Animals
NOD/SCID/γc−/− (NSG) and NOD.Cg-Prkdcscid Il2rgtm1Wjl Tg(CMV-IL3,CSF2,KITLG)1Eav/MloySzJ (NSG-SGM3) mice were obtained from the Jackson Laboratory, housed and bred at the Fred Hutchinson Cancer Research Center (FHCRC) (Seattle, WA). For all experiments, 6–10-week-old age-matched females were randomly assigned to experimental groups. Mice transplanted with engineered CB or AML cell lines were monitored and euthanized when they exhibited symptomatic leukemia (tachypnea, hunchback, persistent weight loss, fatigue or hind-limb paralysis). Experiments were performed after approval by Institutional Animal Care and Use Committee (protocol #51068) and in accordance with institutional and national guidelines and regulations.
Primary Specimens
Human umbilical cord blood samples were obtained from normal deliveries at Swedish Medical Center (Seattle, WA). Frozen aliquots of AML diagnostic bone marrow samples were obtained from the Children’s Oncology Group. Cells were thawed in IMDM supplemented with 20% FBS and 100 U/mL DNaseI (Sigma, Cat#D5025). A bone marrow biopsy from a C/G patient was obtained from a patient treated at the University of Minnesota Masonic Children's Hospital. Healthy donor T cells were obtained from Bloodworks Northwest (Seattle, WA). We confirmed these cells lacked infections agents (EBV, HCMV, Hepatitis A, Hepatitis B, Hepatitis C, HHV 6, HHV 8, HIV1, HIV2, HPV16, HPV18, HSV1, HSV2, HTLV 1, HTLV 2, and Mycoplasma sp) through IDEXX Bioanalytics (West Sacramento, CA). All specimens used in this study were obtained after written content from patients and donors. The research was performed after approval by the FHCRC Institutional Review Board (protocol #9950). The study was conducted in accordance with the Declaration of Helsinki.
Cell lines
M07e (DSMZ, Cat# ACC104), WSU-AML (BioIVT, Cat# HCL-WSUAML-AC), Kasumi-1 (ATCC, Cat# CRL-2724) cell lines were maintained per manufacturer’s instructions. We engineered Kasumi-1 FOLR1+ cell line by transducing Kasumi-1 cells with a lentivirus containing the FOLR1 transgene driven by the EF1a promoter (Genecopoeia, Cat# LPP-C0250-Lv156-050). Jurkat Nur77 reporter cells25 were maintained in RPMI supplemented with 20% FBS and 2 mM L-Glutamine.
Constructs and Lentivirus production
The MSCV-CBFA2T3-GLIS2-IRES-mCherry construct was a gift from Dr. Tanja Gruber (Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tn, Ref2). The C/G fusion gene from this construct and the MND promoter were inserted into pRRLhPGK-GFP lentivirus vector26 as described in Ref.4.
CAR constructs containing IgG4 short, intermediate and long spacers are previously described in Ref.27. The VL and VH sequences from Farletuzumab were used to construct the anti-FOLR1 scFv with VL/VH orientation using G4SX4 linker. Anti-FOLR1 scFv DNA fragment was human codon optimized and synthesized by IDT gBlock gene fragment and cloned into the CAR vectors with NheI and RsrII restriction sites upstream of the IgG4 spacer.
Farletuzumab scFv:
DIQLTQSPSSLSASVGDRVTITCSVSSSISSNNLHWYQQKPGKAPKPWIYGTSNLASGVPSRFSGSGSGTDYTFTISSLQPEDIATYYCQQWSS
YPYMYTFGQGTKVEIKGGGGSGGGGSGGGGSGGGGSEVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISS
GGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSS
Lentivirus particles were produced in 293T cells (ATCC, Cat#CRL-3216). 293T cells were transfected with transfer vector, viral packaging vector (psPAX2), and viral envelope vector (pMD2G) at 4:2:1 ratio using Mirus 293Trans-IT transfection agent (Mirus, Cat# MIR2700) as directed by manufacturer’s protocol. Viral particles were collected each day for 4 days post transfection, filtered through 0.45 μm membrane (Thermo Fisher; Cat NAL-166-0045) and concentrated (overnight spin at 4ºC, 5000rpm) before use.
Transduction of cord blood CD34+ cells
CB samples were processed with red blood cell lysis buffer and enriched for CD34+ cells using CliniMACS CD34 MicroBeads (Miltenyi Biotec, Cat# 130-017-501). CB CD34+ cells were then seeded onto retronectin (5 ug/mL, Takara, Cat#T100A) + Notch ligand Delta1 (2.5 ug/mL, Ref28) coated plates overnight in SFEM II medium (StemCell Technologies, Cat# 09650FH) containing 50ng/mL stem cell factor (SCF, StemCell Technologies, Cat# 78062), 50ng/mL thrombopoietin (TPO, StemCell Technologies, Cat# 78210) and 50ng/mL Fms-like tyrosine kinase 3 ligand (FLT3L, StemCell Technologies, Cat# 78009). Cells were transduced the following day with the C/G construct at an MOI of 200 or GFP control construct at MOI of 50. Transduced cells were grown on Notch ligand at 37oC in 5% CO2 for 6 days then sorted for GFP+ cells. Sorted GFP+ cells were either transplanted into NSG-SGM3 mice at 200,000 cells per mouse or placed in EC co-culture or myeloid promoting condition (MC, see Ref.16 and below) for long term culture at 75,000 cells per 6-well. In a subsequent experiment using a CB CD34+ sample from another donor (CB 2, see Extended Fig. 4), transduced cells were grown on Notch ligand for 2 days prior to placement in EC co-culture or MC plating at 100,000 cells per 12-well.
Long term culture of transduced cord blood CD34+ cells
Transduced cells were placed in either EC co-culture with SFEM II medium supplemented with 50ng/mL SCF, 50ng/mL TPO, 50ng/mL FLT3L, and 100U/mL Pen/Strep, or MC containing Iscove’s Modified Dulbecco’s Medium (IMDM, Gibco 12-440-053) supplemented with 15% fetal bovine serum (FBS, Corning, 35-010-CV), 100U/mL Penicillin-Streptomycin (Pen/Strep, Gibco, 15-140-122), 10ng/mL SCF, 10ng/mL TPO, 10ng/mL FLT3L, 10ng/mL IL-6 (Shenandoah Biotechnology, Cat#100-10), and 10ng/mL IL3 (Shenandoah, Cat#100-80). For EC co-cultures, human umbilical vein endothelial cells (HUVECs) transduced with E4ORF1 construct (E4 ECs) were propagated as previously described13,29. One day prior to co-culture, E4 ECs were seeded into 6-well or 12-well plates at 800,000 or 300,000 cells per well, respectively, and cultured in medium 199 (Biowhittaker #12-117Q) supplemented with FBS (20%, Hyclone, Cat#SH30088.03), endothelial mitogen (Biomedical Technologies, Cat#BT203), Heparin (Sigma, Cat# H3149), HEPES (Gibco, Cat# 15630080), L-Glutamine (Gibco, Cat# 25030), and Pen/Strep. After 24 hours, E4 ECs were washed with PBS and cultured with transduced CB cells in media described above. Transduced CB cells in either culture condition were propagated with fresh media and E4 ECs replaced every week until cells stopped proliferating. Three-to-twenty percent of the cultures were re-plated each week for long-term culture.
We confirmed C/G and FOLR1 expression in engineered cells over weeks in culture using RT-PCR (Supplemental Fig. 3). Tranduced CB cells were sorted for GFP+ cells on an FACSAria II using FACSDiva Software (BD Biosciences). DNA and RNA from sorted cells were extracted with AllPrep DNA/RNA/miRNA Universal Kit using the QIAcube platform (QIAGEN). Expression of the fusion transcript in GFP+ cells was confirmed by RT-qPCR TaqMan assay and QuantStudio 5 real-time PCR system using the primers: Forward 5-CCCTGACGGTCATCAACCA-3, Reverse 5-CACCATCCAAATAGCGCAGTG-3, and TaqMan probe 5-[FAM]-CAGCGAGGACTTCCAG-[MGB]-3. FOLR1 expression was determined using RT-qPCR TaqMan assay (Hs01124177_m1, cat# 4331182).
Cell surface analysis
For xenograft CB cells, mouse bone marrow, peripheral blood, spleen, and liver were harvested at necropsy and processed with red blood cell lysis buffer. Spleen and liver were processed into cell suspension with glass slides and passed through a 70-um cell strainer. CB cells in EC co-culture and MC were harvested after vigorously pipetting to resuspend CB cells. CB cells from processed mouse tissues and cultures were washed in 2% FBS in PBS, blocked with 2% human AB serum in PBS, then stained with a cocktail of fluorescently labeled monoclonal antibodies for 20 min on ice (see Supplemental Information for antibodies used). Labeled cells were washed with PBS and resuspended in 2% FBS/PBS prior to flow cytometric analysis. FACSymphony equipped with FACSDiva Software (BD Biosciences) was used to assess cell surface expressions and FlowJo Software was used for the analysis. Dead cells were excluded based on LIVE/DEADTM Fixable Violet Dead Cell Stain (FVD, Invitrogen, cat# L34955). For EC co-cultures, ECs were excluded by gating on CD45+ cells or CD45+CD144- cells.
A fraction of the C/G-CB cells isolated from xenograft models or cultured in EC co-culture or MC at various timepoints were sent to Hematologics, Inc (Seattle, WA) for assessment of the RAM immunophenotype along with C/G patient samples.
Histology and immunocytochemistry
Sample tissues were fixed in 10% formalin, processed into paraffin sections and stained with hematoxylin and eosin (H&E). Immunohistochemistry was performed using antibodies to ERG (EP111; Cell Marque) and CD56 (MRQ-42; Cell Marque) following citrate pretreatment and visualized with 3, 3'-diaminobenzidine (DAB) on a Ventana Bench Mark Ultra.
All tissues were examined by a board certified Hematopathologist (KRL). The bone marrow core biopsy specimen was fixed in acetic acid-zinc-formalin (AZF), decalcified, and embedded in paraffin, and sections were stained for CD56 (clone MRQ-42; Cell Marque, Rockin, California).
RNA-seq analysis
RNA-sequencing Library Construction. Total RNA was extracted using the QIAcube automated system with AllPrep DNA/RNA/miRNA Universal Kits (QIAGEN, Valencia, CA, #80224) for diagnostic pediatric AML samples from peripheral blood or bone marrow, as well as, bulk healthy bone marrows, and healthy CD34+ peripheral blood samples. Total RNA from C/G-CB and GFP-CB cells in EC co-culture and MC at indicated timepoints was purified as described above. The 75bp strand-specific paired-end mRNA libraries were prepared using the ribodepletion 2.0 protocol by the British Columbia Genome Sciences Center (BCGSC, Vancouver, BC) and sequenced on the Illumina HiSeq 2000/2500. Sequenced reads were quantified using Kallisto v0.45.030 with a GRCh38 transcriptome reference prepared using the coding and noncoding transcript annotations in in Gencode v29 and RepBase v24.01 and gene-level counts and abundances were produced using tximport v1.16.131.
Screening of C/G Fusion in patient samples. The C/G fusion transcript was detected by Fragment length analysis or fusion detection algorithms STAR-fusion v1.1.0 and TransAbyss v1.4.1032,33. Details of the procedure are described previously4.
Transcriptome Analysis: Differentially expressed genes between C/G-CB and GFP-CB cells were identified using the limma voom (v3.44.3 R package) with trimmed mean of M values (TMM) normalized gene counts34. Genes with absolute log2 fold-change > 1 and Benjamini–Hochberg adjusted p-values < 0.05 were retained. Unsupervised hierarchical clustering was completed using the ComplexHeatmap R package (v2.4.3), utilizing Euclidean distances with the ward.D2 linkage algorithm. Log2 transformed TMM normalized counts per million (CPM) were used as input, with a count of 1 added to avoid taking the log of zero. Hierarchical clustering of primary C/G AML samples and C/G-CB cells using a C/G transcriptome signature was carried out. The signature genes (N=1,116 genes) were defined as those within the 75th percentile of absolute log2 fold-changes and adj. p.value < 0.001, when contrasting C/G fusion positive patients (N=39) against a heterogenous AML reference cohort (N=1,355). The 85th percentile of this signature (N = 167 genes) was used to define a C/G gene set in GSEA.
Gene-set enrichment scores were calculated using the single-sample gene-set enrichment (ssGSEA) method (GSVA v1.32.0), which transforms normalized count data from a gene by sample matrix to a gene-set by sample matrix35. Counts were TMM normalized and log2(x+1) transformed prior to gene-set analysis. Curated signaling and metabolic gene-sets from the KEGG database were included in the analysis (gageData v2.26.0). Significant gene-sets (Benjamini–Hochberg adjusted p-values < 0.05) associated with C/G-CB cells were identified using limma v3.44.3 with the GSVA transformed gene-set by sample matrix as input.
GSEA was performed using the ‘unpaired’ comparison in the GAGE R package (v2.38.3), which tests for differential expression of gene-sets by contrasting C/G-CB against GFP-CB cells in each condition to define pathways enriched in EC co-culture versus MC. Non-redundant gene-sets were extracted for further analysis, followed by the identification of core genes that contribute to the pathway enrichment. Gene-sets from the Molecular Signatures Database (MSigDB) and the KEGG pathway database were used. Enrichment score plots for the HSC and C/G signatures were generated using the R package fgsea (v1.14.0). Log fold change values obtained from limma (contrasting C/G-CB EC week 6 against C/G-CB MC week 6) were used as a ranking metric for genes in the two signatures.
Unsupervised clustering of C/G-CB cells with pediatric AML primary diagnostic samples (N=1,033) and healthy normal bone marrows (N=68) was performed by uniform manifold approximation and projection (UMAP) using the uwot v0.1.8 R package36. For UMAP clustering, gene counts underwent variance stabilizing transformation (VST) using the DESeq2 v1.28.1 package. Input genes for clustering (N=6,678 genes) were selected using the mean versus dispersion parametric model trend (SeqGlue v0.1) to identify genes with high variability.
Identification of fusion-specific CAR targets involves three main steps: 1) Determine the ratio of expression for AML primary samples versus healthy normal hematopoietic tissue samples (bulk normal bone marrow, N=68, in combination with CD34+ selected peripheral blood samples, N=16) from log10 transformed normalized expression as transcripts per million, (TPM). Normalization was completed on the full gene expression matrix followed by ratio analysis on 19,901 annotated protein-coding genes for the identification of therapeutic targets. The ratio is calculated per gene from the mean expression in AML and normal tissues, where normal healthy hematopoietic tissue mean expression is the divisor, which acts as a measure of over or under expression. A normal curve is fit to the ratio values, and genes with ratios greater than +2 standard deviations were retained. This process is carried out for all heterogenous AML samples (N=1483) as a group and then repeated iteratively within AML fusion and mutation subtypes, including C/G, to ensure the inherent variability of gene expression in different fusion classes is addressed and all viable targets are identified for any given subtype. Genes are then further refined to include those with maximum expression < 1.0 TPM in normal healthy hematopoietic tissue samples, and thus considered to have AML restricted expression when compared to healthy controls. 2) AML restricted genes were further selected if found to be significantly overexpressed by RNA-seq for bulk fusion positive patient samples compared to bulk healthy bone marrows and were likewise overexpressed in C/G-CB at weeks 6 and 12 in EC co-culture with an absence of expression (< 1.0 TPM) in GFP-CB controls providing several candidate targets. 3). Final selection of optimal CAR-T targets was determined by the identification of candidate genes with cell surface localization potential as annotated by the Human Protein Atlas (https://www.proteinatlas.org/) or Jensen Lab compartments database (https://compartments.jensenlab.org/), in addition to having moderate to high expression in C/G patient samples (maximum expression ≥ 10 TPM), expression in a majority (> 75%) of patient samples, and an absence of expression in healthy hematopoietic tissues as noted in step 1 above.
Generation of human FOLR1 CAR T cells
CAR T cells were generated by transducing healthy donor T cells (Bloodworks Northwest) with lentivirus carrying the FOLR1 CAR vectors. Peripheral blood mononuclear cells from healthy donors were isolated over Lymphoprep (StemCell Technologies, Cat# 07851). CD4 or CD8 T cells were isolated by negative magnetic selection using Easy Sep Human CD4+ T cell Isolation Kit II (StemCell Technologies, Cat # 17952) and Easy Sep Human CD8+ T cell Isolation Kit II (StemCell Technologies, Cat # 17953). Purified T cells were cultured in CTL media [RPMI supplemented with 10% Human serum (Bloodworks Northwest), 2% L-glutamine (Gibco, Cat# 25030-081 1% pen-strep (Gibco, Cat#15140-122), 0.5 M beta-mercaptoethanol (Gibco, Cat# 21985-023), and 50 U/ml IL-2 (aldesleukin, Prometheus)] at 37°C in 5% CO2. T cells were activated with anti-CD3/CD28 beads (3:1 beads: cell, Gibco, 11131D) on Retronectin-coated plates (5 ug/mL, coated overnight at 4°C; Takara, Cat# T100B) and transduced with CAR lentivirus (MOI = 50) one day after activation via spinoculation at 800g for 90 min at 25°C in CTL media (+50 U/mL IL-2) supplemented with 8ug/mL protamine sulfate. Transduction used 200,000 cells per well in 24-well plates. Transduced cells were expanded in CTL media (+50 U/mL IL-2) and separated from beads on day 5. As truncated CD19 was co-expressed with the CAR by a T2A ribosomal skip element, it was used to select for transduced cells. Transduced cells were sorted for CD19 expression [using anti-human CD19 microbeads (Miltenyi Biotec, Cat# 130-050-301)] on Automacs 8-10 days post activation. Sorted cells were further expanded in CTL (+50 U/mL IL-2) media for an additional 4-6 days prior to in vitro and in vivo cytotoxicity assays.
In vitro cytotoxicity studies
Target cells (C/G-CB >9 weeks in EC co-culture, M07e, WSU-AML, Kasumi-1 FOLR1+ and Kasumi-1 parental) were split 1-2 days prior to cytotoxicity assay. Target leukemia cells were labeled with 2.5 uM CFSE (Invitrogen, Cat # C34554) per manufacturer’s protocol, washed with 1X PBS, and resuspended in CTL media (without IL-2). For T cell proliferation assay, effector cells (unmodified or CAR T cells) were labeled with 2.5 uM Violet Cell Proliferation Dye (Invitrogen, Cat # C34557) washed with 1X PBS, serial diluted in CTL media (without IL-2) and combined with target cells at various effector:target (E:T) ratios in 96-well U-bottom plate. Cytotoxicity (at indicated time points) and T cell proliferation (4 days) were assessed by flow cytometry after staining cells with live/dead fixable viability dyes [FVD; Invitrogen, Cat# L34964 (cytotoxicity) or L10120 (T cell proliferation)]. Percent dead amongst target cells was assessed by gating on FVD+ amongst CFSE+ target cells. Percent specific lysis was calculated by subtracting the average of the three replicate wells containing target cells only from each well containing target and effector cells at each E:T ratio. After 24 hours of co-culture, media supernatant was assessed for IL-2, IFN-𝛄, and TNF-𝛂 production by Luminex microbead technology (provided by FHCRC Immune Monitoring Core).
Optimization of IgG4 spacer region for efficient CAR T activity
To evaluate the therapeutic potential of targeting FOLR1, we generated FOLR1-directed CARs by fusing the single-chain variable fragment (scFv) derived from anti-FOLR1 antibody Farletuzumab to the IgG4 spacer, CD28 transmembrane, 4-1BB co-stimulatory and CD3z signaling domains (Supplemental Fig. 4a). We optimized the IgG4 spacer region against fusion-positive cells lines (M0-7e and WSU-AML), C/G-CB cells, Kasumi-1 cells engineered to express FOLR1 (Kasumi-1 FOLR1+) and Kasumi-1 parental cells (Supplemental Fig. 4b). Although all constructs conferred similar cytotoxicity against FOLR1+ cells, intermediate spacer CAR produced higher levels of proinflammatory cytokines (IL-2, IFN-g and TNF-a) compared to short and long IgG4 spacers (Supplemental Fig. 4c, d). We assayed NFAT, NFkB and AP-1 expression in Jurkat Nur77 reporter cells25 transduced with the CAR constructs either cultured alone or co-cultured with Kasumi-1 FOLR1+ cells. None of the FOLR1 CAR constructs demonstrated tonic signaling in the absence of target binding (Supplemental Fig. 4e, f).
In vivo cytotoxicity studies
Target leukemia cells were transduced with mCherry/ffluciferase (C/G-CB, weeks 9-12 in EC co-culture; Plasmid #104833, Addgene) or eGFP/ffluciferase construct (WSU-AML, Kasumi-1 FOLR1+ and Kasumi-1 parental; Plasmid #104834, Addgene) and sorted for mCherry+ or GFP+ cells, respectively. Luciferase-expressing cells were injected intravenously into NSG-SGM3 (C/G-CB) at 5x106 cells per mouse or NSG (WSU-AML, Kasumi-1 FOLR1+ and Kasumi-1 parental) mice at 1x106 cells through the tail vein. Mice were treated with FOLR1 CAR T or unmodified T cells via tail vein intravenous injection one week following leukemia cell injection. Leukemia burden was measured by bioluminescence imaging weekly. Leukemia burden and T cell expansion were monitored by flow cytometric analysis of mouse peripheral blood, which was drawn by retro-orbital bleeds for the indicated time points starting from the first week of T cell injection. Flow cytometric analysis of peripheral blood and tissues was performed as described above (see Supplemental information for antibodies).
Colony-forming cell assay
Following 6 and 12 weeks of culture, cells were placed in or Megacult (Megacult-C, Collagen & Medium with Cytokines Stemcell Technologies, Cat #04961) and incubated at 37°C in 5% CO2 for 10-14 days. Colonies from megacult cultures were fixed in 3.7% formaldehyde, and then washed in PBS, and stained with MegaCult™-C Staining Kit for CFU-Mk (StemCell Technologies Cat# 04962) per manufacturer’s instructions; or were permeabilized after fixation in 0.1% Triton X-100 for 10min, blocked in in 1% BSA in PBST(PBS+0.1% Tween-20) for 30min, then stained with biotin-conjugated mouse anti-human CD41 (Biolegend, cat# 303734) and FITC-conjugated goat anti-GFP(abcam, cat# ab6662) followed by secondary stain with Alexa 647-labeled Streptavidin (Biolegend, cat# 405237) per manufacturer’s instructions, and colonies were stained with DAPI prior to imaging using the TissueFAX microscope. Mk colonies were scored based on positive staining for CD41 and enumerated.
C/G-CB and normal HPSCs after co-cultured with unmodified or CAR T cells for 4 hours were placed in Methocult H4034 Optimum (Stemcell Technologies, Cat #04034). Colonies derived from erythroid (E), granulocyte-macrophage (G, M, and GM) and multipotential granulocyte, erythroid, macrophage, megakaryocyte (GEMM) progenitors were scored and enumerated after 7-10 days as directed by manufacturer’s instructions.
Statistical analysis for in vitro and in vivo studies
Unpaired, two-tailed Student’s t test was used to determine statistical significance for all in vitro studies. Log-rank (Mantel-Cox) test was used to compare Kaplan-Meier survival curves between experimental groups. Statistical significance is defined for p<0.05.
Data and code Availability
RNA-seq data on primary patient samples are deposited in GDC, SRA and Target Data Matrix. RNA-seq data on engineered CB are deposited in GEO. All codes used in this are publicly available.