You are on page 1of 26

Advanced Drug Delivery Reviews 159 (2020) 94–119

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews

journal homepage: www.elsevier.com/locate/addr

Interaction between high-density lipoproteins and inflammation:


Function matters more than concentration!
Sumra Nazir a,b, Vera Jankowski a, Guzide Bender a,b, Stephen Zewinger c,d,
Kerry-Anne Rye e, Emiel P.C. van der Vorst a,b,f,g,h,⁎
a
Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
b
Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
c
Department of Internal Medicine 4, Nephrology and Hypertension, Saarland University Medical Centre, Homburg, Germany
d
Department of Nephrology, Hôpitaux Robert Schuman, Hôpital Kirchberg, Luxembourg, Luxembourg
e
Lipid Research Group, School of Medical Sciences, The University of New South Wales Sydney, New South Wales, Australia
f
Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
g
Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
h
German Centre for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany

a r t i c l e i n f o a b s t r a c t

Article history: High-density lipoprotein (HDL) plays an important role in lipid metabolism and especially contributes to the re-
Received 6 January 2020 verse cholesterol transport pathway. Over recent years it has become clear that the effect of HDL on immune-
Received in revised form 20 September 2020 modulation is not only dependent on HDL concentration but also and perhaps even more so on HDL function.
Accepted 13 October 2020
This review will provide a concise general introduction to HDL followed by an overview of post-translational
Available online 17 October 2020
modifications of HDL and a detailed overview of the role of HDL in inflammatory diseases. The clinical potential
Keywords:
of HDL and its main apolipoprotein constituent, apoA-I, is also addressed in this context. Finally, some conclu-
High-density lipoproteins sions and remarks that are important for future HDL-based research and further development of HDL-focused
Inflammation therapies are discussed.
Metabolic diseases © 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
Infectious diseases (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Auto-immune diseases
Neurological diseases
Chronic kidney disease

Contents

1. General introduction and aim of this review . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95


2. Introduction of the HDL metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
2.1. Discovery of HDL particle(s) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
2.2. HDL structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
2.3. HDL biogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
3. Post translational modifications of apoA-I and HDL . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
3.1. Oxidation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
3.2. Carbamylation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
3.3. Glycation and advanced glycation end products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
4. HDL and immune-mediated diseases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
4.1. Metabolic diseases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
4.1.1. Cardiovascular disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
4.1.2. Diabetes mellitus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
4.1.3. Liver disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
4.1.4. Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104

⁎ Corresponding author at: Pauwelstraße 30, 52074 Aachen, Germany.


E-mail address: evandervorst@ukaachen.de (E.P.C. van der Vorst).

https://doi.org/10.1016/j.addr.2020.10.006
0169-409X/© 2020 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

4.2. Infectious diseases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104


4.2.1. Bacterial, viral and parasitic infections. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
4.2.2. Sepsis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
4.3. Auto-immune diseases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
4.3.1. Rheumatoid arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
4.3.2. Systemic lupus erythematosus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
4.3.3. Psoriasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
4.3.4. Inflammatory bowel disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
4.4. Neurological disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
4.4.1. Effects of HDL on the pathogenesis of neurological disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
4.4.2. Clinical potential of apoA-I and HDL in neurological disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
4.5. Chronic kidney disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
4.5.1. Effects of HDL on the pathogenesis of chronic kidney disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
4.5.2. Clinical potential of apoA-I and HDL in chronic kidney disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
4.6. Other diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
5. Discussion and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
Sources of funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110

1. General introduction and aim of this review apolipoproteins apoA-I and apoA-II [11,12]. Due to differences in apoli-
poproteins, phospholipids, cholesterol and cholesteryl esters, HDLs
High-density lipoprotein (HDL) comprises of a wide range of parti- have differences in surface charge and density and can thereby be clas-
cles, varying in size, composition and density. HDL particles can be sep- sified into γ-, α- or preβ-migrating particles or HDL2 and HDL3, respec-
arated from other lipoproteins, based on their unique physicochemical tively [13,14].
properties. Gofman et al., identified the first clinically beneficial role of
HDL, demonstrating that low levels of HDL were positively associated 2.2. HDL structure
with the development of atherosclerosis [1]. Over the years, this inverse
correlation between the risk for developing coronary heart disease and HDL particles are heterogeneous with respect to charge, density,
the levels of HDL cholesterol (HDL-C) has been widely confirmed, sug- size, and shape, chemical and physiologic properties [15]. In their ma-
gesting a protective role of HDL-C in atherosclerosis development [2]. ture form, HDL contains a nonpolar lipid core (mainly consisting of
However, several recent investigations, including many clinical trials fo- cholesteryl esters and triglycerides) surrounded by a single surface
cusing on raising HDL-C levels that did not show any beneficial effect on monolayer of phospholipids (mainly phosphatidylcholine), unesterified
cardiovascular event rates [3,4], started a lively debate about the appro- cholesterol and amphipathic apolipoproteins (Fig. 1). Based the pres-
priateness of HDL as a therapeutic target. This debate was further fueled ence of absence of cholesteryl esters, HDL has either a discoidal or a
by Mendelian randomization studies, which demonstrated that poly- spherical shape. The density of HDL varies from 1.063–1.21 g/ml, with
morphisms that exclusively alter HDL-C levels did not affect the risk of a size-range of 8-10 nm [16,17]. HDL can be further subdivided into
myocardial infarction (MI) [5]. This led to the suggestion that the func- larger-sized, lower-density (HDL2a and HDL2b) and smaller-sized,
tionality of HDL, that is determined by its composition and post- higher-density (HDL3a, HDL3b, and HDL3c) sub-classes [18]. Apolipopro-
translational modifications are more important determinants for pro- teins associated with HDL can undergo post-translational modifications
tection against cardiovascular disease (CVD) than the levels of HDL-C as will be discussed in more detail later. The major apolipoprotein con-
[6,7]. Over the last years, the question whether HDL function is more im- stituents of HDL are apoA-I (70%) and apoA-II (20%), with small
portant than HDL-C has also been investigated in other pathologies, es- amounts of other apoproteins such as apoA-IV, apoA-V, apoC and apoE
pecially since it has been shown that HDL influences immune cell [19]. Additionally, HDL has been shown to contain a lot more proteins
reactivity in inflammatory and infectious diseases. In this review these (>80) like the lipocalins apoD and apoM [20].
interactions will be discussed as well as the importance of HDL function
vs concentration in the context of pathogenesis and clinical application. 2.3. HDL biogenesis

2. Introduction of the HDL metabolism HDL formation is initiated by the secretion of lipid free apoA-I by the
liver (comprising about 70% of total apoA-I production) [21] and the in-
2.1. Discovery of HDL particle(s) testine (comprising about 30% of total apoA-I production) [22]. This
lipid free apoA-I interacts with the ATP-binding cassette transporter
“HDL” was discovered in 1929 when a lipid rich α-globulin was iso- A1 (ABCA1) on peripheral cells, leading to the transfer of cholesterol
lated from horse serum by Macheboeuf at the Pasteur Institute in Paris and cellular phospholipids from the cell membrane to apoA-I (Fig. 2).
[8]. Several years later, lipoproteins were also isolated from human Through a series of intermediate steps, the lipidated apoA-I is progres-
plasma using density gradient ultra-centrifugation [9]. The use of repet- sively converted into discoidal pre-β-migrating HDL particles that con-
itive ultra-centrifugations was a big breakthrough as it was shown that sist of a phospholipid bilayer surrounded by two apoA-I molecules/
raising the serum density by the addition of concentrated salt solutions particle [23]. Subsequently, the enzyme lecithin:cholesterol acyltrans-
allows the flotation and isolation of three different lipoprotein fractions ferase (LCAT) mediates the esterification of cholesterol, converting dis-
with densities ranging from <1.019 g/ml, 1019–1.063 g/ml, and coidal pre-β-migrating HDL particles into α-migrating, spherical HDL
1.063–1.21 g/ml [10]. This results in the classification of these lipopro- particles with a central core of cholesteryl esters [24] and two molecules
tein fractions, defined as very low density lipoprotein (VLDL), low of apoA-I. Small spherical HDL subsequently acquire additional free cho-
density lipoprotein (LDL) and HDL [10]. Over the years, more lesterol that effluxes from peripheral cells via ABCA1 and ABCG1. Ester-
detailed analysis of HDL revealed the existence of two major HDL ification of this newly acquired cholesterol by LCAT generates large HDL

95
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

Fig. 1. Generalized structure of spherical HDL. High-density-lipoproteins particles consist of a core of triglycerides and cholesteryl esters. This core is surrounded by a phospholipid
monolayer containing unesterified cholesterol and several apoproteins (mostly apoA-I).

Fig. 2. General overview of HDL metabolism. ApoA-I is produced in the liver and intestines and subsequently forms discoidal HDL after interacting with the ATP-binding cassette
transporter A1 (ABCA1). Continued cholesterol export via ABCG1 (and via scavenger receptor class B member 1 (SR-B1) in macrophages) and enzymatic reactions mediated by
lecithin-cholesterol acyltransferase (LCAT) result in the conversion into spherical high-density-lipoproteins (HDL)3 and HDL2 particles. Cholesterol and cholesteryl esters can be
transferred to the liver by two main pathways, directly through binding to SR-B1 in the liver and subsequent excretion via the kidneys, or indirectly by being transferred to very-low-
density lipoproteins (VLDL), intermediate-density-lipoproteins (IDL) and low-density lipoproteins (LDL) by cholesteryl ester transfer protein (CETP), followed by uptake of these
lipoproteins via the LDL receptor (LDLR) in hepatocytes. Additionally, HDL can be catabolized in the liver via its uptake as holoparticles through a yet to be identified HDL receptor
(HDLR). HDL2 can be remodeled by processes that are catalyzed by hepatic lipase (HL), endothelial lipase (EL) and plasma phospholipid transfer protein (PLTP), resulting in the
formation of lipid-poor HDL particles which again interact with ABCA1 for a next lipidation cycle.

96
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

particles with three or more apoA-I molecules/particle. Furthermore, 3.2. Carbamylation


phospholipid transfer protein (PLTP) plays a vital role in the remodel-
ling of HDL, by transferring phospholipids from chylomicrons and Carbamylation is a non-enzymatic and irreversible PTM, catalyzed
VLDL to HDL [25]. This transfer of phospholipids can destabilize HDL, by urea and MPO, that results from the interaction between isocyanic
leading to fusion and the formation of larger α-migrating HDL particles acid and amino groups of proteins. Various studies have confirmed
[26]. Spherical HDL particles can also undergo modification through bi- that apoA-I can be carbamylated and that this is associated with CVD,
directional exchange of cholesteryl esters and triglycerides with apoB with higher levels of apoA-I carbamylation being observed in human
containing lipoproteins (VLDL and LDL) mediated by cholesteryl ester atherosclerotic plaques compared to normal arterial tissues [48]. There-
transfer protein (CETP). As CETP predominately supports the transfer fore, carbamylation of HDL-associated apolipoproteins might be one of
of cholesteryl esters from HDL towards apoB containing lipoproteins the mechanisms linking inflammation and CVD.
in exchange for triglycerides, this process will result in triglyceride–
rich HDL [27,28]. The exact mechanism of the HDL-CETP interactions re- 3.3. Glycation and advanced glycation end products
mains part of active research, as some studies suggest that CETP bind to
HDL by hydrophobic interaction whereas others suggest it is rather a Advanced glycation end products (AGEs) are formed when a non-
protein-protein interactions [29]. The formation of a hydrophobic chan- enzymatic reaction takes place between a protein and reducing sugars.
nel by SR-B1, allowing cholesterol diffusion from HDL into the plasma These compounds have been associated with the development of vari-
membrane, can further remodel HDL [30]. Additionally, hepatic lipase ous cardiovascular disorders such as endothelial dysfunction, increased
can hydrolyse HDL phospholipids and triglycerides, driving the transi- myocardial and vascular stiffness, diastolic dysfunction, and the forma-
tion of HDL2 into HDL3 particles [31]. This remodelling is accompanied tion of atherosclerotic lesions [49]. Non-enzymatic glycation of apoA-I
by the dissociation of lipid-free or lipid-poor apoA-I, while endothelial adversely affects the metabolism of HDL and impairs the ability of
lipase remodels HDL without such dissociation [32]. In the kidney, this LCAT to esterify cholesterol [50]. This could be explained by the observa-
lipid-free apoA-I undergoes glomerular filtration and can subsequently tion that modifications of lysine, arginine and tryptophan residues me-
either be degraded, excreted through the urine or partially reabsorbed, diated by methylglyoxal altered the conformation of an epitope of
thereby regulating the plasma levels of apoA-I and HDL [33,34]. apoA-I in regions required for LCAT activation and binding of lipids
[50]. Glycation of apoA-I also impairs the anti-inflammatory properties
of HDL and reduces its ability to inhibit expression of intercellular adhe-
sion molecule (ICAM-1) and vascular cell adhesion molecule (VCAM-1)
3. Post translational modifications of apoA-I and HDL
on endothelial cells, and its ability to reduce neutrophil infiltration into
the intima of carotid arteries [51]. Combined with the recent observa-
Here, three well-described Post translational modifications (PTMs)
tion that glycation of apoA-I reduces its half-life by threefold [52],
of apoA-I, being oxidation, carbamylation and glycation will be briefly
these studies clearly demonstrate the important detrimental effects of
highlighted.
non-enzymatic glycation on the anti-inflammatory functions of HDL.

4. HDL and immune-mediated diseases


3.1. Oxidation
It has been shown that HDL plays an important role in a wide-range
Oxidation of HDL takes predominantly place in inflammatory micro- of immune cells, inflammatory and infectious diseases. In this chapter,
environments [35]. For example, activated phagocytes secrete superox- the role of HDL in various pathologies will be described, especially to
ide anion radical (O− 2 ), H2O2 and myeloperoxidase (MPO) which all elucidate the role of HDL concentration vs. function. Besides the role
have oxidative capacities [36]. The clinical relevance of such modifica- of HDL in the pathophysiology, also the clinical potential of HDL therapy
tions has been shown by a study that demonstrated that the apoA-I in will be discussed for the specific diseases.
patients with cardiovascular disease contains a higher degree of MPO-
dependent oxidative modifications (nitration and chlorination) [37]. 4.1. Metabolic diseases
These modifications impair the lipid binding potential of apoA-I and
thereby result in a dose-dependent loss of ABCA1-mediated cholesterol 4.1.1. Cardiovascular disease
efflux from macrophages [37]. More recent studies additionally demon-
strate that (patho) physiologically relevant levels of MPO also results 4.1.1.1. Effects of HDL on the pathogenesis of cardiovascular disease. CVD is
in the loss of the anti-apoptotic and anti-inflammatory functions still the major cause of hospitalization and mortality worldwide. The
of HDL [38]. main underlying cause of CVD is atherosclerosis, a chronic inflammatory
In addition, sulfur oxidation of the methionine residues in apoC-I, C- disease of the arteries that develops over time. It has been demonstrated
III and A-I of HDL regulates the activity of these proteins [39,40]. For ex- that a multitude of immune cells play an important role in atherosclero-
ample, methionine oxidation of apoA-I caused partial unfolding of the sis [53]. Here we will limit ourselves to the description of four main cell-
protein, decreased its stability and reduced the formation of apoA-I di- types that are of paramount importance in this pathology and have been
mers and tetramers [41]. Methionine sulfoxidation of apoA-I causes demonstrated to interact with HDL (Fig. 3).
HDL to become pro-inflammatory, as it induces pro-inflammatory cyto- 4.1.1.1.1. Macrophages. Macrophages, the most essential immune
kine secretion (TNFα and IL-6) in mouse bone marrow-derived macro- cells in various CVDs, have recently been highlighted in a seminal re-
phages as well as mouse monocytes [42]. view series [54–57]. During atherosclerosis, lesional macrophages accu-
Furthermore, tryptophan residues were identified as major oxida- mulate large amounts of cholesterol esters forming foam cells. Since
tion sites in proteins because of the high reactivity of sulfur atoms and HDL can only remove unesterified cholesterol from macrophages it is
aromatic rings towards various reactive oxygen species causing a signif- important that intracellular cholesterol ester hydrolyses occurs [58].
icant reduction in the viability of tryptophan of the organism [43,44]. In Macrophages comprise a heterogeneous population of innate myeloid
addition, it was recently shown that tryptophan oxidation of apoA-I oc- cells that acclimatise in response to a variety of signals from the
curs during inflammation and is characterized by its pro-inflammatory micro-environment. For example, stimulation by interferon gamma
properties on endothelial cells and impairs the ABCA1-dependent HDL (IFN-γ) or lipopolysaccharide (LPS) induces classically activated, pro-
biogenesis [45]. Tryptophan oxidation has been recently extensively inflammatory M1 macrophages. In contrast, non-classically activated
reviewed in [46,47]. M2 macrophages are induced, for example, by IL-4 or IL-13 and are

97
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

Fig. 3. Overview of selected Immune-HDL crosstalk mechanisms in atherosclerosis. Increasing HDL levels has several effects on immune cells that play a crucial role in atherosclerosis
development. Although some of these effects are still rather contradictory, it is clear that there are significant interactions between HDL and the described immune cells in this pathology.

known to decrease inflammation by secreting IL-10 [59]. A more elabo- cholesterol levels, which mediate IRE1a/ASK1/p38 MAPK activation and
rate review focusing on the understanding of macrophage heterogene- endoplasmic reticulum (ER) stress. Although further studies using differ-
ity in the context of atherosclerosis has recently been published by ent HDL preparations with varying composition are still needed to fully
Nagenborg et al. [59]. elucidate how these seemingly contradictory findings can be explained,
HDL has been linked to lesional macrophage polarization. Increasing it indicates that HDL composition plays an important role in how HDL in-
the levels of HDL by overexpression of human apoA-I in mice not only fluences inflammatory responses in macrophages.
decreased the size of atherosclerotic lesions and lesion lipid content, Several other observations stress the importance of HDL function,
but also changed the phenotypic polarization of monocyte-derived rather than HDL-C concentration, on inflammation in macrophages.
cells (predominantly macrophages) from an M1 to M2 phenotype [2]. For example, apoA-I and HDL have been shown to exert their
However, the effects of HDL on macrophage inflammation turned out anti-inflammatory effects in macrophages by stimulating cholesterol ef-
to be not so straightforward. Several studies over the last decade evalu- flux via ABCA1 and ABCG1and attenuation of inflammatory TLR-
ated the effects of HDL and apoA-I on macrophage inflammation with signalling [66]. ApoA-I and HDL not only extract intracellular cholesterol
seemingly contradicting results [60–63]. One of the studies showed from the cells but also cholesterol which is present in the cell membrane
that HDL exerts anti-inflammatory effects by down regulating type 1 in- in lipid rafts. Lipid rafts are specific regions in the membrane which are
terferon responses in macrophages downstream of TRAM/TRIF signal- enriched in glycosphingolipids, cholesterol and protein receptors [67].
ling [62]. These inhibitory effects of HDL on IFN signalling were Signalling of many TLRs is highly dependent on the presence of
confirmed in a later study in which the anti-inflammatory effects of cholesterol in lipid rafts, and this can be modulated by apoA-I or HDL.
HDL (CSL-111, reconstituted HDL) are attributed to the induction of For example, macrophages deficient in ABCA1 display enlarged
Atf3, a transcriptional repressor, expression in macrophages [60]. cholesterol-rich lipid rafts with high TLR4 expression and an elevated
Additionally, It has been reported that apoA-I may also mediate response to stimulation by LPS [68–70]. Similarly, in monocyte-
anti-inflammatory effects by modulating Toll-like receptor (TLR) derived macrophages treated with LPS, the apoA-I mimetic peptide 4F
4-dependent signalling in macrophages [64]. down-regulates the expression of several TLRs by reducing the choles-
In sharp-contrast to the previously described studies, it has been terol content of lipid rafts, which reduces the expression of various
shown that HDL can have pro-inflammatory effects in macrophages. inflammatory genes [71,72].
These effects seem to be mediated by passive cholesterol efflux from In recent years, considerable attention has also been paid to the role
the cell membrane, which activates a protein kinase C (PKC)-NF-κB- of sphingosine-1-phosphate (S1P), a bioactive sphingolipid that modu-
STAT1-IRF1 signalling axis [63]. Some of these discrepancies could be ex- lates the inflammatory effects of HDL on macrophages [73]. In general
plained by significant differences in experimental conditions, but also in five receptors are described for S1P of which two are highly expressed
the HDL preparations, highlighting the importance of HDL composition on macrophages, the S1P1 and S1P2 receptor. It has been shown that
and function [65]. For example, the reconstituted HDL product, CSL-111 the S1P1 receptor is responsible for the anti-inflammatory effects of
consists of apoA-I complexed to soybean-derived phospholipids, which S1P and induces a shift in macrophage polarization from the classical
are anti-inflammatory. The importance of the phospholipid component pro-inflammatory M1 to the anti-inflammatory M2 phenotype [74]. Ad-
will be discussed in more detail in chapter 5. Interestingly, a recent pub- ditionally, S1P has been shown to selectively decrease the activation of
lication by Fotakis et al. is the first study to provide evidence for both TLR2 in macrophages, thereby mediating the inhibition of pro-
anti- and pro-inflammatory effects of HDL in macrophages [61]. These inflammatory cytokine expression [75]. HDL-associated S1P also in-
investigators proposed that these effects follow a biphasic pattern at hibits the apoptosis of macrophages by stimulating STAT3 signalling
the gene expression level where initially anti-inflammatory effects dom- and survivin expression [76]. These effects of HDL-associated S1P illus-
inate and pro-inflammatory effects prevail later on. Moreover, pro- trate how small changes in HDL composition can influence atheroscle-
inflammatory effects were suggested to be highly dependent on cellular rosis and CVD.

98
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

PTMs, such as oxidation, also determine how HDL influences inflam- Moreover, apoA-I modulates the cellular fate of Treg cells during ath-
mation [77]. For example, the acute phase protein, serum amyloid A erosclerotic lesion development. Normally, during atherogenesis Foxp3
(SAA), is clearly linked to macrophage inflammation and CVD. HDL iso- expression decreases in Treg cells which reduces their immunosuppres-
lated from acute coronary syndrome patients have increased levels of sive function and leads to pro-atherogenic T follicular helper (Tfh) cells
HDL-bound SAA [78]. Furthermore, in patients undergoing coronary an- formation [103]. Injection of apoA-I into Apoe−/− mice reduced the con-
giography a low SAA, and a high plasma HDL-C was associated with version of Treg cells into Tfh cells and therefore had an athero-protective
lower all-cause and cardiovascular mortality. On the other hand, in pa- effect [103]. A similar result was obtained when Apoe−/− and Ldlr−/−
tients with high SAA, a high HDL-C was associated with increased mor- mice were injected with apoA-I, resulting in increased Treg cell counts
tality, indicating that SAA may impair the cardioprotective functions of in the lymph nodes, with a concomitant decrease in effector memory
HDL by reducing its anti-inflammatory function [55,56,79,80]. The bind- T-cells [101]. Interestingly, and seemingly in contrast to the above
ing of SAA to HDL also reduces the ability of HDL to mediate cholesterol described studies, HDL-associated S1P inhibits the differentiation
efflux from macrophages [81–83], which inherently limits the anti- of Treg cells, while reciprocally increasing the development of pro-
atherogenic potential of HDL. It has also been confirmed that SAA is atherogeneic Th1 cells [104]. Therefore, the exact role of HDL composi-
present in the atherosclerotic lesions of both mice [84] and humans tion in this process remains an active area of research.
[85,86]. Combined, these studies highlight that HDL-associated SAA HDL also influences activation of the T-cell receptor (TCR) which,
clearly has detrimental effects and that pathological conditions can upon activation, associates with lipid rafts [105]. As described above,
change HDL composition and thereby also shift the balance between this association is highly dependent on the modulation of lipid raft cho-
anti- or pro-inflammatory effects of HDL [87–89]. lesterol content by apoA-I or HDL and is therefore highly dependent on
4.1.1.1.2. Dendritic cells. Dendritic cells (DCs) are potent antigen pre- HDL cholesterol efflux capacity [106]. This has been confirmed in an ath-
senting cells that initiate T-cell mediated immune responses [90]. Sev- erogenic Ldlr−/− mouse model, in which apoA-I inhibited the activation
eral studies have demonstrated that DCs play an important but of T-cells by preventing the accumulation of cholesterol in lymph nodes
complex role in atherosclerosis. Mature DCs are pro-atherogenic, [107]. Further supporting the importance of HDL composition is the ob-
while immature DCs appear to be anti-atherogenic [91,92]. servation that HDL-S1P inhibits lymphopoiesis and consequently adap-
Similar to their effects on macrophages, apoA-I and HDL have also tive immune responses [108]. In conclusion, these observations indicate
been shown to mediate anti-inflammatory effects on DCs by regulating that HDL can influence T-cells on multiple levels: by modulating the in-
lipid rafts. These effects are again highly dependent on a completely flammatory profile, by determining cellular fate and by activation of TCR
functional HDL particle. The major histocompatibility complex (MHC) which all have been shown to be influenced by HDL composition and
class II, which is required for the antigen presentation to T-cells and function.
their subsequent activation, resides in lipid rafts [93]. Treatment of 4.1.1.1.4. B-cells. B-cells, which play an important role in humoral im-
DCs with either apoA-I or HDL disrupts lipid rafts, which decreases munity by secreting antibodies, have emerged as an important immune
DC-dependent activation of T-cells and IL-2 production. Further cell in cardiovascular disease. Over the years, it has become clear that
supporting the notion that HDL function and composition play are im- various B-cell subsets have differential effects on atherosclerosis devel-
portant in relation to DCs is the observation that S1P plays an important opment, which is comprehensively reviewed in [109].
role in the anti-inflammatory effects of HDL. Treatment of mature DCs Although the effects of HDL on B-cells has not been extensively stud-
with S1P inhibits the secretion of the pro-inflammatory cytokines ied, it seems that HDL cholesterol efflux capacity plays an important
TNF-α and IL-12, and stimulates secretion of anti-inflammatory cyto- role. Similar to the TCR, the B-cell receptor (BCR) is also located in
kine, IL-10, thereby facilitating immunosuppressive effects [94]. This lipid rafts which play an important role in B cell activation. Removal of
has been confirmed in a more recent study which showed that S1P cholesterol, by apoA-I or HDL, from lipid rafts decreases BCR-initiated
stimulates the S1P1 receptor in activated DCs, thereby inducing an signal transduction, loading of antigenic peptides onto MHC-II mole-
anti-inflammatory cytokine profile with reduced IL-12 and IL-23 pro- cules, endocytosis of BCR–antigen complexes and MHC-II associated an-
duction and a concomitant increase in IL-27 production [95]. The com- tigen presentation to T-cells [110]. This suggests that HDL influences B-
position of HDL has also been shown to play an important role on the cell functions in a manner that is dependent on the capacity of HDL to
effects of HDL on DC-mediated activation of T-cells. It could be observed act as cholesterol acceptor. Further studies are needed to investigate
that the phospholipid fraction of HDL (in particular 1-palmitoyl-2- the exact effects of HDL on different B-cell subsets and the influence of
linoleyl-phosphatidylcholine (PLPC)) was primarily responsible for the HDL composition and function in this context.
inhibitory effects of HDL on DC-mediated induction of a Th1 response
of T-cells [96]. Taken together, although the exact role of DCs in athero- 4.1.1.2. Clinical potential of apoA-I and HDL in cardiovascular disease. In
sclerosis and CVD remains a matter of debate, apoA-I and HDL composi- this chapter various approaches that have been used to develop agents
tion have clearly been shown to modulate DC function, making this for increasing HDL levels and function will be discussed in the context of
interaction an interesting target for future therapeutic interventions. cardiovascular disease (Fig. 4).
4.1.1.1.3. T-cells. It was reported several years ago that 7–20% of infil- 4.1.1.2.1. Dietary intervention. A direct but rather difficult way of in-
trating cells within human atherosclerotic plaques are T-cells [97]. Over creasing HDL levels are lifestyle interventions, such as consumption of
the years, the role of T-cells in atherosclerosis has become more clear, a low-fat diet and regular exercise. Several studies have focussed on
although there is still a great need to better understand their exact con- the effects of lifestyle changes on HDL functionality.
tribution to disease development. For example, regulatory T-cells (Treg) 4.1.1.2.1.1. Macronutrients. A well-studied dietary intervention aiming to
have an athero-protective phenotype and modulate inflammation by increase apoA-I and thereby HDL concentrations, is supplementation
secreting anti-inflammatory molecules including IL-10 and TGF-β with various fatty acids. A recent meta-analysis, demonstrated that a
[98,99]. Administration of apoA-I or HDL in mice has been shown to in- significant increase in serum apoA-I concentrations is observed when
hibit inflammation by increasing Treg cell counts in the spleen of Ldlr−/− only 1% of the carbohydrate energy is replaced by saturated or unsatu-
mice [100,101]. In line with this and highlighting the importance of HDL rated fatty acids [111]. Oleic acid and linoleic acid are among the most
function in this context, the infusion of oxidized, dysfunctional HDL into effective unsaturated fatty acids in this respect. Another meta-analysis
Ldlr−/− mice reduces Treg cell counts and thereby promotes atheroscle- described similar effects on apoA-I concentrations, at least in a fasting
rosis development [100]. These positive effects of HDL on Treg cell counts state, when carbohydrates are replaced by trans-fatty acids [112]. Inter-
may be caused by improved cell survival, as it was recently shown that estingly, it has been shown that omega-3 fatty acids, such as
HDL increases Treg cell survival by increasing oxidative phosphorylation eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), do not
in the mitochondria [102]. influence apoA-I concentrations [113]. A recent study also confirmed

99
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

Fig. 4. Highlighted mechanism of action of HDL-focussed therapies. Therapies focussing on modulating HDL metabolism or plasma HDL-cholesterol levels have different sites of interaction,
as discussed in more detail in the text below. Abbreviations: ABCA1: ATP-binding cassette subfamily A member 1; ABCG1: ATP-binding cassette subfamily G member 1; CETP: cholesteryl
ester transfer protein; EL: endothelial lipase; FFA: free-fatty acids; HDL: high-density-lipoproteins; HDLR: HDL receptor; HL: hepatic lipase; IDL: intermediate-density-lipoproteins; LCAT:
lecithin-cholesterol acyltransferase; LDL: low-density lipoproteins; LDLR: LDL receptor; LPL: lipoprotein lipase; PLTP: plasma phospholipid transfer protein; SR-B1: scavenger receptor
class B member 1; VLDL: very-low-density lipoproteins.

that EPA/DHA supplementation does not change HDL-C levels in Mediterranean diet. This clearly demonstrates that the HDL lipidome
humans, although it could be demonstrated that HDL composition can be remodeled within 4 days of diet change [117].
was significantly changed upon supplementation [114]. A down- Besides the described food supplementations, drink consumption
regulation of several HDL-associated proteins involved in inflammation can also influence apoA-I levels. Several studies have shown that coffee
was observed with both fatty-acids supplements, but to a greater extent or tea drinking does not influence fasting apoA-I levels [113]. However,
with EPA. Furthermore, apoM abundance in HDL was increased after a very consistent and striking improvement of both fasting and
EPA/DHA supplementation. ApoM forms a complex with S1P that has postprandial apoA-I concentrations have been found after alcohol
anti-atherosclerotic functions [115], which suggests that dietary fatty consumption [118,119]. As demonstrated by an increased cholesterol
acid supplementation influences HDL composition and its function. efflux capacity, HDL functionality was also improved in one of these
The effects of soy proteins on fasting apoA-I concentrations are studies [120].
rather inconsistent. Although most studies did not find any effects of 4.1.1.2.1.2. Micronutrients. Theobromine, a supplement found in cocoa
soy protein supplementation [113], one study showed that soy- has been shown to increase serum HDL-C concentrations in a 4-week
containing products did increase apoA-I concentrations, while another randomized controlled trial [121]. Interestingly, it has been shown
study demonstrated the opposite, with apoA-I levels decreasing after that the potency of theobromine depends on the baseline lipoprotein
soy supplementation [116]. Further studies are needed to clarify the un- profile of the studied subjects, as theobromine was not effective in sub-
derlying mechanisms of these differential effects, and especially the ef- jects with low HDL-C levels and high VLDL triglyceride levels [122,123].
fects of soy protein supplementation on HDL composition and However, one of these studies could not fully confirm the beneficial ef-
functionality. fects of theobromine on HDL levels, although non-significant trends
A recent randomized two-way crossover clinical trial supported the were observed [123]. The underlying mechanism by which theobro-
notion that dietary interventions can significantly alter HDL composi- mine affects HDL has been postulated to be mediated by the inhibition
tion. This trial investigated the effects of a short-term Mediterranean of phosphodiesterases, resulting in the degradation of cAMP and
or fast food diet on HDL composition and function [117]. The subjects re- increased ABCA1 activity [122]. The effects of theobromine are indepen-
ceived both diets for 4 days, with a 4-day wash-out period in between. dent of intestinal apoA-I production [124]. Whether theobromine also
In total 170 HDL-associated lipid species were analysed of which 41 affects HDL function remains to be determined.
were differentially affected by the diet. Saturated fatty acids and odd Vitamins have also been shown to influence HDL concentration and
chain fatty acids were enriched by the fast food diet, while very-long composition. For example, in mice vitamin C and E supplementation re-
chain fatty acids and unsaturated fatty acids were enriched after the duces the phospholipid content in HDL by decreasing PLTP activity

100
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

[125]. Besides lipid remodelling, these vitamins also promoted HDL This trial has started recruiting patients to test this PPARα agonist in
protein modifications by increasing paraoxonase 1 (PON1) and apoD, about 10,000 statin-treated patients with diabetes and high triglycer-
which correlated with improved HDL antioxidant activity [125]. The ide/low HDL-C plasma levels. First results from this study are expected
effect of PON1 on oxidation has been confirmed by studies in which in 2022 and are eagerly anticipated to determine whether fibrates
PON1 deficiency is associated with decreased HDL antioxidant activity, have an greater therapeutic potential in this sub-group of patients.
whereas the addition of PON1 increases the antioxidant activity This ongoing trial will also provide more information whether
of HDL [126]. This may be one of the main underlying reasons pemafibrate, a next generation, more potent and selective PPARα mod-
why PON1 deficient mice are more susceptible to the development of ulator [140] has less adverse side effects, such as gastrointestinal issues,
atherosclerosis [126]. skin rash, muscle pain and in some cases liver dysfunction following
Taken together, various dietary interventions have been proven to chronic use, than the currently available PPARα agonists [141,142]. It
be beneficial for HDL concentrations, but also especially for HDL compo- would be highly interestingly to also assess whether pemafibrate has ef-
sition and functionality, although many inconsistencies remain to be fects on HDL composition, besides HDL-C. It has previously been shown
further elucidated. From a preventive point of view these strategies that fenofibrate affects HDL composition in rabbits, by decreasing satu-
are difficult to successfully implement as it remains challenging to trig- rated fatty acids and increasing unsaturated fatty acids, which could
ger people to comply with strict dietary regimes. thereby also influence HDL functions like the cholesterol efflux capacity
[143]. Further studies in a human setting would be needed to confirm
the effects of fibrates on HDL composition.
4.1.1.2.2. Drugs
The activation of PPARα is the main mechanism of action of fibrates
4.1.1.2.2.1. Niacin. Niacin or nicotinic acid has been used widely to raise
or fibric acid [144]. This modulates several processes, including increas-
HDL-C levels, by 15–25% [127], although, the underlying mechanism of
ing the oxidation of free fatty acids in the liver and reducing the hepatic
action is not fully understood. The Arterial Biology for the Investigation
synthesis of triglycerides [138]. Additionally, PPARα increases expres-
of the Treatment Effects of Reducing Cholesterol (ARBITER 2) study
sion of lipoprotein lipase (LPL), which hydrolyzes triglycerides and
clearly demonstrated benefit by showing that niacin stabilized carotid
phospholipids in VLDL, chylomicrons and HDL [145] and reduces
intima-media thickening, on top of statin use [128]. In a follow-up
circulating triglyceride levels. Fibrates also increase HDL-C levels and in-
study, ARBITER 6-HDL and LDL Treatment Strategies in Atherosclerosis
crease the synthesis of apoA-I, apoA-II and ABCA1 [146,147]. Interest-
(ARBITER 6-HALTS), niacin was shown to be superior to ezetimibe in re-
ingly, in contrast to these human observations, fibrates do not exert
ducing carotid intima-media thickening in patients [129]. These out-
similar effects on apoA-I synthesis in rodents [148]. These differential
comes led to the worldwide and widespread use of niacin to increase
effects of PPARα are likely due to species-specific regulation of apoA-I,
HDL-C for several years [127,130], until two major and more recent clin-
as there are 3 nucleotides difference between rat and human ApoA1
ical trials cast doubt on the efficacy and especially also the safety of
promotor sites, rendering the PPARα-response element in the ApoA1
niacin.
promotor non-functional in rats [149]. These differences complicate
The Atherothrombosis Intervention in Metabolic Syndrome with
studies of the underlying molecular mechanisms.
Low HDL/High Triglycerides: Impact on Global Health Outcomes
(AIM-HIGH) trial was the first double-blind, randomized, multi-center
trial to assess whether addition of extended-release niacin to statin- 4.1.1.2.2.3. CETP inhibitors. CETP regulates lipoprotein cholesterol/tri-
treated patients reduces atherogenic dyslipidemia and cardiovascular glyceride ratios, as discussed above. There is a negative correlation be-
risk [131]. Although niacin treatment resulted in improvement of mul- tween CETP activity and plasma HDL-C levels, making CETP inhibition
tiple lipid parameters, the primary endpoints, including cardiovascular an interesting therapeutic target. There have been several clinical stud-
risk, were not met and the trial was terminated after 3-years of ies focusing on the effects of CETP inhibition on HDL levels and assessing
follow-up on the basis of futility. The Second Heart Protection Study – the therapeutic potential of these agents in CVDs [29,138].
Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2- One of the first trials, ILLUMINATE, assessed the effects of the CETP
THRIVE) was another large double-blind, randomized, multi-center inhibitor, torcetrapib. It was terminated prematurely due to an unex-
trial studying the effect of niacin in combination with laropiprant, in pected increase in mortality rates due to sepsis [150]. Subsequent stud-
statin-treated patients [132]. Laropiprant is a prostaglandin (a type of ies discovered that torcetrapib has a significant effect on the renin-
eicosanoid – physiologically active lipid compound) D2 receptor antag- angiotensin-aldosterone system (RAAS), resulting in hypertension and
onist which is suggested to reduce previously observed side effects of hypokalemia which possibly contributed to the detrimental outcome
niacin, such as flushing. Similar to AIM-HIGH, this study revealed favor- [151]. The sepsis-related mortality of torcetrapib could not be attributed
able effects on lipid parameters, but no significant reduction was ob- to any direct effects on lipopolysaccharide binding protein (LBP), nor to
served in major vascular events. Subjects treated with niacin also had inhibition of the interaction of CETP with LPS [152]. Therefore, it could
significantly increased adverse effects, including gastrointestinal prob- be speculated that on top of the aldosterone elevation, changes in
lems, infection, bleeding and a trend towards increased risk of death. plasma lipoprotein metabolism and composition may have contributed
Based on these studies, niacin was withdrawn from several markets, al- to the increase in sepsis. Based on these results, other trials turned their
though the use of niacin is still extensively being investigated in various focus to novel CETP inhibitors such as dalcetrapib (dal-OUTCOMES) and
pathologies. evacetrapib (ACCELERATE). These studies did not show any of the pre-
viously observed adverse side-effects, but were also terminated early
4.1.1.2.2.2. Fibrates. Fibrates, PPARα agonists, have been evaluated in due to futility [153,154]. The recently published trial of anacetrapib (RE-
several clinical outcome trials [133–137]. However, the results of VEAL), demonstrated for the first time that CETP inhibition can have
these studies were variable [138], possibly because different types of beneficial effects on clinical outcomes [155]. Anacetrapib increased
fibrates (e.g. gemfibrozil, bezafibrate, fenofibrate) were used and differ- HDL-C plasma levels by more than 2-fold, while decreasing LDL-C levels
ent patient groups (with or without diabetes) were investigated. Al- by more than 20%, resulting in a decreased occurrence of first major cor-
though the association of fibrate use with CVD mortality was limited onary events by 9% without any safety concerns. Although these results
in most studies, disproportionately large benefits have been observed seem promising, the clinical benefit is rather modest and perhaps more
in patients with high plasma triglyceride and low HDL-C levels at the related to the small decrease in LDL-C plasma levels. Therefore, the
start of the study. This observation triggered the initiation of a new clin- pharmacological development of anacetrapib has been stopped, which
ical trial called Pemafibrate to Reduce Cardiovascular OutcoMes by Re- at least at this moment is the end of clinical trials focussing on this
ducing Triglycerides IN patiENts With diabetes (PROMINENT) [139]. class of drugs for CVD-therapy [156]. A potential explanation for this

101
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

futility of CETP inhibition in CVD is provided by a recent Mendelian Pre-clinical studies of another mimetic peptide called 4F, showed
randomization analyses [157]. In this study, 102,837 participants that this peptide was only able to inhibit early atherogenesis, but was
from 14 studies were analysed and the observed associations were ex- ineffective against more mature lesions [171]. 4F peptide was tested in
ternally validated in 189,539 participants from 48 studies. Considered humans for the first time in a randomized clinical trial that assessed
alone, a CETP score was associated with higher levels of HDL-C, lower the efficacy and safety of orally administrated D-4F peptide in high-
LDL-C and a corresponding lower risk of major CV events. However, risk coronary heart disease patients. Treatment with D-4F resulted in
when combined with a 3-hydroxy-3-methylglutaryl-CoA reductase HDL with improved anti-inflammatory properties, suggesting that D-
(HMGCR) score, the CETP score was still associated with a similar reduc- 4F might be a potential therapeutic agent to improve HDL function
tion in LDL-C, but an attenuated non-significant risk of major CV events. [172]. However, large clinical trials have not been performed to evaluate
These results indicate that a monotherapy of CETP inhibition, without the effect of these mimetic peptides on major adverse cardiovascular
statin treatment, may have more pronounced beneficial effects. This hy- events, though based on the pre-clinical work it seems that 4F could
pothesis could be tested in future clinical trials in statin-intolerant only be effective in an early stage of atherosclerosis development
patients. which is difficult to test in a human setting as patients are often only
In addition to the REVEAL-trial, the dal-ACUTE randomized trial, identified in the later stages of the disease.
which investigated the effects of the CETP inhibitor dalcetrapib on Direct infusion of reconstituted HDL is another therapeutic ap-
markers of HDL function, fuelled the debate that future studies should proach. To date monomeric apoA-I which is present in the HDL fraction
focus on modulating HDL functionality rather than HDL-C levels [158]. of most humans and the dimeric form of apoA-I (apoA-IMilano) which
This study showed that CETP inhibition resulted in approximately 30% has an enhanced cholesterol efflux potential have been formulated as
increase in HDL-C levels, while the apoA-I content and total cholesterol reconstituted HDL for use in clinical trials [138].
efflux capacity were only increased by 10%, clearly supporting the con- In a randomized clinical trial, infusions of reconstituted HDL contain-
cept of dissociation between increased HDL-C levels and improvements ing monomeric apoA-I (CSL-111), significantly improved plaque char-
in HDL function. Recently, it was also shown that elderly subjects have acteristics although there were no changes in atheroma volume [173].
remarkably high cholesterol efflux capacity, compared to middle-aged The successor of CSL-111, which has an enhanced ability to accept cho-
individuals, although the implications of this for therapeutic approaches lesterol from ABCA1, is called CSL-112 [174]. In a dose-ranging Phase IIb
and clinical trials remains to be determined [159]. trial (AEGIS-I), infusions of CSL-112 were well tolerated, without any se-
rious safety concerns, and acutely enhanced cholesterol efflux [175].
4.1.1.2.3. Direct targeting of apoA-I and HDL. Changing HDL levels and Currently, a large-scale Phase III trial of patients with acute coronary
composition can also be done in a more direct manner. For example, syndrome is being undertaken to evaluate the ability of CSL-112 to re-
the endogenous synthesis of apoA-I is increased to some extent with duce major adverse cardiovascular events [176].
a small molecule called apabetalon or RVX-208 or RVX000222, which CER-001 is another reconstituted HDL preparation that contains apoA-
inhibits the bromodomain and extra terminal (BET), resulting in an I and sphingomyelin. Pre-clinical studies have demonstrated that CER-
epigenetic modulation of the genes responsible for apoA-I synthesis. 001 stimulates reverse cholesterol transport and induces atherosclerosis
A Phase II clinical trial showed that RVX-208 was associated with in- regression in mice [177]. Recently, the results of a Phase II double-blind,
creased plasma apoA-I and HDL-C levels in statin-treated patients randomized, multicenter trial (CARAT) was published, in which the effect
with coronary artery disease [160]. These beneficial effects were con- of CER-001 infusion on coronary atherosclerosis progression was evalu-
firmed in a Phase IIb trail (ASSURE) which employed serial intravas- ated in statin-treated acute coronary syndrome patients [178]. Unfortu-
cular ultrasound measures of coronary atheroma in 281 patients nately, the lipid profile in these patients did not change and regression
treated with RVX-208 or placebo for 26 weeks [161]. RVX-208 treat- of coronary atherosclerosis was not influenced by CER-001 infusion.
ment modulated plaque vulnerability favourably, though interest- Another study using a recombinant apoA-IMilano/phospholipid com-
ingly only changes in HDL particle concentration could be observed plex (ETC-216) showed significant regression of coronary atherosclero-
upon treatment, not in HDL-C or apoA-I. Although RVX-208 treat- sis, demonstrating great promise for this approach [179]. It has been
ment of non-human primates demonstrated striking increases in demonstrated that plaque regression after ETC-216 treatment in a rab-
serum apoA-I and HDL-C levels (60% and 97%, respectively) [162], bit model, was associated with a shift towards a more stable lesion phe-
the observed increases in human trial were on average below 10% notype after four days [180]. A follow-up study confirmed that these
[160]. Although the treatment protocols differed, this significant dif- beneficial effects are long lasting [181]. However, the clinical develop-
ference in effect on lipid levels indicates that there may be an as yet ment of ETC-216 was halted after a serious adverse reaction occurred
unknown species-dependent mechanism of action of RVX-208. Cur- early during an follow-up clinical trial in one patient. During infusion
rently, there is an ongoing Phase III study, investigating the effects the patient developed a severe reaction consisting of flushing, chills, hy-
of apabetalone (RVX000222) on major adverse cardiac events potension and ultimately multi-organ failure. This all pointed to a sys-
(BETonMACE) [163,164]. These results are eagerly awaited to evalu- temic inflammatory reaction which would suggest a contamination of
ate the potential of this therapeutic approach. the infused product, which was indeed the case as small quantities of re-
Another strategy to increase apoA-I levels is to infuse artificially syn- sidual host cell proteins that elicit immune responses were found in
thesized apoA-I mimetic peptides. These can be designed to mimic the ETC-216. Hereafter, a similar complex containing apoA-IMilano was in-
lipid binding properties of apoA-I and improve their stability relative troduced as MDCO-216. This formulation did not induce any adverse
to the parent apolipoprotein [165,166]. Several pre-clinical studies immunostimulation in healthy volunteers or patients with stable coro-
have shown that apoA-I mimetic peptides have anti-inflammatory, an- nary artery disease [182]. However, in a double-blind, randomized clin-
tioxidant and antitumor activities [167,168]. One drawback until now is ical trial of acute coronary syndrome patients, infusion of MDCO-216
that most of these apoA-I mimetic peptides are designed to enhance did not promote atherosclerotic lesion regression [183].
only one or two specific functions of apoA-I rather than the full spec- Several direct targeting approaches have been developed and are
trum of physiological relevant functions. One of these mimetic peptides being tested. Especially in the approaches where reconstituted HDL is
that showed great promise in pre-clinical studies is the peptide 5A, being used, a major focus has been to increase HDL function by, for ex-
which induces cholesterol efflux via ABCA1 and ABCG1, thereby reduc- ample, developing agents that have an enhanced cholesterol efflux ca-
ing atherosclerosis in mice [169]. Furthermore, 5A inhibits acute inflam- pacity or that contain specific proteins that improve HDL function,
mation and oxidative stress in human endothelial cells and rabbit such as sphingomyelin or apoA-IMilano. As the importance of HDL func-
carotid arteries [170]. Future clinical trials should determine the poten- tion is being more recognized, future approaches of HDL therapy should
tial of this apoA-I mimetic peptide in humans. focus on this aspect.

102
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

4.1.2. Diabetes mellitus 4.1.2.2. Clinical potential of apoA-I and HDL in diabetes mellitus. In a recent
Diabetes Mellitus (DM) is a disease involving inappropriately el- systematic review and network meta-analysis of patients with T2DM,
evated blood glucose levels. The main subtypes of DM are type 1 the impact of dietary interventions on the blood lipid profile was
(T1DM), caused by defective insulin secretion, and type 2 (T2DM), assessed, with a Mediterranean diet raising HDL-C and decreasing TG
caused by defective insulin function [184]. T1DM generally presents levels compared to control diet. This indicates that a Mediterranean
at a very young age, while T2DM is occurs in adolescents and adults diet can be used to manage diabetic dyslipidaemia [205].
that have prolonged hyperglycemia due to lifestyle problems. As Besides dietary interventions, fibrates have also been tested in the
the pathogenesis of the two types of DM is different, different context of DM. Initially, the Action to Control Cardiovascular Risk in Di-
therapeutical approaches are required. Both types of DM are associ- abetes (ACCORD) lipid study did not show major beneficial effect of
ated with a high risk of developing chronic complications, such as adding a fibrate on top of a statin, compared to statin alone, on cardio-
retinopathy, nephropathy, neuropathy, endothelial dysfunction, vascular outcomes and mortality in T2DM patients, although diabetic
and atherosclerosis. retinopathy progression was reduced [206]. However, a beneficial effect
in CV-events was shown in a pre-specified sub-group of patients with
4.1.2.1. Effects of HDL on the pathogenesis of diabetes mellitus. During the dyslipidemia. This triggered a follow-up study (ACCORDION) in which
course of T2DM, plasma triglycerides and LDL-C increase, while HDL-C a secondary analysis was performed in a pre-specified sub-group of par-
decrease [185]. A meta-analysis evaluated the association of HDL-C ticipants with dyslipidemia, demonstrating that fibrates reduce mortal-
and risk of diabetic nephropathy in T1DM patients, demonstrating ity rates in dyslipidemic patients [207]. In addition, a multicenter
that higher HDL-C levels indeed result in a reduced risk of diabetic com- randomized controlled study showed that in T2DM patients, adminis-
plications [186]. In contrast, a recent retrospective population-based co- tration of ezetimibe/rosuvastatin combination therapy significantly re-
hort study involving 2113 patients with T2DM demonstrated that the duces the apoB/A-I ratio and improves the lipid profile compared to
levels of HDL-C are non-protective in T2DM patients [187]. patients receiving rosuvastatin monotherapy [208].
HDL particles also undergo structural and functional modifications rHDL infusions also have beneficial effects in DM patients, by im-
in DM, resulting in loss and HDL function [185] (Fig. 6). For example, el- proving endothelial function and nitric oxide availability, compared to
evated levels of oxidative stress lead to oxidative modification of apoA-I controls [209]. In line with this, a small randomized clinical trial demon-
in HDL and thereby contributes to the generation of dysfunctional HDL strated that a single infusion of rHDL in patients with T2DM decreased
in DM [188,189]. These modifications have also been shown to result in plasma glucose levels and increased plasma insulin levels [210]. Fur-
decreased cholesterol efflux capacity [190], and impaired antioxidant thermore, focusing on diagnosis instead of therapy, it has been shown
properties in T1DM [191]. Moreover, PON1 expression is decreased dur- that both oxLDL/LDL and oxLDL/HDL are potent biomarkers for T2DM
ing T1DM and T2DM development, which possibly contributes to the [211] and can be used for discriminating the presence of macrovascular
observed cardiovascular complications [192,193]. This is supported by disease in diabetic patients [212].
the observation that elevated levels of HDL-associated PON1 were ob- All in all, the therapeutic efficiency of improving HDL function in the
served in a subset of people with T1DM that are protected from vascular context of DM based on above results would be an interesting research
complications [192]. Additionally, advanced glycation end products focus for the future.
(AGEs) which are elevated in DM and contribute to diabetic complica-
tions have also been shown to modify apoA-I and impair its 4.1.3. Liver disease
cardioprotective functions [194]. In line with this, macrophages treated The liver plays a vital role in the synthesis, secretion, catabolism, and
with albumin isolated from T2DM patients have significantly impaired storage of lipids and lipoproteins. The two main types of fatty liver dis-
cholesterol efflux and intracellular lipid accumulation, compared to ease are nonalcoholic fatty liver disease (NAFLD) and alcoholic liver dis-
cells treated with non-diabetic albumin, further demonstrating an im- ease (ALD). NAFLD includes simple steatosis (SS) and the more severe
portant role of glycation in lipid handling [195]. Lipid accumulation nonalcoholic steatohepatitis (NASH), which is characterized by inflam-
has also been demonstrated to play a role in diabetic nephropathy, as mation and fibrosis, while ALD can lead to fibrosis, hepatic steatosis and
evidenced in streptozotocin-induced diabetes in mice with significantly cirrhosis [213].
reduced renal expression of ABCA1, ABCG1 and SR-B1. As these changes
in ABCA1, ABCG1 and SR-B1 preceded the development of nephropathy 4.1.3.1. Effects of HDL on the pathogenesis of liver disease. Several studies
in these mice, it follows that defective cholesterol efflux may be the have shown that moderate, regular consumption of alcohol leads to fa-
cause of intracellular accumulation of lipids in renal cells and the devel- vorable changes in HDL lipids and apolipoproteins [214]. Moderate con-
opment of diabetic nephropathy [196]. In T2DM the distribution of HDL sumption of alcohol also affects HDL function by improving their anti-
particles is also affected, with a shift from large HDL particles enriched inflammatory and anti-oxidant properties [214]. Although very little is
in cholesteryl esters to smaller, denser triglyceride-rich HDL particles, known about the HDL composition and function in patients with SS
thereby also affecting the HDL function [197]. and NASH, it has been shown that NAFLD is associated with an altered
Besides the effects that diabetes has on HDL composition and func- HDL proteome, such as decreased plasminogen and antithrombin III
tion, apoA-I and HDL also have anti-diabetic properties [198]. For exam- levels [215], and an increased number of small HDL particles [216].
ple, it has been demonstrated that apoA-I increases insulin secretion HDL isolated from NAFLD patients also showed a reduced cholesterol ef-
from pancreatic beta-cells in an ABCA1-dependent manner, while HDL flux capacity [217] and the HDL of patients with liver cirrhosis are
does the same in an ABCG1-dependent manner [199]. Furthermore, it enriched with triglycerides, free cholesterol and phospholipids [218].
has been shown that overexpression of apoA-I in mice increases insulin Furthermore, enzymes that are involved in HDL maturation and metab-
sensitivity, while apoA-I deficiency impaired glucose tolerance olism, such as CETP and LCAT, are significantly decreased in patients
[200,201]. In line with these findings, Torcetrapib and dalcetrapib (in- with cirrhosis, resulting in a shift towards the larger HDL2 subclass as
hibitors of CETP activity) increase plasma HDL-C and apoA-I and im- well as diminished cholesterol efflux capacity [219]. These composi-
prove glycaemic control in patients with T2DM [202,203]. Glucose tional and functional changes were in addition to decreased HDL-C
uptake by skeletal muscle cells is also increased by apoA-I, in an levels with deterioration of liver function in cirrhotic patients [220].
ABCA1 and SR-B1 dependent manner [204]. HDL-C levels are inversely associated with the development of other
All in all, it seems that diabetes impacts on HDL function, while liver diseases. For example, HDL-C levels are increased in the early
vice versa HDL function also impacts on diabetes, clearly demon- stages of Primary Biliary Cirrhosis (PBC) and decline with disease pro-
strating that HDL functionality is an important determinant for its ef- gression [221], which is mediated by increased circulating hepatic lipase
fects on DM. activity and HDL catabolism. Hepatitis C virus (HCV) and Hepatitis B

103
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

virus (HBV) related infections that mainly affect the liver are also asso- migratory behaviour of various carcinoma cells (reviewed in: [229]).
ciated with low serum HDL-C levels. The interaction of lipids and lipo- Animal cancer models have confirmed these effects by showing that
proteins with liver diseases has also recently been reviewed in [222]. apoA-I inhibits the growth of tumors and their metastatic progression
(reviewed in: [229]). For example, a genetic approach was used to test
4.1.3.2. Clinical potential of HDL in liver disease. As outlined above, there is the role of apoA-I in a melanoma model. ApoA-I deficient mice showed
a clear link between HDL and liver disease. However, very few studies an increased tumor burden and reduced survival, while conversely
have focussed on the clinical potential of HDL in this disease. For exam- transgenic overexpression of human apoA-I reduced the malignant bur-
ple, niacin has been shown to beneficially affect NASH/NAFLD which has den and metastases, thereby increasing survival [234]. These effects
recently been elegantly reviewed by Kashyap et al. [223]. In short, both were mainly attributable to immunomodulatory properties of apoA-I,
in-vitro (primary hepatocytes and human hepatoblastoma cell lines) by which tumor-associated macrophages were re-programmed from a
and in-vivo (rat model of NAFLD) studies have demonstrated that niacin pro-tumor M2 to an anti-tumor M1 phenotype. In line with this, an-
decreases hepatic fat deposits, inflammation, steatosis and fibrosis, other study demonstrated that apoA-I mimetic peptides inhibited
which are all major complications associated with NASH/NAFLD tumor development in a mouse model of ovarian cancer [235]. Although
[224,225]. Clearly, more studies are needed to elucidate whether HDL apoA-I and HDL have shown promising results in preclinical studies, it is
targeting has any clinical potential in liver disease. As HDL composition challenging to apply such therapies to the highly diverse and variable
also plays an important role, it would be important that these studies pool of cancer patients. Therefore, feasibility remains a matter of debate
not only increase HDL-C levels but also regulate HDL composition and and more research into the underlying mechanisms, and especially
thereby its function. large clinical trials, would be needed to explore the possibility of using
apoA-I and/or HDL as anticancer agent.
4.1.4. Cancer Another interesting approach that is currently being explored within
Chronic inflammation has been linked to steps that promote the cancer field is the use of HDL as drug delivery platform [236–238].
tumorgenesis, including cellular transformation, survival, proliferation, Such systems are believed to provide tumor-selectivity and site-
angiogenesis and metastasis [226]. The majority of cancers are linked specificity, while reducing off-target effects [239]. As noted above, can-
to somatic mutations and environmental factors, such as smoking, cer cells overexpress SR-B1 and reconstituted HDL nanoparticles are
whereas diet also plays an important role [227]. Accumulating evidence being produced to specifically target this receptor in cancer cells [240].
also suggests that apoA-I and HDL play a role in cancer development The main advantage of such a delivery system is that apoA-I and HDL
and disease progression. are naturally occurring and infusions will not result in immune re-
sponses as has been shown in the CVD-focussed studies described
4.1.4.1. Effects of HDL on the pathogenesis of cancer. As cancer is a very above. Taken together, apoA-I and/or HDL seems to be an interesting ap-
heterogeneous disease, it is challenging to describe disease-specific ef- proach for cancer, either being used as direct effectors or as delivery
fects of HDL. Therefore, selected cancer types, for which a clear effect platforms. Future research should first address the question of the ex-
of HDL has been demonstrated, will be highlighted in this section. This tent to which HDL plays a causal role in the development of different
does not exclude that HDL is not involved in other subtypes. types of cancer.
Recently, RNA sequencing analysis of hepatocellular carcinomas
(HCC) revealed that apoA-I mRNA levels were significantly reduced in 4.2. Infectious diseases
diseased tissue compared to healthy liver tissue [228]. This coincided
with reduced protein levels of apoA-I in cancerous liver tissue and Infectious diseases are disorders caused by organisms including bac-
serum of HCC patients [229], suggesting a tumor suppressor role of teria, viruses, fungi or parasites. A wide diversity of infectious diseases
apoA-I and HDL. Other studies have confirmed this negative association exists and the way of transmission, signs and symptoms vary depending
between apoA-I and HDL serum levels with parameters cancer develop- on the organism causing the infection.
ment, summarized in [229]. Although the majority of studies have
shown a clear inverse association of apoA-I with the development of 4.2.1. Bacterial, viral and parasitic infections
cancer, positive correlations have also been reported. For example,
apoA-I levels are upregulated in the serum of patients with early stage 4.2.1.1. Effects of HDL on the pathogenesis of infectious diseases. Bacterial
gastric adenocarcinoma and retinoblastoma [230,231]. These discrep- infections caused by Gram-negative bacteria modulate HDL composi-
ancies highlight the difficulty of determining a precise role for apoA-I tion and size by reducing the apoA-I and phospholipid content and in-
and HDL in cancer. Furthermore, it is not known whether alterations creasing SAA [241]. Endotoxemia also induces changes in enzymes
in HDL and apoA-I levels could be either a cause or consequence of the that are important for HDL remodelling, such as CETP and LCAT [241].
carcinogenesis. HDL also plays an important role in more severe infections causing sep-
Several studies have confirmed that cancer cells require large sis, which will be discussed in detail later.
amounts of cholesterol to maintain their high degree of proliferation. HDL also interacts with Gram-positive bacteria, by binding directly
This means that cancer cells and tissues over-express specific compo- to lipoteichoic acid (LTA) [242] and attenuating inflammation and cell
nents involved in cholesterol transport, especially SR-B1 [232]. There- death in-vitro [243]. These beneficial effects have also been validated
fore it is plausible that HDL is the major source of cholesterol for in mice, where apoA-I attenuated LTA-induced acute lung injury and cy-
cancer cells. If this is the case, HDL functionality would also be an impor- tokine production [243]. These observations support the hypothesis
tant determinant in regulating the upregulated cholesterol homeostasis. that raising plasma HDL levels may be a viable therapeutic approach
One study that highlights the importance of HDL functionality in cancer, for the treatment/prevention of infectious diseases.
has shown that HDL completely inhibits the oxidative stress-induced However, recently it became clear that this relationship is not
proliferation of prostate cancer cells [233]. The exact role that HDL func- straightforward [244]. In two well-characterized and large-scale cohorts
tion plays in cancer should therefore be one of the main focus points of from Copenhagen, it has been demonstrated that HDL-C levels are asso-
future research, especially in light of the important role of HDL function- ciated with infection-related hospitalization [245]. Interestingly, this as-
ality in other diseases. sociated followed a U-shaped pattern where low and very high HDL-C
levels were associated with increased infection risk. Supporting this
4.1.4.2. Clinical potential of apoA-I and HDL in cancer. In-vitro studies finding, SNPs in the genes encoding for CETP and hepatic lipase (LIPC)
using recombinant apoA-I or apoA-I mimetic peptides have demon- were associated with a reduced risk for infections (especially bacterial
strated that apoA-I can decrease the proliferative, survival and infections). It remains challenging to explain the intriguing association

104
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

between high HDL-C and increased infectious risk [244]. As inflamma- IL-1ß, TNF-α, VCAM-1 and ICAM-1 that are known to be involved in
tion results in changes in the composition of HDL, including the accu- the progression of septic shock.
mulation of SAA, it is possible that such compositional changes may be
responsible for the observed association. Furthermore, PTMs may 4.2.2.1. Effects of HDL on the pathogenesis of sepsis. Interestingly it has
occur in inflammatory environments, as described above, which again been shown that HDL, especially apoA-I, can neutralize LPS [257] by in-
induce changes in HDL composition and function. All in all, this again creasing its biliary excretion and clearance by the liver, thereby
highlights the need to further investigate the exact role of HDL function protecting against septic shock (Fig. 5) [258]. Further support of LPS-
in bacterial infections. lipoprotein interactions comes from a biosensor study demonstrating
The interaction between HDL and parasitic infections mainly relates that the majority of LPS is associated with HDL [259]. This interaction is
to the ability of apoL-1 to limit Trypanosoma brucei and Leishmania in- mediated by lipid transfer proteins such as CETP, PLTP and LBP, which
fections [242]. ApoL-1 is part of the trypanosome lytic factor-1 (TLF-1) transfer LPS to lipoproteins [260]. However the exact contributions of
complex, that mediates uptake by parasites. After uptake, apoL-1 enters these transfer proteins to this process remain unclear. Although CETP
lysosomes where the acidic pH causes a conformational change binds weakly to LPS compared to LBP [152], mice overexpressing
resulting in lysosomal swelling that kills the trypanosome [246,247]. human CETP have improved survival when exposed to experimental sys-
Due to these specific effects of apoL-1, it could be speculated that ap- temic inflammation [261]. Furthermore, it has been shown in hamsters,
proaches to increase the apoL-1 content in HDL could have beneficial that endotoxin and pro-inflammatory cytokines such as TNF-α and IL-1
outcomes in parasitic infections. decrease serum CETP activity, as well as CETP mRNA levels and protein
Changes in plasma HDL-C levels also occur during viral infections, expression in several tissues including the heart, muscle and adipose tis-
such as human immunodeficiency virus (HIV). HIV infected patients sue [262]. This increases circulating HDL levels, which is beneficial for the
have significantly decreased HDL levels, which align with upregulated clearance of endotoxins, even though the pro-inflammatory environ-
inflammasome-related gene expression and serum levels of IL-6 and ment will reduce its endotoxin clearing potential. Decreased CETP activ-
CRP [248]. These negative correlations demonstrate that HDL might af- ity during inflammation has also been reported in sepsis patients
fect the progression of HIV infection by modulating the inflammatory [241,263]. In line with this, Mendelian randomization studies have con-
state, although determining whether changes in HDL are cause or con- firmed a causal role for CETP in sepsis, as a missense variant in CETP re-
sequence remains to be determined. On the other hand, the pro- duced HDL-C levels and decreased sepsis survival [264].
inflammatory state in these patients also increases apoA-I and HDL In humans clear associations have been made between HDL and the
levels following antiretroviral therapy depending on the extent of in- development of sepsis. Clinical studies have highlighted that HDL-C
flammation at baseline [249]. In a small retrospective study, it was levels decline during sepsis [265,266] and that this is associated with in-
shown that HIV infection also corresponded with a decrease in the anti- creased mortality and adverse clinical outcomes [267,268]. Reduced
oxidant and anti-inflammatory functions of HDL [250]. The importance levels of HDL-associated S1P in patients with sepsis are associated
of HDL function has also been confirmed in another study, demonstrat- organ failure [269]. Low levels of S1P are associated with cardiac dys-
ing that antiretroviral treatment was associated with higher HDL anti- function in mice with sepsis [270]. Additionally, it has been shown
oxidant function and PON-1 activity [251]. that HDL levels decreases, HDL size increases due to association with
SAA and HDL function is compromised during the acute phase reaction
in sepsis patients [271,272]. This was confirmed in a pilot study of older
4.2.1.2. Clinical potential of apoA-I and HDL in infectious diseases. Several
patients where isolated HDL had impaired cholesterol efflux capacity
pre-clinical studies have investigated the effect of apoA-I mimetic pep-
and a pro-inflammatory profile, compared to healthy controls [273].
tides in infectious diseases or related processes. For example, the 4F mi-
Therefore HDL function also seems to play an important role in the path-
metic peptide inhibits the expression of pro-inflammatory mediators in
ogenesis of sepsis.
LPS-treated cells in-vitro by neutralizing LPS [252]. These beneficial ef-
The above associations do not prove that a causal relationship exists
fects have been confirmed in-vivo in endotoxemic rats, where 4F treat-
between HDL and sepsis in humans. However, a causal role of apoA-I in
ment results in the attenuation of acute lung injury and increases
protection against sepsis has been confirmed in mouse models of sepsis
survival rates [253]. Future studies are warranted to translate these
where apoA-I deficiency robustly increased production of inflammatory
findings into the clinical setting.
cytokines, while overexpression of apoA-I increased survival rates com-
Regarding parasitic infections, it has been demonstrated that a re-
pared to control mice [274]. Treatment of septic mice with HDL signifi-
combinant apoL-1-nanobody binds to trypanosomes and lyses them
cantly reduced the release of IL-8, IL-6 and TNF-α [275,276],
in-vivo [254]. Treatment of mice infected with Trypanosoma brucei
demonstrating a potential role of HDL as therapeutic target for
brucei with this apoL-1 nanobody resulted in a striking clearance of
endotoxemia.
the parasite from the circulation, indicating the therapeutic potential
A potentially causal relationship between HDL and sepsis in humans
of this approach [254].
has recently been demonstrated in a prospective study of >400,000 par-
Although, HDL clearly plays a potentially important role in infectious
ticipants that examined the association of lipid levels and the risk of in-
diseases, the clinical potential of apoA-I and HDL in these diseases is un-
fectious hospitalizations and 28-day sepsis mortality [277]. A 1 mmol/l
explored. Therefore, it would be worthwhile for future studies to focus
increase in HDL-C levels associated with a hazard risk for infectious dis-
on the clinical potential of increasing HDL functionality in these diseases
ease of 0.84. More importantly, Mendelian randomization using HDL-C-
as an interesting new approach for treatment or possibly prevention.
related genetic variants further demonstrated a causal relationship be-
tween raised HDL-C and reduced risk of infectious hospitalization and
4.2.2. Sepsis there was a significant inverse relation existed between HDL-C and
LPS, also known as lipoglycan or endotoxin, is found in the outer 28-day sepsis mortality [277]. Together these results demonstrate that
membrane of gram-negative bacteria and is considered to be the HDL-C is causally related to sepsis. Future studies should investigate
major cause of sepsis. LPS binds to LBP, an acute phase protein, which whether this causal relationship also exists between HDL function and
transfers LPS from the bacterial wall to membrane-bound CD14. Subse- sepsis as this is most likely even a more important parameter for disease
quently, the LPS-CD14 complex binds and signals through TLR4 on outcome.
immune cells to activate the transcription factor NF-κB and mitogen-
activated protein kinases (MAPKs), such as p38 and c-Jun NH2- 4.2.2.2. Clinical potential of apoA-I and HDL in sepsis. Based on the above
terminal kinase (JNK) [255,256]. This activation regulates the expres- described relationship of HDL and sepsis with the reductions of HDL
sion of several endogenous mediators of inflammation such as IL-8, concentration and HDL dysfunction, there seems to be a great potential

105
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

Fig. 5. Mechanism of action by which HDL protects against sepsis. In sepsis, lipopolysaccharide (LPS) binds to LPS-binding protein (LBP) and interacts with the CD14 receptor, initiating
inflammatory cytokine signalling via TLR4. HDL can inhibit these inflammatory processes via two pathways, (i) inhibition of the inflammatory signalling or (ii) binding and capturing the
LPS-LBP complex resulting in excretion via the liver.

Several studies have investigated the potential therapeutic effect of


for HDL-based therapies in sepsis. Until now, all studies focused on a di- the apoA-I mimetic peptide 4F in a rat model of sepsis (cecal ligation
rect targeting approach using apoA-I mimetic peptides or reconstituted and puncture (CLP)). Administration of 4F, 6 h after CLP surgery, signif-
HDL infusions. icantly decreased plasma IL-6 concentrations, improved cardiac func-
tion and reduced mortality [278]. In another study which used the
same experimental setup, further beneficial effects of 4F treatment
were observed for kidney, vascular and lung function [279]. Together
these studies demonstrate that 4F has beneficial effects on sepsis-
related injury and by using a therapeutically relevant model (treatment
after sepsis induction) these studies highlight the therapeutic potential.
Reconstituted HDL has also been used to study sepsis in animal
models [275,280–284]. For example, reconstituted HDL infusion limited
organ injury during endotoxic shock in rodents [282]. The main limitation
of these studies is the timing of the infusion. Infusion of reconstituted HDL
before initiation of sepsis is of limited relevance in a clinical setting. Re-
cently, infusion of CSL-111 2 h after induction of sepsis in mice reduced
inflammation in plasma, liver and lungs [283]. This suggests that
reconstituted HDL has therapeutic potential in septic conditions.
To date, no randomized human study using apoA-I mimetic peptides
or reconstituted HDL has been performed in septic patients. However, in
a randomized, placebo-controlled study in volunteers infused with
reconstituted HDL as pre-treatment followed by an endotoxin challenge
there was reduced release of pro-inflammatory cytokines and downreg-
ulation of CD14 in monocytes [276]. Although the group size was ex-
Fig. 6. Diabetes alters HDL composition and function. In diabetes, high levels of oxidative tremely small (n = 8), this study highlights the therapeutic potential
stress, advanced glycation end products (AGE) and reactive oxygen species (ROS),
of HDL in human endotoxemia and stresses the need for randomized
induce direct or indirect via PON-1 and myeloperoxidase (MPO) compositional changes
in HDL particles, rendering them dysfunctional. Thereby HDL is less effective in studies with greater power.
extracting cholesterol from cells.

106
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

4.3. Auto-immune diseases antagonist infliximab [303]. The importance of HDL function in RA has
also been demonstrated in the Abatacept Versus Adalimumab Compar-
4.3.1. Rheumatoid arthritis ison in Biologic-Naïve RA Subjects with Background Methotrexate
Rheumatoid Arthritis (RA) is a chronic inflammatory disease charac- (AMPLE) study [304] where decreased CRP levels were associated
terized by symmetrical synovitis and mediated by autoimmune mecha- with improved HDL function and composition. Additionally, improve-
nisms, leading to bone destruction with progressive pain [285]. RA is ment in disease activity was associated with higher PON1 activity and
also associated with inflammation and affected patients have a signifi- lower HDL-SAA content, highlighting that this treatment regime re-
cantly increased risk of CVD [286,287]. A meta-analysis has demon- models the HDL proteome. Collectively, these outcomes indicate that
strated that the risk of CVD mortality in patients with RA is increased HDL composition and function play important roles in RA and should
by a striking 50%, compared to healthy controls [287]. be the focus of future research to optimize HDL-based treatment
approaches.
4.3.1.1. Effects of HDL on the pathogenesis of rheumatoid arthritis. Clinical
studies showed that RA patients have decreased serum HDL-C and 4.3.2. Systemic lupus erythematosus
apoA-I levels compared to healthy controls [288–291]. In particular, Systemic Lupus Erythematous (SLE) is a systemic autoimmune dis-
atheroprotective small sized HDL particles are reduced in patients ease of multifactorial origin, in which environmental and genetic factors
with RA [292]. However, as such modifications are mirrored by in- induce immune disorders [293]. Typically, SLE coincides with the pro-
creased LDL-C and total cholesterol levels, the actual clinical significance duction of type I interferon and is characterized by dysregulation of T-
of HDL-C variations remains unclear [293]. Therefore, elucidation of the and B-cells. SLE is associated with an increased cardiovascular risk and
exact role of HDL-C in RA and especially the question as to whether low accelerated atherosclerosis [305].
HDL-C levels are a consequence of the disease or a causal mediator re-
mains an active field of research. A recent study explored the causal ef- 4.3.2.1. Effects of HDL on the pathogenesis of systemic lupus erythematosus.
fect of HDL on collagen induced arthritis (CIA) in mice, and Patients with SLE show changes in their serum lipids and lipoproteins
demonstrated that HDL decreased the expression of pro-inflammatory similar to those observed in patients with RA [306–308]. The systemic
cytokines and the development of RA [294]. These beneficial effects inflammation, oxidative stress and autoimmunity in SLE patients also
have been confirmed by another study, where apoA-I and reconstituted induces compositional changes leading to the formation of pro-
HDL attenuated arthritis in rats, likely by reducing TLR2 expression in inflammatory and dysfunctional HDL [309]. Accumulating evidence
macrophages [295]. Although these studies suggest that HDL can have suggests that changes in HDL composition and function are responsible
causal effects, it should be kept in mind that these models might not re- for the 7–50 fold increased risk of coronary artery disease in SLE patients
flect the complex mechanisms that underlie alterations in lipid metab- [309–311]. Carotid intimal-medial thickening was also observed in chil-
olism in RA patients. Supporting a role for HDL-C in RA in humans is dren and adolescents with SLE, demonstrating the early onset of athero-
the observation from the Copenhagen General Population Study and sclerosis in these patients [312]. Furthermore, almost half of the SLE
the Copenhagen City Heart Study that the hazard ratio for RA is 1.51 patients have pro-inflammatory HDL, which correlates with increased
when comparing low vs. high HDL cholesterol levels, although this oxidized LDL and carotid plaque formation and carotid intimal-medial
only resulted in a non-significant statistical trend [296]. thickening [313]. Similar to HDL found in RA patients, HDL from SLE pa-
Interestingly, the composition and functional characteristics of HDL tients also has increased levels of SAA and decreased levels of PON-1
are also significantly altered in RA. HDL isolated from RA patients [314]. Interestingly, anti-apoA-I antibodies are also correlated with dis-
shows profound modifications, including increased SAA and reduced ease activity in SLE patients [315]. The ABCA1- and ABCG1-mediated
PON-1 levels, resulting in a pro-inflammatory HDL particle [297]. cholesterol efflux capacity of HDL is also significantly reduced in SLE pa-
These modifications also result in functional changes, as HDL isolated tients [299].
from patients with RA have a reduced ability to protect LDL from oxida-
tion and possibly facilitate LDL oxidation [298]. Furthermore, HDL from 4.3.2.2. Clinical potential of apoA-I and HDL in systemic lupus erythemato-
RA patients have a reduced capacity to act as cholesterol acceptors, in- sus. Although the above studies indicate that SLE has an adverse effect
dependent of HDL-C levels [299]. ABCG1-mediated cholesterol efflux on HDL function, there are at present no clear indications that apoA-I
is also impaired in RA patients and inversely correlates with disease ac- or HDL can causally or directly affect the development of SLE. However,
tivity [299]. Furthermore, anti-apoA-I autoantibodies are detectable in dysfunctional HDL might be an important biomarker for the identifica-
the serum of RA patients and predict acute cardiovascular events tion of CVD risk in SLE patients. The therapeutic potential of hypolipid-
[300]. Together, modifications of HDL composition in RA impair the emic treatment in SLE has been confirmed by treating SLE patients with
anti-inflammatory, antioxidant and cholesterol efflux-promoting ca- pravastatin plus ezetimibe, which improved the ratio of LDL-C and HDL-
pacity, highlighting that HDL function plays an important role in the C levels [316]. However, whether direct HDL-targeted approaches, such
progression of RA. as infusion of apoA-I or reconstituted HDL, has beneficial effects in SLE
patients still needs to be determined.
4.3.1.2. Clinical potential of apoA-I and HDL in rheumatoid arthritis. Several
treatments for RA influence HDL levels and functionality. For instance, a 4.3.3. Psoriasis
meta-analysis of 3552 patients demonstrated that baricitinub, a selec- Psoriasis is a chronic inflammatory skin disease related to immune
tive inhibitor of JAK1/2 signalling in RA, increases HDL-C levels in a inheritance. However, to date, the true cause of the disease remain un-
dose dependent manner [301]. However, a similar increase was also ob- clear [317]. Nowadays, psoriasis is considered to be a multisystem dis-
served in LDL-C levels, and no changes in CV risk were observed. There- ease that may be related to other diseases, including rheumatological,
fore the exact contribution of specific HDL-associated lipid changes in cardiovascular and psychiatric complications [317].
RA and the relationship with CV risk remains to be determined. More-
over, the treatment of RA patients with methotrexate reduces the in- 4.3.3.1. Effects of HDL on the pathogenesis of psoriasis. Several studies that
flammation and disease activity and restores the function of HDL back have compared serum lipid levels in patients with psoriasis to healthy
to normal [302]. Decreases in RA disease activity over time were also as- controls, demonstrate a trend towards increased serum triglyceride
sociated with increases in PON-1 and apoA-I levels and decreases in the levels, with a concomitant decrease in HDL levels [318–320].
HDL inflammatory index and levels of MPO, clearly demonstrating res- This change in lipid profile has been shown to increase MI risk in pa-
toration of normal HDL function. HDL function can even be further im- tients with psoriasis [318,320–322]. More generally, a meta-analysis of
proved by combining methotrexate treatment with the TNF-α 14 studies reported a 1.37-fold increased risk for cardiovascular

107
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

mortality in patients with psoriasis, compared to the general, healthy role in this, as the level was profoundly reduced during disease exacer-
population [323]. Additionally, IL-10 showed a strong inverse correla- bation. Furthermore, HDL isolated from Crohn's disease patients has a
tion with HDL-C in psoriasis patients. In a small randomized, placebo- reduced ability to attenuate oxidation [336]. Altogether, it is clear that
controlled clinical trial it could be shown that there is also a direct causal HDL plays an important role in IBD, though it is not yet clearly eluci-
role for IL-10 in psoriasis, as administration of recombinant human IL- dated whether HDL-changes are causal or a consequence of disease
10 to psoriatic arthritis patients sharply decreased HDL-C levels [324]. status.
After discontinuation of the IL-10 therapy, HDL-C levels recovered and
returned to baseline. Thereby, IL-10 seems to play an important role in 4.3.4.2. Clinical potential of apoA-I and HDL in inflammatory bowel disease.
the so called Disappearing HDL Syndrome, which is characterized by se- It has been shown that the circulating apoA-I mimetic peptide 4F selec-
vere HDL deficiency in non-critically ill patients [324]. tively targets the small intestine, where it is transported into the intes-
Besides these changes in HDL levels, patients with psoriasis also tinal lumen followed by reabsorption by the intestinal mucosa [337].
have significant changes in HDL composition and functionality as This mimetic peptide also reduced the levels of pro-inflammatory
reflected by decreased cholesterol efflux capacity together with de- fatty acid metabolites in enterocytes [338]. Recently, it has been
creased anti-inflammatory properties and decreased anti-oxidant prop- confirmed that 4F treatment has beneficial effects on IBD in mice, by
erties [325]. In line with this, patients with psoriasis have low PON-1 mitigating disease development and reducing the levels of pro-
activity, which is also inversely correlated with the psoriasis area and inflammatory mediators in tissues and plasma [339]. The mechanism
severity index [326]. HDL-associated proteins also undergo changes in underlying these effects include inhibition of LPS responses in macro-
psoriasis, as apoA-I and apoM are significantly reduced, while SAA in- phages and the intestinal epithelium, and direct clearance of pro-
creased. To corroborate these findings, and to show the therapeutic po- inflammatory lipids from intestinal tissue and plasma. The beneficial
tential of HDL, anti-psoriatic therapy has been shown to restore both effects of apoA-I mimetic peptides on IBD have been confirmed in an-
HDL composition and function [327–329]. other study in which mice with colitis were treated with the 5A peptide
[340] which reduced intestinal inflammation, intestinal MPO levels and
4.3.3.2. Clinical potential of apoA-I and HDL in psoriasis. It is clear that HDL plasma pro-inflammatory cytokine levels. Combined in-vitro and in-
composition and function play a role in psoriasis. However, as is the case vivo approaches demonstrated that inhibition of monocyte infiltration
for SLE, it remains to be determined whether changes in HDL function and activation by 5A was the main underlying mechanism [340]. All in
have a causal role in disease development or is only a consequence all, apoA-I mimetic peptide treatment seems to be a potential novel
of disease status. It is at this point unclear whether more direct treatment for IBD. As HDL functionality is also disturbed in IBD, future
HDL-targeted approaches would benefit psoriasis patients. More studies should also focus on therapeutic approaches in which HDL func-
pre-clinical research is needed to establish whether HDL levels and/or tionality and composition are restored. Such approaches might have a
functionality do indeed have a causal role in this pathology, after greater therapeutic effect and value than raising apoA-I levels.
which therapeutic approaches can be tested to improve the HDL func-
tion and perhaps impair the development of psoriasis. 4.4. Neurological disease

4.3.4. Inflammatory bowel disease Interest in lipoprotein metabolism in the brain gained interest after
Inflammatory bowel disease (IBD), mainly consisting of Crohn's dis- it was demonstrated that apoE was connected with the development
ease and ulcerative colitis, is a chronic and relapsing inflammatory dis- of several neurological disorders [341]. The brain is highly enriched in
ease of the gastrointestinal tract [330]. There is not one single cholesterol and accounts for ~25% of total body cholesterol. Net transfer
pathogenic mechanism underlying IBD, but various host and environ- of cholesterol from the periphery into the central nervous system (CNS)
mental triggers can dysregulate the mucosal immune response to gut is restricted as the blood-brain barrier (BBB) limits the movement of
microbiota, fuelling disease development [331,332]. The absence of a plasma lipoproteins into the brain [342]. Here we will describe the
specific trigger makes research into IBD complex and careful elucidation role of HDL on two main neurodegenerative disorders, Alzheimer's dis-
of a wide array of causal contributors is essential. ease (AD) which is the leading cause of senile dementia and Parkinson's
disease (PD) which mainly affects movement and is characterized by
4.3.4.1. Effects of HDL on the pathogenesis of inflammatory bowel disease. tremors.
In a mouse model of colitis, apoA-I and HDL suppresses intestinal in-
flammation. This is exemplified in apoA-I-deficient mice with increased 4.4.1. Effects of HDL on the pathogenesis of neurological disease
mucosal damage coinciding with increased intestinal MPO activity and For AD, several studies have demonstrated a clear negative associa-
TNF expression [333]. In line with this, mice overexpressing human tion between plasma HDL-C levels and risk of AD. For example, a small
apoA-I showed less severe symptoms in the same colitis model. HDL- prospective cohort study of 1,130 individuals in Manhattan revealed
induced autophagy that inhibits cytokine expression could be a plausi- that high HDL-C levels are associated with a decreased risk of AD
ble mechanism underlying these effects [333]. Similar trends have also [343]. In line with this, a study of Japanese-American men indicated
been observed in patients with Crohn's disease and ulcerative colitis that the levels of apoA-I and HDL-C were inversely associated with the
in whom total cholesterol and HDL-C levels were significantly lower, risk of AD and dementia [344]. In AD patients without dementia, this
while LDL-C was elevated, compared to controls [334]. Further beneficial effect of HDL-C has also been observed, as high HDL-C levels
supporting a role for HDL-C in IBD is the observation that in The Copen- correlate with an improved verbal memory [345]. Furthermore, another
hagen General Population Study and the Copenhagen City Heart Study study with subjects from the Sydney Memory and Aging Study (MAS)
the hazard ratio for Crohn's disease and ulcerative colitis is 3.22 and stated that elderly individuals with mild cognitive impairment had
1.94, respectively, when comparing low vs. high HDL cholesterol levels lower levels of apoA-I and apoH and higher levels of apoJ. Interestingly,
[296]. Interestingly, low HDL-C levels in childhood seem to prime these among the apolipoproteins, apoA-I was the most significant predictor of
individuals for IBD. In a Finnish study low HDL-C was inversely corre- cognitive decline [346], highlighting the importance of HDL composi-
lated with IBD after 34 years of follow-up [335], stressing the need for tion in AD. Although these studies demonstrate clear associations,
preventive measures. they were undertaken in small cohorts so some caution is warranted
Crohn's disease is also associated with accelerated atherosclerosis. In in interpretation and larger studies would be needed to confirm these
a small cohort of 60 Crohn's disease patients and 122 matching controls, results.
carotid intimal-medial thickness, which is indicative of accelerated ath- Recently, two large genome-wide meta-analyses of AD found that li-
erosclerosis, was significantly increased [336]. HDL may have played a poprotein metabolism and genes that encode for HDL biogenesis/

108
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

metabolism and HDL apolipoproteins, such as APOE, ABCA1, APOC1, increased risk for CKD. Two other recent studies confirmed this U-
APOM, APOA2, PON1, CLU, LCAT, CETP, and APOA-I [347,348], suggesting shaped association between HDL-C levels and CKD and also demon-
that besides HDL-C levels HDL composition also plays an important strated that a similar association exists between HDL-C and the risk
role in AD. This was confirmed in a recent prospective case-cohort study for total and CVD-related mortality [365,366].
of older adults with dementia. Higher levels of apoE in HDL were asso- However, a Mendelian randomization study that investigated 68
ciated with lower dementia risk, but only when HDL did not contain SNPs that are associated with HDL-C did not show any association
apoC-III [349]. Likewise, higher levels of apoE in whole plasma and in with kidney function [367]. This suggests that HDL-C is likely not the
HDL were linked to better cognitive function on the Alzheimer's Disease most important determinant of kidney function, but do not exclude
Assessment Scale-Cognitive Subscale (ADAS-Cog) [350]. Combined, the possibility that HDL composition and function might be involved.
these studies clearly show that HDL-C, but especially HDL composition Indeed, the HDL in hemodialysis patients has increased SAA, surfac-
are associated with AD. Further studies are needed to elucidate the un- tant protein B, albumin and apoC-III levels [368,369]. The enrichment of
derlying mechanisms of these effects and to determine whether these HDL with SAA and surfactant protein B is associated with cardiac events
effects play a causal role or a consequence of disease state. and all-cause mortality, independent of HDL-C [369]. Impaired cholesterol
Several studies have shown that apoA-I levels were significantly de- efflux capacity of HDL from hemodialysis patients and a more pro-
creased in the cerebral spinal fluid and in plasma of patients with PD, inflammatory macrophage response to patient HDL has also been de-
particularly in the early stage of disease development suggesting scribed [370]. Further supporting the fact that HDL functionality is im-
apoA-I may be a suitable biomarker for PD [351,352]. Supporting the no- paired in CKD is the observation that HDL isolated from CKD patients
tion that HDL composition also plays a role in PD it has been shown that contain high levels of symmetric dimethylarginine and reduce the pro-
the activity of plasma PON1 decreases in PD patients [353]. Several stud- duction of nitric oxide in endothelial cells [371]. Such abnormal HDL func-
ies have also reported that individuals with polymorphisms in the PON1 tion in CKD patients is also a plausible explanation for the observation that
gene that are associated with decreased enzyme activity, have an in- eGFR modifies the association between HDL-C and CVD risk [372]. Further
creased risk of developing PD [354,355]. Although a causal role for supporting a role for HDL composition in CKD is the observation that
apoA-I or HDL in PD remains to be elucidated, these associations indi- APOL1 variants have been associated with an increased CKD risk in
cate that there is an important link in which HDL composition could African Americans and Hispanics [373]. Additionally, acquired LCAT defi-
be an important determinant. ciency is a major cause of low plasma HDL levels in patients with CKD
[374]. In patients from the Ludwigshafen Risk and Cardiovascular Health
4.4.2. Clinical potential of apoA-I and HDL in neurological disease study, participants with normal kidney function (eGFR>90 ml/ml/
Although HDL-based therapeutic strategies have not been tested for 1.73m2) showed a clear inverse association between HDL-C and CVD-
AD in clinical trials, numerous pre-clinical studies have been performed related mortality [372]. However, in patients with mild or advanced re-
in mouse models of AD. For example, in a mouse model of AD, genetic duced kidney function, higher HDL-C was no longer associated with re-
overexpression of human apoA-I that increases plasma HDL levels pre- duced CVD risk. The role of HDL in CKD has recently been reviewed in
vents the development of age-related cognitive deficits [356]. Further- [375]. All in all, there are strong indications that HDL composition and
more, intravenous administration of rHDL reduced soluble brain function is a more important predictor of CKD than HDL-C.
amyloid beta (Aβ) levels, microgliosis and memory deficits in mice
[357,358]. Though these studies show potential beneficial effects of 4.5.2. Clinical potential of apoA-I and HDL in chronic kidney disease
HDL treatment, further studies are needed to confirm and validate HDL is clearly associated with CKD and CKD-related CVD risk. Though
these results in clinical settings. it seems clear that CKD influences HDL-C and HDL function, it remains to
Niacin is another promising way to influence AD. In a cohort study be determined whether there is a causal role for HDL in CKD development
dietary intake of niacin reduced the risk of AD and cognitive decline and progression. At this point, studies that address this question are still
[359], though these studies lacked direct measurement of blood niacin missing and should be a focus point for future research. Based on the de-
levels. Niacin could also play a beneficial role in PD, which is character- scribed associations, it seems that efforts should be put into the elucida-
ized by neuroinflammation [360]. Niacin has a high affinity for the tion of the role of HDL function in this context and approaches that
hydrocarboxylic acid receptor 2 (HCAR2 or GPR109A) [361] which is have the potential to modify HDL composition and thereby function.
highly expressed on macrophages, through which niacin can induce
anti-inflammatory responses. In a clinical trial it was confirmed that 4.6. Other diseases
low doses of niacin in PD patients induced a shift in macrophage polar-
ization, from the pro-inflammatory M1 towards the anti-inflammatory Many other diseases are associated with changed HDL levels and
M2 phenotype in a HCAR2 dependent manner [362], clearly demon- composition. Although indications for a causal role of HDL in these pa-
strating the potential of niacin treatment in this neuropathology. thologies is still missing, it has become clear that HDL composition
and function is changed during disease development. For example, de-
4.5. Chronic kidney disease creased HDL-C levels have been observed in patients with Sjögren's syn-
drome, Kawasaki disease, and ankylosing spondylitis [376–378].
Chronic kidney disease (CKD) describes the gradual loss of kidney Furthermore, a recently conducted meta-analysis suggested abnormal-
function. Early stages of CKD has few symptoms and is often unrecog- ities in serum lipid levels in patients with periodontal disease (PD), in-
nized. However, in more advanced stages accumulation of dangerous cluding reduced HDL levels and elevated LDL levels [379]. These
levels of fluid, electrolytes and waste products can cause serious compli- patients also show a clear increased risk for the development of CVD,
cations and increase the risk of kidney failure, CVD and death. in particular atherosclerosis [380]. Another study, demonstrated that
in Behçet's disease patients, HDL concentrations are decreased, and
4.5.1. Effects of HDL on the pathogenesis of chronic kidney disease there is a shift in HDL subpopulation distribution from HDL2 to HDL3
Mildly impaired estimated glomerular filtration rate (eGFR) is asso- [381]. The potential causal role of HDL in these diseases as well as the
ciated with low HDL-C levels [363]. A recent longitudinal cohort study in therapeutic potential of HDL-targeting remains to be determined.
almost 2 million participants demonstrated that individuals with low
HDL-C levels had a higher risk for CKD development and progression 5. Discussion and conclusions
[364]. Interestingly, similar to the described U-shape association be-
tween HDL-C and infectious diseases, a similar shaped association exists HDL has already long been known as a key player in the lipid metab-
between HDL-C and CKD as participants with high HDL-C levels had an olism. Over the past decades it has become clear that HDL has diverse

109
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

functions that affect physiological and pathological processes. Although References


it was originally believed that the level of HDL-C was the most impor-
[1] J.W. Gofman, Serum lipoproteins and the evaluation of atherosclerosis, Ann. N. Y.
tant determinant of these processes, emerging evidence indicates that Acad. Sci. 64 (4) (1956) 590–595.
the composition and thereby function of HDL is perhaps the most im- [2] J.E. Feig, J.X. Rong, R. Shamir, M. Sanson, Y. Vengrenyuk, J. Liu, K. Rayner, K. Moore,
portant factor. In this review the effects of HDL in various inflammatory M. Garabedian, E.A. Fisher, HDL promotes rapid atherosclerosis regression in mice
and alters inflammatory properties of plaque monocyte-derived cells, Proc. Natl.
and infectious diseases has been discussed, especially focusing on the
Acad. Sci. U. S. A. 108 (17) (2011) 7166–7171.
importance of HDL function, together with an overview of the clinical [3] J. Armitage, M.V. Holmes, D. Preiss, Cholesteryl ester transfer protein inhibition for
potential. preventing cardiovascular events: JACC review topic of the week, J. Am. Coll.
In recent years, there has been a lively debate about the effectiveness Cardiol. 73 (4) (2019) 477–487.
[4] D. Keene, C. Price, M.J. Shun-Shin, D.P. Francis, Effect on cardiovascular risk of high
and feasibility of many of the discussed therapeutic strategies. This was density lipoprotein targeted drug treatments niacin, fibrates, and CETP inhibitors:
mainly fueled by an meta-analysis of 40 clinical trials in CVD, which meta-analysis of randomised controlled trials including 117,411 patients, BMJ. 349
showed that niacin, fibrates and CETP inhibitors have not been effective (2014) g4379.
[5] B.F. Voight, G.M. Peloso, M. Orho-Melander, R. Frikke-Schmidt, M. Barbalic, M.K.
in decreasing all-cause mortality, coronary heart disease mortality, Jensen, G. Hindy, H. Holm, E.L. Ding, T. Johnson, H. Schunkert, N.J. Samani, R.
myocardial infarction or stroke in patients treated with statins [4]. Al- Clarke, J.C. Hopewell, J.F. Thompson, M. Li, G. Thorleifsson, C. Newton-Cheh, K.
though anacetrapib has shown benefits, there has been a change in Musunuru, J.P. Pirruccello, D. Saleheen, L. Chen, A. Stewart, A. Schillert, U.
Thorsteinsdottir, G. Thorgeirsson, S. Anand, J.C. Engert, T. Morgan, J. Spertus, M.
the conceptual framework that future therapies should focus on manip-
Stoll, K. Berger, N. Martinelli, D. Girelli, McKeown PP, C.C. Patterson, S.E. Epstein,
ulating HDL function rather than HDL-C levels. J. Devaney, M.S. Burnett, V. Mooser, S. Ripatti, I. Surakka, M.S. Nieminen, J.
As HDL may lose its functionality in many inflammatory and patho- Sinisalo, M.L. Lokki, M. Perola, A. Havulinna, U. de Faire, B. Gigante, E. Ingelsson,
logical conditions, the main therapeutic outcome should be the im- T. Zeller, P. Wild, P.I. de Bakker, O.H. Klungel, A.H. Maitland-van der Zee, B.J. Peters,
A. de Boer, D.E. Grobbee, P.W. Kamphuisen, V.H. Deneer, C.C. Elbers, N.C. Onland-
provement of HDL function, rather than simply raising its plasma Moret, M.H. Hofker, C. Wijmenga, W.M. Verschuren, J.M. Boer, Y.T. van der Schouw,
concentration. Although there are controversial data and opinions A. Rasheed, P. Frossard, S. Demissie, C. Willer, R. Do, J.M. Ordovas, G.R. Abecasis, M.
about the effectiveness of reconstituted HDL and apoA-I mimetic pep- Boehnke, K.L. Mohlke, M.J. Daly, C. Guiducci, N.P. Burtt, A. Surti, E. Gonzalez, S. Pur-
cell, S. Gabriel, J. Marrugat, J. Peden, J. Erdmann, P. Diemert, C. Willenborg, I.R.
tides, this approach seems one of the most promising areas for targeting Konig, M. Fischer, C. Hengstenberg, A. Ziegler, I. Buysschaert, D. Lambrechts, F.
improvements in HDL function. Clinical evidence for the use of Van de Werf, K.A. Fox, N.E. El Mokhtari, D. Rubin, J. Schrezenmeir, S. Schreiber, A.
reconstituted HDL is still limited and difficult to interpret as each Schafer, J. Danesh, S. Blankenberg, R. Roberts, R. McPherson, H. Watkins, A.S. Hall,
K. Overvad, E. Rimm, E. Boerwinkle, A. Tybjaerg-Hansen, L.A. Cupples, M.P. Reilly,
study has focussed on different patients and conditions, and there is O. Melander, P.M. Mannucci, D. Ardissino, D. Siscovick, R. Elosua, K. Stefansson,
no consistency in the composition of the different reconstituted HDL C.J. O’Donnell, V. Salomaa, D.J. Rader, L. Peltonen, S.M. Schwartz, D. Altshuler, S.
particles. Accumulating evidence also suggests that the phospholipid Kathiresan, Plasma HDL cholesterol and risk of myocardial infarction: a mendelian
randomisation study, Lancet (London, England) 380 (9841) (2012) 572–580.
component of rHDL plays an important, and previously overlooked,
[6] C. Constantinou, E.A. Karavia, E. Xepapadaki, P.I. Petropoulou, E. Papakosta, M.
role in HDL functionality as highlighted in [382]. Additional and more Karavyraki, E. Zvintzou, V. Theodoropoulos, S. Filou, A. Hatziri, C. Kalogeropoulou,
elaborate clinical trials are still needed to reliably evaluate the full po- G. Panayiotakopoulos, K.E. Kypreos, Advances in high-density lipoprotein physiol-
ogy: surprises, overturns, and promises, Am. J. Physiol. Endocrinol. Metab. 310 (1)
tential of this therapeutic approach.
(2016) E1–e14.
Additionally, it would be worthwhile to investigate whether existing [7] E.A. Karavia, E. Zvintzou, P.I. Petropoulou, E. Xepapadaki, C. Constantinou, K.E.
therapies improve HDL functionality in addition to increasing HDL-C Kypreos, HDL quality and functionality: what can proteins and genes predict? Ex-
plasma levels. Most clinical studies have not focussed on functionality, pert. Rev. Cardiovasc. Ther. 12 (4) (2014) 521–532.
[8] M. Macheboeuf, Recherches sur les phosphoaminolipides du sérum sanguin. Na-
or this has only been evaluated as a secondary finding. Once again, ture des phospholipides liés aux albumines du sérum de Cheval à l’état de
this emphasizes the need for evaluation of HDL functionality as a pri- cenapses acido-précipitables, Bull. Soc. Chim. Biol. (Paris). 11 (1929) 485–503.
mary objective in future clinical studies, as also highlighted in recent [9] J.W. Gofman, F.T. Lindgren, H. Elliott, Ultracentrifugal studies of lipoproteins of
human serum, J. Biol. Chem. 179 (2) (1949) 973–979.
other reviews [383,384].
[10] R.J. Havel, H.A. Eder, J.H. Bragdon, The distribution and chemical composition of ul-
The necessity to focus on HDL function does not only hold tracentrifugally separated lipoproteins in human serum, J. Clin. Invest. 34 (9)
true for therapeutic strategies, but also for diagnostic purposes. (1955) 1345–1353.
HDL-C levels per se do not necessary reflect the potential of HDL [11] H.N. Baker, R.L. Jackson, A.M. Gotto Jr., Isolation and characterization of the cyano-
gen bromide fragments from the high-density apolipoprotein glutamine I, Bio-
to exert beneficial effects, stressing the importance of HDL- chemistry. 12 (20) (1973) 3866–3871.
composition/function testing in every clinical setting. Therefore, [12] S.E. Lux, K.M. John, H.B. Brewer Jr., Isolation and characterization of apoLp-Gln-II
future studies should not only focus on raising HDL-C levels, but (apoA-II), a plasma high density apolipoprotein containing two identical polypep-
tide chains, J. Biol. Chem. 247 (23) (1972) 7510–7518.
especially concentrate on improving HDL function to provide use-
[13] P.J. Blanche, E.L. Gong, T.M. Forte, A.V. Nichols, Characterization of human high-
ful diagnostic and therapeutic approaches for cardiovascular and density lipoproteins by gradient gel electrophoresis, Biochim. Biophys. Acta 665
many other diseases [385]. (3) (1981) 408–419.
[14] S.T. Kunitake, K.J. La Sala, J.P. Kane, Apolipoprotein A-I-containing lipoproteins with
pre-beta electrophoretic mobility, J. Lipid Res. 26 (5) (1985) 549–555.
Sources of funding [15] A. von Eckardstein, Y. Huang, G. Assmann, Physiological role and clinical relevance
of high-density lipoprotein subclasses, Curr. Opin. Lipidol. 5 (6) (1994) 404–416.
This work is supported by a grant from the Interdisciplinary Center [16] K.A. Rye, M.A. Clay, P.J. Barter, Remodelling of high density lipoproteins by plasma
factors, Atherosclerosis. 145 (2) (1999) 227–238.
for Clinical Research within the faculty of Medicine at the RWTH Aachen
[17] A. Kontush, M. Lhomme, M.J. Chapman, Unraveling the complexities of the HDL
University, the DZHK (German Centre for Cardiovascular Research) and lipidome, J. Lipid Res. 54 (11) (2013) 2950–2963.
the BMBF (German Ministry of Education and Research), and NWO- [18] A. Kontush, M. Lindahl, M. Lhomme, L. Calabresi, M.J. Chapman, W.S. Davidson,
ZonMw Veni (91619053) to E.P.C.v.d.V; by the DFG (SFB/TRR219-TP- Structure of HDL: particle subclasses and molecular components, Handb. Exp.
Pharmacol. 224 (2015) 3–51.
C09) to S.Z. and (SFB/TRR219-TP-S03) to V.J. [19] D.J. Rader, H.H. Hobbs, Disorders of Lipoprotein Metabolism, 2015.
[20] B. Shao, J.W. Heinecke, Quantifying HDL proteins by mass spectrometry: how
Declaration of Competing Interest many proteins are there and what are their functions? Expert Rev. Proteomics.
15 (1) (2018) 31–40.
[21] J.M. Timmins, J.Y. Lee, E. Boudyguina, K.D. Kluckman, L.R. Brunham, A. Mulya,
None. A.K. Gebre, J.M. Coutinho, P.L. Colvin, T.L. Smith, M.R. Hayden, N. Maeda, J.S.
Parks, Targeted inactivation of hepatic Abca1 causes profound
hypoalphalipoproteinemia and kidney hypercatabolism of apoA-I, J. Clin. In-
Acknowledgements
vest. 115 (5) (2005) 1333–1342.
[22] L.R. Brunham, J.K. Kruit, J. Iqbal, C. Fievet, J.M. Timmins, T.D. Pape, B.A. Coburn, N.
All illustrations are made with the help from [386]. Bissada, B. Staels, A.K. Groen, M.M. Hussain, J.S. Parks, F. Kuipers, M.R. Hayden,

110
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

Intestinal ABCA1 directly contributes to HDL biogenesis in vivo, J. Clin. Invest. 116 [49] S. Zieman, D. Kass, Advanced glycation end product cross-linking: pathophysio-
(4) (2006) 1052–1062. logic role and therapeutic target in cardiovascular disease, Congestive Heart Fail.
[23] E. Favari, A. Chroni, U.J. Tietge, I. Zanotti, J.C. Escola-Gil, F. Bernini, Cholesterol efflux (Greenwich, Conn) 10 (3) (2004) 144–149(quiz 50-1).
and reverse cholesterol transport, Handb. Exp. Pharmacol. 224 (2015) 181–206. [50] E. Nobecourt, M.J. Davies, B.E. Brown, L.K. Curtiss, D.J. Bonnet, F. Charlton, A.S.
[24] V.I. Zannis, A. Chroni, M. Krieger, Role of apoA-I, ABCA1, LCAT, and SR-BI in the bio- Januszewski, A.J. Jenkins, P.J. Barter, K.A. Rye, The impact of glycation on apolipo-
genesis of HDL, J. Mol. Med. (Berlin, Germany) 84 (4) (2006) 276–294. protein A-I structure and its ability to activate lecithin:cholesterol acyltransferase,
[25] D. Masson, X.C. Jiang, L. Lagrost, A.R. Tall, The role of plasma lipid transfer proteins Diabetologia. 50 (3) (2007) 643–653.
in lipoprotein metabolism and atherogenesis, J. Lipid Res. 50 (Suppl) (2009) [51] E. Nobecourt, F. Tabet, G. Lambert, R. Puranik, S. Bao, L. Yan, M.J. Davies, B.E. Brown,
S201–S206. A.J. Jenkins, G.J. Dusting, D.J. Bonnet, L.K. Curtiss, P.J. Barter, K.A. Rye, Nonenzymatic
[26] N. Settasatian, M. Duong, L.K. Curtiss, C. Ehnholm, M. Jauhiainen, J. Huuskonen, K.A. glycation impairs the antiinflammatory properties of apolipoprotein A-I,
Rye, The mechanism of the remodeling of high density lipoproteins by phospho- Arterioscler. Thromb. Vasc. Biol. 30 (4) (2010) 766–772.
lipid transfer protein, J. Biol. Chem. 276 (29) (2001) 26898–26905. [52] S.R. Kashyap, A. Osme, S. Ilchenko, M. Golizeh, K. Lee, S. Wang, J. Bena, S.F. Previs,
[27] L. Camont, M.J. Chapman, A. Kontush, Biological activities of HDL subpopulations J.D. Smith, T. Kasumov, Glycation reduces the stability of ApoAI and increases
and their relevance to cardiovascular disease, Trends Mol. Med. 17 (10) (2011) HDL dysfunction in diet-controlled type 2 diabetes, J. Clin. Endocrinol. Metab.
594–603. 103 (2) (2018) 388–396.
[28] A. Pirillo, G.D. Norata, A.L. Catapano, High-density lipoprotein subfractions–what [53] J.E. Cole, I. Park, D.J. Ahern, C. Kassiteridi, D. Danso Abeam, M.E. Goddard, P. Green,
the clinicians need to know, Cardiology. 124 (2) (2013) 116–125. P. Maffia, C. Monaco, Immune cell census in murine atherosclerosis: cytometry by
[29] M. Zhang, R. Charles, H. Tong, L. Zhang, M. Patel, F. Wang, M.J. Rames, A. Ren, K.-A. time of flight illuminates vascular myeloid cell diversity, Cardiovasc. Res. 114 (10)
Rye, X. Qiu, D.G. Johns, M.A. Charles, G. Ren, HDL surface lipids mediate CETP bind- (2018) 1360–1371.
ing as revealed by electron microscopy and molecular dynamics simulation, Sci. [54] Z.A. Fayad, F.K. Swirski, C. Calcagno, C.S. Robbins, W. Mulder, J.C. Kovacic, Monocyte
Rep. 5 (2015)8741-. and macrophage dynamics in the cardiovascular system: JACC macrophage in CVD
[30] W.V. Rodrigueza, S.T. Thuahnai, R.E. Temel, S. Lund-Katz, M.C. Phillips, D.L. series (part 3), J. Am. Coll. Cardiol. 72 (18) (2018) 2198–2212.
Williams, Mechanism of scavenger receptor class B type I-mediated selective up- [55] K.J. Lavine, A.R. Pinto, S. Epelman, B.J. Kopecky, X. Clemente-Casares, J. Godwin, N.
take of cholesteryl esters from high density lipoprotein to adrenal cells, J. Biol. Rosenthal, J.C. Kovacic, The macrophage in cardiac homeostasis and disease: JACC
Chem. 274 (29) (1999) 20344–20350. macrophage in CVD series (part 4), J. Am. Coll. Cardiol. 72 (18) (2018) 2213–2230.
[31] A. von Eckardstein, J.R. Nofer, G. Assmann, High density lipoproteins and arterio- [56] K.J. Moore, S. Koplev, E.A. Fisher, I. Tabas, J.L.M. Bjorkegren, A.C. Doran, J.C. Kovacic,
sclerosis. Role of cholesterol efflux and reverse cholesterol transport, Arterioscler. Macrophage trafficking, inflammatory resolution, and genomics in atherosclerosis:
Thromb. Vasc. Biol. 21 (1) (2001) 13–27. JACC macrophage in CVD series (part 2), J. Am. Coll. Cardiol. 72 (18) (2018)
[32] A. Jahangiri, D.J. Rader, D. Marchadier, L.K. Curtiss, D.J. Bonnet, K.A. Rye, Evidence 2181–2197.
that endothelial lipase remodels high density lipoproteins without mediating the [57] J.W. Williams, C. Giannarelli, A. Rahman, G.J. Randolph, J.C. Kovacic, Macrophage bi-
dissociation of apolipoprotein A-I, J. Lipid Res. 46 (5) (2005) 896–903. ology, classification, and phenotype in cardiovascular disease: JACC macrophage in
[33] S.M. Hammad, S. Stefansson, W.O. Twal, C.J. Drake, P. Fleming, A. Remaley, H.B. CVD series (part 1), J. Am. Coll. Cardiol. 72 (18) (2018) 2166–2180.
Brewer Jr., W.S. Argraves, Cubilin, the endocytic receptor for intrinsic factor- [58] S. Ghosh, Macrophage cholesterol homeostasis and metabolic diseases: critical role
vitamin B(12) complex, mediates high-density lipoprotein holoparticle endocyto- of cholesteryl ester mobilization, Expert. Rev. Cardiovasc. Ther. 9 (3) (2011)
sis, Proc. Natl. Acad. Sci. U. S. A. 96 (18) (1999) 10158–10163. 329–340.
[34] H. Yang, A.B. Fogo, V. Kon, Kidneys: key modulators of high-density lipoprotein [59] J. Nagenborg, P. Goossens, E.A.L. Biessen, M. Donners, Heterogeneity of atheroscle-
levels and function, Curr. Opin. Nephrol. Hypertens. 25 (3) (2016) 174–179. rotic plaque macrophage origin, phenotype and functions: implications for treat-
[35] G.A. Francis, High density lipoprotein oxidation: in vitro susceptibility and poten- ment, Eur. J. Pharmacol. 816 (2017) 14–24.
tial in vivo consequences, Biochim. Biophys. Acta 1483 (2) (2000) 217–235. [60] D. De Nardo, L.I. Labzin, H. Kono, R. Seki, S.V. Schmidt, M. Beyer, D. Xu, S. Zimmer, C.
[36] U. Panzenboeck, S. Raitmayer, H. Reicher, H. Lindner, O. Glatter, E. Malle, W. Sattler, Lahrmann, F.A. Schildberg, J. Vogelhuber, M. Kraut, T. Ulas, A. Kerksiek, W. Krebs, N.
Effects of reagent and enzymatically generated hypochlorite on physicochemical Bode, A. Grebe, M.L. Fitzgerald, N.J. Hernandez, B.R. Williams, P. Knolle, M.
and metabolic properties of high density lipoproteins, J. Biol. Chem. 272 (47) Kneilling, M. Rocken, D. Lutjohann, S.D. Wright, J.L. Schultze, E. Latz, High-density
(1997) 29711–29720. lipoprotein mediates anti-inflammatory reprogramming of macrophages via the
[37] L. Zheng, M. Settle, G. Brubaker, D. Schmitt, S.L. Hazen, J.D. Smith, M. Kinter, Local- transcriptional regulator ATF3, Nat. Immunol. 15 (2) (2014) 152–160.
ization of nitration and chlorination sites on apolipoprotein A-I catalyzed by [61] P. Fotakis, V. Kothari, D.G. Thomas, M. Westerterp, M.M. Molusky, E. Altin, S.
myeloperoxidase in human atheroma and associated oxidative impairment in Abramowicz, N. Wang, Y. He, J.W. Heinecke, K.E. Bornfeldt, A.R. Tall, Anti-
ABCA1-dependent cholesterol efflux from macrophages, J. Biol. Chem. 280 (1) inflammatory effects of HDL (high-density lipoprotein) in macrophages predomi-
(2005) 38–47. nate over proinflammatory Effects in atherosclerotic plaques, Arterioscler.
[38] A. Undurti, Y. Huang, J.A. Lupica, J.D. Smith, J.A. DiDonato, S.L. Hazen, Modification Thromb. Vasc. Biol. 39 (12) (2019) 253–272ATVBAHA119313253.
of high density lipoprotein by myeloperoxidase generates a pro-inflammatory par- [62] M. Suzuki, D.K. Pritchard, L. Becker, A.N. Hoofnagle, N. Tanimura, T.K. Bammler, R.P.
ticle, J. Biol. Chem. 284 (45) (2009) 30825–30835. Beyer, R. Bumgarner, T. Vaisar, M.C. de Beer, F.C. de Beer, K. Miyake, J.F. Oram, J.W.
[39] R.L. Levine, B.S. Berlett, J. Moskovitz, L. Mosoni, E.R. Stadtman, Methionine residues Heinecke, High-density lipoprotein suppresses the type I interferon response, a
may protect proteins from critical oxidative damage, Mech. Ageing Dev. 107 (3) family of potent antiviral immunoregulators, in macrophages challenged with lipo-
(1999) 323–332. polysaccharide, Circulation. 122 (19) (2010) 1919–1927.
[40] A. von Eckardstein, M. Walter, H. Holz, A. Benninghoven, G. Assmann, Site-specific [63] E.P.C. van der Vorst, K. Theodorou, Y. Wu, M.A. Hoeksema, P. Goossens, C.A. Bursill,
methionine sulfoxide formation is the structural basis of chromatographic hetero- T. Aliyev, L.F.A. Huitema, S.W. Tas, I.M.J. Wolfs, M.J.E. Kuijpers, M.J. Gijbels, C.G.
geneity of apolipoproteins A-I, C-II, and C-III, J. Lipid Res. 32 (9) (1991) 1465–1476. Schalkwijk, D.P.Y. Koonen, S. Abdollahi-Roodsaz, K. McDaniels, C.C. Wang, M.
[41] Y.Q. Wong, K.J. Binger, G.J. Howlett, M.D. Griffin, Methionine oxidation induces am- Leitges, T. Lawrence, J. Plat, M. Van Eck, K.A. Rye, L. Touqui, M.P.J. de Winther,
yloid fibril formation by full-length apolipoprotein A-I, Proc. Natl. Acad. Sci. U. S. A. E.A.L. Biessen, M. Donners, High-density lipoproteins exert pro-inflammatory ef-
107 (5) (2010) 1977–1982. fects on macrophages via passive cholesterol depletion and PKC-NF-kappaB/
[42] A. Witkowski, S. Carta, R. Lu, S. Yokoyama, A. Rubartelli, G. Cavigiolio, Oxidation of STAT1-IRF1 signaling, Cell Metab. 25 (1) (2017) 197–207.
methionine residues in human apolipoprotein A-I generates a potent pro- [64] K.A. Smoak, J.J. Aloor, J. Madenspacher, B.A. Merrick, J.B. Collins, X. Zhu, G.
inflammatory molecule, J. Biol. Chem. 294 (10) (2019) 3634–3646. Cavigiolio, M.N. Oda, J.S. Parks, M.B. Fessler, Myeloid differentiation primary re-
[43] R. Torosantucci, C. Schoneich, W. Jiskoot, Oxidation of therapeutic proteins and sponse protein 88 couples reverse cholesterol transport to inflammation, Cell
peptides: structural and biological consequences, Pharm. Res. 31 (3) (2014) Metab. 11 (6) (2010) 493–502.
541–553. [65] E.P.C. van der Vorst, K. Theodorou, E.A.L. Biessen, M. Donners, Disease- or storage-
[44] G. Uchida, Y. Mino, T. Suzuki, J.I. Ikeda, T. Suzuki, K. Takenaka, Y. Setsuhara, Decom- associated structural modifications are unlikely to explain HDL pro-inflammatory
position and oxidation of methionine and tryptophan following irradiation with a effects on macrophages, Cell Metab. 26 (1) (2017) 4–5.
nonequilibrium plasma jet and applications for killing cancer cells, Sci. Rep. 9 (1) [66] L. Yvan-Charvet, C. Welch, T.A. Pagler, M. Ranalletta, M. Lamkanfi, S. Han, M.
(2019) 6625. Ishibashi, R. Li, N. Wang, A.R. Tall, Increased inflammatory gene expression in
[45] Y. Huang, J.A. DiDonato, B.S. Levison, D. Schmitt, L. Li, Y. Wu, J. Buffa, T. Kim, G.S. ABC transporter-deficient macrophages: free cholesterol accumulation, increased
Gerstenecker, X. Gu, C.S. Kadiyala, Z. Wang, M.K. Culley, J.E. Hazen, A.J. Didonato, signaling via toll-like receptors, and neutrophil infiltration of atherosclerotic le-
X. Fu, S.Z. Berisha, D. Peng, T.T. Nguyen, S. Liang, C.C. Chuang, L. Cho, E.F. Plow, sions, Circulation. 118 (18) (2008) 1837–1847.
P.L. Fox, V. Gogonea, W.H. Tang, J.S. Parks, E.A. Fisher, J.D. Smith, S.L. Hazen, An [67] K. Simons, E. Ikonen, Functional rafts in cell membranes, Nature. 387 (6633)
abundant dysfunctional apolipoprotein A1 in human atheroma, Nat. Med. 20 (2) (1997) 569–572.
(2014) 193–203. [68] M. Koseki, K. Hirano, D. Masuda, C. Ikegami, M. Tanaka, A. Ota, J.C. Sandoval, Y.
[46] M. Ehrenshaft, L.J. Deterding, R.P. Mason, Tripping up Trp: modification of protein Nakagawa-Toyama, S.B. Sato, T. Kobayashi, Y. Shimada, Y. Ohno-Iwashita, F.
tryptophan residues by reactive oxygen species, modes of detection, and biological Matsuura, I. Shimomura, S. Yamashita, Increased lipid rafts and accelerated
consequences, Free Radic. Biol. Med. 89 (2015) 220–228. lipopolysaccharide-induced tumor necrosis factor-alpha secretion in Abca1-
[47] C. Schoneich, Novel chemical degradation pathways of proteins mediated by tryp- deficient macrophages, J. Lipid Res. 48 (2) (2007) 299–306.
tophan oxidation: tryptophan side chain fragmentation, J. Pharm. Pharmacol. 70 [69] X. Zhu, J.Y. Lee, J.M. Timmins, J.M. Brown, E. Boudyguina, A. Mulya, A.K. Gebre, M.C.
(5) (2018) 655–665. Willingham, E.M. Hiltbold, N. Mishra, N. Maeda, J.S. Parks, Increased cellular free
[48] Z. Wang, S.J. Nicholls, E.R. Rodriguez, O. Kummu, S. Horkko, J. Barnard, W.F. cholesterol in macrophage-specific Abca1 knock-out mice enhances pro-
Reynolds, E.J. Topol, J.A. DiDonato, S.L. Hazen, Protein carbamylation links inflam- inflammatory response of macrophages, J. Biol. Chem. 283 (34) (2008)
mation, smoking, uremia and atherogenesis, Nat. Med. 13 (10) (2007) 1176–1184. 22930–22941.

111
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

[70] X. Zhu, J.S. Owen, M.D. Wilson, H. Li, G.L. Griffiths, M.J. Thomas, E.M. Hiltbold, M.B. release in mature human dendritic cells for emergence of Th2 immune responses,
Fessler, J.S. Parks, Macrophage ABCA1 reduces MyD88-dependent toll-like receptor FASEB J. 16 (6) (2002) 625–627.
trafficking to lipid rafts by reduction of lipid raft cholesterol, J. Lipid Res. 51 (11) [95] K. Schaper, M. Kietzmann, W. Baumer, Sphingosine-1-phosphate differently regu-
(2010) 3196–3206. lates the cytokine production of IL-12, IL-23 and IL-27 in activated murine bone
[71] L.E. Smythies, C.R. White, A. Maheshwari, M.N. Palgunachari, G.M. marrow derived dendritic cells, Mol. Immunol. 59 (1) (2014) 10–18.
Anantharamaiah, M. Chaddha, A.R. Kurundkar, G. Datta, Apolipoprotein A-I mi- [96] L. Perrin-Cocon, O. Diaz, M. Carreras, S. Dollet, A. Guironnet-Paquet, P. Andre, V.
metic 4F alters the function of human monocyte-derived macrophages, Am. J. Lotteau, High-density lipoprotein phospholipids interfere with dendritic cell Th1
Physiol. Cell Physiol. 298 (6) (2010) C1538–C1548. functional maturation, Immunobiology. 217 (1) (2012) 91–99.
[72] C.R. White, L.E. Smythies, D.K. Crossman, M.N. Palgunachari, G.M. Anantharamaiah, [97] L. Jonasson, J. Holm, O. Skalli, G. Bondjers, G.K. Hansson, Regional accumulations of
G. Datta, Regulation of pattern recognition receptors by the apolipoprotein A-I mi- T cells, macrophages, and smooth muscle cells in the human atherosclerotic
metic peptide 4F, Arterioscler. Thromb. Vasc. Biol. 32 (11) (2012) 2631–2639. plaque, Arteriosclerosis (Dallas, Tex). 6 (2) (1986) 131–138.
[73] B. Levkau, HDL-S1P: cardiovascular functions, disease-associated alterations, and [98] A.K. Robertson, M. Rudling, X. Zhou, L. Gorelik, R.A. Flavell, G.K. Hansson, Disrup-
therapeutic applications, Front. Pharmacol. 6 (2015) 243. tion of TGF-beta signaling in T cells accelerates atherosclerosis, J. Clin. Invest. 112
[74] J.E. Hughes, S. Srinivasan, K.R. Lynch, R.L. Proia, P. Ferdek, C.C. Hedrick, Sphingo- (9) (2003) 1342–1350.
sine-1-phosphate induces an antiinflammatory phenotype in macrophages, Circ. [99] Z. Mallat, A. Gojova, C. Marchiol-Fournigault, B. Esposito, C. Kamate, R. Merval, D.
Res. 102 (8) (2008) 950–958. Fradelizi, A. Tedgui, Inhibition of transforming growth factor-beta signaling accel-
[75] A.I. Duenas, M. Aceves, I. Fernandez-Pisonero, C. Gomez, A. Orduna, M.S. Crespo, C. erates atherosclerosis and induces an unstable plaque phenotype in mice, Circ.
Garcia-Rodriguez, Selective attenuation of toll-like receptor 2 signalling may ex- Res. 89 (10) (2001) 930–934.
plain the atheroprotective effect of sphingosine 1-phosphate, Cardiovasc. Res. 79 [100] D. Ru, H. Zhiqing, Z. Lin, W. Feng, Z. Feng, Z. Jiayou, R. Yusheng, F. Min, L. Chun, W.
(3) (2008) 537–544. Zonggui, Oxidized high-density lipoprotein accelerates atherosclerosis progression
[76] R. Feuerborn, S. Becker, F. Poti, P. Nagel, M. Brodde, H. Schmidt, C. Christoffersen, U. by inducing the imbalance between treg and teff in LDLR knockout mice, APMIS
Ceglarek, R. Burkhardt, J.R. Nofer, High density lipoprotein (HDL)-associated sphin- 123 (5) (2015) 410–421.
gosine 1-phosphate (S1P) inhibits macrophage apoptosis by stimulating STAT3 ac- [101] A.J. Wilhelm, M. Zabalawi, J.S. Owen, D. Shah, J.M. Grayson, A.S. Major, S. Bhat, D.P.
tivity and survivin expression, Atherosclerosis. 257 (2017) 29–37. Gibbs Jr., M.J. Thomas, M.G. Sorci-Thomas, Apolipoprotein A-I modulates regula-
[77] R.S. Rosenson, H.B. Brewer Jr., B.J. Ansell, P. Barter, M.J. Chapman, J.W. Heinecke, A. tory T cells in autoimmune LDLr−/−, ApoA-I−/− mice, J. Biol. Chem. 285 (46)
Kontush, A.R. Tall, N.R. Webb, Dysfunctional HDL and atherosclerotic cardiovascu- (2010) 36158–36169.
lar disease, Nat. Rev. Cardiol. 13 (1) (2016) 48–60. [102] C.M. Rueda, A.L. Rodriguez-Perea, M. Moreno-Fernandez, C.M. Jackson, J.T.
[78] D.X. Wang, H. Liu, L.R. Yan, Y.P. Zhang, X.Y. Guan, Z.M. Xu, Y.H. Jia, Y.S. Li, The rela- Melchior, W.S. Davidson, C.A. Chougnet, High density lipoproteins selectively pro-
tionship between serum amyloid A and apolipoprotein A-I in high-density lipopro- mote the survival of human regulatory T cells, J. Lipid Res. 58 (8) (2017)
tein isolated from patients with coronary heart disease, Chin. Med. J. 126 (19) 1514–1523.
(2013) 3656–3661. [103] D.E. Gaddis, L.E. Padgett, R. Wu, C. McSkimming, V. Romines, A.M. Taylor, C.A.
McNamara, M. Kronenberg, S. Crotty, M.J. Thomas, M.G. Sorci-Thomas, C.C.
[79] M. Tolle, T. Huang, M. Schuchardt, V. Jankowski, N. Prufer, J. Jankowski, U.J. Tietge,
Hedrick, Apolipoprotein AI prevents regulatory to follicular helper T cell switching
W. Zidek, M. van der Giet, High-density lipoprotein loses its anti-inflammatory ca-
during atherosclerosis, Nat. Commun. 9 (1) (2018) 1095.
pacity by accumulation of pro-inflammatory-serum amyloid a, Cardiovasc. Res. 94
(1) (2012) 154–162. [104] G. Liu, K. Yang, S. Burns, S. Shrestha, H. Chi, The S1P(1)-mTOR axis directs the re-
ciprocal differentiation of T(H)1 and T(reg) cells, Nat. Immunol. 11 (11) (2010)
[80] S. Zewinger, C. Drechsler, M.E. Kleber, A. Dressel, J. Riffel, S. Triem, M. Lehmann, C.
1047–1056.
Kopecky, M.D. Saemann, P.M. Lepper, G. Silbernagel, H. Scharnagl, A. Ritsch, B.
[105] F. Sala, A.L. Catapano, G.D. Norata, High density lipoproteins and atherosclerosis:
Thorand, T. de las Heras Gala, S. Wagenpfeil, W. Koenig, A. Peters, U. Laufs, C.
emerging aspects, JGC. 9 (4) (2012) 401–407.
Wanner, D. Fliser, T. Speer, W. Marz, Serum amyloid A: high-density lipoproteins
[106] P.S. Kabouridis, E.C. Jury, Lipid rafts and T-lymphocyte function: implications for
interaction and cardiovascular risk, Eur. Heart J. 36 (43) (2015) 3007–3016.
autoimmunity, FEBS Lett. 582 (27) (2008) 3711–3718.
[81] A. Artl, G. Marsche, S. Lestavel, W. Sattler, E. Malle, Role of serum amyloid A during
[107] A.J. Wilhelm, M. Zabalawi, J.M. Grayson, A.E. Weant, A.S. Major, J. Owen, M.
metabolism of acute-phase HDL by macrophages, Arterioscler. Thromb. Vasc. Biol.
Bharadwaj, R. Walzem, L. Chan, K. Oka, M.J. Thomas, M.G. Sorci-Thomas, Apolipo-
20 (3) (2000) 763–772.
protein A-I and its role in lymphocyte cholesterol homeostasis and autoimmunity,
[82] J.G. Tsun, S.W. Shiu, Y. Wong, S. Yung, T.M. Chan, K.C. Tan, Impact of serum amyloid
Arterioscler. Thromb. Vasc. Biol. 29 (6) (2009) 843–849.
A on cellular cholesterol efflux to serum in type 2 diabetes mellitus, Atherosclero-
[108] V.A. Blaho, S. Galvani, E. Engelbrecht, C. Liu, S.L. Swendeman, M. Kono, R.L. Proia, L.
sis. 231 (2) (2013) 405–410.
Steinman, M.H. Han, T. Hla, HDL-bound sphingosine-1-phosphate restrains
[83] T. Vaisar, C. Tang, I. Babenko, P. Hutchins, J. Wimberger, A.F. Suffredini, J.W.
lymphopoiesis and neuroinflammation, Nature. 523 (7560) (2015) 342–346.
Heinecke, Inflammatory remodeling of the HDL proteome impairs cholesterol ef-
[109] A.P. Sage, D. Tsiantoulas, C.J. Binder, Z. Mallat, The role of B cells in atherosclerosis,
flux capacity, J. Lipid Res. 56 (8) (2015) 1519–1530.
Nat. Rev. Cardiol. 16 (3) (2019) 180–196.
[84] J.H. Qiao, P.Z. Xie, M.C. Fishbein, J. Kreuzer, T.A. Drake, L.L. Demer, A.J. Lusis, Pathol-
[110] N. Gupta, A.L. DeFranco, Lipid rafts and B cell signaling, Semin. Cell Dev. Biol. 18 (5)
ogy of atheromatous lesions in inbred and genetically engineered mice. Genetic
(2007) 616–626.
determination of arterial calcification, Arterioscler. Thrombosis. 14 (9) (1994)
[111] R.P. Mensink, Effects of Saturated Fatty Acids on Serum Lipids and Lipoproteins: A
1480–1497.
Systematic Review and Regression Analysis, World Health Organization, 2016.
[85] R.L. Meek, S. Urieli-Shoval, E.P. Benditt, Expression of apolipoprotein serum amy- [112] I. Brouwer, Effect of trans-fatty acid intake on blood lipids and lipoproteins: a sys-
loid a mRNA in human atherosclerotic lesions and cultured vascular cells: implica- tematic review and meta-regression analysis, World Health Org. (2016).
tions for serum amyloid a function, Proc. Natl. Acad. Sci. U. S. A. 91 (8) (1994)
[113] L. Smolders, J. Plat, R.P. Mensink, Dietary strategies and novel pharmaceutical ap-
3186–3190.
proaches targeting serum ApoA-I metabolism: a systematic overview, J. Nutr.
[86] T. Yamada, T. Miida, T. Yamaguchi, Y. Itoh, Effect of serum amyloid A on cellular af- Metab. 2017 (2017) 5415921.
finity of low density lipoprotein, Eur. J. Clin. Chem. Clin. Biochem. 35 (6) (1997) [114] Z.H. Yang, M. Amar, M. Sampson, A.B. Courville, A.V. Sorokin, S.M. Gordon, A.M.
421–426. Aponte, M. Stagliano, M.P. Playford, Y.P. Fu, S. Yang, N.N. Mehta, A.T. Remaley,
[87] C.Y. Han, C. Tang, M.E. Guevara, H. Wei, T. Wietecha, B. Shao, S. Subramanian, M. Comparison of Omega-3 eicosapentaenoic acid versus docosahexaenoic acid-rich
Omer, S. Wang, K.D. O’Brien, S.M. Marcovina, T.N. Wight, T. Vaisar, M.C. de Beer, fish oil supplementation on plasma lipids and lipoproteins in normolipidemic
F.C. de Beer, W.R. Osborne, K.B. Elkon, A. Chait, Serum amyloid A impairs the adults, Nutrients. 12 (3) (2020).
antiinflammatory properties of HDL, J. Clin. Invest. 126 (1) (2016) 266–281. [115] K. Ren, Z.L. Tang, Y. Jiang, Y.M. Tan, G.H. Yi, Apolipoprotein M, Clin. Chim. Acta 446
[88] M. Sato, R. Ohkawa, A. Yoshimoto, K. Yano, N. Ichimura, M. Nishimori, S. Okubo, Y. (2015) 21–29.
Yatomi, M. Tozuka, Effects of serum amyloid A on the structure and antioxidant [116] K. Nilausen, H. Meinertz, Variable lipemic response to dietary soy protein in
ability of high-density lipoprotein, Biosci. Rep. 36 (4) (2016). healthy, normolipemic men, Am. J. Clin. Nutr. 68 (6 Suppl) (1998)1380S-4S.
[89] M. Schuchardt, N. Prufer, Y. Tu, J. Herrmann, X.P. Hu, S. Chebli, K. Dahlke, W. Zidek, [117] C. Zhu, L. Sawrey-Kubicek, E. Beals, R.L. Hughes, C.H. Rhodes, R. Sacchi, A.M.
M. van der Giet, M. Tolle, Dysfunctional high-density lipoprotein activates toll-like Zivkovic, The HDL lipidome is widely remodeled by fast food versus Mediterra-
receptors via serum amyloid A in vascular smooth muscle cells, Sci. Rep. 9 (1) nean diet in 4 days, Metabolomics. 15 (8) (2019) 114.
(2019) 3421. [118] S.E. Brien, P.E. Ronksley, B.J. Turner, K.J. Mukamal, W.A. Ghali, Effect of alcohol con-
[90] K. Ni, H.C. O’Neill, The role of dendritic cells in T cell activation, Immunol. Cell Biol. sumption on biological markers associated with risk of coronary heart disease: sys-
75 (3) (1997) 223–230. tematic review and meta-analysis of interventional studies, BMJ. 342 (2011) d636.
[91] D.A. Chistiakov, I.A. Sobenin, A.N. Orekhov, Y.V. Bobryshev, Dendritic cells in ath- [119] M.S. van der Gaag, A. van Tol, L.M. Scheek, R.W. James, R. Urgert, G. Schaafsma, H.F.
erosclerotic inflammation: the complexity of functions and the peculiarities of Hendriks, Daily moderate alcohol consumption increases serum paraoxonase ac-
pathophysiological effects, Front. Physiol. 5 (2014) 196. tivity; a diet-controlled, randomised intervention study in middle-aged men, Ath-
[92] A. Zernecke, Dendritic cells in atherosclerosis: evidence in mice and humans, erosclerosis. 147 (2) (1999) 405–410.
Arterioscler. Thromb. Vasc. Biol. 35 (4) (2015) 763–770. [120] J.W. Beulens, A. Sierksma, A. van Tol, N. Fournier, T. van Gent, J.L. Paul, H.F.
[93] E. Eren, J. Yates, K. Cwynarski, S. Preston, R. Dong, C. Germain, R. Lechler, R. Huby, Hendriks, Moderate alcohol consumption increases cholesterol efflux mediated
M. Ritter, G. Lombardi, Location of major histocompatibility complex class II mole- by ABCA1, J. Lipid Res. 45 (9) (2004) 1716–1723.
cules in rafts on dendritic cells enhances the efficiency of T-cell activation and pro- [121] N. Neufingerl, Y.E. Zebregs, E.A. Schuring, E.A. Trautwein, Effect of cocoa and theo-
liferation, Scand. J. Immunol. 63 (1) (2006) 7–16. bromine consumption on serum HDL-cholesterol concentrations: a randomized
[94] M. Idzko, E. Panther, S. Corinti, A. Morelli, D. Ferrari, Y. Herouy, S. Dichmann, M. controlled trial, Am. J. Clin. Nutr. 97 (6) (2013) 1201–1209.
Mockenhaupt, P. Gebicke-Haerter, F. Di Virgilio, G. Girolomoni, J. Norgauer, Sphin- [122] D.M. Jacobs, L. Smolders, Y. Lin, N. de Roo, E.A. Trautwein, J. van Duynhoven, R.P.
gosine 1-phosphate induces chemotaxis of immature and modulates cytokine- Mensink, J. Plat, V.V. Mihaleva, Effect of theobromine consumption on serum

112
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

lipoprotein profiles in apparently healthy humans with low HDL-cholesterol con- [145] F. Heller, C. Harvengt, Effects of clofibrate, bezafibrate, fenofibrate and probucol on
centrations, Front. Mol. Biosci. 4 (2017) 59. plasma lipolytic enzymes in normolipaemic subjects, Eur. J. Clin. Pharmacol. 25 (1)
[123] L. Smolders, R.P. Mensink, M.V. Boekschoten, R.J.J. de Ridder, J. Plat, Theobromine (1983) 57–63.
does not affect postprandial lipid metabolism and duodenal gene expression, but [146] M.R. Taskinen, D.R. Sullivan, C. Ehnholm, M. Whiting, D. Zannino, R.J. Simes, A.C.
has unfavorable effects on postprandial glucose and insulin responses in humans, Keech, P.J. Barter, investigators Fs, Relationships of HDL cholesterol, ApoA-I, and
Clin. Nutrition (Edinburgh, Scotland). 37 (2) (2018) 719–727. ApoA-II with homocysteine and creatinine in patients with type 2 diabetes treated
[124] L. Smolders, R.P. Mensink, M.V. Boekschoten, R.J.J. de Ridder, J. Plat, The acute ef- with fenofibrate, Arterioscler. Thromb. Vasc. Biol. 29 (6) (2009) 950–955.
fects on duodenal gene expression in healthy men following consumption of a [147] M.A. Hossain, M. Tsujita, F.J. Gonzalez, S. Yokoyama, Effects of fibrate drugs on ex-
low-fat meal enriched with theobromine or fat, Sci. Rep. 8 (1) (2018) 1700. pression of ABCA1 and HDL biogenesis in hepatocytes, J. Cardiovasc. Pharmacol. 51
[125] S. Contreras-Duarte, P. Chen, M. Andia, S. Uribe, P. Irarrazaval, S. Kopp, S. Kern, G. (3) (2008) 258–266.
Marsche, D. Busso, C. Wadsack, A. Rigotti, Attenuation of atherogenic apo B-48-de- [148] S. Kersten, Peroxisome proliferator activated receptors and lipoprotein metabo-
pendent hyperlipidemia and high density lipoprotein remodeling induced by vita- lism, PPAR Res. 2008 (2008) 132960.
min C and E combination and their beneficial effect on lethal ischemic heart [149] N. Vu-Dac, S. Chopin-Delannoy, P. Gervois, E. Bonnelye, G. Martin, J.C. Fruchart, V.
disease in mice, Biol. Res. 51 (1) (2018) 34. Laudet, B. Staels, The nuclear receptors peroxisome proliferator-activated receptor
[126] M. Mackness, B. Mackness, Human paraoxonase-1 (PON1): gene structure and ex- alpha and Rev-erbalpha mediate the species-specific regulation of apolipoprotein
pression, promiscuous activities and multiple physiological roles, Gene. 567 (1) A-I expression by fibrates, J. Biol. Chem. 273 (40) (1998) 25713–25720.
(2015) 12–21. [150] P.J. Barter, M. Caulfield, M. Eriksson, S.M. Grundy, J.J. Kastelein, M. Komajda, J.
[127] P. Mani, A. Rohatgi, Niacin therapy, HDL cholesterol, and cardiovascular disease: is Lopez-Sendon, L. Mosca, J.C. Tardif, D.D. Waters, C.L. Shear, J.H. Revkin, K.A. Buhr,
the HDL hypothesis defunct? Curr. Atheroscler. Rep. 17 (8) (2015) 43. M.R. Fisher, A.R. Tall, B. Brewer, Effects of torcetrapib in patients at high risk for
[128] A.J. Taylor, L.E. Sullenberger, H.J. Lee, J.K. Lee, K.A. Grace, Arterial biology for the in- coronary events, N. Engl. J. Med. 357 (21) (2007) 2109–2122.
vestigation of the treatment effects of reducing cholesterol (ARBITER) 2: a double- [151] J.W. Funder, The off-target effects of torcetrapib, Endocrinology. 150 (5) (2009)
blind, placebo-controlled study of extended-release niacin on atherosclerosis pro- 2024–2026.
gression in secondary prevention patients treated with statins, Circulation. 110 [152] R.W. Clark, D. Cunningham, Y. Cong, T.A. Subashi, G.T. Tkalcevic, D.B. Lloyd, J.G.
(23) (2004) 3512–3517. Boyd, B.A. Chrunyk, G.A. Karam, X. Qiu, I.K. Wang, O.L. Francone, Assessment of
[129] T.C. Villines, E.J. Stanek, P.J. Devine, M. Turco, M. Miller, N.J. Weissman, L. Griffen, cholesteryl ester transfer protein inhibitors for interaction with proteins involved
A.J. Taylor, The ARBITER 6-HALTS trial (arterial biology for the investigation of in the immune response to infection, J. Lipid Res. 51 (5) (2010) 967–974.
the treatment effects of reducing cholesterol 6-HDL and LDL treatment strategies [153] G.G. Schwartz, A.G. Olsson, M. Abt, C.M. Ballantyne, P.J. Barter, J. Brumm, B.R.
in atherosclerosis): final results and the impact of medication adherence, dose, Chaitman, I.M. Holme, D. Kallend, L.A. Leiter, E. Leitersdorf, J.J. McMurray, H.
and treatment duration, J. Am. Coll. Cardiol. 55 (24) (2010) 2721–2726. Mundl, S.J. Nicholls, P.K. Shah, J.C. Tardif, R.S. Wright, dal OI, Effects of dalcetrapib
[130] M. Zeman, M. Vecka, F. Perlík, R. Hromádka, B. Staňková, E. Tvrzická, A. Žák, Niacin in patients with a recent acute coronary syndrome, N. Engl. J. Med. 367 (22)
in the treatment of hyperlipidemias in light of new clinical Trials: has niacin lost its (2012) 2089–2099.
place? Med. Sci. Monit. 21 (2015) 2156–2162. [154] A.M. Lincoff, S.J. Nicholls, J.S. Riesmeyer, P.J. Barter, H.B. Brewer, K.A.A. Fox, C.M.
[131] A.-H. Investigators, W.E. Boden, J.L. Probstfield, T. Anderson, B.R. Chaitman, P. Gibson, C. Granger, V. Menon, G. Montalescot, D. Rader, A.R. Tall, E. McErlean, K.
Desvignes-Nickens, K. Koprowicz, R. McBride, K. Teo, W. Weintraub, Niacin in pa- Wolski, G. Ruotolo, B. Vangerow, G. Weerakkody, S.G. Goodman, D. Conde, D.K.
tients with low HDL cholesterol levels receiving intensive statin therapy, N. Engl. J. McGuire, J.C. Nicolau, J.L. Leiva-Pons, Y. Pesant, W. Li, D. Kandath, S. Kouz, N.
Med. 365 (24) (2011) 2255–2267. Tahirkheli, D. Mason, S.E. Nissen, A. Investigators, Evacetrapib and cardiovascular
[132] Group HTC, M.J. Landray, R. Haynes, J.C. Hopewell, S. Parish, T. Aung, J. Tomson, K. outcomes in high-risk vascular disease, N. Engl. J. Med. 376 (20) (2017)
Wallendszus, M. Craig, L. Jiang, R. Collins, J. Armitage, Effects of extended-release 1933–1942.
niacin with laropiprant in high-risk patients, N. Engl. J. Med. 371 (3) (2014) [155] Group HTRC, L. Bowman, J.C. Hopewell, F. Chen, K. Wallendszus, W. Stevens, R.
203–212. Collins, S.D. Wiviott, C.P. Cannon, E. Braunwald, E. Sammons, M.J. Landray, Effects
[133] M.H. Frick, O. Elo, K. Haapa, O.P. Heinonen, P. Heinsalmi, P. Helo, J.K. Huttunen, P. of Anacetrapib in patients with atherosclerotic vascular disease, N. Engl. J. Med.
Kaitaniemi, P. Koskinen, V. Manninen, et al., Helsinki heart study: primary- 377 (13) (2017) 1217–1227.
prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety [156] L. Badimon, CETP inhibition and HDL: what is the trial REVEALing? Cardiovasc. Res.
of treatment, changes in risk factors, and incidence of coronary heart disease, N. 114 (2) (2018)e15-e6.
Engl. J. Med. 317 (20) (1987) 1237–1245. [157] B.A. Ference, J.J.P. Kastelein, H.N. Ginsberg, M.J. Chapman, S.J. Nicholls, K.K. Ray, C.J.
[134] H.B. Rubins, S.J. Robins, D. Collins, C.L. Fye, J.W. Anderson, M.B. Elam, F.H. Faas, E. Packard, U. Laufs, R.D. Brook, C. Oliver-Williams, A.S. Butterworth, J. Danesh, G.D.
Linares, E.J. Schaefer, G. Schectman, T.J. Wilt, J. Wittes, Gemfibrozil for the second- Smith, A.L. Catapano, M.S. Sabatine, Association of genetic variants related to
ary prevention of coronary heart disease in men with low levels of high-density li- CETP inhibitors and statins with lipoprotein levels and cardiovascular risk, Jama.
poprotein cholesterol. Veterans affairs high-density lipoprotein cholesterol 318 (10) (2017) 947–956.
intervention trial study group, N. Engl. J. Med. 341 (6) (1999) 410–418. [158] K.K. Ray, M. Ditmarsch, D. Kallend, E.J. Niesor, G. Suchankova, R. Upmanyu, J.
[135] Bezafibrate Infarction Prevention s, Secondary prevention by raising HDL choles- Anzures-Cabrera, V. Lehnert, M. Pauly-Evers, I. Holme, J. Stasek, M.W. van
terol and reducing triglycerides in patients with coronary artery disease, Circula- Hessen, P. Jones, The effect of cholesteryl ester transfer protein inhibition on lipids,
tion. 102 (1) (2000) 21–27. lipoproteins, and markers of HDL function after an acute coronary syndrome: the
[136] A. Keech, R.J. Simes, P. Barter, J. Best, R. Scott, M.R. Taskinen, P. Forder, A. Pillai, T. dal-ACUTE randomized trial, Eur. Heart J. 35 (27) (2014) 1792–1800.
Davis, P. Glasziou, P. Drury, Y.A. Kesaniemi, D. Sullivan, D. Hunt, P. Colman, M. [159] F. Zimetti, W.M. Freitas, A.M. Campos, M. Daher, M.P. Adorni, F. Bernini, A.C.
d’Emden, M. Whiting, C. Ehnholm, M. Laakso, investigators Fs, Effects of long- Sposito, I. Zanotti, Brazilian study on healthy a. cholesterol efflux capacity does
term fenofibrate therapy on cardiovascular events in 9795 people with type 2 di- not associate with coronary calcium, plaque vulnerability, and telomere length
abetes mellitus (the FIELD study): randomised controlled trial, Lancet (London, En- in healthy octogenarians, J. Lipid Res. 59 (4) (2018) 714–721.
gland). 366 (9500) (2005) 1849–1861. [160] S.J. Nicholls, A. Gordon, J. Johansson, K. Wolski, C.M. Ballantyne, J.J. Kastelein, A.
[137] Group AS, H.N. Ginsberg, M.B. Elam, L.C. Lovato, J.R. Crouse 3rd, L.A. Leiter, P. Linz, Taylor, M. Borgman, S.E. Nissen, Efficacy and safety of a novel oral inducer of apo-
W.T. Friedewald, J.B. Buse, H.C. Gerstein, J. Probstfield, R.H. Grimm, F. Ismail-Beigi, lipoprotein a-I synthesis in statin-treated patients with stable coronary artery dis-
J.T. Bigger, D.C. Goff Jr., W.C. Cushman, D.G. Simons-Morton, R.P. Byington, Effects ease a randomized controlled trial, J. Am. Coll. Cardiol. 57 (9) (2011) 1111–1119.
of combination lipid therapy in type 2 diabetes mellitus, N. Engl. J. Med. 362 (17) [161] D. Shishikura, Y. Kataoka, S. Honda, K. Takata, S.W. Kim, J. Andrews, P.J. Psaltis, M.
(2010) 1563–1574. Sweeney, E. Kulikowski, J. Johansson, N.C.W. Wong, S.J. Nicholls, The effect of
[138] P.J. Barter, K.A. Rye, New era of lipid-lowering drugs, Pharmacol. Rev. 68 (2) (2016) bromodomain and extra-terminal inhibitor apabetalone on attenuated coronary
458–475. atherosclerotic plaque: insights from the ASSURE trial, Am. J. Cardiovasc. Drugs
[139] A.D. Pradhan, N.P. Paynter, B.M. Everett, R.J. Glynn, P. Amarenco, M. Elam, H. 19 (1) (2019) 49–57.
Ginsberg, W.R. Hiatt, S. Ishibashi, W. Koenig, B.G. Nordestgaard, J.C. Fruchart, P. [162] D. Bailey, R. Jahagirdar, A. Gordon, A. Hafiane, S. Campbell, S. Chatur, G.S. Wagner,
Libby, P.M. Ridker, Rationale and design of the pemafibrate to reduce cardiovascu- H.C. Hansen, F.S. Chiacchia, J. Johansson, L. Krimbou, N.C. Wong, J. Genest, RVX-
lar outcomes by reducing triglycerides in patients with diabetes (PROMINENT) 208: a small molecule that increases apolipoprotein A-I and high-density lipopro-
study, Am. Heart J. 206 (2018) 80–93. tein cholesterol in vitro and in vivo, J. Am. Coll. Cardiol. 55 (23) (2010) 2580–2589.
[140] J.C. Fruchart, Selective peroxisome proliferator-activated receptor alpha modula- [163] Clinical Trials, Effect of RVX000222 on Time to Major Adverse Cardiovascular
tors (SPPARMalpha): the next generation of peroxisome proliferator-activated re- Events in High-Risk T2DM Subjects with CAD (BETonMACE), 2019, September 30.
ceptor alpha-agonists, Cardiovasc. Diabetol. 12 (2013) 82. [164] K.K. Ray, S.J. Nicholls, H.D. Ginsberg, J.O. Johansson, K. Kalantar-Zadeh, E.
[141] (NIDDK) NIoDaDaKD, Fibrates. LiverTox: Clinical and Research Information on Kulikowski, P.P. Toth, N. Wong, J.L. Cummings, M. Sweeney, G.G. Schwartz, Effect
Drug-Induced Liver Injury. Bethesda (MD): National Institute of Diabetes and Di- of selective BET protein inhibitor apabetalone on cardiovascular outcomes in pa-
gestive and Kidney Diseases, 2012. tients with acute coronary syndrome and diabetes: rationale, design, and baseline
[142] Drugs.com, Fibric Acid Derivatives, 2019, November 8. characteristics of the BETonMACE trial, Am. Heart J. 217 (2019) 72–83.
[143] C. Flores-Castillo, M. Luna-Luna, E. Carreon-Torres, V. Lopez-Olmos, S. Frias, M.A. [165] M. Navab, G.M. Anantharamaiah, S.T. Reddy, B.J. Van Lenten, A.M. Fogelman, HDL
Juarez-Oropeza, M. Franco, J.M. Fragoso, G. Vargas-Alarcon, O. Perez-Mendez, Ator- as a biomarker, potential therapeutic target, and therapy, Diabetes. 58 (12)
vastatin and fenofibrate increase the content of unsaturated acyl chains in HDL and (2009) 2711–2717.
modify in vivo kinetics of HDL-cholesteryl esters in New Zealand white rabbits, Int. [166] A. Chattopadhyay, M. Navab, G. Hough, F. Gao, D. Meriwether, V. Grijalva, J.R.
J. Mol. Sci. 20 (10) (2019). Springstead, M.N. Palgnachari, R. Namiri-Kalantari, F. Su, B.J. Van Lenten, A.C.
[144] B. Staels, J. Dallongeville, J. Auwerx, K. Schoonjans, E. Leitersdorf, J.C. Fruchart, Wagner, G.M. Anantharamaiah, R. Farias-Eisner, S.T. Reddy, A.M. Fogelman, A
Mechanism of action of fibrates on lipid and lipoprotein metabolism, Circulation. novel approach to oral apoA-I mimetic therapy, J. Lipid Res. 54 (4) (2013)
98 (19) (1998) 2088–2093. 995–1010.

113
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

[167] M. Ditiatkovski, W. D’Souza, R. Kesani, J. Chin-Dusting, J.B. de Haan, A. Remaley, D. [189] B. Shao, C. Tang, J.W. Heinecke, J.F. Oram, Oxidation of apolipoprotein A-I by
Sviridov, An apolipoprotein A-I mimetic peptide designed with a reductionist ap- myeloperoxidase impairs the initial interactions with ABCA1 required for signaling
proach stimulates reverse cholesterol transport and reduces atherosclerosis in and cholesterol export, J. Lipid Res. 51 (7) (2010) 1849–1858.
mice, PLoS One 8 (7) (2013), e68802. [190] S. Manjunatha, K. Distelmaier, S. Dasari, R.E. Carter, Y.C. Kudva, K.S. Nair, Functional
[168] I. Karalis, J.W. Jukema, HDL mimetics infusion and regression of atherosclerosis: is and proteomic alterations of plasma high density lipoproteins in type 1 diabetes
it still considered a valid therapeutic option? Curr. Cardiol. Rep. 20 (8) (2018) 66. mellitus, Metab. Clin. Exp. 65 (9) (2016) 1421–1431.
[169] M.J. Amar, W. D’Souza, S. Turner, S. Demosky, D. Sviridov, J. Stonik, J. Luchoomun, J. [191] E. Sampaio, D.S. Barbosa, T.L. Mazzuco, V.S. Nunes, M. Passarelli, E.R. Nakandakare,
Voogt, M. Hellerstein, D. Sviridov, A.T. Remaley, 5A apolipoprotein mimetic peptide A.J. Carrilho, Impaired antioxidant action of high density lipoprotein in patients
promotes cholesterol efflux and reduces atherosclerosis in mice, J. Pharmacol. Exp. with type 1 diabetes with normoalbuminuria and microalbuminuria, Diabetes
Ther. 334 (2) (2010) 634–641. Res. Clin. Pract. 99 (3) (2013) 321–326.
[170] F. Tabet, A.T. Remaley, A.I. Segaliny, J. Millet, L. Yan, S. Nakhla, P.J. Barter, K.A. Rye, G. [192] T. Vaisar, J.E. Kanter, J. Wimberger, A.D. Irwin, J. Gauthier, E. Wolfson, V. Bahnam,
Lambert, The 5A apolipoprotein A-I mimetic peptide displays antiinflammatory I.H. Wu, H. Shah, H.A. Keenan, C.J. Greenbaum, G.L. King, J.W. Heinecke, K.E.
and antioxidant properties in vivo and in vitro, Arterioscler. Thromb. Vasc. Biol. Bornfeldt, High concentration of medium-sized HDL particles and enrichment in
30 (2) (2010) 246–252. HDL paraoxonase 1 associate with protection from vascular complications in peo-
[171] G.D. Wool, V.G. Cabana, J. Lukens, P.X. Shaw, C.J. Binder, J.L. Witztum, C.A. Reardon, ple with long-standing type 1 diabetes, Diabetes Care 43 (1) (2020) 178–186.
G.S. Getz, 4F peptide reduces nascent atherosclerosis and induces natural antibody [193] Y. Shokri, A. Variji, M. Nosrati, A. Khonakdar-Tarsi, A. Kianmehr, Z. Kashi, A. Bahar,
production in apolipoprotein E-null mice, FASEB J. 25 (1) (2011) 290–300. A. Bagheri, A. Mahrooz, Importance of paraoxonase 1 (PON1) as an antioxidant and
[172] R.L. Dunbar, R. Movva, L.T. Bloedon, D. Duffy, R.B. Norris, M. Navab, A.M. Fogelman, antiatherogenic enzyme in the cardiovascular complications of type 2 diabetes: ge-
D.J. Rader, Oral Apolipoprotein A-I mimetic D-4F lowers HDL-inflammatory index notypic and phenotypic evaluation, Diabetes Res. Clin. Pract. 161 (2020) 108067.
in high-risk patients: a first-in-human multiple-dose, randomized controlled [194] A. Hoang, A.J. Murphy, M.T. Coughlan, M.C. Thomas, J.M. Forbes, R. O’Brien, M.E.
trial, Clin. Trans. Sci. 10 (6) (2017) 455–469. Cooper, J.P. Chin-Dusting, D. Sviridov, Advanced glycation of apolipoprotein A-I im-
[173] J.C. Tardif, J. Gregoire, P.L. L’Allier, R. Ibrahim, J. Lesperance, T.M. Heinonen, S. Kouz, pairs its anti-atherogenic properties, Diabetologia. 50 (8) (2007) 1770–1779.
C. Berry, R. Basser, M.A. Lavoie, M.C. Guertin, J. Rodes-Cabau, Effect of r HDLoA-S, [195] A. Machado-Lima, R.T. Iborra, R.S. Pinto, G. Castilho, C.H. Sartori, E.R. Oliveira, L.S.
efficacy I. effects of reconstituted high-density lipoprotein infusions on coronary Okuda, E.R. Nakandakare, D. Giannella-Neto, U.F. Machado, M.L. Correa-Giannella,
atherosclerosis: a randomized controlled trial, JAMA. 297 (15) (2007) 1675–1682. P. Traldi, S. Porcu, M. Roverso, A. Lapolla, M. Passarelli, In type 2 diabetes mellitus
[174] S. Diditchenko, A. Gille, I. Pragst, D. Stadler, M. Waelchli, R. Hamilton, A. Leis, S.D. glycated albumin alters macrophage gene expression impairing ABCA1-mediated
Wright, Novel formulation of a reconstituted high-density lipoprotein (CSL112) cholesterol efflux, J. Cell. Physiol. 230 (6) (2015) 1250–1257.
dramatically enhances ABCA1-dependent cholesterol efflux, Arterioscler. Thromb. [196] J.G. Tsun, S. Yung, M.K. Chau, S.W. Shiu, T.M. Chan, K.C. Tan, Cellular cholesterol
Vasc. Biol. 33 (9) (2013) 2202–2211. transport proteins in diabetic nephropathy, PLoS One 9 (9) (2014), e105787.
[175] C. Michael Gibson, S. Korjian, P. Tricoci, Y. Daaboul, M. Yee, P. Jain, J.H. Alexander, [197] W.S. Davidson, A.S. Shah, High-density lipoprotein subspecies in health and human
P.G. Steg, A.M. Lincoff, J.J. Kastelein, R. Mehran, D.M. D’Andrea, L.I. Deckelbaum, B. disease: focus on type 2 diabetes, Methodist. Debakey Cardiovasc. J. 15 (1) (2019)
Merkely, M. Zarebinski, Ophuis TO, R.A. Harrington, Safety and tolerability of 55–61.
CSL112, a reconstituted, infusible, plasma-derived apolipoprotein A-I, after acute [198] B.G. Drew, K.A. Rye, S.J. Duffy, P. Barter, B.A. Kingwell, The emerging role of HDL in
myocardial infarction: the AEGIS-I trial (ApoA-I event reducing in ischemic syn- glucose metabolism, Nat. Rev. Endocrinol. 8 (4) (2012) 237–245.
dromes I), Circulation. 134 (24) (2016) 1918–1930. [199] B.J. Cochran, R.J. Bisoendial, L. Hou, E.N. Glaros, J. Rossy, S.R. Thomas, P.J. Barter, K.A.
[176] Clinical Trials, Study to Investigate CSL112 in Subjects with Acute Coronary Syn- Rye, Apolipoprotein A-I increases insulin secretion and production from pancreatic
drome (AEGIS-II) (NCT03473223), 2019, November 13. beta-cells via a G-protein-cAMP-PKA-FoxO1-dependent mechanism, Arterioscler.
[177] C. Tardy, M. Goffinet, N. Boubekeur, R. Ackermann, G. Sy, A. Bluteau, G. Cholez, C. Thromb. Vasc. Biol. 34 (10) (2014) 2261–2267.
Keyserling, N. Lalwani, J.F. Paolini, J.L. Dasseux, R. Barbaras, R. Baron, CER-001, a [200] R. Han, R. Lai, Q. Ding, Z. Wang, X. Luo, Y. Zhang, G. Cui, J. He, W. Liu, Y. Chen, Apo-
HDL-mimetic, stimulates the reverse lipid transport and atherosclerosis regression lipoprotein A-I stimulates AMP-activated protein kinase and improves glucose me-
in high cholesterol diet-fed LDL-receptor deficient mice, Atherosclerosis. 232 (1) tabolism, Diabetologia. 50 (9) (2007) 1960–1968.
(2014) 110–118. [201] X. Ruan, Z. Li, Y. Zhang, L. Yang, Y. Pan, Z. Wang, G.S. Feng, Y. Chen, Apolipoprotein
[178] S.J. Nicholls, J. Andrews, Kastelein JJP, B. Merkely, S.E. Nissen, K.K. Ray, G.G. A-I possesses an anti-obesity effect associated with increase of energy expenditure
Schwartz, S.G. Worthley, C. Keyserling, J.L. Dasseux, L. Griffith, S.W. Kim, A. and up-regulation of UCP1 in brown fat, J. Cell. Mol. Med. 15 (4) (2011) 763–772.
Janssan, G. Di Giovanni, A.D. Pisaniello, D.J. Scherer, P.J. Psaltis, J. Butters, Effect of [202] P.J. Barter, K.A. Rye, J.C. Tardif, D.D. Waters, S.M. Boekholdt, A. Breazna, J.J. Kastelein,
serial infusions of CER-001, a pre-beta high-density lipoprotein mimetic, on coro- Effect of torcetrapib on glucose, insulin, and hemoglobin A1c in subjects in the in-
nary atherosclerosis in patients following acute coronary syndromes in the CER- vestigation of lipid level management to understand its impact in atherosclerotic
001 atherosclerosis regression acute coronary syndrome trial: a randomized clin- events (ILLUMINATE) trial, Circulation. 124 (5) (2011) 555–562.
ical trial, JAMA Cardiol. 3 (9) (2018) 815–822. [203] G.G. Schwartz, L.A. Leiter, C.M. Ballantyne, P.J. Barter, D.M. Black, D. Kallend, F.
[179] S.E. Nissen, T. Tsunoda, E.M. Tuzcu, P. Schoenhagen, C.J. Cooper, M. Yasin, G.M. Laghrissi-Thode, E. Leitersdorf, J.J.V. McMurray, S.J. Nicholls, A.G. Olsson, D. Preiss,
Eaton, M.A. Lauer, W.S. Sheldon, C.L. Grines, S. Halpern, T. Crowe, J.C. P.K. Shah, J.C. Tardif, J. Kittelson, Dalcetrapib reduces risk of new-onset diabetes
Blankenship, R. Kerensky, Effect of recombinant ApoA-I Milano on coronary ath- in patients with coronary heart disease, Diabetes Care 43 (5) (2020) 1077–1084.
erosclerosis in patients with acute coronary syndromes: a randomized controlled [204] S. Tang, F. Tabet, B.J. Cochran, L.F. Cuesta Torres, B.J. Wu, P.J. Barter, K.A. Rye, Apo-
trial, Jama. 290 (17) (2003) 2292–2300. lipoprotein A-I enhances insulin-dependent and insulin-independent glucose up-
[180] B. Ibanez, G. Vilahur, G. Cimmino, W.S. Speidl, A. Pinero, B.G. Choi, M.U. Zafar, C.G. take by skeletal muscle, Sci. Rep. 9 (1) (2019) 1350.
Santos-Gallego, B. Krause, L. Badimon, V. Fuster, J.J. Badimon, Rapid change in [205] M. Neuenschwander, G. Hoffmann, L. Schwingshackl, S. Schlesinger, Impact of dif-
plaque size, composition, and molecular footprint after recombinant apolipopro- ferent dietary approaches on blood lipid control in patients with type 2 diabetes
tein A-I Milano (ETC-216) administration: magnetic resonance imaging study in mellitus: a systematic review and network meta-analysis, Eur. J. Epidemiol. 34
an experimental model of atherosclerosis, J. Am. Coll. Cardiol. 51 (11) (2008) (9) (2019) 837–852.
1104–1109. [206] Group AS, Group AES, E.Y. Chew, W.T. Ambrosius, M.D. Davis, R.P. Danis, S.
[181] C. Giannarelli, G. Cimmino, B. Ibanez, G. Chiesa, J. Garcia-Prieto, C.G. Santos-Gallego, Gangaputra, C.M. Greven, L. Hubbard, B.A. Esser, J.F. Lovato, L.H. Perdue, D.C. Goff
M. Alique-Aguilar, V. Fuster, C. Sirtori, J.J. Badimon, Acute ApoA-I Milano adminis- Jr., W.C. Cushman, H.N. Ginsberg, M.B. Elam, S. Genuth, H.C. Gerstein, U.
tration induces plaque regression and stabilisation in the long term, Thromb. Schubart, L.J. Fine, Effects of medical therapies on retinopathy progression in
Haemost. 108 (6) (2012) 1246–1248. type 2 diabetes, N. Engl. J. Med. 363 (3) (2010) 233–244.
[182] J.A.A. Reijers, D.G. Kallend, K.E. Malone, J.W. Jukema, P.L.J. Wijngaard, J. Burggraaf, [207] L. Zhu, A. Hayen, K.J.L. Bell, Legacy effect of fibrate add-on therapy in diabetic pa-
M. Moerland, MDCO-216 does not induce adverse immunostimulation, in contrast tients with dyslipidemia: a secondary analysis of the ACCORDION study,
to its predecessor ETC-216, Cardiovasc. Drugs Ther. 31 (4) (2017) 381–389. Cardiovasc. Diabetol. 19 (1) (2020) 28.
[183] S.J. Nicholls, R. Puri, C.M. Ballantyne, J.W. Jukema, J.J.P. Kastelein, W. Koenig, R.S. [208] J. Lee, Y.C. Hwang, W.J. Lee, J.C. Won, K.H. Song, C.Y. Park, K.J. Ahn, J.Y. Park, Com-
Wright, D. Kallend, P. Wijngaard, M. Borgman, K. Wolski, S.E. Nissen, Effect of infu- parison of the efficacy and safety of rosuvastatin/ezetimibe combination therapy
sion of high-density lipoprotein mimetic containing recombinant apolipoprotein and rosuvastatin monotherapy on lipoprotein in patients with type 2 diabetes:
A-I milano on coronary disease in patients with an acute coronary syndrome in multicenter randomized controlled study, Diabetes Ther. 11 (4) (2020) 859–871.
the MILANO-PILOT trial: a randomized clinical trial, JAMA Cardiol. 3 (9) (2018) [209] M. Nieuwdorp, M. Vergeer, R.J. Bisoendial, J. Op’t Roodt, H. Levels, R.S. Birjmohun,
806–814. J.A. Kuivenhoven, R. Basser, T.J. Rabelink, J.J. Kastelein, E.S. Stroes, Reconstituted
[184] A. Sapra, P. Bhandari, Diabetes Mellitus, StatPearls, Treasure Island (FL), 2020. HDL infusion restores endothelial function in patients with type 2 diabetes
[185] D. Farbstein, A.P. Levy, HDL dysfunction in diabetes: causes and possible treat- mellitus, Diabetologia. 51 (6) (2008) 1081–1084.
ments, Expert. Rev. Cardiovasc. Ther. 10 (3) (2012) 353–361. [210] B.G. Drew, S.J. Duffy, M.F. Formosa, A.K. Natoli, D.C. Henstridge, S.A. Penfold, W.G.
[186] Y. Chen, Y. Zhi, C. Li, Y. Liu, L. Zhang, Y. Wang, K. Che, HDL cholesterol and risk of Thomas, N. Mukhamedova, B. de Courten, J.M. Forbes, F.Y. Yap, D.M. Kaye, G. van
diabetic nephropathy in patient with type 1 diabetes: a meta-analysis of cohort Hall, M.A. Febbraio, B.E. Kemp, D. Sviridov, G.R. Steinberg, B.A. Kingwell, High-
studies, Diabetes Res. Clin. Pract. 122 (2016) 84–91. density lipoprotein modulates glucose metabolism in patients with type 2 diabetes
[187] G. Fanni, R. Rosato, L. Gentile, M. Anselmino, S. Frea, V. Ponzo, M. Pellegrini, F. mellitus, Circulation. 119 (15) (2009) 2103–2111.
Broglio, F. Pivari, G.M. De Ferrari, E. Ghigo, S. Bo, Is HDL cholesterol protective in [211] M. Motamed, A.A. Nargesi, B. Heidari, H. Mirmiranpour, A. Esteghamati, M.
patients with type 2 diabetes? A retrospective population-based cohort study, J. Nakhjavani, Oxidized low-density lipoprotein (ox-LDL) to LDL ratio (ox-LDL/
Transl. Med. 18 (1) (2020) 189. LDL) and ox-LDL to high-density lipoprotein ratio (ox-LDL/HDL), Clin. Lab. 62
[188] B. Shao, M.N. Oda, J.F. Oram, J.W. Heinecke, Myeloperoxidase: an oxidative path- (9) (2016) 1609–1617.
way for generating dysfunctional high-density lipoprotein, Chem. Res. Toxicol. 23 [212] J. Girona, J.M. Manzanares, F. Marimon, A. Cabre, M. Heras, M. Guardiola, J. Ribalta,
(3) (2010) 447–454. L. Masana, Oxidized to non-oxidized lipoprotein ratios are associated with

114
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

arteriosclerosis and the metabolic syndrome in diabetic patients, NMCD. 18 (5) [238] M. Gomaraschi, Role of lipoproteins in the microenvironment of hormone-
(2008) 380–387. dependent cancers, Trends Endocrinol. Metab. 31 (3) (2020) 256–268.
[213] N. Toshikuni, M. Tsutsumi, T. Arisawa, Clinical differences between alcoholic liver [239] S. Raut, L. Mooberry, N. Sabnis, A. Garud, A.S. Dossou, A. Lacko, Reconstituted HDL:
disease and nonalcoholic fatty liver disease, World J. Gastroenterol. 20 (26) drug delivery platform for overcoming biological barriers to cancer therapy, Front.
(2014) 8393–8406. Pharmacol. 9 (2018)1154-.
[214] E.A. Brinton, Effects of ethanol intake on lipoproteins, Curr. Atheroscler. Rep. 14 (2) [240] M.M. Shahzad, L.S. Mangala, H.D. Han, C. Lu, J. Bottsford-Miller, M. Nishimura, E.M.
(2012) 108–114. Mora, J.W. Lee, R.L. Stone, C.V. Pecot, D. Thanapprapasr, J.W. Roh, P. Gaur, M.P. Nair,
[215] P.K. Rao, K. Merath, E. Drigalenko, A.Y.L. Jadhav, R.A. Komorowski, M.I. Goldblatt, A. Y.Y. Park, N. Sabnis, M.T. Deavers, J.S. Lee, L.M. Ellis, G. Lopez-Berestein, W.J.
Rohatgi, M.A. Sarzynski, S. Gawrieh, M. Olivier, Proteomic characterization of high- McConathy, L. Prokai, A.G. Lacko, A.K. Sood, Targeted delivery of small interfering
density lipoprotein particles in patients with non-alcoholic fatty liver disease, Clin. RNA using reconstituted high-density lipoprotein nanoparticles, Neoplasia. 13
Proteomics 15 (2018) 10. (4) (2011) 309–319.
[216] F. Bril, J.J. Sninsky, A.M. Baca, H.R. Superko, P. Portillo Sanchez, D. Biernacki, M. [241] Moya M. de la Llera, F.C. McGillicuddy, C.C. Hinkle, M. Byrne, M.R. Joshi, V. Nguyen,
Maximos, R. Lomonaco, B. Orsak, A. Suman, M.H. Weber, M.J. McPhaul, K. Cusi, He- J. Tabita-Martinez, M.L. Wolfe, K. Badellino, L. Pruscino, N.N. Mehta, B.F. Asztalos,
patic steatosis and insulin resistance, but not Steatohepatitis, promote atherogenic M.P. Reilly, Inflammation modulates human HDL composition and function
dyslipidemia in NAFLD, J. Clin. Endocrinol. Metab. 101 (2) (2016) 644–652. in vivo, Atherosclerosis. 222 (2) (2012) 390–394.
[217] R. Fadaei, H. Poustchi, R. Meshkani, N. Moradi, T. Golmohammadi, S. Merat, Im- [242] A. Pirillo, A.L. Catapano, G.D. Norata, HDL in infectious diseases and sepsis, Handb.
paired HDL cholesterol efflux capacity in patients with non-alcoholic fatty liver Exp. Pharmacol. 224 (2015) 483–508.
disease is associated with subclinical atherosclerosis, Sci. Rep. 8 (1) (2018) 11691. [243] Y.L. Jiao, M.P. Wu, Apolipoprotein A-I diminishes acute lung injury and sepsis in
[218] P. Loria, G. Marchesini, F. Nascimbeni, S. Ballestri, M. Maurantonio, F. Carubbi, V. mice induced by lipoteichoic acid, Cytokine. 43 (1) (2008) 83–87.
Ratziu, A. Lonardo, Cardiovascular risk, lipidemic phenotype and steatosis. A com- [244] T. Speer, S. Zewinger, High-density lipoprotein (HDL) and infections: a versatile
parative analysis of cirrhotic and non-cirrhotic liver disease due to varying etiol- culprit, Eur. Heart J. 39 (14) (2018) 1191–1193.
ogy, Atherosclerosis. 232 (1) (2014) 99–109. [245] C.M. Madsen, A. Varbo, A. Tybjaerg-Hansen, R. Frikke-Schmidt, B.G. Nordestgaard,
[219] M. Trieb, A. Horvath, R. Birner-Gruenberger, W. Spindelboeck, V. Stadlbauer, U. U-shaped relationship of HDL and risk of infectious disease: two prospective
Taschler, S. Curcic, R.E. Stauber, M. Holzer, L. Pasterk, A. Heinemann, G. Marsche, population-based cohort studies, Eur. Heart J. 39 (14) (2018) 1181–1190.
Liver disease alters high-density lipoprotein composition, metabolism and func- [246] D. Perez-Morga, B. Vanhollebeke, F. Paturiaux-Hanocq, D.P. Nolan, L. Lins, F.
tion, Biochim. Biophys. Acta 1861 (7) (2016) 630–638. Homble, L. Vanhamme, P. Tebabi, A. Pays, P. Poelvoorde, A. Jacquet, R. Brasseur,
[220] B. Gao, J. Xiao, M. Zhang, F. Zhang, W. Zhang, J. Yang, J. He, Y. Liu, X. Zou, P. Xu, Y. E. Pays, Apolipoprotein L-I promotes trypanosome lysis by forming pores in lyso-
Zhuge, High-density lipoprotein cholesterol for the prediction of mortality in cir- somal membranes, Science (New York, N.Y.) 309 (5733) (2005) 469–472.
rhosis with portal vein thrombosis: a retrospective study, Lipids Health Dis. 18 [247] M. Samanovic, M.P. Molina-Portela, A.D. Chessler, B.A. Burleigh, J. Raper, Trypano-
(1) (2019) 79. some lytic factor, an antimicrobial high-density lipoprotein, ameliorates Leish-
[221] A. Sorokin, J.L. Brown, P.D. Thompson, Primary biliary cirrhosis, hyperlipidemia, mania infection, PLoS Pathog. 5 (1) (2009), e1000276.
and atherosclerotic risk: a systematic review, Atherosclerosis. 194 (2) (2007)
[248] D. Marin-Palma, G.A. Castro, J.A. Cardona-Arias, S. Urcuqui-Inchima, J.C. Hernandez,
293–299.
Lower high-density lipoproteins levels during human immunodeficiency virus
[222] A. Arvind, S.A. Osganian, D.E. Cohen, K.E. Corey, in: K.R. Feingold, B. Anawalt, A. type 1 infection are associated with increased inflammatory markers and disease
Boyce, G. Chrousos, K. Dungan, A. Grossman (Eds.), Lipid and Lipoprotein Metabo- progression, Front. Immunol. 9 (2018) 1350.
lism in Liver Disease, Endotext, South Dartmouth (MA), 2000.
[249] J.V. Baker, J. Neuhaus, D. Duprez, D.A. Cooper, J. Hoy, L. Kuller, F.C. Lampe, A. Liappis,
[223] M.L. Kashyap, S. Ganji, N.K. Nakra, V.S. Kamanna, Niacin for treatment of nonalco-
N. Friis-Moller, J. Otvos, N.I. Paton, R. Tracy, J.D. Neaton, Group ISS, Inflammation
holic fatty liver disease (NAFLD): novel use for an old drug? J. Clin. Lipidol. 13 (6)
predicts changes in high-density lipoprotein particles and apolipoprotein A1 fol-
(2019) 873–879.
lowing initiation of antiretroviral therapy, Aids. 25 (17) (2011) 2133–2142.
[224] S.H. Ganji, M.L. Kashyap, V.S. Kamanna, Niacin inhibits fat accumulation, oxidative
[250] T. Kelesidis, O.O. Yang, M.A. Kendall, H.N. Hodis, J.S. Currier, Dysfunctional HDL and
stress, and inflammatory cytokine IL-8 in cultured hepatocytes: impact on non-
progression of atherosclerosis in HIV-1-infected and -uninfected adults, Lipids
alcoholic fatty liver disease, Metab. Clin. Exp. 64 (9) (2015) 982–990.
Health Dis. 12 (2013) 23.
[225] S.H. Ganji, G.D. Kukes, N. Lambrecht, M.L. Kashyap, V.S. Kamanna, Therapeutic role
[251] S.A. Pereira, J.R. Batuca, U. Caixas, T. Branco, J. Delgado-Alves, I. Germano, F.
of niacin in the prevention and regression of hepatic steatosis in rat model of non-
Lampreia, E.C. Monteiro, Effect of efavirenz on high-density lipoprotein antioxi-
alcoholic fatty liver disease, Am. J. Physiol. Gastrointest. Liver Physiol. 306 (4)
dant properties in HIV-infected patients, Br. J. Clin. Pharmacol. 68 (6) (2009)
(2014) G320–G327.
891–897.
[226] A. Mantovani, Cancer: inflammation by remote control, Nature. 435 (7043) (2005)
[252] H. Gupta, L. Dai, G. Datta, D.W. Garber, H. Grenett, Y. Li, V. Mishra, M.N.
752–753.
Palgunachari, S. Handattu, S.H. Gianturco, W.A. Bradley, G.M. Anantharamaiah,
[227] L.M. Coussens, Z. Werb, Inflammation and cancer, Nature. 420 (6917) (2002)
C.R. White, Inhibition of lipopolysaccharide-induced inflammatory responses by
860–867.
an apolipoprotein AI mimetic peptide, Circ. Res. 97 (3) (2005) 236–243.
[228] Y. Jiang, A. Sun, Y. Zhao, W. Ying, H. Sun, X. Yang, B. Xing, W. Sun, L. Ren, B. Hu, C. Li,
[253] W.Y. Kwon, G.J. Suh, K.S. Kim, Y.H. Kwak, K. Kim, 4F, apolipoprotein AI mimetic
L. Zhang, G. Qin, M. Zhang, N. Chen, M. Zhang, Y. Huang, J. Zhou, Y. Zhao, M. Liu, X.
peptide, attenuates acute lung injury and improves survival in endotoxemic rats,
Zhu, Y. Qiu, Y. Sun, C. Huang, M. Yan, M. Wang, W. Liu, F. Tian, H. Xu, J. Zhou, Z. Wu,
J. Trauma Acute Care Surgery. 72 (6) (2012) 1576–1583.
T. Shi, W. Zhu, J. Qin, L. Xie, J. Fan, X. Qian, F. He, Chinese Human Proteome Project
C, Proteomics identifies new therapeutic targets of early-stage hepatocellular car- [254] T.N. Baral, S. Magez, B. Stijlemans, K. Conrath, B. Vanhollebeke, E. Pays, S.
cinoma, Nature. 567 (7747) (2019) 257–261. Muyldermans, P. De Baetselier, Experimental therapy of African trypanosomiasis
[229] K. Georgila, D. Vyrla, E. Drakos, Apolipoprotein A-I (ApoA-I), immunity, inflamma- with a nanobody-conjugated human trypanolytic factor, Nat. Med. 12 (5) (2006)
tion and cancer, Cancers. 11 (8) (2019). 580–584.
[230] F. Shi, H. Wu, K. Qu, Q. Sun, F. Li, C. Shi, Y. Li, X. Xiong, Q. Qin, T. Yu, X. Jin, L. Cheng, [255] J. Han, Y. Jiang, Z. Li, V.V. Kravchenko, R.J. Ulevitch, Activation of the transcription
Q. Wei, Y. Li, J. She, Identification of serum proteins AHSG, FGA and APOA-I as di- factor MEF2C by the MAP kinase p38 in inflammation, Nature. 386 (6622)
agnostic biomarkers for gastric cancer, Clin. Proteomics 15 (2018) 18. (1997) 296–299.
[231] J. Naru, R. Aggarwal, A.K. Mohanty, U. Singh, D. Bansal, N. Kakkar, N. Agnihotri, [256] M.J. Sweet, D.A. Hume, Endotoxin signal transduction in macrophages, J. Leukoc.
Identification of differentially expressed proteins in retinoblastoma tumors using Biol. 60 (1) (1996) 8–26.
mass spectrometry-based comparative proteomic approach, J. Proteome 159 [257] M.M. Wurfel, S.T. Kunitake, H. Lichenstein, J.P. Kane, S.D. Wright, Lipopolysaccha-
(2017) 77–91. ride (LPS)-binding protein is carried on lipoproteins and acts as a cofactor in the
[232] P.M. Cruz, H. Mo, W.J. McConathy, N. Sabnis, A.G. Lacko, The role of cholesterol me- neutralization of LPS, J. Exp. Med. 180 (3) (1994) 1025–1035.
tabolism and cholesterol transport in carcinogenesis: a review of scientific find- [258] M.A. Freudenberg, T.C. Bog-Hansen, U. Back, C. Galanos, Interaction of lipopolysac-
ings, relevant to future cancer therapeutics, Front. Pharmacol. 4 (2013) 119. charides with plasma high-density lipoprotein in rats, Infect. Immun. 28 (2)
[233] M. Ruscica, M. Botta, N. Ferri, E. Giorgio, C. Macchi, G. Franceschini, P. Magni, L. (1980) 373–380.
Calabresi, M. Gomaraschi, High density lipoproteins inhibit oxidative stress- [259] C.J. de Haas, H.J. van Leeuwen, J. Verhoef, K.P. van Kessel, J.A. van Strijp, Analysis of
induced prostate cancer cell proliferation, Sci. Rep. 8 (1) (2018) 2236. lipopolysaccharide (LPS)-binding characteristics of serum components using gel
[234] M. Zamanian-Daryoush, D. Lindner, T.C. Tallant, Z. Wang, J. Buffa, E. Klipfell, Y. filtration of FITC-labeled LPS, J. Immunol. Methods 242 (1–2) (2000) 79–89.
Parker, D. Hatala, P. Parsons-Wingerter, P. Rayman, M.S. Yusufishaq, E.A. Fisher, [260] S. Tanaka, D. Couret, A. Tran-Dinh, J. Duranteau, P. Montravers, A. Schwendeman,
J.D. Smith, J. Finke, J.A. DiDonato, S.L. Hazen, The cardioprotective protein apolipo- O. Meilhac, High-density lipoproteins during sepsis: from bench to bedside, Critical
protein A1 promotes potent anti-tumorigenic effects, J. Biol. Chem. 288 (29) Care (London, England). 24 (1) (2020) 134.
(2013) 21237–21252. [261] P.M. Cazita, D.F. Barbeiro, A.I. Moretti, E.C. Quintao, F.G. Soriano, Human cholesteryl
[235] F. Su, K.R. Kozak, S. Imaizumi, F. Gao, M.W. Amneus, V. Grijalva, C. Ng, A. Wagner, G. ester transfer protein expression enhances the mouse survival rate in an experi-
Hough, G. Farias-Eisner, G.M. Anantharamaiah, B.J. Van Lenten, M. Navab, A.M. mental systemic inflammation model: a novel role for CETP, Shock (Augusta,
Fogelman, S.T. Reddy, R. Farias-Eisner, Apolipoprotein A-I (apoA-I) and apoA-I mi- Ga). 30 (5) (2008) 590–595.
metic peptides inhibit tumor development in a mouse model of ovarian cancer, [262] I. Hardardottir, A.H. Moser, J. Fuller, C. Fielding, K. Feingold, C. Grunfeld, Endotoxin
Proc. Natl. Acad. Sci. U. S. A. 107 (46) (2010) 19997–20002. and cytokines decrease serum levels and extra hepatic protein and mRNA levels of
[236] S.E. Henrich, C.S. Thaxton, An update on synthetic high-density lipoprotein-like cholesteryl ester transfer protein in Syrian hamsters, J. Clin. Invest. 97 (11) (1996)
nanoparticles for cancer therapy, Expert. Rev. Anticancer. Ther. 19 (6) (2019) 2585–2592.
515–528. [263] J.H. Levels, D. Pajkrt, M. Schultz, F.J. Hoek, A. van Tol, J.C. Meijers, S.J. van Deventer,
[237] S. Sarhadi, S. Ganjali, M. Pirro, A. Sahebkar, The role of high-density lipoproteins in Alterations in lipoprotein homeostasis during human experimental endotoxemia
antitumor drug delivery, IUBMB Life 71 (10) (2019) 1442–1452. and clinical sepsis, Biochim. Biophys. Acta 1771 (12) (2007) 1429–1438.

115
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

[264] M. Trinder, K.R. Genga, H.J. Kong, L.L. Blauw, C. Lo, X. Li, M. Cirstea, Y. Wang, P.C.N. [289] H.K. Choi, J.D. Seeger, Lipid profiles among US elderly with untreated rheumatoid
Rensen, J.A. Russell, K.R. Walley, J.H. Boyd, L.R. Brunham, Cholesteryl Ester transfer arthritis–the third national health and nutrition examination survey, J. Rheumatol.
protein influences high-density lipoprotein levels and survival in sepsis, Am. J. 32 (12) (2005) 2311–2316.
Respir. Crit. Care Med. 199 (7) (2019) 854–862. [290] A.N. Georgiadis, E.C. Papavasiliou, E.S. Lourida, Y. Alamanos, C. Kostara, A.D.
[265] H.J. van Leeuwen, E.C. Heezius, G.M. Dallinga, J.A. van Strijp, J. Verhoef, K.P. van Tselepis, A.A. Drosos, Atherogenic lipid profile is a feature characteristic of patients
Kessel, Lipoprotein metabolism in patients with severe sepsis, Crit. Care Med. 31 with early rheumatoid arthritis: effect of early treatment–a prospective, controlled
(5) (2003) 1359–1366. study, Arthritis Res. Therapy. 8 (3) (2006) R82.
[266] S. Tanaka, J. Labreuche, E. Drumez, A. Harrois, S. Hamada, B. Vigue, D. Couret, J. [291] M.B. Lazarevic, J. Vitic, V. Mladenovic, B.L. Myones, J.L. Skosey, W.I. Swedler,
Duranteau, O. Meilhac, Low HDL levels in sepsis versus trauma patients in inten- Dyslipoproteinemia in the course of active rheumatoid arthritis, Semin. Arthritis
sive care unit, Ann. Intensive Care 7 (1) (2017) 60. Rheum. 22 (3) (1992) 172–178.
[267] S. Barlage, C. Gnewuch, G. Liebisch, Z. Wolf, F.X. Audebert, T. Gluck, D. Frohlich, B.K. [292] C.P. Chung, A. Oeser, P. Raggi, T. Sokka, T. Pincus, J.F. Solus, M.F. Linton, S. Fazio, C.M.
Kramer, G. Rothe, G. Schmitz, Changes in HDL-associated apolipoproteins relate to Stein, Lipoprotein subclasses determined by nuclear magnetic resonance spectros-
mortality in human sepsis and correlate to monocyte and platelet activation, Inten- copy and coronary atherosclerosis in patients with rheumatoid arthritis, J.
sive Care Med. 35 (11) (2009) 1877–1885. Rheumatol. 37 (8) (2010) 1633–1638.
[268] J.Y. Chien, J.S. Jerng, C.J. Yu, P.C. Yang, Low serum level of high-density lipoprotein [293] F. Montecucco, E. Favari, G.D. Norata, N. Ronda, J.R. Nofer, N. Vuilleumier, Impact of
cholesterol is a poor prognostic factor for severe sepsis, Crit. Care Med. 33 (8) systemic inflammation and autoimmune diseases on apoA-I and HDL plasma
(2005) 1688–1693. levels and functions, Handb. Exp. Pharmacol. 224 (2015) 455–482.
[269] M.S. Winkler, K.B. Martz, A. Nierhaus, G. Daum, E. Schwedhelm, S. Kluge, M.H. [294] Y. Wang, S. Lu, G. Zhang, S. Wu, Y. Yan, Q. Dong, B. Liu, Anti-inflammatory effects of
Graler, Loss of sphingosine 1-phosphate (S1P) in septic shock is predominantly HDL in mice with rheumatoid arthritis induced by collagen, Front. Immunol. 9
caused by decreased levels of high-density lipoproteins (HDL), J. Intensive Care 7 (2018) 1013.
(2019) 23. [295] B.J. Wu, K.L. Ong, S. Shrestha, K. Chen, F. Tabet, P.J. Barter, K.A. Rye, Inhibition of ar-
[270] S.M. Coldewey, E. Benetti, M. Collino, J. Pfeilschifter, C. Sponholz, M. Bauer, A. thritis in the Lewis rat by apolipoprotein A-I and reconstituted high-density lipo-
Huwiler, C. Thiemermann, Elevation of serum sphingosine-1-phosphate attenu- proteins, Arterioscler. Thromb. Vasc. Biol. 34 (3) (2014) 543–551.
ates impaired cardiac function in experimental sepsis, Sci. Rep. 6 (2016) 27594. [296] C.M. Madsen, A. Varbo, B.G. Nordestgaard, Low HDL cholesterol and high risk of au-
[271] S. Tanaka, D. Diallo, S. Delbosc, C. Geneve, N. Zappella, J. Yong-Sang, J. Patche, A. toimmune disease: two population-based cohort studies including 117341 indi-
Harrois, S. Hamada, E. Denamur, P. Montravers, J. Duranteau, O. Meilhac, High- viduals, Clin. Chem. 65 (5) (2019) 644–652.
density lipoprotein (HDL) particle size and concentration changes in septic shock [297] J. Watanabe, C. Charles-Schoeman, Y. Miao, D. Elashoff, Y.Y. Lee, G. Katselis, T.D. Lee,
patients, Ann. Intensive Care 9 (1) (2019) 68. S.T. Reddy, Proteomic profiling following immunoaffinity capture of high-density
[272] F. Zimetti, S. De Vuono, M. Gomaraschi, M.P. Adorni, E. Favari, N. Ronda, M.A. Ricci, lipoprotein: association of acute-phase proteins and complement factors with pro-
F. Veglia, L. Calabresi, G. Lupattelli, Plasma cholesterol homeostasis, HDL remodel- inflammatory high-density lipoprotein in rheumatoid arthritis, Arthritis Rheum. 64
ing and function during the acute phase reaction, J. Lipid Res. 58 (10) (2017) (6) (2012) 1828–1837.
2051–2060. [298] B.H. Hahn, J. Grossman, W. Chen, M. McMahon, The pathogenesis of atherosclerosis
[273] F.W. Guirgis, C. Leeuwenburgh, V. Grijalva, J. Bowman, C. Kalynych, L. Moldawer, in autoimmune rheumatic diseases: roles of inflammation and dyslipidemia, J.
F.A. Moore, S.T. Reddy, HDL cholesterol efflux is impaired in older patients with Autoimmun. 28 (2–3) (2007) 69–75.
early sepsis: a subanalysis of a prospective pilot study, Shock (Augusta, Ga). 50 [299] N. Ronda, E. Favari, M.O. Borghi, F. Ingegnoli, M. Gerosa, C. Chighizola, F. Zimetti,
(1) (2018) 66–70. M.P. Adorni, F. Bernini, P.L. Meroni, Impaired serum cholesterol efflux capacity in
rheumatoid arthritis and systemic lupus erythematosus, Ann. Rheum. Dis. 73 (3)
[274] L. Guo, J. Ai, Z. Zheng, D.A. Howatt, A. Daugherty, B. Huang, X.A. Li, High density li-
(2014) 609–615.
poprotein protects against polymicrobe-induced sepsis in mice, J. Biol. Chem. 288
(25) (2013) 17947–17953. [300] F. Carbone, A. Nencioni, F. Mach, N. Vuilleumier, F. Montecucco, Evidence on the
pathogenic role of auto-antibodies in acute cardiovascular diseases, Thromb.
[275] D.M. Levine, T.S. Parker, T.M. Donnelly, A. Walsh, A.L. Rubin, In vivo protection
Haemost. 109 (5) (2013) 854–868.
against endotoxin by plasma high density lipoprotein, Proc. Natl. Acad. Sci. U. S.
[301] C. Qiu, X. Zhao, L. She, Z. Shi, Z. Deng, L. Tan, X. Tu, S. Jiang, B. Tang, Baricitinib in-
A. 90 (24) (1993) 12040–12044.
duces LDL-C and HDL-C increases in rheumatoid arthritis: a meta-analysis of ran-
[276] D. Pajkrt, J.E. Doran, F. Koster, P.G. Lerch, B. Arnet, T. van der Poll, J.W. ten Cate, S.J.
domized controlled trials, Lipids Health Dis. 18 (1) (2019) 54.
van Deventer, Antiinflammatory effects of reconstituted high-density lipoprotein
[302] C. Charles-Schoeman, Y. Yin Lee, A. Shahbazian, X. Wang, D. Elashoff, J.R. Curtis, I.
during human endotoxemia, J. Exp. Med. 184 (5) (1996) 1601–1608.
Navarro-Millan, S. Yang, L. Chen, S.S. Cofield, L.W. Moreland, H. Paulus, J. O’Dell, J.
[277] M. Trinder, K.R. Walley, J.H. Boyd, L.R. Brunham, Causal inference for genetically
Bathon, S. Louis Bridges Jr., S.T. Reddy, Improvement of high-density lipoprotein
determined levels of high-density lipoprotein cholesterol and risk of infectious dis-
function in patients with early rheumatoid arthritis treated with methotrexate
ease, Arterioscler. Thromb. Vasc. Biol. 40 (1) (2020) 267–278.
monotherapy or combination therapies in a randomized controlled trial, Arthritis
[278] Z. Zhang, G. Datta, Y. Zhang, A.P. Miller, P. Mochon, Y.F. Chen, J. Chatham, G.M.
Rheum. 69 (1) (2017) 46–57.
Anantharamaiah, C.R. White, Apolipoprotein A-I mimetic peptide treatment in-
[303] F. O’Neill, M. Charakida, E. Topham, E. McLoughlin, N. Patel, E. Sutill, C.W.M. Kay, F.
hibits inflammatory responses and improves survival in septic rats, Am. J. Physiol.
D’Aiuto, U. Landmesser, P.C. Taylor, J. Deanfield, Anti-inflammatory treatment im-
Heart Circ. Physiol. 297 (2) (2009) H866–H873.
proves high-density lipoprotein function in rheumatoid arthritis, Heart. 103 (10)
[279] R.S. Moreira, M. Irigoyen, T.R. Sanches, R.A. Volpini, N.O. Camara, D.M. Malheiros, (2017) 766–773.
M.H. Shimizu, A.C. Seguro, L. Andrade, Apolipoprotein A-I mimetic peptide 4F at- [304] C. Charles-Schoeman, G.B. Gugiu, H. Ge, A. Shahbazian, Y.Y. Lee, X. Wang, D.E. Furst,
tenuates kidney injury, heart injury, and endothelial dysfunction in sepsis, Am. J. V.K. Ranganath, M. Maldonado, T. Lee, S.T. Reddy, Remodeling of the HDL proteome
Phys. Regul. Integr. Comp. Phys. 307 (5) (2014) R514–R524. with treatment response to abatacept or adalimumab in the AMPLE trial of pa-
[280] A.T. Casas, A.P. Hubsch, J.E. Doran, Effects of reconstituted high-density lipoprotein tients with rheumatoid arthritis, Atherosclerosis. 275 (2018) 107–114.
in persistent gram-negative bacteremia, Am. Surg. 62 (5) (1996) 350–355. [305] Y. Shoenfeld, R. Gerli, A. Doria, E. Matsuura, M.M. Cerinic, N. Ronda, L.J. Jara, M.
[281] A.T. Casas, A.P. Hubsch, B.C. Rogers, J.E. Doran, Reconstituted high-density lipopro- Abu-Shakra, P.L. Meroni, Y. Sherer, Accelerated atherosclerosis in autoimmune
tein reduces LPS-stimulated TNF alpha, J. Surg. Res. 59 (5) (1995) 544–552. rheumatic diseases, Circulation. 112 (21) (2005) 3337–3347.
[282] M.C. McDonald, P. Dhadly, G.W. Cockerill, S. Cuzzocrea, H. Mota-Filipe, C.J. Hinds, [306] E.F. Borba, E. Bonfa, Dyslipoproteinemias in systemic lupus erythematosus: influ-
N.E. Miller, C. Thiemermann, Reconstituted high-density lipoprotein attenuates ence of disease, activity, and anticardiolipin antibodies, Lupus. 6 (6) (1997)
organ injury and adhesion molecule expression in a rodent model of endotoxic 533–539.
shock, Shock (Augusta, Ga). 20 (6) (2003) 551–557. [307] I.N. Bruce, M.B. Urowitz, D.D. Gladman, D. Ibanez, G. Steiner, Risk factors for coro-
[283] S. Tanaka, C. Geneve, N. Zappella, J. Yong-Sang, C. Planesse, L. Louedec, W. nary heart disease in women with systemic lupus erythematosus: the Toronto Risk
Viranaicken, M. Bringart, P. Montravers, E. Denamur, J. Duranteau, D. Couret, O. Factor Study, Arthritis Rheum. 48 (11) (2003) 3159–3167.
Meilhac, Reconstituted high-density lipoprotein therapy improves survival in [308] J.F. de Carvalho, E. Bonfa, E.F. Borba, Systemic lupus erythematosus and “lupus
mouse models of sepsis, Anesthesiology. 132 (4) (2020) 825–838. dyslipoproteinemia”, Autoimmun. Rev. 7 (3) (2008) 246–250.
[284] X. Zhang, L. Wang, B. Chen, Recombinant HDL (Milano) protects endotoxin- [309] S.Y. Kim, M. Yu, E.E. Morin, J. Kang, M.J. Kaplan, A. Schwendeman, High-density li-
challenged rats from multiple organ injury and dysfunction, Biol. Chem. 396 (1) poprotein in lupus: disease biomarkers and potential therapeutic strategy, Arthritis
(2015) 53–60. Rheum. 72 (1) (2019) 20–30.
[285] J.S. Smolen, D. Aletaha, I.B. McInnes, Rheumatoid arthritis, Lancet (London, En- [310] S. Manzi, E.N. Meilahn, J.E. Rairie, C.G. Conte, T.A. Medsger Jr., L. Jansen-
gland). 388 (10055) (2016) 2023–2038. McWilliams, R.B. D’Agostino, L.H. Kuller, Age-specific incidence rates of myocardial
[286] F.A. Pinheiro, D.C. Souza, E.I. Sato, A study of multiple causes of death in rheuma- infarction and angina in women with systemic lupus erythematosus: comparison
toid arthritis, J. Rheumatol. 42 (12) (2015) 2221–2228. with the Framingham study, Am. J. Epidemiol. 145 (5) (1997) 408–415.
[287] J.A. Avina-Zubieta, H.K. Choi, M. Sadatsafavi, M. Etminan, J.M. Esdaile, D. Lacaille, [311] M. McMahon, J. Grossman, J. FitzGerald, E. Dahlin-Lee, D.J. Wallace, B.Y. Thong, H.
Risk of cardiovascular mortality in patients with rheumatoid arthritis: a meta- Badsha, K. Kalunian, C. Charles, M. Navab, A.M. Fogelman, B.H. Hahn, Proinflamma-
analysis of observational studies, Arthritis Rheum. 59 (12) (2008) 1690–1697. tory high-density lipoprotein as a biomarker for atherosclerosis in patients with
[288] R. Agca, S.C. Heslinga, S. Rollefstad, M. Heslinga, McInnes IB, M.J. Peters, T.K. Kvien, systemic lupus erythematosus and rheumatoid arthritis, Arthritis Rheum. 54 (8)
M. Dougados, H. Radner, F. Atzeni, J. Primdahl, A. Sodergren, S. Wallberg Jonsson, J. (2006) 2541–2549.
van Rompay, C. Zabalan, T.R. Pedersen, L. Jacobsson, K. de Vlam, M.A. Gonzalez- [312] L.E. Schanberg, C. Sandborg, H.X. Barnhart, S.P. Ardoin, E. Yow, G.W. Evans, K.L.
Gay, A.G. Semb, G.D. Kitas, Y.M. Smulders, Z. Szekanecz, N. Sattar, D.P. Symmons, Mieszkalski, N.T. Ilowite, A. Eberhard, D.M. Levy, Y. Kimura, E. von Scheven, E.
M.T. Nurmohamed, EULAR recommendations for cardiovascular disease risk man- Silverman, S.L. Bowyer, L. Punaro, N.G. Singer, D.D. Sherry, D. McCurdy, M. Klein-
agement in patients with rheumatoid arthritis and other forms of inflammatory Gitelman, C. Wallace, R. Silver, L. Wagner-Weiner, G.C. Higgins, H.I. Brunner, L.
joint disorders: 2015/2016 update, Ann. Rheum. Dis. 76 (1) (2017) 17–28. Jung, J.B. Soep, A. Reed, Premature atherosclerosis in pediatric systemic lupus

116
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

erythematosus: risk factors for increased carotid intima-media thickness in the [335] M. Voutilainen, N. Hutri-Kahonen, P. Tossavainen, T. Sipponen, N. Pitkanen, T.
atherosclerosis prevention in pediatric lupus erythematosus cohort, Arthritis Laitinen, E. Jokinen, T. Ronnemaa, J.S.A. Viikari, O.T. Raitakari, M. Juonala, Low
Rheum. 60 (5) (2009) 1496–1507. childhood high density lipoprotein cholesterol levels and subsequent risk for
[313] M. McMahon, B.J. Skaggs, L. Sahakian, J. Grossman, J. FitzGerald, N. Ragavendra, C. chronic inflammatory bowel disease, Digest. Liver Dis. 50 (4) (2018) 348–352.
Charles-Schoeman, M. Chernishof, A. Gorn, J.L. Witztum, W.K. Wong, M. [336] S.I. van Leuven, R. Hezemans, J.H. Levels, S. Snoek, P.C. Stokkers, G.K. Hovingh, J.J.
Weisman, D.J. Wallace, A. La Cava, B.H. Hahn, High plasma leptin levels confer in- Kastelein, E.S. Stroes, E. de Groot, D.W. Hommes, Enhanced atherogenesis and al-
creased risk of atherosclerosis in women with systemic lupus erythematosus, tered high density lipoprotein in patients with Crohn’s disease, J. Lipid Res. 48
and are associated with inflammatory oxidised lipids, Ann. Rheum. Dis. 70 (9) (12) (2007) 2640–2646.
(2011) 1619–1624. [337] D. Meriwether, D. Sulaiman, A. Wagner, V. Grijalva, I. Kaji, K.J. Williams, L. Yu, S.
[314] B.H. Hahn, J. Grossman, B.J. Ansell, B.J. Skaggs, M. McMahon, Altered lipoprotein Fogelman, C. Volpe, S.J. Bensinger, G.M. Anantharamaiah, I. Shechter, A.M.
metabolism in chronic inflammatory states: proinflammatory high-density lipo- Fogelman, S.T. Reddy, Transintestinal transport of the anti-inflammatory drug 4F
protein and accelerated atherosclerosis in systemic lupus erythematosus and rheu- and the modulation of transintestinal cholesterol efflux, J. Lipid Res. 57 (7)
matoid arthritis, Arthritis Res. Therapy. 10 (4) (2008) 213. (2016) 1175–1193.
[315] S.G. O’Neill, I. Giles, A. Lambrianides, J. Manson, D. D’Cruz, L. Schrieber, L.M. March, [338] M. Navab, S.T. Reddy, G.M. Anantharamaiah, G. Hough, G.M. Buga, J. Danciger, A.M.
D.S. Latchman, D.A. Isenberg, A. Rahman, Antibodies to apolipoprotein A-I, high- Fogelman, D-4F-mediated reduction in metabolites of arachidonic and linoleic
density lipoprotein, and C-reactive protein are associated with disease activity in acids in the small intestine is associated with decreased inflammation in low-
patients with systemic lupus erythematosus, Arthritis Rheum. 62 (3) (2010) density lipoprotein receptor-null mice, J. Lipid Res. 53 (3) (2012) 437–445.
845–854. [339] D. Meriwether, D. Sulaiman, C. Volpe, A. Dorfman, V. Grijalva, N. Dorreh, R.S.
[316] O.L. Vera-Lastra, A. Olvera-Acevedo, C. Hernandez, G. Medina, A.L. Carrillo- Solorzano-Vargas, J. Wang, E. O’Connor, J. Papesh, M. Larauche, H. Trost, M.N.
Gonzalez, U. Angeles-Garay, A.L. Peralta-Amaro, L. Jara, Effect of pravastatine plus Palgunachari, G.M. Anantharamaiah, H.R. Herschman, M.G. Martin, A.M.
ezetimibe on carotid intima media thickness in patients with lupus erythemato- Fogelman, S.T. Reddy, Apolipoprotein A-I mimetics mitigate intestinal inflamma-
sus, Revista medica del Instituto Mexicano del Seguro Social. 53 (Suppl. 1) tion in COX2-dependent inflammatory bowel disease model, J. Clin. Invest. 129
(2015) S74–S79. (9) (2019) 3670–3685.
[317] C.M. Shih, C.C. Chen, C.K. Chu, K.H. Wang, C.Y. Huang, A.W. Lee, The roles of lipo- [340] T.M. Nowacki, A.T. Remaley, D. Bettenworth, M. Eisenblatter, T. Vowinkel, F. Becker,
protein in psoriasis, Int. J. Mol. Sci. 21 (3) (2020). T. Vogl, J. Roth, U.J. Tietge, A. Lugering, J. Heidemann, J.R. Nofer, The 5A apolipopro-
[318] V.E. Friedewald, J.C. Cather, J.M. Gelfand, K.B. Gordon, G.H. Gibbons, S.M. Grundy, tein a-I (apoA-I) mimetic peptide ameliorates experimental colitis by regulating
M.T. Jarratt, J.G. Krueger, P.M. Ridker, N. Stone, W.C. Roberts, AJC editor’s consen- monocyte infiltration, Br. J. Pharmacol. 173 (18) (2016) 2780–2792.
sus: psoriasis and coronary artery disease, Am. J. Cardiol. 102 (12) (2008) [341] D.A. Hottman, D. Chernick, S. Cheng, Z. Wang, L. Li, HDL and cognition in neurode-
1631–1643. generative disorders, Neurobiol. Dis. 72 (Pt A) (2014) 22–36.
[319] A.B. Gottlieb, F. Dann, Comorbidities in patients with psoriasis, Am. J. Med. 122 [342] J.M. Dietschy, S.D. Turley, Cholesterol metabolism in the brain, Curr. Opin. Lipidol.
(12) (2009)1150 e1-9. 12 (2) (2001) 105–112.
[320] A.M. Tobin, D.J. Veale, O. Fitzgerald, S. Rogers, P. Collins, D. O’Shea, B. Kirby, Cardio- [343] C. Reitz, M.X. Tang, N. Schupf, J.J. Manly, R. Mayeux, J.A. Luchsinger, Association of
vascular disease and risk factors in patients with psoriasis and psoriatic arthritis, J. higher levels of high-density lipoprotein cholesterol in elderly individuals and
Rheumatol. 37 (7) (2010) 1386–1394. lower risk of late-onset Alzheimer disease, Arch. Neurol. 67 (12) (2010)
[321] S.M. Langan, N.M. Seminara, D.B. Shin, A.B. Troxel, S.E. Kimmel, N.N. Mehta, D.J. 1491–1497.
Margolis, J.M. Gelfand, Prevalence of metabolic syndrome in patients with psoria- [344] J.S. Saczynski, L. White, R.L. Peila, B.L. Rodriguez, L.J. Launer, The relation between
sis: a population-based study in the United Kingdom, J. Invest. Dermatol. 132 (3 Pt apolipoprotein A-I and dementia: the Honolulu-Asia aging study, Am. J. Epidemiol.
1) (2012) 556–562. 165 (9) (2007) 985–992.
[322] J.M. Fernandez-Armenteros, X. Gomez-Arbones, M. Buti-Soler, A. Betriu-Bars, V. [345] K.A. Bates, H.R. Sohrabi, S.R. Rainey-Smith, M. Weinborn, R.S. Bucks, M. Rodrigues, J.
Sanmartin-Novell, M. Ortega-Bravo, M. Martinez-Alonso, E. Gari, M. Portero-Otin, Beilby, M. Howard, K. Taddei, G. Martins, A. Paton, T. Shah, S.S. Dhaliwal, J.K. Foster,
L. Santamaria-Babi, J.M. Casanova-Seuma, Psoriasis, metabolic syndrome and car- I.J. Martins, N.T. Lautenschlager, F.L. Mastaglia, S.E. Gandy, R.N. Martins, Serum
diovascular risk factors. A population-based study, JEADV. 33 (1) (2019) 128–135. high-density lipoprotein is associated with better cognitive function in a cross-
[323] E.J. Samarasekera, J.M. Neilson, R.B. Warren, J. Parnham, C.H. Smith, Incidence of sectional study of aging women, Int. J. Neurosci. 127 (3) (2017) 243–252.
cardiovascular disease in individuals with psoriasis: a systematic review and [346] F. Song, A. Poljak, J. Crawford, N.A. Kochan, W. Wen, B. Cameron, O. Lux, H. Brodaty,
meta-analysis, J. Invest. Dermatol. 133 (10) (2013) 2340–2346. K. Mather, G.A. Smythe, P.S. Sachdev, Plasma apolipoprotein levels are associated
[324] A.G. Moraitis, L.A. Freeman, R.D. Shamburek, R. Wesley, W. Wilson, C.M. Grant, S. with cognitive status and decline in a community cohort of older individuals,
Price, S. Demosky, S.G. Thacker, A. Zarzour, R.L. Hornung, F. Pucino, G. Csako, C. PLoS One 7 (6) (2012), e34078.
Yarboro, I.B. McInnes, T. Kuroiwa, D. Boumpas, V.K. Rao, G.G. Illei, A.T. Remaley, El- [347] I.E. Jansen, J.E. Savage, K. Watanabe, J. Bryois, D.M. Williams, S. Steinberg, J. Sealock,
evated interleukin-10: a new cause of dyslipidemia leading to severe HDL defi- I.K. Karlsson, S. Hagg, L. Athanasiu, N. Voyle, P. Proitsi, A. Witoelar, S. Stringer, D.
ciency, J. Clin. Llipidol. 9 (1) (2015) 81–90. Aarsland, I.S. Almdahl, F. Andersen, S. Bergh, F. Bettella, S. Bjornsson, A. Braekhus,
[325] M.J. Paiva-Lopes, Alves J. Delgado, Psoriasis-associated vascular disease: the role of G. Brathen, C. de Leeuw, R.S. Desikan, S. Djurovic, L. Dumitrescu, T. Fladby, T.J.
HDL, J. Biomed. Sci. 24 (1) (2017) 73. Hohman, P.V. Jonsson, S.J. Kiddle, A. Rongve, I. Saltvedt, S.B. Sando, G. Selbaek, M.
Shoai, N.G. Skene, J. Snaedal, E. Stordal, I.D. Ulstein, Y. Wang, L.R. White, J. Hardy,
[326] A. Goren, F. Ozsolak, N. Shoresh, M. Ku, M. Adli, C. Hart, M. Gymrek, O. Zuk, A.
J. Hjerling-Leffler, P.F. Sullivan, W.M. van der Flier, R. Dobson, L.K. Davis, H.
Regev, P.M. Milos, B.E. Bernstein, Chromatin profiling by directly sequencing
Stefansson, K. Stefansson, N.L. Pedersen, S. Ripke, O.A. Andreassen, D. Posthuma,
small quantities of immunoprecipitated DNA, Nat. Methods 7 (1) (2010) 47–49.
Genome-wide meta-analysis identifies new loci and functional pathways influenc-
[327] M. Holzer, P. Wolf, M. Inzinger, M. Trieb, S. Curcic, L. Pasterk, W. Weger, A.
ing Alzheimer’s disease risk, Nat. Genet. 51 (3) (2019) 404–413.
Heinemann, G. Marsche, Anti-psoriatic therapy recovers high-density lipoprotein
[348] B.W. Kunkle, B. Grenier-Boley, R. Sims, J.C. Bis, V. Damotte, A.C. Naj, A. Boland, M.
composition and function, J. Invest. Dermatol. 134 (3) (2014) 635–642.
Vronskaya, S.J. van der Lee, A. Amlie-Wolf, C. Bellenguez, A. Frizatti, V. Chouraki,
[328] G. Marsche, M. Holzer, P. Wolf, Antipsoriatic treatment extends beyond the skin:
E.R. Martin, K. Sleegers, N. Badarinarayan, J. Jakobsdottir, K.L. Hamilton-Nelson, S.
recovering of high-density lipoprotein function, Exp. Dermatol. 23 (10) (2014)
Moreno-Grau, R. Olaso, R. Raybould, Y. Chen, A.B. Kuzma, M. Hiltunen, T.
701–704.
Morgan, S. Ahmad, B.N. Vardarajan, J. Epelbaum, P. Hoffmann, M. Boada, G.W.
[329] G. Marsche, M.D. Saemann, A. Heinemann, M. Holzer, Inflammation alters HDL Beecham, J.G. Garnier, D. Harold, A.L. Fitzpatrick, O. Valladares, M.L. Moutet, A.
composition and function: implications for HDL-raising therapies, Pharmacol. Gerrish, A.V. Smith, L. Qu, D. Bacq, N. Denning, X. Jian, Y. Zhao, M. Del Zompo,
Ther. 137 (3) (2013) 341–351. N.C. Fox, S.H. Choi, I. Mateo, J.T. Hughes, H.H. Adams, J. Malamon, F. Sanchez-
[330] R.J. Xavier, D.K. Podolsky, Unravelling the pathogenesis of inflammatory bowel dis- Garcia, Y. Patel, J.A. Brody, B.A. Dombroski, Naranjo MCD, M. Daniilidou, G.
ease, Nature. 448 (7152) (2007) 427–434. Eiriksdottir, S. Mukherjee, D. Wallon, J. Uphill, T. Aspelund, L.B. Cantwell, F.
[331] A.N. Ananthakrishnan, C.N. Bernstein, D. Iliopoulos, A. Macpherson, M.F. Neurath, Garzia, D. Galimberti, E. Hofer, M. Butkiewicz, B. Fin, E. Scarpini, C. Sarnowski,
R.A.R. Ali, S.R. Vavricka, C. Fiocchi, Environmental triggers in IBD: a review of prog- W.S. Bush, S. Meslage, J. Kornhuber, C.C. White, Y. Song, R.C. Barber, S.
ress and evidence, Nat. Rev. Gastroenterol. Hepatol. 15 (1) (2018) 39–49. Engelborghs, S. Sordon, D. Voijnovic, P.M. Adams, R. Vandenberghe, M. Mayhaus,
[332] J.Z. Liu, S. van Sommeren, H. Huang, S.C. Ng, R. Alberts, A. Takahashi, S. Ripke, J.C. L.A. Cupples, M.S. Albert, P.P. De Deyn, W. Gu, J.J. Himali, D. Beekly, A. Squassina,
Lee, L. Jostins, T. Shah, S. Abedian, J.H. Cheon, J. Cho, N.E. Dayani, L. Franke, Y. A.M. Hartmann, A. Orellana, D. Blacker, E. Rodriguez-Rodriguez, S. Lovestone,
Fuyuno, A. Hart, R.C. Juyal, G. Juyal, W.H. Kim, A.P. Morris, H. Poustchi, W.G. M.E. Garcia, R.S. Doody, C. Munoz-Fernadez, R. Sussams, H. Lin, T.J. Fairchild, Y.A.
Newman, V. Midha, T.R. Orchard, H. Vahedi, A. Sood, J.Y. Sung, R. Malekzadeh, Benito, C. Holmes, H. Karamujic-Comic, M.P. Frosch, H. Thonberg, W. Maier, G.
H.J. Westra, K. Yamazaki, S.K. Yang, International Multiple Sclerosis Genetics C, Roshchupkin, B. Ghetti, V. Giedraitis, A. Kawalia, S. Li, R.M. Huebinger, L. Kilander,
International IBDGC, J.C. Barrett, B.Z. Alizadeh, M. Parkes, T. Bk, M.J. Daly, M. S. Moebus, I. Hernandez, M.I. Kamboh, R. Brundin, J. Turton, Q. Yang, M.J. Katz, L.
Kubo, C.A. Anderson, R.K. Weersma, Association analyses identify 38 susceptibility Concari, J. Lord, A.S. Beiser, C.D. Keene, S. Helisalmi, I. Kloszewska, W.A. Kukull,
loci for inflammatory bowel disease and highlight shared genetic risk across pop- A.M. Koivisto, A. Lynch, L. Tarraga, E.B. Larson, A. Haapasalo, B. Lawlor, T.H.
ulations, Nat. Genet. 47 (9) (2015) 979–986. Mosley, R.B. Lipton, V. Solfrizzi, M. Gill, W.T. Longstreth Jr., T.J. Montine, V.
[333] R. Gerster, J.J. Eloranta, M. Hausmann, P.A. Ruiz, J. Cosin-Roger, A. Terhalle, U. Frisardi, M. Diez-Fairen, F. Rivadeneira, R.C. Petersen, V. Deramecourt, I. Alvarez,
Ziegler, G.A. Kullak-Ublick, A. von Eckardstein, G. Rogler, Anti-inflammatory func- F. Salani, A. Ciaramella, E. Boerwinkle, E.M. Reiman, N. Fievet, J.I. Rotter, J.S.
tion of high-density lipoproteins via autophagy of IkappaB Kinase, Cell. Mol. Reisch, O. Hanon, C. Cupidi, A.G. Andre Uitterlinden, D.R. Royall, C. Dufouil, R.G.
Gastroenterol. Hepatol. 1 (2) (2015)171–87 e1. Maletta, I. de Rojas, M. Sano, A. Brice, R. Cecchetti, P.S. George-Hyslop, K. Ritchie,
[334] R.S. Sappati Biyyani, B.S. Putka, K.D. Mullen, Dyslipidemia and lipoprotein profiles M. Tsolaki, D.W. Tsuang, B. Dubois, D. Craig, C.K. Wu, H. Soininen, D. Avramidou,
in patients with inflammatory bowel disease, J. Clin. Llipidol. 4 (6) (2010) R.L. Albin, L. Fratiglioni, A. Germanou, L.G. Apostolova, L. Keller, M. Koutroumani,
478–482. S.E. Arnold, F. Panza, O. Gkatzima, S. Asthana, D. Hannequin, P. Whitehead, C.S.

117
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

Atwood, P. Caffarra, H. Hampel, I. Quintela, A. Carracedo, L. Lannfelt, D.C. the memory loss of mice with Alzheimer’s disease by accelerating the clearance
Rubinsztein, L.L. Barnes, F. Pasquier, L. Frolich, S. Barral, B. Mcguinness, T.G. of amyloid-beta, ACS Nano 8 (3) (2014) 2345–2359.
Beach, J.A. Johnston, J.T. Becker, P. Passmore, E.H. Bigio, J.M. Schott, T.D. Bird, J.D. [359] M.C. Morris, D.A. Evans, J.L. Bienias, P.A. Scherr, C.C. Tangney, L.E. Hebert, D.A.
Warren, B.F. Boeve, M.K. Lupton, J.D. Bowen, P. Proitsi, A. Boxer, J.F. Powell, J.R. Bennett, R.S. Wilson, N. Aggarwal, Dietary niacin and the risk of incident
Burke, Kauwe JSK, J.M. Burns, M. Mancuso, J.D. Buxbaum, U. Bonuccelli, N.J. Alzheimer’s disease and of cognitive decline, J. Neurol. Neurosurg. Psychiatry 75
Cairns, A. Mcquillin, C. Cao, G. Livingston, C.S. Carlson, N.J. Bass, C.M. Carlsson, J. (8) (2004) 1093–1099.
Hardy, R.M. Carney, J. Bras, M.M. Carrasquillo, R. Guerreiro, M. Allen, H.C. Chui, E. [360] C. Wakade, R. Chong, E. Bradley, B. Thomas, J. Morgan, Upregulation of GPR109A in
Fisher, C. Masullo, E.A. Crocco, C. Decarli, G. Bisceglio, M. Dick, L. Ma, R. Duara, Parkinson’s disease, PLoS One 9 (10) (2014), e109818.
N.R. Graff-Radford, D.A. Evans, A. Hodges, K.M. Faber, M. Scherer, K.B. Fallon, M. [361] H.A. Salem, W. Wadie, Effect of niacin on inflammation and angiogenesis in a mu-
Riemenschneider, D.W. Fardo, R. Heun, M.R. Farlow, H. Kolsch, S. Ferris, M. Leber, rine model of ulcerative colitis, Sci. Rep. 7 (1) (2017) 7139.
T.M. Foroud, I. Heuser, D.R. Galasko, I. Giegling, M. Gearing, M. Hull, D.H. [362] C. Wakade, B. Giri, A. Malik, H. Khodadadi, J.C. Morgan, R.K. Chong, B. Baban, Niacin
Geschwind, J.R. Gilbert, J. Morris, R.C. Green, K. Mayo, J.H. Growdon, T. Feulner, modulates macrophage polarization in Parkinson’s disease, J. Neuroimmunol. 320
R.L. Hamilton, L.E. Harrell, D. Drichel, L.S. Honig, T.D. Cushion, M.J. Huentelman, P. (2018) 76–79.
Hollingworth, C.M. Hulette, B.T. Hyman, R. Marshall, G.P. Jarvik, A. Meggy, E. [363] I.H. de Boer, J.D. Brunzell, HDL in CKD: how good is the “good cholesterol?”, JASN.
Abner, G.E. Menzies, L.W. Jin, G. Leonenko, L.M. Real, G.R. Jun, C.T. Baldwin, D. 25 (5) (2014) 871–874.
Grozeva, A. Karydas, G. Russo, J.A. Kaye, R. Kim, F. Jessen, N.W. Kowall, B. Vellas,
[364] B. Bowe, Y. Xie, H. Xian, S. Balasubramanian, Z. Al-Aly, Low levels of high-density
J.H. Kramer, E. Vardy, LaFerla FM, K.H. Jockel, J.J. Lah, M. Dichgans, J.B. Leverenz,
lipoprotein cholesterol increase the risk of incident kidney disease and its progres-
D. Mann, A.I. Levey, S. Pickering-Brown, A.P. Lieberman, N. Klopp, K.L. Lunetta,
sion, Kidney Int. 89 (4) (2016) 886–896.
H.E. Wichmann, C.G. Lyketsos, K. Morgan, D.C. Marson, K. Brown, F. Martiniuk, C.
[365] H. Moradi, E. Streja, M.L. Kashyap, N.D. Vaziri, G.C. Fonarow, K. Kalantar-Zadeh, El-
Medway, D.C. Mash, M.M. Nothen, E. Masliah, N.M. Hooper, McCormick WC, A.
evated high-density lipoprotein cholesterol and cardiovascular mortality in main-
Daniele, McCurry SM, A. Bayer, McDavid AN, J. Gallacher, McKee AC, H. van den
tenance hemodialysis patients, Nephrol. Dial. Transplant. 29 (8) (2014)
Bussche, M. Mesulam, C. Brayne, B.L. Miller, S. Riedel-Heller, C.A. Miller, J.W.
1554–1562.
Miller, A. Al-Chalabi, J.C. Morris, C.E. Shaw, A.J. Myers, J. Wiltfang, S. O’Bryant, J.M.
[366] K.H. Nam, T.I. Chang, Y.S. Joo, J. Kim, S. Lee, C. Lee, H.R. Yun, J.T. Park, T.H. Yoo, S.A.
Olichney, V. Alvarez, J.E. Parisi, A.B. Singleton, H.L. Paulson, J. Collinge, W.R. Perry,
Sung, K.B. Lee, K.H. Oh, S.W. Kim, J. Lee, S.W. Kang, K.H. Choi, C. Ahn, S.H. Han, K.-C.
S. Mead, E. Peskind, D.H. Cribbs, M. Rossor, A. Pierce, N.S. Ryan, W.W. Poon, B.
Investigators, Association between serum high-density lipoprotein cholesterol
Nacmias, H. Potter, S. Sorbi, J.F. Quinn, E. Sacchinelli, A. Raj, G. Spalletta, M.
Levels and progression of chronic kidney disease: results from the KNOW-CKD, J.
Raskind, C. Caltagirone, P. Bossu, M.D. Orfei, B. Reisberg, R. Clarke, C. Reitz, A.D.
Am. Heart Assoc. 8 (6) (2019), e011162.
Smith, J.M. Ringman, D. Warden, E.D. Roberson, G. Wilcock, E. Rogaeva, A.C.
Bruni, H.J. Rosen, M. Gallo, R.N. Rosenberg, Y. Ben-Shlomo, M.A. Sager, P. [367] S. Coassin, S. Friedel, A. Kottgen, C. Lamina, F. Kronenberg, Is high-density lipopro-
Mecocci, A.J. Saykin, P. Pastor, M.L. Cuccaro, J.M. Vance, J.A. Schneider, L.S. tein cholesterol causally related to kidney function? evidence from genetic epide-
Schneider, S. Slifer, W.W. Seeley, A.G. Smith, J.A. Sonnen, S. Spina, R.A. Stern, R.H. miological studies, Arterioscler. Thromb. Vasc. Biol. 36 (11) (2016) 2252–2258.
Swerdlow, M. Tang, R.E. Tanzi, J.Q. Trojanowski, J.C. Troncoso, V.M. Van Deerlin, [368] M. Holzer, R. Birner-Gruenberger, T. Stojakovic, D. El-Gamal, V. Binder, C. Wadsack,
L.J. Van Eldik, H.V. Vinters, J.P. Vonsattel, S. Weintraub, K.A. Welsh-Bohmer, K.C. A. Heinemann, G. Marsche, Uremia alters HDL composition and function, JASN. 22
Wilhelmsen, J. Williamson, T.S. Wingo, R.L. Woltjer, C.B. Wright, C.E. Yu, L. Yu, Y. (9) (2011) 1631–1641.
Saba, A. Pilotto, M.J. Bullido, O. Peters, P.K. Crane, D. Bennett, P. Bosco, E. Coto, V. [369] C. Kopecky, B. Genser, C. Drechsler, V. Krane, C.C. Kaltenecker, M. Hengstschlager,
Boccardi, P.L. De Jager, A. Lleo, N. Warner, O.L. Lopez, M. Ingelsson, P. Deloukas, W. Marz, C. Wanner, M.D. Saemann, T. Weichhart, Quantification of HDL proteins,
C. Cruchaga, C. Graff, R. Gwilliam, M. Fornage, A.M. Goate, P. Sanchez-Juan, P.G. cardiac events, and mortality in patients with type 2 diabetes on hemodialysis,
Kehoe, N. Amin, N. Ertekin-Taner, C. Berr, S. Debette, S. Love, L.J. Launer, S.G. CJASN. 10 (2) (2015) 224–231.
Younkin, J.F. Dartigues, C. Corcoran, M.A. Ikram, D.W. Dickson, G. Nicolas, D. [370] S. Yamamoto, P.G. Yancey, T.A. Ikizler, W.G. Jerome, R. Kaseda, B. Cox, A. Bian, A.
Campion, J. Tschanz, H. Schmidt, H. Hakonarson, J. Clarimon, R. Munger, R. Shintani, A.B. Fogo, M.F. Linton, S. Fazio, V. Kon, Dysfunctional high-density lipo-
Schmidt, L.A. Farrer, C. Van Broeckhoven, M. COD, DeStefano AL, L. Jones, J.L. protein in patients on chronic hemodialysis, J. Am. Coll. Cardiol. 60 (23) (2012)
Haines, J.F. Deleuze, M.J. Owen, V. Gudnason, R. Mayeux, V. Escott-Price, B.M. 2372–2379.
Psaty, A. Ramirez, L.S. Wang, A. Ruiz, C.M. van Duijn, P.A. Holmans, S. Seshadri, J. [371] T. Speer, L. Rohrer, P. Blyszczuk, R. Shroff, K. Kuschnerus, N. Krankel, G. Kania, S.
Williams, P. Amouyel, G.D. Schellenberg, J.C. Lambert, M.A. Pericak-Vance, Zewinger, A. Akhmedov, Y. Shi, T. Martin, D. Perisa, S. Winnik, M.F. Muller, U.
Alzheimer Disease Genetics C, European Alzheimer’s Disease I, Cohorts for H, Sester, G. Wernicke, A. Jung, U. Gutteck, U. Eriksson, J. Geisel, J. Deanfield, A. von
Aging Research in Genomic Epidemiology C, Genetic, Environmental Risk in Ad/ Eckardstein, T.F. Luscher, D. Fliser, F.H. Bahlmann, U. Landmesser, Abnormal
Defining Genetic P, Environmental Risk for Alzheimer’s Disease C, Genetic meta- high-density lipoprotein induces endothelial dysfunction via activation of Toll-
analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates like receptor-2, Immunity. 38 (4) (2013) 754–768.
Abeta, tau, immunity and lipid processing, Nat. Genet. 51 (3) (2019) 414–430. [372] S. Zewinger, T. Speer, M.E. Kleber, H. Scharnagl, R. Woitas, P.M. Lepper, K. Pfahler, S.
[349] M. Koch, S.T. DeKosky, M. Goodman, J. Sun, J.D. Furtado, A.L. Fitzpatrick, R.H. Seiler, G.H. Heine, W. Marz, G. Silbernagel, D. Fliser, HDL cholesterol is not associ-
Mackey, T. Cai, O.L. Lopez, L.H. Kuller, K.J. Mukamal, M.K. Jensen, Association of ated with lower mortality in patients with kidney dysfunction, JASN. 25 (5) (2014)
Apolipoprotein E in lipoprotein subspecies with risk of dementia, JAMA Netw. 1073–1082.
Open 3 (7) (2020), e209250. [373] J. Nestor, S. Mohan, A focus on APOL1 and kidney disease, Kidney Int. Rep. 4 (7)
[350] J.K. Kueper, M. Speechley, M. Montero-Odasso, The Alzheimer’s disease assessment (2019) 901–903.
scale-cognitive subscale (ADAS-cog): modifications and responsiveness in pre- [374] L. Calabresi, S. Simonelli, P. Conca, G. Busnach, M. Cabibbe, L. Gesualdo, M. Gigante,
dementia populations. A narrative review, J Alzheimers Dis. 63 (2) (2018) S. Penco, F. Veglia, G. Franceschini, Acquired lecithin:cholesterol acyltransferase
423–444. deficiency as a major factor in lowering plasma HDL levels in chronic kidney dis-
[351] J.K. Qiang, Y.C. Wong, A. Siderowf, H.I. Hurtig, S.X. Xie, V.M. Lee, J.Q. Trojanowski, ease, J. Intern. Med. 277 (5) (2015) 552–561.
D.J. Yearout, Montine T.J. BL, M. Stern, S. Mendick, D. Jennings, C. Zabetian, K. [375] J. Rysz, A. Gluba-Brzozka, M. Rysz-Gorzynska, B. Franczyk, The role and function of
Marek, A.S. Chen-Plotkin, Plasma apolipoprotein A1 as a biomarker for Parkinson HDL in patients with chronic kidney disease and the risk of cardiovascular disease,
disease, Ann. Neurol. 74 (1) (2013) 119–127. Int. J. Mol. Sci. 21 (2) (2020).
[352] X. Zhang, X. Yin, H. Yu, X. Liu, F. Yang, J. Yao, H. Jin, P. Yang, Quantitative proteomic [376] V.G. Cabana, S.S. Gidding, G.S. Getz, J. Chapman, S.T. Shulman, Serum amyloid A and
analysis of serum proteins in patients with Parkinson’s disease using an isobaric high density lipoprotein participate in the acute phase response of Kawasaki dis-
tag for relative and absolute quantification labeling, two-dimensional liquid chro- ease, Pediatr. Res. 42 (5) (1997) 651–655.
matography, and tandem mass spectrometry, Analyst. 137 (2) (2012) 490–495. [377] M.P. Migkos, T.E. Markatseli, C. Iliou, P.V. Voulgari, A.A. Drosos, Effect of
[353] K. Ikeda, Y. Nakamura, T. Kiyozuka, J. Aoyagi, T. Hirayama, R. Nagata, H. Ito, K. hydroxychloroquine on the lipid profile of patients with Sjogren syndrome, J.
Iwamoto, K. Murata, Y. Yoshii, K. Kawabe, Y. Iwasaki, Serological profiles of urate, Rheumatol. 41 (5) (2014) 902–908.
paraoxonase-1, ferritin and lipid in Parkinson’s disease: changes linked to disease [378] C. Papagoras, T.E. Markatseli, I. Saougou, Y. Alamanos, A.K. Zikou, P.V. Voulgari, D.N.
progression, Neurodegener. Dis. 8 (4) (2011) 252–258. Kiortsis, A.A. Drosos, Cardiovascular risk profile in patients with spondyloarthritis,
[354] S.N. Akhmedova, A.K. Yakimovsky, E.I. Schwartz, Paraoxonase 1 Met–Leu 54 poly- Joint Bone Spine. 81 (1) (2014) 57–63.
morphism is associated with Parkinson’s disease, J. Neurol. Sci. 184 (2) (2001) [379] R. Nepomuceno, S.C. Pigossi, L.S. Finoti, S.R.P. Orrico, J.A. Cirelli, S.P. Barros, S.
179–182. Offenbacher, R.M. Scarel-Caminaga, Serum lipid levels in patients with periodontal
[355] E. Zintzaras, G.M. Hadjigeorgiou, Association of paraoxonase 1 gene polymor- disease: a meta-analysis and meta-regression, J. Clin. Periodontol. 44 (12) (2017)
phisms with risk of Parkinson’s disease: a meta-analysis, J. Hum. Genet. 49 (9) 1192–1207.
(2004) 474–481. [380] P.B. Lockhart, A.F. Bolger, P.N. Papapanou, O. Osinbowale, M. Trevisan, M.E. Levison,
K.A. Taubert, J.W. Newburger, H.L. Gornik, M.H. Gewitz, W.R. Wilson, S.C. Smith Jr.,
[356] T.L. Lewis, D. Cao, H. Lu, R.A. Mans, Y.R. Su, L. Jungbauer, M.F. Linton, S. Fazio, M.J.
L.M. Baddour, American Heart Association Rheumatic Fever E, Kawasaki Disease
LaDu, L. Li, Overexpression of human apolipoprotein A-I preserves cognitive func-
Committee of the Council on Cardiovascular Disease in the Young CoE, Prevention
tion and attenuates neuroinflammation and cerebral amyloid angiopathy in a
CoPVD, Council on Clinical C, Periodontal disease and atherosclerotic vascular dis-
mouse model of Alzheimer disease, J. Biol. Chem. 285 (47) (2010) 36958–36968.
ease: does the evidence support an independent association?: a scientific state-
[357] J. Robert, S. Stukas, E. Button, W.H. Cheng, M. Lee, J. Fan, A. Wilkinson, I. Kulic, S.D.
ment from the American Heart Association, Circulation. 125 (20) (2012)
Wright, C.L. Wellington, Reconstituted high-density lipoproteins acutely reduce
2520–2544.
soluble brain Abeta levels in symptomatic APP/PS1 mice, Biochim. Biophys. Acta
[381] M. Messedi, K. Jamoussi, M. Frigui, F. Laporte, M. Turki, K. Chaabouni, E. Mnif, M.
1862 (5) (2016) 1027–1036.
Jaloulli, N. Kaddour, Z. Bahloul, F. Ayedi, Atherogenic lipid profile in Behcet’s dis-
[358] Q. Song, M. Huang, L. Yao, X. Wang, X. Gu, J. Chen, J. Chen, J. Huang, Q. Hu, T. Kang, ease: evidence of alteration of HDL subclasses, Arch. Med. Res. 42 (3) (2011)
Z. Rong, H. Qi, G. Zheng, H. Chen, X. Gao, Lipoprotein-based nanoparticles rescue 211–218.

118
S. Nazir, V. Jankowski, G. Bender et al. Advanced Drug Delivery Reviews 159 (2020) 94–119

[382] C.R. Sirtori, M. Ruscica, L. Calabresi, G. Chiesa, R. Giovannoni, J.J. Badimon, HDL ther- [384] N.J. Woudberg, S. Pedretti, S. Lecour, R. Schulz, N. Vuilleumier, R.W. James, M.A.
apy today: from atherosclerosis, to stent compatibility to heart failure, Ann. Med. Frias, Pharmacological intervention to modulate HDL: what do we target? Front.
51 (7–8) (2019) 345–359. Pharmacol. 8 (2017) 989.
[383] C.E. Kosmas, I. Martinez, A. Sourlas, K.V. Bouza, F.N. Campos, V. Torres, P.D. Montan, [385] S. Ertek, High-density lipoprotein (HDL) dysfunction and the future of HDL, Curr.
E. Guzman, High-density lipoprotein (HDL) functionality and its relevance to ath- Vasc. Pharmacol. 16 (5) (2018) 490–498.
erosclerotic cardiovascular disease, Drugs Context. 7 (2018) 212525. [386] Art SSM, SMART Servier Medical Art, Available from https://smart.servier.com/
2020.

119

You might also like