Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
1.
Leukemia ; 32(2): 510-519, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28663580

RESUMO

The concept of arming antibodies with bioactive payloads for a site-specific therapy of cancer has gained considerable interest in recent years. However, a successful antibody-based targeting approach critically relies on the availability of a tumor-associated target that is not only preferentially expressed in the tumor tissue but is also easily accessible for antibody therapeutics coming from the bloodstream. Here, we perfused the vasculature of healthy and acute myeloid leukemia (AML)-bearing rats with a reactive ester derivative of biotin and subsequently quantified the biotinylated proteins to identify AML-associated bone marrow (BM) antigens accessible from the bloodstream. In total, >1400 proteins were identified. Overall, 181 proteins were >100-fold overexpressed in AML as compared with normal BM. Eleven of the most differentially expressed proteins were further validated by immunohistochemistry and confocal microscopic analyses, including novel antigens highly expressed in AML cells (for example, adaptor-related protein complex 3 ß2) and in the leukemia-modified extracellular matrix (ECM) (for example, collagen-VI-α-1). The presented atlas of targetable AML-associated BM proteins provides a valuable basis for the development of monoclonal antibodies that could be used as carriers for a site-specific pharmacodelivery of cytotoxic drugs, cytokines or radionuclides to the BM in AML.


Assuntos
Anticorpos Monoclonais/farmacologia , Medula Óssea/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Citocinas/metabolismo , Humanos , Imuno-Histoquímica/métodos , Masculino , Ratos , Ratos Endogâmicos BN
2.
Leukemia ; 30(10): 2064-2073, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27125308

RESUMO

Although NOD-SCID IL2Rγ-/- (NSG) xenograft mice are currently the most frequently used model to study human leukemia in vivo, the absence of a human niche severely hampers faithful recapitulation of the disease. We used NSG mice in which ceramic scaffolds seeded with human mesenchymal stromal cells were implanted to generate a human bone marrow (huBM-sc)-like niche. We observed that, in contrast to the murine bone marrow (mBM) niche, the expression of BCR-ABL or MLL-AF9 was sufficient to induce both primary acute myeloid leukemia (AML) and acute lymphocytic leukemia (ALL). Stemness was preserved within the human niches as demonstrated by serial transplantation assays. Efficient engraftment of AML MLL-AF9 and blast-crisis chronic myeloid leukemia patient cells was also observed, whereby the immature blast-like phenotype was maintained in the huBM-sc niche but to a much lesser extent in mBM niches. We compared transcriptomes of leukemias derived from mBM niches versus leukemias from huBM-like scaffold-based niches, which revealed striking differences in the expression of genes associated with hypoxia, mitochondria and metabolism. Finally, we utilized the huBM-sc MLL-AF9 B-ALL model to evaluate the efficacy of the I-BET151 inhibitor in vivo. In conclusion, we have established human niche models in which the myeloid and lymphoid features of BCR-ABL+ and MLL-AF9+ leukemias can be studied in detail.


Assuntos
Medula Óssea/patologia , Modelos Animais de Doenças , Proteínas de Fusão bcr-abl , Leucemia Mieloide Aguda/patologia , Proteína de Leucina Linfoide-Mieloide , Proteínas de Fusão Oncogênica , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Animais , Humanos , Camundongos , Transplante Heterólogo
3.
Leukemia ; 29(10): 2039-49, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25975191

RESUMO

Daratumumab is an anti-CD38 monoclonal antibody with lytic activity against multiple myeloma (MM) cells, including ADCC (antibody-dependent cellular cytotoxicity) and CDC (complement-dependent cytotoxicity). Owing to a marked heterogeneity of response to daratumumab therapy in MM, we investigated determinants of the sensitivity of MM cells toward daratumumab-mediated ADCC and CDC. In bone marrow samples from 144 MM patients, we observed no difference in daratumumab-mediated lysis between newly diagnosed or relapsed/refractory patients. However, we discovered, next to an expected effect of effector (natural killer cells/monocytes) to target (MM cells) ratio on ADCC, a significant association between CD38 expression and daratumumab-mediated ADCC (127 patients), as well as CDC (56 patients). Similarly, experiments with isogenic MM cell lines expressing different levels of CD38 revealed that the level of CD38 expression is an important determinant of daratumumab-mediated ADCC and CDC. Importantly, all-trans retinoic acid (ATRA) increased CD38 expression levels but also reduced expression of the complement-inhibitory proteins CD55 and CD59 in both cell lines and primary MM samples. This resulted in a significant enhancement of the activity of daratumumab in vitro and in a humanized MM mouse model as well. Our results provide the preclinical rationale for further evaluation of daratumumab combined with ATRA in MM patients.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Sinergismo Farmacológico , Glicoproteínas de Membrana/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Monoclonais/administração & dosagem , Citotoxicidade Celular Dependente de Anticorpos , Apoptose/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Proliferação de Células/efeitos dos fármacos , Citotoxicidade Imunológica , Proteínas de Ligação a DNA/fisiologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mieloma Múltiplo/patologia , Recidiva Local de Neoplasia/patologia , Terapia de Salvação , Tretinoína/administração & dosagem , Células Tumorais Cultivadas
4.
Ann Rheum Dis ; 74(4): 769-77, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24395558

RESUMO

OBJECTIVES: To explore the immunosuppressive effect and mechanism of action of intraperitoneal (ip) and intra-articular (ia) mesenchymal stem cell (MSC) injection in proteoglycan induced arthritis (PGIA). METHODS: MSC were administered ip or ia after establishment of arthritis. We used serial bioluminescence imaging (BLI) to trace luciferase-transfected MSC. Mice were sacrificed at different time points to examine immunomodulatory changes in blood and secondary lymphoid organs. RESULTS: Both ip and local ia MSC injection resulted in a beneficial clinical and histological effect on established PGIA. BLI showed that MSC ip and ia in arthritic mice are largely retained for several weeks in the peritoneal cavity or injected joint respectively, without signs of migration. Following MSC treatment pathogenic PG-specific IgG2a antibodies in serum decreased. The Th2 cytokine IL-4 was only upregulated in PG-stimulated lymphocytes from spleens in ip treated mice and in lymphocytes from draining lymph nodes in ia treated mice. An increase in production of IL-10 was seen with equal distribution. Although IFN-γ was also elevated, the IFN-γ/IL-4 ratio in MSC treated mice was opposite to the ratio in (untreated) active PGIA. CONCLUSIONS: MSC treatment, both ip and ia, suppresses PGIA, a non-collagen induced arthritis model. MSC are largely retained for weeks in the injection region. MSC treatment induced at the region of injection a deviation of PG-specific immune responses, suggesting a more regulatory phenotype with production of IL-4 and IL-10, but also of IFN-γ, and a systemic decrease of pathogenic PG-specific IgG2a antibodies. These findings underpin the potential of MSC treatment in resistant arthritis.


Assuntos
Artrite Experimental/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Animais , Anticorpos/imunologia , Artrite Experimental/induzido quimicamente , Feminino , Tolerância Imunológica/imunologia , Imunoglobulina G/imunologia , Injeções Intra-Articulares , Injeções Intraperitoneais , Interferon gama/imunologia , Interleucina-4/imunologia , Medições Luminescentes , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteoglicanas/imunologia , Proteoglicanas/toxicidade , Baço/citologia , Baço/imunologia
5.
Cell Transplant ; 22(7): 1171-83, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23031585

RESUMO

Hematopoietic stem cell transplantation (HSCT) using umbilical cord blood (UCB) progenitors is increasingly being used. One of the problems that may arise after UCB transplantation is an impaired engraftment. Either intrabone (IB) injection of hematopoietic progenitors or mesenchymal stem cell (MSC) coadministration has been proposed among the strategies to improve engraftment. In the current study, we have assessed the effects of both approaches. Thus, NOD/SCID recipients were transplanted with human UCB CD34+ cells administered either intravenously (IV) or IB, receiving or not bone marrow (BM)-derived MSCs also IV or IB (in the right femur). Human HSC engraftment was measured 3 and 6 weeks after transplantation. Injected MSCs were tracked weekly by bioluminescence. Also, lodgment within the BM niche was assessed at the latter time point by immuno-fluorescence. Our study shows regarding HSC engraftment that the number of BM human CD45+ cells detected 3 weeks after transplantation was significantly higher in mice cotransplanted with human MSCs. Moreover, these mice had a higher myeloid (CD13+) engraftment and a faster B-cell (CD19+) chimerism. At the late time point evaluated (6 weeks), human engraftment was higher in the group in which both strategies were employed (IB injection of HSC and MSC coadministration). When assessing human MSC administration route, we were able to track MSCs only in the injected femurs, whereas they lost their signal in the contralateral bones. These human MSCs were mainly located around blood vessels in the subendosteal region. In summary, our study shows that MSC coadministration can enhance HSC engraftment in our xenogenic transplantation model, as well as IB administration of the CD34+ cells does. The combination of both strategies seems to be synergistic. Interestingly, MSCs were detected only where they were IB injected contributing to the vascular niche.


Assuntos
Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Mesenquimais , Adulto , Animais , Antígenos CD34/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Osso e Ossos/fisiologia , Quimerismo , Feminino , Sobrevivência de Enxerto/imunologia , Células HEK293 , Células-Tronco Hematopoéticas/citologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Transplante Heterólogo
6.
J Cell Mol Med ; 16(8): 1827-39, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21973026

RESUMO

Although mesenchymal stromal cells (MSCs) have been applied clinically to treat cardiac diseases, it is unclear how and to which extent transplanted MSCs exert their beneficial effects. To address these questions, pre-clinical MSC administrations are needed for which pigs appear to be the species of choice. This requires the use of porcine cells to prevent immune rejection. However, it is currently unknown to what extent porcine MSCs (pMSCs) resemble human MSCs (hMSCs). Aim of this study was to compare MSC from porcine bone marrow (BM) with human cells for phenotype, multi-lineage differentiation potential, immune-modulatory capacity and the effect on cardiac function after transplantation in a mouse model of myocardial infarction. Flow cytometric analysis revealed that pMSC expressed surface antigens also found on hMSC, including CD90, MSCA-1 (TNAP/W8B2 antigen), CD44, CD29 and SLA class I. Clonogenic outgrowth was significantly enriched following selection of CD271+ cells from BM of human and pig (129 ± 29 and 1961 ± 485 fold, respectively). hMSC and pMSC differentiated comparably into the adipogenic, osteogenic or chondrogenic lineages, although pMSC formed fat much faster than hMSC. Immuno-modulation, an important feature of hMSC, was clearly demonstrated for pMSC when co-cultured with porcine peripheral blood cells stimulated with PMA and pIL-2. Finally, pMSC transplantation after myocardial infarction attenuated adverse remodelling to a similar extent as hMSC when compared to control saline injection. These findings demonstrate that pMSCs have comparable characteristics and functionality with hMSCs, making reliable extrapolation of pre-clinical pMSC studies into a clinical setting very well possible.


Assuntos
Diferenciação Celular , Imunomodulação/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Miocárdio/patologia , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia , Animais , Antígenos CD/metabolismo , Células da Medula Óssea/citologia , Proliferação de Células , Separação Celular , Condrogênese , Citometria de Fluxo , Testes de Função Cardíaca , Humanos , Imunofenotipagem , Masculino , Camundongos , Camundongos SCID , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese , Fenótipo , Sus scrofa , Linfócitos T/citologia , Linfócitos T/metabolismo
7.
J Orthop Res ; 22(4): 697-702, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15183423

RESUMO

The use of bone marrow derived stromal cells (BMSC's) for bone tissue engineering has gained much attention as an alternative for autologous bone grafting. Little is known however, about the survival and differentiation of the cells, especially in the clinical application. The aim of this study was to develop a method to trace goat BMSC's in vivo. We investigated retroviral genetic marking, which allows stable expression of the label with cell division. Goat BMSC's were subjected to an amphotropic envelope containing a MoMuLV-based vector expressing the human low affinity nerve growth factor receptor (DeltaLNGFR). Labeling efficiency and effect on the cells were analyzed. Furthermore, transduced cells were seeded onto porous ceramic scaffolds, implanted subcutaneously in nude mice and examined after successive implantation periods. Flow cytometry indicated a transduction efficiency of 40-60%. Immunohistochemistry showed survival and subsequent bone formation of the gene-marked cells in vivo. Besides, marked cells were also found in cartilage and fibrous tissue. These findings indicate the maintenance of the precursor phenotype following gene transfer as well as the ability of the gene to be expressed following differentiation. We conclude that retroviral gene marking with DeltaLNGFR is applicable to trace goat BMSC's in bone tissue engineering research.


Assuntos
Células da Medula Óssea/citologia , Receptor de Fator de Crescimento Neural/genética , Retroviridae/genética , Engenharia Tecidual/métodos , Animais , Desenvolvimento Ósseo , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Osso e Ossos/metabolismo , Sobrevivência Celular , Células Cultivadas , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Técnicas de Transferência de Genes , Marcadores Genéticos , Vetores Genéticos , Cabras , Humanos , Camundongos , Camundongos Nus , Receptor de Fator de Crescimento Neural/metabolismo , Células Estromais/citologia , Células Estromais/transplante
8.
Leukemia ; 14(5): 889-97, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10803522

RESUMO

Among a variety of immunodeficient mouse strains the non-obese diabetic (NOD)/LtSz scid/scid strain appears to be most useful in allowing the engraftment of human AML. However, the large variability in ability to engraft and the levels of engraftment reached have not been explained. To address these issues we have investigated the NOD/SCID repopulating ability of 27 newly diagnosed AML samples. Patients were selected for the absence of internal tandem duplications in the Flt3 gene as we previously reported this mutation to be associated with an enhanced engraftment potential in this model. We observed that secondary AML (n = 6) had a significantly increased level of engraftment when compared to primary AML (n = 21, median levels 73.3% for secondary AML vs 8.94% for primary AML, P = 0.01). Within the primary AML, a significantly higher engraftment was observed in the FAB class M0 than in FAB classes M2, M4 and M5. Within primary AML, samples of patients who failed to respond to the initial therapy gave rise to a higher level of engraftment than samples of patients who did respond to therapy. A similar observation of an increased engraftment correlating with a poorer patient prognosis could be made when applying cytogenetic risk stratification. However, within the primary AML the most important clinical parameter correlating with the level of engraftment appeared to be the patient's WBC count at diagnosis (P = 0.0000). Covariate analysis with the WBC count as a covariate could also fully explain the differences observed in the cytogenetic risk groups, or on the basis of the initial therapy response. Although large differences could be observed, the ability to engraft the NOD/SCID mice was not linked to either the autonomous or cytokine-induced proliferation in vitro. As the leukemic cobblestone area-forming cell frequencies also revealed no correlation with repopulation in the NOD/SCID model, we consider it very likely that the level of engraftment reflects the in vivo proliferative ability of the AML samples assayed rather than the number of leukemia-initiating cells infused into the NOD/SCID mice. Phenotypic analysis based on the expression of CD33, CD34 and CD38 before and after passage in NOD/SCID showed that in 10 out of 16 samples investigated phenotypes were different. In summary, in addition to the Flt3 internal tandem duplications we have identified a series of clinical parameters that determine the NOD/SCID repopulating ability of AML samples, whilst our data strongly suggest that AML in NOD/SCID does not reflect the leukemic process in the patient.


Assuntos
Leucemia Mieloide/patologia , Transplante Heterólogo/patologia , Doença Aguda , Adolescente , Adulto , Idoso , Animais , Antígenos CD/análise , Medula Óssea/patologia , Divisão Celular , Criança , Pré-Escolar , Modelos Animais de Doenças , Humanos , Leucemia Mieloide/classificação , Leucemia Mieloide/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Transplante de Neoplasias , Transplante Heterólogo/imunologia , Células Tumorais Cultivadas
10.
Eur J Cancer ; 35(4): 634-40, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10492639

RESUMO

The in vivo effect of the radiochemoprotectant Amifostine on the therapeutic efficacy of marrow ablative treatment with cyclophosphamide (CP) and total body irradiation (TBI) followed by bone marrow transplantation (BMT) was studied in normal rats as well as in the Brown Norway rat acute myelocytic leukaemia (BNML) model. In normal rats, when the dose of TBI was escalated and the CP dose was kept constant, pretreatment with Amifostine yielded a positive dose modification factor of 1.26. No significant improvement was found after Amifostine pretreatment when the TBI dose was kept constant and CP dose escalated. When leukaemic rats received CP as the only antileukaemia treatment, Amifostine pretreatment did not lead to a reduction in the antileukaemic efficacy of CP, although protection against treatment-related mortality was observed. In the CP only groups, 9 out of 40 animals died of treatment-related toxicity, compared with none of the 40 animals in the Amifostine pretreatment groups. When applying the maximum tolerated treatment of CP and TBI in various combinations to leukaemic rats, 25 out of 36 rats died from treatment-related toxicity, whilst pretreatment with Amifostine reduced this to 11 out of 36, (P = 0.002). Of those animals which survived the CP + TBI conditioning treatment, 10 out of 25 in the Amifostine pretreatment group were cured, versus 8/11 in the CP + TBI only control group (P = 0.146). In conclusion, incorporation of Amifostine as a radiochemoprotectant in a marrow-ablative conditioning regimen allows the use of escalated doses of chemoradiotherapy without reducing the antileukaemic efficacy.


Assuntos
Amifostina/administração & dosagem , Leucemia Mieloide Aguda/terapia , Protetores contra Radiação/administração & dosagem , Irradiação Corporal Total , Animais , Antineoplásicos Alquilantes/uso terapêutico , Transplante de Medula Óssea/métodos , Terapia Combinada , Ciclofosfamida/uso terapêutico , Relação Dose-Resposta a Droga , Masculino , Ratos , Ratos Endogâmicos , Análise de Sobrevida
11.
Gene Ther ; 6(6): 1139-46, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10455417

RESUMO

The in vivo elimination of suicide gene-expressing tumor cells with prodrug treatment can induce protective immunity against wild-type tumors. In this study, we determined the efficacy and safety of the in vivo elimination of HSVtk expressing cells with ganciclovir treatment of a bystander cell killing-insensitive leukemic cell line. The retroviral construct pLTk+NeoDeltaMo, containing the HSVtk gene and the NeoR gene in a bicistronic unit, was introduced into rat leukemic LT12 cells. LT12/Tk+N cells showed a 1000- to 10 000-fold increased sensitivity to ganciclovir in vitro. In vitro mixing experiments demonstrated that LT12 cells were not susceptible to bystander cell lysis by LT12/Tk+N-2 cells exposed to ganciclovir. Rats injected s.c. with cloned LT12/Tk+N-2 cells developed tumors reaching a diameter of 3-4 cm after 40 days. Rats treated with gan- ciclovir twice daily for 5 consecutive days starting at day 7 did not develop s.c. tumors. Large established s.c. LT12/Tk+N-2 tumors completely regressed after ganciclovir treatment. However, recurrences of s.c. tumors were observed that were no longer sensitive to ganciclovir treatment. In vitro analysis of aspirates from the recurrent tumors demonstrated loss of HSVtk expression. In vitro culture of LT12/Tk+N-2 cells in soft agar in the presence of ganciclovir indicated that the frequency with which HSVtk-loss variants occurred is approximately one per 104 cells. The in vivo occurrence of HSVtk-loss variants escaping ganciclovir-induced elimination may have important implications for vaccination protocols using HSVtk gene expressing tumor cells that are not susceptible to bystander cell killing.


Assuntos
Ganciclovir/uso terapêutico , Terapia Genética/métodos , Imunoterapia/métodos , Leucemia Mieloide/terapia , Simplexvirus/genética , Timidina Quinase/genética , Animais , Citotoxicidade Imunológica , Transplante de Neoplasias , Ratos , Simplexvirus/enzimologia , Células Tumorais Cultivadas
12.
Leukemia ; 13(7): 1071-8, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10400423

RESUMO

Recently, in-frame internal tandem duplications have been reported within the regions coding for the juxtamembrane through the first tyrosine kinase domain of the Flt3 gene. These duplications have been reported to lead to autophosphorylation of the receptor. In this study we investigated the effect of such mutations in the Flt3 gene on the in vitro proliferation of human acute myeloid leukemia cells. The mutations were detected in 10 out of 59 AML bone marrow samples analyzed and were not restricted to a specific FAB class or cytogenetic aberration. PCR analysis of those samples showed all mutations to be present in exon 11 of the gene. Whilst samples without a mutation of the Flt3 gene showed an increased cell production in response to either IL-3 and G-CSF or IL-6, SCF, TPO and Flt3L in long-term stroma supported cultures, mutant samples failed to do so. As we could not find a relationship between the absence of a response and either FAB class or cytogenetic aberrations, we interpret these results as an indication that the internal tandem duplications in the Flt3 gene are the prime cause of this unresponsiveness. Although our study does not explain the mechanism by which these mutations cause this unresponsiveness it does suggest that AML cells need a wild-type Flt3 for optimal in vitro proliferation.


Assuntos
Leucemia Mieloide/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Superfície Celular/genética , Células Estromais/patologia , Sequências de Repetição em Tandem , Doença Aguda , Adolescente , Adulto , Idoso , Sequência de Aminoácidos , Adesão Celular , Divisão Celular/genética , Feminino , Humanos , Leucemia Mieloide/patologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação , Fatores de Tempo , Células Tumorais Cultivadas , Tirosina Quinase 3 Semelhante a fms
13.
Leukemia ; 12(12): 1962-70, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9844926

RESUMO

The severe combined immunodeficient (SCID) mouse model may be used to evaluate new approaches for the treatment of acute myeloid leukemia (AML). We have previously demonstrated the killing of SCID mouse leukemia initiating cells by in vitro incubation with human GM-CSF fused to Diphtheria toxin (DT-huGM-CSF). In this report, we show that in vivo treatment with DT-huGM-CSF eliminates AML growth in SCID mice. Seven cases of AML were studied. SCID mice were treated intraperitoneally with the maximally tolerated dose of 75 microg/kg/day for 7 days. Antileukemic efficacy was determined at days 40 and 80 after transplantation, by enumerating the percentages of human cells in SCID bone marrow using flow cytometry and short tandem repeat polymerase chain reaction (STR-PCR) analysis. Four out of seven AML cases were sensitive to in vivo treatment with DT-huGM-CSF at both evaluation time points. In three of these cases, elimination of human cells was demonstrated by flow cytometry and STR-PCR. One AML case showed moderate sensitivity for DT-huGM-CSF, and growth of the two remaining AML cases was not influenced by DT-huGM-CSF. Sensitivity was correlated with GM-CSFR expression. Our data show that DT-huGM-CSF can be used in vivo to reduce growth of AML and warrant further development of DT-huGM-CSF for the treatment of human AML.


Assuntos
Toxina Diftérica/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Imunotoxinas/uso terapêutico , Leucemia Mieloide Aguda/terapia , Animais , Medula Óssea , Divisão Celular/efeitos dos fármacos , DNA de Neoplasias/análise , Relação Dose-Resposta Imunológica , Feminino , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos SCID , Fenótipo , Organismos Livres de Patógenos Específicos
14.
Bone Marrow Transplant ; 21(8): 801-7, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9603404

RESUMO

Ultraviolet-B (UVB) irradiation is known to inhibit lymphocyte activity and consequently to reduce the incidence of graft-versus-host disease (GVHD) in experimental models for allogeneic bone marrow transplantation (BMT). GVHD is frequently associated with morbidity and mortality, but also with the beneficial graft-versus-leukemia (GVL) effect, demonstrated by a reduction in the incidence of leukemia relapse. In this study, we investigated whether UVB treatment of allogeneic T cells could prevent GVHD while sparing the beneficial GVL effect following allogeneic BMT in the Brown Norway myelocytic leukemia (BNML) rat model analogous to human acute myelocytic leukemia (AML). The dose of UVB required to abolish lethal GVHD in the rat allogeneic BMT model (WAG/Rij donors into BN recipients) was 4000 J/m2. However, this UVB dose simultaneously abrogated all GVL activity mediated by the T cells in the graft, while the radio-protective capacity of rat BM cells was strongly reduced. The number of allogeneic BM cells required to protect lethally irradiated BN rats was increased 50 to 100-fold. It is concluded that UVB acts as a non-selective form of T cell inactivation, and that UVB pretreatment of an allogeneic marrow graft is unlikely to be useful clinically as a preventive measure for GVHD, since other means of reduction of the number of functional T cells are less damaging to bone marrow stem cells.


Assuntos
Transplante de Medula Óssea/efeitos adversos , Doença Enxerto-Hospedeiro/prevenção & controle , Leucemia Mieloide Aguda/terapia , Raios Ultravioleta , Animais , Células da Medula Óssea/efeitos da radiação , Masculino , Ratos , Ratos Endogâmicos BN , Transplante Homólogo , Raios Ultravioleta/efeitos adversos
15.
Leukemia ; 12(5): 710-7, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9593269

RESUMO

We have previously demonstrated that diphtheria toxin (DT) fused to human GM-CSF effectively eliminates human long-term leukemia initiating cells in SCID mice. However, because huGM-CSF does not react with the murine GM-CSF receptor possible side-effects to nonleukemic tissues could not be analyzed in the AML/SCID model. To overcome this problem, we used murine GM-CSF fused to DT and studied the therapeutic index in the rat leukemia model BNML/LT12. In DT-mGM-CSF dose escalation experiments, severe dose-dependent toxicity to organs such as liver, kidney and lung was observed. Therefore, the antileukemic effects were evaluated with the lower doses. Daily intraperitoneal bolus injections of 75 microg/kg/day for 7 days induced a 3 log leukemic cell kill. The dose of 75 microg/kg/day had no effect on the hemopoietic progenitor cell subsets. These in vivo studies show that the DT-GM-CSF fusion protein can be used for specifically targeting leukemic cells and thus has potential as a therapeutic agent in the treatment of AML.


Assuntos
Toxina Diftérica/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Leucemia Experimental/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Doenças da Medula Óssea/induzido quimicamente , Toxina Diftérica/toxicidade , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/toxicidade , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Ratos , Ratos Endogâmicos BN , Proteínas Recombinantes de Fusão/toxicidade , Células Tumorais Cultivadas/efeitos dos fármacos
16.
Br J Haematol ; 98(4): 952-9, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9326195

RESUMO

The potential to selectively eliminate acute myeloid leukaemia (AML) cells with the GM-CSF-diphtheria toxin fusion protein (DT-GM-CSF) was studied under conditions of autonomous proliferation in vitro with no growth factors (GFs) added and after growth stimulation with a mixture of human (hu)G-CSF, huIL-3 and huSCF. DNA synthesis was maximally inhibited after 48 h exposure to DT-GM-CSF. Cell viability and AML colony forming ability in vitro were reduced. 18/22 samples were found to be sensitive to DT-GM-CSF, with 50% inhibition of DNA synthesis (ID50) at concentrations ranging from 0.1 to 16 ng/ml, and four samples were minimally or not sensitive to DT-GM-CSF (ID50 > or = 99 ng/ml). From the 15 samples which showed autonomous proliferation, 13 were sensitive to inhibition of proliferation by DT-GM-CSF. The level of GM-CSF receptor (GM-CSFR) expression was determined by flow cytometry after labelling with specific antibodies for the alpha and beta subunits. Although the toxicity to DT-GM-CSF was specifically mediated by the GM-CSFR, no correlation was found between the level of expression of the GM-CSFR alpha or beta subunit and the sensitivity for DT-GM-CSF. These in vitro studies show that the DT-GM-CSF fusion protein can be used for specifically targeting and eliminating leukaemic cells in the majority of AML cases.


Assuntos
Toxina Diftérica/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Leucemia Mieloide/terapia , Doença Aguda , Divisão Celular , Humanos , Leucemia Mieloide/patologia , Células Tumorais Cultivadas
17.
Blood ; 90(9): 3735-42, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9345060

RESUMO

We studied the cell kill induced by granulocyte-macrophage colony-stimulating factor (GM-CSF ) fused to Diphtheria Toxin (DT-GM-CSF ) in acute myeloid leukemia (AML) samples and in populations of normal primitive hemopoietic progenitor cells. AML samples from three patients were incubated in vitro with 100 ng/mL DT-GM-CSF for 48 hours, and AML cell kill was determined in a proliferation assay, a clonogenic assay colony-forming unit-AML (CFU-AML) and a quantitative long-term bone marrow (BM) culture ie, the leukemic-cobblestone area forming cell assay (L-CAFC). To measure an effect on cells with in vivo leukemia initiating potential DT-GM-CSF exposed AML cells were transplanted into immunodeficient mice. In two out of three samples it was shown that all AML subsets, including those with long-term abilities in vivo (severe combined immunodeficient mice) and in vitro (L-CAFC assay) were reduced in number by DT-GM-CSF. Cell kill induced by DT-GM-CSF could be prevented by coincubation with an excess of GM-CSF, demonstrating that sensitivity to DT-GM-CSF is specifically mediated by the GM-CSF receptor. Therefore, binding and internalization of GM-CSF probably occur in immature AML precursors of these two cases of AML. The third AML sample was not responsive to either GM-CSF or DT-GM-CSF. The number of committed progenitors of normal bone marrow (burst-forming unit-erythroid, colony-forming unit granulocyte- macrophage, and cobble stone area forming cell [CAFC] week 2) and also the number of cells with long-term repopulating ability, assayed as week 6 CAFC, were unchanged after exposure to DT-GM-CSF (100 ng/mL, 48 hours). These studies show that DT-GM-CSF may be used to eliminate myeloid leukemic cells with long-term potential in vitro and in immunodeficient mice, whereas normal hemopoietic stem cells are spared.


Assuntos
Toxina Diftérica/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucemia Experimental/tratamento farmacológico , Leucemia Mieloide/tratamento farmacológico , Proteínas Recombinantes de Fusão/uso terapêutico , Doença Aguda , Animais , Morte Celular/efeitos dos fármacos , Toxina Diftérica/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Experimental/patologia , Leucemia Mieloide/patologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Proteínas Recombinantes de Fusão/genética
18.
Prenat Diagn ; 17(10): 953-9, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9358575

RESUMO

Fetal cells are present in the circulation of pregnant women and can be isolated using density gradient centrifugation and magnetic cell sorting. In the present study, maternal cell preparations were depleted for CD45- and CD14-positive cells and enriched for CD71-positive cells. The number of fetal nucleated cells was determined using fluorescence in situ hybridization for X and Y chromosomes. Analysis of maternal blood samples taken before and after transabdominal chorionic villus sampling (TA-CVS) showed an increase in the number of fetal cells in 10 out of 19 male pregnancies after the invasive procedure. This cellular transfusion was found to correlate with elevated maternal serum alpha-fetoprotein levels. TA-CVS-induced cellular transfusion may form a good in vivo system to optimize fetal cell isolation procedures and to study fetal cell dynamics and characteristics.


Assuntos
Amostra da Vilosidade Coriônica/efeitos adversos , Células Precursoras Eritroides/metabolismo , Transfusão Feto-Materna/sangue , alfa-Fetoproteínas/metabolismo , Adulto , Amostra da Vilosidade Coriônica/métodos , Feminino , Idade Gestacional , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Masculino , Gravidez
19.
J Histochem Cytochem ; 44(11): 1323-9, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8918907

RESUMO

Femora and tibiae of rats carrying leukemia from a LacZ-marked acute promyelocytic leukemia-derived leukemic cell line (LT12NL15) were decalcified using EDTA and routinely embedded in paraffin. Sections were used to develop for the first time an immunostaining method for LacZ, employing catalyzed reporter deposition (CARD) based on the deposition of biotinylated tyramine. This method was used to study homing and adhesion of leukemic cells.


Assuntos
Biomarcadores Tumorais , Óperon Lac , Animais , Leucemia Experimental/genética , Leucemia Experimental/patologia , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patologia , Inclusão em Parafina , Ratos , Sensibilidade e Especificidade , Coloração e Rotulagem/métodos , Células Tumorais Cultivadas
20.
Leukemia ; 10(11): 1796-803, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8892684

RESUMO

We studied the applicability of interleukin-6 Pseudomonas exotoxin fusion protein (IL-6PE4E) for treatment of acute myelocytic leukemia (AML). Leukemic cells from five out of 10 AML patients studied expressed IL-6 receptor (IL-6R) and proliferation in vitro was inhibited in four of these cases. The potential of this approach in vivo was tested in a pre-clinical model for AML; the Brown Norway acute myelocytic leukemia (BNML). To obtain IL-6R expression levels on BNML cells comparable to the numbers expressed on human AML, human IL-6R gene transfectants of the BNML sub-line LT12 (LT12/IL-6R) were generated. IL-6PE4E is cytotoxic in vitro to LT12/IL-6R expressing 1400 high affinity IL-6R per cell with 50% inhibition of DNA synthesis at 1 ng/ml. In vivo treatment of leukemic rats carrying LT12/IL-6R leukemia indicated that the maximal tolerated dose of IL-6PE4E was 275 +/- 25 microg/kg/day, when continuously administered for 7 days and resulted in a 90% reduction in leukemic cell load. At this dose level of IL-6PE4E no reduction of normal hemopoietic progenitors was seen in non-leukemic rats. At higher dose levels (350-1050 microg/kg/day) severe systemic toxicity was encountered. On the basis of these pre-clinical studies the feasibility of growth factor-toxins for selective in vivo targeting to AML cells is evaluated.


Assuntos
Exotoxinas/administração & dosagem , Interleucina-6/administração & dosagem , Leucemia Experimental/tratamento farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Recombinantes de Fusão , ADP Ribose Transferases , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Morte Celular/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Humanos , Bombas de Infusão , Leucemia Experimental/metabolismo , Leucemia Experimental/patologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Ratos , Ratos Endogâmicos BN , Receptores de Interleucina/biossíntese , Receptores de Interleucina/genética , Receptores de Interleucina-6 , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...