601
|
Janus ED. Homozygous familial hypercholesterolaemia - Early recognition and early treatment improve outcomes. Atherosclerosis 2017; 260:147-149. [PMID: 28341574 DOI: 10.1016/j.atherosclerosis.2017.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Edward D Janus
- General Internal Medicine Unit, Western Health and Department of Medicine Melbourne Medical School - Western Precinct, University of Melbourne, Sunshine Hospital, St Albans, VIC 3021, Australia.
| |
Collapse
|
602
|
Harada-Shiba M, Ikewaki K, Nohara A, Otsubo Y, Yanagi K, Yoshida M, Chang Q, Foulds P. Efficacy and Safety of Lomitapide in Japanese Patients with Homozygous Familial Hypercholesterolemia. J Atheroscler Thromb 2017; 24:402-411. [PMID: 28154305 PMCID: PMC5392478 DOI: 10.5551/jat.38216] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aim: There is an unmet need in Japan for more optimal lipid-lowering therapy (LLT) for patients with homozygous familial hypercholesterolemia (HoFH) who respond inadequately to available drug therapies and/or apheresis, to achieve goals of low-density lipoprotein cholesterol (LDL-C) reduction by 50% or to < 100 mg/dL. Methods: In this study, Japanese patients with HoFH on stable LLT and diet were treated with lomitapide, initiated at 5 mg/day and escalated to maximum tolerated dose (up to 60 mg/day) over 14 weeks. The primary efficacy endpoint was mean percentage change from baseline to Week 26 in LDL-C. Secondary endpoints included changes in other lipid parameters and safety throughout the 56-week study (including follow-up). Results: Nine patients entered the efficacy phase of the study and, of these, eight completed 56 weeks. Mean LDL-C was reduced by 42% (p < 0.0001) at 26 weeks, from 199 mg/dL (95% CI: 149–250) at baseline to 118 mg/dL (95% CI: 70–166). A 50% reduction in LDL-C and LDL-C < 100 mg/dL was achieved by five and six of nine patients, respectively, at 26 weeks. After 56 weeks, LDL-C was reduced by 38% (p = 0.0032) from baseline. Significant reductions in non-HDL-C, VLDL-C, triglycerides, and apolipoprotein B were also reported at Week 26. There were no new safety signals and, similar to previous studies, gastrointestinal adverse events were the most common adverse events. Conclusion: Lomitapide, added to ongoing treatment with other LLTs, was effective in rapidly and significantly reducing the levels of LDL-C and other atherogenic apolipoprotein B-containing lipoproteins in adult Japanese patients with HoFH.
Collapse
|
603
|
Bruckert E, Kalmykova O, Bittar R, Carreau V, Béliard S, Saheb S, Rosenbaum D, Bonnefont-Rousselot D, Thomas D, Emery C, Khoshnood B, Carrié A. Long-term outcome in 53 patients with homozygous familial hypercholesterolaemia in a single centre in France. Atherosclerosis 2017; 257:130-137. [DOI: 10.1016/j.atherosclerosis.2017.01.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/03/2017] [Accepted: 01/13/2017] [Indexed: 01/09/2023]
|
604
|
Banach M, Jankowski P, Jóźwiak J, Cybulska B, Windak A, Guzik T, Mamcarz A, Broncel M, Tomasik T, Rysz J, Jankowska-Zduńczyk A, Hoffman P, Mastalerz-Migas A. PoLA/CFPiP/PCS Guidelines for the Management of Dyslipidaemias for Family Physicians 2016. Arch Med Sci 2017; 13:1-45. [PMID: 28144253 PMCID: PMC5206369 DOI: 10.5114/aoms.2017.64712] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/05/2016] [Indexed: 02/06/2023] Open
Affiliation(s)
- Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| | - Piotr Jankowski
- 1 Department of Cardiology, Interventional Electrocardiology and Hypertension, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Jóźwiak
- Institute of Health and Nutrition Sciences, Czestochowa University of Technology, Czestochowa, Poland
| | | | - Adam Windak
- Department of Family Medicine, Chair of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz Guzik
- Department of Internal Diseases and Rural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Artur Mamcarz
- 3 Department of Internal Diseases and Cardiology, 2 Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Lodz, Poland
| | - Tomasz Tomasik
- Department of Family Medicine, Chair of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| | | | - Piotr Hoffman
- Department of Congenital Cardiac Defects, Institute of Cardiology, Warsaw, Poland
| | | |
Collapse
|
605
|
Low-density lipoprotein receptor-negative compound heterozygous familial hypercholesterolemia: Two lifetime journeys of lipid-lowering therapy. J Clin Lipidol 2017; 11:301-305. [PMID: 28391901 DOI: 10.1016/j.jacl.2017.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/30/2016] [Accepted: 01/02/2017] [Indexed: 01/08/2023]
Abstract
We present the case history of 2 patients with low-density lipoprotein receptor-negative compound heterozygous familial hypercholesterolemia who did not receive lipoprotein apheresis. We describe the subsequent effect of all lipid-lowering medications during their life course including resins, statins, ezetimibe, nicotinic acid/laropiprant, mipomersen, and lomitapide. These cases tell the story of siblings affected with this rare disease, who are free of symptoms but still are at a very high cardiovascular disease risk, and their treatment from childhood.
Collapse
|
606
|
Toplak H, Ludvik B, Lechleitner M, Dieplinger H, Föger B, Paulweber B, Weber T, Watschinger B, Horn S, Wascher TC, Drexel H, Brodmann M, Pilger E, Rosenkranz A, Pohanka E, Oberbauer R, Traindl O, Roithinger FX, Metzler B, Haring HP, Kiechl S. Austrian Lipid Consensus on the management of metabolic lipid disorders to prevent vascular complications: A joint position statement issued by eight medical societies. 2016 update. Wien Klin Wochenschr 2017; 128 Suppl 2:S216-28. [PMID: 27052248 PMCID: PMC4839054 DOI: 10.1007/s00508-016-0993-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In 2010, eight Austrian medical societies proposed a joint position statement on the management of metabolic lipid disorders for the prevention of vascular complications. An updated and extended version of these recommendations according to the current literature is presented, referring to the primary and secondary prevention of vascular complications in adults, taking into consideration the guidelines of other societies. The "Austrian Lipid Consensus - 2016 update" provides guidance for individualized risk stratification and respective therapeutic targets, and discusses the evidence for reducing vascular endpoints with available lipid-lowering therapies. Furthermore, specific management in key patient groups is outlined, including subjects presenting with coronary, cerebrovascular, and/or peripheral atherosclerosis; diabetes mellitus and/or metabolic syndrome; nephropathy; and familial hypercholesterolemia.
Collapse
Affiliation(s)
- Hermann Toplak
- Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| | - Bernhard Ludvik
- First Medical Department, Rudolfstiftung Hospital, Vienna, Austria
| | | | - Hans Dieplinger
- Department of Medical Genetics, Clinical and Molecular Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Bernhard Föger
- Department of Internal Medicine, Bregenz Hospital, Bregenz, Austria
| | - Bernhard Paulweber
- First Medical Department, Paracelsus Medical University, Salzburg, Austria
| | - Thomas Weber
- Department of Cardiology, Wels Hospital, Wels, Austria
| | - Bruno Watschinger
- Third Medical Department, Medical University of Vienna, Vienna, Austria
| | - Sabine Horn
- Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | | | - Heinz Drexel
- Department of Internal Medicine and Cardiology, Feldkirch Hospital, Feldkirch, Austria
| | - Marianne Brodmann
- Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Ernst Pilger
- Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Alexander Rosenkranz
- Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Erich Pohanka
- Medical Department, Linz General Hospital, Linz, Austria
| | | | - Otto Traindl
- First Medical Department, Mistelbach Hospital, Mistelbach, Austria
| | | | - Bernhard Metzler
- Third Medical Department, Medical University of Innsbruck, Innsbruck, Austria
| | - Hans-Peter Haring
- First Department of Neurology, Kepler University Clinic, Linz, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
607
|
Leng Z, Li R, Li Y, Wang L, Wang Y, Yang Y. Myocardial layer-specific analysis in patients with heterozygous familial hypercholesterolemia using speckle tracking echocardiography. Echocardiography 2017; 34:390-396. [PMID: 28052405 DOI: 10.1111/echo.13442] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Zhaoting Leng
- Department of Echocardiography; Beijing Anzhen Hospital; Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases; Beijing China
| | - Rongjuan Li
- Department of Echocardiography; Beijing Anzhen Hospital; Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases; Beijing China
| | - Yijia Li
- Department of Echocardiography; Beijing Anzhen Hospital; Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases; Beijing China
| | - Lvya Wang
- The Key Laboratory of Remodeling-related Cardiovascular Diseases; Ministry of Education; Department of Atherosclerosis; Beijing Anzhen Hospital; Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases; Beijing China
| | - Yueli Wang
- Department of Echocardiography; Beijing Anzhen Hospital; Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases; Beijing China
| | - Ya Yang
- Department of Echocardiography; Beijing Anzhen Hospital; Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases; Beijing China
| |
Collapse
|
608
|
Santos RD, Bourbon M, Alonso R, Cuevas A, Vasques-Cardenas NA, Pereira AC, Merchan A, Alves AC, Medeiros AM, Jannes CE, Krieger JE, Schreier L, Perez de Isla L, Magaña-Torres MT, Stoll M, Mata N, Dell Oca N, Corral P, Asenjo S, Bañares VG, Reyes X, Mata P. Clinical and molecular aspects of familial hypercholesterolemia in Ibero-American countries. J Clin Lipidol 2017; 11:160-166. [DOI: 10.1016/j.jacl.2016.11.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/10/2016] [Indexed: 11/26/2022]
|
609
|
Chiquette E, Oral EA, Garg A, Araújo-Vilar D, Dhankhar P. Estimating the prevalence of generalized and partial lipodystrophy: findings and challenges. Diabetes Metab Syndr Obes 2017; 10:375-383. [PMID: 29066925 PMCID: PMC5604558 DOI: 10.2147/dmso.s130810] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Lipodystrophy (LD; non-human immunodeficiency virus [HIV]-associated) syndromes are a rare body of disorders for which true prevalence is unknown. Prevalence estimates of rare diseases are important to increase awareness and financial resources. Current qualitative and quantitative estimates of LD prevalence range from ~0.1 to 90 cases/million. We demonstrate an approach to quantitatively estimate LD prevalence (all, generalized, and partial) through a search of 5 electronic medical record (EMR) databases and 4 literature searches. METHODS EMR and literature searches were conducted from 2012 to 2014. For the EMR database searches (Quintiles, IMS LifeLink, General Electric Healthcare, and Humedica EMR), LD cases were identified by the International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) code 272.6 (United Kingdom General Practice Research Database used other diagnostic codes to identify LD) plus additional LD-associated clinical characteristics (patients with HIV or documented HIV treatment were excluded). Expert adjudication of cases was used for the Quintiles database only. Literature searches (PubMed and EMBASE) were conducted for each of the 4 major LD subtypes. Prevalence estimates were determined by extrapolating the total number of cases identified for each search to the database population (EMR search) and European population (literature search). RESULTS The prevalence range of all LD across all EMR databases was 1.3-4.7 cases/million. For the adjudicated Quintiles search, the estimated prevalence of diagnosed LD was 3.07 cases/million (95% confidence interval [CI], 2.30-4.02), 0.23 cases/million (95% CI, 0.06-0.59) and 2.84 cases/million (95% CI, 2.10-3.75) for generalized lipodystrophy (GL) and partial lipodystrophy (PL), respectively. For all literature searches, the prevalence of all LD in Europe was 2.63 cases/million (0.96 and 1.67 cases/million for GL and PL, respectively). CONCLUSION LD prevalence estimates are at the lower range of previously established numbers, confirming that LD is an ultra-rare disease. The establishment of diagnostic criteria and coding specific to the 4 major LD subtypes and future studies/patient registries are needed to further refine our estimates.
Collapse
Affiliation(s)
- Elaine Chiquette
- Aegerion Pharmaceuticals, Cambridge, MA, USA
- Correspondence: Elaine Chiquette, Aegerion Pharmaceuticals, One Main Street, Cambridge, MA 02142, USA, Tel +1 857 242 5876, Fax +1 617 945 7968, Email
| | - Elif A Oral
- Brehm Center for Diabetes Research and Metabolism, Endocrinology and Diabetes Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases, Department of Internal Medicine, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David Araújo-Vilar
- Department of Medicine, UETeM, CIMUS School of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | |
Collapse
|
610
|
Stefanutti C. The 1 st and the 2 nd Italian Consensus Conferences on low-density lipoprotein-apheresis. A practical synopsis and update. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2017; 15:42-48. [PMID: 27416576 PMCID: PMC5269426 DOI: 10.2450/2016.0272-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/12/2016] [Indexed: 01/26/2023]
Abstract
The clinical indications and guidelines for low-density lipoprotein (LDL)-apheresis set by the 1st Italian Consensus Conference held in Ostuni in 1990 and completed in 1992, but never published, are reported schematically. In 1994, within the Project "Prevention and control of the factors of the disease (FATMA)" by the Italian National Research Council, subproject 8 "Control of cardiovascular disease", a "Hearing on therapeutic apheresis: need for a target-oriented project" was organised. The meeting was the last scientific initiative on LDL-apheresis supported by public funds in Italy. After roughly two decades of use of LDL-apheresis, new guidelines were required based on the latest scientific evidence. In 2006, the Italian multicentre study on LDL-apheresis Working Group (IMSLDLa-WP), a scientific initiative at national level, was developed. It initially gathered together 19 Italian centres qualified for the application of lipid apheresis and LDL-apheresis (2007-2008), then 23 in 2010, located in the north, south, centre of Italy and in Sicily and Sardinia. The multicentre study aimed to validate the protocol for selecting patients and to create a network between the Italian centres. A secondary objective was the creation of a database of patients with familial hypercholesterolaemia and other severe forms of dyslipidaemia undergoing treatment with LDL-apheresis using the available techniques. Since LDL-apheresis has multidisciplinary treatment indications, the agreement on the new guidelines was reached through a panel of experts, of different medical and surgical specialties, with scientific and medical interest in the treatment indications, application and development of LDL-apheresis. The initiatives of the IMSLDLa-WP led to the 2nd Italian Consensus Conference on LDL-apheresis held in Rome in 2009. The previous and most recent guidelines are reported here synoptically.
Collapse
Affiliation(s)
- Claudia Stefanutti
- Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Immunohematology and Transfusion Medicine, Department of Molecular Medicine, “Sapienza” University of Rome, “Umberto I” Hospital, Rome, Italy
| |
Collapse
|
611
|
Atypical Presentation and Treatment Response in a Child with Familial Hypercholesterolemia Having a Novel LDLR Mutation. JIMD Rep 2016. [PMID: 27933557 DOI: 10.1007/8904_2016_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an autosomal codominantly inherited disease. The severity of clinical presentation depends on the zygosity of the mutations in the LDLR, APOB, or PCSK9 genes. The homozygous form (HoFH) is associated with high mortality rate by third decade of life, while individuals with HeFH begin to suffer from premature cardiovascular disease in fourth or fifth decade of life. Statin drugs have helped to improve the biochemical profile and life expectancy in HeFH, while they are only minimally effective in HoFH. LDL apheresis remains an effective treatment option in HoFH, though limited by its availability and affordability issues. We present the case that highlights a few novel aspects of clinical and genetic heterogeneity in FH, wherein a child presented with features of both HeFH and HoFH. His clinical picture was that of HoFH; however he responded well clinically and biochemically to pharmacologic treatment only. DNA sequencing showed a novel heterozygous rare splicing variant in the LDLR gene in addition to a relatively high polygenic trait score comprised of LDL-C raising alleles from common polymorphic sites. Interestingly his normolipemic mother showed the same heterozygous mutation. Thus this novel splicing variant in LDLR showed nonclassical co-segregation with the disease phenotype and was associated with a high polygenic trait score comprised of common LDL-C raising polymorphic alleles in the affected proband. Thus it indicates the phenotypic heterogeneity of FH and suggests that secondary causes, such as polygenic factors and possibly as yet undetermined genetic or environmental factors, can exacerbate the metabolic phenotype in an individual who is genotypically heterozygous for FH.
Collapse
|
612
|
Vallejo-Vaz AJ, Akram A, Kondapally Seshasai SR, Cole D, Watts GF, Hovingh GK, Kastelein JJP, Mata P, Raal FJ, Santos RD, Soran H, Freiberger T, Abifadel M, Aguilar-Salinas CA, Alnouri F, Alonso R, Al-Rasadi K, Banach M, Bogsrud MP, Bourbon M, Bruckert E, Car J, Ceska R, Corral P, Descamps O, Dieplinger H, Do CT, Durst R, Ezhov MV, Fras Z, Gaita D, Gaspar IM, Genest J, Harada-Shiba M, Jiang L, Kayikcioglu M, Lam CSP, Latkovskis G, Laufs U, Liberopoulos E, Lin J, Lin N, Maher V, Majano N, Marais AD, März W, Mirrakhimov E, Miserez AR, Mitchenko O, Nawawi H, Nilsson L, Nordestgaard BG, Paragh G, Petrulioniene Z, Pojskic B, Reiner Ž, Sahebkar A, Santos LE, Schunkert H, Shehab A, Slimane MN, Stoll M, Su TC, Susekov A, Tilney M, Tomlinson B, Tselepis AD, Vohnout B, Widén E, Yamashita S, Catapano AL, Ray KK. Pooling and expanding registries of familial hypercholesterolaemia to assess gaps in care and improve disease management and outcomes: Rationale and design of the global EAS Familial Hypercholesterolaemia Studies Collaboration. ATHEROSCLEROSIS SUPP 2016; 22:1-32. [PMID: 27939304 DOI: 10.1016/j.atherosclerosissup.2016.10.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The potential for global collaborations to better inform public health policy regarding major non-communicable diseases has been successfully demonstrated by several large-scale international consortia. However, the true public health impact of familial hypercholesterolaemia (FH), a common genetic disorder associated with premature cardiovascular disease, is yet to be reliably ascertained using similar approaches. The European Atherosclerosis Society FH Studies Collaboration (EAS FHSC) is a new initiative of international stakeholders which will help establish a global FH registry to generate large-scale, robust data on the burden of FH worldwide. METHODS The EAS FHSC will maximise the potential exploitation of currently available and future FH data (retrospective and prospective) by bringing together regional/national/international data sources with access to individuals with a clinical and/or genetic diagnosis of heterozygous or homozygous FH. A novel bespoke electronic platform and FH Data Warehouse will be developed to allow secure data sharing, validation, cleaning, pooling, harmonisation and analysis irrespective of the source or format. Standard statistical procedures will allow us to investigate cross-sectional associations, patterns of real-world practice, trends over time, and analyse risk and outcomes (e.g. cardiovascular outcomes, all-cause death), accounting for potential confounders and subgroup effects. CONCLUSIONS The EAS FHSC represents an excellent opportunity to integrate individual efforts across the world to tackle the global burden of FH. The information garnered from the registry will help reduce gaps in knowledge, inform best practices, assist in clinical trials design, support clinical guidelines and policies development, and ultimately improve the care of FH patients.
Collapse
Affiliation(s)
| | - Antonio J Vallejo-Vaz
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), School of Public Health, Imperial College London, London, UK.
| | - Asif Akram
- Global eHealth Unit, School of Public Health, Imperial College London, London, UK; Centre for Population Health Sciences, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | | - Della Cole
- Cardiovascular and Cell Sciences Research Institute, St George's, University of London, London, UK
| | - Gerald F Watts
- Cardiovascular Medicine, Royal Perth Hospital, University of Western Australia, Perth, Australia
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - John J P Kastelein
- Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Frederick J Raal
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Raul D Santos
- Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Handrean Soran
- University Department of Medicine, Central Manchester University Hospitals, Manchester, UK
| | - Tomas Freiberger
- Centre for Cardiovascular Surgery and Transplantation, Brno, Czech Republic; Ceitec, Masaryk University, Brno, Czech Republic
| | - Marianne Abifadel
- Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Saint-Joseph University, Beirut, Lebanon
| | | | - Fahad Alnouri
- Cardiovascular Prevention and Rehabilitation Unit, Prince Sultan Cardiac Centre Riyadh, Riyadh, Saudi Arabia
| | - Rodrigo Alonso
- Lipid Clinic, Department of Nutrition, Clínica Las Condes, Santiago de Chile, Chile
| | | | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Lodz, Poland
| | - Martin P Bogsrud
- National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Norway
| | - Mafalda Bourbon
- Instituto Nacional de Saúde Doutor Ricardo Jorge and Biosystems & Integrative Sciences Institute (BioISI), Universidade de Lisboa, Portugal
| | - Eric Bruckert
- Endocrinologie, métabolisme et prévention cardiovasculaire, Institut E3M et IHU cardiométabolique (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Josip Car
- Global eHealth Unit, School of Public Health, Imperial College London, London, UK; Centre for Population Health Sciences, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Richard Ceska
- Charles University in Prague, Prague, Czech Republic
| | - Pablo Corral
- FASTA University, School of Medicine, Mar del Plata, Argentina
| | | | - Hans Dieplinger
- Austrian Atherosclerosis Society, c/o Division of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Can T Do
- Vietnam Heart Institute, Bach Mai Hospital, Hanoi, Viet Nam
| | - Ronen Durst
- Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Marat V Ezhov
- Russian Cardiology Research and Production Centre, Moscow, Russia
| | - Zlatko Fras
- University Medical Centre Ljubljana, Division of Medicine, Preventive Cardiology Unit, Ljubljana, Slovenia; Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Dan Gaita
- Universitatea de Medicina si Farmacie Victor Babes din Timisoara, Romania
| | - Isabel M Gaspar
- Medical Genetics Department, Centro Hospitalar de Lisboa Ocidental and Genetics Laboratory, Lisbon Medical School, University of Lisbon, Portugal
| | | | - Mariko Harada-Shiba
- National Cerebral and Cardiovascular Centre Research Institute, Osaka, Japan
| | - Lixin Jiang
- National Clinical Research Centre of Cardiovascular Diseases, Fuwai Hospital, National Centre for Cardiovascular Diseases, Beijing, China
| | - Meral Kayikcioglu
- Ege University Medical School, Department of Cardiology, Izmir, Turkey
| | - Carolyn S P Lam
- National Heart Centre Singapore and Duke-National University of Singapore, Singapore
| | - Gustavs Latkovskis
- Research Institute of Cardiology and Regenerative Therapy, Faculty of Medicine, University of Latvia, Paul Stradins Clinical University Hospital, Riga, Latvia
| | | | | | - Jie Lin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Nan Lin
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), School of Public Health, Imperial College London, London, UK
| | | | | | - A David Marais
- University of Cape Town and National Health Laboratory Service, South Africa
| | - Winfried März
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Germany
| | | | - André R Miserez
- Diagene GmbH, Research Institute, Reinach, Switzerland; Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Olena Mitchenko
- Dyslipidaemia Department, Institute of Cardiology AMS of Ukraine, Ukraine
| | - Hapizah Nawawi
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM) and Faculty of Medicine, Universiti Teknologi MARA, Malaysia
| | - Lennart Nilsson
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Børge G Nordestgaard
- Herlev and Gentofte Hospital, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - György Paragh
- Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary
| | - Zaneta Petrulioniene
- Vilnius University Santariskiu Hospital, Centre of Cardiology and Angiology, Vilnius, Lithuania
| | | | - Željko Reiner
- Department for Metabolic Diseases, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Croatia
| | - Amirhossein Sahebkar
- Biotechnology Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Lourdes E Santos
- Cardinal Santos Medical Centre, University of the Philippines - Philippine General Hospital (UP-PGH), Philippines
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Technische Universität München, Deutsches Zentrum für Herz- und Kreislauferkrankungen (DZHK), Munich Heart Alliance, Germany
| | | | - M Naceur Slimane
- Research Unit on Dyslipidaemia and Atherosclerosis, Faculty of Medicine of Monastir, Tunisia
| | - Mario Stoll
- Cardiovascular Genetic Laboratory, Cardiovascular Health Commission, Montevideo, Uruguay
| | - Ta-Chen Su
- Department of Internal Medicine and Cardiovascular Centre, National Taiwan University Hospital, Taipei, Taiwan
| | - Andrey Susekov
- Department of Clinical Pharmacology and Therapeutics, Russian Medical Academy of Postgraduate Education, Ministry of Health of Russian Federation, Russia
| | - Myra Tilney
- Faculty of Medicine & Surgery, Medical School, Mater Dei Hospital, University of Malta, Malta
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | | | - Branislav Vohnout
- Coordination Centre for Familial Hyperlipoproteinemias, Institute of Nutrition, FOZOS, Slovak Medical University, Department of Epidemiology, School of Medicine, Comenius University, Bratislava, Slovakia
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Shizuya Yamashita
- Rinku General Medical Centre and Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), School of Public Health, Imperial College London, London, UK
| |
Collapse
|
613
|
Alim A, Tokat Y, Erdogan Y, Gokkaya Z, Dayangac M, Yuzer Y, Oezcelik A. Liver transplantation for homozygote familial hypercholesterolemia: the only curative treatment. Pediatr Transplant 2016; 20:1060-1064. [PMID: 27435024 DOI: 10.1111/petr.12763] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2016] [Indexed: 11/28/2022]
Abstract
FH is an autosomal dominant genetic disorder characterized by increased TC and LDL level, which leads to xanthomas, atherosclerosis, and cardiac complications even in childhood. The treatment options are diet, medical treatment, lipid apheresis, and LT. The aim of our study was to analyze our data of patients with FH. Between 2004 and 2015, there were 51 patients who underwent pediatric LT at our center. All patients with FH were identified, and the data were retrospectively analyzed. There were eight patients with homozygous FH in the median age of 10 years (IQR 6-12) who underwent LT. The median pre-operative TC and LDL levels were 611 mg/dL (IQR: 460-844) and 574 mg/dL (IQR: 398-728) and decreased to normal levels 1 week after LT (TC: 193 mg/dL and LDL: 141 mg/dL). Two patients died two and 18 months after LT due to sudden cardiac arrest. Both patients were diagnosed with cardiovascular disease pre-operatively. The LT is the only curative treatment for this disease. To achieve an excellent outcome, it should be performed before the development of cardiovascular disease, because the regression of severe cardiovascular disease after transplantation is limited.
Collapse
Affiliation(s)
- Altan Alim
- Department of General and Transplantation Surgery, University Hospital of the Istanbul Bilim University, Istanbul, Turkey
| | - Yaman Tokat
- Department of General and Transplantation Surgery, University Hospital of the Istanbul Bilim University, Istanbul, Turkey
| | - Yalcin Erdogan
- Department of General and Transplantation Surgery, University Hospital of the Istanbul Bilim University, Istanbul, Turkey
| | - Zafer Gokkaya
- Department of General and Transplantation Surgery, University Hospital of the Istanbul Bilim University, Istanbul, Turkey
| | - Murat Dayangac
- Department of General and Transplantation Surgery, University Hospital of the Istanbul Bilim University, Istanbul, Turkey
| | - Yildiray Yuzer
- Department of General and Transplantation Surgery, University Hospital of the Istanbul Bilim University, Istanbul, Turkey
| | - Arzu Oezcelik
- Department of General and Transplantation Surgery, University Hospital of the Istanbul Bilim University, Istanbul, Turkey
| |
Collapse
|
614
|
Gidding SS. The complexities of homozygous familial hypercholesterolemia management. Pediatr Transplant 2016; 20:1020-1021. [PMID: 27882690 DOI: 10.1111/petr.12812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
615
|
Wang J, Dron JS, Ban MR, Robinson JF, McIntyre AD, Alazzam M, Zhao PJ, Dilliott AA, Cao H, Huff MW, Rhainds D, Low-Kam C, Dubé MP, Lettre G, Tardif JC, Hegele RA. Polygenic Versus Monogenic Causes of Hypercholesterolemia Ascertained Clinically. Arterioscler Thromb Vasc Biol 2016; 36:2439-2445. [DOI: 10.1161/atvbaha.116.308027] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/10/2016] [Indexed: 11/16/2022]
Abstract
Objective—
Next-generation sequencing technology is transforming our understanding of heterozygous familial hypercholesterolemia, including revision of prevalence estimates and attribution of polygenic effects. Here, we examined the contributions of monogenic and polygenic factors in patients with severe hypercholesterolemia referred to a specialty clinic.
Approach and Results—
We applied targeted next-generation sequencing with custom annotation, coupled with evaluation of large-scale copy number variation and polygenic scores for raised low-density lipoprotein cholesterol in a cohort of 313 individuals with severe hypercholesterolemia, defined as low-density lipoprotein cholesterol >5.0 mmol/L (>194 mg/dL). We found that (1) monogenic familial hypercholesterolemia–causing mutations detected by targeted next-generation sequencing were present in 47.3% of individuals; (2) the percentage of individuals with monogenic mutations increased to 53.7% when copy number variations were included; (3) the percentage further increased to 67.1% when individuals with extreme polygenic scores were included; and (4) the percentage of individuals with an identified genetic component increased from 57.0% to 92.0% as low-density lipoprotein cholesterol level increased from 5.0 to >8.0 mmol/L (194 to >310 mg/dL).
Conclusions—
In a clinically ascertained sample with severe hypercholesterolemia, we found that most patients had a discrete genetic basis detected using a comprehensive screening approach that includes targeted next-generation sequencing, an assay for copy number variations, and polygenic trait scores.
Collapse
Affiliation(s)
- Jian Wang
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Jacqueline S. Dron
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Matthew R. Ban
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - John F. Robinson
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Adam D. McIntyre
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Maher Alazzam
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Pei Jun Zhao
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Allison A. Dilliott
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Henian Cao
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Murray W. Huff
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - David Rhainds
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Cécile Low-Kam
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Marie-Pierre Dubé
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Guillaume Lettre
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Jean-Claude Tardif
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| | - Robert A. Hegele
- From the Robarts Research Institute (J.W., J.S.D., M.R.B., J.F.R., A.D.M., A.A.D., H.C., M.W.H., R.A.H.), Department of Biochemistry (J.S.D., M.A., A.A.D., M.W.H., R.A.H.), and Department of Medicine (P.J.Z., M.W.H., R.A.H.), Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Faculté de Médicine, Université de Montréal, Québec, Canada (M.-P.D., G.L., J.-C.T.); and Montréal Heart institute, Québec, Canada (D.R., C.L.-K., M.-P.D., G.L., J.-C.T.)
| |
Collapse
|
616
|
France M, Rees A, Datta D, Thompson G, Capps N, Ferns G, Ramaswami U, Seed M, Neely D, Cramb R, Shoulders C, Barbir M, Pottle A, Eatough R, Martin S, Bayly G, Simpson B, Halcox J, Edwards R, Main L, Payne J, Soran H. HEART UK statement on the management of homozygous familial hypercholesterolaemia in the United Kingdom. Atherosclerosis 2016; 255:128-139. [DOI: 10.1016/j.atherosclerosis.2016.10.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/03/2016] [Accepted: 10/07/2016] [Indexed: 12/16/2022]
|
617
|
Sánchez-Hernández RM, Civeira F, Stef M, Perez-Calahorra S, Almagro F, Plana N, Novoa FJ, Sáenz-Aranzubía P, Mosquera D, Soler C, Fuentes FJ, Brito-Casillas Y, Real JT, Blanco-Vaca F, Ascaso JF, Pocovi M. Homozygous Familial Hypercholesterolemia in Spain. ACTA ACUST UNITED AC 2016; 9:504-510. [DOI: 10.1161/circgenetics.116.001545] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/28/2016] [Indexed: 11/16/2022]
Abstract
Background—
Homozygous familial hypercholesterolemia (HoFH) is a rare disease characterized by elevated plasma levels of low-density lipoprotein cholesterol (LDL-C) and extremely high risk of premature atherosclerotic cardiovascular disease. HoFH is caused by mutations in several genes, including LDL receptor (
LDLR
), apolipoprotein B (
APOB
), proprotein convertase subtilisin/kexin type 9 (
PCSK9
), and LDL protein receptor adaptor 1 (
LDLRAP1
). No epidemiological studies have assessed HoFH prevalence or the clinical and molecular characteristics of this condition. Here, we aimed to characterize HoFH in Spain.
Methods and Results—
Data were collected from the Spanish Dyslipidemia Registry of the Spanish Atherosclerosis Society and from all molecular diagnoses performed for familial hypercholesterolemia in Spain between 1996 and 2015 (n=16 751). Clinical data included baseline lipid levels and atherosclerotic cardiovascular disease events. A total of 97 subjects were identified as having HoFH—of whom, 47 were true homozygous (1 for
APOB
, 5 for
LDLRAP1
, and 41 for
LDLR
), 45 compound heterozygous for
LDLR
, 3 double heterozygous for
LDLR
and
PSCK9
, and 2 double heterozygous for
LDLR
and
APOB
. No
PSCK9
homozygous cases were identified. Two variants in
LDLR
were identified in 4.8% of the molecular studies. Over 50% of patients did not meet the classical HoFH diagnosis criteria. The estimated HoFH prevalence was 1:450 000. Compared with compound heterozygous cases, true homozygous cases showed more aggressive phenotypes with higher LDL-C and more atherosclerotic cardiovascular disease events.
Conclusions—
HoFH frequency in Spain was higher than expected. Clinical criteria would underestimate the actual prevalence of individuals with genetic HoFH, highlighting the importance of genetic analysis to improve familial hypercholesterolemia diagnosis accuracy.
Collapse
|
618
|
Gouni‐Berthold I, Descamps OS, Fraass U, Hartfield E, Allcott K, Dent R, März W. Systematic review of published Phase 3 data on anti-PCSK9 monoclonal antibodies in patients with hypercholesterolaemia. Br J Clin Pharmacol 2016; 82:1412-1443. [PMID: 27478094 PMCID: PMC5099564 DOI: 10.1111/bcp.13066] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/14/2016] [Accepted: 07/04/2016] [Indexed: 01/06/2023] Open
Abstract
AIMS Two anti-proprotein convertase subtilisin/kexin type 9 (PCSK9) monoclonal antibodies, alirocumab and evolocumab, have been approved for the treatment of hypercholesterolaemia in certain patients. We reviewed data from Phase 3 studies to evaluate the efficacy and safety of these antibodies. METHODS We systematically reviewed Phase 3 English-language studies in patients with hypercholesterolaemia, published between 1 January 2005 and 20 October 2015. Congress proceedings from 16 November 2012 to 16 November 2015 were also reviewed. RESULTS We identified 12 studies of alirocumab and nine of evolocumab, including over 10 000 patients overall. Most studies enrolled patients with hypercholesterolaemia and used anti-PCSK9 antibodies with statins. The ODYSSEY FH I, FH II and HIGH FH alirocumab studies and the RUTHERFORD-2 evolocumab study exclusively recruited patients with heterozygous familial hypercholesterolaemia. Two evolocumab studies focused mainly on homozygous familial hypercholesterolaemia (HoFH): TESLA Part B and TAUSSIG (a TESLA sub-study); only those data for HoFH are reported here. All comparator studies demonstrated a reduction in LDL cholesterol (LDL-C) with the anti-PCSK9 antibodies. No head-to-head studies were conducted between alirocumab and evolocumab. Up to 87% of patients receiving alirocumab and up to 98% receiving evolocumab reached LDL-C goals. Both antibodies were effective and well tolerated across a broad population of patients and in specific subgroups, such as those with type 2 diabetes. CONCLUSIONS Using anti-PCSK9 antibodies as add-on therapy to other lipid-lowering treatments or as monotherapy for patients unable to tolerate statins may help patients with high cardiovascular risk to achieve their LDL-C goals.
Collapse
Affiliation(s)
- Ioanna Gouni‐Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine (ZEDP)University of CologneCologneGermany
| | - Olivier S. Descamps
- Department of Internal MedicineCentres Hospitaliers JolimontHaine Saint‐PaulBelgium
| | | | | | | | | | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory DiagnosticsMedical University GrazGrazAustria
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty MannheimUniversity of HeidelbergGermany
- Synlab AcademySynlab Holding Deutschland GmbHMannheim and AugsburgGermany
| |
Collapse
|
619
|
Labib D, Soliman H, Said K, Sorour K. Early severe coronary artery disease and aortic coarctation in a child with familial hypercholesterolaemia. BMJ Case Rep 2016; 2016:bcr-2016-216147. [PMID: 27903575 DOI: 10.1136/bcr-2016-216147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
An 11-year-old boy presented with easy fatigability, multiple xanthomas, and absent pedal pulsations. Laboratory workup showed severe hypercholesterolaemia and non-invasive imaging revealed 'normally functioning' bicuspid aortic valve and tight aortic coarctation. Coronary angiography showed severe right coronary artery (RCA) stenosis. Medical treatment resulted in significant improvement of dyslipidaemia. We successfully performed balloon dilation and stenting of his coarctation, as well as percutaneous coronary intervention for RCA lesion.
Collapse
Affiliation(s)
- Dina Labib
- Department of Cardiovascular Medicine, Cairo University, Giza, Egypt
| | - Haytham Soliman
- Department of Cardiovascular Medicine, Cairo University, Giza, Egypt
| | - Kareem Said
- Department of Cardiovascular Medicine, Cairo University, Giza, Egypt
| | - Khaled Sorour
- Department of Cardiovascular Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
620
|
Collins R, Reith C, Emberson J, Armitage J, Baigent C, Blackwell L, Blumenthal R, Danesh J, Smith GD, DeMets D, Evans S, Law M, MacMahon S, Martin S, Neal B, Poulter N, Preiss D, Ridker P, Roberts I, Rodgers A, Sandercock P, Schulz K, Sever P, Simes J, Smeeth L, Wald N, Yusuf S, Peto R. Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet 2016; 388:2532-2561. [PMID: 27616593 DOI: 10.1016/s0140-6736(16)31357-5] [Citation(s) in RCA: 1254] [Impact Index Per Article: 139.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023]
Abstract
This Review is intended to help clinicians, patients, and the public make informed decisions about statin therapy for the prevention of heart attacks and strokes. It explains how the evidence that is available from randomised controlled trials yields reliable information about both the efficacy and safety of statin therapy. In addition, it discusses how claims that statins commonly cause adverse effects reflect a failure to recognise the limitations of other sources of evidence about the effects of treatment. Large-scale evidence from randomised trials shows that statin therapy reduces the risk of major vascular events (ie, coronary deaths or myocardial infarctions, strokes, and coronary revascularisation procedures) by about one-quarter for each mmol/L reduction in LDL cholesterol during each year (after the first) that it continues to be taken. The absolute benefits of statin therapy depend on an individual's absolute risk of occlusive vascular events and the absolute reduction in LDL cholesterol that is achieved. For example, lowering LDL cholesterol by 2 mmol/L (77 mg/dL) with an effective low-cost statin regimen (eg, atorvastatin 40 mg daily, costing about £2 per month) for 5 years in 10 000 patients would typically prevent major vascular events from occurring in about 1000 patients (ie, 10% absolute benefit) with pre-existing occlusive vascular disease (secondary prevention) and in 500 patients (ie, 5% absolute benefit) who are at increased risk but have not yet had a vascular event (primary prevention). Statin therapy has been shown to reduce vascular disease risk during each year it continues to be taken, so larger absolute benefits would accrue with more prolonged therapy, and these benefits persist long term. The only serious adverse events that have been shown to be caused by long-term statin therapy-ie, adverse effects of the statin-are myopathy (defined as muscle pain or weakness combined with large increases in blood concentrations of creatine kinase), new-onset diabetes mellitus, and, probably, haemorrhagic stroke. Typically, treatment of 10 000 patients for 5 years with an effective regimen (eg, atorvastatin 40 mg daily) would cause about 5 cases of myopathy (one of which might progress, if the statin therapy is not stopped, to the more severe condition of rhabdomyolysis), 50-100 new cases of diabetes, and 5-10 haemorrhagic strokes. However, any adverse impact of these side-effects on major vascular events has already been taken into account in the estimates of the absolute benefits. Statin therapy may cause symptomatic adverse events (eg, muscle pain or weakness) in up to about 50-100 patients (ie, 0·5-1·0% absolute harm) per 10 000 treated for 5 years. However, placebo-controlled randomised trials have shown definitively that almost all of the symptomatic adverse events that are attributed to statin therapy in routine practice are not actually caused by it (ie, they represent misattribution). The large-scale evidence available from randomised trials also indicates that it is unlikely that large absolute excesses in other serious adverse events still await discovery. Consequently, any further findings that emerge about the effects of statin therapy would not be expected to alter materially the balance of benefits and harms. It is, therefore, of concern that exaggerated claims about side-effect rates with statin therapy may be responsible for its under-use among individuals at increased risk of cardiovascular events. For, whereas the rare cases of myopathy and any muscle-related symptoms that are attributed to statin therapy generally resolve rapidly when treatment is stopped, the heart attacks or strokes that may occur if statin therapy is stopped unnecessarily can be devastating.
Collapse
Affiliation(s)
- Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Christina Reith
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jonathan Emberson
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jane Armitage
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Colin Baigent
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Lisa Blackwell
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Roger Blumenthal
- Ciccarone Center for the Prevention of Heart Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - David DeMets
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Stephen Evans
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Malcolm Law
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Stephen MacMahon
- The George Institute for Global Health, University of Sydney, Sydney, Australia
| | - Seth Martin
- Ciccarone Center for the Prevention of Heart Disease, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruce Neal
- The George Institute for Global Health, University of Sydney, Sydney, Australia
| | - Neil Poulter
- International Centre for Circulatory Health & Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - David Preiss
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Paul Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ian Roberts
- Clinical Trials Unit, London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Anthony Rodgers
- The George Institute for Global Health, University of Sydney, Sydney, Australia
| | - Peter Sandercock
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kenneth Schulz
- FHI 360, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Peter Sever
- International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London, London, UK
| | - John Simes
- National Health and Medical Research Council Clinical Trial Centre, University of Sydney, Sydney, Australia
| | - Liam Smeeth
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Nicholas Wald
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Salim Yusuf
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, ON, Canada
| | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit and MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
621
|
The use of targeted exome sequencing in genetic diagnosis of young patients with severe hypercholesterolemia. Sci Rep 2016; 6:36823. [PMID: 27830735 PMCID: PMC5103295 DOI: 10.1038/srep36823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/18/2016] [Indexed: 12/17/2022] Open
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant disorder. Although genetic testing is an important tool for detecting FH-causing mutations in patients, diagnostic methods for young patients with severe hypercholesterolemia are understudied. This study compares the target exome sequencing (TES) technique with the DNA resequencing array technique on young patients with severe hypercholesterolemia. A total of 20 unrelated patients (mean age 14.8 years) with total cholesterol > 10 mmol/L were included. 12 patient samples were processed by DNA resequencing array, 14 patient samples were processed by TES, and 6 patient samples were processed by both methods. Functional characterization of novel mutations was performed by flow cytometry. The mutation detection rate (MDR) of DNA resequencing array was 75%, while the MDR of TES was 100%. A total of 27 different mutations in the LDLR were identified, including 3 novel mutations and 8 mutations with previously unknown pathogenicity. Functional characterization of c.673delA, c.1363delC, p.Leu575Phe and p.Leu582Phe variants found that all of them are pathogenic. Additionally, 7 patients were diagnosed with Heterozygous FH (HeFH) in which lipid levels were significantly higher than common HeFH patients. This data indicates that TES is a very efficient tool for genetic diagnosis in young patients with severe hypercholesterolemia.
Collapse
|
622
|
Greco M, Robinson JD, Eltayeb O, Benuck I. Progressive Aortic Stenosis in Homozygous Familial Hypercholesterolemia After Liver Transplant. Pediatrics 2016; 138:peds.2016-0740. [PMID: 27940769 DOI: 10.1542/peds.2016-0740] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/28/2016] [Indexed: 11/24/2022] Open
Abstract
Early onset coronary artery disease and aortic calcifications are characteristic features of patients with homozygous familial hypercholesterolemia. Standard medical therapy includes dietary modification, pharmacotherapy, and lipoprotein apheresis to lower serum low-density lipoprotein cholesterol (LDL-C). Liver transplant is a surgical option for the treatment of homozygous familial hypercholesterolemia and can lead to normal cholesterol levels. Vascular calcifications are known to progress despite standard medical therapy and have been reported after liver transplant in the setting of rejection. We present the first report of progressive severe aortic valve stenosis in a patient despite liver transplant with normalization of lipid levels and no history of graft rejection.
Collapse
Affiliation(s)
| | - Joshua D Robinson
- Divisions of Cardiology and.,Departments of Pediatrics.,Radiology, and
| | - Osama Eltayeb
- Cardiothoracic Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois; and.,Surgery, Northwestern University Feinberg School of Medicine, Chicago Illinois
| | - Irwin Benuck
- Divisions of Cardiology and.,Departments of Pediatrics
| |
Collapse
|
623
|
Abstract
Homozygous familial hypercholesterolaemia (HoFH) is an inherited disease causing an approximately fourfold increase in blood low-density lipoprotein cholesterol (LDLC) from birth compared with the age-matched normal population owing to reduced low-density lipoprotein receptor (LDLR) activity. Such elevated cholesterol is associated with accelerated atheromatous disease, particularly of the aortic root and coronary arteries. However, HoFH is clinically heterogeneous, reflecting residual low-density lipoprotein receptor (LDLR) activity. The main objective in treating children may be stated to be the avoidance of irreversible cardiac damage requiring heart transplantation by sufficient lowering of blood cholesterol. Lipoprotein apheresis or plasmapheresis are safe means of lowering cholesterol but may be insufficient on their own. Statin drugs, PCSK9 inhibitors ezetimibe and bile acid sequestrants are relatively ineffective if LDLR activity is lacking, but should be used if effective. Two new drugs, lomitapide and mipomersen, have been licensed specifically for HoFH by some regulatory authorities. They work by reducing LDL production rate. They have been associated with fatty liver in adults. Evidence of safety in children is lacking. An alternative is liver transplantation, which replaces the missing LDLR and normalises cholesterol. Clinicians are faced with a dilemma in choosing between these options or deferring such treatment associated with potential harm. Individual case descriptions are an important means of informing clinical judgement. Management of the two cases described in this issue is discussed in the light of modern developments in transplantation and pharmacotherapy.
Collapse
Key Words
- ADH, autosomal dominant hypercholesterolaemia, refers to hypercholesterolaemia owing to a single mutation of an allele of a gene affecting LDLR activity
- APOB, apolipoprotein B, is the main protein component of LDL and is the ligand for LDL receptors in the liver
- ARH, autosomal recessive hypercholesterolaemia, refers to hypercholesterolaemia owing to a mutation of both alleles of a single gene affecting LDLR activity
- Evolucomab
- FH, familial hypercholesterolaemia, is an inherited condition causing reduced LDLR activity with consequent hypercholesterolaemia
- HeFH, heterozygous familial hypercholesterolaemia, is caused by one mutant allele of genes affecting LDLR activity
- HoFH, homozygous familial hypercholesterolaemia is caused by two mutant alleles of genes affecting LDLR activity
- Homozygous familial hypercholesterolaemia
- LDL, low-density lipoprotein, is a complex of cholesterol attached to a lipoprotein particle which is removed from blood mainly by the liver
- LDLC, LDL cholesterol, refers to the cholesterol component of LDL
- LDLR, LDL receptors, mediate LDL uptake by the liver
- LDLRAP1, a protein called LDLR adaptor protein 1, facilitates LDLR function
- Lipoprotein apheresis
- Liver transplantation
- Lomitapide
- Microsomal triglyceride transfer protein is an enzyme involved in the hepatic assembly of triglyceride, cholesterol and APOB into triglyceride-rich particles which are secreted by the liver. These particles are metabolised to LDL
- PCSK9
- PCSK9, a protein called proprotein convertase subtilisin/kexin type 9, increases the rate of degradation of LDLR
Collapse
Affiliation(s)
- Michael France
- a Cardiovascular Trials Unit , Central Manchester University Hospitals Foundation Trust and Cardiovascular Research Group, University of Manchester , Manchester , UK
| |
Collapse
|
624
|
What is the actual epidemiology of familial hypercholesterolemia in Italy? Evidence from a National Primary Care Database. Int J Cardiol 2016; 223:701-705. [DOI: 10.1016/j.ijcard.2016.08.269] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/16/2016] [Indexed: 11/21/2022]
|
625
|
Stempel H, Dodge A, Marriott E, Peterson AL. Referral Patterns and Cascade Screening for Familial Hypercholesterolemia in a Pediatric Lipid Clinic. J Pediatr 2016; 178:285-287. [PMID: 27592098 DOI: 10.1016/j.jpeds.2016.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/11/2016] [Accepted: 08/04/2016] [Indexed: 12/18/2022]
Abstract
Charts of 42 children with familial hypercholesterolemia from a dyslipidemia clinic were reviewed for initial cholesterol screen indication and cascade screening results. Indications were universal screening (8/28 after guideline release, none before), family history (26/42), risk factor (5/42), and other (3/42). Cascade screening identified 63 relatives with unknown familial hypercholesterolemia.
Collapse
Affiliation(s)
- Hilary Stempel
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ann Dodge
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Erin Marriott
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Amy L Peterson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
626
|
Deswal S, Kapoor A, Sibal A, Kohli V. Homozygous familial hypercholesterolaemia in two boys aged 5 and 10 years. Paediatr Int Child Health 2016; 36:308-311. [PMID: 27376185 DOI: 10.1080/20469047.2016.1188497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Familial hypercholesterolaemia (FH) is an autosomal dominant lipid disorder. Homozygous FH (HFH), though rare, presents in early childhood. Two different presentations of HFH are reported. The first child presented at 5 years of age with xanthomas on the knees, elbows and buttocks and failure to thrive since the second year of life. He was found to be hypertensive with moderate aortic regurgitation. He is now stable on statins and antihypertensives. The second child presented at 10 years of age with multiple xanthomas and severe aortic stenosis. He died of refractory cardiac failure despite emergency aortic balloon valvoplasty due to diffuse coronary artery disease. Strong clinical suspicion can aid early diagnosis and delay cardiovascular complications.
Collapse
Affiliation(s)
- Shivani Deswal
- a Department of Pediatrics , Postgraduate Institute of Medical Education and Research and Dr RML Hospital , New Delhi
| | | | | | - Vikas Kohli
- c Pediatric Cardiology and Cardiothoracic surgery, Apollo Centre for Advanced Pediatrics , Indraprastha Apollo Hospital , New Delhi , India
| |
Collapse
|
627
|
Bays HE, Jones PH, Orringer CE, Brown WV, Jacobson TA. National Lipid Association Annual Summary of Clinical Lipidology 2016. J Clin Lipidol 2016; 10:S1-43. [PMID: 26891998 DOI: 10.1016/j.jacl.2015.08.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/03/2015] [Indexed: 01/25/2023]
Abstract
The National Lipid Association (NLA) Annual Summary of Clinical Lipidology is a yearly updated summary of principles important to the patient-centered evaluation, management, and care of patients with dyslipidemia. This summary is intended to be a "living document," with future annual updates based on emerging science, clinical considerations, and new NLA Position, Consensus, and Scientific Statements, thus providing an ongoing resource that applies the latest in medical science towards the clinical management of patients with dyslipidemia. Topics include the NLA Recommendations for Patient-Centered Management of Dyslipidemia, genetics, Familial Hypercholesterolemia, secondary causes of dyslipidemia, biomarkers and advanced lipid testing, nutrition, physical activity, obesity, adiposopathy, metabolic syndrome, diabetes mellitus, lipid pharmacotherapy, lipid-altering drug interactions, lipoprotein apheresis, dyslipidemia management and treatment based upon age (children, adolescents, and older individuals), dyslipidemia considerations based upon race, ethnicity and gender, dyslipidemia and human immune virus infection, dyslipidemia and immune disorders, adherence strategies and collaborative care, and lipid-altering drugs in development. Hyperlinks direct the reader to sentinel online tables, charts, and figures relevant to lipidology, access to online atherosclerotic cardiovascular disease risk calculators, worldwide lipid guidelines, recommendations, and position/scientific statements, as well as links to online audio files, websites, slide shows, applications, continuing medical education opportunities, and patient information.
Collapse
Affiliation(s)
- Harold E Bays
- Louisville Metabolic and Atherosclerosis Research Center, Louisville, KY, USA.
| | | | - Carl E Orringer
- University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | | | | |
Collapse
|
628
|
Landmesser U, John Chapman M, Farnier M, Gencer B, Gielen S, Hovingh GK, Lüscher TF, Sinning D, Tokgözoğlu L, Wiklund O, Zamorano JL, Pinto FJ, Catapano AL. European Society of Cardiology/European Atherosclerosis Society Task Force consensus statement on proprotein convertase subtilisin/kexin type 9 inhibitors: practical guidance for use in patients at very high cardiovascular risk. Eur Heart J 2016; 38:2245-2255. [DOI: 10.1093/eurheartj/ehw480] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022] Open
|
629
|
Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, Hoes AW, Jennings CS, Landmesser U, Pedersen TR, Reiner Ž, Riccardi G, Taskinen MR, Tokgozoglu L, Verschuren WMM, Vlachopoulos C, Wood DA, Zamorano JL, Cooney MT. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. Eur Heart J 2016; 37:2999-3058. [PMID: 27567407 DOI: 10.1093/eurheartj/ehw272] [Citation(s) in RCA: 1973] [Impact Index Per Article: 219.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
630
|
Setia N, Saxena R, Arora A, Verma IC. Spectrum of mutations in homozygous familial hypercholesterolemia in India, with four novel mutations. Atherosclerosis 2016; 255:31-36. [PMID: 27816806 DOI: 10.1016/j.atherosclerosis.2016.10.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS Homozygous familial hypercholesterolemia (FH) is a rare but serious, inherited disorder of lipid metabolism characterized by very high total and LDL cholesterol levels from birth. It presents as cutaneous and tendon xanthomas since childhood, with or without cardiac involvement. FH is commonly caused by mutations in three genes, i.e. LDL receptor (LDLR), apolipoprotein B (ApoB) and PCSK9. We aimed to determine the spectrum of mutations in cases of homozygous FH in Asian Indians and evaluate if there was any similarity to the mutations observed in Caucasians. METHODS Sixteen homozygous FH subjects from eleven families were analyzed for mutations by Sanger sequencing. Large rearrangements in LDLR gene were evaluated by multiplex ligation probe dependent amplification (MLPA) technique. RESULTS Ten mutations were observed in LDLR gene, of which four mutations were novel. No mutation was detected in ApoB gene and common PCSK9 mutation (p.D374Y). Fourteen cases had homozygous mutations; one had compound heterozygous mutation, while no mutation was detected in one clinically homozygous case. We report an interesting "Triple hit" case with features of homozygous FH. CONCLUSIONS The spectrum of mutations in the Asian Indian population is quite heterogeneous. Of the mutations identified, 40% were novel. No mutation was observed in exons 3, 9 and 14 of LDLR gene, which are considered to be hot spots in studies done on Asian Indians in South Africa. Early detection followed by aggressive therapy, and cascade screening of extended families has been initiated to reduce the morbidity and mortality in these patients.
Collapse
Affiliation(s)
- Nitika Setia
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, 110060, India.
| | - Renu Saxena
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, 110060, India
| | - Anjali Arora
- Hyperlipidemia Prevention Clinic, Department of Cardiology, Sir Ganga Ram Hospital, New Delhi, 110060, India
| | - Ishwar C Verma
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, 110060, India
| |
Collapse
|
631
|
Hoe E, Hegele RA. Lipid Management in Diabetes with a Focus on Emerging Therapies. Can J Diabetes 2016; 39 Suppl 5:S183-90. [PMID: 26653256 DOI: 10.1016/j.jcjd.2015.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/07/2015] [Accepted: 09/17/2015] [Indexed: 11/18/2022]
Abstract
We reviewed the current and potential future management of dyslipidemia in patients with diabetes, with a focus on reduction of risk for macrovascular disease. We considered novel dyslipidemia therapies, in particular, inhibitors of proprotein convertase subtilisin kexin 9 (PCSK9), which have been approved in Canada for reducing low-density lipoprotein (LDL) cholesterol in certain patient groups. We searched for English-language randomized clinical trials (RCTs) of lipid-lowering modalities, mainly since 2012, that included patients with diabetes. The results from some RCTs may have future impacts on the approach to patients with diabetes. In particular, ezetimibe added to statins in the context of acute coronary syndromes seems to have particular benefits in patients with diabetes. Also, patients with diabetes show no differences, so far, from patients without diabetes with respect to efficacy of PCSK9 inhibitors in LDL cholesterol reduction and also in the frequency of adverse effects. RCTs of clinical outcomes with PCSK9 inhibitors performed exclusively in patients with diabetes are desirable, but approval of these agents for use in Canada has occurred before the availability of such results. Clinicians will have to gauge whether certain subjects with diabetes might benefit from this therapy, such as those with superimposed familial dyslipidemia, those with recurrent cardiovascular events and recalcitrant LDL cholesterol levels despite maximally tolerated statin therapy and those with high cardiovascular risk who cannot tolerate any dose of statins.
Collapse
MESH Headings
- Combined Modality Therapy/adverse effects
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/therapy
- Drug Therapy, Combination/adverse effects
- Drug Therapy, Combination/trends
- Drugs, Investigational/adverse effects
- Drugs, Investigational/therapeutic use
- Dyslipidemias/complications
- Dyslipidemias/prevention & control
- Humans
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use
- Hypolipidemic Agents/adverse effects
- Hypolipidemic Agents/therapeutic use
- Practice Guidelines as Topic
- Precision Medicine
- Proprotein Convertase 9
- Proprotein Convertases/antagonists & inhibitors
- Proprotein Convertases/metabolism
- Randomized Controlled Trials as Topic
- Serine Endopeptidases/metabolism
- Serine Proteinase Inhibitors/adverse effects
- Serine Proteinase Inhibitors/therapeutic use
Collapse
|
632
|
Abstract
Decreasing low-density lipoprotein cholesterol (LDL-C) is one of the few established and proven principles for the prevention and treatment of atherosclerosis. The higher the individual cardiovascular risk, the higher the benefit of lipid-lowering pharmacotherapy. Therefore, treatment options are chosen based on a patient's total cardiovascular risk. The latter depends not only on the levels of LDL-C but also on the presence of cardiovascular disease (CVD) and on the number and severity of other risk factors. Current guidelines recommend the lowering of LDL-C to 115 mg/dl (3 mmol/l) in patients with low and moderate risk. The LDL-C treatment target is <100 mg/dl (2.6 mmol/l) for patients at high risk and <70 mg/dl (1.8 mmol/l) for patients at very high risk. Although lifestyle measures remain a fundamental part of treatment, many patients require drug therapy to achieve their LDL-C targets. Statins are the drugs of choice, with other options including ezetimibe and the newly available monoclonal antibodies against PCSK9 (proprotein convertase subtilisin/kexin type 9). In some cases, bile acid-binding sequestrants and fibrates can also be considered. Nicotinic acid is no longer available in Germany. PCSK9 antibodies decrease LDL-C about 50-60 % and are well tolerated. Their effects on clinical endpoints are being investigated in large randomized trials. The aim of the present review is to summarize the current guidelines and treatment options for hypercholesterolemia. Moreover, we provide an appraisal of PCSK9 antibodies and propose their use in selected patient populations, particularly in those at very high cardiovascular risk whose LDL-C levels under maximally tolerated lipid-lowering therapy are significantly over their treatment target.
Collapse
|
633
|
Santos RD, Gidding SS, Hegele RA, Cuchel MA, Barter PJ, Watts GF, Baum SJ, Catapano AL, Chapman MJ, Defesche JC, Folco E, Freiberger T, Genest J, Hovingh GK, Harada-Shiba M, Humphries SE, Jackson AS, Mata P, Moriarty PM, Raal FJ, Al-Rasadi K, Ray KK, Reiner Z, Sijbrands EJG, Yamashita S. Defining severe familial hypercholesterolaemia and the implications for clinical management: a consensus statement from the International Atherosclerosis Society Severe Familial Hypercholesterolemia Panel. Lancet Diabetes Endocrinol 2016; 4:850-61. [PMID: 27246162 DOI: 10.1016/s2213-8587(16)30041-9] [Citation(s) in RCA: 310] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/24/2016] [Accepted: 04/06/2016] [Indexed: 12/26/2022]
Abstract
Familial hypercholesterolaemia is common in individuals who had a myocardial infarction at a young age. As many as one in 200 people could have heterozygous familial hypercholesterolaemia, and up to one in 300 000 individuals could be homozygous. The phenotypes of heterozygous and homozygous familial hypercholesterolaemia overlap considerably; the response to treatment is also heterogeneous. In this Review, we aim to define a phenotype for severe familial hypercholesterolaemia and identify people at highest risk for cardiovascular disease, based on the concentration of LDL cholesterol in blood and individuals' responsiveness to conventional lipid-lowering treatment. We assess the importance of molecular characterisation and define the role of other cardiovascular risk factors and advanced subclinical coronary atherosclerosis in risk stratification. Individuals with severe familial hypercholesterolaemia might benefit in particular from early and more aggressive cholesterol-lowering treatment (eg, with PCSK9 inhibitors). In addition to better tailored therapy, more precise characterisation of individuals with severe familial hypercholesterolaemia could improve resource use.
Collapse
Affiliation(s)
- Raul D Santos
- Lipid Clinic Heart Institute (InCor), University of São Paulo Medical School Hospital, and Preventive Medicine Centre and Cardiology Program, Hospital Israelita Albert Einstein, São Paulo, Brazil.
| | - Samuel S Gidding
- Nemours Cardiac Center, A I DuPont Hospital for Children, Wilmington, DE, USA
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine, Western University, London, ON, Canada
| | - Marina A Cuchel
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip J Barter
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Gerald F Watts
- Lipid Disorders Clinic, Royal Perth Hospital, The University of Western Australia, Perth, WA, Australia
| | - Seth J Baum
- Preventive Cardiology, Christine E Lynn Women's Health & Wellness Institute, Boca Raton Regional Hospital, Boca Raton, FL, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; IRCCS Multimedica, Milan, Italy
| | | | - Joep C Defesche
- University of Amsterdam, Academic Medical Center (AMC), Amsterdam, Netherlands
| | | | - Tomas Freiberger
- Molecular Genetics Lab, Centre for Cardiovascular Surgery and Transplantation, and Ceitec, Masaryk University, Brno, Czech Republic
| | - Jacques Genest
- McGill University Health Center, Royal Victoria Hospital, Montreal, QC, Canada
| | - G Kees Hovingh
- University of Amsterdam, Academic Medical Center (AMC), Amsterdam, Netherlands
| | - Mariko Harada-Shiba
- National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Steve E Humphries
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College of London, London, UK
| | - Ann S Jackson
- International Atherosclerosis Society, Houston, TX, USA
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Patrick M Moriarty
- Atherosclerosis and Lipoprotein-Apheresis Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Frederick J Raal
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Kausik K Ray
- School of Public Health, Imperial College London, London, UK
| | - Zelijko Reiner
- European Association for Cardiovascular Prevention and Rehabilitations, Zagreb, Croatia
| | | | | | | |
Collapse
|
634
|
Walsh MT, Hussain MM. Targeting microsomal triglyceride transfer protein and lipoprotein assembly to treat homozygous familial hypercholesterolemia. Crit Rev Clin Lab Sci 2016; 54:26-48. [PMID: 27690713 DOI: 10.1080/10408363.2016.1221883] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Homozygous familial hypercholesterolemia (HoFH) is a polygenic disease arising from defects in the clearance of plasma low-density lipoprotein (LDL), which results in extremely elevated plasma LDL cholesterol (LDL-C) and increased risk of atherosclerosis, coronary heart disease, and premature death. Conventional lipid-lowering therapies, such as statins and ezetimibe, are ineffective at lowering plasma cholesterol to safe levels in these patients. Other therapeutic options, such as LDL apheresis and liver transplantation, are inconvenient, costly, and not readily available. Recently, lomitapide was approved by the Federal Drug Administration as an adjunct therapy for the treatment of HoFH. Lomitapide inhibits microsomal triglyceride transfer protein (MTP), reduces lipoprotein assembly and secretion, and lowers plasma cholesterol levels by over 50%. Here, we explain the steps involved in lipoprotein assembly, summarize the role of MTP in lipoprotein assembly, explore the clinical and molecular basis of HoFH, and review pre-clinical studies and clinical trials with lomitapide and other MTP inhibitors for the treatment of HoFH. In addition, ongoing research and new approaches underway for better treatment modalities are discussed.
Collapse
Affiliation(s)
- Meghan T Walsh
- a School of Graduate Studies, Molecular and Cell Biology Program, State University of New York Downstate Medical Center , Brooklyn , NY , USA.,b Department of Cell Biology , State University of New York Downstate Medical Center , Brooklyn , NY , USA
| | - M Mahmood Hussain
- b Department of Cell Biology , State University of New York Downstate Medical Center , Brooklyn , NY , USA.,c Department of Pediatrics , SUNY Downstate Medical Center , Brooklyn , NY , USA.,d VA New York Harbor Healthcare System , Brooklyn , NY , USA , and.,e Winthrop University Hospital , Mineola , NY , USA
| |
Collapse
|
635
|
Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, Hoes AW, Jennings CS, Landmesser U, Pedersen TR, Reiner Ž, Riccardi G, Taskinen MR, Tokgozoglu L, Verschuren WM, Vlachopoulos C, Wood DA, Zamorano JL. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. Atherosclerosis 2016; 253:281-344. [DOI: 10.1016/j.atherosclerosis.2016.08.018] [Citation(s) in RCA: 562] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
636
|
Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res 2016; 57:1953-1975. [PMID: 27677946 DOI: 10.1194/jlr.r071233] [Citation(s) in RCA: 387] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Indexed: 12/24/2022] Open
Abstract
Human epidemiologic and genetic evidence using the Mendelian randomization approach in large-scale studies now strongly supports that elevated lipoprotein (a) [Lp(a)] is a causal risk factor for cardiovascular disease, that is, for myocardial infarction, atherosclerotic stenosis, and aortic valve stenosis. The Mendelian randomization approach used to infer causality is generally not affected by confounding and reverse causation, the major problems of observational epidemiology. This approach is particularly valuable to study causality of Lp(a), as single genetic variants exist that explain 27-28% of all variation in plasma Lp(a). The most important genetic variant likely is the kringle IV type 2 (KIV-2) copy number variant, as the apo(a) product of this variant influences fibrinolysis and thereby thrombosis, as opposed to the Lp(a) particle per se. We speculate that the physiological role of KIV-2 in Lp(a) could be through wound healing during childbirth, infections, and injury, a role that, in addition, could lead to more blood clots promoting stenosis of arteries and the aortic valve, and myocardial infarction. Randomized placebo-controlled trials of Lp(a) reduction in individuals with very high concentrations to reduce cardiovascular disease are awaited. Recent genetic evidence documents elevated Lp(a) as a cause of myocardial infarction, atherosclerotic stenosis, and aortic valve stenosis.
Collapse
Affiliation(s)
- Børge G Nordestgaard
- Department of Clinical Biochemistry and Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark; and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry and Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark; and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
637
|
Piťha J, Kovář J, Blahová T. Fasting and nonfasting triglycerides in cardiovascular and other diseases. Physiol Res 2016; 64:S323-30. [PMID: 26680665 DOI: 10.33549/physiolres.933196] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Moderately elevated plasma/serum triglycerides (2-10 mmol/l) signalize increased risk for cardiovascular disease or presence of non-alcoholic steatohepatitis. Extremely elevated triglycerides (more than 10 mmol/l) signalize increased risk for pancreatitis and lipemia retinalis. The concentration of triglycerides is regulated by many genetic and nongenetic factors. Extremely elevated triglycerides not provoked by nutritional factors, especially inappropriate alcohol intake are more likely to have a monogenic cause. On the contrary, mildly to moderately elevated triglycerides are often caused by polygenic disorders; these could be also associated with central obesity, insulin resistance, and diabetes mellitus. Concentration of triglycerides is also closely interconnected with presence of atherogenic remnant lipoproteins, impaired reverse cholesterol transport and more atherogenic small LDL particles. In general, there is tight association between triglycerides and many other metabolic factors including intermediate products of lipoprotein metabolism which are frequently atherogenic. Therefore, reliable evaluation of the independent role of triglycerides especially in atherosclerosis and cardiovascular disease is difficult. In individual cases values of HDL cholesterol, non-HDL cholesterol (total minus HDL cholesterol), non-HDL/nonLDL cholesterol (total minus HDL minus LDL cholesterol, especially in nonfasting status), atherogenic index of plasma and/or apolipoprotein B could help in decisions regarding aggressiveness of treatment.
Collapse
Affiliation(s)
- J Piťha
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | | |
Collapse
|
638
|
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant genetic disorder that clinically leads to increased low density lipoprotein-cholesterol (LDL-C) levels. As a consequence, FH patients are at high risk for cardiovascular disease (CVD). Mutations are found in genes coding for the LDLR, apoB, and PCSK9, although FH cannot be ruled out in the absence of a mutation in one of these genes. It is pivotal to diagnose FH at an early age, since lipid lowering results in a decreased risk of cardiovascular complications especially if initiated early, but unfortunately FH is largely underdiagnosed. While a number of clinical criteria are available, identification of a pathogenic mutation in any of the three aforementioned genes is seen by many as a way to establish a definitive diagnosis of FH. It should be remembered that clinical treatment is based on LDL-C levels and not solely on presence or absence of genetic mutations as LDL-C is what drives risk. Traditionally, mutation detection has been done by means of dideoxy sequencing. However, novel molecular testing methods are gradually being introduced. These next generation sequencing-based methods are likely to be applied on broader scale once their efficacy and effect on cost are being established. Statins are the first-line therapy of choice for FH patients as they have been proven to reduce CVD risk across a range of conditions including hypercholesterolemia (though not specifically tested in FH). However, in a significant proportion of FH patients LDL-C goals are not met, despite the use of maximal statin doses and additional lipid-lowering therapies. This underlines the need for additional therapies, and inhibition of PCSK9 and CETP is among the most promising new therapeutic options. In this review, we aim to provide an overview of the latest information about the definition, diagnosis, screening, and current and novel therapies for FH.
Collapse
Affiliation(s)
- Merel L Hartgers
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Kausik K Ray
- Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
639
|
Parizo J, Sarraju A, Knowles JW. Novel Therapies for Familial Hypercholesterolemia. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2016; 18:64. [PMID: 27620638 DOI: 10.1007/s11936-016-0486-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Both HeFH and HoFH require dietary and lifestyle modification. Pharmacotherapy of adult HeFH patients is largely driven by the American Heart Association (AHA) algorithm. A high-potency statin is started initially with a goal low-density lipoprotein cholesterol (LDL-C) reduction of >50 %. The LDL-C target is adjusted to <100 or <70 mg/dL in subjects with coronary artery disease (CAD) with ezetimibe being second line. If necessary, a third adjunctive therapy, such as a PSCK9 inhibitor (not yet approved in children) or bile acid-binding resin, can be added. Finally, LDL-C apheresis can be considered in patients with LDL-C >300 mg/dL (or >200 mg/dL with significant CAD, although now approved for LDL-C as low as 160 mg/dL with CAD). Due to the early, severe LDL-C elevation in HoFH patients, concerning natural history, rarity of the condition, and nuances of treatment, all HoFH patients should be treated at a pediatric or adult center with HoFH experience. LDL-C apheresis should be considered as early as 5 years of age. However, apheresis availability and tolerability is limited and pharmacotherapy is required. Generally, the AHA algorithm with reference to the European Atherosclerosis Society Consensus Panel recommendations is reasonable with all patients initiated on high-dose, high-potency statin, ezetimibe, and bile acid-binding resins. In most, additional LDL-C lowering is required with PCSK9 inhibitors and/or lomitapide or mipomersen. Liver transplantation can also be considered at experienced centers as a last resort.
Collapse
Affiliation(s)
- Justin Parizo
- Stanford University Medical Center, 300 Pasteur Ave, Stanford, CA, 94305, USA
| | - Ashish Sarraju
- Stanford University Medical Center, 300 Pasteur Ave, Stanford, CA, 94305, USA
| | - Joshua W Knowles
- Stanford University School of Medicine and Cardiovascular Institute, Falk CVRC, 300 Pasteur Drive, MC 5406, Stanford, CA, 94305, USA.
| |
Collapse
|
640
|
Double-heterozygous autosomal dominant hypercholesterolemia: Clinical characterization of an underreported disease. J Clin Lipidol 2016; 10:1462-1469. [PMID: 27919364 DOI: 10.1016/j.jacl.2016.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/25/2016] [Accepted: 09/05/2016] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Autosomal dominant hypercholesterolemia (ADH), characterized by high-plasma low-density lipoprotein cholesterol (LDL-C) levels and premature cardiovascular disease (CVD) risk, is caused by mutations in LDLR, APOB, and/or PCSK9. OBJECTIVE To describe the clinical characteristics of "double-heterozygous carriers," with 2 mutations in 2 different ADH causing genes, that is, LDLR and APOB or LDLR and PCSK9. METHODS Double heterozygotes were identified in the database of the national referral laboratory for DNA diagnostics of inherited dyslipidemias. We collected the medical data (comprising lipids and CVD events) from double heterozygotes and compared these with data from their heterozygous and unaffected relatives and homozygote/compound heterozygous LDLR mutation carriers, identified in a previously described cohort (n = 45). RESULTS A total of 28 double heterozygotes (23 LDLR/APOB and 5 LDLR/PCSK9 mutation carriers) were identified. Off treatment, LDL-C levels were significantly higher in double heterozygotes (mean ± SD, 8.4 ± 2.8 mmol/L) compared with 28 heterozygous (5.6 ± 2.2) and 18 unaffected relatives (2.5 ± 1.1; P ≤ .01 for all comparisons) and significantly lower compared with homozygous/compound heterozygous LDLR mutation carriers (13.0 ± 5.1; P < .001). CONCLUSIONS Double-heterozygous carriers of mutations in ADH genes express an intermediate phenotype compared with heterozygous and homozygous/compound heterozygous carriers and might well be misconceived to suffer from a severe form of heterozygous ADH. The molecular identification of double heterozygosity is of relevance from both a screening and an educational perspective.
Collapse
|
641
|
Ajufo E, Cuchel M. Recent Developments in Gene Therapy for Homozygous Familial Hypercholesterolemia. Curr Atheroscler Rep 2016; 18:22. [PMID: 26980316 DOI: 10.1007/s11883-016-0579-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Homozygous familial hypercholesterolemia (HoFH) is a life-threatening Mendelian disorder with a mean life expectancy of 33 years despite maximally tolerated standard lipid-lowering therapies. This disease is an ideal candidate for gene therapy, and in the last few years, a number of exciting developments have brought this approach closer to the clinic than ever before. In this review, we discuss in detail the most advanced of these developments, a recombinant adeno-associated virus (AAV) vector carrying a low-density lipoprotein receptor (LDLR) transgene which has recently entered phase 1/2a testing. We also review ongoing development of approaches to enhance transgene expression, improve the efficiency of hepatocyte transduction, and minimize the AAV capsid-specific adaptive immune response. We include a summary of key gene therapy approaches for HoFH in pre-clinical development, including RNA silencing of the gene encoding HMG-CoA reductase (HMGCR) and induced pluripotent stem cell transplant therapy.
Collapse
Affiliation(s)
- Ezim Ajufo
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marina Cuchel
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
642
|
Catapano AL, Lautsch D, Tokgözoglu L, Ferrieres J, Horack M, Farnier M, Toth PP, Brudi P, Tomassini JE, Ambegaonkar B, Gitt AK. Prevalence of potential familial hypercholesterolemia (FH) in 54,811 statin-treated patients in clinical practice. Atherosclerosis 2016; 252:1-8. [DOI: 10.1016/j.atherosclerosis.2016.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/20/2016] [Accepted: 07/07/2016] [Indexed: 10/21/2022]
|
643
|
Prell C, Koletzko B. Hyperlipidämien im Kindes- und Jugendalter. Monatsschr Kinderheilkd 2016. [DOI: 10.1007/s00112-016-0142-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
644
|
Affiliation(s)
- Anne C Goldberg
- From Washington University School of Medicine, St. Louis, MO (A.C.G.); and A. I. DuPont Hospital for Children, Wilmington, DE (S.S.G.)
| | - Samuel S Gidding
- From Washington University School of Medicine, St. Louis, MO (A.C.G.); and A. I. DuPont Hospital for Children, Wilmington, DE (S.S.G.).
| |
Collapse
|
645
|
Martinez M, Brodlie S, Griesemer A, Kato T, Harren P, Gordon B, Parker T, Levine D, Tyberg T, Starc T, Cho I, Min J, Elmore K, Lobritto S, Hudgins LC. Effects of Liver Transplantation on Lipids and Cardiovascular Disease in Children With Homozygous Familial Hypercholesterolemia. Am J Cardiol 2016; 118:504-10. [PMID: 27365335 DOI: 10.1016/j.amjcard.2016.05.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/12/2022]
Abstract
Homozygous familial hypercholesterolemia (HoFH) is a rare, inherited, life-threatening, metabolic disorder of low-density lipoprotein (LDL) receptor function characterized by elevated serum LDL cholesterol (LDL-C) and rapidly progressive atherosclerotic cardiovascular disease (ACVD). Since LDL receptors are predominantly found on hepatocytes, orthotopic liver transplantation (OLT) has emerged as a viable intervention for HoFH because LDL receptor activity is restored. This study assessed the effects of OLT on ACVD and ACVD risk factors in pediatric patients with HoFH. We analyzed lipids, lipoproteins, body mass index, glucose, blood pressure, and cardiovascular imaging in 8 pediatric patients who underwent OLT for HoFH. Total serum cholesterol, LDL-C, lipoprotein (a), and apolipoprotein B/apolipoprotein A1 ratio decreased to normal values in all subjects (p values <0.001) at 1 month after OLT and were maintained for the length of follow-up (2 to 6 years). There were few complications related to surgery or immunosuppressive therapy. Two patients developed mild hypertension. In the first 4 subjects monitored for 4 to 6 years after OLT, coronary artery disease did not develop or progress except in 1 minor artery in 1 subject and actually regressed in 2 subjects with >50% stenosis. However, aortic valve stenosis progressed in 2 of 4 subjects. In conclusion, OLT is an effective therapeutic option for patients with HoFH with coronary artery disease and persistently elevated serum LDL-C despite maximum medical therapy. Aortic valvular disease may progress. Long-term data are needed to evaluate the true risk-benefit ratio of this surgical approach.
Collapse
|
646
|
Pop GAM, de Boer MJ, Stalenhoef AF. Reducing cardiac after-load by lowering blood viscosity in patients with familial hypercholesterolemia - A pilot study. Possible mechanism for occurrence of anemia in chronic heart failure patients? COR ET VASA 2016. [DOI: 10.1016/j.crvasa.2015.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
647
|
Thedrez A, Sjouke B, Passard M, Prampart-Fauvet S, Guédon A, Croyal M, Dallinga-Thie G, Peter J, Blom D, Ciccarese M, Cefalù AB, Pisciotta L, Santos RD, Averna M, Raal F, Pintus P, Cossu M, Hovingh K, Lambert G. Proprotein Convertase Subtilisin Kexin Type 9 Inhibition for Autosomal Recessive Hypercholesterolemia-Brief Report. Arterioscler Thromb Vasc Biol 2016; 36:1647-50. [PMID: 27079874 DOI: 10.1161/atvbaha.116.307493] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/31/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitors lower low-density lipoprotein (LDL) cholesterol in the vast majority of patients with autosomal dominant familial hypercholesterolemia. Will PCSK9 inhibition with monoclonal antibodies, in particular alirocumab, be of therapeutic value for patients with autosomal recessive hypercholesterolemia (ARH)? APPROACH AND RESULTS Primary lymphocytes were obtained from 28 genetically characterized ARH patients and 11 controls. ARH lymphocytes treated with mevastatin were incubated with increasing doses of recombinant PCSK9 with or without saturating concentrations of alirocumab. Cell surface LDL receptor expression measured by flow cytometry and confocal microscopy was higher in ARH than in control lymphocytes. PCSK9 significantly reduced LDL receptor expression in ARH lymphocytes albeit to a lower extent than in control lymphocytes (25% versus 76%, respectively), an effect reversed by alirocumab. Fluorescent LDL cellular uptake, also measured by flow cytometry, was reduced in ARH lymphocytes compared with control lymphocytes. PCSK9 significantly lowered LDL cellular uptake in ARH lymphocytes, on average by 18%, compared with a 46% reduction observed in control lymphocytes, an effect also reversed by alirocumab. Overall, the effects of recombinant PCSK9, and hence of alirocumab, on LDL receptor expression and function were significantly less pronounced in ARH than in control cells. CONCLUSIONS PCSK9 inhibition with alirocumab on top of statin treatment has the potential to lower LDL cholesterol in some autosomal recessive hypercholesterolemia patients.
Collapse
Affiliation(s)
- Aurélie Thedrez
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Barbara Sjouke
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Maxime Passard
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Simon Prampart-Fauvet
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Alexis Guédon
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Mikael Croyal
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Geesje Dallinga-Thie
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Jorge Peter
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Dirk Blom
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Milco Ciccarese
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Angelo B Cefalù
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Livia Pisciotta
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Raul D Santos
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Maurizio Averna
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Frederick Raal
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Paolo Pintus
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Maria Cossu
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Kees Hovingh
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| | - Gilles Lambert
- From the Inra UMR 1280, Université de Nantes, Faculté de Médecine, Nantes, France (A.T., M.P., S.P.-F., A.G., M.C., G.L.); Department of Vascular Medicine, Academic Medical Centre, Amsterdam, The Netherlands (B.S., G.D.-T., J.P., K.H.); Lipidology Division of Internal Medicine, University of Cape Town, Cape Town, South Africa (D.B.); Dipartimiento di Nefrologia Dialisi e Trapianto, SS Annunziata Hospital, Sassari, Italy (M.C., M.C.); University of Palermo, School of Medicine, Palermo, Italy (A.B.C., M.A.); University of Genoa, Genoa, Italy (L.P.); Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medica School Hospital, Sao Paulo, Brazil (R.D.S.); Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa (F.R.); Dipartimento di Medicina Interna, Brotzu Hospital, Cagliari, Italy (P.P.); Inserm UMR 1188, Sainte Clotilde, France (G.L.); Université de la Réunion, Faculté de Médecine, Saint Denis de la Réunion, France (G.L.); and CHU de la Réunion, Saint-Denis de la Réunion, France (G.L.)
| |
Collapse
|
648
|
Ellis KL, Hooper AJ, Burnett JR, Watts GF. Progress in the care of common inherited atherogenic disorders of apolipoprotein B metabolism. Nat Rev Endocrinol 2016; 12:467-84. [PMID: 27199287 DOI: 10.1038/nrendo.2016.69] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Familial hypercholesterolaemia, familial combined hyperlipidaemia (FCH) and elevated lipoprotein(a) are common, inherited disorders of apolipoprotein B metabolism that markedly accelerate the onset of atherosclerotic cardiovascular disease (ASCVD). These disorders are frequently encountered in clinical lipidology and need to be accurately identified and treated in both index patients and their family members, to prevent the development of premature ASCVD. The optimal screening strategies depend on the patterns of heritability for each condition. Established therapies are widely used along with lifestyle interventions to regulate levels of circulating lipoproteins. New therapeutic strategies are becoming available, and could supplement traditional approaches in the most severe cases, but their long-term cost-effectiveness and safety have yet to be confirmed. We review contemporary developments in the understanding, detection and care of these highly atherogenic disorders of apolipoprotein B metabolism.
Collapse
Affiliation(s)
- Katrina L Ellis
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- Centre for Genetic Origins of Health and Disease, The University of Western Australia and Curtin University, 35 Stirling Highway, Crawley, Perth, Western Australia 6009, Australia
| | - Amanda J Hooper
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- PathWest Laboratory Medicine WA, Royal Perth Hospital and Fiona Stanley Hospital Network, Perth, Western Australia, Australia
- School of Pathology and Laboratory Medicine, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, Western Australia 6009, Australia
| | - John R Burnett
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- PathWest Laboratory Medicine WA, Royal Perth Hospital and Fiona Stanley Hospital Network, Perth, Western Australia, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Wellington Street Perth, Western Australia, Australia
| | - Gerald F Watts
- School of Medicine and Pharmacology, The University of Western Australia, PO Box X2213, Perth, Western Australia 6847, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Wellington Street Perth, Western Australia, Australia
| |
Collapse
|
649
|
Mundal L, Retterstøl K. A systematic review of current studies in patients with familial hypercholesterolemia by use of national familial hypercholesterolemia registries. Curr Opin Lipidol 2016; 27:388-97. [PMID: 27070076 DOI: 10.1097/mol.0000000000000300] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW More than 25 million are expected to have familial hypercholesterolemia worldwide. The risk of cardiovascular disease (CVD) in familial hypercholesterolemia is not clear and register studies represent a valuable tool to get such data, which will be discussed in the present paper. RECENT FINDING A systematic review of current familial hypercholesterolemia studies, by use of National familial hypercholesterolemia registries was performed from 1980 to 2016. This review shows that familial hypercholesterolemia patients have a high prevalence of CVD also in the time period after statins became available. The patient group does not reach recommended target values for lipids, and have a significantly higher CVD mortality as compared with the general population according to age and sex. SUMMARY The review underscores the importance of establishing National familial hypercholesterolemia registries with complete datasets on familial hypercholesterolemia patients to improve early diagnostics, therapeutics and long-term follow-up to prevent the premature CVD events and deaths in this patient group.
Collapse
Affiliation(s)
- Liv Mundal
- aThe Lipid Clinic, Oslo University Hospital bDepartment of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | |
Collapse
|
650
|
Yahya R, Favari E, Calabresi L, Verhoeven A, Zimetti F, Adorni M, Gomaraschi M, Averna M, Cefalù A, Bernini F, Sijbrands E, Mulder M, Roeters van Lennep J. Lomitapide affects HDL composition and function. Atherosclerosis 2016; 251:15-18. [DOI: 10.1016/j.atherosclerosis.2016.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/23/2022]
|