重组MUC1多肽及MUC1-MBP融合蛋白的纯化及抗肿瘤作用比较研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
MUC1是粘蛋白家族中的一员,主要分布在腺上皮细胞及其来源的肿瘤细胞表面,因其在正常组织和肿瘤组织的差异使其成为肿瘤疫苗研究的有效靶点。以MUC1为基础的疫苗已经快速进入人类的临床试验,并且表现出良好的疗效。本研究组成功制备出高纯度的MUC1-MBP融合蛋白,初步研究表明,MUC1-MBP蛋白联合BCG免疫在小鼠体内能预防并抑制肺癌转移及实体瘤的生长,还能明显抑制小鼠皮下人乳腺癌的生长,但对疫苗的各个组分MUC1、MBP、BCG的具体作用还没有进行深入的探讨。本课题的研究目的是在研究组前期研究工作的基础之上进一步比较重组MUC1多肽及MUC1-MBP融合蛋白的抗肿瘤作用,分析MUC1多肽、MBP蛋白、BCG在免疫中所起的作用。
     本研究通过IPTG诱导MUC1-MBP、MBP及MUC1-GST蛋白表达,采用Amylose resin亲和层析制备高纯度MUC1-MBP融合蛋白及MBP蛋白。用Glutathione–Sepherose4B纯化MUC1-GST,采用凝血酶裂解MUC1-GST,获得重组MUC1多肽。用纯化的蛋白在动物肿瘤模型上进行了研究,研究结果表明,MUC1-MBP联合BCG具有显著的抗肿瘤作用,而MUC1多肽联合BCG没有明显的抗肿瘤作用,MUC1-MBP联合BCG作为抗肿瘤疫苗其中的三个组分MUC1、MBP、BCG缺一不可。本研究结果进一步提示了MUC1-MBP联合BCG作为抗肿瘤疫苗的开发价值。
MUC1is a tumor-associated antigen, glycoprotein.of high molecular weight.MUC1-based vaccine has been rapidly applied to human clinical trials, anddemonstrates a good efficacy. Studies have shown that MUC1peptides can notinduce the host to produce antibodies, CTL or DTH; MUC1peptides gearing KLHand GST can induce the host to produce antibodies and DTH, but no CTL; however,MUC1connecting oxidized mannan can induce strong CTL responses andanti-tumor effects. E. coli maltose binding protein (MBP) can regulate the activity ofDCs. Our research team find that MBP can promote cellular immune responses.Therefore, our research group used pMAL-c2, a prokaryotic vector, as a carrier toinsert a repeating sequence of MUC1gene into the downstream of MBPgene—malE gene and established a vector that could successfully expressMUC1-MBP recombinant gene and obtained engineered bacteria which couldexpress MUC1-MBP fusion protein steadily and efficiently. Thus we prepared theMUC1-MBP fusion protein of higher purity and carried out a preliminary study ontheir anti-tumor effects. The purpose of this subject, is to conduct a comparativestudy on the anti-tumor effects of recombinant MUC1peptides and MUC1-MBPfusion protein on the basis of our previous research work.
     Methods and Results of the Research:
     1. Preparation for the protein We cultured a large number of pMAL-MUC1/DH5α, pMAL-c2/DH5α and pGEX-MUC1/BL21and used IPTG for theexpression of MUC1-MBP, MBP and MUC1-GST protein. Bacterial specimenswere lysis by sonication and the supernatant were obtained. MUC1-MBP fusionprotein and MBP protein of high purity were prepared by affinity chromatography of Amylose resin. We purified MUC1-GST through Glutathione-Sepherose4B. Weused thrombin to cleavage the MUC1-GST protein in order to get the recombinantMUC1peptides. The purified protein were identified by Western blotting.
     2. The establishment of the subcutaneous B16-MUC1melanoma model In theexistence of G418we cultured B16melanoma cells stably transfected with humanfull-length MUC1. The expression of MUC1protein was identified by flowcytometry.2*106cells were injeced into each C57BL/6mouse subcutaneouslyand the subcutaneous B16-MUC1melanoma model was successfully established.
