Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Oncologist ; 26(3): 242-249, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33486852

RESUMO

On November 5, 2020, a marketing authorization valid through the European Union (EU) was issued for acalabrutinib monotherapy or acalabrutinib in combination with obinutuzumab (AcalaObi) in adult patients with treatment-naïve (TN) chronic lymphocytic leukemia (CLL) and also for acalabrutinib monotherapy in adult patients with relapsed or refractory (RR) CLL. Acalabrutinib inhibits the Bruton tyrosine kinase, which plays a significant role in the proliferation and survival of the disease. Acalabrutinib was evaluated in two phase III multicenter randomized trials. The first trial (ACE-CL-007) randomly allocated acalabrutinib versus AcalaObi versus chlorambucil plus obinutuzumab (ChlObi) to elderly/unfit patients with TN CLL. The progression-free survival (PFS), as assessed by an independent review committee, was superior for both the AcalaObi (hazard ratio [HR], 0.1; 95% confidence interval [CI], 0.06-0.17) and acalabrutinib (HR, 0.2; 95% CI, 0.13-0.3) arms compared with the ChlObi arm. The second trial (ACE-CL-309) randomly allocated acalabrutinib versus rituximab plus idelalisib or bendamustine to adult patients with RR CLL. Also in this trial, the PFS was significantly longer in the acalabrutinib arm (HR, 0.31; 95% CI, 0.20-0.49). Adverse events for patients receiving acalabrutinib varied across trials, but the most frequent were generally headache, diarrhea, neutropenia, nausea, and infections. The scientific review concluded that the benefit-risk ratio of acalabrutinib was positive for both indications. This article summarizes the scientific review of the application leading to regulatory approval in the EU. IMPLICATIONS FOR PRACTICE: Acalabrutinib was approved in the European Union for the treatment of adult patients with chronic lymphocytic leukemia who have not received treatment before and for those who have received therapy but whose disease did not respond or relapsed afterward. Acalabrutinib resulted in a clinically meaningful and significant lengthening of the time from treatment initiation to further disease relapse or patient's death compared with standard therapy. The overall safety profile was considered acceptable, and the benefit-risk ratio was determined to be positive.


Assuntos
Leucemia Linfocítica Crônica de Células B , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Benzamidas/uso terapêutico , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Estudos Multicêntricos como Assunto , Pirazinas/efeitos adversos , Rituximab/uso terapêutico
2.
Oncologist ; 25(4): e709-e715, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32297447

RESUMO

On November 15, 2018, the Committee for Medicinal Products for Human Use (CHMP) recommended the extension of indication for blinatumomab to include the treatment of adults with minimal residual disease (MRD) positive B-cell precursor acute lymphoblastic leukemia (ALL). Blinatumomab was authorized to treat relapsed or refractory B-precursor ALL, and the change concerned an extension of use. On March 29, 2018, the U.S. Food and Drug Administration (FDA) granted accelerated approval to blinatumomab to treat both adults and children with B-cell precursor ALL who are in remission but still have MRD. On July 26, 2018, the CHMP had originally adopted a negative opinion on the extension. The reason for the initial refusal was that although blinatumomab helped to reduce the amount of residual cancer cells in many patients, there was no strong evidence that it led to improved survival. During the re-examination, the CHMP consulted the scientific advisory group. The CHMP agreed with the expert group's conclusion that, although there was no strong evidence of patients living longer, the available data from the main study (MT103-203) indicated a good durable response to blinatumomab, with an overall complete response rate for the primary endpoint full analysis set (defined as all subjects with an Ig or T-cell receptor polymerase chain reaction MRD assay with the minimum required sensitivity of 1 × 10-4 at central lab established at baseline [n = 113]) as 79.6% (90/113; 95% confidence interval, 71.0-86.6), with a median time to complete MRD response of 29.0 days (range, 5-71). Therefore, the CHMP concluded that the benefits of blinatumomab outweigh its risks and recommended granting the change to the marketing authorization. The Committee for Orphan Medicinal Products, following reassessment, considered that significant benefit continued to be met and recommended maintaining the orphan designation and thus 10 years market exclusivity (the Orphan Designation is a legal procedure that allows for the designation of a medicinal substance with therapeutic potential for a rare disease, before its first administration in humans or during its clinical development). The marketing authorization holder for this medicinal product is Amgen Europe B.V. IMPLICATIONS FOR PRACTICE: Immunotherapy with blinatumomab has excellent and sustainable results, offering new hope for patients with minimal residual disease-positive acute lymphoblastic leukemia, a disease with poor prognosis. New recommendations and change of practice for treatment of this patient group are detailed.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Adulto , Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos/uso terapêutico , Criança , Europa (Continente) , Humanos , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Linfócitos T
3.
Oncologist ; 20(3): 329-34, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25673103

RESUMO

On August 5, 2013, a marketing authorization valid throughout the European Union (EU) was issued for pomalidomide in combination with dexamethasone for the treatment of adult patients with relapsed and refractory multiple myeloma (MM) who have received at least two prior treatment regimens, including both lenalidomide and bortezomib, and have demonstrated disease progression on the last therapy. Pomalidomide is an immunomodulating agent. The recommended starting dose of pomalidomide is 4 mg once daily taken on days 1-21 of repeated 28-day cycles. The main evidence of efficacy for pomalidomide in MM was based on a phase III multicenter, randomized, open-label study (CC-4047-MM-003) in which pomalidomide plus low-dose dexamethasone therapy (POM+LoDEX) was compared with high-dose dexamethasone alone (HiDEX) in previously treated adult patients with relapsed and refractory multiple myeloma who had received at least two prior treatment regimens, including both lenalidomide and bortezomib, and had demonstrated disease progression on the last therapy. For the intent-to-treat population, median progression-free survival based on International Myeloma Working Group criteria was 15.7 weeks (95% confidence interval [CI]: 13.0-20.1) in the POM+LoDEX group versus 8.0 weeks (95% CI: 7.0-9.0) in the HiDEX group (log-rank p value <.001). Overall survival (secondary endpoint) was also different in the two treatment groups (hazard ratio 0.53 [95% CI: 0.37-0.74]). The most commonly reported adverse reactions to pomalidomide in clinical studies were anemia (45.7%), neutropenia (45.3%) and thrombocytopenia (27%), fatigue (28.3%), pyrexia (21%), peripheral edema (13%), and infections including pneumonia (10.7%). Peripheral neuropathy adverse reactions were reported in 12.3% of patients, and venous embolic or thrombotic (VTE) adverse reactions were reported in 3.3% of patients. Pomalidomide is expected to be teratogenic. This paper summarizes the scientific review of the application leading to approval in the EU. The detailed scientific assessment report and product information, including the summary of product characteristics, are available on the EMA website (http://www.ema.europa.eu).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dexametasona/administração & dosagem , Mieloma Múltiplo/tratamento farmacológico , Talidomida/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Ácidos Borônicos/administração & dosagem , Bortezomib , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Europa (Continente) , Humanos , Lenalidomida , Recidiva Local de Neoplasia/tratamento farmacológico , Pirazinas/administração & dosagem , Taxa de Sobrevida , Talidomida/administração & dosagem
4.
Oncologist ; 19(4): 421-5, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24668331

RESUMO

On March 27, 2013, a conditional marketing authorization valid throughout the European Union was issued for bosutinib (Bosulif) for the treatment of adult patients with chronic-phase, accelerated-phase, and blast-phase Philadelphia chromosome positive (Ph⁺) chronic myelogenous leukemia (CML) previously treated with one tyrosine kinase inhibitor or more and for whom imatinib, nilotinib, and dasatinib are not considered appropriate treatment options. Bosutinib is a kinase inhibitor that targets the BCR-ABL kinase. The recommended dose is 500 mg of bosutinib once daily. The main evidence of efficacy for bosutinib was based on a CML subgroup analysis of study 3160A4-200, a phase I/II study of bosutinib in Ph⁺ leukemia in imatinib-resistant or intolerant CML. The subgroup was defined based on the presence of a BCR-ABL kinase domain mutation that would be expected to confer resistance to dasatinib (F317, E255) or nilotinib (E255, Y253, F359) and expected to have sensitivity to bosutinib or based on the presence of medical conditions or prior toxicities that may predispose the patient to unacceptable risk in the setting of nilotinib or dasatinib therapy. A conditional marketing authorization was granted because of the limited evidence of efficacy and safety currently supporting this last-line indication.


Assuntos
Compostos de Anilina/uso terapêutico , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Proteínas de Fusão bcr-abl/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Nitrilas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Quinolinas/uso terapêutico , Compostos de Anilina/efeitos adversos , Aprovação de Drogas , Resistencia a Medicamentos Antineoplásicos/genética , Europa (Continente) , União Europeia , Feminino , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/mortalidade , Masculino , Pessoa de Meia-Idade , Mutação , Nitrilas/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Quinolinas/efeitos adversos , Resultado do Tratamento
5.
Oncologist ; 16(10): 1451-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21963999

RESUMO

The product Teysuno™ (S-1) contains tegafur, a prodrug of 5-fluorouracil (5-FU), and two modulators of 5-FU metabolism, gimeracil and oteracil. The main clinical study in this application was a randomized controlled study comparing S-1 plus cisplatin with 5-FU plus cisplatin. In this study, median overall survival times of 8.6 months and 7.9 months for S-1 plus cisplatin and 5-FU plus cisplatin, respectively, were observed (hazard ratio, 0.92; 95% confidence interval, 0.80-1.05). The Committee for Medicinal Products for Human Use of the European Medicines Agency concluded that S-1 in combination with cisplatin (75 mg/m²) was noninferior to 5-FU plus cisplatin (100 mg/m²) in patients with advanced gastric cancer and adopted a positive opinion recommending the marketing authorization for this product for the treatment of advanced gastric cancer when given in combination with cisplatin. The recommended dose of S-1 is 25 mg/m² (expressed as tegafur content) twice a day, for 21 consecutive days followed by 7 days rest (one treatment cycle), in combination with 75 mg/m² cisplatin i.v. administered on day 1. This treatment cycle is repeated every 4 weeks. The most common side effects reported in the pivotal study were anemia, neutropenia, vomiting, diarrhea, abdominal pain, weight decrease, anorexia, and fatigue. The objective of this paper is to summarize the scientific review of the application leading to approval in the EU. The full scientific assessment report and the summary of product characteristics are available on the European Medicines Agency website (http://www.ema.europa.eu).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Esquema de Medicação , Aprovação de Drogas , União Europeia , Humanos , Ácido Oxônico/administração & dosagem , Ácido Oxônico/efeitos adversos , Guias de Prática Clínica como Assunto , Piridinas/administração & dosagem , Piridinas/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , Tegafur/administração & dosagem , Tegafur/efeitos adversos , Resultado do Tratamento
6.
ESMO Open ; 5(6): e000798, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33188050

RESUMO

On the 15 November 2018, the Committee for Medicinal Products for Human Use adopted an extension to an existing indication for the use of nivolumab (Opdivo) in combination with ipilimumab (Yervoy) for the first-line treatment of adult patients with intermediate/poor-risk advanced renal cell carcinoma (RCC). The approval was based on results from the Pivotal CA209214 study, a randomised, open-label, phase III study, comparing nivolumab +ipilimumab with sunitinib in subjects≥18 years of age with previously untreated advanced RCC (not amenable for surgery or radiotherapy) or metastatic RCC, with a clear-cell component. A total of 1096 patients were randomised in the trial, of which 847 patients had intermediate/poor-risk RCC and received either nivolumab (n=425) in combination with ipilimumab administered every 3 weeks for 4 doses followed by nivolumab monotherapy 3 mg/kg every 2 weeks or sunitinib (n=422) administered orally for 4 weeks followed by 2 weeks off, every cycle. A statistically significant difference in overall survival (OS) was observed in the nivolumab + ipilimumab group compared with the sunitinib group in intermediate/poor-risk subjects (HR 0.63 (99.8% CI 0.44 to 0.89); stratified log-rank 2-sided p-value<0.0001). The median OS was not reached for the nivolumab + ipilimumab group and was 25.95 months for the sunitinib group. The OS rates were 89.5% and 86.2% at 6 months, and 80.1% and 72.1% at 12 months in the nivolumab +ipilimumab and the sunitinib groups, respectively. K-M curves separated after approximately 3 months, favouring nivolumab + ipilimumab. This was not mirrored in the favourable-risk patients where no statistically significant difference was observed between nivolumab + ipilimumab and sunitinib in favourable-risk patients (HR 1.45 (descriptive 99.8% CI 0.51 to 4.12), p =0.2715).


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma de Células Renais/tratamento farmacológico , Humanos , Imunoterapia , Ipilimumab/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Nivolumabe/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto
7.
ESMO Open ; 5(4)2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32847837

RESUMO

The therapeutic landscape in the treatment of advanced/metastatic renal cell cancer has evolved over the last 2 years with the advent of immune checkpoint inhibitors. In 2018 and 2019, marketing authorisations valid throughout the European Union were issued for nivolumab and ipilimumab dual checkpoint inhibition and pembrolizumab or avelumab in combination with the tyrosine kinase inhibitor axitinib. These applications presented numerous regulatory challenges.In this paper, we summarise the main regulatory considerations, originating from the assessment of the dossiers submitted from the applicants for the three combinations. The regulatory issues are grouped in four sections: clinical pharmacology, efficacy, biomarkers and safety. In each section, we describe the issues raised during the regulatory evaluation performed by the Committee for Medicinal Products for Human Use (CHMP) assessors. The CHMP assessments determine whether the medicines concerned meet the necessary quality, safety and efficacy requirements, and whether the benefit-risk balance is positive.In summary, although the overall benefit-risk was considered positive for the three combinations, the immaturity of the outcome data and the absence of long-term safety data remain issues to be addressed. Postauthorisation efficacy studies have been required to confirm the effects of the new combinations.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Axitinibe , Humanos , Ipilimumab , Nivolumabe
8.
Br J Pharmacol ; 148(6): 778-85, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16770327

RESUMO

1. 3,4-Methylenedioxymethamphetamine (MDMA or 'ecstasy') decreases the 5-HT concentration, [3H]-paroxetine binding and tryptophan hydroxylase activity in rat forebrain, which has been interpreted as indicating 5-HT neurodegeneration. This has been questioned, particularly the 5-HT loss, as MDMA can also inhibit tryptophan hydroxylase. We have now evaluated the validity of these parameters as a reflection of neurotoxicity. 2. Male DA rats were administered MDMA (12.5 mg kg(-1), i.p.) and killed up to 32 weeks later. 5-HT content and [3H]-paroxetine binding were measured in the cortex, hippocampus and striatum. Parallel groups of treated animals were administered NSD-1015 for determination of in vivo tryptophan hydroxylase activity and 5-HT turnover rate constant. 3. Tissue 5-HT content and [3H]-paroxetine binding were reduced in the cortex (26-53%) and hippocampus (25-74%) at all time points (1, 2, 4, 8 and 32 weeks). Hydroxylase activity was similarly reduced up to 8 weeks, but had recovered at 32 weeks. The striatal 5-HT concentration and [3H]-paroxetine binding recovered by week 4 and hydroxylase activity after week 1. In all regions, the reduction in 5-HT concentration did not result in an altered 5-HT synthesis rate constant. 4. Administering MDMA to animals when housed at 4 degrees C prevented the reduction in [3H]-paroxetine binding and hydroxylase activity observed in rats housed at 22 degrees C, but not the reduction in 5-HT concentration. 5. These data indicate that MDMA produces long-term damage to serotoninergic neurones, but this does not produce a compensatory increase in 5-HT synthesis in remaining terminals. It also highlights the fact that measurement of tissue 5-HT concentration may overestimate neurotoxic damage.


Assuntos
Encéfalo/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Serotonina/biossíntese , Animais , Temperatura Corporal/efeitos dos fármacos , Encéfalo/metabolismo , Ácido Hidroxi-Indolacético/análise , Masculino , Paroxetina/metabolismo , Ratos , Serotonina/análise , Temperatura , Triptofano Hidroxilase/metabolismo
10.
Hum Vaccin Immunother ; 8(9): 1354-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22863755

RESUMO

Active immunotherapy products (widely known as "cancer vaccines") are products intended to stimulate an immune response to mediate tumor destruction or reduce the progression of disease in patients where cancer has been diagnosed. Some quality attributes of these products are very difficult to characterize or present a high variability (especially if they are for autologous use), further complicating the interpretation of some of the clinical data. Furthermore, questions arise in the evaluation of efficacy and safety data in comparison with current chemical or biological treatments for the same indications. Some of these aspects are discussed in this paper in relationship with the regulatory requirements in the European Union and as applied to two recently assessed medicinal products, Oncophage and Provenge, both considered therapeutic "cancer vaccines" for renal cell carcinoma and prostate cancer, respectively.


Assuntos
Vacinas Anticâncer/uso terapêutico , União Europeia/estatística & dados numéricos , Humanos , Imunoterapia/legislação & jurisprudência , Imunoterapia/normas
11.
Eur J Cancer ; 48(2): 237-42, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22030452

RESUMO

On 13 July 2011 the European Commission issued a marketing authorisation valid throughout the European Union (EU) for ipilimumab for the treatment of advanced (unresectable or metastatic) melanoma in adults who have received prior therapy. Ipilimumab is a monoclonal antibody that specifically blocks the inhibitory signal of cytotoxic T lymphocyte antigen 4 (CTLA-4), resulting in T cell activation, proliferation and lymphocyte infiltration into tumours, leading to tumour cell death. The recommended induction regimen of ipilimumab is 3mg/kg administered intravenously over a 90 min period every 3 weeks for a total of four doses. In a phase 3 trial in patients with advanced melanoma, median overall survival for ipilimumab was 10 months versus 6 months for gp100, an experimental melanoma vaccine (Hazard ratio (HR) 0.66; 95% confidence interval (CI): 0.51, 0.87; p = 0.0026). Ipilimumab was most commonly associated with adverse reactions resulting from increased or excessive immune activity. Most of these, including severe reactions, resolved following initiation of appropriate medical therapy or withdrawal of ipilimumab. The most common side-effects (affecting more than 10% of patients) were diarrhoea, rash, pruritus, fatigue, nausea, vomiting, decreased appetite and abdominal pain. The objective of this paper is to summarise the scientific review of the application leading to approval in the EU. The detailed scientific assessment report and product information, including the summary of product characteristics (SmPC), are available on the European Medicines Agency (EMA) website (www.ema.europa.eu).


Assuntos
Anticorpos Monoclonais/uso terapêutico , Fatores Imunológicos/uso terapêutico , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , União Europeia , Humanos , Ipilimumab , Melanoma/imunologia , Melanoma/secundário , Neoplasias Cutâneas/imunologia
13.
Enferm Intensiva ; 20(4): 159-66, 2009.
Artigo em Espanhol | MEDLINE | ID: mdl-20038384

RESUMO

Adequate monitoring of analgesia and sedation should be one of the main goals in the Intensive Care Units. Once the analgesia is assured, the correct dosage of the sedatives will depend on the correct monitoring of the sedation level. For years, clinical scales have been used to evaluate the sedation level. These scales are appropriate for the evaluation of mild sedation but they cannot correctly evaluate deeply sedated patients or those patients who require continuous intravenous administration of neuromuscular blockade agents. In this chapter, we review the most used monitor to control the adequate level of sedation in the Intensive Care Units as well as the BIS(R) Monitor, which is the one recommended by the Analgesia and Sedation Work Group of the Spanish Society of Critical Care Medicine (SEMICYUC).


Assuntos
Monitores de Consciência , Sedação Profunda , Desenho de Equipamento , Humanos , Monitorização Intraoperatória/instrumentação , Monitorização Intraoperatória/métodos
14.
Transl Androl Urol ; 2(2): 89-91, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26816730
15.
J Neurochem ; 81(5): 961-72, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12065608

RESUMO

The present study examined the mechanisms by which 3,4-methylenedioxymethamphetamine (MDMA) produces long-term neurotoxicity of striatal dopamine neurones in mice and the protective action of the dopamine uptake inhibitor GBR 12909. MDMA (30 mg/kg, i.p.), given three times at 3-h intervals, produced a rapid increase in striatal dopamine release measured by in vivo microdialysis (maximum increase to 380 +/- 64% of baseline). This increase was enhanced to 576 +/- 109% of baseline by GBR 12909 (10 mg/kg, i.p.) administered 30 min before each dose of MDMA, supporting the contention that MDMA enters the terminal by diffusion and not via the dopamine uptake site. This, in addition to the fact that perfusion of the probe with a low Ca(2+) medium inhibited the MDMA-induced increase in extracellular dopamine, indicates that the neurotransmitter may be released by a Ca(2+) -dependent mechanism not related to the dopamine transporter. MDMA (30 mg/kg x 3) increased the formation of 2,3-dihydroxybenzoic acid (2,3-DHBA) from salicylic acid perfused through a probe implanted in the striatum, indicating that MDMA increased free radical formation. GBR 12909 pre-treatment attenuated the MDMA-induced increase in 2,3-DHBA formation by approximately 50%, but had no significant intrinsic radical trapping activity. MDMA administration increased lipid peroxidation in striatal synaptosomes, an effect reduced by approximately 60% by GBR 12909 pre-treatment. GBR 12909 did not modify the MDMA-induced changes in body temperature. These data suggest that MDMA-induced toxicity of dopamine neurones in mice results from free radical formation which in turn induces an oxidative stress process. The data also indicate that the free radical formation is probably not associated with the MDMA-induced dopamine release and that MDMA does not induce dopamine release via an action at the dopamine transporter.


Assuntos
Corpo Estriado/efeitos dos fármacos , Inibidores da Captação de Dopamina/farmacologia , Dopamina/metabolismo , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Piperazinas/farmacologia , Animais , Ácido Ascórbico/química , Temperatura Corporal/efeitos dos fármacos , Cálcio/metabolismo , Corpo Estriado/química , Corpo Estriado/metabolismo , Dopamina/análise , Inibidores da Captação de Dopamina/análise , Compostos Ferrosos/química , Radicais Livres/metabolismo , Hidroxibenzoatos/análise , Hidroxibenzoatos/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Microdiálise , Piperazinas/análise , Ratos , Ácido Salicílico/metabolismo , Sinaptossomos/química , Fatores de Tempo
16.
J Neurochem ; 85(2): 515-24, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12675928

RESUMO

The present study examined the time-course and regionally-selective changes in the levels of the neurofilament protein NF68 in the mouse brain induced by methamphetamine (METH). The ability of low ambient temperature, or of the specific neuronal nitric oxide synthase (nNOS) inhibitor AR-R17477AR, to protect against both long-term striatal NF68 and dopamine loss induced by METH (3 mg/kg, i.p.) was also studied. Seven days after METH administration (3, 6 and 9 mg/kg, i.p., three times at 3 h intervals), mice showed a reduction of about 40% in immunoreactivity for NF68 in the striatum. This effect was not produced in cortex after METH administration at the dose of 3 mg/kg. No difference from controls was observed when measurements were carried out 1 h and 24 h after the last METH injection at the dose of 3 mg/kg. The loss of NF68 immunoreactivity seems to be associated with the long-term dopamine depletion induced by METH, since no change in serotonin concentration is observed in either the striatum or cortex 7 days after dosing. Animals kept at a room temperature of 4 degrees C showed a loss of NF68 similar to those treated at 22 degrees C but an attenuation of dopamine depletion in the striatum. Pre-treatment with AR-R17477AR (5 mg/kg, s.c.) 30 min before each of the three METH (3 mg/kg, i.p.) injections provided complete protection against METH-induced loss of NF68 immunoreactivity and attenuated the decrease in striatal dopamine and HVA concentrations by about 50%. These data indicate that both the reduction of NF68 immunoreactivity and the loss of dopamine concentration are due to an oxidative stress process mediated by reactive nitrogen species, and are not due to changes in body temperature.


Assuntos
Corpo Estriado/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Metanfetamina/farmacologia , Proteínas de Neurofilamentos/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Tirosina/análogos & derivados , Animais , Temperatura Corporal/efeitos dos fármacos , Química Encefálica , Corpo Estriado/metabolismo , Dopamina/metabolismo , Dopaminérgicos/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Febre/induzido quimicamente , Ácido Homovanílico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neurofilamentos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I , Estresse Oxidativo/efeitos dos fármacos , Ácido Peroxinitroso/metabolismo , Temperatura , Tirosina/análise
17.
J Neurochem ; 89(6): 1445-53, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15189347

RESUMO

3,4-Methylenedioxymethamphetamine (MDMA) administration to rats produces acute hyperthermia and 5-HT release. Interleukin-1beta (IL-1beta) is a pro-inflammatory pyrogen produced by activated microglia in the brain. We examined the effect of a neurotoxic dose of MDMA on IL-1beta concentration and glial activation and their relationship with acute hyperthermia and 5-HT depletion. MDMA, given to rats housed at 22 degrees C, increased IL-1beta levels in hypothalamus and cortex from 1 to 6 h and [(3)H]-(1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)3-isoquinolinecarboxamide) binding between 3 and 48 h. Increased immunoreactivity to OX-42 was also detected. Rats became hyperthermic immediately after MDMA and up to at least 12 h later. The IL-1 receptor antagonist did not modify MDMA-induced hyperthermia indicating that IL-1beta release is a consequence, not the cause, of the rise in body temperature. When MDMA was given to rats housed at 4 degrees C, hyperthermia was abolished and the IL-1beta increase significantly reduced. The MDMA-induced acute 5-HT depletion was prevented by fluoxetine coadministration but the IL-1beta increase and hyperthermia were unaffected. Therefore, the rise in IL-1beta is not related to the acute 5-HT release but is linked to the hyperthermia. Contrary to IL-1beta levels, microglial activation is not significantly modified when hyperthermia is prevented, suggesting that it might be a process not dependent on the hyperthermic response induced by MDMA.


Assuntos
Encéfalo/efeitos dos fármacos , Febre/induzido quimicamente , Interleucina-1/metabolismo , Microglia/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Serotonina/metabolismo , Doença Aguda , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Temperatura Corporal/efeitos dos fármacos , Encéfalo/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Febre/tratamento farmacológico , Febre/metabolismo , Fluoxetina/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Proteína Antagonista do Receptor de Interleucina 1 , Isoquinolinas/farmacocinética , Masculino , Microglia/metabolismo , Ratos , Ratos Endogâmicos , Serotonina/deficiência , Serotoninérgicos/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sialoglicoproteínas/farmacologia , Temperatura
18.
Enferm. intensiva (Ed. impr.) ; 20(4): 159-166, oct.-dic. 2009. ilus, tab
Artigo em Espanhol | IBECS (Espanha) | ID: ibc-80340

RESUMO

La correcta monitorización de la analgesia y la sedación debe ser uno de los objetivosprincipales en las Unidades de Cuidados Intensivos. Una vez asegurada la analgesia, lacorrecta dosificación de los sedantes va a depender de la monitorización correcta delnivel de sedación. Desde hace años se han utilizado sistemas de monitorización basadosen escalas. Estas escalas son adecuadas para la monitorización de la sedación superficial,pero no son capaces de evaluar el grado de sedación en pacientes profundamente sedadoso con bloqueantes neuromusculares en perfusión continua. Actualmente disponemosde diferentes sistemas de monitorización que nos facilitan llegar donde las escalas noalcanzan. En este capítulo revisaremos el monitor más empleado en la actualidad en lasUnidades de Cuidados Intensivos, y el recomendado por el grupo de trabajo de Sedacióny Analgesia de la Sociedad Española de Medicina Intensiva, Crítica y Unidades Coronarias(SEMICYUC) para la monitorización de la sedación profunda, el Monitor BIS® (AU)


Adequate monitoring of analgesia and sedation should be one of the main goals in the IntensiveCare Units. Once the analgesia is assured, the correct dosage of the sedatives will dependon the correct monitoring of the sedation level. For years, clinical scales have beenused to evaluate the sedation level. These scales are appropriate for the evaluation of mildsedation but they cannot correctly evaluate deeply sedated patients or those patients whorequire continuous intravenous administration of neuromuscular blockade agents. In thischapter, we review the most used monitor to control the adequate level of sedation in theIntensive Care Units as well as the BIS® Monitor, which is the one recommended by the Analgesiaand Sedation Work Group of the Spanish Society of Critical Care Medicine (SEMICYUC) (AU)


Assuntos
Humanos , Monitorização Fisiológica/instrumentação , Unidades de Terapia Intensiva , Sedação Profunda
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa