TRE sequence motifs
T cells relevant TRE sequence motifs were identified by first searching the TRED30 and TRANSFAC31 database for T cell-centric transcription factors. Eleven transcription factors were present in both databases. For each transcription factor, we obtained the corresponding TRE sequence from the JASPAR32 database of transcription factor binding profiles.
Synthetic promoter library design
TRE-specific and NruI- or XbaI-containing sense and anti-sense 5’ phosphorylated oligonucleotides were resuspended in 100 µM in STE buffer (100 mM NaCl, 50 mM Tris–HCl, 1 mM EDTA, pH 7.8, Sigma Aldrich). Equal volumes of sense and anti-sense oligos were mixed, incubated at 95 oC for 3 min, and gradually cooled down to room temperature. The synthetic promoter library was constructed by ligating these blocks at appropriate stoichiometric molar ratios (6x of NF-κB TRE block, 2x of each other TRE, 1x NruI block, and 1x XbaI) with high concentration T4 DNA ligase (Thermo Fisher).
Resultant fragments ranging from 50 to 500 bp were gel extracted and ligated into NruI:XbaI digested HIV lentiviral backbone,54 upstream of IL2mp and GFP:ffluc. The ligation product was used to transform dam-/dcm- competent E. coli (NEB). A sample of the transformed cells was transferred to a kanamycin agarose plate and the remaining cells cultured in 50 ml LB + kanamycin (50 µg/mL) medium. DNA was extracted using Qiagen Plasmid Midi kit (Qiagen).
Construct design
All studies involving lentivirus featured the FMC63 CD19CAR cloned into an HIV backbone55. PiggyBac-transposon constructs were designed with a human elongation factor 1 alpha (EF1a) promoter driving expression of five-part polycistron: 1) human G01S anti-CD19CAR56, 2) P2A ribosomal skip sequence57, 3) double mutant of the dihydrofolate reductase (DHFRdm) for drug selection58, 4) T2A ribosomal skip sequence, and 5) a truncated epidermal growth factor receptor (EGFRt) to serve as cell surface marker59. iSynPro-regulated transgene sequences, including GFP:ffluc, TurboGFP, PD1:MYD88,; LASI and Marker Only control55, were introduced into piggyBac constructs in forward or reverse orientation upstream of the EF1a-driven cassette. All constructs were assembled from synthesized DNA fragments using standard molecular biology techniques.
Lentivirus production and titering
Lentivirus was produced by co-transfecting the HIV7 transfer plasmid and vectors pCHGP-2, pCMV-Rev2 and pCMV-G encoding packaging proteins (Rev, Gag, Pol, VSVG) in HEK293T cells using Lipofectamine 2000 (Invitrogen). Viral supernatants were harvested 72h after transfection, filtered through a 0.45 µM filter, and centrifuged at 24,500 rpm. The virus pellets were resuspended in serum free medium and stored at -80 oC. Lentiviruses were titrated in Jurkat cells by flow cytometry analysis of the marker expression (e.g. EGFRt, GFP, etc.) and Lenti-X p24 Rapid Titer Kit (Clontech).
Cell line culture
HEK293T cells were cultured in DMEM (Invitrogen) supplemented with 10% FBS (Hyclone), 2.5% HEPES (Invitrogen), 1% L-Glutamine (Invitrogen), and 1% sodium pyruvate (Invitrogen). SK-N-Be2 (also referred to as Be2) cells were cultured in DMEM supplemented with 10% FBS and 1% L-Glutamine. Jurkat, Tm-LCL, Raji, K562, DHL-4 and SupB15 suspension cells were cultured in RPMI (Invitrogen) supplemented with 10% FBS and 1% L-Glutamine.
Primary T cell isolation and culture
Human peripheral blood mononuclear cells (PBMCs) derived from blood discard kits of healthy donors (BloodworksNW) were isolated by Ficoll-Paque (Pharmacia Biotech). CD4+ and CD8+ cells were purified from the PBMC using RoboSep (Stemcell). In some instance, the CD4-, CD8- PBMC fraction was retained. CD4+ T cells were cultured in RPMI with 10% FBS, 5 ng/ml rhIL-7 (Miltenyi) and 0.5 ng/ml rhIL-15 (Miltenyi). CD8+ cells were cultured in RPMI with 10% FBS, 50 U/ml rhIL-2, (Chiron Corporation) and rhIL-15 (0.5 ng/ml). In some instances, CD4+ or CD8+ cells were cultured in 50 U/mL rhIL-2, 20 ng/mL IL-4 (Milteyni), 10 ng/mL IL-7 (Milteyni) and 20 ng/mL Il-21 (Milteyni). Medium was replaced twice a week.
T cell transduction and expansion
CD4+ or CD8+ T cells were resuspended to 2 to 4 x 106 cells / mL in RPMI medium supplemented with cytokines as above and stimulated with Dynabeads™ Human T Activator CD3/CD28 (Invitrogen) at 1:1 ratio overnight. The activated T cells were added with protamine sulfate (40 ug/ml, APP Pharmaceuticals)) and then transferred into a 12-well plate. Viruses were thawed, vortexed and added to each corresponding well. For co-transduction, both viruses were added to the cells at certain ratios. For example, in iSynPro library screening, CD19CAR-T2A-EGFRt virus and the iSynPro-IL2mp-GFP:ffluc library virus were co-transduced at MOI of 2 and 0.1 respectively. The plate was then transferred to 37oC after spinoculation for 30 minutes at 800 x g. Equal volume of warm complete RPMI media supplemented with cytokines was added to each well at 4 hours or overnight post transduction. Six days post transduction, CD3/CD28 beads were removed from the cells and the transduction efficiency was checked by flow cytometry analysis of cell-surface marker or GFP expression.
For T cells transduced with CD19CAR-T2A-EGFRt-T2A-DHFRdm, methotrexate (MTX) was used to select transgene-containing cells. MTX was added to the T cell culture medium at 50 µM 6 days after transduction and supplemented when medium was changed. Transduced T-cells were expanded by stimulation with irradiated (8000 rad) TM-LCL at a 1:7 E:T ratio in the presence of 50U/mL IL-2 (CD8+), 5ng/mL IL-7 (CD4+) and 1ng/ml IL-15. The in vitro and in vivo assays were performed 10 days after T cell expansion.
T cell nucleofection and expansion
CD4+ or CD8+ cells were nucleofected using 10 nM donor plasmid and 10 nM RNA (encoding for piggyBac transposase) with the Lonza 4D system using pulse program EO-115. Nucleofected CD4+ or CD8+ cells were transferred to a G-REX culture vessel pre-seeded with a CD4-/CD8- PBMC fraction in X-Vivo 15 (Lonza) supplemented with 2% KnockOut serum replacement (ThermoFisher), 80 U/mL IL-2 (StemCell), 20 ng/mL IL-4 (Miltenyi), 10 ng/mL IL-7 (Miltenyi) and 20 ng/mL IL-21 (Miltenyi). Media was replaced twice weekly and cell expanded for 21 days.
Sorting of CD8/iSynPro-GFP/CAR T cells using FACS
Eight days after CD19CAR and iSynPro-GFP:ffluc co-transduction, CD8+ T cells were sorted for EGFRt+GFP- population by FACS to exclude cells containing constitutively expressing synthetic promoters.
Cells were resuspended in PBS/10% FBS staining solution at 1 x 106/mL, filtered (30 µm filter) and stained with APC conjugated anti-EGFRt (APC-cetuximab, custom made by BD Biosciences). Cells were washed with the staining solution, resuspended at 1x 106/ml for cell sorting (FACSJazzTM, BD Biosciences). The sorted EGFRt+GFP- cells were pelleted, pooled and seeded in RPMI supplemented with IL-2/IL-15 and 1X Penicillin /Streptomycin (Gibco).
Four days later, these T cells were washed with RPMI medium to remove the cytokines and co-cultured with irradiated Tm-LCL cells at 1:2 ratio. The cell suspension was harvested separately at 24, 48 and 72h after co-culturing, resuspended in 1X PBS+10%FBS and filtered (30 µm filter). The GFP positive population was sorted out by FACS. The sorted cells were centrifuged down and stored at -80oC until DNA extraction.
NGS library preparation
Libraries were generated from DNA samples prepared from the plasmid, virus and GFP sorted cell populations (Fig. 1b and 2a) for sequencing on the Illumina Miseq platform as follows. DNA was extracted using QIAamp DNA Micro Kit (Qiagen #56304) and concentration measured by Qubit DNA HS assay (Thermo Fisher Scientific #Q32851). A first round PCR was performed for targeted amplification of the synthetic promoter library (refer sequence schema below). First round PCR primers comprised of (1) a locus-specific region (letters in ‘bold red’) that matched invariant bases immediately adjacent to the 5’ and 3’ ends of the variable synthetic promoter region (letters in italics), and (2) a linker oligo (green overhang) to permit subsequent conjugation of Illumina adapter sequences. For efficient library recovery, a touchdown PCR protocol was employed with KAPA HiFi HotStart Ready Mix (KAPA Biosystems #KK2601). Each reaction used 100 ng of DNA template based on our PCR optimization result. Samples with material in excess of 100 ng were split into several PCR reactions. Resulting products were combined and purified by Agencourt AMPure XP (Beckman Coulter #A63880).
From 15ng of each first round PCR reaction, dual index amplicon libraries were generated by adding Illumina Index-Adapter sequences to the DNA templates. This was accomplished by a second round PCR with KAPA HiFi HotStart Ready Mix (KAPA Biosystems #KK2601) using Illumina Nextera-XT Index primers that incorporate linker oligos at their 3’ end (Illumina #FC-131-1001). The shared linker oligo between primers of both rounds of PCR facilitate conjugation of the Index-Adapters to the DNA templates. Completed libraries were purified with Agencourt AMPure XP beads (Beckman Coulter #A63880) and quantified using Qubit DNA HS assay (Thermo Fisher Scientific #Q32851). Separately, a PCR-Free amplicon library was prepared from 100ng of purified DNA sourced from the raw ligation product with the Accel-NGS 2S DNA Library Kit (IDT #10009877). This PCR-Free library was quantified by qPCR using the KAPA Library Quant Kit (Roche Diagnostics #KK4824). Following each PCR round, amplicon libraries were quality assessed by Agilent’s 2200 TapeStation D5000 assay (Agilent #5067-5588).
Sequencing
All sequencing was performed on an Illumina MiSeq with MiSeq Reporter v2.5.1 (Illumina), using Illumina’s v3 reagents (#MS-102-3003) and following manufacturer’s instructions. Our libraries featured amplicons with stretches of monotemplate sequence (~30 bases) at both ends juxtaposing variable synthetic promoter sequences in between – this is problematic for Illumina sequencers and can result in poor quality reads. As a diversity countermeasure, 10% PhiX Control v3 Library (Illumina FC-110-3001) was supplemented for sequencing. From this attempt it was evident significantly higher PhiX levels would be needed to improve read quality, in turn sacrificing usable read throughput. This prompted a search for custom Read 1 and 2 primers that would bypass the monotemplate sequence in our libraries. We identified a suitable candidate for use as Read 1 primer (refer previous sequence schema) but were unable to design a custom Read 2 primer counterpart. For Read 2, we relied on the Illumina supplied sequencing primer mix, that primes off the 3’ linker. Illumina sequencer color matrix calibrations that are based on the initial bases of Read 1 are of primary importance – thus we proceeded with the custom Read 1 primer (GCGTTTTGCGCTGCTTCGCGATCGA) and to work in conjunction with it, a custom PhiX Read 1 Primer (ACACTCTTTCCCTACACGACGCTCTTCCGATCT). We spiked 10% PhiX Control to support improved quality for Read 2. Such an approach greatly improved the overall quality of both Read 1 and Read 2. The raw library templates which lacked terminal monotemplate bases representative of plasmid backbone sequences and were directly ligated to Illumina adapters, were accordingly sequenced with the default Illumina sequencing primers. PhiX was added as a quality control for cluster generation, sequencing, alignment and matrix calibration. Sequencing configuration was 300 bp paired end. Read recovery is listed in Supplementary Table 3.
Transient and repeated stimulation of CD19CAR+ iSynPros-GFP:ffluc T cells
For repeated in vitro stimulation, T cells were stimulated with CD3/CD28 Dynabeads overnight and co-transduced with CD19CAR and iSynPros-GFP:ffluc. On day 7, beads were removed, and a sample of the cells was analyzed for EGFRt and GFP expression by flow cytometry. The remainder of the cells were continuously cultured for 7 days, stimulated with irradiated (8000 rad) TM-LCL at 1:2 ratio for 16 to 18 hours and analyzed by flow cytometry. This procedure was repeated two more times on 14-day intervals.
In vitro Incucyte assay
Incucyte (Sartorius) live imaging was used to track the cytotoxicity, proliferation, and iSynPro1 driven GFP expression CAR T cells. CD19t+/mCherry+ Be2 tumor cells and CAR T cells were co-cultured in triplicates in 384-well imaging plates. The number of tumor cells and CAR T cells that were seeded varied depending on the E:T ratios of interest. For the serial re-challenge experiment, subsequent doses of Be2 tumor cells were added every 3 days for a total 9 days. Images were captured every 4-hours at 10x or 20x magnification. IncuCyte 2020C software was used for analysis. Cytotoxicity of CAR T cells, represented by the target cell presence, was quantified by RCU. GFP expression upon iSynPro1 induction was quantified by GCU. Cytotoxicity and iSynPro1 induction were assessed using the Basic Analyzer analysis. Proliferation of CAR T cells was quantified using Non-Adherent Cell-by-Cell analysis.
Dasatinib assay
CD4+ and CD8+ CAR T cells expressing iSynPro1-GFP:ffluc were co-cultured with unmodified, OKT3+, or CD19t+ K562 target cells at a 1:1 E:T ratio, or stimulated with PMA/Ionomycin, with or without Dasatinib at 60 nM. After 16 hours, cells were harvested and analyzed by flow cytometry.
Co-stimulation assay
CD4+ and CD8+ CAR T cells expressing iSynPro1-GFP:ffluc were left untreated or co-cultured with unmodified, CD19t+, CD80+CD86+, or CD19t+CD80+CD86+ K562 target cells at a 1:1 E:T ratio. After 16 hours, cells were harvested and analyzed by flow cytometry.
Jurkat reporter assay
Jurkat reporter cell lines were generated via lentiviral transduction to contain either 7x NF-κB, 6x NFAT, 7x GAS or iSynPro1 driving GFP expression. Cells were cultured in a base media of RPMI supplemented with 10% FBS and 1% glutamine and cell stimulants, TNFα 50 ng/mL (Bio-Techne), IFNγ 50 ng/mL (Bio-Techne) and PMA/Ionomycin 1x (Thermo Fisher), and TLR5 agonist, flagellin 1.25 ug/mL (InvivoGen) for 16 hours at 37°C. 7xNF-κB served as a positive control for TNFα, and 7xGAS for IFNγ. After the stimulation period, cells were harvested and analyzed by flow cytometry for GFP expression. As the PMA/Ionomycin stimulated condition presented the maximal GFP expression for all Jurkat lines, relative percent induction was calculated by dividing the GFP positive percentage for each stimulation condition by the GFP positive percentage induced by PMA/Ionomycin stimulation in each cell line.
RNA kinetics study
Cell pellets were collected and spun at 300 X g for 7 minutes, media aspirated and frozen at -80 oC. Pellets were processed for RNA using a RNeasy Mini kit (Qiagen), cDNA was synthesized using a SuperScript™ IV Reverse Transcriptase (ThermoFisher), and cDNA was quantified using the QX-200 droplet digital PCR system (Bio-Rad) according to the manufacturer’s recommendations. Primer and probes sets are detailed in Supplemental Table 5.
RNA-seq
PD1:MyD88 or GFP expressing CD19CAR CD4+ or CD8+ T cells were subjected to CD19t and mCherry expressing Be2 cells for 24 hours at a 2:1 effector to target ratio. Following 24h, effectors were subjected to another bolus of Be2 targets and, after an additional 24 hours, effectors and targets removed from culture, labeled with live dead stain and anti-CD4, anti CD8, anti-CD3, Herceptin, and Erbitux antibodies. One million live CD4+ and CD8+ cell populations were sorted using a Sony MA900 cell sorter, and cell pellets were frozen at -80° C in preparation for RNAseq.
Cell pellets were shipped on dry ice to Genewiz for library preparation and sequencing. RNAseq libraries were pooled and sequenced to generate 25M 150bp X 2 reads per sample (Illumina). RNA-seq data was analyzed using the nf-core RNA-seq pipeline version 3.4, code available from nf-core/rnaseq (https://nf-co.re/rnaseq/3.12.0). Briefly, reads were mapped to the human genome (GRCh38) using STAR aligner (v2.6.1d). Gene expression counts were generated using salmon (v1.5.2). Differential gene expression analysis between two phenotypes (iSynPro1-PD1:MYD88 and iSynPro1-GFP) was performed using edgeR version 3.32.157.57. First, genes whose expression was less than 10 counts in 2 samples were discarded from further analyses. Across samples, normalization was then performed using trimmed mean of M-values, via the function calcNormFactors. Next, estimateGLMCommonDisp and estimateGLMTrendedDisp were run to properly handle over-dispersion at the global and single gene level. Differentially expressed genes were determined using the glmQLFit function, as those showing a two-sided raw p value <0.01 and |log2(fold change)| >=1.
In vivo studies
All mouse experiments were approved by the SCRI (Seattle Children’s Research Institute) Animal Care and Use Committee. NOD/Scid IL2RγCnull mice were obtained from The Jackson Laboratory or bred in-house.
Subcutaneous Single-Challenge Model: 9- to 13-week old NOD/Scid IL2RγCnull (NSG) mice were injected subcutaneously (SC) with 5 X 106 Be2 or Be2/CD19 cells on day 0. Fourteen days later, mice were intravenously (IV) injected with 2.5 x 106 of CD8+/iSynPro1-GFP:ffluc/CD19CAR.
Subcutaneous Double-Challenge Model: 9- to 13-week old NSG mice were injected SC on the right flank with 5 x 106 Be2/CD19 cells on day 0. Six days later, mice were injected IV with 2.5 x 106 of T cells and monitored thereafter for tumor progression. At any point, mice with tumors that exceeded 1500 mm3 were euthanized. On day 55, surviving mice were re-challenged with 5 x 106 Be2/CD19 on the left flank via a SC injection. Mice were thereafter monitored for tumor progression until day 110.
Intraperitoneal Raji Model: 10- to 11-week old female NSG mice were injected intraperitoneally (i.p.) with 3 x 106 or 5 x 106 CD8+/iSynPro1-GFP:ffluc/CD19CAR cells. 500,000 live or 10 x 106 irradiated Raji (CD19+) tumor cells were i.p. injected 15 to 17 days later. The multiple doses of irradiated Raji cells were given through i.p. injections for the rechallenging study.
Bioluminescent imaging was performed weekly by (i.p.) injection of 4.29 mg/mouse D-luciferin (Xenogen), anesthesia by isoflurane and imaging 10 minutes post D-luciferin injection using the IVIS Spectrum Imaging System (Perkin Elmer). Luciferase activity was analyzed using Living Image Software Version 4.3 (Perkin Elmer) and photon flux was analyzed within regions of interest.
Systemic Nalm-6 Model: 11-13 week old NSG mice were injected with one million human Nalm-6 leukemia cells via the tail vein, modified to express a fusion protein of mCherry and firefly luciferase. Six days after tumor injection, tumor engraftment in each mouse was quantified via bioluminescent imaging as described above. Mice were then assigned to treatment groups to equalize average tumor engraftment across groups. Seven days after tumor injection, mice were systemically injected via the tail vain with two or four million T cells. Thereafter, mice were monitored for tumor progression by bioluminescent imaging. Mice were euthanized when they showed moderate to severe hind-limb paralysis, an effect of leukemia progression, or otherwise as recommended by veterinary staff. Retro-orbital blood samples were taken on a weekly basis to track T cell engraftment in the peripheral blood.
T Cell Tracking by Retro-Orbital Bleeds. Beginning at day 17 after tumor injection, peripheral blood retro-orbital bleeds of mice were taken on a weekly basis, and flow cytometry was performed on blood samples to quantify T cell engraftment. First, samples were subjected to red blood cell lysis using Pharm Lyse Buffer (BD, Cat. # 555899) and treated with Fc blocking reagent (Miltenyi, Cat. # 130-059-901) to prevent indiscriminate antibody binding. Next, cells were stained with the following panel of reagents: anti-CD3, anti-human CD45, anti-mouse CD45, and fixable viability stain 520 to discriminate live human T cells. Finally, samples were fixed in 0.5% paraformaldehyde (Electron Microscopy Sciences, Cat. # 15713) in PBS before analysis. CountBright absolute counting beads (Invitrogen, Cat. # 2207530) were then added to each sample to allow for calculation of T cell concentrations in analyzed blood.
Statistical analyses
Data wrangling and visualization were conducted using Prism (GraphPad software), R (www.r-project.com) and RStudio (www.rstudio.com). The number of replicates or T cell donors are indicated in figure legends. Comparisons of means between two groups was conducted using two-tailed t-tests. Enrichment of TREs within promoter clusters was evaluated using one-sided (alternative=”greater") Fisher’s test (see Supplemental Methods for additional details). Survival comparisons following adoptive transfer of tumor xenografts used log-rank (Mantel-Cox) tests. The number of mice are indicated in the figure legends. For all tests, p < 0.05 was considered significant and was corrected for multiple comparisons. Exact values of test statistics, degrees of freedom, variance assumptions, and power are listed in Supplementary Table 5.