1
|
Shi Z, Han S. Personalized statin therapy: Targeting metabolic processes to modulate the therapeutic and adverse effects of statins. Heliyon 2025; 11:e41629. [PMID: 39866414 PMCID: PMC11761934 DOI: 10.1016/j.heliyon.2025.e41629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/28/2025] Open
Abstract
Statins are widely used for treating lipid disorders and cardiovascular diseases. However, the therapeutic efficiency and adverse effects of statins vary among different patients, which numerous clinical and epidemiological studies have attributed to genetic polymorphisms in statin-metabolizing enzymes and transport proteins. The metabolic processes of statins are relatively complex, involving spontaneous or enzyme-catalyzed interconversion between more toxic lactone metabolites and active acid forms in the liver and bloodstream, influenced by multiple factors, including the expression levels of many metabolic enzymes and transporters. Addressing the variable statin therapeutic outcomes is a pressing clinical challenge. Transcription factors and epigenetic modifications regulate the metabolic enzymes and transporters involved in statin metabolism and disposition and, therefore, hold promise as 'personalized' targets for achieving optimized statin therapy. In this review, we explore the potential for customizing therapy by targeting the metabolism of statin medications. The biochemical bases of adverse reactions to statin drugs and their correlation with polymorphisms in metabolic enzymes and transporters are summarized. Next, we mainly focus on the regulatory roles of transcription factors and epigenetic modifications in regulating the gene expression of statin biochemical machinery. The recommendations for future therapies are finally proposed by targeting the central regulatory factors of statin metabolism.
Collapse
Affiliation(s)
- Zhuangqi Shi
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830046, China
| | - Shuxin Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830046, China
| |
Collapse
|
2
|
Kumar P, Kumari I, Prasad R, Ray S, Banerjee A, Prakash A. Elucidating the binding specificity of interactive compounds targeting ATP-binding cassette subfamily G member 2 (ABCG2). Mol Divers 2025:10.1007/s11030-024-11078-2. [PMID: 39786520 DOI: 10.1007/s11030-024-11078-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025]
Abstract
The ATP-binding cassette transporter superfamily plays a pivotal role in cellular detoxification and drug efflux. ATP-binding cassette subfamily G member 2 (ABCG2) referred to as the Breast cancer resistance protein has emerged as a key member involved in multidrug resistance displayed by cancer cells. Understanding the molecular basis of substrate and inhibitor recognition, and binding within the transmembrane domain of ABCG2 is crucial for the development of effective therapeutic strategies. Herein, utilizing state-of-the-art molecular docking algorithms and molecular dynamic (MD) simulations, molecular binding of substrates and inhibitors with ABCG2 are defined, distinctly. We performed extensive virtual screening of Drugbank to identify the potential candidates, and MD simulations of docked complexes were carried out in POPC lipid bilayer. Further, the binding affinities of compounds were estimated by free binding energy employing MM-GBSA. To gain deeper insight into the binding affinities and molecular characteristics contributing to inhibitory potential of certain substrates, we included some well-known inhibitors, like Imatinib, Tariquidar, and Ko 143, in our analysis. Docking results show three compounds, Docetaxel > Tariquidar > Tezacaftor having the highest binding affinities (≤ 12.00 kcal/mol) for ABCG2. Remarkably, MM-GBSA results suggest the most stable binding of Tariquidar with ABCG2 as compared to the other inhibitors. Furthermore, our results suggested that Docetaxel, Ozanimod, Pitavastatin, and Tezacaftor have the strongest affinity for the drug-binding site(s) of ABCG2. These results provide valuable insights into the key residues that may govern substrate/inhibitor recognition, shedding light on the molecular determinants influencing substrate specificity, transport kinetics, and ABCG2-mediated drug efflux.
Collapse
Affiliation(s)
- Pawan Kumar
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Indu Kumari
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India
- Data Science, Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurugram, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, 845401, India
| | - Atanu Banerjee
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India.
| | - Amresh Prakash
- Data Science, Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurugram, India.
| |
Collapse
|
3
|
Kim HL, Kwon S, Joh HS, Lim WH, Seo JB, Kim SH, Zo JH, Kim MA. Impact of three-month treatment with pitavastatin on arterial stiffness in patients with hypercholesterolemia: a prospective observational study. Hellenic J Cardiol 2024:S1109-9666(24)00214-8. [PMID: 39427761 DOI: 10.1016/j.hjc.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVE Although several studies have highlighted the benefits of statins in improving arterial stiffness, there is limited research on whether pitavastatin, a more recently developed statin, has similar effects. This study aimed to investigate the impact of pitavastatin on arterial stiffness in patients with hypercholesterolemia. METHODS This prospective study enrolled 115 patients with hypercholesterolemia (mean age, 59 years; 22% female) who had not previously been treated with statins. Participants underwent lifestyle interventions followed by administration of pitavastatin (2 or 4 mg daily) if target low-density lipoprotein (LDL) cholesterol levels were not achieved. The primary outcome measured was the change in brachial-ankle pulse wave velocity (baPWV) from baseline after three months of treatment. RESULTS Pitavastatin treatment significantly reduced LDL cholesterol by 41.1% (from 158 ± 28 to 93.0 ± 29.6 mg/dL; P < 0.001). Additionally, systolic blood pressure (SBP) decreased significantly from 130 ± 14 to 126 ± 14 mmHg (P < 0.001), representing a 2.9% reduction, and baPWV decreased significantly from 1522 ± 325 to 1407 ± 289 cm/s (P < 0.001), a 7.6% reduction. There was a significant correlation between changes in SBP and changes in baPWV (r = 0.565; P < 0.001). Even after adjusting for changes in SBP, the reduction in baPWV induced by pitavastatin remained statistically significant (P < 0.001). CONCLUSION A three-month treatment with pitavastatin effectively reduced arterial stiffness in patients with hypercholesterolemia, alongside significant improvements in lipid profiles and blood pressure reductions. These findings support the use of pitavastatin for managing key cardiovascular risk factors.
Collapse
Affiliation(s)
- Hack-Lyoung Kim
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea.
| | - Soonil Kwon
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun Sung Joh
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Woo-Hyun Lim
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Jae-Bin Seo
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang-Hyun Kim
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Joo-Hee Zo
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Myung-A Kim
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
4
|
Oliai SF, Shippy DC, Ulland TK. Mitigation of CXCL10 secretion by metabolic disorder drugs in microglial-mediated neuroinflammation. J Neuroimmunol 2024; 391:578364. [PMID: 38718558 PMCID: PMC11165694 DOI: 10.1016/j.jneuroim.2024.578364] [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: 02/13/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Metabolic disorders are associated with several neurodegenerative diseases. We previously identified C-X-C motif chemokine ligand 10 (CXCL10), also known as interferon gamma-induced protein 10 (IP-10), as a major contributor to the type I interferon response in microglial-mediated neuroinflammation. Therefore, we hypothesized FDA-approved metabolic disorder drugs that attenuate CXCL10 secretion may be repurposed as a treatment for neurodegenerative diseases. Screening, dose curves, and cytotoxicity assays in LPS-stimulated microglia yielded treprostinil (hypertension), pitavastatin (hyperlipidemia), and eplerenone (hypertension) as candidates that significantly reduced CXCL10 secretion (in addition to other pro-inflammatory mediators) without impacting cell viability. Altogether, these data suggest metabolic disorder drugs that attenuate CXCL10 as potential treatments for neurodegenerative disease through mitigating microglial-mediated neuroinflammation.
Collapse
Affiliation(s)
- Sophia F Oliai
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Daniel C Shippy
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
5
|
Chen L, Xu YX, Wang YS, Zhou JL. Lipid metabolism, amino acid metabolism, and prostate cancer: a crucial metabolic journey. Asian J Androl 2024; 26:123-134. [PMID: 38157428 PMCID: PMC10919422 DOI: 10.4103/aja202363] [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: 06/16/2023] [Accepted: 10/08/2023] [Indexed: 01/03/2024] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies in males worldwide, and its development and progression involve the regulation of multiple metabolic pathways. Alterations in lipid metabolism affect the proliferation and metastatic capabilities of PCa cells. Cancer cells increase lipid synthesis and regulate fatty acid oxidation to meet their growth and energy demands. Similarly, changes occur in amino acid metabolism in PCa. Cancer cells exhibit an increased demand for specific amino acids, and they regulate amino acid transport and metabolic pathways to fulfill their proliferation and survival requirements. These changes are closely associated with disease progression and treatment response in PCa cells. Therefore, a comprehensive investigation of the metabolic characteristics of PCa is expected to offer novel insights and approaches for the early diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
6
|
Arsh H, Ali A, Khenhrani RR, Simran F, Dino U, Tamang S, Manoj F, Bai S, Bai M, Panjwani GR, Kumar D, Rani D, Partab F, Malik J. Efficacy and Safety of Pitavastatin in Patients with Impaired Glucose Tolerance: An Updated Review. Curr Probl Cardiol 2023; 48:101981. [PMID: 37473935 DOI: 10.1016/j.cpcardiol.2023.101981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/16/2023] [Indexed: 07/22/2023]
Abstract
This review provides an updated overview of the efficacy and safety of pitavastatin in patients with impaired glucose tolerance (IGT). IGT is a prediabetic state characterized by elevated blood glucose levels that do not meet the criteria for diabetes. The review explores the potential benefits of pitavastatin in reducing cardiovascular risk and improving lipid profiles in individuals with IGT. It also examines the glycemic effects of pitavastatin, including its impact on fasting blood glucose levels, insulin sensitivity, and beta-cell function. The review highlights the need for individualized treatment approaches, taking into account the patient's overall cardiovascular risk profile and glycemic control needs. While pitavastatin has shown modest improvements in glycemic control, it is not a substitute for lifestyle modifications or standard antidiabetic medications. Future directions for research include long-term follow-up studies, mechanistic investigations, and comparative analyses to further understand the glycemic effects of pitavastatin in IGT. Overall, this narrative review provides valuable insights for healthcare professionals involved in the management of individuals with IGT, emphasizing the importance of a comprehensive approach to reduce cardiovascular risk and optimize glycemic control.
Collapse
Affiliation(s)
- Hina Arsh
- Department of Medicine, THQ Hospital, Pasrur, Pakistan
| | - Asif Ali
- Department of Medicine, Chandka Medical College, Larkana, Pakistan
| | - Raja Ram Khenhrani
- Department of Medicine, Shaheed Mohtarma Benazir Bhutto Medical College, Lyari, Pakistan
| | - Fnu Simran
- Department of Medicine, Sheikh Khalifa Bin Zayed Al Nayhan Medical and Dental College, Lahore, Pakistan
| | - Umbish Dino
- Department of Medicine, Sheikh Khalifa Bin Zayed Al Nayhan Medical and Dental College, Lahore, Pakistan
| | - Sweta Tamang
- Nepal Medical College and Teaching Hospital, Nepal
| | - Fnu Manoj
- Department of Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Shuaita Bai
- Department of Medicine, People's University of Medical and Health Sciences, Nawabshah, Pakistan
| | - Monika Bai
- Department of Medicine, People's University of Medical and Health Sciences, Nawabshah, Pakistan
| | | | - Deepak Kumar
- Department of Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Deepa Rani
- Department of Medicine, Shaheed Mohtarma Benazir Bhutto Medical College, Lyari, Pakistan
| | - Fnu Partab
- Department of Medicine, Chandka Medical College, Larkana, Pakistan
| | - Jahanzeb Malik
- Department of Cardiovascular Medicine, Cardiovascular Analytics Group, Islamabad, Pakistan.
| |
Collapse
|
7
|
Tarim BA, Fici F, Tengiz I, Avunduk S, Ozcan Y, Faikoglu G, Ari E, Robles NR, Grassi G. Do Statins Counteract the Effect of Antidiabetic Drugs? Results of the SCEAD Study. Yonsei Med J 2023; 64:175-180. [PMID: 36825343 PMCID: PMC9971433 DOI: 10.3349/ymj.2022.0287] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/25/2023] Open
Abstract
PURPOSE Diabetes and dyslipidemia are leading causes of mortality and morbidity. According to international guidelines, statins are the cornerstone of treatment in patients with diabetes and/or dyslipidemia. However, statins and antidiabetic agents have opposite pharmacological effects, because statins, particularly atorvastatin and rosuvastatin, impair glucose homeostasis, increasing the risk of new-onset diabetes, whereas antidiabetic drugs improve glycemic homeostasis. The aim of this study was to investigate the effect of atorvastatin, rosuvastatin, and pitavastatin on glucose homeostasis in patients with type 2 diabetes mellitus (T2DM) and dyslipidemia during stable treatment with hypoglycemic drugs. MATERIALS AND METHODS The study was conducted as a pilot, prospective, randomized, open label, parallel group with blinded-endpoints (PROBE) study. Of 180 recruited patients with T2DM and dyslipidemia, 131 were randomized to atorvastatin (n=44), rosuvastatin (n=45), and pitavastatin (n=42) and treated for 6 months. RESULTS Fasting plasma glucose (FPG) marginally decreased in patients assigned to atorvastatin (-3.5 mg/dL, p=0.42) and rosuvastatin (-6.5 mg/dL, p=0.17), while it decreased much more in patients treated with pitavastatin (-19.0 mg/dL, p<0.001). Mean glycated hemoglobin A1c (HbA1c ) values remained unchanged during treatment with atorvastatin (-0.10%, p=0.53) and rosuvastatin (0.20%, p=0.40), but were significantly reduced with pitavastatin (-0.75%, p=0.01). Atorvastatin, rosuvastatin, and pitavastatin significantly lowered (p<0.001) plasma levels of total cholesterol, low-density lipoprotein-cholesterol, and triglycerides, while high-density lipoprotein-cholesterol (HDL-C) levels increased significantly (p=0.04) only in the pitavastatin group. CONCLUSION The results of the present study suggest that pitavastatin affects FPG and HbA1c less than atorvastatin and rosuvastatin in patients with T2DM and concomitant dyslipidemia. Lipid-lowering efficacies were not significantly different among the three statins, with the exception of HDL-C, which increased significantly with pitavastatin. Although the pharmacological mechanism of pitavastatin on glucose homeostasis in patients with T2DM during stable antidiabetic therapy is not known, it can be assumed that pitavastatin has less drug interaction with hypoglycemic agents or that it increases plasma levels of adiponectin.
Collapse
Affiliation(s)
- Bahar Arican Tarim
- Department of Internal Medicine, Kartal Research and Education Hospital, Istanbul, Turkey
| | - Francesco Fici
- Department of Cardiovascular Risk, Salamanca University, Salamanca, Spain
- Department of Pharmacology, Milano-Bicocca University, Milan, Italy
| | - Istemihan Tengiz
- Department of Cardiology, Medical Park Hospital, Izmir University, Izmir, Turkey
| | - Saadet Avunduk
- Department of Cardiology, Denizli State Hospital, Denizli, Turkey
| | - Yurdaer Ozcan
- Department of Internal Medicine, Bayrampasa State Hospital, Bayrampasa, Turkey
| | - Gokhan Faikoglu
- Department of Medical Pharmacology, University Cerrahpasa, Istanbul, Turkey
| | - Elif Ari
- Department of Nephrology, Bahcesehir University, Istanbul, Turkey
| | | | - Guido Grassi
- Department of Pharmacology, Milano-Bicocca University, Milan, Italy
- Medical Clinics, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
8
|
Prado Y, Aravena D, Llancalahuen FM, Aravena C, Eltit F, Echeverría C, Gatica S, Riedel CA, Simon F. Statins and Hemostasis: Therapeutic Potential Based on Clinical Evidence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:25-47. [PMID: 37093420 DOI: 10.1007/978-3-031-26163-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Hemostasis preserves blood fluidity and prevents its loss after vessel injury. The maintenance of blood fluidity requires a delicate balance between pro-coagulant and fibrinolytic status. Endothelial cells (ECs) in the inner face of blood vessels maintain hemostasis through balancing anti-thrombotic and pro-fibrinolytic activities. Dyslipidemias are linked to hemostatic alterations. Thus, it is necessary a better understanding of the underlying mechanisms linking hemostasis with dyslipidemia. Statins are drugs that decrease cholesterol levels in the blood and are the gold standard for treating hyperlipidemias. Statins can be classified into natural and synthetic molecules, approved for the treatment of hypercholesterolemia. The classical mechanism of action of statins is by competitive inhibition of a key enzyme in the synthesis pathway of cholesterol, the HMG-CoA reductase. Statins are frequently administrated by oral ingestion and its interaction with other drugs and food supplements is associated with altered bioavailability. In this review we deeply discuss the actions of statins beyond the control of dyslipidemias, focusing on the actions in thrombotic modulation, vascular and cardiovascular-related diseases, metabolic diseases including metabolic syndrome, diabetes, hyperlipidemia, and hypertension, and chronic diseases such as cancer, chronic obstructive pulmonary disease, and chronic kidney disease. Furthermore, we were prompted to delved deeper in the molecular mechanisms by means statins regulate coagulation acting on liver, platelets, and endothelium. Clinical evidence show that statins are effective regulators of dyslipidemia with a high impact in hemostasis regulation and its deleterious consequences. However, studies are required to elucidate its underlying molecular mechanism and improving their therapeutical actions.
Collapse
Affiliation(s)
- Yolanda Prado
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Aravena
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe M Llancalahuen
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Cristobal Aravena
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Eltit
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- Vancouver Prostate Centre, Vancouver, Canada
| | - Cesar Echeverría
- Laboratory of Molecular Biology, Nanomedicine and Genomics, Faculty of Medicine, University of Atacama, Copiapo, Chile
| | - Sebastian Gatica
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Claudia A Riedel
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Simon
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile.
| |
Collapse
|
9
|
Cai R, Chang C, Zhong X, Su Q. Lowering of Blood Lipid Levels with a Combination of Pitavastatin and Ezetimibe in Patients with Coronary Heart Disease: A Meta-Analysis. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2023; 7. [DOI: 10.15212/cvia.2023.0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025] Open
Abstract
Objectives: According to the findings of randomized controlled trials, blood lipid levels in patients with coronary heart disease (CHD) can be significantly decreased through a combination of pitavastatin and ezetimibe; however, the effects and clinical applications of this treatment remain controversial. This meta-analysis was aimed at objectively assessing the efficacy and safety of pitavastatin and ezetimibe in lowering blood lipid levels.
Design: Relevant studies were retrieved from electronic databases, including PubMed, Cochrane Library, Embase, China National Knowledge Infrastructure, VIP, and WanFang Data, from database inception to June 8, 2022. The levels of low-density lipoprotein cholesterol, total cholesterol, triglycerides, and high-density lipoprotein cholesterol in patients’ serum after treatment were the primary endpoint.
Results: Nine randomized controlled trials (2586 patients) met the inclusion criteria. The meta-analysis indicated that pitavastatin plus ezetimibe resulted in significantly lower levels of LDL-C [standardized mean difference (SMD)=−0.86, 95% confidence interval (CI) (−1.15 to −0.58), P<0.01], TC [SMD=−0.84, 95% CI (−1.10 to −0.59), P<0.01], and TG [SMD=−0.59, 95% CI (−0.89 to −0.28), P<0.01] than pitavastatin alone.
Conclusions: Pitavastatin plus ezetimibe significantly decreased serum LDL-C, TC, and TG levels in patients with CHD.
Collapse
|
10
|
Kim KI, Kim SM, Lee YY, Lee Y, Kim CD, Yoon TJ. Pitavastatin Induces Apoptosis of Cutaneous Squamous Cell Carcinoma Cells through Geranylgeranyl Pyrophosphate-Dependent c-Jun N-Terminal Kinase Activation. Ann Dermatol 2023; 35:116-123. [PMID: 37041705 PMCID: PMC10112368 DOI: 10.5021/ad.22.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/01/2022] [Accepted: 11/16/2022] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND Pitavastatin is a cholesterol-lowering drug and is widely used clinically. In addition to this effect, pitavastatin has shown the potential to induce apoptosis in cutaneous squamous cell carcinoma (SCC) cells. OBJECTIVE The purpose of this study is to investigate the effects and possible action mechanisms of pitavastatin. METHODS SCC cells (SCC12 and SCC13 cells) were treated with pitavastatin, and induction of apoptosis was confirmed by Western blot. To examine whether pitavastatin-induced apoptosis is related to a decrease in the amount of intermediate mediators in the cholesterol synthesis pathway, the changes in pitavastatin-induced apoptosis after supplementation with mevalonate, squalene, geranylgeranyl pyrophosphate (GGPP) and dolichol were investigated. RESULTS Pitavastatin dose-dependently induced apoptosis of cutaneous SCC cells, but the viability of normal keratinocytes was not affected by pitavastatin at the same concentrations. In supplementation experiments, pitavastatin-induced apoptosis was inhibited by the addition of mevalonate or downstream metabolite GGPP. As a result of examining the effect on intracellular signaling, pitavastatin decreased Yes1 associated transcriptional regulator and Ras homolog family member A and increased Rac family small GTPase 1 and c-Jun N-terminal kinase (JNK) activity. All these effects of pitavastatin on signaling molecules were restored when supplemented with either mevalonate or GGPP. Furthermore, pitavastatin-induced apoptosis of cutaneous SCC cells was inhibited by a JNK inhibitor. CONCLUSION These results suggest that pitavastatin induces apoptosis of cutaneous SCC cells through GGPP-dependent JNK activation.
Collapse
Affiliation(s)
- Kyung-Il Kim
- Department of Dermatology, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Dermatology, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Seung-Mee Kim
- Department of Dermatology, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
| | - Young-Yoon Lee
- Department of Dermatology, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
| | - Young Lee
- Department of Dermatology, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Chang-Deok Kim
- Department of Dermatology, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Tae-Jin Yoon
- Department of Dermatology, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongsang Institute of Health Sciences, Jinju, Korea
| |
Collapse
|
11
|
Xuan Y, Zhang W, Wang Y, Wang B, Xia F, Zhang K, Li Q, Wang N, Lu Y. Association Between Uric Acid to HDL Cholesterol Ratio and Diabetic Complications in Men and Postmenopausal Women. Diabetes Metab Syndr Obes 2023; 16:167-177. [PMID: 36760595 PMCID: PMC9869791 DOI: 10.2147/dmso.s387726] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/15/2022] [Indexed: 01/20/2023] Open
Abstract
AIM Previous studies have implicated the uric acid to high-density lipoprotein cholesterol (HDL-C) ratio (UHR) was associated with type 2 diabetes. However, the association between UHR and diabetes-related vascular damages is still unclear. METHODS The total of 4551 patients with type 2 diabetes from the cross-sectional Environmental Pollutant Exposure and Metabolic Diseases in Shanghai study (METAL study) were enrolled. UHR was calculated as uric acid to HDL-C ratio. Cardiovascular disease (CVD) was defined as previously diagnosed with stroke, coronary heart disease, or peripheral arterial disease. Chronic kidney disease (CKD) was defined as estimated glomerular filtration rate ≤60 mL/min/1.73 m2 and/or urinary albumin to creatinine ratio ≥30 mg/g. Fundus image was examined by trained individuals and degree of diabetic retinopathy (DR) was evaluated. RESULTS UHR was positively correlated with CVD (OR = 1.28, 95% CI: 1.02-1.61) and CKD (OR = 1.78, 95% CI: 1.39-2.27) after adjusting for all confounders. No association was found between UHR and DR. In stratified analyses, UHR was predominantly correlated with CVD in diabetic patients with age older than 65 (OR = 1.41, 95% CI: 1.08-1.85), female (OR = 1.43, 95% CI: 1.06-1.94) and BMI≥24kg/m2 (OR = 1.57, 95% CI: 1.17-2.11). A 1-SD increment of UHR was also positively associated with CVD (OR 1.26, 95% CI 1.03, 1.15) and CKD (OR 1.28, 95% CI 1.20,1.39). UHR was positively associated with CKD in all subgroups analysis. No significant interaction effect was observed between UHR and all subgroup variables in CVD and CKD risk. CONCLUSION Our study reported a positive association between the UHR and diabetic-related vascular complications in men and postmenopausal women. The relationship between the UHR and DR seems to be uncertain and requires further investigation. And no significant interaction effect was observed between the UHR and all subgroup variables in CVD and CKD risk.
Collapse
Affiliation(s)
- Yan Xuan
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Department of Endocrinology, Luwan Branch, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200020, People’s Republic of China
| | - Wen Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuying Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Bin Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Qing Li
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Yingli Lu, Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, People’s Republic of China, Tel +86-13636352507, Fax +86-21-63136856, Email
| |
Collapse
|
12
|
Liang K, Dai JY. Progress of potential drugs targeted in lipid metabolism research. Front Pharmacol 2022; 13:1067652. [PMID: 36588702 PMCID: PMC9800514 DOI: 10.3389/fphar.2022.1067652] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Lipids are a class of complex hydrophobic molecules derived from fatty acids that not only form the structural basis of biological membranes but also regulate metabolism and maintain energy balance. The role of lipids in obesity and other metabolic diseases has recently received much attention, making lipid metabolism one of the attractive research areas. Several metabolic diseases are linked to lipid metabolism, including diabetes, obesity, and atherosclerosis. Additionally, lipid metabolism contributes to the rapid growth of cancer cells as abnormal lipid synthesis or uptake enhances the growth of cancer cells. This review introduces the potential drug targets in lipid metabolism and summarizes the important potential drug targets with recent research progress on the corresponding small molecule inhibitor drugs. The significance of this review is to provide a reference for the clinical treatment of metabolic diseases related to lipid metabolism and the treatment of tumors, hoping to deepen the understanding of lipid metabolism and health.
Collapse
Affiliation(s)
- Kai Liang
- School of Life Science, Peking University, Beijing, China,*Correspondence: Kai Liang, ; Jian-Ye Dai,
| | - Jian-Ye Dai
- School of Pharmacy, Lanzhou University, Lanzhou, China,Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, China,*Correspondence: Kai Liang, ; Jian-Ye Dai,
| |
Collapse
|
13
|
Wu Y, Meng W, Guan M, Zhao X, Zhang C, Fang Q, Zhang Y, Sun Z, Cai M, Huang D, Yang X, Yu Y, Cui Y, He S, Chai R. Pitavastatin protects against neomycin-induced ototoxicity through inhibition of endoplasmic reticulum stress. Front Mol Neurosci 2022; 15:963083. [PMID: 35992197 PMCID: PMC9381809 DOI: 10.3389/fnmol.2022.963083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Irreversible injury to inner ear hair cells induced by aminoglycoside antibiotics contributes to the formation of sensorineural hearing loss. Pitavastatin (PTV), a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, has been reported to exert neuroprotective effects. However, its role in aminoglycoside-induced hearing loss remains unknown. The objectives of this study were to investigate the beneficial effects, as well as the mechanism of action of PTV against neomycin-induced ototoxicity. We found that PTV remarkably reduced hair cell loss in mouse cochlear explants and promoted auditory HEI-OC1 cells survival after neomycin stimulation. We also observed that the auditory brainstem response threshold that was increased by neomycin was significantly reduced by pretreatment with PTV in mice. Furthermore, neomycin-induced endoplasmic reticulum stress in hair cells was attenuated by PTV treatment through inhibition of PERK/eIF2α/ATF4 signaling. Additionally, we found that PTV suppressed the RhoA/ROCK/JNK signal pathway, which was activated by neomycin stimulation in HEI-OC1 cells. Collectively, our results showed that PTV might serve as a promising therapeutic agent against aminoglycoside-induced ototoxicity.
Collapse
Affiliation(s)
- Yunhao Wu
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Wei Meng
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Ming Guan
- Department of Otolaryngology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolong Zhao
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chen Zhang
- Beijing Key Laboratory of Neural Regeneration and Repair, Department of Neurobiology, Advanced Innovation Center for Human Brain Protection, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qiaojun Fang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yuhua Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Zihui Sun
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Mingjing Cai
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Dongdong Huang
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
| | - Xuechun Yang
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yafeng Yu
- Department of Otolaryngology, First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Yafeng Yu,
| | - Yong Cui
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, South Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Yong Cui,
| | - Shuangba He
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, Nanjing Tongren Hospital, Southeast University, Nanjing, China
- Shuangba He,
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Department of Otolaryngology Head and Neck Surgery, School of Life Sciences and Technology, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Renjie Chai,
| |
Collapse
|
14
|
RSSDI consensus recommendations for dyslipidemia management in diabetes mellitus. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
15
|
Fici F, Faikoglu G, Tarim BA, Robles NR, Tsioufis K, Grassi G, Gungor B. Pitavastatin: Coronary Atherosclerotic Plaques Changes and Cardiovascular Prevention. High Blood Press Cardiovasc Prev 2022; 29:137-144. [PMID: 35064911 DOI: 10.1007/s40292-021-00496-0] [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: 10/03/2021] [Accepted: 11/30/2021] [Indexed: 10/19/2022] Open
Abstract
Stains remain the first therapeutic approach in patients with dyslipidemia to control plasma lipids levels and cardiovascular risk. Multiple clinical trials have demonstrated the benefits of statins in reducing major cardiovascular adverse events in primary and secondary prevention. Moreover, in patients with coronary artery disease, statins decrease coronary atherosclerotic plaque volume and composition, inducing atheroma stabilization. Pitavastatin, is a new-generation lipophilic statin, indicated for the treatment of dyslipidemia and prevention of cardiovascular diseases. The purpose of this review, the first at our knowledge on this topic, is to summarize and examine the current knowledge about the effectiveness of pitavastatin in patients with coronary artery disease. The available data suggest that pitavastatin significantly, lowers the rate of adverse cardiovascular events, in patients at a high risk of atherosclerotic disease, with stable angina pectoris or with acute coronary syndrome. Moreover intravascular ultrasound have shown that pitavastatin induces favorable changes in plaque morphology, increasing the fibrous cap thickness, and decreasing both plaque and lipid volume indexes. Globally the efficacy of pitavastatin is greater or similar to other statins.
Collapse
Affiliation(s)
- Francesco Fici
- Department of Cardiovascular Risk, Salamanca University, Salamanca, Spain
- Milano-Bicocca, University, Milan, Italy
| | - Gokhan Faikoglu
- Department of Medical Pharmacology, University Cerrahpasa, Istanbul, Turkey
| | | | - Nicolas Roberto Robles
- Department of Cardiovascular Risk, Salamanca University, Salamanca, Spain.
- Milano-Bicocca, University, Milan, Italy.
- University Hospital of Badajoz, Badajoz, Spain.
| | - Kostas Tsioufis
- First Cardiology Clinic, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Guido Grassi
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
| | - Barış Gungor
- Department of Cardiology, University of Sciences and Research Hospital, Istanbul, Turkey
| |
Collapse
|
16
|
Yuan M, Han S, Jia Y, Feng J, Liu D, Su Z, Liu X. Statins Are Associated with Improved Survival of Patients with Gastric Cancer: A Systematic Review and Meta-Analysis. Int J Clin Pract 2022; 2022:4938539. [PMID: 35685487 PMCID: PMC9158792 DOI: 10.1155/2022/4938539] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022] Open
Abstract
Statins are associated with gastric cancer (GC) risk. The present study aimed to clarify the efficacy of statins on the overall survival (OS) benefits in patients with GC. Publications were retrieved from PubMed, Embase, and the Cochrane Library as of April 2022. Data from the eligible cohort, case-control studies, and randomized control trials (RCTs) were extracted for the meta-analysis. Hazard ratio (HR) and 95% confidence intervals (CI) were used to assess the association between statins users and OS in GC patients. Subgroup analysis was performed based on the study design (prospective vs. retrospective). A total of 6 studies encompassing 5693 GC patients were included. Statins added to the standard treatment prolonged the patient's OS outcome (HR (95% CI): 0.72 (0.53-0.97), p = 0.032; I 2 = 88.0%, p heterogeneity < 0.001). A prospective study did not find any statistically significant difference in OS between statins users vs. nonstatin users (HR (95% CI): 0.92 (0.68-1.26), p = 0.614; I 2 = 11.7%, p heterogeneity = 0.322), whereas the retrospective studies showed prolonged OS in statins users (HR (95% CI): 0.63 (0.42-0.961), p = 0.032; I 2 = 94.6%, p heterogeneity < 0.001). Statin users had significantly improved OS compared to nonstatin users in GC treatment. This long-term survival benefit was only observed in the pooled analysis of retrospective studies but not in prospective studies.
Collapse
Affiliation(s)
- Mingjie Yuan
- Department of Clinical Laboratory, Binzhou Medical University, Yantai, Shandong, China
| | - Shuyi Han
- Department of Laboratory, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yanfei Jia
- Department of Laboratory, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Jiankai Feng
- Department of Clinical Laboratory, Binzhou Medical University, Yantai, Shandong, China
| | - Duanrui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenguo Su
- Department of Clinical Laboratory, Binzhou Medical University, Yantai, Shandong, China
| | - Xiangdong Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
17
|
Zhou X, Wu L, Chen Y, Xiao H, Huang X, Li Y, Xiao H, Cao X. Forty-eight weeks of statin therapy for type 2 diabetes mellitus patients with lower extremity atherosclerotic disease: Comparison of the effects of pitavastatin and atorvastatin on lower femoral total plaque areas. J Diabetes Investig 2021; 12:1278-1286. [PMID: 33289308 PMCID: PMC8264389 DOI: 10.1111/jdi.13472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 11/12/2020] [Accepted: 11/29/2020] [Indexed: 11/29/2022] Open
Abstract
AIMS/INTRODUCTION Type 2 diabetes mellitus is correlated with systemic atherosclerosis. Statin therapies have been proved to reduce low-density lipoprotein cholesterol (LDL-C) level, protecting type 2 diabetes mellitus patients from cardiovascular events. Recently, more interest has been focused on the regression of lower extremity atherosclerotic disease (LEAD) for the potential prevention of amputation. However, the effects of pitavastatin and atorvastatin on LEAD in type 2 diabetes mellitus patients have not been directly compared. MATERIALS AND METHODS This study compared the effects of pitavastatin and atorvastatin on femoral total plaque areas (FTPA), and lipids and glucose metabolism in type 2 diabetes mellitus patients with elevated LDL-C level and LEAD. Type 2 diabetes mellitus patients with LDL-C level >2.6 mmol/L and LEAD were randomly assigned to receive either pitavastatin 2 mg/day or atorvastatin 10 mg/day for 48 weeks. FTPA were measured at baseline and the end of the study. Levels of glucose and lipids profile were measured periodically. The efficacy was evaluated in 63 patients. RESULTS The percentage change in FTPA measurements was similar between the pitavastatin group and atorvastatin group (-17.79 ± 21.27% vs -14.34 ± 16.33%), as were the changes in LDL-C (-44.0 ± 18.0% vs -40.3 ± 18.2%) and triglyceride (17.6 ± 20.0% vs 16.2 ± 17.0%). However, the level of high-density lipoprotein cholesterol was significantly higher in the pitavastatin group compared with the atorvastatin group after 48 weeks of treatment (12.9 ± 10.3% vs 7.2 ± 11.7%, P < 0.05). There were no significant differences between groups for the measurements of glucose metabolism. CONCLUSION In type 2 diabetes mellitus patients with elevated LDL-C level and LEAD, 48 weeks of treatment with either pitavastatin or atorvastatin was associated with significant regression of FTPA. Pitavastatin treatment resulted in a significantly higher high-density lipoprotein cholesterol level compared with atorvastatin treatment.
Collapse
Affiliation(s)
- Xieda Zhou
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Liting Wu
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yan Chen
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Huangmeng Xiao
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaoyu Huang
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yanbing Li
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Haipeng Xiao
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaopei Cao
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
18
|
Shehab A, Elnour AA, Bhagavathula AS, Kurian JP, Hassan G, AlZaabi S, Gilbert H, Al-Rasadi K. A Multicenter Prospective Hospital-based Cohort Study on the Efficacy and Safety of Pitavastatin. Curr Diabetes Rev 2021; 17:e122820189541. [PMID: 33371852 DOI: 10.2174/1573399817666201228164243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/19/2020] [Accepted: 10/24/2020] [Indexed: 11/22/2022]
Abstract
AIMS We aim to investigate the efficacy and safety of pitavastatin 4 mg in a population of people living in the United Arab Emirates (UAE). BACKGROUND Pitavastatin is a member of the HMG-CoA reductase inhibitors family which was approved for use in adult subjects with primary hyperlipidemia or mixed dyslipidemia. To date, no published studies have assessed the efficacy and safety of pitavastatin in the United Arab Emirates. OBJECTIVE The main objective of the current study was to investigate the efficacy and safety of pitavastatin in subjects with dyslipidemia for the primary prevention of cardiovascular diseases based on total cardiovascular risk. METHODS This was a multicentre (four private hospitals) prospective cohort study to analyze data on the use of pitavastatin for dyslipidemia in adult outpatients in Abu Dhabi and Dubai, United Arab Emirates. We have followed up the clinical profiles of subjects in four hospitals for six-weeks during the period from June 2015 to June 2017. Efficacy was based on the evaluation of the mean (± standard deviation) change in low-density lipoprotein cholesterol between baseline and week six after the initiation of pitavastatin therapy. Safety was reported with respect to the incidence of adverse events occurring with the use of pitavastatin and the development of new-onset diabetes. RESULTS A total of 400 subjects who were receiving pitavastatin 4 mg were included. The mean age of subjects was 50.7 ±10.8 years; of these, 79.0% were males. At the baseline, the mean level of total cholesterol was 185.4 ±41.5 mg/dL, low density lipoprotein was 154.9 ±48.55 mg/dL, high- -density lipoprotein cholesterol was 40.5 ±11.23 mg/dL and fasting blood glucose was 115.0 (±16.63) mg/dl. At the end of six weeks, low density lipoprotein levels significantly decreased to 112.09 ±41.90 mg/dl (standard mean difference (SMD) (-42.8%), 95% CI: -42.88 [-49.17 to -36.58] mg/dl, P <0.001), while high density lipoprotein levels improved (SMD, 95% CI: 1.77% [0.25 to 3.28] mg/dl, P <0.022). There were 55 subjects (13.7%) who reported various adverse events such as myalgia (7.5%), sleep disorders (2.5%), and myopathy (2.2%). Furthermore, 4 (1.0%) have had developed new-onset diabetes post-six-weeks of initiation of pitavastatin therapy. CONCLUSION Pitavastatin 4 mg showed robust efficacy in reducing LDL-C levels and improving HDL-C levels in subjects with dyslipidemia. The use of pitavastatin was associated with a low discontinuation rate, fewer adverse events, and very limited cases of new-onset diabetes.
Collapse
Affiliation(s)
- Abdullah Shehab
- Department of Internal Medicine, College of Medicine, United Arab Emirates University (UAEU), Al Ain, United Arab Emirates
| | - Asim Ahmed Elnour
- Clinical Pharmacy Program, Pharmacy College, Al Ain University, Abu Dhabi Campus,, United Arab Emirates
| | - Akshaya Srikanth Bhagavathula
- Department of Internal Medicine, College of Medicine, United Arab Emirates University (UAEU), Al Ain, United Arab Emirates
| | | | - Gazi Hassan
- Consultant Cardiologist, Gulf Medical Diagnostic Center, Abu Dhabi, United Arab Emirates
| | - Shareen AlZaabi
- Ambulatory Health Services (AHS), Abu Dhabi Heath Services (SEHA), Abu Dhabi, United Arab Emirates
| | - Huburt Gilbert
- Department of Endocrinology and Diabetology, American Hospital, UAE, United Arab Emirates
| | - Khalid Al-Rasadi
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat,, Oman
| |
Collapse
|
19
|
Evaluation of the Pharmacokinetic Drug-Drug Interaction between Micronized Fenofibrate and Pitavastatin in Healthy Volunteers. Pharmaceutics 2020; 12:pharmaceutics12090869. [PMID: 32932576 PMCID: PMC7557955 DOI: 10.3390/pharmaceutics12090869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 11/22/2022] Open
Abstract
Dyslipidemia is a major risk factor for development of atherosclerosis and cardiovascular disease (CVD). Effective lipid-lowering therapies has led to CVD risk reduction. This study evaluated the possible pharmacokinetic interactions between fenofibrate, a peroxisome proliferators-activated receptors α agonist, and pitavastatin, a 3-hydoxy-3-methylglutaryl-coenzyme A reductase inhibitor, in healthy Korean subjects. The study design was an open-label, randomized, multiple-dose, three-period, and six-sequence crossover study with a 10-day washout in 24 healthy volunteers. It had three treatments: 160 mg of micronized fenofibrate once daily for 5 days; 2 mg of pitavastatin once daily for 5 days; and 160 mg of micronized fenofibrate with 2 mg of pitavastatin for 5 days. Serial blood samples were collected at scheduled intervals for up to 48 h after the last dose in each period to determine the steady-state pharmacokinetics of both drugs. Plasma concentrations of fenofibric acid and pitavastatin were measured using a validated high-performance liquid chromatography with the tandem mass spectrometry method. A total of 24 subjects completed the study. Pitavastatin, when co-administered with micronized fenofibrate, had no effect on the Cmax,ss and AUCτ,ss of fenofibric acid. The Cmax,ss and AUCτ,ss of pitavastatin were increased by 36% and 12%, respectively, when co-administered with fenofibrate. Combined treatment with pitavastatin and micronized fenofibrate was generally well tolerated without serious adverse events. Our results demonstrated no clinically significant pharmacokinetic interactions between micronized fenofibrate and pitavastatin when 160 mg of micronized fenofibrate and 2 mg of pitavastatin are co-administered. The treatments were well tolerated during the study, with no serious adverse events.
Collapse
|
20
|
Tokgözoglu L, Zamorano JL. Current perspectives on the use of statins in the treatment of dyslipidaemic patients: focus on pitavastatin. Drugs Context 2020; 9:dic-2020-4-4. [PMID: 32587627 PMCID: PMC7295107 DOI: 10.7573/dic.2020-4-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 01/14/2023] Open
Abstract
A meeting entitled 'Current Perspective on the Use of Statins in the Treatment of Dyslipidemic Patients' was held in Stresa, Italy, on 27-28th June 2019. The presentations covered the 2019 European Society of Cardiology (ESC)/European Atherosclerosis Society (EAS) guidelines on dyslipidaemia, with discussion about the importance of controlling low-density lipoprotein cholesterol (LDL-C) and the pharmacological opportunities to reach the novel lipid goals. The roles of statins to manage dyslipidaemia in patients with different cardiovascular risks were also discussed. In particular, the efficacy and safety of pitavastatin for the treatment of dyslipidaemia were reviewed, highlighting its further advantages beyond LDL-C reduction. Therefore, the impact of statins on the glycaemic profile was discussed in view of the null/lower effect of pitavastatin as compared with other statins, as well as the interaction profile with other drugs commonly used. This meeting report summarizes the main messages of the discussion with a special focus on pitavastatin, whose main features in different settings are described.
Collapse
|
21
|
Statin treatment and increased diabetes risk. Possible mechanisms. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2019; 31:228-232. [PMID: 30737072 DOI: 10.1016/j.arteri.2018.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/26/2018] [Accepted: 12/16/2018] [Indexed: 01/30/2023]
Abstract
Statins have been associated with an increased risk of new-onset diabetes mellitus (NODM), as confirmed in previous observational studies and meta-analyses. Controversy exists as to whether this risk varies depending on statin type or dose. However, there appears to be unanimity regarding the different associated factors that raise this risk. Furthermore, diverse pathophysiologic mechanisms have been described that could explain the increased risk of diabetes in patients with statin treatment. These fundamentally cause a rise in insulin resistance together with a decrease in insulin secretion. The present review aimed to describe the relationship between statin treatment and the presence of diabetes and provide an update of previous published evidence and the possible mechanisms involved.
Collapse
|
22
|
Torres-Adorno AM, Vitrac H, Qi Y, Tan L, Levental KR, Fan YY, Yang P, Chapkin RS, Eckhardt BL, Ueno NT. Eicosapentaenoic acid in combination with EPHA2 inhibition shows efficacy in preclinical models of triple-negative breast cancer by disrupting cellular cholesterol efflux. Oncogene 2018; 38:2135-2150. [PMID: 30459358 PMCID: PMC6430703 DOI: 10.1038/s41388-018-0569-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/26/2018] [Accepted: 10/17/2018] [Indexed: 01/06/2023]
Abstract
Triple-negative breast cancer (TNBC), the most aggressive breast cancer subtype, currently lacks effective targeted therapy options. Eicosapentaenoic acid (EPA), an omega-3 fatty acid and constituent of fish oil, is a common supplement with anti-inflammatory properties. Although it is not a mainstream treatment, several preclinical studies have demonstrated that EPA exerts anti-tumor activity in breast cancer. However, against solid tumors, EPA as a monotherapy is clinically ineffective; thus, we sought to develop a novel targeted drug combination to bolster its therapeutic action against TNBC. Using a high-throughput functional siRNA screen, we identified Ephrin type-A receptor 2 (EPHA2), an oncogenic cell-surface receptor tyrosine kinase, as a therapeutic target that sensitizes TNBC cells to EPA. EPHA2 expression was uniquely elevated in TNBC cell lines and patient tumors. In independent functional expression studies in TNBC models, EPHA2 gene-silencing combined with EPA significantly reduced cell growth and enhanced apoptosis compared with monotherapies, both in vitro and in vivo. EPHA2 specific inhibitors similarly enhanced the therapeutic action of EPA. Finally, we identified that therapy-mediated apoptosis was attributed to a lethal increase in cancer cell membrane polarity due to ABCA1 inhibition and subsequent dysregulation of cholesterol homeostasis. This study provides new molecular and pre-clinical evidence to support a clinical evaluation of EPA combined with EPHA2 inhibition in patients with TNBC.
Collapse
Affiliation(s)
- Angie M Torres-Adorno
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Section of Translational Breast Cancer Research and Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heidi Vitrac
- Department of Biochemistry and Molecular Biology, UTHealth McGovern Medical School, Houston, TX, USA
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kandice R Levental
- Department of Integrative Biology and Pharmacology, UTHealth McGovern Medical School, Houston, TX, USA
| | - Yang-Yi Fan
- Program in Integrative Nutrition & Complex Diseases, Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Peiying Yang
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition & Complex Diseases, Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Bedrich L Eckhardt
- Section of Translational Breast Cancer Research and Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research and Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Pedro-Botet J, Ascaso JF, Barrios V, De la Sierra A, Escalada J, Millán J, Mostaza JM, Pérez-Martínez P, Pintó X, Salas-Salvadó J, Valdivielso P. COSMIC project: consensus on the objectives of the metabolic syndrome in clinic. Diabetes Metab Syndr Obes 2018; 11:683-697. [PMID: 30464566 PMCID: PMC6217133 DOI: 10.2147/dmso.s165740] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome (MetS), a disorder with a high and growing prevalence, is a recognized risk factor for cardiovascular disease (CVD) and type 2 diabetes. It is a constellation of clinical and metabolic risk factors that include abdominal obesity, dyslipidemia, glucose intolerance, and hypertension. Unfortunately, MetS is typically underrecognized, and there is great heterogeneity in its management, which can hamper clinical decision-making and be a barrier to achieving the therapeutic goals of CVD and diabetes prevention. Although no single treatment for MetS as a whole currently exists, management should be targeted at treating the conditions contributing to it and possibly reversing the risk factors. All this justifies the need to develop recommendations that adapt existing knowledge to clinical practice in our healthcare system. In this regard, professionals from different scientific societies who are involved in the management of the different MetS components reviewed the available scientific evidence focused basically on therapeutic aspects of MetS and developed a consensus document to establish recommendations on therapeutic goals that facilitate their homogenization in clinical decision-making.
Collapse
Affiliation(s)
- Juan Pedro-Botet
- Lipids and Vascular Risk Unit, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain,
| | - Juan F Ascaso
- Endocrinology and Nutrition Service, Hospital Clínico, Universitat de Valencia, Valencia, Spain
- INCLIVA Research Institute, Diabetes and Metabolic Diseases Ciber (Networked Biomedical Research Centres - CIBERDEM), Carlos III, Valencia, Spain
| | - Vivencio Barrios
- Cardiology Service, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, Madrid, Spain
- Department of Medicine, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Alejandro De la Sierra
- Internal Medicine Service, Hospital Mutua de Terrassa, Department of Medicine, Universidad de Barcelona, Spain
| | - Javier Escalada
- Department of Endocrinology and Nutrition, Clínica Universitaria de Navarra, IdiSNA
- CIBEROBN "Physiopathology of Obesity and Nutrition", Carlos III Health Institute, Spain
| | - Jesús Millán
- Lipid Unit, Department of Internal Medicine, Hospital Universitario General Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | - Jose M Mostaza
- Lipid and Arteriosclerosis Unit, Internal Medicine Service, Hospital Carlos III, Madrid, Spain
| | - Pablo Pérez-Martínez
- CIBEROBN "Physiopathology of Obesity and Nutrition", Carlos III Health Institute, Spain
- Lipid and Arteriosclerosis Unit, Hospital Universitario Reina Sofía, IMIBIC/University of Cordoba, Cordoba, Spain
| | - Xavier Pintó
- CIBEROBN "Physiopathology of Obesity and Nutrition", Carlos III Health Institute, Spain
- Lipids Unit, Department of Internal Medicine Hospital Universitario de Bellvitge, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- CIBEROBN "Physiopathology of Obesity and Nutrition", Carlos III Health Institute, Spain
- Nutrition Unit, Hospital Universitari Sant Joan de Reus, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili
| | - Pedro Valdivielso
- Internal Medicine Service, Department of Medicine and Dermatology, Hospital Universitario Virgen de la Victoria, Málaga Biomedicine Institute (IBIMA), Universidad de Málaga, Málaga, Spain
| |
Collapse
|
24
|
Iqbal R, Akhtar MS, Hassan MQ, Jairajpuri Z, Akhtar M, Najmi AK. Pitavastatin ameliorates myocardial damage by preventing inflammation and collagen deposition via reduced free radical generation in isoproterenol-induced cardiomyopathy. Clin Exp Hypertens 2018; 41:434-443. [PMID: 30192645 DOI: 10.1080/10641963.2018.1501059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Pitavastatin inhibits 3 hydroxy 3 methyl glutaryl coenzyme A (HMGCoA) reductase enzyme, preventing cholesterol synthesis along with elevating high density apolipoprotein A1 (Apo-A1). The present study was designed to evaluate cardioprotective potential of pitavastatin at 1 mg/kg/day and 3 mg/kg/day dose for 14 days in low dose isoproterenol (ISO) (5 mg/kg/day for 7 consecutive days) induced myocardial damage. ISO administration induced significant reduction in endogenous antioxidant enzymes like reduced glutathione (GSH), superoxide dismutase (SOD), catalase (CAT) and raised thiobarbituric acid reactive substances (TBARS) indicating activated lipid peroxidation. Along with this, a significant increase in level of cardiac injury biomarkers vie, creatine kinase (CK-MB), lactate dehydrogenase (LDH), aspartate amino transferase (AST), tumor necrosis factor (TNF-α) and transforming growth factor (TGF-β) as well as brain natriuretic peptide (BNP). Histological examination also revealed marked myocardial tissue damage in ISO treated rats. However, pretreatment with pitavastatin (3 mg/kg/day) significantly maintained nearly normal levels of cardiac biomarkers and oxidant antioxidant status as well as lipid peroxidation in ISO induced MI rats. Cardiac histological assessment and infarct size assessment also showed marked reduction in myocardial architecture alteration including infarct size as well as collagen deposition by pitavastatin that strongly supported biochemical findings. These observations strongly corroborate that pitavastatin prevents myocardial damages via up regulation of endogenous oxidants along with its hypocholesterolemic activity.
Collapse
Affiliation(s)
- Ramsha Iqbal
- a Department of Pharmacology, Faculty of Pharmacy , Jamia Hamdard , New Delhi , India
| | - Md Sayeed Akhtar
- b Department of Clinical Pharmacy , Ambo University , Ambo , Ethiopia
| | - Md Quamrul Hassan
- c Department of Ilmul Advia-Pharmacology , Faculty of Unani Medicine, Aligarh Muslim University , Aligarh , India
| | - Zeeba Jairajpuri
- d Department of Pathology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard , New Delhi , India
| | - Mohd Akhtar
- a Department of Pharmacology, Faculty of Pharmacy , Jamia Hamdard , New Delhi , India
| | - Abul Kalam Najmi
- a Department of Pharmacology, Faculty of Pharmacy , Jamia Hamdard , New Delhi , India
| |
Collapse
|
25
|
Zhang J, Shao Y, Liu Y, Tao J. A Multi-Center, Open-Label, Two-Arm Parallel Group Non-inferiority Randomized Controlled Trial Evaluating the Effect of Pitavastatin, Compared to Atorvastatin, on Glucose Metabolism in Prediabetics with Hypertension and Dyslipidemia: Rationale and Design for the China Hemoglobin A1c Metabolism Protection Union Study (CAMPUS). Cardiovasc Drugs Ther 2018; 32:581-589. [PMID: 30187345 DOI: 10.1007/s10557-018-6826-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Hypertension and dyslipidemia are major risk factors for cardiovascular disease (CVD). In 2012, over 270 million patients (25.2%) in China were hypertensive and 40.4% was dyslipidemic. The majority of these patients rely on statins for the prevention of cardiovascular disease. However, certain types of statins (e.g., atorvastatin), compared to others (e.g., pitavastatin), may be associated with unfavorable effects on glucose metabolism. This leads to concerns when prescribing statins to patients who also have a predisposition to glucose metabolic disorders (i.e., prediabetes). Thus, this study aims to investigate the effect of pitavastatin, compared to atorvastatin, on glucose metabolism, as measured by hemoglobin A1c (HbA1c), in Chinese prediabetics with hypertension and dyslipidemias. METHODS The China hemoglobin A1c Metabolism Protection Union Study (CAMPUS) is a multi-center, prospective, open-label, 12-month, two-arm parallel group, and non-inferiority randomized controlled trial (RCT). A total of 396 prediabetics with hypertension and dyslipidemias will be randomly assigned 1:1 to either pitavastatin 2 mg/day or atorvastatin 20 mg/day, and followed for 12 months (follow-up visits at 1, 3, 6, and 12 months) for HbA1c levels, as well as other measures of glucose metabolism, serum lipid levels, blood pressure control, measures of inflammation, vascular endothelial function, carotid atherosclerosis, and hypertension-related left ventricular hypertrophy. If the results of low-density lipoprotein cholesterol (LDL-C) levels in month 3 after treatment initiation do not meet individual target, drug dose for the participant would be doubled. DISCUSSION CAMPUS will be the first RCT to investigate the effect of pitavastatin, compared to atorvastatin, on glucose metabolism in Chinese prediabetics with hypertension and dyslipidemias. Further, this study might eventually provide information to design a clinical strategy, and facilitate the improvement of primary prevention in patients at risk for diabetes and CVD. TRIAL REGISTRATION ClinicalTrials.gov number: NCT03532620. Registered 22 May 2018.
Collapse
Affiliation(s)
- Jianning Zhang
- Department of Hypertension and Cardiovascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yijia Shao
- Department of Hypertension and Cardiovascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yin Liu
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jun Tao
- Department of Hypertension and Cardiovascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|