WO2006042465A1 - Composition vaccinale contenant une proteine de fusion recombinee ainsi qu'un adjuvant et son application - Google Patents

Composition vaccinale contenant une proteine de fusion recombinee ainsi qu'un adjuvant et son application Download PDF

Info

Publication number
WO2006042465A1
WO2006042465A1 PCT/CN2005/001715 CN2005001715W WO2006042465A1 WO 2006042465 A1 WO2006042465 A1 WO 2006042465A1 CN 2005001715 W CN2005001715 W CN 2005001715W WO 2006042465 A1 WO2006042465 A1 WO 2006042465A1
Authority
WO
WIPO (PCT)
Prior art keywords
hsp65
bcg
fusion protein
cells
recombinant fusion
Prior art date
Application number
PCT/CN2005/001715
Other languages
English (en)
French (fr)
Inventor
Liying Wang
Shuping Ren
Yongli Yu
Original Assignee
Beijing Hydvax Biotechnology Co., Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Hydvax Biotechnology Co., Ltd. filed Critical Beijing Hydvax Biotechnology Co., Ltd.
Publication of WO2006042465A1 publication Critical patent/WO2006042465A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/35Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Mycobacteriaceae (F)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55588Adjuvants of undefined constitution
    • A61K2039/55594Adjuvants of undefined constitution from bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • Vaccine composition comprising recombinant fusion protein and adjuvant and application thereof
  • the present invention relates to a vaccine composition comprising a recombinant fusion protein CTL vaccine and an adjuvant BCG for preventing and treating tumors, and a vaccine composition comprising the recombinant fusion protein CTL vaccine and the adjuvant BCG vaccine for preventing and treating a viral infection disease Especially for the prevention and treatment of human breast cancer, rectal cancer, colorectal cancer, laryngeal cancer, gastric cancer, bladder cancer, pancreatic cancer, prostate cancer, ovarian cancer, colon cancer, lung cancer, etc., and for prevention And remediation of diseases caused by viral infections such as liver cancer and cirrhosis caused by SARS virus infection, papillomavirus infection, cervical cancer, hepatitis C, hepatitis B, hepatitis C virus infection and hepatitis B virus infection
  • CTLs cytotoxic T lymphocytes
  • Activation of CTL in vivo requires unsensitized T lymphocytes to simultaneously receive two signals provided by antigen-presenting cells.
  • One is that the foreign antigen peptides are processed by antigen presenting ceIl (APC) and combined with MHC class I molecules.
  • the complex is formed and presented on the surface of the APC, which is recognized by the TCR of the T lymphocyte; the other signal is the expression of the co-stimulatory molecule expressed on the surface of the APC, ie, the B7 molecule, which is recognized by the CD28 molecule of the lymphocyte.
  • APC antigen presenting ceIl
  • HSP heat shock proteins
  • a first signal is provided for activation of specific CTLs.
  • the antigen-presenting cells express only the self peptide, but not the B7 molecule, so the T cells against the self peptide cannot be activated. Therefore, if the T lymphocytes only receive a peptide that presents a peptide that binds to the MHC class I molecule on the surface of the APC, the first signal cannot be activated; the APC also needs to provide a second signal to the T lymphocyte.
  • antigen-presenting cells begin to express B7 molecules when the body invades microorganisms. Therefore, if a microbial-like danger signal can be given to the body, a microbial-like danger signal can stimulate APC to express B7 molecules, which activates T lymphocytes. The cell provides a second signal. (Ruslan Medzhitov and Charles Janeway. Innate Immunity, The New England Journal of Medcine, 2000, 343(5): 338-344).
  • BCG is an immunoadjuvant that has been used for many years. Studies have shown that BCG can activate the natural immune system through Toll-like receptors, induce the expression of cytokines and the expression of surface co-stimulatory molecules (B7 molecules) of antigen-presenting cells (Ruslan). Medzhitov, 1999). Activation of unsensitized T lymphocytes requires antigen presenting cells (APCs) to provide two signals. If APC only provides the first signal, it will lead to immune tolerance. APC must also express surface co-stimulatory molecules, the B7 molecule, to activate unsensitized T lymphocytes. According to the above mechanism, BCG can stimulate the expression of synergistic stimulation molecules on the surface of APC, in order to improve the recombinant fusion protein.
  • APCs antigen presenting cells
  • the CTL vaccine activates the potency of specific CTL.
  • the present invention utilizes BCG as an adjuvant for the recombinant fusion protein CTL vaccine, so that the recombinant fusion protein CTL vaccine can produce a strong stimulation-specific CTL production after entering the body. After BCG enters the body, the body recognizes it as a dangerous signal that can cause infection.
  • the B7 molecule on the surface of the antigen-presenting cell begins to express, providing a second signal for activation of CTL.
  • BCG is an attenuated microorganism that does not cause infection in the body, but it still retains the conserved structure of the microorganism, and activates the Toll-like receptor to induce the expression of B7 molecule, thereby enhancing the efficacy and speed of CTL activation (Ruslan Medzhitov and Charles A. Janeway Jr. Decoding the patterns of self and non-self by the innate immune system. Science, 296 (5566): 298.
  • PAMPs are not unique to microorganisms, they are produced by all microorganisms.
  • PAMPs include lipopolysaccharides (LPS) of Gram-negative bacteria, peptidoglycans and lipopeptides of Gram-positive bacteria, and double-stranded RNA of bacterial flagella, bacterial DNA, and viruses.
  • LPS lipopolysaccharides
  • the Toll-like receptor is a transmembrane protein composed of an extracellular leucine-rich domain and an intracellular domain similar to the IL-1 receptor domain.
  • TLR is mainly expressed in tissues related to immune function, such as: spleen, peripheral blood leukocytes, and tissues and organs in contact with the outside world, such as: lung and gastrointestinal i3 ⁇ 4 o www.InvivoGen.Toll-like ⁇ http://www.InvivoGen .Toll-like>') o So far, ten human Toll-like receptors and nine mouse Toll-like receptors have been found, different Toll-like receptors. The bodies have their respective ligands. TLR2 primarily recognizes a variety of PAMPs, including bacterial lipoproteins, peptidoglycans, and lipocalic acids. TLR3 is involved in the recognition of viral-derived double-stranded RNA.
  • TLR4 is preferentially activated by lipopolysaccharide.
  • TLR9 is required for TLR5 to recognize bacterial flagella but not for methylated CpG DNA.
  • TLR7 and TLR8 recognize small synthetic antiviral molecules.
  • Toll-like receptors need to act synergistically with synergistic receptors, such as TLR4, which often acts synergistically with MD2 and CD14.
  • TLR4 is preferentially activated by lipopolysaccharide.
  • TLR9 is required for TLR5 to recognize bacterial flagella but not for methylated CpG DNA.
  • TLR7 and TLR8 recognize small synthetic antiviral molecules.
  • Toll-like receptors need to act synergistically with synergistic receptors, such as TLR4, which often acts synergistically with MD2 and CD14.
  • Toll-like receptors are essential components of the body's activation of the natural immune system against microbial responses and are acquired by the acquired immune system
  • This vaccine can be used to prevent and treat diseases caused by tumors and viral infections.
  • the diseases caused by the viral infection include SARS virus-infected diseases, papillomavirus infection, hepatitis C, hepatitis B and the like. Since the papillomavirus infection can cause cervical cancer, the recombinant fusion protein of the present invention can also be used for the prevention and/or treatment of cervical cancer.
  • Hepatitis C virus infection and the initial stage of hepatitis B virus infection may cause inflammatory changes in the liver of an infected person. As the infection is repeated, the infected person eventually develops pathological changes of liver cancer and liver cirrhosis. Therefore, the recombinant fusion protein of the present invention It can also be used to prevent and/or treat liver cancer and cirrhosis.
  • one aspect of the invention relates to a vaccine composition characterized by comprising a recombinant fusion protein which is a fusion protein fused with heat shock protein 65, and an adjuvant which is BCG.
  • the recombinant fusion protein may be a recombinant fusion protein HSP65-MUC1 composed of Mycobacterium tuberculosis heat shock protein 65 fusion mucin 1, and a Mycobacterium tuberculosis heat shock protein 65 fusion human epithelial growth factor receptor 2 Recombinant fusion protein HSP65-HER2, Mycobacterium tuberculosis heat shock protein 65 fusion human prostate specific antigen recombinant fusion protein HSP65-PSA, Mycobacterium tuberculosis heat shock protein 65 fusion papillomavirus multi-epitope antigen recombinant fusion protein Recombinant fusion protein HSP consisting of HSP65-HPV, Mycobacterium tuberculosis heat shock protein 65 fusion human SARS virus multi-epitope antigen recombinant fusion protein HSP65-SA S or Mycobacterium tuberculosis heat shock protein 65 fusion hepatitis B core anti
  • HSP65-MUC1, HSP65-HER2, HSP65-PSA, HSP65-HBcAg, HSP65-HPV HSP65-SARS, HSP65-HCV are products developed by the applicant and have applied for national patents (see ZL01102614.6, respectively). , ZL01136347.9, 01134935.2, 200410070084.2, 200410074742.5, 200410074743. X, 02122116.2).
  • Another aspect of the present invention relates to the use of a vaccine composition for the preparation of a medicament for preventing and treating a tumor, which can be used for human breast cancer, rectal cancer, colorectal cancer, laryngeal cancer, gastric cancer, bladder cancer, pancreatic cancer, prostate cancer, Prevention and treatment of ovarian cancer, colon cancer, lung cancer, etc.
  • Another aspect of the invention relates to the use of a vaccine composition for the preparation of a medicament for the prevention and treatment of a human viral infectious disease, which can be used for human SA S virus infection, papillomavirus infection, cervical cancer, hepatitis C, type B Prevention and treatment of liver cancer, liver cirrhosis, etc. caused by hepatitis, hepatitis C virus infection and hepatitis B virus infection.
  • the recombinant fusion protein CTL vaccine of the present invention and the adjuvant thereof can be used in the following three ways: 1. The BCG vaccine is administered, and the recombinant fusion protein CTL vaccine of the present invention is inoculated; 2. The recombinant fusion protein CTL of the present invention is first inoculated. Vaccine, post-administration of BCG; 3. Recombinant fusion protein CTL vaccine of the present invention and BCG vaccine are used simultaneously.
  • the recombinant fusion protein of the present invention After the recombinant fusion protein of the present invention enters the body, it can enter the MHC class I antigen presentation pathway and activate specific CTL. After the adjuvant BCG enters the body, it can activate the antigen-presenting cells to express the costimulatory molecules through the Toll-like receptor. Provide a stronger second signal for activating CTL.
  • Figure 3 Comparison of the activation of dendritic cells by BCG+HSP65-MUC1 and HSP65-MUC1.
  • FIG. 4 BCG potentiates the specific anti-tumor effect stimulated by HSP65-MUC1 injection.
  • FIG. 6 BCG enhances the specific anti-tumor effect stimulated by HSP65-MUC1 immunization.
  • Figure 8 Identification of agarose gel electrophoresis after pET28a-HSP65-HER2 digestion
  • Figure 10 Comparison of BCG+HSP65-HER2 and HSP65-HER2 activation of dendritic cells.
  • FIG. 11 BCG enhances the specific anti-tumor effect stimulated by HSP65-HER2 injection.
  • Figure 12 Comparison of BCG+HSP65-HER2 and HSP65-HER2 induced specific CTL effects.
  • Figure 13 BCG enhances the specific anti-tumor effect stimulated by HSP65-HER2 immunization.
  • Figure 15 Comparison of BCG+HSP65-PSA and HSP65-PSA activation of dendritic cells.
  • FIG. 16 BCG enhancement The specific anti-tumor effect stimulated by HSP65-PSA injection.
  • Figure 17 Comparison of BCG+HSP65-PSA and HSP65-PSA induced specific CTL effects.
  • FIG. 18 BCG potentiates the specific anti-tumor effect stimulated by HSP65-PSA immunization.
  • Figure 21 Comparison of BCG+HSP65-HPV and HSP65-HPV activation of dendritic cells.
  • Figure 22 BCG potentiates the effect of specific killing of HPV-infected cells by HSP65-HPV injection.
  • Figure 23 Comparison of BCG+HSP65-HPV and HSP65-HPV induced specific CTL effects.
  • Figure 24 The effect of specific killing of HPV-infected cells by BCG-forced HSP65-HPV immunization.
  • Figure 25 pMD-18T-SARS/3CL 161. Enzyme digestion of 264 recombinant plasmid
  • Figure 26 HSP65-SARS/ 3CL 161-264 SDS-PAGE identification and Western blot analysis
  • Figure 27 Comparison of BCG+HSP65-SARS and HSP65-SARS activation of dendritic cells.
  • Figure 28 GFP-SARS/3CL 161 . 264 transfected cell identification map
  • Figure 29 BCG potentiates the effect of specific killing of SARS-infected cells by HSP65-SARS injection.
  • Figure 30 Comparison of BCG+HSP65-SARS and HSP65-SARS induced specific CTL effects.
  • Figure 31 BCG enhances the effect of specific killing of SARS-infected cells by HSP65-SARS immunization.
  • Figure 32 ⁇ HSP65-HBV recombinant fusion protein expression map
  • Figure 34 Comparison of BCG+HSP65-HBV and HSP65-HBV activation of dendritic cells.
  • Figure 35 BCG potentiates the effect of specific killing of HBV-infected cells by HSP65-HBV injection.
  • Figure 36 Comparison of BCG+HSP65-HBV and HSP65-HBV induced specific CTL effects.
  • Figure 37 BCG enhances the effect of specific killing of HBV-infected cells stimulated by HSP65-HBV immunization.
  • Figure 38 HSP65-HCV plasmid digestion map
  • Figure 40 Comparison of BCG+HSP65-HCV and HSP65-HCV activation of dendritic cells.
  • Figure 41 BCG potentiates the effect of specific killing of HCV-infected cells stimulated by HSP65-HCV injection.
  • Figure 42 Comparison of BCG+HSP65-HCV and HSP65-HCV induced specific CTL effects.
  • Figure 43 BCG enhances the effect of specific killing of HCV-infected cells stimulated by HSP65-HCV immunization.
  • BCG Originated from Changchun Institute of Biological Products. BCG was cultured in Sutong potato medium at a temperature of 37-39 ⁇ , and the growing BCG showed a dry and yellowish yellow film. The bacterial membrane was collected and BCG genomic DNA was extracted therefrom.
  • BCG genomic DNA was extracted from the bacterial membrane collected in the previous step.
  • HSP65 structural gene was isolated from BCG by PCR.
  • the 5' end primer sequence used was 5' CCATG GCC AAGACAATT GCG3' (SEQ ID NO: 1); the 3' end primer sequence was 5' ACC GAA TTC GCT AGC CAT ATG GAA ATC CAT GCC ACC CAT 3' (SEQ ID NO) : 2).
  • Reaction conditions 94 ° C, 30"; 55 ° C, ⁇ ; 72 ° C, 2 30 cycles, 72 ° C extension for 10 min.
  • PCR product of step 3 was cloned into TA vector by TA cloning method to obtain recombinant plasmid pMD-18T-HSP65.
  • the TA-ligation kit Promega
  • the recombinant plasmid-transformed host strain JM109C was purchased from Invitrogen
  • the positive clones were cultured, and plasmid DNA was prepared by a conventional method (Joseph. Sambrook, David W. Russell. SDS alkaline lysis method, Molecular cloning). : A Laboratory Manual, 3 rd ed.
  • BCG HSP65 gene is SEQ ID NO: 3.
  • Example 2 A fusion gene of HSP65 and human MUCl protein epitope gene was constructed.
  • the expression vector was selected from E. coli expression vector pET28a (+), and the recipient strain was Escherichia coli BL21 (DE3). Strain, (all purchased from Novagen).
  • the MUC1 protein VNTR gene sequence was artificially synthesized, and the £coRI and Hmffll sites were introduced at both ends.
  • the MUC1 protein VNTR gene sequence is as follows:
  • pET28a(+)-HSP65 plasmid as a framework, a double-copy VNTR gene fragment was cloned into this vector to obtain a fusion gene sequence encoding the double copy VNTR sequence of HSP65 and MUC1 protein.
  • the BL21(DE3) host strain was transformed with the recombinant plasmid, and the recombinant plasmid was extracted, and the recombinant inserted into the VNTR sequence of the MUC1 protein was identified, and the obtained fusion protein expression vector was named pET28a(+)/HSP-MUCl.
  • the specific methods are as follows: 1.
  • the HSP65 and human MUC1 epitope fusion protein genes were synthesized by PCR: the template is the coding gene of BCG HSP65 (SEQ E NO: 3) (concentration is 0.01 pmol/L), and the primers for PCR are: 5
  • the 'end primer sequence is: 5, TTC GCC ATG GCC AAG ACA ATT GCG 3' (SEQ ID NO: 5),
  • the sequence of the 3' primer is: 5' GGC CGC AAG CTT CAG AGC CGG ACG GTT GTC CGG
  • the reaction conditions were: 94 ° C, 30 °; 55 ° C, ⁇ ; 72 ° C, 4', after 30 cycles, 72 ° C extension for 10 min.
  • the resulting product sequence is shown in SEQ ID NO: 7.
  • PCR product was first cloned into a TA vector by TA cloning.
  • the TA-ligation kit Promega
  • the recombinant plasmid was transformed into the host strain JM109.
  • the positive clones were cultured, and the plasmid was extracted by a conventional method (see above), and the insert was sequenced by the dideoxy terminator method (ABIPrism310TM, USA ).
  • the sequence was subcloned into the pET28a vector (Novagen) using Ncol and Hindlll cut-points to obtain the pET28a-HSP65-MUCl plasmid.
  • the host strain JM109 was transformed, the positive transformant was cultured, the plasmid was extracted, and the insert was sequenced by the dideoxy terminator method (ABIPrism 310TM, USA).
  • the 2nd lane arrow refers to the 123 bp MUC1 epitope DNA fragment.
  • Example 3 Expression and purification of recombinant HSP65-MUC 1 fusion protein
  • the constructed expression plasmid pET28a(+) /HSP-MUCl was transformed into the prepared E. coli BL21 (DE3) competent cells, and the LB plate containing 100 ⁇ ⁇ / ⁇ kanamycin was coated on the transformation plate. Monoclones were picked, inoculated into 3nd LB liquid medium, shake cultured at 37 rpm on a 37 ° C constant shaker overnight, and extracted by alkaline lysis and assayed for plasmid. The monoclonal containing the expression plasmid pET28a(+) /HSP-MUC1 was confirmed to be plated on LB plates containing 100 ⁇ ⁇ / ⁇ kanamycin.
  • a confirmed engineering strain was selected, and a 10-liter fermentor was used, cultured in LB medium, and induced by IPTG.
  • the high-pressure bacteria-breaking method was used to circulate the bacteria 4 to 5 times under the pressure of 500-600 bar, and the supernatant was centrifuged. The supernatant was subjected to nickel affinity chromatography, 10K membrane block ultrafiltration desalting, and finally passed through the Q-Sepharose FF layer. Analysis, a recombinant HSP-MUC 1 fusion protein (molecular weight 61 KD) was obtained. Sampling was performed by HPLC analysis, and the purity of the recombinant HSP65-MUC1 fusion protein was 96.1%.
  • PBS buffer hereinafter referred to as PBS/EDTA/human serum buffer
  • MDM (Sigma Chemical Co. contains 10% inactivated fetal bovine serum (GIBCO BRL) 100 IU penicillin, 100 mg ml of streptomycin, ⁇ -mercaptoethanol 5 ⁇ 1/1000 ⁇ 1, sodium bicarbonate 3.5g/1000ml)
  • GEBCO BRL inactivated fetal bovine serum
  • ⁇ -mercaptoethanol 5 ⁇ 1/1000 ⁇ 1, sodium bicarbonate 3.5g/1000ml
  • a negative control group was set up: only culture medium was added; BCG group: BCG (Changchun Bioproducts) was added to give a final concentration of 3 ( ⁇ g/ml; BCG+HSP65-MUC1 group: added The final concentrations of BCG and HSP65-MUC1 were 3 ( ⁇ g/ml and 10 ( ⁇ g/ml ; HSP65-MUC1 group: HSP65-MUC1 was added to make the final concentration reach ⁇ ; positive control group: add the following Factor and its final concentration reached 10 ng/ml TNFa (Pharmingen), 10 ng/ml IL-6 (Pharmingen), 1 Ong/ml IL-1 ⁇ (Pharmingen) and 10 ⁇ / ⁇ 1 PGE 2 (Pharmingen).
  • the above BCG and HSP65-MUC 1 proteins and cytokines were added to immature dendritic cells (DC) for 2 days.
  • Dendritic cells were harvested on day 7 and assays for activation and maturation were performed. Determination of dendritic cell activation and maturation indicators
  • DC is the most important antigen-presenting cell in human body.
  • the activation and maturation of DC indicates that the antigen-presenting ability of DC is enhanced, and the ability to activate cytotoxic killer T lymphocytes (CTL) against MUC1 is strengthened; Activated CTLs in vivo kill tumor cells expressing MUCl in vivo and, therefore, can be used to prevent and/or treat MUC1-positive tumors.
  • CTL cytotoxic killer T lymphocytes
  • the pET26-IRES-GFP plasmid was supplied by Novagen, and the pET26-IRES-GFP plasmid was digested with NcoI and Sail enzymes, and the digested product was separated by agarose gel electrophoresis and the GFP fragment slightly larger than 750 bp was recovered.
  • the pET28 vector (Invitrogen) was digested with NcoI and Sail, and the digested product was separated by agarose gel electrophoresis and the linear vector was recovered.
  • the pET28 linear vector and the GFP fragment were ligated with ligase to obtain a pET28-GFP recombinant plasmid.
  • the host strain JM109 was transformed with the pET28-GFP recombinant plasmid, and positive clones were cultured, and the plasmid was extracted by a conventional method (see above), and the insert was sequenced by the dideoxy terminator method (ABIPrism 310TM, USA).
  • pET28-GFP as a template, using primers
  • the above PCR product (GFP gene fragment) was cloned into a TA vector (see Example 1 for the method).
  • Hindlll.BamHI digests pcDNA3 (provided by Invitrogen) and pET28-GFP, respectively, and separates and recovers the digested product by agarose gel electrophoresis, and the recovered digested product is ligated with T4 DNA ligase to obtain a pcDNA3-GFP recombinant plasmid.
  • HSP65-MUC1 is a template, 5' end bow I 5'CTCGAGCCGGTACCGATTCCATGGAATT CAGAT3' (SEQ ID NO: 10), 3' end primer 5'GGCCTCTAGACTCGAGTTATTAGCCAGA AGTAG3' (SEQ ID ⁇ : 11),
  • PCR conditions 94 ° C, 30"; 60 ° C, ⁇ ; 72 ° C, 130", after 27 cycles, 72 ° C extension for 10 min.
  • the digested product was separated and recovered by agarose gel electrophoresis (recovery kit was produced by Beijing Dingguo Biotechnology Co., Ltd., and the operation was carried out according to the instructions), and the recovered digested product was ligated with T4 DNA ligase to obtain pcDNA3-GFP-MUCl.
  • Solution A ⁇ serum-free IMDMo containing 1-2 ⁇ ⁇ pcDNA3-GFP-MUCl plasmid (plasmid containing epitope)
  • Solution B ⁇ serum-free MDM containing 10-20 ⁇ l Liposome reagent (Invitrogen).
  • B16 cells purchased from ATCC were washed once with 2 ml of serum-free DM medium during incubation of the mixture.
  • B16 cells transfected with pcDNA3-GFP-MUCl were screened for transfected positive clones by G418 antibiotic (GIBCO), and these clones were separately digested into culture flasks and expanded and cultured, and RT-PCR and Western Blot were used. Clones with high expression of MUC1 were identified for functional testing.
  • G418 antibiotic G418 antibiotic
  • PBS group negative control group
  • BCG group HSP65-MUC1 group
  • BCG+HSP65-MUC1 group carboplatin group (positive control group)
  • carboplatin group positive control group
  • all mice were inoculated with 1.5 ⁇ 10 5 / 200 ⁇ l PBS of B16 cells transfected with the gene encoding the MUC1 epitope antigen polypeptide, and the inoculated site was subcutaneously in the right side of the mouse.
  • the mice were injected for the first time in different groups, and the injection site was subcutaneous to the cardiac end of the limbs of the mice.
  • HSP65-MUC1 group 10 ⁇ ⁇ HSP65-MUC1 / 200 ⁇ 1 PBS / mouse; BCG + HSP65-MUC1 group: lO g HSP65 - MUC1 + 1.32mg BCG / 200W PBS / mouse; PBS group: 200 ⁇ 1 PBS / Mouse; BCG group: 1.32mg BCG /200 ⁇ 1 PBS / mouse; Positive control group: Positive control mice were injected with carboplatin every 4 days (produced by Qilu Pharmaceutical Factory, China), intraperitoneal injection of 0.5mg carboplatin / only Mice were injected four times in succession.
  • mice On the 16th day, a second injection was performed on each group of mice, and the dose and site were injected with the first immunization. The state of the mice was observed daily, and the growth of the tumor was started. When the PBS control group began to die, or when less than 20% of the PBS control mice had a tumor weight of less than 0.4 g, the mice were killed. The tumor weight of each group of mice was weighed.
  • the tumor weights of the BCG+HSP65-MUC1 group and the HSP65-MUC1 group and other groups are shown in Fig. 4.
  • BCG+HSP65-MUC1 group The injection dose was 1.32 mg BCG+ l ( ⁇ g HSP65-MUC1 I 200 ⁇ 1 PBS I mice; PBS injection group: 200 ⁇ 1 PBS I mice; HSP65-MUC1 group: l ( ⁇ g HSP65- MUC1 I 200 ⁇ 1 PBS I mice; BCG group: 1.32mg BCG/200W PBS I mice.
  • the spleen and lymph node cells of the mice were isolated as effector cells.
  • CTL activity was determined by conventional methods (C. Stremmel, EA Greenfield, E. Howard, GJ. Freeman and VKKuchroo. B7-2 Expressed on EL4 lymphoma Suppresses Antitumor Immunity by an Interleukin4-dependent Mechanism. J. Exp. Med, 1999, 189(6): 919-930. ). 2.
  • mice Seven-week-old C57BL/6 female mice were immunized on days 0, 14, 28, according to different groups. The experimental mice were divided into 4 groups, 10 mice in each group: PBS group, HSP65-MUC1 group, BCG group,
  • BCG+HSP65-MUC1 group Immune site: subcutaneous concentric end of mouse limbs, dose: PBS group, 200 ⁇ 1 PBS I mice; HSP65-MUC1 group, 10 ⁇ HSP65-MUC1 I 200 ⁇ 1 PBS I mice; BCG group, 1.32mg BCG /200 ⁇ 1 PBS / mice; BCG + HSP65-MUC1 group, 10 ⁇ ⁇ HSP65 - MUC1 + 1.32 mg BCG / 200 W PBS I mice.
  • mice were subcutaneously inoculated into the right side of the mouse to inoculate 1.5 ⁇ 10 5 I 200 ⁇ 8 I mice of the B16 cells transfected with the gene encoding the MUC1 epitope antigen polypeptide.
  • the state of the mice, the growth of the tumor, and the body weight of the mice were observed daily.
  • the mice were killed and the tumor weight of each group was weighed.
  • a comparison of tumor weights between groups is shown in Figure 6.
  • the sequence of primers is:
  • the sequence of the product synthesized in the first round of PCR is:
  • primer 2 The sequence of primer 2 is -
  • the sequence of primer 3 is:
  • primer 3' The sequence of primer 3' is:
  • the sequence of the product is: TCTGACT3' (SEQ ED NO: 20)
  • the sequence of the bow 4 is: 5' GAATTCGACGAAGCATACGTTATGGCT GGTGTTGGTTCCCCG 3' (SEQ ID NO: 21)
  • sequence of primer 4' is 5' AAGCTTACCTTGCAGAGTCAGAGAGTAAGC 3YSEQ ID NO:
  • This sequence represents the HER2 multi-epitope antigen gene.
  • the gene encoding heat shock protein 65 (HSP65) and the pET28a (Novagen) vector were digested with NcoI and ECORI, respectively, rrc, 2h.
  • the digested product was separated by agarose gel electrophoresis and the digested product was recovered (recycling kit was produced by Beijing Dingguo Biotechnology Co., Ltd., and the operation was carried out according to the instructions).
  • the conditions for electrophoresis were : 1% agarose gel, ⁇ buffer, 150-200 mA, electrophoresis 0.5-lh.
  • HSP65 heat shock protein 65
  • pET28a-HSP65-HER2 The gene encoding heat shock protein 65 (HSP65) digested with NcoI and EcoRI and the pET28a vector 05 001715 Connected with ligase.
  • the ligation product was transformed into E. coli JM109, take positive transformants were cultured, plasmids were extracted (see above), using the dideoxy termination method, inserts were sequenced (ABIPri S m310 TM, USA; ).
  • the HER2 polyepitopic antigen gene (as described in Example 9) and the pET28a vector (pET28a-HSP65) inserted with the HSP65 gene were separately digested with EcoRI and Hindlll for two hours at 37 °C. Digested product The digested product was separated and recovered by agarose gel electrophoresis as above (recycling kit was produced by Beijing Dingguo Biotechnology Co., Ltd., and the operation was carried out according to the instructions).
  • the HER2 polyepitopic antigen gene digested with EcoRI and Hindlll and the pET28a vector into which the HSP65 gene was inserted were ligated with T4 DNA ligase.
  • the product pET28a(+)-HSP65-HER2 plasmid was ligated into the host strain JMI09, and the positive transformant was cultured.
  • Lane 1 HER2 multi-epitope antigen gene fragment (315 bp) obtained after digestion with EcoRI and Hindlll
  • Lane 2 molecular weight scale (Takara) 2000 bp; lOOOObp; 750 bp; 500 bp; 250 bp; lOObp
  • Plasmids were extracted according to conventional methods (see above), to obtain a plasmid containing the HSP65-HER2 gene, (ABIPrism310 TM, USA) HSP65-HER2 gene sequence was measured by the sequencer, the results show that the obtained HSP65 - HER2 fusion gene and The HSP65-HER2 gene sequence designed by the present invention is completely identical.
  • the nucleotide sequence of HSP65-HER2 is shown in SEQ ⁇ ) NO: 24.
  • the expression plasmid BL21 DE3 (Novagen, America) containing the recombinant plasmid pET28a-HSP65-HER2 was picked and inoculated into a 250 ml Erlenmeyer flask containing 50 ml of LB medium, and cultured in a 37 ° C water bath to an OD 600 value of 0.6. IPTG was added to a final concentration of 0.4 mM, and cultured in a 37 ° C water bath for 2-3 h. The flask containing the expression bacteria was placed on ice for 5 min and centrifuged at 4 ° C for 5 min (5000 x g). Aspirate the supernatant, collect the bacteria, use immediately or freeze.
  • the recombinant fusion protein HSP65-HER2 was purified by the following method. '
  • the fermented cells were resuspended using Solution A (50 mM NaCK 20 mM sodium acetate pH 6.0), ultrasonically lysed, and the supernatant was collected by centrifugation.
  • Solution A 50 mM NaCK 20 mM sodium acetate pH 6.0
  • Solution A 50mM NaCK 20mM sodium acetate pH6.0
  • Solution B 20 mM sodium acetate, 1 M NaCl pH 6.0
  • Phenyl Sepharose FF Phenyl Sepharose FF (Pharmingen)
  • Solution F 1.5M NaCK 20mM Tris, pH7.0
  • Recombinant fusion protein HSP65-HER2 was obtained after desalting of the sample.
  • Example 14 BCG and HSP65-HER2 induce activation and maturation of human dendritic cells in vitro I. Separating human peripheral blood mononuclear cells as described in part (a) of Example 4
  • the control group was set up separately: only the culture medium was added; BCG group: BCG was added to a final concentration of 3 ( ⁇ g/ml; BCG+HSP65-HER2 group: BCG and HSP65-HER2 were added to make it The concentrations were 3 (g/ml and 10 ( ⁇ g/ml ; HSP65-HER2 group: HSP65-HER2 was added to achieve a final concentration of 10 (g/ml; positive control group: the following factors were added separately and their final concentrations were respectively 10 ng/ml TNFa, 10 ng/ml IL-6, 10 ng/ml IL- ⁇ and 1 (g/ml PGE2.
  • the above BCG and HSP65-HER2 proteins and cytokines were directly added to immature dendritic cells (DC). Cultivate for 2d.
  • Dendritic cells were harvested on day 7 and assays for activation and maturation were performed.
  • FIG. 10 A comparison of CD86 fluorescence values between groups is shown in Figure 10.
  • the results of flow cytometry showed that BCG+HSP65-HER2 significantly induced DC activation and maturation compared with HSP65-HER2 group.
  • DC is the most important antigen-presenting cell in human body.
  • the activation and maturation of DC indicates that the antigen-presenting ability of DC is enhanced, and the ability to activate cytotoxic killer T lymphocytes (CTL) against HER2 is strengthened; Activated CTLs in vivo kill tumor cells expressing HER2 in vivo and, therefore, can be used to prevent and/or treat HER2-positive tumors.
  • Example 15 A B16 (C57BL/6 mouse melanoma cell line) cell line stably expressing the HER2 multi-epitope antigen gene was obtained.
  • the PCR reaction was carried out using the DNA fragment of SEQ ID NO: 24 as a template: the sequence of primer 5 was: 5, -CTGCAGGATCCATGGACGAAG CATACGTTA TG GC-3, (SEQ ID NO: 25)
  • the sequence of the bow I, 5 is: 5'-GCGGCCGCAAGCTTAAGATCTACCTTG CAGAGTCAGAGAGT AA-3' (SEQ ID NO: 26)
  • the reaction conditions of PCR were: 94 ° C, 30"; 55 ° C, ⁇ ; 72 ° C, ⁇ , 30 cycles and 72 ° C extension for 10 min.
  • the sequence of the product HER2 polyepitope antigen gene was obtained as follows:
  • the HER2 polyepitopic antigen gene (SEQ ID NO: 27) and the eukaryotic expression plasmid pcDNA3 (Invitrogen) were digested with restriction endonucleases BamHI and Notl, respectively.
  • the digestion conditions were 37 ⁇ 2 hours, and the avian digestion product was separated by agarose gel electrophoresis as above and the digested product was recovered.
  • HER2 digested with BamHI and Notl and the eukaryotic expression plasmid pcDNA3 treated with the same enzyme were ligated with ligase to obtain pcDNA3-HER2.
  • mice in the BCG+HSP65-HER2 group was significantly lower than that in the HSP65-HER2 group.
  • BCG can significantly enhance the specific anti-tumor effect induced by HSP65-HER2 injection and inhibit the growth of HER2-positive tumors.
  • Example 17 BCG Enhancement HSP65-HER2 Induction HER2-specific CTL
  • BCG can significantly enhance the HER2-specific CTL induced by HSP65-HER2 and inhibit the growth of HER2-positive tumor cells.
  • Example 18 BCG Enhancement Specific Antitumor Effect Induced by HSP65-HER2 Immunization
  • Example 8 Except that HSP65-HER2 was substituted for HSP65-MUC1 of Example 8 (1), and B16 cells transfected with the HER2 epitope antigen polypeptide-encoding gene were replaced by B16 cells of Example 8 (1), according to the implementation. Part (1) of Example 8 was tested.
  • mice in the BCG+HSP65-HER2 group was significantly lower than that in the HSP65-HER2 group.
  • a comparison of tumor weights between groups is shown in Figure 13.
  • BCG can significantly enhance the specific anti-tumor effect induced by HSP65-HER2 immunization, and can significantly inhibit the growth of HER2-positive tumor cells in mice.
  • Example 19 Construction of heat shock protein 65 fusion gene with prostate specific antigen
  • HSP65 prostate specific antigen
  • CTL cytotoxic T lymphocyte
  • the 5' end primer sequence is: 5'-CCATG GCC AAG ACA ATT GCG-3' ( SEQ ID NO: 28);
  • the sequence of the 3' primer is:
  • the reaction conditions are: 94 ° C, 30"; 55 ° C, ⁇ ; 72 ° C, 2', 30 cycles and 72 ° C extension for 10 minutes.
  • the obtained product is the HSP65-PSA fusion gene, see the sequence SEQ ID NO: 30.
  • the PCR product was digested with Ncol and Hindlll at 37'C for 2 hours.
  • the digested product is separated by agarose gel electrophoresis, and the digested product is recovered (the recovery kit is produced by Beijing Dingguo Biotechnology Co., Ltd., and the operation is carried out according to the instructions).
  • the recovered PCR product was cloned into prokaryotic expression expressed by restriction endonucleases Ncol and Hindlll.
  • the pET-28a (+) plasmid (Novagen, USA) has a 6-histidine codon upstream.
  • the recombinant pET-28a(+)-HSP65-PSA plasmid containing the HSP65 and PSA fusion gene was transformed into Escherichia coli. Take positive transformant culture.
  • the plasmid containing the HSP65-PSA gene was obtained by a conventional method for extracting a plasmid (see above), and the gene sequence of HSP65-PSA was determined by a sequencer (ABIPrism310TM, USA).
  • the nucleotide sequence of HSP65-PSA is shown in SEQ ID NO:30.
  • the recombinant HSP65-PSA fusion protein was expressed in a 10 liter fermentor with lactose as an inducer. After the cleavage, the recombinant HSP-PSA fusion protein was obtained by nickel affinity chromatography, desalting, ion exchange chromatography, and desalting. The purity of the gene fusion protein of the T cell epitope of HSP-65 and prostate specific antigen (PSA) was determined by conventional SDS-PAGE method: 98%.
  • Human peripheral blood mononuclear cells are isolated as described in part (a) of Example 4.
  • Example 4 Inducing immature dendritic cells.
  • control group was set up separately: only the culture solution was added; BCG group: BCG was added to make the final concentration reach
  • BCG + HSP65-PSA group BCG and HSP65-PSA were added to give a final concentration of 3 ( ⁇ g/ml and 10 ( ⁇ g/ml ; HSP65-PSA group: HSP65-PSA was added)
  • the final concentration was 10 ( ⁇ g/ml ; positive control group: the following factors were added to make the final concentration of 1 Ong/ml TNFa, 1 Ong/ml IL-6, lOng/mllL-1 ⁇ and 1 ( ⁇ g/ml respectively).
  • PGE2 The above BCG and HSP65-PSA proteins and cytokines were directly added to immature dendritic cells (DC) for 2 days. 2. Harvested dendritic cells on day 7 and assayed for activation and maturation indicators.
  • FIG. 15 A comparison of CD86 fluorescence values between groups is shown in Figure 15.
  • the results of flow cytometry showed that BCG+HSP65-PSA significantly induced DC activation and maturation compared with HSP65-PSA group.
  • DC is the most important antigen-presenting cell in human body.
  • the activation and maturation of DC indicates that the antigen-presenting ability of DC is enhanced, and the ability to activate cytotoxic killer T lymphocytes (CTL) against PSA is strengthened; Activated CTLs in vivo kill PSA-positive tumor cells in vivo and, therefore, can be used to prevent and or treat PSA-positive tumors.
  • CTL cytotoxic killer T lymphocytes
  • pcDNA3-GFP_PSSA plasmid HSP65-PSA as template, 5'-end primer 5 '-GGATCC ATGGAGCCTGAAGAGTTC-3 " (SEQ ID NO: 31), 3' primer 5'-GGGTCGACTCTAGATCAACACAGCATGAATTT-3' ( SEQ ID NO: 32),
  • PCR conditions 94. C, 30"; 60 ° C, l' ; 72 ° C, ⁇ 30", after 27 cycles, 72 ° C extension for 10 min.
  • the pcDNA3-GFP plasmid and PCR product were separately digested with BamHI and Xbal, and the digested product was separated and recovered by agarose gel electrophoresis (recovery kit was produced by Beijing Dingguo Biotechnology Co., Ltd., operation was carried out according to the instructions), and T4 DNA ligase was used.
  • the recovered digested product is ligated to obtain pcDNA3-GFP-PSA.
  • BCG can significantly enhance the specific anti-tumor effect induced by HSP65-PSA injection and inhibit the growth of PSA tumor cells.
  • Example 25 BCG Enhancement HSP65-PSA Induction PSA Specific CTL Effect
  • HSP65-PSA replace HSP65-MUC1 in part (1) of Example 7, with PSA form
  • the B16 cells transfected with the epitope-encoding polypeptide and the B16 cells not transfected with the PSA epitope antigen-encoding gene are used as target cells in place of the target cells in part (1) of Example 7, in accordance with the portion of Example 7 (1) ) conduct experiment.
  • primer 1 (SEQ ID NO: 33): -
  • the sequence of the primer ⁇ is (SEQ ID NO: 34):
  • the upstream primer has a BamH I cleavage site at the 5' end and a Kpnl, Bgl II, Hind III cleavage site at the 3' end of the downstream primer.
  • SEQ ID NO: 47 is the HPV-Tet sequence. 2.
  • the PCR product was cloned into the vector plasmid pMD-18T (Takara).
  • Lane 2 pMD18T-HPV plasmid was digested, and the arrow showed the HPV gene fragment.
  • the 5' end primer sequence is (SEQ ID NO: 48): 5 ' AAGCTTACTCCGACTCTGC AC-3 '
  • the 3' end primer sequence is (SEQ ID NO: 49): 5'-GAATTCTCACTCCAGTTTCTTCAG-3', wherein the 5' end primer carries The EcoR I cleavage site and the 3' primer have a Hind III restriction site.
  • the 5' primer has a Hind m restriction site and the 3' primer has an EcoR I restriction site.
  • the PCR conditions were: 94 ° C 30 sec, 55 ° C 30 sec, 72 ° C 30 sec, 25 cycles. After the end of the cycle, 72 0 C extended for 10 min.
  • HSP65 heat shock protein 65
  • pET28a Novagen vector
  • the gene encoding heat shock protein 65 (HSP65) and pET28a (Novagen) vector were digested with NcoI and EcoRI, respectively, at 37 ° C for 2 h.
  • the digested product was separated by agarose gel electrophoresis and the digested product was recovered as above (the recovery kit was produced by Beijing Dingguo Biotechnology Co., Ltd., and the operation was carried out according to the instructions).
  • the gene encoding the heat shock protein 65 (HSP65) digested with NcoI, EcoRI and the pET28a vector were ligated with a ligase.
  • the ligation product pET28a-HSP65 was transformed into the host strain JM109, the positive transformant was cultured, the plasmid was extracted (see above), and the insert was sequenced by the dideoxy terminator method (ABIPrism 310TM, USA).
  • the recovery kit is produced by Beijing Dingguo Biotechnology Co., Ltd., and the operation is carried out according to the instructions).
  • the recovered product was ligated with T4 DNA ligase.
  • the ligation product pET28a-HSP65-HPV was transformed into the host strain JM109, and the positive transformant was cultured, the plasmid was extracted (see above), and the insert was sequenced by the dideoxy terminator method (ABIPrism 310TM, USA).
  • the resulting HSP65-HPV fusion gene is identical to the designed gene sequence.
  • Lane 1 pET-28-HSP65-HPV digestion fragment
  • Lane 2 DNAMarker
  • the recombinant HSP65-HPV fusion protein was expressed in a 10 liter fermentor with lactose as an inducer. After the cleavage, the recombinant HSP-HPV fusion protein was obtained by nickel affinity chromatography, desalting, ion exchange chromatography, and desalting. Sampling was performed by HPLC analysis, and the purity of the recombinant HSP65-HPV fusion protein was 96.1%.
  • control group was set up separately: only the culture solution was added; BCG group: BCG was added to make the final concentration reach
  • BCG + HSP65 -HPV group BCG HSP65-HPV, and the final concentration of 30 g / ml and 100 ⁇ ⁇ / ⁇ 1; HSP65-HPV group: HSP65-HPV final concentration of lOO g /ml; Positive control group: The following factors were added to make the final concentration of 1 Ong/mlT Fa, 1 Ong/mlIL-6, 1 Ong/mlEL-1 ⁇ and ⁇ t ⁇ g ni PGE2.
  • the above BCG and HSP65-HPV fusion proteins and cytokines were directly added to immature dendritic cells (DC) for further 2 days.
  • FIG. 21 A comparison of the fluorescence values of CD86 in each group is shown in Figure 21.
  • the results of flow cytometry showed that BCG+HSP6 5-HPV significantly induced DC activation and maturation compared with HSP65-HPV group.
  • DC is the most important antigen-presenting cell in human body.
  • the activation and maturation of DC indicates that the antigen-presenting ability of DC is enhanced, and the ability to activate cytotoxic killer T lymphocytes (CTL) against HPV is enhanced; CTL kills cells infected with HPV in vivo and, therefore, can be used to prevent and or treat HPV infection.
  • CTL cytotoxic killer T lymphocytes
  • Example 31 Construction of a eukaryotic expression plasmid for recombinant HPV antigen multi-epitope gene
  • Example 27 The pMDIS-T-HPV antigen multi-epitope gene recombinant plasmid constructed in Example 27 was used as a template, and the HPV antigen polyepitope gene restriction site was replaced by PCR to obtain a subcloning into a eukaryotic expression plasmid. HPV antigen multi-epitope gene.
  • the 5' primer primer sequence is (SEQ ID NO: 51):
  • the 3' primer sequence is (SEQ ID NO: 52):
  • the 5' primer has a Hind cleavage site
  • the 3' primer has an EcoR I restriction site.
  • the product sequence of the PCR synthesis was as shown in SEQ ID NO: 53, which was 360 bp in total.
  • the PCR product was cloned into the pcDNA3 plasmid: the PCR product was subcloned using Hind III and EcoR I. To the pcDNA3 plasmid. Transformed into E. coli JM109, take positive transformants were cultured, plasmids were extracted (see above), using the dideoxy termination method, inserts were sequenced (ABn> rism310 TM, USA) ) 0
  • CTL can kill HPV-infected cells.
  • Example 34 BCG Enhancement Specific killing by HSP65-HPV immunization HPV-infected cells
  • primer 1 The sequence of primer 1 is (SEQ ID NO: 54):
  • the sequence of the primer ⁇ is (SEQ ID NO: 55):
  • the sequence of the first round of PCR synthesis product is SEQ ID NO: 56
  • the sequence of primer 2 is (SEQ ID NO: 57):
  • sequence of primer 2' is (SEQ ID NO: 58):
  • the primer 3 sequence is (SEQ ID NO: 60):
  • sequence of primer 3' is (SEQ ID NO: 61):
  • sequence of the product is shown as SEQ ED NO: 62.
  • the primer 4 sequence is (SEQ ID NO: 63):
  • sequence of primer 4' is (SEQ ID NO: 64):
  • the sequence of the product is shown in SEQ ID NO: 65.
  • the 4th end of the PCR product has an EcoRI cleavage site at the 5' end and Hind III and Xhol cleavage sites at the 3' end of the downstream primer.
  • This sequence is a copy of an optimized SAS coronavirus epitope antigen sequence.
  • the PCR reaction conditions were: 94 ° C, 30"; 55 ° C, ; 72 ° C, 2', after 30 cycles, 72 ° C extension for 10 min.
  • the PCR product was cloned into the TA vector by TA cloning method to obtain pMD-18T-SARS/3CL 161.
  • 264 plasmid as described in the kit (Katara, Takara).
  • the positive transformant was cultured, the plasmid was extracted (see above), and the insert was sequenced by the dideoxy terminator method.
  • Example 36 HSP65-SARS / 3CL 161 - 264 of the fusion gene, expression and purification
  • the pMD-18T-SARS/ 3CL 161-264 plasmid constructed in Example 35 was digested with EcoRI and Hindlll enzymes and recovered.
  • Escherichia coli BL DE 3 Novagen containing HSP65-SA S/3CL 161 . 264- encoding gene expression plasmid was cultured in a 10-liter fermentor with lactose as an inducer. After the cleavage, HSP65-SA S/ 3CL 161-264 was obtained by nickel affinity chromatography, desalting, ion exchange chromatography and desalting. 10 ⁇ l samples were loaded for 12% SDS-PAGE. After Coomassie blue staining, HSP65-SA S /3CL 161 . 264 appeared as a single zone with no bands. A 10 ⁇ l sample was taken for HPLC analysis, and the purity of HSP65-SARS/ 3CL 161-264 was 96.1%.
  • HSP65-SARS/ 3CL 161-264 SDS-PAGE and Western Blot identification of HSP65-SARS/ 3CL 161-264 are shown in Figure 26: HSP65-SARS/ 3CL 16 i -264 SDS-PAGE identification and Western blot analysis.
  • Human peripheral blood mononuclear cells are isolated as described in part (a) of Example 4.
  • the control group was set up separately: only the culture medium was added; BCG group: BCG was added to make the final concentration reach 30 g/ml; BCG+HSP65-SA S group: BCG and HSP65-SARS were added to make the final concentration. respectively 30 ⁇ ⁇ / ⁇ 1 and 10 ( ⁇ g / ml; HSP65 -SARS group: HSP65-SA S was added at a final concentration of 10 ( ⁇ g / ml; positive control group: The following factors were added and the final concentration It is 10 ng/ml TNFa, 10 ng/ml IL-6, lOng/ml IL- ⁇ and 10 ⁇ ⁇ / ⁇ 1 PGE2.
  • the above BCG and HSP65-SARS proteins and cytokines are directly added to immature dendritic cells (DC) to continue culture. 2d.
  • Example 4 Determination of dendritic cell activation and maturation indicators.
  • FIG. 27 A comparison of the CD86 fluorescence values between groups is shown in Figure 27.
  • the results of flow cytometry showed that 'BCG+HSP65-SA S can significantly induce DC activation and maturation compared with HSP65-SARS group.
  • DC is the most important antigen-presenting cell in human body.
  • the activation and maturation of DC indicates that the antigen-presenting ability of DC is enhanced, and the ability to activate cytotoxic killing lymphocytes (CTL) against SARS is enhanced; CTL kills SARS-infected cells in vivo and, therefore, can be used to prevent and or treat SARS infections.
  • CTL cytotoxic killing lymphocytes
  • Example 38 Construction of eukaryotic expression plasmid of recombinant SARS/3CL 16 264 epitope antigen gene
  • SARS/3CL 161 The 264 gene was constructed downstream of the GFP gene, and the GFP and SARS/3CL 161 . 264 genes were in the same reading frame, and the recombinant p C DNA3- GFP-SARS/3CL 16 ⁇ 6 4 eukaryotic cells were obtained. Expression plasmid.
  • the mouse melanoma cell line B16 was transfected with this plasmid, and transfected cells expressing GFP-SARS/ 3CL 161-264 were obtained. This cell was used as a cell model for SARS Cov infection to detect HSP65-SARS/3CL. Activity of 161-264 .
  • PcDNA3-GFP-SARS/3CL 161 . 264 transfected B16 cells were identified, and positive clones were identified by confocal microscopy and FACS.
  • Stable transfected GFP-SA S/3CL, 61 .264 gene mouse melanoma B16 The cells showed green color under confocal microscopy, and the FACS test showed strong green fluorescence in the transfected cells.
  • Example 7 In addition to replacing HSP65-MUC1 in part (1) of Example 7 with HSP65-SA S, B16 cells transfected with the SARS epitope antigen polypeptide encoding gene and B16 transfected with the SARS epitope antigen polypeptide encoding gene The experiment was carried out in accordance with the portion (1) of Example 7 except that the target cell was replaced with the target cell in the fraction (1) of Example 7 as a target cell. 2. Results: Compared with the HSP65-SA S group, when the B16 cells transfected with the SARS epitope antigen polypeptide encoding gene were used as target cells, the killing rate of spleen and lymph node CTL in the BCG+HSP65-SA S immunized group was 46%. .
  • mice immunized with HSP65-SARS fusion protein had a CTL killing rate of 38%. This suggests that BCG can enhance the role of specific CTL induced by HSP65-SARS immunization.
  • a comparison of the effects of each group of induced specific CTLs is shown in Figure 30.
  • BCG can enhance the specific CTL induced by HSP65-SARS, and the induced specific CTL can kill HPV infected cells.
  • Example 41 BCG Enhancement Specific Killing by HSP65-SARS Immunization The Role of SARS Infected Cells
  • BCG can significantly enhance the specific killing of SARS-infected cells stimulated by HSP65-SARS immunization.
  • Example 42 Acquisition of multi-epitope hepatitis B core antigen gene
  • HBcAg multi-epitope hepatitis B core antigen gene
  • primer 1 The sequence of primer 1 is:
  • the sequence of the primer ⁇ is -
  • sequence of the product obtained in the first round of PCR is shown in SEQ ID NO:68.
  • primer 2 The sequence of primer 2 is:
  • primer 2' The sequence of primer 2' is:
  • SEQ ID NO: 71 The sequence of the product obtained by the second round of PCR is shown in SEQ ID NO: 71, which represents the hepatitis B core antigen multi-epitope gene.
  • HSP65 heat shock protein 65
  • the gene encoding heat shock protein 65 (HSP65) was digested with Ncol and EcoRI, and the multi-epitope hepatitis B virus core antigen gene was digested with EcoRI and Hindlll at 37 ° C for 2 hours.
  • the digested product was separated by agarose gel electrophoresis as above and the digested product was recovered.
  • HSP65-HBcAg (abbreviated as HSP-HBV) was obtained by ligating the gene encoding the heat shock protein 65KD (HSP65) digested with restriction endonuclease and the multi-epitope hepatitis B virus core antigen gene with T4 DNA ligase.
  • the nucleotide sequence of HSP-HBV is shown in SEQ ID NO:72.
  • the ligation product obtained in Example 43 was digested with Ncol and Hindlll at 37 ° C, and cloned into the prokaryotic expression vector pET-28a (+) plasmid (Novagen, USA) digested with restriction endonucleases Ncol and Hindlll. Upstream of the histidine codon, pET-28a(+)-HSP65-HBcAg was obtained.
  • Single colony bacteria were inoculated into 50 ml of LB medium and shaken in a 250 ml Erlenmeyer flask in a 37 Q C water bath to an OD600 value of 0.6. IPTG was added to make the bee concentration 0.4 mM, and cultured in a 37 ° C water bath for 2-3 hours. The flask was placed on ice for 5 minutes and centrifuged at 4 G C for 5 minutes (5000 xg). Aspirate the supernatant, collect the bacteria, use immediately or freeze.
  • the above Escherichia coli cells (3 g wet weight) containing the HSP-HBV recombinant fusion protein were thawed at room temperature, resuspended in 20 ml of the binding solution, and after ice bath for 2 hours, centrifuged for 20 minutes (10000 X g), and the supernatant was collected for use.
  • the supernatant is added to a nickel affinity chromatography column (lXlOcm), and the target protein is eluted after washing.
  • lXlOcm nickel affinity chromatography column
  • HSP-HBV recombinant fusion protein HSP-HBV recombinant fusion protein
  • Human peripheral blood mononuclear cells are isolated as described in part (i) of Example 4. 2. Inducing immature dendritic cells as described in part (ii) of Example 4.
  • control group was set up separately: only the culture medium was added; BCG group: BCG was added to a final concentration of 3 ( ⁇ g/ml; BCG + HSP-HBV group: BCG and HSP-HBV were added to make it concentrations of 3 ( ⁇ g / ml and 100 ⁇ ⁇ / ⁇ 1; HSP-HBV group: HSP-HBV was added to a final concentration of lOO g / ml; positive control group: The following factors were added to a final concentration of 10ng / mlTNFa, 10 ng/ml IL-6, lOng/ml IL- ⁇ and l ( ⁇ g/ml PGE 2 . The above BCG and HSP-HBV proteins were directly added to immature dendritic cells (DC) for 2 days.
  • DC dendritic cells
  • FIG. 34 A comparison of CD86 fluorescence values between groups is shown in Figure 34.
  • the results of flow cytometry showed that BCG+HSP65- HBcAg significantly induced DC activation and maturation compared with HSP-HBV group.
  • DC is the most important antigen-presenting cell in human body.
  • the activation and maturation of DC indicates that the antigen-presenting ability of DC is enhanced, and the ability to activate cytotoxic killer T lymphocytes (CTL) against HBV is enhanced; CTL kills cells infected with HBV in vivo and, therefore, can be used to prevent and or treat HBV infection.
  • CTL cytotoxic killer T lymphocytes
  • the hepatitis B virus core antigen gene fragment (obtained in Example 38) was cloned into the transfection vector pcDNA3-GFP (Invitrogen) using EcoRI and Hindlll restriction sites to form pcDNA3-GFP-hepatitis B virus core antigen. Recombinant plasmid (pcDNA3 - GFP - HBV).
  • BCG can enhance the specific killing of HBV-infected cells stimulated by HSP-HBV injection.
  • Example 50 BCG Enhances HSP65-HBcAg Inducing HBV-Specific Killer T Lymphocytes in Mice 1.
  • Method: In addition to replacing HSP65-MUC1 in part (1) of Example 7 with HSP-HBV, HBV epitope antigen B16 cells transfected with the polypeptide-encoding gene and B16 cells transfected with the HBV epitope antigen-encoding gene were used as target cells instead of the target cells in part (1) of Example 7, according to part (1) of Example 7 experiment. 2.
  • mice in the BCG+HSP-HBV group was significantly lower than that in the HSP-HBV group. This indicates that BCG can enhance the specific killing of HBV-infected cells stimulated by HSP65-HBcAg immunity. The effect of specific killing of HBV-infected cells stimulated by each group is shown in Figure 37.
  • primer 1 The sequence of primer 1 is:
  • reaction conditions for PCR are: 94 ° C, ⁇ ; 60 ° C, 1'; 72 ° C, 2', after 3 cycles? , extending at 72 ° C for 10 minutes.
  • the sequence of the first round of PCR synthesis product is shown in SEQ ID NO:75.
  • primer 2 The sequence of primer 2 is:
  • reaction conditions for PCR are: 94 ° C, r; 60 ° C, 1 '; 72 ° C, 2 ', after 30 cycles, 72 ° C extension
  • the sequence of the product is shown in SEQ ID NO : 78, which is the HCV core antigen gene.
  • the PCR product was cloned into the vector plasmid pMD18-T (Takara): The PCR product (HCV core antigen gene) was cloned into the TA vector by TA cloning method, and the obtained product was pMD-18T-HCV. See the TA Linkage Kit (Promega) for the method. After transformation of the host strain JM109, positive clones were taken for culture, and the extract was extracted by a conventional method (see above), and the insert was sequenced by the dideoxy terminator method (A ⁇ n>rism310TM, USA) o Example 53 Cloning of HSP65 gene
  • the pET28(a)-HSP65 recombinant plasmid and the pMD-18T-HCV recombinant plasmid were digested with EcoRI and Hindlll (Takara), respectively, for 37 hours.
  • the digested product was separated by agarose gel electrophoresis and the digested product was recovered as above (the recovery kit was produced by Beijing Dingguo Biotechnology Co., Ltd., and the operation was carried out according to the instructions).
  • Lane 2 HCV multi-epitope gene fragment (about 300 bp in length).
  • Example 55 Expression of HSP65-HCV fusion gene
  • E. coli cells were thawed at room temperature. Each gram of bacteria (wet weight) was resuspended in 10 ml of lysis buffer. Add 1 M MgS0 4 lOul, 10 ug/ml DNase 1 10 ul per ml of lysis buffer. Place on ice for 30 min.
  • the eluted target protein was subjected to phenyl Sepharose hydrophobic chromatography (chromatographic medium purchased from Pharmacia) to remove endotoxin.
  • the salt in the protein of interest was removed using Sephadex-G-25 ion exchange chromatography (chromatographic medium purchased from Pharmacia).
  • Lanes 1 and 2 are HSP65-HCV proteins expressed in E. coli.
  • Lanes 3 and 4 are purified HSP65-HCV proteins.
  • Example 57 BCG and HSP65-HCV induce activation and maturation of human dendritic cells in vitro
  • Human peripheral blood mononuclear cells are isolated as described in part (i) of Example 4. 2. Inducing immature dendritic cells as described in part (ii) of Example 4.
  • control group was set up separately: only the culture medium was added; BCG group: BCG was added to a final concentration of 3 ( ⁇ g/ml ; BCG+HSP65-HCV group: BCG and HSP65-HCV were added to make it concentrations of 30 ⁇ ⁇ / ⁇ 1 and lOO g / ml; HSP65-HCV group: HSP65-HCV was added to a final concentration of lOO g / ml; positive control group: The following factors were added to a final concentrations of 10ng / ml TNFa > 10 ng/ml IL-6, lOng/ml IL- ⁇ and l ( ⁇ g/ml PGE 2 .
  • the above BCG and HSP65-HCV proteins were directly added to immature dendritic cells (DC) for 2 days.
  • FIG. 40 A comparison of CD86 fluorescence values between groups is shown in Figure 40.
  • the results of flow cytometry showed that BCG+HSP65-HCV significantly induced DC activation and maturation compared with HSP65-HCV group.
  • DC is the most important antigen-presenting cell in human body.
  • the activation and maturation of DC indicates that the antigen-presenting ability of DC is enhanced, and the ability to activate cytotoxic killer T lymphocytes (CTL) against HBV is enhanced; CTL kills cells infected with HCV in vivo and, therefore, can be used to prevent and or treat HCV infection.
  • CTL cytotoxic killer T lymphocytes
  • Example 47 I and Xho l (Takara) were digested, and the linearized vector was separated and recovered by agarose gel electrophoresis; the pMD-18T-HCV recombinant plasmid obtained in Example 47 was also digested with BamH I and Xho l to recover the HCV DNA fragment. The two were ligated with T4 DNA ligase and transformed into competent bacteria JM109. The positive recombinant plasmid was extracted (the method is the same as before).
  • mice in BCG+HSP65-HCV group was significantly lower than that in HSP65-HCV group.
  • Test, P ⁇ 0.05 indicating that BCG can enhance the specific killing of HCV-infected cells stimulated by HSP65-HCV injection.
  • a comparison of the effects of specific killing of HCV-infected cells stimulated by each group is shown in Figure 41.
  • BCG can enhance the specific killing of HCV-infected cells stimulated by HSP65-HCV injection.
  • mice in the BCG+HSP65-HCV group was significantly lower than that in the HSP65-HCV group. This indicates that BCG can enhance the specific killing of HCV-infected cells stimulated by HSP65-HCV immunization. The effect of specific killing of HCV-infected cells stimulated by each group is shown in Figure 43.
  • BCG can enhance the specific killing of HCV-infected cells stimulated by HSP65-HCV immunization.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Communicable Diseases (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Description

