1
|
Brewer HB, Schaefer EJ, Foldyna B, Ghoshhajra BB. High-density lipoprotein infusion therapy: A review. J Clin Lipidol 2024; 18:e374-e383. [PMID: 38782655 DOI: 10.1016/j.jacl.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 05/25/2024]
Abstract
Increased cholesterol-rich, low-density, non-calcified atheromas as assessed by computer coronary tomography angiography analyses have been shown to predict myocardial infarction significantly better than coronary artery calcium score or the presence of obstructive coronary artery disease (CAD) as evaluated with standard coronary angiography. Low serum high-density lipoprotein (HDL) cholesterol values are an independent risk factor for CAD. Very small, lipid-poor preβ-1 HDL particles have been shown to be most effective in promoting cellular cholesterol efflux. HDL infusions have been documented to reduce aortic atherosclerosis in cholesterol-fed animal models. However, human studies using infusions of either the HDL mimetic containing recombinant apolipoprotein (apo) A-I Milano or Cerenis Compound-001 with native recombinant apoA-I have been mainly negative in promoting coronary atherosclerosis progression as assessed by intravascular ultrasound. In contrast, a study using 7 weekly infusions of autologous delipidated HDL in six homozygous familial hypercholesterolemic patients was effective in promoting significant regression of low-density non-calcified coronary atheroma regression as assessed by computed coronary angiography. This therapy has received Food and Drug Administration approval. Commonwealth Serum Laboratories has carried out a large clinical endpoint trial using an HDL complex (native apoA-I with phospholipid), and the results were negative. Our purpose is to review animal and human studies using various forms of HDL infusion therapy to promote regression of atherosclerosis. In our view, differences in results may be due to: 1) the HDL preparations used, 2) the subjects studied, and 3) the methods used to assess coronary atherosclerosis.
Collapse
Affiliation(s)
| | - Ernst J Schaefer
- Boston Heart Diagnostics, Framingham, MA, USA (Dr Schaefer); Department of Medicine, Tufts University School of Medicine, Boston, MA, USA (Dr Schaefer).
| | - Borek Foldyna
- Division of Cardiovascular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA (Drs Foldyna and Ghoshhajra)
| | - Brian B Ghoshhajra
- Division of Cardiovascular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA (Drs Foldyna and Ghoshhajra)
| |
Collapse
|
2
|
Rani A, Marsche G. A Current Update on the Role of HDL-Based Nanomedicine in Targeting Macrophages in Cardiovascular Disease. Pharmaceutics 2023; 15:1504. [PMID: 37242746 PMCID: PMC10221824 DOI: 10.3390/pharmaceutics15051504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
High-density lipoproteins (HDL) are complex endogenous nanoparticles involved in important functions such as reverse cholesterol transport and immunomodulatory activities, ensuring metabolic homeostasis and vascular health. The ability of HDL to interact with a plethora of immune cells and structural cells places it in the center of numerous disease pathophysiologies. However, inflammatory dysregulation can lead to pathogenic remodeling and post-translational modification of HDL, rendering HDL dysfunctional or even pro-inflammatory. Monocytes and macrophages play a critical role in mediating vascular inflammation, such as in coronary artery disease (CAD). The fact that HDL nanoparticles have potent anti-inflammatory effects on mononuclear phagocytes has opened new avenues for the development of nanotherapeutics to restore vascular integrity. HDL infusion therapies are being developed to improve the physiological functions of HDL and to quantitatively restore or increase the native HDL pool. The components and design of HDL-based nanoparticles have evolved significantly since their initial introduction with highly anticipated results in an ongoing phase III clinical trial in subjects with acute coronary syndrome. The understanding of mechanisms involved in HDL-based synthetic nanotherapeutics is critical to their design, therapeutic potential and effectiveness. In this review, we provide a current update on HDL-ApoA-I mimetic nanotherapeutics, highlighting the scope of treating vascular diseases by targeting monocytes and macrophages.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria;
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
3
|
Bonilha I, Luchiari B, Nadruz W, Sposito AC. Very low HDL levels: clinical assessment and management. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:3-18. [PMID: 36651718 PMCID: PMC9983789 DOI: 10.20945/2359-3997000000585] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
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.
Collapse
Affiliation(s)
- Isabella Bonilha
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Beatriz Luchiari
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Wilson Nadruz
- Universidade de Campinas (Unicamp), Divisão de Cardiologia, Campinas, SP, Brasil
| | - Andrei C Sposito
- Universidade de Campinas (Unicamp), Laboratório de Biologia Vascular e Aterosclerose (AtheroLab), Divisão de Cardiologia, Campinas, SP, Brasil,
| |
Collapse
|
4
|
Cui H, Du Q. HDL and ASCVD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:109-118. [DOI: 10.1007/978-981-19-1592-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
5
|
Lee CK, Liao CW, Meng SW, Wu WK, Chiang JY, Wu MS. Lipids and Lipoproteins in Health and Disease: Focus on Targeting Atherosclerosis. Biomedicines 2021; 9:biomedicines9080985. [PMID: 34440189 PMCID: PMC8393881 DOI: 10.3390/biomedicines9080985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Despite advances in pharmacotherapy, intervention devices and techniques, residual cardiovascular risks still cause a large burden on public health. Whilst most guidelines encourage achieving target levels of specific lipids and lipoproteins to reduce these risks, increasing evidence has shown that molecular modification of these lipoproteins also has a critical impact on their atherogenicity. Modification of low-density lipoprotein (LDL) by oxidation, glycation, peroxidation, apolipoprotein C-III adhesion, and the small dense subtype largely augment its atherogenicity. Post-translational modification by oxidation, carbamylation, glycation, and imbalance of molecular components can reduce the capacity of high-density lipoprotein (HDL) for reverse cholesterol transport. Elevated levels of triglycerides (TGs), apolipoprotein C-III and lipoprotein(a), and a decreased level of apolipoprotein A-I are closely associated with atherosclerotic cardiovascular disease. Pharmacotherapies aimed at reducing TGs, lipoprotein(a), and apolipoprotein C-III, and enhancing apolipoprotein A-1 are undergoing trials, and promising preliminary results have been reported. In this review, we aim to update the evidence on modifications of major lipid and lipoprotein components, including LDL, HDL, TG, apolipoprotein, and lipoprotein(a). We also discuss examples of translating findings from basic research to potential therapeutic targets for drug development.
Collapse
Affiliation(s)
- Chih-Kuo Lee
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu 300, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Che-Wei Liao
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Department of Internal Medicine, National Taiwan University Cancer Center, Taipei 106, Taiwan
| | - Shih-Wei Meng
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu 300, Taiwan
| | - Wei-Kai Wu
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Jiun-Yang Chiang
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei 100, Taiwan
- Correspondence: (J.-Y.C.); (M.-S.W.)
| | - Ming-Shiang Wu
- College of Medicine, National Taiwan University, Taipei 100, Taiwan; (C.-K.L.); (C.-W.L.); (S.-W.M.); (W.-K.W.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Correspondence: (J.-Y.C.); (M.-S.W.)
| |
Collapse
|
6
|
B Uribe K, Benito-Vicente A, Martin C, Blanco-Vaca F, Rotllan N. (r)HDL in theranostics: how do we apply HDL's biology for precision medicine in atherosclerosis management? Biomater Sci 2021; 9:3185-3208. [PMID: 33949389 DOI: 10.1039/d0bm01838d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
High-density lipoproteins (HDL) are key players in cholesterol metabolism homeostasis since they are responsible for transporting excess cholesterol from peripheral tissues to the liver. Imbalance in this process, due to either excessive accumulation or impaired clearance, results in net cholesterol accumulation and increases the risk of cardiovascular disease (CVD). Therefore, significant effort has been focused on the development of therapeutic tools capable of either directly or indirectly enhancing HDL-guided reverse cholesterol transport (RCT). More recently, in light of the emergence of precision nanomedicine, there has been renewed research interest in attempting to take advantage of the development of advanced recombinant HDL (rHDL) for both therapeutic and diagnostic purposes. In this review, we provide an update on the different approaches that have been developed using rHDL, focusing on the rHDL production methodology and rHDL applications in theranostics. We also compile a series of examples highlighting potential future perspectives in the field.
Collapse
Affiliation(s)
- Kepa B Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramon 182, 20014, Donostia San Sebastián, Spain.
| | - Asier Benito-Vicente
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Cesar Martin
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco, Apdo.644, 48080 Bilbao, Spain.
| | - Francisco Blanco-Vaca
- Servei de Bioquímica, Hospital Santa Creu i Sant Pau-Institut d'Investigacions Biomèdiques (IIB) Sant Pau, 08041 Barcelona, Spain. and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain and Departament de Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Spain and Institut de Recerca de l'Hospital Santa Creu i Sant Pau-Institut d'Investigacions Biomèdiques (IIB) Sant Pau, 08025 Barcelona, Spain.
| | - Noemi Rotllan
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain and Institut de Recerca de l'Hospital Santa Creu i Sant Pau-Institut d'Investigacions Biomèdiques (IIB) Sant Pau, 08025 Barcelona, Spain.
| |
Collapse
|
7
|
Escolà-Gil JC, Rotllan N, Julve J, Blanco-Vaca F. Reverse Cholesterol Transport Dysfunction Is a Feature of Familial Hypercholesterolemia. Curr Atheroscler Rep 2021; 23:29. [PMID: 33914189 DOI: 10.1007/s11883-021-00928-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 02/21/2023]
Abstract
PURPOSE OF REVIEW We seek to establish whether high-density lipoprotein HDL metabolism and reverse cholesterol transport (RCT) impairment is an intrinsic feature of familial hypercholesterolemia (FH). RECENT FINDINGS RCT from macrophages (m-RCT), a vascular cell type of major influence on atherosclerosis, is impaired in FH due to defective low-density lipoprotein receptor (LDLR) function via both the HDL- and LDL-mediated pathways. Potential mechanisms include impaired HDL metabolism, which is linked to increased LDL levels, as well as the increased transport of cellular unesterified cholesterol to LDL, which presents a defective catabolism. RCT dysfunction is consistently associated with mutation-positive FH linked to decreased HDL levels as well as impaired HDL remodeling and LDLR function. It remains to be explored whether these alterations are also present in less well-characterized forms of FH, such as cases with no identified mutations, and whether they are fully corrected by current standard treatments.
Collapse
Affiliation(s)
- Joan Carles Escolà-Gil
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain. .,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain. .,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Noemí Rotllan
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain
| | - Josep Julve
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisco Blanco-Vaca
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Madrid, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain
| |
Collapse
|
8
|
Ossoli A, Strazzella A, Rottoli D, Zanchi C, Locatelli M, Zoja C, Simonelli S, Veglia F, Barbaras R, Tupin C, Dasseux JL, Calabresi L. CER-001 ameliorates lipid profile and kidney disease in a mouse model of familial LCAT deficiency. Metabolism 2021; 116:154464. [PMID: 33309714 DOI: 10.1016/j.metabol.2020.154464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/25/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE CER-001 is an HDL mimetic that has been tested in different pathological conditions, but never with LCAT deficiency. This study was designed to investigate whether the absence of LCAT affects the catabolic fate of CER-001, and to evaluate the effects of CER-001 on kidney disease associated with LCAT deficiency. METHODS Lcat-/- and wild-type mice received CER-001 (2.5, 5, 10 mg/kg) intravenously for 2 weeks. The plasma lipid/ lipoprotein profile and HDL subclasses were analyzed. In a second set of experiments, Lcat-/- mice were injected with LpX to induce renal disease and treated with CER-001 and then the plasma lipid profile, lipid accumulation in the kidney, albuminuria and glomerular podocyte markers were evaluated. RESULTS In Lcat-/- mice a decrease in total cholesterol and triglycerides, and an increase in HDL-c was observed after CER-001 treatment. While in wild-type mice CER-001 entered the classical HDL remodeling pathway, in the absence of LCAT it disappeared from the plasma shortly after injection and ended up in the kidney. In a mouse model of renal disease in LCAT deficiency, treatment with CER-001 at 10 mg/kg for one month had beneficial effects not only on the lipid profile, but also on renal disease, by limiting albuminuria and podocyte dysfunction. CONCLUSIONS Treatment with CER-001 ameliorates the dyslipidemia typically associated with LCAT deficiency and more importantly limits renal damage in a mouse model of renal disease in LCAT deficiency. The present results provide a rationale for using CER-001 in FLD patients.
Collapse
Affiliation(s)
- Alice Ossoli
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Arianna Strazzella
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Daniela Rottoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Cristina Zanchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Monica Locatelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Carlamaria Zoja
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Sara Simonelli
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | | | | | | | | | - Laura Calabresi
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
9
|
Song F, Zhou Y, Zhang K, Liang YF, He X, Li L. The role of the plasma glycosylated hemoglobin A1c/Apolipoprotein A-l ratio in predicting cardiovascular outcomes in acute coronary syndrome. Nutr Metab Cardiovasc Dis 2021; 31:570-578. [PMID: 33358616 DOI: 10.1016/j.numecd.2020.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND AIMS Glucose and lipid metabolism are major prognostic indicators of coronary heart disease. The ratio of plasma glycosylated hemoglobin A1c (HbA1c) to apolipoprotein A-l (ApoA-l) is an indirect measure of insulin resistance. The study aimed to evaluate whether the HbA1c/ApoA-1 ratio can predict the prognosis in patients with the acute coronary syndrome (ACS). METHODS AND RESULTS A total of 476 ACS patients diagnosed by coronary angiography were enrolled in this longitudinal, observational, retrospective study. Plasma HbA1c, fasting blood glucose and lipid profile were measured. Patients were stratified according to the tertiles of HbA1c/ApoA-l levels. Cox proportional hazard model was used to examine the predictive value of HbA1c/ApoA-l for study endpoints. The association between the Log HbA1c/ApoA-l ratio and major adverse cardiovascular events (MACEs) was estimated using multiple logistic regression. Baseline characteristics showed a mean age of 66 ± 8 years, and 52.5% were hypertensive, 26.8% diabetic, and 54.5% current or prior smokers. During a mean follow-up period of 22.3 ± 1.7 months, 59 deaths occurred. After adjusting for age, gender, smoking, hypertension, diabetes, and coronary artery disease severity, patients in the highest HbA1c/ApoA-l ratio tertile had a 4.36-fold increased risk of mortality compared with those in the lowest tertile. The multivariate logistic regression showed that the Log HbA1c/ApoA-l ratio was associated with MACEs (Odds ratio 2.95, p = 0.013). CONCLUSION After adjusting for traditional cardiovascular risk factors and ACS severity scores, the HbA1c/ApoA-1 ratio remained an independent predictor of all-cause mortality and MACEs in the ACS patients undergoing angiography.
Collapse
Affiliation(s)
- Feier Song
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Yu Zhou
- Division of Vascular Surgery, National-Local Joint Engineering Laboratory of Vascular Disease Treatment, Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangdong Engineering Laboratory of Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Kunyi Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Yuan-Feng Liang
- General Division, Guangdong Geriatric Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Xuyu He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Liwen Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| |
Collapse
|
10
|
Yuan W, Yu B, Yu M, Kuai R, Morin EE, Wang H, Hu D, Zhang J, Moon JJ, Chen YE, Guo Y, Schwendeman A. Synthetic high-density lipoproteins delivering liver X receptor agonist prevent atherogenesis by enhancing reverse cholesterol transport. J Control Release 2021; 329:361-371. [PMID: 33188828 DOI: 10.1016/j.jconrel.2020.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 10/22/2020] [Accepted: 11/08/2020] [Indexed: 12/14/2022]
Abstract
Liver X nuclear receptor (LXR) agonists are promising anti-atherosclerotic agents that increase the expression of cholesterol transporters on atheroma macrophages leading to increased efflux of cholesterol to endogenous high-density lipoprotein (HDL) acceptors. HDL subsequently delivers effluxed cholesterol to the liver by the process of reverse cholesterol transport, resulting in reduction of atherosclerotic plaques. However, LXR agonists administration triggers undesirable liver steatosis and hypertriglyceridemia due to increased fatty acid and sterol synthesis. LXR-induced liver toxicity, poor drug aqueous solubility and low levels of endogenous HDL acceptors in target patient populations limit the clinical translation of LXR agonists. Here, we propose a dual-antiatherogenic strategy for administration of the LXR agonist, T0901317 (T1317), by encapsulating in synthetic HDL (sHDL) nanoparticles. sHDL had been clinically proven to serve as cholesterol acceptors, resulting in plaque reduction in atherosclerosis patients. In addition, the hydrophobic core and endogenous atheroma-targeting ability of sHDL allow for encapsulation of water-insoluble drugs and their subsequent delivery to atheroma. Several compositions of sHDL were tested to optimize both T1317 encapsulation efficiency and ability of T1317-sHDL to efflux cholesterol. Optimized T1317-sHDL exhibited more efficient cholesterol efflux from macrophages and enhanced atheroma-targeting relative to free drug. Most importantly, in an apolipoprotein E deficient (ApoE-/-) atherosclerosis progression murine model, T1317-sHDL showed superior inhibition of atherogenesis and reduced hypertriglyceridemia side effects in comparison to the free drug and blank sHDL. The T1317-sHDL pharmacological efficacy was observed at doses lower than those previously described for LXR agents, which may have additional safety benefits. In addition, the established clinical manufacturing, safety and efficacy of blank sHDL nanoparticles used in this study could facilitate future clinical translation of LXR-loaded sHDLs.
Collapse
Affiliation(s)
- Wenmin Yuan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Bilian Yu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States; Department of Cardiovascular medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Emily E Morin
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Huilun Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Die Hu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Y Eugene Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Yanhong Guo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
11
|
Abstract
Earlier epidemiological studies have shown an inverse correlation between high-density lipoprotein cholesterol (HDLc) and coronary heart disease (CHD). This observation along with the finding that reverse cholesterol transport is mediated by HDL, supported the hypothesis that the HDL molecule has a cardioprotective role. More recently, epidemiological data suggest a U-shaped curve correlating HDLc and CHD. In addition, randomized clinical trials of drugs that significantly increase plasma HDLc levels, such as nicotinic acid and cholesterol ester transfer protein (CETP) inhibitors failed to show a reduction in major adverse cardiovascular events. These observations challenge the hypothesis that HDL has a cardioprotective role. It is possible that HDL quality and function is optimal only when de novo synthesis of apo A-I occurs. Inhibition of turnover of HDL with currently available agents yields HDL molecules that are ineffective in reverse cholesterol transport. To test this hypothesis, newer therapeutic drugs that increase de novo production of HDL and apo A-I should be tested in clinical trials.
Collapse
Affiliation(s)
- Julien J Feghaly
- Department of Medicine, School of Medicine, Saint Louis University, Saint Louis, MO, USA
| | - Arshag D Mooradian
- Department of Medicine, University of Florida College of Medicine, 653-1 West 8th Street, 4th Floor-LRC, Jacksonville, FL, 32209, USA.
| |
Collapse
|
12
|
Stadler JT, Marsche G. Obesity-Related Changes in High-Density Lipoprotein Metabolism and Function. Int J Mol Sci 2020; 21:E8985. [PMID: 33256096 PMCID: PMC7731239 DOI: 10.3390/ijms21238985] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
In obese individuals, atherogenic dyslipidemia is a very common and important factor in the increased risk of cardiovascular disease. Adiposity-associated dyslipidemia is characterized by low high-density lipoprotein cholesterol (HDL-C) levels and an increase in triglyceride-rich lipoproteins. Several factors and mechanisms are involved in lowering HDL-C levels in the obese state and HDL quantity and quality is closely related to adiponectin levels and the bioactive lipid sphingosine-1-phosphate. Recent studies have shown that obesity profoundly alters HDL metabolism, resulting in altered HDL subclass distribution, composition, and function. Importantly, weight loss through gastric bypass surgery and Mediterranean diet, especially when enriched with virgin olive oil, is associated with increased HDL-C levels and significantly improved metrics of HDL function. A thorough understanding of the underlying mechanisms is crucial for a better understanding of the impact of obesity on lipoprotein metabolism and for the development of appropriate therapeutic approaches. The objective of this review article was to summarize the newly identified changes in the metabolism, composition, and function of HDL in obesity and to discuss possible pathophysiological consequences.
Collapse
Affiliation(s)
- Julia T. Stadler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
13
|
Zheng KH, Kaiser Y, van Olden CC, Santos RD, Dasseux JL, Genest J, Gaudet D, Westerink J, Keyserling C, Verberne HJ, Leitersdorf E, Hegele RA, Descamps OS, Hopkins P, Nederveen AJ, Stroes ES. No benefit of HDL mimetic CER-001 on carotid atherosclerosis in patients with genetically determined very low HDL levels. Atherosclerosis 2020; 311:13-19. [DOI: 10.1016/j.atherosclerosis.2020.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/27/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022]
|
14
|
The Role of HDL and HDL Mimetic Peptides as Potential Therapeutics for Alzheimer's Disease. Biomolecules 2020; 10:biom10091276. [PMID: 32899606 PMCID: PMC7563116 DOI: 10.3390/biom10091276] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
The role of high-density lipoproteins (HDL) in the cardiovascular system has been extensively studied and the cardioprotective effects of HDL are well established. As HDL particles are formed both in the systemic circulation and in the central nervous system, the role of HDL and its associated apolipoproteins in the brain has attracted much research interest in recent years. Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder and the leading cause of dementia worldwide, for which there currently exists no approved disease modifying treatment. Multiple lines of evidence, including a number of large-scale human clinical studies, have shown a robust connection between HDL levels and AD. Low levels of HDL are associated with increased risk and severity of AD, whereas high levels of HDL are correlated with superior cognitive function. Although the mechanisms underlying the protective effects of HDL in the brain are not fully understood, many of the functions of HDL, including reverse lipid/cholesterol transport, anti-inflammation/immune modulation, anti-oxidation, microvessel endothelial protection, and proteopathy modification, are thought to be critical for its beneficial effects. This review describes the current evidence for the role of HDL in AD and the potential of using small peptides mimicking HDL or its associated apolipoproteins (HDL-mimetic peptides) as therapeutics to treat AD.
Collapse
|
15
|
Gaudet D, Durst R, Lepor N, Bakker-Arkema R, Bisgaier C, Masson L, Golden L, Kastelein JJ, Hegele RA, Stein E. Usefulness of Gemcabene in Homozygous Familial Hypercholesterolemia (from COBALT-1). Am J Cardiol 2019; 124:1876-1880. [PMID: 31685212 DOI: 10.1016/j.amjcard.2019.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 12/17/2022]
Abstract
Homozygous familial hypercholesterolemia (HoFH) is a rare genetic disorder characterized by severely elevated plasma low-density lipoprotein-cholesterol (LDL-C), and premature atherosclerotic cardiovascular disease. Depending on residual LDL receptor (LDLR) function, most HoFH patients respond modestly to statins, ezetimibe, and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors. However, LDL-C typically remains markedly elevated necessitating additional therapies, including apheresis. Gemcabene is a novel lipid-lowering agent with a mechanism of action independent of the LDLR, which has previously demonstrated the ability to reduce levels of LDL-C on top of maximally tolerated statins. The present study (COBALT-1) assessed efficacy, tolerability, and safety of gemcabene as an adjunctive therapy to current lipid-lowering treatment for familial hypercholesterolemia patients. Eight patients with either a clinical or genetic diagnosis of HoFH on stable standard of care, including statins, ezetimibe, and PCSK9 inhibitors, were treated with gemcabene in an open-label study for 12 weeks. DNA analysis for mutations in the LDLR, apolipoprotein B, and PCSK9 genes was performed. Patients received 300 mg gemcabene for the first 4 weeks, 600 mg for the next 4 weeks, and 900 mg for the final 4 weeks. All patients completed the 12-week study. Mean change from baseline in LDL-C was -26% (p = 0.004) at Week 4 (300 mg), -30% (p = 0.001) at Week 8 (600 mg), and -29% (p = 0.001) at Week 12 (900 mg). In conclusion, the COBALT-1 study demonstrates gemcabene has potential to significantly reduce LDL-C levels when used as an adjunctive therapy to current lipid-lowering treatment for familial hypercholesterolemia patients.
Collapse
Affiliation(s)
- Daniel Gaudet
- ECOGENE-21 and Department of Medicine, Université de Montréal, Chicoutimi, Quebec, Canada.
| | - Ronen Durst
- Center for Research, Prevention, and Treatment of Atherosclerosis, Jerusalem, Israel
| | - Norman Lepor
- Westside Medical Associates, Beverly Hills, California
| | - Rebecca Bakker-Arkema
- Gemphire Therapeutics, Inc., Livonia, Michigan; LIB Therapeutics, LLC, Cincinnati, Ohio
| | | | - Liz Masson
- Gemphire Therapeutics, Inc., Livonia, Michigan; CorMedix, Berkeley Heights, New Jersey
| | - Lee Golden
- Gemphire Therapeutics, Inc., Livonia, Michigan; Espero BioPharma, Inc., Jacksonville, Florida
| | | | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Western University, London, Ontario, Canada
| | - Evan Stein
- Metabolic & Atherosclerosis Research Center, Cincinnati, Ohio
| |
Collapse
|
16
|
Sirtori CR, Ruscica M, Calabresi L, Chiesa G, Giovannoni R, Badimon JJ. HDL therapy today: from atherosclerosis, to stent compatibility to heart failure. Ann Med 2019; 51:345-359. [PMID: 31729238 PMCID: PMC7877888 DOI: 10.1080/07853890.2019.1694695] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epidemiologically, high-density lipoprotein (HDL) cholesterol levels have been inversely associated to cardiovascular (CV) events, although a Mendelian Randomisation Study had failed to establish a clear causal role. Numerous atheroprotective mechanisms have been attributed to HDL, the main being the ability to promote cholesterol efflux from arterial walls; anti-inflammatory effects related to HDL ligands such as S1P (sphingosine-1-phosphate), resolvins and others have been recently identified. Experimental studies and early clinical investigations have indicated the potential of HDL to slow progression or induce regression of atherosclerosis. More recently, the availability of different HDL formulations, with different phospholipid moieties, has allowed to test other indications for HDL therapy. Positive reports have come from studies on coronary stent biocompatibility, where the use of HDL from different sources reduced arterial cell proliferation and thrombogenicity. The observation that low HDL-C levels may be associated with an enhanced risk of heart failure (HF) has also suggested that HDL therapy may be applied to this condition. HDL infusions or apoA-I gene transfer were able to reverse heart abnormalities, reduce diastolic resistance and improve cardiac metabolism. HDL therapy may be effective not only in atherosclerosis, but also in other conditions, of relevant impact on human health.Key messagesHigh-density lipoproteins have as a major activity that of removing excess cholesterol from tissues (particularly arteries).Knowledge on the activity of high-density lipoproteins on health have however significantly widened.HDL-therapy may help to improve stent biocompatibility and to reduce peripheral arterial resistance in heart failure.
Collapse
Affiliation(s)
- C R Sirtori
- Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - M Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - L Calabresi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - G Chiesa
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - R Giovannoni
- Department of Biology, University of Pisa, Pisa, Italy
| | - J J Badimon
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
17
|
Nicholls SJ, Andrews J, Kastelein JJP, Merkely B, Nissen SE, Ray KK, Schwartz GG, Worthley SG, Keyserling C, Dasseux JL, Griffith L, Kim SW, Janssan A, Di Giovanni G, Pisaniello AD, Scherer DJ, Psaltis PJ, Butters J. Effect of Serial Infusions of CER-001, a Pre-β High-Density Lipoprotein Mimetic, on Coronary Atherosclerosis in Patients Following Acute Coronary Syndromes in the CER-001 Atherosclerosis Regression Acute Coronary Syndrome Trial: A Randomized Clinical Trial. JAMA Cardiol 2019; 3:815-822. [PMID: 30046828 DOI: 10.1001/jamacardio.2018.2121] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance CER-001 is a negatively charged, engineered pre-β high-density lipoprotein (HDL) mimetic containing apolipoprotein A-I and sphingomyelin. Preliminary studies demonstrated favorable effects of CER-001 on cholesterol efflux and vascular inflammation. A post hoc reanalysis of a previously completed study of intravenous infusion of CER-001, 3 mg/k, showed that the intravenous infusion in patients with a high coronary plaque burden promoted regression as assessed by intravascular ultrasonography. Objective To determine the effect of infusing CER-001 on coronary atherosclerosis progression in statin-treated patients. Design, Setting, and Participants A double-blind, randomized, multicenter trial evaluating the effect of 10 weekly intravenous infusions of CER-001, 3 mg/kg, (n = 135) or placebo (n = 137) in patients with an acute coronary syndrome (ACS) and baseline percent atheroma volume (PAV) greater than 30% in the proximal segment of an epicardial artery by intravascular ultrasonography. The study included 34 academic and community hospitals in Australia, Hungary, the Netherlands, and the United States in patients with ACS presenting for coronary angiography. Patients were enrolled from August 15, 2015, to November 19, 2016. Interventions Participants were randomized to receive weekly CER-001, 3 mg/kg, or placebo for 10 weeks in addition to statins. Main Outcomes and Measures The primary efficacy measure was the nominal change in PAV from baseline to day 78 measured by serial intravascular ultrasonography imaging. The secondary efficacy measures were nominal change in normalized total atheroma volume and percentage of patients demonstrating plaque regression. Safety and tolerability were also evaluated. Results Among 293 patients (mean [SD] age, 59.8 [9.4] years; 217 men [79.8%] and 261 white race/ethnicity [96.0%]), 86 (29%) had statin prior use prior to the index ACS and 272 (92.8%) had evaluable imaging at follow-up. The placebo and CER-001 groups had similar posttreatment median levels of low-density lipoprotein cholesterol (74 mg/dL vs 79 mg/dL; P = .15) and high-density lipoprotein cholesterol (43 mg/dL vs 44 mg/dL; P = .66). The primary efficacy measure, PAV, decreased 0.41% with placebo (P = .005 compared with baseline), but not with CER-001 (-0.09%; P = .67 compared with baseline; between group differences, 0.32%; P = .15). Similar percentages of patients in the placebo and CER-001 groups demonstrated regression of PAV (57.7% vs 53.3%; P = .49). Infusions were well tolerated, with no differences in clinical and laboratory adverse events observed between treatment groups. Conclusions and Relevance Infusion of CER-001 did not promote regression of coronary atherosclerosis in statin-treated patients with ACS and high plaque burden. Trial Registration ClinicalTrials.gov Identifier: NCT2484378.
Collapse
Affiliation(s)
- Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Jordan Andrews
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - John J P Kastelein
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Kausik K Ray
- School of Public Health, Imperial College London, London, England
| | | | | | | | | | - Liddy Griffith
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Susan W Kim
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Alex Janssan
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Giuseppe Di Giovanni
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Anthony D Pisaniello
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Daniel J Scherer
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Peter J Psaltis
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Julie Butters
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
18
|
Review of the long-term safety of lomitapide: a microsomal triglycerides transfer protein inhibitor for treating homozygous familial hypercholesterolemia. Expert Opin Drug Saf 2019; 18:403-414. [DOI: 10.1080/14740338.2019.1602606] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
19
|
Yu XH, Zhang DW, Zheng XL, Tang CK. Cholesterol transport system: An integrated cholesterol transport model involved in atherosclerosis. Prog Lipid Res 2018; 73:65-91. [PMID: 30528667 DOI: 10.1016/j.plipres.2018.12.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/30/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the pathological basis of most cardiovascular disease (CVD), is closely associated with cholesterol accumulation in the arterial intima. Excessive cholesterol is removed by the reverse cholesterol transport (RCT) pathway, representing a major antiatherogenic mechanism. In addition to the RCT, other pathways are required for maintaining the whole-body cholesterol homeostasis. Thus, we propose a working model of integrated cholesterol transport, termed the cholesterol transport system (CTS), to describe body cholesterol metabolism. The novel model not only involves the classical view of RCT but also contains other steps, such as cholesterol absorption in the small intestine, low-density lipoprotein uptake by the liver, and transintestinal cholesterol excretion. Extensive studies have shown that dysfunctional CTS is one of the major causes for hypercholesterolemia and atherosclerosis. Currently, several drugs are available to improve the CTS efficiently. There are also several therapeutic approaches that have entered into clinical trials and shown considerable promise for decreasing the risk of CVD. In recent years, a variety of novel findings reveal the molecular mechanisms for the CTS and its role in the development of atherosclerosis, thereby providing novel insights into the understanding of whole-body cholesterol transport and metabolism. In this review, we summarize the latest advances in this area with an emphasis on the therapeutic potential of targeting the CTS in CVD patients.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
20
|
Bourdi M, Amar M, Remaley AT, Terse PS. Intravenous toxicity and toxicokinetics of an HDL mimetic, Fx-5A peptide complex, in cynomolgus monkeys. Regul Toxicol Pharmacol 2018; 100:59-67. [PMID: 30359697 PMCID: PMC6893859 DOI: 10.1016/j.yrtph.2018.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 01/22/2023]
Abstract
Fx-5A peptide complex (Fx-5A), a High Density Lipoproteins (HDL) mimetic, has been shown to reduce atherosclerosis. The safety and toxicokinetics of Fx-5A administered IV by 30 min infusion at 8, 25 or 75 mg/kg body weight or vehicle, once every other day for 27 days, were assessed in cynomolgus monkeys. The Fx-5A was well tolerated at all doses. At the highest dose, there were statistically significant effects on hematology and clinical chemistry parameters that were considered non-adverse. Dose-dependent recoverable non-adverse erythrocytes morphological changes (acanthocytes, echinocytes, spherocytes, microcytes, and/or schistocytes) were observed. Fx-5A was not hemolytic in in-vitro fresh NHP or human blood assay. There were no Fx-5A-related statistically significant changes for any cardiovascular function, ECG or respiratory parameters, when compared to control. In addition, there were no Fx-5A-related effects on organ weights, macroscopic or microscopic endpoints. Finally, Fx-5A exhibited sporadic non-appreciable detection of anti-Fx-5A antibodies and a dose-dependent linear toxicokinetics with T1/2 value ranges from 2.7 to 6.2 h. In conclusion, the No Observed Adverse Effect Level was considered to be 75 mg/kg/day with associated exposures average Cmax and AUC0-last of 453 μg/mL and 2232 h μg/mL, respectively, on Day 27.
Collapse
Affiliation(s)
- Mohammed Bourdi
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, 20850, USA
| | - Marcelo Amar
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Alan T Remaley
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Pramod S Terse
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, 20850, USA.
| |
Collapse
|
21
|
Tsujita M, Wolska A, Gutmann DAP, Remaley AT. Reconstituted Discoidal High-Density Lipoproteins: Bioinspired Nanodiscs with Many Unexpected Applications. Curr Atheroscler Rep 2018; 20:59. [PMID: 30397748 DOI: 10.1007/s11883-018-0759-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE OF REVIEW Summarize the initial discovery of discoidal high-density lipoprotein (HDL) in human plasma and review more recent innovations that span the use of reconstituted nanodisc HDL for membrane protein characterization to its use as a drug carrier and a novel therapeutic agent for cardiovascular disease. RECENT FINDINGS Using a wide variety of biophysical techniques, the structure and composition of endogenous discoidal HDL have now largely been solved. This has led to the development of new methods for the in vitro reconstitution of nanodisc HDL, which have proven to have a wide variety of biomedical applications. Nanodisc HDL has been used as a platform for mimicking the plasma membrane for the reconstitution and investigation of the structures of several plasma membrane proteins, such as cytochrome P450s and ABC transporters. Nanodisc HDL has also been designed as drug carriers to transport amphipathic, as well as hydrophobic small molecules, and has potential therapeutic applications for several diseases. Finally, nanodisc HDL itself like native discoidal HDL can mediate cholesterol efflux from cells and are currently being tested in late-stage clinical trials for cardiovascular disease. The discovery of the characterization of native discoidal HDL has inspired a new field of synthetic nanodisc HDL, which has offered a growing number of unanticipated biomedical applications.
Collapse
Affiliation(s)
- Maki Tsujita
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
22
|
Current and Emerging Reconstituted HDL-apoA-I and HDL-apoE Approaches to Treat Atherosclerosis. J Pers Med 2018; 8:jpm8040034. [PMID: 30282955 PMCID: PMC6313318 DOI: 10.3390/jpm8040034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 01/14/2023] Open
Abstract
Atherosclerosis affects millions of people worldwide. However, the wide variety of limitations in the current therapeutic options leaves much to be desired in future lipid-lowering therapies. For example, although statins, which are the first-line treatment for coronary heart disease (CHD), reduce the risk of cardiovascular events in a large percentage of patients, they lead to optimal levels of low density lipoprotein-cholesterol (LDL-C) in only about one-third of patients. A new promising research direction against atherosclerosis aims to improve lipoprotein metabolism. Novel therapeutic approaches are being developed to increase the levels of functional high density lipoprotein (HDL) particles. This review aims to highlight the atheroprotective potential of the in vitro synthesized reconstituted HDL particles containing apolipoprotein E (apoE) as their sole apolipoprotein component (rHDL-apoE). For this purpose, we provide: (1) a summary of the atheroprotective properties of native plasma HDL and its apolipoprotein components, apolipoprotein A-I (apoA-I) and apoE; (2) an overview of the anti-atherogenic functions of rHDL-apoA-I and apoA-I-containing HDL, i.e., natural HDL isolated from transgenic Apoa1−/− × Apoe−/− mice overexpressing human apoA-I (HDL-apoA-I); and (3) the latest developments and therapeutic potential of HDL-apoE and rHDL-apoE. Novel rHDL formulations containing apoE could possibly present enhanced biological functions, leading to improved therapeutic efficacy against atherosclerosis.
Collapse
|
23
|
Mytilinaiou M, Kyrou I, Khan M, Grammatopoulos DK, Randeva HS. Familial Hypercholesterolemia: New Horizons for Diagnosis and Effective Management. Front Pharmacol 2018; 9:707. [PMID: 30050433 PMCID: PMC6052892 DOI: 10.3389/fphar.2018.00707] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022] Open
Abstract
Familial hypercholesterolemia (FH) is a common genetic cause of premature cardiovascular disease (CVD). The reported prevalence rates for both heterozygous FH (HeFH) and homozygous FH (HoFH) vary significantly, and this can be attributed, at least in part, to the variable diagnostic criteria used across different populations. Due to lack of consistent data, new global registries and unified guidelines are being formed, which are expected to advance current knowledge and improve the care of FH patients. This review presents a comprehensive overview of the pathophysiology, epidemiology, manifestations, and pharmacological treatment of FH, whilst summarizing the up-to-date relevant recommendations and guidelines. Ongoing research in FH seems promising and novel therapies are expected to be introduced in clinical practice in order to compliment or even substitute current treatment options, aiming for better lipid-lowering effects, fewer side effects, and improved clinical outcomes.
Collapse
Affiliation(s)
- Maria Mytilinaiou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom.,Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, United Kingdom.,Division of Translational and Experimental Medicine, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Centre of Applied Biological and Exercise Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, United Kingdom
| | - Mike Khan
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Dimitris K Grammatopoulos
- Division of Translational and Experimental Medicine, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Institute of Precision Diagnostics and Translational Medicine, Coventry and Warwickshire Pathology Service, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Harpal S Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom.,Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, United Kingdom.,Division of Translational and Experimental Medicine, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Centre of Applied Biological and Exercise Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, United Kingdom.,Institute of Precision Diagnostics and Translational Medicine, Coventry and Warwickshire Pathology Service, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| |
Collapse
|
24
|
Karalis I, Jukema JW. HDL Mimetics Infusion and Regression of Atherosclerosis: Is It Still Considered a Valid Therapeutic Option? Curr Cardiol Rep 2018; 20:66. [PMID: 29926215 PMCID: PMC6010501 DOI: 10.1007/s11886-018-1004-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose of Review This review aims to summarize and discuss the recent findings in the field of using HDL mimetics for the treatment of patients with coronary artery disease. Recent Findings Following the largely disappointing results with the cholesteryl ester transfer protein inhibitors, focus moved to HDL functionality rather than absolute HDL cholesterol values. A number of HDL/apoA-I mimicking molecules were developed, aiming to enhance reverse cholesterol transport that has been associated with an atheroprotective effect. Three HDL mimetics have made the step from bench-testing to clinical trials in humans and are discussed here: apoA-I Milano, CSL-112, and CER-001. Unfortunately, with the exception of CSL-112 where the results of the clinical trial are not yet known, none of the agents was able to demonstrate a clinical benefit. Summary HDL mimetics have failed to date to prove a beneficial effect in clinical practice. Reverse cholesterol transport remains a challenging therapeutic pathway to be explored.
Collapse
Affiliation(s)
- I Karalis
- Department of Cardiology C5-P, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Postbus 9600, 2300 RC, Leiden, The Netherlands
| | - J W Jukema
- Department of Cardiology C5-P, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Postbus 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
25
|
Abstract
High-density lipoproteins (HDLs) have presented an attractive target for development of new therapies for cardiovascular prevention on the basis of epidemiology and preclinical studies demonstrating their protective properties. Development of HDL mimetics provides an opportunity to administer functional HDL. However, clinical trials have produced variable results, with no evidence to date that they reduce cardiovascular events. This article reviews development programs of HDL mimetics.
Collapse
Affiliation(s)
- Kohei Takata
- South Australian Health and Medical Research Institute, University of Adelaide, PO Box 11060, Adelaide, SA 5001, Australia
| | - Belinda A Di Bartolo
- South Australian Health and Medical Research Institute, University of Adelaide, PO Box 11060, Adelaide, SA 5001, Australia
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, PO Box 11060, Adelaide, SA 5001, Australia.
| |
Collapse
|
26
|
Soran H, Adam S, Mohammad JB, Ho JH, Schofield JD, Kwok S, Siahmansur T, Liu Y, Syed AA, Dhage SS, Stefanutti C, Donn R, Malik RA, Banach M, Durrington PN. Hypercholesterolaemia - practical information for non-specialists. Arch Med Sci 2018; 14:1-21. [PMID: 29379528 PMCID: PMC5778427 DOI: 10.5114/aoms.2018.72238] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/03/2017] [Indexed: 12/21/2022] Open
Abstract
Hypercholesterolaemia is amongst the most common conditions encountered in the medical profession. It remains one of the key modifiable cardiovascular risk factors and there have been recent advances in the risk stratification methods and treatment options available. In this review, we provide a background into hypercholesterolaemia for non-specialists and consider the merits of the different risk assessment tools available. We also provide detailed considerations as to: i) when to start treatment, ii) what targets to aim for and iii) the role of low density lipoprotein cholesterol.
Collapse
Affiliation(s)
- Handrean Soran
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
- Cardiovascular Trials Unit, University Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Safwaan Adam
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
- Cardiovascular Trials Unit, University Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jamal B. Mohammad
- Department of Medicine, University of Duhok, Duhok, Kurdistan region, Iraq
| | - Jan H. Ho
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
- Cardiovascular Trials Unit, University Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jonathan D. Schofield
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
- Cardiovascular Trials Unit, University Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - See Kwok
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
- Cardiovascular Trials Unit, University Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Tarza Siahmansur
- Cardiovascular Trials Unit, University Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Yifen Liu
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
| | - Akheel A. Syed
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
- Department of Diabetes, Endocrinology and Obesity Medicine, Salford Royal NHS Foundation Trust, Manchester, UK
| | - Shaishav S. Dhage
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
- Cardiovascular Trials Unit, University Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Claudia Stefanutti
- Immunohematology and Transfusion Medicine, Department of Molecular Medicine, University of Rome, Rome, Italy
| | - Rachelle Donn
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
| | | | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Poland
| | - Paul N. Durrington
- Cardiovascular Research Group, Faculty of Biology, Medicine & Health, University of Manchester, UK
| |
Collapse
|
27
|
|
28
|
Abstract
Low-density lipoproteins (LDL) play a causal role in the development of atherosclerosis, and reduction of LDL cholesterol with a statin is a cornerstone in prevention of cardiovascular disease. However, it remains an unmet need to reduce the residual risk on maximally tolerated statin alone or in combination with other drugs such as ezetimibe. Among the new LDL-lowering therapies, PCSK9 inhibitors appear the most promising class. Genetic studies suggest that triglyceride-rich lipoproteins are associated with cardiovascular risk and several promising triglyceride-lowering therapies are at various stages of development. At the opposite end, high-density lipoprotein (HDL) cholesterol seems to not be causally associated with cardiovascular risk, and thus far, trials designed to reduce cardiovascular risk by mainly raising HDL cholesterol levels have been disappointing. Nevertheless, new drugs targeting HDL are still in development. This review describes the new drugs reducing LDL, apolipoprotein(a), and triglyceride-rich lipoproteins, and the strategies to modulate HDL metabolism.
Collapse
|
29
|
Badimon L, Padró T, Cubedo J. Protein changes in non-LDL-lipoproteins in familial hypercholesterolemia: implications in cardiovascular disease manifestation and outcome. Curr Opin Lipidol 2017; 28:427-433. [PMID: 28682808 DOI: 10.1097/mol.0000000000000441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Familial hypercholesterolemia, represents one of the most extreme clinical entities associated with premature coronary artery disease (CAD). However, clinical manifestation of CAD varies across cohorts and individual patients suggesting the existence of additional non-LDL factors potentially contributing to their cardiovascular burden. RECENT FINDINGS Changes in HDL-associated proteins appear as one of the potential additional factors contributing to the cardiovascular risk in familial hypercholesterolemia. Specifically, the content of Apo M-SP1 in HDL3 has been directly associated with cholesterol efflux capacity. In addition, a coordinated decrease in the content of Apo L1 and LCAT in HDL3 has been related to the presence of corneal arcus and to bad prognosis in familial hypercholesterolemia patients after an acute ischemic event. In fact, HDL3 particles of familial hypercholesterolemia patients have diminished antioxidant and anti-inflammatory function. SUMMARY The identification of the specific changes in HDL-associated proteins that contribute to the increased cardiovascular risk of familial hypercholesterolemia patients could be useful for the development of novel therapeutic targets. These novel strategies, in combination with current lipid-lowering therapies, may help to reduce the residual risk found in these patients.
Collapse
Affiliation(s)
- Lina Badimon
- aCardiovascular Science Institute - ICCC, Biomedical Research Institute Sant Pau (IIB-Sant Pau) and CiberCV bCardiovascular Research Chair UAB, Barcelona, Spain
| | | | | |
Collapse
|
30
|
Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Mechanisms of foam cell formation in atherosclerosis. J Mol Med (Berl) 2017; 95:1153-1165. [DOI: 10.1007/s00109-017-1575-8] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/04/2017] [Accepted: 07/28/2017] [Indexed: 12/21/2022]
|
31
|
Toward an international consensus—Integrating lipoprotein apheresis and new lipid-lowering drugs. J Clin Lipidol 2017; 11:858-871.e3. [DOI: 10.1016/j.jacl.2017.04.114] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/30/2017] [Accepted: 04/15/2017] [Indexed: 12/11/2022]
|
32
|
Lipid Metabolism and Emerging Targets for Lipid-Lowering Therapy. Can J Cardiol 2017; 33:872-882. [DOI: 10.1016/j.cjca.2016.12.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/12/2016] [Accepted: 12/26/2016] [Indexed: 12/25/2022] Open
|
33
|
Smolders L, Plat J, Mensink RP. Dietary Strategies and Novel Pharmaceutical Approaches Targeting Serum ApoA-I Metabolism: A Systematic Overview. J Nutr Metab 2017; 2017:5415921. [PMID: 28695008 PMCID: PMC5485365 DOI: 10.1155/2017/5415921] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/16/2017] [Indexed: 12/19/2022] Open
Abstract
The incidence of CHD is still increasing, which underscores the need for new preventive and therapeutic approaches to decrease CHD risk. In this respect, increasing apoA-I concentrations may be a promising approach, especially through increasing apoA-I synthesis. This review first provides insight into current knowledge on apoA-I production, clearance, and degradation, followed by a systematic review of dietary and novel pharmacological approaches to target apoA-I metabolism. For this, a systematic search was performed to identify randomized controlled intervention studies that examined effects of whole foods and (non)nutrients on apoA-I metabolism. In addition, novel pharmacological approaches were searched for, which were specifically developed to target apoA-I metabolism. We conclude that both dietary components and pharmacological approaches can be used to increase apoA-I concentrations or functionality. For the dietary components in particular, more knowledge about the underlying mechanisms is necessary, as increasing apoA-I per se does not necessarily translate into a reduced CHD risk.
Collapse
Affiliation(s)
- Lotte Smolders
- Department of Human Biology and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, Netherlands
| | - Jogchum Plat
- Department of Human Biology and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, Netherlands
| | - Ronald P. Mensink
- Department of Human Biology and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, P.O. Box 616, 6200 MD Maastricht, Netherlands
| |
Collapse
|
34
|
Abstract
Background CER-001 comprises recombinant human apolipoprotein A-I complexed with phospholipids that mimics natural, nascent, pre-β high-density lipoprotein (HDL). We present animal model data showing dose-dependent increases in cholesterol efflux with CER-001 and its subsequent elimination by reverse lipid transport, together with inhibition of atherosclerotic plaque progression. We report the first phase I study results with CER-001 in humans, starting at 0.25 mg/kg, which is 1/80th of the safe dose (20 mg/kg) established in 4-week multiple-dose animal studies dosed every second day. Methods Healthy volunteers, 18–55 years old with a low-density lipoprotein-cholesterol:HDL-cholesterol ratio greater than 3.0, received single intravenous escalating doses of CER-001 (0.25–45.0 mg/kg) and placebo in a double-blind randomised cross-over fashion. Subjects were followed up for 3 weeks post-dose. Assessments included adverse event monitoring, blood sampling, and clinical laboratory measurements. Results Thirty-two subjects were enrolled. All CER-001 doses (0.25–45 mg/kg) were safe and well tolerated, with an adverse event profile similar to placebo. Effects on clinical chemistry, haematology and coagulation parameters were comparable to placebo. No adverse effects of CER-001 on electrocardiograms were observed. No antibodies to apolipoprotein A-I were detected following single-dose administration of CER-001. Plasma apolipoprotein A-I levels increased in a dose-related manner and returned to baseline by 24 h post-dose for doses up to 10 mg/kg but remained in circulation for >72 h post-dose for doses >10 mg/kg. CER-001 caused elevations in plasma cholesterol and total and unesterified cholesterol in the HDL fraction. Mobilisation of unesterified cholesterol in the HDL fraction was seen with CER-001 at doses as low as 2 mg/kg. Conclusion CER-001 is well tolerated when administered to humans as single doses up to 45 mg/kg and mobilises and eliminates cholesterol via reverse lipid transport. Electronic supplementary material The online version of this article (doi:10.1007/s40261-017-0506-3) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
New Drugs for Atherosclerosis. Can J Cardiol 2017; 33:350-357. [DOI: 10.1016/j.cjca.2016.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 12/18/2022] Open
|
36
|
Andrews J, Janssan A, Nguyen T, Pisaniello AD, Scherer DJ, Kastelein JJP, Merkely B, Nissen SE, Ray K, Schwartz GG, Worthley SG, Keyserling C, Dasseux JL, Butters J, Girardi J, Miller R, Nicholls SJ. Effect of serial infusions of reconstituted high-density lipoprotein (CER-001) on coronary atherosclerosis: rationale and design of the CARAT study. Cardiovasc Diagn Ther 2017; 7:45-51. [PMID: 28164012 DOI: 10.21037/cdt.2017.01.01] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND High-density lipoprotein (HDL) is believed to have atheroprotective properties, but an effective HDL-based therapy remains elusive. Early studies have suggested that infusion of reconstituted HDL promotes reverse cholesterol transport and vascular reactivity. The CER-001 Atherosclerosis Regression Acute Coronary Syndrome Trial (CARAT) is investigating the impact of infusing an engineered pre-beta HDL mimetic containing sphingomyelin (SM) and dipalmitoyl phosphatidlyglycerol (CER-001) on coronary atheroma volume in patients with a recent acute coronary syndrome (ACS). METHODS The CARAT is a phase 2, multicenter trial in which 292 patients with an ACS undergoing intracoronary ultrasonography and showing percent atheroma volume (PAV) greater than 30% are randomly assigned to treatment with ten infusions of CER-001 3 mg/kg or matching placebo, administered at weekly intervals. Intracoronary ultrasonography is repeated at the end of the treatment period. RESULTS The primary endpoint is the nominal change in PAV. Safety and tolerability will also be evaluated. CONCLUSIONS CARAT will establish whether serial 3 mg/kg infusions of an engineered pre-beta HDL mimetic containing SM and dipalmitoyl phosphatidlyglycerol (CER-001) will regress atherosclerotic plaque in patients with a recent ACS.
Collapse
Affiliation(s)
- Jordan Andrews
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Alex Janssan
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Tracy Nguyen
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Anthony D Pisaniello
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Daniel J Scherer
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - John J P Kastelein
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | | | - Kausik Ray
- School of Public Health, Imperial College London, London, UK
| | | | - Stephen G Worthley
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | | | | | - Julie Butters
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Jacinta Girardi
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Rosemary Miller
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
37
|
Zheng KH, Stroes ESG. HDL infusion for the management of atherosclerosis: current developments and new directions. Curr Opin Lipidol 2016; 27:592-596. [PMID: 27653220 DOI: 10.1097/mol.0000000000000349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Because human genetic studies and large clinical trials have demonstrated that HDL-cholesterol levels are not causally related to cardiovascular disease risk, attention has shifted toward the functional properties of HDL. Infusion of HDL mimetics containing apolipoprotein A-I remains a potential strategy to exploit the atheroprotective effects of HDL. RECENT FINDINGS Three HDL mimetic drugs are under development and currently being evaluated in clinical trials. Upon infusion, these drugs increase cholesterol efflux capacity. Although proof-of-concept studies are promising, large outcome studies are awaited. Alternatively, HDL particles may be used for targeted drug delivery in a nanomedicine approach. Finally, links between cholesterol efflux and myelopoeisis may prove to be a target for HDL infusion in the future. SUMMARY Clinical studies are currently ongoing to evaluate the potential of several HDL mimetic drugs. Novel nanomedicinal approaches and emerging pathophysiological insights may further expand the relevance of HDL infusion.
Collapse
Affiliation(s)
- Kang H Zheng
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
38
|
Thaxton CS, Rink JS, Naha PC, Cormode DP. Lipoproteins and lipoprotein mimetics for imaging and drug delivery. Adv Drug Deliv Rev 2016; 106:116-131. [PMID: 27133387 PMCID: PMC5086317 DOI: 10.1016/j.addr.2016.04.020] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/02/2016] [Accepted: 04/19/2016] [Indexed: 12/22/2022]
Abstract
Lipoproteins are a set of natural nanoparticles whose main role is the transport of fats within the body. While much work has been done to develop synthetic nanocarriers to deliver drugs or contrast media, natural nanoparticles such as lipoproteins represent appealing alternatives. Lipoproteins are biocompatible, biodegradable, non-immunogenic and are naturally targeted to some disease sites. Lipoproteins can be modified to act as contrast agents in many ways, such as by insertion of gold cores to provide contrast for computed tomography. They can be loaded with drugs, nucleic acids, photosensitizers or boron to act as therapeutics. Attachment of ligands can re-route lipoproteins to new targets. These attributes render lipoproteins attractive and versatile delivery vehicles. In this review we will provide background on lipoproteins, then survey their roles as contrast agents, in drug and nucleic acid delivery, as well as in photodynamic therapy and boron neutron capture therapy.
Collapse
Affiliation(s)
- C Shad Thaxton
- Department of Urology, Northwestern University, Chicago, IL, USA; Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, USA; International Institute for Nanotechnology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Jonathan S Rink
- Department of Urology, Northwestern University, Chicago, IL, USA; Simpson Querrey Institute for Bionanotechnology, Northwestern University, Chicago, IL, USA
| | - Pratap C Naha
- Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA
| | - David P Cormode
- Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA; Department of Cardiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Affiliation(s)
- Vinaya Simha
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| | - Yogish C Kudva
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| |
Collapse
|
40
|
Zheng KH, van der Valk FM, Smits LP, Sandberg M, Dasseux JL, Baron R, Barbaras R, Keyserling C, Coolen BF, Nederveen AJ, Verberne HJ, Nell TE, Vugts DJ, Duivenvoorden R, Fayad ZA, Mulder WJ, van Dongen GA, Stroes ES. HDL mimetic CER-001 targets atherosclerotic plaques in patients. Atherosclerosis 2016; 251:381-388. [DOI: 10.1016/j.atherosclerosis.2016.05.038] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/09/2016] [Accepted: 05/25/2016] [Indexed: 12/17/2022]
|
41
|
Raggi P, Baldassarre D, Day S, de Groot E, Fayad Z. Non-invasive imaging of atherosclerosis regression with magnetic resonance to guide drug development. Atherosclerosis 2016; 251:476-482. [DOI: 10.1016/j.atherosclerosis.2016.06.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022]
|
42
|
Annema W, von Eckardstein A. Dysfunctional high-density lipoproteins in coronary heart disease: implications for diagnostics and therapy. Transl Res 2016; 173:30-57. [PMID: 26972566 DOI: 10.1016/j.trsl.2016.02.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
Low plasma levels of high-density lipoprotein (HDL) cholesterol are associated with increased risks of coronary heart disease. HDL mediates cholesterol efflux from macrophages for reverse transport to the liver and elicits many anti-inflammatory and anti-oxidative activities which are potentially anti-atherogenic. Nevertheless, HDL has not been successfully targeted by drugs for prevention or treatment of cardiovascular diseases. One potential reason is the targeting of HDL cholesterol which does not capture the structural and functional complexity of HDL particles. Hundreds of lipid species and dozens of proteins as well as several microRNAs have been identified in HDL. This physiological heterogeneity is further increased in pathologic conditions due to additional quantitative and qualitative molecular changes of HDL components which have been associated with both loss of physiological function and gain of pathologic dysfunction. This structural and functional complexity of HDL has prevented clear assignments of molecules to the functions of normal HDL and dysfunctions of pathologic HDL. Systematic analyses of structure-function relationships of HDL-associated molecules and their modifications are needed to test the different components and functions of HDL for their relative contribution in the pathogenesis of atherosclerosis. The derived biomarkers and targets may eventually help to exploit HDL for treatment and diagnostics of cardiovascular diseases.
Collapse
Affiliation(s)
- Wijtske Annema
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
43
|
Abstract
The introduction of statins ≈ 30 years ago ushered in the era of lipid lowering as the most effective way to reduce risk of atherosclerotic cardiovascular disease. Nonetheless, residual risk remains high, and statin intolerance is frequently encountered in clinical practice. After a long dry period, the field of therapeutics targeted to lipids and atherosclerosis has entered a renaissance. Moreover, the demonstration of clinical benefits from the addition of ezetimibe to statin therapy in subjects with acute coronary syndromes has renewed the enthusiasm for the cholesterol hypothesis and the hope that additional agents that lower low-density lipoprotein will decrease risk of atherosclerotic cardiovascular disease. Drugs in the orphan disease category are now available for patients with the most extreme hypercholesterolemia. Furthermore, discovery and rapid translation of a novel biological pathway has given rise to a new class of cholesterol-lowering drugs, the proprotein convertase subtilisin kexin-9 inhibitors. Trials of niacin added to statin have failed to demonstrate cardiac benefits, and 3 cholesterol ester transfer protein inhibitors have also failed to reduce atherosclerotic cardiovascular disease risk, despite producing substantial increases in HDL levels. Although the utility of triglyceride-lowering therapies remains uncertain, 2 large clinical trials are testing the influence of omega-3 polyunsaturated fatty acids on atherosclerotic events in hypertriglyceridemia. Novel antisense therapies targeting apolipoprotein C-III (for triglyceride reduction) and apo(a) (for lipoprotein(a) reduction) are showing a promising trajectory. Finally, 2 large clinical trials are formally putting the inflammatory hypothesis of atherosclerosis to the test and may open a new avenue for cardiovascular disease risk reduction.
Collapse
Affiliation(s)
- Michael D Shapiro
- From the Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR
| | - Sergio Fazio
- From the Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health & Science University, Portland, OR.
| |
Collapse
|
44
|
Abstract
Several recent reports have raised doubts about the atheroprotective role of high-density lipoprotein cholesterol (HDL-C). Nevertheless, a substantial body of work supports the validity of pharmacological interventions able to enhance HDL function, as opposed to raising HDL-C levels per se. In this article, we briefly review the development of pharmacological interventions that target apoA-I and HDL function as a means of reducing atherosclerotic risk: small molecule pharmaceuticals, small HDL mimetic peptides, and infusion of apoA-I-containing particles.
Collapse
|
45
|
Qu B, Qu T, Liu Y, Jia Y, Han X, Su J, Wang H, Liu L, Wang Z, Wang Y, Pan J, Ren G. Risk Factors Associated With Increased Carotid Intima-Media Thickness in a Male Population With Chronic Alcohol Consumption: A Prospective Observational Study. Medicine (Baltimore) 2016; 95:e3322. [PMID: 27082578 PMCID: PMC4839822 DOI: 10.1097/md.0000000000003322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Previous studies have reported a relationship between alcohol consumption and carotid intima-media thickness (CIMT). However, the exact associations between different severities of CIMT and dyslipidemia, dyslipoproteinemia, inflammatory immune markers, and oxidative markers associated with chronic alcohol consumption remain unknown. The aim of this study was to explore whether there are associations between different severities of CIMT and dyslipidemia, dyslipoproteinemia, inflammatory immune markers, and oxidative markers associated with chronic alcohol consumption. We enrolled 173 males with chronic alcohol consumption and categorized them into 2 groups: 104 chronic alcohol consumers with normal CIMT (group A) and 69 chronic alcohol consumers with increased CIMT (group B). Nonparametric statistics showed that age, body mass index (BMI), and serum TC, TG, Apo A1, and ApoB levels were significantly higher in group B than in group A (P = 0.002, 0.019, 0.021, 0.023, 0.001, and 0.001, respectively). Additionally, tumor necrosis factor alpha (TNFα) and HSP70 serum levels were significantly lower in group B than in group A (P = 0.023 and 0.017, respectively). A binary logistic regression analysis showed that age (OR: 1.077, 95% CI: 1.024-1.13, P = 0.004), ApoB (OR: 6.828, 95% CI: 1.506-30.956, P = 0.013), and TNF-α (OR: 0.999, 95% CI: 0.998-1.00) were independent risk factors associated with CIMT. The present study demonstrated that age, ApoB, and TNFα are independent risk factors associated with CIMT. Thus, older subjects with increased serum ApoB levels are more likely to present with increased CIMT, suggesting that age and ApoB promote such thickening and that TNFα downregulation might play a protective role against the progression of subclinical atherosclerosis in subjects with chronic alcohol consumption.
Collapse
Affiliation(s)
- Baoge Qu
- From the Department of Internal Medicine, Taishan Hospital of Shandong Province; Taishan Medical College, Taian, Shandong (BQ, YL, YJ, XH, JS, HW, LL, ZW, YW, JP, GR); and Zhuhai Campus of Zunyi Medical College, Zhuhai, Guangdong, China (TQ)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Novel therapies for severe dyslipidemia target a wide range of unmet medical needs: severe familial hypercholesterolemia, severe hypertriglyceridemia and chylomicronemia, elevated lipoprotein (a), lipodystrophies, high-density lipoprotein particle diseases, lysosomal acid lipase deficiency and storage diseases, nonalcoholic fatty liver disease and others. The purpose of this review is to describe the contribution of human genetics to the development of therapeutic approaches targeting severe dyslipidemia. RECENT FINDINGS Recent advances in human genetics and the identification of rare genetic variants having strong effects on disease risk not only accelerated the development of therapies for severe dyslipidemia, they also revealed new pathways, genes and mechanisms of health, disease or drug response, and facilitated molecular diagnosis, which may prove essential as the authorized use of some of these novel drugs is limited to specific conditions. In addition, the dissection of the gene and cell machinery gave rise to new technologies, gene-based therapies and biodrugs covering a broad range of novel agents currently available or in clinical development to treat severe lipid disorders. SUMMARY Several novel therapies are recently available or under development to treat severe dyslipidemia and associated risk stem directly from genetic research. Altogether, these therapies target a broad variety of severe dyslipidemia pathways or mechanisms and illustrate that clinical lipidology has now entered the era of precision medicine.
Collapse
Affiliation(s)
- Daniel Gaudet
- Department of Medicine, Lipidology Unit, Community Genomic Medicine Center, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada
| |
Collapse
|
47
|
Cardiovascular risk reduction: the future of cholesterol lowering drugs. Curr Opin Pharmacol 2016; 27:62-9. [DOI: 10.1016/j.coph.2016.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 11/21/2022]
|
48
|
Kuai R, Li D, Chen YE, Moon JJ, Schwendeman A. High-Density Lipoproteins: Nature's Multifunctional Nanoparticles. ACS NANO 2016; 10:3015-41. [PMID: 26889958 PMCID: PMC4918468 DOI: 10.1021/acsnano.5b07522] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
High-density lipoproteins (HDL) are endogenous nanoparticles involved in the transport and metabolism of cholesterol, phospholipids, and triglycerides. HDL is well-known as the "good" cholesterol because it not only removes excess cholesterol from atherosclerotic plaques but also has anti-inflammatory and antioxidative properties, which protect the cardiovascular system. Circulating HDL also transports endogenous proteins, vitamins, hormones, and microRNA to various organs. Compared with other synthetic nanocarriers, such as liposomes, micelles, and inorganic and polymeric nanoparticles, HDL has unique features that allow them to deliver cargo to specific targets more efficiently. These attributes include their ultrasmall size (8-12 nm in diameter), high tolerability in humans (up to 8 g of protein per infusion), long circulating half-life (12-24 h), and intrinsic targeting properties to different recipient cells. Various recombinant ApoA proteins and ApoA mimetic peptides have been recently developed for the preparation of reconstituted HDL that exhibits properties similar to those of endogenous HDL and has a potential for industrial scale-up. In this review, we will summarize (a) clinical pharmacokinetics and safety of reconstituted HDL products, (b) comparison of HDL with inorganic and other organic nanoparticles,
Collapse
Affiliation(s)
- Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Y. Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence should be addressed to A. S. () or J.J.M. ()
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence should be addressed to A. S. () or J.J.M. ()
| |
Collapse
|
49
|
Darabi M, Guillas-Baudouin I, Le Goff W, Chapman MJ, Kontush A. Therapeutic applications of reconstituted HDL: When structure meets function. Pharmacol Ther 2015; 157:28-42. [PMID: 26546991 DOI: 10.1016/j.pharmthera.2015.10.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Reconstituted forms of HDL (rHDL) are under development for infusion as a therapeutic approach to attenuate atherosclerotic vascular disease and to reduce cardiovascular risk following acute coronary syndrome and ischemic stroke. Currently available rHDL formulations developed for clinical use contain apolipoprotein A-I (apoA-I) and one of the major lipid components of HDL, either phosphatidylcholine or sphingomyelin. Recent data have established that quantitatively minor molecular constituents of HDL particles can strongly influence their anti-atherogenic functionality. Novel rHDL formulations displaying enhanced biological activities, including cellular cholesterol efflux, may therefore offer promising prospects for the development of HDL-based, anti-atherosclerotic therapies. Indeed, recent structural and functional data identify phosphatidylserine as a bioactive component of HDL; the content of phosphatidylserine in HDL particles displays positive correlations with all metrics of their functionality. This review summarizes current knowledge of structure-function relationships in rHDL formulations, with a focus on phosphatidylserine and other negatively-charged phospholipids. Mechanisms potentially underlying the atheroprotective role of these lipids are discussed and their potential for the development of HDL-based therapies highlighted.
Collapse
Affiliation(s)
- Maryam Darabi
- UMR INSERM-UPMC 1166 ICAN, Pavillon Benjamin Delessert, Hôpital de la Pitié, 83 boulevard de l'Hôpital, 75651 Paris Cedex 13, France.
| | - Isabelle Guillas-Baudouin
- UMR INSERM-UPMC 1166 ICAN, Pavillon Benjamin Delessert, Hôpital de la Pitié, 83 boulevard de l'Hôpital, 75651 Paris Cedex 13, France.
| | - Wilfried Le Goff
- UMR INSERM-UPMC 1166 ICAN, Pavillon Benjamin Delessert, Hôpital de la Pitié, 83 boulevard de l'Hôpital, 75651 Paris Cedex 13, France.
| | - M John Chapman
- UMR INSERM-UPMC 1166 ICAN, Pavillon Benjamin Delessert, Hôpital de la Pitié, 83 boulevard de l'Hôpital, 75651 Paris Cedex 13, France.
| | - Anatol Kontush
- UMR INSERM-UPMC 1166 ICAN, Pavillon Benjamin Delessert, Hôpital de la Pitié, 83 boulevard de l'Hôpital, 75651 Paris Cedex 13, France.
| |
Collapse
|
50
|
Lacko AG, Sabnis NA, Nagarajan B, McConathy WJ. HDL as a drug and nucleic acid delivery vehicle. Front Pharmacol 2015; 6:247. [PMID: 26578957 PMCID: PMC4620406 DOI: 10.3389/fphar.2015.00247] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/12/2015] [Indexed: 01/13/2023] Open
Abstract
This review is intended to evaluate the research findings and potential clinical applications of drug transport systems, developed based on the concepts of the structure/function and physiological role(s) of high density lipoprotein type nanoparticles. These macromolecules provide targeted transport of cholesteryl esters (a highly lipophilic payload) in their natural/physiological environment. The ability to accommodate highly water insoluble constituents in their core regions enables High density lipoproteins (HDL) type nanoparticles to effectively transport hydrophobic drugs subsequent to systemic administration. Even though the application of reconstituted HDL in the treatment of a number of diseases is reviewed, the primary focus is on the application of HDL type drug delivery agents in cancer chemotherapy. The use of both native and synthetic HDL as drug delivery agents is compared to evaluate their respective potentials for commercial and clinical development. The current status and future perspectives for HDL type nanoparticles are discussed, including current obstacles and future applications in therapeutics.
Collapse
Affiliation(s)
- Andras G Lacko
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, TX, USA ; Department of Pediatrics, University of North Texas Health Science Center , Fort Worth, TX, USA
| | - Nirupama A Sabnis
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, TX, USA ; Department of Pediatrics, University of North Texas Health Science Center , Fort Worth, TX, USA
| | - Bhavani Nagarajan
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, TX, USA
| | | |
Collapse
|