Improvement of UniCAR T cell effectiveness against EGFR+ tumor cells by using different αEGFR targeting module formats


Improvement of UniCAR T cell effectiveness against EGFR+ tumor cells by using different αEGFR targeting module formats

Jureczek, J.; Feldmann, A.; Albert, S.; Bergmann, R.; Berndt, N.; Arndt, C.; Koristka, S.; Bachmann, M.

Since epithelial growth factor receptor (EGFR) mutations or overexpression is linked with variety of malignancies, including lung, breast, stomach, colorectal, head and neck, and pancreatic carcinomas as well as glioblastomas it is an attractive target for tailored treatment of solid cancer. Thus over the last twenty years many strategies targeting EGFR were developed and even clinically approved, including disrupting intracellular signalling involving tyrosine kinase inhibitors (TKIs) or the inhibition of ligand binding using therapeutic monoclonal antibodies like e.g. Cetuximab, Panitumumab or Necitumumab. Unfortunately, cancers treated with these targeted drugs commonly become resistant to them. These limitations justify the need of more efficient therapy options. As chimeric antigen receptor (CAR) engineered T cells highly effectively eliminate hematological malignancies already in the clinics, one idea is to redirect CAR T cells also against EGFR expressing solid cancers. However, CAR T cell therapy can lead to severe even life-threatening side effects and its effectiveness against solid tumors is still limited. Particular worrying is that EGFR is a widespread antigen commonly expressed also on healthy tissues bearing a high risk of severe on-target/off-tumor side effects due to EGFR-targeted therapies, which cannot be controlled in patients. In order to overcome these challenges our UniCAR technology might be an appropriate answer combining high anti-tumor effectiveness, tumor specificity, flexibility, and safety control mechanisms. In contrast to conventional CARs, UniCAR T cells are per se inert because UniCARs are directed against a small peptide epitope, which is not present on living cells. The redirection of UniCAR T cells to tumor cells occurs only in the presence of a tumor specific targeting module (TM). TMs, on one hand carry the specificity for a certain tumor antigen and on the other hand contain the UniCAR peptide epitope recognized by UniCARs mediating the cross-linkage of UniCAR T cells and antigen presenting tumor cells. As TMs have a very short half-life in vivo they can be used as a switch to control UniCAR T cell activity on demand in patients. In detail, UniCAR T cells are only switched on in the presence of antigen specific TMs realized by permanent TM infusion, but could be rapidly switched off when the application of the TM is stopped and the TM is eliminated. Meanwhile we successfully generated a series of different TMs against different tumor antigens and entities. Interestingly, TMs can be made of different molecules showing various structures and can flexible be exchanged in order to target any tumor antigen and overcome tumor escape variants. Commonly our TMs consist of a humanized single-chain variable fragment (scFv) derived from the variable heavy and light chain domains of a murine monoclonal antibody. In addition, we successfully generated TMs based on different monovalent and bivalent antibody derivatives, nanobodies derived from one variable camelid antibody domain, affibodies and even small peptide molecules.
Recently we demonstrated proof-of-concept for the redirection of UniCAR T cells to EGFR expressing tumor cells by a nanobody based αEGFR TM derived from the camelid αEGFR antibody 7C12. Considering that the affinity and anti-tumor efficiency of the eucaryotically expressed αEGFR nanobody based TM was limited, we therefore asked the question, whether we could further improve the therapeutic effect against EGFR positive tumor cells using the UniCAR technology. In order to answer this question, we generated a novel TM based on a scFv derived from the clinically used chimeric monoclonal antibody Cetuximab (IMC C-225). In detail, we designed a murine and humanized αEGFR scFv TM, successfully expressed them in mammalian cell lines and compared their functionality with the eucaryotic αEGFR nanobody-based TM in vitro and in vivo. In principle, we observed that both TM formats, the αEGFR nanobody as well as the scFv-based TM, are able to redirect UniCAR T cells eliminate EGFR-expressing tumor cells in an antigen-specific and TM-dependent manner. As both the murine and humanized scFv TM variants worked equally well, obviously humanization of the αEGFR scFv does not affect its functionality. However most interestingly, the tumor killing efficiency of the αEGFR scFv TM was significantly superior in comparison to the αEGFR nanobody based TM. Here, the half maximal effective TM concentration (EC50) value of scFv based TM was improved 1000-fold, from nM to pM range. Consequently, UniCAR T cells in combination with the scFv based TM efficiently eliminate also target cells expressing a low EGFR density level, while UniCAR T cells redirected by the nb based TM clearly attack only highly EGFR expressing tumor cells. Furthermore, the high anti-tumor efficacy of the αEGFR scFv TM over nb TM was manifested in experimental mice.
In summary, we successfully established different αEGFR TM formats that are able to redirect UniCAR T cells to eliminate EGFR-positive tumor cells. However, the analysed αEGFR TM formats differ with respect to their anti-tumor efficiency, which might decide whether UniCAR T cells attack target cells showing different EGFR density levels.

  • Poster
    Tumor immunology meets oncology (TIMO XV), 25.-27.04.2019, Halle, Deutschland

Permalink: https://www.hzdr.de/publications/Publ-29214