1
|
Lamia TH, Shah-Riar P, Khanam M, Khair F, Sadat A, Tania MK, Haque SM, Saaki SS, Ferdausi A, Naurin SA, Tabassum M, Rahie RET, Hasan R. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Inhibitors as Adjunct Therapy to Statins: A New Frontier in Cardiovascular Risk Reduction. Cureus 2024; 16:e71365. [PMID: 39539858 PMCID: PMC11558015 DOI: 10.7759/cureus.71365] [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: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Lowering low-density lipoprotein cholesterol (LDL-C) plasma levels is crucial for the prevention of primary and secondary cardiovascular diseases (CVDs). Many patients struggle to obtain goal LDL-C levels, despite the availability of several lipid-lowering medications, because of limited efficaciousness and unfavorable side effects. Proprotein convertase subtilisin/kexin type 9 (PCSK9) targeting has drawn interest recently as a novel approach to further lower cardiovascular (CV) risk. The number of receptors accessible to remove LDL-C from the bloodstream is reduced when PCSK9 attaches to LDL-C receptors and directs them toward lysosomal destruction. LDL receptor activity is increased by PCSK9 inhibition, which attracts therapeutic intervention. Despite concurrent statin therapy, phase 3 clinical trials have demonstrated encouraging outcomes with monoclonal antibodies against PCSK9, such as evolocumab and alirocumab, resulting in significant reductions in LDL-C levels. This study intends to investigate recent advancements in the field to evaluate PCSK9 inhibitors' safety, effectiveness, and potential for preventing CVD. The investigation will also review potential future paths and wider effects of using PCSK9 inhibitors in therapeutic settings.
Collapse
Affiliation(s)
| | - Prince Shah-Riar
- Internal Medicine, Doctors Hospital at Renaissance (DHR) Health, Edinburg, USA
| | - Mousumi Khanam
- Internal Medicine, Dhaka Medical College and Hospital, Dhaka, BGD
| | - Farzana Khair
- Internal Medicine, Bangladesh Medical College, Dhaka, BGD
| | - Anahita Sadat
- Internal Medicine, Dhaka Medical College and Hospital, Dhaka, BGD
| | - Maksuda Khan Tania
- Internal Medicine, Ibrahim Medical College and Birdem General Hospital, Dhaka, BGD
| | - Siddiqi M Haque
- Internal Medicine, University of California, Riverside School of Medicine, Riverside, USA
| | - Shaila S Saaki
- Internal Medicine, Dhaka Medical College and Hospital, Dhaka, BGD
| | | | | | - Maliha Tabassum
- Internal Medicine, Holy Family Red Crescent Medical College, Dhaka, BGD
| | | | - Rashedul Hasan
- Internal Medicine, Desert Valley Hospital, Victorville, USA
| |
Collapse
|
2
|
Debernardi C, Savoca A, De Gregorio A, Casalone E, Rosselli M, Herman EJ, Di Primio C, Tumino R, Sieri S, Vineis P, Panico S, Sacerdote C, Ardissino D, Asselta R, Matullo G. Population Heterogeneity and Selection of Coronary Artery Disease Polygenic Scores. J Pers Med 2024; 14:1025. [PMID: 39452533 PMCID: PMC11508882 DOI: 10.3390/jpm14101025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES The identification of coronary artery disease (CAD) high-risk individuals is a major clinical need for timely diagnosis and intervention. Many different polygenic scores (PGSs) for CAD risk are available today to estimate the genetic risk. It is necessary to carefully choose the score to use, in particular for studies on populations, which are not adequately represented in the large datasets of European biobanks, such as the Italian one. This work aimed to analyze which PGS had the best performance within the Italian population. METHODS We used two Italian independent cohorts: the EPICOR case-control study (576 individuals) and the Atherosclerosis, Thrombosis, and Vascular Biology (ATVB) Italian study (3359 individuals). We evaluated 266 PGS for cardiovascular disease risk from the PGS Catalog, selecting 51 for CAD. RESULTS Distributions between patients and controls were significantly different for 49 scores (p-value < 0.01). Only five PGS have been trained and tested for the European population specifically. PGS003727 demonstrated to be the most accurate when evaluated independently (EPICOR AUC = 0.68; ATVB AUC = 0.80). Taking into account the conventional CAD risk factors further enhanced the performance of the model, particularly in the ATVB study (p-value = 0.0003). CONCLUSIONS European CAD PGS could have different risk estimates in peculiar populations, such as the Italian one, as well as in various geographical macro areas. Therefore, further evaluation is recommended for clinical applicability.
Collapse
Affiliation(s)
- Carla Debernardi
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Angelo Savoca
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Alessandro De Gregorio
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Elisabetta Casalone
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Miriam Rosselli
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Elton Jalis Herman
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Cecilia Di Primio
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, Azienda Ospedaliera “Civile-M.P. Arezzo”, 97100 Ragusa, Italy;
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20100 Milan, Italy;
| | - Paolo Vineis
- MRC-PHE Centre for Environment and Health, Imperial College London, London W12 0BZ, UK;
| | - Salvatore Panico
- Department of Clinical and Experimental Medicine, University Federico II, 80100 Naples, Italy;
| | - Carlotta Sacerdote
- Piedmont Reference Centre for Epidemiology and Cancer Prevention (CPO Piemonte), 10126 Turin, Italy;
| | - Diego Ardissino
- Cardiology Department, Azienda Ospedaliero-Universitaria of Parma, 43100 Parma, Italy;
- Department of Medicine and Surgery, University of Parma, 43100 Parma, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Giuseppe Matullo
- Genomic Variation, Complex Diseases and Population Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (C.D.); (A.S.); (A.D.G.); (E.C.); (M.R.); (E.J.H.); (C.D.P.)
- Medical Genetic Service, Città della Salute e della Scienza, 10126 Turin, Italy
| |
Collapse
|
3
|
Khayatan D, Zare K, Khanahmadi M, Momtaz S, Butler AE, Jamialahmadi T, Almahmeed W, Abdolghaffari AH, Sahebkar A. The role of natural products as PCSK9 modulators: A review. Phytother Res 2024; 38:4081-4098. [PMID: 38899632 DOI: 10.1002/ptr.8260] [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: 02/03/2023] [Revised: 04/25/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
A variety of mechanisms and drugs have been shown to attenuate cardiovascular disease (CVD) onset and/or progression. Recent researchers have identified a potential role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in modulating lipid metabolism and reducing plasma low density lipoprotein (LDL) levels. PCSK9 is the central protein in the metabolism of LDL cholesterol (LDL-C) owing to its major function in LDL receptor (LDLR) degradation. Due to the close correlation of cardiovascular disease with lipid levels, many in vivo and in vitro investigations are currently underway studying the physiological role of PCSK9. Furthermore, many studies are actively investigating the mechanisms of various compounds that influence lipid associated-disorders and their associated cardiovascular diseases. PCSK9 inhibitors have been shown to have significant impact in the prevention of emerging cardiovascular diseases. Natural products can effectively be used as PCSK9 inhibitors to control lipid levels through various mechanisms. In this review, we evaluate the role of phytochemicals and natural products in the regulation of PCSK9, and their ability to prevent cardiovascular diseases. Moreover, we describe their mechanisms of action, which have not to date been delineated.
Collapse
Affiliation(s)
- Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Kimia Zare
- School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Maryam Khanahmadi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Weerawatanakorn M, Kamchonemenukool S, Koh YC, Pan MH. Exploring Phytochemical Mechanisms in the Prevention of Cholesterol Dysregulation: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6833-6849. [PMID: 38517334 PMCID: PMC11018292 DOI: 10.1021/acs.jafc.3c09924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/05/2024] [Accepted: 03/09/2024] [Indexed: 03/23/2024]
Abstract
Although cholesterol plays a key role in many physiological processes, its dysregulation can lead to several metabolic diseases. Statins are a group of drugs widely used to lower cholesterol levels and cardiovascular risk but may lead to several side effects in some patients. Therefore, the development of a plant-based therapeutic adjuvant with cholesterol-lowering activity is desirable. The maintenance of cholesterol homeostasis encompasses multiple steps, including biosynthesis and metabolism, uptake and transport, and bile acid metabolism; issues arising in any of these processes could contribute to the etiology of cholesterol-related diseases. An increasing body of evidence strongly indicates the benefits of phytochemicals for cholesterol regulation; traditional Chinese medicines prove beneficial in some disease models, although more scientific investigations are needed to confirm their effectiveness. One of the main functions of cholesterol is bile acid biosynthesis, where most bile acids are recycled back to the liver. The composition of bile acid is partly modulated by gut microbes and could be harmful to the liver. In this regard, the reshaping effect of phytochemicals on gut microbiota has been widely reported in the literature for its significance. Therefore, we reviewed studies conducted over the past 5 years elucidating the regulatory effects of phytochemicals or herbal medicines on cholesterol metabolism. In addition, their effects on the recomposition of gut microbiota and bile acid metabolism due to modulation are discussed. This review aims to provide novel insights into the treatment of cholesterol dysregulation and the anticipated development of natural-based compounds in the near and far future.
Collapse
Affiliation(s)
- Monthana Weerawatanakorn
- Department
of Agro-Industry, Naresuan University, 99 Moo 9, Thapho, Muang, Phitsanulok 65000, Thailand
- Centre
of Excellence in Fats and Oils, Naresuan
University Science Park, 99 M 9, Thapho, Muang, Phitsanulok 65000, Thailand
| | - Sudthida Kamchonemenukool
- Department
of Agro-Industry, Naresuan University, 99 Moo 9, Thapho, Muang, Phitsanulok 65000, Thailand
| | - Yen-Chun Koh
- Institute
of Food Science and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Min-Hsiung Pan
- Institute
of Food Science and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Medical Research, China Medical University Hospital, China Medical University, Taichung City 40447, Taiwan
- Department
of Health and Nutrition Biotechnology, Asia
University, Taichung City 41354, Taiwan
| |
Collapse
|
5
|
Braczko A, Harasim G, Kawecka A, Walczak I, Kapusta M, Narajczyk M, Stawarska K, Smoleński RT, Kutryb-Zając B. Blocking cholesterol formation and turnover improves cellular and mitochondria function in murine heart microvascular endothelial cells and cardiomyocytes. Front Physiol 2023; 14:1216267. [PMID: 37745244 PMCID: PMC10512729 DOI: 10.3389/fphys.2023.1216267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Statins and proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) are cornerstones of therapy to prevent cardiovascular disease, acting by lowering lipid concentrations and only partially identified pleiotropic effects. This study aimed to analyze impacts of atorvastatin and synthetic peptide PCSK9i on bioenergetics and function of microvascular endothelial cells and cardiomyocytes. Methods: Mitochondrial function and abundance as well as intracellular nucleotides, membrane potential, cytoskeleton structure, and cell proliferation rate were evaluated in mouse heart microvascular endothelial cells (H5V) and cardiomyocytes (HL-1) under normal and hypoxia-mimicking conditions (CoCl2 exposure). Results: In normal conditions PCSK9i, unlike atorvastatin, enhanced mitochondrial respiratory parameters, increased nucleotide levels, prevented actin cytoskeleton disturbances and stimulated endothelial cell proliferation. Under hypoxia-mimicking conditions both atorvastatin and PCSK9i improved the mitochondrial respiration and membrane potential in both cell types. Conclusion: This study demonstrated that both treatments benefited the endothelial cell and cardiomyocyte bioenergetics, but the effects of PCSK9i were superior.
Collapse
Affiliation(s)
- Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Gabriela Harasim
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Ada Kawecka
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Iga Walczak
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | | | | | - Klaudia Stawarska
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | | | | |
Collapse
|
6
|
Waiz M, Alvi SS, Khan MS. Potential dual inhibitors of PCSK-9 and HMG-R from natural sources in cardiovascular risk management. EXCLI JOURNAL 2022; 21:47-76. [PMID: 35221836 PMCID: PMC8859648 DOI: 10.17179/excli2021-4453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) stands amongst the leading causes of mortality worldwide and has attracted the attention of world's leading pharmaceutical companies in order to tackle such mortalities. The low-density lipoprotein-cholesterol (LDL-C) is considered the most prominent biomarker for the assessment of ASCVD risk. Distinct inhibitors of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMG-R), the chief hepatic cholesterogenic enzyme, are being used since last seven decades to manage hypercholesterolemia. On the other hand, discovery and the association of proprotein convertase subtilisin/kexin type-9 (PCSK-9) with increased ASCVD risk have established PCSK-9 as a novel therapeutic target in cardiovascular medicine. PCSK-9 is well reckoned to facilitate the LDL-receptor (LDL-R) degradation and compromised LDL-C clearance leading to the arterial atherosclerotic plaque formation. The currently available HMG-R inhibitors (statins) and PCSK-9 inhibitors (siRNA, anti-sense oligonucleotides, and monoclonal antibodies) have shown great promises in achieving LDL-C lowering goals, however, their life long prescriptions have raised significant concerns. These deficits associated with the synthetic HMG-R and PCSK-9 inhibitors called for the discovery of alternative therapeutic candidates with potential dual HMG-R and PCSK-9 inhibitory activities from natural origins. Therefore, this report firstly describes the mechanistic insights into the cholesterol homeostasis through HMG-R, PCSK-9, and LDL-R functionality and then compiles the pharmacological effects of natural secondary metabolites with special emphasis on their dual HMG-R and PCSK-9 inhibitory action. In conclusion, various natural products exhibit atheroprotective effects via targeting HMG-R and PCSK-9 activities and lipoprotein metabolism, however, further clinical assessments are still warranted prior their approval for ASCVD risk management in hypercholesterolemic patients.
