Evidence supports the protective role of nonsteroidal anti-inflammatory drugs (NSAID) and statins against colon cancer. Experiments were designed to evaluate the efficacies atorvastatin and NSAIDs administered individually and in combination against colon tumor formation. F344 rats were fed AIN-76A diet, and colon tumors were induced with azoxymethane. One week after the second azoxymethane treatment, groups of rats were fed diets containing atorvastatin (200 ppm), sulindac (100 ppm), naproxen (150 ppm), or their combinations with low-dose atorvastatin (100 ppm) for 45 weeks. Administration of atorvastatin at 200 ppm significantly suppressed both adenocarcinoma incidence (52% reduction, P = 0.005) and multiplicity (58% reduction, P = 0.008). Most importantly, colon tumor multiplicities were profoundly decreased (80%–85% reduction, P < 0.0001) when given low-dose atorvastatin with either sulindac or naproxen. Also, a significant inhibition of colon tumor incidence was observed when given a low-dose atorvastatin with either sulindac (P = 0.001) or naproxen (P = 0.0005). Proliferation markers, proliferating cell nuclear antigen, cyclin D1, and β-catenin in tumors of rats exposed to sulindac, naproxen, atorvastatin, and/or combinations showed a significant suppression. Importantly, colon adenocarcinomas from atorvastatin and NSAIDs fed animals showed reduced key inflammatory markers, inducible nitric oxide synthase and COX-2, phospho-p65, as well as inflammatory cytokines, TNF-α, interleukin (IL)-1β, and IL-4. Overall, this is the first report on the combination treatment using low-dose atorvastatin with either low-dose sulindac or naproxen, which greatly suppress the colon adenocarcinoma incidence and multiplicity. Our results suggest that low-dose atorvastatin with sulindac or naproxen might potentially be useful combinations for colon cancer prevention in humans. Cancer Prev Res; 4(11); 1895–902. ©2011 AACR.

Colorectal cancer is one of the major leading causes of death from cancer in the United States as well as in the worldwide. It is predicted to be responsible for the death of almost 50,000 people each year in the United States alone (1, 2). Because the majority of the cause of colon cancer is attributable to lifestyle, diet, and genetic factors, there has been increasing awareness and focus on the prevention of colon cancer (3, 4). In particular, the presence of chronic inflammatory conditions in the colonic environment has been implicated in the development of colorectal cancer, and treatment regimens against inflammatory markers have reduced the risk of colon cancer (5–7).

Increased aberrant expression of inflammatory genes, such as inducible nitric oxide synthase (iNOS) and COX, has been shown in the azoxymethane-induced colon cancer model from the early stage of hyperplastic aberrant crypt foci (ACF) to late-stage adenocarcinoma (8–14). Because many studies report that selective iNOS and COX-2 inhibitors exerted suppressive effects against colon cancer (8, 11, 15–22), there is a rationale for investigating the ability of the combination of low-dose atorvastatin and nonsteroidal anti-inflammatory drugs (NSAIDs) to inhibit iNOS and COX-2 in a colon cancer model where inflammatory genes play a key role in carcinogenesis.

Evidence supports the protective role of NSAIDs and statin (15, 16, 23–27). In our earlier study, we showed that statins such as atorvastatin (Lipitor), as well as NSAIDs as effective agents in suppressing colon cancer in animals (7, 16, 24). Azoxymethane-induced tumors result from mutations in the Wnt/β-catenin pathway (28–30). Aberrant expression of β-catenin can be regarded as a key event during colorectal tumorigenesis (31) and is linked to the increased transcription of a number of genes such as cyclin D1 (32, 33). Cyclin D1 is overexpressed in patients with adenomatous polyps, primary colorectal adenocarcinoma, and familial adenomatous polyposis (32, 34). Cyclin D1 is a target gene of the Wnt signaling pathway (35), and mutations in this pathway are responsible for approximately 90% of colorectal cancer (36). Mutations in genes belonging to the Wnt pathway, such as inactivating mutations in the adenomatous polyposis coli (APC) gene or activating mutations in β-catenin, result in the nuclear accumulation of β-catenin and subsequent complex formation with T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors to activate gene transcription (37). TCF/LEF-binding sites on promoters of cell proliferation genes, such as cyclin D1 and c-MYC (35, 38), thus serve to transmit the aberrant mutations to tumorigenic signals within the colonic crypts.

As discussed above, we and others have shown the chemopreventive effects of statins and number of NSAIDs (7, 16, 20, 23, 24, 39). However, many of these studies used higher dose levels and also no efficacy data on the most commonly used NSAID, naproxen, in the colon cancer model is available. Importantly, our aim to establish whether low-dose combinational approaches targeting different pathways would be ideal for human colorectal cancer prevention. Thus, in the present study, experiments were designed to evaluate the efficacies of atorvastatin and NSAIDs, sulindac and naproxen, administered individually and in combination against colon tumorigenesis. We evaluated the chemopreventive potential of a low-dose atorvastatin in combination with NSAIDs with colonic tumor formation as the endpoint and further determined the action of atorvastatin and in combination with NSAIDs in regulating the expression of key protein markers and signaling pathway during colon carcinogenesis.

Compounds

Atorvastatin, sulindac, and naproxen (Fig. 1) were provided by the DCP Repository at the National Cancer Institute. Conversion products from [3H]-l-arginine to [3H]-l-citrulline were obtained from New England Nuclear Corporation.

Figure 1.

Structures of sulindac, naproxen, and atorvastatin.

Figure 1.

Structures of sulindac, naproxen, and atorvastatin.

Close modal

Animals, diet, and in vivo experimental procedures

Weanling male F344 rats obtained from Charles River Breeding Laboratories were randomly distributed by weight into control and experimental groups. Animals had access to food and water at all times. Food cups were replenished with fresh diet twice weekly. Experimental diets were purchased from Research Diets and stored at 4°C. Beginning at 5 weeks of age, all rats were fed the modified American Institute of Nutrition-76A (AIN-76A) diet. At 7 weeks of age, the animals were given subcutaneous injections of azoxymethane (CAS no. 25843-45-2; Ash Stevens) at a dose rate of 15 mg/kg body weight or saline as solvent control once weekly for 2 weeks. One week after the second azoxymethane treatment, groups of rats were fed AIN-76A diet containing atorvastatin (200 ppm), sulindac (100 ppm), naproxen (150 ppm), or their combinations with low-dose atorvastatin (100 ppm) for 45 weeks. At autopsy, animals were sacrificed by CO2 asphyxiation, and the colon was removed, rinsed in PBS, opened longitudinally, and flattened on a filter paper. The location and size of each tumor was noted. Mucosal scrapings were collected and stored at −80°C for further analysis. Tumors were removed, fixed in 10% buffered formalin for 24 hours, and transferred to 70% ethanol for histopathologic analysis.

Histopathology and immunohistochemistry

The tumor tissues were dehydrated, embedded in paraffin, and cut into 4 μm thick sections. For histopathology, the sections were hydrated and stained with hematoxylin and eosin according to the standard protocol. The stained sections were analyzed for tumor grades by a pathologist. For immunohistochemical analysis, only noninvasive adenocarcinomas were selected for the evaluation of protein markers. The detailed procedures for immunohistochemical analysis are reported previously (40). The primary antibodies against proliferating cell nuclear antigen (PCNA; 1:1,500 diluted) from BD PharMingen; cyclin D1 (1:500 diluted), β-catenin (1:500 diluted), phospho-p65 (1:250 diluted), and iNOS (1:500 diluted) all from Santa Cruz Biotechnology; and COX-2 (1:200 diluted) from Cayman Chemical were treated on the sections. The images were taken randomly at 400× using Zeiss AxioCam HRc camera fitted to a Zeiss Axioskope 2 Plus microscope. For β-catenin quantification, Image Pro 6.2 Plus (Media Cybernetics, Inc.) was used to obtain the IOD (integrated optical density = average intensity/density of each object) values.

Measurement of iNOS activity

iNOS activities were determined in colonic tumor samples of rats exposed to various experimental diets. Conversion of [3H]-l-arginine to [3H]-l-citrulline was measured by a modification described previously (7). iNOS activity is expressed as nanomoles of [3H]-l-citrulline per milligram of protein per minute.

Measurement of cytokine production by ELISA

Colonic mucosa samples were homogenized in a PBS-based buffer solution (PBS, 0.4 mol/L NaCl, 10 mmol/L EDTA, 0.1 mmol/L phenylmethylsulfonylfluoride, 0.1 mol/L benzethonium ion, 0.5% bovine serum albumin, 3.0% aprotinin, and 0.05% Tween 20) on ice using a Tekmar Tissuemiser (Fisher Scientific International, Inc.). The homogenized solution was centrifuged at 10,000 rpm at 4°C for 10 minutes. The supernatant was collected for determination of protein concentration and stored at −20°C. For determination of the levels of interleukin (IL)-1β, IL-4, and TNF-α, tissue homogenates were normalized down to a concentration of 1.0 mg/mL of total protein and then diluted 10-fold in diluent buffer for analysis, following the manufacturer's protocols. Invitrogen Immunoassay kits (BioSource International Inc.) were used to determine the levels of IL-1β (catalogue no. KRC0012), IL-4 (catalogue no. KRC0042), and TNF-α (catalogue no. KRC3012).

Statistical analysis

Statistical significance was analyzed using Student's t test or ANOVA test followed by Tukey's multiple comparison test. Tumor incidence was analyzed by 2-tailed Fisher's exact probability test.

General observations

Body weights of animals fed the experimental diets containing atorvastatin, sulindac, or naproxen individually or in combination were comparable with those fed the control diet throughout the study, indicating that the dose of atorvastatin, sulindac, or naproxen used did not cause any overt toxicity. The maximum tolerated dose (MTD) for each agent was previously determined (sulindac ∼400 ppm, naproxen ∼700 ppm, and atorvastatin >600 ppm). Therefore, the doses were determined on the basis of the information with these agents in AIN-76A diet on the F344 rats (16, 20, 24, 25). In the present study, we used the lower MTD doses of sulindac (∼25% MTD), naproxen (∼20% MTD), and atorvastatin (∼30% and 15%), respectively. Importantly, administration of these dose levels would produce plasma area under the curve (AUC) levels in rats that would somewhat equal the plasma AUC levels of humans given low to mid doses of these agents.

A low-dose atorvastatin with sulindac or naproxen reduces tumor incidence and tumor multiplicity in azoxymethane-injected rats

The effects of administration of a low-dose atorvastatin with sulindac or naproxen on azoxymethane-induced colon tumorigenesis were evaluated, and the results are summarized in Table 1. None of the rats in the saline groups (without azoxymethane injection, n = 6 per group) developed tumors when autopsied at week 45 (data not shown). Most of azoxymethane-treated control diet fed rats developed adenocarcinomas at 45 weeks. Histopathologic analysis by hematoxylin and eosin staining revealed more than 90% of the tumors from the control group as adenocarcinomas and the remaining less than 10% were carcinoma in situ. Approximately 95% of the total adenocarcinomas belonged to the noninvasive adenocarcinoma grade whereas the remaining 5% was invasive adenocarcinoma (Table 1). Administration of sulindac and naproxen individually had modest inhibitory (∼25% incidence and ∼33% multiplicity) effect on colon adenocarcinomas. However, atorvastatin (200 ppm) significantly suppressed both colon tumor incidence (52% reduction, P = 0.005) and multiplicity (58% reduction, P = 0.008). Most importantly, total colon tumor incidence was significantly decreased when rats were given low-dose atorvastatin with either sulindac (58% reduction, P = 0.001) or naproxen (63% reduction, P = 0.0005), respectively (Table 1). Colon tumor multiplicities were also profoundly reduced when rats were given low-dose atorvastatin with either sulindac or naproxen (80%–85%, P < 0.0001; Table 2).

Table 1.

Chemopreventive effects of atorvastatin, sulindac, naproxen alone, or combination of low-dose atorvastatin with either sulindac or naproxen on azoxymethane-induced colon adenocarcinoma incidence and multiplicity in male F344 rats

Tumor incidenceTumor multiplicity
Experimental groupaNumber of rats at autopsy% of rats with adenocarcinomasb% inhibitionAdenocarcinomas/rat,c mean ± SE% inhibition
AOM-control (AIN-76A) diet 31 23/31 (74.2%)  1.77 ± 0.31  
AOM-sulindac (100 ppm) 33 19/33 (57.6%) 22.4% 1.15 ± 0.26 35.0% 
AOM-naproxen (150 ppm) 30 17/30 (56.7%) 23.6% 1.23 ± 0.25 30.5% 
AOM-atorvastatin (200 ppm) 31 11/31 (35.5%; P = 0.005) 52.2% 0.74 ± 0.19 (P = 0.008) 58.2% 
AOM-atorvastatin (100 ppm) + sulindac (100 ppm) 32 10/32 (31.3%; P = 0.001) 57.8% 0.31 ± 0.09 (P1 < 0.0001; P2 = 0.005) 82.5% 
AOM-atorvastatin (100 ppm) + naproxen (150 ppm) 33 9/33 (27.3%; P = 0.0004) 63.2% 0.27 ± 0.08 (P1 < 0.0001; P2 = 0.004) 84.8% 
Tumor incidenceTumor multiplicity
Experimental groupaNumber of rats at autopsy% of rats with adenocarcinomasb% inhibitionAdenocarcinomas/rat,c mean ± SE% inhibition
AOM-control (AIN-76A) diet 31 23/31 (74.2%)  1.77 ± 0.31  
AOM-sulindac (100 ppm) 33 19/33 (57.6%) 22.4% 1.15 ± 0.26 35.0% 
AOM-naproxen (150 ppm) 30 17/30 (56.7%) 23.6% 1.23 ± 0.25 30.5% 
AOM-atorvastatin (200 ppm) 31 11/31 (35.5%; P = 0.005) 52.2% 0.74 ± 0.19 (P = 0.008) 58.2% 
AOM-atorvastatin (100 ppm) + sulindac (100 ppm) 32 10/32 (31.3%; P = 0.001) 57.8% 0.31 ± 0.09 (P1 < 0.0001; P2 = 0.005) 82.5% 
AOM-atorvastatin (100 ppm) + naproxen (150 ppm) 33 9/33 (27.3%; P = 0.0004) 63.2% 0.27 ± 0.08 (P1 < 0.0001; P2 = 0.004) 84.8% 

Abbreviations: AOM, azoxymethane; SE, standard error.

aTest agents were administered in the diet following the second AOM or saline treatment and continuously thereafter for the duration of the experiment which is 45 weeks from the start of AOM or saline treatment.

bTumor incidence was analyzed by 2-tailed Fisher's exact probability test in comparison with the control group.

cStatistical significance was analyzed using Student's t test. P1 is the value for the comparison of rats treated with chemopreventive agents with control rats; P2 is the value for the comparison of rats treated with combination of low-dose atorvastatin (100 ppm) with rats treated with either sulindac or naproxen alone.

Table 2.

Atorvastatin, in combination with sulindac or naproxen, decreases mucosal and colonic tumor levels of the proinflammatory cytokines, TNF-α, IL-1β, and IL-4

Experimental groupaTNF-α, pg/mgIL-1β, pg/mgIL-4, pg/mg
AOM-control (AIN-76A) diet 1,182.9 ± 114.9 2,194.7 ± 209.4 321.0 ± 35.6 
AOM-sulindac (100 ppm) 754.1 ± 64.7 (P = 0.004) 1,610.0 ± 173.0 (P = 0.04) 212.4 ± 26.4 (P = 0.02) 
AOM-naproxen (150 ppm) 910.7 ± 85.5 1,917.9 ± 316.6 292.5 ± 51.1 
AOM-atorvastatin (200 ppm) 812.8 ± 117.7 (P = 0.03) 1,431.3 ± 195.1 (P = 0.01) 186.2 ± 30.8 (P = 0.01) 
AOM-atorvastatin (100 ppm) + sulindac (100 ppm) 747.7 ± 109.2 (P = 0.03) 1,410.9 ± 200.7 (P = 0.01) 193.9 ± 39.2 (P = 0.03) 
AOM-atorvastatin (100 ppm) + naproxen (150 ppm) 759.7 ± 205.0 (P = 0.01) 1,499.9 ± 226.7 (P = 0.03) 191.7 ± 29.8 (P = 0.01) 
Experimental groupaTNF-α, pg/mgIL-1β, pg/mgIL-4, pg/mg
AOM-control (AIN-76A) diet 1,182.9 ± 114.9 2,194.7 ± 209.4 321.0 ± 35.6 
AOM-sulindac (100 ppm) 754.1 ± 64.7 (P = 0.004) 1,610.0 ± 173.0 (P = 0.04) 212.4 ± 26.4 (P = 0.02) 
AOM-naproxen (150 ppm) 910.7 ± 85.5 1,917.9 ± 316.6 292.5 ± 51.1 
AOM-atorvastatin (200 ppm) 812.8 ± 117.7 (P = 0.03) 1,431.3 ± 195.1 (P = 0.01) 186.2 ± 30.8 (P = 0.01) 
AOM-atorvastatin (100 ppm) + sulindac (100 ppm) 747.7 ± 109.2 (P = 0.03) 1,410.9 ± 200.7 (P = 0.01) 193.9 ± 39.2 (P = 0.03) 
AOM-atorvastatin (100 ppm) + naproxen (150 ppm) 759.7 ± 205.0 (P = 0.01) 1,499.9 ± 226.7 (P = 0.03) 191.7 ± 29.8 (P = 0.01) 

aThe mucosa samples were homogenized and assayed by ELISA for the different cytokines, as described under Materials and Methods. Colon mucosa samples were randomly selected from each group and cytokine levels were analyzed (n = 12). The mean ± SD values are shown.

A low-dose atorvastatin, in combination with sulindac or naproxen, decreases cell proliferation markers, PCNA, β-catenin, and cyclin D1 in the colon adenocarcinomas

As shown in Figure 2A (first row), the histologic evaluation revealed that the majority of tumors were noninvasive adenocarcinomas. The expression of PCNA, a marker for cell proliferation, was determined in the adenocarcinomas from the control and treatment groups. The colon tumors from a low-dose atorvastatin, in combination with sulindac or naproxen, fed group showed significant reduction of PCNA nuclear staining compared with the control group (Fig. 2A, second row). Aberrant expression of β-catenin can be considered as a key event during colorectal tumorigenesis and is linked to the increased transcription of a number of genes such as cyclin D1 (32, 33). β-Catenin was identified along the membrane of the epithelial cells in the control group. Compared with the control, all treatment groups showed marked inhibition of β-catenin membrane staining: sulindac (35.6% inhibition), naproxen (41.6% inhibition), atorvastatin (41.7% inhibition), atorvastatin + sulindac (59.4% inhibition), and atorvastatin + naproxen (54.6% inhibition; Fig. 2A, third row). The colonic crypt cells in the control group showed homogeneous and intense staining for β-catenin in the cytosol as well as in the membrane, with lower and scattered staining in the nucleus. In contrast, the tumors from the treatment groups had no observable nuclear staining. Furthermore, the cytoplasmic expression of β-catenin was also markedly inhibited by the treatment with atorvastatin alone and in combination with sulindac or naproxen (Fig. 2A, third row). Because cyclin D1 is a downstream signaling target of β-catenin, and overexpression of cyclin D1 is reported in patients with colorectal tumors where its lowering has therapeutic significance (32, 33), we determined whether treatment reduces cyclin D1 levels in colon tumors. Positive brownish staining of cyclin D1 in the control group predominantly localized in both the cytoplasm and nucleus. Administration of a low dose of atorvastatin, in combination with sulindac or naproxen, markedly reduced the staining for cyclin D1 in both the cytoplasm and the nucleus (Fig. 2A, fourth row).

Figure 2.

A, a low-dose atorvastatin (ATO), in combination with sulindac or naproxen, inhibits cell proliferation in colon adenocarcinomas. Hematoxylin and eosin (H&E) staining (first row) and PCNA staining (second row) of the colon tumors. β-Catenin (third row) and cyclin D1 (fourth row) staining was high in the cytosol and also present in the nucleus to a lower extent whereas nuclear staining was predominant with PCNA. Colon tumor sections were processed and incubated with the respective primary antibodies as described in Materials and Methods. B, a low-dose atorvastatin, in combination with sulindac or naproxen, reduces the expression of iNOS and COX-2 and decreases nuclear staining of phospho-p65 (p-p65) in colon adenocarcinomas. The colon tumor sections were processed and incubated with the respective primary antibodies as explained in Materials and Methods. iNOS and COX-2 showed cytoplasmic staining whereas nuclear staining was predominant with phospho-p65. n = 3 per group for each analysis. A representative section is shown. Image magnification, 400×.

Figure 2.

A, a low-dose atorvastatin (ATO), in combination with sulindac or naproxen, inhibits cell proliferation in colon adenocarcinomas. Hematoxylin and eosin (H&E) staining (first row) and PCNA staining (second row) of the colon tumors. β-Catenin (third row) and cyclin D1 (fourth row) staining was high in the cytosol and also present in the nucleus to a lower extent whereas nuclear staining was predominant with PCNA. Colon tumor sections were processed and incubated with the respective primary antibodies as described in Materials and Methods. B, a low-dose atorvastatin, in combination with sulindac or naproxen, reduces the expression of iNOS and COX-2 and decreases nuclear staining of phospho-p65 (p-p65) in colon adenocarcinomas. The colon tumor sections were processed and incubated with the respective primary antibodies as explained in Materials and Methods. iNOS and COX-2 showed cytoplasmic staining whereas nuclear staining was predominant with phospho-p65. n = 3 per group for each analysis. A representative section is shown. Image magnification, 400×.

Close modal

A low-dose atorvastatin, in combination with sulindac or naproxen, reduces the expression of inflammatory enzymes, iNOS, and COX-2 and decreases nuclear staining of phospho-p65 in colon adenocarcinomas

Overexpression of inflammatory markers is a hallmark of colorectal tumors (41, 42). This knowledge led us to examine the effects of long-term feeding of treatment with atorvastatin and NSAIDs on the inflammatory markers in the azoxymethane-injected rats. There was significant inhibition of the expression of iNOS and COX-2 proteins within the crypts in the adenocarcinomas from the treatment groups, compared with those from the control group (Fig. 2B). We also determined the effects of each treatment on a key nuclear factor kappaB (NF-κB) signaling molecule, p65, because NF-κB is an upstream factor of both iNOS and COX-2 transcription, and it is critical in the tumorigenesis where ablation of the proteins in this pathway caused the regression of tumors in animal models (43). The activated form of NF-κB subunit p65, that is, phospho-p65, is markedly reduced in the nucleus of the colon tumors from the treatment groups, when compared with those from the control group (Fig. 2B). In addition, significant inhibition of the iNOS enzyme activity was shown in tumors from naproxen, atorvastatin, and combination treatment groups. Importantly, combinational treatment of atorvastatin with sulindac or naproxen showed maximal inhibition on iNOS enzyme activity compared with single agents (Fig. 3). In summary, colon adenocarcinomas from atorvastatin and NSAIDs fed animals showed reduced expression of key inflammatory markers as well as nuclear staining for phospho-p65, a key molecule in the NF-κB pathway.

Figure 3.

Atorvastatin alone or in combination of sulindac or naproxen significantly suppress the iNOS enzyme activity in colon adenocarcinomas. The colon tumor samples were homogenized, and cytosolic extracts were subjected to assay for iNOS activity. The iNOS activity is shown as mean ± standard error (n = 6–8 per group). Control (CON); sulindac (SUL); naproxen (NAP); atorvastatin (ATO); sulindac + atorvastatin (SUL + ATO); and naproxen + atorvastatin (NAP + ATO).

Figure 3.

Atorvastatin alone or in combination of sulindac or naproxen significantly suppress the iNOS enzyme activity in colon adenocarcinomas. The colon tumor samples were homogenized, and cytosolic extracts were subjected to assay for iNOS activity. The iNOS activity is shown as mean ± standard error (n = 6–8 per group). Control (CON); sulindac (SUL); naproxen (NAP); atorvastatin (ATO); sulindac + atorvastatin (SUL + ATO); and naproxen + atorvastatin (NAP + ATO).

Close modal

A low-dose atorvastatin, in combination with sulindac or naproxen, inhibits colonic mucosal levels of cytokines TNF-α, IL-1β, and IL-4

Inflammatory cytokines are found to be present in human cancers including those of the colorectum, breast, prostate, and bladder (44, 45). The action of cytokines to facilitate carcinogenesis is multifold: DNA damage by reactive oxygen species and reactive nitrogen species; inhibition of DNA repair by reactive oxygen species; functional inactivation of tumor suppressor genes; tissue remodeling via activation of matrix metalloproteinases; and stimulation of angiogenesis and control of cell adhesion molecules (45). ELISA conducted for inflammatory cytokines on mucosal scrapings derived from the azoxymethane-injected rats are shown in Table 2. Sulindac treatment alone strongly inhibited the production of cytokines, TNF-α by 36.2% (P = 0.004), IL-1β by 26.6% (P = 0.04), and IL-4 by 34.0% (P = 0.03). More importantly, administration of a low-dose atorvastatin, in combination with sulindac or naproxen, significantly lowered the levels of cytokines in the colon; TNF-α by 36.8% (P = 0.03) and 33.3% (P = 0.01); IL-1β by 35.7% (P = 0.01) and 31.7% (P = 0.03); IL-4 by 39.9% (P = 0.03) and 40.4% (P = 0.01), respectively.

This is the first report on the combination treatment using low-dose atorvastatin with either low-dose sulindac or naproxen, which greatly suppress the colon adenocarcinoma incidence and multiplicity. Our results suggest that decreased inflammatory cytokines and signaling molecules, particularly inhibition of nuclear p65, β-catenin, and cyclin D1, are responsible for suppression of colonic adenocarcinomas. The present study is an extension of our previous work, which identified atorvastatin, sulindac, and naproxen as effective agents in suppressing colon cancer in animals (7, 16, 24). The results from the current research conducted in colon cancer reveal that administration of a low dose of atorvastatin, in combination with sulindac or naproxen, reduces the colon tumor multiplicity and regulates intermediate signaling pathways of proliferation and inflammation in the colon (Fig. 4).

Figure 4.

The Wnt/β-catenin and NF-κB pathways and their downstream targets in colon cancer. APC gene or activating mutations in β-catenin result in the accumulation of β-catenin and subsequent complex formation with TCF/LEF transcription factors. Excessive β-catenin can interact with TCF to activate transcription of proliferating genes, such as c-MYC and cyclin D1, in the colon. Inflammatory cytokines activate NF-κB by releasing p65, which is then translocalized to the nucleus, leading to increased transcription of target genes such as iNOS, COX-2, TNF-α, interleukins, and cyclin D1. Atorvastatin, in combination with sulindac or naproxen, targets the NF-κB and Wnt/β-catenin pathways and inhibits downstream signaling, iNOS, COX-2, cyclin D1, and others. LPR, low density lipoprotein receptor.

Figure 4.

The Wnt/β-catenin and NF-κB pathways and their downstream targets in colon cancer. APC gene or activating mutations in β-catenin result in the accumulation of β-catenin and subsequent complex formation with TCF/LEF transcription factors. Excessive β-catenin can interact with TCF to activate transcription of proliferating genes, such as c-MYC and cyclin D1, in the colon. Inflammatory cytokines activate NF-κB by releasing p65, which is then translocalized to the nucleus, leading to increased transcription of target genes such as iNOS, COX-2, TNF-α, interleukins, and cyclin D1. Atorvastatin, in combination with sulindac or naproxen, targets the NF-κB and Wnt/β-catenin pathways and inhibits downstream signaling, iNOS, COX-2, cyclin D1, and others. LPR, low density lipoprotein receptor.

Close modal

A comparison of tumor numbers across the different grades of tumor shows an overall reduction by the treatment with a low dose of atorvastatin, in combination with sulindac or naproxen. Importantly, statistical analysis on tumor data revealed a profound inhibitory effect of low-dose atorvastatin with either sulindac or naproxen on adenocarcinomas. Unlike the APCMin/+ mice, in which most of tumors are localized to the small intestine and those are predominantly adenomas, in the azoxymethane-induced rat, intestinal tumors are mostly localized to distal colon and adenocarcinomas, similar to human etiology. Thus, the results of our present study provide potential significance for human clinical trials. Also, it is important to note that in our previous studies, we have shown that 150 ppm atorvastatin inhibits colon adenocarcinoma incidence and multiplicity (∼34% inhibition, P ≤ 0.05) whereas, in this study, use of 200 ppm of atorvastatin suppressed more than 52% incidence (P = 0.005) and more than 58% multiplicity (P < 0.008) of colon adenocarcinomas. These results suggest that a modest dose increase in atorvastatin (from 150 ppm to 200 ppm) significantly enhances the chemopreventive efficacy.

Azoxymethane-induced tumors result from mutations in the Wnt/β-catenin pathway (28) as does the APCMin/+ model. However, unlike the APCMin/+ model, azoxymethane-induced tumors are caused by mutations in the β-catenin gene (29, 30). These mutations result in β-catenin stabilization and aberrant expression of β-catenin, which is considered as a key event during colon tumorigenesis (31). Immunohistochemical analysis revealed abundance of β-catenin mostly in the cytoplasm and relatively low nuclear staining in the adenocarcinomas of rats injected with azoxymethane whereas administration of a low dose of atorvastatin, in combination with sulindac or naproxen, markedly reduced the staining for β-catenin in both the cytoplasm and the nucleus (Fig. 2A).

Cyclin D1 is a very well-known cell-cycle protein targeted by β-catenin (35) and is known to be overexpressed in colonic tumors (32, 34). c-MYC is yet another important protein for cell proliferation regulated by β-catenin and Wnt pathway (38). These observations on cyclin D1 were corroborated by the potency of a low dose of atorvastatin, in combination with sulindac or naproxen, to affect the β-catenin levels in the colon tumors. In our studies, we identified a low dose of atorvastatin, in combination with sulindac or naproxen, to significantly lower the levels of cyclin D1 in the colon tumors induced with azoxymethane (Fig. 2A). More importantly, nuclear levels of β-catenin and cyclin D1 are reported to play more important role in tumorigenesis than the total protein levels (46, 47). In our studies, a low dose of atorvastatin, in combination with sulindac or naproxen, reduced the levels of cyclin D1 and β-catenin in the nucleus (Fig. 2A).

In addition to the effects on β-catenin and cell proliferation, our results indicate the anti-inflammatory property of a low dose of atorvastatin, in combination with sulindac or naproxen. We observed marked reduction in the staining intensities for iNOS, COX-2, and phospho-p65 markers as well as for the iNOS enzyme activity in colon tumors from the combination treatment groups (Figs. 2B and 3). Mucosal levels of inflammatory cytokines, such as TNF-α, IL-1β and IL4, were also significantly downregulated by a low dose of atorvastatin, in combination with sulindac or naproxen (Table 2). Several anti-inflammatory agents that target the nitric oxide or the prostaglandin pathway are reported to present chemopreventive action in the colon (4, 7, 48). A clinical trial on celecoxib, the selective COX-2 inhibitor, at a dose of 400 mg daily reduced advanced adenoma formation in the colon by almost 50% compared with the placebo through a 3-year treatment period (49). In addition to anti-inflammatory and antiproliferative mechanisms, azoxymethane treatment may also generate oxidative stress and significant genotoxicity by inducing methyl–DNA adducts; however, these processes may significantly subside within 12 to 18 hours after the azoxymethane treatment. In this study, we administered chemopreventive agents 1 week after the carcinogen treatment and thus possibility of carcinogenic action of azoxymethane via inhibition of oxidative stress or DNA adducts is very minimal to none by the chemopreventive agents. Promising results with other agents, such as the use of low concentrations of difluoromethylornithine and sulindac as chemopreventive agents in colorectal cancer, highlight the potential role of inflammation in its pathogenesis and the importance of combination strategies (48).

In conclusion, a low dose of atorvastatin, in combination with sulindac or naproxen, inhibits profoundly colon tumorigenesis by regulating the p65/β-catenin/cyclin D1 signaling pathway and the inflammatory responses. Overall, the data indicate that a low dose of atorvastatin, in combination with sulindac or naproxen, holds great promise in the field of colon cancer chemoprevention in humans.

No potential conflicts of interest were disclosed.

The authors thank Maria Hyra and Lamberto R. Navoa of the Animal Facility in the Department of Chemical Biology for their technical assistance in taking care of the animals.

This work was supported by NCI-N01-CN-53300, R01-CA94962, and the Trustees Research Fellowship Program at Rutgers, The State University of New Jersey.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Jemal
A
,
Siegel
R
,
Xu
J
,
Ward
E
. 
Cancer statistics, 2010
.
CA Cancer J Clin
2010
;
60
:
277
300
.
2.
Jemal
A
,
Bray
F
,
Center
MM
,
Ferlay
J
,
Ward
E
,
Forman
D
. 
Global cancer statistics
.
CA Cancer J Clin
2011
;
61
:
69
90
.
3.
Center
MM
,
Jemal
A
,
Ward
E
. 
International trends in colorectal cancer incidence rates
.
Cancer Epidemiol Biomarkers Prev
2009
;
18
:
1688
94
.
4.
Half
E
,
Arber
N
. 
Colon cancer: preventive agents and the present status of chemoprevention
.
Expert Opin Pharmacother
2009
;
10
:
211
9
.
5.
Kawamori
T
,
Takahashi
M
,
Watanabe
K
,
Ohta
T
,
Nakatsugi
S
,
Sugimura
T
, et al
Suppression of azoxymethane-induced colonic aberrant crypt foci by a nitric oxide synthase inhibitor
.
Cancer Lett
2000
;
148
:
33
7
.
6.
Pereg
D
,
Lishner
M
. 
Non-steroidal anti-inflammatory drugs for the prevention and treatment of cancer
.
J Intern Med
2005
;
258
:
115
23
.
7.
Rao
CV
,
Indranie
C
,
Simi
B
,
Manning
PT
,
Connor
JR
,
Reddy
BS
. 
Chemopreventive properties of a selective inducible nitric oxide synthase inhibitor in colon carcinogenesis, administered alone or in combination with celecoxib, a selective cyclooxygenase-2 inhibitor
.
Cancer Res
2002
;
62
:
165
70
.
8.
Takahashi
M
,
Wakabayashi
K
. 
Gene mutations and altered gene expression in azoxymethane-induced colon carcinogenesis in rodents
.
Cancer Sci
2004
;
95
:
475
80
.
9.
DuBois
RN
,
Radhika
A
,
Reddy
BS
,
Entingh
AJ
. 
Increased cyclooxygenase-2 levels in carcinogen-induced rat colonic tumors
.
Gastroenterology
1996
;
110
:
1259
62
.
10.
Ohta
T
,
Takahashi
M
,
Ochiai
A
. 
Increased protein expression of both inducible nitric oxide synthase and cyclooxygenase-2 in human colon cancers
.
Cancer Lett
2006
;
239
:
246
53
.
11.
Takahashi
M
,
Mutoh
M
,
Shoji
Y
,
Sato
H
,
Kamanaka
Y
,
Naka
M
, et al
Suppressive effect of an inducible nitric oxide inhibitor, ONO-1714, on AOM-induced rat colon carcinogenesis
.
Nitric Oxide
2006
;
14
:
130
6
.
12.
Kawamori
T
,
Rao
CV
,
Seibert
K
,
Reddy
BS
. 
Chemopreventive activity of celecoxib, a specific cyclooxygenase-2 inhibitor, against colon carcinogenesis
.
Cancer Res
1998
;
58
:
409
12
.
13.
Reddy
BS
,
Rao
CV
. 
Novel approaches for colon cancer prevention by cyclooxygenase-2 inhibitors
.
J Environ Pathol Toxicol Oncol
. 
2002
;
21
:
155
64
.
14.
Lala
PK
,
Chakraborty
C
. 
Role of nitric oxide in carcinogenesis and tumour progression
.
Lancet Oncol
2001
;
2
:
149
56
.
15.
Rao
CV
,
Reddy
BS
,
Steele
VE
,
Wang
CX
,
Liu
X
,
Ouyang
N
, et al
Nitric oxide-releasing aspirin and indomethacin are potent inhibitors against colon cancer in azoxymethane-treated rats: effects on molecular targets
.
Mol Cancer Ther
2006
;
5
:
1530
8
.
16.
Reddy
BS
,
Wang
CX
,
Kong
AN
,
Khor
TO
,
Zheng
X
,
Steele
VE
, et al
Prevention of azoxymethane-induced colon cancer by combination of low doses of atorvastatin, aspirin, and celecoxib in f 344 rats
.
Cancer Res
2006
;
66
:
4542
6
.
17.
Rao
CV
,
Kawamori
T
,
Hamid
R
,
Reddy
BS
. 
Chemoprevention of colonic aberrant crypt foci by an inducible nitric oxide synthase-selective inhibitor
.
Carcinogenesis
1999
;
20
:
641
4
.
18.
Chen
T
,
Hwang
H
,
Rose
ME
,
Nines
RG
,
Stoner
GD
. 
Chemopreventive properties of black raspberries in N-nitrosomethylbenzylamine-induced rat esophageal tumorigenesis: down-regulation of cyclooxygenase-2, inducible nitric oxide synthase, and c-Jun
.
Cancer Res
2006
;
66
:
2853
9
.
19.
Reddy
BS
,
Hirose
Y
,
Cohen
LA
,
Simi
B
,
Cooma
I
,
Rao
CV
. 
Preventive potential of wheat bran fractions against experimental colon carcinogenesis: implications for human colon cancer prevention
.
Cancer Res
2000
;
60
:
4792
7
.
20.
Rao
CV
,
Rivenson
A
,
Simi
B
,
Zang
E
,
Kelloff
G
,
Steele
V
,
Reddy
BS
. 
Chemoprevention of colon carcinogenesis by sulindac, a nonsteroidal anti-inflammatory agent
.
Cancer Res
1995
;
55
:
1464
72
.
21.
Rao
CV
. 
Nitric oxide signaling in colon cancer chemoprevention
.
Mutat Res
2004
;
555
:
107
19
.
22.
Luceri
C
,
Caderni
G
,
Sanna
A
,
Dolara
P
. 
Red wine and black tea polyphenols modulate the expression of cycloxygenase-2, inducible nitric oxide synthase and glutathione-related enzymes in azoxymethane-induced f344 rat colon tumors
.
J Nutr
2002
;
132
:
1376
9
.
23.
Reddy
BS
,
Kawamori
T
,
Lubet
RA
,
Steele
VE
,
Kelloff
GJ
,
Rao
CV
. 
Chemopreventive efficacy of sulindac sulfone against colon cancer depends on time of administration during carcinogenic process
.
Cancer Res
1999
;
59
:
3387
91
.
24.
Swamy
MV
,
Patlolla
JM
,
Steele
VE
,
Kopelovich
L
,
Reddy
BS
,
Rao
CV
. 
Chemoprevention of familial adenomatous polyposis by low doses of atorvastatin and celecoxib given individually and in combination to APCMin mice
.
Cancer Res
2006
;
66
:
7370
7
.
25.
Steele
VE
,
Rao
CV
,
Zhang
Y
,
Patlolla
J
,
Boring
D
,
Kopelovich
L
, et al
Chemopreventive efficacy of naproxen and nitric oxide-naproxen in rodent models of colon, urinary bladder, and mammary cancers
.
Cancer Prev Res
2009
;
2
:
951
6
.
26.
Rao
KV
,
Detrisac
CJ
,
Steele
VE
,
Hawk
ET
,
Kelloff
GJ
,
McCormick
DL
. 
Differential activity of aspirin, ketoprofen and sulindac as cancer chemopreventive agents in the mouse urinary bladder
.
Carcinogenesis
1996
;
17
:
1435
8
.
27.
Samaha
HS
,
Kelloff
GJ
,
Steele
V
,
Rao
CV
,
Reddy
BS
. 
Modulation of apoptosis by sulindac, curcumin, phenylethyl-3-methylcaffeate, and 6-phenylhexyl isothiocyanate: apoptotic index as a biomarker in colon cancer chemoprevention and promotion
.
Cancer Res
1997
;
57
:
1301
5
.
28.
Takahashi
M
,
Nakatsugi
S
,
Sugimura
T
,
Wakabayashi
K
. 
Frequent mutations of the {beta}-catenin gene in mouse colon tumors induced by azoxymethane
.
Carcinogenesis
2000
;
21
:
1117
20
.
29.
Kaiser
S
,
Park
Y-K
,
Franklin
J
,
Halberg
R
,
Yu
M
,
Jessen
W
, et al
Transcriptional recapitulation and subversion of embryonic colon development by mouse colon tumor models and human colon cancer
.
Genome Biol
2007
;
8
:
R131
.
30.
Wang
QS
,
Papanikolaou
A
,
Sabourin
CL
,
Rosenberg
DW
. 
Altered expression of cyclin D1 and cyclin-dependent kinase 4 in azoxymethane-induced mouse colon tumorigenesis
.
Carcinogenesis
1998
;
19
:
2001
6
.
31.
Morin
PJ
,
Sparks
AB
,
Korinek
V
,
Barker
N
,
Clevers
H
,
Vogelstein
B
, et al
Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC
.
Science
1997
;
275
:
1787
90
.
32.
Arber
N
,
Hibshoosh
H
,
Moss
SF
,
Sutter
T
,
Zhang
Y
,
Begg
M
, et al
Increased expression of cyclin D1 is an early event in multistage colorectal carcinogenesis
.
Gastroenterology
1996
;
110
:
669
74
.
33.
Alao
JP
. 
The regulation of cyclin D1 degradation: roles in cancer development and the potential for therapeutic invention
.
Mol Cancer
2007
;
6
:
24
.
34.
Zhang
T
,
Nanney
LB
,
Luongo
C
,
Lamps
L
,
Heppner
KJ
,
DuBois
RN
, et al
Concurrent overexpression of cyclin D1 and cyclin-dependent kinase 4 (Cdk4) in intestinal adenomas from multiple intestinal neoplasia (Min) mice and human familial adenomatous polyposis patients
.
Cancer Res
1997
;
57
:
169
75
.
35.
Tetsu
O
,
McCormick
F
. 
Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells
.
Nature
1999
;
398
:
422
6
.
36.
Giles
RH
,
van Es
JH
,
Clevers
H
. 
Caught up in a Wnt storm: Wnt signaling in cancer
.
Biochim Biophys Acta
2003
;
1653
:
1
24
.
37.
Klaus
A
,
Birchmeier
W
. 
Wnt signalling and its impact on development and cancer
.
Nat Rev Cancer
2008
;
8
:
387
98
.
38.
He
T-C
,
Sparks
AB
,
Rago
C
,
Hermeking
H
,
Zawel
L
,
da Costa
LT
, et al
Identification of c-MYC as a target of the APC pathway
.
Science
1998
;
281
:
1509
12
.
39.
Ulrich
CM
,
Bigler
J
,
Potter
JD
. 
Non-steroidal anti-inflammatory drugs for cancer prevention: promise, perils and pharmacogenetics
.
Nat Rev Cancer
2006
;
6
:
130
40
.
40.
Suh
N
,
Paul
S
,
Hao
X
,
Simi
B
,
Xiao
H
,
Rimando
AM
, et al
Pterostilbene, an active constituent of blueberries, suppresses aberrant crypt foci formation in the azoxymethane-induced colon carcinogenesis model in rats
.
Clin Cancer Res
2007
;
13
:
350
5
.
41.
Feagins
LA
,
Souza
RF
,
Spechler
SJ
Carcinogenesis in IBD: potential targets for the prevention of colorectal cancer
.
Nat Rev Gastroenterol Hepatol
2009
;
6
:
297
305
.
42.
Fantini
MC
,
Pallone
F
, 
Cytokines: from gut inflammation to colorectal cancer
.
Curr Drug Targets
2008
;
9
:
375
80
.
43.
Karin
M
. 
Nuclear factor-kappaB in cancer development and progression
.
Nature
2006
;
441
:
431
6
.
44.
Kundu
JK
,
Surh
YJ
. 
Inflammation: gearing the journey to cancer
.
Mutat Res
2008
;
659
:
15
30
.
45.
Balkwill
F
,
Mantovani
A
. 
Inflammation and cancer: back to Virchow?
Lancet
2001
;
357
:
539
.
46.
Nelson
WJ
,
Nusse
R
. 
Convergence of Wnt, beta-catenin, and cadherin pathways
.
Science
2004
;
303
:
1483
7
.
47.
Kim
JK
,
Diehl
JA
. 
Nuclear cyclin D1: an oncogenic driver in human cancer
.
J Cell Physiol
2009
;
220
:
292
6
.
48.
Reddy
BS
. 
Strategies for colon cancer prevention: combination of chemopreventive agents
.
Subcell Biochem
2007
;
42
:
213
25
.
49.
Arber
N
,
Eagle
CJ
,
Spicak
J
,
Racz
I
,
Dite
P
,
Hajer
J
, et al
Celecoxib for the prevention of colorectal adenomatous polyps
.
N Engl J Med
2006
;
355
:
885
95
.