HDL-Based Nanomedicine in Cardiovascular Disease: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

High-density lipoproteins (HDL) are complex endogenous nanoparticles involved in important functions such as reverse cholesterol transport and immunomodulatory activities, ensuring metabolic homeostasis and vascular health. The ability of HDL to interact with a plethora of immune cells and structural cells places it in the centre of numerous disease pathophysiologies. With a better understanding of the properties of synthetic and reconstituted HDL formulations and an increase in preclinical research, several HDL based nanoformulations have been tested in clinical trials.

  • lipoprotein
  • apolipoprotein
  • HDL
  • nanoparticle
  • nanodisc
  • inflammation
  1. Introduction

Traditionally, HDL cholesterol levels have been used as a biomarker to assess cardiovascular risk. However, there is a shift in approach from simply raising HDL cholesterol levels to improving HDL function. This has led to significant efforts to restore HDL quality and enhance its ability to protect against atherosclerosis. HDL particles are highly enriched in apolipoprotein A-I (ApoA-I), accounting for approximately 70% of the total HDL protein [1]. The self-assembly of HDL as definitive nanoparticles is largely attributed to the structural uniqueness of ApoA-I. ApoA-I interacts with endothelial and immune cells to exert a plethora of anti-inflammatory and protective homeostatic functions, making it a natural choice for HDL nanoparticle design.

The intrinsic characteristics of native HDL have inspired therapeutic strategies for synthetic HDL nanoparticles. Their size, natural and specific targeting properties, long circulating time and stability in vivo make them an ideal drug delivery platform with endogenous protective effects. Apart from these significant advantages, one of the most beneficial aspects associated with synthetic HDL nanoparticles is the elimination of extensive biocompatibility validation. This reduces the tolerability issues which are associated with other classes of non-lipoprotein nanoparticles, their clearance being the most concerning question [2].

  1. ApoA-I Full-Length Protein: Reconstituted HDL (rHDL) Nanoparticles

2.1. Clinical Trials with Non-Lipidated ApoA-I

The administration of ApoA-I isolated from pooled human plasma was attempted in humans owing to favourable data from mouse models of atherosclerosis by overexpressing ApoA-I [3-7]. In theory, this treatment mimics synthesis and release of nascent ApoA-I to some extent, which would mark the first step of HDL biogenesis. ApoA-I applied to the blood leads to the formation of pre-beta HDL through lipid incorporation via ABCA1, increasing the pre-beta HDL pool, and it later develops into fully mature HDL nanoparticles, analogous to native HDL biogenesis. The study confirmed its tolerability in humans, but functional protective readouts were inconclusive in terms of improved cholesterol efflux capacity, owing to high catabolism of the free protein [8]. This clearly highlights the need for the lipidation of ApoA-I prior to administration.

2.2. Clinical Trials with Lipidated ApoA-I: rHDL Formulations

Another strategy of harnessing cardioprotective benefits of ApoA-I was inspired from the discovery of a mutant variant of ApoA-I in Milan, which was termed as ApoA-I Milano (ApoA-IM) thereafter. As an example of a single nucleotide polymorphism, the variant was characterized by the replacement of arginine by cysteine at position 173, which appears to be associated with better vascular health in carriers [9,10]. ApoA-IM administration in atherosclerotic mice showed a significant decline in plaque volume and promoted the resolution of coronary artery disease (CAD) [11,12]. Based on these promising effects, the first randomized controlled clinical trial was designed with the concept of administering full-length recombinant ApoA-IM complexed with POPC, which was called ETC-216 [13]. The results were favourable in terms of reducing plaque volume and CAD regression. A follow-up trial suggested a strong correlation between decreased atheroma volume and compensatory vascular elastic remodeling in a specific, focal manner [14]. Furthermore, an increased understanding of the pharmacokinetic and pharmacodynamic attributes of such interventions were investigated [15]. A similar trial that used the ApoA-IM variant for human trials was the MDCO 216 MILANO PILOT trial and its phase III trial. However, numerous side effects were reported in a dose-dependent manner in association with ETC-216 administration. The studies using POPC with the MDCO-216 formulation (ApoA-IM-POPC) reported a profound increase in ABCA-1 mediated cholesterol efflux in healthy and stable CAD patients [16-19]. The primary differences between the MDCO-216 and ETC-216 studies were the lipid composition of the formulations, protein to phospholipids ratio and the number of patients recruited. Later, it was shown that the MDCO-216 formulation was better tolerated by the immune system than the ETC-216 formulation [20]. However, there is no drug based on the ApoA-IM variant, as the trial has been interrupted several times, e.g., due to difficulties in producing large quantities of the full-length protein, increased production costs, change of company owners, etc. Commercially, the strategy of using recombinant full-length proteins has a few drawbacks. The synthesis of large quantities of the protein is expensive, and maintaining the quality can be a challenging aspect. A reduced half-life of about 3 h and rapid renal catabolism is another major challenge that needs to be overcome. The immuno-stimulation problems of ETC-216 were attributed to presence of host antigens, which puts stricter regulation criteria on the concept of full-length, ApoA-I based therapies.

Following up, wild-type ApoA-I was evaluated for its possible therapeutic potential in HDL-mimicking nanodisc formulations. Combining recombinant ApoA-I with two different types of phospholipids, sphingomyelin and dipalmitoyl phosphoglycerol, CER-001 was synthesized and tested in clinical trials funded by Cerenis Pharmaceuticals. CER-001 has shown considerable potential in mouse models of atherosclerosis [21,22]. In the first double-blind study of multiple intravenous doses of 3 mg/kg, 6 mg/kg, or 12 mg/kg of CER-001 in 507 randomized subjects, the results were not significant in terms of atheroma volume reduction and cumulative coronary stenosis score [23]. A follow-up trial of with serial infusions of CER-001 at a lower dose of 3 mg/kg was designed with 272 patients with acute coronary syndrome, again failing to show a significant improvement in atherosclerotic plaque regression [24]. In 2017, CER-001 was re-studied in a phase I dose optimization clinical trial. The study showed that a single infusion of 45 mg/kg is safe and efficiently mobilizes cholesterol in 32 healthy volunteers [25]. In patients with homozygous familial hypercholesterolemia, prolonged CER-001 infusions demonstrated beneficial effects by significantly reducing mean carotid vessel wall area, suggesting a possible reversal in arterial wall remodeling [26]. Furthermore, in a phase II trial in patients with familial hypoalphalipoproteinemia, CER-001 significantly increased cholesterol mobilization [27]. However, CER-001 did not improve carotid atherosclerosis in patients with genetically very low HDL levels [28]. Overall, CER-001 proved to be well tolerated, but the dose required to potentially see beneficial effects of CER-001 in CAD remains uncertain.

Studies with isolated human ApoA-I were continued by Commonwealth Serum Laboratories (CSL) Behring, testing ApoA-I conjugated with soy phosphatidylcholine in the ERASE trial. Their first formulation, CSL111, was evaluated as short-term infusions in 171 patients testing a lower and a higher dose (40 and 80 mg/kg, respectively). However, the results were not significant in atheroma characterization and coronary angiography results when compared to the placebo group [29]. Moreover, the treatments were associated with a significant increase in bilirubin in the higher dose group, and hypotension was seen in the lower dose group. Following up on the formulation, CSL112 was first tested in rabbits [30] and was later administered in single and multiple doses in healthy subjects, showing a dramatic and immediate increase in ApoA-I levels, cholesterol efflux capacity and cholesterol esterification [31]. In a randomized, multicentre, double-blind phase IIa study in patients with stable atherosclerosis, a single infusion of CSL112 in groups of three doses, 1.7 g, 3.4 g, or 6.8 g, was well tolerated, and the treatment resulted in significant cholesterol mobilization [32,33]. The first part of the AEGIS I study conducted in approximately 1200 CAD patients who had recently experienced an acute myocardial infarction event, was designed to characterize the dosing and safety profile of CSL112 [32]. CSL112 at a dose of 6 g dramatically increased cholesterol efflux to about 300%, which was much more compared to all other previous studies [34]. The efflux capacity was reported to be similar in healthy and stable atherosclerotic patients [35] that remained unaffected in conditions of moderate underlying renal complications [36]. This outcome is of specific relevance, as kidney disease severely limits HDL functionality [37,38]. In the CSL112-related studies, significant emphasis has been placed on improving study design, gaining mechanistic insights, and continuously monitoring off-target/additive drug effects and co-treatment options [39-41]. The CSL112-based studies are one of the few that seek to determine and understand native HDL remodeling and pre-beta HDL formation with exogenous sHDL nanotherapy. The potential beneficial effects of CSL112 are being investigated in an ongoing multicentre, double-blind, randomized, placebo-controlled phase III study (AEGIS II study) including about 17,400 participants [42]. The primary objective of AEGIS II is to investigate the efficacy of CSL112 in reducing the risk of death, myocardial infarction, or stroke from the time of randomization to 90 days in patients with acute coronary symptoms.

  1. Clinical Trials with ApoA-I Mimetics peptides

Despite the protective and anti-inflammatory effects of full-length ApoA-I, there are disadvantages associated with them, such as high production costs and quality control. The first mimetic peptide developed to study the ApoA-I nanodisc formation, 18A, gave deeper understanding of the HDL nanodisc assembly and paved the way for introducing nuances in the structural design to enhance vascular protective features [43,44].

The sHDL nanodisc formulation ETC-642 was developed by Esperion and has been extensively tested for its anti-atherogenic characteristics [45-47]. The ETC-642 complex consisted of ESP-2418, a 22 amino acid ApoA-I mimetic peptide complexed to two phospholipids, sphingomyelin and DPPC. The trials in human subjects proceeded to phase I with good tolerability and evidence of LCAT activation [47]. However, the study was discontinued in 2006 due to Pfizer ownership. Following this first mimetic peptide leading to a clinical trial [48], several studies reported anti-inflammatory properties of ETC-642 in mice and in vitro [47,49,50,51]. Currently, ETC-642 is not being tested in clinical trials.

The 4F peptide developed by Novartis was tested, for the first time, in oral formulations [52-56]. The 4F peptide was formulated with both D- and L-amino acids, which had similar biological characteristics but different bio-availabilities due to different helicity and amphipathicity. Here, D-4F was well tolerated in a phase I trial, and preliminary data suggested a potent anti-inflammatory activity [57,58]. D-4F showed promising results and the study indicated the intestine as the main site of action with plasma concentrations of D-4F being independent of the route of administration. However, L-4F did not show significant positive results in CAD prevention [59]. In vitro studies have demonstrated 4F-mediated HDL remodeling [60]. It should be emphasized here that the peptides were not lipidated before administration.

The ApoA-I mimetic peptide 5A produced by KlineMed. Inc. is currently another promising candidate for atherosclerosis prevention, as shown by in vitro and in vivo data from mouse and primate models [61-63]. 5A peptide incorporated into nanodiscs with sphingomyelin has demonstrated high cholesterol efflux capacity, anti-inflammatory and anti-oxidant properties in the animal models and is under safety profile and pharmacokinetic evaluation in a phase I clinical trial. Table 1 summarizes the current strategies and clinical trials with ApoA-I based HDL nanoparticles and mimetic peptides.

 

  1. Conclusion

An overwhelming number of studies highlight the role of HDL in cardiovascular disease. Continuous optimization efforts are required to improve the pharmacological characteristics, such as the half-life and targeting accuracy of synthetic HDL nanoparticles, as demonstrated by recent reports on modified and loaded HDL nanodiscs. The focus of new HDL therapeutics has shifted from increasing circulating HDL-C concentrations to improving HDL function, particularly HDL cholesterol efflux capacity, which is impaired in patients with cardiovascular disease. In summary, there is now sufficient evidence from preclinical and clinical studies that the enhancement/restoration of cholesterol efflux by HDL mimetic nanoparticles is a valid strategy to stabilize atherosclerotic plaque through various anti-inflammatory/immunoregulatory processes. In addition, the immunomodulatory effects of HDL nanoparticles on monocytes and macrophages enable multiple cardioprotective properties that improve vascular health. Data from the ongoing phase III AEGIS-II trial testing CSL112 may represent a breakthrough in the treatment paradigm for acute myocardial infarction, particularly in the immediate weeks and months after the event, when patients are at high risk for the recurrence of ischemic events.

Table 1. Summary of HDL nanoparticles and ApoA-I mimetics in clinical trials.

HDL
Mimetic

Protein Type

Peptide Sequence
(P)

Phospholipid
(L)

Nanoparticle Size

Ratio
(P/L)

Administration

Patients
Recruited

Clinical Trial

Major Findings

Reason to Discontinue Clinical Trial

ETC-216

Recombinant ApoA-IM

ApoA-I
Milano

DPPC

7–30 nm

1:2.7 (w/w)

5 weekly,
intravenous, 15 mg/kg and 45 mg/kg

57

Milano, Phase II

Significantly decreased atheroma volume

Change of company ownership; protein production and quality constrain

MDCO-206

Recombinant ApoA-IM

ApoA-I
Milano

POPC

7–30 nm

1:1.1 (w/w)

5 weekly, intravenous,
20 mg/kg

126

MILANO-PILOT, Phase III,
NCT02678923

80.4% increase in cholesterol efflux, no increment in plaque regression

Change of company ownership; protein production and quality constrain

CER-001

Recombinant ApoA-I

ApoA-I
native

SM, DPPG

7–13 nm

1:2.7 (w/w)

10 weekly, intravenous, 3 mg/kg with statins and 6 weekly, intravenous,
12 mg/kg

507
293

CHI SQUARE, Phase II NCT01201837 and CARAT, Phase II NCT2484378

Failed to promote coronary atherosclerosis regression

Insufficient beneficial effects in CAD

CSL111

Plasma purified human ApoA-I

ApoA-I
native

Soy PC

7–30 nm

1:150 molar ratio

4 weekly,
intravenous,
40 mg/kg and 80 mg/kg

111

ERASE Phase II, NCT00225719

Significant improvement in the plaque characterization index, elevation in liver enzymes in 80 mg/kg group

Modified and improved to CSL112

CSL112

Plasma purified human ApoA-I

ApoA-I
native

Soy PC

7–13 nm

1:55 molar ratio

4 weekly, intravenous, 2 g and 6 g

1258

AEGIS-I, Phase II, NCT02108262

No renal/hepatic toxicity, well tolerated

_

CSL112

Plasma purified human ApoA-I

ApoA-I
native

Soy PC

7–13 nm

1:55 molar ratio

4 weekly intravenous, 6 g

17400

AEGIS-II, Phase III, NCT03473223

Results yet to be concluded

_

ETC-642

ApoA-I mimetic- 22 amino acids, single helix

P-V-L-D-L-F-R-E-L-L-NE-L-L-E-AL-K-Q-K-L-K

DPPC
and
SM

7–13 nm

1:1:1 molar ratio

4 weekly intravenous,
up to 30 mg/kg

28

Phase I

Safe and well tolerated, LCAT activation

Change of company ownership

D-4F

ApoA-I mimetic peptide, 18 amino acids, bihelical

Ac-D-W-F-K-A-F-Y-D-KV-A-E-K-F-K-E-A-FNH 2

Non-
lipidated

-

-

Three separate groups, sequential, ascending doses 100 mg, 300 mg, or 500 mg, oral

104

Phase I, NCT00907998

Increased hydrophobicity, blocks absorption of production of oxidized lipids, anti-inflammatory

Insufficient
bioavailability

L-4F

ApoA-I mimetic peptide, 18 amino acids long, bihelical

4F enantiomer,
L-amino acids

Non-
lipidated

-

-

Single and 7-daily infusions, Intravenous, subcutaneous

176

Phase I, NCT00568594

Similar to 4F but shorter circulating time due to proteolysis

Insufficient
HDL-function-biomarker improvements

5A

ApoA-I mimetic peptide, 37 amino acids, bihelical

D-W-L-K-A-F-Y-D-K-V-A-E-K-L-K-E-A-F-P-D-W-A-K-A-A-Y-D-K-A-A-E-K-A-K-E-A-A

SM

-

1:8
(primate study)

5 weekly, intravenous,
2.5, 5.0, 10.0 and 20.0 mg/kg

64, currently recruiting

Phase I, NCT04216342

ABCA-1 specificity, results yet to be concluded

_

Legends: dipalmitoyl phosphatidylcholine (DPPC), 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), dipalmitoyl phosphatidylglycerol (DPPG), palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1-palmitoyl-2-linoleoyl phosphatidylcholine (PLPC), phosphatidylcholine (PC), sphingomyelin (SM).

References:

  1. Feingold, K.R.; Grunfeld, C. Introduction to Lipids and Lipoproteins. In Endotext; Feingold, K.R., Anawalt, B., Boyce, A., Chrousos, G., Dungan, K., Grossman, A., Hershman, J.M., Kaltsas, G., Koch, C., Kopp, P., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000.
  2. Kuai, R.; Li, D.; Chen, Y.E.; Moon, J.J.; Schwendeman, A. High-Density Lipoproteins: Nature’s Multifunctional Nanoparticles. ACS Nano2016, 10, 3015–3041
  3. Plump, A.S.; Scott, C.J.; Breslow, J.L. Human Apolipoprotein A-I Gene Expression Increases High Density Lipoprotein and Suppresses Atherosclerosis in the Apolipoprotein E-Deficient Mouse. Natl. Acad. Sci. USA1994, 91, 9607–9611. [
  4. Liu, A.C.; Lawn, R.M.; Verstuyft, J.G.; Rubin, E.M. Human Apolipoprotein A-I Prevents Atherosclerosis Associated with Apolipoprotein[a] in Transgenic Mice. Lipid Res.1994, 35, 2263–2267.
  5. Duverger, N.; Kruth, H.; Emmanuel, F.; Caillaud, J.-M.; Viglietta, C.; Castro, G.; Tailleux, A.; Fievet, C.; Fruchart, J.C.; Houdebine, L.M.; et al. Inhibition of Atherosclerosis Development in Cholesterol-Fed Human Apolipoprotein A-I–Transgenic Rabbits. Circulation1996, 94, 713–717.
  6. Zhang, Y.; Zanotti, I.; Reilly, M.P.; Glick, J.M.; Rothblat, G.H.; Rader, D.J. Overexpression of Apolipoprotein A-I Promotes Reverse Transport of Cholesterol from Macrophages to Feces In Vivo. Circulation2003, 108, 661–663.
  7. Valenta, D.T.; Bulgrien, J.J.; Banka, C.L.; Curtiss, L.K. Overexpression of Human ApoAI Transgene Provides Long-Term Atheroprotection in LDL Receptor-Deficient Mice. Atherosclerosis2006, 189, 255–263.
  8. Nanjee, M.N.; Crouse, J.R.; King, J.M.; Hovorka, R.; Rees, S.E.; Carson, E.R.; Morgenthaler, J.-J.; Lerch, P.; Miller, N.E. Effects of Intravenous Infusion of Lipid-Free Apo A-I in Humans. Thromb. Vasc. Biol.1996, 16, 1203–1214.
  9. Franceschini, G.; Sirtori, C.R.; Capurso, A.; Weisgraber, K.H.; Mahley, R.W. A-IMilano Apoprotein. Decreased High Density Lipoprotein Cholesterol Levels with Significant Lipoprotein Modifications and without Clinical Atherosclerosis in an Italian Family. Clin. Investig.1980, 66, 892–900.
  10. Sirtori, C.R.; Calabresi, L.; Franceschini, G.; Baldassarre, D.; Amato, M.; Johansson, J.; Salvetti, M.; Monteduro, C.; Zulli, R.; Muiesan, M.L.; et al. Cardiovascular Status of Carriers of the Apolipoprotein A-I MilanoCirculation 2001, 103, 1949–1954.
  11. Shah, P.K.; Nilsson, J.; Kaul, S.; Fishbein, M.C.; Ageland, H.; Hamsten, A.; Johansson, J.; Karpe, F.; Cercek, B. Effects of Recombinant Apolipoprotein A-IMilanoon Aortic Atherosclerosis in Apolipoprotein E–Deficient Mice. Circulation 1998, 97, 780–785.
  12. Ameli, S.; Hultgardh-Nilsson, A.; Cercek, B.; Shah, P.K.; Forrester, J.S.; Ageland, H.; Nilsson, J. Recombinant Apolipoprotein A-I Milano Reduces Intimal Thickening after Balloon Injury in Hypercholesterolemic Rabbits. Circulation1994, 90, 1935–1941.
  13. Nissen, S.E.; Tsunoda, T.; Tuzcu, E.M.; Schoenhagen, P.; Cooper, C.J.; Yasin, M.; Eaton, G.M.; Lauer, M.A.; Sheldon, W.S.; Grines, C.L.; et al. Effect of Recombinant ApoA-I Milano on Coronary Atherosclerosis in Patients with Acute Coronary Syndromes. JAMA2003, 290, 2292.
  14. Nicholls, S.J.; Tuzcu, E.M.; Sipahi, I.; Schoenhagen, P.; Crowe, T.; Kapadia, S.; Nissen, S.E. Relationship Between Atheroma Regression and Change in Lumen Size After Infusion of Apolipoprotein A-I Milano. Am. Coll. Cardiol.2006, 47, 992–997.
  15. Bisgaier, C.L.; Ackermann, R.; Rea, T.; Rodrigueza, W.V.; Hartman, D. ApoA-IMilano Phospholipid Complex (ETC-216) Infusion in Human Volunteers. Insights into the Phenotypic Characteristics of ApoA-IMilano Carriers. Res2016, 111, 86–99.
  16. Kallend, D.G.; Reijers, J.A.A.; Bellibas, S.E.; Bobillier, A.; Kempen, H.; Burggraaf, J.; Moerland, M.; Wijngaard, P.L.J. A Single Infusion of MDCO-216 (ApoA-1 Milano/POPC) Increases ABCA1-Mediated Cholesterol Efflux and Pre-Beta 1 HDL in Healthy Volunteers and Patients with Stable Coronary Artery Disease. Heart J. Cardiovasc. Pharm.2016, 2, 23–29.
  17. Kempen, H.J.; Gomaraschi, M.; Simonelli, S.; Calabresi, L.; Moerland, M.; Otvos, J.; Jeyarajah, E.; Kallend, D.; Wijngaard, P.L.J. Persistent Changes in Lipoprotein Lipids after a Single Infusion of Ascending Doses of MDCO-216 (ApoA-IMilano/POPC) in Healthy Volunteers and Stable Coronary Artery Disease Patients. Atherosclerosis2016, 255, 17–24
  18. Kempen, H.J.; Asztalos, B.F.; Moerland, M.; Jeyarajah, E.; Otvos, J.; Kallend, D.G.; Bellibas, S.E.; Wijngaard, P.L.J. High-Density Lipoprotein Subfractions and Cholesterol Efflux Capacities After Infusion of MDCO-216 (Apolipoprotein A-IMilano/Palmitoyl-Oleoyl-Phosphatidylcholine) in Healthy Volunteers and Stable Coronary Artery Disease Patients. Thromb. Vasc. Biol.2016, 36, 736–742.
  19. Nicholls, S.J.; Puri, R.; Ballantyne, C.M.; Jukema, J.W.; Kastelein, J.J.P.; Koenig, W.; Wright, R.S.; Kallend, D.; Wijngaard, P.; Borgman, M.; et al. Effect of Infusion of High-Density Lipoprotein Mimetic Containing Recombinant Apolipoprotein A-I Milano on Coronary Disease in Patients with an Acute Coronary Syndrome in the MILANO-PILOT Trial. JAMA Cardiol.2018, 3, 806.
  20. Reijers, J.A.A.; Kallend, D.G.; Malone, K.E.; Jukema, J.W.; Wijngaard, P.L.J.; Burggraaf, J.; Moerland, M. MDCO-216 Does Not Induce Adverse Immunostimulation, in Contrast to Its Predecessor ETC-216. Drugs2017, 31, 381–389.
  21. Tardy, C.; Goffinet, M.; Boubekeur, N.; Cholez, G.; Ackermann, R.; Sy, G.; Keyserling, C.; Lalwani, N.; Paolini, J.F.; Dasseux, J.-L.; et al. HDL and CER-001 Inverse-Dose Dependent Inhibition of Atherosclerotic Plaque Formation in ApoE-/-Mice: Evidence of ABCA1 Down-Regulation. PLoS ONE 2015, 10, e0137584.
  22. Tardy, C.; Goffinet, M.; Boubekeur, N.; Ackermann, R.; Sy, G.; Bluteau, A.; Cholez, G.; Keyserling, C.; Lalwani, N.; Paolini, J.F.; et al. CER-001, a HDL-Mimetic, Stimulates the Reverse Lipid Transport and Atherosclerosis Regression in High Cholesterol Diet-Fed LDL-Receptor Deficient Mice. Atherosclerosis2014, 232, 110–118.
  23. Tardif, J.-C.; Ballantyne, C.M.; Barter, P.; Dasseux, J.-L.; Fayad, Z.A.; Guertin, M.-C.; Kastelein, J.J.P.; Keyserling, C.; Klepp, H.; Koenig, W.; et al. Effects of the High-Density Lipoprotein Mimetic Agent CER-001 on Coronary Atherosclerosis in Patients with Acute Coronary Syndromes: A Randomized Trial. Heart J.2014, 35, 3277–3286.
  24. Nicholls, S.J.; Andrews, J.; Kastelein, J.J.P.; Merkely, B.; Nissen, S.E.; Ray, K.K.; Schwartz, G.G.; Worthley, S.G.; Keyserling, C.; Dasseux, J.-L.; et al. Effect of Serial Infusions of CER-001, a Pre-β High-Density Lipoprotein Mimetic, on Coronary Atherosclerosis in Patients Following Acute Coronary Syndromes in the CER-001 Atherosclerosis Regression Acute Coronary Syndrome Trial. JAMA Cardiol.2018, 3, 815.
  25. Keyserling, C.H.; Barbaras, R.; Benghozi, R.; Dasseux, J.-L. Development of CER-001: Preclinical Dose Selection Through to Phase I Clinical Findings. Drug Investig.2017, 37, 483–491.
  26. Hovingh, G.K.; Smits, L.P.; Stefanutti, C.; Soran, H.; Kwok, S.; de Graaf, J.; Gaudet, D.; Keyserling, C.H.; Klepp, H.; Frick, J.; et al. The Effect of an Apolipoprotein A-I–Containing High-Density Lipoprotein–Mimetic Particle (CER-001) on Carotid Artery Wall Thickness in Patients with Homozygous Familial Hypercholesterolemia. Heart J.2015, 169, 736–742.e1.
  27. Kootte, R.S.; Smits, L.P.; van der Valk, F.M.; Dasseux, J.-L.; Keyserling, C.H.; Barbaras, R.; Paolini, J.F.; Santos, R.D.; van Dijk, T.H.; Dallinga-van Thie, G.M.; et al. Effect of Open-Label Infusion of an ApoA-I-Containing Particle (CER-001) on RCT and Artery Wall Thickness in Patients with FHA. Lipid Res.2015, 56, 703–712.
  28. Zheng, K.H.; Kaiser, Y.; van Olden, C.C.; Santos, R.D.; Dasseux, J.-L.; Genest, J.; Gaudet, D.; Westerink, J.; Keyserling, C.; Verberne, H.J.; et al. No Benefit of HDL Mimetic CER-001 on Carotid Atherosclerosis in Patients with Genetically Determined Very Low HDL Levels. Atherosclerosis2020, 311, 13–19.
  29. Tardif, J.-C.; Grégoire, J.; L’Allier, P.L.; Ibrahim, R.; Lespérance, J.; Heinonen, T.M.; Kouz, S.; Berry, C.; Basser, R.; Lavoie, M.-A.; et al. Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: A randomized controlled trial. JAMA2007, 297, 1675–1682.
  30. Diditchenko, S.; Gille, A.; Pragst, I.; Stadler, D.; Waelchli, M.; Hamilton, R.; Leis, A.; Wright, S.D. Novel Formulation of a Reconstituted High-Density Lipoprotein (CSL112) Dramatically Enhances ABCA1-Dependent Cholesterol Efflux. Thromb. Vasc. Biol.2013, 33, 2202–2211.
  31. Easton, R.; Gille, A.; D’Andrea, D.; Davis, R.; Wright, S.D.; Shear, C. A Multiple Ascending Dose Study of CSL112, an Infused Formulation of ApoA-I. Clin. Pharmacol.2014, 54, 301–310.
  32. Tricoci, P.; D’Andrea, D.M.; Gurbel, P.A.; Yao, Z.; Cuchel, M.; Winston, B.; Schott, R.; Weiss, R.; Blazing, M.A.; Cannon, L.; et al. Infusion of Reconstituted High-Density Lipoprotein, CSL112, in Patients with Atherosclerosis: Safety and Pharmacokinetic Results from a Phase 2a Randomized Clinical Trial. Am. Heart Assoc.2015, 4, e002171.
  33. Gille, A.; Easton, R.; D’Andrea, D.; Wright, S.D.; Shear, C.L. CSL112 Enhances Biomarkers of Reverse Cholesterol Transport After Single and Multiple Infusions in Healthy Subjects. Thromb. Vasc. Biol.2014, 34, 2106–2114.
  34. Michael Gibson, C.; Korjian, S.; Tricoci, P.; Daaboul, Y.; Yee, M.; Jain, P.; Alexander, J.H.; Steg, P.G.; Lincoff, A.M.; Kastelein, J.J.P.; et al. Safety and Tolerability of CSL112, a Reconstituted, Infusible, Plasma-Derived Apolipoprotein A-I, After Acute Myocardial Infarction. Circulation2016, 134, 1918–1930.
  35. Gille, A.; D’Andrea, D.; Tortorici, M.A.; Hartel, G.; Wright, S.D. CSL112 (Apolipoprotein A-I [Human]) Enhances Cholesterol Efflux Similarly in Healthy Individuals and Stable Atherosclerotic Disease Patients. Thromb. Vasc. Biol.2018, 38, 953–963.
  36. Gille, A.; Duffy, D.; Tortorici, M.A.; Wright, S.D.; Deckelbaum, L.I.; D’Andrea, D.M. Moderate Renal Impairment Does Not Impact the Ability of CSL112 (Apolipoprotein A-I [Human]) to Enhance Cholesterol Efflux Capacity. Clin. Pharmacol.2019, 59, 427–436.
  37. Marsche, G.; Heine, G.H.; Stadler, J.T.; Holzer, M. Current Understanding of the Relationship of HDL Composition, Structure and Function to Their Cardioprotective Properties in Chronic Kidney Disease. Biomolecules2020, 10, 1348.
  38. Bauer, L.; Kern, S.; Rogacev, K.S.; Emrich, I.E.; Zawada, A.; Fliser, D.; Heinemann, A.; Heine, G.H.; Marsche, G. HDL Cholesterol Efflux Capacity and Cardiovascular Events in Patients With Chronic Kidney Disease. Am. Coll. Cardiol.2017, 69, 246–247.
  39. Gurbel, P.A.; Tantry, U.S.; D’Andrea, D.; Chung, T.; Alexander, J.H.; Bliden, K.P.; Wright, S.D.; Tricoci, P. Evaluation of Potential Antiplatelet Effects of CSL112 (Apolipoprotein A-I [Human]) in Patients with Atherosclerosis: Results from a Phase 2a Study. Thromb. Thrombolysis2018, 45, 469–476.
  40. Zheng, B.; Duffy, D.; Tricoci, P.; Kastrissios, H.; Pfister, M.; Wright, S.D.; Gille, A.; Tortorici, M.A. Pharmacometric Analyses to Characterize the Effect of CSL112 on Apolipoprotein A-I and Cholesterol Efflux Capacity in Acute Myocardial Infarction Patients. J. Clin. Pharm.2021, 87, 2558–2571.
  41. Beyerle, A.; Greene, B.; Dietrich, B.; Kingwell, B.A.; Panjwani, P.; Wright, S.D.; Herzog, E. Co-Administration of CSL112 (Apolipoprotein A-I [Human]) with Atorvastatin and Alirocumab Is Not Associated with Increased Hepatotoxic or Toxicokinetic Effects in Rats. Appl. Pharm.2021, 422, 115557.
  42. Gibson, C.M.; Kastelein, J.J.P.; Phillips, A.T.; Aylward, P.E.; Yee, M.K.; Tendera, M.; Nicholls, S.J.; Pocock, S.; Goodman, S.G.; Alexander, J.H.; et al. Rationale and Design of ApoA-I Event Reducing in Ischemic Syndromes II (AEGIS-II): A Phase 3, Multicenter, Double-Blind, Randomized, Placebo-Controlled, Parallel-Group Study to Investigate the Efficacy and Safety of CSL112 in Subjects after Acute Myocardial Infarction. Heart J.2021, 231, 121–127.
  43. Chung, B.H.; Anatharamaiah, G.M.; Brouillette, C.G.; Nishida, T.; Segrest, J.P. Studies of Synthetic Peptide Analogs of the Amphipathic Helix. Correlation of Structure with Function. Biol. Chem.1985, 260, 10256–10262.
  44. Ben-Aicha, S.; Casaní, L.; Muñoz-García, N.; Joan-Babot, O.; Peña, E.; Aržanauskaitė, M.; Gutierrez, M.; Mendieta, G.; Padró, T.; Badimon, L.; et al. HDL (High-Density Lipoprotein) Remodeling and Magnetic Resonance Imaging–Assessed Atherosclerotic Plaque Burden. Thromb. Vasc. Biol.2020, 40, 2481–2493.
  45. Di Bartolo, B.A.; Nicholls, S.J.; Bao, S.; Rye, K.-A.; Heather, A.K.; Barter, P.J.; Bursill, C. The Apolipoprotein A-I Mimetic Peptide ETC-642 Exhibits Anti-Inflammatory Properties That Are Comparable to High Density Lipoproteins. Atherosclerosis2011, 217, 395–400.
  46. Iwata, A.; Miura, S.; Zhang, B.; Imaizumi, S.; Uehara, Y.; Shiomi, M.; Saku, K. Antiatherogenic Effects of Newly Developed Apolipoprotein A-I Mimetic Peptide/Phospholipid Complexes against Aortic Plaque Burden in Watanabe-Heritable Hyperlipidemic Rabbits. Atherosclerosis2011, 218, 300–307.
  47. di Bartolo, B.A.; Vanags, L.Z.; Tan, J.T.; Bao, S.; Rye, K.-A.; Barter, P.J.; Bursill, C.A. The Apolipoprotein A-I Mimetic Peptide, ETC-642, Reduces Chronic Vascular Inflammation in the Rabbit. Lipids Health Dis.2011, 10, 224.
  48. Khan, M.L.N.; Drake, S.L.; Crockatt, J.G.; Dasseux, J.L.H. Single-dose intravenous infusion of ETC-642, a 22-Mer ApoA-I analogue and phospholipids complex, elevates HDL-C in atherosclerosis patients. Circulation2003, 108, 563–564.
  49. Guo, L.; Morin, E.E.; Yu, M.; Mei, L.; Fawaz, M.V.; Wang, Q.; Yuan, Y.; Zhan, C.-G.; Standiford, T.J.; Schwendeman, A.; et al. Replenishing HDL with Synthetic HDL Has Multiple Protective Effects against Sepsis in Mice. Signal.2022, 15, eabl9322.
  50. Smith, C.K.; Seto, N.L.; Vivekanandan-Giri, A.; Yuan, W.; Playford, M.P.; Manna, Z.; Hasni, S.A.; Kuai, R.; Mehta, N.N.; Schwendeman, A.; et al. Lupus High-Density Lipoprotein Induces Proinflammatory Responses in Macrophages by Binding Lectin-like Oxidised Low-Density Lipoprotein Receptor 1 and Failing to Promote Activating Transcription Factor 3 Activity. Rheum. Dis.2017, 76, 602–611.
  51. Taylor, M.J.; Sanjanwala, A.R.; Morin, E.E.; Rowland-Fisher, E.; Anderson, K.; Schwendeman, A.; Rainey, W.E. Synthetic High-Density Lipoprotein (SHDL) Inhibits Steroid Production in HAC15 Adrenal Cells. Endocrinology2016, 157, 3122–3129.
  52. Navab, M.; Anantharamaiah, G.M.; Reddy, S.T.; Hama, S.; Hough, G.; Grijalva, V.R.; Wagner, A.C.; Frank, J.S.; Datta, G.; Garber, D.; et al. Oral D-4F Causes Formation of Pre-β High-Density Lipoprotein and Improves High-Density Lipoprotein–Mediated Cholesterol Efflux and Reverse Cholesterol Transport from Macrophages in Apolipoprotein E–Null Mice. Circulation2004, 109, 3215–3220.
  53. Ou, Z.; Ou, J.; Ackerman, A.W.; Oldham, K.T.; Pritchard, K.A. L-4F, an Apolipoprotein A-1 Mimetic, Restores Nitric Oxide and Superoxide Anion Balance in Low-Density Lipoprotein-Treated Endothelial Cells. Circulation2003, 107, 1520–1524.
  54. Wool, G.D.; Cabana, V.G.; Lukens, J.; Shaw, P.X.; Binder, C.J.; Witztum, J.L.; Reardon, C.A.; Getz, G.S. 4F Peptide Reduces Nascent Atherosclerosis and Induces Natural Antibody Production in Apolipoprotein E-null Mice. FASEB J.2011, 25, 290–300.
  55. Datta, G. HDL Mimetic Peptide Administration Improves Left Ventricular Filling and Cardiac Output in Lipopolysaccharide-Treated Rats. Clin. Exp. Cardiol.2011, 2, 1000172. [
  56. Moreira, R.S.; Irigoyen, M.C.; Capcha, J.M.C.; Sanches, T.R.; Gutierrez, P.S.; Garnica, M.R.; de Noronha, I.L.; Andrade, L. Synthetic Apolipoprotein A–I Mimetic Peptide 4F Protects Hearts and Kidneys after Myocardial Infarction. J. Physiol.-Regul. Integr. Comp. Physiol.2020, 318, R529–R544.
  57. Bloedon, L.T.; Dunbar, R.; Duffy, D.; Pinell-Salles, P.; Norris, R.; DeGroot, B.J.; Movva, R.; Navab, M.; Fogelman, A.M.; Rader, D.J. Safety, Pharmacokinetics, and Pharmacodynamics of Oral ApoA-I Mimetic Peptide D-4F in High-Risk Cardiovascular Patients. Lipid Res.2008, 49, 1344–1352.
  58. Dunbar, R.L.; Movva, R.; Bloedon, L.T.; Duffy, D.; Norris, R.B.; Navab, M.; Fogelman, A.M.; Rader, D.J. Oral Apolipoprotein A-I Mimetic D-4F Lowers HDL-Inflammatory Index in High-Risk Patients: A First-in-Human Multiple-Dose, Randomized Controlled Trial. Transl. Sci.2017, 10, 455–469.
  59. Watson, C.E.; Weissbach, N.; Kjems, L.; Ayalasomayajula, S.; Zhang, Y.; Chang, I.; Navab, M.; Hama, S.; Hough, G.; Reddy, S.T.; et al. Treatment of Patients with Cardiovascular Disease with L-4F, an Apo-A1 Mimetic, Did Not Improve Select Biomarkers of HDL Function. Lipid Res.2011, 52, 361–373.
  60. Troutt, J.S.; Alborn, W.E.; Mosior, M.K.; Dai, J.; Murphy, A.T.; Beyer, T.P.; Zhang, Y.; Cao, G.; Konrad, R.J. An Apolipoprotein A-I Mimetic Dose-Dependently Increases the Formation of Preβ1 HDL in Human Plasma. Lipid Res.2008, 49, 581–587.
  61. Amar, M.J.A.; D’Souza, W.; Turner, S.; Demosky, S.; Sviridov, D.; Stonik, J.; Luchoomun, J.; Voogt, J.; Hellerstein, M.; Sviridov, D.; et al. 5A Apolipoprotein Mimetic Peptide Promotes Cholesterol Efflux and Reduces Atherosclerosis in Mice. Pharmacol. Exp. Ther.2010, 334, 634–641.
  62. Tabet, F.; Remaley, A.T.; Segaliny, A.I.; Millet, J.; Yan, L.; Nakhla, S.; Barter, P.J.; Rye, K.-A.; Lambert, G. The 5A Apolipoprotein A-I Mimetic Peptide Displays Antiinflammatory and Antioxidant Properties In Vivo and In Vitro. Thromb. Vasc. Biol.2010, 30, 246–252.
  63. Bourdi, M.; Amar, M.; Remaley, A.T.; Terse, P.S. Intravenous Toxicity and Toxicokinetics of an HDL Mimetic, Fx-5A Peptide Complex, in Cynomolgus Monkeys. Toxicol. Pharmacol.2018, 100, 59–67.

 

This entry is adapted from the peer-reviewed paper 10.3390/pharmaceutics15051504

This entry is offline, you can click here to edit this entry!
Video Production Service