Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Immunol ; 212: 108248, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31382036
2.
Biol Blood Marrow Transplant ; 20(3): 345-53, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24315842

RESUMEN

Busulfan (Bu) is used as a myeloablative agent in conditioning regimens before allogeneic hematopoietic cell transplantation (allo-HCT). In line with strategies explored in adults, patient outcomes may be optimized by replacing cyclophosphamide (Cy) with or without melphalan (Mel) with fludarabine (Flu). We compared outcomes in 2 consecutive cohorts of HCT recipients with a nonmalignant HCT indication, a myeloid malignancy, or a lymphoid malignancy with a contraindication for total body irradiation (TBI). Between 2009 and 2012, 64 children received Flu + Bu at a target dose of 80-95 mg·h/L, and between 2005 and 2008, 50 children received Bu targeted to 74-80 mg·h/L + Cy. In the latter group, Mel was added for patients with myeloid malignancy (n = 12). Possible confounding effects of calendar time were studied in 69 patients receiving a myeloablative dose of TBI between 2005 and 2012. Estimated 2-year survival and event-free survival were 82% and 78%, respectively, in the FluBu arm and 78% and 72%, respectively, in the BuCy (Mel) arm (P = not significant). Compared with the BuCy (Mel) arm, less toxicity was noted in the FluBu arm, with lower rates of acute (noninfectious) lung injury (16% versus 36%; P = .007), veno-occlusive disease (3% versus 28%; P = .003), chronic graft-versus-host disease (9% versus 26%; P = .047), adenovirus infection (3% versus 32%; P = .001), and human herpesvirus 6 infection reactivation (21% versus 44%; P = .005). Furthermore, the median duration of neutropenia was shorter in the FluBu arm (11 days versus 22 days; P < .001), and the patients in this arm required fewer transfusions. Our data indicate that Flu (160 mg/m(2)) with targeted myeloablative Bu (90 mg·h/L) is less toxic than and equally effective as BuCy (Mel) in patients with similar indications for allo-HCT.


Asunto(s)
Busulfano/uso terapéutico , Ciclofosfamida/uso terapéutico , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Agonistas Mieloablativos/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Vidarabina/análogos & derivados , Infecciones por Adenoviridae/mortalidad , Adolescente , Niño , Preescolar , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/mortalidad , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Humanos , Lactante , Recién Nacido , Pulmón/inmunología , Pulmón/patología , Masculino , Infecciones por Roseolovirus/mortalidad , Análisis de Supervivencia , Trasplante Homólogo , Resultado del Tratamiento , Vidarabina/uso terapéutico , Adulto Joven
3.
Stem Cell Res Ther ; 15(1): 125, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38679715

RESUMEN

BACKGROUND: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for leukemia and a range of non-malignant disorders. The success of the therapy is hampered by occurrence of acute graft-versus-host disease (aGvHD); an inflammatory response damaging recipient organs, with gut, liver, and skin being the most susceptible. Intestinal GvHD injury is often a life-threatening complication in patients unresponsive to steroid treatment. Allogeneic mesenchymal stromal/stem cell (MSC) infusions are a promising potential treatment for steroid-resistant aGvHD. Data from our institution and others demonstrate rescue of approximately 40-50% of aGvHD patients with MSCs in Phase I, II studies and minor side effects. Although promising, better understanding of MSC mode of action and patient response to MSC-based therapy is essential to improve this lifesaving treatment. METHODS: Single cell human small intestine organoids were embedded in Matrigel, grown for 5 days and treated with busulfan for 48 h. Organoids damaged by treatment with busulfan or control organoids were co-cultured with 5000, 10,000, and 50,000 MSCs for 24 h, 48 h or 7 days and the analyses such as surface area determination, proliferation and apoptosis assessment, RNA sequencing and proteomics were performed. RESULTS: Here, we developed a 3D co-culture model of human small intestinal organoids and MSCs, which allows to study the regenerative effects of MSCs on intestinal epithelium in a more physiologically relevant setting than existing in vitro systems. Using this model we mimicked chemotherapy-mediated damage of the intestinal epithelium. The treatment with busulfan, the chemotherapeutic commonly used as conditioning regiment before the HSCT, affected pathways regulating epithelial to mesenchymal transition, proliferation, and apoptosis in small intestinal organoids, as shown by transcriptomic and proteomic analysis. The co-culture of busulfan-treated intestinal organoids with MSCs reversed the effects of busulfan on the transcriptome and proteome of intestinal epithelium, which we also confirmed by functional evaluation of proliferation and apoptosis. CONCLUSIONS: Collectively, we demonstrate that our in vitro co-culture system is a new valuable tool to facilitate the investigation of the molecular mechanisms behind the therapeutic effects of MSCs on damaged intestinal epithelium. This could benefit further optimization of the use of MSCs in HSCT patients.


Asunto(s)
Mucosa Intestinal , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Regeneración/efectos de los fármacos , Organoides/metabolismo , Técnicas de Cocultivo , Enfermedad Injerto contra Huésped/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Busulfano/farmacología , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos
4.
Bone Marrow Transplant ; 58(7): 762-768, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37002411

RESUMEN

Busulfan exposure has previously been linked to clinical outcomes, hence the need for therapeutic drug monitoring (TDM). Study objective was to evaluate the effect of day 1 TDM-guided dosing (regimen d1) versus days 1 + 2 TDM-guided dosing (regimen d1 + 2) on attaining adequate busulfan exposure. In this observational study, we included all children receiving busulfan-based allogeneic hematopoietic cell transplantation. Primary outcome was the percentage of patients achieving busulfan target attainment in both TDM regimens. Secondary outcomes were the variance in busulfan exposure and day-4 clearance (Clday4) estimates between both TDM regimens and dosing day 1 and 2. In regimen d1, 84.3% (n = 91/108) attained a therapeutic busulfan exposure, while in regimen d1 + 2 a proportion of 90.9% was found (n = 30/33, not-significant). Variance of Clday4 estimate based on busulfan day 2 concentrations was significantly smaller than the variance of Clday4 estimates based on day 1 concentrations (p < 0.001). Therefore, day 1-guided TDM (pharmacometric model-based) of busulfan may be sufficient for attaining optimal target exposure, provided that subsequent TDM is carried out if required. However, performing TDM on subsequent days may be beneficial, as measurements on day 2 seemed to reduce the variance in the estimated clearance as compared to day 1 sampling.


Asunto(s)
Busulfano , Trasplante de Células Madre Hematopoyéticas , Niño , Humanos , Monitoreo de Drogas , Acondicionamiento Pretrasplante
5.
Front Pharmacol ; 13: 828094, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35370695

RESUMEN

Anti-thymocyte globulin (ATG), a polyclonal antibody, is used in allogeneic hematopoietic cell transplantation (HCT) to prevent graft-vs.-host-disease (GvHD) and graft failure (GF). Overexposure to ATG leads to poor early T-cell recovery, which is associated with viral infections and poor survival. Patients with severe inflammation are at high risk for GF and GvHD, and may have active infections warranting swift T-cell recovery. As ATG exposure may be critical in these patients, individualized dosing combined with therapeutic drug monitoring (TDM) may improve outcomes. We describe the individualized dosing approach, an optimal sampling scheme, the assay to measure the active fraction of ATG, and the workflow to perform TDM. Using a previously published population pharmacokinetic (PK) model, we determine the dose to reach optimal exposures associated with low GvHD and rejection, and at the same time promote T-cell recovery. Based on an optimal sampling scheme, peak and trough samples are taken during the first 3 days of once-daily dosing. The fraction of ATG able to bind to T-cells (active ATG) is analyzed using a bio-assay in which Jurkat cells are co-cultured with patient's plasma and the binding is quantified using flow cytometry. TDM is performed based on these ATG concentrations on the third day of dosing; subsequent doses can be adjusted based on the expected area under the curve. We show that individualized ATG dosing with TDM is feasible. This approach is unique in the setting of antibody treatment and may result in better immune reconstitution post-HCT and subsequently better survival chances.

6.
Bone Marrow Transplant ; 57(1): 38-42, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34608276

RESUMEN

Umbilical cord blood is the preferred donor cell source for children with Inherited Metabolic disorders undergoing Hematopoietic Cell Transplant (HCT), and its use has been associated with improved "engrafted survival" and higher donor chimerism compared to other cell sources. However, as in other pediatric cord blood transplants for non-malignant disease, immune-mediated cytopenia and primary graft failure limit its use, and the latter remains the commonest cause of death following cord blood transplant for non-malignant disease. We have previously shown an association between immune-mediated cytopenia and graft failure in inherited metabolic diseases suggesting that both immune-mediated cytopenia and graft failure could be mediated by antibodies from the residual recipient B cells. Since rituximab is effective in depletion of B cells and management of refractory immune-mediated cytopenia following HCT, we have added rituximab to the conditioning regimen. We studied 57 patients in 2 centers who received myeloablative conditioning for cord blood transplant in Hurler syndrome, and report a significant improvement in event-free survival with reduced incidence of graft failure and without any evidence of immune-mediated cytopenia in those patients that had received rituximab.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Mucopolisacaridosis I , Niño , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Rituximab/uso terapéutico , Acondicionamiento Pretrasplante/efectos adversos
7.
Orphanet J Rare Dis ; 15(1): 16, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941528

RESUMEN

The low prevalence of European paediatric transplanted patients and scarcity of resources and expertise led to the need for a multidisciplinary network able to improve the quality of life of paediatric patients and families requiring a solid organ or haematopoietic stem cell transplantation. The European Reference Network (ERN) TransplantChild is one of the 24 ERNs established in a European legal framework to improve the care of patients with rare diseases. ERN TransplantChild is the only ERN focused on both solid organ and haematopoietic stem cell paediatric transplantation, based on the understanding of paediatric transplantation as a complex and highly specialised process where specific complications appear regardless the organ involved, thus linking the skills and knowledge of different organ disciplines. Gathering European centres of expertise in paediatric transplantation will give access to a correct and timely diagnosis, share expertise and knowledge and collect a critical mass of patients and data that increases the speed and value of clinical research outcomes. Therefore, the ERN TransplantChild aims for a paediatric Pan-European, Pan-transplant approach.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Órganos/métodos , Europa (Continente) , Geografía , Humanos , Modelos Teóricos , Calidad de Vida , Procedimientos Quirúrgicos Operativos
8.
Monaldi Arch Chest Dis ; 71(2): 71-5, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19719039

RESUMEN

Here we report two children with a pulmonary inflammatory pseudotumour; a rare entity in children, that often initially presents as a pneumonia, but with the possibility of serious consequences if unrecognised and untreated. One of the children presented is 6 months which is extremely young for this tumour. Difficulties in presentation, management strategies and prognosis are described. Certainly, this is a condition that should be considered even in a very young child with an inflammatory condition presenting as a solid lesion in the lung which does not resolve or even progresses.


Asunto(s)
Granuloma de Células Plasmáticas del Pulmón/diagnóstico , Granuloma de Células Plasmáticas del Pulmón/cirugía , Niño , Medios de Contraste , Femenino , Humanos , Lactante , Pulmón/diagnóstico por imagen , Pulmón/patología , Pulmón/cirugía , Masculino , Tomografía Computarizada por Rayos X/métodos , Ultrasonografía
9.
Blood Adv ; 3(14): 2179-2187, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31324638

RESUMEN

Fludarabine is the most frequently used agent in conditioning regimens for allogeneic hematopoietic cell transplantation (HCT). Body surface area-based dosing leads to highly variable fludarabine exposure. We studied the relation between fludarabine exposure and clinical outcomes. A retrospective, pharmacokinetic-pharmacodynamic analysis was conducted with data from patients undergoing HCT with fludarabine (160 mg/m2) as part of a myeloablative conditioning (busulfan targeted to an area under the plasma-concentration-time curve [AUC] of 90 mg*h/L) and rabbit antithymocyte globulin (6-10 mg/kg; from day -9/-12) between 2010 and 2016. Fludarabine exposure as AUC was calculated for each patient using a previously published population pharmacokinetic model and related to 2-year event-free survival (EFS) by means of (parametric) time-to-event models. Relapse, nonrelapse mortality (NRM), and graft failure were considered events. One hundred ninety-two patients were included (68 benign and 124 malignant disorders). The optimal fludarabine exposure was determined as an AUC of 20 mg*h/L. In the overexposed group, EFS was lower (hazard ratio [HR], 2.0; 95% confidence interval [CI], 1.1-3.5; P = .02), due to higher NRM (HR, 3.4; 95% CI, 1.6-6.9; P <001) associated with impaired immune reconstitution (HR, 0.43; 95% CI, 0.26-0.70; P <001). The risks of NRM and graft failure were increased in the underexposed group (HR, 3.3; 95% CI, 1.2-9.4; P = .02; HR, 4.8; 95% CI, 1.2-19; P = .02, respectively). No relationship with relapse was found. Fludarabine exposure is a strong predictor of survival after HCT, stressing the importance of optimum fludarabine dosing. Individualized dosing, based on weight and "renal function" or "therapeutic drug monitoring," to achieve optimal fludarabine exposure might improve survival.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Agonistas Mieloablativos/uso terapéutico , Acondicionamiento Pretrasplante , Vidarabina/análogos & derivados , Adolescente , Adulto , Causas de Muerte , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Masculino , Persona de Mediana Edad , Mortalidad , Agonistas Mieloablativos/administración & dosificación , Agonistas Mieloablativos/efectos adversos , Agonistas Mieloablativos/farmacocinética , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Acondicionamiento Pretrasplante/efectos adversos , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Vidarabina/administración & dosificación , Vidarabina/efectos adversos , Vidarabina/farmacocinética , Vidarabina/uso terapéutico , Adulto Joven
10.
Sci Immunol ; 4(42)2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31811055

RESUMEN

Despite the importance of intestinal stem cells (ISCs) for epithelial maintenance, there is limited understanding of how immune-mediated damage affects ISCs and their niche. We found that stem cell compartment injury is a shared feature of both alloreactive and autoreactive intestinal immunopathology, reducing ISCs and impairing their recovery in T cell-mediated injury models. Although imaging revealed few T cells near the stem cell compartment in healthy mice, donor T cells infiltrating the intestinal mucosa after allogeneic bone marrow transplantation (BMT) primarily localized to the crypt region lamina propria. Further modeling with ex vivo epithelial cultures indicated ISC depletion and impaired human as well as murine organoid survival upon coculture with activated T cells, and screening of effector pathways identified interferon-γ (IFNγ) as a principal mediator of ISC compartment damage. IFNγ induced JAK1- and STAT1-dependent toxicity, initiating a proapoptotic gene expression program and stem cell death. BMT with IFNγ-deficient donor T cells, with recipients lacking the IFNγ receptor (IFNγR) specifically in the intestinal epithelium, and with pharmacologic inhibition of JAK signaling all resulted in protection of the stem cell compartment. In addition, epithelial cultures with Paneth cell-deficient organoids, IFNγR-deficient Paneth cells, IFNγR-deficient ISCs, and purified stem cell colonies all indicated direct targeting of the ISCs that was not dependent on injury to the Paneth cell niche. Dysregulated T cell activation and IFNγ production are thus potent mediators of ISC injury, and blockade of JAK/STAT signaling within target tissue stem cells can prevent this T cell-mediated pathology.


Asunto(s)
Interferón gamma/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Animales , Muerte Celular , Mucosa Intestinal/patología , Ratones
11.
Bone Marrow Transplant ; 52(2): 270-278, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27991895

RESUMEN

Patients with prior invasive fungal infection (IFI) increasingly proceed to allogeneic hematopoietic cell transplantation (HSCT). However, little is known about the impact of prior IFI on survival. Patients with pre-transplant IFI (cases; n=825) were compared with controls (n=10247). A subset analysis assessed outcomes in leukemia patients pre- and post 2001. Cases were older with lower performance status (KPS), more advanced disease, higher likelihood of AML and having received cord blood, reduced intensity conditioning, mold-active fungal prophylaxis and more recently transplanted. Aspergillus spp. and Candida spp. were the most commonly identified pathogens. 68% of patients had primarily pulmonary involvement. Univariate and multivariable analysis demonstrated inferior PFS and overall survival (OS) for cases. At 2 years, cases had higher mortality and shorter PFS with significant increases in non-relapse mortality (NRM) but no difference in relapse. One year probability of post-HSCT IFI was 24% (cases) and 17% (control, P<0.001). The predominant cause of death was underlying malignancy; infectious death was higher in cases (13% vs 9%). In the subset analysis, patients transplanted before 2001 had increased NRM with inferior OS and PFS compared with later cases. Pre-transplant IFI is associated with lower PFS and OS after allogeneic HSCT but significant survivorship was observed. Consequently, pre-transplant IFI should not be a contraindication to allogeneic HSCT in otherwise suitable candidates. Documented pre-transplant IFI is associated with lower PFS and OS after allogeneic HSCT. However, mortality post transplant is more influenced by advanced disease status than previous IFI. Pre-transplant IFI does not appear to be a contraindication to allogeneic HSCT.


Asunto(s)
Aspergilosis , Aspergillus , Candida , Candidiasis , Trasplante de Células Madre de Sangre del Cordón Umbilical , Neoplasias Hematológicas , Sistema de Registros , Adolescente , Adulto , Anciano , Aloinjertos , Aspergilosis/etiología , Aspergilosis/mortalidad , Aspergilosis/terapia , Candidiasis/etiología , Candidiasis/mortalidad , Candidiasis/terapia , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/terapia , Humanos , Lactante , Masculino , Persona de Mediana Edad , Tasa de Supervivencia
12.
Bone Marrow Transplant ; 51(8): 1113-20, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27042847

RESUMEN

Several studies have reported an association between CMV reactivation and a decreased incidence of relapse for AML after adult donor allogeneic hematopoietic cell transplantation (HCT). Limited data, however, are available on the impact of CMV reactivation on relapse after cord blood (CB) stem cell transplantation. The unique combination of higher incidence of CMV reactivation in the seropositive recipient and lower incidence of graft versus host disease (GvHD) in CB HCT permits a valuable design to analyze the impact of CMV reactivation. Data from 1684 patients transplanted with CB between 2003 and 2010 for AML and ALL were analyzed. The median time to CMV reactivation was 34 days (range: 2-287). CMV reactivation and positive CMV serology were associated with increased non-relapse mortality (NRM) among both AML and ALL CB recipients (reactivation, AML: relative risk (RR) 1.41 (1.07-1.85); ALL: 1.60 (1.14-2.23); Serology, AML: RR 1.39 (1.05-1.85), ALL: RR 1.61 (1.18-2.19)). For patients with ALL, but not those with AML, this yielded inferior overall survival (P<0.005). Risk of relapse was not influenced by CMV reactivation or positive CMV serostatus for either disease.


Asunto(s)
Citomegalovirus/fisiología , Leucemia Mieloide Aguda/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Activación Viral , Adolescente , Adulto , Anciano , Niño , Preescolar , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Trasplante de Células Madre de Sangre del Cordón Umbilical/mortalidad , Femenino , Enfermedad Injerto contra Huésped , Humanos , Lactante , Leucemia Mieloide Aguda/complicaciones , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Recurrencia , Estudios Retrospectivos , Análisis de Supervivencia , Adulto Joven
13.
Bone Marrow Transplant ; 51(4): 573-80, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26726945

RESUMEN

Pneumocystis jiroveci pneumonia (PJP) is associated with high morbidity and mortality after hematopoietic stem cell transplantation (HSCT). Little is known about PJP infections after HSCT because of the rarity of disease given routine prophylaxis. We report the results of a Center for International Blood and Marrow Transplant Research study evaluating the incidence, timing, prophylaxis agents, risk factors and mortality of PJP after autologous (auto) and allogeneic (allo) HSCT. Between 1995 and 2005, 0.63% allo recipients and 0.28% auto recipients of first HSCT developed PJP. Cases occurred as early as 30 days to beyond a year after allo HSCT. A nested case cohort analysis with supplemental data (n=68 allo cases, n=111 allo controls) revealed that risk factors for PJP infection included lymphopenia and mismatch after HSCT. After allo or auto HSCT, overall survival was significantly poorer among cases vs controls (P=0.0004). After controlling for significant variables, the proportional hazards model revealed that PJP cases were 6.87 times more likely to die vs matched controls (P<0.0001). We conclude PJP infection is rare after HSCT but is associated with high mortality. Factors associated with GVHD and with poor immune reconstitution are among the risk factors for PJP and suggest that protracted prophylaxis for PJP in high-risk HSCT recipients may improve outcomes.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Pneumocystis carinii , Neumonía por Pneumocystis , Aloinjertos , Autoinjertos , Femenino , Humanos , Incidencia , Masculino , Neumonía por Pneumocystis/etiología , Neumonía por Pneumocystis/mortalidad , Neumonía por Pneumocystis/prevención & control , Factores de Riesgo
14.
Leukemia ; 29(9): 1839-46, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25836589

RESUMEN

We performed a prospective phase II study to evaluate clinical safety and outcome in 48 patients with steroid-refractory grade II-IV acute graft-versus-host disease (aGVHD) treated with mesenchymal stromal cells (MSCs). Clinical outcomes were correlated to comprehensive analyses of soluble and cellular biomarkers. Complete resolution (CR) of aGVHD at day 28 (CR-28) occurred in 12 (25%) patients, CR lasting >1 month (CR-B) occurred in 24 (50%) patients. One-year overall survival was significantly improved in CR-28 (75 versus 33%, P=0.020) and CR-B (79 versus 8%, P<0.001) versus non-CR patients. A six soluble biomarker-panel was predictive for mortality (HR 2.924; CI 1.485-5.758) when measured before MSC-administration. Suppression of tumorigenicity 2 (ST2) was only predictive for mortality 2 weeks after but not before MSC-administration (HR 2.389; CI 1.144-4.989). In addition, an increase in immature myeloid dendritic cells associated with decreased mortality (HR 0.554, CI 0.389-0.790). Patients had persisting T-cell responses against defined virus- and leukemia-associated antigens. In conclusion, our data emphasize the need to carefully assess biomarkers in cohorts with homogeneous GVHD treatments. Biomarkers might become an additional valuable component of composite end points for the rapid and efficient testing of novel compounds to decrease lifecycle of clinical testing and improve the success rate of phase II/III trials.


Asunto(s)
Resistencia a Medicamentos , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Enfermedad Aguda , Adolescente , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Biomarcadores/sangre , Biomarcadores/metabolismo , Niño , Preescolar , Ciclosporina/uso terapéutico , Citocinas/sangre , Citocinas/metabolismo , Femenino , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/mortalidad , Humanos , Inmunofenotipificación , Inmunosupresores/uso terapéutico , Lactante , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Persona de Mediana Edad , Prednisona/uso terapéutico , Pronóstico , Esteroides/uso terapéutico , Resultado del Tratamiento , Adulto Joven
15.
Adv Exp Med Biol ; 171: 209-39, 1984.
Artículo en Inglés | MEDLINE | ID: mdl-6609542

RESUMEN

The cross-sectional study of patients with RA receiving LDGC, compared with those on NSAID alone (or patients with AS) showed that LDGC significantly affects bone mass at midshaft and even more so at the distal radius. The loss of bone seems to be brisk but continuous on the long run, at least at the distal scanning site, and thus increases the C/T ratio, especially in aged men. The loss of bone mass in the LDGC group correlates with the duration of the disease as well as with carpal destruction (especially at mid shaft radius), with both parameters being correlated with one another. At equal carpal destruction, LDGC still affects bone mass. Whether receiving NSAID alone or LDGC in addition, patients with RA, as compared with controls, are more liable to lose bone when they grow older. In a longitudinal study, premenopausal women were unaffected by the administration of LDGC at both scanning sites. In contrast, postmenopausal women receiving LDGC lost at least twice as much bone as did normal women after the menopause. Men of all ages on LDGC lost bone at a rate equal to that of normal women after the menopause. Men with RA or with AS on NSAID alone did not significantly lose bone. It is concluded that LDGC may be given to premenopausal women without harm to their bone mass. After the menopause, hormonal replacement therapy, if not contra-indicated, should be given in association with LDGC. Men fortunately have a higher peak bone mass and therefore can afford to lose bone during a decade before they attain the same situation as women at the time of their menopause. If treatment is then continued for another two decades, their bone mass might behave as does that of postmenopausal women if bone loss is continuous over such long periods of time. This latter assumption has yet to be verified.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Huesos/análisis , Glucocorticoides/efectos adversos , Minerales/análisis , Adulto , Anciano , Antiinflamatorios/uso terapéutico , Artritis Reumatoide/metabolismo , Estudios Transversales , Femenino , Glucocorticoides/administración & dosificación , Glucocorticoides/uso terapéutico , Humanos , Estudios Longitudinales , Masculino , Menopausia , Métodos , Persona de Mediana Edad , Radio (Anatomía)/análisis , Radio (Anatomía)/anatomía & histología
16.
Ann Endocrinol (Paris) ; 40(2): 173-4, 1979.
Artículo en Francés | MEDLINE | ID: mdl-475313

RESUMEN

Two cases with vitamin D-deficient osteomalacia of digestive origin (coeliac disease) have been treated orally with 1,25(OH)2D3 in dosages of respectively 8 and 3 microgram/24 h. Serial transiliac bone biopsies have been performed. Mineralization fronts were normalized after respectively 4 and 1.5 months, whereas mineralization rates, as demonstrated by double tetracycline labeling, were normalized respectively after 2 and 1.5 months. In both cases osteomalacia healed completely despite persistently low 25(OH)D levels in the serum. It is obvious from these studies that 1,25(OH)2D3 per se is able to cure vitamin D-deficient osteomalacia, a finding that has been disputed. Whether 25(OH)D acts more rapidly on bone, as suggested by the studies performed in one case with anticonvulsant osteomalacia, remains to be proven.


Asunto(s)
Huesos/fisiopatología , Enfermedad Celíaca/tratamiento farmacológico , Dihidroxicolecalciferoles/uso terapéutico , Hidroxicolecalciferoles/uso terapéutico , Osteomalacia/tratamiento farmacológico , Deficiencia de Vitamina D/tratamiento farmacológico , Enfermedad Celíaca/complicaciones , Humanos , Osteomalacia/complicaciones , Deficiencia de Vitamina D/complicaciones
17.
Bone Marrow Transplant ; 49(1): 95-101, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24121212

RESUMEN

This study was aimed at finding predictors of invasive fungal infection (IFI) after pediatric allogeneic hematopoietic SCT (HSCT). All children who received allogeneic HSCT in the Wilhelmina Children's Hospital Utrecht between 2004 and 2012 were included. HSCT data were prospectively collected. Patients were retrospectively classified into high- or low-risk groups for developing IFI using criteria based on available literature. Predictors for the occurrence of IFI were analyzed using Cox regression models. We used logistic regression models to analyze the association between other HSCT-related complications and IFI. Secondary outcomes were overall survival and treatment-related mortality (TRM). Two-hundred nine patients were included in the analysis; median age was 6.6 years. The cumulative incidence of IFI was 12%. In patients classified as 'low risk' (n=75), only 5.3% developed IFI (odds ratio (OR): 0.325; P=0.047). In multivariate analysis, a predictor for the occurrence of IFI was an a priori determined HSCT TRM risk >20% (based on EBMT-risk score). Post-HSCT, the administration of high-dose steroids was associated with IFI (OR: 4.458; P=0.010). Patients who developed IFI showed an increased risk of TRM (OR: 3.773; P=0.004). These results confirm that risk group stratification should guide intensity of monitoring for IFI and use of antifungal prophylaxis.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Micosis/complicaciones , Micosis/diagnóstico , Adolescente , Adulto , Anfotericina B/uso terapéutico , Antifúngicos/uso terapéutico , Aspergilosis/complicaciones , Candidiasis/complicaciones , Caspofungina , Niño , Preescolar , Equinocandinas/uso terapéutico , Femenino , Fusariosis/complicaciones , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Humanos , Lactante , Lipopéptidos , Modelos Logísticos , Masculino , Neutrófilos/citología , Estudios Prospectivos , Pirimidinas/uso terapéutico , Estudios Retrospectivos , Riesgo , Resultado del Tratamiento , Triazoles/uso terapéutico , Voriconazol , Adulto Joven
18.
Leukemia ; 27(6): 1328-38, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23277330

RESUMEN

Human cytomegalovirus (CMV) infections and relapse of disease remain major problems after allogeneic stem cell transplantation (allo-SCT), in particular in combination with CMV-negative donors or cordblood transplantations. Recent data suggest a paradoxical association between CMV reactivation after allo-SCT and reduced leukemic relapse. Given the potential of Vδ2-negative γδT cells to recognize CMV-infected cells and tumor cells, the molecular biology of distinct γδT-cell subsets expanding during CMV reactivation after allo-SCT was investigated. Vδ2(neg) γδT-cell expansions after CMV reactivation were observed not only with conventional but also cordblood donors. Expanded γδT cells were capable of recognizing both CMV-infected cells and primary leukemic blasts. CMV and leukemia reactivity were restricted to the same clonal population, whereas other Vδ2(neg) T cells interact with dendritic cells (DCs). Cloned Vδ1 T-cell receptors (TCRs) mediated leukemia reactivity and DC interactions, but surprisingly not CMV reactivity. Interestingly, CD8αα expression appeared to be a signature of γδT cells after CMV exposure. However, functionally, CD8αα was primarily important in combination with selected leukemia-reactive Vδ1 TCRs, demonstrating for the first time a co-stimulatory role of CD8αα for distinct γδTCRs. Based on these observations, we advocate the exploration of adoptive transfer of unmodified Vδ2(neg) γδT cells after allo-SCT to tackle CMV reactivation and residual leukemic blasts, as well as application of leukemia-reactive Vδ1 TCR-engineered T cells as alternative therapeutic tools.


Asunto(s)
Citomegalovirus/fisiología , Leucemia/cirugía , Trasplante de Células Madre , Linfocitos T/inmunología , Activación Viral , Humanos , Leucemia/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA