Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 180(3): 346-355, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29076145

RESUMEN

Allogeneic haematopoietic stem-cell transplantation remains the only curative treatment for relapsed/refractory acute myeloid leukaemia (AML) and high-risk myelodysplasia but has previously been limited to patients who achieve remission before transplant. New sequential approaches employing T-cell depleted transplantation directly after chemotherapy show promise but are burdened by viral infection and require donor lymphocyte infusions (DLI) to augment donor chimerism and graft-versus-leukaemia effects. T-replete transplantation in sequential approaches could reduce both viral infection and DLI usage. We therefore performed a single-arm prospective Phase II clinical trial of sequential chemotherapy and T-replete transplantation using reduced-intensity conditioning without planned DLI. The primary endpoint was overall survival. Forty-seven patients with relapsed/refractory AML or high-risk myelodysplasia were enrolled; 43 proceeded to transplantation. High levels of donor chimerism were achieved spontaneously with no DLI. Overall survival of transplanted patients was 45% and 33% at 1 and 3 years. Only one patient developed cytomegalovirus disease. Cumulative incidences of treatment-related mortality and relapse were 35% and 20% at 1 year. Patients with relapsed AML and myelodysplasia had the most favourable outcomes. Late-onset graft-versus-host disease protected against relapse. In conclusion, a T-replete sequential transplantation using reduced-intensity conditioning is feasible for relapsed/refractory AML and myelodysplasia and can deliver graft-versus-leukaemia effects without DLI.


Asunto(s)
Efecto Injerto vs Leucemia/inmunología , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/terapia , Adulto , Anciano , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/mortalidad , Depleción Linfocítica , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/mortalidad , Recurrencia , Inducción de Remisión , Quimera por Trasplante , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Resultado del Tratamiento , Activación Viral , Adulto Joven
2.
Mol Ther ; 24(6): 1126-1134, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27049761

RESUMEN

Cell therapy with antigen-specific regulatory T-cells (Treg) has great potential to selectively control unwanted immune responses after allogeneic stem-cell or solid organ transplantation and in autoimmune diseases. Ex vivo allostimulation with costimulatory blockade (alloanergization) of human T-cells expands populations of alloantigen-specific Treg, providing a cellular strategy to control donor T-cell alloresponses causing graft-versus-host disease after allogeneic hematopoietic stem-cell transplantation. Crucially, it is not known if Treg expanded in this way are stable in proinflammatory conditions encountered after transplantation, or if they possess capacity to migrate to key target organs. Using an in vitro model to functionally characterize human Treg expanded after alloanergization, we now show that these cells remain potently allosuppressive in the presence of relevant exogenous inflammatory signals. Expanded allospecific Treg retained expression of molecules conferring migratory capacity to several organs but small intestine-specific chemotaxis was markedly impaired, in keeping with the preponderance of gut graft-versus-host disease in previous clinical studies using this strategy. Importantly, impaired gut-specific chemotaxis could be partially corrected by pharmacological treatment. These findings will facilitate more effective application of this cellular approach to limit T-cell alloresponses after hematopoietic stem-cell transplantation and the wider application of the strategy to other clinical settings.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Isoantígenos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Leucocitos Mononucleares/inmunología , Linfocitos T Reguladores/metabolismo
3.
Proc Natl Acad Sci U S A ; 110(33): 13576-81, 2013 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-23901108

