Acessibilidade / Reportar erro

Very low HDL levels: clinical assessment and management

ABSTRACT

In individuals with very low high-density lipoprotein (HDL-C) cholesterol, such as Tangier disease, LCAT deficiency, and familial hypoalphalipoproteinemia, there is an increased risk of premature atherosclerosis. However, analyzes based on comparisons of populations with small variations in HDL-C mediated by polygenic alterations do not confirm these findings, suggesting that there is an indirect association or heterogeneity in the pathophysiological mechanisms related to the reduction of HDL-C. Trials that evaluated some of the HDL functions demonstrate a more robust degree of association between the HDL system and atherosclerotic risk, but as they were not designed to modify lipoprotein functionality, there is insufficient data to establish a causal relationship. We currently have randomized clinical trials of therapies that increase HDL-C concentration by various mechanisms, and this HDL-C elevation has not independently demonstrated a reduction in the risk of cardiovascular events. Therefore, this evidence shows that (a) measuring HDL-C as a way of estimating HDL-related atheroprotective system function is insufficient and (b) we still do not know how to increase cardiovascular protection with therapies aimed at modifying HDL metabolism. This leads us to a greater effort to understand the mechanisms of molecular action and cellular interaction of HDL, completely abandoning the traditional view focused on the plasma concentration of HDL-C. In this review, we will detail this new understanding and the new horizon for using the HDL system to mitigate residual atherosclerotic risk.

Keywords
High-density lipoprotein; Tangier disease; LCAT deficiency; familial hypoalphalipoproteinemia; polygenic dyslipidemias; atherosclerosis

INTRODUCTION

Important findings from the Framingham (11 Castelli WP, Doyle JT, Gordon T, Hames CG, Hjortland MC, Hulley SB, et al. HDL cholesterol and other lipids in coronary heart disease. The cooperative lipoprotein phenotyping study. Circulation. 1977;55(5):767-72.), Tromsø Heart (22 Mora S, Glynn RJ, Ridker PM. High-density lipoprotein cholesterol, size, particle number, and residual vascular risk after potent statin therapy. Circulation. 2013;128(11):1189-97.) and Prospective Cardiovascular Munster (PROCAM) (33 Assmann G, Schulte H, von Eckardstein A, Huang Y. High-density lipoprotein cholesterol as a predictor of coronary heart disease risk. The PROCAM experience and pathophysiological implications for reverse cholesterol transport. Atherosclerosis. 1996;124 Suppl:S11-20.) studies revealed an inverse relationship between serum concentrations of high-density lipoprotein (HDL) and the risk of cardiovascular disease (CVD), independently of the levels of low-density lipoprotein cholesterol (LDL-C). However, in recent decades it has been increasingly questioned whether approaches to improve HDL-related atheroprotective system function will actually reduce the risk of atherosclerosis. The CANHEART (44 Ko DT, Alter DA, Guo H, Koh M, Lau G, Austin PC, et al. High-Density Lipoprotein Cholesterol and Cause-Specific Mortality in Individuals Without Previous Cardiovascular Conditions: The CANHEART Study. J Am Coll Cardiol. 2016;68(19):2073-83.) study showed that high levels of HDL cholesterol (HDL-C) did not reduce mortality from CVD, indicating that high HDL-C is not necessarily associated with cardioprotection. Also, a genetic study showed that three functional variants of hepatic lipase associated with a modest increase in HDL-C levels did not reduce cardiovascular risk (55 Vergeer M, Holleboom AG, Kastelein JJ, Kuivenhoven JA. The HDL hypothesis: does high-density lipoprotein protect from atherosclerosis? J Lipid Res. 2010;51(8):2058-73.). On the other hand, a 29.3% reduction in HDL-C levels due to functional mutations in the ATP Binding Cassette-A1 (ABCA1) transporter did not adversely affect cardiovascular risk (66 Frikke-Schmidt R, Nordestgaard BG, Stene MC, Sethi AA, Remaley AT, Schnohr P, et al. Association of loss-of-function mutations in the ABCA1 gene with high-density lipoprotein cholesterol levels and risk of ischemic heart disease. JAMA. 2008;299(21):2524-32.). Mendelian randomization studies demonstrated a lack of causal link between low HDL-C and the development of atherosclerosis (77 Haase CL, Tybjærg-Hansen A, Qayyum AA, Schou J, Nordestgaard BG, Frikke-Schmidt R. LCAT, HDL cholesterol and ischemic cardiovascular disease: a Mendelian randomization study of HDL cholesterol in 54,500 individuals. J Clin Endocrinol Metab. 2012;97(2):E248-56.), that is, low HDL-C levels are strongly associated with an increased risk of myocardial infarction, but when these levels are genetically determined, an association does not exist (77 Haase CL, Tybjærg-Hansen A, Qayyum AA, Schou J, Nordestgaard BG, Frikke-Schmidt R. LCAT, HDL cholesterol and ischemic cardiovascular disease: a Mendelian randomization study of HDL cholesterol in 54,500 individuals. J Clin Endocrinol Metab. 2012;97(2):E248-56.). However, studies of HDL functionality showed an even more evident association with cardiovascular risk than HDL-C levels (88 Sposito AC. HDL metrics, let’s call the number thing off? Atherosclerosis. 2016;251:525-7.), indicating that HDL functionality plays a more relevant role in atheroprotection than its circulating levels.

Very low HDL-C is seen in clinical conditions that differ from each other in terms of their potential for the development of atherosclerotic disease. Individuals afflicted with single-gene diseases are at high risk of premature atherosclerosis (99 Posadas-Sánchez R, Posadas-Romero C, Ocampo-Arcos WA, Villarreal-Molina MT, Vargas-Alarcón G, Antúnez-Argüelles E, et al. Premature and severe cardiovascular disease in a Mexican male with markedly low high-density-lipoprotein-cholesterol levels and a mutation in the lecithin: cholesterol acyltransferase gene: a family study. Int J Mol Med. 2014;33(6):1570-6.). On the other hand, less severe mutations in genes related to intravascular metabolism or HDL synthesis (1010 Brunham LR, Hayden MR. Human genetics of HDL: Insight into particle metabolism and function. Prog Lipid Res. 2015;58:14-25.) do not result in an increased risk of atherosclerotic disease (1111 Voight BF, Peloso GM, Orho-Melander M, Frikke-Schmidt R, Barbalic M, Jensen MK, et al. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet. 2012;380(9841):572-80.). In contrast, low plasma HDL-C can also be seen in insulin resistance and subsequent atherogenic dyslipidemia, characterized by low HDL-C levels, high triglyceride levels, and an increased proportion of small, dense lipoproteins both HDL and low-density lipoprotein (LDL) (1212 Nesto RW. Beyond low-density lipoprotein: addressing the atherogenic lipid triad in type 2 diabetes mellitus and the metabolic syndrome. Am J Cardiovasc Drugs. 2005;5(6):379-87.). In this review, we will detail the clinical conditions that occur with low levels of HDL-C.

HDL metabolism

HDL particles are present in the circulation in different sizes (7-12 nm) and densities (1,063-1,21 g/mL) and represent the HDL pool in the course of its maturation after hepatic and intestinal production of apolipoprotein (apo)A-I, which is functionally and structurally the most important protein of HDL. Pre-β, discoid, small and lipid-poor HDL captures free cholesterol after binding between apoA-I and the ABCA1 receptor of peripheral cells, allowing reverse cholesterol transport (1313 Rothblat GH, Phillips MC. High-density lipoprotein heterogeneity and function in reverse cholesterol transport. Curr Opin Lipidol. 2010;21(3):229-38.).

At the same time, HDL particles are also enriched with phospholipids derived from other lipoproteins, especially very low-density lipoprotein (VLDL), by phospholipid transfer protein (PLTP). For the formation of HDL particles with a lipid and hydrophilic core, one of the enzymes associated with HDL, the enzyme lecithin-cholesterol acyltransferase (LCAT), catalyzes the transfer of fatty acids from phospholipids to cholesterol, esterifying it and promoting the formation of HDL particles rich in cholesterol esters. Reverse cholesterol transport is completed when HDL is taken up by hepatic Scavenger Receptor-B1 (SR-B1) receptors, initiating the process of excretion through the bile. On the other hand, cholesteryl esters can also be transferred to lipoproteins that contain apoB by the action of Cholesteryl Ester Transfer Protein (CETP), through exchange for triglycerides (1414 Eckardstein VA, Kardassis D. High Density Lipoproteins From biological understanding to clinical exploitation. Springer Open; 2015.).

As a carrier of several proteins, including acute phase enzymes, and a small amount of non-polar lipids, HDL is found in the circulation with different phenotypes and biological properties. While some of these actions (such as antioxidant activity) are intensely exerted by immature forms of HDL (HDL3), reverse cholesterol transport and anti-inflammatory action on the endothelium appear to require more mature forms of HDL (HDL2) (1515 Kontush A, Chapman MJ. Antiatherogenic function of HDL particle subpopulations: focus on antioxidative activities. Curr Opin Lipidol. 2010;21(4):312-8.). This broad set of actions is the result of several components of HDL, such as apoA-I, apoA-II, apoJ, as well as other proteins, enzymes, phospholipids and even probably microRNAs that are transported by HDL particles (1616 Prosser HC, Ng MKC, Bursill CA. The role of cholesterol efflux in mechanisms of endothelial protection by HDL. Curr Opin Lipidol. 2012;23(3):182-9.,1717 Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13(4):423-33.).

Monogenic dyslipidemias

Extremely low serum HDL-C levels (<30-35 mg/dL) in the absence of secondary causes occur in 1% of the general population (1818 Beheshti SO, Madsen CM, Varbo A, Nordestgaard BG. Worldwide Prevalence of Familial Hypercholesterolemia: Meta-Analyses of 11 Million Subjects. J Am Coll Cardiol. 2020;75(20):2553-66.) and correspond to rare and underdiagnosed genetic syndromes caused by mutations in genes that regulate HDL production or catabolism. According to population studies, 18.7% of individuals with very low HDL-C carry rare genetic variants of great effect and 19.3% carry common low-effect variants (1919 Dron JS, Wang J, Low-Kam C, Khetarpal SA, Robinson JF, McIntyre AD, et al. Polygenic determinants in extremes of high-density lipoprotein cholesterol. J Lipid Res. 2017;58(11):2162-70.). Thus, the genetic basis of very low HDL-C disease is often polygenic.

Tangier disease

The ABCA1 gene resides on chromosome 9q22-q31, contains 50 exons and encodes a long membrane protein of 2,261 amino acids. It consists of two transmembrane domains, each formed by six alpha helices and two intracellular nucleotide-binding domains (2020 von Eckardstein A. Differential diagnosis of familial high density lipoprotein deficiency syndromes. Atherosclerosis. 2006;186(2):231-9.). Tangier disease is a rare monogenic autosomal recessive disorder that occurs due to mutations in both alleles of the ABCA1 gene, both exonic (2121 El Khoury P, Couvert P, Elbitar S, Ghaleb Y, Abou-Khalil Y, Azar Y, et al. Identification of the first Tangier disease patient in Lebanon carrying a new pathogenic variant in ABCA1. J Clin Lipidol. 2018;12(6):1374-82.) and intronic in ABCA1 causing aberrant splicing of mRNA (2222 Maranghi M, Truglio G, Gallo A, Grieco E, Verrienti A, Montali A, et al. A novel splicing mutation in the ABCA1 gene, causing Tangier disease and familial HDL deficiency in a large family. Biochem Biophys Res Commun. 2019;508(2):487-93.). Overall, as changes in the ABCA1 gene negatively influence cellular cholesterol uptake, the concentration of HDL-C decreases more sharply and the incidence of coronary artery disease (CAD) increases. Fibroblasts are very deficient in the export of cholesterol and, as a result, complete particles of HDL are not formed (2323 Quintão ECR, Nakandakare ER, Passarelli M. Lípides: do metabolismo à aterosclerose. 1ᵃ ed. São Paulo: Sarvier; 2011.). Therefore, ABCA1 deficiency leads to intracellular accumulation of cholesteryl esters, precluding the conversion of the lipid-poor apoA-I particles into pre-β HDL (2424 Ceccanti M, Cambieri C, Frasca V, Onesti E, Biasiotta A, Giordano C, et al. A Novel Mutation in ABCA1 Gene Causing Tangier Disease in an Italian Family with Uncommon Neurological Presentation. Front Neurol. 2016;7:185.) (Figure 1).

Figure 1
HDL regulation and monogenic disorders with low levels of HDL-C. Black arrows show HDL metabolism and red arrows where monogenic disorders occur in the three conditions reported in this review. 1. The subjects with familial hypoalphalipoproteinemia have been reported to have either decreased HDL production or increased HDL ApoA-I catabolism. 2. The disorder results from homozygous mutations in the ABCA1 transport protein, which mediates the efflux of cellular cholesterol to the HDL particle in plasma for transport to the liver. 3. Both FLD and FED are caused by the mutations in the LCAT gene. Both disorder lead to a marked reduction in plasma HDL-C. ApoA-I: apolipoprotein A-I; PL: phospholipids; FC: free cholesterol; PLTP: Phospholipid Transfer Protein; LPL: lipoprotein lipase; ABCA1: ATP-binding cassette transporter A1; LCAT: Lecithin-cholesterol acyltransferase; ABCG5/8: ATP-binding cassette, subfamily G, member 5 and member 8.

Clinical assessment

Characterized by the almost complete absence of HDL-C (always less than 5 mg/dL), homozygous individuals show a marked increase in apoA-I catabolism, with plasma residence time of about 0.5 days. On the other hand, heterozygous individuals show increased clearance with plasma residence time of about 2 days (2525 Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.). Also, heterozygotes may show a reduction of approximately 50% in ABCA1-mediated cellular cholesterol efflux (2424 Ceccanti M, Cambieri C, Frasca V, Onesti E, Biasiotta A, Giordano C, et al. A Novel Mutation in ABCA1 Gene Causing Tangier Disease in an Italian Family with Uncommon Neurological Presentation. Front Neurol. 2016;7:185.). There is an accumulation of cholesterol esters in the reticuloendothelial system and adipose tissue, in addition to yellow-orange hyperplastic tonsils, which, together with very low levels of HDL-C, are considered indicative of the disease. Individuals have elevated TG and a reduction of up to 50% in LDL-C concentration.

