Next Article in Journal
Impact of Age and Sex Interaction on Post-Acute Sequelae of COVID-19: An Italian Cohort Study on Adults and Children
Next Article in Special Issue
Current Management of Highly Calcified Coronary Lesions: An Overview of the Current Status
Previous Article in Journal
Differences in Cerebral Oxygenation in Cardiogenic and Respiratory Cardiac Arrest Before, During, and After Cardiopulmonary Resuscitation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Soluble PCSK9 Inhibition: Indications, Clinical Impact, New Molecular Insights and Practical Approach—Where Do We Stand?

1
Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
2
Division of Cardiology, Ospedale Degli Infermi, ASL Biella, 13900 Biella, Italy
3
Structural Interventional Cardiology, Department of Clinical and Experimental Medicine, Clinica Medica, Careggi University Hospital, 50139 Florence, Italy
4
Vascular Physiopathology Unit, IRCCS Neuromed Mediterranean Neurological Institute, 86077 Pozzilli, Italy
5
Division of Cardiology, AOU “Policlinico G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98166 Messina, Italy
6
Division of Cardiology, IRCCS Hospital Galeazzi-Sant’Ambrogio, 20161 Milan, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(8), 2922; https://doi.org/10.3390/jcm12082922
Submission received: 22 March 2023 / Revised: 12 April 2023 / Accepted: 14 April 2023 / Published: 18 April 2023
(This article belongs to the Special Issue Interventional Cardiology: Current Challenges in Clinical Practice)

Abstract

:
Current research on cardiovascular prevention predominantly focuses on risk-stratification and management of patients with coronary artery disease (CAD) to optimize their prognosis. Several basic, translational and clinical research efforts aim to determine the etiological mechanisms underlying CAD pathogenesis and to identify lifestyle-dependent metabolic risk factors or genetic and epigenetic parameters responsible for CAD occurrence and/or progression. A log-linear association between the absolute exposure of LDL cholesterol (LDL-C) and the risk of atherosclerotic cardio-vascular disease (ASCVD) was well documented over the year. LDL-C was identified as the principal enemy to fight against, and soluble proprotein convertase subtilisin kexin type 9 (PCSK9) was attributed the role of a powerful regulator of blood LDL-C levels. The two currently available antibodies (alirocumab and evolocumab) against PCSK9 are fully human engineered IgG that bind to soluble PCSK9 and avoid its interaction with the LDLR. As documented by modern and dedicated “game-changer” trials, antibodies against soluble PCSK9 reduce LDL-C levels by at least 60 percent when used alone and up to 85 percent when used in combination with high-intensity statins and/or other hypolipidemic therapies, including ezetimibe. Their clinical indications are well established, but new areas of use are advocated. Several clues suggest that regulation of PCSK9 represents a cornerstone of cardiovascular prevention, partly because of some pleiotropic effects attributed to these newly developed drugs. New mechanisms of PCSK9 regulation are being explored, and further efforts need to be put in place to reach patients with these new therapies. The aim of this manuscript is to perform a narrative review of the literature on soluble PCSK9 inhibitor drugs, with a focus on their indications and clinical impact.

1. Avoidable Cardiovascular Risk: Global Burden, Target of Therapy and New Frontiers

In the context of a highly dynamic scenario with substantial medical achievements, coronary artery disease (CAD) remains the leading cause of mortality worldwide [1,2]. Therefore, current research predominantly focuses on the efficient prevention, risk-stratification, and management of patients with CAD to optimize their prognosis. Concurrently, several basic, translational and clinical research efforts aim to determine the etiological mechanisms underlying CAD pathogenesis [3,4,5,6,7,8,9,10] and to identify lifestyle-dependent metabolic risk factors or genetic and epigenetic parameters responsible for CAD occurrence and/or progression [11]. Thereby, clinicians could ultimately develop feasible and accurate risk assessment and prediction models with the potential to be incorporated into routine clinical practice.
Low-density lipoprotein (LDL) is the most widely recognized and studied modifiable risk factor associated with atherosclerotic cardiovascular disease (ASCVD). A log-linear association between the absolute exposure of LDL cholesterol (LDL-C) and the risk of ASCVD has been well documented over the years due to prospective cohort studies and randomize clinical trials [12,13,14,15,16].
LDL particle concentration can be lowered by reductions in saturated fat consumption and caloric intake, as well as by multiple classes of cholesterol-lowering therapies that have gone through continuous updates and improvements [17,18].
Efforts made to reduce LDL-C levels, manage major cardiovascular risk factors and identify new primary and secondary prevention targets aim to stabilize coronary plaques and consequently reduce cardiovascular events. Unstable coronary plaques that are prone to rupture and cause a wide range of immediate and long-term adverse cardiac events are characterized by large plaque burden, large lipid content, and thin fibrous caps. It is well established that statins can slow the progression of coronary atherosclerosis, reducing the inflammatory plaque burden. However, the effect of novel therapeutic targets, such as lipoprotein (a) (Lp(a)) and proprotein convertase subtilisin kexin type 9 (PCSK9), on plaque burden, composition and inflammatory pathways involved remains largely unknown [19,20].
PCSK9 was implicated in cholesterol metabolism, as shown by converging lines of investigation from the early 2000s. Cohen et al.’s experiments, performed in 2005, revealed that PCSK9 loss of function (LOF) gene variants were associated with lower LDL-C levels [21]. Individuals with PCSK9 LOF variants, who benefit from a lifetime of low LDL-C levels, also had reduced ASCVD risk. Circulating levels of PCSK9 represent the strongest regulator of cholesterol trafficking in the body.
PCSK9, a proteolytic enzyme, indirectly regulates serum LDL-C by causing the destruction of LDL receptors. Physiologically, the complex PCSK9/LDL receptor (LDLR) is internalized and destroyed by lysosomes. A lower number of LDL-R molecules on the cell surface is associated with a higher level of plasma LDL-C.
Anti-PCSK-9 monoclonal antibodies prevent the binding of extracellular PCSK9 to LDL-R and prevent its internalization and degradation. The effect is the reduction of plasma LDL-C [22].
Although entry of PCSK9 into cells is dependent on the LDLR, the bulk of internalized PCSK9 remains intact inside the cells for several hours. This time lag suggests that the embrace between PCSK9 and LDLR does not lead to immediate targeting of both ligands and receptors to the lysosome and that perhaps a recycling circuit is present that allows a single molecule of PCSK9 to return to the surface and act on new LDLR molecules that recycle [23,24]. PCSK9 action during time, as its inhibition, justifies a powerful effect on circulating LDL particles.
Given that PCSK9 inhibition has changed the therapeutic approach to LDL-C reduction and the outcome of patients at cardiovascular risk, moreover with fast-changing therapeutic platforms, the aim of this manuscript is to perform a narrative review of the literature on soluble PCSK9 inhibitor drugs with a focus on their indications and clinical impact.

2. PCSK9 Inhibitors’ Pleiotropic Effects

Outcome benefits with PCSK9 inhibition are related mainly to LDL lowering. However, cardiovascular risk reduction appears to exceed the benefit exclusively derived from LDL-C reduction. Various hypotheses ascribe to PCSK9 inhibition effects beyond reductions in LDL-C level [25]. PCSK9 is expressed by various cell types that are naturally involved in inflammation, atherosclerosis, thrombosis, and oncogenesis; thus, the inhibition of this protease may have different pleiotropic effects.
(a)
Anti-atherosclerotic effect: PCSK9 inhibitors reduce the concentrations of pro-inflammatory cytokines and increase the levels of interleukin-10 (an anti-inflammatory interleukin), decreasing the expression of TNF-α and the C-C chemokine receptor type 2 (CCR2) and thereby inhibiting the mechanisms that promote atherosclerosis. [26]. Moreover, PCSK9 monoclonal antibodies reduce the expression of NADPH oxidase, involved in oxidative stress [27], and lower the expression of membrane-adhesive molecules (ICAM and VCAM) interfering with the TLR4/NF-kB cascade [28].
(b)
Stabilization of atherosclerotic plaque: PCSK9 inhibition reduces the necrotic core of atheroma, constituting one of the main players in plaque instabilization, induces autophagy of inflammatory cells, and favors elimination of necrotic cells. These mechanisms promote plaque stabilization [29]. However, it was also reported that PCSK9 has direct proinflammatory effects on vessels, possibly through effects on another member of the LDLR family, the LDLR-related protein 1, or LRP1, which exerts a strong regulatory effect on the inflammatory stance of plaque macrophages. Loss of LRP1 induces inflammation, and PCSK9 in the plaque reduces LRP1 levels [30,31]. Thus, therapeutic blockades of PCSK9 via monoclonal antibodies could have on coronary plaques a combination of positive and negative effects. The balance of these is difficult to assess in exclusively clinical studies, given the tremendously beneficial effect of plasma LDL-C reduction [32,33].
(c)
Anti-aggregation and anticoagulant effects: PCSK-9 is involved in platelet activation and aggregation through indirect and direct stimulation of CD36 (scavenger receptor) and low-density lipoprotein receptor-1 (LOX-1) on the surface of platelets. CD36 is activated directly by PCSK-9, but also by LDL and ox-LDL, while LOX-1 is activated by inflammatory stimuli, and its expression is increased by PCSK-9. Platelet aggregation is also favored by stimulation of TLR2 on the cell surface through lipid–peroxide-modified phospholipids transported by Lp(a). Inhibition of PCSK-9, therefore, has an antithrombotic effect for several reasons: reducing levels of LDL and ox-LDL, reducing Lp(a), and decreasing LOX-1 expression [34]. PCSK-9 is involved in coagulation through the downregulation of the low-density lipoprotein receptor-related protein (LRP-1). This one physiologically decreases the levels of tissue factor and factor VIII [35].
(d)
Antineoplastic effect: LDL-C and triglycerides have a negative impact on the risk of developing cancer. Mutations causing a decrease in PCSK-9 activity are associated with inhibition of the progression of colorectal and breast cancer, mainly, but not only, through a decrease in LDL-C concentration [36].
(e)
PCSK9-I and sepsis: the Toll-like receptor (TLR) plays a key role in the immune response and is activated by lipid molecules associated with the pathogen cell walls (lipopolysaccharides, lipoteichoic acid and phospholipomannan). PCSK9 reduces the elimination of lipids by downregulating LDL-R, thereby promoting the development of sepsis and septic shock. Use of PCSK-9 inhibitors may reverse this pathway [37].

3. PCSK9 Monoclonal Antibodies

The two currently available antibodies (alirocumab and evolocumab) are fully human-engineered IgG antibodies that bind to soluble PCSK9 and avoid its interaction with the LDLR. A PCSK9-deficiency condition results in tremendous accumulation of LDLR on the membrane of hepatocytes and a vigorous clearance of LDL particles, which result in significant lowering of plasma LDL-C levels. The subcutaneous injection of alirocumab or evolocumab introduces antibodies that, within few hours, can capture not only all the circulating PCSK9 but also the newly secreted PCSK9 with a long-lasting effect, determining a tremendous reduction of LDL-C circulating levels. Alirocumab and evolocumab were initially approved by the United States Food and Drug Administration (FDA) “as an adjunct to diet and maximally tolerated statin therapy for treatment of adults with heterozygous familial hypercholesterolemia (HeFH) or clinical ASCVD who require additional lowering of LDL-C”, but for the scientific rewards conquered, the therapeutic indications of these two molecules have greatly expanded.

Pharmacokinetics and Drug Interactions

Alirocumab is commonly administered at a dosage of 75 or 150 mg every two weeks and reaches maximum plasma concentration in a time range of 3 to 7 days, with a predominant distribution in the bloodstream and a bioavailability of about 85 percent [38].
For several months to date, a 300 mg formulation has also been available in some European countries in monthly administration, and its diffusion is progressively growing.
The plasma half-life of alirocumab is about 17–20 days. The interval is reduced to 12 days for patients on statin therapy due to the normal increase in PCSK9 expression induced by statin. It is assumed that alirocumab, after binding to PCSK9, is degraded to amino acids by the endothelial reticulum system. There is no need for dosage adjustment according to age, sex, weight, renal or mild-to-moderate hepatic failure. There are limited data for patients with severe hepatic or renal impairment [39].
On the other hand, evolocumab is administered at a dosage of 140 or 420 mg. At the 140 mg dosage, it reaches peak median serum concentrations in 3–4 days with an absolute bioavailability of 72%. A proportional increase in bioavailability was observed for higher dosages. Evolocumab has a prevalent distribution in the bloodstream and a half-life of 11–17 days. It is eliminated in two different phases. At low concentrations, elimination occurs via saturation of PCSK9 binding; at higher concentrations, it occurs via a nonsaturable proteolytic pathway. There is no need for dosage adjustment according to age, sex, weight, renal or mild-to-moderate hepatic insufficiency [40,41].
As for alirocumab, because of increased PCSK9 expression, increased binding to PCSK9 was observed for Evolocumab in patients on statin therapy [42].

4. PCSK9 Monoclonal Antibodies’ Indications for Cardiovascular Primary Prevention

PCSK9 monoclonal antibodies, as documented by modern and dedicated “game-changer” trials (Table 1), reduce LDL-C levels by at least 60 percent when used alone and up to 85 percent when used in combination with high-intensity statins and/or other hypolipidemic therapies, including ezetimibe. Specifically, alirocumab and evolocumab reduce LDL-C levels by 46–73% when compared with placebo and by 30% when compared with ezetimibe [43,44,45].
Both alirocumab and evolocumab showed great efficacy in reducing LDL-C levels in patients with high or very high cardiovascular risk factors, including those with diabetes mellitus and chronic kidney failure, reducing ASCVD events [21,23]. In addition, PCSK9i increases HDL-C levels and reduces triglyceride and Lp(a) levels.
In the context of primary cardiovascular prevention, great attention should be paid to familial heterozygous hypercholesterolemia (HeFH). HeFH is a monogenic codominant disease responsible for early onset CVD (55 years for males, 60 years for females) due to persistently extreme plasma LDL-C values throughout life. It is generally caused by LOF mutations affecting the LDLR or apoB genes, or by gain-of-function mutations in PCSK9 genes. About 95% of HeFH forms can be attributed to LDLR mutations. Hypolipidemic therapy in these patients should be initiated as soon as possible, considering that they have a very high cardiovascular risk profile.
According to the latest ESC 2021 guidelines for the prevention of cardiovascular risk, the use, in primary prevention, of PCSK9 monoclonal antibodies in patients younger than 70 years of age is indicated, with an evidence class IC, in patients with high-risk HeFH who do not reach the LDL-C target (LDL-C ≤ 70 mg/dL or ≤1.8 mmol/L) and who are already on the maximally tolerated statin dose and ezetimibe therapy. However, not all patients tolerate statins. In this case, PCSK9 monoclonal antibodies are recommended in addition to ezetimibe with evidence class IIb (34–35).
Similarly, in patients without HeFH but with very high cardiovascular risk, already on maximally tolerated statin therapy and ezetimibe, a combination with PCSK9i (evidence class IIb) may be considered if the LDL-C target is not reached (Figure 1). In addition, 2018 AHA/ACC guidelines support the use of PCSK9i for primary prevention.
According to the reports, the use of PCSK9i should be evaluated in patients with HeFH aged 30 to 75 years, with LDL-C values of 100 mg/dL or higher (≥2.6 mmol/L), already on maximally tolerated statin dose and ezetimibe, and not reaching the desired therapeutic target [69,70].
Similarly, the use of PCSK9i can be evaluated in patients aged 40 to 75 with LDL-C at baseline of 200 mg/dL or higher (≥5.7 mmol/L), already on maximally tolerated statin therapy and ezetimibe, with achieved LDL-C values of 130 mg/dL or higher (≥3.4 mmol/L) [71].
On the other hand, homozygous FH (HoFH) is a rare genetic syndrome presenting with complete absence of functioning LDL-R. In this case, use of conventional therapy (statin, ezetimibe, PCSK9-i), which acts on the LDL-R pathway, is associated with scarce effects on LDL-C reduction. The therapeutic strategies in this group of patients do not act on the LDL-receptor [72].

5. PCSK9 Monoclonal Antibodies’ Indications for Cardiovascular Secondary Prevention in Chronic Setting

PCSK9 monoclonal antibodies successfully showed, in cardiovascular outcome trials, to consistently reduce, in the context of secondary prevention, the risk for ASCVD. The efficacy of soluble PCSK9 inhibition for secondary ASCVD prevention is supported by two large cardiovascular outcomes trials—the ODYSSEY OUTCOMES and the FOURIER [41,42].
The efficacy and safety of alirocumab was evaluated in the ODISSEY OUTCOMES randomized placebo-controlled trial, which included 18,924 post-ACS patients. Over a 2.8 year follow-up, the primary outcome (composite of death for coronary heart disease, non-fatal MI, fatal or non-fatal ischemic stroke, or unstable angina requiring hospitalization) for alirocumab vs. placebo was 9.5% vs. 11.1%, respectively, resulting in a relative risk reduction of 15% (absolute risk reduction 1.6%) [42].
Among secondary outcomes, the alirocumab group showed lower major coronary heart disease and cardiovascular events but not mortality reduction.
Likewise, in the FOURIER trial, 27,564 patients with clinical ASCVD and LDL-C ≥ 70 mg/dL or a non-HDL-C level ≥ 100 mg/dL on mostly high intensity statin ± ezetimibe were randomized to evolocumab 140 mg subcutaneously every 2 weeks or 420 mg monthly versus placebo. At a median follow-up of 2.2 years, there was a 15% relative risk reduction (1.5% absolute risk reduction) in the primary composite outcome of cardiovascular death, myocardial infarction, stroke, hospitalization for unstable angina, or coronary revascularization in the evolocumab group compared with controls. Among individual endpoints, those randomized to evolocumab had significantly lower rates of myocardial infarction (MI), strokes and coronary revascularization but did not have significant difference in cardiovascular deaths compared with placebo. More recently, the results from FOURIER-OLE (open label extension) were presented at the ESC 2022 congress. FOURIER-OLE was therefore conducted to better understand the long-term safety, tolerability, and risk of major cardiovascular events in patients receiving prolonged treatment with evolocumab. The median follow-up in the extension study was 5.0 years. The maximum exposure to evolocumab in FOURIER plus FOURIER-OLE was 8.4 years. Long-term LDL cholesterol lowering with evolocumab was safe and well tolerated for more than eight years and led to further reductions in cardiovascular events compared with delayed treatment initiation [73]. Furthermore, evaluation of treatment with evolocumab on coronary artery plaque using intravascular ultrasound among statin-treated individuals, in the GLAGOV randomized controlled trial, found a modest but significant reduction in percent atheroma volume (−1.0%, 95% CI: −1.8% to −0.64%). After 76 weeks of treatment, there was also a greater proportion of patients exhibiting plaque regression in the evolocumab group compared with control (64.3% vs. 47.3%) [59].

6. Very Early Treatment of Acute Coronary Syndrome with Monoclonal Antibodies of Soluble PCSK9

Beyond the improvement in percutaneous revascularization techniques and antithrombotic strategies that contributed to change the scenario of acute coronary syndromes (ACS) [74,75,76,77,78,79,80,81], large attention has been paid to immediate and aggressive lipid-lowering therapies with high-intensity statins that are strongly recommended after ACS by the ESC/EAS dyslipidemia guidelines. Because of the delayed onset of action of statins and the risk of event recurrence during the first weeks after ACS, rapid lowering of LDL-C below the recommended targets might be beneficial in this group of patients. However, whether non-statin lipid-lowering agents could induce a further reduction in the risk of major adverse cardiovascular events is still unknown [82].
The use of anti-PCSK9 therapies, in the currently recommended guidelines, has not been included in the short-term after ACS but only after 4–12 weeks if the LDL-C target is not reached with oral therapies [69].
Moreover, the two main trials that investigated the anti-PCSK9, ODYSSEY OUTCOMES and FOURIER trials did not include patients with recent ACS (within 30 days of randomization) [41,42,83].
Nevertheless, recent trials have assessed the use of evolocumab and alirocumab early after an ACS.
The EVOPACS (Evolocumab for Early Reduction of LDL Cholesterol Levels in Patients with Acute Coronary Syndromes) trial evaluated the feasibility, safety, and LDL-C-lowering efficacy of in-hospital administration of evolocumab (<72 h from ACS symptom onset). Eligible patients were randomly assigned through a 1:1 ratio to receive evolocumab 420 mg every 4 weeks or matching placebo, on top of atorvastatin 40 mg. The reduction in LDL-C levels was significantly evident at 4 weeks and was maintained at 8 weeks. LDL-C was reduced below target (<55 mg/dL) in 90.1% of patients in the evolocumab group as compared with 10.7% in the placebo group, with no differences in terms of safety between the two groups [84].
In addition, the EVACS (Evolocumab in Acute Coronary Syndrome) trial enrolled patients with NSTEMI within 24 h of presentation. In-hospital initiation of evolocumab 420 mg rapidly reduced LDL-C, and at day 3, LDL-C levels in the group treated with evolocumab were significantly lower than those in the placebo group (49.2 +/− 24 vs. 76.1 +/− 33 mg/dL), remaining lower until day 30 (Table 1) [67].
These two studies were not powered for cardiovascular outcomes or serious adverse events, and thus, this point needs further investigation.
Based on this consideration, the EMSIACS (Evolocumab added to Moderate-Intensity Statin Therapy on LDL-C lowering and Cardiovascular Adverse Events in Patients with ACS) trial is an ongoing trial evaluating the efficacy of evolocumab added to moderate-intensity statin therapy early after ACS in adult Chinese patients, on both LDL-C lowering effect and cardiovascular outcomes after 12 weeks and 1 year of treatment [68].
The risk of recurrent events, in patients who experienced ACS, is largely attributable to frequent coexistence of multiple nonobstructive lesions in non-culprit arteries.
The PACMAN-AMI (Effects of the PCSK9 Antibody Alirocumab on Coronary Atherosclerosis in Patients with Acute Myocardial Infarction) trial and the HUYGENS (High-Resolution Assessment of Coronary Plaques in a Global Evolocumab Randomized Study) trial evaluated the effect of early administration of alirocumab and evolocumab, respectively, on coronary atherosclerosis. It was assessed by intracoronary imaging (OCT for both trials, IVUS and NIRS only for PACMAN-AMI) of the non-culprit arteries in patients presenting with ACS, after a year of treatment in addition to high-intensity statin therapy. These studies demonstrated a trend toward plaque stabilization, with significant reduction in percent of atheroma volume (assessed by IVUS), reduction in maximum lipid core burden index within 4 mm (assessed by NIRS), and increase in minimal fibrous cap thickness (assessed by OCT) [56,85], confirming, also in the acute setting, the results of the GLAGOV trial [59].
None of these trials investigated cardiovascular outcomes, but it is likely that LDL-C lowering and plaque regression will improve clinical outcomes in this population.

7. Prescriptive Flow-Chart and Real-World Adoption

“The lower the better” has been the conceptual goal that led to the publication of the latest ESC/EAS dyslipidemia guidelines in 2019. It emphasizes the proportional reduction of CV events associated with absolute reduction of LDL-C. According to this, a more important role has been attributed to PCSK9 monoclonal antibodies in primary and secondary prevention [66].
In patients who do not achieve therapeutic LDL-C goal, first-line therapy remains maximally tolerated statins, with or without ezetimibe. However, data from different registries show that this association alone is not enough to reach the goal, and PCSK9-i would guarantee a huge benefit for this population. Consequentially, PCSK9i prescription has been introduced, with different classes of recommendation, in different clinical settings [66].
In high-risk CV patients who do not reach their LDL-C goal with high-intensity statin and ezetimibe, the use of PCSK9-i is recommended in secondary prevention (I, A), while it could be useful in primary prevention (IIb, C) [66].
New attention has been paid to the setting of the ACS and to the early use of PCSK9i. ODYSSEY OUTCOMES and FOURIER showed that in this setting of patients, PCSK9i therapy is associated with a significant reduction of absolute risk of ischemic events and overall mortality [41,42].
Therefore, in high-risk patients who have experienced an ACS, not reaching their therapeutic goal, the ESC/EAS guidelines recommend high-intensity statin and a new evaluation of LDL-C serum levels after 4–6 weeks. If LDL-C goal is not achieved, PCSK9i could be added as a combination therapy [66] (Figure 2).
Nevertheless, this gap in time might allow for a new CV event to occur with an avoidable increase in CV mortality, as several studies have shown [86].
Therefore, does a stepwise approach still make sense in this population? This is the reason why new guidelines suggest, for patients who present with an ACS and whose LDL-C levels are not within ideal parameters, despite already being on maximally tolerated statin dose and ezetimibe, the addition of a PCSK9 monoclonal antibody soon after the event (better during hospitalization if possible) (IIa, C).
Finally, FH, PAD and patients with statin-associated muscle symptoms (SAMS) are other high-CV-risk categories for which PCSK9i should be considered in association with high-intensity statin (without in case of SAMS) and ezetimibe to lower the LDL-C serum levels of patients who do not reach their goal [62,87,88,89].
Despite this and well-known higher therapeutic adherence, real-world data suggest that the use of PCSK9 therapy in daily practice is still very limited, probably for different reasons [90].
The first limit is that only few “prescription hubs” are available, despite the large number of eligible patients. Moreover, there are few local pharmacies where it is possible to withdraw these drugs. Finally, only a small register through which monitoring drug prescription, tolerability and safety is currently fillable. All these barriers dampen the extensive use of these drugs, as the DA VINCI study has shown. Even if the use of this therapy could have a significant positive impact on global cardiovascular health and on the economy of national healthcare systems, there is still need for great effort to ensure the outreach of these drugs to eligible patients [91].

8. Future Directions

Use of PCSK9-i helps to rapidly reach very low values of LDL-C, reducing the global CV risk, and confirming the postulate that not only “the lower”, but also “the earlier” is of crucial importance.
In the near future, it is likely that patients at very high risk with not-a-target LDL-C values will start directly with triple association (statin plus ezetimibe plus PCSK9-i) [91].
In the acute setting, it is presumable that the next set of guidelines will allow for the administration of PCSK9-i in the catheter lab during PCI, introducing the temporal concept of “door to PCSK9-i”, similar to the already known “door to balloon” concept.
The objective of reducing PCSK9 action to control LDL-C levels can also be achieved by inhibiting hepatic PCSK9 synthesis via targeted approaches to transcriptional regulation. The employment of small interfering ribonucleic acid (siRNA) oligonucleotides as pharmacological agents is appearing on the modern therapeutic scene.
Inclisiran is a double-stranded, chemically synthesized, long-acting anti-PCSK9 siRNA molecule. The antisense strand corresponds to PCSK9 mRNA, while the sense strand is conjugated with triantennary N-acetylgalactosamine carbohydrates (tri-GalNAc). This one is recognized by hepatocyte-specific asialoglycoprotein receptors. In hepatocytes, inclisiran activates the RNA-induced silencing complex (RISC), which promotes cleavage of the intracellular PCSK9 mRNA, inhibiting the protein translation of PCSK9 itself [92].
There are significant differences between the two approaches of PCSK9 inhibition that might eventually prove to have clinical relevance. Antisense intervention (a) mimics true relative PCSK9 deficiency seen in patients with PCSK9 LOF, (b) reduces intracellular levels of PCSK9, (c) shows no blockade of PCSK9 in the atheroma, (d) and does not induce nonphysiologic immune complex circulation. Entry of the siRNA approach (inclisiran) for PCSK9 inhibition into the marketplace could change the ecosystem of cholesterol therapies even more than the PCSK9-inhibiting antibodies have. This injectable agent could not only exhibit an even higher profile of efficacy and safety, but it is approved as a two-injection per year intervention, showing excellent adherence to the treatment [93].
Other approaches include oral small-molecule inhibitors, vaccination with the development of natural antibodies against PCSK9 and genic therapy with editing of the PCSK9 gene using clustered regularly interspaced short palindromic repeats (CRISPR) [94].
Vaccines. Two PCSK-9 vaccines (AT04A and AT06A) have been developed. They elicit the production of natural antibodies against PCSK9. Further investigations are needed for the durability of their effects [95].
CRISPR/Cas system (clustered regularly interspaced short palindromic repeats). This is a gene-editing technology that aims to introduce permanent genomic changes to alter gene function. The complex CRISPR/Cas introduces a double-strand break in the PCSK9 gene. Endogenous DNA repair mechanisms lead to random insertions or deletions, which render the gene dysfunctional [96].
Monobodies. Adnectin BMS-962476 is a small protein that inhibits free extracellular PCSK9. The drug is in preclinical phase but has shown, in non-human primates, adequate efficacy in reducing LDL [97].
Preclinical PCSK9 Inhibitors. EGF-A are mimetic peptides that block free PCSK9, enhancing, in vitro, LDL-R expression on the cell surface [98].
Several clues suggest that PCSK9 represents a pivotal factor in cardiovascular disease, in part independent from its effects on lipid metabolism. Statins, for example, have many pleiotropic effects on endothelial function, immune responses, inflammation, and thrombosis [99]. PCSK9-I, with a reasonable probability, has similar additive effects [100]. Results from trials are not univocal. Further research is needed to unveil pleiotropic activities of these drugs and to extent indications to other clinical conditions.
Today, the role of PCSK9 inhibition in lowering cardiovascular risk is well delineated. The modes of inhibition are increasingly being refined as well as the indications, which are being expanded and tailored. Future strategies for lowering CLDL and cardiovascular risk will allow for more scenarios to be attacked safely and effectively.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nowbar, A.N.; Gitto, M.; Howard, J.P.; Francis, D.P.; Al-Lamee, R. Mortality From Ischemic Heart Disease. Circ. Cardiovasc. Qual. Outcomes 2019, 12, e005375. [Google Scholar] [CrossRef] [PubMed]
  2. Silverio, A.; Di Maio, M.; Citro, R.; Esposito, L.; Iuliano, G.; Bellino, M.; Baldi, C.; De Luca, G.; Ciccarelli, M.; Vecchione, C.; et al. Cardiovascular risk factors and mortality in hospitalized patients with COVID-19: Systematic review and meta-analysis of 45 studies and 18,300 patients. BMC Cardiovasc. Disord. 2021, 21, 23. [Google Scholar] [CrossRef] [PubMed]
  3. Groenendyk, J.W.; Greenland, P.; Khan, S.S. Incremental Value of Polygenic Risk Scores in Primary Prevention of Coronary Heart Disease: A Review. JAMA Intern Med. 2022, 182, 1082–1088. [Google Scholar] [CrossRef] [PubMed]
  4. Verdoia, M.; Schaffer, A.; Barbieri, L.; Aimaretti, G.; Marino, P.; Sinigaglia, F.; Suryapranata, H.; De Luca, G.; Novara Atherosclerosis Study Group (NAS). Impact of diabetes on neutrophil-to-lymphocyte ratio and its relationship to coronary artery disease. Diabetes Metab. 2015, 41, 304–311. [Google Scholar] [CrossRef]
  5. De Luca, G.; Verdoia, M.; Cassetti, E.; Schaffer, A.; Cavallino, C.; Bolzani, V.; Marino, P.; Novara Atherosclerosis Study Group (NAS). High fibrinogen level is an independent predictor of presence and extent of coronary artery disease among Italian population. J. Thromb. Thrombolysis. 2011, 31, 458–463. [Google Scholar] [CrossRef]
  6. Barbieri, L.; Verdoia, M.; Schaffer, A.; Marino, P.; Suryapranata, H.; De Luca, G.; Novara Atherosclerosis Study Group (NAS). Impact of sex on uric acid levels and its relationship with the extent of coronary artery disease: A single-centre study. Atherosclerosis 2015, 241, 241–248. [Google Scholar] [CrossRef]
  7. Kedhi, E.; Berta, B.; Roleder, T.; Hermanides, R.S.; Fabris, E.; Ijsselmuiden, A.J.J.; Kauer, F.; Alfonso, F.; von Birgelen, C.; Escaned, J.; et al. Thin-cap fibroatheroma predicts clinical events in diabetic patients with normal fractional flow reserve: The COMBINE OCT–FFR trial. Eur. Heart J. 2021, 42, 4671–4679. [Google Scholar] [CrossRef]
  8. Baldi, C.; Silverio, A.; Esposito, L.; Di Maio, M.; Tarantino, F.; De Angelis, E.; Fierro, G.; Attisano, T.; Di Muro, M.R.; Maione, A.; et al. Clinical outcome of patients with ST-elevation myocardial infarction and angiographic evidence of coronary artery ectasia. Catheter. Cardiovasc. Interv. 2022, 99, 340–347. [Google Scholar] [CrossRef]
  9. Esposito, L.; Di Maio, M.; Silverio, A.; Cancro, F.P.; Bellino, M.; Attisano, T.; Tarantino, F.F.; Esposito, G.; Vecchione, C.; Galasso, G.; et al. Treatment and Outcome of Patients with Coronary Artery Ectasia: Current Evidence and Novel Opportunities for an Old Dilemma. Front. Cardiovasc. Med. 2022, 8, 805727. [Google Scholar] [CrossRef]
  10. Silverio, A.; Buccheri, S.; Venetsanos, D.; Alfredsson, J.; Lagerqvist, B.; Persson, J.; Witt, N.; James, S.; Sarno, G. Percutaneous Treatment and Outcomes of Small Coronary Vessels: A SCAAR Report. JACC Cardiovasc. Interv. 2020, 13, 793–804. [Google Scholar] [CrossRef]
  11. Mozaffarian, D.; Wilson, P.W.; Kannel, W.B. Beyond established and novel risk factors: Lifestyle risk factors for cardiovascular disease. Circulation 2008, 117, 3031–3038. [Google Scholar] [CrossRef] [PubMed]
  12. Galasso, G.; De Angelis, E.; Silverio, A.; Di Maio, M.; Cancro, F.P.; Esposito, L.; Bellino, M.; Scudiero, F.; Damato, A.; Parodi, G.; et al. Predictors of Recurrent Ischemic Events in Patients with ST-Segment Elevation Myocardial Infarction. Am. J. Cardiol. 2021, 159, 44–51. [Google Scholar] [CrossRef] [PubMed]
  13. Scudiero, F.; Canonico, M.E.; Sanna, G.D.; Dossi, F.; Silverio, A.; Galasso, G.; Esposito, G.; Porto, I.; Parodi, G. Dual Antiplatelet Therapy with 3rd Generation P2Y12 Inhibitors in STEMI Patients: Impact of Body Mass Index on Loading Dose–Response. Cardiovasc. Drugs Ther. 2022, 1–9. [Google Scholar] [CrossRef] [PubMed]
  14. Ference, B.A.; Ginsberg, H.N.; Graham, I.; Ray, K.K.; Packard, C.J.; Bruckert, E.; Hegele, R.A.; Krauss, R.M.; Raal, F.J.; Schunkert, H.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2017, 38, 2459–2472. [Google Scholar] [CrossRef]
  15. Baigent, C.; Keech, A.; Kearney, P.M.; Blackwell, L.; Buck, G.; Pollicino, C.; Kirby, A.; Sourjina, T.; Peto, R.; Collins, R.; et al. Efficacy and safety of cholesterol-lowering treatment: Prospective meta-analysis of data from 90,056 participants in 14 randomized trials of statins. Lancet 2005, 366, 1267–1278. [Google Scholar] [CrossRef] [PubMed]
  16. Silverio, A.; Benvenga, R.M.; Piscione, F.; Gulizia, M.M.; Meessen, J.M.T.A.; Colivicchi, F.; Nardi, F.; Baldi, C.; Galasso, G.; Vecchione, C.; et al. Prevalence and Predictors of Out-of-Target LDL Cholesterol 1 to 3 Years After Myocardial Infarction. A Subanalysis From the EYESHOT Post-MI Registry. J. Cardiovasc. Pharmacol. Ther. 2021, 26, 149–157. [Google Scholar] [CrossRef]
  17. Stone, N.J.; Robinson, J.G.; Lichtenstein, A.H.; Merz, C.N.B.; Blum, C.B.; Eckel, R.H.; Goldberg, A.C.; Gordon, D.; Levy, D.; Lloyd-Jones, D.M.; et al. 2013 ACC/AHA Guideline on the Treatment of Blood Cholesterol to Reduce Atherosclerotic Cardiovascular Risk in Adults: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guideline. J. Am. Coll. Cardiol. 2014, 63, 2889–2934. [Google Scholar] [CrossRef]
  18. Catapano, A.L.; Graham, I.; De Backer, G.; Wiklund, O.; Chapman, M.J.; Drexel, H.; Hoes, A.W.; Jennings, C.S.; Landmesser, U.; Pedersen, T.R.; et al. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias: The Task Force for the Management of Dyslipidaemias of the European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) Developed with the special contribution of the European Assocciation for Cardiovascular Prevention & Rehabilitation (EACPR). Atherosclerosis 2016, 253, 281–344. [Google Scholar] [CrossRef]
  19. Koskinas, K.C.; Siontis, G.C.M.; Piccolo, R.; Mavridis, D.; Räber, L.; Mach, F.; Windecker, S. Effect of statins and non-statin LDL-lowering medications on cardiovascular outcomes in secondary prevention: A meta-analysis of randomized trials. Eur. Heart J. 2018, 39, 1172–1180. [Google Scholar] [CrossRef]
  20. Silverio, A.; Cancro, F.P.; Di Maio, M.; Bellino, M.; Esposito, L.; Centore, M.; Carrizzo, A.; Di Pietro, P.; Borrelli, A.; De Luca, G.; et al. Lipoprotein(a) levels and risk of adverse events after myocardial infarction in patients with and without diabetes. J. Thromb. Thrombolysis 2022, 54, 382–392. [Google Scholar] [CrossRef]
  21. Cohen, J.; Pertsemlidis, A.; Kotowski, I.K.; Graham, R.; Garcia, C.K.; Hobbs, H.H. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat. Genet. 2005, 37, 161–165. [Google Scholar] [CrossRef] [PubMed]
  22. Coppinger, C.; Movahed, M.R.; Azemawah, V.; Peyton, L.; Gregory, J.; Hashemzadeh, M. A Comprehensive Review of PCSK9 Inhibitors. J. Cardiovasc. Pharmacol. Ther. 2022, 27, 10742484221100107. [Google Scholar] [CrossRef]
  23. Tavori, H.; Fan, D.; Blakemore, J.L.; Yancey, P.G.; Ding, L.; Linton, M.F.; Fazio, S. Serum Proprotein Convertase Subtilisin/Kexin Type 9 and Cell Surface Low-Density Lipoprotein Receptor: Evidence for a reciprocal regulation. Circulation 2013, 127, 2403–2413. [Google Scholar] [CrossRef] [PubMed]
  24. Nguyen, M.-A.; Kosenko, T.; Lagace, T.A. Internalized PCSK9 dissociates from recycling LDL receptors in PCSK9-resistant SV-589 fibroblasts. J. Lipid Res. 2014, 55, 266–275. [Google Scholar] [CrossRef] [PubMed]
  25. Benn, M.; Nordestgaard, B.G.; Grande, P.; Schnohr, P.; Tybjærg-Hansen, A. PCSK9R46L, Low-Density Lipoprotein Cholesterol Levels, and Risk of Ischemic Heart Disease: 3 Independent Studies and Meta-Analyses. J. Am. Coll. Cardiol. 2010, 55, 2833–2842. [Google Scholar] [CrossRef]
  26. Basiak, M.; Kosowski, M.; Cyrnek, M.; Bułdak, Ł.; Maligłówka, M.; Machnik, G.; Okopień, B. Pleiotropic Effects of PCSK-9 Inhibitors. Int. J. Mol. Sci. 2021, 22, 3144. [Google Scholar] [CrossRef]
  27. Ding, Z.; Liu, S.; Wang, X.; Deng, X.; Fan, Y.; Sun, C.; Wang, Y.; Mehta, J.L. Hemodynamic Shear Stress via ROS Modulates PCSK9 Expression in Human Vascular Endothelial and Smooth Muscle Cells and along the Mouse Aorta. Antioxidants Redox Signal. 2015, 22, 760–771. [Google Scholar] [CrossRef]
  28. Tang, Z.-H.; Peng, J.; Ren, Z.; Yang, J.; Li, T.-T.; Li, T.-H.; Wang, Z.; Wei, D.-H.; Liu, L.-S.; Zheng, X.-L.; et al. New role of PCSK9 in atherosclerotic inflammation promotion involving the TLR4/NF-κB pathway. Atherosclerosis 2017, 262, 113–122. [Google Scholar] [CrossRef]
  29. Omori, H.; Ota, H.; Hara, M.; Kawase, Y.; Tanigaki, T.; Hirata, T.; Sobue, Y.; Okubo, M.; Kamiya, H.; Matsuo, H. Effect of PCSK-9 Inhibitors on Lipid-Rich Vulnerable Coronary Plaque Assessed by Near-Infrared Spectroscopy. JACC Cardiovasc. Imaging 2020, 13, 1639–1641. [Google Scholar] [CrossRef]
  30. Zhu, L.; Giunzioni, I.; Tavori, H.; Covarrubias, R.; Ding, L.; Zhang, Y.; Ormseth, M.; Major, A.S.; Stafford, J.M.; Linton, M.F.; et al. Loss of Macrophage Low-Density Lipoprotein Receptor–Related Protein 1 Confers Resistance to the Antiatherogenic Effects of Tumor Necrosis Factor-α Inhibition. Arter. Thromb. Vasc. Biol. 2016, 36, 1483–1495. [Google Scholar] [CrossRef]
  31. Desai, N.R.; Giugliano, R.; Wasserman, S.M.; Gibbs, J.P.; Liu, T.; Scott, R.; Sabatine, M.S. Association between Circulating Baseline Proprotein Convertase Subtilisin Kexin Type 9 Levels and Efficacy of Evolocumab. JAMA Cardiol. 2017, 2, 556–560. [Google Scholar] [CrossRef] [PubMed]
  32. Tomlinson, B.; Hu, M.; Zhang, Y.; Chan, P.; Liu, Z.M. Alirocumab for the treatment of hypercholesterolemia. Expert Opin. Biol. Ther. 2017, 17, 633–643. [Google Scholar] [CrossRef]
  33. Kasichayanula, S.; Grover, A.; Emery, M.G.; Gibbs, M.A.; Somaratne, R.; Wasserman, S.M.; Gibbs, J.P. Clinical Pharmacokinetics and Pharmacodynamics of Evolocumab, a PCSK9 Inhibitor. Clin. Pharmacokinet. 2018, 57, 769–779. [Google Scholar] [CrossRef] [PubMed]
  34. Puccini, M.; Landmesser, U.; Rauch, U. Pleiotropic Effects of PCSK9: Focus on Thrombosis and Haemostasis. Metabolites 2022, 12, 226. [Google Scholar] [CrossRef] [PubMed]
  35. Canuel, M.; Sun, X.; Asselin, M.-C.; Paramithiotis, E.; Prat, A.; Seidah, N.G. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Can Mediate Degradation of the Low Density Lipoprotein Receptor-Related Protein 1 (LRP-1). PLoS ONE 2013, 8, e64145. [Google Scholar] [CrossRef]
  36. Liu, X.; Bao, X.; Hu, M.; Chang, H.; Jiao, M.; Cheng, J.; Xie, L.; Huang, Q.; Li, F.; Li, C.-Y. Inhibition of PCSK9 potentiates immune checkpoint therapy for cancer. Nature 2020, 588, 693–698. [Google Scholar] [CrossRef]
  37. Walley, K.R.; Thain, K.R.; Russell, J.A.; Reilly, M.P.; Meyer, N.J.; Ferguson, J.F.; Christie, J.D.; Nakada, T.-A.; Fjell, C.D.; Thair, S.A.; et al. PCSK9 is a critical regulator of the innate immune response and septic shock outcome. Sci. Transl. Med. 2014, 6, 258ra143. [Google Scholar] [CrossRef]
  38. Gibbs, J.P.; Slatter, J.G.; Egbuna, O.; Geller, M.; Hamilton, L.; Dias, C.S.; Xu, R.Y.; Johnson, J.; Wasserman, S.M.; Emery, M.G. Evaluation of Evolocumab (AMG 145), a Fully Human Anti-PCSK9 IgG2 Monoclonal Antibody, in Subjects with Hepatic Impairment. J. Clin. Pharmacol. 2017, 57, 513–523. [Google Scholar] [CrossRef]
  39. Lee, E.; Gibbs, J.P.; Emery, M.G.; Block, G.; Wasserman, S.M.; Hamilton, L.; Kasichayanula, S.; Hanafin, P.; Somaratne, R.; Egbuna, O. Influence of Renal Function on Evolocumab Exposure, Pharmacodynamics, and Safety. Clin. Pharmacol. Drug Dev. 2019, 8, 281–289. [Google Scholar] [CrossRef]
  40. Dubuc, G.; Chamberland, A.; Wassef, H.; Davignon, J.; Seidah, N.G.; Bernier, L.; Prat, A. Statins Upregulate PCSK9, the Gene Encoding the Proprotein Convertase Neural Apoptosis-Regulated Convertase-1 Implicated in Familial Hypercholesterolemia. Arter. Thromb. Vasc. Biol. 2004, 24, 1454–1459. [Google Scholar] [CrossRef]
  41. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M.; et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef] [PubMed]
  42. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A.; et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef] [PubMed]
  43. Cho, L.; Rocco, M.; Colquhoun, D.; Sullivan, D.; Rosenson, R.S.; Dent, R.; Xue, A.; Scott, R.; Wasserman, S.M.; Stroes, E. Clinical Profile of Statin Intolerance in the Phase 3 GAUSS-2 Study. Cardiovasc. Drugs Ther. 2016, 30, 297–304. [Google Scholar] [CrossRef] [PubMed]
  44. Moriarty, P.M.; Thompson, P.D.; Cannon, C.P.; Guyton, J.R.; Bergeron, J.; Zieve, F.J.; Bruckert, E.; Jacobson, T.A.; Kopecky, S.L.; Baccara-Dinet, M.T.; 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. [Google Scholar] [CrossRef]
  45. Nissen, S.E.; Stroes, E.; Dent-Acosta, R.E.; Rosenson, R.S.; Lehman, S.J.; Sattar, N.; Preiss, D.; Bruckert, E.; Ceška, 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. [Google Scholar] [CrossRef] [PubMed]
  46. Ray, K.K.; Colhoun, H.M.; Szarek, M.; Baccara-Dinet, M.; Bhatt, D.L.; Bittner, A.V.; Budaj, A.J.; Diaz, R.; Goodman, S.G.; Hanotin, C.; et al. Effects of alirocumab on cardiovascular and metabolic outcomes after acute coronary syndrome in patients with or without diabetes: A prespecified analysis of the ODYSSEY OUTCOMES randomised controlled trial. Lancet Diabetes Endocrinol. 2019, 7, 618–628. [Google Scholar] [CrossRef] [PubMed]
  47. Roth, E.M.; McKenney, J.M. ODYSSEY MONO: Effect of alirocumab 75 mg subcutaneously every 2 weeks as monotherapy versus ezetimibe over 24 weeks. Futur. Cardiol. 2015, 11, 27–37. [Google Scholar] [CrossRef]
  48. Kereiakes, D.J.; Robinson, J.G.; Cannon, C.P.; Lorenzato, C.; Pordy, R.; Chaudhari, U.; Colhoun, H.M. 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.e13. [Google Scholar] [CrossRef]
  49. Cannon, C.P.; Cariou, B.; Blom, D.; McKenney, J.M.; Lorenzato, C.; Pordy, R.; Chaudhari, U.; Colhoun, H.M. 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. [Google Scholar] [CrossRef]
  50. Bays, H.; Gaudet, D.; Weiss, R.; Ruiz, J.L.; Watts, G.; 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. [Google Scholar] [CrossRef]
  51. Farnier, M.; Jones, P.; Severance, R.; Averna, M.; Steinhagen-Thiessen, E.; Colhoun, H.M.; 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. [Google Scholar] [CrossRef] [PubMed]
  52. Ginsberg, H.N.; Rader, D.J.; Raal, F.J.; Guyton, J.R.; Baccara-Dinet, M.T.; 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. [Google Scholar] [CrossRef] [PubMed]
  53. Robinson, J.G.; Farnier, M.; Krempf, M.; Bergeron, J.; Luc, G.; Averna, M.; Stroes, E.S.; Langslet, G.; Raal, F.J.; El Shahawy, M.; et al. Efficacy and Safety of Alirocumab in Reducing Lipids and Cardiovascular Events. N. Engl. J. Med. 2015, 372, 1489–1499. [Google Scholar] [CrossRef] [PubMed]
  54. Kastelein, J.J.P.; Robinson, J.G.; Farnier, M.; Krempf, M.; Langslet, G.; Lorenzato, C.; Gipe, D.A.; Baccara-Dinet, M.T. Efficacy and Safety of Alirocumab in Patients with Heterozygous Familial Hypercholesterolemia not Adequately Controlled with Current Lipid-Lowering Therapy: Design and Rationale of the ODYSSEY FH Studies. Cardiovasc. Drugs Ther. 2014, 28, 281–289. [Google Scholar] [CrossRef] [PubMed]
  55. Stroes, E.; Guyton, J.R.; Lepor, N.; Civeira, F.; Gaudet, D.; Watts, G.F.; Baccara-Dinet, M.T.; 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. [Google Scholar] [CrossRef]
  56. Räber, L.; Ueki, Y.; Otsuka, T.; Losdat, S.; Häner, J.D.; Lonborg, J.; Fahrni, G.; Iglesias, J.F.; van Geuns, R.-J.; Ondracek, A.S.; et al. Effect of Alirocumab Added to High-Intensity Statin Therapy on Coronary Atherosclerosis in Patients With Acute Myocardial Infarction: The PACMAN-AMI Randomized Clinical Trial. JAMA 2022, 327, 1771. [Google Scholar] [CrossRef]
  57. Giugliano, R.P.; Pedersen, T.R.; Park, J.-G.; De Ferrari, G.M.; Gaciong, A.Z.; 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. [Google Scholar] [CrossRef]
  58. Blom, D.J.; Hala, T.; Bolognese, M.; Lillestol, M.J.; Toth, P.D.; Burgess, L.; Ceska, R.; Roth, E.; Koren, M.J.; Ballantyne, C.M.; et al. A 52-Week Placebo-Controlled Trial of Evolocumab in Hyperlipidemia. N. Engl. J. Med. 2014, 370, 1809–1819. [Google Scholar] [CrossRef]
  59. Nicholls, S.J.; Puri, R.; Anderson, T.; Ballantyne, C.M.; Cho, L.; Kastelein, J.J.P.; 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. [Google Scholar] [CrossRef]
  60. Stroes, E.; Colquhoun, D.; Sullivan, D.; Civeira, F.; Rosenson, R.S.; Watts, G.F.; 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. [Google Scholar] [CrossRef]
  61. Koren, M.J.; Lundqvist, P.; Bolognese, M.; Neutel, J.M.; Monsalvo, M.L.; Yang, J.; Kim, J.B.; Scott, R.; Wasserman, S.M.; Bays, H. Anti-PCSK9 Monotherapy for Hypercholesterolemia: The MENDEL-2 randomized, controlled phase III clinical trial of evolocumab. J. Am. Coll. Cardiol. 2014, 63, 2531–2540. [Google Scholar] [CrossRef] [PubMed]
  62. Raal, F.J.; Stein, A.E.; Dufour, R.; Turner, T.; Civeira, F.; Burgess, L.; Langslet, G.; Scott, R.; Olsson, A.G.; 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. [Google Scholar] [CrossRef] [PubMed]
  63. Robinson, J.G.; Nedergaard, B.S.; Rogers, W.J.; Fialkow, J.; Neutel, J.M.; Ramstad, D.; Somaratne, R.; Legg, J.C.; 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, 1870–1883. [Google Scholar] [CrossRef] [PubMed]
  64. Raal, F.J.; Honarpour, N.; Blom, D.J.; Hovingh, G.K.; Xu, F.; Scott, R.; Wasserman, S.M.; Stein, A.E. Inhibition of PCSK9 with evolocumab in homozygous familial hypercholesterolaemia (TESLA Part B): A randomised, double-blind, placebo-controlled trial. Lancet 2015, 385, 341–350. [Google Scholar] [CrossRef] [PubMed]
  65. Sabatine, M.S.; Giugliano, R.P.; Wiviott, S.D.; Raal, F.J.; Blom, D.J.; Robinson, J.; Ballantyne, C.M.; Somaratne, R.; Legg, J.; Wasserman, S.M.; et al. Efficacy and Safety of Evolocumab in Reducing Lipids and Cardiovascular Events. N. Engl. J. Med. 2015, 372, 1500–1509. [Google Scholar] [CrossRef] [PubMed]
  66. Nicholls, S.J.; Nissen, S.E.; Prati, F.; Windecker, S.; Kataoka, Y.; Puri, R.; Hucko, T.; Kassahun, H.; Liao, J.; Somaratne, R.; et al. Assessing the impact of PCSK9 inhibition on coronary plaque phenotype with optical coherence tomography: Rationale and design of the randomized, placebo-controlled HUYGENS study. Cardiovasc. Diagn. Ther. 2021, 11, 120–129. [Google Scholar] [CrossRef]
  67. Koskinas, K.C.; Windecker, S.; Pedrazzini, G.; Mueller, C.; Cook, S.; Matter, C.M.; Muller, O.; Häner, J.; Gencer, B.; Crljenica, C.; et al. Evolocumab for Early Reduction of LDL Cholesterol Levels in Patients With Acute Coronary Syndromes (EVOPACS). J. Am. Coll. Cardiol. 2019, 74, 2452–2462. [Google Scholar] [CrossRef]
  68. Leucker, T.M.; Blaha, M.J.; Jones, S.R.; Vavuranakis, M.A.; Williams, M.S.; Lai, H.; Schindler, T.H.; Latina, J.; Schulman, S.P.; Gerstenblith, G. Effect of Evolocumab on Atherogenic Lipoproteins During the Peri- and Early Postinfarction Period: A Placebo-Controlled, Randomized Trial. Circulation 2020, 142, 419–421. [Google Scholar] [CrossRef]
  69. Grundy, S.M.; Stone, N.J.; Bailey, A.L.; Beam, C.; Birtcher, K.K.; Blumenthal, R.S.; Braun, L.T.; de Ferranti, S.; Faiella-Tommasino, J.; Forman, D.E.; 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. [Google Scholar] [CrossRef]
  70. Silverman, M.G.; Ference, B.A.; Im, K.; Wiviott, S.D.; Giugliano, R.; Grundy, S.M.; Braunwald, E.; Sabatine, M.S. Association Between Lowering LDL-C and Cardiovascular Risk Reduction Among Different Therapeutic Interventions: A Systematic Review and Meta-analysis. JAMA 2016, 316, 1289–1297. [Google Scholar] [CrossRef]
  71. Vallejo-Vaz, A.J.; Robertson, M.; Catapano, A.L.; Watts, G.F.; Kastelein, J.J.; Packard, C.J.; Ford, I.; Ray, K.K. Low-Density Lipoprotein Cholesterol Lowering for the Primary Prevention of Cardiovascular Disease among Men With Primary Elevations of Low-Density Lipoprotein Cholesterol Levels of 190 mg/dL or above: Analyses From the WOSCOPS (West of Scotland Coronary Prevention Study) 5-Year Randomized Trial and 20-Year Observational Follow-Up. Circulation 2017, 136, 1878–1891. [Google Scholar] [CrossRef] [PubMed]
  72. Palacio, C.H.; Harring, T.R.; Nguyen, N.T.T.; Goss, J.A.; O’Mahony, C.A. Homozygous Familial Hypercholesterolemia: Case Series and Review of the Literature. Case Rep. Transplant. 2011, 2011, 154908. [Google Scholar] [CrossRef] [PubMed]
  73. O’donoghue, M.L.; Giugliano, R.P.; Wiviott, S.D.; Atar, D.; Keech, A.C.; Kuder, J.F.; Im, K.; Murphy, S.A.; Flores-Arredondo, J.H.; López, J.A.G.; et al. Long-Term Evolocumab in Patients With Established Atherosclerotic Cardiovascular Disease. Circulation 2022, 146, 1109–1119. [Google Scholar] [CrossRef]
  74. De Luca, G.; Navarese, E.P.; Suryapranata, H. A meta-analytic overview of thrombectomy during primary angioplasty. Int. J. Cardiol. 2013, 166, 606–612. [Google Scholar] [CrossRef] [PubMed]
  75. De Luca, G.; Schaffer, A.; Wirianta, J.; Suryapranata, H. Comprehensive meta-analysis of radial vs femoral approach in primary angioplasty for STEMI. Int. J. Cardiol. 2013, 168, 2070–2081. [Google Scholar] [CrossRef]
  76. De Luca, G.; Suryapranata, H.; Stone, G.W.; Antoniucci, D.; Tcheng, J.E.; Neumann, F.J.; Bonizzoni, E.; Topol, E.J.; Chiariello, M. Relationship between patient’s risk profile and benefits in mortality from adjunctive abciximab to mechanical revascularization for ST-segment elevation myocardial infarction: A meta-regression analysis of randomized trials. J. Am. Coll. Cardiol. 2006, 47, 685–686. [Google Scholar] [CrossRef]
  77. De Luca, G.; Navarese, E.P.; Cassetti, E.; Verdoia, M.; Suryapranata, H. Meta-analysis of randomized trials of glycoprotein IIb/IIIa inhibitors in high-risk acute coronary syndromes patients undergoing invasive strategy. Am. J. Cardiol. 2011, 107, 198–203. [Google Scholar] [CrossRef]
  78. Verdoia, M.; Schaffer, A.; Barbieri, L.; Cassetti, E.; Piccolo, R.; Galasso, G.; Marino, P.; Sinigaglia, F.; De Luca, G. Benefits from new ADP antagonists as compared with clopidogrel in patients with stable angina or acute coronary syndrome undergoing invasive management: A meta-analysis of randomized trials. J. Cardiovasc. Pharmacol. 2014, 63, 339–350. [Google Scholar] [CrossRef]
  79. Costa, F.; Montalto, C.; Branca, M.; Hong, S.J.; Watanabe, H.; Franzone, A.; Vranckx, P.; Hahn, J.Y.; Gwon, H.C.; Feres, F.; et al. Dual antiplatelet therapy duration after percutaneous coronary intervention in high bleeding risk: A meta-analysis of randomized trials. Eur. Heart J. 2023, 44, 954–968. [Google Scholar] [CrossRef]
  80. Secco, G.G.; Ghione, M.; Mattesini, A.; Dall’ara, G.; Ghilencea, L.; Kilickesmez, K.; De Luca, G.; Fattori, R.; Parisi, R.; Marino, P.N.; et al. Very high-pressure dilatation for undilatable coronary lesions: Indications and results with a new dedicated balloon. Eurointervention 2016, 12, 359–365. [Google Scholar] [CrossRef]
  81. De Luca, G.; Smits, P.; Hofma, S.H.; Di Lorenzo, E.; Vlachojannis, G.; Van’t Hof, A.W.J.; van Boven, A.J.; Kedhi, E.; Stone, G.W.; Suryapranata, H. Drug-Eluting Stent in Primary Angioplasty (DESERT 3) cooperation. Everolimus eluting stent vs first generation drug-eluting stent in primary angioplasty: A pooled patient-level meta-analysis of randomized trials. Int. J. Cardiol. 2017, 244, 121–127. [Google Scholar] [CrossRef] [PubMed]
  82. Cimaglia, P.; Fortini, F.; Vieceli Dalla Sega, F.; Cardelli, L.S.; Massafra, R.F.; Morelli, C.; Trichilo, M.; Ferrari, R.; Rizzo, P.; Campo, G. Relationship between PCSK9 and endothelial function in patients with acute myocardial infarction. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 2105–2111. [Google Scholar] [CrossRef] [PubMed]
  83. Ferri, N.; Ruscica, M.; Lupo, M.G.; Vicenzi, M.; Sirtori, C.R.; Corsini, A. Pharmacological rationale for the very early treatment of acute coronary syndrome with monoclonal antibodies anti-PCSK9. Pharmacol. Res. 2022, 184, 106439. [Google Scholar] [CrossRef]
  84. Sinnaeve, P.R.; Schwartz, G.G.; Wojdyla, D.M.; Alings, M.; Bhatt, D.L.; Bittner, A.V.; Chiang, C.-E.; Flores, R.M.C.; Diaz, R.; Dorobantu, M.; et al. Effect of alirocumab on cardiovascular outcomes after acute coronary syndromes according to age: An ODYSSEY OUTCOMES trial analysis. Eur. Heart J. 2019, 41, 2248–2258. [Google Scholar] [CrossRef] [PubMed]
  85. Gao, J.; Liu, J.-Y.; Lu, P.-J.; Xiao, J.-Y.; Gao, M.-D.; Li, C.-P.; Cui, Z.; Liu, Y. Effects of Evolocumab Added to Moderate-Intensity Statin Therapy in Chinese Patients With Acute Coronary Syndrome: The EMSIACS Trial Study Protocol. Front. Physiol. 2021, 12, 750872. [Google Scholar] [CrossRef]
  86. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur. Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef] [PubMed]
  87. Navarese, E.P.; Andreotti, F.; Raggi, P.; Kołodziejczak, M.; Buffon, A.; Bliden, K.; Tantry, U.; Kubica, J.; Sardella, G.; Lauten, A.; et al. Baseline low-density lipoprotein cholesterol to predict the extent of cardiovascular benefit from lipid-lowering therapies: A review. Eur. Heart J. Cardiovasc. Pharmacother. 2019, 5, 47–54. [Google Scholar] [CrossRef]
  88. Aday, A.W.; Matsushita, K. Epidemiology of Peripheral Artery Disease and Polyvascular Disease. Circ. Res. 2021, 128, 1818–1832. [Google Scholar] [CrossRef]
  89. Bonaca, M.P.; Nault, P.; Giugliano, R.; Keech, A.C.; Pineda, A.L.; Kanevsky, E.; Kuder, J.; Murphy, S.A.; Jukema, J.W.; Lewis, B.S.; et al. Low-Density Lipoprotein Cholesterol Lowering with Evolocumab and Outcomes in Patients with Peripheral Artery Disease: Insights from the FOURIER Trial (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk). Circulation 2018, 137, 338–350. [Google Scholar] [CrossRef]
  90. Raal, F.; Scott, R.; Somaratne, R.; Bridges, I.; Li, G.; Wasserman, S.M.; Stein, E.A. 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. [Google Scholar] [CrossRef]
  91. März, W.; Dippel, F.-W.; Theobald, K.; Gorcyca, K.; Iorga, R.; Ansell, D. Utilization of lipid-modifying therapy and low-density lipoprotein cholesterol goal attainment in patients at high and very-high cardiovascular risk: Real-world evidence from Germany. Atherosclerosis 2018, 268, 99–107. [Google Scholar] [CrossRef] [PubMed]
  92. Hardy, J.; Niman, S.; Pereira, E.; Lewis, T.; Reid, J.; Choksi, R.; Goldfaden, R.F. A Critical Review of the Efficacy and Safety of Inclisiran. Am. J. Cardiovasc. Drugs 2021, 21, 629–642. [Google Scholar] [CrossRef]
  93. Ray, K.K.; Reeskamp, L.F.; Laufs, U.; Banach, M.; Mach, F.; Tokgözoğlu, L.S.; Connolly, D.L.; Gerrits, A.J.; Stroes, E.S.G.; Masana, L.; et al. Combination lipid-lowering therapy as first-line strategy in very high-risk patients. Eur. Heart J. 2022, 43, 830–833. [Google Scholar] [CrossRef] [PubMed]
  94. Rosenson, R.S.; Hegele, R.A.; Fazio, S.; Cannon, C.P. The Evolving Future of PCSK9 Inhibitors. J. Am. Coll. Cardiol. 2018, 72, 314–329. [Google Scholar] [CrossRef] [PubMed]
  95. Karagiannis, A.D.; Liu, M.; Toth, P.P.; Zhao, S.; Agrawal, D.K.; Libby, P.; Chatzizisis, Y.S. Pleiotropic Anti-atherosclerotic Effects of PCSK9 Inhibitors From Molecular Biology to Clinical Translation. Curr. Atheroscler. Rep. 2018, 20, 20. [Google Scholar] [CrossRef]
  96. Katzmann, J.L.; Cupido, A.J.; Laufs, U. Gene Therapy Targeting PCSK9. Metabolites 2022, 12, 70. [Google Scholar] [CrossRef]
  97. Mitchell, T.; Chao, G.; Sitkoff, D.; Lo, F.; Monshizadegan, H.; Meyers, D.; Low, S.; Russo, K.; DiBella, R.; Denhez, F.; et al. Pharmacologic Profile of the Adnectin BMS-962476, a Small Protein Biologic Alternative to PCSK9 Antibodies for Low-Density Lipoprotein Lowering. J. Pharmacol. Exp. Ther. 2014, 350, 412–424. [Google Scholar] [CrossRef]
  98. McNutt, M.C.; Kwon, H.J.; Chen, C.; Chen, J.R.; Horton, J.D.; Lagace, T.A. Antagonism of Secreted PCSK9 Increases Low Density Lipoprotein Receptor Expression in HepG2 Cells. J. Biol. Chem. 2009, 284, 10561–10570. [Google Scholar] [CrossRef]
  99. Hapiro, M.D.; Tavori, H.; Fazio, S. PCSK9: From Basic Science Discoveries to Clinical Trials. Circ. Res. 2018, 122, 1420–1438. [Google Scholar] [CrossRef]
  100. Paciullo, F.; Momi, S.; Gresele, P. PCSK9 in Haemostasis and Thrombosis: Possible Pleiotropic Effects of PCSK9 Inhibitors in Cardiovascular Prevention. Thromb. Haemost. 2019, 119, 359–367. [Google Scholar] [CrossRef]
Figure 1. Flowchart of interventions for primary cardiovascular prevention according to the European Society of Cardiology guidelines. CVR: cardiovascular risk, C-LDL: LDL cholesterol, FH: familiar hypercholesterolemia.
Figure 1. Flowchart of interventions for primary cardiovascular prevention according to the European Society of Cardiology guidelines. CVR: cardiovascular risk, C-LDL: LDL cholesterol, FH: familiar hypercholesterolemia.
Jcm 12 02922 g001
Figure 2. Flowchart of interventions for secondary cardiovascular prevention according to the European Society of Cardiology guidelines. CVR: cardiovascular risk, LDL-C: LDL cholesterol.
Figure 2. Flowchart of interventions for secondary cardiovascular prevention according to the European Society of Cardiology guidelines. CVR: cardiovascular risk, LDL-C: LDL cholesterol.
Jcm 12 02922 g002
Table 1. List of trials on PCSK9i with characteristics of patients’ population, number of patients enrolled and percentage of LDL-C reduction achieved. ACS: acute coronary syndrome; CV: cardiovascular; HeFH: heterozygous familiar hypercholesterolemia; HoFH: homozygous familial hypercholesterolemia; IRA: infarct related artery; LLT: lipid lowering therapy; NK: not known; Q2W: once every 2 weeks; SC: subcutaneous.
Table 1. List of trials on PCSK9i with characteristics of patients’ population, number of patients enrolled and percentage of LDL-C reduction achieved. ACS: acute coronary syndrome; CV: cardiovascular; HeFH: heterozygous familiar hypercholesterolemia; HoFH: homozygous familial hypercholesterolemia; IRA: infarct related artery; LLT: lipid lowering therapy; NK: not known; Q2W: once every 2 weeks; SC: subcutaneous.
Trial NamePatients’ PopulationPatientsTreatmentsLDL Reduction
ALIROCUMAB
ODYSSEY OUTCOMES [46]Prior ACS (1–12 months earlier) and LDL > 70 mg/dL on statin therapy18924Alirocumab 75 mg Q2W vs. placebo55%
ODYSSEY MONO [47]Hypercholesterolemia (with or without statin)103Alirocumab 75/150 mg and oral placebo vs. Ezetimibe and subcutaneous placebo47.2%
ODYSSEY ALTERNATIVE [44]Moderate (or higher) CV risk with statin intolerance251Alirocumab 75/150 mg plus oral placebo vs. ezetimibe plus SC placebo vs. atorvastatin 20 mg plus SC placebo52.2%
ODYSSEY COMBO I [48]High CV risk in uncontrolled hypercholesterolemia on maximally tolerated statin therapy +/− other LLT316Alirocumab 75/150 mg Q2W vs. placebo48.2%
ODYSSEY COMBO II [49]High CV risk in uncontrolled hypercholesterolemia on maximally tolerated statin therapy +/− other LLT720Alirocumab 75 mg Q2W (plus oral placebo) vs. ezetimibe (plus SC placebo)50.6%
ODYSSEY OPTIONS I [50]Very high CV risk and LDL > 70 mg/dl or high CV risk and LDL > 100 mg/dl on atorvastatin 20/40 mg355Alirocumab 75 mg Q2W vs. ezetimibe vs. double-dose atorvastatin vs. switch to rosuvastatin 40 mg44.1/54%
ODYSSEY OPTIONS II [51]Very high CV risk and LDL > 70 mg/dl or high risk and LDL > 100 mg/dL on rosuvastatin 10/20 mg305Alirocumab 75 mg Q2W vs. ezetimibe vs. double-dose rosuvastatin50.6/36.3%
ODYSSEY HIGH FH [52]HeFH and LDL > 160 mg/dL on maximally tolerated statin +/− other LLT107Alirocumab 150 mg Q2W vs. placebo46%
ODYSSEY LONG TERM [53]High CV risk and LDL > 70 mg/dL on maximally tolerated statin +/− other LLT2341Alirocumab 150 mg Q2W vs. placebo62%
ODYSSEY FH I and II [54]HeFH and uncontrolled LDL on maximally tolerated statin +/− other LLT486/249Alirocumab 75/150 mg Q2W vs. placebo57.9/51.4%
ODYSSEY CHOICE II [55]Statin intolerance and hypercholesterolemia233Alirocumab 150 mg Q4W or 75 mg Q2W vs. placebo51.7/53.5%
PACMAN-AMI [56]Intracoronary imaging of non-IRAs in early ACS on rosuvastatin 20 mg300Alirocumab 150 mg Q2W vs. placebo−54.7 mg/dL
EVOLOCUMAB
FOURIER [57]Prior CVD and LDL > 70 mg/dL
Background therapy of statin +/− ezetimibe
27564Evolocumab 140 mg Q2W or 420 mg monthly vs. placebo59%
DESCARTES [58]LDL > 75 mg/dL
Background therapy of statin +/− ezetimibe
901Evolocumab 420 mg monthly vs. placebo55%
GLAGOV [59]Angiographic coronary disease and LDL > 80 mg/dL or 60-80 mg/dL plus any risk factor968Evolocumab 420 mg monthly vs. placebo−56.5 mg/dL
GAUSS-2 [60]Hypercholesterolemia with statin intolerance3071. Evolocumab 140 mg Q2W or 420 mg monthly2. Ezetimibe3. Oral and subcutaneous placebo54%
MENDEL-2 [61]Hypercholesterolemia (LDL > 100 mg/dL) and CV risk < 10%6141. Evolocumab 140 mg Q2W or 420 mg monthly2. Ezetimibe3. Oral and subcutaneous placebo58%
RUTHERFORD-2 [62]HeFH and LDL > 100%
Background therapy of statin +/− ezetimibe
331Evolocumab 140 mg Q2W or 420 mg monthly vs. placebo56%
LAPLACE-2 [63]Hypercholesterolemia
Background therapy of statin +/− ezetimibe
20671. Moderate-intensity vs. high-intensity statin2. Evolocumab 140 mg Q2W or 420 mg monthly3. Ezetimibe4. Oral and subcutaneous placebo70%
TESLA Part B [64]HoFH and LDL > 130 mg/dL on maximal therapy50Evolocumab 420 mg monthly vs. placebo23,1%
OSLER-1 and OSLER-2 (open label) [65]Hypercolesterolemia on standard therapy4465Evolocumab 420 mg monthly or 140 mg Q2W plus standard therapy vs. standard therapy alone61%
HUYGENS [66]Intracoronary imaging of non-IRAs in early ACS on maximally tolerated statin therapy150Evolocumab 420 mg monthly vs. placeboNK
EVOPACS [67]In-hospital phase of ACS on atorvastatin 40 mg308Evolocumab 420 mg monthly vs. placebo40,7%
EVACS [68]In-hospital phase of ACS on maximally tolerated statin therapy60Evolocumab 420 mg vs. placebo−27 mg/dL
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bellino, M.; Galasso, G.; Silverio, A.; Tedeschi, M.; Formisano, C.; Romei, S.; Esposito, L.; Cancro, F.P.; Vassallo, M.G.; Accarino, G.; et al. Soluble PCSK9 Inhibition: Indications, Clinical Impact, New Molecular Insights and Practical Approach—Where Do We Stand? J. Clin. Med. 2023, 12, 2922. https://doi.org/10.3390/jcm12082922

AMA Style

Bellino M, Galasso G, Silverio A, Tedeschi M, Formisano C, Romei S, Esposito L, Cancro FP, Vassallo MG, Accarino G, et al. Soluble PCSK9 Inhibition: Indications, Clinical Impact, New Molecular Insights and Practical Approach—Where Do We Stand? Journal of Clinical Medicine. 2023; 12(8):2922. https://doi.org/10.3390/jcm12082922

Chicago/Turabian Style

Bellino, Michele, Gennaro Galasso, Angelo Silverio, Michele Tedeschi, Ciro Formisano, Stefano Romei, Luca Esposito, Francesco Paolo Cancro, Maria Giovanna Vassallo, Giulio Accarino, and et al. 2023. "Soluble PCSK9 Inhibition: Indications, Clinical Impact, New Molecular Insights and Practical Approach—Where Do We Stand?" Journal of Clinical Medicine 12, no. 8: 2922. https://doi.org/10.3390/jcm12082922

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

Article Metrics

Back to TopTop