T-cell receptor gene therapy in human melanoma-bearing immune-deficient mice: human but not mouse T cells recapitulate outcome of clinical studies

T Straetemans, M Coccoris, C Berrevoets… - Human gene …, 2012 - liebertpub.com
T Straetemans, M Coccoris, C Berrevoets, E Treffers-Westerlaken, CEV Scholten…
Human gene therapy, 2012liebertpub.com
Adoptive cell therapy using T-cell receptor (TCR)–engineered T cells is a clinically feasible
and promising approach to target tumors, but is currently faced with compromised antitumor
efficacies in patients. Here, we extensively validated immune-deficient mice to facilitate
further development of the therapeutic potential of TCR-engineered T cells. Treatment of
human melanoma-bearing SCID or NSG mice with high doses of human T cells transduced
with an hgp100/HLA-A2-specific TCR did not result in antitumor responses irrespective of …
Abstract
Adoptive cell therapy using T-cell receptor (TCR)–engineered T cells is a clinically feasible and promising approach to target tumors, but is currently faced with compromised antitumor efficacies in patients. Here, we extensively validated immune-deficient mice to facilitate further development of the therapeutic potential of TCR-engineered T cells. Treatment of human melanoma-bearing SCID or NSG mice with high doses of human T cells transduced with an hgp100/HLA-A2-specific TCR did not result in antitumor responses irrespective of chemotherapeutic preconditioning. Imaging of human green fluorescent protein–labeled T cells demonstrated significant T-cell accumulation in intratumoral vasculature directly upon T-cell transfer, which was followed by loss of T cells within 72 hr. Peripheral persistence of human T cells was highly compromised and appeared related to T-cell differentiation. On the contrary, adoptive transfer (AT) of relatively low numbers of hgp100/HLA-A2 TCR-transduced mouse T cells resulted in rapid clearance of large established human melanomas. Unexpectedly and in contrast to reported studies with chimeric antibody receptor–engineered T cells, antitumor activity and homeostatic expansion of T cells were independent of TCR transgene as evidenced in two SCID strains and using two different human melanoma cell lines. Interestingly, the xeno-reactive melanoma response of mouse T cells appeared to be dictated by CD4+ tumor-infiltrating lymphocytes and did not require in vitro T-cell activation, retroviral gene transfer, or subcutaneous interleukin-2 support. Taken together, AT of human but not mouse T cells in human melanoma-bearing immune-deficient mice is in close accordance with clinical studies.
Mary Ann Liebert