CN112957475B - Composition for preventing and/or treating tumors and application thereof - Google Patents

Composition for preventing and/or treating tumors and application thereof Download PDF

Info

Publication number
CN112957475B
CN112957475B CN202110155443.8A CN202110155443A CN112957475B CN 112957475 B CN112957475 B CN 112957475B CN 202110155443 A CN202110155443 A CN 202110155443A CN 112957475 B CN112957475 B CN 112957475B
Authority
CN
China
Prior art keywords
tumor
agonist
cpg
cgamp
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202110155443.8A
Other languages
Chinese (zh)
Other versions
CN112957475A (en
Inventor
李文峰
苏小平
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to CN202110155443.8A priority Critical patent/CN112957475B/en
Publication of CN112957475A publication Critical patent/CN112957475A/en
Application granted granted Critical
Publication of CN112957475B publication Critical patent/CN112957475B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the technical field of biological medicines, and particularly relates to a composition for preventing and/or treating tumors and application thereof. A composition for the prevention and/or treatment of tumors therein, comprising one or more TLR agonists, one or more TNFRSF agonists and one or more STING agonists. The invention creatively combines the TLR agonist, the TNFRSF agonist and the STING agonist, is applied in local tumors, can induce strong anti-tumor immune response to cause the regression of tumors at injection sites and non-injection sites, and avoids systemic immune adverse reaction because the combination of the TLR agonist, the TNFRSF agonist and the STING agonist is applied in the local tumors.

Description

Composition for preventing and/or treating tumor and application thereof
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to a composition for preventing and/or treating tumors and application thereof.
Background
According to the data published by the world health organization, cancer poses the greatest burden worldwide. Malignant tumors can be considered as the primary public health care problem, which imposes a huge burden on the clinic, disturbs social standards, and erodes a large amount of economic resources. Although the incidence and mortality of malignant tumors has increased year by year, most malignant tumors lack an effective treatment regimen, and the traditional treatment modalities are still limited to radiation therapy, chemotherapy, and surgical resection. In fact, less than 30% of patients with tumors can receive the opportunity for surgical treatment, while many patients experience severe adverse effects immediately or chronically after receiving systemic or local chemotherapy, and in some cases, the adverse effects associated with treatment outweigh the benefits of treatment, or even worsen the patient's condition. Radiotherapy, chemotherapy and surgery have significant limitations in the treatment of tumors and are difficult to cure, especially metastatic tumors.
In recent years, immunotherapy has become a hot research topic for tumor therapy. Monoclonal antibodies against immune checkpoint cytotoxic T lymphocyte-associated protein 4(CTLA-4) and programmed cell death protein 1(PD-1) have shown significant therapeutic efficacy and have been approved by the FDA for cancer treatment. In addition, chimeric antigen receptor-T (CAR-T) cell therapy has been approved for the treatment of certain hematological malignancies. Tumor immunotherapy has made more and more breakthrough progress. At present, the main tumor immunotherapy means include agonistic or blockade therapy for immune checkpoints, adoptive cell therapy, genetic engineering cell therapy, tumor vaccines, and the like. These approaches, while each are well developed, tumor immunotherapy still faces a number of obstacles due to the complexity of the tumor microenvironment and the refractory nature of metastatic tumors.
Currently, tumor immunotherapy still faces three major challenges:
problem 1. in most solid tumors, the tumor immunogenicity is low. Dendritic Cells (DCs) are the most powerful Antigen Presenting Cells (APCs) that induce the formation of specific Cytotoxic T Lymphocytes (CTLs). However, in the tumor microenvironment, the DCs are mostly immature phenotypes, have poor activities, cannot present tumor antigens to effector T cells, and cannot initiate anti-tumor immune responses. This "priming disability" results in limited tumor immune efficacy.
Problem 2. A single immunotherapy or immunotherapy drug does not achieve the desired effect. Genome-wide analysis has shown that heterogeneity between tumor-bearing individuals is very significant, which results in large differences in the responsiveness of different patients to the same monotherapy, and unsatisfactory results are not achieved. For example, while immune checkpoint inhibitors can block the inhibitory effects of cell contact-dependent protein pathways on effector T cells for pancreatic cancer, there are many soluble immunosuppressive factors that inhibit the function of effector T cells in the tumor microenvironment of pancreatic cancer. Pancreatic cancer, a typical "cold" tumor, contains a large number of dense stromal cells and few Tumor Infiltrating Lymphocytes (TILs), leading to the failure of some immune checkpoint inhibitors. This forces tumor immunotherapy to shift from monotherapy directed at a single target or pathway to a combination of multi-target and multi-pathway approaches.
Problem 3. immunotoxic reaction due to systemic immune activation. Due to the systemic administration of immunotherapy, the systemic immune system of the subject is rapidly activated, which on the one hand causes objective responses against the tumor and on the other hand also brings about autoimmune adverse reactions that are difficult to recognize. For example, the gastrointestinal system is one of the organ systems most commonly affected by autoimmune inflammation. The incidence of autoimmune enterocolitis varies from 1% to 25% in patients receiving tumor immunotherapy, depending on the type of drug the patient receives. Systemic administration inevitably over-activates systemic immunity, resulting in normal tissue involvement. This forces the tumor immunotherapy to switch to local, precise dosing.
Disclosure of Invention
The invention aims to overcome the defects of the prior art and provide a composition for preventing and/or treating tumors and application thereof.
The technical scheme adopted by the invention is as follows: a composition for preventing and/or treating a tumor comprising one or more TLR agonists, one or more TNFRSF agonists, and one or more STING agonists.
The TLR agonist is a TLR9 agonist.
The TLR9 agonist is an oligonucleotide and/or hemozoin; the oligonucleotides are cytosine-phosphorothioate-guanine oligodeoxynucleotides containing unmethylated cytosine and guanine in A class, B class or C class.
The TNFRSF agonist is an OX-40 agonist.
The OX-40 agonist is one or more of an OX-40 agonist antibody, an OX40L agonist fragment, an OX-40 oligomeric receptor, and OX-40 immunoadhesin.
The OX-40 agonist is a trimeric OX40L-Fc protein, which trimeric OX40L-Fc protein comprises a fragment of one or more extracellular domains of OX 40L.
The STING agonist is a cyclic dinucleotide or a derivative thereof.
The cyclic dinucleotide is one or more of cyclic adenosine di-phosphate monophosphate, cyclic guanosine di-phosphate monophosphate, cyclic inosine monophosphate, cyclic guanosine monophosphate or cyclic adenosine monophosphate-inosine monophosphate and cyclic guanosine monophosphate-inosine monophosphate.
The use of a composition for the prevention and/or treatment of a tumour as described above in the manufacture of a medicament for the treatment or prevention of a tumour.
A medicament or a kit, characterized in that the medicament or the kit contains the composition for preventing and/or treating tumor.
The invention has the following beneficial effects: the invention innovatively combines the TLR agonist, the TNFRSF agonist and the STING agonist, and simultaneously, the TNFRSF agonist, the STING agonist and the STING agonist are applied in local tumors, so that the TNFRSF agonist can induce strong anti-tumor immune response to cause the regression of tumors at injection sites and non-injection sites.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the embodiments or the prior art descriptions will be briefly described below, it is obvious that the drawings in the following description are only some embodiments of the present invention, and it is within the scope of the present invention for those skilled in the art to obtain other drawings based on the drawings without inventive labor.
In FIG. 1, (A) is a graph of the tumor volume at the injection site after different treatments on the group A-E cervical cancer-bearing mice, confirming the antitumor ability of CPG + OX40+ cGAMP; (B) as a plot of the volume of uninjected tumors after differential treatment of cervical cancer-bearing mice in the A-E group, the results showed that only CPG + OX40+ cGAMP treatment was able to produce distant tumoricidal capacity, confirming the superiority of CPG + OX40+ cGAMP therapy for metastatic tumors.
FIG. 2 is a graph of mouse survival after various treatments of groups A-E cervical cancer-bearing mice, showing that CPG + OX40+ cGAMP significantly extended the survival time of the tumor-bearing mouse model.
FIG. 3 is a graph showing the body weight change of mice treated differently with group A-E cervical cancer-bearing mice, demonstrating that the body weight of mice treated with CPG + OX40+ cGAMP is not different from that of mice treated with blank treatment, i.e., the therapy of the present invention has no obvious toxic side effect when being applied to tumors.
In FIG. 4, CD3, CD4 and CD8 tumors in the non-injected mice were infiltrated after different treatments of mice bearing cervical cancer in groups A-E. After CPG + OX40+ cGAMP treatment, the number of tumor infiltrating lymphocytes at the position without injection is greatly increased, wherein the increase of CD4 positive lymphocytes is most remarkable, so that the anti-tumor effect of the triple combination group is optimal.
In FIG. 5, (A) is a graph of the tumor volume at the injection site after different treatments of melanoma-bearing mice in groups A-E, again demonstrating the antitumor ability of CPG + OX40+ cGAMP. (B) Is a tumor volume map of the non-injection part of a group A-E melanoma tumor-bearing mice after different treatments, and the result is similar to the experiment result of the cervical cancer tumor-bearing mice: only CPG + OX40+ cGAMP treatment was able to produce distant tumoricidal capacity, confirming the superiority of CPG + OX40+ cGAMP therapy for metastatic tumors.
FIG. 6 is a graph showing the survival of mice treated differently with melanoma-bearing mice of groups A-E, similar to the experimental results for mice bearing cervical cancer: only CPG + OX40+ cGAMP treatment significantly extended survival in tumor-bearing mouse models.
In FIG. 7, (A) shows the tumor volume of melanoma tumor-bearing mice treated by injection with combinations of CpG + cGAMP and different immunodetection point inhibitors, demonstrating that CpG + cGAMP in combination with different immunodetection point inhibitors all have anti-tumor effects in situ. (B) Tumor volumes were examined in untreated sites after injection treatment of melanoma-bearing mice with combinations of CpG + cGAMP and different immunodetection point inhibitors, demonstrating that only CpG + OX40+ cGAMP treatment was able to produce a distant tumor killing capability, demonstrating the superiority of CpG + OX40+ cGAMP therapy for metastatic tumors.
Detailed Description
To make the objects, technical solutions and advantages of the present invention more apparent, the present invention will be described in further detail with reference to the accompanying drawings.
A composition for preventing and/or treating a tumor comprising one or more TLR agonists, one or more TNFRSF agonists, and one or more STING agonists.
The invention employs a TLR agonist which, after being administered locally within a tumor, is specifically recognized by dendritic cells, activates and mediates maturation of dendritic cells. The dendritic cell surface co-stimulatory molecules are up-regulated, the antigen presenting capability is enhanced, and meanwhile, the dendritic cells generate more interferon-alpha to promote the maturation of the bone marrow dendritic cells, so that the T cells are indirectly activated. The STING agonist is used, and after the agonist is applied, the STING agonist leads to the activation of STING pathway of dendritic cells in a tumor microenvironment, promotes antigen cross presentation, increases the expression of CCR7 on the dendritic cells, and improves the transportation of homing effector T cells of antigen presenting cells by various Th1 cytokines. Therefore, the dendritic cells as the antigen presenting cells with the strongest functions are activated and proliferated in a tumor microenvironment, and the antigen presenting capacity is greatly enhanced, so that the problem of low tumor immunogenicity is solved.
The invention combines three immune targets or paths at the same time, and changes from a monotherapy mode to a mode of combining multiple targets and multiple paths. First, Toll-like receptors (TLRs) are present on many cells of the immune system and have been shown to be involved in the innate immune response, which is a key means for vertebrates to recognize and establish immune responses to foreign molecules, to recognize pathogen-associated molecular patterns of bacteria, fungi, parasites and viruses, and to provide a means to link innate and adaptive immune responses. The invention uses TLR agonist which can activate NF-kB signal and stimulate IFN-a secretion; the TNFRSF agonist is used, so that the survival time of T cells can be prolonged, the killing capacity of the T cells can be promoted, and the immunosuppressive action in a tumor microenvironment can be improved; the STING agonist used in the invention stimulates the induction of INF-beta through a STING pathway, mediates the activation of NF-kappa B and IRF-3, and then starts the transcription of INF-beta gene to generate beta-interferon. In conclusion, the combination of the agonists of the three types of targets or pathways can effectively solve the problem of tumor heterogeneity.
The invention can be applied in local tumor, avoids systemic medication, achieves the purpose of treating the tumor at the injection site by modifying local tumor microenvironment, simultaneously causes the tumor decline at the site without injection by improving the tumor immunogenicity, modifying the tumor infiltration cell types, cell factors and other factors, and avoids the occurrence of systemic autoimmune toxic and side effects.
In some embodiments of the invention, the TLR agonist is a TLR9 agonist, and a common TLR9 agonist includes an oligonucleotide that is a cytosine-phosphorothioate-guanine oligodeoxynucleotide containing unmethylated cytosine and guanine, including class a (also referred to as class D), class B (also referred to as class K), and class C, and hemozoin. The A class is composed of a PO skeleton with a CpG palindromic sequence as the center and a 3' end poly-G chain modified by PS. CpG-A ODNs stimulate plasmacytoid dendritic cells (pDCs) to produce IFN-a more strongly, but activate the TLR 9-dependent NF-kB signaling pathway less strongly, producing less pro-inflammatory cytokines (e.g., interleukin 6). Class B is the complete PS backbone structure sequence containing one or more CpG dinucleotides. CpG-B ODNs can strongly activate B cells and TLR 9-mediated NF-kB signals, but have weaker effect on stimulating the secretion of IFN-a. The Class C structure is a mixture of classes A and B. It contains the complete PS backbone and CpG palindromic sequences. CpG-C ODNs can strongly cause pDC to secrete IFN-a and can also obtain better effect when stimulating B cells. In some embodiments of the invention, the C-class TLR9 agonist is specifically selected to be a CpG ODN 2395, and the sequence of the CpG ODN 2395 is 5'-TCGTCGTTTTCGGCGCGCGCCG-3'.
In some embodiments of the invention, the TNFRSF agonist is an OX-40 agonist, and the OX-40 agonist can be an OX-40 agonist antibody, an OX40L agonist fragment, an OX-40 oligomeric receptor, and an OX-40 immunoadhesin. In some embodiments of the invention, OX-40 agonist antibodies are specifically selected, wherein the OX-40 agonist antibodies belong to a member of tumor necrosis factor superfamily, are agonist receptors expressed on the cell surface of activated CD4 positive T cells and CD8 positive T cells, and the signals of the OX-40 agonist antibodies can activate downstream NF-kappa B, PI3K and PKB pathways, and the continuous activation of the pathways can finally prolong the survival time of T cells, expand T cell memory and promote the killing capacity of T cells, and in addition, the OX-40 agonist antibodies can also improve the immunosuppressive action in a tumor microenvironment by inhibiting the differentiation and activity of regulatory T cells (Tregs) and further enhance the function of effector T cells. In some embodiments of the invention, the OX-40 agonist antibody is specifically selected from a trimeric OX40L-Fc protein comprising a fragment of one or more extracellular domains of OX40L, or a full length human IgG1 antibody.
In some embodiments of the invention, the STING agonist is a cyclic dinucleotide, or a derivative thereof, wherein the cyclic dinucleotide is cyclic-di-adenylate monophosphate (CDA), cyclic-di-guanylate monophosphate (CDG), cyclic-di-inosine monophosphate (CDI), cyclic-guanylate monophosphate (cGAMP), cyclic-adenosine monophosphate-inosine monophosphate, or cyclic-guanylate-inosine monophosphate, and in some embodiments of the invention, cyclic-guanylate monophosphate (cGAMP) is selected. The STING agonist, cGAMP, is a cytoplasmic DNA sensor that serves as a second messenger to stimulate the induction of INF-beta via the STING pathway, mediate the activation of NF- κ B and IRF-3, and then initiate the transcription of the INF-beta gene, producing beta-interferon.
The application of the composition for preventing and/or treating the tumor in preparing the medicine for treating or preventing the tumor. The composition for preventing and/or treating tumors provided by the invention can be used as an active ingredient to be combined with a pharmaceutically acceptable carrier to prepare a composite biological preparation as a medicinal preparation for preventing and/or treating tumors.
A kit for preventing and/or treating tumor comprising the composition for preventing and/or treating tumor as described above, wherein one or more TLR agonist, one or more TNFRSF agonist and one or more STING agonist are mixed as a single agent or separately provided as a plurality of agents. The agonists for the three targets or pathways may be administered as a combined biological formulation on a localized tumor, or may be administered separately and simultaneously to provide enhanced therapeutic or prophylactic effects on the disease. Other chemical and/or biological therapeutic agents may also be included in the kit, such as anti-angiogenic agents, growth inhibitory agents, other anti-neoplastic agents currently known or combinations thereof.
The following is a sample preparation process used in some embodiments of the invention.
Culture of TC-1 cervical cancer cell line
TC-1 murine cervical carcinoma cells were cultured in RPMI 1640 complete medium containing 10% heat-inactivated fetal bovine serum, penicillin 100U/mL, streptomycin 100U/mL, cultured in a 5% CO2 incubator at 37 ℃ with the culture medium changed daily and digested with 0.1% trypsin every 3 days for passaging. When the cells are in the logarithmic growth phase, digesting the cells, centrifuging at 1000 r/min, counting the number of living cells, and then suspending in balanced salt solution PBS to prepare suspension containing 4 x 10^6 cervical cancer cells per 1 mL.
Secondly, establishing a TC-1 cervical carcinoma C57BL/6 tumor-bearing mouse model
Healthy, pure C57BL/6 mice, 30 females, on SPF scale, 6-8 weeks, body weight (20. + -.2) g. The temperature of the breeding environment is 22-25 ℃, the humidity is 45-50%, the illumination period is 12h illumination/12 h darkness, and water is freely drunk/eaten. The replacement of feed, drinking water and bedding materials is carried out once every 3 days by the mice, wherein the feed, the drinking water and the bedding materials required by the mice are sterilized by high pressure. Mice were acclimatized for one week prior to experimental treatment. And (2) inoculating the cultured TC-1 tumor cells to the left groin of a normal mouse through subcutaneous inoculation (2 x 10 a 5 cells are inoculated to each mouse), performing the same operation on the right groin of the mouse after two days to construct a cervical cancer bilateral tumorigenic animal model, and successfully modeling when the left groin tumor grows to 20-30mm 2.
Thirdly, B16 culture of murine melanoma cell line
B16 murine melanoma cell line was cultured in DMEM complete medium containing 10% heat-inactivated fetal bovine serum, penicillin 100U/mL, streptomycin 100U/mL in a 5% CO2 incubator at 37 ℃ with daily change of the culture medium and digestion with 0.1% trypsin every 3 days for passaging. When the cells are in the logarithmic growth phase, the cells are digested, centrifuged at 1000 r/min, the number of the living cells is counted and then the living cells are resuspended in a balanced salt solution PBS to prepare a suspension containing 4 multiplied by 10^6 melanoma cells per 1 mL.
Fourthly, establishing a B16 melanoma C57BL/6 tumor-bearing mouse model
Healthy, pure C57BL/6 mice, 30 females, on SPF scale, 6-8 weeks, body weight (20. + -.2) g. The temperature of the breeding environment is 22-25 ℃, the humidity is 45-50%, the illumination period is 12h illumination/12 h darkness, and water is freely drunk/eaten. The replacement of feed, drinking water and bedding materials is carried out once every 3 days by the mice, wherein the feed, the drinking water and the bedding materials required by the mice are sterilized by high pressure. Mice were acclimatized for one week prior to experimental treatment. The cultured B16 melanoma cells are inoculated to the left groin of a normal mouse through subcutaneous inoculation (each mouse is inoculated with 2 x 10^5 cells), the same operation is carried out on the right groin of the mouse after two days, a melanoma bilateral tumorigenesis animal model is constructed, and the molding is successful when the left groin tumor grows to 20-30mm 2.
The following are some specific examples of the present invention.
Example 1 TC-1 cervical carcinoma C57BL/6 tumor-bearing mouse models were grouped and treated with different doses
The mice were randomly divided into 5 groups of 5-6 mice each, and a total of 27 mice were included. Group A: blank control group, each mouse left side inguinal tumor injection PBS 50 ul; group B: CpG 2395 treatment groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this trial)); group C: CPG + OX40 treated groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), OX40(1-1000ug (preferably 5-200ug, as 30ug in this experiment)); group D: cGAMP treated groups, each mouse was intratumorally injected with cGAMP (1-1000ug (preferably 1-200ug, as 20ug in this experiment)) in the left groin; group E: groups treated with triple (CPG + OX40+ cGAMP) were each injected intratumorally with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), OX40(1-1000ug (preferably 5-200ug, as 30ug in this experiment)), cGAMP (1-1000ug (preferably 1-200ug, as 20ug in this experiment)). After the administration, measurement of tumor size and observation of mouse status were performed every 2 days, and tumor volume = (long diameter × short diameter) = short diameter)/2, and the observation was continued.
The following are observations from the different test groups described above:
a,Observe the tumor volume difference between the injection site and the non-injection site of each group of cervical carcinoma
The A-E group of tumor-bearing mice were administered intratumorally to the left inguinal: tumor volume sizes were observed and recorded for the a-E group at injection (left groin) and non-injection (right groin) following PBS treatment, CPG + OX40 treatment, cGAMP treatment, CPG + OX40+ cGAMP treatment. The results are shown in FIG. 1 (A): for the injected tumors, the CPG group and CPG + OX40+ cGAMP treated group were the best and there was no significant statistical difference between the two. The tumor volumes of the groups A-E tumor-bearing mice at the non-injection site are shown in FIG. 1 (B), and the results show that: only after CPG + OX40+ cGAMP treatment, growth inhibition of the non-injected tumors appeared, and none of the remaining groups appeared. This confirms the excellent efficacy of the present invention against metastatic tumors.
II,Observing the life cycle difference of different tumor-bearing mice of each group of cervical cancer after different drug administration treatments
The A-E group of tumor-bearing mice were administered intratumorally to the left inguinal: survival of group a-E tumor-bearing mice was observed and recorded following PBS treatment, CPG + OX40 treatment, cGAMP treatment, CPG + OX40+ cGAMP treatment. The results are shown in FIG. 2: the CPG + OX40+ cGAMP treatment group remarkably prolongs the survival period of the tumor-bearing mouse model, the CPG + cGAMP treatment has the effect of slightly prolonging the survival period, and the rest groups have no obvious benefit on the survival period of the tumor-bearing mice.
III,By monitoring the body weight of each group of tumor-bearing mice with cervical cancer, the toxic and side effects of different treatments and each group of mice are observed Abnormal manifestation of (2)
The A-E group of tumor-bearing mice were administered intratumorally to the left inguinal: after PBS treatment, CPG + OX40 treatment, cGAMP treatment, CPG + OX40+ cGAMP treatment, group a-E tumor-bearing mouse body weights were observed and recorded. The results are shown in FIG. 3: abnormal performance does not appear in all groups of mice, and the body weight of the mice in the B-E group and the body weight of the mice in the blank control A group do not have statistical difference, namely: after the mice are administrated with CPG + OX40+ cGAMP in tumors, no obvious toxic or side effect occurs, the general condition is good, and no abnormal expression occurs.
Fourthly, the,The CD3, CD4 and CD8 cell condition of the tumor infiltrated at the non-injection part of each tumor-bearing mouse of the cervical cancer groups
The A-E group of tumor-bearing mice were administered intratumorally to the left inguinal: after PBS treatment, CPG + OX40 treatment, cGAMP treatment, CPG + OX40+ cGAMP treatment, the tumor-infiltrated CD3, CD4 and CD8 cells of the uninjected tumor-bearing mice in the A-E group were examined by flow cytometry. The results are shown in FIG. 4: the tumors treated by CPG, CPG + OX40 and CPG + OX40+ cGAMP all caused the increase of the number of tumor infiltrating lymphocytes at the non-injected sites, but the increased number of CD4 positive T lymphocytes in each group is most remarkable as in the CPG + OX40+ cGAMP treated group, so that the treatment effect is optimal.
Example 2 melanoma B16 mice model C57BL/6 bearing tumor were grouped and treated with different doses
The mice were randomly divided into 5 groups of 5-6 mice each, and a total of 27 mice were included. Group A: blank control group, each mouse left side inguinal tumor injection PBS 50 ul; group B: CpG 2395 treatment groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this trial)); group C: CPG + OX40 treated groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), OX40(1-1000ug (preferably 1-200ug, as 30ug in this experiment)); group D: cGAMP-treated group, each mouse was intratumorally injected with cGAMP (1-1000ug (preferably 1-200ug, as 20ug in this experiment)); group E: the triple (CPG + OX40+ cGAMP) treated group was injected intratumorally with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), OX40(1-1000ug (preferably 1-200ug, as 30ug in this experiment)), cGAMP (1-1000ug (preferably 1-200ug, as 20ug in this experiment)) per mouse in the left groin. After the administration, tumor size measurement and observation of mouse status were performed every 2 days, and tumor volume = (long diameter = short diameter)/2, and observation was continued.
The following are observations made in the different test groups described above:
a,Observing the tumor volume difference between the injection site and the non-injection site of each group of melanoma
The group A-E tumor-bearing mice were administered with the following drugs: tumor volume sizes were observed and recorded for the a-E group injection site (left groin) and non-injection site (right groin) after PBS treatment, CPG + OX40 treatment, cGAMP treatment, CPG + OX40+ cGAMP treatment. Results as shown in fig. 5, the results of the melanoma animal experiments were similar to cervical cancer: for the injected tumors, the CPG group and CPG + OX40+ cGAMP treated group performed best without significant statistical difference between the two. Similarly, the tumor volumes of the groups A-E tumor-bearing mice at the non-injection sites are shown in FIG. 6, and the results show that: only after CPG + OX40+ cGAMP treatment, growth inhibition of the non-injected tumors appeared, and none of the remaining groups appeared. This confirms the excellent efficacy of the present invention on metastatic tumors and does not differentiate between cancer species.
II,Observing the life cycle difference of various groups of melanoma mice after different drug administration treatments
For A-E group tumor-bearing miceGroinThe tumor was given separately: survival of group a-E tumor-bearing mice was observed and recorded following PBS treatment, CPG + OX40 treatment, cGAMP treatment, CPG + OX40+ cGAMP treatment. The results are shown in fig. 7, which are similar to those of cervical cancer animal experiments: only the CPG + OX40+ cGAMP treatment group remarkably prolongs the survival period of the tumor-bearing mouse model, the CPG + cGAMP treatment has the effect of slightly prolonging the survival period, and the rest groups have no obvious benefit on the survival period of the tumor-bearing mice.
In conclusion, the anti-tumor composition or method containing the TLR agonist, the TNFRSF agonist and the STING agonist has a strong immunotherapy effect on tumors, modifies a tumor microenvironment to generate an anti-tumor distant vaccine effect, has an excellent curative effect on metastatic tumors, and more importantly, the anti-tumor effect obtains similar effect evidence in different cancer species, so that the anti-tumor composition or method can be applied to anti-tumor treatment without distinguishing the cancer species. Meanwhile, the invention avoids the toxic and side effects of immunotherapy caused by systemic administration, and overcomes the general problems of the current tumor immunotherapy.
Example 3 grouping and administration of B16 melanoma C57BL/6 tumor-bearing mouse models
To confirm whether other drug combinations also have such systemic anti-tumor effects, we also performed various drug combination experiments. The mice were randomly divided into 9 groups of 6 mice each, and a total of 54 mice were included. Group A: PBS treatment group, 50ul PBS was injected intratumorally in the left groin of each mouse; group B: CPG + cGAMP treated groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), cGAMP (1-1000ug (preferably 1-200ug, as 10ug in this experiment)); group C: CPG + OX40+ cGAMP treated groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), OX40(1-1000ug (preferably 1-200ug, as 30ug in this experiment)), cGAMP (1-1000ug (preferably 1-200ug, as 10ug in this experiment)); group D: CPG + LAG-3+ cGAMP treated groups, each mouse was injected intratumorally with CpG 2395(1-1000ug (preferably 1-200ug, e.g. 50ug in this experiment)), LAG-3(1-1000ug (preferably 5-200ug, e.g. 30ug in this experiment), cGAMP (1-1000ug (preferably 1-200ug, e.g. 10ug in this experiment)); group E: CPG + TIM-3+ cGAMP treated groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), TIM-3(1-1000ug (preferably 1-200ug, as 30ug in this experiment), cGAMP (1-1000ug (preferably 1-200ug, as 10ug in this experiment)); and F group: CPG + BTLA + cGAMP treated groups, each mouse was intratumorally injected with CpG 2395(1-1000ug (preferably 1-200ug, e.g., 50ug in this experiment)), BTLA (1-1000ug (preferably 1-200ug, e.g., 30ug in this experiment)), cGAMP (1-1000ug (preferably 1-200ug, e.g., 10ug in this experiment)); group G: CPG + NKG2A + cGAMP treated groups, each mouse was injected intratumorally with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), NKG2A (1-1000ug (preferably 1-200ug, as 30ug in this experiment)), cGAMP (1-1000ug (preferably 1-200ug, as 10ug in this experiment)); group H: CPG + CD28+ cGAMP treated groups, each mouse was injected intratumorally with CpG 2395(1-1000ug (preferably 1-200ug, as 50ug in this experiment)), CD28(1-1000ug (preferably 1-200ug, as 30ug in this experiment), cGAMP (1-1000ug (preferably 1-200ug, as 10ug in this experiment)); group I: the CPG +41BB + cGAMP treated group, each mouse was injected intratumorally with CpG 2395(1-1000ug (preferably 1-200ug, e.g., 50ug in this experiment)), 41BB (1-1000ug (preferably 1-200ug, e.g., 30ug in this experiment)), cGAMP (1-1000ug (preferably 1-200ug, e.g., 10ug in this experiment)). After the administration, measurement of tumor size and observation of mouse status were performed every 2 days, and tumor volume = (long diameter × short diameter) = short diameter)/2, and the observation was continued.
The following are observations made in the different test groups described above:
firstly, observing the tumor volume difference between the injection site and the non-injection site of each group of melanoma
The A-I group tumor-bearing mice are respectively given with the following tumors in the left inguinal: after PBS treatment, CPG + cGAMP treatment, CPG + OX40+ cGAMP treatment, CPG + LAG-3+ cGAMP treatment, CPG + TIM-3+ cGAMP treatment, CPG + BTLA + cGAMP treatment, CPG + NKG2A + cGAMP treatment, CPG + CD28+ cGAMP treatment, and CPG +41BB + cGAMP treatment, the tumor volume sizes at the group A-I injection site (left groin) and the non-injection site (right groin) were observed and recorded. The results are shown in FIG. 7 (A): for the injected tumors, all treatment groups were significantly effective relative to the PBS group. The tumor volumes of the groups A-I tumor-bearing mice at the non-injection sites are shown in FIG. 7 (B), and the results show that: only after CPG + OX40+ cGAMP treatment did growth arrest in the non-injected tumors and none of the remaining groups. This confirms the excellent efficacy of the present invention against metastatic tumors.
While the invention has been described in connection with what is presently considered to be the most practical and preferred embodiment, it is to be understood that the invention is not to be limited to the disclosed embodiment, but on the contrary, is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.

Claims (3)

1. A composition for preventing and/or treating tumors, comprising: comprising a TLR agonist, a TNFRSF agonist, and a STING agonist;
the TLR agonist is a C-class TLR9 agonist CpG ODN 2395;
the TNFRSF agonist is an OX-40 agonist, and the OX-40 agonist is an OX-40 agonist antibody;
the STING agonist is cGAMP.
2. Use of the composition for treating tumor according to claim 1 in the preparation of a medicament or kit for treating tumor, wherein the tumor is cervical cancer or melanoma.
3. A medicament or kit comprising the composition for treating tumor according to claim 1, wherein the tumor is cervical cancer or melanoma.
CN202110155443.8A 2021-02-04 2021-02-04 Composition for preventing and/or treating tumors and application thereof Active CN112957475B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110155443.8A CN112957475B (en) 2021-02-04 2021-02-04 Composition for preventing and/or treating tumors and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110155443.8A CN112957475B (en) 2021-02-04 2021-02-04 Composition for preventing and/or treating tumors and application thereof

Publications (2)

Publication Number Publication Date
CN112957475A CN112957475A (en) 2021-06-15
CN112957475B true CN112957475B (en) 2022-07-22

Family

ID=76273766

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110155443.8A Active CN112957475B (en) 2021-02-04 2021-02-04 Composition for preventing and/or treating tumors and application thereof

Country Status (1)

Country Link
CN (1) CN112957475B (en)

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2609118T (en) * 2010-08-23 2017-03-22 Univ Texas Anti-ox40 antibodies and methods of using the same
JP6684215B2 (en) * 2013-08-28 2020-04-22 ピーシーアイ バイオテック エイエス Vaccination compound and immunization compound, and vaccination method and immunization method
PE20180926A1 (en) * 2015-05-29 2018-06-08 Bristol Myers Squibb Co ANTIBODIES AGAINST MEMBER 4 OF THE TUMOR NECROSIS FACTOR RECEPTOR SUPERFAMILY (OX40) AND ITS USES
CN106539757A (en) * 2016-03-20 2017-03-29 聊城市奥润生物医药科技有限公司 Application of the ring dinucleotide cGAMP- liposomees in antitumor
CA3030647A1 (en) * 2016-07-20 2018-01-25 Igm Biosciences, Inc. Multimeric ox40 binding molecules and uses thereof
CN110003338B (en) * 2019-04-16 2021-04-23 北京免疫方舟医药科技有限公司 anti-OX 40 antibodies and uses thereof
WO2020259667A1 (en) * 2019-06-28 2020-12-30 江苏恒瑞医药股份有限公司 Use of combination of tlr agonist and anti-ox40 antibody or antigen binding fragment thereof in preparation of medicament for treating tumors
CN111704671B (en) * 2020-08-19 2020-11-24 广东赛尔生物科技有限公司 OX40 antibodies and their use in treating cancer

Also Published As

Publication number Publication date
CN112957475A (en) 2021-06-15

Similar Documents

Publication Publication Date Title
JP6954648B2 (en) Treatment of solid tumors or lymphoid tumors with combination therapy
Murad et al. CpG oligodeoxynucleotides as TLR9 agonists: therapeutic applications in cancer
Wood et al. A pilot study of autologous cancer cell vaccination and cellular immunotherapy using anti-CD3 stimulated lymphocytes in patients with recurrent grade III/IV astrocytoma
AU2019255370B2 (en) Synthetic RIG-I-like receptor agonists
CN107488636A (en) A kind of immunocyte of anti-HER2 Chimeric antigen receptors modification for carrying molecular switch and its application
CN107557337A (en) A kind of immunocyte of safety-type Chimeric antigen receptor modifications of anti-ROR1 and its application
EP0941317B1 (en) Insulin-like growth factor 1 receptor (igf-1r) antisense oligonucleotide-treated breast cancer cells composition
CN101161288B (en) Enhanced tumour cell cracking article of oligonucleotide and application thereof in preparing medicine for treating tumour
Rayburn et al. Experimental therapy for colon cancer: anti-cancer effects of TLR9 agonism, combination with other therapeutic modalities, and dependence upon p53
Fujii et al. Cancer immunotherapy using artificial adjuvant vector cells to deliver NY‐ESO‐1 antigen to dendritic cells in situ
US7662792B2 (en) Modulation of Fas and FasL expression
CN109937051A (en) Treat the raised method of TIM-3
CN112957475B (en) Composition for preventing and/or treating tumors and application thereof
CN113117088A (en) Use of calcium-activated chloride channel inhibitors in tumor immunotherapy
JP5079962B2 (en) A genetically modified cell that expresses a TGFβ inhibitor and is a lung cancer cell
KR102584276B1 (en) Combination therapy with her2 vaccine, and immune checkpoint inhibitors
CN113789347B (en) Application of FGL1/CAIX double-target vaccine in treatment of renal cancer and renal cancer lung metastasis
CN116585315A (en) Compositions for modulating PD-1 signaling
US20230346901A1 (en) Methods and vaccine compositions to treat cancers
KR101976782B1 (en) Composition for preventing or treating cancer comprising soluble common gamma chain inhibitor
Tarhini et al. Early development of the toll-like receptor 9 agonist, PF-3512676, for the treatment of patients with advanced cancers
Cavanagh et al. Chemotherapy followed by syngeneic dendritic cell injection in the mouse: findings and implications for human treatment
Rwandamuriye Biomaterial-assisted delivery of immunotherapy: a novel approach to prevent cancer recurrence
KR20160026034A (en) Pharmaceutical composition for preventing or treating cancers comprising dendritic cells with Foxp3 gene silenced
CN116271090A (en) DNA nano cage with TLR9 agonist activity and application thereof in preparation of antitumor drugs

Legal Events

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