Next Article in Journal
Sagittal Balance Parameters and Proximal Junctional Kyphosis in Adolescent Idiopathic Scoliosis
Previous Article in Journal
Primary Radial Nerve Lesions in Humerus Shaft Fractures—Revision or Wait and See
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Rosuvastatin-Based Lipid-Lowering Therapy for the Control of LDL Cholesterol in Patients at High Vascular Risk

by
Jose María Mostaza
1 and
Carlos Escobar
2,*
1
Lipid and Vascular Risk Unit, Department of Internal, University Hospital La Paz-Carlos III, 28046 Madrid, Spain
2
Cardiology Department, University Hospital La Paz-Carlos III, 28046 Madrid, Spain
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(7), 1894; https://doi.org/10.3390/jcm13071894
Submission received: 13 February 2024 / Revised: 15 March 2024 / Accepted: 19 March 2024 / Published: 25 March 2024
(This article belongs to the Section Cardiovascular Medicine)

Abstract

:
Vascular diseases are the leading cause of death in Spain. Hypercholesterolemia is not only a cardiovascular risk factor, but also underlies the etiopathogenesis of atherosclerosis. Therefore, reducing LDL cholesterol (LDL-C) to the goals recommended by clinical practice guidelines, is essential to decrease the risk of vascular complications. Despite this, current LDL-C control is scarce, even in subjects with high and very high risk. This is mainly due to an insufficient intensification of lipid-lowering treatment. In this context, it is essential to prescribe the appropriate therapy, adjusted to patient’s needs based on their LDL-C and their vascular risk. Rosuvastatin, alone or in combination with ezetimibe, provides intensive LDL-C reductions (up to 50–55% and 60–75%, respectively), with a low risk of side effects and in an efficient manner, in patients both without and with established atherosclerotic vascular disease.

1. Introduction

Despite the continuous decrease in cardiovascular mortality over the last decades, vascular diseases remain the leading cause of death in Spain [1]. Thus, during the first semester of 2023, 27.1% of deaths were related to cardiovascular conditions, 25.8% to cancer and 11.5% to respiratory diseases [1]. The decrease in cardiovascular mortality has contributed significantly to the prolonging of the life expectancy of the Spanish population [1,2]. Thus, between 1980 and 2009, life expectancy increased by 6 years in Spain [2]. The decrease in vascular mortality contributed, by 63% in women and 53% in men, to this increase in longevity [2]. The factors that have motivated the decrease in coronary mortality in Spain have also been evaluated [3]. Between 1988 and 2005, coronary mortality decreased by 40%. Half of this decline was due to an improvement in the population’s cardiovascular risk factor profile, specifically, a reduction in cholesterol and blood pressure levels, a decrease in smoking and an increase in physical activity. However, there were opposing factors that increased its incidence, such as obesity and diabetes, and these negatively impacted on mortality. The other half of the decline was explained by the use of drugs in primary and, mainly, secondary prevention, and by better management during the acute coronary episode. These data are similar to those found in other countries, where, similarly, half of the benefits derived from the population’s improvement in cardiovascular risk factors and the other half from treatments used in the management of risk factors and cardiovascular disease [4]. In addition, a comprehensive approach is required in order to actually reduce cardiovascular disease. Thus, obesity is a recognized risk factor for the development of comorbid conditions such as cardiovascular disease and hypercholesterolemia. Achieving weight loss through a healthy lifestyle seems to be an ideal solution [5,6]. In addition, emerging risk factors, including sex-related factors (i.e., early menopause, gynecological tumors, etc.), should also be considered [7,8]. Given all the above, it seems evident that an optimal management of risk factors may have implications for the incidence of cardiovascular diseases and related mortality.
Hypercholesterolemia is not only a major, independent cardiovascular risk factor, but is also the underlying cause of atherosclerosis, particularly cholesterol that binds to low-density lipoproteins (LDL-C) and to apolipoprotein B-containing lipoproteins [9,10]. Clinical trials have shown that reducing LDL-C levels with lipid-lowering therapy is associated with a decrease in the risk of developing cardiovascular disease; the greater the reduction in LDL-C levels, the greater the protective effect against atherosclerotic cardiovascular disease [11]. In fact, there is no limit to the amount that LD-CL can be lowered while still reducing CV risk, and without a harmful effect [12]. Despite this, the control of hypercholesterolemia has been found to be suboptimal by all the studies that have evaluated this issue. In a review of 21 studies conducted between 2010 and 2014 that analyzed the rate of goal-attainment in patients in secondary prevention, the overall proportion of adequate control was only 31% [13].
As new drugs capable of reducing LDL-C concentrations to very low levels have become available, our knowledge of the relationship between cholesterol and vascular disease has improved and has motivated the downward modification of treatment goals. Those currently in force are the ones recommended by the European Society of Atherosclerosis (EAS) and the European Society of Cardiology (ESC) in their 2019 document [14] and by the European Society of Cardiology in their 2021 document [15]. These guidelines recommend the rapid attainment of LDL-C goals to maximize the preventive benefits [14,15].
The basis of the lipid-lowering treatment is statins. However, not all statins have demonstrated the same ability to reduce LDL-C, with atorvastatin and rosuvastatin having been found to be the most powerful statins for reducing it [14]. This review will focus on the importance of intensive rosuvastatin-based lipid-lowering therapy, researching its place in clinical practice.

2. Current Situation of Lipid Control

2.1. Europe

The SANTORINI study included more than 9000 patients at high and very high vascular risk from 14 European countries, including 990 subjects from Spain [16]. Overall, the percentage of patients who achieved LDL-C goals was 20.1%, with 24% among high-risk and 18.6% among very high-risk patients. The use of lipid-lowering drugs was suboptimal, with only 4.5% using proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), 24% combination therapy, and 21.8% not receiving any treatment. The mean LDL-C concentration in high-risk patients was 93 mg/dL, and in very high-risk patients it was 78 mg/dL (Figure 1) [16]. If these patients had achieved the recommended goals (<70 mg/dL and <55 mg/dL for patients at high and very high vascular risk, respectively), they would have decreased their LDL-C by an additional 23 mg/dL and would have avoided potential increases in vascular risk of between 12 and 15%. Unfortunately, these low rates of achievement of LDL-C deprived these patients of an additional benefit.

2.2. Spain

The dyslipidemia observatory was a study carried out exclusively in Spain by 435 physicians [17]. This study recruited 4010 patients from different clinical settings (34% from cardiology, 28% from primary care, 24% from internal medicine, and 14% from endocrinology). Overall, 31% had LDL-C levels within targets: 47%, 36%, 22% and 25% among low, moderate, high, and very high-risk patients, respectively. As in the SANTORINI study, the use of lipid-lowering therapies was suboptimal. Among the high and very high-risk participants, 7% and 1% did not receive any lipid-lowering treatment, 21% and 8% received low or moderate intensity statins, 44% and 38% received high intensity statins in monotherapy, 21% and 45% a combination of statins and ezetimibe, and 4% and 6% PCSK9i, respectively.
The SNAPSHOT study [18] included 443 patients with hypertension and dyslipidemia, with the aim of knowing how many of them were adequately controlled. The data were just as disappointing as in previous studies, with only 24% achieving the recommended targets, 16.7% among those at low or moderate risk, 21.8% in those at high risk, and 25.1% among those at very high risk. Furthermore, joint control of dyslipidemia and hypertension was only observed in 9% of the study population.
The low rate of goal achievement observed in previous studies represents a loss of opportunity for our patients. There is abundant evidence showing that the lower the cholesterol, the lower the rate of cardiovascular complications, without any threshold below which benefit is no longer observed and without an increase in the number of adverse effects [19]. Most studies on the benefit of treating hypercholesterolemia, however, have not focused specifically on the utility of achieving therapeutic goals. In the Treat Stroke to Target study [20], patients with a history of stroke were randomized to groups with an LDL-C goal of less than 70 mg/dL, the recommended goal for patients with vascular disease on the date the study began, or a less strict objective, aimed at maintaining LDL-C between 90 and 110 mg/dL. The arm that achieved the more strict objectives reduced the primary end point of ischemic stroke, acute myocardial infarction, new urgent coronary or carotid revascularization, or cardiovascular death by 22%, compared to the control group.
Remarkably, in all these studies, physicians erroneously perceived that their patients were better controlled than they were. Thus, in an epidemiological study carried out in Spain that included 300 primary care physicians [21], respondents considered that 61.5% of their patients were within LDL-C targets. This irrational optimism is perhaps also responsible for the systematic underestimation of risk. Although most of the patients who were recruited in the previous studies had a very high calculated vascular risk, the physician who enrolled them perceived their risk to be simply high. This fact had clinical relevance because underestimating the risk led to a less intensive search for objectives and contributed to obtaining fewer patients reaching the goal.
The results of these studies also showed that the use of combination lipid-lowering therapy was associated with a higher rate of goal achievement. In the SANTORINI study [16], 32% of patients receiving combination therapy were on target, compared to only 20.9% of patients receiving monotherapy treatment. Of note, combination treatment is associated with a greater decrease in cholesterol levels, as well as in the rate of cardiovascular complications [22]. This fact is even more relevant if the combination treatment is received in a single pill. In a retrospective analysis [23], it was observed that patients receiving combination treatment with statins and ezetimibe reduced their cholesterol levels more if both drugs were combined in a single tablet than if they were taken separately.

3. Role of Rosuvastatin in the Control of Cholesterol and the Reduction of Vascular Events

3.1. Efficacy

Rosuvastatin is a relatively hydrophilic, potent and highly selective statin for the enzyme 3-hydroxy-3-methylglutaryl-CoA reductase which significantly reduces cholesterol synthesis. Rosuvastatin undergoes limited metabolism, as approximately only 10% is metabolized by cytochrome P450, which translates into a low risk of interactions with other drugs. About 90% of the rosuvastatin is excreted unchanged in the feces and the remaining 10% in the urine. Age has no relevant impact on the pharmacokinetics of rosuvastatin, nor does kidney disease, except in the case of severe renal insufficiency, which markedly increases exposure to rosuvastatin [24,25].
Several clinical trials have analyzed the effects of rosuvastatin on LDL-C. In particular, the STELLAR study showed that it was the most powerful statin, since, after 6 weeks of treatment, rosuvastatin 10–40 mg was able to reduce LDL-C by 46–55%, compared to 37–51% with atorvastatin 10–80 mg, 28–39% with simvastatin 10–40 mg, and 20–30% with pravastatin 10–40 mg. Likewise, rosuvastatin increased HDL cholesterol by 8–10% (vs. 2–6%, 5% and 3–6%, respectively), and decreased triglyceride levels by 20–26% (vs. 20–28%, 12–15% and 8–13%, respectively) [26]. These results were confirmed in the VOYAGER study, a pooled analysis based on data from 32,258 individual patients of studies comparing the efficacy of rosuvastatin with that of atorvastatin or simvastatin [27]. A meta-analysis of 50 studies, with a total of 51,956 patients, analyzed the effectiveness of different statins in reducing LDL-C levels, with rosuvastatin being found the most potent statin [28]. This superiority has been confirmed in a more recent meta-analysis [29]. Nonetheless, rosuvastatin dosage reductions have been recommended in Asian patients, as it has been observed in pharmacokinetic studies that there is a two-fold increase in median exposure to rosuvastatin in Asian subjects when compared to Caucasian individuals [30,31]. On the other hand, the incidence of side effects seems lower with rosuvastatin than with atorvastatin [32], a finding also reached in real-life studies [33]. Likewise, rosuvastatin has not only demonstrated direct positive effects on the lipid profile, but could have additional benefits, due to its anti-inflammatory, antioxidant, antithrombotic and vascular protective properties, among other aspects [34,35,36,37,38].
The role of rosuvastatin in the prevention of vascular complications across the entire spectrum of patients with hypercholesterolemia has been evaluated in several studies. One of the most important was the JUPITER clinical trial [39], a phase III study, which included 17,802 subjects with LDL-C < 130 mg/dL, high-sensitivity C-reactive protein ≥ 2.0 mg/L, and without known cardiovascular disease. That is, these were patients without indications for treatment with statins at the time the study was carried out. Patients were randomized to receive rosuvastatin 20 mg or placebo. The study was stopped prematurely, after a follow-up of only 1.9 years, due to the beneficial effects of rosuvastatin in reducing events. LDL-C reductions with rosuvastatin were around 50%, in line with previous studies. Additionally, there was a 37% decrease in high-sensitivity C-reactive protein levels during the study, confirming the anti-inflammatory properties of rosuvastatin. Compared to placebo, rosuvastatin 20 mg significantly reduced the risk of the primary composite endpoint (myocardial infarction, stroke, arterial revascularization, hospitalization for unstable angina, and cardiovascular death) by 44% (projected 5-year NNT of 25) [40], as well as that of MACE by 47%. Of note, the JUPITER study is one of the few clinical trials with lipid-lowering treatments that has demonstrated a significant reduction in mortality from any cause (Figure 2) [39]. The results of the JUPITER study were consistent in the different subgroups of patients studied, and were independent of age, sex, the patient’s baseline cardiovascular risk, and the presence of diabetes or chronic kidney disease, emphasizing the benefits of rosuvastatin across the spectrum of patients with dyslipidemia [41,42,43,44,45,46]. Similarly, the HOPE-3 study found, in 12,705 patients without previous cardiovascular disease but with an intermediate cardiovascular risk, that after a median follow-up of 5.6 years, compared to placebo, the addition of rosuvastatin 10 mg to the treatment was associated with a significant reduction of 24% (HR 0.76; 95% CI 0.64–0.91; p = 0.002) in the risk of MACE (cardiovascular death, myocardial infarction or stroke); of 25% in the composite secondary variable of cardiovascular death, myocardial infarction, stroke, revascularization, heart failure or recovered cardiac arrest (HR 0.75; 95% CI 0.64–0.88; p < 0.001); 35% in the risk of myocardial infarction (HR 0.65; 95% CI 0.44–0.94); 30% in the risk of stroke (HR 0.70; 95% CI 0.52–0.95) and 32% in the need for revascularization (HR 0.68; 95% CI 0.48–0.95). Furthermore, the results were consistent within the different subgroups analyzed, including baseline cardiovascular risk, as well as lipid levels, C-reactive protein and blood pressure [47].
The role of rosuvastatin has been analyzed not only in patients in primary prevention, but different clinical trials and real-life studies have also been developed in subjects with established atherosclerotic vascular disease [25]. Thus, different clinical trials have shown that rosuvastatin improves the lipid profile (reduction of LDL-C and increase of HDL cholesterol) in this population, even to a greater extent than other statins, including atorvastatin in some of them [48,49,50,51,52,53,54,55,56]. Likewise, it has also been observed that treatment with rosuvastatin can reduce the volume of and stabilize atherosclerotic plaque, in addition to reducing the progression of carotid intima-media thickness and inflammation, as well as ventricular remodeling and myocardial fibrosis [49,50,51,52,53,55,57,58,59]. These positive results translate into a lower risk of recurrence of vascular events, as has been shown in different studies. Thus, in a recent real-life study of patients who had had an acute coronary syndrome, after one year of follow-up, the number of MACE, as well as its separate components, was found to be low and similar between patients treated with rosuvastatin and atorvastatin [60]. A similar trend has been observed in the LODESTAR clinical trial, in 4400 subjects with coronary artery disease who were followed for 3 years [61]. These results have also been observed in studies specifically performed in the Spanish population. Thus, in a nationwide retrospective and multicenter study, after a mean follow-up of 3 years, the recurrence rate of atherosclerotic vascular events was 2.73 cases/100 person-years for patients treated with atorvastatin vs. 2.34 cases/100 person-years for those treated with rosuvastatin, with no statistically significant differences between the two groups [62]. In the SAFEHEART study, which analyzed 1939 patients with familial hypercholesterolemia followed prospectively in Spain, after a median follow-up of 6.6 years, the incidence of atherosclerotic vascular events was similar between patients treated with atorvastatin and rosuvastatin (1.1 vs. 1.2 events/100 patient-years, respectively; p = 0.58) [63]. In summary, rosuvastatin has also been shown to be an effective and safe statin in patients with established atherosclerotic vascular disease [64].

3.2. Safety/Interactions

Clinical trials have shown that rosuvastatin is a very safe statin, with a very low risk of adverse events and drug interactions. Thus, for example, in the JUPITER study, rosuvastatin did not increase the risk of myopathy or cancer, and although in the HOPE-3 study there were more cases of muscle weakness or pain with rosuvastatin, the rates of discontinuations due to muscle symptoms, cases of rhabdomyolysis and myopathy were similar to those of the placebo group.
In the JUPITER study there was an increased risk of physician-reported diabetes, which was not the case in the HOPE-3 study. Additionally, in the JUPITER study, HbA1c increased from 5.7% to 5.9% with rosuvastatin (vs. 5.8% with placebo). Consequently, although the risk of diabetes was statistically increased with rosuvastatin in the JUPITER study, considering the robust reduction in vascular events, overall, it could be concluded that this increase was of little clinical relevance [39,47,65]. On the other hand, despite the fact that an increased incidence of proteinuria with high doses or rosuvastatin, particularly 40 mg/day, has been reported, this is a rare complication, with a good kidney prognosis, and normalizing observed after withdrawal of rosuvastatin [66].
Finally, since many patients with dyslipidemia have other associated cardiovascular risk factors and comorbidities, they are frequently polymedicated, which increases the risk of drug–drug interactions and potential side effects. In this context, unlike what happens with other statins such as atorvastatin, simvastatin and lovastatin, which are metabolized by the 3A4 isoform of cytochrome P450 (CYP3A4), rosuvastatin differs in its metabolization, which greatly reduces the risk of pharmacological interactions, and therefore confers greater safety, especially in polymedicated patients. This is important because numerous frequently used drugs such as verapamil, diltiazem, nifedipine, midazolam, alprazolam, erythromycin, fluoxetine, sertraline, amiodarone, mirtazapine, esomeprazole, omeprazole, digoxin and warfarin, among others, are metabolized through this route. Nonetheless, since levels of rosuvastatin markedly increase with the concomitant use of ciclosporin, co-administration of both drugs is contraindicated. Similarly, concomitant protease inhibitor use is not recommended due to the increase in rosuvastatin exposure with concomitant treatment [24,25].

3.3. Cost-Effectiveness

It is not only important to reduce LDL-C levels and vascular complications, but also to be efficient in the use of these therapies, that is, to ensure the greatest benefit with the lowest cost. In this sense, both rosuvastatin monotherapy and the combination of rosuvastatin and ezetimibe have been shown to be dominant treatments (more effective and less expensive) in a high proportion of patients, especially in those with a higher vascular risk (Table 1) [67,68]. Therefore, rosuvastatin is a high-intensity statin, which may contribute to the sustainability of the health care system [69].

4. Role of Rosuvastatin-Based Combination Lipid-Lowering Therapy

The IMPROVE-IT study demonstrated in subjects hospitalized for acute coronary syndrome that the combination of a statin with ezetimibe was more effective than statin monotherapy, not only in reducing LDL-C, but also in reducing recurrences of atherosclerotic vascular events, without increasing the risk of adverse effects [70]. Furthermore, the addition of ezetimibe to statins is much more effective than doubling the dose of statins in reducing LDL-C levels. This is not surprising, since statins and ezetimibe have complementary mechanisms of action (reduction of cholesterol synthesis and reduction of cholesterol absorption, respectively) that enhance their lipid-lowering action [24,25,71,72]. Thus, while potent statins at maximum doses achieve approximately an average reduction in LDL-C of around 50–55%, the decrease in LDL-C with the combination of potent statins and ezetimibe reaches approximately 60–75% [73].
Different clinical trials have specifically been developed to analyze the effects of the combination of rosuvastatin and ezetimibe on LDL-C levels and the achievement of lipid control objectives compared to rosuvastatin in monotherapy or other combinations of statins with ezetimibe in patients with hypercholesterolemia, mainly in subjects with a high vascular risk. In all of them, greater reductions in LDL-C and a greater degree of achievement of the objectives set by the clinical practice guidelines have been observed with the combination of rosuvastatin and ezetimibe, without increasing the risk of side effects (Table 2) [74,75,76,77,78,79,80,81].
Consequently, in those patients who require LDL-C reductions above 50%, the combination of rosuvastatin and ezetimibe offers additional reductions in LDL-C and a higher control rate [25,82,83,84].

5. Discussion

Hypercholesterolemia is directly involved in the etiopathogenesis of atherosclerosis [14]. Despite this, the percentage of patients whose LDL-C levels are within therapeutic targets is very low, including patients with a higher vascular risk [16,17,18]. Although several reasons have been proposed, it seems that underestimation of the risk, as well as an inadequate perception of actual LDL-C control, play essential roles, since they lead to insufficient intensification of lipid-lowering treatment, which is key to the improvement of these figures [17,21,85].
In this context, rosuvastatin, a high intensity statin that provides marked reductions in LDL-C levels, significantly reduces the risk of cardiovascular events and death, as the JUPITER trial demonstrated [39], in addition to its efficacy in secondary prevention patients, although evidence in these patients is less strong [48,49,50,51,52,53,54,55,56]. On the other hand, despite the fact that an increased risk of diabetes has been observed among predisposed patients, this is not clinically relevant, due to the cardiovascular benefits of rosuvastatin; particular attention should be paid, however, with these patients [39,65]. Moreover, the addition of ezetimibe may be helpful in this context, as lower doses of rosuvastatin may be required to attain LDL-C, thus, reducing the risk of developing diabetes [25,65].
From a clinical point of view, the step-by-step lipid-lowering treatment approach recommended by the European guidelines (starting with statins up to the maximum tolerated doses, if the objectives are not achieved, adding ezetimibe, and if this is not sufficient, adding PCSK9i) facilitates therapeutic inertia and delays goal achievement [14]. Different scientific societies, such as the Spanish Society of Cardiology and the Spanish Society of Arteriosclerosis, have advocated establishing an individualized lipid-lowering treatment strategy from the beginning, one aimed at achieving the required LDL-C target without delay. That is, given that the average reduction in LDL-C produced by each lipid-lowering therapy, alone or in combination, is already known, as well as the LDL-C levels and the LDL-C target required in each patient, it is possible to estimate the precise treatment for each subject (i.e., statins in monotherapy, combination of statins and ezetimibe, etc.) [73]. The application of this initial individualized approach can improve the degree of LDL-C control, even in patients at high/very high vascular risk [86]. In this context, rosuvastatin is a high-intensity statin which produces average reductions in LDL-C of around 50–55%, and when combined with ezetimibe, reductions of up to 60–75% [73], which undoubtedly contributes to the improvement of LDL-C control in our patients. In the light of evidence, and with the aim of “the earlier the better”, guidelines should clarify which patients would benefit more from monotherapy or combined therapy, particularly those patients at higher risk. In addition, future research is needed to clarify whether this approach will translate into a reduction of cardiovascular events compared to the traditional approach, as well as the role of rosuvastatin in this setting.

6. Conclusions

Reducing LDL-C to the recommended targets is essential for decreasing the risk of vascular complications. Unfortunately, the current control figures are very poor. To improve LDL-C control, especially in high/very high-risk patients, it is essential to prescribe the appropriate lipid-lowering treatment, adjusting to the specific needs of each patient. In these patients, rosuvastatin, alone or combined with ezetimibe, provides intensive reductions in LDL-C, with a low risk of side effects and a lower cost, which is associated with a lower risk of vascular complications, both in patients without previous vascular disease, and in those with established atherosclerotic vascular disease.

Funding

This study received funding from Almirall S.A. The funder was not involved in the study design, collection, analysis, interpretation of data, the writing of this article or the decision to submit it for publication.

Conflicts of Interest

J.M.M. has received honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Almirall, Amgen, Sanofi, Novartis, Daichi Sankyo and C.E. has received honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Almirall, Amgen, Novartis, Sanofi, Daichii Sankyo, Viatris.

References

  1. Deaths According to Cause. Statistics National Institute. Available online: https://www.ine.es/dyngs/INEbase/es/operacion.htm?c=Estadistica_C&cid=1254736176780&menu=ultiDatos&idp=1254735573175 (accessed on 11 March 2024).
  2. García González, J.M. Contributions of cardiovascular mortality to Spanish life expectancy from 1980 to 2009. Rev. Esp. Cardiol. 2013, 66, 848–853. [Google Scholar] [CrossRef] [PubMed]
  3. Flores-Mateo, G.; Grau, M.; O’Flaherty, M.; Ramos, R.; Elosua, R.; Violan-Fors, C.; Quesada, M.; Martí, R.; Sala, J.; Marrugat, J.; et al. Analyzing the coronary heart disease mortality decline in a Mediterranean population: Spain 1988–2005. Rev. Esp. Cardiol. 2011, 64, 988–996. [Google Scholar] [CrossRef]
  4. Ford, E.S.; Ajani, U.A.; Croft, J.B.; Critchley, J.A.; Labarthe, D.R.; Kottke, T.E.; Giles, W.H.; Capewell, S. Explaining the decrease in U.S. deaths from coronary disease, 1980–2000. N. Engl. J. Med. 2007, 356, 2388–2398. [Google Scholar] [CrossRef] [PubMed]
  5. Mulita, F.; Lampropoulos, C.; Kehagias, D.; Verras, G.I.; Tchabashvili, L.; Kaplanis, C.; Liolis, E.; Iliopoulos, F.; Perdikaris, I.; Kehagias, I. Long-term nutritional deficiencies following sleeve gastrectomy: A 6-year single-centre retrospective study. Prz. Menopauzalny 2021, 20, 170–176. [Google Scholar] [CrossRef] [PubMed]
  6. Verras, G.I.; Mulita, F.; Pouwels, S.; Parmar, C.; Drakos, N.; Bouchagier, K.; Kaplanis, C.; Skroubis, G. Outcomes at 10-Year Follow-Up after Roux-en-Y Gastric Bypass, Biliopancreatic Diversion, and Sleeve Gastrectomy. J. Clin. Med. 2023, 12, 4973. [Google Scholar] [CrossRef] [PubMed]
  7. Mulita, F.; Leivaditis, V.; Dimopoulos, P.; Ibra, A.; Iliopoulos, F.; Tasios, K.; Pitros, C.; Kaplanis, C.; Peteinaris, A.; Bouchagier, K.; et al. Correlation between gynecological tumors and atherosclerotic diseases. Arch. Med. Sci. Atheroscler. Dis. 2023, 8, e118–e122. [Google Scholar] [CrossRef] [PubMed]
  8. Pakhare, M.; Anjankar, A. Critical Correlation Between Obesity and Cardiovascular Diseases and Recent Advancements in Obesity. Cureus 2024, 16, e51681. [Google Scholar] [CrossRef]
  9. Boekholdt, S.M.; Arsenault, B.J.; Mora, S.; Pedersen, T.R.; LaRosa, J.C.; Nestel, P.J.; Simes, R.J.; Durrington, P.; Hitman, G.A.; Welch, K.M.; et al. Association of LDL cholesterol, non-HDL cholesterol, and apolipoprotein B levels with risk of cardiovascular events among patients treated with statins: A meta-analysis. JAMA 2012, 307, 1302–1309. [Google Scholar] [CrossRef]
  10. Ference, B.A.; Ginsberg, H.N.; Graham, I.; Ray, K.K.; Packard, C.J.; Bruckert, E.; Hegele, R.A.; Krauss, R.M.; Raal, F.J.; Schunkert, H.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2017, 38, 2459–2472. [Google Scholar] [CrossRef]
  11. Boekholdt, S.M.; Hovingh, G.K.; Mora, S.; Arsenault, B.J.; Amarenco, P.; Pedersen, T.R.; LaRosa, J.C.; Waters, D.D.; DeMicco, D.A.; Simes, R.J.; et al. Very low levels of atherogenic lipoproteins and the risk for cardiovascular events: A meta-analysis of statin trials. J. Am. Coll. Cardiol. 2014, 64, 485–494. [Google Scholar] [CrossRef]
  12. Masana, L.; Girona, J.; Ibarretxe, D.; Rodríguez-Calvo, R.; Rosales, R.; Vallvé, J.C.; Rodríguez-Borjabad, C.; Guardiola, M.; Rodríguez, M.; Guaita-Esteruelas, S.; et al. Clinical and pathophysiological evidence supporting the safety of extremely low LDL levels-The zero-LDL hypothesis. J. Clin. Lipidol. 2018, 12, 292–299. [Google Scholar] [CrossRef]
  13. de la Sierra, A.; Pintó, X.; Guijarro, C.; Miranda, J.L.; Callejo, D.; Cuervo, J.; Subirà, R.; Rubio, M. Prevalence, Treatment, and Control of Hypercholesterolemia in High Cardiovascular Risk Patients: Evidences from a Systematic Literature Review in Spain. Adv. Ther. 2015, 32, 944–961. [Google Scholar] [CrossRef]
  14. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef]
  15. Visseren, F.L.J.; Mach, F.; Smulders, Y.M.; Carballo, D.; Koskinas, K.C.; Bäck, M.; Benetos, A.; Biffi, A.; Boavida, J.M.; Capodanno, D.; et al. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice. Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef] [PubMed]
  16. Ray, K.K.; Haq, I.; Bilitou, A.; Manu, M.C.; Burden, A.; Aguiar, C.; Arca, M.; Connolly, D.L.; Eriksson, M.; Ferrières, J.; et al. Treatment gaps in the implementation of LDL cholesterol control among high- and very high-risk patients in Europe between 2020 and 2021: The multinational observational SANTORINI study. Lancet Reg. Health Eur. 2023, 29, 100624. [Google Scholar] [CrossRef]
  17. Cosín-Sales, J.; Campuzano Ruiz, R.; Díaz Díaz, J.L.; Escobar Cervantes, C.; Fernández Olmo, M.R.; Gómez-Doblas, J.J.; Mostaza, J.M.; Pedro-Botet, J.; Plana Gil, N.; Valdivielso, P. Dyslipidemia observatory: Treatment of hypercholesterolemia in Spain, context and levers for improvement in clinical practice. Clin. Investig. Arterioscler. 2022, 34, 253–260. [Google Scholar] [CrossRef] [PubMed]
  18. SNAPSHOT study. Cross-sectional multinational epidemiological study of hypertensive patients with dyslipidemia and patient-reported outcomes. In Proceedings of the 28th SEH-LELHA 2023 Congress, Madrid, Spain, 28–29 September 2023.
  19. Fulcher, J.; O’Connell, R.; Voysey, M.; Emberson, J.; Blackwell, L.; Mihaylova, B.; Simes, J.; Collins, R.; Kirby, A.; Colhoun, H.; et al. Efficacy and safety of LDL-lowering therapy among men and women: Meta-analysis of individual data from 174,000 participants in 27 randomised trials. Lancet 2015, 385, 1397–1405. [Google Scholar] [PubMed]
  20. Amarenco, P.; Kim, J.S.; Labreuche, J.; Charles, H.; Abtan, J.; Béjot, Y.; Cabrejo, L.; Cha, J.K.; Ducrocq, G.; Giroud, M.; et al. A Comparison of Two LDL Cholesterol Targets after Ischemic Stroke. N. Engl. J. Med. 2020, 382, 9. [Google Scholar] [CrossRef] [PubMed]
  21. Pallarés-Carratalá, V.; Barrios, V.; Fierro-González, D.; Polo-García, J.; Cinza-Sanjurjo, S. Cardiovascular Risk in Patients with Dyslipidemia and Their Degree of Control as Perceived by Primary Care Physicians in a Survey-TERESA-Opinion Study. Int. J. Environ. Environ. Res. Public Health 2023, 20, 2388. [Google Scholar] [CrossRef] [PubMed]
  22. Khunti, K.; Danese, M.D.; Kutikova, L.; Catterick, D.; Sorio-Vilela, F.; Gleeson, M.; Kondapally Seshasai, S.R.; Brownrigg, J.; Ray, K.K. Association of a Combined Measure of Adherence and Treatment Intensity with Cardiovascular Outcomes in Patients with Atherosclerosis or Other Cardiovascular Risk Factors Treated with Statins and/or Ezetimibe. JAMA Netw. Open 2018, 1, e185554. [Google Scholar] [CrossRef] [PubMed]
  23. Katzmann, J.L.; Sorio-Vilela, F.; Dornstauder, E.; Fraas, U.; Smieszek, T.; Zappacosta, S.; Laufs, U. Non-statin lipid-lowering therapy over time in very-high-risk patients: Effectiveness of fixed-dose statin/ezetimibe compared to separate pill combination on LDL-C. Clin. Res. Cardiol. 2022, 111, 243–252. [Google Scholar] [CrossRef] [PubMed]
  24. Rosuvastatin and Ezetimibe. Summary of Product Characteristics. Available online: https://www.hpra.ie/img/uploaded/swedocuments/Licence_PA2010-019-001_24082021170348.pdf (accessed on 16 January 2024).
  25. Barrios, V.; Escobar, C. Fixed-dose combination of rosuvastatin and ezetimibe: Treating hypercholesteremia according to cardiovascular risk. Expert Rev. Clin. Pharmacol. 2021, 14, 793–806. [Google Scholar] [CrossRef]
  26. Jones, P.H.; Davidson, M.H.; Stein, E.A.; Bays, H.E.; McKenney, J.M.; Miller, E.; Cain, V.A.; Blasetto, J.W. Comparison of the efficacy and safety of rosuvastatin vs. atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial). Am. J. Cardiol. 2003, 92, 152–160. [Google Scholar] [CrossRef]
  27. Nicholls, S.J.; Brandrup-Wognsen, G.; Palmer, M.; Barter, P.J. Meta-analysis of comparative efficacy of increasing dose of atorvastatin vs. rosuvastatin vs. simvastatin on lowering levels of atherogenic lipids (from VOYAGER). Am. J. Cardiol. 2010, 105, 69–76. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, X.; Xing, L.; Jia, X.; Pang, X.; Xiang, Q.; Zhao, X.; Ma, L.; Liu, Z.; Hu, K.; Wang, Z.; et al. Comparative Lipid-Lowering/Increasing Efficacy of 7 Statins in Patients with Dyslipidemia, Cardiovascular Diseases, or Diabetes Mellitus: Systematic Review and Network Meta-Analyses of 50 Randomized Controlled Trials. Cardiovasc. Ther. 2020, 2020, 3987065. [Google Scholar] [CrossRef] [PubMed]
  29. Jaam, M.; Al-Naimi, H.N.; Haddad, M.M.; Abushanab, D.; Al-Badriyeh, D. Comparative efficacy and safety among high-intensity statins. Syst. Rev. Meta-Anal. J. Comp. Eff. Res. 2023, 12, e220163. [Google Scholar] [CrossRef]
  30. Tse, M.L. Cluster of cases of high-dose rosuvastatin-associated rhabdomyolysis and recent reduction of rosuvastatin dose for Asians in other countries. Hong Kong Med. J. 2023, 29, 474. [Google Scholar] [CrossRef]
  31. Wu, H.F.; Hristeva, N.; Chang, J.; Liang, X.; Li, R.; Frassetto, L.; Benet, L.Z. Rosuvastatin Pharmacokinetics in Asian and White Subjects Wild Type for Both OATP1B1 and BCRP Under Control and Inhibited Conditions. J. Pharm. Sci. 2017, 106, 2751–2757. [Google Scholar] [CrossRef]
  32. Brewer, H.B. Benefit-Risk Assessment of Rosuvastatin 10 to 40 Milligrams. Am. J. Cardiol. 2003, 92, 23K–29K. [Google Scholar] [CrossRef]
  33. Stein, B.; Ward, T.; Hale, G.; Lyver, E. Safety of High-Intensity Statins in the Veteran Population: Atorvastatin 40 to 80 mg Compared with Rosuvastatin 20 to 40 mg. Ann. Pharmacother. 2020, 54, 405–413. [Google Scholar] [CrossRef]
  34. Cortese, F.; Gesualdo, M.; Cortese, A.; Carbonara, S.; Devito, F.; Zito, A.; Ricci, G.; Scicchitano, P.; Ciccone, M.M. Rosuvastatin: Beyond the cholesterol-lowering effect. Pharmacol. Res. 2016, 107, 1–18. [Google Scholar] [CrossRef] [PubMed]
  35. Zhao, Z.; Wang, X.; Lu, M.; Gao, Y. Rosuvastatin Improves Endothelial Dysfunction in Diabetes by Normalizing Endoplasmic Reticulum Stress via Calpain-1 Inhibition. Curr. Pharm. Des. 2023, 29, 2579–2590. [Google Scholar] [CrossRef]
  36. Tsilimigras, D.I.; Thanopoulou, K.; Salagianni, M.; Siasos, G.; Oikonomou, E.; Perrea, D.D.; Nirakis, N.; Filis, K.; Tsioufis, K.; Tousoulis, D.; et al. Rosuvastatin Attenuates Progression of Atherosclerosis and Reduces Serum IL6 and CCL2 Levels in Apolipoprotein-E-deficient Mice. In Vivo 2023, 37, 994–1002. [Google Scholar] [CrossRef]
  37. Joseph, P.; Glynn, R.; Lonn, E.; Ramasundarahettige, C.; Eikelboom, J.; MacFadyen, J.; Ridker, P.; Yusuf, S. Rosuvastatin for the prevention of venous thromboembolism: A pooled analysis of the HOPE-3 and JUPITER randomized controlled trials. Cardiovasc. Res. 2022, 118, 897–903. [Google Scholar] [CrossRef]
  38. Vavlukis, A.; Vavlukis, M.; Mladenovska, K.; Dimovski, A.; Muñoz-García, N.; de Santisteban Villaplana, V.; Padro, T.; Badimon, L. Antioxidative Effects of Rosuvastatin in Low-to-Moderate Cardiovascular Risk Subjects. Prilozi 2022, 43, 65–75. [Google Scholar] [CrossRef] [PubMed]
  39. Ridker, P.M.; Danielson, E.; Fonseca, F.A.; Genest, J.; Gotto, A.M., Jr.; Kastelein, J.J.; Koenig, W.; Libby, P.; Lorenzatti, A.J.; MacFadyen, J.G.; et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N. Engl. J. Med. 2008, 359, 2195–2207. [Google Scholar] [CrossRef]
  40. Ridker, P.M.; MacFadyen, J.G.; Fonseca, F.A.; Genest, J.; Gotto, A.M.; Kastelein, J.J.; Koenig, W.; Libby, P.; Lorenzatti, A.J.; Nordestgaard, B.G.; et al. Number needed to treat with rosuvastatin to prevent first cardiovascular events and death among men and women with low low-density lipoprotein cholesterol and elevated high-sensitivity C-reactive protein: Justification for the use of statins in prevention: An intervention trial evaluating rosuvastatin (JUPITER). Circ. Cardiovasc. Qual. Outcomes 2009, 2, 616–623. [Google Scholar]
  41. Glynn, R.J.; Koenig, W.; Nordestgaard, B.G.; Shepherd, J.; Ridker, P.M. Rosuvastatin for primary prevention in older persons with elevated C-reactive protein and low to average low-density lipoprotein cholesterol levels: Exploratory analysis of a randomized trial. Ann. Intern. Med. 2010, 152, 488–496. [Google Scholar] [CrossRef] [PubMed]
  42. Everett, B.M.; Glynn, R.J.; MacFadyen, J.G.; Ridker, P.M. Rosuvastatin in the prevention of stroke among men and women with elevated levels of C-reactive protein: Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin JUPITER. Circulation 2010, 121, 143–150. [Google Scholar] [CrossRef]
  43. Ridker, P.M.; Macfadyen, J.G.; Nordestgaard, B.G.; Koenig, W.; Kastelein, J.J.; Genest, J.; Glynn, R.J. Rosuvastatin for primary prevention among individuals with elevated high-sensitivity c-reactive protein and 5% to 10% and 10% to 20% 10-year risk. Implications of the Justification for Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER) trial for “intermediate risk”. Circ. Cardiovasc. Qual. Outcomes 2010, 3, 447–452. [Google Scholar]
  44. Koenig, W.; Ridker, P.M. Rosuvastatin for primary prevention in patients with European systematic coronary risk evaluation risk = 5% or Framingham risk >20%: Post hoc analyses of the JUPITER trial requested by European health authorities. Eur. Heart J. 2011, 32, 75–83. [Google Scholar] [CrossRef]
  45. Ridker, P.M.; Pradhan, A.; MacFadyen, J.G.; Libby, P.; Glynn, R.J. Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: An analysis from the JUPITER trial. Lancet 2012, 380, 565–571. [Google Scholar] [CrossRef]
  46. Ridker, P.M.; MacFadyen, J.; Cressman, M.; Glynn, R.J. Efficacy of rosuvastatin among men and women with moderate chronic kidney disease and elevated high-sensitivity C-reactive protein: A secondary analysis from the JUPITER (Justification for the Use of Statins in Prevention-an Intervention Trial Evaluating Rosuvastatin) trial. J. Am. Coll. Cardiol. 2010, 55, 1266–1273. [Google Scholar] [PubMed]
  47. Yusuf, S.; Bosch, J.; Dagenais, G.; Zhu, J.; Xavier, D.; Liu, L.; Pais, P.; López-Jaramillo, P.; Leiter, L.A.; Dans, A.; et al. HOPE-3 Investigators. Cholesterol lowering in intermediate-risk persons without Cardiovascular Disease. N. Engl. J. Med. 2016, 374, 2021–2031. [Google Scholar] [CrossRef] [PubMed]
  48. Clearfield, M.B.; Amerena, J.; Bassand, J.P.; Hernández García, H.R.; Miller, S.S.; Sosef, F.F.; Palmer, M.K.; Bryzinski, B.S. Comparison of the efficacy and safety of rosuvastatin 10 mg and atorvastatin 20 mg in high-risk patients with hypercholesterolemia–Prospective study to evaluate the Use of Low doses of the Statins Atorvastatin and Rosuvastatin (PULSAR). Trials 2006, 7, 35. [Google Scholar] [CrossRef] [PubMed]
  49. Nissen, S.E.; Nicholls, S.J.; Sipahi, I.; Libby, P.; Raichlen, J.S.; Ballantyne, C.M.; Davignon, J.; Erbel, R.; Fruchart, J.C.; Tardif, J.C.; et al. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: The ASTEROID trial. JAMA 2006, 295, 1556–1565. [Google Scholar] [CrossRef] [PubMed]
  50. Crouse, J.R., 3rd; Raichlen, J.S.; Riley, W.A.; Evans, G.W.; Palmer, M.K.; O’Leary, D.H.; Grobbee, D.E.; Bots, M.L. Effect of rosuvastatin on progression of carotid intima-media thickness in low-risk individuals with subclinical atherosclerosis: The METEOR Trial. JAMA 2007, 297, 1344–1353. [Google Scholar] [CrossRef] [PubMed]
  51. Underhill, H.R.; Yuan, C.; Zhao, X.Q.; Kraiss, L.W.; Parker, D.L.; Saam, T.; Chu, B.; Takaya, N.; Liu, F.; Polissar, N.L.; et al. Effect of rosuvastatin therapy on carotid plaque morphology and composition in moderately hypercholesterolemic patients: A high-resolution magnetic resonance imaging trial. Am. Heart J. 2008, 155, 584. [Google Scholar] [CrossRef] [PubMed]
  52. Takayama, T.; Hiro, T.; Yamagishi, M.; Daida, H.; Hirayama, A.; Saito, S.; Yamaguchi, T.; Matsuzaki, M. Effect of rosuvastatin on coronary atheroma in stable coronary artery disease: Multicenter coronary atherosclerosis study measuring effects of rosuvastatin using intravascular ultrasound in Japanese subjects (COSMOS). Circ. J. 2009, 73, 2110–2117. [Google Scholar] [CrossRef]
  53. Hall, A.S.; Jackson, B.M.; Farrin, A.J.; Efthymiou, M.; Barth, J.H.; Copeland, J.; Bailey, K.M.; Romaine, S.P.; Balmforth, A.J.; McCormack, T.; et al. A randomized, controlled trial of simvastatin vs. rosuvastatin in patients with acute myocardial infarction: The secondary prevention of acute coronary events–reduction of cholesterol to key European targets trial. Eur. J. Cardiovasc. Prev. Rehabil. 2009, 16, 712–721. [Google Scholar] [CrossRef]
  54. Lablanche, J.M.; Leone, A.; Merkely, B.; Morais, J.; Alonso, J.; Santini, M.; Eha, J.; Demil, N.; Licour, M.; Tardif, J.C. Comparison of the efficacy of rosuvastatin vs. atorvastatin in reducing apolipoprotein B/apolipoprotein A-1 ratio in patients with acute coronary syndrome: Results of the CENTAURUS study. Arch. Cardiovasc. Dis. 2010, 103, 160–169. [Google Scholar] [CrossRef] [PubMed]
  55. Nicholls, S.J.; Ballantyne, C.M.; Barter, P.J.; Chapman, M.J.; Erbel, R.M.; Libby, P.; Raichlen, J.S.; Uno, K.; Borgman, M.; Wolski, K.; et al. Effect of two intensive statin regimens on progression of coronary disease. N. Engl. J. Med. 2011, 365, 2078–2087. [Google Scholar] [CrossRef] [PubMed]
  56. Pitt, B.; Loscalzo, J.; Monyak, J.; Miller, E.; Raichlen, J. Comparison of lipid-modifying efficacy of rosuvastatin vs. atorvastatin in patients with acute coronary syndrome (from the LUNAR study). Am. J. Cardiol. 2012, 109, 1239–1246. [Google Scholar] [CrossRef] [PubMed]
  57. Luo, R.; Sun, X.; Shen, F.; Hong, B.; Wang, Z. Effects of high-dose rosuvastatin on ventricular remodelling and cardiac function in ST-segment elevation Myocardial infarction. Drug Des. Dev. Ther. 2020, 14, 3891–3898. [Google Scholar] [CrossRef] [PubMed]
  58. Calza, L.; Colangeli, V.; Borderi, M.; Beci, G.; Esposito, F.; Bon, I.; Re, M.C.; Viale, P. Rosuvastatin decreases serum inflammatory markers and slows atherosclerosis progression rate in treated HIV-infected patients with metabolic syndrome. Infect. Dis. 2021, 53, 81–88. [Google Scholar] [CrossRef]
  59. Zheng, H.; Li, H.; Wang, Y.; Li, Z.; Hu, B.; Li, X.; Fu, L.; Hu, H.; Nie, Z.; Zhao, B.; et al. Rosuvastatin Slows Progression of Carotid Intima-Media Thickness: The METEOR-China Randomized Controlled Study. Stroke 2022, 53, 3004–3013. [Google Scholar] [CrossRef]
  60. Rahhal, A.; Khir, F.; Orabi, B.; Chbib, S.; Al-Khalaila, O.; Abdelghani, M.S.; Osman, O.; Ashour, A.A.; Al-Awad, M.; Mahfouz, A.; et al. A Comparative Study of High-intensity Rosuvastatin Vs. Atorvastatin Therapy Post-acute Coronary Syndrome Using Real-world Data. Curr. Probl. Cardiol. 2022, 47, 100956. [Google Scholar] [CrossRef]
  61. Lee, Y.J.; Hong, S.J.; Kang, W.C.; Hong, B.K.; Lee, J.Y.; Lee, J.B.; Cho, H.J.; Yoon, J.; Lee, S.J.; Ahn, C.M.; et al. Rosuvastatin vs. atorvastatin treatment in adults with coronary artery disease: Secondary analysis of the randomised LODESTAR trial. BMJ 2023, 383, e075837. [Google Scholar] [CrossRef]
  62. Perez-Calahorra, S.; Laclaustra, M.; Marco-Benedi, V.; Pinto, X.; Sanchez-Hernandez, R.M.; Plana, N.; Ortega, E.; Fuentes, F.; Civeira, F. Comparative efficacy between atorvastatin and rosuvastatin in the prevention of cardiovascular disease recurrence. Lipids Health Dis. 2019, 18, 216. [Google Scholar] [CrossRef]
  63. Pérez de Isla, L.; Arroyo-Olivares, R.; Muñiz-Grijalvo, O.; Diaz-Díaz, J.L.; Zambón, D.; Fuentes, F.; Sánchez Muñoz-Torrero, J.F.; Mediavilla, J.D.; González-Estrada, A.; Miramontes-González, J.P.; et al. Long-term effect of 2 intensive statin regimens on treatment and incidence of cardiovascular events in familial hypercholesterolemia: The SAFEHEART study. J. Clin. Lipidol. 2019, 13, 989–996. [Google Scholar] [CrossRef]
  64. Barrios, V.; Escobar, C. Rosuvastatin along the cardiovascular continuum: From JUPITER to AURORA. Expert Rev. Cardiovasc. Ther. 2009, 7, 1317–1327. [Google Scholar] [CrossRef] [PubMed]
  65. Barrios, V.; Escobar, C. Rosuvastatin and diabetes: When the evidences talk. Cardiovasc. Hematol. Agents Med. Chem. 2013, 11, 115–124. [Google Scholar] [CrossRef] [PubMed]
  66. Ward, F.L.; John, R.; Bargman, J.M.; McQuillan, R.F. Renal Tubular Toxicity Associated with Rosuvastatin Therapy. Am. J. Kidney Dis. 2017, 69, 473–476. [Google Scholar] [CrossRef] [PubMed]
  67. Fácila Rubio, L.; Pintó Sala, X.; Cinza Sanjurjo, S.; García Goñi, M.; Cortés Gil, X.; Martí Ragué, I.; Soler Martinez, M.; Aceituno Mata, S. Cost-effectiveness of rosuvastatin vs. atorvastatin, simvastatin pitavastatin, fluvastatin, pravastatin and lovastatin in the treatment of patients with moderate, high or very high cardiovascular risk in Spain. Rev. Esp. Econ. Salud 2018, 13, 678–691. [Google Scholar]
  68. Olmo-Quintana, V.; Zamora, B.; Pinto Sala, X.; Mart Rague, I.; Cortes Gil, X.; Gari Peris, C.; Aceituno Mata, S. Cost-effectiveness of the combination rosuvastatin/ezetimibe vs. the combinations of simvastatin and atorvastatin with ezetimibe to reduce the risk of cardiovascular events. Rev. Esp. Econ. Salud 2021, 16, 42–57. [Google Scholar]
  69. Mata, P.; Cortes, X.; Marti, I.; Saborit Canals, G.; Pomares, E. Analysis of cost-consequence of rosuvastatin vs. atorvastatin in the Spanish setting. Rev. Esp. Econ. Salud 2022, 17, 120–133. [Google Scholar]
  70. Cannon, C.P.; Blazing, M.A.; Giugliano, R.P.; McCagg, A.; White, J.A.; Theroux, P.; Darius, H.; Lewis, B.S.; Ophuis, T.O.; Jukema, J.W.; et al. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N. Engl. J. Med. 2015, 372, 2387–2397. [Google Scholar] [CrossRef]
  71. Kim, C.H.; An, H.; Kim, S.H.; Shin, D. Pharmacokinetic and pharmacodynamic interaction between ezetimibe and rosuvastatin in healthy male subjects. Drug Des. Dev. Ther. 2017, 11, 3461–3469. [Google Scholar] [CrossRef]
  72. Rhee, M.Y.; Kim, K.J.; Kim, S.H.; Yoon, Y.W.; Rha, S.W.; Hong, S.J.; Kwak, C.H.; Kim, W.; Nam, C.W.; Park, T.H.; et al. Ezetimibe and rosuvastatin combination treatment can reduce the dose of rosuvastatin without compromising its lipid-lowering efficacy. Clin. Ther. 2019, 41, 2571–2592. [Google Scholar] [CrossRef]
  73. Escobar, C.; Anguita, M.; Arrarte, V.; Barrios, V.; Cequier, Á.; Cosín-Sales, J.; Egocheaga, I.; López de Sa, E.; Masana, L.; Pallarés, V.; et al. Recommendations to improve lipid control. Consensus document of the Spanish Society of Cardiology. Rev. Esp. Cardiol. 2020, 73, 161–167. [Google Scholar] [CrossRef]
  74. Ballantyne, C.M.; Weiss, R.; Moccetti, T.; Vogt, A.; Eber, B.; Sosef, F.; Duffield, E. Efficacy and safety of rosuvastatin 40 mg alone or in combination with ezetimibe in patients at high risk of cardiovascular disease (results from the EXPLORER study). Am. J. Cardiol. 2007, 99, 673–680. [Google Scholar] [CrossRef]
  75. Bays, H.E.; Davidson, M.H.; Massaad, R.; Flaim, D.; Lowe, R.S.; Tershakovec, A.M.; Jones-Burton, C. Safety and efficacy of ezetimibe added on to rosuvastatin 5 or 10 mg vs. up-titration of rosuvastatin in patients with hypercholesterolemia (the ACTE Study). Am. J. Cardiol. 2011, 108, 523–530. [Google Scholar] [CrossRef]
  76. Ballantyne, C.M.; Hoogeveen, R.C.; Raya, J.L.; Cain, V.A.; Palmer, M.K.; Karlson, B.W. Efficacy, safety and effect on biomarkers related to cholesterol and lipoprotein metabolism of rosuvastatin 10 or 20 mg plus ezetimibe 10 mg vs simvastatin 40 or 80 mg plus ezetimibe 10 mg in high-risk patients: Results of the GRAVITY randomized study. Atherosclerosis 2014, 232, 86–93. [Google Scholar] [CrossRef]
  77. Kim, K.J.; Kim, S.H.; Yoon, Y.W.; Rha, S.W.; Hong, S.J.; Kwak, C.H.; Kim, W.; Nam, C.W.; Rhee, M.Y.; Park, T.H.; et al. Effect of fixed-dose combinations of ezetimibe plus rosuvastatin in patients with primary hypercholesterolemia: MRS-ROZE (Multicenter Randomized Study of ROsuvastatin and eZEtimibe). Cardiovasc. Ther. 2016, 34, 371–382. [Google Scholar] [CrossRef]
  78. Yang, Y.J.; Lee, S.H.; Kim, B.S.; Cho, Y.K.; Cho, H.J.; Cho, K.I.; Kim, S.Y.; Ryu, J.K.; Cho, J.M.; Park, J.I.; et al. Combination therapy of rosuvastatin and ezetimibe in patients with high cardiovascular risk. Clin. Ther. 2017, 39, 107–117. [Google Scholar] [CrossRef]
  79. Kim, W.; Yoon, Y.E.; Shin, S.H.; Bae, J.W.; Hong, B.K.; Hong, S.J.; Sung, K.C.; Han, S.H.; Kim, W.; Rhee, M.Y.; et al. Efficacy and safety of ezetimibe and rosuvastatin combination therapy vs. those of rosuvastatin monotherapy in patients with primary hypercholesterolemia. Clin. Ther. 2018, 40, 993–1013. [Google Scholar] [CrossRef]
  80. Hong, S.J.; Jeong, H.S.; Ahn, J.C.; Cha, D.H.; Won, K.H.; Kim, W.; Cho, S.K.; Kim, S.Y.; Yoo, B.S.; Sung, K.C.; et al. A phase III, multicenter, randomized, double-blind, active comparator clinical trial to compare the efficacy and safety of combination therapy with ezetimibe and rosuvastatin vs. rosuvastatin monotherapy in patients with hypercholesterolemia: I-ROSETTE (Ildong rosuvastatin & ezetimibe for hypercholesterolemia) randomized controlled trial. Clin. Ther. 2018, 40, 226–241.e4. [Google Scholar] [PubMed]
  81. Su, Q.; Liu, Y.; Zhang, G.; Xu, L.; Wang, M.; Mei, S.; Garon, G.; Wu, Y.; Lv, Q.; Ma, C. Efficacy and Safety of Single-Pill Combination of Rosuvastatin and Ezetimibe in Chinese Patients with Primary Hypercholesterolemia Inadequately Controlled by Statin Treatment (ROZEL): A Randomized, Double-Blind, Double Dummy, Active-Controlled Phase 3 Clinical Trial. Adv. Ther. 2023, 40, 5285–5299. [Google Scholar] [PubMed]
  82. Chilbert, M.R.; VanDuyn, D.; Salah, S.; Clark, C.M.; Ma, Q. Combination Therapy of Ezetimibe and Rosuvastatin for Dyslipidemia: Current Insights. Drug Des. Dev. Ther. 2022, 16, 2177–2186. [Google Scholar] [CrossRef]
  83. Strilchuk, L.; Tocci, G.; Fogacci, F.; Cicero, A.F.G. An overview of rosuvastatin/ezetimibe association for the treatment of hypercholesterolemia and mixed dyslipidemia. Expert Opin. Pharmacother. 2020, 21, 531–539. [Google Scholar] [CrossRef] [PubMed]
  84. Boutari, C.; Karagiannis, A.; Athyros, V.G. Rosuvastatin and ezetimibe for the treatment of dyslipidemia and hypercholesterolemia. Expert Rev. Cardiovasc. Ther. 2021, 19, 575–580. [Google Scholar] [CrossRef] [PubMed]
  85. Barrios, V.; Pintó, X.; Escobar, C.; Varona, J.F.; Gámez, J.M. Real-World Attainment of Low-Density Lipoprotein Cholesterol Goals in Patients at High Risk of Cardiovascular Disease Treated with High-Intensity Statins: The TERESA Study. J. Clin. Med. 2023, 12, 3187. [Google Scholar] [CrossRef] [PubMed]
  86. Escobar, C.; Barrios, V.; Cequier, A.; Cosin-Sales, J.; Seijas, J.; Doblas, J.J.G.; Arrarte, V.; Tuñon, J.; Banach, M. Impact of the Spanish consensus for improving lipid control on patients admitted for an acute coronary syndrome. J. Clin. Lipidol. 2023, 17, 756–764. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proportion of patients that achieve LDL-C targets and proportion of patients using lipid-lowering drugs in the SANTORINI study according to the presence of vascular disease. LDL-C: low-density lipoprotein cholesterol; PCSK9i: proprotein convertase subtilisin/kexin type 9 inhibitors. Figure created with data from reference [16].
Figure 1. Proportion of patients that achieve LDL-C targets and proportion of patients using lipid-lowering drugs in the SANTORINI study according to the presence of vascular disease. LDL-C: low-density lipoprotein cholesterol; PCSK9i: proprotein convertase subtilisin/kexin type 9 inhibitors. Figure created with data from reference [16].
Jcm 13 01894 g001
Figure 2. Effect of rosuvastatin vs. placebo on main clinical outcomes in the JUPITER trial. Primary endpoint: myocardial infarction, stroke, arterial revascularization, hospitalization for unstable angina or cardiovascular death. MACE: cardiovascular death, myocardial infarction, or stroke. CVD: cardiovascular disease. HR: Hazard Ratio. 95% CI: 95% confidence interval. LDL-C: low-density lipoprotein cholesterol. Figure made with data from reference [39].
Figure 2. Effect of rosuvastatin vs. placebo on main clinical outcomes in the JUPITER trial. Primary endpoint: myocardial infarction, stroke, arterial revascularization, hospitalization for unstable angina or cardiovascular death. MACE: cardiovascular death, myocardial infarction, or stroke. CVD: cardiovascular disease. HR: Hazard Ratio. 95% CI: 95% confidence interval. LDL-C: low-density lipoprotein cholesterol. Figure made with data from reference [39].
Jcm 13 01894 g002
Table 1. Estimated LDL cholesterol reduction of rosuvastatin and the rosuvastatin/ezetimibe combination, compared to atorvastatin, as well as the associated costs per cycle.
Table 1. Estimated LDL cholesterol reduction of rosuvastatin and the rosuvastatin/ezetimibe combination, compared to atorvastatin, as well as the associated costs per cycle.
TreatmentLDL-C ReductionCost per Cycle (2018)TreatmentLDL-C ReductionCost per Cycle (2021)
Rosuvastatin 10 mg46%87.73€Rosu/EZE 10/10 mg56.7%340.5€
Atorvastatin 40 mg49%150.78€Ator/EZE 40/10 mg58.8%396.03€
Rosuvastatin 20 mg50%175.46€Rosu/EZE 20/10 mg59.8%422.23€
Atorvastatin 80 mg50%306.24€Ator/EZE 80/10 mg59.9%530.94€
Ator: atorvastatin; EZE: ezetimibe; Rosu: rosuvastatin. Table created with data from references [67,68].
Table 2. Effect of the rosuvastatin/ezetimibe combination on LDL cholesterol in phase 3 clinical trials.
Table 2. Effect of the rosuvastatin/ezetimibe combination on LDL cholesterol in phase 3 clinical trials.
Study (Year of Publication)TreatmentsLDL-C Reduction (%)Proportion of Patients That Attain LDL-C Targets (%)
EXPLORER (2007) [74]RSV/EZ 40 mg/10mg−70.0 (p < 0.001)94.0 (p < 0.001)
RSV 40 mg−57.079.1
GRAVITY (2014) [76]RSV/EZE 10 mg/10 mg−59.7 (p < 0.05 vs. SIM/EZE 40/10 mg)93.3 (p < 0.05 vs. SIM/EZE 40/10 mg)
RSV/EZE 20 mg/10 mg−63.5 (p < 0.05 vs. SIM/EZE 40–80/10 mg)95.6 (p < 0.05 vs. SIM/EZE 40–80/10 mg)
SIM/EZE 40 mg/10 mg−55.287.4
SIM/EZE 80 mg/10 mg−57.488.6
MRS-ROZE (2016) [77]RSV/EZE 5–20 mg/10 mg−59.1 (p < 0.001)94.1 (p ≤ 0.01)
RSV 5–20 mg−49.486.3
ROSE (2017) [78]RSV/EZE 5–20 mg/10 mg−59.5 (p < 0.001)90.7 (p ≤ 0.01)
RSV 5–20 mg−51.172.9
SP-RE-003 (2018) [79]RSV/EZE 5–20 mg/10 mg−56.5 (p ≤ 0.01)94.2 (p < 0.05)
RSV 5–20 mg−45.286.6
I-ROSETTE (2018) [80]RSV/EZE 5–20 mg/10 mg−57.0 (p < 0.001)92.3 (p < 0.001)
RSV 5–20 mg−44.479.9
LDL-C: Low-density lipoprotein cholesterol; EZE: ezetimibe; RSV: rosuvastatin; SIM: simvastatin. Table created with data from references [74,76,77,78,79,80].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mostaza, J.M.; Escobar, C. Rosuvastatin-Based Lipid-Lowering Therapy for the Control of LDL Cholesterol in Patients at High Vascular Risk. J. Clin. Med. 2024, 13, 1894. https://doi.org/10.3390/jcm13071894

AMA Style

Mostaza JM, Escobar C. Rosuvastatin-Based Lipid-Lowering Therapy for the Control of LDL Cholesterol in Patients at High Vascular Risk. Journal of Clinical Medicine. 2024; 13(7):1894. https://doi.org/10.3390/jcm13071894

Chicago/Turabian Style

Mostaza, Jose María, and Carlos Escobar. 2024. "Rosuvastatin-Based Lipid-Lowering Therapy for the Control of LDL Cholesterol in Patients at High Vascular Risk" Journal of Clinical Medicine 13, no. 7: 1894. https://doi.org/10.3390/jcm13071894

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