Efficient gene transfer into human primary blood lymphocytes by surface-engineered lentiviral vectors that display a T cell–activating polypeptide

M Maurice, E Verhoeyen, P Salmon… - Blood, The Journal …, 2002 - ashpublications.org
M Maurice, E Verhoeyen, P Salmon, D Trono, SJ Russell, FL Cosset
Blood, The Journal of the American Society of Hematology, 2002ashpublications.org
In contrast to oncoretroviruses, lentiviruses such as human immunodeficiency virus 1 (HIV-1)
are able to integrate their genetic material into the genome of nonproliferating cells that are
metabolically active. Likewise, vectors derived from HIV-1 can transduce many types of
nonproliferating cells, with the exception of some particular quiescent cell types such as
resting T cells. Completion of reverse transcription, nuclear import, and subsequent
integration of the lentivirus genome do not occur in these cells unless they are activated via …
In contrast to oncoretroviruses, lentiviruses such as human immunodeficiency virus 1 (HIV-1) are able to integrate their genetic material into the genome of nonproliferating cells that are metabolically active. Likewise, vectors derived from HIV-1 can transduce many types of nonproliferating cells, with the exception of some particular quiescent cell types such as resting T cells. Completion of reverse transcription, nuclear import, and subsequent integration of the lentivirus genome do not occur in these cells unless they are activated via the T-cell receptor (TCR) or by cytokines or both. However, to preserve the functional properties of these important gene therapy target cells, only minimal activation with cytokines or TCR-specific antibodies should be performed during gene transfer. Here we report the characterization of HIV-1–derived lentiviral vectors whose virion surface was genetically engineered to display a T cell-activating single-chain antibody polypeptide derived from the anti-CD3 OKT3 monoclonal antibody. Interaction of OKT3 IgGs with the TCR can activate resting peripheral blood lymphocytes (PBLs) by promoting the transition from G0 to G1 phases of the cell cycle. Compared to unmodified HIV-1–based vectors, OKT3-displaying lentiviral vectors strongly increased gene delivery in freshly isolated PBLs by up to 100-fold. Up to 48% transduction could be obtained without addition of PBL activation stimuli during infection. Taken together, these results show that surface-engineered lentiviral vectors significantly improve transduction of primary lymphocytes by activating the target cells. Moreover these results provide a proof of concept for an approach that may have utility in various gene transfer applications, including in vivo gene delivery.
ashpublications.org