CN110922450B - PSMA activated antitumor prodrug CPT-X and preparation method and application thereof - Google Patents

PSMA activated antitumor prodrug CPT-X and preparation method and application thereof Download PDF

Info

Publication number
CN110922450B
CN110922450B CN201811091945.3A CN201811091945A CN110922450B CN 110922450 B CN110922450 B CN 110922450B CN 201811091945 A CN201811091945 A CN 201811091945A CN 110922450 B CN110922450 B CN 110922450B
Authority
CN
China
Prior art keywords
cpt
compound
preparation
psma
nmr
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.)
Active
Application number
CN201811091945.3A
Other languages
Chinese (zh)
Other versions
CN110922450A (en
Inventor
徐冰
王鹏龙
郭文博
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.)
Xinhuozhiyao Beijing Technology Co ltd
Original Assignee
Xinhuozhiyao Beijing 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 Xinhuozhiyao Beijing Technology Co ltd filed Critical Xinhuozhiyao Beijing Technology Co ltd
Priority to CN201811091945.3A priority Critical patent/CN110922450B/en
Publication of CN110922450A publication Critical patent/CN110922450A/en
Application granted granted Critical
Publication of CN110922450B publication Critical patent/CN110922450B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/02Linear peptides containing at least one abnormal peptide link
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

The invention provides a compound with a structural general formula 1, a preparation method thereof and application thereof in preparing antitumor drugs. The PSMA activated prodrug CPT-HT-J-ZLn shows strong cytotoxic selectivity to PSMA positively expressed and PSMA negatively expressed tumor cells, and the activity of the PSMA activated prodrug on LNCaP-FGC positively expressed tumor cells is stronger than that of PSMA negatively expressed tumor cells such as PC-3, DU145, HepG2, Hela and MCF-7. Wherein the compound CPT-HT-J-ZL12For LNCaP-FGC (IC)50=1.00±0.20μM)(PSMA+) Respectively, HepG2 (IC) of cells negatively expressing PSMA50>40.00μM),Hela(IC50>40.00μM),MCF‑7(IC50=21.68±4.96μM),DU145(IC505.40 ± 1.22 μ M) and PC-3 (IC)5042.96 ± 3.69 μ M) of 40,40,21,5 and 40 times.

Description

PSMA activated antitumor prodrug CPT-X and preparation method and application thereof
Technical Field
The invention relates to a compound and a preparation method and application thereof, in particular to a compound with targeted antitumor activity and a preparation method and application thereof, belonging to the field of pharmaceutical chemistry.
Background
Cancer remains one of the major diseases that seriously threaten human health and life, especially in China, and the situation is much more optimistic. Recent data published by the cancer center of china in 2018 in 2 months show that about 1 million people per day are diagnosed as cancers in china, and on average, 7 people per minute, which imposes a heavy economic burden on individuals, families and society. The most common and effective means for treating cancer is to remove the cancerous tissue by surgery, with chemotherapy or radiotherapy as an adjunct. Radiotherapy and chemotherapy remain the most effective methods for cancer patients who are inoperable or patients with advanced stages of cancer metastasis. However, the conventional chemotherapy drugs are not selective, inevitably cause damage to normal cells, cause large toxic and side effects, and seriously reduce the life quality of cancer patients and the compliance to subsequent treatment; therefore, the development of specific targeting drugs for cancer or the improvement of the targeting specificity of chemotherapeutic drugs to cancer cells will undoubtedly greatly improve the therapeutic effect on cancer.
The Prostate Specific Membrane Antigen (PSMA) is a type II transmembrane glycoprotein located on the surface of Prostate epithelial cells. Research finds that PSMA is highly expressed in prostate tumor tissues and tumor-related neovascular endothelial cells of various malignant non-prostate solid tumors such as liver cancer, pancreatic cancer, breast cancer and the like, but is not expressed in blood vessels of normal tissues adjacent to the tumor tissues, and the PSMA has the activities of folate hydrolase and N-acetylated alpha-linked acidic dipeptidase (NAALADa-se) and can specifically hydrolyze oligopeptide substrates consisting of Asp and Glu. Based on the characteristic of high expression of PSMA in tumor neovascular endothelial cells and the activity of carboxypeptidase, PSMA becomes a good target and a research hotspot for the tumor anti-vascular targeted therapy.
Camptothecin (CPT) is a monomeric active ingredient separated from traditional Chinese medicine Camptotheca acuminata (Camptotheca acuminata decne. var. acuminata), which is currently the third plant anticancer drug in the world, and Camptothecin drugs developed based on Camptothecin are clinically common broad-spectrum anticancer drugs and are long-lasting. Although camptothecin and derivatives thereof show better antitumor activity in vivo and in vitro experiments, the compounds mostly have the defects of poor targeting property and large toxic and side effects, have strong toxic and side effects on digestive systems and urinary systems, are easy to cause diarrhea, hemorrhagic cystitis, severe bone marrow suppression and the like, and have the defects of poor water solubility and poor stability, and the lactone ring of the compounds is easy to crack in human plasma to cause inactivation or activity reduction, so that the further development and clinical application of the compounds are limited, and the development of a new generation of camptothecin derivatives with high efficiency, low toxicity and good water solubility is significant.
The invention uses the splicing principle of medicinal chemistry for reference, and carries out splicing on an antitumor medicament Camptothecin (CPT) and a PSMA specific enzymolysis oligopeptide substrate X (Glu Asp-Glu) by a chemical method to synthesize a PSMA enzyme activated targeted antitumor prodrug CPT-X; activity on such compoundsThe evaluation was mainly spread around the antitumor aspect, and the analogs were tested for positive expression of PSMA (PSMA), respectively+) Tumor cell line LNCaP-FGC, PSMA negative expression (PSMA)-) Tumor cell lines HepG2, Hela, MCF-7, DU145, PC-3 cytotoxic activity.
Disclosure of Invention
The invention aims to provide a compound with a structural general formula 1 and a preparation method thereof.
The invention also aims to provide application of the compound 1 in preparing antitumor drugs.
The invention also aims to provide a pharmaceutical composition with an anti-tumor effect.
The purpose of the invention is realized by the following technical scheme:
a compound having the structure of formula 1 or a pharmaceutically acceptable salt thereof,
Figure GDA0003079749070000021
n is 1,3, 5, 11.
Further, the compound numbers and structural formulas of the invention are as follows:
Figure GDA0003079749070000022
Figure GDA0003079749070000031
Figure GDA0003079749070000041
Figure GDA0003079749070000051
furthermore, the compound can be added with conventional auxiliary materials in the field of preparation to prepare conventional formulations such as tablets, capsules, granules, powder, oral liquid, injection and the like.
The preparation method of the compound comprises the following steps:
the compound of the invention is prepared by the following method:
compound CPT-A-L2The preparation method comprises the following steps: dissolving camptothecin in organic solvent, and reacting with Boc-L-glycine under the action of condensing agent and catalyst to obtain CPT-A-L2
Compound CPT-A-L4The preparation method comprises the following steps: dissolving camptothecin in organic solvent, and reacting with N-Boc-gamma-aminobutyric acid under the action of condensing agent and catalyst to generate CPT-A-L4
Compound CPT-A-L6The preparation method comprises the following steps: dissolving camptothecin in organic solvent, reacting with tert-butyloxycarbonyl acyl 6-aminocaproic acid in the presence of condensing agent and catalyst to obtain CPT-A-L6
Compound CPT-A-L12The preparation method comprises the following steps: dissolving camptothecin in organic solvent, and reacting with 12- (Boc-amino) dodecanoic acid under the action of condensing agent and catalyst to obtain CPT-A-L12
Compound CPT-B-L2The preparation method comprises the following steps: mixing CPT-A-L2Dissolving in organic solvent, and generating CPT-B-L under the action of TFA2
Compound CPT-B-L4The preparation method comprises the following steps: mixing CPT-A-L4Dissolving in organic solvent, and generating CPT-B-L under the action of TFA4
Compound CPT-B-L6The preparation method comprises the following steps: mixing CPT-A-L6Dissolving in organic solvent, and generating CPT-B-L under the action of TFA6
Compound CPT-B-L12The preparation method comprises the following steps: mixing CPT-A-L12Dissolving in organic solvent, and generating CPT-B-L under the action of TFA12
Compound CPT-HT-J-L2The preparation method comprises the following steps: mixing CPT-B-L2Dissolving in organic solvent, and reacting with HT-J under the action of condensing agent and catalyst to obtain CPT-HT-J-L2
Compound CPT-HT-J-L4The preparation method comprises the following steps: mixing CPT-B-L4Is dissolved inOrganic solvent, and HT-J under the action of condensing agent and catalyst to produce CPT-HT-J-L4
Compound CPT-HT-J-L6The preparation method comprises the following steps: mixing CPT-B-L6Dissolving in organic solvent, and reacting with HT-J under the action of condensing agent and catalyst to obtain CPT-HT-J-L6
Compound CPT-HT-J-L12The preparation method comprises the following steps: mixing CPT-B-L12Dissolving in organic solvent, and reacting with HT-J under the action of condensing agent and catalyst to obtain CPT-HT-J-L12
Compound CPT-HT-J-ZL2The preparation method comprises the following steps: mixing CPT-HT-J-L2Dissolved in an organic solvent in TFA Thioanole H2CPT-HT-J-ZL is generated under the action of 95 percent of O, 2.5 percent of O and 2.5 percent of O2
Compound CPT-HT-J-ZL4The preparation method comprises the following steps: mixing CPT-HT-J-L4Dissolved in an organic solvent in TFA Thioanole H2CPT-HT-J-ZL is generated under the action of 95 percent of O, 2.5 percent of O and 2.5 percent of O4
Compound CPT-HT-J-ZL6The preparation method comprises the following steps: mixing CPT-HT-J-L6Dissolved in an organic solvent in TFA Thioanole H2CPT-HT-J-ZL is generated under the action of 95 percent of O, 2.5 percent of O and 2.5 percent of O6
Compound CPT-HT-J-ZL12The preparation method comprises the following steps: mixing CPT-HT-J-L12Dissolved in an organic solvent in TFA Thioanole H2CPT-HT-J-ZL is generated under the action of 95 percent of O, 2.5 percent of O and 2.5 percent of O12
Compound CPT-C-GL2The preparation method comprises the following steps: mixing CPT-B-L2Dissolving in organic solvent, and reacting with N-Cbz-L-glutamic acid-5-benzyl ester under the action of condensing agent and catalyst to obtain CPT-C-GL2
Compound CPT-C-GL4The preparation method comprises the following steps: mixing CPT-B-L4Dissolving in organic solvent, and reacting with N-Cbz-L-glutamic acid-5-benzyl ester under the action of condensing agent and catalyst to obtain CPT-C-GL4
Compound CPT-C-GL6The preparation method comprises the following steps: mixing CPT-B-L6Dissolving in organic solvent, and reacting with N-Cbz-L-glutamic acid-5-benzyl ester under the action of condensing agent and catalyst to obtain CPT-C-GL6
Compound CPT-C-GL12The preparation method comprises the following steps: mixing CPT-B-L12Dissolving in organic solvent, and reacting with N-Cbz-L-glutamic acid-5-benzyl ester under the action of condensing agent and catalyst to obtain CPT-C-GL12
Compound CPT-D-GL2The preparation method comprises the following steps: mixing CPT-C-GL2Dissolving in organic solvent, and generating CPT-D-GL under the action of deprotection agent2
Compound CPT-D-GL4The preparation method comprises the following steps: mixing CPT-C-GL2Dissolving in organic solvent, and generating CPT-D-GL under the action of deprotection agent4
Compound CPT-D-GL6The preparation method comprises the following steps: mixing CPT-C-GL6Dissolving in organic solvent, and generating CPT-D-GL under the action of deprotection agent6
Compound CPT-D-GL12The preparation method comprises the following steps: mixing CPT-C-GL12Dissolving in organic solvent, reacting with N-Cbz-L-glutamic acid-5-benzyl ester under the action of deprotection agent to generate CPT-D-GL12
Wherein the reaction is carried out at-20 ℃ to 250 ℃; the organic solvent is ether, alcohol, alkane, aromatic hydrocarbon, ketone, alkyl halide, amide, nitrile, ester or their mixture in various proportion containing 1-20 carbon atoms; the catalyst is 4-Dimethylaminopyridine (DMAP); the condensing agent is 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI), 1, 3-Dicyclohexylcarbodiimide (DCC), N' -Diisopropylcarbodiimide (DIC) and 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDC); the deprotection agent used was trifluoroacetic acid (TFA).
Further, in the preparation method, the molar ratio of the corresponding raw materials to HT-J is 1: 0.1-1: 10; the molar ratio of the corresponding raw materials to the condensing agent is 1: 0.1-1: 10; the molar ratio of the corresponding raw materials to the catalyst is 1: 0.1-1: 10.
The reaction route of the invention is as follows:
route 1 Synthesis of intermediate HT-J
Figure GDA0003079749070000071
Reaction conditions and reagents (a) EDCI, HOBt, DIPEA, DCM,25 ℃,3 h; (b) piperidine, DMF,1 h; (c) wet Pd/C (5%), MeOH,12 h.
Scheme 2 Synthesis of PSMA activating prodrug CPT-HT-J-ZLn
Figure GDA0003079749070000081
Reaction conditions and reagents (d) EDCI, DMAP, DCM,25 ℃,12 h; (e) TFA, DCM,25 ℃ C, 2 h; (f) EDCI, HOBt, DIPEA, DCM,25 ℃ and 3 h; (g) TFA Thioaisole H2O=95:2.5:2.5,25℃,3h;
Route 3, Synthesis of PSMA metabolite CPT-D-GLn
Figure GDA0003079749070000082
Reaction conditions and reagents (h) EDCI, HOBt, DIPEA, DCM,25 ℃,3 h; (i) wet Pd/C (5%), MeOH,12 h.
The invention also provides application of the compound shown in the formula 1 in preparation of antitumor drugs.
Furthermore, the tumor is a lung cancer cell line, a liver cancer cell line, a stomach cancer cell line, a colon cancer cell line and a cervical cancer cell line.
The present invention also provides a pharmaceutical composition comprising a compound of formula 1, or a pharmaceutically acceptable salt thereof, in a therapeutically effective amount in admixture with at least one pharmaceutically acceptable excipient.
Further, the composition further comprises at least one conventional anticancer drug.
Still further, the anticancer drug is selected from cyclophosphamide, 5-fluorouracil, paclitaxel, doxorubicin, etoposide, irinotecan, oxaliplatin, cisplatin, or gemcitabine.
The present invention also provides a method of treating cancer comprising administering to a patient an effective amount of a compound of formula 1 or a pharmaceutically acceptable salt thereof.
To achieve the above dosage forms, pharmaceutically acceptable excipients, such as fillers, disintegrants, lubricants, suspending agents, binders, sweeteners, flavoring agents, preservatives, etc., are added in the preparation of the dosage forms, and the fillers include: starch, pregelatinized starch, lactose, mannitol, chitin, microcrystalline cellulose, sucrose, etc., and the disintegrant comprises: starch, pregelatinized starch, microcrystalline cellulose, sodium carboxymethyl starch, cross-linked polyvinylpyrrolidone, low-substituted hydroxypropylcellulose, cross-linked sodium carboxymethylcellulose, and the like, and the lubricant comprises: magnesium stearate, sodium dodecyl sulfate, talcum powder, silicon dioxide and the like, and the suspending agent comprises: polyvinylpyrrolidone, microcrystalline cellulose, sucrose, agar, hydroxypropyl methylcellulose, and the like, binders include starch slurry, polyvinylpyrrolidone, hydroxypropyl methylcellulose, and the like, and sweeteners include: saccharin sodium, aspartame, sucrose, sodium cyclamate, glycyrrhetinic acid and the like, and the flavoring agent comprises: sweetening agent and various essences, and the preservative comprises: parabens, benzoic acid, sodium benzoate, sorbic acid and its salts, benzalkonium bromide, chloroacetidine acetate, eucalyptus oil, etc.
By "pharmaceutically acceptable" in the context of this invention is meant that the compound or composition must be compatible chemically and/or toxicologically with the other ingredients included in the formulation.
By "therapeutically effective amount" is meant that the compounds of the present invention treat or prevent a particular disease or condition; reducing, ameliorating, or eliminating one or more symptoms of a particular disease; or an amount that prevents or delays the onset of a particular disease or condition.
The PSMA activated prodrug CPT-HT-J-ZLn shows a certain proliferation inhibition effect on various tumor cells, but the antitumor activity is obviously lower than that of a parent drug CPT; meanwhile, the activity of most of the prodrug CPT-HT-J-ZLn on PSMA (PSMA-negatively expressed tumor cells) reported in the literature of LNCaP-FGC is stronger than that of PC-3, DU145, HepG2, Hela and MCF-7, for example, the compound CPT-HT-J-ZL12LNCaP-FGC (IC) for cells positively expressing PSMA501.00 ± 0.20 μ M) was determined to be PSMA-positively expressed cell HepG2 (IC), respectively50>40.00μM),Hela(IC50>40.00μM),MCF-7(IC50=21.68±4.96μM),DU145(IC505.40 ± 1.22 μ M) and PC-3 (IC)5042.96 ± 3.69 μ M) of 40,40,21,5 and 40 times. Experimental example 1MTT method for observing the Effect of CPT-X of the composition of the present invention on the proliferation of PSMA-positively and negatively expressed tumor cells
1. Apparatus and materials
Thermo 3111 type CO2An incubator; HFsafe biosafety cabinet; multiskan GO microplate reader; jingli brand LD5-2B type desk type low speed centrifuge; olympus IX71 inverted fluorescence microscope modified RPMI-1640 medium, fetal bovine serum, 0.25% trypsin solution, thiazole blue, phosphate buffer (seimer feishell biochemical beijing limited); dimethyl sulfoxide (DMSO);
human hepatoma cell line HepG 2; human cervical cancer cell line Hela; human breast cancer cell line MCF-7; human prostate cancer cell line DU 145; human prostate cell line PC-3; human prostate cell line LNCaP-FGC.
Experimental drugs: compounds of the invention CPT-X (prepared as in examples 1-32, respectively); camptothecin (shanghai beide pharmaceutical science and technology limited);
2. method of producing a composite material
2.1 culture of different cell lines
DU145, HepG2, MCF-7 and LNCaP-FGC cells were cultured in 1640 medium containing 10% fetal bovine serum, and placed at 37 ℃ in 5% CO2Incubation in an incubator. The cells all grow in an adherent state, the growth condition is observed under an inverted microscope, and subculture is carried out when the number of the cells is proper.
Culturing PC-3 and Hela cells in DMEM containing 10% fetal calf serum, and standing at 37 deg.C with 5% CO2Incubation in an incubator. The cells all grow in an adherent state, the growth condition is observed under an inverted microscope, and subculture is carried out when the number of the cells is proper.
2.2 cell inhibition
DU145, HepG2, MCF-7, LNCaP-FGC, PC-3 and Hela cells were taken in logarithmic growth phase at 3X 103The amount per well was inoculated in 96-well culture plates in a medium containing 5% CO2The wet incubator is used for 24 hours at 37 ℃; each hole is respectivelyA gradient of 100. mu.L of test compound was added. Cell control and blank control groups were set, with 4 replicates per concentration in the drug group and 3 replicates in the cell control and blank control groups. Continuing culturing for 72h in incubator, adding 20 μ L MTT into each well, incubating for 4h, discarding supernatant, adding 100 μ L DMSO, oscillating for 10min, measuring absorbance at 490nm wavelength with microplate reader, recording result, and calculating IC of compound50Value, cell inhibition (%) - (1- (a dose-a blank)/(a normal-a blank)]×100%。
3. Results
3.1 IC of prodrugs CPT-HT-J-ZLn and PSMA metabolite CPT-D-GLn on 7 tumor cell lines (DU145, HepG2, MCF-7, NCaP-FGC, PC-3, Hela)50The value (n ═ 3) is shown in table 1
TABLE 1 IC of prodrugs CPT-HT-J-ZLn and PSMA metabolite CPT-D-GLn on different tumor cell lines50Value of
Figure GDA0003079749070000101
As can be seen from Table 1, most of the prodrug CPT-HT-J-ZLn shows a certain effect of inhibiting proliferation on various tumor cells, but the antitumor activity is obviously lower than that of the parent drug CPT; meanwhile, the activity of most of the prodrug CPT-HT-J-ZLn on PSMA (PSMA-negatively expressed tumor cells) reported in the literature of LNCaP-FGC is stronger than that of PC-3, DU145, HepG2, Hela and MCF-7, for example, the compound CPT-HT-J-ZL12For LNCaP-FGC (IC)50=1.00±0.20μM)(PSMA+) Respectively, HepG2 (IC)50>40.00μM),Hela(IC50>40.00μM),MCF-7(IC50=21.68±4.96μM),DU145(IC505.40 ± 1.22 μ M) and PC-3 (IC)50=42.96±3.69μM)(PSMA-) 40,40,21,5 and 40 times. Meanwhile, in five PSMA negative expression tumor cells of PC-3, DU145, HepG2, Hela and MCF-7, the activity of most of the prodrug CPT-HT-J-ZLn on the human breast cancer cell line MCF-7, the human prostate cell line PC-3 and the human prostate cell line DU145 is obviously superior to that of the human liver cancer cell lineThe cell line HepG2 and the human ovarian cancer cell line Hela present different cell sensitivities, and further structure-activity relationship analysis shows that the length of the carbon chain of the linker can also influence the anti-tumor activity of the linker, and the anti-tumor activity of the linker is reduced along with the increase of the carbon chain, such as the anti-tumor activity on MCF-7 and PC-3 cells CPT-HT-J-ZL2>CPT-HT-J-ZL4>CPT-HT-J-ZL6>CPT-HT-J-ZL12(ii) a Most metabolites CPT-D-GLn also show certain proliferation inhibition activity on various tumor cells, wherein the activities on MCF-7, PC-3 and DU145 are better, and the proliferation inhibition activity on Hela and HepG2 is poorer; the same structure-activity relationship shows that the length of the carbon chain of the linker can also influence the anti-tumor activity of the linker, and the anti-tumor activity is reduced along with the increase of the carbon chain; for example, CPT-D-GL, which is an antitumor Activity on MCF-7 and PC-3 cells2,CPT-D-GL4>CPT-D-GL6,CPT-D-GL12
4. Conclusion
The PSMA activated prodrug CPT-HT-J-ZLn shows strong cytotoxic selectivity on PSMA positively expressed and PSMA negatively expressed tumor cells, and the activity on PSMA highly expressed tumor cells reported by LNCaP-FGC is stronger than that of five PSMA negatively expressed tumor cells, namely PC-3, DU145, HepG2, Hela and MCF-7. Wherein the compound CPT-HT-J-ZL12For LNCaP-FGC (IC)50=1.00±0.20μM)(PSMA+) Respectively, HepG2 (IC) of cells negatively expressing PSMA50>40.00μM),Hela(IC50>40.00μM),MCF-7(IC50=21.68±4.96μM),DU145(IC505.40 ± 1.22 μ M) and PC-3 (IC)5042.96 ± 3.69 μ M) of 40,40,21,5 and 40 times. The compounds can be used for research of antitumor drugs.
Detailed Description
EXAMPLE 1 preparation of HT-A
500mg (1.215mmol) of Fmoc-L-aspartic acid 4-tert-butyl ester, 349.37mg (1.8225mmol) of EDCI and 246.26mg (1.8225mmol) of HOBT were weighed in this order into a 50mL single-neck flask, dissolved by addition of 20mL of anhydrous dichloromethane, stirred at room temperature for 0.5h, and 395.48mg (1.337mmol) of L-glutamic acid di-tert-butyl ester hydrochloride and 392.57mg (3.037 mmol) of L-glutamic acid di-tert-butyl ester hydrochloride were added5mmol) DIPEA, stirred at room temperature for 4 hours, TLC [ V (petroleum ether): V (ethyl acetate) ═ 2:1]The reaction was monitored for completion and the reaction was diluted to 50ml with dichloromethane and washed three times with 25ml of 1M hydrochloric acid solution each time, then with 25ml of saturated sodium bicarbonate solution, followed by dehydration with saturated brine, collection of the organic layer, drying of the organic layer with appropriate amount of anhydrous sodium sulfate, filtration, evaporation to dryness under reduced pressure, sample mixing with silica gel, Flash silica gel column chromatography (PE/EA ═ 10/1 to 3/1) to give 680mg of HT-a as a white solid. mp 116.6-116.9 ℃; 85.8 percent of Yield;1H-NMR(500MHz,CDCl3):δ1.44(m,27H,9×-CH3),1.90(m,1H),2.16(m,1H),2.31(m,2H),2.60(m,1H),2.99(m,1H),4.24(t,J=7.1Hz,1H),4.42(m,2H),4.45(m,1H),4.56(m,1H),5.99(d,J=8.5Hz,1H),7.14(d,J=7.6Hz,1H),7.32(t,J=7.5Hz,2H),7.40(t,J=7.5Hz,2H),7.60(m,2H),7.76(d,J=7.6Hz,2H).13C-NMR(126MHz,CDCl3):δ27.8,28.1,28.2,31.4,37.7,47.2,51.2,52.6,67.4,80.7,82.0,82.4,120.1,125.2,127.2,127.9,141.4,144.0,156.1,170.4,170.6,171.5,172.3;HRMS(ESI)m/z:[M+H]+653.3429,calcd.for C36H48N2O9 652.33598。
EXAMPLE 2 preparation of intermediate HT-B
588mg (0.900mmol) of HT-A are weighed out in a 50mL single vial, dissolved by sonication with 20mL of anhydrous DMF, followed by the addition of 229mg (2.7mmol) of piperidine and stirring at room temperature for 0.5h, TLC [ V (petroleum ether): V (ethyl acetate): 2:1]The reaction was monitored for completion, and the reaction solution was directly spun dry with a diaphragm pump, then dissolved in 50ml of ethyl acetate, and the organic layer was washed 3 times with 30ml of distilled water each time, dehydrated with saturated saline, dried over anhydrous sodium sulfate, filtered, stirred with silica gel, and subjected to Flash silica gel column chromatography (PE/EA ═ 3/1 to 0/1) to give 340mg of HT-B as a colorless oil. 87.9 percent of Yield;1H-NMR(500MHz,CDCl3):δ1.43(s,9H,3×-CH3),1.44(s,9H,3×-CH3),1.46(s,9H,3×-CH3),1.89(m,2H),2.29(m,2H),2.58(dd,J=16.7,8.0Hz,1H),2.79(dd,J=16.7,3.7Hz,1H),3.66(dd,J=8.0,3.7Hz,1H),4.45(td,J=8.4,4.9Hz,1H),7.89(d,J=8.4Hz,1H).13C-NMR(126MHz,CDCl3):δ28.1,28.2,28.3,29.8,31.6,40.6,52.1,52.2,80.7,81.4,82.3,171.2,171.3,172.3,173.5;HRMS(ESI)m/z:[M+H]+431.2751,calcd.for C21H38N2O7 430.26790。
EXAMPLE 3 preparation of intermediate HT-C
255mg (0.6mmol) of fluorenylmethyloxycarbonyl-L-glutamic acid 1-tert-butyl ester, 172mg (0.9mmol) of EDCI and 122mg (0.9mmol) of HOBT were sequentially weighed out and placed in a 50mL single-neck flask, 10mL of anhydrous dichloromethane was added to dissolve the components, the reaction was stirred at room temperature for 0.5h, 284mg (0.66mmol) of HT-B and 194mg (1.5mmol) of DIPEA were further added, the reaction was stirred at room temperature for 4h, and TLC [ V (petroleum ether): V (ethyl acetate): 1 ] was used]The reaction was monitored for completion, the reaction was diluted to 50ml with dichloromethane, washed three times with 25ml of 1M hydrochloric acid solution each time, then washed with 25ml of saturated sodium bicarbonate solution, then dehydrated with saturated brine, the organic layer was collected, dried over anhydrous sodium sulfate in appropriate amount, filtered, evaporated to dryness under reduced pressure, stirred on silica gel, and subjected to Flash silica gel column chromatography (PE/EA ═ 5/1 to 2/1) to give 450mg of HT-C as a white solid. mp 106.3-107.3 ℃; 88.1 percent of Yield;1H-NMR(500MHz,CDCl3):δ1.41(s,9H,3×-CH3),1.42(s,9H,3×-CH3),1.43(s,9H,3×-CH3),1.47(s,9H,3×-CH3),1.91(m,3H),2.12(m,1H),2.28(m,5H),2.60(dd,J=17.1,6.3Hz,1H),2.91(dd,J=17.2,3.8Hz,1H),4.22(dd,1H),4.40(m,3H),4.78(d,J=4.3Hz,1H),5.63(d,J=7.7Hz,1H),6.94(d,J=7.4Hz,1H),7.17(d,J=7.7Hz,1H),7.31(t,J=7.3Hz,2H),7.39(t,J=7.3Hz,2H),7.61(d,J=7.3Hz,2H),7.75(d,J=7.4Hz,2H).13C-NMR(126MHz,CDCl3):δ27.7,28.1,28.2,28.8,31.5,32.4,37.2,47.3,49.4,52.5,53.9,67.1,80.7,81.9,82.3,82.6,120.1,125.3,127.2,127.8,141.4,143.9,144.0,156.3,170.5,171.2,171.6,171.9,172.2.HRMS(ESI)m/z:[M+H]+838.4416,calcd.for C45H63N3O12 837.44117。
EXAMPLE 4 preparation of intermediate HT-D
427mg (0.5mmol) of HT-C was weighed into a 50mL single vial, dissolved by sonication with 20mL of anhydrous DMF, followed by addition of 127.7mg (1.5mmol) of piperidine and stirring at room temperature for 0.5h, TLC [ V (dichloromethane): V (methanol): 20: 1:)]Monitoring the reaction completion and the reaction solutionSpin-dry on a diaphragm pump, then dissolve with 50ml of ethyl acetate, wash the organic layer 3 times with 30ml of distilled water each time, dehydrate with saturated brine, dry over anhydrous sodium sulfate, filter, stir-sample with silica gel, Flash silica gel column chromatography (DCM/MeOH 1/0 to 20/1) to give 285mg of HT-D as a colorless oil. 90.4 percent of Yield;1H-NMR(500MHz,CDCl3):δ1.43(d,36H,12×-CH3),1.82(m,2H),1.91(m,2H),2.08(m,2H),2.28(m,2H),2.42(m,1H),2.51(m,1H),2.98(m,1H),3.30(m,1H),4.44(d,J=4.5Hz,1H),4.80(d,J=3.7Hz,1H),7.06(d,J=8.5Hz,1H),7.49(d,J=8.0Hz,1H).13C-NMR(126MHz,CDCl3):δ28.0,28.1,28.2,29.7,29.8,31.5,33.9,37.1,49.1,52.4,55.4,80.6,81.4,81.8,82.2,170.7,170.8,171.8,172.3,172.7,174.9.HRMS(ESI)m/z:[M+H]+616.3830,calcd.for C30H53N3O10615.37309。
EXAMPLE 5 preparation of intermediate HT-E
170mg (0.4mmol) of fluorenylmethyloxycarbonyl-L-glutamic acid 1-tert-butyl ester, 115mg (0.6mmol) of EDCI and 81mg (0.6mmol) of HOBT were weighed in this order and placed in a 50mL single-neck flask, 10mL of anhydrous dichloromethane was added to dissolve the resulting mixture, the reaction was stirred at room temperature for 0.5h, 278mg (0.44mmol) of HT-D and 129mg (1.0mmol) of DIPEA were further added, the reaction was stirred at room temperature for 4h, and TLC [ V (petroleum ether): V (ethyl acetate): 1 ] was used]The reaction was monitored for completion and the reaction was diluted to 50ml with dichloromethane and washed three times with 25ml of 1M hydrochloric acid solution each time, then 25ml of saturated sodium bicarbonate solution, followed by dehydration with saturated brine, collection of the organic layer, drying of the organic layer over anhydrous sodium sulfate, filtration, evaporation to dryness under reduced pressure, sample mixing on silica gel, Flash silica gel column chromatography (DCM/MeOH 1/0 to 30/1) to afford 360mg of HT-E as a white solid. mp 91.6-92.2 ℃; yield of Yield 86.7%;1H-NMR(500MHz,CDCl3):δ1.40(s,9H,3×-CH3),1.41(s,9H,3×-CH3),1.43(s,9H,3×-CH3),1.45(s,9H,3×-CH3),1.46(s,9H,3×-CH3),1.89(m,4H),2.25(m,10H),2.62(dd,J=17.2,6.8Hz,1H),2.84(dd,J=17.2,4.7Hz,1H),4.23(m,1H),4.40(m,2H),4.48(m,1H),4.76(m,1H),5.62(d,J=8.2Hz,1H),6.58(d,J=7.6Hz,1H),7.13(d,J=7.7Hz,1H),7.18(d,J=7.7Hz,1H),7.30(m,2H),7.38(m,2H),7.62(d,J=7.4Hz,2H),7.75(d,J=7.5Hz,2H).13C-NMR(126MHz,CDCl3):δ27.7,28.0,28.1,28.2,28.6,28.8,31.5,32.1,32.3,37.2,42.1,47.3,49.5,52.3,52.5,53.9,67.1,80.7,81.8,82.2,82.5,82.6,120.1,125.3,127.2,127.8,141.4,141.5,143.8,144.1,156.5,170.5,170.6,171.2,171.3,171.3,172.0,172.2,172.2.HRMS(ESI)m/z:[M+H]+1023.5528,calcd.for C54H78N4O15 1022.54637。
EXAMPLE 6 preparation of intermediate HT-F
350mg (0.337mmol) of HT-E was weighed into a 50mL single vial, dissolved by sonication with 20mL of anhydrous DMF, followed by 86.1mg (1.01mmol) of piperidine and stirred at room temperature for 0.5h, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, the reaction solution was directly spun dry with a diaphragm pump, then dissolved in 50ml of ethyl acetate, and the organic layer was washed 3 times with 30ml of distilled water each time, dehydrated with saturated brine, dried over anhydrous sodium sulfate, filtered, stirred on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0 to 20/1) to give 240mg of HT-F as a colorless oil. 87.3 percent of Yield;1H-NMR(500MHz,CDCl3):δ1.41(s,9H,3×-CH3),1.42(s,9H,3×-CH3),1.43(s,9H,3×-CH3),1.44(s,9H,3×-CH3),1.45(s,9H,3×-CH3),1.89(m,4H),2.09(m,2H),2.22(m,2H),2.31(t,J=7.3Hz,2H),2.38(t,J=7.0Hz,2H),2.61(dd,J=17.2,6.6Hz,1H),2.86(m,1H),3.36(m,J=8.4,4.8Hz,1H),4.42(m,2H),4.76(m,1H),6.89(d,J=7.6Hz,1H),7.13(d,J=7.8Hz,1H),7.22(d,J=7.8Hz,1H).13C-NMR(126MHz,CDCl3):δ27.7,28.1,28.2,28.5,30.2,31.5,32.5,32.9,37.2,49.5,52.3,52.5,54.5,80.7,81.5,81.8,82.3,82.4,170.5,170.6,171.2,171.4,172.2,172.3,172.8,174.8.HRMS(ESI)m/z:[M+H]+801.4883,calcd.for C39H68N4O13800.47829。
EXAMPLE 7 preparation of intermediate HT-I
80mg (0.248mmol) of 1-benzyl N- (t-butoxycarbonyl) -L-glutamate, 71.2mg (0.371mmol) of EDCI and 50.1mg (0.371mmol) of HOBT were weighed out in this order and placed in a 50mL one-neck flask, and 10mL of anhydrous dichloromethane was added thereto for dissolution, and the reaction was stirred at room temperature for 0.5h, and 223mg (0.273 m) of HOBT was further addedmol) HT-F and 80mg (0.62mmol) DIPEA, stirred at RT for 4 hours, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion and the reaction was diluted to 50ml with dichloromethane and washed three times with 25ml of 1M hydrochloric acid solution each time, then 25ml of saturated sodium bicarbonate solution, followed by dehydration with saturated brine, collection of the organic layer, drying of the organic layer over anhydrous sodium sulfate, filtration, evaporation to dryness under reduced pressure, sample mixing on silica gel, Flash silica gel column chromatography (DCM/MeOH 1/0 to 30/1) to afford 230mg of HT-I as a white solid. mp107.0-107.7 ℃; 83.6 percent of Yield;1H-NMR(500MHz,CDCl3):δ1.39(s,27H,9×-CH3),1.41(s,18H,6×-CH3),1.42(s,9H,3×-CH3),1.74(m,2H),2.13(s,6H),2.14(s,2H),2.22(m,2H),2.27(m,4H),2.62(dd,J=17.1,6.9Hz,1H),2.78(m,J=17.0,4.8Hz,1H),4.41(m,3H),4.75(dd,J=12.8,6.8Hz,1H),5.14(m,2H),5.57(d,J=8.3Hz,1H),6.52(m,1H),6.86(d,J=7.5Hz,1H),7.21(d,J=7.8Hz,1H),7.32(m,5H).13C-NMR(126MHz,CDCl3):δ27.7,28.0,28.1,28.4,31.0,31.5,31.6,32.0,32.2,37.3,49.5,52.0,52.2,52.4,53.1,67.2,80.1,80.6,81.6,82.1,82.4,128.4,128.5,128.7,135.5,156.0,170.6,170.6,171.0,171.1,171.3,171.9,172.1,172.2,172.3.HRMS(ESI)m/z:[M+H]+1120.6292,calcd.for C56H89N5O18 1119.62026。
EXAMPLE 8 preparation of intermediate HT-J
221mg (0.2mmol) of HT-I are weighed into a 50mL single vial, 20mL of anhydrous methanol is added, 5mg of 10% wet Pd/C is added, and the mixture is stirred under hydrogen catalysis at room temperature for 12h, TLC [ V (dichloromethane): V (methanol) ═ 10: 1%]The reaction was monitored for completion, the reaction solution was filtered to remove Pd/C, the filtrate was evaporated to dryness under reduced pressure, the mixture was stirred on silica gel and subjected to Flash silica gel column chromatography (DCM/MeOH: 40/0 to 20/1) to give 190mg of HT-J as a white solid. mp 94.3-95.0 deg.C; 92.2 percent of Yield;1H-NMR(500MHz,CDCl3):δ1.41(s,9H,3×-CH3),1.42(s,27H,9×-CH3),1.44(s,9H,3×-CH3),1.45(s,9H,3×-CH3),1.87(m,3H),2.08(m,1H),2.24(m,4H),2.33(m,4H),2.40(m,4H),2.73(m,2H),4.29(m,1H),4.43(m,2H),4.54(t,J=7.3Hz,1H),4.79(dd,J=12.7,7.2Hz,1H),5.56(d,J=7.3Hz,1H),6.57(s,1H),7.10(d,J=7.8Hz,1H),7.34(d,J=7.9Hz,1H),7.53(d,J=7.8Hz,1H).13C-NMR(126MHz,CDCl3):δ27.7,28.1,28.2,28.3,28.5,31.2,31.5,32.0,32.4,37.3,49.5,51.7,52.5,52.6,52.8,80.3,81.2,81.8,82.1,82.3,82.6,156.2,170.7,170.8,171.2,171.4,172.2,172.3,172.8,173.0,174.0.HRMS(ESI)m/z:[M+H]+1130.5793,calcd.for C49H83N5O181029.57331。
EXAMPLE 9 Compound CPT-A-L2Preparation of
2g (5.6mmol) of Camptothecin, 1.18g (6.72mmol) of N-Boc-glycine, 1.61g (8.4mmol) of EDCI and 0.068g (0.56mmol) of DMAP were weighed in this order and placed in a 100mL single-necked flask, and dissolved by adding 50mL of anhydrous dichloromethane, and the reaction was stirred at room temperature for 12 hours, TLC [ V (dichloromethane): V (methanol) ═ 20:1]The reaction was monitored for completion, and the reaction solution was diluted with dichloromethane to 150ml, washed with 50ml of distilled water three times each time, then dehydrated with saturated brine, the organic layer was collected, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0 to 30/1) to give 1.84g of CPT-A-L as a pale yellow solid2。mp>220℃;Yield:65.0%;1H-NMR(500MHz,CDCl3):δ0.98(t,J=7.4Hz,3H,-CH3),1.42(s,9H,3×-CH3),2.16(m,2H),4.06(dd,J=18.0,4.8Hz,1H),4.21(dd,J=18.4,6.1Hz,1H),5.07(s,1H),5.25(m,2H),5.38(d,J=17.1Hz,1H),5.67(d,J=17.1Hz,1H),7.30(s,1H),7.65(t,J=7.4Hz,1H),7.81(t,J=7.5Hz,1H),7.91(d,J=8.1Hz,1H),8.23(d,J=8.4Hz,1H),8.37(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,28.4,31.8,42.5,50.1,67.2,80.3,96.5,120.2,128.2,128.3,128.5,129.8,130.8,131.3,145.7,146.5,148.9,152.3,155.7,157.4,167.3,169.7.HRMS(ESI)m/z:[M+H]+506.1917,calcd.for C27H27N3O7505.18490。
EXAMPLE 10 Compound CPT-B-L2Preparation of
1.8g (3.64mmol) of CPT-A-L are weighed2Placing in a 100mL single-neck flask, adding 30mL of anhydrous dichloromethane to dissolve, adding 9mL of TFA under ice bath, and stirring at room temperature for 1 hr, TLC [ V (dichloromethane): V (methanol) ]10:1]Monitoring the reaction completion, spin-drying the reaction solution at low temperature by a diaphragm pump, mixing the sample with silica gel, and performing Flash silica gel column chromatography (DCM/MeOH: 20/1-8/1) to obtain 0.9g of CPT-B-L as a pale yellow solid2。mp 194.8-195.2℃;Yield:61.2%;1H-NMR(500MHz,CD3OD):δ1.08(t,J=7.5Hz,3H,-CH3),2.27(m,2H),4.18(m,1H),4.29(m,1H),5.35(m,2H),5.50(d,J=16.7Hz,1H),5.64(d,J=16.7Hz,1H),7.44(s,1H),7.1(t,J=7.3Hz,1H),7.88(t,J=7.5Hz,1H),8.07(d,J=8.0Hz,1H),8.15(d,J=8.0Hz,1H),8.64(s,1H).13C-NMR(126MHz,CD3OD):δ8.0,31.9,41.0,51.6,67.7,79.6,97.7,120.6,129.3,129.8,130.0,130.9,132.1,133.1,133.5,147.4,147.9,149.6,153.4,159.0,168.1,168.6.HRMS(ESI)m/z:[M+H]+406.1397,calcd.for C22H19N3O5 405.13247。
EXAMPLE 11 Compound CPT-HT-J-L2Preparation of
618.0mg (0.60mmol) of HT-J, 159.0mg (0.83mmol) of EDCI and 112.1mg (0.83mmol) of HOBT were weighed out in sequence and placed in a 50mL single-neck flask, 20mL of anhydrous dichloromethane were added to dissolve the mixture, the reaction was stirred at room temperature for 0.5h, and 223mg (0.55mmol) of CPT-B-L was added2And 178.0mg (1.38mmol) of DIPEA, stirred at room temperature for 4 hours, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted to 50ml with dichloromethane, washed three times with 25ml of distilled water each time, then dehydrated with saturated brine, collected as an organic layer, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, stirred on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH ═ 1/0 to 50/1) to give 240mg of CPT-HT-J-L as a pale yellow solid2。mp 167.4-168.1℃;Yield:30.0%;1H-NMR(500MHz,CDCl3):δ0.97(t,J=7.5Hz,3H,-CH3),1.26(s,2H),1.35(s,9H,3×-CH3),1.37(s,9H,3×-CH3),1.42(s,36H,12×-CH3),1.90(m,2H),2.19(m,14H),2.64(m,1H),2.83(m,1H),4.16(m,2H),4.42(m,4H),4.18(s,1H),5.29(s,2H),5.38(d,J=17.4Hz,1H),5.67(d,J=17.4Hz,1H),6.67(m,1H),7.11(m,1H),7.33(m,4H),7.67(t,J=6.9Hz,1H),7.83(t,J=7.7Hz,1H),7.94(d,J=7.9Hz,1H),8.24(d,J=8.3Hz,1H),8.40(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,27.7,28.0,28.1,28.2,28.4,29.4,31.6,31.9,32.5,37.4,41.4,49.4,50.2,51.8,52.3,52.5,53.9,67.2,79.9,80.7,81.6,82.0,82.3,82.5,82.6,96.5,120.3,128.2,128.3,128.6,129.7,130.8,131.4,145.6,146.4,148.8,152.3,156.0,157.4,167.2,169.5,170.9,171.0,171.7,172.3,172.6,172.7,172.9.HRMS(ESI)m/z:[M+H]+1417.7008,calcd.for C71H100N8O22 1416.69522。
EXAMPLE 12 Compound CPT-HT-J-ZL2Preparation of
Weighing 220mg (0.155mmol) of CPT-HT-J-L2Put into a 50mL single-neck flask, 10mL of hydrolysate (TFA: Thioanisole: H) was added295 percent of O, 2.5 percent of O and 2.5 percent of O, stirring and reacting for 3 hours at room temperature, evaporating the reaction solution to dryness at low temperature under reduced pressure, adding 10ml of ethyl acetate for precipitation, centrifuging and collecting the precipitate, continuously adding 10ml of ethyl acetate for precipitation, centrifuging and collecting the precipitate to obtain CPT-HT-J-ZL2Dissolving the crude product in methanol, separating liquid phase, and freeze drying to obtain 56mg of CPT-HT-J-ZL2。mp 179.0-179.5℃;Yield:19.9%;1H-NMR(500MHz,DMSO):δ0.93(t,J=7.4Hz,3H,-CH3),1.52(d,J=6.6Hz,2H),1.76(s,3H),1.96(s,6H),2.16(s,6H),2.26(m,5H),4.15(m,4H),4.58(m,2H),5.27(s,1H),5.35(s,1H),7.18(s,1H),7.73(t,J=7.5Hz,1H),7.88(t,J=7.5Hz,1H),8.06(d,J=7.1Hz,1H),8.16(m,6H),8.27(d,J=6.6Hz,1H),8.71(s,1H),8.93(s,1H).13C-NMR(101MHz,DMSO):δ7.6,26.2,27.1,29.9,30.4,31.5,31.7,36.2,36.5,49.3,51.3,51.5,51.7,66.3,76.6,95.1,119.0,127.8,128.0,128.7,128.9,129.8,130.5,131.7,145.0,146.1,147.9,152.4,156.5,167.0,168.6,169.0,171.0,171.2,171.4,171.7,173.1,173.3,173.4,173.8;HRMS(ESI)m/z:[M+H]+1037.3386,calcd.for C46H52N8O201036.32979。
EXAMPLE 13 Compound CPT-A-L4Preparation of
2.0g (5.6mmol) of Camptothecin, 1.36g (6.72mmol) of 57294-38-9, 1.61g (8.4mmol) of EDCI and 0.068g (0.56mmol) of DMAP were sequentially weighed out and placed in a 100mL single-neck flask, and 50mL of anhydrous dichloromethane was added thereto to dissolve the mixture, and the reaction was stirred at room temperature for 12 hours, TLC [ V (dichloromethane): V (methanol): 20: 1)]The reaction was monitored for completion, the reaction solution was diluted to 150ml with dichloromethane, washed three times with 50ml of distilled water each time, then dehydrated with saturated brine, the organic layer was collected, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0 to 30/1) to give 1.37g of CPT-A-L as a pale yellow solid4。mp 167.8-170.2℃;Yield:46.0%;1H-NMR(500MHz,CDCl3):δ0.98(t,J=7.5Hz,3H,-CH3),1.42(s,9H,3×-CH3),1.86(m,2H),2.14(m,2H),2.54(m,2H),3.14(m,2H),5.28(m,1H),5.38(m,1H),5.66(m,1H),7.23(m,1H),7.66(m,1H),7.82(m,1H),7.93(m,1H),8.25(m,1H),8.40(m,1H).13C-NMR(126MHz,CDCl3):δ7.7,25.1,28.5,31.1,31.8,39.6,50.1,67.2,76.0,79.3,96.1,120.3,128.2,128.3,128.6,129.6,130.9,131.5,146.1,146.3,148.8,152.3,156.2,157.5,167.7,172.3;HRMS(ESI)m/z:[M+H]+534.2231,calcd.for C29H31N3O7 533.21620。
EXAMPLE 15 Compound CPT-B-L4Preparation of
1.3g (2.44mmol) of CPT-A-L are weighed4Placing in a 100mL single-neck flask, adding 30mL of anhydrous dichloromethane to dissolve, adding 9mL of TFA under ice bath, and stirring at room temperature for 1 hr, TLC [ V (dichloromethane): V (methanol): 10:1]Monitoring the reaction completion, spin-drying the reaction solution at low temperature by a diaphragm pump, mixing the sample with silica gel, and performing Flash silica gel column chromatography (DCM/MeOH: 20/1-8/1) to obtain 0.69g of CPT-B-L as a pale yellow solid4。mp>220℃;Yield:65.3%;1H-NMR(500MHz,DMSO):δ0.93(t,J=7.4Hz,3H,-CH3),1.84(m,2H),2.16(m,2H),2.70(m,2H),2.83(m,2H),5.28(s,2H),5.53(m,2H),7.06(s,1H),7.71(t,J=6.0Hz,1H),7.86(d,J=6.7Hz,1H),8.13(d,J=7.5Hz,1H),8.16(d,J=7.5Hz,1H),8.69(s,1H).13C-NMR(126MHz,DMSO):δ7.7,22.5,30.33,38.0,50.3,66.4,76.1,94.8,118.9,127.8,128.1,128.7,129.0,129.9,130.5,131.7,145.4,146.2,148.0,152.4,156.6,167.4,171.6;HRMS(ESI)m/z:[M+H]+434.1715,calcd.for C24H23N3O5 433.16377。
EXAMPLE 16 Compound CPT-HT-J-L4Preparation of
618.0mg (0.60mmol) of HT-J, 159.0mg (0.83mmol) of EDCI and 112.1mg (0.83mmol) of HOBT were weighed out in sequence and placed in a 50mL single-neck flask, 20mL of anhydrous dichloromethane were added and dissolved, the reaction was stirred at room temperature for 0.5h, and 238.2mg (0.55mmol) of CPT-B-L were added4And 178.0mg (1.38mmol) of DIPEA, stirred at room temperature for 4 hours, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted to 50ml with dichloromethane, washed three times with 25ml of distilled water each time, then dehydrated with saturated brine, collected as an organic layer, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, stirred on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH ═ 1/0 to 50/1) to give 400mg of CPT-HT-J-L as a pale yellow solid4。mp 129.0-129.6℃;Yield:49.3%;1H-NMR(500MHz,CDCl3):δ0.95(t,J=7.4Hz,3H,-CH3),1.39(s,9H,3×-CH3),1.40(s,18H,6×-CH3),1.41(s,18H,6×-CH3),1.44(s,9H,3×-CH3),1.72(m,2H),1.84(m,4H),2.15(m,2H),2.26(m,4H),2.37(m,8H),2.56(m,2H),2.83(m,1H),3.30(m,2H),3.64(m,1H),4.11(m,1H),4.43(m,4H),4.79(m,1H),5.29(s,2H),5.39(d,J=17.2Hz,1H),5.65(d,J=17.2Hz,1H),5.79(m,1H),6.38(d,J=8.4Hz,1H),7.20(d,J=7.7Hz,1H),7.33(m,2H),7.53(s,1H),7.66(t,J=7.4Hz,1H),7.83(t,J=7.4Hz,1H),7.93(d,J=8.0Hz,1H),8.23(d,J=8.0Hz,1H),8.40(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,24.5,24.8,27.6,28.1,28.2,28.4,28.5,29.0,31.3,31.4,31.6,31.9,32.5,37.4,38.7,49.4,50.1,51.7,52.3,52.5,53.2,53.4,67.2,76.0,79.9,80.6,81.6,81.9,82.2,82.4,96.4,120.5,128.2,128.3,128.7,129.7,130.9,131.5,145.9,146.2,148.8,152.4,156.2,157.5,167.7,170.9,171.0,171.1,171.5,172.0,172.1,172.2,172.5,172.6,172.7;HRMS(ESI)m/z:[M+H]+1445.7339,calcd.for C73H104N8O221444.72652。
EXAMPLE 17 Compound CPT-HT-J-ZL4Preparation of
380mg (0.258mmol) of CPT-HT-J-L are weighed out4Put into a 50mL single-neck flask, 10mL of hydrolysate (TFA: Thioanisole: H) was added295% of O, 2.5% of O and 2.5% of O), stirring and reacting for 3h at room temperature, and reactingEvaporating the application solution at low temperature under reduced pressure, adding 10ml of diethyl ether for precipitation, centrifuging, collecting the precipitate to obtain CPT-HT-J-ZL4Dissolving the crude product in methanol, separating liquid phase, and freeze drying to obtain 56mg of CPT-HT-J-ZL4。mp 189.0-189.5℃;Yield:19.9%;1H-NMR(500MHz,DMSO):δ0.92(t,J=7.4Hz,3H,-CH3),1.73(m,4H),1.92(s,6H),2.16(s,6H),2.22(m,12H),2.63(m,2H),4.11(m,3H),4.55(s,1H),5.31(s,2H),5.48(d,J=16.9Hz,1H),5.52(d,J=17.1Hz,1H),7.07(s,1H),7.72(t,J=7.4Hz,1H),7.87(t,J=7.6Hz,1H),7.93(s,1H),8.09(s,1H),8.15(m,4H),8.24(s,2H),8.48(s,1H),8.70(s,1H).13C-NMR(126MHz,DMSO):δ7.6,24.3,27.1,27.2,27.6,29.6,30.3,30.6,30.8,31.5,31.7,36.3,38.1,50.3,51.3,51.4,51.7,51.9,52.1,66.4,75.9,94.7,118.9,127.8,128.0,128.6,129.0,129.9,130.5,131.7,145.4,146.1,147.9,152.4,156.6,167.4,168.5,170.7,171.1,171.5,171.8,172.4,172.9,173.6.HRMS(ESI)m/z:[M+H]+1065.3690,calcd.for C48H56N8O201064.36109。
EXAMPLE 18 Compound CPT-A-L6Preparation of
2g (5.6mmol) of Camptothecin, 1.55g (6.72mmol) of t-butoxycarbonyl 6-aminocaproic acid, 1.61g (8.4mmol) of EDCI and 0.068g (0.56mmol) of DMAP were sequentially weighed and placed in a 100mL single-neck flask, and 50mL of anhydrous dichloromethane was added thereto to dissolve the mixture, and the reaction was stirred at room temperature for 12 hours, TLC [ V (dichloromethane): V (methanol): 20:1 ]]The reaction was monitored for completion, the reaction solution was diluted to 150ml with dichloromethane, washed three times with 50ml of distilled water each time, then dehydrated with saturated brine, the organic layer was collected, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0-30/1) to give 1.73g of CPT-A-L as a pale yellow solid6。mp 152.0-152.5℃;Yield:55.0%;1H-NMR(500MHz,CDCl3):δ0.97(t,J=7.5Hz,3H,-CH3),1.39(s,9H,3×-CH3),1.50(m,2H),1.66(m,4H),2.14(m,2H),2.49(m,2H),3.07(m,2H),5.29(s,2H),5.39(d,J=17.0Hz,1H),5.67(d,J=17.0Hz,1H),7.23(s,1H),7.67(m,1H),7.83(m,1H),7.94(d,J=8.0Hz,1H),8.22(d,J=8.0Hz,1H),8.40(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,24.4,26.2,28.5,29.9,32.0,33.8,40.4,50.1,67.2,75.9,96.3,120.5,128.2,128.3,128.4,128.6,129.6,130.9,131.5,146.2,146.2,148.9,152.4,157.5,167.7,172.7;HRMS(ESI)m/z:[M+H]+562.2548,calcd.for C31H35N3O7 561.24750。
EXAMPLE 19 Compound CPT-B-L6Preparation of
1.7g (3.03mmol) of CPT-A-L are weighed6Placing in a 100mL single-neck flask, adding 30mL of anhydrous dichloromethane to dissolve, adding 9mL of TFA under ice bath, and stirring at room temperature for 1 hr, TLC [ V (dichloromethane): V (methanol): 10:1]Monitoring the reaction completion, spin-drying the reaction solution at low temperature by a diaphragm pump, mixing the sample with silica gel, and performing Flash silica gel column chromatography (DCM/MeOH: 20/1-8/1) to obtain 0.9g of CPT-B-L as a pale yellow solid6。mp 207.5-208.0℃;Yield:65.0%;1H-NMR(500MHz,CD3OD):δ1.05(t,J=7.4Hz,3H,-CH3),1.48(m,2H),1.70(m,4H),2.23(m,2H),2.62(m,2H),2.91(m,2H),5.27(s,2H),5.47(d,J=16.7Hz,1H),5.61(d,J=16.7Hz,1H),7.35(s,1H),7.70(m,1H),7.87(t,J=7.0Hz,1H),8.04(d,J=8.0Hz,1H),8.15(d,J=8.0Hz,1H),8.60(s,1H).13C-NMR(126MHz,CD3OD):δ8.1,25.1,26.6,28.2,32.1,34.2,40.5,51.5,67.7,77.5,97.8,120.6,129.2,129.7,129.7,130.0,130.9,132.0,133.4,147.5,148.4,149.5,153.4,159.0,169.6,173.9.HRMS(ESI)m/z:[M+H]+462.2030,calcd.for C26H27N3O5461.19507。
EXAMPLE 20 Compound CPT-HT-J-L6Preparation of
618.0mg (0.60mmol) of HT-J, 159.0mg (0.83mmol) of EDCI and 112.1mg (0.83mmol) of HOBT were weighed out in sequence and placed in a 50mL single-neck flask, 20mL of anhydrous dichloromethane were added and dissolved, the reaction was stirred at room temperature for 0.5h, and 254mg (0.55mmol) of CPT-B-L was added6And 178.0mg (1.38mmol) of DIPEA, stirred at room temperature for 4 hours, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted with dichloromethane to 50ml, washed three times with 25ml of distilled water each time, then dehydrated with saturated brine, and collectedDrying the organic layer and the organic layer with anhydrous sodium sulfate, filtering, evaporating to dryness under reduced pressure, mixing with silica gel, and performing Flash silica gel column chromatography (DCM/MeOH: 1/0-50/1) to obtain 400mg of CPT-HT-J-L as light yellow solid6。mp 138.5-139.0℃;Yield:45.2%;1H-NMR(500MHz,CDCl3):δ0.96(t,J=7.5Hz,3H,-CH3),1.41(s,27H,9×-CH3),1.42(s,9H,3×-CH3),1.43(s,9H,3×-CH3),1.45(s,9H,3×-CH3),1.52(m,2H),1.76(m,6H),2.12(m,4H),2.32(m,12H),2.48(m,2H),2.61(m,1H),2.86(m,1H),3.21(m,2H),4.11(s,1H),4.45(m,3H),4.80(m,1H),5.30(s,2H),5.40(d,J=17.2Hz,1H),5.66(d,J=17.2Hz,1H),5.84(d,J=7.0Hz,1H),6.34(d,J=8.3Hz,1H),7.23(d,J=7.4Hz,1H),7.27(s,1H),7.34(t,J=8.3Hz,1H),7.67(t,J=7.4Hz,1H),7.84(t,J=7.2Hz,H),7.94(d,J=8.0Hz,1H),8.25(d,J=8.5Hz,1H),8.41(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,24.4,26.3,27.6,27.9,28.1,28.2,28.6,29.0,29.2,31.6,32.0,32.1,32.5,33.7,37.4,39.4,49.4,50.1,51.7,52.3,52.6,53.3,67.2,75.8,79.9,80.6,81.6,81.9,82.2,82.5,96.5,120.6,128.3,128.4,128.7,129.5,131.0,131.7,146.1,148.6,152.3,156.2,157.5,167.7,170.9,171.1,171.5,171.7,172.2,172.5,172.7,172.8.HRMS(ESI)m/z:[M+H]+1473.7646,calcd.for C75H108N8O221472.75782。
EXAMPLE 21 Compound CPT-HT-J-ZL6Preparation of
380mg (0.258mmol) of CPT-HT-J-L are weighed out6Put into a 50mL single-neck flask, 10mL of hydrolysate (TFA: Thioanisole: H) was added295 percent of O, 2.5 percent of O and 2.5 percent of O, stirring and reacting for 3 hours at room temperature, evaporating the reaction solution to dryness at low temperature under reduced pressure, adding 10ml of ethyl acetate for precipitation, centrifuging and collecting the precipitate, continuously adding 10ml of ethyl acetate for precipitation, centrifuging and collecting the precipitate to obtain CPT-HT-J-ZL6Dissolving the crude product in methanol, separating liquid phase, and freeze drying to obtain 56mg of CPT-HT-J-ZL6。mp 190.0-190.4℃;Yield:25.0%;1H-NMR(500MHz,DMSO):δ0.93(t,J=7.5Hz,3H,-CH3),1.36(m,4H),1.48(m,2H),1.60(m,2H),1.79(m,3H),1.94(m,5H),2.21(m,9H),2.36(m,1H),2.66(m,1H),3.11(m,2H),3.62(m,1H),3.72(s,1H),4.20(m,3H),4.60(m,1H),5.31(s,2H),5.49(m,2H),7.06(s,1H),7.72(t,J=7.1Hz,1H),7.87(t,J=7.7Hz,1H),8.07(d,J=6.8Hz,1H),8.15(m,5H),8.33(m,2H),8.70(s,1H).13C-NMR(126MHz,DMSO):δ7.6,24.0,25.7,27.0,27.7,28.5,29.7,30.0,30.3,30.4,31.6,31.7,33.1,36.2,38.6,49.3,50.3,51.3,51.5,51.7,51.8,54.9,66.4,75.7,94.7,118.9,127.8,128.1,128.6,129.0,129.9,130.5,131.7,145.5,146.0,147.9,152.3,156.6,163.1,167.4,167.9,170.9,171.0,171.2,171.5,171.7,172.1,172.8,172.9,173.1,173.4,173.8.HRMS(ESI)m/z:[M+H]+1093.4007,calcd.for C50H60N8O20 1092.39239。
EXAMPLE 22 Compound CPT-A-L12Preparation of
2g (5.6mmol) of Camptothecin, 2.18g (6.72mmol) of 12- (Boc-amino) dodecanoic acid, 1.61g (8.4mmol) of EDCI and 0.068g (0.56mmol) of DMAP were sequentially weighed and placed in a 100mL single-necked flask, and 50mL of anhydrous dichloromethane was added to dissolve them, and the reaction was stirred at room temperature for 12 hours, TLC [ V (dichloromethane): V (methanol): 20: 1)]The reaction was monitored for completion, and the reaction solution was diluted with dichloromethane to 150ml, washed three times with 50ml of distilled water each time, then dehydrated with saturated brine, the organic layer was collected, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0 to 30/1) to give 2.42g of CPT-A-L as a pale yellow solid12。mp 134.6-140.0℃;Yield:67.0%;1H-NMR(500MHz,CDCl3):δ0.96(t,J=7.4Hz,3H,-CH3),1.15(m,8H),1.26(m,6H),1.42(s,9H,3×-CH3),1.62(m,2H),2.16(m,1H),2.27(m,1H),2.47(m,2H),3.06(s,2H),5.26(m,2H),5.40(d,J=17.1Hz,1H),5.66(d,J=17.1Hz,1H),7.22(s,1H),7.66(t,J=7.3Hz,1H),7.82(t,J=7.3Hz,1H),7.93(d,J=8.1Hz,1H),8.20(d,J=8.5Hz,1H),8.39(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,24.7,26.9,28.5,29.1,29.3,29.5,29.5,30.1,32.0,33.9,40.7,50.0,67.2,75.7,79.1,96.2,120.5,128.2,128.3,128.3,128.6,129.6,130.8,131.4,146.1,146.2,148.9,152.4,156.1,157.5,167.7,172.9.HRMS(ESI)m/z:[M+H]+646.3484,calcd.for C37H47N3O7 645.34140。
Practice ofEXAMPLE 23 Compound CPT-B-L12Preparation of
2.2g (3.41mmol) of CPT-A-L were taken12Placing in a 100mL single-neck flask, adding 30mL of anhydrous dichloromethane to dissolve, adding 9mL of TFA under ice bath, and stirring at room temperature for 1 hr, TLC [ V (dichloromethane): V (methanol): 10:1]Monitoring the reaction completion, spin-drying the reaction solution at low temperature by a diaphragm pump, mixing the sample with silica gel, and performing Flash silica gel column chromatography (DCM/MeOH: 20/1-8/1) to obtain 1.34g of CPT-B-L as a pale yellow solid12。mp 215.5-216.0℃;Yield:72.0%;1H-NMR(500MHz,CD3OD):δ1.04(t,J=7.4Hz,3H,-CH3),1.23(m,8H),1.33(m,6H),1.63(m,4H),2.23(m,2H),2.54(m,2H),2.89(m,2H),5.27(s,2H),5.47(d,J=16.8Hz,1H),5.60(d,J=16.8Hz,1H),7.35(s,1H),7.69(dd,J=8.0,7.1Hz,1H),7.86(t,J=7.7Hz,1H),8.04(d,J=8.2Hz,1H),8.14(d,J=8.5Hz,1H),8.58(s,1H).13C-NMR(126MHz,CD3OD):δ8.0,25.8,27.4,28.6,30.0,30.1,30.3,30.4,30.5,32.2,34.6,40.8,51.5,67.7,77.3,97.9,120.7,129.2,129.7,129.8,130.0,130.8,131.9,133.3,147.5,148.4,149.6,153.4,159.0,169.6,174.1.HRMS(ESI)m/z:[M+H]+546.2972,calcd.for C32H39N3O5 545.28897。
EXAMPLE 24 Compound CPT-HT-J-L12Preparation of
618.0mg (0.60mmol) of HT-J, 159.0mg (0.83mmol) of EDCI and 112.1mg (0.83mmol) of HOBT were weighed out in sequence and placed in a 50mL single-neck flask, 20mL of anhydrous dichloromethane were added and dissolved, the reaction was stirred at room temperature for 0.5h, and 300mg (0.55mmol) of CPT-B-L was added12And 178.0mg (1.38mmol) of DIPEA, stirred at room temperature for 4 hours, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted to 50ml with dichloromethane, washed three times with 25ml of distilled water each time, then dehydrated with saturated brine, collected as an organic layer, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, stirred on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH ═ 1/0 to 50/1) to give 360mg of CPT-HT-J-L as a pale yellow solid12。mp 121.5-121.9℃;Yield:42.0%;1H-NMR(500MHz,CDCl3):δ0.96(t,J=7.4Hz,3H,-CH3),1.14(m,10H),1.24(s,4H),1.30(m,4H),1.41(s,27H,9×-CH3),1.42(s,15H,5×-CH3),1.44(s,12H,4×-CH3),1.62(m,4H),1.86(m,2H),2.07(m,4H),2.26(m,6H),2.37(m,4H),2.48(m,4H),2.60(m,1H),2.87(m,1H),3.10(m,1H),3.22(m,1H),4.13(s,1H),4.42(m,2H),4.52(m,1H),4.80(s,1H),5.28(m,2H),5.40(d,J=17.0Hz,1H),5.66(d,J=17.0Hz,1H),5.87(d,J=7.1Hz,1H),6.31(d,J=8.1Hz,1H),7.21(s,1H),7.28(d,J=7.5Hz,1H),7.37(t,J=8.6Hz,1H),7.44(s,1H),7.66(t,J=7.4Hz,1H),7.82(t,J=7.6Hz,1H),7.94(d,J=8.1Hz,1H),8.20(d,J=8.5Hz,1H),8.40(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,24.8,27.0,27.6,27.8,28.1,28.2,28.5,29.0,29.2,29.4,29.5,29.6,29.6,31.5,32.0,32.1,32.5,33.9,37.4,39.8,49.3,50.1,51.7,52.2,52.5,53.3,67.2,75.7,80.0,80.6,81.6,81.9,82.2,82.5,96.2,120.5,128.2,128.3,128.4,128.6,129.7,130.8,131.4,146.1,146.3,149.0,152.5,156.3,157.5,167.7,170.9,171.1,171.5,171.7,172.2,172.5,172.9,172.9.HRMS(ESI)m/z:[M+H]+1557.8584,calcd.for C81H120N8O221556.85172。
EXAMPLE 25 Compound CPT-HT-J-ZL12Preparation of
Weighing 320mg (0.206mmol) of CPT-HT-J-L12Put into a 50mL single-neck flask, 10mL of hydrolysate (TFA: Thioanisole: H) was added2O is 95%: 2.5%: 2.5 percent) of the total saponin, stirring and reacting for 3 hours at room temperature, evaporating the reaction solution at low temperature under reduced pressure, adding 10ml of ethyl acetate for precipitation, centrifuging and collecting the precipitate, continuously adding 10ml of ethyl acetate for precipitation, centrifuging and collecting the precipitate to obtain CPT-HT-J-ZL12Dissolving the crude product in methanol, separating liquid phase, and freeze drying to obtain 60mg of CPT-HT-J-ZL12。mp 183.9-184.3℃;Yield:24.8%;1H-NMR(500MHz,DMSO):δ0.93(t,J=7.4Hz,3H,-CH3),1.12(m,10H),1.26(m,5H),1.37(m,4H),1.45(m,1H),1.54(m,3H),1.80(m,2H),1.93(m,4H),2.22(m,7H),2.35(m,2H),3.10(m,2H),3.30(m,1H),3.40(m,1H),3.59(s,1H),3.62(s,1H),3.72(s,1H),4.17(m,3H),4.57(m,1H),5.31(s,2H),5.50(m,2H),7.05(s,1H),7.73(t,J=7.5Hz,1H),7.87(t,J=7.5Hz,1H),8.15(m,6H),8.32(m,2H),8.71(s,1H).13C-NMR(126MHz,DMSO):δ7.6,24.0,24.5,25.3,26.0,26.4,26.7,27.0,27.6,28.4,28.7,28.8,28.9,29.7,30.3,30.4,33.2,38.8,42.0,45.8,50.3,51.4,51.6,52.0,66.3,75.6,94.7,119.0,127.8,128.1,128.6,129.0,129.8,130.4,131.6,145.4,146.0,147.9,152.3,156.6,163.1,167.3,167.9,169.3,171.5,171.7,172.1,172.9,172.8,173.4,173.6,173.8.HRMS(ESI)m/z:[M+H]+1177.4963,calcd.for C81H120N8O22 1176.48629。
EXAMPLE 26 Compound CPT-C-GL2Preparation of
222.6mg (0.60mmol) of N-Cbz-L-glutamic acid-5-benzyl ester, 59.0mg (0.83mmol) of EDCI and 112.0mg (0.83mmol) of HOBT were weighed in this order and placed in a 100mL single-neck flask, and 20mL of anhydrous dichloromethane was added thereto and dissolved, and the reaction was stirred at room temperature for 10min, and 223mg (0.55mmol) of CPT-B-L was further added2And 178mg (1.38mmol) of DIPEA, stirred at room temperature for 12h, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted with dichloromethane to 100ml, washed with 50ml of distilled water three times each time, then dehydrated with saturated brine, collected as an organic layer, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH ═ 1/0 to 30/1) to give 210mg of CPT-C-GL as a pale yellow solid2. The yield thereof was found to be 50.4%. mp 213.9-214.2 ℃; 50.4 percent of Yield;1H-NMR(500MHz,CDCl3):δ0.97(t,J=7.4Hz,3H,-CH3),1.97(dd,J=14.3,7.4Hz,1H),2.14(m,2H),2.26(dd,J=14.3,7.4Hz,1H),2.48(m,2H),4.12(m,1H),4.31(m,1H),4.37(dd,J=18.2,6.1Hz,1H),4.93(t,J=14.5Hz,2H),5.02(t,J=14.5Hz,2H),5.13(d,J=18.9Hz,1H),5.22(d,J=18.9Hz,1H),5.37(d,J=17.1Hz,1H),5.66(d,J=17.1Hz,1H),6.99(s,1H),7.18(m,2H),7.24(m,2H),7.28(m,6H),7.63(t,J=7.5Hz,1H),7.78(t,J=7.6Hz,1H),7.89(d,J=8.2Hz,1H),8.22(d,J=8.5Hz,1H),8.30(d,J=8.5Hz,1H).13C-NMR(126MHz,CDCl3):7.7,28.1,30.5,31.8,41.4,50.1,54.1,66.7,67.1,67.2,96.4,120.2,128.1,128.2,128.3,128.4,128.6,128.7,129.7,130.8,131.4,135.7,136.1,145.6,146.4,148.8,152.3,156.4,157.4,167.2,168.9,171.7,173.2.HRMS(ESI)m/z:[M+H]+759.2653,calcd.for C42H38N4O10 758.25879。
EXAMPLE 27 Compound CPT-D-GL2Preparation of
Weighing 180mg (0.237mmol) of CPT-C-GL2Placing into a 100mL single-neck bottle, adding 50mL anhydrous methanol to dissolve, adding appropriate amount of 5% Pd/C, stirring under hydrogen protection at room temperature for 12h, and reacting by TLC [ V (dichloromethane): V (methanol): 20:1]Monitoring reaction completion, filtering reaction solution, evaporating filtrate under reduced pressure, mixing with silica gel, and performing Flash silica gel column chromatography (DCM/MeOH: 30/1-10/1) to obtain 50mg of CPT-D-GL as a pale yellow solid2。Yield:39.5%;1H-NMR(500MHz,DMSO):δ0.92(s,3H),2.16(d,J=6.3Hz,2H),2.42(dd,J=16.1,7.8Hz,1H),2.82(d,J=16.1Hz,1H),3.47(s,1H),4.08(d,J=17.6Hz,1H),4.19(d,J=16.9Hz,1H),5.27(s,2H),5.50(s,2H),7.19(s,1H),7.71(t,J=6.5Hz,1H),7.85(d,J=6.7Hz,1H),8.11(d,J=7.5Hz,1H),8.20(d,J=8.1Hz,1H),8.67(s,1H),9.02(s,1H).13C-NMR(126MHz,DMSO):δ8.0,28.0,30.8,36.4,40.9,50.7,51.2,66.8,76.7,95.9,119.4,128.2,128.4,129.0,129.5,130.2,130.9,132.1,145.6,146.4,148.4,152.8,157.0,167.6,169.4,171.7.HRMS(ESI)m/z:[M+H]+535.1800,calcd.for C27H26N4O8534.17506。
EXAMPLE 28 Compound CPT-C-GL4Preparation of
222.6mg (0.60mmol) of N-Cbz-L-glutamic acid-5-benzyl ester, 59.0mg (0.83mmol) of EDCI and 112.0mg (0.83mmol) of HOBT were weighed in this order and placed in a 100mL single-neck flask, and 20mL of anhydrous dichloromethane was added thereto and dissolved, and the reaction was stirred at room temperature for 10min, followed by addition of 238mg (0.55mmol) of CPT-B-L4And 178mg (1.38mmol) of DIPEA, stirred at room temperature for 12h, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted with dichloromethane to 100ml, washed with 50ml of distilled water three times each time, then dehydrated with saturated brine, collected as an organic layer, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0 to 30/1) to give 260mg of CPT-C-GL as a pale yellow solid4。mp 118.0-118.7℃;Yield:60.0%;1H-NMR(500MHz,CDCl3):δ0.96(t,J=7.4Hz,3H,-CH3),1.82(m,2H),1.93(m,1H),2.13(m,2H),2.25(m,1H),2.47(m,4H),3.28(m,2H),4.22(d,J=4.8Hz,1H),5.04(s,2H),5.09(s,2H),5.22(d,J=18.9Hz,1H),5.27(d,J=18.9Hz,1H),5.38(d,J=17.1Hz,1H),5.68(d,J=17.1Hz,1H),5.76(d,J=7.5Hz,1H),6.58(s,1H),7.23(s,1H),7.30(m,10H),7.65(d,J=7.5Hz,1H),7.81(dd,J=11.3,4.1Hz,1H),7.92(d,J=8.1Hz,1H),8.22(d,J=8.5Hz,1H),8.36(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,24.2,28.2,30.6,31.2,31.9,38.7,50.1,54.4,66.6,67.1,67.3,76.2,96.2,120.2,128.1,128.2,128.3,128.4,128.6,128.7,129.6,130.9,131.5,135.8,136.3,146.0,146.3,148.8,152.4,156.4,157.4,168.0,171.5,172.3,173.2.HRMS(ESI)m/z:[2M+H]+1573.7668,calcd.for C44H42N4O10 786.29009。
EXAMPLE 29 Compound CPT-D-GL4Preparation of
Weighing 230mg (0.293mmol) of CPT-C-GL4Placing into a 100mL single-neck bottle, adding 50mL anhydrous methanol to dissolve, adding appropriate amount of 5% Pd/C, stirring under hydrogen protection at room temperature for 12h, and reacting by TLC [ V (dichloromethane): V (methanol): 20:1]Monitoring reaction completion, filtering reaction liquid, decompressing and evaporating filtrate, mixing silica gel, and performing Flash silica gel column chromatography (DCM/MeOH is 30/1-10/1) to obtain 70mg of light yellow solid CPT-D-GL4。mp 127.5-128.1℃;Yield:42.6%;1H-NMR(500MHz,CDCl3):δ0.93(t,J=7.4Hz,3H,-CH3),1.69(m,2H),1.74(m,1H),1.88(m,1H),2.16(m,2H),2.23(t,J=7.7Hz,2H),2.24(m,2H),3.12(d,J=7.1Hz,2H),3.33(s,1H),3.95(dd,J=13.9,8.3Hz,1H),5.02(s,1H),5.03(s,1H),5.31(m,2H),5.48(d,J=16.8Hz,1H),5.52(d,J=16.8Hz,1H),7.09(s,1H),7.31(dd,J=8.5,4.2Hz,1H),7.36(s,1H),7.73(t,J=7.1Hz,1H),7.87(t,J=7.1Hz,1H),7.98(t,J=5.6Hz,1H),8.16(dd,J=18.4,8.2Hz,2H),8.70(s,1H).13C-NMR(126MHz,DMSO):δ7.6,24.4,27.2,30.2,30.3,30.7,37.8,50.3,54.1,65.4,75.8,94.8,118.9,127.7,128.0,128.3,128.6,129.0,129.8,130.4,131.6,145.4,146.1,147.9,152.4,156.6,167.3,171.4,171.9,173.9.HRMS(ESI)m/z:[M+H]+563.2149,calcd.for C29H30N4O8 562.20636。
EXAMPLE 30 Compound CPT-C-GL6Preparation of
222.6mg (0.60mmol) of N-Cbz-L-glutamic acid-5-benzyl ester, 59.0mg (0.83mmol) of EDCI and 112.0mg (0.83mmol) of HOBT were weighed in this order and placed in a 100mL single-neck flask, and 20mL of anhydrous dichloromethane was added thereto and dissolved, and the reaction was stirred at room temperature for 10min, followed by addition of 254mg (0.55mmol) of CPT-B-L6And 178mg (1.38mmol) of DIPEA, stirred at room temperature for 12h, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted with dichloromethane to 100ml, washed with 50ml of distilled water three times each time, then dehydrated with saturated brine, collected as an organic layer, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0 to 30/1) to give 280mg of CPT-C-GL as a pale yellow solid6。mp 106.3-107.3℃;Yield:62.6%;1H-NMR(400MHz,CDCl3):δ0.90(t,J=7.4Hz,3H,-CH3),1.27(m,2H),1.42(m,2H),1.50(m,2H),1.83(m,2H),2.14(m,6H),3.04(m,2H),3.63(s,1H),3.67(s,1H),4.19(m,1H),4.33(m,2H),5.30(d,J=17.5Hz,1H),5.49(d,J=17.5Hz,1H),7.32(dd,J=17.0,9.6Hz,1H),7.48(d,J=7.5Hz,1H),7.71(m,1H),7.88(dd,J=17.0,9.6Hz,1H),8.15(m,1H),8.69(s,1H).13C-NMR(101MHz,CDCl3):7.7,24.3,25.9,27.8,28.9,30.6,31.9,33.7,39.2,50.1,52.7,54.3,66.6,67.1,67.3,75.9,96.3,120.4,128.1,128.3,128.4,128.7,129.4,131.0,131.6,135.9,136.4,146.1,146.2,148.7,152.4,157.4,168.0,171.1,172.7,173.2.HRMS(ESI)m/z:[M+H]+815.3279,calcd.for C46H46N4O10814.32139。
EXAMPLE 31 Compound CPT-D-GL6Preparation of
Weighing 250mg (0.307mmol) of CPT-C-GL6Placing into a 100mL single-neck bottle, adding 50mL anhydrous methanol to dissolve, adding appropriate amount of 5% Pd/C, stirring under hydrogen protection at room temperature for 12h, and reacting by TLC [ V (dichloromethane): V (methanol): 20:1]Monitoring reaction completion, filtering reaction liquid, decompressing and evaporating filtrate, mixing silica gel, and performing Flash silica gel column chromatography (DCM/MeOH is 30/1-10/1) to obtain 65mg of light yellow solid CPT-D-GL6。Yield:35.9%;1H-NMR(500MHz,CDCl3):δ0.93(t,J=7.4Hz,3H,-CH3),1.69(m,2H),1.74(m,1H),1.88(m,1H),2.16(m,2H),2.23(t,J=7.7Hz,2H),2.24(m,2H),3.12(d,J=7.1Hz,2H),3.33(s,1H),3.95(dd,J=13.9,8.3Hz,1H),5.02(s,1H),5.03(s,1H),5.31(m,2H),5.48(d,J=16.8Hz,1H),5.52(d,J=16.8Hz,1H),7.09(s,1H),7.31(dd,J=8.5,4.2Hz,1H),7.36(s,1H),7.73(t,J=7.1Hz,1H),7.87(t,J=7.1Hz,1H),7.98(t,J=5.6Hz,1H),8.16(dd,J=18.4,8.2Hz,2H),8.70(s,1H).13C-NMR(101MHz,DMSO):7.6,24.5,25.4,25.6,28.7,29.3,30.3,33.1,38.3,53.1,54.7,55.9,65.5,75.7,94.8,118.9,127.3,127.8,128.1,129.4,129.9,130.5,131.7,145.5,146.0,147.9,152.3,156.6,167.3,172.1,173.4,177.1,177.4.HRMS(ESI)m/z:[M+H]+591.2467,calcd.for C31H34N4O8 590.23766。
EXAMPLE 32 Compound CPT-C-GL12Preparation of
222.6mg (0.60mmol) of N-Cbz-L-glutamic acid-5-benzyl ester, 59.0mg (0.83mmol) of EDCI and 112.0mg (0.83mmol) of HOBT were weighed in this order and placed in a 100mL single-neck flask, and 20mL of anhydrous dichloromethane was added thereto and dissolved, and the reaction was stirred at room temperature for 10min, and 300mg (0.55mmol) of CPT-B-L was further added12And 178mg (1.38mmol) of DIPEA, stirred at room temperature for 12h, TLC [ V (dichloromethane): V (methanol): 20:1]The reaction was monitored for completion, and the reaction solution was diluted with dichloromethane to 100ml, washed with 50ml of distilled water three times each time, then dehydrated with saturated brine, collected as an organic layer, dried over an appropriate amount of anhydrous sodium sulfate, filtered, evaporated to dryness under reduced pressure, sampled on silica gel, and subjected to Flash silica gel column chromatography (DCM/MeOH. RTM. 1/0 to 30/1) to give 240mg of CPT-C-GL as a pale yellow solid12。mp 90.7-91.5℃;Yield:48.6%;1H-NMR(500MHz,CDCl3):δ0.97(t,J=7.5Hz,3H,-CH3),1.16(s,10H),1.25(s,2H),1.30(m,2H),1.40(m,2H),1.63(m,2H),1.95(m,1H),2.14(m,1H),2.28(m,1H),2.46(m,4H),3.17(m,2H),4.20(m,1H),5.07(m,2H),5.11(m,2H),5.24(d,J=19.0Hz,1H),5.29(d,J=19.0Hz,1H),5.40(d,J=17.0Hz,1H),5.66(d,J=17.0Hz,1H),5.71(s,1H),6.24(s,1H),7.23(s,1H),7.32(s,6H),7.32(s,4H),7.66(t,J=7.4Hz,1H),7.82(t,J=7.4Hz,1H),7.93(d,J=8.1Hz,1H),8.21(d,J=8.5Hz,1H),8.38(s,1H).13C-NMR(126MHz,CDCl3):δ7.7,24.7,26.9,28.4,29.1,29.3,29.3,29.4,29.5,29.5,30.5,32.0,33.9,39.7,50.0,54.3,66.7,67.1,67.2,75.7,96.2,120.5,128.1,128.2,128.3,128.3,128.4,128.6,128.6,128.7,129.6,130.8,131.4,135.8,136.3,146.1,146.2,148.9,152.4,156.4,157.5,167.7,171.0,172.9,173.3.HRMS(ESI)m/z:[M+H]+899.4223,calcd.for C52H58N4O10 898.41529。
EXAMPLE 33 Compound CPT-D-GL12Preparation of
Weighing 220mg (0.245mmol) of CPT-C-GL12Placing into a 100mL single-neck bottle, adding 50mL anhydrous methanol to dissolve, adding appropriate amount of 5% Pd/C, stirring under hydrogen protection at room temperature for 12h, and reacting by TLC [ V (dichloromethane): V (methanol): 20:1]Monitoring reaction completion, filtering reaction solution, evaporating filtrate under reduced pressure, mixing with silica gel, and performing Flash silica gel column chromatography (DCM/MeOH: 30/1-10/1) to obtain 40mg of CPT-D-GL as a pale yellow solid12。mp>220℃;Yield:24.2%;1H-NMR(500MHz,DMSO):δ0.91(t,J=7.4Hz,3H,-CH3),1.26(m,16H),1.54(m,2H),1.65(m,2H),1.78(m,2H),2.13(m,4H),2.24(m,2H),3.03(m,2H),3.31(s,1H),5.26(d,2H),5.44(m,1H),5.49(s,1H),6.82(s,1H),7.04(s,1H),7.31(dd,J=14.7,7.1Hz,1H),7.46(d,J=7.1Hz,1H),7.71(t,J=7.5Hz,1H),7.86(t,J=7.6Hz,1H),8.04(s,1H),8.13(m,2H),8.69(s,1H).13C-NMR(126MHz,DMSO):δ7.5,24.5,26.3,26.4,28.4,28.7,28.7,28.8 28.9,29.0,29.4,30.3,32.3,33.2,38.4,39.0,39.2,39.3,39.5,39.7,39.8,40.0,50.2,53.6,66.3,75.6,94.7,119.0,127.7,128.0,128.6,128.9,129.8,130.4,131.6,145.4,146.0,147.9,152.3,156.6,165.1,167.3,172.1,172.4,174.7.HRMS(ESI)m/z:[M+H]+675.3390,calcd.for C37H46N4O8 674.33156。
Example 34
Taking 10g of the compound prepared in any one of the embodiments 9-33, adding appropriate adjuvants of injection (including lyophilized powder for injection and sterile packaged dry powder for injection), and preparing into antitumor drug injection according to the injection (including lyophilized powder for injection and sterile packaged dry powder for injection) process.
Example 35
Taking 10g of the compound prepared in any one of the embodiments 9-33, adding appropriate auxiliary materials into tablets (including sustained release tablets, matrix tablets, coated tablets, dispersible tablets and the like), and preparing the compound into the anti-tumor tablets according to the process of the tablets (including sustained release tablets, matrix tablets, coated tablets, dispersible tablets and the like).
Example 36
Taking 10g of the compound prepared in any one of the embodiments 9-33, adding appropriate capsule auxiliary materials, and preparing the compound into an anti-tumor capsule according to a capsule process.
Example 37
Taking 10g of the compound prepared in any one of the embodiments 9-33, adding appropriate auxiliary materials of an emulsion (including microemulsion, nanoemulsion and the like), and preparing the anti-tumor emulsion according to the process of the emulsion (including microemulsion, nanoemulsion and the like).
Example 38
Taking 10g of the compound prepared in any one of the embodiments 9-33, adding proper auxiliary materials of granules, and preparing the antitumor granules according to a granule process.
Example 39
Taking 10g of the compound prepared in any one of the embodiments 9-33, adding a proper auxiliary material of the sustained-release controlled-release agent, and preparing the sustained-release controlled-release agent of the antitumor drug according to the sustained-release controlled-release agent process.
Example 40
And (3) taking 10g of the compound prepared in any one of the embodiments 9-33, adding proper auxiliary materials of the oral liquid, and preparing the anti-tumor oral liquid according to an oral liquid process.
EXAMPLE 41
Taking 10g of the compound prepared in any one of the embodiments 9-33, adding proper auxiliary materials of a liposome formulation, and preparing the antitumor liposome formulation according to a liposome process.

Claims (9)

1. A compound with anti-tumor effect and shown in a general formula 1,
Figure FDA0003143004580000011
n is 1,3, 5, 11.
2. Compound CPT-HT-J-ZL with anti-tumor effect12
Figure FDA0003143004580000012
3. The compound or the pharmaceutically acceptable salt thereof according to claim 2, wherein the compound or the pharmaceutically acceptable salt thereof is added with conventional auxiliary materials in the field of preparation to prepare tablets, capsules, granules, powder, oral liquid and injections.
4. A process for the preparation of a compound according to claim 2, which process comprises:
the method comprises the following steps:
step 1, camptothecin is dissolved in an organic solvent, and CPT-A-L is generated with 12- (Boc-amino) dodecanoic acid under the action of a condensing agent and a catalyst12
Figure FDA0003143004580000013
Step 2, Compound CPT-B-L12The preparation method comprises the following steps: mixing CPT-A-L12Dissolving in organic solvent, and generating CPT-B-L under the action of TFA12
Figure FDA0003143004580000021
Step 3, Compound CPT-HT-J-L12The preparation method comprises the following steps: mixing CPT-B-L12Dissolving in organic solvent, and reacting with HT-J under the action of condensing agent and catalyst to obtain CPT-HT-J-L12
Figure FDA0003143004580000022
Step 4, Compound CPT-HT-J-ZL12The preparation method comprises the following steps: mixing CPT-HT-J-L12Dissolving the mixture in an organic solvent to obtain a solution,in TFA Thioaisole H2CPT-HT-J-ZL is generated under the action of 95 percent of O, 2.5 percent of O and 2.5 percent of O12
5. The method of claim 4, wherein the reaction is carried out at-20 ℃ to 250 ℃; wherein the organic solvent is ether, alcohol, alkane, aromatic hydrocarbon, ketone, alkyl halide, amide, nitrile, ester or their mixture containing 1-20 carbon atoms; the catalyst is 4-Dimethylaminopyridine (DMAP); the condensing agent is 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI), 1, 3-Dicyclohexylcarbodiimide (DCC), N' -Diisopropylcarbodiimide (DIC) and 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDC).
6. Use of a compound according to any one of claims 1 and 2, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of cancer selected from the group consisting of liver cancer, prostate cancer, breast cancer and cervical cancer.
7. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claims 1 and 2, or a pharmaceutically acceptable salt thereof, in admixture with at least one pharmaceutically acceptable excipient.
8. The composition of claim 7, further comprising at least one conventional anticancer agent.
9. The composition of claim 8, wherein the anticancer agent is selected from the group consisting of cyclophosphamide, 5-fluorouracil, paclitaxel, doxorubicin, etoposide, irinotecan, oxaliplatin, cisplatin, and gemcitabine.
CN201811091945.3A 2018-09-19 2018-09-19 PSMA activated antitumor prodrug CPT-X and preparation method and application thereof Active CN110922450B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811091945.3A CN110922450B (en) 2018-09-19 2018-09-19 PSMA activated antitumor prodrug CPT-X and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811091945.3A CN110922450B (en) 2018-09-19 2018-09-19 PSMA activated antitumor prodrug CPT-X and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN110922450A CN110922450A (en) 2020-03-27
CN110922450B true CN110922450B (en) 2021-11-02

Family

ID=69855018

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811091945.3A Active CN110922450B (en) 2018-09-19 2018-09-19 PSMA activated antitumor prodrug CPT-X and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN110922450B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111057127A (en) * 2018-10-15 2020-04-24 薪火炙药(北京)科技有限公司 PSMA activated antitumor prodrug CPT-X and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104873982A (en) * 2007-08-17 2015-09-02 普渡研究基金会 PSMA binding ligand-linker conjugates and methods for using

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104873982A (en) * 2007-08-17 2015-09-02 普渡研究基金会 PSMA binding ligand-linker conjugates and methods for using

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Synthesis and Biological Analysis of Prostate-Specific Membrane Antigen-Targeted Anticancer Prodrugs;Sumith A. Kularatne, et al;《JOURNAL OF MEDICINAL CHEMISTRY》;20101111;第53卷(第21期);第7767-7777页 *

Also Published As

Publication number Publication date
CN110922450A (en) 2020-03-27

Similar Documents

Publication Publication Date Title
CN112592331B (en) Oseltamivir PROTAC compound, preparation method thereof and application thereof in anti-influenza virus drugs
WO2022028421A1 (en) Fluorescent traceable amino acid derivative, preparation method therefor, and application thereof
CN107417752A (en) One kind has compound of active anticancer and its preparation method and application
WO2015096640A1 (en) Thiazolyl-containing rapamycin derivative and application thereof
CN108350023B (en) Compound with anticancer effect and preparation method and application thereof
CN110964078B (en) Hederagenin compound H-X with anti-lung cancer effect and preparation method and application thereof
CN110922450B (en) PSMA activated antitumor prodrug CPT-X and preparation method and application thereof
CN114380864A (en) Dihydroartemisinin derivative, preparation method, pharmaceutical composition and application of dihydroartemisinin derivative in preparation of antitumor drugs
CN111253411B (en) Berberine linoleic acid conjugate and preparation method and application thereof
CN111471080B (en) ocotillol type ginsengenin A-ring amino thiazole ring derivative and preparation method thereof
WO2018019188A1 (en) Polymorph of nucleoside phosphoramidate prodrug and preparation method therefor
CN108727208A (en) A kind of connexon compound, polyethylene glycol-connexon conjugate and its derivative and polyethylene glycol-connexon-drug conjugates
CN110577526B (en) Salt of bromodomain structural protein inhibitor and preparation method and application thereof
CN108137644B (en) Compound with anti-tumor effect and preparation method and application thereof
CN107739381B (en) Curcumenol derivative and application thereof in preparation of antitumor drugs
CN111057127A (en) PSMA activated antitumor prodrug CPT-X and preparation method and application thereof
CN101928323B (en) 23-hydroxy betulinic acid derivative with modified sites 3, 23 and 28 as well as preparation method, preparation and application thereof
CN110981865B (en) Medicine for treating brain glioma and preparation method thereof
CN110857295B (en) Flavone-ligustrazine compound CH-X with selective anti-liver cancer effect and preparation method and application thereof
CN109836477B (en) Phenylalanine derivative containing benzothiadiazine-3-ketone 1, 1-dioxide and preparation method and application thereof
CN109867709B (en) Preparation method and application of glycyrrhetinic acid series derivatives (TOGA-X) with anti-tumor effect
CN102267952B (en) Quinazoline compound and preparation method and application thereof
CN110563739A (en) Podophyllotoxin compound P-X with selective anti-lung cancer effect and preparation method and application thereof
CN105732458B (en) Pyrrole amides class compound and preparation method thereof and purposes
CN112694507B (en) Tetrahydro anthraquinone glycoside compound and application thereof in preparation of antitumor drugs

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
GR01 Patent grant
GR01 Patent grant