Route of administration modulates the induction of dendritic cell vaccine–induced antigen-specific T cells in advanced melanoma patients

WJ Lesterhuis, IJM de Vries, G Schreibelt… - Clinical Cancer …, 2011 - AACR
WJ Lesterhuis, IJM de Vries, G Schreibelt, AJA Lambeck, EHJG Aarntzen, JFM Jacobs…
Clinical Cancer Research, 2011AACR
Purpose: It is unknown whether the route of administration influences dendritic cell (DC)-
based immunotherapy. We compared the effect of intradermal versus intranodal
administration of a DC vaccine on induction of immunologic responses in melanoma
patients and examined whether concomitant administration of interleukin (IL)-2 increases
the efficacy of the DC vaccine. Experimental Design: HLA-A2. 1+ melanoma patients
scheduled for regional lymph node dissection were vaccinated four times biweekly via …
Abstract
Purpose: It is unknown whether the route of administration influences dendritic cell (DC)-based immunotherapy. We compared the effect of intradermal versus intranodal administration of a DC vaccine on induction of immunologic responses in melanoma patients and examined whether concomitant administration of interleukin (IL)-2 increases the efficacy of the DC vaccine.
Experimental Design: HLA-A2.1+ melanoma patients scheduled for regional lymph node dissection were vaccinated four times biweekly via intradermal or intranodal injection with 12 × 106 to 17 × 106 mature DCs loaded with tyrosinase and gp100 peptides together with keyhole limpet hemocyanin (KLH). Half of the patients also received low-dose IL-2 (9 MIU daily for 7 days starting 3 days after each vaccination). KLH-specific B- and T-cell responses were monitored in blood. gp100- and tyrosinase-specific T-cell responses were monitored in blood by tetramer analysis and in biopsies from delayed-type hypersensitivity (DTH) skin tests by tetramer and functional analyses with 51Cr release assays or IFNγ release, following coculture with peptide-pulsed T2 cells or gp100- or tyrosinase-expressing tumor cells.
Results: In 19 of 43 vaccinated patients, functional tumor antigen–specific T cells could be detected. Although significantly more DCs migrated to adjacent lymph nodes upon intranodal vaccination, this was also highly variable with a complete absence of migration in 7 of 24 intranodally vaccinated patients. Intradermal vaccinations proved superior in inducing functional tumor antigen–specific T cells. Coadministration of IL-2 did not further augment the antigen-specific T-cell response but did result in higher regulatory T-cell frequencies.
Conclusion: Intradermal vaccination resulted in superior antitumor T-cell induction when compared with intranodal vaccination. No advantage of additional IL-2 treatment could be shown. Clin Cancer Res; 17(17); 5725–35. ©2011 AACR.
AACR