301
|
Mauri M, Calmarza P, Ibarretxe D. Dyslipemias and pregnancy, an update. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2020; 33:41-52. [PMID: 33309071 DOI: 10.1016/j.arteri.2020.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
During pregnancy there is a physiological increase in total cholesterol (TC) and triglycerides (TG) plasma concentrations, due to increased insulin resistance, oestrogens, progesterone, and placental lactogen, although their reference values are not exactly known, TG levels can increase up to 300mg/dL, and TC can go as high as 350mg/dL. When the cholesterol concentration exceeds the 95th percentile (familial hypercholesterolaemia (FH) and transient maternal hypercholesterolaemia), there is a predisposition to oxidative stress in foetal vessels, exposing the newborn to a greater fatty streaks formation and a higher risk of atherosclerosis. However, the current treatment of pregnant women with hyperlipidaemia consists of a diet and suspension of lipid-lowering drugs. The most prevalent maternal hypertriglyceridaemia (HTG) is due to secondary causes, like diabetes, obesity, drugs, etc. The case of severe HTG due to genetic causes is less prevalent, and can be a higher risk of maternal-foetal complications, such as, acute pancreatitis (AP), pre-eclampsia, preterm labour, and gestational diabetes. Severe HTG-AP is a rare but potentially lethal pregnancy complication, for the mother and the foetus, usually occurs during the third trimester or in the immediate postpartum period, and there are no specific protocols for its diagnosis and treatment. In conclusion, it is crucial that dyslipidaemia during pregnancy must be carefully evaluated, not just because of the acute complications, but also because of the future cardiovascular morbidity and mortality of the newborn child. That is why the establishment of consensus protocols or guidelines is essential for its management.
Collapse
Affiliation(s)
- Marta Mauri
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital de Terrassa, Consorci Sanitari de Terrassa, Terrassa, Barcelona, España
| | - Pilar Calmarza
- Servicio de Bioquímica Clínica, Hospital Universitario Miguel Servet, Universidad de Zaragoza, Zaragoza, España.
| | - Daiana Ibarretxe
- Unidad de Medicina Vascular y Metabolismo (UVASMET), Hospital Universitario de Reus, Universidad Rovira y Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| |
Collapse
|
302
|
Watts GF, Sullivan DR, Hare DL, Kostner KM, Horton AE, Bell DA, Brett T, Trent RJ, Poplawski NK, Martin AC, Srinivasan S, Justo RN, Chow CK, Pang J. Integrated Guidance for Enhancing the Care of Familial Hypercholesterolaemia in Australia. Heart Lung Circ 2020; 30:324-349. [PMID: 33309206 DOI: 10.1016/j.hlc.2020.09.943] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022]
Abstract
Familial hypercholesterolaemia (FH) is a dominant and highly penetrant monogenic disorder present from birth that markedly elevates plasma low-density lipoprotein (LDL)-cholesterol concentration and, if untreated, leads to premature atherosclerosis and coronary artery disease (CAD). There are approximately 100,000 people with FH in Australia. However, an overwhelming majority of those affected remain undetected and inadequately treated, consistent with FH being a leading challenge for public health genomics. To further address the unmet need, we provide an updated guidance, presented as a series of systematically collated recommendations, on the care of patients and families with FH. These recommendations have been informed by an exponential growth in published works and new evidence over the last 5 years and are compatible with a contemporary global call to action on FH. Recommendations are given on the detection, diagnosis, assessment and management of FH in adults and children. Recommendations are also made on genetic testing and risk notification of biological relatives who should undergo cascade testing for FH. Guidance on management is based on the concepts of risk re-stratification, adherence to heart healthy lifestyles, treatment of non-cholesterol risk factors, and safe and appropriate use of LDL-cholesterol lowering therapies, including statins, ezetimibe, proprotein convertase subtilisin/kexin type 9 inhibitors and lipoprotein apheresis. Broad recommendations are also provided for the organisation and development of health care services. Recommendations on best practice need to be underpinned by good clinical judgment and shared decision making with patients and families. Models of care for FH need to be adapted to local and regional health care needs and available resources. A comprehensive and realistic implementation strategy, informed by further research, including assessments of cost-benefit, will be required to ensure that this new guidance benefits all Australian families with or at risk of FH.
Collapse
Affiliation(s)
- Gerald F Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia; Lipid Disorders Clinic, Cardiometabolic Service, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia.
| | - David R Sullivan
- Department of Chemical Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - David L Hare
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Vic, Australia; Department of Cardiology, Austin Health, Melbourne, Vic, Australia
| | - Karam M Kostner
- Department of Cardiology, Mater Hospital, University of Queensland, Brisbane, Qld, Australia
| | - Ari E Horton
- Monash Heart and Monash Children's Hospital, Monash Health, Melbourne, Vic, Australia; Monash Cardiovascular Research Centre, Melbourne, Vic, Australia; Department of Paediatrics, Monash University, Melbourne, Vic, Australia
| | - Damon A Bell
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia; Lipid Disorders Clinic, Cardiometabolic Service, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia; Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital and Fiona Stanley Hospital Network, Perth, WA, Australia; Department of Clinical Biochemistry, Clinipath Pathology, Perth, WA, Australia; Sonic Genetics, Sonic Pathology, Sydney, NSW, Australia
| | - Tom Brett
- General Practice and Primary Health Care Research, School of Medicine, University of Notre Dame Australia, Fremantle, WA, Australia
| | - Ronald J Trent
- Department of Medical Genomics, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Central Clinical School, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Nicola K Poplawski
- Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Andrew C Martin
- Department General Paediatrics, Perth Children's Hospital, Perth, WA, Australia; Division of Paediatrics, Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia
| | - Shubha Srinivasan
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Robert N Justo
- Department of Paediatric Cardiology, Queensland Children's Hospital, Brisbane, Qld, Australia; School of Medicine, University of Queensland, Brisbane, Qld, Australia
| | - Clara K Chow
- Westmead Applied Research Centre, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; George Institute for Global Health, Sydney, NSW, Australia
| | - Jing Pang
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia
| | | |
Collapse
|
303
|
McCormick D, Bhatt DL, Bays HE, Taub PR, Caldwell KA, Guerin CK, Steinhoff J, Ahmad Z, Singh R, Moreo K, Carter J, Heggen CL, Sapir T. A regional analysis of payer and provider views on cholesterol management: PCSK9 inhibitors as an illustrative alignment model. J Manag Care Spec Pharm 2020; 26:1517-1528. [PMID: 33251993 PMCID: PMC10391214 DOI: 10.18553/jmcp.2020.26.12.1517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND: Multiple barriers exist for appropriate use of the proprotein convertase subtilisin/kexin type 9 enzyme inhibitors (PCSK9i) in patients with atherosclerotic cardiovascular disease (ASCVD) or familial hypercholesterolemia (FH) with inadequately controlled hypercholesterolemia despite standard therapies. Among these barriers, high payer rejection rates and inadequate prior authorization (PA) documentation by providers hinder optimal use of PCSK9i. OBJECTIVES: To (a) identify and discuss provider and payer discordances on barriers to authorization and use of PCSK9i based on clinical and real-world evidence and (b) align understanding and application of clinical, cost, safety, and efficacy data of PCSK9i. METHODS: Local groups of 3 payers and 3 providers met in 6 separate locations across the United States through a collaborative project of AMCP and PRIME Education. Responses to selected pre- and postmeeting survey questions measured changes in attitudes and beliefs regarding treatment barriers, lipid thresholds for considering PCSK9i therapy, and tactics for improving PA processes. Statistical analysis of inter- and intragroup changes in attitudes were performed by Cox proportional hazards test and Fisher's exact test for < 5 variables. RESULTS: The majority of providers and payers (67%-78%) agreed that high patient copayments and inadequate PA documentation were significant barriers to PCSK9i usage. However, payers and providers differed on beliefs that current evidence does not support PCSK9i cost-effectiveness (6% providers, 56% payers; P = 0.003) and that PA presents excessive administrative burden (72% providers, 44% payers; P = 0.09) Average increases pre- to postmeeting were noted in provider beliefs that properly documented PA forms expedite access to PCSK9i (22%-50% increase) and current authorization criteria accurately distinguish patients who benefit most from PCSK9i (6%-22%). Payers decreased in their belief that current authorization criteria accurately distinguish benefiting patients (72%-50%). Providers and payers increased in their belief that PCSK9i are cost-effective (44%-61% and 28%-50%, respectively) and were more willing to consider PCSK9i at the low-density lipoprotein cholesterol threshold of > 70 mg/dL for patients with ASCVD (78%-83% and 44%-67%, respectively) or FH (22%-39% and 22%-33%). Payers were more agreeable to less stringent PA requirements for patients with FH (33%-72%, P = 0.019) and need for standardized PA requirements (50%-83%, P = 0.034); these considerations remained high (89%) among providers after the meeting. Most participants supported educational programs for patient treatment adherence (83%) and physician/staff PA processes (83%-94%). CONCLUSIONS: Provider and payer representatives in 6 distinct geographic locations provided recommendations to improve quality of care in patients eligible for PCSK9i. Participants also provided tactical recommendations for streamlining PA documentation processes and improving awareness of PCSK9i cost-effectiveness and clinical efficacy. The majority of participants supported development of universal, standardized patient eligibility criteria and PA forms. DISCLOSURES: The study reported in this article was part of a continuing education program funded by an independent educational grant awarded by Sanofi US and Regeneron Pharmaceuticals to PRIME Education. The grantor had no role in the study design, execution, analysis, or reporting. AMCP received grant funding from PRIME to assist in the study, as well as in writing the manuscript. McCormick, Bhatt, Bays, Taub, Caldwell, Guerin, Steinhoff, and Ahmad received an honorarium from PRIME for serving as faculty for the continuing education program. McCormick, Bhatt, Bays, Taub, Caldwell, Guerin, Steinhoff, and Ahmad were involved as participants in the study. Bhatt discloses the following relationships: Advisory board: Cardax, CellProthera, Cereno Scientific, Elsevier Practice Update Cardiology, Level Ex, Medscape Cardiology, PhaseBio, PLx Pharma, Regado Biosciences; Board of directors: Boston VA Research Institute, Society of Cardiovascular Patient Care, TobeSoft; Chair: American Heart Association Quality Oversight Committee; Data monitoring committees: Baim Institute for Clinical Research (formerly Harvard Clinical Research Institute, for the PORTICO trial, funded by St. Jude Medical, now Abbott), Cleveland Clinic (including for the ExCEED trial, funded by Edwards), Contego Medical (Chair, PERFORMANCE 2), Duke Clinical Research Institute, Mayo Clinic, Mount Sinai School of Medicine (for the ENVISAGE trial, funded by Daiichi Sankyo), Population Health Research Institute; Honoraria: American College of Cardiology (Senior Associate Editor, Clinical Trials and News, ACC.org; Vice chair, ACC Accreditation Committee), Baim Institute for Clinical Research (formerly Harvard Clinical Research Institute; RE-DUAL PCI clinical trial steering committee funded by Boehringer Ingelheim; AEGIS-II executive committee funded by CSL Behring), Belvoir Publications (Editor in Chief, Harvard Heart Letter), Duke Clinical Research Institute (clinical trial steering committees, including for the PRONOUNCE trial, funded by Ferring Pharmaceuticals), HMP Global (Editor in Chief, Journal of Invasive Cardiology), Journal of the American College of Cardiology (Guest Editor; Associate Editor), K2P (Co-Chair, interdisciplinary curriculum), Level Ex, Medtelligence/ReachMD (CME steering committees), MJH Life Sciences, Population Health Research Institute (for the COMPASS operations committee, publications committee, steering committee, and USA national co-leader, funded by Bayer), Slack Publications (Chief Medical Editor, Cardiology Today's Intervention), Society of Cardiovascular Patient Care (Secretary/Treasurer), WebMD (CME steering committees); Other: Clinical Cardiology (Deputy Editor), NCDR-ACTION Registry Steering Committee (Chair), VA CART Research and Publications Committee (Chair); Research funding: Abbott, Afimmune, Amarin, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, Cardax, Chiesi, CSL Behring, Eisai, Ethicon, Ferring Pharmaceuticals, Forest Laboratories, Fractyl, Idorsia, Ironwood, Ischemix, Lexicon, Lilly, Medtronic, Pfizer, PhaseBio, PLx Pharma, Regeneron, Roche, Sanofi Aventis, Synaptic, The Medicines Company; Royalties: Elsevier (Editor, Cardiovascular Intervention: A Companion to Braunwald's Heart Disease); Site co-investigator: Biotronik, Boston Scientific, CSI, St. Jude Medical (now Abbott), Svelte; Trustee: American College of Cardiology; Unfunded research: FlowCo, Merck, Novo Nordisk, Takeda. Bays' research site has received research grants from 89Bio, Acasti, Akcea, Allergan, Alon Medtech/Epitomee, Amarin, Amgen, AstraZeneca, Axsome, Boehringer Ingelheim, Civi, Eli Lilly, Esperion, Evidera, Gan and Lee, Home Access, Janssen, Johnson and Johnson, Lexicon, Matinas, Merck, Metavant, Novartis, Novo Nordisk, Pfizer, Regeneron, Sanofi, Selecta, TIMI, and Urovant. Bays has served as a consultant/advisor for 89Bio, Amarin, Esperion, Matinas, and Gelesis, and speaker for Esperion. McCormick, Caldwell, Guerin, Ahmad, Singh, Moreo, Carter, Heggen, and Sapir have nothing to disclose.
Collapse
Affiliation(s)
| | - Deepak L Bhatt
- Brigham and Women’s Hospital Heart and Vascular Center and Harvard Medical School, Boston, MA
| | - Harold E Bays
- Louisville Metabolic and Atherosclerosis Research Center, Louisville, KY
| | - Pam R Taub
- Division of Cardiovascular Medicine, University of California San Diego School of Medicine
| | | | - Chris K Guerin
- Tri-City Medical Center and University of California San Diego School of Medicine
| | - Jeff Steinhoff
- Largo Medical Center, Largo, FL; HCA Healthcare/USF Morsani College of Medicine, Tampa, FL; and Nova Southeastern University, Davie, FL
| | - Zahid Ahmad
- Division of Nutrition and Metabolic Disease, UT Southwestern Medical Center, Dallas, TX
| | | | | | | | | | | |
Collapse
|
304
|
Sampietro T, Sbrana F, Bigazzi F, Ripoli A, Dal Pino B. Null receptor homozygous familial hypercholesterolaemia: Quoad valetudinem long life treatment. Eur J Prev Cardiol 2020; 27:2105-2108. [DOI: 10.1177/2047487319864191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
305
|
Cardiovascular Outcome of Pediatric Patients With Bi-Allelic (Homozygous) Familial Hypercholesterolemia Before and After Initiation of Multimodal Lipid Lowering Therapy Including Lipoprotein Apheresis. Am J Cardiol 2020; 136:38-48. [PMID: 32946862 DOI: 10.1016/j.amjcard.2020.09.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
Twenty-four patients with bi-allelic familial hypercholesterolemia commencing chronic lipoprotein apheresis (LA) at a mean age of 8.5 ± 3.1 years were analysed retrospectively and in part prospectively with a mean follow-up of 17.2 ± 5.6 years. Mean age at diagnosis was 6.3 ± 3.4 years. Untreated mean LDL-C concentrations were 752 mg/dl ± 193 mg/dl (19.5 mmol/l ± 5.0 mmol/l). Multimodal lipid lowering therapy including LA resulted in a mean LDL-C concentration of 184 mg/dl (4.8 mmol/l), which represents a 75.5% mean reduction. Proprotein convertase subtilisin/kexin type 9-antibodies contributed in 3 patients to LDL-C lowering with 5 patients remaining to be tested. After commencing chronic LA, 16 patients (67%) remained clinically stable with only subclinical findings of atherosclerotic cardiovascular disease (ASCVD), and neither cardiovascular events, nor need for vascular interventions or surgery. In 19 patients (79%), pathologic findings were detected at the aortic valve (AV), which in the majority were mild. AV replacement was required in 2 patients. Mean Lipoprotein(a) concentration was 42.4 mg/dl, 38% had >50 mg/dl. There was no overt correlation of AV pathologies with other ASCVD complications, or Lipoprotein(a) concentration. Physicochemical elimination of LDL particles by LA appears indispensable for patients with bi-allelic familial hypercholesterolemia and severe hypercholesterolemia to maximize the reduction of LDL-C. In conclusion, in this rare patient group regular assessment of both the AV, as well as all arteries accessible by ultrasound should be performed to adjust the intensity of multimodal lipid lowering therapy with the goal to prevent ASCVD events and aortic surgery.
Collapse
|
306
|
Abstract
Familial hypercholesterolemia (FH) is considered the genetic cause of coronary heart disease and ischemic stroke. FH is mainly an autosomal codominant pattern-based disorder and is primarily determined by point mutations within the low-density lipoprotein receptor, apolipoprotein B, and proprotein convertase subtilisin/kexin type 9 genes, causing increased low-density lipoprotein cholesterol levels in the serum of untreated individuals. The accumulation will eventually lead to atherosclerotic cardiovascular disease. Although clinical criteria comprising several prognosis scores, such as the Simon Broome, Dutch Lipid Clinic Network, Make Early Diagnosis to Prevent Early Death, and the recently proposed Montreal-FH-SCORE, are the conventional basis of diagnosing FH, the genetic diagnosis made by single nucleotide polymorphism genotyping, multiplex ligation-dependent probe amplification analysis, and sequencing (both Sanger and Next-Generation sequencing) offers unequivocal diagnosis. Given the heterogeneity of known mutations, the genetic diagnosis of FH is often difficult to establish, despite the growing evidence of the causative mutations, as well as the polygenic aspect of this pathology and the importance of cascade screening of the FH patient’s healthy family members. This review article details different genetic techniques that can be used in FH identification when there is a clinical FH suspicion based on criteria comprised in prognosis scores, knowing that none of these are exhaustive in the diagnosis, yet they efficaciously overlap and complement each other for confirming the disease at the molecular level.
Collapse
|
307
|
GENetic characteristics and REsponse to lipid-lowering therapy in familial hypercholesterolemia: GENRE-FH study. Sci Rep 2020; 10:19336. [PMID: 33168860 PMCID: PMC7653043 DOI: 10.1038/s41598-020-75901-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Among the 146 patients enrolled in the Korean FH registry, 83 patients who had undergone appropriate LLT escalation and were followed-up for ≥ 6 months were analyzed for pathogenic variants (PVs). The achieved percentage of expected low-density lipoprotein-cholesterol (LDL-C) reduction (primary variable) and achievement rates of LDL-C < 70 mg/dL were assessed. The correlations between the treatment response and the characteristics of PVs, and the weighted 4 SNP-based score were evaluated. The primary variables were significantly lower in the PV-positive patients than in the PV-negative patients (p = 0.007). However, the type of PV did not significantly correlate with the primary variable. The achievement rates of LDL-C < 70 mg/dL was very low, regardless of the PV characteristics. Patients with a higher 4-SNP score showed a lower primary variable (R2 = 0.045, p = 0.048). Among evolocumab users, PV-negative patients or those with only defective PVs revealed higher primary variable, whereas patients with at least one null PV showed lower primary variables. The adjusted response of patients with FH to LLT showed significant associations with PV positivity and 4-SNP score. These results may be helpful in managing FH patients with diverse genetic backgrounds.
Collapse
|
308
|
Blinc L, Mlinaric M, Battelino T, Groselj U. High-Sensitivity C-Reactive Protein and Carotid Intima Media Thickness as Markers of Subclinical Inflammation and Atherosclerosis in Pediatric Patients with Hypercholesterolemia. Molecules 2020; 25:E5118. [PMID: 33158028 PMCID: PMC7663751 DOI: 10.3390/molecules25215118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Hypercholesterolemia is a major cause of atherosclerosis development and premature cardiovascular disease (CVD). It leads to inflammation, which further accelerates atherosclerosis progression. Familial hypercholesterolemia (FH) is an autosomal dominant disorder characterized by elevated serum LDL-c from birth, due to a disease-causing variant in one of the causative genes (LDLR, APOB, PCSK9). In polygenic hypercholesterolemia (PH), the disease-causing genetic variant is absent; it is likely the cumulative result of multiple single nucleotide polymorphisms in LDL metabolism-related genes and other factors, such as lifestyle and environment. In high risk groups, such as patients with FH, an effective primary prevention of CVD must begin in childhood. High-sensitivity C-reactive protein (hsCRP) and carotid intima media thickness (cIMT) are two potential minimally invasive correlates of inflammation and subclinical atherosclerosis progression. hsCRP and cIMT have been shown to be significantly increased in patients with FH and PH relative to healthy controls, with some studies yielding conflicting results. In this review, we aim to summarize current knowledge and recent findings regarding the applicability of hsCRP and cIMT as markers of low-grade inflammation and subclinical atherosclerosis, focusing especially on children and adolescents with hypercholesterolemia.
Collapse
Affiliation(s)
- Lana Blinc
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (L.B.); (T.B.)
| | - Matej Mlinaric
- University Children’s Hospital, University Medical Center Ljubljana, Bohoriceva ulica 20, 1000 Ljubljana, Slovenia;
| | - Tadej Battelino
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (L.B.); (T.B.)
- University Children’s Hospital, University Medical Center Ljubljana, Bohoriceva ulica 20, 1000 Ljubljana, Slovenia;
| | - Urh Groselj
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (L.B.); (T.B.)
- University Children’s Hospital, University Medical Center Ljubljana, Bohoriceva ulica 20, 1000 Ljubljana, Slovenia;
| |
Collapse
|
309
|
Iqbal Z, Ho JH, Adam S, France M, Syed A, Neely D, Rees A, Khatib R, Cegla J, Byrne C, Qureshi N, Capps N, Ferns G, Payne J, Schofield J, Nicholson K, Datta D, Pottle A, Halcox J, Krentz A, Durrington P, Soran H, Heart UK's Medical Scientific and Research Committee. Managing hyperlipidaemia in patients with COVID-19 and during its pandemic: An expert panel position statement from HEART UK. Atherosclerosis 2020; 313:126-136. [PMID: 33045618 PMCID: PMC7490256 DOI: 10.1016/j.atherosclerosis.2020.09.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
Abstract
The emergence of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which causes Coronavirus Disease 2019 (COVID-19) has resulted in a pandemic. SARS-CoV-2 is highly contagious and its severity highly variable. The fatality rate is unpredictable but is amplified by several factors including advancing age, atherosclerotic cardiovascular disease, diabetes mellitus, hypertension and obesity. A large proportion of patients with these conditions are treated with lipid lowering medication and questions regarding the safety of continuing lipid-lowering medication in patients infected with COVID-19 have arisen. Some have suggested they may exacerbate their condition. It is important to consider known interactions with lipid-lowering agents and with specific therapies for COVID-19. This statement aims to collate current evidence surrounding the safety of lipid-lowering medications in patients who have COVID-19. We offer a consensus view based on current knowledge and we rated the strength and level of evidence for these recommendations. Pubmed, Google scholar and Web of Science were searched extensively for articles using search terms: SARS-CoV-2, COVID-19, coronavirus, Lipids, Statin, Fibrates, Ezetimibe, PCSK9 monoclonal antibodies, nicotinic acid, bile acid sequestrants, nutraceuticals, red yeast rice, Omega-3-Fatty acids, Lomitapide, hypercholesterolaemia, dyslipidaemia and Volanesorsen. There is no evidence currently that lipid lowering therapy is unsafe in patients with COVID-19 infection. Lipid-lowering therapy should not be interrupted because of the pandemic or in patients at increased risk of COVID-19 infection. In patients with confirmed COVID-19, care should be taken to avoid drug interactions, between lipid-lowering medications and drugs that may be used to treat COVID-19, especially in patients with abnormalities in liver function tests.
Collapse
Affiliation(s)
- Zohaib Iqbal
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jan Hoong Ho
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Safwaan Adam
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom,The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Michael France
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Akheel Syed
- Department of Diabetes, Endocrinology and Obesity Medicine, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Dermot Neely
- Department of Blood Sciences and NIHR MedTech and IVD Centre, Newcastle Upon Tyne Hospitals, Newcastle Upon Tyne, United Kingdom
| | - Alan Rees
- HEART UK, Maidenhead, United Kingdom
| | - Rani Khatib
- Departments of Cardiology & Pharmacy, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom,Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Jaimini Cegla
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, Du Cane Road, W12 0NN, London, United Kingdom
| | - Christopher Byrne
- Department of Nutrition and Metabolism, Faculty of Medicine, University of Southampton, United Kingdom
| | - Nadeem Qureshi
- Division of Primary Care, University of Nottingham, Nottingham, United Kingdom
| | - Nigel Capps
- The Shrewsbury and Telford Hospital NHS Trust, United Kingdom
| | - Gordon Ferns
- Department of Medical Education, Brighton and Sussex Medical School, Brighton, United Kingdom
| | | | - Jonathan Schofield
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kirsty Nicholson
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Dev Datta
- Department of Metabolic Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Alison Pottle
- Department of Cardiology, Harefield Hospital, United Kingdom
| | - Julian Halcox
- Department of Medicine, Swansea University, Swansea, United Kingdom
| | - Andrew Krentz
- Institute of Cardiovascular & Metabolic Research, University of Reading, United Kingdom
| | - Paul Durrington
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Handrean Soran
- Cardiovascular Trials Unit, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.
| | | |
Collapse
|
310
|
Underberg JA, Cannon CP, Larrey D, Makris L, Blom D, Phillips H. Long-term safety and efficacy of lomitapide in patients with homozygous familial hypercholesterolemia: Five-year data from the Lomitapide Observational Worldwide Evaluation Registry (LOWER). J Clin Lipidol 2020; 14:807-817. [DOI: 10.1016/j.jacl.2020.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022]
|
311
|
Bertolini S, Calandra S, Arca M, Averna M, Catapano AL, Tarugi P, Bartuli A, Bucci M, Buonuomo PS, Calabrò P, Casula M, Cefalù AB, Cicero A, D'Addato S, D'Erasmo L, Fasano T, Iannuzzo G, Ibba A, Negri EA, Pasta A, Pavanello C, Pisciotta L, Rabacchi C, Ripoli C, Sampietro T, Sbrana F, Sileo F, Suppressa P, Trenti C, Zenti MG. Homozygous familial hypercholesterolemia in Italy: Clinical and molecular features. Atherosclerosis 2020; 312:72-78. [DOI: 10.1016/j.atherosclerosis.2020.08.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/07/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
|
312
|
Mahdavi A, Bagherniya M, Fakheran O, Reiner Ž, Xu S, Sahebkar A. Medicinal plants and bioactive natural compounds as inhibitors of HMG-CoA reductase: A literature review. Biofactors 2020; 46:906-926. [PMID: 33053603 DOI: 10.1002/biof.1684] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 08/15/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVDs) are one of the most important causes for mortality worldwide. Elevated levels of total cholesterol, and particularly LDL-cholesterol (LDL-C) are the main risk factor for acute myocardial infarction (AMI) and ischemic heart disease. The risk of CVDs could be reduced by decreasing the elevated cholesterol levels. β-hydroxy β-methylglutaryl-CoA reductase (HMGCoAR) is the primary and rate-limiting enzyme in the cholesterol biosynthesis pathway. Recently, the crucial role of nutraceuticals in maintaining normal physiological function was established. Nutraceuticals play an important role in preventing several non-communicable diseases such as obesity, CVDs, cancer, diabetes, and reducing hyperlipidemia. Although the effect of nutraceuticals and herbal medicine on CVDs and dyslipidemia was previously investigated thoroughly, the effect of these natural products on HMGCoAR as one of the important enzymes involved in CVDs etiopathogenesis has not yet been investigated. Therefore, the major aim of this paper was to review the effects of nutraceuticals and medicinal plants on HMGCoAR. Results indicate that different types of natural foods, isolated nutrients, herbal products, and dietary supplements as nutraceuticals decrease the expression and activity of HMGCoAR. This review shows that medicinal plants and nutraceuticals could be used to decrease HMGCoAR activity as accessible and convenient and economical natural compounds to prevent dyslipidemia and CVDs.
Collapse
Affiliation(s)
- Atena Mahdavi
- Department of Community Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Omid Fakheran
- Dental research center, Department of Periodontics, Dental research institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, Zagreb, Croatia
| | - Suowen Xu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Halal Research Center of IRI, FDA, Tehran, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| |
Collapse
|
313
|
Leclercq T, Falcon-Eicher S, Farnier M, Le Bret E, Maudinas R, Litzler-Renault S, Mousson C, Lorgis L, Cottin Y. A case report of an acute coronary syndrome in a 10-year-old boy with homozygous familial hypercholesterolaemia. EUROPEAN HEART JOURNAL-CASE REPORTS 2020; 4:1-5. [PMID: 33123673 PMCID: PMC7574956 DOI: 10.1093/ehjcr/ytz233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/30/2019] [Accepted: 12/04/2019] [Indexed: 11/28/2022]
Abstract
Background Familial hypercholesterolaemia is a well-known disorder, but clinical diagnoses tend to be delayed. Acute coronary syndrome may occur in childhood. Case summary Our patient, a young boy with homozygous familial hypercholesterolaemia, complained of persistent chest pain at rest and suffered a non-ST-elevation myocardial infarction (NSTEMI). The diagnosis of NSTEMI was made on the basis of his clinical features, dynamic electrocardiogram changes, troponin elevation, and cardiac computed tomography findings. The patient was managed surgically by intrathoracic artery (ITA) bypass graft. During post-operative follow-up, the young patient suffered from angina pectoris from unexpected and exceptional atheroma stenosis on the ITA. Discussion Familial hypercholesterolaemia needs to be identified quickly in young patients and lipid lowering therapies should be started without delay.
Collapse
Affiliation(s)
- Thibault Leclercq
- Department of Cardiology, Dijon University Hospital, 21000 Dijon, France
| | | | - Michel Farnier
- Department of Cardiology, Dijon University Hospital, 21000 Dijon, France.,Lipid Clinic, Point Médical, 21000 Dijon, France
| | - Emmanuel Le Bret
- Department of Congenital Heart Surgery, Marie Lannelongue Hospital, 92350 Le Plessis Robinson, France
| | - Raphaëlle Maudinas
- Department of Pediatrics, Dijon University Hospital, 21000 Dijon, France
| | | | - Christiane Mousson
- Department of Nephrology, Dijon University Hospital, 21000 Dijon, France
| | - Luc Lorgis
- Department of Cardiology, Dijon University Hospital, 21000 Dijon, France
| | - Yves Cottin
- Department of Cardiology, Dijon University Hospital, 21000 Dijon, France
| |
Collapse
|
314
|
Harada-Shiba M, Ali S, Gipe DA, Gasparino E, Son V, Zhang Y, Pordy R, Catapano AL. A randomized study investigating the safety, tolerability, and pharmacokinetics of evinacumab, an ANGPTL3 inhibitor, in healthy Japanese and Caucasian subjects. Atherosclerosis 2020; 314:33-40. [PMID: 33130482 DOI: 10.1016/j.atherosclerosis.2020.10.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Evinacumab, an angiopoietin-like protein 3 monoclonal antibody, reduced low-density lipoprotein cholesterol (LDL-C) significantly in a Phase 2 study of patients with homozygous familial hypercholesterolemia. In this double-blind, placebo-controlled Phase 1 study, we compared safety, tolerability, pharmacokinetics, and pharmacodynamics of evinacumab between healthy Japanese and Caucasian adults. METHODS Subjects with LDL-C ≥2.6 and <4.1 mmol/L were enrolled to one of four dose cohorts: evinacumab subcutaneous (SC) 300 mg single dose, SC 300 mg once weekly for eight doses, intravenous (IV) 5 mg/kg, or IV 15 mg/kg once every 4 weeks for two doses. Each cohort comprised 24 subjects (12 Japanese; 12 Caucasian), randomized (3:1) to receive evinacumab or placebo within each ethnic group with a 24-week follow-up. RESULTS The safety profile of evinacumab (IV and SC) in both ethnicities was comparable with placebo, with no serious or severe treatment-emergent adverse events. Pharmacokinetic profiles were comparable between Japanese and Caucasian subjects across IV and SC groups. Mean calculated LDL-C decreased from baseline with both IV doses, beginning on day 3 up to week 8. Triglyceride changes observed with evinacumab IV were rapid (seen by day 2) and sustained up to week 8. Evinacumab SC doses also reduced LDL-C and triglyceride levels, although lower doses induced smaller changes. Evinacumab (IV and SC) reduced other lipids, including apolipoprotein B, versus placebo. CONCLUSIONS In both ethnicities, evinacumab (IV and SC) was generally well tolerated, exhibiting comparable pharmacokinetic profiles. Dose-related reductions in LDL-C and triglycerides were observed with evinacumab in both ethnic groups.
Collapse
Affiliation(s)
- Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | - Shazia Ali
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, USA
| | | | | | - Vladimir Son
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, USA
| | - Yi Zhang
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, USA
| | - Robert Pordy
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences University of Milan and IRCCS Multimedica, Milan, Italy
| |
Collapse
|
315
|
Vrablik M, Tichý L, Freiberger T, Blaha V, Satny M, Hubacek JA. Genetics of Familial Hypercholesterolemia: New Insights. Front Genet 2020; 11:574474. [PMID: 33133164 PMCID: PMC7575810 DOI: 10.3389/fgene.2020.574474] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
Familial hypercholesterolemia (FH) is one of the most common monogenic diseases, leading to an increased risk of premature atherosclerosis and its cardiovascular complications due to its effect on plasma cholesterol levels. Variants of three genes (LDL-R, APOB and PCSK9) are the major causes of FH, but in some probands, the FH phenotype is associated with variants of other genes. Alternatively, the typical clinical picture of FH can result from the accumulation of common cholesterol-increasing alleles (polygenic FH). Although the Czech Republic is one of the most successful countries with respect to FH detection, approximately 80% of FH patients remain undiagnosed. The opportunities for international collaboration and experience sharing within international programs (e.g., EAS FHSC, ScreenPro FH, etc.) will improve the detection of FH patients in the future and enable even more accessible and accurate genetic diagnostics.
Collapse
Affiliation(s)
- Michal Vrablik
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Lukas Tichý
- Centre of Molecular Biology and Gene Therapy, University Hospital, Brno, Czechia
| | - Tomas Freiberger
- Centre for Cardiovascular Surgery and Transplantation, Brno, and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Vladimir Blaha
- Internal Gerontometabolic Department, Charles University and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Martin Satny
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Jaroslav A Hubacek
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia.,Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| |
Collapse
|
316
|
Abrignani MG. Evaluating statin effect on LDL-cholesterol: when predicted is not measured. Eur J Prev Cardiol 2020; 27:1627-1629. [PMID: 31615292 DOI: 10.1177/2047487319882818] [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] [Indexed: 02/05/2023]
|
317
|
Al-Rasadi K, Alhabib KF, Al-Allaf F, Al-Waili K, Al-Zakwani I, AlSarraf A, Almahmeed W, AlSayed N, Alghamdi M, Batais MA, Almigbal TH, Alnouri F, Kinsara A, Hammouda A, Awan Z, Kary H, Elamin OA, Zadjali F, Al-Jarallah M, Shehab A, Sabbour H, Amin H, Altaradi H. The Gulf Familial Hypercholesterolemia Registry (Gulf FH): Design, Rationale and Preliminary Results. Curr Vasc Pharmacol 2020; 18:57-64. [PMID: 30289081 PMCID: PMC7040511 DOI: 10.2174/1570161116666181005125459] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/01/2018] [Accepted: 10/01/2018] [Indexed: 01/05/2023]
Abstract
Aim: To determine the prevalence, genetic characteristics, current management and outcomes of familial hypercholesterolaemia (FH) in the Gulf region. Methods: Adult (18-70 years) FH patients were recruited from 9 hospitals and centres across 5 Arabian Gulf countries. The study was divided into 4 phases and included patients from 3 different categories. In phase 1, suspected FH patients (category 1) were collected according to the lipid profile and clinical data obtained through hospital record systems. In phase 2, patients from category 2 (patients with a previous clinical diagnosis of FH) and category 1 were stratified into definitive, probable and possible FH according to the Dutch Lipid Clinic Network criteria. In phase 3, 500 patients with definitive and probable FH from categories 1 and 2 will undergo genetic testing for 4 common FH genes. In phase 4, these 500 patients with another 100 patients from category 3 (patients with previous genetic diagnosis of FH) will be followed for 1 year to evaluate clinical management and cardiovascular outcomes. The Gulf FH cohort was screened from a total of 34,366 patients attending out-patient clinics. Results: The final Gulf FH cohort consisted of 3,317 patients (mean age: 47±12 years, 54% females). The number of patients with definitive FH is 203. In this initial phase of the study, the prevalence of (probable and definite) FH is 1/232. Conclusion: The prevalence of FH in the adult population of the Arabian Gulf region is high. The Gulf FH registry, a first-of-a-kind multi-national study in the Middle East region, will help in improving underdiagnosis and undertreatment of FH in the region.
Collapse
Affiliation(s)
- Khalid Al-Rasadi
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Khalid F Alhabib
- Department of Cardiac Sciences, King Fahad Cardiac Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Faisal Al-Allaf
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, Makka, Saudi Arabia
| | - Khalid Al-Waili
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Ibrahim Al-Zakwani
- Department of Pharmacology & Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman.,Gulf Health Research, Muscat, Oman
| | - Ahmad AlSarraf
- Department of Medical Biochemistry, Ministry of Health, Kuwait City, Safat, Kuwait
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | | | | | - Mohammed A Batais
- Department of Family and Community Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Turky H Almigbal
- Department of Family and Community Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fahad Alnouri
- Cardiovascular Prevention Unit, Prince Sultan Cardiac Centre, Riyadh, Saudi Arabia
| | - Abdulhalim Kinsara
- King Saud bin Abdulaziz University for Health Sciences, College of Medicine, King Abdul Aziz Medical City, Jeddah, Saudi Arabia
| | | | - Zuhier Awan
- King Abdulaziz University, Jeddah, Saudi Arabia
| | - Heba Kary
- King Faisal Specialist Hospital & Research Center, Jeddah, Saudi Arabia
| | - Omer A Elamin
- King Faisal Specialist Hospital & Research Center, Jeddah, Saudi Arabia
| | - Fahad Zadjali
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | | | - Abdullah Shehab
- Department of Internal Medicine, College of Medicine and Health Sciences, UAE; University, Al Ain, United Arab Emirates
| | - Hani Sabbour
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | | | - Hani Altaradi
- Department of Cardiac Sciences, King Fahad Cardiac Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
318
|
Marusic T, Sustar U, Sadiq F, Kotori V, Mlinaric M, Kovac J, Shafi S, Khan I, Cevc M, Trebusak Podkrajsek K, Battelino T, Groselj U. Genetic and Clinical Characteristics of Patients With Homozygous and Compound Heterozygous Familial Hypercholesterolemia From Three Different Populations: Case Series. Front Genet 2020; 11:572176. [PMID: 33093846 PMCID: PMC7528874 DOI: 10.3389/fgene.2020.572176] [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] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/20/2020] [Indexed: 11/17/2022] Open
Abstract
Homozygous familial hypercholesterolemia (HoFH) and compound heterozygous familial hypercholesterolemia (cHeFH) are rare disorders generated by disease-causing variants in both alleles of the LDLR or other familial hypercholesterolemia (FH)-related genes. HoFH and cHeFH are characterized by severely elevated low-density lipoprotein-cholesterol (LDL-C), frequently leading to early cardiovascular disease. We investigated the genetic and clinical characteristics of HoFH and cHeFH patients from the Slovenian FH registry and/or those who were previously diagnosed or managed at our institution (Slovenian, Pakhtun and Albanian ethnicity), where genetic testing is not available. Our study includes seven patients. Their median age at the time of clinical diagnosis was 6.3 years (2.9-12.9 years); 2/7 were females. Two patients were diagnosed through the universal FH screening and five patients were diagnosed due to the presence of xanthomas. All the mutations are present in LDLR gene: 7 different genotypes for HoFH (p.Cys167Leu, p.Asp178Asn, p.Cys243Tyr, p.Gly549Asp, p.Cys27Trp, p.Ile585Thr and p.Val797Met) and p.Gly549Asp/p.Gln384Pro genotype for cHeFH patient. The median initial level of LDL-C was 17.0 mmol/L [655 mg/dL] (range 7.6-21.6 mmol/L). The HoFH/cHeFH patients are clinically and genetically very diverse. The clinical criteria (as Simon Broome criteria) might be applicable already in children to raise suspicion of FH but in some cases fail to distinguish heterozygous FH and HoFH/cHeFH patients. However, genetic testing is helpful in confirming the diagnosis, also for a prompt awareness, better compliance to treatment and family screening.
Collapse
Affiliation(s)
- Tatiana Marusic
- University Children’s Hospital, University Medical Center, Ljubljana, Slovenia
| | - Ursa Sustar
- University Children’s Hospital, University Medical Center, Ljubljana, Slovenia
| | - Fouzia Sadiq
- Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Vjosa Kotori
- Department of Endocrinology, Pediatric Clinic, University Clinical Center of Kosovo, Pristina, Kosovo
| | - Matej Mlinaric
- University Children’s Hospital, University Medical Center, Ljubljana, Slovenia
| | - Jernej Kovac
- University Children’s Hospital, University Medical Center, Ljubljana, Slovenia
| | - Saeed Shafi
- Department of Anatomy, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Iqbal Khan
- Shifa Tameer-e-Millat University, Islamabad, Pakistan
- Department of Vascular Surgery, Shifa International Hospital, Islamabad, Pakistan
| | - Matija Cevc
- Division of Medicine, Centre for Preventive Cardiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Katarina Trebusak Podkrajsek
- University Children’s Hospital, University Medical Center, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tadej Battelino
- University Children’s Hospital, University Medical Center, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Urh Groselj
- University Children’s Hospital, University Medical Center, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
319
|
Abstract
PURPOSE OF REVIEW Cardiovascular disease (CVD) can begin in youth. Prevention is essential to reducing the burden of CVD-related risk factors in childhood and disease development in adulthood. This review addresses the clinical scope of CVD prevention, including a review of conditions encountered, proposed diagnostic criteria, and management strategies. We also highlight the impact of the intrauterine environment on the development of CVD risk. Finally, we highlight the potential role of telehealth in the management of pediatric patients with risk factors for premature CVD. RECENT FINDINGS Growing evidence suggests that maternal obesity, diabetes, and preeclampsia may play an important role in the development of CVD risk among offspring contributing to the development of known traditional CVD risk factors among offspring. As the prevalence of CVD continues to rise, knowledge as well as appropriate diagnosis and management of primordial and traditional risk factors for CVD is needed. The diagnosis and management of CVD risk factors is a central role of the preventive pediatric cardiologist, but it is imperative that the general physician and other pediatric subspecialists be aware of these risk factors, diagnoses, and management strategies. Finally, telehealth may offer an additional method for providing preventive care, including screening and counseling of at risk children and adolescents for traditional risk factors and for providing education regarding risk factors in cases of long distance care and/or during periods of social distancing.
Collapse
|
320
|
Roy A, Kamalanathan S, Naik D, Sahoo JP. Extensive tendon and tuberous xanthomas in a patient with familial hypercholesterolaemia. BMJ Case Rep 2020; 13:e236759. [PMID: 32900739 PMCID: PMC7478013 DOI: 10.1136/bcr-2020-236759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2020] [Indexed: 11/03/2022] Open
Affiliation(s)
- Ayan Roy
- Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Sadishkumar Kamalanathan
- Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Dukhabandhu Naik
- Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Jaya Prakash Sahoo
- Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
321
|
Raal FJ, Bahassi EM, Stevens B, Turner TA, Stein EA. Cascade Screening for Familial Hypercholesterolemia in South Africa: The Wits FIND-FH Program. Arterioscler Thromb Vasc Biol 2020; 40:2747-2755. [PMID: 32878475 DOI: 10.1161/atvbaha.120.315040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Due to gene founder effects, familial hypercholesterolemia (FH) has a prevalence of ≈1:80 in populations of Afrikaner ancestry and is a major contributor to premature atherosclerotic cardiovascular disease in South Africans of Jewish and Indian descent. No systematic program exists to identify these families. Furthermore, information regarding FH prevalence in Black Africans is sparse. The Wits FIND-FH program was initiated in late 2016 to address these issues. Approach and Results: Based on index subjects with definite or probable FH, first-degree relatives were contacted, informed consent obtained, and targeted medical history, physical examination, and blood samples collected. In patients with likely FH using the Simon Broome criteria, DNA analysis for LDLR (low-density lipoprotein receptor), APOB (apolipoprotein B), PCSK9 (proprotein convertase subtilisin/kexin type 9), and LDLRAP1 (LDLR adaptor protein 1) variants was analyzed by next-generation sequencing. Of the initial 700 subjects screened of whom 295 (42%) were index cases, 479 (68.4%) were clinically diagnosed with probable or definite FH. Genetic analysis confirmed 285 of 479 (59.5%) as having variants consistent with FH. Three subjects met the clinical diagnosis for homozygous FH, but DNA analysis revealed a further 34 patients, including 4 Black African subjects, with ≥2 FH-causing variants. CONCLUSIONS Using phenotype cascade screening, the Wits FIND-FH program has screened an average of 30 subjects monthly of whom 68% had a clinical diagnosis of FH with ≈60% genetically confirmed. The program is identifying a small but growing number of Black South Africans with FH. Interestingly, 37 subjects (7.7%) who underwent DNA testing were found to have ≥2 FH-causing variants.
Collapse
Affiliation(s)
- Frederick J Raal
- Department of Medicine, Stein Center for FH, Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (F.J.R., B.S.)
| | - El Mustapha Bahassi
- Medpace and Medpace Reference Laboratories, Cincinnati, OH (E.M.B., T.A.T., E.A.S.)
| | - Belinda Stevens
- Department of Medicine, Stein Center for FH, Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (F.J.R., B.S.)
| | - Traci A Turner
- Medpace and Medpace Reference Laboratories, Cincinnati, OH (E.M.B., T.A.T., E.A.S.)
| | - Evan A Stein
- Medpace and Medpace Reference Laboratories, Cincinnati, OH (E.M.B., T.A.T., E.A.S.)
| |
Collapse
|
322
|
Rieck L, Bardey F, Grenkowitz T, Bertram L, Helmuth J, Mischung C, Spranger J, Steinhagen-Thiessen E, Bobbert T, Kassner U, Demuth I. Mutation spectrum and polygenic score in German patients with familial hypercholesterolemia. Clin Genet 2020; 98:457-467. [PMID: 32770674 DOI: 10.1111/cge.13826] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022]
Abstract
Autosomal-dominant familial hypercholesterolemia (FH) is characterized by increased plasma concentrations of low-density lipoprotein cholesterol (LDL-C) and a substantial risk to develop cardiovascular disease. Causative mutations in three major genes are known: the LDL receptor gene (LDLR), the apolipoprotein B gene (APOB) and the proprotein convertase subtilisin/kexin 9 gene (PCSK9). We clinically characterized 336 patients suspected to have FH and screened them for disease causing mutations in LDLR, APOB, and PCSK9. We genotyped six single nucleotide polymorphisms (SNPs) to calculate a polygenic risk score for the patients and 1985 controls. The 117 patients had a causative variant in one of the analyzed genes. Most variants were found in the LDLR gene (84.9%) with 11 novel mutations. The mean polygenic risk score was significantly higher in FH mutation negative subjects than in FH mutation positive patients (P < .05) and healthy controls (P < .001), whereas the score of the two latter groups did not differ significantly. However, the score explained only about 3% of the baseline LDL-C variance. We verified the previously described clinical and genetic variability of FH for German hypercholesterolemic patients. Evaluation of a six-SNP polygenic score recently proposed for clinical use suggests that it is not a reliable tool to classify hypercholesterolemic patients.
Collapse
Affiliation(s)
- Lorenz Rieck
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Frieda Bardey
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Grenkowitz
- Department of Cardiology, Charité - University Medicine Berlin (Campus Benjamin Franklin), Berlin, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany.,Center for Lifespan Changes in Brain and Cognition (LCBC), Dept of Psychology, University of Oslo, Oslo, Norway
| | - Johannes Helmuth
- Department Molecular Diagnostics, Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Claudia Mischung
- Department Molecular Diagnostics, Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Bobbert
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ursula Kassner
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ilja Demuth
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Charité - Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
323
|
Khoury M, McCrindle BW. The Rationale, Indications, Safety, and Use of Statins in the Pediatric Population. Can J Cardiol 2020; 36:1372-1383. [PMID: 32735868 DOI: 10.1016/j.cjca.2020.03.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 10/24/2022] Open
Abstract
Together with heart-healthy lifestyle habits, statins serve as the cornerstone of primary and secondary prevention of atherosclerotic cardiovascular disease in adults. Several conditions, most notably familial hypercholesterolemia (FH), cause early dyslipidemia and vascular disease, contributing to the development and progression of atherosclerosis from childhood and increased cardiovascular risk. In recent decades, studies increasingly have evaluated the safety and efficacy of statins in such high-risk youth. The strongest evidence for pediatric statin use is for the heterozygous FH population, whereby statin use has been shown to lower low-density lipoprotein cholesterol effectively, slow the progression of atherosclerosis and vascular dysfunction, and significantly reduce cardiovascular risk in early adulthood. Numerous meta-analyses and Cochrane reviews have demonstrated that attributed adverse effects, including liver toxicity, myositis, and rhabdomyolysis, occur no more frequently in youth receiving statins than placebos, with no impact on growth or development. However, further studies evaluating the long-term safety of pediatric statin use are required. In the current review, we summarize the pediatric experience of statin use to date, focusing on its utility for FH, Kawasaki disease, post-heart transplantation, and other at-risk populations. Current guidelines and indications for use are summarized, and the short- and medium-term safety experience is reviewed. Finally, a clinical approach to the indications, initiation, and monitoring of statins in youth is provided.
Collapse
Affiliation(s)
- Michael Khoury
- Division of Pediatric Cardiology, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| | - Brian W McCrindle
- Labatt Family Heart Center, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
324
|
Gulizia MM, Maggioni AP, Abrignani MG, Bilato C, Mangiacapra F, Sanchez FA, Piovaccari G, Montagna L, Marini M, De Biasio M, Averna M, Casula M, Colivicchi F, Fabbri G, Lucci D, Zampoleri V, Catapano AL. Prevalence Of familial hypercholeSTerolaemia (FH) in Italian Patients with coronary artERy disease: The POSTER study. Atherosclerosis 2020; 308:32-38. [PMID: 32823190 DOI: 10.1016/j.atherosclerosis.2020.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Familial hypercholesterolaemia (FH) is a powerful risk factor for cardiovascular (CV) events. High levels of low-density lipoprotein cholesterol (LDL-C) since birth are linked to the early onset of atherosclerotic disease. A genetic mutation determining FH is present in about one subject out of 250; FH should be more represented among subjects with a documented diagnosis of coronary artery disease (CAD). The POSTER Study evaluated the prevalence of FH in Italian patients with a recent CAD event. METHODS Eighty-two cardiology centres enrolled patients with a documented CAD event; CV risk profile, drug therapy and biochemical parameters were collected. Dutch Lipid Clinic Network (DLCN) criteria were used to define patients with a potential FH diagnosis (score ≥6); these patients underwent molecular testing for genetic diagnosis of FH. RESULTS Overall, 5415 patients were enrolled and the main index events were myocardial infarction with ST-elevation, non ST-elevation acute coronary syndrome (ACS), or a recent coronary revascularization (34.8%, 37.2%, and 28% respectively). Mean age was 66 ± 11 years, men were 78%; about 40% were already treated with statins, proportion that increased after the acute event (96.5%). Based on the DLCN score, the prevalence of potential FH was 5.1%, 0.9% of them had a diagnosis of definite FH (score >8). These patients were younger than patients with a score <6 (56 ± 10 vs 66 ± 11, p < 0.001), and LDL-C levels were in most of them (~87%) >190 mg/dL. FH was genetically confirmed in 42 subjects (15.9%); genetic diagnosis was defined as not conclusive for FH in 63 patients (23.9%). Finally, in 159 subjects (60.2%) no pathogenic mutations in the tested genes were identified, defining them as negative for monogenic familial hypercholesterolemia. CONCLUSIONS Results underscore a relatively high prevalence of potential FH in patients with a recent CAD event. Therefore, an early identification of these subjects may help improve the management of their high CV risk and, by cascade screening, identify possible FH relatives.
Collapse
Affiliation(s)
- Michele Massimo Gulizia
- Cardiology Division, Ospedale Garibaldi-Nesima, Azienda di Rilievo Nazionale e Alta Specializzazione "Garibaldi" Catania, Italy; Heart Care Foundation, Florence, Italy.
| | - Aldo P Maggioni
- ANMCO Research Center of the Heart Care Foundation, Florence, Italy.
| | | | - Claudio Bilato
- Division of Cardiology, West Vicenza General Hospitals, Arzignano, Italy
| | | | | | | | - Laura Montagna
- Cardiology Department, Azienda Ospedaliero-Universitaria San Luigi Gonzaga, Orbassano, Italy
| | - Marco Marini
- Cardiology Department, Riuniti Hospital, Ancona, Italy
| | - Marzia De Biasio
- Cardiology Department, Santa Maria Della Misericordia Hospital, Udine, Italy
| | - Maurizio Averna
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Manuela Casula
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | | | - Gianna Fabbri
- ANMCO Research Center of the Heart Care Foundation, Florence, Italy
| | - Donata Lucci
- ANMCO Research Center of the Heart Care Foundation, Florence, Italy
| | - Veronica Zampoleri
- Department of Excellence of Pharmacological and Biomolecular Sciences (DisFeB), University of Milan, Milan, Italy; SISA Centre, Bassini Hospital, Cinisello Balsamo, Italy
| | - Alberico L Catapano
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy; Department of Excellence of Pharmacological and Biomolecular Sciences (DisFeB), University of Milan, Milan, Italy
| |
Collapse
|
325
|
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
|
326
|
Affiliation(s)
- Sander Kersten
- From the Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| |
Collapse
|
327
|
Sreedharan AV, Pek SLT, Tan TH, Tavintharan S, Yap F. Successful pharmacological management of a child with compound heterozygous familial hypercholesterolemia and review of the recent literature. J Clin Lipidol 2020; 14:639-645. [PMID: 32800790 DOI: 10.1016/j.jacl.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/25/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022]
Abstract
Severe familial hypercholesterolemia (SFH) is characterized by markedly elevated low-density lipoprotein cholesterol (LDL-C) and severe early-onset cardiovascular disease if left untreated. We report on the decade-long therapeutic journey of a 15-year-old boy with SFH due to a severe compound heterozygous genotype. He presented at the age of 5 years with widespread xanthomas and LDL-C of 17.4 mmol/L. He was diagnosed with SFH, initially treated with colestyramine that was subsequently combined with simvastatin. At the age of 12 years, he was diagnosed to have supravalvular aortic stenosis and ezetimibe/atorvastatin was introduced in place of colestyramine/simvastatin. At the age of 14 years, he received triple therapy with evolocumab, initially at the recommended dose of 420 mg monthly and then reduced to 140 mg biweekly. Currently at the age of 15 years, he is on atorvastatin 40 mg ON, ezetimibe 10 mg OM, and evolocumab 140 mg biweekly, achieving LDL-C levels of 2.4 mmol/L. Genetic testing identified compound heterozygous mutations in the LDL receptor genes [c.(940 + 1_941-1) (1845 + 1_1846-1)dup] and exon 12, nucleotide c.1747 C > T, amino acid p.(His583Tyr). Medical management without lipoprotein apheresis can achieve target LDL-C in children with SFH. Our patient, who developed supravalvular aortic stenosis at the age of 12 years, needed early aggressive treatment when SFH guidelines and newer drugs for young children were unavailable. Our patient demonstrated that 140 mg biweekly of evolocumab has the same cholesterol-lowering effect as the recommended 420 mg monthly dose.
Collapse
Affiliation(s)
- Aravind Venkatesh Sreedharan
- Division of Medicine, Department of Paediatric Endocrinology, KK Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | | | - Teng Hong Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Division of Medicine, Department of Paediatric Cardiology, KK Hospital, Singapore, Singapore
| | - Subramaniam Tavintharan
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore; Diabetes Centre, Admiralty Medical Centre, Singapore, Singapore; Division of Endocrinology, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Fabian Yap
- Division of Medicine, Department of Paediatric Endocrinology, KK Hospital, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
328
|
Truong TH, Do DL, Kim NT, Nguyen MNT, Le TT, Le HA. Genetics, Screening, and Treatment of Familial Hypercholesterolemia: Experience Gained From the Implementation of the Vietnam Familial Hypercholesterolemia Registry. Front Genet 2020; 11:914. [PMID: 32922439 PMCID: PMC7457124 DOI: 10.3389/fgene.2020.00914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Familial hypercholesterolemia (FH) is underdiagnosed and undertreated in a majority of the low- and middle-income countries. FH registries could prove useful in bridging the knowledge gaps, supporting genetic and clinical research, and improving health-care planning and patient care. Here, we report the first usage experience of the Vietnam FH (VINAFH) Registry. The VINAFH Registry was established in 2016 as a long-term database for prospective cohorts. FH patients were detected based on the opportunistic and cascade screening. Diagnosis of FH was assessed using the Dutch Lipid Clinic Network criteria, plasma levels of low-density lipoprotein (LDL) cholesterol, and genetic testing. To date, a total of 130 patients with FH have been registered, with 48 index cases and 82 relatives. Of the 130 patients, 8 were homozygous FH patients and 38 were children. Of FH individuals, 46.7% was confirmed by genetic testing: 61 patients (96.8%) carried the LDLR mutation (c.681C > G, c.1427C > G, c.1187-?_2140 ± ?del, c.2529_2530delinsA), and two patients (3.2%) carried the PCSK9 (protein convertase subtilisin/kexin type 9) mutation (c.42_43insTG). The c.2529_2530delinsA mutation detected in this study is novel and reported only in the Vietnamese population. However, only 53.8% of FH patients were followed up post diagnosis, and only 15.3% of these were approved for lipid-lowering therapy and specialized care. Notably, factors such as knowledge about FH in patients and/or guardians of FH children and support of primary care physicians affected patient participation with respect to treatment strategies and follow-up. Genetic identification, screening, and treatment of FH were feasible in Vietnam. The VINAFH Registry significantly contributed to the formation of the government agencies legislative acts that established the importance of FH as a socially and medically important disease requiring appropriate management strategies. Other low- and middle-income countries could, thus, use the VINAFH Registry model as a reference to establish programs for FH management according to the current status.
Collapse
Affiliation(s)
- Thanh-Huong Truong
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam.,Department of Cardiology, Hanoi Medical University, Hanoi, Vietnam
| | - Doan-Loi Do
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam.,Department of Cardiology, Hanoi Medical University, Hanoi, Vietnam
| | - Ngoc-Thanh Kim
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam.,Department of Cardiology, Hanoi Medical University, Hanoi, Vietnam
| | - Mai-Ngoc Thi Nguyen
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam.,Department of Cardiology, Hanoi Medical University, Hanoi, Vietnam
| | - Thanh-Tung Le
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
| | - Hong-An Le
- School of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| |
Collapse
|
329
|
Vujić N, Korbelius M, Sachdev V, Rainer S, Zimmer A, Huber A, Radović B, Kratky D. Intestine-specific DGAT1 deficiency improves atherosclerosis in apolipoprotein E knockout mice by reducing systemic cholesterol burden. Atherosclerosis 2020; 310:26-36. [PMID: 32882484 PMCID: PMC7116265 DOI: 10.1016/j.atherosclerosis.2020.07.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/25/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022]
Abstract
Background and aims Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) is the rate-limiting enzyme catalyzing the final step of triglyceride synthesis by esterifying a diglyceride with a fatty acid. We have previously shown that apolipoprotein E-knockout (ApoE−/−) mice lacking Dgat1 have reduced intestinal cholesterol absorption and potentiated macrophage cholesterol efflux, and consequently, exhibit attenuated atherogenesis. However, he-matopoietic Dgat1 deficiency lacked beneficial effects on atherosclerosis. Due to our recent results on the critical role of intestinal Dgat1 in murine cholesterol homeostasis, we delineated whether intestinal Dgat1 deficiency regulates atherogenesis in mice. Methods We generated intestine-specific Dgat1−/− mice on the ApoE−/− background (iDgat1−/−ApoE−/−) and determined cholesterol homeostasis and atherosclerosis development. Results When fed a Western-type diet, iDgat1−/−ApoE−/− mice exhibited a substantial decrease in fasting plasma cholesterol content in ApoB-containing lipoproteins. Although lipid absorption was delayed, iDgat1−/−ApoE−/− mice had reduced acute and fractional cholesterol absorption coupled with an elevated fecal caloric loss. In line, increased appearance of i.v. administered [3H]cholesterol in duodena and stool of iDgat1−/−ApoE−/− animals suggested potentiated cholesterol elimination. Atherosclerotic lesions were markedly smaller with beneficial alterations in plaque composition as evidenced by reduced macrophage infiltration and necrotic core size despite unaltered collagen content, indicating improved plaque stability. Conclusions Disruption of Dgat1 activity solely in the small intestine of ApoE−/− mice strongly decreased plasma cholesterol levels by abrogating the assimilation of dietary cholesterol, partly by reduced absorption and increased excretion. Consequently, the reduced cholesterol burden significantly attenuated atherogenesis and improved the lesion phenotype in iDgat1−/−ApoE−/− mice.
Collapse
Affiliation(s)
- Nemanja Vujić
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Melanie Korbelius
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Vinay Sachdev
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Silvia Rainer
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Andreas Zimmer
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Anton Huber
- Institute of Chemistry, University of Graz, Graz, Austria
| | - Branislav Radović
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
330
|
Aljenedil S, Alothman L, Bélanger AM, Brown L, Lahijanian Z, Bergeron J, Couture P, Baass A, Ruel I, Brisson D, Khoury E, Gaudet D, Genest J. Lomitapide for treatment of homozygous familial hypercholesterolemia: The Québec experience. Atherosclerosis 2020; 310:54-63. [PMID: 32906018 DOI: 10.1016/j.atherosclerosis.2020.07.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Homozygous familial hypercholesterolemia (HoFH) is an orphan disease, most often caused by bi-allelic mutations of the LDLR gene. Patients with HoFH have elevated LDL-C levels >13 mmol/L, tendinous xanthomata and severe, premature atherosclerotic cardiovascular disease (ASCVD). Untreated, most HoFH patients die of ASCVD in youth. New therapeutic modalities include lomitapide, an inhibitor of microsomal triglyceride transfer protein that lowers hepatic LDL-C production. We have recently identified 79 Canadian patients with HoFH. Here, we describe our experience with lomitapide in the province of Quebec, a geographic area known to have a high prevalence of HoFH. METHODS This is a retrospective case series of 12 HoFH patients followed at three lipidology centers in the province of Quebec. RESULTS Mean age of the patients was 44 ± 18 years; age at time of HoFH diagnosis ranged from 2 to 59 years. All patients were on a statin and ezetimibe 10 mg/day and five patients were treated with LDL apheresis. Treatment with lomitapide reduced LDL-C levels by 38% (intention-to-treat). Intolerable gastrointestinal side effects were observed in 3/12 patients and were the main reason for treatment discontinuation. Three patients tolerated lomitapide at doses ranging between 5 and 30 mg/day without major side effects. Downwards drug titration was necessary in the 6 remaining patients because of gastrointestinal side effects (n = 5) and elevated liver enzymes (n = 1), and 2 of them finally discontinued treatment. CONCLUSIONS Lomitapide may be used to further decrease LDL-C in HoFH patients; gastrointestinal side effects and hepatic toxicity may limit adherence.
Collapse
Affiliation(s)
- Sumayah Aljenedil
- Research Institute of the McGill University Health Centre, Montreal, Québec, Canada; Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital, Riyadh, Saudi Arabia
| | - Latifah Alothman
- Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Alexandre M Bélanger
- Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Leslie Brown
- Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Zubin Lahijanian
- Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Jean Bergeron
- Endocrinology and Nephrology Unit, CHU de Québec, Université Laval Research Center, Québec City, Québec, Canada
| | - Patrick Couture
- Endocrinology and Nephrology Unit, CHU de Québec, Université Laval Research Center, Québec City, Québec, Canada
| | - Alexis Baass
- Division of Experimental Medicine and Medical Biochemistry, Department of Medicine, McGill University, Québec, Canada; Nutrition, Metabolism, and Atherosclerosis Clinic, Institut de Recherches Cliniques de Montréal, Québec, Canada
| | - Isabelle Ruel
- Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Diane Brisson
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal, ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| | - Etienne Khoury
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal, ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| | - Daniel Gaudet
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal, ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Québec, Canada
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, Montreal, Québec, Canada.
| |
Collapse
|
331
|
Edelman A, Wetzel M, Owen C, Schadt CR, Callen JP. Xanthomas heralding pediatric coronary artery disease. JAAD Case Rep 2020; 6:753-754. [PMID: 32715071 PMCID: PMC7369508 DOI: 10.1016/j.jdcr.2020.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
332
|
Bélanger AM, Akioyamen L, Alothman L, Genest J. Evidence for improved survival with treatment of homozygous familial hypercholesterolemia. Curr Opin Lipidol 2020; 31:176-181. [PMID: 32520777 DOI: 10.1097/mol.0000000000000686] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW Homozygous familial hypercholesterolemia (HoFH) is an orphan disease caused by biallelic mutations at the LDL receptor (LDLR) gene, with a prevalence estimated at 1 : 250 000 to 1 : 630 000. HoFH is characterized by extremely elevated plasma levels of LDL-C greater than 10 mmol/l (>387 mg/dl), tendinous and cutaneous xanthomas in youth and premature atherosclerotic cardiovascular disease (ASCVD). The expected prevalence varies from country to country depending on the presence of founder effects, genetic probability and life expectancy. Untreated, HoFH is a fatal condition before age 30. Plasma levels of LDL-C are the major cause of mortality and the therapeutic target. Statin therapy led to a remarkable improvement in survival but is of limited use in loss-of-function LDLR gene variants or 'null' mutations. Inhibitors of PCSK9 are a useful adjunct in patients with LDLR mutations with residual activity. Extracorporeal LDL filtration has improved survival since its introduction three decades ago. RECENT FINDINGS Novel therapies, not dependent on a functioning LDLR include lomitapide and mipomersen, which decrease hepatic apolipoprotein B secretion, and evinacumab, directed at the angiopoietin like-3 protein (ANGPLT-3). SUMMARY Over the past 3-4 decades, the survival of patients with HoFH has increased markedly. New therapeutic options offer new hope.
Collapse
Affiliation(s)
- Alexandre M Bélanger
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, Quebec
| | - Leo Akioyamen
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Latifah Alothman
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, Quebec
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, Quebec
| |
Collapse
|
333
|
Wilson MH, Rajan S, Danoff A, White RJ, Hensley MR, Quinlivan VH, Recacha R, Thierer JH, Tan FJ, Busch-Nentwich EM, Ruddock L, Hussain MM, Farber SA. A point mutation decouples the lipid transfer activities of microsomal triglyceride transfer protein. PLoS Genet 2020; 16:e1008941. [PMID: 32760060 PMCID: PMC7444587 DOI: 10.1371/journal.pgen.1008941] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/18/2020] [Accepted: 06/17/2020] [Indexed: 01/08/2023] Open
Abstract
Apolipoprotein B-containing lipoproteins (B-lps) are essential for the transport of hydrophobic dietary and endogenous lipids through the circulation in vertebrates. Zebrafish embryos produce large numbers of B-lps in the yolk syncytial layer (YSL) to move lipids from yolk to growing tissues. Disruptions in B-lp production perturb yolk morphology, readily allowing for visual identification of mutants with altered B-lp metabolism. Here we report the discovery of a missense mutation in microsomal triglyceride transfer protein (Mtp), a protein that is essential for B-lp production. This mutation of a conserved glycine residue to valine (zebrafish G863V, human G865V) reduces B-lp production and results in yolk opacity due to aberrant accumulation of cytoplasmic lipid droplets in the YSL. However, this phenotype is milder than that of the previously reported L475P stalactite (stl) mutation. MTP transfers lipids, including triglycerides and phospholipids, to apolipoprotein B in the ER for B-lp assembly. In vitro lipid transfer assays reveal that while both MTP mutations eliminate triglyceride transfer activity, the G863V mutant protein unexpectedly retains ~80% of phospholipid transfer activity. This residual phospholipid transfer activity of the G863V mttp mutant protein is sufficient to support the secretion of small B-lps, which prevents intestinal fat malabsorption and growth defects observed in the mttpstl/stl mutant zebrafish. Modeling based on the recent crystal structure of the heterodimeric human MTP complex suggests the G865V mutation may block triglyceride entry into the lipid-binding cavity. Together, these data argue that selective inhibition of MTP triglyceride transfer activity may be a feasible therapeutic approach to treat dyslipidemia and provide structural insight for drug design. These data also highlight the power of yolk transport studies to identify proteins critical for B-lp biology.
Collapse
Affiliation(s)
- Meredith H. Wilson
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, United States of America
| | - Sujith Rajan
- New York University Long Island School of Medicine, Mineola, New York, United States of America
| | - Aidan Danoff
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, United States of America
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Richard J. White
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Monica R. Hensley
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, United States of America
| | - Vanessa H. Quinlivan
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, United States of America
| | - Rosario Recacha
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - James H. Thierer
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, United States of America
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Frederick J. Tan
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, United States of America
| | - Elisabeth M. Busch-Nentwich
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Lloyd Ruddock
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - M. Mahmood Hussain
- New York University Long Island School of Medicine, Mineola, New York, United States of America
| | - Steven A. Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland, United States of America
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
334
|
Reeskamp LF, Tromp TR, Defesche JC, Grefhorst A, Stroes ESG, Hovingh GK, Zuurbier L. Next-generation sequencing to confirm clinical familial hypercholesterolemia. Eur J Prev Cardiol 2020; 28:875-883. [PMID: 34298557 DOI: 10.1093/eurjpc/zwaa451] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/28/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Familial hypercholesterolemia is characterised by high low-density lipoprotein-cholesterol levels and is caused by a pathogenic variant in LDLR, APOB or PCSK9. We investigated which proportion of suspected familial hypercholesterolemia patients was genetically confirmed, and whether this has changed over the past 20 years in The Netherlands. METHODS Targeted next-generation sequencing of 27 genes involved in lipid metabolism was performed in patients with low-density lipoprotein-cholesterol levels greater than 5 mmol/L who were referred to our centre between May 2016 and July 2018. The proportion of patients carrying likely pathogenic or pathogenic variants in LDLR, APOB or PCSK9, or the minor familial hypercholesterolemia genes LDLRAP1, ABCG5, ABCG8, LIPA and APOE were investigated. This was compared with the yield of Sanger sequencing between 1999 and 2016. RESULTS A total of 227 out of the 1528 referred patients (14.9%) were heterozygous carriers of a pathogenic variant in LDLR (80.2%), APOB (14.5%) or PCSK9 (5.3%). More than 50% of patients with a Dutch Lipid Clinic Network score of 'probable' or 'definite' familial hypercholesterolemia were familial hypercholesterolemia mutation-positive; 4.8% of the familial hypercholesterolemia mutation-negative patients carried a variant in one of the minor familial hypercholesterolemia genes. The mutation detection rate has decreased over the past two decades, especially in younger patients in which it dropped from 45% in 1999 to 30% in 2018. CONCLUSIONS A rare pathogenic variant in LDLR, APOB or PCSK9 was identified in 14.9% of suspected familial hypercholesterolemia patients and this rate has decreased in the past two decades. Stringent use of clinical criteria algorithms is warranted to increase this yield. Variants in the minor familial hypercholesterolemia genes provide a possible explanation for the familial hypercholesterolemia phenotype in a minority of patients.
Collapse
Affiliation(s)
- Laurens F Reeskamp
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - Tycho R Tromp
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - Joep C Defesche
- Department of Clinical Genetics, University of Amsterdam, The Netherlands
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, University of Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - Linda Zuurbier
- Department of Clinical Genetics, University of Amsterdam, The Netherlands
| |
Collapse
|
335
|
Reeskamp LF, Tromp TR, Defesche JC, Grefhorst A, Stroes ES, Hovingh GK, Zuurbier L. Next-generation sequencing to confirm clinical familial hypercholesterolemia. Eur J Prev Cardiol 2020:2047487320942996. [PMID: 32718233 DOI: 10.1177/2047487320942996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
BACKGROUND Familial hypercholesterolemia is characterised by high low-density lipoprotein-cholesterol levels and is caused by a pathogenic variant in LDLR, APOB or PCSK9. We investigated which proportion of suspected familial hypercholesterolemia patients was genetically confirmed, and whether this has changed over the past 20 years in The Netherlands. METHODS Targeted next-generation sequencing of 27 genes involved in lipid metabolism was performed in patients with low-density lipoprotein-cholesterol levels greater than 5 mmol/L who were referred to our centre between May 2016 and July 2018. The proportion of patients carrying likely pathogenic or pathogenic variants in LDLR, APOB or PCSK9, or the minor familial hypercholesterolemia genes LDLRAP1, ABCG5, ABCG8, LIPA and APOE were investigated. This was compared with the yield of Sanger sequencing between 1999 and 2016. RESULTS A total of 227 out of the 1528 referred patients (14.9%) were heterozygous carriers of a pathogenic variant in LDLR (80.2%), APOB (14.5%) or PCSK9 (5.3%). More than 50% of patients with a Dutch Lipid Clinic Network score of 'probable' or 'definite' familial hypercholesterolemia were familial hypercholesterolemia mutation-positive; 4.8% of the familial hypercholesterolemia mutation-negative patients carried a variant in one of the minor familial hypercholesterolemia genes. The mutation detection rate has decreased over the past two decades, especially in younger patients in which it dropped from 45% in 1999 to 30% in 2018. CONCLUSIONS A rare pathogenic variant in LDLR, APOB or PCSK9 was identified in 14.9% of suspected familial hypercholesterolemia patients and this rate has decreased in the past two decades. Stringent use of clinical criteria algorithms is warranted to increase this yield. Variants in the minor familial hypercholesterolemia genes provide a possible explanation for the familial hypercholesterolemia phenotype in a minority of patients.
Collapse
Affiliation(s)
- Laurens F Reeskamp
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - Tycho R Tromp
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - Joep C Defesche
- Department of Clinical Genetics, University of Amsterdam, The Netherlands
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, University of Amsterdam, The Netherlands
| | - Erik Sg Stroes
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, University of Amsterdam, The Netherlands
| | - Linda Zuurbier
- Department of Clinical Genetics, University of Amsterdam, The Netherlands
| |
Collapse
|
336
|
Raal FJ, Chilton R, Ranjith N, Rambiritch V, Leisegang RF, Ebrahim IO, Tonder AV, Shunmoogam N, Bouharati C, Musa MG, Karamchand S, Naidoo P, Blom DJ. PCSK9 Inhibitors: From Nature’s Lessons to Clinical Utility. Endocr Metab Immune Disord Drug Targets 2020; 20:840-854. [DOI: 10.2174/1871530320666200213114138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/31/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
Abstract
Background:
Proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors are a novel
class of non-statin lipid lowering therapy that reduce LDL-cholesterol by 50 - 60%. PCSK9 inhibitors
decrease LDL-cholesterol by preventing intracellular degradation of LDL receptors; subsequently, a
greater number of LDL-receptors are available on the cell surface to extract circulating LDL.
Objective:
To describe the origins of PCSK9 inhibitors and their current use in clinical practice.
Methods:
We performed a narrative review of the PCSK9 inhibitor class of drugs.
Results:
Current data indicate that PCSK9 inhibitors effectively reduce LDL-cholesterol and are well
tolerated and safe. PCSK9 inhibitors have also been shown to reduce cardiovascular event rates in
patients with stable atherosclerotic cardiovascular disease and in patients with a recent (up to one year)
acute coronary syndrome. Given the costs, chronicity of the treatment and the potential budget impact,
PCSK9 inhibitors are often limited to patients with the highest absolute risk for major adverse cardiovascular
events despite optimal treatment with high-intensity statin and ezetimibe.
Conclusion:
PCSK9 inhibitors have a favorable safety, efficacy and tolerability profile. Postmarketing
safety surveillance and real-world studies are needed to further support the long-term safety
profile of this class of medicine.
Collapse
Affiliation(s)
- Frederick J. Raal
- Department of Medicine, Division of Endocrinology and Metabolism, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Robert Chilton
- Department of Medicine, Faculty of Medicine, Division of Cardiology and Interventional Cardiology, University of Texas Health Science Center at San Antonia, Texas, United States
| | - Naresh Ranjith
- Department of Cardiology, Cardiovascular Research Centre, Durban, South Africa
| | - Virendra Rambiritch
- Department of Pharmacology, Faculty of Health Sciences, Discipline of Pharmaceutical Sciences, University of Kwa-Zulu Natal, Durban, South Africa
| | - Rory F. Leisegang
- Department of Pharmaceutical Biosciences, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| | | | - Alet van Tonder
- Department of Diabetes and Cardiovascular Medicine, Medical Affairs, Sanofi, Midrand, South Africa
| | - Nelusha Shunmoogam
- Department of Diabetes and Cardiovascular Medicine, Medical Affairs, Sanofi, Midrand, South Africa
| | - Célia Bouharati
- Department of Clinical Trials, Clinical Study Unit, Sanofi, Midrand, South Africa
| | - Moji G. Musa
- Department of Diabetes and Cardiovascular Medicine, Medical Affairs, Sanofi, Midrand, South Africa
| | - Sumanth Karamchand
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Poobalan Naidoo
- Department of Diabetes and Cardiovascular Medicine, Medical Affairs, Sanofi, Midrand, South Africa
| | - Dirk J. Blom
- Department of Medicine, Division of Lipidology and Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
337
|
Greco MF, Sirtori CR, Corsini A, Ezhov M, Sampietro T, Ruscica M. Lipoprotein(a) Lowering-From Lipoprotein Apheresis to Antisense Oligonucleotide Approach. J Clin Med 2020; 9:jcm9072103. [PMID: 32635396 PMCID: PMC7408876 DOI: 10.3390/jcm9072103] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
It is well-known that elevated lipoprotein(a)—Lp(a)—levels are associated with a higher risk of cardiovascular (CV) mortality and all-cause mortality, although a standard pharmacotherapeutic approach is still undefined for patients with high CV risk dependent on hyperlipoproteinemia(a). Combined with high Lp(a) levels, familial hypercholesterolemia (FH) leads to a greater CVD risk. In suspected FH patients, the proportion of cases explained by a rise of Lp(a) levels ranges between 5% and 20%. In the absence of a specific pharmacological approach able to lower Lp(a) to the extent required to achieve CV benefits, the most effective strategy today is lipoprotein apheresis (LA). Although limited, a clear effect on Lp(a) is exerted by PCSK9 antagonists, with apparently different mechanisms when given with statins (raised catabolism) or as monotherapy (reduced production). In the era of RNA-based therapies, a new dawn is represented by the use of antisense oligonucleotides APO(a)Lrx, able to reduce Lp(a) from 35% to over 80%, with generally modest injection site reactions. The improved knowledge of Lp(a) atherogenicity and possible prevention will be of benefit for patients with residual CV risk remaining after the most effective available lipid-lowering agents.
Collapse
Affiliation(s)
- Maria Francesca Greco
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
| | - Cesare R. Sirtori
- Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Alberto Corsini
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- IRCCS Multimedica, 20099 Milan, Italy
| | - Marat Ezhov
- National Medical Research Center of Cardiology of the Ministry of Health, Moscow, Russia;
| | - Tiziana Sampietro
- U.O. Lipoapheresis and Center for Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, 56126 Pisa, Italy;
| | - Massimiliano Ruscica
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- Correspondence: ; Tel.: +39-0250318220
| |
Collapse
|
338
|
PCSK9 Inhibitors for Homozygous Familial Hypercholesterolemia. J Am Coll Cardiol 2020; 76:143-145. [DOI: 10.1016/j.jacc.2020.05.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 11/22/2022]
|
339
|
Giammanco A, Cefalù AB, Noto D, Averna MR. Therapeutic Options for Homozygous Familial Hypercholesterolemia: The Role of Lomitapide. Curr Med Chem 2020; 27:3773-3783. [DOI: 10.2174/0929867326666190121120735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/25/2018] [Accepted: 12/28/2018] [Indexed: 11/22/2022]
Abstract
Background:Lomitapide (Juxtapid® in US and Lojuxta® in Europe) is the first developed inhibitor of the Microsomal Triglyceride Transfer Protein (MTP) approved as a novel drug for the management of Homozygous Familial Hypercholesterolemia (HoFH). It acts by binding directly and selectively to MTP thus decreasing the assembly and secretion of the apo-B containing lipoproteins both in the liver and in the intestine.Aims:The present review aims at summarizing the recent knowledge on lomitapide in the management of HoFH.Results:The efficacy and safety of lomitapide have been evaluated in several trials and it has been shown a reduction of the plasma levels of Low-Density Lipoprotein Cholesterol (LDL-C) by an average of more than 50%. Although the most common side effects are gastrointestinal and liver events, lomitapide presents generally with a good tolerability and satisfactory patients compliance. Recently, in Europe, to evaluate the long-term safety and efficacy of lomitapide, the LOWER registry (ClinicalTrials.gov Identifier: NCT02135705) has been established in order to acquire informations on HoFH lomitapidetreated patients from “real life” clinical practice.:Furthermore, the observation that lomitapide decreases triglyceride levels may be considered for patients affected by severe forms of hypertriglyceridemia who undergo recurrent episodes of pancreatitis and are poor responders to conventional treatment.Conclusion:Lomitapide represents an innovative and efficacious drug for the treatment of HoFH. Longterm safety data, treatment of pediatric and pregnant HoFH patients and management of severe hypertriglyceridemia still require further investigations.
Collapse
Affiliation(s)
- Antonina Giammanco
- Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.Mi.S), Policlinico “Paolo Giaccone”, Università di Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Angelo B. Cefalù
- Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.Mi.S), Policlinico “Paolo Giaccone”, Università di Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Davide Noto
- Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.Mi.S), Policlinico “Paolo Giaccone”, Università di Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Maurizio R. Averna
- Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.Mi.S), Policlinico “Paolo Giaccone”, Università di Palermo, Via del Vespro 129, 90127 Palermo, Italy
| |
Collapse
|
340
|
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: 114] [Impact Index Per Article: 22.8] [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]
|
341
|
A systematic review of LDLR, PCSK9, and APOB variants in Asia. Atherosclerosis 2020; 305:50-57. [DOI: 10.1016/j.atherosclerosis.2020.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/18/2020] [Accepted: 05/07/2020] [Indexed: 01/08/2023]
|
342
|
Stefanutti C. Lomitapide-a Microsomal Triglyceride Transfer Protein Inhibitor for Homozygous Familial Hypercholesterolemia. Curr Atheroscler Rep 2020; 22:38. [PMID: 32557261 PMCID: PMC7303073 DOI: 10.1007/s11883-020-00858-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Homozygous familial hypercholesterolemia (HoFH) is a rare, genetic condition characterized by high levels of Low density lipoprotein cholesterol (LDL-C); overt, early-onset atherosclerotic cardiovascular disease (ASCVD); and premature cardiovascular events and mortality. Lomitapide is a first-in-class microsomal triglyceride transfer protein inhibitor for the treatment of HoFH. This review provides an update on data emerging from real-world studies of lomitapide following on from its pivotal phase 3 clinical trial in HoFH. RECENT FINDINGS Recent registry data have confirmed that HoFH is characterized by delayed diagnosis, with many patients not receiving effective therapy until they are approaching the age when major adverse cardiovascular events may occur. Data from case series of varying sizes, and from a 163-patient registry of HoFH patients receiving lomitapide, have demonstrated that lomitapide doses are lower and adverse events less severe than in the phase 3 study. Lomitapide enables many patients to reach European Atherosclerosis Society LDL-C targets. Some patients are able to reduce frequency of lipoprotein apheresis or, in some cases, stop the procedure altogether-unless there is significant elevation of lipoprotein (a). Modelling analyses based on historical and clinical trial data indicate that lomitapide has the potential to improve cardiovascular outcomes and survival in HoFH. Real-world clinical experience with lomitapide has shown the drug to be effective with manageable, less marked adverse events than in formal clinical studies. Event modelling data suggest a survival benefit with lomitapide in HoFH.
Collapse
Affiliation(s)
- Claudia Stefanutti
- Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Regional Centre (Lazio) for Rare Diseases, Immunohematology and Transfusion Medicine, Department of Molecular Medicine, "Sapienza" University of Rome, "Umberto I" Hospital, Rome, Italy.
| |
Collapse
|
343
|
Jia X, Al Rifai M, Liu J, Agarwala A, Gulati M, Virani SS. Highlights of Studies in Cardiovascular Disease Prevention Presented at the 2020 American College of Cardiology Annual Scientific Session. Curr Atheroscler Rep 2020; 22:32. [PMID: 32556778 PMCID: PMC7301047 DOI: 10.1007/s11883-020-00856-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW The review highlights selected studies related to cardiovascular disease (CVD) prevention that were presented at the American College of Cardiology 2020 Virtual Scientific Session (ACC.20)/World Cardiology Congress (WCC). RECENT FINDINGS The studies reviewed include clinical trials on the efficacy and safety of alirocumab (Study in Participants with Homozygous Familial Hypercholesterolemia [ODYSSEY HoFH]) and evinacumab in the treatment of homozygous familial hypercholesterolemia (HoFH); Evaluating the Efficacy of E-cigarettes for Smoking Cessation (E3); the use of renal denervation in the treatment of hypertension (SPYRAL HTN-OFF MED PIVOTAL); and the assessment of vericiguat in the treatment of heart failure (A Study of Vericiguat in Participants with Heart Failure with Reduce Ejection Fraction [VICTORIA]). In addition, results from the pooled analysis of phase III trials on inclisiran and secondary analysis examining eicosapentaenoic acid levels and cardiovascular outcomes from the Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT) were included. Finally, we discuss studies examining the use of polygenic risk score with low density lipoprotein cholesterol (LDL-C) and systolic blood pressure (SBP) on lifetime cardiovascular risk. The studies presented at the ACC.20/WCC represent notable contributions in the field of CVD prevention.
Collapse
Affiliation(s)
- Xiaoming Jia
- Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Mahmoud Al Rifai
- Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Jing Liu
- Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Anandita Agarwala
- Cardiovascular Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Martha Gulati
- Division of Cardiology, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Salim S Virani
- Section of Cardiology, Baylor College of Medicine, Houston, TX, USA.
- Section of Cardiology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
- Health Policy, Quality & Informatics Program, Health Services Research and Development Center for Innovations, Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
344
|
Hu P, Dharmayat KI, Stevens CA, Sharabiani MT, Jones RS, Watts GF, Genest J, Ray KK, Vallejo-Vaz AJ. Prevalence of Familial Hypercholesterolemia Among the General Population and Patients With Atherosclerotic Cardiovascular Disease. Circulation 2020; 141:1742-1759. [DOI: 10.1161/circulationaha.119.044795] [Citation(s) in RCA: 353] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background:
Contemporary studies suggest that familial hypercholesterolemia (FH) is more frequent than previously reported and increasingly recognized as affecting individuals of all ethnicities and across many regions of the world. Precise estimation of its global prevalence and prevalence across World Health Organization regions is needed to inform policies aiming at early detection and atherosclerotic cardiovascular disease (ASCVD) prevention. The present study aims to provide a comprehensive assessment and more reliable estimation of the prevalence of FH than hitherto possible in the general population (GP) and among patients with ASCVD.
Methods:
We performed a systematic review and meta-analysis including studies reporting on the prevalence of heterozygous FH in the GP or among those with ASCVD. Studies reporting gene founder effects and focused on homozygous FH were excluded. The search was conducted through Medline, Embase, Cochrane, and Global Health, without time or language restrictions. A random-effects model was applied to estimate the overall pooled prevalence of FH in the general and ASCVD populations separately and by World Health Organization regions.
Results:
From 3225 articles, 42 studies from the GP and 20 from populations with ASCVD were eligible, reporting on 7 297 363 individuals/24 636 cases of FH and 48 158 patients/2827 cases of FH, respectively. More than 60% of the studies were from Europe. Use of the Dutch Lipid Clinic Network criteria was the commonest diagnostic method. Within the GP, the overall pooled prevalence of FH was 1:311 (95% CI, 1:250–1:397; similar between children [1:364] and adults [1:303],
P
=0.60; across World Health Organization regions where data were available,
P
=0.29; and between population-based and electronic health records–based studies,
P
=0.82). Studies with ≤10 000 participants reported a higher prevalence (1:200–289) compared with larger cohorts (1:365–407;
P
<0.001). The pooled prevalence among those with ASCVD was 18-fold higher than in the GP (1:17 [95% CI, 1:12–1:24]), driven mainly by coronary artery disease (1:16; [95% CI, 1:12–1:23]). Between-study heterogeneity was large (
I
2
>95%). Tests assessing bias were nonsignificant (
P
>0.3).
Conclusions:
With an overall prevalence of 1:311, FH is among the commonest genetic disorders in the GP, similarly present across different regions of the world, and is more frequent among those with ASCVD. The present results support the advocacy for the institution of public health policies, including screening programs, to identify FH early and to prevent its global burden.
Collapse
Affiliation(s)
- Pengwei Hu
- Imperial Center for Cardiovascular Disease Prevention (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V.), Imperial College London, UK
- Department of Primary Care and Public Health (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V., M.T.A.S.), Imperial College London, UK
- Department of Health Service, Logistics University of People’s Armed Police Force, Tianjin, China (P.H.)
| | - Kanika I. Dharmayat
- Imperial Center for Cardiovascular Disease Prevention (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V.), Imperial College London, UK
- Department of Primary Care and Public Health (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V., M.T.A.S.), Imperial College London, UK
| | - Christophe A.T. Stevens
- Imperial Center for Cardiovascular Disease Prevention (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V.), Imperial College London, UK
- Department of Primary Care and Public Health (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V., M.T.A.S.), Imperial College London, UK
| | - Mansour T.A. Sharabiani
- Department of Primary Care and Public Health (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V., M.T.A.S.), Imperial College London, UK
| | - Rebecca S. Jones
- School of Public Health, and Charing Cross Campus Library (R.S.J.), Imperial College London, UK
| | - Gerald F. Watts
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth (G.F.W.)
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Australia (G.F.W.)
| | - Jacques Genest
- McGill University Health Center, Montreal, QC, Canada (J.G.)
| | - Kausik K. Ray
- Imperial Center for Cardiovascular Disease Prevention (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V.), Imperial College London, UK
- Department of Primary Care and Public Health (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V., M.T.A.S.), Imperial College London, UK
| | - Antonio J. Vallejo-Vaz
- Imperial Center for Cardiovascular Disease Prevention (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V.), Imperial College London, UK
- Department of Primary Care and Public Health (P.H., K.I.D., C.A.T.S., K.K.R., A.J.V.-V., M.T.A.S.), Imperial College London, UK
| |
Collapse
|
345
|
Hovingh GK, Lepor NE, Kallend D, Stoekenbroek RM, Wijngaard PL, Raal FJ. Inclisiran Durably Lowers Low-Density Lipoprotein Cholesterol and Proprotein Convertase Subtilisin/Kexin Type 9 Expression in Homozygous Familial Hypercholesterolemia. Circulation 2020; 141:1829-1831. [DOI: 10.1161/circulationaha.119.044431] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- G. Kees Hovingh
- Department of Internal Medicine, Amsterdam UMC, The Netherlands (G.K.H.)
| | - Norman E. Lepor
- Geffen School of Medicine, University of California, Los Angeles (N.E.L.)
| | - David Kallend
- Cardiovascular, The Medicines Company, Parsippany, NJ (D.K., R.M.S., P.L.J.W.)
| | | | | | - Frederick J. Raal
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (F.J.R.)
| |
Collapse
|
346
|
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
|
347
|
Brandts J, Dharmayat KI, Ray KK, Vallejo-Vaz AJ. Familial hypercholesterolemia: is it time to separate monogenic from polygenic familial hypercholesterolemia? Curr Opin Lipidol 2020; 31:111-118. [PMID: 32332432 DOI: 10.1097/mol.0000000000000675] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW This review explores the concepts of monogenic and the so-called polygenic familial hypercholesterolemia and how the identification of familial hypercholesterolemia as a monogenic condition and its separation from polygenic primary hypercholesterolemia may have implications for clinical practice. RECENT FINDINGS Through genetic testing, a mutation in any of the three known autosomal dominant familial hypercholesterolemia-causing genes is found in 60-80% of cases with a clinical diagnosis of definite familial hypercholesterolemia. As individuals with a polygenic basis for their hypercholesterolemia do not follow the same inheritance pattern observed in monogenic familial hypercholesterolemia, the use of family-based cascade screening in individuals with a polygenic origin is not recommend, as only 30% of relatives have an elevated LDL-C compared to the 50% in monogenic families. The presence of a causative monogenic mutation associates the highest cardiovascular risk vs. not having a mutation or having a polygenic background, providing prognostic information independent of LDL-C. It may also help assess intensity of interventions. Treatment adherence also seems to be higher after monogenic confirmation of hypercholesterolemia. SUMMARY Knowledge about the genetic status of an individual with clinical familial hypercholesterolemia (monogenic vs. polygenic) can provide a more informed understanding to evaluating risk, managing disease and opportunities for screening strategies.
Collapse
Affiliation(s)
- Julia Brandts
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
- Department of Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | - Kanika I Dharmayat
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Antonio J Vallejo-Vaz
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
348
|
Lipid lowering therapy in cardiovascular disease: From myth to molecular reality. Pharmacol Ther 2020; 213:107592. [PMID: 32492513 DOI: 10.1016/j.pharmthera.2020.107592] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022]
Abstract
Lipid-lowering therapy is one major cornerstone of medical treatment of cardiovascular disease in order to modulate atherosclerosis. Statins, ezetimibe and novel PCSK9-inhibitors are already recommended in current guidelines and were shown to improve lipid profiles and have positive effects on the rate of ischemic events and cardiovascular mortality. Recent studies suggest that the concept of "The lower the better" might be valid at least regarding low density lipoproteins. In addition, lowering lipoprotein (a) still displays a major challenge in lipid therapy. Furthermore, also lowering triglycerides seems to improve cardiovascular outcome. Regarding triglycerides, icosapent ethyl, a polyunsaturated fatty acid recently attracted attention showing cardiovascular risk reduction due to triglyceride lowering. Therefore, new therapeutic strategies and drug classes are eagerly awaited. Targeting LDL, bempedoic acid and the siRNA inclisiran provide promising results. Moreover, regarding TG a monoclonal antibody called evinacumab and an antisense-oligonucleotide against ANGPTL3 showed effective TG-lowering. At least, using antisense-oligonucleotides against ApoC-III and Lp(a) resulted in promising outcomes. In this review, current and future options for lipid management are presented depending on different drug classes.
Collapse
|
349
|
Yu H, Rimbert A, Palmer AE, Toyohara T, Xia Y, Xia F, Ferreira LMR, Chen Z, Chen T, Loaiza N, Horwitz NB, Kacergis MC, Zhao L, Soukas AA, Kuivenhoven JA, Kathiresan S, Cowan CA. GPR146 Deficiency Protects against Hypercholesterolemia and Atherosclerosis. Cell 2020; 179:1276-1288.e14. [PMID: 31778654 DOI: 10.1016/j.cell.2019.10.034] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 09/12/2019] [Accepted: 10/25/2019] [Indexed: 02/07/2023]
Abstract
Although human genetic studies have implicated many susceptible genes associated with plasma lipid levels, their physiological and molecular functions are not fully characterized. Here we demonstrate that orphan G protein-coupled receptor 146 (GPR146) promotes activity of hepatic sterol regulatory element binding protein 2 (SREBP2) through activation of the extracellular signal-regulated kinase (ERK) signaling pathway, thereby regulating hepatic very low-density lipoprotein (VLDL) secretion, and subsequently circulating low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG) levels. Remarkably, GPR146 deficiency reduces plasma cholesterol levels substantially in both wild-type and LDL receptor (LDLR)-deficient mice. Finally, aortic atherosclerotic lesions are reduced by 90% and 70%, respectively, in male and female LDLR-deficient mice upon GPR146 depletion. Taken together, these findings outline a regulatory role for the GPR146/ERK axis in systemic cholesterol metabolism and suggest that GPR146 inhibition could be an effective strategy to reduce plasma cholesterol levels and atherosclerosis.
Collapse
Affiliation(s)
- Haojie Yu
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| | - Antoine Rimbert
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Center, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands; Institute of Thorax, INSERM, CNRS, UNIV Nantes, Nantes, 44007, France
| | - Alice E Palmer
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA
| | - Takafumi Toyohara
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Yulei Xia
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Fang Xia
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Leonardo M R Ferreira
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Zhifen Chen
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Tao Chen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Natalia Loaiza
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Center, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | | | - Michael C Kacergis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Liping Zhao
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Alexander A Soukas
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Center, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Sekar Kathiresan
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Disease Initiative of the Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Chad A Cowan
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
350
|
Mszar R, Buscher S, Taylor HL, Rice-DeFosse MT, McCann D. Familial Hypercholesterolemia and the Founder Effect Among Franco-Americans: A Brief History and Call to Action. CJC Open 2020; 2:161-167. [PMID: 32462130 PMCID: PMC7242505 DOI: 10.1016/j.cjco.2020.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 01/19/2020] [Indexed: 01/01/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an inherited disorder characterized by chronically elevated low-density lipoprotein cholesterol levels and an increased risk of premature atherosclerotic cardiovascular disease. FH has been shown to disproportionately affect French Canadians and other ethnic populations due to the presence of a founder effect characterized by reduced genetic diversity resulting from relatively few individuals with FH-causing genetic mutations establishing self-contained populations. Beginning in the mid-1800s, approximately 1 million French Canadians immigrated to the Northeastern United States and largely remained in these small, tight-knit communities. Despite extensive genetic- and population-based research involving the French-Canadian founder population, primarily in the Province of Quebec, little is known regarding Franco-Americans in the United States. Concurrent with addressing the underdiagnosis rate of FH in the general population, we propose the following steps to leverage this founder effect and meet the cardiovascular needs of Franco-Americans: (1) increase cascade screening in regions of the United States with a high proportion of individuals of French-Canadian descent; (2) promote registry-based, epidemiological research to elucidate accurate prevalence estimates as well as diagnostic and treatment gaps in Franco-Americans; and (3) validate contemporary risk stratification strategies such as the Montreal-FH-SCORE to enable optimal lipid management and prevention of premature atherosclerotic cardiovascular disease among French-Canadian descendants.
Collapse
Affiliation(s)
- Reed Mszar
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Sara Buscher
- Division of General Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
| | - Heidi L. Taylor
- Department of Sociology, Bates College, Lewiston, Maine, USA
| | - Mary T. Rice-DeFosse
- Department of French and Francophone Studies, Bates College, Lewiston, Maine, USA
| | - Dervilla McCann
- Department of Cardiology, Central Maine Medical Center, Lewiston, Maine, USA
| |
Collapse
|