Targeting HER-2/neu in Early Breast Cancer Development Using Dendritic Cells with Staged Interleukin-12 Burst Secretion

BJ Czerniecki, GK Koski, U Koldovsky, S Xu, PA Cohen… - Cancer research, 2007 - AACR
BJ Czerniecki, GK Koski, U Koldovsky, S Xu, PA Cohen, R Mick, H Nisenbaum, T Pasha…
Cancer research, 2007AACR
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-γ and bacterial lipopolysaccharide to …
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/neupos 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-γ 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 CD8pos 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-γ–secreting CD4pos (85%) and CD8pos (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. [Cancer Res 2007;67(4):1842–52]
AACR