WO2023169480A1 - Deuterated compound, preparation method therefor and use thereof - Google Patents

Deuterated compound, preparation method therefor and use thereof Download PDF

Info

Publication number
WO2023169480A1
WO2023169480A1 PCT/CN2023/080351 CN2023080351W WO2023169480A1 WO 2023169480 A1 WO2023169480 A1 WO 2023169480A1 CN 2023080351 W CN2023080351 W CN 2023080351W WO 2023169480 A1 WO2023169480 A1 WO 2023169480A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
reaction
formula
deuterated
compound according
Prior art date
Application number
PCT/CN2023/080351
Other languages
French (fr)
Chinese (zh)
Other versions
WO2023169480A9 (en
Inventor
姚郑林
尹磊
Original Assignee
甘李药业股份有限公司
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 甘李药业股份有限公司 filed Critical 甘李药业股份有限公司
Publication of WO2023169480A1 publication Critical patent/WO2023169480A1/en
Publication of WO2023169480A9 publication Critical patent/WO2023169480A9/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • R 1 to R 2 are each independently selected from any group among CH 3 , CH 2 D, CHD 2 and CD 3 , R 3 to R 14 are each independently H or D, and R 1 to R 14 There is at least one D-containing atom in , provided that:
  • R2 is CD3 , CH2D , or CHD2 .
  • R 1 and R 2 of the compound are CD 3 ; or R 1 and R 2 are CD 3 , R 3 is D; or R 11 and R 14 are D; or R 3 , R 9 - R 10 is D; or R 11 and R 14 are D; or R 3 , R 11 and R 14 are D; or R 1 and R 2 are CD 3 and R 9 -R 10 is D; or R 1 and R 2 is CD 3 , R 11 and R 14 are D; or R 1 and R 2 are CD 3 , R 3 , R 9 -R 10 are D; or R 1 and R 2 are CD 3 , R 3 , R 8 and R 9 -R 10 are D; or R 1 and R 2 are CD 3 , R 3 , R 11 and R 14 are D; or R 3 and R 7 are D; Or R 3 and R 6 are D; or R 1 and R 2 are CD 3 , R 3 and R 7 are D; or R 6 , and R 9 -R 10 are D; or R 6 -R 7 and R 9
  • the deuterated compounds obtained by these embodiments of the present invention can significantly increase the action time and half-life of the drug, significantly improve metabolic stability, bioavailability, and improve pharmacokinetic properties compared to Ulixertinib. .
  • Step 3) (S)-4-(5-chloro-2-(isopropylamino)pyridin-4-yl-3,6-[D2])-N-(1-(3-chlorophenyl) -2-hydroxyethyl)-1H-pyrrole-2-amide
  • Deuterated compounds 16 and 17 (the structural formulas are shown in Table 4 respectively) each adopt corresponding intermediates and can be synthesized through a synthesis method similar to deuterated compound 15. The results are shown in Table 4:
  • Diastereomers can be separated by chromatography, preferably by separation/resolution techniques based on differences in solubility. The optically pure enantiomers are then recovered, together with the resolving reagents, by any practical means that does not involve racemization.
  • a more detailed description of techniques suitable for the resolution of stereoisomers of compounds starting from racemic mixtures can be found in Jean Jacques, Andre Collet, Samuel H. Wilen, "Enantiomers, Racemates and Resolutions” , Racemates and Resolutions" , John Wiley And Sons, Inc., l98l.
  • the metabolic stability of the deuterated compound 39 and BVD-523 provided by the present invention in monkeys is further verified.
  • the animals will be observed for clinical symptoms and blood collection on the monkey chair 1 hour before the blood collection point. After 1 hour, the animals will be returned to the original cage, and subsequent blood collection and observation will be completed in the cage.
  • Blood is collected mainly through the cephalic vein of the forelimb and the saphenous vein of the hind limb, directly collected with a 1 ml syringe. After collection, it is poured into a vacuum blood collection tube, transferred and centrifuged on wet ice, and the centrifugation time is recorded on the centrifuge table. Blood samples were collected for UPLC-MS/MS analysis and detection. PK parameters were estimated using WinNonlin 6.1 non-compartmental model.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided are a deuterated compound represented by formula IA or a pharmaceutically acceptable salt thereof, a preparation method therefor and a use thereof. Some deuterated compounds have a half-life period of up to 2-4 times that of BVD-523, therefore have a longer action time in in-vivo metabolism, generate more effective clinical application values, and are expected to solve the side effects caused by metabolic defects at present in BVD-523.

Description

氘代化合物,及其制备方法和应用Deuterated compounds, preparation methods and applications thereof 技术领域Technical field
本发明属于医药技术领域,具体涉及一种作为ERK1/2抑制剂的氘代化合物,尤其涉及对(S)-4-(5-氯-2-(异丙基氨基)吡啶-4-基)-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-2-酰胺进行氘代后获得的氘代化合物及其制备方法和应用。The invention belongs to the field of medical technology, and specifically relates to a deuterated compound as an ERK1/2 inhibitor, especially to (S)-4-(5-chloro-2-(isopropylamino)pyridin-4-yl) - A deuterated compound obtained by deuteration of N-(1-(3-chlorophenyl)-2-hydroxyethyl)-1H-pyrrole-2-amide and its preparation method and application.
背景技术Background technique
MAPK(丝裂原活化蛋白激酶)由一系列丝氨酸/苏氨酸激酶组成,可磷酸化其细胞质蛋白,从而使磷酸化的细胞质蛋白能够从胞浆移位到细胞核中进而实现调节转录因子的活性。MAPK通路是细胞内重要的信号传导途径,它将细胞外刺激传递至细胞核,参与细胞的生长、发育、分化等一系列生理过程,并在细胞的恶性转化和肿瘤的发生发展中起重要作用。经典的MAPK信号转导通路可以将细胞外信号转导至细胞内,主要通过三级激酶级联的形式MAPKKK(MAP3K)-MAPKKs(MAP2Ks)-MAPK传导细胞信号,从而调节细胞的增殖,分化,凋亡等生理功能。其中,胞外信号调节激酶(extracellular signal-regulated kinase:ERK)分为ERK1和ERK2,统称为ERK1/2,EKR1(参考文献:Sciences 1990,249(4964):64-67)和ERK2(参考文献:Cell 1991,65(4):663-675)是由TERI G.BOULTON等人于90年代初首次先后分离鉴定的,ERK1的相对分子量为44KDa,ERK2的相对分子量为42KDa。ERK1/2的信号传递途径涉及调节细胞生长、发育及***的信号网络核心,从细胞外刺激作用于细胞,使细胞产生相应的生物学效应。在经典的RAS-RAF-MEK1/2-ERK1/2通路中的各个蛋白及其通路被激活的机制表现为,以RAS作为上游激活蛋白,以RAF作为MAP3K,以MEK1/2激酶作为MAP2K,而ERK1/2即作为MAPK发挥作用(参考文献:Cell Cycle 2009,8(8):1168-1175)。其中,RAS蛋白包含H-RAS、N-RAS和K-RAS等不同类型的21kDa的蛋白,在RTK的细胞内激酶域的邻近位点处被激活的GTP酶让其从非激活的RAS-GDP状态转换成激活的RAS-GTP而刺激下游的RAF。RAF激酶包括A-RAF、B-RAF和C-RAF三个不同的亚型,是RAS的中下游效应物通过结合到负载GTP的RAS蛋白并对RAF的丝氨酸338、苏氨酸341双位点磷酸化而激活。RAF激酶通过对MEK(MEK1和MEK2)的两个紧密相邻的丝氨酸残基(丝氨酸217、丝氨酸221)进行双位点磷酸化使MEK 激活。MEK是双重特异性苏氨酸/酪氨酸激酶,MEK1/2激活后可以使ERK1的苏氨酸202和酪氨酸204以及ERK2的苏氨酸173和酪氨酸185双位点发生磷酸化从而激活ERK1/2。磷酸化激活的ERK1/2由胞质转位到细胞核内,进而介导Elk-1、ATF、MYC、NF-κΒ、Ap-1、c-FOS等多种转录因子和基因的活化和转录,参与细胞增殖与分化、细胞形态维持、细胞骨架的构建、细胞凋亡和细胞的癌变等多科生物学反应。(参考文献:Acta Pharmaceutica Sinica B 2018,8(4):552-562)。MAPK (mitogen-activated protein kinase) consists of a series of serine/threonine kinases that can phosphorylate its cytoplasmic proteins, allowing the phosphorylated cytoplasmic proteins to translocate from the cytoplasm to the nucleus to regulate the activity of transcription factors . The MAPK pathway is an important signaling pathway in cells. It transmits extracellular stimuli to the nucleus, participates in a series of physiological processes such as cell growth, development, and differentiation, and plays an important role in the malignant transformation of cells and the occurrence and development of tumors. The classic MAPK signal transduction pathway can transduce extracellular signals into cells. It mainly conducts cell signals through the three-level kinase cascade form MAPKKK (MAP3K)-MAPKKs (MAP2Ks)-MAPK, thereby regulating cell proliferation and differentiation. Apoptosis and other physiological functions. Among them, extracellular signal-regulated kinase (ERK) is divided into ERK1 and ERK2, collectively referred to as ERK1/2, EKR1 (reference: Sciences 1990, 249 (4964): 64-67) and ERK2 (reference : Cell 1991, 65(4):663-675) were first isolated and identified by TERI G.BOULTON et al. in the early 1990s. The relative molecular weight of ERK1 is 44KDa and that of ERK2 is 42KDa. The signaling pathway of ERK1/2 involves the core of the signaling network that regulates cell growth, development and division. It acts on cells from extracellular stimulation, causing cells to produce corresponding biological effects. In the classic RAS-RAF-MEK1/2-ERK1/2 pathway, each protein and the mechanism by which the pathway is activated is as follows: RAS is used as the upstream activating protein, RAF is used as MAP3K, MEK1/2 kinase is used as MAP2K, and ERK1/2 functions as MAPK (Reference: Cell Cycle 2009, 8(8): 1168-1175). Among them, RAS proteins include different types of 21kDa proteins such as H-RAS, N-RAS and K-RAS. The activated GTPase at the adjacent site of the intracellular kinase domain of RTK allows it to convert from inactive RAS-GDP. The state changes to activated RAS-GTP to stimulate downstream RAF. RAF kinase includes three different subtypes: A-RAF, B-RAF and C-RAF. It is a mid-stream and downstream effector of RAS by binding to the GTP-loaded RAS protein and targeting the serine 338 and threonine 341 double sites of RAF. activated by phosphorylation. RAF kinase catalyzes MEK by dual-site phosphorylation of two closely adjacent serine residues (serine 217, serine 221) of MEK (MEK1 and MEK2). activation. MEK is a dual-specificity threonine/tyrosine kinase. MEK1/2 activation can phosphorylate threonine 202 and tyrosine 204 of ERK1 and threonine 173 and tyrosine 185 of ERK2. thereby activating ERK1/2. Phosphorylated ERK1/2 is translocated from the cytoplasm to the nucleus, and then mediates the activation and transcription of various transcription factors and genes such as Elk-1, ATF, MYC, NF-κΒ, Ap-1, c-FOS, etc. Involved in multidisciplinary biological reactions such as cell proliferation and differentiation, cell morphology maintenance, cytoskeleton construction, cell apoptosis and cell carcinogenesis. (Reference: Acta Pharmaceutica Sinica B 2018,8(4):552-562).
RAS-RAF-MEK1/2-ERK1/2信号级联反应在多种疾病,包括脑损伤、癌症、心肌肥厚、糖尿病和炎症等的发生发展中起着关键作用。特别是在癌症中,RAS已被确认为致癌基因,在大约三分之一的癌症中发现其产生突变激活,主要表现在90%的胰腺癌、50%的结直肠癌、50%的甲状腺癌、30%的肺癌以及25%的黑色素瘤中均发现RAS基因的突变激活。(参考文献:J Cell Sci 2016,129:1287–92.)。B-RAF突变在人类恶性肿瘤中占比高达7%,主要表现在100%的HCL,50%-60%的黑色素瘤,40%-60%的甲状腺癌,5%-10%结直肠癌等中都发生了B-RAF的突变激活。(参考文献:Nat Rev Cancer 2014,14:455–67.)。大量研究表明:RAS和RAF基因突变导致肿瘤细胞中的ERK1/2被持续激活,从而引起细胞过度增殖。在各种类的癌症中多个RAF和MEK小分子抑制剂的临床效果已经将该信号通路作为癌症治疗的靶点进行最终验证(参考文献:TopicsAnti-Cancer Res,2013,2:63-94)。The RAS-RAF-MEK1/2-ERK1/2 signaling cascade plays a key role in the development of various diseases, including brain injury, cancer, cardiac hypertrophy, diabetes, and inflammation. Especially in cancer, RAS has been confirmed as an oncogene and is found to be mutated and activated in about one-third of cancers, mainly in 90% of pancreatic cancers, 50% of colorectal cancers, and 50% of thyroid cancers. Mutated activation of the RAS gene has been found in 30% of lung cancers and 25% of melanomas. (Reference: J Cell Sci 2016, 129:1287–92.). B-RAF mutations account for up to 7% of human malignant tumors, mainly in 100% of HCL, 50%-60% of melanoma, 40%-60% of thyroid cancer, 5%-10% of colorectal cancer, etc. Mutational activation of B-RAF occurred in all. (Reference: Nat Rev Cancer 2014,14:455–67.). A large number of studies have shown that RAS and RAF gene mutations lead to the continuous activation of ERK1/2 in tumor cells, thus causing excessive cell proliferation. The clinical effects of multiple RAF and MEK small molecule inhibitors in various types of cancer have ultimately validated this signaling pathway as a target for cancer treatment (Reference: Topics Anti-Cancer Res, 2013, 2: 63-94) .
近年,有一些RAF抑制剂和MEK抑制剂陆续上市,例如2021年5月由安进公司自主开发的靶向KRASG12C突变的药物Sotorasib(为RAS抑制剂)也获得了FDA批准上市,而且在临床上取得了不错的疗效。但是病人使用一段时间后,发现产生了新的耐药性突变,导致RAS-RAF-MEK-ERK1/2通路的重新激活而使癌细胞恢复增殖能力。而作为RAS-RAF-MEK1/2-ERK1/2通路下游的“最终管理器”,靶向抑制ERK1/2也有望用于治疗MAPK通路异常激活(RAS-RAF-MEK1/2等激活变异)造成的癌变,并且可能对由于ERK1/2重新激活而产生RAS、RAF或MEK1/2抑制剂耐药的患者有效。但到目前为止,还没有出现正式上市的靶向ERK1/2的抑制剂药物。In recent years, some RAF inhibitors and MEK inhibitors have been launched one after another. For example, in May 2021, Sotorasib (a RAS inhibitor), a drug independently developed by Amgen that targets the KRAS G12C mutation, was approved by the FDA and is in clinical use. Achieved good results. However, after the patient used it for a period of time, it was found that new drug-resistant mutations had developed, which led to the reactivation of the RAS-RAF-MEK-ERK1/2 pathway and restored the proliferation ability of cancer cells. As the "final manager" downstream of the RAS-RAF-MEK1/2-ERK1/2 pathway, targeted inhibition of ERK1/2 is also expected to be used to treat abnormal activation of the MAPK pathway (activation mutations such as RAS-RAF-MEK1/2). cancer, and may be effective in patients who develop resistance to RAS, RAF, or MEK1/2 inhibitors due to ERK1/2 reactivation. But so far, there are no officially marketed inhibitor drugs targeting ERK1/2.
WO2005113541A1中公开了一种中文名为(S)-4-(5-氯-2(异丙基氨基)吡啶-4-基)-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-2-酰胺的靶向ERK1/2的抑制剂,与其关联的是目前已经进入临床2期阶段的药物Ulixertinib(又名 BVD-523),虽然其在临床实验中展现出了较好的生物学活性,但是在药物代谢方面的却存在明显的缺陷,导致使用中需要施用较高的临床剂量以及非常频繁的给药方式,这些缺陷将对其进一步的临床开发产生较大限制。WO2005113541A1 discloses a product whose Chinese name is (S)-4-(5-chloro-2(isopropylamino)pyridin-4-yl)-N-(1-(3-chlorophenyl)-2-hydroxy Ethyl)-1H-pyrrole-2-amide is an ERK1/2-targeting inhibitor, related to the drug Ulixertinib (also known as Ulixertinib), which has entered Phase 2 clinical trials. BVD-523), although it has shown good biological activity in clinical trials, it has obvious defects in drug metabolism, resulting in the need for higher clinical doses and very frequent dosing methods. , these defects will greatly limit its further clinical development.
很多药物由于较差的吸收(absorption)、分布(distribution)、代谢(metabolism)及***(excretion)(ADME)性质从而限制了它们的在某些疾病上的使用,也是导致许多候选药物最终未能通过临床试验的原因。特别是针对药物的快速代谢问题,导致许多本来可以高效治疗疾病的药物由于过快的从体内代谢掉而难以成药。另外,快速代谢的药物也可能会使患者暴露于不良的毒性或反应性代谢物中。Many drugs have poor absorption, distribution, metabolism, and excretion (ADME) properties that limit their use in certain diseases. This also leads to the eventual failure of many candidate drugs. Reasons for passing clinical trials. Especially for the problem of rapid drug metabolism, many drugs that can effectively treat diseases are difficult to become drugs because they are metabolized from the body too quickly. Additionally, rapidly metabolized drugs may also expose patients to undesirable toxic or reactive metabolites.
因此,仍然需要代谢稳定性更好、半衰期更长、药代动力学性质更好、生物利用度更高的ERK1/2的抑制剂。Therefore, there is still a need for ERK1/2 inhibitors with better metabolic stability, longer half-life, better pharmacokinetic properties, and higher bioavailability.
发明内容Contents of the invention
本发明提供了一种新型的氘代修饰的Ulixertinib作为ERK1/2抑制剂,发明人通过大量实验,意外发现,本发明在特定位点进行氘代化修饰后获得的化合物在对ERK激酶具有相当或更高的抑制活性的同时,能够显著提升药物在体内的半衰期,提高生物利用度,具有更好的代谢稳定性,明显降低或消除体内代谢过程中产生的不良代谢物,能够使产生疗效所需要的施用剂量和/或施用频次明显降低。The present invention provides a new type of deuterated modified Ulixertinib as an ERK1/2 inhibitor. Through a large number of experiments, the inventor unexpectedly discovered that the compound obtained after deuterated modification at a specific site of the present invention has considerable effects on ERK kinase. or higher inhibitory activity, it can significantly increase the half-life of the drug in the body, improve bioavailability, have better metabolic stability, significantly reduce or eliminate adverse metabolites produced during the metabolism process in the body, and enable the production of therapeutic effects. Significantly lower dosages and/or frequency of administration are required.
本发明第一方面,提供了式IA氘代化合物:
In a first aspect, the present invention provides a deuterated compound of formula IA:
其中,R1-R2各自独立地选自CH3、CH2D、CHD2和CD3中的任一基团,R3-R14各自独立地是H或D,且R1-R14中至少有一个含D原子,Wherein, R 1 to R 2 are each independently selected from any group among CH 3 , CH 2 D, CHD 2 and CD 3 , R 3 to R 14 are each independently H or D, and R 1 to R 14 At least one of them contains D atom,
条件是:requirement is:
当式IA仅含1个D原子时,R8不是D,When formula IA contains only 1 D atom, R 8 is not D,
当式IA含3个D原子时,R8-R10不同时是D, When formula IA contains 3 D atoms, R 8 -R 10 are not D at the same time,
当式IA含4个D原子时,R6-R7、R11和R14不同时是D,When formula IA contains 4 D atoms, R 6 -R 7 , R 11 and R 14 are not D at the same time,
当式IA含7个D原子、且R1-R2是CD3时,R7-R8不是D;和/或When formula IA contains 7 D atoms and R 1 -R 2 is CD 3 , R 7 -R 8 is not D; and/or
当式IA含9个D原子、且R1-R2是CD3时,R6、R11和R14不同时是D。When formula IA contains 9 D atoms and R 1 -R 2 are CD 3 , R 6 , R 11 and R 14 are not D at the same time.
本发明第二方面,提供了式I所示的氘代化合物:
In a second aspect, the present invention provides a deuterated compound represented by formula I:
其中,R1-R2各自独立地选自CH3、CH2D、CHD2和CD3中的任一基团,R3-R14各自独立地是H或D,且R1-R14中至少有一个含D原子,条件是:Wherein, R 1 to R 2 are each independently selected from any group among CH 3 , CH 2 D, CHD 2 and CD 3 , R 3 to R 14 are each independently H or D, and R 1 to R 14 There is at least one D-containing atom in , provided that:
当式I仅含1个D原子时,R8不是D,When formula I contains only 1 D atom, R 8 is not D,
当式I含3个D原子时,R8-R10不同时是D,When formula I contains 3 D atoms, R 8 -R 10 are not D at the same time,
当式I含4个D原子时,R6-R7、R11和R14不同时是D,When formula I contains 4 D atoms, R 6 -R 7 , R 11 and R 14 are not D at the same time,
当式I含7个D原子、且R1-R2是CD3时,R7-R8不是D;和/或When formula I contains 7 D atoms and R 1 -R 2 is CD 3 , R 7 -R 8 is not D; and/or
当式I含9个D原子、且R1-R2是CD3时,R6、R11和R14不同时是D。When formula I contains 9 D atoms and R 1 to R 2 are CD 3 , R 6 , R 11 and R 14 are not D at the same time.
在根据本发明的一个实施方案中,该化合物的R1是CD3、CH2D、或CHD2In one embodiment according to the invention, R 1 of the compound is CD 3 , CH 2 D, or CHD 2 ;
在一个实施方案中,R2是CD3、CH2D、或CHD2In one embodiment, R2 is CD3 , CH2D , or CHD2 .
在一个实施方案中,R3是D;在一个实施方案中,R4是D;In one embodiment, R 3 is D; in one embodiment, R 4 is D;
在一个实施方案中,R5是D;在一个实施方案中,R6是D;In one embodiment, R 5 is D; in one embodiment, R 6 is D;
在一个实施方案中,R7是D;在一个实施方案中,R8是D;In one embodiment, R 7 is D; in one embodiment, R 8 is D;
在一个实施方案中,R9-R10是D;In one embodiment, R 9 -R 10 are D;
在一个实施方案中,R11和R14是D;在一个实施方案中,R12-R13是D。In one embodiment, R 11 and R 14 are D; in one embodiment, R 12 -R 13 are D.
本发明的一个实施方案中,该化合物的R1和R2是CD3;或R1和R2是CD3,R3是D;或R11和R14是D;或R3、R9-R10是D;或R11和R14是D;或R3、R11和R14是D;或R1和R2是CD3,R9-R10是D;或R1和R2是CD3、R11和R14是D;或R1和R2是CD3,R3、R9-R10是D;或R1和R2是CD3,R3、R8和R9-R10是D;或R1和R2是CD3、R3、R11和R14是D;或R3和R7是D; 或R3和R6是D;或R1和R2是CD3,R3和R7是D;或R6、和R9-R10是D;或R6-R7和R9-R10是D;或R3、R7、和R9-R10是D;或R1和R2是CD3,R7和R9-R10是D;或R1和R2是CD3,R3、R7和R9-R10是D;或R3、R6、和R9-R10是D;或R1和R2是CD3,R3、R8是D;或R1和R2是CD3,R3、R6、R7是D;或R1和R2是CD3,R3、R6、R7、R8是D;或R1和R2是CD3,R3、R6、R7、R8、R9、R10是D。发明人意外发现,本发明的这些实施方案所得到的氘代化合物,相比于Ulixertinib,能够显著提高药物的作用时间和半衰期,显著提高代谢稳定性、生物利用度、以及改善药代动力学性质。In one embodiment of the invention, R 1 and R 2 of the compound are CD 3 ; or R 1 and R 2 are CD 3 , R 3 is D; or R 11 and R 14 are D; or R 3 , R 9 - R 10 is D; or R 11 and R 14 are D; or R 3 , R 11 and R 14 are D; or R 1 and R 2 are CD 3 and R 9 -R 10 is D; or R 1 and R 2 is CD 3 , R 11 and R 14 are D; or R 1 and R 2 are CD 3 , R 3 , R 9 -R 10 are D; or R 1 and R 2 are CD 3 , R 3 , R 8 and R 9 -R 10 are D; or R 1 and R 2 are CD 3 , R 3 , R 11 and R 14 are D; or R 3 and R 7 are D; Or R 3 and R 6 are D; or R 1 and R 2 are CD 3 , R 3 and R 7 are D; or R 6 , and R 9 -R 10 are D; or R 6 -R 7 and R 9 - R 10 is D; or R 3 , R 7 , and R 9 -R 10 are D; or R 1 and R 2 are CD 3 and R 7 and R 9 -R 10 are D; or R 1 and R 2 are CD 3 , R 3 , R 7 and R 9 -R 10 are D; or R 3 , R 6 , and R 9 -R 10 are D; or R 1 and R 2 are CD 3 , R 3 and R 8 are D; Or R 1 and R 2 are CD 3 , R 3 , R 6 , and R 7 are D; or R 1 and R 2 are CD 3 , R 3 , R 6 , R 7 , and R 8 are D; or R 1 and R 2 is CD 3 , R 3 , R 6 , R 7 , R 8 , R 9 and R 10 are D. The inventor unexpectedly discovered that the deuterated compounds obtained by these embodiments of the present invention can significantly increase the action time and half-life of the drug, significantly improve metabolic stability, bioavailability, and improve pharmacokinetic properties compared to Ulixertinib. .
在根据本发明的一个实施方案中,该化合物的R12和R13是H。In one embodiment according to the invention, R 12 and R 13 of the compound are H.
在根据本发明的一个实施方案中,该化合物选自下述结构式所示的化合物中的任一种:









In one embodiment according to the present invention, the compound is selected from any one of the compounds represented by the following structural formula:









在根据本发明的一个实施方案中,该化合物选自下述结构式所示的化合物中的任一种:








In one embodiment according to the present invention, the compound is selected from any one of the compounds represented by the following structural formula:








本发明的另一方面提供了上述的氘代化合物的制备方法,所述氘代化合物是通过反应式Ⅰ制备得到的:
Another aspect of the present invention provides a method for preparing the above-mentioned deuterated compound, which is prepared by reaction formula I:
其中,R1-R2各自独立地选自CH3、CH2D、CHD2或CD3中的任一种,R3-R14各自独立地选自H或D中的一种;X1选自Br、I、OTf或Oms中的任一种;Y为离去基团Ts或Boc。 Wherein, R 1 to R 2 are each independently selected from any one of CH 3 , CH 2 D, CHD 2 or CD 3 , and R 3 to R 14 are each independently selected from one of H or D; X 1 Selected from any one of Br, I, OTf or Oms; Y is the leaving group Ts or Boc.
在优选的一个实施方案中,还涉及 In a preferred embodiment, it also involves
中间体化合物4可以通过与中间体化合物5通过铃木反应得到中间体化合物6,所述中间体化合物6水解得到中间体化合物7,最后中间体化合物7和中间体化合物8进行缩合反应,即得到式I化合物。Intermediate compound 4 can be obtained by Suzuki reaction with intermediate compound 5 to obtain intermediate compound 6. The intermediate compound 6 is hydrolyzed to obtain intermediate compound 7. Finally, intermediate compound 7 and intermediate compound 8 undergo a condensation reaction to obtain the formula I compound.
在根据本发明的一个实施方案中,所述中间体化合物4是通过反应式Ⅱ或反应式Ⅲ制备得到的:
In one embodiment according to the present invention, the intermediate compound 4 is prepared by reaction formula II or reaction formula III:
其中,R1-R2各自独立地选自CH3、CH2D、CHD2或CD3中的任一种,R3-R14各自独立地选自H和D中的任一种;X1为OTf或OMs,X2选自卤素F、Cl、Br或I中的任一种;Y为离去基团Ts或Boc。Wherein, R 1 to R 2 are each independently selected from any one of CH 3 , CH 2 D, CHD 2 or CD 3 , and R 3 to R 14 are each independently selected from any one of H and D; X 1 is OTf or OMs, X 2 is selected from any one of the halogens F, Cl, Br or I; Y is the leaving group Ts or Boc.
在一个优选地实施方案中,所述反应式Ⅱ的中间体化合物1经还原反应制备得到中间体化合物2;所述中间体化合物2与中间体化合物3经取代反应得到中间体化合物4;In a preferred embodiment, the intermediate compound 1 of the reaction formula II is prepared through a reduction reaction to obtain the intermediate compound 2; the intermediate compound 2 and the intermediate compound 3 are subjected to a substitution reaction to obtain the intermediate compound 4;
所述反应式Ⅲ中,中间体化合物1’和中间体化合物2’经还原胺化制备得到中间体化合物4。In the reaction formula III, intermediate compound 1' and intermediate compound 2' are prepared through reductive amination to obtain intermediate compound 4.
在根据本发明的进一步的实施方案中,所述中间体化合物4选自In a further embodiment according to the invention, said intermediate compound 4 is selected from
中的任一种。 any of them.
在根据本发明的一个实施方案中,中间体化合物4与下述的中间体化合物5反应得到中间体6:
In one embodiment according to the present invention, intermediate compound 4 is reacted with the following intermediate compound 5 to obtain intermediate 6:
在根据本发明的进一步的实施方案中,中间体化合物5的前体化合物选自In a further embodiment according to the invention, the precursor compound of intermediate compound 5 is selected from the group consisting of
中的任一种。 any of them.
在根据本发明的进一步的实施方案中,中间体化合物8选自In a further embodiment according to the invention, intermediate compound 8 is selected from
中的任一种。 any of them.
本发明的再一方面提供了上述氘代化合物,或其药学上可接受的盐,在制备用于预防和/或治疗增生性疾病或者由RAS-RAF-MEK1/2-ERK1/2激酶调节的疾病的药物中的应用。Another aspect of the present invention provides the above-mentioned deuterated compound, or a pharmaceutically acceptable salt thereof, in the preparation of drugs for preventing and/or treating proliferative diseases or regulated by RAS-RAF-MEK1/2-ERK1/2 kinase. Applications in medicines for diseases.
在根据本发明的一个实施方案中,所述由RAS-RAF-MEK1/2-ERK1/2激酶调节的疾病为对ERK的抑制敏感的肿瘤。In one embodiment according to the present invention, the disease regulated by RAS-RAF-MEK1/2-ERK1/2 kinase is a tumor that is sensitive to inhibition of ERK.
本发明进一步提供了一种用于预防和/或治疗增生性疾病或者由RAS-RAF-MEK1/2-ERK1/2激酶调节的病症的药物组合物,其含有药学上可接受的载体和上述的氘代化合物,或其药学上可接受的盐。 The present invention further provides a pharmaceutical composition for preventing and/or treating proliferative diseases or conditions regulated by RAS-RAF-MEK1/2-ERK1/2 kinase, which contains a pharmaceutically acceptable carrier and the above-mentioned Deuterated compounds, or pharmaceutically acceptable salts thereof.
进一步地,本发明所提供的上述任一化合物或任一药物组合物,其用作药物。Further, any of the above compounds or any pharmaceutical composition provided by the present invention is used as a medicine.
进一步地,本发明所提供的上述任一化合物或任一药物组合物,其用作药物,其用于预防和/或治疗对ERK的抑制敏感的肿瘤。Furthermore, any of the above compounds or any pharmaceutical composition provided by the present invention is used as a medicine for preventing and/or treating tumors that are sensitive to ERK inhibition.
进一步地,本发明还提供了预防和/或治疗对ERK的抑制敏感的肿瘤的方法,包括施用治疗有效量的上述任一化合物或任一药物组合物。Furthermore, the present invention also provides methods for preventing and/or treating tumors that are sensitive to ERK inhibition, including administering a therapeutically effective amount of any of the above compounds or any pharmaceutical composition.
优选地,上述疾病或者对ERK的抑制敏感的肿瘤为人黑色素瘤或人结肠癌。Preferably, the above-mentioned disease or tumor sensitive to ERK inhibition is human melanoma or human colon cancer.
本发明提供了药学效果较佳的氘代的吡咯酰胺类化合物,通过在某些位点或位点组合进行氘代后,获得的氘代化合物与对照化合物BVD-523相比具有相当或更好的抑制活性的同时,还具有更长的半衰期,体内代谢更趋于稳定、药代动力学更优、生物利用度更好,有望降低临床使用剂量。本发明提供的部分氘代化合物的半衰期甚至出乎意料地达到BVD-523的2-4倍,在体内代谢中的作用时间显著延长,有助于解决BVD-523因代谢缺陷导致的单次施用剂量较大、施用频次较高的情况,并减轻或消除因BVD-523代谢缺陷而产生的副作用,进而可以扩展用于更多适应症的治疗。同时,本发明还提供了相应氘代化合物的制备方法和应用。The present invention provides deuterated pyrrolamide compounds with better pharmaceutical effects. After deuteration at certain sites or site combinations, the deuterated compounds obtained have equivalent or better properties than the reference compound BVD-523. In addition to its inhibitory activity, it also has a longer half-life, more stable metabolism in the body, better pharmacokinetics, and better bioavailability, which is expected to reduce the clinical dosage. The half-life of some deuterated compounds provided by the present invention even unexpectedly reaches 2-4 times that of BVD-523, and the action time in the metabolism in the body is significantly prolonged, which helps to solve the problem of single administration of BVD-523 caused by metabolic defects. With larger doses and higher frequency of administration, and reducing or eliminating side effects caused by BVD-523 metabolic defects, it can be expanded for the treatment of more indications. At the same time, the present invention also provides preparation methods and applications of corresponding deuterated compounds.
附图说明Description of the drawings
图1为化合物BVD-523在食蟹猴中1mg静脉给药(IV)和10mg口服灌胃给药(PO)血药浓度曲线图,横轴为时间(单位:hr),纵轴为血药浓度(单位:ng/ml);Figure 1 is a graph showing the blood concentration curve of compound BVD-523 in cynomolgus monkeys after 1 mg intravenous administration (IV) and 10 mg oral administration (PO). The horizontal axis is time (unit: hr), and the vertical axis is blood drug. Concentration (unit: ng/ml);
图2为根据本发明制备得到的氘代化合物39在食蟹猴中1mg静脉给药(IV)和10mg口服灌胃给药(PO)血药浓度曲线图,横轴为时间(单位:hr),纵轴为血药浓度(单位:ng/ml)。Figure 2 is a graph showing the plasma concentration curve of deuterated compound 39 prepared according to the present invention in cynomolgus monkeys after 1 mg intravenous administration (IV) and 10 mg oral administration (PO). The horizontal axis is time (unit: hr) , the vertical axis is the blood drug concentration (unit: ng/ml).
图3为根据本发明制备得到的氘代化合物28在食蟹猴中1mg静脉给药(IV)和10mg口服灌胃给药(PO)血药浓度曲线图,横轴为时间(单位:hr),纵轴为血药浓度(单位:ng/ml)。Figure 3 is a graph showing the plasma concentration curve of deuterated compound 28 prepared according to the present invention in cynomolgus monkeys after 1 mg intravenous administration (IV) and 10 mg oral administration (PO). The horizontal axis is time (unit: hr) , the vertical axis is the blood drug concentration (unit: ng/ml).
具体实施例Specific embodiments
下面结合具体实施例,作进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下述实施例中的实验方法,如无 特殊说明,均为常规方法。下述实施例中所用的化学原料、试剂等,如无特殊说明,均为市售购买产品。The present invention will be further described below in conjunction with specific embodiments. It should be understood that these examples are only used to illustrate the invention and are not intended to limit the scope of the invention. Experimental methods in the following examples, if not Special instructions are all common methods. The chemical raw materials, reagents, etc. used in the following examples are all commercially available products unless otherwise specified.
如无特别说明,术语“本发明的化合物”,是指式(I)化合物及其盐、包括化合物的药学上可接受的盐以及所有立体异构体(包括但不限于非对映异构体和对映异构体)、互变异构体、其前药和同位素化合物。溶剂合物和水合物通常被认为是本发明的化合物的药物组成物。Unless otherwise specified, the term "compounds of the present invention" refers to compounds of formula (I) and salts thereof, including pharmaceutically acceptable salts of the compounds and all stereoisomers (including but not limited to diastereomers). and enantiomers), tautomers, their prodrugs and isotopic compounds. Solvates and hydrates are generally considered pharmaceutical compositions of the compounds of the invention.
术语“药学上可接受的盐”指保留了特定化合物的游离酸和碱的生物学效力而没有生物学不良作用的盐。药学上可接受的盐的例子包括但不限于:(1)酸加成盐,和无机酸例如盐酸、硫酸、氢溴酸、硝酸、磷酸等形成的盐;或和有机酸例如苹果酸、富马酸、马来酸、苯甲酸、苯乙酸、琥珀酸、酒石酸、柠檬酸、甲磺酸、乙磺酸、羟基乙酸、肉桂酸、丙酮酸、甲酸、乙酸、丙酸、草酸、丙二酸、丙烯酸、扁桃酸等形成的盐;或者(2)碱加成盐,和碱金属例如锂、钠、钾等形成的盐;和碱土金属例如钙、镁等形成的盐;和有机碱例如铵、胆碱、二乙醇胺、赖氨酸、乙二胺、叔丁胺、叔辛胺、三(羟甲基)氨基甲烷、N-甲基葡萄糖胺、三乙醇胺、脱氢松香胺等形成的盐。对于本领域的技术人员而言,其他的药学上可接受的盐是已知的。The term "pharmaceutically acceptable salts" refers to salts that retain the biological potency of the free acids and bases of a particular compound without adverse biological effects. Examples of pharmaceutically acceptable salts include, but are not limited to: (1) acid addition salts, salts formed with inorganic acids such as hydrochloric acid, sulfuric acid, hydrobromic acid, nitric acid, phosphoric acid, etc.; or with organic acids such as malic acid, rich acid, etc. Maleic acid, benzoic acid, phenylacetic acid, succinic acid, tartaric acid, citric acid, methanesulfonic acid, ethanesulfonic acid, glycolic acid, cinnamic acid, pyruvic acid, formic acid, acetic acid, propionic acid, oxalic acid, malonic acid , acrylic acid, mandelic acid, etc.; or (2) alkali addition salts, salts formed with alkali metals such as lithium, sodium, potassium, etc.; salts formed with alkaline earth metals such as calcium, magnesium, etc.; and organic bases such as ammonium , choline, diethanolamine, lysine, ethylenediamine, tert-butylamine, tert-octylamine, tris(hydroxymethyl)aminomethane, N-methylglucosamine, triethanolamine, dehydrorosinamine, etc. Other pharmaceutically acceptable salts are known to those skilled in the art.
术语“同位素化合物”是指本发明的通式(I)化合物中含有一个或多个天然或非天然丰度的原子同位素。非天然丰度的原子同位素包括但不限于氘(2H或D)、氚(3H或T)、碘-125(125I)、磷-32(32P)、碳-13(13C)或碳-14(14C)。本发明中所合成的化合物的任何原子若没有特别指定,可代表该原子的任何一种稳定的同位素。除非特别说明,当结构中某一位置被定义为H即氢(H-1)时,该位置仅含天然存在的同位素量(0.015%)。The term "isotopic compound" means that the compound of general formula (I) of the present invention contains one or more atomic isotopes in natural or unnatural abundance. Atomic isotopes of non-natural abundance include, but are not limited to, deuterium ( 2 H or D), tritium ( 3 H or T), iodine-125 ( 125 I), phosphorus-32 ( 32 P), carbon-13 ( 13 C) or carbon-14( 14 C). Unless otherwise specified, any atom of the compound synthesized in the present invention may represent any stable isotope of the atom. Unless otherwise stated, when a position in the structure is defined as H, i.e. hydrogen (H-1), that position contains only the naturally occurring isotope (0.015%).
“氘”或“D”或“d”表示氢的同位素,其核含有一个质子和一个中子。当特别的位置被指定为含有氘时,应当理解的是该位置的氘的丰度大于氘的天然丰度(通常为0.015%)。除非另外说明,否则当某位置被特别指定为“D”或“氘”时,该位置应当理解为含有丰度大于氘的天然丰度的氘。"Deuterium" or "D" or "d" refers to an isotope of hydrogen whose nucleus contains one proton and one neutron. When a particular position is designated as containing deuterium, it is understood that the abundance of deuterium at that position is greater than the natural abundance of deuterium (typically 0.015%). Unless otherwise stated, when a position is specifically designated as "D" or "deuterium," that position is understood to contain deuterium in an abundance greater than the natural abundance of deuterium.
本发明中所合成的化合物的氘代率是指合成的同位素含量与天然存在的同位素量的比值。本发明中所合成的化合物的每个指定氘原子的氘代率可至少为3333.3倍(50%)、至少为4000倍(60%)、至少为4500倍(67.5%)、至少为5000倍(75%)、至少为5333.3倍(80%)、至少为6000倍(90%)、至少为6333.3倍(95%)、至少为6466.7倍(97%)、至少为6566.7倍(98.5%)、至少为6600 倍(99%)、至少为6633.3倍(99.5%)。The deuteration rate of the compound synthesized in the present invention refers to the ratio of the synthesized isotope content to the naturally occurring isotope amount. The deuteration rate of each designated deuterium atom of the compound synthesized in the present invention can be at least 3333.3 times (50%), at least 4000 times (60%), at least 4500 times (67.5%), at least 5000 times ( 75%), at least 5333.3 times (80%), at least 6000 times (90%), at least 6333.3 times (95%), at least 6466.7 times (97%), at least 6566.7 times (98.5%), at least to 6600 times (99%), at least 6633.3 times (99.5%).
本发明中所合成的化合物中的某位置的氢同位素体的量取决许多因素,其中包括氘代试剂(如D2O、D2、NaOD、NaBD4、LiAlD4等)的氘同位素纯度以及引入氘同位素合成方法的有效性。然而,如前所述这种某位置的氢同位素体的量总数将少于49.9%。本发明中所合成的化合物中的某位置的氢同位素体的量总数将少于47.5%、40%、32.5%、25%、17.5%、10%、5%、3%、1%或0.5%。The amount of hydrogen isotope at a certain position in the compound synthesized in the present invention depends on many factors, including the purity of the deuterium isotope of the deuterated reagent (such as D 2 O, D 2 , NaOD, NaBD 4 , LiAlD 4, etc.) and the introduction of Validity of deuterium isotope synthesis methods. However, as mentioned above, the total amount of hydrogen isotopes at a certain location will be less than 49.9%. The total amount of hydrogen isotopes at a certain position in the compound synthesized in the present invention will be less than 47.5%, 40%, 32.5%, 25%, 17.5%, 10%, 5%, 3%, 1% or 0.5% .
本文中,任何未指定为氘的各原子以其天然同位素丰度存在。As used herein, any atom not designated as deuterium exists in its natural isotope abundance.
术语“溶剂合物”是指通过与溶剂分子配位形成固态或液态的配合物的本专利发明的化合物的形式。此类形式的实例为水合物、醇合物等。The term "solvate" refers to a form of a compound of the present invention that forms a solid or liquid complex by coordination with a solvent molecule. Examples of such forms are hydrates, alcoholates, etc.
术语“前药”是指在体内转化成母体药物的任何药剂。前药常常是有用的,因为,在某些情况下,他们比母体药物更易于给药。例如,通过口服给药,它们是可生物利用的,而母体药物却不是。前药相对于母体药物还可改善药物组合物中的溶解度。前药可经由酶的方法以及代谢水解的途径被转化为母体药物。The term "prodrug" refers to any agent that is converted in the body to the parent drug. Prodrugs are often useful because, in some cases, they are easier to administer than the parent drug. For example, they are bioavailable through oral administration, whereas the parent drug is not. Prodrugs may also improve solubility in pharmaceutical compositions relative to the parent drug. Prodrugs can be converted to the parent drug via enzymatic methods as well as metabolic hydrolysis pathways.
英文缩写:English abbreviations:
1,4-dioxone:1,4-二氧六环;ACN:乙腈;AOPI:一种荧光染料;1,4-dioxone:1,4-dioxane; ACN: acetonitrile; AOPI: a fluorescent dye;
Bin2P:双联频哪醇硼酸酯;dtbpy:4,4’-二叔丁基-2,2’-联吡啶;Bin 2 P: Dipinopinacol borate; dtbpy: 4,4'-di-tert-butyl-2,2'-bipyridine;
MeOH:甲醇;DCM:二氯甲烷;D2:氘气;DMF:N,N-二甲基甲酰胺;MeOH: methanol; DCM: dichloromethane; D2 : deuterium; DMF: N,N-dimethylformamide;
DMAP:4-二甲氨基吡啶;DIEA:N,N-二异丙基乙胺;EA:乙酸乙酯;DMAP: 4-dimethylaminopyridine; DIEA: N,N-diisopropylethylamine; EA: ethyl acetate;
ee%:对映体过量百分比;EDCI:1-(3-二甲胺基丙基)-3-乙基碳二亚胺;ee%: enantiomeric excess percentage; EDCI: 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide;
HATU:2-(7-氮杂苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯;HATU: 2-(7-azabenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate;
HOBt:1-羟基苯并三氮唑;In(OTf)3:三氟甲烷磺酸铟;Ms:甲基磺酰基;HOBt: 1-hydroxybenzotriazole; In(OTf) 3 : indium trifluoromethanesulfonate; Ms: methylsulfonyl group;
Ir(OMe)(cod)2:甲氧基(环辛二烯)铱(I)二聚体;LiAlH4:四氢锂铝;Ir(OMe)(cod) 2 : methoxy (cyclooctadiene) iridium (I) dimer; LiAlH 4 : lithium aluminum tetrahydrogen;
LiOH.H2O:氢氧化锂一水合物;LiAlD4:氘代四氢锂铝;Na2CO3:碳酸钠;LiOH . H 2 O: lithium hydroxide monohydrate; LiAlD 4 : lithium aluminum deuterated tetrahydrogen; Na 2 CO 3 : sodium carbonate;
NaBH3CN:氰基硼氢化钠;NaOD:氘代氢氧化钠;NaOH:氢氧化钠;NaBH 3 CN: sodium cyanoborohydride; NaOD: deuterated sodium hydroxide; NaOH: sodium hydroxide;
NCS:N-氯代丁二酰亚胺;NIS:N-碘代丁二酰亚胺;NaH:钠氢;NCS: N-chlorosuccinimide; NIS: N-iodosuccinimide; NaH: sodium hydrogen;
Tf:三氟甲基磺酰基;Tf2O:三氟甲磺酸酐;TFA:三氟乙酸;Tf: trifluoromethanesulfonyl; Tf 2 O: trifluoromethanesulfonic anhydride; TFA: trifluoroacetic acid;
TEA:三乙胺;PE:石油醚;Pd(pph3)4:四(三苯基膦)钯;TEA: triethylamine; PE: petroleum ether; Pd (pph 3 ) 4 : tetrakis (triphenylphosphine) palladium;
Ts:对甲苯磺酰基;Pd/C:钯碳;Ts: p-toluenesulfonyl; Pd/C: palladium carbon;
Pd(dppf)Cl2 .DCM:[1,1’-双(二苯基膦基)二茂铁]二氯化钯二氯甲烷络合物;Pd(dppf)Cl 2 . DCM: [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride dichloromethane complex;
Flash:低压快速制备色谱;GC-MS:气相质谱;HPLC:高压制备液相色谱;Flash: low-pressure rapid preparative chromatography; GC-MS: gas chromatography mass spectrometry; HPLC: high-pressure preparative liquid chromatography;
PH:氢离子浓度指数;Pre-TLC:预制硅胶薄层色谱;TLC:薄层色谱法; PH: hydrogen ion concentration index; Pre-TLC: prefabricated silica gel thin layer chromatography; TLC: thin layer chromatography;
SFC:超临界流体色谱法;SFC: supercritical fluid chromatography;
KDa:千道尔顿;BID:每天2次。KDa: kilodaltons; BID: 2 times a day.
本发明提供的氘代化合物(包括其盐)可使用已知有机合成技术来制备,且可按照多种可能合成途径中的任一种(诸如下文方案中的那些)来合成。用于制备本发明化合物的反应可在合适的溶剂中进行,有机合成领域的技术人员可容易地选择溶剂。合适的溶剂可在进行反应的温度(例如,在溶剂冻结温度至溶剂沸点温度范围内的温度)下与起始物质(反应物)、中间体或产物实质上不反应。既定反应可在一种溶剂或一种以上溶剂的混合物中进行。技术人员可依据具体反应步骤来选择用于具体反应步骤的溶剂。The deuterated compounds provided by the present invention (including salts thereof) can be prepared using known organic synthesis techniques and can be synthesized according to any of a variety of possible synthetic pathways (such as those in the schemes below). The reactions used to prepare the compounds of the present invention can be carried out in suitable solvents, which can be readily selected by those skilled in the art of organic synthesis. Suitable solvents may be substantially unreactive with the starting materials (reactants), intermediates, or products at the temperatures at which the reaction is conducted (eg, at temperatures ranging from the freezing temperature of the solvent to the boiling temperature of the solvent). A given reaction can be carried out in one solvent or a mixture of more than one solvent. Skilled personnel can select solvents for specific reaction steps based on the specific reaction steps.
一、药物中间体及其合成1. Drug intermediates and their synthesis
在氘代化合物的合成过程中,由于可氘代位置较多,因此针对在不同位置进行氘代时,就需要基于对应的中间体化合物进行,在此本申请基于药物的整体合成路线提出以下关键中间体,并基于关键中间体的骨架将关键中间体划分为氘取代异丙基系列关键中间体(A系列中间体)、氘取代氯苯系列关键中间体(B系列中间体)、氘取代吡啶环系列关键中间体(C系列中间体)以及氘取代吡咯环系列关键中间体(D系列中间体),各关键中间体的合成过程具体描述如下:In the synthesis process of deuterated compounds, since there are many deuterated positions, deuteration at different positions needs to be based on corresponding intermediate compounds. Here, this application proposes the following key points based on the overall synthesis route of the drug. Intermediates, and based on the skeleton of the key intermediates, the key intermediates are divided into deuterium-substituted isopropyl series key intermediates (A series intermediates), deuterium-substituted chlorobenzene series key intermediates (B series intermediates), deuterium-substituted pyridine series Ring series key intermediates (C series intermediates) and deuterium-substituted pyrrole ring series key intermediates (D series intermediates). The synthesis process of each key intermediate is described in detail as follows:
1、氘取代异丙基系列关键中间体(A系列中间体)的制备1. Preparation of deuterium-substituted isopropyl series key intermediates (A series intermediates)
1.1中间体A-1的制备
1.1 Preparation of intermediate A-1
步骤1:中间体A-1-2的制备Step 1: Preparation of intermediate A-1-2
用冰浴控制反应瓶的温度在0℃,在80mL的无水THF中缓慢加入LiAlD4(5.0g,68.49mmol),然后在0℃下缓慢滴加溶解在20mL的无水THF丙酮肟(5.0g,68.49mmol)。接下来,撤掉冰浴,氮气保护密封环境下,采用控温在10℃左右低温冷凝管缓慢升温到70℃剧烈反应3小时。反应完成后,缓慢降温到0℃,然后在冰浴下加入十水硫酸钠终止反应。接下来采用常压蒸馏,收集40-60℃左右的馏分,并向馏分中加入4N的盐酸1,4-二氧六环溶液,浓缩干得到A-1-2产物为黄色油状物3.7g。GC-MS[M+H]+=61.1。Use an ice bath to control the temperature of the reaction flask at 0°C, slowly add LiAlD 4 (5.0g, 68.49mmol) to 80mL of anhydrous THF, and then slowly add acetone oxime (5.0) dissolved in 20mL of anhydrous THF at 0°C. g, 68.49mmol). Next, remove the ice bath, use a low-temperature condenser tube with temperature control at about 10°C, and slowly raise the temperature to 70°C for a vigorous reaction of 3 hours in a sealed environment protected by nitrogen. After the reaction is completed, the temperature is slowly lowered to 0°C, and then sodium sulfate decahydrate is added in an ice bath to terminate the reaction. Next, use normal pressure distillation to collect the fractions at about 40-60°C, add 4N hydrochloric acid 1,4-dioxane solution to the fractions, and concentrate to dryness to obtain 3.7g of product A-1-2 as a yellow oil. GC-MS[M+H] + =61.1.
步骤2:中间体A-1的制备Step 2: Preparation of Intermediate A-1
将化合物5-氯-2-氟-4-碘吡啶(500mg,1.95mmol)溶解在乙醇(22mL)中,依次向反应瓶加入TEA(1.3mL)和A-1-2(1.9g,19.5mmol)。然后将该反应加热到90℃反应过夜。TLC检测反应毕,将有机溶剂旋干,萃取,柱 层析纯化(PE/EA=1:1)得到A-1产物为无色油状物300mg。Compound 5-chloro-2-fluoro-4-iodopyridine (500 mg, 1.95 mmol) was dissolved in ethanol (22 mL), and TEA (1.3 mL) and A-1-2 (1.9 g, 19.5 mmol) were added to the reaction bottle in sequence. ). The reaction was then heated to 90°C overnight. After the reaction is detected by TLC, the organic solvent is spin-dried, extracted, and column After chromatography purification (PE/EA=1:1), 300 mg of product A-1 was obtained as colorless oil.
LC-MS[M+H]+=298.1。1H-NMR(400MHz,DMSO-d6)δ7.98(s,1H),7.03(s,1H),6.67(brs,1H),1.11(s,3H),1.09(s,3H)。LC-MS[M+H] + =298.1. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.98 (s, 1H), 7.03 (s, 1H), 6.67 (brs, 1H), 1.11 (s, 3H), 1.09 (s, 3H).
1.2.1中间体A-2的制备
1.2.1 Preparation of intermediate A-2
步骤1:中间体A-2-1的制备Step 1: Preparation of intermediate A-2-1
将氘代丙酮(25mL)溶解在100mL水中,然后将碳酸钠(20g,188.67mmol)缓慢加到氘代丙酮溶液中。接着,将盐酸羟胺(23.6g,342.02mmol)溶解在100mL水中缓慢加入到上述反应液中,然后室温搅拌过夜。反应毕,用二氯甲烷进行萃取,分液,无水硫酸钠干燥,浓缩得到A-2-1产物为白色固体29g。GC-MS[M+H]+=80.3。Dissolve deuterated acetone (25 mL) in 100 mL of water, and then slowly add sodium carbonate (20 g, 188.67 mmol) to the deuterated acetone solution. Next, hydroxylamine hydrochloride (23.6g, 342.02mmol) was dissolved in 100 mL of water and slowly added to the above reaction solution, and then stirred at room temperature overnight. After the reaction was completed, the mixture was extracted with dichloromethane, separated into liquids, dried over anhydrous sodium sulfate, and concentrated to obtain 29 g of product A-2-1 as a white solid. GC-MS[M+H] + =80.3.
步骤2:中间体A-2-2的制备Step 2: Preparation of intermediate A-2-2
用冰浴控制反应瓶的温度在0℃,在80mL的无水THF中缓慢加入LiAlH4(16.8g,441mmol),然后在0℃下缓慢滴加溶解在20mL的无水THF中的A-2-1(29g,367mmol)。接下来,撤掉冰浴,氮气保护密封环境下,采用控温在10℃左右低温冷凝管缓慢升温到70℃剧烈反应3小时。反应毕,缓慢降温到0℃,然后,在冰浴下加入Na2SO4·10H2O终止反应。接下来采用常压蒸馏,收集40-60℃左右的馏分,并向馏分中加入4N的盐酸1,4-二氧六环溶液,浓缩至干,得到黄色油状物产物8.0g,即中间体A-2-2。GC-MS[M+H]+=66.1。Use an ice bath to control the temperature of the reaction bottle at 0°C, slowly add LiAlH 4 (16.8g, 441mmol) to 80 mL of anhydrous THF, and then slowly add A-2 dissolved in 20 mL of anhydrous THF dropwise at 0°C. -1 (29g, 367mmol). Next, remove the ice bath, use a low-temperature condenser tube with temperature control at about 10°C, and slowly raise the temperature to 70°C for a vigorous reaction of 3 hours in a sealed environment protected by nitrogen. After the reaction was completed, the temperature was slowly lowered to 0°C, and then Na 2 SO 4 ·10H 2 O was added in an ice bath to terminate the reaction. Next, use normal pressure distillation to collect the fractions at about 40-60°C, add 4N hydrochloric acid 1,4-dioxane solution to the fractions, and concentrate to dryness to obtain 8.0g of yellow oily product, which is Intermediate A. -2-2. GC-MS[M+H] + =66.1.
步骤3:中间体A-2的制备Step 3: Preparation of Intermediate A-2
将化合物5-氯-2-氟-4-碘吡啶(2.0g,7.8mmol)溶解在乙醇(20mL)中,依次向反应瓶加入TEA(10.8mL)和A-2-2(1.9g,19.5mmol)。然后将该反应加热到95℃反应15小时。TLC检测反应毕,将有机溶剂旋干,萃取,柱层析纯化(PE/EA=0~15%)得到A-2产物为白色固体1.8g。LC-MS[M+H]+=303.1。Dissolve compound 5-chloro-2-fluoro-4-iodopyridine (2.0g, 7.8mmol) in ethanol (20mL), and add TEA (10.8mL) and A-2-2 (1.9g, 19.5 mmol). The reaction was then heated to 95°C for 15 hours. After TLC detection, the reaction was completed. The organic solvent was spin-dried, extracted, and purified by column chromatography (PE/EA=0-15%) to obtain 1.8 g of product A-2 as a white solid. LC-MS[M+H] + =303.1.
1.2.2中间体A-2’的制备
1.2.2 Preparation of intermediate A-2'
在氮气保护下,向反应瓶中依次加入氘代丙酮(617mg,9.64mmol)和4- 溴-5-氯吡啶-2-胺(1.0g,4.83mmol)溶解在10mL氘代甲醇中,加入2滴醋酸,然后室温反应过夜。第二天向反应体系中在加入NaBH3CN(910.7mg,28.92mmol),然后继续反应12小时。将反应液倒入水中,萃取,分液,无水硫酸钠干燥,柱层析纯化(PE/EA=0~10%)得到A-2’产物为白色固体180mg。LC-MS[M+H]+=255.3。1H-NMR(400MHz,DMSO-d6)δ8.06(s,1H),6.81(s,1H),6.80(d,J=7.6Hz,1H),3.89(d,J=7.6Hz,1H)。Under nitrogen protection, deuterated acetone (617 mg, 9.64 mmol) and 4- Bromo-5-chloropyridin-2-amine (1.0g, 4.83mmol) was dissolved in 10mL deuterated methanol, 2 drops of acetic acid were added, and the reaction was carried out at room temperature overnight. The next day, NaBH 3 CN (910.7 mg, 28.92 mmol) was added to the reaction system, and the reaction was continued for 12 hours. The reaction solution was poured into water, extracted, separated, dried over anhydrous sodium sulfate, and purified by column chromatography (PE/EA=0~10%) to obtain 180 mg of product A-2' as a white solid. LC-MS[M+H] + =255.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ8.06 (s, 1H), 6.81 (s, 1H), 6.80 (d, J = 7.6Hz, 1H), 3.89 (d, J = 7.6Hz, 1H ).
1.3中间体A-3的制备
1.3 Preparation of intermediate A-3
步骤1:中间体A-2-1的制备Step 1: Preparation of intermediate A-2-1
将氘代丙酮(5.0g)溶解在40mL水中,然后将碳酸钠(9.9g,93.7mmol)缓慢加到氘代丙酮溶液中。接着,将盐酸羟胺(5.9g,85.9mmol)溶解在30mL水中缓慢加入到上述反应液中,然后室温搅拌过夜。反应毕,用二氯甲烷进行萃取,分液,无水硫酸钠干燥,浓缩得到A-2-1产物为白色固体5.0g。GC-MS[M+H]+=80.3。Dissolve deuterated acetone (5.0g) in 40mL water, and then slowly add sodium carbonate (9.9g, 93.7mmol) to the deuterated acetone solution. Next, hydroxylamine hydrochloride (5.9 g, 85.9 mmol) was dissolved in 30 mL of water and slowly added to the above reaction solution, and then stirred at room temperature overnight. After the reaction is completed, extract with dichloromethane, separate the layers, dry with anhydrous sodium sulfate, and concentrate to obtain 5.0 g of product A-2-1 as a white solid. GC-MS[M+H] + =80.3.
步骤2:中间体A-3-1的制备Step 2: Preparation of intermediate A-3-1
用冰浴控制反应瓶的温度在0℃,在30mL的无水THF中缓慢加入LiAlD4(1.7g,40.47mmol),然后在0℃下缓慢滴加溶解在20mL的无水THF中的A-2-1(2.0g,25.31mmol)。接下来,撤掉冰浴,氮气保护密封环境下,采用控温在10℃左右低温冷凝管缓慢升温到70℃剧烈反应3小时。反应毕,缓慢降温到0℃,然后在冰浴下加入十水硫酸钠终止反应。接下来采用常压蒸馏,收集40-60℃左右的馏分,并向馏分中加入4N的盐酸1,4-二氧六环溶液,浓缩干得到A-3-1产物为黄色油状物2.7g。GC-MS[M+H]+=67.2。Use an ice bath to control the temperature of the reaction bottle at 0°C, slowly add LiAlD 4 (1.7g, 40.47mmol) to 30 mL of anhydrous THF, and then slowly add A- dissolved in 20 mL of anhydrous THF at 0°C. 2-1 (2.0g, 25.31mmol). Next, remove the ice bath, use a low-temperature condenser tube with temperature control at about 10°C, and slowly raise the temperature to 70°C for a vigorous reaction of 3 hours in a sealed environment protected by nitrogen. After the reaction is completed, the temperature is slowly lowered to 0°C, and then sodium sulfate decahydrate is added in an ice bath to terminate the reaction. Next, use normal pressure distillation to collect the fractions at about 40-60°C, add 4N hydrochloric acid 1,4-dioxane solution to the fractions, and concentrate to dryness to obtain 2.7g of product A-3-1 as a yellow oil. GC-MS[M+H] + =67.2.
步骤3:中间体A-3的制备Step 3: Preparation of Intermediate A-3
将化合物5-氯-2-氟-4-碘吡啶(100mg,0.39mmol)溶解在乙醇(2mL)中,依次向反应瓶加入TEA(0.32mL)和A-3-1(200mg,1.95mmol)。然后将该反应加热到95℃反应15小时。TLC检测反应毕,将有机溶剂旋干,萃取,柱层析纯化(PE/EA=0~8%)得到A-3产物为白色固体27mg。LC-MS[M+H]+=304.2。1H-NMR(400MHz,DMSO-d6)δ7.99(s,1H),7.03(s,1H),6.66(brs,1H)。Compound 5-chloro-2-fluoro-4-iodopyridine (100 mg, 0.39 mmol) was dissolved in ethanol (2 mL), and TEA (0.32 mL) and A-3-1 (200 mg, 1.95 mmol) were added to the reaction bottle in sequence. . The reaction was then heated to 95°C for 15 hours. After TLC detection, the reaction was completed. The organic solvent was spin-dried, extracted, and purified by column chromatography (PE/EA=0~8%) to obtain 27 mg of product A-3 as a white solid. LC-MS[M+H] + =304.2. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.99 (s, 1H), 7.03 (s, 1H), 6.66 (brs, 1H).
2、氘取代氯苯系列关键中间体(B系列中间体)的制备2. Preparation of deuterium-substituted chlorobenzene series key intermediates (B series intermediates)
2.1中间体B-1的制备
2.1 Preparation of intermediate B-1
将反应物(S)-2-氨基-2-(3-氯苯基)乙酸(500mg,2.69mmol)溶解在10mL无水THF中,在冰浴条件下缓慢加入LiAlD4(169.5mg,4.04mmol)。然后在氮气保护下将反应液加热到70℃反应12小时。反应毕,将反应液缓慢倒入冰中,DCM萃取,分液,无水硫酸钠干燥,浓缩,C-18反向在220nm条件下纯化得到B-1产物为白色固体302mg。LC-MS[M+H]+=174.3。1H-NMR(400MHz,DMSO-d6)δ7.42(s,1H),7.34-7.25(m,3H),4.76(brs,1H),3.84(s,1H),2.01(brs,2H)。The reactant (S)-2-amino-2-(3-chlorophenyl)acetic acid (500mg, 2.69mmol) was dissolved in 10mL anhydrous THF, and LiAlD 4 (169.5mg, 4.04mmol) was slowly added under ice bath conditions. ). The reaction solution was then heated to 70°C for 12 hours under nitrogen protection. After the reaction is completed, the reaction solution is slowly poured into ice, extracted with DCM, separated, dried over anhydrous sodium sulfate, concentrated, and purified in the C-18 reverse direction at 220 nm to obtain 302 mg of the B-1 product as a white solid. LC-MS[M+H] + =174.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.42 (s, 1H), 7.34-7.25 (m, 3H), 4.76 (brs, 1H), 3.84 (s, 1H), 2.01 (brs, 2H) .
2.2中间体B-2的制备
2.2 Preparation of intermediate B-2
步骤1:中间体B-2-1的合成Step 1: Synthesis of intermediate B-2-1
将反应物2-氨基-2-(3-氯苯基)乙酸(3.54g,19.07mmol)溶解在20mL的重水(D2O)中,然后向反应液中在缓慢加入3.5mL的NaOD。接着加热到100℃反应过夜。反应毕,将反应液降低到室温,在用1N的盐酸调节PH值到6-7后析出大量白色固体,水洗,干燥得到B-2-1产物为白色固体1.6g。LC-MS[M-H]+=185.1。1H-NMR(400MHz,DMSO-d6)δ8.09(brs,1H),7.48(s,1H),7.36-7.35(m,3H)。The reactant 2-amino-2-(3-chlorophenyl)acetic acid (3.54g, 19.07mmol) was dissolved in 20mL of heavy water (D 2 O), and then 3.5mL of NaOD was slowly added to the reaction solution. Then it was heated to 100°C for overnight reaction. After the reaction is completed, the reaction solution is lowered to room temperature, and a large amount of white solid is precipitated after adjusting the pH value to 6-7 with 1N hydrochloric acid, washed with water, and dried to obtain 1.6 g of the B-2-1 product as a white solid. LC-MS[MH] + =185.1. 1 H-NMR (400MHz, DMSO-d 6 ) δ8.09 (brs, 1H), 7.48 (s, 1H), 7.36-7.35 (m, 3H).
步骤2:中间体B-2-2的合成Step 2: Synthesis of intermediate B-2-2
将反应物B-2-1(300mg,1.61mmol)溶解在7mL无水THF中,在冰浴条件下缓慢加入LiAlH4(90.5mg,2.42mmol)。然后在氮气保护下将反应液加热到70℃反应12小时。反应毕,将反应液缓慢倒入冰中,DCM萃取,分液,无水硫酸钠干燥,浓缩,C-18反向在220nm条件下纯化得到B-2-2产物为白色固体254mg。Reactant B-2-1 (300 mg, 1.61 mmol) was dissolved in 7 mL anhydrous THF, and LiAlH 4 (90.5 mg, 2.42 mmol) was slowly added under ice bath conditions. The reaction solution was then heated to 70°C for 12 hours under nitrogen protection. After the reaction is completed, the reaction solution is slowly poured into ice, extracted with DCM, separated, dried over anhydrous sodium sulfate, concentrated, and purified in the C-18 reverse direction at 220 nm to obtain 254 mg of the B-2-2 product as a white solid.
LC-MS[M+H]+=173.1。LC-MS[M+H] + =173.1.
步骤3:中间体B-2的合成Step 3: Synthesis of Intermediate B-2
将中间体B-2-2(200mg)用甲醇溶解,采用SFC进行手型拆分,拆分条件为改性剂为甲醇:乙腈=1:1加千分之五的二乙胺,比例为15%等度,总流速80mL/min,拆分后浓缩分离得到B-2为白色固体84mg。LC-MS[M+H]+=173.1。1H-NMR(400MHz,DMSO-d6)δ7.44(s,1H),7.30-7.25(m,3H),4.79(s,1H),3.34-3.29(m,2H),1.92(brs,2H)。Dissolve intermediate B-2-2 (200mg) with methanol, and use SFC for manual separation. The separation conditions are as follows: the modifier is methanol: acetonitrile = 1:1 plus 5/1000 of diethylamine, the ratio is: 15% isocratic, total flow rate 80mL/min, separated and concentrated to obtain 84 mg of B-2 as a white solid. LC-MS[M+H] + =173.1. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.44(s,1H),7.30-7.25(m,3H),4.79(s,1H),3.34-3.29(m,2H),1.92(brs, 2H).
2.3中间体B-3的制备
2.3 Preparation of intermediate B-3
步骤1:中间体B-2-1的合成Step 1: Synthesis of intermediate B-2-1
将反应物2-氨基-2-(3-氯苯基)乙酸(3.54g,19.07mmol)溶解在20mL的重水(D2O)中,然后向反应液中在缓慢加入3.5mL的NaOD。接着加热到100℃反应过夜。反应毕,将反应液降低到室温,在用1N的盐酸调节PH值到6-7后析出大量白色固体,水洗,干燥得到B-2-1产物为白色固体1.6g。LC-MS[M-H]+=185.1。1H-NMR(400MHz,DMSO-d6)δ8.09(brs,1H),7.48(s,1H),7.36-35(m,3H)。The reactant 2-amino-2-(3-chlorophenyl)acetic acid (3.54g, 19.07mmol) was dissolved in 20mL of heavy water (D 2 O), and then 3.5mL of NaOD was slowly added to the reaction solution. Then it was heated to 100°C for overnight reaction. After the reaction is completed, the reaction solution is lowered to room temperature, and a large amount of white solid is precipitated after adjusting the pH value to 6-7 with 1N hydrochloric acid, washed with water, and dried to obtain 1.6 g of the B-2-1 product as a white solid. LC-MS[MH] + =185.1. 1 H-NMR (400MHz, DMSO-d 6 ) δ8.09 (brs, 1H), 7.48 (s, 1H), 7.36-35 (m, 3H).
步骤2:中间体B-3-1的合成Step 2: Synthesis of intermediate B-3-1
将反应物B-2-1(500mg,2.69mmol)溶解在10mL无水THF中,在冰浴条件下缓慢加入LiAlD4(169.2mg,4.04mmol)。然后在氮气保护下将反应液加热到70℃反应12小时。反应毕,将反应液缓慢倒入冰中,DCM萃取,分液,无水硫酸钠干燥,浓缩,C-18反向在220nm条件下纯化得到B-3-1产物为白色固体370mg。LC-MS[M+H]+=175.2。Reactant B-2-1 (500 mg, 2.69 mmol) was dissolved in 10 mL anhydrous THF, and LiAlD 4 (169.2 mg, 4.04 mmol) was slowly added under ice bath conditions. The reaction solution was then heated to 70°C for 12 hours under nitrogen protection. After the reaction is completed, the reaction solution is slowly poured into ice, extracted with DCM, separated, dried over anhydrous sodium sulfate, concentrated, and purified in the C-18 reverse direction at 220 nm to obtain 370 mg of the B-3-1 product as a white solid. LC-MS[M+H] + =175.2.
步骤3:中间体B-3的合成Step 3: Synthesis of Intermediate B-3
将中间体B-3-1(280mg)用甲醇溶解,采用SFC进行手型拆分,拆分条件为改性剂为甲醇:乙腈=1:1加千分之五的二乙胺,比例为15%等度,总流速80mL/min,拆分后浓缩分离得到B-3为白色固体104mg。LC-MS[M+H]+=175.2。1H-NMR(400MHz,DMSO-d6)δ7.43(s,1H),7.34-7.24(m,3H),4.81(brs,1H),3.44-3.26(m,2H)。Dissolve intermediate B-3-1 (280mg) with methanol, and use SFC for manual separation. The separation conditions are as follows: the modifier is methanol: acetonitrile = 1:1 plus 5/1000 of diethylamine, the ratio is: 15% isocratic, total flow rate 80mL/min, separated and concentrated to obtain 104 mg of B-3 as a white solid. LC-MS[M+H] + =175.2. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.43 (s, 1H), 7.34-7.24 (m, 3H), 4.81 (brs, 1H), 3.44-3.26 (m, 2H).
2.4中间体B-4的制备
2.4 Preparation of intermediate B-4
将(S)-2-氨基-2-(3-氯苯基)乙-1-醇(200mg,1.17mmol)溶解在5mL无水THF中,然后在加入B2Pin(3.0g,11.7mmol)。再在氮气保护下向上述体系中加入Ir(OMe)(cod)2(16mg,0.02mmol)、dtbpy(13mg,0.05mmol)和重水(2mL)。该反应加热到100℃反应19小时后,检测反应生成中间态,再将反应体系放入到微波中140℃反应2小时。反应毕,将反应将至室温,倒入水中,萃取,分离,无水硫酸钠干燥,C-18反向HPLC纯化得到B-4产物为白色固体25mg。LC-MS[M+H]+=174.2。1H-NMR(400MHz,DMSO-d6)δ7.35(d,J=5.2Hz,1H),7.27(d,J=5.2Hz,1H),4.81-4.80(m,1H),3.87-3.85(m,1H),3.43-3.41(m,1H),3.31-3.29(m,1H),1.98-1.97(m,2H)。Dissolve (S)-2-amino-2-(3-chlorophenyl)ethan-1-ol (200mg, 1.17mmol) in 5mL anhydrous THF, and then add B 2 Pin (3.0g, 11.7mmol) . Then, Ir(OMe)(cod) 2 (16 mg, 0.02 mmol), dtbpy (13 mg, 0.05 mmol) and heavy water (2 mL) were added to the above system under nitrogen protection. After the reaction was heated to 100°C for 19 hours, the reaction was detected to form an intermediate state, and then the reaction system was placed in the microwave at 140°C for 2 hours. After the reaction is completed, the reaction is brought to room temperature, poured into water, extracted, separated, dried over anhydrous sodium sulfate, and purified by C-18 reverse HPLC to obtain 25 mg of the B-4 product as a white solid. LC-MS[M+H] + =174.2. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.35 (d, J = 5.2 Hz, 1H), 7.27 (d, J = 5.2 Hz, 1H), 4.81-4.80 (m, 1H), 3.87-3.85 (m,1H),3.43-3.41(m,1H),3.31-3.29(m,1H),1.98-1.97(m,2H).
3、氘取代吡啶环系列关键中间体(C系列中间体)的制备3. Preparation of key intermediates of deuterium-substituted pyridine ring series (C series intermediates)
3.1.1中间体C-1的合成
3.1.1 Synthesis of intermediate C-1
步骤1:中间体C-1-1的合成Step 1: Synthesis of intermediate C-1-1
将化合物4-羟基-2-氨基吡啶(10g,90.90mmol)加入到50mL重水中,接着加入2.5g的Pd/C,并向反应液中充氢气5分钟。接着将反应加热到210℃反应8小时。反应毕,将反应液降到室温,过滤,滤饼在75℃下真空干燥3小时得到产物C-1-1为白色固体7.5g。LC-MS[M+H]+=114.3。1H-NMR(400MHz,DMSO-d6)δ9.88(brs,1H),5.61(brs,2H)。Compound 4-hydroxy-2-aminopyridine (10 g, 90.90 mmol) was added to 50 mL of heavy water, then 2.5 g of Pd/C was added, and hydrogen gas was charged into the reaction solution for 5 minutes. The reaction was then heated to 210°C for 8 hours. After the reaction is completed, the reaction solution is lowered to room temperature, filtered, and the filter cake is vacuum dried at 75° C. for 3 hours to obtain 7.5 g of product C-1-1 as a white solid. LC-MS[M+H] + =114.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ 9.88 (brs, 1H), 5.61 (brs, 2H).
步骤2:中间体C-1-2的合成Step 2: Synthesis of intermediate C-1-2
将化合物C-1-1(4.0g,35.4mmol)溶解在50mL无水DMF后,在氮气保护下向反应瓶中加入NaH(1.7g.70.8mmol),该反应在室温下搅拌30分钟后,在向上述体系加入对甲氧基苄氯(5.6g,35.4mmol)。然后将反应液加热到60℃反应过夜。反应毕,将反应液降到室温倒入冰中,析出大量固体,过滤,得到产物C-1-2为白色固体2.0g。LC-MS[M+H]+=234.3。1H-NMR(400MHz,DMSO-d6)δ7.36(d,J=8.8Hz,2H),6.95(d,J=6.8Hz,2H),5.78(brs,2H),4.96(s,2H),3.75(s,3H)。After dissolving compound C-1-1 (4.0g, 35.4mmol) in 50mL anhydrous DMF, NaH (1.7g.70.8mmol) was added to the reaction flask under nitrogen protection. After the reaction was stirred at room temperature for 30 minutes, To the above system, p-methoxybenzyl chloride (5.6g, 35.4mmol) was added. The reaction solution was then heated to 60°C for overnight reaction. After the reaction is completed, the reaction solution is lowered to room temperature and poured into ice. A large amount of solid is precipitated and filtered to obtain 2.0 g of product C-1-2 as a white solid. LC-MS[M+H] + =234.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.36 (d, J = 8.8 Hz, 2H), 6.95 (d, J = 6.8 Hz, 2H), 5.78 (brs, 2H), 4.96 (s, 2H ), 3.75(s,3H).
步骤3:中间体C-1-3的合成Step 3: Synthesis of intermediate C-1-3
将C-1-2(1.8g,6.4mmol)溶解在15mL无水二氯甲烷中,然后依次向反应瓶中加入二碳酸二叔丁酯(1.1g,5.1mmol)、TEA(2.6mL,19.2mmol)和DMAP(78mg,0.64mmol)。接着该反应在室温反应3小时。反应毕,旋干溶剂,萃取,分液,有机相浓缩,粗品进行柱层析纯化得到C-1-3为黄色油状物0.8g。LC-MS[M+H]+=334.2。Dissolve C-1-2 (1.8g, 6.4mmol) in 15mL anhydrous dichloromethane, then add di-tert-butyl dicarbonate (1.1g, 5.1mmol) and TEA (2.6mL, 19.2 mmol) and DMAP (78 mg, 0.64 mmol). The reaction was then allowed to proceed at room temperature for 3 hours. After the reaction was completed, the solvent was spin-dried, extracted, separated, and the organic phase was concentrated. The crude product was purified by column chromatography to obtain 0.8 g of C-1-3 as a yellow oil. LC-MS[M+H] + =334.2.
步骤4:中间体C-1-4的合成Step 4: Synthesis of intermediate C-1-4
将C-1-3(4.0g,12.01mmol)溶解在80mL无水DMF中,在氮气保护和0℃时,缓慢向反应瓶中加入NaH(1.08g,25.22mmol)。室温搅拌30分钟后,再向反应瓶中加入2-碘丙烷(2.86g,16.82mmol),室温搅拌反应过夜。反应毕,将反应液倒入冰水中淬灭,萃取,分液,无水硫酸钠干燥,浓缩,柱层析分离得到C-1-4为白色固体3.0g。LC-MS[M+H]+=376.2。 Dissolve C-1-3 (4.0g, 12.01mmol) in 80mL anhydrous DMF, and slowly add NaH (1.08g, 25.22mmol) to the reaction bottle under nitrogen protection and 0°C. After stirring at room temperature for 30 minutes, 2-iodopropane (2.86g, 16.82mmol) was added to the reaction flask, and the reaction was stirred at room temperature overnight. After the reaction is completed, the reaction solution is poured into ice water to quench, extracted, separated, dried over anhydrous sodium sulfate, concentrated, and separated by column chromatography to obtain 3.0 g of C-1-4 as a white solid. LC-MS[M+H] + =376.2.
步骤5:中间体C-1-5的合成Step 5: Synthesis of intermediate C-1-5
将中间体C-1-4(100mg,0.265mmol)溶解在3mLTFA中,然后将反应液加热到70℃反应4小时。反应毕,将TFA旋干,粗品采用C-18的flash进行纯化得到C-1-5为白色固体45mg。LC-MS[M+H]+=156.1。1H-NMR(400MHz,DMSO-d6)δ6.22-6.21(m,1H),3.87-3.85(m,1H),3.55-3.43(m,1H),1.11(d,J=6.4Hz,6H)。Intermediate C-1-4 (100 mg, 0.265 mmol) was dissolved in 3 mL of TFA, and then the reaction solution was heated to 70°C for 4 hours. After the reaction is completed, TFA is spun to dryness, and the crude product is purified using C-18 flash to obtain 45 mg of C-1-5 as a white solid. LC-MS[M+H] + =156.1. 1 H-NMR (400MHz, DMSO-d 6 ) δ6.22-6.21(m,1H),3.87-3.85(m,1H),3.55-3.43(m,1H),1.11(d,J=6.4Hz, 6H).
步骤6:中间体C-1-6的合成Step 6: Synthesis of intermediate C-1-6
将中间体C-1-5(10mg,0.064mmol)在0℃下溶解到二氯甲烷中并搅拌10分钟,在氮气保护下将NCS(10mg,0.075mmol)加入到上述反应液中。然后在0℃反应40分钟,反应毕。将溶剂旋干,萃取,分液,无水硫酸钠干燥,过滤,pre-TLC纯化得到C-1-6为白色固体8mg。LC-MS[M+H]+=189.3。1H-NMR(400MHz,DMSO-d6)δ5.55(d,J=5.2Hz,1H),4.19-4.10(m,1H),2.58(s,1H),1.16(d,J=4.4Hz,6H)。Intermediate C-1-5 (10 mg, 0.064 mmol) was dissolved in dichloromethane at 0° C. and stirred for 10 minutes. NCS (10 mg, 0.075 mmol) was added to the above reaction solution under nitrogen protection. Then react at 0°C for 40 minutes to complete the reaction. The solvent was spin-dried, extracted, separated, dried over anhydrous sodium sulfate, filtered, and purified by pre-TLC to obtain 8 mg of C-1-6 as a white solid. LC-MS[M+H] + =189.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ5.55 (d, J = 5.2Hz, 1H), 4.19-4.10 (m, 1H), 2.58 (s, 1H), 1.16 (d, J = 4.4Hz ,6H).
步骤6:中间体C-1的合成Step 6: Synthesis of intermediate C-1
将中间体C-1-6(50mg,0.266mmol)溶解在DCM中加入到5mL的反应瓶,然后0℃条件下向反应瓶中依次加入Tf2O(74.7mg,0.266mmol)和TEA(53.7mg,0.532mmol),室温搅拌反应过夜。反应毕,将溶剂旋干,加入冰水后萃取,分液,无水硫酸钠干燥,过滤,pre-TLC纯化得到C-1为白色固体35mg。LC-MS[M+H]+=321.3。Dissolve intermediate C-1-6 (50 mg, 0.266 mmol) in DCM and add it to a 5 mL reaction flask, then add Tf 2 O (74.7 mg, 0.266 mmol) and TEA (53.7) into the reaction flask in sequence at 0°C. mg, 0.532mmol), and the reaction was stirred at room temperature overnight. After the reaction is completed, spin the solvent to dryness, add ice water, extract, separate the liquids, dry over anhydrous sodium sulfate, filter, and purify by pre-TLC to obtain 35 mg of C-1 as a white solid. LC-MS[M+H] + =321.3.
3.1.2中间体C-1’的合成
3.1.2 Synthesis of intermediate C-1'
步骤1:中间体C-1’-1的合成Step 1: Synthesis of intermediate C-1’-1
将化合物4-溴-5-氯-2-氨基吡啶(5g,24.3mmol)加入到40mL重水中,接着将反应加热到250℃反应2小时。反应毕,将反应液降到室温,过滤,滤饼在75℃下真空干燥3小时得到产物C-1’-1为白色固体503mg。LC-MS[M+H]+=209.3。Compound 4-bromo-5-chloro-2-aminopyridine (5g, 24.3mmol) was added to 40 mL of heavy water, and then the reaction was heated to 250°C for 2 hours. After the reaction was completed, the reaction solution was lowered to room temperature, filtered, and the filter cake was vacuum dried at 75°C for 3 hours to obtain 503 mg of product C-1'-1 as a white solid. LC-MS[M+H] + =209.3.
步骤2:中间体C-1’的合成Step 2: Synthesis of intermediate C-1’
在氮气保护下,向反应瓶中依次加入丙酮(139.4mg,2.41mmol)和C-1’-1(100mg,0.48mmol)溶解在10mL甲醇中,加入2滴醋酸,然后室温反应过夜。第二天向反应体系中在加入NaBH3CN(151.83mg,2.41mmol),然后继续反应12小时。将反应液倒入水中,萃取,分液,无水硫酸钠干燥,柱层析纯化(PE/EA=0~12%)得到C-1’产物为白色固体25mg。LC-MS[M+H]+=253.1。Under nitrogen protection, add acetone (139.4 mg, 2.41 mmol) and C-1'-1 (100 mg, 0.48 mmol) in sequence to the reaction flask, dissolve it in 10 mL methanol, add 2 drops of acetic acid, and then react at room temperature overnight. The next day, NaBH 3 CN (151.83 mg, 2.41 mmol) was added to the reaction system, and the reaction was continued for 12 hours. The reaction solution was poured into water, extracted, separated, dried over anhydrous sodium sulfate, and purified by column chromatography (PE/EA=0~12%) to obtain 25 mg of the C-1' product as a white solid. LC-MS[M+H] + =253.1.
4、氘取代吡咯环系列关键中间体(D系列中间体)的制备4. Preparation of key intermediates of deuterium-substituted pyrrole ring series (D series intermediates)
4.1中间体D-1的合成
4.1 Synthesis of intermediate D-1
步骤1:中间体D-1-2的合成Step 1: Synthesis of intermediate D-1-2
将5-溴-1H-吡咯-2-甲酸甲酯(1.0g,4.91mmol)溶解在10mL的氘代甲醇中,然后向反应液中加入40mg的Pd/C后,在氘气环境下置换3次,室温反应4小时。反应毕,将反应液过滤,并用碳酸氢钠水溶液调节pH到7,乙酸乙酯萃取,分液,无水硫酸钠干燥,过滤,有相机浓缩得到产物D-1-2为黄色油状物610mg。LC-MS[M+H]+=127.3。Dissolve 5-bromo-1H-pyrrole-2-carboxylic acid methyl ester (1.0g, 4.91mmol) in 10 mL of deuterated methanol, then add 40 mg of Pd/C to the reaction solution, and replace 3 in a deuterium atmosphere. times, react at room temperature for 4 hours. After the reaction was completed, the reaction solution was filtered, and the pH was adjusted to 7 with a sodium bicarbonate aqueous solution, extracted with ethyl acetate, separated, dried over anhydrous sodium sulfate, filtered, and concentrated with a camera to obtain 610 mg of product D-1-2 as a yellow oil. LC-MS[M+H] + =127.3.
步骤2:中间体D-1-3的合成Step 2: Synthesis of intermediate D-1-3
将D-1-2(500mg,4mmol)溶解在5mL乙腈中,然后在0℃下缓慢加入NIS(1.08g,4.4mmol)和In(OTf)3(225mg,0.4mmol)。在氮气惰性环境下,该反应在0℃继续反应4小时。反应毕,将反应液倒入冰水中,乙酸乙酯萃取,分液,无水硫酸钠干燥,过滤,浓缩,C-18反向flash纯化得到产物D-1-3为白色固体620mg。LC-MS[M+H]+=253.1。1H-NMR(400MHz,DMSO-d6)δ12.26(brs,1H),6.87(s,1H),3.75(s,3H)。D-1-2 (500 mg, 4 mmol) was dissolved in 5 mL of acetonitrile, and then NIS (1.08 g, 4.4 mmol) and In(OTf) 3 (225 mg, 0.4 mmol) were slowly added at 0°C. The reaction was continued at 0°C for 4 hours under an inert atmosphere of nitrogen. After the reaction was completed, the reaction solution was poured into ice water, extracted with ethyl acetate, separated, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by C-18 reverse flash to obtain 620 mg of product D-1-3 as a white solid. LC-MS[M+H] + =253.1. 1 H-NMR (400MHz, DMSO-d 6 ) δ12.26 (brs, 1H), 6.87 (s, 1H), 3.75 (s, 3H).
步骤3:中间体D-1-4的合成Step 3: Synthesis of intermediate D-1-4
将D-1-3(360mg,1.43mmol)溶解在4mL的二氯甲烷中,然后在0℃条件下依次向反应体系中加入对甲苯磺酰氯(407mg,2.14mmol)、TEA(0.3mL)和DMAP(174mg,1.43mmol)。接着将反应升至室温搅拌过夜。反应毕,将反应液倒入冰水中,萃取,分液,浓缩,硅胶柱层析Flash纯化分离(EA:PE=0~15%)得到产物D-1-4为白色固体510mg。LC-MS[M+H]+=407.3。Dissolve D-1-3 (360 mg, 1.43 mmol) in 4 mL of methylene chloride, and then add p-toluenesulfonyl chloride (407 mg, 2.14 mmol), TEA (0.3 mL) and DMAP (174mg, 1.43mmol). The reaction was then warmed to room temperature and stirred overnight. After the reaction is completed, the reaction solution is poured into ice water, extracted, separated, concentrated, and purified and separated by Flash silica gel column chromatography (EA:PE=0~15%) to obtain 510 mg of product D-1-4 as a white solid. LC-MS[M+H] + =407.3.
步骤4:中间体D-1的合成Step 4: Synthesis of Intermediate D-1
在氮气保护下,向密闭的20mL的微波反应瓶中依次加入D-1-4(600mg,1.5mmol)溶解在6mL无水DMF中,双联频哪醇硼酸酯(762mg,3mmol)、1,1’-二(二苯基膦基)二茂铁二氯化钯的二氯甲烷络合物(122mg,0.15mmol)和乙酸钾(441mg,4.5mmol)。然后将该反应液加热到95℃反应5小时。反应毕,将反应液加入少量乙酸乙酯,过硅藻土处理,接下来萃取,分液,有机相浓缩,粗品采用C-18Flash纯化分离得到产物D-1为白色固体200mg。LC-MS[M+H]+=407.1。 Under nitrogen protection, add D-1-4 (600 mg, 1.5 mmol) dissolved in 6 mL anhydrous DMF, double pinacol borate (762 mg, 3 mmol), 1 , 1'-bis(diphenylphosphino)ferrocene palladium dichloride dichloromethane complex (122 mg, 0.15 mmol) and potassium acetate (441 mg, 4.5 mmol). The reaction solution was then heated to 95°C for 5 hours. After the reaction is completed, a small amount of ethyl acetate is added to the reaction solution, and it is treated with diatomaceous earth, followed by extraction, liquid separation, and concentration of the organic phase. The crude product is purified and isolated using C-18 Flash to obtain 200 mg of product D-1 as a white solid. LC-MS[M+H] + =407.1.
4.2中间体D-2的合成
4.2 Synthesis of intermediate D-2
步骤1:中间体D-2-2的合成Step 1: Synthesis of intermediate D-2-2
将3-溴-1H-吡咯-2-甲酸甲酯(5.0g,24.5mmol)溶解在50mL的氘代甲醇中,然后向反应液中加入500mg的Pd/C和TEA(3.4mL)后,在氘气环境下置换3次,室温反应4小时。反应毕,将反应液过滤,并用碳酸氢钠水溶液调节PH到7,乙酸乙酯萃取,分液,无水硫酸钠干燥,过滤,有相机浓缩得到产物D-2-2为黄色固体4.2g。LC-MS[M+H]+=127.3。Dissolve 3-bromo-1H-pyrrole-2-carboxylic acid methyl ester (5.0g, 24.5mmol) in 50mL of deuterated methanol, and then add 500mg of Pd/C and TEA (3.4mL) to the reaction solution. Replace 3 times under deuterium gas environment and react at room temperature for 4 hours. After the reaction is completed, the reaction solution is filtered, and the pH is adjusted to 7 with a sodium bicarbonate aqueous solution, extracted with ethyl acetate, separated, dried over anhydrous sodium sulfate, filtered, and concentrated with a camera to obtain 4.2g of product D-2-2 as a yellow solid. LC-MS[M+H] + =127.3.
步骤2:中间体D-2-3的合成Step 2: Synthesis of intermediate D-2-3
将D-2-2(200mg,1.59mmol)溶解在2mL乙腈中,然后在0℃下缓慢加入NIS(376mg,1.67mmol)和In(OTf)3(89mg,0.159mmol)。在氮气惰性环境下,该反应在0℃继续反应1.5小时。反应毕,将反应液倒入冰水中,乙酸乙酯萃取,分液,无水硫酸钠干燥,过滤,浓缩,C-18反向flash纯化得到产物D-2-3为黄色固体200mg。LC-MS[M+H]+=253.1。1H-NMR(400MHz,DMSO-d6)δ12.19(brs,1H),7.16(s,1H),3.75(s,3H)。D-2-2 (200 mg, 1.59 mmol) was dissolved in 2 mL acetonitrile, and then NIS (376 mg, 1.67 mmol) and In(OTf) 3 (89 mg, 0.159 mmol) were slowly added at 0°C. The reaction was continued at 0°C for 1.5 hours under an inert atmosphere of nitrogen. After the reaction was completed, the reaction solution was poured into ice water, extracted with ethyl acetate, separated, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by C-18 reverse flash to obtain 200 mg of product D-2-3 as a yellow solid. LC-MS[M+H] + =253.1. 1 H-NMR (400MHz, DMSO-d 6 ) δ 12.19 (brs, 1H), 7.16 (s, 1H), 3.75 (s, 3H).
步骤3:中间体D-2-4的合成Step 3: Synthesis of intermediate D-2-4
将D-2-3(200mg,0.75mmol)溶解在2mL的二氯甲烷中,然后在0℃条件下依次向反应体系中加入对甲苯磺酰氯(215mg,1.13mmol)、TEA(0.2mL)和DMAP(92mg,0.75mmol)。接着将反应升至室温搅拌过夜。反应毕,将反应液倒入冰水中,萃取,分液,浓缩,硅胶柱层析Flash纯化分离(EA:PE=0~15%)得到产物D-2-4为白色固体301mg。LC-MS[M+H]+=407.3。Dissolve D-2-3 (200 mg, 0.75 mmol) in 2 mL of methylene chloride, and then add p-toluenesulfonyl chloride (215 mg, 1.13 mmol), TEA (0.2 mL) and DMAP (92 mg, 0.75 mmol). The reaction was then warmed to room temperature and stirred overnight. After the reaction is completed, the reaction solution is poured into ice water, extracted, separated, concentrated, and purified and separated by silica gel column chromatography (EA:PE=0~15%) to obtain 301 mg of product D-2-4 as a white solid. LC-MS[M+H] + =407.3.
步骤4:中间体D-2的合成Step 4: Synthesis of Intermediate D-2
在氮气保护下,向密闭的20mL的微波反应瓶中依次加入D-2-4(300mg,0.75mmol)溶解在6mL无水DMF中,双联频哪醇硼酸酯(381mg,1.5mmol)、1,1’-二(二苯基膦基)二茂铁二氯化钯的二氯甲烷络合物(61mg,0.075mmol)和乙酸钾(220.5mg,2.25mmol)。然后将该反应液加热到95℃反应5小时。反应毕,将反应液加入少量乙酸乙酯,过硅藻土处理,接下来萃取,分液,有机相浓缩,粗品采用C-18Flash纯化分离得到产物白色固体151mg D-2。LC-MS[M+H]+=407.1。 Under nitrogen protection, add D-2-4 (300 mg, 0.75 mmol) dissolved in 6 mL anhydrous DMF, double pinacol borate (381 mg, 1.5 mmol), and 1,1'-bis(diphenylphosphino)ferrocene palladium dichloride in dichloromethane complex (61 mg, 0.075 mmol) and potassium acetate (220.5 mg, 2.25 mmol). The reaction solution was then heated to 95°C for 5 hours. After the reaction is completed, a small amount of ethyl acetate is added to the reaction solution, and it is treated with diatomaceous earth, followed by extraction, liquid separation, and the organic phase is concentrated. The crude product is purified and isolated using C-18 Flash to obtain 151 mg of D-2 as a white solid product. LC-MS[M+H] + =407.1.
4.3中间体D-3的合成
4.3 Synthesis of intermediate D-3
步骤1:中间体D-3-1的合成Step 1: Synthesis of intermediate D-3-1
将1H-吡咯-2-甲酸甲酯(3.0g,24mmol)溶解在30mL的乙腈中,然后在氮气保护下室温条件下依次向上述反应液中加入NIS(16.7g,74.4mmol)和In(OTf)3(4.0g,7.2mmol)。然后该反应继续在室温反应15小时。反应完毕,旋干乙腈,萃取,分液,无水硫酸钠干燥,过滤,有机相浓缩。粗品采用氘代甲醇进行重结晶,过滤,干燥得到产物棕色固体6.5g,即D-3-1。LC-MS[M+H]+=504.3。1H-NMR(400MHz,DMSO-d6)δ13.04(brs,1H),3.77(s,3H)。Dissolve 1H-pyrrole-2-carboxylic acid methyl ester (3.0g, 24mmol) in 30 mL of acetonitrile, and then add NIS (16.7g, 74.4mmol) and In(OTf) to the above reaction solution in sequence under nitrogen protection at room temperature. ) 3 (4.0g, 7.2mmol). The reaction was then continued at room temperature for 15 hours. After the reaction is completed, the acetonitrile is spun off, extracted, liquid separated, dried over anhydrous sodium sulfate, filtered, and the organic phase is concentrated. The crude product was recrystallized using deuterated methanol, filtered, and dried to obtain 6.5 g of brown solid product, namely D-3-1. LC-MS[M+H] + =504.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ13.04 (brs, 1H), 3.77 (s, 3H).
步骤2:中间体D-3-2的合成Step 2: Synthesis of intermediate D-3-2
将D-3-1(6.5g,12.9mmol)溶解在40mL的氘代甲醇中,然后向反应液中加入Ni(8mL悬浮在氘代甲醇中)和NaOH(2.6g,64.6mmol)后,在氘气环境下置换3次,室温反应4小时。反应毕,将反应液过滤,并用碳酸氢钠水溶液调节PH到7,乙酸乙酯萃取,分液,无水硫酸钠干燥,过滤,有相机浓缩得到产物黄色液体1.5g,即D-3-2为。LC-MS[M+H]+=129.3。Dissolve D-3-1 (6.5g, 12.9mmol) in 40mL of deuterated methanol, and then add Ni (8mL suspended in deuterated methanol) and NaOH (2.6g, 64.6mmol) to the reaction solution. Replace 3 times under deuterium gas environment and react at room temperature for 4 hours. After the reaction is completed, filter the reaction solution, adjust the pH to 7 with sodium bicarbonate aqueous solution, extract with ethyl acetate, separate the liquids, dry with anhydrous sodium sulfate, filter, and concentrate with a camera to obtain 1.5g of the product yellow liquid, namely D-3-2 for. LC-MS[M+H] + =129.3.
步骤3:中间体D-3-3的合成Step 3: Synthesis of intermediate D-3-3
将D-3-2(1.3g,10.15mmol)溶解在18mL乙腈中,然后在0℃下缓慢加入NIS(2.0g,8.88mmol)和In(OTf)3(572mg,1.01mmol)。在氮气惰性环境下,该反应在0℃继续反应2小时。反应毕,将反应液倒入冰水中,乙酸乙酯萃取,分液,无水硫酸钠干燥,过滤,浓缩,柱层析flash纯化得到产物D-3-3为无色液体2.3g。LC-MS[M+H]+=254.3。D-3-2 (1.3g, 10.15mmol) was dissolved in 18 mL acetonitrile, and then NIS (2.0g, 8.88mmol) and In(OTf) 3 (572mg, 1.01mmol) were slowly added at 0°C. The reaction was continued at 0°C for 2 hours under an inert atmosphere of nitrogen. After the reaction is completed, the reaction solution is poured into ice water, extracted with ethyl acetate, separated, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by flash column chromatography to obtain 2.3 g of product D-3-3 as a colorless liquid. LC-MS[M+H] + =254.3.
步骤4:中间体D-3-4的合成Step 4: Synthesis of intermediate D-3-4
将D-3-3(2.3g,9.1mmol)溶解在20mL的二氯甲烷中,然后在0℃条件下依次向反应体系中加入对甲苯磺酰氯(2.6g,13.6mmol)、TEA(2mL)和DMAP(1.1g,9.1mmol)。接着将反应升至室温搅拌过夜。反应毕,将反应液倒入冰水中,萃取,分液,浓缩,硅胶柱层析Flash纯化分离(EA:PE=0~15%) 得到白色固体产物3.0g,即为D-3-4。LC-MS[M+H]+=408.3。1H-NMR(400MHz,DMSO-d6)δ7.95(d,J=6.0Hz,2H),7.48(d,J=8.0Hz,2H),3.68(s,3H),2.40(s,3H)。Dissolve D-3-3 (2.3g, 9.1mmol) in 20mL of methylene chloride, then add p-toluenesulfonyl chloride (2.6g, 13.6mmol) and TEA (2mL) to the reaction system at 0°C. and DMAP (1.1g, 9.1mmol). The reaction was then warmed to room temperature and stirred overnight. After the reaction is completed, pour the reaction solution into ice water, extract, separate, concentrate, and purify and separate using silica gel column chromatography (EA:PE=0~15%) 3.0 g of white solid product was obtained, which was D-3-4. LC-MS[M+H] + =408.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ7.95 (d, J = 6.0 Hz, 2H), 7.48 (d, J = 8.0 Hz, 2H), 3.68 (s, 3H), 2.40 (s, 3H) ).
步骤5:中间体D-3的合成Step 5: Synthesis of intermediate D-3
在氮气保护下,向反应瓶中依次加入D-3-4(3.4g,8.4mmol)溶解在30mL无水DMF中,双联频哪醇硼酸酯(2.3g,9.2mmol)、1,1’-二(二苯基膦基)二茂铁二氯化钯的二氯甲烷络合物(685mg,0.84mmol)和乙酸钾(2.7g,27.6mmol)。然后将该反应液加热到95℃反应2小时。反应毕,将反应液加入少量乙酸乙酯,过硅藻土处理,接下来萃取,分液,有机相浓缩,粗品采用C-18Flash纯化分离得到白色固体产物3.0g即中间体D-3。LC-MS[M+H]+=408.3。Under nitrogen protection, add D-3-4 (3.4g, 8.4mmol) dissolved in 30mL anhydrous DMF, double pinacol borate (2.3g, 9.2mmol), 1,1 '-Bis(diphenylphosphino)ferrocene palladium dichloride in dichloromethane complex (685 mg, 0.84 mmol) and potassium acetate (2.7 g, 27.6 mmol). The reaction solution was then heated to 95°C for 2 hours. After the reaction is completed, a small amount of ethyl acetate is added to the reaction solution, and it is treated with diatomaceous earth, followed by extraction, liquid separation, and the organic phase is concentrated. The crude product is purified and isolated using C-18 Flash to obtain 3.0 g of a white solid product, which is intermediate D-3. LC-MS[M+H] + =408.3.
二、通用合成路线2. General synthetic route
本发明式(I)结构的化合物可通过下述通用制备路线合成。合成路线如下所示:The compound of formula (I) of the present invention can be synthesized by the following general preparation route. The synthesis route is as follows:
反应式Ⅰ:
Reaction formula I:
其中,R1-R2各自独立地选自CH3、CH2D、CHD2或CD3中的任一种,R3-R14各自独立地选自H或D中的一种;X1选自Br、I、OTf或Oms中的任一种;Y为离去基团Ts或Boc。 Wherein, R 1 to R 2 are each independently selected from any one of CH 3 , CH 2 D, CHD 2 or CD 3 , and R 3 to R 14 are each independently selected from one of H or D; X 1 Selected from any one of Br, I, OTf or Oms; Y is the leaving group Ts or Boc.
在优选的一个实施方案中,还涉及 In a preferred embodiment, it also involves
中间体化合物4可以与中间体化合物5通过铃木反应得到中间体化合物6,所述中间体化合物6水解得到中间体化合物7,最后中间体化合物7和中间体化合物8进行缩合反应,即得到式I化合物。Intermediate compound 4 can be reacted with intermediate compound 5 through Suzuki reaction to obtain intermediate compound 6. The intermediate compound 6 is hydrolyzed to obtain intermediate compound 7. Finally, intermediate compound 7 and intermediate compound 8 undergo a condensation reaction to obtain formula I. compound.
中间体化合物4可以通过下述反应式Ⅱ或反应式Ⅲ制备得到的:
Intermediate compound 4 can be prepared by the following reaction formula II or reaction formula III:
其中,R1-R2定义为CH3、CH2D、CHD2、CD3,R3-R14可以定义为H、D。X1定义为OTf,OMs等,X2定义为F、Cl、Br或I等卤素,Y是一个离去基团,可以为Ts,Boc等。Among them, R 1 to R 2 are defined as CH 3 , CH 2 D, CHD 2 , CD 3 , and R 3 to R 14 can be defined as H or D. X 1 is defined as OTf, OMs, etc., X 2 is defined as halogen such as F, Cl, Br or I, Y is a leaving group, which can be Ts, Boc, etc.
具体而言,在反应式Ⅱ中,中间体化合物1经还原反应制备得到中间体化合物2;所述中间体化合物2与中间体化合物3经取代反应得到中间体化合物4;Specifically, in Reaction Formula II, intermediate compound 1 is prepared through reduction reaction to obtain intermediate compound 2; intermediate compound 2 and intermediate compound 3 are subjected to substitution reaction to obtain intermediate compound 4;
在反应式Ⅲ中,中间体化合物1’和中间体化合物2’经还原胺化制备得到中间体化合物4。In Reaction Formula III, intermediate compound 1' and intermediate compound 2' are prepared by reductive amination to obtain intermediate compound 4.
基于上述关键中间体以及通用合成路线,本发明合成获得了以下氘代化合物。Based on the above key intermediates and general synthetic routes, the following deuterated compounds are synthesized in this invention.
典型氘代化合物的合成过程具体如下述实施例。The synthesis process of typical deuterated compounds is detailed in the following examples.
实施例1:Example 1:
1、(S)-4-(5-氯-2-((异丙基-2-基-2-[D])氨基)吡啶-4-基)-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-2-酰胺(氘代化合物1)的合成, 1. (S)-4-(5-chloro-2-((isopropyl-2-yl-2-[D])amino)pyridin-4-yl)-N-(1-(3-chlorobenzene) Synthesis of (ethyl)-2-hydroxyethyl)-1H-pyrrole-2-amide (deuterated compound 1),
结构式为: The structural formula is:
合成步骤如下所示:The synthesis steps are as follows:
1)4-(5-氯-2-((异丙基-2-基-2-[D])氨基)吡啶-4-基)-1-甲苯磺酰基-1H-吡咯-2-甲酸甲酯的合成
1)4-(5-chloro-2-((isopropyl-2-yl-2-[D])amino)pyridin-4-yl)-1-toluenesulfonyl-1H-pyrrole-2-carboxylic acid methyl Synthesis of esters
将5-氯-4-碘-N-(异丙基-2-基-2-[D])吡啶-2-胺(200mg,0.67mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧基硼酸-2-基)-1-甲苯磺酰基-1H-吡咯-2-甲酸甲酯(300mg,0.74mmol)溶解在4mL的1,4-二氧六环中,然后依次向反应瓶中加入四三苯基膦钯(77mg,0.067mmol)和碳酸钠(213mg,2.01mmol)溶解于1mL水中。接着向反应体系通氮气5分钟后,在氮气保护下,升温到100℃微波反应1小时。反应毕,将反应液倒入水中,萃取,分液,有机相浓缩,柱层析flash分离得到黄色固体化合物240mg。LC-MS[M+H]+=449.3。5-Chloro-4-iodo-N-(isopropyl-2-yl-2-[D])pyridin-2-amine (200 mg, 0.67 mmol) and 4-(4,4,5,5-tetrakis Methyl-1,3,2-dioxyboronic acid-2-yl)-1-toluenesulfonyl-1H-pyrrole-2-carboxylate (300 mg, 0.74 mmol) was dissolved in 4 mL of 1,4-dioxanol Six rings, then add tetrakis triphenylphosphine palladium (77 mg, 0.067 mmol) and sodium carbonate (213 mg, 2.01 mmol) to the reaction flask in sequence and dissolve them in 1 mL of water. Then, nitrogen gas was introduced into the reaction system for 5 minutes, and then the temperature was raised to 100°C and microwaved for 1 hour under the protection of nitrogen gas. After the reaction is completed, the reaction solution is poured into water, extracted, separated, the organic phase is concentrated, and 240 mg of the yellow solid compound is obtained by column chromatography flash separation. LC-MS[M+H] + =449.3.
2)4-(5-氯-2-((异丙基-2-基-2-[D])氨基)吡啶-4-基)-1H-吡咯-2-羧酸
2) 4-(5-chloro-2-((isopropyl-2-yl-2-[D])amino)pyridin-4-yl)-1H-pyrrole-2-carboxylic acid
将4-(5-氯-2-((异丙基-2-基-2-[D])氨基)吡啶-4-基)-1-甲苯磺酰基-1H-吡咯-2-甲酸甲酯(300mg,0.67mmol)溶解在3mL的THF中加入到反应瓶中,然后加入氢氧化锂一水合物(169mg,4.02mmol)溶解在1mL水中。将反应加热到75℃反应15小时。将四氢呋喃旋掉,残留液用6N的盐酸调节PH到4-5,析出大量白色固体,过滤,粗品采用pre-TLC纯化得到纯化产物150mg。LC-MS[M+H]+=281.2。4-(5-Chloro-2-((isopropyl-2-yl-2-[D])amino)pyridin-4-yl)-1-toluenesulfonyl-1H-pyrrole-2-carboxylic acid methyl ester (300 mg, 0.67 mmol) dissolved in 3 mL of THF and added to the reaction flask, then lithium hydroxide monohydrate (169 mg, 4.02 mmol) dissolved in 1 mL of water was added. The reaction was heated to 75°C for 15 hours. Spin off the tetrahydrofuran, and adjust the pH of the residual liquid to 4-5 with 6N hydrochloric acid. A large amount of white solid precipitates, and is filtered. The crude product is purified by pre-TLC to obtain 150 mg of the purified product. LC-MS[M+H] + =281.2.
3)(S)-4-(5-氯-2-((异丙基-2-基-2-[D])氨基)吡啶-4-基)-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-2-酰胺
3)(S)-4-(5-chloro-2-((isopropyl-2-yl-2-[D])amino)pyridin-4-yl)-N-(1-(3-chlorobenzene (Hydroxyethyl)-2-Hydroxyethyl)-1H-pyrrole-2-amide
依次将4-(5-氯-2-((异丙基-2-基-2-[D])氨基)吡啶-4-基)-1H-吡咯-2-羧酸(150mg,0.54mmol)、(S)-2-氨基-2-(3-氯苯基)乙-1-醇(109mg,0.64mmol)和HATU(308mg,0.81mmol)溶解的无水DMF中,再加入TEA(164mg,1.62mmol)。将反应瓶抽真空,并充氮气5分钟后,在氮气环境下室温反应2小时。反应毕,将反应液倒入水中,萃取,分液,无水硫酸钠干燥,过滤,浓缩,pre-TLC和HPLC纯化得到白色固体产物18mg,即为氘代化合物1。LC-MS[M+H]+=434.3。1H-NMR(400MHz,DMSO-d6)δ11.88(brs,1H),8.49(d,J=8.4Hz,1H),7.97(s,1H),7.45(s,1H),7.38-7.29(m,5H),6.60(s,1H),6.41(s,1H),5.08-5.03(m,2H),3.67-3.65(m,2H),1.13(s,6H)。4-(5-chloro-2-((isopropyl-2-yl-2-[D])amino)pyridin-4-yl)-1H-pyrrole-2-carboxylic acid (150 mg, 0.54 mmol) , (S)-2-amino-2-(3-chlorophenyl)ethan-1-ol (109mg, 0.64mmol) and HATU (308mg, 0.81mmol) were dissolved in anhydrous DMF, and then TEA (164mg, 1.62mmol). The reaction bottle was evacuated and filled with nitrogen for 5 minutes, and then reacted at room temperature for 2 hours in a nitrogen environment. After the reaction is completed, the reaction solution is poured into water, extracted, separated, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by pre-TLC and HPLC to obtain 18 mg of a white solid product, which is deuterated compound 1. LC-MS[M+H] + =434.3. 1 H-NMR (400MHz, DMSO-d 6 ) δ11.88 (brs, 1H), 8.49 (d, J = 8.4Hz, 1H), 7.97 (s, 1H), 7.45 (s, 1H), 7.38-7.29 (m,5H),6.60(s,1H),6.41(s,1H),5.08-5.03(m,2H),3.67-3.65(m,2H),1.13(s,6H).
2、氘代化合物2和化合物3的合成2. Synthesis of Deuterated Compound 2 and Compound 3
氘代化合物2和氘代化合物3采用各自对应的中间体,可以通过与实施例1相似的合成方法进行合成,其结果如表1所示:Deuterated compound 2 and deuterated compound 3 can be synthesized by using their corresponding intermediates through a synthesis method similar to Example 1, and the results are shown in Table 1:
表1
Table 1
实施例4:Example 4:
1、(S)-4-(5-氯-2-(异丙基氨基)吡啶-4-基)-N-(1-(3-氯苯基)-2-羟基乙基-2,2-[D2])-1H-吡咯-2-酰胺(氘代化合物4)的合成1. (S)-4-(5-chloro-2-(isopropylamino)pyridin-4-yl)-N-(1-(3-chlorophenyl)-2-hydroxyethyl-2,2 Synthesis of -[D2])-1H-pyrrole-2-amide (deuterated compound 4)
结构式为 The structural formula is
合成步骤如下述反应式所示,具体为:
The synthesis steps are as shown in the following reaction formula, specifically:
依次将4-(5-氯-2-((异丙基氨基)吡啶-4-基)-1H-吡咯-2-羧酸(100mg,0.357mmol)、HATU(203.5mg,0.563mmol)和TEA(108mg,1.125mmol)溶解的无水DMF中。将反应瓶抽真空,并充氮气5分钟后,在氮气环境下室温反应30分钟。再将(S)-2-氨基-2-(3-氯苯基)乙-1,1-[D2]-1-醇(67.9mg,0.413mmol)溶解在无水DMF中加入到上述反应液中,继续反应2小时。反应毕,将反应液倒入水中,萃取,分液,无水硫酸钠干燥,过滤,浓缩,pre-TLC和HPLC纯化得到白色固体产物23mg,即为氘代化合物4。LC-MS[M+H]+=435.3。1H-NMR(400MHz,DMSO-d6)δ11.86(brs,1H),8.46(d,J=8.4Hz,1H),7.97(s,1H),7.45(s,1H),7.38-7.30(m,5H),6.60(s,1H),6.43(d,J=7.6Hz,1H),5.06(d,J=8.4Hz,1H),4.96(s,1H),3.99-3.93(m,1H),1.14(d,J=6.4Hz,6H)。4-(5-chloro-2-((isopropylamino)pyridin-4-yl)-1H-pyrrole-2-carboxylic acid (100mg, 0.357mmol), HATU (203.5mg, 0.563mmol) and TEA (108mg, 1.125mmol) was dissolved in anhydrous DMF. The reaction flask was evacuated and filled with nitrogen for 5 minutes, and then reacted at room temperature for 30 minutes in a nitrogen environment. Then (S)-2-amino-2-(3- Chlorophenyl)ethane-1,1-[D2]-1-ol (67.9mg, 0.413mmol) was dissolved in anhydrous DMF and added to the above reaction solution, and the reaction was continued for 2 hours. After the reaction was completed, the reaction solution was poured into in water, extracted, separated, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by pre-TLC and HPLC to obtain 23 mg of white solid product, which is deuterated compound 4. LC-MS [M+H] + = 435.3. 1 H -NMR(400MHz, DMSO-d 6 )δ11.86(brs,1H),8.46(d,J=8.4Hz,1H),7.97(s,1H),7.45(s,1H),7.38-7.30(m ,5H),6.60(s,1H),6.43(d,J=7.6Hz,1H),5.06(d,J=8.4Hz,1H),4.96(s,1H),3.99-3.93(m,1H) ,1.14(d,J=6.4Hz,6H).
2、氘代化合物7-11的合成2. Synthesis of deuterated compounds 7-11
对比化合物1、2,氘代化合物7-11(结构式分别表2所示)基于各自对应的中间体,可以通过与氘代化合物4相似的合成方法合成获得,结果如表2所示:Comparing compounds 1 and 2, deuterated compounds 7-11 (the structural formulas are shown in Table 2 respectively) are based on their corresponding intermediates and can be synthesized by a similar synthesis method to deuterated compound 4. The results are shown in Table 2:
表2


Table 2


实施例10:Example 10:
1、(S)-4-(5-氯-2-(异丙基氨基)吡啶-4-基-3,6-[D2])-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-2-酰胺(氘代化合物12)的合成1. (S)-4-(5-chloro-2-(isopropylamino)pyridin-4-yl-3,6-[D2])-N-(1-(3-chlorophenyl)-2 Synthesis of -hydroxyethyl)-1H-pyrrole-2-amide (deuterated compound 12)
结构式 Structural formula
合成步骤Synthetic steps
步骤1):4-(5-氯-2(异丙基氨基)吡啶-4-基-3,6-[D2])-1-甲苯磺酰基-1H-吡咯-2-甲酸甲酯
Step 1): 4-(5-chloro-2(isopropylamino)pyridin-4-yl-3,6-[D2])-1-toluenesulfonyl-1H-pyrrole-2-carboxylic acid methyl ester
将5-氯-2-(异丙基氨基)吡啶-4-基-3,6-[D2]三氟甲磺酸(100mg,0.32mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧基硼酸-2-基)-1-甲苯磺酰基-1H-吡咯-2-甲酸甲酯(139.22mg,0.343mmol)溶解在2mL的1,4-二氧六环中,然后依次向反应瓶中加入四三苯基膦钯(36.99mg,0.032mmol)和碳酸钾(132.48mg,0.96mmol)溶解于1mL水中。接着向反应体系通氮气5分钟后,在氮气保护下,升温到120℃微波反应1小时。反应毕,将反应液倒入水中,萃取,分液,有机相浓缩,柱层析flash分离得到白色固体产物105mg。LC-MS[M+H]+=450.3。5-Chloro-2-(isopropylamino)pyridin-4-yl-3,6-[D2]trifluoromethanesulfonic acid (100 mg, 0.32 mmol) and 4-(4,4,5,5-tetrafluoromethane Methyl-1,3,2-dioxyboronic acid-2-yl)-1-toluenesulfonyl-1H-pyrrole-2-carboxylate (139.22 mg, 0.343 mmol) was dissolved in 2 mL of 1,4-dioxyboronic acid. Then add tetrakis triphenylphosphine palladium (36.99 mg, 0.032 mmol) and potassium carbonate (132.48 mg, 0.96 mmol) to the reaction flask in sequence and dissolve them in 1 mL of water. Then, nitrogen gas was introduced into the reaction system for 5 minutes, and then the temperature was raised to 120°C and microwaved for 1 hour under the protection of nitrogen gas. After the reaction is completed, the reaction solution is poured into water, extracted, separated, the organic phase is concentrated, and 105 mg of the white solid product is obtained by column chromatography flash separation. LC-MS[M+H] + =450.3.
步骤2):4-(5-氯-2-((异丙基氨基)吡啶-4-基-3,6-[D2])-1H-吡咯-2-羧酸
Step 2): 4-(5-chloro-2-((isopropylamino)pyridin-4-yl-3,6-[D2])-1H-pyrrole-2-carboxylic acid
将4-(5-氯-2(异丙基氨基)吡啶-4-基-3,6-[D2]-1-甲苯磺酰基-1H-吡咯-2-甲酸甲酯(150mg,0.33mmol)溶解在5mL的THF中加入到反应瓶中,然后加入氢氧化锂一水合物(138.6mg,3.3mmol)溶解在2mL水中。将反应加热到78℃反应12小时。将四氢呋喃旋掉,残留液用6N的盐酸调节PH到4-5,析出大量白色固体,过滤,粗品采用C-18flash纯化得到产物90mg。LC-MS[M+H]+=282.2。4-(5-Chloro-2(isopropylamino)pyridin-4-yl-3,6-[D2]-1-toluenesulfonyl-1H-pyrrole-2-carboxylic acid methyl ester (150 mg, 0.33 mmol) Dissolve in 5 mL of THF and add to the reaction flask, then add lithium hydroxide monohydrate (138.6 mg, 3.3 mmol) and dissolve in 2 mL of water. Heat the reaction to 78°C for 12 hours. Spin off the tetrahydrofuran and use the remaining solution with Adjust the pH to 4-5 with 6N hydrochloric acid, and precipitate a large amount of white solid, which is filtered and the crude product is purified by C-18 flash to obtain 90 mg of product. LC-MS [M+H] + =282.2.
步骤3):(S)-4-(5-氯-2-(异丙基氨基)吡啶-4-基-3,6-[D2])-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-2-酰胺
Step 3): (S)-4-(5-chloro-2-(isopropylamino)pyridin-4-yl-3,6-[D2])-N-(1-(3-chlorophenyl) -2-hydroxyethyl)-1H-pyrrole-2-amide
依次将4-(5-氯-2-((异丙基氨基)吡啶-4-基-3,6-[D2])-1H-吡咯-2-羧酸(50mg,0.178mmol),(S)-2-氨基-2-(3-氯苯基)乙-1-醇(33.48mg,0.195mmol),EDCI(37.44mg,0.195mmol),HOBt(29.64mg,0.195mmol)溶解的无水DMF中,再加入DIEA(0.3mL)。将反应瓶抽真空,并充氮气5分钟后,在氮气环境下室温反应12小时。将反应液倒入水中,萃取,分液,浓缩,HPLC纯化得到白色固体45mg,即为氘代化合物12。LC-MS[M+H]+=435.3。1H-NMR(600MHz,DMSO-d6)δ11.92(brs,1H),8.57(d,J=7.8Hz,1H),7.97(s,1H),7.45(s,1H),7.37-7.29(m,4H),6.41(d,J=7.8Hz,1H),5.09-5.04(m,2H),3.98-3.93(m,1H),3.70-3.65(m,2H),1.14(d,J=6.6Hz,6H)。4-(5-chloro-2-((isopropylamino)pyridin-4-yl-3,6-[D2])-1H-pyrrole-2-carboxylic acid (50mg, 0.178mmol), (S )-2-Amino-2-(3-chlorophenyl)ethan-1-ol (33.48mg, 0.195mmol), EDCI (37.44mg, 0.195mmol), HOBt (29.64mg, 0.195mmol) dissolved in anhydrous DMF , then add DIEA (0.3mL). Evacuate the reaction flask and fill it with nitrogen for 5 minutes, then react at room temperature for 12 hours in a nitrogen environment. Pour the reaction solution into water, extract, separate, concentrate, and purify by HPLC to obtain a white color 45 mg of solid is deuterated compound 12. LC-MS [M+H] + = 435.3. 1 H-NMR (600MHz, DMSO-d 6 ) δ11.92 (brs, 1H), 8.57 (d, J = 7.8 Hz,1H),7.97(s,1H),7.45(s,1H),7.37-7.29(m,4H),6.41(d,J=7.8Hz,1H),5.09-5.04(m,2H),3.98 -3.93(m,1H),3.70-3.65(m,2H),1.14(d,J=6.6Hz,6H).
2、氘代化合物13、14的合成2. Synthesis of deuterated compounds 13 and 14
氘代化合物13、14(结构式和合成结果分别如表4所示)采用相对应的中间体与氘代化合物12相似的合成方法进行合成,结果如表3所示:Deuterated compounds 13 and 14 (the structural formulas and synthesis results are shown in Table 4 respectively) were synthesized using the corresponding intermediates and a similar synthesis method to deuterated compound 12, and the results are shown in Table 3:
表3

table 3

实施例13:Example 13:
1、(S)-4-(5-氯-2-(异丙基氨基)吡啶-4-基)-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-3-[D]-2-酰胺(氘代化合物15)的合成1. (S)-4-(5-chloro-2-(isopropylamino)pyridin-4-yl)-N-(1-(3-chlorophenyl)-2-hydroxyethyl)-1H- Synthesis of pyrrole-3-[D]-2-amide (deuterated compound 15)
结构式 Structural formula
步骤1):4-(5-氯-2(异丙基氨基)吡啶-4-基)-1-甲苯磺酰基-1H-吡咯-3-[D]-2-甲酸甲酯
Step 1): 4-(5-chloro-2(isopropylamino)pyridin-4-yl)-1-toluenesulfonyl-1H-pyrrole-3-[D]-2-carboxylic acid methyl ester
将5-氯-4-碘-N-异丙基吡啶-2-胺(1.0g,2.5mmol)和4-(4,4,5,5-四甲基-1,3,2-二氧基硼酸-2-基)-1-甲苯磺酰基-1H-吡咯-3-[D]-2-甲酸甲酯(962mg,3.3mmol)溶解在1,4-二氧六环(10mL)中加入到20mL的微波管中,然后依次四三苯基膦钯(289mg,0.25mmol),Na2CO3(795mg,7.5mmol)溶解在水中。然后向反应瓶中鼓氮气5分钟后,密封,在油浴中加热100℃反应3小时。TLC检测反应毕,将反应液倒入水中,萃取,分离,柱层析flash纯化(EA:PE=0~15%)即得到白色固体1.5g。LC-MS[M+H]+=449.3。 5-Chloro-4-iodo-N-isopropylpyridin-2-amine (1.0g, 2.5mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxy Methyl boronic acid-2-yl)-1-tosyl-1H-pyrrole-3-[D]-2-carboxylate (962 mg, 3.3 mmol) was dissolved in 1,4-dioxane (10 mL) and added into a 20 mL microwave tube, and then dissolve tetrakis triphenylphosphine palladium (289 mg, 0.25 mmol) and Na 2 CO 3 (795 mg, 7.5 mmol) in water in sequence. Then, nitrogen gas was bubbled into the reaction bottle for 5 minutes, sealed, and heated at 100°C in an oil bath for 3 hours. After TLC detection, the reaction was completed. The reaction solution was poured into water, extracted, separated, and purified by flash column chromatography (EA:PE=0~15%) to obtain 1.5g of white solid. LC-MS[M+H] + =449.3.
步骤2):4-(5-氯-2-((异丙基氨基)吡啶-4-基)-1H-吡咯-3-[D]-2-羧酸
Step 2): 4-(5-chloro-2-((isopropylamino)pyridin-4-yl)-1H-pyrrole-3-[D]-2-carboxylic acid
将4-(5-氯-2(异丙基氨基)吡啶-4-基)-1-甲苯磺酰基-1H-吡咯-3-[D]-2-甲酸甲酯(2.0g,4.5mmol)溶解在20mL的THF中加入到反应瓶中,然后加入NaOH(540mg,13.5mmol)溶解在4mL水中。将反应加热到60℃反应6小时。将四氢呋喃旋掉,残留液用6N的盐酸调节PH到4-5,析出大量白色固体,过滤,粗品采用C-18flash纯化得到白色固体180mg。LC-MS[M+H]+=281.3。4-(5-Chloro-2(isopropylamino)pyridin-4-yl)-1-toluenesulfonyl-1H-pyrrole-3-[D]-2-carboxylic acid methyl ester (2.0g, 4.5mmol) Dissolve in 20 mL of THF and add to the reaction flask, then add NaOH (540 mg, 13.5 mmol) and dissolve in 4 mL of water. The reaction was heated to 60°C for 6 hours. Spin off the tetrahydrofuran and adjust the pH of the residual liquid to 4-5 with 6N hydrochloric acid. A large amount of white solid precipitates and is filtered. The crude product is purified using C-18 flash to obtain 180 mg of white solid. LC-MS[M+H] + =281.3.
步骤3):(S)-4-(5-氯-2(异丙基氨基)吡啶-4-基)-N-(1-(3-氯苯基)-2-羟基乙基)-1H-吡咯-3-[D]-2-酰胺
Step 3): (S)-4-(5-chloro-2(isopropylamino)pyridin-4-yl)-N-(1-(3-chlorophenyl)-2-hydroxyethyl)-1H -pyrrole-3-[D]-2-amide
依次将4-(5-氯-2-((异丙基氨基)吡啶-4-基)-1H-吡咯-3-[D]-2-羧酸(100mg,0.36mmol)、HATU(205mg,0.54mmol)和TEA(0.2mL,1.08mmol)溶解的无水DMF中。将反应瓶抽真空,并充氮气5分钟后,在氮气环境下室温反应30分钟。再将(S)-2-氨基-2-(3-氯苯基)乙-1-醇(67mg,0.39mmol)溶解在无水DMF中加入到上述反应液中,继续反应1小时。反应毕,将反应液倒入水中,萃取,分液,无水硫酸钠干燥,过滤,浓缩,pre-TLC和HPLC纯化得到白色固体25mg,即为氘代化合物15。LC-MS[M+H]+=434.3。1H-NMR(400MHz,DMSO-d6)δ11.86(brs,1H),8.47(d,J=8.0Hz,1H),7.97(s,1H),7.45(s,1H),7.38-7.29(m,4H),6.60(s,1H),6.44(d,J=8.0Hz,1H),5.09-4.99(m,2H),3.98-3.93(m,1H),3.69-3.64(m,2H),1.14(d,J=6.4Hz,6H)。4-(5-chloro-2-((isopropylamino)pyridin-4-yl)-1H-pyrrole-3-[D]-2-carboxylic acid (100mg, 0.36mmol), HATU (205mg, 0.54mmol) and TEA (0.2mL, 1.08mmol) dissolved in anhydrous DMF. The reaction flask was evacuated and filled with nitrogen for 5 minutes, and then reacted at room temperature for 30 minutes in a nitrogen environment. Then (S)-2-amino -2-(3-Chlorophenyl)ethan-1-ol (67mg, 0.39mmol) was dissolved in anhydrous DMF and added to the above reaction solution, and the reaction was continued for 1 hour. After the reaction was completed, the reaction solution was poured into water and extracted. , liquid separation, drying over anhydrous sodium sulfate, filtration, concentration, pre-TLC and HPLC purification to obtain 25 mg of white solid, which is deuterated compound 15. LC-MS [M+H] + = 434.3. 1 H-NMR (400MHz ,DMSO-d 6 )δ11.86(brs,1H),8.47(d,J=8.0Hz,1H),7.97(s,1H),7.45(s,1H),7.38-7.29(m,4H), 6.60(s,1H),6.44(d,J=8.0Hz,1H),5.09-4.99(m,2H),3.98-3.93(m,1H),3.69-3.64(m,2H),1.14(d, J=6.4Hz, 6H).
2、氘代化合物16、17的合成2. Synthesis of deuterated compounds 16 and 17
氘代化合物16、17(结构式分别如表4所示)各自采用相应的中间体,可以通过与氘代化合物15相似的合成方法合成获得,其结果如表4所示:Deuterated compounds 16 and 17 (the structural formulas are shown in Table 4 respectively) each adopt corresponding intermediates and can be synthesized through a synthesis method similar to deuterated compound 15. The results are shown in Table 4:
表4:

Table 4:

对照化合物3、4、5、6,以及氘代化合物18-168(结构式分别如表5所示),各自分别采用相应的中间体,可以通过如上述氘代化合物1、4、12或15所示的合成路线合成获得,合成结果如表5所示:Comparative compounds 3, 4, 5, 6, and deuterated compounds 18-168 (structural formulas are shown in Table 5 respectively), each using corresponding intermediates, can be obtained by using the above deuterated compounds 1, 4, 12 or 15. It was synthesized according to the synthetic route shown, and the synthesis results are shown in Table 5:
表5:






























table 5:






























本发明提供的氘代化合物可通过化合物的消旋混合物与光学活性的拆分剂反应形成一对非对映异构体化合物、分离非对映异构体并回收光学纯度的対映体,制备成其单个立体异构体。对映体拆分时可使用本发明化合物的非对映体衍生物进行,优先可解离的复合物(例如,结晶非对映体盐)。非对映体具有显著不同的物理性质(例如,熔点、沸点、溶解度、反应性等),并可通过这些不相似性的优势容易地得到分离。非对映体可通过色谱,优选通过基于溶解度的差异的分离/拆分技术进行分离。然后通过不会消旋化的任何实际手段,回收光学纯对映体,连同拆分试剂。适用于从消旋混合物开始拆分得到化合物立体异构体的技术的更详细的描述可见于Jean Jacques,Andre Collet,Samuel H.Wilen,“对映体、消旋体和拆分”(“Enantiomers,Racemates and Resolutions”),JohnWileyAnd Sons,Inc.,l98l。The deuterated compound provided by the present invention can be prepared by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomer compounds, separating the diastereoisomers and recovering optically pure enantiomers. into its single stereoisomer. Enantioresolution can be performed using diastereomeric derivatives of the compounds of the invention, preferably dissociable complexes (eg, crystalline diastereomeric salts). Diastereomers have significantly different physical properties (eg, melting point, boiling point, solubility, reactivity, etc.) and can be readily separated by taking advantage of these dissimilarities. Diastereomers can be separated by chromatography, preferably by separation/resolution techniques based on differences in solubility. The optically pure enantiomers are then recovered, together with the resolving reagents, by any practical means that does not involve racemization. A more detailed description of techniques suitable for the resolution of stereoisomers of compounds starting from racemic mixtures can be found in Jean Jacques, Andre Collet, Samuel H. Wilen, "Enantiomers, Racemates and Resolutions" , Racemates and Resolutions"), John Wiley And Sons, Inc., l98l.
实验例1:ERK1、ERK2激酶实验Experimental example 1: ERK1, ERK2 kinase experiment
1、实验目的:1. Experimental purpose:
测定本发明提供的上述氘代化合物对于ERK1、ERK2的抑制活性。 The inhibitory activity of the above deuterated compounds provided by the present invention on ERK1 and ERK2 was determined.
2、实验仪器、耗材和试剂,如表6所示:2. Experimental instruments, consumables and reagents, as shown in Table 6:
表6:
Table 6:
3、实验过程3. Experimental process
(1)实验对象:阳性对照物BVD-523(根据公开于专利WO2005113541中的合成方法合成)和本发明提供的部分的氘代化合物。(1) Experimental objects: positive control BVD-523 (synthesized according to the synthesis method disclosed in patent WO2005113541) and some of the deuterated compounds provided by the present invention.
(2)实验准备:(2)Experimental preparation:
A.溶液配置A.Solution configuration
(1)4×kinase reaction buffer:使用5×kinase reaction buffer稀释+终浓度为200nM的DTT;(1) 4×kinase reaction buffer: Use 5×kinase reaction buffer to dilute + DTT with a final concentration of 200nM;
(2)ERK1激酶工作液(2.5×):100ng/μL的母液用buffer稀释50倍,配置成2ng/μL的工作液;(2) ERK1 kinase working solution (2.5×): 100ng/μL stock solution is diluted 50 times with buffer to prepare a 2ng/μL working solution;
(3)ERK2激酶工作液(2.5×):100ng/μL的母液用buffer稀释40倍,配置成2.5ng/μL的工作液;(3) ERK2 kinase working solution (2.5×): 100ng/μL stock solution is diluted 40 times with buffer to prepare a 2.5ng/μL working solution;
(4)substrate/ATP MIX工作液(2.5×):10mM的ATP母液和1mg/mL的substrate母液用buffer稀释80倍、4倍,配置成含125μMATP、0.25mg/mL substrate的MIX工作液;(4) Substrate/ATP MIX working solution (2.5×): 10mM ATP stock solution and 1mg/mL substrate stock solution are diluted 80 times and 4 times with buffer, and configured into a MIX working solution containing 125μ MTP and 0.25mg/mL substrate;
B.化合物工作液的配置B. Configuration of compound working solution
a.取2μL的10mM的对照化合物或者待测化合物的母液,使用38μL DMSO稀释至0.5mM起始浓度,混匀,加到96孔板的B1孔中。a. Take 2μL of 10mM control compound or the stock solution of the compound to be tested, dilute it to a starting concentration of 0.5mM with 38μL DMSO, mix well, and add it to well B1 of a 96-well plate.
b.在B2-B11孔中加入25μL DMSO,从B1孔中取5μL溶液加到B2孔 中,混匀,依次稀释到B9孔,B10-B11孔不稀释(只有DMSO,10孔为空白对照),得到依次6倍稀释的溶液。b. Add 25 μL DMSO to wells B2-B11, and add 5 μL solution from well B1 to well B2. medium, mix well, and dilute it into well B9 in sequence, and holes B10-B11 are not diluted (only DMSO, well 10 is a blank control), to obtain a solution diluted 6 times in sequence.
c.在96孔板C1-C11孔中加入38μL的1×kinase reactionbuffer,取B1-B11孔稀释溶液2μL加到C1-C11中,混匀,配置成首孔浓度25μM,6倍稀释的工作液。c. Add 38 μL of 1×kinase reaction buffer to the C1-C11 wells of the 96-well plate, add 2 μL of the dilute solution from B1-B11 holes to C1-C11, mix well, and prepare a working solution with a concentration of 25 μM in the first well and a 6-fold dilution. .
(3)实验步骤:(3) Experimental steps:
a)取4μL激酶工作液液加入到384孔板中(第11孔为阴性对照,不加激酶溶液,以1×kinase reactionbuffer代替),对应孔再加2μL各浓度的化合物工作液,1000rpm离心1min,室温300rpm孵育10min;a) Add 4 μL of the kinase working solution into the 384-well plate (the 11th well is a negative control, no kinase solution is added, replaced by 1×kinase reactionbuffer), add 2 μL of the compound working solution of each concentration to the corresponding well, and centrifuge at 1000 rpm for 1 min. , incubate at room temperature 300rpm for 10min;
b)对应每孔加入4μL substrate/ATP MIX工作液,1000rpm离心1min,ERK1激酶37℃300rpm孵育60min;ERK2激酶室温300rpm孵育60min;b) Add 4 μL substrate/ATP MIX working solution to each well, centrifuge at 1000 rpm for 1 min, and incubate ERK1 kinase at 37°C and 300 rpm for 60 min; ERK2 kinase incubate at room temperature at 300 rpm for 60 min;
c)每孔加入10μL的ADP-Glo reagent,1000rpm离心1min,室温300rpm孵育50min;c) Add 10 μL of ADP-Glo reagent to each well, centrifuge at 1000 rpm for 1 min, and incubate at 300 rpm at room temperature for 50 min;
d)每孔加入20μL的kinase detection reagent,1000rpm离心1min,室温300rpm孵育40min;d) Add 20 μL of kinase detection reagent to each well, centrifuge at 1000 rpm for 1 min, and incubate at room temperature at 300 rpm for 40 min;
e)酶标仪读取Luminescence下的数值,并记录;e) Read the value under Luminescence with a microplate reader and record it;
f)数据处理f)Data processing
按如下公式计算各化合物在各浓度点的抑制率,并通过软件Graphpad Prism 8.0进行曲线拟合得到IC50值。
The inhibition rate of each compound at each concentration point was calculated according to the following formula, and the IC 50 value was obtained by curve fitting using the software Graphpad Prism 8.0.
4、实验结果4. Experimental results
表7:



NT: No Tested。
Table 7:



NT: No Tested.
由表7中的实验数据可见,与未氘代的对照化合物BVD-523相比,整体而言,本发明提供的供试氘代化合物对ERK1激酶和ERK2激酶的抑制活性与对照化合物相当。It can be seen from the experimental data in Table 7 that compared with the non-deuterated control compound BVD-523, overall, the inhibitory activity of the test deuterated compound provided by the present invention on ERK1 kinase and ERK2 kinase is equivalent to that of the control compound.
实验例2本发明的部分氘代化合物对A375细胞、Colo 205细胞、HCT116细胞的增殖抑制测定Experimental Example 2 Determination of proliferation inhibition of A375 cells, Colo 205 cells, and HCT116 cells by some deuterated compounds of the present invention
1、实验目的1. Experimental purpose
测定本发明的化合物对于A375细胞、Colo205细胞、HCT116细胞的增殖的抑制活性Determination of the inhibitory activity of the compound of the present invention on the proliferation of A375 cells, Colo205 cells, and HCT116 cells
2、实验仪器、耗材和试剂,如表8所示:2. Experimental instruments, consumables and reagents, as shown in Table 8:
表8
Table 8
3、实验过程:3. Experimental process:
(1)实验对象:阳性对照物BVD-523,以及本发明提供的上述氘代化合 物。(1) Experimental objects: positive control BVD-523, and the above-mentioned deuterated compounds provided by the present invention things.
(2)实验准备:(2)Experimental preparation:
A、培养基配置A. Medium configuration
a)人恶性黑色素瘤细胞A375培养基配制:DMEM(高糖)+10%FBS+1%Penicillin-Streptomycina) Human malignant melanoma cell A375 medium preparation: DMEM (high sugar) + 10% FBS + 1% Penicillin-Streptomycin
b)人结肠癌细胞colo 205培养基配制:1640+10%FBS+1%Penicillin-Streptomycinb) Human colon cancer cell colo 205 medium preparation: 1640+10% FBS+1% Penicillin-Streptomycin
c)人结肠癌细胞HCT116培养基配制:McCoy's 5A+10%FBS+1%Penicillin-Streptomycinc) Human colon cancer cell HCT116 medium preparation: McCoy's 5A+10% FBS+1% Penicillin-Streptomycin
B、化合物工作液的配置B. Configuration of compound working fluid
a).取4μL的10mM的对照化合物或待测化合物的母液,使用16μL DMSO稀释至2mM起始浓度,混匀,加到96孔板的B1孔中。a). Take 4μL of 10mM control compound or the mother solution of the test compound, dilute it to a starting concentration of 2mM with 16μL DMSO, mix well, and add it to well B1 of a 96-well plate.
b).在B2-B10孔中加入12μL DMSO,从B1孔中取8μL溶液加到B2孔中,混匀,依次稀释到B9孔,B10孔不稀释(只有DMSO,为空白对照孔),得到依次2.5倍稀释的溶液。b). Add 12 μL DMSO to the B2-B10 holes, add 8 μL solution from the B1 hole to the B2 hole, mix, and dilute it to the B9 hole in sequence. The B10 hole is not diluted (only DMSO is the blank control hole), and we get Dilute the solution 2.5 times in sequence.
c).在96孔板C1-C10孔中加入95μL的培养基,取B1-B10孔稀释溶液5μL加到C1-C10中,混匀,配置成首孔浓度100μM,2.5倍稀释的工作液。c). Add 95 μL of culture medium to wells C1-C10 of a 96-well plate, add 5 μL of the dilute solution from wells B1-B10 to C1-C10, mix well, and prepare a working solution with a concentration of 100 μM in the first well and a 2.5-fold dilution.
(3)实验步骤:(3) Experimental steps:
a)A375细胞分别以2000细胞/90μL/孔接种至96孔平底透明细胞板,colo 205细胞分别以1000细胞/90μL/孔接种至96孔平底透明细胞板,HCT116细胞分别以600细胞/90μL/孔接种至96孔平底透明细胞板,最外圈加入200μL PBS,于37℃,5%CO2培养24h;a) A375 cells were seeded into a 96-well flat-bottom clear cell plate at 2000 cells/90μL/well, colo 205 cells were seeded into a 96-well flat-bottom clear cell plate at 1000 cells/90μL/well, and HCT116 cells were seeded at 600 cells/90μL/ The wells were inoculated into a 96-well flat-bottom transparent cell plate, 200 μL PBS was added to the outermost circle, and cultured at 37°C and 5% CO2 for 24 hours;
b)取出96孔板,将上述稀释好的化合物工作液分别以10μL/孔加入至接种有细胞的培养板中,于37℃,5%CO2培养120h;b) Take out the 96-well plate, add the above diluted compound working solution at 10 μL/well to the culture plate seeded with cells, and incubate at 37°C and 5% CO 2 for 120 hours;
c)取出96孔板,于室温下加入Cell Titer-Glo Reagent试剂,50μL/well,室温避光300rpm孵育30min,转移100μL至96孔白板。c) Take out the 96-well plate, add Cell Titer-Glo Reagent reagent at room temperature, 50 μL/well, incubate at room temperature at 300 rpm for 30 minutes in the dark, and transfer 100 μL to the 96-well white plate.
d)酶标仪读取Luminescence下的数值,并记录;d) Read the value under Luminescence with a microplate reader and record it;
e)数据处理e)Data processing
按如下公式计算各化合物在各浓度点的抑制率,并通过软件Graphpad Prism 8.0进行曲线拟合得到IC50值。
The inhibition rate of each compound at each concentration point was calculated according to the following formula, and the IC 50 value was obtained by curve fitting using the software Graphpad Prism 8.0.
4、实验结果:4. Experimental results:
表9:实验对象对A375细胞、Colo205细胞和HCT116细胞的抑制活性检测结果

NT:No Tested。
Table 9: Test results of inhibitory activity of experimental subjects on A375 cells, Colo205 cells and HCT116 cells

NT:No Tested.
由表9实验数据可见,与未氘代的对照化合物BVD-523相比,本发明提 供的供试氘代化合物对A375细胞、Colo205细胞、HCT116细胞的抑制活性与对照化合物相比活性更优或者相当。It can be seen from the experimental data in Table 9 that compared with the non-deuterated control compound BVD-523, the present invention has The inhibitory activity of the deuterated compounds provided for testing on A375 cells, Colo205 cells, and HCT116 cells is better or equivalent to that of the control compounds.
实验例3:本发明的氘代化合物在肝微粒体中的稳定性Experimental Example 3: Stability of the deuterated compound of the present invention in liver microsomes
1、实验目的:1. Experimental purpose:
测定本发明的化合物在肝微粒体中的代谢稳定性。The metabolic stability of the compounds of the invention in liver microsomes was determined.
2、实验耗材:2. Experimental consumables:
微粒体保存于-80℃冰箱,具体信息见表10。使用之前先于37℃水浴中融化,溶化后放置于冰上待用。其他试剂均为通用试剂。Microsomes were stored in a -80°C refrigerator. See Table 10 for detailed information. Before use, melt in a 37°C water bath, then place on ice until used. Other reagents are general reagents.
表10:
Table 10:
3、实验过程:3. Experimental process:
1)对照化合物:BVD-523,对照化合物1-6,其他均为待测化合物。1) Control compounds: BVD-523, control compounds 1-6, and the others are compounds to be tested.
2)按照下表配置反应体系,准备开始实验。
2) Configure the reaction system according to the table below and prepare to start the experiment.
3)将该反应体系放在37℃水浴中预孵育10分钟。向反应体系中加入40μL10mM NADPH溶液,NADPH的最终浓度分别为1mM。用40μL超纯水代替NADPH溶液作为阴性对照。3) Pre-incubate the reaction system in a 37°C water bath for 10 minutes. Add 40 μL of 10mM NADPH solution to the reaction system, and the final concentration of NADPH is 1mM. Use 40 μL ultrapure water instead of NADPH solution as a negative control.
4)在反应中加入4μL的100μM的待测化合物和对照药物启动反应,药物的最终浓度为1μM。4) Add 4 μL of 100 μM test compound and control drug to the reaction to start the reaction. The final concentration of the drug is 1 μM.
5)在0、5、15、30、45和60分钟分别取出50μL反应样品,用4倍的含有内标(200nM阿普***、200nM拉贝洛尔、2μM酮洛芬、200nM咖啡因)的冷乙腈淬灭。样品在3,220g转速下离心45分钟。离心完成后取90μL上清液和90μL超纯水混匀用于LC-MS/MS分析检测。5) Take out 50μL reaction samples at 0, 5, 15, 30, 45 and 60 minutes respectively, and use 4 times the amount of internal standard (200nM alprazolam, 200nM labetalol, 2μM ketoprofen, 200nM caffeine) Quench with cold acetonitrile. Samples were centrifuged at 3,220g for 45 minutes. After centrifugation, take 90 μL of the supernatant and mix it with 90 μL of ultrapure water for LC-MS/MS analysis and detection.
4、数据分析4. Data analysis
所有的数据计算均通过Microsoft Excel软件进行。通过提取离子图谱检 测峰面积。通过对母药的消失百分比与时间进行线性拟合,检测母药的体外半衰期(T1/2)。体外(T1/2)通过斜率计算:
in vitro t1/2=-(0.693/k)
All data calculations were performed using Microsoft Excel software. Detection by extracted ion spectrum Measure the peak area. The in vitro half-life (T 1/2 ) of the parent drug was detected by linearly fitting the disappearance percentage of the parent drug with time. In vitro (T 1/2 ) is calculated from the slope:
in vitro t 1/2 =-(0.693/k)
用下列公式把体外(T1/2)转化为体外清除率(单位μL/min/mg):
Use the following formula to convert in vitro (T 1/2 ) into in vitro clearance (unit μL/min/mg):
5、实验结果5. Experimental results
(1)本发明提供的对照化合物和示例性氘代化合物在人肝微粒中代谢稳定性的实验结果如表11所示:(1) The experimental results of the metabolic stability of the control compounds and exemplary deuterated compounds provided by the present invention in human liver microparticles are as shown in Table 11:
表11:



NT:No Tested。
Table 11:



NT:No Tested.
(2)本发明提供的对照化合物和示例性氘代化合物在在人和猴的肝微粒代谢稳定性(2) The stability of the control compounds and exemplary deuterated compounds provided by the present invention in the liver microparticle metabolism of humans and monkeys
实验结果如表12所示:The experimental results are shown in Table 12:
表12

Table 12

由表11、12数据可以看出,氘代化合物28、39、41、82、95、105、107、在人,猴的肝微粒体中代谢稳定性均优于BVD-523,尤其是氘代化合物28、39、41、82、105,其在人肝微粒体半衰期是BVD-523的2-4倍,同时在其他种属动物的肝微粒稳定性实验结果展示出一致的结果,半衰期也同样提升了2-4倍。说明本发明中的部分氘代化合物具有更长的作用时间,可具有更好的临床应用价值,有望解决BVD-523目前临床给药剂量600mgBID可能因为代谢带来的副作用及限制BVD-523扩展更多适应症,从而让更多患者受益。 It can be seen from the data in Tables 11 and 12 that the metabolic stability of deuterated compounds 28, 39, 41, 82, 95, 105, 107 in human and monkey liver microsomes is better than that of BVD-523, especially the deuterated compounds The half-life of compounds 28, 39, 41, 82, and 105 in human liver microsomes is 2-4 times that of BVD-523. At the same time, the stability experiments of liver microsomes in other species of animals show consistent results, and the half-life is also the same. Improved by 2-4 times. It shows that some of the deuterated compounds in the present invention have longer action time and can have better clinical application value, which is expected to solve the possible side effects caused by metabolism of BVD-523 at the current clinical dosage of 600 mg BID and limit the expansion of BVD-523. Multiple indications to benefit more patients.
同时,从上述人微粒体的代谢稳定性试验结果进一步证明,相对于BVD-523的氘代位点的数量而言,并非氘代位点越多的化合物的代谢稳定性更高。另外,在某些位点发生氘代的化合物(例如对照化合物1、2和6)不仅半衰期没有延长,反而缩短了,该氘代化合物的药代动力学性质发生了劣化。At the same time, the above-mentioned metabolic stability test results of human microsomes further prove that compared to the number of deuterated sites of BVD-523, compounds with more deuterated sites do not have higher metabolic stability. In addition, the half-life of compounds deuterated at certain sites (such as reference compounds 1, 2 and 6) was not extended but shortened, and the pharmacokinetic properties of the deuterated compounds were deteriorated.
实验例4:本发明部分氘代化合物在肝细胞中的稳定性Experimental Example 4: Stability of some deuterated compounds of the present invention in liver cells
1、实验目的:1. Experimental purpose:
进一步验证氘代化合物28、39、82和BVD-523的代谢稳定性。The metabolic stability of deuterated compounds 28, 39, 82 and BVD-523 was further verified.
2、实验耗材:2. Experimental consumables:
肝细胞保存于液氮中,具体信息见表13。Liver cells were stored in liquid nitrogen. See Table 13 for detailed information.
表13:
Table 13:
3、实验过程:3. Experimental process:
(1)对照化合物:BVD-523。其他均为受试化合物。(1) Control compound: BVD-523. Others are test compounds.
(2)化合物工作液的制备(2) Preparation of compound working solution
将受试化合物和对照化合物BVD-523用DMSO配置成高浓度储备液,使用前用50%乙腈/水稀释到100μM的工作液,受试化合物和BVD-523的终浓度为1μM。The test compound and the control compound BVD-523 were prepared into a high-concentration stock solution using DMSO, and diluted to a 100 μM working solution with 50% acetonitrile/water before use. The final concentration of the test compound and BVD-523 was 1 μM.
(3)肝细胞的制备(3) Preparation of liver cells
a)混合49.5mLWilliams’Medium E和0.5mL GlutaMAX作为孵育液。将肝细胞复苏液和孵育液于使用前置于37℃水浴中预热。a) Mix 49.5mLWilliams’Medium E and 0.5mL GlutaMAX as incubation solution. Preheat the hepatocyte recovery solution and incubation solution in a 37°C water bath before use.
b)取一管超低温保存的肝细胞,将肝细胞迅速置于37℃水浴中并轻摇直至所有冰晶全部分散,喷洒70%乙醇后转移至生物安全柜中。b) Take a tube of cryopreserved liver cells, quickly place the liver cells in a 37°C water bath and shake gently until all ice crystals are dispersed, spray 70% ethanol and transfer to a biological safety cabinet.
c)将肝细胞小管的内容物倾入盛有50mL复苏培养基的离心管中,将其于离心力为100g下离心10分钟。离心后,吸出复苏培养基并加入足量孵育培养基得到细胞密度约1.5×106个细胞/mL的细胞混悬液。c) Pour the contents of the hepatocyte tubule into a centrifuge tube containing 50 mL of recovery medium, and centrifuge it at 100 g for 10 minutes. After centrifugation, aspirate the recovery medium and add sufficient incubation medium to obtain a cell suspension with a cell density of approximately 1.5×10 6 cells/mL.
d)用AOPI染液对肝细胞进行计数及确定活细胞密度,肝细胞成活率必须大于75%。利用孵育培养基稀释肝细胞混悬液至活细胞密度为0.5×106个细胞/mL。 d) Use AOPI staining solution to count liver cells and determine the density of viable cells. The survival rate of liver cells must be greater than 75%. Use the incubation medium to dilute the hepatocyte suspension to a viable cell density of 0.5 × 10 6 cells/mL.
e)将一部分密度为0.5×106个细胞/mL的肝细胞混悬液开水煮沸灭活5分钟作为阴性对照。细胞灭活后,便于考察非细胞酶系介导的底物转化。e) Inactivate a portion of the hepatocyte suspension with a density of 0.5×10 6 cells/mL by boiling water for 5 minutes as a negative control. After cells are inactivated, substrate conversion mediated by non-cellular enzymes can be easily investigated.
(4)实验方法(4) Experimental methods
a)转移198μL活细胞或者灭活细胞的混悬液到96孔深孔板,将深孔板置于涡旋上于孵箱中预热10分钟。活细胞进行双平行孵育,灭活细胞进行单平行孵育。a) Transfer 198 μL of live or inactivated cell suspension to a 96-well deep-well plate, place the deep-well plate on a vortex and preheat it in the incubator for 10 minutes. Live cells were incubated in double parallels, and inactivated cells were incubated in single parallels.
b)每孔加入2μL 100μM受试物或BVD-523进行反应起始,将深孔板放回孵箱涡旋器上。b) Add 2 μL of 100 μM test substance or BVD-523 to each well to start the reaction, and put the deep well plate back on the incubator vortexer.
c)孵育样品,分别于0、15、30、60、90和120分钟,取25μL混悬液,加入150μL含内标的乙腈(200nM阿普***、200nM拉贝洛尔、2μM酮洛芬、200nM咖啡因)终止反应。涡旋10分钟,于离心力为3220g、4℃条件离心30分钟,离心完成后取100μL上清液和100μL超纯水混匀用于UPLC-MS/MS分析检测。c) Incubate the sample. At 0, 15, 30, 60, 90 and 120 minutes, take 25 μL of the suspension and add 150 μL of acetonitrile containing internal standard (200 nM alprazolam, 200 nM labetalol, 2 μM ketoprofen, 200 nM caffeine) to terminate the reaction. Vortex for 10 minutes, and centrifuge for 30 minutes at a centrifugal force of 3220g and 4°C. After centrifugation, take 100 μL of the supernatant and mix it with 100 μL of ultrapure water for UPLC-MS/MS analysis and detection.
4、数据分析4. Data analysis
所有的数据计算均通过Microsoft Excel软件进行。通过提取离子图谱检测峰面积。通过对母药消除百分比的自然对数与时间进行线性拟合,检测母药的体外半衰期(T1/2)。All data calculations were performed using Microsoft Excel software. Peak areas were detected by extracted ion spectra. The in vitro half-life (T 1/2 ) of the parent drug was determined by linearly fitting the natural logarithm of the percent elimination of the parent drug with time.
体外半衰期(T1/2)通过斜率计算:
in vitro t1/2=0.693/k
The in vitro half-life (T 1/2 ) is calculated from the slope:
in vitro t 1/2 =0.693/k
体外固有清除率(单位μL/min/106个细胞)用下列公式计算:
in vitro CLint=kV/N
V=每孔孵育体积(0.2mL);
N=每孔细胞的数量(0.1×106个细胞)
The intrinsic clearance rate in vitro (unit μL/min/106 cells) is calculated using the following formula:
in vitro CL int =kV/N
V=incubation volume per well (0.2mL);
N = number of cells per well (0.1×10 6 cells)
5、氘代化合物28、39、82以及BVD-523在人或猴的肝细胞中代谢稳定性实验结果如表14所示:5. The experimental results of the metabolic stability of deuterated compounds 28, 39, 82 and BVD-523 in human or monkey liver cells are shown in Table 14:
表14

Table 14

由表14数据可以看出,氘代化合物28、39、82在人肝细胞中的代谢稳定性优于BVD-523,尤其是在猴肝细胞的中的稳定性远优于BVD-523。进一步验证了发明的氘代化合物28、39、82的代谢性质相对于对照化合物BVD-523有显著的优化,进而有助于临床用药适应症的扩展。It can be seen from the data in Table 14 that the metabolic stability of deuterated compounds 28, 39, and 82 in human hepatocytes is better than that of BVD-523, especially the stability in monkey hepatocytes is much better than that of BVD-523. It is further verified that the metabolic properties of the invented deuterated compounds 28, 39, and 82 are significantly optimized compared to the control compound BVD-523, which in turn contributes to the expansion of clinical drug indications.
实验例5:氘代化合物在猴中PK实验Experimental Example 5: PK test of deuterated compounds in monkeys
1、实验目的1. Experimental purpose
以本发明提供的氘代化合物39为例,进一步验证本发明提供的氘代化合物39和BVD-523在猴中体内的代谢稳定性Taking the deuterated compound 39 provided by the present invention as an example, the metabolic stability of the deuterated compound 39 and BVD-523 provided by the present invention in monkeys is further verified.
2、实验步骤2. Experimental steps
(1)实验制剂配制:(1) Experimental preparation preparation:
A)BVD-523静脉注射制剂配制过程:A) Preparation process of BVD-523 intravenous injection preparation:
a)称取16.62mg BVD-523于100mL药瓶中;a) Weigh 16.62mg BVD-523 into a 100mL medicine bottle;
b)向瓶中加入1.662mL DMSO和13.296mL PEG 400,涡旋均匀后超声至澄清透明;b) Add 1.662mL DMSO and 13.296mL PEG 400 to the bottle, vortex evenly and sonicate until clear and transparent;
c)向瓶中加入18.282mL 10%HP-β-CD的生理盐水溶液,涡旋均匀;c) Add 18.282mL of 10% HP-β-CD physiological saline solution to the bottle and vortex evenly;
d)将配置好的溶液用0.22um滤膜过滤。备用。d) Filter the prepared solution with a 0.22um filter membrane. spare.
B)BVD-523灌胃制剂配制过程:B) Preparation process of BVD-523 intragastric preparation:
a)称取157.07mg BVD-523于250mL药瓶中;a) Weigh 157.07mg BVD-523 into a 250mL medicine bottle;
b)向瓶中加入3.927mL DMSO和31.414mL PEG 400,涡旋均匀后超声至澄清透明;b) Add 3.927mL DMSO and 31.414mL PEG 400 to the bottle, vortex evenly and sonicate until clear and transparent;
c)向瓶中加入43.194mL 10%HP-β-CD的生理盐水溶液,涡旋均匀。备用。c) Add 43.194mL of 10% HP-β-CD physiological saline solution into the bottle and vortex evenly. spare.
C)氘化化合物39静脉注射制剂配制过程:C) Preparation process of intravenous injection preparation of deuterated compound 39:
a)称取18.36mg氘代化合物39于100mL药瓶中; a) Weigh 18.36mg of deuterated compound 39 into a 100mL vial;
b)向瓶中加入1.836mL DMSO和14.688mL PEG 400,涡旋均匀后超声至澄清透明;b) Add 1.836mL DMSO and 14.688mL PEG 400 to the bottle, vortex evenly and sonicate until clear and transparent;
c)向瓶中加入20.196mL 10%HP-β-CD的生理盐水溶液,涡旋均匀。c) Add 20.196mL of 10% HP-β-CD physiological saline solution to the bottle and vortex evenly.
d)将配置好的溶液用0.22um滤膜过滤。备用。d) Filter the prepared solution with a 0.22um filter membrane. spare.
D)氘代化合物39灌胃制剂配制过程:D) Preparation process of deuterated compound 39 intragastric preparation:
a)称取180.70mg氘代化合物39于250mL药瓶中;a) Weigh 180.70mg of deuterated compound 39 into a 250mL medicine bottle;
b)向瓶中加入4.518mL DMSO和36.140mL PEG 400,涡旋均匀后超声至澄清透明;b) Add 4.518mL DMSO and 36.140mL PEG 400 to the bottle, vortex evenly and sonicate until clear and transparent;
c)向瓶中加入49.693mL 10%HP-β-CD的生理盐水溶液,涡旋均匀。备用。c) Add 49.693mL of 10% HP-β-CD physiological saline solution into the bottle and vortex evenly. spare.
(2)给药过程:(2) Administration process:
a)给药前猴子从笼子里抓出来,称重,放在猴椅上;a) Before administration, the monkey is taken out of the cage, weighed, and placed on the monkey chair;
b)根据动物体重计算给药量;b) Calculate the dosage based on the animal’s weight;
c)收到化合物以后,摇匀,抽药前用注射器吸取吹打制剂,使之混合均匀;c) After receiving the compound, shake it well, and use a syringe to pipette the preparation before withdrawing the medicine to mix it evenly;
d)抽取指定体积的制剂;d) Draw the specified volume of preparation;
e)猴子插管,判断灌胃管在食道或胃部,把制剂通过灌胃管完成给药,用5毫升空气冲洗灌胃管,确保制剂完全给到动物体内。e) Intubate the monkey, determine whether the gavage tube is in the esophagus or stomach, complete the administration of the preparation through the gavage tube, and flush the gavage tube with 5 ml of air to ensure that the preparation is completely administered to the animal.
(3)采血过程:(3) Blood collection process:
1小时采血点前动物会在猴椅上观察临床症状和采血,1小时之后动物放回原笼子,之后的采血和观察在笼子里完成。采血主要通过前肢头静脉和后肢隐静脉,用1毫升注射器直接采集,采集之后打入真空采血管中,并在湿冰上转移并离心,离心时间记录在离心表上。采完血样用于UPLC-MS/MS分析检测。PK参数采用WinNonlin 6.1非室模型估计。The animals will be observed for clinical symptoms and blood collection on the monkey chair 1 hour before the blood collection point. After 1 hour, the animals will be returned to the original cage, and subsequent blood collection and observation will be completed in the cage. Blood is collected mainly through the cephalic vein of the forelimb and the saphenous vein of the hind limb, directly collected with a 1 ml syringe. After collection, it is poured into a vacuum blood collection tube, transferred and centrifuged on wet ice, and the centrifugation time is recorded on the centrifuge table. Blood samples were collected for UPLC-MS/MS analysis and detection. PK parameters were estimated using WinNonlin 6.1 non-compartmental model.
氘代化合物28的实验操作按照上述操作平行进行。数据汇总到结果中。The experimental operation of deuterated compound 28 was carried out in parallel with the above operation. Data are summarized into results.
(4)实验结果:(4)Experimental results:
化合物BVD-523和氘代化合物28、39的静脉给药(iv)和口服灌胃给药(po)的在猴中的实验结果如表15、图1、图2及图3所示: The experimental results of intravenous administration (iv) and oral administration (po) of compound BVD-523 and deuterated compounds 28 and 39 in monkeys are shown in Table 15, Figure 1, Figure 2 and Figure 3:
表15
Table 15
AUC0-∞=药物从零时间至所有原形药物全部消除这一段时间的血药浓度-时曲线下总面积,C0=静脉注射后瞬时的血药浓度,Cmax=最大实测血药浓度,Tmax=最大实测血药浓度对应的时间,T1/2=末端消除半衰期,Cl=清除率,Vd=表观分布容积,F=生物利用度AUC 0-∞ = the total area under the blood drug concentration-time curve from time zero to the time when all prototype drugs are eliminated, C 0 = the instantaneous blood drug concentration after intravenous injection, C max = the maximum measured blood drug concentration, T max = time corresponding to the maximum measured blood drug concentration, T 1/2 = terminal elimination half-life, Cl = clearance rate, Vd = apparent volume of distribution, F = bioavailability
由表15、图1、图2、图3数据可见,在猴中PK数据显示氘代化合物28、39显著优于BVD-523,尤其是通过口服给药的生物利用度以及浓度曲线下面积(AUC)暴露量都显著优于BVD-523。本发明提供的氘代化合物28、39能够有效改善对照化合物BVD-523的口服生物利用度低的缺陷,从而有望克服临床用药个体差异的缺陷,最终使患者受益。It can be seen from the data in Table 15, Figure 1, Figure 2, and Figure 3 that the PK data in monkeys show that deuterated compounds 28 and 39 are significantly better than BVD-523, especially the bioavailability and area under the concentration curve of oral administration ( AUC) exposures were significantly better than BVD-523. The deuterated compounds 28 and 39 provided by the present invention can effectively improve the defect of low oral bioavailability of the control compound BVD-523, thereby hopefully overcoming the defect of individual differences in clinical medication and ultimately benefiting patients.
本发明已经通过上述实施例进行了说明,但应当理解的是,上述实施例只是用于举例和说明的目的,而非意在将本发明限制于所描述的实施例范围内。此外本领域技术人员可以理解的是,本发明并不局限于上述实施例,根据本发明的教导还可以做出更多种的变型和修改,这些变型和修改均落在本发明所要求保护的范围以内。本发明的保护范围由附属的权利要求书及其等效范围所界定。 The present invention has been described through the above-mentioned embodiments, but it should be understood that the above-mentioned embodiments are only for the purpose of illustration and illustration, and are not intended to limit the present invention to the scope of the described embodiments. In addition, those skilled in the art can understand that the present invention is not limited to the above embodiments, and more variations and modifications can be made according to the teachings of the present invention. These variations and modifications all fall within the scope of the protection claimed by the present invention. within range. The protection scope of the present invention is defined by the appended claims and their equivalent scope.

Claims (22)

  1. 一种结构式如式ⅠA所示的氘代化合物、或其药学上可接受的盐:
    A deuterated compound with a structural formula such as formula IA, or a pharmaceutically acceptable salt thereof:
    其中,R1和R2各自独立地选自CH3、CH2D、CHD2和CD3中的任一基团,R3-R14各自独立地是H或D,且R1-R14中至少有一个含D原子,Wherein, R 1 and R 2 are each independently selected from any group among CH 3 , CH 2 D, CHD 2 and CD 3 , R 3 to R 14 are each independently H or D, and R 1 to R 14 At least one of them contains D atom,
    条件是:requirement is:
    当式IA仅含1个D原子时,R8不是D,When formula IA contains only 1 D atom, R 8 is not D,
    当式IA含3个D原子时,R8-R10不同时是D,When formula IA contains 3 D atoms, R 8 -R 10 are not D at the same time,
    当式IA含4个D原子时,R6、R7、R11和R14不同时是D,When formula IA contains 4 D atoms, R 6 , R 7 , R 11 and R 14 are not D at the same time,
    当式IA含7个D原子、且R1和R2是CD3时,R7和R8不是D;和/或When formula IA contains 7 D atoms and R 1 and R 2 are CD 3 , R 7 and R 8 are not D; and/or
    当式IA含9个D原子、且R1和R2是CD3时,R6、R11和R14不同时是D。When formula IA contains 9 D atoms and R 1 and R 2 are CD 3 , R 6 , R 11 and R 14 are not D at the same time.
  2. 一种结构式如式I所示的氘代化合物、或其药学上可接受的盐:
    A deuterated compound with a structural formula as shown in formula I, or a pharmaceutically acceptable salt thereof:
    其中,R1和R2各自独立地选自CH3、CH2D、CHD2和CD3中的任一基团,R3-R14各自独立地是H或D,且R1-R14中至少有一个含D原子,条件是:Wherein, R 1 and R 2 are each independently selected from any group among CH 3 , CH 2 D, CHD 2 and CD 3 , R 3 to R 14 are each independently H or D, and R 1 to R 14 There is at least one D-containing atom in , provided that:
    当式I仅含1个D原子时,R8不是D,When formula I contains only 1 D atom, R 8 is not D,
    当式I含3个D原子时,R8-R10不同时是D,When formula I contains 3 D atoms, R 8 -R 10 are not D at the same time,
    当式I含4个D原子时,R6、R7、R11和R14不同时是D, When formula I contains 4 D atoms, R 6 , R 7 , R 11 and R 14 are not D at the same time,
    当式I含7个D原子、且R1和R2是CD3时,R7和R8不是D;和/或When formula I contains 7 D atoms and R 1 and R 2 are CD 3 , R 7 and R 8 are not D; and/or
    当式I含9个D原子、且R1和R2是CD3时,R6、R11和R14不同时是D。When formula I contains 9 D atoms and R 1 and R 2 are CD 3 , R 6 , R 11 and R 14 are not D at the same time.
  3. 如权利要求1或2所述的化合物,其特征在于,R1-R7、和R9-R14中至少有一个含D原子;优选地,R1-R5、和R9-R14中至少有一个含D原子;优选地,R1-R5、R9-R11、和R14中至少有一个含D原子。The compound according to claim 1 or 2, characterized in that at least one of R 1 -R 7 and R 9 -R 14 contains a D atom; preferably, R 1 -R 5 and R 9 -R 14 At least one of R 1 -R 5 , R 9 -R 11 , and R 14 contains a D atom; preferably, at least one of R 1 -R 5 , R 9 -R 11 , and R 14 contains a D atom.
  4. 如权利要求1-3任一项所述的化合物,其特征在于,The compound according to any one of claims 1-3, characterized in that,
    R1是CD3、CH2D、或CHD2;和/或R 1 is CD 3 , CH 2 D, or CHD 2 ; and/or
    R2是CD3、CH2D、或CHD2;和/或R 2 is CD 3 , CH 2 D, or CHD 2 ; and/or
    R3是D;和/或R 3 is D; and/or
    R4是D;和/或R 4 is D; and/or
    R5是D;和/或R 5 is D; and/or
    R6是D;和/或R 6 is D; and/or
    R7是D;和/或R 7 is D; and/or
    R8是D;和/或R 8 is D; and/or
    R9和R10是D;和/或R 9 and R 10 are D; and/or
    R11和R14是D;和/或R 11 and R 14 are D; and/or
    R12和R13是D。R 12 and R 13 are D.
  5. 如权利要求1-4任一项所述的化合物,其特征在于,The compound according to any one of claims 1 to 4, characterized in that,
    R1和R2是CD3;和/或R 1 and R 2 are CD 3 ; and/or
    R3是D;和/或R 3 is D; and/or
    R6是D;和/或R 6 is D; and/or
    R7是D;和/或R 7 is D; and/or
    R8是D;和/或R 8 is D; and/or
    R9和R10是D;和/或R 9 and R 10 are D; and/or
    R11和R14是D。R 11 and R 14 are D.
  6. 如权利要求1-5中任一项所述的化合物,其特征在于,The compound according to any one of claims 1-5, characterized in that,
    R1和R2是CD3;或R 1 and R 2 are CD 3 ; or
    R1和R2是CD3,R3是D;或R 1 and R 2 are CD 3 and R 3 is D; or
    R11和R14是D;或R 11 and R 14 are D; or
    R3、R9和R10是D;或 R 3 , R 9 and R 10 are D; or
    R3、R11和R14是D;或R 3 , R 11 and R 14 are D; or
    R1和R2是CD3,R9和R10是D;或R 1 and R 2 are CD 3 and R 9 and R 10 are D; or
    R1和R2是CD3、R11和R14是D;或R 1 and R 2 are CD 3 and R 11 and R 14 are D; or
    R1和R2是CD3,R3、R9和R10是D;或R 1 and R 2 are CD 3 and R 3 , R 9 and R 10 are D; or
    R1和R2是CD3,R3、R8、R9和R10是D;或R 1 and R 2 are CD 3 and R 3 , R 8 , R 9 and R 10 are D; or
    R1和R2是CD3、R3、R11和R14是D;或R 1 and R 2 are CD 3 , R 3 , R 11 and R 14 are D; or
    R3和R7是D;或R 3 and R 7 are D; or
    R3和R6是D;或R 3 and R 6 are D; or
    R1和R2是CD3,R3和R7是D;或R 1 and R 2 are CD 3 and R 3 and R 7 are D; or
    R6、R9和R10是D;或R 6 , R 9 and R 10 are D; or
    R6、R7、R9和R10是D;或R 6 , R 7 , R 9 and R 10 are D; or
    R3、R7、R9和R10是D;或R 3 , R 7 , R 9 and R 10 are D; or
    R1和R2是CD3,R7、R9和R10是D;或R 1 and R 2 are CD 3 and R 7 , R 9 and R 10 are D; or
    R1和R2是CD3,R3、R7、R9和R10是D;或R 1 and R 2 are CD 3 and R 3 , R 7 , R 9 and R 10 are D; or
    R3、R6、R9和R10是D。R 3 , R 6 , R 9 and R 10 are D.
  7. 如权利要求1-4任一所述的化合物,其特征在于,The compound according to any one of claims 1 to 4, characterized in that,
    R11和R14是D;或R 11 and R 14 are D; or
    R3、R11和R14是D;或R 3 , R 11 and R 14 are D; or
    R1和R2是CD3、R11和R14是D;或R 1 and R 2 are CD 3 and R 11 and R 14 are D; or
    R1和R2是CD3,R3、R9和R10是D;或R 1 and R 2 are CD 3 and R 3 , R 9 and R 10 are D; or
    R1和R2是CD3,R3、R8、R9和R10是D;或R 1 and R 2 are CD 3 and R 3 , R 8 , R 9 and R 10 are D; or
    R1和R2是CD3、R3、R11和R14是D;或R 1 and R 2 are CD 3 , R 3 , R 11 and R 14 are D; or
    R1和R2是CD3,R3和R7是D;或R 1 and R 2 are CD 3 and R 3 and R 7 are D; or
    R6、R7、R9和R10是D;或R 6 , R 7 , R 9 and R 10 are D; or
    R3、R7、R9和R10是D;或R 3 , R 7 , R 9 and R 10 are D; or
    R1和R2是CD3,R3、R7、R9和R10是D;或R 1 and R 2 are CD 3 and R 3 , R 7 , R 9 and R 10 are D; or
    R3、R6、R9和R10是D;或R 3 , R 6 , R 9 and R 10 are D; or
    R1和R2是CD3,R3、R4和R5是D;或R 1 and R 2 are CD 3 and R 3 , R 4 and R 5 are D; or
    R4、R5、R6和R7是D;或R 4 , R 5 , R 6 and R 7 are D; or
    R1和R2是CD3,R3、R8是D;或R 1 and R 2 are CD 3 and R 3 and R 8 are D; or
    R1和R2是CD3,R3、R6、R7是D;或 R 1 and R 2 are CD 3 and R 3 , R 6 and R 7 are D; or
    R1和R2是CD3,R3、R6、R7、R8是D;或R 1 and R 2 are CD 3 and R 3 , R 6 , R 7 and R 8 are D; or
    R1和R2是CD3,R3、R6、R7、R8、R9、R10是D;或R 1 and R 2 are CD 3 and R 3 , R 6 , R 7 , R 8 , R 9 and R 10 are D; or
    R1和R2是CD3,R3、R5、R6和R7是D。R 1 and R 2 are CD 3 and R 3 , R 5 , R 6 and R 7 are D.
  8. 如权利要求1-7任一项所述的化合物,其特征在于,R12和R13是H。The compound of any one of claims 1-7, wherein R 12 and R 13 are H.
  9. 如权利要求1所述的化合物,其特征在于,选自下述结构式所示的化合物中的任一种:









    The compound according to claim 1, characterized in that it is selected from any one of the compounds represented by the following structural formulas:









  10. 如权利要求1、2或9任一所述的化合物,其特征在于,选自下述结构式所示的化合物中的任一种:








    The compound of any one of claims 1, 2 or 9, characterized in that it is selected from any one of the compounds represented by the following structural formulas:








  11. 如权利要求1-10中任一项所述的化合物的制备方法,其特征在于,通过反应式Ⅰ制备得到的:
    The preparation method of the compound according to any one of claims 1 to 10, characterized in that it is prepared by reaction formula I:
    其中,R1-R2各自独立地选自CH3、CH2D、CHD2或CD3中的任一种, R3-R14各自独立地是H或D;X1选自Br、I、OTf和OMs中的任一种;和Y为Ts或Boc。Wherein, R 1 -R 2 are each independently selected from any one of CH 3 , CH 2 D, CHD 2 or CD 3 , R 3 to R 14 are each independently H or D; X 1 is selected from any one of Br, I, OTf and OMs; and Y is Ts or Boc.
  12. 如权利要求11中所述的化合物的制备方法,其特征在于,所述中间体化合物4是通过反应式Ⅱ或反应式Ⅲ制备得到的:
    The method for preparing a compound as claimed in claim 11, wherein the intermediate compound 4 is prepared by reaction formula II or reaction formula III:
    其中,R1-R2各自独立地选自CH3、CH2D、CHD2或CD3中的任一种,R3-R14各自独立地是H或D;X1为OTf或OMs,X2是F、Cl、Br或I中的任一种;Y为Ts或Boc。Wherein, R 1 to R 2 are each independently selected from any one of CH 3 , CH 2 D, CHD 2 or CD 3 , R 3 to R 14 are each independently H or D; X 1 is OTf or OMs, X 2 is any one of F, Cl, Br or I; Y is Ts or Boc.
  13. 如权利要求11所述的化合物的制备方法,其特征在于,所述中间体化合物4选自
    中的任一种。
    The method for preparing a compound according to claim 11, wherein the intermediate compound 4 is selected from the group consisting of
    any of them.
  14. 如权利要求11所述的化合物的制备方法,其特征在于,中间体化合物4与下述的中间体化合物5反应得到中间体6:
    The method for preparing a compound according to claim 11, wherein intermediate compound 4 reacts with the following intermediate compound 5 to obtain intermediate 6:
  15. 如权利要求11所述的化合物的制备方法,其特征在于,中间体化合物5的前体化合物选自
    中的任一种。
    The method for preparing a compound according to claim 11, wherein the precursor compound of intermediate compound 5 is selected from the group consisting of:
    any of them.
  16. 如权利要求11所述的化合物的制备方法,其特征在于,中间体化合物8选自
    中的任一种。
    The method for preparing a compound according to claim 11, wherein intermediate compound 8 is selected from the group consisting of
    any of them.
  17. 药物组合物,其特征在于,含有药学上可接受的载体和权利要求1-10中任一项所述的化合物。A pharmaceutical composition, characterized by containing a pharmaceutically acceptable carrier and the compound according to any one of claims 1-10.
  18. 如权利要求1-10中任一项所述的化合物或权利要求17所述的药物组合物,在制备用于预防和/或治疗增生性疾病或者由RAS-RAF-MEK1/2-ERK1/2激酶调节的疾病的药物中的应用。The compound according to any one of claims 1 to 10 or the pharmaceutical composition according to claim 17, when prepared for the prevention and/or treatment of proliferative diseases or composed of RAS-RAF-MEK1/2-ERK1/2 Applications in drugs for kinase-modulated diseases.
  19. 如权利要求18所述的应用,其特征在于,所述由RAS-RAF-MEK1/2-ERK1/2激酶调节的疾病为对ERK的抑制敏感的肿瘤;The application according to claim 18, wherein the disease regulated by RAS-RAF-MEK1/2-ERK1/2 kinase is a tumor that is sensitive to the inhibition of ERK;
    优选地,所述疾病为人恶性黑色素瘤或人结肠癌。Preferably, the disease is human malignant melanoma or human colon cancer.
  20. 权利要求1-10中任一项所述的化合物或权利要求17所述的药物组合物,其用作药物。A compound according to any one of claims 1 to 10 or a pharmaceutical composition according to claim 17 for use as a medicament.
  21. 权利要求1-10中任一项所述的化合物或权利要求17所述的药物组合物,其用于预防和/或治疗的疾病为对ERK的抑制敏感的肿瘤;The compound according to any one of claims 1 to 10 or the pharmaceutical composition according to claim 17, the disease used for prevention and/or treatment is a tumor that is sensitive to the inhibition of ERK;
    优选地,所述疾病为人恶性黑色素瘤或人结肠癌。 Preferably, the disease is human malignant melanoma or human colon cancer.
  22. 预防和/或治疗对ERK的抑制敏感的肿瘤疾病的方法,包括施用治疗有效量的权利要求1-10中任一项所述的化合物或权利要求17所述的药物组合物;A method for preventing and/or treating tumor diseases that are sensitive to the inhibition of ERK, comprising administering a therapeutically effective amount of the compound of any one of claims 1-10 or the pharmaceutical composition of claim 17;
    优选地,所述疾病为人恶性黑色素瘤或人结肠癌。 Preferably, the disease is human malignant melanoma or human colon cancer.
PCT/CN2023/080351 2022-03-08 2023-03-08 Deuterated compound, preparation method therefor and use thereof WO2023169480A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202210227792 2022-03-08
CN202210227792.0 2022-03-08
CN202211671643.X 2022-12-26
CN202211671643 2022-12-26

Publications (2)

Publication Number Publication Date
WO2023169480A1 true WO2023169480A1 (en) 2023-09-14
WO2023169480A9 WO2023169480A9 (en) 2024-01-11

Family

ID=87936124

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/080351 WO2023169480A1 (en) 2022-03-08 2023-03-08 Deuterated compound, preparation method therefor and use thereof

Country Status (1)

Country Link
WO (1) WO2023169480A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1976919A (en) * 2004-05-14 2007-06-06 沃泰克斯药物股份有限公司 Pyrrole inhibitors of ERK protein kinase, synthesis thereof and intermediates thereto
CN107406413A (en) * 2015-01-30 2017-11-28 生物医学谷探索股份有限公司 C21H22Cl2N4O2Crystal formation
CN110526916A (en) * 2018-05-23 2019-12-03 成都海创药业有限公司 Deuterated Capmatinib compound and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1976919A (en) * 2004-05-14 2007-06-06 沃泰克斯药物股份有限公司 Pyrrole inhibitors of ERK protein kinase, synthesis thereof and intermediates thereto
CN107406413A (en) * 2015-01-30 2017-11-28 生物医学谷探索股份有限公司 C21H22Cl2N4O2Crystal formation
CN110526916A (en) * 2018-05-23 2019-12-03 成都海创药业有限公司 Deuterated Capmatinib compound and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LIANG, TINGTING; WANG, WENJIE; HAO, SIYUAN; HE, GUANGCHAO; XU, YUNGEN: "Research Progress of ERK Small Molecule Inhibitors", JOURNAL OF CHINA PHARMACEUTICAL UNIVERSITY, NAJING, CN, vol. 51, no. 3, 25 June 2020 (2020-06-25), CN , pages 260 - 269, XP009548992, ISSN: 1000-5048, DOI: 10.11665/j.issn.1000-5048.20200302 *
ZHANG, YINSHENG: "Development of Deuterated Drugs: Past, Present and Future", PROGRESS IN PHARMACEUTICAL SCIENCES, vol. 41, no. 12, 25 December 2017 (2017-12-25), pages 902 - 918, XP009521674, ISSN: 1001-5094 *

Also Published As

Publication number Publication date
WO2023169480A9 (en) 2024-01-11

Similar Documents

Publication Publication Date Title
CN110088105B (en) Small molecule inhibitors of JAK family kinases
CN109563060B (en) IDO1 inhibitor and preparation method and application thereof
BR112018008877B1 (en) RET-INHIBITOR COMPOUNDS, PHARMACEUTICAL COMPOSITION AND USES OF SUCH COMPOUNDS
WO2020259683A1 (en) 2,4-disubstituted pyrimidine derivative, preparation method for same, and uses thereof
CN104136411A (en) Therapeutically active compounds and their methods of use
JP2023535746A (en) Compounds used as CDK7 kinase inhibitors and their applications
CN110092779B (en) Substituted phenyl compound and application thereof
CN108658983A (en) N- benzenesulfonyl benzamide compounds for inhibiting Bcl-2 albumen and combinations thereof and application
WO2022268066A1 (en) Protein degradation agent
JP2022549923A (en) Crystal forms of N-hetero pentacyclic ring-containing capsid protein assembly inhibitors and uses thereof
TW202102511A (en) A thienyl fused heterocyclic derivative, a preparation method thereof, and medical applications thereof
CN106554314A (en) Benzamide derivatives
CN109096272B (en) Indole hydroxamic acid compound with anti-tumor activity and application thereof
WO2023169480A1 (en) Deuterated compound, preparation method therefor and use thereof
CN112898286A (en) Benzothiophene compound or pharmaceutically acceptable salt and isomer thereof, and preparation method, pharmaceutical composition and application thereof
WO2017148290A1 (en) Substituted adenine compound and pharmaceutical composition thereof
WO2022083706A1 (en) Salts of fxia inhibitor compounds, preparation method therefor, and pharmaceutical use thereof
WO2020233618A1 (en) Inhibitors for programmed cell necrosis and preparation method therefor and use thereof
CN114685532A (en) Macrocyclic compound and medical application thereof
CN108623555B (en) Benzoxel compound, preparation method thereof, pharmaceutical composition and application thereof
US9505743B2 (en) Matrix metalloproteinase inhibitors and methods for the treatment of pain and other diseases
WO2019149090A1 (en) Crystal form of urate transporter 1 inhibitor and preparation method and use thereof
WO2020156571A1 (en) Pyridazine derivative, and preparation method and medicinal use thereof
CN111825608A (en) Tetrahydroquinoline and tetrahydroisoquinoline compounds and application thereof
CN104788372B (en) A kind of deuterated card is rich to replace Buddhist nun's derivative, its preparation method, application and its intermediate

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23766057

Country of ref document: EP

Kind code of ref document: A1