Journal Information
Vol. 42. Issue S1.
Pages 9-10 (October 2020)
Share
Share
Download PDF
More article options
Vol. 42. Issue S1.
Pages 9-10 (October 2020)
SP 17
Open Access
Ex vivo activation of pleural T cells in pleural malignancies
Visits
1386
Vera S. Donnenberg, James D. Luketich, Albert D. Donnenberg
This item has received

Under a Creative Commons license
Article information
Full Text

Introduction: MPE are uniformly fatal. It is estimated that the incidence of MPE in the United States is more than 150,000 cases per year, making this a common terminal pathway for a variety of cancers and a dire problem without a solution. Currently available cellular therapeutics are costly and often lack polyclonality, polyfunctionality, and the ability to persist as central memory. The treatment of this deadly complication is potentially at a turning point if the rich immune infiltrates that characterize the majority of effusions can be redirected to an efficacious anti-tumor response. Despite this promise, pleural immune infiltrates have not been used to generate effector cells for adoptive cellular therapy.

Objectives: We have exploited the heterogeneous cellular composition of MPE by piloting the generation of therapeutic T-cell products, using conventional methods used for expanding tumor-infiltrating lymphocytes (TIL). The advantages of plural T cells are: (1) Fewer cycles of expansion owing to several orders of magnitude greater starting number of T cells; (2) Greater initial clonal and functional heterogeneity; (3) Likelihood of preserving polyclonality, polyfunctionality and central memory.

Results: MPE have abundant tumor infiltrating CD3+ T-cells, CD19+ B-cells, CD14+ macrophages, and EpCAM-/Cytokeratin+ mesothelial cells. Regulatory T-cells, which may be abundant in TIL, are low or absent in MPE. Our laboratory's average recovery of viable nucleated cells per MPE is 7.8±4.0×108 cells, with viability exceeding 95%. The cellular composition (tumor, lymphocytes, macrophages, neutrophils, mesothelial cells) varies from patient to patient, but T-cell recovery averages 2.0±1.6×108 (mean, SD). In pilot experiments we cultured whole breast cancer MPE in the presence of anti-CD3/anti-CD28 Dynal beads, IL-2 and IL-7 for 96h. CD3+ T cells were FACS-sorted and added to autologous tumor monolayer cultures and expanded for an additional passage (2 weeks). Expanded passage 2 T cells were compared to freshly isolated T cells (2nd MPE drainage) for ability to kill autologous tumor and non-tumor targets (live cell imaging). Expanded T cells were potently cytotoxic, whereas freshly isolated MPE had no activity against autologous tumor. Expanded T cells did not kill the autologous non-tumor target (adherent cells isolated from peripheral blood). Additionally, we tested freshly isolated breast cancer MPE T cells for the ability to secrete cytokines associated with expansion and effector generation (IL-2, IFNγ and TNFα). We also measured the immunosuppressive cytokine IL-10. Freshly isolated plastic nonadherent cells from a breast cancer MPE were incubated with TPA+ ionomycin for 1h, followed by brefeldin for 2h. CD4+ T cells (85%) and CD8+ T cells (9%) were gated on cells co-expressing intracellular IL-2 and IFNγ. Polyfunctional T cells, defined as IL-2+/IFNγ+/TNFα+/IL-10-, comprised 0.38%, and 0.82% of CD4+ and CD8+ T cells. Unstimulated control cultures constitutively secreted IL-10 and IFNγ but not IL-2 or TNFα.

Conclusions: Pleural infiltrating T-cells represent an attractive source of T cells for immunotherapy. They are numerous, readily expandable without protracted passage and can be induced to secrete immunostimulatory and effector cytokines and specifically kill autologous tumor.

Idiomas
Hematology, Transfusion and Cell Therapy
Article options
Tools