51
|
Abbasi S, Khan A, Choudhry MW. New Insights Into the Treatment of Hyperlipidemia: Pharmacological Updates and Emerging Treatments. Cureus 2024; 16:e63078. [PMID: 38919858 PMCID: PMC11196920 DOI: 10.7759/cureus.63078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 06/27/2024] Open
Abstract
Cardiovascular diseases are the leading causes of global mortality and morbidity. Hyperlipidemia is a significant risk factor for atherosclerosis and subsequent cardiovascular diseases. Hyperlipidemia is characterized by imbalances in blood cholesterol levels, particularly elevated low-density lipoprotein cholesterol and triglycerides, and is influenced by genetic and environmental factors. Current management consists of lifestyle modifications and pharmacological interventions most commonly consisting of statins. This review paper explores pathophysiology, management strategies, and pharmacotherapies including commonly used well-established medications including statins, fibrates, and ezetimibe, exciting novel therapies including proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, and RNA interference therapies (inclisiran), lomitapide, and bempedoic acid, highlighting their mechanisms of action, clinical efficacy, and safety profiles. Additionally, emerging therapies under clinical trials including ApoC-III inhibitors, DGAT2 inhibitors, ACAT2 Inhibitors, and LPL gene therapies are examined for their potential to improve lipid homeostasis and cardiovascular outcomes. The evolving landscape of hyperlipidemia management underscores the importance of continued research into both established therapies and promising new candidates, offering hope for more effective treatment strategies in the future.
Collapse
Affiliation(s)
| | - Adnan Khan
- Cardiology, St. Joseph's Medical Center, Stockton, USA
| | | |
Collapse
|
52
|
Natale F, Franzese R, Marotta L, Mollo N, Solimene A, Luisi E, Gentile C, Loffredo FS, Golino P, Cimmino G. Evolving Concepts of the SCORE System: Subtracting Cholesterol from Risk Estimation: A Way for a Healthy Longevity? Life (Basel) 2024; 14:679. [PMID: 38929662 PMCID: PMC11204887 DOI: 10.3390/life14060679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
The role of cholesterol, mainly low-density lipoproteins (LDL-C), as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) is now established and accepted by the international scientific community. Based on this evidence, the European and American guidelines recommend early risk stratification and "rapid" achievement of the suggested target according to the risk estimation to reduce the number of major cardiovascular events. Prolonged exposure over the years to high levels of LDL-C is one of the determining factors in the development and progression of atherosclerotic plaque, on which the action of conventional risk factors (cigarette smoking, excess weight, sedentary lifestyle, arterial hypertension, diabetes mellitus) as well as non-conventional risk factors (gut microbiota, hyperuricemia, inflammation), alone or in combination, favors the destabilization of the atherosclerotic lesion with rupture/fissuration/ulceration and consequent formation of intravascular thrombosis, which leads to the acute clinical manifestations of acute coronary syndromes. In the current clinical practice, there is a growing number of cases that, although extremely common, are emblematic of the concept of long-term exposure to the risk factor (LDL hypercholesterolemia), which, not adequately controlled and in combination with other risk factors, has favored the onset of major cardiovascular events. The triple concept of "go lower, start earlier and keep longer!" should be applied in current clinical practice at any level of prevention. In the present manuscript, we will review the current evidence and documents supporting the causal role of LDL-C in determining ASCVD and whether it is time to remove it from any score.
Collapse
Affiliation(s)
- Francesco Natale
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
| | - Rosa Franzese
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Luigi Marotta
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Noemi Mollo
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Achille Solimene
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Ettore Luisi
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Carmine Gentile
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Francesco S. Loffredo
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Paolo Golino
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy; (F.N.); (R.F.); (L.M.); (N.M.); (A.S.); (E.L.); (C.G.); (F.S.L.); (P.G.)
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
- Cardiology Unit, AOU Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
53
|
Eisenberg S, Gold D, Mehta PK, Leshnower B, Sperling LS. Supravalvular Aortic Stenosis in Homozygous Familial Hypercholesterolemia: Contemporary Management. JACC Case Rep 2024; 29:102342. [PMID: 38682003 PMCID: PMC11047779 DOI: 10.1016/j.jaccas.2024.102342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024]
Abstract
We report a case of a patient diagnosed with homozygous familial hypercholesterolemia and progressive supravalvular aortic stenosis. Treatment with long-term low-density lipoprotein apheresis and management with novel lipid-lowering agents including an angiopoetin-like protein inhibitor led to significant low-density lipoprotein reduction. The case highlights the challenges in managing the manifestations of homozygous familial hypercholesterolemia.
Collapse
Affiliation(s)
| | - Daniel Gold
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Puja K. Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bradley Leshnower
- Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Laurence S. Sperling
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
54
|
Muzurović E, Borozan S, Rizzo M. Clinical impact of genetic testing for lipid disorders. Curr Opin Cardiol 2024; 39:154-161. [PMID: 38456469 DOI: 10.1097/hco.0000000000001133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW Genetic testing is increasingly becoming a common consideration in the clinical approach of dyslipidemia patients. Advances in research in last decade and increased recognition of genetics in biological pathways modulating blood lipid levels created a gap between theoretical knowledge and its applicability in clinical practice. Therefore, it is very important to define the clinical justification of genetic testing in dyslipidemia patients. RECENT FINDINGS Clinical indications for genetic testing for most dyslipidemias are not precisely defined and there are no clearly established guideline recommendations. In patients with severe low-density lipoprotein cholesterol (LDL-C) levels, the genetic analysis can be used to guide diagnostic and therapeutic approach, while in severe hypertriglyceridemia (HTG), clinicians can rely on triglyceride level rather than a genotype along the treatment pathway. Genetic testing increases diagnostic accuracy and risk stratification, access and adherence to specialty therapies, and cost-effectiveness of cascade testing. A shared decision-making model between the provider and the patient is essential as patient values, preferences and clinical characteristics play a very strong role. SUMMARY Genetic testing for lipid disorders is currently underutilized in clinical practice. However, it should be selectively used, according to the type of dyslipidemia and when the benefits overcome costs.
Collapse
Affiliation(s)
- Emir Muzurović
- Department of Internal Medicine, Endocrinology Section, Clinical Centre of Montenegro
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Sanja Borozan
- Department of Internal Medicine, Endocrinology Section, Clinical Centre of Montenegro
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
55
|
Ates I, Stuart C, Rathbone T, Barzi M, He G, Major AM, Shankar V, Lyman RA, Angner SS, Mackay TFC, Srinivasan S, Farris AB, Bissig KD, Cottle RN. Ex vivo gene editing and cell therapy for hereditary tyrosinemia type 1. Hepatol Commun 2024; 8:e0424. [PMID: 38668730 DOI: 10.1097/hc9.0000000000000424] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/13/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND We previously demonstrated the successful use of in vivo CRISPR gene editing to delete 4-hydroxyphenylpyruvate dioxygenase (HPD) to rescue mice deficient in fumarylacetoacetate hydrolase (FAH), a disorder known as hereditary tyrosinemia type 1 (HT1). The aim of this study was to develop an ex vivo gene-editing protocol and apply it as a cell therapy for HT1. METHODS We isolated hepatocytes from wild-type (C57BL/6J) and Fah-/- mice and then used an optimized electroporation protocol to deliver Hpd-targeting CRISPR-Cas9 ribonucleoproteins into hepatocytes. Next, hepatocytes were transiently incubated in cytokine recovery media formulated to block apoptosis, followed by splenic injection into recipient Fah-/- mice. RESULTS We observed robust engraftment and expansion of transplanted gene-edited hepatocytes from wild-type donors in the livers of recipient mice when transient incubation with our cytokine recovery media was used after electroporation and negligible engraftment without the media (mean: 46.8% and 0.83%, respectively; p=0.0025). Thus, the cytokine recovery medium was critical to our electroporation protocol. When hepatocytes from Fah-/- mice were used as donors for transplantation, we observed 35% and 28% engraftment for Hpd-Cas9 ribonucleoproteins and Cas9 mRNA, respectively. Tyrosine, phenylalanine, and biochemical markers of liver injury normalized in both Hpd-targeting Cas9 ribonucleoprotein and mRNA groups independent of induced inhibition of Hpd through nitisinone, indicating correction of disease indicators in Fah-/- mice. CONCLUSIONS The successful liver cell therapy for HT1 validates our protocol and, despite the known growth advantage of HT1, showcases ex vivo gene editing using electroporation in combination with liver cell therapy to cure a disease model. These advancements underscore the potential impacts of electroporation combined with transplantation as a cell therapy.
Collapse
Affiliation(s)
- Ilayda Ates
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Callie Stuart
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Tanner Rathbone
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Mercedes Barzi
- Department of Pediatrics, Division of Medical Genetics, Alice and Y.T. Chen Center for Genetics and Genomics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gordon He
- Department of Pediatrics, Division of Medical Genetics, Alice and Y.T. Chen Center for Genetics and Genomics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Angela M Major
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Vijay Shankar
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Rachel A Lyman
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Sidney S Angner
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Trudy F C Mackay
- Department of Biochemistry and Genetics, Clemson University, Clemson, South Carolina, USA
- Center for Human Genetics, Clemson University, Greenwood, South Carolina, USA
| | - Shanthi Srinivasan
- Department of Medicine, Digestive Diseases Division, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alton Brad Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Karl-Dimiter Bissig
- Department of Pediatrics, Division of Medical Genetics, Alice and Y.T. Chen Center for Genetics and Genomics, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biomedical Engineering (BME) at the Duke University Pratt School of Engineering, Durham, North Carolina, USA
- Duke Cancer Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Renee N Cottle
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
56
|
Luo F, Das A, Khetarpal SA, Fang Z, Zelniker TA, Rosenson RS, Qamar A. ANGPTL3 inhibition, dyslipidemia, and cardiovascular diseases. Trends Cardiovasc Med 2024; 34:215-222. [PMID: 36746257 DOI: 10.1016/j.tcm.2023.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
Optimal management of low-density lipoprotein cholesterol (LDL-C) is a central tenet in the primary and secondary prevention of atherosclerotic cardiovascular disease (ASCVD). However, significant residual cardiovascular risk remains despite achieving guideline-directed LDL-C levels, in part due to mixed hyperlipidemia with elevated fasting and non-fasting triglyceride-rich lipoprotein levels. Advances in human genetics have identified angiopoietin-like 3 (ANGPTL3) as a promising therapeutic target to lower cardiovascular risk. Evidence accrued from genetic epidemiological studies demonstrate that ANGPTL3 loss of function is strongly associated with lowering of circulating LDL-C, triglyceride-rich lipoproteins and concurrent risk reduction in development of coronary artery disease. Pharmacological inhibition of ANGPTL3 with monoclonal antibodies, antisense oligonucleotides and gene editing are in development with early studies showing their safety and efficacy in lowering in both, LDL-C and TGs, circumventing a key limitation of previous therapies. Monoclonal antibodies targeting ANGPTL3 are approved for clinical use in homozygous familial hypercholesteremia in USA and Europe. Although promising, future studies focusing on long-term beneficial effect in reducing cardiovascular events with inhibition of ANGPTL3 are warranted.
Collapse
Affiliation(s)
- Fei Luo
- Department of Cardiovascular Medicine, Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Avash Das
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sumeet A Khetarpal
- Division of Cardiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Zhenfei Fang
- Department of Cardiovascular Medicine, Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Thomas A Zelniker
- Division of Cardiology, Vienna General Hospital and Medical University of Vienna, Austria
| | - Robert S Rosenson
- Metabolism and Lipids Unit, Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R Kravis Center for Cardiovascular Health, Mount Sinai Icahn School of Medicine, New York, NY, United States
| | - Arman Qamar
- Section of Interventional Cardiology & Vascular Medicine, NorthShore University Health System, University of Chicago Pritzker School of Medicine, 2650 Ridge Avenue, Evanston, IL, United States.
| |
Collapse
|
57
|
Reijman MD, Kusters DM, Groothoff JW, Arbeiter K, Dann EJ, de Boer LM, de Ferranti SD, Gallo A, Greber-Platzer S, Hartz J, Hudgins LC, Ibarretxe D, Kayikcioglu M, Klingel R, Kolovou GD, Oh J, Planken RN, Stefanutti C, Taylan C, Wiegman A, Schmitt CP. Clinical practice recommendations on lipoprotein apheresis for children with homozygous familial hypercholesterolaemia: An expert consensus statement from ERKNet and ESPN. Atherosclerosis 2024; 392:117525. [PMID: 38598969 DOI: 10.1016/j.atherosclerosis.2024.117525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Homozygous familial hypercholesterolaemia is a life-threatening genetic condition, which causes extremely elevated LDL-C levels and atherosclerotic cardiovascular disease very early in life. It is vital to start effective lipid-lowering treatment from diagnosis onwards. Even with dietary and current multimodal pharmaceutical lipid-lowering therapies, LDL-C treatment goals cannot be achieved in many children. Lipoprotein apheresis is an extracorporeal lipid-lowering treatment, which is used for decades, lowering serum LDL-C levels by more than 70% directly after the treatment. Data on the use of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia mainly consists of case-reports and case-series, precluding strong evidence-based guidelines. We present a consensus statement on lipoprotein apheresis in children based on the current available evidence and opinions from experts in lipoprotein apheresis from over the world. It comprises practical statements regarding the indication, methods, treatment goals and follow-up of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia and on the role of lipoprotein(a) and liver transplantation.
Collapse
Affiliation(s)
- M Doortje Reijman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - D Meeike Kusters
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jaap W Groothoff
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Klaus Arbeiter
- Division of Paediatric Nephrology and Gastroenterology, Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Eldad J Dann
- Blood Bank and Apheresis Unit Rambam Health Care Campus, Haifa, Israel
| | - Lotte M de Boer
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - Sarah D de Ferranti
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Antonio Gallo
- Sorbonne Université, INSERM, UMR 1166, Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpêtrière, F-75013, Paris, France
| | - Susanne Greber-Platzer
- Clinical Division of Paediatric Pulmonology, Allergology and Endocrinology, Department of Paediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Jacob Hartz
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Lisa C Hudgins
- The Rogosin Institute, Weill Cornell Medical College, New York, NY, USA
| | - Daiana Ibarretxe
- Vascular Medicine and Metabolism Unit (UVASMET), Hospital Universitari Sant Joan, Spain; Universitat Rovira i Virgili, Spain; Institut Investigació Sanitària Pere Virgili (IISPV)-CERCA, Spain; Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Meral Kayikcioglu
- Department of Cardiology, Medical Faculty, Ege University, 35100, Izmir, Turkey
| | - Reinhard Klingel
- Apheresis Research Institute, Stadtwaldguertel 77, 50935, Cologne, Germany(†)
| | - Genovefa D Kolovou
- Metropolitan Hospital, Department of Preventive Cardiology, 9, Ethn. Makariou & 1, El. Venizelou, N. Faliro, 185 47, Athens, Greece
| | - Jun Oh
- University Medical Center Hamburg/Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - R Nils Planken
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location AMC, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands
| | - Claudia Stefanutti
- Department of Molecular Medicine, Lipid Clinic and Atherosclerosis Prevention Centre, 'Umberto I' Hospital 'Sapienza' University of Rome, I-00161, Rome, Italy
| | - Christina Taylan
- Paediatric Nephrology, Children's and Adolescents' Hospital, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Albert Wiegman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Claus Peter Schmitt
- Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, Germany
| |
Collapse
|
58
|
Gu J, Epland M, Ma X, Park J, Sanchez RJ, Li Y. A machine-learning algorithm using claims data to identify patients with homozygous familial hypercholesterolemia. Sci Rep 2024; 14:8890. [PMID: 38632285 PMCID: PMC11024086 DOI: 10.1038/s41598-024-58719-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
Homozygous familial hypercholesterolemia (HoFH) is an underdiagnosed and undertreated ultra-rare disease. We utilized claims data from the Komodo Healthcare Map database to develop a machine-learning model to identify potential HoFH patients. We tokenized patients enrolled in MyRARE (patient support program for those prescribed evinacumab-dgnb in the United States) and linked them with their Komodo claims. A true positive HoFH cohort (n = 331) was formed by including patients from MyRARE and patients with prescriptions for evinacumab-dgnb or lomitapide. The negative cohort (n = 1423) comprised patients with or at risk for cardiovascular disease. We divided the cohort into an 80% training and 20% testing set. Overall, 10,616 candidate features were investigated; 87 were selected due to clinical relevance and importance on prediction performance. Different machine-learning algorithms were explored, with fast interpretable greedy-tree sums selected as the final machine-learning tool. This selection was based on its satisfactory performance and its easily interpretable nature. The model identified four useful features and yielded precision (positive predicted value) of 0.98, recall (sensitivity) of 0.88, area under the receiver operating characteristic curve of 0.98, and accuracy of 0.97. The model performed well in identifying HoFH patients in the testing set, providing a useful tool to facilitate HoFH screening and diagnosis via healthcare claims data.
