Purpose: Colorectal cancer is a worldwide cancer with rising annual incidence. Inflammation is a well-known cause of colorectal cancer carcinogenesis. Metabolic inflammation (metaflammation) and altered gut microbiota (dysbiosis) have contributed to colorectal cancer. Chemoprevention is an important strategy to reduce cancer-related mortality. Recently, various polypharmacologic molecules that dually inhibit histone deacetylases (HDAC) and other therapeutic targets have been developed.

Experimental Design: Prevention for colitis was examined by dextran sodium sulfate (DSS) mouse models. Prevention for colorectal cancer was examined by azoxymethane/dextran sodium sulfate (AOM/DSS) mouse models. Immunohistochemical staining was utilized to analyze the infiltration of macrophages and neutrophils and COX-II expression in mouse tissue specimens. The endotoxin activity was evaluated by Endotoxin Activity Assay Kit.

Results: We synthesized a statin hydroxamate that simultaneously inhibited HDAC and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR). Its preventive effect on colitis and colitis-associated colorectal cancer in mouse models was examined. Oral administration of this statin hydroxamate could prevent acute inflammation in the DSS-induced colitis and AOM/DSS–induced colorectal cancer with superior activity than the combination of lovastatin and SAHA. It also reduced proinflammatory cytokines, chemokines, expression of COX-II, and cyclin D1 in inflammation and tumor tissues, as well as decreasing the infiltration of macrophages and neutrophils in tumor-surrounding regions. Stemness of colorectal cancer and the release of endotoxin in AOM/DSS mouse models were also attenuated by this small molecule.

Conclusions: This study demonstrates that the polypharmacological HDAC inhibitor has promising effect on the chemoprevention of colorectal cancer, and serum endotoxin level might serve as a potential biomarker for its chemoprevention. Clin Cancer Res; 22(16); 4158–69. ©2016 AACR.

Translational Relevance

Colorectal cancer is a worldwide cancer, causing significant morbidity and mortality, and chemoprevention is one of the potentials to reduce cancer-related mortality. Here, we design a polypharmacologic small molecule, statin hydroxamate, that dually inhibits histone deacetylases (HDAC) and 3-hydroxy-3-methylglutaryl coenzyme A reductase. It attenuated dextran sodium sulfate (DSS)–induced colitis and azoxymethane/DSS (AOM/DSS)–induced colorectal cancer in mice through reducing proinflammatory cytokines, chemokines, expression of COX-II, and cyclin D1 in inflammation and tumor tissues, as well as decreasing the infiltration of macrophages and neutrophils in tumor-surrounding regions. Stemness of Colorectal cancer and the release of endotoxin in AOM/DSS mouse models were also attenuated by this small molecule. These results demonstrate that the polypharmacologic HDAC inhibitor has promising effect on the chemoprevention of Colorectal cancer, and serum endotoxin level might serve as a potential biomarker for its chemoprevention.

Colorectal cancer is a worldwide cancer, causing significant morbidity and mortality, and chemoprevention is one of the potentials to reduce cancer-related mortality by suppressing the initial phase of carcinogenesis or the progression of premalignant cells (1). Meta-analysis studies have shown that aspirin could reduce the incidence of colon polyps and cancer (2). The mechanisms were in part through inhibition of COX-II–related pathways and normalizing EGFR expression (3). However, adverse effects prohibit its clinical use. Thus, new strategy for chemoprevention of colorectal cancer meets an unmet medical need.

Inflammation through inflammation–dysplasia–carcinoma process is an important cause of carcinogenesis to induce colorectal cancer (4, 5). Inflammatory bowel disease (IBD) referring to both Crohn's disease and ulcerative colitis is a chronic disorder. Patients with IBD exhibiting a 2- to 8-fold higher risk of colorectal cancer, is a classic example to highlight the association between chronic inflammation (colitis) and colorectal cancer (4, 6). In addition, both sporadic and heritable colorectal cancer are also related to inflammation (6, 7). Metabolic syndrome (i.e., hypertension, type II diabetes, hyperlipidemia, and obesity) commonly resulted from excess nutrients and energy has been found to induce a low-grade, chronic, metabolically linked inflammatory state termed metaflammation (8). Patients having metabolic syndromes show higher incidence of colorectal cancer and higher recurrence rate of colon adenoma that demonstrated the close association between inflammation and colorectal cancer (8). Epidemiologic studies reported that anti-inflammatory medications effectively reduce the incidence of colorectal cancer among IBD patients (9, 10). Therefore, inhibition of inflammation is a potential strategy to prevent cancer initiation.

Huge amount of bacteria flora inhabited in the intestine interacting with mucosa maintaining gut immunity and homeostasis (11). Recent studies suggest that imbalance of bacterial flora, termed dysbiosis, favors higher abundance of gram-negative bacteria (GNB; refs. 4, 12, 13). Endotoxin, which is a lipopolysaccharide (LPS) from the cell wall of GNB, is associated with various chronic diseases, such as atherosclerosis, diabetes, and cancers (13). The release of endotoxin by GNB is an important mechanism of dysbiosis-induced colorectal cancer (14). In addition to endotoxin, cancer stem cell (CSC) is another crucial factor for colorectal cancer initiation (15). Chronic gut inflammation leads to crypt damage and repeated regeneration, resulting in CSC expansion leading to colorectal tumorigenesis (16, 17).

Statins are 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase (HMGR) inhibitors treating hypercholesterolemia in patients with coronary artery and atherosclerotic diseases (18, 19). They have also shown promise in cancer prevention from observational and preclinical and certain aspects of randomized controlled studies (20). We first demonstrated statins with a carboxylic acid–containing long chain possessing the structure similar to hydroxamate that also inhibits histone deacetylase (HDAC) activity, which is an attractive target against cancer (21, 22). To further improve statins' activity against HDAC, three statin hydroxamates were designed to inhibit class I and II HDACs and HMGR (23). We named compound 12, 13, and 14 as JMF3086, 3171, and 3173, respectively, in this study. Recently, the strategy of developing polypharmacological molecules that dually inhibit HDACs and other therapeutic targets has been reported (24).

