Abstract
Purpose
Glioblastoma multiforme (GBM) is a lethal cancer that responds poorly to therapy. GBM cancer-initiating cells have been shown to mediate resistance to both chemotherapy and radiation; however, it is unknown to what extent these cells contribute to the profound immune suppression in GBM patients and if strategies that alter their differentiation state can reduce this immune suppression.
Experimental Design
We isolated a subpopulation of cells from GBMs that possessed the capacity for self-renewal, formed neurospheres in vitro, were capable of pluripotent differentiation and could initiate tumors in vivo. These cells immune phenotype was characterized including the elaboration of immunosuppressive cytokines and chemokines by enzyme-linked immunosorbent assay. Functional immunosuppressive properties were characterized based on the inhibition of T cell proliferation and effector responses, triggering of T cell apoptosis and the induction FoxP3+ regulatory T cells. Upon altering their differentiation state, the immune suppressive phenotype and functional assays were reevaluated.
Results
We found that the cancer-initiating cells markedly inhibited T cell proliferation and activation, induced regulatory T cells and triggered T cell apoptosis that were mediated by B7-H1 and soluble Galectin-3. These immunosuppressive properties were diminished upon altering the differentiation of the cancer-initiating cells.
Conclusion
Cancer-initiating cells contribute to tumor evasion of the immune surveillance and approaches that alter the differentiation state may have immune therapeutic potential.
Keywords: Cancer-initiating cells, glioblastoma multiforme, immune suppression
STATEMENT OF TRANSLATIONAL RELEVANCE.
Many malignancies, and especially glioblastoma multiforme (GBM), have notable chemo- and radiation therapeutic resistance which is due, in part, to cancer-initiating cells capable of pluripotent differentiation and marked tumorigenesis. GBM patients are notable for profound immunosuppression and we hypothesized that GBM-associated cancer-initiating cells contribute to the immune suppression evident in these patients. This manuscript demonstrates that GBM-associated cancer-initiating cells play a key role in mediating immune suppression mechanistically by both cell-to-cell contact and secreted products, resulting in the inhibition T cell activation and proliferation, induction of regulatory T cells and the initiation of T cell apoptosis. These immune suppression properties are diminished upon altering the differentiation state of the cancer-initiating cells. Thus, we propose the novel concept that strategies that induce the altered differentiation state of cancer-initiating cells could be used to reverse immune suppression and as an immune therapeutic approach.
INTRODUCTION
Cancer-initiating cells are a heterogeneous population of undifferentiated cells with the capacity for self-renewal and a high proliferative potential. Treatments that are designed to eradicate tumors should also target the cancer-initiating cells (1). Glioblastoma multiforme (GBM), the most malignant among the adult human primary central nervous system tumors, contain these cancer-initiating cells that are multi-potent, and can recapitulate the characteristics of GBM including high motility, diversity of progeny, tendency to migrate along white matter tracts, and expression of immature antigenic phenotypes such as epidermal growth factor receptor (EGFR) and nestin (2). Cancer-initiating cells may express CD133 (3), although cancer-initiating cells have been identified that do not express CD133 (4–6), form neurospheres that are nonadherent, have marker characteristics for all three astrocytic, neuronal, and oligodendroglial lineages (7) and are tumorigenic in vivo. These cells are also believed to confer the resistance to chemotherapy and radiation observed in GBM patients (8, 9).
Malignant gliomas express tumor-associated and tumor-specific antigens that should make these tumors detectable to the immune system (10). However, there is a distinct lack of immune-mediated tumor eradication in glioma patients, and most attempts at immunotherapy have met with little clinical success (11). Many factors work in concert to inhibit anti-glioma immunity, including immunosuppressive cytokines such as IL-10, TGF-β, and prostaglandin E2 (PGE2), the induction of regulatory T cells, and down modulating co-stimulation molecules by antigen presenting cells (APCs) resulting in loss of T cell effector function - all of which that have been shown to be operational in GBM patients (Reviewed in ref 12) (12). Although central nervous system (CNS) tumors are recognized by the immune system, this is insufficient for their suppression or eradication. Primed CD8+ cytotoxic T cells gain CNS access (13); however, the lack of tumor eradication indicates that the T cells are functionally impaired within the local tumor microenvironment. It is unknown whether the cancer-initiating cells within the GBM participate in this tumor mediated immune suppression.
Only the immune suppressive properties of human mesenchymal stem cells isolated from normal human donors have been characterized to date and these cells have been used in clinical trials for treatment of graft-versus host disease (14). The mesenchymal stem cells typically have a spindle-shape morphology and grow as single cells, express CD105 and CD90 but not CD133 (15), differentiate into mesenchymal cells and are not tumorigenic in vivo but can enhance tumorigenesis of malignant cells (16). The mesenchymal stem cells express MHC I but not the co-stimulatory molecules (17). Additionally, the mesenchymal stem cells induce dendritic cells to secrete IL-10, increased the number of regulatory T cells, and decrease the secretion of IFN-γ from immune cells. This immune modulation is mechanistically attributed to elevated prostaglandin E2 (PGE2) levels and inhibitors of PGE2 mitigated the immune suppressive effects (18).
In order to understand the mechanism involved in glioma mediated immune suppression, we isolated cancer-initiating cells to ascertain if they possess similar immune suppressive properties that influence the GBM microenvironment. We hypothesized that the GBM-associated cancer-initiating cells, by either direct cell-to-cell contact perhaps by expressing the co-stimulatory inhibitory molecule B7-H1 (19, 20) and/or by secreted immunosuppressive cytokines or factors (21, 22) would induce T cell apoptosis (23–25) and/or the induction of regulatory T cells (26, 27) that would inhibit T cell proliferation. We then ascertained what would occur to the immune suppressive properties of cancer-initiating cells if we altered their differentiation state as a potential approach to overcome cancer-initiating cell-mediated immune suppression.
MATERIAL AND METHODS
Human glioma cell lines
Human normal astrocytes and glioma cell lines U-251 and U-87 were purchased from the American Type Culture Collection (Manassas, VA) and cultured in RPMI-1640 medium (astrocytes), modified Eagle’s medium (MEM) (U-251) or MEM plus 0.1 mM nonessential amino acids (U-87). To all media, 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin were added.
Ethical Treatment of Research Subjects and Patient Consent
Each patient provided written informed consent for tumor tissues and this study was conducted under protocol #LAB03-0687, which was approved by the institutional review board of The University of Texas M. D. Anderson Cancer Center.
Human tumors
Tumor tissues from newly diagnosed GBM patients (n=9) were obtained from surgery specimens and were graded pathologically according to the World Health Organization’s classification system by a neuropathologist.
Human glioma cancer-initiating cell derivation
GBM specimens were processed within 4 hours after resection. They were washed with DMEM-F-12 medium and disassociated as previously described (9). Briefly, the tissues were enzymatically digested with Papain dissociation system (Worthington Biomedical Corp. Lakewood, NJ). After a single-cell suspension was prepared, erythrocytes were lysed using 1x RBC lysis buffer (eBioscience, San Diego, CA). Trypan blue staining confirmed >80% cell viability. Dissociated tumor cells were cultured in DMEM-F-12 medium containing 20 ng/ml of EGF, basic fibroblast growth factor (Sigma, St. Louis, MO), and B27 (1:50; Invitrogen, Carlsbad, CA) as a neural stem cell-permissive medium (neurosphere medium) at a density of 3 × 106 cells/60-mm dish to form spheres. In parallel, single-cell suspensions from the GBM specimens were cultured in U-87 medium (MEM medium) with or without differentiation factors (10 ng/ml retinoic acid, and 20 ng/ml PDGF-AA). After primary sphere formation was noted, sphere cells were dissociated for characterization of their properties as GBM cancer-initiating cells such as immune phenotyping, cell self-renewal, differentiation and tumorigenesis.
Neurosphere formation
Dissociated primary sphere cells were plated at a density of 500 cells/well in 24-well plates in 0.8 ml volumes of neurosphere medium. After 2–6 days, the neurospheres were formed for all GBM specimens and the percentage of wells containing spheres ranged from 21% to 90%. And the formation of neurospheres was maintained for multiple passages in neurosphere medium.
Antibodies and reagents
Tissue culture grade monoclonal antibodies to CD3 (OKT3) and CD28 (28.6) were obtained from eBioscience. Anti-human IL-6 (1936) and anti-human TGF-β1 (27235) antibodies were obtained from R&D Systems (Minneapolis, MN). Blocking antibody to B7-H1 and related isotype control IgG1 were purchased from eBioscience. The cell surface was stained with PE, FITC, or allo-phycocyanin-conjugated antibodies against the following proteins: CD3, CD4, CD8, MHC I, MHC II, CD40, CD80, CD86, and B7-H1 (BD Pharmingen, San Diego, CA) and CD133 (Miltenyi Biotech, Auburn, CA). To detect intracellular cytokines, PE-conjugated antibodies against IL-2 and IFN-γ (R&D Systems) were used. Appropriate isotype controls were used for each antibody. Recombinant human Galectin-3 was obtained from R&D Systems.
ELISAs
Supernatants from the GBM tissue, the human glioma cell lines U-87 and U-251, and the glioma-associated cancer-initiating cells were measured for cytokine concentrations using ELISA kits as described (R&D Systems). These supernatants were collected from 3 × 106 cells after 5 days in culture and stored at −20°C. The supernatants were added in duplicate to appropriate pre-coated plates. After the plates were washed, horseradish peroxidase-conjugated detection antibody was added. The substrate used for color development was tetramethylbenzidine. The optical density was measured at 450 nm with a microplate reader (Spectra Max 190; Molecular Devices, Sunnyvale, CA), and chemokine concentrations were quantitated with SoftMax Pro software (Molecular Devices). The detection limits for CCL-2 were 5 pg/ml; TGF-β1, 16 pg/ml; IL-10, 5 pg/ml; IL-6, 1 pg/ml; PGE2, 10 pg/ml; VEGF, 5 pg/ml; Galectin-3, 10 pg/ml; and soluble Fas, 10 pg/ml.
Human PBMCs
PBMCs were prepared from healthy donor blood (Gulf Coast Blood Center, Houston, TX) and GBM patients’ blood (the same patients whose cancer-initiating cells were isolated by centrifugation on a Ficoll-Hypaque density gradient (Sigma-Aldrich, St. Louis, MO)). Aliquots of the isolated PBMCs were frozen and stored at −180°C until use. Prior to co-culture experiments, frozen PBMCs were thawed at 37°C for 5 min and then washed with warm 10% FBS in RPMI 1640 medium. CD3+ T cells were purified from PBMCs by negative selection using a Pan T Cell Isolation Kit II (Miltenyi Biotech, Auburn, CA), according to the manufacturer’s instructions.
Flow cytometry
FITC-conjugated anti-CD4 (RPA-T4) and APC-conjugated anti-CD8 (RPA-T8) antibodies were used for cell surface staining. Sub-analysis of the T cell populations was based on the gated surface expression of CD4 and CD8. To detect FoxP3 protein expression, the surface stained cells were further subjected to intracellular staining with PE-conjugated monoclonal antibodies to human FoxP3 (clone PCH101, eBiosciences) using staining buffers and conditions specified by the manufacturer. For intracellular cytokine staining, cells were stimulated for 6 hours in the presence of 50 ng/ml phorbol myristate acetate (PMA), 500 ng/ml ionomycin (Sigma-Aldrich), and 2 μM monensin (GolgiStop, BD Sciences). Then the cells were incubated with FITC-conjugated anti-CD4 and APC-conjugated anti-CD8 (RPA-T8) antibodies for surface staining followed by intracellular staining using PE-conjugated anti-mouse IFN-γ (4S.B3) or PE-conjugated anti-mouse IL-2 (MQ1-17H12) antibodies and FIX/PERM buffers (BD Pharmingen) according to the manufacturer’s instructions. Flow cytometry acquisition was done with a FACSCaliber (Becton Dickinson, San Diego, CA) and data analysis was with FlowJo software (TreeStar, Ashland, OR).
Cell proliferation assay and regulatory T cell induction assay
The glioma-associated cancer-initiating cells and the differentiated glioma cells in the U-87 medium were plated into 48-well plates (3 × 104 cells/ml) containing 3 × 105 PBMCs/ml n the presence of 1 μg/ml pre-bound anti-CD3/anti-CD28 antibodies or 2.5 μg/ml phytohemagglutinin (PHA, Sigma-Aldrich) with and without the B7-H1 (10μg/ml) blocking antibody Alternatively, conditioned media from the glioma associated cancer-initiating cells were also added to the stimulated PBMCs. After 72 hours, 100 μl of cells from each well was transferred to new 96-well plates with 10 μl of Cell Counting Kit-8 (Dojindo Laboratories, Rockville, MD). After incubation for 4 hours at 37°C, absorbance was measured at 450 nm with a microplate reader (Spectra Max 190). To detect FoxP3+ regulatory T cells, CD4 surface staining and then FoxP3 intracellular staining were performed on immune cells cultured for 96 hours.
FoxP3+ regulatory T cell functional assay
Healthy donor PBMCs were labeled with 2 μM carboxyfluorescein diacetate succinimidyl ester (CFSE) for 5 min at room temperature in PBS with 0.1% bovine serum albumin, and then the reaction was quenched with RPMI 1640 medium with 10% FBS for 10 min at 37°C. 1 × 106/ml CFSE-labeled PBMCs, and 1 × 106/ml autologous T cells which were cultured with conditioned media from glioma-associated cancer-initiating cells for 4 days, were plated into 96-well plates in the presence of 2 × 106/ml allogeneic irradiated PBMCs in RPMI 1640 medium with10% FBS in a total volume of 0.2 ml. After 72 hours, the cells were harvested, and analysis of cell division was performed by flow cytometry.
Apoptosis assay
The T cell apoptosis assay was performed with the Annexin V/7-AAD staining kit (BD Pharmingen). Healthy donors PBMCs were cultured for 5 days with medium, glioma associated cancer-initiating cells supernatants or human Galactin-3 at 1 and 10 ng/mls and then harvested by centrifugation. Additionally, autologous GBM patient PBMCs were co-cultured with their respective glioma associated cancer-initiating cells with and without anti-B7-H1 blocking antibody (10 ug/ml) at the beginning of the culture conditions in the cell-contact dependent apoptotic assay. The cells were stained with APC-conjugated anti-CD3 antibodies and then washed twice with cold PBS and resuspended in 1× binding buffer (BD Pharmingen) at a concentration of 1 × 106 cells/ml. Next, PE-conjugated annexin V and 7-AAD were added, the cells were incubated for 20 min at 25°C in the dark, and CD3+ T cell apoptosis was analyzed by flow cytometry within 1 hour.
Cloning of single cell cancer-initiating cells
After confirmed with marked capacity for self-renewal, differentiation and tumor formation at a low cell number, accutase (Sigma, St. Louis, MO) dissociated cancer-initiating cells were sorted using the CD133 cell isolation kit (Miltenyi Biotech, Auburn, CA), and > 90% purity by FACS. CD133+ sorted cells were seeded into 96-well plates at a theoretical density of 1 cell per well. After overnight culture, microscopic observation was utilized to identify wells that contained a single cell. These wells were monitored and the medium changed every 5–7 days for 45 days before immune functional analysis. In vivo tumorigenic potential were confirmed by formation of lethal tumor after intracranial implantation into nude mice.
Alteration of differentiation state of cancer-initiating cell
Accutase-dissociated sphere cells were cultured in differentiation medium consisting of 10% FBS, 10 ng/ml retinoic acid, and 20 ng/ml PDGF-AA (both from Sigma-Aldrich). Confluent monolayer cells were detached every 5–7 days by trypsinization, and retinoic acid and PDGF-AA were replenished during the culture. Similarly, the U-87 differentiated medium (MEM medium supplemented with 10 ng/ml retinoic acid and 20 ng/ml PDGF-AA) is utilized to differentiate total GBM cells.
Immunohistochemistry
Differentiated cancer-initiating cells were cultured on eight-chamber slides (Nunc, Rochester, NY) at 5,000/well. After 3 days, cells were fixed with 4% paraformaldehyde, permeabilized with 3% Triton X-100 in PBS, and then blocked with 5% horse serum. Primary antibodies were rabbit anti-GFAP (1:40; Dako, Golstrup, Denmark), mouse anti-GalC (1:100; Chemicon, Ramona, CA), and mouse anti-MAP2 (1:50; Chemicon). After incubation for 90 min, the slides were washed with 5% horse serum. Secondary antibodies, goat anti-rabbit Alexa 546 (1:300; Invitrogen) and donkey anti-mouse Alexa 488 (1:300; Invitrogen), were added for 30 min. Slides were mounted using Vectashield Hard Set mounting medium with DAPI (Vector Laboratories, Burlingame, CA).
Intracranial xenografting of cancer-initiating cells
Single cell suspensions of glioma associated cancer-initiating cells in serum-free medium at 1 × 103 cells per 5 μL were injected into the right frontallobes of 5–8-week-old nude mice (MDACC, Houston) using a stereotactic frame system (Kopf Instruments, Tujunga, CA) as previously described (28). Animals were anesthetized with xylazine/ketamine during the procedure. Mice were maintained in the M. D. Anderson Isolation Facility in accordance with Laboratory Animal Resources Commission standards and conducted according to an approved protocol, 08-06-11831.
Statistical analysis
All values were calculated as means and 95% confidence intervals (CIs) from at least three independent experiments The Student t test was used to test for differences in the means between two groups. P values less than 0.05 were considered to be statistically significant. All statistical analyses were performed using the Statistical Package for the Social Sciences v.12.0.0 (SPSS, Chicago, IL). Error bars represent s.d.
RESULTS
Characterization of glioma associated cancer-initiating cells
From newly diagnosed GBM patients (n=9) at the time of surgery, we isolated glioma associated cancer-initiating cells and the patients’ autologous T cells. The glioma associated cancer-initiating cells from the patients expressed CD133 (range 3–76%, mean 32%, data not shown), formed neurospheres (Fig. 1A) in serum-free medium containing EGF and bFGF after 5–10 days of culture and were capable of differentiating into glial fibrillary acidic protein (GFAP+) astrocyte-like cells, neuron-like cells that were immunoreactive for microtubule associated protein 2 (MAP2), and galactosylceramidase (GalC)-immunoreactive oligodendrocyte-like cells (Fig. 1B). Furthermore, when the glioma associated cancer-initiating cells (n=3; 1000 cells per mouse; 6 mice per glioma associated cancer-initiating cells line) were injected in the right frontal lobes of 5–8-week-old nude mice, the mice developed tumors that were highly infiltrative along white matter tracts--a characteristic of human GBM (Fig. 1C). After confirmation of their capacity for self renewal and recapitulation of the original tumor, the isolated glioma associated cancer-initiating cells were utilized for the characterization of their immune properties.
Immunological phenotype of glioma associated cancer-initiating cells
To characterize their immunological phenotype, the glioma associated cancer-initiating cells (n=5) were assessed for their expression of MHC I, MHC II, CD40, CD80, CD86, and B7-H1 by flow cytometry. The glioma associated cancer-initiating cells expressed high levels of MHC I (mean 99.3%, range 98.5–99.8%) and low levels of CD86 (mean 6.7%, range 5.9–7.9%) and CD40 (mean 5.8%, range 0.7–15.8%), but not MHC II (mean 2.4%, range 1.6–3.2%) or CD80 (mean 0.6%, range 0.2–0.6%) (a representative example is shown in Fig. 2A), indicating that glioma associated cancer-initiating cells lack the capacity for antigen presentation necessary to stimulate T cell activation or proliferation. Furthermore, the inhibitory co-stimulatory molecule B7-H1 (mean 31.2%, range 28.5–34.9%) was expressed indicating that direct contact between T cells and glioma associated cancer-initiating cells would be inhibitory on immune cells.
Glioma associated cancer-initiating cells produce immunosuppressive cytokines
To determine if the glioma associated cancer-initiating cells produce immunosuppressive cytokines, glioma associated cancer-initiating cells (n=4) were assayed for immunosuppressive cytokines by ELISA. The glioma associated cancer-initiating cells did not produce any appreciable IL-6, IL-10, soluble Fas, or TRAIL but did produce TGF-β1 (24–73.8 pg/106 cells/24 hours), the regulatory T cell chemokine attractant CCL-2 (8–710 pg/106 cells/24 hours), VEGF (14–61 pg/106 cells/24 hours) and PGE2 (34–60 pg/106 cells/24 hours).
Glioma associated cancer-initiating cells inhibit T cell activation and proliferation
To determine if the glioma associated cancer-initiating cells produce factors that would inhibit the activation and subsequent proliferation of immune cells, peripheral blood mononuclear cells (PBMCs) from healthy donors were activated with anti-CD3/CD28 or phytohemagglutinin (PHA) in the presence of conditioned medium obtained from 3-day cultures of glioma associated cancer-initiating cells and T cell proliferation was assessed by flow cytometry. The media from a representative glioma associated cancer-initiating cell was capable of inhibiting T cell proliferation (Fig. 2B). This inhibition was seen regardless of the mechanism of stimulation, i.e., anti-CD3/CD28 or PHA; however, no inhibition of T cell proliferation was detected when the conditioned medium was obtained from normal human astrocytes or the U-87 cell line (Table 1). To further demonstrate that individual glioma-associated cancer-initiating cells were capable of clonogenic growth and immunosuppression, CD133+ cells were sorted from neurospheres and diluted for single colony formation. Over 80% of seeded single cells grew out and ten clones from two different neurospheres were selected at random and expanded for further immunological characterization. As shown in Table 1, conditioned media of all clonogenic lines displayed potent immunosuppression on CD3+ T cells of normal donors, which potently inhibited T cell proliferation by 91 ± 12% (p=0.0005).
Table 1.
Neurosphere Parental Line | Clone number | CD133+ (%) | CFSE labeled dividing T cells (%) | Change compared to medium (%) | Apoptotic T cells (%) | Change compared to medium (%) | Foxp3+ Tregs (%) | Change compared to medium (%) |
---|---|---|---|---|---|---|---|---|
6–15 | 1 | 25.9 | 6.6 | ↓92 | 60.7 | ↑163 | 40 | ↑205 |
2 | 88.9 | 3.8 | ↓95 | 48.9 | ↑112 | 27.8 | ↑112 | |
3 | 84.5 | 3.2 | ↓96 | 47.5 | ↑106 | 31.7 | ↑142 | |
4 | 47 | 7.2 | ↓91 | 55.8 | ↑142 | 35.9 | ↑174 | |
5 | 34.6 | 46.7 | ↓42 | 51.7 | ↑124 | 36.1 | ↑176 | |
6 | 89.6 | 3.1 | ↓96 | 53.4 | ↑131 | 24.4 | ↑86 | |
7 | 86.3 | 3.5 | ↓96 | 55.6 | ↑141 | 35.7 | ↑173 | |
8 | 84.7 | 5.7 | ↓93 | 61.1 | ↑165 | 44 | ↑236 | |
9 | 64 | 3.7 | ↓96 | 66.5 | ↑188 | 29 | ↑121 | |
10 | 70.2 | 4 | ↓95 | 67.3 | ↑191 | 31.7 | ↑142 | |
11–28 | 1 | 97.5 | 4.1 | ↓95 | 47.8 | ↑107 | 24.2 | ↑85 |
2 | 93.5 | 4.8 | ↓94 | 64.1 | ↑177 | 18.6 | ↑42 | |
3 | 76 | 5.6 | ↓93 | 54.5 | ↑136 | 28.7 | ↑119 | |
4 | 68.4 | 11.2 | ↓86 | 51.5 | ↑123 | 21.6 | ↑65 | |
5 | 93.6 | 5.6 | ↓93 | 55.9 | ↑142 | 21.6 | ↑65 | |
6 | 91.1 | 1.4 | ↓98 | 70 | ↑203 | 11.8 | ↓10 | |
7 | 81.5 | 5.2 | ↓94 | 54.5 | ↑136 | 29.8 | ↑127 | |
8 | 96.5 | 4.3 | ↓95 | 52.2 | ↑126 | 28.3 | ↑116 | |
9 | 96.2 | 6.1 | ↓92 | 50.6 | ↑119 | 29.6 | ↑126 | |
10 | 81.1 | 3.9 | ↓95 | 57.6 | ↑149 | 13.3 | ↑1.5 | |
Control (cancer- initiating) medium | None | None | 81 | --- | 23.1 | --- | 13.1 | --- |
U-87 | None | None | 55.2 | ↓12 | 15.7 | ↓10 | 8.9 | ↓8 |
U-87 medium | None | None | 63.3 | --- | 17.4 | --- | 9.7 | --- |
U-87 cells propagated in cancer-initiating medium | None | None | 82.9 | ↑2 | 21.8 | ↓6 | 14.5 | ↑11 |
Astrocyte | None | None | 42.7 | ↓8 | 10.9 | ↓7 | 10.6 | ↑36 |
Astrocyte medium | None | None | 46.4 | --- | 11.7 | --- | 7.8 | --- |
U-87 medium * | None | None | 34.1 | --- | 29.3 | --- | 11.5 | --- |
GBM cell suspension in U-87 medium* | None | --- | 14.8 | ↓56 | 51.2 | ↑78 | 22.7 | ↑97 |
GBM cell supsension in U-87 differentation medium* | None | --- | 28.7 | ↓16 | 33.8 | ↑15 | 10.9 | ↓5 |
contact-dependent co-culture with autologuous PBMCs
Co-culture experiments with matched autologous PBMCs and the glioma associated cancer-initiating cells also demonstrated inhibition of T cell proliferation (Fig. 2C), and this inhibition was partially reversed by the addition of B7-H1 blocking antibody in autologous co-culture assay, indicating B7-H1 mediated cell-to-cell contact is a mechanism contributing to cancer-initiating cells mediated inhibition of T cell proliferation (Fig. 2D). To further characterize this inhibition of immune cell proliferation, the glioma associated cancer-initiating cells supernatants were co-incubated with healthy donors’ PBMCs in the presence of anti-CD3/CD28 stimulation, and the percentages of CD4+ and CD8+ T cells producing IL-2 and interferon γ (IFN-γ) effector cytokines were determined by intracellular staining via flow cytometry. The number of IFN-γ and IL-2 producing CD4 and CD8 T cells were both reduced by 30% and 10%, respectively, by the cancer-initiating cell supernatants (n=3). Similarly, the glioma associated cancer-initiating cells inhibited IFN-γ and IL-2 generation by autologous CD3+ T cells in cell-to-cell contact experiments (Fig. 2E). These data demonstrate that glioma associated cancer-initiating cells suppress T cell proliferative and pro-inflammatory responses.
Glioma associated cancer-initiating cells induce regulatory T cells
To determine if the decrease in T cell proliferation and effector response was secondary to the TGF-β producing glioma associated cancer-initiating cells inducing regulatory T cells, we ascertained the ability of the cancer-initiating cells to induce regulatory T cells. Incubation with supernatants from the glioma associated cancer-initiating cells markedly expanded the number of CD4+FoxP3+ regulatory T cells in healthy donor PBMCs by 128 ± 51 % (p=0.0007) (Table 1; representative example in Fig. 3A). These Foxp3+ regulatory T cells are functionally suppressive on autologous T cell proliferation (Fig. 3B). Furthermore, the glioma associated cancer-initiating cells induced FoxP3+ regulatory T cells in co-culture experiments with PBMCs from each respective autologous GBM patient (Fig. 3C). In this cell-to-cell contact context, addition of B7-H1 neutralizing antibody partially blocked the induction of FoxP3+ regulatory T cells (Fig. 2D), indicating that not only secreted factor(s) but also a cell-to-cell contact mechanism mediated by B7-H1 on the surface of cancer-initiating cells play a role in cancer-initiating cell mediated immune suppression. Conditioned media from the U-87 glioma cell line did not induce regulatory T cells but the media from astrocytes could but to a much lesser degree than the glioma associated cancer-initiating cells (Table 1).
Glioma associated cancer-initiating cells induce T cell apoptosis
The glioma associated cancer-initiating cells supernatants were able to induce immune cell apoptosis in healthy donor PBMCs (Fig 4A). All supernatants of the clonogenic cancer-initiating cells (n=20) were able to increase immune cell apoptosis by 144 ± 29% (p=0.0001) in healthy donor PBMCs (Table 1). Furthermore, when GBM patients’ PBMCs were co-incubated with the respective patients’ glioma associated cancer-initiating cells, as predicted from the phenotypic expression of B7-H1 on glioma associated cancer-initiating cells, both pre-apoptosis and apoptosis were induced in the immune cells (Fig. 4B), which was partially rescued by B7-H1 blockade (Fig. 4C), indicating that B7-H1 induces T cell apoptosis by cell-to-cell contact with the cognate receptor on the T cells. Conditioned media from normal human astrocytes and the U-87 glioma cell line did not induce T cell apoptosis (Table 1). Activated immune cells also underwent apoptosis when co-cultured with conditioned medium from glioma associated cancer-initiating cells, indicating that activation did not protect immune cells from the apoptosis induced by conditioned medium from glioma associated cancer-initiating cells (data not shown). This indicates that glioma associated cancer-initiating cells can mediate immunosuppression by apoptotic elimination of immune cells, regardless of their activation state, likely by both secretion of product(s) and direct cell-to-cell contact.
Immunosuppressive properties of glioma associated cancer-initiating cells are lost upon alteration of their differentiation state
We next investigated whether the glioma associated cancer-initiating cells could mediate immunosuppression after altering their state of differentiation. The glioma associated cancer-initiating cells were exposed to differentiating medium (29) resulting in all of the glioma associated cancer-initiating cells lines having alterations in cell morphology and increased expression of astrocytic (GFAP+), neuronal (MAP2+), and oligodendroglial (GalC+) lineage markers (Fig. 1B). In addition, the expression level of CD133 was reduced after exposure to differentiation medium (mean: 5%; range 0–11%, with one representative shown in Fig. 5A).
Conditioned media from the more differentiated glioma associated cancer-initiating cells were then harvested, and the immunosuppressive properties were evaluated. We found that the inhibition of immune cell proliferation (n=3) by conditioned media from the glioma associated cancer-initiating cells was reversed upon altering differentiation (Fig. 5B). This was likely secondary to the fact that fewer FoxP3+ regulatory T cells were induced (Fig. 5C) and the T cell apoptosis was diminished in the presence of conditioned media from the more differentiated glioma associated cancer-initiating cells (Fig. 5D). Furthermore, single cell suspensions of glioma cells isolated from GBM specimens that would contain the sub-population of cancer-initiating cells were found to be immune suppressive, which was lost upon inducing differentiation (Table 1).
Galectin-3 secreted by glioma associated cancer-initiating cells mediates induction of T cell apoptosis
To ascertain the mechanism for the reduced T cell apoptosis after differentiation of cancer-initiating cells, the supernatants from the cancer-initiating cells before and after differentiation and U-87 were assessed for Galectin-3 by ELISA. Before differentiation the mean Galectin-3 produced by the cancer-initiating cells (n=3) was 468 ± 77, 179 ± 10, 139 ± 8 pg/106 cells/24 hours and after inducing an altered differentiated state this was reduced to 18 ± 2, 41 ± 5 and 17± 2 pg/106 cells/24 hours. U-87 cells did not produce Galectin-3. Next, recombinant Galectin-3 was added to the T cell cultures to demonstrate that it is directly responsible for the immune cell apoptosis. At physiological doses of Galectin-3 (1.5~3.2 ng/ml as determined by ELISA from the conditioned media from cancer-initiating cells) there was dose response induced T cell apoptosis (Fig. 4D). Similar doses of Galectin-3 did not affect T cell proliferation or induce FoxP3+ Regulatory T cell induction (data not shown) suggesting multiple factors and mechanisms are involved in the immune suppression mediated by glioma associated cancer-initiating cells.
DISCUSSION
The immunological properties of human cancer-initiating cells have not been defined previously, and to our knowledge this is the first study to show that these cells mediate many of the key features of immunosuppression and explains a possible mechanism for resistance to immunotherapy in the clinic. To investigate the immune properties of glioma associated cancer-initiating cells, we used two different experimental approaches. In the first approach, the supernatants from glioma associated cancer-initiating cells were used in immunological assays with T cells from healthy donors to determine the effects of glioma associated cancer-initiating cells in the absence of pre-existing T cell immunosuppression while avoiding allogeneic responses that could confound the interpretation of the data. In the second approach, using GBM patients’ T cells and the respective patients’ glioma associated cancer-initiating cells, allogeneic interactions would not confound the data, allowing for analysis of direct cell-to-cell contact; however, pre-existing immune suppression in the patient T cells and secreted factor(s) from autologous cancer-initiating cells might dampen the extent of cell-to-cell contacting immunosuppression exerted by the glioma associated cancer-initiating cells. Regardless of the experimental approaches, the data consistently demonstrated that the glioma associated cancer-initiating cells mediate immunosuppression by several redundant mechanisms
In this study, although the glioma associated cancer-initiating cells expressed MHC I, they lacked MHC II, CD40, and CD80, which would be anticipated to induce T cell anergy (30) and this was confirmed in our functional assays of T cell proliferation. Of note, this immune phenotype was homogeneous across the glioma associated cancer-initiating cells from various GBM patients regardless of CD133 expression. Although the immune phenotype of the cancer-initiating cells contribute, in part, to the T cell immune suppression, other mechanisms such as the expression of B7-H1 and the secretion of Galactin-3 and TGF-β also play significant roles. The glioma associated cancer-initiating cells expressed the co-stimulatory inhibitory molecule B7-H1, which was previously demonstrated to be a key factor mediating immune resistance in gliomas (19) and induces T cell apoptosis (31). It was therefore not unexpected to find in the direct cell-to-cell contact experiments that the glioma associated cancer-initiating cells induced T cell apoptosis in the scenario of B7-H1 expression. We also found that a cell-secreted product(s) were mediating T cell apoptosis; however the exact factor eluded determination until we conducted a human antibody microarray to evaluate those factors whose expression was lost upon inducing an altered differentiated state which included the candidate Galectin-3. Soluble Galectin-3 has been shown to induce T cell apoptosis (32), is constitutively expressed in glioma cell lines but not normal astrocytes or oligodendrocytes (33), and has been shown to enhance glioma proliferation and migration (34). Thus, the secretion of Galectin-3 appears to be a feature of nonterminally differentiated cells, which can be reduced, along with the immune suppressive properties, upon altering the differentiation state. Additionally, it was not entirely surprising to see that cancer-initiating cells could induce regulatory T cells that were functionally active since the cancer-initiating cells did elaborate modest levels of TGF-β and this amount (10–100pg/ml) of TGF-β that have been shown previously to be sufficient for regulatory T cell induction (35). Of note, the glioma associated cancer-initiating cells did not elaborate NO as a possible mechanism for regulatory T cell induction (unpublished data). Cumulatively, our data indicate that in addition to the previously identified key role of glioma associated cancer-initiating cells in radiation resistance (9) and chemotherapy resistance (3, 36, 37), glioma associated cancer-initiating cells also contribute to immunosuppression.
Adult human stem cells have been shown to have potential clinical applications for a variety of degenerative diseases and for organ tissue replacement (38). As far as we are aware, we are the first group to propose altering the differentiation state of cancer-initiating cells as an approach to reverse immune suppression that could be utilized in an immune modulation/therapeutic context. In this manuscript we demonstrated that the cancer-initiating mediated immune suppression can be reversed by altering the differentiation state. However, our findings pertaining to the reversal of immune suppression upon altering the differentiation state are in contrast to the studies of Le Blanc et al. who found there was no change in alloreactive lymphocyte proliferative responses between differentiated and undifferentiated mesenchymal stem cells (39). The differences between allogeneic versus autologous T cell proliferation assays, the final differentiated cell phenotype, or a fundamental differences in the ability to reverse immune suppression between these cell types may account for the differences in our findings compared to the previous study. Strategies that force glioma associated cancer-initiating cells into a more differentiated phenotype (40) such as agents that block the signal transducer and activator of transcription pathway (41) may actually be promising agents as an immune therapeutic approach (42). Induction of differentiation as a therapeutic strategy has been used with success in promyelocytic leukemia with all-trans retinoic acid (43) and many new compounds have been identified based on the isoquinoline sulfonamide scaffold that have been shown to induce differentiation in stem cells that may also be of clinical utility (44). Ultimately, the optimal agent would only induce differentiation in the cancer-initiating cells without effecting normal somatic stem cells. Further studies will be necessary to ascertain what type of differentiation agents would be optimal in combination with other immune therapeutics.
Acknowledgments
We thank Lamonne Crutcher for assistance in obtaining tissue specimens and Melissa Burkett and Adelina “Keats” Fuentes for editorial assistance.
Financial Support: This work was supported by the Anthony Bullock III Foundation (ABH), the Dr. Marnie Rose Foundation (ABH), The University of Texas M. D. Anderson Cancer Center (ABH), and the U.S. National Institute of Health (CA120813-01) (ABH). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Footnotes
Parts of these data were presented at the American Society of Clinical Oncology Annual Meeting, May 2008, Chicago, IL and AACR Special Conference in Cancer Research: Tumor Immunology, December 2008, Miami, FL.
References
- 1.Dingli D, Michor F. Successful therapy must eradicate cancer stem cells. Stem Cells. 2006;24:2603–10. doi: 10.1634/stemcells.2006-0136. [DOI] [PubMed] [Google Scholar]
- 2.Sanai N, Alvarez-Buylla A, Berger MS. Neural stem cells and the origin of gliomas. N Engl J Med. 2005;353:811–22. doi: 10.1056/NEJMra043666. [DOI] [PubMed] [Google Scholar]
- 3.Liu G, Yuan X, Zeng Z, et al. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer. 2006;5:67–79. doi: 10.1186/1476-4598-5-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sun Y, Kong W, Falk A, et al. CD133 (Prominin) negative human neural stem cells are clonogenic and tripotent. PLoS One. 2009;4:e5498. doi: 10.1371/journal.pone.0005498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Shmelkov SV, Butler JM, Hooper AT, et al. CD133 expression is not restricted to stem cells, and both CD133+ and CD133− metastatic colon cancer cells initiate tumors. J Clin Invest. 2008;118:2111–20. doi: 10.1172/JCI34401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bleau AM, Hambardzumyan D, Ozawa T, et al. PTEN/PI3K/Akt pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell. 2009;4:226–35. doi: 10.1016/j.stem.2009.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Galli R, Binda E, Orfanelli U, et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004;64:7011–21. doi: 10.1158/0008-5472.CAN-04-1364. [DOI] [PubMed] [Google Scholar]
- 8.Murat A, Migliavacca E, Gorlia T, et al. Stem cell-related “self-renewal” signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol. 2008;26:3015–24. doi: 10.1200/JCO.2007.15.7164. [DOI] [PubMed] [Google Scholar]
- 9.Bao S, Wu Q, McLendon RE, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–60. doi: 10.1038/nature05236. [DOI] [PubMed] [Google Scholar]
- 10.Kurpad SN, Zhao XG, Wikstrand CJ, Batra SK, McLendon RE, Bigner DD. Tumor antigens in astrocytic gliomas [Review] Glia. 1995;15:244–56. doi: 10.1002/glia.440150306. [DOI] [PubMed] [Google Scholar]
- 11.Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–15. doi: 10.1038/nm1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dey M, Hussain SF, Heimberger AB. The role of glioma microenvironment in immune modulation: potential targets for intervention. Lett Drug Des Discov. 2006;3:443–51. [Google Scholar]
- 13.Hickey WF, Hsu BL, Kimura H. T-lymphocyte entry into the central nervous system. J Neurosci Res. 1991;28:254–60. doi: 10.1002/jnr.490280213. [DOI] [PubMed] [Google Scholar]
- 14.Le Blanc K, Frassoni F, Ball L, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371:1579–86. doi: 10.1016/S0140-6736(08)60690-X. [DOI] [PubMed] [Google Scholar]
- 15.Vogel W, Grunebach F, Messam CA, Kanz L, Brugger W, Buhring HJ. Heterogeneity among human bone marrow-derived mesenchymal stem cells and neural progenitor cells. Haematologica. 2003;88:126–33. [PubMed] [Google Scholar]
- 16.Djouad F, Plence P, Bony C, et al. Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood. 2003;102:3837–44. doi: 10.1182/blood-2003-04-1193. [DOI] [PubMed] [Google Scholar]
- 17.Majumdar MK, Keane-Moore M, Buyaner D, et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci. 2003;10:228–41. doi: 10.1007/BF02256058. [DOI] [PubMed] [Google Scholar]
- 18.Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105:1815–22. doi: 10.1182/blood-2004-04-1559. [DOI] [PubMed] [Google Scholar]
- 19.Parsa AT, Waldron JS, Panner A, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007;13:84–8. doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
- 20.Wintterle S, Schreiner B, Mitsdoerffer M, et al. Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res. 2003;63:7462–7. [PubMed] [Google Scholar]
- 21.Roszman T, Elliott L, Brooks W. Modulation of T-cell function by gliomas [Review] Immunol Today. 1991;12:370–4. doi: 10.1016/0167-5699(91)90068-5. [DOI] [PubMed] [Google Scholar]
- 22.Tilg H, Trehu E, Atkins MB, Dinarello CA, Mier J. Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55. Blood. 1994;83:113–8. [PubMed] [Google Scholar]
- 23.Kim R, Emi M, Tanabe K, Uchida Y, Toge T. The role of Fas ligand and transforming growth factor beta in tumor progression: molecular mechanisms of immune privilege via Fas-mediated apoptosis and potential targets for cancer therapy. Cancer. 2004;100:2281–91. doi: 10.1002/cncr.20270. [DOI] [PubMed] [Google Scholar]
- 24.Fanger NA, Maliszewski CR, Schooley K, Griffith TS. Human dendritic cells mediate cellular apoptosis via tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) J Exp Med. 1999;190:1155–64. doi: 10.1084/jem.190.8.1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hegardt P, Widegren B, Sjögren HO. Nitric-oxide-dependent systemic immunosuppression in animals with progressively growing malignant gliomas. Cell Immunol. 2000;200:116–27. doi: 10.1006/cimm.2000.1625. [DOI] [PubMed] [Google Scholar]
- 26.Heimberger AB, Reina-Ortiz C, Yang DS, et al. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res. 2008;14:5166–72. doi: 10.1158/1078-0432.CCR-08-0320. [DOI] [PubMed] [Google Scholar]
- 27.Fecci PE, Mitchell DA, Whitesides JF, et al. Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res. 2006;66:3294–302. doi: 10.1158/0008-5472.CAN-05-3773. [DOI] [PubMed] [Google Scholar]
- 28.Heimberger AB, Crotty LE, Archer GE, et al. Epidermal growth factor receptor VIII peptide vaccination is efficacious against established intracerebral tumors. Clin Cancer Res. 2003;9:4247–54. [PubMed] [Google Scholar]
- 29.Lee A, Kessler JD, Read TA, et al. Isolation of neural stem cells from the postnatal cerebellum. Nat Neurosci. 2005;8:723–9. doi: 10.1038/nn1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gomez GG, Kruse CA. Mechanisms of malignant glioma immune resistance and sources of immunosuppression. Gene Ther Mol Biol. 2006;10a:133–46. [PMC free article] [PubMed] [Google Scholar]
- 31.Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
- 32.Peng W, Wang HY, Miyahara Y, Peng G, Wang RF. Tumor-associated galectin-3 modulates the function of tumor-reactive T cells. Cancer Research. 2008;68:7228–36. doi: 10.1158/0008-5472.CAN-08-1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kuklinski S, Pesheva P, Heimann C, et al. Expression pattern of galectin-3 in neural tumor cell lines. J Neurosci Res. 2000;60:45–57. doi: 10.1002/(SICI)1097-4547(20000401)60:1<45::AID-JNR5>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
- 34.Strik HM, Schmidt K, Lingor P, et al. Galectin-1 expression in human glioma cells: modulation by ionizing radiation and effects on tumor cell proliferation and migration. Oncol Rep. 2007;18:483–8. [PubMed] [Google Scholar]
- 35.Wei J, Duramad O, Perng OA, Reiner SL, Liu YJ, Qin FX. Antagonistic nature of T helper 1/2 developmental programs in opposing peripheral induction of Foxp3+ regulatory T cells. Proc Natl Acad Sci U S A. 2007;104:18169–74. doi: 10.1073/pnas.0703642104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kang SK, Park JB, Cha SH. Multipotent, dedifferentiated cancer stem-like cells from brain gliomas. Stem Cells Dev. 2006;15:423–35. doi: 10.1089/scd.2006.15.423. [DOI] [PubMed] [Google Scholar]
- 37.Ghods AJ, Irvin D, Liu G, et al. Spheres isolated from 9L gliosarcoma rat cell line possess chemoresistant and aggressive cancer stem-like cells. Stem Cells. 2007;25:1645–53. doi: 10.1634/stemcells.2006-0624. [DOI] [PubMed] [Google Scholar]
- 38.Korbling M, Estrov Z. Adult stem cells for tissue repair - a new therapeutic concept? N Engl J Med. 2003;349:570–82. doi: 10.1056/NEJMra022361. [DOI] [PubMed] [Google Scholar]
- 39.Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31:890–6. doi: 10.1016/s0301-472x(03)00110-3. [DOI] [PubMed] [Google Scholar]
- 40.Piccirillo SG, Reynolds BA, Zanetti N, et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature. 2006;444:761–5. doi: 10.1038/nature05349. [DOI] [PubMed] [Google Scholar]
- 41.Colman H, Sai K, Wang S, et al. Effect of a small molecule inhibitor of the JAK2/STAT3 pathway on self-renewal of glioblastoma stem cells. J Clin Oncol. 2008;26(suppl):89s. [Google Scholar]
- 42.Hussain SF, Kong L-Y, Jordan J, et al. A novel small molecule inhibitor of signal transducers and activators of transcription 3 reverses immune tolerance in malignant glioma patients. Cancer Res. 2007;67:9630–6. doi: 10.1158/0008-5472.CAN-07-1243. [DOI] [PubMed] [Google Scholar]
- 43.Tallman MS. Differentiating therapy in acute myeloid leukemia. Leukemia. 1996;10:1262–8. [PubMed] [Google Scholar]
- 44.Hwang KC, Kim JY, Chang W, et al. Chemicals that modulate stem cell differentiation. Proc Natl Acad Sci U S A. 2008;105:7467–71. doi: 10.1073/pnas.0802825105. [DOI] [PMC free article] [PubMed] [Google Scholar]