Adoptive T-cell therapy of rhabdomyosarcoma

作者:Simon Keller K*; Paschen A; Eichmueller S; Gattenloehner S; Barth S; Koscielniak E; Leuschner I; Stoebel P; Hombach A; Abken H; Marx A
来源:Pathologe, 2010, 31: 215-220.
DOI:10.1007/s00292-010-1344-8

摘要

Aims. To improve survival of patients with advanced rhabdomyosarcomas (RMS), we aimed to adoptively transfer T-cells with redirected specificity for the fetal acetylcholine receptor (AChR), an RMS-specific cell surface antigen.
Methods. A "second generation" chimeric antigen receptor (CAR) with a combined CD28-CD3 zeta signaling domain was derived from our previously described chimeric antigen receptor composed of an extracellular human anti-fAChR antibody fragment, an Fc hinge region, and the intracellular T-cell receptor zeta chain. Lymphocytes from the peripheral blood were modified by retroviral transduction and monitored by FACS analysis. Cytotoxicity of modified T-cells towards RMS cells was recorded by MTT-based viability tests; expression of co-stimulatory molecules and anti-apoptotic genes was studied by FACS and qRT-PCR analysis.
Results. Co-stimulatory molecules were expressed in low levels on RMS cells giving the rationale to generate a CD28-CD3 zeta signalling CAR (chimeric antigen receptor) for redirecting T-cells. T-cells were successfully engineered with the "second generation" AChRspecific chimeric antigen receptor. Despite of high CAR expression engineered T-cells showed low killing efficiency towards RMS compared to redirected killing of CD20+ lymphoma or CEA-expressing adenocarcinoma cell lines when redirected by CD20- and/or CEA-specific CAR.
Conclusions. Data suggest that RMS cells exhibit resistance to a T-cell attack redirected by a fAChR-specific CAR. Inhibition of anti-apoptotic pathways in those cells may improve sensitivity to conventional as well as T-cell-based therapeutics.

  • 出版日期2010-10