Moderate-Intensity Rosuvastatin Plus Ezetimibe Versus High-Intensity Rosuvastatin for Target Low-Density Lipoprotein Cholesterol Goal Achievement in Patients With Recent Ischemic Stroke: A Randomized Controlled Trial

Article information

J Stroke. 2023;25(2):242-250
Publication date (electronic) : 2023 April 11
doi : https://doi.org/10.5853/jos.2022.02957
1Department of Neurology, Ilsan Paik Hospital, Inje University, Goyang, Korea
2Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
3Department of Neurology, Myongji Hospital, Hanyang University College of Medicine, Goyang, Korea
4Department of Neurology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
5Department of Neurology, Seoul Hospital, Ewha Women’s University College of Medicine, Seoul, Korea
6Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
7Department of Neurology, Inha University Hospital, Incheon, Korea
8Department of Neurology, Seoul National University College of Medicine, Seoul, Korea
9Department of Neurology, Kyung Hee University Medical Center, Seoul, Korea
10Department of Neurology, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
11Department of Neurology, Hallym University Sacred Heart Hospital, Anyang, Korea
12Department of Neurology, Chung-Ang University Hospital, Seoul, Korea
13Department of Neurology, Korea University Guro Hospital, Seoul, Korea
14Department of Biostatistics, Korea University College of Medicine, Seoul, Korea
Correspondence: Keun-Sik Hong Department of Neurology, Inje University Ilsan Paik Hospital, 170 Juhwa-ro, Ilsanseo-gu, Goyang 10380, Korea Tel: +82-31-910-7680 Email: nrhks455@gmail.com
Received 2022 September 15; Revised 2023 January 17; Accepted 2023 February 14.

Abstract

Background and Purpose

Moderate-intensity statin plus ezetimibe versus high-intensity statin alone may provide a greater low-density lipoprotein cholesterol (LDL-C) reduction in patients with recent ischemic stroke.

Methods

This randomized, open-label, controlled trial assigned patients with recent ischemic stroke <90 days to rosuvastatin/ezetimibe 10/10 mg once daily (ROS10/EZT10) or to rosuvastatin 20 mg once daily (ROS20). The primary endpoint was LDL-C reduction ≥50% from baseline at 90 days. Key secondary endpoints were LDL-C <70 mg/dL and multiple lipid goal achievement, and composite of major vascular events.

Results

Of 584 randomized, 530 were included in the modified intention-to-treat analysis. The baseline LDL-C level was 130.2±34.7 mg/dL in the ROS10/EZT10 group and 131.0±33.9 mg/dL in the ROS20 group. The primary endpoint was achieved in 198 patients (72.5%) in the ROS10/EZT10 group and 148 (57.6%) in the ROS20 group (odds ratio [95% confidence interval], 1.944 [1.352–2.795]; P= 0.0003). LDL-C level <70 mg/dL was achieved in 80.2% and 65.4% in the ROS10/EZT10 and ROS20 groups (P=0.0001). Multiple lipid goal achievement rate was 71.1% and 53.7% in the ROS10/EZT10 and ROS20 groups (P<0.0001). Major vascular events occurred in 1 patient in the ROS10/EZT10 group and 9 in the ROS20 group (P=0.0091). The adverse event rates did not differ between the two groups.

Conclusion

Moderate-intensity rosuvastatin plus ezetimibe was superior to high-intensity rosuvastatin alone for intensive LDL-C reduction in patients with recent ischemic stroke. With the combination therapy, more than 70% of patients achieved LDL-C reduction ≥50% and 80% had an LDL-C <70 mg/dL at 90 days.

Introduction

In patients with ischemic stroke or transient ischemic attack due to atherosclerosis or having concomitant atherosclerotic diseases, intensive low-density lipoprotein cholesterol (LDL-C) lowering is recommended to reduce the risk of subsequent vascular events. In meta-analyses, the benefit of stroke prevention was greater as the magnitude of LDL-C reduction was larger and the achieved LDL-C level was lower [1,2]. For secondary stroke prevention, guidelines recommend LDL-C reduction of ≥50% from baseline or a target LDL-C goal of <70 mg/dL [3,4].

As a first-line strategy for intensive LDL-C lowering, high-intensity statins are generally recommended. However, high-intensity statins as compared to moderate- or low-intensity statins increase the risks of diabetes, hepatic dysfunction, and myopathy [5-7]. Furthermore, the additional reduction in LDL-C with increasing statin dose is limited, only 5% to 7% absolute reduction with doubling the dose of statins [8].

Non-statin lipid-lowering therapies added to statins were effective for preventing cardiovascular events, and the benefit was likely driven by further LDL-C lowering [9-12]. Ezetimibe inhibits intestinal cholesterol absorption by binding to the Niemann–Pick C1-like 1 (NPC1L1) protein [13]. When added to statins, ezetimibe resulted in a substantial LDL-C reduction, and the reduction appeared greater than that achieved with doubling statin dose [8,14,15]. For patients with recent ischemic stroke, there has been no randomized trial that compared moderate-intensity statin plus ezetimibe and high-intensity statin for LDL-C reduction.

We conducted the ROSuvastatin plus Ezetimibe Treatment for Target LDL-C goal Achievement in patients with recent ischemic Stroke (ROSETTA-Stroke) trial to evaluate moderate-intensity rosuvastatin plus ezetimibe, as compared with high-intensity rosuvastatin alone for target LDL-C goal achievement in patients with recent ischemic stroke of atherosclerotic origin or having concomitant atherosclerotic cardiovascular diseases.

Methods

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Trial design

In this multicenter, randomized, open-label, parallel-group trial conducted at 13 sites in South Korea, we compared rosuvastatin 10 mg plus ezetimibe 10 mg once daily (ROS10/EZT10) and rosuvastatin 20 mg alone once daily (ROS20) for target LDL-C goal achievement in patients with recent ischemic stroke. In Korea, rosuvastatin 40 mg has not been approved, and accordingly we selected the dose of 20 mg as a high-intensity rosuvastatin and 10 mg as a moderate-intensity rosuvastatin [16].

This was an investigator-initiated trial and funded by Hanmi Pharm. Co., Ltd. The sponsor participated in the trial design and provided study medications, but it had no role in the conduct of the trial, data collection and analysis, and writing and submitting the manuscript for publication. All the data were collected by the site investigators, and were monitored and maintained by an independent research organization contracted by the principal investigator affiliated academic institution. All the authors vouch for the completeness and accuracy of the data and the fidelity of the trial to the protocol. The principal investigator and independent statisticians had full access to and vouch for the data and analysis. The trial was approved by local institutional review boards. Enrolled patients or their legally authorized representatives provided written informed consent. The trial is registered with ClinicalTrials.gov (NCT03993236).

Study population

Patients were eligible for enrollment if they were 19 years or older; they had recent ischemic stroke within 90 days confirmed by diffusion-weighted imaging; statin therapy was indicated according to the recommendations of the 2014 American Heart Association/American Stroke Association guidelines [17]; they had not taken statins within 4 weeks before the index stroke; and the baseline LDL-C level was measured after onset of the index stroke. Prior to randomization, administration of non-study statins immediately after acute stroke hospitalization according to each institution’s practice protocol was allowed. In this case, the baseline LDL-C level should be measured within 3 days of initiating non-study statins, and the randomization and initiation of study medications should be completed within 7 days of the baseline LDL-C level measurement. Detailed inclusion and exclusion criteria are provided in Supplementary Table 1.

Randomization, intervention, and follow-up

Eligible patients were randomly assigned, in a 1:1 ratio, to receive, once daily, either rosuvastatin 10 mg plus ezetimibe 10 mg (ROS10/EZT10 group) or rosuvastatin 20 mg alone (ROS20 group). Randomization was stratified by centers and baseline LDL-C levels (<100 mg/dL vs. ≥100 mg/dL), using an interactive web response system. All patients received the standard secondary stroke prevention therapy recommended by current practice guidelines. Concomitant use of additional lipid-lowering agents (non-study statins, fibrates, or omega-3 fatty acids) was not allowed during the trial.

Patients were scheduled for follow-up visits at 30 days and at 90 days after the enrollment. Unscheduled visits were made as needed. Major vascular events (including recurrent stroke, coronary event, or vascular death) and adverse events were collected during the follow-up. Lipid profiles and laboratory tests for monitoring adverse events were measured at the 90-day follow-up visit. For patients who withdrew during the trial, we recommended to measure lipid profiles at the time of withdrawal, which were used for endpoint analysis if available.

Endpoints

The primary endpoint was the proportion of patients who achieved a target LDL-C goal, defined as LDL-C reduction of ≥50% from baseline. We selected the LDL-C reduction of ≥50% as a target goal, which was widely recommended by the guidelines when we designed this trial [16].

Secondary endpoints included (1) proportion of patients achieving LDL-C level <70 mg/dL, (2) proportion of patients with LDL-C reduction ≥50% or achieved LDL-C level <70 mg/dL, (3) the absolute and percentage changes in LDL-C level, (4) proportion of patients achieving multiple lipid goals of total cholesterol <200 mg/dL, LDL-C <70 mg/dL, and triglyceride <150 mg/dL [18], (5) composite of major vascular events including stroke (ischemic or hemorrhagic), coronary event (myocardial infarction or coronary revascularization), or vascular death, (6) all cause of death, (7) new onset diabetes, (8) fatigue assessed by the Fatigue Severity Scale [19], (9) rhabdomyolysis, and (10) significant liver enzyme elevation (alanine aminotransferase or aspartate aminotransferase elevation >3 times from baseline).

Safety endpoints included any adverse event, treatment-emergent adverse event (TEAE), any serious adverse event, and serious TEAE. TEAEs were defined as adverse events for which a causal relationship with study medications could not be excluded. Serious events included those that (1) resulted in death or were life-threatening, (2) required or prolonged hospitalization, (3) resulted in persistent or significant disability/incapacity, (4) caused a congenital anomaly/birth defect, or (5) resulted in the development of drug dependency/abuse or significant medical events.

Sample size calculation and statistical analysis

This trial was designed to detect the superiority of rosuvastatin 10 mg plus ezetimibe 10 mg once daily over rosuvastatin 20 mg once daily for achieving LDL-C reduction ≥50% from baseline. Based on the prior study findings [20], the sample size was calculated by assuming that the primary endpoint rate would be 82% in the ROS10/EZT10 group and 72% in the ROS20 group. Under two-sided significance level of 5%, a total of 554 patients (277 patients per group) were needed to ensure 80% of power of detecting 10% of absolute difference in the primary endpoint rate using the chi-square test. Assuming a 5% dropout rate, a total of 584 patients (292 patients per group) was planned to enroll. Given that lipid-lowering with statins and ezetimibe are approved and widely used in daily clinical practice, neither an independent data safety monitoring board was organized, nor interim analysis was planned.

The primary and secondary endpoints were assessed in the modified intent-to-treat (ITT) population, which included patients who were randomized, received at least 1 dose of study medications, and completed follow-up LDL-C level measurement. For patients who withdrew during the trial after initiation of study medications, but their LDL-C levels were measured at the time of withdrawal, they were included in the modified ITT population. Per-protocol (PP) analysis was additionally conducted, which included patients who had no major protocol violation, took >80% of the assigned study medications, and did not take non-study lipid-lowering agents after enrollment among the modified ITT population. Adverse events were assessed in the safety population, which included all patients who were randomized and received at least 1 dose of study medications.

The primary endpoint was analyzed using the chi-square test. Additionally, an adjusted analysis was planned if there were significant differences in the baseline characteristics with biological plausibility, but no such variable was found. For the baseline LDL-C levels, we predetermined that they would not be adjusted even if there was a significant imbalance between the two groups because randomization was stratified by the baseline LDL-C levels (<100 mg/dL vs. ≥100 mg/dL). Secondary and safety endpoints were analyzed, using the chi-square test, Fisher’s exact test, Student’s t-test, or Mann-Whitney U-test as indicated in the statistical analysis plan (SAP). All statistical analyses were performed using the SAS software version 9.4 (SAS Institute, Cary, NC, USA), and a two-sided P-value <0.05 was considered significant. All analyses were pre-planned in the SAP and performed by two independent statisticians who were blinded to the treatment allocation.

Results

Patients

Between September 9, 2019, and September 27, 2021, 584 patients were randomized (295 to the ROS10/EZT10 group and 289 to the ROS20 group). After randomization, 2 patients in the ROS20 group did not take any study medication, and 52 (22 in the ROS10/EZT10 group and 30 in the ROS20 group) did not undergo follow-up LDL-C measurements. Therefore, 530 patients (273 in the ROS10/EZT10 group and 257 in the ROS20 group) were included in the modified ITT population. Of the modified ITT population, 31 patients took less than 80% of study medications or received other lipid-lowering agents, and 499 patients (262 in the ROS10/EZT10 group and 237 in the ROS20 group) were included in the PP population. The safety population included 582 patients (295 in the ROS10/EZT10 group and 287 in the ROS20 group) (Figure 1).

Figure 1.

Trial flow. LDL-C, low-density lipoprotein cholesterol.

Baseline demographics and clinical characteristics were well balanced between the two groups (Table 1). The mean LDL-C level at baseline was 130.2 mg/dL in the ROS10/EZT10 group and 131.0 mg/dL in the ROS20 group. In each group, the median interval from the index ischemic stroke onset to randomization was 4 days, and the median National Institutes of Health Stroke Scale (NIHSS) score at randomization was 2. Before randomization, 193 patients (70.7%) in the ROS10/EZT10 group and 188 (73.2%) in the ROS20 group received non-study statins immediately after acute stroke hospitalization. The proportion of patients taking non-study statins before randomization and the average dose (converted to atorvastatin equivalent dose) did not differ between the two groups. The interval from initiating non-study statins to randomization was 3.0±1.7 days in the ROS10/EZT10 group and 3.1±1.6 days in the ROS20 group (P=0.8958) (Table 1 and Supplementary Table 2).

Baseline characteristics of the modified intention-to-treat population

The follow-up LDL-C levels were obtained at 90 days in 516 patients (266 in the ROS10/EZT10 group and 250 in the ROS20 group). In 14 (7 in each group) patients, the follow-up LDL-C levels were measured at the time of withdrawal, and these values were used for the endpoint analysis.

Endpoints

The primary endpoint of LDL-C reduction ≥50% from baseline was achieved in 198 patients (72.5%) in the ROS10/EZT10 group and 148 (57.6%) in the ROS20 group of the modified ITT population, and the difference was significant (odds ratio [95% confidence interval], 1.944 [1.352–2.795]; P=0.0003) (Table 2). Of secondary endpoints regarding lipid profiles, the ROS10/EZT10 group compared to the ROS20 group had higher proportions of patients with the achieved LDL-C level <70 mg/dL (80.2% vs. 65.4%; OR [95% CI], 2.148 [1.450–3.184]; P=0.0001), LDL-C reduction ≥50% or the achieved LDL-C <70 mg/dL (84.2% vs. 73.2%; OR [95% CI], 1.963 [1.281–3.008]; P=0.0018), and multiple lipid goal achievement (71.1% vs. 53.7%; OR [95% CI], 2.117 [1.479– 3.030]; P<0.0001). The absolute LDL-C reduction was greater in the ROS10/EZT10 group than in the ROS20 group (72.7 mg/dL vs. 64.7 mg/dL; P=0.0111), and accordingly the achieved LDL-C level was lower in the ROS10/EZT10 group than in the ROS20 group (57.4±24.6 mg/dL vs. 66.3±26.2 mg/dL; P<0.0001) (Supplementary Table 3).

Endpoints in the modified intent-to-treat population

Major vascular events occurred in 1 patient (0.4%) in the ROS10/EZT10 group and 9 (3.5%) in the ROS20 group (OR [95% CI], 0.101 [0.013–0.805]; P=0.0091). Of the 10 major vascular events, 7 were recurrent ischemic strokes and 3 were coronary events. Death occurred in 1 patient (sudden cardiac death) in the ROS20 group. There was no rhabdomyolysis during the trial. The two groups did not differ in the incidences of new onset diabetes and significant liver enzyme elevation, and the Fatigue Severity Scale. The results of the PP population analyses were consistent with those of the modified ITT population analyses (Supplementary Table 4).

Total cholesterol, high-density lipoprotein cholesterol, and triglyceride levels were comparable between the two groups at baseline (Table 1). At follow-up, the total cholesterol level was lower in the ROS10/EZT10 group than in the ROS20 group. However, the 90-day high-density lipoprotein cholesterol and triglyceride levels did not differ between the two groups (Supplementary Table 3).

Of the modified ITT population, 14 patients (4 in the ROS10/EZT10 group and 10 in the ROS20 group) discontinued study medications transiently or permanently between the 1st followup visit (at day 30) and the final visit (at day 90). Analysis excluding these patients showed consistent results regarding LDL-C related endpoints (Supplementary Table 5). Additional analysis excluding 22 patients (8 in the ROS10/EZT10 group and 14 in the ROS20 group) who transiently or permanently discontinued study medications at any point during the trial also showed similar results (Supplementary Table 6).

Subgroup analyses were not prespecified, but were undertaken in a post-hoc manner. For the primary endpoint, treatment effects were consistent across major subgroups of age (<64 vs. ≥64), sex, baseline LDL-C level (<100 mg/dL vs. ≥100 mg/dL), hypertension, diabetes, and smoking (Figure 2).

Figure 2.

Subgroup analyses of primary endpoint, according to post hoc subgroups. LDL-C reduction ≥50% (the primary end point) at 90 days among patients in the ROS10/EZT10 group and in the ROS20 group, according to post hoc subgroups. LDL-C, low-density lipoprotein cholesterol; OR, odds ratio; CI, confidence interval; DM, diabetes mellitus.

Adverse events

In the safety population, there were no significant differences between the ROS10/EZT10 and ROS20 groups in the rates of any adverse event (36.9% vs. 36.9%; P=0.9969), any TEAE (4.7% vs. 3.5%; P=0.3814), any serious adverse event (7.5% vs. 7.7%; P=0.9244), and serious TEAE (0% vs. 0%; P>0.9999) (Table 3). Details of any serious adverse events and TEAE according to individual organs are described in Supplementary Table 7 and 8. For muscle-related adverse events, there was no clinically significant myopathy. Myalgia occurred in 5 patients (2.3%) in the ROS10/EZT10 group and 3 (1.4%) in the ROS20 group (P=0.5000). Of these, myalgia in 6 patients (3 in each group) was considered as TEAEs.

Adverse events in the safety population

Discussion

In this randomized trial, we found that the combination of moderate-intensity rosuvastatin 10 mg plus ezetimibe 10 mg was superior to high-intensity rosuvastatin 20 mg alone for LDL-C reduction in patients with recent ischemic stroke who initiated lipidlowering therapy. With the combination therapy, more than 70% of patients achieved an LDL-C reduction ≥50% from baseline, and more than 80% had their LDL-C levels <70 mg/dL at 90 days. For every 100 patients indicated for LDL-C lowering, approximately additional 15 patients would achieve these target LDL-C goals with the combination therapy as compared to rosuvastatin 20 mg alone. In addition, 71% of patients with the combination therapy achieved the multiple lipid goals, an absolute increase of about 17% as compared to that with rosuvastatin 20 mg alone.

Our findings are in accord with earlier studies. A prior meta-analysis showed that the LDL-C reduction was greater with adding ezetimibe to ongoing statins (26.0% reduction from baseline) than with doubling the dose of ongoing statins (9.7% reduction) [15]. In the randomized comparison of efficacy and safety of lipid-lowering with statin monotherapy versus statin ezetimibe combination for high-risk cardiovascular disease (RACING) conducted in South Korea, 73% of patients receiving rosuvastatin 10 mg plus ezetimibe 10 mg and 55% of those receiving rosuvastatin 20 mg alone achieved an LDL-C level of <70 mg/dL at 1 year [21]. The difference in the proportion (about 18%) between the 2 arms in RACING was quite comparable to that observed in our trial (about 15%). Both RACING and our trial would indicate that 7–8 out of 10 Korean patients with atherosclerotic diseases could achieve an LDL-C level of <70 mg/dL with rosuvastatin 10 mg plus ezetimibe 10 mg once daily.

In this trial, the clinical endpoints of major vascular events occurred significantly less in the ROS10/EZT10 group than in the ROS20 group. However, this trial was neither designed nor powered to detect the difference in the clinical endpoint. Given the short trial period and the magnitude of the achieved LDL-C level difference between the two groups, the clinical benefit observed in this study is highly likely due to by chance. Meta-analyses and a clinical trial indicated the greater benefit for secondary stroke prevention with greater LDL-C reduction or lower LDL-C levels achieved [1,2,22], and accordingly moderate-intensity rosuvastatin plus ezetimibe could be superior to high-intensity rosuvastatin to prevent vascular events. The RACING trial demonstrated that rosuvastatin 10 mg plus ezetimibe 10 mg was non-inferior to rosuvastatin 20 mg alone for the 3-year clinical composite outcome of major vascular events. The rate was slightly lower with the combination therapy (9.1%) than with rosuvastatin alone (9.9%) [21]. Therefore, both RACING and our trial suggest that rosuvastatin 10 mg plus ezetimibe 10 mg compared to rosuvastatin 20 mg alone was, at least, non-inferior for preventing vascular events, and was better for intensive LDL-C reduction. Clinical efficacy of statin plus ezetimibe combination therapy was further supported by a post hoc analysis of the Treat Stroke to Target (TST) trial [23]. In the lower LDL-C target group (LDL-C <70 mg/dL), the proportion of patients on combination therapy increased from <5% at baseline to >20% at 6 months and >50% at 3 years. The risk reduction of primary endpoint (major cardiovascular event) in the lower target group as compared with the higher target group (LDL-C 100±10 mg/dL) was significant during combination therapy, but not during statin monotherapy.

We observed a numerically higher incidence of alanine aminotransferase or aspartate aminotransferase elevation >3 times from baseline in the ROS10/EZT10 group than in the ROS20 group. However, the incidence was low and did not significantly differ between the two groups. There was no case of clinically significant hepatic dysfunction. The low TEAE incidences and no case of serious TEAE in the two groups support the safety of the two treatments.

In this trial, we selected rosuvastatin 20 mg as a high-intensity statin strategy because rosuvastatin 40 mg has not been approved for clinical use in Korea. The 2013 American College of Cardiology/American Heart Association guidelines categorized rosuvastatin 20 mg as high-intensity statin [16]. Studies have shown an increased risk of intracerebral hemorrhage with high-intensity statins, and the risk could be greater during the acute stage of ischemic stroke. However, in a randomized Korean trial involving 316 patients with acute ischemic stroke within 48 hours, the incidence of any intracranial hemorrhage detected on follow-up gradient-recalled echo magnetic resonance imaging at 14 days was not higher, but rather lower, in the rosuvastatin 20 mg group than in the placebo group [24]. In our study, patients were enrolled, on average, 4–5 days after acute ischemic stroke, and there was no hemorrhagic stroke during the follow-up. Therefore, both the combination therapy and high-intensity rosuvastatin appear safe in patients with acute ischemic stroke with regard to intracerebral hemorrhage.

Our study has several limitations. This was an open-label study, which was at risk of a reporting bias for clinical events. However, key endpoints were objective laboratory results, and independent statisticians blinded to the treatment allocation analyzed the data. We exclusively enrolled Korean patients. The response to lipid-lowering therapy could differ across ethnicities [25], and thereby the generalizability of our findings might be limited. In addition, most of the included patients had a mild stroke, which could limit the applicability of our findings to patients with more severe strokes. Whether the greater LDL-C reduction with the combination therapy versus high-intensity rosuvastatin alone could be translated into better secondary stroke prevention remains unclear, which should be formally tested by a well-designed large trial.

Conclusions

The ROSETTA-Stroke trial showed that moderate-intensity rosuvastatin plus ezetimibe was superior to high-intensity rosuvastatin alone for intensive LDL-C lowering in patients with recent ischemic stroke who initiated lipid-lowering therapy. For every 100 patients treated with rosuvastatin 10 mg plus ezetimibe 10 mg once daily, almost 73 achieved LDL-C reduction ≥50%, and about 80 had an LDL-C level <70 mg/dL at 90 days.

Supplementary materials

Supplementary materials related to this article can be found online at https://doi.org/10.5853/jos.2022.02957.

Supplementary Table 1.

Inclusion and exclusion criteria

jos-2022-02957-Supplementary-Table-1-2.pdf
Supplementary Table 2.

Non-study statin use before randomization

jos-2022-02957-Supplementary-Table-1-2.pdf
Supplementary Table 3.

ipid profiles at follow-up in the modified intention- to-treat population

jos-2022-02957-Supplementary-Table-3-5.pdf
Supplementary Table 4.

End points in the per-protocol population

jos-2022-02957-Supplementary-Table-3-5.pdf
Supplementary Table 5.

LDL-C related endpoints after excluding patients who discontinued study medications transiently or permanently between the 1st follow-up visit (at day 30) and the final visit (at day 90)

jos-2022-02957-Supplementary-Table-3-5.pdf
Supplementary Table 6.

LDL-C related endpoints after excluding patients who trainsiently or permanently discontinued study medications during the trial

jos-2022-02957-Supplementary-Table-6-8.pdf
Supplementary Table 7.

Any serious adverse events by MedDRA body system in the safety population

jos-2022-02957-Supplementary-Table-6-8.pdf
Supplementary Table 8.

TEAEs by MedDRA body system in the safety population

jos-2022-02957-Supplementary-Table-6-8.pdf

Notes

Funding statement

This study was funded by Hanmi Pharm. Co., Ltd.

Conflicts of interest

K.- S. Hong, O. Y. Bang, J.-H. Park, J.-M. Jung, S.-H. Lee, T.-J. Song, H. S. Nam, H.-K. Park, K.-H. Jung, S. H. Heo, J. Koo, K.-H. Yu, K.-Y. Park, C. K. Kim, H.-K Park, J. Lee, W.-K. Seo received research grants from Hanmi Pharm. Co., Ltd.

Author contribution

Conceptualization: KSH, OYB, HKP, WKS. Study design: KSH, OYB, HKP, WKS, JL (Juneyoung Lee). Methodology: KSH, OYB, HKP, WKS, JL (Juneyoung Lee). Data collection: all authors. Investigation: all authors. Statistical analysis: KSH, OYB. JL (Jiyoon Lee), JL (Juneyoung Lee). Writing—original draft: KSH, JL (Juneyoung Lee). Writing—review & editing: all authors. Funding acquisition: KSH, OYB. Approval of final manuscript: all authors.

References

1. Amarenco P, Labreuche J. Lipid management in the prevention of stroke: review and updated meta-analysis of statins for stroke prevention. Lancet Neurol 2009;8:453–463.
2. Shin J, Chung JW, Jang HS, Lee J, Hong KS, Bang OY, et al. Achieved low-density lipoprotein cholesterol level and stroke risk: a meta-analysis of 23 randomised trials. Eur J Prev Cardiol 2021;28:905–916.
3. Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, et al. 2016 ESC/EAS guidelines for the management of dyslipidaemias. Eur Heart J 2016;37:2999–3058.
4. Kleindorfer DO, Towfighi A, Chaturvedi S, Cockroft KM, Gutierrez J, Lombardi-Hill D, et al. 2021 Guideline for the prevention of stroke in patients with stroke and transient ischemic attack: a guideline from the American Heart Association/American Stroke Association. Stroke 2021;52:e364–e467.
5. Alsheikh-Ali AA, Maddukuri PV, Han H, Karas RH. Effect of the magnitude of lipid lowering on risk of elevated liver enzymes, rhabdomyolysis, and cancer: insights from large randomized statin trials. J Am Coll Cardiol 2007;50:409–418.
6. Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, et al. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010;376:1670–1681.
7. Preiss D, Seshasai SR, Welsh P, Murphy SA, Ho JE, Waters DD, et al. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA 2011;305:2556–2564.
8. Nicholls SJ, Brandrup-Wognsen G, Palmer M, Barter PJ. Metaanalysis of comparative efficacy of increasing dose of atorvastatin versus Rosuvastatin versus Simvastatin on lowering levels of atherogenic lipids (from VOYAGER). Am J Cardiol 2010;105:69–76.
9. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med 2017;376:1713–1722.
10. Giugliano RP, Wiviott SD, Blazing MA, De Ferrari GM, Park JG, Murphy SA, et al. Long-term safety and efficacy of achieving very low levels of low-density lipoprotein cholesterol: a prespecified analysis of the IMPROVE-IT trial. JAMA Cardiol 2017;2:547–555.
11. Giugliano RP, Pedersen TR, Park JG, De Ferrari GM, Gaciong ZA, Ceska R, et al. Clinical efficacy and safety of achieving very low LDL-cholesterol concentrations with the PCSK9 inhibitor evolocumab: a prespecified secondary analysis of the FOURIER trial. Lancet 2017;390:1962–1971.
12. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med 2015;372:2387–2397.
13. Van Heek M, France CF, Compton DS, McLeod RL, Yumibe NP, Alton KB, et al. In vivo metabolism-based discovery of a potent cholesterol absorption inhibitor, SCH58235, in the rat and rhesus monkey through the identification of the active metabolites of SCH48461. J Pharmacol Exp Ther 1997;283:157–163.
14. Morrone D, Weintraub WS, Toth PP, Hanson ME, Lowe RS, Lin J, et al. Lipid-altering efficacy of ezetimibe plus statin and statin monotherapy and identification of factors associated with treatment response: a pooled analysis of over 21,000 subjects from 27 clinical trials. Atherosclerosis 2012;223:251–261.
15. Ambegaonkar BM, Tipping D, Polis AB, Tomassini JE, Tershakovec AM. Achieving goal lipid levels with ezetimibe plus statin add-on or switch therapy compared with doubling the statin dose. A pooled analysis. Atherosclerosis 2014;237:829–837.
16. Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum CB, Eckel RH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association task force on practice guidelines. Circulation 2014;129(25 Suppl 2):S1–S45.
17. Kernan WN, Ovbiagele B, Black HR, Bravata DM, Chimowitz MI, Ezekowitz MD, et al. Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2014;45:2160–2236.
18. Jellinger PS, Handelsman Y, Rosenblit PD, Bloomgarden ZT, Fonseca VA, Garber AJ, et al. American Association of Clinical Endocrinologists and American College of Endocrinology guidelines for management of dyslipidemia and prevention of cardiovascular disease. Endocr Pract 2017;23(Suppl 2):1–87.
19. Krupp LB, LaRocca NG, Muir-Nash J, Steinberg AD. The fatigue severity scale. Application to patients with multiple sclerosis and systemic lupus erythematosus. Arch Neurol 1989;46:1121–1123.
20. Kim KJ, Kim SH, Yoon YW, Rha SW, Hong SJ, Kwak CH, 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.
21. Kim BK, Hong SJ, Lee YJ, Hong SJ, Yun KH, Hong BK, et al. Long-term efficacy and safety of moderate-intensity statin with ezetimibe combination therapy versus high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (RACING): a randomised, open-label, non-inferiority trial. Lancet 2022;400:380–390.
22. Amarenco P, Kim JS, Labreuche J, Charles H, Abtan J, Béjot Y, et al. A comparison of two LDL cholesterol targets after ischemic stroke. N Engl J Med 2020;382:9–19.
23. Amarenco P, Kim JS, Labreuche J, Charles H, Giroud M, Lee BC, et al. Yield of dual therapy with statin and ezetimibe in the Treat Stroke to Target trial. Stroke 2022;53:3260–3267.
24. Heo JH, Song D, Nam HS, Kim EY, Kim YD, Lee KY, et al. Effect and safety of rosuvastatin in acute ischemic stroke. J Stroke 2016;18:87–95.
25. Tamargo J, Kaski JC, Kimura T, Barton JC, Yamamoto K, Komiyama M, et al. Racial and ethnic differences in pharmacotherapy to prevent coronary artery disease and thrombotic events. Eur Heart J Cardiovasc Pharmacother 2022;8:738–751.

Article information Continued

Figure 1.

Trial flow. LDL-C, low-density lipoprotein cholesterol.

Figure 2.

Subgroup analyses of primary endpoint, according to post hoc subgroups. LDL-C reduction ≥50% (the primary end point) at 90 days among patients in the ROS10/EZT10 group and in the ROS20 group, according to post hoc subgroups. LDL-C, low-density lipoprotein cholesterol; OR, odds ratio; CI, confidence interval; DM, diabetes mellitus.

Table 1.

Baseline characteristics of the modified intention-to-treat population

ROS10/EZT10 (n=273) ROS20 (n=257) P
Age (yr) 63.9±11.9 63.7±11.9 0.8124
Male sex 207 (75.8) 180 (70.0) 0.1337
Onset to randomization (day) 4.8±4.3 4.4±3.1 0.1869
NIHSS at randomization 2.0 [1.0–4.0] 2.0 [1.0–4.0] 0.0580
Body-mass index (kg/m2) 23.9±3.2 24.5±3.6 0.0523
Systolic blood pressure (mm Hg) 142.9±19.4 140.3±23.3 0.1649
Diastolic blood pressure (mm Hg) 82.5±12.2 82.5±13.5 0.9826
Vascular risk factors
Hypertension 169 (61.9) 154 (59.9) 0.6401
Diabetes 83 (30.4) 73 (28.4) 0.6139
Current smoking 110 (40.3) 106 (41.2) 0.8236
Coronary heart disease 11 (4.0) 9 (3.5) 0.7502
Prior stroke history 14 (5.1) 18 (7.0) 0.3649
Lipid profiles (mg/dL)
Low-density lipoprotein cholesterol 130.2±34.7 131.0±33.9 0.7777
Total cholesterol 194.3±37.7 196.5±39.2 0.5191
Triglyceride 141.4±74.0 138.4±80.7 0.2033
High-density lipoprotein cholesterol 45.4±11.8 46.3±11.3 0.3791
Glucose (mg/dL) 130.4±43.1 113.3±25.2 0.1021
Glycated hemoglobin (%) 6.4±1.6 6.3±1.4 0.5385
Non-study statin use after hospitalization before randomization 193 (70.7) 188 (73.2) 0.5297

Data are presented as mean±standard deviation, n (%), or median [interquartile range].

NIHSS, National Institutes of Health Stroke Scale.

Table 2.

Endpoints in the modified intent-to-treat population

Endpoints ROS10/EZT10 (n=273) ROS20 (n=257) Odds ratio (95% confidence interval) P
Primary endpoint
LDL-C reduction ≥50% 198 (72.5) 148 (57.6) 1.944 (1.352–2.795) 0.0003
Secondary endpoints
LDL-C <70 mg/dL 219 (80.2) 168 (65.4) 2.148 (1.450–3.184) 0.0001
LDL-C reduction ≥50% or LDL-C <70 mg/dL 230 (84.2) 188 (73.2) 1.963 (1.281–3.008) 0.0018
Absolute LDL-C reduction (mg/dL) 72.7±37.0 64.7±35.1 0.0111
LDL-C reduction percentage (%) 53.9±21.1 47.9±20.5 0.0010
Multiple lipid goal achievement 194 (71.1) 138 (53.7) 2.117 (1.479–3.030) <0.0001
Major vascular events 1 (0.4) 9 (3.5) 0.101 (0.013–0.805) 0.0091
All death 0 (0.0) 1 (0.4) 0.469 (0.042–5.203) 0.4849
New onset diabetes 8 (4.3) 7 (3.8) 1.117 (0.397–3.147) 0.8334
Rhabdomyolysis 0 (0.0) 0 (0.0) >0.9999
Fatigue Severity Scale 14.5±10.0 14.1±8.9 0.5755
Significant liver enzyme elevation* 5 (1.8) 1 (0.4) 4.776 (0.554–41.16) 0.2174

Data are presented as mean±standard deviation or n (%).

LDL-C, low-density lipoprotein cholesterol.

*

Significant liver enzyme elevation: alanine aminotransferase or aspartate aminotransferase elevation >3 times from baseline.

Table 3.

Adverse events in the safety population

Event ROS10/EZT10 (n=295) ROS20 (n=287) P
Any adverse event 109 (36.9) 106 (36.9) 0.9969
TEAE 14 (4.7) 10 (3.5) 0.3814
Any serious adverse event 22 (7.5) 22 (7.7) 0.9244
Serious TEAE 0 (0) 0 (0) >0.9999

Data are presented as n (%).

TEAE, treatment-emergent adverse event.