There are controversies in the literature as to whether individuals who are homozygous for Tangiers have an increased risk of developing premature CVD. An analysis involving 185 cases showed that 25% had CVD but among individuals over 40 years of age this prevalence reached 52% compared to 11% in age- and gender-matched controls (2525 Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.). This suggests that these individuals do not develop early CVD because LDL-C levels are about 50% of normal. Furthermore, this report suggested the presence of two distinct phenotypic groups: (i) patients with marked hepatosplenomegaly, anemia, low levels of non-HDL-C (<70 mg/dL) and absence of premature CAD; and (ii) patients without hepatosplenomegaly or severe anemia, normal or near-normal non-HDL-C levels (>70 mg/dL) and premature CAD (2525 Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.). The variability in CVD risk in homozygotes may be, in part, explained by non-HDL-C levels.

In many cases, peripheral neuropathy is absent or only detectable by electrophysiological investigation of nerve conductance velocity. On the other hand, symptomatic individuals present: (i) a multifocal demyelinating form that is mononeuropathic or asymmetric polyneuropathic and affects the motor and sensory nerves of the limbs or head or (ii) a syndrome similar to syringomyelia, being progressive and often debilitating (2020 von Eckardstein A. Differential diagnosis of familial high density lipoprotein deficiency syndromes. Atherosclerosis. 2006;186(2):231-9.).

Clinical management

ABCA1 molecular gene sequencing is the gold standard for diagnosing Tangier disease. However, two tests can help make the diagnosis more likely: two-dimensional non-denaturing electrophoresis and anti-apoA-I immunoblotting help distinguish part of HDL that has alpha electrophoretic mobility (α-HDL) from a quantitatively smaller proportion that has pre-electrophoretic mobility (pre β1-HDL) (2626 von Eckardstein A, Huang Y, Kastelein JJ, Geisel J, Real JT, Kuivenhoven JA, et al. Lipid-free apolipoprotein (apo) A-I is converted into alpha-migrating high density lipoproteins by lipoprotein-depleted plasma of normolipidemic donors and apo A-I-deficient patients but not of Tangier disease patients. Atherosclerosis. 1998;138(1):25-34.).

Other complementary tests may be performed: nerve conduction studies and electroneuromyography to determine the presence of peripheral neuropathy; ophthalmologic evaluation to identify any corneal opacities; abdominal ultrasound to assess hepatosplenomegaly; carotid Doppler ultrasound to identify the thickness of the intima and media layers of the carotid arteries (cIMT) and plaques (2727 Alshaikhli A, Vaqar S. Tangier Disease. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2022 January.); and echocardiogram to assess coronary atherosclerosis.

LCAT deficiency

LCAT deficiency is a very rare autosomal recessive disorder. The majority of described genetic variations are loss-of-function mutations leading to proteins with total or partial lack of LCAT activity (2828 Ossoli A, Lucca F, Boscutti G, Remaley AT, Calabresei L. Familial LCAT deficiency: from pathology to enzyme replacement therapy. Clin Lipidol. 2015;10:405-13.). Genetic LCAT deficiency is associated with the development of two syndromes: (i) familial LCAT deficiency (FLD) and (ii) fish-eye disease (FED). Among the described mutations, 53 are associated with an FLD phenotype and 19 mutations lead to a FED phenotype (2828 Ossoli A, Lucca F, Boscutti G, Remaley AT, Calabresei L. Familial LCAT deficiency: from pathology to enzyme replacement therapy. Clin Lipidol. 2015;10:405-13.). FLD is characterized by mutations that result in the absence or complete inactivity of the LCAT enzyme and present the disease in its severe form, while FED is the result of mutations that inhibit the ability of LCAT to esterify cholesterol to HDL, but do not affect the ability of esterifying cholesterol to lipoproteins that have apoB, so patients are relatively less symptomatic (2929 Saeedi R, Li M, Frohlich J. A review on lecithin: cholesterol acyltransferase deficiency. Clin Biochem. 2015;48(7-8):472-5.) (Figure 1).

In a case series of a Canadian family with LCAT deficiency, no cardiovascular events or deaths were reported for 25 years after the initial diagnosis. In the two homozygous individuals, cIMT was above the 75th percentile expected for age and sex. However, the abnormalities were much more pronounced in the heterozygous individuals, four of whom had detectable plaques, indicating that heterozygosity may be associated with an atherogenic lipid profile and vascular abnormalities (3030 Ayyobi AF, McGladdery SH, Chan S, John Mancini GB, Hill JS, Frohlich JJ. Lecithin: cholesterol acyltransferase (LCAT) deficiency and risk of vascular disease: 25 year follow-up. Atherosclerosis. 2004;177(2):361-6.). Notably, homozygous carriers have low plasma concentration of LDL-C, which would explain protection against atherogenesis.

A more extensive study evaluated cIMT in 40 carriers of LCAT gene mutations and in 80 healthy controls and found no significant difference between mutation carriers (3131 Calabresi L, Baldassarre D, Castelnuovo S, Conca P, Bocchi L, Candini C, et al. Functional lecithin: cholesterol acyltransferase is not required for efficient atheroprotection in humans. Circulation. 2009;120(7):628-35). Divergent results between studies remain unclear about whether low LCAT activity, genetically determined, is associated with increased preclinical atherosclerosis. LCAT, by itself, is not the only regulator of reverse cholesterol transport pathways (3232 Kunnen S, Van Eck M. Lecithin: cholesterol acyltransferase: old friend or foe in atherosclerosis? J Lipid Res. 2012;53(9):1783-99.). Both overexpression and LCAT deficiency showed reverse cholesterol transport from macrophages preserved in an animal model (3333 Tanigawa H, Billheimer JT, Tohyama J, Fuki IV, Ng DS, Rothblat GH, et al. Lecithin: cholesterol acyltransferase expression has minimal effects on macrophage reverse cholesterol transport in vivo. Circulation. 2009;120(2):160-9.). Furthermore, plasma from LCAT-deficient individuals has the same ability to reduce the cholesterol content of macrophages compared to plasma from control individuals (3434 Calabresi L, Favari E, Moleri E, Adorni MP, Pedrelli M, Costa S, et al. Functional LCAT is not required for macrophage cholesterol efflux to human serum. Atherosclerosis. 2009;204(1):141-6.). Thus, it remains questionable whether the elevation of LCAT activity is a promising therapeutic strategy to reduce cardiovascular risk.

Clinical assessment

Individuals affected by FLD develop corneal opacification, anemia and notable proteinuria (3535 Kuivenhoven JA, Pritchard H, Hill J, Frohlich J, Assmann G, Kastelein J. The molecular pathology of lecithin: cholesterol acyltransferase (LCAT) deficiency syndromes. J Lipid Res. 1997;38(2):191-205.). Also, the patients develop early and progressive chronic kidney disease leading to early end-stage renal disease, which is the main cause of morbidity and mortality in this population (3636 Vitali C, Bajaj A, Nguyen C, Schnall J, Chen J, Stylianou K, et al. A systematic review of the natural history and biomarkers of primary Lecithin: Cholesterol Acyltransferase (LCAT) deficiency. J Lipid Res. 2022;63(3):100169.). The reason for the deterioration of renal function remains unknown. Heterogeneous glomerular pathology implies several possible mechanisms, including abnormal LDL trapping and C3 complement deposition (3737 Strøm EH, Sund S, Reier-Nilsen M, Dørje C, Leren TP. Lecithin: Cholesterol Acyltransferase (LCAT) Deficiency: renal lesions with early graft recurrence. Ultrastruct Pathol. 2011;35(3):139-45.). The presence of anemia may occur in association with the disease and is attributed to an increase in the fragility of erythrocytes due to the abnormal lipid composition of their cell membrane. On the other hand, individuals with FED clinically have corneal opacification but are spared anemia and kidney disease and are therefore considered to have a milder form of LCAT deficiency (3838 Suda T, Akamatsu A, Nakaya Y, Masuda Y, Desaki J. Alterations in erythrocyte membrane lipid and its fragility in a patient with familial lecithin: cholesterol acyltrasferase (LCAT) deficiency. J Med Invest. 2002;49(3-4):147-55.).

Corneal opacification, the characteristic physical finding in LCAT deficiency state, is more severe than the accumulation of cholesterol in the cornea that can be sporadically seen in apoA-I deficiency and Tangier disease. Patients with FLD and FED slowly develop progressive corneal opacification accompanied by a white or gray ring at the corneal margin similar to arcus senilis (2020 von Eckardstein A. Differential diagnosis of familial high density lipoprotein deficiency syndromes. Atherosclerosis. 2006;186(2):231-9.). Surprisingly, vision remains intact in most cases.

Prevalence of CAD is higher in FED than in FLD, and this is supported by significantly lower LDL-C in carriers of FLD mutations compared with carriers of FED mutations (3939 Oldoni F, Baldassarre D, Castelnuovo S, Ossoli A, Amato M, van Capelleveen J, et al. Complete and Partial Lecithin:Cholesterol Acyltransferase Deficiency Is Differentially Associated With Atherosclerosis. Circulation. 2018;138(10):1000-7.); however, there are case reports of mutations in LCAT that present low or normal plasma levels of HDL-C without premature CAD (3434 Calabresi L, Favari E, Moleri E, Adorni MP, Pedrelli M, Costa S, et al. Functional LCAT is not required for macrophage cholesterol efflux to human serum. Atherosclerosis. 2009;204(1):141-6.). Assessment of cIMT, a surrogate imaging measure of cardiovascular risk, also suggests that there is no significant worsening of the vascular phenotype among individuals that are homozygous for LCAT mutations.

Clinical management

Initial clinical suspicion is raised based on corneal opacification. The diagnosis of these disorders can be performed through the quantification of LCAT. Definitive diagnosis requires molecular genetic testing of the LCAT gene and functional analysis of the gene product.

Patients with FLD and FED have not only very low levels of HDL-C (<10 mg/dL), but also elevations in VLDL enriched with free cholesterol (2525 Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.). In two-dimensional gel electrophoresis there is the presence of pre-β HDL and α4-HDL and the absence of mature α-HDL (2828 Ossoli A, Lucca F, Boscutti G, Remaley AT, Calabresei L. Familial LCAT deficiency: from pathology to enzyme replacement therapy. Clin Lipidol. 2015;10:405-13.). ApoA-I levels are usually between 20-30 mg/dL and LDL-C concentration is often low. In addition, they have a large, heterogeneous LDL phenotype enriched in free cholesterol, phospholipids and TG, with a very low cholesterol ester content. These particles are also low in apoB and enriched in apoC. The lipid profile is also characterized by the presence of lipoprotein X (Lp-X). Lp-X is a particle that has low protein, esterified cholesterol and TG content, and high free cholesterol and phospholipid content (4040 Fellin R, Manzato E. Lipoprotein-X fifty years after its original discovery. Nutr Metab Cardiovasc Dis. 2019;29(1):4-8.). Experimental studies demonstrate that Lp-X is nephrotoxic, and there is an association with FLD-related kidney disease, showing distinctive lipid deposits in glomeruli in histology, but not with FED (4141 Narayanan S. Lipoprotein-X. CRC Crit Rev Clin Lab Sci. 1979;11(1):31-51.).

Familial hypoalphalipoproteinemia

Familial hypoalphalipoproteinemia is a very rare autosomal dominant disorder characterized by a heterogeneous group of mutations that cause apoA-I deficiency resulting from a biallelic mutation in the APOA1 gene, located on chromosome 11q23.3, which contains four coding regions and is clustered with the apoC-III and apoC-IV genes (4242 Hegele RA, Borén J, Ginsberg HN, Arca M, Averna M, Binder CJ, et al. Rare dyslipidaemias, from phenotype to genotype to management: a European Atherosclerosis Society task force consensus statement. Lancet Diabetes Endocrinol. 2020;8(1):50-67.). Functionally significant mutations have also been described, including gene disruptions, frame changes, nonsense mutations, chromosomal aberrations or deletions, as well as APOA1/APOA3/APOA4 gene cluster inversion associated with decreased HDL-C levels (4343 Schaefer EJ, Genest JJ, Ordovas JM, Salem DN, Wilson PW. Familial lipoprotein disorders and premature coronary artery disease. Atherosclerosis. 1994;108 Suppl:S41-54.

44 Pisciotta L, Miccoli R, Cantafora A, Calabresi L, Tarugi P, Alessandrini P, et al. Recurrent mutations of the apolipoprotein A-I gene in three kindreds with severe HDL deficiency. Atherosclerosis. 2003;167(2):335-45.
-4545 Ikewaki K, Matsunaga A, Han H, Watanabe H, Endo A, Tohyama J, et al. A novel two nucleotide deletion in the apolipoprotein A-I gene, apoA-I Shinbashi, associated with high density lipoprotein deficiency, corneal opacities, planar xanthomas, and premature coronary artery disease. Atherosclerosis. 2004;172(1):39-45.). ApoA-I variants are generally heterozygous premature terminations, frameshifts, or amino acid substitutions in the 243 amino acid sequence of apoA-I (2525 Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.) (Figure 1).

Clinical assessment

Although extremely low HDL-C can be detected in any patient from birth, the age of onset of symptoms and the clinical presentation vary widely. Homozygous or compound heterozygous carriers have two different clinical features: (i) xanthomas or (ii) corneal opacities. Cutaneous xanthomas (tuber-eruptive, tendinous, palmar or planar) have been described in adult patients who are homozygous or compound heterozygous carrying null alleles and therefore do not have apoA-I in their plasma. They often have premature CAD and carotid atherosclerosis (4646 Schaefer EJ, Santos RD, Asztalos BF. Marked HDL deficiency and premature coronary heart disease. Curr Opin Lipidol. 2010;21(4):289-97.).

Heterozygous variants of apoA-I that cause low HDL-C and decreased LCAT activation are not associated with premature CVD. However, apoA-I variants associated with low HDL-C and normal LCAT activity have been associated with premature CVD (2525 Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.). Heterozygous carriers of apoA-I variants do not show specific clinical symptoms. An important exception is some structural variants of apoA-I with amino-terminus amino acid substitutions, detected in patients with familial amyloidosis (4747 Andreola A, Bellotti V, Giorgetti S, Mangione P, Obici L, Stoppini M, et al. Conformational switching and fibrillogenesis in the amyloidogenic fragment of apolipoprotein a-I. J Biol Chem. 2003;278(4):2444-51.

48 Joy T, Wang J, Hahn A, Hegele RA. APOA1 related amyloidosis: a case report and literature review. Clin Biochem. 2003;36(8):641-5.
-4949 Obici L, Palladini G, Giorgetti S, Bellotti V, Gregorini G, Arbustini E, et al. Liver biopsy discloses a new apolipoprotein A-I hereditary amyloidosis in several unrelated Italian families. Gastroenterology. 2004;126(5):1416-22.). These amyloidogenic mutations lead to the accumulation of amyloid containing amino-terminus apoA-I fragments (9-11 kd N terminal fragments (2525 Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.) in the liver, intestine, kidney, heart, peripheral nerves and skin (4747 Andreola A, Bellotti V, Giorgetti S, Mangione P, Obici L, Stoppini M, et al. Conformational switching and fibrillogenesis in the amyloidogenic fragment of apolipoprotein a-I. J Biol Chem. 2003;278(4):2444-51.

48 Joy T, Wang J, Hahn A, Hegele RA. APOA1 related amyloidosis: a case report and literature review. Clin Biochem. 2003;36(8):641-5.
-4949 Obici L, Palladini G, Giorgetti S, Bellotti V, Gregorini G, Arbustini E, et al. Liver biopsy discloses a new apolipoprotein A-I hereditary amyloidosis in several unrelated Italian families. Gastroenterology. 2004;126(5):1416-22.). ApoA-I deficiency can also manifest with sensorineural signs such as cerebellar ataxia, sensorineural hearing loss, and proliferative retinopathy.

In addition, some apoA-I variants such as apoA-I (L178P) or apoA-I (L159P) were associated with increased risk of premature CAD or increased progression of cIMT (5050 Miller M, Aiello D, Pritchard H, Friel G, Zeller K. Apolipoprotein A-I(Zavalla) (Leu159-->Pro): HDL cholesterol deficiency in a kindred associated with premature coronary artery disease. Arterioscler Thromb Vasc Biol. 1998;18(8):1242-7.,5151 Hovingh GK, Brownlie A, Bisoendial RJ, Dube MP, Levels JH, Petersen W, et al. A novel apoA-I mutation (L178P) leads to endothelial dysfunction, increased arterial wall thickness, and premature coronary artery disease. J Am Coll Cardiol. 2004;44(7):1429-35.), while others did not show this association, or even proved to reduce cardiovascular risk remarkably, such as apoA-I Milano (5252 Chiesa G, Sirtori CR. Apolipoprotein A-I(Milano): current perspectives. Curr Opin Lipidol. 2003;14(2):159-63.).

Clinical management

Homozygous carriers have undetectable apoA-I (less than 5 mg/dL) and HDL-C lower than 10 mg/dL. On the other hand, heterozygous carriers generally have HDL-C levels that are often below the fifth percentile or, at least, below the cardiovascular risk threshold level of 40 mg/dL for men and 50 mg/dL for women. As would be expected, apoA-I levels are also frequently below the fifth percentile (<105 mg/dL in men and <110 mg/dL in women) (5353 von Eckardstein A, Funke H, Walter M, Altland K, Benninghoven A, Assmann G. Structural analysis of human apolipoprotein A-I variants. Amino acid substitutions are nonrandomly distributed throughout the apolipoprotein A-I primary structure. J Biol Chem. 1990;265(15):8610-7.).

In a patient with xanthomas, histological examination of the skin lesions reveals numerous foam cells. Molecular diagnosis can be performed by the technique of two-dimensional gel electrophoresis observing the absence or reduction of apoA-I. DNA analysis can also be useful for an accurate diagnosis, performed by sequencing the APOA1 gene and demonstrating a functionally relevant mutation. Many structural variants of apoA-I can be detected by isoelectric focusing and anti-apoA-I immunoblotting (5353 von Eckardstein A, Funke H, Walter M, Altland K, Benninghoven A, Assmann G. Structural analysis of human apolipoprotein A-I variants. Amino acid substitutions are nonrandomly distributed throughout the apolipoprotein A-I primary structure. J Biol Chem. 1990;265(15):8610-7.).

Polygenic dyslipidemias

Recent studies indicate that the genetic basis of extreme concentrations of HDL-C identified clinically is often polygenic, having previously been considered an archetypal “monogenic” disorder. As mentioned above, about 18.7% of individuals with very low HDL-C are heterozygous for rare mutations of great effect and 19.3% of the cases of very low HDL-C show an accumulation of common mutations (1919 Dron JS, Wang J, Low-Kam C, Khetarpal SA, Robinson JF, McIntyre AD, et al. Polygenic determinants in extremes of high-density lipoprotein cholesterol. J Lipid Res. 2017;58(11):2162-70.). In addition, some patients with polygenic dyslipidemia may also have mixed dyslipidemias (low HDL-C and elevations of TG) associated with a different clinical condition such as metabolic syndrome or obesity. Identification of single nucleotide polymorphisms (SNP) that have modest effects on HDL-C plasma have been used to confirm or refute the role of HDL in the development of atherosclerotic disease and as a tool to identify individuals with polygenic dyslipidemias. A classic Mendelian randomization study demonstrated that polymorphism in the lipase gene endothelial cell and a genetic score of 14 common SNP that specifically increased HDL-C were not associated with risk of acute myocardial infarction, suggesting that some genetic mechanisms that increase or decrease HDL-C do not reduce cardiovascular risk (88 Sposito AC. HDL metrics, let’s call the number thing off? Atherosclerosis. 2016;251:525-7.). Another study that used SNP of the target genes as an instrument demonstrated that not all metabolic alterations that modify serum HDL-C levels influence cardiovascular risk (5454 Rohatgi A, Khera A, Berry JD, Givens EG, Ayers CR, Wedin KE, et al. HDL cholesterol efflux capacity and incident cardiovascular events. N Engl J Med. 2014;371(25):2383-93.).

Consistently, studies that used randomization Mendelian research revealed that while the increase of polygenic origin in the concentration of lipoproteins associated with apoB is associated with increased risk of CVD, no association was found with SNP-mediated variation in lipoproteins associated with apoA-I (5555 Richardson TG, Sanderson E, Palmer TM, Ala-Korpela M, Ference BA, Davey Smith G, et al. Evaluating the relationship between circulating lipoprotein lipids and apolipoproteins with risk of coronary heart disease: A multivariable Mendelian randomisation analysis. PLoS Med. 2020;17(3):e1003062.,5656 Karjalainen MK, Holmes MV, Wang Q, Anufrieva O, Kähönen M, Lehtimäki T, et al. Apolipoprotein A-I concentrations and risk of coronary artery disease: A Mendelian randomization study. Atherosclerosis. 2020;299:56-63.). Although these findings may suggest the absence of a causal relationship, plasma levels of HDL-C or apoA-I do not represent reliable instrumental variables, considering that the increase in the circulating concentration of HDL-C or apoA-I does not always equate with a change of roles of the HDL particle or its interaction with cells, i.e., in the HDL system.

Secondary dyslipidemias with low HDL-C

Type 2 diabetes mellitus, insulin resistance, and obesity

Some dyslipidemias are not explained by mutations but are related to epigenetic mechanisms that may be influenced by drug interventions, lifestyle and environmental factors. Reduced levels of HDL-C are often present in resistance to insulin, obesity and type 2 diabetes mellitus (T2DM) and are associated with hypertension and dyslipidemia, factors that can lead to the early development of CAD. In fact, dyslipidemia in T2DM is observed in 60%-70% of patients and is characterized by high levels of TG and decrease in HDL-C, with this reduction being an independent factor not only for the development of CVD, but also for the manifestation of T2DM (5757 Abbasi A, Corpeleijn E, Gansevoort RT, Gans RO, Hillege HL, Stolk RP, et al. Role of HDL cholesterol and estimates of HDL particle composition in future development of type 2 diabetes in the general population: the PREVEND study. J Clin Endocrinol Metab. 2013;98(8):E1352-9.). Also, insulin resistance is known to be associated with lower circulating HDL-C (5858 DeFronzo RA, Ferrannini E. Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care. 1991;14(3):173-94.). These changes together with the presence of small and dense LDL particles contribute to accelerating atherogenesis. In this clinical condition, one of the mechanisms that promote the reduction of HDL-C is elevation of TG-rich lipoprotein levels that, through CETP, transfer TG to HDL in exchange for cholesterol esters (5959 Rashid S, Watanabe T, Sakaue T, Lewis GF. Mechanisms of HDL lowering in insulin resistant, hypertriglyceridemic states: the combined effect of HDL triglyceride enrichment and elevated hepatic lipase activity. Clin Biochem. 2003;36(6):421-9.).

Subsequent removal of this excess HDL-TG by hepatic lipase results in smaller and denser HDL particles. HDL size of person with diabetes is altered, with loss of large and very large HDL2 and gain of small HDL3 (6060 Bonilha I, Zimetti F, Zanotti I, Papotti B, Sposito AC. Dysfunctional High-Density Lipoproteins in Type 2 Diabetes Mellitus: Molecular Mechanisms and Therapeutic Implications. J Clin Med. 2021;10(11).). The same mechanism promotes the formation of small and dense LDL (6161 Bonilha I, Hajduch E, Luchiari B, Nadruz W, Le Goff W, Sposito AC. The Reciprocal Relationship between LDL Metabolism and Type 2 Diabetes Mellitus. Metabolites. 2021;11(12).). In this group of individuals, sub analyses of studies with fibrates and studies with an eicosapentaenoic acid derivative, icosapent ethyl, were associated with a reduction in cardiovascular risk (6262 Jakob T, Nordmann AJ, Schandelmaier S, Ferreira-González I, Briel M. Fibrates for primary prevention of cardiovascular disease events. Cochrane Database Syst Rev. 2016;11:CD009753.). It is not possible, however, to attribute the benefit of this therapy, even if partially, to the increase in HDL-C. So, although the isolated role of HDL is unclear in atherogenic dyslipidemia, its diagnosis remains a prognostic marker in therapies.

Use of anabolic androgenic steroids

Anabolic androgenic steroids (AAS) are synthetic derivatives of testosterone and are widely used by athletes to improve their physical performance. Observational studies have shown that the use of AAS is associated with adverse effects, including significant decrease in HDL-C levels and increase in LDL-C (6363 Fontana K, Oliveira HC, Leonardo MB, Mandarim-de-Lacerda CA, da Cruz-Höfling MA. Adverse effect of the anabolic-androgenic steroid mesterolone on cardiac remodelling and lipoprotein profile is attenuated by aerobicz exercise training. Int J Exp Pathol. 2008;89(5):358-66.). The mechanisms by which AAS affect HDL-C concentrations are not completely elucidated. Until now, it is known that AAS stimulate hepatic lipase enzyme activity in order to favor the catabolism of HDL and inhibit the biosynthesis of apoA-I (6464 Langfort J, Jagsz S, Dobrzyn P, Brzezinska Z, Klapcinska B, Galbo H, et al. Testosterone affects hormone-sensitive lipase (HSL) activity and lipid metabolism in the left ventricle. Biochem Biophys Res Commun. 2010;399(4):670-6.). In contrast to cross-sectional and prospective observational studies, one study showed hypogonadal patients, characterized by very low testosterone, with a lipoprotein profile within the normal range, suggesting a relationship with the absence of clearly defined obesity. However, there was a marked reduction in HDL cholesterol efflux capacity and an increase in serum cholesterol carrying capacity (6565 Adorni MP, Zimetti F, Cangiano B, Vezzoli V, Bernini F, Caruso D, et al. High-Density Lipoprotein Function Is Reduced in Patients Affected by Genetic or Idiopathic Hypogonadism. J Clin Endocrinol Metab. 2019;104(8):3097-107.). Also, there was a decrease in HDL-C concentrations during transgender hormone therapy. HDL cholesterol efflux capacity decreased during hormone therapy with specific reduction in ABCA1, contributing to an increased risk of CVD (6666 van Velzen DM, Adorni MP, Zimetti F, Strazzella A, Simsek S, Sirtori CR, et al. The effect of transgender hormonal treatment on high density lipoprotein cholesterol efflux capacity. Atherosclerosis. 2021;323:44-53). Cholesterol efflux capacity is a metric of HDL functionality that quantifies the ability of an individual’s HDL to extract cholesterol from macrophages. It has been shown to be a better predictor of atherosclerotic burden than HDL-C levels alone (6767 Khera AV, Demler OV, Adelman SJ, Collins HL, Glynn RJ, Ridker PM, et al. Cholesterol Efflux Capacity, High-Density Lipoprotein Particle Number, and Incident Cardiovascular Events: An Analysis From the JUPITER Trial (Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin). Circulation. 2017;135(25):2494-504.).

Infectious diseases

During infection, significant changes occur in lipid metabolism and lipoprotein composition. Lipoprotein concentrations rapidly change and can be reduced to 50% of recovery concentrations. Also, the levels of circulating HDL-C and LDL-C decrease, while the levels of TG and VLDL-C increase (6868 Miao Q, Ndao M. Trypanosoma cruzi infection and host lipid metabolism. Mediators Inflamm. 2014;2014:902038.), so it is thought that HDL-C may be a negative marker for systemic or local inflammations. More importantly, endotoxemia modulates HDL composition and size: phospholipids and apoA-I are reduced, while serum amyloid A (SAA) and secretory phospholipase A2 (sPLA2) increase dramatically. Although the number of total HDL does not change, a significant reduction is observed in the number of small and medium-sized particles (6969 de la Llera Moya M, McGillicuddy FC, Hinkle CC, Byrne M, Joshi MR, Nguyen V, et al. Inflammation modulates human HDL composition and function in vivo. Atherosclerosis. 2012;222(2):390-4.).

HDL can bind to and neutralize bacterial lipopolysaccharide gram-negative and gram-positive bacterial lipoteichoic acid, favoring the debugging of these products (7070 Parker TS, Levine DM, Chang JC, Laxer J, Coffin CC, Rubin AL. Reconstituted high-density lipoprotein neutralizes gram-negative bacterial lipopolysaccharides in human whole blood. Infect Immun. 1995;63(1):253-8.). Interestingly, HDLs also play a role in fighting infection parasites, and a specific component of HDL, the apolipoprotein L-1 (apoL-1), confers innate immunity against Trypanosoma cruzi by favoring the lysosomal swelling that kills the parasite (6868 Miao Q, Ndao M. Trypanosoma cruzi infection and host lipid metabolism. Mediators Inflamm. 2014;2014:902038.). At the same time that HDL-C undergoes a reduction in infectious diseases, low concentrations of HDL-C are also associated with an increased risk of acquiring infections, and prospective cohort studies observed a U-shaped relationship between HDL levels and risk of infectious disease (7171 Madsen CM, Varbo A, Tybjærg-Hansen A, Frikke-Schmidt R, Nordestgaard BG. U-shaped relationship of HDL and risk of infectious disease: two prospective population-based cohort studies. Eur Heart J. 2018;39(14):1181-90.). In addition, a Mendelian randomization study demonstrated that, genetically, certain HDL-C levels have a significant influence on risk of hospitalization for infectious diseases (7272 Trinder M, Walley KR, Boyd JH, Brunham LR. Causal Inference for Genetically Determined Levels of High-Density Lipoprotein Cholesterol and Risk of Infectious Disease. Arterioscler Thromb Vasc Biol. 2020;40(1):267-78.).

Therapeutic approaches

Mortality from CVD remains the leading cause of death in industrialized countries, and although the use of lipid-lowering therapies focused on LDL-C reduction has made a remarkable contribution to its control, there is still a substantial residual risk of CVD mortality. The observation of this residual risk has driven the search for new therapeutic targets, including conventional and new pharmacological therapies that are still under development. The great challenge lies in improving the functionality of the HDL system to favor the antiatherothrombotic effect. In the following paragraphs, we will briefly comment on some tested or researched therapies for this purpose. A table summarizing the main clinical trials and their outcomes is found at the end of this review (Table 1).

Table 1
Major clinical trials conducted involving therapies to increase plasma HD levels

Statins

Statin therapy has been shown to increase the level of plasma HDL-C and the mechanism most likely involves reduced transfer of cholesteryl ester from HDL to VLDL but other factors such as hepatic lipase and other statin-induced effects may also contribute (7373 McTaggart F, Jones P. Effects of statins on high-density lipoproteins: a potential contribution to cardiovascular benefit. Cardiovasc Drugs Ther. 2008;22(4):321-38.). Due to the inhibition in HMG-CoA reductase and suppression of Rho activity, statins can stimulate the synthesis of apoA-I in a dose-dependent manner and increase ABCA1 expression (7474 Maejima T, Yamazaki H, Aoki T, Tamaki T, Sato F, Kitahara M, et al. Effect of pitavastatin on apolipoprotein A-I production in HepG2 cell. Biochem Biophys Res Commun. 2004;324(2):835-9.,7575 Zanotti I, Favari E, Sposito AC, Rothblat GH, Bernini F. Pitavastatin increases ABCA1-mediated lipid efflux from Fu5AH rat hepatoma cells. Biochem Biophys Res Commun. 2004;321(3):670-4.). In parallel, statins inhibit CETP synthesis and bioavailability of lipoproteins rich in TG, which may contribute to the increase of HDL-C (7676 Sposito AC, Carmo HR, Barreto J, Sun L, Carvalho LSF, Feinstein SB, et al. HDL-Targeted Therapies During Myocardial Infarction. Cardiovasc Drugs Ther. 2019;33(3):371-81.).

Data from randomized clinical trials observed a two-fold increase in HDL-C levels compared to apoA-I levels (7777 Jones PH, Davidson MH, Stein EA, Bays HE, McKenney JM, Miller E, et al. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial). Am J Cardiol. 2003;92(2):152-60.). Furthermore, the percentage increases in HDL-C levels were greater in those individuals who had lower baseline plasma HDL-C levels (7878 Barter PJ, Brandrup-Wognsen G, Palmer MK, Nicholls SJ. Effect of statins on HDL-C: a complex process unrelated to changes in LDL-C: analysis of the VOYAGER Database. J Lipid Res. 2010;51(6):1546-53.).

These data show that statins alter HDL to a more cholesterol-rich form, characteristic of healthy populations of low cardiovascular risk (7878 Barter PJ, Brandrup-Wognsen G, Palmer MK, Nicholls SJ. Effect of statins on HDL-C: a complex process unrelated to changes in LDL-C: analysis of the VOYAGER Database. J Lipid Res. 2010;51(6):1546-53.). According to the STELLAR clinical study, among a group of five statins, the ability to increase HDL-C was greatest with rosuvastatin (9.2%), followed by simvastatin (6.8%), atorvastatin (5.7%) and, finally, pravastatin (5.6%) (7777 Jones PH, Davidson MH, Stein EA, Bays HE, McKenney JM, Miller E, et al. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial). Am J Cardiol. 2003;92(2):152-60.). Data from large studies of morbidity and mortality suggested that the effects induced by statins in HDL-C and apoA-I were sustained over time, offering an interesting advantage in relation to HDL infusion therapies, for example, whose effect is short-term (7979 Kingwell BA, Chapman MJ. Future of high-density lipoprotein infusion therapies: potential for clinical management of vascular disease. Circulation. 2013;128(10):1112-21.). The clinical impact of increased HDL-C by statin treatment is independent of its effects on LDL-C levels, TG-rich lipoproteins, or other side effects.

CETP inhibitors

CETP inhibitors were developed with the aim of blocking or interfering with CETP activity. They can be characterized into CETP inhibitors (torcetrapib, anacetrapib and evacetrapib) and CETP modulators (dalcetrapib) according to their chemical structure (8080 Ferri N, Corsini A, Sirtori CR, Ruscica M. Present therapeutic role of cholesteryl ester transfer protein inhibitors. Pharmacol Res. 2018;128:29-41.). The main function of CETP is the transfer of cholesteryl esters and TG between plasma lipoprotein particles. CETP inhibitors target a part of this mechanism by blocking the transfer of cholesteryl esters (8181 Nurmohamed NS, Ditmarsch M, Kastelein JJP. CETP-inhibitors: from HDL-C to LDL-C lowering agents? Cardiovasc Res. 2021:cvab350.).

CETP inhibitors increase HDL-C levels significantly (40% to 160%) due to the decrease in the transfer of cholesterol from HDL particles to rich lipoproteins in TG (8282 Tall AR, Rader DJ. Trials and Tribulations of CETP Inhibitors. Circ Res. 2018;122(1):106-12.). However, three clinical trials failed to demonstrate any cardiovascular benefits with treatment with these inhibitors (8383 Moreno C, Greil R, Demirkan F, Tedeschi A, Anz B, Larratt L, et al. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20(1):43-56.

84 Schwartz GG, Olsson AG, Abt M, Ballantyne CM, Barter PJ, Brumm J, et al. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N Engl J Med. 2012;367(22):2089-99.
-8585 Lincoff AM, Nicholls SJ, Riesmeyer JS, Barter PJ, Brewer HB, Fox KAA, et al. Evacetrapib and Cardiovascular Outcomes in High-Risk Vascular Disease. N Engl J Med. 2017;376(20):1933-42.). The REVEAL study (8686 HPS3/TIMI55–REVEAL Collaborative Group, Bowman L, Hopewell JC, Chen F, Wallendszus K, Stevens W, Collins R, et al. Effects of Anacetrapib in Patients with Atherosclerotic Vascular Disease. N Engl J Med. 2017;377(13):1217-27.) showed that cardiovascular events occurred less frequently in patients with atherosclerotic vascular disease treated with anacetrapib after 4.1 years of follow-up. In this last trial there was an 18% reduction in non-HDL cholesterol and a 104% increase in HDL-C with anacetrapib. A new CETP inhibitor, obicetrapib, reduced LDL-C and apoB levels by 45.3% and 33.7%, respectively. While HDL-C levels increased by 179.1% and ApoA-I by up to 63.4% (8787 Hovingh GK, Kastelein JJ, van Deventer SJ, Round P, Ford J, Saleheen D, et al. Cholesterol ester transfer protein inhibition by TA-8995 in patients with mild dyslipidaemia (TULIP): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet. 2015;386(9992):452-60.).

A Mendelian 2 × 2 factorial randomization study including 425,354 UK Biobank participants showed an additive association of a genetically reduced combined concentration of CETP and PCSK9 for lipid levels and risk of CAD. Suggesting that the joint inhibition of CETP and PCSK9 has additive effects on lipid concentrations (8888 Cupido AJ, Reeskamp LF, Hingorani AD, Finan C, Asselbergs FW, Hovingh GK, et al. Joint Genetic Inhibition of PCSK9 and CETP and the Association With Coronary Artery Disease: A Factorial Mendelian Randomization Study. JAMA Cardiol. 2022;7(9):955-64.). However, the double-blind dal-GenE study in patients with acute coronary syndrome of 1-3 months and the AA genotype in the rs1967309 variant in the ADCY9 gene showed that dalcetrapib did not significantly reduce the risk of ischemic cardiovascular events at the end of the study (8989 Tardif JC, Pfeffer MA, Kouz S, Koenig W, Maggioni AP, McMurray JJV, et al. Pharmacogenetics-guided dalcetrapib therapy after an acute coronary syndrome: the dal-GenE trial. Eur Heart J. 2022;43(39):3947-56.). A prospective, multicenter, cohort study verified the association between very high HDL-C levels and mortality in patients with CAD and the association of HDL-C genotypes with high HDL-C outcomes. The authors found a U-shaped association with higher risk in those with low and high levels of HDL-C compared with those with medium values (9090 Liu C, Dhindsa D, Almuwaqqat Z, Ko YA, Mehta A, Alkhoder AA, et al. Association Between High-Density Lipoprotein Cholesterol Levels and Adverse Cardiovascular Outcomes in High-risk Populations. JAMA Cardiol. 2022;7(7):672-80.). Paradoxically, the study suggests that very high levels of HDL-C are associated with a higher risk of mortality in individuals with CAD.

Fibrates

Fibrates are agonist drugs at peroxisome proliferator-activated receptor-α (PPAR-α) nuclear receptors, responsible for regulating the transcription of the LPL, ApoC-III and ApoA-I genes, leading to the clearance of TG-rich lipoproteins (9191 Chapman MJ. Fibrates in 2003: therapeutic action in atherogenic dyslipidaemia and future perspectives. Atherosclerosis. 2003;171(1):1-13.). Fibrates promote stimulation of HDL production by inducing hepatic synthesis of ApoA-I and ApoA-II and reducing VLDL production due to the reduction of free fatty acids in the liver, in addition to inhibiting the exchange of cholesterol and TG between HDL and VLDL (9292 Woudberg NJ, Pedretti S, Lecour S, Schulz R, Vuilleumier N, James RW, et al. Pharmacological Intervention to Modulate HDL: What Do We Target? Front Pharmacol. 2017;8:989.). By inducing elevation of HDL-C levels, reduction of TG-rich lipoproteins, and a shift in the phenotype from dense LDL to receptor-active, floating LDL, fibrates act to attenuate the atherosclerotic burden in atherogenic dyslipidemia (9191 Chapman MJ. Fibrates in 2003: therapeutic action in atherogenic dyslipidaemia and future perspectives. Atherosclerosis. 2003;171(1):1-13.).

A reduction in cardiovascular events was observed in patients with low HDL-C after treatment with gemfibrozil without the addition of statins (9393 Manninen V, Tenkanen L, Koskinen P, Huttunen JK, Mänttäri M, Heinonen OP, et al. Joint effects of serum triglyceride and LDL cholesterol and HDL cholesterol concentrations on coronary heart disease risk in the Helsinki Heart Study. Implications for treatment. Circulation. 1992;85(1):37-45.,9494 Robins SJ, Collins D, Wittes JT, Papademetriou V, Deedwania PC, Schaefer EJ, et al. Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial. JAMA. 2001;285(12):1585-91.). Although bezafibrate has been more effective than gemfibrozil in raising HDL-C levels, its use in secondary prevention has not proven to be beneficial in reducing cardiovascular events in patients with low HDL-C (9595 Bezafibrate Infarction Prevention (BIP) study. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease. Circulation. 2000;102(1):21-7.). In patients with T2DM, fenofibrate therapy also did not show cardiovascular benefits even in those with concomitant use of statins (9696 Elam M, Lovato L, Ginsberg H. The ACCORD-Lipid study: implications for treatment of dyslipidemia in Type 2 diabetes mellitus. Clin Lipidol. 2011;6(1):9-20.,9797 Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. 2005;366(9500):1849-61.). A new category of PPAR-α selective modulators that has been studied, pemafibrate, significantly reduces TG, apoC-III, and cholesterol remnants, in addition to increasing HDL-C. The PROMINENT study aims to evaluate the effect of pemafibrate on cardiovascular events in approximately 10,000 persons with diabetes with moderately elevated TG and low HDL-C (9898 Pradhan AD, Paynter NP, Everett BM, Glynn RJ, Amarenco P, Elam M, et al. Rationale and design of the Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) study. Am Heart J. 2018;206:80-93.). However, the Kowa Research Institute announces the decision not to continue the Phase 3 PROMINENT study. Based on a review of a planned interim analysis, they concluded that the primary endpoint would likely not be met. Full study data will be presented as soon as possible at a future conference.

Niacin

Through its GPR109A receptor (G-protein-coupled receptor) present in adipocytes, niacin acts by inhibiting the mobilization of free fatty acids, thus decreasing the supply of substrate for hepatic synthesis. It is possible that niacin increases HDL-C by its ability to inhibit hepatic lipase, decrease the catabolic rate of HDL-C, and decrease catabolic HDL-C receptors. These various effects result in higher levels and larger HDL-C molecules and more efficient reverse cholesterol transport (9999 Kamanna VS, Kashyap ML. Mechanism of action of niacin. Am J Cardiol. 2008;101(8A):20B-26B.). In hepatocytes, niacin inhibits the activity of diacylglycerol O-acyltransferase 2 (DGAT-2), decreasing the hepatic synthesis of TG and VLDL (100100 Song WL, FitzGerald GA. Niacin, an old drug with a new twist. J Lipid Res. 2013;54(10):2586-94.), in addition to promoting selective inhibition of ApoA-I uptake. Among the therapies that increase the serum concentration of HDL-C, niacin has the greatest effect, with an increase of 15 to 40% (101101 McKenney JM, Proctor JD, Harris S, Chinchili VM. A comparison of the efficacy and toxic effects of sustained- vs immediate-release niacin in hypercholesterolemic patients. JAMA. 1994;271(9):672-7.), preferentially raising the HDL2 subfraction. A meta-analysis showed that niacin was able to increase HDL-C levels by 23%, in addition to reducing TG levels by 40% and LDL-C levels by 20% (102102 Birjmohun RS, Hutten BA, Kastelein JJ, Stroes ES. Efficacy and safety of high-density lipoprotein cholesterol-increasing compounds: a meta-analysis of randomized controlled trials. J Am Coll Cardiol. 2005;45(2):185-97.). Despite being very effective in increasing HDL-C there is no evidence that the addition of niacin to therapy with statins results in cardiovascular benefit. Randomized clinical trials tested this hypothesis and failed to demonstrate any benefit from adding niacin to therapy with statins (103103 AIM-HIGH Investigators, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365(24):2255-67.,104104 HPS2-THRIVE Collaborative Group. HPS2-THRIVE randomized placebo-controlled trial in 25 673 high-risk patients of ER niacin/laropiprant: trial design, pre-specified muscle and liver outcomes, and reasons for stopping study treatment. Eur Heart J. 2013;34(17):1279-91.).

Therapies based on recombinant HDL

HDL infusion therapies induce a rapid, dose-proportional, time-dependent elevation in apoA-I and pre-βHDL particles. These effects were seen for recombinant HDL (CSL-111 and CSL-112), where plasma concentrations of apoA-I increased 2-fold and pre-βHDL increased >30-fold (7979 Kingwell BA, Chapman MJ. Future of high-density lipoprotein infusion therapies: potential for clinical management of vascular disease. Circulation. 2013;128(10):1112-21.,105105 Patel S, Drew BG, Nakhla S, Duffy SJ, Murphy AJ, Barter PJ, et al. Reconstituted high-density lipoprotein increases plasma high-density lipoprotein anti-inflammatory properties and cholesterol efflux capacity in patients with type 2 diabetes. J Am Coll Cardiol. 2009;53(11):962-71.). Despite this, the ERASE trial (106106 Tardif JC, Grégoire J, L’Allier PL, Ibrahim R, Lespérance J, Heinonen TM, et al. Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial. JAMA. 2007;297(15):1675-82.) showed that CSL-111 infusions did not result in significant reductions in atheroma volume or nominal change in plaque volume but did significantly impact the plaque characterization index on intravascular ultrasound. The AEGISII Phase 3 Study (expected completion 2022) (NCT03473223) assesses the efficacy and safety of CSL-112 in reducing larger cardiovascular events and will provide more information about the effect of this treatment in the progression of CVD.

The CER-001, an engineered negatively charged lipoprotein particle that contains recombinant human apoA-I (107107 Kalayci A, Gibson CM, Ridker PM, Wright SD, Kingwell BA, Korjian S, et al. ApoA-I Infusion Therapies Following Acute Coronary Syndrome: Past, Present, and Future. Curr Atheroscler Rep. 2022;24(7):585-97.), induced dose-dependent increases in plasma HDL-C of up to 7-fold (108108 Kee P, Rye KA, Taylor JL, Barrett PH, Barter PJ. Metabolism of apoA-I as lipid-free protein or as component of discoidal and spherical reconstituted HDLs: studies in wild-type and hepatic lipase transgenic rabbits. Arterioscler Thromb Vasc Biol. 2002;22(11):1912-7.). In the MODE study (109109 Hovingh GK, Smits LP, Stefanutti C, Soran H, Kwok S, de Graaf 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: The Modifying Orphan Disease Evaluation (MODE) study. Am Heart J. 2015;169(5):736-42.e1.), CER-001 was administered in serial infusions in 23 patients with homozygous familial hypercholesterolemia suggesting reduced mean carotid wall thickness. In the CHI-SQUARE study (110110 Tardif JC, Ballantyne CM, Barter P, Dasseux JL, Fayad ZA, Guertin MC, et al. Effects of the high-density lipoprotein mimetic agent CER-001 on coronary atherosclerosis in patients with acute coronary syndromes: a randomized trial. Eur Heart J. 2014;35(46):3277-86.), however, its administration did not reduce the coronary atherosclerotic burden. Interestingly, CER-001 appears to have a therapeutic target in LCAT deficiency, as shown in LCAT-/- mice treated with CER-001, there was improvement in dyslipidemia and renal function (111111 Ossoli A, Strazzella A, Rottoli D, Zanchi C, Locatelli M, Zoja C, et al. CER-001 ameliorates lipid profile and kidney disease in a mouse model of familial LCAT deficiency. Metabolism. 2021;116:154464.). They also verified the ability of CER-001 to remove cholesterol, reducing the deposition burden on the kidney in a patient with FLD. The beneficial effect is mediated by a dual action of CER-001, which directly effuses cholesterol from podocytes, but also induces the normalization of plasma lipoproteins (112112 Pavanello C, Turri M, Strazzella A, Tulissi P, Pizzolitto S, De Maglio G, et al. The HDL mimetic CER-001 remodels plasma lipoproteins and reduces kidney lipid deposits in inherited lecithin:cholesterol acyltransferase deficiency. J Intern Med. 2022;291(3):364-70.). This beneficial effect occurs earlier than predictable and may offer a new form of treatment for this serious condition (113113 Faguer S, Colombat M, Chauveau D, Bernadet-Monrozies P, Beq A, Delas A, et al. Administration of the High-Density Lipoprotein Mimetic CER-001 for Inherited Lecithin-Cholesterol Acyltransferase Deficiency. Ann Intern Med. 2021;174(7):1022-5.).

In a phase 2 clinical study, administration of ETC-216, recombinant HDL with apoA-I Milano, produced significant regression in atherosclerotic plaque volume in subjects with acute coronary syndrome, measured by intravascular ultrasound (114114 Nissen SE, Tsunoda T, Tuzcu EM, Schoenhagen P, Cooper CJ, Yasin M, et al. Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. JAMA. 2003;290(17):2292-300.) suggesting that infusion in the first weeks after an acute event would stimulate the reverse cholesterol transport. However, serious side effects prevented further evaluation of this formulation and development of this formulation stopped. MDCO-216, tested in a multicenter trial, showed that after five weekly infusions, the percentage volume of the atheroma in the diseased segment did not differ significantly between patients with coronary heart disease documented by angiogram and the control group, ending its development (115115 Reijers JAA, Kallend DG, Malone KE, Jukema JW, Wijngaard PLJ, Burggraaf J, et al. MDCO-216 Does Not Induce Adverse Immunostimulation, in Contrast to Its Predecessor ETC-216. Cardiovasc Drugs Ther. 2017;31(4):381-9.).

Recombinant LCAT (rhLCAT)

Most data from preclinical studies suggest that increasing the amount of LCAT stimulates reverse cholesterol transport and reduces atherosclerosis. These data are based on studies that observed a reduction in the progression of atherosclerosis in mice with increased expression of LCAT (116116 Mertens A, Verhamme P, Bielicki JK, Phillips MC, Quarck R, Verreth W, et al. Increased low-density lipoprotein oxidation and impaired high-density lipoprotein antioxidant defense are associated with increased macrophage homing and atherosclerosis in dyslipidemic obese mice: LCAT gene transfer decreases atherosclerosis. Circulation. 2003;107(12):1640-6.), in addition to the increase in atherosclerotic lesions in mice with enzyme deficiency (117117 Furbee JW, Sawyer JK, Parks JS. Lecithin:cholesterol acyltransferase deficiency increases atherosclerosis in the low density lipoprotein receptor and apolipoprotein E knockout mice. J Biol Chem. 2002;277(5):3511-9.). In this context, a phase I clinical trial demonstrated that a single infusion injection of rhLCAT (ACP-501) had an acceptable safety profile and favorably altered HDL metabolism, showing a dose-proportional increase in HDL-C, thus supporting the investigation of this therapy in individuals with atherosclerotic disease and LCAT deficiency (118118 Shamburek RD, Bakker-Arkema R, Shamburek AM, Freeman LA, Amar MJ, Auerbach B, et al. Safety and Tolerability of ACP-501, a Recombinant Human Lecithin:Cholesterol Acyltransferase, in a Phase 1 Single-Dose Escalation Study. Circ Res. 2016;118(1):73-82.). Next, in the FLD patient study, intravenous drip administration of ACP-501 improved the abnormal distribution of HDL subfractions (119119 Shamburek RD, Bakker-Arkema R, Auerbach BJ, Krause BR, Homan R, Amar MJ, et al. Familial lecithin:cholesterol acyltransferase deficiency: First-in-human treatment with enzyme replacement. J Clin Lipidol. 2016;10(2):356-67.). MEDI6012 is a more active rhLCAT that resulted in increased HDL function in a dose-related manner and also promoted endothelial protection (120120 Ossoli A, Simonelli S, Varrenti M, Morici N, Oliva F, Stucchi M, et al. Recombinant LCAT (Lecithin:Cholesterol Acyltransferase) Rescues Defective HDL (High-Density Lipoprotein)-Mediated Endothelial Protection in Acute Coronary Syndrome. Arterioscler Thromb Vasc Biol. 2019;39(5):915-24.). Furthermore, LCAT overexpression improved LDL receptor-mediated reverse cholesterol transport, leading to regression of atherosclerosis, suggesting synergistic effects of MEDI6012 with statins (121121 George RT, Abuhatzira L, Stoughton SM, Karathanasis SK, She D, Jin C, et al. MEDI6012: Recombinant Human Lecithin Cholesterol Acyltransferase, High-Density Lipoprotein, and Low-Density Lipoprotein Receptor-Mediated Reverse Cholesterol Transport. J Am Heart Assoc. 2021;10(13):e014572.).

ApoA-I mimetic peptides

ApoA-I is the main apolipoprotein present in HDL particles. Although it is composed of 243 amino acids arranged in 10 amphipathic helices, there has been interest in using short apoA-I peptides containing at least one amphipathic helix to mimic the functions of native apoA-I. The first apoA-I mimetic peptide was synthesized comprising 18 amino acids (18A). Subsequently, several modifications to 18A were made to create peptides that more closely mimic apoA-I in its antiatherogenic functions (122122 Anantharamaiah GM, Jones JL, Brouillette CG, Schmidt CF, Chung BH, Hughes TA, et al. Studies of synthetic peptide analogs of the amphipathic helix. Structure of complexes with dimyristoyl phosphatidylcholine. J Biol Chem. 1985;260(18):10248-55.). Then the addition of an acetyl group and an amide group promoted stability and the lipid binding properties were improved, the peptide was named 2F. Although the 2F peptide mimicked many of the lipid-binding properties of apoA-I, it failed to alter the lesions in a mouse model of atherosclerosis (123123 Datta G, Chaddha M, Hama S, Navab M, Fogelman AM, Garber DW, et al. Effects of increasing hydrophobicity on the physical-chemical and biological properties of a class A amphipathic helical peptide. J Lipid Res. 2001;42(7):1096-104.). Subsequent, the administration of peptide 5F by injection significantly inhibited lesion formation in mice fed an atherogenic diet without significantly altering lipoprotein profiles (124124 Garber DW, Datta G, Chaddha M, algunachari MN, Hama SY, Navab M, et al. A new synthetic class A amphipathic peptide analogue protects mice from diet-induced atherosclerosis. J Lipid Res. 2001;42(4):545-52.). Another apoA-I mimetic peptide synthesized from D-amino acids (D-4F) to LDL receptor null mice on a Western diet made HDL anti-inflammatory and reduced lesions by 79% without altering HDL-C levels (125125 Navab M, Anantharamaiah GM, Hama S, Garber DW, Chaddha M, Hough G, et al. Oral administration of an Apo A-I mimetic Peptide synthesized from D-amino acids dramatically reduces atherosclerosis in mice independent of plasma cholesterol. Circulation. 2002;105(3):290-2.). Recently, P12 showed efficient binding with cytomembrane phospholipids, cholesterol and HDL, and showed the ability to reverse cholesterol transport and treat atherosclerosis (126126 Gou S, Wang L, Zhong C, Chen X, Ouyang X, Li B, et al. A novel apoA-I mimetic peptide suppresses atherosclerosis by promoting physiological HDL function in apoE. Br J Pharmacol. 2020;177(20):4627-444.). With its phospholipid affinity, P12 facilitated cholesterol transport through the ABCA1. Also, promoted the function of HDL by remodeling α‐HDL into preβ‐HDL which can more efficiently transport cholesterol in the atherosclerotic plaques to the liver and remove excess cholesterol from the arteries (126126 Gou S, Wang L, Zhong C, Chen X, Ouyang X, Li B, et al. A novel apoA-I mimetic peptide suppresses atherosclerosis by promoting physiological HDL function in apoE. Br J Pharmacol. 2020;177(20):4627-444.). The use of peptides that mimic HDL-associated apolipoproteins may prove to be a successful strategy in the treatment of vascular diseases.

ApoA-I transcriptional supraregulators

RVX-208 is a therapeutic agent that selectively induces hepatic de novo synthesis of apoA-I and thereby increases HDL levels accompanied by a change in the pattern of HDL distribution. In monkeys, it primarily increased the levels of pre-β1 and α-1 HDL particles, representing the lipid-poor cholesterol efflux receptors and the larger lipid-rich HDL particles, respectively (127127 Bailey D, Jahagirdar R, Gordon A, Hafiane A, Campbell S, Chatur S, et al. RVX-208: a small molecule that increases apolipoprotein A-I and high-density lipoprotein cholesterol in vitro and in vivo. J Am Coll Cardiol. 2010;55(23):2580-9.). Treatment with RVX-208 may cause, through de novo synthesis of apoA-I and/or decrease in LCAT activity, favorable changes in HDL distribution. In humans, a 42% and 11% increase in pre-β1 HDL levels and ABCA1-mediated cholesterol efflux, respectively, occurred after only 7 days of RVX-208 therapy (127127 Bailey D, Jahagirdar R, Gordon A, Hafiane A, Campbell S, Chatur S, et al. RVX-208: a small molecule that increases apolipoprotein A-I and high-density lipoprotein cholesterol in vitro and in vivo. J Am Coll Cardiol. 2010;55(23):2580-9.). However, the ASSURE study showed modest increases in apoA-I and HDL-C with RVX-208, these changes did not differ from the placebo group. These data demonstrate that treatment with RVX-208 did not result in any measurable incremental benefit in plaque regression for patients with CAD and low HDL-C levels (128128 Nicholls SJ, Puri R, Wolski K, Ballantyne CM, Barter PJ, Brewer HB, et al. Effect of the BET Protein Inhibitor, RVX-208, on Progression of Coronary Atherosclerosis: Results of the Phase 2b, Randomized, Double-Blind, Multicenter, ASSURE Trial. Am J Cardiovasc Drugs. 2016;16(1):55-65.).

microRNA antagonists

Studies have identified miR-33a/b as essential regulators of lipid metabolism, regulating HDL-C levels and the multi-step reverse cholesterol transport process, therefore, the therapeutic potential of miR-33 to treat CVD is promising. The antagonism of miR-33 significantly increased plasma levels of HDL-C directed macrophage polarization to an M2-like phenotype and improved atherosclerosis regression (129129 Rayner KJ, Sheedy FJ, Esau CC, Hussain FN, Temel RE, Parathath S, et al. Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J Clin Invest. 2011;121(7):2921-31.). However, two independent groups showed a beneficial effect of miR-33 inhibition in attenuating atherosclerosis progression (130130 Rotllan N, Ramírez CM, Aryal B, Esau CC, Fernández-Hernando C. Therapeutic silencing of microRNA-33 inhibits the progression of atherosclerosis in Ldlr-/- mice--brief report. Arterioscler Thromb Vasc Biol. 2013;33(8):1973-7.,131131 Ouimet M, Ediriweera HN, Gundra UM, Sheedy FJ, Ramkhelawon B, Hutchison SB, et al. MicroRNA-33-dependent regulation of macrophage metabolism directs immune cell polarization in atherosclerosis. J Clin Invest. 2015;125(12):4334-48.), while another group did not observe any improvement in atherogenesis under similar conditions (132132 Marquart TJ, Wu J, Lusis AJ, Baldán Á. Anti-miR-33 therapy does not alter the progression of atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2013;33(3):455-8.). Interestingly, all three studies demonstrated that miR-33 antagonism in hypercholesterolemic mice does not increase plasma levels of HDL-C. These findings suggest that miR-33 inhibition may promote atheroprotection through mechanisms independent of the increase in circulating HDL-C.

Antisense oligonucleotides for CETP and ApoC-III

One study compared an antisense oligonucleotides (ASO) inhibitor of CETP with the inhibitor of anacetrapib in hyperlipidemic mice and both therapies resulted in a decrease in total plasma cholesterol, decrease in CETP activity and increase in levels of HDL-C (133133 Bell TA, Graham MJ, Lee RG, Mullick AE, Fu W, Norris D, et al. Antisense oligonucleotide inhibition of cholesteryl ester transfer protein enhances RCT in hyperlipidemic, CETP transgenic, LDLr-/- mice. J Lipid Res. 2013;54(10):2647-57.). ASO against ApoC-III decreases its production and, consequently, lipolysis enhancement of TG-rich lipoproteins results in an increase in HDL-C levels and a decrease in the levels of TG (134134 Schmitz J, Gouni-Berthold I. APOC-III Antisense Oligonucleotides: A New Option for the Treatment of Hypertriglyceridemia. Curr Med Chem. 2018;25(13):1567-76.). These findings suggest that ASOs from CETP and ApoC-III may represent a promising therapeutic alternative.

Endothelial lipase inhibitors

Endothelial lipase (EL) knockout studies (135135 Ishida T, Choi S, Kundu RK, Hirata K, Rubin EM, Cooper AD, et al. Endothelial lipase is a major determinant of HDL level. J Clin Invest. 2003;111(3):347-55.,136136 Ishida T, Choi SY, Kundu RK, Spin J, Yamashita T, Hirata K, et al. Endothelial lipase modulates susceptibility to atherosclerosis in apolipoprotein-E-deficient mice. J Biol Chem. 2004;279(43):45085-92.) and anti-EL antibodies (137137 Jin W, Millar JS, Broedl U, Glick JM, Rader DJ. Inhibition of endothelial lipase causes increased HDL cholesterol levels in vivo. J Clin Invest. 2003;111(3):357-62.) in mice demonstrated that inhibiting EL function resulted in increased plasma HDL-C levels. Furthermore, the overexpression of the human EL gene in mouse liver significantly reduced levels of circulating HDL-C and apoA-I (138138 Goodman KB, Bury MJ, Cheung M, Cichy-Knight MA, Dowdell SE, Dunn AK, et al. Discovery of potent, selective sulfonylfuran urea endothelial lipase inhibitors. Bioorg Med Chem Lett. 2009;19(1):27-30.). Loss-of-function mutations in EL expression have been observed to lead to increased levels of HDL-C and support the idea that inhibition of endothelial lipase may be an effective mechanism to increase HDL-C (139139 Edmondson AC, Brown RJ, Kathiresan S, Cupples LA, Demissie S, Manning AK, et al. Loss-of-function variants in endothelial lipase are a cause of elevated HDL cholesterol in humans. J Clin Invest. 2009;119(4):1042-50.). In contrast, some studies point to an atherogenic role of EL, with a positive association between plasma levels of this enzyme and coronary artery calcification and inflammation (140140 Badellino KO, Wolfe ML, Reilly MP, Rader DJ. Endothelial lipase concentrations are increased in metabolic syndrome and associated with coronary atherosclerosis. PLoS Med. 2006;3(2):e22.,141141 Badellino KO, Wolfe ML, Reilly MP, Rader DJ. Endothelial lipase is increased in vivo by inflammation in humans. Circulation. 2008;117(5):678-85.).

Liver X receptors (LXR) agonists

By coordinating the expression of target genes in various tissues, LXR agonists increase the flow of cholesterol from the periphery to the liver, where it is metabolized and excreted (142142 Zelcer N, Tontonoz P. Liver X receptors as integrators of metabolic and inflammatory signaling. J Clin Invest. 2006;116(3):607-14.). Short-term administration of synthetic LXR agonist T0901317 in mice promoted reverse cholesterol transport in vivo from macrophages, at least in part, inducing an enrichment of HDL subclasses that enhance the plasma’s ability to promote cholesterol efflux by passive diffusion. and mechanisms mediated by SR-BI (143143 Zanotti I, Potì F, Pedrelli M, Favari E, Moleri E, Franceschini G, et al. The LXR agonist T0901317 promotes the reverse cholesterol transport from macrophages by increasing plasma efflux potential. J Lipid Res. 2008;49(5):954-60.). In healthy subjects and hypercholesterolemic patients, reverse cholesterol transport pathways were similarly induced as in animal models by BMS-852927. However, an increase in plasma and liver TG, plasma LDL-C, apoB, apoE and CETP were also evident (144144 Kirchgessner TG, Sleph P, Ostrowski J, Lupisella J, Ryan CS, Liu X, et al. Beneficial and Adverse Effects of an LXR Agonist on Human Lipid and Lipoprotein Metabolism and Circulating Neutrophils. Cell Metab. 2016;24(2):223-33.). In addition, there are potent synthetic LXR agonists, including GW3965, LXR-623, GW6340, AZ876 and ATI-111. LXRs play a central role in the regulation of ABCA1 expression in macrophages (145145 Laffitte BA, Repa JJ, Joseph SB, Wilpitz DC, Kast HR, Mangelsdorf DJ, et al. LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proc Natl Acad Sci U S A. 2001;98(2):507-12.), and also induce expression of ABCG1 (146146 Baldán A, Bojanic DD, Edwards PA. The ABCs of sterol transport. J Lipid Res. 2009;50 Suppl:S80-5.), thus increasing the formation of HDL mediated by these transporters.

In conclusion, despite evidence from observational studies, there are no indications to use HDL-C as a risk marker or even to consider increased plasma HDL-C as a therapeutic means to prevent CAD. In patients with very low HDL-C, diagnosis and intervention is important to exclude secondary causes and identify single-gene causes in anticipation of potential risks. Therapeutic approaches targeting HDL such as niacin, fibrates, and CETP inhibitors increased plasma levels of HDL-C but failed to reduce cardiovascular events. Therefore, we can say that it is insufficient to measure plasma levels of HDL-C as a way of estimating its atheroprotective function. Still, ways to increase cardiovascular protection with therapies aimed at HDL metabolism are not well established. Thus, further studies are needed to understand the mechanisms of molecular action and cellular interaction of HDL, enhancing the function of the HDL system.

REFERENCES

  • 1
    Castelli WP, Doyle JT, Gordon T, Hames CG, Hjortland MC, Hulley SB, et al. HDL cholesterol and other lipids in coronary heart disease. The cooperative lipoprotein phenotyping study. Circulation. 1977;55(5):767-72.
  • 2
    Mora S, Glynn RJ, Ridker PM. High-density lipoprotein cholesterol, size, particle number, and residual vascular risk after potent statin therapy. Circulation. 2013;128(11):1189-97.
  • 3
    Assmann G, Schulte H, von Eckardstein A, Huang Y. High-density lipoprotein cholesterol as a predictor of coronary heart disease risk. The PROCAM experience and pathophysiological implications for reverse cholesterol transport. Atherosclerosis. 1996;124 Suppl:S11-20.
  • 4
    Ko DT, Alter DA, Guo H, Koh M, Lau G, Austin PC, et al. High-Density Lipoprotein Cholesterol and Cause-Specific Mortality in Individuals Without Previous Cardiovascular Conditions: The CANHEART Study. J Am Coll Cardiol. 2016;68(19):2073-83.
  • 5
    Vergeer M, Holleboom AG, Kastelein JJ, Kuivenhoven JA. The HDL hypothesis: does high-density lipoprotein protect from atherosclerosis? J Lipid Res. 2010;51(8):2058-73.
  • 6
    Frikke-Schmidt R, Nordestgaard BG, Stene MC, Sethi AA, Remaley AT, Schnohr P, et al. Association of loss-of-function mutations in the ABCA1 gene with high-density lipoprotein cholesterol levels and risk of ischemic heart disease. JAMA. 2008;299(21):2524-32.
  • 7
    Haase CL, Tybjærg-Hansen A, Qayyum AA, Schou J, Nordestgaard BG, Frikke-Schmidt R. LCAT, HDL cholesterol and ischemic cardiovascular disease: a Mendelian randomization study of HDL cholesterol in 54,500 individuals. J Clin Endocrinol Metab. 2012;97(2):E248-56.
  • 8
    Sposito AC. HDL metrics, let’s call the number thing off? Atherosclerosis. 2016;251:525-7.
  • 9
    Posadas-Sánchez R, Posadas-Romero C, Ocampo-Arcos WA, Villarreal-Molina MT, Vargas-Alarcón G, Antúnez-Argüelles E, et al. Premature and severe cardiovascular disease in a Mexican male with markedly low high-density-lipoprotein-cholesterol levels and a mutation in the lecithin: cholesterol acyltransferase gene: a family study. Int J Mol Med. 2014;33(6):1570-6.
  • 10
    Brunham LR, Hayden MR. Human genetics of HDL: Insight into particle metabolism and function. Prog Lipid Res. 2015;58:14-25.
  • 11
    Voight BF, Peloso GM, Orho-Melander M, Frikke-Schmidt R, Barbalic M, Jensen MK, et al. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet. 2012;380(9841):572-80.
  • 12
    Nesto RW. Beyond low-density lipoprotein: addressing the atherogenic lipid triad in type 2 diabetes mellitus and the metabolic syndrome. Am J Cardiovasc Drugs. 2005;5(6):379-87.
  • 13
    Rothblat GH, Phillips MC. High-density lipoprotein heterogeneity and function in reverse cholesterol transport. Curr Opin Lipidol. 2010;21(3):229-38.
  • 14
    Eckardstein VA, Kardassis D. High Density Lipoproteins From biological understanding to clinical exploitation. Springer Open; 2015.
  • 15
    Kontush A, Chapman MJ. Antiatherogenic function of HDL particle subpopulations: focus on antioxidative activities. Curr Opin Lipidol. 2010;21(4):312-8.
  • 16
    Prosser HC, Ng MKC, Bursill CA. The role of cholesterol efflux in mechanisms of endothelial protection by HDL. Curr Opin Lipidol. 2012;23(3):182-9.
  • 17
    Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13(4):423-33.
  • 18
    Beheshti SO, Madsen CM, Varbo A, Nordestgaard BG. Worldwide Prevalence of Familial Hypercholesterolemia: Meta-Analyses of 11 Million Subjects. J Am Coll Cardiol. 2020;75(20):2553-66.
  • 19
    Dron JS, Wang J, Low-Kam C, Khetarpal SA, Robinson JF, McIntyre AD, et al. Polygenic determinants in extremes of high-density lipoprotein cholesterol. J Lipid Res. 2017;58(11):2162-70.
  • 20
    von Eckardstein A. Differential diagnosis of familial high density lipoprotein deficiency syndromes. Atherosclerosis. 2006;186(2):231-9.
  • 21
    El Khoury P, Couvert P, Elbitar S, Ghaleb Y, Abou-Khalil Y, Azar Y, et al. Identification of the first Tangier disease patient in Lebanon carrying a new pathogenic variant in ABCA1. J Clin Lipidol. 2018;12(6):1374-82.
  • 22
    Maranghi M, Truglio G, Gallo A, Grieco E, Verrienti A, Montali A, et al. A novel splicing mutation in the ABCA1 gene, causing Tangier disease and familial HDL deficiency in a large family. Biochem Biophys Res Commun. 2019;508(2):487-93.
  • 23
    Quintão ECR, Nakandakare ER, Passarelli M. Lípides: do metabolismo à aterosclerose. 1ᵃ ed. São Paulo: Sarvier; 2011.
  • 24
    Ceccanti M, Cambieri C, Frasca V, Onesti E, Biasiotta A, Giordano C, et al. A Novel Mutation in ABCA1 Gene Causing Tangier Disease in an Italian Family with Uncommon Neurological Presentation. Front Neurol. 2016;7:185.
  • 25
    Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59(2):97-106.
  • 26
    von Eckardstein A, Huang Y, Kastelein JJ, Geisel J, Real JT, Kuivenhoven JA, et al. Lipid-free apolipoprotein (apo) A-I is converted into alpha-migrating high density lipoproteins by lipoprotein-depleted plasma of normolipidemic donors and apo A-I-deficient patients but not of Tangier disease patients. Atherosclerosis. 1998;138(1):25-34.
  • 27
    Alshaikhli A, Vaqar S. Tangier Disease. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2022 January.
  • 28
    Ossoli A, Lucca F, Boscutti G, Remaley AT, Calabresei L. Familial LCAT deficiency: from pathology to enzyme replacement therapy. Clin Lipidol. 2015;10:405-13.
  • 29
    Saeedi R, Li M, Frohlich J. A review on lecithin: cholesterol acyltransferase deficiency. Clin Biochem. 2015;48(7-8):472-5.
  • 30
    Ayyobi AF, McGladdery SH, Chan S, John Mancini GB, Hill JS, Frohlich JJ. Lecithin: cholesterol acyltransferase (LCAT) deficiency and risk of vascular disease: 25 year follow-up. Atherosclerosis. 2004;177(2):361-6.
  • 31
    Calabresi L, Baldassarre D, Castelnuovo S, Conca P, Bocchi L, Candini C, et al. Functional lecithin: cholesterol acyltransferase is not required for efficient atheroprotection in humans. Circulation. 2009;120(7):628-35
  • 32
    Kunnen S, Van Eck M. Lecithin: cholesterol acyltransferase: old friend or foe in atherosclerosis? J Lipid Res. 2012;53(9):1783-99.
  • 33
    Tanigawa H, Billheimer JT, Tohyama J, Fuki IV, Ng DS, Rothblat GH, et al. Lecithin: cholesterol acyltransferase expression has minimal effects on macrophage reverse cholesterol transport in vivo. Circulation. 2009;120(2):160-9.
  • 34
    Calabresi L, Favari E, Moleri E, Adorni MP, Pedrelli M, Costa S, et al. Functional LCAT is not required for macrophage cholesterol efflux to human serum. Atherosclerosis. 2009;204(1):141-6.
  • 35
    Kuivenhoven JA, Pritchard H, Hill J, Frohlich J, Assmann G, Kastelein J. The molecular pathology of lecithin: cholesterol acyltransferase (LCAT) deficiency syndromes. J Lipid Res. 1997;38(2):191-205.
  • 36
    Vitali C, Bajaj A, Nguyen C, Schnall J, Chen J, Stylianou K, et al. A systematic review of the natural history and biomarkers of primary Lecithin: Cholesterol Acyltransferase (LCAT) deficiency. J Lipid Res. 2022;63(3):100169.
  • 37
    Strøm EH, Sund S, Reier-Nilsen M, Dørje C, Leren TP. Lecithin: Cholesterol Acyltransferase (LCAT) Deficiency: renal lesions with early graft recurrence. Ultrastruct Pathol. 2011;35(3):139-45.
  • 38
    Suda T, Akamatsu A, Nakaya Y, Masuda Y, Desaki J. Alterations in erythrocyte membrane lipid and its fragility in a patient with familial lecithin: cholesterol acyltrasferase (LCAT) deficiency. J Med Invest. 2002;49(3-4):147-55.
  • 39
    Oldoni F, Baldassarre D, Castelnuovo S, Ossoli A, Amato M, van Capelleveen J, et al. Complete and Partial Lecithin:Cholesterol Acyltransferase Deficiency Is Differentially Associated With Atherosclerosis. Circulation. 2018;138(10):1000-7.
  • 40
    Fellin R, Manzato E. Lipoprotein-X fifty years after its original discovery. Nutr Metab Cardiovasc Dis. 2019;29(1):4-8.
  • 41
    Narayanan S. Lipoprotein-X. CRC Crit Rev Clin Lab Sci. 1979;11(1):31-51.
  • 42
    Hegele RA, Borén J, Ginsberg HN, Arca M, Averna M, Binder CJ, et al. Rare dyslipidaemias, from phenotype to genotype to management: a European Atherosclerosis Society task force consensus statement. Lancet Diabetes Endocrinol. 2020;8(1):50-67.
  • 43
    Schaefer EJ, Genest JJ, Ordovas JM, Salem DN, Wilson PW. Familial lipoprotein disorders and premature coronary artery disease. Atherosclerosis. 1994;108 Suppl:S41-54.
  • 44
    Pisciotta L, Miccoli R, Cantafora A, Calabresi L, Tarugi P, Alessandrini P, et al. Recurrent mutations of the apolipoprotein A-I gene in three kindreds with severe HDL deficiency. Atherosclerosis. 2003;167(2):335-45.
  • 45
    Ikewaki K, Matsunaga A, Han H, Watanabe H, Endo A, Tohyama J, et al. A novel two nucleotide deletion in the apolipoprotein A-I gene, apoA-I Shinbashi, associated with high density lipoprotein deficiency, corneal opacities, planar xanthomas, and premature coronary artery disease. Atherosclerosis. 2004;172(1):39-45.
  • 46
    Schaefer EJ, Santos RD, Asztalos BF. Marked HDL deficiency and premature coronary heart disease. Curr Opin Lipidol. 2010;21(4):289-97.
  • 47
    Andreola A, Bellotti V, Giorgetti S, Mangione P, Obici L, Stoppini M, et al. Conformational switching and fibrillogenesis in the amyloidogenic fragment of apolipoprotein a-I. J Biol Chem. 2003;278(4):2444-51.
  • 48
    Joy T, Wang J, Hahn A, Hegele RA. APOA1 related amyloidosis: a case report and literature review. Clin Biochem. 2003;36(8):641-5.
  • 49
    Obici L, Palladini G, Giorgetti S, Bellotti V, Gregorini G, Arbustini E, et al. Liver biopsy discloses a new apolipoprotein A-I hereditary amyloidosis in several unrelated Italian families. Gastroenterology. 2004;126(5):1416-22.
  • 50
    Miller M, Aiello D, Pritchard H, Friel G, Zeller K. Apolipoprotein A-I(Zavalla) (Leu159-->Pro): HDL cholesterol deficiency in a kindred associated with premature coronary artery disease. Arterioscler Thromb Vasc Biol. 1998;18(8):1242-7.
  • 51
    Hovingh GK, Brownlie A, Bisoendial RJ, Dube MP, Levels JH, Petersen W, et al. A novel apoA-I mutation (L178P) leads to endothelial dysfunction, increased arterial wall thickness, and premature coronary artery disease. J Am Coll Cardiol. 2004;44(7):1429-35.
  • 52
    Chiesa G, Sirtori CR. Apolipoprotein A-I(Milano): current perspectives. Curr Opin Lipidol. 2003;14(2):159-63.
  • 53
    von Eckardstein A, Funke H, Walter M, Altland K, Benninghoven A, Assmann G. Structural analysis of human apolipoprotein A-I variants. Amino acid substitutions are nonrandomly distributed throughout the apolipoprotein A-I primary structure. J Biol Chem. 1990;265(15):8610-7.
  • 54
    Rohatgi A, Khera A, Berry JD, Givens EG, Ayers CR, Wedin KE, et al. HDL cholesterol efflux capacity and incident cardiovascular events. N Engl J Med. 2014;371(25):2383-93.
  • 55
    Richardson TG, Sanderson E, Palmer TM, Ala-Korpela M, Ference BA, Davey Smith G, et al. Evaluating the relationship between circulating lipoprotein lipids and apolipoproteins with risk of coronary heart disease: A multivariable Mendelian randomisation analysis. PLoS Med. 2020;17(3):e1003062.
  • 56
    Karjalainen MK, Holmes MV, Wang Q, Anufrieva O, Kähönen M, Lehtimäki T, et al. Apolipoprotein A-I concentrations and risk of coronary artery disease: A Mendelian randomization study. Atherosclerosis. 2020;299:56-63.
  • 57
    Abbasi A, Corpeleijn E, Gansevoort RT, Gans RO, Hillege HL, Stolk RP, et al. Role of HDL cholesterol and estimates of HDL particle composition in future development of type 2 diabetes in the general population: the PREVEND study. J Clin Endocrinol Metab. 2013;98(8):E1352-9.
  • 58
    DeFronzo RA, Ferrannini E. Insulin resistance. A multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Care. 1991;14(3):173-94.
  • 59
    Rashid S, Watanabe T, Sakaue T, Lewis GF. Mechanisms of HDL lowering in insulin resistant, hypertriglyceridemic states: the combined effect of HDL triglyceride enrichment and elevated hepatic lipase activity. Clin Biochem. 2003;36(6):421-9.
  • 60
    Bonilha I, Zimetti F, Zanotti I, Papotti B, Sposito AC. Dysfunctional High-Density Lipoproteins in Type 2 Diabetes Mellitus: Molecular Mechanisms and Therapeutic Implications. J Clin Med. 2021;10(11).
  • 61
    Bonilha I, Hajduch E, Luchiari B, Nadruz W, Le Goff W, Sposito AC. The Reciprocal Relationship between LDL Metabolism and Type 2 Diabetes Mellitus. Metabolites. 2021;11(12).
  • 62
    Jakob T, Nordmann AJ, Schandelmaier S, Ferreira-González I, Briel M. Fibrates for primary prevention of cardiovascular disease events. Cochrane Database Syst Rev. 2016;11:CD009753.
  • 63
    Fontana K, Oliveira HC, Leonardo MB, Mandarim-de-Lacerda CA, da Cruz-Höfling MA. Adverse effect of the anabolic-androgenic steroid mesterolone on cardiac remodelling and lipoprotein profile is attenuated by aerobicz exercise training. Int J Exp Pathol. 2008;89(5):358-66.
  • 64
    Langfort J, Jagsz S, Dobrzyn P, Brzezinska Z, Klapcinska B, Galbo H, et al. Testosterone affects hormone-sensitive lipase (HSL) activity and lipid metabolism in the left ventricle. Biochem Biophys Res Commun. 2010;399(4):670-6.
  • 65
    Adorni MP, Zimetti F, Cangiano B, Vezzoli V, Bernini F, Caruso D, et al. High-Density Lipoprotein Function Is Reduced in Patients Affected by Genetic or Idiopathic Hypogonadism. J Clin Endocrinol Metab. 2019;104(8):3097-107.
  • 66
    van Velzen DM, Adorni MP, Zimetti F, Strazzella A, Simsek S, Sirtori CR, et al. The effect of transgender hormonal treatment on high density lipoprotein cholesterol efflux capacity. Atherosclerosis. 2021;323:44-53
  • 67
    Khera AV, Demler OV, Adelman SJ, Collins HL, Glynn RJ, Ridker PM, et al. Cholesterol Efflux Capacity, High-Density Lipoprotein Particle Number, and Incident Cardiovascular Events: An Analysis From the JUPITER Trial (Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin). Circulation. 2017;135(25):2494-504.
  • 68
    Miao Q, Ndao M. Trypanosoma cruzi infection and host lipid metabolism. Mediators Inflamm. 2014;2014:902038.
  • 69
    de la Llera Moya M, McGillicuddy FC, Hinkle CC, Byrne M, Joshi MR, Nguyen V, et al. Inflammation modulates human HDL composition and function in vivo. Atherosclerosis. 2012;222(2):390-4.
  • 70
    Parker TS, Levine DM, Chang JC, Laxer J, Coffin CC, Rubin AL. Reconstituted high-density lipoprotein neutralizes gram-negative bacterial lipopolysaccharides in human whole blood. Infect Immun. 1995;63(1):253-8.
  • 71
    Madsen CM, Varbo A, Tybjærg-Hansen A, Frikke-Schmidt R, Nordestgaard BG. U-shaped relationship of HDL and risk of infectious disease: two prospective population-based cohort studies. Eur Heart J. 2018;39(14):1181-90.
  • 72
    Trinder M, Walley KR, Boyd JH, Brunham LR. Causal Inference for Genetically Determined Levels of High-Density Lipoprotein Cholesterol and Risk of Infectious Disease. Arterioscler Thromb Vasc Biol. 2020;40(1):267-78.
  • 73
    McTaggart F, Jones P. Effects of statins on high-density lipoproteins: a potential contribution to cardiovascular benefit. Cardiovasc Drugs Ther. 2008;22(4):321-38.
  • 74
    Maejima T, Yamazaki H, Aoki T, Tamaki T, Sato F, Kitahara M, et al. Effect of pitavastatin on apolipoprotein A-I production in HepG2 cell. Biochem Biophys Res Commun. 2004;324(2):835-9.
  • 75
    Zanotti I, Favari E, Sposito AC, Rothblat GH, Bernini F. Pitavastatin increases ABCA1-mediated lipid efflux from Fu5AH rat hepatoma cells. Biochem Biophys Res Commun. 2004;321(3):670-4.
  • 76
    Sposito AC, Carmo HR, Barreto J, Sun L, Carvalho LSF, Feinstein SB, et al. HDL-Targeted Therapies During Myocardial Infarction. Cardiovasc Drugs Ther. 2019;33(3):371-81.
  • 77
    Jones PH, Davidson MH, Stein EA, Bays HE, McKenney JM, Miller E, et al. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial). Am J Cardiol. 2003;92(2):152-60.
  • 78
    Barter PJ, Brandrup-Wognsen G, Palmer MK, Nicholls SJ. Effect of statins on HDL-C: a complex process unrelated to changes in LDL-C: analysis of the VOYAGER Database. J Lipid Res. 2010;51(6):1546-53.
  • 79
    Kingwell BA, Chapman MJ. Future of high-density lipoprotein infusion therapies: potential for clinical management of vascular disease. Circulation. 2013;128(10):1112-21.
  • 80
    Ferri N, Corsini A, Sirtori CR, Ruscica M. Present therapeutic role of cholesteryl ester transfer protein inhibitors. Pharmacol Res. 2018;128:29-41.
  • 81
    Nurmohamed NS, Ditmarsch M, Kastelein JJP. CETP-inhibitors: from HDL-C to LDL-C lowering agents? Cardiovasc Res. 2021:cvab350.
  • 82
    Tall AR, Rader DJ. Trials and Tribulations of CETP Inhibitors. Circ Res. 2018;122(1):106-12.
  • 83
    Moreno C, Greil R, Demirkan F, Tedeschi A, Anz B, Larratt L, et al. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20(1):43-56.
  • 84
    Schwartz GG, Olsson AG, Abt M, Ballantyne CM, Barter PJ, Brumm J, et al. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N Engl J Med. 2012;367(22):2089-99.
  • 85
    Lincoff AM, Nicholls SJ, Riesmeyer JS, Barter PJ, Brewer HB, Fox KAA, et al. Evacetrapib and Cardiovascular Outcomes in High-Risk Vascular Disease. N Engl J Med. 2017;376(20):1933-42.
  • 86
    HPS3/TIMI55–REVEAL Collaborative Group, Bowman L, Hopewell JC, Chen F, Wallendszus K, Stevens W, Collins R, et al. Effects of Anacetrapib in Patients with Atherosclerotic Vascular Disease. N Engl J Med. 2017;377(13):1217-27.
  • 87
    Hovingh GK, Kastelein JJ, van Deventer SJ, Round P, Ford J, Saleheen D, et al. Cholesterol ester transfer protein inhibition by TA-8995 in patients with mild dyslipidaemia (TULIP): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet. 2015;386(9992):452-60.
  • 88
    Cupido AJ, Reeskamp LF, Hingorani AD, Finan C, Asselbergs FW, Hovingh GK, et al. Joint Genetic Inhibition of PCSK9 and CETP and the Association With Coronary Artery Disease: A Factorial Mendelian Randomization Study. JAMA Cardiol. 2022;7(9):955-64.
  • 89
    Tardif JC, Pfeffer MA, Kouz S, Koenig W, Maggioni AP, McMurray JJV, et al. Pharmacogenetics-guided dalcetrapib therapy after an acute coronary syndrome: the dal-GenE trial. Eur Heart J. 2022;43(39):3947-56.
  • 90
    Liu C, Dhindsa D, Almuwaqqat Z, Ko YA, Mehta A, Alkhoder AA, et al. Association Between High-Density Lipoprotein Cholesterol Levels and Adverse Cardiovascular Outcomes in High-risk Populations. JAMA Cardiol. 2022;7(7):672-80.
  • 91
    Chapman MJ. Fibrates in 2003: therapeutic action in atherogenic dyslipidaemia and future perspectives. Atherosclerosis. 2003;171(1):1-13.
  • 92
    Woudberg NJ, Pedretti S, Lecour S, Schulz R, Vuilleumier N, James RW, et al. Pharmacological Intervention to Modulate HDL: What Do We Target? Front Pharmacol. 2017;8:989.
  • 93
    Manninen V, Tenkanen L, Koskinen P, Huttunen JK, Mänttäri M, Heinonen OP, et al. Joint effects of serum triglyceride and LDL cholesterol and HDL cholesterol concentrations on coronary heart disease risk in the Helsinki Heart Study. Implications for treatment. Circulation. 1992;85(1):37-45.
  • 94
    Robins SJ, Collins D, Wittes JT, Papademetriou V, Deedwania PC, Schaefer EJ, et al. Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial. JAMA. 2001;285(12):1585-91.
  • 95
    Bezafibrate Infarction Prevention (BIP) study. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease. Circulation. 2000;102(1):21-7.
  • 96
    Elam M, Lovato L, Ginsberg H. The ACCORD-Lipid study: implications for treatment of dyslipidemia in Type 2 diabetes mellitus. Clin Lipidol. 2011;6(1):9-20.
  • 97
    Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. 2005;366(9500):1849-61.
  • 98
    Pradhan AD, Paynter NP, Everett BM, Glynn RJ, Amarenco P, Elam M, et al. Rationale and design of the Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) study. Am Heart J. 2018;206:80-93.
  • 99
    Kamanna VS, Kashyap ML. Mechanism of action of niacin. Am J Cardiol. 2008;101(8A):20B-26B.
  • 100
    Song WL, FitzGerald GA. Niacin, an old drug with a new twist. J Lipid Res. 2013;54(10):2586-94.
  • 101
    McKenney JM, Proctor JD, Harris S, Chinchili VM. A comparison of the efficacy and toxic effects of sustained- vs immediate-release niacin in hypercholesterolemic patients. JAMA. 1994;271(9):672-7.
  • 102
    Birjmohun RS, Hutten BA, Kastelein JJ, Stroes ES. Efficacy and safety of high-density lipoprotein cholesterol-increasing compounds: a meta-analysis of randomized controlled trials. J Am Coll Cardiol. 2005;45(2):185-97.
  • 103
    AIM-HIGH Investigators, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365(24):2255-67.
  • 104
    HPS2-THRIVE Collaborative Group. HPS2-THRIVE randomized placebo-controlled trial in 25 673 high-risk patients of ER niacin/laropiprant: trial design, pre-specified muscle and liver outcomes, and reasons for stopping study treatment. Eur Heart J. 2013;34(17):1279-91.
  • 105
    Patel S, Drew BG, Nakhla S, Duffy SJ, Murphy AJ, Barter PJ, et al. Reconstituted high-density lipoprotein increases plasma high-density lipoprotein anti-inflammatory properties and cholesterol efflux capacity in patients with type 2 diabetes. J Am Coll Cardiol. 2009;53(11):962-71.
  • 106
    Tardif JC, Grégoire J, L’Allier PL, Ibrahim R, Lespérance J, Heinonen TM, et al. Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial. JAMA. 2007;297(15):1675-82.
  • 107
    Kalayci A, Gibson CM, Ridker PM, Wright SD, Kingwell BA, Korjian S, et al. ApoA-I Infusion Therapies Following Acute Coronary Syndrome: Past, Present, and Future. Curr Atheroscler Rep. 2022;24(7):585-97.
  • 108
    Kee P, Rye KA, Taylor JL, Barrett PH, Barter PJ. Metabolism of apoA-I as lipid-free protein or as component of discoidal and spherical reconstituted HDLs: studies in wild-type and hepatic lipase transgenic rabbits. Arterioscler Thromb Vasc Biol. 2002;22(11):1912-7.
  • 109
    Hovingh GK, Smits LP, Stefanutti C, Soran H, Kwok S, de Graaf 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: The Modifying Orphan Disease Evaluation (MODE) study. Am Heart J. 2015;169(5):736-42.e1.
  • 110
    Tardif JC, Ballantyne CM, Barter P, Dasseux JL, Fayad ZA, Guertin MC, et al. Effects of the high-density lipoprotein mimetic agent CER-001 on coronary atherosclerosis in patients with acute coronary syndromes: a randomized trial. Eur Heart J. 2014;35(46):3277-86.
  • 111
    Ossoli A, Strazzella A, Rottoli D, Zanchi C, Locatelli M, Zoja C, et al. CER-001 ameliorates lipid profile and kidney disease in a mouse model of familial LCAT deficiency. Metabolism. 2021;116:154464.
  • 112
    Pavanello C, Turri M, Strazzella A, Tulissi P, Pizzolitto S, De Maglio G, et al. The HDL mimetic CER-001 remodels plasma lipoproteins and reduces kidney lipid deposits in inherited lecithin:cholesterol acyltransferase deficiency. J Intern Med. 2022;291(3):364-70.
  • 113
    Faguer S, Colombat M, Chauveau D, Bernadet-Monrozies P, Beq A, Delas A, et al. Administration of the High-Density Lipoprotein Mimetic CER-001 for Inherited Lecithin-Cholesterol Acyltransferase Deficiency. Ann Intern Med. 2021;174(7):1022-5.
  • 114
    Nissen SE, Tsunoda T, Tuzcu EM, Schoenhagen P, Cooper CJ, Yasin M, et al. Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. JAMA. 2003;290(17):2292-300.
  • 115
    Reijers JAA, Kallend DG, Malone KE, Jukema JW, Wijngaard PLJ, Burggraaf J, et al. MDCO-216 Does Not Induce Adverse Immunostimulation, in Contrast to Its Predecessor ETC-216. Cardiovasc Drugs Ther. 2017;31(4):381-9.
  • 116
    Mertens A, Verhamme P, Bielicki JK, Phillips MC, Quarck R, Verreth W, et al. Increased low-density lipoprotein oxidation and impaired high-density lipoprotein antioxidant defense are associated with increased macrophage homing and atherosclerosis in dyslipidemic obese mice: LCAT gene transfer decreases atherosclerosis. Circulation. 2003;107(12):1640-6.
  • 117
    Furbee JW, Sawyer JK, Parks JS. Lecithin:cholesterol acyltransferase deficiency increases atherosclerosis in the low density lipoprotein receptor and apolipoprotein E knockout mice. J Biol Chem. 2002;277(5):3511-9.
  • 118
    Shamburek RD, Bakker-Arkema R, Shamburek AM, Freeman LA, Amar MJ, Auerbach B, et al. Safety and Tolerability of ACP-501, a Recombinant Human Lecithin:Cholesterol Acyltransferase, in a Phase 1 Single-Dose Escalation Study. Circ Res. 2016;118(1):73-82.
  • 119
    Shamburek RD, Bakker-Arkema R, Auerbach BJ, Krause BR, Homan R, Amar MJ, et al. Familial lecithin:cholesterol acyltransferase deficiency: First-in-human treatment with enzyme replacement. J Clin Lipidol. 2016;10(2):356-67.
  • 120
    Ossoli A, Simonelli S, Varrenti M, Morici N, Oliva F, Stucchi M, et al. Recombinant LCAT (Lecithin:Cholesterol Acyltransferase) Rescues Defective HDL (High-Density Lipoprotein)-Mediated Endothelial Protection in Acute Coronary Syndrome. Arterioscler Thromb Vasc Biol. 2019;39(5):915-24.
  • 121
    George RT, Abuhatzira L, Stoughton SM, Karathanasis SK, She D, Jin C, et al. MEDI6012: Recombinant Human Lecithin Cholesterol Acyltransferase, High-Density Lipoprotein, and Low-Density Lipoprotein Receptor-Mediated Reverse Cholesterol Transport. J Am Heart Assoc. 2021;10(13):e014572.
  • 122
    Anantharamaiah GM, Jones JL, Brouillette CG, Schmidt CF, Chung BH, Hughes TA, et al. Studies of synthetic peptide analogs of the amphipathic helix. Structure of complexes with dimyristoyl phosphatidylcholine. J Biol Chem. 1985;260(18):10248-55.
  • 123
    Datta G, Chaddha M, Hama S, Navab M, Fogelman AM, Garber DW, et al. Effects of increasing hydrophobicity on the physical-chemical and biological properties of a class A amphipathic helical peptide. J Lipid Res. 2001;42(7):1096-104.
  • 124
    Garber DW, Datta G, Chaddha M, algunachari MN, Hama SY, Navab M, et al. A new synthetic class A amphipathic peptide analogue protects mice from diet-induced atherosclerosis. J Lipid Res. 2001;42(4):545-52.
  • 125
    Navab M, Anantharamaiah GM, Hama S, Garber DW, Chaddha M, Hough G, et al. Oral administration of an Apo A-I mimetic Peptide synthesized from D-amino acids dramatically reduces atherosclerosis in mice independent of plasma cholesterol. Circulation. 2002;105(3):290-2.
  • 126
    Gou S, Wang L, Zhong C, Chen X, Ouyang X, Li B, et al. A novel apoA-I mimetic peptide suppresses atherosclerosis by promoting physiological HDL function in apoE. Br J Pharmacol. 2020;177(20):4627-444.
  • 127
    Bailey D, Jahagirdar R, Gordon A, Hafiane A, Campbell S, Chatur S, et al. RVX-208: a small molecule that increases apolipoprotein A-I and high-density lipoprotein cholesterol in vitro and in vivo. J Am Coll Cardiol. 2010;55(23):2580-9.
  • 128
    Nicholls SJ, Puri R, Wolski K, Ballantyne CM, Barter PJ, Brewer HB, et al. Effect of the BET Protein Inhibitor, RVX-208, on Progression of Coronary Atherosclerosis: Results of the Phase 2b, Randomized, Double-Blind, Multicenter, ASSURE Trial. Am J Cardiovasc Drugs. 2016;16(1):55-65.
  • 129
    Rayner KJ, Sheedy FJ, Esau CC, Hussain FN, Temel RE, Parathath S, et al. Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J Clin Invest. 2011;121(7):2921-31.
  • 130
    Rotllan N, Ramírez CM, Aryal B, Esau CC, Fernández-Hernando C. Therapeutic silencing of microRNA-33 inhibits the progression of atherosclerosis in Ldlr-/- mice--brief report. Arterioscler Thromb Vasc Biol. 2013;33(8):1973-7.
  • 131
    Ouimet M, Ediriweera HN, Gundra UM, Sheedy FJ, Ramkhelawon B, Hutchison SB, et al. MicroRNA-33-dependent regulation of macrophage metabolism directs immune cell polarization in atherosclerosis. J Clin Invest. 2015;125(12):4334-48.
  • 132
    Marquart TJ, Wu J, Lusis AJ, Baldán Á. Anti-miR-33 therapy does not alter the progression of atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2013;33(3):455-8.
  • 133
    Bell TA, Graham MJ, Lee RG, Mullick AE, Fu W, Norris D, et al. Antisense oligonucleotide inhibition of cholesteryl ester transfer protein enhances RCT in hyperlipidemic, CETP transgenic, LDLr-/- mice. J Lipid Res. 2013;54(10):2647-57.
  • 134
    Schmitz J, Gouni-Berthold I. APOC-III Antisense Oligonucleotides: A New Option for the Treatment of Hypertriglyceridemia. Curr Med Chem. 2018;25(13):1567-76.
  • 135
    Ishida T, Choi S, Kundu RK, Hirata K, Rubin EM, Cooper AD, et al. Endothelial lipase is a major determinant of HDL level. J Clin Invest. 2003;111(3):347-55.
  • 136
    Ishida T, Choi SY, Kundu RK, Spin J, Yamashita T, Hirata K, et al. Endothelial lipase modulates susceptibility to atherosclerosis in apolipoprotein-E-deficient mice. J Biol Chem. 2004;279(43):45085-92.
  • 137
    Jin W, Millar JS, Broedl U, Glick JM, Rader DJ. Inhibition of endothelial lipase causes increased HDL cholesterol levels in vivo. J Clin Invest. 2003;111(3):357-62.
  • 138
    Goodman KB, Bury MJ, Cheung M, Cichy-Knight MA, Dowdell SE, Dunn AK, et al. Discovery of potent, selective sulfonylfuran urea endothelial lipase inhibitors. Bioorg Med Chem Lett. 2009;19(1):27-30.
  • 139
    Edmondson AC, Brown RJ, Kathiresan S, Cupples LA, Demissie S, Manning AK, et al. Loss-of-function variants in endothelial lipase are a cause of elevated HDL cholesterol in humans. J Clin Invest. 2009;119(4):1042-50.
  • 140
    Badellino KO, Wolfe ML, Reilly MP, Rader DJ. Endothelial lipase concentrations are increased in metabolic syndrome and associated with coronary atherosclerosis. PLoS Med. 2006;3(2):e22.
  • 141
    Badellino KO, Wolfe ML, Reilly MP, Rader DJ. Endothelial lipase is increased in vivo by inflammation in humans. Circulation. 2008;117(5):678-85.
  • 142
    Zelcer N, Tontonoz P. Liver X receptors as integrators of metabolic and inflammatory signaling. J Clin Invest. 2006;116(3):607-14.
  • 143
    Zanotti I, Potì F, Pedrelli M, Favari E, Moleri E, Franceschini G, et al. The LXR agonist T0901317 promotes the reverse cholesterol transport from macrophages by increasing plasma efflux potential. J Lipid Res. 2008;49(5):954-60.
  • 144
    Kirchgessner TG, Sleph P, Ostrowski J, Lupisella J, Ryan CS, Liu X, et al. Beneficial and Adverse Effects of an LXR Agonist on Human Lipid and Lipoprotein Metabolism and Circulating Neutrophils. Cell Metab. 2016;24(2):223-33.
  • 145
    Laffitte BA, Repa JJ, Joseph SB, Wilpitz DC, Kast HR, Mangelsdorf DJ, et al. LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proc Natl Acad Sci U S A. 2001;98(2):507-12.
  • 146
    Baldán A, Bojanic DD, Edwards PA. The ABCs of sterol transport. J Lipid Res. 2009;50 Suppl:S80-5.

Publication Dates

  • Publication in this collection
    27 Jan 2023
  • Date of issue
    Feb 2023

History

  • Received
    02 May 2022
  • Accepted
    18 Sept 2022
Sociedade Brasileira de Endocrinologia e Metabologia Rua Botucatu, 572 - Conjuntos 81/83, 04023-061 São Paulo SP Brasil, Tel: (55 11) 5575-0311 - São Paulo - SP - Brazil
E-mail: aem.editorial.office@endocrino.org.br