CN110856717A - Tumor immunomodulator and application thereof - Google Patents

Tumor immunomodulator and application thereof Download PDF

Info

Publication number
CN110856717A
CN110856717A CN201810887556.5A CN201810887556A CN110856717A CN 110856717 A CN110856717 A CN 110856717A CN 201810887556 A CN201810887556 A CN 201810887556A CN 110856717 A CN110856717 A CN 110856717A
Authority
CN
China
Prior art keywords
tumor
immunomodulator
group
mice
derivative
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201810887556.5A
Other languages
Chinese (zh)
Inventor
尹汉维
曾慧慧
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.)
Shanghai Yuanxi Pharmaceutical Technology Co Ltd
Original Assignee
Shanghai Yuanxi Pharmaceutical 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 Shanghai Yuanxi Pharmaceutical Technology Co Ltd filed Critical Shanghai Yuanxi Pharmaceutical Technology Co Ltd
Priority to CN201810887556.5A priority Critical patent/CN110856717A/en
Publication of CN110856717A publication Critical patent/CN110856717A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants

Landscapes

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

Abstract

The invention belongs to the technical field of tumor treatment, and discloses a tumor immunomodulator and application thereof. The immunomodulator at least comprises a benzisoselenazole derivative, preferably comprises a benzisoselenazole derivative and a platinum anti-cancer drug, and the molar ratio of the benzisoselenazole derivative to the platinum anti-cancer drug is (1-99) to (1-99). The immunomodulator can be used for tumor therapy, such as early tumor immunomodulator and postoperative tumor recurrence immunomodulator. The BS has the coordination of apoptosis induction and immunoregulation, so that the BS can be used as a postoperative tumor recurrence control regulator which is used for a long time and benefits the organism of a patient. Further, the use of BS in combination with cisplatin amplifies this coordination and thereby benefits more patients clinically.

Description

Tumor immunomodulator and application thereof
Technical Field
The invention belongs to the technical field of tumor treatment, and particularly relates to a tumor immunomodulator and application thereof.
Background
Tumor cells evade recognition and attack by the body's immune system through various mechanisms, resulting in a phenomenon of survival and proliferation in vivo, called tumor immune escape. The immune system of the body has an immune monitoring function, and when malignant cells appear in the body, the immune system can recognize and specifically eliminate the non-self cells through an immune mechanism to resist the occurrence and development of tumors. However, malignant cells can in some cases escape immune surveillance of the body by multiple mechanisms, proliferate rapidly in vivo, and form tumors. That is to say: on the one hand, the body can resist the development of tumors by natural and acquired immunity; on the other hand, tumor cells can evade recognition and attack by the body's immunity through a variety of mechanisms. Whether a tumor occurs or not and how it is classified depends on the overall effect of both aspects. The tumor immune escape mechanism mainly has the following aspects: firstly, low immunogenicity; secondly, the antigen is identified as self-antigen; thirdly, antigen modulation; fourthly, immunosuppression induced by tumor; and fifthly, inducing the tumor to generate a exempted area.
The existing tumor treatment means comprise surgery treatment, chemotherapy, radiotherapy, immunotherapy and other treatment means, wherein the surgery, the radiotherapy and the chemotherapy are three main roles which cannot be shocked by the tumor treatment, but the immunity of the organism is reduced and the immunity of the organism is inhibited in a more serious way in the processes of the surgery, the radiotherapy and the chemotherapy, so that residual tumor cells can be continuously and further transferred, and the fatal threat is brought to tumor patients. Especially, the operation is used as a mechanical means, the local treatment is thorough, but the cancer cell is dropped after the cancer is attacked due to the metastasis of the cancer cell, and the cancer cell is proliferated or metastasized to the periphery and far through local diffusion, blood vessels, lymphatic vessels and other ways. Furthermore, the patient's own immunity is in a very weak state for a long time after the operation, and the normal immunity of the body cannot be exerted, so how to improve the immunity of the body and stabilize the immune system of the body after the operation is a difficult problem of controlling the recurrence of the tumor after the operation. According to the monitoring data of the cancer condition of the disease pre-control center in China, the recurrence and metastasis rate of cancer patients in China after operation for 3 months is 50%, the recurrence and metastasis rate of cancer patients in China after operation for 6 months is as high as 69%, and the recurrence or metastasis rate of cancer patients after operation for five years is as high as more than 90%.
After the 21 st century, with the development of oncology and immunology, the treatment of tumors around the human immune system is gradually accepted by the scientific research and industrial fields, and becomes a popular field for the research and development of new drugs. Tumor immunotherapy (I/O) refers to a method for effectively treating a tumor patient by directly or indirectly using the human immune system during the course of treatment. For example, killing or inhibiting tumors is achieved by interfering with the direct antitumor effects of cell overgrowth, transformation and metastasis or activating effector cells of the immune system and factors secreted by them.
Therefore, the development of an immunomodulator for postoperative tumor recurrence, which can improve the immunity of the organism, especially the postoperative tumor organism immunity, has low toxicity and long-term use, and can benefit the organism of a patient, is a technical problem to be solved urgently.
Disclosure of Invention
The invention provides a tumor immunomodulator, which at least comprises a benzisoselenazole derivative.
According to the invention, the benzisoselenazole derivative has a structure of a compound shown as a formula A, and is selected from at least one of the compound shown as the formula A, a precursor, an active metabolite, a stereoisomer, a pharmaceutically acceptable salt, a prodrug and a solvate thereof,
Figure BDA0001756036070000021
wherein R is1、R2Identical or different, independently of one another, from H or the following radicals: c1-12Alkyl radical, C3-20A cycloalkyl group; preferably, it is selected from H, or the following groups: c1-6Alkyl radical, C3-10A cycloalkyl group; illustratively, R1、R2Is selected from H.
Wherein R is selected from C1-12Alkylene, phenylene, biphenylene, triphenylene, or
Figure BDA0001756036070000022
Wherein M represents Pt, Pd orRh; preferably, R represents C1-6Alkylene radicals, e.g. C1-4Alkylene, for example R is butylene.
Preferably, in the compound shown in the formula A, R1、R2Identical or different, independently of one another, from H, -CH3、-CH2CH3、-CH(CH3)2、-C(CH3)3、-CH(CH2)4or-CH (CH)2)5(ii) a R is selected from-CH2-、-C2H4-、-C4H8-, phenylene-C6H4-。
According to an exemplary embodiment of the present invention, the benzisoselenazole derivative is selected from 1, 2-bis [2- (1, 2-benzisoselenazol-3 (2H) -one) ] -Butane (BS), the structure of which is shown below:
Figure BDA0001756036070000023
according to the present invention, the tumor immunomodulator comprises a benzisoselenazole derivative and a platinum anticancer drug. Specifically, the tumor immunomodulator is formed by combining a benzoisoselenazole derivative and a platinum anti-cancer drug properly, and shows an immunoregulation effect.
The platinum anticancer drug is at least one selected from Cisplatin (CDDP), Carboplatin (CBP), oxaliplatin (1-OHP), nedaplatin (CDGP) and the like; illustratively, the platinum-based anti-cancer drug is selected from Cisplatin (CDDP).
According to the invention, the molar ratio of the benzisoselenazole derivative to the platinum anticancer drug is (1-99) to (1-99); for example, the molar ratio is (50-90): (1-30), (60-70): 1-10), (1-40): 1-40), (1-25): 1-25), (1-10): 1-10), (1-6): 1-6); illustratively, the molar ratio is 1:1, 2:1 (in vitro combination) or 60:1, 70:1, 80:1, etc. (in vivo combination). Specifically, when the molar ratio of the benzisoselenazole derivative to the platinum-based anticancer drug is not very different, such as the molar ratio of (1-10): 1-10), (1-6): 1-6), and exemplarily, the molar ratio is 1:1 and 2:1, the benzisoselenazole derivative and the platinum-based anticancer drug are used together as an immunomodulator and preferably can be applied to in vitro tumor cells. In vivo combination, because the in vivo metabolism of the drug to reach the tumor site concentration has a great relationship with in vivo properties, the dosage of the oral benzisoselenazole derivative is far greater than that of the platinum anticancer drug (conventional injection administration), i.e. when the dosage of the benzisoselenazole derivative is far greater than that of the platinum anticancer drug, for example, the molar ratio is (50-90): 1-30), (60-70): 1-10, exemplarily, the molar ratio is 60:1, 70:1, and 80:1 (in vivo combination), the benzisoselezole derivative and the platinum anticancer drug are used as immunomodulators, and preferably, the benzisoselezole derivatives can be applied to in vivo tumor cells.
Preferably, the tumor immunomodulator may further optionally comprise at least one pharmaceutically acceptable adjuvant.
Preferably, the pharmaceutically acceptable excipients are various excipients commonly used or known in the pharmaceutical field, including but not limited to: diluents, binders, antioxidants, pH adjusters, preservatives, lubricants, disintegrants, and the like.
For example, the diluent is selected from lactose, starch, cellulose derivatives, inorganic calcium salts, sorbitol and the like. The binder is, for example: starch, gelatin, sodium carboxymethylcellulose, polyvinylpyrrolidone, and the like. For example, the antioxidant is selected from vitamin E, sodium bisulfite, sodium sulfite, butylated hydroxyanisole, and the like. For example, the pH adjusting agent is selected from hydrochloric acid, sodium hydroxide, citric acid, tartaric acid, Tris, acetic acid, sodium dihydrogen phosphate, disodium hydrogen phosphate, and the like. For example, the preservative is selected from methyl paraben, ethyl paraben, m-cresol, benzalkonium chloride, and the like. For example, the lubricant is selected from magnesium stearate, aerosil, talc, and the like. The disintegrant is, for example: starch, methyl cellulose, xanthan gum, croscarmellose sodium, and the like.
The formulation of the immunomodulator may be in the form of an oral preparation, such as tablets, capsules, pills, powders, granules, suspensions, syrups, and the like; it can also be administered by injection, such as injection solution, powder for injection, etc., by intravenous, intraperitoneal, subcutaneous or intramuscular route. All dosage forms used are well known to those of ordinary skill in the pharmaceutical arts.
Furthermore, the invention also provides the application of the immunomodulator in tumor treatment.
Preferably, the immunomodulator is used in an amount of 10-500 mg/kg based on the weight of a subject (such as a mouse); for example, 50 to 400 mg/kg; illustratively, the amount is 90, 180, 360 mg/kg.
Preferably, the immunomodulator can be selected from benzisoselenazole derivatives, and benzisoselenazole derivatives and platinum anticancer drugs for combined application. When the immunomodulator is combined with the benzisoselenazole derivative and the platinum anticancer drug, the dosage of the platinum anticancer drug is 0.1-5 mg/kg based on the weight of an administration object (such as a mouse); for example, 0.5 to 4mg/kg, 1 to 3 mg/kg; illustratively, the amount is 2 mg/kg.
In another example, the immunomodulator is used for inhibiting the expression of PD-L1 protein on the surface of tumor cells and promoting the secretion of cytokines (such as IFN-gamma, IL-2 and TNF- α), and in another example, the immunomodulator is used for promoting lymphocyte CD8 +An increase in the percentage of subpopulation positive cells; as another example, the immunomodulator is used to promote secretion of interferons (e.g., granzyme B, Gz-B).
According to another embodiment of the present invention, the immunomodulator can also be used for immunomodulation of postoperative tumor recurrence. Illustratively, the tumor immunomodulator is used for promoting the secretion of immune active substances (such as perforin and granzyme B), and improving the killing activity of cytotoxic T lymphocytes and NK cells; as another example, the tumor immunomodulator is used to promote lymphocyte proliferation; in another example, the tumor immunomodulator is used to upregulate expression of a lymphocyte-surface NKG2D activating receptor, preferably, expression of a lymphocyte-surface NKG2D ligand RAE-1.
According to the invention, the tumors include solid tumors and non-solid tumors, and may be benign and malignant. For example, the tumors include, but are not limited to: liver cancer, lung cancer, breast cancer, colon cancer, nasopharyngeal cancer, gastric cancer, skin cancer, bladder cancer, ovarian cancer, prostate cancer, bone cancer, brain cancer, rectal cancer, esophageal cancer, tongue cancer, lymph cancer, oral epithelial cancer, epithelial cervical cancer or chronic myelogenous leukemia. Specifically, tumor cells can be selected, and the tumor cells include human cancer cells and mouse cancer cells. Such human cancer cells include, but are not limited to: human liver cancer cell HepG2, human liver cancer cell Be17402, human liver cancer cell Huh7721, human colorectal cancer cell LoVo, human colorectal cancer cell RKO, human colorectal cancer cell SW480, human lung cancer cell A549, human lung cancer cell H1299, human lung cancer cell SPCA-1, human epithelial cervical cancer cell HeLa, human breast cancer cell MCF-7, human chronic myelogenous leukemia cell k562, human esophageal cancer cell KYSE150, human esophageal cancer cell KYSE450 or human esophageal cancer cell KYSE 510. The mouse cancer cells include, but are not limited to: mouse liver cancer cell H22, mouse liver cancer cell Hepa 1-6, mouse erythroleukemia cell MEL, mouse kidney cancer cell Renca, mouse lymphoma cell EL-4, mouse lung cancer cell Lewis, and mouse breast cancer cell 4T 1. Illustratively, mouse liver cancer cells H22, mouse lung cancer cells Lewis and mouse breast cancer cells 4T1 are selected.
Further, the present invention also provides a method of treating tumor or postoperative tumor recurrence by the above-mentioned tumor immunomodulator, by administering a therapeutically effective amount of the immunomodulator to an individual in need thereof.
Preferably, the subject may be a mammal, such as a human or a mouse.
Preferably, the tumor has the meaning as described above, preferably liver cancer, lung cancer, breast cancer.
The invention has the beneficial effects that:
the invention unexpectedly discovers that the BS has extremely low toxicity, can obviously improve the toxic damage of the drug to the organism, can also promote the coefficient of immune organs (such as spleen, thymus and the like) to be increased, and can promote the increase of the number of peripheral blood leukocytes, for example, can obviously inhibit the expression of PD-L1 protein on the surface of tumor cells, promote the secretion of cytokines (such as IFN-gamma, IL-2 and TNF- α), and can promote cytotoxic T lymphocyte CD (compact disc) of T lymphocytes8 +Subgroup positiveAn increase in the percentage of cells; can promote the secretion of interferon (such as granzyme B, Gz-B). The BS has the functions of enhancing immunity and weakening toxic injury to organisms, and can improve the cellular immunity level of the organisms when being used as an immunomodulator, thereby inhibiting the proliferation of tumor cells and the relapse of postoperative tumors.
The invention also unexpectedly discovers that when the BS is combined with a platinum anti-cancer drug (particularly cisplatin CDDP), the BS can synergistically promote the secretion of immune active substances (such as perforin and granzyme B) and improve the killing activity of cytotoxic T lymphocytes and NK cells; can promote lymphocyte proliferation; can up-regulate the expression of RAE-1, a lymphocyte surface NKG2D ligand. The BS and the CDDP can synergistically promote the secretion of immune active substances, and can improve the immunity of the organism and inhibit the proliferation of tumor cells and postoperative tumor recurrence as an immunomodulator.
The immune regulation function of the BS and the combined use of the BS and the cisplatin plays an important role in controlling postoperative tumor recurrence. At present, the postoperative recurrence control is realized by directly killing tumors with treatment and/or chemotherapy drugs clinically, so as to control the tumor recurrence. However, the chemical killing drugs used in clinical practice at present may damage the immune function of the body to a certain extent and cause immunosuppressive reactions, for example, adverse reactions (for example, renal toxicity: repeated administration at multiple times and in a short period of time, irreversible renal dysfunction, bone marrow suppression: reduction of leukocytes and/or platelets) generated by the body of a patient after platinum drugs are used may gradually lead to the final body being unable to bear the adverse reactions.
The BS and the cisplatin jointly show that the activity of the BS on NK is improved, the function of the additive pharmacological action is generated on the basis of the apoptosis action of the BS on the inhibition of the tumor growth, the BS medicament has the coordination action of apoptosis induction and immunoregulation and extremely low toxicity of the BS, the toxic injury brought to an organism by long-term administration can be obviously improved, and the BS can be used as a postoperative tumor recurrence control regulator which is used for a long time and benefits the organism of a patient. Further, the use of BS in combination with cisplatin amplifies this coordination and thereby benefits more patients clinically.
Definition and description of terms
Unless otherwise indicated, the definitions of groups and terms described in the specification and claims of the present application, including definitions thereof as examples, exemplary definitions, preferred definitions, definitions described in tables, definitions of specific compounds in the examples, and the like, may be arbitrarily combined and coupled with each other. The definitions of the groups and the structures of the compounds in such combinations and after the combination are within the scope of the present specification.
The term "C1-12Alkyl is understood to preferably mean a straight-chain or branched, saturated monovalent hydrocarbon radical having from 1 to 12 carbon atoms, preferably C1-6An alkyl group. "C1-6Alkyl "is understood to preferably mean a straight-chain or branched, saturated monovalent hydrocarbon radical having 1,2, 3, 4, 5, 6 carbon atoms. The alkyl group is, for example, a methyl group, an ethyl group, a propyl group, a butyl group, a pentyl group, a hexyl group, an isopropyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an isopentyl group, a 2-methylbutyl group, a 1-ethylpropyl group, a 1, 2-dimethylpropyl group, a neopentyl group, a 1, 1-dimethylpropyl group, a 4-methylpentyl group, a 3-methylpentyl group, a 2-ethylbutyl group, a 1-ethylbutyl group, a 3, 3-dimethylbutyl group, a 2, 2-dimethylbutyl group, a 1, 1-dimethylbutyl group, a 2, 3-dimethylbutyl group, a 1, 3-dimethylbutyl group or a 1, 2-dimethylbutyl group. In particular, the radicals have 1,2, 3, 4, 5, 6 carbon atoms ("C)1-6Alkyl groups) such as methyl, ethyl, propyl, butyl, isopropyl, isobutyl, sec-butyl, tert-butyl, more particularly groups having 1,2, 3 or 4 carbon atoms ("C)1-4Alkyl groups) such as methyl, ethyl, n-propyl, isopropyl or butyl.
The term "C3-20Cycloalkyl is understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring having 3 to 20 carbon atoms, preferably "C3-10Cycloalkyl groups ". The term "C3-10Cycloalkyl radicals "shouldIs understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring having 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms. Said C is3-10Cycloalkyl groups may be monocyclic hydrocarbon groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or bicyclic hydrocarbon groups such as decalin rings.
The term "C1-12Alkylene is understood to mean preferably a straight-chain or branched, saturated monovalent hydrocarbon radical having from 1 to 12 carbon atoms, preferably C, with the loss of two hydrogen atoms1-4An alkylene group. "C1-4Alkylene "is understood to preferably mean a straight-chain or branched, saturated monovalent hydrocarbon radical having 1,2, 3, 4 carbon atoms which has lost two hydrogen atoms. The alkylene group is, for example, methylene, ethylene, propylene, butylene.
The term "effective amount" or "therapeutically effective amount" refers to an amount of a compound of the present invention sufficient to effect the intended use, including but not limited to the treatment of a disease as defined below. The therapeutically effective amount may vary depending on the following factors: the intended application (in vitro or in vivo), or the subject and disease condition being treated, such as the weight and age of the subject, the severity of the disease condition and the mode of administration, etc., can be readily determined by one of ordinary skill in the art. The specific dosage will vary depending on the following factors: the particular compound selected, the dosage regimen to be followed, whether to administer it in combination with other compounds, the timing of administration, the tissue to be administered and the physical delivery system carried.
Drawings
FIG. 1 is a graph showing the effect of BS on mouse body weight and tumor volume in example 1;
wherein, A: change in body weight of the mouse; b: mice relative body weight changes. Data are presented as mean ± standard deviation (n ═ 6).
FIG. 2 is a graph showing the tumor growth time and tumor growth rate of each group of mice in example 1.
FIG. 3 is a graph showing tumor growth in the groups of mice in example 1;
wherein (a): photographs of each group of tumors at the end of the experiment; (b) the method comprises the following steps Tumor volume changes in mice of each group. .
FIG. 4 is a graph showing the effect of BS on the inhibition of the expression of PD-L1 in tumor cells in example 1.
FIG. 5 shows lymphocyte CD4 in example 1+、CD8+Graph of percentage of positive cells of subpopulations;
wherein, A: control group (control); b: BSL; c: BSM; d: BSH.
FIG. 6 is a graph showing the change in body weight of mice in example 1;
wherein (a): the weight change of the mice in the administration process; (b) the method comprises the following steps Weight gain during administration in each group of mice.
FIG. 7 shows the change in the body weight of mice in example 2;
wherein (a): the weight change of the mice in the administration process; (b) the method comprises the following steps Weight gain during administration in each group of mice.
FIG. 8 shows the change in the body weight of mice in example 2;
wherein (a): the weight change of the mice in the administration process; (b) the method comprises the following steps Weight gain during administration in each group of mice.
FIG. 9 shows RAE-1 protein expression levels in mouse tumor tissues in example 3;
wherein (a): mouse tumor tissue RAE-1 expression level; (b) the method comprises the following steps Relative expression level of RAE-1. Data are presented as mean ± standard deviation, n ═ 3. P <0.05, indicating a statistical difference compared to the control group.
Detailed Description
The technical solution of the present invention will be further described in detail with reference to specific embodiments. The following examples are merely illustrative and explanatory of the present invention and should not be construed as limiting the scope of the invention. All the technologies realized based on the above-mentioned contents of the present invention are covered in the protection scope of the present invention.
Unless otherwise indicated, the raw materials and reagents used in the following examples are all commercially available products or can be prepared by known methods.
The experimental method comprises the following steps:
1. test drugs, cells and animals:
BS: the chemical name of the product is 1, 2-bis [2- (1, 2-benzisoselenazol-3 (2H) -ketone) ] -butane, which is referred to Chinese patent No. 02158917.8.
Cisplatin (CDDP): purchased from Hai Wei, China (Beijing) Gene science and technology, Inc.
Mouse hepatoma cell line H22: presented by a Chinese toon researcher in the institute of tumor prevention and treatment of Beijing university.
Mouse lung cancer cell line Lewis: purchased from basic medical college of cooperative medical university.
Mouse breast cancer cell line 4T 1: purchased from basic medical college of cooperative medical university.
Kunming inbred mice (KM mice): male, 4 weeks, with a weight of 18-22g, purchased from the experimental animals center of the department of medicine of Beijing university, the license number of experimental animals: SYXK (Kd) 2012 and 0036. The breeding environment is clean, the breeding temperature is 25 +/-2 ℃, the illumination is 12 hours and the darkness is 12 hours after 12 hours, and the food and water supply is sufficient.
Balb/c mice: the experimental animal is purchased from the center of experimental animals of department of medicine of Beijing university, and the production license number SYXK (Jing) 2012-0036 of the experimental animals, wherein the experimental animal is 4 weeks old, the weight of the experimental animal is 16-18 g, and male (lung cancer and postoperative recurrence of liver cancer) and female (postoperative recurrence of breast cancer). The breeding environment is clean, the breeding temperature is 25 +/-2 ℃, the illumination is 12h for 12h, the breeding environment is dark, and the food and water supply is sufficient.
2. The test method comprises the following steps:
mouse H22 liver cancer/Lewis lung cancer/4T 1 breast cancer transplantation tumor excision model
Respectively taking out H22 (liver cancer) cryopreserved cells/Lewis (lung cancer) cryopreserved cells/4T 1 (breast cancer) cryopreserved cells, taking out after the cells become liquid at 37 ℃ for 1-2 min; and (4) centrifuging after resuspension, discarding the supernatant after centrifuging at 1200rpm/min for 2min, resuspending in 1mL serum-free medium, and injecting cells into the abdominal cavity.
Approximately one week later, when the abdomen of the mouse was touched to have a swelling sensation, ascites was extracted. The abdominal cavity outer skin of the mouse is slightly reduced with a small opening by scissors and tweezers, then the two sides of the hemostatic forceps are slightly pulled open to expose the abdomen, and the ascites is extracted by a syringe.
Preparing ascites of Balb/c mouse into single cell suspension, counting the number of living cells by trypan blue dye exclusion method, and ensuring high cell survival rateAt a rate of 95%; injecting the tumor cell suspension into the right axilla of male Balb/c mice with the weight of 16g-18g, 10g each6And (4) cells.
2.1 mouse H22 model for excision of liver cancer transplantable tumor
Balb/c mice subcutaneously transplanted with mouse hepatoma cells H22 were subjected to local tumor resection as follows: when the right axillary tumor of the mouse reaches about 500mm3In this case, 5% chloral hydrate solution (0.08mL/10g) was intraperitoneally injected for anesthesia and hair preparation. After the mice enter the general anesthesia state, a 1-2cm incision is cut near the tumor, the tumor tissue is blunt stripped and cut off 90%, and the residual is about 50mm3Finally, the tumor tissue is sutured by using sterile medical silk threads, and the wound is coated with iodophor for disinfection. The above operations are all completed in a super clean bench. Mice were placed in a clean, warm place and given sterile aqueous glucose for their consumption after awakening. The day of surgery is considered to be 0 days after surgery. After the mice were fully awakened, the mice were randomly divided into 4 groups of 6 mice each, and the drug treatment protocol was as follows:
a blank control group (5% CMC-Na, i.g., q.d.);
BS Low dose group (90 mg. kg)-1,i.g.,q.d.);
BS Medium dose group (180 mg. kg)-1,i.g.,q.d.);
BS high dose group (360 mg. kg)-1,i.g.,q.d.)。
Mice body weight, tumor volume and activity were recorded at intervals during the experiment. The mice were sacrificed eight days after administration, and the eyeballs were removed to obtain blood for the detection of TrxR activity in serum. Tumors, liver, kidney, thymus and spleen were rapidly isolated, observed and weighed. Tumor tissues were kept in liquid nitrogen for subsequent experiments.
2.2 mouse Lewis Lung cancer transplantation tumor excision model
As above, Balb/c mice subcutaneously transplanted with murine lung carcinoma cells Lewis were subjected to local tumor resection as follows: when the right axillary tumor of the mouse reaches about 500mm3In this case, 5% chloral hydrate solution (0.08mL/10g) was intraperitoneally injected for anesthesia and hair preparation. After the mice enter the general anesthesia state, a 1-2cm incision is cut near the tumor, and the tumor group is divided into two groupsThe blunt tissue is stripped off and cut 90% with a residual of approximately 50mm3Finally, the tumor tissue is sutured by using sterile medical silk threads, and the wound is coated with iodophor for disinfection. The above operations are all completed in a super clean bench. Mice were placed in a clean, warm place and given sterile aqueous glucose for their consumption after awakening. The day of surgery is considered to be 0 days after surgery. After the mice were fully awakened, the mice were randomly divided into 4 groups of 6 mice each, and the drug treatment protocol was as follows:
a blank control group (5% CMC-Na, i.g., q.d.);
BS Low dose group (90 mg. kg)-1,i.g.,q.d.);
BS Medium dose group (180 mg. kg)-1,i.g.,q.d.);
BS high dose group (360 mg. kg)-1,i.g.,q.d.)。
Mice body weight, tumor volume and activity were recorded at intervals during the experiment. The mice were sacrificed eight days after administration, and the eyeballs were removed to obtain blood for the detection of TrxR activity in serum. Tumors, liver, kidney, thymus and spleen were rapidly isolated, observed and weighed. Tumor tissues were kept in liquid nitrogen for subsequent experiments.
2.3 mouse 4T1 Breast cancer graft tumor excision model
Meanwhile, Balb/c mice subcutaneously transplanted with mouse mammary cancer cells 4T1 were subjected to local tumor resection as follows: when the right axillary tumor of the mouse reaches about 500mm3In this case, 5% chloral hydrate solution (0.08mL/10g) was intraperitoneally injected for anesthesia and hair preparation. After the mice enter the general anesthesia state, a 1-2cm incision is cut near the tumor, the tumor tissue is blunt stripped and cut off 90%, and the residual is about 50mm3Finally, the tumor tissue is sutured by using sterile medical silk threads, and the wound is coated with iodophor for disinfection. The above operations are all completed in a super clean bench. Mice were placed in a clean, warm place and given sterile aqueous glucose for their consumption after awakening. The day of surgery is considered to be 0 days after surgery. After the mice were fully awakened, the mice were randomly divided into 4 groups of 6 mice each, and the drug treatment protocol was as follows:
a blank control group (5% CMC-Na, i.g., q.d.);
BS inhibitorDosage group (90mg kg)-1,i.g.,q.d.);
BS Medium dose group (180 mg. kg)-1,i.g.,q.d.);
BS high dose group (360 mg. kg)-1,i.g.,q.d.)。
Mice body weight, tumor volume and activity were recorded at intervals during the experiment. The mice were sacrificed eight days after administration, and the eyeballs were removed to obtain blood for the detection of TrxR activity in serum. Tumors, liver, kidney, thymus and spleen were rapidly isolated, observed and weighed. Tumor tissues were kept in liquid nitrogen for subsequent experiments.
2.4 Western Blot method:
extracting total protein of tissue, determining protein concentration (30-50 μ g), adding 5 × loading buffer solution, adding RIPA lysate to make up volume, mixing, and performing denaturation treatment at 95 deg.C for 10 min. After fully and uniformly mixing, adding a clean 1.5mm rubber plate to a proper position, and sealing with n-butanol. Adding 1mL of 1 XSEPARATION GEL buffer solution, and standing at room temperature overnight; inserting a 1.5mm sample adding comb, and standing at room temperature for 2h to wait for gelation.
Pulling out the sample adding comb, and loading the sample by a conventional method; and (4) performing constant voltage electrophoresis at 80V until the front edge of the bromophenol blue enters the separation gel, increasing the voltage to 160V, continuing the constant voltage electrophoresis, and stopping the electrophoresis until the bromophenol blue migrates to the end of the separation gel.
Accurately shearing 6 pieces of filter paper with the same size as the separation gel and a 0.2 mu m PVDF membrane, soaking the PVDF membrane in methanol for 10s, soaking in deionized water for 3min, and soaking in an electrotransformation buffer solution for 3 min;
the electricity changes sandwich and lays by the order of positive pole to negative pole in proper order: one sponge, three pieces of filter paper, a PVDF membrane, separation glue, three pieces of filter paper and one sponge (all soaked in an electric transfer buffer solution in advance), air bubbles are expelled from the layers by a glass rod, and an electric transfer clamp and an ice box are clamped and put into an electric transfer tank;
and (5) placing the electric rotating tank in an ice-water bath, and rotating for 2 hours at a constant current of 250 mA.
After the electrotransfer is finished, taking out the PVDF membrane, and placing the PVDF membrane in a 5% closed state; washing the membrane with TBST for 3 times, sealing the PVDF membrane and a diluted secondary antibody solution with a proper proportion in a hybridization bag, incubating for 1h at room temperature, washing the membrane with TBST for 3 times, and developing;
and opening a gel imaging system, uniformly mixing the solution A and the solution B of the ECL luminous liquid in equal volume, uniformly distributing the mixture on the PVDF membrane, and carrying out exposure analysis.
2.5 ELISA method: detection was performed according to the ELISA commercial kit protocol.
Example 1 Effect of BS on the immune System of mice
The method comprises randomly dividing 24 normal male KM mice with weight of 18-22g into 4 groups, including BS low, medium and high dose groups and control group, and inoculating liver cancer H22 cells (2 × 10)6One/only) was started on day 2. The administration dose is as follows: a blank control group (5% CMC-Na, i.g., q.d.); BS Low dose group (90 mg. kg)-1I.g., q.d.); BS Medium dose group (180 mg. kg)-1I.g., q.d.); BS high dose group (360 mg. kg)-1I.g., q.d). The status of the mice was observed daily for 8 consecutive days.
Hereinafter, BSL, BSM, BSH represent the low, medium and high dosage groups of BS.
1.1 Effect of BS on mouse body weight
As shown in FIG. 1, FIG. 1 summarizes the body weight changes of the mice in each group during the administration. Compared with the control group, the body weight of the mice of the administration group has no obvious difference. At the end of the experiment, the body weights of the mice of the low, medium and high dosage treatment groups of the BS are respectively increased to 1.11, 1.10 and 1.12 times of the initial body weight, which indicates that the BS has no obvious toxic and side effects on the mice.
1.2 influence of BS on mouse tumorigenicity time and tumorigenicity rate
The state and the tumorigenesis of the mice were observed every day, and a tumorigenesis time curve was plotted according to the Kaplan-Meier method, and the results are shown in FIG. 2. After H22 cells are inoculated, tumors begin to appear on the fourth day, the tumor appearance rate of the administration group is obviously lower than that of the control group or equivalent to that of the control group within the same time, and the dosage correlation is shown, which indicates that the BS has a certain inhibition effect on the proliferation of mouse liver cancer H22 cells. All mice in each group had complete tumor formation on day 7, and the tumor incidence rate of each group was 100% at the end, indicating that the inhibition effect of BS was better in the early stage. No significant difference was seen between groups by log rank test P ═ 0.098.
1.3 Effect of BS on mouse tumor volume
As shown in figure 3, the tumor volume increases and decreases with the increase of the administration concentration, a certain dose dependence is shown, the tumor growth trend is obviously slowed down compared with that of a control group in the first 6 days, a better tumor growth inhibition effect is achieved, and the effect of BS in the early treatment stage of the tumor is also shown to be more obvious.
After 8 days of administration, we performed statistics on the tumor volume of the mice, and the tumor volume of each group of mice is shown in table 1. The tumor volumes of the BS middle and high groups are 388.8 +/-69.8 mm respectively3、215.1±42.9mm3The tumor proliferation inhibition rates were 35.5% and 64.3%, which were significantly different from those of the control group.
TABLE 1 tumor volume at the end of the experiment in each group of mice
Figure BDA0001756036070000111
Data are presented as mean ± standard deviation (n ═ 6). P <0.05, indicating a statistical difference compared to the control group.
1.4 Effect of BS on organ coefficients and blood conventions in mice
In order to examine the effect of BS on mice, at the end of the experiment, thymus, spleen, liver and kidney, which are immune organs, were harvested from the mice, observed, weighed, and the number of leukocytes, erythrocytes and platelets in peripheral blood was measured, and the results are shown in table 2. Compared with the control group, the spleen coefficient and the thymus coefficient of the BS administration group are obviously increased, the corresponding peripheral blood leukocyte number is obviously increased, and the significant difference (P is less than 0.05) shows that the BS has certain immunity enhancing function. Meanwhile, the liver coefficient and the kidney coefficient of the mice have no obvious difference between groups, which indicates that the BS has no obvious hepatotoxicity or renal toxicity.
TABLE 2 influence of BS on organ coefficients and blood routine
Figure BDA0001756036070000122
Note: data are presented as mean ± standard deviation (n ═ 6). P <0.05, representing a statistical difference compared to the control group; p < 0.01, representing a statistical difference compared to the control group. BSL, BSM, BSH refer to BS low, medium, high dose groups; RBC: the number of red blood cells; WBC: white blood cell count; PLT: number of platelets.
1.5 BS downregulation of mouse tumor tissue cell surface PD-L1 expression
And (3) detecting whether the BS affects the immune system of the mouse. Firstly, the influence of BS on the expression of PD-L1 protein on the surface of a mouse tumor cell is considered, tumor tissues are ground, counted and stained, a flow cytometer is adopted to detect the change of PD-L1 on the surface of the tumor cell, and the result is shown in figure 4, the expression level of PD-L1 on the surface of the tumor cell is reduced in a dose-dependent manner along with the increase of the dosage of the BS; when the concentration of BS reaches 360mg/kg-1When the expression of PD-L1 on the surface of the tumor cell is reduced by 51.41%, and BS can obviously inhibit the expression of PD-L1 protein on the surface of the tumor cell.
1.6 BS on mouse lymphocyte CD4 +、CD8 +Effect of percentage of subpopulation positive cells
Study of the effects of BS on splenic lymphocytes, we examined lymphocyte CD after BS treatment4 +、CD8 +Change in percentage of positive cells of the subpopulation. Separated tumor-bearing KM mouse spleen lymphocytes are labeled with FITC-labeled CD4 +Monoclonal antibodies and PE-labeled CD8 +Staining with monoclonal antibody, detecting lymphocyte CD by flow cytometry4 +、CD8 +The percentage of positive cells of the subpopulation is shown in figure 5.
The results obtained were subjected to statistical analysis, and as shown in Table 3, spleen lymphocyte CD of BSL group4 +The positive rate is obviously improved compared with the control group (P is less than 0.01); spleen lymphocyte CD of BSM and BSH group8 +The positive rate is obviously improved compared with the control group, and the splenic lymphocyte CD is increased along with the increase of the dosage8 +The content of the positive cells of the subgroup is increased. The above experimental results show that the influence of BS on the immune system is dominantTo act on CD8 +Toxic T cells, can improve the cellular immunity level of the organism. This result is also consistent with the down-regulation of PD-L1 by BS at the molecular level.
TABLE 3 BS on mouse lymphocyte CD4+、CD8+Effect of percentage of subpopulation positive cells
Figure BDA0001756036070000131
Note: data are presented as mean ± standard deviation (n ═ 3). P <0.05, representing a statistical difference compared to the control group; p < 0.01, representing a statistical difference from the control group;#p < 0.01, which indicates a statistical difference from the BSL group;p <0.05, which indicates a statistical difference from the BSM group.
1.7 Effect of BS on the Activity of splenic lymphocytes in mice
It has been reported in the literature that when PD-L1 is activated, the expression of various cytokines such as IFN-. gamma.and IL-2 is reduced. We have observed a decrease in the expression of PD-L1 on the surface of tumor cells, and to further confirm that BS is mediated by the immunomodulating action exerted by PD-L1, we extracted splenic lymphocytes from various groups of tumor-bearing mice, administered ionomycin and PMA at 20ng/mL each-1The activation of lymphocytes was observed, the supernatant was collected after 48 hours, and the secretion of the lymphokines IFN-. gamma.IL-2 and TNF- α was examined by ELISA method, and the results are shown in Table 4.
After the ionomycin and PMA are stimulated for 24 hours, the number of splenic lymphocyte clusters of a BS administration group is obviously observed to be larger than that of a control group under a mirror, the size of the clusters is larger than that of the control group, after the stimulation is carried out for 48 hours, the secretion amounts of splenic lymphocyte cytokines IFN-gamma, IL-2 and TNF- α are obviously increased, and compared with the control group, the statistic difference exists, and the secretion amount of the cytokines is increased along with the increase of the administration concentration, so that the dose dependence is presented.
Table 4 effect of BS on secretion of mouse lymphokine.
Figure BDA0001756036070000141
Note: data are presented as mean ± standard deviation (n ═ 3). P <0.05, representing a statistical difference compared to the control group;#p < 0.01, which indicates a statistical difference from the BSL group;p <0.05, which indicates a statistical difference from the BSM group.
The influence of BS on tumor-bearing KM mice is found. The BS can better inhibit the growth of the tumor especially in the early stage of the tumor occurrence, has no obvious influence on the weight, the liver coefficient and the kidney coefficient of a mouse, can play a role in inhibiting the growth of the tumor, and has no obvious hepatorenal toxicity or side effect. The tumor growth inhibiting effect of BS can substantially affect the generation of PD-L1 on the surface of tumor cells and the generation of mouse lymphocyte CD4+、CD8+The research on the influence of the percentage of the subgroup positive cells and the secretion condition of cytokines downstream of PD-L1 shows that the BS can remarkably reduce the expression of PD-L1 on the surface of the tumor cells and is dose-dependent; at the same time, the BS medium and high dose group lymphocyte CD8+The secretion of PD-L1 mainly acts on T cells, and the secretion of cytokines IFN-gamma, IL-2 and TNF- α at the downstream of PD-L1 also shows a rising trend in a BS high-dose group, so that the BS can play a role in inhibiting tumors by down-regulating PD-L1 on the surface of the tumor cells, the immune escape of the tumors through PD-L1 is inhibited to a certain extent, the secretion of the cytokines is promoted, and the tumor immunity of an organism is enhanced.
Example 2 immunosurveillance of BS in a model of postoperative recurrence
2.1 BS Effect on body weight of tumor-bearing H22 mice
In a Balb/c mouse established mouse H22 liver cancer transplantation tumor excision model, the weight change monitoring of the mouse in the administration process shows that as shown in figure 6, the weight of each administration group has no significant difference compared with the control group, and gradually increases, and the weight of the low-dose administration group and the high-dose administration group slightly decreases at the eighth day, and has no statistical difference. In the control group, the body weight of the low (90mg/kg), medium (180mg/kg) and high (360mg/kg) dosage administration groups of the BS is respectively increased to 1.12 +/-0.06, 1.12 +/-0.05, 1.15 +/-0.15 and 1.10 +/-0.07 times of the initial body weight at the eighth day, and no significant difference is seen, which indicates that the BS has no obvious toxicity to the mice.
2.2 Effect of BS on organ coefficients of H22 tumor-bearing mice
After the experiment, the liver, kidney, spleen and thymus of the mice were respectively picked up for observation and weighed. In the process of dissecting mice, the visceral organs of each group of mice are normal, and have no organic changes such as necrosis and swelling. The results are shown in Table 5. There was no significant difference in organ coefficients of liver, kidney and spleen in each group of mice. Spleen coefficients each dose group was increased relative to the control group, but there was no significant difference. There was no significant difference in the slight decrease in liver and kidney coefficients. The thymus coefficient of each dose administration group is obviously increased, has obvious difference (P is less than 0.05), and shows that the thymus coefficient of each dose administration group possibly has the function of improving the immunity of tumor-bearing mice, wherein the function of improving the immunity of the medium dose administration group is the greatest.
TABLE 5 influence of BS on organ coefficients
Figure BDA0001756036070000151
Note: data are presented as mean ± standard deviation (n ═ 6). P <0.05, indicating a statistical difference compared to the control group.
2.3 the Effect of BS on body weight of tumor-bearing Lewis lung carcinoma mice
Balb/c mice were selected to establish a mouse Lewis lung carcinoma graft tumor resection model and sacrificed eight consecutive days after administration on the second day of surgery. The body weight change monitoring of the mice during the administration process is shown in fig. 7, and fig. 7 shows the body weight change of the mice during the administration process. It can be observed that the body weight of each administration group has no significant difference compared with the control group, and the body weight of each administration group gradually increases without statistical difference. In the control group, the body weight of the low, medium and high dose administration groups of the BS respectively increases to 1.16 +/-0.10, 1.14 +/-0.05, 1.13 +/-0.06 and 1.12 +/-0.06 times of the initial body weight on the eighth day, and no significant difference is seen, which indicates that the BS has no obvious toxicity to the mice.
2.4 influence of BS on organ coefficients of tumor-bearing Lewis lung cancer mice
After the experiment, the liver, kidney, spleen and thymus of the mice were respectively picked up for observation and weighed. In the process of dissecting mice, the visceral organs of each group of mice are normal, and have no organic changes such as necrosis and swelling. The results are shown in Table 6. There was no significant difference in organ coefficients of liver, kidney and spleen in each group of mice. The thymus coefficient of the high-dose administration group is increased, wherein the medium-dose administration group has significant difference (P is less than 0.05), which indicates that the BS has the function of improving the immunity of tumor-bearing mice, wherein the function of improving the immunity of the medium-dose administration group is the greatest.
TABLE 6 influence of BS on organ coefficients
Figure BDA0001756036070000152
Note: data are presented as mean ± standard deviation (n ═ 6). P <0.05, indicating a statistical difference compared to the control group.
2.5 BS Effect on body weight of tumor-bearing 4T1 Breast cancer mice
Balb/c mice were selected to establish a mouse 4T1 breast cancer graft tumor resection model and sacrificed eight consecutive days after the administration on the second day of surgery. FIG. 8 shows the body weight change of mice during the administration. It can be observed that the body weight of each administration group has no significant difference compared with the control group, and the body weight of each administration group gradually increases without statistical difference. In the control group, the body weight of the low, medium and high dose administration groups of the BS respectively increases to 1.14 +/-0.04, 1.15 +/-0.03, 1.13 +/-0.02 and 1.10 +/-0.02 times of the initial body weight on the eighth day, and no significant difference is found, which indicates that the BS has no obvious toxicity to the mice.
2.6 influence of BS on organ coefficients of tumor-bearing 4T1 Breast cancer mice
After the experiment, the liver, kidney, spleen and thymus of the mice were respectively picked up for observation and weighed. In the process of dissecting mice, the visceral organs of each group of mice are normal, and have no organic changes such as necrosis and swelling. The results are shown in Table 7. Spleen coefficients and kidney coefficients were increased but not significantly different for each dose group. The thymus coefficient of each dose group was significantly increased (P < 0.05), and the thymus coefficient of the high dose group was the largest and was dose-dependent. The BS has the function of improving the immunity of tumor-bearing mice, wherein the function of improving the immunity of a high-dose administration group is the greatest.
TABLE 7 influence of BS on organ coefficients in mice
Note: data are presented as mean ± standard deviation (n ═ 6). P <0.05, indicating a statistical difference compared to the control group.
Example 3 Induction of immunomodulatory Effect of postoperative combination of BS and Cisplatin (CDDP)
BS-administered group (180 mg/kg): weighing 180mg of BS raw drug, dissolving the BS raw drug in 5mL of 5 per thousand CMC-Na solution to prepare suspension, wherein the administration dose is 5 mL/kg;
cisplatin administration group (2 mg/kg): cisplatin 2mg was weighed and dissolved in 5mL of physiological saline to prepare a solution, and the administration dose was 5 mL/kg.
Combination group: BS (180mg/kg) and cisplatin (2mg/kg) were administered simultaneously at a dose of 5 mL/kg.
Selecting Balb/c mice to establish a mouse Lewis lung cancer transplantation tumor resection model, killing the mice after continuously administering the drug for eight days on the second day of the operation, dissecting the mice, completely stripping the tumor, weighing the measured volume and carrying out corresponding analysis. When the derivative is used in vivo, the concentration of the drug metabolized in vivo to reach the tumor part is greatly related to the in vivo property, so the dosage of the oral benzisoselenazole derivative is far larger than that of a platinum anti-cancer drug (conventional injection administration).
3.1 Effect on secretion of immunologically active substances
Interferon IFN- γ is normally produced by activated T cells and natural killer NK cells, and has antiviral, immunomodulatory and tumor growth-inhibiting properties. Perforin (perforin) and granzyme b (granzyme b) are produced by cytotoxic T lymphocytes and NK cells and are the major effector molecules of cell killing of target cells.
Evaluation of the effects of BS drugs on immunomodulation and induction, lymphocyte secretion after 48h stimulation with 20ng/mL of LPMA, 20ng/mL of ionomycin and 100U/mL of IL-2 was examined by the Elisa method, and the results are shown in Table 8. CDDP single drug can significantly improve the secretion of perforin (P < 0.05); BS remarkably improves the secretion of granzyme (P is less than 0.05); the combined administration group simultaneously and remarkably improves the secretion of perforin and granzyme, and the secretion amount is larger than that of a single medicine group (P is less than 0.05); both the BS and the combination administration group can increase the secretion level of interferon. It is shown that BS and CDDP can synergistically promote the secretion of immunocompetent substances, improve the killing activity of cytotoxic T lymphocytes and NK cells, and improve the immunity of the organism, thereby inhibiting the growth of tumors.
TABLE 8 Effect of BS and CDDP combination on mouse Immunoactive Agents
Figure BDA0001756036070000171
Data are presented as mean ± standard deviation (n ═ 3). P <0.05, indicating a significant difference compared to the control group.
3.2 lymphocyte proliferation Activity assay
The lymphocytes obtained were plated in a medium containing 20ng/mL PMA, 20ng/mL ionomycin and 100U/mL IL-2, and the viability of the cells in each well after 0h and 48h of culture was measured. As a result, as shown in Table 9, the cell counts of the control group, the BS-administered group, the CDDP-administered group and the combination-administered group increased to 151.5. + -. 41.0, 285.4. + -. 44.7, 136.1. + -. 26.3 and 355.9. + -. 21.2%, respectively; the increase rates of the BS-administered group, the CDD-administered group and the combination-administered group were 1.88, 0.90 and 2.35 times as high as those of the control group, respectively. Thus, the BS and the CDD have the function of synergistically promoting the proliferation of the lymphocyte.
TABLE 9 Effect of the combination of BS and CDDP on lymphocyte proliferative Activity
Figure BDA0001756036070000172
Data are presented as mean ± standard deviation (n ═ 3). P <0.05, indicating a significant difference compared to the control group.
3.3 lymphocyte killing Activity assay
The extracted lymphocytes and Lewis lung cancer cells are plated by a culture medium which is stimulated by IL-2 containing 20ng/mL PMA and 20ng/mL ionomycin 100U/mL according to the mode described above, and the killing activity of each lymphocyte on the Lewis lung cancer cells is calculated after 24h of culture. As shown in Table 10, the killing activity of the lymphocytes of the control group, the BS-administered group, the CDDP-administered group and the combination-administered group against Lewis lung cancer cells was 17.06. + -. 2.45, 22.78. + -. 3.22, 19.80. + -. 4.10 and 29.71. + -. 2.55%, respectively. The result shows that the BS and the CDDP have the activity of synergistically improving the killing activity of the lymphocytes on the tumor cells.
TABLE 10 Effect of the combination of BS and CDDP on lymphocyte killing Activity
Figure BDA0001756036070000182
Data are presented as mean ± standard deviation (n ═ 3). P <0.05, indicating a significant difference compared to the control group.
3.4 mouse lymphocyte surface NKG2D ligand expression assay
NKG2D is expressed on the surface of all human and mouse NK cells and T cell subsets and is an activating receptor that plays a key role in tumor immunosuppression. Tumors can achieve the aim of suppressing immune response by reducing NKG2D receptors in vivo, so that immunity escapes. The expression of NKG2D receptor on the surface of mouse lymphocytes was examined by flow cytometry, and the results are shown in Table 11, for the control group, the BS administration group, the CDDP administration group and the combination administration group, 15018. + -. 2581, 27187. + -. 1453, 19789. + -. 1720, 34001. + -. 4426; the expression of NKG2D was increased by 31.8%, 81.0% and 126.4% in the BS-administered group, CDDP-administered group and combination-administered group, respectively. The BS and the CDDP are proved to have the function of synergistically up-regulating the expression of the NKG2D activating receptor on the surface of the lymphocyte.
TABLE 11 influence of the combination of BS and CDDP on the expression of NKG2D
Figure BDA0001756036070000191
Data are presented as mean ± standard deviation (n ═ 3). P <0.05, indicating a significant difference compared to the control group. # P <0.05, indicating a significant difference compared to the CDDP group.
3.5 detection of RAE-1 receptor expression on the surface of mouse tumor cells
RAE-1 is an NKG 2D-activating ligand expressed on the surface of mouse tumor cells. Tumor cells can prevent cytotoxic lymphocytes from recognizing the cells by down-regulating RAE-1 expression, so as to achieve the aim of immune escape. We used the WesternBlot method to detect RAE-1 expression on the surface of mouse tumor cells, and the results are shown in FIG. 9, in which RAE-1 levels were all up-regulated in the BS-administered group, CDDP group and combination group compared to the control group, and the combination group was significantly different (P < 0.05). The result shows that the combination of BS and CDDP can synergistically promote the expression of mouse tumor cell surface NKG2D ligand RAE-1, thereby improving the immune recognition and killing of cytotoxic lymphocytes to tumor cells and inhibiting the tumor growth.
The embodiments of the present invention have been described above. However, the present invention is not limited to the above embodiment. 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 (10)

1. A tumor immunomodulator, wherein the immunomodulator comprises at least a benzisoselenazole derivative.
2. The tumor immunomodulator according to claim 1, wherein the benzoisoselenazole derivative has a structure of a compound represented by formula A, and is selected from at least one of a compound represented by formula A, a precursor thereof, an active metabolite, a stereoisomer, a pharmaceutically acceptable salt, a prodrug, and a solvate thereof,
Figure FDA0001756036060000011
wherein R is1、R2Identical or different, independently of one another, from H or the following radicals: c1-12Alkyl radical, C3-20A cycloalkyl group;
wherein R is selected from C1-12Alkylene, phenylene, biphenylene, triphenylene, or
Figure FDA0001756036060000012
Wherein M represents Pt, Pd or Rh.
3. The tumor immunomodulator according to claim 2, wherein in the compound of formula A, R is1、R2Identical or different, independently of one another, from H, -CH3、-CH2CH3、-CH(CH3)2、-C(CH3)3、-CH(CH2)4or-CH (CH)2)5(ii) a R is selected from-CH2-、-C2H4-、-C4H8-, phenylene-C6H4-。
4. The tumor immunomodulator according to any one of claims 1 to 3, wherein the benzisoselenazole derivative is selected from 1, 2-bis [2- (1, 2-benzisoselenazol-3 (2H) -one) ] -Butane (BS), which has the following structure:
Figure FDA0001756036060000013
5. the tumor immunomodulator according to any one of claims 1 to 4, wherein the tumor immunomodulator comprises a benzisoselenazole derivative and a platinum anticancer drug;
preferably, the platinum-based anticancer drug is selected from at least one of Cisplatin (CDDP), Carboplatin (CBP), oxaliplatin (1-OHP), and nedaplatin (CDGP).
6. The tumor immunomodulator according to claim 5, wherein the molar ratio of the benzisoselenazole derivative to the platinum anticancer drug is (1-99): (1-99);
preferably, the molar ratio of the benzisoselenazole derivative to the platinum-based anticancer drug is (50-90): (1-30), (60-70): 1-10), (1-40): 1-40), (1-25): 1-25), (1-10): 1-10), (1-6): 1-6);
more preferably, the molar ratio of the benzisoselenazole derivative to the platinum-based anticancer drug is 1:1, 2:1 (in vitro combination) or 60:1, 70:1, 80:1 (in vivo combination).
7. The tumor immunomodulator according to any one of claims 1 to 6, wherein the tumor immunomodulator further comprises at least one pharmaceutically acceptable auxiliary material.
8. Use of a tumor immunomodulator according to any one of claims 1 to 7 in the treatment of tumors; preferably, the dose of the immunomodulator is 10-500 mg/kg based on the weight of the object to be applied.
9. The use according to claim 8, of said immunomodulator in early tumour immunomodulation.
10. The use according to claim 8, of an immunomodulator in the immunomodulation of postoperative tumor recurrence.
CN201810887556.5A 2018-08-06 2018-08-06 Tumor immunomodulator and application thereof Pending CN110856717A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201810887556.5A CN110856717A (en) 2018-08-06 2018-08-06 Tumor immunomodulator and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201810887556.5A CN110856717A (en) 2018-08-06 2018-08-06 Tumor immunomodulator and application thereof

Publications (1)

Publication Number Publication Date
CN110856717A true CN110856717A (en) 2020-03-03

Family

ID=69634720

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810887556.5A Pending CN110856717A (en) 2018-08-06 2018-08-06 Tumor immunomodulator and application thereof

Country Status (1)

Country Link
CN (1) CN110856717A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115518064A (en) * 2022-11-21 2022-12-27 北京市神经外科研究所 Application of benzisoselenazole compound in preparation of medicine for treating glioma

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1594299A (en) * 2003-09-10 2005-03-16 北京大学药学院 Dibenzo-isozaolone compounds and their synthesis process and application
US20090117204A1 (en) * 2001-06-08 2009-05-07 Peking University Benzoisoselenazole derivatives with anti-inflammation, antivirus and antithrombosis activity and their use
CN101781283A (en) * 2009-01-16 2010-07-21 曾慧慧 Thioredoxin reductase inhibiter compounds and preparation method and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090117204A1 (en) * 2001-06-08 2009-05-07 Peking University Benzoisoselenazole derivatives with anti-inflammation, antivirus and antithrombosis activity and their use
CN1594299A (en) * 2003-09-10 2005-03-16 北京大学药学院 Dibenzo-isozaolone compounds and their synthesis process and application
CN101781283A (en) * 2009-01-16 2010-07-21 曾慧慧 Thioredoxin reductase inhibiter compounds and preparation method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
QIAO ZOU等: "Butaselen and cisplatin synergistically inhibit human cancer cells proliferation", 《JOURNAL OF CHINESE PHARMACEUTICAL SCIENCES》 *
XIAOQING ZHENG等: "Butaselen prevents hepatocarcinogenesis and progression through inhibiting thioredoxin reductase activity", 《REDOX BIOLOGY》 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115518064A (en) * 2022-11-21 2022-12-27 北京市神经外科研究所 Application of benzisoselenazole compound in preparation of medicine for treating glioma
WO2024109574A1 (en) * 2022-11-21 2024-05-30 北京市神经外科研究所 Use of benzisoselenazole compound in preparation of drug for treatment of spinaglioma

Similar Documents

Publication Publication Date Title
Zhou et al. Immunomodulatory effects of herbal formula of astragalus polysaccharide (APS) and polysaccharopeptide (PSP) in mice with lung cancer
Noori et al. Dihydroartemisinin shift the immune response towards Th1, inhibit the tumor growth in vitro and in vivo
EP3311841B1 (en) Anticancer agent
EP2749292A1 (en) Tumor chemotherapeutic drug formulation and preparation method thereof
CN106943432B (en) Exosome derived from umbilical cord mesenchymal stem cells and application of exosome in preparation of liver cancer treatment drug
CA2583464A1 (en) Ecteinascidin compounds as anti -inflammatory agents
US20230256066A1 (en) Pharmaceutical composition for use in the treatment of cancer
CN108498497B (en) Pharmaceutical composition for treating kidney cancer and application thereof
CA2988989A1 (en) Composition containing carboplatin and use
CN114425080B (en) Application of bacteroides fragilis and PD-1 or PD-L1 antibody combined medicament in treatment of genitourinary system cancer
CN114469987A (en) Application of bacteroides fragilis zwitter ion capsular polysaccharide and immune checkpoint inhibitor in combined drug treatment of genitourinary system tumor
US6436411B1 (en) Cancer treatment method
CN110856717A (en) Tumor immunomodulator and application thereof
CN110801449A (en) Application of benzisoselenazole derivative in preparation of tumor treatment drug
KR20210016995A (en) Exosome derived from radiated cancer cell, cancer vaccine comprising dendritic cell prepared by using the same and preparing method thereof
WO2022100535A1 (en) Antitumor pharmaceutical composition and application thereof
CN109420167B (en) Combined medicine for treating tumor
CN110856718A (en) Application of benzisoselenazole derivative and platinum medicine in preparation of medicine for treating tumor and postoperative tumor recurrence
WO2017177515A1 (en) Application of 4-hydroxy salicylanilide in preparation of anti-myeloma or anti-lymphoma drugs
Tseng et al. Induction of immune responses and phosphatidylserine exposure by TLR9 activation results in a cooperative antitumor effect with a phosphatidylserine-targeting prodrug
Li et al. Pilose antler polypeptides promote chemosensitization and T-cell infiltration of triple-negative breast cancer
CN112791106A (en) Pharmaceutical compositions and their use for the treatment of diseases
CN116617222B (en) Application of small molecular ion channel blocker MK-801 in preparation of medicines for treating tumors or resisting infection
CN106539812B (en) Application of the ring dinucleotides cGAMP in anti-colorectal carcinoma hepatic metastases
CN110801450A (en) Application of benzisoselenazole derivative and antimetabolite in preparation of tumor treatment drug

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