Generation of an optimized polyvalent monocyte-derived dendritic cell vaccine by transfecting defined RNAs after rather than before maturation

N Schaft, J Dörrie, P Thumann, VE Beck… - The Journal of …, 2005 - journals.aai.org
N Schaft, J Dörrie, P Thumann, VE Beck, I Müller, ES Schultz, E Kämpgen…
The Journal of Immunology, 2005journals.aai.org
Transfection with RNA is an attractive method of Ag delivery to dendritic cells (DCs), but has
not yet been standardized. We describe in this study the methods to efficiently generate an
optimized mature monocyte-derived DC vaccine at clinical scale based on the
electroporation of several RNAs either into immature DC followed by maturation or,
alternatively, directly into mature DCs, which has not been possible so far with such high
efficiency. Electroporation of DCs resulted in high yield, high transfection efficiency (> 90%) …
Abstract
Transfection with RNA is an attractive method of Ag delivery to dendritic cells (DCs), but has not yet been standardized. We describe in this study the methods to efficiently generate an optimized mature monocyte-derived DC vaccine at clinical scale based on the electroporation of several RNAs either into immature DC followed by maturation or, alternatively, directly into mature DCs, which has not been possible so far with such high efficiency. Electroporation of DCs resulted in high yield, high transfection efficiency (> 90%), and high migration capacity. Intracellular staining allowed the study of the expression kinetics of Ags encoded by the transfected RNAs (MelanA, MAGE-3, and survivin) and a validation of the vaccine (≥ 90% transfection efficiency). Expression of all three Ags peaked 3–4 h after electroporation in DC transfected either before or after maturation, but decreased differently. The DC vaccine can also be cryopreserved and nevertheless retains its viability, stimulatory capacity as well as migratory activity. In addition, we uncover that DC transfected after rather than before maturation appear to be preferable vaccines not only from a production point of view but also because they appear to be immunologically superior for CTL induction in sharp contrast to common belief. DCs transfected after maturation not only more effectively generate and present the Mage-3. A1 and MelanA. A2. 1 epitopes to T cell clones, but they even are superior in priming to the standard proteasome-dependent MelanA. A2. 1 wild-type prototype tumor epitope, both in terms of T cell expansion and effector function on a per cell basis.
journals.aai.org