WO2016119308A1 - Préparation antitumorale et son procédé de préparation - Google Patents

Préparation antitumorale et son procédé de préparation Download PDF

Info

Publication number
WO2016119308A1
WO2016119308A1 PCT/CN2015/075948 CN2015075948W WO2016119308A1 WO 2016119308 A1 WO2016119308 A1 WO 2016119308A1 CN 2015075948 W CN2015075948 W CN 2015075948W WO 2016119308 A1 WO2016119308 A1 WO 2016119308A1
Authority
WO
WIPO (PCT)
Prior art keywords
preparation
tumor
antitumor
cancer
alginate
Prior art date
Application number
PCT/CN2015/075948
Other languages
English (en)
Chinese (zh)
Inventor
王琳
李永奎
方敏
王征
张剑
宋煜
石洁
徐妞妞
王健
Original Assignee
华中科技大学同济医学院附属协和医院
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 华中科技大学同济医学院附属协和医院 filed Critical 华中科技大学同济医学院附属协和医院
Publication of WO2016119308A1 publication Critical patent/WO2016119308A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/63Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide
    • A61K31/635Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide having a heterocyclic ring, e.g. sulfadiazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum

Definitions

  • the invention relates to a pharmaceutical preparation and a preparation method thereof, in particular to an antitumor preparation and a preparation method thereof.
  • Tumors are one of the greatest threats to human health and life.
  • the World Health Organization reports that 14.1 million people worldwide contracted cancer in 2012, with 8.2 million deaths, exceeding the combined death toll from AIDS, malaria and tuberculosis. China's new cancer cases rank first in the world, and cancer has become the number one cause of death among Chinese residents. Every year, 2.07 million people die of cancer.
  • the main treatment methods for tumors are surgical resection, chemotherapy and radiotherapy.
  • chemotherapy and radiotherapy can assist the treatment of malignant tumors to some extent, the strong side effects cause additional damage to the body. Huge damage and a low cure rate.
  • Most chemotherapy drugs cause irreversible damage to normal tissues and organs of the body while killing tumor cells. Therefore, traditional treatment techniques cannot solve the major threat to human health caused by tumors, especially malignant tumors.
  • Tumor immunotherapy technology is a kind of tumor treatment technology developed and developed based on the basic principles of tumor immunity to achieve effective killing of tumor cells by immunological means.
  • the immune system has strong pertinence and specificity for the killing of tumor cells, so the immunological treatment technology of such diseases has the natural high efficiency and targeting unmatched by traditional therapeutic techniques.
  • a successful immunological treatment program can activate the body to produce enough tumor cell characteristics of cytotoxic T lymphocytes, which can selectively kill all tumor cells.
  • immunotherapy is currently The only known means that is expected to completely eliminate tumor cells has no adverse effects on normal cells of the body.
  • ipilimumab as a blocker of B7/CTLA-4 costimulatory pathways
  • ipilimumab has been shown to be effective in improving advanced malignant melanoma.
  • Patient's life The rate of deposit, which was approved by the FDA in 2011 for the treatment of metastatic malignant melanoma, but Iprimima can cause serious and fatal autoimmune-related diseases.
  • the programmed death molecule-1 (PD-1) pathway blocker has also made a breakthrough in anti-PD-1 and PD-L1 monoclonal antibodies, which can block the inhibitory effect of PD-1 on T cells.
  • PD-1 programmed death molecule-1
  • Multiple tumor-bearing animal experiments have shown that blocking the PD-L1/PD-1 pathway can enhance anti-tumor effects, prolong survival, and improve prognosis.
  • monoclonal antibodies against PD-L1/PD-1 pathway blockade are used in phase III clinical trials for advanced malignancies.
  • PD-1 antibodies are due to their broad-spectrum anticancer effects on kidney cancer.
  • the clinical efficacy of cancers such as gastric cancer, breast cancer, bladder cancer, leukemia, head and neck cancer, intestinal cancer and brain tumors is also in Phase II or Phase III clinical trials.
  • cancers such as gastric cancer, breast cancer, bladder cancer, leukemia, head and neck cancer, intestinal cancer and brain tumors is also in Phase II or Phase III clinical trials.
  • continuous infusion or frequent administration is required to obtain the desired therapeutic effect.
  • the dose of PD-1 antibody is usually 250ug/mouse per mouse, and each It is administered once every two days to maintain the efficacy.
  • Such high doses cause side effects during systemic treatment, such as side effects such as immunogenic enteritis and severe autoimmune diseases, which limits its application.
  • the present invention provides an antitumor preparation which is an antitumor preparation in the form of a hydrogel formed by blocking an aqueous solution of an antibody and celecoxib with an active ingredient PD-1.
  • the formulation has good in vivo stability, a markedly enhanced inhibition of tumor cell growth and metastasis, an expanded anti-tumor profile, and reduced toxic side effects on the body.
  • the invention also provides a preparation method of an anti-tumor preparation, which comprises dispersing an active ingredient PD-1 blocking antibody and celecoxib in an alginate solution, and forming an injectable anti-tumor preparation in the form of a hydrogel.
  • the anti-tumor preparation prepared by the method is applied to an individual, and the PD-1 blocking antibody and celecoxib exhibit good in vivo stability and prolonged sustained release effect, which is more conducive to the exertion of the active ingredient and lowering It uses the dose.
  • Non-steroidal anti-inflammatory drugs such as Celecoxib
  • Celecoxib have been shown to inhibit a variety of Tumor cell proliferation, promote cell apoptosis, inhibit tumor neovascularization and prevent tumorigenesis and development, and prevent tumor growth, inhibit tumor growth and metastasis, and achieve good results in animal experiments.
  • its clinical application is still limited, mainly because many problems remain to be solved, such as longer medication time and larger dosage.
  • Mouse experiments show that it is usually necessary to administer a dose of 25 mg/kg body weight of celecoxib per day. It has anti-tumor effect, resulting in large side effects in the gastrointestinal tract and cardiovascular, and narrow anti-tumor spectrum (mainly used for prevention and early treatment of colorectal cancer).
  • non-steroidal anti-inflammatory drugs such as celecoxib combined with PD-1 blocking antibody, which can reduce the amount of drug, expand the anti-tumor spectrum, and reduce the side effects of the body.
  • the present invention provides an antitumor preparation which is an antitumor preparation in the form of a hydrogel formed by dissolving an aqueous solution of an active ingredient PD-1 blocking antibody and celecoxib, which is an injection. preparation.
  • the alginate has a molecular weight of from 75,000 to 300,000 g.
  • the alginate solution mixture is crosslinked in the presence of a crosslinking agent.
  • the crosslinking agent is a divalent metal cation.
  • the divalent metal cation is provided by a sulfate or a hydrochloride of a divalent metal, which is calcium or magnesium.
  • the divalent metal cation is provided by CaSO 4 , MgCl 2 or CaCl 2 .
  • the anti-tumor preparation is an anti-tumor preparation for melanoma, breast cancer, kidney cancer, gastric cancer, bladder cancer, leukemia, head and neck cancer, intestinal cancer or brain tumor.
  • the invention provides a method for preparing an anti-tumor preparation, comprising:
  • the alginate solution mixture is crosslinked to form an antitumor preparation in the form of a hydrogel, which is an injection preparation.
  • the alginate has a molecular weight of 75,000 to 300,000 g.
  • those skilled in the art can select as needed within the above molecular weight range.
  • One of the above molecular weight ranges of one or more alginate can also be used directly by those skilled in the art.
  • the alginate solution mixture is crosslinked in the presence of a crosslinking agent.
  • the crosslinking agent is a divalent metal cation.
  • the divalent metal cation is provided by a sulfate or a hydrochloride of a divalent metal, which is calcium or magnesium.
  • the divalent metal cation is provided by CaSO 4 , MgCl 2 or CaCl 2 .
  • the anti-tumor preparation is an anti-tumor preparation for melanoma, breast cancer, kidney cancer, gastric cancer, bladder cancer, leukemia, head and neck cancer, intestinal cancer or brain tumor.
  • celecoxib since celecoxib is insoluble in water in a powder form, it can be subjected to ultrasonication in the process of dispersing it in an alginate solution.
  • the process of preparing the antitumor preparation of the present invention should be carried out under aseptic conditions.
  • the antitumor preparation provided by the present invention can be administered by subcutaneous injection to inhibit tumorigenesis or kill tumor cells.
  • the antitumor preparation of the present invention may be a unit preparation.
  • the unit preparation is a preparation which satisfies the active ingredient required for one administration, such as a unit (needle) injection or the like.
  • the amount of drug required for a single administration of a patient can be conveniently obtained by calculating the product of the patient's body weight and the unit weight dose required for the patient to take the drug once.
  • the adult body weight is 50-70 kg, and the dose can be determined initially by an equivalent dose conversion relationship between the experimental animal and the human body weight dose.
  • the human and mouse doses can be converted according to the body surface area conversion coefficient of 0.0026 for humans and mice.
  • a mouse weighing 25 g is injected subcutaneously with 0.2 ml of an antitumor preparation of the invention, wherein the PD-1 blocking antibody is 100 ug and the celecoxib is 625 ug, ie: 4 mg. /kg mouse body weight of PD-1 blocking antibody, 25mg/kg mouse body weight of celecoxib, can be maintained once every two weeks.
  • the pharmaceutical manufacturer can obtain the content of the active ingredient in the unit preparation for one administration of the adult according to the above conversion method, and apply it to the pharmaceutical process.
  • the unit preparation contains 20-30 mg of the PD-1 blocking antibody and 130-200mg Celecoxib.
  • the solution concentration of the alginate solution may be in the range of 10-40 mg/mL, and the PD-1 blocking antibody may be dispersed in the above alginate in an amount of 500 to 1000 ⁇ g/mL.
  • the celecoxib may be dispersed in the above alginate solution in an amount of 3.75 to 7.5 mg/mL.
  • Applicants may appropriately select the solution concentration of the alginate solution according to the specifications of the antitumor preparation to be prepared, for example, the amount of the PD-1 blocking antibody and celecoxib to be contained in the unit preparation, and PD-1 blocks the antibody,
  • the amount of celecoxib dispersed in the alginate is achievable by those skilled in the art.
  • the present invention also provides a method for treating or preventing a tumor comprising the step of administering an antitumor preparation to an individual having a tumor or an individual having a tendency to develop a tumor, the antitumor preparation being blocked by the active ingredient PD-1
  • An antitumor preparation in the form of a hydrogel formed by breaking an antibody and an aqueous solution of alginate of celecoxib, and the antitumor preparation is an injection preparation.
  • the anti-tumor preparation further comprises a crosslinking agent for crosslinking the aqueous alginate solution.
  • the alginate has a molecular weight of 75,000 - 300,000 g
  • the crosslinking agent is a divalent metal cation.
  • the anti-tumor preparation is a unit preparation.
  • the unit preparation contains a therapeutically effective amount of 20-30 mg of PD-1 blocking antibody and 130-200 mg of celecoxib.
  • An individual having a tendency to develop a tumor in the present invention refers to an individual who is genetically at risk for a tumor or an individual who exhibits a risk of developing a tumor on other physical indicators.
  • the antitumor preparation prepared by the method of the invention can make PD-1 blocking antibody and celecoxib have good stability in vivo, and has an extended sustained release effect, thereby being more beneficial to the effect of the drug. , reduce the dose.
  • the anti-tumor preparation of the present invention is administered to an individual by injection, that is, the PD-1 blocking antibody and celecoxib are simultaneously encapsulated into the alginate hydrogel to provide the individual, as compared to Administration of alginate hydrogel containing only PD-1 blocking antibody and/or alginate hydrogel containing only celecoxib increases the percentage of survival of tumor-bearing mice and promotes the production of active CD8 T cells
  • the raw side has an unexpected synergy.
  • the anti-tumor preparation provided by the invention enhances the acquired immune response against tumor cells by the body, blocks the immunosuppressive pathway, activates the effector cells to specifically kill the tumor cells, and has almost no harm to the normal tissues and cells of the body. Compared with the damage caused by surgery, chemotherapy and radiotherapy, the advantages are obvious.
  • the material used in the solution of the present invention has a wide range of sources, and can be extracted from seaweed in large amounts, and a significantly reduced PD-1 blocking antibody and celecoxib dose can be used to obtain a good tumor suppressing effect, remarkable Reduce the cost of medication for patients.
  • the anti-tumor preparation provided by the invention can be used for treating solid tumors and inhibiting tumor growth; and can be used for treating tumors with malignant metastasis; and for adjuvant treatment of post-operative radiotherapy and chemotherapy for malignant tumors;
  • the treatment of tumors that cannot be surgically removed has a wide range of applications.
  • Figure 1 is a flow chart showing the preparation of the antitumor preparation of the present invention.
  • Figure 2A shows the concentration of celecoxib in the serum of each group of mice
  • Figure 2B shows the content of celecoxib in the tumor tissues of each group of mice
  • Figure 2C shows the ratio of cytotoxic T lymphocytes (CD8 and IFN- ⁇ double positive) and regulatory T lymphocytes (CD4 and FoxP3 double positive) in each group of mice.
  • Figure 2D shows the percent survival of each group of mice.
  • Figures 3A-3B show the results of detection of CD8 T cells that are effective in killing tumor cells in the tumor microenvironment.
  • Figure 4A shows the expression level of relative messenger RNA of the COX-2 gene.
  • Figure 4B shows the expression level of the relative messenger RNA of the IL-1 ⁇ gene.
  • Figure 4C shows the expression level of the relative messenger RNA of the IL-6 gene.
  • * indicates a p value of ⁇ 0.05, and ** indicates a p value of ⁇ 0.01.
  • Figure 5A shows a picture of the tumor appearance of mice of each group on days 0, 10, 13, 17, 20, and 22.
  • Figure 5B shows tumor size changes for mice of each group on days 0, 10, 13, 17, 20, and 22.
  • * indicates a p value ⁇ 0.05.
  • Figure 6A shows a picture of a pulmonary metastases from each group of mice.
  • Figure 6B shows the number of lung metastases in each group of mice. Where $ indicates that the group is compared with the group a; # indicates that the group is compared with the group b. *, $, # indicates p value ⁇ 0.05; $$, ## indicates p value ⁇ 0.01.
  • Example 1 Preparation of an antitumor preparation of the present invention.
  • the antitumor preparation of the present invention can be prepared according to the procedure shown in Fig. 1.
  • the alginate solution of Philadelphia was stirred overnight at 4 ° C with a magnetic stirrer and filtered through a filter having a pore size of 0.22 ⁇ m.
  • Step (2)-a 3.75 to 7.5 mg of celecoxib powder (LC laboratory, LC Laboratories, Massachusetts, USA) is weighed per ml of alginate solution and mixed;
  • Step (2)-b using a probe type ultrasonic instrument (QSONICA, USA) to ultrasonically 60 seconds for 60 seconds, so that the celecoxib powder is uniformly dispersed in the alginate solution to obtain a mixed system;
  • QSONICA probe type ultrasonic instrument
  • Step (3) 500-1000 ⁇ g of PD-1 blocking antibody per ml of the alginate solution (BioXCell, USA) is added to the mixed system obtained in the step (2)-b, and the shaker is shaken and thoroughly mixed;
  • Step (4)-a weigh calcium sulfate solid powder (CaSO 4 , Sinopharm Chemical Reagent Co., Ltd.), mix it with purified water at a mass to volume ratio of 0.21 g/ml, and autoclave to obtain a calcium sulfate suspension ( If aggregation occurs during use, coarse particles are generated, which can be ultrasonicated by an ultrasonic machine to form a uniform suspension);
  • Step (4)-b sucking the mixture obtained in the step (3) with a syringe with a screw;
  • Step (4)-c adding another suspension of CaSO 4 to the syringe with screw, adding 40 ⁇ L of CaSO 4 suspension per mL of alginate solution;
  • Step (5) through the joint and the screw, the two syringes are tightly connected to eliminate excess air as much as possible;
  • Step (6) quickly push the syringe 20 times each other;
  • Step (7) standing for 1 minute, the antitumor preparation of the present invention in the form of a hydrogel is completed.
  • the syringe can be injected by unscrewing the syringe containing the hydrogel and attaching the injection needle. Or can The prepared antitumor preparation was stored at 4 °C.
  • Example 2 In vivo sustained release effect and drug effect prolongation effect of antitumor preparation provided by the invention
  • the antitumor preparation was prepared in the same manner as in Example 1 so that the anti-tumor preparation contained 500 ug of PD-1 blocking antibody and 3125 ug of celecoxib per ml of the antitumor preparation.
  • mice were purchased from Beijing Huakangkang Biotechnology Co., Ltd., each weighing about 25 grams.
  • B16-F10 melanoma cells were purchased from the American Type Culture Collection.
  • B16-F10 melanoma cells are recognized as the most invasive tumor cells. After each subcutaneous injection of 2.5 ⁇ 10 4 B16-F10 melanoma cells in C57BL/6 mice, the protoplasts can be produced after 1 week. A melanoma that then grows rapidly and causes the mouse to die.
  • the C57BL/6 mouse melanoma model is a mature animal model widely used to evaluate the anti-tumor effect of drugs.
  • the inventors will subcutaneously inject B16-F10 melanoma cells purchased from the ATCC (American Type Culture Collection) with C57BL/6 male mice weighing about 25 grams at 6-8 weeks of age (purchased from Beijing Huakangkang Biotechnology). Technology Co., Ltd.), about 7-10 days later, melanoma formation can be seen at the site of injection of cells.
  • the anti-tumor preparation provided by the invention has an effect of sustained release effect and prolonged effect of the drug in vivo.
  • mice After one week of inoculation of melanoma cells in C57BL/6 mice, the following three groups of mice were set up:
  • mice serum and tumor tissue were taken 24 hours after administration, the other two Mouse sera and tumor tissues were taken on the first day, the third day, the 7th day and the 14th day after administration, and the mouse serum was detected by liquid chromatography-mass spectrometry (HPLC-MS).
  • HPLC-MS liquid chromatography-mass spectrometry
  • FIG 2A shows the concentration of celecoxib in the serum of each group of mice.
  • Figure 2B shows the celecoxib content in tumor tissues of each group of mice. It can be seen that the serum concentration of celecoxib in the experimental group injected with the antitumor preparation of the present invention is significantly higher than that of the control group at least within one week; the celecoxib content in the tumor tissue is significantly higher at least within two weeks. In the control group.
  • mice After one week of inoculation of melanoma cells in C57BL/6 mice, the following 4 groups of mice were set up:
  • inguinal lymph nodes were sampled on the first day, the third day, the 7th day and the 14th day after treatment, and the cytotoxic T lymphocytes were detected by flow cytometry.
  • the number of cells double positive for CD8 and IFN- ⁇
  • regulatory T lymphocytes double positive for CD4 and FoxP3
  • Figure 2C shows cytotoxic T lymphocytes (CD8 and IFN- ⁇ double positive) in each group of mice.
  • the ratio of regulatory T lymphocytes CD4 and FoxP3 double positive.
  • the anti-tumor preparation provided by the present invention is described by the data of the present embodiment, and the effective drug concentration and effect for a long time are maintained while reducing the dose.
  • mice After one week of inoculation of melanoma cells in C57BL/6 mice, the following 4 groups of mice were set up:
  • Figure 2D shows the percent survival of each group of mice. It is indicated that the administration of the antitumor agent group of the present invention (formed by simultaneously encapsulating the PD-1 antibody and celecoxib into the alginate hydrogel) gives an active ingredient group which is coated into the alginate hydrogel in a stepwise manner ( That is, administration of an alginate hydrogel containing only the PD-1 antibody, and simultaneous administration of an alginate hydrogel containing only celecoxib, resulted in a significantly increased percentage of mouse survival.
  • the antitumor preparation of the present invention does not simply use the three components of celecoxib, PD-1 antibody and alginate hydrogel, and celecoxib and PD-1 antibodies must be simultaneously embedded in alginic acid. In a saline gel (i.e., the antitumor preparation of the present invention), a superior antitumor effect can be produced.
  • Example 3 The antitumor preparation of the present invention can promote the production of active CD8 T cells
  • the antitumor preparation was prepared in the same manner as in Example 1 so that the anti-tumor preparation contained 500 ug of PD-1 blocking antibody and 3125 ug of celecoxib per ml of the antitumor preparation.
  • Example 2 The same mouse melanoma model as in Example 2 was constructed.
  • mice After one week of inoculation of melanoma cells in C57BL/6 mice, the following 4 groups of mice were set up:
  • Figures 3A and 3B show the results of detection of CD8 T cells that are effective in killing tumor cells in the tumor microenvironment. It was shown that administration of the antitumor agent group of the present invention (formed by simultaneously encapsulating PD-1 antibody and celecoxib into alginate hydrogel) can greatly promote the production of active CD8 T cells. This indicates that the antitumor preparation of the present invention is not only used as a drug sustained release carrier in the treatment of tumors, but also the volume and space occupied by the body is a place for promoting the production of antitumor immune cells. Both celecoxib and PD-1 antibody are present in the "site” and they are activated The synergistic effect of the plaque cells can be produced.
  • Example 4 The anti-tumor preparation provided by the present invention has reduced toxic side effects.
  • the antitumor preparation was prepared in the same manner as in Example 1 so that the anti-tumor preparation contained 500 ug of PD-1 blocking antibody and 3125 ug of celecoxib per ml of the antitumor preparation.
  • Example 2 The same mouse melanoma model as in Example 2 was constructed.
  • mice One week after inoculation of melanoma cells in C57BL/6 mice, the following groups of mice were set up:
  • mice were further cultured for one week, the mice were sacrificed, the subcutaneous tumors were removed, total RNA was extracted according to the RNA extraction reagent, and COX-2 was analyzed by real-time PCR.
  • Figures 4A-4C which is the expression level of the relative messenger RNA of the COX-2 gene.
  • Figure 4B is the expression level of relative messenger RNA of the IL-1 ⁇ gene.
  • Figure 4C shows the level of expression of the relative messenger RNA of the IL-6 gene.
  • the experimental group injecting the antitumor preparation group of the present invention has a significantly reduced expression of inflammatory factors as compared with the alginate hydrogel group injected with the PD-1 antibody. It is shown that the antitumor preparation provided by the present invention has a significantly reduced toxic side effect on the body.
  • Example 5 The antitumor preparation provided by the invention is used for inhibiting tumor growth of B16 melanoma
  • the antitumor preparation was prepared in the same manner as in Example 1 so that the anti-tumor preparation contained 500 ug of PD-1 blocking antibody and 3125 ug of celecoxib per ml of the antitumor preparation.
  • Example 2 The same mouse melanoma model as in Example 2 was constructed.
  • the antitumor preparation provided by the present invention is used for the anti-B16 melanoma tumor growth effect.
  • mice One week after inoculation of melanoma cells in C57BL/6 mice, the following groups of mice were set up:
  • Figure 5A shows a picture of the tumor appearance of mice of each group on days 0, 10, 13, 17, 20, and 22.
  • Figure 5B shows the size changes of tumors of mice of each group on days 0, 10, 13, 17, 20, and 22.
  • the experimental group in which the antitumor preparation of the present invention was subcutaneously injected was able to inhibit the growth of malignant melanoma in situ tumors as compared with the control group.
  • the anti-tumor effect of the anti-tumor preparation of the invention is better than that of the alginate hydrogel alone occluding celecoxib or PD-1 blocking antibody, and is also superior to the saline mixed celecoxib and PD-1 blocking.
  • the antibody group that is, all the components in the antitumor preparation of the present invention, is necessary for obtaining the antitumor effect of the present application, and an effect that the PD-1 blocking antibody alone or celecoxib is not administered alone is obtained.
  • Example 6 The anti-tumor preparation provided by the invention is used for controlling the metastasis of 4T1 malignant breast cancer
  • the antitumor preparation was prepared in the same manner as in Example 1 so that the anti-tumor preparation contained 500 ug of PD-1 blocking antibody and 3125 ug of celecoxib per ml of the antitumor preparation.
  • mice were purchased from the Medical Laboratory Animal Center of Wuhan University, each weighing about 25 grams.
  • Mouse breast cancer cells were purchased from the American Type Culture Collection.
  • the 4T1 breast cancer metastasis model is the most commonly used experimental model for studying malignant tumor metastasis.
  • the inventors injected 4T1 mouse breast cancer cells (purchased from the American Type Culture Collection) in vitro into the second breast site on the right side of Balb/c mice with a female body weight of about 25 grams at 6-8 weeks old.
  • the amount of cells injected by only one mouse was 1 ⁇ 10 6 .
  • a week later it was seen that the breast site was injected with obvious breast tumors, and after 4-5 weeks, obvious lung metastases were observed.
  • By observing the appearance of the lungs in mice after invasion by cancer cells, and counting the number of lung metastases qualitative and quantitative analysis of cancer cell metastasis can be performed.
  • the antitumor preparation provided by the present invention inhibits the effect of tumor metastasis.
  • mice One week after the above Balb/c mice were inoculated with 4T1 breast cancer cells, the following groups of mice were set up:
  • mice in each group were sacrificed by cervical dislocation, and the whole lung was taken, and the number of tumors on the surface of the lungs was counted from the lungs.
  • the tissue was surgically stripped of the metastatic tumor, and the whole lung and the exfoliated metastatic tumor of the mouse were photographed.
  • Figure 6A shows a picture of a pulmonary metastases from each group of mice.
  • Figure 6B shows the number of lung metastases in each group of mice.
  • the injection of the antitumor preparation group of the present invention showed an excellent effect of inhibiting tumor metastasis as compared with the control group, and the number of lung metastases was suppressed by 70%.
  • the number of metastases was significantly increased significantly in the control group lacking any one component, and the effect of inhibiting tumor metastasis was far less than the present invention, indicating all the components in the antitumor preparation of the present invention. It is necessary to obtain the anti-tumor effect desired by the present application.
  • Example 7 The antitumor preparation of the present invention is equally effective for melanoma and other tumor types other than breast cancer
  • Different tumor-bearing mice include: kidney cancer mice, gastric cancer mice, bladder cancer mice, leukemia mice, head and neck cancer mice, intestinal cancer mice, brain tumor mice, respectively, purchased from Wuhan University Medical Laboratory Animal Center, Weighing about 25 grams.
  • the antitumor preparation was prepared in the same manner as in Example 1 so that the anti-tumor preparation contained 500 ug of PD-1 blocking antibody and 3125 ug of celecoxib per ml of the antitumor preparation.
  • tumor-bearing mice The method of constructing a tumor-bearing mouse model in this example is a conventional method in the art, and these tumor-bearing mouse models are mature animal models widely used to evaluate the anti-tumor effect of drugs.
  • the experimental results show that the kidney cancer mice, the gastric cancer mice, the bladder cancer mice, the leukemia mice, the head and neck cancer mice, the intestinal cancer mice, and the brain tumor mice to which the antitumor preparation of the present invention is administered are compared with the respective control groups. Mice, which exhibit excellent tumor growth inhibition and reduced toxic side effects.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

La présente invention concerne une préparation antitumorale et son procédé de préparation. La préparation antitumorale est une préparation sous la forme d'un hydrogel formé à partir d'une solution aqueuse d'alginate de principes actifs dispersés d'un anticorps de blocage de PD-1 et de célécoxib, et la préparation est une préparation injectable.
PCT/CN2015/075948 2015-01-28 2015-04-07 Préparation antitumorale et son procédé de préparation WO2016119308A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510043930.XA CN105983097B (zh) 2015-01-28 2015-01-28 一种抗肿瘤制剂及其制备方法
CN201510043930X 2015-01-28

Publications (1)

Publication Number Publication Date
WO2016119308A1 true WO2016119308A1 (fr) 2016-08-04

Family

ID=56542266

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/075948 WO2016119308A1 (fr) 2015-01-28 2015-04-07 Préparation antitumorale et son procédé de préparation

Country Status (2)

Country Link
CN (1) CN105983097B (fr)
WO (1) WO2016119308A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018045058A1 (fr) * 2016-08-30 2018-03-08 Dana-Farber Cancer Institute, Inc. Compositions d'administration de médicament et leurs utilisations
US10344090B2 (en) 2013-12-12 2019-07-09 Shanghai Hangrui Pharmaceutical Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
CN111358752A (zh) * 2020-04-14 2020-07-03 大连市慧恩生物科技开发有限公司 一种抗癌水凝胶组合物及其制备方法
EP3656400A4 (fr) * 2017-07-20 2021-10-20 National University Corporation Hokkaido University Utilisation en association d'un inhibiteur ciblant pd-1/pd-l1 et d'un inhibiteur de cox-2

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019118686A1 (fr) * 2017-12-13 2019-06-20 North Carolina State University Compositions comprenant des agents chimiothérapeutiques et des inhibiteurs de points de contrôle et procédés d'utilisation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101423564A (zh) * 2008-12-08 2009-05-06 青岛明月海藻集团有限公司 一种用于快速制备水凝胶的海藻酸盐的制备方法
WO2014046983A1 (fr) * 2012-09-21 2014-03-27 Intensity Therapeutic Procédé de traitement du cancer

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010027423A2 (fr) * 2008-08-25 2010-03-11 Amplimmune, Inc. Compositions d'antagonistes de pd-1 et methodes d'utilisation associees
CN102218140B (zh) * 2010-04-15 2013-03-27 上海天甲生物医药有限公司 丹酚酸b和昔布类药物联合用药的抗肿瘤效果
EP2897962A1 (fr) * 2012-09-21 2015-07-29 Advinus Therapeutics Limited Composés tricycliques condensés substitués, compositions et applications médicales correspondantes

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101423564A (zh) * 2008-12-08 2009-05-06 青岛明月海藻集团有限公司 一种用于快速制备水凝胶的海藻酸盐的制备方法
WO2014046983A1 (fr) * 2012-09-21 2014-03-27 Intensity Therapeutic Procédé de traitement du cancer

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10344090B2 (en) 2013-12-12 2019-07-09 Shanghai Hangrui Pharmaceutical Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
US11365255B2 (en) 2013-12-12 2022-06-21 Suzhou Suncadia Biopharmaceuticals Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
KR20230023060A (ko) * 2016-08-30 2023-02-16 다나-파버 캔서 인스티튜트 인크. 약물 전달 조성물 및 그의 용도
EP3922279A1 (fr) * 2016-08-30 2021-12-15 Dana Farber Cancer Institute, Inc. Compositions pour l'administration de médicaments et leurs utilisations
US10435469B2 (en) 2016-08-30 2019-10-08 Dana-Farber Cancer Institute, Inc. Drug delivery compositions and uses thereof
KR102650076B1 (ko) * 2016-08-30 2024-03-20 다나-파버 캔서 인스티튜트 인크. 약물 전달 조성물 및 그의 용도
US10836826B2 (en) 2016-08-30 2020-11-17 Dana-Farber Cancer Institute, Inc. Drug delivery compositions and uses thereof
US11021539B2 (en) 2016-08-30 2021-06-01 Dana-Farber Cancer Institute, Inc. Drug delivery compositions and uses thereof
AU2017321609C1 (en) * 2016-08-30 2023-11-09 Dana-Farber Cancer Institute, Inc. Drug delivery compositions and uses thereof
US10413612B2 (en) 2016-08-30 2019-09-17 Dana-Farber Cancer Institute, Inc. Method of treating cancer with a biomaterial and a toll-like receptor agonist
KR20190043162A (ko) * 2016-08-30 2019-04-25 다나-파버 캔서 인스티튜트 인크. 약물 전달 조성물 및 그의 용도
KR102497742B1 (ko) * 2016-08-30 2023-02-10 다나-파버 캔서 인스티튜트 인크. 약물 전달 조성물 및 그의 용도
WO2018045058A1 (fr) * 2016-08-30 2018-03-08 Dana-Farber Cancer Institute, Inc. Compositions d'administration de médicament et leurs utilisations
AU2017321609B2 (en) * 2016-08-30 2023-08-17 Dana-Farber Cancer Institute, Inc. Drug delivery compositions and uses thereof
EP3656400A4 (fr) * 2017-07-20 2021-10-20 National University Corporation Hokkaido University Utilisation en association d'un inhibiteur ciblant pd-1/pd-l1 et d'un inhibiteur de cox-2
CN111358752B (zh) * 2020-04-14 2023-07-14 大连市慧恩生物科技开发有限公司 一种抗癌水凝胶组合物及其制备方法
CN111358752A (zh) * 2020-04-14 2020-07-03 大连市慧恩生物科技开发有限公司 一种抗癌水凝胶组合物及其制备方法

Also Published As

Publication number Publication date
CN105983097A (zh) 2016-10-05
CN105983097B (zh) 2021-06-08

Similar Documents

Publication Publication Date Title
JP6963385B2 (ja) 抗CTLA−4抗体又は抗PD−1抗体との併用におけるIL−2Rβ選択的作動薬
WO2016119308A1 (fr) Préparation antitumorale et son procédé de préparation
US20200323957A1 (en) Combined preparations for the treatment of cancer
CN109692326A (zh) 一种蜂毒脂质纳米颗粒的应用
Chen et al. Ferritin Nanocaged Doxorubicin Potentiates Chemo‐Immunotherapy against Hepatocellular Carcinoma via Immunogenic Cell Death
AU2017370226B2 (en) Pharmaceutical composition containing insulin-like growth factor-2 and use thereof
US20230149347A1 (en) CXCR4/CXCR7 Blockade and Treatment of Human Papilloma Virus-Associated Disease
TW202114647A (zh) 用於治療具有高間質壓力的腫瘤受試者的癌症的小分子抑制劑
WO2018101831A1 (fr) Composition pharmaceutique pour utilisation dans le traitement du cancer
WO2022047815A1 (fr) Composition pharmaceutique pour immunothérapie antitumorale
CN113117088B (zh) 钙激活氯离子通道的抑制剂在肿瘤免疫疗法中的应用
CN111166878B (zh) 靶向肿瘤抗原的抗体与iNKT细胞的组合的制备方法与用途
JP2019031470A (ja) Nk細胞培養上清を含む抗腫瘍組成物およびその製造方法
CN107441076B (zh) 一种治疗癌症的联合用药物
JP2016537428A (ja) エフェドラ・アラタ抽出物及びその使用方法
CN105705164A (zh) 用于癌症治疗的免疫刺激性hiv tat衍生多肽
WO2024037521A1 (fr) Composition de traitement combinée pour traitement de tumeur et méthode de traitement combinée
CN112823808B (zh) 用于制备治疗上皮细胞癌的医药组合物及其用途
CN110177552A (zh) 用于调节pd-1信号转导的组合物
CN117323418B (zh) 一种含乳铁蛋白的胶囊在阻止hpv病毒感染中的用途
TW200908997A (en) Treatment of cellular proliferative disorders
CN118184759A (zh) Atap多肽及其疫苗的制备方法和应用
CN116966147A (zh) 一种新型sting激动剂递送体系及免疫治疗应用
CN117695242A (zh) 环二核苷酸自组装纳米粒子及其制备和应用
CN111728960A (zh) 富马酸比索洛尔联合多西他赛在制备抗肿瘤药物中的应用

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15879516

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15879516

Country of ref document: EP

Kind code of ref document: A1