Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 390(8): 687-700, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38381673

RESUMEN

BACKGROUND: Treatment for autoimmune diseases such as systemic lupus erythematosus (SLE), idiopathic inflammatory myositis, and systemic sclerosis often involves long-term immune suppression. Resetting aberrant autoimmunity in these diseases through deep depletion of B cells is a potential strategy for achieving sustained drug-free remission. METHODS: We evaluated 15 patients with severe SLE (8 patients), idiopathic inflammatory myositis (3 patients), or systemic sclerosis (4 patients) who received a single infusion of CD19 chimeric antigen receptor (CAR) T cells after preconditioning with fludarabine and cyclophosphamide. Efficacy up to 2 years after CAR T-cell infusion was assessed by means of Definition of Remission in SLE (DORIS) remission criteria, American College of Rheumatology-European League against Rheumatism (ACR-EULAR) major clinical response, and the score on the European Scleroderma Trials and Research Group (EUSTAR) activity index (with higher scores indicating greater disease activity), among others. Safety variables, including cytokine release syndrome and infections, were recorded. RESULTS: The median follow-up was 15 months (range, 4 to 29). The mean (±SD) duration of B-cell aplasia was 112±47 days. All the patients with SLE had DORIS remission, all the patients with idiopathic inflammatory myositis had an ACR-EULAR major clinical response, and all the patients with systemic sclerosis had a decrease in the score on the EUSTAR activity index. Immunosuppressive therapy was completely stopped in all the patients. Grade 1 cytokine release syndrome occurred in 10 patients. One patient each had grade 2 cytokine release syndrome, grade 1 immune effector cell-associated neurotoxicity syndrome, and pneumonia that resulted in hospitalization. CONCLUSIONS: In this case series, CD19 CAR T-cell transfer appeared to be feasible, safe, and efficacious in three different autoimmune diseases, providing rationale for further controlled clinical trials. (Funded by Deutsche Forschungsgemeinschaft and others.).


Asunto(s)
Antígenos CD19 , Inmunoterapia Adoptiva , Lupus Eritematoso Sistémico , Agonistas Mieloablativos , Miositis , Esclerodermia Sistémica , Humanos , Antígenos CD19/administración & dosificación , Síndrome de Liberación de Citoquinas/etiología , Estudios de Seguimiento , Lupus Eritematoso Sistémico/terapia , Miositis/terapia , Esclerodermia Sistémica/terapia , Agonistas Mieloablativos/administración & dosificación , Ciclofosfamida/administración & dosificación , Infecciones/etiología , Resultado del Tratamiento
2.
Eur J Immunol ; 51(10): 2478-2484, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34350584

RESUMEN

Treatment with convalescent plasma has been shown to be safe in coronavirus disease in 2019 (COVID-19) infection, although efficacy reported in immunocompetent patients varies. Nevertheless, neutralizing antibodies are a key requisite in the fight against viral infections. Patients depleted of antibody-producing B cells, such as those treated with rituximab (anti-CD20) for hematological malignancies, lack a fundamental part of their adaptive immunity. Treatment with convalescent plasma appears to be of general benefit in this particularly vulnerable cohort. We analyzed clinical course and inflammation markers of three B-cell-depleted patients suffering from COVID-19 who were treated with convalescent plasma. In addition, we measured serum antibody levels as well as peripheral blood CD38/HLA-DR-positive T-cells ex vivo and CD137-positive T-cells after in vitro stimulation with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-derived peptides in these patients. We observed that therapy with convalescent plasma was effective in all three patients and analysis of CD137-positive T-cells after stimulation with SARS-CoV-2 peptides showed an increase in peptide-specific T-cells after application of convalescent plasma. In conclusion, we here demonstrate efficacy of convalescent plasma therapy in three B-cell-depleted patients and present data that suggest that while application of convalescent plasma elevates systemic antibody levels only transiently, it may also boost specific T-cell responses.


Asunto(s)
Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , COVID-19/terapia , Linfocitos T/inmunología , Adolescente , Anciano , Anticuerpos Neutralizantes/sangre , Linfocitos B/citología , Humanos , Inmunidad Celular/inmunología , Inmunización Pasiva/métodos , Recuento de Linfocitos , Depleción Linfocítica , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células del Manto/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Rituximab/efectos adversos , SARS-CoV-2/inmunología , Resultado del Tratamiento , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Sueroterapia para COVID-19
3.
Eur J Immunol ; 51(6): 1436-1448, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33784417

RESUMEN

COVID-19 is a life-threatening disease leading to bilateral pneumonia and respiratory failure. The underlying reasons why a smaller percentage of patients present with severe pulmonary symptoms whereas the majority is only mildly affected are to date not well understood. Comparing the immunological phenotype in healthy donors and patients with mild versus severe COVID-19 shows that in COVID-19 patients, NK-/B-cell activation and proliferation are enhanced independent of severity. As an important precondition for effective antibody responses, T-follicular helper cells and antibody secreting cells are increased both in patients with mild and severe SARS-CoV-2 infection. Beyond this, T cells in COVID-19 patients exhibit a stronger activation profile with differentiation toward effector cell phenotypes. Importantly, when looking at the rates of pulmonary complications in COVID-19 patients, the chemokine receptor CCR4 is higher expressed by both CD4 and CD8 T cells of patients with severe COVID-19. This raises the hypothesis that CCR4 upregulation on T cells in the pathogenesis of COVID-19 promotes stronger T-cell attraction to the lungs leading to increased immune activation with presumably higher pulmonary toxicity. Our study contributes significantly to the understanding of the immunological changes during COVID-19, as new therapeutic agents, preferentially targeting the immune system, are highly warranted.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Pulmón/inmunología , Activación de Linfocitos , Receptores CCR4/inmunología , SARS-CoV-2/inmunología , Regulación hacia Arriba/inmunología , Adulto , Linfocitos T CD8-positivos/patología , COVID-19/patología , Femenino , Humanos , Pulmón/patología , Pulmón/virología , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
4.
Clin Oral Investig ; 25(4): 1705-1713, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32754787

RESUMEN

OBJECTIVES: Recently, multiple studies addressed the importance of lymph node ratio (LNR) in specifying patients' risk of disease recurrence in various malignancies. The present study examines the prognostic significance of LNR in predicting outcome of oral squamous cell carcinoma (OSCC) patients after surgical treatment with curative intent. METHODS: Here, we describe a retrospective population-based cohort with 717 patients previously diagnosed with OSCC. Histopathologically verified lymph node metastasis was diagnosed in 290 patients. Among these patients, we evaluated the impact of LNR on overall survival (OAS) and recurrence-free survival (RFS) in uni- as well as multivariate analysis. RESULTS: A median cutoff (0.055) in LNR was found to significantly predict outcome in OSCC patients. Five-year OAS was 54.1% in patients with a low LNR, whereas a high LNR was associated with a 5-year OAS of 33.3% (p < 0.001). Similar results were detected for RFS with a 5-year survival rate of 49.8% (LNR low) and 30.3% (LNR high) (p = 0.002). Results were confirmed in multivariate Cox regression which substantiated the importance of LNR in predicting survival in OSCC patients. CONCLUSIONS: LNR was shown to be an independent prognostic factor for outcome of OSCC in a population-based cohort in uni- as well as multivariate analysis. Hereby, a LNR ≥ 0.055 predicted a shorter OAS and RFS in our cohort. CLINICAL RELEVANCE: Besides established histopathological factors, LNR can be used as a reliable predictor of outcome in OSCC and might therefore be further applied in evaluating adjuvant treatment after resection in curative intention.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/cirugía , Estudios de Cohortes , Humanos , Escisión del Ganglio Linfático , Índice Ganglionar , Ganglios Linfáticos , Neoplasias de la Boca/patología , Neoplasias de la Boca/cirugía , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Pronóstico , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello
5.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-35008509

RESUMEN

Oral cancer often presents with aggressive behavior and a high risk of recurrence and metastasis. For oral squamous cell carcinoma (OSCC), which is the most frequent histological subtype, therapy strategies include surgery, radiation therapy, chemotherapy, immune checkpoint inhibitors, and EGFR inhibitors. Recently, a Trop-2 antibody-drug conjugate (ADC) has been approved in the United States of America for the treatment of advanced triple-negative breast cancer. However, this ADC has also been tested in other solid tumors including head & neck squamous cell carcinoma. The prognostic impact of Trop-2 has already been reported for several cancers. We studied the prognostic influence of Trop-2 protein expression on OSCC patients' survival. The cohort comprised n = 229 OSCC patients with available archived tumor tissue and corresponding non-neoplastic oral mucosa tissue. Using immunohistochemistry, we investigated Trop-2 expression in both the central and peripheral regions of each tumor and in corresponding non-neoplastic oral mucosa. In patients suffering from OSCC with combined high central and low peripheral Trop-2 expression, five-year overall survival (OS) was 41.2%, whereas 55.6% of OSCC patients who presented lower central and/or higher peripheral tumoral Trop-2 expression were alive after five years (p = 0.075). In multivariate Cox regression, the expression pattern of high central tumoral and lower peripheral Trop-2 expression was significantly correlated with impaired OS (HR = 1.802, 95%-CI: 1.134-2.864; p = 0.013) and recurrence-free survival (RFS) (HR = 1.633, 95%-CI: 1.042-2.560; p = 0.033), respectively, when adjusting for co-variables. Hence, Trop-2 may serve as an independent prognostic biomarker in OSCC. In subsequent studies, the pathophysiological meaning of downregulated Trop-2 expression in the OSCC periphery has to be analyzed.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Carcinoma de Células Escamosas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias de la Boca/metabolismo , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/patología , Femenino , Humanos , Inmunoconjugados/metabolismo , Inmunohistoquímica/métodos , Masculino , Persona de Mediana Edad , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Neoplasias de la Boca/patología , Pronóstico
7.
J Immunol ; 199(7): 2356-2365, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28842469

RESUMEN

Maintaining immune tolerance requires the production of Foxp3-expressing regulatory T (Treg) cells in the thymus. Activation of NF-κB transcription factors is critically required for Treg cell development, partly via initiating Foxp3 expression. NF-κB activation is controlled by a negative feedback regulation through the ubiquitin editing enzyme A20, which reduces proinflammatory signaling in myeloid cells and B cells. In naive CD4+ T cells, A20 prevents kinase RIPK3-dependent necroptosis. Using mice deficient for A20 in T lineage cells, we show that thymic and peripheral Treg cell compartments are quantitatively enlarged because of a cell-intrinsic developmental advantage of A20-deficient thymic Treg differentiation. A20-deficient thymic Treg cells exhibit reduced dependence on IL-2 but unchanged rates of proliferation and apoptosis. Activation of the NF-κB transcription factor RelA was enhanced, whereas nuclear translocation of c-Rel was decreased in A20-deficient thymic Treg cells. Furthermore, we found that the increase in Treg cells in T cell-specific A20-deficient mice was already observed in CD4+ single-positive CD25+ GITR+ Foxp3- thymic Treg cell progenitors. Treg cell precursors expressed high levels of the tumor necrosis factor receptor superfamily molecule GITR, whose stimulation is closely linked to thymic Treg cell development. A20-deficient Treg cells efficiently suppressed effector T cell-mediated graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, suggesting normal suppressive function. Holding thymic production of natural Treg cells in check, A20 thus integrates Treg cell activity and increased effector T cell survival into an efficient CD4+ T cell response.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T Reguladores/fisiología , Timo/citología , Timo/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis , Diferenciación Celular , Citometría de Flujo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Enfermedad Injerto contra Huésped/prevención & control , Interleucina-2/inmunología , Activación de Linfocitos , Ratones , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/genética , Transducción de Señal , Trasplante de Células Madre , Timo/inmunología , Factor de Transcripción ReIA/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/deficiencia
8.
Eur J Immunol ; 47(11): 1982-1988, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28833031

RESUMEN

The NF-κB regulator A20 limits inflammation by providing negative feedback in myeloid cells and B cells. Functional lack of A20 has been linked to several inflammatory and autoimmune diseases. To define how A20 affects the functionality of T effector cells in a highly inflammatory environment, we performed conventional allogeneic hematopoietic stem cell transplantation (allo-HSCT) with A20-deficient CD4+ and CD8+ donor T cells in mice. Severity and mortality of graft-versus-host disease (GVHD) after allo-HSCT was drastically reduced in recipients transplanted with conventional doses of A20-deficient T cells. Consistently, we found that the A20-deficient donor T-cell compartment was strongly diminished at various timepoints after allo-HSCT. However, proportionally more A20-deficient donor T cells produced IFN-γ and systemic inflammation was elevated early after allo-HSCT. Consequently, increasing the dose of transplanted A20-deficient T cells reversed the original phenotype and resulted in enhanced GVHD mortality compared to recipients that received A20+/+ T cells. Still, A20-deficient T cells, activated either through T cell receptor-dependent or -independent mechanisms, were less viable than control A20+/+ T cells, highlighting that A20 balances both, T-cell activation and survival. Thus, our findings suggest that targeting A20 in T cells may allow to modulate T-cell-mediated inflammatory diseases like GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Linfocitos T/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Animales , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante Homólogo
9.
Eur J Immunol ; 47(5): 872-879, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28295265

RESUMEN

Activation of the C-type lectin receptor Dectin-1 by ß-glucans triggers multiple signals within DCs that result in activation of innate immunity. While these mechanisms can potently prime CD8+ cytotoxic T-cell (CTL) responses without additional adjuvants, the Dectin-1 effector pathways that control CTL induction remain unclear. Here we demonstrate that Dectin-1-induced CTL cross-priming in mice does not require inflammasome activation but strictly depends on the adapter protein Card9 in vitro. In vivo, Dectin-1-mediated Card9 activation after vaccination drives both expansion and activation of Ag-specific CTLs, resulting in long-lasting CTL responses that are sufficient to protect mice from tumor challenge. This Dectin-1-induced antitumor immune response was independent of NK cell function and completely abrogated in Card9-deficient mice. Thus, our results demonstrate that Dectin-1-triggered Card9 signaling but not inflammasome activation can potently cross-prime Ag-specific CTLs, suggesting that this pathway would be a candidate for immunotherapy and vaccine development.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Lectinas Tipo C/metabolismo , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas Adaptadoras de Señalización CARD/genética , Reactividad Cruzada , Inmunidad Innata , Inflamasomas/inmunología , Ratones , Ratones Endogámicos C57BL , Neoplasias/fisiopatología , Transducción de Señal , Vacunación
10.
Transfusion ; 58(7): 1772-1780, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29732573

RESUMEN

BACKGROUND: Cell separators are routinely used to collect CD34+ blood stem cells in the context of customized stem cell transplantation procedures. The Spectra Optia (Terumo BCT) is a novel development of the precursor instrument, the Cobe Spectra (Terumo BCT). STUDY DESIGN AND METHODS: In this report, 146 autologous and 42 allogeneic donors undergoing apheresis on the Cobe Spectra using the mononuclear cell (MNC) program 4.7 or on the Spectra Optia using the new continuous mononuclear cell (cMNC) program 11.2 are compared. RESULTS: Viability of cells and collection efficacy within the apheresis products was comparable for autologous and allogeneic products collected with the MNC or cMNC method. However, we found a reduced duration of the apheresis procedure and lower hematocrit within the apheresis products when using the cMNC in autologous and allogeneic donors. Moreover, allogeneic donors collected substantially more CD34+ cells per kilogram of body weight when using the cMNC method. Differences in platelets before and after apheresis were substantially smaller in this cohort when compared to the cohort collected with the MNC method. Neutrophil and platelet engraftment after autologous or allogeneic transplantation with a product collected with the MNC procedure was comparable to a transplantation with a product processed according to the cMNC method. CONCLUSION: Comparison of the MNC (Cobe Spectra) and the cMNC (Spectra Optia) methods demonstrated an equal performance and outcome. However, advantages were present using the cMNC method with respect to apheresis duration and hematocrit within the apheresis product (autologous/allogeneic donors) and numbers of CD34+ cells collected, especially in allogeneic donors.


Asunto(s)
Eliminación de Componentes Sanguíneos/métodos , Adulto , Anciano , Antígenos CD34/metabolismo , Donantes de Sangre , Femenino , Movilización de Célula Madre Hematopoyética , Humanos , Leucocitos Mononucleares/citología , Masculino , Persona de Mediana Edad , Células Madre de Sangre Periférica/citología , Trasplante Homólogo/métodos , Adulto Joven
11.
Transfusion ; 57(1): 115-121, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27859332

RESUMEN

BACKGROUND: Plerixafor is predominantly used for patients mobilizing inadequate stem cell numbers for autologous transplantation after stimulation with granulocyte-colony-stimulating factor (G-CSF). STUDY DESIGN AND METHODS: We here report on 300 patients undergoing stem cell mobilization with G-CSF, among them 36 poor mobilizers (CD34+ cell counts < 50 × 106 /L blood) receiving G-CSF alone and 49 receiving G-CSF in combination with plerixafor for rescue intervention. Mobilization efficacy and short-time outcome after autologous stem cell transplantation were analyzed and compared in the respective subgroups. RESULTS: Out of 49 patients treated with plerixafor and G-CSF, 46 (94%) collected sufficient hematopoietic stem cell numbers although the number was clearly inferior in poor mobilizers. Compared to good mobilizers, viability of CD34+ cells analyzed after collection was slightly reduced in poor mobilizers independent of the application of plerixafor. A total of 232 patients underwent autologous stem cell transplantation, among them 26 poor mobilizers who received only G-CSF and 31 patients who received G-CSF in combination with plerixafor. Time until neutrophil engraftment was in median 1 day later in poor mobilizers irrespective of the application of plerixafor. Platelet engraftment was in median 2 days delayed in patients mobilized with G-CSF and plerixafor compared to 1 day in poor mobilizers treated with G-CSF only. Frequency of detected CD38+ CD138+ CD45- CD56+ plasma cells in the apheresis products of myeloma patients was comparable for all groups. CONCLUSION: Our data demonstrate that plerixafor is highly effective as rescue measurement after mobilization failure with G-CSF alone and short-term clinical outcome after stem cell transplantation is comparable.


Asunto(s)
Eliminación de Componentes Sanguíneos , Movilización de Célula Madre Hematopoyética/métodos , Compuestos Heterocíclicos/administración & dosificación , Mieloma Múltiple/terapia , Trasplante de Células Madre de Sangre Periférica , Células Madre de Sangre Periférica , Autoinjertos , Bencilaminas , Ciclamas , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Humanos , Masculino , Mieloma Múltiple/sangre , Células Plasmáticas/metabolismo , Estudios Retrospectivos
12.
Blood ; 123(24): 3832-42, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24711661

RESUMEN

Graft-versus-host-disease (GVHD) is a severe complication of allogeneic hematopoietic cell transplantation (allo-HCT) characterized by the production of high levels of proinflammatory cytokines. Activated Janus kinases (JAKs) are required for T-effector cell responses in different inflammatory diseases, and their blockade could potently reduce acute GVHD. We observed that inhibition of JAK1/2 signaling resulted in reduced proliferation of effector T cells and suppression of proinflammatory cytokine production in response to alloantigen in mice. In vivo JAK 1/2 inhibition improved survival of mice developing acute GVHD and reduced histopathological GVHD grading, serum levels of proinflammatory cytokines, and expansion of alloreactive luc-transgenic T cells. Mechanistically, we could show that ruxolitinib impaired differentiation of CD4(+) T cells into IFN-γ- and IL17A-producing cells, and that both T-cell phenotypes are linked to GVHD. Conversely, ruxolitinib treatment in allo-HCT recipients increased FoxP3(+) regulatory T cells, which are linked to immunologic tolerance. Based on these results, we treated 6 patients with steroid-refractory GVHD with ruxolitinib. All patients responded with respect to clinical GVHD symptoms and serum levels of proinflammatory cytokines. In summary, ruxolitinib represents a novel targeted approach in GVHD by suppression of proinflammatory signaling that mediates tissue damage and by promotion of tolerogenic Treg cells.


Asunto(s)
Enfermedad Injerto contra Huésped/tratamiento farmacológico , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Animales , Incompatibilidad de Grupos Sanguíneos/complicaciones , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Nitrilos , Pirimidinas , Índice de Severidad de la Enfermedad , Trasplante Homólogo/efectos adversos , Resultado del Tratamiento
13.
Adv Exp Med Biol ; 951: 67-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27837555

RESUMEN

Autologous and allogeneic stem cell transplantation (SCT) represents a therapeutic option widely used for hematopoietic malignancies. One important milestone in the development of this treatment strategy was the development of effective cryopreservation technologies resulting in a high quality with respect to cell viability as well as lack of contamination of the graft.Stem cell preparations have been initially performed within standard laboratories as it is routinely still the case in many countries. With the emergence of cleanrooms, manufacturing of stem cell preparations within these facilities has become a new standard mandatory in Europe. However, due to high costs and laborious procedures, novel developments recently emerged using closed bag systems as reliable alternatives to conventional cleanrooms. Several hurdles needed to be overcome including the addition of the cryoprotectant dimethylsulfoxide (DMSO) as a relevant manipulation. As a result of the development, closed bag systems proved to be comparable in terms of product quality and patient outcome to cleanroom products. They also comply with the strict regulations of good manufacturing practice.With closed systems being available, costs and efforts of a cleanroom facility may be substantially reduced in the future. The process can be easily extended for other cell preparations requiring minor modifications as donor lymphocyte preparations. Moreover, novel developments may provide solutions for the production of advanced-therapy medicinal products in closed systems.


Asunto(s)
Criopreservación/métodos , Crioprotectores/farmacología , Dimetilsulfóxido/farmacología , Ambiente Controlado , Trasplante de Células Madre de Sangre Periférica , Células Madre de Sangre Periférica/efectos de los fármacos , Glicerol/farmacología , Neoplasias Hematológicas/terapia , Humanos , Células Madre de Sangre Periférica/citología , Células Madre de Sangre Periférica/fisiología , Guías de Práctica Clínica como Asunto , Control de Calidad , Trasplante Autólogo , Trasplante Homólogo
14.
Transfusion ; 55(11): 2702-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26172379

RESUMEN

BACKGROUND: In several European countries, preparation of cellular products with open manufacturing systems as used for cryopreservation of peripheral blood stem cells (PBSCs) needs to be performed in a clean-room facility. However, this form of manufacturing is highly expensive and laborious. Thus, safe techniques providing improved efficacy regarding time and material, which are in accordance with legal requirements are highly desirable. STUDY DESIGN AND METHODS: We have developed, validated, and applied a simple method for cryopreservation of PBSCs within a functionally closed-bag system using the closed cryo freeze prep set. This process fulfills good manufacturing practice requirements and allows for the cryopreservation of PBSCs without a clean-room facility. In addition to cryopreservation of PBSCs, we have recently successfully modified our system for processing, portioning, and cryopreservation of allogeneic donor lymphocytes. RESULTS: Since 2010, cryopreservation of PBSCs using a closed-bag system has been performed in our facility on a routine basis and 210 patients and healthy donors have been included in this analysis. No significant reduction in viability of CD34+ cells and no process-related contamination were observed. Outcome of hematopoietic stem cell transplantation regarding time of engraftment and infectious complications is comparable to products manufactured in conventional clean-room facilities. CONCLUSION: Our data confirm that cryopreservation of PBSCs within a functionally closed-bag system is safe, effective, and economical. Furthermore, the system has the potential to be extended to other manufacturing processes of cellular products.


Asunto(s)
Criopreservación/métodos , Células Madre Hematopoyéticas/citología , Células Cultivadas , Humanos
15.
J Immunol ; 188(2): 892-901, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22147766

RESUMEN

OX40 is a T cell costimulatory molecule that belongs to the TNFR superfamily. In the absence of immune activation, OX40 is selectively expressed by Foxp3(+) regulatory T cells (Tregs), but not by resting conventional T cells. The exact role of OX40 in Treg homeostasis and function remains incompletely defined. In this study, we demonstrate that OX40 engagement in vivo in naive mice induces initial expansion of Foxp3(+) Tregs, but the expanded Tregs have poor suppressive function and exhibit features of exhaustion. We also show that OX40 enables the activation of the Akt and Stat5 pathways in Tregs, resulting in transient proliferation of Tregs and reduced levels of Foxp3 expression. This creates a state of relative IL-2 deficiency in naive mice that further impacts Tregs. This exhausted Treg phenotype can be prevented by exogenous IL-2, as both OX40 and IL-2 agonists drive further expansion of Tregs in vivo. Importantly, Tregs expanded by both OX40 and IL-2 agonists are potent suppressor cells, and in a heart transplant model, they promote long-term allograft survival. Our data reveal a novel role for OX40 in promoting immune tolerance and may have important clinical implications.


Asunto(s)
Receptores OX40/fisiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Tolerancia al Trasplante/inmunología , Animales , Proliferación Celular , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/biosíntesis , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Trasplante de Corazón/patología , Inmunidad Celular/genética , Interleucina-2/deficiencia , Interleucina-2/fisiología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ligando OX40 , Receptores OX40/deficiencia , Receptores OX40/metabolismo , Linfocitos T Reguladores/trasplante , Tolerancia al Trasplante/genética , Factores de Necrosis Tumoral/metabolismo
16.
J Autoimmun ; 40: 86-95, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23031801

RESUMEN

The clearance of apoptotic cells occurs in a non-inflammatory context. Defects in this clearance process have been linked to the emergence of human autoimmune diseases like systemic lupus erythematosus (SLE). A characteristic of apoptotic cell death is the shedding of membrane coated vesicles from the cellular surfaces. Those vesicles have recently been recognized as mediators of intercellular communication or as adjuvant in the pathogenesis of autoimmune diseases. We analyzed the interactions between these apoptotic cell-derived membrane vesicles and professional antigen presenting cells. These vesicles were engulfed by monocyte-derived dendritic cells (mDC) and stimulated their maturation towards a phenotype comprising an upregulation of CD80, CD83, CD86, and a remarkable downregulation of MHC class II molecules. We observed only a minor release of proinflammatory cytokines from these mDC when compared to LPS stimulation. mDC stimulated by apoptotic vesicles did not cause significant T-cell expansion. Interestingly, when compared to normal healthy donors SLE patients-derived dendritic cells showed a significantly different phenotype lacking the downregulation of MHC class II, which correlated to disease activity.


Asunto(s)
Apoptosis , Vesículas Citoplasmáticas/inmunología , Células Dendríticas/inmunología , Lupus Eritematoso Sistémico/inmunología , Adulto , Anciano , Antígenos CD/metabolismo , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfocitos T CD4-Positivos/inmunología , Comunicación Celular , Proliferación Celular , Células Cultivadas , Células Dendríticas/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunoglobulinas/metabolismo , Interleucina-12/metabolismo , Interleucina-8/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Fagocitosis/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven , Antígeno CD83
17.
J Immunol ; 186(3): 1338-42, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21199897

RESUMEN

Costimulatory molecules regulate the functional outcome of T cell activation, and disturbance of the balance between activating and inhibitory signals results in increased susceptibility to infection or the induction of autoimmunity. Similar to the well-characterized CD28/CTLA-4 costimulatory pathway, a newly emerging pathway consisting of CD226 and T cell Ig and ITIM domain (TIGIT) has been associated with susceptibility to multiple autoimmune diseases. In this study, we examined the role of the putative coinhibitory molecule TIGIT and show that loss of TIGIT in mice results in hyperproliferative T cell responses and increased susceptibility to autoimmunity. TIGIT is thought to indirectly inhibit T cell responses by the induction of tolerogenic dendritic cells. By generating an agonistic anti-TIGIT Ab, we demonstrate that TIGIT can inhibit T cell responses directly independent of APCs. Microarray analysis of T cells stimulated with agonistic anti-TIGIT Ab revealed that TIGIT can act directly on T cells by attenuating TCR-driven activation signals.


Asunto(s)
Inhibidores de Crecimiento/fisiología , Activación de Linfocitos/inmunología , Receptores Inmunológicos/fisiología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Proliferación Celular , Células Cultivadas , Cricetinae , Cricetulus , Regulación hacia Abajo/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Predisposición Genética a la Enfermedad , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/deficiencia , Inmunosupresores/metabolismo , Inmunosupresores/farmacología , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/deficiencia , Transducción de Señal/genética , Linfocitos T/citología , Linfocitos T/metabolismo
18.
Biomedicines ; 11(2)2023 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-36830906

RESUMEN

The anti-cancer properties of statins have attracted much attention recently, but little is known about the prognostic role of statins in oral squamous cell carcinoma (OSCC). In a retrospective approach, we analyzed a population-based cohort of 602 OSCC patients with primary curative tumor resection to negative margins and concomitant neck dissection between 2005-2017. Long-term medication with statins was correlated with overall survival (OAS) as well as recurrence-free survival (RFS) using uni- and multivariable Cox regression. Additionally, propensity score matching was applied to adjust for confounders. Statin use was present in 96 patients (15.9%) at a median age of 65.7 years. Statin treatment correlated with ameliorated survival in multivariable Cox regression in the complete cohort (OAS: HR 0.664; 95% CI 0.467-0.945, p = 0.023; RFS: HR 0.662; 95% CI 0.476-0.920, p = 0.014) as well as matched-pair cohort of OSCC patients (OAS: HR 0.691; 95% CI 0.479-0.997, p = 0.048; RFS: HR 0.694; 95% CI 0.493-0.976, p = 0.036) when compared to patients not taking statins at time of diagnosis. These findings were even more pronounced by sub-group analysis in the matched-pair cohort (age < 70 years). These data indicate that statin use might ameliorate the oncological outcome in primarily resected OSCC patients, but prospective clinical trials are highly recommended.

19.
Cancers (Basel) ; 15(3)2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36765630

RESUMEN

A20, known as a potent inhibitor of NF-κB signaling, has been characterized in numerous clinical as well as preclinical studies. Recently, especially in various malignant diseases, the prognostic and therapeutic relevance of A20 was investigated. In oral squamous cell carcinoma (OSCC) however, the characterization of A20 is uncharted territory. We analyzed a tissue microarray (TMA) of 229 surgically-treated OSCC patients (2003-2013). Immunohistochemical (IHC) stainings were performed for A20 and CD3; additionally, standard haematoxylin-eosin staining was applied. IHC findings were correlated with a comprehensive dataset, comprising clinical and pathohistological information. A20 expression was analyzed in tumor cells as well as in tumor infiltrating lymphocytes (TILs) and correlated with the overall survival (OS) and recurrence-free survival (RFS) using uni- and multivariable Cox regression. The median follow-up time was 10.9 years and the A20 expression was significantly decreased in CD3+ TILs compared to mucosa-infiltrating lymphocytes (MILs). In the Kaplan-Meier analyses, higher A20 expression in TILs was correlated with better OS (p = 0.017) and RFS (p = 0.020). In the multivariable survival analysis, A20 overexpression correlated with improved OS (HR: 0.582; 95% CI 0.388-0.873, p = 0.009) and RFS (HR 0.605; 95% CI 0.411-0.889, p = 0.011). Our results indicate a novel prognostic role for A20 in OSCC. Due to its elevated expression in TILs, further research is highly desirable, which therefore could offer new therapeutic opportunities for patients suffering from OSCC.

20.
Cancers (Basel) ; 15(24)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38136436

RESUMEN

Molecular Tumor Boards (MTBs) converge state-of-the-art next-generation sequencing (NGS) methods with the expertise of an interdisciplinary team consisting of clinicians, pathologists, human geneticists, and molecular biologists to provide molecularly informed guidance in clinical decision making to the treating physician. In the present study, we particularly focused on elucidating the factors impacting on the clinical translation of MTB recommendations, utilizing data generated from gene panel mediated comprehensive genomic profiling (CGP) of 554 patients at the MTB of the Comprehensive Cancer Center Erlangen, Germany, during the years 2016 to 2020. A subgroup analysis of cases with available follow-up data (n = 332) revealed 139 cases with a molecularly informed MTB recommendation, which was successfully implemented in the clinic in 44 (31.7%) of these cases. Here, the molecularly matched treatment was applied in 45.4% (n = 20/44) of cases for ≥6 months and in 25% (n = 11/44) of cases for 12 months or longer (median time to treatment failure, TTF: 5 months, min: 1 month, max: 38 months, ongoing at data cut-off). In general, recommendations were preferentially implemented in the clinic when of high (i.e., tier 1) clinical evidence level. In particular, this was the case for MTB recommendations suggesting the application of PARP, PIK3CA, and IDH1/2 inhibitors. The main reason for non-compliance to the MTB recommendation was either the application of non-matched treatment modalities (n = 30)/stable disease (n = 7), or deteriorating patient condition (n = 22)/death of patient (n = 9). In summary, this study provides an insight into the factors affecting the clinical implementation of molecularly informed MTB recommendations, and careful considerations of these factors may guide future processes of clinical decision making.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA