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
1.
Br J Haematol ; 102(2): 522-7, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9695968

RESUMO

SCID mice were inoculated intravenously with cells from the human HL60 myeloblastic leukaemia cell line and then treated with the 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase inhibitor, simvastatin, by subcutaneous continuous infusion. The effect of the drug was measured by subsequent colony formation of surviving HL60 cells in vitro and flow cytometry. The number of clonogenic HL60 cells was reduced in the bone marrow of mice that received simvastatin compared with control mice by 65% and 68% in two separate experiments. The number of clonogenic, normal, murine, bone marrow progenitor cells concomitantly exposed to simvastatin in vivo, was not affected in either experiment. Flow cytometric analysis of bone marrow and spleen cells confirmed these results by showing that simvastatin had reduced the percentage of human leukaemia cells in these tissues by 70% and 88% respectively. The data show that the reported selective effect of simvastatin against acute myeloid leukaemia cells in vitro, can be extended to this in vivo model. HL60 bears an N-ras mutation. In further in vitro studies, ketoconazole, an inhibitor of cholesterol biosynthesis post farnesyl pyrophosphate synthesis, had a similar effect to simvastatin on HL60 colony development. Furthermore, the clonogenicity of a population of N-ras mutated, primary acute myeloid leukaemia (AML) cells was no more sensitive to simvastatin than a population without the mutation. The data suggest that the inhibition of AML cell proliferation by simvastatin may be independent of the RAS signalling pathway.


Assuntos
Divisão Celular/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Leucemia Mieloide/tratamento farmacológico , Sinvastatina/uso terapêutico , Doença Aguda , Animais , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Células HL-60 , Humanos , Leucemia Mieloide/patologia , Camundongos , Camundongos SCID , Transplante de Neoplasias
2.
Br J Haematol ; 102(3): 718-21, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9722298

RESUMO

Non-obese diabetic NOD/SCID mice have been used to grow human leukaemia as a systemic disease. The animals were inoculated with leukaemic cells obtained from a 36-year-old male with early B-cell precursor acute lymphoblastic leukaemia and on day 15 were given the first of three weekly injections of 1 mg/kg vincristine or equimolar liposomal vincristine. The development of leukaemia in the mice was monitored by taking weekly blood samples and measuring the cell content by flow cytometry. The median time to 50% human cells in the peripheral blood of mice treated with free vincristine was 41 d from the start of treatment compared with 49 d for mice treated with liposomal vincristine (P < 0.01). The median day of death for mice treated with free vincristine was 47 d from the start of treatment and 57 d for mice receiving liposomal vincristine (P<0.01), thus providing a 21% increase in lifespan for animals treated with the liposomal preparation. There was slightly greater weight loss in mice treated with free vincristine than those given liposomal vincristine. Measurement of in vitro colony forming bone marrow progenitor cells in similarly treated, tumour-free mice, showed no difference in progenitor cell survival between mice that received either type of vincristine. We conclude that encapsulating vincristine in liposomes improves the therapeutic index of this drug measured in mice bearing human leukaemia. This may lead to use of the drug in conventional combination chemotherapy with greater safety or, in this setting, at higher dosage.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Vincristina/uso terapêutico , Adulto , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Peso Corporal/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Humanos , Lipossomos , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Análise de Sobrevida , Vincristina/administração & dosagem
3.
Blood ; 90(5): 2015-9, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9292537

RESUMO

Primary leukemic cells from patients with acute lymphoblastic leukemia (ALL) can be injected intravenously into mice with severe combined immunodeficiency (SCID) to create a model of human leukemia. Leukemic cells disseminate to murine tissues in a clinicopathologic pattern similar to that seen in humans. Thus far, reports of engraftment of lymphoid leukemia in SCID mice have mainly been from patients with B-cell lineage ALL, for which engraftment occurs more frequently with cells from high-risk patients. There are few data on the engraftment of T-cell lineage ALL in SCID mice. Leukemic cells from 19 patients (16 adult and three pediatric) with T-cell lineage ALL were injected into SCID mice, with overt engraftment of 12 cases (63%). Engraftment of leukemia in SCID mice was associated with earlier death due to leukemia of the patient donors (P < .01, log-rank test). The recently developed non-obese diabetic (NOD)/SCID mouse may expand the uses of the SCID model. Cells from the seven patients with T-cell lineage ALL that failed to cause leukemia in SCID mice were injected into NOD/SCID mice. Overt leukemia engraftment was observed in all seven cases. Thus, growth of human T-cell lineage ALL cells in SCID mice was associated with a high-risk patient group. However, this association was not observed when NOD/SCID mice were used, suggesting that this model would no longer predict patients likely to die early of leukemia, but may provide a more realistic system for studying the biology and treatment of the disease.


Assuntos
Leucemia Experimental , Leucemia de Células T/patologia , Doença Aguda , Adolescente , Adulto , Animais , Linhagem da Célula , Pré-Escolar , Modelos Animais de Doenças , Feminino , Humanos , Leucemia Experimental/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Linfócitos T/patologia
4.
Bone Marrow Transplant ; 20(4): 325-32, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9285548

RESUMO

We have studied granulocyte colony-stimulating factor (G-CSF)-induced mobilization of haemopoietic cells in severe combined immune-deficient (SCID) mice engrafted with human leukaemia. Three leukaemia cell lines were investigated: the HL60 myeloblastic cell line, a chronic myeloid leukaemia (CML) xenograft cell line and an acute myeloid leukaemia (AML) xenograft line. Engraftment was detected using immunofluorescent staining of class I human leukocyte antigens and flow cytometry. All the tumours grew as disseminated disease with engraftment of bone marrow preceding involvement of peripheral blood (PB). After treatment with G-CSF (250 microg/kg/day) for 5 days, mobilization of haemopoietic progenitor cells (HPCs) was observed in non-engrafted SCID mice (40-fold) and in mice engrafted with human leukaemia (20-fold). G-CSF stimulated increases in PB HPCs and total numbers of host nucleated cells in leukaemia-bearing mice but did not induce rises in numbers of circulating HL60 colony-forming cells. Similarly, in mice engrafted with human CML or AML, G-CSF did not increase the number of malignant cells in the PB. These results provided evidence that the migration of normal and malignant haemopoietic cells into the PB are controlled by different mechanisms, and that contamination of PBSC harvests with leukaemic cells in SCID-human chimaeric mice is not enhanced by G-CSF-stimulated mobilization.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucemia Experimental/sangue , Animais , Feminino , Células HL-60 , Humanos , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Transplante Heterólogo , Células Tumorais Cultivadas
5.
Leuk Lymphoma ; 24(5-6): 533-7, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9086443

RESUMO

Simvastatin is an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase and also selectively inhibits the growth of leukaemic progenitor cells. The antileukaemic action of simvastatin was compared in vitro with that of lovastatin and pravastatin, chemically related compounds which are also competitive inhibitors of HMG-CoA reductase. After 18 hours incubation with 2.5-20 microM of inhibitor, no effect was observed by any of the compounds on the subsequent clonogenic growth of normal bone marrow (BM) progenitor cells from 4 donors and BM cells from one patient in remission. However, simvastatin and lovastatin produced inhibition of acute myeloid leukaemia (AML) progenitor cell growth of between 25% and 100% in 5 populations tested (4 primary AMLs and the HL60 cell line). Pravastatin showed similar growth inhibitory effects to simvastatin and lovastatin in 2 out of 3 primary AMLs but was less active against one primary AML cell population and HL60 cells. These results indicate that, in addition to simvastatin, lovastatin and pravastatin are also selective inhibitors of leukaemic cell growth, however simvastatin was chosen for clinical trial in patients with leukaemia.


Assuntos
Antineoplásicos/farmacologia , Células da Medula Óssea , Inibidores Enzimáticos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases , Leucemia Mieloide/patologia , Lovastatina/análogos & derivados , Lovastatina/farmacologia , Pravastatina/farmacologia , Doença Aguda , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Células HL-60/efeitos dos fármacos , Células HL-60/enzimologia , Células HL-60/patologia , Células-Tronco Hematopoéticas/enzimologia , Humanos , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/enzimologia , Sinvastatina
6.
Blood ; 87(11): 4797-803, 1996 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8639851

RESUMO

Human interleukin-4 (huIL-4) has been shown to inhibit the growth in vitro of cells from patients with acute lymphoblastic leukemia (ALL). With the aim of determining whether this cytokine might be useful in the treatment of patients with ALL, the effects of huIL-4 on human B-cell precursor ALL engrafted in severe combined immunodeficient (SCID) mice were examined. The inhibition of [3H] thymidine uptake of primary ALL cells by huIL-4 was maintained following engraftment and passage of leukemia in SCID mice. Five of seven xenograft leukemias showed significant inhibition in vitro by huIL-4 at concentrations as low as 0.5 ng/mL; furthermore, huIL-4 counteracted the proliferative effects of IL-7. When used to treat two human leukemias engrafted in SCID mice, huIL-4 200 microgram/kg/d, as a continuous 14-day subcutaneous infusion, suppressed the appearance of circulating lymphoblasts and extended survival of mice by 39% and 108%, respectively, the first demonstration of IL-4 activity against human leukemia in vivo. The mean steady-state huIL-4 level in mouse plasma during the infusion was 1.46 ng/mL (SEM +/- 0.14 ng/mL), which was similar to concentrations found to be effective in vitro. ALL cells obtained from mice relapsing after huIL-4 treatment continued to show inhibition by the cytokine in vitro. These data suggest that IL-4 may be useful in the treatment of patients with ALL.


Assuntos
Fatores Imunológicos/uso terapêutico , Interleucina-4/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Animais , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Fatores Imunológicos/farmacologia , Interleucina-4/farmacologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Células-Tronco Neoplásicas/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Quimera por Radiação , Imunodeficiência Combinada Severa/complicações , Transplante Heterólogo
7.
J Investig Med ; 43(3): 269-74, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7614073

RESUMO

BACKGROUND: Autologous bone marrow transplantation (ABMT) is frequently used in the treatment of malignant disease but carries the risk of reintroducing tumor cells into the patient. Methods are required for removing malignant cells from harvested bone marrow (BM) without impairing hematopoietic reconstitution. We have shown that simvastatin is toxic to leukemic progenitor cells at a concentration that conserves normal BM progenitors and may be of use clinically as a novel BM purging agent. METHODS: A two-stage culture system was used to compare the effects of simvastatin on both normal BM progenitor and primary acute myeloid leukemia (AML) cells. AML cells and normal BM mononuclear cells were incubated for 18 hours in suspension culture with 10 micrograms per mL simvastatin and the numbers of surviving clonogenic progenitor cells assayed in semisolid agar culture. RESULTS: Following simvastatin treatment of 18 AML cell populations, the mean surviving fraction of progenitor cells was 21.3 +/- 4.8% ( +/- standard error of the mean [SEM]). In contrast, the mean survival of normal BM progenitors from 16 donors was 89.6 +/- 8.6% ( +/- SEM). Samples were taken from 6 AML patients before treatment and after remission of disease had been induced by chemotherapy. In 5 of these cases the AML sample was significantly more sensitive to simvastatin than the remission sample, 4 of the 5 showed > 80% difference in progenitor cell survival. CONCLUSIONS: AML progenitor cells are sensitive to a short-term exposure to simvastatin that spares normal BM hematopoietic progenitor cells. We conclude that simvastatin may be an effective in vitro purging agent in ABMT for AML.


Assuntos
Purging da Medula Óssea/métodos , Transplante de Medula Óssea/métodos , Medula Óssea/efeitos dos fármacos , Leucemia Mieloide/patologia , Lovastatina/análogos & derivados , Doença Aguda , Morte Celular , Células Cultivadas , Humanos , Lovastatina/farmacologia , Indução de Remissão , Sinvastatina , Transplante Autólogo
8.
Blood ; 85(4): 893-901, 1995 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-7849311

RESUMO

Recurrent abnormalities of the short arm of chromosome 9, including translocations and interstitial deletions, have been reported in both leukemia and lymphoma. The pathologic consequences of these abnormalities remain unknown. The cyclin-dependent kinase 4 inhibitor (CDKN2) gene, which maps to 9p21, has been implicated by the finding of a high frequency of biallelic deletions in leukemic cell lines. We have determined the incidence of structural abnormalities affecting CDKN2 by DNA blot in a panel of 231 cases of leukemia and lymphoma and 66 cell lines derived from patients with lymphoid malignancies with defined cytogenetic abnormalities. Structural alterations of CDKN2 were seen in 20 (8.3%) of all fresh cases and 10 (15.1%) of all cell lines. Biallelic CDKN2 deletions were seen in 11 of 53 (21%) cases of B-cell precursor acute lymphoblastic leukemia (BCP-ALL). There was no association with any particular cytogenetic abnormality. Biallelic deletions were also found in high-grade and transformed non-Hodgkin's lymphoma (NHL) of both B- and T-cell lineages. In two cases of transformed NHL, analysis of sequential samples showed loss of CDKN2 with transformation. Neither deletions nor rearrangements of the CDKN2 gene were seen in any of the 119 leukemias of mature B or T cells analyzed. Biallelic deletions of CDKN2 were observed in 6 of 13 NHL cell lines. Three of the 6 cases had undergone transformation from low- to high-grade disease: in 2 of these cases it was possible to show that the CDKN2 deletions were present in fresh material from the patient and were therefore not an artifact of in vitro culture. Rearrangements of CDKN2 were seen in 2 cases (4%) of BCP-ALL, in 1 case of B-NHL, and in 1 Burkitt's lymphoma cell line and suggest the presence of a "hot spot" for recombination in the vicinity of the CDKN2 gene. These data indicate that the loss of CDKN2 expression may be involved in the pathogenesis of a subset of BCP-ALL, some high-grade NHL, and in the transformation of NHL from low- to high-grade disease. CDKN2 deletions and rearrangements occurred in the absence of detectable cytogenetic changes of chromosome 9p in 25 of 30 (83%) cases. Finally, of 10 cases of BCP-ALL that produced overt, transplantable leukemia in mice with severe combined immunodeficiency (SCID), seven showed biallelic CDKN2 deletions. In contrast, none of 11 cases that failed to engraft showed biallelic CDKN2 deletions. BCP-ALL cases that lack CDKN2 expression may have a particular propensity to grow in SCID mice.


Assuntos
Proteínas de Transporte/genética , Aberrações Cromossômicas , Transtornos Cromossômicos , Cromossomos Humanos Par 9 , Deleção de Genes , Rearranjo Gênico , Leucemia/genética , Linfoma/genética , Inibidores de Proteínas Quinases , Adolescente , Adulto , Idoso , Animais , Proteínas de Transporte/biossíntese , Linhagem Celular , Criança , Pré-Escolar , Mapeamento Cromossômico , Cromossomos Humanos Par 8 , Inibidor p16 de Quinase Dependente de Ciclina , Éxons , Feminino , Humanos , Leucemia/enzimologia , Linfoma/enzimologia , Masculino , Camundongos , Camundongos SCID , Mapeamento por Restrição , Translocação Genética , Transplante Heterólogo , Células Tumorais Cultivadas
9.
Leukemia ; 8(11): 2023-9, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7967748

RESUMO

Primary human acute myeloid leukaemic (AML) cells from bone marrow (BM) and peripheral (PB), the human myeloblastic leukaemia cell line (HL60) and normal human BM mononuclear cells were cultured in serum-free medium. The survival of progenitor cells from normal BM, HL60 and AML cell populations was reduced over a range of concentrations of simvastatin. This dose response relationship was more pronounced in HL60 and AML cell cultures, indicating greater sensitivity of AML progenitor cells compared with normal BM progenitors. Short-term exposure (18 h) to a range of concentrations of simvastatin showed the same differential response between leukaemic and normal BM cells in terms of clonogenicity. At a concentration of 10 micrograms/ml progenitor cell survival remained above 65% for normal BM while at this concentration leukaemia progenitor cell survival fell below 25% of the untreated values. The differential effect of simvastatin on normal and leukaemic progenitor cells may have value in the clinical management of AML. The possible use of simvastatin, or related drugs, as adjuvants to conventional chemotherapy including in vitro BM purging, merits consideration.


Assuntos
Células da Medula Óssea , Colesterol/farmacologia , Hematopoese/efeitos dos fármacos , Leucemia Mieloide/patologia , Lovastatina/análogos & derivados , Doença Aguda , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Clonais , Células-Tronco Hematopoéticas/citologia , Humanos , Lovastatina/farmacologia , Células-Tronco Neoplásicas/citologia , Sinvastatina , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos
10.
Leukemia ; 8(2): 274-80, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8309251

RESUMO

Primary human acute myeloid leukaemic (AML) cells from bone marrow (BM) and peripheral blood (PB), the human myeloblastic leukaemia cell line (HL60) and normal human BM mononuclear cells were cultured in serum-free medium. The survival of progenitor cells from normal BM, HL60 and AML cell populations was reduced over a range of concentrations of lovastatin. This dose response relationship was more pronounced in HL60 and AML cell cultures, indicating greater sensitivity of AML progenitor cells compared with normal BM progenitors. Short-term exposure (18 h) to a range of concentrations of lovastatin showed the same differential response between leukaemic and normal BM cells in terms of clonogenicity. At a concentration of 10 micrograms/ml progenitor cell survival remained above 65% for normal BM while at this concentration leukaemia progenitor cell survival fell below 25% of the untreated values. The differential effect of lovastatin on normal and leukaemic progenitor cells may have value in the clinical management of AML. The possible use of lovastatin, or related drugs, as adjuvants to conventional chemotherapy including in vitro BM purging, merits consideration.


Assuntos
Leucemia Mieloide Aguda/patologia , Lovastatina/farmacologia , Adulto , Medula Óssea/patologia , Células da Medula Óssea , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colesterol/farmacologia , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
11.
Exp Hematol ; 21(6): 826-8, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8500581

RESUMO

We have developed an in vivo model of human chronic myeloid leukemia (CML). A peripheral blood (PB) sample of Philadelphia (Ph) chromosome-positive CML cells in lymphoid blast crisis was transplanted intravenously (IV) into sublethally irradiated severe combined immunodeficient (SCID) mice, and this resulted in engraftment with systemic proliferation. Growth of leukemia was monitored by PB cell morphology and by flow cytometric analysis of murine PB cells labelled with an anti-human leukocyte antigen (HLA) monoclonal antibody. Human cells were first detected in the PB at 4 weeks and comprised a mean of 57% of the total nucleated cells in the PB of these mice by 15 weeks. The Ph chromosome was retained and the population has been successfully passaged. BCR/ABL fusion gene expression was detected in a subsequent passage. Experiments are underway to use this in vivo model to assess the antileukemic activity of BCR/ABL antisense oligonucleotides.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Proteínas Tirosina Quinases , Animais , Anticorpos Monoclonais/análise , Anticorpos Monoclonais/imunologia , Linfócitos B/patologia , Clonagem Molecular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Fluorescência , Regulação Neoplásica da Expressão Gênica/genética , Antígenos HLA/imunologia , Homozigoto , Humanos , Cariotipagem , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Camundongos SCID , Oligonucleotídeos Antissenso/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-bcr , Linfócitos T/patologia , Células Tumorais Cultivadas
12.
Leukemia ; 3(9): 637-42, 1989 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2668655

RESUMO

Normal human bone marrow was grown as xenografts in mice immune-suppressed by thymectomy and total body irradiation. Mononuclear cell fractions isolated from marrow harvests from 17 donors all gave rise to subcutaneous nodules which grew to a variable maximum size and then regressed. Human granulocyte/macrophage progenitors (CFU-GM) were recovered from xenografts up to 20 days postimplantation. Xenograft growth, measured by maximum nodule volume, area under the growth curve, and rate of regression, did not correlate with the speed of neutrophil or platelet recovery in bone marrow transplant patients infused with the same marrow. Assay of numbers of stromal fibroblastoid colony forming cells (CFU-F) in donor marrow was also not predictive of subsequent hemopoietic recovery in recipients. Treatment of host animals with daily intraperitoneal injections of 100 micrograms/kg human recombinant granulocyte/macrophage colony stimulating factor produced a more rapid growth of subcutaneous nodules. This technique may therefore be of use in determining the in vivo efficacy of human hemopoietic regulatory factors.


Assuntos
Transplante de Medula Óssea , Fatores Estimuladores de Colônias/farmacologia , Substâncias de Crescimento/farmacologia , Hematopoese , Animais , Ensaio de Unidades Formadoras de Colônias , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Humanos , Terapia de Imunossupressão , Camundongos , Camundongos Endogâmicos CBA , Proteínas Recombinantes , Timectomia , Transplante Heterólogo
14.
Br J Cancer ; 53(5): 607-14, 1986 May.
Artigo em Inglês | MEDLINE | ID: mdl-3718818

RESUMO

Verapamil had previously been shown to increase cellular melphalan uptake and cytotoxicity in fibrosarcomas, and increased the area under the blood concentration versus time curve (AUC) for melphalan in CBA mice. Verapamil (10 mg kg-1 i.p.) had no effect on the fractional distribution of cardiac output (FDCO), measured with 86Rb-rubidium chloride, to subcutaneous fibrosarcomas. 14C-Melphalan uptake by FS13 fibrosarcomas was increased 60 min after verapamil (10 mg kg-1 i.p.), but not after lower doses which did not affect the AUC. Flunarizine (5 mg kg-1 i.p.) also had no effect on FDCO to FS13 fibrosarcomas, and tended to increase 14C-melphalan content of blood and the fibrosarcomas and to promote growth delay by melphalan. Alcohol increased FDCO to FS13 fibrosarcomas, maximally at a 1:20 dilution in saline, but had no effect on 14C-melphalan uptake or growth delay. Thus, melphalan cytotoxicity correlated with tumour melphalan uptake, and both followed changes in the AUC for melphalan but not changes in FDCO. In these murine fibrosarcomas melphalan uptake and cytotoxicity were not limited by blood flow. In subcutaneous human melanoma HX46 xenografts, verapamil had no effect on the FDCO, nor on 14C-melphalan uptake, and did not affect blood 14C-melphalan levels, suggesting absence of effects on the AUC and on cellular uptake. Alcohol did not increase the FDCO to HX46 xenografts, providing evidence for a different vascular supply.


Assuntos
Etanol/farmacologia , Fibrossarcoma/irrigação sanguínea , Melanoma/irrigação sanguínea , Melfalan/metabolismo , Verapamil/farmacologia , Animais , Cinarizina/análogos & derivados , Cinarizina/farmacologia , Sinergismo Farmacológico , Feminino , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/metabolismo , Flunarizina , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Melfalan/uso terapêutico , Camundongos , Camundongos Endogâmicos CBA , Fluxo Sanguíneo Regional/efeitos dos fármacos
15.
Br J Cancer ; 52(6): 813-22, 1985 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-4074636

RESUMO

Growth delay by melphalan of two fibrosarcomas in CBA mice was prolonged by intraperitoneal (i.p.) verapamil, 10 mg kg-1. Verapamil also increased the area under the blood concentration time curve and the gastrointestinal toxicity of melphalan. Verapamil promoted melphalan cytotoxicity to murine bone marrow both in vivo, by CFU-S assay, and in vitro, by CFU-GM assay. In 1 microgram ml-1 [14C]-melphalan, verapamil (10 micrograms ml-1) increased by 1.5 times the [14C]-melphalan accumulation by murine bone marrow, reversibly and independently of external calcium. Efflux of [14C]-melphalan from murine bone marrow was retarded by verapamil. Verapamil increased [14C]-melphalan uptake by disaggregated fibrosarcoma cells but had no effect on melphalan accumulation and cytotoxicity in human bone marrow. Although verapamil affected melphalan pharmacokinetics, enhancement of cellular melphalan uptake by verapamil in murine fibrosarcoma and bone marrow appeared to account for much of the increase in melphalan cytotoxicity. The lack of potentiation of melphalan by verapamil in human marrow suggests differences in melphalan transport or in verapamil membrane interactions in mouse and man.


Assuntos
Medula Óssea/efeitos dos fármacos , Fibrossarcoma/tratamento farmacológico , Melfalan/uso terapêutico , Verapamil/uso terapêutico , Animais , Medula Óssea/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Sinergismo Farmacológico , Feminino , Fibrossarcoma/sangue , Fibrossarcoma/metabolismo , Humanos , Hidrólise , Técnicas In Vitro , Jejuno/efeitos dos fármacos , Cinética , Masculino , Melfalan/sangue , Melfalan/metabolismo , Camundongos , Camundongos Endogâmicos CBA , Verapamil/sangue , Verapamil/metabolismo
16.
Br J Cancer ; 52(5): 733-7, 1985 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3877517

RESUMO

The effect of epidermal growth factor (hEGF) on intestinal epithelial damage by melphalan was explored in CBA mice. Human EGF was administered in doses of 100 micrograms kg-1 or 1000 micrograms kg-1 using a variety of schedules. Mucosal damage was assessed 4, 8 and 13 days later, by [14C]-xylose uptake and by microcolony survival of jejunum, ileum and colon. The only regimen to show enhanced jejunal crypt survival was administration of hEGF, 100 micrograms kg-1, i.p., 8 hourly, beginning 24 h before melphalan treatment. Oral administration of hEGF had no effect on melphalan induced damage nor on subsequent recovery of intestinal mucosa. Activity of hEGF in mice was confirmed by demonstration of precocious eyelid opening in newborn mice. No consistent protective or restorative effect of hEGF on melphalan-induced intestinal epithelial damage could be demonstrated with the doses and schedules used.


Assuntos
Fator de Crescimento Epidérmico/uso terapêutico , Enteropatias/prevenção & controle , Melfalan/toxicidade , Regeneração/efeitos dos fármacos , Animais , Esquema de Medicação , Fator de Crescimento Epidérmico/administração & dosagem , Feminino , Enteropatias/induzido quimicamente , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos CBA
17.
Leuk Res ; 9(12): 1511-8, 1985.
Artigo em Inglês | MEDLINE | ID: mdl-3878437

RESUMO

Human AML cells from the blood of a series of patients have been implanted subcutaneously into mice immune-suppressed by thymectomy and total-body irradiation. Solid tumours resulted from 18 out of 19 samples and their growth was compared with the proliferation of AML cells in culture. In 17 cases tumours grew to a maximum size and then spontaneously regressed. Cells from one patient produced tumours which did not regress and could be retransplanted into freshly immune-suppressed mice. Cells from a human promyelocytic cell line (HL60) also produced nonregressing and retransplantantable tumours. Normal human mononuclear bone marrow cells implanted s.c. produced a growth pattern similar to that of the majority of AML cells. A second inoculum of AML cells into animals with regressing tumours also produced tumours and thus regression cannot be accounted for on the basis of returning immunity. AML cells placed into short-term suspension culture invariably matured to monocyte/macrophage type cells and/or granulocytic cells as identified by cytochemical staining. However, no correlation was observed between proliferation or maturation of cells in culture, and tumour growth in vivo. Cells derived from disaggregated AML tumours also showed evidence of myeloid differentiation suggesting that tumour regression is due to maturation of leukaemic cells.


Assuntos
Tolerância Imunológica , Leucemia Mieloide Aguda/patologia , Transplante Heterólogo , Animais , Diferenciação Celular , Células Cultivadas , Fatores Estimuladores de Colônias/farmacologia , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos CBA , Transplante de Neoplasias
18.
Br J Cancer ; 49(1): 43-7, 1984 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-6318789

RESUMO

Very high dose cyclophosphamide (Cy) (500-600 mg kg-1) given by single bolus i.p. injection in mice caused acute deaths in all animals within 48 h of treatment (0/10 survivors). These acute deaths were abolished or very significantly reduced if Cy was administered in divided dosage over 8 h (10/10 survivors) or 12 h (14/15 survivors). The effect was maintained at doses of up to 600 mg kg-1 administered in divided dosage over 24 h (15/15 survivors). In 2 human small cell carcinoma xenografts anti-tumour efficacy was not diminished by divided dosage. In both xenografts tumour growth delay was enhanced, although not significantly so, when treated with divided dosage compared with single dose, and in one of the xenografts 3 complete remissions were achieved with divided dosage compared with none after single dosage. It is postulated that the underlying mechanism concerns diminished cardiotoxicity. These results may have significance in clinical studies investigating very high dose Cy.


Assuntos
Ciclofosfamida/administração & dosagem , Animais , Carcinoma de Células Pequenas/tratamento farmacológico , Ciclofosfamida/toxicidade , Relação Dose-Resposta a Droga , Esquema de Medicação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Transplante de Neoplasias , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...