Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lancet ; 403(10435): 1460-1471, 2024 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-38554725

RESUMO

BACKGROUND: Afamitresgene autoleucel (afami-cel) showed acceptable safety and promising efficacy in a phase 1 trial (NCT03132922). The aim of this study was to further evaluate the efficacy of afami-cel for the treatment of patients with HLA-A*02 and MAGE-A4-expressing advanced synovial sarcoma or myxoid round cell liposarcoma. METHODS: SPEARHEAD-1 was an open-label, non-randomised, phase 2 trial done across 23 sites in Canada, the USA, and Europe. The trial included three cohorts, of which the main investigational cohort (cohort 1) is reported here. Cohort 1 included patients with HLA-A*02, aged 16-75 years, with metastatic or unresectable synovial sarcoma or myxoid round cell liposarcoma (confirmed by cytogenetics) expressing MAGE-A4, and who had received at least one previous line of anthracycline-containing or ifosfamide-containing chemotherapy. Patients received a single intravenous dose of afami-cel (transduced dose range 1·0 × 109-10·0 × 109 T cells) after lymphodepletion. The primary endpoint was overall response rate in cohort 1, assessed by a masked independent review committee using Response Evaluation Criteria in Solid Tumours (version 1.1) in the modified intention-to-treat population (all patients who received afami-cel). Adverse events, including those of special interest (cytokine release syndrome, prolonged cytopenia, and neurotoxicity), were monitored and are reported for the modified intention-to-treat population. This trial is registered at ClinicalTrials.gov, NCT04044768; recruitment is closed and follow-up is ongoing for cohorts 1 and 2, and recruitment is open for cohort 3. FINDINGS: Between Dec 17, 2019, and July 27, 2021, 52 patients with cytogenetically confirmed synovial sarcoma (n=44) and myxoid round cell liposarcoma (n=8) were enrolled and received afami-cel in cohort 1. Patients were heavily pre-treated (median three [IQR two to four] previous lines of systemic therapy). Median follow-up time was 32·6 months (IQR 29·4-36·1). Overall response rate was 37% (19 of 52; 95% CI 24-51) overall, 39% (17 of 44; 24-55) for patients with synovial sarcoma, and 25% (two of eight; 3-65) for patients with myxoid round cell liposarcoma. Cytokine release syndrome occurred in 37 (71%) of 52 of patients (one grade 3 event). Cytopenias were the most common grade 3 or worse adverse events (lymphopenia in 50 [96%], neutropenia 44 [85%], leukopenia 42 [81%] of 52 patients). No treatment-related deaths occurred. INTERPRETATION: Afami-cel treatment resulted in durable responses in heavily pre-treated patients with HLA-A*02 and MAGE-A4-expressing synovial sarcoma. This study shows that T-cell receptor therapy can be used to effectively target solid tumours and provides rationale to expand this approach to other solid malignancies. FUNDING: Adaptimmune.


Assuntos
Anemia , Lipossarcoma Mixoide , Sarcoma Sinovial , Trombocitopenia , Adulto , Humanos , Sarcoma Sinovial/tratamento farmacológico , Sarcoma Sinovial/genética , Lipossarcoma Mixoide/etiologia , Síndrome da Liberação de Citocina/etiologia , Ifosfamida , Trombocitopenia/etiologia , Anemia/etiologia , Antígenos HLA-A , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
2.
Nat Med ; 29(1): 104-114, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36624315

RESUMO

Affinity-optimized T cell receptors can enhance the potency of adoptive T cell therapy. Afamitresgene autoleucel (afami-cel) is a human leukocyte antigen-restricted autologous T cell therapy targeting melanoma-associated antigen A4 (MAGE-A4), a cancer/testis antigen expressed at varying levels in multiple solid tumors. We conducted a multicenter, dose-escalation, phase 1 trial in patients with relapsed/refractory metastatic solid tumors expressing MAGE-A4, including synovial sarcoma (SS), ovarian cancer and head and neck cancer ( NCT03132922 ). The primary endpoint was safety, and the secondary efficacy endpoints included overall response rate (ORR) and duration of response. All patients (N = 38, nine tumor types) experienced Grade ≥3 hematologic toxicities; 55% of patients (90% Grade ≤2) experienced cytokine release syndrome. ORR (all partial response) was 24% (9/38), 7/16 (44%) for SS and 2/22 (9%) for all other cancers. Median duration of response was 25.6 weeks (95% confidence interval (CI): 12.286, not reached) and 28.1 weeks (95% CI: 12.286, not reached) overall and for SS, respectively. Exploratory analyses showed that afami-cel infiltrates tumors, has an interferon-γ-driven mechanism of action and triggers adaptive immune responses. In addition, afami-cel has an acceptable benefit-risk profile, with early and durable responses, especially in patients with metastatic SS. Although the small trial size limits conclusions that can be drawn, the results warrant further testing in larger studies.


Assuntos
Antígenos de Neoplasias , Neoplasias de Cabeça e Pescoço , Masculino , Humanos , Proteínas de Neoplasias , Antígenos HLA-A , Terapia Baseada em Transplante de Células e Tecidos , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos
3.
BMC Cardiovasc Disord ; 22(1): 49, 2022 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-35152886

RESUMO

BACKGROUND: In a Phase I study treatment with the serum amyloid P component (SAP) depleter miridesap followed by monoclonal antibody to SAP (dezamizumab) showed removal of amyloid from liver, spleen and kidney in patients with systemic amyloidosis. We report results from a Phase 2 study and concurrent immuno-positron emission tomography (PET) study assessing efficacy, pharmacodynamics, pharmacokinetics, safety and cardiac uptake (of dezamizumab) following the same intervention in patients with cardiac amyloidosis. METHODS: Both were uncontrolled open-label studies. After SAP depletion with miridesap, patients received ≤ 6 monthly doses of dezamizumab in the Phase 2 trial (n = 7), ≤ 2 doses of non-radiolabelled dezamizumab plus [89Zr]Zr-dezamizumab (total mass dose of 80 mg at session 1 and 500 mg at session 2) in the immuno-PET study (n = 2). Primary endpoints of the Phase 2 study were changed from baseline to follow-up (at 8 weeks) in left ventricular mass (LVM) by cardiac magnetic resonance imaging and safety. Primary endpoint of the immuno-PET study was [89Zr]Zr-dezamizumab cardiac uptake assessed via PET. RESULTS: Dezamizumab produced no appreciable or consistent reduction in LVM nor improvement in cardiac function in the Phase 2 study. In the immuno-PET study, measurable cardiac uptake of [89Zr]Zr-dezamizumab, although seen in both patients, was moderate to low. Uptake was notably lower in the patient with higher LVM. Treatment-associated rash with cutaneous small-vessel vasculitis was observed in both studies. Abdominal large-vessel vasculitis after initial dezamizumab dosing (300 mg) occurred in the first patient with immunoglobulin light chain amyloidosis enrolled in the Phase 2 study. Symptom resolution was nearly complete within 24 h of intravenous methylprednisolone and dezamizumab discontinuation; abdominal computed tomography imaging showed vasculitis resolution by 8 weeks. CONCLUSIONS: Unlike previous observations of visceral amyloid reduction, there was no appreciable evidence of amyloid removal in patients with cardiac amyloidosis in this Phase 2 trial, potentially related to limited cardiac uptake of dezamizumab as demonstrated in the immuno-PET study. The benefit-risk assessment for dezamizumab in cardiac amyloidosis was considered unfavourable after the incidence of large-vessel vasculitis and development for this indication was terminated. Trial registration NCT03044353 (2 February 2017) and NCT03417830 (25 January 2018).


Assuntos
Amiloidose , Anticorpos Monoclonais , Ácidos Carboxílicos , Cardiomiopatias , Tomografia por Emissão de Pósitrons , Pirrolidinas , Componente Amiloide P Sérico , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Amiloidose/sangue , Amiloidose/diagnóstico por imagem , Amiloidose/tratamento farmacológico , Amiloidose/imunologia , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Ácidos Carboxílicos/efeitos adversos , Ácidos Carboxílicos/uso terapêutico , Cardiomiopatias/sangue , Cardiomiopatias/diagnóstico por imagem , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/imunologia , Quimioterapia Combinada , Imageamento por Ressonância Magnética , Miocárdio/metabolismo , Miocárdio/patologia , Valor Preditivo dos Testes , Pirrolidinas/efeitos adversos , Pirrolidinas/uso terapêutico , Componente Amiloide P Sérico/antagonistas & inibidores , Componente Amiloide P Sérico/imunologia , Fatores de Tempo , Resultado do Tratamento , Reino Unido , Estados Unidos , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
4.
Front Physiol ; 11: 324, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32425806

RESUMO

Deposition of amyloid in the heart can lead to cardiac dilation and impair its pumping ability. This ultimately leads to heart failure with worsening symptoms of breathlessness and fatigue due to the progressive loss of elasticity of the myocardium. Biomarkers linked to the clinical deterioration can be crucial in developing effective treatments. However, to date the progression of cardiac amyloidosis is poorly characterized. There is an urgent need to identify key predictors for disease progression and cardiac tissue function. In this proof of concept study, we estimate a group of new markers based on mathematical models of the left ventricle derived from routine clinical magnetic resonance imaging and follow-up scans from the National Amyloidosis Center at the Royal Free in London. Using mechanical modeling and statistical classification, we show that it is possible to predict disease progression. Our predictions agree with clinical assessments in a double-blind test in six out of the seven sample cases studied. Importantly, we find that multiple factors need to be used in the classification, which includes mechanical, geometrical and shape features. No single marker can yield reliable prediction given the complexity of the growth and remodeling process of diseased hearts undergoing high-dimensional shape changes. Our approach is promising in terms of clinical translation but the results presented should be interpreted with caution due to the small sample size.

5.
Front Oncol ; 5: 236, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26557502

RESUMO

Atypical teratoid rhabdoid tumors (AT/RTs) are rare pediatric brain tumors characterized by bialleic loss of the SMARCB1 tumor suppressor gene. In contrast to pediatric AT/RT that has a simple genome, very little is known about the adult AT/RT genomic landscape. Using a combination of whole-exome sequencing and high-resolution SNP array in a single adult pituitary AT/RT, we identified a total of 47 non-synonymous mutations, of which 20 were predicted to cause non-conservative amino acid substitutions, in addition to a subclone of cells with trisomy 8. We suggest that adult AT/RT may not be markedly dissimilar to other adult brain tumors where mutations in a range of genes, reflecting the functional specialization of different brain regions, but including SMARCB1 inactivation, may be required for its pathogenesis.

6.
Expert Opin Investig Drugs ; 23(5): 629-45, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24579771

RESUMO

INTRODUCTION: The majority of human sarcomas, particularly soft tissue sarcomas, are relatively resistant to traditional cytotoxic therapies. The proof-of-concept study by Ray-Coquard et al., using the Nutlin human double minute (HDM)2-binding antagonist RG7112, has recently opened a new chapter in the molecular targeting of human sarcomas. AREAS COVERED: In this review, the authors discuss the challenges and prospective remedies for minimizing the significant haematological toxicities of the cis-imidazole Nutlin HDM2-binding antagonists. Furthermore, they also chart the future direction of the development of p53-reactivating (p53-RA) drugs in 12q13-15 amplicon sarcomas and as potential chemopreventative therapies against sarcomagenesis in germ line mutated TP53 carriers. Drawing lessons from the therapeutic use of Imatinib in gastrointestinal tumours, the authors predict the potential pitfalls, which may lie in ahead for the future clinical development of p53-RA agents, as well as discussing potential non-invasive methods to identify the development of drug resistance. EXPERT OPINION: Medicinal chemistry strategies, based on structure-based drug design, are required to re-engineer cis-imidazoline Nutlin HDM2-binding antagonists into less haematologically toxic drugs. In silico modelling is also required to predict toxicities of other p53-RA drugs at a much earlier stage in drug development. Whether p53-RA drugs will be therapeutically effective as a monotherapy remains to be determined.


Assuntos
Compreensão , Descoberta de Drogas/tendências , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Sarcoma/tratamento farmacológico , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Previsões , Humanos , Imidazolinas/farmacologia , Imidazolinas/uso terapêutico , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Sarcoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo
7.
Carcinogenesis ; 33(9): 1717-25, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22777959

RESUMO

Hypoxia-inducible factors, HIF-1α and HIF-2α, are expressed in the majority of clear-cell renal cell carcinoma (CC-RCC). In vitro, HIFα isoforms regulate a differential set of genes, and their effects in vivo within CC-RCC tumours may affect outcome. The role of angiogenesis and HIFα transcriptional products, including those involved in cell metabolism and morphological dedifferentiation have not been extensively investigated and might have relevance to the development of antiangiogenic or anti-HIFα trials in primary CC-RCC, either before or after radical nephrectomy. We analysed 168 consecutive clear-cell renal tumours from 1983 to 1999 within tissue microarrays and assessed expression of HIF-1α and HIF-2α together with the protein expression of seven of their target genes (BNIP3, CA9, Cyclin D1, GLUT-1, LDH5, Oct-4 and VEGF). The expression of these factors was compared with patient overall survival and CD31 angiogenesis. We found that HIFα antigenicity deteriorated with the age of the paraffin block (P < 0.0001) and in tumours from 1983 to 1992 was deemed not to be reliable. Similar findings were found in aged archival osteosarcoma samples. This might have important implications for retrospective biomarker studies that rely on archival tissue material. HIF-1α(HIGH)/HIF-2α(LOW) tumours had a worse overall survival compared with HIF-1α(LOW)/HIF-2α(LOW) tumours (P = 0.04). Surprisingly, on multivariate analysis, high levels of CD31(+) angiogenesis was shown to be an independent prognostic marker of increased overall survival (P = 0.003). We propose that better differentiation of vascular endothelium may be a reflection of a greater production of vessel stabilization factors versus pro-angiogenic factors, and therefore a less aggressive phenotype.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Carcinoma de Células Renais/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias Renais/tratamento farmacológico , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/análise , Biomarcadores Tumorais/análise , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Ciclina D1/análise , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Pessoa de Meia-Idade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Prognóstico
8.
Cancer Metastasis Rev ; 31(3-4): 689-98, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22706846

RESUMO

Cancer biologists seem to have overlooked tumor metabolism in their research endeavors over the last 80 years of the last century, only to have "rediscovered Warburg" (Warburg et al. 1930; Warburg, Science 123(3191):309-314, 1956) within the first decade of the twenty-first century, as well as to suggest the importance of other, non-glucose-dependent, metabolic pathways such as such as fatty acid de novo synthesis and catabolism (ß-oxidation) (Mashima et al., Br J Cancer 100:1369-1372, 2009) and glutamine catabolism (glutaminolysis) (DeBerardinis et al., Proc Nat Acad Sci 104(49):19345-19350, 2007). These non-glucose metabolic pathways seem to be just as important as the Warburg effect, if not potentially more so in human cancer. The purpose of this review is to highlight the importance of fatty acid metabolism in cancer cells and, where necessary, identify gaps in current knowledge and postulate hypothesis based upon findings in the cellular physiology of metabolic diseases and normal cells.


Assuntos
Neoplasias/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Transformação Celular Neoplásica , Citoplasma/metabolismo , Ácidos Graxos/metabolismo , Humanos , Oxirredução
9.
Pediatr Blood Cancer ; 58(3): 327-33, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21990244

RESUMO

The similarities between human and canine osteosarcoma with regard to histology, biological behavior and molecular genetic alterations suggest that the dog provides a supplementary model for the development and preclinical testing of novel therapeutics. Counter intuitively, careful examination of the differences between OS in the two species may also be rewarding in terms of increasing our understanding of the pathogenesis of this cancer. This review will discuss the arguments in favor of the "dog model" and outline how the evaluation of treatment strategies in dogs has indicated avenues for improvement of protocols for human patients.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/veterinária , Modelos Animais de Doenças , Doenças do Cão/fisiopatologia , Cães , Osteossarcoma/tratamento farmacológico , Osteossarcoma/veterinária , Animais , Biomarcadores Tumorais , Neoplasias Ósseas/fisiopatologia , Descoberta de Drogas , Humanos , Salvamento de Membro , Terapia de Alvo Molecular , Osteossarcoma/fisiopatologia , Fisiologia Comparada , Pesquisa Translacional Biomédica
10.
Eur J Cancer ; 46(15): 2671-3, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20800475

RESUMO

BACKGROUND: A phase 2 study of vatalanib (PTK787/ZK222584) an oral tyrosine kinase inhibitor of VEGFR 1, 2 and 3 was undertaken in patients with metastatic melanoma. METHODS: Adults with pathologically confirmed metastatic melanoma, WHO Performance status 0-2, and adequate haematological, hepatic and renal function, were treated with vatalanib until disease progression. The trial used Fleming's single stage design. RESULTS: Tumour control rate (CR+PR+SD) was 35% at 16 weeks, with objective response seen in only 1 patient. Median progression-free survival was 1.8 months (95% CI 1.8-3.7 months) and median overall survival was 6.5 months (95% CI 3.9-10.2 months). CONCLUSION: Vatalanib stabilised disease in a proportion of patients, although overall survival was disappointing.


Assuntos
Antineoplásicos/administração & dosagem , Melanoma/tratamento farmacológico , Melanoma/secundário , Ftalazinas/administração & dosagem , Piridinas/administração & dosagem , Neoplasias Cutâneas/tratamento farmacológico , Administração Oral , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
11.
J Oncol ; 2010: 757908, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20652061

RESUMO

In cultured clear-cell renal carcinoma (CCRCC) 786-0 cells transfected with HIF1alpha (HIF-1+), HIF-2alpha (HIF-2+), or empty vector (EV), no significant differences were observed in the growth rates in vitro, but when grown in vivo as xenografts HIF-2alpha significantly increased, and HIF-1alpha significantly decreased growth rates, compared to EV tumors. Factors associated with proliferation were increased and factors associated with cell death were decreased in HIF-2+ tumors. Metabolite profiles showed higher glucose and lower lactate and alanine levels in the HIF-2+ tumors whilst immunostaining demonstrated higher pyruvate dehydrogenase and lower pyruvate dehydrogenase kinase 1, compared to control tumors. Taken together, these results suggest that overexpression of HIF-2alpha in CCRCC 786-0 tumors regulated growth both by maintaining a low level of glycolysis and by allowing more mitochondrial metabolism and tolerance to ROS induced DNA damage. The growth profiles observed may be mediated by adaptive changes to a more oxidative phenotype.

12.
Expert Rev Anticancer Ther ; 9(11): 1583-98, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19895243

RESUMO

Metastatic melanoma (MM) carries a dismal prognosis, as it is largely resistant to conventional cytotoxic chemotherapy, biochemotherapy and immunotherapy. There is, therefore, a pressing need to identify new, effective treatments to improve outcomes from MM. Innovative approaches in oncology drug development include anti-angiogenic strategies, in the form of monoclonal antibodies and small-molecule kinase inhibitors. In this review we aim to present current concepts and controversies surrounding the role of angiogenesis and anti-angiogenic therapies in MM, alluding to other tumor types in which increasing knowledge may supply avenues for future directions in melanoma research and management. An overview of angiogenesis and its importance in melanoma progression is presented, highlighting the key molecules that represent potential therapeutic targets. The results of using anti-angiogenic strategies in preclinical and clinical trials are discussed and future perspectives for anti-angiogenic therapies in MM are considered.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Melanoma/irrigação sanguínea , Melanoma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Animais , Resistencia a Medicamentos Antineoplásicos , Humanos , Melanoma/patologia , Neovascularização Patológica/patologia , Neoplasias Cutâneas/patologia
13.
Nat Rev Clin Oncol ; 6(8): 478-87, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19546865

RESUMO

FDA approval of the multitargeted, antiangiogenic tyrosine kinase inhibitors (TKIs) sunitinib and sorafenib, and the serine and threonine mammalian target of rapamycin inhibitor, temsirolimus, has revolutionized the management of metastatic clear-cell renal-cell carcinoma (CC-RCC). The inability of these targeted therapies to provide durable complete responses, however, is a serious limiting factor to their clinical usefulness. Although immunotherapeutic approaches in advanced disease are increasingly regarded as a historical treatment paradigm, we propose that a fundamental understanding of immunobiology in CC-RCC can improve the selection of patients for high-dose intravenous interleukin 2 and facilitate the development of novel immunotherapeutic strategies. In our opinion, immunotherapeutic strategies have an important place in the management of advanced CC-RCC in the era of biological targeted therapy.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/secundário , Imunoterapia , Interferon-alfa/uso terapêutico , Interleucina-2/uso terapêutico , Neoplasias Renais/terapia , Inibidores da Angiogênese/uso terapêutico , Antígenos de Neoplasias/análise , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/análise , Vacinas Anticâncer/uso terapêutico , Anidrase Carbônica IX , Anidrases Carbônicas/análise , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/terapia , Ensaios Clínicos como Assunto/estatística & dados numéricos , Terapia Combinada , Citocinas/uso terapêutico , Sistemas de Liberação de Medicamentos , Humanos , Imunoconjugados/uso terapêutico , Infusões Intravenosas , Interferon-alfa/administração & dosagem , Interleucina-2/administração & dosagem , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Metanálise como Assunto , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/fisiologia , Seleção de Pacientes , Inibidores de Proteínas Quinases/uso terapêutico , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia
14.
Pediatr Blood Cancer ; 52(7): 796-803, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19202566

RESUMO

BACKGROUND: Supratentorial PNET (sPNET) are rare CNS tumors of embryonal origin arising in children and adults. The treatment of sPNET for all age groups at our cancer center has been based on the management of medulloblastoma (MB), involving neurosurgical debulking followed by cranio-spinal irradiation (CSI) and systemic chemotherapy. METHODS: Medical records were reviewed to gather demographic and clinical data about all embryonal CNS tumors in children and adults from 2001 to 2007. Tumor pathology, clinical management and survival data were also assessed, particularly as regards those patients who received the Packer chemotherapy regimen for either sPNET or MB. RESULTS: Eleven patients (five children and six adults) were identified with non-pineal sPNET, three children with pineal sPNET, and 19 patients (18 children and 1 adult) with MB. There was no difference in overall survival (OS) rates between pediatric and adult sPNET. When all sPNET were compared to all MB, 5-year OS was 14% versus 73%, respectively, but was only 9% for non-pineal sPNET. When only considering those patients treated with the Packer chemotherapy regimen, the 5-year OS was 12% for sPNET versus 79% for MB. CONCLUSION: This retrospective study demonstrates that non-pineal sPNET are clinically distinct from MB and are resistant to the Packer chemotherapy regimen. We suggest that it is time to reconsider the use of this regimen in teenage and young adult non-pineal sPNET and to investigate the utility of alternative approaches.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Cerebelares/terapia , Irradiação Craniana , Meduloblastoma/terapia , Tumores Neuroectodérmicos Primitivos/terapia , Neoplasias Supratentoriais/terapia , Adolescente , Adulto , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/radioterapia , Criança , Cisplatino/administração & dosagem , Seguimentos , Humanos , Lomustina/administração & dosagem , Meduloblastoma/tratamento farmacológico , Meduloblastoma/radioterapia , Estadiamento de Neoplasias , Tumores Neuroectodérmicos Primitivos/tratamento farmacológico , Tumores Neuroectodérmicos Primitivos/radioterapia , Estudos Retrospectivos , Neoplasias Supratentoriais/tratamento farmacológico , Neoplasias Supratentoriais/radioterapia , Taxa de Sobrevida , Resultado do Tratamento , Vincristina/administração & dosagem , Adulto Jovem
15.
Oncologist ; 14(1): 52-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19147692

RESUMO

Cytoreductive nephrectomy in combination with adjuvant immunotherapy is an established treatment option for selected patients with metastatic clear-cell renal cell carcinoma (mCC-RCC). Multitargeted antiangiogenic and mammalian target of rapamycin tyrosine kinase inhibitors (TKIs) are now established treatment paradigms in patients with mCC-RCC. Given that all the recent seminal TKI trials in mCC-RCC provide no evidence base for the use of cytoreductive nephrectomy in the TKI era, it is not presently clear where such a surgical approach fits into the treatment paradigm. This review summarizes the evidence for the management of mCC-RCC and outlines novel approaches to be tested within future trials if the initial proposed phase III trials in this setting, using sunitinib, are successful. Overall, two principal questions need addressing. First, is cytoreductive nephrectomy necessary in the TKI era? Second, if so, what is the most appropriate scheduling of TKI therapy with cytoreductive nephrectomy?


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/cirurgia , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/cirurgia , Inibidores de Proteínas Quinases/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/enzimologia , Carcinoma de Células Renais/patologia , Ensaios Clínicos Fase III como Assunto , Humanos , Indóis/uso terapêutico , Neoplasias Renais/enzimologia , Neoplasias Renais/patologia , Metástase Neoplásica , Nefrectomia/métodos , Pirróis/uso terapêutico , Sunitinibe
16.
Cancer Res ; 67(23): 11244-53, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18056450

RESUMO

The vascular endothelial growth factor (VEGF) plays a key role in tumor angiogenesis. However, clinical trials targeting the VEGF pathway are often ineffective, suggesting that other factors/pathways are also important in tumor angiogenesis. We have previously shown that the Notch ligand Delta-like 4 (DLL4) is up-regulated in tumor vasculature. Here, we show that DLL4, when expressed in tumor cells, functions as a negative regulator of tumor angiogenesis by reducing the number of blood vessels in all five types of xenografts, but acts as a positive driver for tumor growth in two of them (human glioblastoma and prostate cancer). The growth of in vivo models was not related to the effects on growth in vitro. DLL4 expressed in the tumor cells activated Notch signaling in host stromal/endothelial cells, increased blood vessel size, and improved vascular function within tumors. The promotion of tumor growth was, to some extent, due to a reduction of tumor hypoxia and apoptosis. DLL4-expressing tumor cells responded to anti-VEGF therapy with bevacizumab. A soluble form of DLL4 (D4ECD-Fc) blocked tumor growth in both bevacizumab-sensitive and bevacizumab-resistant tumors by disrupting vascular function despite increased tumor vessel density. In addition, we show that DLL4 is up-regulated in tumor cells and tumor endothelial cells of human glioblastoma. Our findings provide a rational basis for the development of novel antiangiogenic strategies via blockade of DLL4/Notch signaling and suggest that combined approaches for interrupting both DLL4 and VEGF pathways may improve antiangiogenic therapy.


Assuntos
Endotélio Vascular/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/patologia , Receptores Notch/metabolismo , Veias Umbilicais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Bevacizumab , Células CHO , Proteínas de Ligação ao Cálcio , Proliferação de Células , Células Cultivadas , Cricetinae , Cricetulus , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neoplasias Experimentais/metabolismo , Neovascularização Patológica/patologia , Transdução de Sinais , Células Estromais/patologia , Transplante Heterólogo , Veias Umbilicais/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...