首页> 外文学位 >Development and pre-clinical evaluation of a BCG-MUC1-based novel breast cancer vaccine.
【24h】

Development and pre-clinical evaluation of a BCG-MUC1-based novel breast cancer vaccine.

机译:基于BCG-MUC1的新型乳腺癌疫苗的开发和临床前评估。

获取原文
获取原文并翻译 | 示例

摘要

Despite aggressive therapy, breast cancer still has a mortality rate of 25%. Therefore, the development of an effective vaccine may be beneficial for the prevention or adjuvant treatment of this malignancy. We have constructed a novel breast cancer vaccine, BCG-hIL2MUC1, that was designed to preferentially stimulate a Th1 immune response. BCG-hIL2MUC1 consists of BCG that express a truncated form of MUC1 and human interleukin-2 (hIL2). In vitro analysis of BCG-hIL2MUC1 by restriction enzyme mapping, DNA sequencing, and Western blot analysis confirmed the expression of MUC1 and hIL2. The immune response to BCG-hIL2MUC1 was evaluated in SCID mice. SCID mice reconstituted with 50 × 106 human peripheral lymphocytes (hu-PBL-SCID) received three biweekly injections of BCG-hIL2MUC1 (0.5 cfu) and the serum and lymphocyte populations analyzed two weeks after the third immunization. Control animals received PBS, MUC1 peptide (100μg) or BCG-261 (0.5 cfu) vaccination. Flow cytometry documented the increased lymphocyte expression of IL2α receptor on lymphocytes obtained from BCG-hIL2MUC1 immunized animals. These lymphocytes expressed IL2 when exposed to MUC1 peptide. There was minimal MUC1 antibody production. These results suggested that our vaccine had stimulated a Th1 immune response. The ability of BCG-hIL2MUC1 to inhibit breast cancer growth was then evaluated in hu-PBL-SCID mice. After immunization with 3 biweekly vaccinations of BCG-hIL2MUC1, hu-PBL-SCID mice were xenografted with 4 × 106 ZR75-1 human breast cancer cells. Control animals received PBS, MUC1 peptide or BCG-261 vaccination. Outcomes of interest were time to tumor detection and rate of tumor growth. All the animals receiving the control vaccines developed a tumor; only 75% of BCG-hIL2MUC1 immunized animals developed a palpable tumor. The rate of tumor growth was slower in the BCG-hIL2MUC1 immunized animals as compared to the control groups (p 0.05). Histological analysis of the primary tumors in BCG-hIL2MU1 animals revealed areas of apoptosis and reduced MUC1 expression. Our results suggested that BCG-hIL2MUC1 immunization inhibited breast cancer growth in hu-PBL-SCID mice. This inhibition may be due to its ability to stimulate a Th1 immune response. BCG-hIL2MUC1 may be a promising candidate as a breast cancer vaccine.
机译:尽管进行了积极的治疗,乳腺癌的死亡率仍为25%。因此,开发有效的疫苗对于预防或辅助治疗该恶性肿瘤可能是有益的。我们构建了一种新型的乳腺癌疫苗BCG-hIL2MUC1,旨在优先刺激Th1免疫应答。 BCG-hIL2MUC1由表达截短形式的MUC1和人白介素2(hIL2)的BCG组成。通过限制酶定位,DNA测序和Western blot分析对BCG-hIL2MUC1的体外分析证实了MUC1和hIL2的表达。在SCID小鼠中评估了对BCG-hIL2MUC1的免疫应答。用50×10 6 人外周血淋巴细胞(hu-PBL-SCID)重组的SCID小鼠每两周两次注射BCG-hIL2MUC1(0.5 cfu),第三次免疫后两周分析血清和淋巴细胞群。对照动物接受PBS,MUC1肽(100μg)或BCG-261(0.5 cfu)疫苗接种。流式细胞术记录了从经BCG-hIL2MUC1免疫的动物获得的淋巴细胞上IL2α受体的淋巴细胞表达增加。当暴露于MUC1肽时,这些淋巴细胞表达IL2。 MUC1抗体的产生极少。这些结果表明我们的疫苗刺激了Th1免疫反应。然后在hu-PBL-SCID小鼠中评估BCG-hIL2MUC1抑制乳腺癌生长的能力。用BCG-hIL2MUC1的双周免疫接种3次后,将hu-PBL-SCID小鼠异种移植4×10 6 ZR75-1人乳腺癌细胞。对照动物接受PBS,MUC1肽或BCG-261疫苗接种。感兴趣的结果是检测肿瘤的时间和肿瘤的生长速度。所有接受对照疫苗的动物都患有肿瘤;经BCG-hIL2MUC1免疫的动物中,只有75%出现了明显的肿瘤。与对照组相比,经BCG-hIL2MUC1免疫的动物的肿瘤生长速度较慢(p <0.05)。对BCG-hIL2MU1动物的原发肿瘤进行组织学分析,发现其凋亡区域和MUC1表达降低。我们的结果表明,BCG-hIL2MUC1免疫可抑制hu-PBL-SCID小鼠的乳腺癌生长。这种抑制作用可能是由于其刺激Th1免疫反应的能力。 BCG-hIL2MUC1可能是有前途的候选乳腺癌疫苗。

著录项

  • 作者

    Chung, Maureen Angela.;

  • 作者单位

    Brown University.;

  • 授予单位 Brown University.;
  • 学科 Health Sciences Immunology.
  • 学位 Ph.D.
  • 年度 2001
  • 页码 133 p.
  • 总页数 133
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类 预防医学、卫生学;
  • 关键词

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号