1
|
Delabays B, De Paoli C, Miller-Nesbitt A, Mooser V. Genetically Enriched Clinical Trials for Precision Development of Noncancer Therapeutics: A Scoping Review. Annu Rev Pharmacol Toxicol 2025; 65:149-167. [PMID: 39348854 DOI: 10.1146/annurev-pharmtox-031524-021631] [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: 10/02/2024]
Abstract
Genetically driven clinical trial enrichment has been proposed to accelerate and reduce the cost of developing new therapeutics. Usage of this approach has not been comprehensively reviewed. We searched Ovid MEDLINE, Embase, Web of Science, Cochrane Library, ClinicalTrials.gov, and WHO ICTRP for articles published between 2010 and 2023. Excluding absorption, distribution, metabolism, and elimination pharmacogenetic studies and anti-infectives, we found 95 completed, 4 terminated, and 22 ongoing prospective genetically enriched trials on 110 drugs for 48 nononcology, nonrare syndromic indications. Trial sizes ranged from 4 to 6,147 participants (median 72) and covered numerous disease areas, particularly neurology (30), metabolism (22), and psychiatry (17). Fifty-six completed studies (60%) met their primary end point. Overall, this scoping review demonstrates that genetically enriched trials are feasible and scalable across disease areas and provide critical information for further development, or attrition, of investigational drugs. Large, appropriately designed disease-, hospital-, or population-based biobanks will undoubtedly facilitate this type of precision drug development approach.
Collapse
Affiliation(s)
- Benoît Delabays
- Canada Excellence Research Chair in Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Quebec, Canada;
| | - Chiara De Paoli
- School of Medicine, University of Eastern Piedmont, Novara, Italy
| | | | - Vincent Mooser
- Canada Excellence Research Chair in Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, Quebec, Canada;
| |
Collapse
|
2
|
Lagunas-Rangel FA, Liepinsh E, Fredriksson R, Alsehli AM, Williams MJ, Dambrova M, Jönsson J, Schiöth HB. Off-target effects of statins: molecular mechanisms, side effects and the emerging role of kinases. Br J Pharmacol 2024; 181:3799-3818. [PMID: 39180421 DOI: 10.1111/bph.17309] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 08/26/2024] Open
Abstract
Statins are one of the most important classes of drugs. In this analytical review, we elucidate the intricate molecular mechanisms and toxicological rationale regarding both the on- (targeting 3-hydroxy-3-methylglutaryl-coenzyme A reductase [HMGCR]) and off-target effects of statins. Statins interact with a number of membrane kinases, such as epidermal growth factor receptor (EGFR), erb-b2 receptor tyrosine kinase 2 (HER2) and MET proto-oncogene, receptor tyrosine kinase (MET), as well as cytosolic kinases, such as SRC proto-oncogene, non-receptor tyrosine kinase (Src) and show inhibitory activity at nanomolar concentrations. In addition, they interact with calcium ATPases and peroxisome proliferator-activated receptor α (PPARα/NR1C1) at higher concentrations. Statins interact with mitochondrial complexes III and IV, and their inhibition of coenzyme Q10 synthesis also impairs the functioning of complexes I and II. Statins act as inhibitors of kinases, calcium ATPases and mitochondrial complexes, while activating PPARα. These off-target effects likely contribute to the side effects observed in patients undergoing statin therapy, including musculoskeletal symptoms and hepatic effects. Interestingly, some off-target effects of statins could also be the cause of favourable outcomes, relating to repurposing statins in conditions such as inflammatory disorders and cancer.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Edgars Liepinsh
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ahmed M Alsehli
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Michael J Williams
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Pharmaceutical Chemistry, Riga Stradiņš University, Riga, Latvia
| | - Jörgen Jönsson
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
Cheshire MD, Akbar UA. Akira Endo: Father of Statins. Cureus 2024; 16:e68198. [PMID: 39347268 PMCID: PMC11439472 DOI: 10.7759/cureus.68198] [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] [Accepted: 08/30/2024] [Indexed: 10/01/2024] Open
Abstract
There was an unprecedented increase in cardiovascular mortality during the 20th century, which ultimately led to a deeper understanding of cholesterol and its role in cardiovascular disease. Akira Endo is a medical innovator who sought to change the tide of this epidemic. He is often referred to as the "Father of Statins" because of his identification of the first 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) inhibitor, which fostered a new era in cholesterol treatment. The offspring of this discovery - statin medications - changed the landscape of cardiovascular prevention and improved outcomes for millions of patients across the globe. Arika Endo used his prior research experience in biochemistry and fungi to create a new way to meet the objective of reducing cardiovascular morbidity and mortality. In this way, he should be recognized not only for this discovery of statins but as a role model for researchers at any stage of their careers.
Collapse
Affiliation(s)
- Michael D Cheshire
- Internal Medicine/Clinical Lipidology, West Virginia University School of Medicine, Parkersburg, USA
| | - Usman A Akbar
- Internal Medicine, West Virginia University School of Medicine, Parkersburg, USA
| |
Collapse
|
4
|
Providencia R. Preoperative statins in cardiac surgery: a tale of small study bias or 'the truth, and nothing but the truth'. Cochrane Database Syst Rev 2024; 7:ED000167. [PMID: 39037830 PMCID: PMC11262553 DOI: 10.1002/14651858.ed000167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Affiliation(s)
- Rui Providencia
- GENEs health and social care evidence SYnthesiS unit, Institute of Health InformaticsUniversity College LondonUnited Kingdom
| |
Collapse
|
5
|
Okamura T, Tsukamoto K, Arai H, Fujioka Y, Ishigaki Y, Koba S, Ohmura H, Shoji T, Yokote K, Yoshida H, Yoshida M, Deguchi J, Dobashi K, Fujiyoshi A, Hamaguchi H, Hara M, Harada-Shiba M, Hirata T, Iida M, Ikeda Y, Ishibashi S, Kanda H, Kihara S, Kitagawa K, Kodama S, Koseki M, Maezawa Y, Masuda D, Miida T, Miyamoto Y, Nishimura R, Node K, Noguchi M, Ohishi M, Saito I, Sawada S, Sone H, Takemoto M, Wakatsuki A, Yanai H. Japan Atherosclerosis Society (JAS) Guidelines for Prevention of Atherosclerotic Cardiovascular Diseases 2022. J Atheroscler Thromb 2024; 31:641-853. [PMID: 38123343 DOI: 10.5551/jat.gl2022] [Citation(s) in RCA: 69] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Affiliation(s)
- Tomonori Okamura
- Preventive Medicine and Public Health, Keio University School of Medicine
| | | | | | - Yoshio Fujioka
- Faculty of Nutrition, Division of Clinical Nutrition, Kobe Gakuin University
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine
| | - Hirotoshi Ohmura
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka Metropolitan University Graduate school of Medicine
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine
| | - Hiroshi Yoshida
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital
| | | | - Juno Deguchi
- Department of Vascular Surgery, Saitama Medical Center, Saitama Medical University
| | - Kazushige Dobashi
- Department of Pediatrics, School of Medicine, University of Yamanashi
| | | | | | - Masumi Hara
- Department of Internal Medicine, Mizonokuchi Hospital, Teikyo University School of Medicine
| | - Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute
| | - Takumi Hirata
- Institute for Clinical and Translational Science, Nara Medical University
| | - Mami Iida
- Department of Internal Medicine and Cardiology, Gifu Prefectural General Medical Center
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, School of Medicine
- Current affiliation: Ishibashi Diabetes and Endocrine Clinic
| | - Hideyuki Kanda
- Department of Public Health, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Shinji Kihara
- Medical Laboratory Science and Technology, Division of Health Sciences, Osaka University graduate School of medicine
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University Hospital
| | - Satoru Kodama
- Department of Prevention of Noncommunicable Diseases and Promotion of Health Checkup, Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Masahiro Koseki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine
| | - Daisaku Masuda
- Department of Cardiology, Center for Innovative Medicine and Therapeutics, Dementia Care Center, Doctor's Support Center, Health Care Center, Rinku General Medical Center
| | - Takashi Miida
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine
| | | | - Rimei Nishimura
- Department of Diabetes, Metabolism and Endocrinology, The Jikei University School of Medicine
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University
| | - Midori Noguchi
- Division of Public Health, Department of Social Medicine, Graduate School of Medicine, Osaka University
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Isao Saito
- Department of Public Health and Epidemiology, Faculty of Medicine, Oita University
| | - Shojiro Sawada
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Minoru Takemoto
- Department of Diabetes, Metabolism and Endocrinology, International University of Health and Welfare
| | | | - Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital
| |
Collapse
|
6
|
Kobayashi J, Minamizuka T, Tada H, Yokote K. Familial hypercholesterolemia with special focus on Japan. Clin Chim Acta 2024; 556:117847. [PMID: 38417778 DOI: 10.1016/j.cca.2024.117847] [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: 01/23/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Familial hypercholesterolemia (FH) is an inherited disorder characterized by increased low-density lipoprotein LDL) cholesterol and atherosclerotic cardiovascular disease. Although initial genetic analysis linked FH to LDL receptor mutations, subsequent work demonstrated that a gain-of-function mutation in the proprotein convertase subtilisin/kexin type 9 (PCSK9), which causes LDL-R degradation, was shown to be the cause of FH. In this review, we describe the history of research on FH, its clinical phenotyping and genotyping and advances in treatment with special focus on Japan.
Collapse
Affiliation(s)
- Junji Kobayashi
- Department of Endocrinology, Metabolism, Hematology and Geriatrics, Chiba University; Department of Clinical Laboratory Science, Graduate School of Medical Sciences, Kanazawa University.
| | - Takuya Minamizuka
- Department of Endocrinology, Metabolism, Hematology and Geriatrics, Chiba University
| | - Hayato Tada
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University
| | - Koutaro Yokote
- Department of Endocrinology, Metabolism, Hematology and Geriatrics, Chiba University
| |
Collapse
|
7
|
Sadowska A, Osiński P, Roztocka A, Kaczmarz-Chojnacka K, Zapora E, Sawicka D, Car H. Statins-From Fungi to Pharmacy. Int J Mol Sci 2023; 25:466. [PMID: 38203637 PMCID: PMC10779115 DOI: 10.3390/ijms25010466] [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: 11/30/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Statins have been used in the treatment of hyperlipidemia, both as monotherapy and in combination therapy. Natural fermentation processes of fungi such as Monascus spp., Penicillium spp., Aspergillus terreus, and Pleurotus ostreatus have given rise to natural statins. Compactin (mevastatin), the original naturally occurring statin, is the primary biotransformation substrate in the manufacturing process of marketed drugs. Statins are classified into natural, semi-synthetic derivatives of natural statins, and synthetic ones. Synthetic statins differ from natural statins in their structural composition, with the only common feature being the HMG-CoA-like moiety responsible for suppressing HMG-CoA reductase. Statins do not differ significantly regarding their pleiotropic and adverse effects, but their characteristics depend on their pharmacokinetic parameters and chemical properties. This paper focuses on describing the processes of obtaining natural statins, detailing the pharmacokinetics of available statins, divided into natural and synthetic, and indicating their pleiotropic effects.
Collapse
Affiliation(s)
- Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (D.S.); (H.C.)
| | - Patryk Osiński
- Student’s Pharmacological Club, Lazarski University, Świeradowska 43, 02-662 Warsaw, Poland; (P.O.); (A.R.); (K.K.-C.)
| | - Alicja Roztocka
- Student’s Pharmacological Club, Lazarski University, Świeradowska 43, 02-662 Warsaw, Poland; (P.O.); (A.R.); (K.K.-C.)
| | - Karolina Kaczmarz-Chojnacka
- Student’s Pharmacological Club, Lazarski University, Świeradowska 43, 02-662 Warsaw, Poland; (P.O.); (A.R.); (K.K.-C.)
| | - Ewa Zapora
- Department of Silviculture and Forest Use, Institute of Forest Sciences, Bialystok University of Technology, Wiejska 45E, 15351 Bialystok, Poland;
| | - Diana Sawicka
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (D.S.); (H.C.)
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (D.S.); (H.C.)
| |
Collapse
|
8
|
Srivastava RAK. New opportunities in the management and treatment of refractory hypercholesterolemia using in vivo CRISPR-mediated genome/base editing. Nutr Metab Cardiovasc Dis 2023; 33:2317-2325. [PMID: 37805309 DOI: 10.1016/j.numecd.2023.08.010] [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: 05/18/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 10/09/2023]
Abstract
AIMS Refractory hypercholesterolemia (RH), caused primarily by the loss-of-function mutation of LDL receptor (LDLR) gene seen in HoFH and HeFH patients, remains a major risk factor for atherosclerotic cardiovascular disease (ASCVD). Statin and ezetimibe combination therapy lower circulating LDL by 30% in HoFH patients. PCSK9 mAB, being an LDLR-dependent therapy, is not effective in HoFH, but lowers LDL by 25% in HeFH patients. A maximum reduction of 50% was noted in HoFH patients treated with ANGPTL3 mAB, which was not enough to achieve therapeutic goal of LDL. Therefore, new approaches are warranted to offer hopes to individuals intolerant to higher dose statins and not able to achieve recommended LDL level. DATA SYNTHESIS New approaches to lower LDL include gene therapy and gene editing. AAV-based gene therapy has shown encouraging results in animal models. Using CRISPR/Cas9-mediated genome/base editing, gain of function and loss of function have been successfully done in animal models. Recent progress in the refinement of genome/base editing has overcome the issues of off-target mutagenesis with ∼1% mutagenesis in case of PCSK9 and almost no off-target mutagenesis in inactivating ANGPTL3 in animal models showing 50% reduction in cholesterol. Current approaches using CRISPR-Cas9 genome/base editing targeting LDLR-dependent and LDLR-independent pathways are underway. CONCLUSIONS The new information on gain of LDLR function and inactivation of ANGPTL3 together with developments in genome/base editing technology to overcome off-target insertion and deletion mutagenesis offer hope to refractory hypercholesterolemic individuals who are at a higher risk of developing ASCVD.
Collapse
|
9
|
Begue F, Apalama ML, Lambert G, Meilhac O. HDL as a Treatment Target: Should We Abandon This Idea? Curr Atheroscler Rep 2023; 25:1093-1099. [PMID: 38051472 DOI: 10.1007/s11883-023-01176-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2023] [Indexed: 12/07/2023]
Abstract
PURPOSE OF REVIEW High-density lipoproteins (HDL) have long been regarded as an antiatherogenic lipoprotein species by virtue of their role in reverse cholesterol transport (RCT), as well as their established anti-inflammatory and antioxidant properties. For decades, HDL have been an extremely appealing therapeutic target to combat atherosclerotic cardiovascular diseases (ASCVD). RECENT FINDINGS Unfortunately, neither increasing HDL with drugs nor direct infusions of reconstituted HDL have convincedly proven to be positive strategies for cardiovascular health, raising the question of whether we should abandon the idea of considering HDL as a treatment target. The results of two large clinical trials, one testing the latest CETP inhibitor Obicetrapib and the other testing the infusion of patients post-acute coronary events with reconstituted HDL, are still awaited. If they prove negative, these trials will seal the fate of HDL as a direct therapeutic target. However, using HDL as a therapeutic agent still holds promise if we manage to optimize their beneficial properties for not only ASCVD but also outside the cardiovascular field.
Collapse
Affiliation(s)
- Floran Begue
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97410, Saint-Pierre, France
| | - Marie Laurine Apalama
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97410, Saint-Pierre, France
| | - Gilles Lambert
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97410, Saint-Pierre, France.
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97410, Saint-Pierre, France
- CHU de La Réunion, 97400, Saint-Denis, France
| |
Collapse
|
10
|
van der Ende EL, In ‘t Veld SGJG, Hanskamp I, van der Lee S, Dijkstra JIR, Hok-A-Hin YS, Blujdea ER, van Swieten JC, Irwin DJ, Chen-Plotkin A, Hu WT, Lemstra AW, Pijnenburg YAL, van der Flier WM, del Campo M, Teunissen CE, Vermunt L. CSF proteomics in autosomal dominant Alzheimer's disease highlights parallels with sporadic disease. Brain 2023; 146:4495-4507. [PMID: 37348871 PMCID: PMC10629764 DOI: 10.1093/brain/awad213] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023] Open
Abstract
Autosomal dominant Alzheimer's disease (ADAD) offers a unique opportunity to study pathophysiological changes in a relatively young population with few comorbidities. A comprehensive investigation of proteome changes occurring in ADAD could provide valuable insights into AD-related biological mechanisms and uncover novel biomarkers and therapeutic targets. Furthermore, ADAD might serve as a model for sporadic AD, but in-depth proteome comparisons are lacking. We aimed to identify dysregulated CSF proteins in ADAD and determine the degree of overlap with sporadic AD. We measured 1472 proteins in CSF of PSEN1 or APP mutation carriers (n = 22) and age- and sex-matched controls (n = 20) from the Amsterdam Dementia Cohort using proximity extension-based immunoassays (PEA). We compared protein abundance between groups with two-sided t-tests and identified enriched biological pathways. Using the same protein panels in paired plasma samples, we investigated correlations between CSF proteins and their plasma counterparts. Finally, we compared our results with recently published PEA data from an international cohort of sporadic AD (n = 230) and non-AD dementias (n = 301). All statistical analyses were false discovery rate-corrected. We detected 66 differentially abundant CSF proteins (65 increased, 1 decreased) in ADAD compared to controls (q < 0.05). The most strongly upregulated proteins (fold change >1.8) were related to immunity (CHIT1, ITGB2, SMOC2), cytoskeletal structure (MAPT, NEFL) and tissue remodelling (TMSB10, MMP-10). Significant CSF-plasma correlations were found for the upregulated proteins SMOC2 and LILR1B. Of the 66 differentially expressed proteins, 36 had been measured previously in the sporadic dementias cohort, 34 of which (94%) were also significantly upregulated in sporadic AD, with a strong correlation between the fold changes of these proteins in both cohorts (rs = 0.730, P < 0.001). Twenty-nine of the 36 proteins (81%) were also upregulated among non-AD patients with suspected AD co-pathology. This CSF proteomics study demonstrates substantial biochemical similarities between ADAD and sporadic AD, suggesting involvement of the same biological processes. Besides known AD-related proteins, we identified several relatively novel proteins, such as TMSB10, MMP-10 and SMOC2, which have potential as novel biomarkers. With shared pathophysiological CSF changes, ADAD study findings might be translatable to sporadic AD, which could greatly expedite therapy development.
Collapse
Affiliation(s)
- Emma L van der Ende
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sjors G J G In ‘t Veld
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Iris Hanskamp
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sven van der Lee
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Janna I R Dijkstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Elena R Blujdea
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - John C van Swieten
- Alzheimer Center and Department of Neurology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28003 Madrid, Spain
- Barcelonabeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
11
|
Dai W, Zhang H, Lund H, Zhang Z, Castleberry M, Rodriguez M, Kuriakose G, Gupta S, Lewandowska M, Powers HR, Valmiki S, Zhu J, Shapiro AD, Hussain MM, López JA, Sorci-Thomas MG, Silverstein RL, Ginsberg HN, Sahoo D, Tabas I, Zheng Z. Intracellular tPA-PAI-1 interaction determines VLDL assembly in hepatocytes. Science 2023; 381:eadh5207. [PMID: 37651538 PMCID: PMC10697821 DOI: 10.1126/science.adh5207] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/13/2023] [Indexed: 09/02/2023]
Abstract
Apolipoprotein B (apoB)-lipoproteins initiate and promote atherosclerotic cardiovascular disease. Plasma tissue plasminogen activator (tPA) activity is negatively associated with atherogenic apoB-lipoprotein cholesterol levels in humans, but the mechanisms are unknown. We found that tPA, partially through the lysine-binding site on its Kringle 2 domain, binds to the N terminus of apoB, blocking the interaction between apoB and microsomal triglyceride transfer protein (MTP) in hepatocytes, thereby reducing very-low-density lipoprotein (VLDL) assembly and plasma apoB-lipoprotein cholesterol levels. Plasminogen activator inhibitor 1 (PAI-1) sequesters tPA away from apoB and increases VLDL assembly. Humans with PAI-1 deficiency have smaller VLDL particles and lower plasma levels of apoB-lipoprotein cholesterol. These results suggest a mechanism that fine-tunes VLDL assembly by intracellular interactions among tPA, PAI-1, and apoB in hepatocytes.
Collapse
Affiliation(s)
- Wen Dai
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Heng Zhang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Hayley Lund
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ziyu Zhang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | | | - Maya Rodriguez
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- College of Arts and Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sweta Gupta
- Indiana Hemophilia and Thrombosis Center, Indianapolis, IN 46260, USA
| | | | - Hayley R. Powers
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Swati Valmiki
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY 11501, USA
| | - Jieqing Zhu
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amy D. Shapiro
- Indiana Hemophilia and Thrombosis Center, Indianapolis, IN 46260, USA
| | - M. Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY 11501, USA
| | - José A. López
- Bloodworks Research Institute, Seattle, WA 98102, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Mary G. Sorci-Thomas
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Roy L. Silverstein
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Daisy Sahoo
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ze Zheng
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
12
|
Trajanoska K, Bhérer C, Taliun D, Zhou S, Richards JB, Mooser V. From target discovery to clinical drug development with human genetics. Nature 2023; 620:737-745. [PMID: 37612393 DOI: 10.1038/s41586-023-06388-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/29/2023] [Indexed: 08/25/2023]
Abstract
The substantial investments in human genetics and genomics made over the past three decades were anticipated to result in many innovative therapies. Here we investigate the extent to which these expectations have been met, excluding cancer treatments. In our search, we identified 40 germline genetic observations that led directly to new targets and subsequently to novel approved therapies for 36 rare and 4 common conditions. The median time between genetic target discovery and drug approval was 25 years. Most of the genetically driven therapies for rare diseases compensate for disease-causing loss-of-function mutations. The therapies approved for common conditions are all inhibitors designed to pharmacologically mimic the natural, disease-protective effects of rare loss-of-function variants. Large biobank-based genetic studies have the power to identify and validate a large number of new drug targets. Genetics can also assist in the clinical development phase of drugs-for example, by selecting individuals who are most likely to respond to investigational therapies. This approach to drug development requires investments into large, diverse cohorts of deeply phenotyped individuals with appropriate consent for genetically assisted trials. A robust framework that facilitates responsible, sustainable benefit sharing will be required to capture the full potential of human genetics and genomics and bring effective and safe innovative therapies to patients quickly.
Collapse
Affiliation(s)
- Katerina Trajanoska
- Canada Excellence Research Chair in Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - Claude Bhérer
- Canada Excellence Research Chair in Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - Daniel Taliun
- Canada Excellence Research Chair in Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - Sirui Zhou
- Canada Excellence Research Chair in Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - J Brent Richards
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Department of Epidemiology and Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Vincent Mooser
- Canada Excellence Research Chair in Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
13
|
Srivastava RAK. A Review of Progress on Targeting LDL Receptor-Dependent and -Independent Pathways for the Treatment of Hypercholesterolemia, a Major Risk Factor of ASCVD. Cells 2023; 12:1648. [PMID: 37371118 DOI: 10.3390/cells12121648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Since the discovery of the LDL receptor in 1973 by Brown and Goldstein as a causative protein in hypercholesterolemia, tremendous amounts of effort have gone into finding ways to manage high LDL cholesterol in familial hypercholesterolemic (HoFH and HeFH) individuals with loss-of-function mutations in the LDL receptor (LDLR) gene. Statins proved to be the first blockbuster drug, helping both HoFH and HeFH individuals by inhibiting the cholesterol synthesis pathway rate-limiting enzyme HMG-CoA reductase and inducing the LDL receptor. However, statins could not achieve the therapeutic goal of LDL. Other therapies targeting LDLR include PCSK9, which lowers LDLR by promoting LDLR degradation. Inducible degrader of LDLR (IDOL) also controls the LDLR protein, but an IDOL-based therapy is yet to be developed. Among the LDLR-independent pathways, such as angiopoietin-like 3 (ANGPTL3), apolipoprotein (apo) B, apoC-III and CETP, only ANGPTL3 offers the advantage of treating both HoFH and HeFH patients and showing relatively better preclinical and clinical efficacy in animal models and hypercholesterolemic individuals, respectively. While loss-of-LDLR-function mutations have been known for decades, gain-of-LDLR-function mutations have recently been identified in some individuals. The new information on gain of LDLR function, together with CRISPR-Cas9 genome/base editing technology to target LDLR and ANGPTL3, offers promise to HoFH and HeFH individuals who are at a higher risk of developing atherosclerotic cardiovascular disease (ASCVD).
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions LLC, Boston, MA 02101-02117, USA
- College of Professional Studies, Northeastern University, Boston, MA 02101-02117, USA
| |
Collapse
|
14
|
Kallapur A, Sallam T. Pharmacotherapy in familial hypercholesterolemia - Current state and emerging paradigms. Trends Cardiovasc Med 2023; 33:170-179. [PMID: 34968676 DOI: 10.1016/j.tcm.2021.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/01/2022]
Abstract
Familial hypercholesterolemia is a highly prevalent but underdiagnosed disease marked by increased risk of cardiovascular morbidity and mortality. Aggressive reduction of LDL-cholesterol is a hallmark of cardiovascular risk mitigation in familial hypercholesterolemia. More recently, we have witnessed an expanded repertoire of pharmacologic agents that directly target LDL-cholesterol and/or reduce heart disease burden. In this state-of-the-art review, we explore the development, clinical efficacy and limitations of existing and potential future therapeutics in familial hypercholesterolemia.
Collapse
Affiliation(s)
- Aneesh Kallapur
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, CA, United States; Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, United States
| | - Tamer Sallam
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, CA, United States; Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, United States.
| |
Collapse
|
15
|
Jeeyavudeen MS, Pappachan JM, Arunagirinathan G. Statin-related Muscle Toxicity: An Evidence-based Review. TOUCHREVIEWS IN ENDOCRINOLOGY 2022; 18:89-95. [PMID: 36694885 PMCID: PMC9835810 DOI: 10.17925/ee.2022.18.2.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/12/2022] [Indexed: 12/12/2022]
Abstract
The efficacy of statins in the primary and secondary prevention of cardiovascular disease has been proven beyond doubt. The number needed to treat to prevent one cardiovascular event is 1 in 30 over 10 years, and the number needed to treat for secondary prevention is much lower. However, a recent study demonstrated that only 68% of eligible patients are on statin therapy. Moreover, there seems to be a reluctance to escalate statin doses due to the fear of adverse effects. The adverse effects that worries patients and their physicians most frequently are those related to muscular symptoms. N-of-1 trial evidence suggests that muscular symptoms attributed to statins are often caused by the nocebo effect. This article aims to provide a structured, evidence-based approach to suspected statin-related muscle toxicity.
Collapse
Affiliation(s)
| | - Joseph M Pappachan
- Lancashire Teaching Hospitals NHS Trust, Lancashire, UK
- Manchester Metropolitan University, Manchester, UK
| | | |
Collapse
|
16
|
Zechner J, Britza SM, Farrington R, Byard RW, Musgrave IF. Flavonoid-statin interactions causing myopathy and the possible significance of OATP transport, CYP450 metabolism and mevalonate synthesis. Life Sci 2021; 291:119975. [PMID: 34560084 DOI: 10.1016/j.lfs.2021.119975] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 11/16/2022]
Abstract
3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors, statins, are a primary treatment for hyperlipidemic cardiovascular diseases which are a leading global cause of death. Statin therapy is life saving and discontinuation due to adverse events such as myotoxicity may lead to unfavourable outcomes. There is no known mechanism for statin-induced myotoxicity although it is theorized that it is due to inhibition of downstream products of the HMG-CoA pathway. It is known that drug-drug interactions with conventional medicines exacerbate the risk of statin-induced myotoxicity, though little attention has been paid to herb-drug interactions with complementary medicines. Flavonoids are a class of phytochemicals which can be purchased as high dose supplements. There is evidence that flavonoids can raise statin plasma levels, increasing the risk of statin-induced myopathy. This could be due to pharmacokinetic interactions involving hepatic cytochrome 450 (CYP450) metabolism and organic anion transporter (OATP) absorption. There is also the potential for flavonoids to directly and indirectly inhibit HMG-CoA reductase which could contraindicate statin-therapy. This review aims to discuss what is currently known about the potential for high dose flavonoids to interact with the hepatic CYP450 metabolism, OATP uptake of statins or their ability to interact with HMG-CoA reductase. Flavonoids of particular interest will be covered and the difficulties of examining herbal products will be discussed throughout.
Collapse
Affiliation(s)
- Joshua Zechner
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Susan M Britza
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Rachael Farrington
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Roger W Byard
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Forensic Science SA, Adelaide, SA 5000, Australia
| | - Ian F Musgrave
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
17
|
Siasos G, Oikonomou E, Tousoulis D. Alirocumab and evolocumab: an indirect comparison of cardiovascular benefits. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 7:236-237. [PMID: 32375175 DOI: 10.1093/ehjcvp/pvaa031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Gerasimos Siasos
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Oikonomou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Tousoulis
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
18
|
Rynearson KD, Ponnusamy M, Prikhodko O, Xie Y, Zhang C, Nguyen P, Hug B, Sawa M, Becker A, Spencer B, Florio J, Mante M, Salehi B, Arias C, Galasko D, Head BP, Johnson G, Lin JH, Duddy SK, Rissman RA, Mobley WC, Thinakaran G, Tanzi RE, Wagner SL. Preclinical validation of a potent γ-secretase modulator for Alzheimer's disease prevention. J Exp Med 2021; 218:211838. [PMID: 33651103 PMCID: PMC7931646 DOI: 10.1084/jem.20202560] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/20/2022] Open
Abstract
A potent γ-secretase modulator (GSM) has been developed to circumvent problems associated with γ-secretase inhibitors (GSIs) and to potentially enable use in primary prevention of early-onset familial Alzheimer's disease (EOFAD). Unlike GSIs, GSMs do not inhibit γ-secretase activity but rather allosterically modulate γ-secretase, reducing the net production of Aβ42 and to a lesser extent Aβ40, while concomitantly augmenting production of Aβ38 and Aβ37. This GSM demonstrated robust time- and dose-dependent efficacy in acute, subchronic, and chronic studies across multiple species, including primary and secondary prevention studies in a transgenic mouse model. The GSM displayed a >40-fold safety margin in rats based on a comparison of the systemic exposure (AUC) at the no observed adverse effect level (NOAEL) to the 50% effective AUC or AUCeffective, the systemic exposure required for reducing levels of Aβ42 in rat brain by 50%.
Collapse
Affiliation(s)
- Kevin D Rynearson
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Moorthi Ponnusamy
- Department of Molecular Medicine and Byrd Alzheimer's Institute, University of South Florida, Morsani College of Medicine, Tampa, FL
| | - Olga Prikhodko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Yuhuan Xie
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Can Zhang
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA
| | - Phuong Nguyen
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Brenda Hug
- Veterans Administration San Diego Healthcare System, La Jolla, CA
| | - Mariko Sawa
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Ann Becker
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Brian Spencer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Jazmin Florio
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Bahar Salehi
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Carlos Arias
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Brian P Head
- Veterans Administration San Diego Healthcare System, La Jolla, CA.,Department of Anesthesiology, University of California, San Diego, La Jolla, CA
| | | | | | - Steven K Duddy
- Integrated Nonclinical Development Solutions, Ann Arbor, MI
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA.,Veterans Administration San Diego Healthcare System, La Jolla, CA
| | - William C Mobley
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Gopal Thinakaran
- Department of Molecular Medicine and Byrd Alzheimer's Institute, University of South Florida, Morsani College of Medicine, Tampa, FL
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA
| | - Steven L Wagner
- Department of Neurosciences, University of California, San Diego, La Jolla, CA.,Veterans Administration San Diego Healthcare System, La Jolla, CA
| |
Collapse
|
19
|
Markowska A, Antoszczak M, Markowska J, Huczyński A. Statins: HMG-CoA Reductase Inhibitors as Potential Anticancer Agents against Malignant Neoplasms in Women. Pharmaceuticals (Basel) 2020; 13:ph13120422. [PMID: 33255609 PMCID: PMC7760915 DOI: 10.3390/ph13120422] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Statins, also known as HMG-CoA inhibitors, are a class of bioactive small molecules that efficiently reduce the levels of cholesterol, and therefore are commonly used to manage and prevent various cardiovascular diseases. With respect to their original medical indications, statins are currently in the group of the most prescribed drugs worldwide. Of note is that statins are perceived actually rather as agents that have pleiotropic activities; in addition to their inhibitory activity on the production of endogenous cholesterol. Statins may also affect cell proliferation, angiogenesis and/or migration (metastasis) of different cancer cells, and play a positive role in the chemoprevention of cancer, thus being the excellent candidates to be repurposed in oncology. Particularly intriguing in this context seems to be the promising role of statins on both the incidence and course of common malignant neoplasms in women. In this article, we review and discuss the effect of the use of statins in the treatment of three types of cancer, i.e., breast, endometrial and ovarian cancer, with the highest mortality among gynecological cancers.
Collapse
Affiliation(s)
- Anna Markowska
- Department of Perinatology and Women’s Health, Poznań University of Medical Sciences, 60-535 Poznań, Poland;
| | - Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61–614 Poznań, Poland;
| | - Janina Markowska
- Department of Oncology, Gynecological Oncology, Poznań University of Medical Sciences, 60-569 Poznań, Poland;
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61–614 Poznań, Poland;
- Correspondence: ; Tel.: +48-61-829-1673
| |
Collapse
|
20
|
Statin use is associated with lower disease severity in COVID-19 infection. Sci Rep 2020; 10:17458. [PMID: 33060704 PMCID: PMC7562925 DOI: 10.1038/s41598-020-74492-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/29/2020] [Indexed: 12/30/2022] Open
Abstract
We aim to study the association of hyperlipidemia and statin use with COVID-19 severity. We analysed a retrospective cohort of 717 patients admitted to a tertiary centre in Singapore for COVID-19 infection. Clinical outcomes of interest were oxygen saturation ≤ 94% requiring supplemental oxygen, intensive-care unit (ICU) admission, invasive mechanical-ventilation and death. Patients on long term dyslipidaemia medications (statins, fibrates or ezetimibe) were considered to have dyslipidaemia. Logistic regression models were used to study the association between dyslipidaemia and clinical outcomes adjusted for age, gender and ethnicity. Statin treatment effect was determined, in a nested case-control design, through logistic treatment models with 1:3 propensity matching for age, gender and ethnicity. All statistical tests were two-sided, and statistical significance was taken as p < 0.05. One hundred fifty-six (21.8%) patients had dyslipidaemia and 97% of these were on statins. Logistic treatment models showed a lower chance of ICU admission for statin users when compared to non-statin users (ATET: Coeff (risk difference): - 0.12 (- 0.23, - 0.01); p = 0.028). There were no other significant differences in other outcomes. Statin use was independently associated with lower ICU admission. This supports current practice to continue prescription of statins in COVID-19 patients.
Collapse
|
21
|
Ray KK, Corral P, Morales E, Nicholls SJ. Pharmacological lipid-modification therapies for prevention of ischaemic heart disease: current and future options. Lancet 2019; 394:697-708. [PMID: 31448741 DOI: 10.1016/s0140-6736(19)31950-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/24/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
Abstract
Atherosclerosis and its clinical manifestation as ischaemic heart disease remains a considerable health burden. Given that many factors contribute to ischaemic heart disease, a multifactorial approach to prevention is recommended, starting with lifestyle advice, smoking cessation, and control of known cardiovascular risk factors, such as blood pressure and lipids. Within the lipid profile, the principal target is lowering LDL cholesterol, firstly with lifestyle interventions and subsequently with pharmacological therapy. Statins are the recommended first-line pharmacological treatment. Some individuals might require further lowering of LDL cholesterol or be unable to tolerate statins. Additional therapies targeting different pathways in cholesterol metabolism are now available, ranging from small molecules taken orally, to injectable therapies. Examples include ezetimibe, which targets Niemann-Pick C1-like protein, and monoclonal antibodies that target PCSK9. Phase 3 trials have also been completed for bempedoic acid (targeting ATP-citrate lyase) and inclisiran (an interference RNA-based therapeutic targeting hepatic PCSK9 synthesis). In addition to LDL cholesterol, mendelian randomisation studies support a causal role for lipoprotein(a) and triglycerides in ischaemic heart disease. In this Series paper, we appraise currently available and emerging therapies for lowering LDL cholesterol, lipoprotein(a), and triglycerides for prevention of ischaemic heart disease.
Collapse
Affiliation(s)
- Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, School of Public Health, Imperial College London, London, UK.
| | - Pablo Corral
- Pharmacology Department, School of Medicine, FASTA University, Mar del Plata, Argentina
| | - Enrique Morales
- Cardiometabolic Research Center, MAC Hospital, Aguascalientes, Mexico
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
22
|
Kinoshita M, Yokote K, Arai H, Iida M, Ishigaki Y, Ishibashi S, Umemoto S, Egusa G, Ohmura H, Okamura T, Kihara S, Koba S, Saito I, Shoji T, Daida H, Tsukamoto K, Deguchi J, Dohi S, Dobashi K, Hamaguchi H, Hara M, Hiro T, Biro S, Fujioka Y, Maruyama C, Miyamoto Y, Murakami Y, Yokode M, Yoshida H, Rakugi H, Wakatsuki A, Yamashita S, Committee for Epidemiology and Clinical Management of Atherosclerosis. Japan Atherosclerosis Society (JAS) Guidelines for Prevention of Atherosclerotic Cardiovascular Diseases 2017. J Atheroscler Thromb 2018; 25:846-984. [PMID: 30135334 PMCID: PMC6143773 DOI: 10.5551/jat.gl2017] [Citation(s) in RCA: 566] [Impact Index Per Article: 80.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/11/2018] [Indexed: 11/30/2022] Open
Affiliation(s)
| | - Koutaro Yokote
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidenori Arai
- National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Mami Iida
- Department of Internal Medicine and Cardiology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Yasushi Ishigaki
- Division of Diabetes and Metabolism, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Seiji Umemoto
- Center for Integrated Medical Research, Hiroshima University Hospital, Hiroshima, Japan
| | | | - Hirotoshi Ohmura
- Department of Cardiovascular Medicine, Juntendo University, Tokyo, Japan
| | - Tomonori Okamura
- Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Kihara
- Biomedical Informatics, Osaka University, Osaka, Japan
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Isao Saito
- Department of Community Health Systems Nursing, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University, Tokyo, Japan
| | - Kazuhisa Tsukamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Juno Deguchi
- Department of Vascular Surgery, Saitama Medical Center, Saitama, Japan
| | - Seitaro Dohi
- Chief Health Management Department, Mitsui Chemicals Inc., Tokyo, Japan
| | - Kazushige Dobashi
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | | | - Masumi Hara
- Department of Internal Medicine, Mizonokuchi Hospital, Teikyo University School of Medicine, Kanagawa, Japan
| | - Takafumi Hiro
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | | | - Yoshio Fujioka
- Faculty of Nutrition, Division of Clinical Nutrition, Kobe Gakuin University, Hyogo, Japan
| | - Chizuko Maruyama
- Department of Food and Nutrition, Faculty of Human Sciences and Design, Japan Women's University, Tokyo, Japan
| | - Yoshihiro Miyamoto
- Department of Preventive Cardiology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Masayuki Yokode
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Yoshida
- Department of Laboratory Medicine, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akihiko Wakatsuki
- Department of Obstetrics and Gynecology, Aichi Medical University, Aichi, Japan
| | - Shizuya Yamashita
- Department of Community Medicine, Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Rinku General Medical Center, Osaka, Japan
| | | |
Collapse
|
23
|
Robinson JR, Denny JC, Roden DM, Van Driest SL. Genome-wide and Phenome-wide Approaches to Understand Variable Drug Actions in Electronic Health Records. Clin Transl Sci 2018; 11:112-122. [PMID: 29148204 PMCID: PMC5866959 DOI: 10.1111/cts.12522] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/14/2017] [Indexed: 12/24/2022] Open
Affiliation(s)
- Jamie R. Robinson
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Joshua C. Denny
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Dan M. Roden
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of PharmacologyVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Sara L. Van Driest
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of PediatricsVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
24
|
Lipid lowering agents of natural origin: An account of some promising chemotypes. Eur J Med Chem 2017; 140:331-348. [DOI: 10.1016/j.ejmech.2017.09.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/07/2017] [Accepted: 09/12/2017] [Indexed: 12/22/2022]
|
25
|
Kim RG, Loomba R, Prokop LJ, Singh S. Statin Use and Risk of Cirrhosis and Related Complications in Patients With Chronic Liver Diseases: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol 2017; 15:1521-1530.e8. [PMID: 28479502 PMCID: PMC5605397 DOI: 10.1016/j.cgh.2017.04.039] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/28/2017] [Accepted: 04/20/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Statins have been variably shown to decrease risk and complications of chronic liver diseases (CLDs). We performed a systematic review and meta-analysis to evaluate the association between statins and risk of cirrhosis and related complications in patients with CLDs. METHODS Through a systematic literature search up to March 2017, we identified 13 studies (3 randomized trials, 10 cohort studies) in adults with CLDs, reporting the association between statin use and risk of development of cirrhosis, decompensated cirrhosis, improvements in portal hypertension, or mortality. Pooled relative risk (RR) estimates with 95% confidence interval (CIs) were calculated using random effects model. Grading of Recommendations Assessment, Development and Evaluation criteria were used to assess quality of evidence. RESULTS Among 121,058 patients with CLDs (84.5% with hepatitis C), 46% were exposed to statins. In patients with cirrhosis, statin use was associated with 46% lower risk of hepatic decompensation (4 studies; RR, 0.54; 95% CI, 0.46-0.62; I2 = 0%; moderate-quality evidence), and 46% lower mortality (5 studies; RR, 0.54; 95% CI, 0.47-0.61; I2 = 10%; moderate-quality evidence). In patients with CLD without cirrhosis, statin use was associated with a nonsignificant (58% lower) risk of development of cirrhosis or fibrosis progression (5 studies; RR, 0.42; 95% CI, 0.16-1.11; I2 = 99%; very-low-quality evidence). In 3 randomized controlled trials, statin use was associated with 27% lower risk of variceal bleeding or progression of portal hypertension (hazard ratio, 0.73; 95% CI, 0.59-0.91; I2 = 0%; moderate-quality evidence). CONCLUSIONS Based on a systematic review and meta-analysis, statin use is probably associated with lower risk of hepatic decompensation and mortality, and might reduce portal hypertension, in patients with CLDs. Prospective observational studies and randomized controlled trials are needed to confirm this observation.
Collapse
Affiliation(s)
- Rebecca G Kim
- Division of Internal Medicine, Department of Medicine, University of California at San Diego, La Jolla, California
| | - Rohit Loomba
- Division of Gastroenterology, Department of Medicine, University of California at San Diego, La Jolla, California; NAFLD Research Center, Department of Medicine, University of California at San Diego, La Jolla, California; Division of Epidemiology, Department of Family Medicine and Public Health, University of California at San Diego, La Jolla, California
| | - Larry J Prokop
- Department of Library Services, Mayo Clinic, Rochester, Minnesota
| | - Siddharth Singh
- Division of Gastroenterology, Department of Medicine, University of California at San Diego, La Jolla, California; Division of Biomedical Informatics, Department of Medicine, University of California at San Diego, La Jolla, California.
| |
Collapse
|
26
|
Endo A. Discovery and Development of Statins. Nat Prod Commun 2017. [DOI: 10.1177/1934578x1701200801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Mevastatin (Compactin) belonging to the statin class is a hypolipidemic agent. It was isolated from the ascomycetous fungus, Penicillium citrinum by A. Endo in the 1970s, and he discovered that it acted as 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase inhibitor. Mevastatin is considered the first statin drug. The clinical trials on mevastatin were carried out in the late 1970s in Japan. The first commercial statin drug was lovastatin, the second statin, which was found in Aspergillus terreus by Merc and Endo, independently in 1979. Since lovastatin was commercialized, 10 statins were synthesized among which paravastatin was launched to the market by Sankyo in 1989. Pravastatin is used in the lowering of cholesterol and preventing cardiovascular disease. At present, statins are the largest-selling class of drugs currently taken by patients throughout the world. Sales for this one class of drugs on 2005 were $25 billion. Today an estimated 40 million people worldwide are taking statin drugs.
Collapse
Affiliation(s)
- Akira Endo
- Biopharm Research Laboratories, Inc., Tokyo, Japan
| |
Collapse
|
27
|
Abstract
Familial hypercholesterolemia (FH) is a disease characterized by a triad: elevated low-density lipoprotein (LDL) cholesterol, tendon xanthomas, and premature coronary heart disease. Thus, it can be considered as a model disease for hypercholesterolemia and atherosclerotic cardiovascular disease (ASCVD). For the diagnosis of hetero-FH, the detection of Achilles tendon xanthomas by palpation or on X-ray is an indispensable diagnostic skill in clinical lipidology. To prevent the under-diagnosis and under-treatment of FH, the diagnostic criteria should be more convenient and user-friendly. For a patient with cutaneous or tendon xanthomas, the probability of FH is very high; however, an absence of xanthoma does not rule out FH. Brown and Goldstein elucidated the pathogenesis of FH by their work on LDL-receptor (LDL-R), for which they were awarded the Nobel Prize in 1985. In the 1950s, FH patients were divided into heterozygous (hetero-) and homozygous (homo-) FH, and diagnosing homo- and hetero-FH based on the phenotypic features of ASCVD or xanthomas frequently became difficult without the DNA analysis of FH genes. It is estimated that heterozygous mutations in the LDL-R or the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene will be found at a combined frequency of 0.005, which corresponds to 1/199 people in the general population in Japan. Statins and anti-PCSK9 monoclonal antibodies are highly specific and efficient drugs for treating hetero- or homo-FH patients. Most clinical studies have reported an amelioration of ASCVD using long-term statin therapy. Clinical results using anti-PCSK9 monoclonal antibodies will emerge in a few years. In homo-FH patients, mipomersen and lomitapide are expected to yield good results. It is important to sequentially unravel the unrecognized pathogenetic mechanisms of FH to reduce its under-recognition and develop new management strategies for it.
Collapse
Affiliation(s)
- Hiroshi Mabuchi
- Division of Clinical Lipidology, Department of Cardiology, Kanazawa University
| |
Collapse
|
28
|
PCSK9: the Critical Role of Familial Hypercholesterolemia from Discovery to Benefit for all. Cardiovasc Drugs Ther 2016; 30:427-431. [DOI: 10.1007/s10557-016-6690-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
29
|
Im S, Kim BH, Lee K, Kwack K, Yim SV. Screening study for genetic polymorphisms affecting pharmacokinetics of simvastatin. Transl Clin Pharmacol 2016. [DOI: 10.12793/tcp.2016.24.1.43] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Sohee Im
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Bo-Hyung Kim
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Kidong Lee
- Department of BioMedical Science, College of Life Science, CHA University, Pangyo-ro, Bundang-gu, SeongNam 13488, Korea
| | - KyuBum Kwack
- Department of BioMedical Science, College of Life Science, CHA University, Pangyo-ro, Bundang-gu, SeongNam 13488, Korea
| | - Sung-Vin Yim
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
30
|
From the Editor: The Akira Endo Award. Foreword. J Clin Lipidol 2015; 9:479-83. [PMID: 26228664 DOI: 10.1016/j.jacl.2015.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/03/2015] [Indexed: 11/23/2022]
|
31
|
Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell 2015; 161:161-172. [PMID: 25815993 PMCID: PMC4525717 DOI: 10.1016/j.cell.2015.01.036] [Citation(s) in RCA: 793] [Impact Index Per Article: 79.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Indexed: 01/06/2023]
Abstract
One-fourth of all deaths in industrialized countries result from coronary heart disease. A century of research has revealed the essential causative agent: cholesterol-carrying low-density lipoprotein (LDL). LDL is controlled by specific receptors (LDLRs) in liver that remove it from blood. Mutations that eliminate LDLRs raise LDL and cause heart attacks in childhood, whereas mutations that raise LDLRs reduce LDL and diminish heart attacks. If we are to eliminate coronary disease, lowering LDL should be the primary goal. Effective means to achieve this goal are currently available. The key questions are: who to treat, when to treat, and how long to treat.
Collapse
Affiliation(s)
- Joseph L Goldstein
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Michael S Brown
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
32
|
Affiliation(s)
- Hiroshi Mabuchi
- Department of Lipidology, Kanazawa University Graduate School of Medical Science, Takara-machi 13-1, Kanazawa, 920-8640, Japan
| | - Atsushi Nohara
- Department of Lipidology, Kanazawa University Graduate School of Medical Science, Takara-machi 13-1, Kanazawa, 920-8640, Japan
| |
Collapse
|
33
|
Grundy SM. Statins: definitive translational research. Mol Med 2014; 20 Suppl 1:S20-3. [PMID: 25549229 PMCID: PMC4374515 DOI: 10.2119/molmed.2014.00194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 11/06/2022] Open
Affiliation(s)
- Scott M Grundy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Medical Service, Veterans Affairs Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
34
|
Shinohara M, Sato N, Shimamura M, Kurinami H, Hamasaki T, Chatterjee A, Rakugi H, Morishita R. Possible modification of Alzheimer's disease by statins in midlife: interactions with genetic and non-genetic risk factors. Front Aging Neurosci 2014; 6:71. [PMID: 24795626 PMCID: PMC4005936 DOI: 10.3389/fnagi.2014.00071] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/30/2014] [Indexed: 12/28/2022] Open
Abstract
The benefits of statins, commonly prescribed for hypercholesterolemia, in treating Alzheimer's disease (AD) have not yet been fully established. A recent randomized clinical trial did not show any therapeutic effects of two statins on cognitive function in AD. Interestingly, however, the results of the Rotterdam study, one of the largest prospective cohort studies, showed reduced risk of AD in statin users. Based on the current understanding of statin actions and AD pathogenesis, it is still worth exploring whether statins can prevent AD when administered decades before the onset of AD or from midlife. This review discusses the possible beneficial effects of statins, drawn from previous clinical observations, pathogenic mechanisms, which include β-amyloid (Aβ) and tau metabolism, genetic and non-genetic risk factors (apolipoprotein E, cholesterol, sex, hypertension, and diabetes), and other clinical features (vascular dysfunction and oxidative and inflammatory stress) of AD. These findings suggest that administration of statins in midlife might prevent AD in late life by modifying genetic and non-genetic risk factors for AD. It should be clarified whether statins inhibit Aβ accumulation, tau pathological features, and brain atrophy in humans. To answer this question, a randomized controlled study using amyloid positron emission tomography (PET), tau-PET, and magnetic resonance imaging would be useful. This clinical evaluation could help us to overcome this devastating disease.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Naoyuki Sato
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Munehisa Shimamura
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Hitomi Kurinami
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Toshimitsu Hamasaki
- Department of Biomedical Statistics, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Amarnath Chatterjee
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| |
Collapse
|
35
|
Mabuchi H, Nohara A. MTP inhibitor for treating severe LDL cholesterolemia. ACTA ACUST UNITED AC 2013. [DOI: 10.2217/clp.13.38] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
36
|
Wagner SL, Tanzi RE, Mobley WC, Galasko D. Potential use of γ-secretase modulators in the treatment of Alzheimer disease. ACTA ACUST UNITED AC 2013; 69:1255-8. [PMID: 22801784 DOI: 10.1001/archneurol.2012.540] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Although significant progress has occurred in the past 20 years regarding our understanding of Alzheimer disease pathogenesis, we have yet to identify disease-modifying therapeutics capable of substantially altering the clinical course of this prevalent neurodegenerative disease. In this short review, we discuss 2 approaches that are currently being tested clinically (γ-secretase inhibition and γ-secretase modulation) and emphasize the significant differences between these 2 therapeutic approaches. We also discuss certain genetic- and biomarker-based translational and clinical trial paradigms that may assist in developing a useful therapeutic agent.
Collapse
Affiliation(s)
- Steven L Wagner
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92063, USA.
| | | | | | | |
Collapse
|
37
|
Teramoto T, Sasaki J, Ishibashi S, Birou S, Daida H, Dohi S, Egusa G, Hiro T, Hirobe K, Iida M, Kihara S, Kinoshita M, Maruyama C, Ohta T, Okamura T, Yamashita S, Yokode M, Yokote K. Treatment B) Drug Therapy. J Atheroscler Thromb 2013; 20:850-60. [DOI: 10.5551/jat.19166] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
38
|
Long Term Clinical Tolerance of Lovastatin (Mevinolin) and Simvastatin (Epistatin). ACTA ACUST UNITED AC 2012. [DOI: 10.1007/bf03258198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
|
40
|
Affiliation(s)
- Rachel Hajar
- Department of Cardiology, Heart Hospital, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
41
|
Sabri M, Macdonald RL. Statins: A Potential Therapeutic Addition to Treatment for Aneurysmal Subarachnoid Hemorrhage? World Neurosurg 2010; 73:646-53. [DOI: 10.1016/j.wneu.2010.03.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Accepted: 03/19/2010] [Indexed: 11/16/2022]
|
42
|
Endo A. A historical perspective on the discovery of statins. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2010; 86:484-93. [PMID: 20467214 PMCID: PMC3108295 DOI: 10.2183/pjab.86.484] [Citation(s) in RCA: 312] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 03/18/2010] [Indexed: 05/04/2023]
Abstract
Cholesterol is essential for the functioning of all human organs, but it is nevertheless the cause of coronary heart disease. Over the course of nearly a century of investigation, scientists have developed several lines of evidence that establish the causal connection between blood cholesterol, atherosclerosis, and coronary heart disease. Building on that knowledge, scientists and the pharmaceutical industry have successfully developed a remarkably effective class of drugs--the statins--that lower cholesterol levels in blood and reduce the frequency of heart attacks.
Collapse
Affiliation(s)
- Akira Endo
- Biopharm Research Laboratories, Inc., Tokyo 181-0013, Japan.
| |
Collapse
|
43
|
Harada-Shiba M, Sugisawa T, Makino H, Abe M, Tsushima M, Yoshimasa Y, Yamashita T, Miyamoto Y, Yamamoto A, Tomoike H, Yokoyama S. Impact of Statin Treatment on the Clinical Fate of Heterozygous Familial Hypercholesterolemia. J Atheroscler Thromb 2010; 17:667-74. [DOI: 10.5551/jat.4143] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
44
|
Thadhani R. Is calcitriol life-protective for patients with chronic kidney disease? J Am Soc Nephrol 2009; 20:2285-90. [PMID: 19679671 DOI: 10.1681/asn.2009050494] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Ravi Thadhani
- Renal Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
45
|
Bateman RJ, Siemers ER, Mawuenyega KG, Wen G, Browning KR, Sigurdson WC, Yarasheski KE, Friedrich SW, Demattos RB, May PC, Paul SM, Holtzman DM. A gamma-secretase inhibitor decreases amyloid-beta production in the central nervous system. Ann Neurol 2009; 66:48-54. [PMID: 19360898 PMCID: PMC2730994 DOI: 10.1002/ana.21623] [Citation(s) in RCA: 244] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Accumulation of amyloid-beta (Abeta) by overproduction or underclearance in the central nervous system (CNS) is hypothesized to be a necessary event in the pathogenesis of Alzheimer's disease. However, previously, there has not been a method to determine drug effects on Abeta production or clearance in the human CNS. The objective of this study was to determine the effects of a gamma-secretase inhibitor on the production of Abeta in the human CNS. METHODS We utilized a recently developed method of stable-isotope labeling combined with cerebrospinal fluid sampling to directly measure Abeta production during treatment of a gamma-secretase inhibitor, LY450139. We assessed whether this drug could decrease CNS Abeta production in healthy men (age range, 21-50 years) at single oral doses of 100, 140, or 280mg (n = 5 per group). RESULTS LY450139 significantly decreased the production of CNS Abeta in a dose-dependent fashion, with inhibition of Abeta generation of 47, 52, and 84% over a 12-hour period with doses of 100, 140, and 280mg, respectively. There was no difference in Abeta clearance. INTERPRETATION Stable isotope labeling of CNS proteins can be utilized to assess the effects of drugs on the production and clearance rates of proteins targeted as potential disease-modifying treatments for Alzheimer's disease and other CNS disorders. Results from this approach can assist in making decisions about drug dosing and frequency in the design of larger and longer clinical trials for diseases such as Alzheimer's disease, and may accelerate effective drug validation. Ann Neurol 2009.
Collapse
Affiliation(s)
- Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Montecucco F, Mach F. Update on statin-mediated anti-inflammatory activities in atherosclerosis. Semin Immunopathol 2009; 31:127-142. [PMID: 19415282 DOI: 10.1007/s00281-009-0150-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 04/15/2009] [Indexed: 12/28/2022]
Abstract
Anti-inflammatory activities of statins in atherosclerosis have been well documented by both basic research and clinical studies. Statins have been introduced in the 1980s as 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors to block cholesterol synthesis and lower cholesterol serum levels. In the last three decades, statins have been shown to possess several anti-inflammatory and antioxidant activities resulting in the beneficial reduction of atherosclerotic processes and cardiovascular risk in both humans and animal models. Inflammatory intracellular pathways involving kinase phosphorylation and protein prenylation are modulated by statins. The same intracellular mechanisms might also cause statin-induced myotoxicity. In the present review, we will update evidence on statin-mediated regulation of inflammatory pathways in atherogenesis.
Collapse
Affiliation(s)
- Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, University Hospital, Geneva, Switzerland.
| | | |
Collapse
|
47
|
Affiliation(s)
- Akira Endo
- Biopharm Research Laboratories, Inc., 3-41-3 501 Shimorenjaku, Mitaka, Tokyo 181-0013, Japan.
| |
Collapse
|
48
|
Teramoto T, Sasaki J, Ueshima H, Egusa G, Kinoshita M, Shimamoto K, Daida H, Biro S, Hirobe K, Funahashi T, Yokote K, Yokode M. Treatment - Drug Therapy. J Atheroscler Thromb 2008; 15:167-78. [DOI: 10.5551/jat.e619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
49
|
Lin YL, Wang TH, Lee MH, Su NW. Biologically active components and nutraceuticals in the Monascus-fermented rice: a review. Appl Microbiol Biotechnol 2007; 77:965-73. [PMID: 18038131 DOI: 10.1007/s00253-007-1256-6] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 10/24/2007] [Accepted: 10/25/2007] [Indexed: 11/25/2022]
Abstract
Monascus-fermented rice has traditionally been used as a natural food colorant and food preservative of meat and fish for centuries. It has recently become a popular dietary supplement because of many of its bioactive constituents being discovered, including a series of active drug compounds, monacolins, indicated as the 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors for reducing serum cholesterol level. The controversy of its safety has been provoked because a mycotoxin, citrinin, is also produced along with the Monascus secondary metabolites by certain strains or under certain cultivation conditions. This review introduces the basic production process and addresses on the compounds with bioactive functions. Current advances in avoiding the harmful ingredient citrinin are also discussed.
Collapse
Affiliation(s)
- Yii-Lih Lin
- Department of Agricultural Chemistry, National Taiwan University, no. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | | | | | | |
Collapse
|
50
|
Berthold HK, Naini A, Di Mauro S, Hallikainen M, Gylling H, Krone W, Gouni-Berthold I. Effect of ezetimibe and/or simvastatin on coenzyme Q10 levels in plasma: a randomised trial. Drug Saf 2006; 29:703-12. [PMID: 16872244 DOI: 10.2165/00002018-200629080-00007] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND HMG-CoA reductase inhibitors ('statins') have been associated with a decrease in ubidecarenone (ubiquinone) levels, a lipophilic enzyme also known as coenzyme Q10 (CoQ10), due to inhibition of mevalonate synthesis. There is speculation that a decrease in CoQ10 levels may be associated with statin-induced myopathy. The cholesterol absorption inhibitor ezetimibe increases endogenous cholesterol synthesis. The purpose of this study was to examine (i) the effects of ezetimibe and simvastatin on plasma CoQ10 levels and (ii) whether ezetimibe coadministered with simvastatin abrogates the suggested statin-induced decrease in the CoQ10 plasma levels. METHODS Seventy-two healthy male subjects were enrolled in a single-centre, randomised, parallel-group study with three arms. Subjects received ezetimibe 10 mg/day, simvastatin 40 mg/day or the combination of ezetimibe 10 mg/day plus simvastatin 40 mg/day for 14 days. RESULTS Baseline CoQ10 (0.99 +/- 0.30 mg/L) levels for the combined groups remained unchanged in the ezetimibe group (0.95 +/- 0.24 mg/L), and significantly decreased in the simvastatin and combination groups (0.82 +/- 0.18 mg/L, p = 0.0002 and 0.7 +/- 0.22 mg/L, p < 0.0001, respectively). There was a correlation between the percentage change in the levels of low-density lipoprotein-cholesterol (LDL-C) and the percentage change in CoQ10 levels in all treatment groups (correlation coefficient [R] = 0.67, p < 0.0001). The ratios of CoQ10 levels to LDL-C levels were significantly increased in all treatment groups (p < 0.0001). CoQ10 level was independent of cholesterol synthesis or absorption markers. CONCLUSIONS Simvastatin and the combination of simvastatin and ezetimibe significantly decrease plasma CoQ10 levels whereas ezetimibe monotherapy does not. There is a significant correlation between the CoQ10 level decrease and the decrease in total and LDL-C levels in all three treatment groups, suggesting that the CoQ10 decrease may reflect the decrease in the levels of its lipoprotein carriers and might not be statin-specific. The statin-associated CoQ10 reduction is not abrogated through ezetimibe coadministration. Changes of CoQ10 levels are independent of cholesterol synthesis and absorption.
Collapse
|