Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
2.
Transfusion ; 43(1): 58-64, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12519431

RESUMO

BACKGROUND: Peripheral blood progenitor cells (PBPCs) can be collected on various cell separators. Two leukapheresis programs (LP-MNC and LP-PBSC-Lym) were evaluated for computerized collection of PBPCs on a new cell separator. STUDY DESIGN AND METHODS: Leukapheresis assisted by the LP-MNC or LP-PBSC-Lym software was performed for the harvesting of PBPCs in 52 oncology patients after chemotherapy plus G-CSF treatment and in 18 healthy subjects after G-CSF mobilization alone. RESULTS: A total of 38 components from 33 donors via LP-MNC and 43 components from 37 donors via LP-PBSC-Lym were collected with a median of one (range, one to two) standard-volume leukapheresis procedures (9.2-13.3 L) per donor. There were no significant differences between the two groups concerning median counts of WBCs, CD34+ cells, CD34+ cell yields per harvest, and CD34+ cell yields of cumulative harvests. The blood cell counts after leukapheresis revealed that the LP-MNC resulted in significantly higher platelet loss than LP-PBSC-Lym (p = 0.024): 35.9 percent (range, 19.2%-66.1%) versus 29.7 percent (11.6%-52.3%). Regarding the CD34+ cell collection efficiency, the LP-MNC program was significantly better than the LP-PBSC-Lym program (p < 0.001): 77.5 percent (range, 35.5%-98.9%) versus 58.3 percent (range, 20.4%-98.9%). However, concentrates collected by the LP-PBSC-Lym program had significantly higher percentages of MNCs (p < 0.001) and CD34+ cells (p = 0.028) than harvests with the LP-MNC program: 90 percent (range, 69%-99%) versus 70 percent (range, 35%-98%) and 1.2 percent (range, 0.2%-7.3%) versus 0.7 percent (range, 0.2%-6.0%), respectively. No leukapheresis-related serious adverse events were seen, and time for hematopoietic engraftment was equivalent to data published in the literature. CONCLUSION: The LP-MNC program shows a significantly better CD34+ cell collection efficiency than the LP-PBSC-Lym program. However, collections with the LP-MNC program result in PBPC components with a lower MNC and CD34+ cell concentrations and a higher apheresis-related loss of patient's platelets.


Assuntos
Células-Tronco Hematopoéticas , Leucaférese/métodos , Antígenos CD34/análise , Computadores , Transplante de Células-Tronco Hematopoéticas , Humanos
3.
J Hematother Stem Cell Res ; 10(6): 777-85, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11798504

RESUMO

The rationale for purging of autologous acute myeloid leukemia (AML) grafts is to eradicate contaminating leukemic cells that might contribute to relapse. However, in vitro purging generally delays post-transplant hematopoietic recovery, thus increasing treatment-related complication rates. Theoretically, this prolonged aplasia might be shortened by the additional transplantation of ex vivo-generated progenitor cells. Therefore, we investigated whether nonleukemic progenitors could be expanded ex vivo from AML cell-contaminated CD34(+) peripheral blood progenitor cell (PBPC) preparations. Nonleukemic CD34(+)-selected PBPC and AML cells (Kasumi-1, KG-1, primary AML blasts) were cultured in cytokine-supplemented liquid culture for up to 19 days. Cells were used either unmanipulated or following in vitro purging with mafosfamide (30, 50, 75 microg/ml). Ex vivo-generated cells were assessed by flow cytometry, progenitor cell assays, and polymerase chain reaction. Without prior purging, ex vivo culture markedly amplified AML cells as well as nonleukemic CD34(+) PBPC (day 12: Kasumi-1, 18.5 +/- 0.6-fold; KG-1, 52.2 +/- 2.6-fold; CD34(+), 74.1 +/- 5.6-fold). Co-culture with leukemic cells did not affect CD34(+) cell growth and vice versa. Following in vitro purging, CD34(+) PBPC were expanded even at the highest mafosfamide dose (day 19: 25 +/- 15-fold), whereas leukemic cells were markedly depleted (approx. 1.5 log). Furthermore, normal colony-forming units (CFU) could be effectively recovered (day 19: 10 +/- 3.1% of prepurging input CFU), whereas CFU-L were depleted to undetectable levels in six of seven experiments. Finally, leukemic cells were undetectable following ex vivo co-culture of purged cells (CD34(+) PBPC plus 10% Kasumi-1 cells or primary blasts), but were clearly detectable without purging. Taken together, these data demonstrated that ex vivo expansion of normal progenitors from mafosfamide-purged AML cell-contaminated grafts might be feasible.


Assuntos
Antineoplásicos/farmacologia , Purging da Medula Óssea/métodos , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacologia , Células-Tronco Hematopoéticas/citologia , Leucemia Mieloide/patologia , Doença Aguda , Antígenos CD34 , Células Sanguíneas/citologia , Células Sanguíneas/imunologia , Divisão Celular/efeitos dos fármacos , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/efeitos dos fármacos
4.
Stem Cells ; 18(2): 87-92, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10742380

RESUMO

Recently, a number of devices have been developed for the positive selection of CD34(+) peripheral blood progenitor cells (PBPC) for clinical use in autologous or allogeneic transplantation. The rationale for CD34(+) selection is based on clinical studies showing a two- to five-log reduction of contaminating tumor cells in patients with breast cancer, multiple myeloma and low-grade lymphoma. In addition, a three- to five-log reduction of T cells can be obtained by CD34(+) selection in both autologous grafts for patients with autoimmune disease resistant to conventional therapy and allogeneic grafts to reduce the incidence and severity of acute graft-versus-host disease. Transplantation of positively selected autologous CD34(+) PBPC results in a rapid and stable neutrophil and platelet engraftment in patients who received an infused dose of at least 2.0 x 10(6) CD34(+) cells/kg. Results from randomized trials suggest that time to engraftment is not different compared to unmanipulated PBPC autografts. However, close monitoring for infectious complications (e.g., cytomegalovirus disease) is required. Allogeneic CD34(+) PBPC have also been successfully transplanted and, using novel technologies, megadoses of purified CD34(+) PBPC can be obtained and used to overcome histocompatibility differences betweeen allogeneic donor and patient resulting in stable engraftment, even in a haploidentical setting. Additional randomized phase III trials are required to determine whether tumor cell purging or lymphocyte depletion by CD34(+) cell selection will have a significant impact on progression-free and overall survival in both autologous and allogeneic transplantation.


Assuntos
Antígenos CD34 , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Separação Celular/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Leucócitos Mononucleares/citologia , Transplante Homólogo
5.
Exp Hematol ; 28(4): 460-70, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10781904

RESUMO

OBJECTIVE: Neutropenia following high-dose chemotherapy and peripheral blood progenitor cell (PBPC) transplantation might be abrogated by an additional transplantation of ex vivo generated granulopoietic postprogenitor cells (GPPC). Therefore, the ex vivo expansion of CD34(+) PBPC was systematically studied aiming for optimum GPPC production. MATERIALS AND METHODS: CD34(+) PBPC were cultured in serum-free medium comparing different (n = 32) combinations of stem cell factor (S), interleukin 1 (1), interleukin 3 (IL-3) (3), interleukin-6 (6), erythropoietin (E), granulocyte colony-stimulating factor (G), granulate-macrophage colony-stimulating factor (GM), daniplestim (D, a novel IL-3 receptor agonist), and Flt3 ligand (FL) under various culture conditions. Ex vivo generated cells were assessed by flow cytometry, morphology, and progenitor cell assays. RESULTS: Addition of G +/- GM but not GM alone to cultures stimulated with S163E effectively induced the generation of GPPC. GPPC production was maximum after 12 to 14 days. Best expansion rates were observed when cells were cultured at 1.5x10(4)/mL in 21% O(2). Modifications of culture conditions were either less or equally effective (i.e., modification of starting cell concentrations, low oxygen, addition of serum albumin or autologous plasma, repetitive feeding). Comparison of different cytokine combinations revealed that the optimum GPPC expansion cocktail consisted of S6GD+FL (day 12: 130-fold cellular expansion, 32% myeloblasts/promyelocytes, 49.4% myelocytes/metamyelocytes, 12.4% bands/segmented), which furthermore expanded CD34(+) cells (3.4-fold) and clonogenic progenitors (13.4-fold). CONCLUSION: Using the S6DG+FL expansion cocktail, GPPC could be effectively produced ex vivo starting from positively selected CD34 PBPC, possibly enabling amelioration or even abrogation of posttransplant neutropenia.


Assuntos
Antígenos CD34/biossíntese , Granulócitos/citologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Adjuvantes Imunológicos/farmacologia , Antígenos CD/biossíntese , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Meios de Cultura Livres de Soro/farmacologia , Eritropoetina/farmacologia , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interleucina-1/farmacologia , Interleucina-3/farmacologia , Interleucina-6/farmacologia , Proteínas de Membrana/farmacologia , Fragmentos de Peptídeos , Peptídeos/farmacologia , Fator de Células-Tronco/farmacologia
6.
Semin Hematol ; 37(1 Suppl 2): 42-9, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10718158

RESUMO

Ex vivo manipulations of hematopoietic stem and progenitor cells are Increasingly used in the context of autologous and allogeneic stem-cell transplantation. These manipulations include the positive selection of CD34+ cells for tumor-cell reduction and/or T-cell depletion, the ex vivo expansion of hematopoietic progenitor and stem cells under appropriate cytokine-stimulated culture conditions, and the ex vivo generation of myeloid or megakaryocytic postprogenitor cells and Immune effector cells. This article summarizes both the preclinical data on the ex vivo expansion of hematopoietic progenitor and stem cells from purified CD34+ cells and the Initial clinical studies with ex vivo-expanded stem and progenitor cells for hematopoietic support after high-dose chemotherapy.


Assuntos
Células-Tronco Hematopoéticas/citologia , Animais , Antígenos CD34/fisiologia , Técnicas de Cultura de Células/métodos , Divisão Celular , Citaferese , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/imunologia , Humanos , Depleção Linfocítica
7.
Exp Hematol ; 28(3): 335-46, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10720698

RESUMO

OBJECTIVE: The additional transplantation of ex vivo-generated megakaryocytic cells might enable the clinician to ameliorate or abrogate high-dose chemotherapy-induced thrombocytopenia. Therefore, the ex vivo expansion of CD34(+) PBPC was systematically studied aiming for an optimum production of megakaryocytic cells. MATERIALS AND METHODS: CD34(+) PBPC were cultured in serum-free medium comparing different (n = 23) combinations of stem cell factor (SCF) (S), IL-1beta (1), IL-3 (3), IL-6 (6), erythropoietin (EPO) (E), thrombopoietin (TPO) (T) and promegapoietin (PMP, a novel chimeric IL-3/TPO receptor agonist). Ex vivo-generated cells were assessed by flow cytometry, morphology, and progenitor cell assays. RESULTS: Addition of TPO to cultures stimulated with S163E, a potent progenitor cell expansion cocktail previously described by our group, effectively induced the generation of CD61(+) cells (day 12: 31.4 +/- 7.9%). The addition of PMP tended to be more effective than TPO +/- IL-3. Whereas EPO was not required to maximize TPO- or PMP-induced megakaryocytic cell production, the use of IL-6 and IL-1beta augmented cellular expansion as well as CD61(+) cell production rates in the majority of cytokine combinations studied. Thus, the most effective CD61(+) cell expansion cocktail consisted of S163 + PMP which resulted in 65.9 +/- 3.0% CD61(+) at day 12 and an overall production of 40.7 +/- 4.5 CD61(+) cells per seeded CD34(+) PBPC. However, the basic 2-factor combination S + PMP also allowed for an effective CD61(+) cell production (day 12 CD61(+) cell production: 15.1 +/- 1.6). Moreover, maximum amplification of CFU-Meg was observed after 7 days using this two-factor cocktail (12.9 +/- 2.6-fold). The majority of CD61(+) cells generated in TPO- or PMP-based medium were low-ploidy 4N and 8N cells, and ex vivo-generated CD61(+), CD41(+), and CD42b(+) cells were mainly double positive for FACS-measured intracellular von Willebrand Factor (vWF) (76.7 +/- 3.3%, 58.8 +/- 4.4%, and 82.7 +/- 2.5%, respectively). CONCLUSIONS: Taken together, this study demonstrates that megakaryocytic cells can be effectively produced ex vivo with as little as two-factors (SCF + PMP), an approach that might be favorably employed in a clinical expansion trial aiming to ameliorate high-dose chemotherapy-induced thrombocytopenia.


Assuntos
Glicoproteínas/farmacologia , Substâncias de Crescimento/farmacologia , Hematopoese , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Megacariócitos/patologia , Fator de Células-Tronco/farmacologia , Trombopoetina , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos
8.
Ann N Y Acad Sci ; 872: 25-38; discussion 38-9, 1999 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-10372108

RESUMO

The purpose of this report is to demonstrate the expression of very recently identified surface antigens on CD34+ and AC133+ bone marrow (BM) cells. Coexpression analysis of AC133 and defined antigens on CD34+ BM cells revealed that the majority of the CD164+, CD135+, CD117+, CD38low, CD33+, and CD71low cells resides in the AC133+ population. In contrast, most of the CD10+ and CD19+ B cell progenitors and a fraction of the CD71high population are AC133-, indicating that CD34+AC133+ cells are enriched in primitive and myeloid progenitor cells, whereas CD34+AC133- cells mainly consist of B cell and late erythroid progenitors. This corresponds to the highly reduced percentage of CD10+ B cells and the absence of CD71high erythroid progenitors on AC133+ selected BM cells. A portion of 0.2-0.7% of the AC133+ selected cells do not coexpress CD34. These cells are very small and define a uniform CD71-, CD117-, CD10-, CD38low, CD135+, HLA-DRhigh, CD45+ population with unknown delineation. Four color analysis on CD34+CD38- BM cells revealed that virtually all of these primitive cells express AC133. Using an improved liposome-enhanced labeling technique for the staining of weakly expressed antigens, subsets of this population could be identified which express the angiopoietin receptors TIE (67.6%) and TEK (36.8%), the vascular endothelial growth factor receptors FLT1 (7%), FLT4 (3.2%), and KDR (10.4%), or the receptor tyrosine kinases HER-2 (15.4%) and FLT3 (CD135; 77.6%). Our results suggest that the CD34+CD38- population is heterogeneous with respect to the expression of the analyzed receptor tyrosine kinases.


Assuntos
Antígenos CD34/análise , Antígenos CD/análise , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Glicoproteínas/análise , Células-Tronco Hematopoéticas/imunologia , Peptídeos/análise , Antígeno AC133 , ADP-Ribosil Ciclase , ADP-Ribosil Ciclase 1 , Antígenos de Diferenciação/análise , Linfócitos B/citologia , Células da Medula Óssea/imunologia , Células Cultivadas , Citometria de Fluxo , Antígenos HLA-DR/análise , Células-Tronco Hematopoéticas/citologia , Humanos , Separação Imunomagnética/métodos , Glicoproteínas de Membrana , NAD+ Nucleosidase/análise , Receptores Proteína Tirosina Quinases/análise
9.
Ann N Y Acad Sci ; 872: 200-7; discussion 207-10, 1999 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-10372123

RESUMO

Research on the biology, regulation, and transplantation of human hematopoietic stem cells requires test systems for the detection, monitoring, and quantitation of these cells. Xenografted animal models provide suitable stem cell assays, since they allow long-term engraftment, multilineage differentiation, and serial transfer of human hematopoietic cells. Recent techniques for the separation of hematopoietic cells have provided highly purified cellular subsets selected on the basis of the surface marker phenotype. The stem cell content of these subsets, however, is still unclear. Also, innovative approaches for the induction of hematopoietic cell proliferation and differentiation have generated ex vivo manipulated cells whose biological properties and functions still remain to be assessed. This paper reports on the biological characterization of these cell populations by the use of xenograft models.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Transplante Heterólogo , Animais , Antígenos CD/análise , Antígenos CD34/análise , Biomarcadores , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID
10.
Ann N Y Acad Sci ; 872: 363-71, 1999 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-10372138

RESUMO

High-dose chemotherapy with peripheral blood progenitor cell transplantation (PBPCT) is a potentially curative treatment option for patients with both hematological malignancies and solid tumors, including breast cancer. However, based on a number of clinical studies, there is strong evidence that minimal residual disease (MRD) persists after high-dose chemotherapy in a number of patients, which eventually results in disease recurrence. Therefore, several approaches to the treatment of MRD are currently being evaluated, including treatment with dendritic cell (DC)-based cancer vaccines. DCs, which play a crucial role with regard to the initiation of T-lymphocyte responses, can be generated ex vivo either from CD34+ hematopoietic progenitor cells or from blood monocytes. They can be pulsed in vitro with tumor-derived peptides or proteins, and then used as a professional antigen-presenting cell (APC) vaccine for the induction of antigen-specific T-lymphocytes in vivo. This paper summarizes our preclinical studies on the induction of primary HER-2/neu specific cytotoxic T-lymphocyte (CTL) responses using peptide-pulsed DC. As HER-2/neu is overexpressed on 30-40% of breast and ovarian cancer cells, this novel vaccination approach might be particularly applicable to advanced breast or ovarian cancer patients after high-dose chemotherapy and autologous PBPCT.


Assuntos
Células Dendríticas/imunologia , Transplante de Células-Tronco Hematopoéticas , Imunoterapia , Neoplasia Residual/terapia , Neoplasias/terapia , Antígenos CD/análise , Antígenos CD34/análise , Vacinas Anticâncer , Células Cultivadas , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/terapia , Humanos , Neoplasia Residual/imunologia , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia
11.
J Clin Oncol ; 17(5): 1535-44, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10334541

RESUMO

PURPOSE: The expression of the carcinoma-associated mucin MUC-1 is thought to be restricted to epithelial cells and is used for micrometastatic tumor cell detection in patients with solid tumors, including those with breast cancer. Little is known, however, about the expression of MUC-1 epitopes in normal hematopoietic cells. MATERIALS AND METHODS: MUC-1 expression was analyzed by flow cytometry and immunocytology on bone marrow (BM) mononuclear cells and purified CD34+ cells from healthy volunteers, using different anti-MUC-1-specific monoclonal antibodies. In addition, Western blotting of MUC-1 proteins was performed. RESULTS: Surprisingly, 2% to 10% of normal human BM mononuclear cells expressed MUC-1, as defined by the anti-MUC-1 antibodies BM-2 (2E11), BM-7, 12H12, MAM-6, and HMFG-1. In contrast, two antibodies recognizing the BM-8 and the HMFG-2 epitopes of MUC-1 were not detected. MUC-1+ cells from normal BM consisted primarily of erythroblasts and normoblasts. In agreement with this, normal CD34+ cells cultured in vitro to differentiate into the erythroid lineage showed a strong MUC-1 expression on day 7 proerythroblasts. Western blotting of these cells confirmed that the reactive species is the known high molecular weight MUC-1 protein. CONCLUSION: Our data demonstrate that some MUC-1 epitopes are expressed on normal BM cells and particularly on cells of the erythroid lineage. Hence the application of anti-MUC-1 antibodies for disseminated tumor cell detection in BM or peripheral blood progenitor cells may provide false-positive results, and only carefully evaluated anti-MUC-1 antibodies (eg, HMFG-2) might be selected. Furthermore, MUC-1-targeted immunotherapy in cancer patients might be hampered by the suppression of erythropoiesis.


Assuntos
Medula Óssea/imunologia , Epitopos/análise , Células-Tronco Hematopoéticas/imunologia , Mucina-1/imunologia , Anticorpos Monoclonais , Citometria de Fluxo , Humanos
12.
Exp Hematol ; 27(5): 956-65, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10340412

RESUMO

Although hematopoietic recovery following high-dose chemotherapy (HD-CT) and peripheral blood progenitor cell (PBPC) transplantation is rapid, there is still a 5- to 7-day period of severe neutropenia which, theoretically, might be abrogated by an additional transplantation of more differentiated myeloid post-progenitor cells (MPPC). However, both the number of MPPC required to abrogate neutropenia as well as the optimum scheduling of MPPC infusions are currently unknown. Therefore, these questions were addressed by applying a computer model of human granulopoiesis. First, model calculations simulating varying levels of chemotherapy dose intensity were performed and compared with typical clinical neutrophil recovery curves. Using this approach, the data for HD-CT without PBPC transplantation could be reproduced by assuming a reduction of stem cells, committed granulopoietic progenitors and proliferating precursors to about 0.001% of normal. PBPC-supported HD-CT was reproduced by increasing the starting values to at least 0.1%, which corresponded to about 1 to 2 x 10(5)/kg transplanted CFU-GM. Interestingly, reproduction of PBPC-supported HD-CT data could be observed for a wide range of starting values (0.1%-10% of normal), thus confirming the clinical observation that hematopoietic recovery after PBPCT cannot be improved by increasing the dose of transplanted cells over a certain threshold. Using the same simulation model, we then studied the effects of an additional MPPC transplantation. The results showed, that at least 5.7 X 10(8) MPPC/kg have to be provided in addition to the normal PBPC graft to avoid neutropenia <100/microL, and that MPPC are best transplanted on days 0 and 6 after HD-CT. Assuming a 100- to 120-fold cellular ex-vivo expansion rate and MPPC representing about 70% of total expanded cells, 5.7 X 10(8) MPPC/kg could be generated starting from 1 to 2 leukapheresis preparations with about 7 to 8 x 10(6) CD34+ PBPC/kg. Considering furthermore, that only a fraction of ex-vivo generated cells will seed and effectively produce neutrophils in-vivo, the required number of MPPC is most likely even higher and, therefore, might be difficult to be achieved clinically. However, the validity of the model results remains to be proven in appropriate clinical studies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Neutropenia/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Terapia Combinada , Simulação por Computador , Relação Dose-Resposta a Droga , Humanos , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/terapia , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Neutropenia/etiologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-11000987

RESUMO

The increasing use of haematopoietic stem and progenitor cells from the peripheral blood (PBPC) to restore haematopoiesis following high-dose chemotherapy has widely propagated the development of techniques for the ex vivo manipulation of haematopoietic cells. In particular, protocols for the ex vivo expansion of PBPC have been developed for different clinical purposes. Quantitative expansion of PBPC may provide a successful strategy for tumour cell purging of autologous grafts, or may generate sufficient cell numbers for sequential transplantation protocols. Furthermore, allogeneic transplantation of megadoses of PBPC may enable us to overcome immunological barriers, and may substantially increase the number of suitable donors for an individual patient. Clinical applications also include the use of ex vivo generated, partially differentiated, post-progenitor cells, antigen presenting cells for immunotherapy of minimal residual disease, and ex vivo transduced haematopoietic cells as attractive vehicles for genetic therapy.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Técnicas de Cultura de Células , Protocolos Clínicos , Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/normas , Células-Tronco Hematopoéticas/imunologia , Humanos
14.
Semin Hematol ; 35(3): 232-40, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9685169

RESUMO

The development of efficient stem and progenitor cell selection methods in combination with the development of hematopoietic growth factors facilitated the development of ex vivo expansion techniques. Currently, this novel domain of cellular therapy aims to generate stem and progenitor cells, as well as more differentiated post-progenitor cells and antigen-presenting dendritic cells. The feasibility of generating and transplanting hematopoietic progenitor cells ex vivo (using various cytokine combinations) has been successfully shown preclinically as well as clinically. Furthermore, cytokines (eg, Flt-3-ligand; thrombopoietin) have been identified that play important roles with regard to amplification of undifferentiated early hematopoietic cells. The use of lineage-specific cytokines such as granulocyte colony-stimulating factor and thrombopoietin facilitated the generation of large numbers of myeloid and megakaryocytic post-progenitor cells. The clinical usefulness of such ex vivo generated cells, however, has not yet been convincingly shown. Last, ex vivo expansion techniques can be used to generate large numbers of antigen-presenting dendritic cells from CD34+ peripheral blood progenitor cells that might be ideally used for immunotherapeutic approaches.


Assuntos
Células-Tronco Hematopoéticas/citologia , Contagem de Células , Fatores de Crescimento de Células Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Humanos , Megacariócitos/citologia , Trombopoetina
15.
Stem Cells ; 16(2): 144-51, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9554039

RESUMO

Recombinant human granulocyte colony-stimulating factor, rhG-CSF, is widely applied to ameliorate neutropenia following chemotherapy. However, rhG-CSF can exert negative effects on megakaryocytopoiesis that might cause a delay of megakaryocyte recovery. Therefore, the present study was designed to test different rhG-CSF administration protocols with regard to their megakaryocytic inhibitory potential in a 5-fluorouracil (5-FU)-induced experimental model system. Splenectomized B6D2F1 mice received a single injection of 5-FU (150 mg/kg) on day 0 followed by 50 micrograms/kg/day rhG-CSF given daily for either zero, four, or eight days. Five days after 5-FU, bone marrow and blood hematopoiesis were reduced significantly when compared with controls, independent of whether or not animals received rhG-CSF. However, nine days after 5-FU, granulopoietic recovery from 5-FU-induced toxicity was faster for rhG-CSF-treated versus untreated mice as demonstrated by higher values for colony forming unit-granulocyte macrophage (CFU-GM) and granulocytes (CFU-GM: 7.2 +/- 0.4 versus 5 +/- 0.6 x 10(4)/femur, granulocytes: 4.3 +/- 2 versus 1.4 +/- 0.4 x 10(5)/ml, respectively). Furthermore, significant mobilization of CFU-megakaryocyte (CFU-Meg) and CFU-GM into the peripheral blood was induced by the eight-day administration of rhG-CSF following 5-FU (day 9: 911 +/- 102 CFU-Meg/ml, 2330 +/- 152 CFU-GM/ml). However, megakaryocytic cells in these same mice were considerably lower when compared with those of animals receiving no rhG-CSF (CFU-Meg: 2.7 +/- 0.2 x 10(3) versus 4.2 +/- 0.2 x 10(3)/femur; small acetylcholinesterase positive (SAChE+) cells: 4.9 +/- 0.3 x 10(3) versus 7.3 +/- 0.9 x 10(3)/femur; megakaryocytes: 2.5 +/- 0.2 x 10(3) versus 4.1 +/- 0.7 x 10(3)/femur; platelets: 2.67 +/- 0.5 x 10(9) versus 3.1 +/- 0.5 x 10(9)/ml, respectively). On the other hand, the shortening of the rhG-CSF treatment from eight to four days caused a rapid granulopoietic recovery comparable to animals receiving eight days of G-CSF with no significant delay in megakaryocytic recovery when compared with mice treated with 5-FU alone; however, with four days of rhG-CSF, the mobilization of CFU into the peripheral blood was significantly less effective. Taken together, the results showed that a shortening of rhG-CSF treatment after chemotherapy is capable of ameliorating neutropenia without negatively affecting megakaryocytopoietic recovery. If, however, maximum recruitment of CFU into the peripheral blood circulation by rhG-CSF for subsequent harvest and transplantation is needed, any shortening of rhG-CSF administration is not advisable.


Assuntos
Fluoruracila/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Imunossupressores/administração & dosagem , Megacariócitos/efeitos dos fármacos , Animais , Diferenciação Celular , Feminino , Granulócitos , Hematopoese , Humanos , Masculino , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas Recombinantes , Esplenectomia
16.
Stem Cells ; 16 Suppl 1: 199-204, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-11012163

RESUMO

Approaches to manipulate peripheral blood progenitor cells (PBPC) ex vivo currently include the selection of CD34+ cells as a means to purge contaminating tumor cells from leukapheresis preparations or to provide a homogeneous starting population for the expansion of hematopoietic progenitor cells as well as the induction of postprogenitor cells of either the myeloid or megakaryocytic lineage. The latter cell populations might be used for an additional transplantation together with PBPC to possibly shorten the period of aplasia. In addition, ex vivo expansion of CD34+ cells can be used to generate autologous tumor-antigen-presenting dendritic cells for immunotherapeutic approaches aiming to treat minimal residual disease following high-dose chemotherapy.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Neoplasias/terapia , Antígenos CD34/análise , Células da Medula Óssea/citologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Humanos , Imunoterapia , Leucaférese , Megacariócitos/citologia
17.
Leukemia ; 11(11): 1842-9, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9369416

RESUMO

A gene encoding the p53 val135 mutant, which assumes mutant conformation at 38.5 degrees C and wild-type conformation at 32.5 degrees C, was introduced into p53-deficient K562 myeloid leukemia cells. Forced expression of wild-type, but not mutant, p53 resulted in growth arrest, accumulation of p21 and Bax proteins, and delayed cell death. Wild-type p53 enhanced the cytotoxic effects of some drugs and attenuated those of others. Compared with wild-type p53, mutant p53 induced much stronger sensitization to drug cytotoxicity. This occurred in the absence of effects on cell cycle progression or activation of several p53 target genes. Although both mutant and wild-type p53 induced changes of immunophenotype, no specific pattern of differentiation was associated with enhanced chemosensitivity. Thus, (1) induction of growth arrest and activation of p53 target genes such as p21 and bax are linked to the wild-type conformation of p53; (2) p53 induces immunophenotypic changes of myeloid leukemia cells suggestive of multidirectional differentiation in a conformation-dependent manner; and (3) (so-called) mutant p53 induces chemosensitization in the absence of effects on cell cycle progression, activation of bax, p21, gadd45 and mdm-2, or a specific pattern of differentiation; and (4) chemosensitization mediated by wild-type p53 may be masked by transcription-dependent induction of growth arrest.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Animais , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Leucemia Mieloide/patologia , Leucemia Mieloide/fisiopatologia , Camundongos , Microscopia Eletrônica , Mutação , Temperatura , Transfecção , Células Tumorais Cultivadas/ultraestrutura , Proteína Supressora de Tumor p53/genética
18.
Stem Cells ; 15 Suppl 1: 159-65, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9368336

RESUMO

Clinical success of autologous peripheral blood progenitor cell (PBPC) transplantation is challenged by relapse of malignant disease which might at least in part be mediated by graft-contaminating tumor cells. Although the clinical efficacy of tumor cell depletion still remains to be demonstrated, multiple purging strategies are currently pursued in the context of autologous stem cell transplantation. This report discusses ex vivo manipulations of PBPC transplants with respect to purging of tumor cells, including positive selection of CD34+ cells with or without negative depletion of tumor cells as well as ex vivo expansion techniques. Moreover, strategies with an adoptive immunotherapy using ex vivo-generated autologous dendritic cells for the treatment of minimal residual disease after stem cell transplantation will be discussed here.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Neoplasia Residual/cirurgia , Antígenos CD34/análise , Separação Celular , Ensaios Clínicos como Assunto , Células Dendríticas/imunologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Imunoterapia Adotiva , Neoplasia Residual/terapia , Neoplasias Epiteliais e Glandulares/cirurgia , Transplante Autólogo
19.
Blood ; 88(7): 2707-13, 1996 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-8839866

RESUMO

Cytokine-supported ex vivo expansion of peripheral blood progenitor cells (PBPCs) offers new perspectives for autografting after high-dose chemotherapy. One of the potential advantages is the possibility to reduce the volume of blood processed from the patient, thus allowing reduction of the overall tumor cell number in the final autograft. However, ex vivo expansion will only be advantageous if contaminating tumor cells are not expanded concomitantly. This question has not previously been addressed. Therefore, we analyzed unseparated PBPC preparations, CD34(+)-selected cell fractions, and ex vivo-expanded cell preparations from stage IV (n = 16) and high-risk stage II/III (n = 8) breast cancer patients for the presence of human epithelial antigen- (HEA) or cytokeratin (CK)-positive tumor cells. We found that three of 16 (18.8%) of the unseparated PBPC products from stage IV patients were HEA- and/or CK-positive, whereas none of the stage II/III patients were found to be positive after two cycles of induction chemotherapy with etoposide (VP16), ifosfamide, cisplatin, and epirubicin (VIP-E). After CD34+ cell selection (Ceprate SC; CellPro, Bothell, WA) and stem-cell factor (SCF), interleukin (IL)-1, IL-3, IL-6, and erythropoietin (EPO)-mediated ex vivo expansion of the CD34+ cells for 14 to 21 days, no tumor cells could be detected in these primary breast cancer patients at a sensitivity of 1 tumor cell per 4 x 10(5) nucleated cells. Thus, to answer the question of whether tumor cells are expanded concomitantly on ex vivo expansion of normal CD34+ cells, we cocultured defined numbers of primary renal carcinoma cells (RS-85), xenograft-derived breast cancer cells, and small-cell lung cancer cells with CD34+ cells selected from normal donors or cancer patients, either in serum or serum-free culture media. We found that none of the three epithelial tumor cell types increased significantly in number during a 14-day coculture period when compared with normal CD34+ cells alone or tumor cells alone, which increased 110- +/- 77-fold and 45- +/- 26-fold, respectively. However, during coculture, the tumor cells did not undergo cell death and were able to regrow when maintained in serum for longer time periods. We conclude that cytokine-supported expansion cultures of positively selected CD34+ PBPCs from primary high-risk stage II/III or stage IV breast cancer patients do not contain detectable tumor cells, which suggests that there is no increased risk of concomitantly expanding tumor cells. Moreover, cocultures of exogenously mixed tumor cell lines with normal CD34+ cells showed a relative disadvantage of tumor cell growth compared with the growth of hematopoietic cells, again without an apparent risk of concomitantly expanding tumor cells. However, considering the pronounced heterogeneity of tumor cell kinetics, ex vivo-expanded PBPC from cancer patients should be monitored for minimal residual disease.


Assuntos
Antígenos CD34/análise , Células Sanguíneas/citologia , Neoplasias da Mama/sangue , Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Células Neoplásicas Circulantes/patologia , Animais , Biomarcadores Tumorais/análise , Fenômenos Fisiológicos Sanguíneos , Neoplasias da Mama/patologia , Carcinoma de Células Renais/patologia , Bovinos , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura Livres de Soro , Feminino , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Queratinas/análise , Neoplasias Renais/patologia , Mucina-1/análise , Proteínas de Neoplasias/análise , Estadiamento de Neoplasias , Neoplasia Residual , Células Neoplásicas Circulantes/efeitos dos fármacos , Risco , Sensibilidade e Especificidade , Fatores de Tempo , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA