Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Contemp Clin Trials ; 29(2): 157-64, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17707140

RESUMO

Evaluation of time to event outcomes usually is examined by the Kaplan-Meier method and Cox proportional hazards models. We developed a modified statistical model based on histologic grade and other variables to describe the time-dependent outcome for autologous stem cell transplant (autotransplant) performed for non-Hodgkin's lymphoma (NHL) based on histologic grade and other variables. One hundred and fourteen relapsed or refractory NHL patients were treated using BCNU 600 mg/m2, etoposide 2400 mg/m2, and cisplatin 200 mg/m2 IV followed by autotransplant. Median age was 53.5 (range: 25-70) years, 78 patients had aggressive NHL and 36 indolent NHL. Seventy-five patients received involved-field radiotherapy just prior to transplant. At a median follow-up of 33 (range: 3 to 118) months, the estimated 5-year Kaplan-Meier probabilities of overall survival and disease-free survival were 61% and 51%, respectively. Cox proportional hazards model analysis showed that proportionality did not hold for lymphoma grade, indicating that the relationship between the grade and disease-free survival differed over time. By piece-wise Cox model, the relative risk for experiencing relapse or death after 1 year in patients with indolent compared with patients with aggressive NHL was 2.81 (p=0.019) with 95% confidence interval (1.19, 6.65). The time-dependent effect of lymphoma grade on disease-free survival suggests the need for early (within first year) incorporation of novel therapeutic approaches in management of patients with indolent NHL undergoing autotransplant.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfoma não Hodgkin/mortalidade , Linfoma não Hodgkin/terapia , Transplante de Células-Tronco , Adulto , Idoso , Carmustina/administração & dosagem , Cisplatino/administração & dosagem , Intervalo Livre de Doença , Etoposídeo/administração & dosagem , Feminino , Humanos , Linfoma não Hodgkin/patologia , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Modelos de Riscos Proporcionais , Tempo , Transplante Autólogo , Resultado do Tratamento
2.
Bone Marrow Transplant ; 38(3): 189-96, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16850032

RESUMO

We examined pre-mobilization blood CD34+ cell count to predict ability to mobilize adequate peripheral blood progenitor cells (PBPC) in 106 cancer patients and 36 allogeneic donors. Mean pre-mobilization therapy blood CD34+ cell count was 3.1 cells x 10(6)/l (s.d. = 3.9, r = 0.3-37) and mean CD34+ cells collected were 5.3 x 10(6) cells/kg/leukapheresis procedure (s.d. = 7.0, r = 0.03-53). Yields correlated with pre-mobilization CD34+ cells x 10(6)/l (r = 0.37, P-value < 0.0001); correlation was stronger in allogeneic donors (r = 0.56, P-value = 0.0004) and males (r = 0.46, P-value < 0.0001). Based on classification and regression tree multivariate analysis, the predictive value of pre-mobilization blood CD34+ cell count was confounded by other variables, including age, gender, mobilization regimen and malignancy type. We generated an algorithm to predict a minimum PBPC yield of 1 x 10(6) CD34+ cells/kg/leukapheresis procedure after mobilization. A threshold pre-mobilization blood CD34+ cell count of 2.65 cells x 10(6)/l was the most important decision point in predicting successful mobilization. Only 2% of subjects with pre-mobilization blood CD34+ cell counts > 2.65 cells x 10(6)/l did not achieve the minimum per apheresis, whereas 24% with pre-mobilization values below threshold had inadequate mobilization. Prospectively identifying individuals at risk for mobilization failure would allow for improved treatment planning, resource utilization and time saving.


Assuntos
Algoritmos , Mobilização de Células-Tronco Hematopoéticas/métodos , Leucaférese/métodos , Neoplasias/sangue , Adolescente , Adulto , Idoso , Antígenos CD34/análise , Criança , Pré-Escolar , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Humanos , Contagem de Leucócitos/métodos , Masculino , Pessoa de Meia-Idade , Neoplasias/terapia , Contagem de Plaquetas/métodos , Valor Preditivo dos Testes , Estudos Retrospectivos , Resultado do Tratamento
3.
Cancer Res ; 57(22): 5093-9, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9371508

RESUMO

A retroviral gene therapy approach was developed to protect early hematopoietic progenitors from 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), a stem cell toxin, and O6-benzylguanine (BG), an inhibitor of a key BCNU resistance protein, O6-alkylguanine DNA alkyltransferase (AGT). The retroviral vector MFG was used to transfer the G156A MGMT (deltaMGMT) cDNA, encoding a mutant AGT that is resistant to inhibition by BG, into murine bone marrow-derived hematopoietic progenitors. Following transplantation into lethally irradiated mice, the transduced cells were subjected to in vivo BG and BCNU treatment to examine the ability to enrich for transduced cells expressing deltaAGT. Transplantation of deltaMGMT-transduced cells resulted in deltaAGT expression in 30% of bone marrow nucleated cells 13 weeks after transplantation. After one cycle of BG and BCNU, deltaAGT expression was observed in 60% of bone marrow cells, and the percentage of colony-forming units (culture; CFU-C) containing proviral sequence increased from 67 to 100%. CFU-C obtained from BG and BCNU-treated deltaMGMT animals up to 23 weeks after transplantation were more resistant to combination BG and BCNU than CFU-C from mice transplanted with lacZ-transduced cells and treated with BG and BCNU or from mice transplanted with deltaMGMT-transduced cells and left untreated. The degree of drug resistance in deltaMGMT-transduced hematopoietic progenitors to BG and BCNU was much greater than we observed previously with wild-type MGMT gene transfer and treatment with BCNU alone. Furthermore, whereas 21 of 22 mice transplanted with deltaMGMT-transduced cells survived in vivo BG and BCNU administration, only 3 of 13 mice transplanted with lacZ-transduced progenitors survived similar drug treatment. Thus, deltaMGMT-transduced murine bone marrow cells selectively survive in vivo BG and BCNU exposure, resulting in prolonged enrichment for the transduced cells and protection from mortality induced by this drug combination.


Assuntos
Antineoplásicos/farmacologia , Carmustina/farmacologia , Guanina/análogos & derivados , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/genética , Transfecção , Animais , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Vetores Genéticos , Guanina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Retroviridae/genética
4.
Oncogene ; 15(17): 2127-32, 1997 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-9366529

RESUMO

We evaluated induction of lymphomas by the methylating carcinogen, N-methylnitrosourea [MNU], in transgenic mice expressing both LMO1 and the DNA repair gene, MGMT, in the thymus. The goal was to determine whether environmental mutagens shorten the latency or increase the incidence of LMO1 + lymphomas and whether mice transgenic for both LMO1 and MGMT, and thereby able to repair O6-methylguanine DNA adducts induced by MNU, would be protected. Mice heterozygous for LMO1 or MGMT were crossed and offspring treated with MNU at 6 weeks of age. MNU induced lymphoma incidence was highest in the LMO1 mice, 91% and lowest in the hMGMT + mice, 15%. MNU induced K-ras mutations in codon 12 in non-MGMT transgenics resulted in a shorter latency of tumors and accounting for half of the early lymphomas in LMO1 mice. The effect of MNU was abrogated in the LMO1/hMGMT transgenic mice, indicating the ability of MGMT expression to block the carcinogenic effect of MNU even in cancer prone mice. Thus, methylating agents potentiate lymphomagenesis of LMO1, in part through activation of K-ras and the MAPK pathway, a process which appear to synergize with LMO1 mediated transcription activation. O6-alkylguanine DNA-alkyltransferase mediated DNA repair effectively blocks chemical carcinogenesis in mice carrying the LMO1 oncogene.


Assuntos
Proteínas de Ligação a DNA/genética , Genes ras/fisiologia , Linfoma/prevenção & controle , Metaloproteínas/genética , Proteínas de Neoplasias/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Proteínas Oncogênicas , Neoplasias do Timo/prevenção & controle , Animais , Carcinógenos , Códon/genética , Proteínas com Domínio LIM , Linfoma/induzido quimicamente , Linfoma/enzimologia , Metilnitrosoureia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/prevenção & controle , Proteínas Nucleares , O(6)-Metilguanina-DNA Metiltransferase/genética , Mutação Puntual , Neoplasias do Timo/induzido quimicamente , Neoplasias do Timo/enzimologia , Fatores de Transcrição
5.
J Clin Oncol ; 18(2): 307-16, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10637244

RESUMO

PURPOSE: Multipotential mesenchymal stem cells (MSCs) are found in human bone marrow and are shown to secrete hematopoietic cytokines and support hematopoietic progenitors in vitro. We hypothesized that infusion of autologous MSCs after myeloablative therapy would facilitate engraftment by hematopoietic stem cells, and we investigated the feasibility, safety, and hematopoietic effects of culture-expanded MSCs in breast cancer patients receiving autologous peripheral-blood progenitor-cell (PBPC) infusion. PATIENTS AND METHODS: We developed an efficient method of isolating and culture-expanding a homogenous population of MSCs from a small marrow-aspirate sample obtained from 32 breast cancer patients. Twenty-eight patients were given high-dose chemotherapy and autologous PBPCs plus culture-expanded MSC infusion and daily granulocyte colony-stimulating factor. RESULTS: Human MSCs were successfully isolated from a mean +/- SD of 23.4 +/- 5.9 mL of bone marrow aspirate from all patients. Expansion cultures generated greater than 1 x 10(6) MSCs/kg for all patients over 20 to 50 days with a mean potential of 5.6 to 36.3 x 10(6) MSCs/kg after two to six passages, respectively. Twenty-eight patients were infused with 1 to 2.2 x 10(6) expanded autologous MSCs/kg intravenously over 15 minutes. There were no toxicities related to the infusion of MSCs. Clonogenic MSCs were detected in venous blood up to 1 hour after infusion in 13 of 21 patients (62%). Median time to achieve a neutrophil count greater than 500/microL and platelet count >/= 20,000/microL untransfused was 8 days (range, 6 to 11 days) and 8.5 days (range, 4 to 19 days), respectively. CONCLUSION: This report is the first describing infusion of autologous MSCs with therapeutic intent. We found that autologous MSC infusion at the time of PBPC transplantation is feasible and safe. The observed rapid hematopoietic recovery suggests that MSC infusion after myeloablative therapy may have a positive impact on hematopoiesis and should be tested in randomized trials.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Transplante de Células-Tronco Hematopoéticas/métodos , Neutropenia/terapia , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Células da Medula Óssea , Técnicas de Cultura de Células/métodos , Relação Dose-Resposta a Droga , Feminino , Sobrevivência de Enxerto , Humanos , Mesoderma/citologia , Pessoa de Meia-Idade , Transplante Autólogo
6.
J Clin Oncol ; 18(9): 1824-30, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10784622

RESUMO

PURPOSE: Patient response to hematopoietic progenitor-cell mobilizing regimens seems to vary considerably, making comparison between regimens difficult. To eliminate this inter-patient variability, we designed a cross-over trial and prospectively compared the number of progenitors mobilized into blood after granulocyte-macrophage colony-stimulating factor (GM-CSF) days 1 to 12 plus granulocyte colony-stimulating factor (G-CSF) days 7 to 12 (regimen G) with the number of progenitors after cyclophosphamide plus G-CSF days 3 to 14 (regimen C) in the same patient. PATIENTS AND METHODS: Twenty-nine patients were randomized to receive either regimen G or C first (G1 and C1, respectively) and underwent two leukaphereses. After a washout period, patients were then crossed over to the alternate regimen (C2 and G2, respectively) and underwent two additional leukaphereses. The hematopoietic progenitor-cell content of each collection was determined. In addition, toxicity and charges were tracked. RESULTS: Regimen C (n = 50) resulted in mobilization of more CD34(+) cells (2.7-fold/kg/apheresis), erythroid burst-forming units (1.8-fold/kg/apheresis), and colony-forming units-granulocyte-macrophage (2.2-fold/kg/apheresis) compared with regimen G given to the same patients (n = 46; paired t test, P<.01 for all comparisons). Compared with regimen G, regimen C resulted in better mobilization, whether it was given first (P =.025) or second (P =.02). The ability to achieve a target collection of > or =2x10(6) CD34(+) cells/kg using two leukaphereses was 50% after G1 and 90% after C1. Three of the seven patients in whom mobilization was poor after G1 had > or =2x10(6) CD34(+) cells/kg with two leukaphereses after C2. In contrast, when regimen G was given second (G2), seven out of 10 patients failed to achieve the target CD34(+) cell dose despite adequate collections after C1. Thirty percent of the patients (nine of 29) given regimen C were admitted to the hospital because of neutropenic fever for a median duration of 4 days (range, 2 to 10 days). The higher cost of regimen C was balanced by higher CD34(+) cell yield, resulting in equivalent charges based on cost per CD34(+) cell collected. CONCLUSION: We report the first clinical trial that used a cross-over design showing that high-dose cyclophosphamide plus G-CSF results in mobilization of more progenitors then GM-CSF plus G-CSF when tested in the same patient regardless of sequence of administration, although the regimen is associated with greater morbidity. Patients who fail to achieve adequate mobilization after regimen G can be treated with regimen C as an effective salvage regimen, whereas patients who fail regimen C are unlikely to benefit from subsequent treatment with regimen G. The cross-over design allowed detection of significant differences between regimens in a small cohort of patients and should be considered in design of future comparisons of mobilization regimens.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Ciclofosfamida/farmacologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas , Adolescente , Adulto , Idoso , Antígenos CD34/análise , Estudos Cross-Over , Relação Dose-Resposta a Droga , Feminino , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/terapia
7.
Clin Cancer Res ; 4(2): 475-81, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9516939

RESUMO

Alkyltransferase (AGT) repairs alkylation at O6-guanine in DNA and is a major determinant of susceptibility to alkylating chemotherapeutic agents and carcinogens. Using a newly developed flow cytometry assay with the monoclonal anti-AGT antibody, mT3.1, we compared AGT expression in single-cell suspensions with standard biochemical and Western blot assays to validate the fluorescence-activated cell sorting (FACS) method and develop potential applications. From Chinese hamster ovary cells (CHO) transfected with human O6-methylguanine-DNA methyl-transferase cDNA, 6 CHO-O6-methylguanine-DNA methyl-transferase clones were isolated that expressed 0.3 to 64 fmol/microgram DNA (by biochemical assay) of human AGT. FACS yielded a linear relationship between mean fluorescence intensity and both AGT activity by biochemical assay and AGT protein by Western blot. Using this standard curve, FACS-analyzed AGT protein content in human peripheral blood mononuclear cells (PBMCs) from normal donors ranged from 6.1 to 12.8 fmol/microgram DNA, similar to those obtained by biochemical assay and Western blot. This suggests that the level of immunoreactive protein appears to be an accurate predictor of AGT activity in the steady state. FACS-AGT in PBMCs from normal donors had a low index of heterogeneity within the sample. In contrast, by FACS-AGT analysis of human bone marrow samples and granulocyte-colony-stimulating factor-mobilized PBMCs, AGT was lower and had an 8-fold higher index of heterogeneity than observed in PBMCs from normal donors. After treatment with O6-benzylguanine (O6-bG), Western and FACS-AGT detected significant levels of AGT protein for up to 24 h, whereas biochemical assay showed AGT activity less than 5% of the basal level. Because only the biochemical assay accurately measures net AGT activity, the AGT-FACS assay will not be useful in clinical trials to assess the efficacy of O6-bG or other AGT inhibitors. Thus, AGT-FACS can rapidly assess the heterogeneity of steady-state AGT in single-cell suspensions and may be useful for assay in lymphocytes, bone marrow cells, leukemic myeloma plasma cells, or cells transfected with the AGT gene; Western blot analysis is better for small samples such as tumor biopsies, whereas biochemical assay is best able to measure enzyme activity and its inactivation by O6-bG or other agents.


Assuntos
Células da Medula Óssea/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/sangue , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Animais , Anticorpos Monoclonais , Western Blotting , Células CHO/enzimologia , Permeabilidade da Membrana Celular , Separação Celular/métodos , Cricetinae , Ativação Enzimática , Citometria de Fluxo , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Sensibilidade e Especificidade
8.
Clin Cancer Res ; 2(8): 1353-9, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9816307

RESUMO

Myelosuppression is the dose-limiting toxicity for nitrosourea chemotherapy due to low levels of the DNA repair protein O6-alkylguanine-DNA alkyltransferase in myeloid precursors. We have shown that high-efficiency myeloproliferative sarcoma virus (vM5MGMT)-mediated transduction of the human MGMT cDNA into murine bone marrow (BM) cells leads to high MGMT expression and increased progenitor resistance to 1,3-bis-(2-chloroethyl) nitrosourea (BCNU) in vitro immediately after infection and after BM transplantation. These experiments were designed to increase MGMT expression in human hematopoietic progenitors. CD34(+) BM cells were isolated over an immunoaffinity column (CEPRATE, CellPro, Inc.), resulting in a mean 66-fold enrichment in clonogenic progenitors (colony-forming unit granulocyte-macrophage + burst-forming unit erythroid + colony-forming unit granulocyte erythroid macrophage = megakaryocyte), with an average progenitor yield of 58 +/- 11.5% and a final population that was 54% CD34(+). Seventy % of progenitors derived from CD34(+) cells were transduced after coculture with AM12-vM5MGMT retroviral producers. vM5MGMT-transduced progenitors were over 2-fold more resistant to concentrations of BCNU between 30 and 50 micrometer than were concurrently LacZ-transduced progenitors (P < 0.003). In vitro selection of transduced, cytokine-stimulated CD34(+) cells with 20 micrometer BCNU resulted in survival of 4.7% of MGMT+ clonogenic progenitors compared to 0.05% of LacZ+ progenitors. These studies indicate that MGMT-transduced human hematopoietic progenitors have increased resistance to nitrosoureas, and in a clinical transplant setting, this strategy may reduce alkylating agent myelosuppression.


Assuntos
Carmustina/efeitos adversos , DNA Complementar/genética , Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/genética , Retroviridae/genética , Antígenos CD34/análise , Resistencia a Medicamentos Antineoplásicos , Humanos
9.
Exp Hematol ; 27(11): 1675-81, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10560915

RESUMO

Human bone marrow contains mesenchymal stem cells (MSCs) that can differentiate into various cells of mesenchymal origin. We developed an efficient method of isolating and culture expanding a homogenous population of MSCs from bone marrow and determined that MSCs express alpha-L-iduronidase, arylsulfatase-A and B, glucocerebrosidase, and adrenoleukodystrophy protein. These findings raised the possibility that MSCs may be useful in the treatment of storage disorders. To determine if donor derived MSCs are transferred to the recipients with lysosomal or peroxisomal storage diseases by allogeneic hematopoietic stem cell (HSC) transplantation, we investigated bone marrow derived MSCs of 13 patients 1-14 years after allogeneic transplantation. Highly purified MSCs were genotyped either by fluorescence in situ hybridization using probes for X and Y-chromosomes in gender mis-matched recipients or by radiolabeled PCR amplification of polymorphic simple sequence repeats. Phenotype was determined by the measurement of disease specific protein/enzyme activity in purified MSCs. We found that MSCs isolated from recipients of allogeneic HSC transplantation are not of donor genotype and have persistent phenotypic defects despite successful donor type hematopoietic engraftment. Whether culture expanded normal MSCs can be successfully transplanted into patients with storage diseases and provide therapeutic benefit needs to be determined.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Doenças por Armazenamento dos Lisossomos/terapia , Mesoderma/citologia , Transtornos Peroxissômicos/terapia , Estudos de Casos e Controles , Criança , Pré-Escolar , Humanos , Lactente , Fenótipo , Polimorfismo Genético , Transplante Homólogo , Resultado do Tratamento
10.
Hum Gene Ther ; 10(6): 1021-30, 1999 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-10223735

RESUMO

O6-Benzylguanine (BG) is a potent inhibitor of the DNA repair protein 06-alkylguanine DNA alkyltransferase (AGT), and sensitizes tumors to BCNU in vitro and in xenografts. The combination of BG and BCNU is now undergoing phase I clinical testing. The maximally tolerated dose of BCNU given after BG is expected to be lower then the doses tolerated as a single agent owing to BG sensitization of hematopoietic progenitors. We have previously shown that retroviral expression of G156A mutant MGMT (deltaMGMT) in mouse and human marrow cells results in significant BG and BCNU resistance. In this study we evaluated the effect of deltaMGMT-transduced marrow infusion on the therapeutic index of multiple BG and BCNU treatments in tumor-bearing nude (nu/nu athymic) mice. Prior to subcutaneous implantation of BCNU-resistant SW480 human colon cancer cells, cohorts of mice were given intraperitoneal injections of nonablative doses of BG (30 mg/kg) and BCNU (10 mg/kg, one-half of the LD10) and then infused with 1-2 x 10(6) isogeneic deltaMGMT (n = 29 mice) or lacZ-transduced (n = 20 mice) marrow cells. The xenograft-bearing mice were treated with multiple cycles of BG (30 mg/kg) and BCNU (10-25 mg/kg). After three cycles, deltaMGMT mouse bone marrow was repopulated with CFU containing the provirus, and demonstrated a 2.7-fold increase in AGT activity and a 5.5-fold increase in BCNU IC90 compared with LacZ mice. After five cycles, the BCNU IC90 of CFU cells increased nine-fold over control cells, indicating selective enrichment of CFU precursor cells expressing high levels of deltaMGMT. Starting with the third cycle of therapy, tolerance to BG and BCNU was significantly improved in deltaMGMT mice compared with LacZ mice, as evidenced by preserved peripheral blood counts, bone marrow cellularity, and CFU content 1 and 2 weeks posttreatment and a significantly higher survival rate. Xenograft growth was significantly delayed in mice tolerating multiple cycles and higher dose intensity of BG and BCNU as compared with mice receiving less intensive therapy. We conclude that deltaMGMT-transduced marrow cells can improve the therapeutic index of BG and BCNU by selectively repopulating the marrow and providing significant marrow tolerance to this combination, allowing intensive therapy of a BCNU-resistant tumor.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Transplante de Medula Óssea , Carmustina/uso terapêutico , Neoplasias do Colo/terapia , Guanina/análogos & derivados , O(6)-Metilguanina-DNA Metiltransferase/genética , Transdução Genética , Animais , Antineoplásicos Alquilantes/farmacologia , Carmustina/farmacologia , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos , Vetores Genéticos , Guanina/uso terapêutico , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Retroviridae/genética
11.
Cancer Gene Ther ; 6(4): 340-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10419052

RESUMO

Human hematopoietic progenitors express low levels of O6-alkylguanine-DNA alkyltransferase and are sensitive to 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), particularly following O6-benzylguanine (BG)-mediated O6-alkylguanine-DNA alkyltransferase inhibition. Expression of the BG-resistant mutant (G156A) methylguanine methyltransferase (deltaMGMT) gene in hematopoietic cells confers resistance to BG and BCNU. Because BCNU targets both early and late human hematopoietic cells and results in prolonged and cumulative myelosuppression, we attempted to protect early hematopoietic progenitors (long-term culture initiating cells (LTC-ICs)) by retroviral-mediated transfer of the deltaMGMTgene. A total of 33-56% of LTC-ICs were transduced with MFG-deltaMGMT retrovirus as determined by evidence of provirus in secondary colony-forming units at 5 weeks of culture under conditions optimal for the survival and proliferation of early hematopoietic progenitors. The addition of flt-3 ligand to cultures increased the transduction rate of LTC-ICs. Furthermore, 17.8 +/- 8.1% of deltaMGMT-transduced LTC-ICs survived doses of BG and BCNU; these doses allowed the survival of only 0-1% of untransduced LTC-ICs. This finding compares favorably with the 8-12% of CD34+ cell-derived colony-forming units that we previously showed became resistant to BG and BCNU after deltaMGMTgene transfer. Thus, deltaMGMT transduction of human early hematopoietic progenitor LTC-ICs confers resistance to BG and BCNU and may allow transduced LTC-ICs selective survival and enrichment over untransduced cells in patients undergoing BG and BCNU chemotherapy.


Assuntos
Carmustina/farmacologia , Sobrevivência Celular/genética , Técnicas de Transferência de Genes , Guanina/análogos & derivados , Células-Tronco Hematopoéticas/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/genética , Antígenos CD34/imunologia , Citometria de Fluxo , Guanina/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Células K562 , Mutação
12.
Semin Oncol ; 23(1): 46-65, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8607032

RESUMO

A number of drug resistance genes have been identified that may be useful in gene therapy approaches to ameliorate chemotherapy toxicity. Hematopoietic tissue is the most suitable target for drug resistance gene therapy because myelosuppression is the dose-limiting toxicity of the many chemotherapeutic agents. Recent studies have shown that murine and human hematopoietic progenitors can be transduced ex vivo using retroviral vectors to overexpress P-glycoprotein, dihydrofolate reductase, and O6-alkylguanine DNA alkyltransferase. In all instances, gene transfer results in significant drug resistance in hematopoietic progenitors both in vitro and in vivo. Clinical trials are underway to evaluate the role of MDR-1 gene therapy in amelioration of chemotherapy induced myelosuppression. Other genes being examined for their potential to transfer drug resistance to hematopoietic cells include genes encoding aldehyde dehydrogenase, nucleotide excision repair proteins, multidrug resistant protein, and superoxide dismutase. As a group these proteins could confer significant levels of chemotherapy drug resistance to bone marrow cells. When compared with other somatic gene therapy approaches, drug resistance gene therapy has the aim of protecting normal cells and preventing toxicity. In addition many of these genes could be used to select for cells carrying the drug resistance gene as well as cotransduced therapeutic gene. Thus, gene transfer of drug resistance genes will have broad applications in the field of gene therapy as well as in protecting hematopoietic cells from chemotherapy toxicity.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Terapia Genética/métodos , Neoplasias/terapia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Aldeído Desidrogenase/genética , Resistência a Múltiplos Medicamentos/genética , Tolerância a Medicamentos , Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas , Humanos , Metiltransferases/genética , O(6)-Metilguanina-DNA Metiltransferase , Superóxido Dismutase/genética , Tetra-Hidrofolato Desidrogenase/genética
13.
Bone Marrow Transplant ; 27(3): 235-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11277170

RESUMO

In recent years, there has been an increasing interest in non-hematopoietic pluripotent progenitor cells that are found in the bone marrow. Mesenchymal stem cells (MSCs) are the first non-hematopoietic progenitors to be isolated from the bone marrow and extensively characterized. In addition to their ability to support hematopoiesis, MSCs can differentiate into osteocytes, chondrocytes, tenocytes, adipocytes and smooth muscle cells. This article will review our current understanding of bone marrow stroma and MSCs and their potential therapeutic role in the setting of hematopoietic stem cell transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Mesoderma/transplante , Animais , Células da Medula Óssea , Ensaios Clínicos como Assunto , Humanos , Mesoderma/citologia
14.
Bone Marrow Transplant ; 26(8): 831-6, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11081381

RESUMO

High-dose chemotherapy followed by CD34+ enriched peripheral blood progenitor cell (PBPC) transplantation is used for the treatment of primary refractory or relapsed Hodgkin's and non-Hodgkin's lymphomas. The CD34+ enrichment procedure, while reducing tumor burden, may compromise immunological reconstitution in the transplanted patient and result in increased rates of post-transplant infection. We compared infectious complications in patients with lymphoma who were treated with high-dose chemotherapy and supported either with CD34+ enriched PBPC (n = 19) or unmanipulated PBPCs (n = 24). Analysis was limited to patients discharged from initial hospitalization for transplantation with a minimum of 1 year followup and free of lymphoma recurrence. We found a statistically significant increase in the number of patients with one or more infectious events in the CD34+ transplant group (14/19) compared with the unmanipulated PBPC group (9/24, P < 0.01). Greater numbers of patients with two or more infectious events were observed in the CD34+ group (7/19 vs 2/24, P < 0.03) and an increased incidence of bacterial infections was observed in the CD34+ group (10/19 vs 5/24, P < 0.05). Two deaths due to infectious complications were observed in the CD34+ group. There was no significant difference in blood lymphocyte or monocyte recovery between the groups. These data demonstrate a significant increase in the long-term incidence of infectious events in lymphoma patients transplanted with autologous CD34+ enriched PBPCs compared to unmanipulated PBPCs. Thus, patients who undergo CD34+ enriched PBPC transplantation should be followed closely for infectious complications and prolonged infectious prophylaxis should be considered.


Assuntos
Antígenos CD34/análise , Transplante de Células-Tronco Hematopoéticas , Linfoma/terapia , Adulto , Feminino , Humanos , Contagem de Leucócitos , Linfoma/sangue , Linfoma/complicações , Masculino , Pessoa de Meia-Idade , Morbidade , Transplante Autólogo
15.
Bone Marrow Transplant ; 30(4): 215-22, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12203137

RESUMO

Patients with Hurler syndrome (mucopolysaccharidosis type-IH) and metachromatic leukodystrophy (MLD) develop significant skeletal and neurologic defects that limit their survival. Transplantation of allogeneic hematopoietic stem cells results in partial correction of the clinical manifestations. We postulated that some of these defects may be corrected by infusion of allogeneic, multipotential, bone marrow-derived mesenchymal stem cells (MSC). Patients with Hurler syndrome (n = 5) or MLD (n = 6) who previously underwent successful bone marrow transplantation from an HLA-identical sibling were infused with 2-10 x 10(6)/kg MSCs, isolated and expanded from a bone marrow aspirate of the original donor. There was no infusion-related toxicity. In most recipients culture-purified MSCs at 2 days, 30-60 days and 6-24 months after MSC infusion remained of host type. In two patients the bone marrow-derived MSCs contained 0.4 and 2% donor MSCs by FISH 60 days after MSC infusion. In four patients with MLD there were significant improvements in nerve conduction velocities after MSC infusion. The bone mineral density was either maintained or slightly improved in all patients. There was no clinically apparent change in patients' overall health, mental and physical development after MSC infusion. We conclude that donor allogeneic MSC infusion is safe and may be associated with reversal of disease pathophysiology in some tissues. The role of MSCs in the management of Hurler syndrome and MLD should be further evaluated.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Leucodistrofia Metacromática/terapia , Mesoderma/transplante , Mucopolissacaridose I/terapia , Adolescente , Adulto , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Técnicas de Cultura de Células , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Mesoderma/citologia , Condução Nervosa , Quimeras de Transplante , Transplante Homólogo/métodos , Resultado do Tratamento
16.
Bone Marrow Transplant ; 33(6): 597-604, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14716336

RESUMO

Bone marrow-derived mesenchymal stem cells (MSCs) are known to interact with hematopoietic stem cells (HSCs) and immune cells, and represent potential cellular therapy to enhance allogeneic hematopoietic engraftment and prevent graft-versus-host disease (GVHD). We investigated the role of human MSCs in NOD-SCID mice repopulation by unrelated human hematopoietic cells and studied the immune interactions between human MSCs and unrelated donor blood cells in vitro. When hematopoietic stem cell numbers were limited, human engraftment of NOD-SCID mice was observed only after coinfusion of unrelated human MSCs, but not with coinfusion of mouse mesenchymal cell line. Unrelated human MSCs did not elicit T-cell activation in vitro and suppressed T-cell activation by Tuberculin and unrelated allogeneic lymphocytes in a dose-dependent manner. Cell-free MSC culture supernatant, mouse stromal cells and human dermal fibroblasts did not elicit this effect. These preclinical data suggest that unrelated, human bone marrow-derived, culture-expanded MSCs may improve the outcome of allogeneic transplantation by promoting hematopoietic engraftment and limiting GVHD and their therapeutic potential should be tested in clinic.


Assuntos
Células-Tronco Hematopoéticas/citologia , Ativação Linfocitária/imunologia , Mesoderma/citologia , Transplante de Células-Tronco/métodos , Linfócitos T/imunologia , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Citometria de Fluxo , Humanos , Terapia de Imunossupressão , Interferon gama/análise , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante Heterólogo
17.
Bone Marrow Transplant ; 21(4): 337-43, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9509966

RESUMO

In order to demonstrate the feasibility of mobilization, enrichment and engraftment of autologous peripheral blood CD34+ cells in patients with relapsed lymphoma, 59 peripheral blood progenitor cell (PBPC) collections from 21 patients were enriched for CD34+ cells using CEPRATE SC (CellPro, Bothell, WA, USA) immunoaffinity column. Following high-dose chemotherapy, a mean of 17 x 10(8) (range, 3-34) nucleated cells/kg containing 8.7 x 10(6) (0.3-26) CD34+ cells/kg were re-infused. Blood cell recovery in these patients was compared to engraftment capacity of unenriched PBPCs in a cohort of lymphoma patients treated with an identical high-dose chemotherapy regimen. Neutrophil and platelet engraftment was rapid in both groups including five patients who received < or = 1 x 10(6) CD34+ cells/kg. After infusion of CD34+ enriched cells, neutrophils exceeded 0.5 x 10(9)/l in 11 (8-14) days and platelets exceeded 20 x 10(9)/l (untransfused) in 15 (9-39) days. In order to optimize the immunoaffinity column utilization we stored the first PBPC collections overnight at 4 degrees C and combined them with the next day's collection prior to the CD34+ enrichment procedure in 11 patients. This maneuver resulted in a significant decrease in the CD34+ cell recovery (resulting in reinfusion of a mean of 42% less CD34+ cells). Although overnight storage did not affect neutrophil engraftment, platelet engraftment was prolonged in this group of patients even when > 2.0 x 10(6) CD34+ cells/kg were re-infused. The overnight storage procedure should be further evaluated for its effects on the CD34+ immunoaffinity enrichment procedure, megakaryocyte progenitors and platelet engraftment. We conclude that CD34+ cells enriched from peripheral blood result in rapid engraftment after high-dose chemotherapy in patients with advanced lymphoma that is comparable to that of patients receiving unenriched PBPCs.


Assuntos
Antígenos CD34/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Linfoma/terapia , Adulto , Preservação de Sangue , Feminino , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/imunologia , Doença de Hodgkin/terapia , Humanos , Leucaférese , Linfoma não Hodgkin/terapia , Masculino , Pessoa de Meia-Idade , Fatores de Tempo , Transplante Autólogo
18.
Bone Marrow Transplant ; 25(5): 559-66, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10713636

RESUMO

The purpose of this study was to investigate whether storing mobilized peripheral blood progenitor cell (PBPC) collections overnight before CD34+ selection may delay platelet count recovery after high-dose chemotherapy and CD34+-enriched PBPC re-infusion. Lymphoma patients underwent PBPC mobilization with cyclophosphamide 4 g/m2 i.v. and G-CSF 10 microg/kg/day subcutaneously. Patients were prospectively randomized to have each PBPC collection enriched for CD34+ cells with the CellPro CEPRATE SC System either immediately or after overnight storage at 4 degrees C. Thirty-four patients were randomized to overnight storage and 34 to immediate processing of PBPC; 15 were excluded from analysis due to tumor progression or inadequate CD34+ cell mobilization. PBPC from 23 patients were stored overnight, while 30 subjects underwent immediate CD34+ selection and cryopreservation. Median yield of CD34+ enrichment was 43.6% in the immediate processing group compared to 39.1% in the overnight storage group (P = 0.339). Neutrophil recovery >500 x 10(9)/l occurred a median of 11 days (range 9-16 days) in the overnight storage group compared to 10.5 days (range 9-21 days) in the immediate processing group (P = 0.421). Median day to platelet transfusion independence was 13 (range 7-43) days in the overnight storage group vs 13.5 (range 8-35) days in those assigned to immediate processing (P = 0.933). We conclude that storage of PBPC overnight at 4 degrees C allows pooling of consecutive-day collections resulting in decreased costs and processing time without compromising neutrophil and platelet engraftment after infusion of CD34+-selected progenitor cells. Bone Marrow Transplantation(2000) 25, 559-566.


Assuntos
Antígenos CD34/sangue , Preservação de Sangue , Transplante de Células-Tronco Hematopoéticas/economia , Transplante de Células-Tronco Hematopoéticas/métodos , Leucaférese/economia , Linfoma/terapia , Adolescente , Adulto , Idoso , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Plaquetas/imunologia , Contagem de Células , Criopreservação , Feminino , Sobrevivência de Enxerto , Mobilização de Células-Tronco Hematopoéticas , Humanos , Linfoma/economia , Linfoma/patologia , Masculino , Pessoa de Meia-Idade , Neutrófilos/citologia , Contagem de Plaquetas , Transfusão de Plaquetas , Estudos Prospectivos , Transplante Autólogo/economia
19.
Cancer Treat Res ; 87: 123-46, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8886451

RESUMO

The mechanism of cytotoxicity of a number of chemotherapeutic agents involves alkylation at the O6 position of guanine, a site that strongly influences cytotoxicity. Repair of these lesions by the alkyltransferase protects from cytotoxicity and is a major mechanism of resistance to these agents. O6-benzylguanine inhibition of alkyltransferase sensitizes tumor cells, and clinical trials are underway to determine its efficacy. The use of gene therapy to enhance the expression of alkyltransferase in hematopoietic cells may prevent dose-limiting myelosuppression and may enhance the utility of this class of chemotherapeutic agents.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Reparo do DNA , Guanina/análogos & derivados , Metiltransferases/fisiologia , Sequência de Aminoácidos , Animais , Resistência a Medicamentos , Terapia Genética , Guanina/farmacologia , Humanos , Metiltransferases/antagonistas & inibidores , Metiltransferases/química , Dados de Sequência Molecular , O(6)-Metilguanina-DNA Metiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA