[HTML][HTML] Empirical identification and validation of tumor-targeting T cell receptors from circulation using autologous pancreatic tumor organoids

Q Meng, S Xie, GK Gray, MH Dezfulian… - … for immunotherapy of …, 2021 - ncbi.nlm.nih.gov
Q Meng, S Xie, GK Gray, MH Dezfulian, O Gandarilla, W Li, L Huang, D Akshinthala, E Ferrer
Journal for immunotherapy of cancer, 2021ncbi.nlm.nih.gov
Background Tumor-specific cytotoxic T cells and T cell receptors are effective tools for
cancer immunotherapy. Most efforts to identify them rely on known antigens or lymphocytes
that have infiltrated into the tumor bed. Approaches to empirically identify tumor-targeting T
cells and T cell receptors by exploiting all antigens expressed on tumor cell surfaces are not
well developed for most carcinomas, including pancreatic cancer. Methods Autologous
tumor organoids were stimulated with T cells from the patients' peripheral blood for 2 weeks …
Abstract
Background
Tumor-specific cytotoxic T cells and T cell receptors are effective tools for cancer immunotherapy. Most efforts to identify them rely on known antigens or lymphocytes that have infiltrated into the tumor bed. Approaches to empirically identify tumor-targeting T cells and T cell receptors by exploiting all antigens expressed on tumor cell surfaces are not well developed for most carcinomas, including pancreatic cancer.
Methods
Autologous tumor organoids were stimulated with T cells from the patients’ peripheral blood for 2 weeks to generate the organoid-primed T (opT) cells. opT cell phenotype was analyzed by monitoring changes in the expression levels of 28 cell surface and checkpoint proteins. Expression of ligands of the immune checkpoints was investigated by immunohistochemistry staining. T cells were labeled with carboxyfluorescein succinimidyl ester (CFSE) and assayed by flow cytometry to monitor tumor-induced T cell proliferation changes. opT cell-mediated killing of three-dimensional organoids was measured using an M30 ELISA kit. T cell receptors (TCRs) were identified by deep sequencing of gDNA isolated from T cells, and the TCR specificity was confirmed by transferring TCRs to the T cell line SKW-3 or donor T cells.
Results
The co-culture was effective in the generation of CD8+ or CD4+ opT cells. The opT cells killed autologous tumors in a granzyme B or Fas-Fas ligand-dependent manner and expressed markers of tissue-resident memory phenotype. Each patient-derived opT cell culture displayed a unique complement of checkpoint proteins. Interestingly, only NKG2A blockade showed a potent increase in the interferon-γ production compared with blocking programmed cell death protein 1 (PD-1) or programmed cell death ligand 1 (PD-L1) or TIM3 or TIGIT or LAG3. Importantly, TCR sequencing demonstrated a dramatic clonal expansion of T cells with a restricted subset of TCRs. Cloning and transferring the TCRs to heterologous T cells was sufficient to confer tumor cell recognition and cytotoxic properties in a patient-specific manner.
Conclusion
We report a platform for expanding tumor-targeting T cells from the peripheral blood of patients with pancreatic cancer. We identify the NKG2A-HLA-E axis as a potentially important checkpoint for CD8+ T cells for pancreatic cancer. Lastly, we demonstrate empirical identification of tumor-targeting TCRs that can be used for TCR-therapeutics.
ncbi.nlm.nih.gov