WO2014075631A1 - Method for preparing autologous tumor vaccine and use thereof - Google Patents

Method for preparing autologous tumor vaccine and use thereof Download PDF

Info

Publication number
WO2014075631A1
WO2014075631A1 PCT/CN2013/087266 CN2013087266W WO2014075631A1 WO 2014075631 A1 WO2014075631 A1 WO 2014075631A1 CN 2013087266 W CN2013087266 W CN 2013087266W WO 2014075631 A1 WO2014075631 A1 WO 2014075631A1
Authority
WO
WIPO (PCT)
Prior art keywords
tumor
vaccine
tumor cells
cell
cell inactivation
Prior art date
Application number
PCT/CN2013/087266
Other languages
French (fr)
Chinese (zh)
Inventor
韩德平
林建华
张鲁榕
张美�
张震寰
张亨山
吕文龙
洪金省
Original Assignee
厦门鹭佳生物科技有限公司
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 厦门鹭佳生物科技有限公司 filed Critical 厦门鹭佳生物科技有限公司
Publication of WO2014075631A1 publication Critical patent/WO2014075631A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5152Tumor cells

Definitions

  • the present invention is in the field of biotechnology and immunology, and in particular, the present invention relates to a method for preparing an autologous tumor vaccine and an application thereof. Background technique
  • Tumors are one of the leading causes of death in humans. Although the level of diagnosis and treatment of tumors has been continuously improved and improved in recent years, the programs of chemotherapy and radiotherapy are constantly improving, but in the end, most patients still have difficulty to escape the fate of death.
  • malignant tumors have multiple immune escape mechanisms, including low antigen content in tumor cells, destruction or constant mutation of tumor antigens, poor treatment of tumor antigens, or lack of expression of some immune cofactors, or low immune function. Etc., the clinical treatment effect of existing tumor tumor vaccines is not good. Because of the heterogeneity, diversity, and variability of malignant tumors, the individual tumor antigens of different individuals are very different when they are suffering from the same kind of tumor. Thus, tumor immunotherapy should be individualized to induce active immunity of individual-specific killing autologous tumor cells.
  • mice are usually immunized with inactivated tumor cells to protect the mice from inoculation of the inoculated tumor.
  • the vaccine loses the ability to protect the mouse.
  • the current clinical response to tumor cell vaccines is relatively poor, and it is only suitable for preventing recurrence of tumor patients without special tumor antigens. Very few good results have been obtained in clinical studies for advanced patients.
  • An ideal tumor-specific antigen should be immunogenic, expressed by tumor cells but not expressed in normal cells. Unfortunately, most tumor antigens do not have sufficient immunogenicity to induce an effective immune response.
  • Another object of the present invention is to provide an autologous tumor vaccine and use thereof.
  • a method of preparing an autologous tumor vaccine comprising the steps of:
  • step (1) treating the autologous tumor cells of step (1) by radiotherapy and/or heating to obtain tumor cells having enhanced antigenicity and antigen-promoting properties;
  • the inactivated tumor cells obtained in the step (3) were prepared as an autologous tumor vaccine.
  • the tumor is: a solid tumor or a non-solid tumor.
  • the solid tumor is derived from: brain, lung, head and neck, skin, pancreas, gastrointestinal tract, bladder, genital tract, spinal cord, spleen, kidney, liver, limbs, bones, tongue, Or throat.
  • the non-solid tumor is derived from blood or bone marrow.
  • the tumor is a primary tumor or a secondary metastatic tumor.
  • the tumor is derived from a tissue of a non-human mammalian tissue or a human.
  • the method for obtaining the isolated autologous tumor cells according to the step (1) is selected from the group consisting of: enzymatic digestion, mechanical shearing, or artificial grinding of tumor tissue to treat tumor tissue to be culturable. Isolated autologous tumor cells.
  • the radiation method described in the step (2) is selected from the group consisting of: selected from ⁇ , ⁇ , ⁇ , proton, or heavy ion radiation, optionally selected from the range of 0.1-50 Gy, optionally
  • the autologous tumor cells of step (1) were treated at a dose rate of 0.1-10 Gy/min to obtain antigenic and antigen-presenting tumor cells.
  • the radiation is derived from a natural or artificial source of radiation.
  • heating method described in the step (2) is selected from the group consisting of: heating temperature is selected from the group consisting of
  • the heating time is selected from 1 min to 2 hours (preferably 30 min - l hour).
  • the method further comprises the step of: culturing the tumor cells obtained by the step (2) to obtain antigenicity and antigen-promoting enhancement.
  • the culture treatment time is selected from the group consisting of: 6 hours to 2 weeks.
  • a basic cell culture solution is used, and/or a growth factor or a small molecule compound is added.
  • the inactivation treatment described in the step (3) is selected from the group consisting of microwave cell inactivation, electromagnetic wave cell inactivation, laser cell inactivation, high temperature cell inactivation, sonication and cell disruption. Live method, repeated cell freeze-thaw, homogenate, centrifugation, pellet cell inactivation, enzymatic digestion cell inactivation, hypotonic rupture, or a combination thereof.
  • step (4) the inactivated tumor cells obtained in the step (3) are mixed with an adjuvant to obtain an autologous tumor vaccine.
  • the adjuvant is Freund's complete adjuvant or incomplete adjuvant.
  • the adjuvant is selected from the group consisting of liposomes, cytokines (eg, GM-CSF, G-CSF,
  • CSF CSF, IFN, IL-2, etc.
  • various plant proteins DNA, deactivated or inactivated viruses, bacteria, fungi, microorganisms and their extracts, or combinations thereof.
  • an autologous tumor vaccine prepared by the method of the first aspect.
  • the vaccine further comprises an adjuvant.
  • the autologous tumor vaccine of the second aspect which is used for the preparation of a composition for preventing or treating a tumor.
  • the composition is a pharmaceutical composition or a vaccine composition.
  • the composition is in the form of an injection.
  • the injection is administered by a route selected from the group consisting of intradermal single/multiple injection, subcutaneous single/multiple injection, intralymphatic or peripheral injection, intramuscular injection, intrathoracic/peritoneal injection, Intravenous, peritumoral normal tissue injection.
  • it is the distal end of the intradermal limb.
  • the injection site of the injection is selected from the group consisting of: primary tumor, metastases, lymphatic drainage area around the primary tumor, lymphatic drainage area around the metastasis, distal extremities, proximal extremities, chest Back, abdomen, neck, locally accessible lymph nodes or lymphatic drainage areas.
  • the vaccine can be used in combination with a drug selected from the group consisting of cytokines, electromagnetic wave therapy, or a combination thereof.
  • the drug is selected from the group consisting of IFN, TNF, IL-2, or a combination thereof.
  • the inactivation treatment described in the step (3) is selected from the group consisting of microwave cell inactivation, electromagnetic wave cell inactivation, laser cell inactivation, high temperature cell inactivation, sonication and cell disruption. Live method, repeated cell freeze-thaw, homogenate, centrifugation, pellet cell inactivation, enzymatic digestion, cell inactivation, hypotonic rupture, chemical covalent Combined method, chemical denaturing method, or a combination thereof.
  • a method of preventing or treating a tumor comprising administering an effective amount of the tumor vaccine of the second aspect of the invention to a subject is provided.
  • the administration time is selected from the group consisting of: postoperative, pre-chemotherapy, or WBC rebound to >3000 per ul.
  • the number of administrations may be 4 courses (3-4 weeks per course) to life.
  • the method further comprises: monitoring the extent of the immune response using an immunological assay.
  • the immunological assay is selected from the group consisting of a cellular immune response, a humoral immune response, an immunomodulatory response, or a combination thereof.
  • the method for detecting cellular immune response comprises:
  • the anti-tumor delayed allergic reaction can be seen as a cumulus 0.5 cm or more for a good immune response, ⁇ 0.5 cm for poor immune response;
  • Lymphocyte transformation reaction The lymphocytes of the patient one month after immunization are separated, and the vaccine of the second aspect of the invention is added to observe the conversion rate (preferably by BrdU method, MTT method), and the conversion rate is high. Good immune response;
  • the lymphokine is selected from the group consisting of IL-2, IL-4, IL-6, IL-10, IL-12, IL-17, IL-18, TNF-a, IFN- ⁇ , Or a combination thereof;
  • the detection method is selected from the group consisting of ELISPOT, ELISA, FCM, or a combination thereof.
  • the subset of immune cells is selected from the group consisting of CD4+, CD8+, CD1 lb+, Grl+, or a combination thereof;
  • the killer activation marker is selected from the group consisting of Fas L, LIGHT, CD25, TNF-a, IFN- ⁇ , perforin or a combination thereof.
  • each subpopulation is detected by flow cytometry.
  • the humoral immunoassay method comprises:
  • the prepared tumor vaccine is used as an antigen coating to add plasma before and after autoimmunization, and the antibody titer against autoantitumor antigen is compared before and after immunization. The higher the titer, the stronger the immunoreactivity.
  • the molecular weight of the autoantigen is determined by the WESTERN BLOT method.
  • the immunomodulatory reaction detecting method comprises:
  • Detection of a subset of immunoregulatory cells comprises: regulatory T cells: Treg: CD4 + CD25 + , myeloid-derived suppressor cells MDSC: CD14 - CD11b + , tumor-associated macrophages TAM: Ml/M2, regulatory dendritic cell DCreg: CD11b+/Gr-l+, or a combination thereof.
  • the detection method is flow cytometry.
  • the immunoregulatory factors include: IL-2, IL-4, IL-6, IL-10, IL-12, IL-17, IL-18 , TNF-a, IFN- ⁇ , or a combination thereof;
  • the assay comprises: ELISA, LUMINEX bead-array assay, or a combination thereof.
  • the immunological assay is performed within 15-60 days after the first immunization.
  • the method further comprises: determining the therapeutic effect and/or adjusting the treatment regimen based on the degree of immune response measured by the immunological experiment.
  • the adjusted treatment regimen refers to adjusting one or more therapeutic parameters selected from the group consisting of: immunoadjuvant binding, immunization dose, immunization site, immunization time, number of immunizations, low dose radiation, other physics Or biological methods to stimulate immune organs.
  • the immune organ is bone marrow.
  • the method includes:
  • the administration of the vaccine and its effect observation time may vary flexibly due to individual differences. It is to be understood that within the scope of the present invention, the various technical features of the present invention and the technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here. DRAWINGS
  • Figure 1 shows the results of anti-tumor antibody IgG assay 3 weeks after injection of the auto-tumor vaccine.
  • Figure 2 shows an anti-tumor T cell killing experiment.
  • FIG. 3 shows the experimental grouping and the operational flow.
  • Figure 4 shows the results of real-time dynamic measurement of anti-tumor effect fluorescence (Live Image).
  • Fig. 5 shows the results of the vaccine antigenic cancer (4T1-Luc) effect (test 1) prepared by the method of Example 1 of the present invention.
  • Fig. 6 shows the results of the vaccine antigenic cancer (4T1-Luc) effect (test 2) prepared by the method of Example 1 of the present invention.
  • Figure 7 is a graph showing the results of a comparison between the vaccine prepared by the method of Example 1 of the present invention and the anticancer effect of Taxol (Trial 3, compared with the chemotherapy drug Taxol).
  • Figure 8 shows the killing inhibition effect of the method of one embodiment of the present invention on experimental lung metastasis.
  • FIG. 9 shows the flow of an autologous tumor vaccine prepared in accordance with one embodiment of the present invention.
  • Fig. 10 shows a tumor cell illuminator; wherein, the Y-ray is used to break the DNA and cause DNA error repair, and at the same time, the characteristics of the oncogenic gene which promotes growth in a harsh environment are highly expressed, and the tumor cells are treated with the Y illuminator of the exclusive invention.
  • the oncogene is highly expressed, and the activation of tumor antigen is enhanced to increase the efficacy of the method.
  • Figure 11 shows a bone marrow growth stimulator; wherein, using medium/high frequency electromagnetic waves to stimulate cell growth, the bone marrow growth stimulator is placed in the humerus of tumor-free cells to stimulate hematopoietic stem cell proliferation and differentiation, so that autoantibodies The number of cancer immune cells is increased to increase the efficacy of this method.
  • the present inventors have discovered for the first time that the treatment of radiation and/or heating of autologous tumor cells can greatly stimulate the antigenicity of autologous tumor cells, and heat shock proteins enhance antigen presentation;
  • the cells are inactivated and supplemented with adjuvant to make a vaccine.
  • the vaccine has the special advantages of strong presentation and strong immunogenicity of the tumor, and can strongly stimulate the humoral and cellular immune responses of the tumor patients to the autologous tumor, so as to improve the antigenic and metastatic tumor effect and prolong the survival of the tumor patient.
  • the purpose of time On this basis, the invention of a novel autologous tumor vaccine was completed. Its application is also based on monitoring the patient's response to autologous tumor vaccines, changing the amount of vaccine, the location and time of injection, the length and interval of treatment, and truly Active immunotherapy. Preparation
  • the present invention provides a method of preparing an autologous tumor vaccine, and in a preferred embodiment, comprising the steps of: (1) obtaining isolated autologous tumor cells;
  • step (1) treating the autologous tumor cells of step (1) by radiotherapy and/or heating to obtain tumor cells having enhanced antigenicity and antigen-promoting properties;
  • the inactivated tumor cells obtained in the step (3) were prepared as an autologous tumor vaccine.
  • the tumor cells as follows: Obtain the tumor-free, non-necrotic part of the tumor under sterile conditions, store in DMEM or other culture medium, store at 4 ° C, rinse with PBS, and digest by enzyme. Purified tumor cells were obtained by method or / and mechanical separation method, transferred into a culture flask, subcultured at 37 ° C, 5% CO 2 incubator, and reached the required number of cells to enter the tumor whole cell tumor preparation procedure;
  • the tumor cells in culture are treated by radiation and/or heating to excite the antigen, and the increased heat shock protein enhances antigen presentation.
  • the treated tumor cells are inactivated by the following methods: microwave cell inactivation method, electromagnetic wave cell inactivation method, laser cell inactivation method, high temperature cell inactivation method, ultrasonic disruption cell inactivation method, repeated cells At least one of freezing and thawing, homogenization, centrifugation, sedimentation cell inactivation, enzymatic digestion, cell inactivation, hypotonic rupture, and the like;
  • the invention is supplemented by the above tumor whole cell vaccine with an adjuvant
  • the adjuvant may be a complete Freund's adjuvant or an incomplete adjuvant, after a water-in-oil adjuvant, a liposome, a cytokine (such as GM-CSF, various Plant protein DNA), attenuating or inactivating an adjuvant such as a virus, a bacterium, a fungus, a microorganism or an extract thereof. After adjuvanting, it can better enhance the active immune response, reduce the negative regulation of immunity, and enhance the stimulation and memory of anti-tumor immune cells.
  • a cytokine such as GM-CSF, various Plant protein DNA
  • the method of the invention is to treat the tumor cells by antigenic stimulation treatment and enhance the antigen presenting ability, and then inactivate the treatment to lose the tumorigenicity, and retain the immunogenicity after the stimulation, and simultaneously combine the immunosupsis;
  • an individualized tumor vaccine we will establish an individualized treatment method and keep track of individual immunity. State and tumor conditions continue to adjust individualized immunotherapy programs to achieve optimal anti-tumor efficacy. Immune effect monitoring
  • One of ordinary skill in the art can determine the anti-tumor immune response of a vaccine using conventional methods. Including: anti-tumor humoral immune response evaluation, anti-tumor cell immune response evaluation, anti-tumor clinical response evaluation.
  • the anti-tumor humoral immune response was evaluated by using an individualized tumor vaccine as an antigen-coated ELISA method to detect the anti-autoantibody antibody titer in the serum of the individual before and after immunization of the individualized tumor vaccine to objectively judge the anti-tumor humoral immune response.
  • Anti-tumor cell immune response evaluation generally, including the new individualized tumor vaccine prepared by intradermal injection, observe the patient's own pimple size within 1-3 days (late-type individualized tumor vaccine immune response); or self-tumor stimulation specific Sexual lymphocyte transformation experiment: including steps: taking anticoagulation, separating lymphocytes, serum-free culture, adding individualized tumor vaccine or control vector, culturing, judging lymphocyte transformational proliferation level (BrdU incorporation method or MTT method); Individualized tumor vaccine immunomodulatory response.
  • Immunomodulatory responses to individualized tumor vaccines include: detection of leukocyte populations (eg, CD4/CD8/CD1 lb/Gr-l, etc.) and their surface killing or activating markers (eg, FasL/LIGHT/CD25/TNF-a/IFN-) y, etc., according to the results of these in vitro and in vivo experiments can be divided into two types: good response and poor response: high antibody titer, pyel is greater than 0.5 cm, so can maintain the original individualized immunization program; poor response antibody titer Low, pipi is less than 0.5 cm, so you can change the use of individualized tumor vaccine and use different means to enhance the immune response.
  • leukocyte populations eg, CD4/CD8/CD1 lb/Gr-l, etc.
  • their surface killing or activating markers eg, FasL/LIGHT/CD25/TNF-a/IFN-
  • the subset of immunoregulatory cells includes: regulatory T cell Treg (CD4+CD25+), myeloid-derived suppressor cell MDSC (CD14-CD1 lb+), tumor-associated macrophage TAM (M1/M2), regulatory dendritic
  • the cells were DCreg (CDl lb+/Gr-l+) and each subpopulation was detected by flow cytometry.
  • Anti-tumor clinical response According to the tumor image size evaluation, according to the experimental results, the patient's immune status, tumor condition and other anti-tumor treatment programs, decide when to strengthen anti-tumor immunity, adjust individualized treatment plan, continue 2, 3, 4 Or more treatments. Efficacy monitoring includes, but is not limited to, imaging criteria, tumor-free survival time, and overall survival time.
  • the immune response effect of individualized tumor vaccine can be monitored by cellular immunity, humoral immunity, and clinical tumor response.
  • the monitoring time can be about 15 days to 60 days after the first immunization, and the data can be evaluated if the reaction
  • individualized immunotherapy regimens can be re-adjusted, such as by adjusting immune adjuvant binding, immunization dose, immune site, immunization time, number of immunizations, and low-dose radiation-stimulated immune organs such as bone marrow.
  • the present invention also provides a composition, which may be a pharmaceutical composition or a vaccine composition.
  • the compositions of the invention may be therapeutic or prophylactic.
  • the compositions of the present invention comprise an effective amount of an autologous tumor vaccine of the invention, and at least one pharmaceutically acceptable carrier, diluent or excipient.
  • compositions of the present invention can be prepared into various conventional dosage forms including, but not limited to, injections, granules, tablets, pills, suppositories, capsules, suspensions, sprays and the like.
  • compositions of the present invention comprise an effective amount of an autologous tumor vaccine prepared by the method of the present invention.
  • an effective amount refers to an amount of a therapeutic agent that treats, alleviates or prevents a target disease or condition, or an amount that exhibits a detectable therapeutic or prophylactic effect. This effect can be detected by, for example, antigen level. Therapeutic effects also include a reduction in physiological symptoms.
  • the precise effective amount for a subject will depend on the size and health of the subject, the nature and extent of the condition, and the combination of therapeutic and/or therapeutic agents selected for administration. Therefore, it is useless to specify an accurate effective amount in advance. However, for a given situation, routine experimentation can be used to determine the effective amount.
  • an effective dose is from about 0.2 micrograms per kilogram to 2 micrograms per kilogram of the subject.
  • the pharmaceutical composition may also contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to a carrier for administration to a therapeutic agent, such as a protein, VLP or other therapeutic agent.
  • the term refers to pharmaceutical carriers which do not themselves induce the production of antibodies harmful to the individual receiving the composition and which are not excessively toxic after administration.
  • Suitable carriers can be large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acid, polyglycolic acid and the like. These vectors are well known to those of ordinary skill in the art. A full discussion of pharmaceutically acceptable carriers or excipients can be found in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. 1991).
  • the pharmaceutically acceptable carrier in the composition may include liquids such as water, saline, glycerol and ethanol.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like may also be present in these carriers.
  • the composition can be formulated as an injectable, such as a liquid solution or suspension; it can also be formulated to be suitable for dissolution prior to injection. Solid form of liquid or suspension, liquid excipient. Liposomes are also included in the definition of pharmaceutically acceptable carriers.
  • the vaccine compositions of the invention may be prophylactic or therapeutic.
  • the vaccine composition comprises an immunogenic antigen (autologous tumor vaccine of the invention) and is typically combined with a "pharmaceutically acceptable carrier" which includes antibodies which do not themselves induce the production of antibodies harmful to the individual receiving the composition.
  • a pharmaceutically acceptable carrier which includes antibodies which do not themselves induce the production of antibodies harmful to the individual receiving the composition.
  • Any carrier. Suitable carriers are generally large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acid, polyglycolic acid, amino acid polymers, amino acid copolymers, lipid agglutinates (e.g., oil droplets or liposomes), and the like. These vectors are well known to those of ordinary skill in the art.
  • these carriers can function as immunostimulating agents ("adjuvants").
  • the antigen can be coupled to bacterial toxoids (such as toxoids of pathogens such as diphtheria, tetanus, cholera, Helicobacter pylori) or bacterial extracts (such as endotoxin or flagellin).
  • bacterial toxoids such as toxoids of pathogens such as diphtheria, tetanus, cholera, Helicobacter pylori
  • bacterial extracts such as endotoxin or flagellin
  • Preferred adjuvants for enhancing the effect of the immunological composition include, but are not limited to: (1) aluminum salts (alum) such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, and the like; (2) oil-in-water emulsion formulations, for example, (a) MF59 (see WO 90/14837), (b) SAF, and (c) RibiTM Adjuvant System (RAS) (Ribi Immunochem, Hamilton, MT), (3) saponin adjuvant; (4 ⁇ 61111 ( 1 Complete adjuvant (?) and ?61111 ( 1 incomplete adjuvant (1?); (5) cytokines, such as interleukins (such as IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc., interferon (such as gamma interferon), macrophage colony-stimulating factor (M-CFS), mononuclear-macrophage colony-stimulating factor (GM-CFS
  • Vaccine compositions including immunogenic compositions (e.g., can include antigens, pharmaceutically acceptable carriers, and adjuvants), typically contain a diluent such as water, saline, glycerol, ethanol, and the like.
  • a diluent such as water, saline, glycerol, ethanol, and the like.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like may be present in such carriers.
  • vaccines including immunogenic compositions, comprise an immunologically effective amount of an immunogenic polypeptide, as well as other desired components as described above.
  • immunologically effective amount means that the amount administered to a subject in a single dose or in a continuous dose is effective for treatment or prevention. The amount may be based on the health and physiological condition of the individual being treated, the type of individual being treated (eg, human), the ability of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation of the vaccine, the assessment of the medical condition by the treating physician, And other relevant factors. This amount is expected to be in a relatively wide range and can be determined by routine experimentation.
  • the vaccine composition or immunogenic composition can be formulated as an injectable preparation, such as a liquid solution or suspension; it can also be formulated as a solid form suitable for solution or suspension, liquid excipient prior to injection.
  • the formulation may also be emulsified or encapsulated in liposomes to enhance the adjuvant effect.
  • the composition can be administered directly to a subject.
  • the subject can be a human or a non-human mammal, preferably a human.
  • When used as a vaccine it can be directly administered to an individual by a known method.
  • These compositions are usually administered by the same route of administration as conventional vaccines.
  • Routes for administering a pharmaceutical composition or vaccine composition of the invention include, but are not limited to: Formulated pharmaceutical compositions can be administered by conventional routes, including but not limited to: such as intradermal single/multiple Point injection, subcutaneous single/multiple injection, intra- or peripheral lymphatic injection, intramuscular injection, intrathoracic/peritoneal injection, intravenous injection, peritumoral/intratumoral injection.
  • routes including but not limited to: such as intradermal single/multiple Point injection, subcutaneous single/multiple injection, intra- or peripheral lymphatic injection, intramuscular injection, intrathoracic/peritoneal injection, intravenous injection, peritumoral/intratumoral injection.
  • the above approaches may be applied separately depending on the specific situation, and may be applied in combination if necessary.
  • the patient's own site may be, but is not limited to, the primary tumor, metastases or surrounding lymphatic drainage areas and distal or proximal extremities, thoracodorsal, abdomen, neck, locally accessible lymph nodes or lymphatic drainage areas
  • dose control should be considered within a safe and effective range.
  • the specific dose should also take into account the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
  • the vaccine of the present invention may also be used in combination with other drugs or physical therapies, including but not limited to: various cytokines such as IFN, TNF, IL-2, GM-CSF, G-CSF, SCF, And electromagnetic waves, etc.
  • various cytokines such as IFN, TNF, IL-2, GM-CSF, G-CSF, SCF, And electromagnetic waves, etc.
  • the main advantages of the present invention are as follows - (1) The present invention actively stimulates tumor antigen expression and antigen processing presentation function by irradiation and/or heating, and then inactivates, supplements with adjuvant, and modulates immunotherapy according to individual immune response. Program, to achieve the purpose of truly individualized tumor immunotherapy;
  • the present invention can promote immunity in the body, especially for cellular immunity and humoral immunity of tumors;
  • the technical method for treating or preventing tumors according to the present invention is suitable for use in the medical field, particularly for preventing or treating tumors, and is also suitable for postoperative tumor residuals and patients with micrometastases, and provides a field in the art.
  • the new choice has broad application prospects.
  • the invention is further illustrated below in conjunction with specific embodiments. It should be understood that these embodiments are for illustrative purposes only. The invention is not intended to limit the scope of the invention.
  • the experimental methods in the following examples which do not specify the specific conditions are usually carried out according to the conditions described in conventional conditions such as Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturer. The suggested conditions.
  • Example 1 Example 1
  • the tumor vaccine prepared by the method of the present invention has strong antigen presentation ability and ability to elicit an antibody immune response.
  • the specific results are as follows:
  • mice Conventional Balb/c mice were immunized 3 weeks after immunization with the tumor cell antigen prepared in Example 1, and the plasma was diluted and added to the ELISA plate coated with tumor cells to anti-mouse IgG-HRP and TMB substrates. The antibody titer is shown (this experiment is 1:100000).
  • mice were immunized with the tumor cell antigen prepared in Example 1 for 3 weeks, and lymphocytes (LN) and spleen cells (SP) were taken and the constructed Luciferase-containing 4T1 tumor cells (4T1-luc) were 10:1.
  • LN lymphocytes
  • SP spleen cells
  • Fig. 2A is an antitumor T in the adjuvant group.
  • Cell killing experiment, Fig. 2 is the tumor cell antigen anti-tumor cell killing experiment prepared by the method group of Example 1, Rlu refers to the activity of Luciferase; NC refers to the normal control group; LN refers to lymph node cells; SP refers to spleen cells.
  • Example 3
  • mice anti-tumor immunity test is divided into two steps:
  • Tumor immunosensitization The treated tumor antigen novel tumor vaccine was first inoculated subcutaneously in the test mice, and 14 days later, the immunization was boosted once again, and the homologous mouse tumor cells were inoculated 7 days later (Fig. 3).
  • a. Inhibition of carcinoma in situ BALB/c mice and 4T1 breast cancer cell lines were used; b. Inhibition of metastatic tumors: C57BL/6 mice and Lewis lung cancer cell lines were used. Lewis and 4T1 cancer cells are highly malignant, grow fast, are difficult to control, and can form lung metastases.
  • a, living tumor fluorescence quantitative method the above 4T1 cells with Luciferase gene, so 4T1-Luc tumor size can be perfused into the anesthesia tumor-bearing mice peritoneal injection of Luciferin substrate, 5 minutes after the determination of fluorescence intensity; b, vernier caliper to determine the tumor diameter; Both methods can objectively reflect tumor size.
  • Anti-pulmonary metastasis effect Due to the rapid growth of experimental Lewis lung cancer, the cancer nodules are rapidly fused into a piece, and the number of nodules cannot be calculated. Therefore, the results are calculated by whole lung weighing.
  • mice immunized with the tumor antigen prepared in Example 1 were subjected to intravenous or subcutaneous injection of live tumor cells to form metastatic cancer and carcinoma in situ for the antitumor effect.
  • the tumor-injected Balb/c mouse tumor fluorescence imaging showed that compared with the control group (adjuvant immunization) and the original method of preparation of tumor vaccine (adjuvant + inactivated tumor cells) group,
  • the vaccine group prepared in Example 1 had a small number of tumors and a small volume.
  • the tumor growth curve (Fig. 5) showed that the growth rate of the in situ carcinoma of the new tumor vaccine group was significantly slower than that of the original tumor vaccine preparation group, the adjuvant group alone, and the saline group (p ⁇ 0.01). Similar results were obtained (Fig. 6, Fig. 7). It is worth noting that the fastest growing tumor of 4T1-Luc cells could not be inhibited by Taxol 5 mg/kg (daily injection) but was inhibited by the vaccine prepared by the method of the present invention.
  • Metastasis is the main cause of tumor death.
  • experimental lung metastasis of Lewis lung cancer cells on homologous C57BL/6 mice was used as a detection model.
  • A saline control group, subcutaneous injection of simulated saline immunization process
  • Taxol group (Paclitaxel group): Conventional chemotherapy drugs, as a positive control group.
  • mice Four groups of mice were injected with lx lO 5 live Lewis lung cancer cells 21 days after receiving different immunotherapy. After 23 days, the lung tumor weight of the mice was weighed to obtain the mean and standard deviation.

Abstract

Provided are a method for preparing an autologous tumor vaccine and uses thereof, specifically comprising: radiating and/or heating autologous tumor cells to obtain the tumor cells having enhanced antigenicity and antigen presentation, cultivating the obtained tumor cells, deactivating the cultivated tumor cells, and adding adjuvant to prepare the anti-tumor vaccine.

Description

一种自体肿瘤疫苗的制备方法及其应用  Preparation method of autologous tumor vaccine and application thereof
技术领域  Technical field
本发明属于生物技术和免疫学领域, 具体地, 本发明涉及一种自体肿瘤疫苗 的制备方法及其应用。 背景技术  The present invention is in the field of biotechnology and immunology, and in particular, the present invention relates to a method for preparing an autologous tumor vaccine and an application thereof. Background technique
肿瘤是目前引起人类的主要死亡原因之一。 尽管近年来在肿瘤的诊断和治疗 方面的水平不断得到改进和提高, 化疗和放疗的方案也在不断地改进, 但最终大 部分病人仍难以逃脱死亡的厄运。  Tumors are one of the leading causes of death in humans. Although the level of diagnosis and treatment of tumors has been continuously improved and improved in recent years, the programs of chemotherapy and radiotherapy are constantly improving, but in the end, most patients still have difficulty to escape the fate of death.
在恶性肿瘤不同的治疗模式中, 免疫治疗因为其特异性的杀伤肿瘤细胞, 具 有高效率和低毒副作用的特点, 越来越受到关注。 近年来, 分子生物学的进展和 对免疫系统功能的进一步的了解使得的研制和开发生物治疗方法进入了一个飞速 发展的阶段。 肿瘤疫苗的研制是目前肿瘤生物治疗的最主要的方向。  In different treatment modes of malignant tumors, immunotherapy has attracted more and more attention because of its specific killing of tumor cells, high efficiency and low toxic side effects. In recent years, advances in molecular biology and further understanding of the function of the immune system have led to the development and development of biotherapeutics that has entered a stage of rapid development. The development of tumor vaccine is the most important direction of tumor biotherapy.
然而由于恶性肿瘤具有多种免疫逃逸机制, 主要包括肿瘤细胞抗原含量低, 肿瘤抗原的破坏或不断变异, 或者肿瘤抗原的处理提呈差, 或者缺乏一些免疫辅 助因子的表达, 或者机体免疫功能低下等, 使得现有肿瘤瘤苗的临床治疗效果不 佳。 由于恶性肿瘤异质性, 多样性, 多变性使不同个体在患同种肿瘤时, 其个体 肿瘤抗原极不相同。 因而, 肿瘤免疫治疗应为个体化, 以诱导个体特异性的杀伤 自体肿瘤细胞的主动免疫。  However, malignant tumors have multiple immune escape mechanisms, including low antigen content in tumor cells, destruction or constant mutation of tumor antigens, poor treatment of tumor antigens, or lack of expression of some immune cofactors, or low immune function. Etc., the clinical treatment effect of existing tumor tumor vaccines is not good. Because of the heterogeneity, diversity, and variability of malignant tumors, the individual tumor antigens of different individuals are very different when they are suffering from the same kind of tumor. Thus, tumor immunotherapy should be individualized to induce active immunity of individual-specific killing autologous tumor cells.
目前全细胞作为肿瘤疫苗, 可以提供那些未知的肿瘤抗原来激活免疫系统肿 瘤抗原。 小鼠肿瘤模型中, 通常使用灭活的肿瘤细胞免疫小鼠, 以保护小鼠免受 接种的肿瘤的侵袭。但当肿瘤细胞疫苗使用的时间推迟到接种肿瘤细胞后一周时, 疫苗就失去了保护小鼠的能力。 导致目前肿瘤细胞疫苗的临床治疗反应比较差, 仅仅适用于无特殊肿瘤抗原的肿瘤病人的预防复发。 对于进展期病人在临床研究 方面很少获得良好效果。  Whole cells are now used as tumor vaccines to provide unknown tumor antigens to activate immune system tumor antigens. In a mouse tumor model, mice are usually immunized with inactivated tumor cells to protect the mice from inoculation of the inoculated tumor. However, when the time of use of the tumor cell vaccine is delayed until one week after inoculation of the tumor cells, the vaccine loses the ability to protect the mouse. The current clinical response to tumor cell vaccines is relatively poor, and it is only suitable for preventing recurrence of tumor patients without special tumor antigens. Very few good results have been obtained in clinical studies for advanced patients.
理想的肿瘤-特异的抗原应具有免疫原性,为肿瘤细胞所表达但不表达于正常 的细胞。 不幸的是, 大多数肿瘤抗原都没有足够的免疫原性以诱导有效的免疫反 应。  An ideal tumor-specific antigen should be immunogenic, expressed by tumor cells but not expressed in normal cells. Unfortunately, most tumor antigens do not have sufficient immunogenicity to induce an effective immune response.
目前尚缺乏具有良好的免疫原性和特异性自体肿瘤疫苗及其制备方法, 本领 域迫切需要开发相应方法和疫苗。 发明内容 At present, there is still a lack of good immunogenicity and specific autologous tumor vaccine and preparation method thereof. The field urgently needs to develop methods and vaccines. Summary of the invention
本发明的目的就是提供一种自体肿瘤疫苗的制备方法。  It is an object of the present invention to provide a method of preparing an autologous tumor vaccine.
本发明的另一目的提供一种自体肿瘤疫苗及其应用。 在本发明的第一方面, 提供了一种制备自体肿瘤疫苗的方法, 包括步骤: Another object of the present invention is to provide an autologous tumor vaccine and use thereof. In a first aspect of the invention, a method of preparing an autologous tumor vaccine is provided, comprising the steps of:
(1) 获得分离的自体肿瘤细胞; (1) obtaining isolated autologous tumor cells;
(2) 用放射法和 /或加热法处理步骤 (1)的自体肿瘤细胞, 获得抗原性和抗原提 呈性增强的肿瘤细胞;  (2) treating the autologous tumor cells of step (1) by radiotherapy and/or heating to obtain tumor cells having enhanced antigenicity and antigen-promoting properties;
(3) 对步骤 (2)获得的肿瘤细胞进行灭活处理, 获得灭活的肿瘤细胞;  (3) inactivating the tumor cells obtained in the step (2) to obtain inactivated tumor cells;
(4) 将步骤 (3)获得的灭活的肿瘤细胞制备为自体肿瘤疫苗。  (4) The inactivated tumor cells obtained in the step (3) were prepared as an autologous tumor vaccine.
在另一优选例中, 所述的肿瘤为: 实体瘤或非实体瘤。  In another preferred embodiment, the tumor is: a solid tumor or a non-solid tumor.
在另一优选例中, 所述的实体瘤来源于: 脑、 肺、 头颈部、 皮肤、 胰、 胃肠 道、 膀胱、 生殖道、 脊髓、 脾、 肾、 肝、 四肢、 骨骼、 舌、 或喉。  In another preferred embodiment, the solid tumor is derived from: brain, lung, head and neck, skin, pancreas, gastrointestinal tract, bladder, genital tract, spinal cord, spleen, kidney, liver, limbs, bones, tongue, Or throat.
在另一优选例中, 所述的非实体瘤来源于血液或骨髓。  In another preferred embodiment, the non-solid tumor is derived from blood or bone marrow.
在另一优选例中, 所述的肿瘤为原发性肿瘤或继发转移性肿瘤。  In another preferred embodiment, the tumor is a primary tumor or a secondary metastatic tumor.
在另一优选例中, 所述的肿瘤来源于非人哺乳动物组织或人的组织。  In another preferred embodiment, the tumor is derived from a tissue of a non-human mammalian tissue or a human.
在另一优选例中, 步骤 (1)所述获得分离的自体肿瘤细胞的方法选自下组: 酶消化法、 机械剪切法、 或人工磨碎肿瘤组织法将肿瘤组织处理为可培养的 分离的自体肿瘤细胞。  In another preferred embodiment, the method for obtaining the isolated autologous tumor cells according to the step (1) is selected from the group consisting of: enzymatic digestion, mechanical shearing, or artificial grinding of tumor tissue to treat tumor tissue to be culturable. Isolated autologous tumor cells.
在另一优选例中, 步骤 (2)所述的放射法选自下组: 用选自 α, β、 γ、 质子、 或 重离子放射线、 以任选自 0.1-50 Gy剂量范围、 任选自 0.1-10 Gy/min剂量率, 处理 步骤 (1)的自体肿瘤细胞, 获得抗原性和抗原提呈性增强的肿瘤细胞。  In another preferred embodiment, the radiation method described in the step (2) is selected from the group consisting of: selected from α, β, γ, proton, or heavy ion radiation, optionally selected from the range of 0.1-50 Gy, optionally The autologous tumor cells of step (1) were treated at a dose rate of 0.1-10 Gy/min to obtain antigenic and antigen-presenting tumor cells.
在另一优选例中, 放射线来源于天然的或人工的放射源。  In another preferred embodiment, the radiation is derived from a natural or artificial source of radiation.
在另一优选例中, 步骤 (2) 所述的加热法选自下组: 加热温度任选自  In another preferred embodiment, the heating method described in the step (2) is selected from the group consisting of: heating temperature is selected from the group consisting of
37.5 °C -50°C (优选 38 °C -45 °C), 加热时间任选自 1 min-2小时 (优选 30min-l小时)。 37.5 ° C -50 ° C (preferably 38 ° C -45 ° C), the heating time is selected from 1 min to 2 hours (preferably 30 min - l hour).
在另一优选例中, 在步骤 (2)和步骤 (3)之间, 还包括步骤: 对步骤 (2)获得抗原 性和抗原提呈性增强的肿瘤细胞进行培养处理。 在另一优选例中, 培养处理时间选自: 6小时到 2星期。 In another preferred embodiment, between the step (2) and the step (3), the method further comprises the step of: culturing the tumor cells obtained by the step (2) to obtain antigenicity and antigen-promoting enhancement. In another preferred embodiment, the culture treatment time is selected from the group consisting of: 6 hours to 2 weeks.
在另一优选例中, 在培养处理中, 使用基本细胞培养液, 和 /或加入生长因子 或小分子化合物。  In another preferred embodiment, in the culture treatment, a basic cell culture solution is used, and/or a growth factor or a small molecule compound is added.
在另一优选例中, 步骤 (3)所述的灭活处理选自下组: 微波细胞灭活法、 电磁波 细胞灭活法, 激光细胞灭活法、 高温细胞灭活法、 超声破碎细胞灭活法、 反复细胞冻 融、 匀浆、 离心、 沉淀细胞灭活法、 酶学消化细胞灭活法、 低渗破裂法、 或其组合。  In another preferred embodiment, the inactivation treatment described in the step (3) is selected from the group consisting of microwave cell inactivation, electromagnetic wave cell inactivation, laser cell inactivation, high temperature cell inactivation, sonication and cell disruption. Live method, repeated cell freeze-thaw, homogenate, centrifugation, pellet cell inactivation, enzymatic digestion cell inactivation, hypotonic rupture, or a combination thereof.
在另一优选例中, 在步骤 (4)中, 将步骤 (3)获得的灭活的肿瘤细胞与佐剂混合, 获得自体肿瘤疫苗。  In another preferred embodiment, in step (4), the inactivated tumor cells obtained in the step (3) are mixed with an adjuvant to obtain an autologous tumor vaccine.
在另一优选例中, 所述的佐剂为福氏完全佐剂或不完全佐剂。  In another preferred embodiment, the adjuvant is Freund's complete adjuvant or incomplete adjuvant.
在另一优选例中,所述的佐剂选自下组:脂质体、细胞因子 (如 GM-CSF、 G-CSF、 In another preferred embodiment, the adjuvant is selected from the group consisting of liposomes, cytokines (eg, GM-CSF, G-CSF,
CSF、 IFN、 IL-2等), 各种植物蛋白、 DNA、 减活或灭活病毒、 细菌、 真菌、 微生物 及其提取物、 或其组合。 CSF, IFN, IL-2, etc.), various plant proteins, DNA, deactivated or inactivated viruses, bacteria, fungi, microorganisms and their extracts, or combinations thereof.
在本发明的第二方面,提供了一种自体肿瘤疫苗,所述疫苗是用第一方面所述的 方法制备的。  In a second aspect of the invention, there is provided an autologous tumor vaccine prepared by the method of the first aspect.
在另一优选例中, 所述的疫苗还包括佐剂。  In another preferred embodiment, the vaccine further comprises an adjuvant.
在本发明的第三方面,提供了第二方面所述自体肿瘤疫苗的用途,它被用于制备 预防或治疗肿瘤的组合物。  In a third aspect of the invention, there is provided the use of the autologous tumor vaccine of the second aspect, which is used for the preparation of a composition for preventing or treating a tumor.
在另一优选例中, 所述的组合物为药物组合物或疫苗组合物。  In another preferred embodiment, the composition is a pharmaceutical composition or a vaccine composition.
在另一优选例中, 所述的组合物是注射剂形式。 较佳地, 所述的注射剂通过选自 下组的途径进行给药: 皮内单点 /多点注射、 皮下单点 /多点注射、 淋巴结内或周围 注射、 肌肉注射、 胸腔 /腹腔注射、 静脉注射、 瘤周正常组织注射。 优选为皮内肢 体远端。  In another preferred embodiment, the composition is in the form of an injection. Preferably, the injection is administered by a route selected from the group consisting of intradermal single/multiple injection, subcutaneous single/multiple injection, intralymphatic or peripheral injection, intramuscular injection, intrathoracic/peritoneal injection, Intravenous, peritumoral normal tissue injection. Preferably, it is the distal end of the intradermal limb.
在另一优选例中, 所述的注射剂的注入部位选自下组: 原发灶、 转移灶、 原发 灶周围淋巴引流区、 转移灶周围淋巴引流区、 四肢远端、 四肢近端、 胸背部、 腹 部、 颈部、 局部可触及的淋巴结或淋巴引流区。  In another preferred embodiment, the injection site of the injection is selected from the group consisting of: primary tumor, metastases, lymphatic drainage area around the primary tumor, lymphatic drainage area around the metastasis, distal extremities, proximal extremities, chest Back, abdomen, neck, locally accessible lymph nodes or lymphatic drainage areas.
在另一优选例中, 所述疫苗可与选自下组的药物及物理疗法联用: 细胞因子、 电磁波疗法, 或其组合。 较佳地, 所述的药物选自下组: IFN、 TNF、 IL-2, 或其组 合。  In another preferred embodiment, the vaccine can be used in combination with a drug selected from the group consisting of cytokines, electromagnetic wave therapy, or a combination thereof. Preferably, the drug is selected from the group consisting of IFN, TNF, IL-2, or a combination thereof.
在另一优选例中, 步骤 (3)所述的灭活处理选自下组: 微波细胞灭活法、 电磁波 细胞灭活法, 激光细胞灭活法、 高温细胞灭活法、 超声破碎细胞灭活法、 反复细胞冻 融、 匀浆、 离心、 沉淀细胞灭活法、 酶学消化细胞灭活法、 低渗破裂法、 化学共价交 联法、 化学性变性法, 或其组合。 In another preferred embodiment, the inactivation treatment described in the step (3) is selected from the group consisting of microwave cell inactivation, electromagnetic wave cell inactivation, laser cell inactivation, high temperature cell inactivation, sonication and cell disruption. Live method, repeated cell freeze-thaw, homogenate, centrifugation, pellet cell inactivation, enzymatic digestion, cell inactivation, hypotonic rupture, chemical covalent Combined method, chemical denaturing method, or a combination thereof.
本发明的第四方面, 提供了一种预防或治疗肿瘤的方法, 所述方法包括对对象施 用有效量的本发明第二方面所述的肿瘤疫苗。  In a fourth aspect of the invention, a method of preventing or treating a tumor comprising administering an effective amount of the tumor vaccine of the second aspect of the invention to a subject is provided.
在另一优选例中, 所述施用时间选自下组: 术后、化疗前, 或 WBC回升至 >3000 个 /ul时。  In another preferred embodiment, the administration time is selected from the group consisting of: postoperative, pre-chemotherapy, or WBC rebound to >3000 per ul.
在另一优选例中, 所述的施用次数可为 4个疗程 (每疗程 3-4周)至终生。  In another preferred embodiment, the number of administrations may be 4 courses (3-4 weeks per course) to life.
在另一优选例中, 所述方法还包括: 用免疫学实验来监测免疫反应程度。  In another preferred embodiment, the method further comprises: monitoring the extent of the immune response using an immunological assay.
在另一优选例中, 所述的免疫学实验选自下组: 细胞免疫反应、 体液免疫反 应、 免疫调节反应, 或其组合。  In another preferred embodiment, the immunological assay is selected from the group consisting of a cellular immune response, a humoral immune response, an immunomodulatory response, or a combination thereof.
在另一优选例中, 所述的细胞免疫反应检测方法包括:  In another preferred embodiment, the method for detecting cellular immune response comprises:
1、皮内注射个体化瘤苗, 其抗瘤迟发性过敏反应可见为皮丘 0.5 cm以上者 为免疫反应良好, <0.5cm为免疫反应差;  1. Intradermal injection of individualized tumor vaccine, the anti-tumor delayed allergic reaction can be seen as a cumulus 0.5 cm or more for a good immune response, <0.5 cm for poor immune response;
2、 淋巴细胞转化反应: 将免疫后一个月的患者的淋巴细胞分离出来, 加入本 发明第二方面所述的疫苗, 观察其转化率 (优选为用 BrdU法、 MTT法), 转化 率高则免疫反应好;  2. Lymphocyte transformation reaction: The lymphocytes of the patient one month after immunization are separated, and the vaccine of the second aspect of the invention is added to observe the conversion rate (preferably by BrdU method, MTT method), and the conversion rate is high. Good immune response;
3、 检测 T/B细胞免疫反应后分泌的淋巴因子;  3. Detection of lymphokines secreted by T/B cell immune response;
较佳地, 所述的淋巴因子选自下组: IL-2、 IL-4、 IL-6、 IL-10、 IL-12、 IL-17、 IL-18、 TNF-a、 IFN-γ, 或其组合;  Preferably, the lymphokine is selected from the group consisting of IL-2, IL-4, IL-6, IL-10, IL-12, IL-17, IL-18, TNF-a, IFN-γ, Or a combination thereof;
较佳地, 所述的检测方法选自下组: ELISPOT, ELISA, FCM, 或其组合。  Preferably, the detection method is selected from the group consisting of ELISPOT, ELISA, FCM, or a combination thereof.
4、 检测免疫细胞亚群及表面杀伤性标记;  4. Detection of immune cell subpopulations and surface killing markers;
较佳地, 所述的免疫细胞亚群选自下组: CD4+、 CD8+、 CDl lb+、 Grl+, 或 其组合;  Preferably, the subset of immune cells is selected from the group consisting of CD4+, CD8+, CD1 lb+, Grl+, or a combination thereof;
较佳地, 所述的杀伤性激活性标记选自下组: Fas L、 LIGHT, CD25、 TNF-a、 IFN-γ, perforin或其组合。  Preferably, the killer activation marker is selected from the group consisting of Fas L, LIGHT, CD25, TNF-a, IFN-γ, perforin or a combination thereof.
在另一优选例中, 用流式细胞术检测各亚群。  In another preferred embodiment, each subpopulation is detected by flow cytometry.
在另一优选例中, 所述的体液免疫检测方法包括:  In another preferred embodiment, the humoral immunoassay method comprises:
1、抗自身肿瘤抗体的检测: 以所制备的自身瘤苗为抗原包被加入自身免疫前 后的血浆, 比较免疫前后抗自身肿瘤抗原的抗体效价, 效价越高则免疫反应性 越强。 在另一优选例中, 所述自身抗原的分子量用 WESTERN BLOT方法测定。 在另一优选例中, 所述的免疫调节反应检测方法包括: 1. Detection of anti-autoantigen antibody: The prepared tumor vaccine is used as an antigen coating to add plasma before and after autoimmunization, and the antibody titer against autoantitumor antigen is compared before and after immunization. The higher the titer, the stronger the immunoreactivity. In another preferred embodiment, the molecular weight of the autoantigen is determined by the WESTERN BLOT method. In another preferred embodiment, the immunomodulatory reaction detecting method comprises:
1、 检测免疫调节细胞亚群; 较佳地, 所述的亚群包括: 调节性 T 细胞 Treg:CD4+CD25+、 髓系来源的抑制性细胞 MDSC: CD14— CDllb+、 肿瘤相关巨 噬细胞 TAM: Ml/M2、 调节性树突状细胞 DCreg:CDllb+/Gr-l+, 或其组合。 较佳地, 所述的检测方法为流式细胞术。 1. Detection of a subset of immunoregulatory cells; preferably, said subpopulation comprises: regulatory T cells: Treg: CD4 + CD25 + , myeloid-derived suppressor cells MDSC: CD14 - CD11b + , tumor-associated macrophages TAM: Ml/M2, regulatory dendritic cell DCreg: CD11b+/Gr-l+, or a combination thereof. Preferably, the detection method is flow cytometry.
2、测定血浆中免疫调节因子的变化,较佳地,所述的免疫调节因子包括: IL-2、 IL-4、 IL-6、 IL-10、 IL-12、 IL-17、 IL-18、 TNF-a、 IFN-γ, 或其组合; 所述的 测定方法包括: ELISA、 LUMINEX bead-array assay, 或其组合。  2. Determination of changes in immunoregulatory factors in plasma. Preferably, the immunoregulatory factors include: IL-2, IL-4, IL-6, IL-10, IL-12, IL-17, IL-18 , TNF-a, IFN-γ, or a combination thereof; the assay comprises: ELISA, LUMINEX bead-array assay, or a combination thereof.
在另一优选例中, 所述的免疫学实验在首次免疫后 15-60天内进行。  In another preferred embodiment, the immunological assay is performed within 15-60 days after the first immunization.
在另一优选例中, 所述的方法还包括: 根据免疫学实验测得的免疫反应程度, 判定治疗效果和 /或对治疗方案进行调整。  In another preferred embodiment, the method further comprises: determining the therapeutic effect and/or adjusting the treatment regimen based on the degree of immune response measured by the immunological experiment.
在另一优选例中, 所述的调整治疗方案指调整选自下组的一个或多个治疗参 数: 免疫佐剂结合、 免疫剂量、 免疫部位、 免疫时间、 免疫次数, 低剂量放射、 其它物理或生物方法刺激免疫器官。  In another preferred embodiment, the adjusted treatment regimen refers to adjusting one or more therapeutic parameters selected from the group consisting of: immunoadjuvant binding, immunization dose, immunization site, immunization time, number of immunizations, low dose radiation, other physics Or biological methods to stimulate immune organs.
在另一优选例中, 所述的免疫器官是骨髓。  In another preferred embodiment, the immune organ is bone marrow.
在另一优选例中, 所述方法包括:  In another preferred embodiment, the method includes:
(i) 术后第三天, 对患者施用所述疫苗;  (i) administering the vaccine to the patient on the third postoperative day;
(ii) 术后 7-14天, 再次对患者施用所述疫苗;  (ii) administering the vaccine to the patient again 7-14 days after surgery;
(iii) 术后 14-24天, 再次对患者施用所述疫苗;  (iii) administering the vaccine to the patient again 14-24 days after surgery;
(iv) 术后第 31-34天用免疫学实验来监测免疫反应程度, 并通过检测结果调 节免疫治疗方案。  (iv) Immunological tests were used to monitor the degree of immune response on days 31-34 after surgery, and the immunotherapy protocol was adjusted by the test results.
在另一优选例中, 所述的施用所述疫苗及其效果观察时间可因个体差异而灵 活变动。 应理解, 在本发明范围内中, 本发明的上述各技术特征和在下文 (如实施例) 中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。 限于篇幅, 在此不再一一累述。 附图说明  In another preferred embodiment, the administration of the vaccine and its effect observation time may vary flexibly due to individual differences. It is to be understood that within the scope of the present invention, the various technical features of the present invention and the technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here. DRAWINGS
下列附图用于说明本发明的具体实施方案, 而不用于限定由权利要求书所界 定的本发明范围。 The following figures are used to illustrate specific embodiments of the invention and are not intended to be limited by the scope of the claims The scope of the invention is intended.
图 1显示注射自身肿瘤疫苗后 3周抗肿瘤抗体 IgG的测定结果。  Figure 1 shows the results of anti-tumor antibody IgG assay 3 weeks after injection of the auto-tumor vaccine.
图 2显示抗肿瘤 T细胞杀伤实验。  Figure 2 shows an anti-tumor T cell killing experiment.
图 3显示实验分组以及操作流程。  Figure 3 shows the experimental grouping and the operational flow.
图 4显示抗肿瘤效应荧光实时动态测定 (Live Image)结果。  Figure 4 shows the results of real-time dynamic measurement of anti-tumor effect fluorescence (Live Image).
图 5显示本发明实施例 1的方法制备的疫苗抗原位癌 (4T1-Luc)效应 (试验一) 结果。  Fig. 5 shows the results of the vaccine antigenic cancer (4T1-Luc) effect (test 1) prepared by the method of Example 1 of the present invention.
图 6显示本发明实施例 1的方法制备的疫苗抗原位癌 (4T1-Luc)效应 (试验二) 结果。  Fig. 6 shows the results of the vaccine antigenic cancer (4T1-Luc) effect (test 2) prepared by the method of Example 1 of the present invention.
图 7显示本发明实施例 1的方法制备的疫苗与 Taxol抗癌效应的比较 (试验三, 与化疗药 Taxol比较)结果。  Figure 7 is a graph showing the results of a comparison between the vaccine prepared by the method of Example 1 of the present invention and the anticancer effect of Taxol (Trial 3, compared with the chemotherapy drug Taxol).
图 8显示本发明一个实施例的方法对实验性肺转移癌的杀伤抑制作用。  Figure 8 shows the killing inhibition effect of the method of one embodiment of the present invention on experimental lung metastasis.
图 9显示本发明一个实施例制备自体肿瘤疫苗的流程。  Figure 9 shows the flow of an autologous tumor vaccine prepared in accordance with one embodiment of the present invention.
图 10显示了肿瘤细胞照射仪; 其中, 利用 Y射线断裂 DNA 并导致 DNA错 误修复, 同时使促恶劣环境下生长的癌基因高度表达的特性, 将肿瘤细胞用此专 属发明的 Y照射仪处理, 使其癌基因高度表达, 肿瘤抗原激活提呈加强, 以增加 本法疗效。  Fig. 10 shows a tumor cell illuminator; wherein, the Y-ray is used to break the DNA and cause DNA error repair, and at the same time, the characteristics of the oncogenic gene which promotes growth in a harsh environment are highly expressed, and the tumor cells are treated with the Y illuminator of the exclusive invention. The oncogene is highly expressed, and the activation of tumor antigen is enhanced to increase the efficacy of the method.
图 11显示了骨髓生长刺激仪; 其中, 利用中 /高频电磁波有刺激细胞生长的 特性, 将此骨髓生长刺激仪置于无肿瘤细胞的髂骨部, 以刺激造血干细胞增殖分 化, 使自体抗癌免疫细胞数增加, 以增加本法疗效。 具体实施方式  Figure 11 shows a bone marrow growth stimulator; wherein, using medium/high frequency electromagnetic waves to stimulate cell growth, the bone marrow growth stimulator is placed in the humerus of tumor-free cells to stimulate hematopoietic stem cell proliferation and differentiation, so that autoantibodies The number of cancer immune cells is increased to increase the efficacy of this method. detailed description
本发明人经过广泛而深入的研究, 首次意外地发现, 对自体肿瘤细胞进行放 射和 /或加热的处理, 可以大大激发自体肿瘤细胞的抗原性, 热休克蛋白增强抗原 提呈性; 再进行肿瘤细胞灭活, 辅以佐剂, 制成疫苗。 所述疫苗具有被提呈性强, 肿瘤免疫原性强的特殊优点, 能强烈激发肿瘤患者对自体肿瘤的体液及细胞免疫 反应, 以达到提高抗原发性及转移性肿瘤效应, 延长肿瘤患者存活时间的目的。 在此基础上完成了对新型自体肿瘤疫苗发明。 其应用亦根据监测患者对自体肿瘤 疫苗反应状况而改变疫苗用量, 注射部位及时间, 疗程长短及间隔, 真正做到个 体化主动免疫治疗。 制备方法 After extensive and intensive research, the present inventors have discovered for the first time that the treatment of radiation and/or heating of autologous tumor cells can greatly stimulate the antigenicity of autologous tumor cells, and heat shock proteins enhance antigen presentation; The cells are inactivated and supplemented with adjuvant to make a vaccine. The vaccine has the special advantages of strong presentation and strong immunogenicity of the tumor, and can strongly stimulate the humoral and cellular immune responses of the tumor patients to the autologous tumor, so as to improve the antigenic and metastatic tumor effect and prolong the survival of the tumor patient. The purpose of time. On this basis, the invention of a novel autologous tumor vaccine was completed. Its application is also based on monitoring the patient's response to autologous tumor vaccines, changing the amount of vaccine, the location and time of injection, the length and interval of treatment, and truly Active immunotherapy. Preparation
本发明提供了一种制备自体肿瘤疫苗的方法, 在一个优选例中, 包括步骤: (1) 获得分离的自体肿瘤细胞;  The present invention provides a method of preparing an autologous tumor vaccine, and in a preferred embodiment, comprising the steps of: (1) obtaining isolated autologous tumor cells;
(2) 用放射法和 /或加热法处理步骤 (1)的自体肿瘤细胞, 获得抗原性和抗原提 呈性增强的肿瘤细胞;  (2) treating the autologous tumor cells of step (1) by radiotherapy and/or heating to obtain tumor cells having enhanced antigenicity and antigen-promoting properties;
(3) 对步骤 (2)获得的肿瘤细胞进行灭活处理, 获得灭活的肿瘤细胞;  (3) inactivating the tumor cells obtained in the step (2) to obtain inactivated tumor cells;
(4) 将步骤 (3)获得的灭活的肿瘤细胞制备为自体肿瘤疫苗。  (4) The inactivated tumor cells obtained in the step (3) were prepared as an autologous tumor vaccine.
具体地, 包括如下步骤 (图 9):  Specifically, it includes the following steps (Figure 9):
通过外科手术或活检获取肿瘤组织;  Obtaining tumor tissue by surgery or biopsy;
将所得肿瘤组织按如下方法准备肿瘤细胞: 在无菌条件下取肿瘤新鲜、 血供 好的未坏死部分, 放入 DMEM或其它培养液中在 4°C下保存, 用 PBS冲洗, 通过 酶消化法或 /及机械分离法得到纯化的肿瘤细胞,将其移入培养瓶中, 37°C, 5%CO2 恒温培养箱传代培养, 达到所需细胞数后进入肿瘤全细胞瘤苗制备程序; Prepare the tumor cells as follows: Obtain the tumor-free, non-necrotic part of the tumor under sterile conditions, store in DMEM or other culture medium, store at 4 ° C, rinse with PBS, and digest by enzyme. Purified tumor cells were obtained by method or / and mechanical separation method, transferred into a culture flask, subcultured at 37 ° C, 5% CO 2 incubator, and reached the required number of cells to enter the tumor whole cell tumor preparation procedure;
通过放射和 /或加热的方法处理培养中的肿瘤细胞使其抗原激发,利用增加的 热休克蛋白增强抗原提呈性。  The tumor cells in culture are treated by radiation and/or heating to excite the antigen, and the increased heat shock protein enhances antigen presentation.
进一步的, 处理培养后的肿瘤细胞采用下述方法进行灭活:微波细胞灭活法、 电磁波细胞灭活法, 激光细胞灭活法, 高温细胞灭活法, 超声破碎细胞灭活法, 反复细胞冻融、 匀浆、 离心、 沉淀细胞灭活法, 酶学消化细胞灭活法、 低渗破裂 等方法中至少一种;  Further, the treated tumor cells are inactivated by the following methods: microwave cell inactivation method, electromagnetic wave cell inactivation method, laser cell inactivation method, high temperature cell inactivation method, ultrasonic disruption cell inactivation method, repeated cells At least one of freezing and thawing, homogenization, centrifugation, sedimentation cell inactivation, enzymatic digestion, cell inactivation, hypotonic rupture, and the like;
灭活后, 即得肿瘤全细胞疫苗。  After inactivation, a tumor whole cell vaccine is obtained.
本发明辅以上述肿瘤全细胞疫苗以佐剂, 其佐剂可为福氏完全佐剂或不完全 佐剂, 经过油包水佐剂、 脂质体、 细胞因子 (如 GM-CSF、 各种植物蛋白 DNA), 减活或灭活病毒、 细菌、 真菌、 微生物或其提取物等佐剂。 在辅以佐剂后, 能更 好的增强主动免疫反应, 减少免疫负调节, 增强抗肿瘤免疫细胞的激发及记忆。  The invention is supplemented by the above tumor whole cell vaccine with an adjuvant, and the adjuvant may be a complete Freund's adjuvant or an incomplete adjuvant, after a water-in-oil adjuvant, a liposome, a cytokine (such as GM-CSF, various Plant protein DNA), attenuating or inactivating an adjuvant such as a virus, a bacterium, a fungus, a microorganism or an extract thereof. After adjuvanting, it can better enhance the active immune response, reduce the negative regulation of immunity, and enhance the stimulation and memory of anti-tumor immune cells.
本发明方法是将肿瘤细胞经过抗原性刺激处理和增强抗原提呈能力后, 再经 过灭活处理, 使其丧失致瘤性, 并且保留了刺激后的免疫原性, 同时联合免疫佐 齐 ^, 以此作为个体化肿瘤瘤苗, 建立起个体化的治疗方法, 不断跟踪个体免疫状 态及肿瘤病情不断调整个体化免疫治疗方案, 以达最佳抗肿瘤疗效。 免疫效果监测 The method of the invention is to treat the tumor cells by antigenic stimulation treatment and enhance the antigen presenting ability, and then inactivate the treatment to lose the tumorigenicity, and retain the immunogenicity after the stimulation, and simultaneously combine the immunosupsis; As an individualized tumor vaccine, we will establish an individualized treatment method and keep track of individual immunity. State and tumor conditions continue to adjust individualized immunotherapy programs to achieve optimal anti-tumor efficacy. Immune effect monitoring
本领域的普通技术人员可以使用常规方法判断疫苗抗肿瘤免疫反应。 包括: 抗肿瘤体液免疫反应评价、 抗肿瘤细胞免疫反应评价、 抗肿瘤临床反应评价等。  One of ordinary skill in the art can determine the anti-tumor immune response of a vaccine using conventional methods. Including: anti-tumor humoral immune response evaluation, anti-tumor cell immune response evaluation, anti-tumor clinical response evaluation.
抗肿瘤体液免疫反应评价是以个体化瘤苗为抗原包被的 ELISA方法检测对 个体化瘤苗免疫前后患者自身血清中抗自身肿瘤抗体滴度, 以客观判断抗肿瘤体 液免疫反应。  The anti-tumor humoral immune response was evaluated by using an individualized tumor vaccine as an antigen-coated ELISA method to detect the anti-autoantibody antibody titer in the serum of the individual before and after immunization of the individualized tumor vaccine to objectively judge the anti-tumor humoral immune response.
抗肿瘤细胞免疫反应评价, 一般地, 包括皮内注射所制备的新型个体化瘤苗 后, 1-3天内观察患者自身皮丘大小 (迟发型个体化瘤苗免疫反应); 或自身肿瘤刺 激特异性淋巴细胞转化实验: 包括步骤: 取抗凝血, 分离淋巴细胞, 无血清培养, 加入个体化瘤苗或对照载体, 培养, 判断淋巴细胞转化性增殖水平 (BrdU掺入法 或 MTT法); 个体化瘤苗的免疫调节反应。 个体化瘤苗的免疫调节反应包括: 检 测白细胞分群 (如 CD4/CD8/CDl lb/Gr-l等)及其表面杀伤性或激活性标志 (如 FasL/LIGHT/CD25/TNF-a/IFN-y等), 根据这些体内外实验结果可以分为反应好和 反应差两类: 反应好的抗体滴度高, 皮丘大于 0.5 cm, 因此可以维持原个体化免 疫方案; 反应差的抗体滴度低, 皮丘少于 0.5 cm, 因此可以改变个体化瘤苗使用 方法及使用不同手段增强免疫反应。 免疫调节细胞亚群包括: 调节性 T细胞 Treg(CD4+CD25+)、 髓系来源的抑制性细胞 MDSC(CD14-CDl lb+)、 肿瘤相关巨 噬细胞 TAM(M1/M2)、 调节性树突状细胞 DCreg(CDl lb+/Gr-l+), 用流式细胞术 可检测各亚群。  Anti-tumor cell immune response evaluation, generally, including the new individualized tumor vaccine prepared by intradermal injection, observe the patient's own pimple size within 1-3 days (late-type individualized tumor vaccine immune response); or self-tumor stimulation specific Sexual lymphocyte transformation experiment: including steps: taking anticoagulation, separating lymphocytes, serum-free culture, adding individualized tumor vaccine or control vector, culturing, judging lymphocyte transformational proliferation level (BrdU incorporation method or MTT method); Individualized tumor vaccine immunomodulatory response. Immunomodulatory responses to individualized tumor vaccines include: detection of leukocyte populations (eg, CD4/CD8/CD1 lb/Gr-l, etc.) and their surface killing or activating markers (eg, FasL/LIGHT/CD25/TNF-a/IFN-) y, etc., according to the results of these in vitro and in vivo experiments can be divided into two types: good response and poor response: high antibody titer, pyel is greater than 0.5 cm, so can maintain the original individualized immunization program; poor response antibody titer Low, pipi is less than 0.5 cm, so you can change the use of individualized tumor vaccine and use different means to enhance the immune response. The subset of immunoregulatory cells includes: regulatory T cell Treg (CD4+CD25+), myeloid-derived suppressor cell MDSC (CD14-CD1 lb+), tumor-associated macrophage TAM (M1/M2), regulatory dendritic The cells were DCreg (CDl lb+/Gr-l+) and each subpopulation was detected by flow cytometry.
抗肿瘤临床反应: 以肿瘤影像大小评价, 根据实验结果, 病人免疫状态, 肿 瘤病情及其它抗肿瘤治疗方案的应用, 决定何时加强抗肿瘤免疫, 调整个体化治 疗方案, 继续 2、 3、 4或更多疗程。 效果监测包括但不限于影像学指标观察、 无 瘤生存时间、 总生存时间等肿瘤疗效判断标准来判断。  Anti-tumor clinical response: According to the tumor image size evaluation, according to the experimental results, the patient's immune status, tumor condition and other anti-tumor treatment programs, decide when to strengthen anti-tumor immunity, adjust individualized treatment plan, continue 2, 3, 4 Or more treatments. Efficacy monitoring includes, but is not limited to, imaging criteria, tumor-free survival time, and overall survival time.
总之,不断跟踪个体免疫状态及根据肿瘤病情不断调整个体化免疫治疗方案, 以达最佳抗肿瘤疗效。  In short, continuous tracking of individual immune status and continuous adjustment of individualized immunotherapy according to the condition of the tumor, in order to achieve the best anti-tumor efficacy.
如上所述, 个体化瘤苗的免疫反应效应可用细胞免疫、 体液免疫、 临床肿瘤 反应来监测, 监测时间可在首次免疫后约 15天 -60天, 并通过数据评价, 若反应 效果不佳时, 可重新调整个体化免疫治疗方案, 如通过调整免疫佐剂结合、 免疫 剂量、 免疫部位、 免疫时间、 免疫次数, 低剂量放射刺激骨髓等免疫器官。 组合物和给药方式 As mentioned above, the immune response effect of individualized tumor vaccine can be monitored by cellular immunity, humoral immunity, and clinical tumor response. The monitoring time can be about 15 days to 60 days after the first immunization, and the data can be evaluated if the reaction In the case of poor results, individualized immunotherapy regimens can be re-adjusted, such as by adjusting immune adjuvant binding, immunization dose, immune site, immunization time, number of immunizations, and low-dose radiation-stimulated immune organs such as bone marrow. Composition and mode of administration
本发明还提供了一种组合物, 所述的组合物可以是药物组合物, 也可以是疫 苗组合物。 本发明的组合物可以是治疗性的或预防性的。 本发明的组合物包括有 效量的本发明的自体肿瘤疫苗, 以及至少一种药学上可接受的载体、 稀释剂或赋形 剂。  The present invention also provides a composition, which may be a pharmaceutical composition or a vaccine composition. The compositions of the invention may be therapeutic or prophylactic. The compositions of the present invention comprise an effective amount of an autologous tumor vaccine of the invention, and at least one pharmaceutically acceptable carrier, diluent or excipient.
本发明的药物组合物可制备成各种常规剂型, 其中包括 (但并不限于): 注射剂、 粒剂、 片剂、 丸剂、 栓剂、 胶囊、 悬浮液、 喷雾剂等。  The pharmaceutical compositions of the present invention can be prepared into various conventional dosage forms including, but not limited to, injections, granules, tablets, pills, suppositories, capsules, suspensions, sprays and the like.
(i) 药物组合物  (i) pharmaceutical composition
本发明的药物组合物包括有效量的用本发明方法制备的自体肿瘤疫苗。  The pharmaceutical compositions of the present invention comprise an effective amount of an autologous tumor vaccine prepared by the method of the present invention.
本文所用的术语"有效量 "指治疗剂治疗、 缓解或预防目标疾病或状况的量, 或是表现出可检测的治疗或预防效果的量。 该效果可通过例如抗原水平来检测。 治疗效果也包括生理性症状的减少。 对于某一对象的精确有效量取决于该对象的 体型和健康状况、病症的性质和程度、以及选择给予的治疗剂和 /或治疗剂的组合。 因此, 预先指定准确的有效量是没用的。 然而, 对于某给定的状况而言, 可以用 常规实验来确定该有效量。  The term "effective amount" as used herein refers to an amount of a therapeutic agent that treats, alleviates or prevents a target disease or condition, or an amount that exhibits a detectable therapeutic or prophylactic effect. This effect can be detected by, for example, antigen level. Therapeutic effects also include a reduction in physiological symptoms. The precise effective amount for a subject will depend on the size and health of the subject, the nature and extent of the condition, and the combination of therapeutic and/or therapeutic agents selected for administration. Therefore, it is useless to specify an accurate effective amount in advance. However, for a given situation, routine experimentation can be used to determine the effective amount.
为了本发明的目的, 有效的剂量为给予个体约 0.2微克 /千克至 2微克 /千克。 药物组合物还可含有药学上可接受的载体。 术语"药学上可接受的载体 "指用 于治疗剂 (例如蛋白质、 VLP或其它治疗剂)给药的载体。 该术语指这样一些药剂 载体: 它们本身不诱导产生对接受该组合物的个体有害的抗体, 且给药后没有过 分的毒性。 合适的载体可以是大的、 代谢缓慢的大分子, 如蛋白质、 多糖、 聚乳 酸 (polylactic acid)、 聚乙醇酸等。 这些载体是本领域普通技术人员所熟知的。 在 Remington's Pharmaceutical Sciences(Mack Pub. Co. , N.J. 1991)中可找到关于药学 上可接受的载体或赋形剂的充分讨论。  For the purposes of the present invention, an effective dose is from about 0.2 micrograms per kilogram to 2 micrograms per kilogram of the subject. The pharmaceutical composition may also contain a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" refers to a carrier for administration to a therapeutic agent, such as a protein, VLP or other therapeutic agent. The term refers to pharmaceutical carriers which do not themselves induce the production of antibodies harmful to the individual receiving the composition and which are not excessively toxic after administration. Suitable carriers can be large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acid, polyglycolic acid and the like. These vectors are well known to those of ordinary skill in the art. A full discussion of pharmaceutically acceptable carriers or excipients can be found in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. 1991).
组合物中药学上可接受的载体可包括液体, 如水、 盐水、 甘油和乙醇。 另外, 这些载体中还可能存在辅助性的物质,如润湿剂或乳化剂、 pH缓冲物质等。通常, 可将组合物制成可注射剂, 例如液体溶液或悬液; 还可制成在注射前适合配入溶 液或悬液、 液体赋形剂的固体形式。 脂质体也包括在药学上可接受的载体的定义 中。 The pharmaceutically acceptable carrier in the composition may include liquids such as water, saline, glycerol and ethanol. In addition, auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like may also be present in these carriers. In general, the composition can be formulated as an injectable, such as a liquid solution or suspension; it can also be formulated to be suitable for dissolution prior to injection. Solid form of liquid or suspension, liquid excipient. Liposomes are also included in the definition of pharmaceutically acceptable carriers.
(ii) 疫苗组合物  (ii) Vaccine composition
本发明的疫苗组合物可以是预防性的, 也可以是治疗性的。 所述的疫苗组合 物包含免疫性抗原 (本发明的自体肿瘤疫苗), 并且通常与"药学上可接受的载体" 组合, 这些载体包括本身不诱导产生对接受该组合物的个体有害的抗体的任何载 体。 合适的载体通常是大的、 代谢缓慢的大分子, 如蛋白质、 多糖、 聚乳酸、 聚 乙醇酸、 氨基酸聚合物、 氨基酸共聚物、 脂质凝集物 (如油滴或脂质体)等。 这些 载体是本领域普通技术人员所熟知的。 另外, 这些载体可起免疫刺激剂 ("佐剂") 作用。 另外, 抗原也可以和细菌类毒素 (如白喉、 破伤风、 霍乱、 幽门螺杆菌等病 原体的类毒素)或细菌提取物 (如内毒素或鞭毛素)偶联。  The vaccine compositions of the invention may be prophylactic or therapeutic. The vaccine composition comprises an immunogenic antigen (autologous tumor vaccine of the invention) and is typically combined with a "pharmaceutically acceptable carrier" which includes antibodies which do not themselves induce the production of antibodies harmful to the individual receiving the composition. Any carrier. Suitable carriers are generally large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acid, polyglycolic acid, amino acid polymers, amino acid copolymers, lipid agglutinates (e.g., oil droplets or liposomes), and the like. These vectors are well known to those of ordinary skill in the art. In addition, these carriers can function as immunostimulating agents ("adjuvants"). In addition, the antigen can be coupled to bacterial toxoids (such as toxoids of pathogens such as diphtheria, tetanus, cholera, Helicobacter pylori) or bacterial extracts (such as endotoxin or flagellin).
增强免疫组合物效果的优选佐剂包括但不限于: (1)铝盐 (alum), 如氢氧化铝、 磷酸铝、 硫酸铝等; (2)水包油型乳剂配方, 例如, (a)MF59(参见 WO 90/14837), (b)SAF, 禾口(c)Ribi™佐剂系统 (RAS)(Ribi Immunochem, Hamilton, MT), (3)皂素 佐剂; (4^ 61111(1完全佐剂( ? )和? 61111(1不完全佐剂(1? ); (5)细胞因子, 如白 介素(如 IL-1、 IL-2、 IL-4、 IL-5、 IL-6、 IL-7、 IL-12等)、 干扰素(如 γ干扰素)、 巨 噬细胞集落刺激因子 (M-CFS)、 单核-巨噬细胞集落刺激因子 (GM-CFS), 肿瘤坏死 因子 (TNF)等; (6)细菌 ADP-核糖基化毒素 (如霍乱毒素 CT, 百日咳毒素 PT或大 肠杆菌热不稳定毒素 LT)的脱毒变异体, 参见例如 WO93/13302和 WO92/19265 ; 以及 (7)作为免疫刺激剂来增强组合物效果的其它物质。 Preferred adjuvants for enhancing the effect of the immunological composition include, but are not limited to: (1) aluminum salts (alum) such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, and the like; (2) oil-in-water emulsion formulations, for example, (a) MF59 (see WO 90/14837), (b) SAF, and (c) RibiTM Adjuvant System (RAS) (Ribi Immunochem, Hamilton, MT), (3) saponin adjuvant; (4^ 61111 ( 1 Complete adjuvant (?) and ?61111 ( 1 incomplete adjuvant (1?); (5) cytokines, such as interleukins (such as IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc., interferon (such as gamma interferon), macrophage colony-stimulating factor (M-CFS), mononuclear-macrophage colony-stimulating factor (GM-CFS), tumor necrosis factor ( TNF), etc.; (6) Detoxification variants of bacterial ADP-ribosylating toxins (such as cholera toxin CT, pertussis toxin PT or E. coli heat labile toxin LT), see for example WO93/13302 and WO92/19265; 7) Other substances that act as immunostimulating agents to enhance the effect of the composition.
包括免疫原性组合物在内的疫苗组合物 (例如, 可包括抗原、 药学上可接受的 载体以及佐剂), 通常含有稀释剂, 如水, 盐水, 甘油, 乙醇等。 另外, 辅助性物 质, 如润湿剂或乳化剂、 pH缓冲物质等可存在于这类运载体中。  Vaccine compositions, including immunogenic compositions (e.g., can include antigens, pharmaceutically acceptable carriers, and adjuvants), typically contain a diluent such as water, saline, glycerol, ethanol, and the like. In addition, auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like may be present in such carriers.
更具体地, 包括免疫原性组合物在内的疫苗, 包含免疫学有效量的免疫原性 多肽, 以及上述其它所需的组分。 "免疫学有效量"指以单剂或连续剂一部分给予 个体的量对治疗或预防是有效的。 该用量可根据所治疗个体的健康状况和生理状 况、 所治疗个体的类别 (如人)、 个体免疫系统合成抗体的能力、 所需的保护程度、 疫苗的配制、 治疗医师对医疗状况的评估、 及其它的相关因素而定。 预计该用量 将在相对较宽的范围内, 可通过常规实验来确定。 通常, 可将疫苗组合物或免疫原性组合物制成可注射剂, 例如液体溶液或悬 液; 还可制成在注射前适合配入溶液或悬液、 液体赋形剂的固体形式。 该制剂还 可乳化或包封在脂质体中, 以增强佐剂效果。 More specifically, vaccines, including immunogenic compositions, comprise an immunologically effective amount of an immunogenic polypeptide, as well as other desired components as described above. "Immunologically effective amount" means that the amount administered to a subject in a single dose or in a continuous dose is effective for treatment or prevention. The amount may be based on the health and physiological condition of the individual being treated, the type of individual being treated (eg, human), the ability of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation of the vaccine, the assessment of the medical condition by the treating physician, And other relevant factors. This amount is expected to be in a relatively wide range and can be determined by routine experimentation. In general, the vaccine composition or immunogenic composition can be formulated as an injectable preparation, such as a liquid solution or suspension; it can also be formulated as a solid form suitable for solution or suspension, liquid excipient prior to injection. The formulation may also be emulsified or encapsulated in liposomes to enhance the adjuvant effect.
(iii) 给药途径和剂量  (iii) route of administration and dosage
所述组合物可以直接给予对象。对象可以是人或非人哺乳动物,较佳地为人。 当用作疫苗时, 可用已知的方法直接施用于个体。 通常采用与常规疫苗相同的施用 途径施用这些组合物。  The composition can be administered directly to a subject. The subject can be a human or a non-human mammal, preferably a human. When used as a vaccine, it can be directly administered to an individual by a known method. These compositions are usually administered by the same route of administration as conventional vaccines.
给予本发明药物组合物或疫苗组合物的途径包括 (但并不限于): 配制好的药物 组合物可以通过常规途径进行给药, 其中包括 (但并不限于): 如皮内单点 /多点注 射、 皮下单点 /多点注射、 淋巴结内或周围注射、 肌肉注射、 胸腔 /腹腔注射、 静脉 注射、 瘤周 /瘤内注射。 上述途径可视具体情况单独运用, 必要时联合应用。 注入 患者自身的部位, 可为但不限于原发灶、 转移灶或其周围淋巴引流区及四肢远端 或近端、 胸背部、 腹部、 颈部、 局部可触及的淋巴结或淋巴引流区。  Routes for administering a pharmaceutical composition or vaccine composition of the invention include, but are not limited to: Formulated pharmaceutical compositions can be administered by conventional routes, including but not limited to: such as intradermal single/multiple Point injection, subcutaneous single/multiple injection, intra- or peripheral lymphatic injection, intramuscular injection, intrathoracic/peritoneal injection, intravenous injection, peritumoral/intratumoral injection. The above approaches may be applied separately depending on the specific situation, and may be applied in combination if necessary. The patient's own site may be, but is not limited to, the primary tumor, metastases or surrounding lymphatic drainage areas and distal or proximal extremities, thoracodorsal, abdomen, neck, locally accessible lymph nodes or lymphatic drainage areas.
使用药物组合物时, 应考虑使用剂量控制在安全有效量的范围内, 具体剂量 还应考虑给药途径、 病人健康状况等因素, 这些都是熟练医师技能范围之内的。  When using a pharmaceutical composition, dose control should be considered within a safe and effective range. The specific dose should also take into account the route of administration, the health of the patient, etc., which are within the skill of the skilled physician.
此外, 本发明的疫苗还可与其他药物或物理疗法联用, 其中包括 (但并不限于): 各种细胞因子, 如 IFN、 TNF、 IL-2, GM-CSF, G-CSF, SCF, 以及电磁波等。 本发明的主要优点如下- (1) 本发明用照射和 /或加热主动激发肿瘤抗原表达和抗原加工提呈功能, 再 经灭活后, 辅以佐剂, 并按照个体免疫反应情况调节免疫治疗方案, 达到真正个 体化肿瘤免疫治疗的目的;  In addition, the vaccine of the present invention may also be used in combination with other drugs or physical therapies, including but not limited to: various cytokines such as IFN, TNF, IL-2, GM-CSF, G-CSF, SCF, And electromagnetic waves, etc. The main advantages of the present invention are as follows - (1) The present invention actively stimulates tumor antigen expression and antigen processing presentation function by irradiation and/or heating, and then inactivates, supplements with adjuvant, and modulates immunotherapy according to individual immune response. Program, to achieve the purpose of truly individualized tumor immunotherapy;
(2) 本发明能促进机体内提高免疫能力, 特别是针对肿瘤的细胞免疫和体液 免疫;  (2) The present invention can promote immunity in the body, especially for cellular immunity and humoral immunity of tumors;
(3) 本发明中所述治疗或预防肿瘤的技术方法, 适用于医疗领域, 特别是预 防或者治疗肿瘤, 亦适用于术后肿瘤残留及存在微转移灶的患者, 为本领域提供 了一种新的选择, 具有广阔的应用前景。 下面结合具体实施例, 进一步阐述本发明。 应理解, 这些实施例仅用于说明 本发明而不用于限制本发明的范围。 下列实施例中未注明具体条件的实验方法, 通常按照常规条件如 Sambrook等人, 分子克隆: 实验室手册 (New York: Cold Spring Harbor Laboratory Press, 1989)中所述的条件, 或按照制造厂商所建议的条 件。 实施例 1 (3) The technical method for treating or preventing tumors according to the present invention is suitable for use in the medical field, particularly for preventing or treating tumors, and is also suitable for postoperative tumor residuals and patients with micrometastases, and provides a field in the art. The new choice has broad application prospects. The invention is further illustrated below in conjunction with specific embodiments. It should be understood that these embodiments are for illustrative purposes only. The invention is not intended to limit the scope of the invention. The experimental methods in the following examples which do not specify the specific conditions are usually carried out according to the conditions described in conventional conditions such as Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to the manufacturer. The suggested conditions. Example 1
个体化瘤苗的制备  Preparation of individualized tumor vaccine
取处于对数生长期的 4T1细胞或 Lewis肺癌细胞 1*107个,经 γ射线 (4-10 Gy, 1.4 Gy/min)照射处理后 30-60分钟, 37-40°C水浴加热 20 分钟; 在 37 °C, 5%C02 恒温培养箱 48小时, 再经超声破碎细胞后加入福氏佐剂, 制备成乳化剂即为肿瘤 全细胞疫苗。 实施例 2 Take 1*10 7 cells in Lewis logarithmic growth phase or Lewis lung cancer cells, 30-60 minutes after irradiation with γ-rays (4-10 Gy, 1.4 Gy/min), and heat for 20 minutes in 37-40 °C water bath. At 37 ° C, 5% CO 2 incubator for 48 hours, then ultrasonically disrupted cells and then added Freund's adjuvant to prepare an emulsifier is a tumor whole cell vaccine. Example 2
个体化瘤苗治疗小鼠皮下乳腺癌肿瘤  Individualized tumor vaccine for treatment of subcutaneous breast cancer in mice
在本实施例中, 经细胞、 动物实验各相关指标证实, 本发明方法制备的瘤苗 有很强的抗原递呈性及激发抗体免疫反应的能力。 具体结果如下:  In the present example, it was confirmed by the relevant indexes of the cell and animal experiments that the tumor vaccine prepared by the method of the present invention has strong antigen presentation ability and ability to elicit an antibody immune response. The specific results are as follows:
1.促进抗肿瘤抗体的产生  1. Promote the production of anti-tumor antibodies
实验方法:常规的 Balb/c小鼠经过实施例 1制备的肿瘤细胞抗原免疫后 3周, 取血浆倍比稀释, 加入用肿瘤细胞包被的 ELISA板, 以抗鼠 IgG-HRP及 TMB底 物显示抗体滴度 (本实验为 1 : 100000)。  Experimental method: Conventional Balb/c mice were immunized 3 weeks after immunization with the tumor cell antigen prepared in Example 1, and the plasma was diluted and added to the ELISA plate coated with tumor cells to anti-mouse IgG-HRP and TMB substrates. The antibody titer is shown (this experiment is 1:100000).
结果表明, 用实施例 1制备的肿瘤抗原与对照方法制备的瘤苗相比, 有更强 的促抗肿瘤抗体产生能力(图 1): 在免疫 Balb /c小鼠 3周后, 实施例 1制备的抗 原刺激更多的抗 4T1乳腺癌细胞的抗体 (t检验, p<0.05, n=10), NC:对照; NS : 佐剂对照; ORIGINAL: 原旧方法 (单纯杀死肿瘤细胞后加入佐剂)制备的瘤苗 (作 为对照); EW: 本发明制备的新型瘤苗。  The results showed that the tumor antigen prepared in Example 1 was more potent against tumor antibody production than the tumor vaccine prepared by the control method (Fig. 1): After 3 weeks of immunization of Balb/c mice, Example 1 The prepared antigen stimulated more antibodies against 4T1 breast cancer cells (t test, p<0.05, n=10), NC: control; NS: adjuvant control; ORIGINAL: original method (added only after killing tumor cells) Adjuvant) prepared tumor vaccine (as a control); EW: a novel tumor vaccine prepared by the present invention.
2.促进抗肿瘤细胞免疫反应  2. Promote anti-tumor cell immune response
方法同上。 Balb/c小鼠经实施例 1制备的肿瘤细胞抗原免疫 3周后, 取淋巴 细胞 (LN)及脾细胞 (SP)与构建的含有 Luciferase的 4T1肿瘤细胞 (4Tl-luc)以 10: 1, 100: 1, 1000: 1的比例混合培养, 72小时后测定由死亡 4T1细胞释放的 Luciferase 的活性 (RLu表示)。 The method is the same as above. Balb/c mice were immunized with the tumor cell antigen prepared in Example 1 for 3 weeks, and lymphocytes (LN) and spleen cells (SP) were taken and the constructed Luciferase-containing 4T1 tumor cells (4T1-luc) were 10:1. 100: 1, 1000: 1 ratio mixed culture, 72 hours after the release of Luciferase released from dead 4T1 cells Activity (represented by RLu).
结果如图 2所示, 小鼠接受实施例 1制备的肿瘤细胞抗原免疫能够产生较佐 剂免疫更强的 T细胞杀伤 (p<0.05, n=5); 图 2A为佐剂组抗肿瘤 T细胞杀伤实验, 图 2Β为实施例 1方法组制备的肿瘤细胞抗原抗肿瘤 Τ细胞杀伤实验, Rlu指 Luciferase的活性; NC指正常对照组; LN指淋巴结细胞; SP指脾细胞。 实施例 3 As a result, as shown in Fig. 2, mice immunized with the tumor cells prepared in Example 1 were able to produce T cell killing more strongly than adjuvant (p < 0.05, n = 5) ; Fig. 2A is an antitumor T in the adjuvant group. Cell killing experiment, Fig. 2 is the tumor cell antigen anti-tumor cell killing experiment prepared by the method group of Example 1, Rlu refers to the activity of Luciferase; NC refers to the normal control group; LN refers to lymph node cells; SP refers to spleen cells. Example 3
对小鼠肿瘤的抑制作用  Inhibition of mouse tumors
根据免疫学原理, 小鼠抗肿瘤免疫试验分两步:  According to the principle of immunology, the mouse anti-tumor immunity test is divided into two steps:
1. 肿瘤免疫致敏: 将处理后的肿瘤抗原新型瘤苗先接种在待试小鼠皮下, 14 天后, 再强化免疫一次, 7天后接种同源性小鼠肿瘤细胞 (图 3)。  1. Tumor immunosensitization: The treated tumor antigen novel tumor vaccine was first inoculated subcutaneously in the test mice, and 14 days later, the immunization was boosted once again, and the homologous mouse tumor cells were inoculated 7 days later (Fig. 3).
2. 免疫抗癌效应: 分两类:  2. Immunization against cancer: There are two categories:
a、 对原位癌的抑制作用: 使用 BALB/c小鼠与 4T1乳腺癌细胞株; b、 对转移瘤的抑制作用: 使用 C57BL/6小鼠和 Lewis肺癌细胞株。 Lewis 及 4T1癌细胞均为高度恶性, 生长快, 难以控制, 且能够形成肺转移癌。  a. Inhibition of carcinoma in situ: BALB/c mice and 4T1 breast cancer cell lines were used; b. Inhibition of metastatic tumors: C57BL/6 mice and Lewis lung cancer cell lines were used. Lewis and 4T1 cancer cells are highly malignant, grow fast, are difficult to control, and can form lung metastases.
抗肿瘤效应的判断:  Judging anti-tumor effect:
1. 抗原位癌疗效判断:  1. Antigenic cancer judgment:
a、 活体肿瘤荧光定量法: 上述 4T1细胞带有 Luciferase基因, 故 4T1-Luc肿 瘤大小可经向麻醉荷瘤小鼠腹腔灌注 Luciferin底物, 5分钟后测定荧光强度; b、 游标卡尺测定肿瘤直径; 这两种方法均能客观反应肿瘤大小。  a, living tumor fluorescence quantitative method: the above 4T1 cells with Luciferase gene, so 4T1-Luc tumor size can be perfused into the anesthesia tumor-bearing mice peritoneal injection of Luciferin substrate, 5 minutes after the determination of fluorescence intensity; b, vernier caliper to determine the tumor diameter; Both methods can objectively reflect tumor size.
2. 抗肺转移癌疗效: 由于实验性 Lewis肺癌生长速度快, 癌结节迅速融合成 片, 无法计算结节数目, 故以全肺称重计算结果。  2. Anti-pulmonary metastasis effect: Due to the rapid growth of experimental Lewis lung cancer, the cancer nodules are rapidly fused into a piece, and the number of nodules cannot be calculated. Therefore, the results are calculated by whole lung weighing.
经过实施例 1制备的肿瘤抗原免疫后的小鼠, 再接受活肿瘤细胞的静脉注射 或皮下注射形成转移癌和原位癌, 供抗肿瘤效应的研究。  The mice immunized with the tumor antigen prepared in Example 1 were subjected to intravenous or subcutaneous injection of live tumor cells to form metastatic cancer and carcinoma in situ for the antitumor effect.
实验结果如下:  The experimental results are as follows:
1. 对原位癌的杀伤抑制作用  1. Killing inhibition of carcinoma in situ
如图 4所示, 荷瘤 Balb/c小鼠肿瘤荧光活体成像所示, 与对照组 (佐剂免疫) 及原旧方法制备的瘤苗 (佐剂 +灭活肿瘤细胞)组相比, 实施例 1制备的疫苗组肿瘤 个数少, 且体积小。 肿瘤生长曲线 (图 5)表明新型瘤苗免疫组原位癌生长速度较原旧瘤苗制备法 免疫组、 单独佐剂组、 及生理盐水组明显缓慢 (p<0.01), 该实验经过三次重复均获 得相似结果 (图 6、图 7)。值得注意的是, 4T1-Luc细胞增长最快的肿瘤不能为 Taxol 5mg/kg (每日注射)所抑制, 却能被本发明方法制备的疫苗所抑制。 As shown in Figure 4, the tumor-injected Balb/c mouse tumor fluorescence imaging showed that compared with the control group (adjuvant immunization) and the original method of preparation of tumor vaccine (adjuvant + inactivated tumor cells) group, The vaccine group prepared in Example 1 had a small number of tumors and a small volume. The tumor growth curve (Fig. 5) showed that the growth rate of the in situ carcinoma of the new tumor vaccine group was significantly slower than that of the original tumor vaccine preparation group, the adjuvant group alone, and the saline group (p<0.01). Similar results were obtained (Fig. 6, Fig. 7). It is worth noting that the fastest growing tumor of 4T1-Luc cells could not be inhibited by Taxol 5 mg/kg (daily injection) but was inhibited by the vaccine prepared by the method of the present invention.
2. 对肺转移癌的杀伤抑制  2. Kill inhibition of lung metastases
转移是肿瘤致死的主要原因, 为探讨本发明疫苗对转移性肿瘤的杀伤抑制作 用, 将 Lewis肺癌细胞在同源性 C57BL/6小鼠上的实验性肺转移作为检测模型。  Metastasis is the main cause of tumor death. To investigate the killing inhibition effect of the vaccine of the present invention on metastatic tumors, experimental lung metastasis of Lewis lung cancer cells on homologous C57BL/6 mice was used as a detection model.
常规市售的 C57BL/6小鼠 (8周龄、 雌性、 5只 /组)分为四组:  Conventional commercially available C57BL/6 mice (8 weeks old, female, 5/group) were divided into four groups:
A、 生理盐水对照组, 以生理盐水皮下注射模拟免疫过程;  A, saline control group, subcutaneous injection of simulated saline immunization process;
B、 佐剂对照组, 只接受佐剂而无肿瘤细胞抗原;  B. Adjuvant control group, only receiving adjuvant without tumor cell antigen;
C、 新方法 (新型瘤苗)组, 佐剂 +处理后的肿瘤抗原;  C, new method (new tumor vaccine) group, adjuvant + treated tumor antigen;
D、 Taxol组 (紫杉醇组): 常规化疗药, 作为阳性对照组。  D, Taxol group (Paclitaxel group): Conventional chemotherapy drugs, as a positive control group.
四组小鼠均在接受不同免疫处理后 21天, 经静脉注入 l x lO5个活 Lewis肺癌 细胞。 23天后, 称量小鼠肺肿瘤重量, 得到均值和标准差。 Four groups of mice were injected with lx lO 5 live Lewis lung cancer cells 21 days after receiving different immunotherapy. After 23 days, the lung tumor weight of the mice was weighed to obtain the mean and standard deviation.
结果表明(图 8):三个对照组 (无免疫、佐剂、 Taxol)之间的肺重量无差异, Taxol The results showed (Fig. 8): no difference in lung weight between the three control groups (no immunization, adjuvant, Taxol), Taxol
5mg/Kg不能抑制住 Lewis肺肿瘤的生长; 但新方法 (新型瘤苗)组却使肿瘤减少 400mg, t-test分析表明该数据具有统计学差异 (p=0.0069)。 在本发明提及的所有文献都在本申请中引用作为参考, 就如同每一篇文献被 单独引用作为参考那样。 此外应理解, 在阅读了本发明的上述讲授内容之后, 本 领域技术人员可以对本发明作各种改动或修改, 这些等价形式同样落于本申请所 附权利要求书所限定的范围。 5mg/Kg did not inhibit the growth of Lewis lung tumors; however, the new method (new tumor vaccine) group reduced tumors by 400 mg, and t-test analysis showed statistically significant differences (p=0.0069). All documents mentioned in the present application are hereby incorporated by reference in their entirety in their entireties in the the the the the the the the the In addition, it is to be understood that various modifications and changes may be made by those skilled in the art in the form of the appended claims.

Claims

权 利 要 求 Rights request
1. 一种制备自体肿瘤疫苗的方法, 其特征在于, 包括步骤: 1. A method for preparing an autologous tumor vaccine, characterized by comprising the steps:
(1) 获得分离的自体肿瘤细胞; (1) Obtain isolated autologous tumor cells;
(2) 用放射法和 /或加热法处理步骤 (1)的自体肿瘤细胞, 获得抗原性和抗原提 呈性增强的肿瘤细胞; (2) Treat the autologous tumor cells in step (1) with radiation and/or heating to obtain tumor cells with enhanced antigenicity and antigen-presenting properties;
(3) 对步骤 (2)获得的肿瘤细胞进行灭活处理, 获得灭活的肿瘤细胞; (3) Inactivate the tumor cells obtained in step (2) to obtain inactivated tumor cells;
(4) 将步骤 (3)获得的灭活的肿瘤细胞制备为自体肿瘤疫苗。 (4) Prepare an autologous tumor vaccine from the inactivated tumor cells obtained in step (3).
2. 如权利要求 1所述的方法,其特征在于,所述的肿瘤为:实体瘤或非实体瘤。 3. 如权利要求 1所述的方法, 其特征在于, 步骤 (1)所述获得分离的自体肿瘤 细胞的方法选自下组: 用酶消化法、 机械剪切法、 或人工磨碎肿瘤组织法将肿瘤 组织处理为可培养的分离的自体肿瘤细胞。 2. The method of claim 1, wherein the tumor is: a solid tumor or a non-solid tumor. 3. The method of claim 1, wherein the method of obtaining isolated autologous tumor cells in step (1) is selected from the group consisting of enzymatic digestion, mechanical shearing, or manual grinding of tumor tissue. The method is to process tumor tissue into isolated autologous tumor cells that can be cultured.
4. 如权利要求 1所述的方法, 其特征在于, 步骤 (2)所述的放射法选自下组: 用 选自 α, β、 γ、 质子、 或重离子放射线、 以任选自 0.1-50 Gy剂量范围、 任选自 0.1-10 Gy/min剂量率, 处理步骤 (1)的自体肿瘤细胞, 获得抗原性和抗原提呈性增强的肿 瘤细胞。 4. The method of claim 1, wherein the radiation method in step (2) is selected from the following group: using radiation selected from α, β, γ, protons, or heavy ion radiation, optionally selected from 0.1 The autologous tumor cells in step (1) are processed in a dose range of -50 Gy and a dose rate optionally selected from 0.1-10 Gy/min to obtain tumor cells with enhanced antigenicity and antigen-presenting properties.
5. 如权利要求 1所述的方法, 其特征在于, 步骤 (2) 所述的加热法选自下组: 加热温度任选自 37.5 °C-50°C (优选 38°C-45 °C),加热时间任选自 l min-2小时 (优选 30min-l小时)。 5. The method according to claim 1, characterized in that the heating method in step (2) is selected from the following group: The heating temperature is optionally selected from 37.5°C-50°C (preferably 38°C-45°C ), the heating time is optionally selected from 1 min to 2 hours (preferably 30 min to 1 hour).
6. 如权利要求 1所述的方法, 其特征在于, 在步骤 (2)和步骤 (3)之间, 还包括步 骤: 对步骤 (2)获得抗原性和抗原提呈性增强的肿瘤细胞进行培养处理。 6. The method according to claim 1, characterized in that, between step (2) and step (3), it further includes the step of: performing a test on the tumor cells with enhanced antigenicity and antigen-presenting properties obtained in step (2). Cultivation processing.
7. 如权利要求 1所述的方法, 其特征在于, 步骤 (3)所述的灭活处理选自下组: 微波细胞灭活法、 电磁波细胞灭活法, 激光细胞灭活法、 高温细胞灭活法、 超声破碎 细胞灭活法、 反复细胞冻融、 匀浆、 离心、 沉淀细胞灭活法、 酶学消化细胞灭活法、 低渗破裂法、 或其组合。 7. The method of claim 1, wherein the inactivation treatment in step (3) is selected from the group consisting of: microwave cell inactivation method, electromagnetic wave cell inactivation method, laser cell inactivation method, high temperature cell inactivation method Inactivation method, ultrasonic disruption cell inactivation method, repeated cell freezing and thawing, homogenization, centrifugation, sedimentation cell inactivation method, enzymatic digestion cell inactivation method, hypotonic rupture method, or a combination thereof.
8. 如权利要求 1所述的方法, 其特征在于, 在步骤 (4)中, 将步骤 (3)获得的灭活 的肿瘤细胞与佐剂混合, 获得自体肿瘤疫苗。 8. The method of claim 1, wherein in step (4), the inactivated tumor cells obtained in step (3) are mixed with an adjuvant to obtain an autologous tumor vaccine.
9. 一种自体肿瘤疫苗, 其特征在于, 所述疫苗是用权利要求 1所述的方法制备 的。 9. An autologous tumor vaccine, characterized in that the vaccine is prepared by the method of claim 1.
10. 权利要求 9所述自体肿瘤疫苗的用途, 其特征在于, 它被用于制备预防或治 疗肿瘤的组合物。 10. The use of the autologous tumor vaccine according to claim 9, characterized in that it is used to prepare a composition for preventing or treating tumors.
1 1. 如权利要求 1所述的方法, 其特征在于, 步骤 (3)所述的灭活处理选自下组: 微波细胞灭活法、 电磁波细胞灭活法, 激光细胞灭活法、 高温细胞灭活法、 超声破碎 细胞灭活法、 反复细胞冻融、 匀浆、 离心、 沉淀细胞灭活法、 酶学消化细胞灭活法、 低渗破裂法、 化学共价交联法、 化学性变性法, 或其组合。 1 1. The method of claim 1, wherein the inactivation treatment in step (3) is selected from the group consisting of: microwave cell inactivation method, electromagnetic wave cell inactivation method, laser cell inactivation method, high temperature Cell inactivation method, ultrasonic disruption cell inactivation method, repeated cell freezing and thawing, homogenization, centrifugation, sedimentation cell inactivation method, enzymatic digestion cell inactivation method, Hypotonic rupture method, chemical covalent cross-linking method, chemical denaturation method, or a combination thereof.
12. —种预防或治疗肿瘤的方法, 其特征在于, 包括对对象施用有效量的如权利 要求 9所述的肿瘤疫苗。 12. A method for preventing or treating tumors, characterized by comprising administering an effective amount of the tumor vaccine according to claim 9 to a subject.
13. 如权利要求 12所述的方法, 其特征在于, 所述施用时间选自下组: 术后、 化疗前, 或 WBC回升至 >3000个 /ul时。 13. The method of claim 12, wherein the administration time is selected from the following group: after surgery, before chemotherapy, or when WBC rises to >3000/ul.
14. 如权利要求 12所述的方法, 其特征在于, 所述的施用次数可为 4个疗程 (每 疗程 3-4周)至终生。 14. The method of claim 12, wherein the number of administrations can range from 4 treatment courses (3-4 weeks per treatment course) to a lifetime.
15. 如权利要求 12所述的方法, 其特征在于, 所述方法还包括: 用免疫学实验 来监测免疫反应程度。 15. The method of claim 12, wherein the method further includes: using immunological experiments to monitor the degree of immune response.
16. 如权利要求 15所述的方法, 其特征在于, 所述的免疫学实验选自下组: 细胞免疫反应、 体液免疫反应、 免疫调节反应, 或其组合。 16. The method of claim 15, wherein the immunological experiment is selected from the group consisting of: cellular immune response, humoral immune response, immune regulatory response, or a combination thereof.
17. 如权利要求 12所述的方法, 其特征在于, 所述的免疫学实验在首次免疫 后 15-60天内进行。 17. The method of claim 12, wherein the immunological experiment is performed within 15-60 days after the first immunization.
18. 如权利要求 12所述的方法, 其特征在于, 所述的方法还包括: 根据免疫 学实验测得的免疫反应程度, 判定治疗效果和 /或对治疗方案进行调整。 18. The method of claim 12, wherein the method further includes: determining the therapeutic effect and/or adjusting the treatment plan based on the degree of immune response measured by immunological experiments.
19. 如权利要求 12所述的方法, 其特征在于, 所述方法包括: 19. The method of claim 12, characterized in that, the method includes:
(i) 术后第三天, 对患者施用所述疫苗; (i) on the third postoperative day, administer the vaccine to the patient;
(ii) 术后 7-14天, 再次对患者施用所述疫苗; (ii) 7-14 days after surgery, administer the vaccine to the patient again;
(iii) 术后 14-24天, 再次对患者施用所述疫苗; (iii) 14-24 days after surgery, administer the vaccine to the patient again;
(iv) 术后第 31-34天用免疫学实验来监测免疫反应程度, 并通过检测结果调 节免疫治疗方案。 (iv) Use immunological experiments to monitor the degree of immune response on days 31-34 after surgery, and adjust the immunotherapy plan based on the test results.
PCT/CN2013/087266 2012-11-15 2013-11-15 Method for preparing autologous tumor vaccine and use thereof WO2014075631A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201210466990.9A CN103800896A (en) 2012-11-15 2012-11-15 Preparation method and application of autologous tumor vaccine
CN201210466990.9 2012-11-15

Publications (1)

Publication Number Publication Date
WO2014075631A1 true WO2014075631A1 (en) 2014-05-22

Family

ID=50698589

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2013/087266 WO2014075631A1 (en) 2012-11-15 2013-11-15 Method for preparing autologous tumor vaccine and use thereof

Country Status (2)

Country Link
CN (2) CN103800896A (en)
WO (1) WO2014075631A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019204391A1 (en) * 2018-04-19 2019-10-24 Robert Caruso Cryo-inactivated cancer cells for cancer immunotherapy
WO2021016261A3 (en) * 2019-07-22 2021-04-08 Todd Alamin Cell treatments and therapeutic reinfusion methods
WO2024069531A1 (en) * 2022-09-30 2024-04-04 Novocure Gmbh Compositions, systems, and methods for treating cancer using alternating electric fields and apoptotic cancer cell vaccination

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108114274B (en) * 2017-12-26 2019-10-25 中国医学科学院生物医学工程研究所 It is a kind of based on tumour cell be template tumor vaccine and preparation method thereof
CN108175853A (en) * 2018-01-24 2018-06-19 上海赛群生物科技有限公司 A kind of tumor cell vaccine and preparation method thereof
CN109999187A (en) * 2019-05-09 2019-07-12 英诺激光科技股份有限公司 A method of cancer cell vaccine is prepared using laser
CN113827710A (en) * 2020-06-23 2021-12-24 四川大学华西医院 Tumor whole-cell vaccine based on lactic acid
CN111603554A (en) * 2020-07-07 2020-09-01 华中科技大学同济医学院附属协和医院 Preparation method and application of anti-tumor vaccine antigen raw material
CN113476594A (en) * 2021-06-30 2021-10-08 澳门大学 Quantum dot enhanced tumor inactivated vaccine and preparation method and application thereof
WO2023114273A1 (en) * 2021-12-14 2023-06-22 Lurong Zhang Methods of treating cancer and monitoring anti-cancer immunity
CN115192707B (en) * 2022-06-30 2023-05-30 重庆医科大学附属第二医院 Tumor antigen trapping nanoparticle and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004018659A1 (en) * 2002-08-16 2004-03-04 Glycotope Gmbh Process for the production of temperature-induced tumor cell lysates for use as immunogenic compounds

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL140796A0 (en) * 2001-01-08 2002-02-10 Hadasit Med Res Service An autologous anti-cancer vaccine
CN1754576A (en) * 2004-09-29 2006-04-05 本元正阳基因技术股份有限公司 Tumour cell vaccine preparation method
CN102258772B (en) * 2010-05-25 2014-08-13 中国人民解放军第二军医大学 Preparation method and application of new therapeutic vaccine for dendritic tumor cells
CN102343086A (en) * 2010-05-28 2012-02-08 四川大学 Drug and tumor whole-cell vaccine for treating or preventing tumor, and preparation methods and applications of drug and whole-cell vaccine

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004018659A1 (en) * 2002-08-16 2004-03-04 Glycotope Gmbh Process for the production of temperature-induced tumor cell lysates for use as immunogenic compounds

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BENENCIA, F. ET AL.: "Whole tumor antigen vaccination usingdendritic cells: Comparison of RNA electroporation andpulsing with UV-irradiated tumor cells", JOURNAL OF TRANSLATIONAL MEDICINE, vol. 6, no. 21, 2008, pages 1 - 14 *
CHIANG, C.L. ET AL.: "Whole Tumor Antigen Vaccines", SEMIN IMMUNOL., vol. 22, no. 3, 2010, pages 132 - 143, XP027080488, DOI: doi:10.1016/j.smim.2010.02.004 *
LIN, C.C. ET AL.: "Potentiation of the immunotherapeutic effectof autologous dendritic cells by pretreatinghepatocellular carcinoma with low-dose radiation", CLIN INVEST MED., vol. 31, no. 3, 2008, pages E150 - E159 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019204391A1 (en) * 2018-04-19 2019-10-24 Robert Caruso Cryo-inactivated cancer cells for cancer immunotherapy
WO2021016261A3 (en) * 2019-07-22 2021-04-08 Todd Alamin Cell treatments and therapeutic reinfusion methods
CN114466666A (en) * 2019-07-22 2022-05-10 托德·阿拉明 Cell therapy and therapeutic reinfusion method
WO2024069531A1 (en) * 2022-09-30 2024-04-04 Novocure Gmbh Compositions, systems, and methods for treating cancer using alternating electric fields and apoptotic cancer cell vaccination

Also Published As

Publication number Publication date
CN109876137A (en) 2019-06-14
CN103800896A (en) 2014-05-21

Similar Documents

Publication Publication Date Title
WO2014075631A1 (en) Method for preparing autologous tumor vaccine and use thereof
JP5579586B2 (en) Tumor vaccine
JP2018090605A (en) Methods and compositions for clearance of tumors
US11833173B2 (en) Th1 vaccination priming for active immunotherapy
AU2009242471A1 (en) Vaccine compositions and methods
JP2010507584A (en) Extracellular matrix cancer vaccine adjuvant
Zhang et al. Novel immunotherapy for metastatic bladder cancer using vaccine of human interleukin-2 surface-modified MB 49 cells
US20080166369A1 (en) Cancer Vaccine Preparation
GR1009868B (en) Personalized implant against cancer
CN114177282B (en) Use of fluorinated polyethylenimine for preparing vaccine or preparation for preventing/treating diseases caused by virus/bacteria
AU2003277641B2 (en) Remedy for cancer
von Hofe A new ally against cancer
CN115364204A (en) Comprehensive mobilization of individualized active anti-cancer immunity and monitoring of anti-cancer immunity
Guha et al. Immune System Modulation with LOFU And HIFU Treatment of Prostate Cancer
CN1899607A (en) Double effect tumor cell vaccine and its preparing method
Baogang et al. Fixed-tumor vaccine: a practical formulation with cytokine-microspheres for protective and therapeutic antitumor immunity
JP2005281266A (en) Pharmaceutical composition containing thermocell vaccine and cytokine and method for preparing the same
WO2002055103A1 (en) Tumor vaccines
KR20060116850A (en) Intratumoral delivery of dendritic cells

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: 13855275

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13855275

Country of ref document: EP

Kind code of ref document: A1