Statins are potential anticancerous agents (Review)

  • Authors:
    • Abdullah K. Altwairgi
  • View Affiliations

  • Published online on: January 20, 2015     https://doi.org/10.3892/or.2015.3741
  • Pages: 1019-1039
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Statins are inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), which is a rate-limiting enzyme in the mevalonate pathway. The pleiotropic effects of statins may be mediated by the inhibition of downstream products such as small GTP-binding proteins, Rho, Ras and Rac whose localization and function are dependent on isoprenylation. Preclinical studies of statins in different cancer cell lines and animal models showed antiproliferative, pro‑apoptotic and anti-invasive effects. Notably, statins showed targeted action in cancerous cell lines compared to normal cells. Previous studies have also shown the synergistic effects of statins with chemotherapeutic agents and radiotherapy. This effect of statins was also observed in chemotherapeutic-resistant tumors. Statins were reported to sensitize the cells to radiation by arresting them in the late G1 phase of the cell cycle. Similarly, population-based studies also demonstrated a chemopreventive and survival benefit of statins in various types of cancers. However, this benefit has yet to be proven in clinical trials. The inter-individual variation in response to statins may be contributed to many genetic and non-genetic factors, including single-nucleotide polymorphisms in HMGCR gene and the overexpression of heterogeneous nuclear ribonucleoprotein A1, which was reported to reduce HMGCR enzyme activity. However, more studies with large phase III randomized controlled trials in cancer patients should be conducted to establish the effect of statins in cancer prevention and treatment.

1. Introduction

The global burden due to cancer increased to 14.1 million new cases and 8.2 million cancer-associated mortalities in 2012 (1). The outcome of cancer patients remains poor, despite recent advances in the understanding of the molecular mechanism of tumorigenesis. Thus, more effective initial treatments for this intractable disease are required. Recent therapies under investigation include immunotherapy, chemotherapy, targeted molecular, antiangiogenic and gene therapy, radiation enhancement and drugs for overcoming resistance (2). Statins are inhibitors of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), commonly used as cholesterol-lowering agents (3), that have proven their effectiveness in the treatment of cardiovascular diseases. Preclinical evidence has indicated their antiproliferative, pro-apoptotic, anti-invasive and radio-sensitizing properties (4), and there are emerging interests in the use of statins as anticancer agents. In the present study, we reviewed the current data of statins in cancer.

2. Mechanism of action of statins

Inhibition of HMGCR by statins is a rate-limiting step in the mevalonate pathway. The products of the mevalonate pathway include isoprene units incorporated into sterol and non-sterol compounds. This inhibition by a statin may result in decreased levels of mevalonate and its downstream products, which affects critical cell functions such as membrane integrity, cell signaling, protein synthesis and cell cycle progression. The effect of statins on these processes and consequently on tumor cells, may therefore be able to control tumor initiation, growth and metastasis (Fig. 1) (517).

3. Antitumor effects of statins in preclinical studies

The in vitro preclinical studies in different cell lines have shown the ability of statins to suppress tumor growth and development. Statins exert antiproliferative, pro-apoptotic and anti-invasive effects in different cancer cell lines with varying sensitivity. The antimyeloma activity of statins in humans was first reported with the concomitant simvastatin administration in refractory multiple myeloma (MM), which showed reduced drug resistance (18). However, high-dose simvastatin treatment (15 mg/kg/day) in heavily pretreated MM patients transiently increased osteoclast activity and gastrointestinal side-effects, leading to premature discontinuation (19). A pre-operative study in primary invasive breast cancer patients investigated atorvastatin-induced effects on tumor proliferation and HMGCR expression while analyzing HMGCR as a predictive marker for statin response. Results of that study suggest HMGCR is targeted by statins in breast cancer cells in vivo, and that statins may have an antiproliferative effect in HMGCR-positive tumors (20). Furthermore, fluvastatin reduced tumor proliferation and increased apoptotic activity in high-grade, stage 0/1 breast cancer in invasive breast cancer patients (21). Statin-induced effects and the underlined mechanism in different cancer cell lines are presented in Table I.

Table I

Postulated molecular mechanisms of statin-induced anticancer activity in different cancer cell lines and animal models.

Table I

Postulated molecular mechanisms of statin-induced anticancer activity in different cancer cell lines and animal models.

Authors, year (Refs.)StatinCancer cell line/animal model usedOutcomeMolecular mechanism
Sławińska-Brych et al, 2014 (94)FluvastatinC6 rat malignant glioma cellsInhibitory and cytotoxic effect; no inhibition on the growth of normal neuronal cellsDecreased p-ERK1/2 expression, upregulation of p-JNK1/2, and reduction in the MMP-9 and VEGF concentrations
Yongjun et al, 2013 (95)AtorvastatinMicrogliaAtorvastatin reduced the microglial expression of MT1-MMPDownregulation of MT1-MMP is controlled by a p38 MAPK pathway in microglia
Crosbie et al, 2013 (96)StatinsHuman leukemic NK cell lineStatins inhibit YT-INDY proliferation, disrupt cell cycle progression and abrogate NK cell cytotoxicityBlockage of products in the latter part of the mevalonate pathway
Al-Haidari et al, 2014 (97)SimvastatinCCL17-induced colon cancer cell (HT-29)Dose-dependent decrease in cell migrationInhibition of geranylgeranylation and RhoA activation
Ishikawa et al, 2014 (98)StatinsColorectal cancer cell linesSimvastatin, fluvastatin and atorvastatin (except pravastatin) showed antiproliferative effectsStatins induced p27KIP1 expression by downregulation of histone methyltransferase enhancer of EZH2
Chang et al, 2013 (99)SimvastatinHCT116 colorectal cancer cellApoptosis with a decrease in cell viabilityActivation of p38MAPK-p53-survivin cascade; associated with the modulation of p21(cip/Waf1) and survivin
Rentala et al, 2013 (100)Atorvastatin (16–64 μM)CD133(+) CD44(+) cells derived from prostate cancer biopsies and peripheral bloodDose-dependent inhibition of cell adhesion and cell differentiationDecreased expression of integrins α1 and β1 and phosphorylated MYPT1 and FAK, reduced ROCK1 and FAK-mediated cell differentiation
Peng et al, 2013 (101)AtorvastatinLNCaP and PC3 human prostate cancer cellsDose-dependent inhibition of proliferation; greater induction of autophagy in PC3 cellsDownregulation of Bcl2, upregulation of p21, and increased expression of LC3-II; upregulation of miR-182, which may be a stress-responsive mRNA
Al-Husein et al, 2013 (102)SimvastatinHuman PC3 prostate cancer cellsInhibition of transendothelial migration in vitro; direct activation of endothelial cells and enhancement of endothelial-barrier resistanceInhibition of integrin αvβ3 activity and suppression of interaction between integrin αvβ3 and endothelial ICAM 1; modulation of angiopoietins and VEGF-A, thus preventing endothelial-barrier disruption
Fang et al, 2013 (103)SimvastatinRenal cell carcinoma cells, A498 and 786-O cellsDose-dependent suppression of proliferation and motility; induction of apoptosisInhibition of AKT/mTOR, ERK, and JAK2/STAT3 pathways
Islam et al, 2013 (104)AtorvastatinIn vitro and in vivo studies of HNSCCReduced cell motility, invasion, proliferation, and colony formation; inhibition of angiogenesis and lung metastases in vivoReduction in the active form of RhoC in vitro; a significant decrease in p-ERK1/2 and p-STAT3
Yu et al, 2013 (105)SimvastatinA549 Human lung cancer cell lineBlocks cells in G1 phase of the cell cycle and induces apoptosisDecreased Bcl-2, increased Bax protein, downregulation of cyclin D1 and CDK expression, increased caspase-3, -8 and -9 mRNA and protein expression, downregulated XIAP levels, and decreased MMP-9 levels, possibly by inhibiting NF-κB
Pelaia et al, 2012 (106)SimvastatinHuman non-small lung cancer cellsInduction of apoptosis after 36 h; increase in caspase-3 activation and TUNEL-positive cells, associated with a reduction in cell numbersIn susceptible lung cancer cell phenotypes, pro-apoptotic and anti-proliferative activity appears to be mediated by inhibition of the Ras/Raf/MEK/ERK signaling cascade
Chen et al, 2012 (107)AtorvastatinHuman non-small cell lung carcinomas (NSCLCs)Inhibits angiogenesisInhibition of VEGF expression in vitro and in vivo via inhibition of ROS production; ROS inhibited partly through suppression of Rac1/NADPH oxidase activity
Guterres et al, 2013 (108)SimvastatinHuman metastatic melanoma cells (WM9)Induction of a senescent phenotype, characterized by G1 arrestActivation of p53/p21 pathway, and increased intracellular ROS accompanied by elevated expression of catalase and peroxiredoxin-1
Pich et al, 2013 (109)StatinsHuman metastatic melanoma cellsMelanoma cells more sensitive to in vitro lysis by NK cellsIncrease of MICA involving peroxisome proliferator-activated receptor γ, which is independent of Ras and Rho signaling pathways
Gopalan et al, 2013 (110)SimvastatinHuman breast cancer cellsInduces apoptosisActivation of JNK/CHOP/DR5 pro-apoptotic pathway
Park et al, 2013 (111)SimvastatinTNBC cellsDecreased cell viabilities; PTEN mutant-type TNBC cells showed a decreased response vs. PTEN wild-type TNBC cellsInhibition of TNBC cells via PI3K pathway activation
Zhao et al, 2012 (112)SimvastatinHER2-positive breast cancer cellsTime- and dose-dependent cell death in HER2-overexpressing cell linesInhibition of HER2 promoter and increased expression of PEA3 (HER2 promoter inhibitor)
Qi et al, 2013 (113)Atorvastatin, fluvastatin and simvastatinLymphoma cells including A20 and EL4 cellsStatins induced apoptosisActivation caspase-3, PARP and Bax; suppressed activation of anti-apoptotic molecule Bcl-2; increased intracellular ROS generation and p38 activation and suppressed activation of AKT and ERK pathways
Zeng et al, 2012 (114)SimvastatinSHI-1 Human acute monocytic leukemia cell lineTime- and dose-dependent inhibition of proliferation and induction of apoptosisChange in gene expression level in PI3K-AKT signaling pathway
Yang et al, 2012 (115)AtorvastatinHuman monocytic leukemia cells (THP-1)Possible interaction with innate immunitySignificant decrease in the levels of TLR4 protein and mRNA, NF-κB expression and levels of TNF-α, IL-6 and IL-1β in LPS-induced THP-1 cells
Shi et al, 2013 (116)StatinsHuman esophageal squamous cell carcinoma (ESCC)Inhibition of cell growth and proliferationDecreased extracellular signal-regulated kinase activation and proliferating cell nuclear antigen, cyclin D1 expression, and increased cleavage of poly(ADP-ribose) polymerase
Ma et al, 2012 (117)LovastatinSquamous cell carcinoma (SCC) cell lines SCC9 and SCC25Synergistic cytotoxicity in combination with the EGFR inhibitor gefitinibTargets multiple metabolic stress pathways including the LKB1/AMPK pathway
Tu et al, 2011 (118)SimvastatinMultiple myeloma cellsS-phase cell cycle arrest and apoptosisActivation of Chk1, downregulation of Cdc25A, cyclin A and CDK2 expression; diminished Bcl-2 protein expression, increased cytosolic cytochrome c level, and activation of caspase 9 and 3
Araki et al, 2012 (119)Hydrophobic statinsRhabdomyosarcoma cells (A204 cells)AutophagyDepleting cellular levels of GGPP through an unknown pathway; induction of autophagy parallels statin toxicity (both can be suppressed by GGPP)
Animal model studies
Jani et al, 1993 (32)LovastatinHighly metastatic B16F10 mouse melanoma in nude miceIn vitro inhibition of attachment, motility and invasion that comprise the metastatic cascadeAntimetastatic effect on B16F10 cells is probably not mediated by a growth inhibitory action
Narisawa et al, 1996 (23)PravastatinColon tumor; 1,2-dimethylhydrazine.2HCl (DMH)-induced colon tumorigenesis was evaluated in ICR miceIncidence of colon tumors at week 35 was significantly lower in the pravastatin-treated groups vs. the control group. However, pravastatin increased tumor multiplicity/tumor-bearing animalSmall dose of pravastatin may be related, at least in part, to modulation of cholesterol synthesis in situ at the colonic mucosa
Narisawa et al, 1996 (24)PravastatinColon tumor; N-methyl-N-nitrosourea-induced colon carcinogenesis in F344 ratsIncidence of colon carcinomas at week 40 was significantly lower in the 25 and 5 ppm groups vs. the control group. However, it was similar in the 200 ppm groupsInhibition of the stage carcinogenesis (promotion), possibly through suppression of farnesyl isoprenylation of growth-regulating proteins such as p21 ras in the colonic mucosa
Broitman et al, 1996 (31)D-limonene (non-toxic monoterpene)CT-26, a murine transplantable colon tumor (hepatic ‘metastasis’ model)Dietary mevalonate and d-limonene inhibited the growth of tumors both alone and in combination by ~80% vs. controls at day 35D-limonene and mevalonate differentially affects the same pathway, and their individual actions may appear antagonistic in vitro
Hawk et al, 1996 (27)LovastatinIn vitro mouse and human non-transformed and neoplastically transformed lung epithelial cells; NNK-induced lung adenomas in miceInhibition of cell growth; no effect on tumor incidence or size, yet significant dose-dependent reduction in tumor multiplicity notedTumor suppression by lovastatin does not appear to be associated with the presence of mutated K-ras or to changes in K-ras expression
Liu et al, 2013 (120)SimvastatinHuman lung cancer cell line A549 and Balb/c nude mouse were usedSignificant inhibition of tumor cell invasion, growth and bone metastasis in lung cancer xenograft mouse modelSimvastatin may be associated with regulation of CD44, P53, MMP family and inactivation of MAPK/ERK signaling pathway
Inano et al, 1997 (22)SimvastatinMammary tumor (Wistar-MS rats)Increased development of ER PgR tumors and a reduced incidence of ER+ PgR+ tumorsSimvastatin has a potent preventive activity during the DES-dependent promotion/progression phase of radiation-induced mammary tumorigenesis
Alonso et al, 1998 (30)LovastatinF3II sarcomatoid mammary carcinoma tumor modelIn vitro inhibition of tumor cell attachment and migration; these actions were prevented by addition of mevalonate, but not by farnesyl pyrophosphateThese effects were not associated with the modification in Ras oncoproteins
Kikuchi et al, 1997 (26)SimvastatinHuman glioma cell lines in nude miceInhibition of cell growth in all cell lines tested. Simvastatin showed additive effects on pLDL-induced cytotoxicity. Inhibition of cell growth after intra- tumoral injection of simvastatin and pLDLMevalonic acid or a metabolite in the cholesterol synthesis pathway is necessary for glioma cell growth
Clutterbuck et al, 1998 (25)SimvastatinSCID mice injected with human HL60 myeloblastic leukaemia cell line. HL60 bears an N-Ras mutationNumber of clonogenic HL60 cells was reduced in the bone marrow of mice that received simvastatin vs. control mice by 65 and 68%, respectivelyInhibition of AML cell proliferation by simvastatin may be independent of the Ras signaling pathway
Matar et al, 1998 (29)LovastatinRat fibrosarcomaShort treatment with lovastatin diminished primary tumor growth and the number, and size of lung experimental metastasisp21ras protein may have a significant role in metastatic behavior of tumor cells
Matar et al, 1999 (28)LovastatinL-TACB rat lymphomaNo effect on cell motility, metalloprotease secretion, and neovascularization but significant inhibition of lymphoma cell invasivenessInhibition of invasiveness, possibly a consequence of impaired cell adhesion
Feleszko et al, 1999 (34)LovastatinRas-3T3 and HBL100-ras cells; Ha-ras-transformed murine tumor modelInhibition of cells transformed with Ha-ras oncogene more effectively than control NIH-3T3 and HBL100 -neo cells in in vitro studies. In in vivo experiments, the Ras-3T3 tumor demonstrated significantly increased sensitivity to combined treatment with lovastatin and TNF-α vs. either agent aloneLovastatin increases antitumor activity of TNF-α against tumor cells transformed with v-Ha-ras oncogene via inhibition of tumor-induced blood vessel formation (reduced VEGF production)
Feleszko et al, 2000 (33)LovastatinMurine tumor cell lines (colon-26 cells, v-Ha-ras-transformed NIH-3T3 sarcoma cells, and Lewis lung carcinoma cells)Significantly increased sensitivity to the combined treatment with lovastatin and doxorubicin vs. either agent aloneLovastatin induced reduction of troponin T release by cardiomyocytes. Lovastatin is known to reduce doxorubicin-induced cardiac injury
Liao et al, 2013 (121)AtorvastatinLSL-KrasG12D-LSL- Trp53R172H-Pdx1-Cre mouse model (known as Pankras/p53 mice)Significant increase in survival in mice fed 100 ppm atorvastatin vs. control mice with significant reduction in tumor volume and Ki-67-labeled cell proliferationAtorvastatin inhibited prenylation in several key proteins, including Kras protein. Waf1p21, cyp51A1 and soluble epoxide hydrolase were crucial atorvastatin-targeted genes involved in inflammation and carcinogenesis
Vitols et al, 1997 (122)In vivo study of malignant B lymphocytes from humansSimvastatin orally, 40 mg daily for 12 weeksPreviously untreated B-cell chronic lymphocytic leukemiaCells from four patients showed moderate to minor increases in the degradation rate of 1251-LDL, three patients showed an increase in HMGCR activity, and one patient showed both. No significant change in the clinical disease status was observed. However, during the subsequent year, 4/10 patients developed a therapy- demanding progressive disease
Higgins et al, 2012 (123)Short-term biomarker modulation studySimvastatin 40 mg orally daily for 24–28 weeksThe contralateral breast of women with a previous history of breast cancer was used as a high-risk model (n=50)Total cholesterol, LDL cholesterol, triglyceride and hsCRP decreased significantly during the study (P-values: <0.001, <0.001, 0.003 and 0.05, respectively). Estrone sulfate concentrations decreased with simvastatin treatment (P=0.01 overall), particularly among post-menopausal participants (P=0.006)
Bjarnadottir et al, 2013 (20)Window-of- opportunity trialHigh-dose atorvastatin (i.e., 80 mg/day) was prescribed to patients for two weeks before surgery. Pre- and post-statin-paired tumor samples were analyzed for Ki67 and HMGCR immunohistochemical expressionInvasive breast cancer (n=50)Upregulation of HMGCR was observed in 68% of the paired samples (P=0.0005). The average relative decrease in Ki67 expression was 7.6% (P=0.39) in all paired samples. The corresponding decrease in Ki67 expression in tumors expressing HMGCR in the pre-treatment sample was 24% (P=0.02). Furthermore, post-treatment Ki67 expression was inversely correlated to post-treatment HMGCR expression (rs, −0.42; P=0.03)

[i] ADP, adenosine diphosphate; AML, acute myeloid leukemia; AMPK, adenine monophosphate-activated protein kinase; CCL17, chemokine (C-C motif) ligand 17; CDK, cyclin-dependent kinase; CHOP, CCAAT/enhancer-binding protein homologous protein; DES, diethylstilbestrol; DR5, death receptor-5; ERK, extracellular signal-regulated kinase; ERK1/2, extracellular signal-regulated kinase 1 and 2; EZH2, enhancer of zeste homolog 2; FAK, focal adhesion kinase; GGPP, geranylgeranyl pyrophosphate; GI, gastrointestinal; HER2, human epidermal growth factor receptor 2; HMGCR, 3-hydroxy-3-methyl-glutaryl-CoA reductase; HNSCC, head and neck squamous cell carcinomas; hsCRP, high-sensitivity C-reactive protein; ICAM-1, intercellular adhesion molecule-1; IL, interleukin; JAK, janus kinase; STAT3, signal transducer and activator of transcription 3; JNK 1/2, c-Jun N-terminal kinase 1 and 2; LC3-II, microtubule-associated protein 1A/1B-light chain 3-phosphatidylethanolamine conjugate; LDL, low-density lipid; LKB1, liver kinase B1; LPS, lipopolysaccharide; NNK, 4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone; MAPK, mitogen-activated protein kinase; MICA, major histocompatibility complex (MHC) class I chain-related protein A; MMP-9, matrix metalloproteinase-9; MT1-MMP, membrane type 1 metalloproteinase; mTOR, mammalian target of rapamycin; MYPT1, myosin phosphatase target subunit 1; NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor κB; NK, natural killer; PARP, poly(ADP-ribose) polymerase; pJNK, phosphorylated c-Jun N-terminal kinase; p-LDL, peroxidized low-density lipoprotein; p21WAF1/CIP1 and p27KIP1, two cyclin-dependent kinase inhibitors; p38MAPK, p38 mitogen-activated protein kinase; ppm, parts per million; PTEN, phosphatase and tensin homolog; ROCK1, Rho-associated kinase 1; ROS, reactive oxygen species; SCID, severe combined immunodeficient; TLR4, toll-like receptor 4; TNBC, triple negative breast cancer; TNF, tumor necrosis factor; TUNEL, terminal dUTP nick end-labeling; VEGF, vascular endothelial growth factor; VEGF-A, vascular endothelial growth factor A; XIAP, X-linked inhibitor of apoptosis protein.

Besides their in vitro efficacy, statins have also been shown to have in vivo antitumor effects in various animal models of cancer. Their efficacy as chemopreventive agents has been demonstrated in radiation-induced mammary tumorigenesis (22), chemical-induced colon tumorigenesis in rodent models (23,24), human myeloid leukemia and glioma cancer cells inoculated in severe combined immunodeficient mice (25,26), and chemical-induced lung tumor in mice (27). Statins have also been shown to reduce metastasis in rat lymphoma (28), rat fibrosarcoma (29), mouse mammary tumor (30), murine colon tumor (31) and mouse melanoma (32). Furthermore, statins increased the in vivo antitumor effect of doxorubicin in three tumor models accompanied by attenuation of its cardiotoxicity (33). Similarly, statins increased the antitumor effect of tumor necrosis factor by inhibiting the tumor-induced angiogenesis in a murine tumor model (34). The effect of statin-induced anticancer activity in different animal models is presented in Table I.

4. Antitumor effects of statins in clinical studies

Observational and retrospective studies reporting cancer risk with statin usage

Accumulating evidence has focused on pre-diagnostic use of statins in reducing risk of lethal prostate cancer (35). In a prospective cohort study of 34,989 USA male health professionals, the use of statins was associated with a reduced risk of advanced prostate cancer. The risk of advanced disease was lower with longer statin use (P trend=0.003) vs. never use. The relative risk (RR) was 0.60 [95% confidence interval (CI), 0.35–1.03] for <5 years of use and 0.26 (95% CI, 0.08–0.83) for ≥5 years of use. There was no association between statin use and risk of total prostate cancer (RR, 0.96; 95% CI, 0.85–1.09) (36). In a Denmark-based case-control study (n=42,480), statin use was associated with an overall risk reduction (6%) that was specifically higher among patients with advanced prostate cancer (10%) (37). Similarly, a decreased risk in mortality was noted among 11,772 newly diagnosed non-metastatic prostate cancer patients in the UK. Furthermore, decreased risks of prostate cancer mortality and all-cause mortality were reported in patients who used statins prior to diagnosis [hazard ratio (HR), 0.55, 95% CI, 0.41–0.74; and HR, 0.66, 95% CI, 0.53–0.81, respectively]. The results were higher compared to those obtained from patients who initiated the treatment only after diagnosis (HR, 0.82, 95% CI, 0.71–0.96; and HR, 0.91, 95% CI, 0.82–1.01, respectively) (38). In another prospective, population-based cohort study (n=1001), statin use prior to prostate cancer diagnosis was unrelated to prostate cancer recurrence/progression, but was associated with a decrease in the risk of prostate cancer-specific mortality (39).

Data from observational studies have addressed the risk of glioma among statin users (40,41). The use of simvastatin and lovastatin for >6 months was inversely associated with glioma risk (40). A recent large nationwide case-control study (41) conducted in Denmark in patients with glioma (2,656 cases and 18,480 controls) also showed a reduction in the risk of glioma among long-term statin users compared with non-users, and the risk was inversely related to the intensity of statin treatment among users [odds ratio (OR), 0.71, 95% CI, 0.44–1.15 for highest intensity statin users]. This potential chemopreventive effect was limited to users of lipophilic statins (41).

Population-based studies have shown 19% reductions in esophageal cancer incidence where statins have been used. Observational studies have shown that statins reduced the incidence of adenocarcinoma in patients with Barrett’s esophagus (BE) by 43%; this effect was further enhanced by a 74% decrease in risk reduction in patients taking a combination of nonsteroidal anti-inflammatory drugs and statins (42).

Findings of a meta-analysis showed that statins are associated with a reduced risk of esophageal cancer, particularly in patients with BE. In a subset of patients with BE (5 studies, 312 esophageal adenocarcinomas in 2,125 patients), statins were associated with a significantly decreased risk (41%) of esophageal adenocarcinomas after adjusting for potential confounders (adjusted OR, 0.59, 95% CI, 0.45–0.78) (43). These findings were consistent with those of another meta-analysis of 11 observational studies. The pooled adjusted data showed statin use was associated with a lower incidence of the combined esophageal cancers (OR, 0.81, 95% CI, 0.75–0.88). Furthermore, their chemopreventive effect was increased in combination with cyclo-oxygenase inhibitors in reducing the risk of adenocarcinoma in BE (OR, 0.26, 95% CI, 0.1–0.68) (44).

A meta-analysis of 26 randomized controlled trials (RCTs) involving 290 gastric cancer and 8 observational studies, totaling 7,321 gastric cancers indicated a reduced risk of gastric cancer with statin use (45). In addition, a meta-analysis of published studies showed a modest reduction in colorectal cancer risk among statin users (46).

A review of 723 patients diagnosed with primary inflammatory breast cancer in 1995–2011 showed that hydrophilic statins were associated with significantly improved progression-free survival (PFS) rates (47). However, long-term use of statins was associated with increased risk of invasive ductal carcinoma (IDC; n=916) and invasive lobular carcinoma (ILC; n=1,068) in a contemporary population-based case-control study conducted in the Seattle-Puget Sound region. It was also reported that women diagnosed with hypercholesterolemia currently using statins for ≥10 years had more than double the risk of IDC (OR, 2.04, 95% CI, 1.17–3.57) and ILC (OR, 2.43, 95% CI, 1.40–4.21) compared with never users (48).

In the meta-analysis of all the observational studies published up to January 2012, statin use and long-term statin use did not significantly affect breast cancer risk. However, the cumulative meta-analysis showed a change in trend of reporting risk of breast cancer from positive to negative in statin users between 1993 and 2011. These findingd do not support the hypothesis that statins exert a protective effect against breast cancer (49).

The Cancer in The Ovary and Uterus Study (CITOUS; case-control study) assessed the use of statins prior to and following diagnosis in a subset of 424 cases of ovarian and endometrial cancers and 341 controls using pharmacy records. Use of statins >1 year prior to diagnosis was associated with risk reduction, whereas survival improvement was observed among the two malignancies when statins were ingested only after diagnosis (50).

The Nurses’ Health and Health Professionals Follow-Up Study investigated the association between statin use and renal cell carcinoma (RCC) risk. The reported results were similar between ever vs. never users of statins. The subgroup analyses of that study reported that statin use may be associated with a lower risk of RCC among women with no history of hypertension (51).

Statin use is associated with a reduced risk of hepatocellular cancer, most strongly in Asian, but also in Western populations (52). Similarly, another meta-analysis suggested a favorable effect of statins on hepatocellular carcinoma in the absence of a duration-risk relationship (53).

A meta-analysis of all the published articles up to December 2007 showed no association between statin use on pancreatic cancer risk among patients using statins daily for managing hypercholesterolemia (54). These findings were consistent with those from another meta-analysis, which reported no association between statin use and pancreatic cancer risk among patients using statins daily for preventing cardiovascular event (55).

A meta-analysis of observational trials and RCTs did not support a protective effect of statins on overall lung cancer risk, and the lung cancer risk among elderly people (56). Nineteen studies (5 RCTs and 14 observational studies) involving 38,013 lung cancer cases suggested no association between statin use and risk of lung cancer (57). Similarly, a meta-analysis of published literature did not support the role of statins in prevention of skin cancer (58).

A retrospective evaluation of 1,502 patients with urothelial carcinoma of the bladder treated with radical cystectomy and pelvic lymphadenectomy without neoadjuvant therapy showed statin users were at a higher risk for disease recurrence and cancer-specific mortality in a univariate, but not a multivariate analysis. However, the present study also reported that statin users were older (P=0.003), had higher body mass index (median 32 vs. 28 kg/m2, P<0.001), and were more likely to have positive soft tissue surgical margins (9 vs. 4%, P<0.001) (59). Another meta-analysis with limited RCTs suggested no association between statin use and risk of bladder cancer (60).

Numerous population-based case-control studies conducted in Taiwan did not provide evidence to support an association between statin use and risk of breast cancer (n=565; control, n=2,260) (61), esophageal cancer (n=197; controls, n=788) (62), bladder cancer (n=325; controls, n=1,300) (63), kidney cancer (n=177; controls, n=708) (64), and female lung cancer (n=297; controls, n=1,188) (65).

Another retrospective evaluation of the entire Danish population diagnosed with cancer between 1995 and 2007 was performed on 18,721 patients using statins regularly before the cancer diagnosis vs. 277,204 patients who had never used statins. The present study concluded that statin use in cancer patients was associated with reduced cancer-associated mortality as compared to that in non-users (HR, 0.85, 95% CI, 0.82–0.87) for each of the 13 types of cancer (66). In another meta-analysis (27 randomized trials), a median of 5 years of statin therapy was reported to have no effect on the incidence of, or mortality from, any type of cancer, or the aggregate of all cancers (67). The effect of statins on the incidence of different types of cancer reported in various observational and retrospective studies is presented in Table II.

Table II

Effect of the use of statins on different types of cancer reported in various observational and retrospective studies.

Table II

Effect of the use of statins on different types of cancer reported in various observational and retrospective studies.

Authors, year (Refs.)Country and database usedPatient populationTumorOutcome
Platz et al, 2006 (36)Prospective cohort study of USA male health professionalsCancer-free participants (n=34,989) in 1990 followed to 2002. A total of 2,579 prostate cancer cases determined during 376,939 person-year, follow-up, of which 316 were advanced (regionally invasive, metastatic or fatal)Prostrate cancerAge-standardized incidence rates of advanced prostate cancer, 38 vs. 89 in current statin users vs. past or never users/100,000 person-years. Multivariable-adjusted relative risk for current statin use vs. no current use, advanced disease, 0.51 (95% CI, 0.30–0.86); metastatic or fatal disease, 0.39 (95% CI, 0.19–0.77)
Bansal et al, 2012 (124)Literature search for observational studies through February 2012Fifteen cohort and 12 case-control studies; included 56,847 prostate cancer cases (n=1,893,571) followed-up for 2–17 yearsProstate cancerSignificant reduction in the risk of total prostate cancer by 7% (RR, 0.93, 95% CI, 0.87–0.99, P=0.03) and clinically important advanced prostate cancer by 20% (RR, 0.80, 95% CI, 0.70–0.90, P<0.001)
Geybels et al, 2013 (39)Patients (n=1,001) diagnosed with prostate cancer in 2002–2005 were followed-up by survey and the SEER registry (King Country, WA, USA)Statin use was assessed by detailed in-person interview; 289 were ever users of statin drugs (n=1,001)Prostate cancerProstate cancer recurrence/progression events, n=151; total deaths, n=123; prostate cancer-specific deaths, n=39. At 10 years, significantly lower risk of prostate cancer-specific mortality was reported for statin users (1%) vs. non-users (5%), P<0.01 (HR, 0.19, 95% CI, 0.06–0.56)
Park et al, 2013 (125)Systematic search of literature through August 2012; 17 eligible studies from 794 referencesThis 13-study meta-analysis evaluated associations between statins and RFS following treatment of localized prostate cancer, among patients (n=21,185) treated primarily with RT vs. radical prostatectomyLocalized prostate cancerOverall, statins did not affect RFS (HR, 0.90, 95% CI, 0.74–1.08). In RT patients (6 studies), statins were associated with a statistically significant improvement in RFS (HR, 0.68, 95% CI, 0.49–0.93) which was not observed in radical prostatectomy patients (7 studies)
Yu et al, 2014 (38)Population-based electronic database, UKNewly diagnosed patients (n=11,772; between 1998 and 2009) followed until 2012Non- metastatic prostate cancerProstate cancer deaths (n=1,791); post-diagnostic statin use was associated with a decreased risk of prostate cancer mortality (HR, 0.76, 95% CI, 0.66–0.88) and all-cause mortality (HR, 0.86, 95% CI, 0.78–0.95)
Jespersen et al, 2014 (37)Denmark-based case-control study, 1997–2010 from a national cancer registryPatients (n=42,480) diagnosed with incident prostate cancerProstate cancerStatin users had a 6% lower risk of prostate cancer vs. non-users (adjusted OR, 0.94, 95% CI, 0.91–0.97)
Ferris et al, 2012 (40)Case-control study recruited newly diagnosed glioma cases and frequency-matched controls (Columbia University and the University of California San Francisco)Recruited 517 glioma cases and 400 controls from 2007 to 2010GliomaSimvastatin and lovastatin showed significant inverse associations with glioma (OR, 0.49, 95% CI, 0.30–0.81 and OR, 0.47, 95% CI, 0.24–0.93, respectively). Statin intake for >120 months demonstrated the most significant associations (trend tests P=0.03)
Gaist et al, 2013 (41)Nationwide case-control study in Denmark based on population- based medical registriesPatients with a first diagnosis glioma during 2000–2009 (n=2, 656 cases and 18,480 controls). Prior statin use, short-term (<5 years) and long-term (5+ years)GliomaGlioma risk reduced among long-term statin users (OR, 0.76, 95% CI, 0.59–0.98) vs. never users, and was inversely related to the intensity of statin treatment among users (OR, 0.71, 95% CI, 0.44–1.15 for highest intensity)
Singh et al, 2013 (43)Systematic search of databases through August 2012Thirteen studies (one post-hoc analysis reporting 9,285 cases of of 22 RCTs) esophageal cancer among 1,132,969 patientsEsophageal cancerReported a significant reduction (28%) in esophageal cancer risk among patients on statins (adjusted OR, 0.72, 95% CI, 0.60–0.86)
Beales et al, 2013 (44)Systematic search performed across multiple databasesEleven observational studies. Studies examining adenocarcinoma development in BE: included 317 cancers and 1,999 controls. Esophageal cancers included 371,203 cancers and 6,083,150 controlsEsophageal cancerIn the Barrett’s population, statin use (OR, 0.57, 95% CI, 0.43–0.75) was independently associated with a reduced incidence of adenocarcinoma
Wu et al, 2013 (45)Literature search of databases up to March 2013Three post-hoc analyses of 26 RCTs involving 290 gastric cancers and 8 observational studies, totaling 7,321 gastric cancersGastric cancerStatins use significantly associated with a 27% reduction in gastric cancer risk (RR, 0.73, 95% CI, 0.58–0.93), with considerable heterogeneity among studies [I (2): 88.9%]
Singh et al, 2013 (126)Systematic search of multiple databases up to December 2012Included 11 studies (8 observational and three post-hoc analyses of 26 clinical trials) reporting 5,581 cases of gastric cancer (n=5,459,975)Gastric cancerStatin use associated with significant reduction of 32% in gastric cancer risk (adjusted OR, 0.68, 95% CI, 0.51–0.91)
Lytras et al, 2014 (46)Updated meta-analysis of published studiesForty studies, involving >8 million subjectsColorectal cancerRisk of colorectal cancer with statin use was not statistically significant among RCTs (RR, 0.89, 95% CI, 0.74–1.07; n=8), but reached statistical significance among cohort studies (RR, 0.91, 95% CI, 0.83–1.00; n=13) and case-control studies (RR, 0.92, 95% CI, 0.87–0.98; n=19)
Undela et al, 2012 (49)PubMed database and bibliographies of retrieved articles up to January 2012Twenty-four (13 cohort and 11 case-control) studies including 76,759 breast cancer cases and >2.4 million participantsBreast cancerStatin use and long-term statin use did not significantly affect breast cancer risk (RR, 0.99, 95% CI, 0.94–1.04 and RR, 1.03, 95% CI, 0.96–1.11, respectively)
McDougall et al, 2013 (48)Population-based case-control study (Seattle-Puget Sound region)Cases (916 IDC and 1,068 ILC) diagnosed between 2000 and 2008 were compared with 902 control womenBreast cancerCurrent users of statins for ≥10 years had a 1.83-fold increased risk of IDC (95% CI, 1.14–2.93) and a 1.97-fold increased risk of ILC (95% CI, 1.25–3.12) vs. never users
Brewer et al, 2013 (47)Retrospective cohort study (University of Texas, MD Anderson Cancer Center)A total of 723 patients diagnosed with primary IBC in 1995–2011. Clinical outcomes were compared by statin use and type [weakly lipophilic to hydrophilic (H-statin) vs. lipophilic statins (L-statin)Inflammatory breast cancerH-statin use was associated with significantly improved PFS vs. no statin (HR, 0.49, 95% CI, 0.28–0.84; P<0.01); OS and DSS P-values were 0.80 and 0.85, respectively. For L-statins vs. no statin, P-values for PFS, DSS and OS were 0.81, 0.4 and 0.74, respectively
Lavie et al, 2013 (50)Case-control study of newly diagnosed gynecological malignancies (CITOUS) (Clalit Health Services)Statin use assessed in a subset of 424 cases of ovarian and endometrial cancers and 341 controlsGynecological malignanciesStatin use >1 year prior to diagnosis associated with a significantly reduced risk of ovarian cancer (OR, 0.56, 95% CI, 0.33–0.94) and of endometrial cancer (OR, 0.59, 95% CI, 0.40–0.87)
Liu et al, 2012 (51)Nurses’ Health Study and Health Professionals Follow-Up StudyStatin use assessed in 80,782 women and 37,869 men, followed for 14 and 16 years, respectivelyRCCTwo-hundred seventy seven incident RCC cases (164 women and 113 men) were identified. Multivariate RR of current statin use vs. no current use was 0.68 (95% CI, 0.46–1.00) in women and 1.17 (95% CI, 0.75–1.82) in men
Singh et al, 2013 (52)Systematic search of databases through May 2012Ten studies reporting 4,298 cases of hepatocellular carcinoma in 1,459,417 patientsHepatocellular carcinomaAlthough the results were heterogeneous [P=0.01, I (2): 59%], statin users were less likely to develop hepatocellular carcinoma vs. non-users (adjusted OR, 0.63, 95% CI, 0.52–0.76)
Pradelli et al, 2013 (53)MEDLINE search until March 2012Five observational studies based on 2,574 cases of hepatocellular carcinomaHepatocellular carcinomaStatin treatment vs. no treatment was inversely related to hepatocellular carcinoma (summary RR, 0.58, 95% CI, 0.46–0.74). Between-study heterogeneity was significant (P<0.001) and numerically relevant (I, 65%)
Bonovas et al, 2008 (54)Comprehensive search for relevant articles published up to December 2007Twelve studies (3 randomized placebo-controlled trials, 4 cohort and 5 case-control studies)Pancreatic cancerNo association between statin use and pancreatic cancer in randomized placebo-controlled trials (RR, 0.99, 95% CI, 0.44–2.21) or observational studies (RR, 0.86, 95% CI, 0.60–1.24)
Cui et al, 2012 (55)Comprehensive search up to August 2011 for the relevant studiesSixteen studies involving 1,692,863 participants and 7,807 pancreatic cancer casesPancreatic cancerNon-significant decrease of pancreatic cancer risk among all statin users (RR, 0.89, 95% CI, 0.74–1.07). Significant heterogeneity detected among all studies (P<0.00001, I (2), 81%)
Deng et al, 2013 (56)Meta-analysis of databases up to September 2013Twenty-three studies (15 observational studies and 8 RCTs; n=4,864,582)Lung cancerNo association between statins and lung cancer risk reported among RCTs (RR, 0.95, 95% CI, 0.85–1.06) or observational studies (RR, 0.89, 95% CI, 0.77–1.04). Similar association noted in elderly individuals (6 observational studies; RR, 1.03, 95% CI, 0.96–1.11)
Tan et al, 2013 (57)Meta-analysis of the relevant articles in PubMed database up to March 2012Nineteen studies [5 RCTs (n=29,658) and 14 observational studies (n=4,979,746)] involving 38,013 lung cancer casesLung cancerNo association reported for statin use and risk of lung cancer either among RCTs (RR, 0.91, 95% CI, 0.76–1.09), cohort studies (RR, 0.94, 95% CI, 0.82–1.07), or case-control studies (RR, 0.82, 95% CI, 0.57–1.16)
Li et al, 2014 (58)Literature search of databases up to June 2013Twenty-one articles with 29 studiesSkin cancerNo association between statin and skin cancer among melanoma (RR, 0.94, 95% CI, 0.85–1.04) and non-melanoma skin cancer (RR, 1.03, 95% CI, 0.90–1.19)
da Silva et al, 2013 (59)Retrospective evaluation of records at 4 institutionsPatients treated with radical cystectomy and pelvic lymphadenectomy without neoadjuvant therapy (n=1,502)Urothelial carcinoma of the bladderMedian follow-up, 34 months; statin use associated with disease recurrence (P≤0.05) and cancer-specific mortality (P≤0.02) on univariable Cox regression analysis
Zhang et al, 2013 (60)Literature search of databases between January 1966 and October 2012Thirteen studies (3 RCTs, 5 cohort and 5 case-control)Bladder cancerNon-significant increase in total bladder cancer risk among all statin users (RR, 1.07, 95% CI, 0.95–1.21). Long-term statin use did not significantly affect total bladder cancer risk (RR, 1.21, 95% CI, 0.92–1.59)
Nielsen et al, 2012 (66)Entire Danish populationBetween 1995 and 2007, among patients ≥40 years, 18,721 were regular statin users prior to cancer diagnosis and 277,204 never usersCancer Multivariate-adjusted HR for statin users vs. never users was 0.85 (95% CI, 0.83–0.87) for death from any cause and 0.85 (95% CI, 0.82–0.87) for death from cancer
Cholesterol Treatment Trialists’ (CTT)Meta-analysis of individual patient recordsTwenty-two randomized trials. of statin vs control (n=134,537) and 5 trials of more intensive vs. lessCancerNo effect of statins for~5 years on newly diagnosed cancer or death from such cancers [statin vs. control; cancer incidence, 3,755 (1.4%/year) vs. 3,738 (1.4%/year), RR, 1.00
Collaboration et al, 2012 (67)intensive statin therapy (n=39,612)(95% CI, 0.96–1.05); cancer mortality, 1,365 (0.5%/year) vs. 1,358 (0.5%/year), RR, 1.00 (95% CI, 0.93–1.08)]
Katz et al, 2005 (90)Retrospective studyAny statin; median radiation therapy dose, 50.4 Gy (range, 45–55.8 Gy); evaluated in 349 and 308 patients (88%) receiving 5-FU-based chemotherapyNon- metastatic rectal cancersThirty-three (9%) patients used a statin, with no differences in clinical stage vs. other 324 patients. At the time of surgery, metastatic disease was observed in 23 (7%) non-statin vs. 0 statin patients. Unadjusted pCR rates with and without statin use were 30 and 17%, respectively (P=0.10). ORs for statin use on pCR was 4.2 (95% CI,1.7–12.1; P=0.003) after adjusting for NSAID use, clinical stage and chemotherapy type
Graf H et al, 2008 (78)Prospective cohort studyFifty-two patients received TACE combined with pravastatin (20–40 mg/day) and 131 patients received chemoembolization alone; observation period of up to 5 yearsHepatocellular carcinomaThirty-one (23.7%) out of 131 patients treated with TACE alone and 19 (36.5%) out of 52 patients treated with TACE and pravastatin survived. Significant longer median survival of patients in the TACE and pravastatin group (20.9 months, 95% CI, 15.5–26.3, P=0.003) vs. TACE alone (12.0 months, 95% CI, 10.3–13.7)

[i] BE, Barrett’s esophagus; CI, confidence interval; CITOUS, The Cancer in The Ovary and Uterus Study; DSS, disease-specific survival; HR, hazard ratio; IBC, inflammatory breast cancer; IDC, invasive ductal carcinoma; ILC, invasive lobular carcinoma; NSAID, non-steroidal anti-inflammatory drug; OR, odds ratio; OS, overall survival; pCR, pathologic complete response; PFS, progression-free survival; RCC, renal cell carcinoma; RCT, randomized controlled trial; RFS, recurrence-free survival; RR, relative risk; RT, radiotherapy; TACE, transarterial chemoembolization.

Antitumor effect of statins in experimental studies Lovastatin

In a phase I–II trial of lovastatin in anaplastic astrocytoma and glioblastoma multiforme, 18 patients received lovastatin between 20 and 30 mg/kg/day for 7 days followed by a 3-week rest. Lovastatin was considered well tolerated, as no patient reported myalgia and only 2 patients reported mild joint pain. Nine of 18 patients received concurrent radiation with no neurological toxicity, indicating that the combination was potentially safe. Of those who received concurrent radiation, 2 minor and 2 partial responses (duration range, 160–236 days) were observed. One patient each on lovastatin monotherapy showed partial and minor response, and stable disease. Notably, the patient who had partial response accomplished a response duration of >405 days, at which time lovastatin was discontinued due to cost-related issues (68).

Similarly, another phase I study evaluated the safety and tolerability of lovastatin using escalating doses in 88 cancer patients with advanced solid tumors. A majority of patients had prostate cancer or central nervous system tumors. Myopathy was found to be a dose-limiting toxicity and ubiquinone administration was associated with reversal of lovastatin-induced myopathy. Myopathy was prevented by its prophylactic administration in a 56-patient cohort. In the absence of supplementation, lovastatin was well tolerated up to 25 mg/kg/day for 7 days followed by a 3-week rest. One anaplastic astrocytoma patient treated with lovastatin at 30 and 35 mg/kg/day who progressed after surgical resection of the tumor, irradiation and 2 cycles of carmustine had a minor response (45% tumor size reduction) maintained for 8 months (69).

Prolongation of overall survival and PFS was documented in MM patients with lovastatin plus thalidomide and dexamethasone (TDL) vs. thalidomide and dexamethasone alone. The TDL regimen was safe and well tolerated (70).

Simvastatin

Simvastatin in combination with conventional FOLFIRI [irinotecan, 5-fluorouracil (5-FU), and leucovorin] in metastatic colorectal cancer patients showed promising antitumor activities (71). An exploratory subgroup analysis in non-small cell lung carcinoma patients with wild-type epidermal growth factor receptor (EGFR) non-adenocarcinomas showed higher RR, 40 vs. 0%, P=0.043) and longer PFS (3.6 vs. 1.7 months, P=0.027) with simvastatin plus gefitinib vs. gefitinib alone (72). Moreover, low-dose simvastatin to gemcitabine in advanced pancreatic cancer does not provide clinical benefit or results in increased toxicity (73).

Atorvastatin

The 6-month interventions with atorvastatin did not provide convincing evidence of colorectal cancer risk reduction in a multicenter phase II trial, although the relatively small sample size limited statistical power (74).

Fluvastatin

In patients with RCC and metastasis, zoledronate with fluvastatin or atorvastatin as bone-targeting therapy affected certain bone biomarkers and provided bone response in several patients. However, no statistically significant improvement in time to skeletal events was observed (75). The survival of pediatric brain stem tumor patients was significantly increased with metronomic treatment with carboplatin and vincristine associated with fluvastatin and thalidomide (76).

Pravastatin

In patients with acute myeloid leukemia (AML), pravastatin with idarubicin plus high-dose cytarabine (Ida-HDAC) decreased the total and low-density lipoprotein (LDL) cholesterol in almost all patients. The encouraging response rates suggest further trials evaluating the effect of cholesterol modulation on response in AML should be conducted (77). Chemoembolization and pravastatin combination significantly improved (P=0.003) survival of patients with advanced hepatocellular carcinoma vs. those receiving chemoembolization alone (78).

Pravastatin in combination with epirubicin, cisplatin, and capecitabine did not improve outcome in advanced gastric cancer patients in a randomized phase II trial (79). Summary of clinical trials that used statins as monotherapy or as a combination in patients with different cancer types are presented in Table III.

Table III

Summary of clinical trials that used statins as monotherapy or as combination in patients with different types of cancer.

Table III

Summary of clinical trials that used statins as monotherapy or as combination in patients with different types of cancer.

Authors, year (Refs.)PhaseTreatment regimen and durationPatient population and tumor typeTreatment outcomes
Kornblau et al, 2007 (77)IPravastatin (40–1,680 mg/day, days 1–8); Ida [(12 mg/(M2 × day), days 4–6] + high-dose cytarabine (Ara-C; HDAC) [1.5 g/(M2 × day) by CI, days 4–7]AML; included 15 newly diagnosed, 22 salvage patients with unfavorable (n=26) or intermediate (n=10) prognosis cytogeneticsCR/CRp was obtained in 11 of 15 new patients including 8 of 10 with unfavorable cytogenetics and 9 of 22 salvage patients. An MTD for pravastatin + Ida-HDAC was not reached
Sondergaard et al, 2009 (19)IIHigh-dose simvastatin (15 mg/kg/day) for 7 days followed by a 21-day rest in 24-week cyclesHeavily pretreated patients with multiple myeloma (n=6)TRACP (osteoclast) activity in serum and levels of collagen fragments (NTX) in urine increased for all patients. No reduction in free monoclonal light chains or monoclonal proteins with high-dose simvastatin was observed. The study was terminated prematurely due to transient increase in osteoclast activity and gastrointestinal side-effects
Schmidmaier et al, 2007 (18)IIConcomitant administration of simvastatin in myeloma patients refractory to two cycles of bortezomib or bendamustine during further two cyclesRefractory multiple myeloma (n=6)Intrapatient (cycles I/II vs. III/IV) and interpatient comparison (vs. 10 patients without simvastatin) showed reduction of drug resistance
Hus et al, 2011 (70)IIThalidomide, dexamethasone and lovastatin (TDL group, 49 patients) or thalidomide and dexamethasone (TD group, 42 patients)Relapsed or refractory myelomaA 50% reduction of monoclonal band observed in 32% of TD and 44% of TDL patients, with prolongation of OS and PFS in the latter
van der Spek et al, 2007 (127)IISimvastatin 15 mg/kg/day was orally prescribed on days 1–7 of a 28-day cycle, divided into two daily dosages followed by rapid IV infusion of vincristine (0.4 mg) and doxorubicin (9 mg/m2), and dexamethasone 40 mg (VAD) orally on day 7–10Relapsed or refractory myeloma (n=12)Treatment was feasible with mild side effects. However, only 1 of 12 patients achieved a PR
Larner et al, 1998 (68)I-II trialLovastatin at doses between 20 and 30 mg/kg/day for 7 days followed by a 3-week rest, with or without radiationAnaplastic astrocytoma and glioblastoma multiforme (n=18)Monotherapy (N=9), one PR and one minor response; combined therapy (N=9), two minor and two PR; overall dose well tolerated without neurological toxicity
López-Aguilar et al, 2008 (76)IIPatients received four courses of chemotherapy every 28 days [thalidomide alternating with fluvastatin every 14 days and combined with carboplatin and vincristine every 14 days followed by radiotherapy (56 cGy)] and four more courses of the same chemotherapyBrain stem tumors (n=9)Five patients had low-grade astrocytomas, three had glioblastoma multiforme, and one presented high-grade astrocytoma. Significant reduction in tumor volume and a significant increase in survival at 24 months observed
Han et al, 2011 (72)IIGefitinib (G) alone or 250 mg/day, (n=54) or gefitinib plus simvastatin (GS) (250 and 40 mg/day, respectively, n=52). Each cycle of 4 weeks continued until disease progression or intolerable toxicityAdvanced non-small cell lung cancerRR was 38.5% (95% CI, 25.3–51.7) for GS and 31.5% (95% CI, 19.1–43.9) for G. Median PFS was 3.3 (95% CI, 1.4–5.2) and 1.9 months (95% CI, 1.0–2.8) for GS and G, respectively. Median OS was 13.6 (95% CI, 7.1–20.1) and 12.0 months (95% CI, 7.8–16.2) for GS and G, respectively
Han et al, 2011 (128)IIIrinotecan (65 mg/m2) and cisplatin (30 mg/m2) (n=61) on days 1 and 8 every 3 weeks until death or disease progression along with oral simvastatin (40 mg, daily)Chemotherapy-naive patients with extensive-disease small cell lung cancerOne-year survival rate, 39.3%, median OS, 11 months; and median PFS, 6.1 months. Overall, RR, 75%
Konings et al, 2010 (79)IISix cycles of 3-weekly ECC with or without pravastatin (n=30) (40 mg once daily from day 1 of the first cycle until day 21 of the last cycle)Advanced gastric carcinomaECC + pravastatin, PFR at 6 months, 6/14 patients (42.8%); RR, 5/15 (33.3%); median PFS and OS, 6 and 8 months, respectively ECC, PFR at 6 months, 7/15 patients; (46.7%); RR, 7/15 (46.7%) median PFS and OS, 5 and 6 months, respectively
Kim et al, 2001 (129)IILovastatin 35 mg/kg/day for 7 consecutive days with ubiquinone (60 mg 4 times daily orally). Treatment was repeated every 28 days for 28 cycles. The median number of cycles was 2 (range, 1–4)Advanced measurable gastric adenocarcinoma (n=16)Fourteen patients were evaluated for response and toxicity. No patient achieved a response
Lee et al, 2009 (71)IISimvastatin oral 40 mg tablet once daily + conventional FOLFIRI chemotherapy. Treatment repeated every 2 weeks until disease progression or unacceptable toxicityMetastatic adenocarcinoma of the colon or rectum (n=49)ORR, ITT analysis, 46.9% (95% CI, 31.0–58.8), PP analysis, 45.8% (95% CI, 33.3–62.8); 1 complete response and 22 PRs. Disease-control rate, 83.7% (95% CI, 73.4–94.0). Median survival of all patients was 21.8 months (95% CI, 14.4–29.2); median TTP 9.9 months (95% CI, 6.4–13.3)
Limburg et al, 2011 (74)II(a) Atorvastatin 20 mg daily; (b) sulindac 150 mg twice daily; (c) oligofructose-enriched inulin (as ORAFTI® Synergy 1) 6 g twice daily; or (d) control (maltodextrin) 6 gm twice daily for 6 monthsPreviously resected colon cancer or multiple/advanced colorectal adenomas with ≥5 rectal ACFs at baseline (n=85)76/85 Patients completed the trial.
Median (SD) for % ΔACF was as below:
atorvastatin, 5.6 (−69–143%)
sulindac, −18.6 (−83–160%)
ORAFTI® Synergy 1, −3.6 (−88–83%)
control arms, −10.0 (−100–117%)
No statistically significant comparisons of % ΔACF and within-arm or between-arm
Manoukian et al, 2011 (75)Pilot trialPatients received zoledronate and fluvastatin or atorvastatin and followed for a median of 6 monthsRenal cell carcinoma and bone metastasis (n=11)Skeletal events, 7 (63%) patients; no skeletal events, 4 (36%) patients. Treatment response in 4 (36%) patients with the development of sclerosis in lytic bone lesions. Statistically significant differences in the median changes in biomarker levels between patients who had skeletal events and those who did not for DPD (P=0.03) and NTX (P=0.01), but not for BSAP (P=0.4)
Garwood et al, 2010 (21)Peri-operative window trialHigh-dose (80 mg/day) or low-dose (20 mg/day) fluvastatin for 3–6 weeks prior to surgery (n=45)DCIS or stage 1 breast cancerTwenty-nine paatients had paired Ki-67 primary endpoint data. Proliferation of high-grade tumors decreased by a median of 7.2% (P=0.008), which was statistically greater than the 0.3% decrease for low-grade tumors. More high-grade tumors had an increase in apoptosis (60 vs. 13%; P=0.015)
Lazzeroni et al, 2012 (130)IIParticipants were randomized to receive nimesulide 100 mg/day vs. simvastatin 20 mg/day vs. placebo for 1 year followed by a second year of follow-upWomen at higher risk for breast cancer (according to BRCAPRO), focused particularly on hormone non-responsive tumor risk (n=150)Role of ductal lavage was evaluated to study endpoint biomarkers and the effects of these drugs on breast carcinogenesis. Preliminary results showed that the treatment is well tolerated and the safety blood tests do not show any significant liver toxicity
Hong et al, 2014 (73)IIThree-week regimen with GS (gemcitabine 1,000 mg/m2 on days 1, 8 and 15 plus simvastatin 40 mg once daily) or GP (gemcitabine 1,000 mg/m2 on days 1, 8, and 15 plus placebo)Advanced and metastatic pancreatic cancer (n=114)Median TTP was 2.4 (95% CI, 0.7–4.1) and 3.6 months (95% CI, 3.1–4.1) in the GS and GP arms, respectively (P=0.903)
Thibault et al, 1996 (69)ISeven day courses of lovastatin given monthly at doses ranging from 2 to 45 mg/kg/dayAdvanced solid tumors (most patients had prostate cancer or central nervous system tumor, n=88)One minor response was documented in a patient with recurrent high-grade glioma
Kawata et al, 2001 (131)IIPatients underwent TAE followed by oral 5-FU 200 mg (−1) day for 2 months and then randomized to control (n=42) and pravastatin (n=41) administered at a daily dose of 40 mg for 16.5±9.8 months (means ± SD)Unresectable hepatocellular carcinoma (n=83)Median survival was 18 months in pravastatin vs. 9 months in control (P=0.006)
Lersch et al, 2004 (132)IIPatients received 3×200 mcg/day octreotide for 2 months followed by 20 mg octreotide LAR every 4 weeks (n=30) or 40–80 mg pravastatin (n=20) or 80–90 mg/m2 gemcitabine over 24 h weekly in cycles of 4 weeks (n=8)Hepatocellular carcinoma (n=58)Median OS was 5 months in octreotide, 7.2 and 3.5 months in the gemcitabine group. The difference between the pravastatin and gemcitabine groups was significant
Knox et al, 2005 (133)IEscalation of lovastatin starting at 5/mg/kg/day × 2 weeks, every 21 days, until MTD was reached; MTD was determined to be 7.5 mg/kg/day × 21days, every 28 daysSquamous cell carcinomas of the head and neck and of the cervix (n=26)Median survival of patients, 7.5 months (mean 9.2±1.5 months). Stable disease for >3 months was observed in 23% of patients. One patient achieved stable disease and clinical benefit for 14 months in the study and a further 23 months off treatment. Disease stabilization rate of 23% was observed

[i] ACF, aberrant crypt foci; AML, acute myeloid leukemia; BSAP, bone-specific alkaline phosphatase; CC3, cleaved caspase-3; CI, confidence interval; CR, complete remission; CRp, complete remission criteria met except that the platelet count was <100×109/l; DPD, deoxypyridinoline; ECC, epirubicin, cisplatin and capecitabine; FOLFIRI, irinotecan, 5-fluorouracil and leucovorin; Ida, idarubicin; ITT, intent-to-treat; MTD, maximum tolerable dose; NTX, N-telopeptide; ORR, overall response rate; OS, overall survival; PFR, progression-free rate; PFS, progression-free survival; PP, per protocol; PR, partial response; RR, relative risk; SD, standard deviation; TAE, transcatheter arterial embolization; TRACP, tartrate-resistant acid phosphatase; TTP, time to progression.

5. Synergism of statins with radiotherapy and chemotherapy

Statins act by arresting cells in the late G1 phase of the cell cycle and can affect cell synchronization in the radiosensitive phase (5). The late G1 and G2-M phases are most sensitive to radiation therapy; therefore, statins potentially sensitized cells to radiation in the late G1 phase (80,81). The antitumor effect of lovastatin as a radiosensitizer on B-cell rat lymphoma (L-TACB) was higher than that of individual therapy (82). The underlying molecular mechanism involved Ras, which confers intrinsic resistance to radiation since in vitro studies using osteosarcoma cells demonstrated that lovastatin decreases this radiation resistance (80,81). Furthermore, HMGCR may serve as a predictive marker of response to postoperative radiotherapy in ductal carcinoma in situ (DCIS) (83). A retrospective cohort study suggested an association between statin therapy and improvement in response of rectal cancer to neoadjuvant chemoradiation (84).

Statins have shown anticancer potential with numerous chemotherapeutic agents. Simvastatin showed additive activity and mutual sensitization with doxorubicin by triggering caspase activation in human rhabdomyosarcoma cells (85). When combined with 5-FU or cisplatin as chemotherapy, lovastatin acts by inhibiting geranylgeranylation but not farnesylation of target protein(s) in colon cancer cells (10). Lovastatin or simvastatin with cytosine arabinoside significantly enhances the antiproliferative effect of each drug in leukemia cell lines and this may be beneficial in the leukemia treatment (86,87). A similar synergy of simvastatin with N,N′-bis(2-chloroethyl)-N-nitrosourea or β-interferon produces antiproliferative activity in human glioma cells (88). Cerivastatin also increases the cytotoxicity of 5-FU in chemoresistant colorectal cancer cell lines by inhibiting nuclear factor-κB DNA-binding activity (89).

A phase I study in AML patients showed synergistic effects of the addition of pravastatin to a conventional chemotherapy regimen (idarubicin and high-dose cytarabine) (77). Statin use in combination with concurrent chemoradiotherapy in preoperative rectal carcinoma patients was associated with improved pathologic complete response at the time of surgery (90). HMGCR expression was reported as an independent predictor of prolonged recurrence-free survival in primary ovarian cancer. Future studies are required to evaluate HMGCR expression as a surrogate marker of response to statin treatment, particularly in conjunction with current chemotherapeutic regimens (91). Synergistic effects are observed in patients with relapsed or refractory myeloma by the addition of lovastatin to thalidomide and dexamethasone (70).

6. Conclusion

Preclinical data based on cancer cell lines and animal models demonstrate encouraging anticancer activity of statins. Similarly, several population-based and retrospective studies demonstrate chemopreventive and survival benefit of statins in various types of cancer. However, this benefit has not been confirmed/proven or validated in clinical trials, and is attributed to the absence of well-conducted large-scale phase III RCTs that have addressed the antitumor effects of statins in cancer. In fact, a majority of the trials thus far are phase I and/or small or poorly conducted phase II clinical trials with a small sample size and inadequate power. Moreover, genetic and non-genetic factors also may contribute to the inter-individual variation in statin response. In ovarian cancer, HMGCR expression was reported as an independent predictor of prolonged recurrence-free survival (91). Lipkin et al, identified a single-nucleotide polymorphism in the HMGCR gene that significantly modified the chemopreventive activity of statins for colorectal cancer risk (92). Heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) overexpression was recently reported to reduce HMGCR enzyme activity, enhance LDL-C uptake, and increase cellular apolipoprotein B (93). This may explain the inter-individual variation of drug response to statins. Advances in molecular biology may be useful to identify markers responsive to statin treatment and tailor base statin treatment based on genotypic profile, in the direction of personalized medicine.

Studies suggest statins can modulate the outcome of various cancer types and notably can target cancer vs. normal cells. The microenvironments seem to regulate the statin effect in different types of cancer. The side-effects appear to be limited, manageable and may be associated with genetic and non-genetic factors. Future studies should concentrate on evaluating statins in large-scale phase III RCTs in cancer patients to establish the precise effect of stains in cancer prevention and treatment.

Acknowledgements

This study was supported in part by King Fahad Medical City, Riyadh, Saudi Arabia.

References

1 

Ferlay J, Soerjomataram I, Ervik M, et al: GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 (Internet). Lyon, France: International Agency for Research on Cancer; 2013, Available from: http://globocan.iarc.fr. Accessed.

2 

Kanu OO, Mehta A, Di C, Lin N, et al: Glioblastoma multiforme: a review of therapeutic targets. Expert Opin Ther Targets. 13:701–718. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Goldstein JL and Brown MS: Regulation of the mevalonate pathway. Nature. 343:425–430. 1990. View Article : Google Scholar : PubMed/NCBI

4 

Gazzerro P, Proto MC, Gangemi G, et al: Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev. 64:102–146. 2012. View Article : Google Scholar

5 

Chan KK, Oza AM and Siu LL: The statins as anticancer agents. Clin Cancer Res. 9:10–19. 2003.PubMed/NCBI

6 

Baserga R: Molecular biology of the cell cycle. Int J Radiat Biol Relat Stud Phys Chem Med. 49:219–226. 1986. View Article : Google Scholar : PubMed/NCBI

7 

Tapia-Pérez JH, Kirches E, Mawrin C, Firsching R and Schneider T: Cytotoxic effect of different statins and thiazolidinediones on malignant glioma cells. Cancer Chemother Pharmacol. 67:1193–1201. 2011. View Article : Google Scholar

8 

Rao S, Lowe M, Herliczek TW and Keyomarsi K: Lovastatin mediated G1 arrest in normal and tumor breast cells is through inhibition of CDK2 activity and redistribution of p21 and p27, independent of p53. Oncogene. 17:2393–2402. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Collisson EA, Kleer C, Wu M, et al: Atorvastatin prevents RhoC isoprenylation, invasion, and metastasis in human melanoma cells. Mol Cancer Ther. 2:941–948. 2003.PubMed/NCBI

10 

Agarwal B, Bhendwal S, Halmos B, Moss SF, Ramey WG and Holt PR: Lovastatin augments apoptosis induced by chemotherapeutic agents in colon cancer cells. Clin Cancer Res. 5:2223–2229. 1999.PubMed/NCBI

11 

Marcelli M, Cunningham GR, Haidacher SJ, et al: Caspase-7 is activated during lovastatin-induced apoptosis of the prostate cancer cell line LNCaP. Cancer Res. 58:76–83. 1998.PubMed/NCBI

12 

Koyuturk M, Ersoz M and Altiok N: Simvastatin induces apoptosis in human breast cancer cells: p53 and estrogen receptor independent pathway requiring signalling through JNK. Cancer Lett. 250:220–228. 2007. View Article : Google Scholar

13 

Baetta R, Donetti E, Comparato C, et al: Pro-apoptotic effect of atorvastatin on stimulated rabbit smooth muscle cells. Pharmacol Res. 36:115–121. 1997. View Article : Google Scholar : PubMed/NCBI

14 

Wang IK, Lin-Shiau SY and Lin JK: Induction of apoptosis by lovastatin through activation of caspase-3 and DNase II in leukaemia HL-60 cells. Pharmacol Toxicol. 86:83–91. 2000. View Article : Google Scholar : PubMed/NCBI

15 

He Z, Mangala LS, Theriot CA, Rohde LH, Wu H and Zhang Y: Cell killing and radiosensitizing effects of atorvastatin in PC3 prostate cancer cells. J Radiat Res. 53:225–233. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Denoyelle C, Vasse M, Körner M, et al: Cerivastatin, an inhibitor of HMG-CoA reductase, inhibits the signaling pathways involved in the invasiveness and metastatic properties of highly invasive breast cancer cell lines: an in vitro study. Carcinogenesis. 22:1139–1148. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Bouterfa HL, Sattelmeyer V, Czub S, Vordermark D, Roosen K and Tonn JC: Inhibition of Ras farnesylation by lovastatin leads to downregulation of proliferation and migration in primary cultured human glioblastoma cells. Anticancer Res. 20:2761–2771. 2000.PubMed/NCBI

18 

Schmidmaier R, Baumann P, Bumeder I, Meinhardt G, Straka C and Emmerich B: First clinical experience with simvastatin to overcome drug resistance in refractory multiple myeloma. Eur J Haematol. 79:240–243. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Sondergaard TE, Pedersen PT, Andersen TL, et al: A phase II clinical trial does not show that high dose simvastatin has beneficial effect on markers of bone turnover in multiple myeloma. Hematol Oncol. 27:17–22. 2009. View Article : Google Scholar

20 

Bjarnadottir O, Romero Q, Bendahl PO, et al: Targeting HMG-CoA reductase with statins in a window-of-opportunity breast cancer trial. Breast Cancer Res Treat. 138:499–508. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Garwood ER, Kumar AS, Baehner FL, et al: Fluvastatin reduces proliferation and increases apoptosis in women with high grade breast cancer. Breast Cancer Res Treat. 119:137–144. 2010. View Article : Google Scholar

22 

Inano H, Suzuki K, Onoda M and Wakabayashi K: Anti-carcinogenic activity of simvastatin during the promotion phase of radiation-induced mammary tumorigenesis of rats. Carcinogenesis. 18:1723–1727. 1997. View Article : Google Scholar : PubMed/NCBI

23 

Narisawa T, Fukaura Y, Tanida N, Hasebe M, Ito M and Aizawa R: Chemopreventive efficacy of low dose of pravastatin, an HMG-CoA reductase inhibitor, on 1,2-dimethylhydrazine-induced colon carcinogenesis in ICR mice. Tohoku J Exp Med. 180:131–138. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Narisawa T, Morotomi M, Fukaura Y, Hasebe M, Ito M and Aizawa R: Chemoprevention by pravastatin, a 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor, of N-methyl-N-nitrosourea-induced colon carcinogenesis in F344 rats. Jpn J Cancer Res. 87:798–804. 1996. View Article : Google Scholar : PubMed/NCBI

25 

Clutterbuck RD, Millar BC, Powles RL, et al: Inhibitory effect of simvastatin on the proliferation of human myeloid leukaemia cells in severe combined immunodeficient (SCID) mice. Br J Haematol. 102:522–527. 1998. View Article : Google Scholar : PubMed/NCBI

26 

Kikuchi T, Nagata Y and Abe T: In vitro and in vivo antiproliferative effects of simvastatin, an HMG-CoA reductase inhibitor, on human glioma cells. J Neurooncol. 34:233–239. 1997. View Article : Google Scholar : PubMed/NCBI

27 

Hawk MA, Cesen KT, Siglin JC, Stoner GD and Ruch RJ: Inhibition of lung tumor cell growth in vitro and mouse lung tumor formation by lovastatin. Cancer Lett. 109:217–222. 1996. View Article : Google Scholar : PubMed/NCBI

28 

Matar P, Rozados VR, Binda MM, Roggero EA, Bonfil RD and Scharovsky OG: Inhibitory effect of lovastatin on spontaneous metastases derived from a rat lymphoma. Clin Exp Metastasis. 17:19–25. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Matar P, Rozados VR, Roggero EA and Scharovsky OG: Lovastatin inhibits tumor growth and metastasis development of a rat fibrosarcoma. Cancer Biother Radiopharm. 13:387–393. 1998. View Article : Google Scholar

30 

Alonso DF, Farina HG, Skilton G, Gabri MR, De Lorenzo MS and Gomez DE: Reduction of mouse mammary tumor formation and metastasis by lovastatin, an inhibitor of the mevalonate pathway of cholesterol synthesis. Breast Cancer Res Treat. 50:83–93. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Broitman SA, Wilkinson JT IV, Cerda S and Branch SK: Effects of monoterpenes and mevinolin on murine colon tumor CT-26 in vitro and its hepatic ‘metastases’ in vivo. Adv Exp Med Biol. 401:111–130. 1996. View Article : Google Scholar

32 

Jani JP, Specht S, Stemmler N, et al: Metastasis of B16F10 mouse melanoma inhibited by lovastatin, an inhibitor of cholesterol biosynthesis. Invasion Metastasis. 13:314–324. 1993.PubMed/NCBI

33 

Feleszko W, Mlynarczuk I, Balkowiec-Iskra EZ, et al: Lovastatin potentiates antitumor activity and attenuates cardiotoxicity of doxorubicin in three tumor models in mice. Clin Cancer Res. 6:2044–2052. 2000.PubMed/NCBI

34 

Feleszko W, Bałkowiec EZ, Sieberth E, et al: Lovastatin and tumor necrosis factor-α exhibit potentiated antitumor effects against Ha-ras-transformed murine tumor via inhibition of tumor-induced angiogenesis. Int J Cancer. 81:560–567. 1999. View Article : Google Scholar : PubMed/NCBI

35 

Mucci LA and Stampfer MJ: Mounting evidence for prediagnostic use of statins in reducing risk of lethal prostate cancer. J Clin Oncol. 32:1–2. 2014. View Article : Google Scholar

36 

Platz EA, Leitzmann MF, Visvanathan K, et al: Statin drugs and risk of advanced prostate cancer. J Natl Cancer Inst. 98:1819–1825. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Jespersen CG, Norgaard M, Friis S, Skriver C and Borre M: Statin use and risk of prostate cancer: a Danish population-based case-control study, 1997–2010. Cancer Epidemiol. 38:42–47. 2014. View Article : Google Scholar

38 

Yu O, Eberg M, Benayoun S, et al: Use of statins and the risk of death in patients with prostate cancer. J Clin Oncol. 32:5–11. 2014. View Article : Google Scholar

39 

Geybels MS, Wright JL, Holt SK, Kolb S, Feng Z and Stanford JL: Statin use in relation to prostate cancer outcomes in a population-based patient cohort study. Prostate. 73:1214–1222. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Ferris JS, McCoy L, Neugut AI, Wrensch M and Lai R: HMG CoA reductase inhibitors, NSAIDs and risk of glioma. Int J Cancer. 131:E1031–E1037. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Gaist D, Andersen L, Hallas J, Sørensen HT, Schroder HD and Friis S: Use of statins and risk of glioma: a nationwide case-control study in Denmark. Br J Cancer. 108:715–720. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Tsibouris P, Vlachou E and Isaacs PE: Role of chemoprophylaxis with either NSAIDs or statins in patients with Barrett’s esophagus. World J Gastrointest Pharmacol Ther. 5:27–39. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Singh S, Singh AG, Singh PP, Murad MH and Iyer PG: Statins are associated with reduced risk of esophageal cancer, particularly in patients with Barrett’s esophagus: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 11:620–629. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Beales IL, Hensley A and Loke Y: Reduced esophageal cancer incidence in statin users, particularly with cyclo-oxygenase inhibition. World J Gastrointest Pharmacol Ther. 4:69–79. 2013.PubMed/NCBI

45 

Wu XD, Zeng K, Xue FQ, Chen JH and Chen YQ: Statins are associated with reduced risk of gastric cancer: a meta-analysis. Eur J Clin Pharmacol. 69:1855–1860. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Lytras T, Nikolopoulos G and Bonovas S: Statins and the risk of colorectal cancer: an updated systematic review and meta-analysis of 40 studies. World J Gastroenterol. 20:1858–1870. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Brewer TM, Masuda H, Liu DD, et al: Statin use in primary inflammatory breast cancer: a cohort study. Br J Cancer. 109:318–324. 2013. View Article : Google Scholar : PubMed/NCBI

48 

McDougall JA, Malone KE, Daling JR, Cushing-Haugen KL, Porter PL and Li CI: Long-term statin use and risk of ductal and lobular breast cancer among women 55 to 74 years of age. Cancer Epidemiol Biomarkers Prev. 22:1529–1537. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Undela K, Srikanth V and Bansal D: Statin use and risk of breast cancer: a meta-analysis of observational studies. Breast Cancer Res Treat. 135:261–269. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Lavie O, Pinchev M, Rennert HS, Segev Y and Rennert G: The effect of statins on risk and survival of gynecological malignancies. Gynecol Oncol. 130:615–619. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Liu W, Choueiri TK and Cho E: Statin use and the risk of renal cell carcinoma in 2 prospective US cohorts. Cancer. 118:797–803. 2012. View Article : Google Scholar

52 

Singh S, Singh PP, Singh AG, Murad MH and Sanchez W: Statins are associated with a reduced risk of hepatocellular cancer: a systematic review and meta-analysis. Gastroenterology. 144:323–332. 2013. View Article : Google Scholar

53 

Pradelli D, Soranna D, Scotti L, et al: Statins and primary liver cancer: a meta-analysis of observational studies. Eur J Cancer Prev. 22:229–234. 2013. View Article : Google Scholar

54 

Bonovas S, Filioussi K and Sitaras NM: Statins are not associated with a reduced risk of pancreatic cancer at the population level, when taken at low doses for managing hyper-cholesterolemia: evidence from a meta-analysis of 12 studies. Am J Gastroenterol. 103:2646–2651. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Cui X, Xie Y, Chen M, et al: Statin use and risk of pancreatic cancer: a meta-analysis. Cancer Causes Control. 23:1099–1111. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Deng Z, Zhang S, Yi L and Chen S: Can statins reduce risk of lung cancer, especially among elderly people? A meta-analysis. Chin J Cancer Res. 25:679–688. 2013.

57 

Tan M, Song X, Zhang G, et al: Statins and the risk of lung cancer: a meta-analysis. PLoS One. 8:e573492013. View Article : Google Scholar : PubMed/NCBI

58 

Li X, Wu XB and Chen Q: Statin use is not associated with reduced risk of skin cancer: a meta-analysis. Br J Cancer. 110:802–807. 2014. View Article : Google Scholar :

59 

da Silva RD, Xylinas E, Kluth L, et al: Impact of statin use on oncologic outcomes in patients with urothelial carcinoma of the bladder treated with radical cystectomy. J Urol. 190:487–492. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Zhang XL, Geng J, Zhang XP, et al: Statin use and risk of bladder cancer: a meta-analysis. Cancer Causes Control. 24:769–776. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Chan TF, Wu CH, Lin CL and Yang CY: Statin use and the risk of breast cancer: a population-based case-control study. Expert Opin Drug Saf. 13:287–293. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Chan TF, Chiu HF, Wu CH, Lin CL and Yang CY: Statin use and the risk of esophageal cancer: a population-based case-control study. Expert Opin Drug Saf. 12:293–298. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Kuo CC, Chiu HF, Lee IM, Kuo HW, Lee CT and Yang CY: Statin use and the risk of bladder cancer: a population-based case-control study. Expert Opin Drug Saf. 11:733–738. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Chiu HF, Kuo CC, Kuo HW, Lee IM, Lee CT and Yang CY: Statin use and the risk of kidney cancer: a population-based case-control study. Expert Opin Drug Saf. 11:543–549. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Cheng MH, Chiu HF, Ho SC and Yang CY: Statin use and the risk of female lung cancer: a population-based case-control study. Lung Cancer. 75:275–279. 2012. View Article : Google Scholar

66 

Nielsen SF, Nordestgaard BG and Bojesen SE: Statin use and reduced cancer-related mortality. N Engl J Med. 367:1792–1802. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Cholesterol Treatment Trialists’ (CTT) Collaboration. Emberson JR, Kearney PM, Blackwell L, et al: Lack of effect of lowering LDL cholesterol on cancer: meta-analysis of individual data from 175,000 people in 27 randomised trials of statin therapy. PLoS One. 7:e298492012. View Article : Google Scholar : PubMed/NCBI

68 

Larner J, Jane J, Laws E, Packer R, Myers C and Shaffrey M: A phase I–II trial of lovastatin for anaplastic astrocytoma and glioblastoma multiforme. Am J Clin Oncol. 21:579–583. 1998. View Article : Google Scholar : PubMed/NCBI

69 

Thibault A, Samid D, Tompkins AC, et al: Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer. Clin Cancer Res. 2:483–491. 1996.PubMed/NCBI

70 

Hus M, Grzasko N, Szostek M, et al: Thalidomide, dexamethasone and lovastatin with autologous stem cell transplantation as a salvage immunomodulatory therapy in patients with relapsed and refractory multiple myeloma. Ann Hematol. 90:1161–1166. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Lee J, Jung KH, Park YS, et al: Simvastatin plus irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI) as first-line chemotherapy in metastatic colorectal patients: a multicenter phase II study. Cancer Chemother Pharmacol. 64:657–663. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Han JY, Lee SH, Yoo NJ, et al: A randomized phase II study of gefitinib plus simvastatin versus gefitinib alone in previously treated patients with advanced non-small cell lung cancer. Clin Cancer Res. 17:1553–1560. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Hong JY, Nam EM, Lee J, et al: Randomized double-blinded, placebo-controlled phase II trial of simvastatin and gemcitabine in advanced pancreatic cancer patients. Cancer Chemother Pharmacol. 73:125–130. 2014. View Article : Google Scholar

74 

Limburg PJ, Mahoney MR, Ziegler KL, et al: Randomized phase II trial of sulindac, atorvastatin, and prebiotic dietary fiber for colorectal cancer chemoprevention. Cancer Prev Res. 4:259–269. 2011. View Article : Google Scholar

75 

Manoukian GE, Tannir NM, Jonasch E, Qiao W, Haygood TM and Tu SM: Pilot trial of bone-targeted therapy combining zoledronate with fluvastatin or atorvastatin for patients with metastatic renal cell carcinoma. Clin Genitourin Cancer. 9:81–88. 2011. View Article : Google Scholar : PubMed/NCBI

76 

López-Aguilar E, Sepúlveda-Vildósola AC, Betanzos-Cabrera Y, et al: Phase II study of metronomic chemotherapy with thalidomide, carboplatin-vincristine-fluvastatin in the treatment of brain stem tumors in children. Arch Med Res. 39:655–662. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Kornblau SM, Banker DE, Stirewalt D, et al: Blockade of adaptive defensive changes in cholesterol uptake and synthesis in AML by the addition of pravastatin to idarubicin + high-dose Ara-C: a phase 1 study. Blood. 109:2999–3006. 2007.

78 

Graf H, Jüngst C, Straub G, et al: Chemoembolization combined with pravastatin improves survival in patients with hepatocellular carcinoma. Digestion. 78:34–38. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Konings IR, van der Gaast A, van der Wijk LJ, de Jongh FE, Eskens FA and Sleijfer S: The addition of pravastatin to chemotherapy in advanced gastric carcinoma: a randomised phase II trial. Eur J Cancer. 46:3200–3204. 2010. View Article : Google Scholar : PubMed/NCBI

80 

McKenna WG, Weiss MC, Bakanauskas VJ, et al: The role of the H-ras oncogene in radiation resistance and metastasis. Int J Radiat Oncol Biol Phys. 18:849–859. 1990. View Article : Google Scholar : PubMed/NCBI

81 

Miller AC, Kariko K, Myers CE, Clark EP and Samid D: Increased radioresistance of EJras-transformed human osteo-sarcoma cells and its modulation by lovastatin, an inhibitor of p21ras isoprenylation. Int J Cancer. 53:302–307. 1993. View Article : Google Scholar : PubMed/NCBI

82 

Rozados VR, Hinrichsen LI, McDonnell J and Scharovsky OG: Lovastatin enhances in vitro radiation-induced apoptosis of rat B-cell lymphoma cells. J Exp Clin Cancer Res. 24:55–61. 2005.PubMed/NCBI

83 

Butt S, Butt T, Jirström K, et al: The target for statins, HMG-CoA reductase, is expressed in ductal carcinoma-in situ and may predict patient response to radiotherapy. Ann Surg Oncol. 21:2911–2919. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Mace AG, Gantt GA, Skacel M, Pai R, Hammel JP and Kalady MF: Statin therapy is associated with improved pathologic response to neoadjuvant chemoradiation in rectal cancer. Dis Colon Rectum. 56:1217–1227. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Werner M, Sacher J and Hohenegger M: Mutual amplification of apoptosis by statin-induced mitochondrial stress and doxorubicin toxicity in human rhabdomyosarcoma cells. Br J Pharmacol. 143:715–724. 2004. View Article : Google Scholar : PubMed/NCBI

86 

Holstein SA and Hohl RJ: Interaction of cytosine arabinoside and lovastatin in human leukemia cells. Leuk Res. 25:651–660. 2001. View Article : Google Scholar : PubMed/NCBI

87 

Lishner M, Bar-Sef A, Elis A and Fabian I: Effect of simvastatin alone and in combination with cytosine arabinoside on the proliferation of myeloid leukemia cell lines. J Investig Med. 49:319–324. 2001. View Article : Google Scholar : PubMed/NCBI

88 

Soma MR, Pagliarini P, Butti G, Paoletti R, Paoletti P and Fumagalli R: Simvastatin, an inhibitor of cholesterol biosynthesis, shows a synergistic effect with N,N′-bis(2-chloroethyl)-N-nitrosourea and β-interferon on human glioma cells. Cancer Res. 52:4348–4355. 1992.PubMed/NCBI

89 

Wang W, Collie-Duguid E and Cassidy J: Cerivastatin enhances the cytotoxicity of 5-fluorouracil on chemosensitive and resistant colorectal cancer cell lines. FEBS Lett. 531:415–420. 2002. View Article : Google Scholar : PubMed/NCBI

90 

Katz MS, Minsky BD, Saltz LB, Riedel E, Chessin DB and Guillem JG: Association of statin use with a pathologic complete response to neoadjuvant chemoradiation for rectal cancer. Int J Radiat Oncol Biol Phys. 62:1363–1370. 2005. View Article : Google Scholar : PubMed/NCBI

91 

Brennan DJ, Brandstedt J, Rexhepaj E, et al: Tumour-specific HMG-CoAR is an independent predictor of recurrence free survival in epithelial ovarian cancer. BMC Cancer. 10:1252010. View Article : Google Scholar : PubMed/NCBI

92 

Lipkin SM, Chao EC, Moreno V, et al: Genetic variation in 3-hydroxy-3-methylglutaryl CoA reductase modifies the chemo-preventive activity of statins for colorectal cancer. Cancer Prev Res. 3:597–603. 2010. View Article : Google Scholar

93 

Yu CY, Theusch E, Lo K, et al: HNRNPA1 regulates HMGCR alternative splicing and modulates cellular cholesterol metabolism. Hum Mol Genet. 23:319–332. 2014. View Article : Google Scholar

94 

Sławińska-Brych A, Zdzisińska B and Kandefer-Szerszeń M: Fluvastatin inhibits growth and alters the malignant phenotype of the C6 glioma cell line. Pharmacol Rep. 66:121–129. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Yongjun Y, Shuyun H, Lei C, Xiangrong C, Zhilin Y and Yiquan K: Atorvastatin suppresses glioma invasion and migration by reducing microglial MT1-MMP expression. J Neuroimmunol. 260:1–8. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Crosbie J, Magnussen M, Dornbier R, Iannone A and Steele TA: Statins inhibit proliferation and cytotoxicity of a human leukemic natural killer cell line. Biomark Res. 1:332013. View Article : Google Scholar : PubMed/NCBI

97 

Al-Haidari AA, Syk I and Thorlacius H: HMG-CoA reductase regulates CCL17-induced colon cancer cell migration via geranylgeranylation and RhoA activation. Biochem Biophys Res Commun. 446:68–72. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Ishikawa S, Hayashi H, Kinoshita K, et al: Statins inhibit tumor progression via an enhancer of zeste homolog 2-mediated epigenetic alteration in colorectal cancer. Int J Cancer. 135:2528–2536. 2014. View Article : Google Scholar :

99 

Chang HL, Chen CY, Hsu YF, et al: Simvastatin induced HCT116 colorectal cancer cell apoptosis through p38MAPK-p53-survivin signaling cascade. Biochim Biophys Acta. 1830:4053–4064. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Rentala S, Chintala R, Guda M, Chintala M, Komarraju AL and Mangamoori LN: Atorvastatin inhibited Rho-associated kinase 1 (ROCK1) and focal adhesion kinase (FAK) mediated adhesion and differentiation of CD133+CD44+ prostate cancer stem cells. Biochem Biophys Res Commun. 441:586–592. 2013. View Article : Google Scholar : PubMed/NCBI

101 

Peng X, Li W, Yuan L, Mehta RG, Kopelovich L and McCormick DL: Inhibition of proliferation and induction of autophagy by atorvastatin in PC3 prostate cancer cells correlate with downregulation of Bcl2 and upregulation of miR-182 and p21. PLoS One. 8:e704422013. View Article : Google Scholar : PubMed/NCBI

102 

Al-Husein B, Goc A and Somanath PR: Suppression of interactions between prostate tumor cell-surface integrin and endothelial ICAM-1 by simvastatin inhibits micrometastasis. J Cell Physiol. 228:2139–2148. 2013. View Article : Google Scholar : PubMed/NCBI

103 

Fang Z, Tang Y, Fang J, et al: Simvastatin inhibits renal cancer cell growth and metastasis via AKT/mTOR, ERK and JAK2/STAT3 pathway. PLoS One. 8:e628232013. View Article : Google Scholar : PubMed/NCBI

104 

Islam M, Sharma S, Kumar B and Teknos TN: Atorvastatin inhibits RhoC function and limits head and neck cancer metastasis. Oral Oncol. 49:778–786. 2013. View Article : Google Scholar : PubMed/NCBI

105 

Yu X, Pan Y, Ma H and Li W: Simvastatin inhibits proliferation and induces apoptosis in human lung cancer cells. Oncol Res. 20:351–357. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Pelaia G, Gallelli L, Renda T, et al: Effects of statins and farnesyl transferase inhibitors on ERK phosphorylation, apoptosis and cell viability in non-small lung cancer cells. Cell Prolif. 45:557–565. 2012. View Article : Google Scholar : PubMed/NCBI

107 

Chen J, Liu B, Yuan J, et al: Atorvastatin reduces vascular endothelial growth factor (VEGF) expression in human non-small cell lung carcinomas (NSCLCs) via inhibition of reactive oxygen species (ROS) production. Mol Oncol. 6:62–72. 2012. View Article : Google Scholar

108 

Guterres FA, Martinez GR, Rocha ME and Winnischofer SM: Simvastatin rises reactive oxygen species levels and induces senescence in human melanoma cells by activation of p53/p21 pathway. Exp Cell Res. 319:2977–2988. 2013. View Article : Google Scholar : PubMed/NCBI

109 

Pich C, Teiti I, Rochaix P, et al: Statins reduce melanoma development and metastasis through MICA overexpression. Front Immunol. 4:622013. View Article : Google Scholar : PubMed/NCBI

110 

Gopalan A, Yu W, Sanders BG and Kline K: Simvastatin inhibition of mevalonate pathway induces apoptosis in human breast cancer cells via activation of JNK/CHOP/DR5 signaling pathway. Cancer Lett. 329:9–16. 2013. View Article : Google Scholar

111 

Park YH, Jung HH, Ahn JS and Im YH: Statin induces inhibition of triple negative breast cancer (TNBC) cells via PI3K pathway. Biochem Biophys Res Commun. 439:275–279. 2013. View Article : Google Scholar : PubMed/NCBI

112 

Zhao Z, Cao X, Pan Y, Sha S, Zhao T and Zhang T: Simvastatin downregulates HER2 via upregulation of PEA3 to induce cell death in HER2-positive breast cancer cells. Oncol Res. 20:187–195. 2012. View Article : Google Scholar : PubMed/NCBI

113 

Qi XF, Zheng L, Lee KJ, et al: HMG-CoA reductase inhibitors induce apoptosis of lymphoma cells by promoting ROS generation and regulating Akt, Erk and p38 signals via suppression of mevalonate pathway. Cell Death Dis. 4:e5182013. View Article : Google Scholar : PubMed/NCBI

114 

Zeng M, Gu WY, Jiang TX, et al: Effects of simvastatin on PI3K/AKT signaling pathway in human acute monocytic leukemia cell line SHI-1. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 20:268–272. 2012.(In Chinese). PubMed/NCBI

115 

Yang SS, Li R, Qu X, Fang W and Quan Z: Atorvastatin decreases Toll-like receptor 4 expression and downstream signaling in human monocytic leukemia cells. Cell Immunol. 279:96–102. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Shi J, Zhu J, Zhao H, Zhong C, Xu Z and Yao F: Mevalonate pathway is a therapeutic target in esophageal squamous cell carcinoma. Tumour Biol. 34:429–435. 2013. View Article : Google Scholar

117 

Ma L, Niknejad N, Gorn-Hondermann I, Dayekh K and Dimitroulakos J: Lovastatin induces multiple stress pathways including LKB1/AMPK activation that regulate its cytotoxic effects in squamous cell carcinoma cells. PLoS One. 7:e460552012. View Article : Google Scholar : PubMed/NCBI

118 

Tu YS, Kang XL, Zhou JG, Lv XF, Tang YB and Guan YY: Involvement of Chk1-Cdc25A-cyclin A/CDK2 pathway in simvastatin induced S-phase cell cycle arrest and apoptosis in multiple myeloma cells. Eur J Pharmacol. 670:356–364. 2011. View Article : Google Scholar : PubMed/NCBI

119 

Araki M, Maeda M and Motojima K: Hydrophobic statins induce autophagy and cell death in human rhabdomyosarcoma cells by depleting geranylgeranyl diphosphate. Eur J Pharmacol. 674:95–103. 2012. View Article : Google Scholar

120 

Liu H, Wang Z, Li Y, Li W and Chen Y: Simvastatin prevents proliferation and bone metastases of lung adenocarcinoma in vitro and in vivo. Neoplasma. 60:240–246. 2013. View Article : Google Scholar : PubMed/NCBI

121 

Liao J, Chung YT, Yang AL, et al: Atorvastatin inhibits pancreatic carcinogenesis and increases survival in LSL-KrasG12D-LSL-Trp53R172H-Pdx1-Cre mice. Mol Carcinog. 52:739–750. 2013. View Article : Google Scholar

122 

Vitols S, Angelin B and Juliusson G: Simvastatin impairs mitogen-induced proliferation of malignant B-lymphocytes from humans - in vitro and in vivo studies. Lipids. 32:255–262. 1997. View Article : Google Scholar : PubMed/NCBI

123 

Higgins MJ, Prowell TM, Blackford AL, et al: A short-term biomarker modulation study of simvastatin in women at increased risk of a new breast cancer. Breast Cancer Res Treat. 131:915–924. 2012. View Article : Google Scholar

124 

Bansal D, Undela K, D’Cruz S and Schifano F: Statin use and risk of prostate cancer: a meta-analysis of observational studies. PLoS One. 7:e466912012. View Article : Google Scholar : PubMed/NCBI

125 

Park HS, Schoenfeld JD, Mailhot RB, et al: Statins and prostate cancer recurrence following radical prostatectomy or radiotherapy: a systematic review and meta-analysis. Ann Oncol. 24:1427–1434. 2013. View Article : Google Scholar : PubMed/NCBI

126 

Singh PP and Singh S: Statins are associated with reduced risk of gastric cancer: a systematic review and meta-analysis. Ann Oncol. 24:1721–1730. 2013. View Article : Google Scholar : PubMed/NCBI

127 

van der Spek E, Bloem AC, Sinnige HA and Lokhorst HM: High dose simvastatin does not reverse resistance to vincris-tine, adriamycin, and dexamethasone (VAD) in myeloma. Haematologica. 92:e130–e131. 2007. View Article : Google Scholar : PubMed/NCBI

128 

Han JY, Lim KY, Yu SY, Yun T, Kim HT and Lee JS: A phase 2 study of irinotecan, cisplatin, and simvastatin for untreated extensive-disease small cell lung cancer. Cancer. 117:2178–2185. 2011. View Article : Google Scholar : PubMed/NCBI

129 

Kim WS, Kim MM, Choi HJ, et al: Phase II study of high-dose lovastatin in patients with advanced gastric adenocarcinoma. Invest New Drugs. 19:81–83. 2001. View Article : Google Scholar : PubMed/NCBI

130 

Lazzeroni M, Guerrieri-Gonzaga A, Serrano D, et al: Breast ductal lavage for biomarker assessment in high risk women: rationale, design and methodology of a randomized phase II clinical trial with nimesulide, simvastatin and placebo. BMC Cancer. 12:5752012. View Article : Google Scholar : PubMed/NCBI

131 

Kawata S, Yamasaki E, Nagase T, et al: Effect of pravastatin on survival in patients with advanced hepatocellular carcinoma. A randomized controlled trial. Br J Cancer. 84:886–891. 2001. View Article : Google Scholar : PubMed/NCBI

132 

Lersch C, Schmelz R, Erdmann J, et al: Treatment of HCC with pravastatin, octreotide, or gemcitabine - a critical evaluation. Hepatogastroenterology. 51:1099–1103. 2004.PubMed/NCBI

133 

Knox JJ, Siu LL, Chen E, et al: A Phase I trial of prolonged administration of lovastatin in patients with recurrent or metastatic squamous cell carcinoma of the head and neck or of the cervix. Eur J Cancer. 41:523–530. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2015
Volume 33 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Altwairgi AK: Statins are potential anticancerous agents (Review). Oncol Rep 33: 1019-1039, 2015
APA
Altwairgi, A.K. (2015). Statins are potential anticancerous agents (Review). Oncology Reports, 33, 1019-1039. https://doi.org/10.3892/or.2015.3741
MLA
Altwairgi, A. K."Statins are potential anticancerous agents (Review)". Oncology Reports 33.3 (2015): 1019-1039.
Chicago
Altwairgi, A. K."Statins are potential anticancerous agents (Review)". Oncology Reports 33, no. 3 (2015): 1019-1039. https://doi.org/10.3892/or.2015.3741