CN113121527A - Tricyclic compound and use thereof - Google Patents

Tricyclic compound and use thereof Download PDF

Info

Publication number
CN113121527A
CN113121527A CN202011552396.2A CN202011552396A CN113121527A CN 113121527 A CN113121527 A CN 113121527A CN 202011552396 A CN202011552396 A CN 202011552396A CN 113121527 A CN113121527 A CN 113121527A
Authority
CN
China
Prior art keywords
cancer
indole
tetrahydro
reaction mixture
halogen
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
CN202011552396.2A
Other languages
Chinese (zh)
Inventor
程耀邦
王永辉
董志强
栗增
沈孝坤
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.)
Fukang Shanghai Health Technology Co ltd
Shanghai Huiqi Biomedical Technology Co ltd
Original Assignee
Fukang Shanghai Health Technology Co ltd
Shanghai Huiqi Biomedical Technology 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 Fukang Shanghai Health Technology Co ltd, Shanghai Huiqi Biomedical Technology Co ltd filed Critical Fukang Shanghai Health Technology Co ltd
Publication of CN113121527A publication Critical patent/CN113121527A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles
    • C07D209/88Carbazoles; Hydrogenated carbazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/94[b, c]- or [b, d]-condensed containing carbocyclic rings other than six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Transplantation (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Diabetes (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention disclosesA tricyclic compound and its application are provided. The invention particularly discloses a compound shown as a formula I or pharmaceutically acceptable salt thereof. The tricyclic compound disclosed by the invention has a good inhibition effect on HDAC (Histone deacetylase), particularly HDAC6, has good selectivity, and can inhibit the proliferation of various tumor cells;

Description

Tricyclic compound and use thereof
Technical Field
The invention belongs to the technical field of chemical medicines, and particularly relates to a tricyclic compound and application thereof.
Background
Epigenetic modification plays a very important role in the expression regulation process of genes, and Histone Deacetylases (HDACs) have attracted wide attention of scientists in recent decades as important functional proteins for epigenetic regulation. On one hand, HDACs can mediate deacetylation of histone substrate lysine, thereby facilitating chromatin to form a more compact structure, and some HDACs can interact with other chromatin regulatory proteins to form a co-suppression complex, thereby regulating gene expression, cell cycle, cell differentiation and other life processes; on the other hand, some HDACs are also able to catalyze the deacetylation of the non-histone substrate lysine, playing an important role in more cell regulatory pathways (Nat Biotechnol,2015, 10.1038/nbt.3130; Int J Cancer,2019, 10.1002/ijc.32169). Examples of histone deacetylase-mediated diseases which are currently investigated to be associated with HDAC inhibition include cell proliferative diseases, such as malignancies, inflammatory diseases such as inflammatory bowel disease, crohn's disease or ulcerative enteritis, autosomal dominant diseases such as huntington's disease, down's syndrome, Edward's syndrome or patassis syndrome, genetic metabolic diseases such as diabetes, Niemann Pick disease, Gaucher's disease, phenylketonuria, Wilson's disease or fibrotic diseases such as cystic fibrosis, liver fibrosis, kidney fibrosis, lung fibrosis or skin fibrosis, autoimmune diseases such as rheumatoid arthritis, asthma, lupus, psoriasis, psoriatic arthritis, multiple sclerosis, behcet's disease, or organ transplant rejection: acute/chronic neurological, systemic diseases such as stroke or polycystic kidney disease, hypertrophy such as cardiac hypertrophy, heart failure such as congestive heart failure or hemorrhagic heart failure, ocular diseases such as glaucomatous dry eye, syndrome, dry macular degeneration, wet macular degeneration, diabetic retinopathy or uveitis, neurodegenerative diseases such as alzheimer's disease, amyotrophic lateral sclerosis, progressive peroneal muscular atrophy or spinal muscular atrophy, and conditions and diseases caused by abnormal function of HDAC enzymes.
A total of 18 HDACs are currently found in humans and classified into 4 types according to evolutionary analysis and sequence homology analysis (Strahl BD, Allis CD. Nature.2000Jan 6; 403(6765): 41-5.): class I is HDAC1-3, 8 with higher sequence homology to Rpd3 protein in yeast, which are mainly located in the nucleus, wherein HDAC3 is present in the cytoplasm; class II is HDAC4-7, 9,10 with high sequence homology to the Hda1 (hormone deacetylase-1) protein in yeast, which can shuttle between the nucleus and cytoplasm in response to different cellular signals, and have cellular and tissue specificity; class III is a family of sirtuin proteins homologous to Sir2 (silence information regulator 2) proteins in yeast, including SIRT 1-7; class IV contains only one member, HDAC11, with sequence homology between Rpd3 and Hda1, located mainly in the nucleus. HDAC classes I, II and IV can be classified as Rpd3/Had1 deacetylase family, and all of them comprise catalytic core domains with high homology, and the catalytic activity of the HDAC is dependent on the participation of zinc ions. Currently, HDAC inhibitors in clinical trials can be classified into four types according to chemical structure: hydroxamic acid, cyclic peptide, benzamide and short chain fatty acid. SAHA and PXD101 belong to the hydroxamic acids, whereas FK228 is a member of the cyclic peptides, neither of these compounds exhibits any HDAC subtype selectivity (Tomaselli. et al. Med Res Rev.2019Jun20.doi: 10.1002/med.21600).
Since subtype-selective inhibitors can treat biological functions of different subtypes separately, drugs with subtype-selective inhibitors have fewer potential side effects. However, designing selective inhibitors with HDAC isoforms is very difficult at the present stage, mainly for two reasons: existing HDAC structural data is restricted to several limited subtypes (HDAC1, HDAC2, HDAC3, HDAC4, HDAC7 and HDAC 8); in addition, the high degree of sequence conservation in certain HDACs allows them to have a high degree of structural similarity, and the difficulty of designing a target for a certain HDAC in these subtypes is very high. To date, few documents have been reported about selective inhibitors of HDAC isoforms. To date, only non-selective HDAC inhibitors have been identified as anti-cancer agents. Non-selective HDAC inhibitors are known to cause side effects, such as fatigue and nausea, usually at high doses (Piekarz et al pharmaceuticals (Basel) 2010 Sep; 3(9): 2751-2767). This side effect is reported to be due to inhibition of class I HDACs. Due to such side effects, the use of non-selective HDAC inhibitors in drug development other than anticancer drugs is limited (Witt et al, Cancer Lett.2009May 8; 277(1): 8-21.). Meanwhile, it is reported that selective inhibition of class II HDACs does not show toxicity shown in class I HDAC inhibition. Therefore, it is possible that selective HDAC inhibitors could be developed as therapeutic agents effective in the treatment of various diseases (Matthias et al. mol Cell biol. 2008Mar; 28(5): 1688-70).
The selective HDAC6 inhibitor is a research hotspot in the field at present, and is expected to overcome the defects of poor selectivity, large side effect and the like of a broad-spectrum HDAC inhibitor. HDAC6 mainly catalyzes the deacetylation of α -tubulin, heat shock protein Hsp90, cortical actin, and peroxiredoxin reductase. Due to their unique structure, HDAC6 has multiple distinct biological functions and can regulate multiple cellular pathways associated with cell growth, metastasis, and apoptosis through both deacetylase-dependent and independent mechanisms. HDAC6 has also been shown to be important in the translocation of misfolded proteins to aggregates and in the prevention of apoptosis in response to misfolded proteins. HDAC6 is closely related to the pathophysiological processes of tumors, neurodegenerative diseases, inflammations, autoimmune responses, bacterial infections, heart diseases and other diseases, and is a drug target with great application prospect. The HDAC6 inhibitors found in the present study are very few, mainly including Tubacin, Tubastatin A, ACY-1215, Citarinosist (ACY-241), etc.
Tubacin is a highly potent selective, reversible, cell-penetrating HDAC6 inhibitor, IC in cell-free assays50At 4nM, 350-fold more selective for HDAC1 (Butler KV, et al. J Am Chem Soc.2010,132(31), 10842-0846).
Tubastatin A is a potent, selective HDAC6 inhibitor, IC in cell-free assay50At 15nM, the selectivity was much higher (1000 fold) than for the other isoforms except HDAC8(57 fold). These two compounds have not been clinically developed due to their high toxicity (Gradilone et al cancer Res,2013,73(7), 2259-.
ACY-1215 is an orally bioavailable HDAC 6-specific inhibitor with potential anti-tumor activity, and compared to non-selective HDAC inhibitors, ACY-1215 reduces the toxic effects on normal, healthy cells. ACY-1215 is a hydroxamic acid derivative, and is 12, 10 and 11 fold selective for HDAC1, HDAC2 and HDAC3 (HDACs type I), respectively. ACY-1215 minimum Activity (IC)50>1 μ M) on HDAC4, HDAC5, HDAC7, HDAC9, HDAC11, Sirtuin1 and Sirtuin2, on HDAC8 (IC)500.1 μ M) had slight activity. IC of ACY-1215 on T cell toxicity50The value was 2.5. mu.M. ACY-1215 acts on the Bone Marrow (BM) environment to overcome tumor cell growth and survival conferred by BMSCs and cytokines. ACY-1215 in combination with Bortezomib induced synergyanti-MM activity. ACY-1215 induces potent α -tubulin acetylation at very low doses, triggering acetylation of histones H3 and H4 histone lysines only at higher doses, confirming its specific inhibitory effect on HDAC6 activity, ACY-1215 selectively targets and binds to HDAC6, disrupting the Hsp90 chaperone system by hyperacetylation of Hsp90, preventing subsequent degradation of aggrecan. ACY-1215 is currently undergoing secondary clinical trials for hematological and solid tumors, such as bortezomib alone and in combination
Figure BDA0002858379420000031
And dexamethasone for relapsed/refractory multiple myeloma, in combination with PD-1 antibody for breast, lung, etc. (Santo L, et al. blood,2012,119(11), 2579-.
ACY-241 is a selective HDAC6 inhibitor structurally similar to ricolinostat (ACY-1215), an orally active, selective HDAC6 inhibitor, IC for HDAC6 and HDAC3502.6nM and 46nM, respectively. The selectivity to HDAC6 was 13-18 fold greater than to HDAC 1-3. Niesvizky et al developed the first Ia/Ib clinical study on ACY-241, which included 40 relapsed/refractory multiple myeloma (RRMM) patients, with 3 groups of patients taking 180, 360, 480mg of ACY-241 daily for 3 weeks, 1 cycle every 28 days; POM + DEX was combined from cycle 2. From 6 months on 2015, 34 cases with assessed safety were reported, with grade 3 and 4 hematologic toxicities mainly neutropenia (10, 30%). The median follow-up period was 3.5 months, and 22 cases of efficacy were evaluated, with 1 case achieving Very Good Partial Remission (VGPR), 10 cases achieving PR, 2 cases achieving Minimal Remission (MR), 8 cases SD, 1 case of disease Progression (PD), and neither median PFS nor median remission period reached. On the basis of comprehensive pharmacodynamics, pharmacokinetics and safety, 360mg 1 time/day is taken as the recommended dose for future experiments. The combination of ACY-241 with Pom + Dex showed good tolerability and limited adverse effects, and further clinical trials were still in progress. In addition to hematological tumors, phase 1 clinical indications for melanoma and lung cancer are currently ongoing (Huang P, et al. oncotarget.2017,8(2): 2694-2707). However, ACY-1215 andthe ACY-241 also has the defects of poor cell metabolism, half-life period of only 3 hours shown by human body tests and the like, and brings inconvenience to clinical use.
In summary, the development of novel selective HDAC6 inhibitors for the treatment of cancer, inflammatory diseases, autoimmune diseases, neurological diseases and neurodegenerative diseases, and avoiding the side effects is a problem to be solved in clinical application.
Disclosure of Invention
The invention aims to overcome the defect of single structure of the existing HDAC inhibitors, particularly HDAC6 inhibitors, and provides a tricyclic compound and application thereof. The tricyclic compound disclosed by the invention has a good inhibition effect on HDAC (Histone deacetylase), particularly HDAC6, has good selectivity, and can inhibit the proliferation of various tumor cells.
The present invention solves the above-described problems with the following technical solutions.
The invention provides a compound shown as a formula I or a pharmaceutically acceptable salt thereof, which has the following structure,
Figure BDA0002858379420000041
wherein the content of the first and second substances,
z is N or CH;
m1 is 1 or 2;
m2 is 0 or 1;
R1is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl radical, C1-C3Alkoxy radical, C3-C8Cycloalkyl, 3-8 membered heterocycloalkyl with one or more heteroatoms selected from N, O and S and 1-4 heteroatoms, unsubstituted or R1-2Substituted C6-C14Aryl, or unsubstituted or R1-3A 5-12 membered heteroaryl group having one or more of N, O and S as a substituent "hetero atom, and 1-4 as hetero atoms";
R1-1is halogen, C3-C8Cycloalkyl, halogen substituted C3-C8Cycloalkyl, or 3-8 membered heterocycloalkyl with "heteroatom number 1-4" selected from one or more of N, O and S;
R1-2and R1-3Independently cyano or halogen;
R2is hydrogen, cyano, halogen, C1-C3Alkoxy radical, C1-C6Alkyl, halogen substituted C1-C6Alkyl radical, C3-C6Cycloalkyl, halogen substituted C3-C6Cycloalkyl, 3-6 membered heterocycloalkyl with 1-4 heteroatoms selected from N, O and S, or 3-6 membered heterocycloalkyl with 1-4 heteroatoms selected from N, O and S;
R3is hydrogen, halogen, C1-C3Alkoxy radical, C1-C6Alkyl radical, C3-C6Cycloalkyl, or 3-6 membered heterocycloalkyl.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as described above),
z is N or CH;
m1 is 1 or 2;
m2 is 0 or 1;
R1is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl, unsubstituted or R1-2Substituted C6-C14Aryl, or unsubstituted 5-12 membered heteroaryl with 1-4 heteroatoms selected from one or more of N, O and S;
R1-1is halogen, C3-C8Cycloalkyl, halogen substituted C3-C8Cycloalkyl, or 3-8 membered heterocycloalkyl with "heteroatom number 1-4" selected from one or more of N, O and S;
R1-2is halogen;
R2is hydrogen or halogen;
R3is hydrogen.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as described above),
z is N or CH;
m1 is 1 or 2;
m2 is 0 or 1;
R1is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl, unsubstituted or R1-2Substituted C6-C14Aryl, or unsubstituted 5-12 membered heteroaryl with 1-4 heteroatoms selected from one or more of N, O and S;
R1-1is C3-C8Cycloalkyl, or 3-8 membered heterocycloalkyl with "heteroatom number 1-4" selected from one or more of N, O and S;
R1-2is halogen;
R2is hydrogen or halogen;
R3is hydrogen.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above), and R is1Is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl, unsubstituted or R1-2Substituted C6-C14Aryl, or 5-12 membered heteroaryl with one or more unsubstituted "heteroatoms selected from N, O and S, and 1-4 heteroatoms.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above), and R is1-1Is halogen, C3-C8Cycloalkyl, halogen substituted C3-C8Cycloalkyl, or 3-8 membered heterocycloalkyl in which "hetero atom (S)" is (are) selected from N, O and S, and the number of hetero atom (S) is (are) 1 to 4 ".
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are as followsAs defined (undefined groups as defined above), R1-1Is halogen, or 3-8 membered heterocycloalkyl with 1-4 heteroatoms selected from one or more of N, O and S.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above), and R is1-2Is halogen.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above), and R is2Is hydrogen or halogen.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above), and R is3Is hydrogen.
In certain preferred embodiments of the present invention, the compound of formula I may be a compound of formula II,
Figure BDA0002858379420000061
wherein, Z, m1, m2, R1And R2As defined in any of the previous schemes.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1Is unsubstituted or R1-1Substituted C1-C6When it is alkyl, said R1-1The number of (A) may be 1 or more (e.g. 1,2 or 3), when said R is1-1When there are plural, R is1-1May be the same or different.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1Is unsubstituted or R1-1Substituted C1-C6When alkyl, said C1-C6Alkyl is preferably C1-C4Alkyl radicals, e.g. methyl, ethyl, propyl, isopropylAn alkyl group, an n-butyl group, an isobutyl group, a sec-butyl group or a tert-butyl group, more preferably a methyl group, an ethyl group, an isopropyl group or an isobutyl group, and still more preferably a methyl group, an isopropyl group or an isobutyl group.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1Is unsubstituted or R1-2Substituted C6-C14When aryl is said to R1-2The number of (A) may be 1 or more (e.g. 1,2,3 or 4), when said R is1-2When there are plural, R is1-2May be the same or different.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1Is unsubstituted or R1-2Substituted C6-C14When aryl, said C6-C14Aryl is preferably C6-C12More preferably phenyl.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1When the heteroaryl group is a 5-12 membered heteroaryl group having unsubstituted "one or more heteroatoms selected from N, O and S and 1-4 heteroatoms", the 5-12 membered heteroaryl group is preferably one or more "heteroatoms selected from N, O and S and 1-3" heteroatoms, more preferably a 5-6 membered heteroaryl group, e.g., a pyridyl, pyrimidinyl or pyrazinyl group
Figure BDA0002858379420000071
Further preferred is
Figure BDA0002858379420000072
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1-1Is C3-C8When there is a cycloalkyl group, said C3-C8Cycloalkyl is preferably C3-C6Cycloalkyl, more preferably cyclopropyl or cyclopentyl, e.g.
Figure BDA0002858379420000073
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1-1In the case of a 3-to 8-membered heterocycloalkyl group having "one or more hetero atoms selected from N, O and S and a hetero atom number of 1 to 4", the 3-to 8-membered heterocycloalkyl group is preferably a 5-to 6-membered heterocycloalkyl group having "one or more hetero atoms selected from N, O and S and a hetero atom number of 1 to 3", more preferably a tetrahydrofuranyl group or tetrahydropyranyl group, for example
Figure BDA0002858379420000074
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1-2In the case of halogen, the halogen is preferably fluorine, chlorine or bromine, more preferably fluorine or chlorine.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1Is unsubstituted or R1-1Substituted C1-C6When it is alkyl, said R1-1Substituted C1-C6Alkyl is preferred
Figure BDA0002858379420000075
More preferably
Figure BDA0002858379420000076
Figure BDA0002858379420000077
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is1Is unsubstituted or R1-2Substituted C6-C14When aryl is said to R1-2Substituted C6-C14Aryl is preferred
Figure BDA0002858379420000081
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above) when R is2In the case of halogen, the halogen is preferably fluorine, chlorine or bromine, more preferably chlorine.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula (I) are defined below (undefined groups are as described above), and R is1Is H, -CH3、-CH2CH3
Figure BDA0002858379420000082
Figure BDA0002858379420000083
Preferably H, -CH3
Figure BDA0002858379420000084
Figure BDA0002858379420000085
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above), and R is2Is H or Cl.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined below (undefined groups are as defined above), and R is3Is H.
In certain preferred embodiments of the present invention, certain groups of the compounds of formula I are defined as follows (undefined groups are as described above), and have the structure
Figure BDA0002858379420000086
Preference is given to
Figure BDA0002858379420000087
Figure BDA0002858379420000088
Wherein the a-terminal is linked to the C atom, the b-terminal is linked to the N atom, and the C-terminal is linked to
Figure BDA0002858379420000091
Are connected.
In certain preferred embodiments of the present invention, the compound of formula I can be of any of the following structures,
Figure BDA0002858379420000092
Figure BDA0002858379420000101
the invention provides a preparation method of the compound shown in the formula I, which comprises the following steps: in a solvent, in the presence of alkali, carrying out the reaction shown in the formula III on a compound shown in the formula III and hydroxylamine as follows;
Figure BDA0002858379420000102
wherein, Z, m1, m2, R1And R2As defined in any of the previous schemes.
In the preparation method of the compound shown in the formula I, the reaction conditions and operation are the same as those of the reaction in the field.
The preparation method of the compound shown in the formula I can further comprise the following steps:
when Z is N, it comprises the following steps: in a solvent, in the presence of a condensing agent and alkali, carrying out a condensation reaction shown as the following on a compound shown as a formula V-1 and a compound shown as a formula IV to obtain a compound shown as a formula III;
Figure BDA0002858379420000103
when Z is CH, it comprises the following steps: in a solvent, in the presence of a condensing agent and alkali, carrying out a condensation reaction shown as the following on a compound shown as a formula V-2 and a compound shown as a formula IV to obtain a compound shown as a formula III;
Figure BDA0002858379420000111
in the preparation method of the compound shown in the formula I, the conditions and operation of the condensation reaction are the same as those of the reaction in the field.
The invention also provides a pharmaceutical composition, which comprises the compound shown in the formula I or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. The compound shown in the formula I or the pharmaceutically acceptable salt thereof can be used in a therapeutically effective amount.
The invention also provides application of the compound shown as the formula I or pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparation of the HDAC inhibitor. The HDAC inhibitor is preferably an HDAC6 inhibitor.
The invention also provides application of the compound shown as the formula I or pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparation of medicines for treating diseases related to HDAC. The HDAC related diseases are preferably tumors and/or autoimmune diseases.
The invention also provides an application of the compound shown as the formula I or pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparing medicines. The medicament is preferably a medicament for preventing and/or treating tumors and/or autoimmune diseases.
The invention also provides an application of the compound shown in the formula I or the pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparing a medicament for preventing and/or treating tumors, wherein the compound shown in the formula I, the pharmaceutically acceptable salt thereof or the pharmaceutical composition is combined with a chemotherapeutic medicament or an immunotherapeutic medicament.
The invention also provides a method for treating tumors and/or autoimmune diseases, which comprises the step of administering a therapeutically effective amount of the compound shown in the formula I or the pharmaceutically acceptable salt thereof or the pharmaceutical composition to a patient.
Such tumors include, but are not limited to: lung cancer, colon cancer, rectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, brain cancer, kidney cancer, ovarian cancer, stomach cancer, skin cancer, bone cancer, glioma, glioblastoma, hepatocellular carcinoma, papillary renal carcinoma, head and neck cancer, leukemia, lymphoma, myeloma, multiple myeloma and other solid tumors and hematological tumors.
Such autoimmune diseases include, but are not limited to: rheumatoid arthritis, osteoarthritis, rheumatoid spondylitis, psoriasis, post-ischemic perfusion injury, inflammatory bowel disease, chronic inflammatory lung disease, eczema, asthma, psoriasis, ulcerative colitis, acute respiratory distress syndrome, psoriatic arthritis, infectious arthritis, progressive chronic arthritis, osteoarthritis deformans, femoral arthritis, traumatic arthritis, gouty arthritis, reiter's syndrome, polychondritis, acute synovitis and spondylitis, glomerulonephritis, hemolytic anemia, aplastic anemia, idiopathic thrombocytopenia, neutropenia, ulcerative colitis, crohn's disease, graft-versus-host disease, allograft rejection, chronic thyroiditis, graves ' disease, scleroderma, active hepatitis, primary biliary cirrhosis, myasthenia gravis, crohn's disease, graft-versus-host disease, allograft rejection, chronic thyroiditis, graves ' disease, scleroderma, active hepatitis, primary biliary cirrhosis, and other conditions, Multiple sclerosis, systemic lupus erythematosus, allergic dermatitis, contact dermatitis, chronic renal insufficiency, Step-Johnson syndrome, idiopathic steatorrhea, sarcoidosis, Guillain-Barre syndrome, pulmonary fibrosis or chronic inflammatory lung disease.
Definition of terms
The "compounds" described herein may not include all stereoisomers, geometric isomers, tautomers and isotopes.
The "compounds" described herein may include all stereoisomers, geometric isomers, tautomers and isotopes.
If stereoisomers exist, the "compound" of the present invention may exist as a single stereoisomer or a mixture thereof (e.g., as a racemate). The term "stereoisomer" refers to either a cis-trans isomer or an optical isomer. The stereoisomers can be separated, purified and enriched by an asymmetric synthesis method or a chiral separation method (including but not limited to thin layer chromatography, rotary chromatography, column chromatography, gas chromatography, high pressure liquid chromatography and the like), and can also be obtained by chiral resolution in a mode of forming bonds (chemical bonding and the like) or salifying (physical bonding and the like) with other chiral compounds and the like. The term "single stereoisomer" means that the mass content of one stereoisomer of the compound according to the invention is not less than 95% relative to all stereoisomers of the compound.
If tautomers exist, the "compounds" of the present invention may exist as single tautomers or as mixtures thereof, preferably as more stable tautomers, which result from the exchange of a single bond with an adjacent double bond and the accompanying migration of a proton.
The atoms in the "compounds" described herein may be present in their natural or unnatural abundance. In the case of hydrogen atoms, in its natural abundance, it is understood that about 99.985% is protium and about 0.015% is deuterium; in its unnatural abundance, it is meant that about 95% thereof is deuterium. That is, one or more atoms in the terms "compound," "pharmaceutically acceptable salt," "solvate," and "solvate of a pharmaceutically acceptable salt" can be an atom that is present in a non-natural abundance.
The term "pharmaceutically acceptable" means that the salts, solvents, excipients, etc., are generally non-toxic, safe, and suitable for use by the patient. The "patient" is preferably a mammal, more preferably a human.
The term "pharmaceutically acceptable salts" refers to salts prepared from the compounds of the present invention with relatively non-toxic, pharmaceutically acceptable acids. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of a pharmaceutically acceptable acid in neat solution or in a suitable inert solvent. The pharmaceutically acceptable acids include inorganic acids including, but not limited to: hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, phosphorous acid, sulfuric acid, hydrogen sulfate, and the like. The pharmaceutically acceptable acids include organic acids including, but not limited to: acetic acid, propionic acid, oxalic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, salicylic acid, tartaric acid, methanesulfonic acid, isonicotinic acid, acid citric acid, oleic acid, tannic acid, pantothenic acid, hydrogen tartrate, ascorbic acid, gentisic acid, fumaric acid, gluconic acid, saccharic acid, formic acid, ethanesulfonic acid, pamoic acid (i.e. 4, 4' -methylene-bis (3-hydroxy-2-naphthoic acid)), amino acids (e.g. glutamic acid, arginine), and the like. When the compounds of the present invention contain relatively basic functional groups, they may be converted to acid addition salts. See in particular Berge et al, "Pharmaceutical Salts", Journal of Pharmaceutical Science 66:1-19(1977), or, Handbook of Pharmaceutical Salts: Properties, Selection, and Use (P.Heinrich Stahl and Camile G.Wermuth, ed., Wiley-VCH, 2002).
In the present invention, the terms used have the following meanings, unless otherwise specified:
the term "halogen" means fluorine, chlorine, bromine or iodine, preferably fluorine or chlorine.
The term "hydroxy" refers to-OH.
The term "alkyl" denotes a straight or branched chain saturated hydrocarbon group consisting of carbon and hydrogen atoms, such as C1-C20Alkyl, preferably C1-C6Alkyl groups such as methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl or tert-butyl), pentyl (including n-pentyl, isopentyl, neopentyl), n-hexyl, 2-methylhexyl and the like.
The term "cycloalkyl" refers to a monovalent saturated cyclic alkyl group, preferably a monocyclic ring, such as cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
The term "heterocycloalkyl" refers to a saturated cyclic group having a heteroatom, containing 1 or more heteroatoms independently selected from N, O and S, preferably a monocyclic ring. Typically a 5-6 membered heterocyclyl group containing 1 or more heteroatoms independently selected from N, O and S, for example piperazinyl, morpholinyl, piperidinyl, or pyrrolidinyl.
The term "aryl" refers to an all-carbon aromatic group having a fully conjugated pi-electron system, which may be a single ring or a fused ring, generally having 6 to 14 carbon atoms, preferably having 6 to 12 carbon atoms, and most preferably having 6 carbon atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, and anthracenyl.
The term "heteroaryl" refers to an aromatic group containing a heteroatom, which may be a single ring or a fused ring, preferably a 5-12 membered heteroaryl group containing 1-4 heteroatoms independently selected from N, O and S, including but not limited to pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, quinolinyl, isoquinolinyl, triazolyl, tetrahydropyrrolyl.
By "treatment" is meant any treatment of a disease in a mammal, including: (1) preventing disease, i.e., the symptoms that cause clinical disease do not develop; (2) inhibiting disease, i.e., arresting the development of clinical symptoms; (3) alleviating the disease, i.e., causing regression of clinical symptoms.
"prevention" as used herein refers to a reduction in the risk of acquiring or developing a disease or disorder.
"pharmaceutical composition" as used herein, refers to a formulation of one or more compounds of the present invention or salts thereof with a carrier generally accepted in the art for delivery of biologically active compounds to an organism (e.g., a human). The purpose of the pharmaceutical composition is to facilitate delivery of the drug to an organism.
The term "pharmaceutically acceptable carrier" refers to a substance that is co-administered with, and facilitates the administration of, an active ingredient, including, but not limited to, any glidant, sweetener, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersant, disintegrant, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier that is acceptable for use in humans or animals (e.g., livestock) as permitted by the national food and drug administration. Examples include, but are not limited to, calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.
The pharmaceutical composition can be prepared into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, powder, granules, paste, emulsions, suspensions, solutions, suppositories, injections, inhalants, gels, microspheres, aerosols and the like.
The pharmaceutical compositions of the present invention may be manufactured by methods well known in the art, such as conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, lyophilizing, and the like.
The route of administration of the compounds of the present invention or pharmaceutically acceptable salts thereof or pharmaceutical compositions thereof includes, but is not limited to, oral, rectal, transmucosal, enteral, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration. The preferred route of administration is oral.
For oral administration, the pharmaceutical compositions may be formulated by mixing the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, slurries, suspensions and the like, for oral administration to a patient. For example, for pharmaceutical compositions intended for oral administration, tablets may be obtained in the following manner: the active ingredient is combined with one or more solid carriers, the resulting mixture is granulated if necessary, and processed into a mixture or granules, if necessary with the addition of small amounts of excipients, to form tablets or tablet cores. The core may be combined with an optional enteric coating material and processed into a coated dosage form more readily absorbed by an organism (e.g., a human).
On the basis of the common knowledge in the field, the above preferred conditions can be combined randomly to obtain the preferred embodiments of the invention.
The reagents and starting materials used in the present invention are commercially available.
The positive progress effects of the invention are as follows: the tricyclic compound provided by the invention has a novel structure, has a good inhibition effect on HDAC (Histone-like oxidase), particularly HDAC6, and can inhibit the proliferation of various tumor cells.
Drawings
Figure 1 is a tumor volume curve for 21 days of dosing.
Figure 2 is a graph of the rate of change of body weight of animals dosed for 21 days.
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The experimental methods without specifying specific conditions in the following examples were selected according to the conventional methods and conditions, or according to the commercial instructions.
In the preparation method of the target compound, the column chromatography adopts silica gel (300-400 meshes) produced by Ningsan Sun drying agent GmbH; thin layer chromatography using GF254(0.25 mm); nuclear magnetic resonance chromatography (NMR) was measured using a Varian-400 nuclear magnetic resonance spectrometer; LC/MS an Agilent technology ESI 6120 LC/MS instrument was used.
In addition, all operations involving easily oxidizable or hydrolyzable raw materials were carried out under nitrogen protection. Unless otherwise indicated, the starting materials used in the present invention are all commercially available starting materials and can be used without further purification.
Example 1:n- (7- (hydroxyamino) -7-oxoheptyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole- 2-carboxamides
Figure BDA0002858379420000151
Step 1: preparation of 1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylic acid tert-butyl ester
Phenylhydrazine hydrochloride (5.00g,34.6mmol) and tert-butyl 4-oxopiperidine-1-carboxylate (8.3g,4.15mmol) were added to acetic acid (50mL) and the reaction mixture was stirred at 65 ℃ for 16 h. The organic solvent was removed by concentration under reduced pressure. Water (50mL) was added to dilute the solution, and the solution was extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with saturated aqueous sodium bicarbonate (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product. The target compound (5.3g, yield 56.3%, yellow solid) was obtained by column chromatography (silica gel, petroleum ether: ethyl acetate: 10:1 to 5: 1). LC-MS (ESI) M/z [ M + H ]]+273.1。
Step 2: preparation of 2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (1.0g,3.67mmol) was dissolved in a mixed solvent of methanol (10mL) and tetrahydrofuran (10mL), and a methanol solution of hydrochloric acid (3M,10mL) was added. The reaction mixture was stirred at 40 ℃ for 1 hour. The reaction mixture was concentrated under reduced pressure to give a crude product (700mg, pale red solid, crude). Used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+173.1。
And step 3: preparation of methyl 7- (2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (373mg,2.30mmol) was dissolved in N, N-dimethylformamide (3mL), N-diisopropylethylamine (2.48g,19.2mmol) and methyl 7-aminoheptanoate hydrochloride (376mg,1.92mmol) were added, and the reaction mixture was stirred at room temperature for 0.5 hour, followed by addition of 2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] -amine]Indole hydrochloride (400mg,1.92mmol) in N, N-dimethylformamide (1 mL). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (30 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. The target compound (470mg, yield 68.6%, colorless oil) was obtained by column chromatography separation and purification (silica gel, ethyl acetate: petroleum ether ═ 20% to 100%). LC-MS (ESI) M/z [ M + H ]]+358.2。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Indole-2-carboxamido) heptanoic acid methyl ester (470mg, crude) was dissolved in a mixed solvent of methanol (15mL) and methylene chloride (7 mL). Aqueous sodium hydroxide (2N,5mL) and aqueous hydroxylamine (50%, 2.5mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 16 hours. The organic solvent was removed by concentration under reduced pressure. The mixture was adjusted to pH 7 with dilute hydrochloric acid (1N), filtered and the solid washed with water. The solid was purified by liquid phase preparative purification (0.1% HCl) to give the title compound (60mg, two step yield 8.73%, light yellow solid). LC-MS (ESI) M/z [ M + H ]]+359.2。1H NMR(400MHz,DMSO-d6)δ10.83(s,1H),10.32(br s,1H),8.65(br s,1H),7.34(d,J=7.6Hz,1H),7.28(d,J=8.0Hz,1H),7.02(t,J=6.8Hz,1H),6.95(t,J=7.6Hz,1H),6.57(t,J=5.2Hz,1H),4.48(s,2H),3.68(t,J=5.6Hz,2H),3.06-3.01(m,2H),2.75-2.72(m,2H),1.93(t,J=7.2Hz,2H),1.49-1.39(m,4H),1.24-1.23(m,4H)。
Example 2:n- (7- (hydroxyamino) -7-oxoheptyl) -5-methyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3- b]Indole-2-carboxamides
Figure BDA0002858379420000161
Step 1: preparation of tert-butyl 5-methyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (500mg,1.84mmol) was dissolved in N, N-dimethylformamide (5mL), and methyl iodide (287mg,2.02mmol) and potassium hydroxide (310mg, 5.52mmol) were added. The reaction mixture was stirred at 60 ℃ for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. The target compound (300mg, yield 57.0%, white solid) was obtained by column chromatography (silica gel, petroleum ether: ethyl acetate: 20:1 to 5: 1). LC (liquid Crystal)-MS(ESI)m/z[M+H]+287.2。
Step 2: preparation of 5-methyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5-methyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (250mg,0.875mmol) was dissolved in a mixed solvent of methanol (2mL) and tetrahydrofuran (2mL), and 1, 4-dioxane hydrochloride solution (4M,2.5mL) was added. The reaction mixture was stirred at room temperature for 0.5 hour. Concentration under reduced pressure gave the crude product (230mg, pale red solid, crude) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+187.3。
And step 3: preparation of methyl 7- (5-methyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (166mg,1.03mmol) was dissolved in N, N-dimethylformamide (2 mL). N, N-diisopropylethylamine (550mg,4.28mmol) and methyl 7-aminoheptanoate hydrochloride (167mg,0.855mmol) were added successively at 0 ℃. The reaction mixture was stirred at room temperature for 0.5 hour, and 5-methyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (190mg,0.855mmol) in N, N-dimethylformamide (1mL) and N, N-diisopropylethylamine (550mg,4.28 mmol). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product (240mg, crude, colorless oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+372.2。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5-methyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5-methyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Methyl indole-2-carboxamido) heptanoate (240mg, crude) was dissolved in a mixed solvent of methanol (8mL) and methylene chloride (4 mL). Aqueous sodium hydroxide (2N,6mL) and aqueous hydroxylamine (50%, 3mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 16 hours. Concentrating under reduced pressure to remove organic solvent, adjusting pH to 6 with dilute hydrochloric acid (1N), and filteringThe filter cake was washed with water (2 mL). The filter cake was purified by liquid phase preparative purification to give the title compound (34mg, 12.6% yield in three steps, pale green solid). LC-MS (ESI) M/z [ M + H ]]+373.2。1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),7.39-7.36(m,2H),7.11-7.07(m,1H),7.01-6.98(m,1H),6.61-6.60(m,1H),4.49(s,2H),3.71(t,J=5.6Hz,2H),3.61(s,3H),3.03-3.00(m,2H),2.78-2.75(m,2H),1.92(t,J=7.2Hz,2H),1.50-1.38(m,4H),1.24-1.22(m,4H)。
Example 3:n- (7- (hydroxyamino) -7-oxoheptyl) -5-isopropyl-1, 3,4, 5-tetrahydro-2H-pyrido [4, 3-b]indole-2-carboxamides
Figure BDA0002858379420000181
Step 1: preparation of tert-butyl 5-isopropyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (200mg,0.734mmol) was dissolved in N, N-dimethylformamide (5 mL). Sodium hydride (60%, 32.3mg,0.807mmol) was added at 0 ℃. The reaction mixture was stirred at 0 ℃ for half an hour and 2-iodopropane (137mg,0.807mmol) was added. The reaction mixture was stirred at room temperature for 16 hours. The reaction mixture was quenched by addition of water (10mL) at 0 ℃ and extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on a preparative plate (petroleum ether: ethyl acetate ═ 5:1) gave the title compound (120mg, yield 52.0%, colorless oil). LC-MS (ESI) M/z [ M + H ]]+315.2。
Step 2: preparation of 5-isopropyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Mixing 5-isopropyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (120mg,0.382mmol) was dissolved in methanol (3mL), and 1, 4-dioxane hydrochloride solution (4M,1mL) was added. The reaction mixture was stirred at room temperature for 1 hour. Concentration under reduced pressure gave the title compound (95mg, crude, pale red solid) which was used without purificationIn the next reaction. LC-MS (ESI) M/z [ M + H ]]+215.1。
And step 3: preparation of methyl 7- (5-isopropyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (68mg,0.420mmol) was dissolved in N, N-dimethylformamide (1 mL). Methyl 7-aminoheptanoate hydrochloride (81.9mg,0.420mmol) and N, N-diisopropylethylamine (246mg,1.91mmol) were added successively at 0 ℃. The reaction mixture was stirred at room temperature for 0.5 hour, and 5-methyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (95mg, crude, 0.382mmol) and N, N-diisopropylethylamine (246mg,1.91mmol) in N, N-dimethylformamide (3 mL). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (10 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on a preparative plate (silica gel, petroleum ether: ethyl acetate 4: 1) gave the title compound (80mg, 52.3% yield in two steps, pale yellow solid). LC-MS (ESI) M/z [ M + H ]]+400.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5-isopropyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5-isopropyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Indole-2-carboxamido) heptanoic acid methyl ester (80mg,0.200mmol) was dissolved in a mixed solvent of methanol (3mL) and dichloromethane (1.5 mL). Aqueous sodium hydroxide (2N,1mL) and aqueous hydroxylamine (50%, 0.5mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 16 hours. The organic solvent was removed by concentration under reduced pressure, the pH was adjusted to 6 with dilute hydrochloric acid (1N), the filtrate was filtered, and the cake was washed with water (2 mL). The filter cake was purified by liquid phase preparation (0.1% HCl) to give the title compound (22mg, yield 27.5%, off-white solid). LC-MS (ESI) M/z [ M + H ]]+401.2。1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),7.51(d,J=8.4Hz,1H),7.36(d,J=7.6Hz,1H),7.05(t,J=7.2Hz,1H),6.98(t,J=7.6Hz,1H),6.59(br s,1H),4.67-4.64(m,1H),4.47(s,2H),3.70(t,J=5.2Hz,2H),3.05-3.01(m,2H),2.82-2.80(m,2H),1.92(t,J=7.2Hz,2H),1.50-1.39(m,10H),1.24-1.23(m,4H)。
Example 4: n- (7- (hydroxyamino) -7-oxoheptyl) -5- (cyclopropylmethyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Figure BDA0002858379420000191
Step 1: preparation of tert-butyl 5- (cyclopropylmethyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) was dissolved in N, N-dimethylformamide (5 mL). Sodium hydride (60%, 88mg,2.20mmol) was added at 0 ℃ and the reaction was stirred for a further 30 minutes at 0 ℃. Bromomethylcyclopropane (223mg,1.65mmol) was added to the reaction solution. The reaction mixture was warmed to room temperature and stirred at room temperature overnight. The reaction mixture was added to ethyl acetate (50mL) and washed with saturated brine (30 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to give the crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate: 5:1) gave the title compound (200mg, yield 55.7%, white solid). LC-MS (ESI) M/z [ M + H ]]+327.2。
Step 2: preparation of 5- (cyclopropylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (cyclopropylmethyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (200mg,0.612mmol) was dissolved in methanol (2mL) and 1, 4-dioxane hydrochloric acid solution (4M,2mL) was added. The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure to give the title compound (200mg, crude, colorless oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+227.1。
And step 3: preparation of methyl 7- (5- (cyclopropylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
Methyl 7-aminoheptanoate hydrochloride (238mg,1.22mmol) was dissolved in N, N-dimethylformamide (2mL), and N, N-diisopropylethylamine (157mg,1.22 mm) was addedol) and N, N' -carbonyldiimidazole (198mg,1.22 mmol). The reaction mixture was stirred at room temperature for 30 minutes. Adding 5- (cyclopropylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b]Indole hydrochloride (200mg, crude, 0.612mmol) was dissolved in N, N-dimethylformamide (2mL) with N, N-diisopropylethylamine (790mg,6.12 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (50mL) and washed with saturated brine (50 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound (300mg, crude, colorless oil). LC-MS (ESI) M/z [ M + H ]]+412.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (cyclopropylmethyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- (cyclopropylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Indole-2-carboxamido) heptanoic acid methyl ester (300mg, crude, 0.612mmol) was dissolved in a mixed solvent of methanol (2mL) and dichloromethane (2 mL). An aqueous hydroxylamine solution (50%, 1mL) and a saturated methanol solution of sodium hydroxide (1mL) were added at 0 ℃. The reaction mixture was stirred at room temperature for 1 hour. To the mixture was added dilute hydrochloric acid (1N) to adjust pH to 7, and concentrated under reduced pressure to give a crude product. The crude product was prepared in liquid phase (0.1% FA) to give the title compound (48.3mg, 19.1% in three-step yield, white solid). LC-MS (ESI) M/z [ M + H ]]+413.3。1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),8.65(br s,1H),7.44(d,J=8.0Hz,1H),7.37(d,J=7.6Hz,1H),7.10-7.06(m,1H),6.99(t,J=7.4Hz,1H),6.60(t,J=5.2Hz,1H),4.50(s,2H),3.98(d,J=6.8Hz,2H),3.72(t,J=5.4Hz,2H),3.06-3.01(m,2H),2.79(t,J=4.8Hz,2H),1.92(t,J=7.2Hz,2H),1.52-1.37(m,4H),1.27-1.20(m,4H),1.15-1.06(m,1H),0.47-0.41(m,2H),0.35-0.31(m,2H)。
Example 5:n- (7- (hydroxyamino) -7-oxoheptyl) -5-ethyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3- b]Indole-2-carboxamides
Figure BDA0002858379420000201
Step 1: preparation of tert-butyl 5-ethyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
Sodium hydride (66mg,1.65mmol) was added portionwise to 1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] at 0 deg.C]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) in N, N-dimethylformamide (3 mL). The mixture was stirred at 0 ℃ for 0.5 h, and ethyl iodide (206mg,1.32mmol) was added. The reaction mixture was warmed to room temperature and stirred for 2 hours. The reaction mixture was quenched with ice water (20mL) and extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with saturated brine (50 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (325mg, crude, yellow oil). LC-MS (ESI) M/z [ M + H ]]+301.2。
Step 2: preparation of 5-ethyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole trifluoroacetate
Trifluoroacetic acid (1mL) was added to 5-ethyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] at room temperature]Indole-2-carboxylic acid tert-butyl ester (325mg,1.08mmol) in dichloromethane (2 mL). The reaction mixture was stirred at room temperature for 1 hour. The organic solvent was removed by concentration under reduced pressure to give a crude product of the objective compound (321mg, brown oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+201.0。
And step 3: preparation of methyl 7- (5-ethyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N-diisopropylethylamine (695mg,5.40mmol) and methyl 7-aminoheptanoate hydrochloride (211mg,1.08mmol) were added to a solution of N, N' -carbonyldiimidazole (210mg,1.30mmol) in N, N-dimethylformamide (2mL) at 0 ℃. The mixture was stirred at 0 ℃ for 0.5 hour. Reacting 5-ethyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]A mixture of indole trifluoroacetate (321mg,1.08mmol) and N, N-diisopropylethylamine (695mg,5.40mmol) in N, N-dimethylformamide (1mL) was added to the reaction mixture. The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (30mL) and extracted with ethyl acetate (30 mL. times.3). The combined organic phases were washed with saturated brine (50 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification by preparative plate (silicon)Gum, petroleum ether: ethyl acetate ═ 1:1) the title compound (175mg, 41.3% in three-step yield, yellow oil) was obtained. LC-MS (ESI) M/z [ M + H ]]+386.2。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5-ethyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Saturated sodium hydroxide methanol solution (2mL) and aqueous hydroxylamine solution (50%, 1mL) were added to 7- (5-ethyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] in that order at 0 deg.C]Indole-2-carboxamido) heptanoic acid methyl ester (175mg,0.454mmol) in dichloromethane (1 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure to remove the solvent, diluted with water (1mL), cooled in an ice bath, and adjusted to pH 4 with dilute hydrochloric acid (1N). The precipitated solid was filtered, prepared by liquid phase (0.1% HCOOH), and lyophilized to give the title compound (60.1mg, yield 34.3%, light yellow solid). LC-MS (ESI) M/z [ M + H ]]+387.3。1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),8.64(s,1H),7.41-7.36(m,2H),7.10-7.06(m,1H),7.01-6.97(m,1H),6.59(t,J=5.6Hz,1H),4.49(s,2H),4.10(q,J=7.2Hz,2H),3.71(t,J=6.0Hz,2H),3.03(q,J=6.8Hz,2H),2.77(t,J=5.2Hz,2H),1.92(t,J=7.2Hz,2H),1.48-1.37(m,4H),1.23-1.19(m,7H)。
Example 6:n- (7- (hydroxyamino) -7-oxoheptyl) -5- (cyclopentylmethyl) -1,3,4, 5-tetrahydro-2H-pyridine And [4,3-b ]]Indole-2-carboxamides
Figure BDA0002858379420000211
Step 1: preparation of tert-butyl 5- (cyclopentylmethyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (400mg,1.47mmol) was dissolved in N, N-dimethylformamide (10 mL). Sodium hydride (60%, 88mg,2.20mmol) was added at 0 ℃. The reaction mixture was stirred at 0 ℃ for half an hour before (bromomethyl) cyclopentane (264mg,1.62mmol) was added. The reaction mixture was stirred at room temperature for 6 hours. At 0 deg.C, adding to the reaction mixtureQuenched with water (20mL) and extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with saturated brine (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate: 10:1 to 5:1) gave the title compound (150mg, yield 28.8%, colorless oil). LC-MS (ESI) M/z [ M + H ]]+355.2。
Step 2: preparation of 5- (cyclopentylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (cyclopentylmethyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (150mg,0.423mmol) was dissolved in methanol (2mL), and 1, 4-dioxane hydrochloride solution (4M,2mL) was added. The reaction mixture was stirred at room temperature for 1 hour. Concentration under reduced pressure gave the title compound (100mg, crude, white solid) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+255.1。
And step 3: preparation of methyl 7- (5- (cyclopentylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (103mg,0.634mmol) was dissolved in N, N-dimethylformamide (2 mL). Methyl 7-aminoheptanoate hydrochloride (124mg,0.634mmol) and N, N-diisopropylethylamine (546mg,4.23mmol) were added successively at 0 ℃. The reaction mixture was stirred at room temperature for 0.5 hour, and 5- (cyclopentylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (100mg, crude, 0.423mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on a preparative plate (silica gel, petroleum ether: ethyl acetate ═ 2: 1) gave the target compound (100mg, yield in two steps 53.8%, colorless oil). LC-MS (ESI) M/z [ M + H ]]+440.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (cyclopentylmethyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- (cyclopentylmethyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Indole-2-carboxamido) heptanoic acid methyl ester (100mg,0.227mmol) was dissolved in a mixed solvent of methanol (1mL) and dichloromethane (1 mL). Aqueous sodium hydroxide (2N,0.5mL) and aqueous hydroxylamine (50%, 0.5mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 16 hours, and the organic solvent was removed by concentration under reduced pressure. The mixture was adjusted to pH 7 with dilute hydrochloric acid (1N), filtered and the filter cake washed with water (1 mL). The filter cake was purified by liquid phase preparation (0.1% HCl) to give the title compound (21mg, yield 21.0%, yellow solid). LC-MS (ESI) M/z [ M + H ]]+441.4。1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),7.42-7.36(m,2H),7.07(t,J=6.8Hz,1H),6.98(t,J=7.2Hz,1H),6.60-6.58(m,1H),4.49(s,2H),3.98(d,J=7.6Hz,2H),3.70(t,J=5.6Hz,2H),3.05-3.01(m,2H),2.77(t,J=4.4Hz,2H),2.33-2.26(m,1H),1.92(t,J=7.2Hz,2H),1.65-1.39(m,10H),1.27-1.23(m,6H)。
Example 7:n- (7- (hydroxyamino) -7-oxoheptyl) -5-isobutyl-1, 3,4, 5-tetrahydro-2H-pyrido [4, 3-b]indole-2-carboxamides
Figure BDA0002858379420000231
Step 1: preparation of tert-butyl 5-isobutyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
Sodium hydride (121mg,3.03mmol) was added portionwise to 1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] at 0 deg.C]Indole-2-carboxylic acid tert-butyl ester (550mg,2.02mmol) in N, N-dimethylformamide (6 mL). The reaction mixture was stirred at 0 ℃ for 0.5 hour. 1-iodo-2-methylpropane (446mg,2.42mmol) was added at 0 ℃. The reaction solution was heated to 80 ℃ and stirred for reaction overnight. The reaction mixture was cooled to room temperature, diluted with water (50mL) and extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with saturated brine (100 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate ═ 6: 1) gave the title compound (217mg, yield 32.7%, yellow oil). LC-MS (ESI) M/z [ M + H ]]+329.3。1H NMR(400MHz,DMSO-d6)δ7.40(d,J=8.0Hz,2H),7.08(t,J=8.0Hz,1H),6.98(t,J=7.6Hz,1H),4.53(s,2H),3.86(d,J=7.2Hz,2H),3.72(t,J=5.6Hz,2H),2.79(t,J=5.6Hz,2H),2.09-2.02(m,1H),1.43(s,9H),0.84(d,J=6.8Hz,6H)。
Step 2: preparation of 5-isobutyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole trifluoroacetate
Trifluoroacetic acid (1mL) was added to 5-isobutyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] at room temperature]Indole-2-carboxylic acid tert-butyl ester (217mg,0.661mmol) in dichloromethane (2 mL). The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure to give the objective compound (215mg, crude product, brown oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+229.0。
And step 3: 7- (5-isobutyl-2, 3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Preparation of indole-2-carboxamido) methyl heptanoate N, N-diisopropylethylamine (427mg,3.31mmol) and methyl 7-aminoheptanoate hydrochloride (129mg,0.661mmol) were added to a solution of N, N' -carbonyldiimidazole (129mg,0.793mmol) in N, N-dimethylformamide (2mL) at 0 ℃. The reaction mixture was stirred at 0 ℃ for 0.5 hour. At 0 ℃, reacting 5-isobutyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]A mixture of indole trifluoroacetate (215mg,0.661mmol) and N, N-diisopropylethylamine (427mg,3.31mmol) in N, N-dimethylformamide (1mL) was added to the reaction mixture. The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (30mL) and extracted with ethyl acetate (30 mL. times.3). The combined organic phases were washed with saturated brine (100 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (250mg, crude, yellow oil). LC-MS (ESI) M/z [ M + H ]]+414.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5-isobutyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Saturated sodium hydroxide in methanol (1mL) and aqueous hydroxylamine (50%, 1mL) were added to 7- (5-isobutyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] in that order at room temperature]Indole-2-carboxamido) heptanoic acid methyl ester (250mg,0.605mmol) In dichloromethane (1 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure to remove the solvent, and diluted with water (1 mL). The reaction was cooled in an ice bath and adjusted to pH 4 with dilute hydrochloric acid (1N). The precipitated solid was filtered off, and the title compound (75.9mg, yield in three steps, 27.7%, white solid) was obtained via liquid phase preparation (0.1% HCOOH). LC-MS (ESI) M/z [ M + H ]]+415.3。1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),8.64(s,1H),7.40-7.38(m,2H),7.09-7.05(m,1H),7.00-6.97(m,1H),6.59(t,J=5.2Hz,1H),4.97(s,2H),3.86(d,J=7.2Hz,2H),3.70(t,J=5.6Hz,2H),3.06-3.01(m,2H),2.76(t,J=5.2Hz,2H),2.07-2.03(m,1H),1.92(t,J=7.2Hz,2H),1.48-1.39(m,4H),1.24-1.22(m,4H),0.84(d,J=6.4Hz,6H)。
Example 8:n- (7- (hydroxyamino) -7-oxoheptyl) -5- ((tetrahydrofuran-3-yl) methyl) -1,3,4, 5-tetra-ethyl Hydrogen-2H-pyrido [4,3-b]Indole-2-carboxamides
Figure BDA0002858379420000241
Step 1: preparation of 3- (bromomethyl) tetrahydrofuran
(tetrahydrofuran-3-yl) methanol (2.0g,19.6mmol) and carbon tetrabromide (7.9g,24.0mmol) were dissolved in dichloromethane (50mL), and a solution of triphenylphosphine (7.9g,30.0mmol) in dichloromethane (50mL) was added. The reaction mixture was stirred at room temperature overnight. And concentrating the reaction solution under reduced pressure to obtain a crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate: 10:1 to 20:1) gave the title compound (1.3g, yield 40.1%, colorless oil).1H NMR(400MHz,CDCl3)δ3.93-3.87(m,2H),3.81-3.75(m,1H),3.61-3.58(m,1H),3.42-3.36(m,2H),2.71-2.68(m,1H),2.15-2.10(m,1H),1.67-1.71(m,1H)。
Step 2: preparation of tert-butyl 5- ((tetrahydrofuran-3-yl) methyl) -1,3,4, 5-tetrahydro-2H-pyridine [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (500mg,1.83mmol) was dissolved in N, N-dimethylformamide (10 mL). Bringing the reaction solution to 0 ℃ and thenSodium hydride (60%, 120mg,3.00mmol) was added under argon. The reaction mixture was stirred at 0 ℃ for 30 minutes. 3- (bromomethyl) tetrahydrofuran (500mg,3.03mmol) was added to the reaction mixture. The reaction mixture was allowed to warm to room temperature and stirred overnight. The reaction mixture was diluted with water (50mL) and extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on a preparative plate (silica gel, petroleum ether: ethyl acetate ═ 5:1) gave the title compound (110mg, yield 16.8%, yellow solid). LC-MS (ESI) M/z [ M + H ]]+357.2。
And step 3: preparation of 5- ((tetrahydrofuran-3-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (tetrahydrofuran-3-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (110mg,0.309mmol) was added to a solution of 1, 4-dioxane hydrochloride (4M,5 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure to give the title compound (100mg, crude, colorless oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+257.2。
And 4, step 4: preparation of methyl 7- (5- ((tetrahydrofuran-3-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
Methyl 7-aminoheptanoate hydrochloride (98mg,0.500mmol) was dissolved in N, N-dimethylformamide (3mL), and N, N-diisopropylethylamine (132mg,1.026mmol) and N, N' -carbonyldiimidazole (81mg,0.500mmol) were added. The reaction mixture was stirred at room temperature for 1 hour. Adding 5- ((tetrahydrofuran-3-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Indole hydrochloride (100mg, crude, 0.342mmol) and N, N-diisopropylethylamine (132mg,1.026mmol) in N, N-dimethylformamide (3 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (50mL) and extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on preparative plates (silica gel, petroleum ether: ethyl acetate ═ 1:2) gave the title compound (90mg, two-step yield 59).7% yellow oil). LC-MS (ESI) M/z [ M + H ]]+442.3。
And 5: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- ((tetrahydrofuran-3-yl) methyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- ((tetrahydrofuran-3-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Indole-2-carboxamido) heptanoic acid methyl ester (90mg,0.204mmol) was dissolved in a mixed solvent of methanol (3mL) and dichloromethane (3 mL). An aqueous hydroxylamine solution (50%, 2mL) and a saturated methanol solution of sodium hydroxide (2mL) were added at 0 ℃. The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure, and diluted hydrochloric acid (1N) was added to the mixture to adjust the pH to 6. Extraction was carried out with ethyl acetate (20 mL. times.3). The organic phases are combined and concentrated under reduced pressure to obtain a crude product. The title compound (26.6mg, yield 29.4%, yellow solid) was prepared via liquid phase (0.1% FA). LC-MS (ESI) M/z [ M + H ]]+443.3。1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),8.64(s,1H),7.44(d,J=8.0Hz,1H),7.38(d,J=8.0Hz,1H),7.10(t,J=7.6Hz,1H),7.01(t,J=7.4Hz,1H),6.59(t,J=5.4Hz,1H),4.52-4.43(m,2H),4.04(d,J=7.6Hz,2H),3.87-3.82(m,1H),3.75-3.66(m,2H),3.65-3.59(m,1H),3.56-3.52(m,1H),3.41-3.38(m,1H),3.03(q,J=6.8Hz,2H),2.78(t,J=4.4Hz,2H),2.70-2.66(m,1H),1.92(t,J=7.4Hz,2H),1.88-1.83(m,1H),1.61-1.57(m,1H),1.48-1.39(m,4H),1.24-1.23(m,4H)。
Example 9:n- (7- (hydroxyamino) -7-oxoheptyl) -5- ((tetrahydro-2H-pyran-4-yl) methyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxamides
Figure BDA0002858379420000261
Step 1: preparation of tert-butyl 5- ((tetrahydro-2H-pyran-4-yl) methyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) was dissolved in N, N-dimethylformamide (5 mL). Sodium hydride (60%, 66mg,1.65mmol) was added at 0 ℃. The reaction mixture isAfter stirring the reaction at 0 ℃ for half an hour, 4- (bromomethyl) tetrahydro-2H-pyran (217mg,1.21mmol) was added. The reaction mixture was stirred at room temperature for 16 hours. Water (20mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Purification by column chromatography (petroleum ether: ethyl acetate: 10:1 to 5:1) gave the title compound (170mg, yield 41.7%, colorless oil). LC-MS (ESI) M/z [ M + H ]]+371.3。
Step 2: preparation of 5- ((tetrahydro-2H-pyran-4-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- ((tetrahydro-2H-pyran-4-yl) methyl) -1,3,4, 5-tetrahydro-2H-pyrido [4, 3-b)]Indole-2-carboxylic acid tert-butyl ester (170mg,0.459mmol) was dissolved in methanol (2mL), and 1, 4-dioxane hydrochloride solution (4M,2mL) was added. The reaction mixture was stirred at room temperature for 1 hour. Concentration under reduced pressure gave the title compound (140mg, crude, red solid) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+271.1。
And step 3: preparation of methyl 7- (5- ((tetrahydro-2H-pyran-4-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (111mg,0.688mmol) was dissolved in N, N-dimethylformamide (2 mL). Methyl 7-aminoheptanoate hydrochloride (135mg,0.688mmol) and N, N-diisopropylethylamine (592mg,4.59mmol) were added successively at 0 ℃. The reaction mixture was stirred at room temperature for 0.5 hour, and 5- ((tetrahydro-2H-pyran-4-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (140mg, crude) in N, N-dimethylformamide (3 mL). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on preparative plates (silica gel, ethyl acetate) gave the title compound (120mg, 57.4% yield in two steps, yellow solid). LC-MS (ESI) M/z [ M + H ]]+456.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- ((tetrahydro-2H-pyran-4-yl) methyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- ((tetrahydro-2H-pyran-4-yl) methyl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Indole-2-carboxamido) heptanoic acid methyl ester (120mg,0.264mmol) was dissolved in a mixed solvent of methanol (3mL) and dichloromethane (3 mL). Aqueous hydroxylamine (50%, 1mL) and saturated methanolic sodium hydroxide (1mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 3 hours. The organic solvent was removed by concentration under reduced pressure, the mixture was adjusted to pH 7 with dilute hydrochloric acid (1N), filtered, and the cake was washed with water (2 mL). The filter cake was dried in vacuo to give the title compound (28mg, yield 23.2%, yellow solid). LC-MS (ESI) M/z [ M + H ]]+457.4。1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),8.65(s,1H),7.44(d,J=8.0Hz,1H),7.37(d,J=7.6Hz,1H),7.08(t,J=7.2Hz,1H),6.99(t,J=7.6Hz,1H),6.59(t,J=5.2Hz,1H),4.49(s,2H),3.95(d,J=7.2Hz,2H),3.80-3.78(m,2H),3.70(t,J=5.6Hz,2H),3.20-3.15(m,2H),3.06-3.01(m,2H),2.81-2.74(m,2H),1.97-1.90(m,3H),1.48-1.23(m,12H)。
Example 10:n- (7- (hydroxyamino) -7-oxoheptyl) -5-phenyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3- b]Indole-2-carboxamides
Figure BDA0002858379420000271
Step 1: preparation of tert-butyl 5-phenyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Tert-butyl indole-2-carboxylate (200mg,0.882mmol) and iodobenzene (180mg,0.375mmol) were dissolved in toluene (6mL), and potassium phosphate (311mg,1.47mmol), cuprous iodide (14.0mg,0.0375mmol) and N, N' -dimethylethylenediamine (12.9mg,0.147mmol) were added. The reaction mixture was stirred at 150 ℃ for 16 hours. The reaction solution was cooled to room temperature. The reaction mixture was diluted with water (10mL) and extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressureConcentrating to obtain a crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate: 20:1 to 10:1) gave the title compound (230mg, 89.9% yield, colorless oil). LC-MS (ESI) M/z [ M + H ]]+349.3。
Step 2: preparation of 5-phenyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5-phenyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (230mg,0.660mmol) was dissolved in methanol (2mL), and 1, 4-dioxane hydrochloride solution (3M,2mL) was added. The reaction mixture was stirred at room temperature for 1 hour. Concentration under reduced pressure gave the crude product (189mg, crude, light red solid) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+249.1。
And step 3: preparation of methyl 7- (5-phenyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (215mg,1.33mmol) was dissolved in N, N-dimethylformamide (2mL), and N, N-diisopropylethylamine (428mg,3.32mmol) and methyl 7-aminoheptanoate hydrochloride (130mg,0.663mmol) were added at 0 ℃. Followed by the addition of 5-phenyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Indole hydrochloride (189mg,0.663mmol) in N, N-dimethylformamide (3mL) and N, N-diisopropylethylamine (428mg,3.32 mmol). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on preparative plates (petroleum ether: ethyl acetate ═ 5:1) gave the title compound (60mg, 21.0% yield in two steps, colorless oil). LC-MS (ESI) M/z [ M + H ]]+434.4。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5-phenyl-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5-phenyl-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Indole-2-carboxamido) heptanoic acid methyl ester (60mg,0.138mmol) was dissolved in a mixed solvent of methanol (3mL) and dichloromethane (1.5 mL). Aqueous sodium hydroxide (2N,1mL) and aqueous hydroxylamine were added sequentially at 0 deg.C(50%, 0.5 mL). The reaction mixture was stirred at room temperature for 16 hours. The organic solvent was removed by concentration under reduced pressure, the pH was adjusted to 6 with dilute hydrochloric acid (1N), the filtrate was filtered, and the cake was washed with water (1 mL). The filter cake was added to methanol (1mL), stirred at room temperature for 0.5 h, filtered and the filter cake was dried under vacuum to give the title compound (20mg, yield 33.4%, off-white solid). LC-MS (ESI) M/z [ M + H ]]+435.4。1H NMR(400MHz,DMSO-d6)δ10.33(s,1H),8.66(d,J=1.6Hz,1H),7.60-7.56(m,2H),7.49-7.44(m,4H),7.22-7.20(m,1H),7.11-7.09(m,2H),6.63(t,J=5.2Hz,1H),4.58(s,2H),3.68(t,J=5.2Hz,2H),3.08-3.03(m,2H),2.64-2.63(m,2H),1.93(t,J=7.2Hz,2H),1.50-1.41(m,4H),1.25-1.24(m,4H)。
Example 11:n- (7- (hydroxyamino) -7-oxoheptyl) -5- (pyrimidin-2-yl) -1,3,4, 5-tetrahydro-2H-pyridine And [4,3-b ]]Indole-2-carboxamides
Figure BDA0002858379420000291
Step 1: preparation of 5- (pyrimidin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylic acid tert-butyl ester
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) was dissolved in1, 4-dioxane (10mL), and 2-chloropyrimidine (138mg,1.21mmol), cesium carbonate (715mg,2.20mmol), tris (dibenzylideneacetone) dipalladium (50mg,0.055mmol) and 2-dicyclohexylphosphonium-2, 4, 6-triisopropylbiphenyl (52mg,0.110mmol) were added. The reaction mixture was warmed to 100 ℃ under argon and stirred overnight. The reaction mixture was cooled to room temperature, poured into water (100mL), and extracted with ethyl acetate (50mL x 3). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate: 1:0 to 3:1) gave the title compound (130mg, yield 33.7%, white solid). LC-MS (ESI) M/z [ M + H ]]+351.3。
Step 2: preparation of 5- (pyrimidin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Will 5-(pyrimidin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (130mg,0.371mmol) was dissolved in1, 4-dioxane (2mL) and 1, 4-dioxane hydrochloric acid solution (4M,2mL) was added. The reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure to give the title compound (150mg, crude, colorless oil) which was used directly in the next reaction. LC-MS (ESI) M/z [ M + H ]]+251.2。
And step 3: preparation of methyl 7- (5- (pyrimidin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
Methyl 7-aminoheptanoate hydrochloride (95mg,0.482mmol) was dissolved in N, N-dimethylformamide (2mL), and N, N-diisopropylethylamine (62mg,0.482mmol) and N, N' -carbonyldiimidazole (78mg,0.482mmol) were added. After the reaction mixture was stirred at room temperature for 30 minutes, 5- (pyrimidin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (150mg, crude) and N, N-diisopropylethylamine (478mg,3.71mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (50mL) and washed with saturated brine (50 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (200mg, crude, colorless oil) which was used directly in the next reaction. LC-MS (ESI) M/z [ M + H ]]+436.2。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (pyrimidin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- (pyrimidin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Methyl indole-2-carboxamido) heptanoate (200mg, crude) was dissolved in a mixed solvent of methanol (2mL) and methylene chloride (1 mL). An aqueous hydroxylamine solution (50%, 1mL) and a saturated methanol solution of sodium hydroxide (1mL) were added at 0 ℃. The reaction mixture was stirred at room temperature for 1 hour. To the mixture was added dilute hydrochloric acid (1N) to adjust pH to 7, and the mixture was concentrated under reduced pressure to obtain a crude product. The crude product was prepared via liquid phase (0.1% formic acid) and the preparation was lyophilized to give the title compound (24.91mg, 15.3% total yield over three steps, white solid). LC-MS (ESI) M/z [ M + H ]]+437.3。1H NMR(400MHz,DMSO-d6)δ10.33(s,1H),8.89(d,J=4.8Hz,2H),8.66(brs,1H),8.45(d,J=8.0Hz,1H),7.48(d,J=7.2Hz,1H),7.38(t,J=4.8Hz,1H),7.27-7.19(m,2H),6.66(t,J=5.2Hz,1H),4.52(s,2H),3.67(t,J=5.2Hz,2H),3.18(s,2H),3.08-3.04(m,2H),1.93(t,J=7.2Hz,2H),1.50-1.42(m,4H),1.26-1.25(m,4H)。
Example 12:n- (7- (hydroxyamino) -7-oxoheptyl) -5- (pyridin-2-yl) -1,3,4, 5-tetrahydro-2H-pyridine And [4,3-b ]]Indole-2-carboxamides
Figure BDA0002858379420000301
Step 1: preparation of tert-butyl 5- (pyridin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) was dissolved in1, 4-dioxane (10mL), and 2-bromopyridine (191mg,1.21mmol), cesium carbonate (715mg,2.20mmol), tris (dibenzylideneacetone) dipalladium (50mg,0.055mmol) and 4, 5-bis diphenylphosphine-9, 9-dimethylxanthene (52mg,0.11mmol) were added. The reaction mixture was stirred at 100 ℃ overnight under argon. After the reaction mixture was cooled, ethyl acetate (50mL) was added, followed by washing with saturated saline (30mL x 3). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to give the crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate 5:1) gave the title compound (200mg, yield 52.1%, white solid). LC-MS (ESI) M/z [ M + H ]]+350.2。
Step 2: preparation of 5- (pyridin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (pyridin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (200mg,0.573mmol) was dissolved in1, 4-dioxane (2mL), and 1, 4-dioxane hydrochloric acid solution (4M,2mL) was added. The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure to give the title compound (200mg, crude, brown oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+250.2。
And step 3: preparation of methyl 7- (5- (pyridin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N-diisopropylethylamine (96mg,0.745mmol), N, N' -carbonyldiimidazole (120mg,0.745mmol) and methyl 7-aminoheptanoate hydrochloride (146mg,0.745mmol) were dissolved in N, N-dimethylformamide (2mL), and the reaction mixture was stirred at room temperature for 30 minutes and then 5- (pyridin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (200mg, crude, 0.573mmol) and N, N-diisopropylethylamine (740mg,5.73mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (50mL) and washed with saturated brine (50 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to give the title compound (200mg, crude, brown oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+435.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (pyridin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- (pyridine-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Indole-2-carboxamido) heptanoic acid methyl ester (200mg, crude) was dissolved in a mixed solvent of methanol (2mL) and dichloromethane (2 mL). An aqueous hydroxylamine solution (50%, 1mL) and a saturated methanol solution of sodium hydroxide (1mL) were added at 0 ℃. The reaction mixture was stirred at room temperature for 1 hour. To the mixture was added dilute hydrochloric acid (1N) to adjust pH to 7, and the mixture was concentrated under reduced pressure to obtain a crude product. The crude product was passed through a liquid phase (0.1% NH)4OH) to yield the title compound (51.14mg, 20.5% in three-step yield, white solid). LC-MS (ESI) M/z [ M + H ]]+436.2。1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),8.65-8.62(m,2H),8.04(td,J=8.0,1.8Hz,1H),7.64-7.58(m,2H),7.50-7.48(m,1H),7.43-7.40(m,1H),7.19-7.12(m,2H),6.64(t,J=5.2Hz,1H),4.59(s,2H),3.67(t,J=5.4Hz,2H),3.08-3.04(m,2H),2.86-2.85(m,2H),1.94(t,J=7.2Hz,2H),1.50-1.42(m,4H),1.26-1.25(m,4H)。
Example 13:n- (7- (hydroxyamino) -7-oxoheptyl) -5- (4-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyridine And [4,3-b ]]Indole-2-carboxamides
Figure BDA0002858379420000311
Step 1: preparation of tert-butyl 5- (4-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) (MC 18-663-. The reaction mixture was stirred at 150 ℃ overnight under nitrogen. The reaction mixture was cooled to room temperature, poured into water (100mL), and extracted with ethyl acetate (50mL x 3). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate: 1:0 to 5:1) gave the title compound (130mg, yield 32.3%, white solid). LC-MS (ESI) M/z [ M + H ]]+367.2。
Step 2: preparation of 5- (4-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (4-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (130mg,0.354mmol) was dissolved in1, 4-dioxane (2mL) and 1, 4-dioxane hydrochloric acid solution (4M,2mL) was added. The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure to give the objective compound (110mg, crude, colorless oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+267.3。
And step 3: preparation of methyl 7- (5- (4-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (73mg,0.459mmol), N, N-diisopropylethylamine (58mg,0.459mmol) and methyl 7-aminoheptanoate hydrochloride (88mg,0.459mmol) were added to N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature for 30 minutes and then 5- (4-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyridine was addedAnd [4,3-b ]]Indole hydrochloride (110mg, crude, 0.354mmol) and N, N-diisopropylethylamine (451mg,3.54mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (50mL) and washed with saturated brine (50 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound (200mg, crude, colorless oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+452.2。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (4-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- (4-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Indole-2-carboxamido) heptanoic acid methyl ester (200mg, crude, 0.35mmol) was dissolved in a mixed solvent of methanol (2mL) and dichloromethane (2 mL). An aqueous hydroxylamine solution (50%, 1mL) and a saturated methanol solution of sodium hydroxide (2mL) were added at 0 ℃. The reaction mixture was stirred at room temperature for 1 hour. To the mixture was added dilute hydrochloric acid (2N) to adjust pH to 7, and the mixture was concentrated under reduced pressure to obtain a crude product. Lyophilization over liquid phase (0.1% formic acid) preparation yielded the title compound (5.16mg, 3.22% yield over three steps, white solid). LC-MS (ESI) M/z [ M + H ]]+453.3。1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),8.65(s,1H),7.53-7.47(m,3H),7.44-7.39(m,2H),7.17-7.09(m,3H),6.62(t,J=5.2Hz,1H),4.58(s,2H),3.68(t,J=5.2Hz,2H),3.07-3.02(m,2H),2.62-2.59(m,2H),1.93(t,J=7.2Hz,2H),1.49-1.40(m,4H),1.28-1.20(m,4H)。
Example 14:n- (7- (hydroxyamino) -7-oxoheptyl) -5- (2-chlorophenyl) -1,3,4, 5-tetrahydro-2H-pyridine And [4,3-b ]]Indole-2-carboxamides
Figure BDA0002858379420000331
Step 1: preparation of tert-butyl 5- (2-chlorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (2.0g,7.35mmol) and 1-chloro-2-iodoBenzene (3.50g,14.7mmol) was dissolved in N, N-dimethylformamide (20mL), and potassium phosphate (3.12g,14.7mmol), cuprous iodide (279mg,1.47mmol) and L-proline (338mg,2.94mmol) were added. The reaction mixture was stirred at 150 ℃ for 16 hours under argon. The reaction solution was cooled to room temperature. Water (30mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate: 10:1 to 5:1) gave the title compound (102mg, yield 3.62%, colorless oil). LC-MS (ESI) M/z [ M + H ]]+383.3。
Step 2: preparation of 5- (2-chlorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (2-chlorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (102mg,0.266mmol) was dissolved in methanol (1mL), 1, 4-dioxane hydrochloride solution (4M,1mL) was added, and the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure to give the objective compound (84mg, crude, off-white solid) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+283.2。
And step 3: preparation of methyl 7- (5- (2-chlorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (47.5mg,0.293mmol) was dissolved in N, N-dimethylformamide (1mL), and N, N-diisopropylethylamine (343mg,2.66mmol) and methyl 7-aminoheptanoate hydrochloride (57.3mg,0.293mmol) were added at 0 ℃. The reaction mixture was stirred at room temperature for half an hour and then 5- (2-chlorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (84mg, crude, 0.266mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on preparative plates (silica gel, petroleum ether: ethyl acetate 1:1) gave the title compound (62mg, two-step yield)Yield 49.8%, colorless oil). LC-MS (ESI) M/z [ M + H ]]+468.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (2-chlorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- (2-chlorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Indole-2-carboxamido) heptanoic acid methyl ester (62mg,0.132mmol) was dissolved in a mixed solvent of methanol (1mL) and dichloromethane (1mL), and aqueous sodium hydroxide solution (2N,1mL) and aqueous hydroxylamine solution (50%, 0.5mL) were added at 0 ℃. The reaction mixture was stirred at room temperature for 16 hours. The reaction was concentrated under reduced pressure and the crude product was adjusted to pH 7 with dilute hydrochloric acid (1N). Filtration and washing of the filter cake with water (1 mL). The filter cake was isolated and purified by prep (0.1% HCl) to give the title compound (11.85mg, yield 19.1%, light yellow solid). LC-MS (ESI) M/z [ M + H ]]+469.1。1H NMR(400MHz,DMSO-d6)δ10.33(s,1H),7.76-7.74(m,1H),7.60-7.48(m,4H),7.10-7.08(m,2H),6.64-6.62(m,1H),6.63(br s,1H),4.63-4.54(m,2H),3.69-3.61(m,2H),3.12-3.04(m,2H),2.43-2.36(m,2H),1.92(t,J=7.2Hz,2H),1.47-1.41(m,4H),1.30-1.17(m,4H)。
Example 15:n- (7- (hydroxyamino) -7-oxoheptyl) -5- (pyrazin-2-yl) -1,3,4, 5-tetrahydro-2H-pyridine And [4,3-b ]]Indole-2-carboxamides
Figure BDA0002858379420000341
Step 1: preparation of 5- (pyrazin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylic acid tert-butyl ester
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) was dissolved in N, N-dimethylformamide (10mL), and 2-bromopyrazine (192mg,1.21mmol), cesium carbonate (715mg,2.20mmol), cuprous iodide (25mg,0.220mmol) and L-proline (42mg,0.220mmol) were added. The reaction mixture was stirred at 120 ℃ for 4 hours under argon. After the reaction mixture was cooled, ethyl acetate (50mL) was added and the mixture was washed with saturated brine. The organic phase was dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to give the crude product. By column chromatographySeparation and purification (silica gel, petroleum ether: ethyl acetate ═ 5:1) gave the title compound (200mg, yield 51.8%, white solid). LC-MS (ESI) M/z [ M + H ]]+351.2。
Step 2: preparation of 5- (pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (pyrazin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4, 3-b)]Indole-2-carboxylic acid tert-butyl ester (200mg,0.571mmol) was dissolved in1, 4-dioxane (2mL) and 1, 4-dioxane hydrochloric acid solution (4M,2mL) was added. The reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure to give the title compound (200mg, crude, colorless oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+251.2。
And step 3: preparation of methyl 7- (5- (pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (120mg,0.742mmol) was dissolved in N, N-dimethylformamide (2mL), and methyl 7-aminoheptanoate hydrochloride (145mg,0.742mmol) and N, N-diisopropylethylamine (96mg,0.742mmol) were added. The reaction mixture was stirred at room temperature for 30 minutes and 5- (pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] was added]Indole hydrochloride (200mg, crude, 0.571mmol) and N, N-diisopropylethylamine (738mg,5.71mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (50mL) and washed with saturated brine (50 mL. times.3). The organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give the title compound (300mg, crude, brown oil) which was used in the next reaction without purification. LC-MS (ESI) M/z [ M + H ]]+436.2。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (pyrazin-2-yl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5- (pyrazin-2-yl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Methyl indole-2-carboxamido) heptanoate (300mg, crude, 0.571mmol) was dissolved in a mixed solvent of methanol (2mL) and dichloromethane (2 mL). An aqueous hydroxylamine solution (50%, 1mL) and a saturated solution of sodium hydroxide in methanol (1mL) were added at 0 deg.C. The reaction mixture was stirred at room temperature for 1 hour. To the mixture was added dilute hydrochloric acid (1N) to adjust pH to 7, and the mixture was concentrated under reduced pressure to obtain a crude product. The crude product was prepared via liquid phase, and the preparation was lyophilized to give the title compound (9.62mg, 3.8% yield in three steps, white solid). LC-MS (ESI) M/z [ M + H ]]+437.2。1H NMR(400MHz,DMSO-d6)δ10.33(br s,1H),8.95(s,1H),8.71-8.70(m,1H),8.66-8.61(m,2H),7.72-7.68(m,1H),7.52-7.50(m,1H),7.23-7.16(m,2H),6.66(t,J=5.6Hz,1H),4.59(s,2H),3.68(t,J=5.4Hz,2H),3.08-3.01(m,2H),2.90-2.88(m,2H),1.93(t,J=7.4Hz,2H),1.52-1.41(m,4H),1.28-1.21(m,4H)。
Example 16:n- (7- (hydroxyamino) -7-oxoheptyl) -5- (2-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyridine And [4,3-b ]]Indole-2-carboxamides
Figure BDA0002858379420000351
Step 1: preparation of tert-butyl 5- (2-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (300mg,1.10mmol) was dissolved in N, N-dimethylformamide (3mL), and 1-fluoro-2-iodobenzene (268mg,1.21mmol), cesium carbonate (1.075mg,3.30mmol), cuprous iodide (27mg,0.14mmol) and L-proline (152mg,1.32mmol) were added. The reaction mixture was stirred at 120 ℃ overnight under argon. After completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water (10mL), and extracted with ethyl acetate (10 mL. times.3). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to give the crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate 10:1) gave the title compound (91mg, yield 22.5%, oily liquid). LC-MS (ESI) M/z [ M + H-56]+311.1。
Step 2: preparation of 5- (2-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5- (2-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxylic acid tert-butyl ester (91mg,0.248mmol) was dissolved in1, 4-dioxane (3mL) and added1, 4-dioxane hydrochloric acid solution (4M,2mL) was added. The reaction mixture was stirred at room temperature overnight. After completion of the reaction, the reaction solution was concentrated under reduced pressure to give the objective compound (76mg, crude product, colorless oil). LC-MS (ESI) M/z [ M + H ]]+267.3。
And step 3: preparation of methyl 7- (5- (2-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
Methyl 7-aminoheptanoate hydrochloride (11mg,0.060mmol) was dissolved in N, N-dimethylformamide (3mL), and N, N-diisopropylethylamine (32mg,0.250mmol) and N, N' -carbonyldiimidazole (10mg,0.060mmol) were added at 0 ℃. The reaction mixture was stirred at room temperature for 30 minutes. Then adding 5- (2-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b]Indole hydrochloride (15mg,0.044mmol, crude) was dissolved in N, N-diisopropylethylamine (32mg,0.250mmol) in N, N-dimethylformamide (1 mL). The reaction mixture was stirred at room temperature overnight. Water (10mL) was added for dilution, followed by extraction with ethyl acetate (20 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to give the title compound (23mg, crude, colorless oil). LC-MS (ESI) M/z [ M + H ]]+452.2。
Methyl 7-aminoheptanoate hydrochloride (51mg,0.260mmol) was dissolved in N, N-dimethylformamide (3mL), and N, N-diisopropylethylamine (129mg,0.250mmol) and N, N' -carbonyldiimidazole (39mg,0.240mmol) were added at 0 ℃. The reaction mixture was stirred at room temperature for 30 minutes. Then adding 5- (2-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4,3-b]Indole hydrochloride (61mg,0.176mmol, crude) was dissolved in N, N-diisopropylethylamine (129mg,1.00mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature overnight. Water (15mL) was added for dilution, followed by extraction with ethyl acetate (15 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated to give the title compound (90mg, crude, colorless oil). LC-MS (ESI) M/z [ M + H ]]+452.2。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5- (2-fluorophenyl) -1,3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
7- (5- (4-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyridinePyrido [4,3-b]Methyl indole-2-carboxamido) heptanoate (23mg, crude) was dissolved in a mixed solvent of methanol (1mL) and methylene chloride (1 mL). At 0 deg.C, an aqueous hydroxylamine solution (50 wt%, 1.5mL) and a saturated methanol solution of sodium hydroxide (1mL) were added. The reaction mixture was stirred at room temperature overnight. The organic solvent was removed by concentration under reduced pressure, diluted hydrochloric acid (1N) was added to the mixture to adjust pH to 7, and the mixture was concentrated under reduced pressure to obtain a crude product. LC-MS (ESI) M/z [ M + H ]]+453.3。
Reacting 7- (5- (2-fluorophenyl) -2,3,4, 5-tetrahydro-1H-pyrido [4, 3-b)]Methyl indole-2-carboxamido) heptanoate (90mg, crude) was dissolved in a mixed solvent of methanol (2mL) and dichloromethane (2 mL). At 0 deg.C, an aqueous hydroxylamine solution (50%, 2mL) and a saturated methanol solution of sodium hydroxide (2mL) were added. The reaction mixture was stirred at room temperature overnight. The organic solvent was removed by concentration under reduced pressure, diluted hydrochloric acid (1N) was added to the mixture to adjust pH to 7, and the mixture was concentrated under reduced pressure to obtain a crude product. The crude product was combined with a small aliquot (MC19-12-015-A1) and prepared via liquid phase (0.1% formic acid) to afford the title compound (18mg, 15.9% in three steps, as a white solid). LC-MS (ESI) M/z [ M + H ]]+453.4。1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),8.64(s,1H),7.58-7.48(m,4H),7.44-7.39(m,1H),7.13-7.10(m,2H),7.01-6.99(m,1H),6.63(t,J=5.4Hz,1H),4.58(s,2H),3.71-3.66(m,2H),3.05(q,J=6.4Hz,2H),2.61-2.57(m,2H),1.93(t,J=7.6Hz,2H),1.49-1.40(m,4H),1.25-1.24(m,4H)。
Example 20:n-hydroxy-7- (9- (2-chlorophenyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamido) heptanoyl Amines as pesticides
Figure BDA0002858379420000371
Step 1 preparation of ethyl 2,3,4, 9-tetrahydro-1H-carbazole-3-carboxylate
Phenylhydrazine hydrochloride (2.00g,13.9mmol) and ethyl 4-oxocyclohexane-1-carboxylate (2.36g,13.9mmol) were dissolved in acetic acid (30 mL). The reaction mixture was stirred at 65 ℃ overnight. After the organic solvent was removed by concentration under reduced pressure, an aqueous sodium hydrogencarbonate solution (30ml) and ethyl acetate (20ml) were added thereto to conduct liquid separation extraction. The aqueous phase was extracted with ethyl acetate (20)ml × 2) extraction. The combined organic phases were washed with saturated brine (10ml), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give a crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate ═ 10:1) gave the title compound (2.00g, yield 59.3%, pale yellow solid). LC-MS (ESI) M/z [ M + H ]]+244.1。
Step 2: preparation of ethyl 9- (2-chlorophenyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxylate
(200mg,0.823mmol) and 1-chloro-2-iodobenzene (235mg,0.988mmol) were dissolved in toluene (6mL), and cuprous iodide (15.6mg,0.0823mmol), potassium phosphate (349mg,1.65mmol) and dimethylethylenediamine (14.5mg,0.165mmol) were added. The reaction mixture was stirred at 130 ℃ overnight under argon. After the reaction mixture was cooled to room temperature, water (10mL) was added, and the mixture was extracted with ethyl acetate (10 mL. times.2). The combined organic phases were washed with brine (10mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the crude product. Purification by column chromatography (silica gel, petroleum ether: ethyl acetate 10:1) gave the title compound (200mg, yield 68.8%, white solid). LC-MS (ESI) M/z [ M + H ]]+no signal。
And step 3: preparation of 9- (2-chlorophenyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxylic acid
(180mg,0.510mmol) was dissolved in a mixed solvent of tetrahydrofuran (5mL) and methanol (5mL), and a saturated solution of sodium hydroxide (10mL) was added. The reaction mixture was stirred at 30 ℃ overnight. The reaction solution was concentrated to remove the organic solvent, and a dilute hydrochloric acid solution (2N) was added to adjust the pH to 7. Extraction was performed with ethyl acetate (20 mL. times.2). The combined organic phases were washed with brine (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was rotary-dried to give the title compound (150mg, yield 90.4%, yellow solid). LC-MS (ESI) M/z [ M + H ]]+326.3。
And 4, step 4: 7- (9- (2-chlorophenyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamido) heptanoic acid methyl ester
9- (2-chlorophenyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxylic acid (150mg,0.460mmol) and methyl 7-aminoheptanoate hydrochloride (90.1mg,0.460mmol) were added to tetrahydrofuran (10mL), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (175mg,0.460mmol) and triethyleneurea were addedAmine (140mg,1.38 mmol). The reaction mixture was stirred at room temperature overnight. After quenching the reaction with water (10mL), the organic solvent was removed by concentration under reduced pressure. Extraction was performed with ethyl acetate (10 mL. times.2), and the organic phases were combined and concentrated under reduced pressure to remove the organic solvent to give the crude product. Slurried with methanol (2mL), filtered, and the filter cake dried under reduced pressure to give the title compound (150mg, yield 69.7%, white solid). LC-MS (ESI) M/z [ M + H ]]+467.2。
And 5: preparation of N-hydroxy-7- (9- (2-chlorophenyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamido) heptanamide
Methyl 7- (9- (2-chlorophenyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamido) heptanoate (150mg,0.321mmol) was dissolved in a mixed solvent of methanol and dichloromethane (10mL, 1:1), and an aqueous hydroxylamine solution (50%, 2mL) was added at room temperature. A saturated methanol solution of sodium hydroxide (2mL) was added dropwise while cooling on ice. The reaction mixture was stirred at room temperature for 3 hours. The organic solvent was removed by concentration under reduced pressure, and the title compound (13.4mg, yield 8.93%, white solid) was prepared by reverse phase (0.1% formic acid). LC-MS (ESI) M/z [ M + H ]]+468.3。1H NMR(400MHz,DMSO-d6):δ10.34(br s,1H),8.73-8.61(m,1H),7.91-7.88(m,1H),7.76-7.37(m,1H),7.58-7.47(m,4H),7.06-7.04(m,2H),6.79-6.76(m,1H),3.13-3.03(m,2H),2.91-2.72(m,2H),2.62-2.58(m,1H),2.46-2.41(m,1H),2.35-2.29(m,1H),2.07-1.92(m,3H),1.87-1.78(m,1H),1.50-1.42(m,4H),1.27-1.26(m,4H)。
Example 21:n- (7- (hydroxyamino) -7-oxoheptyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamide
Figure BDA0002858379420000381
Step 1: preparation of 2,3,4, 9-tetrahydro-1H-carbazole-3-carboxylic acid
Aqueous sodium hydroxide (5N,2mL) was added to a solution of methyl 2,3,4, 9-tetrahydro-1H-carbazole-3-carboxylate (400mg,1.74mmol) in methanol (2mL) at room temperature. The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure to remove the organic solvent, diluted with water (2mL), and adjusted to pH 4 with dilute hydrochloric acid (5N). Will precipitateThe solid was filtered off and rinsed with water (5 mL. times.2). The solid was dried under reduced pressure to give the objective compound (375mg, yield 100%, light brown solid). LC-MS (ESI) M/z [ M + H ]]+216.1。
Step 2: preparation of methyl 7- (2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamido) heptanoate
N, N, N ', N' -tetramethyl-O- (7-azabenzotriazol-1-yl) urea hexafluorophosphate (459mg,1.21mmol), N, N-diisopropylethylamine (600mg,4.65mmol) and methyl 7-aminoheptanoate hydrochloride (182mg,0.929mmol) were added to a solution of 2,3,4, 9-tetrahydro-1H-carbazole-3-carboxylic acid (200mg,0.929mmol) in N, N-dimethylformamide (2mL) at room temperature. The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (30mL) and extracted with ethyl acetate (30 mL. times.3). The combined organic phases were washed with saturated brine (50 mL. times.2), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to give the objective compound (325mg, crude product, yellow oil). LC-MS (ESI) M/z [ M + H ]]+357.2。
And step 3: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamide
Saturated sodium hydroxide methanol solution (1mL) and aqueous hydroxylamine solution (50%, 1mL) were added to a solution of methyl 7- (2,3,4, 9-tetrahydro-1H-carbazole-3-carboxamido) heptanoate (325mg,0.912mmol) in dichloromethane (1mL) in this order at room temperature. The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure to remove the organic solvent, diluted with water (2mL), cooled in an ice bath, and adjusted to pH 4 with dilute hydrochloric acid (1N). The precipitated solid was filtered off, and the title compound (59.6mg, yield in two steps, 17.9%, white solid) was obtained by liquid phase preparation (0.1% HCOOH). LC-MS (ESI) M/z [ M + H ]]+358.2。1H NMR(400MHz,DMSO-d6)δ10.65(s,1H),10.33(s,1H),8.65(s,1H),7.85(t,J=5.6Hz,1H),7.33(d,J=7.6Hz,1H),7.22(d,J=8.0Hz,1H),6.99-6.95(m,1H),6.92-6.89(m,1H),3.10-3.02(m,2H),2.80-2.63(m,4H),2.55-2.52(m,1H),2.03-1.99(m,1H),1.94(t,J=7.6Hz,2H),1.85-1.78(m,1H),1.52-1.38(m,4H),1.26-1.25(m,4H)。
Example 22:n- (7- (hydroxyamino) -7-oxoheptyl) -4-phenyl-1, 2,3, 4-tetrahydrocyclopenta [ b ] amine]Indole Indole-2-carboxamides
Figure BDA0002858379420000391
Step 1: preparation of methyl 1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxylate
Phenylhydrazine hydrochloride (925mg,6.39mmol) and methyl 3-oxocyclopentane-1-carboxylate (1.00g,7.03mmol) were added to acetic acid (10mL) and the reaction mixture was stirred at 65 ℃ for 16 h. The organic solvent was removed by concentration under reduced pressure. Water (10mL) was added to dilute the solution, and the solution was extracted with ethyl acetate (10 mL. times.3). The combined organic phases were washed with saturated brine solution (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product. The mixture of the target compounds (600mg, crude, red solid) was obtained by column chromatography separation and purification (silica gel, petroleum ether: ethyl acetate: 20: 1-5: 1). LC-MS (ESI) M/z [ M + H ]]+216.0。
Step 2: preparation of methyl 4-phenyl-1, 2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxylate
1,2,3, 4-tetrahydrocyclopenta [ b ] is reacted with]Indole-2-carboxylic acid methyl ester (mixture) (600mg,2.79mmol) was dissolved in toluene (15mL), iodobenzene (683mg,3.35mmol), potassium phosphate (1.18g,5mmol), cuprous iodide (53mg,0.279mmol) and N, N' -dimethylethylenediamine (50mg,0.558mmol) were added. The reaction mixture was stirred at 120 ℃ for 16 hours under argon. The reaction solution was cooled to room temperature. The reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product. The target compound (690mg, 37.1% yield in two steps, colorless oily substance) was obtained by column chromatography separation and purification (silica gel, petroleum ether: ethyl acetate: 20:1 to 2: 1). LC-MS (ESI) M/z [ M + H ]]+292.2。1H NMR(400MHz,CDCl3)δ7.49-7.40(m,6H),7.33-7.30(m,1H),7.13-7.10(m,2H),3.82-7.78(m,2H),3.73(s,3H),3.34-3.10(m,2H)。
Step 3 preparation of 4-phenyl-1, 2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxylic acid
4-phenyl-1, 2,3,4-tetrahydrocyclopenta [ b ] s]Indole-2-carboxylic acid methyl ester (690mg,2.37mmol) was dissolved in ethanol (20mL), and aqueous sodium hydroxide (2N, 10mL) was added. The reaction mixture was stirred at 60 ℃ for 1 hour. The reaction solution was cooled to room temperature, and the pH was adjusted to 6 with dilute hydrochloric acid (1N). Extraction was carried out with ethyl acetate (20 mL. times.3). The combined organic phases were washed with brine (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (470mg, crude, off-white solid). LC-MS (ESI) M/z [ M + H ]]+278.1。
And 4, step 4: 7- (4-phenyl-1, 2,3, 4-tetrahydrocyclopenta [ b ]]Preparation of methyl indole-2-carboxamido) heptanoate 4-phenyl-1, 2,3, 4-tetrahydrocyclopenta [ b ] amine]Indole-2-carboxylic acid (270mg,0.975mmol) and methyl 7-aminoheptanoate hydrochloride (210mg,1.07mmol) were dissolved in N, N-dimethylformamide (5mL), and 2- (7-oxabenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (555mg,1.46mmol) and N, N-diisopropylethylamine (629mg,4.87mmol) were added. The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (10 mL. times.3). The combined organic phases were washed with brine (10 mL. times.2), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the crude product. Ethyl acetate (2mL) was added and stirred for 10 min, filtered, and the filter cake was washed with ethyl acetate (1 mL). The solid was dried under reduced pressure to give the title compound (250mg, two-step yield 43.9%, white solid). LC-MS (ESI) M/z [ M + H ]]+419.4。
And 5: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -4-phenyl-1, 2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxamide
Reacting 7- (4-phenyl-1, 2,3, 4-tetrahydrocyclopenta [ b ]]Methyl indole-2-carboxamido) heptanoate (MC18-663-067-A1) (100mg,0.239mmol) was dissolved in a mixed solvent of methanol (3mL) and dichloromethane (1.5 mL). Aqueous sodium hydroxide (2N,1mL) and aqueous hydroxylamine (50%, 0.5mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 16 hours. The organic solvent was removed by concentration under reduced pressure. The mixture was adjusted to pH 7 with dilute hydrochloric acid (1N), filtered and the filter cake washed with water (2 mL). The solid was dried under reduced pressure to give the objective compound (82.06mg, yield 81.9%, white solid). LC-MS (ESI) M/z [ M + H ]]+420.3。1H NMR(400MHz,DMSO-d6)δ10.34(s,1H),8.65(s,1H),7.96(br s,1H),7.59-7.50(m,4H),7.44-7.37(m,3H),7.09-7.07(m,2H),3.68-3.64(m,1H),3.12-3.10(m,5H),2.93-2.88(m,1H),1.93(t,J=7.2Hz,2H),1.49-1.41(m,4H),1.26-1.25(m,4H)。
Example 23:n- (7- (hydroxyamino) -7-oxoheptyl) -4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta-ane [b]Indole-2-carboxamides
Figure BDA0002858379420000411
Step 1: preparation of methyl 4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxylate
1,2,3, 4-tetrahydrocyclopenta [ b ] is reacted with]Indole-2-carboxylic acid methyl ester (200mg,0.929mmol) and 2-bromopyridine (147mg,0.929mmol) were dissolved in1, 4-dioxane (10mL), and cesium carbonate (606mg,1.86mmol), tris (dibenzylideneacetone) dipalladium (85mg,0.0929mmol) and 4, 5-bis diphenylphosphine-9, 9-dimethylxanthene (108mg,0.186mmol) were added. The reaction mixture was stirred at 100 ℃ for 16 hours under argon. The reaction mixture was cooled to room temperature, and water (20mL) was added to the reaction mixture, followed by extraction with ethyl acetate (20 mL. times.3). The combined organic phases were washed with brine (20mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the crude product. Purification by preparative plate separation (silica gel, petroleum ether: ethyl acetate ═ 1:1) gave the title compound (60mg, yield 22.1%, light yellow oil). LC-MS (ESI) M/z [ M + H ]]+293.2。
Step 2: preparation of 4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxylic acid
Reacting 4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta [ b ]]Indole-2-carboxylic acid methyl ester (60mg,0.205mmol) was dissolved in methanol (2mL), and aqueous sodium hydroxide (2N, 2mL) was added. The reaction mixture was stirred at 60 ℃ for 1 hour. The reaction mixture was cooled to room temperature, concentrated under reduced pressure to remove the organic solvent, and then adjusted to pH 6 with dilute hydrochloric acid (1N). The mixture was filtered, the filter cake was washed with water (2mL), and the solid was dried under reduced pressure to give the title compound (40mg, yield 70.2%, white solid). LC-MS (ESI) M/z [ M + H ]]+279.1。
And step 3: preparation of methyl 7- (4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxamido) heptanoate
Reacting 4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta [ b ]]Indole-2-carboxylic acid (40mg,0.144mmol) was dissolved in N, N-dimethylformamide (1mL), and methyl 7-aminoheptanoate hydrochloride (31mg,0.158mmol), 2- (7-oxabenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (60mg,0.158mmol) and N, N-diisopropylethylamine (56mg,0.432mmol) were added. The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with brine (10mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the crude product. Purification by preparative plate separation (silica gel, petroleum ether: ethyl acetate ═ 1:1) gave the title compound (40mg, yield 66.3%, colorless oil). LC-MS (ESI) M/z [ M + H ]]+420.3。
And 4, step 4: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxamide
Reacting 7- (4- (pyridin-2-yl) -1,2,3, 4-tetrahydrocyclopenta [ b ]]Methyl indole-2-carboxamido) heptanoate (40mg,0.0953mmol) was dissolved in a mixed solvent of methanol (1.5mL) and methylene chloride (1.5 mL). Aqueous hydroxylamine (50%, 0.5mL) and saturated methanolic sodium hydroxide (0.5mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 3 hours. The organic solvent was removed by concentration under reduced pressure. The mixture was adjusted to pH 7 with dilute hydrochloric acid (1N), filtered and the filter cake washed with water (2 mL). The solid was dried under reduced pressure to give the objective compound (13mg, yield 32.4%, white solid). LC-MS (ESI) M/z [ M + H ]]+421.3。1H NMR(400MHz,DMSO-d6)δ10.33(s,1H),8.66(s,1H),8.58-8.56(m,1H),8.01-7.97(m,3H),7.60(d,J=8.0Hz,1H),7.44-7.42(m,1H),7.33-7.30(m,1H),7.17-7.10(m,2H),3.70-3.62(m,1H),3.29-3.27(m,2H),3.09-3.03(m,3H),2.91-2.86(m,1H),1.93(t,J=7.6Hz,2H),1.50-1.40(m,4H),1.27-1.26(m,4H)。
Example 24:n- (7- (hydroxyamino) -7-oxoheptyl) -1,2,3, 4-tetrahydrocyclopenta [ b)]Indole-2-carboxylic acid Amines as pesticides
Figure BDA0002858379420000421
Step 1: preparation of 1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxylic acid
1,2,3, 4-tetrahydrocyclopenta [ b ] is reacted with]Indole-2-carboxylic acid methyl ester (100mg,0.465mmol) was dissolved in methanol (5mL), and aqueous sodium hydroxide (2N, 4mL) was added. The reaction mixture was stirred at 60 ℃ for 1 hour. The reaction mixture was cooled to room temperature, concentrated under reduced pressure to remove the organic solvent, and then adjusted to pH 6 with dilute hydrochloric acid (1N). The mixture was filtered, the filter cake was washed with water (2mL), and the solid was dried under reduced pressure to give the title compound (70mg, yield 74.8%, yellow solid). LC-MS (ESI) M/z [ M + H ]]+202.7。
Step 2: preparation of methyl 7- (1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxamido) heptanoate
1,2,3, 4-tetrahydrocyclopenta [ b ] is reacted with]Indole-2-carboxylic acid (70mg,0.348mmol) was dissolved in N, N-dimethylformamide (2mL), and methyl 7-aminoheptanoate hydrochloride (75mg,0.383mmol), 2- (7-oxabenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (146mg,0.383mmol) and N, N-diisopropylethylamine (135mg,1.04mmol) were added. The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with brine (10mL), dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure to give the crude product. Purification by preparative plate separation (silica gel, petroleum ether: ethyl acetate ═ 1:1) gave the title compound (50mg, crude, colorless oil). LC-MS (ESI) M/z [ M + H ]]+343.3。
And step 3: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -1,2,3, 4-tetrahydrocyclopenta [ b ] indole-2-carboxamide
Reacting 7- (1,2,3, 4-tetrahydrocyclopenta [ b ]]Indole-2-carboxamido) heptanoic acid methyl ester (50mg,0.146mmol) was dissolved in a mixed solvent of methanol (1.5mL) and dichloromethane (1.5 mL). Aqueous hydroxylamine (50%, 0.5mL) and saturated methanolic sodium hydroxide (0.5mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 3 hours. The organic solvent was removed by concentration under reduced pressure. Mixing ofThe mixture was adjusted to pH 7 with dilute hydrochloric acid (1N), filtered and the filter cake washed with water (2 mL). The filter cake was purified by liquid phase preparation (0.1% HCOOH) to give the title compound (2mg, 1.67% yield in two steps, white solid). LC-MS (ESI) M/z [ M + H ]]+344.3。1H NMR(400MHz,DMSO-d6)δ10.79(s,1H),10.34(s,1H),8.67(s,1H),7.92(t,J=5.6Hz,1H),7.29-7.24(m,2H),6.98-6.89(m,2H),3.62-3.58(m,1H),3.09-3.03(m,2H),3.01-2.93(m,3H),2.80-2.75(m,1H),1.93(t,J=7.6Hz,2H),1.52-1.39(m,4H),1.26-1.25(m,4H)。
Example 25:n- (7- (hydroxyamino) -7-oxoheptyl) -1,3,4, 9-tetrahydro-2H-pyrido [3,4-b]Indole- 2-carboxamides
Figure BDA0002858379420000431
Step 1: preparation of methyl 7- (2,3,4, 9-tetrahydro-1H-pyrido [3,4-b ] indole-2-carboxamido) heptanoate
N, N-diisopropylethylamine (1.12g,8.70mmol) and methyl 7-aminoheptanoate hydrochloride (341mg,1.74mmol) were added to a solution of N, N' -carbonyldiimidazole (339mg,2.09mmol) in N, N-dimethylformamide (2mL) at 0 ℃. The mixture was kept at 0 ℃ and stirred for 0.5 hour. Reacting 2,3,4, 9-tetrahydro-1H-pyrido [3,4-b ] at 0 deg.C]A solution of indole (300mg,1.74mmol) and N, N-diisopropylethylamine (1.12mg,8.70mmol) in N, N-dimethylformamide (1mL) was added to the reaction mixture. The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with water (30mL) and extracted with ethyl acetate (30 mL. times.3). The combined organic phases were washed with saturated brine (80 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Separation and purification on a preparative plate (silica gel, petroleum ether: ethyl acetate ═ 1:1) gave the title compound (387mg, yield 62.2%, brown solid). LC-MS (ESI) M/z [ M + H ]]+358.2。
Step 2: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -1,3,4, 9-tetrahydro-2H-pyrido [3,4-b ] indole-2-carboxamide
Saturated sodium hydroxide in methanol (1mL) was added to 7 with aqueous hydroxylamine (50%, 1mL) in that order at room temperature- (2,3,4, 9-tetrahydro-1H-pyrido [3, 4-b)]Indole-2-carboxamido) heptanoic acid methyl ester (387mg,1.08mmol) in dichloromethane (1 mL). The reaction mixture was stirred at room temperature for overnight reaction. The reaction mixture was concentrated under reduced pressure to remove the solvent, diluted with water (2mL), cooled in an ice bath, and adjusted to pH 4 with dilute hydrochloric acid (1N). The precipitated solid was filtered off, and lyophilized via liquid phase preparation (0.1% HCOOH) to give the objective compound (141.7mg, yield 36.6%, white solid). LC-MS (ESI) M/z [ M + H ]]+359.2。1H NMR(400MHz,DMSO-d6)δ10.82(s,1H),10.33(s,1H),8.67(s,1H),7.37(d,J=7.6Hz,1H),7.28(d,J=8.0Hz,1H),7.03-7.00(m,1H),6.96-6.92(m,1H),6.65(t,J=5.2Hz,1H),4.52(s,2H),3.64(t,J=6.0Hz,2H),3.05-3.00(m,2H),2.65(t,J=5.2Hz,2H),1.92(t,J=7.4Hz,2H),1.48-1.38(m,4H),1.24-1.22(m,4H)。
Example 26:n- (7- (hydroxyamino) -7-oxoheptyl) -4-phenyl-3, 4-dihydropyrrolo [3,4-b]Indole-2 (1H) -carboxamides
Figure BDA0002858379420000441
Step 1: preparation of 3-formyl-1H-indole-2-carboxylic acid methyl ester
Phosphorus oxychloride (4.81g,31.4mmol) was added slowly to N, N-dimethylformamide (8.34g,114mmol) at 0 deg.C, followed by the slow addition of a solution of methyl 1H-indole-2-carboxylate (5.00g,28.6mmol) in N, N-dimethylformamide (30 mL). The reaction mixture was stirred at room temperature for 0.5 hour and then at 60 ℃ for 16 hours. The reaction mixture was cooled to room temperature, added to ice water (50mL), and then adjusted to pH 8 with a saturated aqueous solution of sodium hydrogencarbonate. The mixture was filtered, the filter cake was washed with water (20mL), and the solid was dried under reduced pressure to give the title compound (5.80g, 89.0% yield, yellow solid). LC-MS (ESI) M/z [ M + H ]]+204.0。
Step 2: preparation of methyl 3- (((4-methoxybenzyl) amino) methyl) -1H-indole-2-carboxylate
Methyl 3-formyl-1H-indole-2-carboxylate (5.00g,24.6mmol) was dissolved in a mixed solvent of methanol (50mL) and methylene chloride (50mL), and (4-methoxy-4-carboxylate was addedPhenylphenyl) methylamine (3.37g,24.6 mmol). The reaction mixture was stirred at room temperature for 0.5 h before adding sodium cyanoborohydride (3.86g,61.5 mmol). The reaction mixture was stirred at room temperature for 16 hours. The organic solvent was removed by concentration under reduced pressure, and water (50mL) was added to the mixture to conduct extraction with ethyl acetate (100 mL. times.3). The combined organic phases were washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product. The target compound (5.80g, yield 72.6%, yellow solid) was obtained by column chromatography (silica gel, petroleum ether: ethyl acetate 1:1 to 1: 5). LC-MS (ESI) M/z [ M + H ]]+325.1。
And step 3: preparation of 3- (((4-methoxybenzyl) amino) methyl) -1H-indole-2-carboxylic acid
Methyl 3- (((4-methoxybenzyl) amino) methyl) -1H-indole-2-carboxylate (5.0g,15.5mmol) was dissolved in a mixed solvent of methanol (50mL) and tetrahydrofuran (50mL), and an aqueous solution of sodium hydroxide (2N, 50mL) was added. The reaction mixture was stirred at 60 ℃ for 1 hour. The reaction mixture was cooled to room temperature, concentrated under reduced pressure to remove the organic solvent, and then adjusted to pH 6 with dilute hydrochloric acid (1N). The mixture was filtered, the filter cake was washed with water (20mL), and the solid was dried under reduced pressure to give the title compound (3.5g, yield 72.8%, light yellow solid). LC-MS (ESI) M/z [ M + H ]]+311.1。
And 4, step 4: preparation of 2- (4-methoxybenzyl) -1, 4-dihydropyrrolo [3,4-b ] indol-3 (2H) -one
3- (((4-methoxybenzyl) amino) methyl) -1H-indole-2-carboxylic acid (2.0g,6.45mmol) was added to acetonitrile (120mL) and 2- (7-oxabenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (3.68g,9.67mmol) was added. The reaction mixture was stirred at 80 ℃ for 16 hours. After the reaction solution was cooled to room temperature and brought to 0 ℃, water (120mL) was added to the reaction mixture, and the mixture was stirred at 0 ℃ for 0.5 hour. The mixture was filtered, the filter cake was washed with water (20mL), acetonitrile (20mL), and the solid was dried under reduced pressure to give the title compound (1.20g, yield 63.7%, light yellow solid). LC-MS (ESI) M/z [ M + H ]]+293.1。
And 5: preparation of 2- (4-methoxybenzyl) -4-phenyl-1, 4-dihydropyrrolo [3,4-b ] indol-3 (2H) -one
2- (4-methoxybenzyl) -1, 4-dihydropyrrolo [3,4-b]Indol-3 (2H) -one (900mg,3.08mmol) was dissolved in toluene (20mL), and iodobenzene (754mg,3.70mmol), potassium phosphate (1.30g,6.16mmol), cuprous iodide (58mg,0.308mmol) and N, N' -dimethylethylenediamine (54mg,0.616mmol) were added. The reaction mixture was stirred at 120 ℃ for 16 hours under argon. The reaction solution was cooled to room temperature. Water (50mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with saturated brine (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product. The target compound (920mg, yield 81.1%, yellow solid) was obtained by column chromatography (silica gel, petroleum ether: ethyl acetate: 10:1 to 5: 1). LC-MS (ESI) M/z [ M + H ]]+369.1。
Step 6: preparation of 2- (4-methoxybenzyl) -4-phenyl-1, 2,3, 4-tetrahydropyrrolo [3,4-b ] indole
2- (4-methoxybenzyl) -4-phenyl-1, 4-dihydropyrrolo [3, 4-b)]Indol-3 (2H) -one (300mg,0.814mmol) was dissolved in tetrahydrofuran (20mL) and lithium aluminum hydride (155mg,4.07mmol) was added at 0 ℃. The reaction mixture was stirred at 60 ℃ for 16 hours. The reaction was cooled to 0 ℃ and water (0.15mL), NaOH (15%, 0.15mL), H, was added to the reaction mixture2O (0.45mL) and sodium sulfate (1 g). The mixture was stirred at room temperature for 0.5 h, the mixture was filtered, the filter cake was washed with ethyl acetate (20mL), and the filtrate was concentrated under reduced pressure to give the crude product. Purification by preparative plate separation (silica gel, petroleum ether: ethyl acetate ═ 1:1) afforded the title compound (80mg, 50% purity, crude, yellow solid). LC-MS (ESI) M/z [ M + H ]]+355.1。
And 7: preparation of 4-phenyl-1, 2,3, 4-tetrahydropyrrolo [3,4-b ] indole
2- (4-methoxybenzyl) -4-phenyl-1, 2,3, 4-tetrahydropyrrolo [3,4-b]Indole (80mg, crude, 0.226mmol) was dissolved in1, 2-dichloroethane (4mL) and 1-chloroethyl chloroformate (65mg,0.452mmol) was added. The reaction mixture was stirred at 60 ℃ for 16 hours. The organic solvent was removed by concentration under reduced pressure, methanol (5mL) was added to the mixture, and the reaction mixture was stirred at 65 ℃ for 3 hours. The reaction solution was cooled to room temperature,concentration under reduced pressure gave the title compound (40mg, crude, colorless oil). LC-MS (ESI) M/z [ M + H ]]+235.0。
And 8: preparation of methyl 7- (4-phenyl-1, 2,3, 4-tetrahydropyrrolo [3,4-b ] indole-2-formyl) heptanoate
N, N' -carbonyldiimidazole (21mg,0.128mmol) was dissolved in N, N-dimethylformamide (2 mL). Methyl 7-aminoheptanoate hydrochloride (25mg,0.128mmol) and N, N-diisopropylethylamine (110mg,0.854mmol) were added successively at 0 ℃. The reaction mixture was stirred at room temperature for 0.5 hour, and 4-phenyl-1, 2,3, 4-tetrahydropyrrolo [3,4-b ] was added]Indole (MC18-663-135-A1) (40mg,0.0854mmol) in N, N-dimethylformamide (1 mL). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (10 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a crude product. Purification by preparative plate separation (silica gel, petroleum ether: ethyl acetate ═ 1:1) gave the title compound (20mg, 5.86% in three-step yield, colorless oil). LC-MS (ESI) M/z [ M + H ]]+420.2。
And step 9: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -4-phenyl-3, 4-dihydropyrrolo [3,4-b ] indole-2 (1H) -carboxamide
Reacting 7- (4-phenyl-1, 2,3, 4-tetrahydropyrrolo [3,4-b ]]Indole-2-formyl) heptanoic acid methyl ester (20mg,0.0477mmol) was dissolved in a mixed solvent of methanol (1.5mL) and dichloromethane (1.5 mL). Aqueous hydroxylamine (50%, 0.5mL) and saturated methanolic sodium hydroxide (0.5mL) were added sequentially at 0 ℃. The reaction mixture was stirred at room temperature for 3 hours. The organic solvent was removed by concentration under reduced pressure, the pH was adjusted to 7 with dilute hydrochloric acid (1N), the filtrate was filtered, and the cake was washed with water (1 mL). The filter cake was purified by liquid phase preparation (0.1% HCl) to give the title compound (5mg, 25.0% yield, white solid). LC-MS (ESI) M/z [ M + H ]]+421.2。1H NMR(400MHz,DMSO-d6)δ10.33(s,1H),8.65(s,1H),7.62-7.59(m,4H),7.53-7.42(m,3H),7.20-7.13(m,2H),6.35(t,J=5.6Hz,1H),4.63-4.60(m,4H),3.09-3.04(m,2H),1.93(t,J=7.2Hz,2H),1.50-1.42(m,4H),1.26-1.25(m,4H)。
Example 27:N-(7-(hydroxylamino) -7-oxoheptyl) -5-phenyl-8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b]Indole-2-carboxamides
Figure BDA0002858379420000461
Step 1: preparation of 8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
4-Chlorophenylhydrazine hydrochloride (1.0g,5.58mmol) and tert-butyl 4-oxopiperidine-1-carboxylate (1.33g,6.70mmol) were added to ethanol (20mL) and concentrated hydrochloric acid (5mL) was added. The reaction mixture was stirred at 90 ℃ for 16 hours. The reaction was cooled to room temperature, the reaction was filtered, the filter cake was washed with ethanol (5mL), and the solid was dried under vacuum to give the title compound (430mg, yield 22.5%, off-white solid). LC-MS (ESI) M/z [ M + H ]]+207.1。
Step 2: preparation of tert-butyl 8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride (103mg,0.425mmol) was added to dichloromethane (3mL), triethylamine (152mg,1.50mmol) and 4-dimethylaminopyridine (6mg,0.050mmol) were added at 0 deg.C, followed by di-tert-butyl dicarbonate (120mg,0.550 mmol). The reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was diluted with water (10mL), extracted with dichloromethane (10 mL. times.3), the combined organic phases were washed with dilute hydrochloric acid (1N,10 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (150mg, crude, pale yellow oil). LC-MS (ESI) M/z [ M-H ] -305.1.
And step 3: preparation of tert-butyl 5-phenyl-8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxylate
Reacting 8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Tert-butyl indole-2-carboxylate (150mg, crude 0.490mmol) was dissolved in toluene (5mL), and iodobenzene (120mg,0.588mmol), potassium phosphate (208mg,0.980mmol), cuprous iodide (9.33mg,0.0490mmol), and N, N' -dimethylethylenediamine (8.64mg,0.0980mmol) were added. The reaction mixture was stirred at 120 ℃ for 16 hours under argon. Adding water to the reaction mixtureDiluted (10mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic phases were washed with saturated brine (10mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product. Separation and purification on a preparative plate (silica gel, petroleum ether: ethyl acetate ═ 10:1) gave the target compound (100mg, two-step yield 61.5%, white solid). LC-MS (ESI) M/z [ M + H ]]+383.2。
And 4, step 4: preparation of 5-phenyl-8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride
Reacting 5-phenyl-8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ]]Indole-2-carboxylic acid tert-butyl ester (100mg,0.261mmol) was dissolved in methanol (2mL), and 1, 4-dioxane hydrochloride solution (4M,1mL) was added. The reaction mixture was stirred at 40 ℃ for 1 hour. Concentration under reduced pressure gave the crude product (83mg, crude, light red solid). LC-MS (ESI) M/z [ M + H ]]+283.0。
And 5: preparation of methyl 7- (5-phenyl-8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
N, N' -carbonyldiimidazole (50.7mg,0.313mmol) was dissolved in N, N-dimethylformamide (1mL), and N, N-diisopropylethylamine (169mg,1.30mmol) and methyl 7-aminoheptanoate hydrochloride (61.2mg,0.313mmol) were added at 0 ℃. The reaction mixture is stirred at room temperature for half an hour, and then 5-phenyl-8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] is added]Indole hydrochloride (83mg,0.261mmol, crude) and N, N-diisopropylethylamine (169mg,1.30mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was stirred at room temperature for 16 hours. Water (10mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (10 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (100mg, crude, white solid). LC-MS (ESI) M/z [ M + H ]]+468.3。
Step 6: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -5-phenyl-8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (5-phenyl-8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Indole-2-carboxamido) heptanoic acid methyl ester (100mg,0.214mmol) dissolved in methanol (3mL) and dichloromethane (1.5mL)In a mixed solvent. Aqueous sodium hydroxide (2N,1mL) was added at 0 deg.C, followed by aqueous hydroxylamine (50%, 0.5 mL). The reaction mixture was stirred at room temperature for 16 hours. The organic solvent was removed by concentration under reduced pressure and the mixture was adjusted to pH 6 with dilute hydrochloric acid (1N), the mixture was filtered, and the solid was lyophilized from preparative isolation purification (0.1% HCl) to give the title compound (17mg, 13.9% yield in three steps, light yellow solid). LC-MS (ESI) M/z [ M + H ]]+469.3。1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),8.64(br s,1H),7.61-7.57(m,2H),7.53(d,J=2.0Hz,1H),7.50-7.45(m,3H),7.21-7.18(m,1H),7.12-7.09(m,1H),6.57(t,J=5.2Hz,1H),4.57(s,2H),3.67(t,J=5.2Hz,2H),3.05(q,J=6.0Hz,2H),2.65-2.63(m,2H),1.93(t,J=7.6Hz,2H),1.49-1.40(m,4H),1.25-1.24(m,4H)。
Example 28: n- (7- (hydroxyamino) -7-oxoheptyl) -8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Figure BDA0002858379420000481
Step 1: preparation of methyl 7- (8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole-2-carboxamido) heptanoate
Methyl 7-aminoheptanoate hydrochloride (292mg,1.50mmol) was dissolved in N, N-dimethylformamide (5mL), and N, N-diisopropylethylamine (390mg,3.00mmol) and N, N' -carbonyldiimidazole (243mg,1.50mmol) were added. The reaction mixture was stirred at room temperature for 1 hour, and then 8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ] indole hydrochloride (MC19-304-019-A1) (243mg,1.00mmol) and N, N-diisopropylethylamine (390mg,3.00mmol) in N, N-dimethylformamide (1mL) were added. The reaction mixture was stirred at room temperature overnight. Water (50mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The combined organic phases were washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (400mg, crude, yellow oil). LC-MS (ESI) M/z [ M + H ] + 392.2.
Step 2: preparation of N- (7- (hydroxyamino) -7-oxoheptyl) -8-chloro-1, 3,4, 5-tetrahydro-2H-pyrido [4,3-b ] indole-2-carboxamide
Reacting 7- (8-chloro-2, 3,4, 5-tetrahydro-1H-pyrido [4,3-b ]]Methyl indole-2-carboxamido) heptanoate (MC19-304-020-A1) (200mg, crude, 0.500mmol) was dissolved in a mixed solvent of methanol (5mL) and dichloromethane (5 mL). An aqueous hydroxylamine solution (50%, 3mL) and a saturated methanol solution of sodium hydroxide (3mL) were added at 0 ℃. The reaction mixture was stirred at room temperature for 3 hours. The reaction mixture was concentrated under reduced pressure to give a crude product. Purification by liquid phase preparative (0.1% FA) lyophilized to give the title compound (80mg, two step yield 40.8%, white solid). LC-MS (ESI) M/z [ M + H ]]+393.1。1H NMR(400MHz,DMSO-d6)δ11.06(s,1H),10.31(s,1H),8.63(s,1H),7.37(d,J=2.0Hz,1H),7.29(d,J=8.4Hz,1H),7.03-7.00(m,1H),6.52(t,J=5.4Hz,1H),4.46(s,2H),3.67(t,J=5.6Hz,2H),3.05-3.00(m,2H),2.73(t,J=5.4Hz,2H),1.92(t,J=7.2Hz,2H),1.49-1.39(m,4H),1.24-1.22(m,4H)。
Evaluation scheme for Activity of Compounds according to the invention
Experimental example 1: HDAC enzyme activity and subtype selectivity assay
The enzyme inhibitory activity of individual HDAC isoforms of compounds was determined using HDAC Caliper Assay. All HDAC enzymes were purchased from BPS Bioscience, inc, and general reagents not identified were purchased from Sigma, inc.
Preparation of reagents: storing HDAC enzyme stock solution in a refrigerator at-80 ℃; 2. substrate stock (2.5mM) (sequence: 5-FAM-TSRH-Lys (Ac) -M-NH)2) Storing in a refrigerator at-80 deg.C; 3. preparing a sodium chloride solution (1M) at room temperature; 4. preparing a potassium chloride solution (1M) at room temperature; 5. reaction buffer (1000ml) was prepared at room temperature and the composition included: 25ml of Trizma hydrochloride (Sigma company), 137ml of sodium chloride solution and 2.7ml of potassium chloride solution; 1ml of magnesium chloride solution; bovine Serum Albumin (BSA)0.1 g. Substrate working solution (2X), HDAC enzyme working solution (2X); stop solution (1000ml) composition comprising: 100ml of HEPES buffer solution; 20ml of 0.5M EDTA solution; brij 35 solution 1.33 ml; 0.5ml of 5mM Trichostatin A (Sigma). Test compounds were dissolved in DMSO at a concentration of 1mM and diluted at a semilogarithmic concentration to the desired concentration.
The experimental scheme is as follows: add 5. mu.L of substrate working solution into 1.384 well plate; 2.1000rpm for 1 minute; 3. adding 5 mu LHDAC enzyme working solution (containing ZPE or contrast compound) or reaction buffer solution (HPE) into the hole to start reaction; 4.1000rpm for 1 minute; nurturing for 60 minutes at 5.30 ℃; 6. the reaction was stopped by adding a stop solution to the wells. 7.1000rpm for 2 minutes; 8. the 384 well plate was placed in the Caliper EZ Reader II test results.
And (3) data analysis: inhibition calculation, Inhibition% ═ 100- ((Conversion% ZPE-Conversion% test compound)/(Conversion% ZPE-Conversion% HPE)) x 100; IC (integrated circuit)50Values were calculated using GraphPad Prism 5 or XLFit.
Table 1: HDAC6 enzymatic Activity of representative Compounds
Figure BDA0002858379420000491
Figure BDA0002858379420000501
Table 2: HDAC6 enzymatic activity and HDAC subtype selectivity of representative compounds
Figure BDA0002858379420000502
The results show that the compounds of the present patent possess good inhibitory activity against HDAC6 enzyme. Has certain selectivity on HDAC1, HDAC3 and HDAC 8.
Experimental example 2: cell viability assay
The experimental scheme is as follows: a549, Calu-6 and A375 cells were purchased from CAS cell banks, and the remaining cells were purchased from ATCC cell banks. Compounds were prepared in stock solutions with a concentration of 20mM in DMSO. (1) Day 1: plating: cells were trypsinized, resuspended in media, and counted in an automatic cell counter. The cell suspension was diluted to the desired density according to seeding density. 100ul of cell suspension was plated per well in 96-well plates and incubated overnight at 37 ℃. (2) And (3) preparing a compound by using Day 2: compounds were diluted 200-fold in DMSO at final concentration and diluted in 3-fold gradients. By usingThe prepared compound was diluted in the medium to prepare a compound having a final concentration of 3 times, i.e., 3ul of the prepared compound was added to 197ul of the medium to make the total volume 200 ul. 50ul of compound was added to each well, and wells containing the same volume of DMSO were used as controls and incubated at 37 ℃ for 72 hours. (3) And (6) detecting by using Day 4: the cell plate was equilibrated to room temperature. Add 40ul Cell per well
Figure BDA0002858379420000512
The reagent was shaken for 2 minutes, left to stand for 60 minutes and then detected by EnVision.
And (3) data analysis: (1) data were processed using GraphPad Prism 5 software; (2) % Inh ═ Max signal-Compound signal)/(Max signal-Min signal) x 100; (3) max signal is DMSO treatment result; (4) min signal is the medium result.
Table 3: inhibitory Activity of representative Compounds on MM.1S (multiple myeloma) cells
Compound numbering MM.1S(IC50,μM)
Example 2 1.9
Example 10 3.5
Example 1 7.0
Example 27 1.9
Example 20 4.5
Example 22 3.5
Example 3 3.5
Example 11 2.3
Example 12 3.6
Example 13 2.3
Example 14 3.0
Table 4: representative of the inhibitory Activity of the Compound of example 1 on 11 tumor cells
Figure BDA0002858379420000511
Figure BDA0002858379420000521
The results show that the compounds of the invention have certain inhibitory activity on various tumor cell lines including MM.1S cells.
Experimental example 3: in vivo antitumor efficacy testing in animals
The experimental scheme is as follows: culturing and amplifying MM.1S cells in vitro, collecting cells in logarithmic growth phase, suspending in serum-free RPMI1640 culture solution, adding Martrigel at a ratio of 1:1, and adjusting the cell concentration to 6.67X 107/mL; injecting the cell suspension into the axilla of the front right limb of BALB/c nude mice subcutaneously by a 1mL syringe, and injecting 0.15mL into each animal; the growth conditions of animals and transplanted tumors are regularly observed, when the average tumor volume grows to about 100-200mm3, animals with overlarge, undersize or irregular tumor shapes are eliminated, and the animals are divided into groups by a random block method, wherein each group comprises 3-6 animals.
After grouping, the following table 5 dosing was started with a test period of 3 weeks; during the administration period, measuring the tumor diameter 2 times every week, weighing the animal body weight, observing the living state of the animal, and recording abnormal conditions; the test was ended 3 weeks after administration, CO2The animals were euthanized, tumor tissue was stripped, photographed and weighed, and tumor weight inhibition rate was calculated.
The administration scheme is as follows:
blank control group (vehicle): physiological saline.
The positive drug Bortezomib (Bortezomib) (0.5mg/kg) administration preparation is prepared as follows: weighing a proper amount of Bortezomib sample powder, placing the Bortezomib sample powder into a 5mL centrifuge tube, adding a proper amount of normal saline, vortex, shaking and uniformly mixing to prepare a solution with the Bortezomib sample concentration of 0.05mg/mL, and preparing the solution in situ. The preparation and application process need to be protected from light.
Preparation of a test compound administration preparation: weighing a proper amount of sample powder, placing the sample powder into two 5mL centrifuge tubes, sequentially adding a proper amount of DMSO and PEG 400, and performing vortex oscillation to completely dissolve and uniformly mix the sample; adding a proper amount of physiological saline respectively, whirling, shaking and mixing uniformly to prepare solutions with sample concentrations of 3.0mg/mL and 6.0mg/mL respectively, wherein the solvent accounts for 4% DMSO, 30% PEG 400 and 66% physiological saline, and the preparation is carried out on site.
Table 5: dosing regimens
Figure BDA0002858379420000522
Figure BDA0002858379420000531
Note: ip, intraperitoneal injection administration; iv, tail vein injection administration; the administration volume was 10 mL/kg.
Observing the state of the animals at each administration and recording; if the animal died, the animal was roughly dissected, visually inspected for visceral abnormalities, and recorded.
During the experiment, tumor size was measured 2 times per week while animal body weight was weighed.
Data processing: tumor Volume (TV): the formula is that TV is 1/2 multiplied by a multiplied by b2Wherein, a represents the tumor major axis; b represents the tumor minor axis.
The tumor volume curve for 21 days of dosing is shown in figure 1.
Animal weight loss rate: the formula is 100% × (BWinitial-BWfinal)/bwinifinal. Wherein BWinitial represents the body weight of the animal at the time of the group administration; BWfinal represents the animal body weight at the end of the experiment.
The body weight change rate profile of the animals given for 21 days is shown in FIG. 2.
Statistical analysis method
Experimental data were calculated and statistically processed using Microsoft Office Excel 2003 software. Data are expressed as Mean ± standard error (Mean ± s.e) unless otherwise stated, and comparison between groups was performed using t-test. Figure 1 indicates that the results are significantly different from the control group, P < 0.05; indicates that the results are significantly different from the control group, P < 0.01.
And (4) conclusion: the tumor volume curve (fig. 1) after 21 days of administration shows that the compound in example 1 has very good inhibitory activity on transplanted tumors in MM (multiple myeloma) animals, and has significant difference with a control group, and the single drug is equivalent to the positive drug Bortezomib, and the combined drug is better than the positive drug Bortezomib. Meanwhile, the body weight of the animals (figure 2) did not significantly decrease by more than 5% in 21 days, indicating that the compound has good safety. The conclusion shows that the compound of the embodiment has the antitumor effect, has good safety and has the potential to be applied to the treatment of tumor diseases.

Claims (10)

1. A compound shown as a formula I or a pharmaceutically acceptable salt thereof is characterized in that the structure is shown as follows,
Figure FDA0002858379410000011
wherein the content of the first and second substances,
z is N or CH;
m1 is 1 or 2;
m2 is 0 or 1;
R1is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl radical, C1-C3Alkoxy radical, C3-C8Cycloalkyl, 3-8 membered heterocycloalkyl with one or more heteroatoms selected from N, O and S and 1-4 heteroatoms, unsubstituted or R1-2Substituted C6-C14Aryl, or unsubstituted or R1-3A 5-12 membered heteroaryl group having one or more of N, O and S as a substituent "hetero atom, and 1-4 as hetero atoms";
R1-1is halogen, C3-C8Cycloalkyl, halogen substituted C3-C8Cycloalkyl, or 3-8 membered heterocycloalkyl with "heteroatom number 1-4" selected from one or more of N, O and S;
R1-2and R1-3Independently cyano or halogen;
R2is hydrogen, cyano, halogen, C1-C3Alkoxy radical, C1-C6Alkyl, halogen substituted C1-C6Alkyl radical, C3-C6Cycloalkyl, halogen substituted C3-C6Cycloalkyl, 3-6 membered heterocycloalkyl with 1-4 heteroatoms selected from N, O and S, or 3-6 membered heterocycloalkyl with 1-4 heteroatoms selected from N, O and S;
R3is hydrogen, halogen, C1-C3Alkoxy radical, C1-C6Alkyl radical, C3-C6Cycloalkyl, or 3-6 membered heterocycloalkyl.
2. As claimed in claim1 the compound shown as the formula I or pharmaceutically acceptable salt thereof, characterized in that R is1Is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl, unsubstituted or R1-2Substituted C6-C14Aryl, or unsubstituted 5-12 membered heteroaryl with 1-4 heteroatoms selected from one or more of N, O and S;
and/or, R1-1Is halogen, C3-C8Cycloalkyl, halogen substituted C3-C8Cycloalkyl, or 3-to 8-membered heterocycloalkyl having "one or more heteroatoms selected from N, O and S, and 1 to 4 heteroatoms", preferably R1-13-8 membered heterocycloalkyl which is halogen or "heteroatom number is 1-4" selected from one or more of N, O and S;
and/or, R1-2Is halogen;
and/or, R2Is hydrogen or halogen;
and/or, R3Is hydrogen.
3. The compound of formula I, or a pharmaceutically acceptable salt thereof, according to claim 1, wherein the compound of formula I is according to any one of the following schemes:
scheme 1:
z is N or CH;
m1 is 1 or 2;
m2 is 0 or 1;
R1is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl, unsubstituted or R1-2Substituted C6-C14Aryl, or unsubstituted 5-12 membered heteroaryl with 1-4 heteroatoms selected from one or more of N, O and S;
R1-1is halogen, C3-C8Cycloalkyl, halogen substituted C3-C8Cycloalkyl, or 3-8 membered heterocycloalkyl with "heteroatom number 1-4" selected from one or more of N, O and S;
R1-2is halogen;
R2is hydrogen or halogen;
R3is hydrogen;
scheme 2:
z is N or CH;
m1 is 1 or 2;
m2 is 0 or 1;
R1is hydrogen, unsubstituted or R1-1Substituted C1-C6Alkyl, unsubstituted or R1-2Substituted C6-C14Aryl, or unsubstituted 5-12 membered heteroaryl with 1-4 heteroatoms selected from one or more of N, O and S;
R1-1is C3-C8Cycloalkyl, or 3-8 membered heterocycloalkyl with "heteroatom number 1-4" selected from one or more of N, O and S;
R1-2is halogen;
R2is hydrogen or halogen;
R3is hydrogen.
4. A compound of formula I according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R is R1Is unsubstituted or R1-1Substituted C1-C6When it is alkyl, said R1-1When the number of R is 1 or more1-1When there are plural, R is1-1May be the same or different; preferably said R1-1The number of (a) is 1,2 or 3;
and/or when R1Is unsubstituted or R1-1Substituted C1-C6When alkyl, said C1-C6Alkyl is C1-C4An alkyl group, preferably a methyl group, an ethyl group, an isopropyl group or an isobutyl group, further preferably a methyl group, an isopropyl group or an isobutyl group;
and/or when R1Is unsubstituted or R1-2Substituted C6-C14When aryl is said to R1-2When the number of R is 1 or more1-2When there are plural, R is1-2May be the same or different; preferably said R1-2The number of (a) is 1,2,3 or 4;
and/or when R1Is unsubstituted or R1-2Substituted C6-C14When aryl, said C6-C14Aryl is C6-C12Aryl of (a), preferably phenyl;
and/or when R1When the 5-to 12-membered heteroaryl group is an unsubstituted 5-to 12-membered heteroaryl group having one or more heteroatoms selected from N, O and S and 1 to 4 heteroatoms, the 5-to 12-membered heteroaryl group is a 5-to 6-membered heteroaryl group having one or more heteroatoms selected from N, O and S and 1 to 3 heteroatoms, preferably a pyridyl group, a pyrimidinyl group or a pyrazinyl group, more preferably a pyridyl group, a pyrimidinyl group or a pyrazinyl group
Figure FDA0002858379410000031
Figure FDA0002858379410000032
Further preferred is
Figure FDA0002858379410000033
And/or when R1-1Is C3-C8When there is a cycloalkyl group, said C3-C8Cycloalkyl being C3-C6Cycloalkyl, preferably cyclopropyl or cyclopentyl, more preferably
Figure FDA0002858379410000034
And/or when R1-1When the heterocyclic group is a 3-to 8-membered heterocycloalkyl group having "one or more hetero atoms selected from N, O and S and a hetero atom number of 1 to 4", the 3-to 8-membered heterocycloalkyl group is a 5-to 6-membered heterocycloalkyl group having "one or more hetero atoms selected from N, O and S and a hetero atom number of 1 to 3", preferably a tetrahydrofuranyl group or a tetrahydropyranyl group, more preferably a tetrahydrofuranyl group
Figure FDA0002858379410000035
Figure FDA0002858379410000036
And/or when R1-2When halogen, said halogen is fluorine, chlorine or bromine, preferably fluorine or chlorine;
and/or when R2In the case of halogen, the halogen is fluorine, chlorine or bromine, preferably chlorine.
5. A compound of formula I or a pharmaceutically acceptable salt thereof as claimed in any one of claims 1 to 3 wherein R is1Is H, -CH3、-CH2CH3
Figure FDA0002858379410000037
Figure FDA0002858379410000038
Preferably H, -CH3
Figure FDA0002858379410000039
Figure FDA0002858379410000041
And/or, R2Is H or Cl;
and/or, structure
Figure FDA0002858379410000042
Is composed of
Figure FDA0002858379410000043
Figure FDA0002858379410000044
Wherein the a-terminal is linked to the C atom, the b-terminal is linked to the N atom, and the C-terminal is linked to
Figure FDA0002858379410000045
Are connected.
6. The compound of formula I, or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-3, wherein the compound of formula I is of any one of the following structures,
Figure FDA0002858379410000046
Figure FDA0002858379410000051
7. a pharmaceutical composition, which comprises a compound of formula I as described in any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier; the compound shown in the formula I or the pharmaceutically acceptable salt thereof is preferably in a therapeutically effective amount.
8. Use of a compound of formula I according to any one of claims 1 to 6 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 7 for the manufacture of a medicament for inhibiting HDAC or a disease associated with HDAC;
said HDAC inhibitor is preferably an HDAC6 inhibitor;
the HDAC related diseases are preferably tumors and/or autoimmune diseases;
the tumor is preferably solid tumor and blood tumor such as lung cancer, colon cancer, rectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, brain cancer, renal cancer, ovarian cancer, gastric cancer, skin cancer, bone cancer, glioma, glioblastoma, hepatocellular carcinoma, papillary renal cancer, head and neck cancer, leukemia, lymphoma, myeloma, multiple myeloma and the like;
the autoimmune disease is preferably rheumatoid arthritis, osteoarthritis, rheumatoid spondylitis, psoriasis, post-ischemic perfusion injury, inflammatory bowel disease, chronic inflammatory lung disease, eczema, asthma, psoriasis, ulcerative colitis, acute respiratory distress syndrome, psoriatic arthritis, infectious arthritis, progressive chronic arthritis, osteoarthritis deformans, femoral arthritis, traumatic arthritis, gouty arthritis, Reiter's syndrome, polychondritis, acute synovitis and spondylitis, glomerulonephritis, hemolytic anemia, aplastic anemia, idiopathic thrombocytopenia, neutropenia, ulcerative colitis, Crohn's disease, graft-versus-host disease, allograft rejection, chronic thyroiditis, Graves ' disease, scleroderma, active hepatitis, primary biliary cirrhosis, chronic thyroiditis, chronic inflammatory bowel disease, myasthenia gravis, multiple sclerosis, systemic lupus erythematosus, allergic dermatitis, contact dermatitis, chronic renal insufficiency, Step-Johnson syndrome, idiopathic steatorrhea, sarcoidosis, Guillain-Barre syndrome, pulmonary fibrosis, or chronic inflammatory lung disease.
9. Use of a compound of formula I as defined in any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined in claim 7, in the manufacture of a medicament;
the medicament is preferably a medicament for preventing and/or treating tumors and/or autoimmune diseases;
the tumor is preferably solid tumor and blood tumor such as lung cancer, colon cancer, rectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, brain cancer, renal cancer, ovarian cancer, gastric cancer, skin cancer, bone cancer, glioma, glioblastoma, hepatocellular carcinoma, papillary renal cancer, head and neck cancer, leukemia, lymphoma, myeloma, multiple myeloma and the like;
the autoimmune disease is preferably rheumatoid arthritis, osteoarthritis, rheumatoid spondylitis, psoriasis, post-ischemic perfusion injury, inflammatory bowel disease, chronic inflammatory lung disease, eczema, asthma, psoriasis, ulcerative colitis, acute respiratory distress syndrome, psoriatic arthritis, infectious arthritis, progressive chronic arthritis, osteoarthritis deformans, femoral arthritis, traumatic arthritis, gouty arthritis, Reiter's syndrome, polychondritis, acute synovitis and spondylitis, glomerulonephritis, hemolytic anemia, aplastic anemia, idiopathic thrombocytopenia, neutropenia, ulcerative colitis, Crohn's disease, graft-versus-host disease, allograft rejection, chronic thyroiditis, Graves ' disease, scleroderma, active hepatitis, primary biliary cirrhosis, chronic thyroiditis, chronic inflammatory bowel disease, myasthenia gravis, multiple sclerosis, systemic lupus erythematosus, allergic dermatitis, contact dermatitis, chronic renal insufficiency, Step-Johnson syndrome, idiopathic steatorrhea, sarcoidosis, Guillain-Barre syndrome, pulmonary fibrosis, or chronic inflammatory lung disease.
10. Use of a compound of formula I, or a pharmaceutically acceptable salt thereof, as defined in any one of claims 1 to 6, or a pharmaceutical composition as defined in claim 7, in the manufacture of a medicament for the prevention and/or treatment of cancer, in combination with a chemotherapeutic or immunotherapeutic agent;
the tumor is preferably solid tumor and blood tumor such as lung cancer, colon cancer, rectal cancer, breast cancer, prostate cancer, liver cancer, pancreatic cancer, brain cancer, renal cancer, ovarian cancer, gastric cancer, skin cancer, bone cancer, glioma, glioblastoma, hepatocellular carcinoma, papillary renal cancer, head and neck cancer, leukemia, lymphoma, myeloma, multiple myeloma, etc.
CN202011552396.2A 2019-12-31 2020-12-24 Tricyclic compound and use thereof Pending CN113121527A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201911420691 2019-12-31
CN201911420691X 2019-12-31

Publications (1)

Publication Number Publication Date
CN113121527A true CN113121527A (en) 2021-07-16

Family

ID=76772215

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011552396.2A Pending CN113121527A (en) 2019-12-31 2020-12-24 Tricyclic compound and use thereof

Country Status (1)

Country Link
CN (1) CN113121527A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114591323A (en) * 2022-03-17 2022-06-07 济南大学 Hydroxamic acid histone deacetylase inhibitor containing tetrahydrocarboline structure and preparation method and application thereof
CN117865913A (en) * 2024-01-10 2024-04-12 山东金特安全科技有限公司 Preparation method of dinotefuran

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004113336A1 (en) * 2003-06-16 2004-12-29 Chroma Therapeutics Limited Carboline and betacarboline derivatives for use as hdac enzyme inhibitors
US7250514B1 (en) * 2002-10-21 2007-07-31 Takeda San Diego, Inc. Histone deacetylase inhibitors
CN101103000A (en) * 2004-11-23 2008-01-09 Ptc医疗公司 Carbazole, carboline, and indole derivatives useful in the inhibition of VEGF production
CN101133060A (en) * 2005-05-05 2008-02-27 色品疗法有限公司 Enzyme inhibitors
US20100261710A1 (en) * 2007-08-21 2010-10-14 Arqule, Inc. HDAC Inhibitors
CN102408425A (en) * 2004-03-15 2012-04-11 Ptc医疗公司 Carboline derivatives useful in inhibition of angiogenesis and application thereof
JP2013129632A (en) * 2011-12-22 2013-07-04 Ono Pharmaceut Co Ltd Enpp2 inhibiting compound
CN108084178A (en) * 2018-01-04 2018-05-29 济南大学 Hydroxamic acid compound of the one kind containing tetrahydrochysene-β-Ka Lin structures, purposes and preparation method thereof

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7250514B1 (en) * 2002-10-21 2007-07-31 Takeda San Diego, Inc. Histone deacetylase inhibitors
WO2004113336A1 (en) * 2003-06-16 2004-12-29 Chroma Therapeutics Limited Carboline and betacarboline derivatives for use as hdac enzyme inhibitors
CN102408425A (en) * 2004-03-15 2012-04-11 Ptc医疗公司 Carboline derivatives useful in inhibition of angiogenesis and application thereof
CN101103000A (en) * 2004-11-23 2008-01-09 Ptc医疗公司 Carbazole, carboline, and indole derivatives useful in the inhibition of VEGF production
CN101133060A (en) * 2005-05-05 2008-02-27 色品疗法有限公司 Enzyme inhibitors
US20100261710A1 (en) * 2007-08-21 2010-10-14 Arqule, Inc. HDAC Inhibitors
JP2013129632A (en) * 2011-12-22 2013-07-04 Ono Pharmaceut Co Ltd Enpp2 inhibiting compound
CN108084178A (en) * 2018-01-04 2018-05-29 济南大学 Hydroxamic acid compound of the one kind containing tetrahydrochysene-β-Ka Lin structures, purposes and preparation method thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114591323A (en) * 2022-03-17 2022-06-07 济南大学 Hydroxamic acid histone deacetylase inhibitor containing tetrahydrocarboline structure and preparation method and application thereof
CN117865913A (en) * 2024-01-10 2024-04-12 山东金特安全科技有限公司 Preparation method of dinotefuran

Similar Documents

Publication Publication Date Title
US10730863B2 (en) Bridged bicyclic compounds as farnesoid X receptor modulators
CN109071546B (en) Pyrazolo [1,5-A ] pyrazin-4-yl derivatives as JAK inhibitors
CN111440189B (en) Fused ring pyrimidine amino derivative, preparation method, intermediate, pharmaceutical composition and application thereof
CN114096542A (en) Condensed tricyclic pyrroles as alpha-1 antitrypsin modulators
US10016410B2 (en) Tetrahydroisoquinolin-1-one derivative or salt thereof
AU2011311814B2 (en) Substituted pyridazine carboxamide compounds
WO2005094805A1 (en) Imine derivative and amide derivative
JP2022500473A (en) Oxadicycloic acid as an LPA antagonist
TW201302730A (en) Pyrazole compounds
CN107207486A (en) For suppressing method of protein and combination outside Bu Luomo domains and end
DK2599774T3 (en) DEHYDRATED pyridine AS CB2 cannabinoid receptor ligands
CN101918395A (en) Triazole oxadiazoles derivatives
TW201623239A (en) Novel 2-amino-pyridine and 2-amino-pyrimidine derivatives and medical use thereof
CN115315423A (en) Substituted aryl compounds
CN113121527A (en) Tricyclic compound and use thereof
CN111714628A (en) Small molecule PD-1/PD-L1 inhibitor, pharmaceutical composition of small molecule PD-1/PD-L1 inhibitor and PD-L1 antibody and application of small molecule PD-1/PD-L1 inhibitor and pharmaceutical composition
TW202003472A (en) Calpain modulators and therapeutic uses thereof
CN113784950A (en) Novel thyromimetic agents
WO2023202623A1 (en) Polq inhibitor compound and use thereof
CN106458980A (en) Amino pyridine derivatives as phosphatidylinositol 3-kinase inhibitors
CN108586378B (en) Indoleamine 2, 3-bis-oxidase inhibitor and preparation method and application thereof
CN112028815A (en) Indole derivatives and medical application thereof
CN113767102A (en) Novel phenyl and pyridylureas having activity against Hepatitis B Virus (HBV)
CN110177784A (en) One kind containing aromatic compound, preparation method, pharmaceutical composition and application
WO2017034377A1 (en) Pyridopyrimidinone compounds for modulating the catalytic activity of histone lysine demethylases (kdms)

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