Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38926092

RESUMO

Radiographic assessment plays a crucial role in the management of patients with central nervous system (CNS) tumors, aiding in treatment planning and evaluation of therapeutic efficacy by quantifying response. Recently, an updated version of the Response Assessment in Neuro-Oncology (RANO) criteria (RANO 2.0) was developed to improve upon prior criteria and provide an updated, standardized framework for assessing treatment response in clinical trials for gliomas in adults. This article provides an overview of significant updates to the criteria including (1) the use of a unified set of criteria for high and low grade gliomas in adults; (2) the use of the post-radiotherapy MRI scan as the baseline for evaluation in newly diagnosed high-grade gliomas; (3) the option for the trial to mandate a confirmation scan to more reliably distinguish pseudoprogression from tumor progression; (4) the option of using volumetric tumor measurements; and (5) the removal of subjective non-enhancing tumor evaluations in predominantly enhancing gliomas (except for specific therapeutic modalities). Step-by-step pragmatic guidance is hereby provided for the neuroradiologist and imaging core lab involved in operationalization and technical execution of RANO 2.0 in clinical trials, including the display of representative cases and in-depth discussion of challenging scenarios.ABBREVIATIONS: BTIP = Brain Tumor Imaging Protocol; CE = Contrast-Enhancing; CNS = Central Nervous System; CR = Complete Response; ECOG = Eastern Cooperative Oncology Group; HGG = High-Grade Glioma; IDH = Isocitrate Dehydrogenase; IRF = Independent Radiologic Facility; LGG = Low-Grade Glioma; KPS = Karnofsky Performance Status; MR = Minor Response; mRANO = Modified RANO; NANO = Neurological Assessment in Neuro-Oncology; ORR = Objective Response Rate; OS = Overall Survival; PD = Progressive Disease; PFS = Progression-Free Survival; PR = Partial Response; PsP = Pseudoprogression; RANO = Response Assessment in Neuro-Oncology; RECIST = Response Evaluation Criteria In Solid Tumors; RT = Radiation Therapy; SD = Stable Disease; Tx = Treatment.

2.
J Clin Oncol ; 41(33): 5187-5199, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37774317

RESUMO

PURPOSE: The Response Assessment in Neuro-Oncology (RANO) criteria for high-grade gliomas (RANO-HGG) and low-grade gliomas (RANO-LGG) were developed to improve reliability of response assessment in glioma trials. Over time, some limitations of these criteria were identified, and challenges emerged regarding integrating features of the modified RANO (mRANO) or the immunotherapy RANO (iRANO) criteria. METHODS: Informed by data from studies evaluating the different criteria, updates to the RANO criteria are proposed (RANO 2.0). RESULTS: We recommend a standard set of criteria for both high- and low-grade gliomas, to be used for all trials regardless of the treatment modalities being evaluated. In the newly diagnosed setting, the postradiotherapy magnetic resonance imaging (MRI), rather than the postsurgical MRI, will be used as the baseline for comparison with subsequent scans. Since the incidence of pseudoprogression is high in the 12 weeks after radiotherapy, continuation of treatment and confirmation of progression during this period with a repeat MRI, or histopathologic evidence of unequivocal recurrent tumor, are required to define tumor progression. However, confirmation scans are not mandatory after this period nor for the evaluation of treatment for recurrent tumors. For treatments with a high likelihood of pseudoprogression, mandatory confirmation of progression with a repeat MRI is highly recommended. The primary measurement remains the maximum cross-sectional area of tumor (two-dimensional) but volumetric measurements are an option. For IDH wild-type glioblastoma, the nonenhancing disease will no longer be evaluated except when assessing response to antiangiogenic agents. In IDH-mutated tumors with a significant nonenhancing component, clinical trials may require evaluating both the enhancing and nonenhancing tumor components for response assessment. CONCLUSION: The revised RANO 2.0 criteria refine response assessment in gliomas.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Adulto , Neoplasias Encefálicas/tratamento farmacológico , Reprodutibilidade dos Testes , Recidiva Local de Neoplasia , Glioma/patologia , Imageamento por Ressonância Magnética/métodos
3.
Lancet Digit Health ; 3(12): e784-e794, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34688602

RESUMO

BACKGROUND: Gadolinium-based contrast agents (GBCAs) are widely used to enhance tissue contrast during MRI scans and play a crucial role in the management of patients with cancer. However, studies have shown gadolinium deposition in the brain after repeated GBCA administration with yet unknown clinical significance. We aimed to assess the feasibility and diagnostic value of synthetic post-contrast T1-weighted MRI generated from pre-contrast MRI sequences through deep convolutional neural networks (dCNN) for tumour response assessment in neuro-oncology. METHODS: In this multicentre, retrospective cohort study, we used MRI examinations to train and validate a dCNN for synthesising post-contrast T1-weighted sequences from pre-contrast T1-weighted, T2-weighted, and fluid-attenuated inversion recovery sequences. We used MRI scans with availability of these sequences from 775 patients with glioblastoma treated at Heidelberg University Hospital, Heidelberg, Germany (775 MRI examinations); 260 patients who participated in the phase 2 CORE trial (1083 MRI examinations, 59 institutions); and 505 patients who participated in the phase 3 CENTRIC trial (3147 MRI examinations, 149 institutions). Separate training runs to rank the importance of individual sequences and (for a subset) diffusion-weighted imaging were conducted. Independent testing was performed on MRI data from the phase 2 and phase 3 EORTC-26101 trial (521 patients, 1924 MRI examinations, 32 institutions). The similarity between synthetic and true contrast enhancement on post-contrast T1-weighted MRI was quantified using the structural similarity index measure (SSIM). Automated tumour segmentation and volumetric tumour response assessment based on synthetic versus true post-contrast T1-weighted sequences was performed in the EORTC-26101 trial and agreement was assessed with Kaplan-Meier plots. FINDINGS: The median SSIM score for predicting contrast enhancement on synthetic post-contrast T1-weighted sequences in the EORTC-26101 test set was 0·818 (95% CI 0·817-0·820). Segmentation of the contrast-enhancing tumour from synthetic post-contrast T1-weighted sequences yielded a median tumour volume of 6·31 cm3 (5·60 to 7·14), thereby underestimating the true tumour volume by a median of -0·48 cm3 (-0·37 to -0·76) with the concordance correlation coefficient suggesting a strong linear association between tumour volumes derived from synthetic versus true post-contrast T1-weighted sequences (0·782, 0·751-0·807, p<0·0001). Volumetric tumour response assessment in the EORTC-26101 trial showed a median time to progression of 4·2 months (95% CI 4·1-5·2) with synthetic post-contrast T1-weighted and 4·3 months (4·1-5·5) with true post-contrast T1-weighted sequences (p=0·33). The strength of the association between the time to progression as a surrogate endpoint for predicting the patients' overall survival in the EORTC-26101 cohort was similar when derived from synthetic post-contrast T1-weighted sequences (hazard ratio of 1·749, 95% CI 1·282-2·387, p=0·0004) and model C-index (0·667, 0·622-0·708) versus true post-contrast T1-weighted MRI (1·799, 95% CI 1·314-2·464, p=0·0003) and model C-index (0·673, 95% CI 0·626-0·711). INTERPRETATION: Generating synthetic post-contrast T1-weighted MRI from pre-contrast MRI using dCNN is feasible and quantification of the contrast-enhancing tumour burden from synthetic post-contrast T1-weighted MRI allows assessment of the patient's response to treatment with no significant difference by comparison with true post-contrast T1-weighted sequences with administration of GBCAs. This finding could guide the application of dCNN in radiology to potentially reduce the necessity of GBCA administration. FUNDING: Deutsche Forschungsgemeinschaft.


Assuntos
Neoplasias Encefálicas/diagnóstico , Encéfalo/patologia , Meios de Contraste/administração & dosagem , Aprendizado Profundo , Gadolínio/administração & dosagem , Imageamento por Ressonância Magnética/métodos , Redes Neurais de Computação , Algoritmos , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Imagem de Difusão por Ressonância Magnética , Progressão da Doença , Estudos de Viabilidade , Alemanha , Glioblastoma/diagnóstico , Glioblastoma/diagnóstico por imagem , Humanos , Pessoa de Meia-Idade , Neoplasias , Prognóstico , Radiologia/métodos , Estudos Retrospectivos , Carga Tumoral
4.
Eur J Cancer ; 116: 190-198, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31203194

RESUMO

OBJECTIVE: Prognostic value of health-related quality of life (HRQoL) data may be important to inform patients in clinical practice and to guide clinical decision-making. Our study investigated the added prognostic value of HRQoL for overall survival (OS) and progression-free survival (PFS) in a large heterogeneous sample of glioma patients, besides known prognostic factors. METHODS: We included individual baseline data from previously published randomised controlled trials (RCTs) in glioma patients in which HRQoL was assessed through the European Organisation for Research and Treatment of Cancer QLQ-C30 and QLQ-BN20 questionnaires. Multivariable Cox regression models (stratified for newly diagnosed versus recurrent disease) were constructed, first with clinical variables (age, sex, tumour type, performance status, allocated treatment and extent of resection) only and subsequently with HRQoL variables added, separately for OS and PFS. The added prognostic value of HRQoL was calculated using C-indices. RESULTS: Baseline HRQoL and clinical data from 15 RCTs were included, comprising 5217 patients. In the model including both clinical and HRQoL variables, better cognitive and role functioning and less motor dysfunction were independently associated with longer OS, whereas better role and cognitive functioning, less nausea and vomiting and more appetite loss were independently associated with prolonged PFS. However, C-indices indicated only a small prognostic improvement of the models for OS and PFS when adding HRQoL to the clinical prognostic variables (+1.1% for OS and +.7% for PFS). CONCLUSION: Our findings demonstrate that several baseline HRQoL variables are independently prognostic for OS and PFS, yet the added value of HRQoL to the known clinical prognostic variables was small.


Assuntos
Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/mortalidade , Glioma/complicações , Glioma/mortalidade , Qualidade de Vida , Neoplasias Encefálicas/psicologia , Glioma/psicologia , Nível de Saúde , Humanos , Prognóstico , Intervalo Livre de Progressão , Ensaios Clínicos Controlados Aleatórios como Assunto
5.
Neuro Oncol ; 19(5): 625-635, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28453751

RESUMO

Background: The Macdonald criteria and the Response Assessment in Neuro-Oncology (RANO) criteria define radiologic parameters to classify therapeutic outcome among patients with malignant glioma and specify that clinical status must be incorporated and prioritized for overall assessment. But neither provides specific parameters to do so. We hypothesized that a standardized metric to measure neurologic function will permit more effective overall response assessment in neuro-oncology. Methods: An international group of physicians including neurologists, medical oncologists, radiation oncologists, and neurosurgeons with expertise in neuro-oncology drafted the Neurologic Assessment in Neuro-Oncology (NANO) scale as an objective and quantifiable metric of neurologic function evaluable during a routine office examination. The scale was subsequently tested in a multicenter study to determine its overall reliability, inter-observer variability, and feasibility. Results: The NANO scale is a quantifiable evaluation of 9 relevant neurologic domains based on direct observation and testing conducted during routine office visits. The score defines overall response criteria. A prospective, multinational study noted a >90% inter-observer agreement rate with kappa statistic ranging from 0.35 to 0.83 (fair to almost perfect agreement), and a median assessment time of 4 minutes (interquartile range, 3-5). Conclusion: The NANO scale provides an objective clinician-reported outcome of neurologic function with high inter-observer agreement. It is designed to combine with radiographic assessment to provide an overall assessment of outcome for neuro-oncology patients in clinical trials and in daily practice. Furthermore, it complements existing patient-reported outcomes and cognition testing to combine for a global clinical outcome assessment of well-being among brain tumor patients.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/terapia , Neuroimagem/métodos , Neoplasias Encefálicas/patologia , Humanos , Resultado do Tratamento
6.
Lancet Oncol ; 15(2): e78-89, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24480558

RESUMO

Small-cell lung cancer represents about 15% of all lung cancers; increasingly, randomised controlled trials of this disease measure the health-related quality of life of patients. In this Systematic Review we assess the adequacy of reporting of health-related quality-of-life methods in randomised controlled trials of small-cell lung cancer, and the potential effect of this reporting on clinical decision making. Although overall reporting of health-related quality of life was acceptable, improvements are needed to optimise the use of health-related quality of life in randomised controlled trials.


Assuntos
Neoplasias Pulmonares/psicologia , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Projetos de Pesquisa , Carcinoma de Pequenas Células do Pulmão/psicologia , Atividades Cotidianas , Adaptação Psicológica , Efeitos Psicossociais da Doença , Medicina Baseada em Evidências , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/terapia , Prognóstico , Carcinoma de Pequenas Células do Pulmão/diagnóstico , Carcinoma de Pequenas Células do Pulmão/terapia , Inquéritos e Questionários
7.
Future Oncol ; 7(3): 339-54, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21417900

RESUMO

Cilengitide, a cyclicized arginine-glycine-aspartic acid-containing pentapeptide, potently blocks ανß3 and ανß5 integrin activation. Integrins are upregulated in many malignancies and mediate a wide variety of tumor-stroma interactions. Cilengitide and other integrin-targeting therapeutics have preclinical activity against many cancer subtypes including glioblastoma (GBM), the most common and deadliest CNS tumor. Cilengitide is active against orthotopic GBM xenografts and can augment radiotherapy and chemotherapy in these models. In Phase I and II GBM trials, cilengitide and the combination of cilengitide with standard temozolomide and radiation demonstrate consistent antitumor activity and a favorable safety profile. Cilengitide is currently under evaluation in a pivotal, randomized Phase III study (Cilengitide in Combination With Temozolomide and Radiotherapy in Newly Diagnosed Glioblastoma Phase III Randomized Clinical Trial [CENTRIC]) for newly diagnosed GBM. In addition, randomized controlled Phase II studies with cilengitide are ongoing for non-small-cell lung cancer and squamous cell carcinoma of the head and neck. Cilengitide is the first integrin inhibitor in clinical Phase III development for oncology.


Assuntos
Antineoplásicos/uso terapêutico , Glioblastoma/tratamento farmacológico , Integrinas/antagonistas & inibidores , Oligopeptídeos/uso terapêutico , Venenos de Serpentes/uso terapêutico , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/farmacocinética , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Indústria Farmacêutica/tendências , Humanos , Oligopeptídeos/efeitos adversos , Oligopeptídeos/química , Oligopeptídeos/farmacocinética , Venenos de Serpentes/efeitos adversos , Venenos de Serpentes/química , Venenos de Serpentes/farmacocinética , Resultado do Tratamento
8.
J Clin Oncol ; 28(11): 1963-72, 2010 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-20231676

RESUMO

Currently, the most widely used criteria for assessing response to therapy in high-grade gliomas are based on two-dimensional tumor measurements on computed tomography (CT) or magnetic resonance imaging (MRI), in conjunction with clinical assessment and corticosteroid dose (the Macdonald Criteria). It is increasingly apparent that there are significant limitations to these criteria, which only address the contrast-enhancing component of the tumor. For example, chemoradiotherapy for newly diagnosed glioblastomas results in transient increase in tumor enhancement (pseudoprogression) in 20% to 30% of patients, which is difficult to differentiate from true tumor progression. Antiangiogenic agents produce high radiographic response rates, as defined by a rapid decrease in contrast enhancement on CT/MRI that occurs within days of initiation of treatment and that is partly a result of reduced vascular permeability to contrast agents rather than a true antitumor effect. In addition, a subset of patients treated with antiangiogenic agents develop tumor recurrence characterized by an increase in the nonenhancing component depicted on T2-weighted/fluid-attenuated inversion recovery sequences. The recognition that contrast enhancement is nonspecific and may not always be a true surrogate of tumor response and the need to account for the nonenhancing component of the tumor mandate that new criteria be developed and validated to permit accurate assessment of the efficacy of novel therapies. The Response Assessment in Neuro-Oncology Working Group is an international effort to develop new standardized response criteria for clinical trials in brain tumors. In this proposal, we present the recommendations for updated response criteria for high-grade gliomas.


Assuntos
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Diagnóstico por Imagem/normas , Glioma/diagnóstico , Glioma/terapia , Ensaios Clínicos como Assunto , Diagnóstico por Imagem/métodos , Guias como Assunto , Humanos , Prognóstico , Resultado do Tratamento
9.
Expert Rev Pharmacoecon Outcomes Res ; 9(3): 235-41, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19527095

RESUMO

AIM: To perform a systematic review on the costs and cost-effectiveness of concomitant and adjuvant temozolomide with radiotherapy for the treatment of newly diagnosed glioblastoma compared with initial radiotherapy alone. METHODS: Electronic databases were searched for relevant publications on costs and cost-effectiveness until October 2008. RESULTS: We found four relevant clinical trials, one cost study and two economic models. The mean survival benefit in the radiotherapy plus temozolomide group varied between 0.21 and 0.25 life-years. Treatment costs were between 27,365 euros and 39,092 euros. The costs of temozolomide amounted to approximately 40% of the total treatment costs. The incremental cost-effectiveness ratios found in the literature were 37,361 euros per life-year gained and 42,912 euros per quality-adjusted life-year gained. However, the models are not comparable because different outcomes are used (i.e., life-years and quality-adjusted life-years). CONCLUSION: Although the models are not comparable according to outcome, the incremental cost-effectiveness ratios found are within acceptable ranges. We concluded that despite the high temozolomide acquisition costs, the costs per life-year gained and the costs per quality-adjusted life-year gained are comparable with other accepted first-line treatments with chemotherapy in patients with cancer.


Assuntos
Antineoplásicos Alquilantes/economia , Dacarbazina/análogos & derivados , Glioblastoma/economia , Antineoplásicos Alquilantes/uso terapêutico , Ensaios Clínicos como Assunto , Terapia Combinada , Análise Custo-Benefício , Dacarbazina/economia , Dacarbazina/uso terapêutico , Custos de Medicamentos , Glioblastoma/mortalidade , Glioblastoma/terapia , Humanos , Modelos Econômicos , Anos de Vida Ajustados por Qualidade de Vida , Análise de Sobrevida , Temozolomida , Resultado do Tratamento
10.
Lung Cancer ; 61(3): 398-404, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18433927

RESUMO

BACKGROUND: In patients with malignant pleural mesothelioma undergoing a multimodality therapy, treatment toxicity may outweigh the benefit of progression-free survival. The subjective experience across different treatment phases is an important clinical outcome. This study compares a standard with an individual quality of life (QoL) measure used in a multi-center phase II trial. PATIENTS AND METHODS: Sixty-one patients with stage I-III technically operable pleural mesothelioma were treated with preoperative chemotherapy, followed by pleuropneumonectomy and subsequent radiotherapy. QoL was assessed at baseline, at day 1 of cycle 3, and 1, 3 and 6 months post-surgery by using the Rotterdam Symptom Checklist (RSCL) and the Schedule for the Evaluation of Quality of Life-Direct Weighting (SEIQoL-DW), a measure that is based on five individually nominated and weighted QoL-domains. RESULTS: Completion rates were 98% (RSCL) and 92% (SEIQoL) at baseline and 98%/89% at cycle 3, respectively. Of the operated patients (N=45) RSCL and SEIQoL were available from 86%/72%, 93%/74%, and 94%/76% at months 1, 3, and 6 post-surgery. Average assessment time for the SEIQoL was 24min compared to 8min needed for the RSCL. Median changes from baseline indicate that both RSCL QoL overall score and SEIQoL index remained stable during chemotherapy with a clinically significant deterioration (change>or=8 points) 1 month after surgery (median change of -66 and -14 for RSCL and SEIQoL, respectively). RSCL QoL overall scores improved thereafter, but remained beneath baseline level until 6 months after surgery. SEIQoL scores improved to baseline-level at month 3 after surgery, but worsened again at month 6. RSCL QoL overall score and SEIQoL index were moderately correlated at baseline (r=.30; p

Assuntos
Mesotelioma/patologia , Neoplasias Pleurais/patologia , Qualidade de Vida , Adulto , Idoso , Terapia Combinada , Progressão da Doença , Intervalo Livre de Doença , Estudos de Viabilidade , Feminino , Humanos , Masculino , Mesotelioma/terapia , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Pleurais/terapia
11.
Cancer ; 112(6): 1337-44, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18213621

RESUMO

BACKGROUND: The study aimed to compare the cost-effectiveness of concomitant and adjuvant temozolomide (TMZ) for the treatment of newly diagnosed glioblastoma multiforme versus initial radiotherapy alone from a public health care perspective. METHODS: The economic evaluation was performed alongside a randomized, multicenter, phase 3 trial. The primary endpoint of the trial was overall survival. Costs included all direct medical costs. Economic data were collected prospectively for a subgroup of 219 patients (38%). Unit costs for drugs, procedures, laboratory and imaging, radiotherapy, and hospital costs per day were collected from the official national reimbursement lists based on 2004. For the cost-effectiveness analysis, survival was expressed as 2.5 years restricted mean estimates. The incremental cost-effectiveness ratio (ICER) was constructed. Confidence intervals for the ICER were calculated using the Fieller method and bootstrapping. RESULTS: The difference in 2.5 years restricted mean survival between the treatment arms was 0.25 life-years and the ICER was euro37,361 per life-year gained with a 95% confidence interval (CI) ranging from euro19,544 to euro123,616. The area between the survival curves of the treatment arms suggests an increase of the overall survival gain for a longer follow-up. An extrapolation of the overall survival per treatment arm and imputation of costs for the extrapolated survival showed a substantial reduction in ICER. CONCLUSIONS: The ICER of euro37,361 per life-year gained is a conservative estimate. We concluded that despite the high TMZ acquisition costs, the costs per life-year gained are comparable to accepted first-line treatment with chemotherapy in patients with cancer.


Assuntos
Antineoplásicos Alquilantes/economia , Neoplasias Encefálicas/economia , Dacarbazina/análogos & derivados , Glioblastoma/economia , Adolescente , Adulto , Idoso , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Análise Custo-Benefício , Dacarbazina/economia , Dacarbazina/uso terapêutico , Intervalo Livre de Doença , Seguimentos , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Custos de Cuidados de Saúde , Humanos , Pessoa de Meia-Idade , Anos de Vida Ajustados por Qualidade de Vida , Taxa de Sobrevida , Temozolomida
12.
Neuro Oncol ; 7(2): 189-95, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15831237

RESUMO

Effectiveness and costs of care and treatment of recurrent malignant gliomas are largely unknown. In this study, 49 patients (32 males, 17 females; mean age, 49; age range, 23-79) were treated with temozolomide (TMZ) for recurrent or progressive malignant gliomas after standard radiation therapy. Cost assessment (payer's perspective) singled out treatment for first recurrence and all costs of care until death. We computed personnel costs as wages; drugs, imaging, and laboratory tests as prices; and hospitalizations as day rates. Patients were administered a median of five TMZ cycles at recurrence. Drug acquisition costs amounted to euro 2206 per cycle (76% of total costs). Seven patients showed no second recurrence (two are still alive), 16 received no further chemotherapy and died after 3.9 months, and 26 received second-line chemotherapy. After the second progression, median survival was 4.0 months (95% confidence interval, 1.8-6.1). Overall monthly costs of care varied between euro 2450 and euro 3242 among the different groups, and median cost-effectiveness and cost utility ranged from euro 28,817 to euro 38,450 and from euro 41,167 to euro 53,369 per life of year and per quality-adjusted life-year gained, respectively. We conclude that despite high TMZ drug acquisition costs, care of recurrent malignant gliomas is comparable to other accepted therapies.


Assuntos
Antineoplásicos Alquilantes/economia , Neoplasias Encefálicas/economia , Dacarbazina/análogos & derivados , Dacarbazina/economia , Glioma/economia , Recidiva Local de Neoplasia/economia , Adulto , Idoso , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/mortalidade , Efeitos Psicossociais da Doença , Análise Custo-Benefício , Dacarbazina/uso terapêutico , Feminino , Glioma/tratamento farmacológico , Glioma/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/mortalidade , Análise de Sobrevida , Temozolomida
13.
Cancer ; 101(9): 2098-105, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15389472

RESUMO

BACKGROUND: Adding temozolomide (TMZ) to standard radiotherapy as a first-line therapy for glioma may increase costs to a disproportionate degree compared with the resulting survival benefits. METHODS: Forty-six consecutive patients (28 males and 18 females; median age, 52 years; age range, 24-70 years) received concomitant TMZ with radiotherapy for 6 weeks followed by adjuvant TMZ for 6 cycles, and they were followed until disease recurrence and then until death. The authors assessed the costs associated with the four phases of treatment from a hospital-centered perspective. RESULTS: Treatment was discontinued early in 3 patients, 9 patients, and 15 patients during concomitant TMZ, before adjuvant TMZ, and during adjuvant TMZ, respectively. Karnofsky index values varied between 85% (at the beginning of treatment) and 76% (at the end of treatment). The nature of care after disease recurrence was diverse. Overall survival ranged from 1.4 months to 64.3 months (median, 15.8 months) and was better if surgical debulking could be carried out before treatment. Global costs amounted to Euros 39,092 +/- Euros 21,948 (concomitant TMZ, Euros 14,539 +/- Euros 4998; adjuvant TMZ, Euros 13,651 +/- Euros 4320; follow-up, Euros 6363 +/- Euros 6917; and recurrence, Euros 12,344 +/- Euros 18,327), with 53% of these costs being related to the acquisition of TMZ; this represented an eightfold increase in cost compared with radiotherapy alone. CONCLUSIONS: TMZ may be an effective but costly adjuvant outpatient therapy for patients with glioblastoma multiforme. Definite cost-effectiveness/utility must be assessed in a randomized Phase III trial.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Dacarbazina/economia , Dacarbazina/uso terapêutico , Glioblastoma/terapia , Custos de Cuidados de Saúde , Adulto , Idoso , Neoplasias Encefálicas/mortalidade , Terapia Combinada , Análise Custo-Benefício , Europa (Continente) , Feminino , Glioblastoma/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , O(6)-Metilguanina-DNA Metiltransferase/genética , Temozolomida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA