Access to this full-text is provided by Wiley.
Content available from Journal of Clinical Laboratory Analysis
This content is subject to copyright. Terms and conditions apply.
J Clin Lab Anal. 2022;36:e24552.
|
1 of 13
https://doi.org/10.1002/jcla.24552
wileyonlinelibrary.com/journal/jcla
1 | INTRODUCTION
Familial hypercholesterolemia (FH) is an autosomal codominant
genetic disease characterized by high serum levels of low- density
lipoprotein cholesterol (LDL- C).1 The disease is diagnosed by the
presence of mutations in low- density lipoprotein receptor (LDLR),
apolipoprotein (ApoB), or proprotein convertase subtilisin/kexin
9 (PC SK9).2 The LDLR variant is the most common cause of FH
and leads to the malfunction of the LDLR and a defect in the re-
moval of LDL- C from blood. Less common are pathogenic variants
in the ApoB gene, which encodes apolipoprotein B100 (ApoB100),
and these variants may be the predominant cause of 5%– 15% of
Received:20April2022
|
Revised:27May2022
|
Accepted:28May2022
DOI: 10.1002/jcla.24552
REVIEW ESSAY
The promising novel therapies for familial
hypercholesterolemia
Ruoyu Chen1 | Shaoyi Lin2 | Xiaomin Chen2,3
This is an op en access ar ticle under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provide d the original work is properly cited.
© 2022 The Authors. Journal of Clinical Laboratory Analysis published by Wiley Periodicals LLC.
1SchoolofMedicineofNingboUniver sity,
Ningbo,China
2TheAff iliatedNingboFirstHospital,
SchoolofMedicineofNingboUniver sity,
Ningbo,China
3NingboFirstHospitalAffiliatedtoSchool
ofMedicineofZhejiangUniversity,
Ningbo,China
Correspondence
XiaominChen,NingboFirstHospital
AffiliatedtoSchoolofMedicineof
ZhejiangUniversit y,Ningbo,Zhejiang,
China.
Email: chxmin@hotmail.com
Funding information
NaturalScienceFoun dationofZhejiang
Province,Grant/AwardNumber:
LQ20H020001;NingboHealthBranding
Subjec tFund,Grant/AwardNumber :
PPXK2018-01
Abstract
Background: The incidence of premature atherosclerotic cardiovascular disease in fa-
milial hypercholesterolemia (FH) is high. In recent years, novel therapeutic modalities
have shown significant lipid- lowering ability. In this paper, we summarize the recent
developments in novel therapies for FH via the treatment of different targets and
discuss the characteristics of each targeted therapy. Based on the process of protein
synthesis,weattempttosummarizethedirect-effecttargetsincludingprotein,RNA,
andDNA.
Methods: For this systematic review, relevant studies are assessed by searching in
several databasesincluding PubMed,Web of Science,Scopus,andGoogle Scholar.
The publications of original researches are considered for screening.
Results: Most drug s are protein-targeted su ch as molecule-bas ed and monoclonal
antibodies, including statins, ezetimibe, alirocumab, evolocumab, and evinacumab.
Bothantisenseoligonucleotide(ASO)andsmallinterferingRNA(siRNA)approaches,
suchasmipomersen,vupanorsen,inclisiran,andARO-ANG3,aredesignedtoreduce
thenumberofmRNAtranscriptsandthendegradeproteins.DNA-targetedtherapies
such as adeno- associated virus or CRISPR– Cas9 modification could be used to deliver
or edit genes to address a genetic deficiency and improve the related phenotype.
Conclusion: Whilethetherapies based ondifferent targetsincluding protein, RNA,
and DNA are on d ifferent stage s ofd evelopment, the me chanisms of these novel
therapies may provide new ideas for precision medicine.
KEYWORDS
familial hypercholesterolemia, gene therapy, lipidology, precision medicine
2 of 13
|
CHEN Et al.
FH cases, leading to reduced binding of LDL- C to LDLR. Gain- of-
function (GOF) mutations in the PC SK9 gene account for 1% of re-
ported FH cases, such mutations increase the destruction of LDLR
on the hepatocyte sur face and accordingly result in an increase in
levels of circulating LDL- C.3
The global morbidity rate of FH is between 1:200 and 1:300 with
limited therapies.4 Patients with Heterozygous FH (HeFH) in China
usually have a 3 .5- to 16- fold in creased risk of corona ry arter y disease
(CAD) in comparison with non- FH patients. If untreated, FH would
lead to premature atherosclerosis and an increased risk of cardiovas-
cular events.5 The decrease in LDL- C levels has been explicitly shown
to mitigate cardiovascular risk.6,7 The goal of the first- line treatments
is to limit cholesterol and saturated fats via a healthy way of living.8
However, lifestyle alone is insufficient for maintaining the normal av-
erage blood LDL- C levels in patients with FH. Consequently, the use
of lipid- lowering therapy is vital. The pharmacological options now
available include statins, bile acid sequestrants, fibrates, and choles-
terol absorption inhibitors.9,10 Although advances have been made in
potential therapies and treatment of plasma lipids shows improve-
ment, cer tain patients with FH fail to meet the treatment targets
for LDL- C.11 In addition, it is sometimes very difficult for traditional
pharmacotherapy to be ef fective for patients with homozygous FH
(HoFH), unless pharmacotherapy is coupled with LDL apheresis.
Although LDL apheresis or orthotopic liver transplantation has been
considered to be an optional therapeutic method for patients with
HoFH, the complex and risky operation, severely limited availability
of suitable donors, surgical risks, and perpetual use of immunosup-
pressive drugs after transplantation poses great challenges.12,13
Currently, the majority of clinical therapies for patients with FH
are small- molecule- or antibody- based approaches. It is important
to develop new lipid- lowering therapies with novel mechanisms of
action and favorable side effect profiles.14 We assume that the ex-
pansion of novel FH therapy options may have the potential to bring
about considerable economic and social benefits.
In this review, we propose novel therapies for FH according to
thedifferenttargets—proteins,mRNAs,andDNA.Inmolecularbiol-
ogy, such therapies depend on the different stages of central dogma:
the transcriptionofDNAinto mRNA and the translationof mRNA
into proteins.15
2 | PROTEIN- TARGETED THERAPEUTICS
In the clinic, the most commonly used drugs are small compounds.
These drugs interact with target proteins to influence biological
functions.
2.1 | Statins
Protein- targeted therapeutics such as statins can inhibit receptors
or enzymes in the cytoplasm or on the cytomembrane. Statins are
small- molecule lipid- lowering drugs that act on proteins in nanogram
to microgram.16
Statins are hydroxymethylglutaryl coenzyme A (3- hydroxy- 3-
methyl-glutaryl-CoA, HMG-CoA) reductase inhibitors. In clinical
practice, statins are the first- line treatments for patients with FH in
terms of pharmacotherapeutic management.17 Statins inter fere with
cholesterol biosynthesis by restricting the key step in the biosynthe-
sisofisoprenoidsandsterols,andthisprocessiscatalyzedbyHMG-
CoA.18 In a randomized controlled trial (RCT ) of patients with HeFH,
dailyadministrationof80-mgatorvastatinresulted,onaverage,ina
50% decrease in LDL- C levels from baseline. In addition to reducing
LDL- C levels, statins also mitigate the risk of cardiovascular disease
(CVD) and improve certain outcomes in prognosis.7,1 9
Although the options for lipid- lowering therapy have increased
markedly since statins were introduced for clinical applications in
1987,only10%–25% ofdiagnosedpatients havereceived appropri-
ate therapy, and many individuals have failed to achieve their LDL- C
goals.20 At present, most drugs interact with proteins, and they often
bind to nontarget proteins or produce adverse effects via unnoticed
interactions.21 Some patients are intolerant to statins and are easily
affected by side effects such as myalgias and weakness.22Further-
more, statins upregulate the expression of not only LDLR but also
PCSK9, which may limit the pharmacological ef fects of lowering
LDL- C levels. This is the “paradoxical effect” of statin management.23
2.2 | Ezetimibe
Studies show that adding other lowering agents to the statins may
further lower LDL- C with few side effects. In the clinic, the Food
and Drug Administration (FDA) approved the ezetimibe in 2002.
Ezetimibe, a cholesterol absorption inhibitor, is used as an adjunct to
statin forfurtherreduceLDL-C.ItworksbyblockingtheNiemann-
Pick C1-Like (NPC1L1) receptor, which is a multipass membrane
protein, expressed in the small intestine and liver. The NPC1L1
plays an important role in the process in cholesterol movement
into the enterocyte.24 Individuals with the non- GOF mutation of
the NPC1L1genehave lower levels ofLDL-Candacorresponding
risk of ASC VD.25 In addition, the ezetimibe upregulates LDLR in the
liver to reduce the circulating LDL- C; it is an independent mechanism
to statins.26Manyresearchexploredthe relationshipbetweenthe
additive effects of statins and ezetimibe. It provided an additional
23.4% reduction in LDL- C compared with single statins therapy.
What is more, the drug combination reduced high- sensitivity C-
reactive protein (CRP).27
Nowadays,whiletherehasundertreatmentofFH,mainlytreat-
ment priorities for drugs are statins, ezetimibe, and bile acid bind
resins.28
2.3 | Alirocumab and evolocumab
Protein- targeted therapeutics also include antibody- based PCSK9
inhibitor drugs such as alirocumab and evolocumab, which block
proteins in plasma at microgram to milligram.29 PCSK9 is an intrinsic
protein that modulates the amount of LDLR and is very impor tant in
|
3 of 13
CHEN Et al .
LDL-Cmetabolism.Moreover,PCSK9issecretedfromhepatocytes
into blood and binds to LDLR , which targets LDLR for lysosomal
degradation.30 The loss- of- function (LOF) mutation of PCSK9, which
was found to decrease LDL- C levels, reinforces the fact that PCSK9
could be a target for therapy.31 The two antibodies currently in use
against PCSK9 are fully human IgG subtypes that modify this path-
way by binding to circulating PCSK9 and preventing it from binding
LDLR.32 This process results in a lack of PCSK9 and leads to a large
accumulationofLDLRon the membranes oflivercells. Ultimately,
the removal of LDL particles is accelerated, and circulating LDL- C
levels are substantially reduced.33 Subcutaneous injection of PCSK9
inhibitors bound all newly secreted PCSK9 in the serum within hours
of administration, and the effect lasted for the next few days.34,35
However, this approach results in a large accumulation of the
compound in the blood, with an average 10- fold increase. For some
patients, the total concentrations would be increased 20- fold.36 As
part of the immune complex, clearance of PCSK9 may be slow, and
the return of circulating PCSK9 to the liver would be inhibited. This
may stimulate counterregulatory transcriptional pathways that en-
hance PCSK9 synthesis and secretion.
2.4 | Evinacumab
Traditional lipid- lowering therapies, such as statins and PCSK9 in-
hibitors, work by upregulating LDLR expression, but there is virtu-
ally no activity in individuals with two null alleles, such as those with
HoFH. Evinacumab is a monoclonal antibody against angiopoietin-
like protein 3 (ANGPTL3); it has received FDA approval in the
United States and can confer a potential benefitinHoFH.37 Both
ANGPLT3LOFvariantsandpharmacologicinhibitionofANGPTL3
can lower LDL- C levels without LDLR activity.37ANGPTL3isa
secreted hepatic glycoprotein that disrupts the clearance of cir-
culating LDL- C by inhibiting lipases such as endothelial lipase and
lipoprotein lipase, which hydrolyze triglycerides and phospho-
lipids in circulating lipoproteins.38 Genome- wide association and
exome sequencing studies have found correlations bet ween the
LOFgeneticvariantsofANGPTL3andareduction inplasmaLDL-
C, high- density lipoprotein cholesterol (HDL- C), and triglyceride
levels for cardiovascular prevention.39– 41 Prior to these findings,
acombinedhypolipidemia phenotype was observed inANGPTL3
knockout mice.42 In a phase 3 trial, the LDL- C level was decreased
from baseline in the evinacumab group compared with the placebo
group, which ledtoa 49.0%differenceinLDL-C levels anda47%
differ ence in triglyce ride levels bet ween the grou ps at 24 weeks,
regardless of the degree of their LDLR function.37 Evinacumab was
firstapprovedintheUSAonFebruary11,2021,assupplementary
therapy for LDL- C reduction for adult and pediatric patients with
HoFHwhowereolderthan12 years,andithasreceivedapositive
responseintheEuropeanUnion.38
However, the use of antibody- based drugs may create problems
such as patient compliance, problems with the injection technique,
and so on. For evinacumab, the main side effects include nasophar-
yngitis, influenza- like illness, headache, and infusion- site pruritus.37
Therefore, more options are needed because of the interactions and
mechanisms of these drugs.
In summar y, discrimination between the primary t argets and off-
targets is necessar y in the development of protein- targeted drugs.
The related phenotypes, including the curative effects and side ef-
fects, are dependent on the activation or inhibition of target pro-
teins to a large extent.
3 | RNA- TARGETED THERAPEUTICS
Small molecules that affect proteins by inhibiting enzyme function
or receptor activity have no therapeutic effec t on proteins without
enzyme activity. In addition, it may be difficult to achieve adequate
concentrations of humanized monoclonal antibodies in circulation
because of tolerance and cost considerations.43
RNA-targeted therapeutics seemtobe anovelandelegantap-
proach. It can act to regulate genes by directly targeting the nucleic
acids that encode the proteins by interfering with the translation of
mRNAintoprotein.Furthermore,RNA-targetedtherapeutics,which
represent a drug discovery platform involving oligonucleotides,
are akin to small- molecule therapeutics and include small interfer-
ing RNA (siRNA), antisense oligonucleotide (ASO) (Figures 1 and
2), oligonucleotide-induced alternative splicing, anti-miRs, miRNA
mimics,andmRNAupregulation.44 They are synthetic and small, do
not integrate into the host genome, and have limited duration and
activity.45
ASOsorsiRNAcanbindtothemRNAofdisease-causinggenes
and block their activation.46,47 Notably, N-acetylgalactosamine
(GalNAc) modification ofASOs orsiRNAbytheasialoglycoprotein
receptor could facilitate selective uptake into hepatocytes and
FIGURE 1 Antisenseoligonucleotide-basedapproaches:ASO
utilizesasingle-strandedRNaseHmechanism
4 of 13
|
CHEN Et al.
ultimately significantly increase hepatic uptake.48 Therefore, these
approachescouldpreciselyandselectivelymatchthespecificmRNA
intargetedorgansorcells.Ingeneral, thenewgenerationof RNA-
targeted drugs has enhanced potency, thereby reducing plasma drug
levels, improving efficacy, and possibly reducing side effects.49
3.1 | Antisense oligonucleotides (ASOs)
Sincethe1970s,ASOshavefrequentlybeenusedforsilencinggene
expressionandcanlowerthelevelsofspecificmRNAsinasafeand
selective manner.50 ASOs are single- stranded molecules composed
ofmodifiedDNA.ThemiddlebasesareDNA,whilethoseoneither
sideareusuallymodifiedDNAthat hasbeencorrected, often con-
taining 2′methoxyethyl.Anunspecifiednumberofphosphorothio-
ate moieties often replace the phosphodiester backbone, while the
nucleic acid bases are not modified. The chemical modifications con-
tribute to increased plasma stability and high affinity and binding
tomRNA. Thisisthe keyreasonthatASOscanbeused asadrug,
unlike unmodified native oligonucleotides, which degrade promptly
in circulation.43 After subcutaneous injection, ASOs bind to circulat-
ing proteins, reach the liver, enter the cells, and then reach the nu-
clear membranes for ef ficacy. Because of their amphipathicity, ASOs
can be dissolved in normal saline and be injected subcutaneously
with no carrier. The basic premise of the use of ASOs is the abil-
ity to administer shor t- and single- stranded nucleic acid sequences
that bindtocomplementar yRNAsubstrates. Then, the ASOs bind
tospecificsensemRNAsincellswithinthenucleusorcytoplasmby
Watson– Crick base pairing; finally, the resulting complex competi-
tively inhibits translation or is degraded (Figure 1).51 ,52 ASOs have
rapid plasma uptake and a short distribution life (<1 h) but have a
relatively long intracellular life (2–4 weeks), consequently having
a long- lasting effect. They are then cleaved to short fragments by
exonucleases and endonucleases.53 Second- generation ASOs are
composedofsequence-specificduplexesofthe targetmRNA.The
duplexesserveasasubstrateforRNaseH,whichisanendogenous
mechani sm in cells for cleavi ng ASO-bound RN A, result ing in the
degradation of target mRNA and ultimately in a decrease in the
translation of the corresponding protein.54, 55
3.1.1 | ApoB100-specificantisense
oligonucleotides:Mipomersen
Mipomersen(Kynamro)isasecond-generation20-nucleotideASO
that binds to A poB100 mRNA in t he liver and cr eates a subst rate
for intrahepatic endonucleases. Then, the transcripts of ApoB100
are degraded, thereby reducing plasma LDL- C concentrations.56
ApoB100 is o ne of two isoforms fro m the RNA transc ript of the
human ApoB gene, which has great importance in lipoprotein me-
tabolism. And, ApoB100 is a vital structural component of ver y
low- density lipoprotein ( VLDL) and LDL, which are synthesized by
the human liver. In the early phase of atherosclerosis, dysfunctional
arterial endothelial cells allow LDL to enter the endothelial layer
and then adhere to intimal proteoglycans by way of ApoB, where it
accumulates gradually in the intimal layer.57,5 8 The genetic basis of
non- ApoB family hypobetalipoproteinemia (FHBL) suggests the pos-
sibility of ApoB as an attractive target for lipid- lowering therapy.59
FHBL is an autosomal dominant disease characterized by low LDL- C
and ApoB levels, which may be due to the mutation of ApoB.59
Mipomersenincludes10 2′-O-methoxyethylmodifications,and
there are five modifications at each end of the oligonucleotide that
increase the affinit y of target binding, prolong the half- life in the
target organ, and resist degradation by nucleases.60 In the center,
the phosphorothioate nucleotides promote RNase H-mediated
cleavage.55 The plasma concentrations of mipomersen peak within
4 h after ad ministrati on. Mipomer sen is metabo lized in tissues as
a chain- shortened metabolite, which is initially accomplished by
endonucleases and produces oligonucleotides that have no further
pharmacological effects. The smaller oligonucleotides are further di-
gested by exonucleases and excreted mainly in the urine.61 Studies
have shown that in healthy adults, mipomersen could increase the
fractional catabolic rate of ApoB in both VLDL and LDL, thereby
decreasing the levels of lipoproteins that contain ApoB.62 Phase I
and II studies showed that mipomersen produced dose- dependent
decreases in all ApoB- containing lipoproteins. In phase III trials, mi-
pomersen was effective in patient s with HoFH and patients with
HeFH.63 In a pri mary phas e III study, patien ts older tha n 12 years
who were clinically diagnosed with or genetically confirmed to have
HoFH received the maximum tolerated dose of a lipid- lowering
drug andwereassignedtoreceivemipomersen 200 mg or placebo
FIGURE 2 siRNA-basedapproaches:siRNAtechnologyemploys
a double- stranded RISC mechanism
|
5 of 13
CHEN Et al .
weekl y.64LDL-Clevelsdecreasedbyameanpercentageof24.7%in
all subjec ts after 6 months.65 In randomized trials and the open- label
extension phase, long- term mipomersen treatment of FH reduced
the levels of atherogenic lipoproteins and led to a decrease in car-
diovascular events.66
However, mipomersen is approved only for HoFH, and the re-
sponse varies widely among patients. Loss of ApoB is also associated
with on- target adverse effects such as liver steatosis, possibly be-
cause all ApoB- containing lipoproteins are targeted and there may
be no mechanism by which the liver clears excess fat. The use of
mipomersen is complicated by its side effects. For instance, hepato-
toxicity, injection- related side effects, hepatic fat, and the high cost
of the medication are also limiting factors.67
3.1.2 | ANGPTL3-specificantisense
oligonucleotides: Vupanorsen
ANGPTL3has been assessed asapotentialtargetfor loweringthe
levels of LDL- C and triglycerides for CVD prevention.68 Vupanorsen
(AKCEA-ANGPTL3-LRx) is an N-acetyl galactosamine-conjugated
(GalNAc3-modified)ASOtargetedtotheliverthatselectivelyinhib-
itsthesynthesisoftheANGPTL3protein.GalNAc3-modifiedASOs
can target the ASO to the asialoglycoprotein receptor on liver cells,
and it has a treatment effect similar to that of unconjugated ASOs,
although at a 20– 30- fold lower dose. Therefore, it could reduce
systemic exposure risk and be administered at significantly lower
doses and intervals.69 In a phase 2 study, 105 patients had fasting
triglycerides >150 mg/dl(>1.7 mmol/L),type2diabetes,andhepatic
steatosis at baseline. The patients received vupanorsen for 6 months
bysubcutaneousinjectionof40or80 mgmonthlyor20 mgweekly.
The vupanorsen groups achieved a statistically significant dose-
dependent reduction in triglyceride levels (44%) compared with
the placebo group, but no changes in liver fat or HbA1c were ob-
served.Inaddition,thereductionsinapolipoproteinC-III(58%),re-
sidualcholesterol(38%),totalcholesterol(19%),non-HDL-C(18%),
HDL-C(24%),apolipoproteinB(9%),andLDL-C(7%)werealsodose
dependent, and the frequent side effects were generally mild and
occurred at the injection site.70
Vupanorsen is also distinct from evinacumab, which is a mono-
clonal ant ibody targe ted to ANGPTL 3. Vupanorsen a cts in liver
cells, but evinacumab acts in plasma. Whether this would cause
a difference in effect is still not clear. Evinacumab appears to
be deficient regarding compliance, cost, and an inability to self-
administer.Thisisduetothehighintravenousdosages(15–20 mg/
kg) or subcutaneous injections required each week.71 An increas-
ing number of epidemiologic, genetic, and genome- wide associ-
ation stu dies suggest tha t reducing plasma AN GPTL3 levels by
inhibitinghepaticANGPTL3synthesiswillbenefitapolipoprotein
B levels and improve the metabolic measurements related to dys-
lipidemia and atherosclerosis.72 There was no correlation of the
effect with the concentration; rather, it is related only to complete
deficiency.73 Patients who are homozygous for LOF mutations in
the ANGP TL3 gene showed a dditional met abolic benef its, such
as lower circulating free fatty acid levels and better insulin sen-
sitivit y. In addition, vupanorsen could reduce ApoC- III levels in
respons e to the reductio n in ANGPTL3 leve ls. It is well known
that triglyceride levels may be related, but vupanorsen reduces
plasmatriglyceridesby targetinghepaticANGPTL3independent
of apoC- III.74
According to available studies, the adverse effects of vupan-
orsen are generally mild and include flu- like reactions and adverse
effects at the injection site.70 Further studies are needed to explore
the efficacy and safety of different doses of vupanorsen to maximize
target engagement in a population of individuals with elevated non-
HDL- C and triglyceride levels receiving statin treatment . In conclu-
sion, the reduction in triglyceride and atherogenic triglyceride- rich
lipoprotein levels by vupanorsen may suggest a novel strategy for
decreasing the CV risk for patients with FH.
3.2 | Small interfering RNA(siRNA) molecules
Small interferingRNA(siRNA)issmalldouble-strandedRNAmol-
ecules that are critical regulators of eukaryotic genome expression
and function. siRNA post-transcriptionally influences the mRNA
of target genes, ultimately degrading them and preventing trans-
lation.75TheydegradetargetmRNAbybindingwiththeRNA-
induced silencing complex (RISC), after which the antisense strand
ofsiRNAisloadedintotheRISCthroughWatson–Crickbasepair-
ing for seq uence-specif ic cleavage of th e mRNA.76 The RISC has
endonucleaseactivityto specificmRNA ,andthenaftercleavage,
thecellularexonucleasesdegradetheresultingmRNAfragments.77
Thecomplexof thesiRNAstrandandRISCcanthenbereusedto
targetothercomplementarymRNAs,therebyprovidingmoretran-
scriptsfor RNA silencingandmaintaining a long durationof effi-
cacy for an extended period of several months. (Figure 2) s iRNA
has activity in somatic and germline lineages of various eukaryotic
species.Small noncodingRNA regulatesvariousimportantstages
of genome function. Some examples are chromatin, chromosomal
segregation, transcription, RNA processing, RNA stability, and
translation.46 These molecules are also involved in protecting the
genome from invasive nucleic acids.78 In most mammalian cells, long
dou ble-s tra ndedRNAcannotbeusedtosi lenceasp ecificgenebe-
cause it provokes the interferon response to defend against viruses
and then prompts the shutdown of protein synthesis. In contrast,
siR NAc ane vad etheinterferonresponseinm amm alsandgenerate
specific and efficient gene silencing.79
3.2.1 | Inclisiran(ALN-PCSscPCSK9)
As shown in previous studies, PCSK9 inhibitors, such as evolocumab
and alirocumab, have become promising new therapeutic ap-
proachesforLDL-Creduction.Inrecentyears,siRNAhasalsobeen
used to inhibit PCSK9.80
6 of 13
|
CHEN Et al.
Inclisiran(ALN-PCSsc PCSK9) isalong-acting,synthetic siRNA
moleculethattargetsmRNA ofPCSK9tocounteractitandreduce
the production of intracellular PC SK9, resulting in a durable and sub-
stantial reduction in LDL- C levels.81,82 The inclisiran molecule con-
sists ofaguidestrandandpassengerstrand.Thetwo RNAstrands
are complementary. Once inclisiran molecules are incorporated into
hepatocytes, the guide strand binds the RISC and hybridizes with
thecomplementarymRNAofPCSK9,causingdegradation.83 Finall y,
translation of the PCSK9 protein would be limited, resulting in a re-
duction in serum LDL- C levels.84Notably,aspreviouslymentioned,
when degradation of the transcript is initiated, the complex is deliv-
ered to the liver where it remains active for a long period, interfering
wi thmor em RN A .85 In phase 2 and pha se 3 trials, inclisi ran was found
to decrease LDL- C levels by approximately 50% when administered
subcutaneously ever y 6 months. Inclisiran was well tolerated, and
there were no severe adverse events; however, injection- site side
effects were common compared to placebo (2.6% vs. 0.9% in the
ORION-10 trialand4.7% vs.0.5%in theORION-11trial),although
they were basically mild and did not last long.82 Interestingly, pa-
tients with diabetes showed no additional effects after treatment
withPCSK9-targetedsiRNA-drivenstrategies.86 In December 2020,
inclisiranwasfirstapprovedintheEUforuseinadultpatientswith
primary hypercholesterolemia, including HeFH and nonfamilial hy-
percholesterolemia or mixed dyslipidemia, by subcutaneous injec-
tion twice yearly as a dietary supplement. Inclisiran is intended to be
used in combination with statins or with statins combined with other
lipid- lowering treatments in patients who cannot meet LDL- C tar-
gets on the maximum tolerated statin doses. Inclisiran can be used
separately or in conjunc tion with other approaches for patients who
have statin intolerance or contraindications.87
3.2.2 | ARO-ANG3(ANGPTL3mRNA)
PreviousresultsindicatethatANGPTL3isanotablenewtherapeutic
target .Inadditiontomonocl onalantibodiesandA SOs ,siR NAthera-
piestargetingANGPTL3arealsobeingdeveloped.
The siRNA ANGPTL3 mRNA (ARO-ANG3) is a GalNAc3-
conjugated siRNA that effectively and persistently inhibits the
transcription ofANGPTL3 mRNAin liver cells.ThephaseI/IIclin-
icalstudyofARO-ANG3 (NCT03747224)isongoing.Inthecontrol
group, ARO -ANG3 tre atment for 16 weeks at do ses of 100, 200,
and 30 0 mg injected sub cutaneously r educed circulati ng levels of
ANGPT L3 by 96%, triglycerid es by 72%, and LDL-C b y 50%.88 In
the FH grou p, ARO-A NG3 reduce d ANGPTL3 l evels by 62%–92%
at week 16 in a dose- dependent manner. LDL- C levels were reduced
by23%–37%,andTGlevelswerereducedby25%–43%atdosesof
100,200,and300 mginjectedsubcutaneously.Fornon-FHpatients,
ANGPTL3levelswerereducedby85%,and LDL-Clevelswerere-
ducedby28%,comparabletothosepatient swithFH.
AsofMay2020,nodrug-relatedsideeffectsordiscontinuations
werefound.Mostadverseeventsweremild.Themostcommonad-
verseeventsofARO-ANG3treatmentarerespiratorytractinfection
(RTI) in 30% of participants and adverse events at the injection site
in 13% of participants.89
In general, whether the aforementioned siRNA treatment
method would be an effective approach for targeting ANGPTL3
needs further observation.
4 | DNA- TARGETED THERAPEUTICS
AlthoughRNA-targetedtherapiesareusedselectivelytosilenceand
interferewithgeneexpression,DNA-targetedtherapyisusuallyin-
tended to achieve the opposite effect. This technique involves intro-
ducing functional gene copies to restore the function of a mutated
gene.
Accordingtopreviousstudies,certainDNAsequencesofagene
have been observed to change lipoprotein levels, such as the LDLR
variant, which is the most common cause of FH.90 Some kinds of
variants are considered pathogenic, such as missense and nonsense
variants, insertion and deletion mutations, splicing mutations, and
large-scale DNA copy number variation (CNV).91 LDLR mutations
could influence various processes involved in LDL- C uptake and
metabolism and are the key driver of LDL- C clearance without sub-
stitutes in vivo.92Approximately 75%of human LDLR isexpressed
in the liver and determines LDL- C removal from the bloodstream.93
Patients with HoFH have biallelic LOF mutations in LDLR, and or-
thotopic liver transplantation effectively resolves the HoFH pheno-
type; however, the adverse ef fects and risks of transplantation and
long- term immunosuppression are limitations.94 Consequently, pa-
tients with HoFH are considered ideal candidates for gene delivery
and mutation correction.
Accumulating evidence supports the concept that overexpres-
sion of a corrected copy of a gene with an LOF mutation, such as
LDLR, by retrovirus,13 adenovirus, or adeno- associated virus (AAV)
can effectively ameliorate the phenotype.94 The first clinical trial
of gene replacement therapy was performed in five patients with
HoFH and used a recombinant murine retroviral vector that con-
tained LDLR cDNAtotransfect thecellsfromresectedlivertissue
ex vivo, after which the genetically modified cells were injected into
patients through a por tal vein catheter.13 Although the process is
safe and well tolerated, the challenges of the technique and inva-
siveness are present, and the inefficiency of the genetic recombina-
tion of retroviral vectors is the major barrier causing relatively low
ef fic ac y.95 Since then, in vivo preclinical research has star ted to ex-
plore efficient and safe vectors for delivering genetic cargo directly
to liver cells.96 Various methods for gene delivery have been studied
in different kinds of animal models of hypercholesterolemia with
varying degrees of success.97
AAV vectors have low immunogenicity and no pathogenicity in
humans, so AAV vectors could be suitable for gene delivery therapy.
AAV is a branch of the Parvoviridae family, and it has different se-
rotypes with different organ- specific tropisms.98,99Morethan90%
of LDL- C catabolism is mediated by LDLR in the liver. Thus, AAV se-
rotype8(AAV8)hasbeendeveloped,whichisahighlyhepatotropic
|
7 of 13
CHEN Et al .
vector.ArecombinantlivertropicAAV8vectornamedAAV8.TBG.
hLDLR is under development. This vector carries a human wild- type
(WT ) LDLR transgene under the control of a liver- specific promoter
called thyroxin- binding globulin. Recombinant LDLR- expressing
AAV8vectors have high efficiencyingenetransfer andstable he-
patocytetransductionforupto180 days(comparedtoothertissues
in which transduction is 1000- fold higher), and they significantly
improve lipid levels. In HeFH or HoFH humanized mouse animal
models,AAV8genetherapyleadssignificantreductionintotal and
LDL- C with safety.100 Studies have shown the efficiency of AAV vec-
tor transduction in human hepatocytes is lower than that in murine
hepatocytes.101Nonetheless,inthetreatmentofhemophiliaB,re-
combinantFIX-expressingAAV8inducedsignificantlyhigherserum
FIX levels inmicethan inhuman patients.Nevertheless,despite a
reconversionrateofonly1%–7% inpatientswithhemophilia com-
pared to individuals with a normal level, this vector could show sig-
nificantclinicalefficacythatlastedmorethan 3 years.102 The case
ofAAV8isrelatedtoitsusein HoFH.Thisis becauseHoFHindi-
viduals carrying LDLR variants with residual LDLR activity greater
than 2% (LDLR- defective) have a much better prognosis than indi-
viduals with LDLR variants with residual LDLR activity <2% (LDLR-
negative).103 According to the preclinical studies in preparation for a
phase 1clinicaltrial, AAV8.TBG.hLDLRhadgood safety.104,105 The
phase I and IItrials ofAAV-basedliver-directedA AV8.TBG.hLDLR
aiming to assess the safety and effic acy of the treatment for HoFH
are currently underway (NCT02651675; AAV8-mediated LDLR
Gene Replacement in Subjects With HoFH). Subjects in that study
arefollowedfor5 years.TheFDAhasapprovedtwoA AV-mediated
therapies. Other research in this area will allow further treatment
applications to be developed, and the results of the trials for HoFH
will generate new ideas for treating FH.
Genome editing can also permanently repair existing genes in-
stead of introducing a WT gene with gene replacement therapy.
Recently, treatment strategies involving the clustered- regularly
interspaced- short- palindromic- repeat/CRISPR- associated gene 9
(CRISPR– Cas9) system, an efficient gene- editing complex, have
begun to appear. TheRNA-guidednuclease cleavesthestrands of
DNAatthetargetsite.Then,theCRISPR–Cas9systemprovidesen-
gineereddonorDNAforinsertionintothetargetsitewhenthecell's
standardDNArepairmechanismtriestoinitiateDNArepair.106
CRISPR/Cas9 therapy has been explored for most genes associ-
ated with the regulation of lipid homeostasis and the genes targeted
in RNA therapeutics. Although gene editingmay not be poisedto
be adopted in clinical practice in the foreseeable future, achieve-
ments with this technique in animal models are notable.107,108 The
first HeFH mouse model created by in vivo somatic gene editing,
whichdoesnotpassmutationstothenextgeneration,usedA AV8-
delivered CRISPR/Cas9 and disrupted the LDLR gene in mice within
6 weeks, resultingin severe hypercholesterolemiaand atheroscle-
rosis.109Moreover,geneeditingofPCSK9byCRISPR–Cas9isac-
complished in animals through different delivery methods and has
revealed a clinically meaningful reduction of >30% in LDL- C levels.
This finding seems to suggest that permanent silencing of disease-
causing genes has tremendous therapeutic potential.110,111 A vari-
ant of CRISPR–Cas9 na med base editi ng does not nee d the DNA
double-strandbreak.InLDLR(−/−)mice,thismethodisusedtodis-
rupttheANGPTL3gene,andthelipidlevelsofthemicewerelargely
reduced without off- target mutagenesis.111
In another trial using the CRISPR– Cas9 system, T cells from
HoFH individuals were effectively reprogrammed, and then plu-
ripotent stem cells were induced.112 The study showed that LDLRs
are present in the cell membrane, providing a potential therapeu-
tic approach for FH. Another study described an ef ficient approach
for the simultaneous base editing and reprogramming of fibroblasts
using a CRISPR– Cas9 adenine base.113 In this study, the approach
generated gene- edited human- induced pluripotent stem cells iso-
lated from the skin biopsies of patients carrying pathogenic point
mutations and showed restoration of LDLR activity after gene mod-
ification. The approach is presented as being highly efficient while
significantly reducing the time of the in vitro cell culture, thereby
reducing the risk of in vitro changes. This may provide a solid basis
for further experiments in vivo.
In recent studies, somatic cell gene editing has been performed
in vivo by using the CRISPR/Cas9 system transduced via A AV to
treat FH induced by the LDLR mutation using germline editing in
mouse models.114 In this study, AAV- CRISPR /Cas9 was used to mod-
ify the point mutation in hepatocy tes and was delivered subcutane-
ouslyintoLDLRE208XmicecarryinganFH-relatedgenemutation.
Studies have shown that AAV- CRISPR/Cas9- mediated homologous
directed repair of LDLR gene correction can partly salvage LDLR
gene expression in vivo and efficiently improve atherosclerosis
phenotypes such as cholesterol, triglyceride, and LDL- C levels.114
Therefore, we can infer that the CRISPR– Cas9 system may play an
important role in the recovery of LDLR mutations or defect in many
othergenes,includingPCSK9,ApoB,andANGPTL-3,andwouldbea
broadly useful therapeutic approach for FH.
5 | CONCLUSIONS AND PERSPECTIVES
Despite significant advances in the treatment of FH, compound
HeFH and HoFH remain serious genetic disorders that affect the
human qua lity of life. For thes e patients, convent ional treatment may
not be very effective. Residual LDLR activity is critical for a thera-
peutic response, so restoration of LDLR activit y or achievement of
an obvious lipid- lowering effect is a challenging task. Furthermore,
novel therapies with mechanisms of action independent of this
pathway are being explored. Gene- based treatments offer great
potential and possibilities to regulate key points in lipid metabolism
withhighspecificityand efficiency.Noveltherapiesprovidemeth-
ods to address hitherto undruggable targets. While the long- term
safety and efficacy of such strategies still need more exploration,
these novel platforms are in different phases of clinical development
(Tables 1 and 2), which may trigger a paradigm shift in the treatment
8 of 13
|
CHEN Et al.
TABLE 1 NovelandemergingplatformsforFH
Classify (platform) Name Tar ge t Mechanism of actin Biochemical effect Stage Dose
Protein
Enzyme and substrate Statin HMG-CoA HMG-CoAcompetitive
combination
Reduce LDL- C Approved daily po
Enzyme and substrate Ezetimibe NPC1L1 NPC1L1inhibition Reduce LDL- C and hs- CRP combined statins Approved daily po
Antigen antibody Alirocumab, evolocumab PCSK9 Antigen antibody
reaction
Reduce LDL- C and Lp(a) Approved sc, once or twice monthly
Antigen antibody Evinacumab ANGPTL3mAb Antigen antibody
reaction
Reduce LDL- C and apolipoprotein B Approved 15 mg/kgivmonthly
RNA
ASO Mipomersen(Kynamro) APOB Anti- APOB antisense Reduce LDL- C Approved 200 mgsconceweekly
ASO Vupanorsen(ANGPTL3-LRx) ANGPTL3 Anti-ANGPTL3
antisense
Reduce TG, non- HDL- C Phases II 40-80 mgscmonthly
siRNA Inclisiran Anti- PCSK9
siRNA
PCSK9mRNA
degradation
Reduces LDL- C Approved 300 mgsct wiceyearly
siRNA ARO-ANG3 ANGPTL3 ANGPTL3mRNA
degradation
Reduces LDL- C phases I On study
DNA
AAV 8 AAV8.TBG.hLDLR(RGX-501)
RegenXbio, RGX- 501
LDLR LDLR gene therapy Reduces LDL- C Phases I
and II
Single iv infusion
CRI SPR/Cas9 /ANGPTL3/PCSK9 PCSK9 gene CRISPR/
Cas9 editing
/Preclinical /
Abbreviations: iv, intravenous; po, per os; sc, subcutaneous.
|
9 of 13
CHEN Et al .
TABLE 2 TheselectedandlatestclinicaltrialsofnovelandemergingplatformsforFH
Name Clinical trials Population Duration Treatment arms Primary endpoint Effic acy Safety
Alirocumab ODYSSEY HoFH
(NCT03156621)123
HoFH subjects
(N = 69)
12 weeks 150 mgevery2 weeks Percent change
in LDL- C from
baseline
LDL-C(−35.6%) Similar to placebo
Evolocumab OSLER-1(NCT01439880)124 FH subjects
(N = 1255)
1 year and
additional
4 years
420 mgmonthly Percent change
in LDL- C from
baseline
LDL-C(−56%,57%,
56%, 56%)
Similartoplacebo,5.7%
discontinuation
Mipomersen
(Kynamro)
NCT0 060737366 HoFH subjects aged
12 yearsand
older (N = 51)
26 weeks 20 0 mgweekly Percent change
in LDL- C from
baseline
LDL-C(−24.7%) Injection- site reaction
(76%vs24%inplaco)
Vupanorsen
(ANGPTL3-
LRx)
TRANSLATE–TIMI70
(NCT04516291)125
Non-
HDL-C ≥ 100 mg/
dl and
triglycerides 150
to500 mg/dlon
statin therapy
(N = 51)
24 weeks 80 mg,120 mg,or
160 mgevery
4 weeks,or60 mg,
80 mg,120 mg,
or160 mgever y
2 weeks
Percent change in
non– HDL- C from
baseline
Non–HDL-C22.0%
60 mg2weekly,
27.7%80 mg2
weekly
Injection- site reaction
(33.3%), ALT or AST
>3×ULN(44.4%)
Inclisiran ORION-1082 L D L - C > 1.8 mmol/L
(N = 1561 )
540 days 28 4 mgonday1,day
90, and every
6 months over a
periodof5 40 days
Percent change
in LDL- C from
baseline
LDL-C(−52.3%) Injection- site adverse
events (2.6% vs. 0.9%
in placo)
Inclisiran ORION-1182 ASCVD or A SCVD
risk equivalent
(N =1617 )
540 days 28 4 mgonday1,day
90, and every
6 months over a
periodof5 40 days
Percent change
in LDL- C from
baseline
LDL-C(−49.9%) Injection- site adverse
events(4.7%vs.0.5%
in placo)
ARO-ANG3 NCT03747 2241 26 Healthy and
dyslipidemia
individuals
(N = 93)
16 weeks 100 mg,200 mg,
300 mg
/ANGPTL3(−96%),
TG(−72%),LDL-C
(−50 %)
Similar to placebo
10 of 13
|
CHEN Et al.
of patients with FH, especially those who suffer from the severe
clinical symptoms of HoFH or are intolerant to conventional lipid-
lowering treatment.
Monoclo nal antibodie s against PC SK9 are a landmar k therapy
for patients with FH. They are combined with other lipid- lowering
treatment approaches to lower LDL- C levels, and less frequent dos-
ing provides greater convenience. In addition to PCSK9 suppression
based on antibodies, a variety of RNA-focused approaches have
been attempted. Gene- targeted treatments specifically intervene at
the transcriptional level, for instance, by impeding the produc tion
ofproteinsvia siRNA andASOsat themRNAlevel.If theDNAse-
quence is clearly noted to af fect the level of a lipoprotein that causes
certa in disorders , it may be a target fo r an ASO or siRNA , which
willintegratewiththemRNAofthespecificgene.Smallmolecules
are usually found in plasma and cells at similar concentrations, while
antibodies are found only in plasma and other extracellular compart-
ments.ASOsandsiRNAs are designedto reachhigher concentra-
tions in liver cells than in plasma.49
One of the important advantages of ASOs in comparison with
siRNA is th e markedly hig her affin ity for the t argeted mo lecule.80
ASOs presumablyactin thenucleus, whiletheactivityofsiRNAis
confined to the cytoplasm,115 which may decrease off- target hy-
bridization and side effects. However, due to the relatively short
duration of exposure in most clinical trials, the long- term effec ts of
ASOsandsiRNAsarebasicallyunknown.Thus,moreinvestigations
are needed.
In contrast to protein- targeted therapy, gene- based therapy is
sometimes not feasible. Early trials with ASOs targeting PCSK9 were
terminated due to renal tubular toxicity.116 Another option is to in-
crease the production of a specific protein by introducing specific
coding sequences such as LDLR via gene editing.
CRISPR/Cas9 is a very versatile approach with some contro-
versies. This technique produces double- strand breaks, which are
usually restored by nonhomologous end joining but are just as
likely to be repaired by faulty repair strategies.117 Alternatively, the
homology- directed repair is accomplished by careful nucleotide se-
quencereconstructionwith homologouschromosomal DNAorby
exogenous s ingle-stran d template DNA to i ntroduce a spe cific in-
corporation sequence.118 In addition, it is vital to distinguish somatic
gene edit ing and germline ge ne editing. In 2018 , the birth of th e
world's first twingirlsto havetheirgermcellseditedcausedshock
and outrage around the world.119 Germline editing refers to mak-
ing changes to the genome in the embryonic stage, which would be
passed on to future generations. This approach may create troubling
ethical issues.117
One alternative, exosomes, are intracellular vesicles that play a
significant role in intercellular communication.120 Studies have grad-
ually shown that exosomes may be promising treatment vectors,
andtheycaneffectivelyconveymRNA,miRNA,andplasmidDNAto
specific cells. Intracellular vesicles are naturally nanosized and easy
to use and have no cytotoxicit y or immunogenicity compared with
viruses.121TheycouldencapsulateabundantLDLRmRNAandex-
press LDLR in donor liver cells. In the recipient cells, encapsulated
mRNAissta bil izedand fun ctional.M ore over,thephenot ypesofani-
mal models may eventually be reversed using exosomes.122
In summar y, it is plausible to postulate that whether gene- based
therapeutics inhibit or enhance the targeted gene, appropriate drug
affinity, and resistance to nucleases are the key points that need to
be addressed and may help improve the efficiency and frequency
of medication, which requires further research and development in
chemical therapy. This would be an important step toward the prog-
ress of precision medicine. While novel therapies have entered clini-
cal use, problems such as high cost and the specific needs of various
subpopulations may arise. For rare and devastating diseases, gene
therapy may play a decisive therapeutic role. However, there are sig-
nificant ethical challenges related to the abuse of gene- modifying
techniques. As an example, the use of CRISPR/Cas9 technology for
the deliberate editing of human embryos caused much controversy.
This reminds us that we should remain vigilant to ensure the smooth
development of gene therapy in a scientific and ethical manner. To
sum up, it indicates that further research on different therapeutics
for different targets will be helpful in providing new ideas for the
individual treatment of various diseases.
CONFLICT OF INTEREST
We do not have any conflict of interest.
DATA AVAIL ABILI TY STATEMENT
Data sharing is not applicable to this article as no datasets were gen-
erated or analyzed during the current study.
ORCID
Ruoyu Chen https://orcid.org/0000-0002-5210-7702
Xiaomin Chen https://orcid.org/0000-0003-1498-0452
REFERENCES
1. Cuchel M , Bruckert E, Ginsberg HN, et al. Homozygo us famil-
ial hypercholesterolaemia: new insights and guidance for clini-
cians to improve detection and clinical management. A position
paper from the consensus panel on familial hypercholestero-
laemia of the European Atherosclerosis Society. Eur Heart J.
2014;35(32):2146-2157.
2. Nordest gaard BG, Cha pman MJ, Humph ries SE, et al.; f or
the European Atherosclerosis Society Consensus Panel.
Familial hypercholesterolaemia is underdiagnosed and
undertreated in the general population: guidance for cli-
nicians to prevent coronary heart disease: consensus state-
ment of the European Atherosclerosis Society. Eur Heart J.
2013;34(45):3478-3490.
3. WilemonKA,PatelJ,Aguilar-SalinasC,etal.Reducingtheclinical
and public health burden of familial hypercholesterolemia: a global
call to ac tion. JAMA Cardiol.2020;5(2):217-229.
4. Sturm AC,Knowles JW,GiddingSS, etal.Clinicalgenetictesting
forfamilialhypercholesterolemia:JACCscientificexpertpanel.J
Am Coll Cardiol.2018;72(6):662-680.
5. Ference BA,Ginsberg HN, GrahamI, et al. Low-densitylipopro-
teins cause atherosclerotic cardiovascular disease. 1. Evidence
from genetic, epidemiologic, and clinical studies. A consensus
statement from the European atherosclerosis society consensus
panel. Eur Heart J.2017;38(32):2459-2472.
|
11 of 13
CHEN Et al .
6. O'KeefeJHJr,Cordain L, Harris WH,MoeRM, VogelR .Optimal
low-dens itylip oproteinis50 to70mg/dl:lowe ris bet terandphys-
iologically normal. J Am Coll Cardiol. 200 4;4 3(11) :2142- 214 6.
7. LaRosaJC,GrundySM,WatersDD,etal.Intensivelipid lowering
with ator vastatin in patients with stable coronary disease. N Engl J
Med. 2005;352(14):1425- 1435.
8. SinghS, Bittner V. Familialhypercholes terolemia—epidemiology,
diagnosis, and screening. Curr Atheroscler Rep.2015;17(2):482.
9. C atapano AL, Graham I, De Backer G, et al. 2016 ESC/EAS
guidelines for the management of dyslipidaemias: the task force
for the management of dyslipidaemias of the European Society
of Cardiology (ESC) and European Atherosclerosis Society
(EAS) developed with the special contribution of the European
Assocciation for Cardiovascular Prevention & Rehabilitation
(EACPR). Atherosclerosis.2016;253:281-344.
10. Landmesser U, Chapman MJ, Stock JK, et al. 2017 update of
ESC/EAS task force on practical clinical guidance for proprotein
convert ase subtilisin/kexin type 9 inhibition in patients with ath-
erosclerotic cardiovascular disease or in familial hypercholestero-
laemia. Eur Heart J.2018;39(14):1131-1143 .
11. TomlinsonB,HuM,ChowE.Currentstatusoffamilialhypercholester-
olemia in Chinese populations. Curr Opin Lipidol. 2019;30(2):94- 100.
12. Cienfu egos JA, Pardo F, Turrión VS, e t al. Metaboli c effects of
liver replacement in homozygous familial hypercholesterolemia.
Transplant Proc.1987;19(5):3 815-3 817.
13. GrossmanM,RaderDJ,MullerDW,etal.A pilotstudyofexvivo
gene therapy for homozygous familial hypercholesterolaemia. Nat
Med.1995;1(11):1148-11 54.
14. Kosmas CE , Frishman WH. N ew and emerg ing LDL chol esterol-
lowering drugs. Am J Ther. 2015;22(3):234- 241.
15. Crick F. Central dogma of molecular biology. Nature.
1970;227(5258):561-563.
16. Opie LH. Present status of statin therapy. Trends Cardiovasc Med.
2015;25(3):216- 225.
17. Neil A, Cooper J, Betteridge J, et al.; on behalf of the Simon
Broome Familial Hyperlipidaemia Register Group. Reductions in
all- cause, cancer, and coronary mortality in statin- treated patients
with heterozygous familial hypercholesterolaemia: a prospective
registry study. Eur Heart J.2008;29(21):2625-2633.
18. IstvanES,Deisenhofer J. Structural mechanismforst atin inhibi-
tionofHMG-CoAreductase.Science. 2001;292(5519):1160- 1164.
19. VersmissenJ, Ooster veerDM,YazdanpanahM,et al. Efficacyof
statins in familial hypercholesterolaemia: a long term cohort study.
BMJ.2008;337:a2423.
20. Hardc astle SJ, Legg e E, Laundy CS , et al. Patient s' perceptions
and experiences of familial hypercholesterolemia, cascade genetic
screening and treatment. Int J Behav Med. 2015;22(1):92- 100.
21. Guan Y, Hao C, Cha DR, et al. Thiazolidinediones expand body
fluidvolumethroughPPAR gammastimulationofENaC-mediated
renal salt absorption. Nat Med.2005;11(8):861-866.
22. Ridker PM,MoraS,RoseL.PercentreductioninLDL cholesterol
following high- intensit y statin therapy: potential implications for
guidelines and for the prescription of emerging lipid- lowering
agents. Eur Heart J.2016;37(17):1373-1379.
23. GuoYL ,ZhangW,LiJJ.PC SK9andlipidloweringdrugs.Clin Chi m
Acta.2014;437:66-71.
24. Chang TY, Chang C. Ezetimibe blocks internalization of t he
NPC1L1/cholesterolcomplex.Cell Metab.2008;7(6):469-471.
25. Jia L, BettersJL ,Yu L. Niemann-pickC1-like1(NPC1L1)protein
in intestinal and hepatic cholesterol transport. Annu Rev Physiol.
2011;73:239-259.
26. HuangCS,YuX,FordstromP,etal.Cryo-EMstructuresofNPC1L1
reveal mechanisms of cholesterol transport and ezetimibe inhibi-
tion. Sci Adv.2020;6(25):eabb1989.
27. MorroneD, WeintraubWS, TothPP,et al. Lipid-alteringef ficacy
of ezetimibe plus statin and statin monotherapy and identification
of factors associated with treatment response: a pooled analysis
of over 21,000 subjects from 27 clinical trials. Atherosclerosis.
2012;223(2):251- 261.
28. Editor 's note: relates to: 'Familial hypercholesterolaemia is un-
derdiagnosed and undertreated in the general population: guid-
ance for clinicians to prevent coronary heart disease: consensus
statement ofthe European atherosclerosissociety '. Eur Heart J.
2020;41(47):4517.
29. GeyerPE,HoldtLM,TeupserD,MannM.Revisitingbiomarkerdis-
covery by plasma proteomics. Mol Syst Biol.2017;13(9):942.
30. Leren TP. Sorting an LDL receptor with bound PCSK9 to intracel-
lular degradation. Atherosclerosis.2014;237(1):76-81.
31. BerberichAJ, HegeleRA .Thecomplexmolecular geneticsof fa-
milial hypercholesterolaemia. Nat Rev Cardiol. 2019;16(1):9- 20.
32. SabatineMS.PCSK9inhibitors:clinicalevidenceandimplementa-
tion. Nat Rev Cardiol. 2019;16(3) :155- 165.
33. Durai raj A, Saba tes A, Nieves J, M oraes B, Ba um S. Proprotei n
convert ase subtilisin/Kexin type 9 (PCSK9) and it s inhibitor s: a
review of physiology, biology, and clinical data. Curr Treat Options
Cardiovasc Med.2017;19(8):58.
34. Lunven C, Paehler T, Poitiers F, et al. A randomized study of the
relative pharmacokinetics, pharmacodynamics, and safety of aliro-
cumab, a fully human monoclonal antibody to PCSK9, after single
subcutaneous administration at three dif ferent injection sites in
healthy subjects. Cardiovasc Ther.2014;32(6):297-3 01.
35. SchererN, DingsC,BöhmM,LaufsU,LehrT. Alternativetreat-
ment regimens with the P CSK9 inhibitors alirocumab and evo-
locumab: a pharmacokinetic and pharmacodynamic modeling
approach. J Clin Pharmacol.2017;57(7):846-854.
36. ShapiroMD,MilesJ,TavoriH,FazioS.Diagnosingresistancetoa
proprotein convertase subtilisin/Kexin type 9 inhibitor. Ann Intern
Med.2018;168(5):376-379.
37. Raal FJ, Rosenson RS, Reeskamp LF, et al. Evinacumab for
homozygous familial hypercholesterolemia. N Engl J Med.
2020;383(8):711-720.
38. Markham A. Evinacumab: first approval. Drugs.
2021;81(9):1101-1105.
39. Wille r CJ, Sanna S , Jackso n AU, et al. Newly i dentifie d loci that
influence lipid concentrations and risk of coronar y arter y disease.
Nat Genet.2008;40(2):161-169.
40. Musunuru K, Pirruccello JP, Do R, et al. Exome sequencing,
ANGPTL3mutations,andfamilialcombinedhypolipidemia.N Engl
J Med.2010;363(23):2220-2227.
41. TikkaA,Jauhiainen M. The role ofANGPTL3 in controllinglipo-
protein metabolism. Endocrine.2016;52(2):187-193.
42. KoishiR,AndoY,OnoM,etal.Angptl3regulateslipidmetabolism
in mice. Nat Genet.2002;30 (2):151-157.
43. Tsimikas S. RNA-targeted therapeutics for lipid disorders. Curr
Opin Lipidol.2018;29(6):459-466.
44. Goodchild J. Therapeutic oligonucleotides. Methods Mol Biol.
2011;764:1-15.
45. McCloreyG,WoodMJ.Anoverviewoftheclinicalapplicationof
antisenseoligonucleotidesforRNA-targetingtherapies.Curr Opin
Pharmacol.2015;24:52-58.
46. SullengerBA, Nair S. From the RNA world to the clinic. Science.
2016 ;352(6292) :1417-1420.
47. B urnett JC , Rossi JJ. RNA-based t herapeuti cs: current pr ogress
and future prospects. Chem Biol.2012;19(1):60-71.
48. PrakashTP,GrahamMJ,YuJ,etal.Targeteddeliveryofantisense
oligonu cleotides to h epatocy tes using tr iantennar y N-a cetyl ga-
lactosamine improves potency 10- fold in mice. Nucleic Acids Res.
2014;42(13):8796-8807.
49. Nordestgaard BG , Nicholls SJ, Langsted A, Ray KK, Tybjærg-
Hansen A. Advances in lipid- lowering therapy through gene-
silencing technologies. Nat Rev Cardiol.2018;15(5):261-272.
12 of 13
|
CHEN Et al.
50. Benne tt CF,S wayze EE. RNA tar geting therap eutics: mole cular
mechanisms of antisense oligonucleotides as a therapeutic plat-
form. Annu Rev Pharmacol Toxicol. 2010;5 0:259- 293.
51. Wierzbicki AS, Viljoen A. Anti- sense oligonucleotide thera-
pies for the treatment of hyperlipidaemia. Exp ert Opin Biol Ther.
2016;16(9):1125- 1134.
52. LeeRG,Crosby J, Baker BF,Graham MJ, Crooke RM. Antisense
technology: an emerging platform for cardiovascular disease ther-
apeutics. J Cardiovasc Transl Res.2013;6(6):969-980.
53. CrookeST,WitztumJL,BennettCF,BakerBF.RNA-targetedther-
apeutics. Cell Metab.2018;27(4):714-739.
54. WuH,MacLeodAR,LimaWF,CrookeST.Identificationandpartial
purificationofhumandoublestrandRNaseactivity.Anoveltermi-
nating mechanism for oligoribonucleotide antisense drugs. J Biol
Chem.1998;273(5):2532-2542.
55. BellDA ,HooperAJ,Burnett JR. Mipomersen, an antisense apo-
lipoprotein B synthesis inhibitor. Exper t Opin Investig Drugs.
2011;20(2):265-272.
56. Rader DJ,Kastelein JJ.Lomitapideandmipomersen: twofirst-in-
class drugs for reducing low- density lipoprotein cholesterol in pa-
tients with homozygous familial hypercholesterolemia. Circulation.
2014;129(9):1022- 1032 .
57. Ylä-Her ttuala S,SolakiviT,HirvonenJ,etal.Glycosaminoglycans
and apolipoproteins B and A- I in human aortas. Chemical and
immunological analysis of lesion- fre e aortas from children and
adults. Arteriosclerosis.1987;7(4):333-34 0.
58. Borén J, Olin K, Lee I, Chait A, Wight TN, Innerarity TL .
Identification of the principal proteoglycan- binding site in LDL. A
single- point mutation in apo- B100 severely af fects proteoglycan
interaction without affecting LDL receptor binding. J Clin Invest.
1998;101(12):2658-2664.
59. Schonfeld G, Lin X, Yue P. Familial hypobet alipoproteinemia: ge-
netics and metabolism. Cell Mol Life Sci.2005;62(12):1372-1378.
60. Altma nn KH, Fabbro D, D ean NM, et al . Second-gene ration an-
tisense oligonucleotides: structure- activity relationships and the
design of improved signal- transduction inhibitors. Biochem Soc
Trans.1996;24(3):630-637.
61. Yu RZ, Kim TW, Hong A, Watanabe TA, Gaus HJ, Geary RS.
Cross- species pharmacokinetic comparison from mouse to man
of a second- generation antisense oligonucleotide, ISIS 301012,
targeting human apolipoprotein B- 100. Drug Metab Dispos.
2007;35(3):460-468.
62. Reyes-SofferG,MoonB,Hernandez-OnoA,etal.Complexeffects
of inhibiting hepatic apolipoprotein B100 synthesis in humans. Sci
Transl Med.2016;8(323):323ra12.
63. Li N, Li Q, Tian XQ, Qian HY,Yang YJ.Mipomersenisa promis-
ing therapy in the management of hypercholesterolemia: a meta-
analysis of randomized controlled trials. Am J Cardiovasc Drugs.
2014;14(5) :367-376 .
64. ParhamJS,GoldbergAC.Mipomersenanditsuseinfamilialhyper-
cholesterolemia. Expert Opin Pharmacother.2019;20(2):127-131.
65. RaalFJ,SantosRD,BlomDJ,etal.Mipomersen,anapolipoprotein
B synthesis inhibitor, for lowering of LDL cholesterol concentra-
tions in patients with homozygous f amilial hypercholesterolae-
mia: a randomised, double- blind, placebo- controlled trial. Lancet.
2010;375(9719):998-10 06.
66. DuellPB,SantosRD,KirwanBA,Wit ztumJL,TsimikasS,Kastelein
JJP.Long-termmipomersentreatmentisassociatedwithareduc-
tion in cardiovascular events in patients with familial hypercholes-
terolemia. J Clin Lipidol. 2016;10(4):1011- 1021.
67. Santos RD,DuellPB, EastC,et al.Long-termefficacyandsafety
of mipomersen in patients with familial hypercholesterolaemia:
2- year interim results of an open- label extension. Eur Hear t J.
2015;36(9):566-575.
68. Dewey FE,GusarovaV,DunbarRL,et al.Geneticandpharmaco-
logicinactivationofANGP TL3andcardiovasculardisease.N Engl J
Med.2017;377(3):211-221.
69. Crooke ST, Baker BF, Xia S, et al. Integrated assessment of the
clinicalperformanceofGalNAc(3)-conjugated2'-O-methoxyethyl
chimeric antisense oligonucleotides: I. Human volunteer experi-
ence. Nucleic Acid Ther. 2019;29(1):16- 32.
70. GaudetD,Karwatowska-ProkopczukE,BaumSJ,etal.Vupanorsen,
anN-acetylgalactosamine-conjugatedantisensedrugtoANGPTL3
mRNA , lowers triglyce rides and ather ogenic lipoprot eins in pa-
tients with diabetes, hepatic steatosis, and hypertriglyceridaemia.
Eur Heart J. 2020;41(40):3936- 3945.
71. AhmadZ,Banerjee P,HamonS, et al. Inhibitionof angiopoietin-
like protein 3 with a monoclonal antibody reduces triglycerides in
hypertriglyceridemia. Circulation.2019;140(6):470-486.
72. RomeoS,YinW,KozlitinaJ,etal.Rareloss-of-functionmutations
inANGPTLfamilymemberscontributetoplasmatriglyceridelev-
els in humans. J Clin Invest.2009;119(1):70-79.
73. Robciuc MR,MaranghiM,LahikainenA ,etal.Angptl3deficiency
is associated with increased insulin sensitivity, lipoprotein lipase
activity, and decreased serum free fatty acids. Arterioscler Thromb
Vasc Biol.2013;33(7):1706-1713.
74. GrahamMJ,LeeRG,Brandt TA, etal.Cardiovascularandmeta-
bolic effects of ANGPTL3 antisense oligonucleotides. N Engl J
Med.2017;377(3):222-232.
75. A grawalN,DasaradhiPV,MohmmedA,MalhotraP,BhatnagarRK,
MukherjeeSK.RNAinterference:biology,mechanism,andappli-
cations. Microbiol Mol Biol Rev.2003;67(4):657-685.
76. Martinez J, Patkaniowska A, Urlaub H, Lührmann R, Tuschl T.
Single- str anded antis ense siRNAs g uide target R NA cleavage in
RNAi.Cell.2002;110(5):563-574.
77. Rand TA, Petersen S, Du F, Wang X. Argonaute2 cleaves
the anti-guide strand of siRNA during RISC activation. Cell.
2005;123 (4):621- 629.
78. CarthewRW,SontheimerEJ.OriginsandmechanismsofmiRNAs
andsiRNAs.Cell. 20 09;136(4):642- 655.
79. BernardsR.ExploringtheusesofRNAi—geneknockdownandthe
nobel prize. N Engl J Med. 2006;355(23):2391- 2393.
80. Dyrbuś K, Gąsior M, Penson P, Ray KK, Banach M. Inclisiran-
new hope in the management of lipid disorders? J Clin Lipidol.
2020;14(1):16-27.
81. Fitzgerald K, Frank-Kamenetsk y M, Shulga-Morskaya S, et al.
Effec t of an RNA inter ference d rug on the sy nthesis of pr opro-
tein conver tase subtilisin/kexin t ype 9 (PCSK9) and the con-
centration of serum LDL cholesterol in healthy volunteers: a
randomised, single- blind, placebo- controlled, phase 1 trial. Lancet.
2014;383(9911):60-68.
82. Ray KK, Wright RS, Kallend D, et al. Two phase 3 trials of
Inclisir an in patient s with elevated LDL choles terol. N Engl J Med.
2020;382(16):1507-1519.
83. KosmasCE,DeJesusE,MorceloR, GarciaF,Mont anPD,Guzman
E. Lipid- lowering interventions targeting proprotein convertase
subtilisin/kexin type 9 (PCSK9): an emerging chapter in lipid-
lowering therapy. Drugs Context.2017;6:212511.
84. FitzgeraldK ,WhiteS,BorodovskyA,etal.AhighlydurableRNAi
therapeutic inhibitor of PCSK9. N Engl J Med.2017;376(1):41-51.
85. Khvorova A. Oligonucleotide therapeutics—a new class of
cholesterol- lowering drugs. N Engl J Med.2017;376(1):4-7.
86. Leiter L A, Teoh H, Ka llend D, et al. In clisiran lo wers LDL-C a nd
PCSK9 irrespective ofdiabetesstatus: theORION-1 randomized
clinical trial. Diabetes Care.2019;42(1):173-176.
87. LambYN.Inclisiran:firstapproval.Drugs.2021;81(3):389-395.
88. Watts GF, Schwabe C, Scott R, et al. RNAi inhibition of
angiopoietin-like protein 3 (ANGPTL3) with ARO-ANG3 mimics
the lipid and lipoprotein profileoffamilial combined hypolipidemia.
Eur Heart J. 2020;41:ehaa946.3331.
89. Wat tsGF,SchwabeC,ScottR, etal. Pharmacodynamiceffectof
ARO-ANG3,aninvestigationalRNAinterferencetargetinghepatic
angiopoietin- like protein 3, in patients with hypercholesterolemia.
Circulation.2020;142:A15751.
|
13 of 13
CHEN Et al .
90. Alves AC, Azevedo S, Benito- Vicente A , et al. LDLR variants
functional characterization: contribution to variant classification.
Atherosclerosis. 2021;329:14- 21.
91. Defesche JC, Gidding SS, Harada-Shiba M, Hegele R A, Santos
RD, Wierzbicki AS. Familial hypercholesterolaemia. Nat Rev Dis
Primers.2017;3:17093.
92. McG owan MP, Hosseini Dehkordi SH, Moriarty PM, Duell PB.
Diagnosis and treatment of heterozygous familial hypercholester-
olemia. J Am Heart A ssoc.2019;8(24):e013225.
93. Lagace TA, Curtis DE, Garuti R, et al. Secreted PCSK9 decreases
the number of LDL receptors in hepatoc ytes and in livers of para-
biotic mice. J Clin Invest. 2006;116(11):2995- 3005.
94. AjufoE, Cuchel M.Recentdevelopments ingenetherapyforho-
mozygous familial hypercholesterolemia. Curr Atheroscler Rep.
2016;18(5):22.
95. Raper SE, GrossmanM, RaderDJ, et al. Safety and feasibility of
liver- direc ted ex vivo gene therapy for homozygous familial hyper-
cholesterolemia. Ann Surg. 1996;223(2):116- 126.
96. HajighasemiS,Mahdavi GorabiA ,Bianconi V,et al. A review of
gene- and cell- based therapies for familial hypercholesterolemia.
Pharmacol Res. 2 019;143 :119- 132.
97. KerrAG,TamLC,HaleAB, etal.Episomalnonviralgenether-
apy vectors slow progression of atherosclerosis in a model
of familial hypercholesterolemia. Mol Ther Nucleic Acids.
2016;5(11):e383.
98. GaoG,AlviraMR, SomanathanS,etal.Adeno-associatedviruses
undergo substantial evolution in primates during natural infec-
tions. Proc Natl Acad Sci U S A.2003;10 0(10):6081-6086.
99. Gao G, Van denberghe LH , Wilson JM. New re combinant sero-
types of AAV vectors. Curr Gene Ther.2005;5(3):285-297.
100. Kassim SH, Li H, Bell P, et al. Adeno- associated virus serotype
8 gene therapy leads to significant lowering of plasma choles-
terol levels in humanized mouse models of homozygous and
heterozygous familial hypercholesterolemia. Hum Gene Ther.
2013;24(1):19- 26.
101. Lisowski L, Dane AP, Chu K, et al. Selection and evaluation of clin-
ically relevant AAV variants in a xenograft liver model. Nature.
2014;506(7488):382-386.
102. NathwaniAC, Reiss UM, Tuddenham EG,etal.Long-termsafety
and efficacy of factor IX gene therapy in hemophilia B. N Engl J
Med.2014;371(21):1994-2004.
103. Kolansky DM, Cuchel M, Clark BJ, et al. Longitudinal evalu-
ation and assessment of c ardiovascular disease in patients
with homozygous familial hypercholesterolemia. Am J Cardiol.
2008;10 2(11):1438-14 43.
104. Greig JA, LimberisMP,BellP,et al.Non-clinicalstudyexamining
AAV8.TBG.hLDLR vector-associated toxicity in Chow-fed wild-
type and LDLR(+/−). Rhesus Macaques Hum Gene Ther Clin Dev.
2017;28(1):39-50.
105. G reig JA, Limbe ris MP, Bell P, et al. Noncl inical pharma cology/
toxicologystudyofAAV8.TBG.mLDLRandAAV8.TBG.hLDLRina
mouse model of homozygous familial hyperchole sterolemia. Hum
Gene Ther Clin Dev.2017;28(1):28-38.
106. SanderJD, JoungJK.CRISPR-Cas systems for editing,regulating
and targeting genomes. Nat Biotechnol.2014;32(4):347-355.
107. Zhao Y, Yang Y, Xing R, et al. Hyperlipidemia induces typical
atherosclerosis development in Ldlr and Apoe deficient rats.
Atherosclerosis.2018;271:26-35.
108. LuR,Yuan T,WangY,et al.Spontaneousseverehypercholester-
olemia and atherosclerosis lesions in rabbits with deficiency of
low-densit ylipoprotein receptor (LDLR) onexon7.EBioMedicine.
2018;36:29-38.
109. JarrettKE, LeeC,DeGiorgiM, etal. Somatic editing ofLdlrwith
adeno- associated viral- CRISPR is an efficient tool for atherosclero-
sis research. Arterioscler Thromb Vasc Biol.2018;38(9):1997-2006.
110. ZhangL,WangL,XieY,etal.Triple-targetingdeliveryofCRISPR/
Cas9 to reduce the risk of cardiovascular diseases. Angew Chem Int
Ed Engl.2019;58(36):12404-12408.
111. Ding Q, Strong A , Patel KM, et al. Permanent alteration of
PCSK9 with in vivo CRISPR- Cas9 genome editing. Circ Res.
2 0 1 4 ; 1 1 5 ( 5 ) : 4 8 8 - 4 9 2 .
112. O kada H, Naka nishi C, Yoshida S, et a l. Function and i mmuno-
genicit y of gene- corrected iPSC- derived hepatocyte- like cells in
restoring low densit y lipoprotein uptake in homozygous familial
hypercholesterolemia. Sci Rep. 2019;9(1):4695.
113. Jalil S, Keskinen T, Maldonado R, et al. Simultaneous high-
efficiency base editing and reprogramming of patient fibroblasts.
Stem Cell Rep.2021;16(12):3064-3075.
114. ZhaoH,LiY,HeL,etal.InvivoAAV-CRISPR/Cas9-mediatedgene
editing ameliorates atherosclerosis in familial hypercholesterol-
emia. Circulation.2020;141(1):67-79.
115. CastelSE,MartienssenRA .RNAinterferenceinthenucleus:roles
forsmallRNAsintranscription,epigenetics andbeyond.Nat Rev
Genet. 2013;14(2):100- 112.
116. van Poelgeest EP, Hodges MR , Moerland M, et al. Antisense-
mediated reduction of proprotein convertase subtilisin/kexin type
9 (PCSK9): a f irst- in- human randomized, placebo- controlled trial.
Br J Clin Pharmacol.2015;80(6):1350-1361.
117. LanphierE,UrnovF, Haecker SE, WernerM,Smolenski J. Don't
edit the human germ line. Nature.2015;519(7544):410- 411.
118. Nishiga M, QiLS, WuJC .Therapeuticgenomeeditingincardio-
vascular diseases. Adv Drug Deliv Rev.2021;168:147-157.
119. Cyranoski D, Ledford H. Genome- edited baby claim provokes in-
ternational outcry. Nature.2018;563(7733):607-608.
120. RamanathanS,ShenodaBB,LinZ,etal.Inflammationpotentiates
miR- 939 expression and packaging into small extracellular vesi-
cles. J Extracell Vesicles.2019;8(1):1650595.
121. Aqil F, Munagala R, Jeyabalan J, et al. Milk exosomes—natural
nanoparticlesforsiRNAdelivery.Cancer Lett.2019;449:186-195.
122. LiZ,ZhaoP,ZhangY,etal.Exosome-basedLdlr gene therapy for
familial hypercholesterolemia in a mouse model. Theranostics.
2021;11(6):2953- 2965.
123. BlomDJ,Harada-ShibaM,RubbaP,etal.Efficacyandsafet yofali-
rocumab i n adults with h omozygous fam ilial hypercho lesterolemi a:
the ODYSSEY HoFH trial. J A m Coll Cardiol.2020;76(2):131-142.
124. KorenMJ,SabatineMS,GiuglianoRP,etal.Long-termeffic acyand
safety of evolocumab in patients with hypercholesterolemia. J Am
Coll Cardiol.2019;74(17):2132-214 6.
125. BergmarkBA, MarstonNA ,Bramson CR, et al. Effec tof vupan-
orsen on non- high- density lipoprotein cholesterol levels in statin-
treatedpatientswith elevatedcholesterol:TRANSLATE-TIMI 70.
Circulation.2022;145(18):1377-1386.
126. Ying Q, Chan DC, Wat ts GF. Angiopoietin- like protein 3 inhibitors
and contemporary unmet needs in lipid management. Curr Opin
Lipidol. 2021;32(3):210- 212.
How to cite this article: Chen R, Lin S, Chen X. The promising
novel therapies for familial hypercholesterolemia. J Clin Lab
Anal. 2022;36:e24552. doi: 10.10 02/jcla. 24552
Available via license: CC BY
Content may be subject to copyright.