首页 | 本学科首页   官方微博 | 高级检索  
     


Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion
Authors:Czerniecki Brian J  Koski Gary K  Koldovsky Ursula  Xu Shuwen  Cohen Peter A  Mick Rosemarie  Nisenbaum Harvey  Pasha Terry  Xu Min  Fox Kevin R  Weinstein Susan  Orel Susan G  Vonderheide Robert  Coukos George  DeMichele Angela  Araujo Louis  Spitz Francis R  Rosen Mark  Levine Bruce L  June Carl  Zhang Paul J
Affiliation:Department of Surgery, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA. brian.czerniecki@uphs.upenn.edu
Abstract:Overexpression of HER-2/neu (c-erbB2) is associated with increased risk of recurrent disease in ductal carcinoma in situ (DCIS) and a poorer prognosis in node-positive breast cancer. We therefore examined the early immunotherapeutic targeting of HER-2/neu in DCIS. Before surgical resection, HER-2/neu(pos) DCIS patients (n = 13) received 4 weekly vaccinations of dendritic cells pulsed with HER-2/neu HLA class I and II peptides. The vaccine dendritic cells were activated in vitro with IFN-gamma and bacterial lipopolysaccharide to become highly polarized DC1-type dendritic cells that secrete high levels of interleukin-12p70 (IL-12p70). Intranodal delivery of dendritic cells supplied both antigenic stimulation and a synchronized preconditioned burst of IL-12p70 production directly to the anatomic site of T-cell sensitization. Before vaccination, many subjects possessed HER-2/neu-HLA-A2 tetramer-staining CD8(pos) T cells that expressed low levels of CD28 and high levels of the inhibitory B7 ligand CTLA-4, but this ratio inverted after vaccination. The vaccinated subjects also showed high rates of peptide-specific sensitization for both IFN-gamma-secreting CD4(pos) (85%) and CD8(pos) (80%) T cells, with recognition of antigenically relevant breast cancer lines, accumulation of T and B lymphocytes in the breast, and induction of complement-dependent, tumor-lytic antibodies. Seven of 11 evaluable patients also showed markedly decreased HER-2/neu expression in surgical tumor specimens, often with measurable decreases in residual DCIS, suggesting an active process of "immunoediting" for HER-2/neu-expressing tumor cells following vaccination. DC1 vaccination strategies may therefore have potential for both the prevention and the treatment of early breast cancer.
Keywords:
本文献已被 PubMed 等数据库收录!
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号