Skip to main content
Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease logoLink to Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease
. 2022 Sep 8;11(18):e025551. doi: 10.1161/JAHA.122.025551

Network Meta‐Analysis of Randomized Trials Evaluating the Comparative Efficacy of Lipid‐Lowering Therapies Added to Maximally Tolerated Statins for the Reduction of Low‐Density Lipoprotein Cholesterol

Peter P Toth 1,, Sarah Bray 2, Guillermo Villa 3, Tamara Palagashvili 4,*, Naveed Sattar 5, Erik S G Stroes 6, Gavin M Worth 3,*
PMCID: PMC9683660  PMID: 36073669

Abstract

Background

Lowering low‐density lipoprotein cholesterol (LDL‐C) levels decreases major cardiovascular events and is recommended for patients at elevated cardiovascular risk. However, appropriate doses of statin therapy are often insufficient to reduce LDL‐C in accordance with current guidelines. In such cases, treatment could be supplemented with nonstatin lipid‐lowering therapy.

Methods and Results

A systematic literature review and network meta‐analysis were conducted on randomized controlled trials of nonstatin lipid‐lowering therapy added to maximally tolerated statins, including statin‐intolerant patients. The primary objective was to assess relative efficacy of nonstatin lipid‐lowering therapy in reducing LDL‐C levels at week 12. Secondary objectives included the following: LDL‐C level reduction at week 24 and change in non–high‐density lipoprotein cholesterol and apolipoprotein B at week 12. There were 48 randomized controlled trials included in the primary network meta‐analysis. All nonstatin agents significantly reduced LDL‐C from baseline versus placebo, regardless of background therapy. At week 12, evolocumab, 140 mg every 2 weeks (Q2W)/420 mg once a month, and alirocumab, 150 mg Q2W, were the most efficacious regimens, followed by alirocumab, 75 mg Q2W, alirocumab, 300 mg once a month, inclisiran, bempedoic acid/ezetimibe fixed‐dose combination, and ezetimibe and bempedoic acid used as monotherapies. Primary end point results were generally consistent at week 24, and for other lipid end points at week 12.

Conclusions

Evolocumab, 140 mg Q2W/420 mg once a month, and alirocumab, 150 mg Q2W, were consistently the most efficacious nonstatin regimens when added to maximally tolerated statins to lower LDL‐C, non–high‐density lipoprotein cholesterol, and apolipoprotein B levels and facilitate attainment of guideline‐recommended risk‐stratified lipoprotein levels.

Keywords: alirocumab, bempedoic acid, evolocumab, ezetimibe, inclisiran

Subject Categories: Clinical Studies, Cardiovascular Disease, Meta Analysis, Atherosclerosis, Lipids and Cholesterol


Nonstandard Abbreviations and Acronyms

ESC

European Society of Cardiology

FDC

fixed‐dose combination

LLT

lipid‐lowering therapy

mAb

monoclonal antibody

MACE

major adverse cardiovascular event

NMA

network meta‐analysis

PCSK9

proprotein convertase subtilisin/kexin type 9

Q2W

every 2 weeks

Q3M

every 3 months

Q6M

every 6 months

QD

once a day

QM

once a month

SLR

systematic literature review

Clinical Perspective.

What Is New?

  • There are few network meta‐analyses comparing the efficacy of PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors; this study compared PCSK9 inhibitors and new lipid‐lowering therapies (LLTs) (ie, bempedoic acid and inclisiran).

  • Primary network meta‐analysis found all nonstatin LLTs significantly reduced low‐density lipoprotein cholesterol levels versus placebo at week 12; broadly similar results for ranking of the interventions were identified in other lipid end point level changes.

  • Evolocumab, 140 mg every 2 weeks/420 mg once a month, and alirocumab, 150 mg every 2 weeks, treatment resulted in >70% of a simulated population achieving the very high‐risk cardiovascular disease European guideline goal (<55 mg/dL).

What Are the Clinical Implications?

  • Evolocumab and alirocumab, 150 mg every 2 weeks, are the most consistently efficacious nonstatin LLT regimens.

  • The network meta‐analysis of multiple nonstatin LLTs should help inform physicians' treatment choice for patients who would benefit from lower low‐density lipoprotein cholesterol levels and require adjuvant LLTs to statin therapy to achieve current low‐density lipoprotein cholesterol goals.

Low‐density lipoprotein cholesterol (LDL‐C) is an important causal, modifiable risk factor for atherosclerotic cardiovascular disease (ASCVD). 1 , 2 , 3 Evidence from multiple prospective, randomized studies has substantiated that patients achieving the lowest LDL‐C levels have the lowest risk of future major adverse cardiovascular events (MACEs), without associated safety concerns/adverse events, even when LDL‐C is reduced to very low levels (<40 mg/dL [<1 mmol/L]). 2 , 3 , 4 , 5 Managing LDL‐C in a risk‐stratified manner is particularly important in patients at high or very‐high risk of MACEs. 2 , 3

European guidelines now recommend even lower LDL‐C levels in groups at very‐high risk of ASCVD. 3 , 6 , 7 The 2019 European Society of Cardiology (ESC)/European Atherosclerosis Society guidelines and the more recent 2021 ESC guidelines recommend an LDL‐C goal of <55 mg/dL (<1.4 mmol/L) and ≥50% LDL‐C reduction from baseline for very high‐risk patients, whereas US guidelines suggest high‐intensity or maximal statin therapy, with addition of ezetimibe and/or PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor if LDL‐C remains above the threshold of ≥70 mg/dL (≥1.8 mmol/L). 2 , 3 , 7

Although treatment with statins is the predominant treatment for elevated LDL‐C, statin therapy does not allow some patients to achieve LDL‐C values of <55 mg/dL, 2 , 7 some patients may not tolerate statins at higher intensities, 8 and there are a minority of others who are reportedly intolerant entirely, 9 although recent data have highlighted an important role for the nocebo effect in patients with perceived adverse effects following statin therapy. 10 In some patients, even high‐intensity statins alone may not be enough. Where required, statins can be supplemented by nonstatin agents, such as ezetimibe and/or PCSK9 inhibitors, to further optimize LDL‐C reduction. 3 , 7 Recent real‐world European data suggest that LDL‐C goals are rarely met among very high‐risk patients; only 18% (≈1 of 6) of very high‐risk patients achieved the 2019 European guideline LDL‐C goal of <55 mg/dL (<1.4 mmol/L), whereas 39% met the 2016 goal of <70 mg/dL (<1.8 mmol/L). 11 A prospective observational study in the United States looking at treatment patterns over 2 years in patients with ASCVD identified that at 2 years of follow‐up 31.7% of patients overall had LDL‐C levels <70 mg/dL (<1.8 mmol/L). 12 The increased use of nonstatin agents in combination with statins may help higher‐risk patients meet the LDL‐C levels recommended by current guidelines.

There are limited head‐to‐head comparative trial data for nonstatin lipid‐lowering therapies (LLTs) (focused on PCSK9 inhibitors versus other nonstatin LLTs, including newer agents); therefore, indirect treatment comparisons through network meta‐analysis (NMA) may inform evidence‐based treatment decisions. We previously used NMA to compare LDL‐C reduction in nonstatin LLTs, including evolocumab, alirocumab, and ezetimibe, in patients receiving statin background therapy. 13

Since this earlier publication, new studies of the approved monoclonal antibody (mAb) PCSK9 inhibitors alirocumab and evolocumab have been published, and 2 new LLTs have emerged: bempedoic acid, 14 , 15 , 16 , 17 , 18 an ATP citrate lyase inhibitor that has been approved by the US Food and Drug Administration 19 and the European Medicines Agency 20 ; and inclisiran, 21 , 22 , 23 a small interfering RNA (siRNA) PCSK9 inhibitor that has been approved by the European Medicines Agency 24 , 25 and the US Food and Drug Administration. 26 This systematic review and NMA sought to provide a detailed assessment of the relative efficacy of nonstatin agents in reducing LDL‐C.

METHODS

Amgen holds the source data, and all authors had full access to the data. All data are presented in the article and supplementary information.

Objectives

The primary objective of this NMA was to assess the comparative efficacy of nonstatin agents (bempedoic acid, ezetimibe, mAb PCSK9 inhibitors [alirocumab and evolocumab], and siRNA PCSK9 inhibitor [inclisiran]) to reduce LDL‐C (percentage change from baseline at week 12) when added to maximally tolerated statins. Secondary objectives of the study were to assess the reduction in LDL‐C over a longer follow‐up period (percentage change from baseline at week 24), to assess the change in other lipid parameters relevant to ASCVD, including non–high‐density lipoprotein cholesterol (HDL‐C) and apolipoprotein B (ApoB) (percentage change from baseline at week 12), and to analyze the impact of treating a hypothetical population with each intervention on LDL‐C levels to assess the proportion of values that meet the current European guideline‐recommended LDL‐C goal of <55 mg/dL (<1.4 mmol/L).

Study Design

Systematic Literature Review

The systematic literature review (SLR) adhered to methods published by the Centre for Reviews and Dissemination 27 and the Cochrane Collaboration. The Population, Intervention, Comparison, and Outcomes inclusion/exclusion criteria for the SLR are shown in Table 1. Randomized trials were relevant if they compared at least 2 relevant interventions: alirocumab, bempedoic acid, bempedoic acid/ezetimibe fixed‐dose combination (FDC), evolocumab, ezetimibe, inclisiran, or placebo. Trials were included if they enrolled adults (aged ≥18 years) with primary (including familial and nonfamilial) hypercholesterolemia or ASCVD, who require treatment of hyperlipidemia, and were at least 12 weeks in duration with at least 10 patients per study arm.

Table 1.

Inclusion/Exclusion Criteria for the Systematic Literature Review

PICO criteria Inclusion criteria Exclusion criteria
Population

Adults (aged ≥18 y) with primary (including familial and nonfamilial) hypercholesterolemia who require treatment of elevated lipid levels (hyperlipidemia) and are receiving maximally tolerated statins (defined as moderate‐ to high‐intensity statin therapy [per AHA‐ACC] or where lower‐intensity/no statin patients are declared to be statin intolerant*)

For the HoFH subgroup only, patients aged ≥12 y were also eligible for inclusion

Interventions or comparators

Trials comparing at least 2 interventions or comparators of interest:

evolocumab, alirocumab, ezetimibe, bempedoic acid, bempedoic acid/ezetimibe fixed‐dose combination, inclisiran, placebo

Trials including unlicensed doses or regimens
Outcomes

Trials reporting relevant data for at least one of the following outcomes were considered for inclusion:

Lipid outcomes
  • Percentage reduction from baseline in LDL‐C
  • Absolute reduction in LDL‐C from baseline
  • Percentage reduction from baseline in non–HDL‐C
  • Percentage people achieving LDL‐C <70 mg/dL (<1.8 mmol/L)
  • Percentage reduction from baseline in Lpa
  • Percentage reduction from baseline in ApoB
  • Percentage reduction from baseline in total cholesterol
  • Percentage reduction from baseline in triglycerides
Time points
  • Wk 12 or the nearest time point to 12 wk
  • Wk 24 or the nearest time point to 24 wk
  • Wk 48 or the nearest time point to 48 wk
Adverse events
  • Any treatment‐emergent AE
  • Any serious treatment‐emergent AE
  • Any fatal AE
  • AEs leading to discontinuation
  • AEs of interest:
    • Muscle symptoms (specifically, myopathy and rhabdomyolysis)
    • New‐onset diabetes
    • Elevated creatine kinase
    • Neurocognitive AEs
Study design RCTs of at least 12 wk in duration

Trials with <10 participants and preclinical and animal trials

Trials including patients with significant heart failure (NYHA grade III–IV) or significant renal dysfunction (stage 4–5)

ACC indicates American College of Cardiology; AE, adverse event; AHA, American Heart Association; ApoB, apolipoprotein B; HDL‐C, high‐density lipoprotein cholesterol; HoFH, homozygous familial hypercholesterolemia; LDL‐C, low‐density lipoprotein cholesterol; Lpa, lipoprotein a; NYHA, New York Heart Association; PICO, Population, Intervention, Comparison, and Outcomes; and RCT, randomized controlled trial.

*

Definition of statin intensity is less clear in East Asian countries, and lower doses are generally used compared with Western countries. Trials in which populations received lower‐intensity statin therapy than those defined by ACC‐AHA were therefore eligible if conducted in East Asian populations.

Databases searched to identify all relevant records were PUBMED, EMBASE, Cochrane Database of Systematic Reviews, Cochrane Central Register of Controlled Trials, Database of Abstracts of Reviews of Effects, and the Health Technology Assessment Database (an example search strategy for PUBMED is shown in Data S1). The EMBASE search strategies for each set of searches were independently peer reviewed by a second information specialist, using the Canadian Agency for Drugs and Technologies in Health peer review checklist. 28 All data were extracted by one reviewer and independently checked for errors by a second reviewer during the SLR data extraction. Individual patient data were not available for analysis. The risk of bias was assessed using the Cochrane Risk of Bias Assessment Tool.

Network Meta‐Analysis

Trials identified in the SLR were eligible for NMAs if they reported data on LDL‐C, non–HDL‐C, and/or ApoB. Trials were excluded from NMAs if they were specifically conducted in patients with homozygous familial hypercholesterolemia, if <70% of participants were on moderate‐to high‐intensity statins (unless apparently statin intolerant), or if insufficient data were available (eg, variability estimates).

Evolocumab doses of 140 mg every 2 weeks (Q2W) and 420 mg once a month (QM) are clinically similar 29 , 30 , 31 ; therefore, data for both regimens were combined in a single node in the network. This is not the case for alirocumab, where 75 mg Q2W, 300 mg QM, and 150 mg Q2W regimens are not clinically equivalent 32 , 33 , 34 , 35 ; hence, these regimens were included as separate nodes.

The primary network was based on the percentage LDL‐C reduction achieved at week 12. When week 12 data were not available, the nearest time point after week 12 was used. For evolocumab, data for the mean of weeks 10 and 12 were used to ensure the efficacy of monthly dosing was accurately reflected (representing the average LDL‐C reduction across the extended dosing period). Inclisiran is also dosed at extended intervals: days 1 and 90, and in some trials dosing continued on days 270 and 450. 21 , 22 , 23 LDL‐C reduction with inclisiran is maximized at day 150 (ie, around week 21). 25 The coprimary end point, time‐adjusted LDL‐C reduction between days 90 and 540, was used and, given the extended dosing interval, was assumed to provide the most appropriate estimate of inclisiran efficacy rather than using a single time point.

In the NMA focused on time points after week 12 (ie, week 24), data for the nearest time point after the defined follow‐up period were used where necessary. Alirocumab trials of 75 mg Q2W, which allowed up titration at week 12, were excluded from the week 24 data set.

Trials eligible for the NMAs were assessed for clinical and statistical heterogeneity. In the absence of significant treatment effect modifiers one efficacy for reducing LDL‐C, it was considered feasible to combine all eligible trials reporting LDL‐C data in a network. Statistical heterogeneity was assessed through visual inspection of forest plots from pairwise direct meta‐analyses of agents within the network (overlap of effect sizes and 95% CIs) and through subsequent I2 and the χ2 tests (I2>75% may indicate meaningful heterogeneity). The impact of clinical and statistical heterogeneity was explored through sensitivity analyses that excluded individual trials or groups of trials associated with heterogeneity. Consistency of effect was also explored through subgroup analysis by statin background therapy and ASCVD status by including only trials with at least a 50% ASCVD population.

The NMA was conducted using frequentist methods using the Netmeta R package 36 and Bayesian models 37 in WinBUGS (MRC Biostatistics Unit, Cambridge, UK) version 1.4.3. Bayesian analyses were performed as described previously in Toth et al. 13 All analyses used the mean difference between groups and SE (as opposed to the mean and SE for each group). Where >1 treatment difference was reported for a pair of treatments for a single study (eg, for 2 different statin backgrounds), meta‐analysis was used to estimate a single treatment difference for each treatment pair within each study. A random‐effects model was used throughout. Local inconsistency was explored using the Netmeta R package by splitting the network estimates into the contribution of direct and indirect evidence. Network graphs were produced to visualize the weight of evidence and number of trials connecting each pair of treatments.

Simulation of LDL‐C Lowering With Each Intervention

The impact of treating a population with each intervention was explored using simulation techniques. Using the DA VINCI (The EU‐Wide Cross‐Sectional Observational Study of Lipid Modifying Therapy Use in Secondary and Primary Care) study, 11 a European Union–wide cross‐sectional observational study of lipid‐modifying therapy use in secondary and primary care, we estimated the mean (and SD) LDL‐C for the group of patients with ASCVD receiving stabilized statin therapy without ezetimibe or PCSK9i. Assuming a normal distribution, we simulated 10 000 LDL‐C values, and values >70 mg/dL (>1.8 mmol/L) were selected to represent a hypothetical pool of patients requiring additional LLT. We then simulated the LDL‐C reduction achieved by each intervention by randomly sampling from a normal distribution with mean estimated from the primary NMA and SD estimate obtained from a specific clinical trial (Table S1). The NMA does not provide any information about the variability between individuals; hence, to estimate the SD, a single clinical trial was selected for each intervention, with a preference given to the largest study with the time point closest to 12 weeks. For ezetimibe, only studies that included patients stabilized on statin at randomization were considered as a source for the SD, because, for the studies that included patients not stabilized on statin at randomization, percentage reduction in LDL‐C from baseline of ezetimibe versus placebo had been derived from the data provided (statin plus ezetimibe versus statin plus placebo). The posttreatment LDL‐C was calculated, and it was assessed whether each value decreased below 55 mg/dL (1.4 mmol/L), the goal recommended in the 2019 ESC/European Atherosclerosis Society guidelines for very high‐risk patients (eg, those who have ASCVD). 3

RESULTS

Network Construction

The SLR initially identified 5377 records from databases and other sources. This was refined using title and abstract screening and full‐text screening to give 55 relevant trials for the SLR. Further refinement excluded 7 trials, resulting in 48 randomized controlled trials for inclusion in the primary NMA (Figure 1). Of these, 10 trials were phase 2 trials, 36 trials were phase 3 trials, 1 trial was a phase 3b/4 trial, and 1 trial was unclear. The reasons for exclusion of the 7 trials from the NMA are outlined in Table S2, with the main reasons being ineligible population and/or study design and insufficient data.

Figure 1. Study flow diagram of the systematic review.

Figure 1

Trials included in the network meta‐analysis (NMA) included those with patients either receiving background statin treatment or who were statin intolerant. SLR indicates systematic literature review.

The details of the trials included in the primary NMA are shown in Table 2, 14 , 16 , 17 , 18 , 21 , 22 , 23 , 32 , 34 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 , 49 , 50 , 51 , 52 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61 , 62 , 63 , 64 , 65 , 66 , 67 , 68 , 69 , 70 , 71 , 72 , 73 , 74 and the overall network diagram is shown in Figure 2.

Table 2.

Details of Trials Included in the Primary Network

Study Arms (number) Population Duration

McKenney 2012 38

NCT01288443

Alirocumab, 150 mg Q2W (n=31)

vs

placebo for alirocumab, Q2W (n=31)

Hypercholesterolemia

Background: statins

12 wk

ODYSSEY ALTERNATIVE 39

NCT01709513

Alirocumab, 75 mg Q2W (possible up titration to 150 mg Q2W)+placebo for ezetimibe, QD (n=126)

vs

ezetimibe, 10 mg QD+placebo for alirocumab, Q2W (n=126)

Hypercholesterolemia

Background: NR

24 wk

ODYSSEY CHOICE I 34

NCT01926782

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W) (n=78)

vs

placebo for alirocumab, QM (results not reported for placebo, Q2W, arm) (n=157)

Hypercholesterolemia

Background: statins (maximum tolerated), other LLT (including ezetimibe)

48 wk

ODYSSEY CHOICE II 40

NCT02023879

Alirocumab, 75 mg Q2W (possible up titration to 150 mg Q2W) (n=116)

vs

placebo for alirocumab, Q2W (n=58)

Hypercholesterolemia

Background: ezetimibe

24 wk

ODYSSEY COMBO I 32

NCT01644175

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W) (n=107)

vs

placebo (n=209)

Hypercholesterolemia

Background: statins (maximum tolerated), no statins (statin intolerant)

52 wk

ODYSSEY COMBO II 41

EUCTR2011‐004130‐34

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W)+placebo for ezetimibe, QD (n=479)

vs

ezetimibe, 10 mg QD+placebo for alirocumab, Q2W (n=241)

Hypercholesterolemia

Background: statins (maximum tolerated), no statin (statin intolerant)

104 wk

ODYSSEY DM INSULIN 42

NCT02585778, EUCTR2015‐000799‐92

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W) (n=345)

vs

placebo for alirocumab, Q2W (n=172)

Hypercholesterolemia, diabetes

Background: statins (maximum tolerated), other LLT

24 wk

ODYSSEY EAST 43

NCT02715726

Alirocumab, 75 mg Q2W (possibly up titrated to 150 mg Q2W) (n=407)

vs

ezetimibe, 10 mg QD (n=208)

ASCVD

Background: statins (maximum tolerated)

24 wk

ODYSSEY FH I 44

NCT01623115

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W) (n=323)

vs

placebo for alirocumab, Q2W (n=163)

HeFH

Background: statins (maximum tolerated), other LLT (including ezetimibe)

78 wk

ODYSSEY FH II 44

NCT01709500

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W) (n=167)

vs

placebo for alirocumab, Q2W (n=82)

HeFH

Background: statins (maximum tolerated), other LLT (including ezetimibe)

78 wk

ODYSSEY HIGH FH 45

NCT01617655

Alirocumab, 150 mg Q2W (n=72)

vs

placebo for alirocumab, Q2W (n=35)

HeFH

Background: statins (maximum tolerated), other LLT (including ezetimibe)

78 wk

ODYSSEY JAPAN 46

NCT02107898

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W) (n=144)

vs

placebo (n=72)

Hypercholesterolemia, HeFH

Background: statins, other LLT

12 wk

ODYSSEY KT 47

NCT02289963

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W) (n=97)

vs

placebo for alirocumab, Q2W (n=102)

ASCVD, diabetes, hypercholesterolemia

Background: statins (maximum tolerated), other LLT (including ezetimibe)

24 wk

ODYSSEY LONG TERM 48

NCT01507831

Alirocumab, 150 mg Q2W (n=1553)

vs

placebo for alirocumab, Q2W (n=788)

Hypercholesterolemia

Background: statins (maximum tolerated), no statin (statin intolerant), other LLT

78 wk

ODYSSEY NIPPON 49

NCT02584504

Alirocumab, 150 mg Q2W (n=53)

vs

placebo for alirocumab, Q2W (n=56)

Hypercholesterolemia

Background: low/no statin (statin intolerant), ezetimibe, other LLT

12 wk

ODYSSEY OPTIONS I 50

NCT01730040

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W)+placebo for ezetimibe, QD (n=104)

vs

ezetimibe, 10 mg QD+placebo for alirocumab, Q2W (n=102)

vs

placebo for alirocumab, Q2W+placebo for ezetimibe, QD (n=149)

Hypercholesterolemia

Background: statins

24 wk

ODYSSEY OPTIONS II 51

NCT01730053

Alirocumab, 75 mg Q2W (possible dose adjustment to 150 mg Q2W)+placebo for ezetimibe, QD (n=103)

vs

ezetimibe, 10 mg QD+placebo for alirocumab, Q2W (n=101)

vs

placebo for alirocumab, Q2W+placebo for ezetimibe, QD (n=101)

Hypercholesterolemia

Background: statins

24 wk

Stein 2012 52

NCT01266876

Alirocumab, 150 mg Q2W (n=16)

vs

placebo for alirocumab, Q2W (n=15)

HeFH

Background: statins

12 wk

Teramoto 2016 53

NCT01812707

Alirocumab, 75 mg Q2W (n=25)

vs

alirocumab, 150 mg Q2W (n=25)

vs

placebo for alirocumab, Q2W (n=25)

Hypercholesterolemia

Background: statins

12 wk

Ballantyne 2016 54

NCT02072161

Bempedoic acid, 180 mg QD (n=45)

vs

placebo (n=45)

Hypercholesterolemia

Background: statins

12 wk

CLEAR HARMONY 18

NCT02666664, EUCTR2015‐004136‐36

Bempedoic acid, 180 mg QD (n=1488)

vs

placebo for bempedoic acid, QD (n=742)

ASCVD and/or HeFH

Background: statins (maximum tolerated), other LLT

52 wk

CLEAR SERENITY 17

NCT02988115

Bempedoic acid, 180 mg QD (n=234)

vs

placebo for bempedoic acid, QD (n=111)

Hypercholesterolemia

Background: none (statin intolerant)

24 wk

CLEAR TRANQUILITY 14

EUCTR2016‐004084‐39, NCT03001076

Bempedoic acid, 180 mg QD (n=181)

vs

placebo for bempedoic acid, QD (n=81)

Hypercholesterolemia

Background: none (statin intolerant)

12 wk

CLEAR WISDOM 16

NCT02991118

Bempedoic acid, 180 mg QD (n=522)

vs

placebo (n=257)

ASCVD and/or HeFH

Background: LLT (maximum tolerated), statin, no LLT

52 wk

Ballantyne 2020 FDC 55

NCT03337308

Bempedoic acid, 180 mg QD+ezetimibe, 10 mg QD (fixed‐dose combination) (n=108 randomized, 86 analyzed)

vs

bempedoic acid, 180 mg QD (n=110 randomized, 88 analyzed)

vs

ezetimibe, 10 mg QD (n=109 randomized, 86 analyzed)

vs

placebo, QD (n=55 randomized, 41 analyzed)

Hypercholesterolemia, high risk

Background: all (statin intolerant) after no statins

12 wk

Thompson 2016 (statin‐intolerant group only) 56

NCT01941836

Bempedoic acid, 180 mg QD (n=51)

vs

bempedoic acid, 180 mg QD+ezetimibe, 10 mg QD (n=12)

vs

ezetimibe, 10 mg QD (n=51)

Hypercholesterolemia

Background: none (statin intolerant)

12 wk

BANTING 57

NCT02739984, EUCTR2015‐004711‐21

Evolocumab, 420 mg QM (n=281)

vs

placebo for evolocumab, QM (n=143)

Hypercholesterolemia, diabetes

Background: statins (maximum tolerated)

12 wk

BERSON 58

NCT02662569

Evolocumab, 140 mg Q2W (n=327)

vs

placebo for evolocumab, Q2W (n=166)

vs

evolocumab, 420 mg QM (n=332)

vs

placebo for evolocumab, QM (n=161)

Hypercholesterolemia, diabetes

Background: statins

12 wk

DESCARTES/Amgen 20110109 59

NCT01516879

Evolocumab, 420 mg QM (n=402)

vs

placebo for evolocumab, QM (n=202)

vs

evolocumab, 420 mg QM+ezetimibe, 10 mg QD (n=126)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, QM (n=63)

Hypercholesterolemia

Background: statins

48 wk

FOURIER/Amgen 20160250 60

NCT01764633

Evolocumab, 140 mg Q2W/420 mg QM (n=13 784)

vs

placebo for evolocumab, Q2W or QM (n=13 780)

ASCVD

Background: statins, ezetimibe

Median, 26 mo

GAUSS/Amgen 20090159 61

NCT01375764

Evolocumab, 420 mg QM (n=32)

vs

evolocumab, 420 mg QM+ezetimibe, 10 mg QD (n=31)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, QM (n=33)

Hypercholesterolemia

Background: none (statin intolerant)

12 wk

GAUSS‐2/Amgen 20110116 62

NCT01763905

Evolocumab, 140 mg Q2W+placebo for ezetimibe, QD (n=103)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, Q2W (n=51)

vs

evolocumab, 420 mg QM+placebo for ezetimibe, QD (n=102)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, QM (n=51)

Hypercholesterolemia

Background: none (statin intolerant)

12 wk

GAUSS‐3 (part B)/Amgen 20120332 63

NCT01984424

Evolocumab, 420 mg QM+placebo for ezetimibe, QD (n=145)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, QM (n=73)

Hypercholesterolemia

Background: none (statin intolerant)

24 wk

GAUSS‐4 64

NCT02634580

Evolocumab, 140 mg Q2W+placebo for ezetimibe, QD (n=19)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, Q2W (n=10)

vs

evolocumab, 420 mg QM+placebo for ezetimibe, QD (n=21)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, QM (n=11)

Hypercholesterolemia

Background: none (statin intolerant)

12 wk

GLAGOV/Amgen 20120153 65

NCT01813422

Evolocumab, 420 mg QM (n=484)

vs

placebo for evolocumab, QM (n=486)

ASCVD

Background: statins

Median, 78 wk

LAPLACE‐2/Amgen 20110115 66

NCT01763866

Evolocumab, 140 mg Q2W+placebo for ezetimibe, QD (n=394)

vs

placebo for evolocumab, Q2W±placebo for ezetimibe, QD (n=183)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, Q2W (n=80)

vs

evolocumab, 420 mg QM±placebo for ezetimibe, QD (n=391)

vs

placebo for evolocumab, QM±placebo for ezetimibe, QD (n=193)

vs

ezetimibe, 10 mg QD+placebo for evolocumab, QM (n=68)

Hypercholesterolemia

Background: statins

52 wk

LAPLACE‐TIMI 57/Amgen 20101155 67

NCT01380730

Evolocumab, 140 mg Q2W (n=78)

vs

placebo for evolocumab, Q2W (n=78)

vs

evolocumab, 420 mg QM (n=80)

vs

placebo for evolocumab, QM (n=79)

Hypercholesterolemia

Background: statins

12 wk

RUTHERFORD/Amgen 20090158 68

NCT01375751

Evolocumab, 420 mg QM (n=56)

vs

placebo for evolocumab, QM (n=56)

HeFH

Background: statins, ezetimibe, other LLT

12 wk

RUTHERFORD‐2/Amgen 20110117 69

NCT01763918

Evolocumab, 140 mg Q2W (n=111)

vs

placebo for evolocumab, Q2W (n=55)

vs

evolocumab, 420 mg QM (n=110)

vs

placebo for evolocumab, QM (n=55)

HeFH

Background: statins, ezetimibe, other LLT

12 wk

YUKAWA/Amgen 20110231 70

NCT01652703

Evolocumab, 140 mg Q2W (n=52)

vs

placebo for evolocumab, Q2W (n=52)

vs

evolocumab, 420 mg QM (n=51)

vs

placebo for evolocumab, QM (n=53)

Hypercholesterolemia

Background: statins, ezetimibe

12 wk

YUKAWA‐2/Amgen 20120122 71

NCT01953328

Evolocumab, 140 mg Q2W (n=101)

vs

placebo for evolocumab, Q2W (n=101)

vs

evolocumab, 420 mg QM (n=101)

vs

placebo for evolocumab, QM (n=101)

Hypercholesterolemia

Background: statins

12 wk

ENHANCE 72

NCT00552097

Ezetimibe, 10 mg QD (n=357)

vs

placebo for ezetimibe, QD (n=363)

HeFH

Background: statins

104 wk

IMPROVE‐IT 73

NCT00202878

Ezetimibe, 10 mg QD (n=9067)

vs

placebo for ezetimibe, QD (n=9077)

ASCVD

Background: statins

Median, 6 y
Masana 2005 74

Ezetimibe, 10 mg QD (n=355)

vs

placebo for ezetimibe, QD (n=78)

Hypercholesterolemia

Background: statins

48 wk

ORION‐1 23

NCT02597127

Inclisiran, 300 mg (n=62)

vs

placebo for inclisiran (n=65)

Hypercholesterolemia

Background: statins (maximum tolerated), other LLT (including ezetimibe)

26.7 wk

ORION‐9 21

NCT03397121

Inclisiran‐free acid, 284 mg (inclisiran sodium, 300 mg) (n=242)

vs

placebo (n=240)

HeFH

Background: statins (maximum accepted), ezetimibe

77.1 wk

ORION‐10 22

NCT03399370

Inclisiran‐free acid, 284 mg (inclisiran sodium, 300 mg) (n=781)

vs

placebo (n=780)

ASCVD

Background: statins (maximum tolerated), ezetimibe, other LLT

77.1 wk

ORION‐11 22

NCT03400800

Inclisiran‐free acid, 284 mg (inclisiran sodium, 300 mg) (n=810)

vs

placebo (n=807)

ASCVD

Background: statins (maximum tolerated), ezetimibe, other LLT

77.1 wk

ASCVD indicates atherosclerotic cardiovascular disease; HeFH, heterozygous familial hypercholesterolemia; LLT, lipid‐lowering therapy; NR, not reported; Q2W, every 2 weeks; QD, once a day; and QM, once a month.

Figure 2. Primary network: connection of eligible randomized controlled trials reporting percentage change in low‐density lipoprotein cholesterol from baseline to week 12.

Figure 2

The diameter of each circle represents the proportional total weight of all trials in the network that investigated that intervention. The thickness of each line connecting 2 interventions is proportional to the number of trials that investigated that pair of interventions. FDC indicates fixed‐dose combination; Q2W, every 2 weeks; Q3M, every 3 months; Q6M, every 6 months; QD, once a day; QM, once a month; and RCT, randomized controlled trial.

The risk of bias was analyzed to assess the quality of each study. Overall, the risk of bias for the 48 trials in the primary network was generally low or unclear in 31 and 14 trials, respectively. The most common areas where reporting was unclear were allocation of concealment and randomization methods. Some trials (n=4) were observed to have high potential for bias with regard to incomplete reporting of outcomes; however, it was possible to source the required information from ClinicalTrials.gov, and therefore, in practice, risk of bias was low. No studies were excluded from the NMA on the risk of bias. The complete risk of bias assessment is presented in Table S3.

Direct meta‐analyses combining specific trials comparing relevant interventions within the network are shown in Figure S1. I2 values generally indicated high levels of heterogeneity; however, this finding is influenced by narrow CIs from individual trials, resulting in less overlap. Visual inspection indicated some possible heterogeneity associated with East Asian trials and with certain trials in populations with familial hypercholesterolemia. Because this is also clinically plausible, sensitivity analyses for the primary end point excluding these trials were performed.

Table S4 details which trials from the overall network were included in the sensitivity analyses, subgroup analyses, and the secondary objective analyses at week 24 and for the other lipid end points. Although an analysis at week 48 was considered, it was deemed unfeasible because of relatively few data being reported at this time point and, most important, because of differences between trials in approaches to handling missing data over the longer follow‐up period.

Primary Objective: The Placebo‐Corrected LDL‐C Reduction at Week 12

All interventions significantly reduced LDL‐C when compared with placebo (Figure 3). The treatment differences between LLT and placebo for the percentage reduction in LDL‐C from baseline to week 12, or time‐average data in the case of inclisiran, show that among the mAb PCSK9 inhibitors, evolocumab, 140 mg Q2W/420 mg QM, reduced LDL‐C by a mean of 64.68% (95% CI, 67.37%–62.00%), whereas alirocumab, 150 mg Q2W, reduced LDL‐C by a mean of 62.71% (95% CI, 67.56%–57.87%) (Figure 3). Both alirocumab, 75 mg Q2W and 300 mg QM, dosages resulted in a smaller reduction in LDL‐C than 150 mg Q2W. Treatment with 300 mg of siRNA PCSK9 inhibitor inclisiran every 3 months and then every 6 months (Q3M to Q6M) was found to reduce LDL‐C by a mean of 50.17% (95% CI, 54.99%–45.35%). Treatment with ezetimibe, 10 mg once a day (QD), reduced LDL‐C by a mean of 24.49% (95% CI, 27.48%–21.49%) from baseline; and for bempedoic acid, 180 mg QD, LDL‐C was reduced by a mean of 22.83% (95% CI, 26.83%–18.82%). The FDC of ezetimibe, 10 mg, and bempedoic acid, 180 mg, was almost 2‐fold more efficacious than either treatment individually, reducing LDL‐C by a mean of 42.93% (95% CI, 49.96%–35.80%).

Figure 3. The mean difference (MD) in percentage change in low‐density lipoprotein cholesterol (LDL‐C) in response to lipid‐lowering therapy relative to placebo at week 12.

Figure 3

FDC indicates fixed‐dose combination; Q2W, every 2 weeks; Q3M, every 3 months; Q6M, every 6 months; QD, once a day; and QM, once a month.

The primary analysis was performed using frequentist methods. When Bayesian methods were used, similar results were observed (data not shown), which confirmed the frequentist approach.

A pairwise comparison between the agents in the primary network was performed (Table S5) and identified that the reduction in LDL‐C was greater for evolocumab, 140 mg Q2W/420 mg QM, and alirocumab, 150 mg Q2W, compared with each of the other LLTs. Alirocumab, 75 mg Q2W, alirocumab, 300 mg QM, and inclisiran, 300 mg Q3M to Q6M, were not significantly more efficacious than one another. Of these, alirocumab, 75 mg Q2W, was significantly more efficacious than the bempedoic acid/ezetimibe FDC, whereas alirocumab, 300 mg QM, and inclisiran, 300 mg Q3M to Q6M, were not. All these agents were more efficacious than bempedoic acid and ezetimibe alone.

Subgroup Analyses

In 38 of the trials included in the primary NMA, patients received background statin therapy. A subgroup analysis of these trials looking at the mean difference in percentage change in LDL‐C from baseline relative to placebo at week 12 found similar results to the overall population (Figure 4), with evolocumab, 140 mg Q2W/420 mg QM, and alirocumab, 150 mg Q2W, being the most efficacious (65.44% and 61.94% change in LDL‐C from baseline, respectively), followed by other alirocumab doses, inclisiran, and then the bempedoic acid/ezetimibe FDC and its components. A similar pattern was found in an NMA including the 10 trials in patients reporting statin intolerance (Figure S2); however, these findings are based on a much smaller evidence base (lower number of trials and sample size) and a more disconnected network. Furthermore, all the evidence for evolocumab, ezetimibe, and the bempedoic acid/ezetimibe FDC compared with placebo is indirect. These data therefore have greater levels of uncertainty.

Figure 4. Subgroup analysis: the mean difference (MD) in percentage change in low‐density lipoprotein cholesterol (LDL‐C) from baseline in response to lipid‐lowering therapy relative to placebo at week 12 in patients receiving statin background therapy (moderate‐high intensity) (blue), with the primary analysis data plotted for comparison (gray).

Figure 4

FDC indicates fixed‐dose combination; Q2W, every 2 weeks; Q3M, every 3 months; Q6M, every 6 months; QD, once a day; and QM, once a month.

Unlike other agents, the percentage reduction of LDL‐C levels by bempedoic acid was influenced by statin background therapy. Bempedoic acid reduced LDL‐C levels to a lesser extent in the subgroup analysis focusing on trials with moderate‐ to high‐intensity statin background therapy, than it did in the overall analyses, which also included trials that declared lower intensities/no statin patients to be statin intolerant.

A further subgroup NMA included 17 trials that enrolled predominately patient populations with ASCVD (>50% patients with ASCVD included) (Figure 5). Again, analysis of these trials showed a similar trend to the main NMA, with evolocumab, 140 mg Q2W/420 mg QM, and alirocumab, 150 mg Q2W, being most efficacious (62.80% and 64.80% LDL‐C reduction, respectively), followed by alirocumab, 75 mg Q2W (53.97%), and inclisiran (52.11%). Bempedoic acid and ezetimibe were least efficacious, with percentage LDL‐C reduction of 17.96% and 24.63%, respectively.

Figure 5. Subgroup analysis: the mean difference (MD) in percentage change in low‐density lipoprotein cholesterol (LDL‐C) from baseline in response to lipid‐lowering therapy relative to placebo at week 12 in predominantly populations with atherosclerotic cardiovascular disease (ASCVD) (blue), with the primary analysis data plotted for comparison (gray).

Figure 5

FDC indicates fixed‐dose combination; Q2W, every 2 weeks; Q3M, every 3 months; Q6M, every 6 months; QD, once a day; and QM, once a month.

Sensitivity Analyses

Sensitivity analyses were conducted for the primary network, excluding potential sources of statistical and clinical heterogeneity (ie, trials in familial hypercholesterolemia or East Asian populations). The 8 trials involving patients with familial hypercholesterolemia and the 8 trials with East Asian populations were excluded in separate analyses. The trials included in the analyses are detailed in Table S4. The exclusion of these trials from the meta‐analysis provided similar results to the overall NMA (Figure 6).

Figure 6. Sensitivity analyses: treatment difference in percentage change in low‐density lipoprotein cholesterol (LDL‐C) from baseline in response to lipid‐lowering therapy relative to placebo at week 12, excluding trials featuring familial hypercholesterolemia (FH) (A) or East Asian populations (B) (blue), with the primary analysis data plotted for comparison (gray).

Figure 6

FDC indicates fixed‐dose combination; MD, mean difference; Q2W, every 2 weeks; Q3M, every 3 months; Q6M, every 6 months; QD, once a day; and QM, once a month.

Secondary Objectives

Placebo‐Corrected LDL‐C Reduction at Week 24

Of the 48 trials in the primary NMA, 16 were included in an analysis to assess the reduction in LDL‐C levels compared with placebo at week 24 (Table S4). All interventions significantly reduced LDL‐C at week 24 (Table 3). Among the PCSK9 inhibitors, evolocumab was the most efficacious (61.84% change from baseline), followed by alirocumab, 300 mg QM, alirocumab, 150 mg Q2W, and inclisiran (58.70%, 57.11%, and 50.07%, respectively). As with the primary NMA, ezetimibe and bempedoic acid were least efficacious.

Table 3.

Relative Efficacy of Nonstatin LLTs When Added to Maximally Tolerated Statins on the Percentage Change in LDL‐C at Week 24, on ApoB Levels at Week 12, and on Non–HDL‐C Levels at Week 12

Treatment MD 95% CI
MD in percentage change in LDL‐C in response to LLT relative to placebo at wk 24
Evolocumab, 140 mg Q2W/420 mg QM −61.84 −65.70 to −57.99
Alirocumab, 300 mg QM −58.70 −67.30 to −50.10
Alirocumab, 150 mg Q2W −57.11 −63.39 to −50.83
Inclisiran, 300 mg Q3M to Q6M −50.07 −53.82 to −46.31
Ezetimibe, 10 mg QD −25.03 −29.30 to −20.76
Bempedoic acid, 180 mg QD −16.61 −20.99 to −12.24
MD in percentage change in non–HDL‐C in response to LLT relative to placebo at wk 12
Evolocumab, 140 mg Q2W/420 mg QM −58.41 −61.19 to −55.63
Alirocumab, 150 mg Q2W −52.65 −57.97 to −47.32
Inclisiran, 300 mg Q3M to Q6M −45.07 −50.19 to −39.95
Alirocumab, 75 mg Q2W −44.80 −48.03 to −41.57
Alirocumab, 300 mg QM −44.33 −52.04 to −36.62
Bempedoic acid, 180 mg QD/ezetimibe, 10 mg QD FDC −34.45 −43.66 to −25.25
Ezetimibe, 10 mg QD −23.01 −26.38 to −19.64
Bempedoic acid, 180 mg QD −16.46 −20.89 to −12.04
MD in percentage change in ApoB in response to LLT relative to placebo at wk 12
Evolocumab, 140 mg Q2W/420 mg QM −51.16 −53.65 to −48.67
Alirocumab, 150 mg Q2W −50.08 −54.98 to −45.18
Alirocumab, 75 mg Q2W −41.46 −44.35 to −38.56
Inclisiran, 300 mg Q3M to Q6M −39.99 −44.51 to −35.47
Alirocumab, 300 mg QM −36.12 −43.75 to −28.48
Bempedoic acid, 180 mg QD/ezetimibe, 10 mg QD FDC −28.35 −36.82 to −19.88
Ezetimibe, 10 mg QD −18.86 −21.93 to −15.79
Bempedoic acid, 180 mg QD −14.53 −18.56 to −10.51

ApoB indicates apolipoprotein B; FDC, fixed‐dose combination; HDL‐C, high‐density lipoprotein cholesterol; LDL‐C, low‐density lipoprotein cholesterol; LLT, lipid‐lowering therapy; MD, mean difference; Q2W, every 2 weeks; Q3M, every 3 months; Q6M, every 6 months; QD, once a day; and QM, once a month.

Percentage Change in Non–HDL‐C From Baseline at Week 12 Compared With Placebo

Of the 48 trials in the primary NMA, 44 were included in an analysis to assess the percentage change in non–HDL‐C from baseline compared with placebo at week 12 (Table S4). The levels of non–HDL‐C were reduced from baseline compared with placebo by all nonstatin agents at week 12 (Table 3). Evolocumab was the most efficacious, with the greatest reduction in non–HDL‐C, followed by alirocumab, 150 mg Q2W, in a similar trend to the primary NMA. Inclisiran was more efficacious than alirocumab, 75 mg Q2W and 300 mg QM, at reducing levels of non–HDL‐C, in contrast to the findings seen in the primary analysis of LDL‐C reduction compared with placebo at week 12. This was followed by bempedoic acid/ezetimibe FDC, which was more efficacious than either treatment individually, as in the primary NMA.

Percentage Change in ApoB From Baseline at Week 12 Compared With Placebo

Of the 48 trials in the primary NMA, 44 were included in an analysis to assess the percentage change in ApoB from baseline compared with placebo at week 12 (Table S4). All nonstatin agents reduced ApoB levels from baseline compared with placebo (Table 3). A similar trend was seen to the primary NMA, with evolocumab as the most efficacious, followed by alirocumab, 150 mg Q2W, and alirocumab, 75 mg Q2W. However, inclisiran was more efficacious than alirocumab, 300 mg QM, at reducing levels of ApoB; these results differed from that seen in the primary analysis of LDL‐C reduction compared with placebo at week 12. Following alirocumab, 300 mg QM, were bempedoic acid/ezetimibe FDC, ezetimibe, and bempedoic acid as least efficacious.

Impact of Each Intervention on the LDL‐C Levels of a Simulated Population

Of the 10 000 simulated LDL‐C values ≥70 mg/dL (≥1.8 mmol/L) and thus requiring additional LLT, the proportion of values that achieved the 2019 ESC/European Atherosclerosis Society guideline LDL‐C goal of <55 mg/dL (<1.4 mmol/L) was highest for treatment with evolocumab, at 78.41%, followed by alirocumab, 150 mg Q2W, at 74.68% (Figure 7). Alirocumab, 75 mg Q2W, alirocumab, 300 mg QM, and inclisiran achieved proportions of 63.03%, 61.30%, and 60.25%, respectively. This was followed by bempedoic acid/ezetimibe FDC with 49.45%, and each monotherapy at 21.87% and 20.75%, respectively.

Figure 7. The proportion of simulated values that achieved a low‐density lipoprotein cholesterol (LDL‐C) level of <55 mg/dL (<1.4 mmol/L) following treatment with each intervention.

Figure 7

The simulation values represent a hypothetical population with atherosclerotic cardiovascular disease, and the <55‐mg/dL value is the 2019 European Society of Cardiology/European Atherosclerosis Society guideline‐recommended LDL‐C level goal for very high‐risk patients. FDC indicates fixed‐dose combination; Q2W, every 2 weeks; Q3M, every 3 months; Q6M, every 6 months; QD, once a day; and QM, once a month.

DISCUSSION

The results of our NMA indicate that all nonstatin agents significantly reduced LDL‐C compared with placebo, regardless of background therapy. Evolocumab, 140 mg Q2W/420 mg QM, and alirocumab, 150 mg Q2W, were the most efficacious agents, followed by alirocumab, 75 mg Q2W, alirocumab, 300 mg QM, inclisiran, 300 mg Q3M to Q6M, and bempedoic acid, 180 mg QD/ezetimibe, 10 mg QD FDC. Ezetimibe, 10 mg QD, and bempedoic acid, 180 mg QD, monotherapies were shown to be somewhat less efficacious. The results observed in the primary network at week 12 were generally consistent when a longer time point was analyzed at week 24. The percentage LDL‐C reduction achieved with the PCSK9 inhibitor treatments at certain approved dosing regimens compared with placebo indicated that mAb PCSK9 inhibitors were more efficacious at reducing LDL‐C than treatment with an siRNA PCSK9 inhibitor. In addition, there was no significant difference between the LDL‐C–lowering capacity of alirocumab, 75 mg Q2W, alirocumab, 300 mg QM, and inclisiran, nor was there a significant difference between inclisiran and bempedoic acid/ezetimibe FDC, as shown in the pairwise comparison. Overall, these data could help inform physicians' treatment choice for patients who require additional LLT to lower their LDL‐C levels, consistent with current guidelines.

The findings from the primary NMA were consistent regardless of statin background therapy for most interventions, although the analysis in patients who were apparently statin intolerant must be viewed in the context of greater uncertainty. In the case of bempedoic acid, it was shown that the addition of bempedoic acid to moderate‐ to high‐intensity statin therapy resulted in a lower percentage reduction in LDL‐C compared with use in statin‐intolerant patients. The reduced efficacy of bempedoic acid on top of statin therapy is to be expected, because ATP citrate lyase is 2 steps upstream of statin reductase in the cholesterol synthesis pathway 20 , 75 ; hence, the impact of ATP citrate lyase inhibition can be expected to be greater in the absence of statin reductase inhibition. Findings from the primary NMA were consistent in a subgroup analysis of trials with populations of >50% patients with ASCVD, the most common very high‐risk group for whom nonstatin agents are recommended as an adjunct to statins to achieve LDL‐C treatment goals. 3 The maximum reduction achieved in the primary NMA by an additional nonstatin agent was 64.68%, which is around 14% higher than the current European guidelines’ minimum LDL‐C reduction goal of 50%. 3 However, this ≥50% goal reduction is from a baseline of no LDL‐C–lowering treatment, not in addition to statins. 3 The benefit of add‐on LLT can be estimated by assuming a “pre‐PCSK9” LDL‐C level of 96.7 mg/dL (2.5 mmol/L), which can be translated into an additional 13.5‐mg/dL (0.35‐mmol/L) reduction when comparing the reduction goal of 50% with the maximum reduction achieved by a PCSK9 inhibitor of 64.68%. Therefore, assuming the results in the Cholesterol Treatment Trialists' Collaboration meta‐analysis, which indicate a 38.7‐mg/dL (1.0‐mmol/L) reduction in LDL‐C reduces MACEs by 21%, 1 this would imply a reduction in MACEs of ≈7.9% (1–0.790.35), a hypothetical percentage that implies the benefit of the addition of nonstatin LLT agents.

The highest proportion (>70%) of LDL‐C values achieved the 2019 ESC/European Atherosclerosis Society LDL‐C guideline goal of <55 mg/dL (<1.4 mmol/L) with either evolocumab, 140 mg Q2W/420 mg QM, or alirocumab, 150 mg Q2W, treatment, in a simulated population. These 2 treatments also reduced LDL‐C levels by the greatest amount. Studies that also looked at the impact of treatment on whether LDL‐C met guidelines looked at the sequential addition of treatment. One study ran a simulation on a population of patients following a myocardial infarction, where the following treatment pathway was used: maximized high‐intensity statins, followed by ezetimibe, then an additional mAb PCSK9 inhibitor. 76 In those who had yet to achieve an LDL‐C of <55 mg/dL (<1.4 mmol/L) and who already received high‐intensity statins (78.3%), the addition of ezetimibe resulted in a further 27.5% achieving the goal. 76 The addition of evolocumab, 140 mg Q2W/420 mg QM, or alirocumab, 75 mg Q2W, to the previous statin and ezetimibe therapy resulted in a further 42.7% and 39.2% of patients achieving an LDL‐C of <55 mg/dL (<1.4 mmol/L), respectively. 76 Another study looked at the achievement of an LDL‐C of <70 mg/dL (<1.8 mmol/L) using a simulated population with ASCVD, and the following treatments steps sequentially applied with LDL‐C measured after each to identify whether LDL‐C of <70 mg/dL (<1.8 mmol/L) had been achieved: statin, statin up titration, add‐on ezetimibe, add‐on alirocumab, 75 mg, and up titration to alirocumab, 150 mg (base‐case scenario). 77 The base‐case scenario identified that 16.7% people required an ezetimibe add‐on therapy, 14% people required an additional add‐on alirocumab, 75 mg, therapy or further 150 mg up titration to achieve an LDL‐C of <70 mg/dL (<1.8 mmol/L). 77 A related study found that when 10% full and 10% partial statin intolerance were assumed in the simulated population, ezetimibe use was 38.5% and PCSK9 inhibitor use was 21.1% to achieve LDL‐C of <70 mg/dL (<1.8 mmol/L). 78 These studies are a sample of those that show the benefits of add‐on LLT therapies to achieve an LDL‐C consistent with guideline recommendations.

For the time points used in the study, week 12 data were the most reliable to assess relative efficacy. At this time point, most study participants remained on therapy. Fewer trials reported week 24 data. Many alirocumab trials were excluded from analysis at week 24 because the protocol allowed up titration of dose at week 12; hence, week 24 data describe a combination of doses. In clinical practice, the effectiveness of LLT is not only a function of efficacy but also adherence and persistence of patients to treatment in the longer term. The data included in the NMA are drawn from randomized controlled trials in which patients generally adhered to and persisted with treatment. The modes and frequencies of administration differ between LLTs, including oral tablets, Q2W or QM subcutaneous injections (mAb PCSK9 inhibitors), and Q3M to Q6M subcutaneous injection (siRNA PCSK9 inhibitor). 20 , 25 , 29 , 33 , 79 , 80

The NMA reported in this article provides updated information to our previous analysis published in 2017, 13 not only in terms of including new data for established therapies, but also with the introduction of newer agents. In addition, the scope of the analysis was broadened by allowing the inclusion of any study in which nonstatin agents were combined with maximum‐tolerated statin background (or in reported statin‐intolerant patients). This allowed inclusion of additional trials, such as IMPROVE‐IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial), in which patients were not stabilized on statin dose before randomization. 73 Although there are other NMAs published on this subject, our analysis is differentiated by the granular comparison of PCSK9 inhibitors and by the inclusion of bempedoic acid and inclisiran. Several NMAs have been conducted using clinical studies of mAb PCSK9 inhibitors; however, authors have pooled results together into a single class 81 , 82 , 83 , 84 , 85 , 86 , 87 or not made formal indirect comparisons. 88 , 89

A recent NMA performed by Burnett et al concluded that inclisiran, alirocumab, and evolocumab are expected to provide similar clinically meaningful improvements in LDL‐C in patients with hypercholesterolemia on maximally tolerated statins who were at increased cardiovascular risk. 90 There are, however, important methodologic differences between the Burnett et al study and this study. First, our analysis encompassed a broader population, including a larger number of clinical trials (48 versus 16) in the primary analysis, which reduced overall uncertainty. Second, although the percentage LDL‐C reduction reported for evolocumab (≈65%) was consistent in both analyses, Burnett et al reported a larger reduction in LDL‐C for inclisiran (57.49%) and a more modest reduction in LDL‐C for alirocumab (58.25%) than the analysis presented herein. More important, the reduction in LDL‐C for inclisiran was evaluated at day 150, whereas our analysis considered the more appropriate time‐adjusted LDL‐C reduction between days 90 and 540. Notably, the reduction in LDL‐C at day 150 was not a primary or secondary outcome in ORION‐10 (Inclisiran for Participants With Atherosclerotic Cardiovascular Disease and Elevated Low‐density Lipoprotein Cholesterol) or ORION‐11 (Inclisiran for Subjects With ASCVD or ASCVD‐Risk Equivalents and Elevated Low‐density Lipoprotein Cholesterol) 22 ; therefore, Burnett et al also presented a sensitivity analysis considering the time‐adjusted LDL‐C reduction. The results indicate a reduction in LDL‐C for inclisiran of 51.42%, which is closely aligned with the 50.17% reduction reported in our analysis. Finally, alirocumab LDL‐C reduction is estimated after dose titration is allowed such that the efficacy estimate is “averaged” over the doses, whereas our analysis considers the 75‐ and 150‐mg Q2W doses separately.

Our analysis of the effect of additional nonstatin agents on the change from baseline in other lipid end points (ApoB and non–HDL‐C), compared with placebo at week 12, found broadly similar findings to that of the primary analysis, except for inclisiran. Inclisiran was more efficacious than alirocumab, 300 mg QM, at reducing ApoB levels and more efficacious than alirocumab, 75 mg Q2W and 300 mg QM, at reducing non–HDL‐C levels, compared with placebo at week 12.

At present, mAb PCSK9 inhibitors (evolocumab and alirocumab) and ezetimibe have been shown to reduce risk of MACEs in patients with ASCVD or acute coronary syndrome 30 , 35 , 60 , 73 , 91 ; however, bempedoic acid (NCT02993406 and NCT04579367) and inclisiran (NCT03705234) are currently under investigation for their efficacy in reducing cardiovascular events. 92 , 93 , 94 Our NMA did not include a comparison of cardiovascular outcomes, even where data are available for individual interventions, because of differing trial designs and populations. Focusing on LDL‐C reduction is therefore currently the best method of comparing the efficacy of LLTs because it is largely unaffected by treatment effect modifiers. In contrast, cardiovascular outcomes are impacted by the duration of follow‐up, and the type and recency of qualifying events, as well as the cardiovascular risk of the trial populations. Definition of outcome measures can also complicate comparison between agents.

There are limitations associated with this review and NMA. First, the NMA is limited by the quantity and quality of data available from included trials. Our NMA concentrates on efficacy estimates and does not consider adherence and economic value implications. There are relatively few head‐to‐head trials of nonstatin agents or regimens; therefore, most comparisons within the network are largely indirect. However, given that LDL‐C reductions have been shown to be similar across multiple characteristics, we believe that this limitation does not undermine the analysis. The NMA focuses on week 12 follow‐up, which provides the richest data source but could be considered as relatively short‐term. However, for most agents, there is evidence that week 12 data are generalizable to the long‐term. 18 , 45 , 59

CONCLUSIONS

All nonstatin agents significantly reduced LDL‐C levels compared with placebo. However, evolocumab, 140 mg Q2W/420 mg QM, and alirocumab, 150 mg Q2W (mAb PCSK9 inhibitors also shown to reduce MACEs in patients with ASCVD and acute coronary syndrome), were consistently the most efficacious nonstatin agents/regimens, potentially allowing more patients to achieve an LDL‐C consistent with the current guidelines.

Sources of Funding

This systematic review and network meta‐analysis was sponsored by Amgen Inc, as were all trials of evolocumab included in the analysis.

Disclosures

Drs Bray and Villa are Amgen employees and hold Amgen stock. Dr Worth was an employee of Amgen at time of analysis and drafting. Dr Palagashvili was an employee of Amgen at the initiation of analysis and drafting, and holds Amgen stock. Dr Toth reports speakers bureau for Amarin, Amgen, Esperion, and Novo‐Nordisk; and consultant for Amarin, Amgen, bio89, Novartis, and Theravance. Dr Sattar reports consulting fees from Affimune, Amgen, AstraZeneca, Boehringer Ingelheim, Eli Lilly, Hanmi Pharmaceuticals, MSD, Novo Nordisk, Novartis, Pfizer, and Sanofi; and grants from AstraZeneca, Boehringer Ingelheim, Novartis, and Roche Diagnostics. Dr Stroes reports ad‐board/lecturing fees have been paid to his institution by Sanofi‐Regeneron, Esperion, Amgen, Novartis, Novo‐Nordisk, and Athera.

Supporting information

Data S1

Tables S1–S5

Figures S1–S2

Acknowledgments

Support for the systematic literature review was provided by Royal College of Physicians London, UK, and Ingress Health GmbH, Germany. Medical writing support (funded by Amgen Inc) was provided by Daniel Smalley, PhD, and Hayley Owen, PhD (Bioscript Medical Ltd, Macclesfield, UK).

Author contributions: Drs Worth, Bray, and Villa contributed to study design, interpretation of data, and drafting of the article. Drs Toth, Stroes, Sattar, and Palagashvili contributed to interpretation of data and drafting of article. All authors approved the final article for submission.

For Sources of Funding and Disclosures, see page 17.

REFERENCES

  • 1. Cholesterol Treatment Trialists' (CTT) Collaboration , Baigent C, Blackwell L, Emberson J, Holland LE, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, 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. doi: 10.1016/S0140-6736(10)61350-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella‐Tommasino J, Forman DE, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines. Circulation. 2019;139:e1082–e1143. doi: 10.1161/CIR.0000000000000625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, et al. 2019 ESC/EAS guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J. 2020;41:111–188. doi: 10.1093/eurheartj/ehz455 [DOI] [PubMed] [Google Scholar]
  • 4. Giugliano RP, Pedersen TR, Park JG, De Ferrari GM, Gaciong ZA, Ceska R, Toth K, Gouni‐Berthold I, Lopez‐Miranda J, Schiele F, 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. doi: 10.1016/S0140-6736(17)32290-0 [DOI] [PubMed] [Google Scholar]
  • 5. Koren MJ, Sabatine MS, Giugliano RP, Langslet G, Wiviott SD, Ruzza A, Ma Y, Hamer AW, Wasserman SM, Raal FJ. Long‐term efficacy and safety of evolocumab in patients with hypercholesterolemia. J Am Coll Cardiol. 2019;74:2132–2146. doi: 10.1016/j.jacc.2019.08.1024 [DOI] [PubMed] [Google Scholar]
  • 6. Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, Hoes AW, Jennings CS, Landmesser U, Pedersen TR, et al. 2016 ESC/EAS guidelines for the management of dyslipidaemias. Eur Heart J. 2016;37:2999–3058. doi: 10.1093/eurheartj/ehw272 [DOI] [PubMed] [Google Scholar]
  • 7. Visseren FLJ, Mach F, Smulders YM, Carballo D, Koskinas KC, Back M, Benetos A, Biffi A, Boavida JM, Capodanno D, et al. 2021 ESC guidelines on cardiovascular disease prevention in clinical practice. Eur Heart J. 2021;42:3227–3337. doi: 10.1093/eurheartj/ehab484 [DOI] [PubMed] [Google Scholar]
  • 8. 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. doi: 10.1177/1060028019888487 [DOI] [PubMed] [Google Scholar]
  • 9. Serban MC, Colantonio LD, Manthripragada AD, Monda KL, Bittner VA, Banach M, Chen L, Huang L, Dent R, Kent ST, et al. Statin intolerance and risk of coronary heart events and all‐cause mortality following myocardial infarction. J Am Coll Cardiol. 2017;69:1386–1395. doi: 10.1016/j.jacc.2016.12.036 [DOI] [PubMed] [Google Scholar]
  • 10. Gupta A, Thompson D, Whitehouse A, Collier T, Dahlof B, Poulter N, Collins R, Sever P; ASCOT Investigators . Adverse events associated with unblinded, but not with blinded, statin therapy in the Anglo‐Scandinavian Cardiac Outcomes Trial‐Lipid‐Lowering Arm (ASCOT‐LLA): a randomised double‐blind placebo‐controlled trial and its non‐randomised non‐blind extension phase. Lancet. 2017;389:2473–2481. doi: 10.1016/S0140-6736(17)31075-9 [DOI] [PubMed] [Google Scholar]
  • 11. Ray KK, Molemans B, Schoonen WM, Giovas P, Bray S, Kiru G, Murphy J, Banach M, De Servi S, Gaita D. EU‐wide cross‐sectional observational study of lipid‐modifying therapy use in secondary and primary care: the DA VINCI study. Eur J Prev Cardiol. 2021;28:1279–1289. doi: 10.1093/eurjpc/zwaa047 [DOI] [PubMed] [Google Scholar]
  • 12. Cannon CP, de Lemos JA, Rosenson RS, Ballantyne CM, Liu Y, Gao Q, Palagashvilli T, Alam S, Mues KE, Bhatt DL, et al. Use of lipid‐lowering therapies over 2 years in GOULD, a registry of patients with atherosclerotic cardiovascular disease in the US. JAMA Cardiol. 2021;6:1060–1068. doi: 10.1001/jamacardio.2021.1810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Toth PP, Worthy G, Gandra SR, Sattar N, Bray S, Cheng LI, Bridges I, Worth GM, Dent R, Forbes CA, et al. Systematic review and network meta‐analysis on the efficacy of evolocumab and other therapies for the management of lipid levels in hyperlipidemia. J Am Heart Assoc. 2017;6:e005367. doi: 10.1161/JAHA.116.005367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Ballantyne CM, Banach M, Mancini GBJ, Lepor NE, Hanselman JC, Zhao X, Leiter LA. Efficacy and safety of bempedoic acid added to ezetimibe in statin‐intolerant patients with hypercholesterolemia: a randomized, placebo‐controlled study. Atherosclerosis. 2018;277:195–203. doi: 10.1016/j.atherosclerosis.2018.06.002 [DOI] [PubMed] [Google Scholar]
  • 15. Ballantyne CM, Davidson MH, Macdougall DE, Bays HE, Dicarlo LA, Rosenberg NL, Margulies J, Newton RS. Efficacy and safety of a novel dual modulator of adenosine triphosphate‐citrate lyase and adenosine monophosphate‐activated protein kinase in patients with hypercholesterolemia: results of a multicenter, randomized, double‐blind, placebo‐controlled, parallel‐group trial. J Am Coll Cardiol. 2013;62:1154–1162. doi: 10.1016/j.jacc.2013.05.050 [DOI] [PubMed] [Google Scholar]
  • 16. Goldberg AC, Leiter LA, Stroes ESG, Baum SJ, Hanselman JC, Bloedon LT, Lalwani ND, Patel PM, Zhao X, Duell PB. Effect of bempedoic acid vs placebo added to maximally tolerated statins on low‐density lipoprotein cholesterol in patients at high risk for cardiovascular disease: the CLEAR wisdom randomized clinical trial. JAMA. 2019;322:1780–1788. doi: 10.1001/jama.2019.16585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Laufs U, Banach M, Mancini GBJ, Gaudet D, Bloedon LT, Sterling LR, Kelly S, Stroes ESG. Efficacy and safety of bempedoic acid in patients with hypercholesterolemia and statin intolerance. J Am Heart Assoc. 2019;8:e011662. doi: 10.1161/JAHA.118.011662 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Ray KK, Bays HE, Catapano AL, Lalwani ND, Bloedon LT, Sterling LR, Robinson PL, Ballantyne CM; CLEAR Harmony Trial . Safety and efficacy of bempedoic acid to reduce LDL cholesterol. N Engl J Med. 2019;380:1022–1032. doi: 10.1056/NEJMoa1803917 [DOI] [PubMed] [Google Scholar]
  • 19. Esperion Therapeutics, Inc . Nexletol—Prescribing Information. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/211616s000lbl.pdf. Accessed April 8, 2022.
  • 20. European Medicines Agency . Nilemdo—Summary of Product Characteristics. Available at: https://www.ema.europa.eu/en/documents/product‐information/nilemdo‐epar‐product‐information_en.pdf. Accessed April 8, 2022.
  • 21. Raal FJ, Kallend D, Ray KK, Turner T, Koenig W, Wright RS, Wijngaard PLJ, Curcio D, Jaros MJ, Leiter LA, et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N Engl J Med. 2020;382:1520–1530. doi: 10.1056/NEJMoa1913805 [DOI] [PubMed] [Google Scholar]
  • 22. Ray KK, Wright RS, Kallend D, Koenig W, Leiter LA, Raal FJ, Bisch JA, Richardson T, Jaros M, Wijngaard PLJ, et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med. 2020;382:1507–1519. doi: 10.1056/NEJMoa1912387 [DOI] [PubMed] [Google Scholar]
  • 23. Leiter LA, Teoh H, Kallend D, Wright RS, Landmesser U, Wijngaard PLJ, Kastelein JJP, Ray KK. Inclisiran lowers LDL‐C and PCSK9 irrespective of diabetes status: the ORION‐1 randomized clinical trial. Diabetes Care. 2019;42:173–176. doi: 10.2337/dc18-1491 [DOI] [PubMed] [Google Scholar]
  • 24. European Medicines Agency . Leqvio. 2021. Available at: https://www.ema.europa.eu/en/medicines/human/EPAR/leqvio. Accessed April 8, 2022.
  • 25. European Medicines Agency . Leqvio—Summary of Product Characteristics. Available at: https://www.ema.europa.eu/en/documents/product‐information/leqvio‐epar‐product‐information_en.pdf. Accessed April 8, 2022.
  • 26. Novartis Pharmaceuticals Corporation . Leqvio—Prescribing Information. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/214012lbl.pdf. Accessed April 8, 2022.
  • 27. Centre for Reviews and Dissemination . Systematic Reviews: CRD's Guidance for Undertaking Reviews in Health Care. University of York; 2009. Available at: http://www.york.ac.uk/inst/crd/SysRev/!SSL!/WebHelp/SysRev3.htm. Accessed April 8, 2022. [Google Scholar]
  • 28. Canadian Agency for Drugs and Technologies in Health . CADTH peer review checklist for search strategies. CADTH; 2013. Available at: https://www.cadth.ca/sites/default/files/is/Peer_review/CADTH%20Peer%20Review%20Checklist%20for%20Search%20Strategies_e.pdf. Accessed April 8, 2022. [Google Scholar]
  • 29. European Medicines Agency . Repatha—Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product‐information/repatha‐epar‐product‐information_en.pdf. Accessed April 8, 2022.
  • 30. Amgen Inc . Repatha (evolocumab)—Prescribing Information. Available at: https://www.pi.amgen.com/~/media/amgen/repositorysites/pi‐amgen‐com/repatha/repatha_pi_hcp_english.pdf. Accessed April 8, 2022.
  • 31. Wasserman SM, Sabatine MS, Koren MJ, Giugliano RP, Legg JC, Emery MG, Doshi S, Liu T, Somaratne R, Gibbs JP. Comparison of LDL‐C reduction using different evolocumab doses and intervals: biological insights and treatment implications. J Cardiovasc Pharmacol Ther. 2018;23:423–432. doi: 10.1177/1074248418774043 [DOI] [PubMed] [Google Scholar]
  • 32. Kereiakes DJ, Robinson JG, Cannon CP, Lorenzato C, Pordy R, Chaudhari U, Colhoun HM. Efficacy and safety of the proprotein convertase subtilisin/kexin type 9 inhibitor alirocumab among high cardiovascular risk patients on maximally tolerated statin therapy: the ODYSSEY COMBO I study. Am Heart J. 2015;169:906–915.e913. doi: 10.1016/j.ahj.2015.03.004 [DOI] [PubMed] [Google Scholar]
  • 33. European Medicines Agency . Praluent—Summary of Product Characteristics. Available at: https://www.ema.europa.eu/en/documents/product‐information/praluent‐epar‐product‐information_en.pdf. Accessed April 8, 2022.
  • 34. Roth EM, Moriarty PM, Bergeron J, Langslet G, Manvelian G, Zhao J, Baccara‐Dinet MT, Rader DJ; ODYSSEY CHOICE I Investigators . A phase III randomized trial evaluating alirocumab 300 mg every 4 weeks as monotherapy or add‐on to statin: ODYSSEY CHOICE I. Atherosclerosis. 2016;254:254–262. doi: 10.1016/j.atherosclerosis.2016.08.043 [DOI] [PubMed] [Google Scholar]
  • 35. Sanofi‐Aventis U.S . LLC. Praluent—Prescribing Information. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125559s024lbl.pdf. Accessed April 8, 2022.
  • 36. Rücker G, Krahn U, König J, Efthimiou O, Schwarzer G. Netmeta: network meta‐analysis using frequentist methods. R package version 1.2‐1. 2020. Available at: https://CRAN.R‐project.org/package=netmeta. Accessed April 8, 2022.
  • 37. Lu G, Ades AE. Combination of direct and indirect evidence in mixed treatment comparisons. Stat Med. 2004;23:3105–3124. doi: 10.1002/sim.1875 [DOI] [PubMed] [Google Scholar]
  • 38. McKenney JM, Koren MJ, Kereiakes DJ, Hanotin C, Ferrand AC, Stein EA. Safety and efficacy of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 serine protease, SAR236553/REGN727, in patients with primary hypercholesterolemia receiving ongoing stable atorvastatin therapy. J Am Coll Cardiol. 2012;59:2344–2353. doi: 10.1016/j.jacc.2012.03.007 [DOI] [PubMed] [Google Scholar]
  • 39. Moriarty PM, Thompson PD, Cannon CP, Guyton JR, Bergeron J, Zieve FJ, Bruckert E, Jacobson TA, Kopecky SL, Baccara‐Dinet MT, et al. Efficacy and safety of alirocumab vs ezetimibe in statin‐intolerant patients, with a statin rechallenge arm: the ODYSSEY ALTERNATIVE randomized trial. J Clin Lipidol. 2015;9:758–769. doi: 10.1016/j.jacl.2015.08.006 [DOI] [PubMed] [Google Scholar]
  • 40. Stroes E, Guyton JR, Lepor N, Civeira F, Gaudet D, Watts GF, Baccara‐Dinet MT, Lecorps G, Manvelian G, Farnier M, et al. Efficacy and safety of alirocumab 150 mg every 4 weeks in patients with hypercholesterolemia not on statin therapy: the ODYSSEY CHOICE II study. J Am Heart Assoc. 2016;5:e003421. doi: 10.1161/JAHA.116.003421 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Cannon CP, Cariou B, Blom D, McKenney JM, Lorenzato C, Pordy R, Chaudhari U, Colhoun HM; ODYSSEY COMBO II Investigators . Efficacy and safety of alirocumab in high cardiovascular risk patients with inadequately controlled hypercholesterolaemia on maximally tolerated doses of statins: the ODYSSEY COMBO II randomized controlled trial. Eur Heart J. 2015;36:1186–1194. doi: 10.1093/eurheartj/ehv028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Leiter LA, Cariou B, Muller‐Wieland D, Colhoun HM, Del Prato S, Tinahones FJ, Ray KK, Bujas‐Bobanovic M, Domenger C, Mandel J, et al. Efficacy and safety of alirocumab in insulin‐treated individuals with type 1 or type 2 diabetes and high cardiovascular risk: the ODYSSEY DM‐INSULIN randomized trial. Diabetes Obes Metab. 2017;19:1781–1792. doi: 10.1111/dom.13114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Han Y, Chen J, Chopra VK, Zhang S, Su G, Ma C, Huang Z, Ma Y, Yao Z, Yuan Z, et al. ODYSSEY EAST: alirocumab efficacy and safety vs ezetimibe in high cardiovascular risk patients with hypercholesterolemia and on maximally tolerated statin in China, India, and Thailand. J Clin Lipidol. 2020;14:98–108.e108. doi: 10.1016/j.jacl.2019.10.015 [DOI] [PubMed] [Google Scholar]
  • 44. Kastelein JJ, Ginsberg HN, Langslet G, Hovingh GK, Ceska R, Dufour R, Blom D, Civeira F, Krempf M, Lorenzato C, et al. ODYSSEY FH I and FH II: 78 week results with alirocumab treatment in 735 patients with heterozygous familial hypercholesterolaemia. Eur Heart J. 2015;36:2996–3003. doi: 10.1093/eurheartj/ehv370 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Ginsberg HN, Rader DJ, Raal FJ, Guyton JR, Baccara‐Dinet MT, Lorenzato C, Pordy R, Stroes E. Efficacy and safety of alirocumab in patients with heterozygous familial hypercholesterolemia and LDL‐C of 160 mg/dl or higher. Cardiovasc Drugs Ther. 2016;30:473–483. doi: 10.1007/s10557-016-6685-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Teramoto T, Kobayashi M, Tasaki H, Yagyu H, Higashikata T, Takagi Y, Uno K, Baccara‐Dinet MT, Nohara A. Efficacy and safety of alirocumab in Japanese patients with heterozygous familial hypercholesterolemia or at high cardiovascular risk with hypercholesterolemia not adequately controlled with statins—ODYSSEY Japan randomized controlled trial. Circ J. 2016;80:1980–1987. doi: 10.1253/circj.CJ-16-0387 [DOI] [PubMed] [Google Scholar]
  • 47. Koh KK, Nam CW, Chao TH, Liu ME, Wu CJ, Kim DS, Kim CJ, Li I, Li J, Baccara‐Dinet MT, et al. A randomized trial evaluating the efficacy and safety of alirocumab in South Korea and Taiwan (ODYSSEY KT). J Clin Lipidol. 2018;12:162–172.e166. doi: 10.1016/j.jacl.2017.09.007 [DOI] [PubMed] [Google Scholar]
  • 48. Robinson JG, Farnier M, Krempf M, Bergeron J, Luc G, Averna M, Stroes ES, Langslet G, Raal FJ, El Shahawy M, et al. Efficacy and safety of alirocumab in reducing lipids and cardiovascular events. N Engl J Med. 2015;372:1489–1499. doi: 10.1056/NEJMoa1501031 [DOI] [PubMed] [Google Scholar]
  • 49. Teramoto T, Kiyosue A, Ishigaki Y, Harada‐Shiba M, Kawabata Y, Ozaki A, Baccara‐Dinet MT, Sata M. Efficacy and safety of alirocumab 150mg every 4 weeks in hypercholesterolemic patients on non‐statin lipid‐lowering therapy or lowest strength dose of statin: ODYSSEY NIPPON. J Cardiol. 2019;73:218–227. doi: 10.1016/j.jjcc.2018.10.004 [DOI] [PubMed] [Google Scholar]
  • 50. Bays H, Gaudet D, Weiss R, Ruiz JL, Watts GF, Gouni‐Berthold I, Robinson J, Zhao J, Hanotin C, Donahue S. Alirocumab as add‐on to atorvastatin versus other lipid treatment strategies: ODYSSEY OPTIONS I randomized trial. J Clin Endocrinol Metab. 2015;100:3140–3148. doi: 10.1210/jc.2015-1520 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Farnier M, Jones P, Severance R, Averna M, Steinhagen‐Thiessen E, Colhoun HM, Du Y, Hanotin C, Donahue S. Efficacy and safety of adding alirocumab to rosuvastatin versus adding ezetimibe or doubling the rosuvastatin dose in high cardiovascular‐risk patients: the ODYSSEY OPTIONS II randomized trial. Atherosclerosis. 2016;244:138–146. doi: 10.1016/j.atherosclerosis.2015.11.010 [DOI] [PubMed] [Google Scholar]
  • 52. Stein EA, Dufour R, Gagne C, Gaudet D, East C, Donovan JM, Chin W, Tribble DL, McGowan M. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: results of a randomized, double‐blind, placebo‐controlled trial to assess efficacy and safety as add‐on therapy in patients with coronary artery disease. Circulation. 2012;126:2283–2292. doi: 10.1161/CIRCULATIONAHA.112.104125 [DOI] [PubMed] [Google Scholar]
  • 53. Teramoto T, Kobayashi M, Uno K, Takagi Y, Matsuoka O, Sugimoto M, Inoue S, Minami F, Baccara‐Dinet MT. Efficacy and safety of alirocumab in Japanese subjects (phase 1 and 2 studies). Am J Cardiol. 2016;118:56–63. doi: 10.1016/j.amjcard.2016.04.011 [DOI] [PubMed] [Google Scholar]
  • 54. Ballantyne CM, McKenney JM, MacDougall DE, Margulies JR, Robinson PL, Hanselman JC, Lalwani ND. Effect of ETC‐1002 on serum low‐density lipoprotein cholesterol in hypercholesterolemic patients receiving statin therapy. Am J Cardiol. 2016;117:1928–1933. doi: 10.1016/j.amjcard.2016.03.043 [DOI] [PubMed] [Google Scholar]
  • 55. Ballantyne CM, Laufs U, Ray KK, Leiter LA, Bays HE, Goldberg AC, Stroes ES, MacDougall D, Zhao X, Catapano AL. Bempedoic acid plus ezetimibe fixed‐dose combination in patients with hypercholesterolemia and high CVD risk treated with maximally tolerated statin therapy. Eur J Prev Cardiol. 2020;27:593–603. doi: 10.1177/2047487319864671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Thompson PD, MacDougall DE, Newton RS, Margulies JR, Hanselman JC, Orloff DG, McKenney JM, Ballantyne CM. Treatment with ETC‐1002 alone and in combination with ezetimibe lowers LDL cholesterol in hypercholesterolemic patients with or without statin intolerance. J Clin Lipidol. 2016;10:556–567. doi: 10.1016/j.jacl.2015.12.025 [DOI] [PubMed] [Google Scholar]
  • 57. Rosenson RS, Daviglus ML, Handelsman Y, Pozzilli P, Bays H, Monsalvo ML, Elliott‐Davey M, Somaratne R, Reaven P. Efficacy and safety of evolocumab in individuals with type 2 diabetes mellitus: primary results of the randomised controlled BANTING study. Diabetologia. 2019;62:948–958. doi: 10.1007/s00125-019-4856-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Lorenzatti AJ, Eliaschewitz FG, Chen Y, Lu J, Baass A, Monsalvo ML, Wang N, Hamer AW, Ge J. Randomised study of evolocumab in patients with type 2 diabetes and dyslipidaemia on background statin: primary results of the BERSON clinical trial. Diabetes Obes Metab. 2019;21:1455–1463. doi: 10.1111/dom.13680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Blom DJ, Hala T, Bolognese M, Lillestol MJ, Toth PD, Burgess L, Ceska R, Roth E, Koren MJ, Ballantyne CM, et al. A 52‐week placebo‐controlled trial of evolocumab in hyperlipidemia. N Engl J Med. 2014;370:1809–1819. doi: 10.1056/NEJMoa1316222 [DOI] [PubMed] [Google Scholar]
  • 60. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376:1713–1722. doi: 10.1056/NEJMoa1615664 [DOI] [PubMed] [Google Scholar]
  • 61. Sullivan D, Olsson AG, Scott R, Kim JB, Xue A, Gebski V, Wasserman SM, Stein EA. Effect of a monoclonal antibody to PCSK9 on low‐density lipoprotein cholesterol levels in statin‐intolerant patients: the GAUSS randomized trial. JAMA. 2012;308:2497–2506. doi: 10.1001/jama.2012.25790 [DOI] [PubMed] [Google Scholar]
  • 62. Stroes E, Colquhoun D, Sullivan D, Civeira F, Rosenson RS, Watts GF, Bruckert E, Cho L, Dent R, Knusel B, et al. Anti‐PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: the GAUSS‐2 randomized, placebo‐controlled phase 3 clinical trial of evolocumab. J Am Coll Cardiol. 2014;63:2541–2548. doi: 10.1016/j.jacc.2014.03.019 [DOI] [PubMed] [Google Scholar]
  • 63. Nissen SE, Stroes E, Dent‐Acosta RE, Rosenson RS, Lehman SJ, Sattar N, Preiss D, Bruckert E, Ceska R, Lepor N, et al. Efficacy and tolerability of evolocumab vs ezetimibe in patients with muscle‐related statin intolerance: the GAUSS‐3 randomized clinical trial. JAMA. 2016;315:1580–1590. doi: 10.1001/jama.2016.3608 [DOI] [PubMed] [Google Scholar]
  • 64. Koba S, Inoue I, Cyrille M, Lu C, Inomata H, Shimauchi J, Kajinami K. Evolocumab vs. ezetimibe in statin‐intolerant hyperlipidemic Japanese patients: phase 3 GAUSS‐4 trial. J Atheroscler Thromb. 2020;27:471–484. doi: 10.5551/jat.50963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Nicholls SJ, Puri R, Anderson T, Ballantyne CM, Cho L, Kastelein JJ, Koenig W, Somaratne R, Kassahun H, Yang J, et al. Effect of evolocumab on progression of coronary disease in statin‐treated patients: the GLAGOV randomized clinical trial. JAMA. 2016;316:2373–2384. doi: 10.1001/jama.2016.16951 [DOI] [PubMed] [Google Scholar]
  • 66. Robinson JG, Nedergaard BS, Rogers WJ, Fialkow J, Neutel JM, Ramstad D, Somaratne R, Legg JC, Nelson P, Scott R, et al. Effect of evolocumab or ezetimibe added to moderate‐ or high‐intensity statin therapy on LDL‐C lowering in patients with hypercholesterolemia: the LAPLACE‐2 randomized clinical trial. JAMA. 2014;311:1870–1882. doi: 10.1001/jama.2014.4030 [DOI] [PubMed] [Google Scholar]
  • 67. Giugliano RP, Desai NR, Kohli P, Rogers WJ, Somaratne R, Huang F, Liu T, Mohanavelu S, Hoffman EB, McDonald ST, et al. Efficacy, safety, and tolerability of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 in combination with a statin in patients with hypercholesterolaemia (LAPLACE‐TIMI 57): a randomised, placebo‐controlled, dose‐ranging, phase 2 study. Lancet. 2012;380:2007–2017. doi: 10.1016/S0140-6736(12)61770-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Raal F, Scott R, Somaratne R, Bridges I, Li G, Wasserman SM, Stein EA. Low‐density lipoprotein cholesterol‐lowering effects of AMG 145, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 serine protease in patients with heterozygous familial hypercholesterolemia: the Reduction of LDL‐C with PCSK9 Inhibition in Heterozygous Familial Hypercholesterolemia Disorder (RUTHERFORD) randomized trial. Circulation. 2012;126:2408–2417. doi: 10.1161/CIRCULATIONAHA.112.144055 [DOI] [PubMed] [Google Scholar]
  • 69. Raal FJ, Stein EA, Dufour R, Turner T, Civeira F, Burgess L, Langslet G, Scott R, Olsson AG, Sullivan D, et al. PCSK9 inhibition with evolocumab (AMG 145) in heterozygous familial hypercholesterolaemia (RUTHERFORD‐2): a randomised, double‐blind, placebo‐controlled trial. Lancet. 2015;385:331–340. doi: 10.1016/S0140-6736(14)61399-4 [DOI] [PubMed] [Google Scholar]
  • 70. Hirayama A, Honarpour N, Yoshida M, Yamashita S, Huang F, Wasserman SM, Teramoto T. Effects of evolocumab (AMG 145), a monoclonal antibody to PCSK9, in hypercholesterolemic, statin‐treated Japanese patients at high cardiovascular risk‐‐primary results from the phase 2 YUKAWA study. Circ J. 2014;78:1073–1082. doi: 10.1253/circj.CJ-14-0130 [DOI] [PubMed] [Google Scholar]
  • 71. Kiyosue A, Honarpour N, Kurtz C, Xue A, Wasserman SM, Hirayama A. A phase 3 study of evolocumab (AMG 145) in statin‐treated Japanese patients at high cardiovascular risk. Am J Cardiol. 2016;117:40–47. doi: 10.1016/j.amjcard.2015.10.021 [DOI] [PubMed] [Google Scholar]
  • 72. Kastelein JJ, Akdim F, Stroes ES, Zwinderman AH, Bots ML, Stalenhoef AF, Visseren FL, Sijbrands EJ, Trip MD, Stein EA, et al. Simvastatin with or without ezetimibe in familial hypercholesterolemia. N Engl J Med. 2008;358:1431–1443. doi: 10.1056/NEJMoa0800742 [DOI] [PubMed] [Google Scholar]
  • 73. Cannon CP, Blazing MA, Giugliano RP, McCagg A, White JA, Theroux P, Darius H, Lewis BS, Ophuis TO, Jukema JW, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372:2387–2397. doi: 10.1056/NEJMoa1410489 [DOI] [PubMed] [Google Scholar]
  • 74. Masana L, Mata P, Gagne C, Sirah W, Cho M, Johnson‐Levonas AO, Meehan A, Troxell JK, Gumbiner B; Ezetimibe Study Group . Long‐term safety and, tolerability profiles and lipid‐modifying efficacy of ezetimibe coadministered with ongoing simvastatin treatment: a multicenter, randomized, double‐blind, placebo‐controlled, 48‐week extension study. Clin Ther. 2005;27:174–184. doi: 10.1016/j.clinthera.2005.02.011 [DOI] [PubMed] [Google Scholar]
  • 75. Marrs JC, Anderson SL. Bempedoic acid for the treatment of dyslipidemia. Drugs Context. 2020;9:2020‐6‐5. doi: 10.7573/dic.2020-6-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Allahyari A, Jernberg T, Hagstrom E, Leosdottir M, Lundman P, Ueda P. Application of the 2019 ESC/EAS dyslipidaemia guidelines to nationwide data of patients with a recent myocardial infarction: a simulation study. Eur Heart J. 2020;41:3900–3909. doi: 10.1093/eurheartj/ehaa034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Cannon CP, Khan I, Klimchak AC, Reynolds MR, Sanchez RJ, Sasiela WJ. Simulation of lipid‐lowering therapy intensification in a population with atherosclerotic cardiovascular disease. JAMA Cardiol. 2017;2:959–966. doi: 10.1001/jamacardio.2017.2289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Cannon CP, Sanchez RJ, Klimchak AC, Khan I, Sasiela WJ, Reynolds MR, Rosenson RS. Simulation of the impact of statin intolerance on the need for ezetimibe and/or proprotein convertase subtilisin/kexin type 9 inhibitor for meeting low‐density lipoprotein cholesterol goals in a population with atherosclerotic cardiovascular disease. Am J Cardiol. 2019;123:1202–1207. doi: 10.1016/j.amjcard.2019.01.028 [DOI] [PubMed] [Google Scholar]
  • 79. European Medicines Agency . Nustendi—Summary of Product Characteristics. Available at: https://www.ema.europa.eu/en/documents/product‐information/nustendi‐epar‐product‐information_en.pdf. Accessed April 8, 2022.
  • 80. Merck & Co., Inc . Zetia—Product Information. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/021445s033lbl.pdf. Accessed April 8, 2022.
  • 81. Li C, Lin L, Zhang W, Zhou L, Wang H, Luo X, Luo H, Cai Y, Zeng C. Efficiency and safety of proprotein convertase subtilisin/kexin 9 monoclonal antibody on hypercholesterolemia: a meta‐analysis of 20 randomized controlled trials. J Am Heart Assoc. 2015;4:e001937. doi: 10.1161/JAHA.115.001937 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Lipinski MJ, Benedetto U, Escarcega RO, Biondi‐Zoccai G, Lhermusier T, Baker NC, Torguson R, Brewer HB Jr, Waksman R. The impact of proprotein convertase subtilisin‐kexin type 9 serine protease inhibitors on lipid levels and outcomes in patients with primary hypercholesterolaemia: a network meta‐analysis. Eur Heart J. 2016;37:536–545. doi: 10.1093/eurheartj/ehv563 [DOI] [PubMed] [Google Scholar]
  • 83. Navarese EP, Kolodziejczak M, Schulze V, Gurbel PA, Tantry U, Lin Y, Brockmeyer M, Kandzari DE, Kubica JM, D'Agostino RB Sr, et al. Effects of proprotein convertase subtilisin/kexin type 9 antibodies in adults with hypercholesterolemia: a systematic review and meta‐analysis. Ann Intern Med. 2015;163:40–51. doi: 10.7326/M14-2957 [DOI] [PubMed] [Google Scholar]
  • 84. Peng W, Qiang F, Peng W, Qian Z, Ke Z, Yi L, Jian Z, Chongrong Q. Therapeutic efficacy of PCSK9 monoclonal antibodies in statin‐nonresponsive patients with hypercholesterolemia and dyslipidemia: a systematic review and meta‐analysis. Int J Cardiol. 2016;222:119–129. doi: 10.1016/j.ijcard.2016.07.239 [DOI] [PubMed] [Google Scholar]
  • 85. Zhao Z, Du S, Shen S, Luo P, Ding S, Wang G, Wang L. Comparative efficacy and safety of lipid‐lowering agents in patients with hypercholesterolemia: a frequentist network meta‐analysis. Medicine (Baltimore). 2019;98:e14400. doi: 10.1097/MD.0000000000014400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Casula M, Olmastroni E, Boccalari MT, Tragni E, Pirillo A, Catapano AL. Cardiovascular events with PCSK9 inhibitors: an updated meta‐analysis of randomised controlled trials. Pharmacol Res. 2019;143:143–150. doi: 10.1016/j.phrs.2019.03.021 [DOI] [PubMed] [Google Scholar]
  • 87. Guedeney P, Giustino G, Sorrentino S, Claessen BE, Camaj A, Kalkman DN, Vogel B, Sartori S, De Rosa S, Baber U, et al. Efficacy and safety of alirocumab and evolocumab: a systematic review and meta‐analysis of randomized controlled trials. Eur Heart J. 2019;43:e17‐e25. doi: 10.1093/eurheartj/ehz430 [DOI] [PubMed] [Google Scholar]
  • 88. Zhang XL, Zhu QQ, Zhu L, Chen JZ, Chen QH, Li GN, Xie J, Kang LN, Xu B. Safety and efficacy of anti‐PCSK9 antibodies: a meta‐analysis of 25 randomized, controlled trials. BMC Med. 2015;13:123. doi: 10.1186/s12916-015-0358-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Karatasakis A, Danek BA, Karacsonyi J, Rangan BV, Roesle MK, Knickelbine T, Miedema MD, Khalili H, Ahmad Z, Abdullah S, et al. Effect of PCSK9 inhibitors on clinical outcomes in patients with hypercholesterolemia: a meta‐analysis of 35 randomized controlled trials. J Am Heart Assoc. 2017;6:e006910. doi: 10.1161/JAHA.117.006910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Burnett H, Fahrbach K, Cichewicz A, Jindal R, Tarpey J, Durand A, Di Domenico M, Reichelt A, Viljoen A. Comparative efficacy of non‐statin lipid‐lowering therapies in patients with hypercholesterolemia at increased cardiovascular risk: a network meta‐analysis. Curr Med Res Opin. 2022;38:777–784. doi: 10.1080/03007995.2022.2049164 [DOI] [PubMed] [Google Scholar]
  • 91. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, Edelberg JM, Goodman SG, Hanotin C, Harrington RA, et al. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N Engl J Med. 2018;379:2097–2107. doi: 10.1056/NEJMoa1801174 [DOI] [PubMed] [Google Scholar]
  • 92. ClinicalTrials.gov . Evaluation of major cardiovascular events in patients with, or at high risk for, cardiovascular disease who are statin intolerant treated with bempedoic acid (ETC‐1002) or placebo (CLEAR Outcomes). Available at: https://clinicaltrials.gov/ct2/show/NCT02993406?cond=bempedoic+acid&draw=2&rank=9. Accessed April 8, 2022.
  • 93. ClinicalTrials.gov . Treatment With bempedoic acid and/or its fixed‐dose combination with ezetimibe in hypercholesterolemia or mixed dyslipidemia (MILOS). Available at: https://clinicaltrials.gov/ct2/show/NCT04579367?cond=bempedoic+acid&draw=2&rank=1. Accessed April 8, 2022.
  • 94. ClinicalTrials.gov . A randomized trial assessing the effects of inclisiran on clinical outcomes among people with cardiovascular disease (ORION‐4). Available at: https://clinicaltrials.gov/ct2/show/NCT03705234?cond=inclisiran&draw=2&rank=6. Accessed April 8, 2022.
  • 95. ClinicalTrials.gov . Efficacy and safety evaluation of alirocumab in patients with heterozygous familial hypercholesterolemia or high cardiovascular risk patients with hypercholesterolemia on lipid modifying therapy. Avaialble at: https://ClinicalTrials.gov/show/NCT02107898. Accessed April 8, 2022.
  • 96. Bays H, Farnier M, Gaudet D, Weiss R, Ruiz JL, Watts GF, Gouni‐Berthold I, Robinson JG, Jones PH, Severance R, et al. Efficacy and safety of combining alirocumab with atorvastatin or rosuvastatin versus statin intensification or adding ezetimibe in high cardiovascular risk patients: ODYSSEY OPTIONS I and II. Circulation. 2014;130:2118. [Google Scholar]
  • 97. Teramoto T, Kobayashi M, Uno K, Takagi Y, Matsuoka O, Sugimoto M, Inoue S, Minami F, Baccara‐Dinet MT. Efficacy and safety of alirocumab in Japanese patients with hypercholesterolemia on stable statin therapy: first data with the 75 mg every two weeks dose. Circulation. 2014;130:A13651. [Google Scholar]
  • 98. ClinicalTrials.gov . Efficacy and safety evaluation of alirocumab SAR236553 (REGN727) in patients with primary hypercholesterolemia on stable atorvastatin therapy. Available at: http://ClinicalTrials.gov/show/NCT01812707. Accessed April 8, 2022.
  • 99. ClinicalTrials.gov . Goal achievement after utilizing an anti‐PCSK9 antibody in statin intolerant subjects‐4 (GAUSS‐4). Available at: https://clinicaltrials.gov/show/nct02634580. Accessed April 8, 2022. [DOI] [PMC free article] [PubMed]
  • 100. Amgen Inc . A double‐blind, randomized, placebo and ezetimibe controlled, multicenter study to evaluate safety, tolerability and efficacy of AMG 145 on LDL‐C in combination with statin therapy in subjects with primary hypercholesterolemia and mixed dyslipidemia: LAPLACE‐2 [data supplied by Amgen]. 2014.
  • 101. Amgen Inc . A double‐blind, randomized, placebo‐controlled, multicenter study to evaluate safety, tolerability and efficacy of evolocumab (AMG 145) on LDL‐C in combination with statin therapy in Japanese subjects with high cardiovascular risk and with hyperlipidemia or mixed dyslipidemia. YUKAWA 2 [data supplied by Amgen]. 2014.
  • 102. Ray KK, Landmesser U, Leiter LA, Kallend D, Dufour R, Karakas M, Hall T, Troquay RP, Turner T, Visseren FL, et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N Engl J Med. 2017;376:1430–1440. doi: 10.1056/NEJMoa1615758 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data S1

Tables S1–S5

Figures S1–S2


Articles from Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease are provided here courtesy of Wiley

RESOURCES