2,632
Views
4
CrossRef citations to date
0
Altmetric
Back Matter

T-cells with a single tumor antigen-specific T-cell receptor can be generated in vitro from clinically relevant stem cell sources

ORCID Icon, , , , , , , , & show all
Article: 1727078 | Received 19 Aug 2019, Accepted 19 Dec 2019, Published online: 17 Feb 2020

Figures & data

Figure 1. CD34+ HSPC from adult sources show slower in vitro maturation kinetics and less expansion compared to cord blood HSPC. (a) Culture protocol. Numbers indicate time (in days) of co-culture. Abbreviations: CB, cord blood; mPB, mobilized peripheral blood; BM, bone marrow; DP, double positive; SCF, stem cell factor; FLT3-L, FLT3 ligand; IL, interleukin; RV, retroviral. (b) Kinetics of expansion before transduction in OP9-DL1 co-cultures of HSPC from cord blood (n = 7), healthy donors (n = 12), patients in remission after chemotherapy (n = 15) and AML patients at diagnosis (n = 12). Mean ± s.d. is shown. T-cell committed progenitors in cord blood co-cultures were transduced at day 14, in co-cultures from adult HSPC at later timepoints (d19 or d24). (c) Relative cell numbers (i.e. cell numbers obtained when theoretically starting from a single CD34+ cell at day 0) at day 14 of co-cultures from cord blood (n = 7), healthy donor (n = 13), patient in remission after chemotherapy (n = 16) and AML patient at diagnosis (n = 13) HSPC. Values for individual samples and mean ± s.d. are shown. Mann–Whitney U test was used to assess statistical significance. P-value < 0.05 (*), P < 0.01 (**) and P < 0.001 (***)

Figure 1. CD34+ HSPC from adult sources show slower in vitro maturation kinetics and less expansion compared to cord blood HSPC. (a) Culture protocol. Numbers indicate time (in days) of co-culture. Abbreviations: CB, cord blood; mPB, mobilized peripheral blood; BM, bone marrow; DP, double positive; SCF, stem cell factor; FLT3-L, FLT3 ligand; IL, interleukin; RV, retroviral. (b) Kinetics of expansion before transduction in OP9-DL1 co-cultures of HSPC from cord blood (n = 7), healthy donors (n = 12), patients in remission after chemotherapy (n = 15) and AML patients at diagnosis (n = 12). Mean ± s.d. is shown. T-cell committed progenitors in cord blood co-cultures were transduced at day 14, in co-cultures from adult HSPC at later timepoints (d19 or d24). (c) Relative cell numbers (i.e. cell numbers obtained when theoretically starting from a single CD34+ cell at day 0) at day 14 of co-cultures from cord blood (n = 7), healthy donor (n = 13), patient in remission after chemotherapy (n = 16) and AML patient at diagnosis (n = 13) HSPC. Values for individual samples and mean ± s.d. are shown. Mann–Whitney U test was used to assess statistical significance. P-value < 0.05 (*), P < 0.01 (**) and P < 0.001 (***)

Figure 2. Presence of early T-progenitors at the start of co-cultures. Percentage of early T-progenitors (ETP, lin CD34+ CD38+ CD7+) in CD34+ cells isolated at day 0 of co-cultures from cord blood (n = 7), healthy donors (n = 7), patients in remission (n = 5) and AML patients at diagnosis (n = 9). Individual samples, mean percentages per sample group and s.d. are shown. Mann–Whitney U test was used to assess statistical significance. P-value < 0.01 (**). Other differences were not significant

Figure 2. Presence of early T-progenitors at the start of co-cultures. Percentage of early T-progenitors (ETP, lin− CD34+ CD38+ CD7+) in CD34+ cells isolated at day 0 of co-cultures from cord blood (n = 7), healthy donors (n = 7), patients in remission (n = 5) and AML patients at diagnosis (n = 9). Individual samples, mean percentages per sample group and s.d. are shown. Mann–Whitney U test was used to assess statistical significance. P-value < 0.01 (**). Other differences were not significant

Figure 3. Multiple rounds of agonist peptide stimulation are needed to achieve the selection and maturation of HSPC from adult sources. Maturation of TCR-transduced cells after one or more rounds of agonist peptide stimulation. Contour plots show CD1a (x-axis) and CD27 (y-axis) expression before agonist stimulation and after 1, 2 or 3 rounds of agonist stimulation for TCR-transduced cells in co-cultures from cord blood (a), healthy donors (b), patients in remission after chemotherapy (c) and AML patients at diagnosis (d). Gating on eGFP+ TCR-transduced cells. Numbers indicate percentages of cells in each quadrant. A representative sample from each sample group is shown

Figure 3. Multiple rounds of agonist peptide stimulation are needed to achieve the selection and maturation of HSPC from adult sources. Maturation of TCR-transduced cells after one or more rounds of agonist peptide stimulation. Contour plots show CD1a (x-axis) and CD27 (y-axis) expression before agonist stimulation and after 1, 2 or 3 rounds of agonist stimulation for TCR-transduced cells in co-cultures from cord blood (a), healthy donors (b), patients in remission after chemotherapy (c) and AML patients at diagnosis (d). Gating on eGFP+ TCR-transduced cells. Numbers indicate percentages of cells in each quadrant. A representative sample from each sample group is shown

Figure 4. Phenotype of in vitro generated T-cells after agonist peptide stimulation and after polyclonal feeder expansion. (a) Percentage of cells with a TN (CD45RA+ CD62L+ CXCR3 CD95), TSCM (CD45RA+ CD62L+ CXCR3+ CD95+), TCM (CD45RA CD62L+ CXCR3+ CD95+), TEM (CD45RA CD62L CXCR3 CD95+) and TTE (CD45RA+ CD62L CXCR3 CD95+) phenotype before agonist peptide stimulation, and after 1, 2 or 3 rounds of agonist peptide stimulation. (b) Percentage of cells with a TN, TSCM, TCM, TEM, and TTE phenotype after polyclonal feeder expansion. For (a) and (b) T-cells were generated from HSPC from healthy donors (n = 3), patients in remission (n = 6) and AML patients at diagnosis (n = 3). Gating on eGFP+ TCR-transduced cells. Mean and s.d. are shown. (c) Percentage of cells positive for PD-1 expression after polyclonal feeder expansion. T-cells were generated from HSPC from healthy donors (n = 8), patients in remission after chemotherapy (n = 9) and AML patients at diagnosis (n = 6). Gating on eGFP+ TCR-transduced cells. Individual samples and mean ± s.d. are shown. Mann–Whitney U test was used to assess statistical significance. P-value < 0.05 (*)

Figure 4. Phenotype of in vitro generated T-cells after agonist peptide stimulation and after polyclonal feeder expansion. (a) Percentage of cells with a TN (CD45RA+ CD62L+ CXCR3− CD95−), TSCM (CD45RA+ CD62L+ CXCR3+ CD95+), TCM (CD45RA− CD62L+ CXCR3+ CD95+), TEM (CD45RA− CD62L− CXCR3− CD95+) and TTE (CD45RA+ CD62L− CXCR3− CD95+) phenotype before agonist peptide stimulation, and after 1, 2 or 3 rounds of agonist peptide stimulation. (b) Percentage of cells with a TN, TSCM, TCM, TEM, and TTE phenotype after polyclonal feeder expansion. For (a) and (b) T-cells were generated from HSPC from healthy donors (n = 3), patients in remission (n = 6) and AML patients at diagnosis (n = 3). Gating on eGFP+ TCR-transduced cells. Mean and s.d. are shown. (c) Percentage of cells positive for PD-1 expression after polyclonal feeder expansion. T-cells were generated from HSPC from healthy donors (n = 8), patients in remission after chemotherapy (n = 9) and AML patients at diagnosis (n = 6). Gating on eGFP+ TCR-transduced cells. Individual samples and mean ± s.d. are shown. Mann–Whitney U test was used to assess statistical significance. P-value < 0.05 (*)

Figure 5. In vitro generated TA-specific T-cells specifically recognize and kill TA-expressing cell lines. (a) Intracellular staining of T-cells for interferon-gamma (IFNg) after co-culture with THP-1 (HLA-A2+ WT1+) or JY (HLA-A2+ WT1) cells. Culture medium was used as a negative control, stimulation with aCD3/aCD28 as a positive control. Effector/target ratio 1/2. Gating on eGFP+ TCR-transduced cells. T-cells generated from HSPC from healthy donors (n = 4), patients in remission after chemotherapy (n = 5), AML patients at diagnosis (n = 4) and PBL (n = 6). (b) Percentage specific lysis determined via 4-h 51chromium release assay after co-culture of T-cells with T2 cells pulsed with relevant WT1 or irrelevant influenza (INF) peptide (10 µg/ml), HL-60-A2 (HLA-A2+ WT1+), THP-1, or JY cells. Effector/target ratio 10/1. T-cells generated from HSPC from healthy donors (n = 6), patients in remission after chemotherapy (n = 7), AML patients at diagnosis (n = 8) and PBL (n = 6). For (a) and (b) mean and s.d. are shown. Kruskal–Wallis test was used to determine statistical significance between different HSPC sample groups. Differences were not significant. Mann–Whitney U test was used to determine statistical significance for between-group comparisons for HSPC- and PBL-derived T-cells. P-value < 0.05 (*) and P < 0.01 (**)

Figure 5. In vitro generated TA-specific T-cells specifically recognize and kill TA-expressing cell lines. (a) Intracellular staining of T-cells for interferon-gamma (IFNg) after co-culture with THP-1 (HLA-A2+ WT1+) or JY (HLA-A2+ WT1−) cells. Culture medium was used as a negative control, stimulation with aCD3/aCD28 as a positive control. Effector/target ratio 1/2. Gating on eGFP+ TCR-transduced cells. T-cells generated from HSPC from healthy donors (n = 4), patients in remission after chemotherapy (n = 5), AML patients at diagnosis (n = 4) and PBL (n = 6). (b) Percentage specific lysis determined via 4-h 51chromium release assay after co-culture of T-cells with T2 cells pulsed with relevant WT1 or irrelevant influenza (INF) peptide (10 µg/ml), HL-60-A2 (HLA-A2+ WT1+), THP-1, or JY cells. Effector/target ratio 10/1. T-cells generated from HSPC from healthy donors (n = 6), patients in remission after chemotherapy (n = 7), AML patients at diagnosis (n = 8) and PBL (n = 6). For (a) and (b) mean and s.d. are shown. Kruskal–Wallis test was used to determine statistical significance between different HSPC sample groups. Differences were not significant. Mann–Whitney U test was used to determine statistical significance for between-group comparisons for HSPC- and PBL-derived T-cells. P-value < 0.05 (*) and P < 0.01 (**)

Figure 6. Cryopreservation has a negligible effect on maturation kinetics and the functionality of in vitro generated TA-specific T-cells. (a) Relative cell numbers at day 14 of co-cultures (day at which cord blood progenitors were transduced) from fresh and cryopreserved (frozen) HSPC from cord blood (n = 6), healthy donors (n = 6), patients in remission after chemotherapy (n = 7) and AML patients at diagnosis (n = 6). Individual fresh and paired frozen samples are shown. (b) Percentage specific lysis determined via 4-h 51chromium release assay after co-culture of T-cells with T2 cells pulsed with relevant WT1 or irrelevant influenza (INF) peptide (10 µg/ml), HL-60-A2 (HLA-A2+ WT1+), THP-1 (HLA-A2+ WT1+) or JY (HLA-A2+ WT1) cells. Effector/target ratio 5/1. T-cells generated from fresh and cryopreserved (frozen) HSPC (n = 11). Results from different sample groups (healthy donors, patients in remission and AML patients at diagnosis) were pooled. Mean and s.d. are shown. Wilcoxon matched-pairs signed-rank test was used to assess statistical significance. Differences were not significant

Figure 6. Cryopreservation has a negligible effect on maturation kinetics and the functionality of in vitro generated TA-specific T-cells. (a) Relative cell numbers at day 14 of co-cultures (day at which cord blood progenitors were transduced) from fresh and cryopreserved (frozen) HSPC from cord blood (n = 6), healthy donors (n = 6), patients in remission after chemotherapy (n = 7) and AML patients at diagnosis (n = 6). Individual fresh and paired frozen samples are shown. (b) Percentage specific lysis determined via 4-h 51chromium release assay after co-culture of T-cells with T2 cells pulsed with relevant WT1 or irrelevant influenza (INF) peptide (10 µg/ml), HL-60-A2 (HLA-A2+ WT1+), THP-1 (HLA-A2+ WT1+) or JY (HLA-A2+ WT1−) cells. Effector/target ratio 5/1. T-cells generated from fresh and cryopreserved (frozen) HSPC (n = 11). Results from different sample groups (healthy donors, patients in remission and AML patients at diagnosis) were pooled. Mean and s.d. are shown. Wilcoxon matched-pairs signed-rank test was used to assess statistical significance. Differences were not significant

Figure 7. Functional TA-specific T-cells can also be generated from HSPC from HLA-A2 negative donors. Percentage specific lysis determined via 4-h 51chromium release assay after co-culture of T-cells with T2 cells pulsed with relevant WT1 or irrelevant influenza (INF) peptide (10 µg/ml), HL-60-A2 (HLA-A2+ WT1+), THP-1 (HLA-A2+ WT1+) or JY (HLA-A2+ WT1) cells. Effector/target ratio 5/1. Mean and s.d. are shown. T-cells generated from HLA-A2+ (n = 5) and HLA-A2 (n = 4) HSPC. Results from different sample groups (healthy donors, patients in remission and AML patients at diagnosis) were pooled. Mann-Whitney U test was used to assess statistical significance. Differences were not significant

Figure 7. Functional TA-specific T-cells can also be generated from HSPC from HLA-A2 negative donors. Percentage specific lysis determined via 4-h 51chromium release assay after co-culture of T-cells with T2 cells pulsed with relevant WT1 or irrelevant influenza (INF) peptide (10 µg/ml), HL-60-A2 (HLA-A2+ WT1+), THP-1 (HLA-A2+ WT1+) or JY (HLA-A2+ WT1−) cells. Effector/target ratio 5/1. Mean and s.d. are shown. T-cells generated from HLA-A2+ (n = 5) and HLA-A2− (n = 4) HSPC. Results from different sample groups (healthy donors, patients in remission and AML patients at diagnosis) were pooled. Mann-Whitney U test was used to assess statistical significance. Differences were not significant
Supplemental material

Supplemental Material

Download ()

Supplemental Material

Download ()

Supplemental Material

Download ()