Experimental Design
The objective of this controlled laboratory experiment was to determine if sustained TLR7/8 agonism (achieved by formulating small molecule, resiquimod, within a biodegradable polymer scaffold) applied at the time of GBM tumor resection would improve survival. Pharmacokinetic studies confirmed that Ace-Resi extended the duration of resiquimod exposure in the murine brain relative to bolus injection. Mice bearing two different orthotopic luminescent GBM tumors (GL261-mCh-Luc and CT2A-Luc) were utilized for evaluation, as well as confirmation in the same cell line without reporter protein (CT2A-Par). Survival was prospectively selected as the primary endpoint, and for luminescent tumors, serial bioluminescence imaging was used to quantify and monitor local tumor recurrence. Formation of an immune memory response towards the tumor was determined by challenging long-term surviving mice with a new inoculate of tumor cells in the contralateral hemisphere utilizing naïve mice as a control. Immune cell infiltrates were evaluated by flow cytometry after a second tumor challenge. Tolerability of Ace-Resi treatment was evaluated by weight loss. For mechanistic studies, flow cytometry and ELISA were used to assess cell populations and soluble cytokines, respectively.
Sample sizes were determined based on results from prior experiments to ensure statistical significance could be analyzed. No animals that received treatment were excluded from the analysis. One control mouse was excluded as it died unrelated to tumor burden. Mice were randomly assigned to treatment groups the day of surgery. When possible, monitoring mice for survival was done in a blinded manner. Mice were evaluated daily and euthanized if they lost more than 20% body weight, or if they exhibited signs of distress or gait abnormalities.
Chemicals and Reagents
All chemicals were purchased from Sigma-Aldrich (St. Louis, MO) and used as purchased, unless otherwise noted. All disposables were purchased from VWR International (Radnor, PA) unless indicated. High glucose Dulbecco's Modified Eagle Medium (DMEM) cell culture medium, puromycin dihydrochloride, and fetal bovine serum (FBS) were obtained from Corning (Corning, NY). Penicillin/streptomycin was purchased from Hyclone (Pittsburgh, PA). mCherry-firefly luciferase (mCh-FL) was acquired from Invitrogen (Carlsbad, CA). 2-ethoxypropene was obtained from Matrix Scientific (Columbia, SC). Resiquimod (Resi, > 99%) was acquired from Accel PharmTech (East Brunswick, NJ).
Cell Culture
GL261 cells were engineered to express mCherry-firefly luciferase (mCh-Luc) using previously developed lentiviral vectors to allow for fluorescent and bioluminescent imaging as in our previous study (‘GL261-mCh-Luc’).(68) Cells were infected for 24 hours and purified by puromycin selection. Murine glioma cell line CT2A expressing firefly luciferase (‘CT2A-Luc’) was purchased from Millipore Sigma (Burlington, MA, Cat # SC195). The parental CT2A cell line (‘CT2A-Par’) was a gift from Professor Thomas Seyfried. Cells were cultured in DMEM medium supplemented with 1% penicillin/streptomycin and 10% fetal bovine serum.
Scaffold Fabrication
Dextran (molecular weight 450–650 kDa) was chemically modified to generate the hydrophobic biopolymer Ace-DEX as previously described in Kauffman et al.(43) Cyclic acetal coverage (CAC) was determined by NMR.(43) Resiquimod loaded scaffolds were fabricated via electrospinning similar to Graham-Gurysh et al.(37) Briefly, 31% CAC Ace-DEX was dissolved at a concentration of 300 mg/mL in a tri-solvent system consisting of dichloromethane, hexafluoro-2-propanol, and butanol at a ratio of 30%, 30%, and 40% v/v respectively. Resiquimod (10% wt/wt) was added to the polymer solution and extruded from a glass syringe with a 21-guage needle at a flow rate of 1 mL per hour. A voltage bias was applied to the needle and a metal collection plate. The resulting scaffold (termed “Ace-Resi”) was retrieved from the collection plate and stored at -20˚C until further use. A scaffold without any resiquimod was fabricated similarly as a control, termed “Ace-Blank”.
Scaffold Characterization
Electrospun scaffolds were evaluated by scanning electron microscopy to elucidate fiber morphology. A small piece of each scaffold was mounted on aluminum stubs using carbon tape, sputter coated with palladium, and imaged at 2 kV on a Hitachi S-4700 Cold Cathode Field Emission Scanning Electron Microscope (Hitachi High-Technologies, Krefeld, Germany).
To measure resiquimod loading, scaffold samples were weighed, dissolved in DMSO and the absorbance at 260 nm was compared to a standard curve. Resiquimod encapsulation efficiency was defined as the amount of measured resiquimod loaded divided by the theoretical resiquimod loaded. Resiquimod release and scaffold mass loss was determined by adding pre-weighed scaffold samples to a sink of phosphate buffered saline (PBS, pH 7.4) agitated at 37°C. At specific time points, scaffold samples were removed, washed with basic water (water with 0.1% triethylamine), lyophilized, and re-weighed to determine the total scaffold mass loss. Scaffold mass loss was calculated according to Supp Eq. 1. Each scaffold sample was then dissolved in DMSO and the absorbance at 260 nm was compared against a standard curve to determine the amount of resiquimod retained in each sample. Resiquimod release was calculated according to Supp Eq. 2. Ace-Resi and Ace-Blank scaffolds were tested for endotoxin and measured less than 0.1 EU per mg of scaffold (Pierce Chromogenic Endotoxin Quant Kit).
All experimental protocols were approved by the Animal Care and Use Committees at UNC-CH, and care of the mice was in accordance with the standards set forth by the National Institutes of Health Guide for the Care and Use of Laboratory Animals, and the American Veterinary Medical Association. To assess the pharmacokinetics of resiquimod released from Ace-Resi/fast within the mouse brain compared to bolus soluble injection, experimental groups were applied to a resection cavity created in the right frontal lobe in a non-tumor bearing C57BL/6 mouse. At 1hr, 6hr, 1, 3, and 7 days, mice were euthanized, and brain and plasma collected. Any remaining scaffold was removed from the brain. The remaining scaffold was lyophilized, dissolved in DMSO, and evaluated by fluorescence (excitation 260nm, emission 370nm) against a standard curve to quantitate the percentage of resiquimod retained within the scaffold.
Plasma samples and brain tissue were snap frozen. Brain tissue was homogenized in PBS using 1.4 mm porcelain homogenization beads and diluted in PBS for a 4x dilution. Samples were analyzed with LC-MS/MS to measure the concentration of resiquimod. The lower limit of quantification for resiquimod in the brain was 1 ng per gram of tissue and in plasma was 1 ng per mL.
Glioblastoma mouse model
C57BL/6 mice were purchased from Charles River (stock# 027) or Jackson Labs (stock# 00064) for GL261 and CT-2A models, respectively. For all surgical procedures, mice were anesthetized by vapor isoflurane. Bupivacaine was utilized for local analgesia and meloxicam (5 mg/kg) was administered prior to surgery and once daily for the following 3 days. Mice were evaluated daily and euthanized if they lost more than 20% body weight, or if they exhibited signs of distress or gait abnormalities.
A model of GBM resection and recurrence was used to evaluate drug-loaded scaffolds similar to previously described.(37) Complete timelines for all studies are in the main text or supplement (Fig. 2A, Supp Fig. 2A, 3A, 4J, 5A). GBM tumor cells were injected (either GL261-mCh-Luc, CT2A-Luc, or CT2A-Par) at 1 x 105 cells in 2 µL of PBS at a rate of 1 µL per minute into the right frontal lobe of the brain via burr hole approximately 2mm lateral of bregma and 0.5 mm below the dura. For luminescent tumors, growth was monitored by bioluminescent imaging (BLI, Perkin Elmer IVIS Spectrum In Vivo Imaging System). Ten to 14 days after implantation, a cranial window ~ 3mm in diameter was created around the initial burr hole. Tumors were then partially resected, treatment was applied to the surgical cavity, and the skin was closed with Vetbond tissue adhesive (3M, Maplewood, MN). For mice treated with a bolus soluble resiquimod, 100 µg of resiquimod in 5 µL of PBS and DMSO (80%, 20% v/v) was injected into the exposed brain tissue of the resection cavity. For mice treated with scaffolds, the scaffolds were simply placed into the resection cavity. The resiquimod dose implanted via Ace-Resi scaffolds was controlled by changing mass of scaffold implanted knowing the empirical drug loading. With a 9.5% w/w loaded scaffold, a 100 µg dose of resiquimod would require 1.05 mg of scaffold, whereas a 10 µg dose of resiquimod would require 0.11 mg of scaffold. Sample sizes for each study are detailed in Supp Table 2. Mice were monitored for tumor recurrence and survival. Serial BLI was used to quantify tumor growth starting the day after tumor resection (Day 1).
For GL261-mCh-Luc and CT2A-Luc studies, to assess for a memory immune response, survivors were challenged with tumors (2 x 105 GL261-mCh-Luc or CT2A-Luc) in the contralateral hemisphere 9–10 weeks after initial resection and treatment. As a control, naïve mice were also implanted with tumors. Mice were followed for survival and 6–7 weeks later, survivors were re-challenged. One week after the second challenge, mice were euthanized and brain leukocytes were analyzed by flow cytometry.
For the CT2A-Par study, survivors were challenged with 2 x 105 CT2A-Par tumors in the contralateral hemisphere 9 weeks after resection and treatment. One week after tumor challenge, mice were euthanized, and brains and one spleen were collected. Leukocytes were isolated from the pooled brains and CD8+ T cells isolated from the spleen via magnetic bead separation (Miltenyi cat # 130-104-075). Additionally, as a control a healthy naïve mouse was also euthanized and CD8+ T cells were isolated from the spleen. T cells from both groups were expanded through 48hr culture on CD3 and CD28 antibody coated plates (0.5 µg per well, 4hr incubation at 37ºC). For long-term survivors, brain leukocytes (accounting for 5% of the total effector population) and CD8+ T cells isolated from spleen (accounting for 95% of the effector cells) were cocultured with CT2A-Par cells at various ratios. For naïve mice, CD8+ T cells from the spleen (accounting for 100% of the effector population) were cocultured with CT2A-Par cells at various ratios. Media was collected at 24 hours from the coculture and evaluated by cytokine ELISA for IFNγ (Biolegend Cat # 430801).
Evaluation of Early Immunological Effects
Mice were implanted with CT2A-Luc tumors in the right frontal lobe as described above. Two weeks later, tumors were resected, and scaffolds implanted with the following groups: Ace-Blank, Ace-Resi 100 µg. As controls, a subset of mice with and without tumors did not undergo resection, ‘tumor’ and ‘healthy’ respectively. Sample sizes for these groups are detailed in Supp Table 2. At days 1, 3, and 7 following resection mice were euthanized. Brain, cervical lymph nodes, spleen, bone marrow, and blood were collected for leukocyte analysis. Cytokines were measured through ELISA according to manufacturer guidelines: IFN-β (R&D Systems Catalog # DY8234-05), TNF-α (Biolegend Cat # 430901), IFN-γ (Biolegend Cat # 430801).
Cervical lymph nodes, spleen, and bone marrow (extruded from a single femur and tibia) were harvested. Single cell suspensions were generated by grinding through a 70-µm cell strainer. Red blood cells were lysed from spleen and bone marrow samples with ACK lysis buffer. All cell suspensions were washed with PBS + 2% fetal bovine serum (FACs buffer), then passed through another 70 µm strainer. Blood was collected into both K3EDTA coated and serum separator tubes (Greiner). Serum was stored at -80°C for later use. Fifty µL of blood from K3EDTA tubes was treated with ACK lysis buffer to remove red blood cells. The cell suspension was then washed with FACs buffer.
For S1P1 staining, bone marrow (extruded from a single femur) was harvested into S1P1 buffer (500 mL PBS, 5 mL 10% Buffered Neutral Formalin, 2 mL 0.5 M EDTA, 2.5 g BSA) to immediately fix cells. Single cell suspensions were generated by grinding through a 70-µm cell strainer in S1P1 buffer. Red blood cells were lysed from spleen and bone marrow samples with ACK lysis buffer. Cell suspensions were washed with S1P1 buffer and passed through another 70 µm strainer. Cells were washed twice in FACS buffer, stained for 30 minutes on ice, then washed twice and resuspended in FACS buffer.
Whole brains were removed and dissociated via homogenization in Hank’s buffered salt solution (HBSS). For lymphocyte evaluation 7 days after tumor challenges in GL261-mCh-Luc and CT2A-Luc models, brain homogenate was resolved with a 70%, 30% Percoll gradient, collecting single cells at the interface. For leukocyte analysis of the early immunological effects on days 1, 3, and 7 following treatment, brain homogenate was suspended in 30% Percoll gradient and the cell pellet was collected.
Following generation of single-cell suspensions, cells were divided based on downstream application (either surface staining or intracellular cytokine staining (ICCS)). For the identification and exclusion of dead cells in all flow analysis, samples were stained with eBioscience Fixable Viability Dye (ThermoFisher) diluted 1:1000 in FACS buffer for 30 minutes on ice, then washed twice with FACS buffer. All samples were acquired using either an Attune NxT flow cytometer (ThermoFisher) or an LSRFortessa flow cytometer (Becton Dickinson). Flow data analysis was performed using FlowJo (Becton Dickinson). All samples were subject to sequential debris, doublet, and dead cell removal prior to analysis. A complete list of antibody clones, fluorochrome conjugates, vendors, and dilutions is included in Supp Table 3.
For ‘myeloid’, ‘systemic lymphocyte’, and ‘microglia and macrophage’ panels (involving only surface stains), cells were washed twice in FACS buffer, then stained for 30 minutes on ice in FACS buffer, resuspended in 1% formaldehyde in PBS for 30 minutes on ice, then washed twice and resuspended in FACS buffer.
For ICCS, cells were treated with brefeldin A and monensin (5 µg/mL and 2 µM, Biolegend) with and without phorbol 12-myristate 13-acetate and ionomycin (50 ng/mL and 500 ng/mL, Sigma) for 4 hours at 37°C. Cells were then fixed and permeabilized for 30 minutes on ice using the eBioscience™ Foxp3 / Transcription Factor Staining Buffer Set (Thermo Fisher), washed twice with permeabilization buffer, then stained with a mixture of fluorochrome-conjugated antibodies diluted in permeabilization buffer for 30 minutes at room temperature, washed twice with permeabilization buffer and resuspended in FACS buffer.
Myeloid panel: Cells expressing high levels of CD45 were selected for downstream analysis Leukocytes (CD45high), Lymphocytes (CD45high TCR-β/CD19+), DCs (CD45high TCR-β/CD19- CD11c+ MHC-II+), Monocytes (CD45high TCR-β/CD19- CD11bhigh Ly6C+), Macrophages (CD45high TCR-β/CD19- CD11bhigh Ly6C-), Neutrophils (CD45high TCR-β/CD19- CD11bhigh Ly6G+), CD11bint (CD45high TCR-β/CD19- CD11bint), and Lin- (Negative for other lineage-defining markers). Cell types were assessed for MFI of CD80, CD86, and MHC-II. The gating strategy for this panel is in Supp Fig. 11.
Microglia and macrophage panel: Cells expressing high levels of CD45 were selected for downstream analysis and microglia were identified through expression of an intermediate CD45 signal (CD45int). Lymphocytes (CD19+ CD3+ TCRß+) were removed through a dump channel (PE/Dazzle594). Non-lymphocytes were subject to CD11b stratification, and the high-expressing population selected. This population (CD45high CD19- CD3- TCRß- CD11bhigh) designated macrophages. Macrophages and microglia were assessed for frequency and MFI of CD80, CD86, and CD206. The gating strategy for this panel is in Supp Fig. 12.
Systemic lymphocyte panel: B cells (CD19+) and NK cells (NK1.1+) were removed via dump channel and CD4+ T cells (CD90.2+ CD4+) CD8+ T cells (CD90.2+ CD8+) were identified. T cell central memory (CD44+ CD62L+), effector memory (CD44+ CD62L-), and naïve (CD44- CD62L+/-) subpopulations were then stratified. CD69+ and CD44 MFI were analyzed for both CD4 and CD8+ T cells. Total T-cell population S1P1+ differences were assessed by selecting for CD90.2+ CD19- cells (combined CD4s and CD8s).
Brain lymphocyte panel: NK cells (CD90.2+ NK1.1+), CD4+ T cells (CD90.2+ CD4+), and CD8+ T cells (CD90.2+ CD8+) populations were identified. T cell central memory (CD44+ CD62L+), effector memory (CD44+ CD62L-), and naïve (CD44- CD62L+/-) subpopulations were then stratified. CD25+, IFN-γ+, GzmB+, PD1+ frequency and MFI were analyzed for NK cells and CD8+ T cells. IFN-γ+, FoxP3+, IL-10+ frequency and MFI were analyzed for CD4+ T cells.
FlowSOM UMAP Analysis
Following the gating strategy in Supplemental Fig. 13B-C, the flow cytometry data for the myeloid cell panel were down-sampled on a CD45 + gate to ensure each animal was represented equally. Files were concatenated to generate UMAP figures clustered by FlowSOM for the brain and cervical lymph node. FlowSOM clusters were defined via a heatmap in Supplemental Fig. 13D-E.
Statistical Analysis
Statistical analysis was performed using GraphPad Prism (La Jolla, CA). Mouse survival curves were evaluated by Log-Rank (Mantel-Cox) test. Statistical significance for flow cytometry data was determined by one- or two-way ANOVA, with multiple comparisons using Tukey’s or Dunnett’s post-hoc test. When no difference was seen temporally across days 1, 3, and 7, non-resected ‘tumor’ mice results were grouped together to increase power.