包含重组融合蛋白及佐剂的疫苗组合物及其应用
技术领域
本发明涉及用于预防和治疗肿瘤的包括重组融合蛋白 CTL疫苗及其佐剂卡介苗的 疫苗组合物,用于预防和治疗病毒感染疾病的包括重组融合蛋白 CTL疫苗及其佐剂卡介 苗的疫苗组合物, 特别是涉及用来预防和治疗人乳腺癌、 直肠癌、 结直肠癌、 喉癌、 胃癌、 膀胱癌、 胰腺癌、 前列腺癌、 卵巢癌、 结肠癌、 肺癌等的人类肿瘤, 以及用来 预防和治疗 SARS病毒感染疾病、 乳头瘤病毒感染、 宫颈癌、 丙型肝炎、 乙型肝炎、 丙 型肝炎病毒感染和乙型肝炎病毒感染导致的肝癌、 肝硬化等病毒感染所引起的疾病的 包括重组融合蛋白 CTL疫苗及其佐剂 BCG的疫苗组合物。 背景技术
肿瘤、病毒感染所引起的疾病都是世界范围内严重危害人们健康的疾病, 攻克这些 疾病是人类面临的重大难题。
特异性的细胞毒性 T淋巴细胞(cytotoxic T lymphocyte, CTL)是人体杀伤肿瘤细胞 和清除体内病毒感染细胞最高效的免疫细胞。
体内 CTL的激活需要未致敏的 T淋巴细胞同时接受抗原递呈细胞提供的两个信号, 一个是外源抗原肽经抗原递呈细胞 (antigen presenting ceIl,APC)加工处理后与 MHCI类 分子结合形成复合物并递呈在 APC的表面, 被 T淋巴细胞的 TCR所识别; 另一个信号是 APC表面表达的协同刺激分子, 即 B7分子的表达, 被丁淋巴细胞的 CD28分子所识别。 只有 T淋巴细胞同时接受 APC所提供的这样两个信号时, 才能被激活。
大量的研究表明: 热休克蛋白 (heat shock protein, HSP) 能够携带它们所融合的蛋 白进入抗原递呈细胞中,并协助所融合的蛋白经过抗原递呈细胞的加工处理后与 MHCI 类分子结合递呈在 APC的表面。为激活特异性的 CTL提供第一信号。而在机体没有感染 时, 抗原递呈细胞只表达自身肽, 而不表达 B7分子, 因此针对自身肽的 T细胞不能被激 活。 所以, 如果 T淋巴细胞只接受了递呈在 APC表面与 MHCI类分子结合的肽这样的第 一信号是不能被激活的; APC还需要给 T淋巴细胞提供第二信号。 通常情况下, 当机体 有微生物入侵时, 抗原递呈细胞开始表达 B7分子。 因此, 如果能够给机体一个类似微 生物的危险信号, 则类似微生物的危险信号就能够刺激 APC表达 B7分子, 为激活 T淋巴 细胞提供第二信号。 (Ruslan Medzhitov and Charles Janeway. Innate Immunity, The New England Journal of Medcine, 2000, 343(5):338-344 )。
卡介苗(BCG)是应用多年的免疫佐剂, 有研究表明 BCG能够通过 Toll样受体激活 天然免疫系统,诱导细胞因子的表达和抗原递呈细胞的表面协同刺激分子 (B7分子)的表 达(Ruslan Medzhitov, 1999)。未致敏的 T淋巴细胞的激活,需要抗原递呈细胞(antigen presenting cell,APC)提供两个信号, 如果 APC只提供第一信号, 会导致免疫耐受。 APC 还必须表达表面协同刺激分子, 即 B7分子, 才能激活未致敏的 T淋巴细胞。根据上述的 机制, 利用 BCG能够刺激 APC表面协同刺激分子表达的作用, 为了提高重组融合蛋白
CTL疫苗激活特异性 CTL的效能,本发明利用卡介苗作为重组融合蛋白 CTL疫苗的 佐剂, 使重组融合蛋白 CTL疫苗进入机体后能够产生较强的刺激特异性 CTL产生的作 用。 卡介苗进入机体后, 机体会识别其为能够引起感染的危险信号, 抗原递呈细胞表 面的 B7分子开始表达, 为激活 CTL提供第二信号。 BCG是减毒的微生物不能引起机体 的感染, 但其仍保留着微生物的保守结构, 通过激活 Toll样受体, 诱导 B7分子的表达, 进而增强 CTL激活的效力和速度 (Ruslan Medzhitov and Charles A. Janeway Jr. Decoding the patterns of self and non-self by the innate immune system. Science,296(5566):298。
Sybille Thoma-Uszynski,Steffen Stenger, Osamu Takkkeuchi et al. Induction of direct antimicrobial activity through mamalian Toll-like receptors. Science, 291(5508): 1544。 Hans D.Brightbill, Danidl H.Libraty, Stephan R.Kruzzik et al. Host defense mechanisms triggered by microbial liporoteins through Toll-like receptors. Science,285(5428):732,)。 机体识别非己微生物主要是通过 To】】样受体识别微生物生理结构中比较保守的分 子(pathogen-associated molecular patterns, PAMPs), 虽然这些 P AMP不是微生物所独有, 却是所有微生物能够产生的。 PAMPs包括革兰式阴性菌的脂多糖 (lipopolysaccharides LPS) , 革兰氏阳性菌的肽聚糖和脂多肽 (peptidoglycans and lipopeptides) , 以及细菌 的鞭毛、 细菌的 DNA、 病毒的双链 RNA。 Toll样受体是由胞外段富含亮氨酸的结构域 和胞内的结构类似于 IL一 1受体的结构域所构成的跨膜蛋白。 TLR主要表达在与免疫功 能相关的组织, 如: 脾、 外周血白细胞以及与外界有接触的组织器官, 如: 肺和胃肠 i¾ o www.InvivoGen.Toll-like <http://www.InvivoGen.Toll-like>') o 迄今为止, 己发现有十种人的 Toll样受体和九种鼠的 Toll样受体, 不同的 Toll样受 体有其各自的配体。 TLR2主要识别各种 PAMPs, 包括细菌脂蛋白、 肽聚糖和脂壁酸。 TLR3参与病毒来源的双链 RNA的识别。 TLR4优先被脂多糖激活。 TLR5能够识别细菌 的鞭毛而非甲基化的 CpG DNA的识别则需要 TLR9。 最近的研究表明: TLR7 和 TLR8 能够识别小的合成的抗病毒分子。 而且, 在很多情况下, Toll样受体发挥作用时需要与 协同受体的协同发挥作用, 如 TLR4发挥作用时常与 MD2和 CD14协同作用。 (www.InvivoGen.Toll-like <http://www.InvivoGen.Toll-like> ) 新近的发现表明 Toll样受体是机体激活天然免疫系统防御微生物反应的必要成分, 是 获 得 性 免 疫 系 统 激 活 的 前 提 (www.InvivoGen.Toll-like <http://www.InvivoGen.Toll-like> )。
发明内容 .
本发明的目的是提供一种以卡介苗作为佐剂的重组融合蛋白疫苗组合物, 其中卡 介苗能够使重组融合蛋白进入机体后产生较强的刺激特异性 CTL的作用。这种疫苗可用 于预防和治疗肿瘤以及病毒感染所引起的疾病。 所述的病毒感染所引起的疾病包括 SARS病毒感染疾病、 乳头瘤病毒感染、 丙型肝炎、 乙型肝炎等。 由于乳头瘤病毒感染 能够导致宫颈癌, 因此, 本发明中的重组融合蛋白亦可用于预防和 /或治疗宫颈癌。 丙 型肝炎病毒感染和乙型肝炎病毒感染的初期会导致感染者的肝脏炎性改变, 随着感染 的反复, 感染者最终出现肝癌、 肝硬化的病理改变, 所以, 本发明中的重组融合蛋白 也可以用于预防和 /或治疗肝癌和肝硬化。
因此, 本发明的一个方面涉及一种疫苗组合物, 其特征在于包括重组融合蛋白和 佐剂, 所述重组融合蛋白是融合了热休克蛋白 65的融合蛋白, 所述佐剂为卡介苗。
具体地说, 上述重组融合蛋白可以是结核分枝杆菌热休克蛋白 65融合粘蛋白 1构成 的重组融合蛋白 HSP65-MUC1、 结核分枝杆菌热休克蛋白 65融合人上皮细胞生长因子 受体 2构成的重组融合蛋白 HSP65-HER2、 结核分枝杆菌热休克蛋白 65融合人前列腺特 异抗原构成的重组融合蛋白 HSP65-PSA、 结核分枝杆菌热休克蛋白 65融合乳头瘤病毒 多表位抗原构成的重组融合蛋白 HSP65-HPV、结核分枝杆菌热休克蛋白 65融合人 SARS 病毒多表位抗原构成的重组融合蛋白 HSP65-SA S或者结核分枝杆菌热休克蛋白 65融 合乙型肝炎核心抗原构成的重组融合蛋白 HSP-HBV、 结核分枝杆菌热休克蛋白 65融合 丙型肝炎抗原表位构成的重组融合蛋白 HSP65-HCV。 上述的重组融合蛋白 HSP65-MUC1、 HSP65-HER2、 HSP65-PSA、 HSP65-HBcAg、 HSP65-HPV HSP65-SARS、 HSP65-HCV为申请人研制开发的产品, 且已申请国家专 利 ( 分 别 参 见 ZL01102614.6, ZL01136347.9, 01134935.2, 200410070084.2, 200410074742.5, 200410074743. X, 02122116.2)。
本发明的另一个方面涉及疫苗组合物在制备预防和治疗肿瘤的药物中的用途, 其 可用于人乳腺癌、 直肠癌、 结直肠癌、 喉癌、 胃癌、 膀胱癌、 胰腺癌、 前列腺癌、 卵 巢癌、 结肠癌、 肺癌等的预防和治疗。
本发明的另一个方面涉及疫苗组合物在制备预防和治疗人病毒感染性疾病的药物 中的用途, 其可用于人 SA S病毒感染疾病、 乳头瘤病毒感染、 宫颈癌、 丙型肝炎、 乙 型肝炎、 丙型肝炎病毒感染和乙型肝炎病毒感染导致的肝癌、 肝硬化等的预防和治疗。
本发 的重组融合蛋白 CTL疫苗及其佐剂的使用方法可以是以下三种: 1.给予卡介 苗, 后接种本发明中的重组融合蛋白 CTL疫苗; 2.先接种本发明中的重组融合蛋白 CTL 疫苗, 后给予卡介苗; 3. 本发明中的重组融合蛋白 CTL疫苗和卡介苗同时使用。
本发明的重组融合蛋白进入机体后, 能够进入 MHC I类抗原递呈途径,激活特异性 的 CTL, 佐剂 BCG进入机体后, 则能够通过 Toll样受体激活抗原递呈细胞表达协同刺激 分子, 为激活 CTL提供更强的第二信号。 附图说明
图 1 : pET28a-HSP65-MUCl质粒酶切产物的琼脂糖凝胶电泳图
图 2: . HSP-MUC1鉴定图谱
图 3: BCG+HSP65-MUC1和 HSP65-MUC1对树突状细胞激活程度的比较。
图 4: BCG加强 HSP65-MUC1注射所激发的特异性抑瘤作用。
图 5: BCG+HSP65-MUC1和 HSP65-MUC1诱生特异性 CTL作用的比较。
图 6: BCG增强 HSP65-MUC1免疫所激发的特异性抑瘤作用。
图 7: HER2多表位抗原基因的琼脂糖凝胶电泳
图 8: pET28a-HSP65-HER2酶切后的琼脂糖凝胶电泳鉴定图
图 9: HSP65-HRE2重组融合蛋白的表达
图 10: BCG+HSP65-HER2 和 HSP65-HER2对树突状细胞激活程度的比较。
图 11 : BCG增强 HSP65-HER2注射所激发的特异性抑瘤作用。
图 12: BCG+HSP65-HER2和 HSP65-HER2诱生特异性 CTL作用的比较。 图 13: BCG增强 HSP65-HER2免疫所激发的特异性抑瘤作用。
图 14: pET28-HSP65-PSA质粒酶切图谱
图 15: BCG+HSP65-PSA和 HSP65-PSA对树突状细胞激活程度的比较。
图 16: BCG增强 HSP65-PSA注射所激发的特异性抑瘤作用。
图 17: BCG+HSP65-PSA和 HSP65-PSA诱生特异性 CTL作用的比较。
图 18: BCG加强 HSP65-PSA免疫所激发的特异性抑瘤作用。
图 19: pMD-18T-HPV质粒的酶切鉴定
图 20: HSP65-HPV质粒的酶切鉴定
图 21 : BCG+HSP65-HPV和 HSP65-HPV对树突状细胞激活程度的比较。
图 22: BCG加强 HSP65-HPV注射所激发的特异性杀伤 HPV感染细胞作用。 图 23: BCG+HSP65-HPV和 HSP65-HPV诱生特异性 CTL作用的比较。
图 24: BCG力卩强 HSP65-HPV免疫所激发的特异性杀伤 HPV感染细胞的作用。 图 25: pMD-18T- SARS/3CL161.264重组质粒的酶切鉴定
图 26 : HSP65-SARS/3CL161-264 SDS-PAGE鉴定及 Western blot鉴定分析 图 27: BCG+HSP65-SARS和 HSP65-SARS对树突状细胞激活程度的比较。 图 28: GFP-SARS/3CL161.264转染细胞鉴定图谱
图 29: BCG加强 HSP65-SARS注射所激发的特异性杀伤 SARS感染细胞的作用。 图 30: BCG+HSP65-SARS和 HSP65-SARS诱生特异性 CTL作用的比较。
图 31 : BCG增强 HSP65-SARS免疫所激发的特异性杀伤 SARS感染细胞的作用。 图 32· : HSP65-HBV重组融合蛋白表达图谱
图 33 : HSP-HBV融合蛋白 SDS-PAGE鉴定图谱
图 34: BCG+HSP65-HBV和 HSP65-HBV对树突状细胞激活程度的比较。
图 35: BCG加强 HSP65-HBV注射所激发的特异性杀伤 HBV感染细胞的作用。 图 36: BCG+HSP65-HBV和 HSP65-HBV诱生特异性 CTL作用的比较。
图 37: BCG增强 HSP65-HBV免疫所激发的特异性的杀伤 HBV感染细胞的作用。 图 38: HSP65-HCV质粒酶切图谱
图 39: HSP65-HCV蛋白质表达纯化的 SDS-PAGE结果
图 40: BCG+HSP65-HCV和 HSP65-HCV对树突状细胞激活程度的比较。
图 41 : BCG加强 HSP65-HCV注射所激发的特异性杀伤 HCV感染细胞的作用。 图 42: BCG+HSP65-HCV和 HSP65-HCV诱生特异性 CTL作用的比较。 图 43: BCG增强 HSP65-HCV免疫所激发的特异性的杀伤 HCV感染细胞的作用。 具体实施方式
通过参考下述实施例详细描述本发明。 但本领域的普通技术人员应理解, 下述实 施例不是用于限制本发明的目的。 本发明的实质保护范围由后附的权利要求所限定。 实施例 1 获取热休克蛋白 65 (HSP65) 的编码基因
1. 卡介苗: 来源于长春生物制品研究所。 采用苏通马铃薯培养基培养卡介苗, 培养的温度为 37-39Ό , 生长出的卡介苗呈现干皱浅黄色的菌膜。收集菌膜从中提取 卡介苗基因组 DNA。
2.提取卡介苗基因组 DNA:方法参照 Molecular Cloning一书(Joseph. Sambrook, David W.Russell,用蛋白酶 K 和苯酚从哺乳动物细胞中分离高分子质量 DNA。
Molecular cloning: A Laboratory Manual,3rd ed.463― 470, Cold Spring Harbor Laboratory Press, 2001 ), 从上步收集的菌膜中提取卡介苗基因组 DNA。
3. 分离 HSP65结构基因: 采用 PCR方法自卡介苗分离 HSP65结构基因。采用 的 5' 端引物序列为 5'CCATG GCC AAGACAATT GCG3' (SEQ ID NO: 1); 3'端引 物序列为 5'ACC GAA TTC GCT AGC CAT ATG GAA ATC CAT GCC ACC CAT 3' (SEQ ID NO: 2)。
反应条件: 94°C, 30"; 55°C, Γ; 72°C, 2 30个循环周期后, 72°C 延伸 10min。 PCR 产物的 5'端带有 Nco I酶切位点, 下游引物 3'端带有 EcoRI酶切位点 (1638bp)。
4. 克隆 PCR产物: 采用 TA克隆方法, 将步骤 3的 PCR产物克隆入 TA载体, 得到重组质粒 pMD-18T-HSP65。 方法见 TA连接试剂盒 (Promega), 重组质粒转化 宿主菌 JM109C购自 Invitrogen公司),取阳性克隆培养,按常规方法(Joseph. Sambrook, David W.Russell.SDS 碱裂解法制备质粒 DNA, Molecular cloning: A Laboratory Manual,3rd ed.27-30, Cold Spring Harbor Laboratory Press, 2001 ) 提取质粒, 采用双 脱氧末端终止法, 对插入片段进行序列测定(ABn»rism310™, USA)。 获得的卡介苗 (BCG) HSP65 基因的序列为 SEQ ID NO: 3。 实施例 2 构建 HSP65和人 MUC1蛋白抗原表位基因的融合基因。
表达载体选用了大肠杆菌表达载体 pET28a (+), 受体菌为大肠杆菌 BL21 ( DE3 ) 菌 株, (均购自 Novagen公司)。
人工合成 MUC1蛋白 VNTR基因序列, 同时在两端引入 £coRI和 H mffll位点。 MUC1蛋白 VNTR基因序列如下:
Figure imgf000009_0001
CCGTCCGGCTCTG ( SEQ ID NO 4)
以 pET28a(+)-HSP65质粒为框架, 将双拷贝 VNTR基因片段克隆入此载体中, 即 获得编码 HSP65与 MUC1蛋白双拷贝 VNTR序列的融合基因序列。 用重组质粒转化 BL21(DE3)宿主菌, 提取重组质粒,经鉴定获得插入 MUC1蛋白 VNTR序列的重组子, 获得的融合蛋白表达载体命名为 pET28a(+)/HSP-MUCl。
具体方法是: 1. 采用 PCR方法合成 HSP65和人 MUC1抗原表位融合蛋白基因: 模板为 BCG HSP65的编码基因 (SEQ E NO:3)(浓度为 0.01pmol/L), PCR的引物为: 5' 端引物序列为: 5,TTC GCC ATG GCC AAG ACA ATT GCG 3' (SEQ ID NO: 5 ) ,
3'端引物的序列为: 5' GGC CGC AAG CTT CAG AGC CGG ACG GTT GTC CGG
Figure imgf000009_0002
ACC GAA TTC GCT AGC CAT ATG CAAATC 3' ( SEQ ID NO: 6)。
反应条件为: 94°C , 30"; 55°C, Γ; 72°C, 4', 30个循环周期后, 72。C延伸 10min。 得到的产物序列如 SEQ ID NO:7所示。
2. PCR产物的克隆: 首先采用 TA克隆方法将上述 PCR产物克隆入 TA载体。方法见 TA连接试剂盒 (Promega) , 重组质粒转化宿主菌 JM109, 取阳性克隆培养, 按常规方 法 (见上文) 提取质粒, 采用双脱氧末端终止法, 对插入片段进行序列测定 ( ABIPrism310™, USA )。 序列测定正确后, 利用 Ncol和 Hindlll切点将其亚克隆至 pET28a载体 (Novagen) , 得到 pET28a-HSP65-MUCl质粒。 转化宿主菌 JM109, 取阳性 转化菌培养, 提取质粒, 采用双脱氧末端终止法, 对插入片段进行序列测定 (ABIPrism310™, USA)。
图 1 : pET28a-HSP65-MUCl质粒酶切产物的琼脂糖凝胶电泳图
第 2泳道箭头所指的为 123bp的 MUC1表位 DNA片段 实施例 3 重组 HSP65-MUC 1融合蛋白的表达和纯化 将构建好的表达质粒 pET28a(+) /HSP-MUCl 转化进入己制备好的大肠杆菌 BL21 (DE3 )感受态细胞, 涂布含 100μ§/πι1卡那霉素的 LB平板, 然后在转化平板上挑取单 克隆, 接种到 3nd LB液体培养基中, 37°C恒温摇床上 300rpm振荡培养过夜, 用碱裂 解法抽提并检测质粒。 将确证含有表达质粒 pET28a(+) /HSP- MUC1 的单克隆在含 100μ§/πι1卡那霉素的 LB平板上点复板保存。
挑选一株确证好的表达工程菌, 采用 10升的发酵罐, 用 LB培养基进行培养及 用 IPTG诱导表达。采用高压破菌方法, 在 500〜600 bar压力下, 循环破菌 4~5次, 离心收上清, 上清通过镍亲合层析, 10K膜块超滤脱盐, 最后经过 Q-Sepharose FF 层析, 得到重组 HSP-MUC 1融合蛋白 (分子量为 61KD)。 取样做 HPLC分析, 重 组 HSP65-MUC1融合蛋白的纯度为 96.1%。
图 2 Western blotting证实该表达条带为 HSP-MUC1。
表达 SDS-PAGE电泳图 (左), Western blotting图谱 (右)
1 诱导前; 2、3、4、5 不同表达克隆诱导 3小时;3' HSP单抗 Western; 5'MUC1 单抗 Western; 实施例 4 BCG和 HSP65-MUC1体外诱导人的树突状细胞的活化和成熟
一、 分离人外周血单核细胞
1. 向 50 ml离心管中加入 12.5 ml泛影葡胺 (天津 TBD公司生产)。
2. 汲取 50 ml PBS EDTA(pH7.4) 缓冲液移入一烧瓶中。
3. 血袋经 70%乙醇洗涤后, 将其中的血移入于含 PBS/EDTA的烧瓶中, 混匀。
4. 从烧瓶 Φ汲取 25 ml血液和 PBS的混合液移入于含 12.5 ml泛影葡胺的 50 ml离 心管中。
5. 将上述试管离心, 室温, 3000 rpm/min (不使用刹车), 离心 20-25 min。
6. 离心后弃去上清, 汲取界面层细胞, 移入一新离心管中。
7. 向含有分层细胞的离心管中加入 PBS/EDTA缓冲液至 45 ml, 1800 rpm/min, 4°C 离心 10 min。 此为第一次冲洗; 第二次冲洗时为 1200 rpm/min, 4°C离心 7 min。
8. 离心后, 加入含有 2 %人血清和 2mmEDTA的 PBS缓冲液 (以下简称 PBS/EDTA/ 人血清缓冲液) 重悬细胞。
9. 第兰次洗涤, 1200 rpm/min, 4。C离心 7 min (不用刹车)。
10.以 6 ml冷的 PBS I EDTA I人血清缓冲液重悬细胞, 取 2只试管分别加入 6ml 52%冷的 Percol。
11.汲取 3ml细胞悬液缓慢移入(勿搅动 Percol )含6:^冷的 52% Percol(Pharmingen) 试管中, 在无刹车条件下 2000 rpm/min ,4'C离心 20 min。
12. 汲取界面层细胞, 移入一新离心管中。
13.以冷的 PBS/EDTA/人血清缓冲液洗涤分层细胞, 4'C, 1300 πι/min, 无刹车离 心 10 min。
14.以 200目滤网滤过因死亡而成团的细胞。
15.计数单核细胞数。 二、 诱导未成熟的树突状细胞 (dendritic cell,DC)
1. 以 MDM (Sigma Chemical Co. 含有 10%的灭活胎牛血清 (GIBCO BRL) 100IU青霉素, 100mg ml的链霉素, β—巯基乙醇 5μ1/1000πι1, 碳酸氢钠 3.5g/1000ml) 稀释单核细胞, 调整细胞数达2 106 / 1^。
2. 向 12孔板中每一个孔加入 1 ml 细胞悬液和 lml MDM, 将细胞于 37°C, 5% C02孵箱内中培养 2h。
3. 2h后用 DM洗漆去掉每孔中未粘附的细胞。
4. 每孔中加入 2 ml含 100ng (200 U) /ml 的 GM-CSF (Pharmingen) 和 200 U/ml的 IL-4 (Pharmingen) 的 IMDM培养液。
5. 于 37°C, 5% C02孵箱中培养 5d, 每 2-3 d换一次培养液。 三、 用 BCG和 HSP65-MUC1融合蛋白装载树突状细胞
1. 于第五天, 分别设立阴性对照组: 只加培养液; BCG组: 加入 BCG (长春生物 制品所生产)使其终浓度达 3(^g/ml; BCG+HSP65-MUC1组:加入 BCG和 HSP65-MUC1 使其终浓度分别为 3(^g/ml和 10(^g/ml; HSP65-MUC1组: 加入 HSP65-MUC1使其终浓 度达 ΙΟΟμ^η Ι ; 阳性对照组: 加入下列因子并使其终浓度分别达到 10ng/ml TNFa ( Pharmingen )、 10ng/ml IL-6 ( Pharmingen )、 1 Ong/ml IL- 1 β ( Pharmingen )和 10μ§/ηι1 PGE2 ( Pharmingen )。 直接将上述的 BCG和 HSP65-MUC 1蛋白及细胞因子加入未成熟的树突 状细胞 (DC) 中继续培养 2d。
2: 于第 7d收获树突状细胞, 进行活化和成熟指标的测定。 四、 树突状细胞活化和成熟指标的测定
1. 于第 7天收集树突状细胞于 96孔板中, 每孔加入 200μ1的细胞悬液。
2. 1500rpm, 4QC离心 5分钟, 弃掉上清液, 留存孔底的细胞。
3. 每孔中加入 200μ1 ΡΒ5缓冲液, 洗涤两次, 弃掉上清液, 留存孔底细胞。 4. 轻轻弹起孔底的细胞, 分别向每孔中加入 1.5^ FITC标记的 CD86荧光抗 体 (BD, USA)。
5. 避光, 冰上染色 30分钟。
. 6. 染色完毕后, 用 PBS缓冲液, 每孔 200μ1, 洗涤细胞两次。
7. 洗涤完毕后, 用 400μ1 ΡΒ3缓冲液收集细胞, 利用流式细胞仪进行测定。 五、 结果
流式细胞仪的测定结果表明: 同 HSP65-MUC1组相比较, BCG+HSP65-MUC1能 够显著的诱导 DC的活化和成熟。 DC是人体内最为重要的抗原递呈细胞, DC的活化和 成熟标志着 DC的抗原递呈能力加强, 激活针对 MUC1的细胞毒性杀伤性 T淋巴细胞 (CTL)的能力加强;这样有助于人体内被激活的 CTL杀伤体内表达 MUC1的肿瘤细胞, 因此, 可用于预防和 /或治疗 MUC1阳性肿瘤。
HSP65-MUC 1和 BCG+HSP65-MUC 1对树突状细胞激活程度的比较见图 3。 实施例 5 转染 pcDNA3-GFP-MUCl细胞 B16 (C57BL/6小鼠黑色素瘤细胞系)的构建 1. pcDNA3-GFP-MUCl重组质粒的构建
pET26-IRES-GFP质粒由 Novagen公司提供, pET26-IRES-GFP质粒经 NcoI、 Sail 双酶消化后, 用琼脂糖凝胶电泳分离消化产物并回收消化产物略大于 750bp的 GFP片 段。 pET28载体 (Invitrogen) 用 NcoI、 Sail双酶消化后, 用琼脂糖凝胶电泳分离消化 产物并回收线性载体,将 pET28线性载体与 GFP片段用连接酶连接,即得到 pET28-GFP 重组质粒。 用 pET28— GFP重组质粒转化宿主菌 JM109, 取阳性克隆培养, 按常规方 法 (见上文) 提取质粒, 采用双脱氧末端终止法, 对插入片段进行序列测定 (ABIPrism310™, USA) )。 以 pET28-GFP为模板, 利用引物
5,引物 AAGCTTACCATGGAAGTGAGCAAGGGCGAG 3 (SEQ ID NO:8)
3'引物 TGGATCCTTTGTACAGCTCGTCC3 (SEQ ID NO: 9)
进行 PCR反应: 94°C , 30"; 60。C, Γ; 72°C, 1*30", 27个循环周期后, 72°C延伸 lOmino (约 750bp)
将上述 PCR产物 (GFP基因片段)克隆入 TA载体(方法见实施例 1 )。 Hindlll.BamHI 分别消化 pcDNA3 (Invitrogen公司提供)、 pET28-GFP, 用琼脂糖凝胶电泳分离并回收 消化产物, 用 T4DNA连接酶连接回收的消化产物, 即得到 pcDNA3-GFP重组质粒。
用 X ol和 Xbal分别消化 pcDNA3-GFP重组质粒和用 PCR方法得到的 MUC1片段: 以
HSP65-MUC1 为 模 板 , 5' 端 弓 I 物 5'CTCGAGCCGGTACCGATTCCATGGAATT CAGAT3'(SEQ ID NO: 10), 3'端引物 5'GGCCTCTAGACTCGAGTTATTAGCCAGA AGTAG3'(SEQ ID ΝΟ:11),
PCR条件: 94°C, 30"; 60°C, Γ; 72°C, 130" , 27个循环周期后, 72°C延伸 10min。 用琼脂糖凝胶电泳分离并回收消化产物 (回收试剂盒为北京鼎国生物技术有限责 任公司生产, 操作按说明书进行), 用 T4DNA连接酶连接回收的消化产物, 即得到 pcDNA3-GFP-MUCl。
2. 细胞转染:
( 1 ) 准备下列试剂:
溶液 A: 含 1-2μβ pcDNA3-GFP-MUCl质粒 (含表位基因的质粒) 的 ΙΟΟμΙ无血清 IMDMo
溶液 B: 含 10-20μ1 Liposome试剂 (Invitrogen)的 ΙΟΟμΙ无血清 MDM。
(2)将溶液 A慢慢加到溶液 B中,轻轻混合,室温(一般为 20-25°C左右)孵育 45min, 此时混合液中可能出现絮状物, 为正常现象。
(3 )在混合液孵育期间, 用 2ml无血清 DM培养液将 B16细胞(购自 ATCC)洗一 次。
(4) 将 0.8ml无血清 IMDM培养液加到 A/B混合液中, 轻轻混合。
(5 ) 将 A/B混合液轻轻加入到含有 B16细胞的培养孔中。
(6) 37°C, 5%C02培养 5小时。
(7) 直接向培养孔中加入 lml含 20%FBS的 DM, 继续培养 18-24小时。
( 8) 弃培养液, 加入 2ml含 10%FBS和含 G418 (GEBCO) 的新鲜 MDM, 继续培 养。
(9)将 pcDNA3-GFP—MUCl转染的 B16细胞经 G418抗生素(GIBCO)筛选出转染 阳性的克隆, 将这些克隆分别消化进入培养瓶并扩增培养, 经 RT-PCR及 Western Blot鉴 定出高表达 MUC1的克隆, 用于功能试验。 实施例 6 BCG增强 HSP65-MUC1注射所激发的小鼠体内的特异性抑瘤作用
1.方法:设立 PBS组(阴性对照组)、 BCG组、 HSP65-MUC1组、 BCG+HSP65-MUC1 组和卡铂组 (阳性对照组) 。 每组 10只 7周龄的 C57BL/6雌性小鼠 (北京维通利华实验 动物有限公司)。于第 0天给所有小鼠接种上述转染 MUC1表位抗原多肽编码基因的 B16 细胞 1.5χ105 / 200μ1 PBS, 接种部位是小鼠右侧背部皮下。 于第 2天, 按不同分组对小 鼠进行第一次注射, 注射部位为小鼠四肢皮下向心端。 剂量为 HSP65-MUC1组: 10μ§ HSP65-MUC1 / 200μ1 PBS /只小鼠; BCG+HSP65-MUC1组: lO g HSP65— MUC1 + 1.32mg BCG/200W PBS /只小鼠; PBS组: 200μ1 PBS /只小鼠; BCG组: 1.32mg BCG /200μ1 PBS /只小鼠; 阳性对照组: 阳性对照组小鼠每 4天注射一次卡铂 (中国齐鲁制 药厂生产) , 腹腔注射 0.5mg卡铂 /只小鼠, 连续注射四次。 于第 16天时, 对各组小鼠进 行第二次注射, 注射剂量和部位同第一次免疫。 每日观察小鼠的状态, 肿瘤的生长情 况, 当 PBS对照组小鼠开始死亡, 或 PBS对照组小鼠少于 20%的小鼠瘤重低于 0.4g时, 开始杀鼠取瘤。 称取各组小鼠的肿瘤重量。
2. 结果: BCG+HSP65-MUC1组小鼠的肿瘤重量显著低于 HSP65-MUC1组 (秩和检验, P<0.05 ) , 说明 BCG能够加强 HSP65-MUC1注射所激发的特异性抑瘤作用。
BCG+HSP65-MUC1组与 HSP65-MUC1组及其它组肿瘤重量的比较, 见图 4。
3. 结论: BCG能明显增强 HSP65-MUC1注射所激发的特异性抑瘤作用, 抑制 MUC1阳 性肿瘤的生长。 实施例 7 BCG增强 HSP65-MUC1诱生 MUC1特异性 CTL的作用
1. 方法: 分别在 0、 14、 21天, 用 BCG和 HSP65-MUC1经小鼠四肢皮下向心端免 疫 7周龄的 C57BL/6雌性小鼠。 BCG+HSP65-MUC1组: 注射剂量为 1.32mg BCG+ l(^g HSP65-MUC1 I 200μ1 PBS I只小鼠; PBS注射组: 200μ1 PBS I只小鼠; HSP65-MUC1 组: l (^g HSP65-MUC1 I 200μ1 PBS I只小鼠; BCG组: 1.32mg BCG/200W PBS I只小 鼠。 免疫结束后第 5d, 分离小鼠的脾及淋巴结细胞作为效应细胞。 MUC1表位抗原多肽 编码基因转染的 B16细胞及未转染 MUC1表位抗原多肽编码基因的 B16细胞作为靶细 胞, 按常规方法测定 CTL活性 (C.Stremmel, E.A.Greenfield, E.Howard, GJ.Freeman and V.K.Kuchroo.B7-2 Expressed on EL4 lymphoma Suppresses Antitumor Immunity by an Interleukin4-dependent Mechanism. J.Exp.Med, 1999,189(6):919-930. )。 2. 结果: 同 HSP65-MUC1组相比较, MUC1表位抗原多肽编码基因转染的 B16细胞作 为靶细胞时, BCG+HSP65-MUC1免疫组小鼠的脾及淋巴结 CTL在效靶比为 1 : 20时的 杀伤率为 45%, HSP65-MUC1免疫组小鼠的脾和淋巴结 CTL的杀伤率为 34%。 这表明 BCG能够加强 HSP65-MUC1在小鼠所诱生的 MUC1特异性的杀伤性 T淋巴细胞的作用。 BCG+HSP65-MUC1和 HSP65-MUC1诱生特异性的 CTL作用的比较见图 5。
3. 结论: BCG能显著增强 HSP65-MUC1所诱生的特异性 CTL的作用。 实施例 8 BCG增强 HSP65-MUC1免疫所激发的特异性抑瘤作用
1. 方法: 于第 0, 14, 28天, 按照不同的分组, 给予 7周龄的 C57BL/6雌性小鼠进 行免疫。 实验小鼠分为 4组, 每组 10只小鼠: PBS组、 HSP65-MUC1组、 BCG 组、
BCG+HSP65-MUC1组。 免疫部位: 小鼠四肢皮下向心端, 剂量: PBS组, 200μ1 PBS I只小鼠; HSP65-MUC1组, 10μ§ HSP65-MUC1 I 200μ1 PBS I只小鼠; BCG组, 1.32mg BCG /200μ1 PBS /只小鼠; BCG+HSP65-MUC1组, 10μβ HSP65 -MUC1 + 1.32mg BCG/200W PBS I只小鼠。 最后一次免疫后第 5天, 在小鼠右侧背部皮下给小鼠接种上 述转染 MUC1表位抗原多肽编码基因的 B16细胞 1.5X105个 I 200μΐΡΒ8 I只小鼠。 每日 观察小鼠的状态, 肿瘤的生长情况以及小鼠体重。 当 PBS对照组小鼠开始死亡, 或 PBS 对照组小鼠少于 20%的小鼠瘤重低于 0.4g时, 开始杀鼠取瘤, 称取各组小鼠的肿瘤重 量。 各组间肿瘤重量的比较见图 6。
2. 结果: BCG+HSP65-MUC1 组小鼠的肿瘤重量显著低于 HSP65-MUC1组。 3. 结论: BCG能够显著加强 HSP65-MUC1免疫所激发的特异性抑瘤作用, 能明显 抑制小鼠体内 MUC1阳性肿瘤的生长。 实施例 9合成 HER2蛋白多表位抗原基因
用四轮 PCR合成 HER2蛋白多表位抗原基因
1. 第一轮 PCR:下列两条引物互为模板 引物 1的序列为:
5'CTCTCGCATTi
ACCGCTTCA 3YSEO ID NO: 12)
引物 Γ的序列为:
5'CAACGATTTCT
GTGCGGTATT 3YSEO K) NO: 13)
第一轮 PCR合成的产物的序列是:
Figure imgf000016_0001
3'(SEQ ID NO: 14)
2. 第二轮 PCR: 以第一轮 PCR的产物为模板
引物 2的序列是-
5'CTGACCGAGC
GCATTCCTGC 3YSEO ID NO: 15)
引物 2'的序列是
5'CTTTGTTAGCTTTCGGGGAGGTGTTTTCACGCAGAACCTTGATAGCAACGA TTTCTTCCA 3YSEO ID NO: 16)
产物的序列是-
Figure imgf000016_0002
CTGCGTGAAAACACCTCCCCGAAAGCTAACAAAG3'(SEQ ID NO:
3. 第三轮 PCR: 以第二轮 PCR的产物为模板
引物 3的序列是:
AGCAG3YSEO ID NO: 18)
引物 3'的序列是:
5'AGTCAGAGA(
TTTCG3YSEO ID NO: 19)
产物的序列是: TCTGACT3'(SEQ ED NO: 20)
4. 第四轮 PCR: 以第三轮 PCR的产物为模板
弓 I 物 4 的 序 列 是 : 5' GAATTCGACGAAGCATACGTTATGGCT GGTGTTGGTTCCCCG 3' (SEQ ID NO: 21)
引物 4'的序列是 5' AAGCTTACCTTGCAGAGTCAGAGAGTAAGC 3YSEQ ID NO:
22)
产物的序列是:
Figure imgf000017_0001
NO: 23)。 此序列代表的是 HER2多表位抗原基因。
PCR的反应条件为: 94°C , 30"; 55°C, Γ; 72°C, Γ, 30个循环周期后, 72°C延 伸 10min。 用 PCR获得的 HER2多表位抗原基因的琼脂糖凝胶电泳见图 7 实施例 10 构建表达 HSP65基因的表达载体 (pET28a— HSP65)
将热休克蛋白 65 (HSP65 )的编码基因和 pET28a (Novagen)载体分别用 NcoI、 ECORI 消化, rrc, 2h。 采用琼脂糖凝胶电泳分离消化产物并回收消化产物 (回收 试剂盒为北京鼎国生物技术有限责任公司生产,操作按说明书进行)。电泳的条件是 : 1%琼脂糖凝胶, Ιχ ΤΑΕ缓冲液, 150-200mA, 电泳 0.5-lh。
(20χ ΤΑΕ缓冲液: 0.8mol/L Tris碱, 0.4mol/L NaOAc, 0.04mol/L Na2 EDTA, 用冰醋酸调 pH 8.3 )。
将经 NcoI、 EcoRI消化的热休克蛋白 65 (HSP65 ) 的编码基因和 pET28a载体 05 001715 用连接酶连接。 将连接产物转化宿主菌 JM109, 取阳性转化菌培养, 提取质粒 (见上 文), 采用双脱氧末端终止法, 对插入片段进行序列测定 (ABIPriSm310™, USA;)。 实施例 11 HSP65和 HER2多表位抗原基因融合基因 (pET28a-HSP65-HER2 ) 的 构建
1. 在 37°C, 用 EcoRI和 Hindlll分别消化 HER2多表位抗原基因 (如实施例 9 所述) 和插入了 HSP65基因的 pET28a载体 (pET28a— HSP65) 两小时。 消化产物 经如上的琼脂糖凝胶电泳分离并回收消化产物 (回收试剂盒为北京鼎国生物技术有 限责任公司生产, 操作按说明书进行)。
2. 将经过 EcoRI和 Hindlll分别消化的 HER2多表位抗原基因和插入了 HSP65 基因的 pET28a载体, 用 T4DNA连接酶连接。 将连接产物 pET28a(+)-HSP65-HER2 质粒, 转化宿主菌 JMI09, 取阳性转化菌培养。
图 8 pET28a-HSP65-HER2酶切后的琼脂糖凝胶电泳鉴定图
泳道 1 : 经 EcoRI和 Hindlll酶切后,得到的 HER2多表位抗原基因片段 (315bp) 泳道 2: 分子量标尺 (Takara) 2000bp; lOOObp; 750 bp; 500 bp; 250bp; lOObp
3. HSP65和人 HER2多表位抗原基因融合构成的融合基因的测序
按照常规提取质粒的方法(见上文), 得到含有 HSP65-HER2基因的质粒, 用测 序仪 (ABIPrism310TM,USA) 测定 HSP65-HER2的基因序列, 结果表明, 所得到的 HSP65 - HER2 融合基因和本发明设计的 HSP65-HER2 基因序列完全一致。 HSP65-HER2的核苷酸序列如 SEQ Π) NO: 24所示。
4.将含 HSP65-HER2融合基因的重组质粒 pET28a(+)-HSP65-HER2转化 BL21 DE3 (Novagen, America) 0 实施例 12 HSP65-HER2重组融合蛋白的表达
挑取含有重组质粒 pET28a-HSP65-HER2的表达菌 BL21 DE3 (Novagen, America) 单菌落接种于含有 50 ml LB培养基的 250 ml三角烧瓶中, 37 °C水浴震荡培养至 OD600 值为 0.6。 加入 IPTG使其终浓度为 0.4 mM, 37°C水浴震荡培养 2-3h。将含有表达菌的 三角烧瓶置于冰上 5 min, 4°C离心 5min (5000 x g)。 吸弃上清, 收集细菌, 立即使用 或冻存。
图 9 HSP65-HRE2重组融合蛋白的表达 实施例 13 重组融合蛋白 HSP65-HER2 的纯化
重组融合蛋白 HSP65-HER2用下述方法进行纯化。 '
1、 裂菌
使用溶液 A (50mM NaCK 20mM醋酸钠 pH6.0) 重悬发酵菌体, 超声波裂 菌, 离心收集上清。
2、 离子交换层析
层 ^斤填料: Q Sepharose XL(Pharmingen,USA)
溶液 A: 50mM NaCK 20mM 醋酸钠 pH6.0
溶液 B: 20mM醋酸钠、 lM NaCl pH6.0
样品: 超声裂菌上清
步骤: 使用溶液 A平衡 Q Sepharose XL介质, 样品上样后, 使用溶液 B 0— 100% 线性梯度洗脱, 收集目标蛋白所在洗脱峰作为下一步疏水层析样品。
3、 疏水层析
层析填料: Phenyl Sepharose FF (Pharmingen)
溶液 C: 1.5M NaCK 20mMTris pH7.9
溶液 D: 20mM Tris pH7.9
样品: 离子交换层析洗脱样品 (经透析后更换缓冲液为溶液 C)
步骤: 用溶液 C平衡疏水介质 (Phenyl SFF), 样品上样, 溶液 D 35 %— 100%进 行线性梯度洗脱, 收集目标蛋白所在洗脱峰。
4、 DEAE离子交换去除内毒素
层析填料: DEAE Sepharose FF (Pharmingen)
溶液 E: lOOmM NaCK 20mM Tris、 pH7.0
溶液 F: 1.5M NaCK 20mM Tris、 pH7.0
样品: 疏水层析洗脱样品
步骤: 用溶液 E平衡介质 (DEAE SFF), 溶液 F进行梯度洗脱, 收集洗脱峰。 洗 脱样品使用 G25除盐并更换缓冲液为 PBS。
样品进行脱盐后即得到重组融合蛋白 HSP65-HER2 实施例 14 BCG和 HSP65-HER2体外诱导人的树突状细胞的活化和成熟 一、 如实施例 4的部分 (一) 所述分离人外周血单核细胞
二、 如实施例 4的部分 (二) 所述诱导未成熟的树突状细胞
三、 用 BCG和 HSP65-HER2重组融合蛋白装载树突状细胞
1. 于第五天, 分别设立对照组: 只加培养液; BCG组: 加入 BCG使其终浓度达 3(^g/ml; BCG+HSP65-HER2组: 加入 BCG和 HSP65-HER2使其终浓度分别为 3( g/ml 和 10(^g/ml; HSP65-HER2组: 加入 HSP65-HER2使其终浓度达 10( g/ml; 阳性对照组: 分别加入下列因子并使其终浓度分别为 10ng/mlTNFa、 10ng/mlIL-6、 lOng/mlIL-Ιβ和 l( g/ml PGE2。 直接将上述的 BCG和 HSP65-HER2蛋白和细胞因子加入未成熟的树突 状细胞 (DC) 中继续培养 2d。
2.于第 7d收获树突状细胞,进行活化和成熟指标的测定。
四、 如实施例 4的部分 (四) 所述进行树突状细胞活化和成熟指标的测定 五、 结果
各组间 CD86荧光值的比较见图 10。 流式细胞仪的测定结果表明: 同 HSP65-HER2 组相比较, BCG+HSP65-HER2能够显著地诱导 DC的活化和成熟。 DC是人体内最为重 要的抗原递呈细胞, DC的活化和成熟标志着 DC的抗原递呈能力加强, 激活针对 HER2 的细胞毒性杀伤性 T淋巴细胞(CTL) 的能力加强; 这样有助于人体内被激活的 CTL杀 伤体内表达 HER2的肿瘤细胞, 因此, 可用于预防和 /或治疗 HER2阳性肿瘤。 实施例 15获得稳定表达 HER2多表位抗原基因的 B16 (C57BL/6小鼠黑色素瘤细 胞系) 细胞株
1.用 PCR方法获得多聚 HER2多表位基因的序列
以 SEQ ID NO: 24 的 DNA 片段为模板, 进行 PCR 反应:引物 5 的序列 为: 5,-CTGCAGGATCCATGGACGAAG CATACGTTA TG GC- 3, (SEQ ID NO:25)
弓 I 物 5, 的 序 列 为 : 5'-GCGGCCGCAAGCTTAAGATCTACCTTG CAGAGTCAGAGAGT AA-3'(SEQ ID NO:26)
PCR的反应条件为: 94°C , 30"; 55°C, Γ; 72°C, Γ, 30个循环周期后, 72°C延 伸 10min。 得到产物 HER2多表位抗原基因的序列为:
5'CTGCAGGATCCATGGACGAAGCATACGTTATGGCTGGTGTTGGTTCCCC GCAAGGTAGATCTTAAGCTTGCGGCCGC 3, (SEQ ID NO: 27) (含有 Pstl, BamHI, kozak序列, Bgin, Hindm和 Notl酶切位点)。
2.将 HER2多表位基因与真核表达载体 pcDNA3连接
将 HER2多表位抗原基因 (SEQ ID NO:27) 和真核表达质粒 pcDNA3(Invitrogen ) 分别用限制性内切酶 BamHI和 Notl酶切。 酶切条件 37Ό 2小时, 如上釆用琼脂糖凝 胶电泳分禽消化产物并回收消化产物。
将经 BamHI和 Notl酶切消化的 HER2和用同样酶处理的真核表达质粒 pcDNA3 用连接酶连接, 即得到 pcDNA3-HER2。
3. 用 pcDNA3-HER2转染 B16细胞具体方法见实施例 5中的部分 (2) 的细胞转染。 实施例 16 BCG增强 HSP65-HER2注射所激发的小鼠体内的特异性抑瘤作用
1. 方法: 除了以 HSP65-HER2代替 HSP65-MUC1 , 以转染 HER2表位抗原多肽编码 基因的 B16细胞代替实施例 6部分(1 ) 中的 B16细胞以外, 如实施例 6的部分(1 )所述, 进行实验。.
2 . 结果: BCG+HSP65-HER2组的小鼠肿瘤重量显著低于 HSP65-HER2组。
(PO.05 ) 。 各组间肿瘤大小的比较见图 11。
3.结论.,BCG能够显著增强 HSP65-HER2注射所激发的特异性抑瘤作用,抑制 HER2 阳性肿瘤的生长。 实施例 17 BCG加强 HSP65— HER2 诱生 HER2特异性 CTL的作用
1. 方法: 除了以 HSP65-HER2代替实施例 7的部分(1 ) 中 HSP65-MUC1 , 以 HER2 表位抗原多肽编码基因转染的 B16细胞及未转染 HER2表位抗原多肽编码基因的 B16细 胞作为靶细胞代替实施例 7的部分 (1 ) 中的靶细胞以外, 按照实施例 7的部分 (1 ) 进 行实验。 '
2. 结果: 同 HSP65-HER2组相比较, BCG+HSP65-HER2免疫组小鼠的脾及淋巴结 CTL在效靶比为 1 : 20的杀伤率为 52%, HSP65-HER2免疫组小鼠的脾和淋巴结 CTL的杀 伤率为 41%。 这表明 BCG能够加强 HSP65-HER2所诱生的 HER2特异性 CTL的作用。 BCG+HSP65-HER2和 HSP65-HER2诱生特异性 CTL作用的比较见图 12。
3.结论: BCG能够显著加强 HSP65-HER2所诱生的 HER2特异性的 CTL,抑制 HER2 阳性肿瘤细胞的生长。 实施例 18 BCG增强 HSP65-HER2免疫所激发的特异性抑瘤作用
1.方法:除了以 HSP65-HER2代替实施例 8部分(1 )的 HSP65-MUC1 , 以转染 HER2 表位抗原多肽编码基因的 B16细胞代替实施例 8部分(1 )的 B16细胞以外, 按照实施例 8 的部分 (1 ) 进行实验。
2. 结果: BCG+HSP65-HER2组的小鼠肿瘤重量显著低于 HSP65-HER2组。 各组 间肿瘤重量的比较见图 13。
3. 结论: BCG能够显著增强 HSP65-HER2免疫所激发的特异性抑瘤作用, 能明显 抑制小鼠体内 HER2阳性肿瘤细胞的生长。 实施例 19 构建热休克蛋白 65与前列腺特异性抗原的融合基因
采用 PCR方法合成 HSP65和前列腺特异性抗原(prostate specific antigen, PS A )的细 胞毒性 T淋巴细胞 (CTL) 细胞表位的融合基因, 模板为热休克蛋白 65 (HSP65 ) 的编 码基因 (浓度为 0.01pmol/L), PCR引物的为:
5'端引物序列为: 5'-CCATG GCC AAG ACA ATT GCG-3' ( SEQ ID NO: 28 ) ;
3'端引物的序列为:
5'-AAGCTTTTTAGTAACTTTCTGCGGGTGAACCTGAGCGCAAACGTCGTTAG
TGCCACCCATGTC-3' (SEQ ID NO: 29) 。
反应条件为: 94° C , 30"; 55° C, Γ ; 72° C, 2', 30个循环周期后, 72° C延伸 10分 钟。 得到的产物即为 HSP65-PSA融合基因, 序列见 SEQ ID NO: 30。
用 Ncol和 Hindlll在 37'C消化 PCR产物,时间为 2小时。消化产物如上经琼脂糖凝胶 电泳分离后, 回收消化产物 (回收试剂盒为北京鼎国生物技术有限责任公司生产, 操 作按说明书进行)。
将回收的 PCR产物克隆入经限制性内切酶 Ncol和 Hindlll消化的原核细胞表达载 体 pET-28a(+)质粒 (美国 Novagen公司) 的 6聚组氨酸 (histidine) 密码子的上游。
将含 HSP65和 PSA融合基因的重组 pET-28a(+)— HSP65-PSA质粒转化大肠杆 菌。取阳性转化菌培养。按照常规提取质粒的方法(见上文), 得到含有 HSP65-PSA 基因的质粒, 用测序仪 (ABIPrism310™,USA ) 测定 HSP65-PSA 的基因序列。 HSP65-PSA的核苷酸序列如 SEQ ID NO: 30所示。 实施例 20 重组融合蛋白 HSP65-PSA的表达
挑取含有重组质粒 pET28a-HSP65-PSA的表达菌 BL21 DE3 (Novagen, America)单 菌落接种于含有 50 ml LB培养基的 250 ml三角烧瓶中, 37 'C水浴震荡培养至 OD600 值为 0.6。 加入 IPTG使其终浓度为 0.4 mM, 37°C水浴震荡培养 2-3h。将含有表达菌的 三角烧瓶置于冰上 5 min, 4°C离心 5min ( 5000 x g)。 吸弃上清, 收集细菌, 立即使用 或冻存。 . 实施例 21重组融合蛋白 HSP65-PSA的纯化
采用 10升的发酵罐表达重组 HSP65-PSA融合蛋白, 以乳糖为诱导物。 在裂菌后, 经镍亲合层析、 除盐、 离子交换层析、 除盐后得到重组 HSP-PSA融合蛋白。 采用常规 的 SDS-PAGE方法鉴定 HSP-65和前列腺特异性抗原(PSA)的 T细胞表位的基因融合 蛋白的纯度为: 98%。
图 14 HSP-PSA融合蛋白 SDS-PAGE鉴定图谱 实施例 22 BCG和 HSP65-PSA体外诱导人的树突状细胞的活化和成熟
一、 如实施例 4的部分 (一) 所述分离人外周血单核细胞。
二、 如实施例 4的部分 (二) 诱导未成熟的树突状细胞。
三、 用 BCG和 HSP65-PSA融合蛋白装载树突状细胞
1 . 于第五天, 分别设立对照组: 只加培养液; BCG组: 加入 BCG使其终浓度达
3(^g/ml ; BCG +HSP65-PSA组: 加入 BCG和 HSP65-PSA使其终浓度分别为 3(^g/ml和 10(^g/ml; HSP65-PSA组: 加入 HSP65-PSA使其终浓度达 10(^g/ml; 阳性对照组: 分别 加入下列因子使其终浓度分别为 1 Ong/mlTNFa、 1 Ong/mlIL-6、 lOng/mllL- 1 β和 1 (^g/ml PGE2。直接将上述的 BCG和 HSP65-PSA蛋白和细胞因子加入未成熟的树突状细胞( DC ) 中继续培养 2d。 2. 第 7d收获树突状细胞,进行活化和成熟指标的测定。
四、 如实施例 4的部分 (四) 所述进行树突状细胞活化和成熟指标的测定。
五、 结果
各组间 CD86荧光值的比较见图 15。 流式细胞仪的测定结果表明: 同 HSP65-PSA组 相比较, BCG+HSP65-PSA能够显著地诱导 DC的活化和成熟。 DC是人体内最为重要的 抗原递呈细胞, DC的活化和成熟标志着 DC的抗原递呈能力加强, 激活针对 PSA的细胞 毒性杀伤性 T淋巴细胞 (CTL) 的能力加强; 这样有助于人体内被激活的 CTL杀伤体内 PSA阳性的肿瘤细胞, 因此, 可用于预防和或治疗 PSA阳性肿瘤。 实施例 23.转染 PCDNA3— GFP—PSA的 B16细胞的构建
1. pcDNA3— GFP _ PSA质粒的构建: 以 HSP65-PSA为模板, 5'端引物 5 '-GGATCC ATGGAGCCTGAAGAGTTC-3 " ( SEQ ID NO: 31 ) , 3'端引物 5'-GGGTCGACTCTAGATCAACACAGCATGAATTT-3' (SEQ ID NO: 32),
PCR条件: 94。C , 30"; 60°C, l' ; 72°C, Γ30", 27个循环周期后, 72°C延伸 10min。 用 BamHI和 Xbal分别消化 pcDNA3— GFP质粒和 PCR产物, 用琼脂糖凝胶电泳分 离并回收消化产物 (回收试剂盒为北京鼎国生物技术有限责任公司生产, 操作按说明 书进行), 用 T4DNA连接酶连接回收的消化产物, 即得到 pcDNA3— GFP— PSA。
2. 用 pcDNA3-GFP— PSA转染 B16细胞具体方法见实施例 5中的细胞转染。 实施例 24 BCG增强 HSP65-PSA注射所激发的特异性抑瘤作用
1. 方法: 除了以 HSP65-PSA代替 HSP65-MUC1 , 以转染 PSA表位抗原多肽编码基 因的 B16细胞代替实施例 6部分 (1 ) 中的 B16细胞以外, 如实施例 6的部分 (1 ) 所述, 进行实验。
2.结果: HSP65-PSA+BCG组的小鼠肿瘤重量显著低于 HSP65-PSA组(PO05 )。 各组肿瘤重量的比较见图 16。
3. 结论: BCG能够显著增强 HSP65-PSA注射所激发的特异性抑瘤作用, 抑制 表达 PSA肿瘤细胞的生长。 实施例 25 BCG加强 HSP65-PSA诱生 PSA特异性 CTL的作用
1. 方法: 除了以 HSP65-PSA代替实施例 7的部分 (1 ) 中 HSP65-MUC1 , 以 PSA表 位抗原多肽编码基因转染的 B16细胞及未转染 PSA表位抗原多肽编码基因的 B16细胞作 为靶细胞代替实施例 7的部分 (1 ) 中的靶细胞以外, 按照实施例 7的部分 (1 ) 进行实 验。
2. 结.果: 同 HSP65-PSA组相比较, PSA转染的 B16细胞作为靶细胞时, BCG+HSP65-PS A免疫组小鼠的脾及淋巴结 CTL在效靶比为 1: 20的杀伤率为 47%, HSP65-PSA融合蛋白免疫组小鼠的脾和淋巴结 CTL的杀伤率为 36%。这表明 BCG能够加 强 HSP65-PSA免疫所诱生的 PSA特异性的 CTL。BCG+HSP65-PSA和 HSP65-PSA免疫诱 生 PSA特异性 CTL强度的比较见图 17。
3. 结论: BCG能够显著加强 HSP65-PSA所诱生的特异性 CTL的作用。 实施例 26 BCG增强 HSP65-PSA免疫所激发的特异性抑瘤作用
1. 方法: 除了以 HSP65-PSA代替实施例 8部分 (1 ) 的 HSP65-MUC1 , 以转染 PSA 表位抗原多肽编码基因的 B16细胞代替实施例 8部分(1 )的 B16细胞以外, 按照实施例 8 的部分 (ί) 进行实验。
2. 结果: BCG+HSP65-PSA组小鼠的肿瘤重量显著地低于 HSP65—PSA组。各组间 肿瘤重量的比较见图 18。
3. 结论: BCG能够加强 HSP65-PSA免疫所激发的特异性抑瘤作用, 抑制表达 PSA肿瘤细胞的生长。 实施例 27 HSP65-HPV融合基因的获得
1. 采用五轮 PCR 成单拷贝多表位 HPV- Tet抗原基因
以下五轮 PCR条件均为: 94QC 30 sec, 55°C 30sec, 72°C 30sec, 25cycles. 循环结束 后, 72QC ®伸 10min。
1)、 第一轮 PCR (两条引物互为模板进行扩增)
引物 1的序列为 (SEQ ID NO: 33): -
5'-GTTTGGGCAGGCCTGCATTGCTACGAACAACTGGTAGATTCTTCTGAGGA CGAAGTT-3'
引物 Γ的序列为 (SEQ ID NO: 34):
CAGAAGA-3' te-01301V3VIW03V30XDI3V0DDIDVD31V00-.S οε
Figure imgf000026_0001
†7†7 N Cn 3S mi^m^-
,ε-工 voo丄:) Yiojyv丄工工 > i丄 ovoo丄工:) wOi coovi o丄 ooo工 oov -,s
■(zt ON cn
Figure imgf000026_0002
I έ
D丄 丄丄 COVLOVWODOO OE)工: JY09丄:) DIV YL OOV O工
'■(i N(H0as) fi *i Ι oz
。^^ ΐΐ7 ON cn bas am^m^-
.ε-OVDOVD 丄 丄 DVOV DDOlVOOVW DiDDOXVOJLVVDV IV O丄:) VXOIVYLL丄-, S
(ε-工丄 i£»丄:) 5i
:(6£ ΝΟΙ HS) Γ^*ί ε I έ
d^ ^Aii^^^^^x d^ ^w、(ε
.ε-νιονονο
■ i N (Ή HS) fi iiW.2
,ε-丄 v o丄:)
OD£VC)£) £)丄 1丄 £9丄 ICO IDVIDVDOOI O工丄工 O丄:) 10丄工 丄 IV£)£): -,S
g N Cn HS
Figure imgf000026_0003
¾3<I ¾一蚩
SIZ.I00/C00iM3/X3J 請 00ε OAV 引物 5'的序列是 (SEQ ID NO: 46):
5'-GGCAAGCTTAGATCTGGTACCCTCCAGTTTCTTCAGCTCGGTGATACCGAT-
3'
上游引物 5'端带有 BamH I酶切点, 下游引物 3'端带有 Kpnl、 Bgl II、 Hind III 酶切点。
产物的序列如 SEQ ID NO: 47所示, 即为 HPV-Tet序列。 2. 将 PCR产物克隆入载体质粒 pMD-18T(Takara)。
1) . 采用 TA 克隆方法将上述 PCR 产物克隆入 TA 载体, 得到的产物为 pMD-18T-HPV。方法见 TA连接试剂盒(Promega)按常规方法(见上文)提取质粒, 采用双脱氧末端终止法, 对插入片段进行序列测定 (ABIPriSm310TM,USA)。
图 19 HPV基因片段的 TA克隆
注: pMD18T-HPV质粒的酶切鉴定。
第 1道: DNA Marker
第 2道: pMD18T-HPV质粒酶切, 箭头示 HPV基因片段。
2) . 以上述的 pMD18-T-HPV抗原多表位基因重组质粒为模板, 用 PCR方法更换 HPV抗原多表位基因的酶切位点, 获取 HPV抗原多表位基因。
5' 端引物序列为 (SEQ ID NO: 48): 5 ' AAGCTTACTCCGACTCTGC AC-3 ' 3'端引物序列为 (SEQ ID NO: 49): 5'-GAATTCTCACTCCAGTTTCTTCAG-3'其 中, 5'端引物带有 EcoR I酶切位点, 3'端引物带有 Hind III酶切位点。 5'端引物带有 Hind m酶切位点, 3'端引物带有 EcoR I酶切位点。
PCR条件为: 94°C 30 sec, 55°C 30sec, 72°C 30sec, 25cycles. 循环结束后, 720C 延伸 10min。
PCR合成的产物序列如 SEQ ID NO: 50所示, 共计 360bp。 CACCGAGCTGAAGAAACTGGAGTGAGAATTC (360bp)
3) . 将热休克蛋白 65 (HSP65 ) 的编码基因和 pET28a (Novagen) 载体分别用 NcoI、 EcoRI 消化, 37°C , 2h。 如上采用琼脂糖凝胶电泳分离消化产物并回收消化 产物 (回收试剂盒为北京鼎国生物技术有限责任公司生产, 操作按说明书进行)。
将经 NcoI、 EcoRI消化的热休克蛋白 65 (HSP65) 的编码基因和 pET28a载体 用连接酶连接。将连接产物 pET28a— HSP65转化宿主菌 JM109,取阳性转化菌培养, 提取质粒(见上文), 采用双脱氧末端终止法, 对插入片段进行序列测定 (ABIPrism310™, USA)。
4) . 用 EcoR I和 Hind III分别消化步骤 2)中得到 HPV抗原多表位基因和 pET28a -HSP65质粒, 37°C, 2ho 如上采用琼脂糖凝胶电泳分离消化产物并回收消化产物
(回收试剂盒为北京鼎国生物技术有限责任公司生产, 操作按说明书进行)。
将回收产物用 T4DNA 连接酶连接。 连接产物 pET28a-HSP65-HPV 转化宿主菌 JM109, 取阳性转化菌培养, 提取质粒 (见上文), 采用双脱氧末端终止法, 对插入片段 进行序列测定(ABIPrism310TM,USA)。所得到的 HSP65-HPV融合基因与设计的基因 序列完全一致。
图 20 HSP65-HPV质粒的酶切鉴定
注: 第 1道: pET-28-HSP65-HPV酶切片段 第 2道: DNAMarker 实施例 28 HSP65-HPV融合蛋白的表达
挑取含有重组质粒 pET28a-HSP65-HPV的表达菌 BL21 DE3 (Novagen, America)单 菌落接种于含有 50 ml LB培养基的 250 ml三角烧瓶中, 37 °C水浴震荡培养至 OD600 值为 0.6。 加入 IPTG使其终浓度为 0.4 mM, 37°C水浴震荡培养 2-3h。将含有表达菌的 三角烧瓶置于冰上 5 min, 4°C离心 5min (5000 x g)。 吸弃上清, 收集细菌, 立即使用 或冻存。 . 实施例 29 重组融合蛋白 HSP65-HPV 的纯化
采用 10升的发酵罐表达重组 HSP65-HPV融合蛋白, 以乳糖为诱导物。在裂菌后, 经镍亲合层析、 除盐、 离子交换层析、 除盐后得到重组 HSP-HPV 融合蛋白。 取样做 HPLC分析, 重组 HSP65-HPV融合蛋白的纯度为 96.1%。 实施例 30 BCG和 HSP65— HPV体外诱导人的树突状细胞的活化和成熟 一、 如实施例 4的部分 (1 ) 所述分离人外周血单核细胞。
二、 如实施例 4的部分 (2) 所述诱导未成熟的树突状细胞。
三、 用 BCG和 HSP65-HPV融合蛋白装载树突状细胞
1 . 于第五天, 分别设立对照组: 只加培养液; BCG组: 加入 BCG使其终浓度达
30μ^ηι\; BCG+HSP65-HPV组: 加入 BCG和 HSP65-HPV使其终浓度分别为 30 g/ml和 100μβ/ιη1; HSP65-HPV组: 加入 HSP65-HPV使其终浓度达 lOO g/ml; 阳性对照组: 分 别加入下列因子使其终浓度分别为 1 Ong/mlT Fa、 1 Ong/mlIL-6、 1 Ong/mlEL- 1 β和 】t^g ni〗PGE2。 直接将上述的 BCG和 HSP65-HPV融合蛋白和细胞因子加入未成熟的树 突状细胞 (DC ) 中继续培养 2d。
2. 第 7d收获树突状细胞,进行活化和成熟指标的测定。
四、 如实施例 4的部分 (4) 所述进行树突状细胞活化和成熟指标的测定。
五、 结果
各组 CD86荧光值的比较见图 21。 流式细胞仪的测定结果表明: 同 HSP65-HPV组相 比较, BCG+HSP6 5-HPV能够显著地诱导 DC的活化和成熟。 DC是人体内最为重要的 抗原递呈细胞, DC的活化和成熟标志着 DC的抗原递呈能力加强, 激活针对 HPV的细胞 毒性杀伤性 T淋巴细胞 (CTL) 的能力加强; 人体内被激活的 CTL杀伤体内感染 HPV的 细胞, 因此, 可用于预防和或治疗 HPV感染。 实施例 31构建重组 HPV抗原多表位基因的真核细胞表达质粒
1、 以实施例 27构建的 pMDIS-T-HPV抗原多表位基因重组质粒为模板, 用 PCR 方法更换 HPV抗原多表位基因的酶切位点,获取拟亚克隆入真核细胞表达质粒的 HPV 抗原多表位基因。
5' 端引物序列为 (SEQ ID NO: 51):
5 '-AAGCTTACTCCGACTCTGCAC-3 '
3'端引物序列为 (SEQ ID NO: 52):
5'- GAATTCTCACTCCAGTTTCTTCAG-3'。 其中, 5'端引物带有 Hind ΙΠ酶切位 点, 3'端引物带有 EcoR I酶切位点。 PCR合成的产物序列如 SEQ ID NO: 53所示, 共计 360bp。
将 PCR产物克隆入 pcDNA3质粒:采用 Hind III和 EcoR I切点将 PCR产物亚克隆 至 pcDNA3质粒。 转化宿主菌 JM109, 取阳性转化菌培养, 提取质粒 (见上文), 采用双 脱氧末端终止法, 对插入片段进行序列测定 (ABn>rism310™, USA) )0
2. 用 pcDNA3-GFP-HPV质粒转染 B16细胞具体方法见实施例 5中的细胞转染。 实施例 32 BCG加强 HSP65-HPV注射所激发的特异性杀伤 HPV感染细胞的作用
1. 方法: 除了以 HSP65-HPV代替 HSP65-MUC1 , 以转染 HPV表位抗原多肽编码基 因的 B16细胞代替实施例 6部分 (1 ) 中的 B16细胞以外, 如实施例 6的部分 (1 ) 所述, 进行实验。
2. 结果: BCG+HSP65-HPV组小鼠的肿瘤重量显著低于 HSP65-HPV组(秩和检验,
PO.05 ),说明 BCG能够加强 HSP65-HPV注射所激发的特异性杀伤 HPV感染细胞的作用。 各组间肿瘤重量的比较见图 22。
3. 结论: BCG能够加强 HSP65-HPV所激发的小鼠体内特异性的杀伤 HPV感染 细胞的作用。 实施例 33 BCG加强 HSP65— PPV在小鼠诱生 HPV特异性杀伤性 T淋巴细胞的作用
1. 方法: 除了以 HSP65-HPV代替实施例 7的部分(1 ) 中 HSP65-MUC1 , 以 HPV表 位抗原多肽编码基因转染的 B16细胞及未转染 HPV表位抗原多肽编码基因的 B16细胞作 为靶细胞代替实施例 7的部分 (1 ) 中的靶细胞以外, 按照实施例 7的部分 (1 ) 进行实 验。
2. 结果: 同 HSP65-HPV组相比, HPV表位抗原多肽编码基因转染的 B16细胞作为 靶细胞时, BCG+HSP65-HPV免疫组的脾及淋巴结 CTL在效靶比为 1 : 20的杀伤率为 46%, HSP65-HPV融合蛋白免疫小鼠的脾和淋巴结 CTL杀伤率为 37%。各组诱生特异性 CTL作用的比较见图 23。
3. 结论: BCG能够加强 HSP65-HPV免疫所诱生的特异性 CTL的作用, 特异性的
CTL能够杀伤 HPV感染细胞。 实施例 34 BCG加强 HSP65-HPV免疫所激发的特异性杀伤 HPV感染细胞作用
1. 方法: 除了以 HSP65-HPV代替实施例 8部分(1 ) 的 HSP65-MUC1 , 以转染 HPV 表位抗原多肽编码基因的 B16细胞代替实施例 8部分(1 )的 B16细胞以外, 按照实施例 8 的部分 (1 ) 进行实验。
2.结果: BCG+HSP65-HPV 组小鼠的肿瘤重量显著低于 HSP65-HPV组。说明 BCG 能够加强 HSP65-HPV所激发的特异性杀伤 HPV感染细胞的作用。 各组间激发特异性杀 伤 HPV感染细胞作用的比较见图 24。
3. 结论: BCG能够加强 HSP65-HPV免疫所激发的特异性杀伤 HPV感染细胞的 作用。 实施例 35 单拷贝 SARS/3CL161.264表位抗原基因的合成
1、 第一轮 PCR (两条引物互为模板进行扩增)
引物 1的序列为 (SEQ ID NO: 54):
5'-ACTATCACTCTTAATGTTCTGGCTTGGCTGTACGCTGCTGTTATTAACGGTG ACCGC-3'
引物 Γ的序列为 (SEQ ID NO: 55):
5 '-GTTGAAATCGTTCAGGGTAGTAGTAAAGCGGTTCAGGAACCAGCGGTCAC CGTTAAT-3'
第一轮 PCR合成的产物的序列如 SEQ ID NO: 56
5'-ACTATCACTCTTAATGTTCTGGCTTGGCTGTACGCTGCTGTTATTAACGGTG ACCGCTGGTTCCTGAACCGCTTTACTACTACCCTGAACGATTTCAAC-3,。
' 2、 以第一轮 PCR产物为模板进行第二轮 PCR
引物 2的序列是 (SEQ ID NO: 57):
5'-TTCGTTGACCGTCAGACTGCACAGGCTGCTGGTACTGACACTACTATCACT CTTAATG-3'
引物 2'的序列是 (SEQ ID NO: 58):
5'-GTCTTGAGTCAGCGGTTCGTAGTTGTACTTCATAGCAACCAGGTTGAAATC GTTCAG-3'
产物的序列如 SEQ ID NO: 59所示。
3、 以第二轮 PCR产物为模板进行第三轮 PCR
引物 3序列是 (SEQ ID NO: 60):
5,-GTTCACGCAGGTACTGACCTGGAAGGTAAATTTTATGGTCCGTTCGTTGAC CGTCAG-3'
引物 3'的序列是 (SEQ ID NO: 61) :
5'-GATACCAGTCTGAGCAGACAGTGGACCCAGAATGTCTACGTGGTCTTGAG TCAGCGG-3'
产物的序列如 SEQ ED NO: 62所示。
4、 以第三轮 PCR产物为模板进行第四轮 PCR '
引物 4序列是 (SEQ ID NO: 63):
5'-GAATTCGATGAGTACATGCACCACATGGAACTGCCGACCGGTGTTCACGC AGGTACTG-3'
引物 4'的序列是 (SEQ ID NO: 64):
5'-CTCGAGTCACTAAAGCTTGTCTTCGTCGTCCTCCATATCCAGAACAGCGAT ACCAGTCTGAGC-3'
产物的序列如 SEQ ID NO: 65所示。 第四轮 PCR产物的 5'端带有 EcoRI酶切点, 下游引物 3'端带有 Hind III、 Xhol酶切点。该序列为一个拷贝的优化的 SA S冠状病毒 表位抗原序列。
PCR反应条件为: 94° C , 30" ; 55° C, ; 72° C, 2', 30个循环周期后, 72° C 延伸 10min。
5、 将第四轮 PCR产物克隆入载体 pMD-18T(Takara)
采用 TA克隆方法将 PCR产物克隆入 TA载体, 得到 pMD-18T-SARS/3CL161.264质粒, 方法见 (试剂盒, Takara)。
取阳性转化菌培养, 提取质粒 (见上文), 采用双脱氧末端终止法, 对插入片段进 行序列测定。
实施例 36 HSP65-SARS/3CL161-264融合基因的构建、 表达和纯化
1、 重组融合蛋白
Figure imgf000032_0001
实施例 35构建的 pMD-18T-SARS/3CL161-264质粒经 EcoRI和 Hindlll双酶消化后, 回收
SA S/3CL161-264的 DNA片段。 亚克隆连接至用同样的限制性内切酶消化并回收的 pET28a-HSP65线性载体, 即得到 HSP65- SARS/3CLI61.264。 图 25: pMD-18T- SARS/3CL161-264重组质粒的酶切鉴定
2、 HSP65-SARS/3CL161.264的表达和纯化
采用 10升的发酵罐培养含 HSP65-SA S/3CL161.264编码基因表达质粒的大肠杆菌 BL DE 3 (Novagen), 以乳糖为诱导物。 在裂菌后, 经镍亲合层析、 除盐、 离子交换层析、 除盐后得到 HSP65-SA S/3CL161-264。 取 10 μ 1样品上样进行 12%SDS-PAGE, 考马斯亮蓝 染色后, HSP65-SA S /3CL161.264出现为一个单一区带, 无杂带。 取 10 μ 1样品做 HPLC分 析, HSP65-SARS/3CL161-264的纯度为 96.1%。 HSP65-SARS/3CL161-264的 SDS-PAGE和 Western Blot鉴定见图 26: HSP65-SARS/3CL16i-264 SDS-PAGE鉴定及 Western blot鉴定分 析。 实施例 37 BCG和 HSP65— SARS体外诱导人的树突状细胞的活化和成熟
一、 如实施例 4的部分 (一) 所述分离人外周血单核细胞。
二、 如实施例 4的部分 (二) 所述诱导未成熟的树突状细胞。
三、 用 BCG和 HSP65-SA S融合蛋白装载树突状细胞
1. 于第五天, 分别设立对照组: 只加培养液; BCG组: 加入 BCG使其终浓度达 30 g/ml; BCG+HSP65-SA S组: 加入 BCG和 HSP65-SARS使其终浓度分别为 30μβ/ηι1 和 10(^g/ml; HSP65-SARS组: 加入 HSP65-SA S使其终浓度达 10(^g/ml; 阳性对照组: 分别加入下列因子并使其终浓度分别为 10ng/mlTNFa、 10ng/mlIL-6, lOng/mlIL-Ιβ和 10μβ/ηι1 PGE2。直接将上述的 BCG和 HSP65-SARS蛋白和细胞因子加入未成熟的树突状 细胞 (DC) 中继续培养 2d。
2. 7d收获树突状细胞,进行活化和成熟指标的测定。
四、 如实施例 4的部分 (四) 所述树突状细胞活化和成熟指标的测定。
五、 结果
各组间 CD86荧光值的比较见图 27。 流式细胞仪的测定结果表明: 同 HSP65-SARS 组相比较,' BCG+HSP65-SA S能够显著地诱导 DC的活化和成熟。 DC是人体内最为重 要的抗原递呈细胞, DC的活化和成熟标志着 DC的抗原递呈能力加强, 激活针对 SARS 的细胞毒性杀伤性 Γ淋巴细胞(CTL)的能力加强; 人体内被激活的 CTL杀伤体内 SARS 感染细胞, 因此, 可用于预防和或治疗 SARS感染。 实施例 38重组 SARS/3CL16 264表位抗原基因真核细胞表达质粒的构建
1 - 用 EcoRI和 Xhol同时消化实施例 35构建的 pMD-18T-SA S/3CL161^64质粒和携带 GFP基因的 pcDNA3-GFP质粒, 分别回收 SARS/3CL161_264基因片段和线性化的 pcDNA3-GFP质粒, 用 T4DNA连接酶将 SARS/3CL161.264基因片段和线性化的 pcDNA3-GFP质粒连接后, 形成重组质粒 pcDNA3-GFP- SA S/3CL161-264 , 用重组质粒 pcDNA3-GFP-SARS/3CL161.264转化宿主菌 JM109 , 取阳性转化菌培养, 提取质粒 (见上 文), 采用双脱氧末端终止法, 对插入片段进行序列测定。
2. 用 PcDNA3-GFP-SARS 质粒转染 B16细胞
利用携带绿色荧光蛋白 GFP基因的真核细胞表达质粒 pcDNA3-GFP, 把合成的
SARS/3CL161.264基因构建于 GFP基因的下游,使 GFP和 SARS/3CL161.264 基因同处于一个 阅读框架中, 得到了重组 pCDNA3- GFP-SARS/3CL16^64真核细胞表达质粒。用这种质粒 转染小鼠黑色素瘤细胞株 B16, 获得了表达 GFP-SARS/3CL161-264的转染细胞, 这种细胞 被我们用作 SARS Cov感染的细胞模型来检测 HSP65-SARS/3CL161-264的活性。
具体方法见实施例 5中的细胞转染。
pcDNA3-GFP -SARS/3CL161.264转染小鼠黑色素瘤 B16细胞的鉴定
收集 pcDNA3-GFP -SARS/3CL161.264转染的 B16细胞, 用共聚焦显微镜和 FACS技术 对阳性克隆进行鉴定,稳定转染 GFP-SA S/3CL,61.264基因的小鼠黑色素瘤 B16细胞在共 聚焦显微镜下细胞呈现绿色, FACS检测结果显示转染细胞中有较强的绿色荧光。
鉴定结果见图 28 实施例 39 BCG加强 HSP65-SARS注射所激发的特异性杀伤 SARS感染细胞的作用
1 . 方法: 除了以 HSP65-SARS代替 HSP65-MUC1 , 以转染 SARS表位抗原多肽编码 基因的 B16细胞代替实施例 6部分(1 ) 中的 B 16细胞以外, 如实施例 6的部分(1 )所述, 进行实验。
2.结果: BCG+HSP65-SA S组小鼠的肿瘤重量显著低于 HSP65-SA S组(秩和检验, P<0.05 ) , 明 BCG能够加强 HSP65-SA S注射所激发的特异性杀伤 SARS感染细胞的作 用。 各组激发特异性杀伤 SARS感染细胞作用的比较见图 29。 3. 结论: BCG能够显著加强 HSP65-SARS注射所激发的特异性杀伤 SARS感染细胞 的作用。 实施例 40 BCG加强 HSP65— SARS诱生 SARS特异性杀伤性 T淋巴细胞的作用
1. 方法: 除了以 HSP65-SA S代替实施例 7的部分(1 ) 中 HSP65-MUC1 , 以 SARS 表位抗原多肽编码基因转染的 B16细胞及未转染 SARS表位抗原多肽编码基因的 B16细 胞作为靶细胞代替实施例 7的部分 (1 ) 中的靶细胞以外, 按照实施例 7的部分 (1 ) 迸 行实验。 · 2. 结果: 同 HSP65-SA S组相比较, SARS表位抗原多肽编码基因转染的 B16细胞 作为靶细胞时, BCG+HSP65-SA S免疫组的脾及淋巴结 CTL的杀伤率为 46%。
HSP65-SARS融合蛋白免疫小鼠的脾和淋巴结 CTL杀伤率为 38%。 这表明 BCG能够加强 HSP65-SARS免疫所诱生的特异性 CTL的作用。 各组诱生特异性 CTL的作用的比较见图 30 ο
3. 结论: BCG能够加强 HSP65-SARS所诱生的特异性 CTL的作用, 所诱生的特异 性 CTL能够杀伤 HPV感染细胞。 实施例 41 BCG增强 HSP65-SARS免疫所激发的特异性杀伤 SARS感染细胞的作用
1.方法:除了以 HSP65-SARS代替实施例 8部分(1 )的 HSP65-MUC1, 以转染 SARS 表位抗原多肽编码基因的 B16细胞代替实施例 8部分(Γ)的 B16细胞以外, 按照实施例 8 的部分 (1 ) 进行实验。
2. 结果: BCG+HSP65-SARS 组小鼠的肿瘤重量显著低于 HSP65-SARS组。 说明 BCG能够加强 HSP65-SARS免疫所激发的特异性杀伤 SARS感染细胞的作用。 各组激发 特异性杀伤 SARS感染细胞作用的比较见图 31。
3. 结论: BCG能够显著加强 HSP65-SARS免疫所激发的特异性杀伤 SARS感染细胞 作用。 实施例 42 .多表位乙肝核心抗原基因的获得
采用二轮 PCR合成多表位乙肝核心抗原基因(多表位乙肝核心抗原基因以下简称 HBcAg)
1 ) 第一轮 PCR, 下列两条引物互为模板
引物 1的序列为:
5'-GGTTGTTTCTTACGTTAACGTAAACATGGGTCTGAAAATCCGTCAGCTGGC TCCGATCCTGTCCACTCTGCCAGAAACTACTGTTG-3'(SEQ ID NO: 66)
引物 Γ的序列为-
5'-TAGAGGACAGACGAGCAACGTAACGAGACAGACCGGAAGAACCGATGCT ACGACCACGACGACGAACAACAGTAGTTTCTGGCAG-3'(SEQ ID NO: 67)
第一轮 PCR获得产物的序列如 SEQ ID NO: 68所示。
2) 第二轮 PCR, 以第一轮 PCR的产物为模板
引物 2的序列是:
5'-GAATTCGAACTGCTGTCTTTCCTGCCGAGCGACTTCTTCCCGTCCGTTCGT GACCTGCTGCTGGTTGTTTCTTACGTTAACG-3'(SEQ ID NO: 69)
引物 2'的序列是:
5'-ACGCGTCGACCTGTTTAGCCTGGATGCAAGCGTACAGCGGCATCAGAGCC GGGTAACCGCAAGAGT1AGAGGACAGACGAGCAACG-3'(SEQ ID NO: 70)
第二轮 PCR获得产物的序列如 SEQ ID NO: 71所示, 此序列代表的是乙肝核心抗 原多表位基因。
上述 PCR的反应条件为: 94°C, 30"; 55°C, Γ; 72°C, 4', 30个循环周期后, 72°C 延伸 10分钟。 实施例 43 HSP65-HBcAg融合基因的构建
将热休克蛋白 65 (HSP65 )的编码基因用 Ncol和 EcoRI消化, 多表位乙型肝炎 病毒核心抗原基因用 EcoRI和 Hindlll消化, 37°C, 2小时。 如上经琼脂糖凝胶电泳 分离消化产物并回收消化产物。
将经限制性内切酶消化的热休克蛋白 65KD (HSP65 )的编码基因和多表位乙型肝 炎病毒核心抗原基因用 T4DNA连接酶连接, 即得到 HSP65-HBcAg (简称 HSP-HBV)。 HSP-HBV的核苷酸序列如 SEQ ID NO: 72所示。 实施例 44 HSP-HBV融合基因的克隆
用 Ncol和 Hindlll在 37°C消化实施例 43中得到的连接产物, 克隆入经限制性内 切酶 Ncol和 Hindlll消化的原核细胞表达载体 pET-28a(+)质粒 (美国 Novagen公司) 的 6聚组氨酸 (histidine) 密码子的上游, 即得到 pET-28a(+)- HSP65-HBcAg。
将含 HSP65-HBcAg的重组质粒 pET-28a(+)-HSP65-HBcAg转化大肠杆菌, 取阳性菌 培养, 提取质粒 (见上文), 采用双脱氧末端终止法, 对插入片段进行序列测定。 实施例 45 HSP65-HbcAg (简称 HSP-HBV)重组融合蛋白的表达
将单菌落的细菌接种于 50 ml LB培养基中, 于 250 ml三角烧瓶中, 37Q C水浴 震荡培养至 OD600值为 0.6。加入 IPTG使其蜂浓度为 0.4 mM, 37° C水浴震荡培养 2-3小时。 置三角烧瓶于冰上 5分钟, 4 GC离心 5分钟(5000 x g)。 吸弃上清, 收集 细菌, 立即使用或冻存。
图 32 HSP65-HBV重组融合蛋白表达图谱 实施例 46 HSP-HBV重组融合蛋白的纯化
将上述含 HSP-HBV重组融合蛋白的大肠杆菌细胞 (3g湿重) 于室温下解冻, 重 悬于 20ml结合液中, 冰浴 2小时后, 离心 20分钟 (10000Xg), 收集上清备用。
结合液的配制:
20mmoVL Tris.HCL(pH7.9)
0.5 mol/LNaCL
5mmol/L咪唑
6mol/L尿素
将上清液加入镍亲和层析柱 (lXlOcm)上, 洗涤后洗脱目的蛋白, 即得到
HSP-HBV重组融合蛋白。
图 33 SDS-PAGE鉴定纯化蛋白图谱 实施例 47 BCG和 HSP65— HBcAg体外诱导人的树突状细胞的活化和成熟
一、 如实施例 4的部分 (一) 所述分离人外周血单核细胞。 二、 如实施例 4的部分 (二) 所述诱导未成熟的树突状细胞。
三、 用 BCG和 HSP-HBV融合蛋白装载树突状细胞
1. 于第五天, 分别设立对照组: 只加培养液; BCG组: 加入 BCG使其终浓度达 3(^g/ml ; BCG + HSP-HBV组: 加入 BCG和 HSP-HBV使其终浓度分别为 3(^g/ml和 100μβ/ιη1; HSP-HBV组: 加入 HSP-HBV使其终浓度为 lOO g/ml; 阳性对照组: 分别加 入下列因子使其终浓度分别为 10ng/mlTNFa、 10ng/mlIL-6、 lOng/mlIL-Ιβ和 l(^g/mlPGE2。 直接将上述的 BCG和 HSP-HBV蛋白加入未成熟的树突状细胞 (DC) 中 继续培养 2d。
2. 7d收获树突状细胞, 进行活化和成熟指标的测定。
四、 如实施例 4的部分 (四) 所述进行树突状细胞活化和成熟指标的测定。
五、 结果
各组间 CD86荧光值的比较见图 34。流式细胞仪的测定结果表明: 同 HSP-HBV组相 比较, BCG+HSP65- HBcAg能够显著地诱导 DC的活化和成熟。 DC是人体内最为重要 的抗原递呈细胞, DC的活化和成熟标志着 DC的抗原递呈能力加强,激活针对 HBV的细 胞毒性杀伤性 T淋巴细胞(CTL) 的能力加强; 人体内被激活的 CTL杀伤体内感染 HBV 的细胞, 因此, 可用于预防和或治疗 HBV感染 。 实施例 48'转染 pcDNA3—GFP— HBV的 B16细胞的构建
1. 用 EcoRI和 Hindlll酶切位点将乙型肝炎病毒核心抗原基因片段 (实施 例 38中得到的) 克隆入转染载体 pcDNA3— GFP (Invitrogen), 形成 pcDNA3— GFP 一乙型肝炎病毒核心抗原基因重组质粒 (pcDNA3— GFP— HBV)。
2. 用 pcDNA3-GFP-HBV质粒转染 B16细胞具体方法见实施例 5中的细 胞转染。 实施例 49 BCG加强 HSP-HBV注射所激发的特异性杀伤 HBV感染细胞的作用
1. 方法: 除了以 HSP-HBV代替 HSP65-MUC1, 以转染 HBV表位抗原多肽编码基因 的 B16细胞代替实施例 6部分 (1 ) 中的 B16细胞以外, 如实施例 6的部分 (1 ) 所述, 进 行实验。
2. 结果: BCG+HSP-HBV组小鼠的肿瘤重量显著低于 HSP-HBV组 (秩和检验, P<0.05 ),说明 BCG能够加强 HSP-HBV注射所激发的特异性杀伤 HBV感染细胞作用。 各 组所激发的特异性杀伤 HBV感染细胞作用的比较见图 35。
3. 结论: BCG能够加强 HSP-HBV注射所激发的特异性杀伤 HBV感染细胞的作用。 实施例 50 BCG加强 HSP65— HBcAg在小鼠诱生 HBV特异性杀伤性 T淋巴细胞作用 1. 方法: 除了以 HSP-HBV代替实施例 7的部分(1 ) 中 HSP65-MUC1 , 以 HBV表位 抗原多肽编码基因转染的 B16细胞及未转染 HBV表位抗原多肽编码基因的 B16细胞作 为靶细胞代替实施例 7的部分 (1 ) 中的靶细胞以外, 按照实施例 7的部分 (1 ) 进行实 验。 2. 结果: 同 HSP-HBV组相比较, HBV表位抗原多肽编码基因转染的 B16细胞作为 靶细胞时, · BCG+HSP65- HBcAg免疫组的脾及淋巴结 CTL的杀伤率为 49%。 HSP-HBV 融合蛋白免疫小鼠的脾和淋巴结 CTL杀伤率为 38%。这表明 BCG能够加强重组融合蛋白 HSP-HBV免疫所诱生的特异性杀伤 HBV感染细胞的作用。 各组诱生特异性 CTL杀伤 HBV感染细胞作用的比较见图 36。 实施例 51 BCG增强 HSP-HBV免疫所激发的特异性杀伤 HBV感染细胞作用
1. 方法: 除了以 HSP-HBV代替实施例 8部分 (1 ) 的 HSP65-MUC1 , 以转染 HBV 表位抗原多肽编码基因的 B16细胞代替实施例 8部分(1 )的 B16细胞以外, 按照实施例 8 的部分 (1 ) 进行实验。
2.结果: BCG+HSP-HBV组小鼠的肿瘤重量显著低于 HSP-HBV组小鼠的肿瘤重 量。说明 BCG能够增强 HSP65— HBcAg免疫所激发的特异性杀伤 HBV感染细胞的作用。 各组激发的特异性杀伤 HBV感染细胞的作用比较见图 37。
3. 结论: BCG能够增强 HSP-HBV免疫所激发的特异性杀伤 HBV感染细胞作用。 实施例 52合成单拷贝多表位丙型肝炎病毒核心抗原基因
采用两轮 PCR合成单拷贝多表位丙型肝炎病毒 (HCV)核心抗原基因
1. 第一轮 PCR (两条引物互为模板进行扩增)
引物 1的序列为:
5'-ATGGGTTACATCCCGCTGGTTGGTGCTCCGCTGGAAGACTCTGAAGGTGTT TACCTGCTGCCGCGTCGTGGGCCGCGCCTGGGCGTTCnC-3'(SEO ΤΏ ΝΟ·. 73) 引物 Γ的序列为:
5*-GTCGTTACGTTCAGAAGTTTTACGAGTAGCACGAACACCCAGACGCGGAC CTTCGATTTCGTCGTTTTCAGCGCGAACGCCCAGGCGCGG-3'iSEO ID NO: 74) PCR的反应条件为: 94°C , Γ; 60°C, 1'; 72°C, 2', 3个循环周期后? , 72°C延伸 10分钟。
第一轮 PCR合成的产物的序列如 SEQ ID NO: 75所示。
2. 以第一轮 PCR产物为模板进行第二轮 PCR
引物 2的序列是:
5'-GAATTCATGTCTACTAACCCGAAACCGCAGCGTAAAACTAAAGAAATCGA CGACTTCGCTGACCTGATGGGTTACATCCCGCTG-3' (SEQ ID NO: 76)
引物 2'的序列是
5'-AAGCTTTTCGTTGTCGATTTCCTGACGACGACCACGCGGCTGAGAACGTT CGGAGGTTTTACGCAGTTCGTCGTTACGTTCAGAAGT- 3YSEO ID NO: 77)
PCR的反应条件为: 94°C , r; 60°C, 1'; 72°C, 2', 30个循环周期后, 72°C延伸
10分钟。 产物的序列是如 SEQ ID NO:78所示, 此序列为 HCV核心抗原基因。
3. 将 PCR产物克隆入载体质粒 pMD18-T(Takara): 采用 TA克隆方法将 PCR产物 (HCV核心抗原基因)克隆入 TA载体, 得到的产物为 pMD-18T-HCV。 方法见 TA连 接试剂盒 (Promega)。 转化宿主菌 JM109后, 取阳性克隆进行培养, 按常规方法 (见 上文)提取质 , 采用双脱氧末端终止法, 对插入片段进行序列测定(A^n>rism310™, USA) o 实施例 53 HSP65基因的克隆
同实施例 1 实施例 54构建 HSP65和多表位 HCV核心抗原基因融合基因
将 pET28(a)-HSP65重组质粒和 pMD-18T-HCV重组质粒分别用 EcoRI和 Hindlll (Takara) 消化, 37'C, 2小时。 如上采用琼脂糖凝胶电泳分离消化产物并回收消化 产物 (回收试剂盒为北京鼎国生物技术有限责任公司生产, 操作按说明书进行)。
将经 EcoRI/Hindlll消化的 pET28(a)-HSP65和多表位 HCV核心抗原基因用 T4 DNA连接酶 (Takara)连接, 即得到 pET28(a)-HSP65-HCV。连接产物转化感受态细胞 JM109, 取阳性克隆进行培养, 按常规方法 (见上文) 提取质粒, 采用双脱氧末端 终止法, 对插入片段进行序列测定 (ABn»rism310™,USA)。
图 38 HSP65-HCV质粒酶切图谱
第 1道: DAN Marker
第 2道: HCV多表位基因片段 (长度为 300bp左右)。 实施例 55 HSP65-HCV融合基因的表达
将上述转化了融合基因的大肠杆菌 JM109的单菌落接种于 1000 ml LB培养基 中。 37°C水浴震荡培养至 OD600 为 0.6。 加入 EPTG使其终浓度为 1 mM, 37°C水 浴震荡培养 2-3小时。 三角烧瓶于冰上 5 分钟, 4°C离心 5分钟(5000 x g)。 吸弃 上清, 收集细菌, 立即使用或冻存。 实施例 56 HSP65-HCV融合蛋白的纯化
将上述大肠杆菌细胞于室温下解冻。每克菌(湿重)重悬于 10ml裂解缓冲液。 每毫升裂解缓冲液加 1M MgS04 lOul, 10 ug/ml DNase 1 10ul。 冰上放置 30min。
12,000 πι, 4 °C离心 15 min。 弃上清。 每克菌加 1.5ml 结合缓冲液。
冰上放置 2h。 12,000 ipm, 4 °C离心 15 min。 留取上清, 做 Ni2+-Saphrose-4-B 层析 (层析介质购自 Pharmacia公司) , 分离 HSP65-HCV融合蛋白。
洗脱的目的蛋白进行苯基 Sepharose疏水层析 (层析介质购自 Pharmacia公司) 去除内毒素。
采用葡聚糖凝胶 Sephadex-G-25离子交换层析 (层析介质购自 Pharmacia公司) 去除目的蛋白中的盐类。
取 10 μ 1样品用 HPLC进行测定分析,结果表明: HSP65-HCV融合蛋白纯度为: 95%。 图 39 HSP65-HCV蛋白质表达纯化的 SDS-PAGE结果
注: 第 1、 2道为大肠杆菌中表达的 HSP65-HCV蛋白
第 3、 4道为纯化后 HSP65-HCV蛋白。 实施例 57 BCG和 HSP65— HCV体外诱导人的树突状细胞的活化和成熟
一、 如实施例 4的部分 (一) 所述分离人外周血单核细胞。 二、 如实施例 4的部分 (二) 所述诱导未成熟的树突状细胞。
三、 用 BCG和 HSP65-HCV融合蛋白装载树突状细胞
1 . 于第五天, 分别设立对照组: 只加培养液; BCG组: 加入 BCG使其终浓度达 3(^g/ml; BCG+HSP65-HCV组: 加入 BCG和 HSP65-HCV使其终浓度分别为 30μβ/ιη1和 lOO g/ml; HSP65-HCV组: 加入 HSP65-HCV使其终浓度为 lOO g/ml; 阳性对照组: 分 别加入下列因子使其终浓度分别为 10ng/ml TNFa> 10ng/ml IL-6、 lOng/ml IL-Ιβ和 l(^g/ml PGE2。 直接将上述的 BCG和 HSP65-HCV蛋白加入未成熟的树突状细胞 (DC) 中继续培养 2d。
2. 7d收获树突状细胞,进行活化和成熟指标的测定。
四、 如实施例 4的部分 (四) 所述进行树突状细胞活化和成熟指标的测定。
五、 结果
各组间 CD86荧光值的比较见图 40。流式细胞仪的测定结果表明: 同 HSP65-HCV组 相比较, BCG+HSP65- HCV能够显著地诱导 DC的活化和成熟。 DC是人体内最为重要 的抗原递呈细胞, DC的活化和成熟标志着 DC的抗原递呈能力加强,激活针对 HBV的细 胞毒性杀伤性 T淋巴细胞 (CTL) 的能力加强; 人体内被激活的 CTL杀伤体内感染 HCV 的细胞, 因此, 可用于预防和或治疗 HCV感染 。 实施例 58 转染 HCV的 B16细胞的构建
1. 质粒 pET26b-8R ( 8R为八聚精氨酸, 是一段由 8个精氨酸残基连接而成短肽, 构建 8R-多拷贝多表位 HCV核心抗原融合蛋白, 用于转染细胞。 方法同前)。用 BamH
I和 Xho l (Takara) 消化, 用琼脂糖凝胶电泳分离并回收线性化载体; 实施例 47中得 到的 pMD-18T-HCV重组质粒同样用 BamH I和 Xho l消化, 回收 HCV DNA片段。 将 二者用 T4DNA连接酶连接, 转化感受态菌 JM109。 提取阳性重组质粒 (方法同前)。
2. 用 pET26b-8R-HCV质粒转染 B16细胞具体方法见实施例 5中的细胞转染。 实施例 59 BCG加强 HSP65-HCV注射所激发的特异性杀伤 HCV感染细胞的作用
1. 方法: 除了以 HSP65-HCV代替 HSP65-MUC1 , 以转染 HCV表位抗原多肽编码 基因的 B16细胞代替实施例 6部分 (1 ) 中的 B16细胞以外, 如实施例 6的部分 (1 ) 所 述, 进行实验。
2. 结果: BCG+HSP65-HCV组小鼠的肿瘤重量显著低于 HSP65-HCV组 .(秩和检 验, P<0.05 ) ,说明 BCG能够加强 HSP65-HCV注射所激发的特异性杀伤 HCV感染细胞 作用。 各组所激发的特异性杀伤 HCV感染细胞作用的比较见图 41。
3. 结论: BCG能够加强 HSP65-HCV注射所激发的特异性杀伤 HCV感染细胞的作 用。 实施例 60 BCG加强 HSP65— HCV在小鼠诱生 HCV特异性杀伤性 T淋巴细胞作用
1. 方法: 除了以 HSP65-HCV代替实施例 7的部分 (1 ) 中 HSP65-MUC1 , 以 HCV 表位抗原多肽编码基因转染的 B16细胞及未转染 HCV表位抗原多肽编码基因的 B16细 胞作为靶细胞代替实施例 7的部分 (1 ) 中的靶细胞以外, 按照实施例 7的部分 (1 ) 进 行实验。.
2. 结果: 同 HSP65-HCV组相比较, HCV表位抗原多肽编码基因转染的 B 16细胞作 为靶细胞时, BCG+HSP65- HCV免疫组的脾及淋巴结 CTL的杀伤率为 48%。 HSP65-HCV 融合蛋白免疫小鼠的脾和淋巴结 CTL杀伤率为 38%。这表明 BCG能够加强重组融合蛋白 HSP65-HCV免疫所诱生的特异性杀伤 HCV感染细胞的作用。 各组诱生特异性 CTL杀伤 HCV感染细胞作用的比较见图 42。 实施例 61 BCG增强 HSP65-HCV免疫所激发的特异性杀伤 HCV感染细胞作用
1. 方法: 除了以 HSP65-HCV代替实施例 8部分(1 ) 的 HSP65-MUC1 , 以转染 HCV 表位抗原多肽编码基因的 B16细胞代替实施例 8部分(1 )的 B16细胞以外, 按照实施例 8 的部分 (1 )进行实验。
2.结果: BCG+HSP65-HCV组小鼠的肿瘤重量显著低于 HSP65-HCV 组小鼠的肿 瘤重量。 说明 BCG能够增强 HSP65— HCV免疫所激发的特异性杀伤 HCV感染细胞的作 用。 各组激发的特异性杀伤 HCV感染细胞的作用比较见图 43。
3. 结论: BCG能够增强 HSP65-HCV免疫所激发的特异性杀伤 HCV感染细胞作 用。

Claims

权 .利 要 求
1 一种疫苗组合物, 其特征在于包括重组融合蛋白和佐剂, 所述重组融合蛋 白是融合了热休克蛋白的融合蛋白, 所述佐剂为卡介苗。
2 如权利要求 1的疫苗组合物,其特征在于所述热休克蛋白是结核分枝杆菌热 休克蛋白 65。
3 ' 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65位于氨基端的融合蛋白。
4 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65融合粘蛋白 1构成的重组融合蛋白 HSP65-MUC1。
5 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65融合人上皮细胞生长因子受体 2构成的重组融合蛋白 HSP65-HER2。
6 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65融合人前列腺特异抗原构成的重组融合蛋白 HSP65-PSA。
7 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65融合乳头瘤病毒多表位抗原构成的重组融合蛋白 HSP65-HPV。
8 ' 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65融合 SARS病毒抗原构成的重组融合蛋白 HSP65-SARS。
9 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65融合乙型肝炎核心抗原构成的重组融合蛋白 HSP-HBV。
10 如权利要求 2的疫苗组合物,其特征在于所述重组融合蛋白是结核分枝杆菌 热休克蛋白 65融合丙型肝炎抗原表位构成的重组融合蛋白 HSP65-HCV。
11 权利要求 1-10任一项的疫苗组合物在制备预防和治疗肿瘤的药物中的用 途。
12 如权利要求 11的用途, 所述肿瘤包括但不限于人乳腺癌、 直肠癌、 结直肠 癌、 喉癌、 胃癌、 膀胱癌、 胰腺癌、 前列腺癌、 卵巢癌、 结肠癌、 肺癌。
13 权利要求 1-10任一项的疫苗组合物在制备预防和治疗病毒感染所引起的疾 病的药物中的用途。
14 如权利要求 13的用途,所述的病毒感染所引起的疾病包括 SARS病毒感染疾 病、 乳头瘤病毒感染、 宫颈癌、 丙型肝炎、 乙型肝炎、 丙型肝炎病毒感染和乙型肝 炎病毒感染导致的肝癌、 肝硬化。
PCT/CN2005/001715 2004-10-19 2005-10-19 Composition vaccinale contenant une proteine de fusion recombinee ainsi qu'un adjuvant et son application WO2006042465A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNA2004100840458A CN1762491A (zh) 2004-10-19 2004-10-19 Bcg作为激活细胞毒性t淋巴细胞活性蛋白的佐剂
CN200410084045.8 2004-10-19

Publications (1)

Publication Number Publication Date
WO2006042465A1 true WO2006042465A1 (fr) 2006-04-27

Family

ID=36202676

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2005/001715 WO2006042465A1 (fr) 2004-10-19 2005-10-19 Composition vaccinale contenant une proteine de fusion recombinee ainsi qu'un adjuvant et son application

Country Status (2)

Country Link
CN (1) CN1762491A (zh)
WO (1) WO2006042465A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021243417A1 (en) * 2020-06-05 2021-12-09 Centenary Institute Of Cancer Medicine And Cell Biology Immunogenic formulations

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1362263A (zh) * 2001-01-04 2002-08-07 北京迪威华宇生物技术有限公司 预防和治疗人前列腺癌的重组蛋白疫苗
CN1368384A (zh) * 2001-02-08 2002-09-11 北京迪威华宇生物技术有限公司 人乳腺癌muc-1抗原基因工程疫苗
CN1410127A (zh) * 2001-10-10 2003-04-16 北京迪威华宇生物技术有限公司 一种卡介苗热休克蛋白65和多表位her-2抗原融合蛋白重组蛋白疫苗
CN1462636A (zh) * 2002-05-30 2003-12-24 北京迪威华宇生物技术有限公司 预防和治疗人丙型肝炎病毒感染的重组蛋白疫苗及其用途

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1362263A (zh) * 2001-01-04 2002-08-07 北京迪威华宇生物技术有限公司 预防和治疗人前列腺癌的重组蛋白疫苗
CN1368384A (zh) * 2001-02-08 2002-09-11 北京迪威华宇生物技术有限公司 人乳腺癌muc-1抗原基因工程疫苗
CN1410127A (zh) * 2001-10-10 2003-04-16 北京迪威华宇生物技术有限公司 一种卡介苗热休克蛋白65和多表位her-2抗原融合蛋白重组蛋白疫苗
CN1462636A (zh) * 2002-05-30 2003-12-24 北京迪威华宇生物技术有限公司 预防和治疗人丙型肝炎病毒感染的重组蛋白疫苗及其用途

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
LI D ET AL: "Priming of mice lymphocytes for secretion of IFN by heat sock protein-MUCI fusion protein.", JOURNAL OF JILIN UNIVERSITY (MEDICINE EDITION)., vol. 30, no. 3, May 2004 (2004-05-01), pages 405 - 408 *
LI Q.: "Use of genetic recombinant BCG for preventing and curing disease.", CHIN PREV MED., vol. 5, no. 4, August 2004 (2004-08-01), pages 317 - 319 *
SHAO M ET AL: "Construction and expression of HBcA epitope fusion gene.", J MED MOL BIOL., vol. 1, no. 2, 2004, pages 77 - 80 *
TIAN Y ET AL: "Site-directed mutagenesis of HPV Multi-Epitope fusion protein.", J MED MOL BIOL., vol. 1, no. 1, 2004, pages 10 - 14 *
WANG L ET AL: "Amplification and sequence analysis of fusion protein gene of BCG HSP65 and human HER2/neu T cell epitope.", J BENGBU MED COLL., vol. 27, no. 4, July 2002 (2002-07-01), pages 292 - 294 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021243417A1 (en) * 2020-06-05 2021-12-09 Centenary Institute Of Cancer Medicine And Cell Biology Immunogenic formulations

Also Published As

Publication number Publication date
CN1762491A (zh) 2006-04-26

Similar Documents

Publication Publication Date Title
CN108434450B (zh) 基于铁蛋白纳米颗粒的疫苗及其制备方法
EP0013828B1 (en) Recombinant DNA, hosts transformed with it and processes for the preparation of polypeptides
JP5200201B2 (ja) 外来抗原に対する宿主の寛容を壊すためのキメラ抗原
JP4056306B2 (ja) C型肝炎ウイルスアシアロ糖タンパク質
JP2014039557A (ja) 免疫応答を誘導するためのC型肝炎ウイルスポリペプチドおよびFcフラグメントを含むキメラ抗原
KR101181907B1 (ko) 자궁경부암 백신
JP2011521985A (ja) Hcv感染の予防および治療のためのワクチン
JP2008543301A (ja) フィブロネクチンのedaドメインの使用を基にした剤および方法
TW201300418A (zh) 用於抗c型肝炎病毒免疫之組合物及方法
KR20040034511A (ko) 인간 파필로마바이러스에 대한 백신용 벡터 및 그에 의해형질전환된 미생물
KR20190079628A (ko) 백신으로 사용하기 위한 엑소좀-앵커링 단백질을 발현하는 뉴클레오티드 서열
TW200932908A (en) Antigenic compositions and use of same in the targeted delivery of nucleic acids
CA2905511A1 (en) Therapeutic cancer vaccine targeted to haah (aspartyl-[asparaginyl]-beta-hydroxylase)
EP0182442A2 (en) Recombinant DNA molecules and their method of production
EP0518313A2 (en) Gene of hepatitis C virus or fragment thereof, polypeptide encoded by the same
WO2006042465A1 (fr) Composition vaccinale contenant une proteine de fusion recombinee ainsi qu&#39;un adjuvant et son application
WO2022127820A1 (zh) 病原样抗原疫苗及其制备方法
CN114717205A (zh) 一种冠状病毒RBDdm变异体及其应用
CN105367662B (zh) 一种hbv相关的融合蛋白、其制备方法及其应用
Deckhut et al. Localization of common cytotoxic T lymphocyte recognition epitopes on simian papovavirus SV40 and human papovavirus JC virus T antigens
CN105801704B (zh) 一种具有抗宫颈癌细胞活性的重组疫苗构建方法及应用
KR960004264B1 (ko) B형 간염 표면항원, 이의 생성 진핵 세포 및 백신의 제조방법, 검정방법 및 진단용 키트
Bandehpour et al. Cloning and expression of Hepatitis B surface antigen
Parizad et al. Comparison of Immune Response in Mice Immunized with Recombinant PreS2/S-C18-27 Protein Derived from Hepatitis B Virus with Commercial Vaccine
CN117203221A (zh) 用于人类乳突病毒的dna疫苗及其使用方法

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05801963

Country of ref document: EP

Kind code of ref document: A1