     3. Comparative analysis on the protein’s anti-tumor effects C57BL/6micewere immunized with recombinant MUC1peptides, MUC1-MBP protein ofdifferent doses and MBP protein and the above-mentioned protein joint BCGsubcutaneously every seven days and immune totally three times. The weightchanges of the mice were monitored and analyzed by statistical software. After5-7days of the third immunization,2*106mouse melanoma cells stably transfectedwith the MUC1gene (B16-MUC1) were subcutaneously injected into each mouse.We monitored tumor growth carefully. The mice were killed9-13days after theinjection of tumor cells. We weighed the mice, stripped subcutaneous melanomaxenograft tumor completely and weighed the tumor. Subcutaneous melanomaxenograft tumor inhibitory rates of each group were calculated. Separated andweighed the spleen. Combining with the body weights of the mice, spleen indexes ofall groups were culculated. The results showed that:
     1) MUC1peptides immunized alone slightly inhibited the growth of subcutaneoustransplanted melanoma tumor; BCG immunized alone and MUC1peptidesimmunized with BCG had no tumor inhibition effect.
     2) It was not obvious of tumor inhibition when MBP protein was immunizedalone; on the contrary, immunizingMBP protein combined with BCG distinctlyprohibited the growth of subcutaneous melanoma with the rate of55.5%.
     3) Immunizing MUC1-MBP protein alone slightly reduced the growth ofsubcutaneous transplanted melanoma and the inhibitory rate was higher when the doses of MUC1-MBP increased. Inhibitory rates of subcutaneous transplantedmelanoma clearly increased when mice were immunized with MUC1-MBP proteincombined with BCG. Importantly, the inhibitory rate increased to a maximum of98%with the administered dose increased to50μg.
     4) With regard to the mice spleen indexes, the MUC1peptides group,MUC1-MBP fusion protein group and MBP group had no significant differencescompared with the control group (P>0.05). Furthermore, the MUC1peptidescombined with BCG group, MUC1-MBP fusion protein combined with BCG groupand MBP combined with BCG group had distinct differences compared with thecontrol group (P<0.05). So spleen index can be used as an indicator of immuneactivity of the MUC1-MBP vaccine or BCG.
     5) The body weights of the mice immunized with recombinant MUC1peptides,MUC1-MBP fusion protein and MBP had no significant differences compared withthe control group (P>0.05). When joint BCG, the body weights of mice declinedslightly but didn’t significantly compared with the control group (P>0.05). Theresults indicate that BCG prompts a slight toxic effect in mice
     In conclusion, MUC1-MBP protein combined BCG has significant anti-tumoreffect on transplant malignant melanoma while MUC1peptides combined BCGhas no evident anti-tumor effect. The results further suggest the valuation ofMUC1-MBP combined with BCG as a anti-tumor vaccine.
引文
[1] Dillman R. Cancer immunotherapy [J]. Cancer Biotherapy andRadiopharmaceuticals,2011,26(1):1–64.
    [2] Palucka K,Ueno H, Banchereau J. Recent developments in cancervaccines [J]. Journal of Immunology,2011,186(3):1325–1331.
    [3] McGuckin MA. CD227(MUC1)–summary and workshop report. In:Leucocyte Typing VII. Mason D (Ed.). Oxford University Press, Oxford,UK,2001:54–56.
    [4] Apostolopoulos V, Hu XF, Pouniotis DS, Xing PX.MUC1: a molecule ofmany talents [J]. Curr. Trends Immunol,2003(12):629–639.
    [5] Itoh Y, Kamata-Sakurai M, Denda-Nagai K et al. Identification andexpression of human epiglycanin/MUC21: a novel transmembrane mucin[J]. Glycobiology,2008,18(1):74–83.
    [6] Vlad AM, Kettel JC, Alajez NM, Carlos CA, Finn OJ. MUC1immunobiology: from discovery to clinical applications [J]. Adv. Immunol,2004(82):249–293.
    [7] Apostolopoulos V, McKenzie IF, Pietersz GA. Breast cancerimmunotherapy: current status and future prospects [J]. Immunol. Cell Biol,1996,74(5):457–464.
    [8] Xing PX, Michaels M, Apostolopoulos V, Prenzoska J, Bishop J, MckenzieIFC. Phase I study of synthetic MUC1peptides in cancer [J]. Int. J. Oncol,1995(6):1283–9.
    [9] Apostolopoulos V, Pietersz GA, McKenzie IF. MUC1and breast cancer [J].Curr. Opin. Mol. Ther,1999,1(1):98–103.
    [10] Graham RA, Stewart LS, Beverley P, Papadimitriou JT. MUC1-basedimmunogens for tumour therapy: development of murine model systems [J].Tumour Targeting,1995(1):211–21.
    [11] Gendler SJ, Spicer AP. Epithelial mucin genes [J]. Annu. Rev. Physiol,1995(57):607–34.
    [12] Engelmann K, Shen H, Finn OJ. MCF7side population cells withcharacteristics of cancer stem/progenitor cells express the tumor antigenMUC1[J]. Cancer Res,2008,68(7):2419–2426.
    [13] Acres B, Limacher JM. MUC1as a target antigen for cancerimmunotherapy [J]. Expert Rev. Vaccines,2005,4(4):493–502.
    [14] Pilla L, Rivoltini L, Patuzzo R, Marrari A, Valdagni R, Parmiani G.Multipeptide vaccination in cancer patients [J]. Expert Opinion onBiological Therapy,2009,9(8):1043–1055.
    [15] Ramanathan RK, Lee KM, Mckolanis J et al. Phase I study of a MUC1vaccine composed of different doses of MUC1peptide with SB-AS2adjuvant in resected and locally advanced pancreatic cancer [J]. CancerImmunol. Immunother,2005,54(3):254–264.
    [16] Yamamoto K, Ueno T, Kawaoka T et al. MUC1peptide vaccination inpatients with advanced pancreas or biliary tract cancer [J]. AntiCancerRes,2005,25(5):3575–3579.
    [17] Gilewski T, Adluri S, Ragupathi G et al. Vaccination of high-risk breastcancer patients with mucin-1(MUC1) keyhole limpet hemocyaninconjugate plus QS-21[J]. Clin. Cancer Res,2000,6(5):1693–1701.
    [18] Reddish M, Maclean GD, Koganty RR et al. Anti-MUC1class I restrictedCTLs in metastatic breast cancer patients immunized with a syntheticMUC1peptide [J]. Int. J. Cancer,1998,76(6):817–823.
    [19] Musselli C, Ragupathi G, Gilewski T, Panageas KS, Spinat Y, LivingstonPO:Reevaluation of the cellular immune response in breast cancer patientsvaccinated with MUC1[J]. Int. J. Cancer,2002,97(5):660–667.
    [20] Butts C, Murray N, Maksymiuk A et al. Randomized Phase IIB trial ofBLP25liposome vaccine in stage IIIB and IV non-small-cell lung cancer[J]. J. Clin. Oncol,2005,23(27):6674–6681.
    [21] Mellstedt H, Vansteenkiste J, Thatcher N. Vaccines for the treatment ofnon-small cell lung cancer: investigational approaches and clinicalexperience [J]. Lung Cancer,2011,73(1):11–17.
    [22] Lees CJ, Apostolopoulos V, Acres B et al. Immunotherapy withmannan-MUC1and IL12in MUC1transgenic mice [J]. Vaccine,2000,19(2–3):158–162.
    [23] Karanikas V, Lodding J, Maino VC, Mckenzie IF. Flow cytometricmeasurement of intracellular cytokines detects immune responses in MUC1immunotherapy [J]. Clin. Cancer Res,2000,6(3):829–837.
    [24] Apostolopoulos V, Pietersz GA, Tsibanis A et al. Pilot Phase IIIimmunotherapy study in early-stage breast cancer patients using oxidizedmannan-MUC1[ISRC TN71711835]. Breast Cancer Res,2006,8(3), R27.
    [25] Tang CK, Katsara M, Apostolopoulos V. Strategies used for MUC1immunotherapy: human clinical studies [J]. Expert Rev. Vaccines,2008,7(7):963–975.
    [26] Sheng KC, Pouniotis DS, Wright MD et al. Mannan derivatives inducephenotypic and functional maturation of mouse dendritic cells [J].Immunology,2006,118(3):372–383.
    [27] Acres B. Cancer immunotherapy: phase II clinical studies with TG4010(MVA-MUC1-IL2)[J]. J BUON,2007,12: S71-5.
    [28] Ramlau R, Quoix E, Rolski J et al.: A Phase II study of Tg4010(MVA-MUC1-IL2) in association with chemotherapy in patients with stageIII/IV non-small cell lung cancer [J]. J. Thorac. Oncol,2008,3(7):735–744.
    [29] Dreicer R, Stadler WM, Ahmann FR et al. MVA-MUC1-IL2vaccineimmunotherapy (Tg4010) improves PSA doubling time in patients withprostate cancer with biochemical failure [J]. Invest. New Drugs,200927(4):379–386.
    [30] Nencioni A, Grunebach F, Schmidt SM et al. The use of dendritic cells incancer immunotherapy [J]. Crit. Rev. Oncol. Hematol.2008,65(3):191–199.
    [31] Soares MM, Mehta V, Finn OJ. Three different vaccines based on the140-amino acid MUC1peptide with seven tandemly repeatedtumor-specific epitopes elicit distinct immune effector mechanisms inwild-type versus MUC1-transgenic mice with different potential for tumorrejection [J]. J. Immunol,2001,166(11):6555–6563.
    [32] Miles DW, Happerfield LC, Smith P et al. Expression of sialyl-Tn predictsthe effect of adjuvant chemotherapy in node-positive breast cancer [J]. Br. J.Cancer,1994,70(6):1272–1275.
    [33] Miles D, Papazisis K. Rationale for the clinical development of STn-KLH(theratope) and anti-MUC-1vaccines in breast cancer [J]. Clin. BreastCancer,2003,3(Suppl.4): S134–S138.
    [34] Miles DW, Towlson KE, Graham R et al. A randomised Phase II study ofsialyl-Tn and detox-B adjuvant with or without cyclophosphamidepretreatment for the active specific immunotherapy of breast cancer [J]. Br.J. Cancer,1996,74(8):1292–1296.
    [35] Holmberg LA, Sandmaier BM: Vaccination with theratope (STn-KLH) astreatment for breast cancer [J]. Expert Rev. Vaccines,2004,3(6):655–663.
    [36] Julien S, Picco G, Sewell R et al. Sialyl-Tn vaccine inducesantibody-mediated tumour protection in a relevant murine model [J]. Br. J.Cancer,2009,100(11):1746–1754.
    [37] Karanikas V, Hwang LA, Pearson J, et al. Antibody and T cell responses ofpatients with adenocarcinoma immunized with mannan-MUC1fusionprotein [J]. J Clin Invest,1997(100):2738–2792.
    [38] Stefan etal. Potential role for toll-like receptor4in mediating Escherichiacoli maltose-binding protein activation of dendritic cells [J]. Infection andImmunity,2007,75(3):1359–1363.
    [39]台桂香,张吉凤,朱讯.重组人MUC1-MBP融合蛋白的抗肿瘤作用[J].中国肿瘤生物治疗杂志,2003,10(3):202–205.
    [40]宋献美,方芳,马吉春,赵小霞,周静,台桂香.利用小鼠皮下人乳腺癌移植瘤模型观察MUC1-MBP抗乳腺癌作用[J].免疫学杂志,2010,26(5):428–432.
    [41] Apostolopoulos V, Pieterz GA, Xing PX, et al. The immunogenicity ofMUC1peptides and fusion Protein. Cancer Letters [J],1995,90:2
    [42]方芳.重组MUC1-MBP融合蛋白疫苗抗肿瘤作用机制研究[D].长春:吉林大学白求恩医学院,2010.
    [43]张庆勇.大肠杆菌麦芽糖结合蛋白(MBP)免疫增强活性及抗肿瘤作用研究[D].长春:吉林大学白求恩医学院,2011.
    [44] Kang QZ,.Duan GC, Fan QT, Xi YL. Fusion expression of Helieobaeterpylorineutrophil-activating protein in E.coli [J]. World J. Gastroenterol,2005,11:454–456.
    [45] Simmons M, Murphy GS,.Kochel T, Raviprakash K, Hayes CG.Characterization of antibody responses to combinations of a dengue-2DNAand dengue-2recombinant subunit vaccine [J]. Am. J. Trop. Med. Hyg,2001,65:420–426.
    [46] Kushwaha A, Rao PP, Suresh RP. Chauhan VS. Immunogenicity ofreeombinant fragments of Plasmodium falciparum acidic basic repeatantigen produced in Escherichia coli [J]. Parasite Immunol,2001,23:435–444.
    [47] Fernandez Stefan, Palmer DuPeh R, Simmons Monika, etal. Potential roleforToll-like receptor4in mediating Escherichia coli maltose-binding proteinactivation of dendritic cells [J]. Infect. Immun,2007,75:1359–1363.
    [48]赵小霞,马吉春,方芳,台桂香,等.大肠杆菌麦芽糖结合蛋白(MBP)诱导小鼠Thl细胞的活化作用[J].中国免疫学杂志,2009,25:504-507.
    [49] Daugelat S,Ladel CH,Kaufmann SH. Influence of mouse strain andvaccine viability on T cell responses induced by Mycobacterium bovisbacillus Calmette Guerin [J]. Infect Immunol,1995,63:2033.
    [50] Flynn J L, Chan J. Immunology of tuberculosis [J]. Annu Rev Immunol,2001,19:93-129.
    [51] Zhang S, Graeber LA, Helling F, Ragupathi G, Adluri S, Lloyd KO, andLivingston PO. Augmenting Immunogenicity of synthetic MUC1peptidevaccines in mice[J]. Cancer Res,1996,56:3315-3319.