Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Drugs ; 80(14): 1465-1475, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32910441

RESUMEN

Secondary cardiovascular (CV) prevention in patients with vascular disease [e.g. coronary (CAD) and peripheral (PAD) artery disease] is crucial and typically involves antiplatelet therapy with aspirin; however, managing residual ischaemic and bleeding risks in CV disease (CVD) remains a challenge. Combining the oral anticoagulant rivaroxaban (Xarelto®) with aspirin targets both the platelet and thrombotic processes of atherosclerosis, a common pathophysiological process associated with CVD. In the global COMPASS trial (n > 27,000), rivaroxaban 2.5 mg twice daily plus aspirin 100 mg once daily (vs aspirin alone) significantly reduced the risk of the primary composite major adverse CV event (MACE) outcome (i.e. myocardial infarction, stroke or CV death) in adults with stable CAD and/or PAD and, in those with PAD, significantly reduced the risk of the composite major adverse limb event (MALE) outcome. Rivaroxaban + aspirin treatment was generally well tolerated; however, the risk of the composite major bleeding outcome, but not intracranial or fatal bleeding, was significantly higher with rivaroxaban + aspirin than aspirin. The increased risk for the composite major bleeding outcome did not negate the composite net clinical benefits of rivaroxaban + aspirin for secondary CV prevention, with rivaroxaban + aspirin especially beneficial in those with a greater CV risk at baseline. Ongoing clinical experience is required to fully define the role of rivaroxaban + aspirin in secondary CV prevention. In the meantime, dual therapy with rivaroxaban + aspirin is an important emerging option for secondary CV prevention of atherothrombotic events in adults with CAD or symptomatic PAD who are at high risk of ischaemic events.


Asunto(s)
Anticoagulantes/uso terapéutico , Enfermedades Cardiovasculares/prevención & control , Rivaroxabán/uso terapéutico , Administración Oral , Anticoagulantes/administración & dosificación , Aspirina/administración & dosificación , Aspirina/uso terapéutico , Quimioterapia Combinada , Humanos , Rivaroxabán/administración & dosificación , Prevención Secundaria
2.
Drugs ; 79(6): 665-674, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30993651

RESUMEN

Lenvatinib (Lenvima®) is an oral small molecule inhibitor of multiple receptor tyrosine kinases, and is approved for the first-line treatment of patients with unresectable hepatocellular carcinoma (HCC) in the USA, EU, Japan and China. The approval of lenvatinib was based on results of the randomized, open-label, multinational, non-inferiority phase III REFLECT trial in patients with unresectable HCC, who had not received treatment for advanced disease. In REFLECT, lenvatinib was non-inferior, but not superior, to sorafenib (current standard of care) for overall survival (OS). However, lenvatinib was associated with significant improvements compared with sorafenib in terms of all secondary endpoints [higher objective response rate (ORR), and longer progression-free survival (PFS) and time to progression (TTP)]. Lenvatinib had a generally manageable tolerability profile in REFLECT, with the most common treatment-emergent adverse events being hypertension, diarrhoea, decreased appetite and decreased weight. Given its non-inferior efficacy to sorafenib and manageable tolerability profile, lenvatinib represents a long-awaited alternative option to sorafenib for the first-line systemic treatment of patients with unresectable HCC. Further clinical experience may be required to fully define the position of lenvatinib in this setting.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Compuestos de Fenilurea/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinolinas/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/administración & dosificación , Quinolinas/efectos adversos , Quinolinas/farmacología , Sorafenib/farmacología , Sorafenib/uso terapéutico , Resultado del Tratamiento , Tirosina/antagonistas & inhibidores
3.
Drugs ; 78(4): 479-493, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29532438

RESUMEN

Intravenous ferric carboxymaltose (Ferinject®; Injectafer®) is a colloidal solution of nanoparticles which consist of a polynuclear iron (III)-(oxyhydr)oxide core stabilized by carboxymaltose and may be given as a single high-dose, 15-min infusion. This article reviews the clinical use of ferric carboxymaltose in various patient populations with iron deficiency (ID) [± anaemia] and briefly summarizes its pharmacological properties. Based on extensive experience in the clinical trial and real-world settings, ferric carboxymaltose is an effective and generally well tolerated treatment for rapidly replenishing iron stores and correcting anaemia in patients with ID (± anaemia) of various aetiologies, including patients with chronic heart failure (CHF), chronic kidney disease, inflammatory bowel disease or perioperative anaemia, and women with ID during pregnancy, postpartum or associated with heavy uterine bleeding. As it may be given as a single high-dose infusion, ferric carboxymaltose has the potential to provide cost savings from a healthpayer perspective. Thus, ferric carboxymaltose remains an important option for the treatment of ID in adults and, where approved, children aged ≥ 14 years, when oral iron preparations are ineffective or cannot be used.


Asunto(s)
Anemia Ferropénica/tratamiento farmacológico , Compuestos Férricos/uso terapéutico , Maltosa/análogos & derivados , Administración Intravenosa , Anemia Ferropénica/sangre , Anemia Ferropénica/complicaciones , Enfermedad Crónica , Femenino , Compuestos Férricos/administración & dosificación , Compuestos Férricos/efectos adversos , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/complicaciones , Humanos , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Renales/sangre , Enfermedades Renales/complicaciones , Maltosa/administración & dosificación , Maltosa/efectos adversos , Maltosa/uso terapéutico , Neoplasias/sangre , Neoplasias/complicaciones , Periodo Posparto , Embarazo , Resultado del Tratamiento , Hemorragia Uterina/tratamiento farmacológico
4.
Drugs ; 75(16): 1891-902, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26464319

RESUMEN

Oritavancin (Orbactiv(®)) is a new generation lipoglycopeptide approved for use in adult patients with acute bacterial skin and skin structure infections (ABSSSI). It is administered as a single 1200 mg intravenous infusion over 3 h. In phase 3 trials in adult patients with ABSSSI, oritavancin was noninferior to vancomycin in terms of a composite outcome (cessation of spreading or reduction in the size of the baseline lesion, absence of fever and no rescue antibacterials required; primary endpoint) assessed at an US FDA-recommended early timepoint of 48-72 h after initiation of treatment. Oritavancin was also noninferior to vancomycin in terms of a ≥20 % reduction in the baseline lesion size at the early timepoint and clinical cure rate 7-14 days after the end of treatment. Oritavancin was generally well tolerated in the phase 3 trials, with most treatment-emergent adverse reactions being mild in severity. The most common adverse events occurring in oritavancin recipients were nausea, headache, vomiting, limb and subcutaneous abscesses, and diarrhoea. Oritavancin offers a number of potential advantages, including a convenient single dose treatment that may shorten or eliminate hospital stays, a reduction in healthcare resource utilization and cost, no need for dosage adjustment in patients with mild to moderate hepatic or renal impairment, no need for therapeutic drug monitoring, and elimination of compliance concerns. Therefore, oritavancin is a useful treatment option for adults with ABSSSI.


Asunto(s)
Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Enfermedades Cutáneas Bacterianas/tratamiento farmacológico , Antibacterianos/efectos adversos , Antibacterianos/farmacocinética , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Glicopéptidos/efectos adversos , Glicopéptidos/farmacocinética , Humanos , Lipoglucopéptidos , Pruebas de Sensibilidad Microbiana
5.
Drugs ; 74(5): 563-78, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24671907

RESUMEN

Delta-9-tetrahydrocannabinol (THC)/cannabidiol (CBD) [Sativex®] is an oromucosal spray formulation that contains principally THC and CBD at an approximately 1:1 fixed ratio, derived from cloned Cannabis sativa L. plants. The main active substance, THC, acts as a partial agonist at human cannabinoid receptors (CB1 and CB2), and thus, may modulate the effects of excitatory (glutamate) and inhibitory (gamma-aminobutyric acid) neurotransmitters. THC/CBD is approved in a number of countries, including Germany and the UK, as an add-on treatment for symptom improvement in adult patients with moderate to severe spasticity due to multiple sclerosis who have not responded adequately to other anti-spasticity medication and who demonstrate clinically significant improvement in spasticity-related symptoms during an initial trial of therapy. In the largest multinational clinical trial that evaluated the approved THC/CBD regimen in this population, 12 weeks' double-blind treatment with THC/CBD significantly reduced spasticity severity (primary endpoint) compared with placebo in patients who achieved a clinically significant improvement in spasticity after 4 weeks' single-blind THC/CBD treatment, as assessed by a patient-rated numerical rating scale. A significantly greater proportion of THC/CBD than placebo recipients achieved a ≥ 30% reduction (a clinically relevant reduction) in spasticity severity. The efficacy of THC/CBD has been also shown in at least one everyday clinical practice study (MOVE 2). THC/CBD was generally well tolerated in clinical trials. Dizziness and fatigue were reported most frequently during the first 4 weeks of treatment and resolved within a few days even with continued treatment. Thus, add-on THC/CBD is a useful symptomatic treatment option for its approved indication.


Asunto(s)
Esclerosis Múltiple/tratamiento farmacológico , Espasticidad Muscular/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Adulto , Cannabidiol , Agonistas de Receptores de Cannabinoides/efectos adversos , Agonistas de Receptores de Cannabinoides/farmacología , Agonistas de Receptores de Cannabinoides/uso terapéutico , Dronabinol , Combinación de Medicamentos , Humanos , Esclerosis Múltiple/fisiopatología , Espasticidad Muscular/etiología , Extractos Vegetales/efectos adversos , Extractos Vegetales/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB2/agonistas , Índice de Severidad de la Enfermedad
6.
Drugs ; 73(15): 1733-47, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24136090

RESUMEN

Oral fidaxomicin (Dificid(®); Dificlir(®)) is a first-in-class macrocyclic antibacterial that is approved in several countries for the treatment of adult patients with Clostridium difficile-associated diarrhoea. Fidaxomicin 200 mg twice daily for 10 days was an effective and generally well tolerated treatment in adult patients with a first episode or first recurrence of C. difficile infection. In two multinational phase III trials, fidaxomicin treatment was noninferior to vancomycin treatment with regard to clinical cure rates and was associated with statistically significantly lower C. difficile infection recurrence rates and statistically significantly higher global cure rates than vancomycin. The drug has a favourable pharmacological profile, including having a narrow spectrum of activity that targets relevant pathogens, minimal impact on normal faecal microflora, a convenient treatment regimen and attainment of very high faecal concentrations. Albeit further clinical experience is required to fully define the position of fidaxomicin, it is a valuable emerging option for the treatment of first episode and recurrent episodes of C. difficile-associated diarrhoea.


Asunto(s)
Aminoglicósidos/uso terapéutico , Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Enterocolitis Seudomembranosa/tratamiento farmacológico , Aminoglicósidos/administración & dosificación , Aminoglicósidos/efectos adversos , Aminoglicósidos/farmacocinética , Animales , Antibacterianos/administración & dosificación , Antibacterianos/efectos adversos , Antibacterianos/farmacocinética , Ensayos Clínicos Fase III como Asunto , Clostridioides difficile/aislamiento & purificación , Farmacorresistencia Bacteriana Múltiple , Enterocolitis Seudomembranosa/microbiología , Fidaxomicina , Humanos , Pruebas de Sensibilidad Microbiana , Resultado del Tratamiento
7.
CNS Drugs ; 25(12): 1073-85, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22133328

RESUMEN

Oral tetrabenazine is currently the only drug approved by the US FDA for the treatment of chorea associated with Huntington's disease (HD). Although the precise antichorea mechanism of action is unknown, it most likely involves reversible depletion of monoamines, particularly dopamine, from presynaptic terminals via inhibition of human vesicular monoamine transporter type 2. In a 12-week, double-blind, placebo-controlled trial conducted in the US in patients with HD, oral tetrabenazine (≤100 mg/day; n = 54) was significantly (p = 0.0001) more efficacious than placebo (n = 30) at improving adjusted mean Unified HD Rating Scale (UHDRS) total maximum chorea scores (reduced from baseline by 5 vs 1.5) [primary endpoint]. After 12 weeks, improvements in UHDRS total maximum chorea scores of >3 were achieved by significantly (p < 0.0001) more patients in the tetrabenazine group than in the placebo group. The antichorea efficacy of tetrabenazine was maintained in an 80-week extension study (n = 75), with the adjusted mean UHDRS total maximum chorea score significantly (p < 0.001) reduced from baseline (score of 14.9) by 4.6 points (primary outcome). In the 12-week trial and 80-week extension study, treatment-emergent adverse events in the tetrabenazine group mainly occurred during the dosage-titration phase, a period during which the dosage was individually optimized. Most of these events were mild to moderate and were manageable with dosage adjustments or discontinuation of study drug.


Asunto(s)
Corea/tratamiento farmacológico , Enfermedad de Huntington/tratamiento farmacológico , Tetrabenazina/farmacología , Tetrabenazina/uso terapéutico , Animales , Corea/metabolismo , Ensayos Clínicos como Asunto , Dopamina/metabolismo , Método Doble Ciego , Evaluación Preclínica de Medicamentos , Humanos , Enfermedad de Huntington/metabolismo , Estudios Multicéntricos como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas de Transporte Vesicular de Monoaminas/antagonistas & inhibidores , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
8.
Drugs ; 70(5): 631-42, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20329808

RESUMEN

Raltegravir, a first-in-class, oral HIV type-1 (HIV-1) integrase inhibitor, blocks the covalent integration of HIV-1 complementary DNA into the host genome. In a large, randomized, double-blind, multinational, ongoing trial in treatment-naive patients with HIV-1 infection, raltegravir 400 mg twice daily was noninferior to efavirenz 600 mg once daily in achieving plasma HIV-1 RNA viral levels of <50 copies/mL after 48 weeks' treatment (primary endpoint), when used as part of an antiretroviral therapy (ART) combination regimen. The time to achieve a virological response was significantly shorter in the raltegravir group than in the efavirenz group. Furthermore, significantly more raltegravir than efavirenz recipients achieved HIV-1 RNA viral levels of <50 copies/mL at weeks 2-16. The efficacy of raltegravir in achieving HIV-1 RNA levels of <50 copies/mL was also demonstrated in subgroups of patients separated according to baseline viral levels, CD4+ cell counts and viral subtypes. Preliminary evidence suggests that both virological and CD4+ cell count improvements were maintained at 96 weeks for treatment-naive recipients of raltegravir 400 mg twice daily. As part of an ART combination regimen, raltegravir treatment was generally well tolerated and was associated with significantly fewer drug-related adverse events than efavirenz treatment. Moreover, after 96 weeks, raltegravir treatment was associated with a significantly lower impact on serum lipid levels than efavirenz treatment.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Inhibidores de Integrasa VIH/uso terapéutico , VIH-1/patogenicidad , Pirrolidinonas/uso terapéutico , Administración Oral , Recuento de Linfocito CD4 , Quimioterapia Combinada , Medicina Basada en la Evidencia , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Inhibidores de Integrasa VIH/administración & dosificación , Inhibidores de Integrasa VIH/efectos adversos , Inhibidores de Integrasa VIH/farmacocinética , VIH-1/genética , Humanos , Pirrolidinonas/administración & dosificación , Pirrolidinonas/efectos adversos , Pirrolidinonas/farmacocinética , ARN Viral/sangre , Raltegravir Potásico , Factores de Tiempo , Resultado del Tratamiento , Carga Viral
9.
Drugs ; 70(1): 83-97, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20030427

RESUMEN

Besifloxacin is a novel fluoroquinolone that, like other fluoroquinolones, acts by inhibiting the essential bacterial enzymes DNA gyrase and topoisomerase IV. Topical besifloxacin ophthalmic suspension 0.6% is indicated for use in patients with bacterial conjunctivitis caused by susceptible bacteria. Besifloxacin had in vitro activity against a broad spectrum of Gram-positive and -negative bacteria that commonly cause ocular infections (e.g. Haemophilus influenzae, Staphylococcus aureus, S. epidermidis and Streptococcus pneumoniae), including drug-resistant strains. In two randomized, double-blind, multicentre trials, besifloxacin ophthalmic suspension 0.6% administered at the recommended dose for 5 days in patients aged > or =1 year with bacterial conjunctivitis was significantly (p < 0.01) more effective than vehicle in terms of clinical resolution and microbial eradication rates (coprimary endpoints) at study visit two (day 5+/-1) or three (day 8 or 9) [primary timepoints]. Besifloxacin ophthalmic suspension 0.6% was noninferior to moxifloxacin ophthalmic solution 0.5% in patients aged > or =1 year with bacterial conjunctivitis with regard to clinical resolution (58.3% vs 59.4%) and microbial eradication (93.3% vs 91.1%) rates on day 5 +/- 1 of treatment (coprimary endpoints) in a randomized, double-blind, multicentre trial; both drugs were administered at a dosage of one drop in the affected eye(s) three times daily for 5 days. Besifloxacin ophthalmic suspension 0.6% was generally well tolerated in clinical trials, with most adverse events being mild in severity. The tolerability profile of besifloxacin ophthalmic suspension 0.6% was similar to that of moxifloxacin ophthalmic solution 0.5%.


Asunto(s)
Azepinas/uso terapéutico , Fluoroquinolonas/uso terapéutico , Soluciones Oftálmicas/uso terapéutico , Administración Tópica , Antibacterianos/farmacología , Antifúngicos/farmacología , Compuestos Aza/farmacología , Compuestos Aza/uso terapéutico , Recuento de Colonia Microbiana , Conjuntivitis Bacteriana/tratamiento farmacológico , Girasa de ADN/metabolismo , Topoisomerasa de ADN IV/metabolismo , Método Doble Ciego , Esquema de Medicación , Ojo , Humanos , Resistencia a la Meticilina , Staphylococcus aureus Resistente a Meticilina , Pruebas de Sensibilidad Microbiana , Moxifloxacino , Quinolinas/farmacología , Quinolinas/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Streptococcus pneumoniae/efectos de los fármacos , Suspensiones/uso terapéutico
10.
Drugs ; 69(1): 123-36, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19192941

RESUMEN

Oral sodium picosulfate/magnesium citrate (CitraFleet; Picolax), consisting of sodium picosulfate (a stimulant laxative) and magnesium citrate (an osmotic laxative), is approved for use in adults (CitraFleet; Picolax) and/or adolescents and children (Picolax) as a colorectal cleansing agent prior to any diagnostic procedure (e.g. colonoscopy or x-ray examination) requiring a clean bowel and/or surgery. It is dispensed in powder form (sodium picosulfate 0.01 g, magnesium oxide 3.5 g, citric acid 12.0 g per sachet), with the magnesium oxide and citric acid components forming magnesium citrate when the powder is dissolved in water. In adult patients, two sachets of sodium picosulfate/magnesium citrate was at least as effective and well tolerated as oral magnesium citrate 17.7 or 35.4 g, or oral polyethylene glycol 236 g in adult patients undergoing a double-contrast barium enema procedure in three large, randomized, comparative clinical studies. In contrast, sodium picosulfate/magnesium citrate was less effective than a sodium phosphate enema preparation in two studies in patients undergoing flexible sigmoidoscopy. A similar number of patients receiving two sachets of sodium picosulfate/magnesium citrate or two 45 mL doses of oral sodium phosphate the day before a double-contrast barium enema procedure achieved satisfactory barium coating and none/minimal faecal residue in one study. However, the data from three of these studies should be interpreted with caution because the administrative regimens used differed from that recommended. Sodium picosulfate/magnesium citrate is also an effective and generally well tolerated colorectal cleansing agent in children and adolescents; the preparation was more effective than oral bisacodyl 0.01 or 0.02 g plus a sodium phosphate enema preparation in this population. Further research is thus required to accurately position sodium picosulfate/magnesium citrate and fully establish its efficacy and tolerability prior to various exploratory or surgical procedures. Nevertheless, oral sodium picosulfate/magnesium citrate provides a useful option in the preparation of the colon and rectum in adults, adolescents and children undergoing any diagnostic procedure (e.g. colonoscopy or x-ray examination) requiring a clean bowel and/or surgery. Oral sodium picosulfate/magnesium citrate acts locally in the colon as both a stimulant laxative, by increasing the frequency and the force of peristalsis (sodium picosulfate component), and an osmotic laxative, by retaining fluids in the colon (magnesium citrate component), to clear the colon and rectum of faecal contents. It is not absorbed in any detectable quantities. Sodium picosulfate is a prodrug: it is hydrolyzed by bacteria in the colon to the active metabolite 4,4'-dihydroxydiphenyl-(2-pyridyl)methane. Sodium picosulfate/magnesium citrate may be associated with a dehydrating effect, as evidenced by a reduction in bodyweight and increased haemoglobin levels; some at-risk patients may experience postural hypotension and older patients may require additional electrolytes. In three large (n >100), randomized, single-blind clinical studies, two sachets of oral sodium picosulfate/magnesium citrate was at least as effective as oral magnesium citrate 17.7 or 35.4 g, or oral polyethylene glycol 236 g as a colorectal cleansing agent in adult patients undergoing a double-contrast barium enema procedure. In contrast, sodium picosulfate/magnesium citrate was less effective than a sodium phosphate enema preparation in two studies in patients undergoing flexible sigmoidoscopy. A similar number of patients receiving two sachets of sodium picosulfate/magnesium citrate or two 45 mL doses of oral sodium phosphate the day before a double-contrast barium enema procedure achieved satisfactory barium coating and none/minimal faecal residue in one study. However, the data from three of these studies should be interpreted with caution because the administrative regimens used differed from that recommended. In children and adolescents, sodium picosulfate/magnesium citrate was significantly more effective as a colorectal cleansing agent than oral bisacodyl 0.01 or 0.02 g plus a sodium phosphate enema preparation in a randomized, single-blind study; dosages were adjusted for age in this study. Oral sodium picosulfate/magnesium citrate is generally well tolerated in adult patients undergoing various investigational colorectal procedures. Adverse events were generally mild to moderate in intensity and mainly gastrointestinal in nature (e.g. abdominal cramps/pain, nausea); other common treatment-emergent adverse events included disturbance of daily activity, headache and sleep disturbance. This combination is at least as well tolerated as oral sodium phosphate or oral polyethylene glycol, with moderate/severe nausea and vomiting occurring less frequently in sodium picosulfate/magnesium citrate recipients than in those receiving oral sodium phosphate, and abdominal bloating/pain and nausea developing less often with sodium picosulfate/magnesium citrate than polyethylene glycol therapy. The incidence of abdominal pain and sleep disturbance in sodium picosulfate/magnesium citrate versus oral magnesium citrate recipients was similar in one study, but significantly lower with sodium picosulfate/magnesium citrate in another. While the incidence of most adverse events was similar in recipients of sodium picosulfate/magnesium citrate and a sodium phosphate enema preparation, more patients receiving sodium picosulfate/magnesium citrate reported moderate/severe flatulence, incontinence and sleep disturbance, and more patients receiving the enema preparation reported rectal soreness. The tolerability profile of sodium picosulfate/magnesium citrate in patients aged >70 years is reportedly similar to that in patients aged <70 years. Abdominal pain also occurred less frequently with sodium picosulfate/magnesium citrate than with oral bisacodyl plus a sodium phosphate enema preparation in children and adolescents.


Asunto(s)
Catárticos/administración & dosificación , Ácido Cítrico/administración & dosificación , Compuestos Organometálicos/administración & dosificación , Picolinas/administración & dosificación , Administración Oral , Adolescente , Adulto , Catárticos/efectos adversos , Catárticos/farmacocinética , Niño , Citratos , Ácido Cítrico/efectos adversos , Ácido Cítrico/farmacocinética , Colonoscopía/métodos , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Humanos , Compuestos Organometálicos/efectos adversos , Compuestos Organometálicos/farmacocinética , Picolinas/efectos adversos , Picolinas/farmacocinética
11.
Drugs ; 67(2): 269-98, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17284090

RESUMEN

Voriconazole (VFEND), a synthetic second-generation, broad-spectrum triazole derivative of fluconazole, inhibits the cytochrome P450 (CYP)-dependent enzyme 14-alpha-sterol demethylase, thereby disrupting the cell membrane and halting fungal growth. In the US, intravenous and/or oral voriconazole is recommended in adults for the treatment of invasive aspergillosis, candidaemia in non-neutropenic patients, disseminated infections caused by Candida spp., oesophageal candidiasis, and in patients with scedosporiosis and fusariosis who are refractory to or intolerant of other antifungal therapy. In Europe, intravenous and/or oral voriconazole is recommended in adults and paediatric patients of at least 2 years of age for the treatment of invasive aspergillosis, candidaemia in non-neutropenic patients, fluconazole-resistant serious invasive Candida spp. infections, scedosporiosis and fusariosis. In large randomised trials, voriconazole was an effective and generally well tolerated primary treatment for candidiasis and invasive aspergillosis in adults and adolescents. More limited data also support the use of voriconazole for the treatment of invasive fungal infections in children, in those with rare fungal infections, such as Fusarium spp. or Scedosporium spp., and in those refractory to or intolerant of other standard antifungal therapies. The availability of both parenteral and oral formulations and the almost complete absorption of the drug after oral administration provide for ease of use and potential cost savings, and ensure that therapeutic plasma concentrations are maintained when switching from intravenous to oral therapy. On the other hand, the numerous drug interactions associated with voriconazole may limit its usefulness in some patients. Further clinical experience will help to more fully determine the position of voriconazole in relation to other licensed antifungal agents. In the meantime, voriconazole is a valuable emerging option for the treatment of invasive aspergillosis and rare fungal infections, including Fusarium spp. and Scedosporium spp. infections, and provides an alternative option for the treatment of candidiasis, particularly where the causative organism is inherently resistant to other licensed antifungal agents.


Asunto(s)
Antifúngicos/uso terapéutico , Micosis/tratamiento farmacológico , Pirimidinas/uso terapéutico , Triazoles/uso terapéutico , Adolescente , Adulto , Antifúngicos/efectos adversos , Antifúngicos/farmacocinética , Antifúngicos/farmacología , Niño , Farmacorresistencia Fúngica , Hongos/efectos de los fármacos , Hongos/patogenicidad , Humanos , Pruebas de Sensibilidad Microbiana , Micosis/patología , Pirimidinas/efectos adversos , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Triazoles/efectos adversos , Triazoles/farmacocinética , Triazoles/farmacología , Voriconazol
12.
Paediatr Drugs ; 6(3): 177-97, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15170364

RESUMEN

Palivizumab (Synagi) is a humanized monoclonal antibody that provides immunoprophylaxis against serious lower respiratory tract infections (LRTIs) caused by respiratory syncytial virus (RSV). RSV is the leading cause of hospitalization for LRTIs in infants, causing winter- or wet-season epidemics. In two double-blind, placebo-controlled trials, intramuscular palivizumab 15 mg/kg every 30 days for 5 months significantly reduced RSV-related hospitalizations by 55% in 1502 infants with prematurity and/or bronchopulmonary dysplasia/chronic lung disease (BPD/CLD) and by 45% in 1287 infants with hemodynamically significant congenital heart disease (HSCHD). Reductions were statistically significant versus placebo in infants with BPD/CLD, with all degrees of prematurity, and with acyanotic/other heart disease. Palivizumab was generally well tolerated, with < or =1.9% of recipients discontinuing treatment for tolerability reasons. In placebo-controlled trials, the most common potentially drug-related adverse events were fever, nervousness, injection-site reactions, and diarrhea. Drug-related events occurred in 7.2-11% of palivizumab recipients in controlled trials (vs 6.9-10% with placebo) and 0-7.9% in open-label trials. Very few serious potentially drug-related adverse events occurred in clinical trials; four occurred in 2 of 285 patients in one open-label trial. No significant anti-palivizumab antibodies developed during palivizumab use. Palivizumab trough serum concentrations were below the recommended 40 microg/mL in about 33% and up to 14% of children prior to their second and third palivizumab injections. In pharmacoeconomic studies, the cost of palivizumab per hospitalization averted was generally lowest in the highest-risk infants. Drug cost was generally the most influential factor in sensitivity analyses. In conclusion, prophylaxis with palivizumab significantly reduces the incidence of RSV-related hospitalization relative to placebo and is generally well tolerated in high-risk infants aged <2 years, including those with prematurity and BPD/CLD or HSCHD, which are risk factors for early or serious RSV infection. Palivizumab is approved for use in these patients. Other high-risk infants in whom palivizumab has not been formally assessed, such as those with immunodeficiency, cystic fibrosis, or location-specific risk factors (including extended hospital stays) might potentially benefit from palivizumab. The use of palivizumab in these other high-risk populations is likely to be determined as much by pharmacoeconomic considerations as by efficacy outcomes.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/prevención & control , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Displasia Broncopulmonar/complicaciones , Displasia Broncopulmonar/diagnóstico , Displasia Broncopulmonar/tratamiento farmacológico , Niño , Enfermedad Crónica , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Economía Farmacéutica/estadística & datos numéricos , Semivida , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/tratamiento farmacológico , Cardiopatías Congénitas/fisiopatología , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Lactante , Recién Nacido , Inyecciones Intramusculares , Modelos Biológicos , Palivizumab , Ratas , Ratas Endogámicas F344 , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA