CN112279863A - Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof - Google Patents

Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof Download PDF

Info

Publication number
CN112279863A
CN112279863A CN201910675023.5A CN201910675023A CN112279863A CN 112279863 A CN112279863 A CN 112279863A CN 201910675023 A CN201910675023 A CN 201910675023A CN 112279863 A CN112279863 A CN 112279863A
Authority
CN
China
Prior art keywords
compound
condensing agent
conjugate
organic solvent
reaction
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
CN201910675023.5A
Other languages
Chinese (zh)
Inventor
吕伟
朱书雷
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.)
East China Normal University
Original Assignee
East China Normal University
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 East China Normal University filed Critical East China Normal University
Priority to CN201910675023.5A priority Critical patent/CN112279863A/en
Publication of CN112279863A publication Critical patent/CN112279863A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/22Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

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

Abstract

The invention discloses a conjugate of an Hsp90 inhibitor and a camptothecin derivative, and a preparation method and application thereof, wherein the conjugate is connected through a connecting chain; the preparation method comprises the following steps: hsp90 inhibitor and camptothecin derivative are connected through a cleavable connecting chain to prepare a series of conjugates, so that the targeting of corresponding drug molecules to tumor tissues and the enrichment of the camptothecin derivative at tumor parts are realized, and the camptothecin derivative has good antitumor activity and can be applied to the preparation of antitumor drugs.

Description

Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof
Technical Field
The invention relates to the field of antitumor drug chemistry, and particularly relates to a conjugate of an Hsp90 inhibitor and a camptothecin derivative, and a preparation method and application thereof.
Background
In recent years, medical technology is rapidly developed, but cancer still is one of the major diseases seriously threatening the life and social development of human beings, and the incidence rate still tends to rise year by year, thus seriously harming the health of human beings. At present, the main treatment means of cancer is surgical resection, traditional radiotherapy and chemotherapy, targeted therapy, novel cell therapy and the like. Chemotherapy remains one of the main approaches to clinical tumor treatment, with cytotoxic drugs still being the first line of chemotherapy. Cytotoxic drugs act by inhibiting rapidly differentiating tumor cells. However, at present, the antitumor drugs have high toxicity, poor biocompatibility and poor selectivity, and are easy to generate drug resistance, so that the clinical application of the antitumor drugs is limited. Research shows that the combined chemotherapy can overcome the drug resistance of tumors to a certain extent, but different drugs have different action modes and metabolic modes in vivo due to different properties, have a plurality of uncontrollable factors, and are limited in clinical application. The selective drug conjugate can specifically identify tumors and realize the enrichment of drugs at tumor sites, and becomes a hotspot of tumor drug research in recent years. The heat shock protein 90(Hsp90) inhibitor drug conjugate can be combined with extracellular Hsp90(eHsp90) to realize the transportation and accumulation of the drug in tumors, and clinically shows good effects.
Hsp90 is an important covalent chaperone protein in vivo, accounts for about 1-3% of the total number of cellular proteins in vivo, and is one of the important targets for tumor therapy. Hsp90 is one of the important members of heat shock protein family, and the substrate protein is mostly the protein controlling the cell differentiation and growth, including the signal transduction molecules of tumor cell metastasis, such as epidermal growth factor receptor 2, Bcr/Abl fusion gene, protein kinase B, c-Raf, cyclin dependent kinase 4, hypoxia inducible factor I, steroid hormone receptor, etc. The function of these conformationally labile substrate proteins is maintained by Hsp 90. AT present, clinical Hsp90 inhibitors mainly comprise geldanamycin, NVP-AUY922, SNX5422, STA9090 and AT13387, NVP-BEP800, KW2478, XL888, PU-H71 and the like, and the inhibitors all show certain tumor treatment effects clinically, wherein the inhibitors are structurally represented by the NVP-AUY922 and the SNX 5422.
NVP-AUY922 is a potent and newly synthesized small molecule inhibitor of resorcinol isoxazole amide Hsp90, can inhibit Hsp90 alpha/beta, has an IC50 of 13nM/21nM, has a slightly weaker effect on the Hsp90 family members GRP94 and TRAP-1, and can be tightly bound with a small molecule Hsp90 ligand. NVP-AUY922 inhibited proliferation of a number of human tumor cell lines in an in vitro study, with GI50 averaging 9 nM. NVP-AUY922 has an IC50 value of 2 to 40nM on gastric cancer cells. NVP-AUY922 acted on BEAS-2B cells with an IC50 of 28.49 nM. The expression of Hsp90 was not affected by NVP-AUY922, but increased the expression of HSP 70. NVP-AUY922 enhances the binding ability of HSP70 to HSP 90. NVP-AUY922 causes separation of p23 from the HSP complex. NVP-AUY922 enables HSP70 to bind to HSP90 complex. Using NVP-AUY922, reduced expression of tyrosine kinase receptors such as VEGFR1, 2, 3 and PDGFR-alpha, as well as Akt and p-Akt, was induced. In addition, NVP-AUY922 acts on NCI-N87 cells, causing a decrease in HER2 expression. NVP-AUY922 increases the binding force of HSP90 to a target protein, and then protein degradation is performed by protease. NVP-AUY922 can alter multiple signaling pathways to affect cell growth. In addition, the use of the protease inhibitor MG132 restored the decreased thymidine synthase overexpression due to the use of NVP-AUY 922. NVP-AUY922 increases the expression of cleared caspase-3 and causes apoptosis of HSC-2 cells. Meanwhile, in an in vivo experiment, the NVP-AUY922 can be used for causing a strong anti-cancer response and inhibiting the expression of p-Akt and VEGF in a HSC-2 transplantation tumor model. Of the 12 experimental tumors, 5 tumor regressions were observed. Body weight loss was < 5% and the biomarkers associated with HSP90 were significantly altered. In BT474, ERBB2 was completely absent, era was depleted, and CDK4 and phosphorylated ERK1/2 were reduced.
SNX-2112 is a mesylate salt, is an orally active HSP90 inhibitor with a KD of 41nm and induces HER-2 degradation, and an IC50 of 37 μ nm. PF 049113 had a clear effect on HER2 and P-ERK stability in A375 cells, and P-S6 protein stability in A375 cells was 5 + -1, 11 + -3 and 61 + -22 nm, respectively. PF 04929113 induced HSP70 in A375 cells with an IC50 of 13. + -.3 nM. PF 04929113 (0.5, 1, 2, 5 and 10 μm) reduced cell viability in a concentration-dependent manner. In addition, PF 04929113 (1, 3, 5, 7 μm) synergized with equivalent amounts of HDAC inhibitors (PXD101, SAHA and TSA) to induce cell death by inhibiting PI3K/Akt/mTOR signaling in ATC cells. In vivo experiments, SNX5422 can effectively inhibit tumor growth of HT-29 human colon cancer xenograft model, and obviously inhibit tumor growth and angiogenesis of mouse Multiple Myeloma (MM).
AT13387(Onalespib) is a potent, selective Hsp90 inhibitor with an IC50 of 18nM in a375 cells with long-lasting antitumor activity. AT13387 inhibits a variety of kinases, including CDK 1, CDK 2, CDK4, FGFR3, PKB-B, JAK2, VEGFR2, PDGFR-B, and Aurora B. However, AT13387 concentrations below 30 μ M had no significant inhibitory effect on the experimental kinases. AT13387 is effective in inhibiting proliferation and survival of a variety of tumor cell lines, such as MES-SA cell lines. AT13387 acts on a group of 30 tumor cell lines, effectively inhibiting cell proliferation, with GI50 values of 13-260 nM. AT13387 inhibited the proliferation of the non-tumorigenic prostate epithelial cell line PNT2 with a GI50 value of 480 nM. In vivo studies, AT13387 was administered twice weekly AT a dose of 70mg/kg, or once weekly AT a dose of 90 mg/kg. The weight of the mice is reduced by no more than 20 percent. Prolonged non-treatment period, and anticancer activity, associated with the long-term efficacy of AT13387 on mutant EGFR and other biomarkers, and the memory of AT13387 on tumors.
Although Hsp90 inhibitor has a certain clinical effect, the toxic side effects, such as hepatotoxicity and optic nerve toxicity, greatly limit the clinical application of the medicine. Therefore, a new drug delivery method is urgently needed to improve the clinical administration property of the Hsp90 inhibitor.
7-Ethyl-10-hydroxycamptothecin (SN38) is a camptothecin derivative, a classical topoisomerase I inhibitor. Clinically, SN38 is effective in inducing apoptosis of rapidly differentiating tumor cells. However, SN38 shows very low solubility under both physiological and pharmacological conditions, which makes it impossible to administer it directly, directly limiting its wide clinical application. In recent years, in order to improve the water solubility and biocompatibility of SN38, a number of SN38 drug delivery systems have been developed, such as: pegylated conjugates, liposomes, antibody-conjugated drugs, and the like. These drug delivery systems often exhibit poor tumor permeability and low drug carrier dose, limiting their clinical efficacy. It is particularly urgent to develop a new strategy that can improve the administration properties of SN38 while achieving accumulation of SN38 at the tumor site.
Disclosure of Invention
The invention aims to overcome the defects of the existing clinical drugs and provides a conjugate of an Hsp90 inhibitor and a camptothecin derivative as well as a preparation method and application thereof, wherein the Hsp90 inhibitor and the camptothecin derivative are coupled through a connecting chain, and the amino acid ester bonds of 10-position hydroxyl and 20-position hydroxyl of 7-ethyl-10-hydroxycamptothecin (SN38) are mainly used for connection, so that the selection of the connecting chain is wider, and the modification is carried out at the drug position. The conjugate can improve the administration of 7-ethyl-10-hydroxycamptothecin (SN38) in vivo, realize the enrichment and slow release of SN38 in tumor, improve the bioavailability of SN38 in vivo, and prolong the time of drug action.
The specific technical scheme for realizing the purpose of the invention is as follows:
a conjugate of an Hsp90 inhibitor and a camptothecin derivative, the conjugate being linked by a linking chain and having the formula Ia or Ib, I c; IIa or IIb, IIc; IIIa or IIIb, IIIc; IVa or IVb, IV c:
Figure BDA0002142994190000031
Figure BDA0002142994190000041
wherein in formula Ia or Ib, IIa or IIb, IIIa or IIIb, IVa or IVb, n is 2 or 1; the Hsp90 inhibitor is NVP-AUY922, SNX-5422, AT13387, NVP-BEP800, KW2478, XL888 or PU-H71; the camptothecin derivative is camptothecin, 7-ethyl-10-hydroxycamptothecin or topotecan; the connecting chain is an amino acid ester bond, a urethane bond, a carbonate bond, an isocyanate bond or a thioisocyanate bond.
The preparation method of the conjugate comprises the following steps:
Figure BDA0002142994190000051
the reaction steps comprise:
a) condensation reaction: under the action of a condensing agent, reacting the compound VIII with a compound V, VI and a compound VII respectively in an organic solvent to generate compounds Ia, Ib and Ic; the molar ratio of the compound VIII to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound VIII to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
In the step a), the condensing agent is one or a mixture of more of O- (7-azobenzotriazol-1-oxide) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
The preparation method of the conjugate comprises the following steps:
Figure BDA0002142994190000061
the reaction steps comprise:
a) hydrogenation reaction: carrying out hydrogenation reaction on the compound IX in an organic solvent under the conditions of a palladium catalyst and hydrogen to generate a compound X; the mass ratio of the compound IX to the palladium catalyst is 1: 0.1, and the reaction time is 4 hours;
b) condensation reaction: under the action of a condensing agent, reacting a compound X with a compound V, VI and a compound VII in an organic solvent to generate compounds IIa, IIb and IIc; the molar ratio of the compound X to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound X to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
In the step a), the organic solvent is one or a mixture of methanol, ethanol and isopropanol; the palladium catalyst is one or a mixture of more of 10% palladium carbon, palladium and palladium hydroxide; in the step b), the condensing agent is one or a mixture of more of O- (7-azobenzotriazol-1-oxide) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
The preparation method of the conjugate comprises the following steps:
Figure BDA0002142994190000071
the reaction steps comprise:
a) condensation reaction: under the action of a condensing agent, reacting a compound XI with a compound V, VI and a compound VII in an organic solvent to generate compounds IIIa, IIIa and IIIc; the molar ratio of the compound XI to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound XI to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
In the step a), the condensing agent is one or a mixture of more of O- (7-azobenzotriazol-1-oxide) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
The preparation method of the conjugate comprises the following steps:
Figure BDA0002142994190000072
the reaction steps comprise:
a) condensation reaction: under the action of a condensing agent, reacting a compound XII with a compound V, VI and a compound VII in an organic solvent respectively to generate compounds VIa, VIb and VIc; the molar ratio of the compound XII to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound XII to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
In the step a), the condensing agent is one or a mixture of more of O- (7-azobenzotriazol-1-oxide) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
An application of the conjugate in preparing antitumor drugs.
The conjugate of the invention can realize the enrichment of targeting and active drugs in vivo and play the role of anti-tumor. The conjugate is a compound which is formed by modifying two or more medicines in a certain structure and is connected in a certain connection mode to integrally exert the drug effect, and the purpose of providing the medicine is to improve the targeting property of the medicine, realize the enrichment of active medicines in vivo, reduce toxic and side effects and the like.
Drawings
FIG. 1 is a graph showing the results of cell cycle experiments with compounds IIa, IIb and IIc;
FIG. 2 is a Western blot experiment of compounds IIa, IIb and IIc.
Detailed Description
The present invention will be described in further detail with reference to the following specific examples and the accompanying drawings, which are not intended to limit the invention.
Example 1
Synthesis of Compounds Ia-c
Figure BDA0002142994190000081
Preparation of Compound Ia
100mg (0.236mmol) of compound VIII are introduced into a 25mL round-bottomed flask, dissolved in anhydrous DCM (5mL), 135mg (0.354mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethylurea hexafluorophosphate and 130. mu.L (0.708mmol) of N, N-diisopropylethylamine are added thereto, and after stirring for 0.5h in an ice bath, 120mg (0.260mmol) of compound V are added and the reaction is continued for 12h at room temperature. Dilute with dichloromethane (5mL), WaterWashed (10mL x3), washed with saturated sodium chloride solution (10mL), and dried over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure. The crude product was subjected to silica gel column chromatography (DCM: MeOH ═ 30: 1 → 15: 1) to give compound I a as a pale yellow solid, 85mg, 42% yield. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C44H42N7O9F3,[m+Na]+:892.2894,found:892.2894.
1H NMR(400MHz,DMSO-d6)δ8.94(s,1H),8.02(d,J=7.5Hz,1H),7.94(t,J=6.9Hz,1H),7.80(d,J=7.6Hz,1H),7.70(s,2H),7.36(d,J=1.5Hz,1H),7.33-7.32(m,1H),7.32-7.29(m,1H),7.14(t,J=1.0Hz,1H),7.13(d,J=1.6Hz,1H),7.11(dd,J=7.5,1.6Hz,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),3.92-3.74(m,2H),3.56-3.39(m,2H),3.38(s,2H),3.09(dd,J=12.5,8.0Hz,2H),2.79-2.69(m,2H),2.67(d,J=7.0Hz,2H),2.13-1.93(m,2H),1.21(t,J=8.0Hz,3H),1.15(s,3H),1.09(s,3H),1.06(t,J=8.0Hz,3H)。
Preparation of Compound I b
100mg (0.236mmol) of compound VIII are placed in a 25mL round-bottom flask, dissolved in anhydrous DCM (5mL), 135mg (0.354mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethylurea hexafluorophosphate and 130. mu.L (0.708mmol) of N, N-diisopropylethylamine are added thereto, and after stirring for 0.5h in an ice bath, 116mg (0.260mmol) of compound VI are added, and the reaction is continued for 12h at room temperature. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure, and the crude product was subjected to silica gel column chromatography (DCM: MeOH ═ 30: 1 → 15: 1) to give I b as a pale yellow solid, 80mg, yield 41%. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C43H40N7O9F3,[m+Na]+:878.2737,found:878.2749.
1H NMR(400MHz,DMSO-d6)δ8.94(s,1H),8.02(d,J=7.5Hz,1H),7.80(d,J=7.6Hz,1H),7.70(s,2H),7.58-7.46(m,1H),7.39(t,J=7.7Hz,1H),7.36(d,J=1.5Hz,1H),7.32(dd,J=7.5,1.5Hz,1H),7.14(t,J=1.0Hz,1H),7.13(d,J=1.6Hz,1H),7.11(dd,J=7.5,1.6Hz,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),3.98(s,1H),3.96(s,1H),3.92-3.78(m,2H),3.38(s,2H),3.09(ddq,J=77.3,12.5,8.0Hz,2H),2.73(q,J=17.9Hz,2H),2.12-1.93(m,2H),1.21(t,J=8.0Hz,3H),1.15(s,3H),1.09(s,3H),1.06(t,J=8.0Hz,3H)。
Synthesis of Compound I c
100mg (0.236mmol) of compound VIII are placed in a 25mL round-bottom flask, dissolved in dry dichloromethane (5mL), 135mg (0.354mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethylurea hexafluorophosphate and 130. mu.L (0.708mmol) of N, N-diisopropylethylamine are added thereto, and after stirring for 0.5h in an ice bath, 183mg (0.260mmol) of compound VII are added, and the reaction is continued for 12h at room temperature. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure, and the crude product was subjected to silica gel column chromatography (DCM: MeOH ═ 30: 1 → 15: 1) to give I c as a pale yellow solid (98 mg, yield 45%). m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C47H48N8O9F3,[m+H]+:925.3496,found:925.3468.
1H NMR(400MHz,DMSO-d6)δ8.02(d,J=7.5Hz,1H),7.96(d,J=7.6Hz,1H),7.70(s,2H),7.60(t,J=8.5Hz,1H),7.43(d,J=1.5Hz,1H),7.32(td,J=7.3,1.5Hz,2H),7.21(t,J=1.0Hz,1H),7.15(d,J=10.1Hz,1H),7.13(d,J=1.6Hz,1H),5.27(dd,J=2.1,1.0Hz,2H),4.92(s,2H),4.77(s,1H),3.88(dd,J=12.4,8.5Hz,1H),3.82-3.78(m,2H),3.77(d,J=7.0Hz,1H),3.72-3.69(m,1H),3.70-3.67(m,1H),3.68-3.63(m,1H),3.38(s,2H),3.08(dd,J=12.4,8.1Hz,2H),2.73(q,J=17.9Hz,2H),1.97(dq,J=12.3,8.0Hz,1H),1.83(qd,J=7.1,2.9Hz,4H),1.79-1.74(m,1H),1.22(t,J=8.0Hz,3H),1.12(s,6H),0.98(t,J=8.0Hz,3H)。
Example 2
Synthesis of Compounds II a-c
Figure BDA0002142994190000101
Synthesis of Compound X
2.0g (3.236mmol) of Compound IX was charged in a 100mL two-necked flask, dissolved in methanol (30mL), purged with nitrogen, to which 10% -palladium on carbon (400mg) was added, purged with nitrogen and further purged with hydrogen, and the reaction was stirred at room temperature for 4 hours. The celite pad was filtered off with suction, the filtrate was evaporated to dryness under reduced pressure, and the crude product was chromatographed on silica gel (DCM: MeOH ═ 30: 1 → 10: 1) to give 1.3g of compound X as a pale yellow solid with a yield of 90%. m.p.180-182 deg.C
Synthesis of Compound II a
100mg (0.228mmol) of compound X are placed in a 25mL round-bottom flask, dissolved in anhydrous DCM (5mL), 130mg (0.342mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethylurea hexafluorophosphate, 126. mu.L (0.684mmol) of N, N-diisopropylethylamine are added thereto, and after stirring for 0.5h in an ice bath, 112mg (0.250mmol) of compound V are added, and the reaction is continued for 12h at room temperature. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure. The crude product was chromatographed on a silica gel column (DCM: MeOH ═ 30: 1 → 15: 1) to give compound IIa as 89mg as a pale yellow solid in 45% yield. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C49H49N5O11,[m+Na]+:906.9551,found:906.9558.
1HNMR(400MHz,DMSO-d6)δ8.94(s,1H),8.87(s,1H),8.31(t,J=8.1Hz,1H),7.80(d,J=7.6Hz,1H),7.58(dt,J=7.6,1.0Hz,2H),7.41(d,J=0.6Hz,1H),7.38(d,J=1.4Hz,1H),7.37(d,J=1.2Hz,1H),7.36(d,J=1.8Hz,1H),7.14(t,J=1.0Hz,1H),7.11(dd,J=7.5,1.6Hz,1H),6.52(s,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),3.65-3.60(m,5H),3.57-3.40(m,3H),3.27-3.09(m,2H),3.01(dq,J=12.5,8.1Hz,1H),2.71(t,J=7.1Hz,2H),2.48(q,J=7.2Hz,4H),2.17-1.87(m,2H),1.24(d,J=6.8Hz,3H),1.22(d,J=8.1Hz,3H),1.19(d,J=6.8Hz,3H),1.06(t,J=8.0Hz,3H)。
Synthesis of Compound II b
100mg (0.228mmol) of compound X are placed in a 25mL round-bottom flask, dissolved in anhydrous DCM (5mL), 130mg (0.342mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethyluronium hexafluorophosphate, 126. mu.L (0.684mmol) of N, N-diisolactothylamine are added thereto, and after stirring for 0.5h in an ice bath, 106mg (0.250mmol) of compound VI are added, and the reaction is continued at room temperature for 12 h. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure. The crude product was chromatographed on a silica gel column (DCM: MeOH ═ 30: 1 → 15: 1) to give 89mg of compound II b as a pale yellow solid in 44% yield. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C48H47N5O11,[m+H]+:869.9280,found:869.9288.
1H NMR(500MHz,DMSO-d6)δ8.94(s,1H),8.87(s,1H),8.36(t,J=9.1Hz,1H),7.80(d,J=7.6Hz,1H),7.58(dt,J=7.6,1.1Hz,2H),7.41(d,J=0.6Hz,1H),7.40(d,J=1.3Hz,1H),7.39(d,J=1.3Hz,1H),7.36(d,J=1.4Hz,1H),7.14(t,J=1.0Hz,1H),7.11(dd,J=7.5,1.6Hz,1H),6.52(s,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),4.23-4.06(m,2H),3.65-3.59(m,5H),3.52(dt,J=12.5,1.0Hz,1H),3.23-3.18(m,1H),3.17-3.12(m,1H),3.01(dq,J=12.5,8.1Hz,1H),2.48(q,J=7.2Hz,4H),2.15-1.83(m,2H),1.25-1.18(m,9H),1.06(t,J=8.0Hz,3H)。
Synthesis of Compound IIc
100mg (0.228mmol) of Compound X was placed in a 25mL round-bottom flask, dissolved in anhydrous DCM (5mL), to which 130mg (0.342mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethyluronium hexafluorophosphate, 126. mu.L (0.684mmol) of N, N-diisopropylethylamine were added, stirred for 0.5h under ice bath and 128mg (0.250mmol) was addedThe compound VII is stirred at room temperature for 12 h. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure. The crude product was chromatographed on a silica gel column (DCM: MeOH ═ 30: 1 → 15: 1) to give compound II c as a pale yellow solid, 104mg, 49% yield. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C52H54N6O12,[m+H]+:939.0350,found:939.0351.
1H NMR(400MHz,DMSO-d6)δ8.87(s,1H),7.96(d,J=7.6Hz,1H),7.80(d,J=11.0Hz,1H),7.58(dt,J=7.6,1.0Hz,2H),7.43(d,J=1.4Hz,1H),7.41(d,J=0.6Hz,1H),7.38(d,J=1.4Hz,1H),7.37(d,J=1.2Hz,1H),7.31(dd,J=7.5,1.5Hz,1H),7.21(t,J=1.0Hz,1H),6.52(s,1H),5.40-5.17(m,2H),4.92(s,2H),4.77(s,1H),3.86(dp,J=10.8,7.0Hz,1H),3.73(ddt,J=43.2,12.4,7.1Hz,4H),3.63(s,1H),3.61(d,J=7.2Hz,4H),3.52(dt,J=12.5,1.0Hz,1H),3.24-3.09(m,2H),3.00(dq,J=12.5,8.1Hz,1H),2.48(t,J=7.1Hz,4H),2.01-1.94(m,1H),1.90(q,J=7.0Hz,4H),1.77(dq,J=12.3,7.9Hz,1H),1.24(s,1H),1.22(s,4H),1.21(d,J=1.5Hz,4H),0.98(t,J=8.0Hz,3H)。
Example 3
Synthesis of Compounds III a-c
Figure BDA0002142994190000121
Preparation of Compound III a
100mg (0.191mmol) of compound XI are placed in a 25mL round-bottom flask, dissolved in anhydrous DCM (5mL), to which 109mg (0.287mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethyluronium hexafluorophosphate, 106. mu.L (0.573mmol) of N, N-diisopropylethylamine are added, stirred for 0.5h in an ice bath, 105mg (0.229mmol) of compound V are added, and the reaction is continued at room temperature for 12 h. Dilute with dichloromethane (5mL), wash with water (10 mL. times.3), wash with saturated sodium chloride solution (10mL)Washed and dried over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure. The crude product was chromatographed on a silica gel column (DCM: MeOH ═ 30: 1 → 15: 1) to give compound IIIa as a pale yellow solid 83mg, 45% yield. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C53H57N7O11,[m+H]+:968.4194,found:968.4192.
1H NMR(400MHz,DMSO-d6)δ8.94(s,1H),8.87(s,1H),7.96(t,J=7.7Hz,1H),7.83-7.77(m,2H),7.58(dt,J=7.6,1.0Hz,2H),7.41(d,J=0.6Hz,1H),7.39-7.34(m,3H),7.16-7.08(m,2H),6.52(s,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),3.68(dt,J=12.5,1.1Hz,1H),3.59(dt,J=12.5,1.1Hz,1H),3.53-3.31(m,4H),3.25-3.11(m,4H),3.01(dq,J=12.5,8.1Hz,1H),2.68(t,J=7.1Hz,2H),2.63-2.56(m,3H),2.56-2.50(m,5H),2.12-2.01(m,1H),1.97(dt,J=12.5,8.0Hz,1H),1.26-1.17(m,12H),1.06(t,J=8.0Hz,3H)。
Preparation of Compound III b
100mg (0.191mmol) of compound XI are placed in a 25mL round-bottom flask, dissolved in anhydrous DCM (5mL), to which 109mg (0.287mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethyluronium hexafluorophosphate, 106. mu.L (0.573mmol) of N, N-diisopropylethylamine are added, stirred for 0.5h in an ice bath, 102mg (0.229mmol) of compound VI are added, and the reaction is stirred at room temperature for a further 12 h. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure, and the crude product was subjected to silica gel column chromatography (DCM: MeOH ═ 30: 1 → 15: 1) to give compound III b as a pale yellow solid (74 mg, yield 41%). m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C52H55N7O11,[m+H]+:954.4038,found:954.4065.
1H NMR(400MHz,DMSO-d6)δ8.94(s,1H),8.87(s,1H),7.96(t,J=7.7Hz,1H),7.90(t,J=7.9Hz,1H),7.80(d,J=7.6Hz,1H),7.58(dt,J=7.5,1.0Hz,2H),7.41(d,J=0.6Hz,1H),7.39-7.34(m,3H),7.16-7.08(m,2H),6.52(s,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),3.97(d,J=7.8Hz,2H),3.68(dt,J=12.5,1.1Hz,1H),3.59(dt,J=12.5,1.1Hz,1H),3.53-3.41(m,1H),3.41-3.31(m,1H),3.27-3.20(m,2H),3.20-3.11(m,2H),3.01(dq,J=12.5,8.1Hz,1H),2.61-2.51(m,8H),2.12-2.01(m,1H),1.97(dt,J=12.5,8.0Hz,1H),1.25(s,3H),1.23-1.17(m,9H),1.06(t,J=8.0Hz,3H)。
Synthesis of Compound IIIc
100mg (0.236mmol) of compound XI was added to a 25mL round-bottom flask, dissolved in anhydrous dichloromethane (5mL), to which 109mg (0.287mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethylurea hexafluorophosphate and 106. mu.L (0.573mmol) of N, N-diisopropylethylamine were added, stirred for 0.5h in an ice bath, 161mg (0.229mmol) of compound VII was added, and the reaction was stirred at room temperature for 12 h. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure, and the crude product was subjected to silica gel column chromatography (DCM: MeOH ═ 30: 1 → 15: 1) to give 87mg of compound IIIc as a pale yellow solid in 45% yield. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C56H62N8O11, [ m + H]+:1023.4616,found:1023.4584.
1H NMR(400MHz,DMSO-d6)δ8.87(s,1H),7.96(dt,J=7.7,3.9Hz,2H),7.58(dt,J=7.5,1.0Hz,2H),7.42(dd,J=9.3,1.0Hz,2H),7.39-7.34(m,2H),7.31(dd,J=7.5,1.5Hz,1H),7.21(t,J=1.0Hz,1H),7.10(d,J=9.9Hz,1H),6.52(s,1H),5.27(dd,J=2.1,1.0Hz,2H),4.92(s,2H),4.77(s,1H),3.79(dt,J=12.4,7.1Hz,2H),3.74-3.62(m,4H),3.59(dt,J=12.5,1.1Hz,1H),3.52-3.41(m,1H),3.41-3.31(m,1H),3.25-3.10(m,4H),3.00(dq,J=12.5,8.1Hz,1H),2.61-2.51(m,8H),1.97(dq,J=12.3,8.0Hz,1H),1.91-1.80(m,4H),1.80-1.71(m,1H),1.27-1.19(m,12H),0.98(t,J=8.0Hz,3H)。
Example 4
Synthesis of Compounds IV a-c
Figure BDA0002142994190000141
Preparation of Compound IV a
100mg (0.242mmol) of compound XII are placed in a 25mL round-bottomed flask, dissolved in anhydrous DCM (5mL), 138mg (0.364mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethylurea hexafluorophosphate, 134. mu.L (0.728mmol) of N, N-diisopropylethylamine are added thereto, and after stirring for 0.5h in an ice bath, 134mg (0.290mmol) of compound V are added and the reaction is continued for 12h at room temperature. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure. The crude product was chromatographed on a silica gel column (DCM: MeOH ═ 30: 1 → 15: 1) to give compound IV a as 87mg as a pale yellow solid in 42% yield. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C47H47N5O11, [ m + H]+:858.3350,found:858.3375.
1H NMR(400MHz,DMSO-d6)δ9.54(s,1H),8.94(s,1H),8.86(s,1H),7.85(dd,J=7.5,1.4Hz,1H),7.80(d,J=7.6Hz,1H),7.72(t,J=7.0Hz,1H),7.47-7.40(m,2H),7.38-7.32(m,2H),7.16-7.08(m,2H),6.53(s,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),4.26(td,J=7.1,1.2Hz,2H),3.52(dq,J=12.3,7.1Hz,1H),3.43(dq,J=12.5,7.1Hz,1H),3.22-3.11(m,4H),3.01(dq,J=12.5,8.1Hz,1H),2.68(t,J=7.1Hz,2H),2.64-2.47(m,3H),2.44-2.34(m,1H),2.12-2.01(m,1H),1.97(dt,J=12.5,7.9Hz,1H),1.26(s,2H),1.25-1.17(m,8H),1.06(t,J=8.0Hz,3H)。
Preparation of Compound IV b
100mg (0.242mmol) of compound XII are placed in a 25mL round-bottom flask, dissolved in anhydrous DCM (5mL), 138mg (0.364mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethyluronium hexafluorophosphate, 134. mu.L (0.728mmol) of N, N-diisopropylethylamine are added thereto, the mixture is stirred for 0.5h in an ice bath, 129mg (0.290mmol) of compound VI are added, and the reaction is continued at room temperatureThe reaction was stirred for 12 h. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure, and the crude product was subjected to silica gel column chromatography (DCM: MeOH ═ 30: 1 → 15: 1) to give compound IV b as a pale yellow solid (81 mg, yield 41%). m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C46H45N5O11, [ m + H]+:866.3013,found:866.3044.
1H NMR(400MHz,DMSO-d6)δ9.54(s,1H),8.94(s,1H),8.86(s,1H),7.89(t,J=7.9Hz,1H),7.85(dd,J=7.5,1.4Hz,1H),7.80(d,J=7.6Hz,1H),7.47-7.40(m,2H),7.38-7.32(m,2H),7.16-7.08(m,2H),6.53(s,1H),5.17(t,J=1.0Hz,2H),4.92(s,2H),4.26(td,J=7.1,1.2Hz,2H),3.96(d,J=7.9Hz,2H),3.22-3.11(m,4H),3.01(dq,J=12.5,8.1Hz,1H),2.64-2.54(m,1H),2.54-2.45(m,2H),2.44-2.35(m,1H),2.12-2.01(m,1H),1.97(dt,J=12.5,7.9Hz,1H),1.26(s,2H),1.25-1.17(m,8H),1.06(t,J=8.0Hz,3H)。
Synthesis of Compound IV c
100mg (0.242mmol) of compound XII are placed in a 25mL round-bottomed flask, dissolved in anhydrous dichloromethane (5mL), 138mg (0.364mmol) of O- (7-azobenzotriazol-1-oxy) -N, N' -tetramethylurea hexafluorophosphate and 134. mu.L (0.728mmol) of N, N-diisopropylethylamine are added thereto, and after stirring for 0.5h in an ice bath, 204mg (0.290mmol) of compound VII are added, and the reaction is continued for 12h at room temperature. Dilute with dichloromethane (5mL), wash with water (10mL x3), wash with saturated sodium chloride solution (10mL), dry over anhydrous sodium sulfate. The solvent was removed by distillation under the reduced pressure, and the crude product was subjected to silica gel column chromatography (DCM: MeOH ═ 30: 1 → 15: 1) to give compound IV c as a pale yellow solid (99 mg) with a yield of 45%. m.p. is greater than 200 ℃; hrms (esi): m/z Calcd for C50H52N6O11, [ m + Na]+:935.3592,found:935.3577.
1H NMR(400MHz,DMSO-d6)δ9.54(s,2H),8.86(s,2H),7.96(d,J=7.6Hz,2H),7.85(dd,J=7.5,1.5Hz,2H),7.47-7.40(m,6H),7.36-7.28(m,4H),7.21(t,J=1.0Hz,2H),7.00(d,J=9.9Hz,2H),6.53(s,2H),5.27(dd,J=2.1,1.0Hz,4H),4.92(s,4H),4.77(s,2H),4.26(t,J=7.1Hz,4H),3.82-3.76(m,3H),3.75(s,1H),3.73-3.62(m,6H),3.22-3.10(m,8H),3.00(dq,J=12.5,8.1Hz,2H),2.60-2.48(m,6H),2.45-2.35(m,2H),1.97(dq,J=12.3,8.0Hz,2H),1.88(q,J=7.1Hz,8H),1.77(dq,J=12.3,7.9Hz,2H),1.25-1.19(m,18H),0.98(t,J=8.0Hz,6H)。
Example 5
Cell cycle experiments with Compounds IIa, IIb and IIc
To demonstrate the effect of the compounds on the cell cycle, the effect of the compounds on the cell cycle was determined by flow cytometry. Cells in logarithmic growth phase are taken and inoculated in a six-hole culture plate, and the compound is prepared into a certain concentration for treatment. After 24h, cells were collected, stained with propidium iodide (10 μ g/ml) for 30min, and cell cycle and apoptosis were detected using a flow cytometer. The experimental results are shown in FIG. 1; in the figure, A is a graph of the results of cell cycle experiments which have 24h of action in A549 cells; in the figure, B is a graph showing the results of cell cycle experiments performed for 24 hours in the Capan-1 cells; in the figure, C is a graph showing the results of a cell cycle experiment in which HCT116 cells were affected for 24 hours; the result shows that SN38 has strong effect of inducing cell division to stay in S phase. The three compounds have similar effects to AUY922 on three cell strains, and have stronger effect of blocking cell cycle. All three compounds maintain the properties of AUY922 and SN 38.
Example 6
Western blot experiments for Compounds II a, IIb and IIc
Hsp90 inhibitors have a significant inhibitory effect on Hsp90 chaperone, while SN38 has a significant inhibitory effect on topoisomerase 1. In order to study the inhibitory activity of compounds on Hsp90 chaperone protein and Top 1 at the cellular level, western blot experiments were performed using AUY922 and SN38 as positive controls, and the results are shown in fig. 2; in the figure, A is a protein immunoblotting experimental diagram in A549 cells; in the figure, B is a protein immunoblotting experimental picture in the Capan-1 cell; in the figure, C is a protein immunoblotting experiment picture in HCT116 cells; the results show that the compound has certain inhibitory activity on Hsp90 chaperone protein and Top 1 for three cells of A549, Capan-1 and HCT116 at 12h, which indicates that the compound maintains the common properties of an Hsp90 inhibitor and SN38, and reflects that the coupling of the two substances does not influence the properties of the substances to a certain extent.
Example 7
Experiment of proliferation inhibitory Activity of Compounds II a, IIb and IIc on A549, HCT116, MIA-PACA-2 and Capan-1 cell lines
In order to evaluate the influence of the compound on the proliferation capacity of tumor cells, a sulforhodamine B protein staining method is adopted, A549, HCT116, MIA-PACA-2 and Capan-1 cell strains are selected, after 72 hours of action, the proliferation inhibition rate of the compound on the tumor cells is observed, and IC is calculated50
Table 1 shows the proliferation inhibitory activity of compounds I a, Ib, Ic, IIa, IIb and IIc on A549, HCT116, MIA-PACA-2 and Capan-1 cell lines;
TABLE 1
Figure BDA0002142994190000161
The results of the cell experiments in Table 1 show that the compounds all show certain antitumor activity, and are better or equivalent to that of the SN38 prodrug irinotecan. Among them, the compound IIa is the most active, the compound IIb is the least active, and the compound IIc is the least active, consistent with the stability of the connecting chain. Compound IIa has relatively low stability and highest cytotoxic activity. The above experimental results show that the compounds II a, IIb and IIc maintain good antitumor activity against A549, HCT116, MIA-PACA-2 and Capan-1 cell lines.
Example 8
Experiment on proliferation inhibitory Activity of Compounds IIIa, IIIb, IIIc, IVa, IVb and IVc on A549, BGC823, HCT116, MIA-PACA-2, PSN-1 and T47D cell lines
In order to evaluate the influence of the compound on the proliferation capacity of tumor cells, a sulforhodamine B protein staining method is adopted to select A549, BGC823, HCT116, MIA-PACA-2, PSN-1 and T47D cell line, observing the proliferation inhibition rate of the compound on the tumor cells after 72 hours of action and calculating IC50
Table 2 shows the proliferation inhibitory activities of the compounds III a, IIIb, III c and IV a-c on cell lines A549, BGC823, HCT116, MIA-PACA-2, PSN-1 and T47D;
TABLE 2
Figure BDA0002142994190000171
The results of cell experiments in Table 2 show that the compounds all show certain antitumor activity. Of these, the activity of compounds IIIb, IIIc, IVa and IVb is higher compared to ADR. The activity of compounds IIIc and IVa on four cell lines BGC823, MIA-PACA-2, PSN-1 and T47D was about 10 times that of ADR. About 3-fold higher than ADR on a549 cell line; activity on HCT116 cells was comparable to ADR. The above experimental results show that the compounds IIIc and IVa maintain good antitumor activity against A549, BGC823, HCT116, MIA-PACA-2, PSN-1 and T47D cell lines.

Claims (10)

1. A conjugate of an Hsp90 inhibitor and a camptothecin derivative, wherein the conjugate is linked by a linking chain and has the formula Ia or Ib, Ic; IIa or IIb, IIc; IIIa or IIIb, IIIc; IVa or IVb, IVc:
Figure FDA0002142994180000011
Figure FDA0002142994180000021
wherein in formula Ia or Ib, IIa or IIb, IIIa or IIIb, IVa or IVb, n is 2 or 1; the Hsp90 inhibitor is NVP-AUY922, SNX-5422, AT13387, NVP-BEP800, KW2478, XL888 or PU-H71; the camptothecin derivative is camptothecin, 7-ethyl-10-hydroxycamptothecin or topotecan; the connecting chain is an amino acid ester bond, a urethane bond, a carbonate bond, an isocyanate bond or a thioisocyanate bond.
2. A method for preparing the conjugate of claim 1, wherein the method comprises the following steps:
Figure FDA0002142994180000031
the reaction steps comprise:
a) condensation reaction: under the action of a condensing agent, reacting the compound VIII with a compound V, VI and a compound VII respectively in an organic solvent to generate compounds Ia, Ib and Ic; the molar ratio of the compound VIII to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound VIII to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
3. The preparation method according to claim 2, wherein in step a), the condensing agent is one or more of O- (7-azobenzotriazol-1-oxy) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
4. A method for preparing the conjugate of claim 1, wherein the method comprises the following steps:
Figure FDA0002142994180000041
the reaction steps comprise:
a) hydrogenation reaction: carrying out hydrogenation reaction in an organic solvent of a compound IX under the conditions of a palladium catalyst and hydrogen to generate a compound X; the mass ratio of the compound IX to the palladium catalyst is 1: 0.1, and the reaction time is 4 hours;
b) condensation reaction: under the action of a condensing agent, reacting a compound X with a compound V, VI and a compound VII in a right organic solvent to generate compounds IIa, IIb and IIc; the molar ratio of the compound X to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound X to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
5. The preparation method according to claim 4, wherein in step a), the organic solvent is one or more of methanol, ethanol and isopropanol; the palladium catalyst is one or a mixture of more of 10% palladium carbon, palladium and palladium hydroxide; in the step b), the condensing agent is one or a mixture of more of O- (7-azobenzotriazol-1-oxide) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
6. A method for preparing the conjugate of claim 1, wherein the method comprises the following steps:
Figure FDA0002142994180000051
the reaction steps comprise:
a) condensation reaction: under the action of a condensing agent, reacting a compound XI with a compound V, VI and a compound VII in an organic solvent to generate compounds IIIa, IIIa and IIIc; the molar ratio of the compound XI to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound XI to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
7. The preparation method according to claim 6, wherein in step a), the condensing agent is one or more of O- (7-azobenzotriazol-1-oxy) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
8. A method for preparing the conjugate of claim 1, wherein the method comprises the following steps:
Figure FDA0002142994180000052
the reaction steps comprise:
a) condensation reaction: under the action of a condensing agent, reacting a compound XII with a compound V, VI and a compound VII in an organic solvent respectively to generate compounds VIa, VIb and VIc; the molar ratio of the compound XII to the compound V, VI to the compound VII is 1: 1.2, the molar ratio of the compound XII to the condensing agent is 1: 1.5, and the reaction time is 3-4 hours.
9. The preparation method according to claim 8, wherein in step a), the condensing agent is one or more of O- (7-azobenzotriazol-1-oxy) -N, N '-tetramethylurea hexafluorophosphate, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N' -diisopropylcarbodiimide and 1-hydroxybenzotriazole; the organic solvent is one or a mixture of DMF, DCM, DMSO and THF.
10. Use of the conjugate of claim 1 for the preparation of an anti-tumor medicament.
CN201910675023.5A 2019-07-25 2019-07-25 Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof Pending CN112279863A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910675023.5A CN112279863A (en) 2019-07-25 2019-07-25 Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910675023.5A CN112279863A (en) 2019-07-25 2019-07-25 Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN112279863A true CN112279863A (en) 2021-01-29

Family

ID=74418742

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910675023.5A Pending CN112279863A (en) 2019-07-25 2019-07-25 Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112279863A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114306632A (en) * 2022-01-04 2022-04-12 华东师范大学 7-ethyl-10-hydroxycamptothecin derivative prodrug non-covalently bound with human serum albumin, preparation and application

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2015125641A1 (en) * 2014-02-19 2017-03-30 日本化薬株式会社 Pharmaceutical composition comprising a polymeric camptothecin derivative and a polymeric HSP90 inhibitor derivative
CN110049783A (en) * 2016-12-14 2019-07-23 塔弗达治疗有限公司 HSP90- targets conjugate and its preparation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2015125641A1 (en) * 2014-02-19 2017-03-30 日本化薬株式会社 Pharmaceutical composition comprising a polymeric camptothecin derivative and a polymeric HSP90 inhibitor derivative
CN110049783A (en) * 2016-12-14 2019-07-23 塔弗达治疗有限公司 HSP90- targets conjugate and its preparation

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114306632A (en) * 2022-01-04 2022-04-12 华东师范大学 7-ethyl-10-hydroxycamptothecin derivative prodrug non-covalently bound with human serum albumin, preparation and application
CN114306632B (en) * 2022-01-04 2023-11-17 华东师范大学 Non-covalent binding type 7-ethyl-10-hydroxycamptothecin derivative prodrug with human serum albumin, preparation and application

Similar Documents

Publication Publication Date Title
CN108069946B (en) Substituted quinazoline compounds having the ability to cross the blood brain barrier
CA3009850C (en) Deuterated compounds for treating cancer and related diseases and conditions, and compositions and methods thereof
MXPA06011679A (en) Novel water-soluble prodrug.
KR20120113760A (en) Purified pyrroloquinolinyl-pyrrolidine-2,5-dione compositions and methods for preparing and using same
Antoszczak et al. Synthesis and antiproliferative activity of new bioconjugates of Salinomycin with amino acid esters
CN104163823B (en) A kind of camptothecine and Artesunate conjugate and preparation method thereof and application
JPH11511761A (en) Bicyclic 4-aralkylaminopyrimidine derivatives as tyrosine kinase inhibitors
WO2007023778A1 (en) Novel anticancer concomitant drug
CN108026046A (en) The purposes of substituted quinazoline compound and its inhibitor as G12C mutant KRAS, HRAS and/or NRAS protein
EP3418277B1 (en) Substituted amino six-membered nitric heterocyclic ring compound and preparation and use thereof
CN110551102B (en) ALK covalent inhibitors and uses thereof
CN110283162B (en) Epidermal growth factor receptor inhibitor and application thereof
EP3939592A1 (en) Application of combination of quinoline derivative and immunomodulator in preparation of antitumor drugs
CN112279863A (en) Conjugate of Hsp90 inhibitor and camptothecin derivative as well as preparation method and application thereof
CN113366008A (en) CD73 inhibitor, preparation method and application thereof
CN115819418A (en) PLK1 kinase inhibitor and preparation method and application thereof
WO2019001307A1 (en) Amide compound, composition containing same, and use thereof
TWI794576B (en) A class of fluorine-substituted benzothiophene compounds and their pharmaceutical compositions and applications
CN115433207A (en) Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof
CN114072396B (en) Quinoline and cinnoline derivatives as DNA-PK inhibitors
RU2768451C1 (en) Selective receptor antagonist type a2a
CN109153639A (en) Faenum graecum alkaloid compound
CN113292484A (en) 3- (4-methylpiperidine-1-yl) -3-benzylurea compound and analogue, preparation method and application thereof
CN102731516A (en) Novel camptothecin derivatives having antineoplastic activity
JP4305909B2 (en) Hexacyclic compounds

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
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210129

WD01 Invention patent application deemed withdrawn after publication