Composition and application thereof in preparation of medicine for treating cancer

文档序号:493130 发布日期:2022-01-07 浏览:6次 中文

阅读说明:本技术 组合物及其在制备治疗癌症的药物中的应用 (Composition and application thereof in preparation of medicine for treating cancer ) 是由 齐海龙 王晓芳 李伟伟 于 2020-07-06 设计创作,主要内容包括:本发明涉及医药技术领域,尤其涉及组合物及其在制备治疗癌症的药物中的应用。本发明提供了含有式I化合物或其可药用盐、二聚体或三聚体和酪氨酸激酶活性抑制剂的组合物。基于酪氨酸激酶活性抑制剂靶向VEGFR,EGFR等受体酪氨酸激酶的抑制的作用,本发明通过体内,体外抗肿瘤试验表明索格列净与酪氨酸激酶活性抑制剂联合用药可以对肿瘤产生协同抑制作用。两药联用的效果显著由于单一药物。(The invention relates to the technical field of medicines, in particular to a composition and application thereof in preparing a medicine for treating cancer. The present invention provides compositions comprising a compound of formula I, or a pharmaceutically acceptable salt, dimer or trimer thereof, and an inhibitor of tyrosine kinase activity. Based on the inhibition effect of the tyrosine kinase activity inhibitor targeting VEGFR, EGFR and other receptor tyrosine kinases, the invention shows that the joint medication of the sugelliflozin and the tyrosine kinase activity inhibitor can generate the synergistic inhibition effect on tumors through in-vivo and in-vitro anti-tumor experiments. The effect of the combination of the two medicines is remarkable because of the single medicine.)

1. A composition comprising a compound of formula I, or a pharmaceutically acceptable salt, dimer or trimer thereof, and an inhibitor of tyrosine kinase activity;

wherein: r1Is hydrogen or optionally substituted C1-10Alkyl radical, C1-5-cycloalkyl or 5-membered heterocycle, said optional substitution being with one or more R1ASubstitution;

each R1AIndependently is amino, ester, amide, thiol, carboxylic acid, cyano, halogen, hydroxy or optionally substituted C1-4-alkoxy, C1-5-cycloalkyl or 5-membered heterocycle, said optional substitution being with one or more R1BSubstitution; each R1BIndependently is C1-4-alkyl, halogen or hydroxy; n is 0, 1 or 2;

each R2Independently F OR OR2AWherein each R is2AIndependently of each other is hydrogen, C1-4-alkyl or acyl;

each R3Independently is halogen, hydroxy or optionally substituted C1-10-alkyl or C1-10-alkoxy, said optional substitution being with one or more R3ASubstitution;

each R3AIndependently is amino, ester, amide, thiol, carboxylic acid, cyano, halogen, hydroxy or optionally substitutedSubstituted C1-4-alkoxy, C1-5-cycloalkyl or 5-membered heterocycle, said optional substitution being with one or more R3BSubstitution; each R3BIndependently is C1-4-alkyl, amino, cyano, halogen or hydroxy; p is 0, 1 or 2;

each R4Independently is R4A、-N(R4A)(R4B)、-OR4A、-SR4A、-S(O)R4Aor-S (O)2R4A

R4AIs optionally substituted C4-20-alkyl or 4-20 membered heteroalkyl, said optional substitution being with one or more R4CIs substituted, and is optionally attached to another R4AA moiety to provide a dimer or trimer; r4BIs hydrogen or R4A(ii) a Each R4CIndependently is amino, aminoacyl, azo, carbonyl, carboxyl, cyano, formyl, guanidino, halogen, hydroxyl, imidoyl, imino, isothiocyanate, nitrile, nitro, nitroso, nitroxyl, oxy, sulfanyl, sulfinyl, sulfonyl, thioaldehyde, thiocyanate, thione, thiourea, urea, or X1、X1-L1-X2Or X1-L1-X2-L2-X3Wherein each X is1、X2And X3Independently is optionally substituted C1-4Alkyl radical, C1-6-cycloalkyl, 5-or 6-membered heterocycle or aryl, said optional substitution being with one or more R4DIs substituted, and each L1And L2Independently is optionally substituted C1-6-alkyl or 1-10 membered heteroalkyl, said optional substitution being with one or more R4ESubstitution; each R4DIndependently is R4EOr optionally with one or more R4ESubstituted C1-6-an alkyl group; each R4EIndependently is amino, aminoacyl, azo, carbonyl, carboxyl, cyano, formyl, guanidino, halogen, hydroxyl, imidoyl, imino, isothiocyanate, nitrile, nitro, nitroso, nitroxyl, oxo, sulfanyl, sulfinyl, sulfonyl, thioaldehyde, thiocyanate, thione, or urea; and ism is 1, 2 or 3.

2. The composition of claim 1, wherein the tyrosine kinase activity inhibitor comprises: EGFR inhibitors, c-Kit, c-Met, c-Ret, Raf, PDGFR, BTK, PKA/C, FGFR inhibitors, VEGFR inhibitors.

3. The composition of claim 2, wherein the tyrosine kinase activity inhibitor is: multiple target kinase inhibitors, tivozanib, genistein, panatinib, dapoxetine, tilinib, erlotinib, osertinib mesylate, osertinib, natatinib, AZD3759, erlotinib, elvitinib or lasertinib, lidocaine hydrochloride, 4- [ (1E) -2- [5- [ (1R) -1- (3, 5-dichloro-4-pyridinyl) ethoxy ] -1H-indazol-3-yl ] vinyl ] -1H-pyrazol-1-ethanol, axitinib, nidib, cedanib, pazopanib hydrochloride, sunitinib malate, britinib, cabozantinib, alantinib, lenvatinib, regorafenib md, ennetup-2076 tartrate, tiratinib, gefitinib, sunitinib hydrochloride, sunitinib mesylate, gefitinib hydrochloride, gefitinib, neritinib, gefitinib, and sunitinib, Pazopanib, cabozantinib malate, regorafenib hydrate, nintedanib ethanesulfonate, lenvatinib mesylate, cedanib maleate, furoquintinib, sulitinib, imatinib, cetraratinib, vandetanib, gefitinib, afatinib, apatinib, erlotinib or sorafenib, taxifolin or vitamin E.

4. The composition according to any one of claims 1 to 3, wherein the molar ratio of the compound of formula I or the pharmaceutically acceptable salt, dimer or trimer thereof to the tyrosine kinase activity inhibitor is (10 to 40) to (5 to 60).

5. The composition of any one of claims 1 to 3, wherein the compound of formula I is suggestin.

6. Use of a composition according to any one of claims 1 to 5 for the preparation of a medicament for the prevention and/or treatment of cancer.

7. The use of claim 6, wherein the cancer comprises: bladder cancer, blood cancer, bone cancer, brain cancer, breast cancer, central nervous system cancer, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, gallbladder cancer, gastrointestinal cancer, genitourinary cancer, head cancer, kidney cancer, laryngeal cancer, liver cancer, lung cancer, muscle tissue cancer, neck cancer, oral or nasal mucosa cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, spleen cancer, small intestine cancer, large intestine cancer, stomach cancer, testicular cancer, and/or thyroid cancer.

8. The use of claim 6, wherein the treatment comprises inhibiting proliferation of tumor cells and/or inhibiting tumor volume.

9. A medicament for the treatment of cancer comprising a composition according to any one of claims 1 to 5.

10. The medicament of claim 9, wherein the medicament is administered orally, and the dosage form of the medicament comprises granules, pills, powder, tablets, capsules, oral liquid or syrup.

Technical Field

The invention relates to the technical field of medicines, in particular to a composition and application thereof in preparing a medicine for treating cancer.

Background

1. Epidemiology of cancer

Non-infectious diseases are the main causes of death in the world, and cancer is the disease with the highest fatality rate among the non-infectious diseases, and brings heavy burden to the social health medical system. Traditional cancer treatment is mainly performed by surgery and radiotherapy, and chemotherapy is mainly performed on advanced cancer. Classical chemotherapy has great side effects due to poor targeting. The occurrence of targeted chemotherapy drugs represented by gleevec greatly reduces the pain of patients caused by chemotherapy. The targeted drug is designed for molecules different from normal cell growth characteristics and expression of cancer cells, such as gleevec specificity, which has good therapeutic effect on constitutively activated tyrosine kinase in chronic myelogenous leukemia (Flynn and gerries, 2020). Another significant feature of cancer cells, unlike normal cells, is an alteration in the metabolic pattern. In order to balance the need for cellular components for rapid proliferation and to maintain the energy supply required for survival, cancer cells prefer to utilize glucose in an aerobic glycolysis fashion, which is called the Warburg effect (Warburg, 1956). Aerobic glycolysis does not sufficiently oxidize glucose to produce ATP, but can produce a large number of intermediate metabolites for DNA and protein synthesis to promote cancer cell proliferation. It is therefore feasible to target the carbohydrate metabolism of tumor cells, thereby achieving the goal of inhibiting cancer cell proliferation (Kroemer and Pouyssegur, 2008).

2. Suggestin and cancer treatment

Glucose uptake by cancer cells from the external environment is mainly achieved by glucose transporters, which are divided into two major families, the GLUT family, which transports along a glucose concentration gradient in a diffusion-assisted manner, and the SGLT family, which transports glucose uptake by co-transport of sodium ions, and which actively transports external glucose to cells for use in an ATP-consuming manner (Navale and parajame, 2016). The two most prominent members of the SGLT family are SGLT1 and SGLT2, SGLT2 is distributed mainly in the anterior end of the proximal convoluted tubule of the kidney to reabsorb more than 97% of the glucose in the raw urine into the blood by active transport, while SGLT1 is distributed mainly in the epithelial cells of the intestinal serosa and distal end of the proximal convoluted tubule of the kidney to absorb the glucose in the intestinal tract by active transport and the remaining approximately 3% of the glucose in the raw urine after absorption by SGLT2 (douminez Rieg and Rieg, 2019). Because of the important roles of SGLT1 and SGLT2 in sugar absorption and reabsorption, it is an ideal target for the treatment of diabetes. Currently, the empagliflozin targeting SGLT2, canagliflozin and dapagliflozin show good treatment effects when clinically used for treating type II diabetes, and also have the curative effect of reducing cardiovascular diseases. Miglityn alone targeted SGLT1 has entered the clinical study stage. Sogeletin targeting both SGLT1 and SGLT2 was approved for marketing in the european union.

TKI and TKI resistance of cancer

Receptor Tyrosine Kinases (RTKs) are the most common genes in cancer that promote cancer progression, and RTKs, when bound to the corresponding ligands, typically dimerize and catalyze autophosphorylation to initiate a series of downstream signaling cascades that promote cancer cell proliferation (Lemmon and Schlessinger, 2010; Yarden and Pines, 2012). Cancer cells often overexpress RTKs or constitutively activate mutations of RTKs to upregulate RTK activity to promote self-proliferation, and thus targeting RTK activity is a major means for developing anticancer drugs, including small molecule tyrosine kinase activity inhibitors (TKIs) targeting the ATP-binding pocket of RTKs, and targeting ligand-bound mabs and the like (Thomas and Weihua, 2019). The successful development of TKI, which effectively prolongs the disease-free progression survival of patients with over-expressed or constitutively activated RTK, significantly improves the quality of life of the patients, and most TKI are first-line drugs in the corresponding cancer field at present, however, patients who respond well to treatment with TKI even though they are naturally resistant to TKI develop acquired resistance within a treatment period of about one year (Camdge et al; Cortot and Janne, 2014). Summary the reasons for drug resistance are mainly divided into pharmacological resistance and biological resistance, pharmacological resistance is mostly caused by drug interaction in the body and drug concentration around cancer cells cannot be achieved, cancer cells may still be sensitive to drugs, while biological resistance is caused by cancer heterogeneity, drug resistance mutation caused by drug selection pressure, and alternative activation of alternative signal pathways (Minuti, g., a et al). Biological resistance is the main reason for TKI class drug resistance. Against cancer cells

The mechanism of acquired resistance, researchers have devised a number of approaches to overcome cancer resistance, including the development of newer inhibitors for the second, third, etc. generation, against conventional resistance mutations, in combination with radiation or chemotherapy after resistance, and with new targeted drugs. To date, even third-generation inhibitors have acquired resistance, and radiotherapy and chemotherapy have received little benefit. Overcoming resistance in combination with other targeted drugs is the primary choice.

Disclosure of Invention

In view of the above, the technical problem to be solved by the present invention is to provide a composition and its use in preparing a medicament for treating cancer, wherein the composition comprises sueglin and a tyrosine kinase activity inhibitor.

The present invention also provides a composition comprising a compound of formula I, or a pharmaceutically acceptable salt, dimer or trimer thereof, and an inhibitor of tyrosine kinase activity;

wherein: r1Is hydrogen or optionally substituted C1-10Alkyl radical, C1-5-cycloalkyl or 5-membered heterocycle, said optional substitution being with one or more R1ASubstitution;

each R1AIndependently is amino, ester, amide, thiol, carboxylic acid, cyano, halogen, hydroxy or optionally substituted C1-4-alkoxy, C1-5-cycloalkyl or 5-membered heterocycle, said optional substitution being with one or more R1BSubstitution; each R1BIndependently is C1-4-alkyl, halogen or hydroxy; n is 0, 1 or 2;

each R2Independently F OR OR2AWherein each R is2AIndependently of each other is hydrogen, C1-4-alkyl or acyl;

each R3Independently is halogen, hydroxy or optionally substituted C1-10-alkyl or C1-10-alkoxy, said optional substitution being with one or more R3ASubstitution;

each R3AIndependently amino, ester, amide, thiol, carboxylic acid, cyano, halogen, hydroxy or optionally substitutedC1-4-alkoxy, C1-5-cycloalkyl or 5-membered heterocycle, said optional substitution being with one or more R3BSubstitution; each R3BIndependently is C1-4-alkyl, amino, cyano, halogen or hydroxy; p is 0, 1 or 2;

each R4Independently is R4A、-N(R4A)(R4B)、-OR4A、-SR4A、-S(O)R4Aor-S (O)2R4A

R4AIs optionally substituted C4-20-alkyl or 4-20 membered heteroalkyl, said optional substitution being with one or more R4CIs substituted, and is optionally attached to another R4AA moiety to provide a dimer or trimer; r4BIs hydrogen or R4A(ii) a Each R4CIndependently is amino, aminoacyl, azo, carbonyl, carboxyl, cyano, formyl, guanidino, halogen, hydroxyl, imidoyl, imino, isothiocyanate, nitrile, nitro, nitroso, nitroxyl, oxy, sulfanyl, sulfinyl, sulfonyl, thioaldehyde, thiocyanate, thione, thiourea, urea, or X1、X1-L1-X2Or X1-L1-X2-L2-X3Wherein each X is1、 X2And X3Independently is optionally substituted C1-4Alkyl radical, C1-6-cycloalkyl, 5-or 6-membered heterocycle or aryl, said optional substitution being with one or more R4DIs substituted, and each L1And L2Independently is optionally substituted C1-6-alkyl or 1-10 membered heteroalkyl, said optional substitution being with one or more R4ESubstitution; each R4DIndependently is R4EOr optionally with one or more R4ESubstituted C1-6-an alkyl group; each R4EIndependently is amino, aminoacyl, azo, carbonyl, carboxyl, cyano, formyl, guanidino, halogen, hydroxyl, imidoyl, imino, isothiocyanate, nitrile, nitro, nitroso, nitroxyl, oxo, sulfanyl, sulfinyl, sulfonyl, thioaldehyde, thiocyanate, thione, or urea; and ism is 1, 2 or 3.

In the present invention, R1Is methyl, n ═ 0; r2Are all-OH; p is 1, R3Is Cl; m is 1, R4Is ethoxy.

In some embodiments, the compound of formula I is sugelliflozin, having the structure of formula II.

In some embodiments, the composition consists of sugelliflozin and an inhibitor of tyrosine kinase activity.

The tyrosine kinase activity inhibitor comprises: EGFR inhibitors, c-Kit, c-Met, c-Ret, Raf, PDGFR, BTK, PKA/C, FGFR inhibitors, VEGFR inhibitors.

The EGFR inhibitor comprises: gefitinib, erlotinib, afatinib, lapatinib ditosylate, genistein, lapatinib, sapertinib, daparinib, valtinib, erlotinib, lidocaine hydrochloride, osetinib mesylate, osetinib, pozitinib, natatinib, AZD3759, temotinib, elvitinib, neratinib, lazatitinib;

the c-Met inhibitors include: cabozantinib;

the PKA/C inhibitors include: daphnetin;

the BTK inhibitor includes: (ii) tematinib;

the c-Ret inhibitors include: regorafenib hydrate, regorafenib;

the Raf inhibitors include: regorafenib hydrate;

the FGFR inhibitor comprises: 4- [ (1E) -2- [5- [ (1R) -1- (3, 5-dichloro-4-pyridinyl) ethoxy ] -1H-indazol-3-yl ] vinyl ] -1H-pyrazole-1-ethanol; nintedanib, nintedanib ethanesulfonate, ponatinib, brimonib, alanine brimonib;

the c-Kit inhibitors include: axitinib, pazopanib hydrochloride, regorafenib hydrate, sunitinib malate, sunitinib, cetraratinib, tiratinib;

the PDGFR inhibitor includes: axitinib, tivozanib, tiratinib, nintedanib ethanesulfonate, pazopanib hydrochloride, panatinib;

the VEGFR inhibitors include: apatinib, axitinib, nintedanib, cedanib, pazopanib hydrochloride, sunitinib malate, brimonib, cabozantinib, alantinib, lenvatinib, regorafenib, ENMD-2076 tartrate, tivozanib, panatinib, furazotinib, tiratinib, taxifolin, pazopanib, cabozantinib malate, vitamin E, regorafenib hydrate, nidanib ethanesulfonate, lenvatinib mesylate, cedanib maleate, 4- [ (1E) -2- [5- [ (1R) -1- (3, 5-dichloro-4-pyridyl) ethoxy ] -1H-indazol-3-yl ] vinyl ] -1H-pyrazole-1-ethanol, VEGFR-targeted monoclonal antibody drugs such as sunitinib, cetraratinib, antrocinib, sorafenib, vandetanib and bevacizumab.

In the invention, the anti-tumor drugs for verifying the combined drug effect of the suggestin and the anti-tumor drugs comprise: multiple target kinase inhibitors, tivozanib, genistein, panatinib, dapatinib, tilinib, erlotinib, osertinib mesylate, osertinib, natatinib, AZD3759, erlotinib, elvitinib, lasertinib, lidocaine hydrochloride, 4- [ (1E) -2- [5- [ (1R) -1- (3, 5-dichloro-4-pyridinyl) ethoxy ] -1H-indazol-3-yl ] vinyl ] -1H-pyrazol-1-ethanol, axitinib, ninib, cinidanib, cediranib hydrochloride, sunitinib malate, britinib, cabozantinib, alantinib, lenvatinib, regorafenib MD, ENE-2076 tartrate, tiratinib, gefitinib, and mixtures thereof, Pazopanib, cabozantinib malate, regorafenib hydrate, nintedanib ethanesulfonate, lenvatinib mesylate, cedanib maleate, furoquintinib, sunitinib, imatinib, cetraratinib, vandetanib, gefitinib, afatinib, apatinib, erlotinib, sorafenib, taxifolin, or vitamin E.

The molar ratio of the compound of the formula I or the pharmaceutically acceptable salt, dimer or trimer thereof to the tyrosine kinase activity inhibitor is (10-40) to (5-60).

In some embodiments, the molar ratio of suggestin to the tyrosine kinase activity inhibitor is (10-40) to (5-60).

The composition of the invention is applied to the preparation of a medicament for treating cancer.

The cancer includes: bladder cancer, blood cancer, bone cancer, brain cancer, breast cancer, central nervous system cancer, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, gallbladder cancer, gastrointestinal cancer, genitourinary cancer, head cancer, kidney cancer, laryngeal cancer, liver cancer, lung cancer, muscle tissue cancer, neck cancer, oral or nasal mucosa cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, spleen cancer, small intestine cancer, large intestine cancer, stomach cancer, testicular cancer, and/or thyroid cancer.

In the present invention, the treatment comprises inhibiting the proliferation of tumor cells and/or inhibiting tumor volume. In the embodiment of the invention, the effect of combining the lung cancer cell, the colorectal cancer cell, the cervical cancer, the ovarian cancer, the bile duct cancer, the gastric cancer, the esophageal cancer and the liver cancer cell is verified.

The invention also provides a medicament for treating cancer, which comprises the composition.

The drug is orally administered, and the dosage form of the drug comprises granules, pills, powder, tablets, capsules, oral liquid or syrup.

In some embodiments provided herein, the capsule is a hard or soft capsule.

In some embodiments provided herein, the tablet is an oral tablet or buccal tablet.

Tablets refer to tablets intended for oral administration, most of which act by absorption through the gastrointestinal tract, and some of which act locally in the gastrointestinal tract. In some embodiments provided herein, the tablet is a compressed tablet, a dispersible tablet, an effervescent tablet, a chewable tablet, a coated tablet, or a sustained release tablet.

The medicine also comprises pharmaceutically acceptable adjuvants including one or more of fruit powder, edible essence, sweetener, sour agent, bulking agent, lubricant, antiseptic, suspending agent, edible pigment, diluent, emulsifier, disintegrating agent or plasticizer.

The invention also provides a method for treating cancer, which is to administer the medicament.

The present invention provides compositions comprising a compound of formula I, or a pharmaceutically acceptable salt, dimer or trimer thereof, and an inhibitor of tyrosine kinase activity. Based on the inhibition effect of the tyrosine kinase activity inhibitor targeting receptor tyrosine kinases such as VEGFR, EGFR and the like, the invention shows that the joint medication of the sugelliflozin and the tyrosine kinase activity inhibitor can generate the synergistic inhibition effect on tumors through in-vivo and in-vitro anti-tumor experiments. The effect of the combination of the two medicines is remarkable because of the single medicine.

Description of the drawings:

FIG. 1 shows the killing effect of gefitinib on lung cancer A549 cells cultured in a sugar-free medium and a sugar-containing medium;

FIG. 2-a-1 shows the inhibitory effect of varying concentrations of gefitinib on lung cancer cell line A549; FIG. 2-a-2 shows the inhibitory effect of different concentrations of suggestin on lung cancer cell line A549; FIGS. 2-a-3 show the inhibitory effect of varying concentrations of gefitinib +10uM suogliflozin in test group 1 on lung cancer cell line A549; FIGS. 2-a-4 show the inhibitory effect of varying concentrations of gefitinib +20uM suggestin in combination with test group 2 on lung cancer cell line A549; FIGS. 2-a-5 show the inhibitory effect of varying concentrations of gefitinib +30uM suogliflozin in test group 3 on lung cancer cell line A549;

FIG. 2-b shows the growth inhibition rate of gefitinib single drug and sugrelide in combination with gefitinib composition for colorectal cancer cell line LoVo;

FIG. 2-c shows the growth inhibition rate of gefitinib single drug and sugrelide in combination with gefitinib composition for colorectal cancer cell line HT 29;

FIG. 2-d shows the growth inhibition rate of gefitinib single drug and sugrelin in combination with gefitinib composition on colorectal cancer cell line SW 620;

FIG. 2-e shows the growth inhibition rate of gefitinib single drug and sugrelin in combination with gefitinib composition for colorectal cancer cell line HCT 116;

FIG. 2-f-1 shows the growth inhibition rate of gefitinib single drug against ovarian cancer cell line SKOV3 at different concentrations; FIG. 2-f-2 shows the inhibitory effect of suggestin at different concentrations on ovarian cancer cell line SKOV 3; FIG. 2-f-3 shows the growth inhibition rate of 10. mu. mol/L suggestin with various concentrations of gefitinib on ovarian cancer cell line SKOV 3; FIG. 2-f-4 shows the growth inhibition rate of 20. mu. mol/L suggestin with various concentrations of gefitinib for ovarian cancer cell line SKOV 3; FIG. 2-f-5 shows the growth inhibition rate of 30. mu. mol/L suggestin with various concentrations of gefitinib for ovarian cancer cell line SKOV 3; FIG. 2-f-6 shows the growth inhibition rate of 40. mu. mol/L suggestin with various concentrations of gefitinib for ovarian cancer cell line SKOV 3;

FIG. 2-g-1 shows the growth inhibition rate of gefitinib single drug on the esophageal cancer cell line KYSE30 at different concentrations; FIG. 2-g-2 is a graph showing the inhibitory effect of Soxhlet alone on the esophageal cancer cell line KYSE30 at different concentrations; FIG. 2-g-3 shows the growth inhibition rate of 10. mu. mol/L suggestin with different concentrations of gefitinib for the esophageal cancer cell line KYSE 30; FIG. 2-g-4 shows the growth inhibition rate of 20. mu. mol/L suggestin with different concentrations of gefitinib for the esophageal cancer cell line KYSE 30; FIG. 2-g-5 shows the growth inhibition rate of 30. mu. mol/L suggestin with different concentrations of gefitinib for the esophageal cancer cell line KYSE 30; FIG. 2-g-6 shows the growth inhibition rate of 40. mu. mol/L suggestin with different concentrations of gefitinib for the esophageal cancer cell line KYSE 30;

FIG. 2-h-1 shows the growth inhibition rate of a single drug with different concentrations of gefitinib for the gastric cancer cell line HGC-27; FIG. 2-h-2 shows the inhibitory effect of different concentrations of suggestin on the gastric cancer cell line HGC-27; FIG. 2-h-3 shows the growth inhibition rate of 10. mu. mol/L suggestin with different concentrations of gefitinib for the gastric cancer cell line HGC-27; FIGS. 2-h-4 show the growth inhibition rates of 20. mu. mol/L of sugliflozin and different concentrations of gefitinib for the gastric cancer cell line HGC-27; FIGS. 2-h-5 show the growth inhibition rates of 30. mu. mol/L of sugliflozin and different concentrations of gefitinib for the gastric cancer cell line HGC-27; FIGS. 2-h-6 show the growth inhibition rates of 40. mu. mol/L of sugliflozin with different concentrations of gefitinib for the gastric cancer cell line HGC-27;

FIG. 2-i-1 shows the growth inhibition rate of different concentrations of gefitinib single drug on the cervical cancer cell line HeLa; FIG. 2-i-2 is a graph showing the inhibitory effects of sugrezin at various concentrations on the cervical cancer cell line HeLa; 2-i-3 shows the growth inhibition rate of 10 mu mol/L suogranzin and different concentrations of gefitinib on the cervical cancer cell line HeLa; FIGS. 2-i-4 show the growth inhibition rates of 20. mu. mol/L of sugrelin with different concentrations of gefitinib for the cervical cancer cell line HeLa; FIGS. 2-i-5 show the growth inhibition rates of 30. mu. mol/L of sugrelin with different concentrations of gefitinib for the cervical cancer cell line HeLa; FIGS. 2-i-6 show the growth inhibition rates of 40. mu. mol/L of sugrelin with different concentrations of gefitinib for the cervical cancer cell line HeLa;

FIG. 2-j-1 shows the growth inhibition rate of a single drug of gefitinib for bile duct cancer cell line RBE at different concentrations; FIG. 2-j-2 shows the inhibitory effect of Soxhlet alone on bile duct cancer cell line RBE at different concentrations FIG. 2-j-3 shows the growth inhibitory rate of 10. mu. mol/L of Soxhlet alone and Gefitinib at different concentrations on bile duct cancer cell line RBE; FIG. 2-j-4 shows the growth inhibition rates of 20. mu. mol/L of suggestin with different concentrations of gefitinib for bile duct cancer cell line RBE; FIG. 2-j-5 shows the growth inhibition rate of 30. mu. mol/L of suggestin with different concentrations of gefitinib for bile duct cancer cell line RBE; FIG. 2-j-6 shows the growth inhibition rate of 40. mu. mol/L of suggestin with different concentrations of gefitinib for bile duct cancer cell line RBE;

FIG. 2-k-1 shows the inhibitory effect of various concentrations of Afatinib on lung cancer cell line A549; FIG. 2-k-2 shows the inhibitory effect of different concentrations of suggestin on lung cancer cell line A549; FIG. 2-k-3 shows the inhibitory effect of different concentrations of Afatinib +10uM suggestin combination on lung cancer cell line A549 in test group 1; FIG. 2-k-4 shows the inhibitory effect of varying concentrations of Afatinib +20uM suggestin in combination on lung cancer cell line A549 in test group 2; FIG. 2-k-5 shows the inhibitory effect of varying concentrations of Afatinib +30uM suggestin in combination on lung cancer cell line A549 in test group 3; FIG. 2-k-6 shows the inhibitory effect of different concentrations of Afatinib +40uM suggestin combination on lung cancer cell line A549 in the test group 4

FIG. 2-1-1 shows the inhibitory effect of different concentrations of erlotinib on lung cancer cell line A549; FIGS. 2-1-2 show the inhibitory effect of different concentrations of suggestin on lung cancer cell line A549; FIGS. 2-1-3 show the inhibitory effect of erlotinib +10uM suggestin combination on lung cancer cell line A549 in test groups 1 at different concentrations; FIGS. 2-l-4 show the inhibitory effect of erlotinib +20uM suggestin combination test group 2 on lung cancer cell line A549 at different concentrations; FIGS. 2-1-5 show the inhibitory effect of erlotinib +30uM suggestin combination on lung cancer cell line A549 in test groups 3 at different concentrations; FIGS. 2-1-6 show the inhibitory effects of erlotinib +40uM suggestin combination on lung cancer cell line A549 in test groups 4 at different concentrations

FIG. 3 shows the killing effect of gefitinib, suggestin and the combination of gefitinib and suggestin on the screened gefitinib-resistant cell strain A549;

FIG. 4-a-1 shows the growth inhibition rate of apatinib single drug and sugrelide in combination with apatinib composition on the liver cancer cell line HepG 2;

FIG. 4-a-2 shows the growth inhibition rates of apatinib single drug and sugregliflozin in combination with apatinib composition on colorectal cancer cell line LoVo;

FIGS. 4-a-3 show the growth inhibition rates of apatinib single drug and sugrelide in combination with apatinib composition on colorectal cancer cell line HT 29;

FIGS. 4-a-4 show the growth inhibition rates of apatinib single drug and sugregliflozin in combination with apatinib composition on colorectal cancer cell line SW 620;

FIGS. 4-a-5 show the growth inhibition rates of apatinib single drug and sugregliflozin in combination with apatinib composition on colorectal cancer cell lines SW 480;

FIG. 4-b-1 shows the inhibitory effect of apatinib at different concentrations on the cell line HepG 2;

FIG. 4-b-2 shows the inhibitory effect of different concentrations of sugelliflozin on the cell line HepG 2;

FIGS. 4-b-3 show the inhibitory effect of varying concentrations of apatinib + 10. mu.M suggestin combination on cell line HepG 2;

FIGS. 4-b-4 show the inhibitory effect of varying concentrations of apatinib + 20. mu.M suggestin combination on cell line HepG 2;

FIGS. 4-b-5 show the inhibitory effect of varying concentrations of apatinib + 30. mu.M suggestin combination on cell line HepG 2;

FIGS. 4-b-6 show the inhibitory effect of varying concentrations of apatinib + 40. mu.M suggestin combination on cell line HepG2 in test group 4;

FIG. 4-c-1 shows the growth inhibition rates of the cholangiocarcinoma cell line RBE by apatinib single drugs at different concentrations; FIG. 4-c-2 shows the growth inhibition rates of different concentrations of suggestin single drug on the bile duct cancer cell line RBE; FIG. 4-c-3 shows the growth inhibition rate of 10. mu. mol/L of Soxhlet and various concentrations of Apatinib on RBE of the cell line bile duct carcinoma; FIG. 4-c-4 shows the growth inhibition rate of 20. mu. mol/L suggestin with varying concentrations of apatinib on RBE of the cell line; FIGS. 4-c-5 show the growth inhibition rate of 30. mu. mol/L of Soxhlet and varying concentrations of Apatinib on RBE; FIGS. 4-c-6 show the growth inhibition rate of 40. mu. mol/L of suggestin with varying concentrations of apatinib on RBE of the cell line cholangiocarcinoma;

FIG. 4-d-1 shows the growth inhibition rate of the esophageal cancer cell line KYSE30 by apatinib single drug at different concentrations; FIG. 4-d-2 shows the growth inhibition rate of different concentrations of suggestin single drug on the esophageal cancer cell line KYSE 30; FIG. 4-d-3 shows the growth inhibition rate of 10. mu. mol/L suggestin with varying concentrations of apatinib on the esophageal cancer cell line KYSE 30; FIG. 4-d-4 shows the growth inhibition rate of 20. mu. mol/L suggestin with varying concentrations of apatinib on the esophageal cancer cell line KYSE 30; FIG. 4-d-5 shows the growth inhibition rate of 30. mu. mol/L suggestin with varying concentrations of apatinib on the esophageal cancer cell line KYSE 30; FIG. 4-d-6 shows the growth inhibition of 40. mu. mol/L suggestin with varying concentrations of apatinib on the esophageal cancer cell line KYSE 30;

FIG. 4-e-1 shows the growth inhibition rate of apatinib single drug on ovarian cancer cell line SKOV3 at different concentrations; FIG. 4-e-2 shows the growth inhibition rate of sugrezin single drug on ovarian cancer cell line SKOV3 at different concentrations; FIG. 4-e-3 shows the growth inhibition rate of 10. mu. mol/L suggestin with various concentrations of apatinib on ovarian cancer cell line SKOV 3; FIG. 4-e-4 shows the growth inhibition rate of 20. mu. mol/L suggestin with various concentrations of apatinib on ovarian cancer cell line SKOV 3; FIG. 4-e-5 shows the growth inhibition rate of 30. mu. mol/L suggestin with various concentrations of apatinib on ovarian cancer cell line SKOV 3; FIGS. 4-e-6 show the growth inhibition rates of 40. mu. mol/L suggestin with various concentrations of apatinib on ovarian cancer cell line SKOV 3;

FIG. 4-f-1 shows the growth inhibition rate of apatinib single drug on the gastric cancer cell line NGC-27 at different concentrations; FIG. 4-f-2 shows the growth inhibition ratios of the single drug of suggestin against the gastric cancer cell line NGC-27 at different concentrations; FIG. 4-f-3 shows the growth inhibition rate of 10. mu. mol/L suggestin with different concentrations of apatinib on the gastric cancer cell line NGC-27; FIG. 4-f-4 shows the growth inhibition rate of 20. mu. mol/L suggestin with various concentrations of apatinib on the gastric cancer cell line NGC-27; FIG. 4-f-5 shows the growth inhibition rate of 30. mu. mol/L suggestin with various concentrations of apatinib on the gastric cancer cell line NGC-27; FIGS. 4-f-6 show the growth inhibition rates of 40. mu. mol/L suggestin with various concentrations of apatinib on the gastric cancer cell line NGC-27;

FIG. 4-g-1 shows the growth inhibition rate of apatinib single drug on the cervical cancer cell line HeLa at different concentrations; FIG. 4-g-2 shows the growth inhibition rate of solifenacin at different concentrations on the cervical cancer cell line HeLa; FIG. 4-g-3 shows the growth inhibition rate of 10. mu. mol/L suggestin with different concentrations of apatinib on the cervical cancer cell line HeLa; FIG. 4-g-4 shows the growth inhibition rate of 20. mu. mol/L suggestin with various concentrations of apatinib on the cervical cancer cell line HeLa; FIG. 4-g-5 shows the growth inhibition rate of 30. mu. mol/L suggestin with various concentrations of apatinib on the cervical cancer cell line HeLa; FIGS. 4-g-6 show the growth inhibition rates of 40. mu. mol/L suggestin with various concentrations of apatinib on the cervical cancer cell line HeLa;

FIG. 5 shows the inhibition of the growth of liver cancer HepG2 cells, colorectal cancer LoVo, HT29, DLD1, SW480, HCT116 cells by the combination of lenvatinib single drug and suogliflozin combined lenvatinib composition;

FIGS. 6-1 to 6-43 show in sequence sugrel and axitinib (FIG. 6-1), nintedanib (FIG. 6-2), cediranib (FIG. 6-3), pazopanib hydrochloride (FIG. 6-4), sunitinib malate (FIG. 6-5), brimonib (FIG. 6-6), cabozantinib (FIG. 6-7), alantinib (FIG. 6-8), lenvatinib (FIG. 6-9), regorafenib (FIG. 6-10), ENMD-2076 (FIG. 6-11), tivazanib (FIG. 6-12), panatinib (FIG. 6-13), ENMD-2076 tartrate (FIG. 6-14), tiratinib (FIG. 6-15), taxifolin (FIG. 6-16), pazopanib (FIG. 6-17), cabozantinib malate (FIG. 6-18), and, Vitamin E (FIGS. 6-19), regorafenib hydrate (FIGS. 6-20), nintedanib ethanesulfonate (FIGS. 6-21), lenvatinib methanesulfonate (FIGS. 6-22), cedanib maleate (FIGS. 6-23), 4- [ (1E) -2- [5- [ (1R) -1- (3, 5-dichloro-4-pyridinyl) ethoxy ] -1H-indazol-3-yl ] vinyl ] -1H-pyrazol-1-ethanol (FIGS. 6-24), sunitinib (FIGS. 6-25), cetraratinib (FIGS. 6-26), ariotinib (FIGS. 6-27), sorafenib (FIGS. 6-28), vandetanib (FIGS. 6-29), furacitinib (FIGS. 6-30), The effect of combining the combination of the imatinib (fig. 6-31), the ostinib (fig. 6-32), the genistein (fig. 6-33), the ibrutinib (fig. 6-34), the dacomitinib (fig. 6-35), the ostinib mesylate (fig. 6-36), the daphnetin (fig. 6-37), the tilinib (fig. 6-38), the AZD3759 (fig. 6-39), the lazatinib (fig. 6-40), the natatinib (fig. 6-41), the lidocaine hydrochloride (fig. 6-42), the icotinib (fig. 6-43);

FIG. 7-a is a graph showing the tumor volume growth of tumor-bearing mice modeled by lung cancer A549 cells during administration of sugrelide monotherapy, apatinib monotherapy and sugrelide in combination with an apatinib composition;

FIG. 7-b shows a gross tumor view of tumor-bearing mice dissected for modeling of lung cancer A549 cells from various groups after the administration of soligliflozin single drug, gefitinib single drug and soligliflozin in combination with gefitinib composition is completed;

figure 7-c is a graph showing the weight of mouse tumors following the end of dosing of the suggestin monotherapy, gefitinib monotherapy and the suggestin in combination with gefitinib composition.

Detailed Description

The invention provides a composition and application thereof in preparing a medicament for treating cancer, and a person skilled in the art can use the content to realize the composition by appropriately improving process parameters. It is expressly intended that all such similar substitutes and modifications which would be obvious to one skilled in the art are deemed to be included in the invention. While the methods and applications of this invention have been described in terms of preferred embodiments, it will be apparent to those of ordinary skill in the art that variations and modifications in the methods and applications described herein, as well as other suitable variations and combinations, may be made to implement and use the techniques of this invention without departing from the spirit and scope of the invention.

Unless specifically stated otherwise in the present invention, all technical and scientific terms referred to in the present invention are intended to have meanings commonly understood by those skilled in the art. The techniques mentioned for use in the present invention are intended to refer to those techniques commonly understood in the art, including those variations of the techniques or alternatives to equivalents that would be apparent to those skilled in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.

The terms "comprising," "including," "having," "containing," or "involving," and other variations thereof as used herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps

The present inventors found in previous studies that tumor cells have a decreased proliferation rate in low-sugar medium and an increased sensitivity to TKI-type inhibitors such as gefitinib, apatinib, etc. in low-sugar medium. The method comprises the following steps: after A549 cells were grown to 80% density, passaged, and the cells were resuspended at 4X 10 using normal medium (25mM glucose)5The cells were plated in 24-well plates at a concentration of 1 ml per well, and after 24 hours, the cells were replaced with a sugar-free medium (0mM glucose) and a normal medium and simultaneously treated with gefitinib, and after 48 hours, the medium was discarded, one ml of 1 XPBS was washed once, 1 ml of 1 XPBS was added to each well to stain for 10 minutes, the trypan blue staining solution was aspirated, and one ml of 1 XPBS was washed three times and photographed under a light microscope. As shown in FIG. 1, the A549 cells cultured in the sugar-free medium (0mM glucose) were colored substantially in blue, indicating that the cells were all dead, whereas the A549 cells in the normal medium (25mM glucose) were not colored substantially in blue. Indicating that the sugarless environment enhanced the sensitivity of a549 cells to gefitinib. Meanwhile, the cell density in the sugar-free culture medium is lower, which indicates that the sugar-free culture can also inhibit the proliferation of cancer cells. With the introduction of the background, sugar uptake by cancer cells is primarily dependent on SGLT family proteins, therefore, the present invention relates to the dual inhibitor suografungin of the two most major and highly expressed members of the SGLT1 and SGLT2 of the SGLT family and its TKI-containing casamino acid kinase inhibitorUse of a composition of a tyrosine kinase inhibitor for the manufacture of a medicament for the treatment of cancer.

The term "treatment" as used herein means that the experimental animal suffering from the disease or disease condition shows partial or total relief of the symptoms after administration of the medicament of the invention, or does not continue to worsen after treatment. Thus, treatment includes healing.

As used herein, "therapeutic effect" means an effect resulting from a treatment, expressed at the cellular level as a rate of inhibition of cell growth or cell death, expressed at the animal level as a rate of alteration thereof, typically alleviation or amelioration of symptoms of a disease or disease condition, or cure of a disease or disease condition, the present invention uniformly assumes a tumor growth inhibition greater than 60% according to the drug's effective prescription while the statistical difference p-value in tumor volume or weight between the treated group and the control group is less than 0.05.

As used herein, "inhibition of cell growth" refers to the ratio of the mean value of absorbance of cells of the treated group by MTT staining to the mean value of absorbance of the control group after drug treatment, and "inhibition of tumor growth" refers to the ratio of the mean value of the volume or weight of the tumor of the treated group to the mean value of the volume or weight of the control group after drug treatment.

In one embodiment, the suggestin is for use in treating cancer in a subject.

The term "cancer" as used herein refers to malignant proliferation of epithelial cells due to alteration of genetic material. The cancer includes: bladder cancer, blood cancer, bone cancer, brain cancer, breast cancer, central nervous system cancer, cervical cancer, colon cancer, endometrial cancer, esophageal cancer, gallbladder cancer, gastrointestinal cancer, genitourinary cancer, head cancer, kidney cancer, laryngeal cancer, liver cancer, lung cancer, muscle tissue cancer, neck cancer, oral or nasal mucosa cancer, ovarian cancer, pancreatic cancer, prostate cancer, skin cancer, spleen cancer, small intestine cancer, large intestine cancer, stomach cancer, testicular cancer, and/or thyroid cancer.

The invention also relates to the combined use of the suogliflozin and other TKI medicines for treating cancers. In one embodiment, the present invention provides the use of suggestin for the manufacture of a medicament for the treatment of cancer in a subject, wherein the medicament is for use in combination with a tyrosine kinase inhibitor-based medicament for the treatment of cancer.

The term "TKI-type drug" as used herein includes drugs known in the art that can be used for the treatment of cancer, including at least one of EGFR inhibitors, c-Kit, c-Met, c-Ret, Raf, PDGFR, BTK, PKA/C, FGFR inhibitors, VEGFR inhibitors.

The composition provided by the invention comprises sugelliflozin and TKI medicines.

In one embodiment, the composition consists of suogramazin and gefitinib, wherein the molar ratio of suogramazin to gefitinib is (10-30) to (10-60). Specifically, the mol ratio of the sugelliflozin to the gefitinib is 10: 10, 10: 20, 10: 30, 10: 50, 10: 60, 20: 10, 20: 20, 20: 30, 20: 50, 20: 60, 30: 10, 30: 20, 30: 30, 30: 50 and 30: 60.

In one embodiment, the composition consists of suograpzin and apatinib, wherein the molar ratio of the suograpzin to the apatinib is (10-40) to (5-40). Specifically, the mol ratio of the suggestin to the apatinib is 10: 5, 10: 10, 10: 20, 10: 30, 10: 40, 20: 5, 20: 10, 20: 20, 20: 30, 20: 40, 30: 5, 30: 10, 30: 20, 30: 30, 30: 40, 40: 5, 40: 10, 40: 20, 40: 30 and 40: 40; or the molar ratio of the Soxhlet to the apatinib is (10-40) to (5-60). Specifically, the molar ratio of the suggestin to the apatinib is 10: 5, 10: 10, 10: 20, 10: 30, 10: 40, 10: 60, 20: 5, 20: 10, 20: 20, 20: 30, 20: 40, 20: 60, 30: 5, 30: 10, 30: 20, 30: 30, 30: 40, 30: 60, 40: 5, 40: 10, 40: 20, 40: 30, 40: 40, 40: 60.

In one embodiment, the composition consists of sugelliflozin and lenvatinib, wherein the molar ratio of sugelliflozin and lenvatinib is 20: 1.

In another embodiment, the composition consists of sugelliflozin and drug a, wherein drug a is: lapatinib ditosylate, genistein, lapatinib, sapertinib, daparinib, dasatinib, tilinib, erlotinib hydrochloride, osetinib mesylate, osetinib, pozzitinib, natatinib, azatinib, AZD3759, imatinib, ibrutinib, lenatinib, lazertinib, axitinib, nidanib, cedanib, cabotinib, alanatinib, lenvatinib, regorafenib, ENMD-6, ENMD-2076 tartrate, tevozanib, panatinib, furalatinib, tematinib, taxifolin, pazopanib, cabotinib malate, vitamin E, regorafenib hydrate, nilapanib ethanesulfonate, nilapanib mesylate, cilatinib maleate, 4- [ (1R) -1- [ (1R) -2- [ (1R) -1- [ (1-5- [ (1R) -1- [ (1-), 5-dichloro-4-pyridyl) ethoxy ] -1H-indazol-3-yl ] vinyl ] -1H-pyrazole-1-ethanol, sunitinib, cetraratinib, ariotinib, sorafenib and vandetanib.

The test materials adopted by the invention are all common commercial products and can be purchased in the market.

In the embodiments of the present application, the related drugs and their english names are listed in table 1:

TABLE 1 drugs and their English names

The invention is further illustrated by the following examples:

example 1 combination of Soxhlet with gefitinib

1. Sogelliflozin safety concentration determination

Killing effect of gefitinib on lung cancer A549 cells cultured in a sugar-free culture medium and a sugar-containing culture medium; after 48 hours of treatment of a549 cells with 20uM gefitinib, trypan blue staining was used to distinguish live cells from dead cells, which were stained blue with trypan blue, and the results are shown in fig. 1. According to the earlier findings illustrated in FIG. 1, the in vitro inhibition of cancer cell growth of suggestin purchased from Selleck inhibitors was tested by first testing suggestin using normal human umbilical epithelial cells at concentrations above 80 μ M for significant cytotoxicity, and below 80 μ M for substantial inhibition of cell growth, so that the subsequent compositions used in this experiment all used suggestin at concentrations below 80 μ M to prevent effects on normal cells.

2. Tumor cell inhibiting effect of combined medicine

Selecting a lung cancer cell line A549 according to tissue distribution characteristics of gefitinib and suogliflozin drug targets EGFR and SGLT 1/2; colorectal cancer cell lines LoVo, HT29, SW620, HCT 116; cervical cancer HeLa; ovarian cancer SKOV 3; gastric cancer NGC 27; bile duct cancer RBE; the experiment of the esophageal cancer KYSE30 and the like is verified. The method comprises the steps of digesting cells by trypsin when the cells grow to 80% density, carrying out passage and paving the cells in a 96-well plate, changing the culture medium containing the drug with corresponding concentration after 24 hours, and detecting the light absorption value under each concentration by using an MTT method after 48 hours.

The experiment is divided into:

control group: no drug is added, and only normal culture medium is used for culturing cells;

gefitinib test group: cells were treated with gefitinib alone in the medium, and 4 treatments were set at different concentrations, 5. mu.M, 10. mu.M, 20. mu.M, and 30. mu.M, respectively.

Gefitinib + suogliflozin combination test group: cells were treated with addition of sugelliflozin (20 μ M) and gefitinib at 4 different concentrations, 5 μ M, 10 μ M, 20 μ M, 30 μ M gefitinib.

After the culture, the cell growth inhibition rate was calculated by dividing the absorbance at each concentration by the absorbance of the control, and the results are shown in FIGS. 2-a to 2-e. In the figure, the concentration of gefitinib is used as a horizontal coordinate, and the cell growth inhibition rate is used as a vertical coordinate, so that the result shows that the inhibition effect of the gefitinib alone on tumor cells is limited, and the combination of the gefitinib and the gefitinib is beneficial to improving the inhibition effect on tumors.

3. Determination of IC50 value of combination drug

Taking the lung cancer cell line A549 as an example, the IC50 value of the gefitinib and suggestin combined drug is verified, and the experiment is divided into:

gefitinib test group (fig. 2-a-1): and only adding gefitinib into the culture medium to treat the cells, and detecting the cell survival rate after incubating for 1 h. 6 concentration gradients were set at 0. mu.M, 10. mu.M, 20. mu.M, 30. mu.M, 40. mu.M, 50. mu.M, respectively. The results showed that gefitinib had an IC50 value of 24.42 μ M for a549 cells.

Sugelliflozin test group (fig. 2-a-2): cells were treated with sugelliflozin only in the culture medium and the cell viability was measured after 1h incubation. 6 concentration gradients were set at 0. mu.M, 10. mu.M, 30. mu.M, 40. mu.M, 50. mu.M, 60. mu.M, respectively. The results showed that the IC50 value for sugelliflozin against a549 cells was 73.04 μ M.

Gefitinib + sugelliflozin combination test group 1 (fig. 2-a-3): cells were treated with sugelliflozin (10 μ M) and gefitinib added to the medium and assayed for cell viability after 1h of incubation. Gefitinib was set for 6 different concentration treatments, with gefitinib at 0 μ M, 10 μ M, 20 μ M, 30 μ M, 50 μ M, 60 μ M, respectively. The results showed an IC50 value of 17.03 μ M for a549 cells from this test group.

Gefitinib + sugelliflozin combination test group 2 (fig. 2-a-4): cells were treated with sugelliflozin (20 μ M) and gefitinib added to the medium and assayed for cell viability after 2h incubation. Gefitinib was set for 5 different concentration treatments, with gefitinib at 0 μ M, 10 μ M, 20 μ M, 30 μ M, 40 μ M, respectively. The results showed an IC50 value of 12.71 μ M for a549 cells from this test group.

Gefitinib + suogliflozin combination test group 3 (fig. 2-a-5): cells were treated with sugelliflozin (30 μ M) and gefitinib added to the medium and assayed for cell viability after 1h of incubation. Gefitinib was set for 6 different concentration treatments, with gefitinib at 0 μ M, 10 μ M, 20 μ M, 30 μ M, 40 μ M, 50 μ M, respectively. The results showed that the IC50 value for this test group for a549 cells was 9.318 μ M.

The result shows that with the use of the combination of the suogrel and the gefitinib, the inhibition rate of the gefitinib on the A549 cells is obviously enhanced, and the IC50 is reduced to be less than half of that of a single drug, so that the combined effect within the safe concentration range of the suogrel is better than the effect of each single drug. The results of the verification of other cell lines are shown in the attached drawings (2f-2j), and it is worth mentioning that the effect of the combination of sugelliflozin for enhancing the effect of targeting EGFR TKI is not limited to gefitinib alone, in FIG. 2k, FIG. 21, the invention further verifies afatinib and erlotinib which are two other EGFR inhibitors by taking A549 cells as an example, and the result shows that the addition of safe dose of sugelliflozin can effectively reduce the IC50 value of afatinib and erlotinib.

Example 2 combination of Soxhlet and Gefitinib to reverse Gefitinib resistance in tumor cells

1. And (4) screening gefitinib resistant cell strains.

After the gefitinib and sueglin combination in example 1 is obtained to significantly enhance the effectiveness of gefitinib, the present invention continues to explore whether the combination can reverse the gefitinib resistance of tumor cells. The A549 cells are cultured for a long time by using a culture medium containing gefitinib with gradually increased concentration, so that the cells can obtain drug resistance to the gefitinib, and the invention obtains an A549 gefitinib drug-resistant cell strain which can survive in 60uM gefitinib for a long time through 5 months of screening.

2. The combination of sugelliflozin and gefitinib reverses the resistance of tumor cells to gefitinib.

The gefitinib-resistant cell strain obtained by the invention can still be effectively killed by adding 30uM gefitinib and the sugelliflozin composition. It is shown that the combination of sugelliflozin and gefitinib reverses the resistance of tumor cells to gefitinib, and the results are shown in fig. 3.

Example 3 combination of Soxhlet with Apatinib

1. Tumor cell inhibiting effect of combined medicine

After the effectiveness of gefitinib in example 1 is obtained, the invention further verifies the other type of VEGFR-targeted TKI drug apatinib, and the liver cancer cell line HepG2 is preferentially selected according to the tissue distribution characteristics of the target points VEGFR and SGLT1/2 of the apatinib and the sugrezin drugs; colorectal cancer cell lines LoVo, HT29, SW620, SW 480; cervical cancer HeLa; ovarian cancer SKOV 3; gastric cancer NGC 27; bile duct cancer RBE; the experiment of the esophageal cancer KYSE30 and the like is verified. The method comprises the steps of growing cells to 80% density, digesting the cells with trypsin, carrying out passage and paving the cells in a 96-well plate, changing the cells into a culture medium containing apatinib, sueggliflozin and a combined drug of apatinib and sueggliflozin with corresponding concentrations after 24 hours, and detecting the absorbance value under each concentration by using an MTT method after 48 hours.

The experiment is divided into:

control group: no medicine is added, and only a normal culture medium is suitable for culturing cells;

apatinib test group: the cells were treated with apatinib alone in the medium, setting 4 different concentrations of treatment, 5 μ M, 10 μ M, 20 μ M, 30 μ M.

Apatinib + suggestin combination test group: cells were treated with addition of suggestin (20 μ M) and apatinib to the medium, setting 4 different concentrations of treatment, wherein apatinib was 5 μ M, 10 μ M, 20 μ M, 30 μ M, respectively.

The cell growth inhibition was calculated by dividing the absorbance at each concentration by the absorbance of the control, and the results are shown in FIGS. 4-a-1 to 4-a-5. The results of the figure, which uses the concentration of apatinib as the abscissa and the cell growth inhibition rate as the ordinate, show that the inhibition effect of apatinib on tumor cells is limited when the apatinib is used alone, and the combination of the two medicines is beneficial to improving the inhibition effect on tumors.

2. Determination of IC50 value of combination drug

2.1, taking the liver cancer cell line HepG2 as an example, the IC50 value of the apatinib and suggestin combination is verified, and the experiment is divided into:

apatinib test group (FIG. 4-b-1): only apatinib is added into the culture medium to treat the cells, and the cell survival rate is detected after the cells are incubated for 2 hours. 6 concentration gradients were set at 0. mu.M, 5. mu.M, 10. mu.M, 20. mu.M, 30. mu.M, 40. mu.M, respectively. The results showed that apatinib had an IC50 value of 46.02 μ M against HepG2 cells.

Sugelliflozin test group (fig. 4-b-2): cells were treated with sugelliflozin only in the culture medium and assayed for cell viability after 2h incubation. 7 concentration gradients were set, 0. mu.M, 20. mu.M, 30. mu.M, 40. mu.M, 60. mu.M, 80. mu.M, 100. mu.M, respectively. The results showed that the IC50 value of sugelliflozin to HepG2 cells was 115.7 μ M.

Apatinib + suggestin combination test group 1 (fig. 4-b-3): cells were treated with sugelliflozin (10 μ M) and apatinib in culture medium and assayed for cell viability after 2h incubation. Apatinib was set for 6 different concentration treatments, with 0 μ M, 5 μ M, 10 μ M, 20 μ M, 30 μ M, 40 μ M for Apatinib, respectively. The results showed that the IC50 value of this test group to HepG2 cells was 33.3. mu.M.

Apatinib + suggestin combination test group 2 (fig. 4-b-4): cells were treated with sugelliflozin (20 μ M) and apatinib in culture medium and assayed for cell viability after 2h incubation. Apatinib was set for 6 different concentration treatments, with 0 μ M, 5 μ M, 10 μ M, 20 μ M, 30 μ M, 40 μ M for Apatinib, respectively. The results showed that the IC50 value of this test group to HepG2 cells was 29.69. mu.M.

Apatinib + suggestin combination test group 3 (fig. 4-b-5): cells were treated with sugelliflozin (30 μ M) and apatinib in culture medium and assayed for cell viability after 2h incubation. Apatinib was set for 6 different concentration treatments, with 0 μ M, 5 μ M, 10 μ M, 20 μ M, 30 μ M, 40 μ M for Apatinib, respectively. The results showed that the IC50 value of this test group to HepG2 cells was 13.13. mu.M.

Apatinib + suggestin combination test group 4 (fig. 4-b-6): cells were treated with sugelliflozin (40 μ M) and apatinib in culture medium and assayed for cell viability after 2h incubation. Apatinib was set for 6 different concentration treatments, with 0 μ M, 5 μ M, 10 μ M, 20 μ M, 30 μ M, 40 μ M for Apatinib, respectively. The results showed that the IC50 value of this test group to HepG2 cells was 10.89. mu.M.

The result shows that with the use of the combination of soxhlet and apatinib, the inhibition rate of apatinib on HepG2 cells is remarkably enhanced, IC50 is reduced to be less than one fourth of that of a single drug, so the verification result that the combined effect of the combination of the soxhlet in a safe concentration range is better than that of other cell lines with respective single drug effects is shown in the attached drawing (4c-4g), and the fact that the capability of the combination of the soxhlet in enhancing the drug effect of the VEGFR-based TKI is not limited to the single drug of the apatinib is worth proposing, in the figure 5, another inhibition lenvatinib of VEGFR is further verified in a plurality of cell lines, and the result shows that the addition of the safe dose of the soxhlet in can effectively enhance the inhibition effect of lenvatinib on the cell lines.

Example 4

The other TKI drugs were further validated to demonstrate that the suogliflozin combination TKI drug is not a specific drug or drugs.

The selected medicines comprise: axitinib (fig. 6-1), nintedanib (fig. 6-2), cediranib (fig. 6-3), pazopanib hydrochloride (fig. 6-4), sunitinib malate (fig. 6-5), brimonib (fig. 6-6), cabozantinib (fig. 6-7), alanine brimonib (fig. 6-8), lenvatinib (fig. 6-9), regorafenib (fig. 6-10), ENMD-2076 (fig. 6-11), tivozanib (fig. 6-12), panatinib (fig. 6-13), ENMD-2076 tartrate (fig. 6-14), tiratinib (fig. 6-15), pinostin (fig. 6-16), pazopanib (fig. 6-17), cabozantinib malate (fig. 6-18), vitamin E (fig. 6-19), Regorafenib hydrate (FIGS. 6-20), nintedanib ethanesulfonate (FIGS. 6-21), lenvatinib mesylate (FIGS. 6-22), cediranib maleate (FIGS. 6-23), 4- [ (1E) -2- [5- [ (1R) -1- (3, 5-dichloro-4-pyridinyl) ethoxy ] -1H-indazol-3-yl ] vinyl ] -1H-pyrazol-1-ethanol (FIGS. 6-24), sunitinib (FIGS. 6-25), cetraratinib (FIGS. 6-26), arotinib (FIGS. 6-27), sorafenib (FIGS. 6-28), vandetanib (FIGS. 6-29), furoquintinib (FIGS. 6-30), The pharmaceutical composition comprises the following components, i.e., the following components, and the following components, i.e., the following components, imatinib (fig. 6-31), the same components, i.e., the same components, and the like (fig. 6-32), genistein (fig. 6-33), ibrutinib (fig. 6-34), dactinib (fig. 6-35), osetinib mesylate (fig. 6-36), daparinib (fig. 6-37), tilitinib (fig. 6-38), AZD3759 (fig. 6-39), and the same components, and the like (fig. 6-40), and lidocaine hydrochloride (fig. 6-41), and the like (fig. 6-42), and the like.

The invention preferentially selects the liver cancer cell line HepG2 for experimental verification. The method comprises the steps of growing cells to 80% density, digesting the cells with trypsin, carrying out passage and paving the cells in a 96-well plate, changing the cells into a culture medium containing apatinib, sueggliflozin and a combined drug of apatinib and sueggliflozin with corresponding concentrations after 24 hours, and detecting the absorbance value under each concentration by using an MTT method after 48 hours.

The experimental method is the same as before, and the incubation time is 1 h. The experiment shows that the blank control group, namely the HepG2 cells cultured normally, wherein the concentration of the sugelliflozin or the TKI medicament is 0, the survival rate of the cells of the blank control group is 100%, the concentration of the sugelliflozin adopted in other administration groups is 30 mu mol/L, the concentration of the TKI medicament is divided into the experiment groups shown in the table 2, and the results are shown in the accompanying drawings:

TABLE 2 administration group Experimental design

The results show that in each drug combination group, the drug combination group shows good inhibition effect on tumor cells, and the effect of the drug combination group is obviously superior to that of a control group which is singly administrated.

Example 5

Examples 1 to 4 are all thinIn order to further verify that the diabetes treatment drug suggestin and TKI inhibitor drugs discovered by the invention have effects in-vivo anti-tumor, the lung cancer A549 cell is selected and subjected to tumor-bearing treatment test by using Balbc nude mice produced by Witongli Hua. The cells are grown to the logarithmic phase, and A549 cells are collected and are resuspended to be 5 multiplied by 10 by serum-free DMEM medium7Each mouse was inoculated with 100. mu.l of 5X 10 cells per ml6Individual cells, tumor size was measured 19 days later and grouped by size, with the average size of tumors in each group being consistent. The administration is started after grouping, the administration dosage of the sugelliflozin is 200-400 mg according to the current sugelliflozin recommended daily administration dosage of diabetic patients, the administration dosage is selected by converting the dosage into the dosage of 22-44 mg per kg of body weight of mice, and finally, the administration of the sugelliflozin is carried out by selecting the dosage of 30 mg per kg in an oral administration mode. Gefitinib was administered at a dose of 100 mg per kg, determined according to previous studies to treat a549 transplantable tumors. The administration mode of the two medicines is consistent with the oral administration mode in the current clinic, and the administration is carried out by intragastric administration of mice. The dosing period was once every two days. Tumor size was measured every two days. After 40 days of administration, the test is ended and the tumor is dissected and weighed.

TABLE 3 summary of the therapeutic effects of suogliflozin in combination with gefitinib on A549 cell-bearing tumor mice

Tumor volume mean value Ratio of each group to control group Tumor growth inhibition rate p value
Solvent control group 1034.278808
Suogelijing 430.2976498 0.416036417 58.39635826 0.002
Gefitinib 504.8082165 0.488077502 51.19224984 0.0012
Sogelliflozin in combination with gefitinib 211.0447064 0.204050112 79.59498882 0.0001

As shown in FIGS. 7-a-7-b-7-c, the single drugs of both sugliflozin and gefitinib can inhibit tumor growth (FIGS. 7-a-7-b-7-c), and the combination of sugliflozin and gefitinib has better tumor inhibition effect than the two drugs used alone. According to the evaluation of the effectiveness of the medicine, the requirement that the tumor growth inhibition rate is more than 60 percent and the p value is less than 0.05 is met. By calculating the tumor growth inhibition rate and the p value of each group, the single drug administration of the sugelliflozin and the gefitinib is judged to be ineffective finally, and the treatment effect of the sugelliflozin and gefitinib composite is effective.

The foregoing is only a preferred embodiment of the present invention, and it should be noted that it is obvious to those skilled in the art that various modifications and improvements can be made without departing from the principle of the present invention, and these modifications and improvements should also be considered as the protection scope of the present invention.

72页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:一种JO蛋白锚定的载药免疫细胞及其制备方法和应用

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!