Abstract 1209: Screeningex vivoconditions that increase memory T cell frequency using high throughput flow cytometry and an optimized multiplexed assay

Author(s):  
Zhaoping Liu ◽  
Andrea Donart-Gomez ◽  
John O'Rourke
2020 ◽  
Vol 2020 ◽  
pp. 1-2
Author(s):  
Iole Macchia ◽  
Valentina La Sorsa ◽  
Irene Ruspantini ◽  
Massimo Sanchez ◽  
Valentina Tirelli ◽  
...  

1996 ◽  
Vol 38 (1) ◽  
pp. 25-29 ◽  
Author(s):  
Naoki Kunugita ◽  
Nan Mei ◽  
Satoshi Nomoto ◽  
Toshiyuki Norimura

2018 ◽  
Vol 23 (7) ◽  
pp. 603-612
Author(s):  
Emily M. Martinez ◽  
Samuel D. Klebanoff ◽  
Stephanie Secrest ◽  
Gabrielle Romain ◽  
Samuel T. Haile ◽  
...  

High-throughput flow cytometry is an attractive platform for the analysis of adoptive cellular therapies such as chimeric antigen receptor T cell therapy (CAR-T) because it allows for the concurrent measurement of T cell–dependent cellular cytotoxicity (TDCC) and the functional characterization of engineered T cells with respect to percentage of CAR transduction, T cell phenotype, and measurement of T cell function such as activation in a single assay. The use of adherent tumor cell lines can be challenging in these flow-based assays. Here, we present the development of a high-throughput flow-based assay to measure TDCC for a CAR-T construct co-cultured with multiple adherent tumor cell lines. We describe optimal assay conditions (such as adherent cell dissociation techniques to minimize impact on cell viability) that result in robust cytotoxicity assays. In addition, we report on the concurrent use of T cell transduction and activation antibody panels (CD25) that provide further dissection of engineered T cell function. In conclusion, we present the development of a high-throughput flow cytometry method allowing for in vitro interrogation of solid tumor, targeting CAR-T cell–mediated cytotoxicity, CAR transduction, and engineered T cell characterization in a single assay.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2545-2545
Author(s):  
David Wu ◽  
Anna Sherwood ◽  
Stuart S. Winter ◽  
Kimberly Dunsmore ◽  
Mignon L Loh ◽  
...  

Abstract Abstract 2545 There is increasing evidence for the utility of minimal residual disease (MRD) assessment in predicting clinical outcomes of patients with T cell lymphoblastic leukemia (T-ALL). Evaluation of MRD by PCR-based analysis of T-cell receptor (TCR) genes has a sensitivity of 10−5, but requires the use of individualized patient-specific primers, which is laborious, expensive and difficult to implement for real-time, clinical decision-making. Multi-parametric flow cytometry is currently limited to a sensitivity of 10−4, requires viable cells, and is poorly standardized. High-throughput DNA sequencing offers the potential to equal or surpass the higher sensitivity of PCR-based MRD testing with reduced cost, improved turn-around time, and better standardization. Paired samples of pediatric T-ALL from 14 patients enrolled on Children's Oncology Group AALL0434 were obtained at diagnosis and at day 29 post-induction therapy. The complementarity determining regions (CDR3) regions of TCRB and TCRG were sequenced for all 28 specimens using an Illumina GA2 platform as previously described (see Blood, 114(19):4099–4107, 2009 and Sci Transl Med. 3(90):90ra61, 2011). Pre-treatment samples were used to obtain unique TCR sequences for the leukemic clone, and post-treatment samples were assessed for the frequency of each TCR sequence as a percentage of the total. The frequency of each sequence was also enumerated in post-treatment samples from all other patients to evaluate specificity. These results were compared to MRD results obtained by 9-color flow cytometry per trial protocol. Eleven of 14 pre-treatment samples (78.6%) had a detectable clonal population based on TCRG sequence analysis, and 10 of these also had a clonal TCRB sequence. Five samples exhibited an additional unique TCRG sequence, consistent either with rearrangement of both TCRG loci or the presence of two clonal subpopulations. Two of 3 cases without a detectable clonal TCR gene sequence had the immunophenotype of early thymic precursor (ETP) T-ALL and would be expected to have germline TCRB and TCRG genes. No other cases were ETP. Clones were found in all 5 informative post-treatment samples positive for MRD by flow cytometry, as well as at a low level in 3 additional patients without MRD by flow cytometry, suggesting superior sensitivity for sequencing. The background sequence frequencies were very low (0–10−5) in other patient post-treatment samples, being slightly higher for TCRG than for TCRB, consistent with germline sequence diversity. We demonstrate the potential of high-throughput sequencing for analysis of MRD in pediatric T-ALL. The number of cases in which the assay is informative (78.6%) is similar to that seen with standard PCR MRD methods, but evaluation of more cases is needed. MRD by sequencing appears to have a higher sensitivity than current flow cytometric methods, although direct comparison of MRD frequencies from the two techniques is problematic and will require normalization. The strong association of ETP status and lack of clonal TCR sequence identification at diagnosis suggests utility in identifying this poor outcome subset of T-ALL. Disclosures: Sherwood: Adaptive TCR, Seattle, WA: Employment, Equity Ownership. Wood:Becton, Dickinson and Company, NJ, USA: Research Funding. Robins:Adaptive TCR, Seattle, WA: Consultancy, Equity Ownership, Patents & Royalties.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A124-A124
Author(s):  
Brikena Gjeci ◽  
Sadik Kassim ◽  
Julian Scherer

BackgroundCAR T-cells have exhibited efficacious treatment of hematological malignancies, such as Acute Myeloid Leukemia (AML). Upon antigen binding, CARs initiate Ca2+-dependent signaling pathways, leading to an increased intracellular concentration of nuclear factor of activated T cells (NFAT), which stimulates downstream T cell effector functions.1 2 However, an objective high-throughput platform to compare CAR-induced T cell activity has been lacking. Here, we report on a CAR screening platform that utilizes an NFAT-sensitive promoter driving fluorescent protein expression in Jurkat T cells. This approach, termed ‘IL-2 Reporter System’ (IRS), is employed for the identification of functional CD33CARs to improve AML CAR T-cell therapies.MethodsLentiviral transduction of two fluorescent proteins (mOrange2 and mTurquoise2) under either the constitutively active EF1alpha (FP1) or the NFAT-sensitive minimal IL-2 promoter (FP2) into Jurkat cells resulted in two IRS cell lines. IRS cell function was confirmed by treatment with phorbol myristate acetate (PMA) and ionomycin to induce activation and thus the expression of FP2. To screen CARs with previously described potential in AML therapy, IRS cells were transduced with eight distinct CD33CAR constructs and co-cultured with CD33+ cells (MOLM13-WT) and CD33 knock-out (MOLM13-CD33KO) cells. IRS cell analysis was performed using flow cytometry, fluorescent microscopy, and IncuCyte live cell imaging.ResultsAs expected, live cell fluorescence imaging revealed that PMA/ionomycin treatment induced expression of FP2. Peak expression occurred at 12–15 hours post-treatment and consistent, high FP2 signal was detected for 24h in both cell lines. Flow cytometry analysis 24hr post treatment determined that 60–80% of transduced IRS cells expressed FP2. Normalized FP2 expression was comparable between both IRS cell lines but varied across the eight CD33CARs. Six of the eight CD33CAR constructs resulted in significant increases (up to 8-fold) in FP2 signal upon exposure to CD33+ cells compared to control CD33KO cells.ConclusionsOur results indicate that IRS cells can be used as an objective, fast, and reliable reporter system for CD33CAR activity. The constitutively expressed FP1 eliminates false negative outcomes and verifies successful transduction of the reporter construct. FP2 expression, driven only in activated cells, represents antigen recognition by and activity of the CAR. Importantly, the use of IRS cells is not restricted to just CD33CARs but provides a platform for CAR and recombinant TCR screenings against any antigen. Additionally, efforts are underway to adapt this platform entirely to microplates and a liquid handling system, allowing for high-throughput screens of several AML-targeting CAR constructs.ReferencesHogan PG. Calcium–NFAT transcriptional signaling in T cell activation and T cell exhaustion. Cell Calcium 2017;63:66–9.Chow C-W, Rincón Mercedes, Davis RJ. Requirement for Transcription Factor NFAT in Interleukin-2 Expression. Molecular and Cellular Biology 1999;19(3):2300–7.


Sign in / Sign up

Export Citation Format

Share Document