Oral administration of JMF3086 attenuated dextran sodium sulfate (DSS)–induced colitis and azoxymethane (AOM)/DSS–induced colorectal cancer in mice. A positive correlation between HMGR and HDAC activity and prevention of colorectal cancer was seen. JMF3086 exerted anti-inflammatory effect and inhibited the in vivo release of endotoxin. Furthermore, the stemness of colorectal cancer was inhibited in vivo. These results show that our polypharmacological molecule JMF3086 targeting HDACs and HMGR possesses great potential for the prevention of colitis and colitis-associated colorectal cancer.

Reagents

Lovastatin was obtained from Lotus Pharmaceutical Co. SAHA was obtained and purchased from Merck. Anti-acetyl-histone H3 and anti-acetyl-histone H4 antibodies were obtained from Millipore. Anti-acetyl-tubulin antibody was obtained from Sigma. Anti-PARP1/2 antibody was purchased from Santa Cruz Biotechnology. Anti-caspase-3 antibody and anti-BAX antibody were purchased from Cell Signaling Technology. Anti-β-actin antibody was purchased from GeneTex. HDAC Fluorometric Assay/Drug Discovery Kit (AK-500) was purchased from Biomol. HMG-CoA reductase activity kit (CS-1090) and AOM were purchased from Sigma. DSS was purchased from MP Biomedicals. Endotoxin Activity Assay Kit was purchased from Spectral Diagnostics.

Mice

C57BL/6, BALB/c, and NOD/SCID mice were obtained from the National Laboratory Animal Center (Taipei City, Taiwan). Mice were performed in accordance with protocols approved by the Institutional Animal Care and Use Committee of the College of Medicine, National Taiwan University (Taipei, Taiwan).

Animal models of DSS-induced acute colitis

The DSS-induced colitis animal model exhibits many phenotypes that are relevant to human ulcerative colitis, including inflammation and ulceration of the colonic mucosa (4). Colitis was induced in seven-week-old male C57BL/6 mice by adding DSS to their drinking water for 5 days, followed by switching to regular drinking water. In the preventive model, test compounds were dissolved in corn oil and administered orally 7 days before, during, and after 3.5% (w/v) DSS treatment (Fig. 1A). In the therapeutic model, test compounds were dissolved in corn oil and administered orally 5 days following the 2.5% (w/v) DSS treatment (Fig. 2A).

Figure 1.

JMF3086 prevents acute colitis in vivo. A, schematic overview of the experimental design. B, changes in body weights (top), clinical bleeding scores (middle), and clinical diarrhea scores (bottom) are shown. *, P < 0.05; **, P < 0.01 versus vehicle. C, representative whole colons (top) and colon length (bottom) are shown. Control mice received distilled water only. Scale bar, 2 cm. ##, P < 0.01 versus control; **, P < 0.01 versus vehicle. D, colon sections were counterstained with H&E, and high-magnification images of the black-boxed area are presented in the bottom row. Scale bar, 250 μm. E, colon sections were immunostained with anti-F4/80, anti-Ly-6G, and anti-COX-II antibodies. Scale bar, 250 μm. F, the level of various cytokines in colon culture supernatants was seen. ##, P < 0.01 versus control; *, P < 0.05 and **, P < 0.01 versus vehicle.

Figure 1.

JMF3086 prevents acute colitis in vivo. A, schematic overview of the experimental design. B, changes in body weights (top), clinical bleeding scores (middle), and clinical diarrhea scores (bottom) are shown. *, P < 0.05; **, P < 0.01 versus vehicle. C, representative whole colons (top) and colon length (bottom) are shown. Control mice received distilled water only. Scale bar, 2 cm. ##, P < 0.01 versus control; **, P < 0.01 versus vehicle. D, colon sections were counterstained with H&E, and high-magnification images of the black-boxed area are presented in the bottom row. Scale bar, 250 μm. E, colon sections were immunostained with anti-F4/80, anti-Ly-6G, and anti-COX-II antibodies. Scale bar, 250 μm. F, the level of various cytokines in colon culture supernatants was seen. ##, P < 0.01 versus control; *, P < 0.05 and **, P < 0.01 versus vehicle.

Close modal
Figure 2.

JMF3086 treats acute colitis in vivo. A, schematic overview of the experimental design. B, changes in body weights (top), clinical bleeding scores (middle), and clinical diarrhea scores (bottom) are shown. *, P < 0.05; **, P < 0.01; and ***, P < 0.001 versus vehicle. C, representative whole colons (top) and colon length (bottom) are shown. Control mice received distilled water only. Scale bar, 2 cm. D, colon sections were counterstained with H&E, and high-magnification images of the black-boxed area are presented in the bottom row. Scale bar, 100 μm. E, colon sections were immunostained with anti-Ly-6G antibodies. Scale bar, 100 μm. F, the level of various cytokines in colon culture supernatants was seen. ##, P < 0.01 versus control; *, P < 0.05 versus vehicle. GM-CSF, granulocyte macrophage colony-stimulating factor.

Figure 2.

JMF3086 treats acute colitis in vivo. A, schematic overview of the experimental design. B, changes in body weights (top), clinical bleeding scores (middle), and clinical diarrhea scores (bottom) are shown. *, P < 0.05; **, P < 0.01; and ***, P < 0.001 versus vehicle. C, representative whole colons (top) and colon length (bottom) are shown. Control mice received distilled water only. Scale bar, 2 cm. D, colon sections were counterstained with H&E, and high-magnification images of the black-boxed area are presented in the bottom row. Scale bar, 100 μm. E, colon sections were immunostained with anti-Ly-6G antibodies. Scale bar, 100 μm. F, the level of various cytokines in colon culture supernatants was seen. ##, P < 0.01 versus control; *, P < 0.05 versus vehicle. GM-CSF, granulocyte macrophage colony-stimulating factor.

Close modal

Animal model of AOM/DSS–induced colorectal cancer

AOM in conjunction with the additional inflammatory stimulus of DSS results in tumor development that is restricted to the colon in mice (4, 6). This model is widely utilized to recapitulate human colorectal cancer because it results in inflammation and ulceration of the entire colon, similar to what is observed in patients (6). Colorectal cancer was induced in male C57BL/6 mice via the intraperitoneal injection of AOM (12.5 mg/kg) while the mice were maintained with a regular diet and drinking water for 7 days and then subjecting the mice to 3 cycles of DSS treatment, with each cycle consisting of the administration of 3.5% DSS for 5 days, followed by a 14-day recovery period with regular water (Fig. 3A). In the preventive model of AOM/DSS–induced colorectal cancer, test compounds were dissolved in corn oil and administered orally 5 days a week for 8 weeks, simultaneously with AOM exposure.

Figure 3.

Preventive effect of JMF3086 on the colitis symptoms in AOM/DSS mouse models. A, experimental timeline for AOM/DSS–induced colorectal cancer mouse model. B, changes in body weights (top), clinical bleeding scores (middle), and clinical diarrhea scores (bottom) are shown. Error bars, SE. *, P < 0.05; **, P < 0.01 versus vehicle; two-sided student t test. C, representative whole colons (top) and colon length (bottom) are shown. Scale bar, 2 cm. Error bars, SE. *, P < 0.05; **, P < 0.01 versus vehicle; ##, P < 0.01 versus control; two-sided student t test.

Figure 3.

Preventive effect of JMF3086 on the colitis symptoms in AOM/DSS mouse models. A, experimental timeline for AOM/DSS–induced colorectal cancer mouse model. B, changes in body weights (top), clinical bleeding scores (middle), and clinical diarrhea scores (bottom) are shown. Error bars, SE. *, P < 0.05; **, P < 0.01 versus vehicle; two-sided student t test. C, representative whole colons (top) and colon length (bottom) are shown. Scale bar, 2 cm. Error bars, SE. *, P < 0.05; **, P < 0.01 versus vehicle; ##, P < 0.01 versus control; two-sided student t test.

Close modal

Clinical assessment of DSS-induced acute colitis and AOM/DSS–induced colorectal cancer

Body weight, the presence of occult or gross blood in the rectum, and stool consistency were determined daily in the mice. The weight change during the experiment was calculated as the percent change in weight compared with the baseline measurement. Bleeding was scored as 0, when there was no blood in the Hemoccult test; 1, for a positive Hemoccult result; 2, for slight bleeding; or 3, for gross bleeding. Regarding stool consistency, 0 points were given for well-formed pellets, 1 point for semiformed stools that did not adhere to the anus, 2 points for pasty stools, and 3 points for liquid stools that adhered to the anus.

HDAC activity assay

The HDAC activity was performed using HDAC Fluorescent Activity Assay Kit. Total lysate from mouse tissue was mixed with Fluor-de-Lys substrate for 10 minutes at 37°C, followed by adding developer to stop the reaction. Fluorescence was measured by fluorometric reader with excitation at 360 nm and emission at 460 nm. The IC50 values were calculated from SigmaPlot software.

HMG-CoA reductase activity assay

The HMG-CoA reductase activity was performed using HMG-CoA Reductase Assay Kit. Total lysate from mouse tissue was mixed with NADPH and HMG-CoA and incubated for 5 minutes at 37°C. The absorbance at 340 nm was measured. The IC50 values were calculated from SigmaPlot software.

IHC assay

IHC was performed with One Step Polymer-HRP Detection Kit (BioGenex) on sections from 10% paraffin-embedded samples according to the manufacturer's protocols. Pictures were acquired using TissueFAXS (TissueGnostics), and the results of DAB-positive cells were presented as scattergrams plot using HistoQuest software (TissueGnostics). The cut-off values for background staining were chosen manually using the forward/backward gating tools of the software.

Endotoxin activity assay

The endotoxin activity was performed using Endotoxin Activity Assay Kit. Serum from mouse whole blood was mixed with control standard endotoxin and limulus amoebocyte lysate reagent water at 37°C. The absorbance at 405 nm was measured.

Statistical analysis

The SPSS program (SPSS Inc.) was used for all statistical analyses. Statistical analysis was performed by two-sided Student t test. Data shown were representative of at least three independent experiments. Quantitative data are presented as means ± SD.

Additional methods

Detailed methodology is described in the Supplementary Material.

Statin hydroxamates prevent inflammation in the DSS-induced colitis

As inhibition of inflammation is a potential strategy to prevent cancer initiation, the acute colitis induced by DSS in C57BL/6 mouse model was employed to evaluate the anti-inflammation efficacy of JMF3086. JMF3086 (50 mg/kg) was orally administered for 7 days before treatment with 3.5% DSS began and was continued until the end of the experiment (Fig. 1A). It prevented the colitis symptoms (i.e., body weight loss, diarrhea, and rectal bleeding) and was more effective than the combination of lovastatin and SAHA (Fig. 1B).

The reduction in colon length that occurred in DSS-treated mice was prevented by JMF3086 (Fig. 1C). Histopathologically, DSS-treated mice exhibited complete destruction of the epithelial architecture with a loss of crypts and epithelial integrity and significant infiltration of neutrophils (Ly-6G) but not of macrophages (F4/80). In JMF3086-treated mice, the crypt architecture was maintained, and only a mild infiltration of neutrophils in the mucosa was detected (Fig. 1D and E). Furthermore, elevated COX-II expression was observed in the epithelial cells of DSS-treated mice, and this increase was prevented by JMF3086 (Fig. 1E). DSS-induced acute colitis is predominantly characterized by a cytokine response (25). TNFα, IFNγ, IL6, and IL12 secretion from colons in the DSS-treated mice were found to be markedly increased and prevented by JMF3086 treatment (Fig. 1F).

The preventive effect of JMF3171 and JMF3173 on DSS-induced colitis was also examined and found to exert nice efficacy in colitis symptoms, as well as the reductions of colon length, infiltration of neutrophils, destruction of the epithelial architecture, and cytokine releases (Supplementary Fig. S1).

JMF3086 treats DSS-induced colitis

We further examined the efficacy of JMF3086 in treating DSS-induced colitis in mouse models. The colitis symptoms were inhibited by JMF3086, which was more effective than the combination of lovastatin and SAHA (Fig. 2B and C). Microscopically, JMF3086 reduced DSS-induced colonic ulceration (Fig. 2D). Furthermore, infiltration of neutrophils as well as the secretion of proinflammatory cytokines and chemokines in the colons was attenuated by JMF3086 (Fig. 2E and F). Elevated serum endotoxin level found in DSS mice was reduced by JMF3086 (Supplementary Fig. S2).

JMF3086 attenuates the colitis symptoms and tumor growth in AOM/DSS–induced colorectal cancer mouse models

As JMF3086 could prevent and treat acute inflammation in the DSS-induced colitis in mice, the chemoprevention of JMF3086 was further evaluated in AOM/DSS–induced colorectal cancer mouse models. JMF3086 (25 or 50 mg/kg) was administered orally 5 days per week concurrent with AOM injections (Fig. 3A). The effect of lovastatin (50 mg/kg) and SAHA (50 mg/kg) either individually or in combination was compared. Symptomatic parameters, such as loss of body weight, diarrhea, and rectal bleeding, were observed in mice after AOM/DSS treatment, and JMF3086 prevented the occurrence of these symptoms (Fig. 3B). Colon length was decreased in AOM/DSS–treated mice, but not in those treated with JMF3086 (Fig. 3C).

Polyps and colon tumors were macroscopically observed and counted, and H&E staining revealed adenocarcinomas with dysplasia in AOM/DSS–treated mice (Fig. 4A and B and Supplementary Fig. S3A). JMF3086 at a dose of 50 mg/kg decreased tumor numbers and sizes and protected against AOM/DSS–induced dysplasia/adenocarcinoma lesion to surface tumor necrosis (Fig. 4A and B).

Figure 4.

Preventive effect of JMF3086 on tumor growth in AOM/DSS mouse models. A, representative terminal colons (top) and number of tumors per mouse and total tumor size (bottom) are shown. *, P < 0.05; **, P < 0.01 versus vehicle. B, representative of terminal colon sections counterstained with H&E, and high-magnification images of the black-boxed area are presented in the bottom row. Scale bar, 250 μm. C, HMGR and HDAC activities in cell lysates from colon tissues are shown. ##, P < 0.01 versus control; *, P < 0.05; and **, P < 0.01 versus vehicle. D, correlations between HDAC activity, HMGR activity, and the number of tumors and tumor sizes are shown.

Figure 4.

Preventive effect of JMF3086 on tumor growth in AOM/DSS mouse models. A, representative terminal colons (top) and number of tumors per mouse and total tumor size (bottom) are shown. *, P < 0.05; **, P < 0.01 versus vehicle. B, representative of terminal colon sections counterstained with H&E, and high-magnification images of the black-boxed area are presented in the bottom row. Scale bar, 250 μm. C, HMGR and HDAC activities in cell lysates from colon tissues are shown. ##, P < 0.01 versus control; *, P < 0.05; and **, P < 0.01 versus vehicle. D, correlations between HDAC activity, HMGR activity, and the number of tumors and tumor sizes are shown.

Close modal

To examine whether HMGR and HDAC were inhibited by JMF3086 in vivo, their activity was analyzed in cell lysates from colon tissue and found to be increased in AOM/DSS–induced colorectal cancer. JMF3086 inhibited these effects and induced acetylations of histone H3, H4, and tubulin. Its accumulation in colon tissues detected by LC/MS-MS and ESI-MS analyses was seen (Fig. 4C and Supplementary Figs. S3B and S4A–S4C). There was a positive correlation between HMGR and HDAC activity and tumor number and size (Fig. 4D), indicating that inhibition of both activities contributed to the prevention of JMF3086 on AOM/DSS–induced colorectal cancer in mice.

JMF3086 reduces the mRNA expression of proinflammatory cytokines/chemokines and intracolonic infiltration of macrophages and neutrophils in AOM/DSS–induced colorectal cancer mouse models

JMF3086 potently decreased the mRNA expression of proinflammatory cytokines and chemokines in the colons of AOM/DSS–induced colorectal cancer mice, and the mRNA expression of COX-II, which is an important tumor marker for colorectal cancer, as well as the proliferation marker cyclin D1 (Fig. 5A). JMF3086 also reduced the intracolonic infiltration of inflammatory cells, such as macrophages and neutrophils (F4/80 and Ly-6G), and decreased COX-II expression around the sites of tumors (Fig. 5B and Supplementary Fig. S5). The efficacy of JMF3086 was superior to the combination of lovastatin and SAHA (Figs. 5B, 4A and B). In addition, JMF3086 did not induce in vivo apoptosis despite of protecting AOM/DSS–induced colorectal cancer (Fig. 5C and D).

Figure 5.

Preventive effect of JMF3086 on immune responses in AOM/DSS mouse models. A, the level of various cytokines and chemokines in colon culture supernatants was seen. Error bars, SE. **, P < 0.01 versus vehicle, ##, P < 0.01 versus normal mouse tissue lysates (control); two-sided Student t test. B, colon sections were immunostained with anti-F4/80, anti-Ly-6G, and anti-COX-II antibodies, and one representative experiment of three was presented (left). Scale bar, 250 μm. Right, quantification of IHC performed by TissueFAXS software. Error bars, SD. ##, P < 0.01 versus normal mouse paraffin sections (control); *, P < 0.05; and **, P < 0.01 versus vehicle; two-sided Student t test. C, colon tissues were taken from colorectal cancer mice for prevention experiments, and TUNEL assay was employed to detect apoptotic cells. Higher magnification views of cells are indicated by arrows, and one representative experiment of three is presented (original magnification: ×400, ×1,000). D, effect of JMF3086 on apoptotic proteins in colon tissues. Total cell lysates were prepared and the expressions of PARP, caspase-3, BAX, and β-actin were analyzed by Western blot analysis.

Figure 5.

Preventive effect of JMF3086 on immune responses in AOM/DSS mouse models. A, the level of various cytokines and chemokines in colon culture supernatants was seen. Error bars, SE. **, P < 0.01 versus vehicle, ##, P < 0.01 versus normal mouse tissue lysates (control); two-sided Student t test. B, colon sections were immunostained with anti-F4/80, anti-Ly-6G, and anti-COX-II antibodies, and one representative experiment of three was presented (left). Scale bar, 250 μm. Right, quantification of IHC performed by TissueFAXS software. Error bars, SD. ##, P < 0.01 versus normal mouse paraffin sections (control); *, P < 0.05; and **, P < 0.01 versus vehicle; two-sided Student t test. C, colon tissues were taken from colorectal cancer mice for prevention experiments, and TUNEL assay was employed to detect apoptotic cells. Higher magnification views of cells are indicated by arrows, and one representative experiment of three is presented (original magnification: ×400, ×1,000). D, effect of JMF3086 on apoptotic proteins in colon tissues. Total cell lysates were prepared and the expressions of PARP, caspase-3, BAX, and β-actin were analyzed by Western blot analysis.

Close modal

JMF3086 treatment did not exhibit any toxicity based on biochemical examinations and H&E staining of various organs (Supplementary Fig. S6A and S6B). However, combination of lovastatin and SAHA increased serum creatinine levels and reduced the concentration of albumin (Supplementary Fig. S6A), indicating renal toxicity.

JMF3086 prevents cancer stemness and endotoxin release in AOM/DSS–induced colorectal cancer mouse models

It has been suggested that chronic inflammation causes crypt damage and regeneration, resulting in stem cell expansion and leading to colorectal tumorigenesis (17, 26). CD166, EpCAM, CD44, and ALDH1 are putative cell-surface markers associated with colorectal CSCs (26, 27). The mRNA expression of these markers was found to be increased in AOM/DSS–induced colorectal cancer mice (Fig. 6A). JMF3086 prevented the induction of these markers, whereas statins and SAHA were ineffective or weaker (Fig. 6A).

Figure 6.

Preventive effect of JMF3086 on cancer stemness and endotoxin release in AOM/DSS mouse models. A, the mRNA expressions of colorectal CSC markers in colon tissues from 8-week-old AOM/DSS colorectal cancer mice of prevention model are shown. ##, P < 0.01 versus normal mouse tissue lysates (control); *, P < 0.05; and **, P < 0.01 versus vehicle. B, gross pictures of terminal colons are shown, and arrowhead indicates macroscopic lesions (left). Scale bars, 5 mm. Colon sections were counterstained with H&E. Scale bars, 250 μm. Endotoxin concentration (EU/mL) of serum from AOM/DSS–induced colorectal cancer mouse was measured by limulus amebocyte lysate (LAL) assay (right). Error bars, SE. *, P < 0.05; **, P < 0.01 versus vehicle; ##, P < 0.01 versus normal mouse serum (control); two-sided Student t test. C, connection between inflammation and carcinogenesis and prevention by statin hydroxamates. The oral administration of statin hydroxamate prevented AOM/DSS–induced colorectal cancer in mice by inhibiting inflammation. Statin hydroxamate also inhibited the release of endotoxin in vivo. Furthermore, the stemness of colorectal cancer was inhibited in vivo as well. These data provided compelling evidence that statin hydroxamate showed significant efficacy to prevent colorectal cancer in preclinical models.

Figure 6.

Preventive effect of JMF3086 on cancer stemness and endotoxin release in AOM/DSS mouse models. A, the mRNA expressions of colorectal CSC markers in colon tissues from 8-week-old AOM/DSS colorectal cancer mice of prevention model are shown. ##, P < 0.01 versus normal mouse tissue lysates (control); *, P < 0.05; and **, P < 0.01 versus vehicle. B, gross pictures of terminal colons are shown, and arrowhead indicates macroscopic lesions (left). Scale bars, 5 mm. Colon sections were counterstained with H&E. Scale bars, 250 μm. Endotoxin concentration (EU/mL) of serum from AOM/DSS–induced colorectal cancer mouse was measured by limulus amebocyte lysate (LAL) assay (right). Error bars, SE. *, P < 0.05; **, P < 0.01 versus vehicle; ##, P < 0.01 versus normal mouse serum (control); two-sided Student t test. C, connection between inflammation and carcinogenesis and prevention by statin hydroxamates. The oral administration of statin hydroxamate prevented AOM/DSS–induced colorectal cancer in mice by inhibiting inflammation. Statin hydroxamate also inhibited the release of endotoxin in vivo. Furthermore, the stemness of colorectal cancer was inhibited in vivo as well. These data provided compelling evidence that statin hydroxamate showed significant efficacy to prevent colorectal cancer in preclinical models.

Close modal

The microbiota and the immune system mutually interact to maintain homeostasis in the intestine. However, endotoxin released from the microbiota can alter this balance and promote chronic inflammation and lead to intestinal tumor development (28). To evaluate whether JMF3086 maintained gut homeostasis, endotoxin level in the serum of AOM/DSS–induced colorectal cancer mice was detected. High level of endotoxin was detected in AOM/DSS–treated group, and this effect was prevented by JMF3086 with the extent correlated with the reductions in tumor number (Fig. 6B).

Chemoprevention is a rational and appealing approach to reduce the risk of cancer by using pharmacologic means, such as aspirin for colorectal cancer and tamoxifen for breast cancer (29, 30). Aspirin has been reported to prevent carcinogenesis of colorectal cancer in ApcMin/+ mice and prevent 44% tumor formation at a dosage of 200 mg/kg through attenuating inflammation (31, 32). However, its clinical use for chemoprevention is prohibited due to a significant incidence of gastrointestinal upset and bleeding. Epigenetic modulators, such as HDAC2 overexpression, were associated with colorectal cancer progression, and a selective HDAC2 inhibitor was reported to prevent colorectal cancer tumorigenesis in both AOM/DSS mouse models and ApcMin/+ mice (33). A recent study shows that gut microbiota could ferment dietary fiber into butyrate, a class I and II HDAC inhibitor, to exert chemopreventive effect on colorectal cancer in gnotobiotic mouse models (34), suggesting that HDACs are potential targets for colorectal cancer prevention. In addition, epidemiology study suggests that statins attenuated the risk of colorectal cancer (35). In this study, our polypharmacological small molecule, statin hydroxamate (JMF3086), dually inhibits HDACs and HMGR exerting anti-inflammatory effect to prevent colitis and colitis-associated colorectal cancer. HMGR and HDAC activities in colon lysates of AOM/DSS–induced colorectal cancer were increased and inhibited by JMF, indicating that both inhibitions contributed to its chemoprevention. The advantage of JMF being superior to lovastatin plus SAHA is like a two-in-one antibody with superior inhibitory activity, compared with two monospecific antibodies (36). Additional mechanisms beyond the inhibitions on HDAC and HMGR activities or due to different pharmacokinetic property are also possible and awaited for further investigation. However, we reveal a new strategy for colorectal cancer chemoprevention.

IBD is an inflammatory disorder of the gastrointestinal tract (6). Epidemiologic studies suggest that the incidence and prevalence of IBD are increasing around the world. However, the exact etiology remains unknown and disease pathogenesis is not fully understood. DSS-induced colitis has been shown to be similar to human IBD in multiple aspects and has become an important tool to investigate its pathophysiologic mechanisms and immunologic processes (37). DSS-induced colitis is triggered by disrupting the colon epithelial barrier, allowing intestinal bacteria to penetrate the injured mucosa and induce mucosal inflammation, which is characterized by increased infiltration of inflammatory cells and an excessive production of proinflammatory cytokines/chemokines, leading to colitis exacerbation (38). Therefore, inhibition of inflammation is a potential strategy to prevent cancer initiation. In this study, we found JMF3086 could prevent and treat acute inflammation in the DSS-induced colitis mouse models by inhibiting the release of proinflammatory cytokines/chemokines and the infiltration of neutrophils into colitis regions. AOM in conjunction with the additional inflammatory stimulus of DSS results in tumor development that is restricted to the colon in mice (12). This model is widely utilized to recapitulate human colorectal cancer through inflammation and colon ulceration, similar to what is observed in patients (39). JMF3086 is demonstrated to prevent AOM/DSS colorectal cancer in this study, indicating that both initiation and progression of colorectal cancer are inhibited by JMF3086.

Tumors are tightly connected with microbiota and immune system (40). The link between microbiota and colorectal cancer in patients and mice has been studied and found that germ-free animals showed fewer tumors compared with those of normal microbiota. Antibiotic cocktail depleting the intestinal microbiota in mice reduced tumor growth in liver and colon, suggesting that microbiota promotes tumor progression (41). Specifically, Fusobacterium nucleatum compared with healthy people is more common in the gut of IBD patients to promote colorectal cancer (42), and clinical isolate of F. nucleatum promoted carcinogenesis in ApcMin/+ mice (43). The gut microbiota modulates carcinogenesis through three mechanisms (28). First, dysbiosis induces bacterial translocation, leading to induction of inflammation mediated by microorganism-associated molecular patterns and Toll-like receptors in several cell types (41). Intestinal inflammation promotes colorectal cancer development through release of proinflammatory cytokines/chemokines and the infiltration of macrophages and neutrophils into tumor regions (28, 44). Bacterial translocation was detected at tumor sites in ApcMin/+ mice, and antibiotic eradication reduced colorectal cancer development (45). Second, microbiota-derived metabolites could induce DNA damage, genotoxicity, and chromosome instability to induce barrier failure of gastrointestinal tract, which also trigger colorectal cancer carcinogenesis. For example, GNB-produced LPS and cytolethal distending toxin directly trigger double-strand DNA damage responses, leading to barrier disrupt. Third, microbiota could convert bile acid in host into secondary metabolites, such as deoxycholic acid, which in turn promotes the development of hepatocellular carcinoma and colorectal cancer (46, 47). In this study, increased mRNA expressions of proinflammatory cytokines and chemokines, infiltrations of macrophage (F4/80) and neutrophil (Ly-6G), as well as increased serum endotoxin (LPS) level were seen in AOM/DSS colorectal cancer mouse models. All these events were prevented by JMF3086, suggesting that JMF3086 might prevent colorectal cancer development through modulating gut microbiota–induced bacterial translocation and barrier failure of gastrointestinal tract. It is important to find a reliable biomarker to detect the effect of chemopreventive agents. Serum endotoxin level might serve as a potential biomarker to detect the prevention of JMF3086 in colorectal cancer.

The collection of bacteria, viruses, and fungi that live in the human body was known as the “microbiome” (48). It is estimated that less than 1% of all microorganisms in the natural world have been identified (49). It was largely unknown and regarded as a black box. Recently, the advent of new technologies, including high-throughput DNA sequencing and bioinformatics, has increased the capacity and launched a revolution in the microbiome (40). In this study, we found that JMF3086 prevented endotoxin release in AOM/DSS–induced colorectal cancer mouse models as well as the treatment model of DSS-induced colitis. It is possible that JMF3086 acts through modulating the population of gut microbiota. High-throughput microbial 16S rRNA gene sequencing combined with mouse models is awaited to discover functional genes of microbiota impacted by JMF3086.

Colorectal CSCs have been reported to initiate colorectal cancer and to play an important role in the genesis, maintenance, recurrence, and metastasis of cancer (50, 51). The expression of colorectal CSC markers, CD166, EpCAM, CD44, and ALDH1, in AOM/DSS–induced colorectal cancer mouse models was prevented by JMF3086, indicating that inhibition of CSCs might also contribute to its chemoprevention on colorectal cancer. Statins and HDAC inhibitors have been reported to reduce the stemness of colorectal CSCs (52–54).

In conclusion, our findings indicated that JMF3086, which is a potent dual inhibitor targeting HDACs and HMGR, has potential benefit and is a promising lead for the prevention of colitis-associated colorectal cancer.

No potential conflicts of interest were disclosed.

Conception and design: T.-T. Wei, C.-C. Chen

Development of methodology: T.-T. Wei, Y.-T. Lin, R.-Y. Tseng

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T.-T. Wei, Y.-T. Lin, R.-Y. Tseng

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T.-T. Wei, Y.-T. Lin, R.-Y. Tseng, C.-T. Shun

Writing, review, and/or revision of the manuscript: T.-T. Wei, Y.-C. Lin, M.-S. Wu, C.-C. Chen

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): T.-T. Wei, M.-S. Wu, J.-M. Fang, C.-C. Chen

Study supervision: C.-C. Chen

Other (obtained funding): C.-C. Chen

This work was supported by a research grant from the National Science Council of Taiwan and the Institute of Biomedical Sciences, Academia Sinica (IBMS-CRC103-P02).

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.
Sporn
MB
. 
Approaches to prevention of epithelial cancer during the preneoplastic period
.
Cancer Res
1976
;
36
:
2699
702
.
2.
Burn
J
,
Gerdes
AM
,
Macrae
F
,
Mecklin
JP
,
Moeslein
G
,
Olschwang
S
, et al
Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial
.
Lancet
2011
;
378
:
2081
7
.
3.
Fink
SP
,
Yamauchi
M
,
Nishihara
R
,
Jung
S
,
Kuchiba
A
,
Wu
K
, et al
Aspirin and the risk of colorectal cancer in relation to the expression of 15-hydroxyprostaglandin dehydrogenase (HPGD)
.
Sci Transl Med
2014
;
6
:
233re2
.
4.
Terzic
J
,
Grivennikov
S
,
Karin
E
,
Karin
M
. 
Inflammation and colon cancer
.
Gastroenterology
2010
;
138
:
2101
14
.
e5
.
5.
Greten
FR
,
Eckmann
L
,
Greten
TF
,
Park
JM
,
Li
ZW
,
Egan
LJ
, et al
IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer
.
Cell
2004
;
118
:
285
96
.
6.
Ullman
TA
,
Itzkowitz
SH
. 
Intestinal inflammation and cancer
.
Gastroenterology
2011
;
140
:
1807
16
.
7.
Moossavi
S
,
Bishehsari
F
. 
Inflammation in sporadic colorectal cancer
.
Arch Iran Med
2012
;
15
:
166
70
.
8.
Bardou
M
,
Barkun
AN
,
Martel
M
. 
Obesity and colorectal cancer
.
Gut
2013
;
62
:
933
47
.
9.
Tang
J
,
Sharif
O
,
Pai
C
,
Silverman
AL
. 
Mesalamine protects against colorectal cancer in inflammatory bowel disease
.
Dig Dis Sci
2010
;
55
:
1696
703
.
10.
van Staa
TP
,
Card
T
,
Logan
RF
,
Leufkens
HG
. 
5-Aminosalicylate use and colorectal cancer risk in inflammatory bowel disease: a large epidemiological study
.
Gut
2005
;
54
:
1573
8
.
11.
Arumugam
M
,
Raes
J
,
Pelletier
E
,
Le Paslier
D
,
Yamada
T
,
Mende
DR
, et al
Enterotypes of the human gut microbiome
.
Nature
2011
;
473
:
174
80
.
12.
Chen
GY
,
Nunez
G
. 
Inflammasomes in intestinal inflammation and cancer
.
Gastroenterology
2011
;
141
:
1986
99
.
13.
Round
JL
,
Mazmanian
SK
. 
The gut microbiota shapes intestinal immune responses during health and disease
.
Nat Rev Immunol
2009
;
9
:
313
23
.
14.
Sommer
F
,
Backhed
F
. 
The gut microbiota–masters of host development and physiology
.
Nat Rev Microbiol
2013
;
11
:
227
38
.
15.
Macarthur
M
,
Hold
GL
,
El-Omar
EM
. 
Inflammation and Cancer II. Role of chronic inflammation and cytokine gene polymorphisms in the pathogenesis of gastrointestinal malignancy
.
Am J Physiol Gastrointest Liver Physiol
2004
;
286
:
G515
20
.
16.
Hull
M
,
Lagergren
J
. 
Obesity and colorectal cancer
.
Gut
2014
;
63
:
205
.
17.
Carpentino
JE
,
Hynes
MJ
,
Appelman
HD
,
Zheng
T
,
Steindler
DA
,
Scott
EW
, et al
Aldehyde dehydrogenase-expressing colon stem cells contribute to tumorigenesis in the transition from colitis to cancer
.
Cancer Res
2009
;
69
:
8208
15
.
18.
Kapur
NK
. 
Rosuvastatin: a highly potent statin for the prevention and management of coronary artery disease
.
Expert Rev Cardiovasc Ther
2007
;
5
:
161
75
.
19.
Ward
MM
. 
The JUPITER study: statins for the primary prevention of cardiovascular events in patients with inflammatory rheumatic diseases?
F1000 Med Rep
2009
;
1
:
35
.
20.
Hawk
E
,
Viner
JL
. 
Statins and cancer–beyond the "one drug, one disease" model
.
N Engl J Med
2005
;
352
:
2238
9
.
21.
Lin
YC
,
Lin
JH
,
Chou
CW
,
Chang
YF
,
Yeh
SH
,
Chen
CC
. 
Statins increase p21 through inhibition of histone deacetylase activity and release of promoter-associated HDAC1/2
.
Cancer Res
2008
;
68
:
2375
83
.
22.
West
AC
,
Johnstone
RW
. 
New and emerging HDAC inhibitors for cancer treatment
.
J Clin Invest
2014
;
124
:
30
9
.
23.
Chen
JB
,
Chern
TR
,
Wei
TT
,
Chen
CC
,
Lin
JH
,
Fang
JM
. 
Design and synthesis of dual-action inhibitors targeting histone deacetylases and 3-hydroxy-3-methylglutaryl coenzyme A reductase for cancer treatment
.
J Med Chem
2013
;
56
:
3645
55
.
24.
Falkenberg
KJ
,
Johnstone
RW
. 
Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders
.
Nat Rev Drug Discov
2014
;
13
:
673
91
.
25.
Neurath
MF
. 
Cytokines in inflammatory bowel disease
.
Nat Rev Immunol
2014
;
14
:
329
42
.
26.
Huang
EH
,
Wicha
MS
. 
Colon cancer stem cells: implications for prevention and therapy
.
Trends Mol Med
2008
;
14
:
503
9
.
27.
LaBarge
MA
,
Bissell
MJ
. 
Is CD133 a marker of metastatic colon cancer stem cells?
J Clin Invest
2008
;
118
:
2021
4
.
28.
Gagliani
N
,
Hu
B
,
Huber
S
,
Elinav
E
,
Flavell
RA
. 
The fire within: microbes inflame tumors
.
Cell
2014
;
157
:
776
83
.
29.
Reddy
BS
,
Hirose
Y
,
Lubet
R
,
Steele
V
,
Kelloff
G
,
Paulson
S
, et al
Chemoprevention of colon cancer by specific cyclooxygenase-2 inhibitor, celecoxib, administered during different stages of carcinogenesis
.
Cancer Res
2000
;
60
:
293
7
.
30.
Fisher
B
,
Costantino
JP
,
Wickerham
DL
,
Cecchini
RS
,
Cronin
WM
,
Robidoux
A
, et al
Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study
.
J Natl Cancer Inst
2005
;
97
:
1652
62
.
31.
Bosetti
C
,
Rosato
V
,
Gallus
S
,
Cuzick
J
,
La Vecchia
C
. 
Aspirin and cancer risk: a quantitative review to 2011
.
Ann Oncol
2012
;
23
:
1403
15
.
32.
Mahmoud
NN
,
Dannenberg
AJ
,
Mestre
J
,
Bilinski
RT
,
Churchill
MR
,
Martucci
C
, et al
Aspirin prevents tumors in a murine model of familial adenomatous polyposis
.
Surgery
1998
;
124
:
225
31
.
33.
Ravillah
D
,
Mohammed
A
,
Qian
L
,
Brewer
M
,
Zhang
Y
,
Biddick
L
, et al
Chemopreventive effects of an HDAC2-selective inhibitor on rat colon carcinogenesis and APCmin/+ mouse intestinal tumorigenesis
.
J Pharmacol Exp Ther
2014
;
348
:
59
68
.
34.
Donohoe
DR
,
Holley
D
,
Collins
LB
,
Montgomery
SA
,
Whitmore
AC
,
Hillhouse
A
, et al
A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner
.
Cancer Discov
2014
;
4
:
1387
97
.
35.
Poynter
JN
,
Gruber
SB
,
Higgins
PD
,
Almog
R
,
Bonner
JD
,
Rennert
HS
, et al
Statins and the risk of colorectal cancer
.
N Engl J Med
2005
;
352
:
2184
92
.
36.
Schaefer
G
,
Haber
L
,
Crocker
LM
,
Shia
S
,
Shao
L
,
Dowbenko
D
, et al
A two-in-one antibody against HER3 and EGFR has superior inhibitory activity compared with monospecific antibodies
.
Cancer Cell
2011
;
20
:
472
86
.
37.
Mizoguchi
A
. 
Animal models of inflammatory bowel disease
.
Prog Mol Biol Transl Sci
2012
;
105
:
263
320
.
38.
Maloy
KJ
,
Powrie
F
. 
Intestinal homeostasis and its breakdown in inflammatory bowel disease
.
Nature
2011
;
474
:
298
306
.
39.
Alex
P
,
Zachos
NC
,
Nguyen
T
,
Gonzales
L
,
Chen
TE
,
Conklin
LS
, et al
Distinct cytokine patterns identified from multiplex profiles of murine DSS and TNBS-induced colitis
.
Inflamm Bowel Dis
2009
;
15
:
341
52
.
40.
Blaser
MJ
. 
The microbiome revolution
.
J Clin Invest
2014
;
124
:
4162
5
.
41.
Schwabe
RF
,
Jobin
C
. 
The microbiome and cancer
.
Nat Rev Cancer
2013
;
13
:
800
12
.
42.
Allen-Vercoe
E
,
Strauss
J
,
Chadee
K
. 
Fusobacterium nucleatum: an emerging gut pathogen?
Gut Microbes
2011
;
2
:
294
8
.
43.
Kostic
AD
,
Chun
E
,
Robertson
L
,
Glickman
JN
,
Gallini
CA
,
Michaud
M
, et al
Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment
.
Cell Host Microbe
2013
;
14
:
207
15
.
44.
Galdiero
MR
,
Bonavita
E
,
Barajon
I
,
Garlanda
C
,
Mantovani
A
,
Jaillon
S
. 
Tumor associated macrophages and neutrophils in cancer
.
Immunobiology
2013
;
218
:
1402
10
.
45.
Grivennikov
SI
,
Wang
K
,
Mucida
D
,
Stewart
CA
,
Schnabl
B
,
Jauch
D
, et al
Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth
.
Nature
2012
;
491
:
254
8
.
46.
Nesic
D
,
Hsu
Y
,
Stebbins
CE
. 
Assembly and function of a bacterial genotoxin
.
Nature
2004
;
429
:
429
33
.
47.
Bernstein
C
,
Holubec
H
,
Bhattacharyya
AK
,
Nguyen
H
,
Payne
CM
,
Zaitlin
B
, et al
Carcinogenicity of deoxycholate, a secondary bile acid
.
Arch Toxicol
2011
;
85
:
863
71
.
48.
Lederberg
J
. 
Infectious history
.
Science
2000
;
288
:
287
93
.
49.
Fuhrman
JA
. 
Microbial community structure and its functional implications
.
Nature
2009
;
459
:
193
9
.
50.
Eyler
CE
,
Rich
JN
. 
Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis
.
J Clin Oncol
2008
;
26
:
2839
45
.
51.
Pignalosa
D
,
Durante
M
. 
Overcoming resistance of cancer stem cells
.
Lancet Oncol
2012
;
13
:
e187
8
.
52.
Gauthaman
K
,
Fong
CY
,
Bongso
A
. 
Statins, stem cells, and cancer
.
J Cell Biochem
2009
;
106
:
975
83
.
53.
Kodach
LL
,
Jacobs
RJ
,
Voorneveld
PW
,
Wildenberg
ME
,
Verspaget
HW
,
van Wezel
T
, et al
Statins augment the chemosensitivity of colorectal cancer cells inducing epigenetic reprogramming and reducing colorectal cancer cell ‘stemness’ via the bone morphogenetic protein pathway
.
Gut
2011
;
60
:
1544
53
.
54.
Sikandar
S
,
Dizon
D
,
Shen
X
,
Li
Z
,
Besterman
J
,
Lipkin
SM
. 
The class I HDAC inhibitor MGCD0103 induces cell cycle arrest and apoptosis in colon cancer initiating cells by upregulating Dickkopf-1 and non-canonical Wnt signaling
.
Oncotarget
2010
;
1
:
596
605
.