Collapse
Affiliation(s)
- Jing Gu
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York, NY, 10591, USA
| | | | | | | | - Robert J Sanchez
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York, NY, 10591, USA.
| | - Ying Li
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York, NY, 10591, USA
| |
Collapse
|
59
|
Buganza R, Massini G, Di Taranto MD, Cardiero G, de Sanctis L, Guardamagna O. Simplified Criteria for Identification of Familial Hypercholesterolemia in Children: Application in Real Life. J Cardiovasc Dev Dis 2024; 11:123. [PMID: 38667741 PMCID: PMC11050898 DOI: 10.3390/jcdd11040123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The diagnosis of familial hypercholesterolemia (FH) in children is primarily based on main criteria including low-density lipoprotein cholesterol (LDL-C) levels, increased in the proband and relatives, and its inheritance. Two other relevant parameters are symptoms, rarely occurring in children, as rare are the FH homozygous patients, and the mutation detection of related genes. The latter allows the final diagnosis, although it is not commonly available. Moreover, the application of diagnostic scores, useful in adults, is poorly applied in children. The aim of this study was to compare the reliability of criteria here applied with different scores, apart from genetic analysis, for FH diagnosis. The latter was then confirmed by genetic analysis. METHODS n. 180 hypercholesterolemic children (age 10.2 ± 4.6 years) showing LDL-C levels ≥95th percentile (age- and sex-related), the dominant inheritance pattern of hypercholesterolemia (including LDL-C ≥95th percentile in one parent), were considered potentially affected by FH and included in the study. The molecular analysis of the LDLR, APOB and PCSK9 genes was applied to verify the diagnostic accuracy. Biochemical and family history data were also retrospectively categorized according to European Atherosclerosis Society (EAS), Simon Broome Register (SBR), Pediatric group of the Italian LIPIGEN (LIPIGEN-FH-PED) and Dutch Lipid Clinic Network (DLCN) criteria. Detailed kindred biochemical and clinical assessments were extended to three generations. The lipid profile was detected by standard laboratory kits, and gene analysis was performed by traditional sequencing or Next-Generation Sequencing (NGS). RESULTS Among 180 hypercholesterolemic subjects, FH suspected based on the above criteria, 164/180 had the diagnosis confirmed, showing causative mutations. The mutation detection rate (MDR) was 91.1%. The scoring criteria proposed by the EAS, SBR and LIPIGEN-FH-PED (resulting in high probable, possible-defined and probable-defined, respectively) showed high sensitivity (~90%), low specificity (~6%) and high MDR (~91%). It is noteworthy that their application, as a discriminant for the execution of the molecular investigation, would lead to a loss of 9.1%, 9.8% and 9.1%, respectively, of FH-affected patients, as confirmed by the genetic analysis. DLCN criteria, for which LDL-C cut-offs are not specific for childhood, would lead to a loss of 53% of patients with mutations. CONCLUSIONS In the pediatric population, the combination of LDL-C ≥95th percentile in the proband and the dominant inheritance pattern of hypercholesterolemia, with LDL-C ≥95th percentile in one parent, is a simple, useful and effective diagnostic criterion, showing high MDR. This pattern is crucial for early FH diagnosis. EAS, SBR and LIPIGEN-FH-PED criteria can underestimate the real number of patients with gene mutations and cannot be considered strictly discriminant for the execution of molecular analysis.
Collapse
Affiliation(s)
- Raffaele Buganza
- Department of Public Health and Pediatric Sciences, University of Torino, 10133 Torino, Italy; (G.M.); (L.d.S.); (O.G.)
- Pediatric Endocrinology, Ospedale Infantile Regina Margherita, 10126 Torino, Italy
| | - Giulia Massini
- Department of Public Health and Pediatric Sciences, University of Torino, 10133 Torino, Italy; (G.M.); (L.d.S.); (O.G.)
- Pediatric Endocrinology, Ospedale Infantile Regina Margherita, 10126 Torino, Italy
| | - Maria Donata Di Taranto
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (M.D.D.T.); (G.C.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Giovanna Cardiero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (M.D.D.T.); (G.C.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Luisa de Sanctis
- Department of Public Health and Pediatric Sciences, University of Torino, 10133 Torino, Italy; (G.M.); (L.d.S.); (O.G.)
- Pediatric Endocrinology, Ospedale Infantile Regina Margherita, 10126 Torino, Italy
| | - Ornella Guardamagna
- Department of Public Health and Pediatric Sciences, University of Torino, 10133 Torino, Italy; (G.M.); (L.d.S.); (O.G.)
- Pediatric Endocrinology, Ospedale Infantile Regina Margherita, 10126 Torino, Italy
| |
Collapse
|
60
|
Cardiero G, Ferrandino M, Calcaterra IL, Iannuzzo G, Di Minno MND, Buganza R, Guardamagna O, Auricchio R, Di Taranto MD, Fortunato G. Impact of 12-SNP and 6-SNP Polygenic Scores on Predisposition to High LDL-Cholesterol Levels in Patients with Familial Hypercholesterolemia. Genes (Basel) 2024; 15:462. [PMID: 38674396 PMCID: PMC11050365 DOI: 10.3390/genes15040462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Familial hypercholesterolemia (FH) comprises high LDL-cholesterol (LDL-c) levels and high cardiovascular disease risk. In the absence of pathogenic variants in causative genes, a polygenic basis was hypothesized. METHODS In a population of 418 patients (excluding homozygotes) with clinical suspicion of FH, the FH-causative genes and the regions of single nucleotide polymorphisms (SNPs) included in 12-SNP and 6-SNP scores were sequenced by next-generation sequencing, allowing for the detection of pathogenic variants (V+) in 220 patients. To make a comparison, only patients without uncertain significance variants (V-/USV-) were considered (n = 162). RESULTS Higher values of both scores were observed in V+ than in V-. Considering a cut-off leading to 80% of V-/USV- as score-positive, a lower prevalence of patients positive for both 12-SNP and 6-SNP scores was observed in V+ (p = 0.010 and 0.033, respectively). Mainly for the 12-SNP score, among V+ patients, higher LDL-c levels were observed in score-positive (223 mg/dL -IQR 187-279) than in negative patients (212 mg/dL -IQR 162-240; p = 0.006). Multivariate analysis confirmed the association of scores and LDL-c levels independently of age, sex, and presence of pathogenic variants and revealed a greater association in children. CONCLUSIONS The 12-SNP and 6-SNP polygenic scores could explain hypercholesterolemia in patients without pathogenic variants as well as the variability of LDL-c levels among patients with FH-causative variants.
Collapse
Affiliation(s)
- Giovanna Cardiero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (G.C.); (M.F.); (G.F.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Martina Ferrandino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (G.C.); (M.F.); (G.F.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Ilenia Lorenza Calcaterra
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (I.L.C.); (G.I.); (M.N.D.D.M.)
| | - Gabriella Iannuzzo
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (I.L.C.); (G.I.); (M.N.D.D.M.)
| | - Matteo Nicola Dario Di Minno
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (I.L.C.); (G.I.); (M.N.D.D.M.)
| | - Raffaele Buganza
- Dipartimento di Scienze della Sanità Pubblica e Pediatriche, Università di Torino, 10126 Turin, Italy; (R.B.); (O.G.)
| | - Ornella Guardamagna
- Dipartimento di Scienze della Sanità Pubblica e Pediatriche, Università di Torino, 10126 Turin, Italy; (R.B.); (O.G.)
| | - Renata Auricchio
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, 80131 Naples, Italy;
| | - Maria Donata Di Taranto
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (G.C.); (M.F.); (G.F.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Giuliana Fortunato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (G.C.); (M.F.); (G.F.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| |
Collapse
|
61
|
Mulder JWCM, Tromp TR, Al-Khnifsawi M, Blom DJ, Chlebus K, Cuchel M, D’Erasmo L, Gallo A, Hovingh GK, Kim NT, Long J, Raal FJ, Schonck WAM, Soran H, Truong TH, Boersma E, Roeters van Lennep JE. Sex Differences in Diagnosis, Treatment, and Cardiovascular Outcomes in Homozygous Familial Hypercholesterolemia. JAMA Cardiol 2024; 9:313-322. [PMID: 38353972 PMCID: PMC10867777 DOI: 10.1001/jamacardio.2023.5597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/15/2023] [Indexed: 02/17/2024]
Abstract
Importance Homozygous familial hypercholesterolemia (HoFH) is a rare genetic condition characterized by extremely increased low-density lipoprotein (LDL) cholesterol levels and premature atherosclerotic cardiovascular disease (ASCVD). Heterozygous familial hypercholesterolemia (HeFH) is more common than HoFH, and women with HeFH are diagnosed later and undertreated compared to men; it is unknown whether these sex differences also apply to HoFH. Objective To investigate sex differences in age at diagnosis, risk factors, lipid-lowering treatment, and ASCVD morbidity and mortality in patients with HoFH. Design, Setting, and Participants Sex-specific analyses for this retrospective cohort study were performed using data from the HoFH International Clinical Collaborators (HICC) registry, the largest global dataset of patients with HoFH, spanning 88 institutions across 38 countries. Patients with HoFH who were alive during or after 2010 were eligible for inclusion. Data entry occurred between February 2016 and December 2020. Data were analyzed from June 2022 to June 2023. Main Outcomes and Measures Comparison between women and men with HoFH regarding age at diagnosis, presence of risk factors, lipid-lowering treatment, prevalence, and onset and incidence of ASCVD morbidity (myocardial infarction [MI], aortic stenosis, and combined ASCVD outcomes) and mortality. Results Data from 389 women and 362 men with HoFH from 38 countries were included. Women and men had similar age at diagnosis (median [IQR], 13 [6-26] years vs 11 [5-27] years, respectively), untreated LDL cholesterol levels (mean [SD], 579 [203] vs 596 [186] mg/dL, respectively), and cardiovascular risk factor prevalence, except smoking (38 of 266 women [14.3%] vs 59 of 217 men [27.2%], respectively). Prevalence of MI was lower in women (31 of 389 [8.0%]) than men (59 of 362 [16.3%]), but age at first MI was similar (mean [SD], 39 [13] years in women vs 38 [13] years in men). Treated LDL cholesterol levels and lipid-lowering therapy were similar in both sexes, in particular statins (248 of 276 women [89.9%] vs 235 of 258 men [91.1%]) and lipoprotein apheresis (115 of 317 women [36.3%] vs 118 of 304 men [38.8%]). Sixteen years after HoFH diagnosis, women had statistically significant lower cumulative incidence of MI (5.0% in women vs 13.7% in men; subdistribution hazard ratio [SHR], 0.37; 95% CI, 0.21-0.66) and nonsignificantly lower all-cause mortality (3.0% in women vs 4.1% in men; HR, 0.76; 95% CI, 0.40-1.45) and cardiovascular mortality (2.6% in women vs 4.1% in men; SHR, 0.87; 95% CI, 0.44-1.75). Conclusions and Relevance In this cohort study of individuals with known HoFH, MI was higher in men compared with women yet age at diagnosis and at first ASCVD event were similar. These findings suggest that early diagnosis and treatment are important in attenuating the excessive cardiovascular risk in both sexes.
Collapse
Affiliation(s)
- Janneke W. C. M. Mulder
- Department of Internal Medicine, Erasmus Medical Center Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Tycho R. Tromp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Dirk J. Blom
- Department of Medicine, Division of Lipidology and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Krysztof Chlebus
- 1st Department of Cardiology, Medical University of Gdańsk, Gdańsk, Poland
- National Centre of Familial Hypercholesterolaemia, Gdańsk, Poland
| | - Marina Cuchel
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Laura D’Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, Sorbonne Université, Institut national de la santé et de la recherche médicale UMR 1166, Assistance Publique–Hôpitaux de Paris, Hôpital Pitié-Salpètriêre, Paris, France
| | - G. Kees Hovingh
- Department of Vascular Medicine, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Ngoc Thanh Kim
- Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam
- Department of Cardiology, Hanoi Medical University, Hanoi, Vietnam
| | - Jiang Long
- Department of Atherosclerosis, Beijing Anzhen Hospital, Capital Medical University, The Key Laboratory of Remodeling–Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Frederick J. Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Willemijn A. M. Schonck
- Department of Vascular Medicine, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Handrean Soran
- Department of Diabetes, Endocrinology and Metabolism and Manchester National Institute of Health Research/Wellcome Trust Clinical Research Facility, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Thanh-Huong Truong
- Faculty of Medicine, Phenikaa University, Hanoi City, Vietnam
- Vietnam Atherosclerosis Society, Hanoi, Vietnam
| | - Eric Boersma
- Department of Cardiology, Erasmus Medical Center Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jeanine E. Roeters van Lennep
- Department of Internal Medicine, Erasmus Medical Center Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
62
|
Pussadhamma B, Wongvipaporn C, Wutthimanop A, Nuinoon M, Porntadavity S, Jeenduang N. Identification of a novel LDLR p.Glu179Met variant in Thai families with familial hypercholesterolemia and response to treatment with PCSK9 inhibitor. Sci Rep 2024; 14:6785. [PMID: 38514665 PMCID: PMC10957951 DOI: 10.1038/s41598-024-57069-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
Familial hypercholesterolemia (FH) is a genetic disease characterized by elevated LDL-C levels. In this study, two FH probands and 9 family members from two families from northeastern Thailand were tested for LDLR, APOB, and PCSK9 variants by whole-exome sequencing, PCR-HRM, and Sanger sequencing. In silico analysis of LDLR was performed to analyse its structure‒function relationship. A novel variant of LDLR (c.535_536delinsAT, p.Glu179Met) was detected in proband 1 and proband 2 in homozygous and heterozygous forms, respectively. A total of 6 of 9 family members were heterozygous for LDLR p.Glu179Met variant. Compared with proband 2, proband 1 had higher baseline TC and LDL-C levels and a poorer response to lipid-lowering therapy combined with a PCSK9 inhibitor. Multiple sequence alignment showed that LDLR p.Glu179Met was located in a fully conserved region. Homology modelling demonstrated that LDLR p.Glu179Met variant lost one H-bond and a negative charge. In conclusion, a novel LDLR p.Glu179Met variant was identified for the first time in Thai FH patients. This was also the first report of homozygous FH patient in Thailand. Our findings may expand the knowledge of FH-causing variants in Thai population, which is beneficial for cascade screening, genetic counselling, and FH management to prevent coronary artery disease.
Collapse
Affiliation(s)
- Burabha Pussadhamma
- Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Queen Sirikit Heart Center of the Northeast, Khon Kaen University, Khon Kaen, Thailand
| | - Chaiyasith Wongvipaporn
- Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Queen Sirikit Heart Center of the Northeast, Khon Kaen University, Khon Kaen, Thailand
| | - Atthakorn Wutthimanop
- Department of Internal Medicine, Maharaj Nakhon Si Thammarat Hospital, Nakhon Si Thammarat, Thailand
| | - Manit Nuinoon
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | | | - Nutjaree Jeenduang
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand.
| |
Collapse
|
63
|
Paquette M, Baass A. Advances in familial hypercholesterolemia. Adv Clin Chem 2024; 119:167-201. [PMID: 38514210 DOI: 10.1016/bs.acc.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Familial hypercholesterolemia (FH), a semi-dominant genetic disease affecting more than 25 million people worldwide, is associated with severe hypercholesterolemia and premature atherosclerotic cardiovascular disease. Over the last decade, advances in data analysis, screening, diagnosis and cardiovascular risk stratification has significantly improved our ability to deliver precision medicine for these patients. Furthermore, recent updates on guideline recommendations and new therapeutic approaches have also proven to be highly beneficial. It is anticipated that both ongoing and upcoming clinical trials will offer further insights for the care and treatment of FH patients.
Collapse
Affiliation(s)
- Martine Paquette
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
64
|
Chia SPS, Pang JKS, Soh BS. Current RNA strategies in treating cardiovascular diseases. Mol Ther 2024; 32:580-608. [PMID: 38291757 PMCID: PMC10928165 DOI: 10.1016/j.ymthe.2024.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/22/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Cardiovascular disease (CVD) continues to impose a significant global health burden, necessitating the exploration of innovative treatment strategies. Ribonucleic acid (RNA)-based therapeutics have emerged as a promising avenue to address the complex molecular mechanisms underlying CVD pathogenesis. We present a comprehensive review of the current state of RNA therapeutics in the context of CVD, focusing on the diverse modalities that bring about transient or permanent modifications by targeting the different stages of the molecular biology central dogma. Considering the immense potential of RNA therapeutics, we have identified common gene targets that could serve as potential interventions for prevalent Mendelian CVD caused by single gene mutations, as well as acquired CVDs developed over time due to various factors. These gene targets offer opportunities to develop RNA-based treatments tailored to specific genetic and molecular pathways, presenting a novel and precise approach to address the complex pathogenesis of both types of cardiovascular conditions. Additionally, we discuss the challenges and opportunities associated with delivery strategies to achieve targeted delivery of RNA therapeutics to the cardiovascular system. This review highlights the immense potential of RNA-based interventions as a novel and precise approach to combat CVD, paving the way for future advancements in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Shirley Pei Shan Chia
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Jeremy Kah Sheng Pang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Boon-Seng Soh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore.
| |
Collapse
|
65
|
Al-Ashwal A, Alsagheir A, Al Dubayee M, Al-Khnifsawi M, Al-Sarraf A, Awan Z, Ben-Omran T, Al-Yaarubi S, Almutair A, Habeb A, Maatouk F, Alshareef M, Kholaif N, Blom D. Modern approaches to the management of homozygous familial hypercholesterolemia in the Middle East and North Africa. J Clin Lipidol 2024; 18:e132-e141. [PMID: 38158247 DOI: 10.1016/j.jacl.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
Homozygous familial hypercholesterolaemia (HoFH) is a severe form of FH in which inheritance of two defective or null mutations in genes associated with metabolism of low-density lipoprotein cholesterol (LDL-C) results in extremely high LDL-C, premature atherosclerotic cardiovascular disease (ASCVD) and mortality. Treatment of HoFH comprises a multi-modal approach of statins, ezetimibe, lipoprotein apheresis; and inhibitors of proprotein convertase subtilisin/kexin type, angiopoietin-like protein 3 (ANGPTL3) and microsomal triglyceride transfer protein. These treatments are generally costly, and patients also often require treatment for ASCVD consequent to HoFH. Therefore, in the interests of both economics and preservation of life, disease prevention via genetic screening and counselling is rapidly becoming a key element in the overall management of HoFH. Guidelines are available to assist diagnosis and treatment of HoFH; however, while advancements have been made in the management of the disease, there has been little systematic attention paid to prevention. Additionally, the Middle East/North Africa (MENA) region has a higher prevalence of HoFH than most other regions - chiefly due to consanguinity. This has led to the establishment of regional lipid clinics and awareness programs that have thrown education and awareness of HoFH into sharp focus. Incorporation of principles of prevention, education, awareness, and data from real-world use of existing therapeutics will significantly enhance the effectiveness of future guidelines for the management of HoFH, particularly in the MENA region.
Collapse
Affiliation(s)
- Abdullah Al-Ashwal
- Medical & Clinical Affairs, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia (Dr Al-Ashwal)
| | - Afaf Alsagheir
- Pediatrics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia (Dr Alsagheir)
| | - Mohammed Al Dubayee
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia (Dr Al Dubayee)
| | | | - Ahmed Al-Sarraf
- Sabah Al Ahmad Cardiac Center, Department Cardiology, Ministry of Health, Kuwait (Dr Al-Sarraf)
| | - Zuhier Awan
- Division of Clinical Biochemistry, King Abdulaziz University, Abdullah Sulayman, Jeddah, Saudi Arabia (Dr Awan)
| | - Tawfeg Ben-Omran
- Division of Genetics and Genomic Medicine, Sidra Medicine and Hamad Medical Corporation, Doha, Qatar (Dr Ben-Omran)
| | - Saif Al-Yaarubi
- Oman Medical Specialty Board, Muscat, Sultanate of Oman (Dr Al-Yaarubi)
| | - Angham Almutair
- King Abdullah Specialised Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia (Dr Almutair)
| | - Abdelhadi Habeb
- Pediatric Department, Prince Mohamed Bin Abdulaziz Hospital, Madinah, Saudi Arabia (Dr Habeb)
| | - Faouzi Maatouk
- Division of Cardiology, Department of Medicine, Fattouma Bourguiba University Hospital, Tunisia (Dr Maatouk)
| | - Manal Alshareef
- National Guard Hospital, Prince Mutib Ibn Abdullah Rd, National Guard District, Riyadh, Saudi Arabia (Dr Alshareef)
| | - Naji Kholaif
- Heart Centre Cardiology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia (Dr Kholaif); Alfaisal University College of Medicine, Riyadh, Saudi Arabia (Dr Kholaif)
| | - Dirk Blom
- Division of Lipidology, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa (Dr Blom)
| |
Collapse
|
66
|
Zhang Z, Yang R, Zhu J, Yang X, Luo H, Wang H, Luo X. Failure of lipid control by PCSK9 inhibitors in compound heterozygous familial hypercholesterolemia complicated with premature myocardial infarction: A case report. Clin Case Rep 2024; 12:e8498. [PMID: 38487640 PMCID: PMC10939999 DOI: 10.1002/ccr3.8498] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/26/2023] [Accepted: 01/16/2024] [Indexed: 03/17/2024] Open
Abstract
Key Clinical Message A certain level of low-density lipoprotein receptor activity is crucial for the efficacy of PCSK9i. Therapeutic strategies for familial hypercholesterolemia patients should consider drug efficacy, and genetic testing will be helpful. Abstract Familial hypercholesterolemia (FH) is a serious autosomal dominant disorder. Managing blood lipids in FH patients poses greater challenges for clinicians. Drug therapy may not always yield satisfactory results, particularly in individuals with low-density lipoprotein receptor (LDLR) negative mutations. Herein, we report a young female harboring an LDLR frameshift mutation. This patient developed xanthomas at 7 months old and underwent several years of treatment involving four classes of lipid-lowering drugs, including PCSK9i. However, the response to drug therapy was limited in this patient and eventually culminated in premature myocardial infarction. The efficacy of PCSK9i depends on the activity of LDLR. The inefficacy of PCSK9i may arise from the extensive mutations which leading to loss of LDLR activity. Therapy plans for these patients should take into account the efficacy of drug therapy. Early genetic testing is crucial for clinicians to make informed decisions regarding therapy options.
Collapse
Affiliation(s)
- Ziyue Zhang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
- 96608 Hospital of PLAHan ZhongShanxiP. R. China
| | - Rongpei Yang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Jun Zhu
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - XiaoLi Yang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Hao Luo
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Hongyong Wang
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| | - Xiaoli Luo
- Department of Cardiology, Daping HospitalThe Third Military Medical University (Army Medical University)ChongqingP. R. China
| |
Collapse
|
67
|
Duell PB, Banach M, Catapano AL, Laufs U, Mancini GBJ, Ray KK, Broestl C, Zhang Y, Lei L, Goldberg AC. Efficacy and safety of bempedoic acid in patients with heterozygous familial hypercholesterolemia: analysis of pooled patient-level data from phase 3 clinical trials. J Clin Lipidol 2024; 18:e153-e165. [PMID: 38341323 DOI: 10.1016/j.jacl.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND Patients with heterozygous familial hypercholesterolemia (HeFH) often cannot reach guideline-recommended low-density lipoprotein cholesterol (LDL-C) goals despite multidrug therapy. OBJECTIVE To evaluate the efficacy and safety of bempedoic acid as an add-on therapy for lowering LDL-C in patients with HeFH. METHODS Pooled data from two 52-week phase 3 clinical trials of patients with atherosclerotic cardiovascular disease and/or HeFH receiving maximally tolerated statin therapy (randomized 2:1 to bempedoic acid or placebo) were analyzed by HeFH status. Endpoints included changes from baseline to week 12 (and up to week 52) in LDL-C and other lipid parameters, achievement of LDL-C goals, and safety. RESULTS A total of 217 (bempedoic acid, 146; placebo, 71) patients with HeFH and 2,792 (bempedoic acid, 1,864; placebo, 928) without HeFH were included (mean baseline LDL-C, 172.8 mg/dL and 102.6 mg/dL, respectively). Bempedoic acid significantly lowered LDL-C at week 12 vs. placebo regardless of HeFH status (with HeFH, -21.2%; without HeFH, -18.2% [both P<0.0001]). Bempedoic acid significantly reduced other lipid parameters and high-sensitivity C-reactive protein vs. placebo regardless of HeFH status (all P≤0.01). Among patients with HeFH treated with bempedoic acid, 32% and 27% achieved LDL-C <100 mg/dL at weeks 12 and 52, respectively. Overall treatment-emergent adverse event incidence was comparable across all four groups (74.7-77.5%). CONCLUSION Bempedoic acid significantly lowered LDL-C levels vs. placebo and was generally well tolerated in all patients, with no new safety findings in patients with HeFH, despite more intensive lipid-lowering therapy in patients with vs. without HeFH.
Collapse
Affiliation(s)
- P Barton Duell
- Center for Preventive Cardiology, Knight Cardiovascular Institute and Division of Endocrinology, Diabetes, and Clinical Nutrition, School of Medicine, Oregon Health & Science University, Portland, OR, USA (Dr Duell).
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Łódź (MUL), Łódź, Poland (Dr Banach)
| | | | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany (Dr Laufs)
| | - G B John Mancini
- Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada (Dr Mancini)
| | - Kausik K Ray
- Department of Primary Care and Public Health, Imperial College London, London, UK (Dr Ray)
| | - Christine Broestl
- Esperion Therapeutics, Inc., Ann Arbor, MI, USA (Drs Broestl, Zhang, Lei)
| | - Yang Zhang
- Esperion Therapeutics, Inc., Ann Arbor, MI, USA (Drs Broestl, Zhang, Lei)
| | - Lei Lei
- Esperion Therapeutics, Inc., Ann Arbor, MI, USA (Drs Broestl, Zhang, Lei)
| | - Anne C Goldberg
- Department of Medicine, Washington University, St. Louis, MO, USA (Dr Goldberg)
| |
Collapse
|
68
|
Al-Baldawi Z, Brown L, Ruel I, Baass A, Bergeron J, Cermakova L, Couture P, Gaudet D, Francis GA, Hegele RA, Iatan I, Mancini GBJ, McCrindle BW, Ransom T, Sherman MH, McPherson R, Genest J, Brunham LR. Sex differences in the presentation, treatment and outcomes of patients with homozygous familial hypercholesterolemia. J Clin Lipidol 2024; 18:e189-e196. [PMID: 38281851 DOI: 10.1016/j.jacl.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/30/2024]
Abstract
BACKGROUND Homozygous familial hypercholesterolemia (HoFH) is a rare, autosomal semi-dominant lipid metabolism disorder characterized by extremely high low-density lipoprotein cholesterol (LDL-C) levels and premature cardiovascular disease. The objective of this study was to investigate sex-differences in the treatment and outcomes of patients with HoFH. METHODS We examined clinical characteristics, lipid-lowering therapy (LLT), and cardiovascular events using descriptive statistics of patients in the Canadian HoFH registry. Major adverse cardiovascular events (MACE) were defined as the composite of cardiovascular death, non-fatal myocardial infarction, and stroke. Sex differences between continuous and categorical variables were analyzed using Mann-Whitney U test and Fisher's Exact test, respectively. RESULTS This study included 48 patients (27 (56%) female). The median age at diagnosis in females was 14.0 (interquartile range (IQR) 9.0-30.0) and in males was 8.0 (IQR 2.0-23.0) (p = 0.07). Baseline clinical characteristics were comparable between both sexes. The median baseline LDL-C was 12.7 mmol/L (10.0-18.3) in females and 15.3 (10.5-20.0) in males (p = 0.51). Follow up LDL-C levels were 7.6 mmol/L (IQR 4.8-11.0) in females and 6.3 (IQR 4.6-7.5) in males (p = 0.1). Most patients were taking 3 or more LLTs, with comparable proportions in both sexes (p = 0.26). Apheresis was similar in both sexes, 14 (51.8%) vs. 10 (47.6%) (p = 0.2). Over a mean of 10 years of follow-up, MACE occurred in 3 females (11.1%) and 4 males (19.1%) (p = 0.2). CONCLUSION Lipid levels and treatment were similar between sexes. MACE occurred in similar proportions between sexes, indicating that HoFH offsets the inherently lower cardiovascular risk in pre-menopausal females. Further investigation into sex-differences in HoFH in larger sample sizes is warranted.
Collapse
Affiliation(s)
- Zobaida Al-Baldawi
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada (Dr Al-Baldawi)
| | - Leslie Brown
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada (Drs Brown, Ruel, Baass, Sherman, Genest)
| | - Isabelle Ruel
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada (Drs Brown, Ruel, Baass, Sherman, Genest)
| | - Alexis Baass
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada (Drs Brown, Ruel, Baass, Sherman, Genest)
| | - Jean Bergeron
- Endocrinology and Nephrology Unit, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada (Drs Bergeron, Couture)
| | - Lubomira Cermakova
- Centre for Heart Lung Innovation, Providence Health Care Research, Institute, Department of Medicine, University of British Columbia, Vancouver, BC, Canada (Drs Cermakova, Francis, Iatan, Brunham)
| | - Patrick Couture
- Endocrinology and Nephrology Unit, CHU de Québec - Université Laval Research Center, Québec City, QC, Canada (Drs Bergeron, Couture)
| | - Daniel Gaudet
- ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, QC, Canada (Dr Gaudet)
| | - Gordon A Francis
- Centre for Heart Lung Innovation, Providence Health Care Research, Institute, Department of Medicine, University of British Columbia, Vancouver, BC, Canada (Drs Cermakova, Francis, Iatan, Brunham)
| | - Robert A Hegele
- Departments of Medicine and Biochemistry, Schulich School of, Medicine and Robarts Research Institute, Western University, London, ON, Canada (Dr Hegele)
| | - Iulia Iatan
- Centre for Heart Lung Innovation, Providence Health Care Research, Institute, Department of Medicine, University of British Columbia, Vancouver, BC, Canada (Drs Cermakova, Francis, Iatan, Brunham)
| | - G B John Mancini
- Centre for, Cardiovascular Innovation, University of British Columbia, Vancouver, BC, Canada (Dr Mancini)
| | - Brian W McCrindle
- Department of Pediatrics, The Labatt Family Heart Centre, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada (Dr McCrindle)
| | - Thomas Ransom
- Queen Elizabeth II Health Sciences Centre, Dalhousie University, Halifax, NS, Canada (Dr Ransom)
| | - Mark H Sherman
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada (Drs Brown, Ruel, Baass, Sherman, Genest); Department of Endocrinology, McGill University, Health Centre, Montreal, QC, Canada (Dr Sherman)
| | - Ruth McPherson
- Lipid Clinic & Atherogenomics Laboratory, University, of Ottawa Heart Institute, Ottawa, ON, Canada (Dr McPherson)
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada (Drs Brown, Ruel, Baass, Sherman, Genest)
| | - Liam R Brunham
- Centre for Heart Lung Innovation, Providence Health Care Research, Institute, Department of Medicine, University of British Columbia, Vancouver, BC, Canada (Drs Cermakova, Francis, Iatan, Brunham)
| |
Collapse
|
69
|
White RT, Sankey KH, Nawarskas JJ. Evinacumab-dgnb (Evkeeza-REGN1500), A Novel Lipid-Lowering Therapy for Homozygous Familial Hypercholesterolemia. Cardiol Rev 2024; 32:180-185. [PMID: 37071085 DOI: 10.1097/crd.0000000000000522] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Chronically elevated low-density lipoprotein (LDL) has harmful effects on the vasculature including increased vasoconstriction and the formation of plaques which may rupture, causing coronary heart disease and stroke. In patients with familial hypercholesterolemia, adequate reduction of LDL is especially challenging. Although HMG-CoA reductase inhibitors (statins) are the mainstays for LDL lowering, other treatments such as proprotein convertase subtilisin/kexin type 9 inhibitors, bempedoic acid, incliseran, lomitapide, and apheresis have been employed in an effort to achieve adequate LDL reduction in these patients. Despite these available therapies, many patients with familial hypercholesterolemia do not meet the LDL targets suggested in current guidelines. Evinacumab is a novel lipid-lowering therapy that exerts its LDL-lowering effect through inhibition of angiopoietin-like protein 3 (ANGPTL3). ANGPTL3 inhibits the breakdown of triglyceride-rich lipoproteins, such as very low-density lipoprotein and chylomicrons. By inhibiting ANGPTL3, evinacumab allows these lipoproteins to be degraded, ultimately leading to reductions in LDL, high-density lipoprotein, and triglycerides. Clinical trials have demonstrated evinacumab to be safe and effective in reducing LDL. However, data are lacking regarding its potential to reduce risk of atherosclerotic cardiovascular disease. Evinacumab is generally well tolerated with the primary adverse effects comprising infusion reactions, nasopharyngitis, influenza-like illness, dizziness, rhinorrhea, and nausea. While evinacumab is an interesting therapy, until it is proven to reduce cardiovascular events, its high cost leaves its anticipated role in therapy somewhat ambiguous. In the meantime, it may be a useful therapy for those with homozygous familial hypercholesterolemia.
Collapse
Affiliation(s)
- Raechel T White
- From the Department of Pharmacy Practice and Administrative sciences, College of Pharmacy, University of New Mexico Health Science Center, Albuquerque, NM
| | | | - James J Nawarskas
- From the Department of Pharmacy Practice and Administrative sciences, College of Pharmacy, University of New Mexico Health Science Center, Albuquerque, NM
| |
Collapse
|
70
|
Alshahrani A, Kholaif N, Al-Khnifsawi M, Zarif H, Mahzari M. The Effect of PCSK9 Inhibitors on LDL-C Target Achievement in Patients with Homozygous Familial Hypercholesterolemia: A Retrospective Cohort Analysis. Adv Ther 2024; 41:837-846. [PMID: 38169062 PMCID: PMC10838833 DOI: 10.1007/s12325-023-02764-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
Abstract
INTRODUCTION Achieving target low-density lipoprotein-cholesterol (LDL-C) levels remains challenging when treating homozygous familial hypercholesterolemia (HoFH). Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) are prescribed in addition to statins and ezetimibe, but patients' response varies and depends on residual low-density lipoprotein receptor (LDLR) function. METHODS A multicenter, retrospective observational analysis evaluated LDL-C target achievement in response to PCSK9i treatment in 28 patients with HoFH from the Middle East/North Africa region. Effect of genotype was investigated. Demographic and clinical information was retrospectively obtained from medical records. Patient response to PCSK9i treatment was assessed by calculating percentage changes in lipid levels from pre-PCSK9i treatment baseline to most recent follow-up visit where patients were recorded as receiving PCSK9i on top of standard of care lipid-lowering therapies (LLTs; i.e., statins/ezetimibe) and assessing European Atherosclerosis Society (EAS) target achievement up to January 31, 2022. Lowest LDL-C level while receiving PCSK9i was identified. RESULTS The cohort (n = 28) had a mean age (standard deviation; SD) of 22.8 (9.8) years (n = 28) and was 51% female (n = 27). Baseline LDL-C data were available in 24/28 (85.7%) patients (mean [SD] 14.0 [3.0] mmol/L). Median (interquartile range) duration of PCSK9i treatment was 12.0 months (4.0-19.1) months and mean (SD) % change in LDL-C after PCSK9i treatment was - 8.6% (12.1). LDL-C reduction from baseline was below 15% in 17/24 patients (70.8%). In the full cohort, mean (SD) minimum LDL-C during PCSK9i treatment was 11.9 (2.8; n = 28) mmol/L. No patient achieved EAS target LDL-C while receiving PCSK9i; genotype analysis suggested LDLR-null/null patients were most refractory to PCSK9i. CONCLUSION Response to PCSK9i was minimal in this cohort of patients with HoFH. No patients achieved EAS LDL-C targets, and most failed to reach the EAS-recommended 15% LDL-C reduction for PCSK9i therapy continuation. These results suggest additional LLTs are necessary to achieve LDL-C targets in HoFH.
Collapse
Affiliation(s)
- Awad Alshahrani
- Department of Medicine, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Naji Kholaif
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Alfaisal University, Al Takhassousi, Riyadh, Saudi Arabia
| | | | - Hawazen Zarif
- Department of Medicine, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Moeber Mahzari
- Department of Medicine, Ministry of the National Guard-Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
71
|
Medeiros AM, Alves AC, Miranda B, Chora JR, Bourbon M. Unraveling the genetic background of individuals with a clinical familial hypercholesterolemia phenotype. J Lipid Res 2024; 65:100490. [PMID: 38122934 PMCID: PMC10832474 DOI: 10.1016/j.jlr.2023.100490] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Familial hypercholesterolemia (FH) is a common genetic disorder of lipid metabolism caused by pathogenic/likely pathogenic variants in LDLR, APOB, and PCSK9 genes. Variants in FH-phenocopy genes (LDLRAP1, APOE, LIPA, ABCG5, and ABCG8), polygenic hypercholesterolemia, and hyperlipoprotein (a) [Lp(a)] can also mimic a clinical FH phenotype. We aim to present a new diagnostic tool to unravel the genetic background of clinical FH phenotype. Biochemical and genetic study was performed in 1,005 individuals with clinical diagnosis of FH, referred to the Portuguese FH Study. A next-generation sequencing panel, covering eight genes and eight SNPs to determine LDL-C polygenic risk score and LPA genetic score, was validated, and used in this study. FH was genetically confirmed in 417 index cases: 408 heterozygotes and 9 homozygotes. Cascade screening increased the identification to 1,000 FH individuals, including 11 homozygotes. FH-negative individuals (phenotype positive and genotype negative) have Lp(a) >50 mg/dl (30%), high polygenic risk score (16%), other monogenic lipid metabolism disorders (1%), and heterozygous pathogenic variants in FH-phenocopy genes (2%). Heterozygous variants of uncertain significance were identified in primary genes (12%) and phenocopy genes (7%). Overall, 42% of our cohort was genetically confirmed with FH. In the remaining individuals, other causes for high LDL-C were identified in 68%. Hyper-Lp(a) or polygenic hypercholesterolemia may be the cause of the clinical FH phenotype in almost half of FH-negative individuals. A small part has pathogenic variants in ABCG5/ABCG8 in heterozygosity that can cause hypercholesterolemia and should be further investigated. This extended next-generation sequencing panel identifies individuals with FH and FH-phenocopies, allowing to personalize each person's treatment according to the affected pathway.
Collapse
Affiliation(s)
- Ana Margarida Medeiros
- Unidade de I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Catarina Alves
- Unidade de I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Beatriz Miranda
- Unidade de I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Joana Rita Chora
- Unidade de I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Mafalda Bourbon
- Unidade de I&D, Grupo de Investigação Cardiovascular, Departamento de Promoção da Saúde e Prevenção de Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisboa, Portugal; BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
72
|
Kozan DW, Farber SA. Is It Ever Wise to Edit Wild-Type Alleles? Engineered CRISPR Alleles Versus Millions of Years of Human Evolution. Arterioscler Thromb Vasc Biol 2024; 44:328-333. [PMID: 38059350 PMCID: PMC10948015 DOI: 10.1161/atvbaha.123.318069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
The tremendous burden of lipid metabolism diseases, coupled with recent developments in human somatic gene editing, has motivated researchers to propose population-wide somatic gene editing of PCSK9 (proprotein convertase subtilisin/kexin type 9) within the livers of otherwise healthy humans. The best-characterized molecular function of PCSK9 is its ability to regulate plasma LDL (low-density lipoprotein) levels through promoting LDL receptor degradation. Individuals with loss-of-function PCSK9 variants have lower levels of plasma LDL and reduced cardiovascular disease. Gain-of-function variants of PCSK9 are strongly associated with familial hypercholesterolemia. A new therapeutic strategy delivers CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats; CRISPR-associated protein 9) specifically to liver cells to edit the wild-type alleles of PCSK9 with the goal of producing a loss-of-function allele. This direct somatic gene editing approach is being pursued despite the availability of US Food and Drug Administration-approved PCSK9 inhibitors that lower plasma LDL levels. Here, we discuss other characterized functions of PCSK9 including its role in infection and host immunity. We explore important factors that may have contributed to the evolutionary selection of PCSK9 in several vertebrates, including humans. Until such time that more fully understand the multiple biological roles of PCSK9, the ethics of permanently editing the gene locus in healthy, wild-type populations remains highly questionable.
Collapse
Affiliation(s)
- Darby W. Kozan
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States
| | - Steven A. Farber
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
73
|
Anderson S, Botti C. The genetics of autosomal dominant familial hypercholesterolemia. J Am Assoc Nurse Pract 2024; 36:136-142. [PMID: 37624754 DOI: 10.1097/jxx.0000000000000930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/23/2023] [Indexed: 08/27/2023]
Abstract
ABSTRACT Familial hypercholesterolemia (FH) is one of the most common genetic conditions. Affected individuals are unable to metabolize cholesterol due to inherited changes in the low-density lipoprotein (LDL) receptor, which impairs the ability to metabolize cholesterol, resulting in extremely high levels of cholesterol that leads to premature coronary artery disease. Autosomal dominant FH is caused by variants in several genes, which may present as heterozygous FH (less severe) or homozygous FH (more severe). Clinical diagnosis may be more likely when there is a family history of two or more first-degree relatives with total and LDL-cholesterol (LDL-C) level elevations, a child is identified, or the affected individual or close relatives have tendon xanthomas and/or progressive atherosclerosis. This article provides an overview of autosomal dominant FH, including disease prevalence, clinical diagnostic criteria, genetic variants, diagnostic testing, pathognomonic findings, and treatment options. It also shares a brief case, which highlights challenges associated with genetic test interpretation and the importance of including experienced providers in the diagnosis and treatment of this underdiagnosed and often untreated or undertreated genetic condition.
Collapse
Affiliation(s)
- Sharon Anderson
- Division of Medical Genetics, Rutgers Robert Wood Johnson Medical School, Rutgers Health, Child Health Institute of New Jersey, New Brunswick, New Jersey
- Division of Advanced Nursing Practice, School of Nursing, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Christina Botti
- Division of Medical Genetics, Rutgers Robert Wood Johnson Medical School, Rutgers Health, Child Health Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
74
|
Raal F, Durst R, Bi R, Talloczy Z, Maheux P, Lesogor A, Kastelein JJ. Efficacy, Safety, and Tolerability of Inclisiran in Patients With Homozygous Familial Hypercholesterolemia: Results From the ORION-5 Randomized Clinical Trial. Circulation 2024; 149:354-362. [PMID: 37850379 PMCID: PMC10815002 DOI: 10.1161/circulationaha.122.063460] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 09/22/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Homozygous familial hypercholesterolemia is a genetic disease characterized by extremely high levels of low-density lipoprotein cholesterol (LDL-C) and a high risk of premature cardiovascular events. The proof-of-concept study ORION-2 (A Study of Inclisiran in Participants With Homozygous Familial Hypercholesterolemia) showed that inclisiran, a small interfering RNA that prevents production of the hepatic PCSK9 protein (proprotein convertase subtilisin/kexin type 9), could lead to durable reductions in LDL-C levels when added to statins and ezetimibe in patients with homozygous familial hypercholesterolemia. METHODS ORION-5 was a phase 3, 2-part, multicenter study in 56 patients with homozygous familial hypercholesterolemia and elevated LDL-C levels despite maximum tolerated doses of LDL-C-lowering therapies with or without lipoprotein apheresis. Patients eligible for part 1 (double-blind, 6 months) were randomized 2:1 to receive either 300 mg of inclisiran sodium (equivalent to 284 mg of inclisiran) or placebo. Placebo-treated patients from part 1 were transitioned to inclisiran in part 2 (open-label, 18 months). The primary end point was the percentage change in LDL-C levels from baseline to day 150. RESULTS The mean age of the patients was 42.7 years, and 60.7% were women. The mean baseline LDL-C levels were 294.0 mg/dL and 356.7 mg/dL in the inclisiran and placebo groups, respectively. The placebo-corrected percentage change in LDL-C level from baseline to day 150 was -1.68% (95% CI, -29.19% to 25.83%; P=0.90), and the difference was not statistically significant between the inclisiran and placebo groups. The placebo-corrected percentage change in PCSK9 levels from baseline to day 150 was -60.6% with inclisiran treatment (P<0.0001); this was sustained throughout the study, confirming the effect of inclisiran on its biological target of PCSK9. No statistically significant differences between the inclisiran and placebo groups were observed in the levels of other lipids and lipoproteins (apolipoprotein B, total cholesterol, and non-high-density lipoprotein cholesterol). Adverse events and serious adverse events did not differ between the inclisiran and placebo groups throughout the study. CONCLUSIONS Inclisiran treatment did not reduce LDL-C levels in patients with homozygous familial hypercholesterolemia despite substantial lowering of PCSK9 levels. Inclisiran was well-tolerated, and the safety findings were consistent with previously reported studies and the overall safety profile. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03851705.
Collapse
Affiliation(s)
- Frederick Raal
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa (F.R.)
| | - Ronen Durst
- Cardiology Department, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel (R.D.)
| | - Ran Bi
- Novartis Pharmaceuticals Corporation, East Hannover, NJ (R.B., Z.T.)
| | - Zsolt Talloczy
- Novartis Pharmaceuticals Corporation, East Hannover, NJ (R.B., Z.T.)
| | - Pierre Maheux
- Novartis Pharmaceuticals Corporation, Basel, Switzerland (P.M., A.L.)
| | - Anastasia Lesogor
- Novartis Pharmaceuticals Corporation, Basel, Switzerland (P.M., A.L.)
| | - John J.P. Kastelein
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Netherlands (J.J.P.K.)
| |
Collapse
|
75
|
Fularski P, Hajdys J, Majchrowicz G, Stabrawa M, Młynarska E, Rysz J, Franczyk B. Unveiling Familial Hypercholesterolemia-Review, Cardiovascular Complications, Lipid-Lowering Treatment and Its Efficacy. Int J Mol Sci 2024; 25:1637. [PMID: 38338916 PMCID: PMC10855128 DOI: 10.3390/ijms25031637] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Familial hypercholesterolemia (FH) is a genetic disorder primarily transmitted in an autosomal-dominant manner. We distinguish two main forms of FH, which differ in the severity of the disease, namely homozygous familial hypercholesterolemia (HoFH) and heterozygous familial hypercholesterolemia (HeFH). The characteristic feature of this disease is a high concentration of low-density lipoprotein cholesterol (LDL-C) in the blood. However, the level may significantly vary between the two mentioned types of FH, and it is decidedly higher in HoFH. A chronically elevated concentration of LDL-C in the plasma leads to the occurrence of certain abnormalities, such as xanthomas in the tendons and skin, as well as corneal arcus. Nevertheless, a significantly more severe phenomenon is leading to the premature onset of cardiovascular disease (CVD) and its clinical implications, such as cardiac events, stroke or vascular dementia, even at a relatively young age. Due to the danger posed by this medical condition, we have investigated how both non-pharmacological and selected pharmacological treatment impact the course of FH, thereby reducing or postponing the risk of clinical manifestations of CVD. The primary objective of this review is to provide a comprehensive summary of the current understanding of FH, the effectiveness of lipid-lowering therapy in FH and to explain the anatomopathological correlation between FH and premature CVD development, with its complications.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Joanna Hajdys
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Gabriela Majchrowicz
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Magdalena Stabrawa
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
76
|
Picano E, Pierard L, Peteiro J, Djordjevic-Dikic A, Sade LE, Cortigiani L, Van De Heyning CM, Celutkiene J, Gaibazzi N, Ciampi Q, Senior R, Neskovic AN, Henein M. The clinical use of stress echocardiography in chronic coronary syndromes and beyond coronary artery disease: a clinical consensus statement from the European Association of Cardiovascular Imaging of the ESC. Eur Heart J Cardiovasc Imaging 2024; 25:e65-e90. [PMID: 37798126 DOI: 10.1093/ehjci/jead250] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023] Open
Abstract
Since the 2009 publication of the stress echocardiography expert consensus of the European Association of Echocardiography, and after the 2016 advice of the American Society of Echocardiography-European Association of Cardiovascular Imaging for applications beyond coronary artery disease, new information has become available regarding stress echo. Until recently, the assessment of regional wall motion abnormality was the only universally practiced step of stress echo. In the state-of-the-art ABCDE protocol, regional wall motion abnormality remains the main step A, but at the same time, regional perfusion using ultrasound-contrast agents may be assessed. Diastolic function and pulmonary B-lines are assessed in step B; left ventricular contractile and preload reserve with volumetric echocardiography in step C; Doppler-based coronary flow velocity reserve in the left anterior descending coronary artery in step D; and ECG-based heart rate reserve in non-imaging step E. These five biomarkers converge, conceptually and methodologically, in the ABCDE protocol allowing comprehensive risk stratification of the vulnerable patient with chronic coronary syndromes. The present document summarizes current practice guidelines recommendations and training requirements and harmonizes the clinical guidelines of the European Society of Cardiology in many diverse cardiac conditions, from chronic coronary syndromes to valvular heart disease. The continuous refinement of imaging technology and the diffusion of ultrasound-contrast agents improve image quality, feasibility, and reader accuracy in assessing wall motion and perfusion, left ventricular volumes, and coronary flow velocity. Carotid imaging detects pre-obstructive atherosclerosis and improves risk prediction similarly to coronary atherosclerosis. The revolutionary impact of artificial intelligence on echocardiographic image acquisition and analysis makes stress echo more operator-independent and objective. Stress echo has unique features of low cost, versatility, and universal availability. It does not need ionizing radiation exposure and has near-zero carbon dioxide emissions. Stress echo is a convenient and sustainable choice for functional testing within and beyond coronary artery disease.
Collapse
Affiliation(s)
- Eugenio Picano
- Institute of Clinical Physiology of the National Research Council, CNR, Via Moruzzi 1, 56124 Pisa, Italy
| | - Luc Pierard
- University of Liège, Walloon Region, Belgium
| | - Jesus Peteiro
- CHUAC-Complexo Hospitalario Universitario A Coruna, CIBER-CV, University of A Coruna, 15070 La Coruna, Spain
| | - Ana Djordjevic-Dikic
- Cardiology Clinic, University Clinical Centre of Serbia, Medical School, University of Belgrade, 11000 Belgrade, Serbia
| | - Leyla Elif Sade
- University of Pittsburgh Medical Center UPMC Heart & Vascular Institute, Pittsburgh, PA, USA
| | | | | | - Jelena Celutkiene
- Centre of Cardiology and Angiology, Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
| | - Nicola Gaibazzi
- Cardiology Department, Parma University Hospital, 43100 Parma, Italy
| | - Quirino Ciampi
- Cardiology Division, Fatebenefratelli Hospital, 82100 Benevento, Italy
| | - Roxy Senior
- Imperial College, UK
- Royal Brompton Hospital Imperial College London, UK
- Northwick Park Hospital, London, UK
| | - Aleksandar N Neskovic
- Department of Cardiology, University Clinical Hospital Center Zemun-Belgrade Faculty of Medicine, University of Belgrade, Serbia
| | - Michael Henein
- Department of Public Health and Clinical Medicine Units: Section of Medicine, Umea University, Umea, Sweden
| |
Collapse
|
77
|
Wang M, Hong L, Cai L, Zhang Z, Jiang N, Chen Y, Ying Q, Kong L, Wei Z, Xu Y, Jin L. Novel LDLR variants affecting low density lipoprotein metabolism identified in familial hypercholesterolemia. Mol Biol Rep 2024; 51:153. [PMID: 38236436 DOI: 10.1007/s11033-023-09169-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/15/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is an autosomal dominant disease of lipid metabolism mainly caused by mutations in the low-density lipoprotein receptor (LDLR) gene. Genetic detection of patients with FH help with precise diagnosis and treatment, thus reducing the risk of coronary heart disease (CHD) and other related diseases. The study aimed to identify the causative gene mutations in a Chinese FH family and reveal the pathogenicity and the mechanism of these mutations. METHODS AND RESULTS Whole exome sequencing was performed in a patient with severe lipid metabolism dysfunction seeking fertility guidance from a Chinese FH family. Two LDLR variants c.1875 C > G (p.N625K; novel variant) and c.1448G > A (p.W483*) were identified in the family. Wildtype and mutant LDLR constructs were established by the site-direct mutagenesis technique. Functional studies were carried out by cell transfection to evaluate the impact of detected variants on LDLR activity. The two variants were proven to affect LDL uptake and binding, resulting in cholesterol clearance reduction to different degrees. According to The American College of Medical Genetics and Genomics (ACMG) Standards and Guidelines, the W483* variant was classified as "Pathogenic", while the N625K variant as "VUS". CONCLUSIONS Our results provide novel experimental evidence of functional alteration by LDLR variants identified in our study and expand the mutational spectrum of LDLR mutation induced FH.
Collapse
Affiliation(s)
- Miao Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Ling Hong
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Luyi Cai
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Ziyi Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Ningdong Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yijing Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qian Ying
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Lingpeng Kong
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Zhiyun Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
78
|
Rogozik J, Główczyńska R, Grabowski M. Genetic backgrounds and diagnosis of familial hypercholesterolemia. Clin Genet 2024; 105:3-12. [PMID: 37849044 DOI: 10.1111/cge.14435] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023]
Abstract
Lipid disorders play a critical role in the intricate development of atherosclerosis and its clinical consequences, such as coronary heart disease and stroke. These disorders are responsible for a significant number of deaths in many adult populations worldwide. Familial hypercholesterolemia (FH) is a genetic disorder that causes extremely high levels of LDL cholesterol. The most common mutations occur in genes responsible for low-density lipoprotein receptor (LDLR), apolipoprotein B (APOB), or proprotein convertase subtilisin/kexin type 9 (PCSK9). While genetic testing is a dependable method for diagnosing the disease, it may not detect primary mutations in 20%-40% of FH cases.
Collapse
Affiliation(s)
- Joanna Rogozik
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Renata Główczyńska
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Grabowski
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
79
|
Ai JY, Zhao PC, Zhang W, Rao GW. Research Progress in the Clinical Treatment of Familial Hypercholesterolemia. Curr Med Chem 2024; 31:1082-1106. [PMID: 36733200 DOI: 10.2174/0929867330666230202111849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 02/04/2023]
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant inheritable disease with severe disorders of lipid metabolism. It is mainly marked by increasing levels of plasma total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C), xanthoma, corneal arch, and early-onset coronary heart disease (CHD). The prevalence of FH is high, and it is dangerous and clinically underdiagnosed. The clinical treatment for FH includes both pharmacological and non-pharmacological treatment, of which non-pharmacological treatment mainly includes therapeutic lifestyle change and dietary therapy, LDL apheresis, liver transplantation and gene therapy. In recent years, many novel drugs have been developed to treat FH more effectively. In addition, the continuous maturity of non-pharmacological treatment techniques has also brought more hope for the treatment of FH. This paper analyzes the pathogenic mechanism and the progress in clinical treatment of FH. Furthermore, it also summarizes the mechanism and structure-activity relationship of FH therapeutic drugs that have been marketed. In a word, this article provides a reference value for the research and development of FH therapeutic drugs.
Collapse
Affiliation(s)
- Jing-Yan Ai
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| | - Peng-Cheng Zhao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| | - Wen Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, P.R. China
| |
Collapse
|
80
|
de Oliveira J, Moreira ELG, de Bem AF. Beyond cardiovascular risk: Implications of Familial hypercholesterolemia on cognition and brain function. Ageing Res Rev 2024; 93:102149. [PMID: 38056504 DOI: 10.1016/j.arr.2023.102149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Familial hypercholesterolemia (FH) is a metabolic condition caused mainly by a mutation in the low-density lipoprotein (LDL) receptor gene (LDLR), which is highly prevalent in the population. Besides being an important causative factor of cardiovascular diseases, FH has been considered an early risk factor for Alzheimer's disease. Cognitive and emotional behavioral impairments in LDL receptor knockout (LDLr-/-) mice are associated with neuroinflammation, blood-brain barrier dysfunction, impaired neurogenesis, brain oxidative stress, and mitochondrial dysfunction. Notably, today, LDLr-/- mice, a widely used animal model for studying cardiovascular diseases and atherosclerosis, are also considered an interesting tool for studying dementia. Here, we reviewed the main findings in LDLr-/- mice regarding the relationship between FH and brain dysfunctions and dementia development.
Collapse
Affiliation(s)
- Jade de Oliveira
- Laboratory of investigation on metabolic disorders and neurodegenerative diseases, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil.
| | - Eduardo Luiz Gasnhar Moreira
- Neuroscience Coworking Lab, Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil.
| | - Andreza Fabro de Bem
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, University of Brasilia, Brasília, Federal District, DF 70910-900, Brazil; Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040360, Brazil.
| |
Collapse
|
81
|
Wang S, Link F, Han M, Chaudhary R, Asimakopoulos A, Liebe R, Yao Y, Hammad S, Dropmann A, Krizanac M, Rubie C, Feiner LK, Glanemann M, Ebert MPA, Weiskirchen R, Henis YI, Ehrlich M, Dooley S. The Interplay of TGF-β1 and Cholesterol Orchestrating Hepatocyte Cell Fate, EMT, and Signals for HSC Activation. Cell Mol Gastroenterol Hepatol 2023; 17:567-587. [PMID: 38154598 PMCID: PMC10883985 DOI: 10.1016/j.jcmgh.2023.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND & AIMS Transforming growth factor-β1 (TGF-β1) plays important roles in chronic liver diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD involves various biological processes including dysfunctional cholesterol metabolism and contributes to progression to metabolic dysfunction-associated steatohepatitis and hepatocellular carcinoma. However, the reciprocal regulation of TGF-β1 signaling and cholesterol metabolism in MASLD is yet unknown. METHODS Changes in transcription of genes associated with cholesterol metabolism were assessed by RNA sequencing of murine hepatocyte cell line (alpha mouse liver 12/AML12) and mouse primary hepatocytes treated with TGF-β1. Functional assays were performed on AML12 cells (untreated, TGF-β1 treated, or subjected to cholesterol enrichment [CE] or cholesterol depletion [CD]), and on mice injected with adenovirus-associated virus 8-control/TGF-β1. RESULTS TGF-β1 inhibited messenger RNA expression of several cholesterol metabolism regulatory genes, including rate-limiting enzymes of cholesterol biosynthesis in AML12 cells, mouse primary hepatocytes, and adenovirus-associated virus-TGF-β1-treated mice. Total cholesterol levels and lipid droplet accumulation in AML12 cells and liver tissue also were reduced upon TGF-β1 treatment. Smad2/3 phosphorylation after 2 hours of TGF-β1 treatment persisted after CE or CD and was mildly increased after CD, whereas TGF-β1-mediated AKT phosphorylation (30 min) was inhibited by CE. Furthermore, CE protected AML12 cells from several effects mediated by 72 hours of incubation with TGF-β1, including epithelial-mesenchymal transition, actin polymerization, and apoptosis. CD mimicked the outcome of long-term TGF-β1 administration, an effect that was blocked by an inhibitor of the type I TGF-β receptor. In addition, the supernatant of CE- or CD-treated AML12 cells inhibited or promoted, respectively, the activation of LX-2 hepatic stellate cells. CONCLUSIONS TGF-β1 inhibits cholesterol metabolism whereas cholesterol attenuates TGF-β1 downstream effects in hepatocytes.
Collapse
Affiliation(s)
- Sai Wang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Frederik Link
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Mei Han
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Internal Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Roohi Chaudhary
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Anastasia Asimakopoulos
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, Aachen, Germany
| | - Roman Liebe
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Ye Yao
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Seddik Hammad
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anne Dropmann
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marinela Krizanac
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, Aachen, Germany
| | - Claudia Rubie
- Department of General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Laura Kim Feiner
- Department of General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias Glanemann
- Department of General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias P A Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Clinical Cooperation Unit Healthy Metabolism, Center of Preventive Medicine and Digital Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, Aachen, Germany
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Steven Dooley
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
82
|
Gogate A, Belcourt J, Shah M, Wang AZ, Frankel A, Kolmel H, Chalon M, Stephen P, Kolli A, Tawfik SM, Jin J, Bahal R, Rasmussen TP, Manautou JE, Zhong XB. Targeting the Liver with Nucleic Acid Therapeutics for the Treatment of Systemic Diseases of Liver Origin. Pharmacol Rev 2023; 76:49-89. [PMID: 37696583 PMCID: PMC10753797 DOI: 10.1124/pharmrev.123.000815] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
Systemic diseases of liver origin (SDLO) are complex diseases in multiple organ systems, such as cardiovascular, musculoskeletal, endocrine, renal, respiratory, and sensory organ systems, caused by irregular liver metabolism and production of functional factors. Examples of such diseases discussed in this article include primary hyperoxaluria, familial hypercholesterolemia, acute hepatic porphyria, hereditary transthyretin amyloidosis, hemophilia, atherosclerotic cardiovascular diseases, α-1 antitrypsin deficiency-associated liver disease, and complement-mediated diseases. Nucleic acid therapeutics use nucleic acids and related compounds as therapeutic agents to alter gene expression for therapeutic purposes. The two most promising, fastest-growing classes of nucleic acid therapeutics are antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs). For each listed SDLO disease, this article discusses epidemiology, symptoms, genetic causes, current treatment options, and advantages and disadvantages of nucleic acid therapeutics by either ASO or siRNA drugs approved or under development. Furthermore, challenges and future perspectives on adverse drug reactions and toxicity of ASO and siRNA drugs for the treatment of SDLO diseases are also discussed. In summary, this review article will highlight the clinical advantages of nucleic acid therapeutics in targeting the liver for the treatment of SDLO diseases. SIGNIFICANCE STATEMENT: Systemic diseases of liver origin (SDLO) contain rare and common complex diseases caused by irregular functions of the liver. Nucleic acid therapeutics have shown promising clinical advantages to treat SDLO. This article aims to provide the most updated information on targeting the liver with antisense oligonucleotides and small interfering RNA drugs. The generated knowledge may stimulate further investigations in this growing field of new therapeutic entities for the treatment of SDLO, which currently have no or limited options for treatment.
Collapse
Affiliation(s)
- Anagha Gogate
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Jordyn Belcourt
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Milan Shah
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Alicia Zongxun Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Alexis Frankel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Holly Kolmel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Matthew Chalon
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Prajith Stephen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Aarush Kolli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Sherouk M Tawfik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Jing Jin
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Raman Bahal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Theodore P Rasmussen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - José E Manautou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| | - Xiao-Bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
83
|
Reijman M, Defesche J, Wiegman A. Genotype-phenotype correlation in a large cohort of pediatric patients with heterozygous and homozygous familial hypercholesterolemia. Curr Opin Lipidol 2023; 34:287-295. [PMID: 36752612 PMCID: PMC10624405 DOI: 10.1097/mol.0000000000000863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is a genetic disorder characterized by elevated low-density lipoprotein cholesterol (LDL-C) levels and premature cardiovascular disease (CVD). Both the heterozygous form and the very severe homozygous form can be diagnosed by genetic testing and by clinical criteria. Genetic testing can discern FH in a form caused by complete absence of the LDL-receptors, the negative variant and a form leading to reduced activity of the LDL receptors, the defective variant. The aim of this study is to provide more insight in the genotype-phenotype correlation in children and adolescents diagnosed with heterozygous FH (HeFH) and with homozygous FH (HoFH), specifically in relation to the clinical and therapeutic consequences of the negative and defective variant of FH. METHODS AND RESULTS Data of 5904 children with a tentative diagnosis of FH referred to our center for genetic testing were collected. A lipid-profile was present in 3494 children, who became the study cohort. In this large cohort of children, which includes 2714 HeFH and 41 HoFH patients, it is shown that receptor negative variants are associated with significant higher LDL-C levels in HeFH patients than receptor defective variants (6.0 versus 4.9 mmol/L; p < 0.001). A negative/negative variant is associated with a significant higher LDL-C level jn HoFH patients than a negative/defective variant, which in itself has a higher LDL-C level than a defective/defective variant. Significantly more premature CVD is present in close relatives of children with HeFH with negative variants compared to close relatives of HeFH children with defective variants (75% vs 59%; p < 0.001). CONCLUSIONS Performing genetic testing and identifying the type of underlying genetic variant is of added value in order to distinguish between pediatric patients with higher risks of premature CVD and to identify those that will benefit most from new types of lipid-lowering therapies. Since in children the phenotype of FH is less affected by environmental factors, the study substantiates the genotype-phenotype correlation in this large pediatric population.
Collapse
Affiliation(s)
| | - J.C. Defesche
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
84
|
Watts GF, Gidding SS, Hegele RA, Raal FJ, Sturm AC, Jones LK, Sarkies MN, Al-Rasadi K, Blom DJ, Daccord M, de Ferranti SD, Folco E, Libby P, Mata P, Nawawi HM, Ramaswami U, Ray KK, Stefanutti C, Yamashita S, Pang J, Thompson GR, Santos RD. International Atherosclerosis Society guidance for implementing best practice in the care of familial hypercholesterolaemia. Nat Rev Cardiol 2023; 20:845-869. [PMID: 37322181 DOI: 10.1038/s41569-023-00892-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
This contemporary, international, evidence-informed guidance aims to achieve the greatest good for the greatest number of people with familial hypercholesterolaemia (FH) across different countries. FH, a family of monogenic defects in the hepatic LDL clearance pathway, is a preventable cause of premature coronary artery disease and death. Worldwide, 35 million people have FH, but most remain undiagnosed or undertreated. Current FH care is guided by a useful and diverse group of evidence-based guidelines, with some primarily directed at cholesterol management and some that are country-specific. However, none of these guidelines provides a comprehensive overview of FH care that includes both the lifelong components of clinical practice and strategies for implementation. Therefore, a group of international experts systematically developed this guidance to compile clinical strategies from existing evidence-based guidelines for the detection (screening, diagnosis, genetic testing and counselling) and management (risk stratification, treatment of adults or children with heterozygous or homozygous FH, therapy during pregnancy and use of apheresis) of patients with FH, update evidence-informed clinical recommendations, and develop and integrate consensus-based implementation strategies at the patient, provider and health-care system levels, with the aim of maximizing the potential benefit for at-risk patients and their families worldwide.
Collapse
Affiliation(s)
- Gerald F Watts
- School of Medicine, University of Western Australia, Perth, WA, Australia.
- Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia.
| | | | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine, Western University, London, ON, Canada
| | - Frederick J Raal
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amy C Sturm
- Department of Genomic Health, Geisinger, Danville, PA, USA
- 23andMe, Sunnyvale, CA, USA
| | - Laney K Jones
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Mitchell N Sarkies
- School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Khalid Al-Rasadi
- Medical Research Centre, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Dirk J Blom
- Division of Lipidology and Cape Heart Institute, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | | | | | | | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Hapizah M Nawawi
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM) and Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
- Specialist Lipid and Coronary Risk Prevention Clinics, Hospital Al-Sultan Abdullah (HASA) and Clinical Training Centre, Puncak Alam and Sungai Buloh Campuses, Universiti Teknologi MARA, Sungai Buloh, Selangor, Malaysia
| | - Uma Ramaswami
- Royal Free London NHS Foundation Trust, University College London, London, UK
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Imperial College London, London, UK
| | - Claudia Stefanutti
- Department of Molecular Medicine, Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Regional Centre for Rare Diseases, Immunohematology and Transfusion Medicine, Umberto I Hospital, 'Sapienza' University of Rome, Rome, Italy
| | - Shizuya Yamashita
- Department of Cardiology, Rinku General Medical Center, Osaka, Japan
| | - Jing Pang
- School of Medicine, University of Western Australia, Perth, WA, Australia
| | | | - Raul D Santos
- Lipid Clinic, Heart Institute (InCor), University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
85
|
Gu J, Kuznik A, Quon P, Chauhan A, Sravya TS, Raal FJ. Modelling the potential long-term survival benefit of evinacumab treatment vs. standard of care in patients with homozygous familial hypercholesterolaemia. Eur J Prev Cardiol 2023; 30:1874-1880. [PMID: 37314419 DOI: 10.1093/eurjpc/zwad203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/15/2023]
Abstract
AIMS Despite intensive lipid-lowering therapies (LLTs), most patients with homozygous familial hypercholesterolaemia (HoFH) do not achieve guideline recommended low-density lipoprotein cholesterol (LDL-C) targets and are at increased risk of premature cardiovascular death. This analysis aimed to predict the impact of evinacumab and standard-of-care LLTs on life expectancy in an HoFH population using mathematical modelling. METHODS AND RESULTS Mathematical models were developed using efficacy data for evinacumab from the phase 3 ELIPSE HoFH trial plus efficacy data for standard-of-care LLTs from peer-reviewed publications. Treatment strategies evaluated included (i) untreated, (ii) high-intensity statin (HIS) only, (iii) HIS plus ezetimibe, (iv) HIS plus ezetimibe plus proprotein convertase subtilisin/kexin type 9 inhibitor (PCSK9i), and (v) HIS plus ezetimibe plus PCSK9i plus evinacumab. Markov analyses were used to assess differences in survival probability for different LLT strategies. The median survival for untreated HoFH patients was only 33-43 years, depending on different assumptions on baseline untreated LDL-C levels. In the most robust model, we estimated that HIS increased median survival by 9 years and ezetimibe further increased median survival by an additional 9 years. When PCSK9i was added on top of HIS plus ezetimibe, median survival was further improved by 14 years. Finally, the addition of evinacumab to standard-of-care LLTs was estimated to increase median survival by ∼12 years. CONCLUSION In this mathematical modelling analysis, evinacumab treatment could potentially increase long-term survival vs. standard-of-care LLTs for patients with HoFH.
Collapse
Affiliation(s)
- Jing Gu
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill, River Road, Tarrytown, NY 10591, USA
| | - Andreas Kuznik
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill, River Road, Tarrytown, NY 10591, USA
| | - Peter Quon
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill, River Road, Tarrytown, NY 10591, USA
| | | | | | - Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
86
|
Collotta D, Bertocchi I, Chiapello E, Collino M. Antisense oligonucleotides: a novel Frontier in pharmacological strategy. Front Pharmacol 2023; 14:1304342. [PMID: 38044945 PMCID: PMC10690781 DOI: 10.3389/fphar.2023.1304342] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Antisense oligonucleotides (ASOs) are short single stranded synthetic RNA or DNA molecules, whereas double-stranded RNA nucleotide sequences are called small interfering RNA (siRNA). ASOs bind to complementary nucleic acid sequences impacting the associated functions of the targeted nucleic acids. They represent an emerging class of drugs that, through a revolutionary mechanism of action, aim to directly regulate disease-causing genes and their variants, providing an alternative tool to traditional "protein-specific" therapies. The majority of the ASOs are designed to treat orphan genetic disorders that in most of the cases are seriously disabling and still lacking an adequate therapy. In order to translate ASOs into clinical success, constant technological advances have been instrumental in overcoming several pharmacological, toxicological and formulation limitations. Accordingly, chemical structures have been recently implemented and new bio-conjugation and nanocarriers formulation strategies explored. The aim of this work is to offer an overview of the antisense technology with a comparative analysis of the oligonucleotides approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA).
Collapse
Affiliation(s)
- D. Collotta
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - I. Bertocchi
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
| | - E. Chiapello
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - M. Collino
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| |
Collapse
|
87
|
Reijman MD, Kusters DM, Groothoff JW, Arbeiter K, Dann EJ, de Boer LM, de Ferranti SD, Gallo A, Greber-Platzer S, Hartz J, Hudgins LC, Ibarretxe D, Kayikcioglu M, Klingel R, Kolovou GD, Oh J, Planken RN, Stefanutti C, Taylan C, Wiegman A, Schmitt CP. Clinical practice recommendations on lipoprotein apheresis for children with homozygous familial hypercholesterolemia: an expert consensus statement from ERKNet and ESPN. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.14.23298547. [PMID: 38014132 PMCID: PMC10680892 DOI: 10.1101/2023.11.14.23298547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Homozygous familial hypercholesterolaemia is a life-threatening genetic condition, which causes extremely elevated LDL-C levels and atherosclerotic cardiovascular disease very early in life. It is vital to start effective lipid-lowering treatment from diagnosis onwards. Even with dietary and current multimodal pharmaceutical lipid-lowering therapies, LDL-C treatment goals cannot be achieved in many children. Lipoprotein apheresis is an extracorporeal lipid-lowering treatment, which is well established since three decades, lowering serum LDL-C levels by more than 70% per session. Data on the use of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia mainly consists of case-reports and case-series, precluding strong evidence-based guidelines. We present a consensus statement on lipoprotein apheresis in children based on the current available evidence and opinions from experts in lipoprotein apheresis from over the world. It comprises practical statements regarding the indication, methods, treatment targets and follow-up of lipoprotein apheresis in children with homozygous familial hypercholesterolaemia and on the role of lipoprotein(a) and liver transplantation.
Collapse
Affiliation(s)
- M. Doortje Reijman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - D. Meeike Kusters
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Jaap W. Groothoff
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Klaus Arbeiter
- Division of Paediatric Nephrology and Gastroenterology, Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Eldad J. Dann
- Blood Bank and apheresis unit Rambam Health care campus, Haifa, Israel
| | - Lotte M. de Boer
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Sarah D. de Ferranti
- Department of Cardiology, Boston Children’s Hospital, Boston, MA, USA, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Antonio Gallo
- Sorbonne Université, INSERM, UMR 1166, Lipidology and cardiovascular prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpêtrière F-75013 Paris, France
| | - Susanne Greber-Platzer
- Clinical Division of Paediatric Pulmonology, Allergology and Endocrinology, Department of Paediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - Jacob Hartz
- Department of Cardiology, Boston Children’s Hospital, Boston, MA, USA, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Lisa C. Hudgins
- The Rogosin Institute, Weill Cornell Medical College, New York, New York, USA
| | - Daiana Ibarretxe
- Vascular Medicine and Metabolism Unit (UVASMET), Hospital Universitari Sant Joan; Universitat Rovira i Virgili; Institut Investigació Sanitària Pere Virgili (IISPV)-CERCA, Spain; Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Meral Kayikcioglu
- Department of Cardiology, Medical Faculty, Ege University, 35100 Izmir, Turkey
| | - Reinhard Klingel
- Apheresis Research Institute, Stadtwaldguertel 77, 50935 Cologne, Germany (www.apheresis-research.org)
| | - Genovefa D. Kolovou
- Metropolitan Hospital, Department of Preventive Cardiology. 9, Ethn. Makariou & 1, El. Venizelou, N. Faliro, 185 47, Athens, Greece
| | - Jun Oh
- University Medical Center Hamburg/Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - R. Nils Planken
- Department of Radiology and nuclear medicine, Amsterdam UMC, location AMC, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
| | - Claudia Stefanutti
- Department of Molecular Medicine, Lipid Clinic and Atherosclerosis Prevention Centre, ‘Umberto I’ Hospital ‘Sapienza’ University of Rome, I-00161 Rome, Italy
| | - Christina Taylan
- Paediatric Nephrology, Children’s and Adolescents’ Hospital, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Albert Wiegman
- Amsterdam UMC, University of Amsterdam, Department of Paediatrics, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, Amsterdam, Netherlands
| | - Claus Peter Schmitt
- Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, Germany
| |
Collapse
|
88
|
Mirzai S, Chevli PA, Rikhi R, Shapiro MD. Familial Hypercholesterolemia: From Clinical Suspicion to Novel Treatments. Rev Cardiovasc Med 2023; 24:311. [PMID: 39076456 PMCID: PMC11272857 DOI: 10.31083/j.rcm2411311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2024] Open
Abstract
Familial hypercholesterolemia (FH) is the most common monogenic disorder in humans. It affects millions of people globally, increasing the risk of developing cardiovascular disease (CVD) at a younger age due to elevated levels of low-density lipoprotein cholesterol (LDL-C) from birth. While effective traditional and novel treatments are available, the most significant challenge with FH is the lack of timely diagnosis. As a result, many patients remain undertreated leading to an increased risk of CVD. To mitigate risk, initiating early and aggressive LDL-C-lowering therapies is recommended. Moreover, given its autosomal dominant inheritance patterns, it is also recommended to perform cascade lipid and/or genetic testing of all first-degree relatives. This review highlights the importance of early FH diagnosis and available treatment options. Greater awareness and improved screening efforts can help diagnose and treat more individuals, ultimately reducing the CVD risk associated with FH.
Collapse
Affiliation(s)
- Saeid Mirzai
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH 44195,
USA
| | - Parag Anilkumar Chevli
- Section on Hospital Medicine, Department of Internal Medicine, Wake Forest
University School of Medicine, Winston-Salem, NC 27157, USA
| | - Rishi Rikhi
- Section on Cardiovascular Medicine, Department of Internal Medicine, Wake
Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Michael D. Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake
Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
89
|
Raal FJ, Rosenson RS, Reeskamp LF, Kastelein JJ, Rubba P, Duell PB, Koseki M, Stroes E, Ali S, Banerjee P, Chan KC, Khilla N, McGinniss J, Pordy R, Zhang Y, Gaudet D. The Long-Term Efficacy and Safety of Evinacumab in Patients With Homozygous Familial Hypercholesterolemia. JACC. ADVANCES 2023; 2:100648. [PMID: 38938723 PMCID: PMC11198175 DOI: 10.1016/j.jacadv.2023.100648] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/11/2023] [Accepted: 08/22/2023] [Indexed: 06/29/2024]
Abstract
Background Homozygous familial hypercholesterolemia (HoFH) is characterized by early-onset atherosclerotic cardiovascular disease due to the high low-density lipoprotein cholesterol (LDL-C) burden. Patients with null-null low-density lipoprotein receptor (LDLR) variants respond poorly, if at all, to statins and proprotein convertase subtilisin/kexin type 9 inhibitors, which act by upregulating LDLR expression. The 24-week double-blind treatment period (DBTP) of the phase 3 ELIPSE HoFH (Evinacumab Lipid Studies in Patients with Homozygous Familial hypercholesterolemia; NCT03399786) study demonstrated significant LDL-C reductions in patients with HoFH; LDL-C reductions were also observed in those with null-null LDLR mutations. Objectives The purpose of this study was to evaluate longer-term efficacy and safety of evinacumab in patients with HoFH from the ELIPSE HoFH study. Methods Patients with HoFH on stable lipid-lowering therapies (LLTs) ± lipoprotein apheresis and screening LDL-C ≥70 mg/dL who completed the DBTP entered the 24-week open-label treatment period (OLTP) and received intravenous evinacumab 15 mg/kg every 4 weeks. OLTP results were summarized descriptively. Results A total of 64 patients completed the DBTP and received open-label evinacumab. Despite multiple LLTs, the mean baseline LDL-C at DBTP entry was 250.5 ± 162.3 mg/dL. From baseline to week 48 (end of OLTP), evinacumab reduced mean LDL-C by 46.3% (mean reduction, 134.3 ± 117.3 mg/dL), with similar mean LDL-C reductions for patients with null-null (47.2%) and non-null variants (45.9%). Adverse events occurred in 47 (73.4%) patients; 4 (6.3%) patients experienced adverse events considered evinacumab-related (drug hypersensitivity, infusion-related reaction and asthenia, generalized pruritis, and muscle spasms). Conclusions In patients with HoFH, evinacumab demonstrated substantial and sustained LDL-C reduction regardless of LDLR function, and was generally well tolerated.
Collapse
Affiliation(s)
- Frederick J. Raal
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Robert S. Rosenson
- Cardiometabolics Unit, Zena and Michael A Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Laurens F. Reeskamp
- Department of Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - John J.P. Kastelein
- Department of Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Paolo Rubba
- Department of Internal Medicine and Surgery, Federico II University, Naples, Italy
| | - P. Barton Duell
- Knight Cardiovascular Institute and Division of Endocrinology, Diabetes and Clinical Nutrition, Oregon Health & Science University, Portland, Oregon, USA
| | - Masahiro Koseki
- Division of Cardiovascular Medicine, Department of Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Erik Stroes
- Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Shazia Ali
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | | | - Kuo-Chen Chan
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Nagwa Khilla
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | | | - Robert Pordy
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Yi Zhang
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Daniel Gaudet
- Clinical Lipidology and Rare Lipid Disorders Unit, Department of Medicine, Université de Montréal Community Gene Medicine Center, Lipid Clinic Chicoutimi Hospital and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada
| |
Collapse
|
90
|
Klevmoen M, Mulder JWCM, Roeters van Lennep JE, Holven KB. Sex Differences in Familial Hypercholesterolemia. Curr Atheroscler Rep 2023; 25:861-868. [PMID: 37815650 PMCID: PMC10618303 DOI: 10.1007/s11883-023-01155-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/11/2023]
Abstract
PURPOSE OF REVIEW This review aims to summarize the existing research on sex differences in familial hypercholesterolemia (FH) across the lifespan. RECENT FINDINGS From childhood onward, total- and low-density lipoprotein cholesterol (LDL-C) levels in girls are higher than those in boys with FH. By the age of 30 years, women with FH have a higher LDL-C burden than men. In adulthood, women are diagnosed later than men, receive less lipid-lowering treatment, and consequently have higher LDL-C levels. An excessive atherosclerotic cardiovascular disease risk is reported in young female compared to male FH patients. The periods of pregnancy and breastfeeding contribute to treatment loss and increased cholesterol burden. Earlier initiation of treatment, especially in girls with FH, and lifelong treatment during all life stages are important. Future research should aim to recruit both women and men, report sex-specific data, and investigate the impact of the female life course on cardiovascular outcomes. Future guidelines should include sex-specific aspects.
Collapse
Affiliation(s)
- Marianne Klevmoen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Oslo, Norway
| | - Janneke W C M Mulder
- Department of Internal Medicine, Cardiovascular Institute, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Cardiovascular Institute, Erasmus MC Cardiovascular Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Kirsten B Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
91
|
Lv X, Wang C, Liu L, Yin G, Zhang W, Abdu FA, Shi T, Zhang Q, Che W. Screening and verifying the mutations in the LDLR and APOB genes in a Chinese family with familial hypercholesterolemia. Lipids Health Dis 2023; 22:175. [PMID: 37853441 PMCID: PMC10585857 DOI: 10.1186/s12944-023-01935-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/02/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is an autosomal dominant genetic disorder. The primary objective of this study was to identify the major pathogenic mutations in a Chinese family with FH. METHODS Whole-genome sequencing (WGS) was used to identify variants of FH-related genes, including low-density lipoprotein receptor (LDLR), apolipoprotein B (APOB), and proprotein convertase subtilisin/kexin 9 (PCSK9). Bioinformatics software was used to predict signal peptides, transmembrane structures, and spatial construction information of the mutated sequences. Western blotting was performed on the mutant protein to determine the presence of the major structural domains of the LDLR. The PCSK9 and APOB genes were screened and analyzed. Moreover, the proband and his brother were treated with a PCSK9 inhibitor for 1 year, and the effect of the treatment on lipid levels was assessed. RESULTS WGS revealed two potentially pathogenic mutations in the LDLR gene. One was a novel mutation, c.497delinsGGATCCCCCAGCTGCATCCCCCAG (p. Ala166fs), and the other was a known pathogenic mutation, c.2054C>T (p. Pro685Leu). Bioinformatics prediction and in vitro experiments revealed that the novel mutation could not be expressed on the cell membrane. Numerous gene variants were identified in the APOB gene that may have a significant impact on the family members with FH. Thus, it is suggested that the severe manifestation of FH in the proband primarily resulted from the cumulative genetic effects of variants in both LDLR and APOB. However, a subsequent study indicated that treatment with a PCSK9 inhibitor (Evolocumab) did not significantly reduce the blood lipid levels in the proband or his brother. CONCLUSIONS The cumulative effect of LDLR and APOB variants was the primary cause of elevated blood lipid levels in this family. However, PCSK9 inhibitor therapy did not appear to be beneficial for the proband. This study emphasizes the importance of genetic testing in determining the most suitable treatment options for patients with FH.
Collapse
Affiliation(s)
- Xian Lv
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Chunyue Wang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Lu Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Guoqing Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Wen Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Fuad A Abdu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Tingting Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Qingfeng Zhang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital, Clinical Center for Brain and Spinal Cord Research, School of Medicine, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China.
- Department of Cardiology, Shanghai Tenth People's Hospital Chongming Branch, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
92
|
Luca AC, David SG, David AG, Țarcă V, Pădureț IA, Mîndru DE, Roșu ST, Roșu EV, Adumitrăchioaiei H, Bernic J, Cojocaru E, Țarcă E. Atherosclerosis from Newborn to Adult-Epidemiology, Pathological Aspects, and Risk Factors. Life (Basel) 2023; 13:2056. [PMID: 37895437 PMCID: PMC10608492 DOI: 10.3390/life13102056] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity throughout the world, accounting for 16.7 million deaths each year. The underlying pathological process for the majority of cardiovascular diseases is atherosclerosis, a slowly progressing, multifocal, chronic, immune-inflammatory disease that involves the intima of large and medium-sized arteries. The process of atherosclerosis begins in childhood as fatty streaks-an accumulation of lipids, inflammatory cells, and smooth muscle cells in the arterial wall. Over time, a more complex lesion develops into an atheroma and characteristic fibrous plaques. Atherosclerosis alone is rarely fatal; it is the further changes that render fibrous plaques vulnerable to rupture; plaque rupture represents the most common cause of coronary thrombosis. The prevalence of atherosclerosis is increasing worldwide and more than 50% of people with circulatory disease die of it, mostly in modern societies. Epidemiological studies have revealed several environmental and genetic risk factors that are associated with the early formation of a pathogenic foundation for atherosclerosis, such as dyslipidemia, hypertension, diabetes mellitus, obesity, and smoking. The purpose of this review is to bring together the current information concerning the origin and progression of atherosclerosis in childhood as well as the identification of known risk factors for atherosclerotic cardiovascular disease in children.
Collapse
Affiliation(s)
- Alina Costina Luca
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Simona Georgiana David
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Alexandru Gabriel David
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Viorel Țarcă
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana-Alexandra Pădureț
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Dana Elena Mîndru
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Solange Tamara Roșu
- Nursing Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Eduard Vasile Roșu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.C.L.); (D.E.M.); (E.V.R.)
| | - Heidrun Adumitrăchioaiei
- Saint Mary Emergency Hospital for Children, 700309 Iasi, Romania; (S.G.D.); (A.G.D.); (I.-A.P.); (H.A.)
| | - Jana Bernic
- Discipline of Pediatric Surgery, “Nicolae Testemițanu” State University of Medicine and Pharmacy, 2025 Chisinau, Moldova;
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I—Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Elena Țarcă
- Surgery II Department—Pediatric Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| |
Collapse
|
93
|
Pirillo A, Casula M, Catapano AL. European guidelines for the treatment of dyslipidaemias: New concepts and future challenges. Pharmacol Res 2023; 196:106936. [PMID: 37739143 DOI: 10.1016/j.phrs.2023.106936] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 09/24/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality and morbidity worldwide. Low-density lipoprotein cholesterol (LDL-C) is one of the most important causal factors for ASCVD. Based on the evidence of the clinical benefits of lowering LDL-C, the current 2019 European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) guidelines provide guidance for optimal management of people with dyslipidaemia. These guidelines include new and revised concepts, with a general tightening of LDL-C goals to be achieved, especially for patients at high and very high cardiovascular risk, based on the results of clinical trials of the recently approved drugs for the treatment of hypercholesterolaemia. However, some issues are still open for discussion. Among others, the concept of lifetime exposure to elevated LDL-C levels will probably drive the pharmacological approach and future guidelines. In addition, other factors such as non-HDL-C, apolipoprotein B, and lipoprotein(a) are becoming increasingly important in determining cardiovascular risk. Finally, there is the question of whether combination therapy should be used as the first step to maximise the effectiveness of the pharmacological approach, avoiding the stepwise approach, which is likely to have a detrimental effect on adherence. Given the ever-changing landscape and the availability of new drugs targeting other important lipids, future guidelines will need to consider all these issues.
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - Manuela Casula
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Alberico L Catapano
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy.
| |
Collapse
|
94
|
LI JJ, ZHAO SP, ZHAO D, LU GP, PENG DQ, LIU J, CHEN ZY, GUO YL, WU NQ, YAN SK, WANG ZW, GAO RL. 2023 China Guidelines for Lipid Management. J Geriatr Cardiol 2023; 20:621-663. [PMID: 37840633 PMCID: PMC10568545 DOI: 10.26599/1671-5411.2023.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death among urban and rural residents in China, and elevated low-density lipoprotein cholesterol (LDL-C) is a risk factor for ASCVD. Considering the increasing burden of ASCVD, lipid management is of the utmost importance. In recent years, research on blood lipids has made breakthroughs around the world, hence a revision of China guidelines for lipid management is imperative, especially since the target lipid levels in the general population vary in respect to the risk of ASCVD. The level of LDL-C, which can be regarded as appropriate in a population without frisk factors, can be considered abnormal in people at high risk of developing ASCVD. As a result, the "Guidelines for the prevention and treatment of dyslipidemia" were adapted into the "China Guidelines for Lipid Management" (henceforth referred to as the new guidelines) by an Experts' committee after careful deliberation. The new guidelines still recommend LDL-C as the primary target for lipid control, with CVD risk stratification to determine its target value. These guidelines recommend that moderate intensity statin therapy in adjunct with a heart-healthy lifestyle, be used as an initial line of treatment, followed by cholesterol absorption inhibitors or/and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, as necessary. The new guidelines provide guidance for lipid management across various age groups, from children to the elderly. The aim of these guidelines is to comprehensively improve the management of lipids and promote the prevention and treatment of ASCVD by guiding clinical practice.
Collapse
Affiliation(s)
- Jian-Jun LI
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shui-Ping ZHAO
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dong ZHAO
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guo-Ping LU
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dao-Quan PENG
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jing LIU
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhen-Yue CHEN
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuan-Lin GUO
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Na-Qiong WU
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng-Kai YAN
- Affiliated Hospital of Zunyi Medical University, School of Laboratory Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zeng-Wu WANG
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Run-Lin GAO
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
95
|
Luo RF, Wang JH, Hu LJ, Fu QA, Zhang SY, Jiang L. Applications of machine learning in familial hypercholesterolemia. Front Cardiovasc Med 2023; 10:1237258. [PMID: 37823179 PMCID: PMC10562581 DOI: 10.3389/fcvm.2023.1237258] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Familial hypercholesterolemia (FH) is a common hereditary cholesterol metabolic disease that usually leads to an increase in the level of low-density lipoprotein cholesterol in plasma and an increase in the risk of cardiovascular disease. The lack of disease screening and diagnosis often results in FH patients being unable to receive early intervention and treatment, which may mean early occurrence of cardiovascular disease. Thus, more requirements for FH identification and management have been proposed. Recently, machine learning (ML) has made great progress in the field of medicine, including many innovative applications in cardiovascular medicine. In this review, we discussed how ML can be used for FH screening, diagnosis and risk assessment based on different data sources, such as electronic health records, plasma lipid profiles and corneal radian images. In the future, research aimed at developing ML models with better performance and accuracy will continue to overcome the limitations of ML, provide better prediction, diagnosis and management tools for FH, and ultimately achieve the goal of early diagnosis and treatment of FH.
Collapse
Affiliation(s)
- Ren-Fei Luo
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing-Hui Wang
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, Nanchang University Queen Mary School, Nanchang, China
| | - Li-Juan Hu
- Department of Nursing, Nanchang Medical College, Nanchang, China
| | - Qing-An Fu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Si-Yi Zhang
- Department of Clinical Medicine, Nanchang University Queen Mary School, Nanchang, China
| | - Long Jiang
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
96
|
Aldisi R, Hassanin E, Sivalingam S, Buness A, Klinkhammer H, Mayr A, Fröhlich H, Krawitz P, Maj C. Gene-based burden scores identify rare variant associations for 28 blood biomarkers. BMC Genom Data 2023; 24:50. [PMID: 37667186 PMCID: PMC10476296 DOI: 10.1186/s12863-023-01155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 08/28/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND A relevant part of the genetic architecture of complex traits is still unknown; despite the discovery of many disease-associated common variants. Polygenic risk score (PRS) models are based on the evaluation of the additive effects attributable to common variants and have been successfully implemented to assess the genetic susceptibility for many phenotypes. In contrast, burden tests are often used to identify an enrichment of rare deleterious variants in specific genes. Both kinds of genetic contributions are typically analyzed independently. Many studies suggest that complex phenotypes are influenced by both low effect common variants and high effect rare deleterious variants. The aim of this paper is to integrate the effect of both common and rare functional variants for a more comprehensive genetic risk modeling. METHODS We developed a framework combining gene-based scores based on the enrichment of rare functionally relevant variants with genome-wide PRS based on common variants for association analysis and prediction models. We applied our framework on UK Biobank dataset with genotyping and exome data and considered 28 blood biomarkers levels as target phenotypes. For each biomarker, an association analysis was performed on full cohort using gene-based scores (GBS). The cohort was then split into 3 subsets for PRS construction and feature selection, predictive model training, and independent evaluation, respectively. Prediction models were generated including either PRS, GBS or both (combined). RESULTS Association analyses of the cohort were able to detect significant genes that were previously known to be associated with different biomarkers. Interestingly, the analyses also revealed heterogeneous effect sizes and directionality highlighting the complexity of the blood biomarkers regulation. However, the combined models for many biomarkers show little or no improvement in prediction accuracy compared to the PRS models. CONCLUSION This study shows that rare variants play an important role in the genetic architecture of complex multifactorial traits such as blood biomarkers. However, while rare deleterious variants play a strong role at an individual level, our results indicate that classical common variant based PRS might be more informative to predict the genetic susceptibility at the population level.
Collapse
Affiliation(s)
- Rana Aldisi
- Institute of Genomic Statistic and Bioinformatics, University Hospital Bonn, Bonn, Germany.
| | - Emadeldin Hassanin
- Institute of Genomic Statistic and Bioinformatics, University Hospital Bonn, Bonn, Germany
- Luxembourg Center for Systems Biomedicine, University of Luxembourg, Esch-Sur-Alzette, Luxembourg
| | - Sugirthan Sivalingam
- Institute of Genomic Statistic and Bioinformatics, University Hospital Bonn, Bonn, Germany
- Core Unit for Bioinformatics Analysis, University Hospital Bonn, Bonn, Germany
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Andreas Buness
- Institute of Genomic Statistic and Bioinformatics, University Hospital Bonn, Bonn, Germany
- Core Unit for Bioinformatics Analysis, University Hospital Bonn, Bonn, Germany
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Hannah Klinkhammer
- Institute of Genomic Statistic and Bioinformatics, University Hospital Bonn, Bonn, Germany
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Andreas Mayr
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Holger Fröhlich
- Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, Germany
- Bonn-Aachen International Center for IT (b-it), University of Bonn, Bonn, Germany
| | - Peter Krawitz
- Institute of Genomic Statistic and Bioinformatics, University Hospital Bonn, Bonn, Germany
| | - Carlo Maj
- Institute of Genomic Statistic and Bioinformatics, University Hospital Bonn, Bonn, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
| |
Collapse
|
97
|
Brandts J, Ray KK. Novel and future lipid-modulating therapies for the prevention of cardiovascular disease. Nat Rev Cardiol 2023; 20:600-616. [PMID: 37055535 DOI: 10.1038/s41569-023-00860-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/15/2023]
Abstract
Lowering the levels of LDL cholesterol in the plasma has been shown to reduce the risk of atherosclerotic cardiovascular disease (ASCVD). Several other lipoproteins, such as triglyceride-rich lipoproteins, HDL and lipoprotein(a) are associated with atherosclerosis and ASCVD, with strong evidence supporting causality for some. In this Review, we discuss novel and upcoming therapeutic strategies targeting different pathways in lipid metabolism to potentially attenuate the risk of cardiovascular events. Key proteins involved in lipoprotein metabolism, such as PCSK9, angiopoietin-related protein 3, cholesteryl ester transfer protein and apolipoprotein(a), have been identified as viable targets for therapeutic intervention through observational and genetic studies. These proteins can be targeted using a variety of approaches, such as protein inhibition or interference, inhibition of translation at the mRNA level (with the use of antisense oligonucleotides or small interfering RNA), and the introduction of loss-of-function mutations through base editing. These novel and upcoming strategies are complementary to and could work synergistically with existing therapies, or in some cases could potentially replace therapies, offering unprecedented opportunities to prevent ASCVD. Moreover, a major challenge in the prevention and treatment of non-communicable diseases is how to achieve safe, long-lasting reductions in causal exposures. This challenge might be overcome with approaches such as small interfering RNAs or genome editing, which shows how far the field has advanced from when the burden of achieving this goal was placed upon patients through rigorous adherence to daily small-molecule drug regimens.
Collapse
Affiliation(s)
- Julia Brandts
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
98
|
Clegg K, Schubert TJ, Block RC, Burke F, Desai NR, Greenfield R, Karalis D, Kris-Etherton PM, McNeal CJ, Nahrwold R, Peña JM, Plakogiannis R, Wong ND, Jones LK. Translating Evidence-based Approaches into optimal Care for individuals at High-risk of ASCVD: Pilot testing of case-based e-learning modules and design of the TEACH-ASCVD study. J Clin Lipidol 2023; 17:592-601. [PMID: 37550150 DOI: 10.1016/j.jacl.2023.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of death in the United States. Case-based learning using electronic delivery of the modules can educate clinicians and improve translation of evidence-based guidelines into practice for high-risk ASCVD patients. OBJECTIVE To develop and optimize module design, content, and usability of e-learning modules to teach clinicians evidence-based management in accordance with multi-society guidelines for high-risk ASCVD patients that will be implemented and evaluated in U.S. health systems in the TEACH-ASCVD study. METHODS Seven e-learning modules were created by a committee of lipid experts. Focus groups were conducted with lipid experts to elicit feedback on case content followed by interviews with a target audience of clinicians to assess usability of the online module platform. Responses from both groups were evaluated, and appropriate changes were made to improve the e-learning modules. Design of the TEACH-ASCVD study is presented. RESULTS Feedback regarding case content by lipid experts included providing more detailed patient histories, clarifying various diagnostic criteria, and emphasizing clinical best practices based on evidence-based guidelines. The target audience clinician group reported an agreeable experience with the e-learning modules but noted a discordance between the evidence-based guidelines and clinical decision-making in their own practices. Participants felt the modules would help educate clinicians in managing high-risk ASCVD patients. CONCLUSION Clinicians must be informed of best practices as the field of lipidology continues to evolve. E-learning modules provide a concise, valuable, and accessible mechanism for educating clinicians regarding changes in the field to deliver the best patient care.
Collapse
Affiliation(s)
- Katarina Clegg
- Department of Genomic Health, Geisinger, Danville, PA 17822, United States; Geisinger Commonwealth School of Medicine, Scranton, PA, 18510, United States
| | - Tyler J Schubert
- Department of Genomic Health, Geisinger, Danville, PA 17822, United States; Geisinger Commonwealth School of Medicine, Scranton, PA, 18510, United States
| | - Robert C Block
- Department of Public Health; Cardiology Division, Department of Medicine; University of Rochester Medical Center, NY, 14642, United States
| | - Frances Burke
- Division of Cardiology, University of Pennsylvania Health System, Philadelphia, PA, 19104, United States
| | - Nihar R Desai
- Section of Cardiovascular Medicine, Department of Medicine, Yale School of Medicine, New Haven, CT, 06510, United States
| | - Robert Greenfield
- Division of Cardiology, University of California, Irvine School of Medicine, Orange County, CA 92868, United States
| | - Dean Karalis
- Division of Cardiology, Thomas Jefferson University Hospital, Philadelphia, PA 19107, United States
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, Penn State University, University Park, PA, 16802, United States
| | - Catherine J McNeal
- Department of Internal Medicine, Baylor Scott & White Health, Temple, TX, 76502, United States
| | - Rachel Nahrwold
- Lenox Hill Hospital, Northwell Health, New York, NY, 10075, United States
| | - Jessica M Peña
- Departments of Radiology and Medicine, Weill Cornell Medicine, New York, NY 10065, United States
| | - Roda Plakogiannis
- Transdermal Research Pharm Laboratories, Long Island City, NY, 11101, United States
| | - Nathan D Wong
- Division of Cardiology, University of California, Irvine School of Medicine, Orange County, CA 92868, United States
| | - Laney K Jones
- Department of Genomic Health, Geisinger, Danville, PA 17822, United States; Heart and Vascular Institute, Geisinger, Danville, PA, 17822, United States.
| |
Collapse
|
99
|
Li JJ, Zhao SP, Zhao D, Lu GP, Peng DQ, Liu J, Chen ZY, Guo YL, Wu NQ, Yan SK, Wang ZW, Gao RL. 2023 Chinese guideline for lipid management. Front Pharmacol 2023; 14:1190934. [PMID: 37711173 PMCID: PMC10498001 DOI: 10.3389/fphar.2023.1190934] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/26/2023] [Indexed: 09/16/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death among urban and rural residents in China, and elevated low-density lipoprotein cholesterol (LDL-C) is a risk factor for ASCVD. Considering the increasing burden of ASCVD, lipid management is of the utmost importance. In recent years, research on blood lipids has made breakthroughs around the world, hence a revision of Chinese guideline for lipid management is imperative, especially since the target lipid levels in the general population vary in respect to the risk of ASCVD. The level of LDL-C, which can be regarded as appropriate in a population without frisk factors, can be considered abnormal in people at high risk of developing ASCVD. As a result, the "Guidelines for the prevention and treatment of dyslipidemia" were adapted into the "Chinese guideline for Lipid Management" (henceforth referred to as the new guidelines) by an Experts' committee after careful deliberation. The new guidelines still recommend LDL-C as the primary target for lipid control, with cardiovascular disease (CVD) risk stratification to determine its target value. These guidelines recommend that moderate intensity statin therapy in adjunct with a heart-healthy lifestyle, be used as an initial line of treatment, followed by cholesterol absorption inhibitors or/and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, as necessary. The new guidelines provide guidance for lipid management across various age groups, from children to the elderly. The aim of these guidelines is to comprehensively improve the management of lipids and promote the prevention and treatment of ASCVD by guiding clinical practice.
Collapse
Affiliation(s)
- Jian-Jun Li
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shui-Ping Zhao
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dong Zhao
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guo-Ping Lu
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dao-Quan Peng
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jing Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhen-Yue Chen
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuan-Lin Guo
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Na-Qiong Wu
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng-Kai Yan
- Affiliated Hospital of Zunyi Medical University, School of Laboratory Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zeng-Wu Wang
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Run-Lin Gao
- National Center for Cardiovascular Diseases, FuWai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
100
|
Sun L, Wolska A, Amar M, Zubirán R, Remaley AT. Approach to the Patient With a Suboptimal Statin Response: Causes and Algorithm for Clinical Management. J Clin Endocrinol Metab 2023; 108:2424-2434. [PMID: 36929838 PMCID: PMC10438872 DOI: 10.1210/clinem/dgad153] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/27/2023] [Accepted: 03/15/2023] [Indexed: 03/18/2023]
Abstract
CONTEXT Statins are the lipid-lowering therapy of choice for the prevention of atherosclerotic cardiovascular disease (ASCVD) but their effectiveness in lowering low-density lipoprotein cholesterol (LDL-C) can substantially differ between individuals. In this mini-review, we describe the different causes for a suboptimal statin response and an algorithm for the diagnosis and clinical management of these patients. EVIDENCE ACQUISITION A PubMed search using the terms "statin resistance," "statin sensitivity," "statin pharmacokinetics," "cardiovascular disease," and "lipid-lowering therapies" was performed. Published papers in the past 10 years that were relevant to the topic were examined to provide content for this mini-review. EVIDENCE SYNTHESIS Suboptimal lowering of LDL-C by statins is a major problem in the clinical management of patients and limits the value of this therapeutic approach. There are multiple causes of statin hyporesponsiveness with compliance being the most common explanation. Other causes, such as analytical issues with LDL-C measurement and the presence of common lipid disorders (familial hypercholesterolemia, elevated lipoprotein[a] and secondary dyslipidemias) should be excluded before considering primary statin resistance from rare genetic variants in lipoprotein-related or drug-metabolism genes. A wide variety of nonstatin lipid-lowering drugs are now available and can be added to statins to achieve more effective LDL-C lowering. CONCLUSIONS The evaluation of statin hyporesponsiveness is a multistep process that can lead to the optimization of lipid-lowering therapy for the prevention of ASCVD. It may also lead to the identification of distinct types of dyslipidemias that require specific therapies and/or the genetic screening of family members.
Collapse
Affiliation(s)
- Lufan Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcelo Amar
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rafael Zubirán
- Departamento de Endocrinología y Metabolismo de Lípidos, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|