Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Biomolecules ; 11(8)2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34439781

RESUMO

SARS-CoV-2 infection of host cells is driven by binding of the SARS-CoV-2 spike-(S)-protein to lung type II pneumocytes, followed by virus replication. Surfactant protein SP-D, member of the front-line immune defense of the lungs, binds glycosylated structures on invading pathogens such as viruses to induce their clearance from the lungs. The objective of this study is to measure the pulmonary SP-D levels in COVID-19 patients and demonstrate the activity of SP-D against SARS-CoV-2, opening the possibility of using SP-D as potential therapy for COVID-19 patients. Pulmonary SP-D concentrations were measured in bronchoalveolar lavage samples from patients with corona virus disease 2019 (COVID-19) by anti-SP-D ELISA. Binding assays were performed by ELISAs. Protein bridge and aggregation assays were performed by gel electrophoresis followed by silver staining and band densitometry. Viral replication was evaluated in vitro using epithelial Caco-2 cells. Results indicate that COVID-19 patients (n = 12) show decreased pulmonary levels of SP-D (median = 68.9 ng/mL) when compared to levels reported for healthy controls in literature. Binding assays demonstrate that SP-D binds the SARS-CoV-2 glycosylated spike-(S)-protein of different emerging clinical variants. Binding induces the formation of protein bridges, the critical step of viral aggregation to facilitate its clearance. SP-D inhibits SARS-CoV-2 replication in Caco-2 cells (EC90 = 3.7 µg/mL). Therefore, SP-D recognizes and binds to the spike-(S)-protein of SARS-CoV-2 in vitro, initiates the aggregation, and inhibits viral replication in cells. Combined with the low levels of SP-D observed in COVID-19 patients, these results suggest that SP-D is important in the immune response to SARS-CoV-2 and that rhSP-D supplementation has the potential to be a novel class of anti-viral that will target SARS-CoV-2 infection.


Assuntos
COVID-19/metabolismo , Proteína D Associada a Surfactante Pulmonar/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Adulto , Idoso , COVID-19/virologia , Células CACO-2 , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ligação Proteica , Proteína D Associada a Surfactante Pulmonar/genética , Proteína D Associada a Surfactante Pulmonar/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Replicação Viral
3.
CNS Oncol ; 7(3): CNS23, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30124334

RESUMO

AIM: To estimate the mean lifetime survival benefit, an essential component of health economic evaluations in oncology, of adding tumor treating fields (TTFields) to maintenance temozolomide (TMZ) for newly diagnosed glioblastoma patients. METHODS: We integrated EF-14 trial data with glioblastoma epidemiology data. The model provided for an evidence-based approach to estimate lifetime survival for the material number of EF-14 trial patients still alive at 5 years. RESULTS & CONCLUSION: Patients treated with TTFields and TMZ had an incremental mean lifetime survival of 1.8 years (TTFields/TMZ: 4.2 vs TMZ alone: 2.4). Patients alive at year 2 after starting TTFields had a 20.7% probability of surviving to year 10. The results presented here provide the required incremental survival benefit necessary for a future assessment of the incremental cost-effectiveness of TTFields.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas , Terapia por Estimulação Elétrica/métodos , Glioblastoma , Temozolomida/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/mortalidade , Terapia Combinada , Intervalo Livre de Doença , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/epidemiologia , Glioblastoma/mortalidade , Humanos , Estudos Longitudinais , Masculino
4.
ESMO Open ; 3(4): e000381, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30018811

RESUMO

PURPOSE: TrasGEX is a second-generation monoclonal antibody of trastuzumab, glyco-optimised to enhance antibody-dependent cellular cytotoxicity while fully retaining trastuzumab's antigen-binding properties to human epidermal growth factor receptor 2 (HER2). A phase I dose-escalation study was conducted to establish the optimal TrasGEX dose and regimen for phase II studies and to define the safety, pharmacokinetics (PK) and preliminary antitumour activity of TrasGEX. PATIENTS AND METHODS: A total of 37 patients with advanced HER2-positive carcinomas and progressive disease received TrasGEX intravenously every 3 weeks until disease progression in doses of 12-720 mg in a three-plus-three dose escalation design, including an expansion cohort at the highest dose. RESULTS: No dose limiting toxicity was observed, and no maximum tolerated dose was reached. Drug-related adverse events were mainly infusion-related reactions occurring during the first infusion in 51% of patients; all but two were mild-to-moderate. Compared with trastuzumab, the PK parameters were dose dependent, with a mean terminal half-life (t1/2) of 263±99 hours for the 720 mg dose. Clinical benefit in 15 out of 30 (50%) evaluable patients included one ongoing complete response, two partial remissions lasting 16 and 77 weeks and disease stabilisation (SD) in 12 patients lasting a median (range) of 17 (7-26) weeks; three of them had SD of 24, 25 and 26 weeks, respectively. CONCLUSION: TrasGEX was safe, well-tolerated and showed antitumour activity in 50% of evaluable patients, all with progressive disease at study entry. Infusions at an interval of 2-3 weeks should achieve clinically relevant trough levels for future studies (NCT01409343).

5.
ESMO Open ; 3(2): e000303, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29464112

RESUMO

BACKGROUND: Changes in glycosylation of the constant domain (Fc) of monoclonal antibodies (mAbs) enhance antibody-dependent cell-mediated cytotoxicity independently of downstream effects following receptor blockade by the antibody, thus extending their indication. We investigated the safety, pharmacokinetics, pharmacodynamics and antitumour activity of tomuzotuximab, an IgG1 glycoengineered mAb against the epidermal growth factor receptor with enhanced tumour cytotoxicity in a phase I dose-escalation study (NTC01222637). PATIENTS AND METHODS: Forty-one patients with advanced solid tumours refractory to standard therapies received tomuzotuximab weekly (12-1370 mg) or two-weekly (990 mg) on a three-plus-three dose escalation design. RESULTS: A maximum tolerated dose was not reached. The most frequent treatment-related adverse events were infusion-related reactions in 31 (76%) patients (grade 3, 12%), mainly confined to the first dose, and skin toxicities (grade 1 or 2) in 30 (73%) patients. Hypomagnesaemia was observed in 9 out of 23 evaluable patients (39%). Similar to cetuximab, tomuzotuximab concentrations increased proportionally to dose from doses≥480 mg with a median terminal half life (t½) of 82 hours, range 55-113 hours. Antitumour activity included one complete response ongoing since more than 4.5 years in a patient with non-small-cell lung cancer and one partial response lasting 353 days in a patient with colorectal cancer. Twelve patients achieved stable disease (median, 166 days, range, 71-414 days) and two patients had prolonged control (>1 year) of their non-measurable disease. CONCLUSION: Tomuzotuximab was safe and showed promising antitumour activity in heavily pretreated patients with advanced metastatic disease. A phase IIb trial of chemotherapy and weekly tomuzotuximab or cetuximab followed with maintenance therapy with the corresponding mAb in patients with recurrent or metastatic head and neck squamous cell carcinoma is ongoing.

6.
Lung Cancer ; 81(3): 445-450, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23891283

RESUMO

BACKGROUND: Low intensity, intermediate frequency, alternating electric fields (Tumor Treating Fields; TTFields) exhibit anti-mitotic activity in cancer cells. Promising preclinical data have led to a single arm phase I/II trial in NSCLC patients. METHODS: Forty-two inoperable stage IIIB (with pleural effusion) and IV NSCLC patients who had had tumor progression received pemetrexed 500 mg/m(2) iv q3w together with daily TTFields therapy until disease progression. The primary endpoint was time to "in-field" progression. RESULTS: Median age for all patients was 63 years, 76% had stage IV disease, 78% had adenocarcinoma and 17% had performance status of 2. The median time to in-field progression was 28 weeks and the median time to systemic progression was 22 weeks. Six patients (14.6%) had a partial remission (PR) and 20 had stable disease (SD) (48.8%). Median overall survival was 13.8 months and 1 year survival rate was 57%. There were no TTFields-related serious adverse events. CONCLUSIONS: The combination of TTFields and pemetrexed as a second line therapy for NSCLC is safe and potentially more effective than pemetrexed alone. TTFields improved disease control within the treatment field and a phase III study is planned to further investigate its role as a novel treatment in NSCLC.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Glutamatos/uso terapêutico , Guanina/análogos & derivados , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Modalidades de Fisioterapia , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Terapia Combinada , Feminino , Glutamatos/administração & dosagem , Glutamatos/efeitos adversos , Guanina/administração & dosagem , Guanina/efeitos adversos , Guanina/uso terapêutico , Humanos , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Pemetrexede , Modalidades de Fisioterapia/efeitos adversos , Resultado do Tratamento
7.
Clin Pharmacokinet ; 52(9): 783-92, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23681967

RESUMO

BACKGROUND AND OBJECTIVES: Angiotensin-converting enzyme 2 (ACE2) converts angiotensin II (Ang1-8) to angiotensin 1-7 (Ang1-7), a functional antagonist of Ang1-8, with vasodilatory, antiproliferative, antiangiogenic, and anti-inflammatory properties. In conditions with an unbalanced renin-angiotensin-aldosterone system with elevated Ang1-8, administration of ACE2 has shown promising effects in a variety of animal models. Enhancing ACE2 activity by exogenous administration of ACE2 might also be beneficial in human diseases with pathologically elevated Ang1-8. As a first step we performed a first-in-man study to determine pharmacokinetics, pharmacodynamics, safety, and tolerability of recombinant ACE2 in healthy volunteers. METHODS: Recombinant human ACE2 (rhACE2) was administered intravenously to healthy human subjects in a randomized, double-blind, placebo-controlled, single-dose, dose-escalation study followed by an open-label multiple-dose study. ACE2 concentrations were determined by quantifying ACE2 activity and ACE2 content in plasma samples. Concentrations of the angiotensin system effector peptides Ang1-8, Ang1-7, and Ang1-5 were determined using a liquid chromatography-tandem mass spectrometry method. RESULTS: Single rhACE2 doses of 100-1,200 µg/kg caused a dose-dependent increase of systemic exposure with biphasic elimination and a dose-independent terminal half-life of 10 h. In all single-dose cohorts, Ang1-8 decreased within 30 min postinfusion, angiotensin 1-7 (Ang1-7) either increased (100 and 200 µg/kg doses), decreased, or remained unchanged (400-1,200 µg/kg doses), whereas angiotensin 1-5 (Ang1-5) transiently increased for all doses investigated. With the exception of the lowest rhACE2 dose, the decrease in Ang1-8 levels lasted for at least 24 h. Repeated dosing (400 µg/kg for 3 or 6 days) caused only minimal accumulation of ACE2, and Ang1-8 levels were suppressed over the whole application period. CONCLUSIONS: Administration of rhACE2 was well tolerated by healthy human subjects. Exposure was dose dependent with a dose-independent terminal elimination half-life in the range of 10 h. Despite marked changes in angiotensin system peptide concentrations, cardiovascular effects were absent, suggesting the presence of effective compensatory mechanisms in healthy volunteers.


Assuntos
Peptidil Dipeptidase A/farmacocinética , Proteínas Recombinantes/farmacocinética , Angiotensina I/sangue , Angiotensina II/sangue , Enzima de Conversão de Angiotensina 2 , Método Duplo-Cego , Feminino , Humanos , Masculino , Fragmentos de Peptídeos/sangue , Peptidil Dipeptidase A/administração & dosagem , Proteínas Recombinantes/administração & dosagem
8.
Rambam Maimonides Med J ; 3(2): e0007, 2012 04.
Artigo em Inglês | MEDLINE | ID: mdl-23908831

RESUMO

Phase 1 first-in-human studies with anti-cancer products differ from other phase 1 studies in that they are evaluated in patients rather than healthy volunteers. The rationale design of targeted drugs triggers changes in the design of these studies. Patient populations are more precisely defined and pose a challenge to the efficient inclusion of study patients. Objectives shift from the definition of a maximum tolerated dose to the evaluation of a recommended phase 2 dose. Other challenges related to the efficacy and safety profile of novel targeted anti-cancer drugs call for changes in designing first-in-human studies, such as definitions of biological doses, collection of fresh tumor tissue for surrogate marker analyses, and the management of infusion-related reactions with monoclonal antibodies. Consequently, the conduct of phase 1 clinical trials in oncology requires changes. Corresponding education with particular focus on phase 1 trials and on the complex drug development process needs to be an integrated part of the medical oncology curriculum for physicians and nursing staff. This is a crucial element for institutions to remain or become clinical research sites for phase 1 studies, and to participate in the drug development process of novel anti-cancer compounds in the future.

9.
Lancet Oncol ; 9(10): 946-52, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18775668

RESUMO

BACKGROUND: Bacterial infection of the pleural space often causes adherence of the pleural membranes by fibrous tissue, probably mediated by inflammation initiated by bacterial cell-wall motifs, including lipoteichoic acid-T (LTA-T). We postulated that therapeutically administered LTA-T might produce a similar effect, achieving control of malignant pleural effusion (pleurodesis). METHODS: Patients with histocytologically proven symptomatic malignant pleural effusions were included in this phase I toxicity and dose-escalation study, An indwelling pleural catheter was placed in the pleural effusion to drain the fluid fully. A control dose of intrapleural saline was administered after complete drainage (day 1) and pleural-fluid production was recorded for 7 days. On day 7 a single dose of intrapleural LTA-T (increasing in each patient) was administered and pleural-fluid production was monitored for a further 7 days. Long-term fluid control was recorded. This study is registered as an International Standard Randomised Controlled Trial, ISRCTN44367564. FINDINGS: Between November, 2004, and November, 2005, 14 patients were enrolled on the trial at the Oxford Centre for Respiratory Medicine (Oxford, UK). 13 of 14 patients received escalated doses of LTA-T. A dose-limiting toxic effect (ie, systemic inflammation) occurred at 3000 microg, and a therapeutic dose of 750-1500 microg was established. Toxic effects were mild and had no consistent pattern at the therapeutic dose. Pleural-fluid production decreased significantly after a dose of at least 750 microg LTA-T, compared with saline control (mean fluid production after saline control 1244 mL [SD 933], mean fluid production after LTA-T 394 mL [SD 375], mean difference -850 mL [SD 699], p=0.028), and six of seven (86%) patients achieved pleural-fluid control at 1 month with no further intervention. INTERPRETATION: The toxic effects of intrapleural LTA-T seem to be mild and favourable when compared with the toxicity profiles of standard pleurodesis agents. There is early evidence of LTA-T-induced pleurodesis efficacy, suggesting that this might be a viable therapeutic strategy for the control of malignant pleural effusion.


Assuntos
Lipopolissacarídeos/administração & dosagem , Derrame Pleural Maligno/terapia , Pleurodese/métodos , Ácidos Teicoicos/administração & dosagem , Adulto , Citocinas/análise , Relação Dose-Resposta a Droga , Drenagem , Feminino , Humanos , Contagem de Leucócitos , Lipopolissacarídeos/efeitos adversos , Masculino , Pessoa de Meia-Idade , Derrame Pleural Maligno/patologia , Derrame Pleural Maligno/fisiopatologia , Ácidos Teicoicos/efeitos adversos
10.
Onkologie ; 31(7): 362-5, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18596382

RESUMO

BACKGROUND: The transmission of electric fields using insulated electrodes has demonstrated that very low-intensity, properly tuned, intermediate-frequency electric fields, termed tumor-treating fields (TTFields), selectively stunts tumor cell growth and is accompanied by a decrease in tumor angiogenesis. PATIENTS AND METHODS: This open, prospective pilot study was designed to evaluate the safety, tolerability, and efficacy profile of TTFields treatment in patients with locally advanced and/or metastatic solid tumors using the NovoTTF100A(TM) device. All 6 patients were heavily pre-treated with several lines of therapy; no additional standard treatment option was available to them. TTFields treatment using continuous NovoTTF-100A lasted a minimum of 14 days and was very well tolerated. RESULTS: No related serious adverse events occurred. Outcomes showed 1 partial response of a treated skin metastasis from a primary breast cancer, 3 cases where tumor growth was arrested during treatment, and 1 case of disease progression. One mesothelioma patient experienced lesion regression near TTFields with simultaneous tumor stability or progression in distal areas. CONCLUSION: Although the number of patients in this study is small, the lack of therapy toxicity and the efficacy observed in data gathered to date indicate the potential of TTFields as a new treatment modality for solid tumors, definitely warranting further investigation.


Assuntos
Terapia por Estimulação Elétrica/métodos , Recidiva Local de Neoplasia/prevenção & controle , Neoplasias/terapia , Neoplasias Cutâneas/secundário , Neoplasias Cutâneas/terapia , Adulto , Idoso , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Terapia por Estimulação Elétrica/efeitos adversos , Terapia por Estimulação Elétrica/tendências , Eletricidade/efeitos adversos , Campos Eletromagnéticos/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Resultado do Tratamento
11.
J Thorac Oncol ; 3(1): 30-6, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18166838

RESUMO

BACKGROUND: Patients with advanced non-small cell lung cancer (NSCLC) require care that emphasizes symptom palliation in addition to extending survival. The low response rates and minimal survival gains observed in second-line studies underscore the need to assess treatment efficacy with symptomatic end points. METHODS: To characterize the relationship between patient-reported health-related quality of life outcomes and efficacy end points (tumor response, overall survival [OS], progression-free survival [PFS]), retrospective analyses were performed on Lung Cancer Symptom Scale (LCSS) data (n = 488) from the phase III study of pemetrexed (500 mg/m2 once every 3 weeks) versus docetaxel (75 mg/m2 once every 3 weeks) in advanced NSCLC. The LCSS data consisted of patient ratings of six symptoms and three summary items using 100-mm visual analogue scales. The mean maximum improvement for each item was categorized according to best tumor response, with statistical analyses based on a two-factor interaction model (with treatment arm and response group as fixed factors). Additional analyses pooled data between treatment arms and examined correlation (nonparametric and Pearson's) of time to first worsening of symptoms (TWS) with PFS and OS. RESULTS: All LCSS items, except hemoptysis, showed mean maximum improvement over baseline for responders and patients with stable disease (p < 0.01), with greater improvement associated with response. Median TWS for each LCSS item ranged between 2.3 months (fatigue) and 7.0 months (cough), with correlation between TWS and PFS and OS (all p values

Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Ensaios Clínicos Fase III como Assunto , Glutamatos/uso terapêutico , Guanina/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Taxoides/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Docetaxel , Seguimentos , Guanina/uso terapêutico , Humanos , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Estadiamento de Neoplasias , Pemetrexede , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto , Estudos Retrospectivos , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento
12.
Oncology ; 72(3-4): 147-51, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18087175

RESUMO

OBJECTIVES: Pegylated liposomal doxorubicin (PLD) has improved therapy options significantly, as it causes less myelosuppression, nausea, vomiting, and alopecia than conventional doxorubicin, while maintaining efficacy. The goal of this survey was to determine whether the use of PLD in a community-based patient group is comparable regarding chemotherapeutic doses and side effects to preselected study patients. METHODS: 100 questionnaires were randomly sent to Swiss oncologists in private practices, general hospitals and university hospitals. RESULTS: The patient cohort was heterogeneous with respect to prior treatments. PLD was an active agent in metastatic breast cancer and was well tolerated by the majority of patients. The most common non-hematological side effects were hand-foot syndrome (HFS) and mucositis while only patients receiving a dose of 50 mg/m(2) (recommended dose) experienced grade 4 HFS. The reported mean dose of PLD was 38.5 mg/m(2). CONCLUSIONS: This community-based observational study supports previous reports indicating that PLD at a median dose of < or =40 mg/m(2) every 4 weeks is an active, well-tolerated agent in non-selected, pretreated patients with metastatic breast cancer.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/análogos & derivados , Polietilenoglicóis/administração & dosagem , Idoso , Doxorrubicina/administração & dosagem , Feminino , Pesquisas sobre Atenção à Saúde , Humanos , Lipossomos , Pessoa de Meia-Idade , Metástase Neoplásica , Resultado do Tratamento
13.
Onkologie ; 30(7): 355-60, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17596743

RESUMO

BACKGROUND: Prostate cancer is the most common type of cancer in men, however, therapeutic options are limited. 50-90% of hormone-refractory prostate cancer cells show an overexpression of epidermal growth factor receptor (EGFR), which may contribute to uncontrolled proliferation and resistance to chemotherapy. In vitro, gefitinib, an orally administered tyrosine kinase inhibitor, has shown a significant increase in antitumor activity when combined with chemotherapy. PATIENTS AND METHODS: In this phase II study, the safety and efficacy of gefitinib in combination with docetaxel, a chemotherapeutic agent commonly used for prostate cancer, was investigated in patients with hormone-refractory prostate cancer (HRPC). 37 patients with HRPC were treated continuously with gefitinib 250 mg once daily and docetaxel 35 mg/m2 i.v. for up to 6 cycles. PSA response, defined as a =50% decrease in serum PSA compared with trial entry, was the primary efficacy parameter. PSA levels were measured at prescribed intervals. RESULTS: The response rate and duration of response were consistent with those seen with docetaxel monotherapy. The combination of docetaxel and gefitinib was reasonably well tolerated in this study. CONCLUSION: Future studies should investigate whether patients with specific tumor characteristics, e.g. EGFR protein overexpression, respond better to gefitinib than patients without, leading to a more customized therapy option.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/tratamento farmacológico , Quinazolinas/administração & dosagem , Taxoides/administração & dosagem , Administração Oral , Idoso , Idoso de 80 Anos ou mais , Antagonistas de Androgênios/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Progressão da Doença , Docetaxel , Relação Dose-Resposta a Droga , Esquema de Medicação , Resistencia a Medicamentos Antineoplásicos , Gefitinibe , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias da Próstata/sangue , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Quinazolinas/efeitos adversos , Taxa de Sobrevida , Taxoides/efeitos adversos , Resultado do Tratamento
14.
Proc Natl Acad Sci U S A ; 104(24): 10152-7, 2007 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-17551011

RESUMO

We have recently shown that low intensity, intermediate frequency, electric fields inhibit by an anti-microtubule mechanism of action, cancerous cell growth in vitro. Using implanted electrodes, these fields were also shown to inhibit the growth of dermal tumors in mice. The present study extends these findings to additional cell lines [human breast carcinoma; MDA-MB-231, and human non-small-cell lung carcinoma (H1299)] and to animal tumor models (intradermal B16F1 melanoma and intracranial F-98 glioma) using external insulated electrodes. These findings led to the initiation of a pilot clinical trial of the effects of TTFields in 10 patients with recurrent glioblastoma (GBM). Median time to disease progression in these patients was 26.1 weeks and median overall survival was 62.2 weeks. These time to disease progression and OS values are more than double the reported medians of historical control patients. No device-related serious adverse events were seen after >70 months of cumulative treatment in all of the patients. The only device-related side effect seen was a mild to moderate contact dermatitis beneath the field delivering electrodes. We conclude that TTFields are a safe and effective new treatment modality which effectively slows down tumor growth in vitro, in vivo and, as demonstrated here, in human cancer patients.


Assuntos
Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica , Glioblastoma/terapia , Recidiva Local de Neoplasia , Adulto , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Terapia por Estimulação Elétrica/efeitos adversos , Feminino , Glioblastoma/patologia , Humanos , Camundongos , Microeletrodos , Modelos Biológicos , Neoplasias Experimentais/terapia , Projetos Piloto , Ratos , Ratos Endogâmicos F344 , Taxa de Sobrevida , Fatores de Tempo , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Thorac Oncol ; 2(5): 397-401, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17473654

RESUMO

BACKGROUND: In a recent large phase III study, previously treated patients with advanced non-small cell lung cancer who received pemetrexed demonstrated a survival time similar to patients who received docetaxel (median, 8.3 months with pemetrexed versus 7.9 months with docetaxel), with a more favorable toxicity profile, and significantly fewer Common Toxicity Criteria grade 3/4 toxicities. This is a retrospective risk-benefit analysis of survival without grade 3/4 toxicity, defined as the time to the first occurrence of Common Toxicity Criteria grade 3 or 4 toxicity or death, in the prospective phase III study comparing pemetrexed with docetaxel. METHODS: A total of 541 patients (of 571 randomized) received either pemetrexed (500 mg/m intravenously [IV]) supplemented with vitamin B12 injections and oral folic acid or docetaxel (75 mg/m IV) on day 1 of 21-day cycles. Survival without grade 3/4 toxicity was analyzed using Kaplan-Meier and Cox methods. RESULTS: Pemetrexed demonstrated a statistically significantly longer survival without grade 3/4 toxicity compared with docetaxel (hazard ratio = 0.60, 95% confidence interval: 0.50-0.72; p < 0.0001). A supportive analysis based on selected grade 3/4 toxicities (neutropenia lasting >5 days, febrile neutropenia, infection with neutropenia, anemia, thrombocytopenia, fatigue, nausea, vomiting, diarrhea, stomatitis, and neurosensory events) also demonstrated an advantage for pemetrexed (hazard ratio = 0.53; 95% confidence interval: 0.44-0.64; p < 0.0001). CONCLUSION: This analysis of survival without grade 3/4 toxicity suggests a benefit-to-risk profile that favors pemetrexed over docetaxel in the second-line treatment of patients with non-small cell lung cancer.


Assuntos
Antineoplásicos/efeitos adversos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Glutamatos/efeitos adversos , Guanina/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Taxoides/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos , Docetaxel , Feminino , Glutamatos/uso terapêutico , Guanina/efeitos adversos , Guanina/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Pemetrexede , Modelos de Riscos Proporcionais , Estudos Prospectivos , Estudos Retrospectivos , Análise de Sobrevida , Taxoides/uso terapêutico , Resultado do Tratamento
17.
Cancer Chemother Pharmacol ; 57(4): 533-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16052341

RESUMO

Epidermal growth factor receptor (EGFR) expression has been associated with clinical outcome in some studies of renal-cell carcinoma (RCC). We investigated the efficacy and safety of gefitinib (IRESSA), an EGFR tyrosine kinase inhibitor, in RCC patients. This phase II trial recruited 28 patients with advanced, metastatic, or relapsed RCC. Patients received oral gefitinib 500 mg/day. Objective responses (ORs) were assessed every 2 months according to RECIST. Baseline tumor biopsies were analyzed immunohistochemically for EGFR expression. At trial closure (March 2003), no ORs were seen but 14 patients (53.8%) had stable disease. At extended analysis (August 2004), median time to progression was 110 days (95% confidence interval [CI]: 55, 117); median overall survival was 303 days (95% CI 180, 444). Gefitinib was generally well tolerated. Skin rash and diarrhea were the most common drug-related adverse events (AEs) [54 and 39% of patients, respectively] and the most common drug-related grade 3/4 AEs (both 11%). The majority of tumor biopsies (91%) had > or =70% of tumor cells expressing membrane EGFR. Despite the lack of ORs in this study, disease control was observed in 53.8% of patients. Gefitinib was generally well tolerated and no unexpected drug-related AEs were observed.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico , Quinazolinas/uso terapêutico , Adulto , Idoso , Antineoplásicos/efeitos adversos , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Receptores ErbB/biossíntese , Receptores ErbB/genética , Feminino , Gefitinibe , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Quinazolinas/efeitos adversos , Recidiva , Tomografia Computadorizada por Raios X
18.
Invest New Drugs ; 24(4): 299-304, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16237511

RESUMO

Vascular endothelial growth factor (VEGF) is a potent stimulant of angiogenesis. SU5416, is a small molecule tyrosine kinase inhibitor, and a potent inhibitor of VEGF-mediated Flk-1 receptor signaling. Intravenous agent SU5416 has shown evidence of biological activity against a variety of tumor types. The current intravenous dosing regimen is not optimal for long-term administration, which is needed for optimal efficacy. The aim of this study was to evaluate the safety profile and pharmacokinetics of a Nanocrystal Colloidal Dispersion (NCD) SU5416 formulation in humans. Patients with advanced and/or metastatic solid organ tumors were included in the trial; various SU5416 regimens were tested for tolerability, safety and were evaluated concerning pharmacokinetics. The results of this study indicate that induction of clearance after oral dosing of NCD SU5416 in humans occurs and is greater than following i.v. administration. It has been confirmed that SU5416 is a high clearance compound, also as an oral NCD formulation. The NCD formulation was well tolerated, but no effective drug serum levels could be achieved. These data help to understand the ADME (Absorption, Distribution, Metabolism, Excretion) properties of indoline chemical class compounds. The lessons learned should be applied in the development of next generation indoline anti-angiogenic and anti-tumor compounds.


Assuntos
Indóis/farmacocinética , Indóis/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Pirróis/farmacocinética , Pirróis/uso terapêutico , Administração Oral , Adulto , Esquema de Medicação , Feminino , Humanos , Indóis/administração & dosagem , Indóis/efeitos adversos , Infusões Intravenosas , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Pirróis/administração & dosagem , Pirróis/efeitos adversos
19.
Oncology ; 68(4-6): 293-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16020955

RESUMO

Ovarian cancer is the fifth most common cause of death from cancer in women. The standard first-line treatment for advanced ovarian cancer is a combination of paclitaxel and carboplatin or carboplatin alone. Sequential single-agent therapy is of particular interest in patients with symptom-free disease progression. Age, performance status and treatment preferences of the respective patient are further decisive factors which should be taken into account when selecting single or combination therapy. Second-line treatment depends, for instance, on the duration of response to first-line platinum therapy, previous treatment regimens used, tolerability, the patient's performance status and preference of a particular treatment, and cost-effectiveness. If tumor recurrence occurs within 6 months following platinum-based therapy, other agents such as paclitaxel, pegylated liposomal doxorubicin, topotecan or gemcitabine should be used. If the tumor recurs after 6 months, a combination therapy of platinum and paclitaxel has proven to be the most effective. Reasonable options in progressive disease are treatment with platinum, either alone or combined with other agents, especially investigational compounds.


Assuntos
Antineoplásicos/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Feminino , Humanos , Neoplasias Ovarianas/patologia
20.
J Clin Oncol ; 23(23): 5314-22, 2005 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-15955899

RESUMO

PURPOSE: In this study, two doses of temsirolimus (CCI-779), a novel inhibitor of the mammalian target of rapamycin, were evaluated for efficacy, safety, and pharmacokinetics in patients with locally advanced or metastatic breast cancer who had been heavily pretreated. PATIENTS AND METHODS: Patients (n = 109) were randomly assigned to receive 75 or 250 mg of temsirolimus weekly as a 30-minute intravenous infusion. Patients were evaluated for tumor response, time to tumor progression, adverse events, and pharmacokinetics of temsirolimus. RESULTS: Temsirolimus produced an objective response rate of 9.2% (10 partial responses) in the intent-to-treat population. Median time to tumor progression was 12.0 weeks. Efficacy was similar for both dose levels but toxicity was more common with the higher dose level, especially grade 3 or 4 depression (10% of patients at the 250-mg dose level, 0% at the 75-mg dose level). The most common temsirolimus-related adverse events of all grades were mucositis (70%), maculopapular rash (51%), and nausea (43%). The most common, clinically important grade 3 or 4 adverse events were mucositis (9%), leukopenia (7%), hyperglycemia (7%), somnolence (6%), thrombocytopenia (5%), and depression (5%). CONCLUSION: In heavily pretreated patients with locally advanced or metastatic breast cancer, 75 and 250 mg temsirolimus showed antitumor activity and 75 mg temsirolimus showed a generally tolerable safety profile.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Quinases/efeitos dos fármacos , Terapia de Salvação , Sirolimo/análogos & derivados , Neoplasias de Tecidos Moles/tratamento farmacológico , Adulto , Idoso , Antineoplásicos/uso terapêutico , Área Sob a Curva , Neoplasias Ósseas/sangue , Neoplasias Ósseas/secundário , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Segurança , Sirolimo/efeitos adversos , Sirolimo/farmacocinética , Sirolimo/uso terapêutico , Neoplasias de Tecidos Moles/sangue , Neoplasias de Tecidos Moles/secundário , Serina-Treonina Quinases TOR , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...