CN114105751A - Terpenoid and preparation method and application thereof - Google Patents

Terpenoid and preparation method and application thereof Download PDF

Info

Publication number
CN114105751A
CN114105751A CN202210084250.2A CN202210084250A CN114105751A CN 114105751 A CN114105751 A CN 114105751A CN 202210084250 A CN202210084250 A CN 202210084250A CN 114105751 A CN114105751 A CN 114105751A
Authority
CN
China
Prior art keywords
silica gel
compound
terpenoid
preparation
ethyl acetate
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.)
Granted
Application number
CN202210084250.2A
Other languages
Chinese (zh)
Other versions
CN114105751B (en
Inventor
冯育林
何明珍
李志强
杨世林
李军茂
欧阳辉
张皓男
崔玉顺
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.)
Jiangxi University of Traditional Chinese Medicine
Jiangxi Bencao Tiangong Technology Co Ltd
Original Assignee
Jiangxi University of Traditional Chinese Medicine
Jiangxi Bencao Tiangong 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 Jiangxi University of Traditional Chinese Medicine, Jiangxi Bencao Tiangong Technology Co Ltd filed Critical Jiangxi University of Traditional Chinese Medicine
Priority to CN202210084250.2A priority Critical patent/CN114105751B/en
Publication of CN114105751A publication Critical patent/CN114105751A/en
Application granted granted Critical
Publication of CN114105751B publication Critical patent/CN114105751B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/703Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups
    • C07C49/723Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups polycyclic
    • C07C49/727Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups polycyclic a keto group being part of a condensed ring system
    • C07C49/733Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups polycyclic a keto group being part of a condensed ring system having two rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/78Separation; Purification; Stabilisation; Use of additives
    • C07C45/79Separation; Purification; Stabilisation; Use of additives by solid-liquid treatment; by chemisorption
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/78Separation; Purification; Stabilisation; Use of additives
    • C07C45/81Separation; Purification; Stabilisation; Use of additives by change in the physical state, e.g. crystallisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/14All rings being cycloaliphatic
    • C07C2602/26All rings being cycloaliphatic the ring system containing ten carbon atoms
    • C07C2602/28Hydrogenated naphthalenes

Abstract

The invention belongs to the technical field of medicines, and particularly discloses a terpenoid, a preparation method and application thereof. The terpenoid can obviously inhibit the growth of 10 cancer cells such as liver cancer cells (HepG 2), breast cancer cells (MCF-7), cervical cancer cells (HeLa) and the like, has obvious anticancer effect, and is expected to be developed into a new anticancer medicament.

Description

Terpenoid and preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a terpenoid and a preparation method and application thereof, in particular to application in preparation of anti-cancer drugs.
Background
Cancer is a malignant disease seriously threatening human health, and has the characteristics of unlimited proliferation, transformation and easy dispersion and transfer, and the clinical manifestations of cancer are high recurrence rate and death rate, which are now a medical problem all over the world. According to the latest cancer burden data (provided by IARC) in the world of 2020, 1929 ten thousands of new cancer cases and 996 ten thousands of cancer death cases in the world of 2020, wherein 457 thousands of new cancers in China account for 23.7% of the world and far surpass other countries in the world. Worldwide, the burden of cancer presents an increasing trend due to the aggravation of aging problems of the population, smoking, unhealthy diet, and lack of exercise, and effective cancer prevention and therapeutic intervention is urgently required.
Since the discovery and application of cisplatin to tumor therapy by Rosenberg et al in the 60's last century, platinum drugs such as carboplatin and oxaliplatin were introduced in succession, and significant progress was made in the clinical therapy of tumors. With the widespread use of platinum-based antineoplastic drugs, some of their disadvantages, such as poor compliance, myelosuppression, severe neurotoxicity, renal toxicity and acquired drug resistance, have been gradually revealed, which limits their clinical application. At present, a novel efficient and safe antitumor drug is urgently needed clinically.
LilacSyringaoblataLindl is deciduous tree of Syzygium of Oleaceae. The lilac contains rich chemical componentsIs rich in various chemical components such as terpenes, lignans, esters and the like. The invention carries out deep research on two terpenoid chemical components extracted and separated from lilac medicinal materials, and is expected to screen and obtain a new compound or lead compound with anticancer effect.
Disclosure of Invention
The invention aims to deeply research two terpenoid chemical components of syringa oblata stems, and provides terpenoid compounds in syringa oblata and a preparation method and application thereof.
The technical scheme of the invention is as follows:
a terpenoid compound comprises a compound I and a compound II, wherein the chemical names of the compounds are respectively 3- (1, 2-dihydroxy-2-methylethyl) -7-hydroxy-5, 6-dimethyl-2, 3,4,5,9, 10-hexahydronaphthalene-1, 8-dione and 8- (-1, 2-dihydroxyethyl) -2-hydroxy-3, 4, 7-trimethyl-5, 6-dihydronaphthalene-1-ketone, and the structural formula is as follows:
Figure 488805DEST_PATH_IMAGE001
the invention also provides a preparation method of the terpenoid, which specifically comprises the following steps:
(1) taking a lilac stem medicinal material, extracting, filtering, collecting an extracting solution, concentrating under reduced pressure until no alcohol smell exists to obtain an extract concentrated solution, and then sequentially extracting with dichloromethane and ethyl acetate to obtain a dichloromethane extraction part and an ethyl acetate extraction part;
(2) dissolving the ethyl acetate extraction part obtained in the step (1) with methanol, adding silica gel, mixing with dry method, loading to silica gel column, performing column chromatography, gradient eluting with dichloromethane-methanol at volume ratio of 100:0, 90:10, 80:20, 70:30, 50:50, 30:70, 0:100, and eluting with silica gel GF254Inspecting the thin-layer plate, developing with petroleum ether-ethyl acetate (volume ratio of 8:1, 4:1, 2:1, 1:1, and 0: 1), and mixing fractions containing similar spots to obtain 14 fractions 1,2, 3,4,5, 6, 7, 8, 9,10, 11, 12, 13, and 14;
(3) dissolving the fraction 10 with methanol, separating by silica gel column chromatography, gradient eluting with petroleum ether-acetone at volume ratio of 10:1, 8:1, 4:1, 2:1, 1:1, 0:1, and detecting by HPLC with the following conditions: welchrom C18 reverse phase chromatography column, mobile phase: acetonitrile-water solution (volume ratio 30: 70), isocratic elution, full wavelength scan, flow rate: 1mL/min, collecting for 50min, respectively extracting spectra under three different detection wavelengths of 254nm, 280nm and 300nm, and combining fractions with similar chemical compositions under the same detection wavelength to obtain 5 fractions a, b, c, d and e;
(4) dissolving fraction d with methanol, separating by preparative liquid chromatography, eluting with acetonitrile-water at volume ratio of 20:80 as mobile phase, collecting two chromatographic peaks with retention time of 18min and 20min respectively to obtain two crude compounds;
(5) and (4) dissolving the two crude compounds obtained in the step (4) and then recrystallizing to obtain pure compound I and compound II.
Preferably, the extraction method in the step (1) is a cold soaking method, a percolation method, a microwave extraction method, an ultrasonic extraction method, a reflux extraction method or a continuous reflux extraction method, and the solvent used for extraction is 40-95% ethanol water solution, and more preferably 80% ethanol water solution.
Preferably, in the step (1), the dosage of the dichloromethane and the ethyl acetate is 1/5-1/3 of the volume of the concentrated extract liquid each time, and the extraction times are 2-6 times.
Preferably, in the step (2), the silica gel used for sample mixing is 100-200 meshes, the mass ratio of the ethyl acetate extraction part to the sample mixing silica gel is 1 (2-5), and the silica gel used in the silica gel column is 100-200 meshes.
Preferably, in the step (3), the silica gel used in the silica gel column is 200-300 meshes.
Preferably, in the step (4), the chromatographic conditions of the preparative liquid chromatography are as follows: and a YMC-TriartC18 chromatographic column, wherein the size of the chromatographic column is 250 multiplied by 20mm, the particle size is 5 mu m, and the flow rate is 10 mL/min.
The invention also provides application of the terpenoid in preparing anti-cancer drugs.
The invention also provides a pharmaceutical preparation, which comprises terpenoid with effective treatment amount and pharmaceutically acceptable carriers or auxiliary materials.
The terpenoid can be directly or indirectly added into various pharmaceutically acceptable common adjuvants such as filler, disintegrant, lubricant, and binder for preparing different dosage forms, and made into common oral preparation or injection by conventional method.
Preferably, the oral preparation can be tablets, capsules, granules, fat emulsion, microcapsules and dropping pills.
Preferably, the injection preparation can be injection or powder injection.
Has the advantages that: the preparation method of the terpenoid is simple to operate, and a pure compound can be separated and obtained. Experiments show that the compound has obvious anticancer effect and can be used for preparing anticancer drugs.
Drawings
FIG. 1 is a chemical structural formula of a terpenoid of the present invention;
FIG. 2 is a MS diagram of Compound I of the present invention;
FIG. 3 is a MS diagram of Compound II of the present invention;
FIG. 4 shows compounds I according to the invention1H-NMR chart;
FIG. 5 shows compounds II of the present invention1H-NMR chart;
FIG. 6 shows compounds I of the present invention13C-NMR chart;
FIG. 7 shows the preparation of compound II according to the invention13C-NMR chart;
FIG. 8 is a DEPT spectrum of Compound I of the present invention;
FIG. 9 is a DEPT spectrum of Compound II of the present invention;
FIG. 10 is an HSQC spectrum of compound I of the present invention;
FIG. 11 is an HSQC spectrum of compound II of the present invention;
FIG. 12 is an HMBC spectrum of compound I of the present invention;
FIG. 13 is an HMBC spectrum of compound II of the present invention;
FIG. 14 shows compounds I of the present invention1H-1H-COSY spectrum;
FIG. 15 is a drawing showingOf the compounds II of the invention1H-1H-COSY spectrum;
FIG. 16 shows compounds I of the present invention1H-1The structural analysis schematic diagram of H-COSY, HMBC and NOESY maps;
FIG. 17 shows the preparation of compound II of the present invention1H-1Schematic structural analysis diagrams of H-COSY, HMBC and NOESY maps.
Detailed Description
In order to further understand the present invention, the technical solutions in the embodiments of the present invention will be clearly and completely described below with reference to the embodiments of the present invention, and it is obvious that the described embodiments are only a part of the embodiments of the present invention, and not all embodiments. All other embodiments, which can be derived by a person skilled in the art from the embodiments given herein without making any creative effort, shall fall within the protection scope of the present invention.
Unless otherwise specified, the reagents involved in the examples of the present invention are all commercially available products, and all of them are commercially available.
First, preparation method of the compound of the invention
Instrument and material (one):
AVANCE iii 500 type superconducting nuclear magnetic resonance apparatus (Bruker, switzerland); ABCIEX 5600+QTOF LC-MS (AB SCIEX, USA); shimadzu model LC-30 ultra high performance liquid chromatograph (Shimadzu, Japan); model 1260 semi-preparative hplc (equipped with quaternary pump, online degasser, autosampler, autosegregation collector, PDA detector, Agilent, usa); Milli-QACsdemica10 ultra pure water machine (Millipore, USA); analytical balance model BT25S (sartorius technologies ltd). Thin layer chromatography silica gel GF254Column chromatography silica gel (Qingdao ocean chemical plant division); reagents such as methanol, acetonitrile, dichloromethane, petroleum ether, ethyl acetate, acetone, etc. are analytically pure (Shandong Yuwang industries, Ltd.).
Preparation examples of (di) Compounds
(1) Taking 10kg of lilac stem medicinal material, adding 80% ethanol water solution with 8 times of volume, reflux-extracting for 2 times, each time for 1.5 hours, filtering after extraction, combining the extracting solutions, and concentrating under reduced pressure until no alcohol smell exists to obtain an extract concentrated solution; extracting the concentrated solution with 1/3 volume of dichloromethane 5 times, extracting with 1/3 volume of ethyl acetate 5 times, mixing dichloromethane extractive solution and ethyl acetate extractive solution, concentrating under reduced pressure at 40 deg.C, and drying under reduced pressure at 60 deg.C to obtain dichloromethane extraction part and ethyl acetate extraction part;
(2) dissolving the ethyl acetate extraction part obtained in the step (1) with methanol, mixing a 100-200-mesh silica gel (the mass of the silica gel is 2 times that of the ethyl acetate extraction part) by a dry method, loading the sample into a silica gel (the particle size of the silica gel is 100-200 meshes, and the mass of the silica gel is 10 times that of the ethyl acetate extraction part) column, performing column chromatography separation, performing gradient elution by dichloromethane-methanol at the volume ratio of 100:0, 90:10, 80:20, 70:30, 50:50, 30:70 and 0:100, and performing gradient elution by silica gel GF (glass fiber)254Inspecting the thin-layer plate, developing with petroleum ether-ethyl acetate (volume ratio of 8:1, 4:1, 2:1, 1:1, and 0: 1), and mixing fractions containing similar spots to obtain 14 fractions 1,2, 3,4,5, 6, 7, 8, 9,10, 11, 12, 13, and 14;
(3) dissolving the fraction 10 with methanol, performing column chromatography separation on silica gel (200-300 meshes), performing gradient elution by using petroleum ether-acetone in a volume ratio of 10:1, 8:1, 4:1, 2:1, 1:1 and 0:1, and inspecting by using HPLC (high performance liquid chromatography) under the following conditions: welchrom C18 reverse phase chromatography column, mobile phase: acetonitrile-water solution (volume ratio 30: 70), isocratic elution, full wavelength scan, flow rate: 1mL/min, collecting for 50min, respectively extracting spectra under three different detection wavelengths of 254nm, 280nm and 300nm, and combining fractions with similar chemical compositions under the same detection wavelength to obtain 5 fractions a, b, c, d and e;
(4) dissolving fraction d with methanol, and separating by preparative liquid chromatography under the following chromatographic conditions: carrying out elution by using acetonitrile-water with a volume ratio of 20:80 as a mobile phase, and collecting two chromatographic peaks with retention time of 18min and 20min respectively to obtain two crude compounds, wherein the size of the chromatographic column is 250 multiplied by 20mm, the particle size is 5 mu m, and the flow rate is 10 mL/min;
(5) and (4) respectively dissolving the two crude compounds obtained in the step (4) by using acetone, standing at room temperature, filtering after a large amount of white precipitates appear at the bottom of a bottle, and collecting the white precipitates to obtain pure compounds I and II. The purities of the compound I and the compound II are respectively 98.9 percent and 99.3 percent by HPLC.
II, structural analysis and identification of compound
Mainly uses the wave spectrum technology, including ultraviolet, infrared, mass spectrum, nuclear magnetic resonance (1H-NMR、13C-NMR and 2D-NMR), and taking the compound prepared in the above example to analyze the structure, wherein the specific spectrogram is shown in the attached figures 2-15, and the spectral data and the analysis process are as follows:
compound I
(1) White amorphous powder, high resolution mass spectrometry ESI-TOF-MS: 305.1370[ M + Na ]]+
The molecular formula of the compound is C15H22O5The exact molecular weight is 282.1467, and the unsaturation is 5.
Comprehensive analysis1H NMR、13C NMR showed that three methyl groups [ delta C19.9, 25.9 and 9.6 (corresponding to C-13, C-14 and C-15, respectively), delta H1.12 (s, Me-13), delta H1.43 (s, Me-14) and delta H1.83 (s, Me-15) were present in the structure of the compound]Two carbonyl groups [ Delta C210.8 and 191.1 (C-1 and C-8, respectively), four methylene groups [ Delta C41.9, 36.5, 32.0 and 67.3 (corresponding to C-2, C-4, C-9 and C-12, respectively). And determining the planar structure of the compound by combining two-dimensional nuclear magnetic resonance spectrum data such as HMBC, DEPT-135 and HSQC spectrums, wherein the structural formula is shown as the attached drawing 1, and the chemical name is 3- (1, 2-dihydroxy-2-methylethyl) -7-hydroxy-5, 6-dimethyl-2, 3,4,5,9, 10-hexahydronaphthalene-1, 8-diketone.
Process for preparing compounds I1H-1The structure analysis process of H-COSY, HMBC and NOESY is shown in figure 16.
(2) The spectral data for this compound are summarized below:
1H NMR (600 MHz, MeOD) δ 3.49 (d, J = 11 Hz, 1H, H-12a), 3.45 (d, J = 11 Hz, 1H, H-12b), 2.88(dd, J = 2.1, 17.1 Hz, 1H, H-9a), 2.83 (m, 1H, H-10), 2.67 (dd, J = 4.4,17.1 Hz, 1H, H-9b), 2.38 (dd, J = 3.1, 14.1 Hz, 1H, H-4a), 2.35 (m, 2H, H-2), 1.83 (s, 3H, H-15), 1.78 (m, 1H, H-3), 1.68 (t, J = 13.5 Hz,1H, H-4b), 1.18 (s, 3H, H-14), 1.43 (s, 3H, H-14), 1.12 (s, 3H, H-13)。
13C NMR (150 MHz, MeOD) δ210.8 (C-1), 191.1 (C-8), 144.8 (C-7), 131.6 (C-6), 73.0 (C-11), 67.3 (C-12), 53.9 (C-10), 41.9 (C-2), 42.4 (C-5), 40.2 (C-3), 36.5 (C-4), 32.0 (C-9), 25.9(C-14), 19.9 (C-13), 9.6 (C-15)。
(di) Compound II
(1) White amorphous powder, high resolution mass spectrometry ESI-TOF-MS: 287.1266[ M + Na ]]+
The molecular formula of the compound is C15H20O4The exact molecular weight is 264.1362, and the unsaturation is 6.
Comprehensive analysis1H NMR、13C NMR showed that three methyl groups [ delta C14.9, 22.6 and 10.9 (C-13, C-14 and C-15, respectively), delta H2.02 (s, Me-15), delta H1.94 (s, Me-13) and delta H1.18 (s, Me-14) were present in the structure of the compound]Two methylene groups [ δ C39.0 and 41.7 (C-5 and C-6, respectively), δ H3.23 (dd, J = 2.4, 7.6 Hz, H-5a) and δ H1.61 (d, J = 13.5 Hz, H-5b), δ H3.26 (dd, J = 2.4,5.7 Hz, H-6a) and 1.85 (t, J = 12.4 Hz, H-6b)]One carbonyl group [ Delta C180.5 (corresponding to C-1)]。
Then, two-dimensional nuclear magnetic resonance spectrum data such as HMBC, DEPT-135 and HSQC spectra are combined to determine the planar structure of the compound, the structural formula is shown as the attached figure 1, and the chemical name is 8- (-1, 2-dihydroxyethyl) -2-hydroxy-3, 4, 7-trimethyl-5, 6-dihydronaphthalene-1-ketone.
Of compounds II1H-1The structure analysis process of H-COSY, HMBC and NOESY is shown in figure 16.
(2) The spectral data for this compound are summarized below:
1H NMR (600 MHz, MeOD) δ 6.54 (s, 1H, H-10), 4.45 (dd, J = 5.6, 11.9 Hz, 1H, H-11), 4.26 (d, J = 11.9 Hz, 1H, H-12a), 4.15 (d, J = 11.9 Hz, 1H, H-12b), 3.26 (dd, J = 2.4, 5.7 Hz, 1H, H-6a), 1.85 (t, J = 12.4 Hz, 1H, H-6b), 3.23 (dd, J = 2.4, 7.6 Hz, 1H, H-5a), 1.61 (d, J = 13.5 Hz, 1H, H-5b), 2.02 (s, 3H, H-15), 1.94 (s, 3H, H-13), 1.18 (s, 3H, H-14)。
13C NMR (150 MHz, MeOD) δ180.5 (C-1), 172.0 (C-9), 144.8 (C-2), 134.7 (C-3), 132.7 (C-8), 126.7 (C-7), 117.1 (C-1), 67.8 (C-11), 61.8 (C-12), 44.2 (C-4), 41.7 (C-6), 39.0 (C-5), 22.6(C-14), 14.9 (C-13), 10.9 (C-15)。
thirdly, the research of the drug effect experiment of the terpenoid compound of the invention
(I) Experimental materials and instruments
1. Experimental Material
The compounds I and II prepared in the examples (self-extracted, purity 98% or more); 10 cell strains (basic medical cell center of institute of basic medicine of Chinese medical academy of sciences), such as liver cancer cell (HepG 2), breast cancer cell (MCF-7), cervical cancer cell (HeLa), lung cancer cell (A549), ovarian cancer cell (IA-9), renal cancer cell (CAKI-1), prostate cancer (PC-3), nasopharyngeal carcinoma (KB), melanoma (SK-MEL-2), intestinal cancer (HCT-8), and the like; cisplatin (Macklin corporation); DMEM medium (solibao corporation); trypsin (Trypsin-EDTA, Solebao Co.); FBS (Gibco Corp.); DMSO (Dalochi chemical Agents works, Tianjin); PBS buffer (solibao corporation); MTT (SIGMA Co., 5 mg. prepared with PBS buffer before use)-1A solution).
2. Laboratory apparatus
Inverted electron microscope (OLYMPUS: CKX41, Japan), microplate reader (Molecular Devices: SpectraMax i3, USA), MCV-B161S (T) type superclean bench (SANYO, Japan), MCO-20ACI type CO2Thermostated incubator (Thermo company, usa), a ten thousandth balance model BS125S (mettler-toledo, switzerland), a desk top microcentrifuge model TDL-5C (Eppendorf, germany).
3. Preparation of test drugs
The compound I, the compound II and the positive drug cisplatin prepared in the embodiment are respectively weighed, and are added with dimethyl sulfoxide (DMSO) solution for dissolution to prepare 1mg/mL sample solution to be tested, and the sample solution is stored in a refrigerator at the temperature of-20 ℃ for later use. When in use, the culture medium is diluted into a liquid medicine with required concentration according to the experimental requirement (the DMSO content is controlled within 0.1%).
(II) Experimental method
1. Cell culture
DMEM medium containing 10% FBS was used as the cell culture medium in CO2Culturing in a constant temperature incubator. And (5) starting passage when the cell density reaches 80-90%. After removing the cells from the incubator, the cells were observed under a microscope, and then the following operations were performed on a sterile operating table: discarding the old culture medium, adding 2 mL of PBS buffer solution to clean the cells, adding 1mL of trypsin-EDTA solution after discarding, slightly shaking to enable the trypsin-EDTA solution to be paved on the surface of the whole culture bottle, digesting for 2 min at room temperature, observing the cells under a microscope and ensuring complete digestion, adding 2 mL of culture medium to collect the cells, transferring the cells to a 15 mL centrifuge tube, centrifuging the centrifuge tube (1000 rpm for 3 min), discarding the supernatant, adding 2 mL of culture medium to blow and resuspend the cells, adding a proper amount of cells into a new culture dish, slightly shaking to enable the cells to be uniformly distributed, then placing the cells back to an incubator to be continuously cultured, and taking the cells in a logarithmic phase for experiment.
2. Activity assay
The MTT method is adopted for carrying out: taking cells in logarithmic growth phase, adjusting the concentration of cell suspension to 1 × 105Bore et-1Seeded in 96-well plates at 100. mu.L per well in 5% CO2After culturing in an incubator at 37 ℃ for 24 h, the cell monolayer is paved on the bottom of a well (a 96-well flat bottom plate), and the cell monolayer is randomly divided into an administration group, a positive control group and a blank control group, wherein each group is provided with 6 multiple wells. The drug group is added with 100 mu L of drug-containing culture medium with different concentrations (1.5625, 3.125, 6.25, 12.5, 25, 50 mu g/mL). Each well of the positive control group was filled with 100. mu.L of medium containing different concentrations of the positive drug (1.5625, 3.125, 6.25, 12.5, 25, 50. mu.g/mL). The blank control group was added with 100. mu.L of drug-free medium. Placing 96-well plate in 5% CO2Incubator (37 ℃) 48 h, 5 mg/well/. mL-1MTT 10. mu.L, after further culturing for 4 h, the supernatant was discarded, DMSO 150. mu.L was added, and the mixture was dissolved for 10 min with shaking. Using enzyme-linked immunosorbent assayThe OD was measured at 490 nm to calculate the inhibition (%) of the drug on cell growth. According to the calculation formula: inhibition (%) = [1- (mean OD value of administration group)/mean OD value of control group)]X 100%. The IC50 value was calculated according to the formula of Kouzhi.
(III) results of the experiment
The inhibitory activity of the compound of the present invention on 10 kinds of cancer cells (IC 50: μ g/mL) was as shown in Table 1 below.
TABLE 1
Figure 70965DEST_PATH_IMAGE002
The results in Table 1 show that the compounds I and II of the present invention have some inhibitory effect on the above 10 cancer cells (IC 50 < 30. mu.g. mL)-1) The compound has better development prospect and can be used for developing new anti-cancer drugs or new lead compounds.
Finally, it should be noted that the above-mentioned embodiments are only preferred embodiments of the present invention, and not intended to limit the present invention, and although the present invention has been described in detail with reference to the foregoing embodiments, it will be apparent to those skilled in the art that modifications and equivalents can be made in the technical solutions described in the foregoing embodiments, or some technical features thereof can be replaced. Any modification, equivalent replacement, or improvement made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (7)

1. A terpenoid characterized by: the terpenoids comprise a compound I and a compound II, and the structural formula of the terpenoids is as follows:
Figure 894645DEST_PATH_IMAGE001
2. a method of preparing the terpenoid of claim 1, wherein: the method specifically comprises the following steps:
(1) taking a lilac stem medicinal material, extracting and concentrating, and then sequentially extracting with dichloromethane and ethyl acetate to obtain a dichloromethane extraction part and an ethyl acetate extraction part;
(2) dissolving the ethyl acetate extraction part obtained in the step (1) with methanol, adding silica gel into the mixture, mixing the mixture with a dry method, loading the mixture into a silica gel column, performing column chromatography separation, and performing gradient elution by using dichloromethane-methanol with the volume ratio of 100:0, 90:10, 80:20, 70:30, 50:50, 30:70 and 0:100 to obtain 14 fractions 1,2, 3,4,5, 6, 7, 8, 9,10, 11, 12, 13 and 14;
(3) subjecting the fraction 10 to silica gel column chromatography, and gradient eluting with petroleum ether-acetone at volume ratio of 10:1, 8:1, 4:1, 2:1, 1:1, and 0:1 to obtain 5 fractions a, b, c, d, and e;
(4) separating fraction d by preparative liquid chromatography, eluting with acetonitrile-water at a volume ratio of 20:80 as mobile phase, collecting two chromatographic peaks with retention time of 18min and 20min respectively to obtain two crude compounds;
(5) and (4) dissolving the two crude compounds obtained in the step (4) and then recrystallizing to obtain pure compound I and compound II.
3. The method for preparing terpenoids according to claim 2, characterized in that: in the step (2), the silica gel used for sample mixing is 100-200 meshes, and the silica gel used in the silica gel column is 100-200 meshes.
4. The method for preparing terpenoids according to claim 2, characterized in that: in the step (3), the silica gel used in the silica gel column is 200-300 meshes.
5. The method for preparing terpenoids according to claim 2, characterized in that: in the step (4), the chromatographic conditions of the preparative liquid chromatography are as follows: and a YMC-TriartC18 chromatographic column, wherein the size of the chromatographic column is 250 multiplied by 20mm, the particle size is 5 mu m, and the flow rate is 10 mL/min.
6. Use of the terpenoid of claim 1 for the preparation of an anticancer drug.
7. A pharmaceutical formulation characterized by: comprises a therapeutically effective amount of the terpenoid of claim 1 and a pharmaceutically acceptable carrier or adjuvant, and the pharmaceutical preparation is an oral preparation or an injection preparation.
CN202210084250.2A 2022-01-25 2022-01-25 Terpenoid and preparation method and application thereof Active CN114105751B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210084250.2A CN114105751B (en) 2022-01-25 2022-01-25 Terpenoid and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210084250.2A CN114105751B (en) 2022-01-25 2022-01-25 Terpenoid and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN114105751A true CN114105751A (en) 2022-03-01
CN114105751B CN114105751B (en) 2022-04-15

Family

ID=80360942

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210084250.2A Active CN114105751B (en) 2022-01-25 2022-01-25 Terpenoid and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114105751B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114736106A (en) * 2022-04-28 2022-07-12 江西中医药大学 Lignan dimer compound and preparation method and application thereof
CN116903576A (en) * 2023-09-13 2023-10-20 江西中医药大学 Triterpene compound in syringa oblata, and preparation method and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103351419A (en) * 2013-07-09 2013-10-16 陕西中药研究所 Two-step simultaneous preparation method for pedunculoside and syringin
US20160046593A1 (en) * 2014-08-12 2016-02-18 National Taiwan University Preparation of Ambrox from Labdanes of Dysoxylum hongkongense, and the Preparation of New Diterpenoids from Dysoxylum hongkongense
CN108610241A (en) * 2018-06-15 2018-10-02 东北林业大学 A method of extracting hydroxytyrosol from lilac
KR20210104633A (en) * 2019-08-13 2021-08-25 한국화학연구원 Composition for controlling plant diseases including diterpenoid compounds derived from an extract of Platycladus orientalis as an active ingredient and method of controlling plant diseases using the same
CN113563172A (en) * 2021-09-23 2021-10-29 江西中医药大学 Azulene compound and preparation method and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103351419A (en) * 2013-07-09 2013-10-16 陕西中药研究所 Two-step simultaneous preparation method for pedunculoside and syringin
US20160046593A1 (en) * 2014-08-12 2016-02-18 National Taiwan University Preparation of Ambrox from Labdanes of Dysoxylum hongkongense, and the Preparation of New Diterpenoids from Dysoxylum hongkongense
CN108610241A (en) * 2018-06-15 2018-10-02 东北林业大学 A method of extracting hydroxytyrosol from lilac
KR20210104633A (en) * 2019-08-13 2021-08-25 한국화학연구원 Composition for controlling plant diseases including diterpenoid compounds derived from an extract of Platycladus orientalis as an active ingredient and method of controlling plant diseases using the same
CN113563172A (en) * 2021-09-23 2021-10-29 江西中医药大学 Azulene compound and preparation method and application thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HIKINO HIROSHI ET AL.: ""Sesquiterpenoids. XLIX. Stereoselectivity in microbiological transformation of stereoisomers of α-cyperone and dihydro-α-cyperone with Colletotrichum phomoides"", 《CHEMICAL & PHARMACEUTICAL BULLETIN》 *
蔡恩博 等: ""丁香属植物化学成分研究进展"", 《北方园艺》 *
高佳琪 等: ""丁香属植物中萜类成分研究进展"", 《中国中药杂志》 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114736106A (en) * 2022-04-28 2022-07-12 江西中医药大学 Lignan dimer compound and preparation method and application thereof
CN114736106B (en) * 2022-04-28 2023-06-09 江西中医药大学 Lignan dimer compound, and preparation method and application thereof
CN116903576A (en) * 2023-09-13 2023-10-20 江西中医药大学 Triterpene compound in syringa oblata, and preparation method and application thereof
CN116903576B (en) * 2023-09-13 2023-11-24 江西中医药大学 Triterpene compound in syringa oblata, and preparation method and application thereof

Also Published As

Publication number Publication date
CN114105751B (en) 2022-04-15

Similar Documents

Publication Publication Date Title
CN114105751B (en) Terpenoid and preparation method and application thereof
CN113620912B (en) Furanone compound and preparation method and application thereof
CN114262294B (en) Phenyl isoquinoline alkaloid compound and preparation method and application thereof
CN108558980B (en) Cardiac glycoside compound separated from streblus streblumea root and having antitumor activity and application thereof
CN113801000B (en) Compound with anti-tumor activity and preparation method and application thereof
CN113061124B (en) Sesquiterpene dimer compound, and preparation method, application and pharmaceutical composition thereof
CN109180632B (en) A method for preparing compound separated from radix Tripterygii Wilfordii
CN109206392B (en) Coumarin compound and preparation method and application thereof
CN112724113A (en) Flavonoid compound with anti-tumor activity and preparation method and application thereof
CN107325069B (en) Extraction method of sesquiterpenoids
CN106619611B (en) Application of eupatorium lactone in preparing medicine for resisting liver cancer and colon cancer
CN115536532B (en) Crotonane diterpenoid compound, preparation method, pharmaceutical composition and application thereof
CN115677471B (en) Rose alkyl diterpenoid compound, preparation method, pharmaceutical composition and anti-tumor application
CN113980085B (en) Preparation method and application of polyoxapregnane compounds sub-nuostane G
CN112125804B (en) Diterpenoid of euphorbia lathyris, preparation method and anti-leukemia application thereof
CN111410645B (en) Lignans compound and preparation and application thereof
CN110305081B (en) Dihydro agarofuran type sesquiterpene and preparation and application thereof
CN116925054B (en) Lignan compound in syringa oblata, and preparation method and application thereof
CN112300185B (en) Alkaloid compound with reduced hepatotoxicity, and preparation method and application thereof
CN116115602B (en) New application of compound chaetomougilin O in preparation of medicaments for preventing or treating thyroid cancer
CN115974695B (en) Preparation method and application of vibsane diterpenoid compounds in coral tree
CN111995647B (en) Compound with anti-tumor activity separated from Japanese apricot and preparation method thereof
CN111187327B (en) Method for extracting triterpenoid compound gendarussa terpene A from gendarussa and application of triterpenoid compound gendarussa terpene A
CN116354824B (en) Rosin alkane type diterpenoid compound, preparation method, pharmaceutical composition and application thereof
CN114671759B (en) Enantiomer-kaurane diterpenoid compound with anti-tumor activity and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant