Targeted Strategy in Lipid-Lowering Therapy - MDPI

Page created by Regina Taylor
 
CONTINUE READING
Targeted Strategy in Lipid-Lowering Therapy - MDPI
biomedicines
Review
Targeted Strategy in Lipid-Lowering Therapy
Ezgi Dayar           and Olga Pechanova *

                                          Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences,
                                          Sienkiewiczova 1, 813 71 Bratislava, Slovakia; ezgi.dayar@savba.sk
                                          * Correspondence: olga.pechanova@savba.sk; Tel.: +421-911-938-910

                                          Abstract: Dyslipidemia is characterized by a diminished lipid profile, including increased level of
                                          total cholesterol and low-density lipoprotein cholesterol (LDL-c) and reduced level of high-density
                                          lipoprotein cholesterol (HDL-c). Lipid-lowering agents represent an efficient tool for the prevention
                                          or reduction of progression of atherosclerosis, coronary heart diseases and metabolic syndrome.
                                          Statins, ezetimibe, and recently proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are
                                          the most effective and used drugs in clinical lipid-lowering therapy. These drugs are mainly aimed
                                          to lower cholesterol levels by different mechanisms of actions. Statins, the agents of the first-line
                                          therapy—known as 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitors—suppress
                                          the liver cholesterol synthesis. Ezetimibe as the second-line therapy can decrease cholesterol by
                                          inhibiting cholesterol absorption. Finally, the PCSK9 inhibitors act as an inducer of LDL excretion. In
                                          spite of their beneficial lipid-lowering properties, many patients suffer from their serious side effects,
                                          route of administration, or unsatisfactory physicochemical characteristics. Clinical demand for dose
                                          reduction and the improvement of bioavailability as well as pharmacodynamic and pharmacokinetic
                                          profile has resulted in the development of a new targeted therapy that includes nanoparticle carriers,
                                          emulsions or vaccination often associated with another more subtle form of administration. Targeted
                                          therapy aims to exert a more potent drug profile with lipid-lowering properties either alone or in
                                          mutual combination to potentiate their beneficial effects. This review describes the most effective
                                          lipid-lowering drugs, their favorable and adverse effects, as well as targeted therapy and alternative
                                          treatments to help reduce or prevent atherosclerotic processes and cardiovascular events.
Citation: Dayar, E.; Pechanova, O.
Targeted Strategy in Lipid-Lowering       Keywords: dyslipidemia; cholesterol; metabolic syndrome; statins; ezetimibe; PCSK9 inhibitors;
Therapy. Biomedicines 2022, 10, 1090.     nanoparticles; targeted therapy
https://doi.org/10.3390/
biomedicines10051090

Academic Editor: Anca Sima
                                          1. Introduction
Received: 14 April 2022
Accepted: 3 May 2022
                                               Metabolic disorders are disorders that adversely affect the distribution of macronu-
Published: 8 May 2022
                                          trients such as lipids, carbohydrates, and proteins. They are basically a consequence of
                                          abnormal chemical reactions in the body that alter the normal metabolic process. While
Publisher’s Note: MDPI stays neutral
                                          congenital metabolic disorders are caused by genetic defects, acquired metabolic disor-
with regard to jurisdictional claims in
                                          ders are associated with external factors, such as an unhealthy lifestyle, little physical
published maps and institutional affil-
                                          activity, and excessive caloric intake (for review see [1]). Eckel et al. (2010) documented
iations.
                                          that human lifestyle is associated with an inherited epigenetic pattern, which affects gene
                                          expression, and protein activity that leads to the development of metabolic disorders [2].
                                          Metabolic syndrome is the most common metabolic disorder and represents a cluster of
Copyright: © 2022 by the authors.
                                          conditions that occur together and increase the risk of heart disease, stroke, and type 2
Licensee MDPI, Basel, Switzerland.        diabetes. These conditions include increased blood pressure, high blood glucose, obesity,
This article is an open access article    and dyslipidemia [3,4]. Dyslipidemia, manifested by elevated low-density lipoprotein
distributed under the terms and           cholesterol (LDL-c), is the primary cause of the development and progression of atheroscle-
conditions of the Creative Commons        rosis. Atherosclerosis is initiated by multiple interactions between oxidatively modified
Attribution (CC BY) license (https://     lipids and lipoproteins, inflammatory factors, and components of the immune system in the
creativecommons.org/licenses/by/          arterial wall that result in the formation of fatty streaks and fibrous plaques. Plaque buildup
4.0/).                                    and rupture can eventually lead to progressive stenosis and thrombosis. Atherosclerosis

Biomedicines 2022, 10, 1090. https://doi.org/10.3390/biomedicines10051090                               https://www.mdpi.com/journal/biomedicines
Biomedicines 2022, 10, 1090                                                                                            2 of 18

                              may progress silently for a longer period of time until it causes a first cardiovascular event.
                              Analyses of coronary arteries have indicated that dyslipidemia and obesity were predictive
                              of earlier and greater extent of atherosclerosis in large vessels, increase of coronary fatty
                              streaks, and even recurrent coronary events [5,6].
                                   Consequently, dyslipidemia has become a key intervention in the prevention of car-
                              diovascular diseases. In this meaning, therapeutic lifestyle changes and the use of lipid-
                              lowering drugs are the most recommended options for avoiding coronary heart diseases.
                              The main effect of lipid-lowering drugs is the reduction of the plasma low-density lipopro-
                              tein or the enhancement of high-density lipoproteins [5,6]. 3- hydroxy-3-methylglutaryl-
                              CoA (HMG-CoA) reductase inhibitors, Niemann-Pick C1-like 1 (NPC1L1) protein inhibitors,
                              and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are among the most
                              effective and hitherto most used drugs in lipid-lowering therapy. These substances affect
                              respective lipid metabolic pathways and reduce the production or absorption of cholesterol
                              and LDL [7]. However, in addition to their beneficial pleiotropic properties, they have
                              also adverse side effects. Therefore, the goal of a targeted therapy is the reduction of
                              the dose of lipid-lowering drugs while simultaneously achieving a sufficiently effective
                              impact. However, many patients show intolerance to these drugs, so routine treatment
                              needs to be replaced by appropriate alternatives. This review describes the most effective
                              lipid-lowering drugs, their beneficial and adverse side effects, as well as targeted therapy
                              and alternative treatments.

                              2. Routine Lipid-Lowering Therapy
                              2.1. Statins
                                   Statin therapy represents the gold standard of dyslipidemia treatment. Statins are
                              prescribed as the first-line pharmacological therapy for the reduction of cardiovascular
                              risk [8,9]. The mechanism by which statins act to reduce liver cholesterol production is
                              based on the competitive inhibition of rate-controlling enzyme in cholesterol synthesis
                              HMG-CoA reductase. This enzyme catalyzes the conversion of HMG-CoA to a mevalonic
                              acid, a necessary step in the biosynthesis of cholesterol (Figure 1). Among other products
                              of this pathway are also ubiquinones including coenzyme Q [9,10]. HMG-CoA reductase
                              is active during higher blood glucose level. The basic function of insulin and glucagon
                              is the maintenance of glucose homeostasis. Thus, in controlling blood sugar levels, they
                              indirectly affect the activity of HMG-CoA reductase [11].
                                   Apart from cholesterol-lowering effects, statins also have a wide range of well-
                              documented pleiotropic effects including the improvement of the endothelial function
                              and anti-inflammatory, anti-thrombotic, and immunomodulatory effects for vascular en-
                              dothelial as well as smooth muscle cells [8,12]. The improvement of the endothelial function
                              is predominantly associated with positive effects of statins on nitric oxide (NO)/reactive
                              oxygen species (ROS) balance, upregulation of endothelial nitric oxide synthase (eNOS)
                              and tetrahydrobiopterin stabilization [8,13–15], while anti-inflammatory effects of statins
                              are attributed to their ability to modulate cytokine production [16].
                                   According to the guidelines of the American College of Cardiology (ACC)/American
                              Heart Association (AHA), the dose and the type of statin treatment regimen differ by
                              requiring the reduction of LDL-c rate. However, for all statins, the maximum U.S. Food
                              and Drug Administration (FDA)-approved dose is 80 mg, except for rosuvastatin, which
                              is 40 mg/day. For patients who need a reduction in LDL-c of over 50%, atorvastatin
                              40–80 mg, simvastatin 80 mg, rosuvastatin 20 mg or combination statin with ezetimibe
                              10/40 mg are prescribed [17,18]. In spite of highly beneficial effects of statins, they have a
                              low bioavailability. Long-term treatment especially with higher doses may therefore lead
                              to serious side effects such as myopathy, muscle inflammation, joint pains, hemorrhagic
                              stroke, increase of liver enzymes, memory loss, and some neurological disorders [12,19].
                              The original definition of statin-induced myopathy is the unexplained muscle pain or
                              weakness accompanied by more than 10 times higher creatine kinase concentration. Statin-
                              induced rhabdomyolysis is a severe form of myopathy with more than 40 times higher
Biomedicines 2022, 10, x FOR PEER REVIEW                                                                                                      3 of 18

Biomedicines 2022, 10, 1090                                                                                                                            3 of 18

                                   accompanied by more than 10 times higher creatine kinase concentration. Statin-induced
                                   rhabdomyolysis is a severe form of myopathy with more than 40 times higher than the
                                   basal the
                                   than   activity
                                              basalofactivity
                                                       creatineofkinase andkinase
                                                                   creatine  muscleandfibermuscle
                                                                                            necrosis which
                                                                                                  fiber     often which
                                                                                                        necrosis   results often
                                                                                                                           in myoglo-
                                                                                                                                 results in
                                   binuria and acute
                                   myoglobinuria      andrenal failure
                                                           acute  renal[20]. In addition,
                                                                        failure            prolongation
                                                                                [20]. In addition,       of statin of
                                                                                                    prolongation   therapy
                                                                                                                      statin leads to aleads
                                                                                                                             therapy
                                   decrease
                                   to         in coenzyme
                                      a decrease             Q concentration
                                                   in coenzyme                 of up to
                                                                    Q concentration    of40% with
                                                                                          up to   corresponding
                                                                                                40%                 consequences
                                                                                                     with corresponding            (for
                                                                                                                             consequences
                                   review
                                   (for     see [21].
                                        review   see [21].

                                    Figure 1.
                                   Figure   1. The
                                               Themechanism
                                                     mechanismbybywhichwhich statins
                                                                                 statinsactact
                                                                                            to to
                                                                                               reduce   liver
                                                                                                   reduce     cholesterol
                                                                                                           liver          production
                                                                                                                  cholesterol           is based
                                                                                                                                production         on the
                                                                                                                                             is based    on the
                                    competitive inhibition
                                   competitive     inhibition of  rate-controlling enzyme
                                                               of rate-controlling     enzyme in     cholesterol synthesis
                                                                                                  in cholesterol   synthesis 3-hydroxy-3-methylglu-
                                                                                                                              3-hydroxy-3-methylglutaryl-
                                    taryl-CoA (HMG-CoA) reductase. This enzyme catalyzes the conversion of HMG-CoA to a mevalo-
                                   CoA (HMG-CoA) reductase. This enzyme catalyzes the conversion of HMG-CoA to a mevalonic acid,
                                    nic acid, a necessary step in the biosynthesis of cholesterol. Ezetimibe inhibits the absorption of
                                   acholesterol
                                      necessaryfromstepsmall
                                                        in theintestine
                                                                biosynthesis
                                                                         leading  oftocholesterol.
                                                                                       the reduction  Ezetimibe    inhibits
                                                                                                        in intestinal       the absorption
                                                                                                                       cholesterol transmissionof cholesterol
                                                                                                                                                    to the
                                    liver [19,22].
                                   from             The main
                                          small intestine      mechanism
                                                            leading   to the isreduction
                                                                                 associated  in with  the inhibition
                                                                                                intestinal            of the
                                                                                                            cholesterol       Niemann-Pick
                                                                                                                         transmission    to theC1-like   1
                                                                                                                                                 liver [19,22].
                                    (NPC1L1)
                                   The   main protein,
                                                mechanism the key   factor of cholesterol
                                                                is associated                  absorption.of
                                                                                  with the inhibition         Proprotein  convertase C1-like
                                                                                                                the Niemann-Pick       subtilisin/kexin
                                                                                                                                                 1 (NPC1L1)
                                    type 9 (PCSK9)
                                   protein,   the key inhibitors
                                                         factor ofinhibit  the binding
                                                                     cholesterol            of PCSK9
                                                                                     absorption.        with low-density
                                                                                                      Proprotein             lipoprotein
                                                                                                                     convertase           (LDL)-recep-
                                                                                                                                  subtilisin/kexin      type 9
                                    tors and  prevent   the  degradation    of  LDL-receptors.       The  inhibition  of PCSK9    is
                                   (PCSK9) inhibitors inhibit the binding of PCSK9 with low-density lipoprotein (LDL)-receptors and  targeting  also   via
                                    small interfering RNA (siRNA) against PCSK9 synthesis and expression, small molecules, and vac-
                                   prevent the degradation of LDL-receptors. The inhibition of PCSK9 is targeting also via small
                                    cination against PCSK9.
                                   interfering RNA (siRNA) against PCSK9 synthesis and expression, small molecules, and vaccination
                                    2.2. Ezetimibe
                                   against   PCSK9.

                                         Ezetimibe inhibits the absorption of cholesterol from small intestine leading to the
                                   2.2. Ezetimibe
                                   reduction  in intestinal cholesterol transmission to the liver [19,22]. The main mechanism
                                   is associated
                                          Ezetimibewith  the inhibition
                                                       inhibits             of the NPC1L1
                                                                  the absorption               protein, from
                                                                                      of cholesterol       the key    factor
                                                                                                                   small       of cholesterol
                                                                                                                           intestine    leadingab-
                                                                                                                                                 to the
                                   sorption (Figure
                                   reduction            1) located
                                                in intestinal        on the gastrointestinal
                                                                 cholesterol    transmission totract       epithelial
                                                                                                      the liver          cellsThe
                                                                                                                   [19,22].     andmain
                                                                                                                                     hepatocytes
                                                                                                                                           mechanism
                                   [8,9].
                                   is     The lower
                                      associated      level
                                                    with     of inhibition
                                                           the  cholesterolof  in the
                                                                                  the liver
                                                                                       NPC1L1cells protein,
                                                                                                   leads them   thetokey
                                                                                                                      absorb    more
                                                                                                                           factor  of cholesterol
                                                                                                                                       cholesterol ab-
                                   from circulation
                                   sorption   (Figure resulting
                                                        1) locatedinondecrease     blood cholesterol
                                                                         the gastrointestinal      tractlevel    [23]. cells and hepatocytes [8,9].
                                                                                                          epithelial
                                         Ezetimibe
                                   The lower    levelwas   originally developed
                                                       of cholesterol     in the liverascells
                                                                                          a potent
                                                                                               leads inhibitor
                                                                                                        them toofabsorb
                                                                                                                     acyl-coenzyme       A choles-from
                                                                                                                             more cholesterol
                                   terol acyltransferase
                                   circulation   resulting toin block   spontaneous
                                                                 decrease                cholesterol
                                                                              blood cholesterol          efflux,
                                                                                                     level   [23].which may be responsible
                                   for cholesterol
                                          Ezetimibeesterification
                                                      was originally  in macrophages
                                                                           developed as[24]     and results
                                                                                             a potent            in formation
                                                                                                         inhibitor                of intracellular
                                                                                                                      of acyl-coenzyme        A choles-
                                   cholesteryl   ester, and   foam   cells  [25]. Later  it was  described      as  an inhibitor
                                   terol acyltransferase to block spontaneous cholesterol efflux, which may be responsible         of  cholesterol
                                   uptake
                                   for      from theesterification
                                       cholesterol     small intestine  inby   binding to the
                                                                            macrophages         NPC1L1
                                                                                              [24]            transporter
                                                                                                    and results              [26]. of intracellular
                                                                                                                     in formation
                                         Major   indication   of  ezetimibe    is the elevation    of  total  cholesterol,
                                   cholesteryl ester, and foam cells [25]. Later it was described as an inhibitor             LDL andofapolipo-
                                                                                                                                            cholesterol
                                   uptake from the small intestine by binding to the NPC1L1 transporterand
                                   protein   B (ApoB)    levels   [19]. Ezetimibe     also  blocks    aminopeptidase        N        interrupts a
                                                                                                                                 [26].
                                   caveolin   1–annexin
                                          Major  indication A2ofcomplex
                                                                  ezetimibe  involved   in cholesterol
                                                                                is the elevation    of totaltransport    [23].LDL
                                                                                                               cholesterol,     Latestandliterature
                                                                                                                                            apolipopro-
                                   tein B (ApoB) levels [19]. Ezetimibe also blocks aminopeptidase N and interruptsclearly
                                   data  suggest   that ezetimibe    may    exert  several  extra-intestinal      effects, but  these   are  a caveolin
                                   1–annexin A2 complex involved in cholesterol transport [23]. Latest literature data suggest
                                   that ezetimibe may exert several extra-intestinal effects, but these are clearly limited to the
                                   inhibition of macrophage migration, reduction of ROS levels and plaque size [8].
Biomedicines 2022, 10, 1090                                                                                            4 of 18

                                    Ezetimibe belongs to the class II of antihyperlipidemic drugs and is recommended in
                              second-line therapy for coronary artery diseases prevention after statins [9,22]. Numerous
                              clinical trials showed that ezetimibe could be used either as monotherapy or in combination
                              with statins. It has been used as the first option for patients who are unable to tolerate
                              statins or suffer from their side effects [22,27,28]. The recommended daily dose of ezetimibe
                              10 mg/day orally is well-tolerated either as monotherapy or in combination with statins or
                              fenofibrates [29].
                                    To date, there is little documented evidence of serious side effects of ezetimibe. Due to
                              insufficient data, ezetimibe is contraindicated and is not recommended for patients with
                              moderate to severe hepatic impairment. However, in monotherapy trials, liver function
                              tests were found to be similar like placebo [28,30]. Studies of Slim and Thompson (2008) and
                              Havranek et al. (2006) have documented the coherence between ezetimibe and myopathy,
                              the second one even pointed to the same effect of ezetimibe and statins in relation to
                              myopathy [31,32].

                              2.3. PCSK9 Inhibitors
                                    PCSK9, a secretory protease, is a member of the proprotein convertase family. PCSK9
                              is initially secreted as an inactive enzyme precursor which undergoes autocatalytic cleavage
                              in the endoplasmic reticulum of hepatic cells for activation. It moves out of the endoplasmic
                              reticulum to be further handled by the Golgi apparatus before entering the circulation.
                              PCSK9 is secreted into plasma by hepatocytes and has the ability to degrade LDL receptors,
                              inhibiting recycling of receptors to the cell surface (Figure 1). This process thus inhibits
                              uptake of plasma LDL [33,34]. The expression of PCSK9 is induced by sterol regulatory
                              element-binding protein 2 (SREBP-2), leading to LDL-receptor degradation [35]. As result,
                              PCSK9 has become an important cholesterol reduction target. PCSK9 inhibitors inhibit the
                              binding of PCSK9 with LDL-receptors and prevent the degradation of LDL-receptors [36].
                                    There are several strategies that target the inhibition of PCSK9 with different mech-
                              anisms such as monoclonal antibodies, synthetic small interfering RNA (siRNA) against
                              PCSK9, vaccination, and small molecules. From human monoclonal antibodies, evolocumab
                              and alirocumab were approved by FDA in 2015 and are currently being marketed. In addi-
                              tion to their ability to decrease LDL-c levels, they may affect the lipid profile by increasing
                              HDL, reducing total cholesterol, and lipoprotein A levels, thus lowering plaque volume.
                              Bococizumab, the third of monoclonal antibodies has been withdrawn by Pfizer while
                              under the evaluation of phase III clinical trials. Although it was able to decrease LDL-c
                              level to 54% [37], after 12-month follow up, it did not demonstrate any benefits regarding
                              the primary end point of major adverse cardiovascular events [38].
                                    Alirocumab is available as a 75 mg/mL pre-filled pen or syringe and is administered
                              every two weeks by subcutaneous injection at a dose of 75–150 mg [39]. Evolocumab is avail-
                              able as a 140 mg/mL single use prefilled syringe or as an autoinjector activated every two
                              weeks. The monthly dose of evolocumab is more than a double of the dose of two-weekly
                              injections because the drug has non-linear pharmacokinetics. Its plasma concentrations
                              do not increase in proportion to the administered dose [39,40]. No adverse effects were
                              reported concerning monoclonal antibodies except injection-site reaction [41] which is
                              related to immunogenicity [42]. It is already accepted that PCSK9 monoclonal antibodies
                              which are already approved have the highest effectiveness comparing long-term statin
                              therapy or statin-ezetimibe combination or ezetimibe alone therapy regimen [27,43,44].
                              According to the recent studies, monoclonal antibodies have the potential to be used as
                              an alternative to statins [43,45]. However, subcutaneously drug administration, high cost,
                              inaccessibility, and limited long-term clinical outcomes are major obstacles to their wider
                              clinical use [44,46,47].
Biomedicines  2022, 10,
 Biomedicines2022,  10, 1090
                        x FOR PEER REVIEW                                                                                              5 of 185 of 18

                                   3.3.Targeted
                                        TargetedTherapies
                                                 Therapies
                                    3.1.Statin-Loaded
                                   3.1.  Statin-Loaded Nano-Based
                                                      Nano-Based   Drug
                                                                 Drug   Delivery
                                                                      Delivery    System
                                                                               System
                                         According
                                          Accordingtotodifferent
                                                             different human
                                                                         human  studies,    long-term
                                                                                     studies,           statin
                                                                                                long-term       treatment
                                                                                                             statin          oftenoften
                                                                                                                     treatment      causes  sev- sev-
                                                                                                                                          causes
                                   eral adverse effects described shortly in the Part 2 [12,19]. In clinical practice, in such cases,
                                    eral  adverse    effects   described     shortly     in  the Part  2 [12,19].    In  clinical  practice,  in such
                                   statin doses are reduced, combined with ezetimibe or PCSK9 inhibitors, and/or vitamins
                                    cases,   statin  doses    are  reduced,    combined        with  ezetimibe     or PCSK9     inhibitors,   and/or
                                   or coenzyme Q10 are added to the treatment regimen respectively. In addition, there are
                                    vitamins    or  coenzyme      Q10    are added      to the  treatment   regimen      respectively.   In addition,
                                   several
                                    there arestatin-intolerant   patients whopatients
                                                several statin-intolerant         are unable  whoto are
                                                                                                    tolerate statins
                                                                                                        unable        at any dose.
                                                                                                                  to tolerate         Consider-
                                                                                                                                statins  at any dose.
                                   ing  its low water
                                    Considering           solubility,
                                                     its low    water rapid     metabolism,
                                                                        solubility,              low bioavailability,
                                                                                       rapid metabolism,                  and several clinical
                                                                                                               low bioavailability,      and several
                                   complications     of statins, it
                                    clinical complications        ofisstatins,
                                                                       desirable
                                                                               it istodesirable
                                                                                       improve the    therapeutic
                                                                                                   to improve     theefficacy  of the efficacy
                                                                                                                       therapeutic    drug andof the
                                   reduce
                                    drug andits side effects
                                                 reduce    its by developing
                                                               side   effects bydifferent
                                                                                  developing  therapeutic  approaches,
                                                                                                   different  therapeutic   such  as nanomed-
                                                                                                                               approaches,    such as
                                   icine  options [48–50].
                                    nanomedicine        optionsToday,   several
                                                                   [48–50].       nano-formulations,
                                                                               Today,                     including polymeric
                                                                                          several nano-formulations,                 nanoparti-
                                                                                                                              including    polymeric
                                   cles, lipid-based lipid-based
                                    nanoparticles,      nanoparticles,    chitosan-basedchitosan-based
                                                                        nanoparticles,         nanoparticles, nanoliposomes,        nanoemul-
                                                                                                                 nanoparticles, nanoliposomes,
                                   sions,  nanotransfersomal
                                    nanoemulsions,                 carriers, self-nanoemulsifying
                                                          nanotransfersomal                              systems, and cerium
                                                                                      carriers, self-nanoemulsifying               oxideand
                                                                                                                             systems,     nano-cerium
                                   particles   have been formulated
                                    oxide nanoparticles        have beentoformulated
                                                                              increase thetobioavailability      and therapeuticand
                                                                                                 increase the bioavailability        efficacy  of
                                                                                                                                         therapeutic
                                   statins
                                    efficacy(Figure  2). (Figure 2).
                                               of statins

                                    Figure 2. Proposed statin-loaded nanoparticle systems in targeted lipid-lowering therapy.
                                   Figure 2. Proposed statin-loaded nanoparticle systems in targeted lipid-lowering therapy.
                                    3.1.1. Polymeric Nanoparticles
                                   Polymeric Nanoparticles
                                          Among biodegradable polymeric nanoparticles, poly(lactic-co-glycolic acid) (PLGA)
                                    whichAmong      biodegradable
                                              is approved    by FDApolymeric     nanoparticles,
                                                                       (for review                poly(lactic-co-glycolic
                                                                                      see [51]) was   investigated usingacid)    (PLGA)
                                                                                                                            different   statins.
                                   which    is approved    by FDA   (for  review  see  [51]) was  investigated
                                    Statin-loaded PLGA nanoparticles displayed a superior profile concerning theusing  different  statins.
                                                                                                                                      bioavail-
                                   Statin-loaded     PLGA nanoparticles
                                    ability, drug release,    dosing, and displayed
                                                                             minimizing  a superior
                                                                                            adverse profile
                                                                                                     effects concerning   the hyperlipidemic
                                                                                                             [52,53]. In the  bioavaila-
                                   bility, drug release, dosing, and minimizing adverse effects [52,53]. In the hyperlipidemic
                                    rat model, administration of atorvastatin-loaded PLGA nanoparticles every 3 days exhib-
                                   rat model, administration of atorvastatin-loaded PLGA nanoparticles every 3 days exhib-
                                    ited the same efficacy as the once-daily treatment of Lipicure- commercial formulation of
                                   ited the same efficacy as the once-daily treatment of Lipicure- commercial formulation of
                                    atorvastatin calcium. As a result, the daily dose of atorvastatin was reduced by 66% with
                                   atorvastatin calcium. As a result, the daily dose of atorvastatin was reduced by 66% with
                                    PLGA formulation [52]. Moreover, pitavastatin-loaded PLGA nanoparticles attenuated the
                                   PLGA formulation [52]. Moreover, pitavastatin-loaded PLGA nanoparticles attenuated
                                    increase of inflammatory inducible nitric oxide synthase (iNOS) activity [54]. Apart from
                                   the increase of inflammatory inducible nitric oxide synthase (iNOS) activity [54]. Apart
                                    their anti-inflammatory effect, statin-loaded PLGA nanoparticles reduce the progression of
                                   from their anti-inflammatory effect, statin-loaded PLGA nanoparticles reduce the pro-
                                    hypertension and proliferation of pulmonary smooth muscle cells [55], while exhibiting
                                   gression of hypertension and proliferation of pulmonary smooth muscle cells [55], while
                                    cardioprotective properties without any adverse effects in different animal models of car-
                                   exhibiting cardioprotective properties without any adverse effects in different animal
                                    diovascular
                                   models           diseases [56,57].
                                              of cardiovascular         Similarly,
                                                                  diseases         acid-polycaprolactone-based
                                                                             [56,57].                                delivery of simvastatin
                                                                                      Similarly, acid-polycaprolactone-based        deliv-
                                    was   able   to markedly    reduce   the  chemotaxis     of vascular smooth
                                   ery of simvastatin was able to markedly reduce the chemotaxis of vascular smoothmuscle   cells muscle
                                                                                                                                  and intimal
                                    hyperplasia
                                   cells and intimal [58].hyperplasia
                                                           Pitavastatin-loaded    PLGA nanoparticles
                                                                       [58]. Pitavastatin-loaded    PLGA were     able to repair
                                                                                                           nanoparticles           injured
                                                                                                                           were able    to vas-
                                    culature    via the activation  of  PI3K  signaling   pathway   promoting    the re-reendothelialization
                                   repair injured vasculature via the activation of PI3K signaling pathway promoting the re-
                                    and reducing intimal
                                   reendothelialization      andhyperplasia    [59]. The
                                                                 reducing intimal         increased[59].
                                                                                      hyperplasia     phosphorylation     of Akt [59,60] and
                                                                                                         The increased phosphoryla-
                                   tion of Akt [59,60] and eNOS upregulation [54,61] have been mainly responsible for
                                    eNOS     upregulation     [54,61] have    been  mainly    responsible  for these  effects.      these
                                                                                                                                In agreement
                                   effects. In agreement with these studies, combined therapy with simvastatin- and coen- poly-
                                    with   these   studies,  combined     therapy   with  simvastatin-   and  coenzyme     Q10-loaded
                                   zyme
                                    meric Q10-loaded
                                             nanoparticles  polymeric
                                                               enhanced  nanoparticles
                                                                            PI3K-Akt-eNOS  enhanced    PI3K-Akt-eNOS
                                                                                                 pathway    by increasingpathway       by
                                                                                                                             the expressions
                                    of Akt and eNOS both in the heart and aorta of obese Zucker rats [62]. Rosuvastatin-
                                    loaded poly(L-lactide-co-caprolactone) nanoparticles increased the reendothelialization
                                    and reduced the thrombotic potential via increased vascular endothelial growth factor
Biomedicines 2022, 10, 1090                                                                                           6 of 18

                              signaling [63]. Later, atorvastatin calcium-loaded poly(ε-caprolactone) nanoparticles were
                              developed and their sustained drug release, improved efficacy, better drug bioavailability,
                              and reduced adverse effects comparing pure atorvastatin have also been confirmed [64,65].
                              In high-fat diet induced rats, atorvastatin calcium nanoparticles enhanced the NO pro-
                              duction and decreased the lipid peroxidation in the liver while simultaneously decreasing
                              the interleukin-1 beta and interleukin-6 levels [66]. Recently, cellulose-based polymer
                              nanoparticles have shown a 3.5-fold enhancement in drug bioavailability. After successful
                              preclinical studies, atorvastatin-loaded ethyl cellulose nanoparticles are considered as a
                              candidate for further clinical trials [49,67].

                              3.1.2. Chitosan Nanoparticles
                                   Chitosan is known as a biopolymer that coats liposomes to enhance their stability
                              and leads to controlled release of drugs. It increases the nanoformulation efficiency and
                              accessibility of loaded-drug [68]. Chitosan itself may lower cholesterol which has been
                              confirmed in both experimental animal and human studies [69]. Atorvastatin calcium-
                              loaded chitosan nanoparticle formulation showed sustained drug release up to 7 days [70].
                              In an experimental animal study, rosuvastatin-chitosan nanoparticles were found to be
                              more effective in improving the lipid profile than the pure drug [71]. Recently, novel
                              oral chitosan-based atorvastatin nanocrystals formulation with improved bioavailability
                              have been successfully developed and anti-hyperlipidemic activity has been found to
                              be higher than in marketed Lipitor [72]. In an alternative approach, long-circulating
                              polyethylene glycol (PEG) chitosan nanoparticles showed more than a 72-h drug release
                              profile. Moreover, pharmacodynamic parameters of this formulation were superior to the
                              pure drug [73].

                              3.1.3. Cerium Oxide Nanoparticles
                                   Cerium oxide nanoparticles have the capacity to eliminate ROS and actually are
                              known as a ROS scavenger. This particular system could accumulate in kidneys and tar-
                              get mitochondria to eliminate excessive ROS [74]. Moreover, cerium oxide nanoparticles
                              interfered with the adipogenic pathway and hindered the accumulation of triglycerides.
                              Transcriptional analysis following in vivo treatment revealed a down-regulation of Lep,
                              Bmp2, Twist1, Angpt2, and Ddit3, and an up-regulation of Irs1 and Klf4 expression. Overall,
                              cerium oxide nanoparticles contributed to a slowing of weight gain and lowered the plasma
                              levels of insulin, leptin, glucose, and triglycerides [75]. In this sense, ceria nanoparticles
                              coated with ROS-responsive organic polymer (mPEG-TK-PLGA) and loaded with atorvas-
                              tatin demonstrated a greater antioxidant and anti-apoptotic activity than the pure drug,
                              and effectively decreased oxidative stress and inflammatory processes in acute kidney
                              injury mice model [74].

                              3.1.4. Lipid-Base Nanoparticles
                                    Among the lipid-based nanoparticle formulations, solid lipid nanoparticles (SLN)
                              and nanostructured lipid carrier (NLC) are well-studied promising delivery systems [76].
                              Comparing free statins, oral bioavailability and absorption of statin-loaded SLNs were
                              markedly enhanced [77–79]. A sustained release of simvastatin from the lipid core of
                              nanoparticles was confirmed as well [80]. In high-fat diet induced hyperlipidemic rats,
                              the treatment of atorvastatin-loaded SLNs combined with coenzyme Q10 and vitamin E
                              showed superior effect on lipid profile. Atorvastatin-loaded SLNs alone administration
                              illustrated weaker effect in reducing triglycerides and LDL levels than the combined
                              therapy with coenzyme Q10 and vitamin E [81].
                                    Since the NLCs formulation has produced and evolved from SLNs, several studies
                              compared the effects of drug profile between SLNs and NLCs. According to the results,
                              statin-loaded NLCs exhibited superior pharmaco-technical properties in regards to sus-
                              tained and gradual drug release than SLNs [76,82]. Moreover, displaying greater results in
                              lowering total cholesterol, LDL, triglycerides, and elevating HDL was observed in compari-
Biomedicines 2022, 10, 1090                                                                                           7 of 18

                              son with the standard drug [82]. 5-weeks-of-simvastatin-loaded NLCs treatment exhibited
                              the recovery of blood lipid levels, inhibition of smooth muscle cell apoptosis, and the delay
                              in the onset of atherosclerosis in hyperlipidemic rats [83]. Statin-loaded NLCs formulations
                              also exerted prolonged reduction in the total cholesterol and non-high-density lipoprotein
                              cholesterol levels [84]. The pharmacodynamic and pharmacokinetic profile of NLCs with
                              statins has been tested in several studies confirming that this treatment approach could
                              be used to improve the statin oral delivery and bioavailability, clinical efficacy, and anti-
                              hyperlipidemic activity [84–87].
                                   Statin-loaded HDL nanoparticles showed the inhibition of inflammation progression
                              and a decrease of atherosclerotic plaques while simultaneously achieving these effects
                              without any hepatotoxicity. Injectable reconstituted HDL nanoparticle carrier vehicle was
                              able to deliver statin directly to the atherosclerotic plaques [88,89].

                              3.1.5. Nanoliposomes, Nanoemulsions, Nanotransfersomal Carriers
                                    In high-fat diet rats, simvastatin-loaded lipid core nanocapsules exhibited greater
                              efficiency than same lovastatin-loaded nanocapsules [90]. In studies with novel nano-
                              formulations indicated that simvastatin-loaded nanoliposomes exerted a higher plasma sim-
                              vastatin concentration than the pure drug [91] and nanoemulsion of simvastatin promised
                              higher oral bioavailability with almost 100% of drug release [92].
                                    Chen et al. suggested that the lipophilic emulsifier is crucial for the oral absorption
                              of the drug. In their study, the lipophilic emulsifier named Myverol and soybean phos-
                              phatidylcholine were used and successfully formulated lovastatin-loaded NLCs. According
                              to the results, lovastatin administration of Myverol-containing NLCs were found to be
                              more stable in the gastric environment compared to soybean phosphatidylcholine. When
                              compared with the free drug, lovastatin-loaded NLCs from Myverol exhibited greater
                              plasma concentrations [85].
                                    In another unique study, the effect of transdermal delivery system was investigated in
                              hyperlipidemic rats. The developed nanotransfersomal carrier exerted potent effects on
                              hyperlipidemia without serious side effects on liver in comparison to an oral atorvastatin
                              treatment. Interestingly, in hyperlipidemic rats, pure atorvastatin had no effect on lipid
                              profile. On the other hand, nanotransfersomal atorvastatin decreased significantly total
                              cholesterol, triglycerides, and LDL-c levels. This pioneer study showed for the first-time
                              amelioration of dyslipidemia by the treatment with transdermal atorvastatin nanotransfer-
                              somal gel system. Transfersomes may open a window of opportunity for the well-controlled
                              transdermal delivery of drugs that produce side effects following oral administration [93].

                              3.1.6. Self-Nanoemulsifying Drug Delivery System
                                    Self-nanoemulsifying drug delivery system (SNEDDS) of statins could be a novel
                              formulation to enhance drug profile. SNEDDS of statins improved the drug dissolution
                              rate [94], increased the oral bioavailability approximately 2.4-fold [94,95] and the expansion
                              of drug release 4-fold [96] compared to the pure statins. Statin-loaded SNEDDS system has
                              exerted remarkable antihyperlipidemic properties by normalizing serum lipid levels [97]
                              and overall, it has a valuable potential to improve the oral absorption as well as the
                              pharmacodynamic efficacy compared to the pure drug [98,99].
                                    According to the previous findings, it is obvious that statin nano-therapy using differ-
                              ent nanotechnology systems may contribute to the reduction or elimination of common
                              adverse effects related to the statin treatment and helps statin delivery as well as their
                              positive pleiotropic effects.

                              3.2. Ezetimibe-Loaded Nano-Based Drug Delivery Systems
                                   According to the Biopharmaceutics Classification System, ezetimibe belongs to the
                              class II/IV of compounds which display a low aqueous solubility and absorption and high
                              permeability resulting in poor bioavailability [100]. In the literature, many approaches can
                              be found concerning upgrade ezetimibe drug profile (Figure 3).
3.2. Ezetimibe-Loaded Nano-Based Drug Delivery Systems

                                   According to the Biopharmaceutics Classification System, ezetimibe belongs to the
                              class II/IV of compounds which display a low aqueous solubility and absorption and high
Biomedicines 2022, 10, 1090
                              permeability resulting in poor bioavailability [100]. In the literature, many approaches can
                                                                                                                        8 of 18
                              be found concerning upgrade ezetimibe drug profile (Figure 3).

                              Figure 3. Proposed
                                 Figure          ezetimibe-loaded
                                         3. Proposed               nanoparticle
                                                     ezetimibe-loaded           systems
                                                                      nanoparticle      in targeted
                                                                                   systems           lipid-lowering
                                                                                            in targeted              therapy.
                                                                                                         lipid-lowering therapy.

                                   3.2.1. Lipid Carrier Systems
                              Lipid Carrier Systems
                                         Lipid carrier systems have been documented as suitable delivery systems to increase
                                     Lipid carrier systems have been documented as suitable delivery systems to increase
                                   the bioavailability of ezetimibe. Comparing to marketed product and drug suspension,
                              the bioavailability of ezetimibe. Comparing to marketed product and drug suspension,
                                   ezetimibe-loaded SLNs showed greater stability. According to the results, stability of
                              ezetimibe-loaded SLNs showed greater stability. According to the results, stability of
                                   ezetimibe-loaded SLNs remained unchanged for 3 months. However, slow and limited
                              ezetimibe-loaded SLNs remained unchanged for 3 months. However, slow and limited
                                   ezetimibe release lead to decreasing its bioavailability [101]. On the contrary, in high-fat
                              ezetimibe release lead to decreasing its bioavailability [101]. On the contrary, in high-fat
                                   diet rats, ezetimibe-loaded NLCs increased drug bioavailability even with reduced dose
                              diet rats, ezetimibe-loaded NLCs increased drug bioavailability even with reduced dose
                                   compared to the pure drug. Simultaneously, triglyceride, HDL-c, LDL-c and cholesterol
                              compared       to the
                                   levels were       pure drug.and
                                                  normalized       Simultaneously,
                                                                       liver functionstriglyceride,
                                                                                        were improved. HDL-c,    LDL-c and cholesterol
                                                                                                              Ezetimibe-loaded      SLNs are
                              levels    were   normalized     and  liver  functions
                                   ready today for clinical evaluation [102].        were   improved.      Ezetimibe-loaded      SLNs are
                              ready today for clinical evaluation [102].
                                   3.2.2. Green-Based Nanocarriers
                              Green-Based Nanocarriers
                                         Nowadays, the attention has been drawn to biosustainable and biodegradable systems
                                     Nowadays,
                                   like  green-based the nanocarriers.
                                                          attention has Itbeen
                                                                             hasdrawn   to biosustainable
                                                                                 been documented               and biodegradable
                                                                                                        that biodegradable             sys-
                                                                                                                                nanoparticle
                              tems    like green-based
                                   adjusted                 nanocarriers.
                                               drug therapeutic       valueItby
                                                                              has been documented
                                                                                increasing                that biodegradable
                                                                                             its bioavailability,    solubility,nanopar-
                                                                                                                                  and release
                              ticletime
                                     adjusted    drug therapeutic
                                         [103]. Using                  valuealternative
                                                         linseed mucilage     by increasing
                                                                                         in theitsdevelopment
                                                                                                   bioavailability,   solubility,nanoparticle
                                                                                                                 of polymeric      and re-
                              lease   time [103]. pharmaceutical
                                   formulation,     Using linseed mucilage       alternative
                                                                         properties            in theofdevelopment
                                                                                     and efficacy        ezetimibe were of polymeric
                                                                                                                            enhanced.na-  This
                              noparticle     formulation,     pharmaceutical     properties     and   efficacy  of  ezetimibe
                                   study concludes that linseed mucilage seems to be a resourceful alternate for the delivery    were   en-
                              hanced.     This study
                                   of therapeutic      concludes
                                                      agents  with that
                                                                     poorlinseed  mucilage [104].
                                                                           water solubility   seems to be a resourceful alternate for
                              the delivery of therapeutic agents with poor water solubility [104].
                                   3.2.3. Nanoemulsions, Nanosuspensions, Nanocrystals
                              Nanoemulsions,      Nanosuspensions,
                                         Also, nanoemulsions       andNanocrystals
                                                                         nanosuspensions of ezetimibe illustrated better drug absorp-
                                     Also, nanoemulsions and nanosuspensions
                                   tion  than   the marketed    formulation     and belong of to
                                                                                               ezetimibe
                                                                                                  possible illustrated
                                                                                                            alternativesbetter   drug ab-the
                                                                                                                           in improving
                              sorption than the marketed formulation and belong to possible alternatives in improving
                                   drug   profile  [105,106].  Ezetimibe    nanosuspensions      completely    dissolved   in the dissolution
                              the medium
                                    drug profile    [105,106].
                                               within           Ezetimibe
                                                        1 h, while  pure drugnanosuspensions
                                                                                 was dissolved completely
                                                                                                   only up to 42%dissolved
                                                                                                                      duringinthe
                                                                                                                                thesame
                                                                                                                                    disso-
                                                                                                                                         time.
                              lution medium within 1 h, while pure drug was dissolved only up to 42% during the sameThe
                                   Better  drug   dissolution    resulted   in improved    oral   bioavailability  of  ezetimibe   [107].
                              time.drug   release
                                       Better  drugprofile  of nano-ezetimibe
                                                      dissolution                from volatile
                                                                    resulted in improved     oralmicroemulsion
                                                                                                    bioavailabilitytemplate    was increased
                                                                                                                      of ezetimibe   [107].
                                   significantly >98% in 30 min [108].
                                         Similarly, nanocrystals of ezetimibe remarkably increased its solubility and displayed
                                   a superior pharmacodynamic activity upon oral administration. Solubility, dissolution,
                                   and pharmacodynamics in lipid lowering activity using ezetimibe nanocrystals were
                                  preferable [109]. Dual drug nanocrystals loaded nano-embedded microparticles with a
                                   combination of simvastatin and ezetimibe were also studied. Nanocrystals of both drugs
                                   demonstrated a significant enhancement of dissolution in comparison to their physical
                                   mixture [110].
Biomedicines 2022, 10, 1090                                                                                          9 of 18

                              3.2.4. Self-Nanoemulsifying Drug Delivery
                                   It has been proven that self-nanoemulsifying granule system of ezetimibe ensured
                              128% protection while pure ezetimibe can offer only 58%. In high-fat diet rats, this formula-
                              tion yielded a higher reduction in the total cholesterol levels due to enhancement of drug
                              solubility and absorption. On the other hand, there was no significant difference in the
                              HDL levels after 14-days of treatment, by reason it had no direct action on the blood HDL
                              levels like statins [111]. Great potential of optimized super saturable SNEDDS (S-SNEDDS)
                              has been reported for the first time by Gao and Morozowich (2006) [112]. Later, Bandy-
                              opadhyay and Singh (2012) investigated the impact of the same formulation and presented
                              the enhancement of the drug absorption [113].

                              3.2.5. Solid Dispersion Nanoparticle Formulation
                                    Solid dispersion nanoparticle formulation is another approach to find out an effective
                              oral drug delivery system. Ezetimibe-hydroxypropyl cellulose (HPC) solid dispersion
                              nanoparticle system enhanced absorption and low dissolution rate of pure ezetimibe with
                              about 7.5% increasement in maximum dissolution and 9.9% increasement of dissolution
                              efficiency [114]. Torrado-Salmeron et al. described ezetimibe self-micellizing solid dis-
                              persion with a non-ionic emulsifier and solubilizer called Kolliphor. Results showed that
                              serum levels of total cholesterol, LDL, and triglycerides were significantly improved when
                              compared to pure ezetimibe in high-fat induced hyperlipidemic rats [115]. The same group
                              of authors also investigated the comparison between solid dispersion of ezetimibe and
                              micellar system of ezetimibe with Kolliphor. The second one showed greater solubility and
                              better oral bioavailability accompanied by an improvement of the lipid profile with half
                              dose of ezetimibe and using different formulation of micellar system [116].
                                    A comprehensive study that examined the differences between ezetimibe-loaded
                              different formulation approaches such as SNEDDS, surface modified solid dispersion
                              (SMSD), and solvent evaporated solid dispersion (SESD) revealed higher total ezetimibe
                              plasma concentrations and aqueous solubility as compared with the pure ezetimibe. All
                              formulations significantly ameliorated dissolution of the drug in the following order:
                              SNEDDS ≥ SESD > SMSD (200-fold for SNEDDS, 110-fold for SESD, and 80-fold for
                              SMSD). Because SESD gave higher area under curve from zero to infinity than did SNEDDS
                              and SMSD, this system was mostly recommended as a drug delivery system for the oral
                              administration of ezetimibe [117].
                                    Statin combination therapy with ezetimibe may significantly reduce the incidence
                              of severe atherosclerotic events [118]. Indeed, combination of ezetimibe and atorvastatin
                              loaded nano-solid dispersions improved efficiency, serum lipid levels, and reduced the
                              toxic effects of cholesterol on the liver tissue [119,120]. Interestingly, atorvastatin even
                              enhanced solubility of ezetimibe and its dissolution profile. On the other hand, atorvastatin
                              release was decreased in the presence of ezetimibe [120]. Micellar system of ezetimibe and
                              atorvastatin combination therapy using nano solid dispersions with Kolliphor showed
                              significantly greater impact on lipid profile than the same dose of pure drugs [121].

                              3.3. PCSK9 Inhibition Targeting
                                  With the exception of monoclonal antibodies, the inhibition of PCSK9 is targeting via
                              siRNA against PCSK9 synthesis and expression, small molecules, and vaccination against
                              PCSK9 (Figure 4). While siRNA is in the phase III clinical stage, the last two brand-new
                              approaches are still in the preclinical study stage [122–124].
With the exception of monoclonal antibodies, the inhibition of PCSK9 is targeting via
                              siRNA against PCSK9 synthesis and expression, small molecules, and vaccination against
                              PCSK9 (Figure 4). While siRNA is in the phase III clinical stage, the last two brand-new
Biomedicines 2022, 10, 1090
                              approaches are still in the preclinical study stage [122–124].                       10 of 18

                               Figure 4. Proposed PCSK9 inhibition targeting in lipid-lowering therapy.
                              Figure 4. Proposed PCSK9 inhibition targeting in lipid-lowering therapy.
                                3.3.1. siRNA: Inclisiran
                              siRNA: Inclisiran
                                       Inclisiran is the first siRNA-based drug that acts as an inhibitor of PCSK9 expression.
                                It specificallyistargets
                                     Inclisiran        the firstandsiRNA-based
                                                                         binds to PCSK9 drug that     acts asleading
                                                                                                   mRNA            an inhibitor
                                                                                                                              to PCSK9of PCSK9        expression.
                                                                                                                                               degradation          and
                              Itresulting
                                 specifically     targets     and    binds   to   PCSK9       mRNA       leading
                                             in diminished PCSK9 protein levels and long-lasting reduction of LDL-c     to  PCSK9       degradation          andeven
                                                                                                                                                                   re-
                              sulting
                                up to 3–6in diminished
                                              months [125–127].PCSK9 protein          levelsbiological
                                                                          It has a long         and long-lasting
                                                                                                             half-life that reduction
                                                                                                                                   induced  of LDL-c
                                                                                                                                                 sustained even    up
                                                                                                                                                                LDL-c
                              tolowering,
                                  3–6 months        [125–127].       It  has a  long     biological      half-life
                                              which seems to be more preferable than monoclonal antibody therapy [128]. that    induced      sustained        LDL-c
                              lowering,     whichefficacy,
                                       Inclisiran      seems to      be more and
                                                                  toleration,    preferable       than
                                                                                        its ability   to monoclonal
                                                                                                          reduce LDL-cantibody   levels have  therapy      [128].
                                                                                                                                                   been displayed
                                     Inclisiran efficacy,
                                by preclinical       studies, toleration,
                                                                  Phase I andand    Phase its ability    to reduce
                                                                                               II clinical    trials. PhaseLDL-cIII   levels    have
                                                                                                                                         clinical        been
                                                                                                                                                     trials   aredis-
                                                                                                                                                                    still
                              played    by preclinical
                                in progress      [126,129].   studies,    Phase to
                                                                  According        I and
                                                                                       thePhase
                                                                                             ORION-1 II clinical      trials. Phase
                                                                                                             randomized                   III clinical
                                                                                                                                   clinical     trial, onetrials
                                                                                                                                                               or are
                                                                                                                                                                    two
                              still in progress
                                injections           [126,129].
                                              of inclisiran      canAccording
                                                                       remarkably   to the    ORION-1
                                                                                         reduce    PCSK9randomized
                                                                                                               and LDL, decrease   clinicalatherogenic
                                                                                                                                               trial, one orlipids,
                                                                                                                                                                 two
                              injections    of  inclisiran      can    remarkably        reduce     PCSK9        and     LDL,
                                improve lipoprotein profiles and appear to be safe and well tolerated [130]. Patients with        decrease       atherogenic         li- a
                              pids,
                                highimprove        lipoprotein
                                       cardiovascular                 profiles and
                                                              risk received      one appear        to be safe
                                                                                        or two doses                and wellfor
                                                                                                            of inclisiran          tolerated
                                                                                                                                      6 months.   [130].
                                                                                                                                                      TheirPatients
                                                                                                                                                                results
                              with   a highthat
                                showed        cardiovascular
                                                    PCSK9 and risk     LDL-creceived
                                                                                 levelsone  were or markedly
                                                                                                    two doses reduced of inclisiran  from forthe
                                                                                                                                               6 months.
                                                                                                                                                    baselineTheir [131].
                              results   showed that PCSK9 patients
                                After one-year-follow-up,                and LDL-c  who   levels   weretwo-dose-inclisiran
                                                                                             received       markedly reduced have           from still
                                                                                                                                                     the remained
                                                                                                                                                           baseline
                              [131].   After one-year-follow-up,
                                the reductions         on the PCSK9 and       patients
                                                                                    LDL-c    who    received
                                                                                                levels.             two-dose-inclisiran
                                                                                                            According          to the results,have    twice  still  re-
                                                                                                                                                                a year
                              mained      the reductions
                                administration                   on thecould
                                                       of inclisiran       PCSK9      and LDL-c
                                                                                  maintain              levels.
                                                                                                 to stable          According
                                                                                                               reductions             to the results,
                                                                                                                                  in LDL-c                   twice a
                                                                                                                                                 levels [132].
                              year administration
                                       Novartis recently    of inclisiran    couldinclisiran
                                                                    introduced         maintain to   and  stable     reductions
                                                                                                              received        its firstin LDL-c
                                                                                                                                            approval levels    [132].
                                                                                                                                                            from     the
                                     Novartis
                                European          recently
                                               Union            introduced
                                                           in 2020              inclisiran
                                                                        for patients       with and   received
                                                                                                   primary            its first approval fromand
                                                                                                                  hypercholesterolemia                   the dyslipi-
                                                                                                                                                               Euro-
                              pean
                                demia Union     in 2020
                                          [128,129].          for patients
                                                          It could     be usedwithwithprimary
                                                                                           either a hypercholesterolemia
                                                                                                       statin or a statin-ezetimibe         and dyslipidemia
                                                                                                                                                    [129]. Among
                              [128,129].    It couldinjection
                                its side effects,         be used sitewithreactions,
                                                                             either a statin       or asite
                                                                                            injection     statin-ezetimibe
                                                                                                                 pain, erythema       [129].
                                                                                                                                           andAmong
                                                                                                                                                  rash have  its side
                                                                                                                                                                   been
                              effects,
                                reported.injection
                                              Othersite  thanreactions,    injection
                                                                these possible            site pain,
                                                                                     adverse            erythema
                                                                                                  effects,   such asand          rash haverisk
                                                                                                                           an increased          been ofreported.
                                                                                                                                                          cardiovas-
                              Other
                                cular than
                                        eventsthese
                                                 or organ possible     adverse
                                                                toxicity,          effects,
                                                                           are still    unknown,such butas an areincreased
                                                                                                                     being thoroughlyrisk of cardiovascular
                                                                                                                                                 tested in clinical
                              events or organ toxicity, are still unknown, but are being thoroughly tested in clinical trials
                                trials  [127,129].
                              [127,129].
                                       In a preclinical study testing the use of lipid-containing nanoparticles with the precur-
                                sor In   a preclinical
                                     ALN-PCS         it hasstudy
                                                               been testing
                                                                       shownthe  thatuse thisofsystem
                                                                                                 lipid-containing
                                                                                                           was able to       nanoparticles
                                                                                                                                reduce PCSK9        with
                                                                                                                                                       mRNAthe pre- and
                              cursor
                                protein ALN-PCS         it has been
                                           concentrations               shown
                                                                   by 70%    andthatLDL-c this concentrations
                                                                                                system was ableby          to60%.
                                                                                                                               reduce     PCSK9
                                                                                                                                      These           mRNA
                                                                                                                                                effects    lasted and for
                              protein concentrations by 70% and LDL-c concentrations by 60%. These effects lasted for
                                3 weeks     after   a   single    intravenous       administration           in   the    animal      models.       This   treatment
                              3 was
                                 weeksalso after
                                             ablea to single
                                                          reduceintravenous       administration
                                                                     total cholesterol        and apoB     inlevels
                                                                                                                the animal[133]. models.
                                                                                                                                    A phaseThis         treatment
                                                                                                                                                   I clinical    study
                                that  observed       the    effects   of  ALN-PCS          did   not  show       any
                              was also able to reduce total cholesterol and apoB levels [133]. A phase I clinical study  serious     adverse       effects.    On    the
                                contrary,    PCSK9        and    LDL-c    levels    were     significantly        reduced
                              that observed the effects of ALN-PCS did not show any serious adverse effects. On the              for  at  least   6  months       [134].
                                According
                              contrary,        to theand
                                            PCSK9          meta-analysis
                                                               LDL-c levelsofwere  randomized          clinical
                                                                                           significantly            trials, for
                                                                                                               reduced        in patients
                                                                                                                                    at least 6withmonthsfamilial
                                                                                                                                                               [134]. hy-
                                percholesterolemia and/or atherosclerosis, the drug found to be effective, safe, and well
                                tolerated at lower LDL-c levels and exerted only mild adverse effects along with injection
                                site reaction [135].
Biomedicines 2022, 10, 1090                                                                                            11 of 18

                              3.3.2. Vaccination against PCSK9
                                    Vaccination strategies could be a better alternative than monoclonal antibodies be-
                              cause their approach is less expensive and does not require frequent administration
                              intervals [136,137]. The peptide-based PCSK9 vaccine has been shown to induce the pro-
                              duction of antibodies that improve the lipid profile for up to 24 to 40 weeks, according
                              to data from animal studies [138,139]. According to the study of Landlinger, the PCSK9
                              vaccine reduced plasma lipids and systemic and vascular inflammation by reducing plasma
                              inflammatory markers and vascular endothelial growth factor leading to weakening of
                              atherosclerotic lesions in the aorta [140]. Similarly, in a peptide-vaccinated mice, an effective
                              immune response was associated with a significantly improved lipid profile [141]. In vacci-
                              nated hypercholesterolemic mice, Wu et al. have shown a reduction in total cholesterol and
                              LDL-c [142]. Recently, phase I clinical trial is conducted to assess the safety and tolerability
                              of anti-PCSK9 vaccine strategy in healthy subjects. The results have not been published
                              yet [143].
                                    Momtazi-Borojeni et al. introduced a nanoliposomal anti-PCSK9 vaccine [144] and in
                              the first approach administered it to healthy animals to observe antibody production [145].
                              After detection of antibody production, immunized rats were administered intraperi-
                              toneally with streptozotocin to induce diabetes mellitus. It is reported that vaccinated
                              rats showed lower plasma LDL-c levels compared to non-vaccinated diabetic rats. Hy-
                              perglycemia was suppressed in vaccinated rats [146]. In hypercholesterolemic mice, the
                              nanoliposomal vaccine against PCSK9 promoted antibodies that inhibited the interaction
                              between PCSK 9 and the LDL receptor and also led to a reduction in LDL and triglycerides.
                              In addition, a reduction in inflammatory cells as well as a reduction in the size of atheroscle-
                              rotic lesions have been observed [147]. Ortega-Rivera et al. established technology for a
                              single-dose multi-target vaccination strategy targeting ‘cholesterol checkpoint’ proteins in-
                              cluding PCSK9, ApoB, and cholesteryl ester transfer protein (CETP). The candidate vaccine
                              was developed using virus-like particles from bacteriophage Q-beta that exhibit PCSK9,
                              ApoB and CETP antigens. Decreased plasma PCSK9 and ApoB levels, in vitro CETP
                              inhibition and decreased total plasma cholesterol were observed after vaccination [148].

                              3.3.3. Small Molecule PCSK9 Targeting
                                   Agents of small molecule PCSK9 targeting could be a new alternative of lipid-lowering
                              drugs. They are potentially safer and have more advantages such as small size, low cost, and
                              easier production than the other respective treatments [149,150]. Despite that, development
                              of small molecule PCSK9 targeting is still in the preclinical stage [151].
                                   Shifa Biomedical has developed the first small molecule that targets PCSK9, called P-4.
                              It successfully inhibits PCSK9 and LDL binding and reduces circulating LDL-c levels. Due
                              to the low solubility and permeability in the water, a new nano-formulation P-4 called P-21
                              has been developed to overcome these obstacles. The P-4 formulation had better plasma
                              bioavailability, than P-21. However, targeted delivery of the P-21 nano-formulation to the
                              hepatocytes showed higher bioavailability in the liver. The LDL-c lowering efficacy of P-21
                              was about 90% and much higher than that of P-4. In addition, it led to a 2-fold increase
                              in HDL-c after two weeks of oral treatment. The development of a small molecule nano-
                              formulation represents a new alternative to lipid profile modification with the potential for
                              significant benefits for clinical practice [152].

                              4. Conclusions
                                   Nowadays, statins are the key medication for treatment of dyslipidemia in patients
                              who have or are at substantial risk for atherosclerotic cardiovascular diseases. However,
                              with the intensity of statin treatment, intolerance to their use has the increasing tendency.
                              Therefore, new lipid-lowering drugs that interfere with various mechanisms of lipid path-
                              ways are being evaluated. Clinical trials have documented that ezetimibe and PCSK9
                              inhibitors can successfully lower LDL-c level and contribute to lowering cardiovascular
                              events. However, recent studies have shown that also these drugs may have some side
Biomedicines 2022, 10, 1090                                                                                                            12 of 18

                                   effects, or the route of their administration is unsatisfactory. To improve the therapeutic
                                   efficacy of the lipid-lowering drugs and reduce their side effects different nanoparticle
                                   systems with targeted strategy have been developed and described. Thus, enhancing the
                                   pleiotropic effects of lipid-lowering drugs by suitable targeted strategy may represent a
                                   promising tool for the treatment of atherosclerotic cardiovascular diseases.

                                   Author Contributions: Conceptualization, E.D. and O.P.; Formal analysis, E.D. and O.P.; Writing—
                                   original draft preparation, E.D. and O.P.; Writing—review and editing, E.D. and O.P.; supervision,
                                   O.P.; project administration, O.P.; funding acquisition, O.P. All authors have read and agreed to the
                                   published version of the manuscript.
                                   Funding: This work was supported by the national grant agencies APVV 14-0932 and VEGA
                                   2/0112/19; 2/0132/20; 1/0035/19; 2/0151/18 and by the The European Regional Development
                                   Fund “Vývoj biomodelov pre zlepšenie hodnotenia účinnosti liekov a látok, ktoré majú potenciál
                                   pri liečbe COVID-19 (BIOVID-19)”—ITMS2014+:313011AVG3 and “Centrum pre biomedicínsky
                                   výskum—BIOMEDIRES—II. etapa”—ITMS2014+:313011W428, and ITMS project code 313021T081
                                   (supported by the Integrated Infrastructure Operational Programme funded by the ERDF).
                                   Institutional Review Board Statement: Not applicable.
                                   Informed Consent Statement: Not applicable.
                                   Conflicts of Interest: The authors declare no conflict of interest.

References
1.    Ichai, C.; Quintard, H.; Orban, J.C. Metabolic Disorders and Critically III Patients from Pathophysiology to Treatment, 1st ed.; Springer
      International Publishing AG: Cham, Switzerland, 2018; p. 483.
2.    Eckel, R.H.; Alberti, K.G.; Grundy, S.M.; Zimmet, P.Z. The metabolic syndrome. Lancet 2010, 375, 181–183. [CrossRef]
3.    Ott, C.; Schmieder, R.E. The role of statins in the treatment of the metabolic syndrome. Curr. Hypertens. Rep. 2009, 11, 143–149.
      [CrossRef] [PubMed]
4.    Pechanova, O.; Varga, Z.V.; Cebova, M.; Giricz, Z.; Pacher, P.; Ferdinandy, P. Cardiac NO signalling in the metabolic syndrome. Br.
      J. Pharmacol. 2015, 172, 1415–1433. [CrossRef] [PubMed]
5.    Feher, M.D. Lipid lowering to delay the progression of coronary artery disease. Heart 2003, 89, 451–458. [CrossRef] [PubMed]
6.    Lewis, S.J. Lipid-lowering therapy: Who can benefit? Vasc. Health Risk Manag. 2011, 7, 525–534. [CrossRef] [PubMed]
7.    Li, H.; Jing, X.; Deng, X.; Ouyang, D. Targets of anti-hyperlipidemia drugs. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013, 38,
      101–108. (In Chinese) [CrossRef]
8.    Niedzielski, M.; Broncel, M.; Gorzelak-Pabiś, P.; Woźniak, E. New possible pharmacological targets for statins and ezetimibe.
      Biomed. Pharmacother. 2020, 129, 110388. [CrossRef]
9.    Musunuru, K. Treating Coronary Artery Disease: Beyond Statins, Ezetimibe, and PCSK9 Inhibition. Annu. Rev. Med. 2021, 72,
      447–458. [CrossRef]
10.   Cheung, Y.M.; O’Brien, R.; Ekinci, E.I. What is new in lipid-lowering therapies in diabetes? Intern. Med. J. 2019, 49, 1472–1480.
      [CrossRef]
11.   Hardie, D.G. Regulation of fatty acid and cholesterol metabolism by the AMP-activated protein kinase. Biochim. Biophys. Acta
      1992, 1123, 231–238. [CrossRef]
12.   Pinal-Fernandez, I.; Casal-Dominguez, M.; Mammen, A.L. Statins: Pros and cons. Med. Clin. 2018, 150, 398–402. [CrossRef]
      [PubMed]
13.   Förstermann, U.; Li, H. Therapeutic effect of enhancing endothelial nitric oxide synthase (eNOS) expression and preventing
      eNOS uncoupling. Br. J. Pharmacol. 2011, 164, 213–223. [CrossRef]
14.   Torok, J.; Luptak, I.; Matuskova, J.; Pechanova, O.; Zicha, J.; Kunes, J.; Simko, F. Comparison of the effect of simvastatin,
      spironolactone and L-arginine on endothelial function of aorta in hereditary hypertriglyceridemic rats. Physiol. Res. 2007, 56,
      S33–S40. [CrossRef] [PubMed]
15.   Simko, F.; Matuskova, J.; Luptak, I.; Krajcirovicova, K.; Kucharska, J.; Gvozdjakova, A.; Babal, P.; Pechanova, O. Effect of
      simvastatin on remodeling of the left ventricle and aorta in L-NAME-induced hypertension. Life Sci. 2004, 74, 1211–1224.
      [CrossRef] [PubMed]
16.   Greenwood, J.; Mason, J.C. Statins and the vascular endothelial inflammatory response. Trends Immunol. 2007, 28, 88–98.
      [CrossRef]
17.   Helfand, M.; Carson, S.; Kelley, C. Drug Class Review on HMG-CoA Reductase Inhibitors (Statins): Final Report [Internet]; Oregon
      Health & Science University: Portland, OR, USA, 2006.
You can also read