RESUMEN

Acute myeloid leukemia (AML) induces bone marrow (BM) failure in patients, predisposing them to life-threatening infections and bleeding. The mechanism by which AML mediates this complication is unknown but one widely accepted explanation is that AML depletes the BM of hematopoietic stem cells (HSCs) through displacement. We sought to investigate how AML affects hematopoiesis by quantifying residual normal hematopoietic subpopulations in the BM of immunodeficient mice transplanted with human AML cells with a range of genetic lesions. The numbers of normal mouse HSCs were preserved whereas normal progenitors and other downstream hematopoietic cells were reduced following transplantation of primary AMLs, findings consistent with a differentiation block at the HSC-progenitor transition, rather than displacement. Once removed from the leukemic environment, residual normal hematopoietic cells differentiated normally and outcompeted steady-state hematopoietic cells, indicating that this effect is reversible. We confirmed the clinical significance of this by ex vivo analysis of normal hematopoietic subpopulations from BM of 16 patients with AML. This analysis demonstrated that the numbers of normal CD34(+)CD38(-) stem-progenitor cells were similar in the BM of AML patients and controls, whereas normal CD34(+)CD38(+) progenitors were reduced. Residual normal CD34(+) cells from patients with AML were enriched in long-term culture, initiating cells and repopulating cells compared with controls. In conclusion the data do not support the idea that BM failure in AML is due to HSC depletion. Rather, AML inhibits production of downstream hematopoietic cells by impeding differentiation at the HSC-progenitor transition.


Asunto(s)
Células de la Médula Ósea/patología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Leucemia Mieloide Aguda/fisiopatología , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Antígenos CD34/metabolismo , Proliferación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Microscopía Confocal , Estadísticas no Paramétricas
4.
Br J Haematol ; 160(2): 133-45, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23121307

RESUMEN

A major challenge to transplant immunologists and physicians remains the separation of harmful graft-versus-host disease (GvHD) and beneficial graft-versus-tumour (GvT) effects after allogeneic haematopoietic stem cell transplantation. Recent advances in our understanding of the allogeneic immune response provide potential new opportunities to achieve this goal. Three potential new approaches that capitalize on this new knowledge are considered in depth; the manipulation of organ-specific cytokines and other pro-inflammatory signals, the selective manipulation of donor effector T cell migration, and the development of cell-mediated immunosuppressive strategies using donor-derived regulatory T cells. These new approaches could provide strategies for local control of allogeneic immune responses, a new paradigm to separate GvHD and GvT effects. Although these strategies are currently in their infancy and have challenges to successful translation to clinical practice, all have exciting potential for the future.


Asunto(s)
Refuerzo Inmunológico de Injertos/métodos , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Tumor , Trasplante de Células Madre Hematopoyéticas , Trasplante Homólogo/inmunología , Inmunidad Adaptativa , Animales , Antígenos de Neoplasias/administración & dosificación , Movimiento Celular , Ensayos Clínicos como Asunto , Citocinas/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Tumor/efectos de los fármacos , Efecto Injerto vs Tumor/inmunología , Humanos , Inmunidad Innata , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Inflamación/inmunología , Inflamación/prevención & control , Transfusión de Linfocitos , Ratones , Modelos Inmunológicos , Especificidad de Órganos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/trasplante , Investigación Biomédica Traslacional/tendencias , Acondicionamiento Pretrasplante/efectos adversos , Acondicionamiento Pretrasplante/métodos , Vacunación
5.
Br J Haematol ; 162(4): 525-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23718277

RESUMEN

The toxicity burden and long-term anti-leukaemic effect of non-myeloablative (NMA) allogeneic haematopoietic stem-cell transplantation (AHSCT) for acute myeloid leukaemia (AML) and myelodysplasia (MDS) remains undefined. We report the outcome of 56 patients with AML/MDS transplanted from human leucocyte antigen-matched donors using NMA conditioning without T-cell depletion. With a median follow-up of 5 years, treatment-related mortality was 9% and current disease-free survival (CDFS) was 45% (overall) and 55% (patients transplanted in remission). Development of graft-versus-host disease upon withdrawal of post-transplant immunosuppression was associated with less relapse and better CDFS. These data confirm that NMA AHSCT without T-cell depletion is safe and can result in sustained remissions of AML/MDS.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/cirugía , Depleción Linfocítica , Síndromes Mielodisplásicos/cirugía , Linfocitos T , Acondicionamiento Pretrasplante/métodos , Adulto , Infecciones por Citomegalovirus/epidemiología , Supervivencia sin Enfermedad , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/epidemiología , Humanos , Terapia de Inmunosupresión , Estimación de Kaplan-Meier , Tiempo de Internación , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Trastornos Linfoproliferativos/epidemiología , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/mortalidad , Complicaciones Posoperatorias/epidemiología , Sepsis/epidemiología , Acondicionamiento Pretrasplante/mortalidad , Trasplante Homólogo , Adulto Joven
6.
Br J Haematol ; 157(5): 580-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22449197

RESUMEN

The relative merits of reduced-intensity allogeneic stem cell transplantation (RISCT) for high-risk indolent lymphoid malignancies are emerging, although the preferred conditioning regimen to manage the risks of graft-versus-host disease (GVHD) is not clearly defined. Here we report the outcome of 73 patients with lymphoid malignancies who received RISCT with a fludarabine/cyclophosphosphamide conditioning regimen and a median follow-up of 3 years. Median age was 54 years. Forty-eight per cent of patients had previously undergone autologous stem cell transplantation with a median of three prior therapies. Non-relapse mortality at 3 years was 19% but only 5% for patients with multiple myeloma (MM). Three-year overall survival and current progression-free survival was 67% and 63% respectively. Grade 2-4 acute GVHD occurred in 14% of patients while 49% had chronic GVHD requiring systemic immunosuppression. The preparatory regimen in this study has the advantage of reduced acute GVHD and low mortality, notably in patients with MM. In addition, this strategy provides long-term disease control in a significant proportion of patients with particular benefit in those with high-risk follicular lymphoma.


Asunto(s)
Ciclofosfamida/administración & dosificación , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Trastornos Linfoproliferativos/terapia , Acondicionamiento Pretrasplante , Vidarabina/análogos & derivados , Adulto , Anciano , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/mortalidad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/mortalidad , Persona de Mediana Edad , Recurrencia , Análisis de Supervivencia , Linfocitos T/inmunología , Trasplante Homólogo , Vidarabina/administración & dosificación
7.
Anticancer Res ; 41(3): 1123-1141, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33788704

RESUMEN

Haematology has been at the vanguard of cancer immunotherapy. Immune checkpoint inhibitors (ICIs), bispecific T-cell engagers (BiTEs), allogeneic haematopoietic stem cell transplantation (allo-HSCT) and donor lymphocyte infusion (DLI), as well as adoptive T-cell therapies outside the setting of allo-HSCT, have been approved for distinct haematologic malignancies producing durable responses in otherwise untreatable patients. Despite recent advances, immunotherapies do not benefit most patients, due to resistance or lack of response, and are only approved in specific settings. Moreover, immunotherapies are expensive and may produce severe immune related adverse reactions. Combination therapy complicates the picture and requires further evaluation. This review considers the current status and future perspectives of ICIs and BiTEs approved for haematological malignancies by analysing their strengths, weaknesses, opportunities and threats (SWOT). The biological rationale for anti-cancer mechanisms, clinical data for specific haematological cancers, efficacy, toxicity, response and resistance profiles, novel strategies to improve these characteristics as well as the potential targets to enhance or expand the application of ICIs and BiTEs are also discussed.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Linfocitos T/inmunología , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/uso terapéutico , Antígeno CTLA-4/fisiología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/farmacología , Receptor de Muerte Celular Programada 1/fisiología
8.
Anticancer Res ; 41(3): 1143-1156, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33788705

RESUMEN

Haematology has been at the forefront of cancer immunotherapy advancements. Allogeneic haematopoietic stem cell transplant (allo-HSCT) is one of the earliest forms of cancer immunotherapy and continues to cure thousands of patients. Donor lymphocyte infusion (DLI) increases allo-HSCT efficacy and reduces graft-versus-host disease (GVHD). In recent years, chimeric antigen receptor (CAR)-T-cells have been approved for the treatment of distinct haematologic malignancies, producing durable response in otherwise untreatable patients. New target antigen identification and technological advances have enabled the structural and functional evolution of CARs, broadening their applications. Despite successes, adoptive T-cell (ATC) therapies are expensive, can cause severe adverse reactions and their use is restricted to few patients. This review considers the current status and future perspectives of allogeneic transplant and donor lymphocytes, as well as novel ATC therapies, such as CAR-T-cells in haematological malignancies by analysing their strengths, weaknesses, opportunities, and threats (SWOT). The biological rationale for anti-cancer mechanisms and development; current clinical data in specific haematological malignancies; efficacy, toxicity, response and resistance profiles; novel strategies to improve these characteristics; and potential targets to enhance or expand the application of these therapies are discussed.


Asunto(s)
Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunoterapia Adoptiva/métodos , Inmunoterapia/métodos , Linfocitos T/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD19/uso terapéutico , Productos Biológicos , Humanos , Inmunoterapia Adoptiva/efectos adversos , Transfusión de Linfocitos , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/inmunología , Donantes de Tejidos
10.
Blood ; 112(6): 2232-41, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18617635

RESUMEN

We report the outcomes of 24 patients with high-risk hematologic malignancies or bone marrow failure (BMF) who received haploidentical bone marrow transplantation (BMT) after ex vivo induction of alloantigen-specific anergy in donor T cells by allostimulation in the presence of costimulatory blockade. Ninety-five percent of evaluable patients engrafted and achieved full donor chimerism. Despite receiving a median T-cell dose of 29 x10(6)/kg, only 5 of 21 evaluable patients developed grade C (n = 4) or D (n = 1) acute graft-versus-host disease (GVHD), with only one attributable death. Twelve patients died from treatment-related mortality (TRM). Patients reconstituted T-cell subsets and immunoglobulin levels rapidly with evidence of in vivo expansion of pathogen-specific T cells in the early posttransplantation period. Five patients reactivated cytomegalovirus (CMV), only one of whom required extended antiviral treatment. No deaths were attributable to CMV or other viral infections. Only 1 of 12 evaluable patients developed chronic GVHD. Eight patients survive disease-free with normal performance scores (median follow-up, 7 years). Thus, despite significant early TRM, ex vivo alloanergization can support administration of large numbers of haploidentical donor T cells, resulting in rapid immune reconstitution with very few viral infections. Surviving patients have excellent performance status and a low rate of chronic GVHD.


Asunto(s)
Trasplante de Médula Ósea/métodos , Anergia Clonal/inmunología , Transfusión de Linfocitos/métodos , Linfocitos T/inmunología , Adolescente , Adulto , Enfermedades de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/terapia , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/mortalidad , Niño , Preescolar , Infecciones por Citomegalovirus , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped , Haplotipos , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/terapia , Humanos , Lactante , Isoantígenos , Masculino , Persona de Mediana Edad , Tasa de Supervivencia , Quimera por Trasplante , Resultado del Tratamiento
11.
Leukemia ; 33(6): 1540, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30903015

RESUMEN

In the original version of this article the authors noted an omission in the author affiliations where the university details: Queen Mary University of London was not included in the original affiliation for the majority of the authors. The correct affiliations are as follows1. Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK3. Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK6. Evolution and Cancer Laboratory, Barts Cancer Institute, Queen Mary University of London, London, UK.

12.
Clin Cancer Res ; 24(17): 4098-4109, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29769208

RESUMEN

Purpose: Allogeneic hematopoietic stem-cell transplantation (HSCT) is a curative treatment for many hematologic cancers. Use of haploidentical (mismatched) donors increases HSCT availability but is limited by severe graft-versus-host disease (GvHD) and delayed immune reconstitution. Alloanergization of donor T cells is a simple approach to rebuild immunity while limiting GvHD after haploidentical HSCT, but the optimal T-cell dose and impact on immune reconstitution remain unknown.Patients and Methods: We performed a multicenter phase I trial of alloanergized donor lymphocyte infusion (aDLI) after CD34-selected myeloablative haploidentical HSCT. The primary aim was feasibility and safety with secondary aims of assessing the less frequently addressed issue of impact on immune reconstitution.Results: Nineteen patients with high-risk acute leukemia or myelodysplasia were enrolled. Engraftment occurred in 18 of 19 patients (95%). Pre-aDLI, 12 patients (63%) had bacteremia, nine of 17 at-risk patients (53%) reactivated CMV, and one developed acute GvHD. Sixteen patients received aDLI at dose levels 1 (103 T cells/kg, n = 4), 2 (104, n = 8), and 3 (105, n = 4). After aDLI, five patients developed clinically significant acute GvHD, and four of 14 at-risk patients (29%) reactivated CMV. T-cell recovery was significantly greater, and functional virus- and tumor-associated antigen-specific T cells were detectable earlier in patients receiving dose level 2 or 3 versus dose level 1/no aDLI. Alloanergization of donor cells expanded the CD4+ T-regulatory cell frequency within aDLI, which increased further in vivo without impeding expansion of virus- and tumor-associated antigen-specific T cells.Conclusions: These data demonstrate safety and a potential role for aDLI in contributing to immune reconstitution and expanding tolerogenic regulatory T cells in vivo after CD34-selected myeloablative haploidentical HSCT. Clin Cancer Res; 24(17); 4098-109. ©2018 AACR.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia/terapia , Linfocitos/inmunología , Síndromes Mielodisplásicos/terapia , Linfocitos T/inmunología , Adulto , Antígenos CD34/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Inmunidad Innata/inmunología , Leucemia/inmunología , Leucemia/patología , Transfusión de Linfocitos , Masculino , Persona de Mediana Edad , Agonistas Mieloablativos/administración & dosificación , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/patología , Linfocitos T/patología , Donantes de Tejidos , Acondicionamiento Pretrasplante , Trasplante Haploidéntico , Trasplante Homólogo
14.
Cell Transplant ; 21(9): 2047-61, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22507909

RESUMEN

Ex vivo alloanergization of human immune cells, via allostimulation in the presence of costimulatory blockade with either a combination of anti-B7.1 and anti-B7.2 antibodies or first-generation cytotoxic T-lymphocyte antigen 4-immunoglobulin (CTLA4-Ig), induces alloantigen-specific hyporesponsiveness and expands alloantigen-specific regulatory T cells (Treg). We have successfully used this approach in the clinical setting of haploidentical hematopoietic stem cell transplantation. Recently, the in vivo use of a new second-generation CTLA4-Ig, belatacept, has shown promise in controlling alloresponses after transplantation of both human kidneys and islet cells. We therefore compared the efficiency of first- and second-generation CTLA4-Ig in alloanergizing human peripheral blood mononuclear cells (PBMCs) and investigated whether ex vivo alloanergization with belatacept could be used to engineer an alloantigen-specific immunoregulatory population of autologous cells suitable for administration to recipients of cellular or solid organ transplant recipients. Alloanergization of HLA-mismatched human PBMCs with belatacept resulted in a greater reduction in subsequent alloresponses than alloanergization with first generation CTLA4-Ig. Moreover, subsequent ex vivo re-exposure of alloanergized cells to alloantigen in the absence of belatacept resulted in a significant expansion of Tregs with enhanced alloantigen-specific suppressive function. Alloanergized PBMCs retained functional Epstein-Barr virus (EBV)-specific T-cell responses, and expanded Tregs did not suppress EBV-specific proliferation of autologous cells. These results suggest that ex vivo alloanergization with belatacept provides a platform to engineer populations of recipient Treg with specificity for donor alloantigens but without nonspecific suppressive capacity. The potential advantages of such cells for solid organ transplantation include (1) reduction of the need for nonspecific immunosuppression, (2) retention of pathogen-specific immunity, and (3) control of graft rejection, if used as an intervention.


Asunto(s)
Inmunoconjugados/administración & dosificación , Inmunosupresores/administración & dosificación , Isoantígenos/inmunología , Abatacept , Epítopos , Antígenos HLA/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Inmunología del Trasplante
15.
J Vis Exp ; (49)2011 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-21445041

RESUMEN

Allogeneic hematopoietic stem cell transplantation (AHSCT) offers the best chance of cure for many patients with congenital and acquired hematologic diseases. Unfortunately, transplantation of alloreactive donor T cells which recognize and damage healthy patient tissues can result in Graft-versus-Host Disease (GvHD). One challenge to successful AHSCT is the prevention of GvHD without associated impairment of the beneficial effects of donor T cells, particularly immune reconstitution and prevention of relapse. GvHD can be prevented by non-specific depletion of donor T cells from stem cell grafts or by administration of pharmacological immunosuppression. Unfortunately these approaches increase infection and disease relapse. An alternative strategy is to selectively deplete alloreactive donor T cells after allostimulation by recipient antigen presenting cells (APC) before transplant. Early clinical trials of these allodepletion strategies improved immune reconstitution after HLA-mismatched HSCT without excess GvHD. However, some allodepletion techniques require specialized recipient APC production and some approaches may have off-target effects including depletion of donor pathogen-specific T cells and CD4 T regulatory cells .One alternative approach is the inactivation of alloreactive donor T cells via induction of alloantigen-specific hyporesponsiveness. This is achieved by stimulating donor cells with recipient APC while providing blockade of CD28-mediated co-stimulation signals.This "alloanergization" approach reduces alloreactivity by 1-2 logs while preserving pathogen- and tumor-associated antigen T cell responses in vitro. The strategy has been successfully employed in 2 completed and 1 ongoing clinical pilot studies in which alloanergized donor T cells were infused during or after HLA-mismatched HSCT resulting in rapid immune reconstitution, few infections and less severe acute and chronic GvHD than historical control recipients of unmanipulated HLA-mismatched transplantation. Here we describe our current protocol for the generation of peripheral blood mononuclear cells (PBMC) which have been alloanergized to HLA-mismatched unrelated stimulator PBMC. Alloanergization is achieved by allostimulation in the presence of monoclonal antibodies to the ligands B7.1 and B7.1 to block CD28-mediated costimulation. This technique does not require the production of specialized stimulator APC and is simple to perform, requiring only a single and relatively brief ex vivo incubation step. As such, the approach can be easily standardized for clinical use to generate donor T cells with reduced alloreactivity but retaining pathogen-specific immunity for adoptive transfer in the setting of AHSCT to improve immune reconstitution without excessive GvHD.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Isoantígenos/inmunología , Leucocitos Mononucleares/inmunología , Anergia Clonal/inmunología , Técnicas de Cocultivo , Epítopos , Antígenos HLA/inmunología , Trasplante de Células Madre Hematopoyéticas , Humanos , Prueba de Cultivo Mixto de Linfocitos
16.
Cancer Res ; 70(10): 3915-24, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20424114

RESUMEN

Allogeneic hematopoietic stem-cell transplantation can cure some patients with high-risk B-cell malignancies, but disease relapse following transplantation remains a significant problem. One approach that could be used to augment the donor T-cell-mediated antitumor effect is the infusion of allogeneic donor-derived T cells expressing a chimeric antibody receptor (CAR) specific to the B-cell antigen CD19. However, the use of such cells might result in toxicity in the form of graft-versus-host disease mediated by CD19-specific (CD19-CAR) T cells possessing alloreactive endogenous T-cell receptors. We therefore investigated whether nonalloreactive tumor-specific human T cells could be generated from peripheral blood mononuclear cells of healthy donors by the combination of CD19 redirection via CAR expression and subsequent alloanergization by allostimulation and concomitant blockade of CD28-mediated costimulation. Alloanergization of CD19-CAR T cells resulted in efficient and selective reduction of alloresponses in both CD4(+) and CD8(+) T cells, including allospecific proliferation and cytokine secretion. Importantly, T-cell effector functions including CAR-dependent proliferation and specific target cytolysis and cytokine production were retained after alloanergization. Our data support the application of CD19 redirection and subsequent alloanergization to generate allogeneic donor T cells for clinical use possessing increased antitumor activity but limited capacity to mediate graft-versus-host disease. Immunotherapy with such cells could potentially reduce disease relapse after allogeneic transplantation without increasing toxicity, thereby improving the outcome of patients undergoing allogeneic transplantation for high-risk B-cell malignancies.


Asunto(s)
Antígenos CD19/inmunología , Linfoma de Burkitt/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Anergia Clonal , Linfocitos T/inmunología , Adulto , Animales , Linfoma de Burkitt/terapia , Citometría de Flujo , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas , Humanos , Activación de Linfocitos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA