Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.796
Filter
1.
Clin Transl Sci ; 17(6): e13836, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38845393

ABSTRACT

Homozygous familial hypercholesterolemia (HoFH) is a rare and serious genetic condition characterized by premature cardiovascular disease due to severely elevated low-density lipoprotein cholesterol (LDL-C). HoFH primarily results from loss-of-function (LOF) mutations in the LDL receptor (LDLR), reducing LDL-C clearance such that patients experience severe hypercholesterolemia, exacerbating the risk of developing cardiovascular events. Treatment options such as statins, lomitapide, ezetimibe, proprotein convertase subtilisin/kexin type 9 inhibitors, and apheresis help lower LDL-C; however, many patients with HoFH still fail to reach their target LDL-C levels and many of these lipid-lowering therapies are not indicated for pediatric use. Angiopoietin-like protein 3 (ANGPTL3) has been identified as a target to treat elevated LDL-C by acting as a natural inhibitor of lipoprotein lipase (LPL) and endothelial lipase (EL), enzymes involved in the hydrolysis of the triglyceride and phospholipid content of very low-density lipoproteins. Persons heterozygous for LOF mutations in ANGPTL3 were reported to have lower LDL-C than non-carriers and lower risk of coronary artery disease. Evinacumab is a first-in-class human monoclonal antibody that specifically binds to ANGPTL3 to prevent its inhibition of LPL and EL. In clinical trials, a 15 mg/kg intravenous dose every 4 weeks has shown a mean percent change from baseline in LDL-C of ~50% in adult, adolescent, and pediatric patients with HoFH. This mini review article describes the mechanism of action of evinacumab, evinacumab population PK and PD modeling, and clinical development history of evinacumab for the treatment of HoFH.


Subject(s)
Hyperlipoproteinemia Type II , Translational Research, Biomedical , Humans , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/blood , Angiopoietin-Like Protein 3 , Cholesterol, LDL/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , Broadly Neutralizing Antibodies , Animals , Anticholesteremic Agents/therapeutic use , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/administration & dosage , Receptors, LDL/metabolism , Receptors, LDL/genetics
3.
Medicine (Baltimore) ; 103(21): e37883, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38788020

ABSTRACT

BACKGROUND: Hyperlipidemia is a common feature of chronic diseases. The aim of this work was designed to assess the role of probiotics (Lactobacillus casei Zhang, Bifidobactetium animalis subsp. lactis V9, and Lactobacillus plantarum P-8) in the treatment of hyperlipidemia. METHODS: Thirty three patients with hyperlipidemia were randomly divided into a probiotic group (n = 18) and a control group (n = 15). The probiotic group was administered probiotics (2 g once daily) and atorvastatin 20 mg (once daily), and the control group was administered a placebo (2 g once daily) and atorvastatin 20 mg (once daily). Serum and fecal samples were gathered for subsequent analyses. RESULTS: Time had a significant effect on the total cholesterol (TC), triglycerides (TG), and low-density lipoprotein-cholesterol (LDL-C) levels in the probiotic and control groups (P < .05). The gut microbial abundance in the probiotic group was markedly higher than that in the control group following 3-month probiotic treatment (P < .05). At the phylum level, probiotics exerted no notable effects on the relative abundance of Firmicutes, Bacteroidetes, and Actinobacteria but elevated that of Tenericutes and reduced Proteobacteria. At the genus level, probiotics increased the relative abundance of Bifidobacterium, Lactobacillus, and Akkermansia, and decreased that of Escherichia, Eggerthella, and Sutterella relative to the control group in months 1, 2, and 3 (P < .05). CONCLUSIONS: Probiotics optimize the gut microbiota structure and decrease the amount of harmful bacteria in patients with hyperlipidemia. Probiotics can influence the composition of gut microorganisms and increase their diversity and abundance in vivo. It is recommended to use probiotics combined with atorvastatin to treat patients with hyperlipidemia.


Subject(s)
Atorvastatin , Gastrointestinal Microbiome , Hyperlipidemias , Probiotics , Humans , Atorvastatin/administration & dosage , Atorvastatin/therapeutic use , Probiotics/administration & dosage , Probiotics/therapeutic use , Hyperlipidemias/drug therapy , Double-Blind Method , Male , Female , Middle Aged , Gastrointestinal Microbiome/drug effects , Adult , Treatment Outcome , Triglycerides/blood , Cholesterol, LDL/blood , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/therapeutic use , Lactobacillus plantarum , Feces/microbiology , Aged , Combined Modality Therapy
4.
J Am Heart Assoc ; 13(11): e033669, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38818934

ABSTRACT

BACKGROUND: A phase 3 trial was conducted to evaluate the efficacy and safety of ongericimab, a monoclonal antibody that inhibits proprotein convertase subtilisin/kexin type 9, as an add-on treatment to optimized lipid-lowering therapy in Chinese patients with primary hypercholesterolemia and mixed dyslipidemia. METHODS AND RESULTS: A total of 806 patients who were receiving stable and optimized lipid-lowering therapy but did not achieve their low-density lipoprotein cholesterol (LDL-C) targets were enrolled and randomly assigned in a 2:1:2:1 ratio to receive either ongericimab 150 mg or matching placebo every 2 weeks, or ongericimab 300 mg or matching placebo every 4 weeks for 52 weeks. Efficacy and safety were evaluated in 802 patients who received at least 1 dose of ongericimab or placebo. The primary end point was the percentage change in LDL-C from baseline to week 24. Our findings demonstrated that the least-squares mean difference of percentage change in LDL-C from baseline to week 24 was -67.7% (95% CI, -72.5% to -63.0%; P<0.0001) in the ongericimab 150 mg every 2 weeks group compared with the placebo every 2 weeks group, and -61.2% (95% CI, -67.1% to -55.2%; P<0.0001) in the ongericimab 300 mg every 4 weeks group compared with the placebo every 4 weeks group. These reductions were sustained up to week 52. Furthermore, treatment with ongericimab favorably altered other lipid parameters. A similar incidence of adverse events was observed in the ongericimab and placebo groups. CONCLUSIONS: Ongericimab, as an add-on treatment to optimized lipid-lowering therapy, significantly reduced LDL-C and was well-tolerated in Chinese patients with primary hyperlipidemia and mixed dyslipidemia who did not achieve their LDL-C targets. REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT04781114.


Subject(s)
Cholesterol, LDL , Dyslipidemias , Hypercholesterolemia , Humans , Male , Female , Middle Aged , Hypercholesterolemia/drug therapy , Hypercholesterolemia/blood , Hypercholesterolemia/diagnosis , Cholesterol, LDL/blood , China , Dyslipidemias/drug therapy , Dyslipidemias/blood , Dyslipidemias/diagnosis , Treatment Outcome , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Aged , Double-Blind Method , PCSK9 Inhibitors , Adult , Asian People , Proprotein Convertase 9/immunology , Proprotein Convertase 9/metabolism , Biomarkers/blood , Time Factors , Drug Therapy, Combination , Anticholesteremic Agents/therapeutic use , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/administration & dosage , East Asian People
5.
J Am Heart Assoc ; 13(9): e033780, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38686894

ABSTRACT

BACKGROUND: Hypertension and hypercholesterolemia are important risk factors for cardiovascular disease, and treatment with fixed-dose combination (FDC) regimens is recommended by current guidelines. However, the clinical outcomes of different FDC dosages remain unknown. This study aimed to examine the clinical outcomes of FDC regimens and the free combination of amlodipine and atorvastatin at different dosages. METHODS AND RESULTS: Patients with concurrent hypertension and hypercholesterolemia treated daily with an FDC of 5 mg amlodipine and 10 mg atorvastatin (5/10 fixed group), and FDC of 5 mg amlodipine and 20 mg atorvastatin (5/20 fixed group), or free combination of 5 mg amlodipine and 20 mg atorvastatin (5/20 free group) were identified from the National Health Insurance Research Database of Taiwan. The primary outcome was the composite cardiovascular outcomes, including cardiovascular death, acute myocardial infarction, stroke, and coronary intervention. A total of 9095 patients were eligible for inclusion. The incidence of primary outcome per 1000 person-years was 16.6 in the 5/10 fixed group, 12.6 in the 5/20 fixed group, and 16.5 in the 5/20 free group (5/20 fixed versus 5/20 free: hazard ratio [HR], 0.76 [95% CI, 0.64-0.91]; 5/20 fixed versus 5/10 fixed: HR, 0.76 [95% CI, 0.63-0.90]). CONCLUSIONS: Among patients with concomitant hypertension and hypercholesterolemia, treatment with an FDC of amlodipine and high-dose atorvastatin led to a lower risk of a composite of cardiovascular outcomes than treatment with the free combination or a similar FDC with a lower dose of atorvastatin.


Subject(s)
Amlodipine , Atorvastatin , Drug Combinations , Heptanoic Acids , Hypercholesterolemia , Hypertension , Pyrroles , Humans , Amlodipine/administration & dosage , Amlodipine/adverse effects , Male , Hypercholesterolemia/drug therapy , Hypercholesterolemia/complications , Hypercholesterolemia/epidemiology , Hypertension/drug therapy , Hypertension/complications , Hypertension/epidemiology , Female , Middle Aged , Atorvastatin/administration & dosage , Aged , Taiwan/epidemiology , Treatment Outcome , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/adverse effects , Retrospective Studies , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/therapeutic use , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/therapeutic use , Blood Pressure/drug effects
6.
Curr Cardiol Rep ; 26(5): 443-450, 2024 May.
Article in English | MEDLINE | ID: mdl-38557814

ABSTRACT

PURPOSE OF REVIEW: The polypill strategy, originally developed to improve medication adherence, has demonstrated efficacy in improving baseline systolic blood pressures and cholesterol levels in multiple clinical trials. However, the long-term clinical impact of improved major cardiovascular events (MACE) outcomes by the polypill remains uncertain. RECENT FINDINGS: Recent trials with long-term follow-up, which included minority groups and people with low socioeconomic status, have shown non-inferiority with no difference in adverse effects rates for the secondary prevention of MACE. Although the polypill strategy was initially introduced to improve adherence to guideline-directed medical therapy (GDMT) for cardiovascular complications, the strategy has surpassed standard medical treatment for secondary prevention of MACE outcomes. Studies also showed improved medication compliance in underserved populations.


Subject(s)
Cardiovascular Diseases , Medication Adherence , Secondary Prevention , Humans , Cardiovascular Diseases/prevention & control , Secondary Prevention/methods , Drug Combinations , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/administration & dosage , Anticholesteremic Agents/therapeutic use , Anticholesteremic Agents/administration & dosage
7.
Am J Cardiovasc Drugs ; 24(3): 419-431, 2024 May.
Article in English | MEDLINE | ID: mdl-38578578

ABSTRACT

AIM: Statin therapy is considered the gold standard for treating hypercholesterolemia. This updated meta-analysis aims to compare the efficacy and safety of a low/moderate-intensity statin in combination with ezetimibe compared with high-intensity statin monotherapy in patients with atherosclerotic cardiovascular disease (ASCVD). METHODS: A systematic search of two databases (PubMed and Cochrane CENTRAL) was conducted from inception to January 2023 and a total of 21 randomized clinical trials (RCTs) were identified and included in the analysis. Data were pooled using Hedges's g and a Mantel-Haenszel random-effects model to derive standard mean differences (SMDs) and 95% confidence intervals (Cis). The primary outcome studied was the effect of these treatments on lipid parameters and safety events. RESULTS: The results revealed that combination therapy was more effective in reducing low-density lipoprotein cholesterol (LDL-C) levels (SMD= - 0.41; CI - 0.63 to - 0.19; P = 0.0002). There was no significant change in the levels of high-density lipoprotein cholesterol (HDL-C), total cholesterol (TC), triglyceride (TG), high-sensitivity C-reactive protein (hs-CRP), Apo A1, or Apo B. The safety of these treatments was assessed by the following markers alanine aminotransferase (ALT), aspartate aminotransferase (AST), and creatine phosphokinase (CK), and a significant difference was only observed in CK (SMD: - 0.81; CI - 1.52 to - 0.10; P = 0.02). CONCLUSION: This meta-analysis demonstrated that the use of low/moderate-intensity statin combination therapy significantly reduced LDL-C levels compared with high-intensity statin monotherapy, making it preferable for patients with related risks. However, further trials are encouraged to evaluate potential adverse effects associated with combined therapy.


Subject(s)
Anticholesteremic Agents , Atherosclerosis , Drug Therapy, Combination , Ezetimibe , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Ezetimibe/administration & dosage , Ezetimibe/therapeutic use , Ezetimibe/adverse effects , Atherosclerosis/drug therapy , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/therapeutic use , Cholesterol, LDL/blood , Randomized Controlled Trials as Topic , Cardiovascular Diseases , Hypercholesterolemia/drug therapy , Cholesterol, HDL/blood
8.
Semergen ; 50(4): 102156, 2024.
Article in English | MEDLINE | ID: mdl-38310834

ABSTRACT

Monacolin K is the major active component in red yeast rice (RYR) which is structurally identical to lovastatin and has the most powerful effect, in terms of reducing blood cholesterol levels. This review aimed to examine the effect and safety of different doses of monacolin K on blood cholesterol levels. PubMed and Cochrane were searched for articles published between 2012 and 2023 for clinical-trials and randomized-controlled-trials. Eligible studies included participants>18-years-old, of any gender and ethnicity. The intervention/exposure of interest was monacolin K. Hypercholesterolemia was considered the outcome of interest defined as the elevated total or low-density-lipoprotein (LDL) cholesterol levels. 12 randomized-controlled-trials were eligible for inclusion in the analysis including 769 participants>18-years-old. 11 out of 12 studies were assessed with high methodological quality and one study with low methodological quality. Monacolin K supplementation varied between 2mg and 10mg per day and the maximum period of supplementation was 12 weeks. All studies indicated a beneficial effect of monacolin supplementation on LDL and total cholesterol levels (p<0.05) regardless the dose and period of supplementation. Also, 3 of the included studies reported adverse side effects after treatment with monacolin K. Low doses of monacolin K equal to 3mg/day exert potential cholesterol-lowering effects although the number of relative studies is limited. Regarding the safety of monacolin K supplementation, findings seem to be more controversial and therefore, it is suggested for all patients treated with monacolin K to be routinely monitored regardless the dose of supplementation.


Subject(s)
Anticholesteremic Agents , Biological Products , Cholesterol, LDL , Dicarboxylic Acids , Dietary Supplements , Fatty Acids , Hypercholesterolemia , Lovastatin , Randomized Controlled Trials as Topic , Humans , Hypercholesterolemia/drug therapy , Lovastatin/administration & dosage , Lovastatin/adverse effects , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Cholesterol, LDL/blood , Dose-Response Relationship, Drug , Cholesterol/blood
9.
Eur J Intern Med ; 124: 99-107, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38336550

ABSTRACT

OBJECTIVE: Scant data is available on the efficacy and safety of adding ezetimibe to high-intensity statin therapy for early and rapid reduction of low-density lipoprotein cholesterol (LDL-C) within 4-12 weeks of an acute-event in acute coronary syndrome (ACS). We undertook this meta-analysis to address this knowledge-gap. METHODS: Electronic databases were searched for RCTs involving patients with ACS receiving ezetimibe in intervention arm, and placebo/active comparator in control arm. Primary outcome was to evaluate changes in LDL-C levels post-ACS. Secondary outcomes were to evaluate alterations in other lipid parameters and adverse events. RESULTS: From initially screened 4561 articles, data from 11 studies (20,291 patients) were analyzed. Compared to controls, patients receiving ezetimibe had significantly lower LDL-C at 7-days [MD -19.55 mg/dl(95 %CI:-36.46 to -2.63);P = 0.02;I2 = 91 %], 1-month [MD-24.67 mg/dl (95 %CI:-34.59 to -14.76);P < 0.001;I2 = 81 %], 3-months [MD -18.01 mg/dl(95 %CI:-24.11 to -11.90);P < 0.001;I2 = 92 %] and 10-12 months [MD -16.90 mg/dl (95 % CI: -17.67 to -16.12); P < 0.001; I2 = 0 %] of treatment. Compared to controls, patients receiving ezetimibe had significantly lower total cholesterol at 7-days [MD-21.05 mg/dl(95 %CI:-26.73 to -15.37);P < 0.001;I2 = 0 %], 1-month [MD-25.56 mg/dl(95 %CI:-38.29 to -12.83);P < 0.001;I2 = 85 %], 3-months [MD-22.54 mg/dl(95 %CI:-36.90 to -8.19);P = 0.002;I2 = 22 %] and 12-months [MD-19.68 mg/dl(95 %CI:-20.78 to -18.59);P < 0.001;I2 = 0 %] of treatment. Death from any cause, ACS and non-fatal stroke [OR0.89(95 %CI:0.83-0.96);P = 0.002;I2 = 0 %], non-fatal myocardial infarction [OR0.86(95 %CI:0.79-0.94);P = 0.001;I2 = 0 %] and ischemic stroke [OR0.80(95 %CI:0.68-0.94);P = 0.009;I2 = 0 %] was significantly reduced in patients receiving ezetimibe. CONCLUSION: Addition of ezetimibe to high-intensity statin therapy at the time of ACS event is associated with significantly better cholesterol reduction at day-7,1-month, 3- months and 1-year of follow-up, which translates into a significantly lower recurrent cardiovascular events post an index event of ACS. CONCISE SUMMARY OF FINDINGS: Addition of ezetimibe to high-intensity statin therapy at the time of acute coronary syndrome (ACS) index event is associated with significantly better low density lipoprotein cholesterol and total cholesterol reduction at day-7, 1-month, 3-months and 1-year of follow-up, which translates into a significantly lower recurrent cardiovascular events (death from any cause, major ACS, non-fatal stroke, non-fatal myocardial infarction, and ischemic stroke) post an index event of ACS.


Subject(s)
Acute Coronary Syndrome , Anticholesteremic Agents , Cholesterol, LDL , Ezetimibe , Humans , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/blood , Ezetimibe/therapeutic use , Anticholesteremic Agents/therapeutic use , Anticholesteremic Agents/administration & dosage , Cholesterol, LDL/blood , Randomized Controlled Trials as Topic , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Drug Therapy, Combination , Treatment Outcome
10.
Adv Ther ; 40(12): 5285-5299, 2023 12.
Article in English | MEDLINE | ID: mdl-37770770

ABSTRACT

INTRODUCTION: Many patients with primary hypercholesterolemia do not achieve their plasma low-density lipoprotein cholesterol (LDL-C) goals with statin alone under a recommended dose of statin (e.g., 10 mg rosuvastatin) in China. The objective of this phase III study was to evaluate the efficacy and safety of a new single-pill combination (SPC) of rosuvastatin 10 mg/ezetimibe 10 mg (R10/E10) in this population. METHODS: This was a randomized, double-blind, double-dummy, active-controlled study in patients with primary hypercholesterolemia inadequately controlled with statin alone. The participants were randomized 1:1 to receive SPC R10/E10 or R10. The primary objective was to demonstrate the superiority of SPC R10/E10 vs. R10 in reducing the LDL-C levels after 8 weeks. RESULTS: This trial randomized 305 participants to SPC R10/E10 (n = 153) and R10 (n = 152). The superiority of SPC R10/E10 over R10 was demonstrated with the least square (LS) mean difference of percent change in LDL-C from baseline to week 8: - 13.85% (95% confidence interval [CI] - 20.15% to - 7.56%, P < 0.0001). The proportion of participants who achieved the LDL-C target (< 2.6 mmol/l) at week 8 was larger with SPC R10/E10 (n = 80, 54.1%) than with R10 (n = 42, 29.2%) (Odds ratio = 2.80, 95% CI 1.70 to 4.61, P < 0.0001). No unexpected safety findings were reported. CONCLUSION: The results suggest that SPC R10/E10 improve LDL-C reduction and goal achievement in Chinese patients with primary hypercholesterolemia not adequately controlled on statin therapy, without new safety findings. TRIAL REGISTRATION: ClinicalTrials.gov (NCT04669041).


Subject(s)
Anticholesteremic Agents , Ezetimibe , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hypercholesterolemia , Rosuvastatin Calcium , Humans , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/therapeutic use , Cholesterol, LDL , Double-Blind Method , East Asian People , Ezetimibe/administration & dosage , Ezetimibe/therapeutic use , Hypercholesterolemia/drug therapy , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/therapeutic use , Treatment Outcome , Drug Combinations
11.
N Engl J Med ; 387(20): 1855-1864, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36342163

ABSTRACT

BACKGROUND: Lipoprotein(a) is a presumed risk factor for atherosclerotic cardiovascular disease. Olpasiran is a small interfering RNA that reduces lipoprotein(a) synthesis in the liver. METHODS: We conducted a randomized, double-blind, placebo-controlled, dose-finding trial involving patients with established atherosclerotic cardiovascular disease and a lipoprotein(a) concentration of more than 150 nmol per liter. Patients were randomly assigned to receive one of four doses of olpasiran (10 mg every 12 weeks, 75 mg every 12 weeks, 225 mg every 12 weeks, or 225 mg every 24 weeks) or matching placebo, administered subcutaneously. The primary end point was the percent change in the lipoprotein(a) concentration from baseline to week 36 (reported as the placebo-adjusted mean percent change). Safety was also assessed. RESULTS: Among the 281 enrolled patients, the median concentration of lipoprotein(a) at baseline was 260.3 nmol per liter, and the median concentration of low-density lipoprotein cholesterol was 67.5 mg per deciliter. At baseline, 88% of the patients were taking statin therapy, 52% were taking ezetimibe, and 23% were taking a proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitor. At 36 weeks, the lipoprotein(a) concentration had increased by a mean of 3.6% in the placebo group, whereas olpasiran therapy had significantly and substantially reduced the lipoprotein(a) concentration in a dose-dependent manner, resulting in placebo-adjusted mean percent changes of -70.5% with the 10-mg dose, -97.4% with the 75-mg dose, -101.1% with the 225-mg dose administered every 12 weeks, and -100.5% with the 225-mg dose administered every 24 weeks (P<0.001 for all comparisons with baseline). The overall incidence of adverse events was similar across the trial groups. The most common olpasiran-related adverse events were injection-site reactions, primarily pain. CONCLUSIONS: Olpasiran therapy significantly reduced lipoprotein(a) concentrations in patients with established atherosclerotic cardiovascular disease. Longer and larger trials will be necessary to determine the effect of olpasiran therapy on cardiovascular disease. (Funded by Amgen; OCEAN[a]-DOSE ClinicalTrials.gov number, NCT04270760.).


Subject(s)
Anticholesteremic Agents , Atherosclerosis , Hypercholesterolemia , Lipoprotein(a) , RNA, Small Interfering , Humans , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Atherosclerosis/drug therapy , Cardiovascular Diseases/drug therapy , Double-Blind Method , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Lipoprotein(a)/analysis , Lipoprotein(a)/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/adverse effects , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Liver/drug effects , Liver/metabolism , PCSK9 Inhibitors/therapeutic use , Ezetimibe/therapeutic use
12.
Pharm Biol ; 60(1): 282-293, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35138992

ABSTRACT

CONTEXT: Cordia dichotoma Forst. (Boraginaceae) has potent pharmacological impact. Meanwhile, its effect on fertility is unclear. OBJECTIVE: This study investigates the effect of Cordia fresh fruits hydroethanolic extract on fertility. MATERIALS AND METHODS: 120 Wistar albino male rats were divided into four groups (n = 30). The first group was negative control, and the second, third, and fourth groups received 125, 250, and 500 mg extract/kg bodyweight for 56 days. After 56 days, Cordia force-feeding stopped, and all groups were kept under laboratory conditions for another month to study the recovering effect. RESULTS: After day 56, extract at 500 mg/kg significantly reduced sperm total count, motility%, and alive%, to 47.60 ± 2.27 × 106 sperm/mL, 43.33% ± 1.49, and 63.67% ± 1.19, respectively, abnormalities% increased considerably (26.67% ± 0.54), compared to the negative control. Also, significant depletion on follicle-stimulating hormone (2.66 ± 0.21 mIU/L), luteinizing hormone (1.07 ± 0.06 mIU/L), and testosterone (2.69 ± 0.13 nmol/L) level was recorded, compared to the negative control. Cordia negative effect showed on histopathological studies of testes, prostate, and seminal vesicles. Fortunately, these adverse effects of Cordia recovered remarkably after stopping administration for one month. CONCLUSIONS: Cordia antifertility effect may be due to its hypocholesterolemic effect, where cholesterol, the steroid cycle precursor, was significantly reduced. This study can be incorporated in clinical research after being repeated on another small experimental animal, their offspring, and one large experimental animal, then going to a clinical study that we plan to do in the future.


Subject(s)
Cordia/chemistry , Plant Extracts/toxicity , Spermatozoa/drug effects , Testis/drug effects , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/isolation & purification , Anticholesteremic Agents/toxicity , Dose-Response Relationship, Drug , Follicle Stimulating Hormone/metabolism , Fruit , Luteinizing Hormone/metabolism , Male , Plant Extracts/administration & dosage , Rats , Rats, Wistar , Testis/pathology , Testosterone/metabolism
13.
Biomed Pharmacother ; 146: 112519, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34968928

ABSTRACT

INTRODUCTION: Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) have emerged as a therapeutic option for patients with hypercholesterolemia who do not attain low-density lipoprotein cholesterol (LDL-C) goals and/or are intolerant to other lipid-lowering drugs. Our aim was to analyze the effectiveness and safety of PCSK9i in routine clinical practice and factors related to poor outcomes. MATERIALS AND METHODS: We conducted an ambispective study in 115 patients who recieved alirocumab or evolocumab, in a tertiary level hospital. From February 2017 to April 2020, patients were recruited and followed up for a median of 20.4 months. The main outcomes were relative reduction in LDL-C, percentage of patients achieving the therapeutic goals established by 2016 ESC/EAS guidelines, incidence of major cardiovascular events (MACEs) and drug-related adverse events (ADRs). RESULTS: The median LDL-C achieved was 57.0 mg/dL (relative reduction of 59.9% from baseline, p< 0.001). After adjusting for confounders, smaller LDL-C reductions were related to female sex, absence of concomitant lipid-lowering therapy and treatment with alirocumab. Overall, 84.6% of the patients achieved the therapeutic goals. During follow-up, 7 MACEs were detected. ADRs, generally considered mild, affected 38.1% of the participants (mainly mialgias and arthralgias) and triggered discontinuations in 8.7% of cases. CONCLUSIONS: PCSK9i are effective and safe, although certain factors may influence their effectiveness. Interestingly, our results suggest that alirocumab and evolocumab may not be therapeutic equivalents, as initially suggested.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Anticholesteremic Agents/therapeutic use , Hypercholesterolemia/drug therapy , PCSK9 Inhibitors/therapeutic use , Age Factors , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Cardiovascular Diseases/epidemiology , Comorbidity , Dose-Response Relationship, Drug , Female , Humans , Life Style , Lipids/blood , Male , Middle Aged , PCSK9 Inhibitors/administration & dosage , PCSK9 Inhibitors/adverse effects , Sex Factors , Spain
14.
Biochem Pharmacol ; 196: 114731, 2022 02.
Article in English | MEDLINE | ID: mdl-34407453

ABSTRACT

Cholesterol esterification proteins Sterol-O acyltransferases (SOAT) 1 and 2 are emerging prognostic markers in many cancers. These enzymes utilise fatty acids conjugated to coenzyme A to esterify cholesterol. Cholesterol esterification is tightly regulated and enables formation of lipid droplets that act as storage organelles for lipid soluble vitamins and minerals, and as cholesterol reservoirs. In cancer, this provides rapid access to cholesterol to maintain continual synthesis of the plasma membrane. In this systematic review and meta-analysis, we summarise the current depth of understanding of the role of this metabolic pathway in pan-cancer development. A systematic search of PubMed, Scopus, Web of Science, and Cochrane Library for preclinical studies identified eight studies where cholesteryl ester concentrations were compared between tumour and adjacent-normal tissue, and 24 studies where cholesterol esterification was blocked by pharmacological or genetic approaches. Tumour tissue had a significantly greater concentration of cholesteryl esters than non-tumour tissue (p < 0.0001). Pharmacological or genetic inhibition of SOAT was associated with significantly smaller tumours of all types (p ≤ 0.002). SOAT inhibition increased tumour apoptosis (p = 0.007), CD8 + lymphocyte infiltration and cytotoxicity (p ≤ 0.05), and reduced proliferation (p = 0.0003) and metastasis (p < 0.0001). Significant risk of publication bias was found and may have contributed to a 32% overestimation of the meta-analysed effect size. Avasimibe, the most frequently used SOAT inhibitor, was effective at doses equivalent to those previously reported to be safe and tolerable in humans. This work indicates that SOAT inhibition should be explored in clinical trials as an adjunct to existing anti-neoplastic agents.


Subject(s)
Anticholesteremic Agents/administration & dosage , Cholesterol/genetics , Cholesterol/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Tumor Burden/drug effects , Animals , Antineoplastic Agents/administration & dosage , Clinical Trials as Topic/methods , Esterification/drug effects , Esterification/physiology , Humans , Organic Anion Transporters/antagonists & inhibitors , Tumor Burden/physiology , Urea/administration & dosage , Urea/analogs & derivatives , Xenograft Model Antitumor Assays/methods
15.
Cardiovasc Drugs Ther ; 36(1): 173-186, 2022 02.
Article in English | MEDLINE | ID: mdl-32979175

ABSTRACT

The widespread availability of highly effective antiretroviral therapies has reduced mortality from opportunistic infections in persons living with HIV (PLHIV), resulting in an increase in atherosclerotic cardiovascular disease (ASCVD) and other chronic illnesses (Samji et al. 2013). Although there has been a decline in morbidity and mortality from ASCVD in the past several decades, contemporary studies continue to report higher rates of cardiovascular events (Rosenson et al. 2020). HIV has been identified as a risk enhancer for ASCVD by multiple professional guideline writing committees (Grundy Scott et al. 2019, Mach et al. 2020); however, the utilization of cholesterol-lowering therapies in PLHIV remains low (Rosenson et al. 2018). Moreover, the use of statin therapy in PLHIV is complicated by drug-drug interactions that may either elevate or lower the blood statin concentrations resulting in increased toxicity or reduced efficacy respectively. Other comorbidities commonly associated with HIV present other challenges for the use of cholesterol-lowering therapies. This review will summarize the data on lipoprotein-associated ASCVD risk in PLHIV and discuss the challenges with effective treatment. Finally, we present a clinical algorithm to optimize cardiovascular risk reduction in this high-risk population.


Subject(s)
Anticholesteremic Agents/pharmacology , Cardiovascular Diseases/prevention & control , HIV Infections/complications , Algorithms , Anti-HIV Agents/administration & dosage , Anticholesteremic Agents/administration & dosage , Atherosclerosis/etiology , Atherosclerosis/prevention & control , Cardiovascular Diseases/etiology , Cholesterol/blood , Drug Interactions , HIV Infections/drug therapy , Heart Disease Risk Factors , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
16.
Clin Res Cardiol ; 111(3): 243-252, 2022 Mar.
Article in English | MEDLINE | ID: mdl-32949286

ABSTRACT

BACKGROUND: Many patients at very-high atherosclerotic cardiovascular disease risk do not reach guideline-recommended targets for LDL-C. There is a lack of data on real-world use of non-statin lipid-lowering therapies (LLT) and little is known on the effectiveness of fixed-dose combinations (FDC). We therefore studied prescription trends in oral non-statin LLT and their effects on LDL-C. METHODS: A retrospective analysis was conducted of electronic medical records of outpatients at very-high cardiovascular risk treated by general practitioners (GPs) and cardiologists, and prescribed LLT in Germany between 2013 and 2018. RESULTS: Data from 311,242 patients were analysed. Prescriptions for high-potency statins (atorvastatin and rosuvastatin) increased from 10.4% and 25.8% of patients treated by GPs and cardiologists, respectively, in 2013, to 34.7% and 58.3% in 2018. Prescription for non-statin LLT remained stable throughout the period and low especially for GPs. Ezetimibe was the most prescribed non-statin LLT in 2018 (GPs, 76.1%; cardiologists, 92.8%). Addition of ezetimibe in patients already prescribed a statin reduced LDL-C by an additional 23.8% (32.3 ± 38.4 mg/dL), with a greater reduction with FDC [reduction 28.4% (40.0 ± 39.1 mg/dL)] as compared to separate pills [19.4% (27.5 ± 33.8 mg/dL)]; p < 0.0001. However, only a small proportion of patients reached the recommended LDL-C level of < 70 mg/dL (31.5% with FDC and 21.0% with separate pills). CONCLUSIONS: Prescription for high-potency statins increased over time. Non-statin LLT were infrequently prescribed by GPs. The reduction in LDL-C when statin and ezetimibe were prescribed in combination was considerably larger for FDC; however, a large proportion of patients still remained with uncontrolled LDL-C levels.


Subject(s)
Anticholesteremic Agents/administration & dosage , Atherosclerosis/drug therapy , Cholesterol, LDL/drug effects , Ezetimibe/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hypercholesterolemia/drug therapy , Aged , Cross-Sectional Studies , Drug Prescriptions/statistics & numerical data , Drug Therapy, Combination , Female , General Practice/statistics & numerical data , Germany , Humans , Male , Middle Aged , Practice Patterns, Physicians'/statistics & numerical data , Retrospective Studies , Time Factors , Treatment Outcome
17.
Eur J Clin Pharmacol ; 78(1): 111-126, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34423376

ABSTRACT

PURPOSE: We aimed to summarize current evidence regarding the impact of a high-dose statin loading before percutaneous coronary intervention (PCI) on short-term outcomes in patients presenting with the acute coronary syndrome (ACS). METHODS: This meta-analysis was based on a search of the MEDLINE, Cochrane Central Register of Controlled Trials, Ovid Journals, and SCOPUS for randomized controlled trials that compared high-dose atorvastatin or rosuvastatin with no or low-dose statin administered before planned PCI in statin-naive patients with ACS. The primary endpoints were major adverse cardiovascular and cerebrovascular events (MACCE), myocardial infarction (MI), and all-cause mortality at 30 days. Prespecified subanalyses were performed with respect to statin and ACS type. RESULTS: A total of eleven trials enrolling 6291 patients were included, of which 75.4% received PCI. High-dose statin loading was associated with an overall 43% relative risk (RR) reduction in MACCE at 30 days (RR 0.57, 95% CI 0.41-0.77) in whole ACS population. This effect was primarily driven by the 39% reduction in the occurrence of MI (RR 0.61, 95% CI 0.46-0.80). No significant effect on all-cause mortality reduction was observed (RR 0.92, 95% CI 0.67-1.26). In the setting of ST-elevation myocardial infarction (STEMI), atorvastatin loading was associated with a 33% reduction in MACCE (RR 0.67, 95% CI 0.48-0.94), while in non-ST-elevation myocardial infarction ACS (NSTE-ACS), rosuvastatin loading was associated with 52% reduction in MACCE at 30 days (RR 0.48, 95% CI 0.34-0.66). The level of evidence as qualified with GRADE was low to high, depending on the outcome. CONCLUSION: A high-dose loading of statins before PCI in patients with ACS reduces MACCE and reduces the risk of MI with no impact on mortality at 30 days. Atorvastatin reduces MACCE in STEMI while rosuvastatin reduces MACCE in NSTE-ACS at 30 days.


Subject(s)
Acute Coronary Syndrome/therapy , Anticholesteremic Agents/administration & dosage , Atorvastatin/administration & dosage , Percutaneous Coronary Intervention/methods , Rosuvastatin Calcium/administration & dosage , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/surgery , Aged , Cardiovascular Diseases/prevention & control , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Myocardial Infarction/prevention & control , Randomized Controlled Trials as Topic
18.
J Ethnopharmacol ; 282: 114590, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-34487844

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Carthamus tinctorius L. (Safflower) has been widely recommended to treat metabolic disorders in traditional herbal medicine in Persia, China, Korea, Japan, and other East-Asian countries. The anti-hypercholesterolemic and antioxidant effects of this plant have been well documented, but its protective effects against Metabolic Syndrome (MetS) have not been fully illustrated. AIM OF THE STUDY: The present study aimed to evaluate the effects of safflower oil on MetS risk factors. MATERIALS AND METHODS: In this randomized, double-blind, placebo-controlled clinical trial, 67 patients with MetS were administered either divided 8 g safflower oil or placebo daily for 12 weeks. All patients were advised to follow their previous diets and physical activities. RESULTS: Safflower oil resulted in a significant reduction in waist circumference (-2.42 ± 3.24 vs. 0.97 ± 2.53, p<0.001), systolic blood pressure (-8.80 ± 9.77 vs. -2.26 ± 8.56, p = 0.021), diastolic blood pressure (-3.53 ± 7.52 vs. -0.70 ± 6.21, p = 0.041), fasting blood sugar (-5.03 ± 10.62 vs. 2.94 ± 7.57, p = 0.003), and insulin resistance (-0.59 ± 1.43 vs. 0.50 ± 1, p = 0.012), but an increase in adiponectin level (0.38 ± 0.99 vs. -0.09 ± 0.81, p = 0.042) in the treatment group in comparison to the placebo group. The results revealed a direct relationship between leptin level and Body Mass Index (BMI) in both groups (p<0.001). In addition, increase in BMI resulted in a non-significant decrease in adiponectin level in both groups. Moreover, no significant difference was observed between the two groups regarding lipid profiles, leptin serum level, serum creatinine concentration, and other outcomes. CONCLUSION: Safflower oil without lifestyle modification improved abdominal obesity, blood pressure, and insulin resistance in patients with MetS.


Subject(s)
Blood Glucose/analysis , Blood Pressure Determination , Carthamus tinctorius , Metabolic Syndrome , Obesity, Abdominal , Safflower Oil/administration & dosage , Adiponectin/blood , Adult , Anticholesteremic Agents/administration & dosage , Antioxidants/administration & dosage , Blood Pressure Determination/methods , Blood Pressure Determination/statistics & numerical data , Body Mass Index , Double-Blind Method , Drug Monitoring/methods , Female , Humans , Insulin Resistance , Male , Medicine, Persian/methods , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Obesity, Abdominal/diagnosis , Obesity, Abdominal/drug therapy , Obesity, Abdominal/metabolism , Phytotherapy/methods , Treatment Outcome
19.
Biochem Pharmacol ; 196: 114725, 2022 02.
Article in English | MEDLINE | ID: mdl-34384758

ABSTRACT

Aberrant cholesterol metabolism and homeostasis in the form of elevated cholesterol biosynthesis and dysregulated efflux and metabolism is well recognized as a major feature of metabolic reprogramming in solid tumors. Recent studies have emphasized on major drivers and regulators such as Myc, mutant p53, SREBP2, LXRs and oncogenic signaling pathways that play crucial roles in tumor cholesterol metabolic reprogramming. Therapeutics such as statins targeting the mevalonate pathway were tried at the clinic without showing consistent benefits to cancer patients. Nuclear receptors are prominent regulators of mammalian metabolism. Their de-regulation often drives tumorigenesis. RORγ and its immune cell-specific isoform RORγt play important functions in control of mammalian metabolism, circadian rhythm and immune responses. Although RORγ, together with its closely related members RORα and RORß were identified initially as orphan receptors, recent studies strongly support the conclusion that specific intermediates and metabolites of cholesterol pathways serve as endogenous ligands of RORγ. More recent studies also reveal a critical role of RORγ in tumorigenesis through major oncogenic pathways including acting a new master-like regulator of tumor cholesterol biosynthesis program. Importantly, an increasing number of RORγ orthosteric and allosteric ligands are being identified that display potent activities in blocking tumor growth and autoimmune disorders in preclinical models. This review summarizes the recent preclinical and clinical progress on RORγ with emphasis on its role in reprogramming tumor cholesterol metabolism and its regulation. It will also discuss RORγ functional mechanisms, context-specificity and its value as a therapeutic target for effective cancer treatment.


Subject(s)
Anticholesteremic Agents/administration & dosage , Autoimmune Diseases/metabolism , Cholesterol/biosynthesis , Neoplasms/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Oncogenes/physiology , Animals , Antineoplastic Agents/administration & dosage , Autoimmune Diseases/drug therapy , Drug Delivery Systems/trends , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Neoplasms/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Oncogenes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...