1. Introduction
Cell-based therapies, including those using tumor-infiltrating T cells, chimeric antigen receptor (CAR)-T cells, and stem cells, have become major therapeutic strategies for cancers and other diseases [
1,
2,
3]. Cell-based therapies rely on the transferred cells arriving at the target organ or tissue in sufficient numbers to have a therapeutic effect, but this is difficult to non-invasively ascertain. Hence, to optimize the therapeutic potential of cell-based therapies, the ability to measure the distribution of therapeutic cells throughout the body and assess the effects of cell engineering on migration, targeting and engraftment is crucial [
4,
5]. Zirconium-89 (
89Zr)-oxine has been developed as an ex vivo labeling agent for tracking cells in vivo over multiple days using positron emission tomography (PET) imaging [
6,
7]. Unlike indium-111 (
111In)-oxine or technetium-99m (
99mTc)-hexamethylpropyleneamine oxime (HMPAO) cell labeling that utilizes single-photon emission computed tomography (SPECT) imaging [
8,
9,
10,
11], the utilization of PET enables cell detection with higher sensitivity, lower radioactivity, and, thus, lower radiotoxicity to the cells. Furthermore, the short half-life of
99mTc (6-h) is a limitation because it greatly shortens the imaging window for tracking cells [
12,
13]. In comparison,
89Zr and
111In have longer half-lives of 3.3 days and 2.7 days, respectively, making them more suitable for the longer-term tracking of cells [
5,
6,
7,
8].
89Zr-oxine ex vivo cell labeling has been applied to the tracking of many different cell types, such as T cells, natural killer (NK) cells, dendritic cells (DCs), hematopoietic stem cells, eosinophils and plasma cells, in preclinical mouse and non-human primate studies, and has proven to be safe, exhibiting negligible radiotoxicity, when optimal labeling doses are used [
6,
14,
15,
16,
17,
18,
19]. Studies using labeled CAR-T cells indicated no difference in the cytokine production and cytotoxic function between the labeled and non-labeled cells [
20,
21]. Clinical application of
89Zr-oxine cell labeling and tracking has recently started [
22].
89Zr-oxine and
111ln-oxine share structural similarities and labeling protocols. Previous reports indicated that post-labeling dissociation of the
111ln-oxine complexes within the cell leads to the intracellular trapping of the
111ln while releasing the oxine [
9,
23,
24,
25]. Similar studies for
89Zr-oxine have not been performed. Despite their differences in oxidation state, +3 for indium (ln) and +4 for zirconium (Zr), both metals have shown similarities in reactivity and preferred ligand type [
26]. Thus, we postulated that both isotopes share similar cellular binding and intracellular distribution.
In this study, we aimed to determine the intracellular distribution and stability of protein binding of 89Zr-oxine, compared to 111ln-oxine, after cell labeling.
2. Materials and Methods
2.1. Animals
C57BL/6 mice were purchased from Jackson Laboratories and handled in accordance with a protocol approved by the National Cancer Institute Animal Care and Use Committee. Either male or female mice aged 8–12 weeks of age were used for naïve T cell purification, and those aged 10–24 weeks were used for other experiments.
2.2. Cell Culture
RPMI 1640 medium (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 2 mmol/L L-glutamine (Thermo Fisher Scientific), 100 IU/mL penicillin/100 µg/mL streptomycin (Thermo Fisher Scientific), 10% fetal calf serum (FCS, Gemini Bio Products, West Sacramento, CA, USA), and 0.05 mmol/L 2-mercaptoethanol (Millipore Sigma, Burlington, MA, USA) was used for all cell cultures. To generate bone marrow-derived monocytes and macrophages, bone marrow cells flushed out of the femurs and tibias of mice were grown for 5 days (for monocytes) and 9 days (for macrophages) with the addition of 20 ng/mL and 10 ng/mL, respectively, of macrophage-colony stimulating factor (M-CSF, Peprotech, Cranbury, NJ, USA). For differentiation of dendritic cells (DCs), bone marrow cells were grown in 20 ng/mL of granulocyte-macrophage-colony stimulating factor (GM-CSF, Peprotech) for 9 days. Naïve T cells were purified from the spleen of mice as follows: The spleen was passed through a 70 µm-pore strainer (Corning Life Sciences, Tewksbury, MA, USA) in phosphate-buffered saline (PBS), and the obtained single cell suspension was layered over Cell Separation Medium (Lonza, Walkersville, MA, USA), followed by centrifugation at 240× g for 12 min with low acceleration/deceleration. The cells in the interphase were collected, washed with PBS containing 0.5% bovine serum albumin (Miltenyi, Gaithersburg, MA, USA) and 2 mmol/L ethylenediaminetetraacetic acid (EDTA), and underwent a positive selection of T cells using CD8 and CD4 T cell magnetic beads (Miltenyi) and an LS column (Miltenyi) according to the manufacturer’s instructions. EL4 (murine lymphoma) cells and DC2.4 cells (murine DC line) were purchased from American Type Culture Collection and Millipore Sigma, respectively. Kit225K6 (human T cell lymphoma) cells were kindly provided by the late Dr. Thomas Waldmann, National Cancer Institute, National Institutes of Health. MC38 (murine colon adenocarcinoma) cells were purchased from Kerafast (Boston, MA, USA).
Live cells were counted using a LUNA-FL Dual Fluorescence Cell Counter and Acridine Orange/Propidium Iodide Stain, following the manufacturer’s instructions (Logos Biosystems, Anyang, Republic of Korea), throughout the study.
2.3. Flow Cytometry Analysis
Purified naïve T cells were stained with anti-CD8 (clone 53.67) and CD4 (clone RM4-5) antibodies conjugated with fluorescein isothiocyanate (FITC) and allophycocyanin (APC), respectively (Thermo Fisher Scientific), and run on a flow cytometer [FACSCaliber, Becton Dickinson Biosciences (BD), Franklin Lakes, NJ, USA] to examine their purity. When determining forward scatter (FSC), which reflects cellular size, and side scatter (SSC), which reflects granular/vesicular content, of various cell types, a 300 µL cell suspension in PBS with 0.1% FCS (FCM buffer) was prepared, and 0.3 µL of 10 mg/mL propidium iodide (Sigma Aldrich, St. Louis, MO, USA) was added to stain dead cells immediately before the acquisition of the data. The acquisition settings, including voltage and gain, were kept consistent across the cell types, allowing for comparison of the FSC and SSC profiles of live (propidium iodide negative) cells. All flow cytometry data were analyzed using the FlowJo software ver.10 (BD), including the calculation of mean FSC and mean SSC values for each sample, representing cell size and granular/vesicular content, respectively. The determined FSC and SSC values of the samples were further averaged across the replicates in each cell type, and standard deviations were calculated.
2.4. Total Protein Measurement of Cell Lysates
Two million cells were lysed with 200 µL of M-PER Mammalian Protein Extraction Reagent (Thermo Fisher Scientific), vortexed, and incubated on a shaker for 10 min. The samples were centrifuged at 6760× g for 5 min and the supernatants were collected. The sample was then subjected to a DC Protein Assay Kit (Bio-Rad, Hercules, CA, USA) to measure the total protein concentration using a spectrophotometer at 750 nm absorbance.
2.5. 89Zr-Oxine Production and Cell Labeling
89Zr-chloride was produced from
89Zr-oxalate generated at the cyclotron facility of the Clinical Center, National Institutes of Health, and was used to synthesize the
89Zr-oxine complex following a previously established protocol [
6,
27]. Briefly, 2 µL Tween 80 (20%), oxine (102 µL of 20 mmol/L in 0.04 N HCl), and
89Zr-chloride (60 µL, 25.9–40.5 MBq) were mixed and vortexed in a 1.5 mL tube, followed by neutralization using NaHCO
3 (500 mmol/L). The final solution pH ranged between 7 and 7.3.
To determine the
89Zr incorporation levels and labeling efficiencies in various cell types, cells in PBS were incubated with 37 kBq of
89Zr-oxine solution per 10
6 cells at a 30:1 volume ratio for 15 min. In select experiments, the incubation was shortened to 5 min to evaluate the kinetics of protein binding of
89Zr. After the incubation, 500 µL culture medium was added to the incubation mixture, centrifuged at 1900×
g for 2 min, and the supernatant was removed. The cells were washed twice with 800 µL culture medium, resuspended in 800 µL PBS, transferred to a new tube, and centrifuged again.
89Zr activity was measured during the incubation and after washing using a micro-dose calibrator built in-house [
28], and the activity incorporation yield (labeling efficiency) was calculated. To determine the
89Zr incorporation thresholds, 2 × 10
6 EL4, DC2.4, or Kit225K6 cells were incubated with increasing doses of
89Zr-oxine, aiming to label the cells at 18.5, 37, 74, 148, 296, and 592 kBq per 10
6 cells, based on the labeling efficiencies obtained above.
2.6. 111In-Oxine Cell Labeling
111In-oxine was purchased from GE Healthcare (Chicago, IL, USA, specific activity 59.2 kBq/mL). To determine the 111In incorporation levels, 2 × 106 EL4 and DC2.4 cells were incubated with 370 kBq per 106 cells in 100 µL PBS for 15 min at room temperature. After incubation, the samples underwent the same washing procedures as 89Zr-oxine labeled cells. The incubation and incorporated activities were measured using a dose calibrator (Capintec, Florham, NJ, USA).
2.7. Trichloroacetic Acid Protein Precipitation
To determine the intracellular protein binding of 89Zr-oxine or 111In-oxine, 2 × 106 labeled EL4, Kit225K6, or DC2.4 cells were lysed with 200 µL of M-PER Mammalian Protein Extraction Reagent (Thermo Fisher Scientific) immediately after labeling. PBS containing either 89Zr-oxine alone or 111In-oxine alone was used as a protein-free control in the relevant assays. One mL of 10% ice-cold trichloroacetic acid (TCA) in PBS was added to each lysate, incubated on ice for 10 min, and then centrifuged at 18,400× g for 5 min. The supernatants were separated from the pellets, and the activity in the supernatants (non-bound) and pellets (protein-bound) was measured using a γ-counter (Wizard 2, Perkin-Elmer, Shelton, CT, USA).
2.8. Cellular Fractionation and Western Blotting
Five million EL4, DC2.4, and Kit225K6 cells labeled with 89Zr-oxine (185–259 kBq/106 cells) underwent subcellular fractionation using a Subcellular Protein Fractionation Kit for Cultured Cells (Thermo Fisher Scientific), which separates the fractions with approximately 90% accuracy, according to the manufacturer’s instructions. Briefly, the cell pellet volume was estimated to be approximately 30 µL, and the cells were fractionated into cytoplasmic, membrane-bound, soluble nuclear, chromatin-bound, and cytoskeletal fractions in 300 µL Cytoplasmic Extraction Buffer, 300 µL Membrane Extraction Buffer, 150 µL Nuclear Extraction Buffer (NEB), 150 µL Chromatin-bound Extraction Buffer (NEB with 7.5 µL of 100 mM CaCl2 and 4.5 µL of Micrococcal Nuclease), and 150 µL Pellet Extraction Buffer, each containing protease inhibitors, respectively. The 89Zr activity in each cellular component was measured using a γ-counter (Perkin-Elmer). The quality of the fractionation was determined by western blot analysis by running 30 µL each of cytoplasmic and membrane-bound lysates and 15 µL each of soluble nuclear, chromatin-bound, and cytoskeletal lysates (i.e., 1/10th of each fractionated samples) mixed with 10 µL and 5 µL of 4× Laemmli Sample Buffer (Bio-Rad), respectively, on either 4–20% or 10% tris-glycine gels (Thermo Fisher Scientific). The electrophoresed samples were blotted against PVDF membranes (Immobilon-P, Millipore Sigma). A set of membranes was set aside for autoradiography. The PVDF membranes were blocked for non-specific binding with a PBS Blocking Buffer (Thermo Fisher Scientific) and probed with antibodies against histone H3 (clone 3H1), lysine-specific demethylase 1 (LSD1, clone C69G12), or vimentin (clone D21H3) purchased from Cell Signaling Technology for overnight at 4 °C, or with anti-Mek-1 (clone H-8, Santa Cruz Biotechnology, Dallas, TX, USA) or anti-calnexin (polyclonal, Enzo, Farmingdale, NY, USA) antibody for 1 h at room temperature. Then the membranes were washed using PBS with 0.1% Tween 20 for 1 h and incubated with a horseradish peroxidase-conjugated anti-rabbit antibody (1:20,000 dilution, Cytiva/GE Healthcare, Marlborough, MA, USA) or anti-mouse IgG (1:1000 dilution, Pierce, Appleton, WI, USA). The blots were developed using SuperSigna West Dura Extended Duration Substrate (Thermo Fisher Scientific), following the manufacturer’s instructions, and analyzed using a Chemi Doc imaging system (Bio-Rad). 111In-oxine labeled EL4 and DC2.4 cells (139 and 141 kBq/106 cells, respectively) were fractionated into subcellular compartments in a similar manner.
2.9. Autoradiography
PVDF membranes obtained from Western blotting of subcellular fractionated samples of the cells labeled with 89Zr-oxine or 111In-oxine were subjected to autoradiography. Phosphor screens (BAS-IP SR 2025, Cytiva/GE Healthcare) were exposed to the PVDF membranes for 7 days in cassettes and analyzed on a phosphor imager (Typhoon FLA 9000 Gel Scanner, Cytiva/GE Healthcare) at a 0.25 µm resolution.
2.10. Analysis of 89Zr Retention in Labeled Cells
Kit225K6 cells (5.6 × 106 cells) were labeled with increasing doses of 89Zr-oxine. Labeled cells were plated at 3.5 × 105 cells per 350 µL in triplicates. At each time point indicated, cell number and viability were examined using a LUNA-FL Dual Fluorescence Cell Counter and Acridine Orange/Propidium Iodide Stain, and cell-associated activity was measured with a γ-counter after centrifugation (1900× g for 2 min) and removal of the supernatants. The radioactivity results were decay corrected.
2.11. Biodistribution of 89Zr-Labeled and 111In-Oxine-Labeled Cells
Six million DCs (differentiated from the bone marrow collected from female mice) labeled with either 89Zr-oxine (29.6 kBq/106 cells) or 111In-oxine (76.0 kBq/106 cells) were intravenously administered to female recipient mice. One day and 7 days later, the mice were euthanized by CO2 inhalation, weighed, had blood withdrawn, and indicated organs and tissues were harvested. A femur was collected from each mouse as a bone/bone marrow sample. The collected blood and organs/tissues were weighed, and their radioactivity was measured using a γ-counter. The percent injected dose per gram of tissue (%ID/g), normalized to a 20-g mouse, was calculated using the following formula: %ID/g = [decay-corrected radioactivity of the tissue (cpm)]/[injected activity (cpm)]/[tissue weight (g)] × 100 × [body weight (g)]/20 (g), where cpm stands for count per minute.
2.12. Statistical Analysis
The statistical analysis was conducted using GraphPad Prism software (ver. 10.3.1, GraphPad Software). Correlation analyses were based on the Pearson r value. A two-tailed unpaired Student’s t-test was utilized to compare 1 variable between two groups. For the comparison of multiple groups involving 1 variable, an ordinary one-way analysis of variance (ANOVA) was employed, and for that involving 2 variables, a two-way ANOVA was used. For analyzing 2 variables over time, a repeated measure two-way ANOVA was used. Statistical significance was defined as p-values less than 0.05, where ‘n’ represents the number of replicates.
4. Discussion
Cell-based therapies using T cells or NK cells depend on the successful migration of the cells to a target organ or tissue. However, such cell tracking is difficult to perform non-invasively.
89Zr-oxine has emerged as the leading contender for ex vivo labeling of therapeutic cells to enable visualization and quantification of their distribution and migration kinetics in the body after infusion [
6,
7,
15,
18]. In this study, we examined how
89Zr-oxine distributes within cells and what influences its cellular retention to enable successful imaging by PET.
Our previous studies demonstrated that
89Zr-oxine can label many different cell types, albeit at different efficiencies [
6]. In this study, we demonstrated that the percent of
89Zr uptake (labeling efficiency) and activity incorporated positively correlated with the protein mass and size of the cell (r > 0.91).
89Zr-oxine demonstrated high cellular protein binding (>97%), accounting for the dependence on the protein mass of cells. Once labeled with
89Zr-oxine, breakdown of the
89Zr-oxine complex followed by binding of
89Zr to cellular proteins occurred relatively rapidly, and trapped the
89Zr radiometal inside the cells causing little (<3%) free
89Zr. Shortening the incubation time from 15 min to 5 min did not change the intracellular protein binding and distribution of
89Zr. This observation is supported by the known kinetics of neutral and lipid-soluble complexes [
29], of which the
89Zr-oxine complex is an example. The differences in the granular/vesicular contents of each cell type did not affect the uptake and retention of
89Zr-oxine, as the labeling relies on the membrane-permeable nature of the
89Zr-oxine complex [
6,
8].
The protein binding and intracellular distribution of
89Zr were compared to those of
111In-oxine labeling. In contrast to
89Zr-oxine,
111In-oxine labeled cells showed only 59–65% of activity binding to proteins, suggesting possible instability of the
111In complex within the cell. Of note, we used higher doses of
111In-oxine to label cells compared to
89Zr-oxine, because of the requirement of higher radioactive doses for imaging
111In by SPECT [
30]. If labeling saturation exists with
111In-oxine, similar to
89Zr, the high dose used for labeling might have lowered the percentage of intracellular protein-bound
111In. However, we did not observe meaningful
111In-bound proteins on autoradiography, indicating more unstable protein binding for
111In than
89Zr. Nevertheless. it was surprising that so many
89Zr-protein complexes survived the western blotting procedure and were detected by autoradiography in our study.
In
89Zr-oxine labeled cells, the majority of
89Zr was found in the cytoplasmic fraction, cytoplasmic and intracellular membrane fraction, and soluble nuclear fraction, but the distribution ratios differed among EL4, DC2.4, and Kit225K6 cells. We attribute this to differences in protein concentrations across various cell components among different cell types. EL4 cells have a relatively smooth surface and are round, as compared to DC2.4 cells, which are long and branching. DC2.4 thus provides a greater surface area for the membrane, explaining higher membrane protein labeling with
89Zr. Kit225K6 cells are small, irregular, and contain a significant number of vesicles, which helps explain the high binding of
89Zr to the proteins in the membrane. In contrast to
89Zr, the distribution of
111In after
111In-oxine cell labeling was more uniform across cell types (EL4 and DC2.4 cells), with 62–65% in the cytoplasm and 21–26% in the membrane. In a study of
111ln-oxine labeled erythrocytes of human or rat origin, 67% and 59% of activity was distributed to the cytosol, while the membranes retained 33% and 41% of activity, respectively [
23]. It is possible that the slight differences in
111In distribution between our results and the reported studies derived from the absence of nuclei in erythrocytes. We postulate that some
89Zr-bound intracellular proteins exchange within the cell, changing the intracellular distribution of
89Zr over time. This is true in the case of membrane-bound proteins, which can be internalized. Various transport proteins, such as transferrin or signaling molecules, commonly traffic between compartments. As virtually all of the
89Zr-oxine bound to intracellular proteins within 5 min of incubation,
89Zr probably does not readily bind to proteins newly generated after labeling. Although
89Zr alone does not permeate the plasma membrane, it is also possible that some
89Zr-bound proteins are excreted from the cell via exocytosis or as components of vesicles such as exosomes.
While
89Zr-oxine readily permeates cells for labeling, there appears to be a saturation point, which is cell-type-dependent. We observed plateauing of activity when high doses of
89Zr-oxine were added to the cell. As
89Zr uptake is dependent on cell size and protein mass, we believe that these factors influence the saturation point. This conclusion is supported by the lower saturation point observed for the smaller Kit225K6 cells (approximately 60 kBq/10
6 cells), as compared to EL4 and DC2.4 cells (approximately 90 kBq/10
6 cells) that are larger and have a greater protein mass. Our in vitro retention assay suggested that labeling cells below the saturation point improves the ability of the cells to retain
89Zr. In addition, we and others have demonstrated that the
89Zr-oxine activity that does not cause radiotoxicity to the cells is approximately 37 kBq/10
6 cells or below for many primary cell types [
6,
19], except for some radioresistant cells. The effects of
89Zr-oxine labeling on the cell viability and functions have been extensively evaluated in previous studies, as these are crucial steps for tracking cells using PET [
6,
14,
15,
16,
17,
18,
19,
20,
21,
31]. Keeping the specific activity of the cells below 37 kBq/10
6 cells would ensure better retention of
89Zr without inducing radiotoxicity.
In support of our in vitro findings of higher protein binding and labeling stability with
89Zr-oxine compared to
111In-oxine, in vivo infusion of DCs labeled with either tracer revealed significantly higher %ID/g values for
89Zr activity in the liver and spleen, the primary organs to which infused DCs home, compared to
111In. Very similar biodistribution results have been reported, showing higher retention of
89Zr than
111In in the liver and spleen following infusion of eGFP-5T33 murine myeloma cells labeled with
89Zr-oxine (150 kBq/10
6 cells) or
111In-oxine (340 kBq/10
6 cells) [
7]. Again, the higher activity requirement for
111In-oxine cell tracking using SPECT, compared to
89Zr-oxine that utilizes PET, may further accentuate the instability of
111In-oxine labeling. Additionally, several studies have reported radiotoxicity associated with
111In-oxine cell labeling, particularly in radiosensitive cells [
32,
33,
34]. Altogether, our findings suggest that
89Zr-oxine cell tracking using PET offers advantages over
111In-oxine SPECT imaging for detecting low concentrations of cells, with reduced risk of radiotoxicity.
This study has some limitations. First, we were not able to identify the exact intracellular proteins that bind
89Zr. Given the huge number of proteins present in cells, identifying
89Zr-bound protein(s) in electrophoreses gel samples was not possible. Our attempts to precipitate candidate proteins and examine if
89Zr was bound were not successful either. Second, we demonstrated that
89Zr-oxine cell labeling results in different subcellular distribution by cell type, and also when compared to
111In-oxine cell labeling, but the mechanisms causing these differences and whether these differences differentially affect cellular functions remain unknown. It is challenging to directly compare cellular functions, including trafficking, across different subcellular
89Zr distribution patterns in a cell type, as altering these patterns would require modifying the cells in ways that affect their functionality. One notable difference between
89Zr-oxine and
111In-oxine cell labeling is the higher nuclear localization observed with
89Zr. Both
89Zr and
111In emit Auger electrons capable of inducing DNA double-strand breaks [
35]. Do the differences in their intracellular distributions influence the extent of DNA damage? A previous study reported that
89Zr-oxine labeling at 6–20 kBq/10⁶ cells did not induce significant DNA damage in human γδ T cells, whereas labeling at 50–90 kBq/10
6 cells did [
19]. The same group later showed that human white blood cells labeled with
89Zr-oxine (32.9 ± 9.2 kBq/10⁶ cells) and
111In-oxine (52.8 ± 26.1 kBq/10⁶ cells) exhibited no significant difference in DNA double-strand breaks, although a variable dose of
111In was used [
31]. Further studies would be needed to address these questions.
In conclusion, we show that 89Zr-oxine cell labeling results in nearly complete protein binding in contrast to 111In-oxine in which only <65% is protein-bound. Protein mass and the size of cells were determining factors for the cellular incorporation of 89Zr. Intracellular protein binding of 89Zr can contribute to the stability of the label within the cell; however, this binding is saturable, and higher doses can destabilize binding. 89Zr-oxine labeled cells retained radioactivity at higher levels than 111In-oxine labeled cells following infusion to mice. These findings deepen our understanding of the mechanisms underlying 89Zr-oxine cell labeling and its stability both in vitro and in vivo. These characteristics of 89Zr-oxine support its utility for reliable long-term cell tracking with PET imaging and its clinical applications, such as monitoring cell-based therapies and investigating immune cell dynamics.