Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Breast ; 50: 64-70, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32062536

RESUMEN

BACKGROUND: STEPAUT, an Austrian non-interventional study, evaluated the safety and efficacy of everolimus plus exemestane in patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer (ABC) recurring/progressing on/after nonsteroidal aromatase inhibitors (NSAIs) in routine clinical practice. METHODS: Postmenopausal women with HR+, HER2- ABC progressing on/after NSAIs receiving everolimus plus exemestane in accordance with routine practice and the current version of Summary of Product Characteristics were eligible. Planned individual observation period corresponded to the duration of treatment until formal study end. RESULTS: Overall, 236 patients (median age: 65 years) were enrolled at 17 sites across Austria. The median progression-free survival (mPFS) in the overall population was 9.5 months (95% confidence interval [CI]: 8.6-10.7 months). The mPFS (95% CI) in patients who received everolimus 10 and 5 mg was 9.9 months (7.3-11.5 months) and 8 months (4.7-10.7 months), respectively. The median time to progression was numerically longer in patients who had a therapy break (11.9 months, 95% CI: 10.0-14.6 months) versus those who did not have any therapy break (10.7 months, 95% CI: 8.9-12.6 months). Patients experienced grade 1 (53.7%), grade 2 (35.9%), grade 3 (9.9%), grade 4 (0.2%) adverse events (AEs). The most common AEs of any grade were stomatitis, mucositis (53.8%), rash, exanthema (29.7%), loss of appetite, nausea (28.4%). CONCLUSIONS: Real-world safety and efficacy data from STEPAUT were consistent with results from BOLERO-2, supporting everolimus plus exemestane as a suitable treatment option for HR+, HER2- ABC recurring/progressing on/after NSAIs.


Asunto(s)
Androstadienos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Everolimus/uso terapéutico , Pautas de la Práctica en Medicina , Anciano , Austria/epidemiología , Femenino , Humanos , Posmenopausia , Supervivencia sin Progresión , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
2.
J Allergy Clin Immunol ; 143(1): 335-345.e12, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30009843

RESUMEN

BACKGROUND: Donor-specific antibodies of the IgG isotype are measured routinely for diagnostic purposes in renal transplant recipients and are associated with antibody-mediated rejection and long-term graft loss. OBJECTIVE: This study aimed to investigate whether MHC-specific antibodies of the IgE isotype are induced during allograft rejection. METHODS: Anti-MHC/HLA IgE levels were measured in sera of mice grafted with skin or heart transplants from various donor strains and in sera of kidney transplant patients with high levels of HLA IgG. Mediator release was triggered in vitro by stimulating basophils that were coated with murine or human IgE-positive serum, respectively, with specific recombinant MHC/HLA antigens. Kidney tissue samples obtained from organ donors were analyzed by using flow cytometry for cells expressing the high-affinity receptor for IgE (FcεRI). RESULTS: Donor MHC class I- and MHC class II-specific IgE was found on acute rejection of skin and heart grafts in several murine strain combinations, as well as during chronic antibody-mediated heart graft rejection. Anti-HLA IgE, including donor HLA class I and II specificities, was identified in a group of sensitized transplant recipients. Murine and human anti-MHC/HLA IgE triggered mediator release in coated basophils on stimulation with specific MHC/HLA antigens. HLA-specific IgE was not linked to atopy, and allergen-specific IgE present in allergic patients did not cross-react with HLA antigens. FcεRI+ cells were found in the human renal cortex and medulla and provide targets for HLA-specific IgE. CONCLUSION: These results demonstrate that MHC/HLA-specific IgE develops during an alloresponse and is functional in mediating effector mechanisms.


Asunto(s)
Rechazo de Injerto/inmunología , Trasplante de Corazón , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoglobulina E/inmunología , Trasplante de Riñón , Trasplante de Piel , Aloinjertos , Animales , Femenino , Rechazo de Injerto/patología , Humanos , Inmunoglobulina G/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C
3.
Am J Transplant ; 19(2): 591-596, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30346652

RESUMEN

Resistance to parental bone marrow (BM) grafts in F1 hybrid recipients is due to natural killer (NK) cell-mediated rejection triggered through "missing self" recognition. "Hybrid resistance" has usually been investigated in lethally irradiated F1 recipients in conjunction with pharmacological activation of NK cells. Here, we investigated BM-directed NK-cell alloreactivity in settings of reduced conditioning. Nonlethally irradiated (1-3 Gy) or nonirradiated F1 (C57BL6 × BALB/c) recipient mice received titrated doses (5-20 x 106 ) of unseparated parental BALB/c BM without pharmacological NK cell activation. BM successfully engrafted in all mice and multilineage donor chimerism persisted long-term (24 weeks), even in the absence of irradiation. Chimerism was associated with the rearrangement of the NK-cell receptor repertoire suggestive of reduced reactivity to BALB/c. Chimerism levels were lower after transplantation with parental BALB/c than with syngeneic F1 BM, indicating partial NK-mediated rejection of parental BM. Activation of NK cells with polyinosinic-polycytidylic acid sodium salt poly(I:C), reduced parental chimerism in nonirradiated BM recipients but did not prevent hematopoietic stem cell engraftment. In contrast, equal numbers of parental lymph node cells were completely rejected. Hence, hybrid resistance leads to incomplete rejection of parental BM under reduced conditioning settings.


Asunto(s)
Trasplante de Médula Ósea/métodos , Médula Ósea/inmunología , Rechazo de Injerto/inmunología , Tolerancia Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Quimera por Trasplante/inmunología , Animales , Médula Ósea/efectos de la radiación , Femenino , Células Asesinas Naturales/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
4.
J Heart Lung Transplant ; 37(9): 1119-1130, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29699851

RESUMEN

BACKGROUND: The mixed chimerism approach for intentional induction of donor-specific tolerance was shown to be successful in various models from mice to humans. For transplant patients, the approach would obviate the need for long-term immunosuppression and associated side effects; moreover, it would preclude the risk of late graft loss due to chronic rejection. Widespread clinical application is hindered by toxicities related to recipient pre-conditioning. Herein we aimed to investigate a clinically relevant protocol for tolerance induction to cardiac allografts, sparing CD40 blockade or T-cell depletion. METHODS: B6 mice were conditioned with non-myeloablative total body irradiation, fully mismatched BALB/c bone marrow cells, and short-term therapy, based on either anti- lymphocyte function-associated antigen-1 (anti-LFA-1) or anti-CD40L. Multilineage chimerism was followed by flow-cytometric analysis, tolerance was assessed with skin and heart allografts from fully or major histocompatibility complex-mismatched donors. Mechanisms of tolerance were investigated by analysis of donor-specific antibodies (DSAs), mixed lymphocyte reaction (MLR) assays, and deletion of donor-reactive T cells. RESULTS: We found that the combination of cytotoxic T-lymphocyte antigen 4 immunoglobulin (CTLA4Ig) and rapamycin with LFA-1 blockade enhanced bone marrow engraftment and led to more efficient T-cell engraftment and subsequent tolerization. Although fully mismatched skin grafts were chronically rejected, primarily vascularized heart allografts survived indefinitely and without signs of chronic rejection, independent of minor antigen mismatches. CONCLUSIONS: We have demonstarted a robust protocol for the induction of tolerance for cardiac allografts in the absence of CD40 blockade. Our findings demonstrate the potential of a clinically relevant minimal conditioning protocol designed to induce lifelong immunologic tolerance toward cardiac allografts.


Asunto(s)
Rechazo de Injerto/fisiopatología , Supervivencia de Injerto/fisiología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Linfocitos T/fisiología , Quimera por Trasplante/fisiología , Animales , Antígenos CD40/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos
5.
Surgery ; 161(3): 735-746, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27776794

RESUMEN

BACKGROUND: Interleukin-15 has become a promising molecule in the context of eliciting an effective, antitumor immune response because it is able to stimulate cells of the innate and adaptive immune system. METHODS: We generated an interleukin-15-expressing oncolytic influenza A virus for the treatment of an established murine tumor model. RESULTS: Our oncolytic influenza A virus produced large amounts of interleukin-15 and induced proliferation and activation of human T cells in vitro. Intraperitoneal administration increased the amount of mouse natural killer cells and effector memory T cells, as well as T cell reactivity in vivo. Moreover, intratumoral injection induced a profound decrease in growth of established tumors in mice and increased the amount of tumor-infiltrating T cells and natural killer cells. CONCLUSION: We established a stable, IL-15-producing oncolytic influenza A virus with promising immunostimulatory and antitumor attributes.


Asunto(s)
Virus de la Influenza A , Interleucina-15/fisiología , Melanoma Experimental/patología , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Cutáneas/patología , Animales , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Células Asesinas Naturales/efectos de los fármacos , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Neoplasias Cutáneas/terapia , Linfocitos T/efectos de los fármacos
6.
JCI Insight ; 1(7): e85911, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27699263

RESUMEN

Central clonal deletion has been considered the critical factor responsible for the robust state of tolerance achieved by chimerism-based experimental protocols, but split-tolerance models and the clinical experience are calling this assumption into question. Although clone-size reduction through deletion has been shown to be universally required for achieving allotolerance, it remains undetermined whether it is sufficient by itself. Therapeutic Treg treatment induces chimerism and tolerance in a stringent murine BM transplantation model devoid of myelosuppressive recipient treatment. In contrast to irradiation chimeras, chronic rejection (CR) of skin and heart allografts in Treg chimeras was permanently prevented, even in the absence of complete clonal deletion of donor MHC-reactive T cells. We show that minor histocompatibility antigen mismatches account for CR in irradiation chimeras without global T cell depletion. Furthermore, we show that Treg therapy-induced tolerance prevents CR in a linked suppression-like fashion, which is maintained by active regulatory mechanisms involving recruitment of thymus-derived Tregs to the graft. These data suggest that highly efficient intrathymic and peripheral deletion of donor-reactive T cells for specificities expressed on hematopoietic cells preclude the expansion of donor-specific Tregs and, hence, do not allow for spreading of tolerance to minor specificities that are not expressed by donor BM.


Asunto(s)
Trasplante de Médula Ósea , Supresión Clonal , Tolerancia Inmunológica , Antígenos de Histocompatibilidad Menor/inmunología , Linfocitos T Reguladores/citología , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL
7.
EBioMedicine ; 7: 230-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27322476

RESUMEN

BACKGROUND: Therapeutic strategies for the prophylaxis of IgE-mediated allergy remain an unmet medical need. Cell therapy is an emerging approach with high potential for preventing and treating immunological diseases. We aimed to develop a cell-based therapy inducing permanent allergen-specific immunological tolerance for preventing IgE-mediated allergy. METHODS: Wild-type mice were treated with allergen-expressing bone marrow cells under a short course of tolerogenic immunosuppression (mTOR inhibition and costimulation blockade). Bone marrow was retrieved from a novel transgenic mouse ubiquitously expressing the major grass pollen allergen Phl p 5 as a membrane-anchored protein (BALB/c-Tg[Phlp5-GFP], here mPhl p 5). After transplantation recipients were IgE-sensitized at multiple time points with Phl p 5 and control allergen. RESULTS: Mice treated with mPhl p 5 bone marrow did not develop Phl p 5-specific IgE (or other isotypes) despite repeated administration of the allergen, while mounting and maintaining a strong humoral response towards the control allergen. Notably, Phl p 5-specific T cell responses and allergic airway inflammation were also completely prevented. Interestingly allergen-specific B cell tolerance was maintained independent of Treg functions indicating deletional tolerance as underlying mechanism. CONCLUSION: This proof-of-concept study demonstrates that allergen-specific immunological tolerance preventing occurrence of allergy can be established through a cell-based therapy employing allergen-expressing leukocytes.


Asunto(s)
Alérgenos/inmunología , Trasplante de Médula Ósea/métodos , Hipersensibilidad/prevención & control , Inmunoglobulina E/metabolismo , Alérgenos/genética , Animales , Modelos Animales de Enfermedad , Humanos , Hipersensibilidad/inmunología , Tolerancia Inmunológica , Ratones , Ratones Transgénicos , Polen/inmunología , Profilaxis Pre-Exposición/métodos
8.
PLoS One ; 11(1): e0146245, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26731275

RESUMEN

Cell therapy with recipient Tregs achieves engraftment of allogeneic bone marrow (BM) without the need for cytoreductive conditioning (i.e., without irradiation or cytotoxic drugs). Thereby mixed chimerism and transplantation tolerance are established in recipients conditioned solely with costimulation blockade and rapamycin. However, clinical translation would be substantially facilitated if Treg-stimulating pharmaceutical agents could be used instead of individualized cell therapy. Recently, it was shown that interleukin-2 (IL-2) complexed with a monoclonal antibody (mAb) (clone JES6-1A12) against IL-2 (IL-2 complexes) potently expands and activates Tregs in vivo. Therefore, we investigated whether IL-2 complexes can replace Treg therapy in a costimulation blockade-based and irradiation-free BM transplantation (BMT) model. Unexpectedly, the administration of IL-2 complexes at the time of BMT (instead of Tregs) failed to induce BM engraftment in non-irradiated recipients (0/6 with IL-2 complexes vs. 3/4 with Tregs, p<0.05). Adding IL-2 complexes to an otherwise effective regimen involving recipient irradiation (1Gy) but no Treg transfer indeed actively triggered donor BM rejection at higher doses (0/8 with IL-2 complexes vs. 9/11 without, p<0.01) and had no detectable effect at two lower doses (3/5 vs. 9/11, p>0.05). CD8 T cells and NK cells of IL-2 complex-treated naïve mice showed an enhanced proliferative response towards donor antigens in vitro despite the marked expansion of Tregs. However, IL-2 complexes also expanded conventional CD4 T cells, CD8 T cells, NK cells, NKT cells and notably even B cells, albeit to a lesser extent. Notably, IL-2 complex expanded Tregs featured less potent suppressive functions than in vitro activated Tregs in terms of T cell suppression in vitro and BM engraftment in vivo. In conclusion, these data suggest that IL-2 complexes are less effective than recipient Tregs in promoting BM engraftment and in contrast actually trigger BM rejection, as their effect is not sufficiently restricted to Tregs but rather extends to several other lymphocyte populations.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Trasplante de Médula Ósea/métodos , Interleucina-2/uso terapéutico , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Femenino , Interleucina-2/farmacología , Ratones , Linfocitos T Reguladores/efectos de los fármacos , Tolerancia al Trasplante/inmunología
9.
J Immunol Res ; 2015: 562935, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26273682

RESUMEN

Induction of donor-specific tolerance is still considered as the "Holy Grail" in transplantation medicine. The mixed chimerism approach is virtually the only tolerance approach that was successfully translated into the clinical setting. We have previously reported successful induction of chimerism and tolerance using cell therapy with recipient T regulatory cells (Tregs) to avoid cytotoxic recipient treatment. Treg therapy is limited by the availability of cells as large-scale expansion is time-consuming and associated with the risk of contamination with effector cells. Using a costimulation-blockade based bone marrow (BM) transplantation (BMT) model with Treg therapy instead of cytoreductive recipient treatment we aimed to determine the most potent Treg population for clinical translation. Here we show that CD4(+)CD25(+) in vitro activated nTregs are superior to TGFß induced iTregs in promoting the induction of chimerism and tolerance. Therapy with nTregs (but not iTregs) led to multilineage chimerism and donor-specific tolerance in mice receiving as few as 0.5 × 10(6) cells. Moreover, we show that only recipient Tregs, but not donor or third-party Tregs, had a beneficial effect on BM engraftment at the tested doses. Thus, recipient-type nTregs significantly improve chimerism and tolerance and might be the most potent Treg population for translation into the clinical setting.


Asunto(s)
Linfocitos T Reguladores/inmunología , Donantes de Tejidos , Receptores de Trasplantes , Tolerancia al Trasplante/inmunología , Animales , Trasplante de Médula Ósea , Femenino , Supervivencia de Injerto/inmunología , Isoanticuerpos/inmunología , Ratones , Trasplante de Piel , Quimera por Trasplante
10.
Eur J Immunol ; 45(9): 2470-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26200095

RESUMEN

Establishing donor-specific immunological tolerance could improve long-term outcome by obviating the need for immunosuppressive drug therapy, which is currently required to control alloreactivity after organ transplantation. Mixed chimerism is defined as the engraftment of donor hematopoietic stem cells in the recipient, leading to viable coexistence of both donor and recipient leukocytes. In numerous experimental models, cotransplantation of donor bone marrow (BM) into preconditioned (e.g., through irradiation or cytotoxic drugs) recipients leads to transplantation tolerance through (mixed) chimerism. Mixed chimerism offers immunological advantages for clinical translation; pilot trials have established proof of concept by deliberately inducing tolerance in humans. Widespread clinical application is prevented, however, by the harsh preconditioning currently necessary for permitting BM engraftment. Recently, the immunological mechanisms inducing and maintaining tolerance in experimental mixed chimerism have been defined, revealing a more prominent role for regulation than historically assumed. The evidence from murine models suggests that both deletional and regulatory mechanisms are critical in promoting complete tolerance, encompassing also the minor histocompatibility antigens. Here, we review the current understanding of tolerance through mixed chimerism and provide an outlook on how to realize widespread clinical translation based on mechanistic insights gained from chimerism protocols, including cell therapy with polyclonal regulatory T cells.


Asunto(s)
Trasplante de Médula Ósea , Quimerismo , Linfocitos T Reguladores/inmunología , Quimera por Trasplante/inmunología , Tolerancia al Trasplante , Animales , Anticuerpos Monoclonales/uso terapéutico , Ensayos Clínicos como Asunto , Rayos gamma/uso terapéutico , Regulación de la Expresión Génica , Humanos , Inmunosupresores/uso terapéutico , Ratones , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Linfocitos T Reguladores/citología , Quimera por Trasplante/genética , Acondicionamiento Pretrasplante/métodos
11.
Circulation ; 132(2): 122-31, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-25957225

RESUMEN

BACKGROUND: Organ transplantation has seen an increased use of organs from older donors over the past decades in an attempt to meet the globally growing shortage of donor organs. However, inferior transplantation outcomes when older donor organs are used represent a growing challenge. METHODS AND RESULTS: Here, we characterize the impact of donor age on solid-organ transplantation using a murine cardiac transplantation model. We found a compromised graft survival when older hearts were used. Shorter graft survival of older hearts was independent of organ age per se, because chimeric young or old organs repopulated with young passenger leukocytes showed comparable survival times. Transplantation of older organs triggered more potent alloimmune responses via intragraft CD11c+ dendritic cells augmenting CD4+ and CD8+ T-cell proliferation and proinflammatory cytokine production, particularly that of interleukin-17A. Of note, depletion of donor CD11c+ dendritic cells before engraftment, neutralization of interleukin-17A, or transplantation of older hearts into IL-17A(-/-) mice delayed rejection and reduced alloimmune responses to levels observed when young hearts were transplanted. CONCLUSIONS: These results demonstrate a critical role of old donor CD11c+ dendritic cells in mounting age-dependent alloimmune responses with an augmented interleukin-17A response in recipient animals. Targeting interleukin-17A may serve as a novel therapeutic approach when older organs are transplanted.


Asunto(s)
Envejecimiento/inmunología , Antígeno CD11c/inmunología , Células Dendríticas/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón/efectos adversos , Interleucina-17/inmunología , Envejecimiento/patología , Animales , Células Dendríticas/patología , Rechazo de Injerto/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Trasplante Homólogo/efectos adversos
12.
J Heart Lung Transplant ; 33(4): 429-37, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24468120

RESUMEN

BACKGROUND: The mixed chimerism approach induces donor-specific tolerance in both pre-clinical models and clinical pilot trials. However, chronic rejection of heart allografts and acute rejection of skin allografts were observed in some chimeric animals despite persistent hematopoietic chimerism and tolerance toward donor antigens in vitro. We tested whether additional cell therapy with regulatory T cells (Tregs) is able to induce full immunologic tolerance and prevent chronic rejection. METHODS: We recently developed a murine "Treg bone marrow (BM) transplantation (BMT) protocol" that is devoid of cytoreductive recipient pre-treatment. The protocol consists of a moderate dose of fully mismatched allogeneic donor BM under costimulation blockade, together with polyclonal recipient Tregs and rapamycin. Control groups received BMT under non-myeloablative irradiation and costimulation blockade without Treg therapy. Multilineage chimerism was followed by flow cytometry, and tolerance was assessed by donor-specific skin and heart allografts. RESULTS: Durable multilineage chimerism and long-term donor skin and heart allograft survival were successfully achieved with both protocols. Notably, histologic examination of heart allografts at the end of follow-up revealed that chronic rejection is prevented only in chimeras induced with the Treg protocol. CONCLUSIONS: In a mouse model of mixed chimerism, additional Treg treatment at the time of BMT prevents chronic rejection of heart allografts. As the Treg-chimerism protocol also obviates the need for cytoreductive recipient treatment it improves both efficacy and safety over previous non-myeloablative mixed chimerism regimens. These results may significantly impact the development of protocols for tolerance induction in cardiac transplantation.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Corazón , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Quimera por Trasplante/inmunología , Tolerancia al Trasplante/inmunología , Aloinjertos , Animales , Enfermedad Crónica , Femenino , Ratones , Ratones Endogámicos , Sirolimus/farmacología , Trasplante de Piel , Inmunología del Trasplante/inmunología
13.
Eur J Immunol ; 43(9): 2451-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23765421

RESUMEN

The transplantation of allergens (e.g. Phl p 5 or Bet v 1) expressed on BM cells as membrane-anchored full-length proteins leads to permanent tolerance at the T-cell, B-cell, and effector-cell levels. Since the exposure of complete allergens bears the risk of inducing anaphylaxis, we investigated here whether expression of Phl p 5 in the cytoplasm (rather than on the cell surface) is sufficient for tolerance induction. Transplantation of BALB/c BM retrovirally transduced to express Phl p 5 in the cytoplasm led to stable and durable molecular chimerism in syngeneic recipients (∼20% chimerism at 6 months). Chimeras showed allergen-specific T-cell hyporesponsiveness. Further, Phl p 5-specific TH 1-dependent humoral responses were tolerized in several chimeras. Surprisingly, Phl p 5-specific IgE and IgG1 levels were significantly reduced but still detectable in sera of chimeric mice, indicating incomplete B-cell tolerance. No Phl p 5-specific sIgM developed in cytoplasmic chimeras, which is in marked contrast to mice transplanted with BM expressing membrane-anchored Phl p 5. Thus, the expression site of the allergen substantially influences the degree and quality of tolerance achieved with molecular chimerism in IgE-mediated allergy.


Asunto(s)
Alérgenos/inmunología , Antígenos de Plantas/inmunología , Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Tolerancia Inmunológica , Proteínas de Plantas/inmunología , Células 3T3 , Alérgenos/biosíntesis , Animales , Células de la Médula Ósea/virología , Trasplante de Médula Ósea/inmunología , Línea Celular , Proliferación Celular , Quimera/inmunología , Femenino , Hipersensibilidad/inmunología , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología
14.
Transplantation ; 95(12): 1431-8, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23694950

RESUMEN

BACKGROUND: Allogeneic bone marrow transplantation is under investigation for a range of nonmalignant indications, including tolerance induction through mixed chimerism. This strategy has so far been tested experimentally only in young recipients. Due to immunosenescence, older patients have an increase in memory T cells (TMEM) as well as other alterations to their immune system, which may influence the potential to induce tolerance. We therefore investigated the impact of immunosenescence on chimerism-based tolerance induction. METHODS: Groups of young (2 months) and old (12 months) C57BL/6 recipients received BALB/c bone marrow under nonmyeloablative (3 Gy) and minimal (1 Gy) total body irradiation and treatment with costimulation blockade, T-cell depletion, or rapamycin. Multilineage chimerism, clonal deletion, and lymphocyte subsets were analyzed by flow cytometry. Tolerance was assessed by skin and heart grafts and enzyme-linked immunospot, intracellular cytokine, and mixed lymphocyte reaction assays. RESULTS: Unexpectedly, chimerism and tolerance were established in old recipients with comparable-and in some cases increased-efficacy as in young recipients employing costimulation blockade-based or T-cell depletion-based conditioning with 1 or 3 Gy total body irradiation. TMEM reactivity in (naïve) old mice was augmented in response to polyclonal but not to allogeneic stimulation, providing a mechanistic underpinning for the susceptibility to chimerism induction despite increased TMEM frequencies. Tolerance in old recipients was associated with peripheral and central clonal deletion and a higher frequency of regulatory T cells. CONCLUSION: Advanced age does not impair bone marrow engraftment, thereby widening the clinical potential of experimental protocols inducing transplantation tolerance through mixed chimerism.


Asunto(s)
Envejecimiento/inmunología , Trasplante de Médula Ósea/inmunología , Tolerancia al Trasplante/inmunología , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Citocinas/metabolismo , Femenino , Citometría de Flujo , Memoria Inmunológica , Depleción Linfocítica , Linfocitos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Linfocitos T/inmunología , Factores de Tiempo , Quimera por Trasplante/inmunología , Tolerancia al Trasplante/efectos de los fármacos
15.
Immunobiology ; 218(9): 1139-46, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23623394

RESUMEN

Molecular chimerism is a promising strategy to induce tolerance to disease-causing antigens expressed on genetically modified haematopoietic stem cells. The approach was employed successfully in models of autoimmunity and organ transplantation. Recently, we demonstrated that molecular chimerism induces robust and lasting tolerance towards the major grass pollen allergen Phl p 5. Since allergens are a group of antigens differing widely in their function, origin and structure we further examined the effectiveness of molecular chimerism using the Phl p 5-unrelated major birch pollen allergen Bet v 1, co-expressed with the reporter GFP. Besides, inhibition of CD26 was used to promote engraftment of modified stem cells. Retrovirus VSV-Betv1-GFP was generated to transduce 5-FU-mobilized BALB/c hematopoietic cells to express membrane-bound Bet v 1 (VSV-GFP virus was used as control). Myeloablated BALB/c mice received Betv1-GFP or GFP expressing bone marrow cells, pre-treated with a CD26 inhibitor. Chimerism was followed by flow cytometry. Tolerance was assessed by measuring allergen-specific isotype levels in sera, RBL assays and T-cell proliferation assays. Mice transplanted with transduced BMC developed multi-lineage molecular chimerism which remained stable long-term (>8 months). After repeated immunizations with Bet v 1 and Phl p 5 serum levels of Bet v 1-specific antibodies (IgE, IgG1, IgG2a, IgG3 and IgA) remained undetectable in Betv1-GFP chimeras while high levels of Phl p 5-specific antibodies developed. Likewise, basophil degranulation was induced in response to Phl p 5 but not to Bet v 1 and specific non-responsiveness to Bet v 1 was observed in proliferation assays. These data demonstrate successful tolerization towards Bet v 1 by molecular chimerism. Stable long-term chimerism was achieved under inhibition of CD26. These results provide evidence for the broad applicability of molecular chimerism as tolerance strategy in allergy.


Asunto(s)
Antígenos de Plantas/inmunología , Basófilos/inmunología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Quimera por Trasplante/inmunología , Tolerancia al Trasplante/inmunología , Animales , Antígenos de Plantas/genética , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Degranulación de la Célula , Células Cultivadas , Dipeptidil Peptidasa 4/metabolismo , Fluorouracilo/administración & dosificación , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunidad Humoral , Ratones , Ratones Endogámicos BALB C , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Retroviridae , Transducción Genética
16.
Transpl Int ; 26(3): 242-53, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23190423

RESUMEN

Aging affects all compartments of the immune response and has a major impact on transplant outcome and organ quality. Although clinical trials in the aging transplant population remain rare, our current understanding of immunosenescence provides a basis for an age-adapted immunosuppression and organ allocation with the goal to optimize utilization and to improve outcomes in older recipients. From a more general perspective, understanding the mechanisms and consequences of immunosenescence will have a broad impact on immune therapies in and beyond transplantation.


Asunto(s)
Envejecimiento/inmunología , Inmunidad Innata/fisiología , Trasplante de Órganos/métodos , Inmunología del Trasplante/inmunología , Factores de Edad , Anciano , Envejecimiento/fisiología , Rechazo de Injerto , Supervivencia de Injerto , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/inmunología , Trasplante de Riñón/métodos , Trasplante de Hígado/efectos adversos , Trasplante de Hígado/inmunología , Trasplante de Hígado/métodos , Donadores Vivos , Persona de Mediana Edad , Trasplante de Órganos/efectos adversos , Pronóstico , Medición de Riesgo , Donantes de Tejidos , Inmunología del Trasplante/fisiología
17.
Transpl Int ; 26(2): 206-18, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23240587

RESUMEN

While costimulation blockade-based mixed chimerism protocols work well for inducing tolerance in rodents, translation to preclinical large animal/nonhuman primate models has been less successful. One recognized cause for these difficulties is the high frequency of alloreactive memory T cells (Tmem) found in the (pre)clinical setting as opposed to laboratory mice. In the present study, we therefore developed a murine bone marrow transplantation (BMT) model employing recipients harboring polyclonal donor-reactive Tmem without concomitant humoral sensitization. This model was then used to identify strategies to overcome this additional immune barrier. We found that B6 recipients that were enriched with 3 × 10(7) T cells isolated from B6 mice that had been previously grafted with Balb/c skin, rejected Balb/c BM despite costimulation blockade with anti-CD40L and CTLA4Ig (while recipients not enriched developed chimerism). Adjunctive short-term treatment of sensitized BMT recipients with rapamycin or anti-LFA-1 mAb was demonstrated to be effective in controlling Tmem in this model, leading to long-term mixed chimerism and donor-specific tolerance. Thus, rapamycin and anti-LFA-1 mAb are effective in overcoming the potent barrier that donor-reactive Tmem pose to the induction of mixed chimerism and tolerance despite costimulation blockade.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Células de la Médula Ósea/citología , Rechazo de Injerto , Antígeno-1 Asociado a Función de Linfocito/inmunología , Sirolimus/uso terapéutico , Animales , Anticuerpos/metabolismo , Trasplante de Médula Ósea , Ligando de CD40/metabolismo , Antígeno CTLA-4/metabolismo , Quimerismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Trasplante de Piel , Linfocitos T/citología , Tolerancia al Trasplante
18.
Curr Opin Organ Transplant ; 17(1): 63-70, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22186093

RESUMEN

PURPOSE OF REVIEW: Organ transplantation is the state-of-the-art treatment for end-stage organ failure; however, long-term graft survival is still unsatisfactory. Despite improved immunosuppressive drug therapy, patients are faced with substantial side effects and the risk of chronic rejection with subsequent graft loss. The transplantation of donor bone marrow for the induction of mixed chimerism has been recognized to induce donor-specific tolerance a long time ago, but safety concerns regarding toxicities of current bone marrow transplantation (BMT) protocols impede widespread application. RECENT FINDINGS: Recent studies in nonhuman primates and kidney transplant patients have demonstrated successful induction of allograft tolerance even though--in contrast to murine models--only transient chimerism was achieved. Progress toward the development of nontoxic murine BMT protocols revealed that Treg therapy is a potent therapeutic adjunct eliminating the need for cytotoxic recipient conditioning. Furthermore, new insight into the mechanisms underlying tolerization of CD4 and CD8 T cells in mixed chimeras has been gained and has identified possible difficulties impeding clinical translation. SUMMARY: This review will address the recent advances in murine models as well as findings from the first clinical trials for the induction of tolerance through mixed chimerism. Both the potential for more widespread clinical application and the remaining hurdles and challenges of this tolerance approach will be discussed.


Asunto(s)
Trasplante de Médula Ósea , Quimerismo , Enfermedad Injerto contra Huésped/prevención & control , Inmunoterapia Adoptiva , Trasplante de Órganos , Tolerancia al Trasplante/inmunología , Animales , Antígenos CD/inmunología , Trasplante de Médula Ósea/métodos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Humanos , Inmunoterapia Adoptiva/métodos , Ratones , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...