Article Text

Download PDFPDF

Chimeric antigen receptors (CARs) incorporating mutations in the IgG4 Fc spacer region to eliminate Fc receptor recognition results in improved CAR T cell persistence and anti-tumor efficacy
  1. Mahesh Jonnalagadda1,
  2. Armen Mardiros1,
  3. Lauren Hoffman1,
  4. Alyssa Bernanke1,
  5. Wen-Chung Chang1,
  6. William Bretzlaff1,
  7. Renate Starr1,
  8. Xiuli Wang1,
  9. Julie Ostberg1,
  10. Christine Brown1 and
  11. Stephen J Forman1
  1. Aff1 grid.410425.60000000404218357Hematology & Hematopoietic Cell TransplantationBeckman Research Institute and City of Hope National Medical Center Duarte CA USA

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Meeting abstracts

Adoptive immunotherapy using T cells genetically redirected via expression of chimeric antigen receptors (CARs) is a promising approach for cancer treatment. However, this immunotherapy is dependent in part on the optimal molecular design of the CAR, which involves an extracellular ligand-binding domain connected to an intracellular signaling domain by spacer and/or transmembrane sequences. CAR designs frequently incorporate extracellular linker regions based on the immunoglobulin constant regions of either IgG1 or IgG4. In this study we evaluated the potential for the IgG4-Fc linker to result in off-target interactions between the CAR and Fc gamma receptors (FcγRs). As proof of principle, we have focused on a CD19-specific CD19scFv-IgG4-CD28-zeta CAR, and indeed found that CAR+ T cells bound to soluble FcγRs, and did not engraft in NSG mice compared to CAR-negative T cells that only expressed an EGFRt tracking marker. We hypothesized that mutations to avoid FcγR interactions would improve CAR+ T cell persistence and anti-tumor efficacy. To this end, we generated a CD19-specific CAR that has been mutated at two sites within the CH2 region (L235E; N297Q) of the IgG4 Fc spacer, here called CD19R(EQ), as well as a CD19-specific CAR that has a CH2 deletion in its IgG4 Fc spacer (CD19Rch2Δ). These mutations/deletion do not alter the functional ability of the CAR, when expressed by T cells, to mediate antigen-specific lysis of tumor cells. However, compared to T cells that express a non-mutated CAR, T cells expressing the CD19R(EQ) and CD19Rch2Δ exhibit impaired binding to recombinant soluble FcγRs. These CD19R(EQ) and CD19Rch2Δ T cells also exhibit improved engraftment in NSG mice. Indeed the engraftment levels seen with the mutated CAR were similar to that seen with CAR-negative T cells that only expressed the EGFRt tracking marker. Importantly, elimination of CAR/FcγR interactions also significantly improves CD19-specific CAR+ T cell anti-lymphoma efficacy in NSG mice. These studies provide evidence that optimal CAR function necessitates the elimination of cellular FcγR interactions in order to improve T cell persistence and anti-tumor responses.