Cell lines and primary cells culturing conditions
Human lymphoma cell lines DB, and DG-75[37] (Kind gift from Dr G Klein, Karolinska Institute, Stockholm, Sweden), U-698[38] (Kind gift Kenneth Nilsson, Uppsala University), Daudi (ATCC CCL-213), Raji (Sigma Aldrich) were cultured in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum (FBS), 1% penicillin/streptomycin (PeSt) and 1% sodium pyruvate. All reagents were from Invitrogen. All lymphoma cell lines were modified to stably express green fluorescent protein, firefly luciferase (FLuc) and puromycin (for selection) by lentiviral transduction (Magnus Essand, Addgene, Plasmid #80389)[39]. Cells expressing transgenes were selected using 5µg/ml puromycin.
Mouse B-cell lymphoma cell line A20 (ATCC TIB-208) was cultured in culture medium (RPMI 1640) containing 10% heat-inactivated FBS, 1% (PeSt) and 1% sodium pyruvate. To generate A20 cells stably expressing human CD20 (termed as A20-hCD20), A20 cells were transduced with a lentiviral vector encoding hCD20, FLuc and puromycin separated by a self-cleaving peptide (T2A and F2A). A20-hCD20 cells were selected using 3µg/ml puromycin.
Peripheral blood mononuclear cells (PBMCs) were isolated by Ficoll-Paque (GE Healthcare Life Science) from fresh buffy coats taken from healthy donors, collected from the Blood Center at Uppsala University Hospital. They were used as a source for human T cells, which were cultured in RPMI 1640 supplemented with 10% FBS, 1% PeSt and 1% sodium pyruvate with 50IU/ml IL-2 (Proleukin, Novartis) if not stated otherwise.
Retro- and lentiviral vector construction and production
To create lentiviral vectors encoding CAR20 the variable region sequence from Rituximab was incorporated into a CAR cassette with the signaling domain of CD3zeta and the co-stimulatory signaling domain of CD137 as previously described[40]. The CAR constructs were cloned into a third-generation self-inactivating (SIN) lentiviral vector (SBI, System Biosciences) under the control of elongation factor-1 alpha (EF1a) promoter. Green fluorescence protein (GFP) was inserted after the CAR construct using a T2A self-cleaving peptide in both plasmids to allow detection of CAR-T cells. A mock construct only encoding GFP under the control of the EF1a promoter was used as a control. All sequences were purchased from GenScript. Production of third generation viral particles has been previously described[41]. The retroviral vector encoding human CAR20 sequence was purchased from BioNTech and used for therapeutic efficacy studies in xenograft Daudi model.
The variable region of Rituximab was incorporated into a murine CAR construct containing murine CD3ζ and murine CD28 signaling domains, has been described previously[42]. To construct retroviral vector RV(mCAR20), the above constructs were subcloned into mouse stem cell virus-based retroviral vector, pMIG-W (Luk Parijs, Addgene, plasmid #12282)[43]. The RV(mCAR19) and RV(mCAR19-NAP) constructs have been described previously[23]. An empty retroviral vector was used as Mock control. The NAP sequence was inserted in the orientation opposite the CAR20 cassette and under the control of murine NFAT-IL-2 promoter to construct retroviral vector RV(mCAR20-NAP). Retrovirus was produced using the packaging plasmid pCL-Eco in the Gryphon-Eco retroviral packaging cell line (Allele Biotechnology).
Human T cell engineering, enrichment and expansion
Human PBMCs, see above, were activated using OKT-3 (100ng/ml, BioLegend San Diego, CA) or TransAct (1:100, Miltenyi Biotec) and IL-2 (100 IU/ml, Proleukin) for 3 days at a concentration of 2x106 cells per ml. Following activated T cells (1x106 cells) were re-suspended in 20µl concentrated lentivirus together with 10mg/ml protamine sulfate (Sigma-Aldrich) and IL-2 (100 IU) and incubated for 4hrs at 37°C. The following day T cells were transduced in the same manner. After transduction T cells were cultured (in culture medium supplemented with 50IU/ml IL-2) for 7 days before CAR-T cells were enriched by sorting out GFP+ cells (BD FACSAriaIII, BD Biosciences). Sorted cells were expanded using rapid expansion protocol (REP) previously described[44].
Mouse T cell activation and transduction
Spleens were collected from BALB/c mice (Janvier) and mashed against a 70µm cell strainer (Coring) to create single cell suspension. After resuspended in red blood cell lysis buffer (ACK lysing buffer, Invitrogen) for 5min, splenocytes were activated using Concanavalin A (2µg/ml) (Sigma Aldrich) and murine IL-7 (Miltenyi Biotec) in RPMI 1640 containing 10% FBS, 1% PeSt and 1% sodium pyruvate. Two days post activation splenocytes were transduced with retrovirus as previously described[23].
T cell cytotoxicity assay and IFN-γ ELISA
Human T cell cytotoxicity assay:
The cytotoxicity assay was performed using luciferase-expressing target cells. CAR-engineered human T cells were rested for 3d after rapid expansion in a medium containing a low dose of IL-2 (20 IU/ml) before performing the cytotoxicity assay. The T cells were co-cultured with luciferase-tagged target cells (DG75, DB, U698, Raji and Daudi, at 3:1 ratio) in a total volume of 200μl, in a round-bottom 96-well plate. To detect the luciferase expression and activity, ONE-Glo luciferase assay system (Promega) was added according to manufacturer’s protocol. Specific lysis of target cells was calculated by comparing luminescence of co-cultured samples to untreated target cells.
Murine T cell cytotoxicity assay:
The cytotoxicity assay was performed using luciferase-expressing target cells. Murine splenocytes were harvested, activated and transduced with retroviral vectors as above described. The engineered-T cells were co-cultured with firefly luciferase-tagged A20-hCD20 target cells at different ratios (50:1, 20:1, 5:1 and 1:1) in a round bottom 96-well plate (total volume 200µl). After 5 days of co-culture, 80µl supernatant was collected for IFN-γ analysis determined by an ELISA kit (Mabtech). Luciferase expression and activity (as an indicator of target cell viability) were determined by using ONE-Glo luciferase assay system (Promega) according to manufacturer’s protocol. Specific lysis of target cells was calculated by relating luminescence of co-culture with target cells alone.
T-cell cytotoxicity assay towards patient samples
Biopsy samples containing primary mantle cell lymphoma (MCL) cells were either left untreated or co-cultured together with engineered-T cells at a 1:1 (1=25 000 cells) ratio for 3 days in round bottom 96-well plate in a total volume of 200µl per well. After 2 days co-culture, supernatant was collected for analysis of IFNγ secretion by an ELISA kit (Mabtech). After 3 days of co-culture, the remaining amount of CD20+ cells (as an indicator of target cell viability) was assessed by flow cytometry. Specific lysis of target cells was shown as the proportion of CD20+ cells normalized to the one at the start of the experiment.
Cytotoxicity assay using Incucyte
Measurement of immune cell killing of target tumor cells was analyzed with the Incucyte® Zoom image analysis software (Essen BioScience). Briefly, one image per well in a 24-hour repeat scanning was schedule with the Phase contrast, green and red channels in 10x objective. Images were taken every 30 minutes with the standard scan schedule and “Basic Analyzer” module. Image collections and processing definitions for every cell line and assay were stablished following the suggested procedures by the manufacturer. Processing definitions were stablished with phase and fluorescence masks for scarlet expression to quantify target cell proliferation, determined as total red fluorescence confluence. Target cell killing by CAR-T cells was determined by comparing red fluorescent signal of co-cultured (at the effector to target ratio 5:1) samples to untreated target cells and reported as relative cell viability.
Flow cytometry
In order to assess transduction efficiency, CAR and GFP expression of engineered-T cells were detected by flow cytometry. The T cells were stained with CD3-BB700 (Clone SP34-2) and anti-mouse IgG (H+L)-AF647 (to detect CAR expression, A21237, Thermo Fisher Scientific).
Human CD20 expression in human cell lines and in the modified A20-hCD20 cells was assessed by staining the cells with anti-human CD20-APC (Clone 2H7), isotype was used as control (Mouse IgG2b,κ). Stained cells were analyzed by flow cytometry using BD FACSCanto II (BD Biosciences) or CytoFLEX LX (Beckman Coulter) and analyzed using FlowJo_v10.8.1 (FlowJo LLC).
Animal experiment
Survival experiment
Daudi model: NOD-SCID mice (8 weeks) were implanted intravenously (i.v.) with 1×106 firefly luciferase (fLuc) expressing Daudi cells. Mice were then treated with two doses (2×106 cells/dose) intravenously on 4 and 8 days post tumor implantation. The tumor progression was monitored by in vivo imaging system (IVIS) Lumina III (PerkinElmer). Mice were intraperitoneal (i.p.) injected with 100 μL of 30 mg/mL of D-Luciferin potassium salt (Perkin Elmer) and imaged 10min after substrate injection. Mice were monitored regularly for their health status and euthanized when reaching the humane endpoint.
A20-hCD20 model: Female BALB/c mice (8 weeks) were implanted subcutaneously (s.c.) with 2×105 murine lymphoma A20 expressing human CD20 (A20-hCD20). The mice were treated with 3×106 T cells intravenously (i.v.) 10-, 14-, and 18-days post tumor implantation. The mice were monitored regularly and euthanized when reaching the humane endpoint. Mice were sacrificed when reaching the humane endpoint (a tumor size reach 1cm3). The experiment was performed twice, and all data were pooled together (Mock-T: n=20, mCAR20-T: n=20, mCAR20(NAP)-T: n=19).
The mice were monitored for tumor growth until reaching the humane endpoint volume 1000 mm3. The tumor size was calculated as: volume=length×width2×π/6. The time to endpoint (TTE) for each mouse was calculated as TTE= [log (EPV)-b]/m. The constant b is the intercept and m are the slope of the line obtained by linear regression (time vs tumor volume) of a log-transformed tumor volume data set, which comprised of the first measured tumor volume when EPV was exceeded and three consecutive tumor volumes immediately prior to the attainment of EPV. Any animal that died from treatment-related causes was assigned a TTE value equal to the day of death. Any animal that died from non-treatment-related causes was excluded from the analysis. Survival curve was generated based on the TTE value using the Kaplan-Meier method and compared using the log-rank (Mantel-Cox) test.
Safety assessment
Female Balb/c mice (3 mice/group) were subcutaneously implanted on day 0 with A20 tumor (murine lymphoma cell line A20) and treated intravenously with Mock-T, mCAR-T or mCAR(NAP)-T cells (targeting murine CD19) on day 10, 14 and 18. Different organs (brain, heart, kidney, lung, liver, and spleen) were collected on day 20 and, fixed in 4% formalin, embedded paraffin and sent to BioVet AB for pathologic evaluation. Sections were 3-4 um thin and stained with Heamatoxylin-Eosin (HE). Photomicrographs were taken when the observed lesions appeared.
Whole blood loop analyses
Fresh whole blood was taken from ten healthy volunteers (D1-D10) and immediately mixed with a low amount of soluble heparin. Then whole blood was immediately transferred to heparin pre-coated plastic tubes forming the loops, followed by administration of the test items, 5μg/ml NAP proteins and 25μg/ml NAP proteins. PBS was used as negative control and 3μg/ml Alemtuzumab (Lemtrada, Sanofi) was used as positive antibody, and 3μg/ml LPS (Sigma-Aldrich, Sweden) was used as positive control. Additionally, fresh blood from each donor was used for hematology measurements directly after blood collection (described as pre-loop baseline). Additionally, blood collected directly from donors was processed to plasma and included in cytokine and complement analysis. Blood samples were collected at 15 minutes for complement analysis and immune cells activation, while blood samples collected at 4 hours were analyzed for hematology analysis, immune cell activation and cytokine analysis.
For hematology analysis, it included the parameters: red blood cell (1012/L), platelets (109/L), white blood cell (109/L), differential neutrophil (109/L), lymphocyte (109/L), monocyte (109/L), eosinophil (109/L) and basophil (109/L). These parameters were measured at 0h (pre-loop baseline) and 4h time-point by the Hematology Analyzer Sysmex XN-L350 (Sysmex). Hematology analysis of zero time point samples was performed as three subsequent measurements, while samples collected in the loop system at 4 hours were done as single measurements.
For cytokine and complement analysis, plasma harvested at 0 (pre-loop baseline) and 4-hour time-point was analyzed for IFN-γ, TNF-α, IL-2, IL-6 and IL-8 with a Mesoscale V-plex kit (Meso Scale Discovery) according to the manufacturer's instructions. Complement analysis (C3a and C5a) was performed on plasma collected at 0h (pre-loop baseline) and 15min time-point with ELISA kits (Raybiotech) according to the manufacturer’s instructions. Detection limit or limit of detection (LOD) is the smallest concentration of an analyte from which it is possible to deduce the presence of an analyte in the test sample, while quantification limit or limit of quantification (LOQ) is the smallest concentration of an analyte that can be determined with the specified degree of accuracy and precision. Lower limit of detection (LLOD) was calculated by MSD software and defined as 2.5xSD above the zero calibrator (Standard-8).
For immune cells activation, blood was collected at the 0, 15min and/or 4h time points, and the activation of immune cells were analyzed by flow cytometry (CytoFLEX LX, Beckman Coulter). The platelets were gated as CD41+ CD45- cells and analyzed the activation marker CD62P. The lymphocytes were first identified in the forward versus side scatter (FSC/SSC) plot and further gated as T cells (CD3+CD56-), NK cells (CD3-CD56+) and B cells (CD3-CD19+). The activation phenotype of lymphocytes was determined by staining by CD69 and CD107a. Granulocytes and monocytes were as well identified in FSC/SSC plot and further defined as granulocytes (CD14-CD66b+) and monocytes (CD14+CD66b-), respectively. The activation markers were analyzed by staining with CD11b and CD83.
Ethical approval
The Uppsala Research Animal Ethics Committee has approved all the animal studies (5.8.18-19434-2019). The human buffy coat obtained from healthy donors were anonymized. The patient-derived mantle cell lymphoma cells were collected under ethical permit: EPN 233-2014.
Statistical analysis
Two-way ANOVA with Tukey multiple comparisons test was used to compare means between more than two experimental groups. For statistical analysis between two experimental groups, a non-parametric unpaired t-test was used to compare means between experimental groups. Statistical significance of survival (Kaplan-Meier) a log-rank test was used. A p value of less than 0.05 was considered as statistically significant.