Next Article in Journal
The Case of Medication-Related Osteonecrosis of the Jaw Addressed from a Pathogenic Point of View. Innovative Therapeutic Strategies: Focus on the Most Recent Discoveries on Oral Mesenchymal Stem Cell-Derived Exosomes
Next Article in Special Issue
Sex-Differences in Discontinuation of Statin Treatment in Cancer Patients the Year before Death
Previous Article in Journal
Bowman-Birk Inhibitors: Insights into Family of Multifunctional Proteins and Peptides with Potential Therapeutical Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Statins: HMG-CoA Reductase Inhibitors as Potential Anticancer Agents against Malignant Neoplasms in Women

1
Department of Perinatology and Women’s Health, Poznań University of Medical Sciences, 60-535 Poznań, Poland
2
Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61–614 Poznań, Poland
3
Department of Oncology, Gynecological Oncology, Poznań University of Medical Sciences, 60-569 Poznań, Poland
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2020, 13(12), 422; https://0-doi-org.brum.beds.ac.uk/10.3390/ph13120422
Submission received: 1 October 2020 / Revised: 17 November 2020 / Accepted: 23 November 2020 / Published: 25 November 2020
(This article belongs to the Special Issue Statins Use and Cancer)

Abstract

:
Statins, also known as HMG-CoA inhibitors, are a class of bioactive small molecules that efficiently reduce the levels of cholesterol, and therefore are commonly used to manage and prevent various cardiovascular diseases. With respect to their original medical indications, statins are currently in the group of the most prescribed drugs worldwide. Of note is that statins are perceived actually rather as agents that have pleiotropic activities; in addition to their inhibitory activity on the production of endogenous cholesterol. Statins may also affect cell proliferation, angiogenesis and/or migration (metastasis) of different cancer cells, and play a positive role in the chemoprevention of cancer, thus being the excellent candidates to be repurposed in oncology. Particularly intriguing in this context seems to be the promising role of statins on both the incidence and course of common malignant neoplasms in women. In this article, we review and discuss the effect of the use of statins in the treatment of three types of cancer, i.e., breast, endometrial and ovarian cancer, with the highest mortality among gynecological cancers.

1. Introduction

Cardiovascular diseases (CVDs) are the main cause of death worldwide, responsible for more than 75% of all deaths in low- and middle-income countries. According to the World Health Organization, nearly 18 million people die every year because of the disorders of the heart as well as blood vessels, such as cerebrovascular, coronary heart and rheumatic heart diseases [1]. In the group of well-known factors that may initiate and further promote CVDs is a high plasma level of low density-lipoprotein (LDL) cholesterol [2]. With respect to the biosynthetic pathway of cholesterol, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase plays a pivotal role in the control of the rate limiting step in its production, and thus the inhibition of the activity of this crucial enzyme has been pinpointed as the prime target of effective cholesterol-lowering therapies [3]. In this context, the treatment strategies with the use of statins have been found to significantly reduce the elevated levels of atherogenic lipoproteins, primarily LDL cholesterol, mainly through the inhibition of HMG-CoA reductase [4]. Currently, six forms of statins are marketed and commonly used as the cholesterol-lowering drugs, leading finally to the reduction of both morbidity and mortality among patients with CVDs; in this group are atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, as well as simvastatin (Figure 1a). Several dual treatment strategies with the use of statins in the combination with other agents, like simvastatin/niacin extended-release, are also available [5].
Structurally, all statins have a common element (pharmacophore group) (Figure 1) that shows great similarity to the HMG-CoA molecule (Figure 2), thus being the excellent competitive inhibitor of HMG-CoA reductase [6]. With respect to the ‘side arms’ of most commercially available statins, they constitute partially hydrogenated naphthalene ring, with either the 2-methylbutyrate or 2,2-dimethylbutyrate moiety, and one or two methyl groups located on the opposite sites of the bicyclic ring system. On the other hand, the basis of the structure of synthetic statins is indole (fluvastatin), pyrimidine (rosuvastatin), pyrrole (atorvastatin) or quinoline (pitavastatin) aromatic ring substituted with various groups, including 4-fluorophenyl one, identical for all these structures. Nevertheless, as far as statins are concerned, the history of their isolation, synthesis and further development goes back nearly 50 years, and is briefly described in the next section.

2. Brief History of Statins

Before the discovery of the first statins, only a few lipid-lowering agents were known; they included cholestyramine, clofibrate and nicotinic acid [7]. However, as the activity of these compounds was rather moderate, development and introduction of more effective and less toxic inhibitors of cholesterogenesis was of top interest. In 1976, Endo and co-workers discovered compactin, renamed mevastatin (Figure 1b), a multifunctional fungal metabolite produced by Penicillium citrinum [8]; it was also later isolated from cultures of Penicillium brevicompactum [9]. After a series of subsequent animal experiments, this naturally-occurring compound has been shown to exhibit potent competitive inhibitory effects on the activity of HMG-CoA reductase and biosynthesis of cholesterol [10,11,12,13,14,15,16]. As mevastatin has been demonstrated to show therapeutic effects in humans by reducing the plasma levels of LDL cholesterol [17,18], but also some adverse side incidences, like lymphomas in dogs when using it in particularly high doses [7,19], intensive efforts have been made to identify other agents from this group of small bioactive molecules.
As a result of a screening program for HMG-CoA reductase inhibitors, lovastatin, known also as mevinolin (Figure 1b), was isolated from a mold Aspergillus terreus in the late 1970s [20], and then found to be more effective than mevastatin when tested in animal models; for review see [21]. Of note, a series of clinical trials have proven the ability of this compound to reduce the levels of both total and LDL cholesterol in patients with severe hypercholesterolemia [22,23,24,25,26,27,28]. The promising hypocholesterolemic effects of lovastatin led finally to its introduction to clinical therapy in 1987 as the first statin in history [3]. Since a natural lovastatin had been commercialized, six semi-synthetic and synthetic statins have been introduced to the markets in the last 30 years [7]. Among them were pravastatin and simvastatin (Figure 1a), the analogs of mevastatin and lovastatin, respectively, which have been found to effectively decrease the lipid profiles in a series of large and well-controlled clinical trials [29,30,31,32,33]. On the other hand, four synthetic variants of statins, fluvastatin, atorvastatin, rosuvastatin and pitavastatin (Figure 1a), were approved for routine medical practice in 1993, 1996, 2003 and 2009, respectively, thanks to which millions of patients around the world using these pharmaceuticals for primary prevention may have avoided heart attacks altogether [34,35].
Statistically, in 2017, atorvastatin became the second most prescribed pharmaceutical in the United States, with more than 104 million prescriptions (Table 1) [36]. Although the total number of prescriptions of simvastatin has been systematically falling since 2011 (~110 million in 2011 versus ~60 million in 2017), the sale of pravastatin, rosuvastatin and lovastatin remains almost at the same level up to now (Figure 3). Of note, it was estimated that more than one billion people globally would qualify to be prescribed a drug from the statins class [37].
In addition to the original medical indications of statins, some interesting studies have suggested recently that selected compounds from this group may also play a positive role in cancer prevention [38,39], thus they can be considered as promising repurposable oncological drug candidates. Drug repurposing is a relatively novel strategy for the identification of possible alternative uses of well-known medications which are outside the scope of their original therapeutic indications. Importantly, such a strategy could entail a number of advantages compared to standard de novo drug development, such as less time-consuming and reduced costs of introduction of a drug to the market [40,41].
In this context, in the following paragraphs, the effects of statins on three malignant cancers occurring in women are discussed; these are the breast, endometrial and ovarian cancer, with the highest mortality among gynecological types of neoplasm. Although the results seem to be slightly ambiguous, the positive role of statins on both the incidence and course of these selected types of cancer, as well as their possible chemopreventive action is worth the attention.

3. Anticancer Activity of Statins

3.1. Breast Cancer

Breast cancer (BC) is the most common malignant neoplasm in women. In 2018, more than two million women were diagnosed with this type of cancer, which caused about 630,000 deaths; it accounts for 24% of all cases and 15% of total deaths from all malignant neoplasms in women, respectively [42]. The majority of cases occur in women aged 50–70, and the risk factors for the development of this cancer include, but are not limited to, mutations in genes (breast cancer 1 and 2, BRCA1/2) and the influence of hormones (contraception, hormone replacement therapy). Approximately 10–20% of BCs are a heterogeneous group of triple negative BCs (TNBCs), with an earlier age of development, aggressive course of the disease, lack of progesterone and estrogen receptors (ER), and expression for human epidermal growth factor receptor 2 (HER2) [43]. Importantly, the risk of BC development in postmenopausal women is associated with excess weight and obesity, which causes i.a. changes in circulating cholesterol levels and pro-inflammatory cytokine levels. Statins are frequently prescribed as part of women’s long-term treatment programs. There is a need therefore, for further research into the effects of statins from the perspective of cancer prevention and treatment [44,45].
The studies by Islam and co-workers [46], based on electronic databases of 121,399 patients, did not show a clear relationship between statin use and the risk of BC development (RR = 0.94, 95% CI 0.86–1.03). Similar results showing that statin treatment does not influence the progress of BC were obtained by the researchers of the Nurses’ Health Study (NHS) [47]. Very briefly, more than 79,518 postmenopausal women participated in the survey based on questionnaires (confirmed by a physician). The study covered the period from 2000 to 2012, and 3055 women with invasive BC were qualified for the analysis, of which over 1900 individuals had never used the statin-based therapy. Ductal carcinoma was diagnosed in most of the patients (1952 cases), in some lobular carcinoma (313 cases), while in the rest of the participants the carcinoma in situ. In addition, some cases of ER, but also estrogen receptor positive (ER+) carcinomas were determined. Of note, statin use was neither associated with the risk of infiltrating lobular carcinoma, ductal cancer nor estrogen receptor status (HR = 0.96, 95% CI 0.82–1.1 and HR = 1.1, 95% CI 0.92–1.3, respectively). According to the authors of this article, future analyzes need to take into account the type of statins used and the specific histological subtype of BC.
Metabolic syndrome occurs in some women with BC, and in this context the effect of the antidiabetic metformin and statins is frequently analyzed. According to data from the Finnish national diabetes database, 2300 women were diagnosed with BC, and then were treated with metformin, insulin and statins. Case-control analysis clearly showed that there was no difference in cancer incidence between the metformin group (HR = 1.02, 95% CI 0.93–1.11) and the statin group (HR = 0.97, 95% CI 0.89–1.05), while insulin treatment was associated with slightly increased morbidity (HR = 1.18, 95% CI 1.03–1.36) [48].
Furthermore, a study by Yao and co-workers [49] suggested that lipophilic statins, particularly lovastatin (Figure 1b), preferentially target TNBCs compared to other types of BC. The authors believe that the future therapy of TNBCs with statins should take into account the role of these compounds in cancer cells with stem-like phenotype (cancer stem cells, CSCs), develop the possible use of nanoparticles to encapsulate statins, and identify the molecular targets and mechanisms underlying such promising effects. The results of a study by the Danish Breast Cancer Group covering the period 2007–2017, with participation of over 14,000 women, also demonstrated the beneficial effects of the statin-based treatment option. In a multivariate analysis, statin treatment reduced the recurrence rate among women with stage I–III BC with the presence of estrogen receptors treated with aromatase inhibitors (HR = 0.72, 95% CI 0.50–1.04), suggesting that statins may become an additional therapeutic regimen in future [50]. As BC patients are exposed to cardiotoxic therapies—such as anthracyclines and trastuzumab—statins could be included in the BC treatment process for their cardioprotective effect [51,52].
Finally, the use of statins reducing death in BC patients has been shown to be strictly associated with the reduction of metastases; Beckwitt and co-workers [53] observed in their studies the inhibitory effect of atorvastatin (Figure 1a) on the proliferation of metastatic foci on BC cell lines that metastasize to the liver and lungs. Clinical observations indicate that both BC metastases and relapses are a sign of a poor course of the disease, and in this context some recent studies suggest that lipophilic statins can prevent recurrence of BC [54]. However, it is most likely not associated with variants of genes encoding drug transporters and simvastatin (Figure 1a) metabolizing enzymes (CYP enzyme group) [55].

3.2. Endometrial Cancer

On the basis of the world epidemiological statistics, endometrial cancer (EC) is a common malignant neoplasm in women. It ranks 6th as the cause of death after BC, colorectal cancer, lung cancer, cervical cancer and thyroid cancer. In 2018, over 380,000 new cases of EC were diagnosed, which accounted for 4.4% of all cancer cases in women [42]. There are two known types of EC: (i) type I EC, histologically endometrioid, which accounts for about 80% of all ECs, has a good prognosis, and is associated with well-known risk factors, like metabolic syndrome (diabetes, hypertension, obesity), mutations in the mismatch repair (MMR), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA), Kirsten rat sarcoma viral oncogene homolog (KRAS), β1-catenin (CTNNB1) genes, and microsatellite instability (MSI), as well as (ii) non-endometrioid, less common type II EC, which has aggressive clinical course, and is often characterized by mutations in tumor protein p53 (TP53), HER2/neu and BRCA [56,57]. Although genomic analysis divides EC into four subgroups, there is an association of some EC subtypes with body weight (body mass index, BMI), which clearly suggests a modelling effect of obesity on the genetic types of EC malignancy [58,59].
Numerous studies have shown hyperglycemia and type 2 diabetes increase the risk of developing EC, especially EC type I. Likewise, research indicates a reduction in morbidity, as well as a more favorable clinical course as a result of the application of drugs, such as metformin, which reduce these factors [58,59,60]. However, the results of the studies on the use of statins in EC remain inconclusive [61,62,63,64,65], and the observational studies often compare the effects of two drugs, i.e., antidiabetic metformin and a selected compound from the statins class [48,49].
The studies by Kim and co-workers [61] on established three cell lines, including the Ishikawa line, proved that both metformin and simvastatin (Figure 1a) used either separately or together in the experiments inhibit tumor growth and metastasis. These therapeutics mediated cell apoptosis, as determined by executive caspase 3 levels as well as Bax, Bcl-2 and Bim markers, but also inhibited the mammalian target of rapamycin (mTOR) signaling pathway strictly involved in cell proliferation, angiogenesis, and protein synthesis. On the other hand, a retrospective analysis of the Finnish Diabetes Registry, including 92,366 women with newly diagnosed type 2 diabetes and 590 with EC type I, has shown that statin treatment was inversely related to the frequency of EC in these individuals (HR = 0.78, 95% CI 0.65–0.94). Nevertheless, metformin and other antidiabetics had an unfavorable effect, increasing the incidence of EC compared to the group of women not treated for diabetes (HR = 1.24, 95% CI 1.02–1.51 and HR = 1.25, 95% CI 1.04–1.50, respectively) [64].
Interestingly, Yang and co-workers [65] presented a meta-analysis of 9517 women with EC diagnosed between 2001 and 2016 on different continents (North America, Europe and Asia) and treated with statins. These analyses, also based on electronic databases, included two randomized and 11 non-randomized studies (four cohorts and seven case-control studies), with a median duration of 5.2 years. According to the authors of this article, statin use was found to reduce the risk of developing EC only in the group of women from Asia (437 cases) (RR = 0.52, 95% CI 0.37–0.74). It is worth noting that, in this study, the use of cardiovascular drugs, such as aspirin, may have been a confounding factor, as noted by the researchers analysing the data.
Also on the basis of electronic databases, Liu and co-workers [66] analysed the risk of developing gynecological cancers in women using statins, including 10 studies with a total of 9957 patients with EC. Although it was suggested that statin treatment did not significantly reduce the risk of developing EC (RR = 0.90, 95% CI 0.75–1.07), the risk decreased (RR = 0.69, 95% CI 0.44–1.10) if statins were used for more than 5 years. Intriguingly, as previously observed by Yang and co-workers [65], Asian women using statins once again were at lower risk of developing EC than individuals from America or Europe (RR = 0.46, 95% CI 0.28–074) [66]. Contrary to these results, the PRISMA meta-analysis, based on the results of electronic databases of 19 studies that included 199,362 women and 19,849 cancer cases, including 11,901 EC cases, suggested not only that the use of statins reduced the risk of developing EC (RR = 0.88, 95% CI 0.78–1.00), but also revealed no association between long-term statin use (5 years) and a reduction in the risk of developing this type of cancer [62]. On the basis of the Danish Cancer Registry, Sperling and co-workers [67] analysed EC mortality in 6694 women with EC aged 38–84, using statins; type I EC was found in 5982 women (89% of all ECs) and type II in 712 women (11%). Of note is that post-diagnosis statin use was found to be associated with lower mortality (reduced by even 39%) among patients compared to non-statin users (HR = 0.61, 95% CI 0.48–0.77). This relationship was similar for EC I and II type (HR = 0.59, 95% CI 0.45–0.77 and HR = 0.65, 95% CI 0.43–0.99, respectively). The mortality rate in women using statins before diagnosis was also lower, but not as pronounced as in those who used them post diagnosis.
Slightly different results have been presented by Li and co-workers [63]. Using electronic databases, they analysed the data of 5923 EC women from Austria, Denmark, Israel, UK and USA. The use of statins prolonged both overall survival and disease specific survival (HR = 0.80, 95% CI 0.66–0.95 and HR = 0.69, 95% CI 0.61–0.79, respectively). Moreover, the authors of this article were the first to find a dependence of disease specific survival prolongation in type II EC (HR = 0.64, 95% CI 0.50–0.81), which is very aggressive and has a poor prognosis due to relapses even in the early clinical stages. On the other hand, a group of Israeli researchers analysed the cases of women in 11 hospital centres from 2002–2014, with a mean follow-up of 6.2 years. Surprisingly, the 5-year relapse-free survival did not differ between the group of women with EC who had used statins before diagnosis (633 women, 32.8% of all respondents) and those who had not used them at all (1354 women, 67.1%). The 5-year relapse-free survival was 82% and 83% (p = 0.508), respectively, and the overall survival was 77% and 75% (p = 0.901), respectively [68]. The presented results regarding the impact of statin treatment on the incidence of EC and the course of this cancer seems thus far rather inconclusive. The proportion of presented results suggesting a beneficial effect of statins on the risk of developing EC and its course seems a promising development.

3.3. Ovarian Cancer

Ovarian cancer (OC) ranks as the 8th cause of death in women with all malignant neoplasms. In 2018, 295,414 women worldwide were diagnosed with OC, which accounts for 3.4% of all cancer cases [42]. Of note, this heterogeneous group of cancers (genetically determined and sporadic) is the leading cause of mortality among all gynecological cancers, with high-grade serous OC diagnosed in high clinical stages, often with relapses, which displays progressive resistance to platinum derivatives, and thus has a poor prognosis [69].
Similar to BC and EC, the results of the studies on the influence of statins on the development of OC are also inconsistent. In the aforementioned PRISMA meta-analysis, Wang and co-workers [62] presented the results of 10 studies aimed at assessment of the influence of statins on the development of OC. On the basis of the calculations, it was shown that among 7948 cases of OC in women from North America or Europe, statin use did not significantly reduce the risk of developing this type of cancer (RR = 0.88, 95% CI 0.76–1.03). There was also no association between long-term statin use (>5 years) and the risk of developing OC (RR = 0.73, 95% CI 0.51–1.04). A similar lack of relationship between the statins treatment and development of OC was also shown by the Finnish Registry study in a group of women aged over 40 and diagnosed with type 2 diabetes. Among the cohort of 137,643 individuals studied in 1996–2011, 303 cases of OC were diagnosed. It was also the first study in women with type 2 diabetes treated with either metformin or insulin as well as statins, due to the increased risk of cardiovascular diseases and/or hypercholesterolemia. In a full cohort analysis, there was no evidence that metformin or insulin use was associated with a different incidence of OC compared to other oral antidiabetic agents. Furthermore, there was also no effect of statin use on the development of OC (HR = 0.99, 95% CI 0.78–1.25), and no interaction of statin treatment with metformin [70].
However, some other authors have reported a lower risk of developing OC in women using statins. Five studies have been based on the electronic database analyses for 624 women who had developed OC, from North America, Asia and Europe. Statin use was associated with a 21% reduction in the risk of developing this type of cancer without taking into account their heterogeneity (RR = 0.79, 95% CI 0.64–0.98). There was also no difference in the geographical subgroups of women (Asia or other continents) [66]. A systematic review and meta-analysis by Irvin and co-workers [71] in nine studies involving 435,237 women suggested that statin use was associated with a lower risk of developing OC. However, this risk depended on the OC histotype, the class of statins used and the duration of their use. The risk for OC without histotype was reduced (RR = 0.87, 95% CI 0.77–1.03), especially in women with low pravastatin (Figure 1a) treatment (RR = 0.83, 95% CI 0.70–0.99). The risk of developing serous carcinomas was reduced in relation to clear cell carcinomas (RR = 0.95, 95% CI 0.69–1.20 and RR = 1.17, 95% CI 0.54–1.10, respectively). According to the authors of this article, such dependencies on the cancer histotype, type of statins and their dose require further clarification.
The differences in the incidence of OC associated with the use of statins are partly explained by genetic studies on the inhibition of HMG-CoA reductase, which is affected by statins which lower the synthesis of endogenous cholesterol [72]. The GWAS (genome association study) meta-analyses from the Ovarian Cancer Association Consortium, involving more than 22,000 patients with invasive OC, as well as retrospective cohort studies of the Consortium of Investigators of Modifiers performed among BRCA1/2 mutation carriers, involving more than 3,880 women, have shown a clear relationship between LDL cholesterol resulting from inhibition of HMG-CoA reductase and reduction in the risk of developing OC (OR = 60, 95% CI 0.43–0.83). Importantly, in BRCA1/2 mutation carriers, the risk of developing OC was also reduced (RR = 0.69, 95% CI 0.51–0.93). On the other hand, the explanation of the beneficial effects of statins on inhibiting the development of OC is based, according to Liu and co-workers [73], on the activation of the c-Jun N-terminal kinase (JNK) signaling pathway and the reduction of the proapoptotic Bim protein. According to the authors of this article, such a molecular mechanism of action of statins inducing apoptosis in OC cells could lead to novel therapies for advanced OC.
The effect of statin treatment on OC survival has also been assessed in several studies. In a meta-analysis based on an electronic database of 12 studies, an improvement in overall survival was found in women using statins (HR = 0.76, 95% CI 0.68–0.85). This study was a part of a larger meta-analysis (36 studies) that included a group of women who were also treated with other drugs, such as metformin, β-blockers, aspirin or non-steroidal anti-inflammatory drugs (NSAIDs); neither drug has shown an overall survival benefit [74]. Jeong and co-workers [75] have reported results of a study (16 articles) of the influence of statins on survival and mortality of patients treated for various malignant neoplasms, including OC. From among the three meta-analyses of OC, one suggested a beneficial effect in reducing cancer-specific mortality (RR = 0.74, 95% CI 0.63–0.87). Favorable results concerning survival in OC following from the data for more than 2,100 patients with this type of cancer have been also documented by Harding and co-workers [76]. In the group of over 2,100 women with OC (≥66 years of age), statin use was associated with a lower risk of death from cancer (RR = 0.74, 95% CI 0.60–0.91). Similarly, Couttenier and co-workers [77] in a retrospective study of the data from the Belgian Cancer Registry for over 5400 OC patients found that the use of statins, especially simvastatin and rosuvastatin (Figure 1a), after cancer diagnosis prolonged survival (HR = 0.86, 95% CI 0.74–0.99 and HR = 0.71, 95% CI 0.55–0.92, respectively).
The positive effect of statins used in OC patients may be explained by the fact that serous OC with the highest malignancy exhibit specific oxidative metabolism responsible for the inflammatory response and drug resistance. Research by Criscuolo and co-workers [78] revealed that platinum-resistant OC cells show reduced biosynthesis of cholesterol, mainly due to decreased activity of the enzymes involved in this process, and they capture exogenous cholesterol for their needs. With regard to this metabolism, a dual mechanism of statins action has been demonstrated – on the one hand, when used to inhibit cholesterol biosynthesis, they reduce cisplatin-induced apoptosis, but after silencing the enzyme (lipase G, LIPG) involved in lipid metabolism as well as removing lipids from the culture medium, they increase the sensitivity to platinum drugs. These results raise concerns about the use of statins in OC patients and point to the necessity of taking into account lipid metabolism in the treatment.
In connection with the above described results, an article trying to respond to the question "How much of it is true?" has been published [79]. The author of this paper hypothesized that statin therapy could improve the survival of women with OC. The mechanism of this action in OC is based on inhibitory interaction with the signal transducer and activator of transcription 3 (STAT3) signaling pathway, which is activated by the cytokine interleukin 6 (IL-6); IL-6 can be induced in response to pro-inflammatory interleukin 1β (IL-1β), whose elevated levels are seen in OC. Thus, statins not only lower cholesterol, but also suppress inflammation. Further research is however necessary.

4. Conclusions

Statins, HMG-CoA reductase inhibitors commonly used as cholesterol-lowering therapeutics, represent one of the most commonly prescribed classes of drugs worldwide. Six statins are currently marketed, i.e., atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, as well as simvastatin (Figure 1a). Interestingly, some recent findings also suggest the positive role of statin-based regimens in reducing the incidences of non-cardiac mortality, particularly those linked with cancer.
The beneficial effect of statin use on the incidence and course of common malignant neoplasms in various localisations, histological subtypes, advancement in women is ambiguous. This is possibly due to the use of different types of statins and/or heterogeneous populations, dosage and duration of treatment. While some meta-analyses from electronic databases did not show the effect of statins on reducing the incidence of breast cancer, a series of other studies strongly suggested the positive impact of the use of these bioactive small molecules on both reduction in the recurrence rate and in the risk of death from breast cancer; similar results were observed for endometrial and ovarian cancer patients. The studies performed using established cancer cell lines were very promising; statins inhibited i.a. the mTOR signaling pathway strictly associated with cancer progression, and pro-inflammatory cytokine activity.
To sum up, in our opinion, statins should only be considered as an adjuvant therapeutic option in cancer patients with the internal medicine indications, such as hypercholesterolemia, metabolic syndrome or the prevention of circulatory disturbances in the cardiovascular and cerebral systems. Moreover, their use seems to be beneficial due to the cardioprotective effects in cancer patients treated with cardiotoxic therapies. Nevertheless, future studies, including large and well-controlled clinical trials, considering i.a. the type of statin used (hydrophilic or lipophilic), time of statin use (prediagnosis or postdiagnosis) as well as duration of treatment, are vital to validate correctly the promising effects of the use of statins in women with selected malignant neoplasms.

Author Contributions

Writing—original draft preparation, A.M. and M.A.; writing—review and editing, A.H. and J.M.; supervision, A.H.; funding acquisition, A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

M.A. wishes to acknowledge the Polish Science Center (NCN) and the Polish National Agency for Academic Exchange (NAWA) for scholarships under the UWERTURA (2019/32/U/ST4/00092) and the BEKKER program (PPN/BEK/2019/1/00034), respectively, and the Polish Ministry of Science and Higher Education (MNiSW) for a scholarship for outstanding young scientists for the period 2020–2023 (STYP/15/1665/E-336/2020).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization—Cardiovascular Diseases. Available online: https://www.who.int/health-topics/cardiovascular-diseases (accessed on 19 September 2020).
  2. Nissen, S.E.; Tuzcu, E.M.; Schoenhagen, P.; Crowe, T.; Sasiela, W.J.; Tsai, J.; Orazem, J.; Magorien, R.D.; O’Shaughnessy, C.; Ganz, P. Statin therapy, LDL cholesterol, C-reactive protein, and coronary artery disease. N. Engl. J. Med. 2005, 352, 29–38. [Google Scholar] [CrossRef]
  3. Mishra, V.; Mehta, K.D. History and biochemistry of statins. In Statins. Understanding Clinical Use; Mehta, J.L., Ed.; W.B. Saunders: Philadelphia, PA, USA, 2004; pp. 1–12. [Google Scholar]
  4. Hu, M.; Cheung, B.M.Y.; Tomlinson, B. Safety of statins: An update. Ther. Adv. Drug Saf. 2012, 3, 133–144. [Google Scholar] [CrossRef] [Green Version]
  5. U.S. Food and Drug Administration—Statins. Available online: https://www.fda.gov/drugs/information-drug-class/statins (accessed on 19 September 2020).
  6. Jiang, S.Y.; Li, H.; Tang, J.J.; Wang, J.; Luo, J.; Liu, B.; Wang, J.K.; Shi, X.J.; Cui, H.W.; Tang, J.; et al. Discovery of a potent HMG-CoA reductase degrader that eliminates statin-induced reductase accumulation and lowers cholesterol. Nat. Commun. 2018, 9, 5138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Endo, A. A historical perspective on the discovery of statins. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2010, 86, 484–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Endo, A.; Kuroda, M.; Tsujita, Y. ML-236A, ML-236B, and ML-236C: New inhibitors of cholesterogenesis produced by Penicillium citrinum. J. Antibiot. (Tokyo) 1976, 29, 1346–1348. [Google Scholar] [CrossRef] [Green Version]
  9. Brown, A.G.; Smale, T.C.; King, T.J.; Hasenkamp, R.; Thompson, R.H. Crystal and molecular structure of compactin, a new antifungal metabolite from Penicillium brevicompactum. J. Chem. Soc. Perkin Trans. 1 1976, 11, 1165–1170. [Google Scholar] [CrossRef]
  10. Tanzawa, K.; Endo, A. Kinetic analysis of the reaction catalyzed by 3-hydroxy-3-methylglutaryl coenzyme A reductase using two specific inhibitors. Eur. J. Biochem. 1979, 98, 195–201. [Google Scholar] [CrossRef]
  11. Tsujita, Y.; Kuroda, M.; Tanzawa, K.; Kitano, N.; Endo, A. Hypolipidemic effects in dogs of ML-236B, a competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase. Lipids 1979, 14, 585–589. [Google Scholar] [CrossRef]
  12. Kanecko, I.; Hazama-Shimada, Y.; Endo, A. Effects of ML-236B, a competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase, on the lipid metabolism in culture cells. Eur. J. Biochem. 1978, 87, 313–321. [Google Scholar] [CrossRef]
  13. Doi, O.; Endo, A. Specific inhibition of desmosterol synthesis by ML-236B in mouse LM cells grown in suspension in a liquid-free medium. Jpn. J. Med. Sci. Biol. 1978, 31, 225–233. [Google Scholar] [CrossRef] [Green Version]
  14. Fears, R.; Richards, D.H.; Ferres, H. The effect of compactin, a potent inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity, on cholesterogenesis and serum cholesterol levels in rats and chicks. Atherosclerosis 1978, 35, 439–449. [Google Scholar] [CrossRef]
  15. Endo, A.; Tsujita, Y.; Kuroda, M.; Tanazawa, K. Inhibition of cholesterol synthesis in vitro and in vivo by ML-236A and ML-236B, competitive inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase. Eur. J. Biochem. 1977, 77, 31–36. [Google Scholar] [CrossRef] [PubMed]
  16. Endo, A.; Kuroda, M.; Tanzawa, K. Competitive inhibition of HMG CoA reductase by ML-236A and ML-236B fungal metabolites, having hypercholesterolemic activity. FEBS Lett. 1976, 72, 323–326. [Google Scholar] [CrossRef] [Green Version]
  17. Mabuchi, H.; Haba, T.; Tatami, R.; Miyamoto, S.; Sakai, Y.; Wakasugi, T.; Watanabe, A.; Koizumi, J.; Takeda, R. Effects of an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase on serum lipoproteins and ubiquinone-10 levels in patients with familial hypercholesterolemia. N. Engl. J. Med. 1981, 305, 478–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Yamamota, A.; Sudo, H.; Endo, A. Therapeutic effects of ML-236B in primary hypercholesterolemia. Atherosclerosis 1980, 35, 259–266. [Google Scholar] [CrossRef]
  19. Lyons, K.S.; Harbinson, M. Statins: In the beginning. J. R. Coll. Physicians Edinb. 2009, 39, 362–364. [Google Scholar] [CrossRef]
  20. Alberts, A.W.; Chen, J.; Kuron, G.; Hunt, V.; Huff, J.; Hoffman, C.; Rothrock, J.; Lopez, M.; Joshua, H.; Harris, E.; et al. Springer, J. Mevinolin: A highly potent competitive inhibitor of hydroxymethylglutaryl-coenzyme A reductase and a cholesterol-lowering agent. Proc. Natl. Acad. Sci. USA 1980, 77, 3957–3961. [Google Scholar] [CrossRef] [Green Version]
  21. Alberts, A.W. Discovery, biochemistry and biology of lovastatin. Am. J. Cardiol. 1988, 62, 10J–15J. [Google Scholar] [CrossRef]
  22. Vega, G.L.; East, C.A.; Grundy, S.M. Lovastatin therapy in familial dysbetalipoproteinemia: Effect on kinetics of apoprotein B. Atherosclerosis 1988, 70, 131–143. [Google Scholar] [CrossRef]
  23. Garg, A.; Grundy, S.M. Lovastatin for lowering cholesterol levels in noninsulin-dependent diabetes mellitus. N. Engl. J. Med. 1988, 314, 81–86. [Google Scholar] [CrossRef]
  24. Havel, R.J.; Hunninghake, D.B.; Illingworth, D.R.; Lees, R.S.; Stein, E.A.; Tobert, J.A.; Bacon, S.R.; Bolognese, J.A.; Frost, P.H.; Lamkin, G.E. Lovastatin (mevinolin) in the treatment of heterozygous familial hypercholesterolemia: A multicenter study. Ann. Intern. Med. 1987, 107, 609–615. [Google Scholar] [CrossRef] [PubMed]
  25. The Lovastatin Study Group II. Therapeutic response to lovastatin (mevinolin) in nonfamilial hypercholesterolemia: A multicenter study. JAMA 1986, 256, 2829–2834. [Google Scholar]
  26. East, C.A.; Grundy, S.M.; Bilheimer, D.W. Preliminary report: Treatment of type 3 hyperlipoproteinemia with mevinolin. Metabolism 1986, 35, 97–98. [Google Scholar] [CrossRef]
  27. Vega, G.L.; Grundy, S.M. Lovastatin therapy in nephrotic hyperlipidemia: Effects on lipoprotein metabolism. Kidney Int. 1985, 33, 339–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Illingworth, D.R.; Sexton, G.J. Hypocholesterolemic effects of mevinolin in patients with heterozygous familial hypercholesterolemia. J. Clin. Investig. 1984, 74, 1972–1978. [Google Scholar] [CrossRef]
  29. Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group. Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. N. Engl. J. Med. 1998, 339, 1349–1357. [Google Scholar] [CrossRef] [Green Version]
  30. Downs, J.R.; Clearfield, M.; Weis, S.; Whitney, E.; Shapiro, D.R.; Beere, P.A.; Langendorfer, A.; Stein, E.A.; Kruyer, W.; Gotto, A.M., Jr. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: Results of AFCAPS/TexCAPS. Air Force/Texas Coronary Atherosclerosis Prevention Study. JAMA 1998, 279, 1615–1622. [Google Scholar] [CrossRef]
  31. Sacks, F.M.; Pfeffer, M.A.; Moye, L.A.; Rouleau, J.L.; Rutherford, J.D.; Cole, T.D.; Brown, L.; Warnica, J.W.; Arnold, J.M.; Wun, C.C.; et al. The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. N. Engl. J. Med. 1996, 335, 1001–1009. [Google Scholar] [CrossRef]
  32. Shepherd, J.; Cobbe, S.M.; Ford, I.; Isles, C.G.; Ross Lorimer, A.; Macfarlane, P.W.; McKillop, J.H.; Packard, C.J. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. N. Engl. J. Med. 1995, 333, 1301–1307. [Google Scholar] [CrossRef]
  33. Scandinavian Simvastatin Survival Study Group. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: The Scandinavian Simvastatin Survival Study (4S). Lancet 1994, 344, 1383–1389. [Google Scholar]
  34. Stossel, T.P. The discovery of statins. Cell 2008, 134, 903–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Steinberg, D. The Cholesterol Wars: The Cholesterol Skeptics vs. the Preponderance of Evidence; Academic Press-Elsevier: San Diego, CA, USA, 2007. [Google Scholar]
  36. Atorvastatin—Drug Usage Statistics. Available online: https://clincalc.com/drugstats/drugs/atorvastatin (accessed on 19 September 2020).
  37. Ioannidis, J.A. More than a billion people taking statins? Potential implications of the new cardiovascular guidelines. JAMA 2014, 311, 463–464. [Google Scholar] [CrossRef] [PubMed]
  38. Hassanabad, A.F. Current perspectives on statins as potential anti-cancer therapeutics: Clinical outcomes and underlying molecular mechanisms. Transl. Lung Cancer Res. 2019, 8, 692–699. [Google Scholar] [CrossRef] [PubMed]
  39. Ciofu, C. The statins as anticancer agents. Maedica 2012, 7, 377. [Google Scholar] [PubMed]
  40. Antoszczak, M.; Markowska, A.; Markowska, J.; Huczyński, A. Antidepressants and antipsychotic agents as repurposable oncological drug candidates. Curr. Med. Chem. 2020. [Google Scholar] [CrossRef]
  41. Antoszczak, M.; Markowska, A.; Markowska, J.; Huczyński, A. Old wine in new bottles: Drug repurposing in oncology. Eur. J. Pharmacol. 2020, 866, 172784. [Google Scholar] [CrossRef]
  42. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  43. Kumar, P.; Aggarwal, R. An overview of triple-negative breast cancer. Arch. Gynecol. Obstet. 2016, 293, 247–269. [Google Scholar] [CrossRef]
  44. Kang, C.; LeRoith, D.; Gallagher, E.J. Diabetes, obesity, and breast cancer. Endocrinology 2018, 159, 3801–3812. [Google Scholar] [CrossRef] [Green Version]
  45. Li, Y.R.; Ro, V.; Tchou, J.C. Obesity, metabolic syndrome, and breast cancer: From prevention to intervention. Curr. Surg. Rep. 2018, 6, 7. [Google Scholar] [CrossRef]
  46. Islam, M.M.; Yang, H.C.; Nguyen, P.A.; Poly, T.N.; Huang, C.W.; Kekade, S.; Khalfan, A.M.; Debnath, T.; Li, Y.J.; Abdul, S.S. Exploring association between statin use and breast cancer risk: An updated meta-analysis. Arch. Gynecol. Obstet. 2017, 296, 1043–1053. [Google Scholar] [CrossRef] [PubMed]
  47. Borgquist, S.; Tamimi, R.M.; Chen, W.Y.; Garber, J.E.; Eliassen, A.H.; Ahern, T.P. Statin use and breast cancer risk in the Nurses’ Health Study. Cancer Epidemiol. Biomark. Prev. 2016, 25, 201–206. [Google Scholar] [CrossRef] [Green Version]
  48. Hosio, M.; Urpilainen, E.; Marttila, M.; Hautakoski, A.; Arffman, M.; Sund, R.; Puistola, U.; Läärä, E.; Jukkola, A.; Karihtala, P. Association of antidiabetic medication and statins with breast cancer incidence in women with type 2 diabetes. Breast Cancer Res. Treat. 2019, 175, 741–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Yao, H.; He, G.; Yan, S.; Chen, C.; Song, L.; Rosol, T.J.; Deng, X. Triple-negative breast cancer: Is there a treatment on the horizon? Oncotarget 2017, 8, 1913–1924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Harborg, S.; Heide-Jørgensen, U.; Ahern, T.P.; Ewertz, M.; Cronin-Fenton, D.; Borgquist, S. Statin use and breast cancer recurrence in postmenopausal women treated with adjuvant aromatase inhibitors: A Danish population-based cohort study. Breast Cancer Res. Treat. 2020, 183, 153–160. [Google Scholar] [CrossRef] [PubMed]
  51. Padegimas, A.; Clasen, S.; Ky, B. Cardioprotective strategies to prevent breast cancer therapy-induced cardiotoxicity. Trends Cardiovasc. Med. 2020, 30, 22–28. [Google Scholar] [CrossRef]
  52. Calvillo-Argüelles, O.; Abdel-Qadir, H.; Michalowska, M.; Billia, F.; Suntheralingam, S.; Amir, E.; Thavendiranathan, P. Cardioprotective effect of statins in patients with HER2-positive breast cancer receiving trastuzumab therapy. Can. J. Cardiol. 2019, 35, 153–159. [Google Scholar] [CrossRef]
  53. Beckwitt, C.H.; Clark, A.M.; Ma, B.; Whaley, D.; Oltvai, Z.N.; Wells, A. Statins attenuate outgrowth of breast cancer metastases. Br. J. Cancer 2018, 119, 1094–1105. [Google Scholar] [CrossRef] [Green Version]
  54. Manthravadi, S.; Shrestha, A.; Madhusudhana, S. Impact of statin use on cancer recurrence and mortality in breast cancer: A systematic review and meta-analysis. Int. J. Cancer 2016, 139, 1281–1288. [Google Scholar] [CrossRef]
  55. Ahern, T.P.; Damkier, P.; Feddersen, S.; Kjærsgaard, A.; Lash, T.L.; Hamilton-Dutoit, S.; Lythjohan, C.B.; Ejlertsen, B.; Christiansen, P.M.; Cronin-Fenton, D.P. Predictive pharmacogenetic biomarkers for breast cancer recurrence prevention by simvastatin. Acta Oncol. 2020, 59, 1009–1015. [Google Scholar] [CrossRef]
  56. Byrne, F.L.; Martin, A.R.; Kosasih, M.; Caruana, B.T.; Farrell, R. The role of hyperglycemia in endometrial cancer pathogenesis. Cancers 2020, 12, 1191. [Google Scholar] [CrossRef]
  57. Colombo, N.; Creutzberg, C.; Amant, F.; Bosse, T.; González-Martín, A.; Ledermann, J.; Marth, C.; Nout, R.; Querleu, D.; Mirza, M.R.; et al. ESMO-ESGO-ESTRO endometrial consensus conference Working Group. ESMO-ESGO-ESTRO consensus conference on endometrial cancer: Diagnosis, treatment and follow-up. Ann. Oncol. 2016, 27, 16–41. [Google Scholar] [CrossRef]
  58. Espinosa, I.; De Leo, A.; D’Angelo, E.; Rosa-Rosa, J.M.; Corominas, M.; Gonzalez, A.; Palacios, J.; Prat, J. Dedifferentiated endometrial carcinomas with neuroendocrine features: A clinicopathologic, immunohistochemical, and molecular genetic study. Hum. Pathol. 2018, 72, 100–106. [Google Scholar] [CrossRef]
  59. Roque, D.R.; Makowski, L.; Chen, T.H.; Rashid, N.; Hayes, D.N.; Bae-Jump, V. Association between differential gene expression and body mass index among endometrial cancers from The Cancer Genome Atlas Project. Gynecol. Oncol. 2016, 142, 317–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Meireles, C.G.; Pereira, S.A.; Valadares, L.P.; Rêgo, D.F.; Simeoni, L.A.; Guerra, E.N.S.; Lofrano-Porto, A. Effects of metformin on endometrial cancer: Systematic review and meta-analysis. Gynecol. Oncol. 2017, 147, 167–180. [Google Scholar] [CrossRef] [PubMed]
  61. Kim, J.S.; Turbov, J.; Rosales, R.; Thaete, L.G.; Rodriguez, G.C. Combination simvastatin and metformin synergistically inhibits endometrial cancer cell growth. Gynecol. Oncol. 2019, 154, 432–440. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, Y.; Ren, F.; Song, Z.; Chen, P.; Liu, S.; Ouyang, L. Statin use and the risk of ovarian and endometrial cancers: A meta-analysis. BMC Cancer 2019, 19, 730. [Google Scholar] [CrossRef] [Green Version]
  63. Li, J.; Liu, R.; Sun, Z.; Tang, S.; Wang, L.; Liu, C.; Zhao, W.; Yao, Y.; Sun, C. The association between statin use and endometrial cancer survival outcome: A meta-analysis. Medicine (Baltimore) 2018, 97, e13264. [Google Scholar] [CrossRef]
  64. Arima, R.; Marttila, M.; Hautakoski, A.; Arffman, M.; Sund, R.; Ilanne-Parikka, P.; Kangaskokko, J.; Läärä, E.; Puistola, U.; Hinkula, M. Antidiabetic medication, statins and the risk of endometrioid endometrial cancer in patients with type 2 diabetes. Gynecol. Oncol. 2017, 146, 636–641. [Google Scholar] [CrossRef] [Green Version]
  65. Yang, J.; Zhu, Q.; Liu, Q.; Wang, Y.; Xie, W.; Hu, L. Statin use and endometrial cancer risk: A meta-analysis. Oncotarget 2017, 8, 62425–62434. [Google Scholar] [CrossRef]
  66. Liu, Y.; Qin, A.; Li, T.; Qin, X.; Li, S. Effect of statin on risk of gynecologic cancers: A meta-analysis of observational studies and randomized controlled trials. Gynecol. Oncol. 2014, 133, 647–655. [Google Scholar] [CrossRef]
  67. Sperling, C.D.; Verdoodt, F.; Kjaer Hansen, M.; Dehlendorff, C.; Friis, S.; Kjaer, S.K. Statin use and mortality among endometrial cancer patients: A Danish nationwide cohort study. Int. J. Cancer 2018, 143, 2668–2676. [Google Scholar] [CrossRef] [PubMed]
  68. Segev, Y.; Gemer, O.; Helpman, L.; Hag-Yahia, N.; Eitan, R.; Raban, O.; Vaknin, Z.; Ben-Arie, A.; Amit, A.; Levy, T.; et al. An Israeli Gynecologic Oncology Group study of statin use and endometrial cancer prognosis. Int. J. Gynaecol. Obstet. 2020, 148, 79–86. [Google Scholar] [CrossRef] [Green Version]
  69. Colombo, N.; Sessa, C.; du Bois, A.; Ledermann, J.; McCluggage, W.G.; McNeish, I.; Morice, P.; Pignata, S.; Ray-Coquard, I.; Vergote, I.; et al. ESMO-ESGO Ovarian Cancer Consensus Conference Working Group. ESMO-ESGO consensus conference recommendations on ovarian cancer: Pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease. Ann. Oncol. 2019, 30, 672–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Urpilainen, E.; Marttila, M.; Hautakoski, A.; Arffman, M.; Sund, R.; Ilanne-Parikka, P.; Arima, R.; Kangaskokko, J.; Puistola, U.; Läärä, E.; et al. The role of metformin and statins in the incidence of epithelial ovarian cancer in type 2 diabetes: A cohort and nested case-control study. BJOG 2018, 125, 1001–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Irvin, S.; Clarke, M.A.; Trabert, B.; Wentzensen, N. Systematic review and meta-analysis of studies assessing the relationship between statin use and risk of ovarian cancer. Cancer Causes Control 2020, 31, 869–879. [Google Scholar] [CrossRef] [PubMed]
  72. Yarmolinsky, J.; Bull, C.J.; Vincent, E.E.; Robinson, J.; Walther, A.; Smith, G.D.; Lewis, S.J.; Relton, C.L.; Martin, R.M. Association between genetically proxied inhibition of HMG-CoA reductase and epithelial ovarian cancer. JAMA 2020, 323, 646–655. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, H.; Liang, S.L.; Kumar, S.; Weyman, C.M.; Liu, W.; Zhou, A. Statins induce apoptosis in ovarian cancer cells through activation of JNK and enhancement of Bim expression. Cancer Chemother. Pharmacol. 2009, 63, 997–1005. [Google Scholar] [CrossRef] [Green Version]
  74. Majdi, A.; Na, R.; Dixon-Suen, S.; Jordan, S.J.; Webb, P.M. Common medications and survival in women with ovarian cancer: A systematic review and meta-analysis. Gynecol. Oncol. 2020, 157, 678–685. [Google Scholar] [CrossRef]
  75. Jeong, G.H.; Lee, K.H.; Kim, J.Y.; Eisenhut, M.; Kronbichler, A.; van der Vliet, H.J.; Shin, J.I.; Gamerith, G. Statin and cancer mortality and survival: An umbrella systematic review and meta-analysis. J. Clin. Med. 2020, 9, 326. [Google Scholar] [CrossRef] [Green Version]
  76. Harding, B.N.; Delaney, J.A.; Urban, R.R.; Weiss, N.S. Use of statin medications following diagnosis in relation to survival among women with ovarian cancer. Cancer Epidemiol. Biomark. Prev. 2019, 28, 1127–1133. [Google Scholar] [CrossRef] [PubMed]
  77. Couttenier, A.; Lacroix, O.; Vaes, E.; Cardwell, C.R.; De Schutter, H.; Robert, A. Statin use is associated with improved survival in ovarian cancer: A retrospective population-based study. PLoS ONE 2017, 12, e0189233. [Google Scholar] [CrossRef] [PubMed]
  78. Criscuolo, D.; Avolio, R.; Calice, G.; Laezza, C.; Paladino, S.; Navarra, G.; Maddalena, F.; Crispo, F.; Pagano, C.; Bifulco, M.; et al. Cholesterol homeostasis modulates platinum sensitivity in human ovarian cancer. Cells 2020, 9, 828. [Google Scholar] [CrossRef] [PubMed]
  79. Mormile, R. Statin therapy and survival among women with ovarian cancer: How much of it is true? Pathol. Oncol. Res. 2020, 26, 1365–1366. [Google Scholar] [CrossRef]
Figure 1. The structure of (a) six currently marketed statins, and (b) mevastatin (compactin) and lovastatin (mevinolin), the first two isolated and known statins. Both mevastatin and lovastatin are naturally-occurring statins, pravastatin is derived from mevastatin via biotransformation process, simvastatin is a semi-synthetic analog of lovastatin, while fluvastatin, atorvastatin, rosuvastatin and pitavastatin are fully synthetic compounds from the statins class.
Figure 1. The structure of (a) six currently marketed statins, and (b) mevastatin (compactin) and lovastatin (mevinolin), the first two isolated and known statins. Both mevastatin and lovastatin are naturally-occurring statins, pravastatin is derived from mevastatin via biotransformation process, simvastatin is a semi-synthetic analog of lovastatin, while fluvastatin, atorvastatin, rosuvastatin and pitavastatin are fully synthetic compounds from the statins class.
Pharmaceuticals 13 00422 g001
Figure 2. The similarity between structures of HMG-CoA molecule and statins.
Figure 2. The similarity between structures of HMG-CoA molecule and statins.
Pharmaceuticals 13 00422 g002
Figure 3. The number of prescriptions of statins in the United States in the period 2007 to 2017 (source: https://clincalc.com; accessed on 19 September 2020).
Figure 3. The number of prescriptions of statins in the United States in the period 2007 to 2017 (source: https://clincalc.com; accessed on 19 September 2020).
Pharmaceuticals 13 00422 g003
Table 1. A summary of basic information about the most prescribed compounds from the statins class (source: https://clincalc.com; accessed on 19 September 2020).
Table 1. A summary of basic information about the most prescribed compounds from the statins class (source: https://clincalc.com; accessed on 19 September 2020).
StatinBrand Name SynonymsFDA ApprovalTotal Prescriptions (2017)Rank/Change
atorvastatinLipitor®1996104,774,0062/ Pharmaceuticals 13 00422 i0011
simvastatinFlolipid®, Zocor®199156,708,6178/ Pharmaceuticals 13 00422 i0020
pravastatin 1Pravachol®199124,812,69824/ Pharmaceuticals 13 00422 i0033
rosuvastatinCrestor®, Ezallor®200319,628,89739/ Pharmaceuticals 13 00422 i0042
lovastatinAltoprev®, Mevacor®19879,453,81584/ Pharmaceuticals 13 00422 i00512
1 Pravastatin sodium.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Markowska, A.; Antoszczak, M.; Markowska, J.; Huczyński, A. Statins: HMG-CoA Reductase Inhibitors as Potential Anticancer Agents against Malignant Neoplasms in Women. Pharmaceuticals 2020, 13, 422. https://0-doi-org.brum.beds.ac.uk/10.3390/ph13120422

AMA Style

Markowska A, Antoszczak M, Markowska J, Huczyński A. Statins: HMG-CoA Reductase Inhibitors as Potential Anticancer Agents against Malignant Neoplasms in Women. Pharmaceuticals. 2020; 13(12):422. https://0-doi-org.brum.beds.ac.uk/10.3390/ph13120422

Chicago/Turabian Style

Markowska, Anna, Michał Antoszczak, Janina Markowska, and Adam Huczyński. 2020. "Statins: HMG-CoA Reductase Inhibitors as Potential Anticancer Agents against Malignant Neoplasms in Women" Pharmaceuticals 13, no. 12: 422. https://0-doi-org.brum.beds.ac.uk/10.3390/ph13120422

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop