Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2018 Jul 9;284(4):346–357. doi: 10.1111/joim.12806

Statins: a role in breast cancer therapy?

S Borgquist 1,2,, O Bjarnadottir 2, S Kimbung 2, T P Ahern 3
PMCID: PMC6175478  PMID: 29923256

Abstract

Statin drugs have been used for more than two decades to treat hypercholesterolemia and as cardio‐preventive drugs, resulting in a marked decrease in cardiovascular morbidity and mortality worldwide. Statins halt hepatic cholesterol biosynthesis by inhibiting the rate‐limiting enzyme in the mevalonate pathway, hydroxymethylglutaryl‐coenzyme A reductase (HMGCR). The mevalonate pathway regulates a host of biochemical processes in addition to cholesterol production. Attenuation of these pathways is likely responsible for the myriad benefits of statin therapy beyond cholesterol reduction – the so‐called pleiotropic effects of statins. Chief amongst these purported effects is anti‐cancer activity. A considerable body of preclinical, epidemiologic and clinical evidence shows that statins impair proliferation of breast cancer cells and reduce the risk of breast cancer recurrence. Potential mechanisms for this effect have been explored in laboratory models, but remain poorly understood and require further investigation. The number of clinical trials assessing the putative clinical benefit of statins in breast cancer is increasing. Currently, a total of 30 breast cancer/statin trials are listed at the global trial identifier website clinicaltrials.gov. Given the compelling evidence from performed trials in a variety of clinical settings, there have been calls for a clinical trial of statins in the adjuvant breast cancer setting. It would be imperative for such a trial to incorporate tumour biomarkers predictive of statin response in its design and analysis plan. Ongoing translational clinical trials aimed at biomarker discovery will help identify, which breast cancer patients are most likely to benefit from adjuvant statin therapy, and will add valuable clinical knowledge to the field.

Keywords: breast cancer, cholesterol, endocrine therapy, HMGCR, statins

Breast cancer

Breast cancer is globally the most common malignancy amongst women, contributing more than 25% of the total number of new cancer cases diagnosed (http://www.wcrf.org). The number of breast cancer cases has steadily increased over the last few decades, although annual incidence rates vary greatly worldwide, from 19.3 per 100 000 women in Eastern Africa to 89.7 per 100 000 women in Western Europe (http://www.who.int). Worldwide, breast cancer ranks as the fifth most frequent cause of cancer death, and in the female population, it is the second most frequent cause of cancer death 1. In parallel with the incidence rates for breast cancer, the prevalence of overweight and obesity has rapidly risen. Overweight/obesity is often associated with the metabolic syndrome and increases the risk of a number of diseases, including hypercholesterolemia and breast cancer 2. Not only does overweight/obesity influence breast cancer incidence 3, 4, it also worsens prognosis following breast cancer 5. The epidemiological and clinically manifested linkage between overweight/obesity and breast cancer is increasingly clear 6, although important gaps exist in our knowledge. A common comorbidity of overweight/obesity is hypercholesterolemia, and one knowledge gap is, for example, the impact of cholesterol on the progression of breast cancer and on the effectiveness of adjuvant endocrine therapy.

Molecular basis for statin use as breast cancer therapeutic agents

Statins are routinely used to treat hyperlipidaemia, but may also have antineoplastic effects. The target for statins, HMGCR, is a transmembrane glycoprotein found in the endoplasmic reticulum in all cells. HMGCR activity controls the mevalonate pathway, which in addition to cholesterol produces steroid‐based hormones and non‐sterol isoprenoids (Fig. 1) 7, 8. Inhibition of hepatic HMGCR causes reduced intracellular cholesterol levels in hepatocytes. This in turn triggers up‐regulation of low‐density lipoprotein cholesterol (LDL‐C) receptors to scavenge cholesterol from the serum to support cell growth and division. Serum levels of LDL‐C consequently plummet, reducing the rate of adverse cardiovascular events in statin‐treated individuals 9. Cellular cholesterol levels and HMGCR activity are maintained via tightly regulated feedback mechanisms, whereas extracellular serum cholesterol concentrations vary 10. In addition to reducing LDL‐C production, blocking the mevalonate pathway interrupts synthesis of the isoprenoids geranylgeranyl pyrophosphate (GGPP) and farnesyl pyrophosphate (FPP) 7, 8. Isoprenylation of proteins by GGPP and FPP enables subcellular localization and intracellular trafficking of membrane‐associated proteins that are essential for the cell 11. GGPP and FPP post‐translationally prenylate a host of proteins – including the oncogene products Ras, Rac and Rho – to enable their full function 11, 12, 13. The isoprenoids demonstrate tumour‐suppressive properties as regulators of important processes in cancer such as proliferation, migration and angiogenesis 7, 14. Comprehensive analysis of statin‐induced changes in tumour cell lipid profiles may reveal the dominant pathways through which statins attenuate tumour promotion 15.

Figure 1.

Figure 1

Pathways and potential predictive biomarkers that may mediate breast tumour response to statin therapy.

Highly proliferative cells (such as cancer cells) must rapidly produce lipid bilayer membranes, requiring increased cholesterol biosynthesis 16. Whilst the cholesterol biosynthesis pathway is tightly regulated in normal cells, it may be dysregulated in cancer cells 17. It has been implied that HMGCR is a metabolic oncogene, and that dysregulation of the mevalonate pathway promotes transformation 18. In addition, high mRNA levels of HMGCR and other mevalonate pathway genes were associated with impaired prognosis for breast cancer patients 18. This finding was recently validated in two large breast cancer datasets 19.

The mevalonate pathway is a possible therapeutic target for tumours with mutations of the tumour suppressor p53 20, 21. The p53 protein, encoded by the TP53 gene, is dubbed ‘the guardian of the genome’ due its tumour‐suppressing activities. These activities, triggered by DNA damage, include activation of DNA repair mechanisms, initiation of growth arrest at the G1/S boundary and induction of apoptosis if repair fails. The mevalonate pathway is both necessary and sufficient for the phenotypic effects of mutant p53 on breast tissue architecture, and mutant p53 associates with sterol gene promoters 20. In a three‐dimensional culture model, mutant p53 up‐regulated mevalonate pathway genes in breast cancer cells, leading to disordered, invasive morphology 20. In the same model, depletion of mutant p53 by RNA interference caused reversion to normal morphology. Remarkably, the addition of clinically achievable concentrations of simvastatin to the culture system resulted in marked reductions in tumour cell growth, induction of apoptosis and reversion to normal morphology in the various breast cancer cell lines tested 20. The beneficial effects of simvastatin were negated when the mevalonate pathway products GGPP and FPP were simultaneously added to the culture medium. The p53 effect is likely modulated by sterol regulatory element‐binding proteins (SREBPs), and tied to the YAP/TAZ effectors of the Hippo signalling pathway 22. YAP/TAZ activity is also controlled by Rho GTPases, which are dependent on prenylation for activation 23. Therefore, overexpression of genes encoding p53, SREBPs, mevalonate pathway genes and the YAP/TAZ transcriptional regulators may identify breast tumours that will be sensitive to statin treatment.

A large number of in vitro and in vivo cancer studies with statins have been performed (Table 1), with many more underway. So far, these models have shown that statins decrease proliferation and increase apoptosis of breast cancer cells 18, 24. The biological mechanisms for these actions are not yet fully elucidated. Our previous work demonstrated that HMGCR is differentially expressed in human breast cancer samples and holds prognostic value 25, 26. HMGCR may also predict tumour response to endocrine treatment 27, as well as to statin treatment, in a recent phase II clinical trial (ClinicalTrials.gov Identifier: NCT00816244) 28. Other in vitro studies demonstrated substantial statin‐induced increases in HMGCR expression 29, and atorvastatin induced HMGCR up‐regulation when assessed with a novel, well‐validated monoclonal HMGCR antibody 19. Additionally, in vivo studies suggest that HMGCR activity is higher in mammary tumours compared with normal mammary glands, and that tumours are resistant to feedback regulation by sterols 30.

Table 1.

Key experimental data linking statin treatment to decrease proliferation and cell death induction in cancer

Reference Research findings Tumour type
Campbell et al. 67 Cancer cells with activated Ras or ErbB2 pathways and low oestrogen receptor expression were more susceptible to statin‐induced anti‐proliferative and pro‐apoptotic signals. Statin sensitivity also correlated with endogenous levels of activated nuclear factor kappaB (NF‐kappaB) Breast
Wong et al. 68 Half of a panel of 17 genetically distinct multiple myeloma cell lines displayed significant sensitivity to statin‐induced apoptosis. Addition of mevalonate, geranylgeranyl PPi, farnesyl PPi, completely or partially rescued the sensitive cells from the statin‐induced apoptosis, thus highlighting the importance of isoprenylation in this process Multiple myeloma
Clendening et al. 18 Overexpression of HMGCR accentuates growth of transformed and non‐transformed cells in vitro and in mice. This occurs via mechanisms including cooperation with RAS to drive cell transformation Breast, colorectal
Clendening et al. 69 Statin treatment inhibits proliferation and induced apoptosis in a subset of primary myeloma cells. Dysregulation of the mevalonate pathway may distinguish between sensitive and resistant cells Multiple myeloma
Garwood et al. 70 Preoperative treatment with high dose (80 mg day−1) or low dose (20 mg day−1) fluvastatin for 3–6 weeks showed measurable biologic activity by reducing tumor proliferation and increasing apoptosis in high‐grade, stage 0/1 primary tumours Breast
Bjarnadottir et al. 28 Preoperative treatment with high dose (80 mg day−1) atorvastatin for 2 weeks showed measurable biologic activity by reducing tumor proliferation in HMGCR‐expressing primary tumours Breast
Freed‐Pastor et al. 20 Genome‐wide expression analysis revealed that p53 mutants significantly upregulate the mevalonate pathway. Treatment with statins and sterol biosynthesis intermediates reveal that the mevalonate pathway is both necessary and sufficient for the phenotypic effects of mutant p53 on breast tissue architecture implicating the mevalonate pathway as a therapeutic target for tumours bearing mutations in p53 Breast
Nelson et al. 33 Cholesterol through its conversion to 27‐hydroxycholesterol increased ER‐dependent cell and tumor growth. These effects were attenuated by treatment with statins and CYP27A1 inhibitors Breast
Sorrentino et al. 22 Statin treatment prevented YAP/TAZ nuclear localization and transcriptional responses which are necessary to promote tissue proliferation and organ growth thus revealing a link between mevalonate‐YAP/TAZ axis which is required for proliferation and self‐renewal of breast cancer cells Breast
Pandyra et al. 17 Blocking the sterol‐feedback loop initiated by statin treatment, by co‐inhibiting SREBP2 significantly potentiates the anti‐tumour activity of statins Breast, Lung

If statins do target the mevalonate pathway in cancer cells within a tumour, the lowering of intracellular cholesterol may lead to lowered intra‐tumoural autocrine hormone production, as cholesterol is fundamental for all steroid hormone synthesis. Interestingly, one study has reported that atorvastatin and its metabolites are detectable in human breast samples following oral administration 31, indicating that direct inhibition of HMGCR may occur in breast tumours. Most relevant for an endocrine responsive disease such as oestrogen receptor (ER)‐positive breast cancer, statin treatment reduces levels of the cholesterol metabolite 27‐hydroxy cholesterol (27HC) 32. 27HC acts as an ER ligand, potentiating ER‐dependent tumour growth 33, 34, 35, 36. Interestingly, 27HC can agonize both the ER and the liver X receptor (LXR) to drive breast tumour proliferation 33, 34, and it promotes metastasis through interactions with myeloid immune cells 37. The cholesterol biosynthesis pathway was recently shown to be up‐regulated in ER‐positive breast cancer cell lines that are resistant to oestrogen deprivation 38, 39, suggesting that dysregulation of cholesterol biosynthesis may be a mechanism of endocrine resistance in hormone receptor‐positive breast cancer 38, 39. Chronic oestrogen deprivation in ER‐positive breast cancer cells seems to stabilize the epigenetic activation of the mevalonate pathway and cholesterol biosynthesis 39, and this leads to the accumulation of other ligands (such as 27HC), which potentiate ER signalling in the absence of oestrogen, consequently driving the activation of genes that promote a proliferative and invasive cell phenotype 39.

To summarize, cancer cells depend on cholesterol for continued growth and survival. Therefore, attenuating cholesterol biosynthesis seems to be a promising anti‐cancer strategy. Of note, rapidly proliferating cancer cells have an increased cholesterol demand to enable cell membrane synthesis 16, 40. By lowering plasma levels of cholesterol and 27HC, their availability for use by cancer cells is consequently lowered. Additionally, direct inhibition of HMGCR by statins depletes intratumoural reserves of isoprenoids, which are key regulators of cancer cell proliferation and metastasis. Ongoing studies are exploring additional roles that cholesterol, cholesterol metabolites and statins play in breast tumour promotion.

Breast cancer risk reduction by statins

The positive association between overweight and obese body constitutions on risk of postmenopausal breast cancer has been demonstrated by several studies 3, 6, 41, 42, 43. The biological underpinnings of the observed association between obesity and breast cancer are not completely understood; both in terms of effects from the local micro‐environment on progression 44, and also regarding breast cancer initiation (an area that has hardly been explored) 45. Overweight‐associated alterations in circulating cholesterol might partly explain the association between obesity and breast cancer 46. Evidence for an association between cholesterol and cancer incidence is not consistent 47, 48, 49. Some epidemiological studies previously indicated lower cancer incidence amongst statin users 50, 51, although the results have been inconsistent across studies 52, 53, 54, 55. Our recent update from the large, prospective Nurse's Health Study (NHS) showed no association between different statin exposures and risk of invasive breast cancer 56. Results from a prominent meta‐analysis were null 54. Notably in the NHS, statin use was not associated with increased risk of invasive breast cancer, regardless of histological type. Nonetheless, development of a subset of breast cancer – presumably subtypes highly dependent on functional cholesterol metabolism – may possibly be obstructed by treatment with cholesterol‐lowering agents. This hypothesis stresses the need for a much more sophisticated approach to modelling risk of breast cancer molecular subtypes.

Potential future studies of statins in relation to breast cancer incidence should explore effects in an appropriately powered primary prevention trial amongst high‐risk women. Such a trial would benefit from translational studies where the biological relevance of statins to prevent breast cancer is understood. To investigate the biological processes of statins as a primary preventive drug in experimental models have yet not been investigated in relevant models involving normal breast epithelial cells and stromal cells, which will be critical for understanding the potential role of statins in breast cancer initiation, and therefore primary prevention.

Statins and breast cancer prognosis

The U.S. National Cancer Institute estimated that there were over 2.7 million breast cancer survivors living in the United States in 2009 57. Statins are inexpensive and well‐tolerated drugs, taken long term by about one‐third of older US adults, so a beneficial effect of statins on breast cancer outcomes would have important public health impact. There is considerable evidence for a protective effect of statin use against recurrence amongst survivors of nonmetastatic breast cancer (Fig. 2). One study of about 2000 breast cancer survivors observed an imprecise protective association between statin use and recurrence (HR = 0.67, 95% CI: (0.39, 1.13) 58. This association was measured with greater precision in a cohort of 18 769 Danish breast cancer survivors, in which statin users had a reduced rate of recurrence compared with never users (HR = 0.73, 95% CI: (0.60, 0.89) 59. Since publication of these studies, 11 others have measured the association in a variety of source populations 60. A systematic review and meta‐analysis reported a summary relative risk associating statin use with breast cancer recurrence of 0.64 (95% CI: (0.53–0.79) 60. Another meta‐analysis showed that statin use was associated with lower breast cancer‐specific mortality (lipophilic statins) and all‐cause mortality (lipophilic and hydrophilic statins) 61. A third meta‐analysis of observational studies showed that postdiagnostic statin use was associated with lower cancer‐specific mortality in breast cancer, and prediagnostic statin use was negatively associated with both all‐cause and cancer‐specific mortality 62.

Figure 2.

Figure 2

The prognostic value of statin treatment in the adjuvant breast cancer setting illustrated by a forest plot of the currently reported studies.

Statins and response to endocrine therapy in breast cancer

Hypercholesterolemia is an established clinical side effect of aromatase inhibitor (AI) therapy. Given the revelations about the estrogenic cholesterol metabolite 27HC, this phenomenon may counteract the intended effects of AIs 33, 63. Breast cancer patients experiencing hypercholesterolemia during AI treatment may thus benefit from concomitant cholesterol‐lowering treatment to reduce the probability of 27HC‐driven ER actions. Tamoxifen, on the other hand, reduces circulating cholesterol levels. The large‐scale Breast International Group (BIG) 1–98 study examined the clinical efficacy of tamoxifen and AI, respectively, as adjuvant endocrine therapy. Within the BIG 1–98 study population, we investigated whether initiation of cholesterol‐lowering medication (CLM) concomitantly with endocrine therapy in the adjuvant setting was related to prognosis. Initiation of CLM improved disease‐free survival (HR = 0.79, 95% CI: (0.66–0.95), breast cancer‐free interval (HR 0.76, 95% CI: (0.60–0.97) and distant‐recurrence‐free interval (HR 0.74, 95% CI: (0.56–0.97). We concluded that CLM during adjuvant endocrine treatment appears to have a favourable impact on clinical outcome in ER positive breast cancer 64. The beneficial effects of concomitant CLM were apparent for both endocrine drug groups (AI and tamoxifen).

Since cholesterol forms the biochemical scaffold for all steroid hormones (e.g. oestradiol), it is likely to play a crucial role in hormone (oestrogen)‐dependent breast cancer. Importantly, 27HC, the oxysterol produced from cholesterol, is involved in the regulation of intracellular cholesterol homeostasis 33, but also acts as an endogenous selective oestrogen receptor modulator capable of increasing the growth and metastasis of tumours 63. Valuable clinical insight into the impact of statin treatment on 27HC in breast cancer patients was gained through collaboration with the McDonnell Lab at Duke University 32, where biological samples from our “window‐of‐opportunity” statin trial 19, 28, 65 were analysed. This work demonstrated that statin therapy reduces 27HC in parallel with reductions in LDL cholesterol levels 32. A significant up‐regulation of tumour expression of CYP27A1, the enzyme responsible for the production of 27HC from cholesterol (Fig. 1), was, however, observed in breast tumours following statin therapy. Further investigation revealed noteworthy associations between CYP27A1 expression and adverse tumour characteristics and with poorer breast cancer prognosis in women 50 years and older (presumably postmenopausal women). The revelation that prognosis was worse for older women with high tumour CYP27A1 expression (and, presumably, high 27HC), is of relevance as these will be the breast cancer patients who are prescribed AI treatment; measures to address 27HC levels may improve outcomes for these women.

AIs have now been used in postmenopausal breast cancer treatment for more than a decade. However, updated knowledge on the interaction between endocrine treatment and obesity/hypercholesterolemia is needed to ensure that breast cancer patients with lipid‐associated comorbidities, such as obesity and hypercholesterolemia, receive optimal endocrine treatment.

Predictive factors for statin response in breast cancer

The well‐characterized molecular and clinical heterogeneity of breast cancer warrants that for every novel drug showing efficacy against this disease, specific treatment predictive biomarkers should be identified to enable the precise selection and treatment of only those patients who may derive clinical benefit from the treatment. As such, the search for statin treatment predictive markers in breast cancer has been the subject of many studies (Table 2). HMGCR has been shown to be overexpressed in about 80% of breast tumours, and its expression is correlated with less aggressive tumour phenotype and longer recurrence‐free survival 25, 26, 27. In the aforementioned clinical phase II trial when 50 breast cancer patients were treated preoperatively with 80 mg day−1 atorvastatin for 2 weeks, tumour levels of Ki67 (a proliferation marker) decreased from presurgical biopsy to resected tumour only if the tumour expressed HMGCR 28, 65, suggesting a predictive role for HMGCR for efficacy of statin treatment in breast cancer. HMGCR was not only differentially expressed across tumours from breast cancer patients, but was also associated with improved prognosis amongst ER‐positive breast cancer patients, whereas ER‐negative patients seemed to have better outcomes when HMGCR was absent 25, 26. Several other mevalonate pathway biomarkers have been associated with breast tumour response to statins in vitro and in animal models. However, no study has comprehensively evaluated the network comprised of these individual factors, and how it is perturbed by statin exposure to alter recurrence risk. Randomized trials are now warranted to clarify the potential beneficial effects of statins in breast cancer management in the adjuvant and metastatic setting. In October 2016, the first trial with statins in metastatic breast cancer was launched, with the hypothesis that HMGCR expression will identify tumours that will respond to statin treatment (ClinicalTrials.gov Identifier: NCT02958852).

Table 2.

Candidate predictive biomarkers linking statin exposure to breast cancer recurrence risk

Candidate biomarker Prevalence Univariate hypothesis Reference
SLCO1B1 rs4149056 variant 0.15 Statin‐treated breast cancer survivors who carry the variant allele will have a lower rate of breast cancer recurrence compared with statin‐treated survivors who carry the normal allele Ahern et al. 8
HMG‐CoA reductase expression 0.75 Statin‐treated breast cancer survivors whose primary tumours express high levels of HMG‐CoAR will have a lower rate of breast cancer recurrence than statin treated survivors whose tumours express little or no HMG‐CoAR Borgquist et al. 26
YAP/TAZ expression 0.9 Statin‐treated breast cancer survivors whose primary tumours exhibit positive nuclear YAP/TAZ staining will have a lower rate of breast cancer recurrence than statin‐treated survivors whose tumours do not exhibit nuclear YAP/TAZ staining Sorrentino et al. 22
CYP3A4 rs35599367 and CYP3A5 rs776746variants 0.02
0.31
Statin‐treated breast cancer survivors who carry at least one of the CYP3A4/5 variants will have a lower rate of breast cancer recurrence than statin‐treated survivors who are homozygous wild type at both loci Ahern et al. 8
ABCB1 rs2032582 variant 0.45 Statin‐treated breast cancer survivors who carry at least one variant allele will have a lower rate of breast cancer recurrence than statin‐treated survivors who carry only wild‐type alleles Fiegenbaum et al. 71
DDX20 expression 0.80 Statin‐treated breast cancer survivors whose tumours express high levels of DDX20 will have a lower rate of breast cancer recurrence than statin‐treated survivors whose tumours express little or no DDX20 Shin et al. 72
LDL receptor expression 0.75 Statin‐treated breast cancer survivors whose tumours express high levels of LDL receptor will have a lower rate of breast cancer recurrence than statin‐treated survivors whose tumours express little or no LDL receptor Liu et al. 73
Mutant p53 0.30 Statin‐treated breast cancer survivors whose primary tumours express mutant p53 will have a lower rate of breast cancer recurrence than statin‐treated survivors whose tumours do not express mutant p53 Freed‐Pastor et al. 20
“Cholesterol biosynthesis signature” 0.70 Breast cancer survivors whose primary tumours show low activity of the “cholesterol biosynthesis signature” and receive adjuvant statin treatment will have a lower rate of breast cancer recurrence compared with nonstatin treated or survivors whose tumours have a high “cholesterol biosynthesis signature” activity Kimbung et al. 19
EMT‐associated genes (VIM, CDH1, ZEB1, FN1, and CDH2) 0.88 Statin‐treated breast cancer survivors whose tumours express high levels of EMT‐associated genes will have a lower rate of breast cancer recurrence than statin‐treated survivors with lower EMT‐associated genes expressing tumours Yu et al. 74

Exploring transcription profiles associated with breast cancer sensitivity to statin treatment is another method that has been explored to discover genes or gene signatures that are predictive of statin treatment sensitivity. In one such study based on in vitro experiments with atorvastatin in a collection of breast cancer cell lines, we aimed to uncover transcriptional differences associated with statin response in breast cancer 19, 65. The strongest discriminants between the breast cancer cells associated with statin sensitivity at the transcriptional level were the expression of the oestrogen receptor and a gene set enriched for genes involved in the cholesterol biosynthesis pathway. Following statin treatment, the less‐sensitive cells exhibited the classical response of up‐regulating the expression of genes in the cholesterol biosynthesis pathway via the normal negative feedback loop resulting from the statin‐induced inhibition of HMGCR. This classical response was, however, weaker in the sensitive cells, suggesting that these cells may possess an inherent defect in this pathway. This cholesterol biosynthesis gene set showed potential for identifying tumours that experience reduced proliferation upon statin treatment, albeit in a small cohort of patients. Further studies using similar and more advanced approaches are necessary to identify robust biomarkers for identifying patients most likely to derive benefit from the addition of cholesterol‐lowering medications to their therapeutic regimen for controlling breast cancer.

Clinical breast cancer trials with statins

The number of trials assessing the putative clinical benefit of statins in breast cancer is increasing. A total of 30 trials were listed at the global trial identifier website clinicaltrials.gov at the time of this writing (February 23, 2018). Of these, eight trials are currently recruiting; another two trials are active but not yet recruiting, and nine trials are already complete. The remaining trials have either been withdrawn, are not yet active or were terminated for other reasons (e.g. slow accrual). Many of the trials include a translational protocol, reflecting the understanding that the success of novel therapies requires molecular prediction of treatment efficacy. Given the compelling evidence from several trials in a variety of clinical settings, there have been calls for a clinical trial of statins in the adjuvant breast cancer setting 8, 66. It would be imperative for such a trial to incorporate tumour biomarkers predictive of statin response in its design and analysis plan.

Concluding remarks

Taken together, obesity‐associated metabolic disorders such as hypercholesterolemia can have a negative impact on the prognosis of breast cancer patients. This may in particular be of importance in the endocrine treatment setting. Future research activities should evaluate the interplay between host factors (e.g. obesity/hypercholesterolemia), treatment factors (e.g. statin therapy) and breast cancer progression. If such studies detect a group of patients with less expected clinical efficacy of the standard endocrine drug of choice, this may impact future clinical guidelines for the purpose of improved clinical outcome amongst breast cancer patients. Advanced understanding of the molecular mechanisms of breast cancer response to common prescription drugs such as statins stands to increase the effectiveness with which we treat breast cancer and lead to the development of new therapies with specific actions against up‐regulated molecular pathways.

Funding

This work was supported by grants received from the Swedish Research Council, the Crafoord Foundation and the Swedish Cancer Foundation by S. Borgquist. T. Ahern was supported by an award from the National Institute of General Medical Studies at the U.S. National Institutes of Health (P20 GM103644)

Conflicts of interest statement

All authors declare to have no relevant conflict of interest.

Borgquist S, Bjarnadottir O, Kimbung S, Ahern TP (Aarhus University Hospital, Aarhus C, Denmark; Lund University, Lund, Sweden; and The University of Vermont, Burlington, VT, USA). Statins: a role in breast cancer therapy? (Review). J Intern Med 2018; 284: 346–357.

References

  • 1. Ferlay J, Soerjomataram I, Dikshit R et al Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 136: E359–86. [DOI] [PubMed] [Google Scholar]
  • 2. Hauner D, Hauner H. Metabolic syndrome and breast cancer: is there a link? Breast Care (Basel) 2014; 9: 277–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Borgquist S, Jirstrom K, Anagnostaki L et al Anthropometric factors in relation to different tumor biological subgroups of postmenopausal breast cancer. Int J Cancer 2009; 124: 402–11. [DOI] [PubMed] [Google Scholar]
  • 4. Lahmann PH, Hoffmann K, Allen N et al Body size and breast cancer risk: findings from the European Prospective Investigation into Cancer And Nutrition (EPIC). Int J Cancer 2004; 111: 762–71. [DOI] [PubMed] [Google Scholar]
  • 5. Ewertz M, Jensen MB, Gunnarsdottir KA et al Effect of obesity on prognosis after early‐stage breast cancer. J Clin Oncol 2011; 29: 25–31. [DOI] [PubMed] [Google Scholar]
  • 6. Goodwin PJ, Stambolic V. Impact of the obesity epidemic on cancer. Annu Rev Med 2015; 66: 281–96. [DOI] [PubMed] [Google Scholar]
  • 7. Liao JK. Isoprenoids as mediators of the biological effects of statins. J Clin Invest 2002; 110: 285–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Ahern TP, Lash TL, Damkier P et al Statins and breast cancer prognosis: evidence and opportunities. Lancet Oncol 2014; 15: e461–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Collins R, Reith C, Emberson J et al Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet 2016; 388: 2532–61. [DOI] [PubMed] [Google Scholar]
  • 10. Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell 2015; 161: 161–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Liao JK, Laufs U. Pleiotropic effects of statins. Annu Rev Pharmacol Toxicol 2005; 45: 89–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Zhang FL, Casey PJ. Protein prenylation: molecular mechanisms and functional consequences. Annu Rev Biochem 1996; 65: 241–69. [DOI] [PubMed] [Google Scholar]
  • 13. Thurnher M, Nussbaumer O, Gruenbacher G. Novel aspects of mevalonate pathway inhibitors as antitumor agents. Clin Cancer Res 2012; 18: 3524–31. [DOI] [PubMed] [Google Scholar]
  • 14. Mo H, Elson CE. Studies of the isoprenoid‐mediated inhibition of mevalonate synthesis applied to cancer chemotherapy and chemoprevention. Exp Biol Med (Maywood) 2004; 229: 567–85. [DOI] [PubMed] [Google Scholar]
  • 15. Han X, Gross RW. Global analyses of cellular lipidomes directly from crude extracts of biological samples by ESI mass spectrometry: a bridge to lipidomics. J Lipid Res 2003; 44: 1071–9. [DOI] [PubMed] [Google Scholar]
  • 16. Mullen PJ, Yu R, Longo J et al The interplay between cell signalling and the mevalonate pathway in cancer. Nat Rev Cancer 2016; 16: 718–31. [DOI] [PubMed] [Google Scholar]
  • 17. Pandyra AA, Mullen PJ, Goard CA et al Genome‐wide RNAi analysis reveals that simultaneous inhibition of specific mevalonate pathway genes potentiates tumor cell death. Oncotarget 2015; 6: 26909–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Clendening JW, Pandyra A, Boutros PC et al Dysregulation of the mevalonate pathway promotes transformation. Proc Natl Acad Sci USA 2010; 107: 15051–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Kimbung S, Lettiero B, Feldt M et al High expression of cholesterol biosynthesis genes is associated with resistance to statin treatment and inferior survival in breast cancer. Oncotarget 2016; 7: 59640–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Freed‐Pastor WA, Mizuno H, Zhao X et al Mutant p53 disrupts mammary tissue architecture via the mevalonate pathway. Cell 2012; 148: 244–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Ingallina E, Sorrentino G, Bertolio R et al Mechanical cues control mutant p53 stability through a mevalonate‐RhoA axis. Nat Cell Biol 2018; 20: 28–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Sorrentino G, Ruggeri N, Specchia V et al Metabolic control of YAP and TAZ by the mevalonate pathway. Nat Cell Biol 2014; 16: 357–66. [DOI] [PubMed] [Google Scholar]
  • 23. Yu FX, Zhao B, Panupinthu N et al Regulation of the Hippo‐YAP pathway by G‐protein‐coupled receptor signaling. Cell 2012; 150: 780–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Clendening JW, Penn LZ. Targeting tumor cell metabolism with statins. Oncogene 2012; 31: 4967–78. [DOI] [PubMed] [Google Scholar]
  • 25. Borgquist S, Djerbi S, Ponten F et al HMG‐CoA reductase expression in breast cancer is associated with a less aggressive phenotype and influenced by anthropometric factors. Int J Cancer 2008; 123: 1146–53. [DOI] [PubMed] [Google Scholar]
  • 26. Borgquist S, Jogi A, Ponten F et al Prognostic impact of tumour‐specific HMG‐CoA reductase expression in primary breast cancer. Breast Cancer Res 2008; 10: R79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Brennan DJ, Laursen H, O'Connor DP et al Tumor‐specific HMG‐CoA reductase expression in primary premenopausal breast cancer predicts response to tamoxifen. Breast Cancer Res 2011; 13: R12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. 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 2013; 138: 499–508. [DOI] [PubMed] [Google Scholar]
  • 29. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature 1990; 343: 425–30. [DOI] [PubMed] [Google Scholar]
  • 30. El‐Sohemy A, Archer MC. Inhibition of N‐methyl‐N‐nitrosourea‐ and 7,12‐dimethylbenz[a] anthracene‐induced rat mammary tumorigenesis by dietary cholesterol is independent of Ha‐Ras mutations. Carcinogenesis 2000; 21: 827–31. [DOI] [PubMed] [Google Scholar]
  • 31. Arun BK, Gong Y, Liu D et al Phase I biomarker modulation study of atorvastatin in women at increased risk for breast cancer. Breast Cancer Res Treat 2016; 158: 67–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Kimbung S, Chang C, Bendahl PO et al Impact of 27‐hydroxylase (CYP27A1) and 27‐hydroxycholesterol in breast cancer. Endocr Relat Cancer 2017; 24: 339–49. [DOI] [PubMed] [Google Scholar]
  • 33. Nelson ER, Wardell SE, Jasper JS et al 27‐Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology. Science 2013; 342: 1094–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Warner M, Gustafsson JA. On estrogen, cholesterol metabolism, and breast cancer. N Engl J Med 2014; 370: 572–3. [DOI] [PubMed] [Google Scholar]
  • 35. Umetani M, Domoto H, Gormley AK et al 27‐Hydroxycholesterol is an endogenous SERM that inhibits the cardiovascular effects of estrogen. Nat Med 2007; 13: 1185–92. [DOI] [PubMed] [Google Scholar]
  • 36. DuSell CD, Umetani M, Shaul PW et al 27‐hydroxycholesterol is an endogenous selective estrogen receptor modulator. Mol Endocrinol 2008; 22: 65–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Baek AE, Yu YA, He S et al The cholesterol metabolite 27 hydroxycholesterol facilitates breast cancer metastasis through its actions on immune cells. Nat Commun 2017; 8: 864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Simigdala N, Gao Q, Pancholi S et al Cholesterol biosynthesis pathway as a novel mechanism of resistance to estrogen deprivation in estrogen receptor‐positive breast cancer. Breast Cancer Res 2016; 18: 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Nguyen VT, Barozzi I, Faronato M et al Differential epigenetic reprogramming in response to specific endocrine therapies promotes cholesterol biosynthesis and cellular invasion. Nat Commun 2015; 6: 10044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Cruz PM, Mo H, McConathy WJ et al The role of cholesterol metabolism and cholesterol transport in carcinogenesis: a review of scientific findings, relevant to future cancer therapeutics. Front Pharmacol 2013; 4: 119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Borgquist S, Wirfalt E, Jirstrom K et al Diet and body constitution in relation to subgroups of breast cancer defined by tumour grade, proliferation and key cell cycle regulators. Breast Cancer Res 2007; 9: R11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Bianchini F, Kaaks R, Vainio H. Overweight, obesity, and cancer risk. Lancet Oncol 2002; 3: 565–74. [DOI] [PubMed] [Google Scholar]
  • 43. Harvie M, Hooper L, Howell AH. Central obesity and breast cancer risk: a systematic review. Obes Rev 2003; 4: 157–73. [DOI] [PubMed] [Google Scholar]
  • 44. Park J, Morley TS, Kim M et al Obesity and cancer–mechanisms underlying tumour progression and recurrence. Nat Rev Endocrinol 2014; 10: 455–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Renehan AG, Zwahlen M, Egger M. Adiposity and cancer risk: new mechanistic insights from epidemiology. Nat Rev Cancer 2015; 15: 484–98. [DOI] [PubMed] [Google Scholar]
  • 46. Borgquist S, Butt T, Almgren P et al Apo‐lipoproteins, lipids and risk of cancer. Int J Cancer 2016; 138: 2648–56. [DOI] [PubMed] [Google Scholar]
  • 47. Melvin JC, Holmberg L, Rohrmann S et al Serum lipid profiles and cancer risk in the context of obesity: four meta‐analyses. J Cancer Epidemiol 2013; 2013: 823849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Grundy SM. Metabolic complications of obesity. Endocrine 2000; 13: 155–65. [DOI] [PubMed] [Google Scholar]
  • 49. Martin LJ, Melnichouk O, Huszti E et al Serum lipids, lipoproteins, and risk of breast cancer: a nested case‐control study using multiple time points. J Natl Cancer Inst 2015; 107: pii: djv032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Boudreau DM, Yu O, Miglioretti DL et al Statin use and breast cancer risk in a large population‐based setting. Cancer Epidemiol Biomarkers Prev 2007; 16: 416–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Gronich N, Rennert G. Beyond aspirin‐cancer prevention with statins, metformin and bisphosphonates. Nat Rev Clin Oncol 2013; 10: 625–42. [DOI] [PubMed] [Google Scholar]
  • 52. Jacobs EJ, Newton CC, Thun MJ et al Long‐term use of cholesterol‐lowering drugs and cancer incidence in a large United States cohort. Cancer Res 2011; 71: 1763–71. [DOI] [PubMed] [Google Scholar]
  • 53. Undela K, Srikanth V, Bansal D. Statin use and risk of breast cancer: a meta‐analysis of observational studies. Breast Cancer Res Treat 2012; 135: 261–9. [DOI] [PubMed] [Google Scholar]
  • 54. Bonovas S, Filioussi K, Tsavaris N et al Use of statins and breast cancer: a meta‐analysis of seven randomized clinical trials and nine observational studies. J Clin Oncol 2005; 23: 8606–12. [DOI] [PubMed] [Google Scholar]
  • 55. McDougall JA, Malone KE, Daling JR et al Long‐term statin use and risk of ductal and lobular breast cancer among women 55 to 74 years of age. Cancer Epidemiol Biomarkers Prev 2013; 22: 1529–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Borgquist S, Tamimi RM, Chen WY et al Statin use and breast cancer risk in the nurses’ health study. Cancer Epidemiol Biomarkers Prev 2016; 25: 201–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. DeSantis CE, Lin CC, Mariotto AB et al Cancer treatment and survivorship statistics, 2014. CA Cancer J Clin 2014; 64: 252–71. [DOI] [PubMed] [Google Scholar]
  • 58. Kwan ML, Habel LA, Flick ED et al Post‐diagnosis statin use and breast cancer recurrence in a prospective cohort study of early stage breast cancer survivors. Breast Cancer Res Treat 2008; 109: 573–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Ahern TP, Pedersen L, Tarp M et al Statin prescriptions and breast cancer recurrence risk: a Danish nationwide prospective cohort study. J Natl Cancer Inst 2011; 103: 1461–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. 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: 1281–8. [DOI] [PubMed] [Google Scholar]
  • 61. Liu B, Yi Z, Guan X et al The relationship between statins and breast cancer prognosis varies by statin type and exposure time: a meta‐analysis. Breast Cancer Res Treat 2017; 164: 1–11. [DOI] [PubMed] [Google Scholar]
  • 62. Zhong S, Zhang X, Chen L et al Statin use and mortality in cancer patients: Systematic review and meta‐analysis of observational studies. Cancer Treat Rev 2015; 41: 554–67. [DOI] [PubMed] [Google Scholar]
  • 63. McDonnell DP, Chang CY, Nelson ER. The estrogen receptor as a mediator of the pathological actions of cholesterol in breast cancer. Climacteric 2014; 17(Suppl 2): 60–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Borgquist SG‐HA, Ahern TP, Garber JE et al Cholesterol, cholesterol‐lowering medication use, and breast cancer outcome in the BIG 1‐98 study. J Clin Oncol 2017; 35: 1179–88. [DOI] [PubMed] [Google Scholar]
  • 65. Bjarnadottir O, Kimbung S, Johansson I et al Global transcriptional changes following statin treatment in breast cancer. Clin Cancer Res 2015; 21: 3402–11. [DOI] [PubMed] [Google Scholar]
  • 66. Kumar AS, Campbell M, Benz CC et al A call for clinical trials: lipophilic statins may prove effective in treatment and prevention of particular breast cancer subtypes. J Clin Oncol 2006; 24: 2127; author reply 27‐8. [DOI] [PubMed] [Google Scholar]
  • 67. Campbell MJ, Esserman LJ, Zhou Y et al Breast cancer growth prevention by statins. Cancer Res 2006; 66: 8707–14. [DOI] [PubMed] [Google Scholar]
  • 68. Wong WW, Clendening JW, Martirosyan A et al Determinants of sensitivity to lovastatin‐induced apoptosis in multiple myeloma. Mol Cancer Ther 2007; 6: 1886–97. [DOI] [PubMed] [Google Scholar]
  • 69. Clendening JW, Pandyra A, Li Z et al Exploiting the mevalonate pathway to distinguish statin‐sensitive multiple myeloma. Blood 2010; 115: 4787–97. [DOI] [PubMed] [Google Scholar]
  • 70. 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 2010; 119: 137–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Fiegenbaum M, da Silveira FR, Van der Sand CR et al The role of common variants of ABCB1, CYP3A4, and CYP3A5 genes in lipid‐lowering efficacy and safety of simvastatin treatment. Clin Pharmacol Ther 2005; 78: 551–8. [DOI] [PubMed] [Google Scholar]
  • 72. Shin EM, Sin Hay H, Lee MH et al DEAD‐box helicase DP103 defines metastatic potential of human breast cancers. J Clin Invest 2014; 124: 3807–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Liu J, Xu A, Lam KS et al Cholesterol‐induced mammary tumorigenesis is enhanced by adiponectin deficiency: role of LDL receptor upregulation. Oncotarget 2013; 4: 1804–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Yu R, Longo J, van Leeuwen JE et al Statin‐induced cancer cell death can be mechanistically uncoupled from prenylation of RAS family proteins. Cancer Res 2018; 78: 1347–57. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Internal Medicine are provided here courtesy of Wiley

RESOURCES