Collapse
Affiliation(s)
- Mohd Waiz
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, U.P. 226026, India
| | - Sahir Sultan Alvi
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, U.P. 226026, India
| | - M Salman Khan
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, U.P. 226026, India
| |
Collapse
|
7
|
Ben-Naim L, Khalaila I, Papo N. Modifying pH-sensitive PCSK9/LDLR interactions as a strategy to enhance hepatic cell uptake of low-density lipoprotein cholesterol (LDL-C). Protein Eng Des Sel 2022; 35:6529797. [DOI: 10.1093/protein/gzab032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 11/14/2022] Open
Abstract
Abstract
LDL-receptor (LDLR)-mediated uptake of LDL-C into hepatocytes is impaired by lysosomal degradation of LDLR, which is promoted by proprotein convertase subtilisin/kexin type 9 (PCSK9). Cell surface binding of PCSK9 to LDLR produces a complex that translocates to an endosome, where the acidic pH strengthens the binding affinity of PCSK9 to LDLR, preventing LDLR recycling to the cell membrane. We present a new approach to inhibit PCSK9-mediated LDLR degradation, namely, targeting the PCSK9/LDLR interface with a PCSK9-antagonist, designated Flag-PCSK9PH, which prevents access of WT PCSK9 to LDLR. In HepG2 cells, Flag-PCSK9PH, a truncated version (residues 53–451) of human WT PCSK9, strongly bound LDLR at the neutral pH of the cell surface but dissociated from it in the endosome (acidic pH), allowing LDLR to exit the lysosomes intact and recycle to the cell membrane. Flag-PCSK9PH thus significantly enhanced cell-surface LDLR levels and the ability of LDLR to take up extracellular LDL-C.
Collapse
Affiliation(s)
- Lital Ben-Naim
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Isam Khalaila
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
8
|
Lebeau PF, Wassef H, Byun JH, Platko K, Ason B, Jackson S, Dobroff J, Shetterly S, Richards WG, Al-Hashimi AA, Won KD, Mbikay M, Prat A, Tang A, Paré G, Pasqualini R, Seidah NG, Arap W, Chrétien M, Austin RC. The loss-of-function PCSK9Q152H variant increases ER chaperones GRP78 and GRP94 and protects against liver injury. J Clin Invest 2021; 131:128650. [PMID: 33211673 DOI: 10.1172/jci128650] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Individuals harboring the loss-of-function (LOF) proprotein convertase subtilisin/kexin type 9 Gln152His variation (PCSK9Q152H) have low circulating low-density lipoprotein cholesterol levels and are therefore protected against cardiovascular disease (CVD). This uncleavable form of proPCSK9, however, is retained in the endoplasmic reticulum (ER) of liver hepatocytes, where it would be expected to contribute to ER storage disease (ERSD), a heritable condition known to cause systemic ER stress and liver injury. Here, we examined liver function in members of several French-Canadian families known to carry the PCSK9Q152H variation. We report that PCSK9Q152H carriers exhibited marked hypocholesterolemia and normal liver function despite their lifelong state of ER PCSK9 retention. Mechanistically, hepatic overexpression of PCSK9Q152H using adeno-associated viruses in male mice greatly increased the stability of key ER stress-response chaperones in liver hepatocytes and unexpectedly protected against ER stress and liver injury rather than inducing them. Our findings show that ER retention of PCSK9 not only reduced CVD risk in patients but may also protect against ERSD and other ER stress-driven conditions of the liver. In summary, we have uncovered a cochaperone function for PCSK9Q152H that explains its hepatoprotective effects and generated a translational mouse model for further mechanistic insights into this clinically relevant LOF PCSK9 variant.
Collapse
Affiliation(s)
- Paul F Lebeau
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Hanny Wassef
- Laboratory of Functional Endoproteolysis, Clinical Research Institute of Montreal, affiliated with the University of Montreal, Montreal, Quebec, Canada
| | - Jae Hyun Byun
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Khrystyna Platko
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Brandon Ason
- Cardiometabolic Disorders, Amgen Research Inc., South San Francisco, California, USA
| | - Simon Jackson
- Cardiometabolic Disorders, Amgen Research Inc., South San Francisco, California, USA
| | | | - Susan Shetterly
- Cardiometabolic Disorders, Amgen Research Inc., South San Francisco, California, USA
| | | | - Ali A Al-Hashimi
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Kevin Doyoon Won
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| | - Majambu Mbikay
- Laboratory of Functional Endoproteolysis, Clinical Research Institute of Montreal, affiliated with the University of Montreal, Montreal, Quebec, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, affiliated with the University of Montreal, Montreal, Quebec, Canada
| | - An Tang
- Department of Radiology at the Centre Hospitalier Universitaire de Montréal, University of Montreal, Montreal, Quebec, Canada
| | - Guillaume Paré
- Population Health Research Institute and Departments of Medicine, Epidemiology, and Pathology, McMaster University, Hamilton, Ontario, Canada
| | | | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, affiliated with the University of Montreal, Montreal, Quebec, Canada
| | - Wadih Arap
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School and Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Michel Chrétien
- Laboratory of Functional Endoproteolysis, Clinical Research Institute of Montreal, affiliated with the University of Montreal, Montreal, Quebec, Canada
| | - Richard C Austin
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton and Hamilton Centre for Kidney Research, Hamilton, Ontario, Canada
| |
Collapse
|
9
|
Zainab R, Kaleem A, Ponczek MB, Abdullah R, Iqtedar M, Hoessli DC. Finding inhibitors for PCSK9 using computational methods. PLoS One 2021; 16:e0255523. [PMID: 34351937 PMCID: PMC8341581 DOI: 10.1371/journal.pone.0255523] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/17/2021] [Indexed: 12/20/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is one of the key targets for atherosclerosis drug development as its binding with low-density lipoprotein receptor leads to atherosclerosis. The protein-ligand interaction helps to understand the actual mechanism for the pharmacological action. This research aims to discover the best inhibitory candidates targeting PCSK9. To start with, reported ACE inhibitors were incorporated into pharmacophore designing using PharmaGist to produce pharmacophore models. Selected models were later screened against the ZINC database using ZINCPHARMER to define potential drug candidates that were docked with the target protein to understand their interactions. Molecular docking revealed the top 10 drug candidates against PCSK9, with binding energies ranging from -9.8 kcal·mol-1 to -8.2 kcal·mol-1, which were analyzed for their pharmacokinetic properties and oral bioavailability. Some compounds were identified as plant-derived compounds like (S)-canadine, hesperetin or labetalol (an antihypertensive drug). Molecular dynamics results showed that these substances formed stable protein-ligand complexes. (S)-canadine-PCSK9 complex was the most stable with the lowest RMSD. It was concluded that (S)-canadine may act as a potential inhibitor against atherosclerosis for the development of new PCSK9 inhibitory drugs in future in vitro research.
Collapse
Affiliation(s)
- Rida Zainab
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Afshan Kaleem
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
- * E-mail: (AK); (MBP)
| | - Michał B. Ponczek
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
- * E-mail: (AK); (MBP)
| | - Roheena Abdullah
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Mehwish Iqtedar
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Daniel C. Hoessli
- Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Studies, University of Karachi, Karachi, Pakistan
| |
Collapse
|
10
|
Xu Q, Deng Y, Xiao J, Liu X, Zhou M, Ren Z, Peng J, Tang Y, Jiang Z, Tang Z, Liu L. Three Musketeers for Lowering Cholesterol: Statins, Ezetimibe and Evolocumab. Curr Med Chem 2021; 28:1025-1041. [PMID: 32368969 DOI: 10.2174/0929867327666200505091738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/08/2020] [Accepted: 03/19/2020] [Indexed: 11/22/2022]
Abstract
Coronary heart disease (CHD) is closely related to hypercholesterolemia, and lowering serum cholesterol is currently the most important strategy in reducing CHD. In humans, the serum cholesterol level is determined mainly by three metabolic pathways, namely, dietary cholesterol intake, cholesterol synthesis, and cholesterol degradation in vivo. An intervention that targets the key molecules in the three pathways is an important strategy in lowering serum lipids. Statins inhibit 3-hydroxyl-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase) to reduce low-density lipoprotein (LDL) by about 20% to 45%. However, up to 15% of patients cannot tolerate the potential side effects of high statin dosages, and several patients also still do not reach their optimal LDL goals after being treated with statins. Ezetimibe inhibits cholesterol absorption by targeting the Niemann-Pick C1-like 1 protein (NPC1L1), which is related to cholesterol absorption in the intestines. Ezetimibe lowers LDL by about 18% when used alone and by an additional 25% when combined with statin therapy. The proprotein convertase subtilisin/kexin type 9 (PCSK9) increases hepatic LDLR degradation, thereby reducing the liver's ability to remove LDL, which can lead to hypercholesterolemia. Evolocumab, which is a PCSK9 monoclonal antibody, can reduce LDL from baseline by 53% to 56%. The three drugs exert lipid-lowering effects by regulating the three key pathways in lipid metabolism. Combining any with the two other drugs on the basis of statin treatment has improved the lipid-lowering effect. Whether the combination of the three musketeers will reduce the side effects of monotherapy and achieve the lipid-lowering effect should be studied further in the future.
Collapse
Affiliation(s)
- Qian Xu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Yiming Deng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Jun Xiao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Xiangrui Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Min Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Juan Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Yaling Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Zhihan Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Lushan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
11
|
Coronary CT Angiography Guided Medical Therapy in Subclinical Atherosclerosis. J Clin Med 2021; 10:jcm10040625. [PMID: 33562179 PMCID: PMC7914610 DOI: 10.3390/jcm10040625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
The goals of primary prevention in coronary atherosclerosis are to avoid sudden cardiac death, myocardial infarction or the need for revascularization procedures. Successful prevention will rely on accurate identification, effective therapy and monitoring of those at risk. Identification and potential monitoring can be achieved using cardiac computed tomography (CT). Cardiac CT can determine coronary artery calcification (CAC), a useful surrogate of coronary atherosclerosis burden. Cardiac CT can also assess coronary CT angiography (CCTA). CCTA can identify arterial lumen narrowing and highlight mural atherosclerosis hitherto hidden from other anatomical approaches. Herein we consider the role of CCTA and CAC-scoring in subclinical atherosclerosis. We explore the use of these modalities in screening and discuss data that has used CCTA for guiding primary prevention. We examine therapeutic trials using CCTA to determine the effects of plaque-modifying therapies. Finally, we address the role of CCTA and CAC to guide therapy as defined in current primary prevention documents. CCTA has emerged as an essential tool in the detection and management of clinical coronary artery disease. To date, its role in subclinical atherosclerosis is less well defined, yet with modern CT scanners and continued pharmacotherapy development, CCTA is likely to achieve a more prominent place in the primary prevention of coronary atherosclerosis.
Collapse
|
12
|
Riaz H, Khan SU, Lateef N, Talluri S, Khan MS, Desai MY. Residual inflammatory risk after contemporary lipid lowering therapy. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2020; 6:105-111. [PMID: 31609450 PMCID: PMC7850089 DOI: 10.1093/ehjqcco/qcz055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/29/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Recently, there has been an increasing interest in targeting inflammation to reduce major adverse cardiovascular events (MACE) in patients with cardiovascular risk. Statins, PCSK9 inhibitors, and ezetimibe have been shown to reduce MACE owing to reduction in low-density lipoproteins cholesterol (LDL-c). Herein, we investigate whether the intensity of these agents is associated with (i) discernible reduction in inflammation measured by the levels of high-sensitivity C-reactive protein (hsCRP); (ii) reduction in MACE; (iii) if there is an association between the baseline hsCRP and MACE. METHODS AND RESULTS Electronic databases were searched for randomized controlled trials (RCTs) that compared statins, ezetimibe, PCSK9 inhibitors with placebos/active controls and reported MACEs and hsCRP (mg/L). Studies were stratified based on baseline hsCRP (<2, 2-3, >3) with subgroup analysis conducted across each stratum. Fourteen RCTs including 133 109 patients randomized into more intensive therapy (MIT) and less intensive therapy were selected. Meta-analysis did not demonstrate any significant differences between use of MIT and hsCRP levels (mean difference, -0.02; CI, -0.06, 0.02; P = 0.31). The MIT significantly reduced the risk of MACE (RR, 0.82; CI, 0.75, 0.91; P < 0.001). The relative risk and absolute risk remained consistent across the strata. However, there was a 0.5% statistically significant absolute risk reduction in all-cause mortality in patients with higher hsCRP (RD, -0.005; CI, -0.009, -0.001; P = 0.01). CONCLUSION Overall, LDL-c lowering therapies reduce relative risk of MACEs particularly in patients with higher baseline hsCRP. However, there appears to be a residual inflammatory risk despite the use of contemporary lipid lowering agents.
Collapse
Affiliation(s)
- Haris Riaz
- Department of Cardiology, Heart and Vascular Institute, Cleveland Clinic, Euclid Avenue, Cleveland, OH 44195, USA
| | - Safi U Khan
- Department of Internal Medicine, West Virginia University, Morgantown, WV, 26506 USA
| | - Noman Lateef
- Department of Internal Medicine, Creighton University, 7500 Mercy Road, Omaha, NE 68124, USA
| | - Swapna Talluri
- Department of Internal Medicine, Guthrie/Robert Packer Hospital, Guthrie Square, Sayre, PA 18840, USA
| | - Muhammad Shahzeb Khan
- Department of Internal Medicine, John H Stroger Jr, Hospital of Cook County, 969 W Ogden Ave, Chicago, IL 60612, USA
| | - Milind Y Desai
- Department of Cardiology, Heart and Vascular Institute, Cleveland Clinic, Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
13
|
Coudert P, Daulhac-Terrail L. Les nouveaux hypocholestérolémiants. ACTUALITES PHARMACEUTIQUES 2020. [DOI: 10.1016/j.actpha.2019.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Chen B, Shi X, Cui Y, Hou A, Zhao P. A Review of PCSK9 Inhibitors and their Effects on Cardiovascular Diseases. Curr Top Med Chem 2019; 19:1790-1817. [PMID: 31400268 DOI: 10.2174/1568026619666190809094203] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/07/2019] [Accepted: 07/25/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardiovascular diseases remain the leading cause of morbidity and mortality in the world, with elevated Low-Density Lipoprotein-Cholesterol (LDL-C) levels as the major risk factor. Lower levels of LDL-C can effectively reduce the risk of cardiovascular diseases. Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an important role in regulating the degradation of hepatic LDL receptors that remove LDL-C from the circulation. PCSK9 inhibitors are a new class of agents that are becoming increasingly important in the treatment to reduce LDL-C levels. Two PCSK9 inhibitors, alirocumab and evolocumab, have been approved to treat hypercholesterolemia and are available in the United States and the European Union. Through the inhibition of PCSK9 and increased recycling of LDL receptors, serum LDL-C levels can be significantly reduced. OBJECTIVE This review will describe the chemistry, pharmacokinetics, and pharmacodynamics of PCSK9 inhibitors and their clinical effects.
Collapse
Affiliation(s)
- Bo Chen
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 20092, China
| | - Xin Shi
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 20092, China
| | - Yanping Cui
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 20092, China
| | - Aiping Hou
- Department of Pediatric, Shidong Hospital, Shanghai 20092, China
| | - Pengjun Zhao
- Department of Pediatric, Shidong Hospital, Shanghai 20092, China
| |
Collapse
|
15
|
Ding Z, Wang X, Liu S, Shahanawaz J, Theus S, Fan Y, Deng X, Zhou S, Mehta JL. PCSK9 expression in the ischaemic heart and its relationship to infarct size, cardiac function, and development of autophagy. Cardiovasc Res 2019; 114:1738-1751. [PMID: 29800228 DOI: 10.1093/cvr/cvy128] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/17/2018] [Indexed: 11/12/2022] Open
Abstract
Aims Inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as a novel therapy to treat hypercholesterolaemia and related cardiovascular diseases. This study determined if PCSK9 can regulate infarct size, cardiac function, and autophagy during ischaemia. Methods and results Mice hearts were subjected to left coronary artery (LCA) occlusion. There was intense expression of PCSK9 in the zone bordering the infarct area in association with marked cardiac contractile dysfunction in the wild-type mice. This region also revealed intense autophagy. To assess the role of PCSK9 in the evolution of infarct size and function and development of autophagy, we used wild-type mice pre-treated with two different PCSK9 inhibitors (Pep2-8 and EGF-A) or mice lacking PCSK9 gene. Both strategies resulted in smaller infarcts and improved cardiac function following LCA ligation. PCSK9 inhibition also markedly reduced autophagy. Relationship between myocardial ischaemia and PCSK9 expression and autophagy was examined in cultured mouse cardiomyocytes. Exposure of cardiomyocytes to hypoxia resulted in prompt PCSK9 expression and autophagy signals; both were blocked by HIF-1α siRNA. Further, treatment of cardiomyocytes with recombinant PCSK9 during hypoxia induced, and treatment with PCSK9 siRNA reduced, autophagy suggesting a possible role of PCSK9 in the determination of autophagy. Other studies revealed activation of ROS-ATM-LKB1-AMPK axis as a possible mechanism of PCSK-induced autophagy. Hearts of humans with recent infarcts also showed expression of PCSK9 and autophagy in the border zone-similar to that in the infarcted mouse heart. Conclusion PCSK9 is up-regulated in the ischaemic hearts and determines development of infarct size, cardiac function, and autophagy.
Collapse
Affiliation(s)
- Zufeng Ding
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xianwei Wang
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China
| | - Shijie Liu
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jiwani Shahanawaz
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sue Theus
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Xiaoyan Deng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Sichang Zhou
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Jawahar L Mehta
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
16
|
Caron J, Pène V, Tolosa L, Villaret M, Luce E, Fourrier A, Heslan JM, Saheb S, Bruckert E, Gómez-Lechón MJ, Nguyen TH, Rosenberg AR, Weber A, Dubart-Kupperschmitt A. Low-density lipoprotein receptor-deficient hepatocytes differentiated from induced pluripotent stem cells allow familial hypercholesterolemia modeling, CRISPR/Cas-mediated genetic correction, and productive hepatitis C virus infection. Stem Cell Res Ther 2019; 10:221. [PMID: 31358055 PMCID: PMC6664765 DOI: 10.1186/s13287-019-1342-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/03/2019] [Accepted: 07/14/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Familial hypercholesterolemia type IIA (FH) is due to mutations in the low-density lipoprotein receptor (LDLR) resulting in elevated levels of low-density lipoprotein cholesterol (LDL-c) in plasma and in premature cardiovascular diseases. As hepatocytes are the only cells capable of metabolizing cholesterol, they are therefore the target cells for cell/gene therapy approaches in the treatment of lipid metabolism disorders. Furthermore, the LDLR has been reported to be involved in hepatitis C virus (HCV) entry into hepatocytes; however, its role in the virus infection cycle is still disputed. METHODS We generated induced pluripotent stem cells (iPSCs) from a homozygous LDLR-null FH-patient (FH-iPSCs). We constructed a correction cassette bearing LDLR cDNA under the control of human hepatic apolipoprotein A2 promoter that targets the adeno-associated virus integration site AAVS1. We differentiated both FH-iPSCs and corrected FH-iPSCs (corr-FH-iPSCs) into hepatocytes to study statin-mediated regulation of genes involved in cholesterol metabolism. Upon HCV particle inoculation, viral replication and production were quantified in these cells. RESULTS We showed that FH-iPSCs displayed the disease phenotype. Using homologous recombination mediated by the CRISPR/Cas9 system, FH-iPSCs were genetically corrected by the targeted integration of a correction cassette at the AAVS1 locus. Both FH-iPSCs and corr-FH-iPSCs were then differentiated into functional polarized hepatocytes using a stepwise differentiation approach (FH-iHeps and corr-FH-iHeps). The correct insertion and expression of the correction cassette resulted in restoration of LDLR expression and function (LDL-c uptake) in corr-FH-iHeps. We next demonstrated that pravastatin treatment increased the expression of genes involved in cholesterol metabolism in both cell models. Moreover, LDLR expression and function were also enhanced in corr-FH-iHeps after pravastatin treatment. Finally, we demonstrated that both FH-iHeps and corr-FH-iHeps were as permissive to viral infection as primary human hepatocytes but that virus production in FH-iHeps was significantly decreased compared to corr-FH-iHeps, suggesting a role of the LDLR in HCV morphogenesis. CONCLUSIONS Our work provides the first LDLR-null FH cell model and its corrected counterpart to study the regulation of cholesterol metabolism and host determinants of HCV life cycle, and a platform to screen drugs for treating dyslipidemia and HCV infection.
Collapse
Affiliation(s)
- Jérôme Caron
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France
| | | | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | | | - Eléanor Luce
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France
| | - Angélique Fourrier
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Jean-Marie Heslan
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Samir Saheb
- Service d'Endocrinologie Métabolisme, Hôpital Pitié-Salpêtrière, Paris, France
| | - Eric Bruckert
- Service d'Endocrinologie Métabolisme, Hôpital Pitié-Salpêtrière, Paris, France
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.,CIBERehd, FIS, Barcelona, Spain
| | - Tuan Huy Nguyen
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Arielle R Rosenberg
- Université Paris Descartes, EA4474, Paris, France.,AP-HP, Hôpital Cochin, Service de Virologie, Paris, France
| | - Anne Weber
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France
| | - Anne Dubart-Kupperschmitt
- INSERM UMR_S1193, Hôpital Paul Brousse, Villejuif, France; UMR-S1193, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France; DHU Hepatinov, Hôpital Paul Brousse, Villejuif, France.
| |
Collapse
|
17
|
Wang Y, Liu ZP. PCSK9 Inhibitors: Novel Therapeutic Strategies for Lowering LDLCholesterol. Mini Rev Med Chem 2018; 19:165-176. [DOI: 10.2174/1389557518666180423111442] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/17/2017] [Accepted: 12/29/2017] [Indexed: 11/22/2022]
Abstract
Statins are currently the major therapeutic strategies to lower low-density lipoprotein cholesterol
(LDL-C) levels. However, a number of hypercholesterolemia patients still have a residual cardiovascular
disease (CVD) risk despite taking the maximum-tolerated dose of statins. Proprotein convertase
subtilisin/kexin type 9 (PCSK9) binds to low-density lipoprotein receptor (LDLR), inducing its
degradation in the lysosome and inhibiting LDLR recirculating to the cell membranes. The gain-offunction
mutations in PCSK9 elevate the LDL-C levels in plasma. Therefore, PCSK9 inhibitors become
novel therapeutic approaches in the treatment of hypercholesterolemia. Several PCSK9 inhibitors
have been under investigation, and much progress has been made in clinical trials, especially for
monoclonal antibodies (MoAbs). Two MoAbs, evolocumab and alirocumab, are now in clinical use. In
this review, we summarize the development of PCSK9 inhibitors, including antisense oligonucleotides
(ASOs), small interfering RNA (siRNA), small molecule inhibitor, MoAbs, mimetic peptides and
adnectins, and the related safety issues.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Zhao-Peng Liu
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
18
|
Pasipoularides A. Implementing genome-driven personalized cardiology in clinical practice. J Mol Cell Cardiol 2018; 115:142-157. [PMID: 29343412 PMCID: PMC5820118 DOI: 10.1016/j.yjmcc.2018.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/04/2018] [Accepted: 01/12/2018] [Indexed: 12/18/2022]
Abstract
Genomics designates the coordinated investigation of a large number of genes in the context of a biological process or disease. It may be long before we attain comprehensive understanding of the genomics of common complex cardiovascular diseases (CVDs) such as inherited cardiomyopathies, valvular diseases, primary arrhythmogenic conditions, congenital heart syndromes, hypercholesterolemia and atherosclerotic heart disease, hypertensive syndromes, and heart failure with preserved/reduced ejection fraction. Nonetheless, as genomics is evolving rapidly, it is constructive to survey now pertinent concepts and breakthroughs. Today, clinical multimodal electronic medical/health records (EMRs/EHRs) incorporating genomic information establish a continuously-learning, vast knowledge-network with seamless cycling between clinical application and research. It can inform insights into specific pathogenetic pathways, guide biomarker-assisted precise diagnoses and individualized treatments, and stratify prognoses. Complex CVDs blend multiple interacting genomic variants, epigenetics, and environmental risk-factors, engendering progressions of multifaceted disease-manifestations, including clinical symptoms and signs. There is no straight-line linkage between genetic cause(s) or causal gene-variant(s) and disease phenotype(s). Because of interactions involving modifier-gene influences, (micro)-environmental, and epigenetic effects, the same variant may actually produce dissimilar abnormalities in different individuals. Implementing genome-driven personalized cardiology in clinical practice reveals that the study of CVDs at the level of molecules and cells can yield crucial clinical benefits. Complementing evidence-based medicine guidelines from large ("one-size fits all") randomized controlled trials, genomics-based personalized or precision cardiology is a most-creditable paradigm: It provides customizable approaches to prevent, diagnose, and manage CVDs with treatments directly/precisely aimed at causal defects identified by high-throughput genomic technologies. They encompass stem cell and gene therapies exploiting CRISPR-Cas9-gene-editing, and metabolomic-pharmacogenomic therapeutic modalities, precisely fine-tuned for the individual patient. Following the Human Genome Project, many expected genomics technology to provide imminent solutions to intractable medical problems, including CVDs. This eagerness has reaped some disappointment that advances have not yet materialized to the degree anticipated. Undoubtedly, personalized genetic/genomics testing is an emergent technology that should not be applied without supplementary phenotypic/clinical information: Genotype≠Phenotype. However, forthcoming advances in genomics will naturally build on prior attainments and, combined with insights into relevant epigenetics and environmental factors, can plausibly eradicate intractable CVDs, improving human health and well-being.
Collapse
Affiliation(s)
- Ares Pasipoularides
- Consulting Professor of Surgery, Emeritus Faculty of Surgery and of Biomedical Engineering, Duke University School of Medicine and Graduate School, Durham, NC 27710, USA.
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Genetic dyslipidemias contribute to the prevalence of ischemic heart disease. The field of genetic dyslipidemias and their influence on atherosclerotic heart disease is rapidly developing and accumulating increasing evidence. The purpose of this review is to describe the current state of knowledge in regard to inherited atherogenic dyslipidemias. The disorders of familial hypercholesterolemia (FH) and elevated lipoprotein(a) will be detailed. Genetic technology has made rapid advancements, leading to new discoveries in inherited atherogenic dyslipidemias, which will be explored in this review, as well as a description of possible future developments. Increasing attention has come upon the genetic disorders of familial hypercholesterolemia and elevated lipoprotein(a). RECENT FINDINGS This review includes new knowledge of these disorders including description of these disorders, their method of diagnosis, their prevalence, their genetic underpinnings, and their effect on the development of cardiovascular disease. In addition, it discusses major advances in genetic technology, including the completion of the human genome sequence, next-generation sequencing, and genome-wide association studies. Also discussed are rare variant studies with specific genetic mechanisms involved in inherited dyslipidemias, such as in the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme. The field of genetics of dyslipidemia and cardiovascular disease is rapidly growing, which will result in a bright future of novel mechanisms of action and new therapeutics.
Collapse
Affiliation(s)
- Kavita Sharma
- Ohio Health Heart and Vascular Physicians, 765 North Hamilton Road, Suite 120, Gahanna, OH, 43230, USA
| | - Ragavendra R Baliga
- The Ohio State University Wexner Medical Center, Suite 200, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
20
|
Hansen RJ, Berna MJ, Sperry AE, Beyer TP, Wroblewski VJ, Schroeder KM, Eacho PI. Quantitative characterization of the mechanism of action and impact of a 'proteolysis-permitting' anti-PCSK9 antibody. MAbs 2017; 9:285-296. [PMID: 27981884 PMCID: PMC5297531 DOI: 10.1080/19420862.2016.1270490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A recent report described a novel mechanism of action for an anti-proprotein convertase subtilisin-kexin type 9 (PCSK9) monoclonal antibody (LY3015014, or LY), wherein the antibody has improved potency and duration of action due to the PCSK9 epitope for LY binding. Unlike other antibodies, proteolysis of PCSK9 can occur when LY is bound to PCSK9. We hypothesized that this allowance of PCSK9 cleavage potentially improves LY efficiency through two pathways, namely lack of accumulation of intact PCSK9 and reduced clearance of LY. A quantitative modeling approach is necessary to further understand this novel mechanism of action. We developed a mechanism-based model to characterize the relationship between antibody pharmacokinetics, PCSK9 and LDL cholesterol levels in animals, and used the model to better understand the underlying drivers for the improved efficiency of LY. Simulations suggested that the allowance of cleavage of PCSK9 resulting in a lack of accumulation of intact PCSK9 is the major driver of the improved potency and durability of LY. The modeling reveals that this novel 'proteolysis-permitting' mechanism of LY is a means by which an efficient antibody can be developed with a total antibody dosing rate that is lower than the target production rate. We expect this engineering approach may be applicable to other targets and that the mathematical models presented herein will be useful in evaluating similar approaches.
Collapse
Affiliation(s)
- Ryan J Hansen
- a Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , IN , USA
| | - Michael J Berna
- a Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , IN , USA
| | - Andrea E Sperry
- a Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , IN , USA
| | - Thomas P Beyer
- a Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , IN , USA
| | - Victor J Wroblewski
- a Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , IN , USA
| | - Krista M Schroeder
- a Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , IN , USA
| | | |
Collapse
|
21
|
Hess GP, Natarajan P, Faridi KF, Fievitz A, Valsdottir L, Yeh RW. Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitor Therapy: Payer Approvals and Rejections, and Patient Characteristics for Successful Prescribing. Circulation 2017; 136:2210-2219. [PMID: 29084735 DOI: 10.1161/circulationaha.117.028430] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 09/08/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) are a novel class of medications for patients with familial hypercholesterolemia or clinical atherosclerotic cardiovascular disease requiring additional lipid lowering beyond dietary measures and statin use. Because of the drugs' high cost, rates of prescription approval by payers may be low. We aimed to identify payer approval and rejection rates for PCSK9i prescriptions and the potential factors influencing these rates. METHODS This is a retrospective, descriptive cohort study using nationwide pharmacy claims linked to electronic medical records from a nationwide data warehouse. The data set includes >220 million patients from all 50 states and all payer types with 5140 distinct health plans. PCSK9i prescriptions were submitted for 51 466 patients in the pharmacy data set. The main outcome was approval or rejection of PCSK9i prescription claims. Factors associated with approval and rejection of these medications in the United States were assessed. RESULTS Among patients who were prescribed a PCSK9i, 47.0% were approved for coverage by the payer. Variables that were associated with PCSK9i approval included age >65 years (P<0.01), history of atherosclerotic cardiovascular disease (P<0.01), prescription by a cardiologist or nonprimary care provider (P<0.01), statin intolerance (P=0.03), longer statin duration (P=0.01), and noncommercial payers (P<0.01). Higher low-density lipoprotein cholesterol levels were not associated with higher approval rates. Commercial third-party payers had the lowest approval rates (24.4%) and Medicare had the highest (60.9%). CONCLUSIONS Rates of approval for PCSK9i therapy are low, even for patients who appear to meet labeled indications. Although a combination of clinical characteristics increases the likelihood of approval, payer type is the most significant factor.
Collapse
Affiliation(s)
- Gregory P Hess
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia (G.P.H.). Scientific Studies & Projects, Symphony Health, Conshohocken, PA (G.P.H., A.F.). Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.). Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (P.N.). Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (K.F.F., L.V., R.W.Y.)
| | - Pradeep Natarajan
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia (G.P.H.). Scientific Studies & Projects, Symphony Health, Conshohocken, PA (G.P.H., A.F.). Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.). Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (P.N.). Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (K.F.F., L.V., R.W.Y.)
| | - Kamil F Faridi
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia (G.P.H.). Scientific Studies & Projects, Symphony Health, Conshohocken, PA (G.P.H., A.F.). Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.). Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (P.N.). Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (K.F.F., L.V., R.W.Y.)
| | - Anna Fievitz
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia (G.P.H.). Scientific Studies & Projects, Symphony Health, Conshohocken, PA (G.P.H., A.F.). Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.). Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (P.N.). Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (K.F.F., L.V., R.W.Y.)
| | - Linda Valsdottir
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia (G.P.H.). Scientific Studies & Projects, Symphony Health, Conshohocken, PA (G.P.H., A.F.). Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.). Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (P.N.). Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (K.F.F., L.V., R.W.Y.)
| | - Robert W Yeh
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia (G.P.H.). Scientific Studies & Projects, Symphony Health, Conshohocken, PA (G.P.H., A.F.). Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.). Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA (P.N.). Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Boston, MA (K.F.F., L.V., R.W.Y.).
| |
Collapse
|
22
|
Zhang PY. PCSK9 as a therapeutic target for cardiovascular disease. Exp Ther Med 2017; 13:810-814. [PMID: 28450903 PMCID: PMC5403434 DOI: 10.3892/etm.2017.4055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/12/2017] [Indexed: 01/14/2023] Open
Abstract
It is well recognized that the elevated plasma level of low-density lipoprotein-cholesterol (LDL-C) is a major risk factor for atherosclerosis and cardiovascular disease (CVD). Deposition of pro-atherogenic LDL-C, on the intima of arterial wall, contributes to plaque formation and atherosclerosis, which further leads to lowered blood flow to vital organs and increased risk of CVD. The most commonly used statin therapy is effective in reducing dyslipidemia and preventing cardiovascular events only in about half of the patient population. However, in patients with familial hypercholesterolemia, these drugs were not effective to meet the required goals of lower LDL-C, and to reduce the CVD risk. Furthermore, many patients even develop intolerability to statins and resistance. The identification of pro-protein convertase subtilisin/kexin type 9 (PCSK9) and the association of PCSK9 mutations with familial hypercholesterolemia led to the identification of PCSK9 as a new therapeutic target for lowering LDL-C and dyslipidemia-associated CVD. PCSK9 is found to promote the degradation of LDL-receptor (LDLR), thus rendering it unavailable for recycling to hepatocyte plasma membrane, leading to elevated levels of circulating LDL-C, as it cannot be taken up into cells. While gain-of-function mutations aggravate the degradation of LDLR as in familial hypercholesterolemia whereas loss of function mutations reduce the ability of PCSK9 to promote the degradation of LDLR and thus lower the plasma level of LDL-C and dyslipidemia. Monoclonal antibodies against PCSK9 are currently being tested in clinical trials and are found to be efficacious in countering the activity of PCSK9 and thus control the plasma LDL-C and triglycerides even in statin non-responsive patients and protect against dyslipidemia-related CVD.
Collapse
Affiliation(s)
- Pei-Ying Zhang
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
23
|
Benedict PA, Abdou RM, Dion GR, Woo P, Branski RC, Amin MR. Association of alirocumab therapy with inflammatory lesions of the vocal folds: A case report. Laryngoscope 2016; 127:1652-1654. [PMID: 27933632 DOI: 10.1002/lary.26426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2016] [Indexed: 11/05/2022]
Abstract
Therapeutic monocolonal antibodies (MAbs) are a new, rapidly growing class of medications that frequently have poorly characterized side-effect profiles. We present a patient who developed inflammatory lesions of the vocal folds in temporal relation to the initiation of alirocumab. Lesions of the vocal folds represent a previously unreported adverse effect of alirocumab therapy, making it the second MAb documented with such a side effect. The potential laryngeal effects of alirocumab specifically, and of MAbs more broadly, warrant investigation. Laryngoscope, 127:1652-1654, 2017.
Collapse
Affiliation(s)
- Peter A Benedict
- NYU Voice Center, Department of Otolaryngology-Head and Neck Surgery, New York University School of Medicine, New York, New York, U.S.A
| | - Rania M Abdou
- Phoniatrics Unit, Department of Otorhinolaryngology, Alexandria University School of Medicine, Alexandria, Egypt
| | - Gregory R Dion
- NYU Voice Center, Department of Otolaryngology-Head and Neck Surgery, New York University School of Medicine, New York, New York, U.S.A
| | - Peak Woo
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine, New York, New York, U.S.A
| | - Ryan C Branski
- NYU Voice Center, Department of Otolaryngology-Head and Neck Surgery, New York University School of Medicine, New York, New York, U.S.A
| | - Milan R Amin
- NYU Voice Center, Department of Otolaryngology-Head and Neck Surgery, New York University School of Medicine, New York, New York, U.S.A
| |
Collapse
|
24
|
Abstract
A long, healthy life is a desire and priority for most people. Genetic factors for longevity do not fully explain healthy aging. Recent research suggests that, in addition to other factors, healthy aging is at least in part the result of protective genetic variants for Alzheimer's disease (AD) and coronary artery disease (CAD).
Collapse
|