CN116270660A - Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof - Google Patents

Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof Download PDF

Info

Publication number
CN116270660A
CN116270660A CN202310330972.6A CN202310330972A CN116270660A CN 116270660 A CN116270660 A CN 116270660A CN 202310330972 A CN202310330972 A CN 202310330972A CN 116270660 A CN116270660 A CN 116270660A
Authority
CN
China
Prior art keywords
methyl
bcl
piperazin
tetrahydro
benzamide
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN202310330972.6A
Other languages
Chinese (zh)
Inventor
谭芬来
朱聪
徐雪松
尹凯
沈玥
武春
罗妹平
楼叶江
陈夷花
肖湘
欧鹏举
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangzhou Lupeng Pharmaceutical Co Ltd
Original Assignee
Guangzhou Lupeng Pharmaceutical Co Ltd
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 Guangzhou Lupeng Pharmaceutical Co Ltd filed Critical Guangzhou Lupeng Pharmaceutical Co Ltd
Priority to CN202310330972.6A priority Critical patent/CN116270660A/en
Publication of CN116270660A publication Critical patent/CN116270660A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/14Drugs for genital or sexual disorders; Contraceptives for lactation disorders, e.g. galactorrhoea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Inorganic Chemistry (AREA)
  • Pulmonology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a composition containing a Bcl-2/Bcl-xL inhibitor, and a preparation method and application thereof, and belongs to the field of medicines. The Bcl-2/Bcl-xL inhibitor has remarkable pharmaceutical activity for inhibiting and treating cancers, especially SCLC, and the effect of the Bcl-2/Bcl-xL inhibitor is superior to that of conventional therapeutic drugs or other structural forms. The invention also provides a composition formed by using the Bcl-2/Bcl-xL inhibitor or other forms of the Bcl-2/Bcl-xL inhibitor as an API combined DNA alkylating agent, the composition shows the synergistic effect of two active substances, can effectively inhibit and treat cancer tumors, especially SCLC tumors, and quickly lighten the body load caused by the diseases, and simultaneously has high targeting property, high curative effect stability and low biological toxicity in the application process, and the comprehensive effect is far superior to that of the existing products of the same type.

Description

Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof
Technical Field
The invention relates to the technical field of medicines, in particular to a composition containing a Bcl-2/Bcl-xl inhibitor, and a preparation method and application thereof.
Background
Cancer, which is a disease mechanism, disease symptoms and differences in therapeutic means due to differences in disease sites, wherein SCLC (small cell lung cancer) accounts for about 10 to 15% of all lung cancers, wherein about 30% of patients suffer from localized diseases, and 70% of patients exhibit widespread disease at diagnosis. Given the trend of disease spreading extensively at the time of first diagnosis and the limited treatment options available, for SCLC patients not receiving any treatment, the average median survival is only 2 to 4 months, with a typical 5-year survival rate of less than 6%. For patients with a wide range of diseases, the current standard of first-line SCLC initial treatment is chemotherapy or combined chest radiotherapy, as SCLC is sensitive to the initial course of treatment. However, most patients relapse, secondary or further treatment is frequent, but currently approved and effective adjuvant drugs are very limited.
In recent years, targeted therapies and immunotherapies for SCLC have been widely studied, and a large number of drugs have been demonstrated to be effective in the treatment of SCLC, with Bcl-2/Bcl-xl inhibitors as a dual-target efficacy drug having been demonstrated to have positive efficacy in SCLC clinical studies, which is achieved mainly by inhibiting the signaling pathway expression of the two anti-apoptotic proteins Bcl-2 and Bcl-xl: (1) The high expression of Bcl-2 protein can cause abnormal apoptosis and participate in the generation and development of tumors, and the Bcl-2 protein can play an important role in the regulation of apoptosis by forming dimers or self-dimerization with pro-apoptotic proteins such as Bax. In normal cells, the balance of apoptotic states can be maintained by various protein binding. In tumor cells, when Bcl-2 protein is over-expressed, heterodimers formed by the combination with Bax in the body are increased, so that apoptosis is inhibited or the threshold value of apoptosis is increased; when Bcl-2 protein is inhibited, its heterodimer with pro-apoptotic proteins is reduced and more Bax is released resulting in apoptosis of the cell. Thus, the balance between BCL-2 and Bax proteins between cell death signal checkpoints, especially tumor cells, determines cell survival or apoptosis. Meanwhile, research shows that Bcl-2 protein can also participate in the formation of tumor drug resistance, and the level of Bcl-2 protein in tumor cells is greatly increased after tumor patients receive chemotherapy or radiotherapy. (2) Bcl-xl proteins also play an important role in the processes of apoptosis and cell necrosis. Also anti-apoptotic proteins, bcl-xl and Bxl-2 proteins do not play exactly the same role in the different cell types and death signaling processes. In tumor cells, bcl-cl is also highly expressed, and it has been found that Bcl-cl protein is expressed in various solid tumors such as lung cancer (including SCLC and NSCLC), colorectal cancer, glioblastoma, breast cancer, esophageal cancer, liver cancer, pancreatic cancer, and melanoma, as compared with that of normal cells. Meanwhile, the high-expression Bcl-xl protein is related to the disregulation of the apoptosis mechanism of tumor cells and the generation of drug resistance to the chemotherapeutic drugs, and the inhibition of the over-expression of the Bcl-xl protein can induce the apoptosis of the tumor cells or increase the sensitivity of the Bcl-xl protein to the chemotherapeutic drugs.
However, the overall effect of the existing Bcl-2/Bcl-xl inhibitor still has a larger lifting space, and at the same time, under specific details, different Bcl-2/Bcl-xl inhibitors have larger differences in effect and side effect degree on the same cancer cell line and the same Bcl-2/Bcl-xl inhibitor on different cancer cell lines, and further analysis is needed depending on the use scene.
Disclosure of Invention
Based on the drawbacks of the prior art, the present invention aims to provide a Bcl-2/Bcl-xl inhibitor with better therapeutic effect on cancer, especially SCLC, than other Bcl-2/Bcl-xl inhibitors or efficacy drugs alone.
In order to achieve the above purpose, the invention adopts the following technical scheme:
Bcl-2/Bcl-xl inhibitors have the chemical structure shown below:
Figure BDA0004154928500000021
wherein,,
q4 and Q5 are at least one of cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl and spiroheterocyclyl;
R 1 、R 2 、R 7 、R 9 and R is 10 Is H, D, alkyl, spiroalkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, spiroheterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl, halogen, nitro, oxo, cyano, -OR a 、-SR a -alkyl-R a 、-NH(CH 2 ) p R a 、-C(O)R a 、-S(O)Ra、-SO 2 R a 、-C(O)OR a 、-OC(O)R a 、-NR b R c 、-C(O)N(R b )R c 、-N(R b )C(O)R c 、-P(O)R b R c -alkyl-P (O) R b R c 、-S(O)(=N(R b ))R c 、-N=S(O)R b R c 、=NRb、-SO 2 N(R b )Rc、-N(R b )SO 2 R c Wherein at least one of said cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl is substituted with one or more R d Substitution;
R a 、R b 、R c 、R d is at least one of H, D, alkyl, spiroalkyl, alkenyl, alkynyl, alkoxy, alkoxyalkyl, haloalkyl, hydroxyalkyl, aminoalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, spiroheterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl, wherein at least one of the alkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl is substituted with one or more R e Substitution;
R e is at least one of H, D, alkyl, spiroalkyl, alkenyl, alkynyl, halogen, cyano, amine, nitro, hydroxy, =o, -C (O) NHOH, alkoxy, alkoxyalkyl, haloalkyl, hydroxyalkyl, aminoalkyl, alkylcarbonyl, alkoxycarbonyl, alkylcarbonylamino, alkylamino, oxo, haloalkylamino, cycloalkyl, cycloalkenyl, heterocycloalkyl, spiroheterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl;
Z 1 is a bond, - (CH) 2 ) p -、-N(H)-、-O-、-S-、-C(O)-、-S(O 2 )-、-OC(O)-、-C(O)O-、-OSO 2 -、-S(O 2 )O-、-C(O)S-、-SC(O)-、-C(O)C(O)-、-C(O)N(H)-、-N(H)C(O)-、-S(O 2 )N(H)-、-N(H)S(O 2 )-、-OC(O)O-、-OC(O)S-、-OC(O)N(H)-、-N(H)C(O)O-、-N(H)C(O)S-、-N(H)C(O)N(H)-、-(CH 2 ) p N(H)(CH 2 ) q -、-(CH 2 ) p N(H)C(O)(CH 2 ) q -、-(CH 2 ) p C(O)N(H)(CH 2 ) q -、-OC(O)N(H)(CH 2 ) p+ 1 N(H)(CH 2 ) q -, a divalent alkenyl group or a divalent alkynyl group;
l is-L 1 -L 2 -;
Wherein L is 1 Is a bond, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl,Aryl or heteroaryl, at least one of said alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl being substituted with one or more R d Substitution;
L 2 is a bond or an alkyl group, at least one of which is bound to one or more-L i Optionally interposed between any two adjacent carbon atoms;
the said-L i -is-N (Ra) -, -O-, -S-, -C (O) -, -S (O) 2 )-、-OC(O)-、-C(O)O-、-OSO 2 -、-S(O 2 )O-、-C(O)S-、-SC(O)-、-C(O)C(O)-、-C(O)N(Ra)-、-N(Ra)C(O)-、-S(O 2 )N(R a )-、-N(R a )S(O 2 )-、-OC(O)O-、-OC(O)S-、-OC(O)N(R a )-、-N(R a )C(O)O-、-N(R a )C(O)S-、-N(R a )C(O)N(R a ) At least one of a divalent alkenyl group, a divalent alkynyl group, a divalent cycloalkyl group, a divalent heterocycloalkyl group, a divalent aryl group, a divalent heteroaryl group;
the k, n, p, q =0 to 5,s =0 or 1, and f=0 or 1.
Preferably, said R 2 Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R d And (3) substitution.
Preferably, said R 10 Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R d And (3) substitution.
Preferably, said R 7 And L, together with the atoms to which they are attached, form cycloalkyl or heterocycloalkyl, said cycloalkyl or heterocycloalkyl being substituted with one or more R e And (3) substitution.
Preferably, said R b And R is c Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R e And (3) substitution.
Preferably, said R d Two of which, together with the atoms to which they are attached, are formedCycloalkyl or heterocycloalkyl, said cycloalkyl or heterocycloalkyl being substituted by one or more R e And (3) substitution.
Preferably, said R e Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R f Substitution, said R f Is at least one of H, D, alkyl, alkenyl, alkynyl, halogen, cyano, amine, nitro, hydroxy, -C (O) NHOH, alkoxy, alkoxyalkyl, haloalkyl, hydroxyalkyl, aminoalkyl, alkylcarbonyl, alkoxycarbonyl, alkylcarbonylamino, alkylamino, oxo, haloalkylamino, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl.
Preferably, the chemical structure of the Bcl-2/Bcl-xl inhibitor is as follows:
Figure BDA0004154928500000031
more preferably, the chemical structure of the Bcl-2/Bcl-xl inhibitor is as follows:
Figure BDA0004154928500000032
preferably, the Bcl-2/Bcl-xl inhibitor is as follows:
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 3-difluoro-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
Or (b)
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((4- (((4-fluorotetrahydro-2H-pyran-4-yl) methyl) amino) -3-nitrophenyl) sulfonyl) benzamide;
or (b)
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(R) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) benzamide;
Or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((4- (((4-fluorotetrahydro-2H-pyran-4-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -2- (4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- ((4- ((((R) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((S) -4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- ((4- ((((R) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((S) -4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- ((4- ((((R) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((R) -4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
Or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- ((4- ((((S) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((R) -4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
Or (b)
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-4, 4-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((5-nitro-3- (tetrahydro-2H-pyran-4-yl) -3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) benzamide;
or (b)
(R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((5-nitro-3- (tetrahydro-2H-pyran-4-yl) -3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) benzamide;
Or (b)
N- (((R) -3- ((S) -1, 4-dioxane-2-yl) -5-nitro-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- (((S) -3- ((S) -1, 4-dioxan-2-yl) -5-nitro-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide.
More preferably, the Bcl-2/Bcl-xl inhibitor is:
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 3-difluoro-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide.
It should be noted that the Bcl-2/Bcl-xl inhibitors of the present invention are not limited to the specific chemical structures described above, and other specific structures of Bcl-2/Bcl-xl inhibitors that conform to the scope of the chemical structures described herein are within the scope of the present invention.
It is another object of the present invention to provide the use of said Bcl-2/Bcl-xl inhibitors for the preparation of a medicament for the treatment of cancer.
Compared with the existing conventional effective cancer medicines or Bcl-2/Bcl-xl inhibitors with other structures, the Bcl-2/Bcl-xl inhibitor provided by the invention has better curative effect activity, and is suitable for preparing various dosage forms of cancer treatment medicines.
Preferably, the administration of the cancer treatment drug is sequential.
According to the actual needs, the medicines for treating cancers prepared by the Bcl-2/Bcl-xl inhibitor can be used for treating cancers by sequential administration, but are not limited to the medicines.
Preferably, the components of the medicament for treating cancer further comprise excipients and/or pharmaceutically acceptable carriers and/or pharmaceutically acceptable vehicles.
Preferably, the dosage form of the cancer treatment drug is any one of a liquid dosage form, a gas dosage form, a solid dosage form and a semisolid dosage form.
Preferably, the cancer comprises small cell lung cancer, breast cancer, colorectal cancer;
more preferably, the cancer is small cell lung cancer SCLC;
more preferably, the SCLC is Bcl-2 highly expressed SCLC.
It is a further object of the present invention to provide a composition comprising the Bcl-2/Bcl-xl inhibitor of the invention or a nitrogen oxide thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a compound thereof or a prodrug form of a nitrogen oxide, and a DNA alkylating agent.
DNA alkylating agents are a commonly used clinical chemotherapeutic agent that, by covalent binding to the DNA single or double strand of a patient, disrupt its DNA structure and function, resulting in cell death in the patient. Alkylating agents can act on proliferating and resting cells (alkylating agents are a cell cycle non-specific anti-cancer drug), but proliferating cells are more sensitive to drugs, especially cells in G1 and S phases. The prior researches show that the inhibition and treatment effect degree of the DNA alkylating agent is not high when the DNA alkylating agent acts on clinical cancers, especially SCLC diseases, and in the invention, the pharmaceutical composition formed by combining the Active Pharmaceutical Ingredients (API) in the form of the Bcl-2/Bcl-xl inhibitor or the nitrogen oxides thereof and the like and the DNA alkylating agent has obvious curative effect on cancers, especially SCLC, and the two have synergistic effect.
Preferably, the components of the composition further comprise excipients and/or pharmaceutically acceptable carriers and/or pharmaceutically acceptable vehicles.
More preferably, in the composition, the Bcl-2/Bcl-xl inhibitor or a nitroxide thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a compound thereof or a prodrug form of a nitroxide is present in a solid dispersant and is a compound determined for at least one of WO/2017/132474, WO/2019/040550, WO/2019/040573, PCT/US2019/047404 and PCT/US 2019/047403.
Preferably, the DNA alkylating agent comprises a platinum-containing alkylating agent;
more preferably, the platinum-containing alkylating agent is at least one of platinum, cisplatin, carboplatin, dicycloplatin, etaplatin, lobaplatin, milliplatin, nedaplatin, oxaliplatin, pi Pubo, satraplatin and/or triplatin tetranitrate.
Preferably, the DNA alkylating agent is alkyl (C n H 2n+1 ) To DNA;
more preferably, the alkyl group is attached to a guanine base of the DNA. This site is preferably the N-7 position of the purine nucleus.
The DNA alkylating agent of the present invention may be active in a cellular environment, and inhibits tumor growth by crosslinking the avidines within the DNA duplex, e.g., preventing DNA strand expansion or/and separation, thereby inhibiting DNA replication; or the active substance is required to be converted into a certain in-vivo active substance before the in-vivo active substance is provided.
Meanwhile, the DNA alkylating agent of the present invention may contain a typical DNA alkylating agent such as nitrosourea, e.g., carmustine, lomustine, or streptozotocin, and may also contain an atypical alkylating agent such as procarbazine, a Qu Tamin, triazine (dacarbazine, imidazole amine, temozolomide), dacarbazine, or temozolomide.
It is a further object of the present invention to provide the use of said composition for the preparation of a medicament for the treatment of cancer
Because of the synergistic effect of the API of the Bcl-2/Bcl-xl inhibitor and the DNA alkylating agent, the medicine further prepared by the composition provided by the invention has better curative effect activity compared with the traditional conventional cancer effective medicine or the medicine which singly adopts the Bcl-2/Bcl-xl inhibitor or the DNA alkylating agent as the API. Meanwhile, biological in-vivo experiments prove that the composition has high targeting property and low drug toxicity in the application process, and can not generate excessive body load on a treatment object.
Preferably, the administration of the cancer treatment drug is sequential.
According to the actual needs, the cancer treatment medicine prepared by the composition of the invention can be used for treating cancer by using but not limited to sequential administration.
Preferably, the components of the medicament for treating cancer further comprise excipients and/or pharmaceutically acceptable carriers and/or pharmaceutically acceptable vehicles.
Preferably, the dosage form of the cancer treatment drug is any one of a liquid dosage form, a gas dosage form, a solid dosage form and a semisolid dosage form.
Preferably, the cancer comprises small cell lung cancer, breast cancer, colorectal cancer;
More preferably, the cancer is small cell lung cancer SCLC;
more preferably, the SCLC is Bcl-2 highly expressed SCLC.
Preferably, the cancer treatment drug, when used for preventing and/or treating cancer, is administered in an amount of 0.0025 to 1,500 mg/day in the form of a Bcl 2/Bcl xL inhibitor of the cancer treatment drug, a nitroxide thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a compound thereof, or a prodrug of the nitroxide.
Preferably, the agent for treating cancer is administered in an amount of 0.0025 to 1,000 mg/day, as the DNA alkylating agent or a pharmaceutically acceptable salt or solvate thereof in the agent for treating cancer, when used for preventing and/or treating cancer.
Preferably, the cancer treatment drug, when used in the prevention and/or treatment of cancer, the Bcl 2/Bcl xL inhibitor and the DNA alkylating agent in the cancer treatment drug may be administered sequentially, with time intervals of sequential administration including, but not limited to, 1 minute, 5 minutes, 10 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, and 12 weeks.
Preferably, the cancer therapeutic agent, when used in the prevention and/or treatment of cancer, the Bcl 2/Bcl xL inhibitor and DNA alkylating agent in the cancer therapeutic agent may be administered in combination, either in individual dosage unit form or in combination dosage unit form, for a number of administrations including, but not limited to, 1, 2, 3, 4, 5 and 6.
Preferably, the cancer treatment drug, when used in the prevention and/or treatment of cancer, is administered by a method including, but not limited to: oral, buccal, inhalation spray, sublingual, rectal, transdermal, vaginal mucosal, transmucosal, topical, nasal or enteral administration; injection administration, including but not limited to intramuscular, subcutaneous, intramedullary, and intrathecal, direct brain administration, in situ, subcutaneous, intraperitoneal, intravenous, intra-articular, intrasternal, intrahepatic, intralesional, intracranial, intraperitoneal, nasal, and intraocular injection.
Preferably, the cancer treatment drug is administered in an amount of 0.005 to 500 mg/day, preferably 0.05 to 365 mg/day, preferably 0.05 to 317 mg/day, preferably 0.05 to 250 mg/day, more preferably 0.5 to 100 mg/day of the Bcl 2/Bcl xL inhibitor, the nitroxide thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a compound thereof, or a prodrug form of nitroxide in the cancer treatment drug when used for the prevention and/or treatment of cancer; or in an amount of 10 to 1000 mg/week, 10 to 900 mg/week, 10 to 800 mg/week, 10 to 700 mg/week, 10 to 640 mg/week, 10 to 600 mg/week, 10 to 500 mg/week, 10 to 400 mg/week, 10 to 300 mg/week, 10 to 200 mg/week, 20 to 100 mg/week; more preferably, the administration is carried out in an amount of 0.005mg, 0.05mg, 0.5mg, 5mg, 10mg, 20mg, 30mg, 40mg, 50mg, 100mg, 150mg, 200mg, 250mg, 300mg, 350mg, 400mg, 450mg or 500mg per dose.
Preferably, the cancer treatment drug, when used in the prevention and/or treatment of cancer, is administered at a frequency of 1 time per week, 2 times per week, 3 times per week, 4 times per week, 5 times per week, 6 times per week, 7 times per week of the Bcl 2/Bcl xL inhibitor of the cancer treatment drug, its nitroxide, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a compound thereof, or a prodrug form of nitroxide.
Preferably, when the cancer treatment drug is used for preventing and/or treating cancer, the DNA alkylating agent in the cancer treatment drug is administered in an amount ranging from 10mg, 20mg, 30mg, 40mg, 50mg, 61mg, 70mg, 73mg, 80mg, 90mg, 97.6mg, 100mg, 122mg, 150mg, 200mg, 250mg, 300mg, 350mg, 400mg, 450mg, 460mg, 470mg, 480mg, 487mg, 490mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, 800mg, 850mg, 900mg, 950mg or 1000mg, and between the amounts including, but not limited to, 10mg to 1000mg, 20mg to 950mg, 30mg to 900mg, 50mg to 650mg, 61mg to 600mg, 400mg, 73mg to 550mg, 73mg to 522mg, 97.6mg to 600mg, 97.6 to 700mg, 97.700 mg, 700mg to 500mg, 122 to 1000mg, 122mg and 122 to 1000 mg.
The Bcl 2/Bcl xL inhibitor has the beneficial effects that the Bcl 2/Bcl xL inhibitor has remarkable pharmaceutical activity for inhibiting and treating cancers, especially SCLC, and the effect is superior to that of the conventional therapeutic drugs or other structural forms. The invention also provides a composition formed by using the Bcl 2/Bcl xL inhibitor or other forms of the Bcl 2/Bcl xL inhibitor as an API combined DNA alkylating agent, the composition shows the synergistic effect of two active substances, can effectively inhibit and treat cancer tumors, especially SCLC tumors, quickly lighten the body load caused by the diseases, and has high targeting property, high curative effect stability and low biotoxicity in the application process, and the comprehensive effect is far better than that of the existing products of the same type.
Drawings
FIG. 1 is a graph showing the synergistic activity of a combination of Compound 1 (0 to 100 nM) and cisplatin (0 to 10000 nM) in NCI-H69 cell line test in accordance with effect example 2 of the present invention.
FIG. 2 is a graph showing the synergistic activity of a combination of Compound 1 (0 to 100 nM) and cisplatin (0 to 10000 nM) in the NCI-H889 cell line test in accordance with effect example 2 of the present invention.
FIG. 3 is a graph showing the synergistic activity of a combination of Compound 1 (0 to 100 nM) and cisplatin (0 to 10000 nM) in NCI-H146 cell line test in accordance with effect example 2 of the present invention.
Detailed Description
The present invention will be further described with reference to specific examples and comparative examples for better illustrating the objects, technical solutions and advantages of the present invention, and the object of the present invention is to be understood in detail, not to limit the present invention. All other embodiments, which can be made by those skilled in the art without the inventive effort, are intended to be within the scope of the present invention. The experimental reagents, raw materials and instruments designed in the practice and comparative examples of the present invention are common reagents, raw materials and instruments unless otherwise specified. In various embodiments of the present invention, when NMR data is provided, a 1H spectrum can be obtained at XL400 (400 MHz) and reported in ppm of Me4Si to low magnetic fields, and proton numbers, multiples and coupling constants are expressed in Hertz. In providing HPLC data, analysis can be performed using an Agilent 1100 or the like system. In providing LC/MS data, analysis can be performed using a Applied Biosystems API-100 mass spectrometer, a Shimadzu SCL-10A LC column, and the like.
In the specific examples of the present invention, examples 1 to 27 specifically demonstrate the Bcl-2/Bcl-xl inhibitors of the chemical structure of the present invention and the synthetic procedures thereof as follows:
Figure BDA0004154928500000081
The person skilled in the art can synthesize these products by the described methods or by the present invention without mention of other ways.
Example 1
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3-nitro- [ [ (oxa-4-yl) methyl ] amino ] benzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7]]]Synthesis of tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl) benzoate (999.0 mg,1.88mmol,4.00 eq.), toluene (20 mL), 4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7]]]Tridec-1 (9), 2,5, 7-tetraene (150 mg,0.47mmol,1 eq.) Cs 2 CO 3 (764.9 mg,2.35mmol,5 eq.) and XantPhos Pd 2G (333.2 mg,0.38mmol,0.8 eq.) were placed in a round-bottomed flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred at 110℃for 1 night. The resulting solution was diluted with 300mL of water, the resulting solution was extracted with 2X 100mL of ethyl acetate, and the resulting mixture was washed with 1X 300mL of brine. The resulting mixture was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo, and the residue was treated with ethyl acetate/petroleum ether (1:2) on a silica gel column to give 200mg (55.29%) of 4- (4- [ [2- (4-chlorophenyl))Phenyl) -4, 4-dimethylcyclohex-1-en-1-yl]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate as a yellow solid. LC-MS (ES, M/z): M+H=769, R, T= 3.076min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50 x 3.0shim-pack XR-ODS,2.2 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 100% acetonitrile, 5.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.4.0.0 ] [3,7 ]]]Synthesis of methyl tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: in a vial, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) is placed]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraene-1 (200 mg,0.26mmol,1 eq.) THF (20 mL), TBAF.3H 2 O (2.5 g), ethane-1, 2-diamine (1.5 g,24.96mmol,96.15 eq.) the resulting solution was stirred overnight in an oil bath at 70℃and then diluted with 200mL of water and the resulting solution extracted with 3X 30mL of ethyl acetate. The resulting mixture was washed with 2×200mL brine, dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (2:1) on a silica gel column to give 130mg (78.22%) of methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoate as a pale yellow solid. LC-MS (ES, M/z): M+=639, R, T=1.388 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50 x 3.0shim-pack XR-ODS,2.2 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 100% acetonitrile, 2.6min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoic acid: in one vial, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) was placed]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate (130 mg,0.20mmol,1 eq.) MeOH (6 mL), THF (6 mL), H 2 O (2 mL), naOH (81.2 mg,2.03mmol,10.00 eq.). The resulting solution was stirred in an oil bath at 60 ℃ for 1 night. The resulting mixture was concentrated under vacuum. The pH of the solution was adjusted to 5-6 with HCl (2 mol/L). The resulting solution was extracted with 2X 50mL of dichloromethane/MeOH (v: v=10:1). The resulting mixture was washed with 2X 200mL of brine, then dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo to yield 80mg (62.92%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid as a pale yellow solid. LC-MS (ES, M/z): M+=625, R, T=1.336 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50 x 3.0shim-pack XR-ODS,2.2 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 100% acetonitrile, 2.6min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ (3-nitro- [ [ (oxa-4-yl) methyl)]Amino group]Benzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: in a round bottom flask, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) was placed]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (50 mg,0.08mmol,1 eq.) DCM (3 mL), 3-nitro-4- [ [ (oxan-4-yl) methyl]Amino group]Benzene-1-sulfonamide (25.2 mg,0.08mmol,1.00 eq.) EDCI (30.6 mg,0.16mmol,2 eq.) DMAP (39.0 mg,0.32mmol,4 eq.). The resulting solution was stirred overnight at 25 ℃, and the resulting mixture was concentrated under vacuum. The residue was purified by column chromatography on silica gel with dichloroethane/methanolThe alcohol (10:1) was treated. Crude-purified by Prep-HPLC under the following conditions (IntelFlash-1): a column, a C18 reverse phase column; mobile phase, water (10 mmol/L NH) 4 HCO 3 +0.05%NH 3 .H 2 O) and CH 3 CN(20.0%CH 3 CN up to 90.0% in 30 min); detector, UV 220nm. 19.1mg (25.90%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -N- ([ 3-nitro-4- [ (oxalan-4-yl) methyl) amino group]Benzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide is a yellow solid. LC-MS (ES, M/z): M+1=923, R, T= 3.463min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Agilent Poroshell HPH-C18,2.7 μm; eluent a: water (0.05% ammonia); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min. 1 H NMR(300MHz,DMSO-d 6 Ppm) δ11.91 (s, 1H), 11.26 (s, 1H), 8.56 (s, 1H), 8.47 (d, j=2.1 hz, 1H), 7.61 (d, j=9.0 hz, 1H), 7.48 (d, j=9.2 hz, 1H), 7.37 (d, j=8.3 hz, 2H), 7.20 (s, 1H), 7.07 (d, j=8.3 hz, 2H), 6.99-6.83 (m, 2H), 6.76 (d, j=29.2 hz, 2H), 6.14 (s, 1H), 4.21 (s, 2H), 3.85 (d, j=9.3 hz, 2H), 3.52 (s, 2H), 3.30-3.14 (m, 8H), 2.79 (s, 1H), 2.23 (d, j=20.5 hz), 1.99 (s, 1.85 (s, 1H), 1.7.7 (s, 2H), 6.9 (s, 2H), 1.85 (s, 1H), 1.3.3 (s, 2H). NMR spectroscopic measurements were performed with Bruker AvanceIII HD 300MHz and BBOF probes.
Example 2
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (4-fluorooxalan-4-yl) methyl ] amino-3-nitrobenzene) benzenesulfonyl 2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tetracyclic-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
in a small bottle, 4- (4- [ [2- ]4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7]]]Tetracyclic-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (80 mg,0.13mmol,1 eq.) DCM (3 mL), 4- [ [ (4-fluorooxalan-4-yl) methyl]Amino group]3-Nitrophenyl-1-sulfonamide (42.6 MG,0.13MMOL,1.00 eq.) EDCI (49.0 mg,0.26mmol,2 eq.) DMAP (62.4 mg,0.51mmol,4 eq.). The resulting solution was stirred at 25℃for 1 night. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC under the following conditions (Prep-HPLC-006): column, X Bridge Prep C18 OBD column, 19X 150mm 5 μm; mobile phase, water (10 mmol/L NH) 4 HCO 3 +0.1%NH 3 ·H 2 O) and CH 3 CN(41.0%CH 3 CN increased to 61.0% in 6min, maintained at 95.0% in 1min, decreased to 41.0% in 1min, and maintained at 41.0% in 1 min); the detector, UV 210nm. 17mg (14.13%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -N- (4- [ [ (4-fluorooxalan-4-yl) methyl]Amino-3-nitrobenzene) benzenesulfonyl 2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tetracyclic-1 (9), 2,5, 7-tetraen-10-yl]Benzamide is a yellow solid. LC-MS (ES, M/z): M+1=940, R, T=1.583 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 100% acetonitrile, 3.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min. 1 H NMR(300MHz,DMSO-d 6 Ppm) δ11.94 (s, 1H), 11.25 (s, 1H), 8.59 (s, 1H), 8.48 (d, j=2.3 hz, 1H), 7.66 (d, j=9.1 hz, 1H), 7.47 (d, j=8.9 hz, 1H), 7.37 (d, j=8.2 hz, 2H), 7.20 (d, j=3.1 hz, 1H), 7.07 (dd, j=8.9, 3.8hz, 3H), 6.94 (s, 1H), 6.71 (s, 2H), 6.13 (d, j=3.1 hz, 1H), 4.20 (d, j=6.5 hz, 2H), 3.80-3.70 (m, 3H), 3.68-3.60 (m, 1H), 3.58-3.45 (m, 4H), 3.25-3.05 (m, 4.83-2H), 6.94 (s, 1H), 6.71 (s, 2H), 6.13 (d, j=3.1H), 4.68-3.60 (m, 1H), 3.58-3.60 (m, 1H), 2.35 (2H). NMR spectroscopic measurements were performed with Bruker AvanceIII HD 300MHz and BBOF probes.
Example 3
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of 4, 4-trifluoro-3-hydroxybutyramide: in a round bottom flask, 4-trifluoro-3-hydroxybutyramide (500 mg,2.7mmol,1 eq.) and NH were placed 3 MeOH (5 mL, 4.0M). The resulting solution was stirred at 60℃for 16h and the resulting mixture was concentrated. 500mg of 4, 4-trifluoro-3-hydroxybutyramide was obtained as a white solid. 1 H NMR(300MHz,DMSO-d 6 Ppm) delta 7.62 (ds, 1H), 7.01 (ds, 1H), 3.36-6.34 (m, 1H), 4.27-4.40 (m, 1H), 2.39-2.36 (m, 2H). NMR spectroscopic measurements were performed with Bruker AvanceIII HD 300MHz and BBOF probes.
Synthesis of 4-amino-1, 1-trifluorobutan-2-ol: in a round bottom flask, 4-trifluoro-3-hydroxybutyramide (500 mg,3.2mmol,1 eq.) and THF (10 mL) were placed and LAH (242 mg,6.4mmol,2 eq.) was added dropwise under ice-bath. The resulting solution was stirred at room temperature for 16h. After the reaction was completed, the reaction mixture was cooled by adding 0.24mL of water, and 0.24mL of LNaOH (10% H) was continuously added to the solution in an ice bath 2 O) and 0.72mL. The solid was filtered. The resulting mixture was concentrated under vacuum. 380mg (83.43%) of 4-amino-1, 1-trifluorobutan-2-ol were obtained as a colorless oil. 1 H NMR (300 MHz, CDCl3, ppm) 4.10-4.01 (m, 1H), 2.70-2.66 (m, 2H), 1.51-1.47 (m, 2H). NMR spectroscopic measurements were performed with Bruker AvanceIII HD 300MHz and BBOF probes.
Synthesis of 4-methyl-N- (4, 4-trifluoro-3-hydroxybutyl) benzenesulfonamide: in a 100mL round bottom flask, 4-amino-1, 1-trifluorobutan-2-ol (350 mg,2.4mmol,1 eq.) and triethanolamine (480 mg,4.8mmol,2 eq.) with DCM (10 mL) were placed in an ice bath with TsCl (470 mg,2.4mmol,1.0 eq.). The resulting solution was stirred at room temperature for 4h with 50mLAnd (5) diluting with DCM. The resulting mixture was washed with 2X 20mL of water and 1X 20mL of brine. The mixture was dried over anhydrous sodium sulfate. The solid was filtered. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column to finally give 600mg (82.52%) of 4-methyl-N- (4, 4-trifluoro-3-hydroxybutyl) benzene-1-sulfonamide as a yellow solid. 1 H NMR(300MHz,DMSO-d 6 Ppm) delta 7.70-7.68 (m, 2H), 7.42-7.40 (m, 2H), 6.23-6.21 (m, 1H), 4.01-3.96 (m, 1H), 2.87-2.83 (m, 2H), 2.39 (s, 3H), 1.70-1.49 (m, 2H). NMR spectroscopic measurements were performed with Bruker AvanceIII HD 300MHz and BBOF probes.
N- (3- (5-bromo-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrolo [2, 3-b)]Synthesis of pyridin-6-oxy) -4, 4-trifluorobutyl) -4-methylbenzenesulfonamide: in a round bottom flask, 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] was placed]Methyl group]-1H-pyrrole [2,3-b ]]Pyridine (300 mg,0.9mmol,1 eq.) 4-methyl-N- (4, 4-trifluoro-3-hydroxybutyl) benzene-1-sulfonamide (310 mg,1.0mmol,1.2 eq.) Cs 2 CO 3 (566 mg,1.7mmol,2.0 eq.) and 1, 4-dioxane (10 mL). The resulting solution was stirred in an oil bath at 90℃for 16h. The reaction mixture was cooled. The solid was filtered. The resulting solution was diluted with 100mL DCM. The resulting mixture was washed with 5X 50mL of water and 1X 50mL of brine. The mixture was dried over anhydrous sodium sulfate. The solid was filtered. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (0-30%) on a silica gel column. 400mg (73.95%) of N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) are obtained]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl) oxy]-4, 4-trifluorobutyl]-4-methylbenzene-1-sulfonamide as a pale yellow solid. 1 H NMR (300 MHz, CD Cl3, ppm) 8.17 (bs, 1H), 7.76-7.73 (m, 2H), 7.29-7.27 (m, 3H), 6.51-6.50 (m, 1H), 5.93-5.91 (m, 1H), 5.75-5.72 (m, 2H), 5.63-5.58 (m, 1H), 3.60-3.57 (m, 2H), 3.34-3.32 (m, 1H), 3.13-3.11 (m, 1H), 2.46 (s, 3H), 2.31-2.29 (m, 1H), 2.10-2.07 (m, 1H), 1.00-0.85 (m, 2H), 0.01 (s, 9H). NMR spectroscopic measurements were performed with Bruker AvanceIII HD 300MHz and BBOF probes.
1-tosyl-4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5,6]Pyridine compoundAnd [2,3-b ]][1,4]Synthesis of oxazepan: n- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl]Oxy group]-4, 4-trifluorobutyl]-4-methylbenzene-1-sulfonamide (700 mg,1.13mmol,1 eq.) Cs 2 CO 3 (1.1 g,3.39mmol,3.00 eq.) CuI (214 mg,1.13mmol,1.0 eq.) and 2-isobutyrylcyclohexanol-1-one (80 mg,0.56mmol,0.5 eq.) in DMSO (10 mL) were placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred in an oil bath at 120℃for 24h. The resulting solution was diluted with 20mL of water. The resulting solution was extracted with 2X 50mL of ethyl acetate. The resulting mixture was washed with 1X 50mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0-30%) on a silica gel column to give 350mg (57.38%) of 1-tosyl-4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5,6 ]]Pyridine [2,3-b][1,4]Oxazepan as a pale yellow solid. 1 H NMR (300 MHz, CD Cl3, ppm) 8.19 (bs, 1H), 7.50-7.47 (m, 2H), 7.39 (s, 1H), 7.24-7.22 (m, 2H), 6.58-6.57 (m, 1H), 5.69-5.66 (m, 1H), 5.55-5.51 (m, 1H), 4.57-4.52 (m, 1H), 3.96-3.94 (m, 1H), 3.59-3.56 (m, 2H), 3.48-3.44 (m, 1H), 2.41 (s, 3H), 2.31-2.29 (m, 1H), 1.95-1.91 (m, 1H), 0.97-0.91 (m, 2H), 0.05 (s, 9H). NMR spectroscopic measurements were performed with Bruker AvanceIII HD 300MHz and BBOF probes.
4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2':5,6]Pyridine [2,3-b][1,4]Synthesis of oxazepan: in a round bottom flask, in N 2 Na (150 mg,6.5mmol,1.0 eq.), naphthalene (833 mg,6.5mmol,10 eq.) and DME (3 mL) were added. The reaction mixture was stirred at room temperature until Na and naphthalene were completely dissolved. 1-tosyl-4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5,6 ] is added thereto at-78deg.C]Pyridine [2,3-b][1,4]A solution of oxazepan (350 mg,0.65mmol,1 eq.) in THF (5 mL). The resulting solution was stirred at-60 to-40 ℃ for 2-3 h until TLC had consumed the starting material. Then 5mL NH was added at-10deg.C 4 Cl toThe reaction was stopped. The resulting solution was extracted with 3X 10mL ethyl acetate. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was eluted with ethyl acetate/petroleum ether (1/3) on a silica gel column. 220mg (88%) of 4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrole are obtained [3',2':5,6 ]Pyridine [2,3-b][1,4]Oxazepan as a white solid.
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -3, 4-dihydropyrrol [3',2',5,6]Pyrido [2,3-b][1,4]Synthesis of methyl oxazepan-1 (7H) -yl) benzoate: 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) benzoate (1.2 g,2.28mmol,4 eq), toluene (20 mL), 4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5, 6)]Pyridine [2,3-b][1,4]Oxazepan (220 mg,0.57mmol,1 eq.) Cs 2 CO 3 (923 mg,2.84mmol,5 eq.) XantPhos Pd 2G (250 mg,0.46mmol,0.8 eq.) was placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred at 110℃overnight. The resulting solution was diluted with 30mL of water. The resulting solution was extracted with 2X 30mL ethyl acetate. The resulting mixture was washed with 1X 30mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column to give 380mg of crude (80.0%) 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) ]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid methyl ester as a yellow solid. LC-MS (ES, M/z): M+=838, R, T=3.33 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0,Poroshell HPH-C18,2.7 μm; eluent a: water (0.05% NH) 4 HCO 3 ) The method comprises the steps of carrying out a first treatment on the surface of the Eluent B: acetonitrile; linear gradient, from 5% acetonitrile to 100% acetonitrile,5.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min.
Methyl 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyridine [2,3-b][1,4]Synthesis of oxazepan-1 (7H) -yl) benzoate: in a vial, 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) is placed]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoate (380 mg,0.45mmol,1 eq.) THF (20 mL), TBAF.3H 2 O (706 mg,2.25mmol,5 eq.) ethane-1, 2-diamine (540 mg,9.0mmol,20 eq.). The resulting solution was stirred overnight in an oil bath at 70 ℃. The resulting solution was diluted with 20mL of water. The resulting solution was extracted with 3X 30mL of ethyl acetate. The resulting mixture was washed with 2X 20mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column. 300mg (93%) of 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) are obtained]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyridine [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid methyl ester as a pale yellow solid. 1 H NMR(300MHz,CDCL3,ppm)10.78(bs,1H),7.75-7.72(m,1H),7.29-7.24(m,3H),7.00-6.97(m,2H),6.59-6.56(m,2H),6.28-6.26(m,1H),5.28-5.20(m,1H),3.98-3.96(m,1H),3.79-3.77(m,1H),3.59(s,1H),3.24-3.23(m,3H),2.84(m,2H),2.31-2.28(m,4H),2.26-2.24(m,4H),1.49-1.47(m,2H),1.34-1.24(m,4H),1.00(s,6H),0.96-0.90(m,2H)。
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyridine [2,3-b][1,4]Synthesis of oxazepan-1 (7H) -yl) benzoic acid: in a vial, 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) is placed]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoate (200 mg,0.28mmol,1 eq.) M eOH (6 mL), 1, 4-dioxane (6 mL), H 2 O (2 mL), naOH (67 mg,1.68mmol,6 eq.). The resulting solution was stirred overnight in an oil bath at 60 ℃. The resulting mixture was concentrated under vacuum. The pH of the solution was adjusted to 5-6 with HCl (2 mol/L). The resulting solution was extracted with 2X 50mL of dichloromethane/MeOH (v: v=10:1). The resulting mixture was washed with 2X 200mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. 157mg (81.0%) of 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) were obtained]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid as a pale yellow solid. LC-MS (ES, M/z): M+=694, R, T=2.43 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0,Poroshell HPH-C18,2.7 μm; eluent a: water (0.05% NH) 4 HCO 3 ) The method comprises the steps of carrying out a first treatment on the surface of the Eluent B: acetonitrile; linear gradient from 5% acetonitrile to 100% acetonitrile, 5.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min.
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl-piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4-trifluoromethyl-3, 4-dihydro-2H-pyrrole [3',2',5, 6) ]Pyrrolo [2,3-b][1,4]Synthesis of oxazepan-1 (7H) -yl) benzamide: in a round bottom flask, 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) was placed]-2-yl) methyl-piperazin-1-yl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid (57 mg,0.08mmol,1 eq.) DCM (3 mL), 3-nitro-4- [ [ (oxa-4) methyl]Amino group]Benzene-1-sulfonamide (25.2 mg,0.08mmol,1 eq.) EDCI (30.6 mg,0.16mmol,2 eq.) DMAP (39.0 mg,0.32mmol,4 eq.). The resulting solution was stirred overnight at 25 ℃, and the resulting mixture was concentrated under vacuum. The residue was treated with dichloroethane/methanol (10:1) on a silica gel column. The crude product was purified by Prep-HPLC under the following conditions (IntelFlash-1): a column, a C18 reverse phase column; mobile phase, water (10 mmol/L NH) 4 HCO 3 +0.05%NH 3 .H 2 O) and CH 3 CN(20.0%CH 3 CN up to 90.0% in 30 min); detector, UV 220nm. 32mg (40.0%) of 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) are obtained]-2-yl) methyl-piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrole [3',2',5, 6)]Pyrido [2,3-b ][1,4]Oxazepan-1 (7H) -yl) benzamide was a yellow solid. LC-MS (ES, M/z): M+1=991, R, T=2.41 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Agilent Poroshell HPH-C18,2.7 μm; eluent a: water (0.05% ammonia); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min. 1 H NMR (300 MHz, CD Cl3, ppm) delta 11.43 (ds, 1H), 9.99 (ds, 1H), 8.72 (s, 1H), 8.40 (ds, 1H), 7.93-7.85 (m, 2H), 7.37-7.35 (m, 2H), 7.26 (s, 1H), 7.02-6.93 (m, 3H), 6.71-6..64 (m, 3H), 6.27 (s, 1H), 4.58 (s, 1H), 4.05-3.15 (m, 23H), 2.70-2.35 (m, 7H), 2.03 (s, 3H), 1.92-1.90 (m, 1H), 1.89-1.87 (m, 2H), 1.71-1.67 (m, 2H), 1.53-1.45 (m, 3H), 1.00 (s, 6H), and measurement of the spectrum by the probe (37 MHz were performed by the OF BB 62 MHz.
Example 4
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (3-nitro-4- [ [ (oxa-4-yl) methyl ] amino ] benzenesulfonyl) -2- [ 14-thio-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) sulfonyl]Synthesis of methyl propionate: in a round bottom flask, 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] was placed]Methyl group]-1H-pyrrole [2,3-b ]]Pyridine (3 g,8.7mmol,1 eq.) methyl 3-sulphonylpropionate (2.1 g,17.4mmol,2.0 eq.), ACN (30 mL), cs 2 CO 3 (7.1 g,21.7mmol,2.5 g.)Amount). The resulting solution was stirred at 70 ℃ for 1h, the solids were filtered and the resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0-10%) on a silica gel column. 300mg (7.75%) of 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) are obtained]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) sulfonyl]Methyl propionate was a colorless oil. LC-MS (ES, M/z): M+1=445/447, R, T=1.47 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient.
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl) sulfonyl]Synthesis of propan-1-ol: methyl 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) sulfonyl]Methyl propionate (300 mg,0.67mmol,1 eq.) and THF (10 mL) were placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. Subsequently LiAlH is added at-78 DEG C 4 (51 mg,1.3mmol,2 eq.). The resulting solution was stirred at room temperature for 1h, after the reaction was completed, 0.5mL of water was added to cool the reaction mixture, and 0.5mL of NaOH (10% H) was continuously added to the solution in an ice bath 2 O) and 1.5mL of water, and the solid was filtered. The resulting mixture was concentrated under vacuum. 200mg (71.14%) of 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) are obtained]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl) sulfonyl]Propan-1-ol as a yellow oil. H-NMR (CDCl 3,300 ppm): 7.99 (s, 1H), 7.24-7.20 (m, 1H), 6.49-6.41 (m, 1H), 5.32 (s, 2H), 3.84-3.78 (m, 2H), 3.58-3.50 (m, 2H), 3.46-3.40 (m, 2H), 2.09-2.01 (m, 2H), 0.96-0.88 (m, 2H), 0.05 (s, 9H).
N- [6- [ (3-hydroxypropyl) thio ] s]-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-5-yl]-synthesis of 4-methylbenzene-1-sulfonamide: 4-methylbenzene-1-sulfonamide (1.6 g,9.6mmol,2 eq.) and 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) sulfonyl]Propan-1-ol (2 g,4.8mmol,1 eq.) 1, 10-phenanthroline (0.17 g,0.94mmol,0.20 eq.) CuI (0.18 g,0.96mmol,0.2 eq.) Cs 2 CO 3 (3.1 g,9.6mmol,2 eq.) in DMSO (30 mL) was placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. By a means ofThe solution was stirred at 120℃for 72h. The resulting solution was treated with 30mL of H 2 O extraction. The solid was filtered. The resulting solution was extracted with 3×50mL of ethyl acetate, and the mixture was dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0:1-1:1) on a silica gel column. 1.0g (41.11%) of N- [6- [ (3-hydroxypropyl) thio ] are obtained]-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-5-yl]-4-methylbenzene-1-sulfonamide as a white solid. LC-MS (ES, M/z): M+1=508, R, T= 2.845min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient.
10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tridecyl-1 (9), 2,5, 7-tetraene: in a round bottom flask, N- [6- [ (3-hydroxypropyl) thio ] was placed]-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-5-yl]-4-methylbenzene-1-sulfonamide (240 mg,0.47mmol,1 eq.) in THF (5 mL), PPh 3 (248 mg,0.95mmol,2 eq.). DEAD (164 mg,0.95mmol,2 eq.) was then added dropwise with stirring at 0deg.C. The resulting solution was stirred at room temperature for 12h. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:2) on a silica gel column. 200mg (86.40%) of 10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy are obtained]Methyl group]-14-thia-2, 4, 10-triazacyclo [7.4.0.0 ] [3,7 ],]]tridec-1 (9), 2,5, 7-tetraene, is a colorless solid. H-NMR (CDCl 3,300 ppm): 7.82 (s, 1H), 7.69-7.58 (m, 3H), 7.38-7.36 (d, j=6 hz, 2H), 6.55-6.54 (d, j=3 hz, 2H), 5.55 (s, 2H), 4.30-4.23 (m, 2H) 4.06-4.02 (m, 2H), 3.57-3.49 (m, 2H), 2.77 (s, 2H), 2.50 (s, 3H), 2.07-1.99 (m, 3H), 1.24-1.04 (m, 9H), 0.87-0.84 (m, 3H), 0.02 (s, 9H).
4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-thio-2, 4, 10-triazacyclo [7.4.0.0 ] [3,7 ]]]Synthesis of tridecyl-1 (9), 2,5, 7-tetraene: in a round bottom flask, naphthalene (314 mg,2.4mmol,6 eq), na (90 mg,3.9mmol,9.6 eq) and DME (5 mL) were placed and the resulting solution stirred at room temperature for 0.5h. Adding the resulting solution to 10- (4-methylbenzenesulfonyl) in another round bottom flaskPhenyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group ]-14-thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7]]]Tridec-1 (9), 2,5, 7-tetraene (200 mg,0.41mmol,1 eq.) in THF (10 mL). The resulting solution was stirred at room temperature for 3h. Then 1mL NH was added 4 Cl, stopping the reaction. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column. The final product was 120mg (87.57%) of 4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradecane-1 (9), 2,5, 7-tetraene brown solid. LC-MS- (ES, m/z): m+1=649, r, t=0.80 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient.
Synthesis of methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.6.0.0 [3,7] ] penta-1 (9), 2,5, 7-tetraen-10-yl) benzoate: methyl 4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-thio-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraene (120 mg,0.36mmol,1 eq.) 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-yl ] methyl ] piperazin-1-yl) benzoate (380 mg,0.71mmol,2 eq.) caesio methaneperoxoate caesium (233 mg,0.71mmol,2 eq.) toluene (3 mL), xantPhosPd (34 mg,0.04mmol,0.1 eq.) was placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred at 100℃overnight. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column. The final product was 150mg (53.32%) methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate as a yellow oil. LC-MS (ES, M/z): M+1=786, R, T=1.26 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient. H-NMR (CDCl 3,300 ppm): 7.64-7.61 (d, J=9 Hz, 1H), 7.35 (s, 1H), 7.28-7.26 (m, 2H), 7.19-7.18 (m, 1H), 6.99-6.97 (m, 2H), 6.52-6.47 (m, 2H), 6.27-6.26 (d, J=3 Hz, 1H), 5.61 (s, 2H), 3.96-3.84 (m, 2H), 3.60-3.54 (m, 5H), 3.28-3.16 (m, 6H), 2.82 (s, 2H), 2.33-2.24 (m, 6H), 2.06-2.02 (m, 4H), 1.49-1.45 (m, 2H), 1.28-1.19 (m, 4H), 1.03-0.88 (m, 6H), 0.00 (m, 9.00).
Synthesis of methyl-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] pentadec-1 (9), 2,5, 7-tetraen-10-yl ] benzoate: in a round bottom flask was placed 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-thio-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (150 mg,0.19mmol,1 eq), ethane-1, 2-diamine (229 mg,3.8mmol,20 eq), TBAF (997 mg,3.8mmol,20 eq) and THF (10 mL). The resulting solution was stirred at 70℃for 14h. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column. The final product was 50mg (39.95%) methyl-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-thio-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] pentadec-1 (9), 2,5, 7-tetraen-10-yl ] benzoate as a yellow solid. LC-MS (ES, M/z): M+1=656, R, T=1.05 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-thio-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] pentadec-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid: to a round bottom flask was added methyl-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2-thio-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (65 mg,0.10mmol,1 eq.), 1-4-dioxane (1 mL), water (1 mL), and NaOH (23.95 mg,0.60mmol,6 eq.). The resulting solution was stirred at 90℃for 14h. The pH of the solution was adjusted to 5 with hydrochloric acid (2 mol/L). The resulting mixture was concentrated. The residue was treated with dichloroethane/methanol (0-10%) on a silica gel column. The final product was 30mg (46.80%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-thio-2, 4, 10-triazatricyclo [7.6.0.0 ] [3,7] ] pentadec-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid as a white solid.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (3-nitro-4- [ [ (oxa-4-yl) methyl ] amino ] benzenesulfonyl) -2- [ 14-thio-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl ] benzamide: in a round bottom flask was placed 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7] tride-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid (30 mg,0.05mmol,1 eq.), 3-nitro-4- [ [ (oxa-4-yl) methyl ] amino ] benzene-1-sulfonamide (17 mg,0.06mmol,1.20 eq.), EDCI (18 mg,0.09mmol,2 eq.), DMAP (23 mg,0.19mmol,4 eq.) and DCM (3 mL). The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The crude product was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): column, C18 silica gel; mobile phase, water (0.1% fa) and ACN (48.0% ACN rise to 53.0% in 7min, hold 95.0% in 1min, fall to 48.0% in 1 min) for 5 min; detector, UV 254nm. The final product was 10.6mg (24.15%) 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (3-nitro-4- [ [ (oxa-4-yl) methyl ] amino ] benzenesulfonyl) -2- [ 14-thio-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] pentadec-1 (9), 2,5, 7-tetraen-10-yl ] benzamide as a yellow solid. LC-MS (ES, M/z): M+1=939, R, T=3.55 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient. H-NMR (CDCl 3,300 ppm): 8.71 (s, 1H), 8.49 (s, 1H), 7.99-7.97 (m, 1H), 7.81-7.77 (m, 1H), 7.38-7.28 (m, 4H), 7.01-6.93 (m, 2H), 6.88-6.72 (m, 3H), 3.36 (s, 1H), 4.06-3.26 (m, 18H), 2.73-2.22 (m, 6H), 2.13-1.73 (m, 3H), 1.79-1.25 (m, 6H), 1.00 (s, 6H).
Example 5
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-N- (3-nitro-4- [ [ (oxan-4-yl) methyl)]Amino group]Benzenesulfonyl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoate: in a round bottom flask, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) was placed]Methyl group]Piperazin-1-yl) -2- [ 14-thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoate (30 mg,0.046mmol,1 eq.) DCM (3 mL), m-CPBA (19.72 mg,0.114mmol,2.50 eq.). The resulting solution was stirred at room temperature overnight. Then 1mL of water was added to stop the reaction. The resulting solution was extracted with 2X 5mL of dichloromethane. The residue was treated with ethyl acetate/petroleum ether (0:1-1:1) on a silica gel column. 20mg (63.57%) of methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoate as a white solid. LC-MS-PH-PHNW-4-65-8: (ES, M/z) m+1=688, r, t=0.967 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoic acid: in a round bottom flaskPlacing 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -sulfur-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7,]]tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoate (20 mg,0.029mmol,1 eq.) 1, 4-dioxane (1 mL), water (1 mL), naOH (6.97 mg,0.174mmol,6.00 eq.). The resulting solution was stirred at 70 ℃ overnight. The pH of the solution was adjusted to 5 with hydrochloric acid (1 mol/L). The resulting solution was extracted with 2X 3mL ethyl acetate. The resulting mixture was washed with 2X 3mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. 13mg (66.35%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid as a white solid. LC-MS-PH-PHNW-4-65-9: (ES, M/z) m+1=674, r, t=1.945 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-N- (3-nitro-4- [ [ (oxan-4-yl) methyl)]Amino group]Benzenesulfonyl) synthesis of benzamide: in a round bottom flask, 3-nitro-4- [ [ (oxan-4-yl) methyl]Amino group]Benzene-1-sulfonamide (6.08 mg,0.019mmol,1 eq.) and 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ6-thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (13 mg,0.019mmol,1 eq.) DCM (0.47 mL,5.506mmol,381.56 eq.) DMAP (9.42 mg,0.077mmol,4 eq.) EDCI (7.39 mg,0.039mmol,2 eq.). The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The crude product was purified by Prep-HPLC under the following conditions (Waters-I): chromatographic column, XBridge Shield C18 OBD chromatographic column, 5 μm,19 x 150mm; mobile phase: water (0.05% fa) and ACN (46% mobile phase B reached 51% in 7 min); detector, UV 220 &254nm. Yield 2.5mg (6.24%)) 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [14, 14-dioxo-14λ 6 -thio-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-N- (3-nitro-4- [ [ (oxaalkane) methyl)]Amino group]Benzenesulfonyl) benzamide as a yellow solid. LC-MS-PH-PHNW-4-65-0: (ES, M/z) m+1=971.5, r, t= 3.243min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient.
Example 6
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of 2-methoxy-1, 3-methylnicotinonitrile: 2-Methoxymalonic acid 1, 3-dimethyl ester (20 g,123mmol,1 eq.) and THF (250 mL) were placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. Dropwise adding LiAlH in ice bath 4 (23.4 g,616mmol,5 eq.). The resulting solution was stirred at room temperature for 16h. After the reaction was completed, the reaction mixture was cooled by adding 23.4mL of water, and 23.4mL of LNaOH (10% H) was continuously added to the solution in an ice bath 2 O) and 70mL water. The solid was filtered. The resulting mixture was concentrated under vacuum. 12g (91.67%) of 2-dioxane-1, 3-carbaldehyde were obtained as a colorless oil. 1 H NMR(300MHz,CDCL3,ppm)δ3.80-3.63(m,4H),3.47(s,3H),3.36-3.32(m,1H),3.25(bs,2H)。
3- (5-bromo-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrolo [2,3-b]Synthesis of pyridin-6-oxy) -2-methoxypropan-1-ol: in a round bottom flask was placed 2-methoxypropyl-1, 3-diol (1.84 g,17.4mmol,1.2 eq.) in) And THF (70 mL), naH (0.87 g,36.2mmol,2.5 eq.) was added, and after stirring the mixture in an ice bath for 30min, 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] was then added at room temperature]Methyl group]-1H-pyrrolo [2,3-b]Pyridine (5.0 g,14.5mmol,1 eq.). The resulting solution was stirred at 80℃for 4h. The reaction was quenched by the addition of 20mL of water. The resulting solution was extracted with 3X 50mLDCM and the organic layers were combined. The resulting mixture was washed with 1X 50mL of brine. The mixture was dried over anhydrous sodium sulfate. The solid was filtered. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0-40%) on a silica gel column. The final product was 4.0g (64.03%) 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl) oxy]-2-difluoropropan-1-ol as a pale yellow oil. 1 H NMR(300MHz,CDCL3,ppm)8.09(s,1H),7.20(d,J=3.6Hz,1H),6.44(d,J=3.6Hz,1H),5.62(s,2H),4.60-4.58(m,2H),3.97-3.95(m,1H),3.92-3.90(m,2H),3.79(s,3H),3.65-3.60(m,2H),0.98-0.93(m,2H),0.01(s,9H)。
N- (6- (3-hydroxy-2-methoxypropoxy) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrolo [2, 3-b)]Synthesis of pyridin-5-yl) -4-methylbenzenesulfonamide: 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]-2-methoxypropan-1-ol (4.0 g,9.3mmol,1 eq.) T S -NH 2 (4.76 g,27.8mmol,3.00 eq.) Cs 2 CO 3 (9.06 g,27.8mmol,3.0 eq.) CuI (0.88 g,4.6mmol,0.5 eq.) 1, 10-phenanthroline (0.50 g,2.8mmol,0.3 eq.) and DMSO (100 mL) were placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred in an oil bath at 120℃for 24h. The reaction mixture was cooled. The resulting solution was diluted with 500ml cm. The solid was filtered. The mixture was dried over anhydrous sodium sulfate. The solid was filtered. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0-60%) on a silica gel column. 2.7g (55.82%) of N- [6- (3-hydroxy-2-methoxypropoxy) -1- [ [2- (trimethylsilyl) ethoxy ] were obtained]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-5-yl]-4-methylbenzene-1-sulfonamide as a solid. 1 H NMR(300MHz,CDCL3,ppm)8.09(s,1H),7.61-7.59(m,2H),7.17-7.15(m,3H),6.75(s,1H),6.46(d,J=3.3Hz,1H),5.48(s,2H),4.28-4.23(m,2H),3.59-3.44(m,8H),2.37(s,3H),0.92-0.86(m,3H),0.01(s,9H)。
3-methoxy-1-tosyl-7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5,6]Pyrido [2,3-b][1,4]Synthesis of oxazepan: n- [6- (3-hydroxy-2-methoxypropoxy) -1- [ [2- (trimethylsilyl) ethoxy ] ethoxy]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-5-yl]-4-methylbenzene-1-sulfonamide (500 mg,0.96mmol,1 eq.) PPh 3 (1.2 g,4.79mmol,5.0 eq.) and THF (10 mL) were placed in a round bottom flask, nitrogen was purged to maintain an inert atmosphere, and DEAD (830 mg,4.79mmol,5.0 eq.) was added dropwise under ice-bath. The resulting solution was stirred at room temperature for 2h. The resulting solution was diluted with 50mL of DCM. The resulting mixture was washed with 3X 20mL of water and 1X 20mL of brine. The mixture was dried over anhydrous sodium sulfate. The solid was filtered. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (0-60%) on a silica gel column. 370mg (76.65%) of 3-methoxy-1-tosyl-7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrole [3',2',5,6 ] are obtained]Pyridine [2,3-b][1,4]Oxazepan as a white solid. LC-MS (ES, M/z): M+1=504, RT=2.40 min.
3-methoxy-7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5,6 ]Pyridine [2,3-b][1,4]Synthesis of oxazane: in a round bottom flask, in N 2 Na (158 mg,6.9mmol,1.0 eq.) naphthalene (884 mg,6.9mmol,10 eq.) and DME (3 mL) were added. The reaction mixture was stirred at room temperature until Na and naphthalene were completely dissolved. 3-methoxy-1-tosyl-7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5,6 ] is added thereto at-78deg.C]Pyridine [2,3-b][1,4]A solution of oxazepan (350 mg,0.69mmol,1 eq.) in THF (5 mL). The resulting solution was stirred at-60 to-40 ℃ for 2-3 h until TLC consumed the starting material. Then 5mLNH was added at-10 ℃ 4 Cl to stop the reaction. The resulting solution was extracted with 3X 10mL ethyl acetate. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was eluted with ethyl acetate/petroleum ether (1/3) on a silica gel column. 214mg is obtained(88%) 3-methoxy-7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrole [3',2',5,6 ]]Pyridine [2,3-b][1,4]Oxazepan as a white solid. LC-MS (ES, M/z): M+1=350, RT=2.26 min.
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl-piperazin-1-yl) -2- (3-methoxy-7- ((2- (trimethylsilyl) ethoxy) methyl) -3, 4-dihydropyrrole [3',2',5,6 ]Pyrido [2,3-b][1,4]Synthesis of methyl oxazepan-1 (7H) -yl) benzoate: 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) benzoate (1.34 g,2.28mmol,4 eq), toluene (20 mL), 4- (trifluoromethyl) -7- ((2- (trimethylsilyl) ethoxy) methyl) -1,3,4, 7-tetrahydro-2H-pyrrolo [3',2',5, 6)]Pyridine [2,3-b][1,4]Oxazepan (220 mg,0.63mmol,1 eq.) Cs 2 CO 3 (1.02G, 3.15mmol,5 eq.) XantPhos Pd 2G (25 mg,0.06mmol,0.1 eq.) was placed in a round-bottomed flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred at 110℃overnight. The resulting solution was diluted with 30mL of water. The resulting solution was extracted with 2X 30mL ethyl acetate. The resulting mixture was washed with 1X 30mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column. 403mg of crude (80.0%) 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) were obtained]-2-yl) methyl-piperazin-1-yl) -2- (3-methoxy-7- ((2- (trimethylsilyl) ethoxy) methyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6 ]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid methyl ester as a yellow solid.
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyridine [2,3-b][1,4]Synthesis of methyl oxazepan-1 (7H) -yl) benzoate: in a vial, 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) is placed]-2-yl) methyl-piperazin-1-yl) -2- (3-methoxy-7- ((2- (trimethylsilyl) ethoxy) methyl) -3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H)) -yl) benzoate (380 mg,0.48mmol,1 eq.) THF (20 mL), tbaf.3h 2 O (7516 mg,2.4mmol,5 eq.) ethane-1, 2-diamine (576 mg,9.6mmol,20 eq.). The resulting solution was stirred overnight in an oil bath at 70 ℃. The resulting solution was diluted with 20mL of water. The resulting solution was extracted with 3X 30mL of ethyl acetate. The resulting mixture was washed with 2X 20mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0-50%) on a silica gel column. 338mg (93%) of 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) are obtained ]-2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyridine [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid methyl ester as a pale yellow solid. LC-MS (ES, M/z): M+1=670, RT=2.00 min.
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-dihydro-2H-pyrrole [3',2':5,6]Pyridine [2,3-b][1,4]Synthesis of oxazepan-1 (7H) -yl) benzoic acid: in a vial, 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) is placed]-2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoate (200 mg,0.30mmol,1 eq.) MeOH (6 mL), 1, 4-dioxane (6 mL), H 2 O (2 mL), naOH (72 mg,1.8mmol,6.00 eq.). The resulting solution was stirred overnight in an oil bath at 60 ℃. The resulting mixture was concentrated under vacuum. The pH of the solution was adjusted to 5-6 with HCl (2 mol/L). The resulting solution was extracted with 2X 50mL of dichloromethane/MeOH (v: v=10:1). The resulting mixture was washed with 2X 200mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. 152mg (81.0%) of 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) were obtained ]-2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid as a pale yellow solid. LC-MS (ES, M/z): M+=656, RT=2.43 min.
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-tetralinhydrogen-2H-pyrrole [3',2',5,6 ]]Pyrido [2,3-b][1,4]Synthesis of oxazepin-1 (7H) -yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) benzenesulfonyl) benzamide: in a round bottom flask, 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) was placed]-2-yl) methyl) piperazin-1-yl) -2- (3-methoxy-3, 4-dihydro-2H-pyrrole [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepan-1 (7H) -yl) benzoic acid (50 mg,0.08mmol,1 eq.) DCM (3 mL), 3-nitro-4- [ [ (oxa-4) methyl]Amino group]Benzene-1-sulfonamide (25.2 mg,0.08mmol,1 eq.) EDCI (30.6 mg,0.16mmol,2 eq.) DMAP (39.0 mg,0.32mmol,4 eq.). The resulting solution was stirred overnight at 25 ℃, and the resulting mixture was concentrated under vacuum. The residue was treated with dichloroethane/methanol (10:1) on a silica gel column. Crude-purified by Prep-HPLC under the following conditions (IntelFlash-1): a column, a C18 reverse phase column; mobile phase, water (10 mmol/L NH) 4 HCO 3 +0.05%NH 3 .H 2 O) and CH 3 CN(20.0%CH 3 CN up to 90.0% in 30 min); detector, UV 220nm. 32mg (40.0%) of 4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) are obtained]-2-yl) methyl) piperazin-1-yl) -2- (-3, 4-dihydropyrrol [3',2',5,6 ]]Pyrido [2,3-b][1,4]Oxazin-1 (7H) -yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) benzenesulfonyl) benzamide as a yellow solid. LC-MS (ES, M/z): M+1=953, RT=3.44 min. 1 H NMR(300MHz,DMSO-d6,ppm)δ11.00(ds,1H),8.56(s,2H),7.47-7.44(m,2H),7.36-7.33(m,2H),7.08-7.04(m,3H),6.78-6..65(m,2H),6.57-6.54(m,1H),6.43(s,1H),6.04(s,1H),4.05-3.15(m,20H),3.13(s,1H),2.70-2.35(m,4H),2.03(s,3H),1.92-1.90(m,1H),1.89–1.87(m,2H),1.71-1.67(m,2H),1.53-1.45(m,3H),0.97(s,6H)。
Example 7
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -1.4,4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [4- [ [ (2R) -1, 4-dioxan-2-yl ] methyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7] tride-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- ([ [ (2R) -1, 4-dioxan-2-yl)]Methyl group]Synthesis of amino) -3-nitrobenzene-1-sulfonamide: 4-fluoro-3-nitrobenzene-1-sulfonamide (1.43 g, 0.0070 mmol,1 eq.) 1- [ (2R) -1, 4-dioxane-2-yl]Carboxamide hydrochloride (1 g,6.510mmol,1 eq.), THF (30 mL), cs 2 CO 3 (8.48 g,0.026mmol,4 eq.) was added to a round bottom flask. The resulting solution was stirred overnight in an oil bath at 50 ℃. The solid was collected by filtration. The solid was dried in an oven under reduced pressure. 1.8g (87.14%) of 4- ([ [ (2R) -1, 4-dioxane-2-yl) are obtained]Methyl group]Amino) -3-nitrobenzene-1-sulfonamide as a yellow solid. LC-MS (ES, M/z): M+1=318, RT=0.740 min.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ],]]tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl) benzoate (4.35 g,8.21mmol,1.87 eq), toluene (20 mL), 4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,4.98,7-tetraene (1.4 g,4.39mmol,1 eq.) Cs 2 CO 3 (7.1G, 21.85mmol,4.98 eq.) XantPhos Pd 2G (284 mg,0.66mmol,0.15 eq.) was placed in a round-bottomed flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred at 110℃overnight. The resulting solution was diluted with 300mL of water. The resulting solution was extracted with 2X 100mL ethyl acetate. The resulting mixture was washed with 1X 300mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:2) on a silica gel column. 2.1g (62%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), methyl 2,5, 7-tetraen-10-yl benzoate as brownColor solids. LC-MS (ES, M/z): M+=770, RT= 1.318min.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: in a vial, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) is placed]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraene-1 (2.1 g,3.29mmol,1 eq.) THF (20 mL), TBAF.3H 2 O (5 g), ethane-1, 2-diamine (1.4 g). The resulting solution was stirred overnight in an oil bath at 70 ℃. The resulting solution was diluted with 200mL of water. The resulting solution was extracted with 3X 30mL ethyl acetate. The resulting mixture was washed with 2X 200mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo and recrystallized from ethyl acetate/petroleum ether (1:5). 1.4g (80%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,9 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate as a white solid. LC-MS (ES, M/z): M+=640, RT=1.023 min.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] ] tridec-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid: in a vial, methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7] ] tride-1 (9), 2,5, 7-tetraen-10-yl ] benzoate (1.4 g,2.24mmol,1 eq), etOH (25 mL), dioxane (25 mL), 4M NaOH (5 mL). The resulting solution was stirred in an oil bath at 80℃for 4h. The resulting mixture was concentrated under vacuum. The pH of the solution was adjusted to 5-6 with HCl (2 mol/L). The solid was collected by filtration. 1.1g (80%) 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid was obtained as a white solid. LC-MS (ES, M/z): M+1=626, RT= 2.138min.
4- (4- [ [2- (4-chlorophenyl) -1.4,4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [4- [ [ (2R) -1, 4-dioxan-2-yl]Methyl group]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: in a vial, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) is placed]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tetracyclic-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (150 mg,0.240mmol,1 eq.) DCM (3 mL), 4- [ [ (2R) 4-fluorooxalan-2-yl) methyl]Amino group]3-Nitrophenyl-1-sulfonamide (76 mg,0.240mmol,1 eq.) EDCI (92 mg,0.480mmol,2 eq.) DMAP (117 mg,0.958mmol,4 eq.). The resulting solution was stirred at 25 ℃ overnight. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC under the following conditions (2#SHIMADZU (HPLC-01)): column, XBridge Prep C18 OBD column, 5 μm,19 x 150 mm; mobile phase, ACN and water (0.05% nh 3 .H 2 O) (20% b phase, 75% in 1min, 95% in 7min, 95% in 1min, 20% in 1 min); detector, 254/220nm. 28mg (12.63%) of 4- (4- [ [2- (4-chlorophenyl) -2, 4-dimethylcyclohex-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -N- [4- [ [ (2R) -1, 4-dioxan-2-yl) methyl]Amino) -3-nitrobenzene) benzenesulfonyl]-2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7]]]Tetracyclic-1 (9), 2,5, 7-tetraen-10-yl]Benzamide is a yellow solid. LC-MS (ES, M/z): M+1=925, R, T= 3.425min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0SUPELCO Ascentis Express C18,2.7 micrometers; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min. 1 H NMR(300MHz,DMSO-d 6 ,ppm)δ11.90(s,1H),11.20(s,1H),8.60–8.41(m,2H),7.64(d,J=8.7Hz,1H),7.47(d,J=8.7Hz,1H),7.36(d,J=8.1Hz,2H),7.20(t,J=2.9Hz,1H),7.07(d,J=8.1Hz,2H),6.90(d,J=12.1Hz,2H),6.70(d,J=8.6Hz,2H),6.19–6.06(m,1H),4.20(d,J=6.5Hz,2H),3.88–3.71(m,3H),3.69–3.32(m,8H),3.28(s,0H),3.20(s,4H),2.78(s,2H),2.22(d,J=18.1Hz,6H),1.98(s,4H),1.51–1.34(m,2H),0.95(s,6H)。
Example 8
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [4- [ [ (2S) -1, 4-dioxan-2-yl ] methyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ],]]tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl) benzoate (4.35 g,8.21mmol,1.87 eq), toluene (20 mL), 4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene (1.4 g,4.39mmol,1 eq.) Cs 2 CO 3 (7.1G, 21.85mmol,4.98 eq.) XantPhos Pd 2G (284 mg,0.66mmol,0.15 eq.) was charged to a round bottom flask and nitrogen was purged to maintain an inert atmosphere and the resulting solution was stirred overnight at 110 ℃. The reaction mixture was diluted with water (300 mL), and extracted twice with ethyl acetate (100 mL). The resulting organic phase was washed with brine (300 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The crude product obtained was purified on a silica gel column with ethyl acetate/petroleum ether (1:2) as eluent. Finally, a brown solid product, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) is obtained]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate 2.1g (yield 62%). LC-MS (ES, M/z) [ M+H ]] + =770。
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethyl)Methylcyclohexyl-1-en-1-yl]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) in a reaction flask]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene-1 (2.1 g,3.29mmol,1 eq.) THF (20 mL), TBAF.3H 2 O (5 g), 1, 2-ethylenediamine (1.4 g). The resulting solution was stirred overnight in an oil bath at 70 ℃. The reaction mixture was diluted with water (200 mL), and extracted three times with ethyl acetate (30 mL). The resulting organic phase was washed twice with brine (200 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The crude product obtained is purified by recrystallisation from ethyl acetate/petroleum ether (1:5) to give the product as a white solid, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,9 ] ]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate 1.4g (80% yield). LC-MS (ES, M/z) [ M+H ]] + =640。
4- ([ [ (2S) -1, 4-dioxan-2-yl)]Methyl group]Preparation of amino) -3-nitrobenzene-1-sulfonamide: 4-fluoro-3-nitrobenzene-1-sulfonamide (1.43 g,0.007mmol,1 eq.) 1- [ (2S) -1, 4-dioxane-2-yl]Carboxamide hydrochloride (1 g,6.510mmol,1 eq.), THF (30 mL), cs 2 CO 3 (8.48 g,0.026mmol,4 eq.) in a round bottom flask. The resulting solution was stirred overnight in an oil bath at 50 ℃. After the reaction solution was filtered, the solid was collected. The solid obtained is dried under reduced pressure in an oven to finally obtain a yellow solid product, 4- ([ [ (2S) -1, 4-dioxane-2-yl)]Methyl group]Amino) -3-nitrobenzene-1-sulfonamide 1.82g (yield 88.1%). LC-MS (ES, M/z) [ M+H ]] + =318。
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [4- [ [ (2S) -1, 4-dioxan-2-yl]Methyl group]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: in a reaction flask, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ] ]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (150 mg,0.240mmol,1 eq.) DCM (3 mL), 4- [ [ (2S) -1, 4-dioxan-2-yl) methyl]Amino group]3-Nitrophenyl-1-sulfonamide (76 mg,0.240mmol,1.00 eq.) EDCI (92 mg,0.480mmol,2 eq.) DMAP (117 mg,0.958mmol,4 eq.). The resulting solution was stirred at 25 ℃ overnight. The resulting reaction solution was concentrated under vacuum. The crude product was purified by Prep-HPLC under the following conditions: chromatographic column, XBridge Prep C18 OBD column, 5 μm,19 x 150mm; mobile phase, acetonitrile and 0.05% ammonia (75% acetonitrile in 1min, 95% in 7min and 1min, and then 1min down to 20%); detector, 254/220nm. Finally, a yellow solid product, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) is obtained]Methyl group]Piperazin-1-yl) -N- [4- [ [ (2S) -1, 4-dioxan-2-yl) methyl]Amino) -3-nitrobenzene) benzenesulfonyl group]-2- [ 14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide 29mg (13.1% yield). LC-MS (ES, M/z) [ M+H ]] + =925. 1 H NMR(300MHz,DMSO-d 6 ,ppm)δ11.90(s,1H),11.20(s,1H),8.60–8.41(m,2H),7.64(d,J=8.7Hz,1H),7.47(d,J=8.7Hz,1H),7.36(d,J=8.1Hz,2H),7.20(t,J=2.9Hz,1H),7.07(d,J=8.1Hz,2H),6.90(d,J=12.1Hz,2H),6.70(d,J=8.6Hz,2H),6.19–6.06(m,1H),4.20(d,J=6.5Hz,2H),3.88–3.71(m,3H),3.69–3.32(m,8H),3.28(s,0H),3.20(s,4H),2.78(s,2H),2.22(d,J=18.1Hz,6H),1.98(s,4H),1.51–1.34(m,2H),0.95(s,6H)。
Example 9
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
(S) -N- ((4- (((1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 3-difluoro-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of N- (2, 2-difluoro-3-hydroxypropyl) -4-methylbenzene-1-sulfonamide: in a round bottom flask, 3-amino-2, 2-difluoropropan-1-ol (1 g,9.002mmol,1 eq.) in DCM (10 mL), triethylamine (1.37 g, 13.39 mmol,1.50 eq.) in TsCl (1.72 g,9.002mmol,1.0 eq.). The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:2) on a silica gel column. 600mg (25.13%) of N- (2, 2-difluoro-3-hydroxypropyl) -4-methylbenzene-1-sulfonamide was obtained as a white solid. 1 H-NMR-1(300MHz,CDCl 3 ,ppm)δ7.76–7.73(d,J=9.0Hz,2H),7.35–7.32(d,J=9.0Hz,2H),5.04–5.00(m,1H),3.91–3.83(m,2H),3.46–3.35(m,2H),2.45(s,3H)。
Synthesis of N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -2, 2-difluoropropyl ] -4-methylbenzene-1-sulfonamide: in a round bottom flask, 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridine (2.60 g,7.530mmol,1.00 eq.) N- (2, 2-difluoro-3-hydroxypropyl) -4-methylbenzene-1-sulfonamide (2 g, 7.39 mmol,1 eq.) dioxane (20 mL,236.082mmol,31.31 eq.) was placed, and then NaH (453 mg,11.3mmol,1.5 eq., 60%) was added in portions at 0deg.C and the resulting solution stirred overnight at 90deg.C. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:5) on a silica gel column. 2.4g (53.90%) of N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -2, 2-difluoropropyl ] -4-methylbenzene-1-sulfonamide were obtained as a colorless oil. LC-MS (ES, M/z): m+1=590.
12, 12-difluoro-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy ] ethyl group]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Thirteen-1.3 (7), synthesis of 5, 8-tetraene: in a round bottom flask, N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) oxy) ethoxy ] was placed]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]-2, 2-difluoropropyl]-4-methylbenzene-1-sulfonamide (2.4 g,4.064mmol,1 eq.) 2- (2-methylpropanoyl) cyclohexanone (0.14 g,0.832mmol,0.20 eq.) CuI (0.15 g,0.813mmol,0.2 eq.) Cs 2 CO 3 (2.65 g,8.128mmol,2 eq.) in DMSO (25 mL,351.964mmol,86.61 eq.). The resulting solution was stirred overnight at 120℃and 50mLH for the resulting solution 2 O and 50mLEA dilution. The solid was filtered. The resulting solution was extracted with 3X 50mL ethyl acetate and the organic layer was washed with 3X 50mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 1.5g (72.42%) of 12, 12-difluoro-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy ] were obtained]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1.3 (7), 5, 8-tetraene, as yellow oil. 1 H NMR(300MHz,CDCl 3 ,ppm):δ8.37(s,1H),7.42–7.39(m,3H),7.16–7.14(d,J=6.0Hz,2H),6.61-6.60(d,J=3.0Hz,1H),5.58(s,1H),4.23–4.15(m,2H),3.98–3.91(m,1H),3.58–3.52(m,2H),2.37(s,3H),0.95–0.89(m,2H),0.00(s,9H)。
12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] ]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tridecyl-1 (9), 2,5, 7-tetraene: in a round bottom flask was placed naphthalene (2.26 g,17.660mmol,6 eq.) DME (20 mL,206.633mmol,70.21 eq.) Na (0.65 g,28.255mmol,9.6 eq.) and the resulting solution stirred at room temperature for 0.5h. The resulting solution was placed in a 40mL round bottom flask and placed in 12, 12-difluoro-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraene (1.5 g,2.943mmol,1 eq.) in THF (20 mL,246.860mmol,83.87 eq.). The resulting solution was stirred at room temperature for 1h. Then 10mLNH was added 4 Cl, stopping the reaction. The resulting solution was extracted with 3X 50mL ethyl acetate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 0.5g (47.79%) of 12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] is obtained]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraene, is a white solid. 1 H-NMR(300MHz,CDCl 3 ,ppm):δ7.34–7.28(m,1H),7.25–7.23(d,J=6.0Hz,1H),6.37-6.36(m,1H),5.58-5.53(m,2H),4.57–4.48(m,2H),3.72–3.64(m,2H),3.59–3.52(m,2H),1.30-1.28(m,2H),0.00(s,9H)。
4-Chloro-2- (12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy)]Methyl group](-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: in a round bottom flask was placed methyl 2-bromo-4-chlorobenzoate (210.56 mg,0.844mmol,1.5 eq.) 12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraene (200 mg,0.563mmol,1 eq.) pyridine-2-carboxylic acid (13.85 mg,0.113mmol,0.2 eq.), DMSO (5 mL), cs 2 CO 3 (549.97 mg,1.688mmol,3 eq.) CuI (21.43 mg,0.113mmol,0.2 eq.). The resulting solution was stirred at 130℃for 12h, then 10mL of water was added to stop the reaction. The resulting solution was extracted with 3X 20mL ethyl acetate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:10) on a silica gel column. 180mg (46.40%) of 4-chloro-2- (12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy) are obtained]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate as a white crude solid. LC-MS (ES, M/z) m+1=524.
4- (4- [ [2- (4-chlorophenyl) -4, 12, 12-4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ],]]tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: in a round bottom flask, 4-chloro-2- (12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy) was placed]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ] ]]Tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (180.00 mg,0.343mmol,1 eq.) 1- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ]]Methyl group]Piperazine (131.44 mg,0.412mmol,1.2 eq), toluene (3.00 mL), cs 2 CO 3 (335.74 mg,1.030mmol,3 eq.) tBuXPhos Pd G3 pre-catalyst (27.29 mg,0.034mmol,0.1 eq.). The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 100mg of methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -2- (12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7],]]tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate as a white solid. LC-MS (ES, M/z): M+1= 806.4
Synthesis of methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (12.12-difluoro-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: in a round bottom flask was placed 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (12, 12-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazacyclo [7.5.0.0 [3,7] ] tride-1 (9), 2,5, 7-tetraen-10-yl) benzoate (100 mg,0.124mmol,1 eq), TBAF (648.41 mg,2.480mmol,20 eq), THF (10 mL), ethane-1, 2-diamine (149.04 mg,2.480mmol,20 eq). The resulting solution was stirred at 70℃for 12h and the resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:1) on a silica gel column. 50mg (59.63%) of methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [12, 12-difluoro-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl ] benzoate were obtained as a white solid. LC-MS (ES, M/z): m+1=676.3.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [12, 12-difluoro-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoic acid: in a round bottom flask, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) was placed]Methyl group]Piperazin-1-yl) -2- [12, 12-difluoro-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoate (50 mg,0.074mmol,1 eq.) dioxane (1 mL), meOH (1 mL), H 2 O (1 mL), naOH (17.74 mg, 0.447 mmol,6 eq.). The resulting solution was stirred at 70℃for 12 hours, and the pH of the solution was adjusted to 6 with hydrochloric acid (1 mol/L). The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 20mg (40.85%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -2- [12, 12-difluoro-14-oxoHetero-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid as a white solid. H-NMR (300 MHz, CDCl) 3 ,ppm):δ7.70-7.63(m,1H),7.31-7.25(m,2H),7.03-6.93(m,2H),6.77(s,1H),6.67-6.63(m,2H),6.06(s,1H),3.64-3.47(m,3H),3.40-3.31(m,4H),2.88(s,1H),2.42-2.27(m,5H),2.10-2.07(m,3H),1.38-1.26(m,8H),1.25-1.24(m,2H),1.02(s,6H),1.42(m,2H),0.88-0.85(m,2H)。
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) ]-2-yl) methyl) piperazin-1-yl) -2- (3, 3-difluoro-3, 4-dihydro-2H-pyrrolo [3',2',5,6]Pyrido [2,3-b][1,4]Synthesis of oxazepin-1 (7H) -yl) benzamide: in a round bottom flask, 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) was placed]Methyl group]Piperazin-1-yl) -2- [ -12, 12-difluoro-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (20.00 mg,0.030mmol,1 eq.) and (S) -4- (((1, 4-dioxan-2-yl) methyl) amino) -3-nitrobenzenesulfonamide (11.43 mg,0.036mmol,1.2 eq.) in DCM (3 mL), DMAP (14.76 mg,0.121mmol,4 eq.) EDCI (11.58 mg,0.060mmol,2 eq.). The resulting solution was stirred at room temperature for 12h. The crude product was purified by Prep-HPLC under the following conditions (Waters-2767): column, X-bridge RP18,5 μm, 19X 100mm; mobile phase, 0.03% ammonia (0.03% NH) 4 HCO 3 And NH 4 OH) and CH 3 CN(32%CH 3 CN up to 52% in 6 min); detector, UV 254nm. 11.5mg (39.68%) of (S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) are obtained]-2-yl) methyl) piperazin-1-yl) -2- (3, 3-difluoro-3, 4-dihydro-2H-pyrrolo [3',2':5,6 ]Pyrido [2,3-b][1,4]Oxazepin-1 (7H) -yl) benzamide was a white solid. LC-MS-0: (ES, M/z) m+1= 961.47. 1 H-NMR-0(300MHz,d-DMSO,ppm):δ11.01(s,1H),8.34-8.33(d,J=3.0Hz,1H),7.47-7.45(m,2H),7.38-7.35(m,2H),6.82-6.80(m,3H),6.02(s,1H),3.86–3.51(m,7H),3.39-3.17(m,5H),2.77-2.73(m,2H),2.24-2.20(m,6H),2.00(s,2H),1.42(m,2H),1.30(s,1H),0.95(s,6H)。
Example 10
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ 3-nitro-4- [ (tetrahydropyran-4-ylmethyl) amino ] benzenesulfonyl ] -2- [13- (pyridin-2-yl) -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] -tetradec-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of 3-hydroxy-3- (pyridin-2-yl) propionitrile: to a round bottom flask was added 3-oxo-3- (pyridin-2-yl) propionitrile (4.00 g,27.369mmol,1.00 eq.) and methanol (40.00 mL). Subsequently NaBH is added in portions at 0 ℃ 4 (2.07 g,54.739mmol,2 eq.). The resulting solution was stirred at room temperature for 3h. The reaction was cooled to 0deg.C and quenched by the addition of 5mL of water. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:3) on a silica gel column. The final product was 2.3g (56.72%) of 3-hydroxy-3- (pyridin-2-yl) propionitrile as a white solid. LC-MS-1: (ES, m/z): 149.1[ M+H ] ] +
Synthesis of 3-amino-1- (pyridin-2-yl) -1-propanol: to a round bottom flask was added 3-hydroxy-3- (pyridin-2-yl) propionitrile (2.30 g,15.523mmol,1.00 eq.) and THF (30.00 mL). Subsequently LiAlH is added in portions at 0 ℃ 4 (1.18 g,31.046mmol,2.00 eq.). The resulting solution was stirred in a water/ice bath at 0 ℃ for 2h. Then 3mL of water was added to stop the reaction. The resulting mixture was concentrated. The final product was 2.1g (crude) of 3-amino-1- (pyridin-2-yl) -1-propanol as a brown solid. LC-MS-2: 148.2[ M+H ]] +
N- [ 3-hydroxy-3- (pyridin-2-yl) propyl]Synthesis of tert-butyl carbamate: to a round bottom flask was added 3-amino-1- (pyridin-2-yl) -1-propanol (2.00 g, crude), DCM (30 mL), (Boc) 2 O (5.74 g,26.282mmol,2 eq.) and TEA (2.66 g,26.287mmol,2 eq.). The resulting solution was stirred at room temperature for 14h. The resulting mixture was concentrated. The residue was purified on a silica gel column with ethyl acetateThe ester/petroleum ether (1:2) eluted. The final product was 700mg (21.11%) of N- [ 3-hydroxy-3- (pyridin-2-yl) propyl]Tert-butyl carbamate as white solid. LC-MS-2: (ES, m/z): 253.1[ M+H ]] +
N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy ]-3- (pyridin-2-yl) propyl]Synthesis of tert-butyl carbamate brown oil: n- [ 3-hydroxy-3- (pyridin-2-yl) propyl ] was added to a round bottom flask]Tert-butyl carbamate (700.00 mg,2.774mmol,1 eq.) dioxane (15.00 mL), naH (222.00 mg,9.251mmol,3.33 eq.) and 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridine (957.94 mg,2.774mmol,1 eq.). The resulting solution was stirred at 70℃for 3h. The reaction was cooled to room temperature and then quenched by the addition of 2mL of water. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:3) on a silica gel column. The final product was 600mg (37.44%) of N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]-3- (pyridin-2-yl) propyl]Tert-butyl carbamate was a brown oil. LC-MS-4 (ES, m/z): 577.2[ M+H ]] +
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]-synthesis of 3- (pyridin-2-yl) propan-1-amine: n- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) oxy) ethoxy ] was added to a round bottom flask]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]-3- (pyridin-2-yl) propyl ]Tert-butyl carbamate (600.00 mg,1.039mmol,1 eq.) CH 3 CN (5.00 mL) and ZnBr 2 (2.34 g, 10.3838 mmol,10 eq.). The resulting solution was stirred in an oil bath at 80 ℃ for 14h. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:2) on a silica gel column. The final product was 350mg (70.56%) 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]-3- (pyridin-2-yl) propan-1-amine yellow oil. LC-MS-4: (ES, m/z): 477.2[ M+H ]] +
13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-synthesis of tetradec-1 (9), 2,5, 7-tetraene: to one directionA round bottom flask (the flask was purged and kept under nitrogen inert atmosphere) was charged with 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]3- (pyridin-2-yl) propan-1-amine (300.00 mg, 0.6278 mmol,1 eq.), toluene (5.00 mL,0.054mmol,0.09 eq.), brettPhos Pd G3 (56.96 mg,0.063mmol,0.10 eq.) and t-Buona (181.15 mg,1.885mmol,3 eq.). The resulting solution was stirred in an oil bath at 80℃for 6h. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:3) on a silica gel column. The final product was 150mg of 13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy ]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Fourteen-1 (9), 2,5, 7-tetraene brown solid (60.20%). LC-MS-6: (ES, m/z): 397.2[ M+H ]] +
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: 13- (pyridin-2-yl) -4- (trimethylsilyl) ethoxy was added to a vial (the vial was purged and kept in a nitrogen inert atmosphere)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Fourteen-1 (9), 2,5, 7-tetraene (80.00 mg,0.202mmol,1 eq.), toluene (3.00 mL,0.033mmol,0.16 eq.), 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ]]Methyl group]Methyl piperazin-1-yl benzoate (128.77 mg,0.242mmol,1.20 eq.) Cs 2 CO 3 (197.18 mg,0.605mmol,3 eq.) and xanthos Pd 2G (17.89 mg,0.020mmol,0.10 eq.). The resulting solution was stirred in an oil bath at 110 ℃ for 3h. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:1) on a silica gel column. The final product was 75mg (43.86%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate was a pale yellow oil. LC-MS-7 (ES, m/z): 847.5[ M+H ]] +
4-(4-[ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ]]Methyl group]Piperazin-1-yl) -2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoic acid: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a round bottom flask]Methyl group]Piperazin-1-yl) -2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate (80.00 mg,0.094mmol,1 eq), methanol (0.50 mL), dioxane (0.50 mL), water (0.25 mL,0.014mmol,0.15 eq.) and NaOH (15.10 mg,0.378mmol,4 eq.). The resulting solution was stirred in an oil bath at 70 ℃ for 14h. The resulting mixture was concentrated. The resulting solution was treated with 2mLH 2 O extraction. The pH of the solution was adjusted to 6 with AcOH. The resulting solution was concentrated and extracted with 3X 2mL of methylene chloride. The residue was applied to a silica gel column and eluted with dichloromethane/methanol (10:1). The final product was 60mg (76.26%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid as a white solid. LC-MS-8 (ES, m/z): 833.4[ M+H ]] +
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ 3-nitro-4- [ (tetrahydropyran-4-ylmethyl) amino group]Benzenesulfonyl group]-2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a vial]Methyl group]Piperazin-1-yl) -2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (20.00 mg,0.024mmol,1 eq.) 3-nitro-4- [ (tetrahydropyran-4-ylmethyl) amino group]Benzenesulfonamide (7.57 mg,0.024mmol,1 eq.), DCM (1 mL), EDCI (9.20 mg,0.048mmol,2 eq.) and DMAP (5.86 mg,0.048mmol,2 eq.)Amount). The resulting solution was stirred at room temperature for 14h. The resulting mixture was concentrated. The residue was purified by Prep-TLC with dichloromethane/methanol (10:1). The final product was 17mg (62.65%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- [ 3-nitro-4- [ (tetrahydropyran-4-ylmethyl) amino group]Benzenesulfonyl group]-2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide was a yellow solid. LC-MS-8 (ES, m/z): 1130.7[ M+H ]] +
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ 3-nitro-4- [ (tetrahydropyran-4-ylmethyl) amino group]Benzenesulfonyl group]-2- [13- (pyridin-2-yl) -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide hydrochloride: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a round bottom flask]Methyl group]Piperazin-1-yl) -N- [ 3-nitro-4- [ (tetrahydropyran-4-ylmethyl) amino group]Benzenesulfonyl group]-2- [13- (pyridin-2-yl) -4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide (15.00 mg), TBAF in THF (2M, 1.00 mL), and ethylenediamine (0.25 mL). The resulting solution was stirred in an oil bath at 70 ℃ for 14h. The resulting mixture was concentrated. The residue was purified by Prep-TLC with dichloromethane/methanol (10:1). The crude product was purified by high performance liquid chromatography under the following conditions: chromatographic column, XBridge Prep C18 OBD 19 x 150mm 5 μm; mobile phase, a:0.1% hydrochloric acid water; b: ACN; gradient: 6-95% B in 7.9 min; flow rate: 20mL/min; detector, UV220nm. The final product was 3.2mg (23.8%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- [ 3-nitro-4- [ (tetrahydropyran-4-ylmethyl) amino group]Benzenesulfonyl group]-2- [13- (pyridin-2-yl) -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide hydrochloride was a yellow solid. LC-MS-0 (ES, m/z): 1000.5[ M-HCl+H] + , 1 H NMR-0(300MHz,CDCl 3 ,ppm):δ8.81(s,1H),8.62-8.57(m,1H),8.42-8.25(m,1H),8.01-8.75(m,3H),7.45-7.34(m,3H),7.18-7.10(m,1H),7.05-6.88(m,3H),6.83-6.55(m,3H),6.27-6.17(m,1H),5.19(s,1H),4.13-3.90(m,3H),3.82-3.33(m,10H),3.24-3.02(m,2H),2.70-2.48(m,2H),2.43-2.31(m,2H),2.17-2.08(m,2H),1.96-1.8(m,1H),1.70-1.63(m,2H),1.58-1.52(m,2H),1.46-1.20(m,6H),1.01(d,J=2.1Hz,6H)。
Example 11
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ 13-oxa-2, 4, 10-triazatricyclo [7.5.0.0
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of methyl 6-amino-3-bromo-5-iodopyridine-2-carboxylate: to a round bottom flask was added 6-amino-3-bromopyridine-2-carboxylic acid methyl ester (20.00 g,86.562mmol,1 eq.), HOAc (200.00 mL) and TFA (10.00 mL). NIS (29.35 g, 130.457 mmol,1.51 eq.) was then added in portions at 25 ℃. The resulting solution was stirred overnight at 25 ℃. The stirred solution was diluted with 2000mL of water. The resulting solution was extracted with 3X 500mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 2X 2000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. Crude product from PE: EA is given 1: 1. The solid was collected by filtration. The final product was 25.8g of 6-amino-3-bromo-5-iodopyridine-2-carboxylic acid methyl ester as a red solid (83.50%). 1 H-NMR (300 MHz, chloroform-d, ppm) delta 8.14 (s, 1H), 5.21 (br, 2H), 3.99 (s, 3H).
Synthesis of methyl 6-amino-3-bromo-5- (2-ethoxyvinyl) pyridine-2-carboxylate: to a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere) was added 6-amino-3-bromo-5-iodopyridine-2-carboxylic acid methyl ester (24.00 g,67.237mmol,1 eq.), 2- (2-ethoxyvinyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (26.50 g,133.791mmol,1.99 eq.), i-PrOH (300 mL), K 3 PO 4 (42.60 g,200.691mmol,2.98 eq.) Pd (OAc) 2 (1.50 g,6.681mmol,0.10 eq.) and Ruphos (3.13 g,6.708mmol,0.10 eq.). The resulting solution was stirred overnight in an oil bath at 30 ℃. The reaction mixture was cooled to room temperature. The solid was filtered. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. The final product was 10.7g (52.85%) of methyl 6-amino-3-bromo-5- (2-ethoxyvinyl) pyridine-2-carboxylate as a brown oil. 1 H NMR (300 MHz, chloroform-d, ppm) delta 8.10 (s, 0.5H), 7.57 (s, 0.5H), 6.90 (d, j=12.6 hz, 0.5H), 6.41 (d, j=7.1 hz, 0.5H), 3.97 (s, 3H).
5-bromo-1H-pyrrolo [2,3-b]Synthesis of pyridine-6-carboxylic acid methyl ester: to a round bottom flask was added 6-amino-3-bromo-5- (2-ethoxyvinyl) pyridine-2-carboxylic acid methyl ester (10.70 g,35.532mmol,1 eq.), methanol (100.00 mL) and concentrated hydrochloric acid (20.00 mL,12 n). The resulting solution was stirred overnight at 25 ℃. The resulting mixture was concentrated in a vacuum environment. The pH of the solution was adjusted to 7 with NaOH (2 mol/L). The solid was collected by filtration. The final product was 9g of 5-bromo-1H-pyrrolo [2,3-b ]Pyridine-6-carboxylic acid methyl ester as brown solid (99.30%). 1 H NMR (300 MHz, chloroform-d, ppm) δ11.60 (s, 1H), 8.29 (s, 1H), 7.69 (t, j=3.0 hz, 1H), 6.54 (dd, j=3.4, 1.7hz, 1H), 4.10 (s, 3H).
5-bromo-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]Pyrrolo [2,3-b]Synthesis of pyridine-6-carboxylic acid methyl ester: to a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere) was added 5-bromo-1H-pyrrolo [2,3-b]Pyridine-6-carboxylic acid methyl ester (9.00 g,35.284mmol,1 eq.) and DMF (100.00 mL). Subsequently NaH (2.83 g,70.757mmol,2.01 eq., 60%) was added in portions at 0deg.C. The resulting solution was stirred at 0℃for 30min. SEM-Cl (8.82 g,52.903mmol,1.50 eq.) was added to the mixture at 0deg.C. The resulting solution was stirred at 25℃for 2h, then 500mL of water was added to quench the reaction. The resulting solution was extracted with 2X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. The final product was 8.2g (60.31%) 5-bromo-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]Pyrrolo [2,3-b ]Pyridine-6-carboxylic acid methyl ester as yellow oil. 1 H NMR (300 MHz, chloroform-d, ppm) delta 8.17 (s, 1H), 7.50 (d, j=3.7 hz, 1H), 6.53 (d, j=3.6 hz, 1H), 5.68 (s, 2H), 4.02 (s, 3H), 3.60-3.45 (m, 2H), 0.99-0.88 (m, 2H), 0.05 (s, 9H).
(5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methanol synthesis: into a round bottom flask was added 5-bromo-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]Pyrrolo [2,3-b]Pyridine-6-carboxylic acid methyl ester (5.00 g,12.976mmol,1 eq.) and THF (50.00 mL). LAH (990.00 mg,26.084mmol,2.01 eq.) was then added in portions at 0 ℃. The resulting solution was stirred at 25℃for 2h. Subsequent addition of Na 2 SO 4 ·10H 2 O quench reaction. The solid was filtered. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. The final product was 1.7g (36.67%) (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methanol as a pale yellow oil. 1 H NMR (300 MHz, chloroform-d, ppm) delta 8.10 (s, 1H), 7.36 (d, j=3.6 hz, 1H), 6.51 (d, j=3.6 hz, 1H), 5.68 (s, 2H), 4.85 (s, 2H), 3.63-3.50 (m, 2H), 0.98-0.88 (m, 2H), 0.05 (s, 9H).
2- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methoxy]Synthesis of ethyl acetate: to a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere) was added (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methanol (1.70 g,4.758mmol,1 eq.) and THF (20.00 mL). Subsequently NaH (669.00 mg,16.727mmol,3.52 eq., 60%) was added in portions at 0deg.C. The resulting solution was stirred at 0℃for 30min. To the mixture was added ethyl bromoacetate (2.38 g,14.251mmol,3 eq.) at 0deg.C. The resulting solution was stirred overnight in an oil bath at 70 ℃. The reaction mixture was cooled to room temperature. 200mLNH was then added 4 The reaction was stopped with Cl aqueous solution. The resulting solution was extracted with 2X 100mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. Residual ofThe material was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. The final product was 1.7g (80.58%) 2- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methoxy]Ethyl acetate as a pale yellow oil. 1 H NMR (300 MHz, chloroform-d, ppm) delta 8.10 (s, 1H), 7.39 (d, j=3.6 hz, 1H), 6.48 (d, j=3.6 hz, 1H), 5.68 (s, 2H), 4.99 (s, 2H), 4.28 (s, 2H), 4.23 (t, j=7.1 hz, 2H), 3.58-3.51 (m, 2H), 1.34-1.27 (m, 3H), 0.95-0.88 (m, 2H), 0.05 (s, 9H).
2- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methoxy]Synthesis of acetamide: 2- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) was charged to a pressure tank reactor]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methoxy]Ethyl acetate (1.70 g,3.834mmol,1 eq.) and NH 3 Methanol solution (20.00 mL). The resulting solution was stirred overnight in an oil bath at 60 ℃. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. Crude product from PE: EA is shown as 5: 1. The solid was collected by filtration. The final product was 1.2g (75.53%) 2- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methoxy]Acetamide as white solid. 1 H NMR (300 MHz, chloroform-d, ppm) delta 8.13 (s, 1H), 7.40 (d, j=3.6 hz, 1H), 6.50 (d, j=3.6 hz, 1H), 5.65 (s, 2H), 4.95 (s, 2H), 4.19 (s, 2H), 3.64-3.46 (m, 2H), 1.03-0.85 (m, 2H), -0.04 (s, 9H).
4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-synthesis of tetradec-1 (9), 2,5, 7-tetraen-11-one: to a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere) was added 2- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) methoxy]Acetamide (1.20 g,2.896mmol,1 eq.), dioxane (20.00 mL), methyl [2- (methylamino) ethyl]Amine (256.00 mg, 2.906 mmol,1 eq.) CuI (553.00 mg, 2.906 mmol,1 eq.) and K 2 CO 3 (1.20 g,8.683mmol,3 eq.). The resulting solution was stirred overnight in an oil bath at 100 ℃. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by column chromatography on silica gel with ethyl acetate/petroleum ether(1:3) treatment. The final product was 527mg (54.57%) of 4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-11-one as white solid. 1 H NMR (300 MHz, chloroform-d, ppm) delta 8.65 (br, 1H), 7.55 (s, 1H), 7.38 (d, j=3.6 hz, 1H), 6.49 (d, j=3.6 hz, 1H), 5.65 (s, 2H), 5.00 (s, 2H), 4.64 (s, 2H), 3.66-3.45 (m, 2H), 0.98-0.87 (m, 2H), -0.04 (s, 9H).
4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-synthesis of tetradec-1 (9), 2,5, 7-tetraene: 4- [ [2- (trimethylsilyl) ethoxy ] was added to a round bottom flask]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Fourteen-1 (9), 2,5, 7-tetraen-11-one (527.00 mg,1.580mmol,1 eq.), THF (10.00 mL) and LAH (150.00 mg,3.952mmol,2.50 eq.). The resulting solution was stirred at 25℃for 3h. Then Na is added 2 SO 4 ·10H 2 O quench reaction. The solid was filtered. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. The final product was 350mg (69.32%) of 4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraene yellow oil. LC-MS (ES, M/z) m+1=320.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-13-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ],]]tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: to a vial (the vial was purged and stored in a nitrogen inert atmosphere) was added 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl) benzoate (1.16 g,2.181mmol,1.99 eq), toluene (20.00 mL), 4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]-tetradec-1 (9), 2,5, 7-tetraene (350.00 mg,1.096mmol,1 eq.) Cs 2 CO 3 (1.07G, 3.284mmol,3 eq.) and Xanphos Pd 2G (146.00 mg,0.165mmol,0.15 eq.). The resulting solution was stirred in an oil bath at 110℃for 2 days. The reaction mixture was cooled to room temperature. The obtained mixture is trueConcentrating under the air. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. The crude product was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): chromatographic column, C18 reverse phase chromatographic column; mobile phase, water (0.01% TFA) and CH 3 A CN; gradient: 20% CH 3 CN increased to 80% in 15 min; flow rate: 80mL/min; the detector comprises: 254/220nm. The final product was 150mg (17.77%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate as yellow oil. LC-MS (ES, M/z): m+1=770.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] fourteen-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid: to a round bottom flask was added methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (150.00 mg,0.195mmol,1 eq), methanol (5.00 mL), dioxane (5.00 mL), naOH (1.00 mL,4.000mmol,20.55 eq). The resulting solution was stirred in an oil bath at 70℃for 4h. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The pH of the solution was adjusted to 6-7 with HCl (2 mol/L). The resulting solution was extracted with 3X 50mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 200mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. The final product was 100mg (67.90%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid as a pale yellow oil. LC-MS (ES, M/z): M+1=756.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -13-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 ] [3,7] ] -tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide: to a vial was added 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] fourteen-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid (100.00 mg,0.132mmol,1 eq), DCM (5.00 mL), 4- [ [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonamide (42.00 mg,0.132mmol,1 eq), EDCI (51.00 mg, 0.268 mmol,2.01 eq) and DMAP (64.00 mg,0.524mmol,3.96 eq). The resulting solution was stirred overnight in an oil bath at 30 ℃. The resulting mixture was concentrated under vacuum. The residue was applied to a silica gel column (dichloromethane: ethyl acetate=1:1). The final product was 50mg (35.83%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] -tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide as a yellow solid.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ 13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide hydrochloride: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a vial]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Fourteen-1 (9), 2,5, 7-tetraen-10-yl) benzamide (50.00 mg,0.047mmol,1 eq.), 1.0M TBAF/THF (5.00 mL) and ethylenediamine (100.00 mg, 1.264 mmol,35.13 eq.). The resulting solution was stirred in an oil bath at 70℃for 3.5h. The reaction mixture was cooled to room temperature. The resulting solution was treated with 100mLH 2 O extraction. The resulting solution was treated with 3X 30mL of dimethylformamideAlkane extraction and combining organic layers. The resulting mixture was washed with 1X 200mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was purified by preparative thin layer chromatography (dichloromethane: ethyl acetate=1:1) (including 10% methanol). The crude product was purified by high performance liquid chromatography under the following conditions: chromatographic column, sunFire Prep C18,19×150 mm,5um; mobile phase, water (0.1% fa) and CH 3 A CN; gradient: 20% increases to up to 85% within 15 min; flow rate: 15mL/min; the detector comprises: 254/220nm. The collected solution was concentrated under vacuum to remove CH 3 CN, the resulting solution was dried by lyophilization (during which time an appropriate amount of concentrated hydrochloric acid was added). The final product was 4.5mg (9.88%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexen-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ 13-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide hydrochloride as a yellow solid. LC-MS (ES, M/z) m+1=925. 1 H NMR(300MHz,DMSO-d 6 ,ppm)δ11.80(s,1H),11.22(s,1H),10.71(d,J=10.0Hz,1H),8.62(t,J=5.2Hz,1H),8.32(d,J=2.0Hz,1H),7.55–7.21(m,5H),7.14(d,J=7.9Hz,2H),6.98–6.58(m,4H),6.13(s,1H),4.76(s,2H),4.03–3.46(m,17H),3.13(d,J=30.4Hz,2H),2.78(q,J=11.2,10.0Hz,2H),2.40(s,2H),2.05(s,2H),1.46(q,J=6.3Hz,2H),1.23(s,2H),0.97(s,6H)。
Examples 12 to 13
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3S or 3R) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] fourteen-1 (9), 2,5, 7-tetraen-10-yl ] benzamide and 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3R or 3S) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] -2- [ 14-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 [3,7] ] tetradec 1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of methyl 4- (4- [ [2- (4-chlorophenyl) -3, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (12R) -12-methyl-13-oxa-2, 4, 10-triazacyclo [7.4.0.0 [3,7] ] tride-1 (9), 2,5, 7-tetraen-10-yl ] -N- (3S) -5-nitro-3- (oxa-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] benzamide hydrochloride: to a vial was added 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13R) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid (25.00 mg,0.033mmol,1 eq.), DCM (10.00 mL), 4- [ [ [ (2S) -3, 4-dioxan-2-ylmethyl ] amino ] -7-nitrobenzenesulfonamide (11.35 mg,0.033mmol,1 eq.), EDCI (12.67 mg,0.066mmol,2 eq.) and DMAP (12.11 mg,0.099mmol,3 eq.). The resulting solution was stirred in an oil bath at 35 ℃ for 14h. The resulting mixture was concentrated. The residue was purified by Prep-TLC with dichloromethane/methanol (10:1). The final product was 22mg (61.53%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3S) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 [3,7] ] tetradec 1 (9), 2,5, 7-tetraen-10-yl) benzamide as a yellow solid. LC-MS (ES, m/z): 1081[ M+H ].
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ (3S) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a vial]Methyl group]Piperazin-1-yl) -N- [ (3S) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoatesAmide (20.00 mg,0.018mmol,1 eq.), ethylenediamine (0.20 mL) and TBAF in THF (1 mL, 2M). The resulting solution was stirred in an oil bath at 70 ℃ for 48h. The resulting mixture was concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane: methanol=10:1). The crude product was purified by high performance liquid chromatography under the following conditions: chromatographic column, X Bridge Prep C18 OBD 19X 150mm 5 μm; mobile phase, a:0.1% ammonia water; b: ACN; gradient: 6-85% B in 7.9 min; flow rate: 20mL/min; detector, UV220nm. The final product was 6mg (34.11%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- [ (3S) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide was a yellow solid. LC-MS (ES, m/z): 951.4[ M+H ]] 1 H NMR(300MHz,CDCl 3 ,ppm):δ12.22(s,1H),8.73(s,1H),8.47(d,J=2.1Hz,1H),8.37(s,1H),8.00(d,J=9.6Hz,1H),7.50(s,1H),7.27(s,1H),7.18(s,1H),7.07(s,1H),7.02–6.98(m,2H),6.76–6.72(m,2H),6.25(s,1H),4.40(s,1H),4.10–3.90(m,4H),3.78-3.20(m,9H),2.85(s,2H),2.45–2.14(m,6H),2.03(s,2H),1.90–1.32(m,10H),0.96(s,6H)。
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3R) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] -tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide: to a vial was added 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] fourteen-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid (25.00 mg,0.033mmol,1 eq), (3R) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazine-7-sulfonamide (11.35 mg,0.033mmol,1 eq), EDCI (12.67 mg,0.066mmol,2 eq.), DMAP (12.11 mg,0.099mmol,3 eq) and DCM (1.00 mL). The resulting solution was stirred in an oil bath at 35 ℃ for 14h. The resulting mixture was concentrated. The residue was purified by Prep-TLC with dichloromethane/methanol (10:1). The final product was 20mg (55.94%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3R) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 [3,7] ] tetradec 1 (9), 2,5, 7-tetraen-10-yl) benzamide as a yellow solid. LC-MS (ES, m/z): 1081[ M+H ].
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ (3R) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a vial]Methyl group]Piperazin-1-yl) -N- [ (3R) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide (18.00 mg,0.018mmol,1 eq.), ethylenediamine (0.20 mL) and TBAF in THF (1 mL, 2M). The resulting solution was stirred in an oil bath at 70 ℃ for 48h. The resulting mixture was concentrated. The residue was purified by preparative thin layer chromatography (dichloromethane: methanol=10:1). The crude product was purified by high performance liquid chromatography under the following conditions: chromatographic column, X Bridge Prep C18 OBD 19X 150mm 5 μm; mobile phase, a:0.1% ammonia water; b: ACN; gradient: 6-80% B in 7.9 min; flow rate: 20mL/min; detector, UV220nm. The final product was 7mg (44.21%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- [ (3R) -5-nitro-3- (tetrahydropyran-4-yl) -3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide was a yellow solid. LC-MS-PH-PHNW-4-141-0: (ES, m/z): 951.4[ M+H ]] 1 H NMR-PH-PHNW-4-141-0(300MHz,CDCl 3 ,ppm):δ12.23(s,1H),8.68(s,1H),8.48(d,J=2.1Hz,1H),8.37(s,1H),8.00(d,J=9.6Hz,1H),7.51(s,1H),7.27(s,1H),7.17(s,1H),7.07(s,1H),7.02–6.98(m,z,2H),6.77-6.71(m,2H),6.25(s,1H),4.40(s,1H),4.10–3.90(m,4H),3.78-3.20(m,9H),2.45–2.13(m,6H),2.03(s,2H),1.89–1.37(m,10H),0.98(s,6H)。
Examples 14 to 15
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3S or 3R) -3- [ (2S) -1, 4-dioxan-2-yl ] -5-nitro-3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] fourteen-1 (9), 2,5, 7-tetraen-10-yl ] benzamide and 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexen-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [ (3R or 3S) -3- [ (2S) -1, 4-dioxan-2-yl ] -5-nitro-3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl ] -2- [ 14-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 [3,7] ] tetradec 1 (9), 2,5, 7-tetraen-10-yl ] benzamide.
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
Synthesis of (2R) -1, 4-dioxane-2-yl) formaldehyde: to a round bottom flask was added (2S) -1, 4-dioxan-2-ylmethanol (30.00 g,253.953mmol,1 eq.) CH 3 CN (250.00 mL) and IBX (120.89 g,431.724mmol,1.70 eq.). The resulting solution was stirred in an oil bath at 70℃for 4h. The solid was filtered. The resulting mixture was concentrated. The final product was 30g (crude) (2R) -1, 4-dioxane-2-carbaldehyde as a colorless oil. H-NMR-1: 1 H NMR(300MHz,CDCl 3 ,ppm)δ9.66(s,1H),4.12-3.77(m,7H)。
synthesis of 2-amino-2- [ (2S) -1, 4-dioxan-2-yl ] acetonitrile: to a pressure tank reactor were added (2R) -1, 4-dioxane-2-carbaldehyde (30.00 g,258.362mmol,1.00 eq.) and ammonia/methanol (7M) (300.00 mL). TMSCN (38.40 g,387.071mmol,1.50 eq.) was then added. The resulting solution was stirred overnight in an oil bath at 70 ℃. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (3:1) on a silica gel column. The collected fractions were combined and concentrated. The final product was 18g (49.01%) of (2S) -2-amino-2- [ (2S) -1, 4-dioxan-2-yl ] acetonitrile as a yellow oil. LCMS-2 (ES, m/z): m+1:143.
Synthesis of((2S) -1, 4-Dioxahexacyclic-2-yl) acetic acid: in a round bottom flask was placed 2-amino-2- [ (2S) -1, 4-dioxan-2-yl ] acetonitrile (18.00 g,126.620mmol,1 eq.) NaOH (4M, 40.00 mL). The resulting solution was stirred overnight in an oil bath at 70 ℃. The pH of the solution was adjusted to 6 with AcOH. The solid was collected by filtration. The solid was dried in an oven under reduced pressure. The final product was 8.8g (43.13%) amino ((2S) -1, 4-dioxan-2-yl) acetic acid as a white solid, LCMS-3 (ES, m/z): m+1:162.
2-amino-2- [ (2S) -1, 4-dioxan-2-yl]Synthesis of ethanol: to a round bottom flask was added amino ((2S) -1, 4-dioxan-2-yl) acetic acid (8.80 g,54.605mmol,1 eq.) and THF (120.00 mL,1.664mmol,0.03 eq.). LiAlH is then added in portions at 0 ℃ 4 (6.22 g,163.883mmol,3 eq.). The resulting solution was stirred at 60℃in an oil bath for 6h. Then 12g Na was added 2 SO 4 ·10H 2 O quench reaction. The solid was filtered. The resulting mixture was concentrated. The final product was 4.5g (55.99%) 2-amino-2- [ (2S) -1, 4-dioxan-2-yl]Ethanol was a white solid. LCMS-4 (ES, m/z): m+1:148.
3-bromo-4- ([ 1- [ (2S) -1, 4-dioxan-2-yl)]-2-hydroxyethyl group]Synthesis of amino) -5-nitrobenzenesulfonamide: to a round bottom flask was added 3-bromo-4-chloro-5-nitrobenzenesulfonamide (4.00 g,12.677mmol,1 eq.) CH 3 CN (120.00 mL), DIEA (6.55 g,0.051mmol,4 eq.) and 2-amino-2- [ (2S) -1, 4-dioxan-2-yl]Ethanol (4.48 g,0.030mmol,2.4 eq.). The resulting solution was stirred in an oil bath at 80℃for 48h. The resulting mixture was concentrated. The residue was loaded onto a silica gel column and eluted with ethyl acetate/petroleum ether (9:1). The collected fractions were combined and concentrated. 3.2g (59.22%) of 3-bromo-4- ([ 1- [ (2S) -1, 4-dioxane-2-yl) are obtained ]-2-hydroxyethyl group]Amino) -5-nitrobenzenesulfonamide as a yellow solid. LCMS-5 (ES, m/z): m-1:424.
(3S or 3R) -3- [ (2S) -1, 4-dioxan-2-yl]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazine-7-sulfonamide (3R or 3S) -3- [ (2S) -1, 4-dioxan-2-yl]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazine-synthesis of 7-sulfonamide: to a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere) was added 3-bromo-4- ([ 1- [ (2S) -1, 4-dioxan-2-yl)]-2-hydroxyethyl group]Amino) -5-nitrobenzenesulfonamide (1.00 g,2.346mmol,1 eq.), dioxane (40.00 mL), cs 2 CO 3 (1911.01 mg,5.865mmol,2.50 eq.) and t-BuXPhos Pd G3 (186.05 mg,0.235mmol,0.10 eq.). The resulting solution was stirred in an oil bath at 100℃for 8h. Then 40mL of water was added to stop the reaction. The resulting solution was extracted with 3X 40mL of ethyl acetate, dried over anhydrous sodium sulfate and concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:1) on a silica gel column. The collected fractions were combined and concentrated. The crude product was purified by Prep-HPLC under the following conditions: column, X-bridge RP18; mobile phase, 0.05% ammonia and CH 3 CN(45%CH 3 CN reached 60% in 5 min); detector, UV 254nm. The final product was 20mg (2.47%) (3S or 3R) -3- [ (2S) -1, 4-dioxan-2-yl ]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazine-7-sulfonamide yellow solid and 40mg (4.94%) (3R or 3S) -3- [ (2S) -1, 4-dioxan-2-yl]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazine-7-sulfonamide as a yellow solid. LC-MS-6: (ES, M/z): M-1:344.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ (3R or 3S) -3- [ (2S) -1, 4-dioxan-2-yl]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a vial]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (18.13 mg,0.029mmol,1 eq), (3S or 3R) -3- [ (2S) -1, 4-dioxan-2-yl]5-nitro-3, 4-dihydro-2H-1, 4-benzoxazine-7-sulfonamide (10.00 mg,0.029mmol,1 eq.), DCM (3.00 mL), EDCI (11.10 mg,0.058mmol,2 eq.) and DMAP (14.15 mg,0.116mmol,4 eq.). The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The crude product was purified by Prep-HPLC under the following conditions: column, X-bridge RP18; mobile phase, 0.05% ammonia and CH 3 CN(45%CH 3 CN is in 5minInner up to 60%); detector, UV 254nm. The final product was 3.2mg (11.59%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ (3S or 3R) -3- [ (2S) -1, 4-dioxan-2-yl]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide was a yellow solid. LC-MS-0: (ES, M/z): M+1:953. 1 H NMR (300 MHz, chloroform-d, ppm) delta 12.09 (d, j=26.1 hz, 1H), 8.61 (s, 1H), 8.50 (s, 1H), 8.23 (s, 1H), 7.99 (d, j=8.4 hz, 1H), 7.63 (s, 1H), 7.35 (s, 2H), 7.16 (s, 1H), 7.00 (d, j=8.4 hz, 3H), 6.74 (d, j=8.1 hz, 2H), 6.25 (d, j=15.9 hz, 1H), 4.38 (s, 1H), 4.00-3.37 (m, 14H), 3.31 (s, 2H), 2.86 (s, 1H), 2.63 (s, 2H), 2.32 (d, j=35.6 hz, 5H), 2.08 (d, j=25.8 hz, 2H), 1.48 (s, 1.28, 1H), 1.01 (s, 1H).
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ (3S or 3R) -3- [ (2S) -1, 4-dioxan-2-yl]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a vial ]Methyl group]Piperazin-1-yl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (18.13 mg,0.029mmol,1 eq), (3S or 3R) -3- [ (2S) -1, 4-dioxan-2-yl]5-nitro-3, 4-dihydro-2H-1, 4-benzoxazine-7-sulfonamide (10.00 mg,0.029mmol,1 eq.), DCM (3.00 mL), EDCI (11.10 mg,0.058mmol,2 eq.) and DMAP (14.15 mg,0.116mmol,4 eq.). The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The crude product was purified by Prep-HPLC under the following conditions: column, X-bridge RP18; mobile phase, 0.05% ammonia and CH 3 CN(45%CH 3 CN reached 60% in 5 min); detector, UV 254nm. The final product was 3.1mg (11.23%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [ (3R or 3S) -3- [ (2S) -1, 4-dioxan-2-yl]-5-nitro-3, 4-dihydro-2H-1, 4-benzoxazin-7-ylsulfonyl]-2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide yellowColor solids. LC-MS-0: (ES, M/z): M+1:953. 1 H NMR (300 MHz, chloroform-d, ppm) delta 12.05 (d, j=29.4 hz, 1H), 8.82 (s, 1H), 8.49 (s, 2H), 7.98 (d, j=9.0 hz, 1H), 7.58 (s, 1H), 7.36 (s, 1H), 7.17 (s, 1H), 7.00 (d, j=7.5 hz, 3H), 6.74 (d, j=8.1 hz, 2H), 6.25 (d, j=14.4 hz, 1H), 4.15-3.44 (m, 16H), 3.31 (s, 2H), 2.86 (s, 1H), 2.64 (s, 2H), 2.32 (d, j=37.5 hz, 4H), 2.08 (d, j=25.3 hz, 2H), 1.50 (s, 4H), 1.28 (s, 1H), 1.01 (s, 6H).
Example 16
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
(R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) was added to a round bottom flask]-2-yl) methyl-piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepin-1 (7H) -yl]A solution of benzamide (15 mg,0.015mmol,1 eq.) in methanol (5 mL). The resulting solution was purified (Shimadzu LC-20 AT) under the following conditions: chromatographic column, CHIRALPAK ID-3,4.6 x 50mm,3 μm; mobile phase a: n-hexane/dcm=5/1; and B phase: ethanol/methanol=1/1; a detector, a PDA. The final product was 2.5mg (33%) (R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) ]-2-yl) methyl-piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepin-1 (7H) -yl]Benzamide was a yellow solid. LC-MS (ES, M/z): M+1=991, RT=1.61 min. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0Agilent Poroshell HPH-C18,2.7 μm; elutionLiquid A: water (0.05% ammonia); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min. 1 H NMR(300MHz,CDCL3,ppm)δ11.43(ds,1H),9.99(ds,1H),8.72(s,1H),8.40(ds,1H),7.93-7.85(m,2H),7.37-7.35(m,2H),7.26(s,1H),7.02-6.93(m,3H),6.71-6..64(m,3H),6.27(s,1H),4.58(s,1H),4.05-3.15(m,23H),2.70-2.35(m,7H),2.03(s,3H),1.92-1.90(m,1H),1.89–1.87(m,2H),1.71-1.67(m,2H),1.53-1.45(m,3H),1.00(s,6H)。
Example 17
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl) was added to a round bottom flask]-2-yl) methyl-piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepin-1 (7H) -yl]A solution of benzamide (15 mg,0.015mmol,1 eq.) in methanol (5 mL). The resulting solution was purified (Shimadzu LC-20 AT) under the following conditions: chromatographic column, CHIRALPAK ID-3,4.6 x 50mm,3 μm; mobile phase a: n-hexane/dcm=5/1; and B phase: ethanol/methanol=1/1; a detector, a PDA. The final product was 2.5mg (33%) (S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl)]-2-yl) methyl-piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2',5,6]Pyrido [2,3-b][1,4]Oxazepin-1 (7H) -yl]Benzamide was a yellow solid. LC-MS (ES, M/z): M+1=991, RT=1.61 min. The residence time was measured using a reverse phase column (C18 Is completed. Shimadzu LCMS 2020;50*3.0Agilent Poroshell HPH-C18,2.7 μm; eluent a: water (0.05% ammonia); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min. 1 H NMR(300MHz,CDCL3,ppm)δ11.43(ds,1H),9.99(ds,1H),8.72(s,1H),8.40(ds,1H),7.93-7.85(m,2H),7.37-7.35(m,2H),7.26(s,1H),7.02-6.93(m,3H),6.71-6..64(m,3H),6.27(s,1H),4.58(s,1H),4.05-3.15(m,23H),2.70-2.35(m,7H),2.03(s,3H),1.92-1.90(m,1H),1.89–1.87(m,2H),1.71-1.67(m,2H),1.53-1.45(m,3H),1.00(s,6H)。
Example 18
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [13, 13-difluoro-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] -N- (4- [ [ (2R) -1, 4-dioxa-2-ylmethyl ] amino ] -2-nitrobenzenesulfonyl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of 1-bromo-3, 3-diethoxy-1, 1-difluoropropane to a sealed tube was added vinyl diethyl ether (14.40 g,199.703mmol,1 eq.), ethanol (80.00 mL), difluoromethane (62.85 g,0.300mmol,1.5 eq.), na 2 S 2 O 4 (52.15 g,0.300mmol,1.5 eq.) and NaHCO 3 (50.33 g,0.599mmol,3 eq.). The resulting solution was stirred in an oil bath at 60℃for 4h. The reaction was then stopped by adding 200mL of water/ice. The resulting solution was extracted 3 times with 200mL petroleum ether. The resulting mixture was washed 2 times with 100mL of water. The mixture was dried over anhydrous sodium sulfate. The solid was filtered. The resulting mixture was concentrated under vacuum. The final product was 12g (24.32%) 1-bromo-3, 3-diethoxy-1, 1-difluoropropane as a yellow oil. 1 H NMR(300MHz,DMSO-d 6 ):δ4.82(t,J=5.2Hz,1H),3.7-3.55(m,2H),3.54-3.43(m,2H),2.83(td,J=14.7,5.1Hz,2H),1.13(t,J=7.2Hz,6H)。
Synthesis of ethyl 3-bromo-3, 3-difluoropropionate: into a round bottom flask was charged 1-bromo-3, 3-diethoxy-1, 1-difluoropropane (12.00 g,48.567mmol,1 eq.), DCM (120.00 mL), m-CPBA (11.73 g,0.068mmol,1.4 eq.) and H 2 SO 4 (0.10 g,0.001mmol,0.02 eq.). The resulting solution was stirred overnight in an oil bath at 55 ℃. The solid was filtered. With NaHCO 3 (5 mol/L) the pH of the solution was adjusted to 8. The resulting solution was extracted with 2X 120mL of dichloromethane. The extract solution was concentrated under vacuum. The final product was 8g (75.90%) ethyl 3-bromo-3, 3-difluoropropionate as a pale yellow oil.
Synthesis of 5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-ol: to a round bottom flask was added 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridine (3.00 g,8.688mmol,1 eq.), NMP (50.00 mL), water (5.00 mL), and KOH (1.46 g,0.026mmol,3 eq.). The resulting solution was stirred in an oil bath at 135℃for 3h. Then 50mL of water was added to stop the reaction. The resulting solution was extracted 4 times with 50mL ethyl acetate and concentrated in vacuo. The residue was eluted with ethyl acetate/petroleum ether (1:5) on a silica gel column. The collected fractions were combined and concentrated. The final product was 1.5g (50.29%) of 5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-ol as a white solid. LCMS (ES, M/z): M+1:343/345.
Synthesis of ethyl 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -3, 3-difluoropropionate: to a round bottom flask was added 5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-ol (1.02 g,2.971mmol,1 eq.), DMF (10.00 mL), TEA (0.75 g, 0.0070 mmol,2.5 eq.) and ethyl 3-bromo-3, 3-difluoropropionate (0.64 g,0.003mmol,1 eq.). The resulting solution was stirred at 0 ℃ to room temperature overnight. Then 20mL of water was added to stop the reaction. The resulting solution was extracted 3 times with 20mL ethyl acetate and concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:10) on a silica gel column. The collected fractions were combined and concentrated. The final product was 1.6g (112.33%) ethyl 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -3, 3-difluoropropionate as a yellow oil. LC-MS (ES, M/z): M+1:479/481.
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl) oxy]-synthesis of 3, 3-difluoropropan-1-ol: to a three-necked flask (the flask was purged and kept in a nitrogen inert atmosphere) was added 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl) oxy]-ethyl 3, 3-difluoropropionate (1.60 g,3.338mmol,1 eq.), THF (20.00 mL) and LiAlH 4 (0.51 g,0.013mmol,4 eq.). The resulting solution was stirred at-78℃to-30℃for 2h. The reaction was quenched by the addition of 20mL of water. The solid was filtered. The resulting solution was extracted 3 times with 20mL ethyl acetate and concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:4) on a silica gel column. The collected fractions were combined and concentrated in vacuo. The final product was 900mg (61.66%) 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrolo [2,3-b]Pyridin-6-yl) oxy]-3, 3-difluoropropan-1-ol as a pale yellow oil. LC-MS (ES, M/z): M+1:437/439.
Synthesis of 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -3, 3-difluoropropyl methanesulfonate: to a round bottom flask was added 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -3, 3-difluoropropan-1-ol (900.00 mg,2.058mmol,1 eq), DCM (20.00 mL), TEA (624.69 mg,6.173mmol,3 eq) and methanesulfonyl chloride (259.27 mg,2.26mmol,1.10 eq). The resulting solution was stirred at 0 ℃ to room temperature for 3h. Then 20mL of water was added to stop the reaction. The resulting solution was extracted 3 times with 20mL ethyl acetate and concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:5) on a silica gel column. The collected fractions were combined and concentrated in vacuo. The final product was 1g (94.28%) of 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -3, 3-methanesulfonic acid difluoropropyl ester as a pale yellow oil. LC-MS (ES, M/z): M+1:515/517.
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]-synthesis of 3, 3-difluoropropan-1-amine: 3- [ (5-bromo-1- [ [2- (trimethyl)Silicon-based) ethoxy]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]3, 3-Difluoropropyl methane sulfonate (1.00 g,1.94mmol,1 eq.) was placed in a sealed tube and NH was introduced 3 MeOH (7M) (20.00 mL). The resulting solution was stirred overnight in an oil bath at 70 ℃. The resulting mixture was concentrated under vacuum. To obtain 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]600mg (70.87%) of 3, 3-difluoropropane-1-amine was a yellow oil. LC-MS (ES, M/z): M+1:436/438.
Synthesis of 13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraene: 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -3, 3-difluoropropan-1-amine (600.00 mg,1.375mmol,1 eq.), toluene (10.00 mL), t-Buona (396.42 mg,4.125mmol,3 eq.), XPhos Pd G3 precatalyst (116.38 mg,0.137mmol,0.10 eq.) are placed in a vial (the vial is purged and stored in a nitrogen inert atmosphere). The resulting solution was stirred in an oil bath at 80℃for 2h. The resulting mixture was concentrated under vacuum. The residue was eluted with ethyl acetate/petroleum ether (1:3) on a silica gel column. The collected fractions were combined and concentrated. 13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraene pale 400mg (81.84%) as a yellow oil. LC-MS (ES, M/z): M+1:356.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: 13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen (400.00 mg,1.125mmol,1 eq.), toluene (30.00 mL), methyl 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl benzoate (838.00 mg,1.575mmol,1.40 eq.) Cs 2 CO 3 (1099.93 mg,3.376mmol,3 eq.) XantPhos Pd G2 precatalyst (65.11 mg,0.113 mmol)0.1 equivalent) was placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The resulting solution was stirred overnight in an oil bath at 110 ℃. The solid was filtered. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:3) on a silica gel column. The collected fractions were combined and concentrated. To obtain methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate 400mg (44.08%), yellow oil. LC-MS (ES, M/z): M+1:806.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid: methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (200.00 mg,0.248mmol,1 eq), dioxane (2.00 mL), methanol (2.00 mL), sodium hydroxide (0.40 mL, 1.600mmol, 6.45 eq) was placed in a vial. The resulting solution was stirred overnight in an oil bath at 70 ℃. The resulting mixture was concentrated. The pH of the solution was adjusted to 6 with hydrochloric acid (2 mol/L). The resulting solution was extracted with 3X 20mL ethyl acetate concentrated under vacuum. The residue was applied to a silica gel column and eluted with dichloromethane/methanol (10:1). The collected fractions were combined and concentrated. 100mg (508.85%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid is obtained as a white solid. LC-MS (ES, M/z): M+1:792.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) -N- (4- [ [ (2S) -1, 4-dioxa-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid (100.00 mg,0.126mmol,1 eq.), 4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonamide (40.04 mg,0.126mmol,1 eq.), DCM (20.00 mL), EDCI (48.38 mg,0.25mmol,2 eq.), DMAP (61.67 mg,0.505mmol,4 eq.) were placed in a round bottom flask. The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:1) on a silica gel column. The collected fractions were combined and concentrated in vacuo. 80mg (58.07%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetrafluoro-10-yl) -N- (4- [ (2S) -1, 4-dioxa-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) benzamide is obtained as a yellow solid. LC-MS (ES, M/z): M+1:1091.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [13, 13-difluoro-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]-N- (4- [ [ (2R) -1, 4-dioxan-2-ylmethyl]Amino group]-synthesis of 2-nitrobenzenesulfonyl) benzamide hydrochloride: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (13, 13-difluoro-4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) -N- (4- [ [ (2S) -1, 4-dioxa-2-ylmethyl]Amino group]3-Nitrophenesulfonyl) benzamide (50.00 mg,0.046mmol,1 eq), tetrahydrofuran (5.00 mL), TBAF (119.75 mg,0.458mmol,10 eq.) and ethylenediamine (27.52 mg,0.458mmol,10 eq.) were placed in a vial. The resulting solution was stirred overnight in an oil bath at 70 ℃. The resulting mixture was concentrated. The crude product is obtained by high performance liquid chromatographyPurifying under the condition: a chromatographic column, X-bridge RP18; mobile phase, 0.05% aqueous hydrochloric acid and CH 3 CN(45%CH 3 CN reached 60% in 5 min); detector, UV 254nm. To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [13, 13-difluoro-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]-N- (4- [ [ (2R) -1, 4-dioxan-2-ylmethyl]Amino group]15.1mg (33.04%) of 2-nitrobenzenesulfonyl) benzamide hydrochloride as a yellow solid. LC-MS (ES, M/z): M+1-HCl:961. 1 H NMR(300MHz,DMSO-d 6 )δ11.68(s,1H),11.43(s,1H),9.73(s,1H),8.60(t,J=5.7Hz,1H),8.40(d,J=2.4Hz,1H),7.62(dd,J=9.3,2.4Hz,1H),7.48–7.28(m,4H),7.24–7.07(m,3H),7.02(d,J=9.3Hz,1H),6.70–6.56(m,2H),6.22(dd,J=3.3,1.8Hz,1H),3.94–3.74(m,5H),3.71–3.58(m,4H),3.56–3.39(m,5H),3.31(s,2H),3.22(d,J=12.3Hz,2H),2.89–2.67(m,2H),2.35(d,J=36.3Hz,4H),2.06(s,2H),1.50(d,J=7.5Hz,2H),0.97(s,6H)。
example 19
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [4- [ [ (2R) -1, 4-dioxan-2-yl ] methyl ] amino) -3-nitrobenzenesulfonylbenzene ] -2- [ (13R) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) benzoate: in a round bottom flask, 1- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-enyl-1-yl ] methyl ] piperazine (15.09 g,47.32mmol,1 eq.) in DMA (150 mL), DIEA (12.9 g,99.81mmol,2 eq.) methyl 2-bromo-4-fluorobenzoate (11.6 g,49.78mmol,1 eq.) was placed. The resulting solution was stirred at 100℃for 12h. The reaction mixture was cooled to room temperature. Then 50mL of water was added to stop the reaction. The resulting solution was extracted with 3X 100mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 3X 100mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0:1-1:5) on a silica gel column. 7g of methyl (crude) 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) benzoate were obtained as a yellow oil. LC-MS (ES, M/z): M+1= 533,531.
4- ([ [ (2R) -1, 4-dioxan-2-yl)]Methyl group]Preparation of amino) -3-nitrobenzene-1-sulfonamide: 4-fluoro-3-nitrobenzene-1-sulfonamide (1.43 g, 0.0070 mmol,1 eq.) 1- [ (2R) -1, 4-dioxane-2-yl]Carboxamide hydrochloride (1 g,6.510mmol,1 eq.), THF (30 mL), cs 2 CO 3 (8.48 g,0.026mmol,4 eq.) in a round bottom flask. The resulting solution was stirred overnight in an oil bath at 50 ℃. The solid was collected by filtration. The solid was dried in an oven under reduced pressure. 1.8g (87.14%) of 4- ([ [ (2R) -1, 4-dioxane-2-yl) are obtained]Methyl group]Amino) -3-nitrobenzene-1-sulfonamide as a yellow solid. LC-MS (ES, M/z): M+1=318, RT=0.740 min.
Synthesis of N- (3-hydroxybutyl) -4-toluene-1-sulfonamide: 4-aminobutan-2-ol hydrochloride (4 g,31.847mmol,1 eq), DCM (40 mL), TEA (3.56 g,35.181mmol,1.10 eq) were placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). Then, 4-toluene-1-sulfonyl chloride (6.08 g,31.893mmol,1 eq.) was added at 0deg.C. The resulting solution was stirred at 25℃for 2h. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. 4.4g (56.78%) of N- (3-hydroxybutyl) -4-toluene-1-sulfonamide are obtained as a colorless oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ7.82–7.73(d,J=9.0Hz,2H),7.39–7.30(d,J=9.0Hz,2H),4.06–3.82(m,1H),3.24–3.16(m,1H),3.06–3.00(m,1H),2.45(s,3H),1.70–1.55(m,2H),1.20(d,J=6.2Hz,3H)。
N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]-synthesis of 4-toluene-1-sulfonamide: n- (3-hydroxybutyl) -4-toluene-1-sulfonamide (4.4 g, 18.083)mmol,1.10 eq), THF (60 mL) was placed in a three-necked round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). Subsequently, at 0deg.C, part of NaH (1.97 g,49.255mmol,3 eq., 60%) was added. The resulting solution was stirred at 0℃for 0.5h. To which 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] is added]Methyl group]-1H-pyrrole [2,3-b ]]Pyridine (5.66 g, 16.390 mmol,1 eq.). The resulting solution was stirred overnight in an oil bath at 50 ℃. The reaction mixture was cooled to 25 ℃. 500mL of NH was then added 4 The reaction was quenched by Cl. The resulting solution was extracted with 2X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. To obtain N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]6g (64.37%) of 4-toluene-1-sulfonamide as colorless oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.04(s,1H),7.74–7.63(m,2H),7.23–7.11(m,3H),6.41(d,J=3.6Hz,1H),5.56(d,J=3.3Hz,2H),5.89–5.33(m,1H),3.59–3.46(m,2H),3.17(t,J=6.0Hz,2H),2.37(s,3H),2.08–1.78(m,2H),1.37(d,J=6.2Hz,3H),0.94–0.85(m,2H),-0.06(s,9H)。
13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: n- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]-4-toluene-1-sulfonamide (6 g,10.552mmol,1 eq.) dimethyl sulfoxide (60 mL), pyridine-2-carboxylic acid (1.04 g, 8.4478 mmol,0.80 eq.), cuI (2.41 g, 12.254 mmol,1.20 eq.), K 2 CO 3 (4.38 g,31.692mmol,3 eq.) was placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The resulting solution was stirred in an oil bath at 120℃for 2 days. The reaction mixture was cooled to 25 ℃. The resulting solution was diluted with 500mL of water. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 2X 1000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo.The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. To obtain 13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradeca-1 (9), 2,5, 7-tetraene 2.8g (54.41%) as yellow oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.10(s,1H),7.53–7.40(m,2H),7.30(d,J=3.6Hz,1H),7.24–7.12(m,2H),6.50(d,J=3.6Hz,1H),5.65(d,J=10.7Hz,1H),5.49(d,J=10.7Hz,1H),4.37–4.23(m,1H),3.94–3.80(m,1H),3.62–3.36(m,3H),2.37(s,3H),1.89–1.64(m,2H),1.23(d,J=6.3Hz,3H),1.01–0.76(m,2H),-0.07(s,9H)。
13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: na (793.5 mg,34.51mmol,6.01 eq), naphthalene (4.42 g,34.48mmol,6.01 eq), dimethyl ether (20 mL) were placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The mixture was stirred at room temperature for 40min until sodium/naphthalene was completely formed. 13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene (2.8 g,5.741mmol,1 eq.) and tetrahydrofuran (20 mL) were placed in another round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The above solution was then added at-78 ℃. The resulting solution was stirred at room temperature for 2h. Then 500mLNH was added 4 Cl, stopping the reaction. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. Obtaining 13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene 1.2g (62.67%) as yellow oil. 1 H NMR(300MHz,DMSO-d 6 ,ppm)δ7.43–7.26(m,2H),6.27(d,J=3.5Hz,1H),5.43(d,J=3.4Hz,2H),5.13(s,1H),4.14–4.04(m,1H),3.58–3.39(m,3H),3.28–3.10(m,1H),2.95–2.77(m,1H),2.09–1.85(m,1H),1.85–1.64(m,1H),1.37(d,J=6.3Hz,3H),0.88–078(m,2H),-0.09(s,9H)。
13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: the crude product (0.5 g) was purified by high performance liquid chromatography HPLC: instrument name: SHIMADZU LC-20AD, liquid chromatography parameters: pump mode: binary gradient, pump starting concentration: 50.0% of total flow: 15mL/min, phase A: n-hexane (0.1% dea), phase B: ethanol, column name: CHIRALpak IA-3, length: 50mm, inside diameter: 4.6mm, particle size: 3.0 μm, column temperature: 25 ℃, PDA model: SPD-M20A, wavelength: 190nm to 500nm. Obtain (peak 1 presumes R) [ a ]]= -6.78 ° (in CH 2 Cl 2 (13R or S) -13-methyl-4-ethoxy [2- (trimethylsilyl) oxy (C=0.129 g/100mL, T=27℃)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetraene 220mg (yellow oil) and (peak 2 supposedly S) [ a ]]=+11.84°(CH 2 Cl 2 (13S or R) -13-methyl-4-ethoxy [2- (trimethylsilyl) oxy (C=0.106 g/100mL, T=27℃)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Tetradec-1 (9), 2,5, 7-tetraene 230mg (yellow oil).
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen (210.00 mg,0.630mmol,1 eq.), toluene (5.00 mL), 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl benzoate (669.87 mg, 1.319 mmol,2 eq.) Cs 2 CO 3 (1025.80 mg,3.148mmol,5 eq.) Xantphos Pd 2G precatalyst (111.70 mg,0.126mmol,0.20 eq.) is placed in a vial (the vial is purged and stored under nitrogen inert atmosphere). The resulting solution was stirred overnight in an oil bath at 110 ℃. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was applied to a silica gel columnTreatment with ethyl acetate/petroleum ether (1:1). To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]240mg (31.09%) of methyl benzoate as a yellow oil. LC-MS (ES, M/z): M+1=784.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoic acid: 4- (4- [ [ 2-4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate (200 mg,0.255mmol,1 eq.) H 2 O (1 mL), methanol (2 mL), dioxane (2 mL), sodium hydroxide (61.18 mg,1.530mmol,6 eq.) were placed in a round bottom flask. The resulting solution was stirred at 70℃for 12h and the resulting mixture was concentrated. The pH of the solution was adjusted to 5 with hydrochloric acid (1 mol/L). The solid was collected by filtration. 180mg of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (91.64%) as a white solid. H-NMR (CDCl 3,300 ppm): delta 8.17 (s, 1H), 7.28 (s, 1H), 7.00-6.79 (m, 4H), 6.35 (s, 1H), 5.70-5.67 (d, j=9 hz, 1H), 5.56-5.52 (d, j=12 hz, 1H), 4.38-4.35 (m, 1H), 3.81 (s, 1H), 3.58-3.56 (m, 4H), 3.26 (s, 3H), 2.84 (s, 1H), 2.35 (s, 3H), 2.24 (s, 2H), 2.12 (s, 2H), 2.04 (s, 2H), 1.60 (s, 6H), 1.28 (s, 1H), 1.01 (s, 7H), 0.98-0.95 (m, 3H), 0.00 (s, 9H).
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [4- ([ [ (2R) -1, 4-dioxan-2-yl ] methyl ] amino) -3-nitrobenzenesulfonyl ] -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzamide: 4- ([ [ (2R) -1, 4-dioxan-2-yl ] methyl ] amino) -3-nitrobenzene-1-sulfonamide (41.18 mg,0.130mmol,1 eq.), 4- (4- [ [2- (4-chlorophenyl) 4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid (100.00 mg,0.130mmol,1 eq.), DCM (3 mL), DMAP (63.42 mg,0.519mmol,4 eq.), EDCI (49.76 mg,0.260mmol,2 eq.) was placed in a round bottom flask. The resulting solution was stirred at room temperature for 12h. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:0) on a silica gel column. The resulting 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- [4- ([ [ (2R) -1, 4-dioxan-2-yl ] methyl ] amino) -3-nitrobenzenesulfonyl ] -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzamide 80mg (57.62%) as a yellow solid.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- [4- [ [ (2R) -1, 4-dioxan-2-yl]Methyl group]Amino) -3-nitrobenzenesulfonylenes]-2- [ (13R) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4-4- [ [2- (4-chlorophenyl) 4, 4-dimethylcyclohexyl-1-en-1-yl]Methyl group]Piperazin-1-yl) -N- [4- ([ [ (2R) -1, 4-dioxan-2-yl)]Methyl group]Amino) -3-nitrobenzenesulfonyl]-2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3.7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Benzamide (80.00 mg,0.075mmol,1 eq), tetrahydrofuran (5 mL), ethane-1, 2-diamine (89.89 mg,1496mmol,20 eq.) and TBAF (391.05 mg,1496mmol,20 eq.) were placed in a round bottom flask. The resulting solution was stirred at 70℃for 48h and the resulting mixture was concentrated. The crude product was purified by Prep-HPLC under the following conditions (Waters-2767): column, X-bridge RP18,5 μm, 19X 100mm; mobile phase, 0.03% ammonia (0.03% NH) 4 HCO 3 And NH 4 OH) and CH 3 CN(32%CH 3 CN up to 52% in 6 min); detector, UV 254nm. To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- [4- ([ [ (2R) -1, 4-dioxan-2-yl)]Methyl group]Amino) -3-nitrobenzenesulfonyl]-2- [ (13R) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Benzamide 25mg (35.58%) as a yellow solid. H-NMR (d-DMSO, 300M Hz,ppm): delta 8.87-8.86 (m, 1H), 8.69 (s, 1H), 8.47 (s, 1H), 8.00-7.92 (m, 2H), 7.13 (s, 1H), 7.00-6.93 (m, 2H), 6.75-6.72 (m, 2H), 6.60-6.50 (s, 1H), 6.19 (s, 1H), 3.92-3.67 (m, 7H), 3.45-3.31 (m, 8H), 2.85 (s, 2H), 2.38-2.25 (m, 6H), 2.09-2.03 (m, 3H), 1.71-1.69 (m, 3H), 1.50-1.46 (m, 2H), 1.06-0.99 (m, 6H).
Example 20
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (4-fluorooxalan-4-yl) methyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ (13S) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of methyl 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) benzoate: in a round bottom flask, 1- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-enyl-1-yl ] methyl ] piperazine (15.09 g,47.32mmol,1 eq.) in DMA (150 mL), DIEA (12.9 g,99.81mmol,2 eq.) methyl 2-bromo-4-fluorobenzoate (11.6 g,49.78mmol,1 eq.) was placed. The resulting solution was stirred at 100℃for 12h. The reaction mixture was cooled to room temperature. Then 50mL of water was added to stop the reaction. The resulting solution was extracted with 3X 100mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 3X 100mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (0:1-1:5) on a silica gel column. 7g of methyl (crude) 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) benzoate were obtained as a yellow oil. LC-MS (ES, M/z): M+1= 533,531.
1, 6-Dioxaspiro [2.5 ]]Synthesis of octane-2-carbonitrile: in a 3000mL round bottom flask was placed oxazolidin-4-one (204.2 g,2039.74mmol,1.1 eq), 2-chloroacetonitrile (140 g,1854.30mmol,1 eq), t-BuOH (200 mL). The resulting solution was stirred at 25℃for 30min. Then a solution of t-BuOK (249.7 g,2225.25mmol,1.20 eq.) was added dropwise in t-butanol (2000 mL) and stirred at 25℃for 40min. The resulting solution was stirred at 25 ℃ overnight. The resulting solution was diluted with 400mL of water and the reaction quenched with 80mL of 10% hydrogen chloride. The resulting mixture was concentrated to one third of its volume. The resulting solution was extracted with 3X 800mL diethyl ether and the organic layers were combined. The resulting mixture was washed with 3000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. Obtaining 1, 6-dioxaisothiocyanate [2.5 ]]Octane-2-carbonitrile 162g (62.78%) as a brown oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ3.95-3.81(m,4H),2.17-2.02(m,1H),1.99-1.77(m,2H),1.66-1.54(m,1H)。
Synthesis of 2- (4-fluorooxalan-4-yl) -2-hydroxyacetonitrile: 1, 6-Dioxaspiro [2.5 ]]Octane-2-carbonitrile (162 g,1164.18mmol,1 eq.) and DCM (800 mL) were placed in a 2000mL round bottom flask. Subsequently, 70% HF/Py (150 mL,0.95 eq.) was added dropwise at 0deg.C and stirred. The resulting solution was stirred in an oil bath at 40 ℃ overnight. The resulting solution was diluted with 1500mL ethyl acetate and poured into saturated NaHCO 3 In an aqueous solution. Using additional solid NaHCO 3 The mixture was slowly neutralized until foaming ceased. The resulting solution was extracted with 2X 1000mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 3000mL of 1% hydrogen chloride and 1X 3000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:2) on a silica gel column. 95g (51.27%) of 2- (4-fluorooxalan-4-yl) -2-hydroxyacetonitrile were obtained as a yellow solid. 1 H NMR(300MHz,CDCl 3- d,ppm)δ4.39(d,J=15.6Hz,1H),4.05-3.90(m,2H),3.83-3.70(m,2H),2.08–1.77(m,4H)。
Synthesis of (4-fluorooxalan-4-yl) methanol: 2- (4-Fluoroalk-4-yl) -2-hydroxyacetonitrile (95 g,596.88mmol,1 eq.), i-PrOH (1500 mL), H 2 O (375 mL) was placed in a 2000mL round bottom flask. Subsequently adding part of NaBH at 0 DEG C 4 (35 g,925.12mmol,1.55 eq.). The resulting solution was stirred at 25℃for 2h. The reaction was then quenched by the addition of 100mL of acetone and stirred for an additional 1h. The solid was filtered off. The solid was washed with EtOAc (200 mL). Concentrating and filtering under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. 66g (82.43%) of (4-fluorooxalan-4-yl) methanol are obtained as a yellow oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ3.90–3.70(m,2H),3.62(d,J=20.6Hz,1H),1.88–1.61(m,2H)。
Synthesis of methyl (4-fluorooxalan-4-yl) methylsulfonate: (4-fluorooxalan-4-yl) methanol (66 g,491.99mmol,1 eq), DCM (600 mL), TEA (74.7 g,737.98mmol,1.5 eq) were placed in a round bottom flask. MsCl (84.5 g,737.66mmol,1.50 eq.) was then added dropwise at 0deg.C and stirred. The resulting solution was stirred at 25℃for 2h. The resulting solution was diluted with 2000mL of water. The resulting solution was extracted with 2X 500mL of dichloromethane. The combined organic layers were dried over anhydrous sodium sulfate and concentrated under vacuum. 100g (95.77%) of methyl (4-fluoroalkan-4-yl) methylsulfonate were obtained as yellow oil. 1 H NMR(300MHz,DMSO)δ4.32(d,J=21.6Hz,1H),3.83–3.49(m,2H),1.92–1.62(m,2H)。
Synthesis of 4- (azidomethyl) -4-fluorooxazolidine: methyl (4-fluorooxalan-4-yl) methylsulfonate (100 g,471.16mmol,1 eq.), DMF (1000 mL), naHCO 3 (93.0 g,1107.06mmol,2.35 eq.) NaN 3 (90 g,1384.40mmol,2.94 eq.) is placed in a four-necked round bottom flask. The resulting solution was stirred overnight in an oil bath at 120 ℃. The reaction mixture was cooled to room temperature. Then 3000mL of water was added to stop the reaction. The resulting solution was extracted with 3X 500mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 3X 3000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. Obtaining 4- (azidomethyl) -4-fluorooxazolidine70g (93.34%) as yellow oil without further purification.
Synthesis of 1- (4-fluorooxalan-4-yl) carboxamide: 4- (azidomethyl) -4-fluorooxazolidine (70 g,439.80mmol,1 eq.) EtOAc (700 mL), 10% palladium on carbon (7 g,3472.43 mmol) was placed into a pressure tank reactor with a capacity of 2000 mL. The flask was evacuated, flushed three times with nitrogen, and then flushed with hydrogen (2-5 atmospheres). The resulting solution was stirred at 25 ℃ overnight. The solid was filtered off. The resulting mixture was concentrated under vacuum. 52g (crude) of 1- (4-fluorooxalan-4-yl) carboxamide are obtained as a yellow oil. 1 H NMR(300MHz,MeOD d 4 ,ppm)δ3.88–3.69(m,2H),2.76(d,J=20.5Hz,1H),1.85–1.68(m,2H)。
4- [ [ (4-fluorooxalan-4-yl) methyl]Amino group]-synthesis of 3-nitrobenzene-1-sulfonamide: 1- (4-Fluorooxalan-4-yl) methylamine (52.0 g,390.49mmol,1 eq.) tetrahydrofuran (1000 mL), 4-fluoro-3-nitrobenzene-1-sulfonamide (86 g,390.61mmol,1 eq.) Cs 2 CO 3 (254.5 g,781.21mmol,2 eq.) was placed in a 2000mL round bottom flask. The resulting solution was stirred in an oil bath at 50℃for 4h. The resulting solution was diluted with 3000mL of water. The solid was collected by filtration. Obtaining 4- [ [ (4-fluoro-oxazolidin-4-yl) methyl]Amino group]-3-nitrobenzene-1-sulfonamide 50.6g (38.86%) as yellow solid, and some crude product from the aqueous phase. 1 H NMR(300MHz,DMSO d 6 ,ppm)δ8.58(t,J=6.4Hz,1H),8.48(d,J=2.2Hz,1H),7.83(dd,J=9.1,2.0Hz,1H),7.41(d,J=9.2Hz,1H),7.34(s,2H),3.84–3.70(m,4H),3.59–3.45(m,2H),1.87–1.66(m,4H)。
Synthesis of N- (3-hydroxybutyl) -4-toluene-1-sulfonamide: 4-aminobutan-2-ol hydrochloride (4 g,31.847mmol,1 eq), DCM (40 mL), TEA (3.56 g,35.181mmol,1.10 eq) were placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). Then, 4-toluene-1-sulfonyl chloride (6.08 g,31.893mmol,1 eq.) was added at 0deg.C. The resulting solution was stirred at 25℃for 2h. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. 4.4g (56.7) of N- (3-hydroxybutyl) -4-toluene-1-sulfonamide are obtained 8%) as a colourless oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ7.82–7.73(d,J=9.0Hz,2H),7.39–7.30(d,J=9.0Hz,2H),4.06–3.82(m,1H),3.24–3.16(m,1H),3.06–3.00(m,1H),2.45(s,3H),1.70–1.55(m,2H),1.20(d,J=6.2Hz,3H)。
N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]-synthesis of 4-toluene-1-sulfonamide: n- (3-hydroxybutyl) -4-toluene-1-sulfonamide (4.4 g,18.083mmol,1.10 eq.) and THF (60 mL) were placed in a three-necked round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). Subsequently, at 0deg.C, part of NaH (1.97 g,49.255mmol,3 eq., 60%) was added. The resulting solution was stirred at 0℃for 0.5h. To which 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] is added]Methyl group]-1H-pyrrole [2,3-b ]]Pyridine (5.66 g, 16.390 mmol,1 eq.). The resulting solution was stirred overnight in an oil bath at 50 ℃. The reaction mixture was cooled to 25 ℃. 500mL of NH was then added 4 The reaction was quenched by Cl. The resulting solution was extracted with 2X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. To obtain N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group ]6g (64.37%) of 4-toluene-1-sulfonamide as colorless oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.04(s,1H),7.74–7.63(m,2H),7.23–7.11(m,3H),6.41(d,J=3.6Hz,1H),5.56(d,J=3.3Hz,2H),5.89–5.33(m,1H),3.59–3.46(m,2H),3.17(t,J=6.0Hz,2H),2.37(s,3H),2.08–1.78(m,2H),1.37(d,J=6.2Hz,3H),0.94–0.85(m,2H),-0.06(s,9H)。
13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: n- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]4-toluene-1-sulfonamide (6 g,10.552mmol,1 eq.) dimethyl sulfoxide (60 mL), pyridine-2-carboxylic acid (1.04 g, 8.447 mmol,0.80 eq.) CuI (2.41)g, 12.264 mmol,1.20 eq), K 2 CO 3 (4.38 g,31.692mmol,3 eq.) was placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The resulting solution was stirred in an oil bath at 120℃for 2 days. The reaction mixture was cooled to 25 ℃. The resulting solution was diluted with 500mL of water. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 2X 1000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. To obtain 13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group ]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradeca-1 (9), 2,5, 7-tetraene 2.8g (54.41%) as yellow oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.10(s,1H),7.53–7.40(m,2H),7.30(d,J=3.6Hz,1H),7.24–7.12(m,2H),6.50(d,J=3.6Hz,1H),5.65(d,J=10.7Hz,1H),5.49(d,J=10.7Hz,1H),4.37–4.23(m,1H),3.94–3.80(m,1H),3.62–3.36(m,3H),2.37(s,3H),1.89–1.64(m,2H),1.23(d,J=6.3Hz,3H),1.01–0.76(m,2H),-0.07(s,9H)。
13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: na (793.5 mg,34.51mmol,6.01 eq), naphthalene (4.42 g,34.48mmol,6.01 eq), dimethyl ether (20 mL) were placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The mixture was stirred at room temperature for 40min until sodium/naphthalene was completely formed. 13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene (2.8 g,5.741mmol,1 eq.) and tetrahydrofuran (20 mL) were placed in another round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The above solution was then added at-78 ℃. The resulting solution was stirred at room temperature for 2h. Then 500mLNH was added 4 Cl, stopping the reaction. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue is put on a silica gel column The above was treated with ethyl acetate/petroleum ether (1:3). Obtaining 13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene 1.2g (62.67%) as yellow oil. 1 H NMR(300MHz,DMSO-d 6 ,ppm)δ7.43–7.26(m,2H),6.27(d,J=3.5Hz,1H),5.43(d,J=3.4Hz,2H),5.13(s,1H),4.14–4.04(m,1H),3.58–3.39(m,3H),3.28–3.10(m,1H),2.95–2.77(m,1H),2.09–1.85(m,1H),1.85–1.64(m,1H),1.37(d,J=6.3Hz,3H),0.88–078(m,2H),-0.09(s,9H)。
13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: purification of the crude product by high performance liquid chromatography HPLC (0.5 g): instrument name: SHIMADZU LC-20AD, liquid chromatography parameters: pump mode: binary gradient, pump starting concentration: 50.0% of total flow: 15mL/min, phase A: n-hexane (0.1% dea), phase B: ethanol, column name: CHIRALpak IA-3, length: 50mm, inside diameter: 4.6mm, particle size: 3.0 μm, column temperature: 25 ℃, PDA model: SPD-M20A, wavelength: 190nm to 500nm. Obtain (peak 1 presumes R) [ a ]]= -6.78 ° (in CH 2 Cl 2 (13R or S) -13-methyl-4-ethoxy [2- (trimethylsilyl) oxy (C=0.129 g/100mL, T=27℃)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetraene 220mg (yellow oil) and (peak 2 supposedly S) [ a ]]= +11.84 ° (in CH 2 Cl 2 (13S or R) -13-methyl-4-ethoxy [2- (trimethylsilyl) oxy (C=0.106 g/100mL, T=27℃) ]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene 230mg (yellow oil).
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene (210.00 mg,0.630mmol,1 eq.), toluene (5.00 mL), 2-bromo-4- (4- [ [2- (4- [) ])Chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl]Methyl group]Methyl piperazin-1-yl benzoate (669.87 mg, 1.319 mmol,2 eq.) Cs 2 CO 3 (1025.80 mg,3.148mmol,5 eq.) Xantphos Pd 2G precatalyst (111.70 mg,0.126mmol,0.20 eq.) is placed in a vial (the vial is purged and stored under nitrogen inert atmosphere). The resulting solution was stirred overnight in an oil bath at 110 ℃. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. 240mg (31.09%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate as a yellow oil. Liquid chromatography-mass spectrometry detection: (ES, M/z) m+1=784.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoic acid: 4- (4- [ [ 2-4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate (200 mg,0.255mmol,1 eq.) H 2 O (1.00 mL), methanol (2.00 mL), dioxane (2.00 mL), sodium hydroxide (61.18 mg,1.530mmol,6 eq.) were placed in a round bottom flask. The resulting solution was stirred at 70℃for 12h and the resulting mixture was concentrated. The pH of the solution was adjusted to 5 with hydrochloric acid (1 mol/L). The solid was collected by filtration. 180mg of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (91.64%) as a white solid. H-NMR (CDCl 3,300 ppm): delta 8.17 (s, 1H), 7.28 (s, 1H), 7.00-6.79 (m, 4H), 6.35 (s, 1H), 5.70-5.67 (d, J=9Hz, 1H), 5.56-5.52 (d, J=12Hz, 1H), 4.38-4.35%m,1H),3.81(s,1H),3.58-3.56(m,4H),3.26(s,3H),2.84(s,1H),2.35(s,3H),2.24(s,2H),2.12(s,2H),2.04(s,2H),1.60(s,6H),1.28(s,1H),1.01(s,7H),0.98-0.95(m,3H),0.00(s,9H)。
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (4-fluorooxalan-4-yl) methyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid (100.00 mg,0.130mmol,1 eq.), 4- [ [ [ (4-fluorooxalan-4-yl) methyl ] amino ] -3-nitrobenzene-1-sulfonamide (43.26 mg,0.130mmol,1 eq.), DCM (3 mL), DMAP (63.42 mg,0.519mmol,4 eq.), EDCI (49.76 mg,0.260mmol,2 eq.) were placed in a flask. The resulting solution was stirred at room temperature for 12h. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:0) on a silica gel column. 80mg of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ 4-fluorooxalan-4-yl) methyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradecan-1 (9), 2,5, 7-tetraen-10-yl ] benzamide (56.77%) are obtained as a yellow solid.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (4-fluorooxalan-4-yl) methyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ (13S) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (4-fluorooxalan-4-yl) methyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) oxy]Ethoxy group]Amino group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetralinAlkenyl-10-yl]Benzamide (80.00 mg,0074mmol,1 eq), tetrahydrofuran (5 mL), TBAF (385.28 mg,1474mmol,20 eq.) and ethane-1, 2-diamine (88.56 mg,1474mmol,20 eq.) were placed in a round bottom flask. The resulting solution was stirred at 70℃for 48h and the resulting mixture was concentrated. The crude product was purified by Prep-HPLC under the following conditions (Waters-2767): chromatographic column, X-bridge RP18,5 μm, 19X 100mm; mobile phase, 0.03% ammonia (0.03% NH) 4 HCO 3 And NH 4 OH) and CH 3 CN(32%CH 3 CN reached 52% in 6 min); detector, UV 254nm. To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- (4- [ [ (4-fluorooxalan-4-yl) methyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ (13S) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Benzamide 25mg (35.51%) as a yellow solid. H-NMR (d-DMSO, 300M Hz,ppm): delta 8.59-8.67 (m, 1H), 8.69 (s, 1H), 8.48 (s, 1H), 8.03-7.94 (m, 2H), 7.15-7.14 (m, 2H), 6.97-6.93 (m, 3H), 6.76-6.73 (m, 3H), 6.20 (s, 1H), 3.96-3.69 (m, 5H), 3.49-3.25 (m, 7H), 2.85 (s, 2H), 2.37-2.25 (m, 6H), 2.15-2.04 (m, 3H), 1.89-1.83 (m, 3H), 1.71-1.69 (m, 4H), 1.50-1.46 (m, 2H), 1.06-0.99 (m, 6H).
Example 21
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-methyl ] piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] -N- [ 3-nitro-4- [ (oxa-4-ylmethyl) amino ] benzenesulfonyl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of ethyl 2-cyano-3-hydroxy-2-methylpropionate: ethyl 2-cyanopropionate (20.00 g,157.303mmol,1 eq.) CH 3 CN (200.00 mL), formaldehyde (32.00 g,394.325mmol,2.51 equiv., 37%), TEA (800.00 mg,7.906mmol,0.05 equiv.) were placed in a round bottom flask. The resulting solution was stirred in an oil bath at 50 ℃And (5) at night. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was eluted with ethyl acetate/petroleum ether (1:4) on a silica gel column. 20g (80.90%) of ethyl 2-cyano-3-hydroxy-2-methylpropionate were obtained as colorless oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ5.01–4.72(m,1H),4.38–4.23(m,2H),4.04–3.82(m,2H),1.60(d,J=2.6Hz,3H),1.35(td,J=7.1,2.7Hz,3H)。
Synthesis of 3-hydroxy-2- (hydroxymethyl) -2-methylpropionitrile: ethyl 2-cyano-3-hydroxy-2-methylpropionate (12.00 g,76.351mmol,1 eq.) and methanol (120.00 mL) were placed in a round bottom flask. Subsequently adding part of NaBH at 0 DEG C 4 (4.40 g,116.301mmol,1.52 eq.). The resulting solution was stirred at 25℃for 1.5h. Then 20mL of water was added to quench the reaction. The resulting mixture was concentrated under vacuum. The residue was applied to a silica gel column and eluted with dichloromethane/methanol (10:1). 8g (91.01%) of 3-hydroxy-2- (hydroxymethyl) -2-methylpropanenitrile are obtained as a pale yellow oil. 1 H NMR(300MHz,MeOD-d 4 ,ppm)δ3.74–3.50(m,4H),1.29(s,3H)。
2- [ [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Methyl group]-synthesis of 3-hydroxy-2-methylpropanenitrile: 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] ]Methyl group]Pyrrole [2,3-b ]]Pyridine (12.00 g,34.754mmol,1 eq.) CH 3 CN (200.00 mL), 3-hydroxy-2- (hydroxymethyl) -2-methylpropionitrile (8.00 g, 69.480 mmol,2 eq.) Cs 2 CO 3 (28.34 g,86.981mmol,2.50 eq.) is placed in a round bottom flask. The resulting solution was stirred in an oil bath at 80℃for 12h. The reaction mixture was cooled to room temperature. The resulting solution was treated with 500mLH 2 O extraction. The resulting solution was extracted with 2X 300mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 1000mL of brine. The mixture was dried over anhydrous sodium sulfate. The solid was filtered. The resulting mixture was concentrated under vacuum. The residue was eluted with ethyl acetate/petroleum ether (1:5) on a silica gel column. The obtained 2- [ [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Methyl group]6g (39.20%) of 3-hydroxy-2-methylpropanenitrile as a pale yellow oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.08(s,1H),7.18(d,J=3.6Hz,1H),6.42(d,J=3.6Hz,1H),5.55(s,2H),4.70(d,J=10.8Hz,1H),4.53(d,J=10.9Hz,1H),3.85(d,J=4.0Hz,2H),3.62–3.46(m,2H),1.53(s,3H),1.05–0.85(m,2H),-0.03(s,8H)。
N- [6- [ 2-cyano-2- (hydroxymethyl) -2-methylethoxy ]]-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]Pyrrole [2,3-b ]]Pyridin-5-yl]-synthesis of 4-methylbenzenesulfonamide: 2- [ [ (5-bromo-1- [ [ (2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Methyl group]-3-hydroxy-2-methacrylonitrile (6.00 g, 13.264 mmol,1 eq), dimethyl sulfoxide (50.00 mL), p-toluenesulfonamide (4.67 g,27.276mmol,2 eq), picolinic acid (1.35 g,10.966mmol,0.80 eq), cuI (3.12 g,16.382mmol,1.20 eq), K 2 CO 3 (5.66 g,40.954mmol,3.01 eq.) is placed in a round bottom flask (the flask is purged and kept under nitrogen inert atmosphere). The resulting solution was stirred in an oil bath at 120℃for 2 days. The reaction mixture was cooled to room temperature. The resulting solution was treated with 500mLH 2 O extraction. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 2X 1000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was eluted with ethyl acetate/petroleum ether (1:1) on a silica gel column. Obtaining N- [6- [ 2-cyano-2- (hydroxymethyl) -2-methylethoxy ]]-1- [ [2- (trimethylsilyl) ethoxy ]]Amino group]Pyrrole [2,3-b ]]Pyridin-5-yl]2.45g (33.89%) of 4-methylbenzenesulfonamide as a white solid. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.14(s,1H),7.68–7.58(m,2H),7.25–7.13(m,3H),6.76(s,1H),6.48(d,J=3.6Hz,1H),5.47(d,J=4.4Hz,2H),4.41(d,J=11.2Hz,1H),4.29(d,J=11.3Hz,1H),3.61(dd,J=7.9,6.3Hz,1H),3.56–3.43(m,4H),2.38(s,3H),1.34(s,3H),0.97–0.84(m,2H),-0.05(s,9H)。
12-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene-12-carbonitrile: n- [6- [ 2-cyano-2- (hydroxymethyl) -2-methylethoxy ]]-1- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]Pyrrole [2,3-b ]]Pyridin-5-yl]-4-Methylbenzenesulfonamide (2.45 g,4.616mmol,1 eq.), THF (30 mL), PPh3 (2)42g,9.227mmol,2 eq) was placed in a round bottom flask. DEAD (1.61 g,9.245mmol,2 eq.) was then added dropwise at 0deg.C and stirred. The resulting solution was stirred at 25℃for 1h. With 500mL H 2 O dilutes the resulting solution. The resulting solution was extracted with 2X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was eluted with ethyl acetate/petroleum ether (1:3) on a silica gel column. To obtain 12-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetraene-12-carbonitrile 2g (84.50%) as a yellow solid.
12-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene-12-carbonitrile: na (539.00 mg,23.445mmol,6.01 eq), naphthalene (3.00 g,23.406mmol,6 eq), dimethyl ether (10.00 mL) were placed in a vial (the vial was purged and stored in a nitrogen inert atmosphere). The mixture was stirred at room temperature for 40min until sodium/naphthalene was completely formed. The above solution was then added to 12-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy ] in THF (10.00 mL) at-78deg.C]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Tetradecane-1 (9), 2,5, 7-tetraene-12-carbonitrile (2.00 g,3.901mmol,1 eq.). The resulting solution was stirred at 25℃for 2h. 500mL of NH was then added 4 The reaction was quenched with aqueous Cl. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was eluted with ethyl acetate/petroleum ether (1:3) on a silica gel column. Obtaining 12-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetraene-12-carbonitrile 1g (71.50%) as a yellow oil. 1 H NMR(300MHz,DMSO-d 6 ,ppm)δ7.44(s,1H),7.40(d,J=3.5Hz,1H),6.34(d,J=3.5Hz,1H),5.65–5.52(m,1H),5.44(s,2H),4.51–4.36(m,1H),3.86(d,J=12.2Hz,1H),3.56–3.42(m,3H),2.94(dd,J=13.2,2.3Hz,1H),1.31(s,3H),0.89–0.71(m,2H),-0.08(s,9H).
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (12-cyano-12-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl) benzoate (1.18 g,2.218mmol,1.99 eq), toluene (10.00 mL), 12-methyl-4- [ [2- (trimethylsilyl) ethoxy)]Methyl group ]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetraene-12-carbonitrile (400.00 mg,1.116mmol,1 eq.) Cs 2 CO 3 (1.82G, 5.586mmol,5.01 eq.) Xantphos Pd 2G precatalyst (149.00 mg,0.168mmol,0.15 eq.) is placed in a round bottom flask (the flask is purged and kept under nitrogen inert atmosphere). The resulting solution was stirred in an oil bath at 110 ℃ for 2 days. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was eluted with ethyl acetate/petroleum ether (1:1) on a silica gel column. To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (12-cyano-12-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate 330mg (36.54%) as a yellow oil. LC-MS (ES, M/z): M+1=809.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: methyl 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- (12-cyano-12-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (330.00 mg,0.408mmol,1 eq.), THF (20.00 mL), TBAF.3H 2 O (5.00 g), ethylenediamine (2.50 g,41.597mmol,102.04 eq.) was placed in a round bottom flask. The resulting solution was stirred in an oil bath at 70 ℃ for 2 days. The reaction mixture is coated withCooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (2:1) on a silica gel column. To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Methyl benzoate 220mg (79.45%) as a pale yellow solid. LC-MS (ES, M/z): M+1=679.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzoic acid. 4- (4- [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3.7 ]]]Tetradec-1 (9), 2.5.7-tetraen-10-yl]Methyl benzoate (190.00 mg,0.280mmol,1 eq.), meOH (12.00 mL), dioxane (12.00 mL), H 2 O(4.00mL)、LiOH·H 2 O (353.00 mg,8.412mmol,30.07 eq.) was placed in a vial. The resulting solution was stirred overnight in an oil bath at 60 ℃. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The pH of the solution was adjusted to 6-7 with HCl (2 mol/L). The resulting solution was extracted with 2X 100mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was purified by Prep-TLC with ethyl acetate. To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid 80mg (42.99%) as a pale yellow solid. LC-MS (ES, M/z): M+1=665.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-methyl)]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]-N- [ 3-nitro-4- [ (oxazolidin-4-ylmethyl) amino group]Benzenesulfonyl group]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Benzoic acid (40.00 mg,0.060mmol,1 eq.), DCM (2, 00 mL), 3-nitro-4- [ (oxa-4-ylmethyl) amino]Benzenesulfonamide (19, 00mg,0060mmol,1 eq.), EDCI (23.00 mg,0.120mmol,2 eq.) and DMAP (29.00 mg,0.237mmol,3.95 eq.) were placed in a vial. The resulting solution was stirred overnight at 25 ℃. The resulting mixture was concentrated in a vacuum environment. The crude product was purified by Prep-HPLC under the following conditions (2#SHIMADZU (HPLC-01)): chromatographic column, XBridge Prep C18 OBD chromatographic column, 5um,19 x 150mm; mobile phase, ACN and water (0.05% nh 3 H2O) (20% b phase reached 75% in 1min, 95% in 7min, 95% hold in 1min, 20% drop in 1 min); detector, UV 254/220nm. The obtained 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ 12-cyano-12-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3.7 ] ]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]-N- [ 3-nitro-4- [ (oxazolidin-4-ylmethyl) amino group]Benzamide 18.8mg (32.48%) as a yellow solid. Liquid chromatography mass spectrometry detection: (ES, M/z) m+1=962, retention time=1.461 min. Retention was measured using a reverse phase chromatography column (C18). Shimadzu LCMS 2020; SUPELCO Ascentis Sxpress C18, 50 x 3.0mm,2.7 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 100% acetonitrile, 3.0min; the temperature of the oven is 40 ℃; flow rate: 1.5mL/min. 1 H NMR(300MHz,DMSO-d 6,ppm )δ8.55(s,1H),8.34(s,1H),7.60–7.31(m,4H),7.23–7.00(m,3H),6.76(d,J=26.1Hz,4H),6.04(s,1H),3.88(dd,J=11.7,4.0Hz,2H),3.38–3.12(m,7H),2.77(d,J=19.8Hz,2H),2.24(d,J=20.9Hz,6H),1.99(s,4H),1.66(d,J=12.4Hz,2H),1.42(t,J=6.3Hz,2H),1.36–1.03(m,8H),0.96(s,6H)。
Example 22
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl l-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] -N- (3-nitro-4- [ [ (oxa-4-yl) methyl ] amino ] benzenesulfonyl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of N- (3-hydroxybutyl) -4-toluene-1-sulfonamide: 4-aminobutan-2-ol hydrochloride (4 g,31.847mmol,1 eq), DCM (40 mL), TEA (3.56 g,35.181mmol,1.10 eq) were placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). Then, 4-toluene-1-sulfonyl chloride (6.08 g,31.893mmol,1 eq.) was added at 0deg.C. The resulting solution was stirred at 25℃for 2h. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. 4.4g (56.78%) of N- (3-hydroxybutyl) -4-toluene-1-sulfonamide are obtained as a colourless oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ7.82–7.73(d,J=9.0Hz,2H),7.39–7.30(d,J=9.0Hz,2H),4.06–3.82(m,1H),3.24–3.16(m,1H),3.06–3.00(m,1H),2.45(s,3H),1.70–1.55(m,2H),1.20(d,J=6.2Hz,3H)。
N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]-synthesis of 4-toluene-1-sulfonamide: n- (3-hydroxybutyl) -4-toluene-1-sulfonamide (4.4 g,18.083mmol,1.10 eq.) and THF (60 mL) were placed in a three-necked round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). Subsequently, an appropriate amount of NaH (1.97 g,49.255mmol,3.00 eq., 60%) was added at 0deg.C. The resulting solution was stirred at 0℃for 0.5h. To which 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] is added]Methyl group]-1H-pyrrole [2,3-b ]]Pyridine (5.66 g, 16.390 mmol,1 eq.). The resulting solution was stirred overnight in an oil bath at 50 ℃. The reaction mixture was cooled to 25 ℃. 500mL of NH was then added 4 The reaction was quenched by Cl. The resulting solution was extracted with 2X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. To obtain N- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]6g (64.37%) of 4-toluene-1-sulfonamide as colorless oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.04(s,1H),7.74–7.63(m,2H),7.23–7.11(m,3H),6.41(d,J=3.6Hz,1H),5.56(d,J=3.3Hz,2H),5.89–5.33(m,1H),3.59–3.46(m,2H),3.17(t,J=6.0Hz,2H),2.37(s,3H),2.08–1.78(m,2H),1.37(d,J=6.2Hz,3H),0.94–0.85(m,2H),-0.06(s,9H)。
13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: n- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy]Methyl group]-1H-pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Butyl group]-4-toluene-1-sulfonamide (6 g,10.552mmol,1 eq.) dimethyl sulfoxide (60 mL), pyridine-2-carboxylic acid (1.04 g, 8.4478 mmol,0.80 eq.), cuI (2.41 g, 12.254 mmol,1.20 eq.), K 2 CO 3 (4.38 g,31.692mmol,3 eq.) was placed in a round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The resulting solution was stirred in an oil bath at 120℃for 2 days. The reaction mixture was cooled to 25 ℃. The resulting solution was diluted with 500mL of water. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 2X 1000mL of brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. To obtain 13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradeca-1 (9), 2,5, 7-tetraene 2.8g (54.41%) as yellow oil. 1 H NMR(300MHz,CDCl 3 -d,ppm)δ8.10(s,1H),7.53–7.40(m,2H),7.30(d,J=3.6Hz,1H),7.24–7.12(m,2H),6.50(d,J=3.6Hz,1H),5.65(d,J=10.7Hz,1H),5.49(d,J=10.7Hz,1H),4.37–4.23(m,1H),3.94–3.80(m,1H),3.62–3.36(m,3H),2.37(s,3H),1.89–1.64(m,2H),1.23(d,J=6.3Hz,3H),1.01–0.76(m,2H),-0.07(s,9H)。
13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa [2,4, 10-triazatricyclic- [7.5.0.0 ] [3,7 ]]]Synthesis of tetradec-1 (9), 2,5, 7-tetraene: to a round bottom flask (flask is purged and protected)Sodium metal (793.5 mg,34.51mmol,6.01 eq.) naphthalene (4.42 g,34.48mmol,6.01 eq.) and DME (20 mL) were added in an inert atmosphere in the presence of nitrogen. The mixture was stirred at room temperature for 40min until sodium/naphthalene was completely formed. 13-methyl-10- (4-methylbenzenesulfonyl) -4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene (2.8 g,5.741mmol,1 eq.) and tetrahydrofuran (20 mL) were placed in another round bottom flask (the flask was purged and kept under nitrogen inert atmosphere). The above solution was then added at-78 ℃. The resulting solution was stirred at room temperature for 2h. Then 500mLNH was added 4 Cl, stopping the reaction. The resulting solution was extracted with 3X 200mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 500mL brine. The mixture was dried over anhydrous sodium sulfate, then filtered and concentrated in vacuo. The residue was treated with ethyl acetate/petroleum ether (1:3) on a silica gel column. Obtaining 13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene 1.2g (62.67%) as yellow oil. 1 H NMR(300MHz,DMSO-d 6 ,ppm)δ7.43–7.26(m,2H),6.27(d,J=3.5Hz,1H),5.43(d,J=3.4Hz,2H),5.13(s,1H),4.14–4.04(m,1H),3.58–3.39(m,3H),3.28–3.10(m,1H),2.95–2.77(m,1H),2.09–1.85(m,1H),1.85–1.64(m,1H),1.37(d,J=6.3Hz,3H),0.88–078(m,2H),-0.09(s,9H)。
13-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradecane-1 (9), 2,5, 7-tetraene: purification of the crude product by high performance liquid chromatography HPLC (0.5 g): instrument name: SHIMADZU LC-20AD, liquid chromatography parameters: pump mode: binary gradient, pump starting concentration: 50.0% of total flow: 15mL/min, phase A: n-hexane (0.1% dea), phase B: ethanol, column name: CHIRALpak IA-3, length: 50mm, inside diameter: 4.6mm, particle size: 3.0 μm, column temperature: 25 ℃, PDA model: SPD-M20A, wavelength: 190nm to 500nm. Obtain (peak 1 presumes R) [ a ]]= -6.78 ° (in CH 2 Cl 2 (13R or S) -13-methyl-4-ethoxy [2- (trimethylsilyl) oxy (C=0.129 g/100mL, T=27℃)]Methyl group]-14-oxa-2, 4, 10-triazasHeterotricyclo [7.5.0.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetraene 220mg (yellow oil) and (peak 2 supposedly S) [ a ]]= +11.84 ° (in CH 2 Cl 2 (13S or R) -13-methyl-4-ethoxy [2- (trimethylsilyl) oxy (C=0.106 g/100mL, T=27℃)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Tetradec-1 (9), 2,5, 7-tetraene 230mg (yellow oil).
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen (210.00 mg,0.630mmol,1 eq.), toluene (5.00 mL), 2-bromo-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl benzoate (669.87 mg, 1.319 mmol,2 eq.) Cs 2 CO 3 (1025.80 mg,3.148mmol,5 eq.) Xantphos Pd 2G precatalyst (111.70 mg,0.126mmol,0.20 eq.) is placed in a vial (the vial is purged and stored under nitrogen inert atmosphere). The resulting solution was stirred overnight in an oil bath at 110 ℃. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under vacuum. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. To give 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradecan-1 (9), 2,5, 7-tetraen-10-yl]240mg (31.09%) of methyl benzoate as a yellow oil. LC-MS (ES, M/z): M+1=784.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid: to a round bottom flask was added methyl-4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] benzoate (200 mg,0.255mmol,1 eq), water (1 mL), methanol (2 mL), dioxane (2 mL), and sodium hydroxide (61.18 mg,1.530mmol,6 eq). The resulting solution was stirred at 70℃for 12h and the resulting mixture was concentrated. The pH of the solution was adjusted to 5 with hydrochloric acid (1 mol/L). The solid was collected by filtration. The final product was 180mg (91.64%) 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid as a white solid. H-NMR-PH-PHNW-4-82-1: delta 8.17 (s, 1H), 7.28 (s, 1H), 7.00-6.79 (m, 4H), 6.35 (s, 1H), 5.70-5.67 (d, J=9Hz, 1H), 5.56-5.52 (d, J=12 Hz, 1H), 4.38-4.35 (m, 1H), 3.81 (s, 1H), 3.58-3.56 (m, 4H), 3.26 (s, 3H), 2.84 (s, 1H), 2.35 (s, 3H), 2.24 (s, 2H), 2.12 (s, 2H), 2.04 (s, 2H), 1.60 (s, 6H), 1.28 (s, 1H), 1.01 (s, 7H), 0.98-0.95 (m, 3H), 0.00 (s, 9H).
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradec 1 (9), 2,5, 7-tetraen-10-yl ] -N- (3-nitro-4 [ (oxa-4-yl) methyl ] amino ] benzenesulfonyl) benzamide: to a round bottom flask was added 4- (4- [ [2- (4-chlorophenyl l) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] benzoic acid (100.00 mg,0.130mmol,1 eq.), 3-nitro-4- [ [ (oxalan-4-yl) methyl ] amino ] benzene-1-sulfonamide (40.93 mg,0.130mmol,1 eq.), DCM (3 mL), DMAP (63.42 mg,0.519mmol,4 eq.) and EDCI (49.76 mg,0.260mmol,2 eq.). The resulting solution was stirred at room temperature for 12h. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:0) on a silica gel column. The final product was 80mg (57.72%) 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] -N- (3-nitro-4- [ [ (oxa-4-yl) methyl ] amino ] benzenesulfonyl) benzamide as a yellow solid. LC-MS (ES, M/z): M+1= 1066.46.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]-N- (3-nitro-4- [ [ (oxan-4-yl) methyl)]Amino group]Benzenesulfonyl) benzamide synthesis: 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) was added to a round bottom flask]Methyl group]Piperazin-1-yl l) -2- [ (13S) -13-methyl-4- [ [2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]-N- (3-nitro-4- [ [ (oxan-4-yl) methyl)]Amino group]Benzenesulfonyl) benzamide (80.00 mg,0.075mmol,1 eq), THF (5 mL), TBAF (391.77 mg,1.498mmol,20 eq.) and ethylenediamine (90.05 mg,1.498mmol,20 eq.). The resulting solution was stirred at 70℃for 48h and the resulting mixture was concentrated. The crude product was purified by Prep-HPLC under the following conditions (Waters-2767): chromatographic column: x-bridge RP18,5 μm, 19X 100mm; mobile phase: aqueous solution (NH) containing 0.03% ammonia 4 HCO 3 And NH 4 OH 0.03% total) with acetonitrile (acetonitrile content rises from 32% to 52% in 6 min); the detector comprises: UV 254nm. The final product was 25mg (35.59%) of 4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- [ (13S) -13-methyl-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]-N- (3-nitro-4- [ [ (oxan-4-yl) methyl)]Amino group]Benzenesulfonyl) benzamide, yellow solid. LC-MS-PH-PHNW-4-77-0B: (ES, M/z) m+1= 937.54. Retention time was determined using a reverse phase chromatography column (C18). Shimadzu LCMS 2020;50*3.0Kinetex 2.6u XB-C18,2.6 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient. H-NMR-PH-PHNW-4-77-0B: (300 MHz, chloroform-d) delta 8.84-8.78 (m, 2H), 8.34 (s, 1H), 7.98-7.96 (d, J=6.0 Hz, 2H), 7.28-7.27 (m, 1H), 7.17-7.15 (m, 1H), 7.04-6.94 (m, 3H), 6.76-6.73 (m, 2H), 6.64 (s, 1H), 6.22 (s, 1H), 4.02–3.93(m,3H),3.46–3.30(m,7H),3.10(s,2H),2.85(s,2H),2.37(s,4H),2.25(s,2H),2.15–2.03(m,3H),1.85–1.80(m,1H),1.70(s,4H),1.60(s,2H),1.50–1.46(m,4H),1.00(s,6H)。
Example 23
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- ([ [ (2S) -1, 4-dioxan-2-yl)]Methyl group]Synthesis of amino) -3-nitrobenzene-1-sulfonamide: 4-fluoro-3-nitrobenzene-1-sulfonamide (1.43 g,0.007mmol,1 eq.) 1- [ (2S) -1, 4-dioxane-2-yl]Carboxamide hydrochloride (1 g,6.510mmol,1 eq.), THF (30 mL), cs 2 CO 3 (8.48 g,0.026mmol,4 eq.) in a round bottom flask. The resulting solution was stirred overnight in an oil bath at 50 ℃. The solid was collected by filtration. The solid was dried in an oven under reduced pressure. 1.82g (88.10%) of 4- ([ [ (2S) -1, 4-dioxane-2-yl) are obtained]Methyl group]Amino) -3-nitrobenzene-1-sulfonamide as a yellow solid. LC-MS (ES, M/z): M+1=318.
Synthesis of 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] pyrrole ] isoindole-1, 3-dione: an inert atmosphere of nitrogen was introduced into a three-necked round bottom flask and maintained, and 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridine (53.50 g,154.944mmol,1 eq.) was added thereto. 2- (3-hydroxypropyl) isoindole-1, 3-dione (31.80 g,154.944mmol,1 eq.) was then added in portions followed by dioxane (500.00 mL) and NaH (9.30 g, 232.418 mmol,1.50 eq., 60%). The resulting solution was stirred at 80℃for 4h and the reaction mixture was cooled with a water/ice bath. Then 500ml of LAcOH/ice water solution was added to stop the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate. The resulting mixture was washed with 3X 500mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:10) on a silica gel column. The final product was 58g (70.56%) 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propyl ] isoindole-1, 3-dione as a colorless oil. LC-MS (ES, M/z): M+1=554.
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]Synthesis of propan-1-amine: 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Propyl group]Isoindole-1, 3-dione (58.00 g, 109.336 mmol,1 eq.), ethanol (300.00 mL) and NH 2 NH 2 ·H 2 O (68.42 g,1093.321mmol,10 equiv., 80%) was charged to a round bottom flask. The resulting solution was stirred at room temperature for 4h. The resulting mixture was concentrated. Then 200mL of water was added to stop the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate and the resulting mixture was washed with 2X 300mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:2) on a silica gel column. 37.5g (85.66%) of 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) are obtained]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]Propane-1-amine as a yellow oil.
Synthesis of 4[ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazacyclo [7.4.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraene: in a round bottom flask was placed 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propan-1-amine (37.50G, 93.618 mmol,1 eq.) toluene (500.00 mL), t-BuONa (27.00G, 280.947mmol,3 eq.) BrettPhos Pd G3 (4.25G, 4.688mmol,0.05 eq.). The resulting solution was stirred at 110℃for 4h and the solid was filtered off. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. The final product was 16.1g (53.81%) of 4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] fourteen-1 (9), 2,5, 7-tetraene as a brown solid. LC-MS (ES, M/z): M+1=320.
Synthesis of 2-bromo-5, 5-dimethylcyclohexyl-1-ene-1-carbaldehyde: to a three-necked round bottom flask was added DMF (7.24 g,99.050mmol,2.5 eq.) and DCM (100 mL). Then, at 0 ℃, PBr is added dropwise 3 (24.67 g,91.139mmol,2.30 eq.) and stirred. After stirring the resulting solution at 0deg.C for 1h, 4-dimethylcyclohexen-1-one (5.00 g,39.620mmol,1 eq.) was added in portions at 0deg.C. The resulting solution was stirred at room temperature overnight. The reaction was then stopped by adding 100mL of ice water. With Na 2 CO 3 The pH of the solution was adjusted to 5. The resulting solution was extracted 3 times with 100mL of dichloromethane dried over anhydrous sodium sulfate and concentrated. The final product was 5g (58.13%) 2-bromo-5, 5-dimethylcyclohexyl-1-en-1-carbaldehyde as a yellow oil. LC-MS (ES, M/z): M+1=217/219.
Synthesis of 2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-ene-1-carbaldehyde: into a round bottom flask was charged 2-bromo-5, 5-dimethylcyclohexyl-1-ene-1-carbaldehyde (2.50 g,11.515mmol,1 eq), benzoic acid, 4-chloro (1.80 g,11.515mmol,1 eq), DME (25 mL), water (25 mL), na 2 CO 3 (2.44 g,23.030mmol,2 eq.) and Pd (dppf) Cl 2 CH 2 Cl 2 (0.94 g,1.151mmol,0.10 eq.). The resulting solution was stirred at 60℃for 12h. The reaction mixture was cooled to room temperature. Then 50mL of water was added to stop the reaction. The resulting solution was extracted with 3X 50mL of ethyl acetate, dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. The final product was 1.2g (41.89%) 2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-carbaldehyde as a yellow oil. LC-MS: (ES, M/z) m+1=249.
2-bromo-4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazine 1-yl) methyl benzoate synthesis: to a round bottom flask was added 2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-ene-1-carbaldehyde (1.20 g, 4.284 mmol,1 eq), methyl 2-bromo-4- (piperazin-1-yl) benzoate (1.44 g, 4.284 mmol,1 eq), DCE (50.00 mL) and Ti (Oi-Pr) 4 (4.11 g, 14.470 mmol,3 eq.). The resulting solution was stirred at room temperature for 3h. NaBH (OAc) was then added in portions at room temperature 3 (2.04 g, 9.640 mmol,2 eq.). The resulting solution was stirred at room temperature overnight. Then 10mL of water was added to stop the reaction. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. The final product was 600mg (23.38%) of 2-bromo-4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) benzoic acid methyl esters as white oils. LC-MS (ES, M/z): M+1=531/533.
4- (4- [ [2- (4-chlorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.5.0.0 ] [3,7 ],]]tridec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of methyl benzoate: into a round bottom flask was added 4- [ [2- (trimethylsilyl) ethoxy ] ]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraene (150.15 mg,0.470mmol,1 eq.) methyl-2-bromo-4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) benzoate (250.00 mg,0.470mmol,1 eq.) Cs 2 CO 3 (459.40 mg,1.410mmol,3 eq.) toluene (5.00 mL) and Xantphos Pd 2G palladium pre-catalyst (27.19 mg,0.047mmol,0.10 eq.). The resulting solution was stirred at 110 ℃ for 12h until LCMS showed complete consumption of material. The residue was treated with ethyl acetate/petroleum ether (0:1-1:1) on a silica gel column. The final product was 120mg (33.14%) -4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Ethyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate, as a white solid. LC-MS (ES, M/z): M+1=770.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] fourteen-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid: to a round bottom flask was added methyl-4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (120.00 mg,0.156mmol,1 eq), methanol (1 mL), dioxane (1 mL), water (1 mL), and NaOH (37.38 mg,0.934mmol,6 eq). The resulting solution was stirred at 70℃for 12h, then 5mL of water was added to stop the reaction. The pH of the solution was adjusted to 5 with hydrochloric acid (1 mol/L). The solid was collected by filtration. The final product was 100mg (84.88%) 4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid as a white crude solid. LC-MS (ES, M/z): M+1=756, RT=1.288 min.
Synthesis of 4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 [3,7] ] tetradec 1 (9), 2,5, 7-tetraen-10-yl) benzamide: to a round bottom flask was added 4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] ] tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid (100.00 mg,0.132mmol,1 eq), DCM (5 mL), DMAP (64.60 mg,0.529mmol,4 eq), EDCI (50.68 mg,0.264mmol,2 eq) and 4- [ [ (2S) -1, 4-dioxa-2-ylmethyl ] amino ] -3-nitrobenzenesulfonamide (46.14 mg,0.145mmol,1.10 eq). The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The residue was treated with dichloroethane/methanol (1:0-10:1) on a silica gel column. The final product was 100mg (71.65%) 4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradec 1 (9), 2,5, 7-tetraen-10-yl) benzamide as a yellow solid. LC-MS (ES, M/z): M+1=1056.
4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxane-2-Methyl radical]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Synthesis of benzamide: 4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl) was added to a round bottom flask]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide (100.00 mg,0.095mmol,1 eq.), ethylenediamine (113.85 mg,1.894mmol,20 eq.), TBAF (495.31 mg,1.894mmol,20 eq.) and THF (10 mL). The resulting solution was stirred at 70℃for 12h, then 10mL of water was added to stop the reaction. The resulting solution was extracted with 2X 10mL ethyl acetate and concentrated. The crude product was purified by Prep-HPLC under the following conditions (Waters-2767): column, X-bridge RP18,5 μm, 19X 100mm; mobile phase, 0.03% ammonia (0.03% NH) 4 HCO 3 And NH 4 OH) and CH 3 CN(32%CH 3 CN up to 52% in 6 min); detector, UV 254nm. The final product was 25mg (28.52%) of 4- (4- [ [2- (4-chlorophenyl) -5, 5-dimethylcyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl]Benzamide, yellow solid. LC-MS (ES, M/z): M+1=925. 1 H-NMR:(DMSO-d 6 ,300ppm)δ:11.91(s,1H),11.22(s,1H),8.52-8.48(m,2H),7.68-7.63(m,1H),7.47-7.45(d,J=6Hz,1H),7.38-7.35(d,J=9Hz,2H),7.20(s,1H),7.12-7.09(d,J=9Hz,2H),6.92-6.89(m,2H),6.72(s,1H),6.13-6.11(m,1H),4.21(s,2H),3.82-3.75(m,3H),3.68-3.51(m,8H),3.22-3.19(m,2H),2.73(s,2H),2.3-2.10(m,4H),2.08-1.90(m,3H),1.40(m,2H),1.21(m,1H),0.97(s,6H)。
Example 24
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
synthesis of 2-bromocyclohexyl-1-ene-1-carbaldehyde to a 250mL three-necked round bottom flask was added CHCl 3 (80.00 mL,991.80mmol,19.47 eq.). DMF (9.31 g,0.12mmol,2.5 eq.) was then added dropwise at 0deg.C and stirred. To this mixture was added dropwise PBr with stirring at 0 °c 3 (31.72 g,0.11mmol,2.3 eq.). The resulting solution was stirred at 0℃for 1h. Cyclohexanone (5.00 g,50.94mmol,1.00 eq.) was added dropwise to the mixture at room temperature and stirred. The resulting solution was left at room temperature for continued reaction overnight. The reaction was then stopped by adding 100mL of ice water. With Na 2 CO 3 The pH of the solution was adjusted to 5. The resulting solution was extracted 3 times with 100mL of dichloromethane dried over anhydrous sodium sulfate and concentrated. The final product was 5.2g (53.99%) 2-bromocyclohexyl-1-ene-1-carbaldehyde as a pale yellow oil. LC-MS (ES, M/z): M+1:189.
Synthesis of 2- (4-chlorophenyl) cyclohexyl-1-ene-1-carbaldehyde: into a round bottom flask was charged 2-bromocyclohexyl-1-ene-1-carbaldehyde (5.00 g,26.44mmol,1 eq.), DME (50 mL), 4-chlorophenylboronic acid (4.14 g,26.44mmol,1 eq.), na 2 CO 3 (5.66 g,52.89mmol,2 eq.) Pd 2 (dba) 3 (2.42 g,2.64mmol,0.1 eq.) and water (5 mL). The resulting solution was stirred at 60℃for 12h. The reaction mixture was cooled to room temperature. The resulting solution was treated with 50. 50mLH 2 O extraction. The resulting solution was extracted with 3X 50mL ethyl acetate. The resulting mixture was washed with 50mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was applied to a column containing silica gel (ethyl acetate: petroleum ether=1:10 to 2:5). The final product was 3.5g (59.96%) 2- (4-chlorophenyl) cyclohexyl-1-en-1-carbaldehyde as a pale yellow oil. LC-MS (ES, M/z): M+1:221.
2-bromo-4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Synthesis of methyl piperazin-1-yl) benzoate: to one direction Into a three-necked round bottom flask was charged 2- (4-chlorophenyl) cyclohexyl-1-ene-1-carbaldehyde (3.50 g,15.85mmol,1 eq), DCE (35.00 mL,442.10mmol,27.88 eq), methyl 2-bromo-4- (piperazin-1-yl) -benzoate (4.74 g,15.85mmol,1 eq) and Ti (Oi-Pr) 4 (13.52 g,47.57mmol,3 eq.). The resulting solution was stirred at room temperature for 3h. NaBH (AcO) was then added in portions at room temperature 3 (6.72 g,31.71mmol,2 eq.). The resulting solution was reacted at room temperature for 16 hours. Thereafter, 10mL of methanol was added to quench the reaction. The resulting mixture was concentrated. The residue was applied to a column containing silica gel (ethyl acetate: petroleum ether=1:10 to 2:5). The final product was 4.2g (52.56%) of 2-bromo-4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) methyl benzoate, a pale yellow oil. LC-MS (ES, M/z): M+1:503.
Synthesis of 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propyl ] isoindole-1, 3-dione: to a three-necked round bottom flask (which was purged and kept under nitrogen inert atmosphere) was added 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridine (53.50 g,154.944mmol,1 eq). Then 2- (3-hydroxypropyl) isoindole-1, 3-dione (31.80 g,154.944mmol,1 eq.) was added in portions at room temperature. Dioxane (500 mL) and NaH (9.30 g, 232.418 mmol,1.50 eq., 60%) were added at room temperature. The resulting solution was stirred at 80℃for 4h and the reaction mixture was cooled with an ice bath. Then, 500ml of ice water solution of lacoh was added to stop the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate. The resulting mixture was washed with 3X 500mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:10) on a silica gel column. 58g (70.56%) of 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propyl ] isoindole-1, 3-dione are obtained as a colorless oil. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0SUPELCO Ascentis Express C18,2.7 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 4 ℃; flow rate: 1.5mL/min. LC-MS: (ES, M/z): M+1:554.
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]Synthesis of 1-propylamine: ethoxy groups were added to a round bottom flask]Methyl group]Propyl group]Isoindole-pyrrolo [2,3-b]Pyridin-6-yl) oxy]2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) 1, 3-dione (58.00 g,109.33mmol,1 eq.), ethanol (300 mL) and NH 2 NH 2 ·H 2 O (68.42 g,1093.32mmol,10 eq., 80%). The resulting solution was stirred at room temperature for 4h. The resulting mixture was concentrated. Thereafter, 200mL of water was added to quench the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate. The resulting mixture was washed with 2X 300mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was applied to a silica gel column (ethyl acetate: petroleum ether=0:1 to 1:2). The final product was 37.5g (85.66%) 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]-1-propylamine, yellow oil.
Synthesis of 4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] fourteen-1 (9), 2,5, 7-tetraene: 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] -1-propylamine (37.50G, 93.618 mmol,1 eq), toluene (500 mL), t-Buona (27.00G, 280.947mmol,3 eq.) and BrettPhos Pd G3 (4.25G, 4.688mmol,0.05 eq.) were added to a round bottom flask. The resulting solution was stirred at 110℃for 4h and the solid was filtered off. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 16.1g (53.81%) of 4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazacyclo [7.4.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraene are obtained as a brown solid. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0SUPELCO Ascentis Express C18,2.7 micrometers; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 4 ℃; flow rate: 1.5mL/min. LC-MS: (ES, M/z): M+1:320.
4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) 2- [) Ethoxy group]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: an inert atmosphere of nitrogen was introduced into a flask and maintained, and methyl-2-bromo-4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl) was added thereto]Methyl group]Piperazin-1-yl) benzoate (473.8 mg,2 eq), toluene (5 mL), 4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraene (150 mg,1 equivalent), cs 2 CO 3 (306.4 mg,2 eq.) and X-antphos G2 precatalyst (50 mg) were stirred at 110℃for 12h. The stirred solution was diluted with 10mL of water. The resulting solution was extracted with 4X 10mL ethyl acetate. The resulting mixture was washed with 20mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (1:5-1:1) on a silica gel column. The final product was 280mg (80.33%) of 4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate, as a pale yellow oil. LC-MS (ES, M/z): M+1:742.
4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid: 4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (280.00 mg,0.37mmol,1 eq.), methanol (3.00 mL,0.094mmol,0.25 eq.), dioxane (3.00 mL,35.41mmol,93.90 eq.) and sodium hydroxide (0.57 mL,2.262mmol,6 eq.) were added to a round bottom flask. The resulting solution was stirred at 70℃for 3h. The reaction mixture was cooled to room temperature. The resulting solution was treated with 10mLH 2 O extraction. The pH of the solution was adjusted to 5 with AcOH. The resulting solution was extracted with 4X 10mL ethyl acetate. The resulting mixture was washed with 20mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was purified on a silica gel column using ethyl acetateThe ethyl acetate/petroleum ether (1:2-2:1) is treated. 100mg (36.40%) of (4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid, brown solid. LC-MS (ES, M/z): M+1:728.
Synthesis of 4- ([ [ (2S) -1, 4-dioxane-2-yl ] methyl ] amino) -3-nitrobenzene-1-sulfonamide: 4-fluoro-3-nitrobenzene-1-sulfonamide (1.43 g, 0.0071 mmol,1 eq.), 1- [ (2S) -1, 4-dioxan-2-yl ] carboxamide hydrochloride (1 g,6.510mmol,1 eq.), THF (30 mL), cesium carbonate (8.48 g,0.026mmol,4 eq.) was charged to a round bottom flask. The resulting solution was stirred overnight in an oil bath at 50 ℃. The solid was collected by filtration. The solid was dried in an oven under reduced pressure. The final product was 1.82g (88.10%) 4- ([ [ (2S) -1, 4-dioxane-2-yl ] methyl ] amino) -3-nitrobenzene-1-sulfonamide as a yellow solid. LC-MS (ES, M/z): M+1:318.
4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide 4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (95.00 mg,0.130mmol,1 eq.), DCM (2 mL), DMAP (31.87 mg,0.261mmol,2 eq.), 4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]3-Nitrophenyl sulfonamide (49.60 mg,0.156mmol,1.20 eq.) and EDCI (30.00 mg,0.15mmol,1.2 eq.) were added to a round bottom flask. The resulting solution was stirred at 30℃for 12h. Then use 10mL H 2 O dilutes the resulting solution. The resulting solution was extracted with 4X 10mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 20mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with dichloroethane/methanol (95:5) on a silica gel column. 80mg (59.69%) of 4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide, yellow solid. LC-MS (ES, M/z): M+1:1027.
4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) - - (4- [ [ (2S) 2- (trimethylsilyl) ethoxy]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid: 4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (80.00 mg,0.078mmol,1 eq.), methanol (4 mL), dioxane (3 mL), sodium hydroxide (0.57 mL,2.262mmol,6 eq.) were added to a round bottom flask. The resulting solution was stirred at 70℃for 8h. The reaction mixture was cooled to room temperature. The resulting solution was treated with 10mLH 2 O extraction. The resulting solution was extracted with 4X 10mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 20mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with dichloroethane/methanol (95:5) on a silica gel column. The crude product (35 mg) was purified by Prep-HPLC under the following conditions: column (C18). Shimadzu LCMS 2020;50*3.0SUPELCO Ascentis Express C18,2.7 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 4 ℃; flow rate: 1.5mL/min gives 19mg (27.20%) of 4- (4- [ [2- (4-chlorophenyl) cyclohexyl-1-en-1-yl) ]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide, yellow solid. LC-MS (ES, M/z): M+1:897, 1 H-NMR:(300MHz,DMSO-d 6 ,ppm)δ11.94(s,1H),8.88–8.60(m,2H),8.45(s,1H),8.02(m,1H),7.95(m,1H),7.13(m,1H),7.09–6.93(m,3H),6.74(s,2H),6.20(s,1H),4.13–3.56(m,8H),3.60–3.39(m,2H),3.31(s,6H),2.84(s,2H),2.38(s,5H),2.19(d,J=30.5Hz,5H),1.73(s,5H)。
example 25
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
4- ([ [ (2S) -1, 4-dioxan-2-yl)]Methyl group]Synthesis of amino) -3-nitrobenzene-1-sulfonamide: 4-fluoro-3-nitrobenzene-1-sulfonamide (1.43 g,0.007mmol,1 eq.) 1- [ (2S) -1, 4-dioxane-2-yl]Carboxamide hydrochloride (1 g,6.510mmol,1 eq.), THF (30 mL), cs 2 CO 3 (8.48 g,0.026mmol,4 eq.) in a round bottom flask. The resulting solution was stirred overnight in an oil bath at 50 ℃. The solid was collected by filtration. The solid was dried in an oven under reduced pressure. 1.82g (88.10%) of 4- ([ [ (2S) -1, 4-dioxane-2-yl) are obtained ]Methyl group]Amino) -3-nitrobenzene-1-sulfonamide as a yellow solid. LC-MS (ES, M/z): M+1=554.
Synthesis of 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propyl ] isoindole-1, 3-dione: 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrole [2,3-b ] pyridine (53.50 g,154.944mmol,1 eq.) was placed in a three-necked round bottom flask, purged with nitrogen inert gas and maintained. 2- (3-hydroxypropyl) isoindole-1, 3-dione (31.80 g, 232.418 mmol,1.50 eq., 60%) was then added at 0deg.C. The resulting solution was stirred at 80℃for 4h and the reaction mixture was cooled with a water/ice bath. Then 500ml of LAcOH/ice water solution was added to stop the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate. The resulting mixture was washed with 3X 500mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:10) on a silica gel column. The final product was 58g (70.56%) 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propyl ] isoindole-1, 3-dione as a colorless oil. LC-MS (ES, M/z): M+1=554.
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) ]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Synthesis of propane-1-amine: 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Propyl group]Isoindole-1, 3-dione (58.00 g, 109.336 mmol,1 eq.), etOH (300.00 mL), NH 2 NH 2 .H 2 O (68.42 g,1093.321mmol,10 equiv., 80%) was charged to a round bottom flask. The resulting solution was stirred at room temperature for 4h. The resulting mixture was concentrated. Then 200mL of water was added to stop the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate. The resulting mixture was washed with 2X 300mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:2) on a silica gel column. 37.5g (85.66%) of 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) are obtained]Methyl group]Pyrrolo [2,3-b]Pyridin-6-yl) oxy]Propane-1-amine as a yellow oil.
Synthesis of 4[ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazacyclo [7.4.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraene: 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propan-1-amine (37.50G, 93.618 mmol,1 eq.) toluene (500.00 mL), t-BuONa (27.00G, 280.947mmol,3 eq.) BrettPhos Pd G3 (4.25G, 4.688mmol,0.05 eq.) was placed in a round bottom flask. The resulting solution was stirred at 110℃for 4h and the solid was filtered off. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 16.1g (53.81%) of 4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazacyclo [7.4.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraene are obtained as a brown solid. LC-MS (ES, M/z): M+1=320.
Synthesis of 2-bromocyclohepta-1-ene-1-carbaldehyde: DMF (8.15 g, 111.433 mmol,2.5 eq.) and DCM (100 mL) were added to a three-necked round bottom flask. Subsequently, PBr3 (27.75 g,102.521mmol,2.3 eq.) was added dropwise with stirring at 0deg.C. The resulting solution was stirred at 0℃for 1h. Adding at 0deg.C in portionsCycloheptanone (5.00 g,44.574mmol,1 eq.). The resulting solution was stirred at room temperature overnight. The reaction was then stopped by adding 100mL of ice water. With Na 2 CO 3 The pH of the solution was adjusted to 5. The resulting solution was extracted 3 times with 100mL of dichloromethane dried over anhydrous sodium sulfate and concentrated. 5g (55.24%) of 2-bromocyclohepta-1-en-1-carbaldehyde are obtained as a yellow oily liquid. LC-MS (ES, M/z): M+1=203/205.
Synthesis of 2- (4-chlorophenyl) cyclohepta-1-ene-1-carbaldehyde: 2-Bromocyclohepta-1-en-1-carbaldehyde (2.50 g,12.310mmol,1 eq), (4-chlorophenyl) boronic acid (1.92 g,12.279mmol,1 eq), dimethyl ether (25 mL), H 2 O(25mL),Na 2 CO 3 (2.61 g,24.621mmol,2 eq.) Pd (pph) 3 ) 2 Cl 2 (1.01 g,1.231mmol,0.1 eq.) was placed in a round bottom flask. The resulting solution was stirred at 60℃for 12h. The reaction mixture was cooled to room temperature. Then 50mL of water was added to stop the reaction. The resulting solution was extracted with 3X 50mL ethyl acetate. The resulting mixture was washed with 3X 50mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 0.6g (20.76%) 2- (4-chlorophenyl) cyclohepta-1-ene-1-carbaldehyde was obtained as a yellow oily liquid. LC-MS: (ES, M/z) m+1=235.
2-bromo-4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl)]Methyl group]Synthesis of methyl piperazin-1-yl) benzoate: 2- (4-chlorophenyl) cyclohepta-1-ene-1-carbaldehyde (0.60 g, 2.552 mmol,1 eq.), methyl 2-bromo-4- (piperazin-1-yl) benzoate (0.76 g,2.540mmol,0.99 eq.), DCM (30.00 mL,471.901mmol,184.61 eq.), ti (Oi-Pr) 4 (2.18 g,7.669mmol,3 eq.) was placed in a round bottom flask. NaBH (OAc) was then added in portions at room temperature 3 (1.08 g,5.112mmol,2 eq.). The resulting solution was stirred at room temperature overnight. Then 10mL of water was added to stop the reaction. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 500mg (37.77%) of 2-bromo-4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) methyl benzoate, an off-white oily liquid. LC-MS (ES, M/z): M+1=517/519.
4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: 4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group ]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradeca-1 (9), 2,5, 7-tetraene (154.22 mg,0.483mmol,1 eq.) 2-bromo-4- (4- (4-chlorophenyl) cyclohepta-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl benzoate (250.00 mg,0.483mmol,1 eq.) Cs 2 CO 3 (471.85 mg, 1.4478 mmol,3 eq.) toluene (5.00 mL) and Xantphos Pd 2G (46.00 mg,0.1 eq.) were added to a round bottom flask. The resulting solution was stirred at 110 ℃ for 11h until LCMS showed complete consumption of material. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 110mg (30.12%) of (4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate, yellow solid. LC-MS (ES, M/z): M+1=756.
(4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid: methyl 4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl)]Methyl group ]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (110.00 mg,0.145mmol,1 eq.), meOH (1 mL), dioxane (1 mL), H 2 O (1 mL), naOH (34.90 mg) was added to a round bottom flask. The resulting solution was stirred at 70℃for 12h, then 5mL of water was added to stop the reaction. The pH of the solution was adjusted to 5 with hydrochloric acid (1 mol/L). The solid was collected by filtration. 100mg (92.63%) of (4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid, white crude solid. LC-MS (ES),m/z):M+1=742。
Synthesis of 4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxa-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzamide: 4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl ] methyl ] piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid (100.00 mg,0.135mmol,1 eq.), DCM (5 mL), DMAP (65.82 mg, 0.39 mmol,4 eq.), EDCI (51.64 mg,0.269mmol,2 eq.), 4- [ (2S) -1, 4-dioxane-2-ylmethyl ] amino ] -3-nitrobenzenesulfonamide (47.01 mg,0.148mmol,1.1 eq.) was added to a round bottom flask. The resulting solution was stirred at room temperature overnight. The resulting mixture was concentrated. The residue was treated with dichloroethane/methanol (1:0 to 10:1) on a silica gel column. 120mg (85.52%) of 4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy ] methyl ] -14-oxa-2, 4, 10-triaza-tricyclo [7.5.0.0 [3,7] ] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzamide are obtained as a yellow crude solid. LC-MS (ES, M/z): M+1=1041.
4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide: 4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide (120.00 mg,0.115mmol,1 eq.), ethylenediamine (138.46 mg,2.304mmol,20 eq.), THF (10 mL), TBAF (602.37 mg,2.304mmol,20 eq.) were added to a round bottom flask. The resulting solution was stirred at 70℃for 12h, then 10mL of water was added to reverseShould stop. The resulting solution was extracted with 2X 10mL ethyl acetate and concentrated. The crude product was purified by Prep-HPLC under the following conditions (Waters-2767): column, X-bridge RP18,5 μm, 19X 100mm; mobile phase, 0.03% ammonia (0.03% NH) 4 HCO 3 And NH 4 OH) and CH 3 CN(32%CH 3 CN up to 52% in 6 min); detector, UV 254nm. 26mg (24.76%) of 4- (4- [ [2- (4-chlorophenyl) cyclohepta-1-en-1-yl) are obtained ]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide solid. LC-MS (ES, M/z): M+1=911. 1 H NMR(300MHz,DMSO-d 6 )δ11.90(s,1H),11.20(s,1H),8.46(s,2H),7.65(dd,J=9.1,1.8Hz,2H),7.45(s,1H),7.36(d,J=8.3Hz,4H),7.20(s,2H),7.08(d,J=8.4Hz,4H),6.97–6.86(m,4H),6.70(s,2H),6.12(d,J=2.5Hz,2H),4.21(s,3H),3.86–3.72(m,6H),3.71–3.41(m,13H),3.20(s,4H),2.76(s,4H),2.39(s,7H),2.28(s,7H),1.98(s,2H),1.57(s,3H),1.50(s,3H),1.24(s,1H)。
Example 26
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl ] methyl ] piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ] amino ] -3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7] tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
2-bromo-4- (4- [ [2- (4-chlorophenyl) 5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Synthesis of methyl piperazin-1-yl) benzoate: in a three neck round bottom flask was charged methyl 2-bromo-4- (piperazin-1-yl) benzoate (2.00 g,6.68mmol,1 eq), DCE (20 mL), 2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-carbaldehyde (1.77 g,6.685mmol,1 eq), ti (Oi-Pr) 4 (5.70 g,20.05mmol,3 eq.) the resulting solution was stirred for 3h. The obtained mixture is divided intoBatch addition of NaBH (OAc) 3 (2.83 g,13.37mmol,2 eq.). The resulting solution was allowed to stand at room temperature for further reaction for 16h. Then 10mL of water was added to stop the reaction. The resulting mixture was concentrated. The residue was applied to a column containing silica gel (ethyl acetate: petroleum ether=1:10 to 2:5). 3.0g (81.90%) of 2-bromo-4- (4- [ [2- (4-chlorophenyl) 5-methoxy-5-methylcyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) methyl benzoate, a pale yellow oily liquid. LC-MS (ES, m/z): 547[ M+H ]] +
Synthesis of 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propyl ] isoindole-1, 3-dione: 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridine (53.50 g,154.944mmol,1 eq.) was placed in a round bottom flask and nitrogen was purged to maintain an inert atmosphere. Subsequently, 2- (3-hydroxypropyl) isoindole-1, 3-dione (31.80 g,154.944mmol,1 eq.) was added in portions at room temperature. Dioxane (500 mL) and NaH (9.30 g, 232.418 mmol,1.50 eq., 60%) were added at room temperature. The resulting solution was stirred at 80℃for 4h and the reaction mixture was cooled with an ice bath. Then, 500ml of ice water solution of lacoh was added to stop the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate. The resulting mixture was washed with 3X 500mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:10) on a silica gel column. 58g (70.56%) of 2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] pyrrolo [2,3-b ] pyridin-6-yl) oxy ] propyl ] isoindole-1, 3-dione are obtained as a colorless oil. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0SUPELCO Ascentis Express C18,2.7 micrometers; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 4 ℃; flow rate: 1.5mL/min. LC-MS: (ES, M/z): M+1:554.
3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Synthesis of propane-1-amine: adding ethoxy groups to a round bottom flask]Methyl group]Propyl group]Isoindolopyrroles [2,3-b ]]Pyridin-6-yl) oxy]2- [3- [ (5-bromo-1- [ [2- (trimethylsilyl) 1, 3-dione (58.00)g,109.33mmol,1 eq), etOH (300 mL), NH 2 NH 2 .H 2 O (68.42 g,1093.32mmol,10 eq., 80%). The resulting solution was stirred at room temperature for 4h. The resulting mixture was concentrated. Then 200mL of water was added to stop the reaction. The resulting solution was extracted with 2X 500mL ethyl acetate and the resulting mixture was washed with 2X 300mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:2) on a silica gel column. 37.5g (85.66%) of 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy) are obtained]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Propane-1-amine, yellow oily liquid.
4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]-synthesis of tetradec-1 (9), 2,5, 7-tetraene: 3- [ (5-bromo-1- [ [2- (trimethylsilyl) ethoxy)]Methyl group]Pyrrole [2,3-b ]]Pyridin-6-yl) oxy]Propane-1-amine (37.50G, 93.618 mmol,1 eq.), toluene (500 mL), t-Buona (27.00G, 280.947mmol,3 eq.), brettPhos Pd G 3 (4.25 g,4.688mmol,0.05 eq.) was added to a round bottom flask. The resulting solution was stirred at 110℃for 4h and the solid was filtered off. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1 to 1:1) on a silica gel column. 16.1g (53.81%) of 4- [ [2- (trimethylsilyl) ethoxy ] are obtained]Methyl group]-14-oxa-2, 4, 10-triazacyclo [7.4.0.0 ] [3,7 ],]]tridec-1 (9), 2,5, 7-tetraene, is a brown solid. The residence time measurement was performed using a reverse phase column (C18). Shimadzu LCMS 2020;50*3.0SUPELCO Ascentis Express C18,2.7 μm; eluent a: water (0.05% tfa); eluent B: acetonitrile; linear gradient from 5% acetonitrile to 95% acetonitrile, 7.0min; the temperature of the oven is 4 ℃; flow rate: 1.5mL/min. LC-MS: (ES, M/z): M+1:320.
4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate: 2-bromo-4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Methyl piperazin-1-yl benzoate (515 mg,0.940mmol,1 eq.) methyl formate Benzene (5 mg), 4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0 ] [3,7 ]]]Tetradecane-1 (9), 2,5, 7-tetraene (150.14 mg,0.47mmol,0.50 eq.) Cs 2 CO 3 (306.24 mg,0.94mmol,1 eq.) and X-antphos Pd G3 (50.00 mg,0.05mmol,0.06 eq.) were added to a round bottom flask. The resulting solution was stirred at 110℃for 12h. The resulting solution was treated with 10mLH 2 O extraction. The resulting solution was extracted with 4X 10mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 20mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (1:5-1:1) on a silica gel column. 230mg (62.29%) of (4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate, as a pale yellow oily liquid. LC-MS (ES, m/z): 786[ M+H ]] +
4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Synthesis of tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid: 4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Methyl tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoate (230 mg,0.29mmol,1 eq.), methanol (3 mL), dioxane (3 mL), 4M NaOH (0.44 mL,1.75mmol,6 eq.) were added to a vial. The resulting solution was stirred at 70℃for 3h. The reaction mixture was cooled to room temperature. The resulting solution was treated with 10mLH 2 O extraction. The pH of the solution was adjusted to 5 with HOAc. The resulting solution was extracted with 4X 10mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 20mL brine. The mixture was dried over anhydrous sodium sulfate. The residue was treated with ethyl acetate/petroleum ether (1:2-2:1) on a silica gel column. 150mg (66.40%) of 4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxygenHetero-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid, brown solid. LC-MS (ES, m/z): 772[ M+H ] ] +
4- ([ [ (2S) -1, 4-dioxan-2-yl)]Methyl group]Synthesis of amino) -3-nitrobenzene-1-sulfonamide: 4-fluoro-3-nitrobenzene-1-sulfonamide (1.43 g,0.007mmol,1 eq.) 1- [ (2S) -1, 4-dioxane-2-yl]Carboxamide hydrochloride (1 g,6.510mmol,1 eq.), THF (30 mL), cs 2 CO 3 (8.48 g,0.026mmol,4 eq.) in a round bottom flask. The resulting solution was stirred overnight in an oil bath at 50 ℃. The solid was collected by filtration. The solid was dried in an oven under reduced pressure. The final product was 1.82g (88.10%) 4- ([ [ (2S) -1, 4-dioxane-2-yl)]Methyl group]Amino) -3-nitrobenzene-1-sulfonamide, yellow solid. LC-MS (ES, M/z): M+1:318.
4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide: 4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ] ]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzoic acid (140.00 mg,0.18mmol,1 eq.), DCM (2.00 mL), DMAP (44.28 mg,0.36mmol,2 eq.), 4 [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]3-Nitrophenyl sulfonamide (69.00 mg,0.21mmol,1.20 eq.) and EDCI (41.8 mg,0.21mmol,1.20 eq.) were added to a vial. The resulting solution was stirred at 30℃for 12h. The resulting solution was treated with 10mLH 2 O extraction. The resulting solution was extracted with 4X 10mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 20mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The solid residue was loaded onto preparative thin layer chromatography (dichloromethane: methanol=95:5). 135mg (69.50%) of 4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [[2- (trimethylsilyl) ethoxy ]]Methyl group]-14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide, yellow solid. LC-MS (ES, m/z): 1071[ M+H ]] +
4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl ]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Synthesis of tetradeca-1 (9), 2,5, 7-tetraen-10-yl) benzamide 4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl) was added to a vial]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- (4- [ [2- (trimethylsilyl) ethoxy)]Methyl group]- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7 ]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide (135 mg,0.125mmol,1 eq.) THF (4 mL), ethylenediamine (150.00 mg,2.50mmol,20 eq.) 1M TBAF in THF (2.50 mL,2.50mmol,20 eq.). The resulting solution was stirred at 70℃for 8h. The reaction mixture was cooled to room temperature. The resulting solution was treated with 10mLH 2 O extraction. The resulting solution was extracted with 4X 10mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 20mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with dichloroethane/methanol (95:5) on a silica gel column. The crude product (50 mg) was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): a chromatographic column, a C18 silica gel column; mobile phase, CH 3 CN:H 2 O(0.05NH 3 .H 2 O) =10% to CH 3 CN:H 2 O(0.05NH 3 .H 2 O) =50%, within 6 min; detector, UV 220nm. 23mg (25.30%) of 4- (4- [ [2- (4-chlorophenyl) -5-methoxy-5-methylcyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -N- (4- [ [ (2S) -1, 4-dioxan-2-ylmethyl]Amino group]-3-nitrobenzenesulfonyl) -2- [ 14-oxa-2, 4, 10-triazatricyclo [7.5.0.0 ] [3,7]]]Tetradec-1 (9), 2,5, 7-tetraen-10-yl) benzamide, yellow solid. LC-MS (ES, m/z): 941[ M+H ]] + 1 H NMR(300MHz,DMSO,ppm):δ11.93(s,1H),8.81(s,1H),8.70(s,1H),8.45(s,1H),7.98(m,2H),7.31(s,1H),7.13(s,1H),7.08–6.94(m,3H),6.74(s,2H),6.66(m,1H),6.20(s,1H),4.08–3.54(m,8H),3.47(m,2H),3.29(s,8H),2.84(s,2H),2.61–2.04(m,10H),1.89(m,1H),1.80–1.63(m,2H),1.28(s,3H)。
Example 27
The Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, and the preparation method and the application of the composition are as follows:
4- (4- [ [2- (4-chloro-3-fluorophenyl) -4, 4-dimethylcyclohex-1-en-1-yl ] methyl ] piperazin-1-yl) -2- [ (12R) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraen-10-yl ] -N- [ 3-nitro-4- [ (oxa-4-ylmethyl) amino ] benzenesulfonyl ] benzamide
The preparation method of the Bcl-2/Bcl-xl inhibitor comprises the following steps:
(E) -synthesis of 2- (2-ethoxyvinyl) -4, 5-tetramethyl-1, 3, 2-dioxaborane: in a one round bottom flask, ethyl vinyl ether (1000.00 g,13.9mmol,3.50 eq.) and 4, 5-tetramethyl-1, 3, 2-dioxaborane (508.00 g,3.97mmol,1 eq.) were placed and the solution cooled to 5℃in an ice/salt bath. Then adding an appropriate amount of Pd (OAc) at 5 ℃ 2 (50.00 g,222mmol,0.02 eq.). The resulting solution was stirred at room temperature for 18h. The resulting solution was diluted with 5L PE. The resulting mixture was concentrated. 1100g (crude product, Y=50%) of 2- [ (E) -2-ethoxyvinyl are obtained]-4, 5-tetramethyl-1, 3-dioxolane, yellow oily liquid. LC-MS (ES, M/z): M+1=199; H-NMR (300 MHz, DMSO-d) 6 ,ppm)δ6.91–6.95(m,1H),4.30–4.34(m,1H),3.78–3.82(m,2H),1.14–1.24(m,15H)。
Synthesis of N- [ (2R) -2-hydroxypropyl ] acetamide: purged with nitrogen and charged to a three-necked flask were (2R) -1-aminopropane-2-ol (100 g,1.3mol,1 eq), DCM (100 mL) and TEA (160 g,1.6mol,1.2 eq). A solution of acetoacetate (136 g,1.3mol,1 eq.) in 100mL (DCM) was then added dropwise at 0-10deg.C. The resulting solution was stirred at room temperature for 14h. The resulting mixture was concentrated. The residue was treated with dichloroethane/methanol (100:5) on a silica gel column. 250g (55.24%) of N- [ (2R) -2-hydroxypropyl ] acetamide are obtained as a yellow oily liquid.
Synthesis of 5-bromo-6-fluoropyridin-2-amine: into a four-necked round bottom flask was charged 6-fluoropyridin-2-amine (4500.00 g,40.178mol,1 eq.) ACN (20L). Then NBS (7035.17 g,41.383mol,1.03 eq.) was added as part of the solution at room temperature. The resulting solution was stirred at room temperature for 2h. The obtained solution was treated with 40LH 2 O dilution. The resulting solution was extracted with 2X 36L ethyl acetate. The resulting mixture was washed with 10L of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The resulting mixture was washed with 3X 9L PE. The mixture was dried in an oven to give 7634g (y=90%) of 5-bromo-6-fluoropyridin-2-amine as a light brown solid. LC-MS (ES, M/z): M+1=190; H-NMR (300 MHz, DMSO-d) 6 ,ppm)δ7.63–7.71(m,1H),6.56(s,2H),6.30–631(m,1H)。
Synthesis of 5-bromo-6-fluoro-3-iodopyridin-2-amine: 5-bromo-6-fluoropyridin-2-amine (7000.00 g,36.842mol,1 eq.) and AcOH (34L) were added to a four-necked round bottom flask. The solution was cooled to 10 ℃ in a water/ice bath. An appropriate amount of NIS (8290 g,36.8mol,1 eq.) was then added at 10deg.C. The resulting solution was stirred at room temperature for 2h. The resulting solution was diluted with 100L of water. The mixture was filtered and the filter cake was collected, washed with water (35 x 2L) and then dried through an oven to give 9840g (y=90%) of 5-bromo-6-fluoro-3-iodopyridin-2-amine as a brown solid. LC-MS (ES, M/z): M+1=317; H-NMR (300 MHz, DMSO-d) 6 ,ppm)δ8.16–8.23(m,1H),6.69(s,2H)。
Synthesis of 5-bromo-3- (2-ethoxyvinyl) -6-fluoropyridin-2-amine: 5-bromo-6-fluoro-3-iodopyridin-2-amine (855.00 g,2705.696mmol,1 eq.) i-PrOH (10000 mL), 2- [ (E) -2-ethoxyvinyl]-4, 5-tetramethyl-1, 3-dioxolane (1000.00 g,5050.505mmol,1.87 eq.) K 3 PO 4 (1720.00 g,8113.207mmol,3 eq.), ruphos (12.00 g,27.060mmol,0.02 eq.), pd (OAc) 2 (20.00 g,88mmol,0.02 eq.) was charged to a four-necked round bottom flask purged with nitrogen inert gas and maintained. The resulting solution was stirred in a liquid nitrogen bath at room temperature for 12h. The solid was filtered. The filter cake was washed with 3X 2L DCM. The organic layer was collected and concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:5) on a silica gel column. After concentration 680g (80%) of 5-bromo-3- [ are produced(E, Z) -2-ethoxyvinyl]-6-fluoropyridin-2-amine (Z, E mixture), dark brown oily liquid. LC-MS (ES, M/z) M+1=261; H-NMR (300 MHz, DMSO-d) 6 ,ppm)δ7.96–7.99(m,1H),6.72–6.76(m,1H),5.49–5.53(m,1H),3.99–4.06(m,3H)。
5- (tert-butoxy) -N- (diphenylmethylene) -1H-pyrrole [2,3-b]Synthesis of pyridin-5-amine: 5-bromo-3- [ (E) -2-ethoxyvinyl]6-Fluoropyridin-2-amine (680.00 g, 2605.264 mmol,1 eq), etOH (5000 mL), HCl (1000 mL) were placed in a 10L round bottom flask and nitrogen was purged to maintain an inert atmosphere. The resulting solution was stirred at room temperature for 5h. The resulting mixture was concentrated. The pH of the solution was adjusted to 6 with NaOH (4 mol/L). The solid was collected by filtration and washed with 3X 500mL of water. 560g (Q-NMR=70%) of 5-bromo-6-fluoro-1H-pyrrole [2,3-b ] are obtained ]Pyridine, light brown solid. LC-MS (ES, M/z): M+1=215; H-NMR (300 MHz, DMSO-d) 6 ,ppm)δ9.53(brs,1H),8.19–8.22(d,J=9.0Hz,1H),7.32–7.34(m,1H),6.50–6.52(m,1H)。
5-bromo-6-fluoro-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrole [2,3-b]Synthesis of pyridine: 5-bromo-6-fluoro-1H-pyrrole [2,3-b ]]Pyridine (560.00 g,2.6mmol,1 eq.) and DMF (5000 mL) were placed in a 10L four-necked round bottom flask. The solution was cooled to 0 ℃ in a water/ice bath. NaH (156 g,3.9mmol,1. Eq.) was then added in portions at 0deg.C. SEM-Cl (561 g,3.38mmol,1.3 eq.) was added dropwise with stirring at 0deg.C. The resulting solution was stirred at room temperature for 1h. The reaction was then stopped by adding 2000mL of ice water. The resulting solution was diluted with 5L of water. The resulting solution was extracted with 2X 10L ethyl acetate and the organic layers were combined. The resulting mixture was washed with 3X 5L of water. The resulting mixture was washed with 3L brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was eluted with ethyl acetate/petroleum ether (1:20) on a silica gel column. 550g (87%) of 5-bromo-6-fluoro-1- [ [2- (trimethylsilyl) ethoxy ] are obtained]Methyl group]Pyrrolo [2,3-b]Pyridine was a pale yellow oil. LC-MS (ES, M/z) m+1=345; H-NMR (300 MHz, DMSO-d) 6 ,ppm)δ8.46–8.49(d,J=9.0Hz,1H),7.68–7.69(m,1H),6.57–6.58(m,1H),5.52–5.55(m,2H),3.47–3.60(m,2H),0.79–0.90(m,2H),0.01(s,9H)。
(R) -N- (2- (5-bromo)-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrolidine [2,3-b ]Pyridin-6-oxy) propyl) acetamide synthesis: n- [ (2R) -2-hydroxypropyl]Acetamide (crude, 81.6g,697mmol,3 eq.) and dioxane (800 mL) were placed in a round bottom flask. NaH (28 g,697mmol,3 eq.) was then added at 5-15 ℃. The resulting solution was stirred at room temperature for 30min. To this was added 5-bromo-6-fluoro-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrolo [2,3-b ] in dioxane (100 mL)]Pyridine (80 g,232mmol,1 eq.) solution. The resulting solution was stirred in an oil bath at 80℃for 4h. The reaction was then inhibited by adding 50mL of water at 10 ℃. The resulting solution was concentrated and taken up with 500mL H 2 O dilution. The resulting solution was extracted with 3X 500mL ethyl acetate. The resulting mixture was washed with 1X 300mL of aqueous sodium chloride solution. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with PE/EA (100:20) on a silica gel column. 40g (pure) and 30g (70%) of (R) -N- (2- (5-bromo-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrolidine [2,3-b were obtained]Pyridin-6-yloxy) propyl) acetamide, yellow oily liquid.
1- [ (12R) -4- (trimethylsilyl) ethoxy) methyl) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7]]Tridec-1 (9), 2,5, 7-tetraen-10-yl ]Synthesis of ethane-1-one: (R) -N- (2- (5-bromo-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrrole [2, 3-b)]Pyridin-6-yloxy) propyl acetamide (40 g,90.7mmol,1 eq), dioxane (500 mL), cs 2 CO 3 (88G, 272mmol,3 eq.) and BrettPhos Pd G3 (4.1G, 4.5mmol,0.05 eq.) were placed in a round bottom flask purged with an inert atmosphere of nitrogen and maintained. The resulting solution was concentrated and taken up with 300mL H 2 O dilution. The resulting solution was extracted with 3X 300mL ethyl acetate. The resulting mixture was washed with 1X 200mL brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (40:100) on a silica gel column. 33g (crude, 70%) of 1- [ (12R) -4- (trimethylsilyl) ethoxy) methyl) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 are obtained]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]Ethane-1-one, brown oily liquid.
Synthesis of (12R) -4- (trimethylsilyl) ethoxy ] methyl) -12-2H 4) -13-oxa-2, 4, 10-triazacyclo [7.4.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraene: 1- [ (12R) -4- (trimethylsilyl) ethoxy) methyl) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 [3,7] ] trideca-1 (9), 2,5, 7-tetraen-10-yl ] ethan-1-one (33 g), methanol (300 mL), sodium hydroxide (2M, 300 mL) were placed in a 1L reaction flask. The resulting solution was stirred in an oil bath at 80 ℃ for 6h. The resulting solution was concentrated and methanol was removed, then extracted with 3X 200mL ethyl acetate. The resulting mixture was washed with 200mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (1:1) on a silica gel column. 16.2g of (12R) -4- (trimethylsilyl) ethoxy) methyl) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 [3,7] ] tridec-1 (9), 2,5, 7-tetraenone were obtained as a brown oily liquid.
2-bromo-4- [4- (tert-butoxycarbonyl) piperazin-1-yl]Synthesis of benzoic acid: in a three neck round bottom flask, 4- [ 3-bromo-4- (methoxycarbonyl) phenyl ] is added]Piperazine-1-carboxylic acid tert-butyl ester (4.00 g,10.0mmol,1 eq.), methanol (20 mL), THF (20 mL), H 2 O (20 mL) and NaOH (1.60 g,40.0mmol,4 eq.). The resulting solution was stirred at 30℃for 3h. With 40mL H 2 O dilutes the resulting solution. The pH of the solution was adjusted to 5 with hydrochloric acid (0.5 mol/L). The resulting solution was extracted with 3X 30mL ethyl acetate and the organic layers were combined. The resulting mixture was washed with 1X 50mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. 3.8g (98.45%) of 2-bromo-4- [4- (tert-butoxycarbonyl) piperazin-1-yl are obtained]Benzoic acid, white solid. LC-MS (ES, M/z): M+1:385.
4- [ 3-bromo-4- ([ 3-nitroso-4- [ (pyrrolidin-4-ylmethyl) amino)]Benzenesulfonyl group]Carbamoyl) phenyl group]Synthesis of piperazine-1-carboxylic acid tert-butyl ester hydrate: 2-bromo-4- [4- (tert-Butoxycarbonyl) piperazin-1-yl]Benzoic acid (3.80 g,9.864mmol,1 eq.) 3-nitro-4- [ (pyrrolidin-4-ylmethyl) amino]Benzenesulfonamide (3.11 g,9.864mmol,1 eq.), EDCI (2.27 g,11.836mmol,1.2 eq.), DMAP (2.41 g,19.727mmol,2 eq.) and DCM (40 mL) were placed in a three-necked round bottom flask. The resulting solution was stirred at 30℃for 12h. With 50mL H 2 O dilutes the resulting solution. The resulting solution was extracted with 3X 50mL of DCM and the organics were combinedA layer. The resulting mixture was washed with 50mL of brine. The mixture was dried over anhydrous sodium sulfate and concentrated. The residue was treated with dichloroethane/methanol (90:10) on a silica gel column. 5.4g (79.73%) of 4- [ 3-bromo-4- ([ 3-nitroso-4- [ (pyrrolidin-4-ylmethyl) amino are obtained]Benzenesulfonyl group]Carbamoyl) phenyl]Piperazine-1-carboxylic acid tert-butyl ester hydrate, yellow solid. LC-MS (ES, M/z): M+1:682.
4- [3- [ (12R) -12-methyl-4- [ [2- (trimethylsilyl) ethoxy ] ethyl group]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-4- ([ 3-nitroso-4- [ (oxazolidin-4-ylmethyl) amino)]Benzenesulfonyl group]Carbamoyl) phenyl]Synthesis of piperazine-1-carboxylic acid tert-butyl ester hydrate: in 4- [ 3-bromo-4- ([ 3-nitroso-4- [ (oxazol-4-ylmethyl) amino group]Benzenesulfonyl group]Carbamoyl) phenyl]Piperazine-1-carboxylic acid tert-butyl ester hydrate (4.00 g,5.860mmol,1 eq), (12R) -12-methyl-4- [ [2- (trimethylsilyl) ethoxy ]]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraene (1.87 g,5.860mmol,1 eq.), DMF (60 mL), cuI (0.22 g,1.172mmol,0.20 eq.), cs 2 CO 3 (3.82 g,11.720mmol,2 eq.) N1, N2 bis (4-hydroxy-2, 6-dimethylphenyl) oxamide (0.58 g,1.766mmol,0.30 eq.) was placed in a round bottom flask which was purged and maintained with an inert atmosphere of nitrogen. The resulting solution was stirred at 100℃for 2h. The reaction mixture was cooled to room temperature with a water bath. The resulting solution was treated with 100mLH 2 O extraction. The resulting solution was extracted with 3X 100mL ethyl acetate, the organic layers were combined, dried over anhydrous sodium sulfate and concentrated. The residue was treated with ethyl acetate/petroleum ether (1:3-1:0) on a silica gel column. 3.2g (59.02%) of 4- [3- [ (12R) -12-methyl-4- [ [2- (trimethylsilyl) ethoxy ] were obtained]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-4- ([ 3-nitroso-4- [ (oxazolidin-4-ylmethyl) amino)]Benzenesulfonyl group]Carbamoyl) phenyl]Piperazine-1-carboxylic acid tert-butyl ester hydrate, yellow solid. LC-MS (ES, M/z): M+1:921.
2- [ (12R) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-N- [ 3-nitro-4- [ (oxazolidin-4-ylmethyl) amino group]Benzenesulfonyl group]-4-Synthesis of (piperazin-1-yl) benzamide 4- [3- [ (12R) -12-methyl-4- [ [2- (trimethylsilyl) ethoxy ] ethoxy ]Methyl group]-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-4- ([ 3-nitro-4- [ (oxazolidin-4-ylmethyl) amino group]Benzenesulfonyl group]Carbamoyl) phenyl group]Piperazine-1-carboxylic acid tert-butyl ester (2.40 g,2.605mmol,1 eq), DCM (30 mL), TFA (10 mL) were placed in a round bottom flask and the resulting solution was stirred at room temperature for 4h. The resulting mixture was concentrated. Adding CH 3 CN (30.00 mL), ethylenediamine (0.79 g,13.145mmol,5.05 eq.). The resulting solution was reacted at 60℃for 6 hours. The resulting mixture was concentrated. The crude product (3.0 g) was purified by Flash-Prep-HPLC under the following conditions (CombiFlash-1): a chromatographic column, a C18 silica gel column; mobile phase, CH 3 CN:H 2 O (0.5% fa) =5% increases to CH in 7min 3 CN:H 2 O (0.5% fa) =60; detector, UV 254nm. 600mg (33.34%) of 2- [ (12R) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ] are obtained]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-N- [ 3-nitro-4- [ (oxazolidin-4-ylmethyl) amino group]Benzenesulfonyl group]-4- (piperazin-1-yl) benzamide, yellow solid. LC-MS (ES, M/z): M+1:691.
Synthesis of 2-bromo-1, 4-trimethylcyclohexyl-1-ene hydrate: CHCl was added to a three-necked flask 3 (100.00 mL), DMF (7.24 g). Followed by dropwise addition of PBr with stirring 3 (24.65 g,91.065mmol,2.30 eq.). The resulting solution was stirred at room temperature for 1h. Then cyclohexanone, 3-dimethyl- (5.00 g,39.620mmol,1 eq.) are added dropwise with stirring at 0 ℃. The resulting solution was allowed to react at room temperature for 12h. The reaction was then placed in 200mL ice water. With saturated Na 2 CO 3 The pH of the solution was adjusted to 5. The resulting solution was extracted with 3X 100mL of dichloromethane and the organic layers were combined, then dried over anhydrous sodium sulfate and concentrated. 3.1g (35.38%) of 2-bromo-1, 4-trimethylcyclohexyl-1-ene hydrate were obtained as a colorless oily liquid. LC-MS: (ES, M/z): M+1:217/219.
Synthesis of 2- (4-chloro-3-fluorophenyl) -4, 4-dimethylcyclohexyl-1-ene-1-carbaldehyde: 2-bromo-4, 4-dimethylcyclohexyl-1-ene-1-carbaldehyde (300.00 mg,1.382mmol,1 eq.) and 4-chloro-3-fluorobenzeneboronic acid (289.12 mg, 1)238 mmol,1.2 eq), DME (4 mL), na 2 CO 3 (292.91 mg,2.764mmol,2 eq.) Pd (PPh) 3 ) 2 Cl 2 (48.49 mg,0.069mmol,0.05 eq.) H 2 O (0.20 mL) was placed in a vial purged with inert nitrogen and maintained. The resulting solution was stirred at 100℃for 12h. The reaction mixture was then cooled to room temperature. The resulting mixture was concentrated. The residue was treated with ethyl acetate/petroleum ether (0:1-1:6) on a silica gel column. 170mg (46.12%) of 2- (4-chloro-3-fluorobenzene J-yl) -4, 4-dimethylcyclohexyl-1-en-1-carbaldehyde were obtained as a colorless oily liquid. LC-MS: (ES, M/z): M+1:267.
4- (4- [ [2- (4-chloro-3-fluorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl)]Methyl group]Piperazin-1-yl) -2- [ (12R) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-N- [ 3-nitro-4- [ (oxazolidin-4-ylmethyl) amino group]Benzenesulfonyl group]Synthesis of benzamide: to a vial was added 2- (4-chloro-3-fluorophenyl) -4, 4-dimethylcyclohexyl-1-ene-1-carbaldehyde (55.00 mg,0.21mmol,1 eq.) and [ (12R) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]]]Tridec-1 (9), 2,5, 7-tetraen-10-yl]-N- [ 3-nitro-4- [ (oxazolidin-4-ylmethyl) amino group]Benzenesulfonyl group]-4- (piperazin-1-yl) benzamide (142.43 mg,0.206mmol,1 eq.), meOH (1.50 mL), DCM (1.50 mL), znCl 2 (56.21 mg,0.412mmol,2 eq.) NaBH 3 CN (25.92 mg,0.412mmol,2 eq.). The resulting solution was stirred at 60℃for 5h. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated. The solid residue was loaded onto preparative thin layer chromatography (dichloromethane: methanol=95:5). The crude product (50 mg) was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): a chromatographic column, a C18 silica gel column; mobile phase, CH 3 CN:H 2 O(0.05%NH 3 .H 2 O) =10% to CH in 7min 3 CN:H 2 O(0.05%NH 3 .H 2 O) =60%; detector, UV 254nm. 6.0mg (3.09%) of 4- (4- [ [2- (4-chloro-3-fluorophenyl) -4, 4-dimethylcyclohexyl-1-en-1-yl) are obtained]Methyl group]Piperazin-1-yl) -2- [ (12R) -12-methyl-13-oxa-2, 4, 10-triazatricyclo [7.4.0.0 ] [3,7 ]][ tridec-1 (9), 2,5, 7-tetraen-10-yl]N- [ 3-nitro-)4- [ (oxan-4-ylmethyl) amino group]Benzenesulfonyl group]Benzamide, yellow solid. LC-MS (ES, M/z): M+1:941. 1 H-NMR(300MHz,DMSO-d6,ppm)δ10.89(s,1H),8.48–8.29(m,2H),7.51(dd,J=8.6,7.1Hz,2H),7.33(s,1H),7.11(dd,J=10.3,1.8Hz,1H),7.00(t,J=2.8Hz,1H),6.92(dd,J=8.2,1.9Hz,1H),6.69(d,J=24.5Hz,4H),5.99(s,1H),4.41(s,1H),3.86(dd,J=11.5,4.1Hz,2H),3.29-3.17(m,3H),3.10(d,J=11.7Hz,6H),2.77(s,2H),2.21(d,J=19.8Hz,6H),1.99(s,2H),1.89(d,J=12.7Hz,1H),1.63(d,J=13.0Hz,2H),1.40(t,J=6.2Hz,2H),1.35–1.10(m,6H),0.94(s,6H)。
Example 28
The Bcl-2/Bcl-xl inhibitor, the Bcl-2/Bcl-xl inhibitor-containing composition, the preparation method and the application of the Bcl-2/Bcl-xl inhibitor are one embodiment, the Bcl-2/Bcl-xl inhibitor adopts the compound synthesized in the embodiment 9, and the Bcl-2/Bcl-xl inhibitor-containing composition is obtained by combining the compound with DNA alkylating agent cisplatin.
Example 29
The Bcl-2/Bcl-xl inhibitor, the Bcl-2/Bcl-xl inhibitor-containing composition, the preparation method and the application of the Bcl-2/Bcl-xl inhibitor are one embodiment, the Bcl-2/Bcl-xl inhibitor adopts the compound synthesized in the embodiment 1, and the Bcl-2/Bcl-xl inhibitor-containing composition is obtained by combining the compound with DNA alkylating agent carboplatin.
Example 30
The Bcl-2/Bcl-xl inhibitor, the Bcl-2/Bcl-xl inhibitor-containing composition, the preparation method and the application of the Bcl-2/Bcl-xl inhibitor are one embodiment, the Bcl-2/Bcl-xl inhibitor adopts the compound synthesized in the embodiment 2, and the Bcl-2/Bcl-xl inhibitor-containing composition is obtained by combining the compound with DNA alkylating agent dicycloplatin.
Example 31
According to one embodiment of the Bcl-2/Bcl-xl inhibitor, the Bcl-2/Bcl-xl inhibitor-containing composition, the preparation method and the application thereof, the Bcl-2/Bcl-xl inhibitor adopts the compound synthesized in the embodiment 3, and the Bcl-2/Bcl-xl inhibitor-containing composition is obtained by combining the compound with DNA alkylating agent oxaliplatin.
Example 32
The Bcl-2/Bcl-xl inhibitor, the Bcl-2/Bcl-xl inhibitor-containing composition, the preparation method and the application of the Bcl-2/Bcl-xl inhibitor are one embodiment, the Bcl-2/Bcl-xl inhibitor adopts the compound synthesized in the embodiment 4, and the Bcl-2/Bcl-xl inhibitor-containing composition is obtained by combining the compound with DNA alkylating agent carboplatin.
Example 33
According to the invention, the Bcl-2/Bcl-xl inhibitor, the composition containing the Bcl-2/Bcl-xl inhibitor, the preparation method and the application of the composition are one embodiment, the Bcl-2/Bcl-xl inhibitor adopts the compound synthesized in the embodiment 8, the composition containing the Bcl-2/Bcl-xl inhibitor is obtained by combining the compound with DNA alkylating agent carboplatin, and meanwhile, the composition also contains excipient copolyvidone VA64, soluplus, vitamin C, span 20, polysorbate 80, colloidal silicon dioxide, calcium hydrophosphate, microcrystalline cellulose, crosslinked sodium carboxymethyl cellulose and sodium stearyl fumarate.
Effect example 1
To verify the substantial effect of the Bcl-2/Bcl-xl inhibitors of the invention, the compounds obtained in example 9 were assayed, the test method comprising the steps of:
(1) Bcl-2 competitive binding (fluorescence polarization) assay
Principle of: a fluorescently labeled 23 amino acid peptide BH3 was purchased from Calbiochem (NLWAAQRYGRELRRMSDKFVD; SEQ ID NO: 1). Unbound fluorescein-labeled BH3 peptide emits random light relative to the plane of the polarization plane of the excitation light, resulting in lower polarization degree (mP) values. When the peptide binds to Bcl-2, the complex rolls over slower and the emitted light can have a higher level of polarization, resulting in a higher mP value.
Embodiments are described below: performed in 96-well plates and each sample contained 15 and 30nM of the labeled peptide and purified Bcl-2 protein (purchased from R&D Systems, inc). The assay buffer contained 20mM Hepes (pH 7.0), 50mM KCl, 5mM MgCl 2 、20mM Na 2 MoO 4 0.lmg/ml bovine Gamma globulin and 0.01% NP40. Test compounds were diluted in DMSO and added to the final assay at a concentration range of 20 μm to 2 nM.The polarization (mP) value was determined by BioTek Synergy II after incubation for 3h at room temperature with background subtraction. Calculation of IC using Prism software with sigmoidal dose-response curve fitting 50 . The comparison sample set was set up using the compounds ABT-199 (Bcl-2 inhibitor, molecular formula: C45H50ClN7O 7S) and ABT-263 (Bcl-2/Bcl-xl inhibitor, molecular formula: C47H55ClF3N5O6S 3) and using ABT-737 (Bcl-2/Bcl-xl inhibitor, molecular formula: C42H45ClN6O5S 2) as reference compounds. From experience and routine testing procedures, these compounds exhibit activities substantially in IC 50 In the range of =0.1 to 1000 nM.
(2) BCL-2 dependent Acute Lymphoblastic Leukemia (ALL) cell line RS4;11 in vitro antiproliferative assay
Embodiments are described below: through PerkinElmer ATPlite TM The luminometric assay system measures cell antiproliferative efficacy. The cancer cell line RS4;11 (ATCC, catlog: CRL-1873) at about 1X 10 per well 4 The density of individual cells was seeded in 96-well plates and incubated with different concentrations of compound (0.0056 nM to 1000nM, 12 total) in medium supplemented with 5% fbs for about 72h. A vial of lyophilized substrate solution was then reconstituted by adding 5mL of substrate buffer and gently stirred until the solution was homogeneous. About 50 μl of mammalian cell lysis solution was added to 100 μl of cell suspension per well of the microplate, and the plate was shaken in an orbital shaker at about 700rpm for about 5min for lysing the cells and stabilizing ATP. Subsequently, 50 μl of substrate solution was added to the wells, and the microwell plates were shaken in an orbital shaker at about 700rpm for 5min. Finally, by Perkinelmer
Figure BDA0004154928500000641
The microplate scintillation counter measures luminescence. Such assays with a range of doses of test compounds allow for the determination of cellular antiproliferative ICs of the compounds of the invention 50
The test results are shown in Table 1, and it can be seen that the IC of the Bcl-2/Bcl-xl inhibitor of the present invention 50 The absolute value is far lower than that of the existing commercial Bcl-2 inhibitors and Bcl-2/Bcl-xl inhibitors, which are pharmaceutically active against cancer cell linesAbove these existing products.
TABLE 1
Figure BDA0004154928500000642
Further, the SCLC cell line NCI-H889 (ATCC, catlog: CRL-5817) and NCI-526 (ATCC, catlog: CRL-5811) were tested by the in vitro antiproliferative assay as described above, and the test results are shown in Table 2. Obviously, the pharmaceutical activity of the Bcl-2/Bcl-xl inhibitor of the invention is still higher in different cancer cell lines, especially SCLC cell lines, and the effect of the Bcl-2/Bcl-xl inhibitor is greatly different from that of the existing products. Wherein ABT-199 was not detected in NCI-H889 assay system.
TABLE 2
Figure BDA0004154928500000651
Effect example 2
To verify the combined effect of Bcl-2/Bcl-xl inhibitors and DNA alkylating agents in the Bcl-2/Bcl-xl inhibitor-containing compositions of the invention, in vitro antiproliferative assay tests were performed on different SCLC cell lines:
through PerkinElmer ATPlite TM The luminometric assay system measures cell antiproliferation. Each test cancer cell line (including NCI-H69 (ATCC, catog: HTB-119), NCI-H889 (ATCC, catog: CRL-5817), and NCI-H146 (ATCC, catog: HTB-173) was used at a rate of about 5X 10 per well 3 The density of individual cells was inoculated in 96-well plates and incubated in medium supplemented with 5% FBS in combination with various concentrations of the compound obtained in example 9 (hereinafter referred to as Compound 1) and DNA alkylating agent cisplatin for about 72 hours. A vial of lyophilized substrate solution was then reconstituted by adding 5mL of substrate buffer solution and gently stirred until the solution was homogeneous. About 50 μl of mammalian cell lysis solution was added to 100 μl of cell suspension per well of the microplate, and the plate was shaken in an orbital shaker at about 700rpm for about 5min for lysing the cells and stabilizing ATP. Subsequently, 50. Mu.L of substrate solution was added to the wells and the microplate was placed in an orbital shaker toThe mixture was shaken at about 700rpm for 5min. Finally, by Perkinelmer
Figure BDA0004154928500000652
The scintillation counter measures luminescence. Such assays are performed with a range of doses of the test compound. The following is the synergistic activity of compound 1 of the present invention in combination with cisplatin on several cancer cell lines (5% fbs). The test results are shown in fig. 1-3, and it can be seen that the Bcl-2/Bcl-xl inhibitor and the DNA alkylating agent can achieve strong synergistic activity at sufficient concentrations in different cell lines, which indicates that the composition formed by the Bcl-2/Bcl-xl inhibitor and the DNA alkylating agent has higher synergistic pharmaceutical activity for inhibiting and treating cancers, especially SCLC.
Effect example 3
According to the experimental results of effect example 2, in order to verify the actual effects (pharmaceutical activity and toxicity) of the Bcl-2/Bcl-xl inhibitor and the DNA alkylating agent in the Bcl-2/Bcl-xl inhibitor-containing composition of the present invention in living body treatment, the pharmacokinetic parameters of the Bcl-2/Bcl-xl inhibitor of the present invention are first studied as follows:
the pharmacokinetics of Compound 1 was evaluated in CD-1 mice (animal production license: SCXK (Zhe) 2019-0001) by intravenous and oral administration, liwa laboratory animal technologies Co., ltd. IV doses were administered as a slow bolus in the jugular vein and oral doses were administered by gavage. The formulation for IV administration was 5% dmso in 20% hpbcd in water and the PO formulation was 2.5%DMSO,10%EtOH,20%Cremphor EL,67.5%D5W. PK time points for IV arms are 5, 15, 30min and 1, 2, 4, 6, 8, 12, 24h post-dose, and PK time points for PO arms are 15, 30min and 1, 2, 4, 6, 8, 12, 24h post-dose. About 0.03mL of blood was collected at each time point. The blood of each sample was transferred to a plastic microcentrifuge tube containing EDTA-K2 and plasma was collected by centrifugation at 4000g for 5min in a centrifuge at 4 ℃ over 15 min. Plasma samples were stored in polypropylene tubes. Prior to analysis, the samples were stored in a refrigerator at-75±15 ℃. Plasma samples were analyzed for compound concentration using LC-MS/MS method. WinNonlin (Phoenix) TM Version 6.1) for pharmacokinetic metersAnd (5) calculating. The following pharmacokinetic parameters were calculated from the data of plasma concentration versus time, where possible: IV administration: c (C) 0 ,CL,V d ,T 1/2 ,AUC inf ,AUC last MRT, regression points; PO application: c (C) max ,T max ,T 1/2 ,AUC inf ,AUC last F%, regression points. Pharmacokinetic data are described using descriptive statistics (e.g., mean, standard deviation). The PK results of 10mg/kg for the simultaneous oral administration of PO were also counted as shown in Table 3.
TABLE 3 Table 3
Route of administration IV PO
Dosage for administration 1mg/kg 10mg/kg
Sex of animals Male male Male male
Number of animals 3 3
CL(mL/min/kg) 13.4±3.0
T 1/2 (h) 3.5±0.4 2.7±0.2
T max (h) 2.7±1.2
C 0 or C max (ng/mL) 969.5±142.0 1523.3±372.2
AUClast(h*ng/mL) 1276.7±288.9 11533.2±4000.8
AUCInf(h*ng/mL) 1281.8±288.9 11541.3±4001.0
MRT(h) 2.8±0.3
Vss obs(L/kg) 2.3±0.3
F(%) 90.1±31.2
Subsequently, a human tumor immunodeficiency mouse subcutaneous xenograft tumor model was established by a cell inoculation method for testing:
collecting tumor cells in logarithmic growth phase, counting, re-suspending in 1×PBS, and regulating cell suspension concentration to 2.5-5×10 7 /mL. Tumor cells were inoculated subcutaneously on the right back of immunodeficient mice (BALB/c Nude mice, purchased from Beijing An Kaiyi Bo, production license number: SCXK (Su) 2017-0006) 5 to 10X 10) with a 1mL syringe (needle No. 4) 6 0.2 mL/mouse. All animal experimental procedures strictly follow the middle american coronal biotechnology (beijing) limited laboratory animal usage and management specifications. The calculation of the related parameters refers to the technical guidelines of non-clinical research of cytotoxic anti-tumor drugs of CFDA in China.
Animal body weight and tumor size were measured twice a week during the experiment. Periodically observing the growth condition of the tumor until the tumor grows to an average volume of 100-150 mm 3 At this time, the administration was randomly grouped according to tumor size and mouse weight. The animal is observed every day for death. Conventional monitoring includes the effect of tumor growth and administration on normal animal behavior, including activity, feeding and drinking conditions, weight gain or loss, eyes, hair and other abnormalities in experimental animals. Both the death and clinical symptoms observed during the experiment are recorded in the raw data. The whole administration, the measurement of the body weight of the mice and the tumor volume were performed in an ultra clean bench. Plasma and tumor tissue were collected after the end of the last dose, weighed and photographed for recording, as required by the protocol. The plasma and tumor samples were frozen at 80℃for further use.
The calculation formula of Tumor Volume (TV) is: tv=a×b 2 /2. Wherein a and b represent tumor measurement length and width, respectively. The relative tumor volume (relative tumor volume, RTV) was calculated as: rtv=vt/V1. Where V1 is the tumor volume at the time of group administration and Vt is the tumor volume measured on the day after administration. The evaluation index of the anti-tumor activity is relative tumor proliferation rate T/C (%), and the calculation formulas are respectively as follows: relative tumor proliferation rate T/C (%) = (T) RTV /C RTV )×100%,T RTV For treatment of RTV, C RTV RTV for vehicle control; tumor remission (%) is the number of tumor-bearing mice that developed SD (disease stabilization), PR (partial tumor regression) and CR (complete tumor regression) after treatment divided by the total number of mice in the group x 100%.
Animal weight change (Change of body weight,%) = (weight at weight group measurement/weight at group time×100%).
Efficacy evaluation criteria: according to the guidelines of non-clinical research technology of cytotoxic antitumor drugs (11 months in 2006) of CFDA of China, the T/C (%) value is less than or equal to 40%, and p is effective by statistical analysis. A drug dose is considered severely toxic if the mice lose more than 20% weight or the drug-related mortality exceeds 20%. The synergy analysis uses the following formula: cofactor= ((a/C) × (B/C))/(AB/C); RTV values for a = a drug single group; RTV values for B = B drug single group; RTV values for c=vehicle control, RTV values for ab=ab combination (Clarke r. Issues in experimental design and endpoint analysis in the study ofexperimental cytotoxic agents in vivo in breast cancer and other models [ J ]. Breast Cancer Research & Treatment,1997,46 (2 3): 255 278). If the synergistic factor is more than 1, the synergistic effect is achieved; if the cofactor=1, then there is an additive effect; if the cofactor is <1, then antagonism is provided.
(1) Preparation of compound 1 drug solution: 250mg of Compound 1 was weighed into a mixture of (1.25 mL EtOH+5mL PEG400+10mL Kolliphor HS 15) and sonicated to a uniform solution, then 33.75mL of water was added to make 50mL yellow solution, and the mixture was dispensed into 7 bottles for 7 days, 7mL each.
(2) Preparing DNA alkylating agent carboplatin liquid medicine: 42mg of Carboplatin was weighed and 7mL of physiological saline was added to prepare 7mL of a clear dosing solution.
(3) Sequential dosing intervention: wherein the drug administration method of the compound 1 drug solution is oral administration, and each time, the drug administration is carried out according to 50mg/kg body weight; carboplatin was administered by intraperitoneal injection, and 6mg/mL of carboplatin was prepared with physiological saline, and administered by intraperitoneal injection at 60 mg/kg.
Right side subcutaneous inoculation by Balb/c nude mice 5X 10 6 NCI-H526 small cell lung cancer to construct a nude mouse small cell lung cancer subcutaneous tumor model. When the tumor grows to average 125.86mm 3 After that, compound 1 liquid medicine solution treatment (dosage is 50 mg/kg) is carried out by intragastric administration once a day, carboplatin liquid medicine (60 mg/kg) is administrated by intraperitoneal injection every week for 28 days, and the administration is carried out for 4 times, tumor volume is measured during the period, experimental nude mice are sacrificed after the experiment is finished, tumor weights are recorded, and the results show that each treatment group has no animal death, does not show obvious drug toxicity and is well tolerated during the treatment period. Vehicle control mice had an average tumor volume of 2144.93mm on day 24 post-grouping 3 . The average tumor volume of the test drug Carboplatin (60 mg/kg) +Compound 1 (50 mg/kg) treatment group at day 24 after the grouping was 540.85mm 3 Compared with the control group, the tumor inhibition rate TGI (%) is 75.82% with statistically significant difference (p < 0.001). The tumor re-analysis results are substantially identical to the relative tumor volume analysis results. The result shows that the Bcl-2/Bcl-xl inhibitor and the DNA alkylating agent have obvious combined effect in-vivo experiments, and the pharmaceutical activity of the composition is higher than the use curative effect of the single Bcl-2/Bcl-xl inhibitor or the DNA alkylating agent.
The test results are shown in tables 4 and 5 below.
TABLE 4 Table 4
Figure BDA0004154928500000671
Figure BDA0004154928500000681
Note that: 1. data are expressed as "mean ± standard error"; T/C% = T RTV /C RTV ×100%;TGI%=(1-T/C)×100%。
TABLE 5
Figure BDA0004154928500000682
Note that: 1. data are expressed as "mean ± standard error"A representation; T/C% = T TW /C TW X 100%; TGI% = (1-T/C) ×100%;3. g1#702, #704 mice specifically grouped during the test were more than 3000mm on day 24 post-grouping due to single tumor volume 3 Is euthanized so that tumor weight is not accounted for.
Finally, it should be noted that the above embodiments are only for illustrating the technical solution of the present invention and not for limiting the scope of the present invention, and although the present invention has been described in detail with reference to the preferred embodiments, it should be understood by those skilled in the art that the technical solution of the present invention may be modified or substituted equally without departing from the spirit and scope of the technical solution of the present invention.

Claims (10)

1. A Bcl-2/Bcl-xl inhibitor characterized by the chemical structure shown below:
Figure FDA0004154928480000011
wherein,,
q4 and Q5 are at least one of cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl and spiroheterocyclyl;
R 1 、R 2 、R 7 、R 9 and R is 10 Is H, D, alkyl, spiroalkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, spiroheterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl, halogen, nitro, oxo, cyano, -OR a 、-SR a -alkyl-R a 、-NH(CH 2 ) p R a 、-C(O)R a 、-S(O)Ra、-SO 2 R a 、-C(O)OR a 、-OC(O)R a 、-NR b R c 、-C(O)N(R b )R c 、-N(R b )C(O)R c 、-P(O)R b R c -alkyl-P (O) R b R c 、-S(O)(=N(R b ))R c 、-N=S(O)R b R c 、=NRb、-SO 2 N(R b )Rc、-N(R b )SO 2 R c Wherein at least one of said cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl is substituted with one or more R d Substitution;
R a 、R b 、R c 、R d is at least one of H, D, alkyl, spiroalkyl, alkenyl, alkynyl, alkoxy, alkoxyalkyl, haloalkyl, hydroxyalkyl, aminoalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, spiroheterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl, wherein at least one of the alkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl is substituted with one or more R e Substitution;
R e is at least one of H, D, alkyl, spiroalkyl, alkenyl, alkynyl, halogen, cyano, amine, nitro, hydroxy, =o, -C (O) NHOH, alkoxy, alkoxyalkyl, haloalkyl, hydroxyalkyl, aminoalkyl, alkylcarbonyl, alkoxycarbonyl, alkylcarbonylamino, alkylamino, oxo, haloalkylamino, cycloalkyl, cycloalkenyl, heterocycloalkyl, spiroheterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl;
Z 1 Is a bond, - (CH) 2 ) p -、-N(H)-、-O-、-S-、-C(O)-、-S(O 2 )-、-OC(O)-、-C(O)O-、-OSO 2 -、-S(O 2 )O-、-C(O)S-、-SC(O)-、-C(O)C(O)-、-C(O)N(H)-、-N(H)C(O)-、-S(O 2 )N(H)-、-N(H)S(O 2 )-、-OC(O)O-、-OC(O)S-、-OC(O)N(H)-、-N(H)C(O)O-、-N(H)C(O)S-、-N(H)C(O)N(H)-、-(CH 2 ) p N(H)(CH 2 ) q -、-(CH 2 ) p N(H)C(O)(CH 2 ) q -、-(CH 2 ) p C(O)N(H)(CH 2 ) q -、-OC(O)N(H)(CH 2 ) p+1 N(H)(CH 2 ) q -, a divalent alkenyl group or a divalent alkynyl group;
l is-L 1 -L 2 -;
Wherein L is 1 Is a bond, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl or heteroarylAt least one of the alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl is substituted with one or more R d Substitution;
L 2 is a bond or an alkyl group, at least one of which is bound to one or more-L i Optionally interposed between any two adjacent carbon atoms;
the said-L i -is-N (Ra) -, -O-, -S-, -C (O) -, -S (O) 2 )-、-OC(O)-、-C(O)O-、-OSO 2 -、-S(O 2 )O-、-C(O)S-、-SC(O)-、-C(O)C(O)-、-C(O)N(Ra)-、-N(Ra)C(O)-、-S(O 2 )N(R a )-、-N(R a )S(O 2 )-、-OC(O)O-、-OC(O)S-、-OC(O)N(R a )-、-N(R a )C(O)O-、-N(R a )C(O)S-、-N(R a )C(O)N(R a ) At least one of a divalent alkenyl group, a divalent alkynyl group, a divalent cycloalkyl group, a divalent heterocycloalkyl group, a divalent aryl group, a divalent heteroaryl group;
the k, n, p, q =0 to 5,s =0 or 1, and f=0 or 1.
2. The Bcl-2/Bcl-xl inhibitor of claim 1, comprising at least one of the following (a) - (e):
(a) The R is 2 Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R d Substitution;
(b) The R is 10 Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R d Substitution;
(c) The R is 7 And L, together with the atoms to which they are attached, form cycloalkyl or heterocycloalkyl, said cycloalkyl or heterocycloalkyl being substituted with one or more R e Substitution;
(d) The R is b And R is c Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R e Substitution;
(e) The R is d Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R e Substitution;
the R is e Together with the atoms to which they are attached form a cycloalkyl or heterocycloalkyl group, which is substituted with one or more R f Substitution, said R f Is at least one of H, D, alkyl, alkenyl, alkynyl, halogen, cyano, amine, nitro, hydroxy, -C (O) NHOH, alkoxy, alkoxyalkyl, haloalkyl, hydroxyalkyl, aminoalkyl, alkylcarbonyl, alkoxycarbonyl, alkylcarbonylamino, alkylamino, oxo, haloalkylamino, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl.
3. The Bcl-2/Bcl-xl inhibitor of claim 1, wherein the Bcl-2/Bcl-xl inhibitor has a chemical structure as follows:
Figure FDA0004154928480000021
Preferably, the chemical structure of the Bcl-2/Bcl-xl inhibitor is as follows:
Figure FDA0004154928480000022
4. the Bcl-2/Bcl-xl inhibitor of claim 1, wherein the Bcl-2/Bcl-xl inhibitor is any one of the following compounds:
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 3-difluoro-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((4- (((4-fluorotetrahydro-2H-pyran-4-yl) methyl) amino) -3-nitrophenyl) sulfonyl) benzamide;
or (b)
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(R) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
Or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) benzamide;
or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((4- (((4-fluorotetrahydro-2H-pyran-4-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -2- (4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- ((4- ((((R) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((S) -4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- ((4- ((((R) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((S) -4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
Or (b)
N- ((4- ((((R) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((R) -4-methyl-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -N- ((3-nitro-4- (((tetrahydro-2H-pyran-4-yl) methyl) amino) phenyl) sulfonyl) -2- (4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
Or (b)
N- ((4- ((((S) -1, 4-dioxan-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- ((R) -4- (trifluoromethyl) -3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-4, 4-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
(S) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((5-nitro-3- (tetrahydro-2H-pyran-4-yl) -3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) benzamide;
Or (b)
(R) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) -N- ((5-nitro-3- (tetrahydro-2H-pyran-4-yl) -3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) benzamide;
or (b)
N- (((R) -3- ((S) -1, 4-dioxane-2-yl) -5-nitro-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
or (b)
N- (((S) -3- ((S) -1, 4-dioxane-2-yl) -5-nitro-3, 4-dihydro-2H-benzo [ b ] [1,4] oxazin-7-yl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide;
preferably, the Bcl-2/Bcl-xl inhibitor is as follows:
(S) -N- ((4- (((1, 4-dioxane-2-yl) methyl) amino) -3-nitrophenyl) sulfonyl) -4- (4- ((4 '-chloro-5, 5-dimethyl-3, 4,5, 6-tetrahydro- [1,1' -biphenyl ] -2-yl) methyl) piperazin-1-yl) -2- (3, 3-difluoro-3, 4-dihydro-2H-pyrrolo [3',2':5,6] pyrido [2,3-b ] [1,4] oxazepin-1 (7H) -yl) benzamide.
5. The use of the Bcl-2/Bcl-xl inhibitor of any one of claims 1-4 for the manufacture of a medicament for the treatment of cancer; preferably, the cancer comprises small cell lung cancer, breast cancer, colorectal cancer.
6. The use according to claim 5, comprising at least one of the following (a) - (d):
(a) The administration mode of the medicine for treating cancer is sequential administration;
(b) The components of the medicament for treating cancer also comprise excipients and/or pharmaceutically acceptable carriers and/or pharmaceutically acceptable vehicles;
(c) The dosage form of the cancer treatment medicine is any one of liquid dosage form, gas dosage form, solid dosage form and semisolid dosage form;
(d) The cancer is small cell lung cancer.
7. A composition comprising the Bcl-2/Bcl-xl inhibitor of any one of claims 1-4, or a nitrogen oxide thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a compound thereof, or a prodrug form of a nitrogen oxide, and a DNA alkylating agent; preferably, the components of the composition further comprise excipients and/or pharmaceutically acceptable carriers and/or pharmaceutically acceptable vehicles.
8. The composition of claim 7, wherein the DNA alkylating agent comprises a platinum-containing alkylating agent; preferably, the platinum-containing alkylating agent is at least one of platinum, cisplatin, carboplatin, dicycloplatin, etaplatin, lobaplatin, milliplatin, nedaplatin, oxaliplatin, pi Pubo, satraplatin and/or triplatin tetranitrate.
9. Use of a composition according to any one of claims 7 to 8 in the manufacture of a medicament for the treatment of cancer; preferably, the cancer comprises small cell lung cancer, breast cancer, colorectal cancer.
10. The use according to claim 9, comprising at least one of the following (a) - (d):
(a) The administration mode of the medicine for treating cancer is sequential administration;
(b) The components of the medicament for treating cancer also comprise excipients and/or pharmaceutically acceptable carriers and/or pharmaceutically acceptable vehicles;
(c) The dosage form of the cancer treatment medicine is any one of liquid dosage form, gas dosage form, solid dosage form and semisolid dosage form;
(d) The cancer is small cell lung cancer.
CN202310330972.6A 2023-03-30 2023-03-30 Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof Pending CN116270660A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310330972.6A CN116270660A (en) 2023-03-30 2023-03-30 Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310330972.6A CN116270660A (en) 2023-03-30 2023-03-30 Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN116270660A true CN116270660A (en) 2023-06-23

Family

ID=86818626

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310330972.6A Pending CN116270660A (en) 2023-03-30 2023-03-30 Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN116270660A (en)

Similar Documents

Publication Publication Date Title
US11767313B2 (en) Compounds as nuclear transport modulators and uses thereof
WO2021218110A1 (en) Benzothiazolyl biaryl compound, and preparation method and use
ES2857251T3 (en) Polyfluoro compounds that act as bruton tyrosine kinase inhibitors
RU2745035C1 (en) Fgfr inhibitor and its application
TW201930292A (en) Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors
CN102131390A (en) Triazolopyridine jak inhibitor compounds and methods
CN102333779A (en) Benzopyran and benzoxepin pi3k inhibitor compounds and methods of use
WO2016155545A1 (en) Sulfamyl-containing 1,2,5-oxadiazole derivative, preparation method therefor and use thereof in pharmaceuticals
JP7164203B2 (en) Pyrrolo aromatic heterocyclic compound, method for producing same, and medical use
CN110167941B (en) Substituted fused heteroaryl compounds as kinase inhibitors and uses thereof
WO2021190417A1 (en) Novel aminopyrimidine egfr inhibitor
CN110546150B (en) Pyrazolyl-containing tricyclic derivatives, preparation method and application thereof
CN107312009B (en) Quinoline compound, preparation method, intermediate, pharmaceutical composition and application thereof
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
WO2022228543A1 (en) Bridged ring compound, preparation method therefor, and application thereof in medicine
KR20210066839A (en) FGFR4 inhibitors and uses thereof
WO2020238776A1 (en) Substituted fused bicyclic derivative, preparation method therefor, and application thereof in medicines
CN110312717A (en) The condensed heteroaryl tricyclic compound replaced is as kinase inhibitor and its application
WO2023125928A1 (en) Menin inhibitor and use thereof
JP2018513214A (en) Preparation and use of novel kinase inhibitors
CN116270660A (en) Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof
WO2022253311A1 (en) Hpk1 inhibitor and use thereof
WO2022148439A1 (en) Heterocyclic compound as bcl-2 inhibitor
CN116236583A (en) Bcl-2/Bcl-xl inhibitor-containing composition and preparation method and application thereof
WO2022117090A1 (en) Polycyclic compound, and preparation method therefor and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination