Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biol Blood Marrow Transplant ; 19(6): 904-11, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23416854

RESUMO

Manifestations of and risk factors for graft-versus-host disease (GVHD) after double-unit cord blood transplantation (DCBT) are not firmly established. We evaluated 115 DCBT recipients (median age, 37 years) who underwent transplantation for hematologic malignancies with myeloablative or nonmyeloablative conditioning and calcineurin inhibitor/mycophenolate mofetil immunosuppression. Incidence of day 180 grades II to IV and III to IV acute GVHD (aGVHD) were 53% (95% confidence interval, 44 to 62) and 23% (95% confidence interval, 15 to 31), respectively, with a median onset of 40 days (range, 14 to 169). Eighty percent of patients with grades II to IV aGVHD had gut involvement, and 79% and 85% had day 28 treatment responses to systemic corticosteroids or budesonide, respectively. Of 89 engrafted patients cancer-free at day 100, 54% subsequently had active GVHD, with 79% of those affected having persistent or recurrent aGVHD or overlap syndrome. Late GVHD in the form of classic chronic GVHD was uncommon. Notably, grades III to IV aGVHD incidence was lower if the engrafting unit human leukocyte antigen (HLA)-A, -B, -DRB1 allele match was >4/6 to the recipient (hazard ratio, 0.385; P = .031), whereas engrafting unit infused nucleated cell dose and unit-to-unit HLA match were not significant. GVHD after DCBT was common in our study, predominantly affected the gut, and had a high therapy response, and late GVHD frequently had acute features. Our findings support the consideration of HLA- A,-B,-DRB1 allele donor-recipient (but not unit-unit) HLA match in unit selection, a practice change in the field. Moreover, new prophylaxis strategies that target the gastrointestinal tract are needed.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Trato Gastrointestinal/imunologia , Doença Enxerto-Hospedeiro/terapia , Antígenos HLA/imunologia , Neoplasias Hematológicas/terapia , Agonistas Mieloablativos/uso terapêutico , Condicionamento Pré-Transplante , Adolescente , Corticosteroides/uso terapêutico , Adulto , Idoso , Budesonida/uso terapêutico , Calcineurina/metabolismo , Inibidores de Calcineurina , Criança , Pré-Escolar , Inibidores Enzimáticos/uso terapêutico , Feminino , Trato Gastrointestinal/patologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/mortalidade , Doença Enxerto-Hospedeiro/patologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/mortalidade , Neoplasias Hematológicas/patologia , Teste de Histocompatibilidade , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapêutico , Índice de Gravidade de Doença , Análise de Sobrevida , Transplante Homólogo , Resultado do Tratamento
2.
J Exp Med ; 171(1): 197-210, 1990 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-2295876

RESUMO

We recently described a mAb 3.2.3 (IgG1), that recognizes a 60-kD dimeric molecule expressed exclusively on fresh and rIL-2-activated NK cells and polymorphonuclear cells. mAb 3.2.3 enhances cytolytic activity of NK cells against selected FcR+ tumor target cells by reverse antibody-dependent cellular cytotoxicity (ADCC), indicating that it recognizes an important triggering site on NK cells. The in vivo treatment of F344 rats with mAb 3.2.3 intraperitoneally completely and selectively eliminated NK/ADCC function in the spleen and peripheral blood for up to 10 d after treatment. Total numbers and percentages of T cells, monocytes, or PMN were not decreased and T cell function, as determined by Con A stimulation, was not affected. The reduction in NK function was associated with a decrease in the numbers of LGL and the expression of other NK-related cell surface markers including CD2, CD8, and asialo GM1. Depletion of NK cells with 3.2.3 markedly decreased the survival of F344 rats injected intravenously with MADB106 mammary adenocarcinoma cells, but did not affect the subcutaneous growth of MADB106 tumors. These results indicate that mAb 3.2.3 (in contrast to anti-asialo GM1 and OX8, which are less selective markers) will be useful for studies on the functional role of NK cells in vivo as well as their in vivo differentiation and origin from 3.2.3- precursors.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Citotoxicidade Celular Dependente de Anticorpos , Linhagem Celular , Concanavalina A , Citometria de Fluxo , Injeções Intraperitoneais , Células Matadoras Naturais/citologia , Ativação Linfocitária , Masculino , Ratos , Ratos Endogâmicos F344 , Células Tumorais Cultivadas/imunologia
3.
J Exp Med ; 172(1): 303-13, 1990 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-2358779

RESUMO

In this report, we describe a novel long-term bone marrow culture (LTBMC) system to study the origin and generation of natural killer (NK) cells from NK precursors. Rat bone marrow was cultured for 4 wk in RPMI 1640 with 5% fetal calf serum and 2-mercaptoethanol to allow the formation of an adherent stromal cell layer containing NK precursor cells. After addition of interleukin 2 (IL-2), the LTBMC generated high numbers (up to 100-fold expansion in 7 d) of pure 3.2.3+ large granular lymphocytes with lytic activity against NK-sensitive and -resistant tumor targets, as well as antibody-dependent cellular cytotoxicity. NK activity in LTBMC could be detected 3 d after addition of as little as 1 U/ml rIL-2, whereas lymphokine-activated killer activity was found 5 d after addition of at least 10 U/ml rIL-2. In vivo depletion and in vitro complement lysis studies showed that the NK precursor cells in LTBMC did not express the NK-associated surface markers asialo GM1 or 3.2.3. We also found that LTBMC cells did not exhibit colony growth in granulocyte/macrophage or spleen colony-forming unit assays. The generation of NK cells from NK precursors required, in addition to IL-2, a second growth/maturation factor(s), which was present in the conditioned medium of the LTBMC. This LTBMC system provides a unique in vitro model to study the development of NK cells from precursor cells, the role of the bone marrow stromal microenvironment in this development, and the lineage relationship of NK cells to other hematopoietic cells.


Assuntos
Células da Medula Óssea , Células-Tronco Hematopoéticas/citologia , Células Matadoras Naturais/citologia , Animais , Anticorpos Monoclonais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Interleucina-2/farmacologia , Células Matadoras Naturais/imunologia , Masculino , Camundongos , Camundongos Mutantes , Fenótipo , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes , Células Tumorais Cultivadas
4.
J Exp Med ; 174(2): 467-78, 1991 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-1856629

RESUMO

Transplantation of untreated F344 rat bone marrow into irradiated B10 mouse recipients (non-TCD F344----B10) to produce fully xenogeneic chimeras resulted in stable xenogeneic lymphoid chimerism, ranging from 82% to 97% rat. Survival of animals was excellent, without evidence for GVH disease. The specificity of tolerance which resulted was highly donor-specific; MHC disparate third party mouse and rat skin grafts were promptly rejected while donor-specific F344 grafts were significantly prolonged (MST greater than 130 days). Multi-lineage rat stem cell-derived progeny including lymphoid cells (T- and B-lymphocytes), myeloid cells, erythrocytes, platelets, and natural killer (NK) cells were present in the fully xenogenic chimeras up to 7 months after bone marrow transplantation. Immature rat T-lymphocytes matured and acquired the alpha/beta T-cell receptor in the thymus of chimeras in a pattern similar to normal rat controls, suggesting that immature T-lymphocytes of rat origin could interact with the murine xenogeneic thymic stroma to undergo normal maturation and differentiation. This model may be useful to study the mechanisms responsible for the induction and maintenance of donor-specific transplantation tolerance across a species barrier.


Assuntos
Transplante de Medula Óssea/imunologia , Tolerância Imunológica , Transplante de Células-Tronco , Linfócitos T/imunologia , Transplante Heterólogo/imunologia , Animais , Plaquetas/imunologia , Medula Óssea/imunologia , Medula Óssea/efeitos da radiação , Quimera/imunologia , Citometria de Fluxo , Imunofenotipagem , Tecido Linfoide/imunologia , Masculino , Camundongos , Camundongos Endogâmicos , Ratos , Ratos Endogâmicos , Transplante de Pele/imunologia , Células-Tronco/imunologia , Linfócitos T/citologia
5.
J Mol Med (Berl) ; 85(8): 837-43, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17333096

RESUMO

Strategies to enhance post-transplant immune reconstitution without aggravating graft-vs-host disease (GVHD) can improve the outcome of allogeneic hematopoietic stem cell transplantation. Recent preclinical studies demonstrated that the use of T cell depleted allografts supplemented with committed progenitor cells (vs stem cells only) allows enhanced immune reconstitution of specific hematopoietic lineages including myeloid, B, T, and natural killer lineages in the absence of GVHD. This novel adoptive therapy resulted in significantly improved resistance to microbial pathogens and could, in some cases, even mediate tumor immunity. Clinical protocols using adoptive transfer of committed hematopoietic progenitor cells are currently being evaluated.


Assuntos
Transferência Adotiva/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Imunidade Celular/imunologia , Humanos , Modelos Biológicos , Imunologia de Transplantes
6.
J Clin Invest ; 104(3): 317-25, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10430613

RESUMO

We recently showed that IL-11 prevents lethal graft-versus-host disease (GVHD) in a murine bone marrow transplantation (BMT) model of GVHD directed against MHC and minor antigens. In this study, we have investigated whether IL-11 can maintain a graft-versus-leukemia (GVL) effect. Lethally irradiated B6D2F1 mice were transplanted with either T cell-depleted (TCD) bone marrow (BM) alone or with BM and splenic T cells from allogeneic B6 donors. Animals also received host-type P815 mastocytoma cells at the time of BMT. Recipients were injected subcutaneously with recombinant human IL-11 or control diluent twice daily, from 2 days before BMT to 7 days after BMT. TCD recipients all died from leukemia by day 23. All control- and IL-11-treated allogeneic animals effectively rejected their leukemia, but IL-11 also reduced GVHD-related mortality. Examination of the cellular mechanisms of GVL and GVHD in this system showed that IL-11 selectively inhibited CD4-mediated GVHD, while retaining both CD4- and CD8-mediated GVL. In addition, IL-11 treatment did not affect cytolytic effector functions of T cells after BMT either in vivo or in vitro. Studies with perforin-deficient donor T cells demonstrated that the GVL effect was perforin dependent. These data demonstrated that IL-11 can significantly reduce CD4-dependent GVHD without impairing cytolytic function or subsequent GVL activity of CD8(+) T cells. Brief treatment with IL-11 shortly after BMT may therefore represent a novel strategy for separating GVHD and GVL.


Assuntos
Transplante de Medula Óssea/imunologia , Doença Enxerto-Hospedeiro/imunologia , Efeito Enxerto vs Tumor/imunologia , Interleucina-11/fisiologia , Animais , Transplante de Medula Óssea/efeitos adversos , Antígenos CD4/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Intervalo Livre de Doença , Feminino , Doença Enxerto-Hospedeiro/prevenção & controle , Doença Enxerto-Hospedeiro/terapia , Humanos , Imunossupressores/uso terapêutico , Interleucina-11/uso terapêutico , Leucemia Experimental/imunologia , Leucemia Experimental/terapia , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Perforina , Proteínas Citotóxicas Formadoras de Poros , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas , Receptor fas/fisiologia
7.
Bone Marrow Transplant ; 52(12): 1629-1636, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28991247

RESUMO

CD34+ cell selection significantly improves GvHD-free survival in allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, specific information regarding long-term prognosis and risk factors for late mortality after CD34+ cell-selected allo-HSCT is lacking. We conducted a single-center landmark analysis in 276 patients alive without relapse 1 year after CD34+ cell-selected allo-HSCT for AML (n=164), ALL (n=33) or myelodysplastic syndrome (n=79). At 5 years' follow-up after the 1-year landmark (range 0.03-13 years), estimated relapse-free survival (RFS) was 73% and overall survival (OS) 76%. The 5-year cumulative incidence of relapse and non-relapse mortality (NRM) were 11% and 16%, respectively. In multivariate analysis, Hematopoietic Cell Transplantation Comorbidity Index score⩾3 correlated with marginally worse RFS (hazard ratio (HR) 1.78, 95% confidence interval (CI) 0.97-3.28, P=0.06) and significantly worse OS (HR 2.53, 95% CI 1.26-5.08, P=0.004). Despite only 24% of patients with acute GvHD within 1 year, this also significantly correlated with worse RFS and OS, with increasing grades of acute GvHD associating with increasingly poorer survival on multivariate analysis (P<0.0001). Of 63 deaths after the landmark, GvHD accounted for 27% of deaths and was the most common cause of late mortality, followed by relapse and infection. Although prognosis is excellent for patients alive without relapse 1 year after CD34+ cell-selected allo-HSCT, risks of late relapse and NRM persist, particularly due to GvHD.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicas/terapia , Adolescente , Adulto , Idoso , Antígenos CD34 , Comorbidade , Feminino , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/mortalidade , Transplante de Células-Tronco Hematopoéticas/mortalidade , Humanos , Leucemia Mieloide Aguda/mortalidade , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/mortalidade , Prognóstico , Fatores de Risco , Análise de Sobrevida , Sobreviventes , Transplante Homólogo , Adulto Jovem
8.
Cancer Res ; 51(18): 4931-6, 1991 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-1893382

RESUMO

A monoclonal antibody, designated 3.2.3, which recognizes a novel Mr 60,000 disulfide-linked lytic triggering structure present on rat large granular lymphocytes and natural killer (NK) cells was recently described (W. H. Chambers et al., J. Exp. Med., 169: 1373-1389, 1989). The present study describes the use of 3.2.3 to identify the in situ tissue distribution of large granular lymphocytes/NK cells in different organs from normal and tumor-bearing F344 rats. Frozen tissue sections were prepared and stained with monoclonal antibody 3.2.3 using an avidinbiotin immunoperoxidase technique. 3.2.3+ NK cells were easily identified using this technique, and quantitative analysis of various tissues of normal rats demonstrated that (a) in the spleen, most NK cells were located, sometimes as aggregates, in the red pulp (12.4% of total nucleated cells in that organ compartment) with relatively few noted in the white pulp (0.2-2.3%); (b) in the liver, 3.2.3+ cells were rare, sparsely distributed, and located primarily in the sinusoids (1.2%); (c) in the lungs, 3.2.3+ cells were located in the interstitium (3.7%); (d) in the thymus, 3.2.3+ cells were found primarily in the medulla (1.8%) adjacent to the cortex but not in the cortex itself (0.2%); (e) in the lymph node, most 3.2.3+ cells were contained in the paracortex (6.9%); and finally (f) in the small bowel, 3.2.3+ cells were present in the lamina propria (8.6%) and as aggregates in the interfollicular zone of Peyer's patches (0.7%). To study the distribution of 3.2.3+ NK cells in developing tumor metastases, we induced liver metastases by intrasplenic injection of MADB106 mammary adenocarcinoma cells and prepared frozen tissue sections of the liver 10-14 days later. We found that the frequency of 3.2.3+ cells in the developing liver metastases was 3-6 times higher than in the surrounding normal liver tissue. Moreover, the frequency of 3.2.3+ NK cells was equivalent to the frequency of tumor-infiltrating CD5+ T-cells identified in the same tumor lesions. This suggests specific infiltration of 3.2.3+ NK cells in early developing metastatic lesions. These results indicate that monoclonal antibody 3.2.3 will be valuable in analyzing the involvement of NK cells in various pathological states.


Assuntos
Anticorpos Monoclonais/imunologia , Células Matadoras Naturais/citologia , Adenocarcinoma/patologia , Animais , Anticorpos Monoclonais/metabolismo , Técnicas Imunoenzimáticas , Células Matadoras Naturais/imunologia , Neoplasias Hepáticas Experimentais/imunologia , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/secundário , Masculino , Neoplasias Mamárias Experimentais/patologia , Transplante de Neoplasias , Ratos , Ratos Endogâmicos F344 , Distribuição Tecidual
9.
Leukemia ; 30(5): 1116-25, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26867669

RESUMO

Waldenström's macroglobulinemia (WM) is a B-cell non-Hodgkin's lymphoma (B-NHL) characterized by immunoglobulin M (IgM) monoclonal gammopathy and the medullary expansion of clonal lymphoplasmacytic cells. Neoplastic transformation has been partially attributed to hyperactive MYD88 signaling, secondary to the MYD88 L265P mutation, occurring in the majority of WM patients. Nevertheless, the presence of chronic active B-cell receptor (BCR) signaling, a feature of multiple IgM+ B-NHL, remains a subject of speculation in WM. Here, we interrogated the BCR signaling capacity of primary WM cells by utilizing multiparametric phosphoflow cytometry and found heightened basal phosphorylation of BCR-related signaling proteins, and augmented phosphoresponses on surface IgM (sIgM) crosslinking, compared with normal B cells. In support of those findings we observed high sIgM expression and loss of phosphatase activity in WM cells, which could both lead to signaling potentiation in clonal cells. Finally, led by the high-signaling heterogeneity among WM samples, we generated patient-specific phosphosignatures, which subclassified patients into a 'high' and a 'healthy-like' signaling group, with the second corresponding to patients with a more indolent clinical phenotype. These findings support the presence of chronic active BCR signaling in WM while providing a link between differential BCR signaling utilization and distinct clinical WM subgroups.


Assuntos
Linfócitos B/patologia , Receptores de Antígenos de Linfócitos B/fisiologia , Transdução de Sinais , Macroglobulinemia de Waldenstrom/patologia , Células Clonais/patologia , Feminino , Humanos , Imunoglobulina M/metabolismo , Masculino , Fosforilação
10.
Clin Cancer Res ; 6(6): 2201-8, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10873069

RESUMO

In an effort to develop a biochemotherapy regimen for metastatic melanoma suitable for testing in a cooperative group setting, we modified the concurrent biochemotherapy regimen of S. S. Legha et al. (J. Clin. Oncol., 16: 1752-1759, 1998) by providing enhanced supportive care and developing a strict, conservative approach to the management of treatment-related toxicities. Patients received cisplatin, vinblastine, and dacarbazine (CVD: cisplatin (20 mg/m2) and vinblastine (1.2 mg/m2) on days 1-4, dacarbazine (800 mg/m2) on day 1 only) concurrently with interleukin 2 (9 MIU/m2/day) by continuous i.v. infusion on days 1-4 and IFN-alpha (5 MU/m2/day) on days 1-5, 8, 10, and 12. Prophylactic antibiotics and a maximum of four cycles were administered. Routine granulocyte colony-stimulating factor and aggressive antiemetics were initiated after patients 7 and 14, respectively. Forty-four patients were enrolled in this study. No patients had received prior chemotherapy or interleukin 2; however, 23 (53%) had received prior IFN-alpha, mostly in the adjuvant setting. A total of 131 treatment cycles was administered. Significant toxicities requiring dose modification included: hypotension requiring pressors (15 episodes in 11 patients), grades 3/4 vomiting (12 episodes in 15 cycles; 5 episodes in 12 patients (6 episodes in 9 cycles after initiation of the modified antiemetic regimen), transient renal insufficiency (5 episodes in 5 patients), grade 4 thrombocytopenia (24 episodes, 1 associated with bleeding), neutropenia with or without fever (15 instances, only 11 in 112 cycles after routine use of granulocyte colony-stimulating factor), and catheter-related bacteremia (2 patients). Five (16%) of 30 patients who were treated after the last protocol modification experienced what we defined as unacceptable toxicity for a cooperative group setting. Responses were seen in 19 of 40 evaluable patients (relative risk, 48%) with 8 complete responses (20%). The median response duration was 7 months (range, 1-17+ months) with one currently ongoing. The central nervous system was the initial site of relapse in 11 responding patients. The median survival duration was 11 months (range, 2-31 months). This modified, concurrent biochemotherapy regimen is active and tolerable for use in a cooperative group setting. Central nervous system relapse, however, remains a concern for responders. This regimen is being compared with CVD in a Phase III Intergroup Trial (Eastern Cooperative Oncology Group/Southwest Oncology Group 3695).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Melanoma/tratamento farmacológico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias do Sistema Nervoso Central/secundário , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Terapia Combinada , Dacarbazina/administração & dosagem , Dacarbazina/efeitos adversos , Intervalo Livre de Doença , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Interferon-alfa/efeitos adversos , Interleucina-2/administração & dosagem , Interleucina-2/efeitos adversos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Projetos Piloto , Proteínas Recombinantes , Recidiva , Fatores de Tempo , Resultado do Tratamento , Vimblastina/administração & dosagem , Vimblastina/efeitos adversos
11.
Bone Marrow Transplant ; 50 Suppl 2: S77-81, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26039214

RESUMO

Age-related decline in thymic function is a well-described process that results in reduced T-cell development and thymic output of new naïve T cells. Thymic involution leads to reduced response to vaccines and new pathogens in otherwise healthy individuals; however, reduced thymic function is particularly detrimental in clinical scenarios where the immune system is profoundly depleted such as after chemotherapy, radiotherapy, infection and shock. Poor thymic function and restoration of immune competence has been correlated with an increased risk of opportunistic infections, tumor relapse and autoimmunity. Apart from their primary role in sex dimorphism, sex steroid levels profoundly affect the immune system in general and, in fact, age-related thymic involution has been at least partially attributed to the increase in sex steroids at puberty. Subsequently it has been demonstrated that the removal of sex steroids, or sex steroid ablation (SSA), triggers physiologic changes that ultimately lead to thymic re-growth and improved T-cell reconstitution in settings of hematopoietic stem cell transplant (HSCT). Although the cellular and molecular process underlying these regenerative effects are still poorly understood, SSA clearly represents an attractive therapeutic approach to enhance thymic function and restore immune competence in immunodeficient individuals.


Assuntos
Envelhecimento/imunologia , Hormônios Esteroides Gonadais/antagonistas & inibidores , Hospedeiro Imunocomprometido/imunologia , Imunoterapia/métodos , Medicina Regenerativa/métodos , Timo/imunologia , Envelhecimento/patologia , Hormônios Esteroides Gonadais/imunologia , Humanos , Timo/fisiologia
12.
Leukemia ; 29(7): 1530-42, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25652739

RESUMO

Co-transplantation of hematopoietic stem cells with those engineered to express leukemia-reactive T-cell receptors (TCRs) and differentiated ex vivo into precursor T cells (preTs) may reduce the risk of leukemia relapse. As expression of potentially self-(leukemia-) reactive TCRs will lead to negative selection or provoke autoimmunity upon thymic maturation, we investigated a novel concept whereby TCR expression set under the control of an inducible promoter would allow timely controlled TCR expression. After in vivo maturation and gene induction, preTs developed potent anti-leukemia effects. Engineered preTs provided protection even after repeated leukemia challenges by giving rise to effector and central memory cells. Importantly, adoptive transfer of TCR-transduced allogeneic preTs mediated anti-leukemia effect without evoking graft-versus-host disease (GVHD). Earlier transgene induction forced CD8(+) T-cell development was required to obtain a mature T-cell subset of targeted specificity, allowed engineered T cells to efficiently pass positive selection and abrogated the endogenous T-cell repertoire. Later induction favored CD4 differentiation and failed to produce a leukemia-reactive population emphasizing the dominant role of positive selection. Taken together, we provide new functional insights for the employment of TCR-engineered precursor cells as a controllable immunotherapeutic modality with significant anti-leukemia activity.


Assuntos
Doença Enxerto-Hospedeiro/prevenção & controle , Efeito Enxerto vs Leucemia/imunologia , Leucemia Mieloide/imunologia , Células Precursoras de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/genética , Transferência Adotiva , Animais , Citometria de Fluxo , Engenharia Genética , Doença Enxerto-Hospedeiro/mortalidade , Transplante de Células-Tronco Hematopoéticas , Humanos , Leucemia Mieloide/mortalidade , Leucemia Mieloide/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/genética , Regiões Promotoras Genéticas/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transplante Homólogo
13.
Transplantation ; 69(3): 446-9, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10706060

RESUMO

BACKGROUND: We studied whether T-cell clones, which appear in the periphery as a result of the failure of thymic negative selection during graft-versus-host disease (GVHD), have any in vivo anti-host reactivity and can cause GVHD in an adoptive transfer model. METHODS: We performed our studies in a murine model (B10.BR into CBA/J) for allogeneic bone marrow transplantation with major histocompatibility complex-matched and minor histocompatibility antigen-mismatched unrelated donors and unique Vbeta T-cell deletion patterns in donors and recipients. RESULTS: GVHD resulted in the appearance of Vbeta6+ T cells as a result of a loss of negative selection. We found that Vbeta6+ T cells from normal donors proliferated in vitro and in vivo. Depletion of Vbeta6+ T cells from the donor T-cell inoculum resulted in less GVHD morbidity and a decrease in the loss of thymic cellularity. To test the anti-host reactivity of de novo generated Vbeta6+ T cells in animals with GVHD, we developed an adoptive transfer model of splenic T cells from CBA/J host animals with GVHD into sublethally irradiated CBA/J recipients Depletion of Vbeta6+ T cells from the splenic T cells before adoptive transfer could significantly decrease the transient GVHD morbidity in the sublethally irradiated hosts. CONCLUSIONS: Our data indicate that GVHD-associated thymic damage results in a loss of thymic negative selection, which leads to the appearance of T-cell clones with anti-host reactivity in vitro and in vivo.


Assuntos
Transplante de Medula Óssea , Doença Enxerto-Hospedeiro/imunologia , Subpopulações de Linfócitos T/imunologia , Imunologia de Transplantes , Animais , Apresentação de Antígeno , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Camundongos , Timo/imunologia , Transplante Homólogo
14.
Transplantation ; 55(2): 355-61, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8434388

RESUMO

Reconstitution of B10 recipient mice, conditioned with total body irradiation (950 rads), with 40 x 10(6) untreated F344 or WF rat bone marrow cells results in stable rat stem-cell engraftment with multilineage lymphohematopoietic chimerism. We have now characterized NK cell generation, maturation, and function in fully xenogeneic chimeras (WF rat-->B10 mouse; F344-->B10 mouse). Early during xenogeneic reconstitution, rat-derived NK cells predominated in splenic lymphoid tissue, composing 14-18% of total cells at week 1 and increasing to 35.6-59.9% of total cells at week 2. By week 6, levels of rat NK cells had decreased and stabilized to that expected for normal rat (9-14.2%). The NK chimerism was reliably stable for up to 7 months following reconstitution. Most importantly, rat-derived NK cells were functional in both YAC tumor cytolysis and ADCC assays, suggesting that the xenogeneic mouse host environment was sufficient to support the generation, maturation, and function of rat-derived NK cells.


Assuntos
Transplante de Medula Óssea , Quimera/imunologia , Células Matadoras Naturais/fisiologia , Transplante Heterólogo/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos , Citotoxicidade Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos WF
15.
Transplantation ; 53(4): 815-22, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1566347

RESUMO

We report the induction of stable and reliably detectable mixed xenogeneic chimerism through the coadministration of a mixture of untreated rat bone marrow plus T cell-depleted mouse bone marrow into B10 recipients conditioned with total body irradiation (TCD B10 mouse + untreated F344 rat----B10 mouse). Recipients repopulated as true mixed lymphopoietic chimeras, with from 1-21.6% rat-derived lymphoid cells in peripheral blood and splenic lymphoid tissue. Production of rat platelets was also demonstrated. Rat platelet and lymphoid chimerism was reliably detectable in chimeras from 1 to 7 months following reconstitution, suggesting engraftment of the rat bone marrow stem cell. Production of each stem cell-derived lineage appeared to be under independent regulation since a significantly greater proportion of platelets were rat-derived (24-81%) than were lymphocytes (1-21.6% rat), while erythrocytes were preferentially syngeneic (less than 2% rat). The tolerance induced by this model was highly donor strain-specific: donor-specific rat and mouse skin grafts were accepted while MHC-disparate third-party mouse (C3H; H-2k) and rat (Wistar Furth; Rt1Au) skin grafts were promptly rejected. Although specifically prolonged xenogeneic donor rat skin grafts underwent a slow chronic rejection, and some totally disappeared. In spite of this, chimeras retained their lymphoid chimerism, suggesting the presence of skin-specific antigens. This model for mixed xenogeneic chimerism with reliably detectable rat lymphoid cells may provide a model to study the existence of tissue-specific antigens across a species barrier, as well as mechanisms responsible for the induction and maintenance of this strain-specific transplantation tolerance.


Assuntos
Antígenos/imunologia , Transplante de Medula Óssea , Quimera , Tolerância Imunológica , Pele/imunologia , Transplante Heterólogo , Animais , Citometria de Fluxo , Sobrevivência de Enxerto , Masculino , Camundongos , Camundongos Endogâmicos , Ratos , Ratos Endogâmicos , Transplante de Pele , Especificidade da Espécie , Linfócitos T/fisiologia
16.
Transplantation ; 70(1): 184-91, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10919598

RESUMO

BACKGROUND: The Fas/Fas ligand (FasL) pathway plays an important role in a number of apoptotic processes that could be important for the development of graft-versus-host disease (GVHD) after bone marrow transplantation (BMT), such as cytolysis of target cells by cytotoxic T cells, regulation of inflammatory responses, peripheral deletion of autoimmune cells, costimulation of T cells, and activation-induced cell death. METHODS: To study the role of the Fas/FasL pathway in the complex pathophysiology of graft versus host disease (GVHD), we used FasL-deficient B6.gld mice as recipients in a Major Histocompatibility Antigen Complex-matched minor Histocompatibility Antigen-mismatched murine model for GVHD after allogeneic BMT (C3H.SW-->B6). RESULTS: We found a significantly higher morbidity and mortality from GVHD compared to control B6 recipients. Histopathological analysis of the GVHD target organs demonstrated that B6.gld recipients developed significantly more thymic and intestinal GVHD. B6gld recipients with GVHD demonstrated an increased expansion of donor T cells and monocytes/ macrophages compared to control B6 recipients, whereas serum TNF-alpha levels were equivalent in B6.gld recipients and control B6 recipients. CONCLUSION: This study demonstrates that the expression of FasL in the BMT recipient is important for the host's ability to control GVHD.


Assuntos
Transplante de Medula Óssea/efeitos adversos , Doença Enxerto-Hospedeiro/etiologia , Glicoproteínas de Membrana/fisiologia , Animais , Suscetibilidade a Doenças , Proteína Ligante Fas , Feminino , Macrófagos/fisiologia , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Monócitos/fisiologia , Linfócitos T/imunologia , Transplante Homólogo , Fator de Necrose Tumoral alfa/biossíntese
17.
Neth Heart J ; 11(4): 163-168, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25696203

RESUMO

Both contrast-enhanced magnetic resonance imaging (CE-MRI) and myocardial contrast echocardiography (MCE) are promising tools to detect cardiac inflammation. CE-MRI can be used to characterise the location and extent of myocardial inflammation, since areas of abnormal signal enhancement associated with regional wall motion abnormalities reliably indicate areas of active myocarditis. In MCE, chemically composed microbubbles can be visualised by ultrasound and used to determine the status of the cardiac microvasculature. If there is any inflammation the microbubbles will be phagocytosed by neutrophils and monocytes, thus enabling the degree of inflammation to be assessed. These noninvasive techniques may allow early diagnosis and accurate evaluation of myocardial inflammation.

18.
Bone Marrow Transplant ; 48(1): 99-104, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22750997

RESUMO

Palifermin, a recombinant human keratinocyte growth factor, is commonly given to prevent mucositis following autologous transplantation. In the allogeneic hematopoietic stem cell transplant (allo-HSCT) setting, safety and efficacy data are limited. We conducted a retrospective study in 251 patients undergoing allo-HSCT, 154 of whom received peritransplant palifermin. In all patients, palifermin significantly decreased the mean number of days of total parenteral nutrition (TPN, 13 vs 16 days, P=0.006) and patient-controlled analgesia (PCA, 6 vs 10 days, P=0.023), as well as the length of initial hospital stay (LOS, 32 vs 37 days, P=0.014). However, the effect of palifermin was only significant in patients who received a TBI- but not BU-based chemotherapy conditioning regimen. In TBI recipients, palifermin decreased the mean number of days of TPN (13 vs 17 days, P<0.001) and PCA (7 vs 12 days, P=0.033), and the length of stay (32 vs 38 days, P=0.001). Palifermin did not affect GVHD, graft failure or relapse. Therefore, in the largest analysis with this patient population to date, we demonstrate that palifermin is safe in allo-HSCT patients, decreases TPN and PCA use and decreases LOS following TBI-based but not chemotherapy-based allo-HSCT.


Assuntos
Fator 7 de Crescimento de Fibroblastos/uso terapêutico , Trato Gastrointestinal/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Mucosite/prevenção & controle , Substâncias Protetoras/uso terapêutico , Condicionamento Pré-Transplante/efeitos adversos , Irradiação Corporal Total/efeitos adversos , Adulto , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Estudos de Coortes , Feminino , Fator 7 de Crescimento de Fibroblastos/efeitos adversos , Fator 7 de Crescimento de Fibroblastos/genética , Seguimentos , Trato Gastrointestinal/fisiopatologia , Trato Gastrointestinal/efeitos da radiação , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Mucosite/epidemiologia , Mucosite/etiologia , Mucosite/fisiopatologia , Cidade de Nova Iorque/epidemiologia , Substâncias Protetoras/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Estudos Retrospectivos , Índice de Gravidade de Doença , Análise de Sobrevida , Condicionamento Pré-Transplante/métodos , Transplante Homólogo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA