.69 mL) and then 4 M HCl in dioxane (4.00 mL) was slowly added. After the mixture was stirred at room temperature for 1 h, Et 2 O was added and the resulting precipitate was filtered with Et 2 O. The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH:aq.NH 4 OH (28%) = 9:1:0.1) to 1-(4-aminobutyl)-2-(1-ethyl -3- Methyl - 1H -pyrazole-5-carboxamido)-1H-benzo[ d ]imidazole-5-carboxamide (55.3 mg, 0.144 mmol, 35%) was obtained as a pinkish solid.

1 H NMR (400 MHz, DMSO-d 6 ) δ 8.05-7.91 (m, 2H), 7.81-7.74 (m, 1H), 7.53 (d, J = 8.4 Hz, 1H), 7.31 (s, 1H), 6.63 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.20 (t, J = 7.1 Hz, 2H), 2.58 (t, J = 6.9 Hz, 2H), 2.17 (s, 3H), 1.79 (p, J = 6.9 Hz, 2H), 1.37 (dt, J = 14.1, 7.5 Hz, 5H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.05-7.91 (m, 2H), 7.81-7.74 (m, 1H), 7.53 (d, J = 8.4 Hz, 1H), 7.31 (s, 1H), 6.63 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.20 (t, J = 7.1 Hz, 2H), 2.58 (t, J = 6.9 Hz, 2H), 2.17 (s, 3H), 1.79 (p, J = 6.9 Hz, 2H), 1.37 (dt, J = 14.1, 7.5 Hz, 5H).

1-(4-아세트아미도부틸)-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복사미도)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 20) 1-(4-acetamidobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H - benzo[ d ]imidazole-5-carboxamide (Compound 20)

Figure pat00034
Figure pat00034

1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 19, 100 mg, 0.261 mmol)을 CH2Cl2:MeOH (9:1, 0.392 mL, 0.05 mL)혼합물에 용해시킨 뒤 0 ℃로 냉각시켜 주었다. 5분 후 acetyl chloride (0.028 mL, 0.392 mmol)를 0 ℃에서 천천히 첨가해주었다. 그 후 0 ℃에서 3시간동안 교반시켜준 뒤 실온에서 3시간 추가로 교반해주었다. 용매를 진공하에 제거시켜준 뒤 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 흰색 고체상의 1-(4-아세트아미도부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (35.4 mg, 0.0835 mmol, 32% 수율)를 수득하였다.1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 19, 100 mg, 0.261 mmol) was dissolved in a mixture of CH 2 Cl 2 :MeOH (9:1, 0.392 mL, 0.05 mL) and cooled to 0 °C. After 5 minutes, acetyl chloride (0.028 mL, 0.392 mmol) was slowly added at 0 °C. After that, the mixture was stirred at 0° C. for 3 hours and then stirred at room temperature for 3 hours. After the solvent was removed in vacuo, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) as a white solid 1-(4-acetamidobutyl)-2-(1-ethyl-3-methyl-1 H -Pyrazole-5-carboxamido)-1H-benzo[ d ]imidazole-5-carboxamide (35.4 mg, 0.0835 mmol, 32% yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6 ) δ 12.82 (s, 1H), 7.99 (d, J = 6.5 Hz, 2H), 7.86-7.76 (m, 2H), 7.57 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.66 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 6.9 Hz, 2H), 3.07 (q, J = 6.5 Hz, 2H), 2.18 (s, 3H), 1.75 (s, 5H), 1.49-1.33 (m, 5H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.82 (s, 1H), 7.99 (d, J = 6.5 Hz, 2H), 7.86-7.76 (m, 2H), 7.57 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.66 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 6.9 Hz, 2H), 3.07 (q, J = 6.5 Hz, 2H), 2.18 (s, 3H), 1.75 (s, 5H), 1.49-1.33 (m, 5H).

2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1-(4-피발아미도부틸)-1 H -벤조[ d ]이미다졸-5-카르복스아미드 (화합물 21) 2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1-(4-pivalamidobutyl)-1H - benzo[ d ]imidazole-5-carbox Amide (Compound 21)

Figure pat00035
Figure pat00035

1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 19, 100 mg, 0.261 mmol)을 CH2Cl2:MeOH (9:1, 0.392 mL, 0.05 mL)혼합물에 용해시킨 뒤 0 ℃로 냉각시켜 주었다. 5분 후 pivaloyl chloride (0.048 mL, 0.392 mmol)를 0 ℃에서 천천히 첨가해주었다. 그 후 0 ℃에서 3시간동안 교반시켜준 뒤 실온에서 3시간 추가로 교반해주었다. 용매를 진공하에 제거시켜준 뒤 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 흰색 고체상의 2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1-(4-피발아미도부틸)-1H-벤조[d]이미다졸-5-카르복스아미드 (55.7 mg, 0.120 mmol, 46% 수율)를 수득하였다.1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 19, 100 mg, 0.261 mmol) was dissolved in a mixture of CH 2 Cl 2 :MeOH (9:1, 0.392 mL, 0.05 mL) and cooled to 0 °C. After 5 minutes, pivaloyl chloride (0.048 mL, 0.392 mmol) was slowly added at 0 °C. After that, the mixture was stirred at 0° C. for 3 hours and then stirred at room temperature for 3 hours. After the solvent was removed in vacuo, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) to 2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) as a white solid. Obtained -1-(4- pivalamidobutyl )-1H-benzo[ d ]imidazole-5-carboxamide (55.7 mg, 0.120 mmol, 46% yield).

1 H NMR (400 MHz, DMSO-d 6) δ 12.80 (s, 1H), 8.03-7.93 (m, 2H), 7.80 (dd, J = 8.4, 1.7 Hz, 1H), 7.57 (d, J = 8.4 Hz, 1H), 7.42 (t, J = 5.8 Hz, 1H), 7.33 (s, 1H), 6.66 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 7.1 Hz, 2H), 3.09 (q, J = 6.4 Hz, 2H), 2.17 (s, 3H), 1.73 (p, J = 7.5 Hz, 2H), 1.46 (q, J = 7.1 Hz, 2H), 1.35 (t, J = 7.1 Hz, 3H), 1.02 (s, 9H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.80 (s, 1H), 8.03-7.93 (m, 2H), 7.80 (dd, J = 8.4, 1.7 Hz, 1H), 7.57 (d, J = 8.4) Hz, 1H), 7.42 (t, J = 5.8 Hz, 1H), 7.33 (s, 1H), 6.66 (s, 1H), 4.62 (q, J = 7.1 Hz, 2H), 4.21 (t, J = 7.1) Hz, 2H), 3.09 (q, J = 6.4 Hz, 2H), 2.17 (s, 3H), 1.73 (p, J = 7.5 Hz, 2H), 1.46 (q, J = 7.1 Hz, 2H), 1.35 ( t, J = 7.1 Hz, 3H), 1.02 (s, 9H).

1-(4-벤즈아미도부틸)-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복사미도)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 22) 1-(4-benzamidobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H - benzo[ d ]imidazole-5-carboxamide (Compound 22)

Figure pat00036
Figure pat00036

1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 19, 100 mg, 0.261 mmol)을 CH2Cl2:MeOH (9:1, 0.392 mL, 0.05 mL)혼합물에 용해시킨 뒤 0 ℃로 냉각시켜 주었다. 5분 후 benzoyl chloride (0.046 mL, 0.392 mmol)를 0 ℃에서 천천히 첨가해주었다. 그 후 0 ℃에서 3시간동안 교반시켜준 뒤 실온에서 3시간 추가로 교반해주었다. 용매를 진공하에 제거시켜준 뒤 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 흰색 고체상의 1-(4-벤즈아미도부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (57.2 mg, 0.117 mmol, 45% 수율)를 수득하였다.1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 19, 100 mg, 0.261 mmol) was dissolved in a mixture of CH 2 Cl 2 :MeOH (9:1, 0.392 mL, 0.05 mL) and cooled to 0 °C. After 5 minutes, benzoyl chloride (0.046 mL, 0.392 mmol) was slowly added at 0 °C. After that, the mixture was stirred at 0° C. for 3 hours and then stirred at room temperature for 3 hours. After the solvent was removed in vacuo, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) as a white solid 1-(4-benzamidobutyl)-2-(1-ethyl-3-methyl- 1H ) -Pyrazole -5-carboxamido)-1H-benzo[ d ]imidazole-5-carboxamide (57.2 mg, 0.117 mmol, 45% yield) was obtained.

1 H NMR (400 MHz, DMSO-d 6 ) δ 12.81 (s, 1H), 8.45 (t, J = 5.7 Hz, 1H), 8.03-7.94 (m, 2H), 7.81-7.75 (m, 3H), 7.59 (d, J = 8.4 Hz, 1H), 7.49 (dd, J = 8.4, 6.2 Hz, 1H), 7.41 (dd, J = 8.2, 6.6 Hz, 2H), 7.33 (s, 1H), 6.66 (s, 1H), 4.61 (q, J = 7.1 Hz, 2H), 4.25 (t, J = 7.0 Hz, 2H), 3.31 (t, J = 6.4 Hz, 2H), 2.15 (s, 3H), 1.83 (p, J = 7.2 Hz, 2H), 1.58 (dq, J = 13.7, 7.1 Hz, 2H), 1.34 (t, J = 7.1 Hz, 3H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.81 (s, 1H), 8.45 (t, J = 5.7 Hz, 1H), 8.03-7.94 (m, 2H), 7.81-7.75 (m, 3H), 7.59 (d, J = 8.4 Hz, 1H), 7.49 (dd, J = 8.4, 6.2 Hz, 1H), 7.41 (dd, J = 8.2, 6.6 Hz, 2H), 7.33 (s, 1H), 6.66 (s) , 1H), 4.61 (q, J = 7.1 Hz, 2H), 4.25 (t, J = 7.0 Hz, 2H), 3.31 (t, J = 6.4 Hz, 2H), 2.15 (s, 3H), 1.83 (p , J = 7.2 Hz, 2H), 1.58 (dq, J = 13.7, 7.1 Hz, 2H), 1.34 (t, J = 7.1 Hz, 3H).

2-(1-에틸-3-메틸-1 H -피라졸-5-카르복사미도)-1-(4-(니코틴아미도)부틸)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 23) 2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1-(4-(nicotinamido)butyl)-1H - benzo[ d ]imidazole-5-car Boxamide (Compound 23)

Figure pat00037
Figure pat00037

니코틴 산 (64 mg, 0.522 mmol), HATU (198 mg, 0.522 mmol), HOBt (35 mg, 0.261 mmol) 및 TEA (0.364 mL, 2.61 mmol)를 DMF (6.5 mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, 1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 19, 100 mg, 0.261 mmol)을 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 밝은 분홍색 고체상의 2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1-(4-(니코틴아미도)부틸)-1H-벤조[d]이미다졸-5-카르복사미드 (38.5 mg, 0.0783 mmol, 30% 수율)를 수득하였다.Nicotinic acid (64 mg, 0.522 mmol), HATU (198 mg, 0.522 mmol), HOBt (35 mg, 0.261 mmol) and TEA (0.364 mL, 2.61 mmol) were dissolved in DMF (6.5 mL). After the reaction was stirred at room temperature for 5 min, 1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 19, 100 mg, 0.261 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) to 2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido) as a light pink solid. Obtained -1-(4-(nicotinamido)butyl)-1H-benzo[ d ]imidazole-5-carboxamide (38.5 mg, 0.0783 mmol, 30% yield).

1 H NMR (400 MHz, DMSO-d 6 ) δ 12.82 (s, 1H), 8.97 (d, J = 2.3 Hz, 1H), 8.74 (t, J = 5.6 Hz, 1H), 8.66 (dd, J = 4.8, 1.7 Hz, 1H), 8.14 (dt, J = 8.0, 2.0 Hz, 1H), 8.05-7.95 (m, 2H), 7.81 (dd, J = 8.5, 1.7 Hz, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.45 (dd, J = 7.9, 4.8 Hz, 1H), 7.33 (s, 1H), 6.65 (s, 1H), 4.60 (q, J = 7.1 Hz, 2H), 4.26 (t, J = 7.0 Hz, 2H), 3.33 (t, J = 6.4 Hz, 2H), 2.14 (s, 3H), 1.84 (p, J = 7.1 Hz, 2H), 1.59 (p, J = 7.0 Hz, 2H), 1.34 (t, J = 7.1 Hz, 3H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.82 (s, 1H), 8.97 (d, J = 2.3 Hz, 1H), 8.74 (t, J = 5.6 Hz, 1H), 8.66 (dd, J = 4.8, 1.7 Hz, 1H), 8.14 (dt, J = 8.0, 2.0 Hz, 1H), 8.05-7.95 (m, 2H), 7.81 (dd, J = 8.5, 1.7 Hz, 1H), 7.60 (d, J ) = 8.4 Hz, 1H), 7.45 (dd, J = 7.9, 4.8 Hz, 1H), 7.33 (s, 1H), 6.65 (s, 1H), 4.60 (q, J = 7.1 Hz, 2H), 4.26 (t) , J = 7.0 Hz, 2H), 3.33 (t, J = 6.4 Hz, 2H), 2.14 (s, 3H), 1.84 (p, J = 7.1 Hz, 2H), 1.59 (p, J = 7.0 Hz, 2H) ), 1.34 (t, J = 7.1 Hz, 3H).

1-(4-(1 H -피라졸-5-카르복스아미도)부틸)-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1 H -벤조[ d ]이미다졸-5-카르복스아미드 (화합물 24) 1-(4-( 1H -Pyrazole-5-carboxamido)butyl)-2-(1-ethyl-3-methyl- 1H - pyrazole-5-carboxamido) -1H- Benzo[ d ]imidazole-5-carboxamide (Compound 24)

Figure pat00038
Figure pat00038

1H-피라졸-5-카르복실산 (59 mg, 0.522 mmol), HATU (198 mg, 0.522 mmol), HOBt (35 mg, 0.261 mmol) 및 TEA (0.364 mL, 2.61 mmol)를 DMF (6.5 mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, 1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 19, 100 mg, 0.261 mmol)을 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 밝은 분홍색 고체상의 1-(4-(1H-피라졸-5-카르복스아미도)부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복스아미드 (27.0 mg, 0.0574 mmol, 22% 수율)를 수득하였다.1 H -Pyrazole-5-carboxylic acid (59 mg, 0.522 mmol), HATU (198 mg, 0.522 mmol), HOBt (35 mg, 0.261 mmol) and TEA (0.364 mL, 2.61 mmol) were mixed with DMF (6.5 mL) ) was dissolved in After the reaction was stirred at room temperature for 5 min, 1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 19, 100 mg, 0.261 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) to 1-(4-(1 H -pyrazole-5-carboxamido)butyl)-2 as a light pink solid. -(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1H-benzo[ d ]imidazole-5-carboxamide (27.0 mg, 0.0574 mmol, 22% yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6 ) δ 13.25 (s, 1H), 12.83 (s, 1H), 8.20 (d, J = 6.0 Hz, 1H), 8.06-7.95 (m, 2H), 7.84-7.74 (m, 2H), 7.59 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.65 (s, 1H), 6.61 (s, 1H), 4.61 (q, J = 7.1 Hz, 2H), 4.24 (t, J = 6.9 Hz, 2H), 3.28 (q, J = 6.5 Hz, 2H), 2.16 (s, 3H), 1.86-1.72 (m, 2H), 1.56 (t, J = 7.6 Hz, 2H), 1.34 (t, J = 7.1 Hz, 3H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 13.25 (s, 1H), 12.83 (s, 1H), 8.20 (d, J = 6.0 Hz, 1H), 8.06-7.95 (m, 2H), 7.84 7.74 (m, 2H), 7.59 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.65 (s, 1H), 6.61 (s, 1H), 4.61 (q, J = 7.1 Hz, 2H) ), 4.24 (t, J = 6.9 Hz, 2H), 3.28 (q, J = 6.5 Hz, 2H), 2.16 (s, 3H), 1.86-1.72 (m, 2H), 1.56 (t, J = 7.6 Hz) , 2H), 1.34 (t, J = 7.1 Hz, 3H).

1-(4-(1,3-디메틸-1 H -피라졸-5-카르복스아미도)부틸)-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 25) 1-(4-(1,3-dimethyl- 1H -pyrazole-5-carboxamido)butyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido ) Figure)-1H - benzo[ d ]imidazole-5-carboxamide (compound 25)

Figure pat00039
Figure pat00039

1,3-디메틸-1H-피라졸-5-카르복실산 (73 mg, 0.522 mmol), HATU (198 mg, 0.522 mmol), HOBt (35 mg, 0.261 mmol) 및 TEA (0.364 mL, 2.61 mmol)를 DMF (6.5 mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, 1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 19, 100 mg, 0.261 mmol)을 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 밝은 분홍색 고체상의 1-(4-(1,3-디메틸-1H-피라졸-5-카르복스아미도)부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복사미드 (68.7 mg, 0.136 mmol, 52% 수율)를 수득하였다.1,3-dimethyl- 1H -pyrazole-5-carboxylic acid (73 mg, 0.522 mmol), HATU (198 mg, 0.522 mmol), HOBt (35 mg, 0.261 mmol) and TEA (0.364 mL, 2.61 mmol) ) was dissolved in DMF (6.5 mL). After the reaction was stirred at room temperature for 5 min, 1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 19, 100 mg, 0.261 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) as a light pink solid 1-(4-(1,3-dimethyl- 1H -pyrazole-5-carboxami). do)butyl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (68.7 mg, 0.136 mmol, 52% yield).

1 H NMR (300 MHz, DMSO-d 6 ) δ 12.82 (s, 1H), 8.34 (t, J = 5.7 Hz, 1H), 8.00 (t, J = 3.3 Hz, 2H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.33 (s, 1H), 6.66 (s, 1H), 6.49 (s, 1H), 4.61 (q, J = 7.1 Hz, 2H), 4.24 (t, J = 6.8 Hz, 2H), 3.92 (s, 3H), 3.25 (q, J = 6.6 Hz, 2H), 2.16 (s, 3H), 2.10 (s, 3H), 1.87-1.74 (m, 2H), 1.53 (dt, J = 13.3, 6.6 Hz, 2H), 1.34 (t, J = 7.1 Hz, 3H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.82 (s, 1H), 8.34 (t, J = 5.7 Hz, 1H), 8.00 (t, J = 3.3 Hz, 2H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.33 (s, 1H), 6.66 (s, 1H), 6.49 (s, 1H), 4.61 (q, J = 7.1 Hz, 2H), 4.24 (t, J = 6.8 Hz, 2H), 3.92 (s, 3H), 3.25 (q, J = 6.6 Hz, 2H), 2.16 (s, 3H), 2.10 (s, 3H), 1.87- 1.74 (m, 2H), 1.53 (dt, J = 13.3, 6.6 Hz, 2H), 1.34 (t, J = 7.1 Hz, 3H).

2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-1-(4-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)부틸)-1 H -벤조[ d ]이미다졸-5-카르복사미드 (화합물 26) 2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-1-(4-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido ) Do)Butyl)-1H - benzo[ d ]imidazole-5-carboxamide (Compound 26)

Figure pat00040
Figure pat00040

HATU (99 mg, 0.260 mmol), HOBt (18 mg, 0.130 mmol), TEA (0.181 mL, 1.30 mmol) 및 1-에틸-3-메틸-1H-피라졸-5-카르복실산 (40 mg, 0.260 mmol)을 DMF(3.3 mL, 0.04 M)에 용해시킨 뒤 실온에서 5분 동안 교반한 후, 1-(4-아미노부틸)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 19, 50 mg, 0.130 mmol)를 첨가하고 혼합물을 실온에서 16시간 동안 추가로 교반하였다. 그 다음, 반응 혼합물을 물에 붓고 EtOAc로 추출하였다. 유기층을 물로 세척하고 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH = 9:1)로 정제하여 2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1-(4-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)부틸)-1H-벤조[d]이미다졸-5-카르복사미드 (54.7 mg, 0.105 mmol, 81%)를 백색 고체로서 수득하였다.HATU (99 mg, 0.260 mmol), HOBt (18 mg, 0.130 mmol), TEA (0.181 mL, 1.30 mmol) and 1-ethyl-3-methyl-1 H -pyrazole-5-carboxylic acid (40 mg, 0.260 mmol) was dissolved in DMF (3.3 mL, 0.04 M) and stirred at room temperature for 5 minutes, followed by 1-(4-aminobutyl)-2-(1-ethyl-3-methyl- 1H -pyrazole -5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 19, 50 mg, 0.130 mmol) was added and the mixture was further stirred at room temperature for 16 h. Then, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was washed with water and dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH = 9:1) to 2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-1- (4-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)butyl) -1H -benzo[ d ]imidazole-5-carboxamide (54.7 mg, 0.105 mmol, 81%) as a white solid.

1 H NMR (400 MHz, DMSO-d 6) δ 12.84 (s, 1H), 8.34 (t, J = 5.8 Hz, 1H), 7.99 (d, J = 7.2 Hz, 2H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.66 (s, 1H), 6.46 (s, 1H), 4.61 (q, J = 7.1 Hz, 2H), 4.36 (q, J = 7.1 Hz, 2H), 4.25 (t, J = 6.9 Hz, 2H), 3.25 (q, J = 6.5 Hz, 2H), 2.16 (s, 3H), 2.11 (s, 3H), 1.81 (p, J = 7.1 Hz, 2H), 1.54 (p, J = 7.0 Hz, 2H), 1.35 (t, J = 7.1 Hz, 3H), 1.21 (t, J = 7.1 Hz, 3H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.84 (s, 1H), 8.34 (t, J = 5.8 Hz, 1H), 7.99 (d, J = 7.2 Hz, 2H), 7.79 (dd, J = 8.4, 1.6 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 6.66 (s, 1H), 6.46 (s, 1H), 4.61 (q, J = 7.1 Hz, 2H), 4.36 (q, J = 7.1 Hz, 2H), 4.25 (t, J = 6.9 Hz, 2H), 3.25 (q, J = 6.5 Hz, 2H), 2.16 (s, 3H), 2.11 (s, 3H), 1.81 (p, J = 7.1 Hz, 2H), 1.54 (p, J = 7.0 Hz, 2H), 1.35 (t, J = 7.1 Hz, 3H), 1.21 (t, J = 7.1 Hz, 3H) .

tert -부틸 (4-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-7-메톡시-1 H -벤조[ d ]이미다졸-1-일)부틸)카르바메이트 (화합물 29), 및 tert -Butyl (4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-7-methoxy- 1H -benzo[ d ] imidazol-1-yl)butyl)carbamate (Compound 29), and

tert -부틸 ( E )-(4-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복사미도)-7-메톡시-1 H -벤조[ d ]이미다졸-1-일)부트-2-엔-1-일)카바메이트 (화합물 30) tert -Butyl ( E )-(4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-7-methoxy- 1H -benzo [ d ]imidazol-1-yl)but-2-en-1-yl)carbamate (Compound 30)

Figure pat00041
Figure pat00041

Figure pat00042
Figure pat00042

단계 1: 4-클로로-3-메톡시-5-니트로벤즈아미드Step 1: 4-Chloro-3-methoxy-5-nitrobenzamide

Figure pat00043
Figure pat00043

메틸 4-클로로-3-메톡시-5-니트로벤조에이트 (3.00 g, 12.2 mmol)를 aq.NH4OH (28%) (40 mL)에 용해시킨 뒤 실온에서 16시간 동안 교반하였다. 교반 후 냉수로 고체를 여과하고, 진공 하에 건조시켜 황갈색 고체상의 4-클로로-3-메톡시5-니트로벤즈아미드 (2.26 g, 9.76 mmol, 80% 수율)를 수득하였다. Methyl 4-chloro-3-methoxy-5-nitrobenzoate (3.00 g, 12.2 mmol) was dissolved in aq.NH 4 OH (28%) (40 mL) and stirred at room temperature for 16 hours. After stirring, the solid was filtered with cold water and dried under vacuum to give 4-chloro-3-methoxy5-nitrobenzamide (2.26 g, 9.76 mmol, 80% yield) as a tan solid.

1 H NMR (300 MHz, Chloroform-d) δ 7.90 (d, J = 1.9 Hz, 1H), 7.76 (t, J = 1.8 Hz, 1H), 4.04 (d, J = 1.4 Hz, 3H). 1 H NMR (300 MHz, Chloroform-d) δ 7.90 (d, J = 1.9 Hz, 1H), 7.76 (t, J = 1.8 Hz, 1H), 4.04 (d, J = 1.4 Hz, 3H).

단계 2: Step 2: terttert -부틸 (-Butyl ( EE )-(4-((4-카르바모일-2-메톡시-6-니트로페닐)아미노)부트-2-엔-1-일)카르바메이트)-(4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-en-1-yl)carbamate

Figure pat00044
Figure pat00044

밀봉 튜브에 4-클로로-3-메톡시-5-니트로벤즈아미드 (1.00 g, 4.34 mmol)를 EtOH (4.34 mL)에 용해시킨 뒤 (E)-tert-부틸 (4-아미노부트-2-엔-1-일)카르바메이트 (0.970 g, 5.21 mmol) 및 DIPEA (2.26 mL, 13.0 mmol)를 첨가하였다. 반응물을 120 ℃에서 3일동안 교반하고, 실온으로 냉각시켰다. 생성된 오렌지색 고체를 EtOH로 여과한 뒤 건조시켜 주황색 고체상의 tert-부틸 (E)-(4-((4-카르바모일-2-메톡시-6-니트로 페닐)아미노)부트-2-엔-1-일)카르바메이트 (1.22 g, 3.21 mmol, 74% 수율)를 수득하였다. Dissolve 4-chloro-3-methoxy-5-nitrobenzamide (1.00 g, 4.34 mmol) in EtOH (4.34 mL) in a sealed tube ( E ) -tert -butyl (4-aminobut-2-ene) -1-yl)carbamate (0.970 g, 5.21 mmol) and DIPEA (2.26 mL, 13.0 mmol) were added. The reaction was stirred at 120 °C for 3 days and cooled to room temperature. The resulting orange solid was filtered with EtOH and dried to form an orange solid tert -butyl ( E )-(4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-ene Obtained -1-yl)carbamate (1.22 g, 3.21 mmol, 74% yield).

1 H NMR (400 MHz, methanol-d 4) δ 8.30 (d, J = 2.0 Hz, 1H), 7.54 (d, J = 2.0 Hz, 1H), 5.71-5.58 (m, 2H), 4.20 (d, J = 4.6 Hz, 2H), 3.93 (s, 3H), 3.60 (s, 2H), 1.41 (s, 9H). 1 H NMR (400 MHz, methanol- d 4 ) δ 8.30 (d, J = 2.0 Hz, 1H), 7.54 (d, J = 2.0 Hz, 1H), 5.71-5.58 (m, 2H), 4.20 (d, J = 4.6 Hz, 2H), 3.93 (s, 3H), 3.60 (s, 2H), 1.41 (s, 9H).

단계 3: Step 3: terttert -부틸 (-Butyl ( EE )-(4-((2-아미노-4-카르바모일-6-메톡시페닐)아미노)부트-2-엔-1-일)카르바메이트)-(4-((2-amino-4-carbamoyl-6-methoxyphenyl)amino)but-2-en-1-yl)carbamate

Figure pat00045
Figure pat00045

tert-부틸 (E)-(4-((4-카르바모일-2-메톡시-6-니트로페닐)아미노)부트-2-엔-1-일)카르바메이트 (500 mg, 1.31 mmol)을 MeOH (2.18 mL)에 용해시킨 뒤 aq.NH4OH (28%) (0.436 mL, 13.1 mmol) 및 1.0 M 농도의 아황산수소나트륨 수용액 (0.13 mL, 6.55 mmol)을 0 ℃에서 순서대로 첨가하였다. 반응물을 상온에서 2시간동안 교반한 뒤 진공하에 용매를 제거시켜주었다. 그 후 반응 혼합물에 물에 붓고 CH2Cl2로 추출하였다. 유기층을 Na2SO4로 건조시키고 걸러서 제거해준 뒤 걸러진 액체를 진공하에 제거시켜 tert-부틸 (E)-(4-((2-아미노-4-카르바모일-6-메톡시페닐)아미노)부트-2-엔-1-일)카르바메이트 (225 mg, 0.612 mmol, 49% 수율)을 수득하였다. tert -Butyl ( E )-(4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-en-1-yl)carbamate (500 mg, 1.31 mmol) was dissolved in MeOH (2.18 mL), and then aq.NH 4 OH (28%) (0.436 mL, 13.1 mmol) and a 1.0 M aqueous sodium hydrogen sulfite solution (0.13 mL, 6.55 mmol) were sequentially added at 0 °C. . The reaction mixture was stirred at room temperature for 2 hours, and then the solvent was removed under vacuum. Then, the reaction mixture was poured into water and extracted with CH 2 Cl 2 . The organic layer was dried over Na 2 SO 4 and filtered off, and the filtered liquid was removed under vacuum to remove tert -butyl ( E )-(4-((2-amino-4-carbamoyl-6-methoxyphenyl)amino) But-2-en-1-yl)carbamate (225 mg, 0.612 mmol, 49% yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6 ) δ 7.59 (s, 1H), 7.02-6.84 (m, 3H), 6.79 (d, J = 1.9 Hz, 1H), 5.67-5.47 (m, 2H), 4.65 (s, 2H), 3.81 (t, J = 7.1 Hz, 1H), 3.76 (s, 3H), 3.51 (q, J = 6.6 Hz, 4H), 1.37 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 7.59 (s, 1H), 7.02-6.84 (m, 3H), 6.79 (d, J = 1.9 Hz, 1H), 5.67-5.47 (m, 2H), 4.65 (s, 2H), 3.81 (t, J = 7.1 Hz, 1H), 3.76 (s, 3H), 3.51 (q, J = 6.6 Hz, 4H), 1.37 (s, 9H).

단계 4: (Step 4: ( EE )-2-아미노-1-(4-아미노부트-2-엔-1-일)-7-메톡시-1)-2-amino-1-(4-aminobut-2-en-1-yl)-7-methoxy-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복사미드]Imidazole-5-carboxamide

Figure pat00046
Figure pat00046

tert-부틸 (E)-(4-((2-아미노-4-카르바모일-6-메톡시페닐)아미노)부트-2-엔-1-일)카르바메이트 (100 mg, 0.285 mmol)를 MeOH (0.48 mL)에 용해시킨 뒤 시아노겐 브로마이드 (63 mg, 0.591 mmol)를 첨가한 후, 60 ℃에서 3시간 동안 교반하였다. 이어서 생성물을 함유하는 잔여물을 진공하에 MeOH을 제거시켜 흰색 고체상의 시아노겐 브로마이드를 함유한 (E)-2-아미노-1-(4-아미노부트-2-엔-1-일)-7-메톡시-1H-벤조[d]이미다졸-5-카르복사미드를 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl ( E )-(4-((2-amino-4-carbamoyl-6-methoxyphenyl)amino)but-2-en-1-yl)carbamate (100 mg, 0.285 mmol) was dissolved in MeOH (0.48 mL), and then cyanogen bromide (63 mg, 0.591 mmol) was added, followed by stirring at 60 °C for 3 hours. The residue containing the product was then removed in vacuo with MeOH to ( E )-2-amino-1-(4-aminobut-2-en-1-yl)-7- containing cyanogen bromide as a white solid. Methoxy- 1H -benzo[ d ]imidazole-5-carboxamide was obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6) δ 12.91 (s, 1H), 8.65 (s, 2H), 8.09 (s, 1H), 7.54-7.40 (m, 3H), 6.96 (d, J = 5.9 Hz, 1H), 5.66 (t, J = 4.0 Hz, 2H), 4.85 (d, J = 4.1 Hz, 2H), 3.95 (s, 3H), 3.52 (d, J = 5.9 Hz, 2H), 1.34 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.91 (s, 1H), 8.65 (s, 2H), 8.09 (s, 1H), 7.54-7.40 (m, 3H), 6.96 (d, J = 5.9) Hz, 1H), 5.66 (t, J = 4.0 Hz, 2H), 4.85 (d, J = 4.1 Hz, 2H), 3.95 (s, 3H), 3.52 (d, J = 5.9 Hz, 2H), 1.34 ( s, 9H).

단계 5: tert -부틸 ( E )-(4-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복사미도)-7-메톡시-1 H -벤조[ d ]이미다졸-1-일)부트-2-엔-1-일)카바메이트 (화합물 29) Step 5: tert -Butyl ( E )-(4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-7-methoxy-1 H -benzo[ d ]imidazol-1-yl)but-2-en-1-yl)carbamate (Compound 29)

Figure pat00047
Figure pat00047

(E)-2-아미노-1-(4-아미노부트-2-엔-1-일)-7-메톡시-1H-벤조[d]이미다졸-5-카르복사미드 (128 mg, 0.341 mmol), 1-에틸-3-메틸-1H-피라졸-5-카복실산 (63 mg, 0.409 mmol), HATU (648 mg, 1.71 mmol), 및 TEA (0.570 mL, 4.09 mmol)를 NMP (1.14 mL)에 용해시킨 뒤 80 ℃에서 72시간 동안 가열하였다. 반응물에 물 (30 mL)을 첨가하고 EtOAc (4 X 10 mL)로 추출하였다. 유기층을 무수 MgSO4을 첨가하여 건조시키고 진공하에 용매를 제거시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH=9:1)로 정제하여 보라색 고체상의 tert-부틸 (E)-(4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-7-메톡시-1H-벤조[d]이미다졸-1-일)부트-2-엔-1-일)카바메이트 (43 mg, 0.085 mmol, 25% 수율)를 수득하였다.( E )-2-amino-1-(4-aminobut-2-en-1-yl)-7-methoxy- 1H -benzo[ d ]imidazole-5-carboxamide (128 mg, 0.341 mmol), 1-ethyl-3-methyl-1 H -pyrazole-5-carboxylic acid (63 mg, 0.409 mmol), HATU (648 mg, 1.71 mmol), and TEA (0.570 mL, 4.09 mmol) with NMP (1.14) mL) and then heated at 80 °C for 72 hours. To the reaction was added water (30 mL) and extracted with EtOAc (4 X 10 mL). The organic layer was dried by the addition of anhydrous MgSO 4 and the solvent was removed in vacuo. The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH=9:1) as a purple solid tert -butyl ( E )-(4-(5-carbamoyl-2-(1-ethyl-) 3-methyl- 1H -pyrazole-5-carboxamido)-7-methoxy- 1H -benzo[ d ]imidazol-1-yl)but-2-en-1-yl)carbamate (43 mg, 0.085 mmol, 25% yield).

1 H NMR (300 MHz, methanol-d 4 ) δ 7.57 (s, 1H), 7.34 (d, J = 1.4 Hz, 1H), 6.64 (s, 1H), 5.84-5.58 (m, 2H), 4.95 (d, J = 5.7 Hz, 2H), 4.64 (q, J = 7.3 Hz, 2H), 3.98 (s, 3H), 3.58 (t, J = 4.8 Hz, 2H), 2.22 (s, 3H), 1.43-1.26 (m, 12H). 1 H NMR (300 MHz, methanol- d 4 ) δ 7.57 (s, 1H), 7.34 (d, J = 1.4 Hz, 1H), 6.64 (s, 1H), 5.84-5.58 (m, 2H), 4.95 ( d, J = 5.7 Hz, 2H), 4.64 (q, J = 7.3 Hz, 2H), 3.98 (s, 3H), 3.58 (t, J = 4.8 Hz, 2H), 2.22 (s, 3H), 1.43- 1.26 (m, 12H).

단계 6: tert -부틸 (4-(5-카르바모일-2-(1-에틸-3-메틸-1 H -피라졸-5-카르복스아미도)-7-메톡시-1 H -벤조[ d ]이미다졸-1-일)부틸)카르바메이트, (화합물 30) Step 6: tert -Butyl (4-(5-carbamoyl-2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido)-7-methoxy-1 H -benzo [ d ]imidazol-1-yl)butyl)carbamate, (compound 30)

Figure pat00048
Figure pat00048

tert-부틸 (E)-(4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-7-메톡시-1H-벤조[d]이미다졸-1-일)부트-2-엔-1-일)카바메이트 (화합물 29, 33 mg, 0.0645 mmol)를 MeOH (0.22 mL)에 용해시킨 뒤 10% 습윤 Pd/C (6.0 mg, 0.00645 mmol)을 첨가하고 수소 풍선 하에 실온에서 3시간 동안 교반 시켰다. 이 후 MeOH 20 mL로 세척하면서 셀라이트®를 통해 여과하였다. 생성물을 함유하는 여과물을 진공하에 농축시켜 보라색 액체로서 tert-부틸 (4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-7-메톡시-1H-벤조[d]이미다졸-1-일)부틸)카르바메이트 (31 mg, 0.059 mmol, 92% 수율)를 수득하였다. tert -Butyl ( E )-(4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-7-methoxy- 1H -benzo [ d ]Imidazol-1-yl)but-2-en-1-yl)carbamate (compound 29, 33 mg, 0.0645 mmol) was dissolved in MeOH (0.22 mL) followed by 10% wet Pd/C (6.0 mg, 0.00645 mmol) and stirred under a hydrogen balloon at room temperature for 3 hours. It was then filtered through Celite®, washing with 20 mL of MeOH. The filtrate containing the product was concentrated in vacuo as a purple liquid as tert -butyl (4-(5-carbamoyl-2-(1-ethyl-3-methyl-1 H -pyrazole-5-carboxamido) )-7-methoxy- 1H -benzo[ d ]imidazol-1-yl)butyl)carbamate (31 mg, 0.059 mmol, 92% yield) was obtained.

1 H NMR (400 MHz, methanol-d 4) δ 7.57 (s, 1H), 7.35 (s, 1H), 6.64 (s, 1H), 4.65 (q, J = 7.1 Hz, 2H), 4.35 (t, J = 7.2 Hz, 2H), 3.99 (s, 3H), 3.06 (t, J = 7.0 Hz, 2H), 2.22 (s, 3H), 1.79 (t, J = 7.7 Hz, 2H), 1.51 (t, J = 7.7 Hz, 2H), 1.43-1.35 (m, 12H). 1 H NMR (400 MHz, methanol- d 4 ) δ 7.57 (s, 1H), 7.35 (s, 1H), 6.64 (s, 1H), 4.65 (q, J = 7.1 Hz, 2H), 4.35 (t, J = 7.2 Hz, 2H), 3.99 (s, 3H), 3.06 (t, J = 7.0 Hz, 2H), 2.22 (s, 3H), 1.79 (t, J = 7.7 Hz, 2H), 1.51 (t, J = 7.7 Hz, 2H), 1.43-1.35 (m, 12H).

하기 표 5 화합물(이량체)들의 합성Synthesis of the compounds (dimers) in Table 5 below

Figure pat00049
Figure pat00049

단계 1: 4,4'-(부탄-1,4-디일비스(아잔디일))비스(3-니트로벤즈아미드)Step 1: 4,4'-(butane-1,4-diylbis(azanediyl))bis(3-nitrobenzamide)

Figure pat00050
Figure pat00050

부탄-1,4-디아민 (0.800 g, 9.00 mmol) 및 4-클로로-3-니트로벤즈아미드 (3.00 g, 15.00 mmol)를 DMSO (15 mL)에 용해시킨 뒤 K2CO3 (3.11 g, 22.5 mmol)를 첨가 하고 실온에서 16시간 동안 교반하였다. 16시간 동안 교반 후 고체를 냉수로 여과하고, 진공 하에 건조시켜 노란색 고체상의 4,4'-(부탄-1,4-디일비스(아잔디일))비스(3-니트로벤즈아미드) (4.21 g, 10.1 mmol, 67% 수율)를 수득하였다. Butane-1,4-diamine (0.800 g, 9.00 mmol) and 4-chloro-3-nitrobenzamide (3.00 g, 15.00 mmol) were dissolved in DMSO (15 mL) followed by K 2 CO 3 (3.11 g, 22.5) mmol) and stirred at room temperature for 16 hours. After stirring for 16 hours, the solid was filtered with cold water, dried under vacuum and 4,4'-(butane-1,4-diylbis(azanediyl))bis(3-nitrobenzamide) as a yellow solid (4.21 g) , 10.1 mmol, 67% yield).

1 H NMR (300 MHz, DMSO-d 6) δ 8.65 (d, J = 2.1 Hz, 1H), 8.43 (t, J = 5.8 Hz, 1H), 8.00 (dd, J = 9.7, 2.7 Hz, 2H), 7.30 (s, 1H), 7.13 (d, J = 9.2 Hz, 1H), 1.74 (d, J = 6.2 Hz, 2H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.65 (d, J = 2.1 Hz, 1H), 8.43 (t, J = 5.8 Hz, 1H), 8.00 (dd, J = 9.7, 2.7 Hz, 2H) , 7.30 (s, 1H), 7.13 (d, J = 9.2 Hz, 1H), 1.74 (d, J = 6.2 Hz, 2H).

단계 2: 4,4'-(부탄-1,4-디일비스(아잔디일))비스(3-아미노벤즈아미드)Step 2: 4,4'-(Butane-1,4-diylbis(azanediyl))bis(3-aminobenzamide)

Figure pat00051
Figure pat00051

4,4'-(부탄-1,4-디일비스(아잔디일))비스(3-니트로벤즈아미드) (1.00 g, 2.40 mmol)를 MeOH (40 mL)에 용해시킨 뒤 10% 습윤 Pd/C (26.0 mg, 0.240 mmol)를 첨가한 뒤 수소 풍선 하에 실온에서 18시간 동안 교반 시켰다. 이 후 진공하에 농축시켜 용매를 제거해주어 습윤 Pd/C가 포함된 녹색 고체상의 4,4'-(부탄-1,4-디일비스(아잔디일))비스(3-아미노벤즈아미드) (0.775 g, 2.17 mmol, 91% 수율)을 수득하였다. 4,4′-(butane-1,4-diylbis(azanediyl))bis(3-nitrobenzamide) (1.00 g, 2.40 mmol) was dissolved in MeOH (40 mL) followed by 10% wet Pd/ C (26.0 mg, 0.240 mmol) was added and stirred for 18 hours at room temperature under a hydrogen balloon. After concentration in vacuo to remove the solvent, 4,4'-(butane-1,4-diylbis(azanediyl))bis(3-aminobenzamide) (0.775) as a green solid containing wet Pd/C g, 2.17 mmol, 91% yield).

1 H NMR (300 MHz, DMSO-d 6) δ 7.41 (s, 1H), 7.13-7.07 (m, 2H), 6.71 (s, 1H), 6.39 (d, J = 8.1 Hz, 1H), 4.90 (t, J = 5.2 Hz, 1H), 4.59 (s, 2H), 3.13 (d, J = 5.2 Hz, 2H), 1.73 (s, 2H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 7.41 (s, 1H), 7.13-7.07 (m, 2H), 6.71 (s, 1H), 6.39 (d, J = 8.1 Hz, 1H), 4.90 ( t, J = 5.2 Hz, 1H), 4.59 (s, 2H), 3.13 (d, J = 5.2 Hz, 2H), 1.73 (s, 2H).

단계 3: 1,1'-(부탄-1,4-디일)비스(2-아미노-1Step 3: 1,1′-(butane-1,4-diyl)bis(2-amino-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복스아미드)]imidazole-5-carboxamide)

Figure pat00052
Figure pat00052

4,4'-(부탄-1,4-디일비스(아잔디일))비스(3-아미노벤즈아미드) (775 mg, 2.17 mmol)를 MeOH (5 mL)에 용해시킨 뒤 시아노겐 브로마이드 (575 mg, 5.43 mmol)를 첨가하고, 60 ℃에서 3시간 동안 교반하였다. 이어서 생성물을 함유하는 잔여물을 진공하에 MeOH을 제거시켜 황갈색 고체로서 시아노겐 브로마이드와 습윤 Pd/C를 함유한 녹색 고체상의 1,1'-(부탄-1,4-디일)비스(2-아미노-1H-벤조[d]이미다졸-5-카르복스아미드)을 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. 4,4'-(butane-1,4-diylbis(azanediyl))bis(3-aminobenzamide) (775 mg, 2.17 mmol) was dissolved in MeOH (5 mL) followed by cyanogen bromide (575 mg, 5.43 mmol) and stirred at 60 °C for 3 h. The residue containing the product was then removed in vacuo with MeOH removed as 1,1′-(butane-1,4-diyl)bis(2-amino) as a green solid containing cyanogen bromide and wet Pd/C as a tan solid. -1H -benzo[ d ]imidazole-5-carboxamide) was obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6) δ 12.80 (s, 1H), 8.76 (s, 2H), 8.06 (s, 1H), 7.88-7.80 (m, 2H), 7.63 (d, J = 8.4 Hz, 1H), 7.43 (s, 1H), 4.19 (s, 2H), 3.16 (s, 2H), 1.79 (s, 2H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.80 (s, 1H), 8.76 (s, 2H), 8.06 (s, 1H), 7.88-7.80 (m, 2H), 7.63 (d, J = 8.4) Hz, 1H), 7.43 (s, 1H), 4.19 (s, 2H), 3.16 (s, 2H), 1.79 (s, 2H).

단계 4: 1,1'-(부탄-1,4-디일)비스(2-산치환기아미도-1Step 4: 1,1'-(butane-1,4-diyl)bis(2-acid substituted groupamido-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복사미드)]imidazole-5-carboxamide)

Figure pat00053
Figure pat00053

1,1'-(부탄-1,4-디일)비스(2-아미노-1H-벤조[d]이미다졸-5-카르복스아미드) (200 mg, 0.492 mmol), 치환기카르복실산 (2.2 eq, 1.08 mmol), HATU (935 mg, 2.46 mmol), 및 트리에틸아민 (0.823 mL, 5.91 mmol)을 NMP (0.82 mL)에 융해시킨 뒤 80 ℃에서 48시간 동안 가열하였다. 반응물에 물 (30 mL)을 첨가하고 10분간 교반한 후 생성된 고체를 여과하고 CH2Cl2으로 헹구어 주었다. 걸러진 고체를 건조 로딩하고 실리카 겔 상에서 CH2Cl2 중 0 - 10% MeOH로 정제하여 목적화합물을 수득하였다.1,1′-(butane-1,4-diyl)bis(2-amino- 1H -benzo[ d ]imidazole-5-carboxamide) (200 mg, 0.492 mmol), substituted carboxylic acid (2.2 eq, 1.08 mmol), HATU (935 mg, 2.46 mmol), and triethylamine (0.823 mL, 5.91 mmol) were dissolved in NMP (0.82 mL) and heated at 80 °C for 48 hours. Water (30 mL) was added to the reaction mixture and stirred for 10 minutes. The resulting solid was filtered and washed with CH 2 Cl 2 . The filtered solid was dry-loaded and purified on silica gel with 0-10% MeOH in CH 2 Cl 2 to obtain the target compound.

화합물 31 내지 36의 NMR 측정 결과를 하기 표 2에 종합하여 나타내었다.The NMR measurement results of compounds 31 to 36 are summarized in Table 2 below.

Compound
No.
Compound
No.
IUPAC nameIUPAC name 1H NMR 1 H NMR
3131 1,1'-(부탄-1,4-디일)비스(2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복스아미드)1,1'-(butane-1,4-diyl)bis(2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole -5-carboxamide) 1H NMR (400 MHz, DMSO-d 6) δ 12.82 (s, 2H), 7.97 (s, 4H), 7.76 (d, J = 8.4 Hz, 2H), 7.54 (d, J = 8.4 Hz, 2H), 7.34 (s, 2H), 6.59 (s, 2H), 4.56 (q, J = 7.2 Hz, 4H), 4.27 (s, 4H), 2.09 (s, 6H), 1.87 (s, 4H), 1.30 (t, J = 7.1 Hz, 6H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.82 (s, 2H), 7.97 (s, 4H), 7.76 (d, J = 8.4 Hz, 2H), 7.54 (d, J = 8.4 Hz, 2H) , 7.34 (s, 2H), 6.59 (s, 2H), 4.56 (q, J = 7.2 Hz, 4H), 4.27 (s, 4H), 2.09 (s, 6H), 1.87 (s, 4H), 1.30 ( t, J = 7.1 Hz, 6H). 3232 1,1'-(부탄-1,4-디일)비스(2-(1-알릴-5-메틸-1H-피라졸-3-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드)1,1'-(butane-1,4-diyl)bis(2-(1-allyl-5-methyl- 1H -pyrazole-3-carboxamido) -1H -benzo[ d ]imidazole- 5-carboxamide) 1H NMR (300 MHz, DMSO-d 6) δ 12.66 (s, 2H), 7.97 (d, J = 17.9 Hz, 4H), 7.73 (d, J = 8.3 Hz, 2H), 7.51 (s, 2H), 7.29 (s, 2H), 6.58 (s, 2H), 5.93 (ddt, J = 15.7, 10.2, 5.1 Hz, 2H), 5.12 (d, J = 10.3 Hz, 2H), 4.87 (d, J = 17.2 Hz, 2H), 4.74 (s, 4H), 4.29 (s, 4H), 2.21 (s, 6H), 1.80 (s, 4H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.66 (s, 2H), 7.97 (d, J = 17.9 Hz, 4H), 7.73 (d, J = 8.3 Hz, 2H), 7.51 (s, 2H) , 7.29 (s, 2H), 6.58 (s, 2H), 5.93 (ddt, J = 15.7, 10.2, 5.1 Hz, 2H), 5.12 (d, J = 10.3 Hz, 2H), 4.87 (d, J = 17.2) Hz, 2H), 4.74 (s, 4H), 4.29 (s, 4H), 2.21 (s, 6H), 1.80 (s, 4H). 3333 1,1'-(부탄-1,4-디일)비스(2-(벤조푸란-2-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드)1,1'-(butane-1,4-diyl)bis(2-(benzofuran-2-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide) 1H NMR (300 MHz, DMSO-d 6) δ 12.83 (s, 2H), 7.99 (d, J = 16.2 Hz, 4H), 7.81-7.72 (m, 2H), 7.60 (dd, J = 17.3, 8.6 Hz, 8H), 7.41-7.18 (m, 6H), 4.47 (s, 4H), 2.04-1.80 (m, 4H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.83 (s, 2H), 7.99 (d, J = 16.2 Hz, 4H), 7.81-7.72 (m, 2H), 7.60 (dd, J = 17.3, 8.6) Hz, 8H), 7.41-7.18 (m, 6H), 4.47 (s, 4H), 2.04-1.80 (m, 4H). 3434 1,1'-(부탄-1,4-디일)비스(2-(1-메틸-3-(트리플루오로메틸)-1H-피라졸-4-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드)1,1′-(butane-1,4-diyl)bis(2-(1-methyl-3-(trifluoromethyl) -1H -pyrazole-4-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide) 1H NMR (500 MHz, DMSO-d 6) δ 12.78 (s, 2H), 8.36 (s, 2H), 7.94 (d, J = 11.8 Hz, 4H), 7.72 (d, J = 8.3 Hz, 2H), 7.49 (d, J = 8.4 Hz, 2H), 7.30 (s, 2H), 4.28 (s, 4H), 3.87 (s, 6H), 1.85 (s, 4H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 12.78 (s, 2H), 8.36 (s, 2H), 7.94 (d, J = 11.8 Hz, 4H), 7.72 (d, J = 8.3 Hz, 2H) , 7.49 (d, J = 8.4 Hz, 2H), 7.30 (s, 2H), 4.28 (s, 4H), 3.87 (s, 6H), 1.85 (s, 4H). 3535 디메틸 1,1'-(부탄-1,4-디일)비스(2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복실레이트)Dimethyl 1,1'-(butane-1,4-diyl)bis(2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole -5-carboxylate) 1H NMR (400 MHz, DMSO-d 6) δ 12.83 (s, 2H), 8.05 (d, J = 1.6 Hz, 2H), 7.80 (dd, J = 8.4, 1.6 Hz, 2H), 7.55 (d, J = 8.5 Hz, 2H), 6.57 (s, 2H), 4.55 (q, J = 7.1 Hz, 4H), 4.25 (s, 4H), 3.86 (s, 6H), 2.09 (s, 6H), 1.87 (s, 4H), 1.30 (t, J = 7.1 Hz, 6H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.83 (s, 2H), 8.05 (d, J = 1.6 Hz, 2H), 7.80 (dd, J = 8.4, 1.6 Hz, 2H), 7.55 (d, J = 8.5 Hz, 2H), 6.57 (s, 2H), 4.55 (q, J = 7.1 Hz, 4H), 4.25 (s, 4H), 3.86 (s, 6H), 2.09 (s, 6H), 1.87 ( s, 4H), 1.30 (t, J = 7.1 Hz, 6H). 3636 1,1'-(부탄-1,4-디일)비스(2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-5-카르복실산)1,1'-(butane-1,4-diyl)bis(2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole -5-carboxylic acid) 1H NMR (300 MHz, DMSO-d 6) δ 12.83 (s, 2H), 8.07 (d, J = 1.5 Hz, 2H), 7.82 (dd, J = 8.4, 1.7 Hz, 2H), 7.56 (d, J = 8.5 Hz, 2H), 6.59 (s, 2H), 4.56 (q, J = 6.9 Hz, 4H), 4.27 (s, 4H), 3.17 (s, 2H), 2.09 (s, 6H), 1.88 (s, 4H), 1.30 (t, J = 7.1 Hz, 6H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 12.83 (s, 2H), 8.07 (d, J = 1.5 Hz, 2H), 7.82 (dd, J = 8.4, 1.7 Hz, 2H), 7.56 (d, J = 8.5 Hz, 2H), 6.59 (s, 2H), 4.56 (q, J = 6.9 Hz, 4H), 4.27 (s, 4H), 3.17 (s, 2H), 2.09 (s, 6H), 1.88 ( s, 4H), 1.30 (t, J = 7.1 Hz, 6H).

하기 표 6의 화합물 37, 38 및 39의 합성Synthesis of compounds 37, 38 and 39 of Table 6 below

중간체 1Intermediate 1

1-에틸-3-메틸-11-ethyl-3-methyl-1 HH -피라졸-5-카르보닐 이소티오시아네이트-pyrazole-5-carbonyl isothiocyanate

Figure pat00054
Figure pat00054

1-에틸-3-메틸-1H-피라졸-5-카르복실산 (500 mg, 3.24 mmol)을 CH2Cl2 (5.4 mL)에 용해시켰다. 이 혼합물에 DMF (0.2 mL)를 첨가하고, 이어서 옥살릴 클로라이드 (0.417 mL, 4.86 mmol)를 천천히 첨가하였다. 실온에서 1시간 동안 교반 후, 용매를 진공 하에 제거하고, CH2Cl2으로 2회 (각각 30 mL) 공증발시켰다. 100% 수율을 가정하고 1-에틸-3-메틸-1H-피라졸-5-카르보닐 클로라이드 (559 mg, 3.24 mmol, 100% 수율)를 그대로 후속 반응에 직접 사용하였다.1-Ethyl-3-methyl-1 H -pyrazole-5-carboxylic acid (500 mg, 3.24 mmol) was dissolved in CH 2 Cl 2 (5.4 mL). To this mixture was added DMF (0.2 mL) followed by slow addition of oxalyl chloride (0.417 mL, 4.86 mmol). After stirring at room temperature for 1 h, the solvent was removed in vacuo and co-evaporated with CH 2 Cl 2 twice (30 mL each). Assuming 100% yield, 1-ethyl-3-methyl- 1H -pyrazole-5-carbonyl chloride (559 mg, 3.24 mmol, 100% yield) was used directly in the subsequent reaction as such.

KSCN (472 mg, 4.86 mmol)을 아세톤 (2 mL)에 용해시켜준 뒤 0 ℃로 냉각시켰다. 0 ℃에서 5분 동안 교반한 후, 1-에틸-3-메틸-1H-피라졸-5-카르보닐 클로라이드 (559 mg, 3.24 mmol)를 아세톤 (2 mL)에 녹여 용액으로서 첨가하였다. 첨가 후, 반응물을 0 ℃에서 20분동안 교반 하였다. 이후 haxanes (50 mL)을 반응 혼합물에 첨가하고, 진공 하에 부피의 3분의 1로 농축시켰다. hexanes 첨가 및 농축의 과정을 2회 (각각 50 mL의 hexanes) 반복하였다. 마지막 농축 후, hexanes (50 mL)을 첨가하고, hexanes (100 mL)으로 고체를 여과한 뒤, 여과된 투명한 담황색 여과물을 농축시키고, 크로마토그래피 (30 g 실리카 칼럼; 0-20% EtOAc / hexanes으로 용리시킴)에 의해 정제하여 1-에틸-3-메틸-1H-피라졸-5-카르보닐 이소티오시아네이트 (297 mg, 1.52 mmol, 47% 수율)를 투명한 무색 액체로서 수득하였다. KSCN (472 mg, 4.86 mmol) was dissolved in acetone (2 mL) and then cooled to 0 °C. After stirring at 0° C. for 5 minutes, 1-ethyl-3-methyl-1 H -pyrazole-5-carbonyl chloride (559 mg, 3.24 mmol) was dissolved in acetone (2 mL) and added as a solution. After addition, the reaction was stirred at 0 °C for 20 min. Then haxanes (50 mL) were added to the reaction mixture and concentrated to one third of the volume in vacuo. The process of hexanes addition and concentration was repeated twice (each of 50 mL of hexanes). After the last concentration, hexanes (50 mL) was added, the solid was filtered with hexanes (100 mL), the filtered clear pale yellow filtrate was concentrated, and the filtrate was chromatographed (30 g silica column; 0-20% EtOAc / hexanes) eluted with) to give 1-ethyl-3-methyl-1 H -pyrazole-5-carbonyl isothiocyanate (297 mg, 1.52 mmol, 47% yield) as a clear colorless liquid.

1 H NMR (300 MHz, chloroform-d) δ 6.72 (d, J = 0.7 Hz, 1H), 4.49 (q, J = 7.2 Hz, 2H), 2.29 (d, J = 0.6 Hz, 3H), 1.40 (t, J = 7.2 Hz, 3H). 1 H NMR (300 MHz, chloroform- d ) δ 6.72 (d, J = 0.7 Hz, 1H), 4.49 (q, J = 7.2 Hz, 2H), 2.29 (d, J = 0.6 Hz, 3H), 1.40 ( t, J = 7.2 Hz, 3H).

아실이소티오시아네이트 생성물은 시간이 지나면서 분해되므로 따라서 즉시 후속 반응에 직접 사용하였다. The acylisothiocyanate product degrades over time and is therefore immediately used directly in the subsequent reaction.

중간체 2Intermediate 2

Figure pat00055
Figure pat00055

단계 1: 4-클로로-3-메톡시-5-니트로벤즈아미드Step 1: 4-Chloro-3-methoxy-5-nitrobenzamide

Figure pat00056
Figure pat00056

메틸 4-클로로-3-메톡시-5-니트로벤조에이트 (10.0 g, 40.8 mmol)를 NH4OH (80 mL, 2009 mmol)에 용해시킨 뒤 실온에서 16시간 동안 교반하였다. 교반 후 냉수로 고체를 여과하고, 진공 하에 건조시켜 황갈색 고체상의 4-클로로-3-메톡시5-니트로벤즈아미드 (7.79 g, 33.8 mmol, 83% 수율)를 수득하였다. Methyl 4-chloro-3-methoxy-5-nitrobenzoate (10.0 g, 40.8 mmol) was dissolved in NH 4 OH (80 mL, 2009 mmol) and stirred at room temperature for 16 hours. After stirring, the solid was filtered with cold water and dried under vacuum to give 4-chloro-3-methoxy5-nitrobenzamide (7.79 g, 33.8 mmol, 83% yield) as a tan solid.

1 H NMR (300 MHz, DMSO-d 6) δ 8.29 (s, 1H), 8.05 (d, J = 1.8 Hz, 1H), 7.88 (d, J = 1.9 Hz, 1H), 7.79 (s, 1H), 4.02 (s, 3H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.29 (s, 1H), 8.05 (d, J = 1.8 Hz, 1H), 7.88 (d, J = 1.9 Hz, 1H), 7.79 (s, 1H) , 4.02 (s, 3H).

단계 2: Step 2: terttert -부틸 (-Butyl ( EE )-(4-((4-카르바모일-2-메톡시-6-니트로페닐)아미노)부트-2-엔-1-일)카르바메이트)-(4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-en-1-yl)carbamate

Figure pat00057
Figure pat00057

밀봉 튜브에 4-클로로-3-메톡시-5-니트로벤즈아미드 (1.41 g, 6.13 mmol)를 EtOH (6.13 mL)에 용해시킨 뒤 (E)-tert-부틸 (4-아미노부트-2-엔-1-일)카르바메이트 (1.37 g, 7.36 mmol) 및 DIPEA (2.38 mL, 18.4 mmol)를 첨가하였다. 반응물을 130 ℃에서 4일동안 교반하고, 실온으로 냉각시켰다. 생성된 오렌지색 고체를 EtOH로 여과한 뒤 건조시켜 주황색 고체상의 tert-부틸 (E)-(4-((4-카르바모일-2-메톡시-6-니트로 페닐)아미노)부트-2-엔-1-일)카르바메이트 (1.97 g, 5.18 mmol, 84% 수율)를 수득하였다. Dissolve 4-chloro-3-methoxy-5-nitrobenzamide (1.41 g, 6.13 mmol) in EtOH (6.13 mL) in a sealed tube ( E ) -tert -butyl (4-aminobut-2-ene) -1-yl)carbamate (1.37 g, 7.36 mmol) and DIPEA (2.38 mL, 18.4 mmol) were added. The reaction was stirred at 130 &lt;0&gt;C for 4 days and cooled to room temperature. The resulting orange solid was filtered with EtOH and dried to form an orange solid tert -butyl ( E )-(4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-ene Obtained -1-yl)carbamate (1.97 g, 5.18 mmol, 84% yield).

1 H NMR (300 MHz, DMSO-d 6) δ 8.18 (d, J = 1.9 Hz, 1H), 8.01 (s, 1H), 7.74 (t, J = 6.2 Hz, 1H), 7.55 (d, J = 1.9 Hz, 1H), 7.31 (s, 1H), 6.92 (d, J = 7.0 Hz, 1H), 5.53 (q, J = 3.1, 2.4 Hz, 2H), 4.09 (d, J = 5.6 Hz, 2H), 3.87 (s, 3H), 3.46 (d, J = 4.2 Hz, 2H), 1.35 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.18 (d, J = 1.9 Hz, 1H), 8.01 (s, 1H), 7.74 (t, J = 6.2 Hz, 1H), 7.55 (d, J = 1.9 Hz, 1H), 7.31 (s, 1H), 6.92 (d, J = 7.0 Hz, 1H), 5.53 (q, J = 3.1, 2.4 Hz, 2H), 4.09 (d, J = 5.6 Hz, 2H) , 3.87 (s, 3H), 3.46 (d, J = 4.2 Hz, 2H), 1.35 (s, 9H).

단계 3: (Step 3: ( EE )-4-((4-아미노부트-2-엔-1-일)아미노)-3-메톡시-5-니트로벤즈아미드, 히드로클로라이드)-4-((4-Aminobut-2-en-1-yl)amino)-3-methoxy-5-nitrobenzamide, hydrochloride

Figure pat00058
Figure pat00058

tert-부틸 (E)-(4-((4-카르바모일-2-메톡시-6-니트로페닐)아미노)부트-2-엔-1-일)카르바메이트 (2.00 g, 5.26 mmol)를 MeOH (5 mL)에 용해시킨 뒤 디옥산 중 4 M HCl (16 mL, 44.7 mmol)을 천천히 첨가하였다. 반응 혼합물을 실온에서 1시간 30분동안 교반한 다음, 생성된 고체를 여과한 뒤, Et2O (100ml X3)으로 3회 세척하고, 진공하에 건조시켜 (E)-4-((4-아미노부트-2-엔-1-일)아미노)-3-메톡시-5-니트로벤즈아미드, 히드로클로라이드을 수득하였으며 별도의 추가 정제없이 다음 반응에 사용하였다. tert -Butyl ( E )-(4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-en-1-yl)carbamate (2.00 g, 5.26 mmol) was dissolved in MeOH (5 mL) and then 4 M HCl in dioxane (16 mL, 44.7 mmol) was added slowly. The reaction mixture was stirred at room temperature for 1 hour and 30 minutes, then the resulting solid was filtered, washed three times with Et 2 O (100 ml X3), and dried under vacuum ( E )-4-((4-amino). But-2-en-1-yl)amino)-3-methoxy-5-nitrobenzamide, hydrochloride, was obtained and used in the next reaction without further purification.

1 H NMR (300 MHz, DMSO-d 6) δ 8.21 (d, J = 1.9 Hz, 1H), 8.05 (s, 1H), 7.90 (s, 3H), 7.58 (d, J = 2.0 Hz, 1H), 7.35 (s, 1H), 5.93-5.81 (m, 1H), 5.62 (dd, J = 14.6, 7.3 Hz, 1H), 4.18 (d, J = 5.7 Hz, 2H), 3.89 (s, 3H), 3.44-3.35 (m, 2H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.21 (d, J = 1.9 Hz, 1H), 8.05 (s, 1H), 7.90 (s, 3H), 7.58 (d, J = 2.0 Hz, 1H) , 7.35 (s, 1H), 5.93-5.81 (m, 1H), 5.62 (dd, J = 14.6, 7.3 Hz, 1H), 4.18 (d, J = 5.7 Hz, 2H), 3.89 (s, 3H), 3.44-3.35 (m, 2H).

중간체 3Intermediate 3

Figure pat00059
Figure pat00059

단계 1: 4-클로로-3-히드록시-5-니트로벤즈아미드Step 1: 4-Chloro-3-hydroxy-5-nitrobenzamide

Figure pat00060
Figure pat00060

4-클로로-3-메톡시-5-니트로벤즈아미드 (2.00 g, 8.68 mmol)를 CH2Cl2 (14.5 mL) 중에 용해 시킨 뒤 반응물에 BBr3 (36.4 mL, CH2Cl2 중 1 M)을 천천히 첨가 후 질소 하에 실온에서 밤새 교반하였다. 반응물에 냉수 (300 mL)를 붓고, 30분 동안 격렬히 교반하였다. 생성된 고체를 여과하고, 건조시켜 4-클로로-3-히드록시-5-니트로벤즈아미드 (1.35 g, 6.23 mmol, 72% 수율)을 수득하였다. 4-Chloro-3-methoxy-5-nitrobenzamide (2.00 g, 8.68 mmol) was dissolved in CH 2 Cl 2 (14.5 mL) followed by BBr 3 (36.4 mL, 1 M in CH 2 Cl 2 ) was added slowly and stirred overnight at room temperature under nitrogen. The reaction was poured with cold water (300 mL) and stirred vigorously for 30 minutes. The resulting solid was filtered and dried to give 4-chloro-3-hydroxy-5-nitrobenzamide (1.35 g, 6.23 mmol, 72% yield).

1 H NMR (300 MHz, DMSO-d 6) δ 11.52 (s, 1H), 8.17 (s, 1H), 7.92 (d, J = 1.9 Hz, 1H), 7.74-7.61 (m, 2H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 11.52 (s, 1H), 8.17 (s, 1H), 7.92 (d, J = 1.9 Hz, 1H), 7.74-7.61 (m, 2H).

단계 2: Step 2: terttert -부틸 (3-(5-카르바모일-2-클로로-3-니트로페녹시)프로필)카르바메이트-Butyl (3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propyl)carbamate

Figure pat00061
Figure pat00061

4-클로로-3-히드록시-5-니트로벤즈아미드 (1.35 g, 6.23 mmol)를 DMF (7.79 mL)에 용해시킨 뒤 tert-부틸 (3-클로로프로필)카르바메이트 (1.35 g, 6.23 mmol) 및 K2CO3 (1.12 g, 8.10 mmol)을 첨가하였다. 반응 혼합물을 70 ℃에서 48시간 동안 교반하였다. 48시간 후, 실리카겔을 넣어준 뒤 DMF를 진공 하에 제거하고 잔류물을 건식 로딩 실리카 칼럼 크로마토그래피 (CH2Cl2:MeOH=9:1)에 의해 정제하여 노란색 고체상의 tert-부틸 (3-(5-카르바모일-2-클로로-3-니트로페녹시)프로필)카르바메이트 (1.61 g, 4.31 mmol, 69% 수율)을 수득하였다. 4-Chloro-3-hydroxy-5-nitrobenzamide (1.35 g, 6.23 mmol) was dissolved in DMF (7.79 mL) followed by tert -butyl (3-chloropropyl)carbamate (1.35 g, 6.23 mmol) and K 2 CO 3 (1.12 g, 8.10 mmol) were added. The reaction mixture was stirred at 70 °C for 48 h. After 48 hours, silica gel was added, DMF was removed in vacuo, and the residue was purified by dry loading silica column chromatography (CH 2 Cl 2 :MeOH=9:1) to form a yellow solid tert -butyl (3-( Obtained 5-carbamoyl-2-chloro-3-nitrophenoxy)propyl)carbamate (1.61 g, 4.31 mmol, 69% yield).

1 H NMR (500 MHz, DMSO-d 6) δ 8.28 (s, 1H), 8.04 (d, J = 1.7 Hz, 1H), 7.86 (d, J = 1.9 Hz, 1H), 7.77 (s, 1H), 6.91 (t, J = 5.7 Hz, 1H), 4.23 (t, J = 6.1 Hz, 2H), 3.13 (q, J = 6.7 Hz, 2H), 1.90 (p, J = 6.4 Hz, 2H), 1.36 (s, 9H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 8.28 (s, 1H), 8.04 (d, J = 1.7 Hz, 1H), 7.86 (d, J = 1.9 Hz, 1H), 7.77 (s, 1H) , 6.91 (t, J = 5.7 Hz, 1H), 4.23 (t, J = 6.1 Hz, 2H), 3.13 (q, J = 6.7 Hz, 2H), 1.90 (p, J = 6.4 Hz, 2H), 1.36 (s, 9H).

화합물 37, 38, 및 39의 합성Synthesis of compounds 37, 38, and 39

Figure pat00062
Figure pat00062

Figure pat00063
Figure pat00063

단계 1: Step 1: terttert -부틸 (-Butyl ( EE )-(3-(5-카르바모일-2-((4-((4-카르바모일-2-메톡시-6-니트로페닐)아미노)부트-2-엔-1-일)아미노)-3-니트로페녹시)프로필)카르바메이트)-(3-(5-carbamoyl-2-((4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-en-1-yl)amino) -3-nitrophenoxy)propyl)carbamate

Figure pat00064
Figure pat00064

(E)-4-((4-아미노부트-2-엔-1-일)아미노)-3-메톡시-5-니트로벤즈아미드, 히드로클로라이드 (0.930 g, 3.32 mmol)를 i-PrOH (5.53 mL)에 용해시켜준 뒤 DIPEA (2.31 mL, 13.3 mmol)를 첨가하고, tert-부틸 (3-(5-카르바모일-2-클로로-3-니트로페녹시)프로필)카르바메이트 (1.12 g, 2.99 mmol)를 첨가하였다. 반응 혼합물을 130 ℃에서 72시간 동안 교반시켜 주었다. 반응 혼합물을 실온으로 냉각시키고 실리카겔을 넣어준 뒤 용매를 진공 하에 제거하고 잔류물을 건식 로딩 실리카 칼럼 크로마토그래피 (CH2Cl2:MeOH=9:1)로 정제하여 붉은색 고체상의 tert-부틸 (E)-(3-(5-카르바모일-2-((4-((4-카르바모일-2-메톡시-6-니트로페닐)아미노)부트-2-엔-1-일)아미노)-3-니트로페녹시)프로필)카르바메이트 (500 mg, 0.810 mmol, 25% 수율)를 수득하였다. ( E )-4-((4-aminobut-2-en-1-yl)amino)-3-methoxy-5-nitrobenzamide, hydrochloride (0.930 g, 3.32 mmol) was mixed with i -PrOH (5.53 mL), DIPEA (2.31 mL, 13.3 mmol) was added, and tert -butyl (3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propyl)carbamate (1.12 g) , 2.99 mmol) was added. The reaction mixture was stirred at 130 °C for 72 hours. The reaction mixture was cooled to room temperature, silica gel was added, the solvent was removed under vacuum, and the residue was purified by dry loading silica column chromatography (CH 2 Cl 2 :MeOH=9:1) to form a red solid tert -butyl ( E )-(3-(5-carbamoyl-2-((4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-en-1-yl)amino )-3-nitrophenoxy)propyl)carbamate (500 mg, 0.810 mmol, 25% yield) was obtained.

1 H NMR (300 MHz, DMSO-d 6) δ 8.15 (dd, J = 1.9, 0.9 Hz, 2H), 8.01 (s, 2H), 7.70 (td, J = 6.2, 3.0 Hz, 2H), 7.51 (t, J = 2.2 Hz, 2H), 7.31 (s, 2H), 6.91 (t, J = 5.3 Hz, 1H), 5.63 (q, J = 1.5 Hz, 2H), 4.18-3.97 (m, 6H), 3.82 (s, 3H), 3.08 (q, J = 6.4 Hz, 2H), 1.86 (q, J = 6.4 Hz, 2H), 1.35 (s, 9H). 1 H NMR (300 MHz, DMSO- d 6 ) δ 8.15 (dd, J = 1.9, 0.9 Hz, 2H), 8.01 (s, 2H), 7.70 (td, J = 6.2, 3.0 Hz, 2H), 7.51 ( t, J = 2.2 Hz, 2H), 7.31 (s, 2H), 6.91 (t, J = 5.3 Hz, 1H), 5.63 (q, J = 1.5 Hz, 2H), 4.18-3.97 (m, 6H), 3.82 (s, 3H), 3.08 (q, J = 6.4 Hz, 2H), 1.86 (q, J = 6.4 Hz, 2H), 1.35 (s, 9H).

단계 2: Step 2: terttert -부틸 (-Butyl ( EE )-(3-(3-아미노-2-((4-((2-아미노-4-카르바모일-6-메톡시페닐)아미노)부트-2-엔-1-일)아미노)-5-카르바모일페녹시)프로필)카르바메이트)-(3-(3-amino-2-((4-((2-amino-4-carbamoyl-6-methoxyphenyl)amino)but-2-en-1-yl)amino)-5 -carbamoylphenoxy)propyl)carbamate

Figure pat00065
Figure pat00065

tert-부틸 (E)-(3-(5-카르바모일-2-((4-((4-카르바모일-2-메톡시-6-니트로페닐)아미노)부트-2-엔-1-일)아미노)-3-니트로페녹시)프로필)카르바메이트 (500 mg, 0.810 mmol)를 MeOH (10.2 mL)에 용해시키고, 물 (5 mL)에 녹인 Na2S2O4 (1.98 g, 11.3 mmol)를 첨가하고, 실온에서 25분 동안 교반하였다. 이어서, 고체 탄산수소나트륨 (2.93 g, 34.8 mmol)을 첨가하고, 실온에서 10분간 추가로 교반하였다. 이어서 고체를 여과시켜준 뒤 MeOH로 세척하면서 셀라이트®를 통해 여과하였다. 목적 생성물을 함유하는 여과물을 진공 하에 농축시켜 잔류물을 건식 로딩 실리카 칼럼 크로마토그래피 CH2Cl2 중 0-10% (10:1 MeOH:aq.NH4OH(28%))에 의해 정제하여 노란색 고체상의 tert-부틸 (E)-(3-(3-아미노-2-((4-((2-아미노-4-카르바모일-6-메톡시페닐)아미노)부트-2-엔-1-일)아미노)-5-카르바모일페녹시)프로필)카르바메이트 (297 mg, 0.533 mmol, 66% 수율)을 수득하였다. tert -Butyl ( E )-(3-(5-carbamoyl-2-((4-((4-carbamoyl-2-methoxy-6-nitrophenyl)amino)but-2-ene-1 -yl)amino)-3-nitrophenoxy)propyl)carbamate (500 mg, 0.810 mmol) was dissolved in MeOH (10.2 mL) and Na 2 S 2 O 4 (1.98 g) dissolved in water (5 mL) , 11.3 mmol) and stirred at room temperature for 25 min. Then solid sodium hydrogen carbonate (2.93 g, 34.8 mmol) was added and stirred at room temperature for a further 10 minutes. The solid was then filtered and filtered through Celite®, washing with MeOH. The filtrate containing the desired product was concentrated in vacuo and the residue was purified by dry loading silica column chromatography 0-10% in CH 2 Cl 2 (10:1 MeOH:aq.NH 4 OH (28%)) tert -Butyl ( E )-(3-(3-amino-2-((4-((2-amino-4-carbamoyl-6-methoxyphenyl)amino)but-2-ene- as yellow solid 1-yl)amino)-5-carbamoylphenoxy)propyl)carbamate (297 mg, 0.533 mmol, 66% yield) was obtained.

1 H NMR (500 MHz, methanol-d 4 ) δ 6.93 (d, J = 1.8 Hz, 2H), 6.87 (dd, J = 5.5, 1.9 Hz, 2H), 5.72 (td, J = 4.8, 3.6 Hz, 2H), 3.99 (t, J = 6.1 Hz, 2H), 3.79 (s, 3H), 3.57 (dd, J = 13.2, 4.4 Hz, 4H), 3.23 (t, J = 6.7 Hz, 2H), 1.93 (p, J = 6.4 Hz, 2H), 1.41 (s, 9H). 1 H NMR (500 MHz, methanol- d 4 ) δ 6.93 (d, J = 1.8 Hz, 2H), 6.87 (dd, J = 5.5, 1.9 Hz, 2H), 5.72 (td, J = 4.8, 3.6 Hz, 2H), 3.99 (t, J = 6.1 Hz, 2H), 3.79 (s, 3H), 3.57 (dd, J = 13.2, 4.4 Hz, 4H), 3.23 (t, J = 6.7 Hz, 2H), 1.93 ( p, J = 6.4 Hz, 2H), 1.41 (s, 9H).

단계 3: Step 3: terttert -부틸 (-Butyl ( EE )-(3-((5-카르바모일-1-(4-(5-카르바모일-2-(1-에틸-3-메틸-1)-(3-((5-carbamoyl-1-(4-(5-carbamoyl-2-(1-ethyl-3-methyl-1) HH -피라졸-5-카르복사미도)-7-메톡시-1-Pyrazole-5-carboxamido)-7-methoxy-1 HH -벤조[-benzo[ dd ]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1]imidazol-1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl-1 HH -피라졸-5-카르복사미도)-1-Pyrazole-5-carboxamido)-1 HH -벤조[-benzo[ dd ]이미다졸-7-일)옥시)프로필)카르바메이트 (화합물 37)]imidazol-7-yl)oxy)propyl)carbamate (Compound 37)

Figure pat00066
Figure pat00066

0 ℃에서 tert-부틸 (E)-(3-(3-아미노-2-((4-((2-아미노-4-카르바모일-6-메톡시페닐)아미노)부트-2-엔-1-일)아미노)-5-카르바모일페녹시)프로필)카르바메이트 (100 mg, 0.179 mmol)를 DMF (1.79 mL)에 용해시킨 뒤 1-에틸-3-메틸-1H-피라졸5-카르보닐 이소티오시아네이트 (70 mg, 0.358 mmol)를 서서히 첨가하고 35분간 교반하였다. 이어서 1-에틸-3-(3-디메틸아미노프로필)카르보디이미드 (83 mg, 0.537 mmol)를 첨가하고, 이어서 트리에틸 아민 (0.150 mL, 1.07 mmol)을 첨가한 뒤 상온에서 밤새 교반하였다 (~16시간). 반응물을 3:1 물:포화 aq.NH4Cl 용액 (40 mL)을 첨가하고, 3:1 클로로포름:에탄올 (4 X 40 mL)로 추출하였다. 유기층을 MgSO4로 건조시키고, 농축시켰다. 생성된 잔류물을 실리카 겔 상에서 CH2Cl2 중 2 - 20% (10:1 MeOH:aq.NH4OH)로 정제하여 회백색 고체상의 tert-부틸 (E)-(3-((5-카르바모일-1-(4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-7-메톡시-1H-벤조[d]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-7-일)옥시)프로필)카르바메이트 (35.3 mg, 0.04 mmol, 22% 수율)을 수득하였다. tert -Butyl ( E )-(3-(3-amino-2-((4-((2-amino-4-carbamoyl-6-methoxyphenyl)amino)but-2-ene- at 0° C.) 1-yl)amino)-5-carbamoylphenoxy)propyl)carbamate (100 mg, 0.179 mmol) was dissolved in DMF (1.79 mL) followed by 1-ethyl-3-methyl- 1H -pyrazole 5-carbonyl isothiocyanate (70 mg, 0.358 mmol) was added slowly and stirred for 35 min. Then 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (83 mg, 0.537 mmol) was added, followed by triethyl amine (0.150 mL, 1.07 mmol), followed by stirring at room temperature overnight (~ 16 hours). The reaction was added 3:1 water:saturated aq.NH 4 Cl solution (40 mL) and extracted with 3:1 chloroform:ethanol (4 X 40 mL). The organic layer was dried over MgSO 4 and concentrated. The resulting residue was purified on silica gel with 2 - 20% (10:1 MeOH:aq.NH 4 OH) in CH 2 Cl 2 on silica gel tert -butyl ( E )-(3-((5-car) as an off-white solid. Bamoyl-1-(4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-7-methoxy- 1H -benzo[ d ]imidazol-1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ] Obtained imidazol-7-yl)oxy)propyl)carbamate (35.3 mg, 0.04 mmol, 22% yield).

1 H NMR (500 MHz, DMSO-d 6 ) δ 12.83 (s, 2H), 7.97 (s, 2H), 7.63 (d, J = 4.2 Hz, 2H), 7.41-7.33 (m, 2H), 7.30 (d, J = 6.1 Hz, 2H), 6.88 (t, J = 5.8 Hz, 1H), 6.50 (s, 2H), 5.82 (qd, J = 17.6, 16.8, 7.8 Hz, 2H), 4.98-4.84 (m, 4H), 4.57-4.43 (m, 4H), 3.98 (t, J = 6.0 Hz, 2H), 3.72 (s, 3H), 3.05-2.95 (m, 2H), 2.09 (s, 6H), 1.74-1.62 (m, 2H), 1.31 (s, 9H), 1.25 (t, J = 7.3 Hz, 6H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 12.83 (s, 2H), 7.97 (s, 2H), 7.63 (d, J = 4.2 Hz, 2H), 7.41-7.33 (m, 2H), 7.30 ( d, J = 6.1 Hz, 2H), 6.88 (t, J = 5.8 Hz, 1H), 6.50 (s, 2H), 5.82 (qd, J = 17.6, 16.8, 7.8 Hz, 2H), 4.98-4.84 (m) , 4H), 4.57-4.43 (m, 4H), 3.98 (t, J = 6.0 Hz, 2H), 3.72 (s, 3H), 3.05-2.95 (m, 2H), 2.09 (s, 6H), 1.74- 1.62 (m, 2H), 1.31 (s, 9H), 1.25 (t, J = 7.3 Hz, 6H).

단계 4: (Step 4: ( EE )-7-(3-아미노프로폭시)-1-(4-(5-카바모일-2-(1-에틸-3-메틸-1)-7-(3-aminopropoxy)-1-(4-(5-carbamoyl-2-(1-ethyl-3-methyl-1) HH -피라졸-5-카복스아미도)-7-메톡시-1-Pyrazole-5-carboxamido)-7-methoxy-1 HH -벤조[-benzo[ dd ]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1]imidazol-1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl-1 HH -피라졸-5-카르복사미도)-1-Pyrazole-5-carboxamido)-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복사미드 (화합물 38)]Imidazole-5-carboxamide (Compound 38)

Figure pat00067
Figure pat00067

tert-부틸 (E)-(3-((5-카르바모일-1-(4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-7-메톡시-1H-벤조[d]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1H-벤조[d]이미다졸-7-일)옥시)프로필)카바메이트(화합물 37, 12.0mg, 0.014mmol)에 MeOH(0.006mL)을 용해 시킨 뒤 디옥산 (0.50mL) 중 4 M HCl을 천천히 첨가하였다. 혼합물을 실온에서 1시간 동안 교반한 후, 생성된 고체를 여과 시켜준 뒤, Et2O로 3회 세척하고, 진공 하에 건조시켜 백색 고체상의 (E)-7-(3-아미노프로폭시)-1-(4-(5-카바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카복스아미도)-7-메톡시-1H-벤조[d]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드를 (9.8 mg, 0.013 mmol, 90%) 수득하였다. R f 0.16 (CH2Cl2:MeOH:aq.NH4OH(28%)=8:2:0.2). tert -Butyl ( E )-(3-((5-carbamoyl-1-(4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-car) copymido)-7-methoxy- 1H -benzo[ d ]imidazol-1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl- 1H -pyrazole -5-carboxamido)-1H-benzo[d]imidazol-7-yl)oxy)propyl)carbamate (Compound 37, 12.0mg , 0.014mmol) was dissolved in MeOH (0.006mL) 4 M HCl in oxane (0.50 mL) was added slowly. After the mixture was stirred at room temperature for 1 hour, the resulting solid was filtered, washed 3 times with Et 2 O, and dried under vacuum to form ( E )-7-(3-aminopropoxy)- as a white solid. 1-(4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-7-methoxy- 1H -benzo[ d ]imidazole -1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido) -1H -benzo[ d ]imidazole- 5-carboxamide was obtained (9.8 mg, 0.013 mmol, 90%). R f 0.16 (CH 2 Cl 2 :MeOH:aq.NH 4 OH(28%)=8:2:0.2).

1 H NMR (400 MHz, DMSO-d 6 ) δ 8.13 (s, 3H), 8.04 (s, 2H), 7.65 (d, J = 7.2 Hz, 2H), 7.41 (s, 1H), 7.36 (d, J = 9.6 Hz, 2H), 6.51 (d, J = 12.1 Hz, 2H), 5.86-5.71 (m, 2H), 4.93 (dd, J = 12.3, 5.0 Hz, 4H), 4.49 (t, J = 8.0 Hz, 4H), 4.13 (d, J = 6.3 Hz, 2H), 3.74 (s, 3H), 2.88 (q, J = 6.4 Hz, 2H), 2.10 (d, J = 6.3 Hz, 6H), 1.94 (t, J = 6.8 Hz, 2H), 1.25 (dt, J = 11.6, 5.6 Hz, 6H). 1 H NMR (400 MHz, DMSO- d 6 ) δ 8.13 (s, 3H), 8.04 (s, 2H), 7.65 (d, J = 7.2 Hz, 2H), 7.41 (s, 1H), 7.36 (d, J = 9.6 Hz, 2H), 6.51 (d, J = 12.1 Hz, 2H), 5.86-5.71 (m, 2H), 4.93 (dd, J = 12.3, 5.0 Hz, 4H), 4.49 (t, J = 8.0) Hz, 4H), 4.13 (d, J = 6.3 Hz, 2H), 3.74 (s, 3H), 2.88 (q, J = 6.4 Hz, 2H), 2.10 (d, J = 6.3 Hz, 6H), 1.94 ( t, J = 6.8 Hz, 2H), 1.25 (dt, J = 11.6, 5.6 Hz, 6H).

단계 5: (Step 5: ( EE )-1-(4-(5-카르바모일-2-(1-에틸-3-메틸-1)-1-(4-(5-carbamoyl-2-(1-ethyl-3-methyl-1) HH -피라졸-5-카르복스아미도)-7-(3-(1-에틸-3-메틸-1-Pyrazole-5-carboxamido)-7-(3-(1-ethyl-3-methyl-1) HH -피라졸-5-카르복스아미도)프로폭시)-1-Pyrazole-5-carboxamido)propoxy)-1 HH -벤조[-benzo[ dd ]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1]imidazol-1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl-1 HH -피라졸-5-카르복스아미도)-7-메톡시-1-Pyrazole-5-carboxamido)-7-methoxy-1 HH -벤조[-benzo[ dd ]이미다졸-5-카르복사미드 (화합물 39)]Imidazole-5-carboxamide (Compound 39)

Figure pat00068
Figure pat00068

1-에틸-3-메틸-1H-피라졸-5-카복실산 (8.0mg, 0.051mmol), HATU(26mg, 0.051mmol), HOBt(5mg, 0.034 mmol) 및 TEA(0.047 mL, 0.34 mmol)를 DMF(0.85mL)에 용해시켰다. 반응물을 실온에서 5분 동안 교반한 후, (E)-7-(3-아미노프로폭시)-1-(4-(5-카바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-7-메톡시-1H-벤조[d]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복사미도)-1H-벤조[d]이미다졸-5-카르복사미드(화합물 38, 27.0 mg, 0.034 mmol)를 첨가하였다. 반응을 실온에서 16시간 동안 교반한 후, 반응 혼합물에 물에 붓고 EtOAc로 추출하였다. 유기층을 MgSO4로 건조시켰다. 잔류물을 실리카겔 플래시 컬럼 크로마토그래피(CH2Cl2:MeOH:aq.NH4OH(28%)= 90:10:1)로 정제하여 백색 고체상의 (E)-1-(4-(5-카르바모일-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-7-(3-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)프로폭시)-1H-벤조[d]이미다졸-1-일)부트-2-엔-1-일)-2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-7-메톡시-1H-벤조[d]이미다졸-5-카르복사미드 (15.6 mg, 0.017 mmol, 50%)를 수득하였다. R f 0.57 (CH2Cl2:MeOH:aq. NH4OH(28%)=80:20:2).1-ethyl-3-methyl- 1H -pyrazole-5-carboxylic acid (8.0mg, 0.051mmol), HATU (26mg, 0.051mmol), HOBt (5mg, 0.034mmol) and TEA (0.047 mL, 0.34mmol) It was dissolved in DMF (0.85 mL). After the reaction was stirred at room temperature for 5 min, ( E )-7-(3-aminopropoxy)-1-(4-(5-carbamoyl-2-(1-ethyl-3-methyl- 1H- ) Pyrazole-5-carboxamido)-7-methoxy- 1H -benzo[ d ]imidazol-1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl) -1H- Pyrazole -5-carboxamido) -1H -benzo[ d ]imidazole-5-carboxamide (compound 38, 27.0 mg, 0.034 mmol) was added. After the reaction was stirred at room temperature for 16 h, the reaction mixture was poured into water and extracted with EtOAc. The organic layer was dried over MgSO 4 . The residue was purified by silica gel flash column chromatography (CH 2 Cl 2 :MeOH:aq.NH 4 OH(28%)=90:10:1) as ( E )-1-(4-(5-) as a white solid. Carbamoyl-2-(1-ethyl-3-methyl- 1H -pyrazole-5-carboxamido)-7-(3-(1-ethyl-3-methyl- 1H -pyrazole-5) -carboxamido )propoxy)-1H-benzo[ d ]imidazol-1-yl)but-2-en-1-yl)-2-(1-ethyl-3-methyl- 1H -pyra Obtained sol-5-carboxamido)-7-methoxy-1 H -benzo[ d ]imidazole-5-carboxamide (15.6 mg, 0.017 mmol, 50%). R f 0.57 (CH 2 Cl 2 :MeOH:aq. NH 4 OH(28%)=80:20:2).

1 H NMR (500 MHz, DMSO-d 6) δ 12.82 (d, J = 11.8 Hz, 2H), 8.38 (t, J = 5.8 Hz, 1H), 7.97 (s, 2H), 7.63 (s, 2H), 7.36 (d, J = 7.1 Hz, 2H), 7.29 (d, J = 3.8 Hz, 2H), 6.50 (d, J = 9.3 Hz, 3H), 5.90-5.78 (m, 2H), 4.92 (dd, J = 25.0, 4.9 Hz, 4H), 4.50 (dt, J = 14.4, 7.2 Hz, 4H), 4.30 (q, J = 7.1 Hz, 2H), 4.01 (t, J = 6.0 Hz, 2H), 3.71 (s, 3H), 3.27 (q, J = 6.5 Hz, 2H), 2.12 (s, 3H), 2.10 (s, 3H), 2.08 (s, 3H), 1.80 (p, J = 6.4 Hz, 2H), 1.25 (q, J = 7.5 Hz, 6H), 1.18 (t, J = 7.1 Hz, 3H). 1 H NMR (500 MHz, DMSO- d 6 ) δ 12.82 (d, J = 11.8 Hz, 2H), 8.38 (t, J = 5.8 Hz, 1H), 7.97 (s, 2H), 7.63 (s, 2H) , 7.36 (d, J = 7.1 Hz, 2H), 7.29 (d, J = 3.8 Hz, 2H), 6.50 (d, J = 9.3 Hz, 3H), 5.90-5.78 (m, 2H), 4.92 (dd, J = 25.0, 4.9 Hz, 4H), 4.50 (dt, J = 14.4, 7.2 Hz, 4H), 4.30 (q, J = 7.1 Hz, 2H), 4.01 (t, J = 6.0 Hz, 2H), 3.71 ( s, 3H), 3.27 (q, J = 6.5 Hz, 2H), 2.12 (s, 3H), 2.10 (s, 3H), 2.08 (s, 3H), 1.80 (p, J = 6.4 Hz, 2H), 1.25 (q, J = 7.5 Hz, 6H), 1.18 (t, J = 7.1 Hz, 3H).

세포 배양과 관련 시약 Cell culture and related reagents

인간 monocyte 세포주인 THP1-dual cell 및 THP1-dual STING KO cell은 Invivogen 사에서 구입하여 사용하였다. 세포주는 1X RPMI 1640 2.05 Mm L-Glutamine (HyClone), 10%의 Fetal Bovine Serum (FBS, HyClone) 및 1%의 페니실린(Corning)의 조건으로 37℃의 CO2 in humidified incubator에서 배양하였다. Human monocyte cell lines, THP1-dual cell and THP1-dual STING KO cell, were purchased from Invivogen and used. The cell line was cultured in a humidified incubator at 37° C. in CO 2 under conditions of 1X RPMI 1640 2.05 Mm L-Glutamine (HyClone), 10% Fetal Bovine Serum (FBS, HyClone) and 1% penicillin (Corning).

ISRE 리포터 어세이 ISRE Reporter Assay

IRSE 매개 면역 반응을 모니터링하기 위하여 THP1 세포주를 384 well plate (Grenier)에 분주 후 각각의 화합물을 처리하였다. 24시간이 지난 후, 10 μl의 상등액을 취하여 QUANTI-Luc assay (Invivogen)의 제조 프로토콜에 따라 분석하였으며, 이를 통해 리포터 신호를 측정하였다. Relative Luminescent (RU)은 마이크로플레이트 판독기(TECAN)로 측정하였다. 측정 결과는 DMSO를 기준으로 정규화(normalization)하여 분석하였다.In order to monitor the IRSE-mediated immune response, the THP1 cell line was dispensed into a 384 well plate (Grenier) and each compound was treated. After 24 hours, 10 μl of the supernatant was taken and analyzed according to the manufacturing protocol of the QUANTI-Luc assay (Invivogen), and the reporter signal was measured through this. Relative Luminescent (RU) was measured with a microplate reader (TECAN). The measurement results were analyzed by normalization based on DMSO.

세포 생존성 검사Cell viability test

세포 생존성 검사는 Luciferase assay 후 잔여 세포를 사용하여 수행하였다. 24시간 화합물 처리하였다. 그 후, 제조 프로토콜에 따라 CellTiter 96 Aqueous One Solution Proliferation Assay (MTS, Promega)을 사용하여 세포 생존 여부를 분석하였다. 흡광도 신호는 마이크로플레이트 리더(TECAN)를 사용하여 측정하였으며, 분석 결과는 DMSO를 기준으로 정규화(normalization)하였다.Cell viability test was performed using residual cells after Luciferase assay. 24 hours compound treatment. Thereafter, cell viability was analyzed using CellTiter 96 Aqueous One Solution Proliferation Assay (MTS, Promega) according to the manufacturing protocol. The absorbance signal was measured using a microplate reader (TECAN), and the analysis results were normalized based on DMSO.

Western BlotWestern Blot

웨스턴 블랑팅 분석을 위하여 세포를 PBS로 워싱하고 RIPA 완충제(Bioseasang)를 사용하여 용해하였다. 이후 4 ℃에서 20분 동안 20000g로 원심분리하였다. 그 후, Cell lysate는 SDS-폴리아크릴아미드 젤 전기영동에 의해 분리되어 Trans-Blot Turbo Transfer System (Bio-Rad)을 통해 PVDF membrane으로 트랜스퍼하였다. 실온에서 1시간 동안 TBST에 녹여진 5% BSA에서 membrane을 blocking 한 후, 밤새 1차 항체 용액으로 처리하였다. 모든 항체 용액은 TBST를 사용하여 다음의 농도로 희석되었다: anti-STING (Cell Signaling/no.13647, 1:1000), anti-pSTING (Cell Signaling/no. 19781, 1:1000), anti-TBK1 (Cell Signaling/no.3504, 1:1000), anti-pTBK1(Cell Signaling/no.5483, 1:1000), anti-IRF3(Cell Signaling/no.11904, 1:1000), anti-pIRF3(Cell Signaling/no.4947, 1:1000), anti-STAT1(Cell Signaling/no.9172, 1:1000), anti-pSTAT(Cell Signaling/no.9167, 1:1000), anti-Actin(Cell Signaling/4970, 1:1000). TBST로 세척한 후 anti-Rabbit-HRP 기반 2차 항체(세포 신호/7074, 1:3000)에 의해 웨스턴 블랏 신호를 검출하였다. β-Actin은 총단백질량에 대한 대조군으로서 사용되었다. ECL 시약을 사용하여 발광 신호를 검출하였으며, ChemiDoc 영상촬영 시스템(Bio-Rad)으로 면역반응 밴드를 캡쳐하였다.For Western blotting analysis, cells were washed with PBS and lysed using RIPA buffer (Bioseasang). Thereafter, centrifugation was performed at 20000g at 4°C for 20 minutes. Then, the cell lysate was separated by SDS-polyacrylamide gel electrophoresis and transferred to a PVDF membrane through a Trans-Blot Turbo Transfer System (Bio-Rad). After blocking the membrane in 5% BSA dissolved in TBST for 1 hour at room temperature, it was treated with a primary antibody solution overnight. All antibody solutions were diluted with TBST to the following concentrations: anti-STING (Cell Signaling/no.13647, 1:1000), anti-pSTING (Cell Signaling/no. 19781, 1:1000), anti-TBK1 (Cell Signaling/no.3504, 1:1000), anti-pTBK1 (Cell Signaling/no.5483, 1:1000), anti-IRF3 (Cell Signaling/no.11904, 1:1000), anti-pIRF3 (Cell Signaling/no.4947, 1:1000), anti-STAT1 (Cell Signaling/no.9172, 1:1000), anti-pSTAT (Cell Signaling/no.9167, 1:1000), anti-Actin (Cell Signaling/ 4970, 1:1000). After washing with TBST, a Western blot signal was detected by an anti-Rabbit-HRP-based secondary antibody (cell signal/7074, 1:3000). β-Actin was used as a control for the total protein amount. Luminescence signals were detected using ECL reagent, and immunoreactive bands were captured with ChemiDoc imaging system (Bio-Rad).

ELISA (Enzyme-Linked Immunosorbent Assay) / 효소결합면역흡착검사ELISA (Enzyme-Linked Immunosorbent Assay) / Enzyme-Linked Immunosorbent Assay

IFN-α 및 IP-10 사이토카인 분비는 R&D 시스템의 ELISA 키트를 사용하여 정량화하였다. THP1 세포는 96-웰 플레이트에 분주하였고, 화합물 처리 후 24시간 배양하였다. 100 μl의 세포 배양 상등액을 추출하였고, 이후 제조 프로토콜을 사용하여 분석하였다. IFN-α and IP-10 cytokine secretion was quantified using an ELISA kit from R&D Systems. THP1 cells were seeded in 96-well plates, and cultured for 24 hours after compound treatment. 100 μl of the cell culture supernatant was extracted and then analyzed using the preparation protocol.

rt-PCR 을 통한 유전자 발현 분석 Gene expression analysis by rt-PCR

세포 pellet은 NucleoSpin RNA plus (MN)을 사용하여 제조 프로토콜에 따라 RNA를 분리하였다. 유전자 발현 여부는 IQ SYBR green Supermix (Bio-Rad)를 사용하여 real-time PCR (CFX96 Real-Time PCR detection system, Bio-Rad)에 의해 평가되었다. mRNA level은 GAPDH에 의해 정규화(normalization) 되고 비교 Ct 방법에 의해 계산되었다.For cell pellets, RNA was isolated according to the manufacturing protocol using NucleoSpin RNA plus (MN). Gene expression was assessed by real-time PCR (CFX96 Real-Time PCR detection system, Bio-Rad) using IQ SYBR green Supermix (Bio-Rad). mRNA levels were normalized by GAPDH and calculated by the comparative Ct method.

다음과 같은 프라이머 시퀀스가 사용되었다:The following primer sequences were used:

IFNB (F) 5'-AGGATTCTGCATTACCTGAA-3' (R) 5'-GGCTAGGAGATCTTCAGTTT-3',IFNB (F) 5'-AGGATTCTGCATTACCTGAA-3' (R) 5'-GGCTAGGAGATCTTCAGTTT-3',

CXCL10 (F) 5'-CTAGAACTGTACGCTGTACC-3' (R) 5'-TTGATGGCCTTCGATTCTGG-3',CXCL10 (F) 5'-CTAGAACTGTACCGCTGTACC-3' (R) 5'-TTGATGGCCTTCGATTCTGG-3',

IFIT3 (F) 5'-AACTACGCCTGGGTCTACTATCACT-3' (R) 5'-ACACCTTCGCCCTTTCATTTC-3',IFIT3 (F) 5'-AACTACGCCTGGGTCTACTATCACT-3' (R) 5'-ACACCTTCGCCCTTTCATTTC-3',

IFIM (F) 5'-GGCTTCATAGCATTCGCCTACTC-3' (R) 5'-AGATGTTCAGGCACTTGGCGGT-3'IFIM (F) 5'-GGCTTCATAGCATTCGCCTACTC-3' (R) 5'-AGATGTTCAGGCACTTGGCGGT-3'

IRF7 (F) 5'-GCTGGACGTGACCATCATGTA-3' (R) 5'-GGGCCGTATAGGAACGTGC-3',IRF7 (F) 5'-GCTGGACGTGACCATCATGTA-3' (R) 5'-GGGCCGTATAGGAACGTGC-3',

OAS1 (F) 5'-AGGAAAGGTGCTTCCGAGGTAG-3' (R) 5'-GGACTGAGGAAGACAACCAGGT-3'OAS1 (F) 5'-AGGAAAGGTGCTTCCGAGGTAG-3' (R) 5'-GGACTGAGGAAGACAACCAGGT-3'

ISG15 (F) 5'-CGCAGATCACCCAGAAGATCG-3' (R) 5'-TTCGTCGCATTTGTCCACCA-3'ISG15 (F) 5'-CGCAGATCACCCAGAAGATCG-3' (R) 5'-TTCGTCGCATTTGTCCACCA-3'

STING 결합 어세이 STING binding assay

STING에 결합하고 있는 ligand와 화합물 간의 경쟁반응을 확인하기 위해 STING WT binding assay (CisBio/ NO. 64BDSTGPEG)를 사용하였다. 이후 제조 프로토콜을 사용하여 분석하였다.STING WT binding assay (CisBio/ NO. 64BDSTGPEG) was used to confirm the competitive reaction between the ligand and the compound binding to STING. It was then analyzed using the manufacturing protocol.

CYP (Cytochrome P450) 저해 어세이CYP (Cytochrome P450) inhibition assay

Potassium phosphate buffer로 희석시킨 마이크로솜에 시험 약물을 가해 37 ℃에서 5분간 배양시킨 후 NADPH를 5종의 substrate 물질을 가해 37 ℃에서 20분간 반응시켰다. (test compound final conc. : 10 μM, microsome final conc. : 0.2 mg/mL) 반응 종결을 위해 Internal standard가 포함된 cold acetonitrile을 가한 후 제단백 처리하였다. 원심분리 후 (4000 rpm, 4 ℃, 15 min) 상층액을 LC-MS/MS(분석기기: Mass sepctrometry (AB Sciex Qtrap 4000) with HPLC (Agilent 1260))로 분석하였다.The test drug was added to microsomes diluted with potassium phosphate buffer and incubated at 37°C for 5 minutes, then 5 kinds of substrate materials were added to NADPH and reacted at 37°C for 20 minutes. (test compound final conc.: 10 μM, microsome final conc.: 0.2 mg/mL) To terminate the reaction, cold acetonitrile containing internal standard was added, and then protein was treated. After centrifugation (4000 rpm, 4 °C, 15 min), the supernatant was analyzed by LC-MS/MS (analyzer: Mass sepctrometry (AB Sciex Qtrap 4000) with HPLC (Agilent 1260)).

hERG 패치 클램프 (patch clamp) 어세이 hERG patch clamp assay

액체질소에 동결된 hERG-HEK293 세포를 해동하여 DMEM/F-12 배지 (10% FBS, 7.5 mg/mLblasticidin S HCl, 50 mg/mL hygromycin B)를 이용하여 배양한 뒤, 배양한 세포를 trypsine-EDTA 용액으로 떼어낸 후, 60 mm cell culture dish에 2x105개의 세포를 포함하도록 분주하여 5% CO2 조건의 37 ℃ incubator에서 2일간 배양하였다. PatchXpress 7000A에 준비된 세포와 sealchip을 로딩하고, 세포 안착 및 저항안정화를 거친 뒤 MultiClamp 700A를 이용하여 Voltage-clamp와 data를 얻었다. hERG channel의 활성 측정과 데이터 분석은 tail currentsms DataXpress 2.0.4.5 (Axon Instruments)와 Clampfit 9.2 (Axon Instruments)를 이용하였다.Thaw hERG-HEK293 cells frozen in liquid nitrogen and culture them using DMEM/F-12 medium (10% FBS, 7.5 mg/mLblasticidin S HCl, 50 mg/mL hygromycin B), and then incubate the cultured cells with trypsine- After removing with EDTA solution, it was aliquoted to contain 2x10 5 cells in a 60 mm cell culture dish and cultured for 2 days in an incubator at 37 °C under 5% CO 2 condition. After loading the cells and sealchip prepared in PatchXpress 7000A, cell settling and resistance stabilization, Voltage-clamp and data were obtained using MultiClamp 700A. For measurement of hERG channel activity and data analysis, tail currentsms DataXpress 2.0.4.5 (Axon Instruments) and Clampfit 9.2 (Axon Instruments) were used.

혈장 안정도 테스트 Plasma Stability Test

혈장에 약물을 일정 시간 동안 노출시킨 후 잔존한 약물양을 LC-MS/MS로 정량하여 약물의혈장에서의 안정성을 측정하였고 rat과 human 혈장에서의 안정성이 보고된 enalapril 및 procaine을 reference 물질로 사용하였다. 혈장에 10 mM DMSO 약물 stock solution을 spiking하여 약물농도가 5 μM되도록 한 뒤, 혈장 샘플을 shaking incubator에서 반응시켰다(37℃, 4 h). 반응 종결을 위해 Internal standard가 포함된 cold acetonitrile을 가한 후 제단백 처리하고, 원심분리 후 (4000 rpm, 4℃, 15 min) 상층액을 LC-MS/MS(분석기기: Mass sepctrometry (Agilent 6460) with HPLC (Agilent 1260))로 분석하였다.After exposing the drug to plasma for a certain period of time, the amount of the drug remaining was quantified by LC-MS/MS to measure the stability of the drug in plasma, and enalapril and procaine, which have reported stability in rat and human plasma, were used as reference materials. did. After spiking a 10 mM DMSO drug stock solution into plasma so that the drug concentration was 5 μM, the plasma sample was reacted in a shaking incubator (37° C., 4 h). To terminate the reaction, cold acetonitrile containing internal standard was added, protein treatment, and centrifugation (4000 rpm, 4℃, 15 min), the supernatant was LC-MS/MS (analytical instrument: Mass sepctrometry (Agilent 6460)) with HPLC (Agilent 1260)).

Mouse 약동학 실험Mouse Pharmacokinetic Experiments

동물실험은 순화기간을 거친 7-8 주령 mouse에 약물을 투여한 후 정해진 시간에 채혈하였다. 시료 전처리를 위하여 혈액은 원심분리하여 혈장을 분리하고 Internal standard가 포함된 cold acetonitrile 9배 용량을 가한 후 제단백 처리하였으며, 원심분리 후 (13000 rpm, 4℃, 10 min) 상층액을 LC-MS/MS(분석기기: Mass sepctrometry (Agilent 6460) with HPLC (Agilent 1260))로 분석하였다. PK 파라미터 분석은 Phoenix WinNonlin (Pharsight ver 6.4, USA) Non-compartmental analysis 모델을 이용하여 산출하였다.In the animal experiment, the drug was administered to 7-8 week old mice that had undergone the acclimatization period, and blood was collected at a set time. For sample pretreatment, blood was centrifuged to separate plasma, 9 times the volume of cold acetonitrile containing internal standard was added, and then protein was treated. After centrifugation (13000 rpm, 4℃, 10 min), the supernatant was LC-MS /MS (analysis instrument: Mass sepctrometry (Agilent 6460) with HPLC (Agilent 1260)) was analyzed. PK parameter analysis was calculated using the Phoenix WinNonlin (Pharsight ver 6.4, USA) Non-compartmental analysis model.

구조-활성 상관관계(Structure-Activity Relationship; SAR) 연구 결과 Structure-Activity Relationship (SAR) Study Results

합성된 화합물의 생물학적 활성을 평가하기 위해, 인터페론 민감성 면역 반응 요소(ISRE) 리포터로 조작된 THP-1(인간 단핵구)을 사용하여 루시퍼라제 유전자 발현을 측정하였다. STING 의존성 면역 반응을 검증하기 위해, 화합물의 활성을 야생형(WT) 및 STING 녹아웃(KO) 세포 모두에서 테스트하였다. 개별 화합물에 대한 면역 반응에 대한 리포터 분석에서는 먼저 단일 농도(50 μM)에서 실험한 결과에서 정규화된 배수 변화 값을 비교하는 방식으로 활성을 평가하였다. 이후 활성이 나타난 화합물에 대하여 용량-반응 분석을 통하여 EC50 값을 구하여 그 결과를 하기 표 3, 4, 및 5에 나타내었다.To evaluate the biological activity of the synthesized compounds, luciferase gene expression was measured using THP-1 (human monocytes) engineered as an interferon sensitive immune response element (ISRE) reporter. To validate the STING-dependent immune response, the activity of the compounds was tested in both wild-type (WT) and STING knockout (KO) cells. In the reporter assay for immune responses to individual compounds, activity was first evaluated by comparing normalized fold change values from the experimental results at a single concentration (50 μM). Thereafter, EC 50 values were obtained through dose-response analysis for compounds exhibiting activity, and the results are shown in Tables 3, 4, and 5 below.

Figure pat00069
Figure pat00069

Figure pat00070
Figure pat00070

표 3-5에 있어, 루시페라제 값 a 은 상대적 루시페라제 유닛의 평균 ± 표준 편차로 나타내었다. THP-1 WT 및 STING KO 세포들은 개별적인 화합물 (50 μM)로 처리되었다. b EC50 값들은 THP-1 WT 세포들의 용량-반응 곡선(dose-response curve)을 이용하여 측정되었다. N/A는 not applicable을 의미한다. N/A로 표시된 화합물들은 최대 50 μM의 낮은 배수 변화 값(fold-change value)으로 인해 EC50 값의 계산 대상이 아니었다. In Table 3-5, the luciferase value a is expressed as the mean±standard deviation of the relative luciferase units. THP-1 WT and STING KO cells were treated with individual compounds (50 μM). b EC 50 values were determined using a dose-response curve of THP-1 WT cells. N/A means not applicable. Compounds marked with N/A were not subject to calculation of EC 50 values due to their low fold-change values of up to 50 μM.

표 3에서 R2를 (S)-2-hydroxy-phenethyl 그룹으로 유지하면서 화합물 1의 R1 그룹을 변화시키면서 STING 작용 활성 및 세포 생존성을 확인하였고, 동일 실험 조건에서의 cGAMP 효능을 비교하였다(화합물 2-7과 cGAMP). 테스트한 모든 화합물에 대하여 동일 농도에서의 세포 생존성 검사 결과 세포독성을 보이지 않았다. 트리플루오로메틸-치환된 피라졸(화합물 2 및 3), 퓨란(화합물 4), 벤조퓨란(화합물 5) 및 인다졸(화합물 6 및 7)을 포함한 다양한 헤테로고리를 조사했지만 STING 효능작용에 뚜렷한 효과를 나타내지 않았다.In Table 3, STING action activity and cell viability were confirmed by changing the R 1 group of Compound 1 while maintaining R 2 as a (S)-2-hydroxy-phenethyl group, and cGAMP efficacy under the same experimental conditions was compared ( compounds 2-7 and cGAMP). All tested compounds showed no cytotoxicity as a result of cell viability at the same concentration. Various heterocycles were investigated, including trifluoromethyl-substituted pyrazoles (Compounds 2 and 3), furan (Compound 4), benzofuran (Compound 5) and indazole (Compounds 6 and 7), but not distinct in STING agonism. showed no effect.

화합물 1의 R2 치환체가 (S)-2-하이드록시-페네틸 그룹에서 tert-부틸 부틸카바메이트 그룹으로 대체되었을 때, 생성된 화합물 8는 ISRE 리포터 분석에서 개선된 효능을 나타냈다(화합물 1 및 8). 용량 의존적 효능 분석은 화합물 8(EC50=6.5μM)의 활성이 화합물 1(EC50=102μM) 및 cGAMP(EC50=30.1μM)의 활성보다 각각 약 15배 및 5배 더 높은 것으로 나타났다. When the R 2 substituent of compound 1 was replaced with a tert-butyl butylcarbamate group in the (S)-2-hydroxy-phenethyl group, the resulting compound 8 showed improved efficacy in the ISRE reporter assay (compound 1 and 8). Dose dependent efficacy analysis showed that the activity of compound 8 (EC 50 =6.5 μM) was approximately 15-fold and 5-fold higher than that of compound 1 (EC 50 =102 μM) and cGAMP (EC 50 =30.1 μM), respectively.

또한 R2 그룹으로서 더 효과적인 tert-부틸 부틸카바메이트의 존재 하에 R1 그룹을 변화시켜 화합물의 활성 및 세포생존성을 조사했다. 트리플루오로메틸 치환된 피라졸(화합물 9 및 10), 1-알릴-5-메틸-1H-피라졸(화합물 11), 퓨란(화합물 12) 및 벤조퓨란(화합물 13)을 포함하는 유도체를 합성하였지만 유의미한 활성 결과는 관찰되지 않았다. 피라졸(pyrazole)의 N1 위치에서 다른 치환체를 조사한 결과 에틸 그룹이 가장 강력한 활성을 제공하는 반면 메틸, 이소프로필 및 tert-부틸 그룹은 STING 반응에 해로운 영향을 미치는 것으로 나타났다(화합물 8 및 14-16).In addition, the activity and cell viability of the compound were investigated by changing the R 1 group in the presence of tert-butyl butylcarbamate, which is more effective as the R 2 group. Synthesis of derivatives comprising trifluoromethyl substituted pyrazoles (compounds 9 and 10), 1-allyl-5-methyl-1H-pyrazole (compound 11), furan (compound 12) and benzofuran (compound 13) However, no significant activity results were observed. Examination of other substituents at the N1 position of pyrazole showed that methyl, isopropyl and tert-butyl groups had a detrimental effect on the STING reaction, while the ethyl group provided the most potent activity (compounds 8 and 14-16). ).

Figure pat00071
Figure pat00071

Figure pat00072
Figure pat00072

표 4에서는 추가적인 구조-활성 관계(SAR) 연구를 통해 benzimidazole 하위 구성요소를 변경하여 화합물 8의 최적화 연구를 수행하였다. R2 그룹의 알킬 사슬 길이의 경우 부틸카바메이트(화합물 8)가 해당 프로필카바메이트(화합물 17)및 펜틸카바메이트(화합물 18)보다 더 강력하고 세포독성이 적었다. R2의 아미노 치환기를 변화시켜 보았을 경우, tert-부틸 카바메이트 그룹을 수소 원자로 바꾸자 화합물 19는 활성을 나타내지 않았다. 화합물 20 및 21보다 화합물 22와 같은 방향족 아미드 화합물에서 높은 활성을 확인하였고, 헤테로방향족 치환기를 아미드 치환기로 갖는 화합물 23과 24의 경우 리포터 어세이 활성이 관측되지 않았다. 반면 피라졸의 질소 원자를 알킬 그룹으로 치환하자 활성을 현저하게 향상시켰다. N-메틸-3-메틸 피라졸이 치환된 화합물 25보다 N-에틸-3-메틸 피라졸이 치환된 화합물 26의 경우 훨씬 향상된 활성을 나타내었고, 화합물 27 및 28과 비교하여 화합물 26의 사슬길이가 더 나은 STING 효능제 활성 및 세포 생존 결과를 나타내는 것을 확인하였다. R3 위치에 도입된 메톡시기도 효능에 영향을 미치는 것을 확인하여 화합물 29는 서브마이크로몰 EC50을 보였고, 화합물 30은 29에 비하여 루시퍼라제 활성과 세포 생존력이 회복됨을 확인함으로써 회전 가능한 결합의 수를 줄임으로써 활성 증진에 영향을 주었음을 보였다.In Table 4, an optimization study of compound 8 was performed by modifying the benzimidazole subcomponent through additional structure-activity relationship (SAR) studies. For the alkyl chain length of the R 2 group, butyl carbamate (compound 8) was more potent and less cytotoxic than the corresponding propyl carbamate (compound 17) and pentyl carbamate (compound 18). When the amino substituent of R 2 was changed, compound 19 showed no activity when the tert-butyl carbamate group was replaced with a hydrogen atom. Higher activity was confirmed in the aromatic amide compound such as compound 22 than in compounds 20 and 21, and in the case of compounds 23 and 24 having a heteroaromatic substituent as an amide substituent, reporter assay activity was not observed. On the other hand, when the nitrogen atom of pyrazole was replaced with an alkyl group, the activity was significantly improved. Compound 26 substituted with N-ethyl-3-methyl pyrazole showed significantly improved activity than compound 25 substituted with N-methyl-3-methyl pyrazole, and the chain length of compound 26 compared to compounds 27 and 28 showed better STING agonist activity and cell survival results. By confirming that the methoxy group introduced at the R 3 position also affects the efficacy, compound 29 showed submicromolar EC 50 , and compound 30 compared to 29 restored luciferase activity and cell viability. The number of rotatable bonds was shown to have an effect on activity enhancement by reducing

Figure pat00073
Figure pat00073

표 5에서는 STING의 이량체 구조에 보다 더 잘 결합할 수 있을 것으로 예상되는 이량체 화합물의 활성을 조사하였다. 화합물 31의 증가된 효능을 관찰한 다음, 다른 종류의 헤테로고리화 카복시 그룹으로 R1 그룹이 치환된 화합물 32-34들에 대해 효능을 조사하였다. 화합물 32와 33은 상응하는 단량체 화합물에 비하여 향상 개선을 관찰하지 못하였고, 화합물 34는 상응하는 단량체 화합물에 비하여 활성이 약간 향상되었다. R4 위치의 아미드 작용기를 에스테르와 카복시산으로 변화시킨 화합물 35와 36의 경우에 전혀 효능이 나타나지 않았다. In Table 5, the activity of the dimer compound expected to be better able to bind to the dimer structure of STING was investigated. After observing the increased efficacy of compound 31, the efficacy of compounds 32-34 in which the R 1 group was substituted with another type of heterocyclic carboxy group was investigated. Compounds 32 and 33 did not observe improvement in enhancement compared to the corresponding monomeric compounds, and compound 34 showed a slight improvement in activity compared to the corresponding monomeric compounds. Compounds 35 and 36, in which the amide functional group at the R 4 position was changed to an ester and a carboxylic acid, showed no efficacy at all.

상기 표 3-5에 요약된 정보를 기반으로 하기 화학식 1의 화합물들을 고안하였고, 이들의 효과 평가 결과를 표 6와 도 1에 종합하여 나타내었다. Based on the information summarized in Table 3-5, the compounds of Formula 1 below were devised, and the results of evaluation of their effects are summarized in Table 6 and FIG. 1 .

Figure pat00074
Figure pat00074

상기 화학식 1에서, R5

Figure pat00075
,
Figure pat00076
, 또는
Figure pat00077
임. In Formula 1, R 5 is
Figure pat00075
,
Figure pat00076
, or
Figure pat00077
lim.

Figure pat00078
Figure pat00078

표 6에 있어, 루시페라제 값 a 은 상대적 루시페라제 유닛의 평균 ± 표준 편차로 나타내었다. THP-1 WT 및 STING KO 세포들은 개별적인 화합물 (1 μM)로 처리되었다.In Table 6, luciferase values a are expressed as mean±standard deviation of relative luciferase units. THP-1 WT and STING KO cells were treated with individual compounds (1 μM).

3개의 합성된 화합물 37-39는 리포터 분석에서 포화 수준에 도달한 반면, 화합물 diABZI-3은 더 낮은 리포터 반응을 나타냈다. 낮은 농도에서 용량-반응 측정을 수행한 결과 화합물 37의 EC50은 0.36 nM, 화합물 38의 EC50은 1.62 nM로 매우 좋은 효능을 보였으며, 화합물 39의 경우 4.0 pM의 EC50을 나타냄으로써 STING 매개 활성이 극적으로 증가함을 확인할 수 있었다. 또한 같은 농도 범위에서 본 발명화합물을 처리하였을 시 STING KO 세포주에서는 전혀 활성을 보이지 않음을 확인하여 본 발명 화합물이 STING 의존적으로 면역반응을 유도하는 것을 검증하였다. The three synthesized compounds 37-39 reached saturation levels in the reporter assay, whereas compound diABZI-3 showed a lower reporter response. As a result of performing the dose-response measurement at a low concentration, the EC 50 of compound 37 was 0.36 nM and the EC 50 of compound 38 was 1.62 nM, showing very good efficacy, and in the case of compound 39, the EC 50 of 4.0 pM was shown to mediate STING. It was confirmed that the activity increased dramatically. In addition, when the compound of the present invention was treated in the same concentration range, it was confirmed that no activity was shown in the STING KO cell line, thereby verifying that the compound of the present invention induces an immune response in a STING-dependent manner.

MTS 어세이를 통해 세포생존률을 확인하였으며, 그 결과를 도 2에 나타내었다. 도 2에 나타나는 바와 같이, 본 발명 화합물 37및 39를 400 μM 및 10,000 nM 범위의 고농도까지 처리하였을 시에도 세포에서 전혀 독성을 나타내지 않았다. 이러한 결과와 THP1 세포에서 화합물 37 및 39의 EC50가 약 356.3 pM 및 4.0 pM임을 고려하였을 시 본 발명 화합물들의 safety window가 매우 큼을 확인하였다.Cell viability was confirmed through the MTS assay, and the results are shown in FIG. 2 . As shown in FIG. 2 , even when the compounds 37 and 39 of the present invention were treated up to high concentrations ranging from 400 μM and 10,000 nM, they did not show any toxicity in cells. Considering these results and the EC 50 of compounds 37 and 39 in THP1 cells were about 356.3 pM and 4.0 pM, it was confirmed that the safety window of the compounds of the present invention was very large.

화합물 37의 STING단백질 결합 평가를 위하여 인간 STING 재조합 단백질을 사용하여 시험관 내 리간드-경쟁 결합 실험을 수행하여 도 3에 나타내었다. d2-표지된 인간 STING 리간드와 Tb3+-표지된 STING 항체 사이의 결합에 의해 생성된 FRET 신호를 측정하여 화합물 37이 화합물 31보다 STING에 대해 100배 더 큰 결합 친화도를 가지는 것을 확인하였다. HTRF(homogeneous time-resolved fluorescence) 비율로 계산된 화합물 37의 IC50 값은 0.06nM로 화합물 31의 경우 (6.2nM)에 비하여 약 100배 이상 더 강력하게 결합하는 양상을 확인하였다. In order to evaluate the binding of compound 37 to the STING protein, an in vitro ligand-competition binding experiment was performed using human STING recombinant protein and is shown in FIG. 3 . By measuring the FRET signal generated by the binding between the d2-labeled human STING ligand and the Tb3+-labeled STING antibody, it was confirmed that compound 37 has a 100-fold greater binding affinity for STING than compound 31. The IC 50 value of compound 37 calculated by the HTRF (homogeneous time-resolved fluorescence) ratio was 0.06 nM, confirming the binding pattern about 100 times more strongly than that of compound 31 (6.2 nM).

화합물 37이 인간과 마우스 세포 모두에서 STING에 결합할 수 있는지 확인하였다. CETSA(cellular thermal shift assay)를 이용하여 화합물 37이 결합할 때 STING 단백질의 열역학적 안정성의 변화를 평가하여 결과를 도 4에 나타냈다. CETSA 결과를 통하여 화합물 37이 인간 및 마우스 STING에 결합한다는 것을 확인했다. 화합물 37이 없는 경우 STING의 열 안정성은 온도 증가에 따라 감소되었지만 THP-1 및 RAW264.7 세포를 화합물 37과 함께 배양하면 단백질의 안정성이 증가했다. 이는 단백질과 화합물 사이의 강한 상호작용으로 인한 결과로 사료된다.It was confirmed that compound 37 could bind to STING in both human and mouse cells. CETSA (cellular thermal shift assay) was used to evaluate the change in thermodynamic stability of the STING protein when compound 37 was bound, and the results are shown in FIG. 4 . The results of CETSA confirmed that compound 37 binds to human and mouse STING. In the absence of compound 37, the thermal stability of STING decreased with increasing temperature, but incubation of THP-1 and RAW264.7 cells with compound 37 increased the stability of the protein. This is thought to be the result of the strong interaction between the protein and the compound.

다음으로 본 발명 화합물 37과 39의 선천면역반응 유도 효능을 검증하기 위하여 THP-1 세포주에서 STING 활성화에 의한 하위 경로 중 가장 주요한 인자인 IFN-β과 type I IFN 경로의 하위 인자인 IP-10 사이토카인 분비 정도를 효소흡착면역흡착 분석법(ELISA)를 이용하여 측정하였다. 그 결과를 도 5와 도 6에 나타내었다. Next, in order to verify the efficacy of inducing innate immune response of compounds 37 and 39 of the present invention, IFN-β, which is the most important factor among the sub-pathways by STING activation in THP-1 cell line, and IP-10 cytokine, which is a sub-factor of the type I IFN pathway. The degree of Cain secretion was measured using an enzyme adsorption immunosorbent assay (ELISA). The results are shown in FIGS. 5 and 6 .

본 발명 화합물 37 처리 시, 화합물 31에서보다 훨씬 더 낮은 농도인 12.8 nM 에서부터 STING 활성화에 의한 IFN-β 의 분비를 확인할 수 있었으며 IFN-β 분비에 대한 EC50값은 2.047nM로 기존 물질에 비해 매우 우수함을 증명하였다. 뿐만 아니라 하위 인자인 IP-10 또한 화합물 31은 500 nM 에서도 분비가 미미한 반면, 본 발명 화합물37은 0.41 nM의 매우 저농도에서부터 의존적으로 증가하는 양상을 확인할 수 있었다. 이와 같은 양상은 인간 및 마우스 세포에서 모두 용량 의존적 방식으로 사이토카인 분비가 관찰되었고, 이를 통해 화합물 37의 효능이 매우 우수함을 보였다. 또한 THP1 WT 세포주와 STING KO 세포주에서 본 발명 화합물 37 처리에 따른 IFN-β 생성을 ELISA로 측정한 결과, STING KO 세포에서 본 발명 화합물 37에 의한 IFN-β 분비는 전혀 관찰되지 않았다. 이 결과를 통해 본 발명 화합물 37이 STING 의존적으로 IFN-β 분비에 따른 선천면역 반응을 조절하는 것을 다시 한 번 검증하였다. Upon treatment with compound 37 of the present invention, secretion of IFN-β by STING activation was confirmed from 12.8 nM, which is a much lower concentration than in compound 31, and the EC 50 value for IFN-β secretion was 2.047 nM, which was very high compared to the existing material. proved to be excellent. In addition, while the sub-factor IP-10 and compound 31 were insignificant even at 500 nM, it was confirmed that the compound 37 of the present invention increased dependently from a very low concentration of 0.41 nM. In this aspect, cytokine secretion was observed in both human and mouse cells in a dose-dependent manner, indicating that the efficacy of Compound 37 was very good. In addition, as a result of ELISA measurement of IFN-β production according to the compound 37 treatment of the present invention in the THP1 WT cell line and the STING KO cell line, IFN-β secretion by the compound 37 of the present invention in the STING KO cells was not observed at all. Through these results, it was verified once again that compound 37 of the present invention regulates the innate immune response according to IFN-β secretion in a STING-dependent manner.

도 6에서는 기 보고된 바 있는 STING agonist인 diABZI-3과 화합물 39의 효능을 비교 분석하였다. 분비된 IFN-β 및 IP-10의 용량 의존적 수준에 대한 ELISA 측정은 화합물 39가 diABZI-3에 비해 THP-1 세포에서 사이토카인 분비를 크게 촉진하였다. In FIG. 6, the efficacy of diABZI-3 and compound 39, which is a previously reported STING agonist, was compared and analyzed. ELISA measurements of dose-dependent levels of secreted IFN-β and IP-10 showed that compound 39 significantly promoted cytokine secretion in THP-1 cells compared to diABZI-3.

다음으로 본 발명 화합물 37에 의해 유도되는 type 1 Interferon 경로 활성화에 따른 다양한 ISG (interferon stimulated gene)의 발현 여부를 검증하였으며 그 결과를 도 7에 나타내었다. 이를 위하여 THP1 세포주에서 RT-PCR(real-time Polymerase Chain Reaction)을 진행하였다. Next, the expression of various interferon stimulated genes (ISG) according to the activation of the type 1 interferon pathway induced by compound 37 of the present invention was verified, and the results are shown in FIG. 7 . To this end, RT-PCR (real-time polymerase chain reaction) was performed in the THP1 cell line.

도 7의 ELISA 결과에서 확인한 바와 같이, IFNB, CXCL10의 유전자가 각각 6시간, 18시간에서 유의미하게 증가하는 것을 확인하였다. 특히 type I IFN 중 하나인 IFNB는 6시간 내 급격한 증가를 보이는 데에 비해, type I IFN에 의한 하위인자인 ISG 유전자들은 6시간에 비해 18시간에 유사하거나 더 높은 발현을 보이는 것을 확인하였다. 이를 바탕으로 본 발명 화합물 37이 효과적으로 IFNB에 의한 type I IFN 신호 전달 체계를 활성화시켜 선천면역 반응을 유도함을 검증하였다.As confirmed in the ELISA result of FIG. 7 , it was confirmed that the genes of IFNB and CXCL10 significantly increased at 6 hours and 18 hours, respectively. In particular, it was confirmed that IFNB, one of the type I IFNs, showed a sharp increase within 6 hours, whereas the ISG genes, a subfactor by type I IFN, showed similar or higher expression at 18 hours compared to 6 hours. Based on this, it was verified that compound 37 of the present invention effectively activates the type I IFN signal transduction system by IFNB to induce an innate immune response.

STING이 활성화 되면 STING 자체의 인산화 및 하위 인자인 TBK1, IRF3의 인산화를 통해 신호체계의 활성화가 일어나게 된다. 본 발명 화합물 37이 STING 신호체계 활성에 작용하는지 확인하기 위해, Western Blotting을 통해 주요 인자의 발현양 및 인산화 여부를 확인하였다. 효능 비교를 위하여 화합물 31을 비교군으로 함께 검증하였다. 그 결과를 도 8에 나타내었다. When STING is activated, activation of the signaling system occurs through phosphorylation of STING itself and phosphorylation of sub-factors TBK1 and IRF3. In order to confirm whether the compound 37 of the present invention acts on the STING signaling system activity, the expression level and phosphorylation of major factors was confirmed through Western Blotting. For efficacy comparison, compound 31 was verified together as a control group. The results are shown in FIG. 8 .

화합물 31은 1000 nM을 처리하였음에도 불구하고 p-STAT1, pTBK1, pIRF3, pSTING 등 하위 인자의 인산화가 유도되지 않은 반면, 본 발명 화합물 37의 경우 매우 낮은 농도인 2nM과 10nM 에서 모두 pSTAT1, pTBK1, pIRF3, pSTING이 관찰되었다. 다음과 같은 결과로 미루어 보아, 본 발명 화합물 37은 매우 낮은 농도에서도 STING 신호체계의 하위인자들을 활성화 시키는 것을 검증하였으며 이를 통해 효과적인 STING 작용제로서 작용함을 증명하였다.Although compound 31 was treated with 1000 nM, phosphorylation of sub-factors such as p-STAT1, pTBK1, pIRF3, and pSTING was not induced, whereas in the case of compound 37 of the present invention, both pSTAT1, pTBK1, and pIRF3 at very low concentrations of 2 nM and 10 nM. , pSTING was observed. Judging from the following results, it was verified that the compound 37 of the present invention activates sub-factors of the STING signaling system even at very low concentrations, thereby proving that it acts as an effective STING agonist.

한편, 본 발명 화합물 37의 독성과 안정성을 평가하여 그 결과를 하기 표 7에 나타내었다. Meanwhile, the toxicity and stability of compound 37 of the present invention were evaluated, and the results are shown in Table 7 below.

(means ± SD; n = 3)(means ± SD; n = 3) 본 발명 화합물 37compound 37 of the present invention ■ CYP inhibition (IC50, μM) a ■ CYP inhibition (IC 50 , μM) a 1A21A2 >100.0>100.0 2C92C9 >100.0>100.0 2C192C19 >100.0>100.0 2D62D6 >100.0>100.0 3A43A4 4.24.2 ■ Cardiotoxicity (IC50, μM)■ Cardiotoxicity (IC 50 , μM) hERG patch clamp assayhERG patch clamp assay > 50.0> 50.0 ■ Plasma stability c ■ Plasma stability c Mouse (%)Mouse (%) >99>99 Human (%)Human (%) >99>99

구체적으로 먼저 사람 간 마이크로솜에서의 CYP(cytochrome P450) 저해에 대한 IC50 값을 측정하였다. CYP 대사효소 활성 저해 정도를 측정하기 위하여 사람 간 마이크로솜에 각 CYP 특이적인 기질을 가했을 때 생성된 대사체의 양을 정량하였다. 이를 통해 시험 약물에 의한 특정 대사체 생성이 억제될 경우 해당 대사에 관여하는 CYP 활성을 저해한다고 판단하였다. 총 5 가지의 CYP 아이소자임에 대한 효소 활성저해를 측정한 결과, CYP3A4에 대한 저해가 확인되었으나 알려진 CYP3A4 저해제(ketoconazole)에 비하여 약한 저해를 보였으며, 이를 제외한 나머지 CYP1A2, CYP1C9, CYP2C19 및 CYP2D6에 대하여는 본 발명 화합물에 의한 저해가 전혀 관측되지 않음을 확인하였다. CYP3A4의 경우도, 본 발명 화합물 37의 EC50이 356.3 pM임을 고려하였을 때 11878배 이상의 차이를 나타내므로 효소활성 저해에 큰 영향을 미치지 않을 것으로 사료된다.Specifically, first, IC 50 values for CYP (cytochrome P450) inhibition in human liver microsomes were measured. To measure the degree of inhibition of CYP metabolizing enzyme activity, the amount of metabolites produced when each CYP-specific substrate was added to human liver microsomes was quantified. Through this, it was determined that when the production of a specific metabolite by the test drug was inhibited, the CYP activity involved in the corresponding metabolism was inhibited. As a result of measuring the inhibition of enzyme activity for a total of 5 CYP isozymes, inhibition of CYP3A4 was confirmed, but weak inhibition compared to known CYP3A4 inhibitors (ketoconazole). It was confirmed that no inhibition by the compound of the present invention was observed. In the case of CYP3A4, considering that the EC50 of compound 37 of the present invention is 356.3 pM, it is thought that it does not significantly affect the inhibition of enzyme activity because it shows a difference of 11878 times or more.

다음으로 전기생리학적 방법인 patch clamp법을 이용하여 약물에 의한 hERG K+ channel의 저해에 의한 생체 내 심장독성 유발 가능성을 예측하였다. 본 발명 화합물 37은 시험 최고 농도인 50 μM 이내에서 IC50를 나타내지 않음으로써 hERG K+ channel의 활성저해에 의한 심장독성 유발 가능성에 대하여 안전한 것으로 확인하였다.Next, by using the electrophysiological patch clamp method, the possibility of inducing cardiotoxicity in vivo by drug-induced inhibition of the hERG K + channel was predicted. Compound 37 of the present invention was confirmed to be safe against the possibility of cardiotoxicity caused by inhibition of hERG K+ channel activity by not showing IC 50 within 50 μM, the highest concentration tested.

반면 혈장 안정성 테스트 실험에서는 쥐와 사람의 혈장에 약물을 4시간 노출시킨 후 잔존하는 본 발명 화합물의 양을 측정한 결과 99% 이상의 화합물이 남아있었다. 따라서 혈장 내에 존재하는 가수분해 효소들에 대하여는 화합물이 매우 안정함을 확인하였다.On the other hand, in the plasma stability test experiment, as a result of measuring the amount of the compound of the present invention remaining after exposure of the drug to plasma of rats and humans for 4 hours, 99% or more of the compound remained. Therefore, it was confirmed that the compound is very stable against hydrolytic enzymes present in plasma.

한편, 본 발명 화합물의 약동학적 특성을 평가하여 하기 표 8에 나타내었다. Meanwhile, the pharmacokinetic properties of the compounds of the present invention were evaluated and shown in Table 8 below.

Parameter a,b Parameter a, b Compound 26Compound 26 Compound 37Compound 37 Compound 39Compound 39 T1/2 (h)T 1/2 (h) 0.59 ± 0.400.59 ± 0.40 10.54 ± 4.1010.54 ± 4.10 1.75 ± 0.131.75 ± 0.13 Vss (L/kg)V ss (L/kg) 1.80 ± 0.741.80 ± 0.74 17.74 ± 5.2917.74 ± 5.29 1.81 ± 0.341.81 ± 0.34 CL (L/h/kg)CL (L/h/kg) 9.48 ± 1.119.48 ± 1.11 2.12 ± 0.272.12 ± 0.27 0.72 ± 0.180.72 ± 0.18 AUClast (μg·h/mL)AUC last (μg h/mL) 0.31 ± 0.030.31 ± 0.03 4.20 ± 0.264.20 ± 0.26 4.24 ± 0.964.24 ± 0.96 AUC∞ (μg·h/mL)AUC∞ (μg h/mL) 0.32 ± 0.040.32 ± 0.04 4.78 ± 0.594.78 ± 0.59 4.30 ± 0.974.30 ± 0.97

a Values are shown as the means ± standard deviation of at least three independent experiments. b Parameters from intravenous administration (3 mg/kg, BALB/c mice, n=3). AUClast, areas under the plasma concentration-time curve from time zero to time of last measurable concentration; AUC, areas under the plasma concentration-time curve from time zero to time infinity; CL, total clearance from plasma steady; T1/2, terminal half-life; Vss, state volume of distribution. a Values are shown as the means ± standard deviation of at least three independent experiments. b Parameters from intravenous administration (3 mg/kg, BALB/c mice, n=3). AUC last , areas under the plasma concentration-time curve from time zero to time of last measurable concentration; AUC , areas under the plasma concentration-time curve from time zero to time infinity; CL, total clearance from plasma steady; T 1/2 , terminal half-life; V ss , state volume of distribution.

구체적으로, BALB/c 쥐에 본 발명 화합물을 3 mg/kg IV 투여하여 시간에 따른 혈액 내 화합물 농도변화를 측정하였다. 화합물 26의 반감기가 1시간이 되지 않는 것에 비하여 본 발명 화합물 39의 반감기는 1.75시간, 화합물 37의 반감기는 10.5 시간으로 유사한 구조를 다진 다른 화합물들 대비 반감기에 큰 차이가 있고, 이러한 차이는 의약품의 유효 성분으로서 큰 장점이 될 수 있다. Specifically, 3 mg/kg IV of the compound of the present invention was administered to BALB/c mice to measure the change in the concentration of the compound in the blood over time. Compared to the half-life of compound 26, less than 1 hour, the half-life of compound 39 of the present invention is 1.75 hours, and the half-life of compound 37 is 10.5 hours. It can be a great advantage as an active ingredient.

한편 화합물 37의 AUClast는 4.20 μg·h/ml, AUC는 4.78 μg·h/ml, 화합물 39의 AUClast는 4.24 μg·h/ml, AUC는 4.30 μg·h/ml로 확인되었으며, 이 역시 유사한 구조의 다른 화합물들 대비 높게 나왔다. 마찬가지로 clearance는 화합물 37이 2.12 L/h/kg, 화합물 39가 0.72 L/h/kg로 단량체 화합물 26에 비하여도 매우 낮게 나타났으며, volume of distribution역시 화합물 37에서 17.7 L/kg으로, 유사한 구조를 가지는 다른 화합물들 대비 높게 나타나 체내대사안정성에서 본 발명 화합물이 좋은 장점을 가지는 것을 확인하였다.On the other hand, AUC last of compound 37 was 4.20 μg h/ml, AUC was 4.78 μg h/ml, AUC last of compound 39 was 4.24 μg h/ml, AUC was 4.30 μg h/ml, This was also higher than other compounds with similar structures. Similarly, clearance of compound 37 was 2.12 L/h/kg and compound 39 was 0.72 L/h/kg, which was very low compared to monomeric compound 26, and the volume of distribution was also 17.7 L/kg in compound 37, similar structure It was found to be higher than other compounds having

전체적으로 본 발명의 화합물들은 유사 구조의 화합물들에 비하여 반감기가 길고, clearance가 낮은 편으로 AUC 값이 좋다고 볼 수 있다. Overall, the compounds of the present invention have a longer half-life and a lower clearance than those of a similar structure, and thus it can be seen that the AUC value is good.

도 9에서는 마우스 CT26 종양 모델에서 종양 성장에 대한 화합물 37의 효과를 확인하였다. 종양이 50-100 mm3에 도달했을 때, 마우스는 화합물 37 두 가지 다른 용량(0.015 및 1.5 mg/kg)으로 네 번 투여했다. 두 가지 용량에서 화합물 37로 처리하여 마우스 체중 감소가 거의 나타나지 않았고, 종양 성장이 유의하게 억제되었다(1.5mg/kg의 경우 P=0.018, 0.015mg/kg의 경우 P=0.045). 화합물 37(1.5 mg/kg)은 17일째에 마우스에서 57% 종양 성장 억제를 유발하여 STING 관련 신호전달 경로의 활성화에 의해 매개되는 생체내 화합물의 항암 효능을 확인하였다.In Figure 9, the effect of compound 37 on tumor growth in the mouse CT26 tumor model was confirmed. When tumors reached 50-100 mm 3 , mice were dosed with compound 37 four times at two different doses (0.015 and 1.5 mg/kg). Treatment with Compound 37 at both doses showed little weight loss in mice and significantly inhibited tumor growth (P=0.018 for 1.5 mg/kg, P=0.045 for 0.015 mg/kg). Compound 37 (1.5 mg/kg) induced 57% tumor growth inhibition in mice on day 17, confirming the anticancer efficacy of the compound in vivo mediated by activation of STING-related signaling pathways.

높은 효능과 우수한 PK 특성으로 인해 화합물 26, 37, 및 39를 가지고 CT26 보유 마우스에서 항암 효과에 대해 테스트하였다. STING의 자연리간드인 cGAMP의 효과는 종양 내 주사를 통해 활용되어 왔지만, 종 모양의 반응-효능 곡선으로 인해 낮은 용량에서만 원하는 항암 효과가 관찰되기 때문에 cGAMP의 용량 조절이 어려운 문제가 있다. 따라서 본 발명에서는 화합물 26, 37 및 39를 각각 정맥투여하여 마우스 항암효능을 확인해보았다. 투여한 양은 각 화합물에 대해 1.5 mg/kg이었다. 초기 실험한 화합물 37의 생체 내 평가(도 9)를 기반으로 충분한 면역 자극을 달성하기 위해 투여 횟수를 늘리고 8일 후에 치료를 중단했으며 결과는 도 10에 나타내었다. 세 가지 화합물 26, 37, 및 39는 체중 감소를 일으키지 않고 종양 성장을 억제했다(화합물 39의 경우 P=0.0037, 화합물 37의 경우 P=0.0188). 또한 화합물 39를 처리한 5마리의 마우스 중 1마리는 종양이 완전히 사라지면서 종양이 없는 상태에 도달했고 2마리의 마우스는 매우 더딘 종양 성장을 보였다. Compounds 26, 37, and 39 were tested for anticancer effects in CT26 bearing mice due to their high potency and good PK properties. Although the effect of cGAMP, a natural ligand of STING, has been utilized through intratumoral injection, there is a problem in that it is difficult to control the dose of cGAMP because the desired anticancer effect is observed only at low doses due to the bell-shaped response-efficiency curve. Therefore, in the present invention, compounds 26, 37 and 39 were each administered intravenously to confirm the anticancer effect in mice. The dose administered was 1.5 mg/kg for each compound. Based on the in vivo evaluation of the initially tested compound 37 ( FIG. 9 ), the number of administrations was increased to achieve sufficient immune stimulation, and treatment was stopped after 8 days, and the results are shown in FIG. 10 . Three compounds 26, 37, and 39 inhibited tumor growth without causing weight loss (P=0.0037 for compound 39, P=0.0188 for compound 37). Additionally, 1 out of 5 mice treated with compound 39 reached a tumor-free state with complete disappearance of tumors and 2 mice showed very slow tumor growth.

이후 화합물 39를 투여하였던 마우스에 화합물의 추가 투여없이 CT26을 다시 심어서 종양 재발을 모니터링하였다. 화합물 39가 기처리된 마우스의 종양 성장은 사전에 아무 화합물 처리도 되지 않은 마우스보다 느렸고 1차 종양 테스트에서 종양이 완전히 사라진 개체에서는 두 번째 접종된 재발 종양에서도 종양이 없는 상태를 달성했다. 결과는 도 11에 나타내었다. 이러한 결과는 화합물 39로 유도된 면역 활성화가 추가 치료 없이 면역학적 기억을 통해 종양 재발을 예방할 수 있음을 시사한다.Thereafter, mice treated with compound 39 were re-injected with CT26 without additional compound administration to monitor tumor recurrence. Tumor growth in mice treated with compound 39 was slower than in mice that had not been previously treated with any compound, and subjects that had completely disappeared from the primary tumor test achieved tumor-free status even with a second inoculated recurrent tumor. The results are shown in FIG. 11 . These results suggest that compound 39-induced immune activation can prevent tumor recurrence through immunological memory without additional treatment.

Claims (12)

하기 화학식 1의 화합물 또는 이의 약학적으로 허용 가능한 염.
[화학식 1]
Figure pat00079

상기 화학식 1에서, R5
Figure pat00080
,
Figure pat00081
, 또는
Figure pat00082
임.
A compound of Formula 1 or a pharmaceutically acceptable salt thereof.
[Formula 1]
Figure pat00079

In Formula 1, R 5 is
Figure pat00080
,
Figure pat00081
, or
Figure pat00082
lim.
제1항에 있어서, 상기 R5
Figure pat00083
또는
Figure pat00084
인 화합물 또는 이의 약학적으로 허용 가능한 염.
The method of claim 1, wherein R 5 is
Figure pat00083
or
Figure pat00084
A phosphorus compound or a pharmaceutically acceptable salt thereof.
제2항에 있어서, 상기 R5
Figure pat00085
인 화합물 또는 이의 약학적으로 허용 가능한 염.
The method of claim 2, wherein R 5 is
Figure pat00085
A phosphorus compound or a pharmaceutically acceptable salt thereof.
2-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)-1-(4-(1-에틸-3-메틸-1H-피라졸-5-카르복스아미도)부틸)-1H-벤조[d]이미다졸-5-카르복사미드 (화합물 26) 또는 이의 약학적으로 허용 가능한 염. 2-(1-ethyl-3-methyl-1H-pyrazole-5-carboxamido)-1-(4-(1-ethyl-3-methyl-1H-pyrazole-5-carboxamido) Butyl)-1H-benzo[d]imidazole-5-carboxamide (Compound 26) or a pharmaceutically acceptable salt thereof. 제1항 내지 제4항 중 어느 한 항의 화합물 또는 이의 약학적으로 허용 가능한 염, 및 약학적으로 허용 가능한 첨가제를 포함하는 조성물. A composition comprising the compound of any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable additive. 제1항 내지 제4항 중 어느 한 항의 화합물 또는 이의 약학적으로 허용 가능한 염을 유효 성분으로 포함하는 STING 매개 질환의 치료 또는 예방용 약학 조성물. A pharmaceutical composition for the treatment or prevention of STING-mediated diseases comprising the compound of any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof as an active ingredient. 제6항에 있어서, 상기 STING 매개 질환은 암, 전암성 증후군, 또는 감염성 질환인 약학 조성물. The pharmaceutical composition according to claim 6, wherein the STING-mediated disease is cancer, precancerous syndrome, or infectious disease. 제7항에 있어서, 상기 STING 매개 질환은 암인, 약학 조성물.The pharmaceutical composition according to claim 7, wherein the STING-mediated disease is cancer. 제1항 내지 제4항 중 어느 한 항의 화합물 또는 이의 약학적으로 허용 가능한 염을 백신 애주번트로 포함하는, 백신 조성물.A vaccine composition comprising the compound of any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof as a vaccine adjuvant. 제1항 내지 제4항 중 어느 한 항의 화합물 또는 이의 약학적으로 허용 가능한 염의 치료학적으로 유효한 양을 STING 매개 질환의 치료 또는 예방이 필요한 개체에게 투여하는 것을 특징으로 하는, STING 매개 질환의 치료 또는 예방 방법. Claims 1 to 4, characterized in that the administration of a therapeutically effective amount of the compound of any one of claims 1 to 4 or a pharmaceutically acceptable salt thereof to an individual in need of treatment or prevention of a STING-mediated disease, treatment or Prevention methods. 제10항에 있어서, 상기 STING 매개 질환은 암, 전암성 증후군, 또는 감염성 질환인 치료 또는 예방 방법. The method of claim 10, wherein the STING-mediated disease is cancer, precancerous syndrome, or an infectious disease. 제10항에 있어서, 상기 STING 매개 질환은 암인, 치료 또는 예방 방법.The method of claim 10 , wherein the STING-mediated disease is cancer.
KR1020220006893A 2021-02-17 2022-01-17 Composition for stimulating interferon genes comprising a benzimidazole derivative as an active ingredient KR20220117805A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/KR2022/002319 WO2022177307A1 (en) 2021-02-17 2022-02-17 Interferon gene stimulator composition comprising benzimidazole derivative as active ingredient

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020210021453 2021-02-17
KR20210021453 2021-02-17

Publications (1)

Publication Number Publication Date
KR20220117805A true KR20220117805A (en) 2022-08-24

Family

ID=83111638

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020220006893A KR20220117805A (en) 2021-02-17 2022-01-17 Composition for stimulating interferon genes comprising a benzimidazole derivative as an active ingredient

Country Status (1)

Country Link
KR (1) KR20220117805A (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017175147A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017175147A1 (en) 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators

Similar Documents

Publication Publication Date Title
RU2705579C2 (en) Novel pyrazolo[3,4-d]pyrimidine compound or salt thereof
AU2020200979A1 (en) New substituted indazoles, methods for the production thereof, pharmaceutical preparations that contain said new substituted indazoles, and use of said new substituted indazoles to produce drugs
KR20220122662A (en) Quinazoline Compounds Containing Spiro Rings
EP2799437B1 (en) Quinoline and cinnoline derivatives and use thereof
JP5599611B2 (en) Antiviral protease inhibitor
WO2019170150A1 (en) Protein degradation targeting bcr-abl compound and antitumor application thereof
IL262414B2 (en) Polymorphic form of n-{6-(2-hydroxypropan-2-yl)-2-[2-(methylsulphonyl)ethyl]-2h-indazol-5-yl}-6-(trifluoromethyl)pyridine-2-carboxamide
US20110313167A1 (en) Substituted Heterocycles as Therapeutic agents for treating cancer
WO2017152874A1 (en) Urea compound, preparation method therefor and medical use thereof
WO2019052535A1 (en) Compound for inhibiting and degrading cdk
KR20170003688A (en) Heterocyclic Hydroxamic Acids as Protein Deacetylase Inhibitors and Dual Protein Deacetylase-Protein Kinase Inhibitors and Methods of Use Thereof
RU2650682C2 (en) Pyrrole substituted indolone derivative, method of its preparation including its composition and application
US20110251238A1 (en) Ppar agonist compositions and methods of use
KR20180100373A (en) Uses of Ascochlorin Derivatives and AMPK Activators
US20070004777A1 (en) Methods for treating or preventing acute myelogenous leukemia
KR20220117805A (en) Composition for stimulating interferon genes comprising a benzimidazole derivative as an active ingredient
CN113416181B (en) Quinazoline derivative and application thereof
KR20230133396A (en) Heteroaryl compounds as RIP2 kinase inhibitors, compositions and uses thereof
KR20210132143A (en) Novel pan-RAF kinase inhibitors and uses thereof
WO2019001307A1 (en) Amide compound, composition containing same, and use thereof
US6887864B2 (en) Azepane derivatives
JP6508796B2 (en) Preparation and use of novel kinase inhibitors
CN114929675A (en) Novel adamantane derivatives as inhibitors of focal adhesion kinase
KR102635126B1 (en) Novel pyrrolopyrimidine derivatives as a Ectonucleotide pyrophosphatase-phosphodiesterase inhibitors and use thereof
KR102409595B1 (en) Novel purinone derivatives as protein kinase CSF-1R inhibitor

Legal Events

Date Code Title Description
E902 Notification of reason for refusal