WO2020211857A1 - 用于免疫增强的锰组合物 - Google Patents
用于免疫增强的锰组合物 Download PDFInfo
- Publication number
- WO2020211857A1 WO2020211857A1 PCT/CN2020/085507 CN2020085507W WO2020211857A1 WO 2020211857 A1 WO2020211857 A1 WO 2020211857A1 CN 2020085507 W CN2020085507 W CN 2020085507W WO 2020211857 A1 WO2020211857 A1 WO 2020211857A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- virus
- manganese
- colloidal
- cancer
- precipitated
- Prior art date
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K33/00—Medicinal preparations containing inorganic active ingredients
- A61K33/24—Heavy metals; Compounds thereof
- A61K33/32—Manganese; Compounds thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
- A61P17/06—Antipsoriatics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/10—Antimycotics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
- A61P31/16—Antivirals for RNA viruses for influenza or rhinoviruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
- A61P31/18—Antivirals for RNA viruses for HIV
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
- A61P31/22—Antivirals for DNA viruses for herpes viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
- A61P33/02—Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
- A61P33/02—Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
- A61P33/06—Antimalarials
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
- A61P33/10—Anthelmintics
- A61P33/12—Schistosomicides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/04—Immunostimulants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55505—Inorganic adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2300/00—Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- the present invention provides divalent manganese colloid or divalent manganese neoprecipitate for enhancing immunity, and its use for enhancing immunity, which can be used as an immune adjuvant, antiviral or antitumor, for example.
- the body protects the body from foreign pathogens through the natural immune system and adaptive immune system. Innate immunity can promote adaptive immunity.
- Innate immunity can promote adaptive immunity.
- the body When the body is infected by a pathogen, it first initiates a natural immune response, recognizes the pathogen's pathogen-related molecular patterns through pattern recognition receptors, and activates a variety of signaling pathways, such as TLR pathway (1), RLR pathway (2), cGAS-STING Pathway (3), inflammasome activation (4), etc.
- the activation of these pathways leads to the production of many downstream cytokines, including type I interferon, IL-1 ⁇ , IL-18 and so on.
- Type I interferon can activate the JAK-STAT pathway through autocrine and paracrine pathways, induce the expression of a large number of antiviral genes, and achieve the effect of resisting viral infections (5,6).
- type I interferon can promote the maturation of antigen-presenting cells (7).
- the antigen-presenting cells present pathogenic microorganisms or tumor antigens to T cells, and activate antigen-specific CD4 + T cells and CD8 + T cells; it can also Promote the activation of B cells to produce antigen-specific antibodies; and promote the production of memory B cells and immune memory.
- the pro-inflammatory factor IL-1 ⁇ can directly act on CD4 + T cells and promote the proliferation of T cells (8); IL-18 can effectively enhance Th1 immune response, and can also promote the proliferation and cytotoxicity of T cells and NK cells (9) ).
- Aluminum adjuvants have been widely used since 1920, and are used in hepatitis A (HAV) vaccine, hepatitis B (HBV) vaccine, DTP vaccine, human papillomavirus (HPV) vaccine, Haemophilus influenzae (HiB) ) Vaccines, etc.
- HAV hepatitis A
- HBV hepatitis B
- HPV human papillomavirus
- HiB Haemophilus influenzae
- AS04 is an adjuvant containing MPL and aluminum salts and is used in HPV vaccines and HBV vaccines.
- the adjuvant can activate NF-kB, produce pro-inflammatory factors, and has the ability to activate Th1 response that the aluminum adjuvant does not originally have.
- MF59 is a water-in-oil adjuvant made with squalene as the oil phase. It contains degradable squalene, Tween 80 and Span 85 and is used in influenza vaccines. The mechanism is still unclear. Its half-life in the body is 42 hours, and it can activate both Th1 and Th2 responses.
- AS03 is an oil-in-water adjuvant containing alpha tocopherol, squalene, and Tween 80, which is used in influenza vaccines, but the H1N1 influenza virus vaccine Pandemrix TM containing AS03 may cause narcolepsy (11). AS03 can induce the production of pro-inflammatory factors by activating the NF-kB pathway, recruit immune cells, and induce antibody production.
- ligands that activate the cGAS-STING pathway can be used as adjuvants, such as DMXAA (12), c-di-GMP (13), cGAMP (14), chitosan (15).
- CN107412260A discloses that divalent manganese is a cGAS-STING pathway activator, which has the function of enhancing immunity, for example, it can be used as an immune adjuvant.
- divalent manganese itself still has some defects, and an improved solution is needed to eliminate at least one of the defects.
- the inventors further studied the immuno-enhancing effect of divalent manganese and found that although the divalent manganese solution can produce an immune-enhancing effect in the body, this immune-enhancing effect is not high enough. More importantly, during the research process, it was also found that when the concentration of the divalent manganese solution is increased to improve the immune enhancement effect, it is easy to produce manganese precipitation, which makes it impossible to obtain a uniform and stable high-concentration manganese solution, which makes the experiment difficult. Repeatability cannot be effectively guaranteed. This manganese precipitation gradually agglomerates and grows with increasing storage time.
- the inventors had to use divalent manganese precipitation to continue the research on its immune enhancement effect. Due to the negligence of the experimental operation, the inventor inadvertently used divalent manganese precipitates left for different periods of time. The inventors unexpectedly discovered that the divalent manganese precipitates with different storage time have different immune enhancement effects, and the immuno-enhancing effects of the newborn precipitates are significantly better than the manganese precipitation with longer storage time. The inventor further compared the divalent manganese solution with the divalent manganese precipitation stored for different time, and unexpectedly found that the immunoenhancing effect of the new manganese salt precipitation was even significantly better than that of the divalent manganese solution.
- divalent manganese compounds can form colloidal solutions. What is even more surprising is that this manganese colloid shows an immune-enhancing effect comparable to or even stronger than that of the new manganese precipitation.
- the present invention provides an immune-enhancing composition comprising nascent precipitated manganese, colloidal manganese, and/or a source capable of forming nascent precipitated manganese and/or colloidal manganese.
- the newly precipitated manganese and/or colloidal manganese are selected from the following forms: manganese phosphate, manganese carbonate, manganese hydroxide and any mixtures thereof.
- the time for the nascent precipitated manganese to be converted from a non-precipitated form to a precipitated form does not exceed 1 day, or does not exceed 24, 22, 20, 18, 16, 15, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 hour, or no more than 60, 50, 45, 40, 35, 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 minute.
- the present invention provides a vaccine composition comprising
- Newly precipitated manganese, colloidal manganese and/or can form a source of new precipitated manganese and or colloidal manganese
- components A and B may be in the same and/or separate containers.
- the vaccine immunogen is derived from viruses, bacteria and/or parasites,
- the virus is selected from: DNA virus and RNA virus
- the virus is selected from: Herpesviridae, Rhabdoviridae, Filoviridae, Orthomyxoviridae, Paramyxoviridae, Coronaviridae, Mini RNAviridae, hepatotropic DNAviridae, flaviviridae, papillomaviridae, poxviridae, and retroviridae
- the virus is selected from: herpes simplex virus, vesicular stomatitis virus, vaccinia virus , HIV and HBV;
- the bacteria are selected from Gram-negative bacteria and Gram-positive bacteria, preferably the bacteria are selected from Streptococcus pneumoniae, Haemophilus influenzae, Salmonella, and meningitis. Meningococcus, Staphylococcus epidermidis, Staphylococcus aureus, Escherichia coli, Klebsiella pneumoniae, Klebsiella oxytoca ), Enterobacter cloacae, Citrobacter freundii, Pseudomonas aeruginosa and Acinetobacter baumanni;
- the vaccine immunogen is derived from influenza virus, hepatitis virus (such as hepatitis A virus, hepatitis B virus), polio virus, rabies virus, HPV virus, encephalitis virus (such as Japanese encephalitis virus), mumps Virus, rubella virus, tetanus bacillus, bacillus pertussis, diphtheria bacillus, leprosy bacillus, tuberculosis bacillus, meningococcus, pneumococcus and any combination thereof.
- hepatitis virus such as hepatitis A virus, hepatitis B virus
- polio virus rabies virus
- HPV virus encephalitis virus
- mumps Virus rubella virus
- tetanus bacillus bacillus pertussis
- diphtheria bacillus diphtheria bacillus
- leprosy bacillus tuberculosis bacill
- the present invention provides a method of manufacturing an immune enhancing composition, which comprises
- the newly precipitated manganese and/or colloidal manganese are selected from manganese phosphate, manganese carbonate, manganese hydroxide and any mixtures thereof.
- the time for the nascent precipitated manganese to be converted from a non-precipitated form to a precipitated form does not exceed 1 day, or does not exceed 24, 22, 20, 18, 16, 15, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 hour, or no more than 60, 50, 45, 40, 35, 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 minute.
- the present invention provides a method of manufacturing a vaccine composition, which comprises
- the vaccine immunogen is derived from viruses, bacteria and/or parasites, as described above, and will not be repeated here.
- the present invention provides a source of nascent precipitated manganese, colloidal manganese and/or capable of forming nascent precipitated manganese and or colloidal manganese in the preparation of an immune enhancement composition or vaccine composition for improving innate immunity and/or adaptive immunity Use in.
- the nascent precipitated manganese, colloidal manganese and/or a source capable of forming nascent precipitated manganese and or colloidal manganese improves innate immunity and/or adaptive immunity by increasing the expression of type I interferon.
- the newly-precipitated manganese, colloidal manganese and/or the source capable of forming newly-precipitated manganese and or colloidal manganese improve innate immunity and/or adaptive immunity by inducing cleavage of inflammatory factors to produce active forms.
- the nascent precipitated manganese, colloidal manganese and/or a source capable of forming nascent precipitated manganese and or colloidal manganese improve innate immunity and/or adaptive immunity by promoting antibody production.
- the present invention provides a method of enhancing immunity, which comprises administering the immune enhancing composition to a subject in need thereof.
- the immune enhancement is, for example, A) improving innate immunity and/or adaptive immunity, B) increasing the expression of type I interferon, C) inducing the production of inflammatory factors in active form, D) promoting antibody production, E) promoting T Cell proliferation, and/or F) promote the maturation of dendritic cells.
- the administration is selected from intramuscular injection, intradermal injection, subcutaneous injection, intravenous injection, mucosal administration, and any combination thereof.
- the enhanced immunity is used to prevent and/or treat diseases, such as bacterial infections, fungal infections, viral infections, parasitic infections, tumors, and autoimmune diseases.
- said virus is selected from: DNA virus and RNA virus, preferably said virus is selected from: Herpesviridae, Rhabdoviridae, Filoviridae, Orthomyxoviridae, Paramyxoviridae, Coronaviridae, Mini RNAviridae, hepatotropic DNAviridae, flaviviridae, papillomaviridae, poxviridae, and retroviridae, specifically the virus is selected from: herpes simplex virus, vesicular stomatitis virus, vaccinia virus, HIV, influenza virus, hepatitis virus (such as hepatitis A virus, hepatitis B virus), polio virus, rabies virus, HPV virus, encephalitis virus (such as Japanese encephalitis virus), mumps virus, rubella virus and any combination thereof.
- Herpesviridae Rhabdoviridae, Filoviridae, Orthomyxoviridae, Paramyx
- said bacteria are selected from Gram-negative bacteria and Gram-positive bacteria, preferably said bacteria are selected from Streptococcus pneumoniae, Haemophilus influenzae (Haemophilus influenzae), Salmonella (Salmonella), meningitis double Meningococcus, Staphylococcus epidermidis, Staphylococcus aureus, Escherichia coli, Klebsiella pneumoniae, Klebsiella oxytoca ), Enterobacter cloacae, Citrobacter freundii, Pseudomonas aeruginosa, Acinetobacter baumanni, Tetanus, Pertussis, Diphtheria , Leprosy Bacillus, Mycobacterium tuberculosis, Meningococcus, Pneumococcus and any combination thereof.
- the parasite is an intracellular parasite, preferably selected from Plasmodium, Toxoplasma, Trypanosoma, Schistosoma, Filaria, and Leishmania.
- the autoimmune disease is selected from type I diabetes, psoriasis, rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis.
- the tumor is selected from ovarian cancer, lung cancer, gastric cancer, breast cancer, liver cancer, pancreatic cancer, skin cancer, malignant melanoma, head and neck cancer, sarcoma, cholangiocarcinoma, bladder cancer, kidney cancer, colon cancer, placental choriocarcinoma, Cervical cancer, testicular cancer, uterine cancer and leukemia.
- the immune enhancing composition is administered to a subject in need thereof together with another preventive/therapeutic agent.
- the present invention provides an immunization method, which comprises
- component A and component B can be administered at the same time or at different times when they are in different containers;
- the immune enhancing composition of the present invention and optionally the vaccine immunogen are administered to a subject in need thereof.
- the administration is selected from intramuscular injection, intradermal injection, subcutaneous injection, intravenous injection, mucosal administration, and any combination thereof.
- the immunization is used to prevent diseases such as bacterial infections, fungal infections, viral infections, parasitic infections, tumors and autoimmune diseases.
- diseases such as bacterial infections, fungal infections, viral infections, parasitic infections, tumors and autoimmune diseases.
- the disease is the same as that described above, and will not be repeated here.
- the immune enhancing composition is administered to a subject in need thereof together with another preventive/therapeutic agent.
- the present invention provides the use of newly precipitated manganese, colloidal manganese and/or a source capable of forming newly precipitated manganese and/or colloidal manganese in the preparation of drugs for enhancing type I interferon.
- the present invention provides the use of newly precipitated manganese, colloidal manganese and/or a source capable of forming newly precipitated manganese and/or colloidal manganese in the preparation of a medicament for inducing the cleavage of inflammatory factors to produce an active form.
- the present invention provides the use of nascent precipitated manganese, colloidal manganese and/or a source capable of forming nascent precipitated manganese and/or colloidal manganese in the preparation of a drug for stimulating antibody production.
- the present invention provides the use of nascent precipitated manganese, colloidal manganese and/or a source capable of forming nascent precipitated manganese and/or colloidal manganese as an immune adjuvant.
- the immune adjuvant activates T cell activation and/or antibody production.
- the immune adjuvant is used in a vaccine composition for the treatment of diseases selected from bacterial infections, viral infections, parasites, autoimmune diseases and cancer.
- the present invention provides a kit for immunization, which comprises: a first container containing one or more antigens therein; and a second container containing newly precipitated manganese, colloidal manganese, and /Or can form a source of newly precipitated manganese and/or colloidal manganese.
- the first container and/or the second container further comprise a pharmaceutically acceptable carrier.
- the kit is used for one or more purposes of the present invention.
- the antigen and the nascent precipitated manganese, colloidal manganese and/or the source capable of forming nascent precipitated manganese and/or colloidal manganese are located in the same container.
- the antigen used in the present invention is selected from virus or bacterial or parasite antigens, for example, hepatitis A, B, C, D and E-3 hepatitis virus, HIV, herpes virus 1, Types 2, 6 and 7, cytomegalovirus, varicella-zoster virus, papilloma virus, Epstein-Barr virus, influenza virus, parainfluenza virus, adenovirus, Bunya virus (Hantavirus), Coxsackie virus, Picornavirus, rotavirus, respiratory syncytial virus, poxvirus, rhinovirus, rubella virus, papilloma virus, mumps virus and measles virus, mycobacteria that cause tuberculosis and leprosy, pneumococcus, aerobic leather Collins-negative bacilli, mycoplasma, staphylococcal infections, streptococcal infections, salmonella and chlamydia, Helicobacter pylori, malaria,
- the nascent precipitated manganese, colloidal manganese and/or the source and/or antigen capable of forming nascent precipitated manganese and/or colloidal manganese in the present invention are effective amounts.
- the present invention provides a method for increasing the expression of type I interferon in a subject, which comprises administering to the subject nascent precipitated manganese, colloidal manganese and/or capable of forming nascent precipitated manganese and/or colloidal manganese source.
- the present invention also provides a method for enhancing the activity of type I interferon in cells in vitro, which comprises applying newly precipitated manganese, colloidal manganese and/or a source capable of forming newly precipitated manganese and/or colloidal manganese to the cells, Preferably, the method is for non-therapeutic purposes.
- the present invention provides a method for inducing the cleavage of inflammatory factors to produce an active form in a subject, which comprises administering to the subject nascent precipitated manganese, colloidal manganese and/or capable of forming nascent precipitated manganese and/or colloidal manganese The source.
- the present invention also provides a method for inducing cleavage of inflammatory factors in cells to produce active forms in vitro, which comprises applying to the cells a source of newly precipitated manganese, colloidal manganese and/or capable of forming newly precipitated manganese and/or colloidal manganese.
- the method is for non-therapeutic purposes.
- the present invention provides a method for stimulating antibody production in a subject, which comprises administering to the subject nascent precipitated manganese, colloidal manganese, and/or a source capable of forming nascent precipitated manganese and/or colloidal manganese.
- the present invention also provides a method for stimulating cells to produce antibodies in vitro, which comprises applying newly precipitated manganese, colloidal manganese and/or a source capable of forming newly precipitated manganese and/or colloidal manganese to the cells, preferably, The method described is for non-therapeutic purposes.
- Figure 1 illustrates that newly precipitated manganese and colloidal manganese (Mn 2 OHPO 4 ) have good adjuvant effects.
- Figure 1A shows the different states of divalent manganese in the solution;
- Figure 1B shows the transmission electron micrograph of colloidal manganese (Mn2OHPO4);
- Figure 1C shows the level of OVA antibody IgG1 after immunization;
- Figure 1D shows the MnCl 2 solution and colloidal manganese (Mn 2 OHPO) 4 ) Comparison of residence time after intramuscular injection.
- Figure 2 illustrates the different existence states and properties of MnCl 2 solution mixed with other anions in physiological saline. And determine the ability of these precipitates or colloids to activate type I interferon or inflammatory factors, and the adjuvant effect.
- Figure 2A shows that MnCl 2 solution and different concentrations of carbonate ion, bicarbonate ion, phosphate ion, monohydrogen phosphate ion, and hydroxide ion form different states;
- Figure 2B shows that different states of manganese ion activate Type I interference It can induce the production of IL-1 ⁇ , an inflammatory factor, and the ability to produce antibodies as an adjuvant mixed with OVA protein.
- Figure 2C shows the products formed by laser pointer irradiation of manganese ions and different anions, and observe the Tyndall effect.
- Figure 3 illustrates the colloidal manganese (Mn 2 OHPO 4 ) and aluminum adjuvant mixed with chicken ovalbumin (OVA) to immunize mice by intramuscular injection for 10 days (A), 17 days (B), 24 days (C) and After 31 days (D), the amount of anti-OVA antibody produced.
- Mn 2 OHPO 4 colloidal manganese
- OVA chicken ovalbumin
- Figure 4 illustrates the colloidal manganese (Mn 2 OHPO 4 ) and cholera toxin B (CTB) adjuvants respectively mixed with OVA after nasal drip to immunize mice with alveolar lavage (A), oral lavage (B), and serum IgA (C) and serum IgG1 (D) anti-OVA antibody amount.
- Mn 2 OHPO 4 colloidal manganese
- C serum IgA
- D serum IgG1
- Figure 5 illustrates the use of colloidal manganese (Mn 2 OHPO 4 ) as an adjuvant and mixed OVA intramuscular injection (A) or intranasal immunization (B) of mice according to the method of Figure 1 and Figure 2 and the determination of mice for 1 to 6 months The amount of anti-OVA antibodies in serum, oral lavage fluid, and alveolar lavage fluid.
- Mn 2 OHPO 4 colloidal manganese
- Figure 6 illustrates MnCl 2 activation of type I interferon pathway and inflammasome activation.
- Figure 6A-B shows the expression levels of type I interferon, downstream inducible factors ISG54 and viperin after MnCl 2 treatment of mouse peritoneal macrophages, with aluminum adjuvant as a control.
- Figure 6C-D shows that MnCl 2 can activate inflammasome activation, and aluminum adjuvant is used as a control.
- Figure 6E-F shows that the activation of inflammasomes activated by MnCl 2 is dependent on NLRP3 and ASC.
- Figure 7 illustrates that colloidal manganese (Mn 2 OHPO 4 ) promotes the proliferation of CD4 + T cells, with aluminum adjuvant as a control.
- Figure 8 illustrates that the amount of antibodies produced by mice during intramuscular injection of colloidal manganese (Mn 2 OHPO 4 ) and mucosal immunization depends on the cGAS-STING pathway and the inflammasome pathway.
- Figure 9 illustrates that the ability of colloidal manganese (Mn 2 OHPO 4 ) to promote the maturation of dendritic cells and the activation of T cells depends on the cGAS-STING pathway and the inflammasome pathway.
- Figure 9A shows that colloidal manganese (MnOHPO 4 ) induces the maturation of antigen-presenting cells BMDC.
- Figure 9B shows that colloidal manganese (MnOHPO 4 ) plus antigen OVA stimulates the proliferation of CD4 + T cells.
- Figure 9C shows that colloidal manganese (MnOHPO 4 ) was used as an adjuvant to stimulate CD8 + T cell activation after immunizing mice.
- FIG 10 illustrates that colloidal manganese (Mn 2 OHPO 4 ) as an adjuvant can significantly enhance the immune protection effect of inactivated vesicular stomatitis virus (VSV) and herpes simplex virus (HSV-1).
- Figure 10A shows the experimental flow.
- Figure 10B shows that the inactivated VSV virus was diluted into doses of 10 -1 , 10 -2 , 10 -3 , 10 -4 , 10 -5 , and 10 -6 to immunize mice, and the survival rate of the mice was observed for 14 days.
- Fig. 10C shows the death curve of mice in 14 days after immunization with VSV inactivated virus diluted to a dose of 10 -3 .
- Figure 10D shows the virus titers in the brains of mice on the 4th day of infection with the VSV virus.
- Figures 10E-G show that adding colloidal manganese to the inactivated HSV-1 virus has the same protective effect.
- Figure 11 illustrates that colloidal manganese (Mn 2 OHPO 4 ) as an adjuvant can significantly enhance the immune protective effect of inactivated influenza virus PR8.
- Figure 11A-B shows the survival rate and body weight of mice after the inactivated influenza virus was diluted to 1 , 10 -1 , 10 -2 , and 10 -3 doses after intranasal immunization, 7 days later Variety.
- Figure 11C-D shows the survival rate and body weight changes of mice infected with influenza virus after immunizing mice twice with intranasal drops.
- FIG. 12 illustrates that colloidal manganese (Mn 2 OHPO 4 ) as an adjuvant can significantly enhance the immunoprotective effect of influenza subunit HA vaccine.
- Fig. 12A shows the content of anti-HA protein IgG1 antibody in mouse serum at 14, 21, and 28 days after intramuscular injection of mice.
- Fig. 12B shows the anti-HA protein IgA antibody content in the mouse serum at 14, 21, and 28 days after intranasal immunization of mice.
- Figure 12C shows the weight change of mice after infection with influenza virus.
- Figure 12D shows the pathological changes of mouse lung tissue on day 5 of infection.
- Figure 13 illustrates the use of colloidal manganese Mn 2 OHPO 4 as an adjuvant to enhance the protective effect of inactivated influenza virus and subunit vaccines against homeotype influenza virus (WSN) and heterologous influenza virus (H3N2).
- Figure 13A shows the weight change of WSN virus-infected mice after immunization;
- Figure 13B shows the weight change of H3N2 virus-infected mice after immunization.
- Figure 14 illustrates that colloidal manganese (Mn 2 OHPO 4 ) can significantly inhibit the growth of tumors in situ and metastases.
- Figure 14A shows the size of the subcutaneous tumor taken with the imager at different times.
- Figure 14B shows the tumor volume change after tumor inoculation.
- Figure 14C shows the survival curve of mice after tumor inoculation.
- Figure 14D shows that the tumor metastasized lung tissue was photographed 21 days after different immunization methods.
- Figure 14E shows the statistics of the number of tumor cells in Figure 14D.
- Figure 15 illustrates the effect of colloidal manganese (Mn 2 OHPO 4 ) and the chemotherapy drug cyclophosphamide (CTX) in the treatment of subcutaneous tumor model-melanoma B16-F10.
- Figure 15A shows pictures of tumors 14 days after different treatments.
- Figure 15B shows the growth curve of mouse subcutaneous tumor B16F10.
- Figure 15C shows the tumor weight of the mice in the corresponding group on day 14 in Figure 15B.
- Figure 16 illustrates the effect of colloidal manganese (Mn 2 OHPO 4 ) combined with PD1 antibody drugs on the subcutaneous tumor model-melanoma B16-F10.
- Figure 16A shows pictures of tumors after different treatments.
- Figure 16B shows the growth curve of the tumor.
- Figure 16C shows the tumor weights of the mice in the B panel.
- Figure 16D-E shows the number of CD8 + T cells infiltrated in the tumor analyzed by flow cytometry.
- Figure 17 illustrates the effect of colloidal manganese (Mn 2 OHPO 4 ) combined with PD1 antibody drugs in the treatment of subcutaneous tumor model-colon cancer tumor MC38.
- Figure 17A shows pictures of tumors after different treatments.
- Figure 17B shows the growth curve of the tumor.
- Figure 17C shows the tumor weight of the corresponding group in Figure 17B.
- Figure 17D shows a tissue section of the tumor in the corresponding group in Figure A, with immunofluorescence staining of DAPI and CD8 + T cells.
- innate immunity refers to the natural immune defense function formed by the body during germline development and evolution, that is, the non-specific defense function already possessed after birth, also known as non-specific immunity (non-specific immunity).
- Innate immunity involves a variety of cells and molecules, such as macrophages, natural killer cells, complement, cytokines (IL, CSF, IFN, TNF, TGF- ⁇ ), chemokines (including CC chemokines, such as CCL1, CCL2) , CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCL10, CCL11, CCL12, etc.
- CXC chemokines such as CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8, CXCL9, etc., C Factor, CX3C chemokine), lysozyme and so on.
- adaptive immunity refers to the specific immunity of the body against the antigen formed after the stimulation of the antigen molecule, which involves cellular immunity and humoral immunity.
- adjuvant refers to an agent that does not constitute a specific antigen, but enhances the strength and duration of the immune response to the co-administered antigen.
- divalent manganese or “divalent manganese compound” as used herein can be hydrochloride, carbonate, hydrobromide, sulfate, nitrate, phosphate, tartrate, fumarate , Maleate, lactate, benzenesulfonate, pantothenate, ascorbate, hydroxide, etc., or any combination thereof.
- the divalent manganese compound is pharmaceutically acceptable.
- colloidal manganese refers to the state in which divalent manganese exists in the form of colloid (colloid, also known as colloidal solution) formed with anions.
- colloid has the meaning understood by those of ordinary skill in the art.
- the particle size of colloidal particles is generally about 1 to 100 nm, especially 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 nm, or a value between any two of the above values, such as 1 to 20, 5 to 20 nm, 10 nm.
- colloidal solutions exhibit the Tyndall effect when exposed to light.
- the particle size range of colloidal particles is on the order of nanometers
- “colloidal manganese” in this article can also be referred to as “nano manganese” according to its particle size range.
- cent precipitated manganese refers to the state of existence of the divalent manganese compound in a short period of time after the non-precipitated form (for example, the solution form) is converted to the precipitated form.
- the short period of time generally does not exceed 1 day, and typically does not exceed 24, 22, 20, 18, 16, 15, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1. Hours, such as not exceeding 60, 50, 45, 40, 35, 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 minute.
- derived from viruses, bacteria and/or parasites means to include inactivated viruses, bacteria and/or parasites, or proteins and/or nucleic acids extracted from the above pathogenic substances (including cutting , Engineering and other processed substances), or purified recombinant protein substances, or chemically synthesized peptide substances.
- pharmaceutically acceptable carrier can be selected from: water, buffered aqueous solution, isotonic salt solution such as PBS (phosphate buffered saline), glucose, mannitol, dextrose, lactose, starch, stearin Magnesium acid, cellulose, magnesium carbonate, 0.3% glycerin, hyaluronic acid, ethanol or polyalkylene glycols such as polypropylene glycol, triglycerides, etc.
- the type of pharmaceutically acceptable carrier used depends inter alia on whether the composition according to the invention is formulated for oral, nasal, intradermal, subcutaneous, intramuscular or intravenous administration.
- composition according to the present invention may contain lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts that affect osmotic pressure, buffers, coloring substances, flavoring substances and/or aromatic substances and the like as additives.
- composition according to the present invention can be administered by any suitable route, such as oral, nasal, intradermal, subcutaneous, intramuscular or intravenous administration.
- administration means to provide a substance to a subject in a pharmacologically usable manner.
- pharmaceutical effective amount and “effective amount” refer to a dose sufficient to show its benefit to the administered subject.
- the actual amount administered, as well as the rate and time course of administration, will depend on the condition and severity of the individual being treated.
- the prescription of treatment (such as the decision on dosage, etc.) is ultimately the responsibility of the general practitioner and other doctors and rely on them to make decisions, usually taking into account the disease to be treated, the individual patient’s condition, the delivery site, the method of application, and what is already known to the doctor. Know other factors.
- subject means animals, including warm-blooded mammals such as humans and primates; birds; domesticated domestic or farm animals such as cats, dogs, sheep, goats, cows, horses, and pigs ; Laboratory animals, such as mice, rats and guinea pigs; fish; reptiles; zoo animals and wild animals.
- any component, element, attribute or step disclosed in an embodiment of the method and product can be applied to any other method and product disclosed herein.
- anti-GAPDH antibody (sc-25778) was purchased from Santa Cruz.
- Anti-Viperin antibodies, anti-ISG54 antibodies, anti-Casp1/p20 antibodies, anti-IL1 ⁇ /p17 antibodies and anti-ASC antibodies are all prepared and used by published methods (16).
- the cDNA of the antigen fragment is inserted into the pET-21b vector (Novagen) and expressed in E. coli BL21(DE3), the recombinant protein is purified by Ni-NTA affinity column, and then injected into mice or rabbits , To obtain antiserum that can recognize the corresponding antigen.
- L929-ISRE (Jiang Zhengfan’s laboratory self-made cell line, stably transfected pGL3 ISRE-Luciferase plasmid into L929 cells, CCL-1), BHK21( CCL-10), B16-OVA( CRL-6322) and B16F10( CRL-6475) cells were cultured in DMEM (Gibco) medium supplemented with 10% FBS (Gibco), 5 ⁇ g/ml penicillin and 10 ⁇ g/ml streptomycin.
- BMDCs Bone marrow derived macrophages
- RPMI-1640 (Gibco) medium containing 10ng/mL GM-CSF, 10ng/mL IL-4, and 10% FBS (Gibco)
- FBS FBS
- Peritoneal macrophages were harvested from mice 5 days after the induction of thioglycolate (BD, Sparks, MD) injection, and cultured in DMEM medium supplemented with 5% FBS.
- Tmem173 -/- mice were established by injecting Cas9 mRNA (100ng/ ⁇ l) and gRNA (50ng/ ⁇ l) cytoplasm into C57BL/6J mouse fertilized eggs using the CRISPR-cas9 method.
- Cas9 mRNA and single guide RNA (gRNA) were transcribed in vitro by mMESSAGE mMACHINE T7 Ultra (Ambion, am1345).
- Nlrp3 -/- , Nlrc4 -/- , Pycard -/- and Aim2 -/- gene knockout mice were donated by Vishva Dixit (Genentech Inc, USA).
- Tmem173 -/- Pycard -/- double knockout mice were identified by mating Tmem173 -/- mice and Pycard -/- .
- mice All mice were fed in the Laboratory Animal Center of Peking University under aseptic conditions in accordance with the NIH National Institute of Health Guide for Care and Use of Laboratory Animals.
- Type I IFN Type I-IFN bioassay
- IFN-stimulated response element IFN-stimulated response element
- pGL3-Basic vector Promega
- IFN-sensitive luciferase vector IFN-sensitive luciferase vector
- L929-ISRE cells were seeded into a 96-well plate and incubated with the cell culture supernatant. Recombinant human and mouse IFN- ⁇ (R&D Systems) were used as standards. After 4 hours, L929-ISRE cells were lysed and assayed by Luciferase Reporter Assay System (Promega).
- H1N1 influenza virus PR8 strain from Yonghui Zhang, Tsinghua University, VR-95
- H1N1 influenza virus WSN strain, A/WSN/33(H1N1) from Wenjun Liu, Institute of Microbiology, CAS
- H3N2 influenza virus strain, H3N2 subtype A/Jiangxi/2005 from Min Fang, Institute of Microbiology, CAS
- BHK21 cells were used to determine the titers of HSV-1 and VSV viruses.
- MDCK cells were used to determine influenza virus titer.
- HSV-1 and VSV viruses are amplified with vero cells.
- PR8, WSN, and H3N2 influenza viruses are amplified with chicken embryos.
- the amplified virus was purified with PEG8000.
- the purified virus was inactivated with 0.2% formaldehyde at 37 degrees Celsius for 24 hours. The plaque method was used to measure whether the virus was completely inactivated, and then the inactivated virus was used to immunize mice.
- Mouse survival experiment Infect 8-12 weeks of mice intravenously with HSV-1 (1.4 x 10 7 pfu/mouse) and VSV (8 x 10 8 pfu/mouse), or with PR8 (1 x 10 5 pfu/mouse), WSN (1 x 10 6 pfu/mouse), H3N2 (1 x 10 6 pfu/mouse) intranasally infected mice for 8-12 weeks.
- Plaque assay BHK21 cells were incubated with homogenate from infected mouse organs (a series of dilutions in serum-free DMEM) for 2 hours. Then, the medium was replaced with serum-free DMEM containing 0.5% methylcellulose. After 60 hours, the cells were fixed with 0.5% (vol/vol) glutaraldehyde and stained with 1% (wt/vol) crystal violet (dissolved in 70% ethanol). The plaques are counted to calculate the virus titer in plaque forming units.
- the purified plasmid of influenza virus HA1 protein was donated by Zhang Yonghui's laboratory of Tsinghua University (18).
- the plasmid expresses the 11-324 amino acids of HA protein (Gene ID: 956529).
- the bacterial solution is shaken to an OD 600 of 0.6, it is induced by adding 0.5 mM IPTG and incubated at 37°C for 5 hours.
- the inclusion bodies are collected by ultrasound, and the HA1 protein is obtained after denaturation and renaturation. Then the HA1 protein was purified by gel filtration chromatography.
- Ovalbumin (OVA) as an immunogen to immunize mice
- the method of immunization by intramuscular injection of MnCl 2 solution is to add 10 ⁇ g MnCl 2 and 10 ⁇ g OVA protein to 100 ⁇ L PBS, mix and leave for 5 minutes, and then immunize mice by intramuscular injection.
- Intranasal immunization method of MnCl 2 solution is to add 5 ⁇ g MnCl 2 and 10 ⁇ g OVA protein to 20 ⁇ L PBS, mix and place for 5 minutes, and then immunize mice by intranasal drip.
- Colloidal manganese (take Mn 2 OHPO 4 as an example) intramuscular injection immunization method is to add 10 ⁇ g colloidal manganese and 10 ⁇ g OVA protein to 100 ⁇ L of normal saline, mix well and intramuscularly immunize mice.
- Colloidal manganese nasal drip immunization method is to add 5 ⁇ g colloidal manganese and 10 ⁇ g OVA protein to 20 ⁇ L of normal saline, mix well and immunize mice by nasal drip.
- Aluminum adjuvant intramuscular injection immunization method is to add 10 ⁇ g/20 ⁇ L aluminum adjuvant and 10 ⁇ g OVA protein to 80 ⁇ L normal saline, emulsify with an emulsifier, and then intramuscularly immunize mice.
- Cholera toxin B (CTB) intranasal immunization method is to add 5 ⁇ g CTB and 10 ⁇ g OVA protein to 20 ⁇ L of normal saline, mix well and immunize mice by intrana
- the ELISA method measures OVA-specific IgG1, IgG2c, total IgG, and IgA antibodies: the diluted serum from the immunized mice is incubated on an ELISA plate coated with 100 ⁇ g/ml OVA. After washing, use HRP-conjugated antibody mouse IgG1 (eBioscience, #18-4015-82), IgG2c (GeneTex, GTX77297), IgG total (Invitrogen, G21040), IgA (GeneTex, GTX77223) to detect the bound corresponding antibodies . Then, the plate was incubated with the substrate TMB (eBioscience), the reaction was stopped with 1M H 3 PO 4 , and then the absorbance at 450 nm was measured.
- HRP-conjugated antibody mouse IgG1 eBioscience, #18-4015-82
- IgG2c GeneTex, GTX77297
- IgG total Invitrogen, G21040
- IgA GeneTex,
- Mantel-Cox test was used to compare survival curves.
- Example 1 The state of manganese compound in solution and its effect as an adjuvant
- mice were randomly divided into 6 groups according to their body weight, namely: 1) PBS control group, 2) antigen OVA (10 ⁇ g), 3) MnCl 2 reacted in saline for 2 days (10 ⁇ g MnCl 2 +OVA), 4) MnCl 2 Reaction in PBS for 5 minutes group (10 ⁇ g MnCl 2 +OVA), 5) MnCl 2 reaction in PBS for 2 days group (10 ⁇ g MnCl 2 +OVA), 6) MnCl 2 reaction to generate colloidal manganese for 30 days group (10 ⁇ g Mn 2 OHPO 4 +OVA). 10 ⁇ g here refers to the mass of manganese.
- mice were randomly divided into 2 groups according to their body weight, namely: intramuscular injection of MnCl 2 solution group (20 ⁇ g MnCl 2 dissolved in 100 ⁇ l of saline), intramuscular injection of colloidal manganese (20 ⁇ g of colloidal manganese suspended in 100 ⁇ l of saline), where 20 ⁇ g were both Refers to the quality of manganese.
- MnCl 2 solution group (20 ⁇ g MnCl 2 dissolved in 100 ⁇ l of saline
- colloidal manganese 20 ⁇ g of colloidal manganese suspended in 100 ⁇ l of saline
- the content of manganese was determined by ICP-MS (Thermo X SERIES II).
- colloidal manganese can stay at the injection site longer, which can activate the body to produce antibodies for a longer time, while the solution manganese is quickly metabolized.
- Mn 3 (PO 4 ) 2 is produced at low concentration, and Mn 2 OHPO 4 is produced at high concentration; divalent free manganese reacts with HPO 4 2- to produce white MnHPO 4 ; reacts with OH - to produce slightly yellow Precipitated Mn(OH) 2 .
- P-IRF3 is the main transcription factor regulating the expression of type I interferon.
- Mn 2 OHPO 4 and MnHPO 4 can also significantly induce the production of IL-1 ⁇ .
- non-colloid Mn(OH) 2 can only activate the production of type I interferon, but cannot induce the production of inflammatory factor IL-1 ⁇ .
- the mouse serum was taken to measure the anti-OVA IgG1 antibody content. The effect of Mn 2 OHPO 4 and MnHPO 4 in stimulating the body to produce antibodies is significantly better than that of manganese compounds in the precipitated state.
- the leftmost MnCl 2 in the antibody test is the adjuvant effect of the precipitation of MnCl 2 reacted with PBS for 5 minutes. Other solid precipitates placed overnight also have a partial adjuvant effect.
- the reaction product was resuspended in physiological saline and then irradiated with a laser pointer. It can be seen that the Tyndall effect of Mn 2 OHPO 4 is the most obvious, and MnHPO 4 has a part of Tyndall effect, probably because its diameter is slightly larger than 100 nm. The diameter of Mn 2 OHPO 4 is about 10 nm, which meets the definition of colloid.
- Example 3 Colloidal manganese Mn 2 OHPO 4 as an adjuvant for intramuscular injection and mucosal immunity
- mice were randomly divided into 6 groups according to their body weight, namely: PBS control group, antigen OVA (10 ⁇ g), colloidal manganese Mn 2 OHPO 4 group (10 ⁇ g Mn 2 OHPO 4 +OVA), low-dose aluminum adjuvant group (440 ⁇ g aluminum adjuvant)
- the medium-dose aluminum adjuvant group 800 ⁇ g aluminum adjuvant+OVA
- the high-dose aluminum adjuvant group (1220 ⁇ g aluminum adjuvant+OVA).
- Each group was immunized by intramuscular injection according to the experimental design amount, and immunized once on the 0th day and the 10th day.
- colloidal manganese Mn 2 OHPO 4 at a dose of 10 ⁇ g/mouse is more effective than a high-dose (1220 ⁇ g/mouse) aluminum adjuvant. This indicates that colloidal manganese can achieve a better immune enhancement effect at a dose 100 times lower than that of aluminum adjuvants, which can greatly reduce the side effects of aluminum adjuvants currently widely used.
- Example 4 Colloidal manganese Mn 2 OHPO 4 can be used as an adjuvant for intramuscular injection and mucosal immunity to make the body persistently produce specific antibodies
- mice were immunized with colloidal manganese Mn 2 OHPO 4 as an adjuvant mixed with OVA by intramuscular injection, and the anti-OVA antibody content in the serum of the mice was determined 1 week, 1 month, 3 months, and 6 months after immunization. .
- the anti-OVA antibody in the serum remained at a high level by the sixth month, which indicates that the colloidal manganese Mn 2 OHPO 4 as an adjuvant can produce a lasting protective effect.
- the life span of mice is generally 2 years. It is expected that colloidal manganese Mn 2 OHPO 4 can produce protective antibodies throughout the life cycle of the body.
- Example 3 Refer to the method in Example 3 after mucosal immunization of mice, the blood, alveolar lavage fluid, and oral lavage fluid of the mice were collected at different times to determine the antibody content. As shown in Figure 5B, the concentration of specific antibodies remained at a high level until 6 months after immunization, indicating that colloidal manganese Mn 2 OHPO 4 can also produce a lasting protective effect when used for mucosal immunity.
- Example 5 Manganese (II) activates the production of inflammatory factors
- the inflammatory factor IL-1 ⁇ can directly act on CD4 + T cells and promote the proliferation of T cells (8).
- IL-1 ⁇ and IL-1 ⁇ can promote the infiltration of neutrophils (19); the inflammatory factor IL-18 can be effective Enhancing Th1 immune response can also promote the proliferation and cytotoxicity of T cells and NK cells (9). It has been reported that the adjuvant effect of aluminum adjuvant is achieved by activating NLRP3 inflammasome and releasing inflammatory factors (20-22), so we tested and compared MnCl 2 /colloidal manganese Mn 2 OHPO 4 with aluminum adjuvant to activate inflammatory factors. Ability.
- MnCl 2 and colloidal manganese Mn 2 OHPO 4 have the same ability to activate inflammatory factors (see Figure 2B) and mechanism.
- MnCl 2 is used as an example.
- the primary culture of mouse peritoneal macrophages cultured in vitro were treated with 10, 20, 50, 100 ⁇ g/mL manganese chloride or aluminum adjuvants, and the interferon-inducing genes ISG54 and Viperin were detected by Western blotting, and the supernatant was detected by Bioassay The total type I interferon content (the sum of the concentrations of interferon alpha and interferon beta). The results are shown in Figures 6A and 6B.
- the peritoneal macrophages pretreated with LPS were treated with MnCl 2 , ATP (classical NLRP3 inflammasome activator), and VacV (classical Aim2 inflammasome activator).
- Western blotting was used to detect the cleavage of caspase1 and IL-1 ⁇ in the supernatant Cut (the upper band is the precursor, the lower band is the active form of caspase1 and IL-1 ⁇ ), and the production of inflammatory factors IL-1 ⁇ and IL-18 in the supernatant is detected by ELISA.
- the results are shown in Figures 6E and 6F.
- NLRP3 knockout cells the ability of MnCl 2 to activate inflammatory factors was significantly weakened, so NLRP3 inflammasome was mainly activated by MnCl 2 .
- OT-II cells labeled with CFSE dye into the tail vein of mice.
- the usage amount described here is the amount of one mouse immunization. Lymph nodes were collected three days after the immunized mice, and the proliferation of OT-II cells in the lymph nodes was analyzed by flow cytometry.
- the rightmost peak in the figure is the OT-II cells originally injected into the mouse body.
- the CFSE in the cell will be evenly distributed between the two cells, so the brightness of the CFSE in the cell will decrease, and the cells with decreased CFSE on the left are all dividing cells.
- the results show that when the same amount of antigen is used, the ability of colloidal manganese to promote the proliferation of T cells is stronger than that of aluminum adjuvant, and the amount of colloidal manganese only needs 1/100 of that of aluminum adjuvant to achieve similar effects.
- Example 7 Colloidal manganese Mn 2 OHPO 4 activated antibody production depends on the STING pathway and the inflammasome pathway
- Figure 8A shows the intramuscular injection of WT, Pycard knockout (ASC protein deletion), Sting knockout (STING protein deletion), Pycard and Sting double knockout with 10 ⁇ g of colloidal manganese per mouse plus 10 ⁇ g of OVA protein.
- the mouse serum was taken to measure the IgG1, IgG2c, and total IgG contents of anti-OVA antibodies in the serum by ELISA.
- the results show that knockout alone will only partially lose the adjuvant effect of colloidal manganese, while double knockout will significantly reduce the adjuvant effect.
- Fig. 8B shows that mice were immunized three times with 5 ⁇ g colloidal manganese per mouse and 10 ⁇ g OVA protein mucosa.
- the serum, alveolar lavage fluid, and oral lavage fluid were collected by ELISA to measure the secreted anti-OVA IgA antibody content. The results showed that the adjuvant effect was significantly reduced in double knocked mice.
- Example 8 Colloidal manganese Mn 2 OHPO 4 promotes the maturation of dendritic cells
- Figure 9A shows the use of 100, 200, and 400 ⁇ M colloidal manganese to treat bone marrow-derived dendritic cells (BMDC) cells for 20 hours, and use a flow cytometer to detect the CD86 protein, a marker of dendritic cell maturation.
- BMDC bone marrow-derived dendritic cells
- FIG 9B the method described in Example 6 was used in four mice to test the efficiency of colloidal manganese in promoting T cell proliferation.
- Example 9 Colloidal manganese Mn 2 OHPO 4 as an adjuvant enhances the immune protective effect of inactivated vesicular stomatitis virus (VSV) and herpes simplex virus (HSV-1)
- the experimental procedure is shown in Figure 10A.
- the mice were injected intramuscularly with the vaccine on day 0, and the mice were infected with virus on day 10.
- the virus titer in mouse tissues was measured on day 14.
- the mice were observed and the death curve was recorded to day 14. .
- the PEG-purified VSV virus was inactivated and used as a vaccine, and diluted to 10 -1 , 10 -2 , 10 -3 , 10 -4 , 10 -5 and 10 -6 dilutions to immunize mice by intramuscular injection.
- the virus concentration is high enough, it can produce enough immune effect to protect mice when the virus is used alone. After the virus is diluted by 10 times several gradients, the mice are immunized.
- the survival rate of mice is only 8.3%.
- the concentration was 10 -4 , 10 -5 , and 10 -6 , all mice died; but when Mn 2 OHPO 4 colloid was added as an adjuvant to immunize mice with 10 -3 concentration of inactivated virus, the survival rate of mice Increase to 75%, as shown in Figures 10B and 10C. It shows that colloidal manganese as an adjuvant can enhance the immune effect of low-concentration immunogens, using a small amount of immunogen can play a protective effect, and the active virus titer in mice is also reduced, as shown in Figure 10D.
- the PEG-purified HSV-1 virus was inactivated and used as a vaccine to immunize mice by intramuscular injection.
- the virus dilution is 10 -1 , 10 -2 , it can produce sufficient immune effect to protect mice when the virus is used alone.
- the survival rate of mice was only 16.7%, and all the mice died when the dilution was lower; while adding Mn 2 OHPO 4 colloid to the virus at the 10 -4 dilution, the survival rate of mice increased to 100%, as shown in Figures 10E and 10F.
- the active virus titer in mice was reduced, as shown in Figure 10G. This indicates that colloidal manganese as an adjuvant can achieve the same protective effect while reducing the dose of inactivated HSV-1 virus by 10 times.
- the PEG-purified influenza PR8 virus was inactivated and diluted to 1 , 10 -1 , 10 -2 , and 10 -3 dilutions, and the mice were immunized once by nasal drip. Seven days later, the mice were infected with influenza virus, and the survival rate and weight change of the mice were recorded. All doses have no protective effect when no adjuvant is added, and the weight of the mouse has been reduced until death; and after adding Mn 2 OHPO 4 colloid as an adjuvant, the virus diluted to a dose of 10 -1 can make the weight of the mouse drop to The increase appeared after the 4th and 5th days, the body weight returned to the level before the virus infection after the 10th day, and the condition improved, as shown in Figures 11A and 11B. This result indicates that colloidal manganese can be used as an adjuvant to protect an immunogen that is originally non-immunogenic or weakly immunogenic after immunizing a subject once.
- the PEG-purified influenza PR8 virus was inactivated and diluted to dilutions of 1 , 10 -1 , 10 -2 , and 10 -3 .
- the mice were immunized twice (day 0 and day 7). immunity). After 14 days, the mice were infected with influenza virus, and the survival rate and weight change of the mice were recorded. All doses have no protective effect without adjuvant, and the weight of the mice has been reduced until death; and after adding Mn 2 OHPO 4 colloid as an adjuvant, the mice were immunized twice with the virus diluted to a dose of 10 -1 .
- Example 10 Colloidal manganese Mn 2 OHPO 4 is used as an adjuvant to enhance the protective effect of influenza subunit vaccine
- mice were randomly divided into 4 groups according to their body weight, namely: blank control group (ctrl), HA antigen group (HA), HA antigen and colloidal manganese group (HA+Mn) and HA antigen and aluminum adjuvant group (HA+Al ).
- ctrl blank control group
- HA HA antigen group
- H+Mn HA antigen and colloidal manganese group
- HA+Al HA antigen and aluminum adjuvant group
- Example 11 Colloidal manganese Mn 2 OHPO 4 is used as an adjuvant to enhance inactivated influenza virus and subunit vaccines to have protective effects on homeotype influenza virus (WSN) and heterologous influenza virus (H3N2) bacteria
- mice were randomly divided into 5 groups according to their body weight, namely: blank control group (con), inactivated virus PR8 group, inactivated virus PR8+ colloidal manganese group (PR8+MnJ), PR8-HA1 protein group (HA1) and PR8- HA1 protein + colloidal manganese group (HA1+MnJ).
- blank control group con
- inactivated virus PR8 group inactivated virus PR8+ colloidal manganese group
- PR8+MnJ PR8-HA1 protein group
- HA1+MnJ PR8-HA1 protein + colloidal manganese group
- mice were randomly divided into 5 groups according to their body weight, namely: blank control group (con), inactivated virus PR8 group, inactivated virus PR8+ colloidal manganese group (PR8+MnJ), PR8-HA1 protein group (HA1) and PR8- HA1 protein + colloidal manganese group (HA1+MnJ).
- Blank control group con
- inactivated virus PR8 group inactivated virus PR8+ colloidal manganese group
- PR8+MnJ PR8-HA1 protein group
- HA1+MnJ PR8-HA1 protein + colloidal manganese group
- the weight of the mice will drop to 80% before recovery, and the HA1 group alone will reduce to 90% recovery.
- the HA1 group with colloidal manganese basically did not lose weight, and the inactivated virus group with colloidal manganese added only 5% of the weight to recover.
- Example 12 Colloidal manganese Mn 2 OHPO 4 as an immune enhancer enhances the body's anti-tumor effect
- mice were randomly divided into 4 groups according to their body weight, namely: blank control group (con), OVA control group, OVA and colloidal manganese group (OVA+Mn), and OVA and aluminum adjuvant group (OVA+Al). After immunizing the mice three times according to the experimental design (once every 7 days), the melanoma B16-OVA was subcutaneously inoculated to establish an orthotopic tumor model.
- the use of aluminum adjuvants in advance cannot inhibit tumor growth and improve the survival rate of mice; while the use of colloidal manganese in advance can significantly inhibit tumor growth and increase the survival rate of mice; 21 days after tumor inoculation, the control group is small the average tumor size of mice is 1906mm 3, the average tumor size of mice was OVA 925.5mm 3, manganese colloid tumors in mice immunized with an average size of 222.9mm 3, the average tumor size of mice aluminum adjuvant is 1946mm 3, As shown in Figure 14B; the survival time of control mice was 26 days, and the survival time of mice immunized with colloidal manganese was 49 days, which was significantly longer than that of mice immunized with Lu adjuvant at 27 days, as shown in Figure 14C. The results indicate that colloidal manganese can be used as an immune enhancer to enhance the body's epitopic tumor effect.
- Mn 2 OHPO 4 colloid and aluminum adjuvant were mixed with antigen OVA to immunize mice, immunized every 7 days, immunized 3 times to activate the immune response in the body; B16-OVA tumor was injected into the tail vein to establish a tumor metastasis model. The mice were sacrificed 21 days after the tumor injection, and the lung tissues were taken out. It can be seen that the lungs of the mice in the blank control group were covered with melanomas, and a certain number of melanomas appeared on the lungs of the mice immunized with OVA, while those immunized with OVA + colloidal manganese There were almost no tumors on the lungs of the mice, as shown in Figure 14D.
- mice with melanoma B16F10 After subcutaneously inoculating mice with melanoma B16F10, they were randomly divided into 4 groups: control group (con), Mn 2 OHPO 4 colloid group (5 mg/kg), Cyclophosphamide Monohydrate (CTX) group (140 mg/kg), and Combination medication group (CTX+Mn). Cyclophosphamide (CTX) is a widely used anti-tumor chemotherapy drug in clinical practice. Mn 2 OHPO 4 colloidal intramuscular injection (treatment every two days after tumor inoculation), intraperitoneal injection of CTX (treatment each on the 6, 9 and 12 days after tumor inoculation), and the control group was given an equal volume of normal saline.
- CTX Cyclophosphamide
- the combination of colloidal manganese and CTX can significantly inhibit tumor growth. 14 days after tumor inoculation, the mice were sacrificed, the tumors were dissected and their volumes were measured. The average tumor size of the control group was 960.1mm 3 , the tumor size of mice treated with colloidal manganese alone was 659.5mm 3 , and the size of mice treated with CTX alone was 393.9 mm 3 , the tumor size of the combined mice was 238 mm 3 , as shown in Figure 15B; weighing the weight of each tumor at the same time, the results showed that the combined use of colloidal manganese and CTX inhibited tumor growth, as shown in Figure 15C. This indicates that colloidal manganese as an immune enhancer enhances traditional treatment methods, such as chemotherapy, and its combined use has a significant inhibitory effect on tumor growth.
- mice with melanoma B16F10 After subcutaneously inoculating mice with melanoma B16F10, they were randomly divided into 4 groups: control group (con), Mn 2 OHPO 4 colloid group (5 mg/kg), anti-PD1 antibody group (200 ⁇ g/mouse), and combined Medication group (anti-PD1+Mn).
- PD1 antibody is a new drug used in immunotherapy in recent years.
- PD1 antibody (Clone 29 F.1A12, BioXCell) was treated once each on the 3rd, 7th, and 11th days after tumor inoculation. Each time 200ug of antibody was dissolved in 200ul PBS and injected intraperitoneally.
- mice 15 days after tumor inoculation, the mice were sacrificed, and the tumors were dissected and measured.
- the average tumor size of mice in the control group was 1847 mm 3
- the average tumor size of mice in the colloidal manganese group alone was 797.3 mm 3 .
- tumor bearing mice PD1 antibody alone group average size of 687.3mm 3, while tumors in mice treated with a mean size of 342mm 3, PD1 antibodies significantly lower than used alone.
- Flow cytometry was used to analyze the proportion of CD8 + T cells infiltrating the tumor.
- Colloidal manganese increased the effect of PD1 antibody drug from 27.7% to 45.4%. This indicates that colloidal manganese can cooperate with PD1 antibody drugs to treat melanoma.
- mice were subcutaneously inoculated with colon cancer tumor MC38 and randomly divided into 4 groups, namely: control antibody group (Isotype, 200 ⁇ g/mouse), Mn 2 OHPO 4 colloid and control antibody group (Isotype+Mn), anti-PD1 antibody In the group (200 ⁇ g/mouse), and the combination group (anti-PD1+Mn), the use of PD-1 is the same as in experiment (d). Twenty days after tumor inoculation, the mice were sacrificed and the tumors were dissected and measured.
- mice in the control group were 319.8 mm 3
- the size of mice treated with manganese alone was 126.9 mm 3
- the size of mice treated with PD1 antibody drug alone was 143.1 mm 3
- the tumor size of the mice treated with the combination treatment was 66.74 mm 3 , as shown in Figure 17B.
- the tumor tissue sections were made, and immunofluorescence staining of DAPI and CD8 + T cells was performed. It can be seen that the proportion of CD8 + T cells infiltrating into the tumor of mice treated with PD1 antibody and colloidal manganese was significantly increased, as shown in Figure 17D. This indicates that colloidal manganese can cooperate with PD1 antibody drugs to treat colon cancer tumors.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Public Health (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Virology (AREA)
- Engineering & Computer Science (AREA)
- Oncology (AREA)
- Immunology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Communicable Diseases (AREA)
- Tropical Medicine & Parasitology (AREA)
- Molecular Biology (AREA)
- Diabetes (AREA)
- Epidemiology (AREA)
- Hematology (AREA)
- Rheumatology (AREA)
- Inorganic Chemistry (AREA)
- Physical Education & Sports Medicine (AREA)
- Biotechnology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Endocrinology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Dermatology (AREA)
- Obesity (AREA)
- Emergency Medicine (AREA)
- Pain & Pain Management (AREA)
- AIDS & HIV (AREA)
- Pulmonology (AREA)
- Gastroenterology & Hepatology (AREA)
- Neurosurgery (AREA)
Abstract
Description
Claims (41)
- 一种免疫增强组合物,其包含新生沉淀锰、胶体锰和/或能够形成新生沉淀锰和/或胶体锰的源。
- 根据权利要求1所述的免疫增强组合物,其用作免疫佐剂。
- 根据权利要求1或2所述的免疫增强组合物,其中所述新生沉淀锰和/或胶体锰选自磷酸锰盐、碳酸锰盐、氢氧化锰及其任意混合物。
- 根据权利要求1至3任一项所述的免疫增强组合物,其中所述新生沉淀锰从非沉淀形式转化成沉淀形式的时间不超过1天,或者不超过24、22、20、18、16、15、14、12、10、9、8、7、6、5、4、3、2或1小时,或者不超过60、50、45、40、35、30、25、20、15、10、9、8、7、6、5、4、3、2或1分钟。
- 一种疫苗组合物,其包含A)疫苗免疫原,和B)新生沉淀锰、胶体锰和/或能够形成新生沉淀锰和/或胶体锰的源,任选地,组分A和B可以处于相同和/或分开的容器中。
- 根据权利要求5的疫苗组合物,其中所述疫苗免疫原源自病毒、细菌和/或寄生虫,例如所述病毒选自:DNA病毒和RNA病毒,优选地所述病毒选自:疱疹病毒科、弹状病毒科、丝状病毒科、正粘病毒科、副粘病毒科、冠状病毒科、小RNA病毒科、嗜肝DNA病毒科、黄病毒科、乳头瘤病毒科、痘病毒科、和逆转录病毒科,更优选地所述病毒选自:单纯疱疹病毒、水疱性口炎病毒、牛痘病毒、HIV和HBV;例如所述细菌选自革兰氏阴性菌和革兰氏阳性菌,优选地所述细菌选自肺炎链球菌(Streptococcus pneumoniae)、流感嗜血杆菌(Haemophilus influenzae)、沙门氏菌(Salmonella)、脑膜炎双球菌(Meningococcus)、表皮葡萄球菌(Staphylococcus epidermidis)、金黄色葡萄球菌(Staphylococcus aureus)、大肠杆菌(Escherichia coli)、肺炎克雷伯氏菌(Klebsiella pneumoniae)、产酸克雷伯氏菌(Klebsiella oxytoca)、阴沟肠杆菌(Enterobacter cloacae)、弗氏柠檬酸杆菌(Citrobacter freundii)、绿脓假单胞菌(Pseudomonas aeruginosa)和 波美不动杆菌(Acinetobacter baumanni);具体地,例如所述疫苗免疫原源自流感病毒、肝炎病毒(如甲肝病毒、乙肝病毒)、脊髓灰质炎病毒、狂犬病毒、HPV病毒、脑炎病毒(如乙型脑炎病毒)、腮腺炎病毒、风疹病毒、破伤风杆菌、百日咳杆菌、白喉杆菌、麻风杆菌、结核杆菌、脑膜炎双球菌、肺炎双球菌及其任意组合。
- 根据权利要求5或6的疫苗组合物,其中所述新生沉淀锰和/或胶体锰选自磷酸锰盐、碳酸锰盐、氢氧化锰及其任意混合物。
- 根据权利要求5至7任一项所述的疫苗组合物,其中所述新生沉淀锰从非沉淀形式转化成沉淀形式的时间不超过1天,或者不超过24、22、20、18、16、15、14、12、10、9、8、7、6、5、4、3、2或1小时,或者不超过60、50、45、40、35、30、25、20、15、10、9、8、7、6、5、4、3、2或1分钟。
- 一种制造免疫增强组合物的方法,其包括1)提供能够形成新生沉淀锰和/或胶体锰的源;和2)任选地使所述能够形成新生沉淀锰和/或胶体锰的源转化为新生沉淀锰和/或胶体锰。
- 根据权利要求9所述的方法,其中所述新生沉淀锰和/或胶体锰选自磷酸锰盐、碳酸锰盐、氢氧化锰及其任意混合物。
- 根据权利要求9或10所述的方法,其中所述新生沉淀锰从非沉淀形式转化成沉淀形式的时间不超过1天,或者不超过24、22、20、18、16、15、14、12、10、9、8、7、6、5、4、3、2或1小时,或者不超过60、50、45、40、35、30、25、20、15、10、9、8、7、6、5、4、3、2或1分钟。
- 一种制造疫苗组合物的方法,其包括1)提供疫苗免疫原;和2)提供新生沉淀锰、胶体锰和/或能够形成新生沉淀锰和/或胶体锰的源。
- 根据权利要求12的方法,其中所述疫苗免疫原源自病毒、细菌和/或寄生虫,例如所述病毒选自:DNA病毒和RNA病毒,优选地所述病毒选自:疱疹 病毒科、弹状病毒科、丝状病毒科、正粘病毒科、副粘病毒科、冠状病毒科、小RNA病毒科、嗜肝DNA病毒科、黄病毒科、乳头瘤病毒科、痘病毒科、和逆转录病毒科,更优选地所述病毒选自:单纯疱疹病毒、水疱性口炎病毒、牛痘病毒、HIV和HBV;例如所述细菌选自革兰氏阴性菌和革兰氏阳性菌,优选地所述细菌选自肺炎链球菌(Streptococcus pneumoniae)、流感嗜血杆菌(Haemophilus influenzae)、沙门氏菌(Salmonella)、脑膜炎双球菌(Meningococcus)、表皮葡萄球菌(Staphylococcus epidermidis)、金黄色葡萄球菌(Staphylococcus aureus)、大肠杆菌(Escherichia coli)、肺炎克雷伯氏菌(Klebsiella pneumoniae)、产酸克雷伯氏菌(Klebsiella oxytoca)、阴沟肠杆菌(Enterobacter cloacae)、弗氏柠檬酸杆菌(Citrobacter freundii)、绿脓假单胞菌(Pseudomonas aeruginosa)和波美不动杆菌(Acinetobacter baumanni);具体地,例如所述疫苗免疫原源自流感病毒、肝炎病毒(如甲肝病毒、乙肝病毒)、脊髓灰质炎病毒、狂犬病毒、HPV病毒、脑炎病毒(如乙型脑炎病毒)、腮腺炎病毒、风疹病毒、破伤风杆菌、百日咳杆菌、白喉杆菌、麻风杆菌、结核杆菌、脑膜炎双球菌、肺炎双球菌及其任意组合。
- 根据权利要求12或13所述的方法,其中所述新生沉淀锰和/或胶体锰选自磷酸锰盐、碳酸锰盐、氢氧化锰及其任意混合物。
- 根据权利要求12至14任一项所述的方法,其中所述新生沉淀锰从非沉淀形式转化成沉淀形式的时间不超过1天,或者不超过24、22、20、18、16、15、14、12、10、9、8、7、6、5、4、3、2或1小时,或者不超过60、50、45、40、35、30、25、20、15、10、9、8、7、6、5、4、3、2或1分钟。
- 新生沉淀锰、胶体锰和/或能够形成新生沉淀锰和或胶体锰的源在制备免疫增强组合物或疫苗组合物中的用途。
- 根据权利要求16的用途,其中所述免疫增强组合物是免疫佐剂。
- 根据权利要求16或17的用途,其中所述新生沉淀锰和/或胶体锰选自磷酸锰盐、碳酸锰盐、氢氧化锰及其任意混合物。
- 根据权利要求16至18任一项所述的用途,其中所述新生沉淀锰从非沉淀形式转化成沉淀形式的时间不超过1天,不超过24、22、20、18、16、15、14、12、10、9、8、7、6、5、4、3、2或1小时,或者不超过60、50、 45、40、35、30、25、20、15、10、9、8、7、6、5、4、3、2或1分钟。
- 根据权利要求16至19任一项所述的用途,其中所述免疫增强是改善固有免疫和/或适应性免疫。
- 根据权利要求16至20任一项所述的用途,其中所述免疫增强是增加I型干扰素表达。
- 根据权利要求16至20任一项所述的用途,其中所述免疫增强是诱导炎症因子切割产生活性形式。
- 根据权利要求16至20任一项所述的用途,其中所述免疫增强是促进抗体产生。
- 一种增强免疫的方法,其包括,向有此需要的对象施用根据权利要求1至4任一项所述的或者根据权利要求9至11任一项所述方法获得的免疫增强组合物,具体地,所述免疫增强例如是A)改善固有免疫和/或适应性免疫,B)增加I型干扰素表达,C)诱导产生活性形式的炎症因子,D)促进抗体产生,E)促进T细胞增殖,和/或F)促进树突细胞成熟。
- 根据权利要求24的方法,其中所述施用选自肌肉注射、皮内注射、皮下注射、静脉注射、粘膜施用及其任意组合。
- 根据权利要求24或25的方法,其中所述增强免疫是用于预防和/或治疗疾病,例如细菌感染、真菌感染、病毒感染、寄生虫感染、肿瘤、自身免疫病。
- 根据权利要求24至26任一项所述的方法,其中所述病毒选自:DNA病毒和RNA病毒,优选地所述病毒选自:疱疹病毒科、弹状病毒科、丝状病毒科、正粘病毒科、副粘病毒科、冠状病毒科、小RNA病毒科、嗜肝DNA病毒科、黄病毒科、乳头瘤病毒科、痘病毒科、和逆转录病毒科,具体地所述 病毒选自:单纯疱疹病毒、水疱性口炎病毒、牛痘病毒、HIV、流感病毒、肝炎病毒(如甲肝病毒、乙肝病毒)、脊髓灰质炎病毒、狂犬病毒、HPV病毒、脑炎病毒(如乙型脑炎病毒)、腮腺炎病毒、风疹病毒及其任意组合。
- 根据权利要求24至26任一项所述的方法,其中所述细菌选自革兰氏阴性菌和革兰氏阳性菌,优选地所述细菌选自肺炎链球菌(Streptococcus pneumoniae)、流感嗜血杆菌(Haemophilus influenzae)、沙门氏菌(Salmonella)、脑膜炎双球菌(Meningococcus)、表皮葡萄球菌(Staphylococcus epidermidis)、金黄色葡萄球菌(Staphylococcus aureus)、大肠杆菌(Escherichia coli)、肺炎克雷伯氏菌(Klebsiella pneumoniae)、产酸克雷伯氏菌(Klebsiella oxytoca)、阴沟肠杆菌(Enterobacter cloacae)、弗氏柠檬酸杆菌(Citrobacter freundii)、绿脓假单胞菌(Pseudomonas aeruginosa)、波美不动杆菌(Acinetobacter baumanni)、破伤风杆菌、百日咳杆菌、白喉杆菌、麻风杆菌、结核杆菌、脑膜炎双球菌、肺炎双球菌及其任意组合。
- 根据权利要求24至26任一项所述的方法,其中所述自身免疫性疾病选自I型糖尿病、银屑病、类风湿性关节炎、系统性红斑狼疮和多发性硬化。
- 根据权利要求24至26任一项所述的方法,其中所述肿瘤选自卵巢癌、肺癌、胃癌、乳腺癌、肝癌、胰腺癌、皮肤癌、恶性黑色素瘤、头颈癌、肉瘤、胆管癌、膀胱癌、肾癌、结肠癌、胎盘绒毛膜癌、子宫颈癌、睾丸癌、子宫癌和白血病。
- 根据权利要求24至26任一项所述的方法,其中所述寄生虫是细胞内寄生虫,优选地选自疟原虫、弓形虫、锥虫、血吸虫、丝虫和利什曼原虫。
- 根据权利要求24至26任一项所述的方法,其还包括向所述对象施用另外的预防/治疗剂。
- 一种免疫接种方法,其包括向有此需要的对象施用根据权利要求5至8任一项所述的疫苗组合物,其中当组分A和组分B处于不同容器中时可以同时或不同时施用;和/或向有此需要的对象施用根据权利要求1至4任一项所述的免疫增强组合物和任选地疫苗免疫原。
- 根据权利要求33的方法,其中所述施用选自肌肉注射、皮内注射、 皮下注射、静脉注射、粘膜施用及其任意组合。
- 根据权利要求33或34的方法,其中所述免疫接种用于预防疾病,例如细菌感染、真菌感染、病毒感染、寄生虫感染、肿瘤和自身免疫病。
- 根据权利要求33至35任一项所述的方法,其中所述病毒选自:DNA病毒和RNA病毒,优选地所述病毒选自:疱疹病毒科、弹状病毒科、丝状病毒科、正粘病毒科、副粘病毒科、冠状病毒科、小RNA病毒科、嗜肝DNA病毒科、黄病毒科、乳头瘤病毒科、痘病毒科、和逆转录病毒科,具体地所述病毒选自:单纯疱疹病毒、水疱性口炎病毒、牛痘病毒、HIV、流感病毒、肝炎病毒(如甲肝病毒、乙肝病毒)、脊髓灰质炎病毒、狂犬病毒、HPV病毒、脑炎病毒(如乙型脑炎病毒)、腮腺炎病毒、风疹病毒及其任意组合。
- 根据权利要求33至35任一项所述的方法,其中所述细菌选自革兰氏阴性菌和革兰氏阳性菌,优选地所述细菌选自肺炎链球菌(Streptococcus pneumoniae)、流感嗜血杆菌(Haemophilus influenzae)、沙门氏菌(Salmonella)、脑膜炎双球菌(Meningococcus)、表皮葡萄球菌(Staphylococcus epidermidis)、金黄色葡萄球菌(Staphylococcus aureus)、大肠杆菌(Escherichia coli)、肺炎克雷伯氏菌(Klebsiella pneumoniae)、产酸克雷伯氏菌(Klebsiella oxytoca)、阴沟肠杆菌(Enterobacter cloacae)、弗氏柠檬酸杆菌(Citrobacter freundii)、绿脓假单胞菌(Pseudomonas aeruginosa)、波美不动杆菌(Acinetobacter baumanni)、破伤风杆菌、百日咳杆菌、白喉杆菌、麻风杆菌、结核杆菌、脑膜炎双球菌、肺炎双球菌及其任意组合。
- 根据权利要求33至35任一项所述的方法,其中所述自身免疫性疾病选自I型糖尿病、银屑病、类风湿性关节炎、系统性红斑狼疮和多发性硬化。
- 根据权利要求33至35任一项所述的方法,其中所述肿瘤选自卵巢癌、肺癌、胃癌、乳腺癌、肝癌、胰腺癌、皮肤癌、恶性黑色素瘤、头颈癌、肉瘤、胆管癌、膀胱癌、肾癌、结肠癌、胎盘绒毛膜癌、子宫颈癌、睾丸癌、子宫癌和白血病。
- 根据权利要求33至35任一项所述的方法,其中所述寄生虫是细胞内寄生虫,优选地选自疟原虫、弓形虫、锥虫、血吸虫、丝虫和利什曼原虫。
- 根据权利要求33至35任一项所述的方法,其还包括向所述对象施用另外的预防剂。
Priority Applications (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
KR1020217037373A KR102702643B1 (ko) | 2019-04-19 | 2020-04-20 | 면역 증강용 망간 조합물 |
JP2021561978A JP7308562B2 (ja) | 2019-04-19 | 2020-04-20 | 免疫増強用マンガン組成物 |
EP20791542.2A EP3957312B1 (en) | 2019-04-19 | 2020-04-20 | Manganese combination for immunological enhancement |
US17/594,468 US20220193125A1 (en) | 2019-04-19 | 2020-04-20 | Manganese composition for enhancing immunity |
CN202080029675.2A CN113692280A (zh) | 2019-04-19 | 2020-04-20 | 用于免疫增强的锰组合物 |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201910319344.1 | 2019-04-19 | ||
CN201910319344.1A CN111821316A (zh) | 2019-04-19 | 2019-04-19 | 用于免疫增强的锰组合物 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020211857A1 true WO2020211857A1 (zh) | 2020-10-22 |
Family
ID=72837045
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2020/085507 WO2020211857A1 (zh) | 2019-04-19 | 2020-04-20 | 用于免疫增强的锰组合物 |
Country Status (6)
Country | Link |
---|---|
US (1) | US20220193125A1 (zh) |
EP (1) | EP3957312B1 (zh) |
JP (1) | JP7308562B2 (zh) |
KR (1) | KR102702643B1 (zh) |
CN (2) | CN111821316A (zh) |
WO (1) | WO2020211857A1 (zh) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113274550A (zh) * | 2021-05-31 | 2021-08-20 | 福州大学 | 一种具有抗炎作用的血管化骨仿生多功能组织工程支架及其制备方法 |
CN113797329A (zh) * | 2021-10-19 | 2021-12-17 | 启锰生物科技(江苏)有限公司 | 一种二价锰佐剂和CpG佐剂的疫苗佐剂组合物及其制作方法 |
WO2023123959A1 (zh) * | 2021-12-28 | 2023-07-06 | 广东粤港澳大湾区国家纳米科技创新研究院 | 一种铝锰复合纳米晶及其制备方法和应用 |
Families Citing this family (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111643454B (zh) * | 2020-06-22 | 2021-06-15 | 北京大学 | 用于免疫治疗的含锰微沉淀脂质体及其制备方法 |
CN112791181B (zh) * | 2021-02-05 | 2024-04-02 | 广东粤港澳大湾区国家纳米科技创新研究院 | 一种锰纳米佐剂、其制备方法及用途 |
US20240261382A1 (en) * | 2021-06-07 | 2024-08-08 | Innobm Pharmaceuticals Co., Ltd. | Modified bacterium, preparation method therefor and application thereof |
CN113817677B (zh) * | 2021-09-29 | 2023-08-18 | 四川大学 | 泛酸或其衍生物与α-D-葡萄糖-1,6-二磷酸或其衍生物在促进DC迁移中的用途 |
WO2024012580A1 (zh) * | 2022-07-15 | 2024-01-18 | 上海石趣医药科技有限公司 | 含锰的化合物在降低哺乳动物个体的尿酸水平中的用途 |
CN117100852A (zh) * | 2023-10-24 | 2023-11-24 | 江苏瑞科生物技术股份有限公司 | 复合佐剂及其制备方法和应用 |
Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN1542449A (zh) * | 2003-04-30 | 2004-11-03 | �����ɷ� | 一种核/壳型超顺磁性复合微粒及其制备方法与应用 |
CN101151532A (zh) * | 2005-03-29 | 2008-03-26 | 因弗因斯医药瑞士股份有限公司 | 胶态金属轭合物 |
CN101225092A (zh) * | 2007-12-26 | 2008-07-23 | 首都师范大学 | 一种锰配合物及其制备方法与应用 |
CN102515276A (zh) * | 2011-12-30 | 2012-06-27 | 四川大学 | 一种基于牛血清蛋白为模板制备二氧化锰纳米粒子的方法 |
CN107412260A (zh) | 2016-05-23 | 2017-12-01 | 北京大学 | cGAS-STING通路激活剂及其用途 |
CN107456575A (zh) * | 2017-09-06 | 2017-12-12 | 苏州大学 | 一种二氧化锰纳米佐剂及其制备方法、应用 |
Family Cites Families (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US2031579A (en) * | 1933-04-13 | 1936-02-18 | Swann Res Inc | Preparation of phosphates of manganese |
US4720386A (en) * | 1976-11-24 | 1988-01-19 | Mccollester Duncan L | Vaccine and method for immunotherapy of neoplastic disease |
FR2823119B1 (fr) * | 2001-04-05 | 2004-02-20 | Seppic Sa | Adjuvant d'immunite contenant un cation metallique complexe et vaccin le contenant |
US20040115208A1 (en) * | 2002-12-16 | 2004-06-17 | Li Frank Q. | Method of using colloidal metal-protein composition for treatment of cancer |
-
2019
- 2019-04-19 CN CN201910319344.1A patent/CN111821316A/zh active Pending
-
2020
- 2020-04-20 WO PCT/CN2020/085507 patent/WO2020211857A1/zh unknown
- 2020-04-20 EP EP20791542.2A patent/EP3957312B1/en active Active
- 2020-04-20 CN CN202080029675.2A patent/CN113692280A/zh active Pending
- 2020-04-20 JP JP2021561978A patent/JP7308562B2/ja active Active
- 2020-04-20 KR KR1020217037373A patent/KR102702643B1/ko active IP Right Grant
- 2020-04-20 US US17/594,468 patent/US20220193125A1/en active Pending
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN1542449A (zh) * | 2003-04-30 | 2004-11-03 | �����ɷ� | 一种核/壳型超顺磁性复合微粒及其制备方法与应用 |
CN101151532A (zh) * | 2005-03-29 | 2008-03-26 | 因弗因斯医药瑞士股份有限公司 | 胶态金属轭合物 |
CN101225092A (zh) * | 2007-12-26 | 2008-07-23 | 首都师范大学 | 一种锰配合物及其制备方法与应用 |
CN102515276A (zh) * | 2011-12-30 | 2012-06-27 | 四川大学 | 一种基于牛血清蛋白为模板制备二氧化锰纳米粒子的方法 |
CN107412260A (zh) | 2016-05-23 | 2017-12-01 | 北京大学 | cGAS-STING通路激活剂及其用途 |
CN107456575A (zh) * | 2017-09-06 | 2017-12-12 | 苏州大学 | 一种二氧化锰纳米佐剂及其制备方法、应用 |
Non-Patent Citations (23)
Title |
---|
A. M. BRUNSC. M. HORVATH: "Activation of RIG-I-like receptor signal transduction", CRITICAL REVIEWS IN BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 47, 2012, pages 194 - 206 |
C. K. TANGT. AOSHIN. JOUNAIJ. ITOK. OHATAK. KOBIYAMAB. H. DESSAILLYE. KURODAS. AKIRAK. MIZUGUCHI: "The chemotherapeutic agent DMXAA as a unique IRF3-dependent type-2 vaccine adjuvant.", PLOS ONE, vol. 8, pages e60038 |
C. WANGY. GUANM. LVR. ZHANGZ. GUOX. WEIX. DUJ. YANGT. LIY. WAN: "Manganese Increases the Sensitivity of the cGAS-STING Pathway for Double-Stranded DNA and Is Required for the Host Defense against DNA Viruses", IMMUNITY, vol. 48, pages 675 - 687 |
E. C. CARROLLL. JINA. MORIN. MUNOZ-WOLFE. OLESZYCKAH. B. MORANS. MANSOURIC. P. MCENTEEE. LAMBEE. M. AGGER: "The Vaccine Adjuvant Chitosan Promotes Cellular Immunity via DNA Sensor cGAS-STING-Dependent Induction of Type I Interferons", IMMUNITY, vol. 44, 2016, pages 597 - 608, XP029448981, DOI: 10.1016/j.immuni.2016.02.004 |
E. OLESZYCKAH. B. MORANG. A. TYNANC. H. HEARNDENG. COUTTSM. CAMPBELLS. M. ALLANC. J. SCOTTE. C. LAVELLE: "IL-lalpha and inflammasome-independent IL-lbeta promote neutrophil infiltration following alum vaccination", FEBS J, vol. 283, 2016, pages 9 - 24 |
H. LIS. B. WILLINGHAMJ. P. Y. TINGF. RE: "Cutting Edge: Inflammasome Activation by Alum and Alum's Adjuvant Effect Are Mediated by NLRP3", THE JOURNAL OF IMMUNOLOGY, vol. 181, 2008, pages 17 - 21 |
H. NOHYNEKJ. JOKINENM. PARTINENO. VAARALAT. KIRJAVAINENJ. SUNDMANS. L. HIMANENC. HUBLINI. JULKUNENP. OLSEN: "AS03 adjuvanted AH1N1 vaccine associated with an abrupt increase in the incidence of childhood narcolepsy in Finland", PLOS ONE, vol. 7, 2012, pages e33536 |
K. SCHRODERJ. TSCHOPP, THE INFLAMMASOMES. CELL, vol. 140, 2010, pages 821 - 832 |
L. B. IVASHKIVL. T. DONLIN: "Regulation of type I interferon responses", NATURE REVIEWS. IMMUNOLOGY, vol. 14, 2014, pages 36 - 49 |
M. KOOLV. PETRILLIT. DE SMEDTA. ROLAZH. HAMMADM. VAN NIMWEGENI. M. BERGENR. CASTILLOB. N. LAMBRECHTJ. TSCHOPP: "Cutting Edge: Alum Adjuvant Stimulates Inflammatory Dendritic Cells through Activation of the NALP3 Inflammasome", THE JOURNAL OF IMMUNOLOGY, vol. 181, 2008, pages 3755 - 3759 |
M. P. LONGHIC. TRUMPFHELLERJ. IDOYAGAM. CASKEYI. MATOSC. KLUGERA. M. SALAZARM. COLONNAR. M. STEINMAN: "Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Thl immunity with poly IC as adjuvant", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 206, 2009, pages 1589 - 1602 |
N. S. WILSONP. DUEWELLB. YANGY. LIS. MARSTERSS. KOEMIGE. LATZE. MARASKOVSKYA. B. MORELLIM. SCHNURR: "Inflammasome-dependent and -independent IL-18 production mediates immunity to the ISCOMATRIX adjuvant.", JOURNAL OF IMMUNOLOGY, vol. 192, 2014, pages 3259 - 3268 |
P. MARRACKA. S. MCKEEM. W. MUNKS: "Towards an understanding of the adjuvant action of aluminium", NATURE REVIEWS. IMMUNOLOGY, vol. 9, 2009, pages 287 - 293, XP002719141, DOI: 10.1038/nri2510 |
S. AKIRAK. TAKEDA: "Toll-like receptor signalling", NATURE REVIEWS. IMMUNOLOGY, vol. 4, 2004, pages 499 - 511, XP009128219, DOI: 10.1038/nri1391 |
S. C. EISENBARTHO. R. COLEGIOW. O'CONNORF. S. SUTTERWALAR. A. FLAVELL: "Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants", NATURE, vol. 453, 2008, pages 1122 - 1126 |
S. M. BLAAUBOERV. D. GABRIELLEL. JIN: "MPYS/STING-mediated TNF-alpha, not type I IFN, is essential for the mucosal adjuvant activity of (3'-5')-cyclic-di-guanosine-monophosphate in vivo.", JOURNAL OF IMMUNOLOGY, vol. 192, 2014, pages 492 - 502, XP055690352, DOI: 10.4049/jimmunol.1301812 |
S. Z. BEN-SASSONJ. HU-LIJ. QUIELS. CAUCHETAUXM. RATNERI. SHAPIRAC. A. DINARELLOW. E. PAU: "IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 106, 2009, pages 7119 - 7124, XP055077266, DOI: 10.1073/pnas.0902745106 |
See also references of EP3957312A4 |
W. M. SCHNEIDERM. D. CHEVILLOTTEC. M. RICE: "Interferon-stimulated genes: a complex web of host defenses", ANNU REV IMMUNOL, vol. 32, 2014, pages 513 - 545 |
X. D. LIJ. WUD. GAOH. WANGL. SUNZ. J. CHEN: "Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immunoadjuvant effects", SCIENCE, vol. 341, 2013, pages 1390 - 1394 |
Y. XIAY. XIEZ. YUH. XIAO, G. JIANG, X. ZHOU, Y. YANG, X. LI, M. ZHAO, L. LIM. ZHENGS. HANZ. ZONGX. MENGH. DENGH. YE: "The Mevalonate Pathway Is a Druggable Target for Vaccine Adjuvant Discovery", CELL, vol. 175, pages 1059 - 1073 |
Z. JIANGP. GEORGELX. DUL. SHAMELS. SOVATHS. MUDDM. HUBERC. KALISS. KECKC. GALANOS: "CD14 is required for MyD88-independent LPS signaling", NATURE IMMUNOLOGY, vol. 6, 2005, pages 565 - 570, XP002519138, DOI: 10.1038/ni1207 |
Z. MAB. DAMANIA: "The cGAS-STING Defense Pathway and Its Counteraction by Viruses", CELL HOST MICROBE, vol. 19, 2016, pages 150 - 158, XP029415479, DOI: 10.1016/j.chom.2016.01.010 |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113274550A (zh) * | 2021-05-31 | 2021-08-20 | 福州大学 | 一种具有抗炎作用的血管化骨仿生多功能组织工程支架及其制备方法 |
CN113274550B (zh) * | 2021-05-31 | 2022-06-14 | 福州大学 | 一种具有抗炎作用的血管化骨仿生多功能组织工程支架及其制备方法 |
CN113797329A (zh) * | 2021-10-19 | 2021-12-17 | 启锰生物科技(江苏)有限公司 | 一种二价锰佐剂和CpG佐剂的疫苗佐剂组合物及其制作方法 |
WO2023123959A1 (zh) * | 2021-12-28 | 2023-07-06 | 广东粤港澳大湾区国家纳米科技创新研究院 | 一种铝锰复合纳米晶及其制备方法和应用 |
Also Published As
Publication number | Publication date |
---|---|
KR102702643B1 (ko) | 2024-09-05 |
KR20220002968A (ko) | 2022-01-07 |
US20220193125A1 (en) | 2022-06-23 |
EP3957312A1 (en) | 2022-02-23 |
CN111821316A (zh) | 2020-10-27 |
CN113692280A (zh) | 2021-11-23 |
JP7308562B2 (ja) | 2023-07-14 |
EP3957312A4 (en) | 2023-02-01 |
JP2022529071A (ja) | 2022-06-16 |
EP3957312B1 (en) | 2024-08-28 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2020211857A1 (zh) | 用于免疫增强的锰组合物 | |
JP2021191783A (ja) | 新生児及び乳幼児におけるワクチン接種 | |
EP2306993B1 (en) | Protamine/rna nanoparticles for immunostimulation | |
ES2380481T3 (es) | Sustancias inmunogénicas que comprenden un adyuvante a base de ácido poliinosínico y ácido policitidílico | |
US20100150960A1 (en) | Compositions and methods for chitosan enhanced immune response | |
CN107412260B (zh) | cGAS-STING通路激活剂及其用途 | |
RU2746118C2 (ru) | Состав для введения рнк | |
KR20150102995A (ko) | 경비 인플루엔자 백신 조성물 | |
CN109701010B (zh) | 疫苗复合佐剂系统及其在抗原中的应用 | |
US20100310600A1 (en) | Selective agonist of toll-like receptor 3 | |
CA2719252C (en) | Anti-infective agents comprising muramyl dipeptide microparticles and uses thereof | |
JP2022062175A (ja) | 免疫応答を誘導するための方法および手段 | |
EP2754436A1 (en) | Biodegradable high-efficiency dengue vaccine, method for making the same, and pharmaceutical composition comprising the same | |
EP2950816B1 (en) | The use of dna sequences encoding an interferon as vaccine adjuvants | |
KR20070122056A (ko) | 콜로이드 금의 아쥬반트로의 사용 | |
CN109701011B (zh) | 疫苗复合佐剂系统及其在抗原中的应用 | |
Ohta | Cholesteryl pullulan nanoparticles-encapsulated TNF-α: an effective mucosal vaccine adjuvant against influenza | |
EP4431111A1 (en) | Probiotic as vaccine immunoadjuvant | |
TWI384996B (zh) | 以聚肌苷酸-聚胞苷酸為主的佐劑 | |
CN117100852A (zh) | 复合佐剂及其制备方法和应用 | |
KR20040006196A (ko) | 양이온성 고분자를 함유하는 어쥬반트 유전자 조성물 |
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: 20791542 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2021561978 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 20217037373 Country of ref document: KR Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 2020791542 Country of ref document: EP Effective date: 20211119 |