Abstract
Statins are potent inhibitors of hydroxyl-3-methylglutaryl co-enzyme A (HMG-CoA) reductase, and have emerged as potential anti-cancer agents based on preclinical evidence. In particular, compelling evidence suggests that statins have a wide range of immunomodulatory properties. However, little is known about the role of statins in tumour immune tolerance. Tumour immune tolerance involves the production of immunosuppressive molecules, such as interleukin (IL)-10, transforming growth factor (TGF)-β and indoleamine-2,3-dioxygenase (IDO) by tumours, which induce a regulatory T cell (Treg) response. In this study, we investigated the effect of simvastatin on the production of IL-10, TGF-β and IDO production and the proliferation of Tregs using several cancer cell lines, and Lewis lung cancer (3LL) cells-inoculated mouse tumour model. Simvastatin treatment resulted in a decrease in the number of cancer cells (3LL, A549 and NCI-H292). The production of the immune regulatory markers IL-10, TGF-β in 3LL and NCI-H292 cells increased after treatment with simvastatin. The expression of IDO and forkhead box P3 (FoxP3) transcription factor was also increased in the presence of simvastatin. In a murine 3LL model, there were no significant differences in tumour growth rate between untreated and simvastatin-treated mice groups. Therefore, while simvastatin had an anti-proliferative effect, it also exhibited immune tolerance-promoting properties during tumour development. Thus, due to these opposing actions, simvastatin had no net effect on tumour growth.
Keywords: indoleamine-2,3-dioxygenase; interleukin-10; regulatory T cell; statin; transforming growth factor-β
Introduction
Hydroxyl-3-methylglutaryl co-enzyme A (HMG-CoA) reductase inhibitors (or statins) are in widespread use due to their low-density lipoprotein (LDL) cholesterol-reducing properties. A number of multi-centre clinical trials have demonstrated the efficacy of HMG-CoA reductase inhibitors in reducing morbidity and mortality associated with coronary artery disease [1,2]. A growing body of evidence also suggests that statins might have chemopreventative properties against cancer [3,4]. There is preclinical evidence of the anti-proliferative, pro-apoptotic, anti-invasive and radiosensitizing properties of statins [5–8]; however, based on some lines of meta-analysis, the association between statins and cancer development remains controversial. Considerable evidence suggests that immune-mediated mechanisms play a dominant role in the effects of statins. For example, statins attenuate T cell activation and proliferation, thereby inhibiting the secretion of proinflammatory cytokines and enhancing the secretion of anti-inflammatory cytokines [9,10]. According to recent data, statins appear to play a significant role in the regulation of regulatory T cells (Tregs) [11,12]. Statins decrease the syntheses of the isopernoid intermediates farnesyl pyrophosphate and geranylgeranyl pyrophosphate, which results in decreased activation of the small G-proteins, Ras and Rho. This pathway may be a potential mechanism for the induction of Tregs by statins [13,14]. Tregs, or CD4+CD25+ T cells, play a key role in tumour-related immune tolerance by suppressing immune responses. Because Tregs are related to cancer development and progression, it is important to evaluate the relationship between statins and Tregs with regard to the immune tolerance of the tumour microenvironment [15]. Although the immunomodulatory potential of statins has been suggested, there are currently no reports on the effects of statins on tumour immune tolerance.
To evaluate the role of statins in tumour immune tolerance, we investigated the effects of simvastatin on indoleamine-2,3-dioxygenase (IDO) production and Treg-mediated tumour immune tolerance.
Materials and methods
Mice and cell lines
Seven-week-old C57Bl/6 mice were purchased from the Orient Co., Ltd (Gapyeong, Korea). Animals were housed in climate-controlled quarters (24 ± 1°C at 50% humidity) under a 12-h light/12-h dark cycle and were maintained under specific pathogen-free conditions. We performed all experiments according to international guidelines on the use of laboratory animals. The Lewis lung cancer (3LL) cell line was derived originally from a C57Bl/6 mouse, and was obtained from the American Type Culture Collection (ATCC, Rockville, MD, USA). 3LL cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco BRL, Gaithersburg, MD, USA) supplemented with 10% fetal bovine serum (FBS), L-glutamine, penicillin and streptomycin. Study mice were inoculated with 3LL cells [2 × 105 in 0·1 ml phosphate-buffered saline (PBS)] by subcutaneous injection into the right forelimb. Forty mice were then randomized into four groups as follows: non-inoculated control group, 3LL-inoculated cancer control, low-dose statin and high-dose statin (10 or 50 mg/kg/day simvastatin, respectively; CJ Pharmacia, Seoul, Korea) groups. The drug was administered by oral gavage q.i.d. in a solution of 0·5% methylcellulose (Sigma-Aldrich, St Louis, MO, USA) containing 0·025% Tween 20 (Sigma-Aldrich). Vehicle control mice (control and cancer control) were gavaged with this same solution. Treatment was initiated 7 days after 3LL inoculation and continued for 21 days. Total tumour volume was measured by a blinded observer using a caliper to measure perpendicular diameters every 4 days. Tumour volume was calculated from the shortest and longest diameters of the tumour mass, according to the following formula [16]: volume (mm3) = (shortest diameter)2 × (longest diameter) × 0·5. Mice that developed tumours were killed on day 28, after which the lungs were removed, rinsed with saline and then fixed in 4% paraformaldehyde.
Cell viability assays
Viability assays were based on the reduction of 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenylte-trazolium bromide (MTT; Amresco, Solon, OH, USA). Cells (control or drug-treated) were rinsed twice with PBS, and then seeded into a 96-well flat-bottomes plate in fetal bovine serum (FBS)-free medium (5 × 104/well in 200 µl). After 24 h incubation, MTT solution was added to each well and incubated at 37°C 2 h, and then the MTT solution was removed; 200 µl dimethylsulphoxide (DMSO; Sigma-Aldrich) was added and incubated at room temperature for 30 min in a shaker. Finally, the density of each well was detected at 540 nm using a microplate reader (CODA; Bio-Rad, San Diego, CA, USA). Cell viability was expressed as a relative viability of tumour cells (% of control cultures incubated with medium only) and was calculated using the following formula: (1 − average absorbance of the experimental group/average absorbance of the control group) × 100%.
Cell cycle analysis
Cell cycle analysis was performed following propidium iodide staining [17]. Cells in culture were treated with simvastatin for 24–48 h. Detached and attached cells were harvested and incubated with CycleTEST™ PLUS DNA Reagent (Becton Dickinson, San Jose, CA, USA). DNA content of the samples was analysed by fluorescence activated cell sorter (FACScan) flow cytometry (Beckman Coulter, Fullerton, CA, USA), and cell cycle distribution was analysed using Multicycle AV (Phoenix Flow System, San Diego, CA, USA).
Enzyme-linked immunosorbent assay (ELISA)
3LL and NCI-H292 cells were cultured in FBS-free media alone or media plus simvastatin (5, 10 or 15 µM) for 24 h. Supernatants from 3LL, NCI-H292 cell cultures (1 × 106 cells/ml) were collected and the concentrations of interleukin (IL)-10 and transforming growth factor (TGF)-β were assessed by ELISA (Biosource, Camarillo, CA, USA), according to the manufacturer's instructions. Simvastatin was obtained from CJ Pharmacia.
FACS analysis
We used a mouse Treg staining kit (eBioscience, San Diego, CA, USA) for FACS analysis. Splenocytes of mice from each group were isolated using a CD4+ T cell isolation kit (MACS; Miltenyi Biotec, Bergisch-Gladbach, Germany), and then CD4+ T cells were isolated by depleting non-CD4+ T cells. The isolated CD4+ T cell population was incubated with the following monoclonal antibodies (mAbs) for 30 min (min) at 4°C: fluorescein isothiocyanate (FITC)-conjugated anti-mouse CD4 (RM4-5), phycoerythrin (PE)-conjugated anti-mouse CD25 (PC61·5) and PE-Cy5-conjugated anti-mouse/rat forkhead box P3 (FoxP3). FACS analysis was performed using an Epics XL system (Beckman Coulter).
Real-time polymerase chain reaction (PCR)
FoxP3 gene expression in splenocytes of mice from each group was evaluated by real-time PCR. Following the isolation of splenocytes, RNA was isolated using a total RNA isolation kit (iNtRON Biotechnology, Seongnam, Korea) and cDNA was prepared using a kit (Invitrogen, Carlsbad, CA, USA), according to the manufacturer's instructions. FoxP3 gene expression was quantified using the SYBR Premix Ex Tag quantitative PCR kit (Takara, Otsu, Japan) and an ABI PRISM 7000 system, with glyceraldehydes-3-phosphate-dehydrogenase (GAPDH) expression as a control. Amplification was performed in a total volume of 25 µl for 40 cycles of 5 s at 95°C and 31 s at 60°C. Samples were analysed in triplicate, and relative expression was determined by normalizing the expression of the target gene to that of GAPDH, and then comparing the normalized value to a normalized reference control sample to calculate fold-change. The primers that were used for amplification spanned intron/exon boundaries to minimize the amplification of genomic DNA. The primer sequences were as follows: GAPDH 5′-TGAAGCAGGCATGGAGGG-3′ and 5′-CGAAGGTGGAAGAGTGGGAG-3′; FoxP3 5′-CAGCTGCCTACAGTGCCCCTA-3′ and 5′-CATTTGCCAGCAGTGGGTAG-3′; IDO 5′-CGGACTGAGAGGACACAGGTTAC-3′ and 5′-ACACATACGCCATGGTGATGTAC-3′.
Western blot analysis
Tumours were homogenized by sonification for 30 s on ice at 100 W, and then subjected to centrifugation at 12 000 g for 10 min at room temperature. Total protein was separated by sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) on a 12% acrylamide gel, and then electrotransferred to a polyvinylidene difluoride membrane (Millipore, Billerica, MA, USA). The membrane was incubated in a solution of 5% fat-free skimmed milk in Tris-buffered saline (TBS)/0·05% Tween 20 for 1 h at room temperature, followed by an anti-IDO mAb (1 : 1000; Upstate, Charlottesville, VA, USA) or an anti-FoxP3 antibody (1 : 1000; BioLegend, San Diego, CA, USA). The membranes were stripped and re-probed with an anti-β-actin mAb (Sigma Aldrich) to verify equal loading of protein in each lane. To detect immunoreactive proteins, the membrane was incubated with horseradish peroxidase (HRP)-linked secondary antibody (Vector Laboratories, Burlingame, CA, USA) for 1 h at room temperature. Secondary antibody was detected using WEST-one (iNtRON Biotechnology), according to the manufacturer's instructions. Immunoreactive bands were quantified by scanning the X-ray films, followed by analysis using SigmaScan-Pro, version 5·01 (SPSS Inc., Chicago, IL, USA).
Statistics
All data are expressed as means ± standard error of the mean (s.e.). One-way analysis of variance (anova) was used to determine statistically significant differences between groups. A P-value of less than 0·05 was considered significant.
Results
Simvastatin inhibits the growth of cancer cells
To evaluate the effect of statins on the growth of tumour cells, we treated human lung cancer cell lines (A549, NCI-H292), C57Bl/6 mouse originated 3LL cells with simvastatin, and then measured cell growth by MTT assay after 24 and 48 h (Fig. 1). Simvastatin decreased the number of human and mouse lung cancer cells in a dose-dependent manner. To determine the mechanism of simvastatin-induced growth inhibition, we analysed the cell cycle distribution of simvastatin-treated tumour cells using propidium iodide staining and FACS in A549 cells and 3LL cells. There was a time-dependent decrease in the percentage of cells in S phase and an increase in G0/G1 phase cells following treatment with simvastatin (Fig. 2). In mammalian cells, cell cycle progression through G1 phase is regulated by cyclin D1, in association with cyclin-dependent kinase (cdk) 4 and cdk2. Because simvastatin induced the accumulation of cells in the G0/G1 phase, we examined the effect of simvastatin on cyclin D1 expression in 3LL cells. Treatment of cells with simvastatin for up to 48 h resulted in a reduction in cyclin D1 expression (Fig. 3). These results were consistent with cell cycle analysis and suggested that simvastatin induces G1 arrest by inhibiting the expression of cyclin D1.
Fig. 1.

Simvastatin decreased the number of cancer cells. Cells were incubated at an initial density of 1 × 106 cell/ml in RPMI-1640 containing 10% fetal bovine serum in the presence or absence of increasing concentrations of simvastatin, and were then cultured for 48 h. Samples were removed at 24 and 48 h, and cell proliferation was analysed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenylte-trazolium bromide (MTT) assay. Compared to the 24-h numbers, the proliferations of Lewis lung carcinoma (3LL), A549 and NCI-H292 cells at 48 h were decreased in a dose-dependent manner by treatment with simvastatin. The data represent the means ± standard error (s.e.) of three independent experiments (*P < 0·05).
Fig. 2.

Effect of simvastatin on cell cycle progression. Simvastatin treatment results in a decrease in the percentage of cells in the S phase and an increase in the number of cells in G0/1. Cell cycle progression was analysed at 24 and 48 h after simvastatin treatment in A549 cells and Lewis lung carcinoma (3LL) cells. There was a time-dependent decrease in the percentage of cells in the S phase and an increase in the percentage of cells in the G0/1 phase following simvastatin treatment (*P < 0·05).
Fig. 3.

Simvastatin down-regulates cyclin D1 expression in Lewis lung carcinoma (3LL) cells. Cells were treated with simvastatin for 48 h, then the media was removed and cells were incubated in control or treatment media. Cells were collected at 24 and 48 h, and then whole cell lysate was analysed by Western blot using an anti-cyclin D1 antibody.
Effects of simvastatin on the production of immunomodulatory cytokines in vitro
Simvastatin increases the production of IL-10 and TGF-β in cancer cell lines
IL-10 and TGF-β are associated with tumour immune tolerance and Treg activation. To determine whether simvastatin regulated IL-10 and TGF-β production in vitro, we treated 3LL and NCI-H292 cells with increasing concentrations of simvastatin. Treatment with simvastatin increased IL-10 and TGF-β production (Fig. 4).
Fig. 4.

Simvastatin increases interleukin (IL)-10 and transforming growth factor (TGF)-β production in lung cancer cells in vitro. Cells were incubated in fetal bovine serum-free culture media alone, or in culture media plus simvastatin (5, 10 or 15 µM) for 48 h, and then IL-10 and TGF-β production was analysed by enzyme-linked immunosorbent assay (ELISA). (a) Simvastatin increased IL-10 production in a dose-dependent manner; (b) simvastatin also increased TGF-β production. The data represent the means ± standard error (s.e.) of at least three independent experiments (*P < 0·01 compared to the control group, **P < 0·05 compared to the control group).
Simvastatin increases FoxP3 and IDO expression
To determine whether simvastatin regulated the gene expression of FoxP3, isolated tumour-infiltrating lymphocytes (TILs) were treated with simvastatin and then FoxP3 expression was analysed by real-time PCR. FoxP3 is a transcription factor that serves as a reliable intracellular marker for Tregs, and high numbers of CD4+CD25+FoxP3+ cells can be found in circulation as well as in tumours [18]. FoxP3 expression was higher in simvastatin-treated TILs (Fig. 5a). To confirm the results of real-time PCR analysis of isolated TILs, we analysed FoxP3 protein levels in tumours from simvastatin-treated and untreated control mice by Western blotting. FoxP3 protein levels were higher in tumours from simvastatin-treated mice compared to untreated control mice (Fig. 5b). There was also a corresponding increase in IDO levels in the simvastatin groups. Thus, real-time PCR and Western blot analysis demonstrated that the expression of FoxP3, as well as IDO expression in tumours, increases with simvastatin treatment.
Fig. 5.

Simvastatin increases the expressions of forkhead box P3 (FoxP3) and indoleamine-2,3-dioxygenase (IDO). (a) Tumour-infiltrating lymphocytes of Lewis lung carcinoma (3LL) control mice were isolated, and then FoxP3 gene expression was analysed by real-time polymerase chain reaction (PCR). FoxP3 gene expression was higher in the simvastatin-treated groups than in the control group (10 µM and 15 µM, *P < 0·05 compared to the control group). (b) Tumours were removed from mice from the indicated treatment groups, and IDO and FoxP3 levels were analysed by Western blot. Sample loading was normalized using a probe for beta-actin. The expression of IDO and FoxP3 within the tumour mass was increased by simvastatin treatment.
Simvastatin increases the percentage of Tregs
To assess the relationship between simvastatin and Treg proliferation, splenocytes were isolated from non-inoculated control mice and 3LL-inoculated cancer control, low-dose and high-dose simvastatin mice. The percentage of CD4+CD25+ and CD4+CD25+FoxP3+ cells in each group was analysed by FACS using the Epics XL system. There was an increase in the percentage of CD4+CD25+ and CD4+CD25+FoxP3+ cells in the simvastatin groups compared to the 3LL-inoculated cancer control group (Fig. 6a). A representative dot plot showing the increased percentage of Tregs following simvastatin treatment is shown in Fig. 6b. These results indicated that simvastatin enhances the Tregsin vivo.
Fig. 6.

Simvastatin increases the percentage of regulatory T cells (Treg) cells in vivo. C57Bl/6 mice were inoculated with Lewis lung carcinoma (3LL) cells, and then either untreated or treated with simvastatin (10 or 50 mg/kg daily). After 4 weeks, mice were killed and spleens were extracted. The percentage of CD4+CD25+ and CD4+CD25+forkhead box P3 (FoxP3)+ cells in the spleens of each group was analysed by fluorescence activated cell sorter (FACS). The percentage of CD4+CD25+ and CD4+CD25+FoxP3+ cells was significantly higher in the high-dose (50 mg/kg) simvastatin group compared to the control group (*P < 0·01 compared to the 3LL control group).
Simvastatin has no effect on the growth of tumours
To determine whether simvastatin affect tumour growth, we measured tumour size in mice treated with simvastatin and in untreated 3LL cancer control mice. We did not observe a significant inhibitory effect of simvastatin on tumour growth. Furthermore, there was no significant difference in tumour mass between the low-dose (10 mg/kg) and high-dose (50 mg/kg) statin groups (Fig. 7).
Fig. 7.

Tumour-bearing mice (10 mice per group) were treated with vehicle or simvastatin (10 mg/kg or 50 mg/kg) starting at day 7 after inoculation until day 28. Tumour volume was measured on the indicated days. Data represent the means ± standard deviation. There were no significant differences in tumour size between the untreated Lewis lung carcinoma (3LL) group and the simvastatin-treated groups.
Discussion
In the current study, we provide evidence of a novel mechanism by which statins regulate Treg-mediated tumour immune tolerance. Simvastatin decreased the number of tumour cells in vitro through the induction of a G1/S phase arrest. Simvastatin also activated and enhanced Tregsin vivo. These opposing actions of simvastatin resulted in no net effect on the inhibition of tumour growth in a murine 3LL model.
Statins comprise a therapeutic class of agents that reduce plasma cholesterol levels by inhibiting HMG-CoA reductase, the rate-controlling enzyme in cholesterol production. A growing body of evidence suggests that statins may also have chemopreventative properties against cancer [3]. However, there is a long-standing debate concerning the association between statins and cancer. In a review of rodent carcinogenicity tests, statins initiated or promoted cancer in rats and mice [19]. The putative growth-suppressive properties of statins are believed to be mediated through the inhibition of HMG-CoA reductase, but other mechanisms have also been suggested [5,20]. HMG-CoA reductase inhibitors can synchronize tumour cells, and thereby exert an anti-proliferative effect, by blocking the G1–S cell cycle transition. However, while statins can inhibit cell proliferation, they also appear to enhance tumour progression; the mechanism responsible for this phenomenon was unknown. There have been several reports of the immunomodulatory effects of statin [21,22]. Statins attenuate T cell activation and proliferation, thereby inhibiting the secretion of proinflammatory cytokines and enhancing the secretion of anti-inflammatory cytokines [9,10]. For example, statins have been shown to reduce IL-1, IL-6, IL-8, IL-12 and interferon (IFN)-γ levels, and consequently reduce T cell activation. Recent evidence suggests that statins play a role in the modulation of immune responses through the regulation of Tregs, which are involved in tumour immune tolerance [11,12,23]. In the current study, we demonstrated that simvastatin treatment induces FoxP3 expression, and that this increase in FoxP3 expression is accompanied by an expansion of Tregs, as determined by FACS analysis of CD4+CD25+FoxP3+ cells. The X chromosome-encoded forkhead transcription factor, FoxP3, was identified recently as a key player in Treg biology [24]. This is the most specific marker for Tregs and is expressed specifically in CD4+CD25+ T cells in the thymus and the periphery [25]. FoxP3 was not observed in tumour cells except pancreatic carcinoma cells [26], highly restricted to the subset of T cells and correlating with suppressor activity [27,28]. The induction of Tregs by simvastatin correlated with the production of IL-10 and TGF-β, which are Treg inducers [29].
IDO is a rate-limiting enzyme in tryptophan catabolism. It, too, has emerged as an important immunoregulatory enzyme. A localized reduction in tryptophan, which is an essential amino acid, and the concomitant production of immunomodulatory tryptophan metabolites contributes to the immunosuppressive effects of IDO [30,31]. The up-regulation of IDO at inflammatory sites leads to the depletion of tryptophan and the generation of intermediate catabolites (kynurenines), which exert inhibitory effects on T lymphocytes. IDO is also expressed by tumour cells. Immunogenic tumours that are engineered to overexpress IDO grow more aggressively, and this effect correlates with a decrease in activated T cells at the tumour site [31]. Furthermore, the expression of IDO in cancer cells has been associated with the induction of Tregsin vitro and in vivo[32,33]. In the current study, we evaluated the relationship between simvastatin and IDO expression. In a murine 3LL model, mice that were treated with simvastatin exhibited higher levels of IDO protein expressions than untreated cancer control mice (Fig. 5). Because IDO influences Treg proliferation, we also examined the effect of simvastatin on Treg-mediated immune regulation. The percentage of Tregs in mice was higher in simvastatin-treated mice than in control mice (Fig. 6). Tregs can be generated from conventional CD4+CD25- T cells following exposure to antigens and IL-10, and the production of IL-10 or TGF-β by the induced Tregs appears to mediate their inhibitory properties [34]. The presence of these regulatory cells in cancer patients might be important in the suppression of T cells and subsequent tumour growth. Our results demonstrate that simvastatin increases IL-10 production and IDO expression in cancer cells, and induces the expansion of Tregs.
In conclusion, simvastatin inhibits cancer cell proliferation through the induction of G1/S phase arrest. However, simvastatin also induces the immune suppressive cytokines and the expansion of functionally active Tregs. Increased numbers of Tregs induced by simvastatin in the cancer microenvironment would enable tumour cells to escape immune surveillance and proliferate. These opposing effects of simvastatin would result in no net effect on tumour growth.
Acknowledgments
This research was supported by Basic Science Research Program through the Korea Research Foundation Grant (KRF-2007-331-E00212).
Disclosure
None.
References
- 1.Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S) Lancet. 1994;344:1383–9. [PubMed] [Google Scholar]
- 2.Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. The Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group. N Engl J Med. 1998;339:1349–57. doi: 10.1056/NEJM199811053391902. [DOI] [PubMed] [Google Scholar]
- 3.Graaf MR, Richel DJ, van Noorden CJ, Guchelaar HJ. Effects of statins and farnesyltransferase inhibitors on the development and progression of cancer. Cancer Treat Rev. 2004;30:609–41. doi: 10.1016/j.ctrv.2004.06.010. [DOI] [PubMed] [Google Scholar]
- 4.Jakobisiak M, Golab J. Potential antitumor effects of statins. Int J Oncol. 2003;23:1055–69. Review. [PubMed] [Google Scholar]
- 5.Keyomarsi K, Sandoval L, Band V, Pardee AB. Synchronization of tumor and normal cells from G1 to multiple cell cycles by lovastatin. Cancer Res. 1991;51:3602–9. [PubMed] [Google Scholar]
- 6.Xia Z, Tan MM, Wong WW, Dimitroulakos J, Minden MD, Penn LZ. Blocking protein geranylgeranylation is essential for lovastatin-induced apoptosis of human acute myeloid leukemia cells. Leukemia. 2001;15:1398–407. doi: 10.1038/sj.leu.2402196. [DOI] [PubMed] [Google Scholar]
- 7.Kusama T, Mukai M, Iwasaki T, et al. Inhibition of epidermal growth factor-induced RhoA translocation and invasion of human pancreatic cancer cells by 3-hydroxy-3-methylglutaryl-coenzyme a reductase inhibitors. Cancer Res. 2001;61:4885–91. [PubMed] [Google Scholar]
- 8.Miller AC, Kariko K, Myers CE, Clark EP, Samid D. Increased radioresistance of EJras-transformed human osteosarcoma cells and its modulation by lovastatin, an inhibitor of p21ras isoprenylation. Int J Cancer. 1993;53:302–7. doi: 10.1002/ijc.2910530222. [DOI] [PubMed] [Google Scholar]
- 9.Youssef S, Stuve O, Patarroyo JC, et al. The HMG-CoA reductase inhibitor, atorvastatin, promotes a Th2 bias and reverses paralysis in central nervous system autoimmune disease. Nature. 2002;420:78–84. doi: 10.1038/nature01158. [DOI] [PubMed] [Google Scholar]
- 10.Rosenson RS, Tangney CC, Casey LC. Inhibition of proinflammatory cytokine production by pravastatin. Lancet. 1999;353:983–4. doi: 10.1016/S0140-6736(98)05917-0. [DOI] [PubMed] [Google Scholar]
- 11.Mausner-Fainberg K, Luboshits G, Mor A, et al. The effect of HMG-CoA reductase inhibitors on naturally occurring CD4+CD25+ T cells. Atherosclerosis. 2008;197:829–39. doi: 10.1016/j.atherosclerosis.2007.07.031. [DOI] [PubMed] [Google Scholar]
- 12.Mira E, Leon B, Barber DF, et al. Statins induce regulatory T cell recruitment via a CCL1 dependent pathway. J Immunol. 2008;181:3524–34. doi: 10.4049/jimmunol.181.5.3524. [DOI] [PubMed] [Google Scholar]
- 13.Cordle A, Koenigsknecht-Talboo J, Wilkinson B, Limpert A, Landreth G. Mechanisms of statin-mediated inhibition of small G-protein function. J Biol Chem. 2005;280:34202–9. doi: 10.1074/jbc.M505268200. [DOI] [PubMed] [Google Scholar]
- 14.Blanco-Colio LM, Tunon J, Martin-Ventura JL, Egido J. Anti-inflammatory and immunomodulatory effects of statins. Kidney Int. 2003;63:12–23. doi: 10.1046/j.1523-1755.2003.00744.x. [DOI] [PubMed] [Google Scholar]
- 15.Goldstein MR, Mascitelli L, Pezzetta F. Statins, Tregs and cancer. Atherosclerosis. 2008;196:483–4. doi: 10.1016/j.atherosclerosis.2007.09.028. [DOI] [PubMed] [Google Scholar]
- 16.Corbett TH, Griswold DP, Jr, Roberts BJ, Peckham JC, Schabel FM., Jr Evaluation of single agents and combinations of chemotherapeutic agents in mouse colon carcinomas. Cancer. 1977;40:2660–80. doi: 10.1002/1097-0142(197711)40:5+<2660::aid-cncr2820400940>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 17.Rakkar AN, Katayose Y, Kim M, et al. A novel adenoviral vector expressing human Fas/CD95/APO-1 enhances p53-mediated apoptosis. Cell Death Differ. 1999;6:326–33. doi: 10.1038/sj.cdd.4400498. [DOI] [PubMed] [Google Scholar]
- 18.Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
- 19.Newman TB, Hulley SB. Carcinogenicity of lipid-lowering drugs. JAMA. 1996;275:55–60. [PubMed] [Google Scholar]
- 20.Rao S, Porter DC, Chen X, Herliczek T, Lowe M, Keyomarsi K. Lovastatin-mediated G1 arrest is through inhibition of the proteasome, independent of hydroxymethyl glutaryl-CoA reductase. Proc Natl Acad Sci USA. 1999;96:7797–802. doi: 10.1073/pnas.96.14.7797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kwak B, Mulhaupt F, Myit S, Mach F. Statins as a newly recognized type of immunomodulator. Nat Med. 2000;6:1399–402. doi: 10.1038/82219. [DOI] [PubMed] [Google Scholar]
- 22.Mach F. Immunosuppressive effects of statins. Atheroscler Suppl. 2002;3:17–20. doi: 10.1016/s1567-5688(01)00010-1. [DOI] [PubMed] [Google Scholar]
- 23.Jain MK, Ridker PM. Anti-inflammatory effects of statins: clinical evidence and basic mechanisms. Nat Rev Drug Discov. 2005;4:977–87. doi: 10.1038/nrd1901. [DOI] [PubMed] [Google Scholar]
- 24.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–61. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
- 25.Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:337–42. doi: 10.1038/ni909. [DOI] [PubMed] [Google Scholar]
- 26.Hinz S, Pagerols-Raluy L, Oberg HH, et al. Foxp3 expression in pancreatic carcinoma cells as a novel mechanism of immune evasion in cancer. Cancer Res. 2007;67:8344–50. doi: 10.1158/0008-5472.CAN-06-3304. [DOI] [PubMed] [Google Scholar]
- 27.Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG, Rudensky AY. Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity. 2005;22:329–41. doi: 10.1016/j.immuni.2005.01.016. [DOI] [PubMed] [Google Scholar]
- 28.Yagi H, Nomura T, Nakamura K, et al. Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int Immunol. 2004;16:1643–56. doi: 10.1093/intimm/dxh165. [DOI] [PubMed] [Google Scholar]
- 29.Levings MK, Bacchetta R, Schulz U, Roncarolo MG. The role of IL-10 and TGF-beta in the differentiation and effector function of T regulatory cells. Int Arch Allergy Immunol. 2002;129:263–76. doi: 10.1159/000067596. [DOI] [PubMed] [Google Scholar]
- 30.Taylor MW, Feng GS. Relationship between interferon-gamma, indoleamine 2,3-dioxygenase, and tryptophan catabolism. FASEB J. 1991;5:2516–22. [PubMed] [Google Scholar]
- 31.Uyttenhove C, Pilotte L, Theate I, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med. 2003;9:1269–74. doi: 10.1038/nm934. [DOI] [PubMed] [Google Scholar]
- 32.Curti A, Pandolfi S, Valzasina B, et al. Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells. Blood. 2007;109:2871–7. doi: 10.1182/blood-2006-07-036863. [DOI] [PubMed] [Google Scholar]
- 33.Wobser M, Voigt H, Houben R, et al. Dendritic cell based antitumor vaccination: impact of functional indoleamine 2,3-dioxygenase expression. Cancer Immunol Immunother. 2007;56:1017–24. doi: 10.1007/s00262-006-0256-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sakaguchi S. Regulatory T cells: key controllers of immunologic self-tolerance. Cell. 2000;101:455–8. doi: 10.1016/s0092-8674(00)80856-9. [DOI] [PubMed] [Google Scholar]
