HSP65-MUC1通过分子模拟损伤小鼠胰腺的可能性研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
  • 英文题名:Research on Possible Damage of Murine Pancreas by HSP65-MUC1 via Molecular Mimicry
  • 作者:孙琳
  • 论文级别:博士
  • 学科专业名称:免疫学
  • 学位年度:2007
  • 导师:于永利
  • 学科代码:100102
  • 学位授予单位:吉林大学
  • 论文提交日期:2007-04-01
摘要
卡介苗热休克蛋白65-MUC1多肽融合蛋白(HSP65-MUC1)是一种治疗肿瘤的重组蛋白疫苗,在进入树突状细胞(DCs)后,经MHCI类途径加工提呈,可激活MUC1特异性CTL去杀伤表达MUC1抗原的肿瘤。
     因为卡介苗热休克蛋白65(HSP65)和人体热休克蛋白60(HSP60)存在高度同源性,且人胰岛β细胞高表达HSP60,因此在应用HSP65-MUC1后可能通过分子模拟的方式诱导人体产生针对HSP60的交叉免疫应答,进而造成胰腺β细胞的破坏,引发自身免疫性糖尿病。
     为了证实HSP65-MUC1的安全性,采用正常小鼠和荷瘤小鼠,我们研究了HSP65-MUC1通过分子模拟损伤小鼠胰腺的可能性。结果表明:1、注射后的HSP65-MUC1未在胰腺大量滞留;2、HSP65-MUC1可在小鼠诱生HSP65特异性的细胞毒性T淋巴细胞(CTL);3、HSP65-MUC1诱生的HSP65特异性淋巴细胞能和小鼠的HSP(包括HSP60)发生交叉反应;4、反复注射HSP65-MUC1的小鼠未发生明显的胰腺损伤。
     本工作为HSP65-MUC1临床应用的安全性提供了重要参考。
People expected the treatment with better target and lower toxicity all the times, for traditional radiative treatment and chemical treatment killed normal cell as killing tumor cell. Tumor vaccine belong to one kind of independent tumor biology treatment, yet, it usually combined with standard treatment.The ideal tumor vaccine scarcely harmed the normal cells when it got rid of tumor cells.By contraries,Some tumor vaccine prolong patients’life without reduce tumor size compared with former chemical drug,which become one of standards for evaluating tumor vaccine treatment was curative and effective or not.it is neccssary to carry safety and curative rearch on animals for developing tumor vaccine,then,enter stage III of clinical test.
     HSP-peptide, tumor vaccine, has some charatrers as follows: (1) HSP-peptide was not restricted to recognize special antigen epitope,(2) HSP-peptide induced special CTL that killed tumor cell specially,(3) HSP-peptide can be immunized without adjuvant.
     As vaccine anti-lung cancerHSP65-MUC1 involved two parts:heat BCG shock protein 65 and MUC1 CTL epitope.The mechnism of fusion protein was HSP65 transfer MUC1 CTL epitope to DCs where CTL epitope was processed and presented by MHCI pathway, then, CTL was activated by epitope.
     HSP kept conservation during evolution. great similarity existed between HSP from other kinds of pathogens and human HSP,when bacteria invade host, T lymphocytes were primed by HSP60 as foreign antigen through MHCI molecular, since highly homology existed between mammal HSP60 and bacteria HSP60,autoimmunity reaction occured when primed T lymphocytes recongnized self antigen,HSP60, by molecular mimicry.Autoimmune disease was aroused further.
     So,HSP65-MUC1 fusion protein acted as vaccine to treat lung cancer, researching on curative and safety as HSP65-MUC1vaccine was applied, we concentrate on three parts.
     The first part made research whether absorption and distribution of tumor vaccine was localized selectively or not, the second part made research whether cross reaction happened under different immunity mode, and cross reaction will give rise to insulin dependent diabetes mellitus. The third part made research on antitumor effective of tumor vaccine on mice which were inoculated with tumor. It basically proved that tumor vaccine developed by our lab was able to induce T cell mediated immune response to inhibit MUC1 positive tumor cell growing, and well done in treatment. MUC1 express highly on ovary cancer, pancrea cancer, liver cancer, stomach cancer, lung cancer, prostate cancer, so, HSP65-MUC1, gene engineering vaccine, was may valuable to therapy and prevention of these cancer. Our work involved several parts as following:
     1.absorption and distribution in mice
     To be sure of HSP65-MUC1 destroying tissue by settling, fluorescence quantitative test detect distribution of HSP65-MUC1 in mice.
     1.1 Acquirements of HSP65-MUC1, HSP65 and BSA labeled by FITC
     Under alkaline condition, FITC labeled HSP65-MUC1, HSP65 and BSA by blending, at low temperature, overnight. The dissociative FITC was filtrated by G25. Protein labeled with FITC was qualified in purity and speciality.
     1.2 Establishment of fluorescence protein tracer methode
     First,Total protein concentration in homo-plasm was measured by standard specimen of BSA.Three kinds of fluorescence protein in homo-plasm was determined measured by standard specimen ofthree fluorescence protein . Calculating mass of three fluorescence protein in unit tissue from mice that immunized with three fluorescence protein.
     Calculational formula as follow :
     Mass of fluorescence protein (sample-blank)/ Mass of total protein=Mass of fluorescence protein in unit tissue
     1.3 Research on HSP65-MUC1 distribution in mice
     When mice were immunized with protein labeled with FITC in 40μg dose and for 70 min. Three kinds of protein all distribute a largest amount of drug in intestine, the next one was kidney and liver, the distribute in spleen, lung, pancrear were found less and the distribute in pancrear was the least one among three tissue. The least one was found in heart.
     1.4 Analysis of HSP65-MUC1 on drug metabolize dynamics
     When mice were immunized with protein labeled with FITC in 40μg dose and for different time, Three kinds of protein all distribute a largest amount of drug in intestine, The least one was found in heart. Except 240min of immunization, distribution in kidney and liver was inferior to distribution in intestine; there is increasing amount of protein in spleen, on the contrary, it is decreasing in distribution in lung and pancrear which is slightly much more than quantity of heart.
     1.5 Analysis of time- and dose-dependent on HSP65-MUC1
     Three kinds of protein all distribute a largest amount of drug in intestine, the next one was kidney and liver, the distribute in spleen, lung, pancrear were found less and the distribute in pancrear was the least one among three tissue. The least one was found in heart.
     2 Research on possible damage of normal murine pancreas via autoimmune disease induced by HSP65-MUC1
     Normal mice immunized with HSP-MUC1, specific lyphocytes were produced in vivo, HSP65 as foreign antigen activated T lymphocytes via MHCI molecular. For mammal HSP60 showed high homology with HSP65 from bacteria, multiple clone T lymphocytes activated by HSP65-MUC1 may cross-reactived with self-reactive antigen, HSP60 and lead to autoimmunization. So, we penetrate with possibility that HSP65-MUC1 cross-reactived with self-reactive antigen and organ specific autoimmune disease induced by HSP65-MUC1.
     2.1 Generation of CTL specific to HSP65 of mice induced by HSP65-MUC1
     Mice immunized with HSP65-MUC1 generated multiple specific CTL and HSP65 specific to CTL, except MUC1 epitope specific to CTL.To prove HSP65 as fusion partner have strong immunity, First,proliferation of HSP65-MUC1 specific to CTL were detected via CTL assay in vivo.The results showed mice inoculated HSP65-MUC1 or HSP65 not only produced HSP65 specific to CTL, but generated MUC1 epitope specific to CTL. To explor increasing immunizing treatments impact on cellular immunity effect, Mice was respectively immunized twice and four times under the same condition, then, CTL proliferation of two situation was compared. Specific CTL proliferation of mice immunized was checked by CTL assay in vivo. CTL proliferate remarkably in mice which were detected on 7 days later from twice immunization, the specific killing rate reached over 90%. CTL proliferate slightly in mice which were detected on 7 days later from four times immunization. the specific killing rate missed reaching 60%.The result presented us that mice produced specific CTL in vivo, but it also proved that immunization for multiple time fail to accelerate immune response.
     2.2. Lymphocytes induced by HSP65-MUC1 cross-reactive with HSP60
     To detect lymphocytes activated by HSP65-MUC1 or HSP65 recongnize HSP65 or HSP60, lymphocyts proliferation test in vitro was carried on. Spleen cell was separated,and stained with CFSE. The spleen lymphocytes incubated with macrophge cells that were dealed by heat shock or unsettled before dealed with colchicines according to ratio of 1:10.10μg/mL of HSP65-MUC1, HSP65 and Vp1 was added into cell supernatants for five days, cells collected was tested for FACS. The result presented: Lymphocytes from mice immunized with HSP65-MUC1 and HSP65, proliferated by stimulus, such as two forms of macrophage, HSP65-MUC1 and HSP65. The results suggested that lymphocytes generated from mice immunized with HSP65-MUC1 and HSP65 were multiclone, that is, lymphocytes activated by HSP65-MUC1 include lymphocytes specific for HSP65 epitope and lymphocytes specific for MUC1 epitope. Besides, Lymphocytes responsed better to macrophage cell dealed with heat shock, by control with two forms of macrophage as simulus, the lyphocytes proliferate in the presence of HSP60 expressed by macrophge, and the reponse was accelerated when HSP60 upregulated.
     2.3 Analysis of possible damage of murine pancrear by HSP65-MUC1
     By using European Molecular Biology Open Software Suite, amino acid sequence of mammal HSP60 was contrast with amino acid sequence of HSP65 from Mycobacterium tuberculosis. The results proved similarity between HSP65 and HSP60 reach over 60%.
     3 Research on possible damage of tumor-bearing murine pancreas via autoimmune disease induced by HSP65-MUC1
     Based on paper, HSP60 as member of heat shock protein family was significantly influenced by temperature and pressure. Accordingly, first, macrophage cells were separated from mice ascites, then, the expression of HSP60 on macrophage cells was altered by changing temperature. when HSP65 and HSP60 polycolonal antibody was used for western-blottin test,Macrophage cells incubating under 43℃and 37℃were collected respectively, it was detected that HSP60 upregulate distinctly ,and HSP65 antibody can cross-recognize HSP60 antigen,
     3.1 Anti-tumor cellular immunity response of tumor-bearing mice induced by HSP65-MUC1
     The principle of tumor vaccine is induced specific CTL generated to kill tumor cells, tumor was fadeaway and patients lives were prolonged. Therefore, we researched that whether mice with tumor immunized with HSP65-MUC1 generated specific CTL, and led to eppitope extention as unspecific killing melanin tumor.
     First, we separated spleen cell of mice with tumor, the spleen cell cocultured with MUC1 positive B16 inactivated by colchicum, B16 inactivated by colchicum acted as negative control, HSP65-MUC1 acted as positive antigen. The cells were cultured for five days, and marked by CD8+ monoclone antibody labeled with PE, FACS check after gathered the cell. Compared with PBS group, proportion of CD8+ lymphocytes increased distinctively when lymphocytes of mice immunized with HSP65-MUC1 were pulsed with MUC1 positive B16. It presented CD8+ lymphocytes specific proliferated. B16 fails to stimulate CD8+ cell proliferate specificly, that is, no epitope extention happened. So, mice induced by HSP65-MUC1 produced CTL specific for MUC1 epitope.
     3.2 Generation of CTL specific to HSP65 of mice induced by HSP65-MUC1
     HSP65 as foreign antigen activeted by T lymphocytes via MHC class I molecular, since HSP60 showed high homology with bacteria HSP65, lthe primed T lymphocytes recongnize self HSP60, and lead to autoimmune reaction, the autoimmune disease occured. HSP60 was self antigen on isletβcell, when the primed lymphocytes of mice immunized by HSP65-MUC1 cross- reactived with HSP60, the cross-reaction are likely to cause autoimmun response, then, insulin dependent diabetes mellitus was triggered. First, blood sugar of mice was detected by blood sugar instrument on the time of experiments earlier and each immunization later. The results showed: blood sugar of mice immunized with HSP65-MUC1 or HSP65, fluctuated below normal value, 6.00mmol/L, no abnormity was found. Second, mice was killed and its pancreas were separated which were sent to pathology and physiology lab for slice, the results of HE staining presented that pancreas of mice were clear and found no lymphocytes invading. The mice generated cross-reactive lymphocytes after immunization, yet, the primed lymphocytes failed to response to self antigen, thus, islet was not damage to change blood sugar concentration for lymphocytes invading islet.
     3.3 Lymphocytes induced by HSP65-MUC1 cross-reactive with HSP60
     To detect lymphocytes activated by HSP65-MUC1 in tumor-bearing mice recongnize HSP65 or HSP60, lymphocyts proliferation test in vitro was carried on. Spleen cell was separated,and stained with CFSE. The spleen lymphocytes incubated with macrophge cells that were dealed by heat shock or unsettled before dealed with colchicines according to ratio of 1:10.10μg/mL of HSP65-MUC1, HSP65 and Vp1 was added into cell supernatants for five days, cells collected was tested for FACS. The result presented: Lymphocytes from mice immunized with HSP65-MUC1 and HSP65, proliferated with stimulus, such as two forms of macrophage, HSP65-MUC1 and HSP65. The results suggested that lymphocytes generated from tumor-bearing mice immunized with HSP65-MUC1 were multiclone, that is, lymphocytes activated by HSP65-MUC1 include lymphocytes specific for HSP65 epitope and lymphocytes specific for MUC1 epitope. Besides, Lymphocytes responsed better to macrophage cell dealed with heat shock, by control with two forms of macrophage as simulus, the lyphocytes proliferate in the presence of HSP60 expressed by macrophge, and the reponse was accelerated when HSP60 upregulated. In a whole, to compare with normal mice immunized, ymphocytes in tumor-bearing mice immunized with HSP65-MUC1 proliferated weakly.
     3.4 Analysis of possible damage of pancrear by HSP65-MUC1
     HSP65-MUC1 as foreign antigen activeted by T lymphocytes via MHC class I molecular, since HSP60 showed high homology with bacteria HSP65, lthe primed T lymphocytes recongnize HSP60, and lead to autoimmune reaction, the autoimmune disease occured. HSP60 was self antigen on islet c?ell, when the primed lymphocytes of mice immunized by HSP65-MUC1 cross- reactived with HSP60, the cross-reaction are likely to cause autoimmun response, then, insulin dependent diabetes mellitus was triggered. First, blood sugar of mice was detected by blood sugar instrument on the time of experiments earlier and each immunization later. The results showed:blood sugar of tumor-bearing mice immunized with HSP65-MUC1, fluctuated below normal value, 6.00mmol/L, no abnormity was found. Second, mice was killed and its pancreas were separated which were sent to pathology and physiology lab for slice, the results of HE staining presented that pancreas of tumor-bearing mice were clear and found no lymphocytes invading. The tumor bearing mice generated cross reactive lymphocytes after immunization, yet, the primed lymphocytes failed to response to self antigen, thus, islet was not damage to change blood sugar concentration for lymphocytes invading islet.
     In conclusion, HSP65-MUC1 fusion protein may induced mice produced effective cellular immunity response, thus, it play an important role in prevention and tumor theropy. We design HSP65-MUC1 labeled with FITC, and made a detailed research on metaboliz.ing and distribute, in addition, HSP65 not only acted as vector of antigen, but also induced mice to produce specific lymphocytes,which could recongnize cross-reactive self HSP60. Though HSP60 was self antigen of isletβcell, yet, whatever was normal mice or tumor-bearing mice,cross-reactive immunity failed to destroy pancreas.HSP65-MUC1 , gene engineering vaccine acquaring first stage clinical warrant, these results presented HSP65-MUC1 could company with chemical drug or single used for treat tumor with MUC1 antigen safely and effectively in clinic.
引文
1. Yannelli JR, Wroblewski JM1 On the road to a tumor cell vaccine:20 years of cellular immunotherapy1 Vaccine, 2004, 23: 9721131.
    2. Disis ML, Cheever MA. Oncogenic proteins as tumor antigens. Curr. Opin. Immunol. 1996;8:637–642.
    3. Wang R-F, Rosenberg SA. Human tumor antigens for cancer vaccine development. Immunol. Rev. 1999;170:85–100. .
    4. Berzofsky JA, Ahlers JD, Belyakov IM. Strategies for designing and optimizing new generation vaccines. Nat. Rev. Immunol. 2001;1:209–219.
    5. Yu Z, Restifo NP. Cancer vaccines: progress reveals new complexities. J. Clin. Invest. 2002;110:289–294
    6. Jager E, Jager D, Knuth A. Antigen specific immunotherapy and cancervaccines [ J] . Int J Cancer, 2003, 106( 6) : 817- 820.
    7. Marten A, Renoth S ,Heinicke T ,et al . Allogeneic dendritic cells fusedwith tumor cells :preclinical results and outcome of a clinical phase ⅠPⅡtrial in patients with metastatic renal cell carcinoma. Hum Gene Ther , 2003 ,14 (5) :483-494.
    8. Vilella R , Benitez D ,Mila J ,et al . Treatment of patients with progressive unresectable metastatic melanoma with a heterologous polyvalent melanoma whole cell vaccine. Int J Cancer ,2003 ,106(4) :626-631.
    9. Parmiani G, et al. Cancer immunotherapy with peptide-based vaccines: what have we achieved? where are we going? J. Natl. Cancer. Inst. 2002;94:805–818. 7.
    10. Henderson RA, Finn OJ. Human tumor antigens are ready to fly. Adv. Immunol. 1996;62:217–256.
    11. Dapeng L,Hui L,Peiyin Z, et al.Heat shock fusion protein induces both specific and nospecific anti-tumor immunity Eur.J. Immunol.2006.36:1324-1336 et al.
    12. Morimoto RL,Tissierse A. Stress proteins in biology and medicine. Cold Spring Harbor: CSHL Press,1990:1-36
    13. Salit RB, Kast WM, Velders MP. Ins and outs of clinical trials with peptide-based vaccines. Front. Biosci. 2002;7:e204–e213.
    14. Berzofsky, J.A., and Berkower, I.J. 2003. Immunogenicity and antigen structure. In Fundamental immunology. W.E. Paul, editor. Lippincott Williams & Wilkins. Philadelphia, Pennsylvania, USA. 631–683.
    15. Germain RN, Margulies DH. The biochemistry and cell biology of antigen processing and presentation. Annu. Rev. Immunol. 1993;11:403–450.
    16. Rosenberg AS, Singer A. Cellular basis of skin allograft rejection: an in vivo model of immune-mediated tissue destruction. Ann. Rev. Immunol. 1992;10:333–358.
    17. Waldmann TA. Immunotherapy: past, present and future. Nat. Med. 2003;9:269–277.
    18. Hung K, et al. The central role of CD4(+) T cells in the antitumor immune response. J. Exp. Med. 1998;188:2357–2368.
    19. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252.
    20. Hurwitz AA, Yu TF-Y, Leach DR, Allison JP. CTLA-4 blockade synergizes with tumor-derived granulocyte-macrophage colony-stimulating factor for treatment of an experimental mammary carcinoma. Proc. Natl. Acad. Sci. U. S. A. 1998;95:10067–10071.
    21. Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+ CD4+ T cells: a common basis between tumor immunity and autoimmunity. J. Immunol. 1999;163:5211–5218.
    22. Sutmuller RPM, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25+ regulatory T cells in antitumor therapy reveals alternative cytotoxic T lymphocyte responses. J. Exp. Med. 2001;194:823–832.
    23. Terabe M, Berzofsky JA. Immunoregulatory T cells in tumor immunity. Curr. Opin. Immunol. 2004;16:157–162.
    24. Terabe M, et al. NKT cell-mediated repression of tumor immunosurveillance by IL-13 and the IL-4R-STAT6 pathway. Nat. Immunol. 2000;1:515–520.
    25. Terabe M, et al. Transforming growth factor-β production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence. J. Exp. Med. 2003;198:1741–1752.
    26. Renkvist N, Castelli C, Robbins PF, Parmiani G. A listing of human tumor antigens recognized by T cells. Cancer Immunol. Immunother. 2001;50:3–15.
    27. Berzofsky, J.A., Helman, L.J., and Carbone, D.P. 2000. Cancer vaccines: cancer antigens: oncogenes and mutations. In Principles and practice ofthe biologic therapy of cancer. 3rd edition. S.A. Rosenberg, editor. Lippincott Williams & Wilkins. Philadelphia, Pennsylvania, USA. 526–541.
    28. Gjertsen MK, et al. Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation. Lancet. 1995;346:1399–1400. 23.
    29. Yanuck M, et al. A mutant p53 tumor suppressor protein is a target for peptide-induced CD8+ cytotoxic T cells. Cancer Res. 1993;53:3257–3261.
    30. Pinilla-Ibarz J, et al. Vaccination of patients with chronic myelogenous leukemia with bcr-abl oncogene breakpoint fusion peptides generates specific immune responses. Blood. 2000;95:1781–1787.
    31. Goletz TJ, Mackall CL, Berzofsky JA, Helman LJ. Molecular alterations in pediatric sarcomas: potential targets for immunotherapy. Sarcoma. 1998;2:77–87.
    32. Mackall, C., Berzofsky, J., and Helman, L.J. 2000. Targeting tumor specific translocations in sarcomas in pediatric patients for immunotherapy. Clin. Orthop. 25–31.
    33. Worley BS, et al. Antigenicity of fusion proteins from sarcoma-associated chromosomal translocations. Cancer Res. 2001;61:6868–6875.
    34. Bos JL. ras oncogenes in human cancer: a review. Cancer Res. 1989;49:4682–4689.
    35. Chiba I, et al. Mutations in the p53 gene are frequent in primary, resected non-small cell lung cancer. Oncogene. 1990;5:1603–1610.
    36. Smith MC, et al. Oncogenic mutations in ras create HLA-A2.1 binding peptides but affect their extracellular processing. Int. Immunol. 1997;9:1085–1093.
    37. Peace DJ, et al. Lysis of Ras oncogene-transformed cells by specific cytotoxic T lymphocytes elicited by primary in vitro immunization with mutated Ras peptide. J. Exp. Med. 1994;179:473–479.
    38. Zaremba S, et al. Identification of an enhancer agonist cytotoxic T lymphocyte peptide from human carcinoembryonic antigen. Cancer Res. 1997;57:4570–4577.
    39. Fong L, et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc. Natl. Acad. Sci. U. S. A. 2001;98:8809–8814.
    40. Theobald M, Biggs J, Dittmer D, Levine AJ, Sherman LA. Targeting p53as a general tumor antigen. Proc. Natl. Acad. Sci. U. S. A. 1995;92:11993–11997.
    41. Correale P, et al. In vitro generation of human cytotoxic T lymphocytes specific for peptides derived from prostate-specific antigen. J. Natl. Cancer Inst. 1997;89:293–300.
    42. Roskrow MA, et al. Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes for the treatment of patients with EBV-positive relapsed Hodgkin’s disease. Blood. 1998;91:2925–2934.
    43. Dranoff G, et al. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc. Natl. Acad. Sci. U.S.A. 1993;90:3539–3543.
    44. Simons JW, et al. Induction of immunity to prostate cancer antigens: results of a clinical trial of vaccination with irradiated autologous prostate tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor using ex vivo gene transfer. Cancer Res. 1999;59:5160–5168.
    45. Soiffer R, et al. Vaccination with irradiated autologous melanoma cells engineered to secrete human granulocyte-macrophage colony-stimulating factor generates potent antitumor immunity in patients with metastatic melanoma. Proc. Natl. Acad. Sci. U. S. A. 1998;95:13141–13146.
    46. Jaffee EM, et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J. Clin. Oncol. 2001;19:145–156.
    47. Salgia R, et al. Vaccination with irradiated autologous tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor augments antitumor immunity in some patients with metastatic non-small-cell lung carcinoma. J. Clin. Oncol. 2003;21:624–630.
    48. Maio M, et al. Vaccination of stage IV patients with allogeneic IL-4- or IL-2-gene-transduced melanoma cells generates functional antibodies against vaccinating and autologous melanoma cells. Cancer Immunol. Immunother. 2002;51:9–14.
    49. Antonia SJ, et al. Phase I trial of a B7-1 (CD80) gene modified autologous tumor cell vaccine in combination with systemic interleukin-2 in patients with metastatic renal cell carcinoma. J. Urol. 2002;167:1995–2000.
    50. Unfer RC, Hellrung D, Link CJ Jr. Immunity to the alpha(1, 3)galactosyl epitope provides protection in mice challenged with colon cancer cells expressing alpha(1, 3)galactosyl-transferase: a novel suicide gene for cancer gene therapy. Cancer Res. 2003;63:987–993.
    51. Stern LJ, Wiley DC. Antigenic peptide binding by class I and class II histocompatibility proteins. Structure. 1994;2:245–251.
    52. Rammensee H-G, Friede T, Stevanovíc S. MHC ligands and peptide motifs: first listing. Immunogenetics. 1995;41:178–228.
    53. Traversari C, et al. A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E. J. Exp. Med. 1992;176:1453–1457.
    54. Berzofsky JA. Epitope selection and design of synthetic vaccines: molecular approaches to enhancing immunogenicity and crossreactivity of engineered vaccines. Ann. N. Y. Acad. Sci. 1993;690:256–264.
    55. Rosenberg SA, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat. Med. 1998;4:321–327.
    56. Rivoltini L, et al. A superagonist variant of peptide MART1/Melan A27-35 elicits anti-melanoma CD8+ T cells with enhanced functional characteristics: implication for more effective immunotherapy. Cancer Res. 1999;59:301–306.
    57. Brinckerhoff LH, et al. Terminal modifications inhibit proteolytic degradation of an immunogenic MART-1(27–35) peptide: implications for peptide vaccines. Int. J. Cancer. 1999;83:326–334.
    58. Ahlers JD, et al. A push-pull approach to maximize vaccine efficacy: abrogating suppression with an IL-13 inhibitor while augmenting help with GM-CSF and CD40L. Proc. Natl. Acad. Sci. U. S. A. 2002;99:13020–13025.
    59. Klinman DM, Yi AK, Beaucage SL, Conover J, Krieg AM. CpG motifs present in bacterial DNA rapidly induce lymphocytes to secrete interleukin 6, interleukin 12, and interferon gamma. Proc. Natl. Acad. Sci. U. S. A. 1996;93:2879–2883.
    60. Alexander J, et al. Development of high potency universal DR-restricted helper epitopes by modification of high affinity DR-blocking peptides. Immunity. 1994;1:751–761.
    61. Shirai M, et al. Use of intrinsic and extrinsic helper epitopes for in vivo induction of anti-hepatitis C virus cytotoxic T lymphocytes (CTL) with CTL epitope peptide vaccines. J. Infect. Dis. 1996;173:24–31.
    62. Melief CJ, Van Der Burg SH, Toes RE, Ossendorp F, Offringa R. Effective therapeutic anticancer vaccines based on precision guiding of cytolytic T lymphocytes. Immunol. Rev. 2002;188:177–182.
    63. Ahlers JD, Belyakov IM, Thomas EK, Berzofsky JA. High affinity T-helper epitope induces complementary helper and APC polarization, increased CTL and protection against viral infection. J. Clin. Invest. 2001;108:1677–1685. doi:10.1172/JCI200113463.
    64. Smith JW 2nd, et al. Adjuvant immunization of HLA-A2-positive melanoma patients with a modified gp100 peptide induces peptide-specific CD8+ T-cell responses. J. Clin. Oncol. 2003;21:1562–1573.
    65. Lee P, et al. Effects of interleukin-12 on the immune response to a multipeptide vaccine for resected metastatic melanoma. J. Clin. Oncol. 2001;19:3836–3847.
    66. Weber J, et al. Granulocyte-macrophage-colony-stimulating factor added to a multipeptide vaccine for resected Stage II melanoma. Cancer. 2003;97:186–200.
    67. Schaed SG, et al. T-cell responses against tyrosinase 368-376( 370D) peptide in HLA*A0201+ melanoma patients: randomized trial comparing incomplete Freund’s adjuvant, granulocyte macrophage colony-stimulating factor, and QS-21 as immunological adjuvants. Clin. Cancer Res. 2002;8:967–972.
    68. Phan GQ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen blockade in patients with metastatic melanoma. Proc. Natl. Acad. Sci. U. S. A . 2003;100:8372–8377.
    69. Marshall JL, et al. Phase I study in advanced cancer patients of diversified prime-and-boost vaccination protocol using recombinant vaccinia virus and recombinant nonreplicating avipox virus to elicit anti-carcinoembryonic antigen immune responses. J. Clin. Oncol. 2000;18:3964–3973.
    70. Zhu MZ, Marshall J, Cole D, Schlom J, Tsang KY. Specific cytotoxic T-cell responses to human CEA from patients immunized with recombinant avipox-CEA vaccine. Clin. Cancer Res. 2000;6:24–33.
    71. Belyakov IM, Moss B, Strober W, Berzofsky JA. Mucosal vaccination overcomes the barrier to recombinant vaccinia immunization caused by preexisting poxvirus immunity. Proc. Natl. Acad. Sci. U. S. A. 1999;96:4512–4517.
    72. Hodge JW, et al. A triad of costimulatory molecules synergize to amplify T-cell activation. Cancer Res. 1999;59:5800–5807.
    73. Oh S, et al. Selective induction of high avidity CTL by altering the balance of signals from antigen presenting cells. J. Immunol. 2003;170:2523–2530.
    74. Tang D, DeVit M, Johnston SA. Genetic immunization is a simple method for eliciting an immune response. Nature. 1992;356:152–154.
    75. Ulmer JB, et al. Heterologous protection against influenza by injection of DNA encoding a viral protein. Science. 1993;259:1745–1749.
    76. Timmerman JM, et al. Immunogenicity of a plasmid DNA vaccine encoding chimeric idiotype in patients with B-cell lymphoma. Cancer Res. 2002;62:5845–5852.
    77. Conry RM, et al. Safety and immunogenicity of a DNA vaccine encoding carcinoembryonic antigen and hepatitis B surface antigen in colorectal carcinoma patients. Clin. Cancer Res. 2002;8:2782–2787.
    78. Fong L, Engleman EG. Dendritic cells in cancer immunotherapy. Annu. Rev. Immunol. 2000;18:245–273.
    79. Schuler G, Schuler-Thurner B, Steinman RM. The use of dendritic cells in cancer immunotherapy. Curr. Opin. Immunol. 2003;15:138–147.
    80. Gabrilovich DI, Nadaf S, Corak J, Berzofsky JA, Carbone DP. Dendritic cells in anti-tumor immune responses. II. Dendritic cells grown from bone marrow precursors, but not mature DC from tumor-bearing mice are effective antigen carriers in the therapy of established tumors. Cell Immunol. 1996;170:111–119.
    81. Gabrilovich DI, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells [erratum 1996, 2:1267] Nat. Med. 1996;2:1096–1103.
    82. Mayordomo JI, et al. Bone marrow-derived dendritic cells pulsed with synthetic tumour peptides elicit protective and therapeutic antitumour immunity. Nat. Med. 1995;1:1297–1302.
    83. Porgador A, Gilboa E. Bone marrow-generated dendritic cells pulsed with a class I-restricted peptide are potent inducers of cytotoxic T lymphocytes. J. Exp. Med. 1995;182:255–260.
    84. Nair SK, Snyder D, Rouse BT, Gilboa E. Regression of tumors in mice vaccinated with professional antigen-presenting cells pulsed with tumorextracts. Int. J. Cancer. 1997;70:706–715.
    85. Paglia P, Chiodoni C, Rodolfo M, Colombo MP. Murine dendritic cells loaded in vitro with soluble protein prime cytotoxic T lymphocytes against tumor antigen in vivo. J. Exp. Med. 1996;183:317–322.
    86. Zitvogel L, et al. Therapy of murine tumors with tumor peptide-pulsed dendritic cells: dependence on T cells, B7 costimulation, and T helper cell 1-associated cytokines. J. Exp. Med. 1996;183:87–97.
    87. Ashley DM, et al. Bone marrow-generated dendritic cells pulsed with tumor extracts or tumor RNA induce antitumor immunity against central nervous system tumors. J. Exp. Med. 1997;186:1177–1182.
    88. Mayordomo JI, et al. Therapy of murine tumors with p53 wild-type and mutant sequence peptide-based vaccines. J. Exp. Med. 1996;183:1357–1365.
    89. Siders WM, Vergilis KL, Johnson C, Shields J, Kaplan JM. Induction of specific antitumor immunity in the mouse with the electrofusion product of tumor cells and dendritic cells. Mol. Ther. 2003;7:498–505.
    90. Chen HW, et al. Inducing long-term survival with lasting anti-tumor immunity in treating B cell lymphoma by a combined dendritic cell-based and hydrodynamic plasmid-encoding IL-12 gene therapy. Int. Immunol. 2003;15:427–435.
    91. Akiyama Y, et al. Enhancement of antitumor immunity against B16 melanoma tumor using genetically modified dendritic cells to produce cytokines. Gene Ther. 2000;7:2113–2121.
    92. Wan Y, Bramson J, Carter R, Graham F, Gauldie J. Dendritic cells transduced with an adenoviral vector encoding a model tumor-associated antigen for tumor vaccination. Hum. Gene Ther. 1997;8:1355–1363.
    93. Esslinger C, Romero P, MacDonald HR. Efficient transduction of dendritic cells and induction of a T-cell response by third-generation lentivectors. Hum. Gene Ther. 2002;13:1091–1100.
    94. Bonifaz L, et al. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J. Exp. Med. 2002;196:1627–1638.
    95. Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau J. GM-CSF and TNF-alpha cooperate in the generation of dendritic Langerhans cells. Nature. 1992;360:258–261.
    96. Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen bycultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α J. Exp. Med. 1994;179:1109–1118.
    97. Banchereau J, et al. Immune and clinical responses in patients with metastatic melanoma to CD34(+) progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61:6451–6458.
    98. Mackensen A, et al. Phase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in vitro from CD34(+) hematopoietic progenitor cells. Int. J. Cancer. 2000;86:385–392.
    99. Rosenzwajg M, Jourquin F, Tailleux L, Gluckman JC. CD40 ligation and phagocytosis differently affect the differentiation of monocytes into dendritic cells. J. Leukoc. Biol. 2002;72:1180–1189.
    100.Labeur MS, et al. Generation of tumor immunity by bone marrow-derived dendritic cells correlates with dendritic cell maturation stage. J. Immunol. 1999;162:168–175.
    101.Dhodapkar MV, Steinman RM, Krasovsky J, Munz C, Bhardwaj N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J. Exp. Med. 2001;193:233–238.
    102.Dhodapkar MV, Steinman RM. Antigen-bearing immature dendritic cells induce peptide-specific CD8(+) regulatory T cells in vivo in humans. Blood. 2002;100:174–177.
    103.Jonuleit H, Schmitt E, Schuler G, Knop J, Enk AH. Induction of interleukin 10-producing, nonproliferating CD4(+) T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. J. Exp. Med. 2000;192:1213–1222.
    104.Steinmann RM, Dhodapkar K. Active immunization against cancer with dendritic cells. Int. J. Cancer. 2001;94:459–573.
    105.Nestle FO. Dendritic cell vaccination for cancer therapy. Oncogene. 2000;19:6673–6679.
    106.Hsu FJ, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat. Med. 1996;2:52–58.
    107.Timmerman JM, et al. Idiotype-pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune responses in 35 patients. Blood. 2002;99:1517–1526.
    108.Reichardt VL, et al. Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma—a feasibility study. Blood. 1999;93:2411–2419.
    109.Liso A, et al. Idiotype vaccination using dendritic cells after autologous peripheral blood progenitor cell transplantation for multiple myeloma. Biol. Blood Marrow Transplant. 2000;6:621–627.
    110.Valone FH, et al. Dendritic cell-based treatment of cancer: closing in on a cellular therapy. Cancer J. 2001;7(Suppl. 2):S53–S61.
    111.Fong L, et al. Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy. J. Immunol. 2001;167:7150–7156.
    112.Nestle FO, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat. Med. 1998;4:328–332.
    113.Thurner B, et al. Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J. Exp. Med. 1999;190:1669–1678.
    114.Stift A, et al. Dendritic cell-based vaccination in solid cancer. J. Clin. Oncol. 2003;21:135–142.
    115.Su Z, et al. Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells. Cancer Res. 2003;63:2127–2133.
    116.Serody JS, Collins EJ, Tisch RM, Kuhns JJ, Frelinger JA. T cell activity after dendritic cell vaccination is dependent on both the type of antigen and the mode of delivery. J. Immunol. 2000;164:4961–4967.
    117.Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat. Immunol. 2002;3:611–618.
    118.Lee KM, et al. Molecular basis of T cell inactivation by CTLA-4. Science. 1998;282:2263–2266.
    119.Thornton AM, Shevach EM. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J. Immunol. 2000;164:183–190.
    120.Kronenberg M, Gapin L. The unconventional lifestyle of NKT cells. Nat. Rev. Immunol. 2002;2:557–568.
    121.Wilson SB, Delovitch TL. Janus-like role of regulatory iNKT cells in autoimmune disease and tumour immunity. Nat. Rev. Immunol. 2003;3:211–222.
    122.Ostrand-Rosenberg S, et al. Resistance to metastatic disease in Stat6-deficient mice requires hematopoietic and non-hematopoietic cells and is IFNγ-dependent. J. Immunol. 2002;169:5796–5804.
    123.Snyder JT, Alexander-Miller MA, Berzofsky JA, Belyakov IM. Molecular mechanisms and biological significance of CTL avidity. Curr. HIV Res. 2003;1:287–294.
    124.Alexander-Miller MA, Leggatt GR, Berzofsky JA. Selective expansion of high or low avidity cytotoxic T lymphocytes and efficacy for adoptive immunotherapy. Proc. Natl. Acad. Sci. U. S. A. 1996;93:4102–4107.
    125.Yee C, Savage PA, Lee PP, Davis MM, Greenberg PD. Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers. J. Immunol. 1999;162:2227–2234.
    126.Zeh HJ III, Perry-Lalley D, Dudley ME, Rosenberg SA, Yang JC. High avidity CTLs for two self-antigens demonstrate superior in vitro and in vivo antitumor efficacy. J. Immunol. 1999;162:989–994.
    127.Oh S, Berzofsky JA, Burke DS, Waldmann TA, Perera LP. Coadministration of HIV vaccine vectors with vaccinia viruses expressing IL-15 but not IL-2 induces long-lasting cellular immunity. Proc. Natl. Acad. Sci. U. S. A. 2003;100:3392–3397.
    128.Janssen EM, et al. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature. 2003;421:852–856.
    129.Sun JC, Bevan MJ. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science. 2003;300:339–342.
    130.Shedlock DJ, Shen H. Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science. 2003;300:337–339.
    131.Gett AV, Sallusto F, Lanzavecchia A, Geginat J. T cell fitness determined by signal strength. Nat. Immunol. 2003;4:355–360.
    132.Kawakami Y, Eliyahu S , Jennings C , et al . Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor infiltrating T lymphocytes associated with in vivo tumor regression [J ] . J Immunol , 1995 ,154(8) :3 961 - 3 968.
    133.Mitchell M S , Kan2Mitchell J , Kempf R A , et al . Active specific immunotherapy for melanoma : Phase Ⅰtrial of allogeneic lysates and a novel adjuvant [J ]. Cancer Res, 1988 ,48(20) : 5 883 - 5 893.
    134.Reddish M A , MacLean G D , Koganty R R , et al . Anti MUC1 class Ⅰrestricted CTLs in metastatic breast cancer patients immunized with a synthetic MUC1 peptide[J ] . Int J Cancer , 1998 , 76(6) : 817 - 823.
    135.Marchand M, van Baren N , Boon T, et al . Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE23 and presented by HLA-A1 [J ] . Int J Cancer ,1999 , 80(2) : 219 - 230.
    136.Chang E Y, Chen C H , Ji H , et al . Antigen specific cancer immunotherapy using a GM-CSF secreting allogeneic tumor cellbased vaccine[J ] . Int J Cancer , 2000 , 86(5) :725 - 730.
    137.Yang S , Darrow TL , Seigler H F. Generation of primary tumor specificcytotoxic T lymphocytes from autologous and human lymphocyte antigen class Ⅰ2matched allogeneic peripheral blood lymphocytes by B7 gene2modified melanoma cells [J ] . Cancer Res ,1997 , 57(8) : 1 561 - 1 568
    138.Dunnion DJ, Cywinski AL, Tucker V C , et al . Human antigen presenting cell/ tumour cell hybrids stimulate strong allogeneic responses and present tumour2associated antigens to cytotoxic T cells in vitro[J ]. Immunology, 1999, 98(4) : 541 - 550.
    139.Sastry S , Linderoth N. Molecular mechanisms of peptide loading by the tumor rejection/ heat shock chaperone gp96 (grp94) [J ] . J Biol Chem , 1999 , 274 (17) : 12023-12035.
    140.Arnold2Schild D , Hanau D , Spehner D , et al . Cutting edge : Receptor mediated endocytosis of heat shock proteins by professional antigen presenting cells[J ] . J Immunol , 1999 , 162 : 375723760.
    141.Singh-Jasuja H , Toes REM , Spee P , et al . Cross-presentation of glycoprotein 96-associated antigens on major histocompatibility complex class I molecules requires receptor mediated endocytosis J Exp Med , 2000 , 191 (11) : 1965-1974.
    142.Janetzki S, Blachere NE, Srivasava PK. Generation of tumor specific cytotoxic T lymphocytes and memory T cells by immunization with tumor-derived heat shock protein gp96 [ J ] . J Immunother 1998 , 21 (4) : 269-276.
    143.Chandawarkar RY, Wagh MS , Srivastava PK. The dual nature of specific immunological activity of tumor2derived gp96 preparations [J ] . J Exp Med , 1999 , 189 (9) : 1437-1442.
    144.Heikema A , Agsteribbe E , Wilschut J , et al . Generation of heat shock protein2based vaccines by intracellular loading of gp96 with antigenic peptides[J ]. Immunol Lett , 1997 , 57 : 69274.
    145.Berd D, Maguire H C, Schuchier L M, et al. Autologous hapten modified melanoma vaccine as postsurgical adjuvant treatment after resection of nodal metastatases [J ].J Clin Oncol ,1997 ,15(6) :2 359 - 2 370.
    146.Tsang K Y, Zaremba S, Nieroda C A, et al. Generation of human cytotoxic T cells specific for human carcinoembryonic antigen eiptopes from patients immunized with recombinant CEA vaccine [J ]. J Natl Cancer Inst, 1995, 87(13): 982 -990.
    147.Thurner B , Haendle I , Roder C , et al . Vaccination with mage-3A1 peptide2pulsed mature , monocyte2derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage Ⅳ melanoma [ J ] . J Exp Med ,1999 , 190(11) :1 669 - 1 678.
    148.Ayyoub M, Mazarguil H , Monsarrat B , et al . A structure2based approach to designing non2natural peptides that can activate anti melanoma cytotoxic T cells[J ] . J Biol Chem , 1999 , 274(15) :10 227 - 10 234.
    149.Reynolds S R , Oratz R , Shapiro R L , et al . Stimulation of CD8 + T cell responses to MAGE23 and Melan A/MART21 byimmunization to a polyvalent melanoma vaccine [J ] . Int J Cancer , 1997 , 72(6) : 972 - 976.
    150.Parkhurst M R , Salgaller M L , Southwood S , et al . Improved induction of melanoma2reactive CTL with peptides from themelanoma antigen gp100 modified at HLA2A 3 02012binding residues[J ] . J Immunol , 1996 , 157(6) :2 539 - 2 548.
    151.Rosenberg S A , Yang J C , Schwartzentruber D J , et al . Impact of cytokine administration on the generation of antitumor reactivi y in patients with metastatic melanoma receiving a peptide vaccine[J ] . J Immunol , 1999 , 163(3) : 1 690 - 1 695.
    152.Bueler H , Mulligan R C. Induction of antigen2specific tumor immunity by genetic and cellular vaccines against MAGE: enhanced tumor protection by coexpression of granulocytemacrophage colony2stimulating factor and B7. 1 [J ] . Mol Med ,1996 , 2(5) :545 - 555.
    153.Tuting T, DeLeo A B , Lotze M T, et al . Genetically modifiedbone marrow derived dendritic cells expressing tumor associatedviral or“self”antigens induce antitumor immunity in vivo [J ] .Eur J Immunol ,1997 ,27(10) :2 702 - 2 707.
    154.Martin B K, Frelinger J G, Ting J P , et al . Combination genetherapy with CD86 and the MHC class Ⅱtransactivator in the control of lung tumor growth[J ] . J Immunol , 1999 , 162(11) :6 663 - 6 670.
    155.Tuting T, Wilson C C , Martin D M, et al . Autologous human monocyte2derived dendritic cells genetically modified to express melanoma antigens elicit primary cytotoxic T cell responses in vitro :enhancement by cotransfection of genes encoding the Th12 biasing cytokines IL212 and IFN2α[J ] . J Immunol , 1998 , 160 (3) :1 139 - 1 147.
    156.Toes R E , van der Voort E I H , Schoenberger S P , et al . Enhancement of tumor outgrowth through CTL tolerization after peptide vaccination is avoided by peptide presentation on dendriticcells[J ] . J Immunol , 1998 , 160(9) :4 449 - 4 456.
    157.张立新,李春海。MUC1 粘蛋白的免疫学作用及其在肿瘤生物学治疗中的作用。中国肿瘤生物治疗杂志,2000, 7(3):165-170
    158.阎洪彦,王立顺。MUC1 粘蛋白-肿瘤生物治疗的新靶点。国外医学免疫学分册,2000, 23(2):113-115。
    159.王洪琦,张正,赵燕萍。热休克蛋白在肿瘤应用基础领域中的研究进展。2001,23(2):93-95。
    160.赵 君,杨守京,刘彦仿。实验性汉坦病毒感染乳鼠诱导脑神经细胞表达热休克蛋白 70。细胞与分子免疫学杂志,2000,16(3):232-233
    161.陈慰峰主编.医学免疫学第三版。
    162.162Johnen H, Kulbe H, Pecher G. Long-term tumor growth suppression in mice immunized with naked DNA of the human tumor antigen mucin (MUC1). Cancer Immunol Immunother, 2001 Sep; 50(7): 356-60
    163.163 Karanikas V; Lodding J; Maino VC; McKenzie IF. Flow cytometric measurement of intra cellula r cytokines detects immune responses in MUC1 immunotherapy. Clin Cancer Res, 2000 Mar; 6(3): 829-37
    164.164 Karanikas V, et al. Antibody and T cell responses of patients with adenocarcinoma immunized with mannan-MUC1 fusion protein. J Clin Invest 1997 Dec 1; 100(11): 2783-92
    165.165 Kodama H, Suzuki M, Katayose Y, Shinoda M, Sakurai N, Takemura S, Yoshida H, Jang KT, Chae SW, Sohn JH, Park HR, Shin HS.Coexpression of MUC1 with p53 or MUC2 correlates with Lymph Node Metastasis in Colorectal Carcinomas. J Korean Med Sci, 2002 Feb; 17(1): 29-33
    166.166 Kilger E; Pecher G; Schwenk A; Hammerschmidt W Expression of mucin (MUC-1) from a mini-Epstein-Barr virus in immortalized B-cells to generate tumor antigen specific cytotoxic T cells. J Gene Med, 1999 Mar-Apr; 1(2): 84-92
    167.167 Koido S, Tanaka Y, Chen D, Kufe D, Gong J. The kinetics of in vivo priming of CD4 and CD8 T cells by dendritic/tumor fusion cells in MUC1-transgenic mice. J Immunol,2002 Mar 1;168(5):2111-7
    168.Wen YJ, Barlogie B, Yi Q. Idiotype-specific cytotoxic T lymphocytes in multiple myeloma: evidence for their capacity to lyse autologous primary tumor cells. Blood. 2001;97:1750–1755.
    169.Dutoit V, et al. Heterogeneous T-cell response to MAGE-A10 (254–262): high avidity-specific cytolytic T lymphocytes show superior antitumor activity. Cancer Res. 2001;61:5850–5856.
    170.Heiser A, et al. Human dendritic cells transfected with renal tumor RNA stimulate polyclonal T-cell responses against antigens expressed by primary and metastatic tumors. Cancer Res. 2001;61:3388–3393.
    171.Valmori D, et al. Vaccination with a Melan-A peptide selects an oligoclonal T cell population with increased functional avidity and tumor reactivity. J. Immunol. 2002;168:4231–4240.
    172.Pardoll DM, Topalian SL. The role of CD4+ T cell responses in anti-tumor immunity. Curr. Opin. Immunol. 1998;10:588–594.
    173.Yanagawa H, et al. Presence and potent immunosuppressive role of interleukin-10 in malignant pleural effusion due to lung cancer. Cancer Lett. 1999;136:27–32.
    174.Kudryashov, V., Glunz, P. W., Williams, L. J., Hintermann, S., Danishefsky, S. J. & Lloyd, K. O. (2001. ) Proc. Natl. Acad. Sci. USA 98:, 3264-3269.
    175.陈劲松,夏双印。热休克蛋白的分子遗传学研究进展。国外医学遗传学分册,2001;24(3):128-132。
    176.范云霞。热休克蛋白与免疫应答。国外医学免疫学分册,2001;24(2):2-64。
    177.李晓鲁,彭毅志。热休克蛋白与抗感染免疫。免疫学杂志,2001;17(3):139-142。
    178.孟颂东,高福,田波。热休克蛋白-多肽复合物在肿瘤和传染性疾病免疫中的作用。生物工程学报, 2000;16(4):425-428。
    179.Gaur R ,Gupta PK,Goyal A ,et al . Delivery of nucleic acid into mam2malian cells by anthrax toxin. BBRC ,2002 ,297 :112121127.
    180.Srivastava PK.Udono H,Blachere NE,et al.Heat shock proteins transfer peptides during antigen processing and CTL priming. [J].J immunogenetics,1994:9(2)93-98
    181.Chandawarkar RY, Wagh MS , Srivastava PK. The dual nature of specific immunological activity of tumor2derived gp96 preparations [J ] . J Exp Med , 1999 , 189 (9) : 1437-1442.
    182.Zhang, S. L., Graeber, L. A., & Gulley JL, Arlen PM, Bastian A, et al.Combining a recombinant cancer vaccine with standard definitive radiotherapy in patients with localized prostate cancer. Clin Cancer Res. 2005;11:3353–62.
    183.Arlen PM, Gulley JL, Todd N, et al. Antiandrogen, vaccine and combination therapy in patients with nonmetastatic hormone refractory prostate cancer. [J ]. J Urol. 2005;4:539–46.
    184.Chan OT, Yang LX. The immunological effects of taxanes. Cancer Immunol Immunother. 2000;49:181–5.
    185.Mason K, Staab A, Hunter N, et al. Enhancement of tumor radioresponse by docetaxel: involvement of immune system. Int J Oncol. 2001;18:599–606.
    186.Bubley GJ, Carducci M, Dahut W, et al. Eligibility and response guidelines for phase II clinical trials in androgen-independent prostate cancer: recommendations from the Prostate-Specific Antigen Working Group. [J ].J Clin Oncol. 1999;17:3461–7.
    187.Arlen P, Tsang KY, Marshall JL, et al. The use of a rapid ELISPOT assay to analyze peptide- specific immune responses in carcinoma patients to peptide vs. recombinant poxvirus vaccines. Cancer Immunol Immunother. 2000;49:517–29.
    188.Hodge JW, Grosenbach DW, Aarts WM, Poole DJ, Schlom J. Vaccine therapy of established tumors in the absence of autoimmunity. Clin Cancer Res. 2003;9:1837–49.
    189.Linderoth NA, Popowicz A , Sastry S. Identification of the peptide binging sites in the heat shock chaperone/ tumor rejection anti gen gp96 (grp94) [J ]. J Biol Chem , 2000 , 275 (8) : 547225477.
    190.Kass E, Panicali DL, Mazzara G, Schlom J, Greiner JW. Granulocyte/macrophage-colony stimulating factor produced by recombinant avian poxviruses enriches the regional lymph nodes with antigen-presenting cells and acts as an immunoadjuvant. Cancer Res. 2001;61:206–14.
    191.Wang XY, Kaneko Y, Repasky E , et al . Heat shock proteins and cancer immunotherapy [J ] . Immunol Invest , 2000 , 29 (2) :1312137.
    192.Singh-Jasuja H , Toes REM , Spee P , et al . Crosspresentation of glycoprotein 96 associated antigens on major histocompatibility complex class I molecules requires receptor mediated endocytosis[J ]. J Exp Med , 2000 , 191 (11) : 196521974.
    193.Arnold Schild D, Hanau D, Spehner D, et al. Cutting edge: Receptormediated endocytosis of heat shock proteins by professional antigen2presenting cells. [J ].J Immunol , 1999 , 162 : 3757-3760.
    194.Phan GQ, Yang JC, Sherry RM, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A. 2003;100:8372–8377.
    195.Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med. 1998;4:321–327.
    196.Takahashi T, Tagami T, Yamazaki S, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000;192:303–310.
    197.Taylor PA, Noelle RJ, Blazar BR. CD4(+) CD25(+) immune regulatory cells are required for induction of tolerance to alloantigen via co-stimulatory blockade. J Exp Med. 2001;193:1311–1318.
    198.Kristiansen OP, Larsen ZM, Pociot F. CTLA-4 in autoimmune diseases: A general susceptibility gene to autoimmunity? Genes Immun. 2000;1:170–184.
    199.YiPing Y,Ching-Tai H,XiaoPeng H,Drew M Paradoll, persistent Toll-like receptor signals required for reversal of T cell-mediated, nature immunology,2004,5(5).
    200.W.E. 保 罗 , 编 著 , 吴 玉 章 译 . 基 础 免 疫 学 ( Foungdamental Immunology.Fourth Edition)2003,7