Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Genet Couns ; 29(4): 598-606, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32227564

RESUMO

Healthcare disparities in genomic medicine are well described. Despite some improvements, we continue to see fewer individuals of African American, Asian, and Hispanic ancestry undergo genetic counseling and testing compared to those of European ancestry. It is well established that variant of uncertain significance (VUS) rates are higher among non-European ancestral groups undergoing multi-gene hereditary cancer panel testing. However, pathogenic variant (PV) yields, and genomic data in general, are often reported in aggregate and derived from cohorts largely comprised of individuals of European ancestry. We performed a retrospective review of clinical and ancestral data for individuals undergoing multi-gene hereditary cancer panel testing to determine ancestry-specific PV and VUS rates. An ancestry other than European was reported in 29,042/104,851 (27.7%) of individuals. Compared to Europeans (9.4%), individuals of Middle Eastern ancestry were more likely to test positive for one or more pathogenic variants (12.1%, p = .0025), while African Americans were less likely (7.9%, p < .0001). Asian and Middle Eastern individuals were most likely (34.8% and 33.2%, respectively) to receive a report with an overall classification of VUS, while individuals of Ashkenazi Jewish and European ancestry were least likely (17.1% and 20.4%, respectively). These data suggest that in addition to higher VUS rates, there may be ancestry-specific PV yields. Providing aggregate data derived from cohorts saturated with European individuals does not adequately reflect genetic testing outcomes in minority groups, and interrogation of ancestry-specific data is a step toward a more personalized risk assessment.


Assuntos
Asiático/genética , Negro ou Afro-Americano/genética , Predisposição Genética para Doença , Hispânico ou Latino/genética , Neoplasias/genética , Feminino , Aconselhamento Genético , Testes Genéticos , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Medição de Risco , População Branca
2.
Genet Med ; 21(7): 1497-1506, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30504931

RESUMO

PURPOSE: Several genes on hereditary breast and ovarian cancer susceptibility test panels have not been systematically examined for strength of association with disease. We employed the Clinical Genome Resource (ClinGen) clinical validity framework to assess the strength of evidence between selected genes and breast or ovarian cancer. METHODS: Thirty-one genes offered on cancer panel testing were selected for evaluation. The strength of gene-disease relationship was systematically evaluated and a clinical validity classification of either Definitive, Strong, Moderate, Limited, Refuted, Disputed, or No Reported Evidence was assigned. RESULTS: Definitive clinical validity classifications were made for 10/31 and 10/32 gene-disease pairs for breast and ovarian cancer respectively. Two genes had a Moderate classification whereas, 6/31 and 6/32 genes had Limited classifications for breast and ovarian cancer respectively. Contradictory evidence resulted in Disputed or Refuted assertions for 9/31 genes for breast and 4/32 genes for ovarian cancer. No Reported Evidence of disease association was asserted for 5/31 genes for breast and 11/32 for ovarian cancer. CONCLUSION: Evaluation of gene-disease association using the ClinGen clinical validity framework revealed a wide range of classifications. This information should aid laboratories in tailoring appropriate gene panels and assist health-care providers in interpreting results from panel testing.


Assuntos
Neoplasias da Mama/genética , Neoplasias Ovarianas/genética , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Testes Genéticos , Humanos
3.
Artigo em Inglês | MEDLINE | ID: mdl-31341520

RESUMO

BACKGROUND: Genes in the homologous recombination pathway have shown varying results in the literature regarding ovarian cancer (OC) association. Recent case-control studies have used allele counts alone to quantify genetic associations with cancer. METHODS: A retrospective case-control study was performed on 6,182 women with OC referred for hereditary cancer multi-gene panel testing (cases) and 4,690 mothers from trios who were referred for whole-exome sequencing (controls). We present age-adjusted odds ratios (ORAdj) to determine association of OC with pathogenic variants (PVs) in homologous recombination genes. RESULTS: Significant associations with OC were observed in BRCA1, BRCA2, RAD51C and RAD51D. Other homologous recombination genes, BARD1, NBN, and PALB2, were not significantly associated with OC. ATM and CHEK2 were only significantly associated with OC by crude odds ratio (ORCrude) or by ORAdj, respectively. However, there was no significant difference between ORCrude and ORAdj for these two genes. The significant association of PVs in BRIP1 by ORCrude (2.05, CI = 1.11 to 3.94, P = 0.03) was not observed by ORAdj (0.87, CI = 0.41 to 1.93, P = 0.73). Interestingly, the confidence intervals of the two effect sizes were significantly different (P = 0.04). CONCLUSION: The lack of association of PVs in BRIP1 with OC by ORAdj is inconsistent with some previous literature and current management recommendations, highlighted by the significantly older age of OC onset for BRIP1 PV carriers compared to non-carriers. By reporting ORAdj, this study presents associations that reflect more informed genetic contributions to OC when compared to traditional count-based methods.

4.
Gynecol Oncol ; 151(3): 481-488, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30322717

RESUMO

OBJECTIVE: The recognition of genes implicated in ovarian cancer risk beyond BRCA1, BRCA2, and the Lynch syndrome genes has increased the variety of testing options available to providers and patients. We report the frequency of pathogenic variants identified among individuals with ovarian cancer undergoing clinical genetic testing via a multi-gene hereditary cancer panel. METHODS: Genetic testing of up to 32 genes using a hereditary cancer panel was performed on 4439 ovarian cancer cases, and results were analyzed for frequency of pathogenic variants. Statistical comparisons were made using t-tests and Fisher's exact tests. RESULTS: The positive yield was 13.2%. While BRCA1/2 pathogenic variants were most frequent, one third (33.7%) of positive findings were in other homologous recombination genes, and accounted for over 40.0% of findings in endometrioid and clear cell cases. Women with a personal history of breast cancer (22.1%), who reported a family history of ovarian cancer (17.7%), and/or serous histology (14.7%) were most likely to harbor a pathogenic variant. Those with very early onset (<30 years) and late onset (≥70 years) ovarian cancer had low positive yields. CONCLUSIONS: Our study highlights the genetic heterogeneity of ovarian cancer, showing that a large proportion of cases are not due to BRCA1/2 and the Lynch syndrome genes, but still have an identifiable hereditary basis. These findings substantiate the utility of multi-gene panel testing in ovarian cancer care regardless of age at diagnosis, family history, or histologic subtype, providing evidence for testing beyond BRCA1/2 and the Lynch syndrome genes.


Assuntos
Mutação em Linhagem Germinativa/genética , Neoplasias Ovarianas/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Adulto Jovem
5.
Int J Gynecol Pathol ; 37(2): 117-122, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28463906

RESUMO

We report the first case of distinct, synchronous serous carcinomas of the adnexa arising in a patient with a family history of breast and ovarian cancer and a germline loss of function mutation in BRCA1. Illustrating an exceedingly rare phenomenon of synchronous high-grade carcinomas with distinct histomorphologic, immunohistochemical and cytogenetic features, the case serves as a point of departure for the discussion of phenotypic patterns of carcinomas arising in BRCA1 mutation carriers. We also review patient management, including the importance of risk-reducing salpingo-oophorectomy in women with deleterious BRCA1 mutations, as well as the potential need for an intraoperative pathologic assessment to find occult, high-grade carcinomas in this setting.


Assuntos
Adenocarcinoma/genética , Proteína BRCA1/genética , Biomarcadores Tumorais/genética , Cistadenocarcinoma Seroso/genética , Neoplasias Primárias Múltiplas/genética , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Proteína BRCA1/metabolismo , Biomarcadores Tumorais/metabolismo , Hibridização Genômica Comparativa , Cistadenocarcinoma Seroso/diagnóstico , Cistadenocarcinoma Seroso/patologia , Cistadenocarcinoma Seroso/cirurgia , Éxons/genética , Feminino , Duplicação Gênica , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Humanos , Mutação com Perda de Função , Pessoa de Meia-Idade , Neoplasias Primárias Múltiplas/diagnóstico , Neoplasias Primárias Múltiplas/patologia , Neoplasias Primárias Múltiplas/cirurgia , Salpingo-Ooforectomia
6.
Breast Cancer Res ; 18(1): 15, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26857456

RESUMO

BACKGROUND: BRCA1 and, more commonly, BRCA2 mutations are associated with increased risk of male breast cancer (MBC). However, only a paucity of data exists on the pathology of breast cancers (BCs) in men with BRCA1/2 mutations. Using the largest available dataset, we determined whether MBCs arising in BRCA1/2 mutation carriers display specific pathologic features and whether these features differ from those of BRCA1/2 female BCs (FBCs). METHODS: We characterised the pathologic features of 419 BRCA1/2 MBCs and, using logistic regression analysis, contrasted those with data from 9675 BRCA1/2 FBCs and with population-based data from 6351 MBCs in the Surveillance, Epidemiology, and End Results (SEER) database. RESULTS: Among BRCA2 MBCs, grade significantly decreased with increasing age at diagnosis (P = 0.005). Compared with BRCA2 FBCs, BRCA2 MBCs were of significantly higher stage (P for trend = 2 × 10(-5)) and higher grade (P for trend = 0.005) and were more likely to be oestrogen receptor-positive [odds ratio (OR) 10.59; 95 % confidence interval (CI) 5.15-21.80] and progesterone receptor-positive (OR 5.04; 95 % CI 3.17-8.04). With the exception of grade, similar patterns of associations emerged when we compared BRCA1 MBCs and FBCs. BRCA2 MBCs also presented with higher grade than MBCs from the SEER database (P for trend = 4 × 10(-12)). CONCLUSIONS: On the basis of the largest series analysed to date, our results show that BRCA1/2 MBCs display distinct pathologic characteristics compared with BRCA1/2 FBCs, and we identified a specific BRCA2-associated MBC phenotype characterised by a variable suggesting greater biological aggressiveness (i.e., high histologic grade). These findings could lead to the development of gender-specific risk prediction models and guide clinical strategies appropriate for MBC management.


Assuntos
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias da Mama Masculina/genética , Neoplasias da Mama/genética , Adulto , Idoso , Neoplasias da Mama/patologia , Neoplasias da Mama Masculina/patologia , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Polimorfismo de Nucleotídeo Único
7.
Am J Cardiovasc Drugs ; 10(6): 383-400, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21090831

RESUMO

Rosuvastatin (Crestor®) is an HMG-CoA reductase inhibitor (statin) that has both lipid-lowering and anti-inflammatory effects. The drug has various indications in the US, including the primary prevention of cardiovascular disease (CVD) in patients with no clinical evidence of coronary heart disease who are at increased risk of CVD based on their age, a high-sensitivity C-reactive protein (hsCRP) level of ≥2 mg/L, and at least one other CVD risk factor. The efficacy of rosuvastatin in apparently healthy women (aged ≥60 years) or men (aged ≥50 years) with normal low-density lipoprotein cholesterol (LDL-C) levels and elevated hsCRP levels was demonstrated in the large, randomized, double-blind, multinational, JUPITER trial. Relative to placebo, rosuvastatin 20 mg once daily for a median follow-up of 1.9 years significantly reduced the occurrence of first major cardiovascular events in this trial (primary endpoint). A between-group difference in favor of rosuvastatin was also demonstrated for various other endpoints, including overall deaths and the nonatherothrombotic endpoint of venous thromboembolism. Rosuvastatin remained more effective than placebo when primary endpoint results were stratified according to various baseline factors, including in patient subgroups thought to be at low risk of CVD. In addition, rosuvastatin was associated with reductions in LDL-C and hsCRP levels, and these reductions appeared to occur independently of each other. The greatest clinical benefit was observed in rosuvastatin recipients achieving an LDL-C level of <1.8 mmol/L (<70 mg/dL) and an hsCRP level of <2 mg/L or, even more so, <1 mg/L. Rosuvastatin was well tolerated in the JUPITER trial, with most adverse events being mild to moderate in severity. Myalgia, arthralgia, constipation, and nausea were the most commonly occurring treatment-related adverse events, and the incidence of monitored adverse events and laboratory measurements was generally similar in the rosuvastatin and placebo groups. It is not yet known whether the mechanism of benefit of rosuvastatin is via lipid effects, anti-inflammatory effects, or a mixture of both, and the use of rosuvastatin solely on the basis of elevated hsCRP levels is controversial. Nonetheless, the drug remains an important pharmacologic option in the prevention of CVD, and has demonstrated efficacy in preventing major cardiovascular events in apparently healthy women (aged ≥60 years) or men (aged ≥50 years) with normal LDL-C levels and elevated hsCRP levels.


Assuntos
Proteína C-Reativa/análise , Doenças Cardiovasculares/prevenção & controle , LDL-Colesterol/sangue , Fluorbenzenos/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Pirimidinas/uso terapêutico , Sulfonamidas/uso terapêutico , Feminino , Fluorbenzenos/efeitos adversos , Fluorbenzenos/farmacocinética , Fluorbenzenos/farmacologia , Humanos , Masculino , Pirimidinas/efeitos adversos , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Rosuvastatina Cálcica , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética , Sulfonamidas/farmacologia
8.
CNS Drugs ; 23(6): 523-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19480470

RESUMO

Duloxetine (Cymbalta(R)) is a potent serotonin and noradrenaline (norepinephrine) reuptake inhibitor (SNRI) in the CNS. It is indicated for the treatment of generalized anxiety disorder (GAD) as well as other indications. In patients with GAD of at least moderate severity, oral duloxetine 60-120 mg once daily was effective with regard to improvement from baseline in assessments of anxiety and functional impairment, and numerous other clinical endpoints. Longer-term duloxetine 60-120 mg once daily also demonstrated efficacy in preventing or delaying relapse in responders among patients with GAD. In addition, duloxetine was generally well tolerated, with most adverse events being of mild to moderate severity in patients with GAD in short- and longer-term trials. Additional comparative and pharmacoeconomic studies are required to position duloxetine among other selective serotonin reuptake inhibitors and SNRIs. However, available clinical data, and current treatment guidelines, indicate that duloxetine is an effective first-line treatment option for the management of GAD. Duloxetine is a potent and selective inhibitor of serotonin and noradrenaline transporters, and a weak inhibitor of dopamine transporters. It has a low affinity for neuronal receptors, such as alpha(1)- and alpha(2)-adrenergic, dopamine D(2), histamine H(1), muscarinic, opioid and serotonin receptors, as well as ion channel binding sites and other neurotransmitter transporters, such as choline and GABA transporters. It does not inhibit monoamine oxidase types A or B. The pharmacokinetics of duloxetine in healthy volunteers were dose proportional over the range of 40-120 mg once daily. Steady state was typically reached by day 3 of administration. Duloxetine may be administered without regard to food or time of day. Duloxetine is highly protein bound and is widely distributed throughout tissues. It is rapidly and extensively metabolized in the liver by cytochrome P450 (CYP) 1A2 and 2D6, and its numerous metabolites, which are inactive, are mainly excreted in the urine. The mean elimination half-life of duloxetine is approximately 12 hours. Duloxetine is a substrate for CYP1A2 and CYP2D6 and a moderate inhibitor of CYP2D6. Concomitant use of duloxetine and potent CYP1A2 inhibitors should be avoided and duloxetine should be used with caution in patients receiving drugs that are extensively metabolized by CYP2D6, particularly those with a narrow therapeutic index. Duloxetine was effective in the short-term treatment of patients with primary GAD of at least moderate severity. In four randomized, double-blind, placebo-controlled, multicentre, phase III trials, duloxetine 60-120 mg once daily for 9 or 10 weeks was significantly more effective than placebo with regard to the primary endpoint of mean change in Hamilton Anxiety Rating Scale (HAM-A) total score from baseline to study endpoint. In addition, all other endpoints were generally improved from baseline to a greater extent with duloxetine 60-120 mg once daily than with placebo. Duloxetine also improved patient role functioning (assessed using Sheehan Disability Scale global impairment functioning scores), health-related quality of life and patient well-being compared with placebo. Duloxetine was effective in patients with GAD who were aged >/=65 years. Pooled results of data from the two short-term efficacy trials that also included an active comparator arm showed that the mean change in HAM-A scores with duloxetine relative to placebo were of the same magnitude as those with venlafaxine extended release versus placebo. Duloxetine 60-120 mg once daily was also more effective than placebo in preventing or delaying relapse in responders to duloxetine in a longer-term study. In this study, patients with GAD received duloxetine during a 26-week, open-label, acute treatment phase and responders were then randomized to continue on duloxetine or receive placebo during a 26-week, double-blind, continuation phase. Time to relapse was significantly longer in duloxetine recipients than in placebo recipients. In addition, significantly fewer duloxetine recipients than placebo recipients relapsed during the double-blind phase of the trial and more duloxetine recipients achieved remission. Short- (9-10 weeks) and longer-term (52 weeks) treatment with duloxetine 60-120 mg once daily was generally well tolerated in patients with GAD, with the majority of adverse events being of mild to moderate severity. Nausea, dry mouth, headache, constipation, dizziness and fatigue were among the most common treatment-emergent adverse events. The adverse event profile of duloxetine did not differ with dose or treatment duration. Significantly more patients receiving short-term duloxetine than placebo discontinued treatment because of an adverse event, with nausea being the only event that resulted in significantly more treatment discontinuations in duloxetine recipients than in placebo recipients. Serious adverse events were uncommon with both short- and longer-term duloxetine treatment. Two episodes of attempted suicide and one episode of completed suicide occurred in duloxetine recipients during the 24-week open-label phase of a longer-term trial. No deaths or suicides were reported in any of the short-term trials. Discontinuation-emergent adverse events, most commonly nausea and dizziness, occurred in up to one-third of duloxetine recipients in the short-term trials.


Assuntos
Antidepressivos/uso terapêutico , Transtornos de Ansiedade/tratamento farmacológico , Tiofenos/uso terapêutico , Bases de Dados Bibliográficas/estatística & dados numéricos , Relação Dose-Resposta a Droga , Esquema de Medicação , Avaliação de Medicamentos , Cloridrato de Duloxetina , Humanos , Metanálise como Assunto , Resultado do Tratamento
9.
Paediatr Drugs ; 11(4): 271-91, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19566111

RESUMO

Intravenous micafungin (Mycamine; Funguard) is an echinocandin indicated in Japan and the EU for the treatment of pediatric patients (including neonates) with invasive candidiasis and as prophylaxis against Candida infection in pediatric patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT). In the EU, micafungin is also indicated in pediatric patients who are expected to have neutropenia for >/=10 days. In Japan, children may also receive micafungin for the treatment of, or as prophylaxis against, invasive Aspergillus infection. Micafungin is not currently approved for use in pediatric patients in the US. Micafungin has very good antifungal activity against a wide range of Candida spp. in vitro. It has a favorable pharmacokinetic profile allowing for once-daily administration, has few drug-drug interactions, and reports of resistance are rare. The results of pediatric substudies indicate that intravenous micafungin is effective in a majority of patients for the treatment of candidemia and other types of invasive candidiasis, and provides effective prophylaxis against invasive fungal infections in pediatric patients undergoing HSCT. The tolerability profile of micafungin in pediatric patients was generally acceptable. In the EU, micafungin is indicated for use when other antifungal medications are not appropriate. Therefore, micafungin provides an alternative to other antifungal agents used in the management of candidemia and invasive candidiasis in pediatric patients, or as prophylaxis against fungal infections in pediatric patients undergoing HSCT.


Assuntos
Antifúngicos/uso terapêutico , Candidíase/tratamento farmacológico , Candidíase/prevenção & controle , Equinocandinas/uso terapêutico , Lipopeptídeos/uso terapêutico , Antifúngicos/administração & dosagem , Antifúngicos/efeitos adversos , Antifúngicos/farmacocinética , Antifúngicos/farmacologia , Candidíase/microbiologia , Criança , Equinocandinas/administração & dosagem , Equinocandinas/efeitos adversos , Equinocandinas/farmacocinética , Equinocandinas/farmacologia , Humanos , Lipopeptídeos/administração & dosagem , Lipopeptídeos/efeitos adversos , Lipopeptídeos/farmacocinética , Lipopeptídeos/farmacologia , Micafungina
10.
Drugs ; 68(18): 2645-60, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19093705

RESUMO

Bendamustine (Treanda, Ribomustin) is a bifunctional alkylating agent that also has potential antimetabolite properties, and only partial cross-resistance occurs between bendamustine and other alkylators. In patients with indolent non-Hodgkin lymphoma (NHL), bendamustine monotherapy achieved high objective response rates in those with rituximab-refractory disease in a pivotal noncomparative trial and a similarly designed smaller phase II study. Many of these heavily treated patients were also refractory to standard chemotherapy regimens. Several phase II trials demonstrated good response rates with single-agent bendamustine, or bendamustine in combination with rituximab, in patients with indolent NHL whose disease relapsed after (or was refractory to) chemotherapy. Phase III studies comparing combination regimens as first-line therapy in patients with indolent NHL showed no significant differences in response rates between bendamustine-containing regimens and standard regimens included in treatment guidelines. Bendamustine has been generally well tolerated in clinical trials and has a low propensity to induce alopecia. Results of ongoing trials will help to clarify the optimal role of bendamustine in indolent NHL. Available data indicate that it may be a particularly useful treatment option as monotherapy in patients with indolent NHL whose disease progressed during or following rituximab-based therapy, many of whom are also refractory to standard chemotherapy regimens.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Linfoma não Hodgkin/tratamento farmacológico , Neoplasia Residual/tratamento farmacológico , Compostos de Mostarda Nitrogenada/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Murinos , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cloridrato de Bendamustina , Ensaios Clínicos como Assunto , Resistencia a Medicamentos Antineoplásicos , Meia-Vida , Humanos , Infusões Intravenosas , Linfoma não Hodgkin/fisiopatologia , Compostos de Mostarda Nitrogenada/metabolismo , Compostos de Mostarda Nitrogenada/farmacocinética , Rituximab
11.
Drugs ; 68(5): 691-710, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370449

RESUMO

Enoxaparin (enoxaparin sodium; Lovenox) is a low-molecular-weight heparin (LMWH) that has recently been approved by the US FDA for use in patients with medically managed ST-segment myocardial infarction (STEMI), or STEMI with subsequent percutaneous coronary intervention (PCI). It binds to and potentiates the action of antithrombin, and inhibits coagulation factors XIa, IXa, Xa and IIa (thrombin), thereby preventing formation of blood clots. Unfractionated heparin (UFH) has long been regarded as the antithrombotic agent of choice in the adjunctive treatment of patients with STEMI. However, compared with UFH, enoxaparin has many advantages in terms of its pharmacodynamic profile and, potentially, also its efficacy. Enoxaparin was significantly more effective than UFH in patients presenting with STEMI who underwent fibrinolytic therapy in terms of the 30-day combined incidence of all-cause mortality plus recurrent nonfatal myocardial infarction (MI) [primary endpoint], and all-cause mortality plus recurrent nonfatal MI plus urgent revascularization (secondary endpoint) in the ExTRACT-TIMI 25 trial. The significant difference in the incidence of the composite primary endpoint between these two groups was maintained at the 1-year follow-up. Although bleeding was reported more frequently with enoxaparin than with UFH in the ExTRACT-TIMI 25 trial, enoxaparin was associated with a net clinical benefit relative to UFH. Patients in this trial received enoxaparin as an initial 30 mg intravenous bolus, followed by 1 mg/kg subcutaneously within 15 minutes and then every 12 hours for up to 8 days; the first two subcutaneous dosages were not to exceed 100 mg. Patients > or =75 years of age did not receive the initial bolus of enoxaparin and the 12-hourly dosages were reduced to 0.75 mg/kg; the dose was also reduced to 1 mg/kg every 24 hours in patients of any age who had an estimated creatinine clearance (CL(CR)) of <30 mL/min. Data from several earlier randomized, multicentre, phase III trials support these results.


Assuntos
Anticoagulantes/uso terapêutico , Eletrocardiografia/efeitos dos fármacos , Enoxaparina/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Adulto , Idoso , Angiografia Coronária , Enoxaparina/efeitos adversos , Enoxaparina/farmacocinética , Enoxaparina/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/fisiopatologia
12.
BioDrugs ; 22(5): 279-92, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18778110

RESUMO

Although rare, influenza pandemics are a recurrent event, and influenza A/H5N1 is generally considered to be the most likely causative agent of the next pandemic. Vaccines are widely considered to be the first line of defense for protecting populations in advance of an influenza pandemic. Because it is not known beforehand which strain of influenza A/H5N1 virus could give rise to a pandemic, prepandemic vaccines that impart broad cross-reactive immunogenicity are required. In addition, low doses of H5 hemagglutinin are preferable in order to make antigen supplies go further towards meeting global demands for prepandemic vaccines.Prepandemic influenza vaccine H5N1 [Prepandrix(trade mark); AS03-H5N1 vaccine] is a split virion, inactivated vaccine containing H5 hemagglutinin antigen adjuvanted with a novel 10% oil-in-water emulsion-based adjuvant system (AS03). It is approved in the EU for use as an active immunization against H5N1 subtype influenza A virus (influenza A/H5N1 virus) in adults aged 18-60 years. The recommended dosage in this population is two doses of 0.5 mL containing 3.75 microg of H5 hemagglutinin, administered > or =21 days apart. Adjuvantation of H5N1 vaccine with AS03 allows for a reduction in the H5 hemagglutinin dose required to elicit an adequate immune response, and administration of two doses of the adjuvanted vaccine met all criteria for the licensure of influenza vaccines set out in European Committee for Proprietary Medicinal Products (CPMP) and US FDA documents. In two clinical trials, two doses of AS03-H5N1 vaccine containing 3.75 microg of H5 hemagglutinin induced an immune response in healthy volunteers aged 18-60 years against the homologous, clade 1 vaccine strain, A/Vietnam/1194/2004, and the heterologous, drifted, clade 2 nonvaccine strains, A/Anhui/1/2005, A/Indonesia/5/2005, and A/turkey/Turkey/1/2005. This cross-clade response persisted for > or =6 months following administration of the first vaccine dose in the majority of vaccine recipients. In addition, AS03-H5N1 vaccine protected against lethal challenge with A/Vietnam/1194/2004 or A/Indonesia/5/2005 in animal studies. The vaccine was generally well tolerated and adverse events were transient and predominantly of mild to moderate severity.AS03-H5N1 vaccine has demonstrated antigen dose-sparing properties and cross-clade reactive immunity in clinical trials and challenge studies in animal models. As a result, stockpiling AS03-H5N1 vaccine has the potential to protect populations against a pandemic caused by an influenza A/H5N1 virus and may represent an important measure in pandemic preparedness.


Assuntos
Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Vacinação , Animais , Surtos de Doenças/prevenção & controle , Humanos , Vacinas contra Influenza/efeitos adversos
13.
BioDrugs ; 22(5): 343-5, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18778116

RESUMO

Enoxaparin (enoxaparin sodium; Lovenox) is a low-molecular-weight heparin (LMWH) that has recently been approved by the US FDA for use in patients with medically managed ST-segment myocardial infarction (STEMI), or STEMI with subsequent percutaneous coronary intervention. It binds to and potentiates the action of antithrombin, and inhibits coagulation factors XIa, IXa, Xa, and IIa (thrombin), thereby preventing formation of blood clots. Unfractionated heparin (UFH) has long been regarded as the antithrombotic agent of choice in the adjunctive treatment of patients with STEMI. However, compared with UFH, enoxaparin has many advantages in terms of its pharmacodynamic profile and, potentially, also its efficacy. Enoxaparin was significantly more effective than UFH in patients presenting with STEMI who underwent fibrinolytic therapy in terms of the 30-day combined incidence of all-cause mortality plus recurrent nonfatal myocardial infarction (MI) [primary endpoint], and all-cause mortality plus recurrent nonfatal MI plus urgent revascularization (secondary endpoint) in the ExTRACT-TIMI 25 trial. The significant difference in the incidence of the composite primary endpoint between these two groups was maintained at the 1-year follow-up. Although bleeding was reported more frequently with enoxaparin than with UFH in the ExTRACT-TIMI 25 trial, enoxaparin was associated with a net clinical benefit relative to UFH. Patients in this trial received enoxaparin as an initial 30 mg intravenous bolus, followed by 1 mg/kg subcutaneously within 15 minutes and then every 12 hours for up to 8 days; the first two subcutaneous dosages were not to exceed 100 mg. Patients > or =75 years of age did not receive the initial bolus of enoxaparin and the 12-hourly dosages were reduced to 0.75 mg/kg; the dose was also reduced to 1 mg/kg every 24 hours in patients of any age who had an estimated creatinine clearance of <30 mL/min. Data from several earlier randomized, multicentre, phase III trials support these results.


Assuntos
Anticoagulantes/uso terapêutico , Eletrocardiografia , Enoxaparina/uso terapêutico , Enoxaparina/efeitos adversos , Enoxaparina/farmacologia , Humanos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia
14.
Drugs ; 67(10): 1513-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17600395

RESUMO

Haemate P/Humate-P is a pasteurised human plasma-derived concentrate containing coagulation factor VIII and a near-normal spectrum of von Willebrand factor multimers, including high-molecular weight multimers, for intravenous use in patients with von Willebrand disease or haemophilia A. Extensive clinical experience over the past 25 years has shown that Haemate P/Humate-P provides effective haemostatic control for the prevention and treatment of bleeds in patients with these conditions, with no confirmed cases of viral or prion transmission occurring during this time. In small prospective and retrospective noncomparative studies, Haemate P/Humate-P provided effective haemostatic control for the prevention and treatment of bleeding episodes in the vast majority of paediatric and adult patients with von Willebrand disease. Haemate P/Humate-P was generally well tolerated in patients with von Willebrand disease or haemophilia A.


Assuntos
Coagulantes/uso terapêutico , Fator VIII/uso terapêutico , Hemofilia A/terapia , Doenças de von Willebrand/terapia , Fator de von Willebrand/efeitos adversos , Fator de von Willebrand/uso terapêutico , Coagulantes/efeitos adversos , Coagulantes/farmacologia , Fator VIII/efeitos adversos , Fator VIII/farmacologia , Humanos , Fator de von Willebrand/farmacologia
15.
Drugs ; 67(15): 2257-76, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17927287

RESUMO

Trabectedin (Yondelis); ET-743) is an antineoplastic agent that was originally derived from the Caribbean marine tunicate Ecteinascidia turbinata and is now produced synthetically. It binds to the minor groove of DNA, disrupting the cell cycle and inhibiting cell proliferation. Intravenous trabectedin administered once every 3 weeks is approved as monotherapy in Europe for use in patients with advanced soft tissue sarcoma (STS) after failure of standard therapy with anthracyclines or ifosfamide, or who are unsuited to receive these agents. It also has orphan drug status in STS in the US and in ovarian cancer in the US and Europe, and is under investigation as combination therapy in patients with recurrent ovarian cancer. In clinical trials, trabectedin showed efficacy in the treatment of patients with advanced or metastatic STS, especially those with leiomyosarcoma or liposarcoma, as well as in women with platinum-sensitive advanced or recurrent ovarian cancer. In addition, its tolerability profile was generally manageable. The introduction of trabectedin expands the currently limited range of effective treatment options for patients with advanced or metastatic STS; trabectedin also has the potential to be a beneficial treatment for advanced or recurrent ovarian cancer.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Dioxóis/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Sarcoma/tratamento farmacológico , Tetra-Hidroisoquinolinas/uso terapêutico , Animais , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/efeitos adversos , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacologia , Dioxóis/administração & dosagem , Dioxóis/efeitos adversos , Dioxóis/farmacocinética , Dioxóis/farmacologia , Feminino , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Tetra-Hidroisoquinolinas/administração & dosagem , Tetra-Hidroisoquinolinas/efeitos adversos , Tetra-Hidroisoquinolinas/farmacocinética , Tetra-Hidroisoquinolinas/farmacologia , Trabectedina
16.
Drugs ; 67(15): 2277-88; discussion 2289-90, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17927288

RESUMO

Maraviroc is a specific, slowly reversible, noncompetitive, small-molecule antagonist of the CCR5 chemokine receptor, which also serves as an HIV-1 coreceptor. By acting as an antagonist at the CCR5 coreceptor, maraviroc inhibits HIV-1 from entering host cells. Clinical data for maraviroc are available from two large, well designed, ongoing phase IIb/III trials (MOTIVATE-1 and MOTIVATE-2) conducted in patients infected with R5-tropic HIV-1 who had previously received at least one agent from three of the four classes of antiretroviral drugs and/or were triple-class resistant. According to 24-week interim results of the MOTIVATE-1 and -2 trials, a significantly greater reduction in viral load occurred in patients receiving maraviroc 150 or 300mg (depending on optimised background therapy [OBT]) twice daily plus OBT compared with placebo plus OBT. This significant difference was maintained at 48 weeks in MOTIVATE-1. In the MOTIVATE-1 and -2 trials, a significantly greater proportion of patients receiving maraviroc plus OBT achieved an HIV-1 RNA level <400 and <50 copies/mL compared with those receiving placebo plus OBT. In addition, the CD4+ cell count was increased to a significantly greater extent with maraviroc plus OBT compared with placebo plus OBT. The 48-week results of MOTIVATE-1 also report a significant difference in favour of maraviroc for all these endpoints. In general, maraviroc at dosages of up to 300mg twice daily was well tolerated in treatment-experienced patients infected with R5-tropic HIV-1.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Cicloexanos/uso terapêutico , Infecções por HIV/tratamento farmacológico , Triazóis/uso terapêutico , Animais , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/efeitos adversos , Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/farmacologia , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Cicloexanos/administração & dosagem , Cicloexanos/efeitos adversos , Cicloexanos/farmacocinética , Cicloexanos/farmacologia , Farmacorresistência Viral , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Humanos , Maraviroc , Triazóis/administração & dosagem , Triazóis/efeitos adversos , Triazóis/farmacocinética , Triazóis/farmacologia
17.
Drugs Aging ; 31(1): 67-78, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24276917

RESUMO

Regorafenib (Stivarga) is an inhibitor of multiple protein kinases, including those involved in oncogenesis, tumour angiogenesis and maintenance of the tumour microenvironment. The drug is approved as monotherapy for the treatment of metastatic colorectal cancer (mCRC) in patients who have previously received all standard systemic anticancer treatments (US, EU and Canada) or in patients with unresectable, advanced or recurrent colorectal cancer (Japan). In the randomized, controlled COloRectal cancer treated with REgorafenib or plaCebo after failure of standard Therapy (CORRECT) trial, regorafenib 160 mg once daily for the first 3 weeks of each 4-week cycle plus best supportive care (BSC) was associated with a significantly longer median overall survival than placebo plus BSC in patients with previously treated, progressive mCRC. The drug was also associated with significantly longer progression-free survival and better disease control rates than placebo, although objective response rates were similar in both treatment groups. Regorafenib did not appear to compromise health-related quality of life over the study duration and had a generally acceptable tolerability profile. The introduction of regorafenib expands the currently limited range of effective treatment options in patients with previously treated, progressive mCRC.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Compostos de Fenilureia/uso terapêutico , Piridinas/uso terapêutico , Progressão da Doença , Humanos , Metástase Neoplásica , Compostos de Fenilureia/efeitos adversos , Compostos de Fenilureia/farmacocinética , Compostos de Fenilureia/farmacologia , Piridinas/efeitos adversos , Piridinas/farmacocinética , Piridinas/farmacologia , Resultado do Tratamento
18.
Drugs ; 74(6): 699-712, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24756432

RESUMO

Degarelix (Firmagon(®); Gonax(®)) is a gonadotropin-releasing hormone receptor antagonist that is approved for the treatment of advanced (hormone-dependent) prostate cancer in the US and EU and the treatment of prostate cancer in Japan. In a pivotal randomized, controlled, 12-month phase III study, degarelix (initial subcutaneous dose of 240 mg followed by monthly dosages of 80 mg) was noninferior to leuprolide (monthly intramuscular dosages of 7.5 mg) in patients with prostate cancer of any stage for which endocrine treatment was indicated (except neoadjuvant hormonal therapy) with regard to suppression of testosterone to castration levels (i.e. ≤0.5 ng/mL). Suppression of testosterone and prostate-specific antigen (PSA) levels was faster with degarelix than with leuprolide, and no testosterone surges or microsurges were seen in degarelix recipients. Suppression of testosterone and PSA levels was maintained for the 12-month study duration and continued for up to 5 years in an extension to the main trial (including in patients switching from leuprolide to degarelix in the extension). The drug was generally well tolerated, with most adverse events being mild to moderate in severity. Injection-site reactions and events reflecting the expected effects of testosterone suppression (e.g. hot flushes, weight increase) were the most common treatment-emergent adverse events. Thus, degarelix is a useful option for the treatment of prostate cancer in patients for whom endocrine treatment is indicated.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Oligopeptídeos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Animais , Antineoplásicos Hormonais/efeitos adversos , Antineoplásicos Hormonais/farmacologia , Aprovação de Drogas , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Humanos , Masculino , Oligopeptídeos/efeitos adversos , Oligopeptídeos/farmacologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/patologia , Testosterona/metabolismo
20.
BioDrugs ; 27(3): 263-74, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23575646

RESUMO

Multicomponent meningococcal serogroup B vaccine (4CMenB; Bexsero(®)) is a unique vaccine containing four main immunogenic components: three recombinant proteins combined with outer membrane vesicles derived from meningococcal NZ98/254 strain. After three doses of 4CMenB (administered at 2, 3, and 4 months or 2, 4, and 6 months of age) in vaccine-naive infants, the majority of infants had seroprotective human complement serum bactericidal assay (hSBA) antibody titers against the meningococcal serogroup B test strains selected to be specific for the vaccine antigens in randomized, open-label or observer-blind, multicenter, phase IIb or III trials. In extensions to the phase III trial, two doses of 4CMenB administered between 12 and 15 months of age in vaccine-naive infants, and a single booster dose of 4CMenB administered at 12 months of age in vaccine-experienced infants, also elicited robust immunogenic responses. In a phase IIb/III trial, the majority of adolescents (aged 11-17 years) achieved seroprotective hSBA antibody titers against meningococcal serogroup B test strains after two doses of 4CMenB, and a third dose did not appear to add any extra protection. In adults who were potentially at an increased risk of occupational exposure to meningococcal isolates, seroprotection rates were high after one dose of 4CMenB and increased further after two or three doses in a small noncomparative, two-center, phase II trial. The reactogenicity of 4CMenB was generally acceptable in clinical trials. However, the vaccine was associated with more solicited systemic adverse events (particularly fever) in infants when coadministered with routine infant vaccines than when these vaccines were administered alone. In conclusion, 4CMenB effectively elicited immune responses against meningococcal serogroup B test strains selected to be specific for the vaccine antigens in infants, adolescents, and adults.


Assuntos
Infecções Meningocócicas/prevenção & controle , Vacinas Meningocócicas/administração & dosagem , Neisseria meningitidis Sorogrupo B , Adolescente , Adulto , Humanos , Esquemas de Imunização , Imunização Secundária , Lactente , Vacinas Meningocócicas/efeitos adversos , Vacinas Meningocócicas/imunologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Ensaios de Anticorpos Bactericidas Séricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA