51
|
Ramin-Mangata S, Thedrez A, Nativel B, Diotel N, Blanchard V, Wargny M, Aguesse A, Billon-Crossouard S, Vindis C, Le May C, Hulin P, Armanet M, Gmyr V, Pattou F, Croyal M, Meilhac O, Nobécourt E, Cariou B, Lambert G. Effects of proprotein convertase subtilisin kexin type 9 modulation in human pancreatic beta cells function. Atherosclerosis 2021; 326:47-55. [PMID: 33933263 DOI: 10.1016/j.atherosclerosis.2021.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/09/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND AIMS Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9) is an endogenous inhibitor of the LDL receptor (LDLR). Mendelian randomization studies suggest that PCSK9 deficiency increases diabetes risk, but the underlying mechanisms remain unknown. The aim of our study was to investigate whether PCSK9 or its inhibition may modulate beta cell function. METHODS We assessed PCSK9 and insulin colocalization in human pancreatic sections by epifluorescent and confocal microscopy. We also investigated the expression and the function of PCSK9 in the human EndoC-βH1 beta cell line, by ELISA and flow cytometry, respectively. PCSK9 was inhibited with Alirocumab or siRNA. LDLR expression and LDL uptake were assessed by flow cytometry. RESULTS PCSK9 was expressed and secreted from beta cells isolated from human pancreas as well as from EndoC-βH1 cells. PCSK9 secretion was enhanced by statin treatment. Recombinant PCSK9 decreased LDLR abundance at the surface of these cells, an effect abrogated by Alirocumab. Alirocumab as well as PCSK9 silencing increased LDLR expression at the surface of EndoC-βH1 cells. Neither exogenous PCSK9, nor Alirocumab, nor PCSK9 silencing significantly altered glucose-stimulated insulin secretion (GSIS) from these cells. High-low density lipoproteins (LDL) concentrations decreased GSIS, but the addition of PCSK9 or its inhibition did not modulate this phenomenon. CONCLUSIONS While PCSK9 regulates LDLR abundance in beta cells, inhibition of exogenous or endogenous PCSK9 does not appear to significantly impact insulin secretion. This is reassuring for the safety of PCSK9 inhibitors in terms of beta cell function.
Collapse
Affiliation(s)
| | - Aurélie Thedrez
- Université de Nantes, CRNH Ouest, Inra UMR 1280 PhAN, Nantes, France; L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Brice Nativel
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Nicolas Diotel
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Valentin Blanchard
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Matthieu Wargny
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France; CHU Nantes, INSERM, CIC 1413, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des Données, Nantes, F-44093, France
| | - Audrey Aguesse
- Université de Nantes, CRNH Ouest, Inra UMR 1280 PhAN, Nantes, France
| | | | | | - Cédric Le May
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Philippe Hulin
- Université de Nantes, CHU de Nantes, Inserm UMS 016, Cnrs UMS 3556, Structure Fédérative de Recherche François Bonamy, Micropicell Facility, Nantes, France
| | - Mathieu Armanet
- Cell Therapy Unit, Hôpital Saint Louis, AP-HP, Université Paris Diderot, Paris, France
| | - Valery Gmyr
- European Genomic Institute for Diabetes, Inserm UMR 1190 Translational Research for Diabetes, University of Lille 2, Lille, France
| | - François Pattou
- European Genomic Institute for Diabetes, Inserm UMR 1190 Translational Research for Diabetes, University of Lille 2, Lille, France; Lille University Hospital, Lille, France
| | - Mikaël Croyal
- Université de Nantes, CRNH Ouest, Inra UMR 1280 PhAN, Nantes, France
| | - Olivier Meilhac
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France
| | - Estelle Nobécourt
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France; CHU de La Réunion, Service d'Endocrinologie Nutrition, Saint-Pierre, France
| | - Bertrand Cariou
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Gilles Lambert
- Université de La Réunion, Inserm UMR 1188 DéTROI, Sainte Clotilde, France.
| |
Collapse
|
52
|
Nohara A, Tada H, Ogura M, Okazaki S, Ono K, Shimano H, Daida H, Dobashi K, Hayashi T, Hori M, Matsuki K, Minamino T, Yokoyama S, Harada-Shiba M. Homozygous Familial Hypercholesterolemia. J Atheroscler Thromb 2021; 28:665-678. [PMID: 33867421 PMCID: PMC8265428 DOI: 10.5551/jat.rv17050] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an inherited disorder with retarded clearance of plasma LDL caused by mutations of the genes involved in the LDL receptor-mediated pathway and most of them exhibit autosomal dominant inheritance. Homozygotes of FH (HoFH) may have plasma LDL-C levels, which are at least twice as high as those of heterozygous FH (HeFH) and therefore four times higher than normal levels. Prevalence of HoFH had been estimated as 1 in 1,000,000 before but more recent genetic analysis surveys predict 1 in 170,000 to 300,000. Since LDL receptor activity is severely impaired, HoFH patients do not or very poorly respond to medications to enhance activity, such as statins, and have a poorer prognosis compared to HeFH. HoFH should therefore be clinically distinguished from HeFH. Thorough family studies and genetic analysis are recommended for their accurate diagnosis. Fatal cardiovascular complications could develop even in the first decade of life for HoFH, so aggressive lipid-lowering therapy should be initiated as early as possible. Direct removal of plasma LDL by lipoprotein apheresis has been the principal measure for these patients. However, this treatment alone may not achieve stable LDL-C target levels and combination with drugs should be considered. The lipid-lowering effects of statins and PCSK9 inhibitors substantially vary depending on the remaining LDL receptor activity of individual patients. On the other hand, the action an MTP inhibitor is independent of LDL receptor activity, and it is effective in most HoFH cases. This review summarizes the key clinical issues of HoFH as well as insurance coverage available under the Japanese public healthcare system.
Collapse
Affiliation(s)
- Atsushi Nohara
- Department of Clinical Genetics, Ishikawa Prefectural Central Hospital
| | - Hayato Tada
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences
| | - Masatsune Ogura
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| | - Sachiko Okazaki
- Division for Health Service Promotion, The University of Tokyo
| | - Koh Ono
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine University of Tsukuba
| | - Hiroyuki Daida
- Faculty of Health Science, Juntendo University, Juntendo University Graduate School of Medicine
| | - Kazushige Dobashi
- Department of Pediatrics, School of Medicine, University of Yamanashi
| | - Toshio Hayashi
- School of Health Sciences, Nagoya University Graduate School of Medicine
| | - Mika Hori
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University
| | - Kota Matsuki
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University
| | | | - Mariko Harada-Shiba
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute
| |
Collapse
|
53
|
Chemello K, García-Nafría J, Gallo A, Martín C, Lambert G, Blom D. Lipoprotein metabolism in familial hypercholesterolemia. J Lipid Res 2021; 62:100062. [PMID: 33675717 PMCID: PMC8050012 DOI: 10.1016/j.jlr.2021.100062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 02/06/2023] Open
Abstract
Familial hypercholesterolemia (FH) is one of the most common genetic disorders in humans. It is an extremely atherogenic metabolic disorder characterized by lifelong elevations of circulating LDL-C levels often leading to premature cardiovascular events. In this review, we discuss the clinical phenotypes of heterozygous and homozygous FH, the genetic variants in four genes (LDLR/APOB/PCSK9/LDLRAP1) underpinning the FH phenotype as well as the most recent in vitro experimental approaches used to investigate molecular defects affecting the LDL receptor pathway. In addition, we review perturbations in the metabolism of lipoproteins other than LDL in FH, with a major focus on lipoprotein (a). Finally, we discuss the mode of action and efficacy of many of the currently approved hypocholesterolemic agents used to treat patients with FH, with a special emphasis on the treatment of phenotypically more severe forms of FH.
Collapse
Affiliation(s)
- Kévin Chemello
- Inserm UMR 1188 DéTROI, Université de La Réunion, Saint- Denis de La Réunion, France
| | - Javier García-Nafría
- Institute for Biocomputation and Physics of complex systems (BIFI), University of Zaragoza, Zaragoza, Spain; Laboratorio de Microscopías Avanzadas, University of Zaragoza, Zaragoza, Spain
| | - Antonio Gallo
- Cardiovascular Prevention Unit, Department of Endocrinology and Metabolism, Pitié-Salpêtrière University Hospital, Paris, France; Laboratoire d'imagerie Biomédicale, INSERM 1146, CNRS 7371, Sorbonne University, Paris, France
| | - Cesar Martín
- Instituto Biofisika (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco UPV/EHU, Bilbao, Spain
| | - Gilles Lambert
- Inserm UMR 1188 DéTROI, Université de La Réunion, Saint- Denis de La Réunion, France.
| | - Dirk Blom
- Hatter Institute for Cardiovascular Research in Africa and Division of Lipidology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
54
|
Calcaterra I, Buonaiuto A, Iannuzzo G, Di Minno MND. Authors' response to letter by Sbrana et al. "Evolocumab improve intima media thickness regression in He-FH subjects on lipoprotein apheresis". Nutr Metab Cardiovasc Dis 2021; 31:361-362. [PMID: 33257188 DOI: 10.1016/j.numecd.2020.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/20/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Ilenia Calcaterra
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Italy
| | - Alessio Buonaiuto
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Italy
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Italy
| | - Matteo N D Di Minno
- Department of Translational Medical Sciences, Federico II University of Naples, Italy.
| |
Collapse
|
55
|
Huijgen R, Blom DJ, Hartgers ML, Chemello K, Benito-Vicente A, Uribe KB, Behardien Z, Blackhurst DM, Brice BC, Defesche JC, de Jong AG, Jooste RJ, Solomon GAE, Wolmarans KH, Hovingh GK, Martin C, Lambert G, Marais AD. Novel PCSK9 (Proprotein Convertase Subtilisin Kexin Type 9) Variants in Patients With Familial Hypercholesterolemia From Cape Town. Arterioscler Thromb Vasc Biol 2020; 41:934-943. [PMID: 33147992 DOI: 10.1161/atvbaha.120.314482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Familial hypercholesterolemia (FH) is characterized by elevated low-density lipoprotein-cholesterol and markedly increased cardiovascular risk. In patients with a genetic diagnosis, low-density lipoprotein receptor (LDLR) mutations account for >90% of cases, apolipoprotein B (APOB) mutations for ≈5% of cases, while proprotein convertase subtilisin kexin type 9 (PCSK9) gain of function mutations are rare (<1% of cases). We aimed to evaluate the functional impact of several novel PCSK9 variants in a cohort of patients with FH by genetic cascade screening and in vitro functionality assays. Approach and Results: Patients with clinically diagnosed FH underwent genetic analysis of LDLR, and if negative, sequential testing of APOB and PCSK9. We analyzed cosegregation of hypercholesterolemia with novel PCSK9 variants. Gain of function status was determined by in silico analyses and validated by in vitro functionality assays. Among 1055 persons with clinical FH, we identified nonsynonymous PCSK9 variants in 27 (2.6%) patients and 7 of these carried one of the 4 previously reported gain of function variants. In the remaining 20 patients with FH, we identified 7 novel PCSK9 variants. The G516V variant (c.1547G>T) was found in 5 index patients and cascade screening identified 15 additional carriers. Low-density lipoprotein-cholesterol levels were higher in these 15 carriers compared with the 27 noncarriers (236±73 versus 124±35 mg/dL; P<0.001). In vitro studies demonstrated the pathogenicity of the G516V variant. CONCLUSIONS In our study, 1.14% of cases with clinical FH were clearly attributable to pathogenic variants in PCSK9. Pathogenicity is established beyond doubt for the G516V variant.
Collapse
Affiliation(s)
- Roeland Huijgen
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, The Netherlands (R.H., M.L.H., A.G.d.J., G.K.H.).,Spaarne Gasthuis, Haarlem, The Netherlands (R.H.)
| | - Dirk J Blom
- Division of Lipidology, Department of Medicine, Hatter Institute for Cardiovascular Research in Africa (D.J.B., Z.B., D.M.B., D.M.B., B.C.B., R.J.J., K.H.W.), University of Cape Town, South Africa
| | - Merel L Hartgers
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, The Netherlands (R.H., M.L.H., A.G.d.J., G.K.H.)
| | - Kévin Chemello
- Laboratoire Inserm UMR1188 DéTROI, Université de La Réunion, Sainte Clotilde, France (K.C., G.L.)
| | - Asier Benito-Vicente
- Biofisika Institute (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain (A.B.-V., K.B.U., C.M.)
| | - Kepa B Uribe
- Biofisika Institute (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain (A.B.-V., K.B.U., C.M.)
| | - Zorena Behardien
- Division of Lipidology, Department of Medicine, Hatter Institute for Cardiovascular Research in Africa (D.J.B., Z.B., D.M.B., D.M.B., B.C.B., R.J.J., K.H.W.), University of Cape Town, South Africa
| | - Dee M Blackhurst
- Division of Lipidology, Department of Medicine, Hatter Institute for Cardiovascular Research in Africa (D.J.B., Z.B., D.M.B., D.M.B., B.C.B., R.J.J., K.H.W.), University of Cape Town, South Africa
| | - Brigitte C Brice
- Division of Lipidology, Department of Medicine, Hatter Institute for Cardiovascular Research in Africa (D.J.B., Z.B., D.M.B., D.M.B., B.C.B., R.J.J., K.H.W.), University of Cape Town, South Africa
| | - Joep C Defesche
- Department of Clinical Genetics, Laboratory of Genome Diagnostics, Amsterdam University Medical Center, The Netherlands (J.C.D.)
| | - Annemiek G de Jong
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, The Netherlands (R.H., M.L.H., A.G.d.J., G.K.H.)
| | - Rosemary J Jooste
- Division of Lipidology, Department of Medicine, Hatter Institute for Cardiovascular Research in Africa (D.J.B., Z.B., D.M.B., D.M.B., B.C.B., R.J.J., K.H.W.), University of Cape Town, South Africa
| | - Gabriele A E Solomon
- Division of Chemical Pathology, Department of Pathology (G.A.E.S., A.D.M.), University of Cape Town, South Africa.,Division of Chemical Pathology, Department of Pathology, University of Cape Town, South Africa (G.A.E.S., A.D.M.)
| | - Karen H Wolmarans
- Division of Lipidology, Department of Medicine, Hatter Institute for Cardiovascular Research in Africa (D.J.B., Z.B., D.M.B., D.M.B., B.C.B., R.J.J., K.H.W.), University of Cape Town, South Africa
| | - G Kees Hovingh
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, The Netherlands (R.H., M.L.H., A.G.d.J., G.K.H.)
| | - Cesar Martin
- Biofisika Institute (UPV/EHU, CSIC) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain (A.B.-V., K.B.U., C.M.)
| | - Gilles Lambert
- Laboratoire Inserm UMR1188 DéTROI, Université de La Réunion, Sainte Clotilde, France (K.C., G.L.)
| | - A David Marais
- Division of Chemical Pathology, Department of Pathology (G.A.E.S., A.D.M.), University of Cape Town, South Africa.,Division of Chemical Pathology, Department of Pathology, University of Cape Town, South Africa (G.A.E.S., A.D.M.)
| |
Collapse
|
56
|
Matta A, Taraszkiewicz D, Bongard V, Ferrières J. Ineffective Subtilisin/Kexin Type 9 (PCSK9) Inhibitors Monotherapy in Dyslipidemia with Low-Density Lipoprotein Cholesterol (LDL-C) Receptor Abnormalities: A Report of 2 Cases. AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e923722. [PMID: 32929056 PMCID: PMC7520129 DOI: 10.12659/ajcr.923722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Case series Patients: Male, 44-year-old • Female, 71-year-old Final Diagnosis: Resistance to PCSK9I overcomed by adding statin Symptoms: Dyslipidemia Medication: — Clinical Procedure: — Specialty: Cardiology
Collapse
Affiliation(s)
- Anthony Matta
- Department of Preventive Cardiology, CHU-Rangueil Hospital, Toulouse, France.,Atherosclerosis Risk and Treatment Evaluation towards Risk Reduction Epidemiology (ARTERRE) Team, Epidemiology and Analyses in Public Health: Risks, Chronic Diseases and Disabilities (INSERM UMR1027), Toulouse, France.,Toulouse University School of Medicine, Paul Sabatier University, Toulouse, France.,Faculty of Medicine, Holy Spirit University of Kaslik, Kaslik, Lebanon
| | - Dorota Taraszkiewicz
- Department of Preventive Cardiology, CHU-Rangueil Hospital, Toulouse, France.,Atherosclerosis Risk and Treatment Evaluation towards Risk Reduction Epidemiology (ARTERRE) Team, Epidemiology and Analyses in Public Health: Risks, Chronic Diseases and Disabilities (INSERM UMR1027), Toulouse, France.,Toulouse University School of Medicine, Paul Sabatier University, Toulouse, France
| | - Vanina Bongard
- Department of Preventive Cardiology, CHU-Rangueil Hospital, Toulouse, France.,Atherosclerosis Risk and Treatment Evaluation towards Risk Reduction Epidemiology (ARTERRE) Team, Epidemiology and Analyses in Public Health: Risks, Chronic Diseases and Disabilities (INSERM UMR1027), Toulouse, France.,Toulouse University School of Medicine, Paul Sabatier University, Toulouse, France
| | - Jean Ferrières
- Department of Preventive Cardiology, CHU-Rangueil Hospital, Toulouse, France.,Atherosclerosis Risk and Treatment Evaluation towards Risk Reduction Epidemiology (ARTERRE) Team, Epidemiology and Analyses in Public Health: Risks, Chronic Diseases and Disabilities (INSERM UMR1027), Toulouse, France.,Toulouse University School of Medicine, Paul Sabatier University, Toulouse, France
| |
Collapse
|
57
|
Page MM, Ekinci EI, Burnett JR, Hooper AJ, Reid N, Bishop W, Florkowski CM, Scott R, O'Brien RC, Watts GF. Lipoprotein apheresis and PCSK9 inhibitors for severe familial hypercholesterolaemia: Experience from Australia and New Zealand. J Clin Apher 2020; 36:48-58. [PMID: 32911577 DOI: 10.1002/jca.21839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Severe familial hypercholesterolaemia (FH) causes premature disability and death due to atherosclerotic cardiovascular disease and is refractory to standard lipid-lowering therapies. Lipoprotein apheresis (LA) has long been a standard of care for patients with severe FH, but is invasive, expensive and time-consuming for patients and their caregivers. Newer drug therapies, including the proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, may reduce the need for LA. MATERIALS AND METHODS We audited the records of 16 patients (eight homozygous, eight heterozygous) treated with LA in Australia and New Zealand, 14 of whom subsequently commenced PCSK9 inhibitor therapy. LA was performed by cascade filtration in all centres. RESULTS LDL-cholesterol was acutely lowered by 69 ± 7% in patients with homozygous FH and by 72 ± 9% in those with heterozygous FH, representing time-averaged reductions of 36 ± 12% and 34 ± 5%, respectively. LA was well-tolerated, and patients reported comparable quality of life to population and disease-related norms. After commencement of PCSK9 inhibitors, four of seven patients with homozygous FH had meaningful biochemical responses, with a reduction in the frequency of LA permitted in one patient and complete cessation in another. Four of seven patients with heterozygous FH were able to be managed without LA after commencing PCSK9 inhibitors. CONCLUSION While PCSK9 inhibitors have reduced the need for LA, some patients with severe FH continue to require LA, and will require it for the foreseeable future. However, emerging therapies, including angiopoetin-like 3 inhibitors, may further reduce the need for LA.
Collapse
Affiliation(s)
- Michael M Page
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia.,Western Diagnostic Pathology, Myaree, Western Australia, Australia
| | - Elif I Ekinci
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia.,Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - John R Burnett
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia.,Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital and Fiona Stanley Hospital, Perth, Western Australia, Australia.,Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Amanda J Hooper
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia.,Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital and Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Nicola Reid
- Cardiovascular Prevention and Lipid Disorders Clinic, Christchurch Hospital, Christchurch, New Zealand
| | - Warrick Bishop
- Calvary Cardiac Centre, Calvary Hospital, Lenah Valley, Tasmania, Australia
| | - Chris M Florkowski
- Cardiovascular Prevention and Lipid Disorders Clinic, Christchurch Hospital, Christchurch, New Zealand.,Canterbury Health Laboratories, Christchurch, New Zealand
| | - Russell Scott
- Cardiovascular Prevention and Lipid Disorders Clinic, Christchurch Hospital, Christchurch, New Zealand
| | - Richard C O'Brien
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia.,Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia.,Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
58
|
Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJP, Rubba P, Ali S, Banerjee P, Chan KC, Gipe DA, Khilla N, Pordy R, Weinreich DM, Yancopoulos GD, Zhang Y, Gaudet D. Evinacumab for Homozygous Familial Hypercholesterolemia. N Engl J Med 2020; 383:711-720. [PMID: 32813947 DOI: 10.1056/nejmoa2004215] [Citation(s) in RCA: 476] [Impact Index Per Article: 95.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Homozygous familial hypercholesterolemia is characterized by premature cardiovascular disease caused by markedly elevated levels of low-density lipoprotein (LDL) cholesterol. This disorder is associated with genetic variants that result in virtually absent (null-null) or impaired (non-null) LDL-receptor activity. Loss-of-function variants in the gene encoding angiopoietin-like 3 (ANGPTL3) are associated with hypolipidemia and protection against atherosclerotic cardiovascular disease. Evinacumab, a monoclonal antibody against ANGPTL3, has shown potential benefit in patients with homozygous familial hypercholesterolemia. METHODS In this double-blind, placebo-controlled, phase 3 trial, we randomly assigned in a 2:1 ratio 65 patients with homozygous familial hypercholesterolemia who were receiving stable lipid-lowering therapy to receive an intravenous infusion of evinacumab (at a dose of 15 mg per kilogram of body weight) every 4 weeks or placebo. The primary outcome was the percent change from baseline in the LDL cholesterol level at week 24. RESULTS The mean baseline LDL cholesterol level in the two groups was 255.1 mg per deciliter, despite the receipt of maximum doses of background lipid-lowering therapy. At week 24, patients in the evinacumab group had a relative reduction from baseline in the LDL cholesterol level of 47.1%, as compared with an increase of 1.9% in the placebo group, for a between-group least-squares mean difference of -49.0 percentage points (95% confidence interval [CI], -65.0 to -33.1; P<0.001); the between-group least-squares mean absolute difference in the LDL cholesterol level was -132.1 mg per deciliter (95% CI, -175.3 to -88.9; P<0.001). The LDL cholesterol level was lower in the evinacumab group than in the placebo group in patients with null-null variants (-43.4% vs. +16.2%) and in those with non-null variants (-49.1% vs. -3.8%). Adverse events were similar in the two groups. CONCLUSIONS In patients with homozygous familial hypercholesterolemia receiving maximum doses of lipid-lowering therapy, the reduction from baseline in the LDL cholesterol level in the evinacumab group, as compared with the small increase in the placebo group, resulted in a between-group difference of 49.0 percentage points at 24 weeks. (Funded by Regeneron Pharmaceuticals; ELIPSE HoFH ClinicalTrials.gov number, NCT03399786.).
Collapse
Affiliation(s)
- Frederick J Raal
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Robert S Rosenson
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Laurens F Reeskamp
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - G Kees Hovingh
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - John J P Kastelein
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Paolo Rubba
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Shazia Ali
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Poulabi Banerjee
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Kuo-Chen Chan
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Daniel A Gipe
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Nagwa Khilla
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Robert Pordy
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - David M Weinreich
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - George D Yancopoulos
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Yi Zhang
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| | - Daniel Gaudet
- From the Faculty of Health Sciences, University of the Witwatersrand, Johannesburg (F.J.R.); the Cardiometabolics Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York (R.S.R.), and Regeneron Pharmaceuticals, Tarrytown (S.A., P.B, K.-C.C., D.A.G., N.K., R.P., D.M.W. G.D.Y., Y.Z.) - both in New York; the Department of Vascular Medicine, University of Amsterdam, Amsterdam (L.F.R., G.K.H., J.J.P.K.); the Department of Internal Medicine and Surgery, Federico II University, Naples, Italy (P.R.); and the Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (D.G.)
| |
Collapse
|
59
|
Efficacy and Safety of Alirocumab in Adults With Homozygous Familial Hypercholesterolemia. J Am Coll Cardiol 2020; 76:131-142. [DOI: 10.1016/j.jacc.2020.05.027] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022]
|
60
|
Agrawal H, Choy HHK, Liu J, Auyoung M, Albert MA. Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2020; 40:e185-e192. [PMID: 32579480 DOI: 10.1161/atvbaha.120.313608] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Harsh Agrawal
- From the Center for the Study of Adversity and Cardiovascular Disease (NURTURE Center), Division of Cardiology, Department of Medicine, University of California San Francisco (H.A., M.A.A.)
| | - Ho-Hin K Choy
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Jason Liu
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Matthew Auyoung
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Michelle A Albert
- From the Center for the Study of Adversity and Cardiovascular Disease (NURTURE Center), Division of Cardiology, Department of Medicine, University of California San Francisco (H.A., M.A.A.)
| |
Collapse
|
61
|
Abstract
PURPOSE OF REVIEW Homozygous familial hypercholesterolemia (HoFH) is a rare disorder associated with early atherosclerotic disease due to impairment of the LDL receptor (LDLR) pathway. Because of their molecular defect, current treatment options have limited success in bringing HoFH patient to LDL-C target and morbidity and mortality remain high. We review current and upcoming therapies directed at HoFH, including gene therapy. RECENT FINDINGS Recent real-world studies have confirmed the strength in lomitapide as a treatment adjunct to statins and other lipid-lowering therapies in HoFH patients. The approval of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor monoclonal antibodies has also been a welcome addition to the treatment armamentarium offering an additional average reduction in LDL-C levels of 24% when added to background lipid-lowering therapies in this population. Although achieving adequate LDL-C levels in this population is difficult, there are several therapies on the horizon that may help more patients reach goal. Evinacumab, a monoclonal antibody against ANGPTL3, has been shown to substantially reduce LDL-C of an average of 49%, independently of residual LDLR activity. RNA interference targeting PCSK9 and ANGPTL3 shows promise in clinical trials. Adeno-associated virus-mediated gene transfer and gene editing techniques are in early clinical and preclinical development. SUMMARY LDL-C lowering in HoFH patients remains very challenging. However, novel treatment options are emerging. Upcoming therapies directed at PCSK9 and ANPTL3 may offer additional LDL-C reduction, to help patients achieve adequate LDL-C levels. Gene therapy and gene editing techniques, if proven effective, may offer a unique opportunity to treat patients with a one-time treatment.
Collapse
Affiliation(s)
- Archna Bajaj
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
62
|
MicroRNA-148a regulates low-density lipoprotein metabolism by repressing the (pro)renin receptor. PLoS One 2020; 15:e0225356. [PMID: 32437440 PMCID: PMC7241754 DOI: 10.1371/journal.pone.0225356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 05/06/2020] [Indexed: 01/15/2023] Open
Abstract
High plasma LDL cholesterol (LDL-c) concentration is a major risk factor for atherosclerosis. Hepatic LDL receptor (LDLR) regulates LDL metabolism, and thereby plasma LDL-c concentration. Recently, we have identified the (pro)renin receptor [(P)RR] as a novel regulator of LDL metabolism, which regulates LDLR degradation and hence its protein abundance and activity. In silico analysis suggests that the (P)RR is a target of miR-148a. In this study we determined whether miR-148a could regulate LDL metabolism by regulating (P)RR expression in HepG2 and Huh7 cells. We found that miR-148a suppressed (P)RR expression by binding to the 3’-untranslated regions (3’-UTR) of the (P)RR mRNA. Mutating the binding sites for miR-148a in the 3’-UTR of (P)RR mRNA completely abolished the inhibitory effects of miR-148a on (P)RR expression. In line with our recent findings, reduced (P)RR expression resulted in decreased cellular LDL uptake, likely as a consequence of decreased LDLR protein abundance. Overexpressing the (P)RR prevented miR-148a-induced reduction in LDLR abundance and cellular LDL uptake. Our study supports a new concept that miR-148a is a regulator of (P)RR expression. By reducing (P)RR abundance, miR-148a decreases LDLR protein abundance and consequently cellular LDL uptake.
Collapse
|
63
|
Chemello K, Beeské S, Trang Tran TT, Blanchard V, Villard EF, Poirier B, Le Bail JC, Dargazanli G, Ho-Van-Guimbal S, Boulay D, Bergis O, Pruniaux MP, Croyal M, Janiak P, Guillot E, Lambert G. Lipoprotein(a) Cellular Uptake Ex Vivo and Hepatic Capture In Vivo Is Insensitive to PCSK9 Inhibition With Alirocumab. JACC Basic Transl Sci 2020; 5:549-557. [PMID: 32613143 PMCID: PMC7315184 DOI: 10.1016/j.jacbts.2020.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
Modulating LDL receptor expression genetically (in familial hypercholesterolemia) or pharmacologically (using statins or the PCSK9 inhibitor alirocumab) does not alter the cellular uptake of Lp(a) in primary human lymphocytes. Lp(a) hepatic capture is not modulated by PCSK9 inhibition with alirocumab in liver-humanized mice. LDLR does not appear to play a significant role in mediating Lp(a) plasma clearance in vivo.
Lipoprotein(a) (Lp[a]) is the most common genetically inherited risk factor for cardiovascular disease. Many aspects of Lp(a) metabolism remain unknown. We assessed the uptake of fluorescent Lp(a) in primary human lymphocytes as well as Lp(a) hepatic capture in a mouse model in which endogenous hepatocytes have been ablated and replaced with human ones. Modulation of LDLR expression with the PCSK9 inhibitor alirocumab did not alter the cellular or the hepatic uptake of Lp(a), demonstrating that the LDL receptor is not a major route for Lp(a) plasma clearance. These results have clinical implications because they underpin why statins are not efficient at reducing Lp(a).
Collapse
Key Words
- 3D, 3-dimensional
- AU, arbitrary unit
- BSA, bovine serum albumin
- ELISA, enzyme-linked immunosorbent assay
- FCR, fractional catabolic rate
- FRG, Fah(−/−)Rag2(−/−)Il2rg(−/−)
- HoFH, homozygous familial hypercholesterolemia
- LC-MS/MS, liquid chromatography tandem mass spectrometry
- LDL, low-density lipoprotein
- LDL-C, low-density lipoprotein cholesterol
- LDLR, low-density lipoprotein receptor
- Lp(a), lipoprotein(a)
- MFI, mean fluorescence intensity
- PBMC, peripheral blood mononuclear cell
- PBS, phosphate-buffered saline
- PCSK9, proprotein convertase subtilisin/kexin type 9
- apoB100, apolipoprotein B100
- bodipy, boron dipyrromethene
- lipoprotein(a)
- liver-humanized mice
- low-density lipoprotein receptor
- proprotein convertase subtilisin/kexin type 9
- rPCSK9, recombinant proprotein convertase subtilisin/kexin type 9
Collapse
Affiliation(s)
- Kévin Chemello
- Laboratoire Inserm UMR 1188 DéTROI, Université de La Réunion, Sainte Clotilde, France
| | | | | | - Valentin Blanchard
- Laboratoire Inserm UMR 1188 DéTROI, Université de La Réunion, Sainte Clotilde, France
| | | | | | | | | | | | | | | | | | - Mikaël Croyal
- Université de Nantes, CRNH Ouest, Inra UMR 1280 PhAN, Nantes, France
| | | | | | - Gilles Lambert
- Laboratoire Inserm UMR 1188 DéTROI, Université de La Réunion, Sainte Clotilde, France
| |
Collapse
|
64
|
A small molecule inhibitor of PCSK9 that antagonizes LDL receptor binding via interaction with a cryptic PCSK9 binding groove. Bioorg Med Chem 2020; 28:115344. [DOI: 10.1016/j.bmc.2020.115344] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
|
65
|
Familial hypercholesterolaemia: evolving knowledge for designing adaptive models of care. Nat Rev Cardiol 2020; 17:360-377. [DOI: 10.1038/s41569-019-0325-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2019] [Indexed: 01/05/2023]
|
66
|
Ramin-Mangata S, Wargny M, Pichelin M, Le May C, Thédrez A, Blanchard V, Nativel B, Santos RD, Benseñor IM, Lotufo PA, Lambert G, Cariou B. Circulating PCSK9 levels are not associated with the conversion to type 2 diabetes. Atherosclerosis 2020; 293:49-56. [DOI: 10.1016/j.atherosclerosis.2019.11.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/07/2019] [Accepted: 11/27/2019] [Indexed: 01/09/2023]
|
67
|
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020; 41:111-188. [PMID: 31504418 DOI: 10.1093/eurheartj/ehz455] [Citation(s) in RCA: 5188] [Impact Index Per Article: 1037.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
68
|
Abstract
Loss-of-function variants in PCSK9 (proprotein convertase subtilisin-kexin type 9) are associated with lower lifetime risk of atherosclerotic cardiovascular disease) events. Confirmation of these genetic observations in large, prospective clinical trials in participants with atherosclerotic cardiovascular disease has provided guidance on risk stratification and enhanced our knowledge on hitherto unresolved and contentious issues concerning the efficacy and safety of markedly lowering LDL-C (low-density lipoprotein cholesterol). PCSK9 has a broad repertoire of molecular effects. Furthermore, clinical trials with PCSK9 inhibitors demonstrate that reductions in atherosclerotic cardiovascular disease events are more effective in patients with recent myocardial infarction, multiple myocardial infarctions, multivessel coronary artery disease, and lower extremity arterial disease. The potent LDL-C lowering efficacy of PCSK9 inhibitors provides the opportunity for more aggressive LDL-lowering strategies in high-risk patients with atherosclerotic cardiovascular disease and supports the notion that there is no lower limit for LDL-C. Aggressive LDL-C lowering with fully human PCSK9 monoclonal antibodies has been associated by a safety profile superior to that of other classes of LDL-lowering agents. These clinical trials provide evidence that LDL lowering with PCSK9 inhibitors is an effective therapy for lowering cardiovascular events in high-risk patients with LDL-C levels ≥70 mg/dL on maximally tolerated oral therapies, including statins and ezetimibe.
Collapse
Affiliation(s)
- Robert S Rosenson
- From the Zena and Michael A. Wiener Cardiovascular Institute and Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, NY (R.S.R.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine, Western University, London, Ontario, Canada
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (W.K.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (W.K.).,Institute of Epidemiology and Biostatistics, University of Ulm, Germany (W.K.)
| |
Collapse
|
69
|
Raal FJ, Hovingh GK, Catapano AL. Familial hypercholesterolemia treatments: Guidelines and new therapies. Atherosclerosis 2019; 277:483-492. [PMID: 30270089 DOI: 10.1016/j.atherosclerosis.2018.06.859] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/28/2018] [Accepted: 06/14/2018] [Indexed: 12/16/2022]
Abstract
Familial hypercholesterolemia (FH) is a genetic disorder resulting from mutations in genes encoding proteins involved in the metabolism of low density lipoproteins (LDL) and characterized by premature cardiovascular disease due to the exposure to high levels of LDL-cholesterol (LDL-C) from birth. Thus, the early identification of FH subjects, followed by appropriate treatment is essential to prevent or at least delay the onset of cardiovascular events. However, FH is largely underdiagnosed; in addition, FH patients are frequently not adequately treated, despite the availability of several pharmacological therapies to significantly reduce LDL-C levels. Current guidelines recommend LDL-C targets for FH (either heterozygotes [HeFH] or homozygotes [HoFH]) <100 mg/dL (<2.6 mmol/L) for adults or <70 mg/dL (<1.8 mmol/L) for adults with CHD or diabetes, and <135 mg/dL (<3.5 mmol/L) for children. With the pharmacological options now available, which include statins as a first approach, ezetimibe, and the recently approved monoclonal antibodies targeting PCSK9, the guideline recommended LDL-C target levels can be achieved in the majority of heterozygous FH subjects, while for the most severe forms of homozygous FH, the addition of therapies such as lomitapide either with or without apheresis may be required.
Collapse
Affiliation(s)
- Frederick J Raal
- Carbohydrate & Lipid Metabolism Research Unit, Division of Endocrinology & Metabolism, Department of Medicine, Faculty of Health Sciences, Johannesburg Hospital, University of the Witwatersrand, Parktown, Johannesburg, South Africa
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy; IRCCS Multimedica, Milan, Italy.
| |
Collapse
|
70
|
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, Chapman MJ, De Backer GG, Delgado V, Ference BA, Graham IM, Halliday A, Landmesser U, Mihaylova B, Pedersen TR, Riccardi G, Richter DJ, Sabatine MS, Taskinen MR, Tokgozoglu L, Wiklund O, Windecker S, Aboyans V, Baigent C, Collet JP, Dean V, Delgado V, Fitzsimons D, Gale CP, Grobbee D, Halvorsen S, Hindricks G, Iung B, Jüni P, Katus HA, Landmesser U, Leclercq C, Lettino M, Lewis BS, Merkely B, Mueller C, Petersen S, Petronio AS, Richter DJ, Roffi M, Shlyakhto E, Simpson IA, Sousa-Uva M, Touyz RM, Nibouche D, Zelveian PH, Siostrzonek P, Najafov R, van de Borne P, Pojskic B, Postadzhiyan A, Kypris L, Špinar J, Larsen ML, Eldin HS, Viigimaa M, Strandberg TE, Ferrières J, Agladze R, Laufs U, Rallidis L, Bajnok L, Gudjónsson T, Maher V, Henkin Y, Gulizia MM, Mussagaliyeva A, Bajraktari G, Kerimkulova A, Latkovskis G, Hamoui O, Slapikas R, Visser L, Dingli P, Ivanov V, Boskovic A, Nazzi M, Visseren F, Mitevska I, Retterstøl K, Jankowski P, Fontes-Carvalho R, Gaita D, Ezhov M, Foscoli M, Giga V, Pella D, Fras Z, Perez de Isla L, Hagström E, Lehmann R, Abid L, Ozdogan O, Mitchenko O, Patel RS. 2019 ESC/EAS guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Atherosclerosis 2019; 290:140-205. [PMID: 31591002 DOI: 10.1016/j.atherosclerosis.2019.08.014] [Citation(s) in RCA: 657] [Impact Index Per Article: 109.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
71
|
Banerjee P, Chan KC, Tarabocchia M, Benito-Vicente A, Alves AC, Uribe KB, Bourbon M, Skiba PJ, Pordy R, Gipe DA, Gaudet D, Martin C. Functional Analysis of LDLR (Low-Density Lipoprotein Receptor) Variants in Patient Lymphocytes to Assess the Effect of Evinacumab in Homozygous Familial Hypercholesterolemia Patients With a Spectrum of LDLR Activity. Arterioscler Thromb Vasc Biol 2019; 39:2248-2260. [PMID: 31578082 PMCID: PMC6818983 DOI: 10.1161/atvbaha.119.313051] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Supplemental Digital Content is available in the text. Homozygous familial hypercholesterolemia is a rare disease usually caused by LDLR (low-density lipoprotein receptor) mutations. Homozygous familial hypercholesterolemia is characterized by markedly elevated LDL-C (low-density lipoprotein cholesterol) levels and an extremely high risk of premature atherosclerotic cardiovascular disease. A phase 2, proof-of-concept study (NCT02265952) demonstrated that evinacumab, a fully human monoclonal antibody to ANGPTL3 (angiopoietin-like 3 protein), reduced LDL-C levels in 9 patients with genotypically confirmed homozygous familial hypercholesterolemia and was well tolerated. The aim of this study was to analyze the effects of evinacumab on LDLR activity in lymphocytes purified from patients in the proof-of-concept study.
Collapse
Affiliation(s)
- Poulabi Banerjee
- From Regeneron Pharmaceuticals, Inc, Tarrytown, NY (P.B., K-C.C., M.T., P.J.S., R.P., D.A.G.)
| | - Kuo-Chen Chan
- From Regeneron Pharmaceuticals, Inc, Tarrytown, NY (P.B., K-C.C., M.T., P.J.S., R.P., D.A.G.)
| | - Michel Tarabocchia
- From Regeneron Pharmaceuticals, Inc, Tarrytown, NY (P.B., K-C.C., M.T., P.J.S., R.P., D.A.G.)
| | - Asier Benito-Vicente
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, UPV/EHU, Spain (A.B-V., K.B.U, C.M.)
| | - Ana C Alves
- Unidade de I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal (A.C.A., M.B.)
| | - Kepa B Uribe
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, UPV/EHU, Spain (A.B-V., K.B.U, C.M.)
| | - Mafalda Bourbon
- Unidade de I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal (A.C.A., M.B.)
| | - Paul J Skiba
- From Regeneron Pharmaceuticals, Inc, Tarrytown, NY (P.B., K-C.C., M.T., P.J.S., R.P., D.A.G.)
| | - Robert Pordy
- From Regeneron Pharmaceuticals, Inc, Tarrytown, NY (P.B., K-C.C., M.T., P.J.S., R.P., D.A.G.)
| | - Daniel A Gipe
- From Regeneron Pharmaceuticals, Inc, Tarrytown, NY (P.B., K-C.C., M.T., P.J.S., R.P., D.A.G.)
| | - Daniel Gaudet
- Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada (D.G.)
| | - Cesar Martin
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, UPV/EHU, Spain (A.B-V., K.B.U, C.M.)
| |
Collapse
|
72
|
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]
|
73
|
Chemello K, Martín C, Lambert G. PCSK9 inhibition for autosomal recessive hypercholesterolemia. Atherosclerosis 2019; 284:209-211. [DOI: 10.1016/j.atherosclerosis.2019.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/12/2019] [Indexed: 10/27/2022]
|
74
|
Rodríguez-Jiménez C, Gómez-Coronado D, Frías Vargas M, Cerrato F, Lahoz C, Saban-Ruiz J, González-Nieto D, Lasunción MA, Mostaza JM, Rodríguez-Nóvoa S. A new variant (c.1A>G) in LDLRAP1 causing autosomal recessive hypercholesterolemia: Characterization of the defect and response to PCSK9 inhibition. Atherosclerosis 2019; 284:223-229. [PMID: 30777337 DOI: 10.1016/j.atherosclerosis.2019.01.010] [Citation(s) in RCA: 6] [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: 09/18/2018] [Revised: 12/12/2018] [Accepted: 01/10/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND AND AIMS Autosomal recessive hypercholesterolemia (ARH) is a rare disorder caused by mutations in LDLRAP1, which impairs internalization of hepatic LDL receptor (LDLR). ARH patients respond relatively well to statins or the combination of statins and Ezetimibe, but scarce and variable data on treatment with PCSK9 inhibitors is available. We aimed to identify and characterize the defect in a hypercholesterolemic patient with premature cardiovascular disease and determine the response to lipid-lowering treatment. METHODS AND RESULTS Gene sequencing revealed a homozygous c.1A > G:p.? variant in LDLRAP1. Primary lymphocytes were isolated from the ARH patient, one control and two LDLR-defective subjects, one LDLR:p.(Cys352Ser) heterozygote and one LDLR:p.(Asn825Lys) homozygote. The patient had undetectable full-length ARH protein by Western blotting, but expressed a lower-than-normal molecular weight peptide. LDLR activity was measured by flow cytometry, which showed that LDL binding and uptake were reduced in lymphocytes from the ARH patient as compared to control lymphocytes, but were slightly higher than in those from the LDLR:p.(Cys352Ser) heterozygote. Despite the analogous internalization defect predicted in ARH and homozygous LDLR:p.(Asn825Lys) lymphocytes, LDL uptake was higher in the former than in the latter. LDL-cholesterol levels were markedly reduced by the successive therapy with Atorvastatin and Atorvastatin plus Ezetimibe, and the addition of Evolocumab biweekly decreased LDL-cholesterol by a further 39%. CONCLUSIONS The LDLRAP1:c.1A > G variant is associated with the appearance of an N-terminal truncated ARH protein and to reduced, although still significant, LDLR activity in lymphocytes. Residual LDLR activity may be relevant for the substantial response of the patient to Evolocumab.
Collapse
Affiliation(s)
- Carmen Rodríguez-Jiménez
- Department of Genetics of Metabolic Diseases, Institute of Medical & Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Diego Gómez-Coronado
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain
| | | | - Francisca Cerrato
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Carlos Lahoz
- Department of Internal Medicine, Hospital Carlos III-La Paz, Madrid, Spain
| | - Jose Saban-Ruiz
- Endothelium and Cardiometabolic Medicine Unit, Department of Internal Medicine, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Photonics Technology and Bioengineering Department, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, and CIBERBBN, Spain
| | - Miguel A Lasunción
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain
| | - José M Mostaza
- Department of Internal Medicine, Hospital Carlos III-La Paz, Madrid, Spain
| | - Sonia Rodríguez-Nóvoa
- Department of Genetics of Metabolic Diseases, Institute of Medical & Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Madrid, Spain.
| |
Collapse
|
75
|
Khantalin I, Blanchard V, Viallet N, Lambert G. Recurrent coronary syndromes in a patient with isolated very-high lipoprotein (a) and the prothrombin genetic variant rs1799963 (G20210A): a case report. Eur Heart J Case Rep 2019; 3:ytz019. [PMID: 31020261 PMCID: PMC6439380 DOI: 10.1093/ehjcr/ytz019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/07/2019] [Indexed: 12/24/2022]
Abstract
Background Elevated lipoprotein (a) [Lp(a)] is an under-diagnosed genetically inherited risk factor for coronary heart disease (CHD) and calcific aortic valve stenosis. Premature myocardial infarction (MI) could stem from the association between elevated Lp(a) and other non-traditional cardiovascular risk factors. Case summary Here, we report a male patient with extremely high Lp(a) plasma levels [610 nmol/L (244 mg/dL); normal <75 nmol/L (<30 mg/dL)] associated with the prothrombin genetic variant rs1799963 (G20210A) and no other CHD risk factor. At the age of 32, he suffered recurrent episodes of MI treated by coronary angioplasty and drug eluting stents. The patient who was initially prescribed antiplatelet therapy, beta-blockers, and statins, has subsequently been treated by lipoprotein apheresis every fortnight for 43 months. He has never experienced any recurrent episode of angina or chest pain since. Discussion The rare association between extremely elevated circulating Lp(a) levels and prothrombotic genetic variants of coagulation factors appears to be a deadly combination that can only be adequately treated by antiplatelet therapy and lipoprotein apheresis.
Collapse
Affiliation(s)
- Ilya Khantalin
- Université de La Réunion, Inserm, UMR 1188 DéTROI, Plateforme CYROI, 2 Rue Maxime Rivière, Sainte-Clotilde, France.,CHU de La Réunion, Service de Chirurgie Vasculaire, Allée des Topazes, Saint-Denis, France
| | - Valentin Blanchard
- Université de La Réunion, Inserm, UMR 1188 DéTROI, Plateforme CYROI, 2 Rue Maxime Rivière, Sainte-Clotilde, France
| | - Nicolas Viallet
- CHU de La Réunion, Service de Néphrologie, Allée des Topazes, Saint-Denis, France
| | - Gilles Lambert
- Université de La Réunion, Inserm, UMR 1188 DéTROI, Plateforme CYROI, 2 Rue Maxime Rivière, Sainte-Clotilde, France
| |
Collapse
|
76
|
PCSK9 inhibition 2018: riding a new wave of coronary prevention. Clin Sci (Lond) 2019; 133:205-224. [DOI: 10.1042/cs20171300] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023]
Abstract
AbstractProprotein convertase subtilisin/kexin type 9 (PCSK9) is a hepatic enzyme that regulates the low-density lipoprotein cholesterol (LDL-c) receptor and thus circulating LDL-c levels. With overwhelming evidence now supporting the reduction in LDL-c to lower the risk of cardiovascular disease, PCSK9 inhibitors represent an important therapeutic target, particularly in high-risk populations. Here, we summarise and update the science of PCSK9, including its discovery and the development of various inhibitors, including the now approved monoclonal antibodies. In addition, we summarise the clinical applications of PCSK9 inhibitors in a range of patient populations, as well as the major randomised controlled trials investigating their use in coronary prevention.
Collapse
|
77
|
Santos RD. Expression of LDLRs (Low-Density Lipoprotein Receptors), Dyslipidemia Severity, and Response to PCSK9 (Proprotein Convertase Subtilisin Kexin Type 9) Inhibition in Homozygous Familial Hypercholesterolemia: Connecting the Dots. Arterioscler Thromb Vasc Biol 2018; 38:481-483. [PMID: 29467219 DOI: 10.1161/atvbaha.117.310675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Raul D Santos
- From the Lipid Clinic Heart Institute (InCor), University of Sao Paulo Medical School Hospital, Brazil; and Preventive Medicine Centre and Cardiology Program, Hospital Israelita Albert Einstein, Sao Paulo, Brazil.
| |
Collapse
|
78
|
Abstract
PURPOSE OF REVIEW Our primary objective is to review the most recent findings on the biology of PCSK9 and on two key aspects of PCSK9 inhibition beyond LDL control of great clinical relevance: the regulation of lipoprotein (a) circulating levels by PCSK9 inhibitors and the putative diabetogenic effects of these novel therapies. RECENT FINDINGS The reality of two distinct extracellular and intracellular pathways by which PCSK9 decreases the abundance of the LDLR at the surface of many cell types, most importantly hepatocytes, has recently been established. In contrast, the exact mechanisms by which PCSK9 inhibitors lower the circulating levels of lipoprotein (a) remain a point of major dispute. Despite strong indications from genetic studies that PCSK9 inhibition should increase diabetes risk, no such effect has been observed in clinical trials, and in-vitro and in-vivo studies do not clarify this issue. SUMMARY The trafficking pathways by which PCSK9 enhance LDLR degradation via the endolysosomal extracellular route or via the Golgi-lysosomal intracellular route remain to be fully elucidated. The mechanisms by which PCSK9 inhibitors reduce lipoprotein (a) also merit additional research efforts. The role of PCSK9 on glucose metabolism should likewise be studied in depth.
Collapse
|
79
|
Abstract
Clinical trials have unequivocally shown that inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) efficaciously and safely prevents cardiovascular events by lowering levels of LDL cholesterol. PCSK9 in the circulation is derived mainly from the liver, but the protein is also expressed in the pancreas, the kidney, the intestine and the central nervous system. Although PCSK9 modulates cholesterol metabolism by regulating LDL receptor expression in the liver, in vitro and in vivo studies have suggested that PCSK9 is involved in various other physiological processes. Although therapeutic PCSK9 inhibition could theoretically have undesired effects by interfering with these non-cholesterol-related processes, studies of individuals with genetically determined reduced PCSK9 function and clinical trials of PCSK9 inhibitors have not revealed clinically meaningful adverse consequences of almost completely eradicating PCSK9 from the circulation. The clinical implications of PCSK9 functions beyond lipid metabolism in terms of wanted or unwanted effects of therapeutic PCSK9 inhibition therefore appear to be limited. The objective of this Review is to describe the physiological role of PCSK9 beyond the LDL receptor to provide a rational basis for monitoring the effects of PCSK9 inhibition as these drugs gain traction in the clinic.
Collapse
Affiliation(s)
| | - Gilles Lambert
- Inserm UMR 1188 DéTROI, Université de La Réunion, Saint-Denis de La Réunion, France
| | - Bertrand Cariou
- L'institut du thorax, INSERM, CNRS, Université de Nantes, CHU Nantes, Nantes, France
| | - G Kees Hovingh
- Department of Vascular Medicine, Academisch Medisch Centrum, Amsterdam, Netherlands.
| |
Collapse
|
80
|
Nohara A, Otsubo Y, Yanagi K, Yoshida M, Ikewaki K, Harada-Shiba M, Jurecka A. Safety and Efficacy of Lomitapide in Japanese Patients with Homozygous Familial Hypercholesterolemia (HoFH): Results from the AEGR-733-301 Long-Term Extension Study. J Atheroscler Thromb 2018; 26:368-377. [PMID: 30259883 PMCID: PMC6456458 DOI: 10.5551/jat.45708] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Lomitapide is an approved lipid-lowering agent indicated as adjunct to low-fat diet and standard lipid-lowering therapies (LLTs) including lipoprotein apheresis for the treatment of homozygous familial hypercholesterolemia (HoFH). Clinical data from Phase 3 studies have demonstrated the prolonged lipid-lowering capacity of lomitapide in patients with HoFH. We assessed the long-term lipid-lowering capacity of daily oral lomitapide in a cohort of Japanese patients with HoFH enrolled in a Phase 3 extension study. Methods: Five of 8 Japanese HoFH patients completing a 56-week Phase 3 dose-escalation and safety study of lomitapide continued their maximum tolerated dose (MTD) until study drug was approved or commercially available or until treatment was discontinued. Lipid parameters were measured at Day 1 and at 12-week intervals through study end. Safety and tolerability were assessed. Results: Daily lomitapide treatment with permitted LLTs maintained approximately 50% mean reductions in plasma low-density lipoprotein cholesterol (LDL-C) levels from baseline for > 60 weeks. Reductions in LDL-C levels varied across patients and were not associated with the HoFH genotype. Four patients achieved > 25% reductions and 1 patient achieved > 50% reduction in LDL-C; 2 patients achieved reduction in LDL-C to < 100 mg/dL. Lomitapide significantly reduced total cholesterol (−26.5%), triglycerides (−54.8%), and non-highdensity lipoprotein cholesterol (non-HDL-C) (−37.4%). All 5 patients continued their individual MTD of lomitapide throughout the extension study with acceptable safety and tolerability, and no deaths were reported. Conclusions: Results from this extension study support the long-term safety and efficacy of lomitapide in significantly reducing plasma levels of atherosclerotic lipids in patients with HoFH.
Collapse
Affiliation(s)
- Atsushi Nohara
- Kanazawa University Hospital, Kanazawa University Health Service Center
| | | | | | | | | | | | | |
Collapse
|
81
|
|
82
|
Rosenson RS, Hegele RA, Fazio S, Cannon CP. The Evolving Future of PCSK9 Inhibitors. J Am Coll Cardiol 2018; 72:314-329. [DOI: 10.1016/j.jacc.2018.04.054] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 01/09/2023]
|
83
|
PCSK9 monoclonal antibody on a knife-edge: An article of faith in FH? J Clin Lipidol 2018; 12:844-848. [PMID: 29945779 DOI: 10.1016/j.jacl.2018.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 11/23/2022]
|
84
|
Packard CJ. Determinants of Achieved LDL Cholesterol and "Non-HDL" Cholesterol in the Management of Dyslipidemias. Curr Cardiol Rep 2018; 20:60. [PMID: 29904807 DOI: 10.1007/s11886-018-1003-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The advent of combination therapy to provide LDL lowering beyond that achieved with statins necessitates the development of greater understanding of how drugs work together, what changes occur in key lipoprotein fractions, and what residual risk remains. RECENT FINDINGS Clinical trials of agents that, when added to statins, generate profound LDL lowering have been successful in reducing further the risk of cardiovascular disease. LDL cholesterol can be now decreased to unprecedented levels, so the focus of attention then shifts to other apolipoprotein B-containing, atherogenic lipoprotein classes such as lipoprotein(a) and remnants of the metabolism of triglyceride-rich particles. "Non-HDL cholesterol" is used increasingly (especially if measured in the non-fasting state) as a more comprehensive index of risk. Metabolic studies reveal how current drugs act in combination to achieve profound lipid lowering. However, care is needed in interpreting achieved LDLc and non-HDLc levels in the emerging treatment paradigm.
Collapse
Affiliation(s)
- Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Ground Floor, Room G31, McGregor Building, University Avenue, Glasgow, G12 8QQ, UK.
| |
Collapse
|