Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Rev Clin Exp Pharmacol ; 18(2): 120-147, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35642121

RESUMEN

BACKGROUND: A subpopulation of statin users such as subjects with chronic kidney disease (CKD), Human Immune virus (HIV), acute coronary syndrome (ACS), revascularization, metabolic syndrome, and/or diabetes may particularly benefit from pitavastatin pharmacotherapy. AIM: The current systematic review aimed systematically to evaluate the effect of pitavastatin on primary cardiac events in subjects receiving pitavastatin in comparison to the other four statin members. METHODS: We conducted a systematic review on phases III and IV of randomized controlled trials (RCT-s, 11 trials) for subjects with primary cardiac events who received pitavastatin. Subjects diagnosed with any type of dyslipidemia (population 4804) and received pitavastatin (interventions) versus comparator (comparison) with the primary efficacy endpoint of minimization of LDL-C and non- HDL-C, had an increase in HDL-C and/or reduction in major adverse cardiac events (MACE, cardiovascular death, myocardial infarction (fatal/nonfatal), and stroke (fatal/nonfatal) and/or their composite (outcomes). The secondary safety endpoint was the development of any adverse effects. RESULTS: In the included trials (11), participants (4804) were randomized for pitavastatin or its comparators such as atorvastatin, pravastatin, rosuvastatin, simvastatin and followed up for 12 to 52 weeks. In terms of the primary outcome (reduction in LDL-C), pitavastatin 4 mg was superior to pravastatin 40 mg in three trials, while the 2 mg pitavastatin was comparable to atorvastatin 10 mg in four trials and simvastatin 20 and 40 mg in two 2 trials. However, rosuvastatin 2.5 mg was superior to pitavastatin 2 mg in two trials. Pitavastatin increased HDL-C and reduced non-HDL-C in eleven trials. Regarding the safety profile, pitavastatin has proved to be tolerated and safe. CONCLUSION: The FDA-approved indications for pitavastatin included primary dyslipidemia and mixed dyslipidemia as a supplementary therapy to dietary changes to lower total cholesterol, LDL-C, apolipoprotein B (Apo B), triglycerides (TG), and enhance HDL-C. Pitavastatin might be suitable for subjects with diabetes, ACS (reduced revascularization), metabolic syndrome, CKD, HIV, and subjects with low levels of HDL-C. We highly recommend rational individualization for the selection of statin.


Asunto(s)
Enfermedades Cardiovasculares , Dislipidemias , Infecciones por VIH , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Síndrome Metabólico , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Atorvastatina/uso terapéutico , Rosuvastatina Cálcica/uso terapéutico , Pravastatina/uso terapéutico , LDL-Colesterol/uso terapéutico , Síndrome Metabólico/inducido químicamente , HDL-Colesterol/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Simvastatina/uso terapéutico , Dislipidemias/tratamiento farmacológico , Enfermedades Cardiovasculares/tratamiento farmacológico , Infecciones por VIH/complicaciones
2.
Am J Obstet Gynecol ; 226(2S): S1157-S1170, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-32946849

RESUMEN

There has been increasing research momentum to identify new therapeutic agents for the prevention or treatment of preeclampsia, drugs that can affect the underlying disease pathophysiology. Molecular targets of candidate treatments include oxidative stress, antiangiogenic factors, and the angiotensin, nitric oxide, and proinflammatory pathways. The proposed treatments undergoing preclinical and clinical trial evaluation are thought to act on placental or endothelial disease or both. Most have adopted the pragmatic strategy of repurposing drugs. Of all the therapeutic agents proposed, pravastatin has received the most interest. There are preclinical studies showing that it has pleiotropic actions that favorably impact on multiple molecular targets and can resolve a preeclampsia phenotype in many animal models. An early phase clinical trial suggests that it may have therapeutic activity. Several large prevention trials are planned or ongoing and, when completed, could definitively address whether pravastatin can prevent preeclampsia. Proton-pump inhibitors, metformin, and sulfasalazine are other drugs with preclinical evidence of multiple molecular actions that could resolve the pathophysiology of preeclampsia. These agents are also currently being evaluated in clinical trials. There have been many recent preclinical studies identifying the potential of numerous natural compounds to treat preeclampsia, such as plant extracts and micronutrients that have potent anti-inflammatory or antioxidant activity. Recent preclinical studies have also proposed novel molecular-targeted strategies, such as monoclonal antibodies targeting tumor necrosis factor alpha, placental growth factor, and short interfering RNA technology, to silence the gene expression of soluble fms-like tyrosine kinase-1 or angiotensinogen. Other treatment approaches that have transitioned to human trials (ranging from single-arm to phase III trials that have been completed or are ongoing) include folic acid, nitric oxide donors (such as L-arginine), recombinant antithrombin III, digoxin immune antigen-binding fragment, and melatonin. There have been case series showing the removal of circulating soluble fms-like tyrosine kinase-1 may help stabilize the disease and prolong pregnancy. Interestingly, there are case reports suggesting that monoclonal antibody eculizumab (complement inhibitor) may have therapeutic potential. If new agents are discovered that are proven to be effective in preventing or treating preeclampsia, the potential to improve global maternal and perinatal health will be significant.


Asunto(s)
Preeclampsia/prevención & control , Anticuerpos Monoclonales/uso terapéutico , Antioxidantes/uso terapéutico , Antitrombina III/uso terapéutico , Productos Biológicos/uso terapéutico , Eliminación de Componentes Sanguíneos , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Micronutrientes/uso terapéutico , Factor de Crecimiento Placentario/uso terapéutico , Extractos Vegetales/uso terapéutico , Pravastatina/uso terapéutico , Embarazo , Inhibidores de la Bomba de Protones/uso terapéutico , ARN Interferente Pequeño , Proteínas Recombinantes/uso terapéutico , Sulfasalazina/uso terapéutico , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre
3.
Hepatol Int ; 15(1): 93-104, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33420951

RESUMEN

BACKGROUND AND AIMS: There is limited data regarding the role for systemic treatment in patients with Hepatocellular Carcinoma with Child-Pugh B cirrhosis. METHODS: PRODIGE 21 was a multicentric prospective non-comparative randomized trial. Patients were randomized to receive sorafenib (Arm A), pravastatin (Arm B), sorafenib-pravastatin (Arm C) combination, or best supportive care (Arm D). Primary endpoint was time to progression (TTP), secondary endpoints included safety and overall survival (OS). RESULTS: 160 patients were randomized and 157 patients were included in the final analysis. 86% of patients were BCLC C and 55% had macrovascular invasion. The safety profiles of the drugs were as expected. Median TTP was 3.5, 2.8, 2.0 and 2.2 months in arms A, B, C and D, respectively, but analysis was limited by the number of patients deceased without radiological progression (59%). Median OS was similar between the four arms: 3.8 [95% CI: 2.4-6.5], 3.1 [95% CI: 1.9-4.3], 4.0 [95% CI: 3.2-5.5] and 3.5 months [95% CI: 2.2-5.4] in arms A, B, C and D, respectively. Median OS was 4.0 months [95% CI: 3.3-5.5] for patients treated with sorafenib, vs 2.9 months [95% CI: 2.2-3.9] for patients not treated with sorafenib. In patients with ALBI grade 1/2, median OS was 6.1 months [95% CI: 3.8-8.3] in patients treated with sorafenib vs 3.1 months [95% CI: 1.9-4.8] for patients not treated with sorafenib. CONCLUSION: In the overall Child-Pugh B population, neither sorafenib nor pravastatin seemed to provide benefit. In the ALBI grade 1/2 sub-population, our trial suggests potential benefit of sorafenib. CLINICAL TRIAL REGISTRATION: The study was referenced in clinicaltrials.gov (NCT01357486).


Asunto(s)
Carcinoma Hepatocelular , Cirrosis Hepática , Neoplasias Hepáticas , Pravastatina/uso terapéutico , Sorafenib/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/etiología , Combinación de Medicamentos , Humanos , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/etiología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/uso terapéutico , Estudios Prospectivos , Resultado del Tratamiento
4.
Nutrients ; 12(8)2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32722395

RESUMEN

Statins and omega-3 supplementation have been recommended for cardiovascular disease prevention, but comparative effects have not been investigated. This study aimed to summarize current evidence of the effect of statins and omega-3 supplementation on cardiovascular events. A meta-analysis and a network meta-analysis of 63 randomized controlled trials were used to calculate pooled relative risks (RRs) and 95% confidence intervals (CIs) for the effects of specific statins and omega-3 supplementation compared with controls. Overall, the statin group showed significant risk reductions in total cardiovascular disease, coronary heart disease, myocardial infarction, and stroke; however, omega-3 supplementation significantly decreased the risks of coronary heart disease and myocardial infarction only, in the comparison with the control group. In comparison with omega-3 supplementation, pravastatin significantly reduced the risks of total cardiovascular disease (RR = 0.81, 95% CI = 0.72-0.91), coronary heart disease (RR = 0.75, 95% CI = 0.60-0.94), and myocardial infarction (RR = 0.71, 95% CI = 0.55-0.94). Risks of total cardiovascular disease, coronary heart disease, myocardial infarction, and stroke in the atorvastatin group were statistically lower than those in the omega-3 group, with RRs (95% CIs) of 0.80 (0.73-0.88), 0.64 (0.50-0.82), 0.75 (0.60-0.93), and 0.81 (0.66-0.99), respectively. The findings of this study suggest that pravastatin and atorvastatin may be more beneficial than omega-3 supplementation in reducing the risk of total cardiovascular disease, coronary heart disease, and myocardial infarction.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Suplementos Dietéticos , Ácidos Grasos Omega-3/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Anciano , Atorvastatina/uso terapéutico , Enfermedad Coronaria/prevención & control , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/prevención & control , Metaanálisis en Red , Pravastatina/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Accidente Cerebrovascular/prevención & control , Resultado del Tratamiento
5.
Nat Commun ; 11(1): 1990, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32332749

RESUMEN

Up-regulation of utrophin in muscles represents a promising therapeutic strategy for the treatment of Duchenne Muscular Dystrophy. We previously demonstrated that eEF1A2 associates with the 5'UTR of utrophin A to promote IRES-dependent translation. Here, we examine whether eEF1A2 directly regulates utrophin A expression and identify via an ELISA-based high-throughput screen, FDA-approved drugs that upregulate both eEF1A2 and utrophin A. Our results show that transient overexpression of eEF1A2 in mouse muscles causes an increase in IRES-mediated translation of utrophin A. Through the assessment of our screen, we reveal 7 classes of FDA-approved drugs that increase eEF1A2 and utrophin A protein levels. Treatment of mdx mice with the 2 top leads results in multiple improvements of the dystrophic phenotype. Here, we report that IRES-mediated translation of utrophin A via eEF1A2 is a critical mechanism of regulating utrophin A expression and reveal the potential of repurposed drugs for treating DMD via this pathway.


Asunto(s)
Distrofia Muscular de Duchenne/tratamiento farmacológico , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , Biosíntesis de Proteínas/efectos de los fármacos , Utrofina/genética , Regiones no Traducidas 5'/genética , Animales , Betaxolol/farmacología , Betaxolol/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Humanos , Sitios Internos de Entrada al Ribosoma/genética , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular de Duchenne/genética , Mioblastos , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Pravastatina/farmacología , Pravastatina/uso terapéutico , Biosíntesis de Proteínas/genética , Regulación hacia Arriba/efectos de los fármacos , Utrofina/metabolismo
6.
Bone ; 125: 103-111, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31077852

RESUMEN

PURPOSE: Children with Hutchinson-Gilford progeria syndrome (HGPS), a rare premature aging disease, exhibit extraskeletal calcifications detected by radiographic analysis and on physical examination. The aim of this study was to describe the natural history and pathophysiology of these abnormal calcifications in HGPS, and to determine whether medications and/or supplements tested in clinical trials alter their development. METHODS: Children from two successive clinical trials administering 1) lonafarnib (n = 26) and 2) lonafarnib + pravastatin + zoledronic acid (n = 37) were studied at baseline (pre-therapy), one year on therapy, and at end-of-therapy (3.3-4.3 years after the baseline visit). Calcium supplementation (oral calcium carbonate) was administered during the first year of the second trial and was subsequently discontinued. Information on calcifications was obtained from physical examinations, radiographs, and serum and urinary biochemical measures. The mineral content of two skin-derived calcifications was determined by x-ray diffraction. RESULTS: Extraskeletal calcifications were detected radiographically in 12/39 (31%) patients at baseline. The odds of exhibiting calcifications increased with age (p = 0.045). The odds were unaffected by receipt of lonafarnib, pravastatin, and zoledronate therapies. However, administration of calcium carbonate supplementation, in conjunction with all three therapeutic agents, significantly increased the odds of developing calcifications (p = 0.009), with the odds plateauing after the supplement's discontinuation. Composition analysis of calcinosis cutis showed hydroxyapatite similar to bone. Although serum calcium, phosphorus, and parathyroid hormone (PTH) were within normal limits at baseline and on-therapy, PTH increased significantly after lonafarnib initiation (p < 0.001). Both the urinary calcium/creatinine ratio and tubular reabsorption of phosphate (TRP) were elevated at baseline in 22/39 (56%) and 31/37 (84%) evaluable patients, respectively, with no significant changes while on-therapy. The mean calcium × phosphorus product (Ca × Pi) was within normal limits, but plasma magnesium decreased over both clinical trials. Fibroblast growth factor 23 (FGF23) was lower compared to age-matched controls (p = 0.03). CONCLUSIONS: Extraskeletal calcifications increased with age in children with HGPS and were composed of hydroxyapatite. The urinary calcium/creatinine ratio and TRP were elevated for age while FGF23 was decreased. Magnesium decreased and PTH increased after lonafarnib therapy which may alter the ability to mobilize calcium. These findings demonstrate that children with HGPS with normal renal function and an unremarkable Ca × Pi develop extraskeletal calcifications by an unidentified mechanism that may involve decreased plasma magnesium and FGF23. Calcium carbonate accelerated their development and is, therefore, not recommended for routine supplementation in these children.


Asunto(s)
Calcinosis/patología , Progeria/patología , Calcinosis/sangre , Calcinosis/diagnóstico por imagen , Calcinosis/tratamiento farmacológico , Calcio/sangre , Niño , Preescolar , Creatinina/sangre , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Técnicas In Vitro , Lamina Tipo A/metabolismo , Masculino , Hormona Paratiroidea/sangre , Hormona Paratiroidea/metabolismo , Piperidinas/uso terapéutico , Pravastatina/uso terapéutico , Progeria/sangre , Progeria/diagnóstico por imagen , Progeria/tratamiento farmacológico , Piridinas/uso terapéutico , Ácido Zoledrónico/uso terapéutico
7.
J Hepatol ; 71(3): 516-522, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31125576

RESUMEN

BACKGROUND & AIMS: Sorafenib is the standard of care for advanced hepatocellular carcinoma (HCC). Combining sorafenib with another treatment, to improve overall survival (OS) within an acceptable safety profile, might be the next step forward in the management of patients with advanced HCC. We aimed to assess whether a combination of sorafenib and a statin improved survival in patients with HCC. METHODS: The objective of the PRODIGE-11 trial was to compare the respective clinical outcomes of the sorafenib-pravastatin combination (arm A) versus sorafenib alone (arm B) in patients with advanced HCC. Child-Pugh A patients with advanced HCC who were naive to systemic treatment (n = 323) were randomly assigned to sorafenib-pravastatin combination (n = 162) or sorafenib alone (n = 161). The primary endpoint was OS; secondary endpoints were progression-free survival, time to tumor progression, time to treatment failure, safety, and quality of life. RESULTS: After randomization, 312 patients received at least 1 dose of study treatment. After a median follow-up of 35 months, 269 patients died (arm A: 135; arm B: 134) with no difference in median OS between treatments arms (10.7 months vs. 10.5 months; hazard ratio = 1.00; p = 0.975); no difference was observed in secondary survival endpoints either. In the univariate analysis, the significant prognostic factors for OS were CLIP score, performance status, and quality of life scores. The multivariate analysis showed that the only prognostic factor for OS was the CLIP score. The main toxicity was diarrhea (which was severe in 11% of patients in arm A, and 8.9% in arm B), while severe nausea-vomiting was rare, and no toxicity-related deaths were reported. CONCLUSION: Adding pravastatin to sorafenib did not improve survival in patients with advanced HCC. LAY SUMMARY: Sorafenib has proven efficacy for the treatment of patients with advanced hepatocellular carcinoma. However, overall survival remains poor in these patients, so we were interested to see if the addition of a statin, pravastatin, improved outcomes in patients with advanced HCC. This randomized-controlled trial demonstrated that the sorafenib-pravastatin combination did not improve overall survival in this study population compared to sorafenib alone. Clinical trial number: NCT01075555.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Pravastatina/uso terapéutico , Sorafenib/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma Hepatocelular/mortalidad , Diarrea/inducido químicamente , Femenino , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/mortalidad , Masculino , Persona de Mediana Edad , Pravastatina/efectos adversos , Pronóstico , Supervivencia sin Progresión , Calidad de Vida , Sorafenib/efectos adversos
8.
Pharmacol Res ; 144: 90-98, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30959158

RESUMEN

Hydrogen sulfide gas (H2S) has protective effects in the cardiovascular system that includes preventing the development of atherosclerosis when tested in several in vivo models. Plaque instability is a major risk factor for thromboembolism, myocardial infarction, and stroke, so we examined if H2S can promote plaque stability and the potential underlying mechanisms. Apolipoprotein E knockout mice fed an atherogenic diet were administered the exogenous H2S donor sodium hydrosulfide (NaHS) or pravastatin as a positive control daily for 14 weeks. NaHS significantly enhanced plaque stability by increasing fibrous cap thickness and collagen content compared to vehicle-treated controls. NaHS treatment also reduced blood lipid levels and plaque formation. Preservation of plaque stability by NaHS was associated with reductions in vascular smooth muscle cells (VSMCs) apoptosis and expression of the collagen-degrading enzyme matrix metallopeptidase-9 (MMP-9) in plaque. While pravastatin also increased fibrous cap thickness and reduced VSMC apoptosis, but did not enhance plaque collagen or reduce MMP-9 significantly, suggesting distinct mechanisms of plaque stabilization. in vitro, NaHS also decreased MMP-9 expression in macrophages stimulated with tumor necrosis factor-α by inhibiting ERK/JNK phosphorylation and activator protein 1 nuclear translocation. Moreover, H2S reduced caspase-3/9 activity, Bax/Bcl-2 ratio, and LOX-1 mRNA expression in VSMCs stimulated with oxidized low-density lipoprotein. These results suggest that H2S enhances plaque stability and protects against atherogenesis by increasing plaque collagen content and VSMC count. In conclusion, H2S exerts protective effects against atherogenesis at least partly by stabilizing atherosclerotic plaque.


Asunto(s)
Sulfuro de Hidrógeno/uso terapéutico , Músculo Liso Vascular/efectos de los fármacos , Placa Aterosclerótica/tratamiento farmacológico , Sulfuros/uso terapéutico , Animales , Anticolesterolemiantes/uso terapéutico , Apolipoproteínas E/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/efectos de los fármacos , Placa Aterosclerótica/genética , Pravastatina/uso terapéutico , Ratas Wistar
9.
Placenta ; 75: 1-8, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30712660

RESUMEN

INTRODUCTION: Statins induce heme oxygenase-1 (HO-1) expression in vitro and in vivo. Low HO-1 expression is associated with pregnancy complications, e.g. preeclampsia and recurrent miscarriages. Here, we investigated the effects of pravastatin on HO-1 expression, placental development, and fetal survival in mice with a partial HO-1 deficiency. METHODS: At E14.5, untreated pregnant wild-type (WT, n=13-18), untreated HO-1+/- (Het, n=6-9), and Het mice treated with pravastatin (Het+Pravastatin, n=12-14) were sacrificed. Numbers of viable fetuses/resorbed concepti were recorded. Maternal livers and placentas were harvested for HO activity. Hematoxylin and eosin (H&E) and CD31 immunohistochemical staining were performed on whole placentas. RESULTS: Compared with WT, HO activity in Het livers (65±18%, P<0.001) and placentas (74±7%, P<0.001) were significantly decreased. Number of viable fetuses per dam was significantly lower in Untreated Het dams (6.0±2.2) compared with WT (9.1±1.4, P<0.01), accompanied by a higher relative risk (RR) for concepti resorption (17.1, 95% CI 4.0-73.2). In Hets treated with pravastatin, maternal liver and placental HO activity increased, approaching levels of WT controls (to 83±7% and 87±14%, respectively). The number of viable fetuses per dam increased to 7.7±2.5 with a decreased RR for concepti resorption (2.7, 95% CI 1.2-5.9). In some surviving Untreated Het placentas, there were focal losses of cellular architecture and changes suggestive of reduced blood flow in the labyrinth. These findings were absent in Het+Pravastatin placentas. DISCUSSION: Pravastatin induces maternal liver and placental HO activity, may affect placental function and improve fetal survival in the context of a partial deficiency of HO-1.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Proteínas de la Membrana/metabolismo , Placentación/efectos de los fármacos , Pravastatina/uso terapéutico , Preeclampsia/prevención & control , Animales , Evaluación Preclínica de Medicamentos , Femenino , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hígado/efectos de los fármacos , Hígado/enzimología , Ratones , Pravastatina/farmacología , Embarazo , Distribución Aleatoria
10.
J Cell Physiol ; 234(7): 11047-11059, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30536661

RESUMEN

New onset of diabetes is associated with the use of statins. We have recently demonstrated that pravastatin-treated hypercholesterolemic LDL receptor knockout (LDLr-/- ) mice exhibit reductions in insulin secretion and increased islet cell death and oxidative stress. Here, we hypothesized that these diabetogenic effects of pravastatin could be counteracted by treatment with the antioxidant coenzyme Q 10 (CoQ 10 ), an intermediate generated in the cholesterol synthesis pathway. LDLr -/- mice were treated with pravastatin and/or CoQ 10 for 2 months. Pravastatin treatment resulted in a 75% decrease of liver CoQ 10 content. Dietary CoQ 10 supplementation of pravastatin-treated mice reversed fasting hyperglycemia, improved glucose tolerance (20%) and insulin sensitivity (>2-fold), and fully restored islet glucose-stimulated insulin secretion impaired by pravastatin (40%). Pravastatin had no effect on insulin secretion of wild-type mice. In vitro, insulin-secreting INS1E cells cotreated with CoQ 10 were protected from cell death and oxidative stress induced by pravastatin. Simvastatin and atorvastatin were more potent in inducing dose-dependent INS1E cell death (10-15-fold), which were also attenuated by CoQ 10 cotreatment. Together, these results demonstrate that statins impair ß-cell redox balance, function and viability. However, CoQ 10 supplementation can protect the statins detrimental effects on the endocrine pancreas.


Asunto(s)
Hipercolesterolemia/tratamiento farmacológico , Células Secretoras de Insulina/efectos de los fármacos , Pravastatina/efectos adversos , Receptores de LDL/metabolismo , Ubiquinona/análogos & derivados , Animales , Línea Celular , Supervivencia Celular , Diabetes Mellitus/inducido químicamente , Suplementos Dietéticos , Femenino , Prueba de Tolerancia a la Glucosa , Peróxido de Hidrógeno , Insulina , Hígado/metabolismo , Ratones , Ratones Noqueados , Pravastatina/uso terapéutico , Receptores de LDL/genética , Ubiquinona/farmacología
11.
High Blood Press Cardiovasc Prev ; 25(4): 355-359, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30229462

RESUMEN

INTRODUCTION: Fenofibrate is an effective and safe treatment for hypertriglyceridemia. However, after TG reduction a residual dyslipidemia could appear and require further treatment. AIM: To comparatively evaluate the short-term tolerability and efficacy of a combined lipid-lowering nutraceutical and pravastatin 40 mg in fenofibrate treated patients. METHOD: We prospectively enrolled 40 patients well-tolerating treatment with micronized fenofibrate 145 mg/day and with residual dyslipidemia (LDL-C > 115 mg/dL and TG > 150 mg/dL). Exclusion criteria have been type 2 diabetes, Familial Hypercholesterolemia, previous cardiovascular diseases and severe chronic kidney disease. Then, we have randomly assigned the patients to treatment with pravastatin 40 mg or a combined lipid-lowering nutraceutical (Armolipid Plus®, containing monacolin 3 mg and berberine 500 mg). RESULTS: After 8 weeks of treatment, 80% of pravastatin treated patients (N. 16/20) and 75% of those treated with Armolipid Plus® (N. 15/20) reached the desired LDL-C target, while 50% of pravastatin treated patients (N. 10/20) and 80% of the Armolipid Plus® treated ones reached the desired TG target (N. 16/20). No one adverse event has been registered during Armolipid Plus®, while 1 patient claimed myalgia and 1 reported significant increase of CPK (> 3 ULN) during pravastatin treatment. Both patients were then treated with Armolipid Plus® with resolution of symptoms and CPK increase, respectively. CONCLUSION: In hypertriglyceridemic patients treated with fenofibrate, the association with a combined lipid lowering nutraceutical seem to be more effective in optimizing residual hypertriglyceridemia than pravastatin 40 mg, while being more tolerable and having similar effect on LDL-C plasma level.


Asunto(s)
Suplementos Dietéticos , Fenofibrato/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipertrigliceridemia/tratamiento farmacológico , Pravastatina/uso terapéutico , Triglicéridos/sangre , Biomarcadores/sangre , Suplementos Dietéticos/efectos adversos , Regulación hacia Abajo , Quimioterapia Combinada , Femenino , Fenofibrato/efectos adversos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Hipertrigliceridemia/sangre , Hipertrigliceridemia/diagnóstico , Italia , Masculino , Pravastatina/efectos adversos , Vigilancia de Productos Comercializados , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento
12.
J Reprod Immunol ; 124: 15-20, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29028516

RESUMEN

Pre-eclampsia is a disease of pregnancy affecting 5%-8% of all pregnancies and a leading cause of maternal and fetal mortality. Despite improvements in the diagnosis, there is no effective method for prevention and treatment. While studies in women are of critical importance, investigation of pathological mechanisms in pregnant women is necessarily limited, and the ability to establish cause and effect relationships, difficult. Mouse models have been instrumental in defining pathogenic mechanisms in preeclampsia and in the identification of pravastatin as a potential treatment to prevent pregnancy complications associated with placental dysfunction. Numerous epidemiological studies provided robust evidence demonstrating that pravastatin exposure during pregnancy does not affect fetal development. In addition, pravastatin is hydrophilic and has a limited passage through the placenta, diminishing any safety concerns. Several pilot studies suggest that pravastatin may be a good option to prevent and treat preeclampsia in women. While these studies are promising, the effectiveness of pravastatin to treat preeclampsia needs to be confirmed by randomized clinical trials.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Placenta/fisiología , Pravastatina/uso terapéutico , Preeclampsia/tratamiento farmacológico , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Desarrollo Fetal , Humanos , Ratones , Placenta/efectos de los fármacos , Pravastatina/farmacología , Embarazo
13.
FASEB J ; 31(8): 3636-3648, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28465323

RESUMEN

Rheumatoid arthritis is an inflammatory condition characterized by overzealous inflammation that leads to joint damage and is associated with an increased incidence of cardiovascular disease. Statins are frontline therapeutics for patients with cardiovascular disease and exert beneficial actions in rheumatoid arthritis. The mechanism that mediates the beneficial actions of statins in rheumatoid arthritis remains of interest. In the present study, we found that the administration of 2 clinically relevant statins-atorvastatin (0.2 mg/kg) or pravastatin (0.2 mg/kg)-to mice during inflammatory arthritis up-regulated systemic and tissue amounts of a novel family of proresolving mediators, termed 13-series resolvins (RvTs), and significantly reduced joint disease. Of note, administration of simvastatin (0.2 mg/kg) did not significantly up-regulate RvTs or reduce joint inflammation. We also found that atorvastatin and pravastatin each reduced systemic leukocyte activation, including platelet-monocyte aggregates (∼25-60%). These statins decreased neutrophil trafficking to the joint as well as joint monocyte and macrophage numbers. Atorvastatin and pravastatin produced significant reductions (∼30-50%) in expression of CD11b and major histocompatibility complex class II on both monocytes and monocyte-derived macrophages in joints. Administration of an inhibitor to cyclooxygenase-2, the initiating enzyme in the RvT pathway, reversed the protective actions of these statins on both joint and systemic inflammation. Together, these findings provide evidence for the role of RvTs in mediating the protective actions of atorvastatin and pravastatin in reducing local and vascular inflammation, and suggest that RvTs may be useful in measuring the anti-inflammatory actions of statins.-Walker, M. E., Souza, P. R., Colas, R. A., Dalli, J. 13-Series resolvins mediate the leukocyte-platelet actions of atorvastatin and pravastatin in inflammatory arthritis.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Artritis/tratamiento farmacológico , Atorvastatina/uso terapéutico , Ácidos Docosahexaenoicos/metabolismo , Inflamación/tratamiento farmacológico , Pravastatina/uso terapéutico , Animales , Artritis/inducido químicamente , Artritis/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
14.
J Natl Cancer Inst ; 109(5)2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28040693

RESUMEN

Background: Recent studies have suggested associations between statins and enhanced survival among patients with pancreatic ductal adenocarcinoma (PDAC). However, the relationship between statins, cholesterol, and survival remains unclear. Methods: We conducted a retrospective cohort study on 2142 PDAC patients in a regional integrated healthcare system from 2006 to 2014. Electronic pharmacy records were used to abstract information on the type, length, and dosage of statin exposures starting in the year prior to diagnosis. The cumulative and individual effects of simvastatin, lovastatin, atorvastatin, pravastatin, and rosuvastatin on mortality were assessed using Cox proportional hazards regression. Statins were evaluated as any use (pre- and postdiagnosis as a time-dependent variable) and baseline use (prediagnosis only). We also evaluated whether low-density lipoprotein (LDL) cholesterol, measured at various time windows prior to diagnosis, had an independent influence on survival. Additional analyses were performed to examine whether cholesterol mediated the relationship between statins and mortality. All models included age, race, stage, surgery, gemcitabine-based chemotherapy, and the Charlson comorbidity index as covariates. Results: Any (hazard ratio [HR] = 0.87, 95% confidence interval [CI] = 0.79 to 0.97) and baseline (HR = 0.88, 95% CI = 0.79 to 0.98) statin use were both associated with a decreased risk in mortality. When assessing individual statins, we found reduced mortality among simvastatin (HR = 0.87, 95% CI = 0.77 to 0.98) and atorvastatin (HR = 0.58, 95% CI = 0.46 to 0.72) users. Cholesterol was not associated with mortality and did not mediate any relationships between statins and survival. Conclusions: Statin use rather than cholesterol level was associated with lower mortality risk in patients with pancreatic cancer. Statins appear to improve survival through a lipid-independent mechanism.


Asunto(s)
Carcinoma Ductal Pancreático/mortalidad , LDL-Colesterol/sangre , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Neoplasias Pancreáticas/mortalidad , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Atorvastatina/uso terapéutico , California/epidemiología , Carcinoma Ductal Pancreático/secundario , Carcinoma Ductal Pancreático/terapia , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Prescripciones de Medicamentos , Femenino , Humanos , Lovastatina/uso terapéutico , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Pravastatina/uso terapéutico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Rosuvastatina Cálcica/uso terapéutico , Simvastatina/uso terapéutico , Tasa de Supervivencia , Gemcitabina
15.
Drugs Aging ; 32(12): 1055-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26625880

RESUMEN

BACKGROUND: Improvement in renal function and decreases in serum uric acid (SUA) have been reported following prolonged high-intensity statin (HMG-CoA reductase inhibitor) therapy. This post hoc analysis of the SAGE trial examined the effect of intensive versus less intensive statin therapy on renal function, safety, and laboratory parameters, including SUA, in elderly coronary artery disease (CAD) patients (65-85 years) with or without chronic kidney disease (CKD). METHODS: Patients were randomized to atorvastatin 80 mg/day or pravastatin 40 mg/day and treated for 12 months. Patients were stratified using Modification of Diet in Renal Disease (MDRD) estimated glomerular filtration rates (eGFRs) in CKD (eGFR <60 mL/min/1.73 m(2)) and non-CKD populations. RESULTS: Of the 893 patients randomized, 858 had complete renal data and 418 of 858 (49%) had CKD (99% Stage 3). Over 12 months, eGFR increased with atorvastatin and remained stable with pravastatin (+2.38 vs. +0.18 mL/min/1.73 m(2), respectively; p < 0.0001). MDRD eGFR improved significantly in both CKD treatment arms; however, the increased eGFR in patients without CKD was significantly greater with atorvastatin (+2.08 mL/min/1.73 m(2)) than with pravastatin (-1.04 mL/min/1.73 m(2)). Modest reductions in SUA were observed in both treatment arms, but a greater fall occurred with atorvastatin than with pravastatin (-0.52 vs. -0.09 mg/dL, p < 0.0001). Change in SUA correlated negatively with changes in eGFR and positively with changes in low-density lipoprotein cholesterol. Reports of myalgia were rare (3.6% CKD; 5.7% non-CKD), and there were no episodes of rhabdomyolysis. Elevated serum alanine and aspartate transaminase to >3 times the upper limit of normal occurred in 4.4% of atorvastatin- and 0.2% of pravastatin-treated patients. CONCLUSION: Intensive management of dyslipidemia in older patients with stable coronary heart disease may have beneficial effects on renal function and SUA.


Asunto(s)
Dislipidemias/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Ácido Úrico/sangre , Anciano , Anciano de 80 o más Años , Atorvastatina/uso terapéutico , LDL-Colesterol/sangre , Enfermedad Coronaria/tratamiento farmacológico , Método Doble Ciego , Femenino , Tasa de Filtración Glomerular , Humanos , Riñón/efectos de los fármacos , Masculino , Pravastatina/uso terapéutico , Insuficiencia Renal Crónica/fisiopatología
16.
Curr Opin Pediatr ; 27(2): 227-32, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25689458

RESUMEN

PURPOSE OF REVIEW: All polycystic diseases of the kidney exhibit tubular or saccular cysts. The cysts can either be open to the tubule or isolated sacs that have lost their connections. Polycystic kidney diseases derived from different genetic mutations share basic mechanisms of cytogenesis, formation, and progressive enlargement, involving a cellular organelle called the primary cilium. Given the mechanistic commonalities, this review will focus on the therapeutic approaches currently available or under development that likely apply to all inherited renal cystic diseases. RECENT FINDINGS: Recent advances in clinical trials and preclinical experiments have illuminated common signaling pathway involvement. SUMMARY: Avoidance of nephrotoxic drugs or radio-contrast and maintaining normal BMI are routine preventive measures. Limiting the intake of calories, salt, and protein, together with increased intake of fruits, vegetables, and water are dietary treatments that should be started early in the course of the disease. Potential pharmacological treatments targeting cyst initiation and progression are on the horizon.


Asunto(s)
Dieta Hiposódica , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Enfermedades Renales Poliquísticas/terapia , Pravastatina/uso terapéutico , Sistema Renina-Angiotensina/efectos de los fármacos , Consejo Dirigido , Progresión de la Enfermedad , Ejercicio Físico , Frutas , Humanos , Enfermedades Renales Poliquísticas/fisiopatología , Pronóstico , Conducta de Reducción del Riesgo , Verduras
17.
Placenta ; 35(9): 724-31, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25043692

RESUMEN

INTRODUCTION: Preeclampsia is a serious pregnancy complication. Soluble endoglin (sEng) is released from the placenta and contributes to the maternal endothelial dysfunction seen in preeclampsia. Recently oxysterols, which activate the Liver X Receptor (LXR), have been implicated in producing sEng, by upregulating matrix metalloproteinase-14 (MMP14; cleaves endoglin to produce sEng) and down-regulating tissue inhibitor of metalloproteinase-3 (TIMP-3; inhibitor of MMP14). The functional experiments in that study were performed on JAR cells (human choriocarcinoma cell line) and placental explants. METHODS: We characterized LXR in severe preeclamptic placentas, and assessed whether oxysterols increase release of sEng from primary human umbilical vein endothelial cells (HUVECs), primary trophoblasts and placental explants. Given pravastatin is thought to block oxysterol production and inhibit the LXR, we examined whether pravastatin reduces sEng release. RESULTS: LXRα and ß were localized to the syncytiotrophoblast and villous tips and were significantly up-regulated in preeclamptic placenta. Oxysterols upregulated sEng production in HUVECs and placental explants although the increases were far more modest than that recently reported. Oxysterols did not upregulate sEng in primary trophoblasts. Furthermore, mRNA expression of MMP14 and TIMP-3 were not altered by oxysterols in any tissue. Surprisingly, pravastatin did not decrease oxysterol-induced upregulation of sEng. DISCUSSION: LXR is up-regulated in preeclamptic placenta. Oxysterols upregulate sEng production from human tissues, but the increase is modest, suggesting this may not be the main mechanism for the very significant elevations in sEng seen in preeclampsia. Pravastatin does not decrease sEng production. CONCLUSION: Oxysterols modestly up-regulate sEng production which is not quenched by pravastatin.


Asunto(s)
Antígenos CD/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Receptores Nucleares Huérfanos/metabolismo , Placenta/efectos de los fármacos , Pravastatina/farmacología , Receptores de Superficie Celular/metabolismo , Adulto , Estudios de Casos y Controles , Evaluación Preclínica de Medicamentos , Endoglina , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Técnicas In Vitro , Receptores X del Hígado , Placenta/metabolismo , Pravastatina/uso terapéutico , Preeclampsia/tratamiento farmacológico , Embarazo , Esteroles/metabolismo , Regulación hacia Arriba , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
J Clin Lipidol ; 8(1): 61-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24528686

RESUMEN

BACKGROUND: Primary cardiovascular prevention may be achieved by lifestyle/nutrition improvements and specific drugs, although a relevant role is now emerging for specific functional foods and nutraceuticals. OBJECTIVES: The aim of this study was to evaluate the usefulness of a nutraceutical multitarget approach in subjects with moderate cardiovascular risk and to compare it with pravastatin treatment. SUBJECTS: Thirty patients with moderate dyslipidemia and metabolic syndrome (according to the Third Report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults) were included in an 8-week randomized, double-blind crossover study and took either placebo or a nutraceutical combination that contained red yeast rice extract, berberine, policosanol, astaxanthin, coenzyme Q10, and folic acid (Armolipid Plus). Subsequently, they were subjected to another 8-week treatment with pravastatin 10 mg/d. This dosage was selected on the basis of its expected -20% efficacy in reducing low-density lipoprotein-cholesterol. RESULTS: Treatment with Armolipid Plus led to a significant reduction of total cholesterol (-12.8%) and low-density lipoprotein-cholesterol (-21.1%), similar to pravastatin (-16% and -22.6%, respectively), and an increase of high-density lipoprotein-cholesterol (4.8%). Armolipid Plus improved the leptin-to-adiponectin ratio, whereas adiponectin levels were unchanged. CONCLUSIONS: These results indicate that this nutraceutical approach shows a lipid-lowering activity comparable to pravastatin treatment. Hence, it may be a safe and useful option, especially in conditions of moderate cardiovascular risk, in which a pharmacologic intervention may not be appropriate.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Suplementos Dietéticos , Berberina/uso terapéutico , Productos Biológicos/uso terapéutico , Biomarcadores/sangre , LDL-Colesterol/sangre , Estudios Cruzados , Método Doble Ciego , Determinación de Punto Final , Alcoholes Grasos/uso terapéutico , Femenino , Ácido Fólico/uso terapéutico , Humanos , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , Pravastatina/uso terapéutico , Factores de Tiempo , Ubiquinona/análogos & derivados , Ubiquinona/uso terapéutico , Xantófilas/uso terapéutico
19.
J Orthop Res ; 32(4): 557-65, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24302463

RESUMEN

The objective of the study is to understand the therapeutic effects of lipophilic (simvastatin) and hydrophilic statins (pravastatin) combined with/without hyaluronic acid for osteoarthritis by an in vitro LPS-induced inflammatory model of articular chondrocytes. HA in combination with different doses of simvastatin or pravastatin were used. Beside cytotoxicity, the influence of statins on NO production, pro-inflammatory cytokine, inflammatory mediators, and NF-κB p50 protein were analyzed. Finally, TUNEL assay was performed to detect DNA strand breakage. Two statins were less able to lower NF-κB activity when they were administrated along without HA. The gene expression demonstrates that simvastatin and pravastatin had the ability to decrease pro-inflammatory and inflammatory mediator levels. High dose simvastatin with or without HA down regulated inflammatory cytokines, but resulted in higher cytotoxicity. TUNEL assay confirms the regulatory effect of statins with or without HA over the apoptosis of chondrocytes, especially in hydrophilic statins. The significant down-regulation of inflammatory mediators suggests that intra-articular injection of HA in combination with statins might feasibly slow the progress of osteoarthritis. Administration of simvastatin or pravastatin with hyaluronic acid may produce beneficial effects for OA treatment, but with better results when hydrophilic statin was used.


Asunto(s)
Condrocitos/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Osteoartritis/tratamiento farmacológico , Pravastatina/uso terapéutico , Simvastatina/uso terapéutico , Animales , Condrocitos/metabolismo , Colágeno Tipo II/metabolismo , Ácido Hialurónico/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Etiquetado Corte-Fin in Situ , Inyecciones Intraarticulares , Lipopolisacáridos/toxicidad , Subunidad p50 de NF-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Osteoartritis/patología , Pravastatina/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Simvastatina/farmacología , Porcinos , Viscosuplementos/uso terapéutico
20.
Pharmacoeconomics ; 31(6): 519-31, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23585310

RESUMEN

BACKGROUND: Results from the PROVE IT trial suggest that patients with acute coronary syndrome (ACS) treated with atorvastatin 80 mg/day (A80) have significantly lower rates of cardiovascular events compared with patients treated with pravastatin 40 mg/day (P40). In a genetic post hoc substudy of the PROVE IT trial, the rate of event reduction was greater in carriers of the Trp719Arg variant in kinesin family member 6 protein (KIF6) than in noncarriers. We assessed the cost effectiveness of testing for the KIF6 variant followed by targeted statin therapy (KIF6 Testing) versus not testing patients (No Test) and treating them with P40 or A80 in the USA from a payer perspective. METHODS: A Markov model was developed in which 2-year event rates from PROVE IT were extrapolated over a lifetime horizon. Costs and utilities were derived from published literature. All costs were in 2010 US dollars except the cost of A80, which was in 2012 US dollars because the generic formulation was available in 2012. Expected costs and quality-adjusted life-years (QALYs) were estimated for each strategy over a lifetime horizon. RESULTS: Lifetime costs were US$31,700; US$37,100 and US$41,300 for No Test P40, KIF6 Testing and No Test A80 strategies, respectively. The No Test A80 strategy was associated with more QALYs (9.71) than the KIF6 Testing (9.69) and No Test P40 (9.57) strategies. No Test A80 had an incremental cost-effectiveness ratio (ICER) of US$232,100 per QALY gained compared with KIF6 Testing. KIF6 Testing had an ICER of US$45,300 per QALY compared with No Test P40. CONCLUSIONS: Testing ACS patients for KIF6 carrier status may be a cost-effective strategy at commonly accepted thresholds. Treating all patients with A80 is more expensive than treating patients on the basis of KIF6 results, but the modest gain in QALYs is achieved at a cost/QALY that is generally considered unacceptable compared with the KIF6 Testing strategy. Compared with treating all patients with P40, the KIF6 Testing strategy had an ICER below US$50,000 per QALY. The conclusions from this study are sensitive to the price of generic A80 and the effect on adherence of knowing KIF6 carrier status. The results were based on a post hoc substudy of the PROVE IT trial, which was not designed to test the effectiveness of KIF6 testing.


Asunto(s)
Síndrome Coronario Agudo/tratamiento farmacológico , Ácidos Heptanoicos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Cinesinas/genética , Pirroles/uso terapéutico , Síndrome Coronario Agudo/economía , Síndrome Coronario Agudo/genética , Atorvastatina , Análisis Costo-Beneficio , Relación Dosis-Respuesta a Droga , Femenino , Pruebas Genéticas/economía , Pruebas Genéticas/métodos , Genotipo , Ácidos Heptanoicos/economía , Ácidos Heptanoicos/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/economía , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Masculino , Cadenas de Markov , Persona de Mediana Edad , Terapia Molecular Dirigida , Pravastatina/administración & dosificación , Pravastatina/economía , Pravastatina/uso terapéutico , Pirroles/economía , Pirroles/farmacología , Años de Vida Ajustados por Calidad de Vida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA