Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
2.
Klin Padiatr ; 228(3): 113-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27135271

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) and diffuse intrinsic pontine glioma (DIPG) belong to the most aggressive cancers in children with poor prognosis and limited therapeutic options. Therapeutic targeting of epigenetic proteins may offer new treatment options. Preclinical studies identified Enhancer of Zeste Homolog 2 (EZH2) within polycomb repressor complex 2 (PRC2) as a potential epigenetic anti-tumor target in adult GBM cells but similar inhibition studies in pediatric GBM/DIPG were still missing. Moreover, approximately 30% of pediatric high grade gliomas (pedHGG) including GBM and DIPG harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3) which is correlated with poor outcome and was shown to influence EZH2 function. PATIENTS, MATERIALS AND METHODS: The present study investigated the correlation of expression of EZH2 and other PRC2 genes (EZH1, SUZ12, EED) with overall survival of pediatric GBM patients and the cytotoxic impact of EZH2 inhibition by the novel agent Tazemetostat in pediatric GBM/DIPG cells harboring either a H3.3 mutation or a H3 wildtype. RESULTS: EZH2 gene expression does not correlate with survival of pedHGG patients, and EZH2 inhibition does not induce significant cytotoxicity in pedHGG cells independently of H3.3 mutations. DISCUSSION AND CONCLUSION: We suggest that EZH2 inhibition might not offer an effective single agent treatment option for paedHGG patients. However, the therapeutic efficacy in combination with cytotoxic and/or other epigenetically active agents still has to be elucidated.


Assuntos
Benzamidas/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Análise Mutacional de DNA , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Glioblastoma/genética , Glioblastoma/patologia , Glioma/genética , Glioma/patologia , Histonas/genética , Piridonas/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Adolescente , Compostos de Bifenilo , Linhagem Celular Tumoral , Criança , Pré-Escolar , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Masculino , Morfolinas , Estatística como Assunto , Adulto Jovem
3.
Gene Ther ; 23(7): 615-26, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27092941

RESUMO

Engineering autologous or allogeneic T cells to express a suicide gene can control potential toxicity in adoptive T-cell therapies. We recently reported the development of a novel human suicide gene system that is based on an orphan human cytochrome P450 enzyme, CYP4B1, and the naturally occurring alkylator prodrug 4-ipomeanol. The goal of this study was to systematically develop a clinically applicable self-inactivating lentiviral vector for efficient co-expression of CYP4B1 as an ER-located protein with two distinct types of cell surface proteins, either MACS selection genes for donor lymphocyte infusions after allogeneic stem cell transplantation or chimeric antigen receptors for retargeting primary T cells. The U3 region of the myeloproliferative sarcoma virus in combination with the T2A site was found to drive high-level expression of our CYP4B1 mutant with truncated CD34 or CD271 as MACS suitable selection markers. This lentiviral vector backbone was also well suited for co-expression of CYP4B1 with a codon-optimized CD19 chimeric antigen receptor (CAR) construct. Finally, 4-ipomeanol efficiently induced apoptosis in primary T cells that co-express mutant CYP4B1 and the divergently located MACS selection and CAR genes. In conclusion, we here developed a clinically suited lentiviral vector that supports high-level co-expression of cell surface proteins with a potent novel human suicide gene.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Genes Transgênicos Suicidas , Terapia Genética/métodos , Imunoterapia Adotiva/métodos , Antígenos CD34/genética , Antígenos CD34/metabolismo , Apoptose , Hidrocarboneto de Aril Hidroxilases/metabolismo , Células Cultivadas , Vetores Genéticos/genética , Células HEK293 , Humanos , Células Jurkat , Lentivirus/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Terpenos/uso terapêutico
4.
Klin Padiatr ; 225(6): 347-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24166086

RESUMO

We report on a case of Pseudomonas aeruginosa sepsis and consecutive lung abscess in a 13-year-old patient with acute B-cell leukemia. At first, radiographic findings strongly suggested presence of pulmonary aspergilloma and only microbiological testing of the surgically enucleated mass revealed the correct underlying pathogen and confirmed final diagnosis.


Assuntos
Leucemia de Células B/diagnóstico , Abscesso Pulmonar/diagnóstico , Micetoma/diagnóstico , Infecções Oportunistas/diagnóstico , Infecções por Pseudomonas/diagnóstico , Pseudomonas aeruginosa , Aspergilose Pulmonar/diagnóstico , Adolescente , Diagnóstico Diferencial , Humanos , Pulmão/patologia , Pulmão/cirurgia , Abscesso Pulmonar/patologia , Abscesso Pulmonar/cirurgia , Masculino , Infecções Oportunistas/patologia , Infecções Oportunistas/cirurgia , Tomografia Computadorizada por Raios X
5.
Cancer Gene Ther ; 20(9): 514-20, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23949282

RESUMO

The antifolate methotrexate (MTX) is an important chemotherapeutic agent for treatment of osteosarcoma. This drug is converted intracellularly into polyglutamate derivates by the enzyme folylpolyglutamate synthase (FPGS). MTX polyglutamates show an enhanced and prolonged cytotoxicity in comparison to the monoglutamate. In the present study, we proved the hypothesis that transfer of the human fpgs gene into osteosarcoma cells may augment their MTX sensitivity. For this purpose, we employed the human osteocalcin (OC) promoter, which had shown marked osteosarcoma specificity in promoter studies using different luciferase assays in osteosarcoma and non-osteosarcoma cell lines. A recombinant lentiviral vector was generated with the OC promoter driving the expression of fpgs and the gene for enhanced green fluorescent protein (egfp), which was linked to fpgs by an internal ribosomal entry site (IRES). As the vector backbone contained only a self-inactivating viral LTR promoter, any interference of the OC promoter by unspecific promoter elements was excluded. We tested the expression of FPGS and enhanced green fluorescent protein (EGFP) after lentiviral transduction in various osteosarcoma cell lines (human MG-63 cells and TM 791 cells; rat osteosarcoma (ROS) 17/2.8 cells) and non-osteogenic tumor cell lines (293T human embryonic kidney cells, HeLa human cervix carcinoma cells). EGFP expression and MTX sensitivity were assessed in comparison with non-transduced controls. Whereas the OC promoter failed to enhance MTX sensitivity via FPGS expression in non-osteogenic tumor cell lines, the OC promoter mediated a markedly increased MTX cytotoxicity in all osteosarcoma cell lines after lentiviral transduction. The present chemotherapy-enhancing gene therapy system may have great potential to overcome in future MTX resistance in human osteosarcomas.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Neoplasias Ósseas/genética , Expressão Gênica/efeitos dos fármacos , Metotrexato/farmacologia , Osteossarcoma/genética , Peptídeo Sintases/genética , Linhagem Celular Tumoral , Clonagem Molecular , Ordem dos Genes , Genes Reporter , Vetores Genéticos/genética , Humanos , Lentivirus/genética , Especificidade de Órgãos/genética , Osteocalcina/genética , Osteocalcina/metabolismo , Regiões Promotoras Genéticas , Transdução Genética , Transfecção , Células Tumorais Cultivadas
6.
Br J Cancer ; 109(4): 844-51, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23868007

RESUMO

BACKGROUND: High-grade glioma (HGG) of the cerebellum accounts for only 5% of paediatric HGG. Since little is known about these tumours, the present study aimed at their further characterisation. METHODS: Twenty-nine paediatric patients with centrally reviewed cerebellar HGG were identified from the HIT-GBM/HIT-HGG database. Clinical and epidemiological data were compared with those of 180 paediatric patients with cortical HGG. RESULTS: Patients with cerebellar tumours were younger (median age of 7.6 vs 11.7 years, P=0.028), but both groups did not differ significantly with regard to gender, tumour predisposing syndromes, secondary HGG, primary metastasis, tumour grading, extent of tumour resection, chemotherapy regimen, or radiotherapy. Except for an increased incidence of anaplastic pilocytic astrocytoma (APA) in the cerebellar subset (20.7% vs 3.3%; P<0.001), histological entities were similarly distributed in both groups. As expected, tumour grading had a prognostic relevance on survival. Compared with cortical HGG, overall survival in the cerebellar location was significantly worse (median overall survival: 0.92 ± 0.02 vs 2.03 ± 0.32 years; P=0.0064), and tumour location in the cerebellum had an independent poor prognostic significance as shown by Cox-regression analysis (P=0.019). CONCLUSION: High-grade glioma represents a group of tumours with an obviously site-specific heterogeneity associated with a worse survival in cerebellar location.


Assuntos
Neoplasias Cerebelares/diagnóstico , Glioma/diagnóstico , Adolescente , Distribuição por Idade , Astrocitoma/diagnóstico , Astrocitoma/epidemiologia , Astrocitoma/patologia , Estudos de Casos e Controles , Neoplasias Cerebelares/epidemiologia , Neoplasias Cerebelares/patologia , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Ganglioglioma/diagnóstico , Ganglioglioma/epidemiologia , Ganglioglioma/patologia , Glioblastoma/diagnóstico , Glioblastoma/epidemiologia , Glioblastoma/patologia , Glioma/epidemiologia , Glioma/patologia , Humanos , Lactente , Masculino , Gradação de Tumores , Oligodendroglioma/diagnóstico , Oligodendroglioma/epidemiologia , Oligodendroglioma/patologia , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Distribuição por Sexo , Neoplasias Supratentoriais/diagnóstico , Neoplasias Supratentoriais/epidemiologia , Neoplasias Supratentoriais/patologia
7.
Strahlenther Onkol ; 189(8): 693-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23754473

RESUMO

Primary metastatic diffuse intrinsic pontine glioma (DIPG) is relatively rare and associated with a dismal prognosis. Combining craniospinal irradiation (CSI) with concurrent temozolomide and nimotuzumab therapy may slightly improve tumor control and overall survival. However, little is known about the feasibility and toxicity of this treatment approach. Here, we describe the case of an 8-year-old girl with primary metastatic DIPG who received craniospinal radiotherapy, a local boost, and concurrent temozolomide and nimotuzumab treatment based on an individual therapy recommendation. Radiotherapy could be completed without any interruption. However, concurrent temozolomide had to be disrupted several times due to considerable acute myelotoxicity (grade III-IV).Maintenance immunochemotherapy could be started with a delay of 5 days and was performed according to treatment schedule. The disease could be stabilized for a few months. A routine MRI scan finally depicted disease progression 5.7 months after the start of irradiation. The patient died 1.9 months later.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias do Tronco Encefálico/terapia , Quimiorradioterapia/métodos , Glioma/secundário , Glioma/terapia , Neoplasias da Coluna Vertebral/secundário , Neoplasias da Coluna Vertebral/terapia , Anticorpos Monoclonais Humanizados/administração & dosagem , Criança , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Evolução Fatal , Feminino , Humanos , Temozolomida , Resultado do Tratamento
8.
Klin Padiatr ; 224(3): 156-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22513795

RESUMO

Methotrexate (MTX) is commonly administered in high doses for treatment of childhood acute lymphoblastic leukemia (ALL). The aim of this analysis was to study the influence of 2 common MTHFR polymorphisms (MTHFR 677C>T and 1298 A>C) on MTX toxicity in children with ALL.Retrospective analysis of 129 MTX courses in 34 pediatric patients with ALL.677C>T variants (CT or TT) were found in 19 (14 heterozygous, 5 homozygous) and 1298A>C variants (AC or CC) in 20 (16 heterozygous, 4 homozygous) patients. The MTHFR 677C>T wild type was associated with an increased frequency of grade III and IV leukopenia (60% vs. 31%, p<0.05) compared to the variants. The rate of severe infections (21% vs. 0%, p<0.05) and grade III-IV anemia (26% vs. 5%, p<0.05) was increased in carriers of the MTHFR 677C>T wild type compared to patients with the TT variant. Grade III-IV anemia was more frequent in patients with the MTHFR 1298A>C CC variant compared to the wild type (56% vs. 21%, p<0.05). The differences were not significant in a patient-based analysis.MTX related toxicity might be influenced by the MTHFR 677C>T or the MTHFR 1298A>C polymorphisms. Differences in MTX toxicity are only partially explainable by these 2 polymorphisms.


Assuntos
Alelos , Antimetabólitos Antineoplásicos/toxicidade , Metotrexato/toxicidade , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo Genético/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Antimetabólitos Antineoplásicos/farmacocinética , Antimetabólitos Antineoplásicos/uso terapêutico , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Triagem de Portadores Genéticos , Genótipo , Homozigoto , Humanos , Leucopenia/induzido quimicamente , Leucopenia/genética , Masculino , Metotrexato/farmacocinética , Metotrexato/uso terapêutico , Projetos Piloto , Estudos Retrospectivos , Resultado do Tratamento
9.
Neuro Oncol ; 13(10): 1049-58, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21784756

RESUMO

Pediatric high-grade gliomas (HGGs)--including glioblastoma multiforme, anaplastic astrocytoma, and diffuse intrinsic pontine glioma--are difficult to treat and are associated with an extremely poor prognosis. There are no effective chemotherapeutic regimens for the treatment of pediatric HGG, but many new treatment options are in active investigation. There are crucial molecular differences between adult and pediatric HGG such that results from adult clinical trials cannot simply be extrapolated to children. Molecular markers overexpressed in pediatric HGG include PDGFRα and P53. Amplification of EGFR is observed, but to a lesser degree than in adult HGG. Potential molecular targets and new therapies in development for pediatric HGG are described in this review. Research into bevacizumab in pediatric HGG indicates that its activity is less than that observed in adult HGG. Similarly, tipifarnib was found to have minimal activity in pediatric HGG, whereas gefitinib has shown greater effects. After promising phase I findings in children with primary CNS tumors, the integrin inhibitor cilengitide is being investigated in a phase II trial in pediatric HGG. Studies are also ongoing in pediatric HGG with 2 EGFR inhibitors: cetuximab and nimotuzumab. Other novel treatment modalities under investigation include dendritic cell-based vaccinations, boron neutron capture therapy, and telomerase inhibition. While the results of these trials are keenly awaited, the current belief is that multimodal therapy holds the greatest promise. Research efforts should be directed toward building multitherapeutic regimens that are well tolerated and that offer the greatest antitumor activity in the setting of pediatric HGG.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Oncologia/tendências , Adolescente , Neoplasias Encefálicas/patologia , Criança , Ensaios Clínicos como Assunto , Glioma/patologia , Humanos , Gradação de Tumores
10.
Klin Padiatr ; 221(6): 374-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19890790

RESUMO

BACKGROUND: The risk of severe complications or death during the initial period of acute leukemia was markedly decreased due to the progress in pediatric oncology and use of simple measures like hyperhydration, forced diuresis, treatment of hyperuricemia, correction of electrolyte and coagulation disturbances and the careful use of antileukemic drugs. The incidence of leukostasis and tumor lysis syndrome depends on absolute initial white blood cell counts and the underlying type of leukemia. Leukapheresis or exchange transfusion may improve the prognosis of high risk patients. METHODS: Records of all pediatric patients who were newly diagnosed with acute leukemia between 1 / 1998 und 12 / 2008 were retrospectively reviewed for presence of hyperleukocytosis(white blood cell count > 100 GPT / l) at diagnosis and subsequent leukapheresis or exchange transfusion in regards to the clinical outcome. RESULTS: At diagnosis 11 (14 % ) of 77 children with acute leukemia (7 acute lymphoblastic leukemia / ALL; 4 acute myeloblastic leukemia /AML) had hyperleukocytosis. 4 patients (2 ALL, 2 AML) received exchange transfusion and 2 others (1 ALL, 1 AML) underwent leukapheresis. Marked cytoreduction was achieved in all patients within 24 h after therapy initiation. There were no procedure-related adverse events. Symptoms due to hyperleukocytosis markedly improved after cytoreduction. CONCLUSION: Leukapheresis or exchange transfusion together with conservative management and specific oncological therapy may contribute to rapid leukocyte reduction with acceptable risk. The exact impact of leukapheresis or exchange transfusion on short and long term outcome in pediatric patients with acute leukemia and initial hyperleukocytosis has to be evaluated in future multicentre studies or by the formation of clinical registries.


Assuntos
Transfusão Total , Leucaférese , Leucemia Mieloide Aguda/terapia , Leucocitose/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Criança , Pré-Escolar , Terapia Combinada , Intervalo Livre de Doença , Feminino , Humanos , Lactente , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/mortalidade , Contagem de Leucócitos , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Estudos Retrospectivos
11.
Klin Padiatr ; 218(3): 177-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16688676

RESUMO

Caspofungin was used for the first time with trimethoprim/sulfamethoxazole (TMP/SMX) for treatment of high risk Pneumocystis jiroveci pneumonia (PCP) in a pediatric immunocompromised patient. Despite the need for mechanical ventilation, the pediatric patient with relapsed acute lymphoblastic leukemia improved within nine days of treatment and showed no major side effects. The apparent relative lack of toxicity and of pharmacokinetic drug interactions makes caspofungin an attractive agent.


Assuntos
Antifúngicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Infecções Oportunistas/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Pneumocystis carinii , Pneumonia por Pneumocystis/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Combinação Trimetoprima e Sulfametoxazol/uso terapêutico , Antibioticoprofilaxia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Contagem de Linfócito CD4 , Caspofungina , Pré-Escolar , Quimioterapia Combinada , Equinocandinas , Seguimentos , Humanos , Lipopeptídeos , Masculino , Infecções Oportunistas/imunologia , Pneumonia por Pneumocystis/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Indução de Remissão , Tomografia Computadorizada por Raios X
12.
Pediatr Blood Cancer ; 46(7): 767-72, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16550534

RESUMO

BACKGROUND: Monitoring renal function is crucial in children undergoing chemotherapy. To date, a combination of routine serum creatinine (SCR) monitoring with occasional determination of creatinine clearance ratio (CCR) is widely used as clinical standard for this purpose. Both methods have their limitations regarding diagnostic value (SCR) or practicability (CCR), especially in young children. Diagnostic alternatives, such as glomerular filtration rate (GFR) estimation formulas have not been proved to be superior. The aim of the study was to evaluate whether serum cystatin C (CysC) may have a diagnostic impact on pediatric patients. PROCEDURE: CysC, SCR, several GFR estimation formulas (Counahan-Barratt, Ghazali-Barratt, Schwartz, Shull, Traub), and CCR were studied in 80 pediatric cancer patients (age range: 0.17-17.9 years) during their chemotherapy. Special attention was given to children under the age of 3 in whom accurate urine collection for CCR is difficult. RESULTS: All parameters correlated similarly well with CCR. Total accuracy was 66% and 67% for CysC and SCR, respectively. In very young children (<3 years), correlation with CCR was for CysC r = -0.74 with an area under the curve (AUC) of 0.646, and for SCR r = -0.27 with AUC = 0.594. Total accuracy was 60% for CysC, 50% for SCR. CONCLUSIONS: CysC represents a suitable marker for monitoring renal function in pediatric cancer patients. In young children (<3 years), CysC may have a better diagnostic value than SCR. Future studies should show if CysC can improve renal monitoring by replacing SCR, especially in very young children.


Assuntos
Cistatinas/sangue , Monitoramento de Medicamentos , Testes de Função Renal , Neoplasias/tratamento farmacológico , Adolescente , Fatores Etários , Área Sob a Curva , Biomarcadores/sangue , Criança , Pré-Escolar , Cistatina C , Feminino , Taxa de Filtração Glomerular , Humanos , Lactente , Recém-Nascido , Masculino , Valor Preditivo dos Testes , Sensibilidade e Especificidade
13.
Onkologie ; 27(2): 131-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15138344

RESUMO

OBJECTIVES: Aim of this study was to analyze the feasibility of oncological treatment in pediatric patients belonging to Jehovah's Witnesses and to describe the changing policy in performing transfusions and supportive care measures at two German pediatric cancer institutions. PATIENTS AND METHODS: Over a period of 16 years 21 treatments according to the current cooperative protocols were performed in 14 children of Jehovah's Witnesses. Various hematological supportive care measures such as supplementation with iron, human erythropoietin, interleukin 11, granulocyte colony-stimulating factor and autologous or allogeneic stem cell rescue had been applied. For comparison matched pairs treated in our hospitals not belonging to Jehovah's Witnesses and 50 pediatric and adult oncological patients belonging to Jehovah's Witnesses reviewed from the international literature were analyzed with respect to transfusions and outcome. RESULTS: So far, 9 of 14 children are surviving 16-195 months (median 26 months). During the primary therapy they received markedly less transfusions than the control cohort (-39,1% red blood cell transfusions and -37,5% platelet transfusions). The review of 50 reported cases showed that oncological therapy can also be successfully performed with a restricted transfusion regimen in children and particularly in adults. CONCLUSION: Pediatric cancer patients belonging to Jehovah's Witnesses can be treated similarly to other patients. A restrictive transfusion policy and the broad application of hematopoietic supportive care measures may reduce transfusions. This treatment policy and a continuous collaboration with the Hospital Liaison Committee for Jehovah's Witnesses appears to create an oncological treatment situation with a high compliance of patients and parents where court orders may not be necessary.


Assuntos
Perda Sanguínea Cirúrgica/prevenção & controle , Transfusão de Sangue/ética , Transfusão de Sangue/legislação & jurisprudência , Testemunhas de Jeová , Leucemia/terapia , Administração dos Cuidados ao Paciente/métodos , Recusa do Paciente ao Tratamento/ética , Recusa do Paciente ao Tratamento/legislação & jurisprudência , Transfusão de Sangue/métodos , Criança , Pré-Escolar , Feminino , Alemanha , Humanos , Lactente , Cuidados para Prolongar a Vida/métodos , Masculino , Resultado do Tratamento
14.
Neuropediatrics ; 33(1): 6-9, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11930269

RESUMO

During ganciclovir treatment of an adolescent ependymoma patient two weeks after intracranial implantation of HSVtk retroviral vector producer cells, increasing numbers of peripheral T- and B-cells were found as well as enhanced T-cell activation and elevated serum levels of interleukin 12 and soluble Fas ligand. These findings suggest the systemic activation of the immune system during ganciclovir treatment in our patient. The induction of an immune response by HSVtk/ganciclovir supports the concept of an anti-tumor vaccination effect by prodrug activating gene therapy systems and may open new promising perspectives for enhancing therapeutic efficiency by combined prodrug activating and immunological gene therapy strategies.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Neoplasias Encefálicas/terapia , Ependimoma/terapia , Ganciclovir/farmacologia , Ganciclovir/uso terapêutico , Técnicas de Transferência de Genes , Terapia Genética , Herpesvirus Humano 1/efeitos dos fármacos , Sistema Imunitário/efeitos dos fármacos , Timidina Quinase/efeitos dos fármacos , Adolescente , Antivirais/administração & dosagem , Neoplasias Encefálicas/imunologia , Ependimoma/imunologia , Ganciclovir/administração & dosagem , Herpesvirus Humano 1/imunologia , Humanos , Sistema Imunitário/imunologia , Injeções Intralesionais , Masculino , Timidina Quinase/imunologia
15.
Support Care Cancer ; 9(7): 514-21, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11680831

RESUMO

Infections still remain a major cause of therapy-associated morbidity and death in patients with malignant diseases. To further lower the risk of serious and long-lasting infections by additional supportive measures, detailed information on the frequency and characteristic features of infections is needed. Therefore, patient data from 112 children with acute lymphoblastic leukemia and T-cell lymphoma who were treated according to the COALL-05-92 protocol in our department were analyzed for differences in the frequency and origin of febrile episodes in relation to age, immunological type of leukemia, treatment in group assessed as being at high or low risk of relapse, actual occurrence of relapse, and course of chemotherapy. At the time of diagnosis, low-risk patients more commonly presented with febrile episodes than high-risk patients. In total, patients developed a fever in 313 (24%) of 1,307 evaluated chemotherapy cycles. Febrile episodes were associated with microbiologically or clinically documented infections in 60% of all cases, while in 40% the fever was of unknown origin. Gram-positive pathogens had a markedly higher incidence than gram-negative or fungal ones. The incidence of febrile episodes during therapy appeared to be correlated with certain chemotherapeutic drug combinations. The highest rate was found after high-dose cytarabine and asparaginase causing a long period of leukopenia. However, after other chemotherapy courses with a similar duration of leukopenia the incidence of febrile episodes was significantly lower, suggesting that specific interactions of different chemotherapeutic agents with the immune response might be an important factor in development of infections. Individual factors might also account for an increased incidence of infections, since the number of high-risk patients with recurrent infections was significantly higher than expected on the basis of statistical evaluation. In conclusion, our findings suggest that the risk of infections during chemotherapy may not only be influenced by leukopenia, but that drug-specific effects of the various chemotherapeutic agents and individual factors may also be important contributory factors. These observations must be further expanded in prospective studies so that new tailored supportive care protocols can be elaborated.


Assuntos
Infecções/complicações , Linfoma de Células T/complicações , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicações , Adolescente , Análise de Variância , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Distribuição de Qui-Quadrado , Criança , Pré-Escolar , Feminino , Humanos , Incidência , Lactente , Infecções/epidemiologia , Linfoma de Células T/tratamento farmacológico , Masculino , Neutropenia/complicações , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Estudos Retrospectivos , Fatores de Risco , Análise de Sobrevida
16.
Cancer Gene Ther ; 8(9): 662-8, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11593335

RESUMO

Gene therapy for malignant glioma with the herpes simplex virus thymidine kinase/ganciclovir (HSV-tk/GCV) system is already in the stage of clinical trials, but still needs major improvement to achieve greater clinical efficacy. The aim of this study was to determine whether combining HSV-tk/GCV gene therapy with temozolomide (TMZ), an alkylating drug clinically proven to be efficient in recurrent high-grade gliomas, would result in enhanced antitumor effect in malignant glioma in culture and in vivo. Human U87MG glioblastoma (GBM) cells with or without expression of HSV-tk were treated with different concentrations of GCV, TMZ, or both drugs. Cell viability was accessed by an automated microplate assay (MTT). The isobologram method and the combination index (CI) method of Chou-Talalay were used to measure the interactions between the two drugs when applied simultaneously. U87-tk and control U87 cells (5x10(6) each) were implanted in the flanks of nude mice, and animals were treated with GCV or TMZ or with both drugs. All tumors were measured and weighed at specified time points. IC(50) for GCV was 511 microM in control U87 cells and 14.3 microM in U87-tk cells, resulting in 35.7-fold increase of toxicity in the HSV-tk-expressing cells. TMZ had an IC(50) of 20.2 mM in control cells and 2.35 mM in U87-tk cells, resulting in 8.6-fold increase in sensitivity of the HSV-tk-expressing cells. TMZ and HSV-tk/GCV actions were synergistic (CI<1) in both control and U87-tk cells with higher synergism in U87-tk cells at high effect levels. Tumors expressing HSV-tk and treated with TMZ and GCV were significantly smaller than those treated by TMZ, but not by GCV. There was also a significant difference between the weight of HSV-tk expressing versus control tumors treated with TMZ, with GCV, or with both drugs. These data demonstrate synergism between HSV-tk/GCV and TMZ and higher sensitivity against TMZ in HSV-tk-expressing GBM cells. The potential importance for clinical studies combining both local tumor gene therapy and systemic chemotherapy should be explored further.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Ganciclovir/uso terapêutico , Terapia Genética/métodos , Glioma/terapia , Simplexvirus/enzimologia , Timidina Quinase/genética , Animais , Neoplasias Encefálicas/enzimologia , Divisão Celular/efeitos dos fármacos , Interações Medicamentosas , Sinergismo Farmacológico , Glioma/enzimologia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Simplexvirus/genética , Temozolomida , Células Tumorais Cultivadas
17.
Curr Gene Ther ; 1(4): 367-83, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12109063

RESUMO

Efficient virus and non-virus vector systems for gene transfer to tumor cells have been developed and tested in cell culture and in animal experiments. With some of the earliest and most comprehensively evaluated vectors, such as retroviruses, advanced clinical trials were performed in tumor patients. Malignant primary brain tumors (gliomas) have been chosen for the first clinical studies on novel gene therapy approaches because these tumors are non-metastatic and develop on the largely postmitotic background of normal glial and neuronal tissue. However, the human cancer gene therapy studies performed so far were not as successful as preclinical animal experiments. Furthermore, the clinical studies did not address major limiting factors for in vivo gene therapy, such as insufficient gene transfer rates to the tumor with the used local delivery modalities, and the resulting inability of a particular transgene-prodrug system to confer permanently eradicating cytotoxicity to the whole neoplasm. Critical evaluation of gene transfer and therapy studies has led to the conclusion that, even using identical vectors, the anatomical route of vector administration can dramatically affect both the efficiency of tumor transduction and its spatial distribution, as well as the extent of intratumoral and intracerebral transgene expression. This review concentrates on different physical methods for vector delivery to malignant primary brain tumors in experimental or clinical settings: stereotactic or direct intratumoral injection or convection-enhanced bulk-flow interstitial delivery; intrathecal and intraventricular injection; and intravascular infusion with or without modification of the blood-tumor-barrier. The advantages and drawbacks of the different modes and delivery routes of in vivo vector application, and the possibilities for tumor targeting by modifications of the native tropism of virus vectors or by using tissue-specific or inducible transgene expression are summarized.


Assuntos
Neoplasias Encefálicas/terapia , Marcação de Genes , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/genética , Humanos
18.
Gene Ther ; 7(21): 1853-8, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11110418

RESUMO

This study was conducted to investigate immunological components of somatic gene therapy for primary glioblastoma multiforme (GBM) in adults. It involved 13 patients treated by surgical resection of tumor with subsequent radiation therapy. Seven of them received additional herpes simplex virus thymidine kinase/ganciclovir (HSV-tk/GCV) gene therapy by direct intracerebral injection of retrovirus (RV) vector producing cells (VPC) during tumor surgery and subsequent systemic administration of GCV. Peripheral blood for FACS immunophenotyping, isolation of peripheral mononuclear cells (PMNC), and serum ELISA assays for IL-12 and soluble Fas ligand (sFasL) was collected daily during the first 4 post-operative weeks. Tumor specimens were obtained at primary or recurrent surgery and at autopsy. Tumors from gene therapy patients showed varying degrees of peritumoral necrosis around the former tumor resection cavity. Numbers of tumor-infiltrating lymphocytes found weeks after gene therapy were not significantly increased compared with primary tumors. Mitotic tumor cells were sparse close to the VPC injection sites, but abundant in brain areas somewhat distant from these sites. Serum ELISA revealed significantly increased sFasL and IL-12 levels in the gene therapy group compared with controls. Immunophenotyping of PMNC did not show a significant activation of T cells or NK cells during gene therapy. Interferon gamma secretion was evaluated by ELISPOT assays employing PMNC cocultivated with autologous tumor cells. It demonstrated an antitumor immune response in the gene therapy group, but not in the control group. These findings support the concept of in vivo induction of a systemic immune response by local intracerebral HSV-tk/GCV pharmacogene therapy for primary human GBM.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioblastoma/terapia , Simplexvirus/enzimologia , Timidina Quinase/genética , Transfecção/métodos , Idoso , Antivirais/uso terapêutico , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/imunologia , Estudos de Casos e Controles , Terapia Combinada , Proteína Ligante Fas , Feminino , Ganciclovir/uso terapêutico , Vetores Genéticos/administração & dosagem , Glioblastoma/sangue , Glioblastoma/imunologia , Humanos , Injeções Intralesionais , Interferon gama/imunologia , Interleucina-12/sangue , Masculino , Glicoproteínas de Membrana/sangue , Pessoa de Meia-Idade , Retroviridae/genética , Resultado do Tratamento
19.
J Neurooncol ; 48(3): 207-16, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11100818

RESUMO

Bcl-2 protein plays an important role in inhibiting apoptosis and protecting normal and neoplastic cells from toxicity. Bcl-2 overexpression in malignant tumors, on the other hand, may cause resistance against adjuvant treatment. Since there are subpopulations of patients with glioma that differ considerably in their treatment benefit, it is important to identify prognostic factors for outcome and to tailor adjuvant protocols in accordance with specific biological features of the respective tumor. The present study aimed at investigating the role of bcl-2 expression in higher-grade glioma (WHO grade III and IV). Bcl-2 expression was correlated with clinical and paraclinical parameters, and evaluated in univariate and multivariate statistical models. In addition, bcl-2-overexpressing human glioma cells in culture were used for modeling the in vivo findings and for investigating the importance of bcl-2 for tumor resistance against cytotoxic treatment. A group of 86 patients with higher-grade glioma were investigated. Anaplastic astrocytoma (AA; WHO G III, n = 29) showed bcl-2 expression in 48% of the cases, and immunohistochemical positivity was associated with a significantly shorter survival time (p = 0.0068). In glioblastoma patients (GBM; WHO G IV, n = 57), 51% of tumors were bcl-2 positive, but bcl-2 expression did not correlate significantly with survival (p = 0.39). In a Cox proportional hazards regression model, bcl-2 positivity was confirmed as a negative prognostic parameter in AA, but not in GBM. Bcl-2 overexpressing and control human glioma cell clones (T98MG line) were treated in culture with the cytotoxic drugs carmustine (BCNU), paclitaxel, vincristine, and doxorubicin. In addition, bcl-2-overexpressing and control cells were infected with a retrovirus carrying the herpes-simplex-virus thymidine kinase gene (HSV-tk), and then treated with ganciclovir (GCV). Bcl-2 overexpression significantly increased tumor cell resistance against all of the above cytotoxic drugs, and also against HSV-TK/GCV mediated gene therapy.


Assuntos
Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Glioma/patologia , Proteínas Nucleares , Proteínas Proto-Oncogênicas c-bcl-2/análise , Astrocitoma/mortalidade , Neoplasias Encefálicas/mortalidade , Receptores ErbB/análise , Glioblastoma/mortalidade , Glioma/mortalidade , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Proteínas de Neoplasias/análise , Prognóstico , Modelos de Riscos Proporcionais , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-mdm2 , Análise de Regressão , Estudos Retrospectivos , Taxa de Sobrevida , Fatores de Tempo , Proteína Supressora de Tumor p53/análise
20.
Cancer Gene Ther ; 7(5): 806-12, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10830728

RESUMO

To monitor therapeutic transgene expression, we developed fusion genes of enhanced green fluorescent protein (EGFP) with two different prodrug-activating enzyme genes: herpes simplex virus type 1 thymidine kinase (HSV-tk) and rabbit cytochrome P450 4B1 (cyp4b1). Expression of the resulting fusion proteins, TK-EGFP and 4B1-EGFP, rendered transduced human and rodent glioma cells sensitive to cytotoxic treatment with the corresponding prodrugs ganciclovir and 4-ipomeanol. Ganciclovir and 4-ipomeanol sensitivity was comparable with that achieved with the native HSV-TK and CYP4B1 proteins. As shown by fluorescence microscopy, TK-EGFP was expressed predominantly intranuclearly, whereas 4B1-EGFP was detectable in the cytoplasm, thereby displaying the orthotopic subcellular distribution of the corresponding native enzymes. The fluorescence intensity correlated well with the corresponding prodrug sensitivity, as shown by fluorescence-activated cell sorter analysis. EGFP expression was also used for the selection of stably HSV-tk-transduced cells by flow cytometric cell sorting. Resulting cell populations showed a homogeneity of fluorescence intensity similar to single-cell clones after antibiotic selection. In conclusion, tk-egfp and 4b1-egfp fusion genes are valuable tools for monitoring prodrug-activating gene therapy in living cells. EGFP fusion genes/proteins provide a simple and reproducible means for the detection, selection, and characterization of cells expressing enzyme genes for prodrug activation.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/genética , Terapia Genética/métodos , Herpesvirus Humano 1/enzimologia , Proteínas Luminescentes/metabolismo , Pró-Fármacos/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Timidina Quinase/genética , Animais , Western Blotting , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Glioma/terapia , Gliossarcoma/terapia , Proteínas de Fluorescência Verde , Humanos , Luz , Camundongos , Microscopia de Fluorescência , Modelos Genéticos , Coelhos , Ratos , Espalhamento de Radiação , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA