Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(1): 60-71, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36167031

RESUMEN

Allogeneic hematopoietic stem cell transplantation (alloSCT) is curative for severe inborn errors of immunity (IEIs), with recent data suggesting alloSCT in adulthood is safe and effective in selected patients. However, questions remain regarding the indications for and optimal timing of transplant. We retrospectively compared outcomes of transplanted vs matched nontransplanted adults with severe IEIs. Seventy-nine patients (aged ≥ 15 years) underwent alloSCT between 2008 and 2018 for IEIs such as chronic granulomatous disease (n = 20) and various combined immune deficiencies (n = 59). A cohort of nontransplanted patients from the French Centre de Référence Déficits Immunitaires Héréditaires registry was identified blindly for case-control analysis, with ≤3 matched controls per index patient, without replacement. The nontransplanted patients were matched for birth decade, age at last review greater than index patient age at alloSCT, chronic granulomatous disease or combined immune deficiencies, and autoimmune/lymphoproliferative complications. A total of 281 patients were included (79 transplanted, 202 nontransplanted). Median age at transplant was 21 years. Transplant indications were mainly lymphoproliferative disease (n = 23) or colitis (n = 15). Median follow-up was 4.8 years (interquartile range, 2.5-7.2). One-year transplant-related mortality rate was 13%. Estimated disease-free survival at 5 years was higher in transplanted patients (58% vs 33%; P = .007). Nontransplanted patients had an ongoing risk of severe events, with an increased mean cumulative number of recurrent events compared with transplanted patients. Sensitivity analyses removing patients with common variable immune deficiency and their matched transplanted patients confirm these results. AlloSCT prevents progressive morbidity associated with IEIs in adults, which may outweigh the negative impact of transplant-related mortality.


Asunto(s)
Enfermedad Injerto contra Huésped , Enfermedad Granulomatosa Crónica , Trasplante de Células Madre Hematopoyéticas , Humanos , Adulto , Adulto Joven , Estudios Retrospectivos , Enfermedad Granulomatosa Crónica/terapia , Tratamiento Conservador , Trasplante Homólogo/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre/métodos , Acondicionamiento Pretrasplante/métodos , Enfermedad Injerto contra Huésped/etiología
2.
N Engl J Med ; 385(3): 228-238, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34260836

RESUMEN

BACKGROUND: Chronic graft-versus-host disease (GVHD), a major complication of allogeneic stem-cell transplantation, becomes glucocorticoid-refractory or glucocorticoid-dependent in approximately 50% of patients. Robust data from phase 3 randomized studies evaluating second-line therapy for chronic GVHD are lacking. In retrospective surveys, ruxolitinib, a Janus kinase (JAK1-JAK2) inhibitor, showed potential efficacy in patients with glucocorticoid-refractory or -dependent chronic GVHD. METHODS: This phase 3 open-label, randomized trial evaluated the efficacy and safety of ruxolitinib at a dose of 10 mg twice daily, as compared with the investigator's choice of therapy from a list of 10 commonly used options considered best available care (control), in patients 12 years of age or older with moderate or severe glucocorticoid-refractory or -dependent chronic GVHD. The primary end point was overall response (complete or partial response) at week 24; key secondary end points were failure-free survival and improved score on the modified Lee Symptom Scale at week 24. RESULTS: A total of 329 patients underwent randomization; 165 patients were assigned to receive ruxolitinib and 164 patients to receive control therapy. Overall response at week 24 was greater in the ruxolitinib group than in the control group (49.7% vs. 25.6%; odds ratio, 2.99; P<0.001). Ruxolitinib led to longer median failure-free survival than control (>18.6 months vs. 5.7 months; hazard ratio, 0.37; P<0.001) and higher symptom response (24.2% vs. 11.0%; odds ratio, 2.62; P = 0.001). The most common (occurring in ≥10% patients) adverse events of grade 3 or higher up to week 24 were thrombocytopenia (15.2% in the ruxolitinib group and 10.1% in the control group) and anemia (12.7% and 7.6%, respectively). The incidence of cytomegalovirus infections and reactivations was similar in the two groups. CONCLUSIONS: Among patients with glucocorticoid-refractory or -dependent chronic GVHD, ruxolitinib led to significantly greater overall response, failure-free survival, and symptom response. The incidence of thrombocytopenia and anemia was greater with ruxolitinib. (Funded by Novartis and Incyte; REACH3 ClinicalTrials.gov number, NCT03112603.).


Asunto(s)
Enfermedad Injerto contra Huésped/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Quinasas Janus/antagonistas & inhibidores , Pirazoles/uso terapéutico , Adolescente , Adulto , Anciano , Niño , Infecciones por Citomegalovirus/etiología , Femenino , Glucocorticoides/uso terapéutico , Humanos , Factores Inmunológicos/efectos adversos , Masculino , Persona de Mediana Edad , Nitrilos , Fotoféresis , Pirazoles/efectos adversos , Pirimidinas , Análisis de Supervivencia , Trombocitopenia/inducido químicamente , Insuficiencia del Tratamiento , Adulto Joven
3.
Blood ; 138(18): 1657-1665, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34370823

RESUMEN

Regenerative failure at barrier surfaces and maladaptive repair leading to fibrosis are hallmarks of graft-versus-host disease (GVHD). Although immunosuppressive treatment can control inflammation, impaired tissue homeostasis leads to prolonged organ damage and impaired quality of life. In this Blood Spotlight, we review recent research that addresses the critical failures in tissue regeneration and repair that underpin treatment-resistant GVHD. We highlight current interventions designed to overcome these defects and provide our assessment of the future therapeutic landscape.


Asunto(s)
Enfermedad Injerto contra Huésped/fisiopatología , Enfermedad Injerto contra Huésped/terapia , Animales , Enfermedad Injerto contra Huésped/inmunología , Humanos , Calidad de Vida , Regeneración , Medicina Regenerativa
4.
Blood ; 131(8): 917-931, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29279357

RESUMEN

The primary immunodeficiencies (PIDs), rare inherited diseases characterized by severe dysfunction of immunity, have been successfully treated by allogeneic hematopoietic stem cell transplantation (Allo-HSCT) in childhood. Controversy exists regarding optimal timing and use of Allo-HSCT in adults, due to lack of experience and previous poor outcomes. Twenty-nine consecutive adult patients, with a mean age at transplant of 24 years (range, 17-50 years), underwent Allo-HSCT. Reduced-intensity conditioning (RIC) included fludarabine (Flu)/melphalan/alemtuzumab (n = 20), Flu/busulfan (Bu)/alemtuzumab (n = 8), and Flu/Bu/antithymocyte globulin (n = 1). Stem cell donors were matched unrelated donors or mismatched unrelated donors (n = 18) and matched related donors (n = 11). Overall survival (OS), event-free survival, transplant-related mortality (TRM), acute and chronic graft-versus-host disease incidence and severity, time to engraftment, lineage-specific chimerism, immune reconstitution, and discontinuation of immunoglobulin replacement therapy were recorded. OS at 3 years for the whole cohort was 85.2%. The rarer PID patients without chronic granulomatous disease (CGD) achieved an OS at 3 years of 88.9% (n = 18), compared with 81.8% for CGD patients (n = 11). TRM was low with only 4 deaths observed at a median follow-up of 3.5 years. There were no cases of early or late rejection. In all surviving patients, either stable mixed chimerism or full donor chimerism were observed. At last follow-up, 87% of the surviving patients had no evidence of persistent or recurrent infections. Allo-HSCT is safe and effective in young adult patients with severe PID and should be considered the treatment of choice where an appropriate donor is available.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/mortalidad , Síndromes de Inmunodeficiencia/mortalidad , Adolescente , Adulto , Estudios de Cohortes , Femenino , Humanos , Síndromes de Inmunodeficiencia/patología , Síndromes de Inmunodeficiencia/terapia , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia , Acondicionamiento Pretrasplante , Trasplante Homólogo , Adulto Joven
5.
Haematologica ; 105(11): 2639-2646, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33131253

RESUMEN

Poor graft function is a serious complication following allogeneic hematopoietic stem cell transplantation. Infusion of CD34+-selected stem cells without pre-conditioning has been used to correct poor graft function, but predictors of recovery are unclear. We report the outcome of 62 consecutive patients who had primary or secondary poor graft function who underwent a CD34+-selected stem cell infusion from the same donor without further conditioning. Forty-seven of 62 patients showed hematological improvement and became permanently transfusion and growth factor-independent. In multivariate analysis, parameters significantly associated with recovery were shared CMV seronegative status for recipient/donor, the absence of active infection and matched recipient/donor sex. Recovery was similar in patients with mixed and full donor chimerism. Five -year overall survival was 74.4% (95% CI 59-89) in patients demonstrating complete recovery, 16.7% (95% CI 3-46) in patients with partial recovery and 22.2% (CI 95% 5-47) in patients with no response. In patients with count recovery, those with poor graft function in 1-2 lineages had superior 5-year overall survival (93.8%, 95% CI 82-99) than those with tri-lineage failure (53%, 95% CI 34-88). New strategies including cytokine or agonist support, or second transplant need to be investigated in patients who do not recover.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Donantes de Tejidos , Acondicionamiento Pretrasplante , Trasplante Homólogo
6.
Blood ; 130(11): 1327-1335, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28716862

RESUMEN

Until recently, hematopoietic stem cell transplantation was the only curative option for Wiskott-Aldrich syndrome (WAS). The first attempts at gene therapy for WAS using a ϒ-retroviral vector improved immunological parameters substantially but were complicated by acute leukemia as a result of insertional mutagenesis in a high proportion of patients. More recently, treatment of children with a state-of-the-art self-inactivating lentiviral vector (LV-w1.6 WASp) has resulted in significant clinical benefit without inducing selection of clones harboring integrations near oncogenes. Here, we describe a case of a presplenectomized 30-year-old patient with severe WAS manifesting as cutaneous vasculitis, inflammatory arthropathy, intermittent polyclonal lymphoproliferation, and significant chronic kidney disease and requiring long-term immunosuppressive treatment. Following reduced-intensity conditioning, there was rapid engraftment and expansion of a polyclonal pool of transgene-positive functional T cells and sustained gene marking in myeloid and B-cell lineages up to 20 months of observation. The patient was able to discontinue immunosuppression and exogenous immunoglobulin support, with improvement in vasculitic disease and proinflammatory markers. Autologous gene therapy using a lentiviral vector is a viable strategy for adult WAS patients with severe chronic disease complications and for whom an allogeneic procedure could present an unacceptable risk. This trial was registered at www.clinicaltrials.gov as #NCT01347242.


Asunto(s)
Terapia Genética , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/terapia , Adulto , Proliferación Celular , Preescolar , Ensayos Clínicos como Asunto , Células Clonales , Citocinas/sangre , Humanos , Subgrupos Linfocitarios/inmunología , Linfocitos T/inmunología , Vacunación , Síndrome de Wiskott-Aldrich/sangre
7.
Mol Ther ; 25(2): 504-511, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28153097

RESUMEN

Recombinant lentiviral vectors (LVs) are highly effective vaccination vehicles that elicit protective T cell immunity in disease models. Dendritic cells (DCs) acquire antigen at sites of vaccination and migrate to draining lymph nodes, where they prime vaccine-specific T cells. The potency with which LVs activate CD8+ T cell immunity has been attributed to the transduction of DCs at the immunization site and durable presentation of LV-encoded antigens. However, it is not known how LV-encoded antigens continue to be presented to T cells once directly transduced DCs have turned over. Here, we report that LV-encoded antigen is efficiently cross-presented by DCs in vitro. We have further exploited the temporal depletion of DCs in the murine CD11c.DTR (diphtheria toxin receptor) model to demonstrate that repopulating DCs that were absent at the time of immunization cross-present LV-encoded antigen to T cells in vivo. Indirect presentation of antigen from transduced cells by DCs is sufficient to prime functional effector T cells that control tumor growth. These data suggest that DCs cross-present immunogenic antigen from LV-transduced cells, thereby facilitating prolonged activation of T cells in the absence of circulating LV particles. These are findings that may impact on the future design of LV vaccination strategies.


Asunto(s)
Presentación de Antígeno , Antígenos/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Inmunidad Celular , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos/genética , Biomarcadores , Antígeno CD11c/metabolismo , Vacunas contra el Cáncer/inmunología , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Vectores Genéticos/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/inmunología , Lentivirus/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Modelos Animales , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Transducción Genética , Vacunas/genética , Vacunas/inmunología
8.
Eur J Immunol ; 46(1): 192-203, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26464217

RESUMEN

Dendritic cells (DCs) play a vital role in innate and adaptive immunities. Inducible depletion of CD11c(+) DCs engineered to express a high-affinity diphtheria toxin receptor has been a powerful tool to dissect DC function in vivo. However, despite reports showing that loss of DCs induces transient monocytosis, the monocyte population that emerges and the potential impact of monocytes on studies of DC function have not been investigated. We found that depletion of CD11c(+) cells from CD11c.DTR mice induced the expansion of a variant CD64(+) Ly6C(+) monocyte population in the spleen and blood that was distinct from conventional monocytes. Expansion of CD64(+) Ly6C(+) monocytes was independent of mobilization from the BM via CCR2 but required the cytokine, G-CSF. Indeed, this population was also expanded upon exposure to exogenous G-CSF in the absence of DC depletion. CD64(+) Ly6C(+) monocytes were characterized by upregulation of innate signaling apparatus despite the absence of inflammation, and an increased capacity to produce TNF-α following LPS stimulation. Thus, depletion of CD11c(+) cells induces expansion of a unique CD64(+) Ly6C(+) monocyte population poised to synthesize TNF-α. This finding will require consideration in experiments using depletion strategies to test the role of CD11c(+) DCs in immunity.


Asunto(s)
Células Dendríticas/inmunología , Monocitos/citología , Monocitos/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Antígenos Ly/inmunología , Antígeno CD11c/inmunología , Citometría de Flujo , Ratones , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de IgG/inmunología , Factor de Necrosis Tumoral alfa/inmunología
9.
Blood ; 125(4): 731-9, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25499763

RESUMEN

Cytomegalovirus (CMV) remains a significant cause of morbidity after allogeneic hematopoietic stem cell transplantation (HSCT). Clinical risk varies according to a number of factors, including recipient/donor CMV serostatus. Current dogma suggests risk is greatest in seropositive recipient (R+)/seronegative donor (D-) transplants and is exacerbated by T-cell depletion. We hypothesized that in the setting of reduced-intensity T-cell-depleted conditioning, recipient-derived CMV-specific T cells escaping deletion may contribute significantly to CMV-specific immunity and might therefore also influence chimerism status. We evaluated 105 recipients of alemtuzumab-based reduced-intensity HSCT and collated details on CMV infection episodes and T-cell chimerism. We used CMV-specific HLA multimers to enumerate CMV-specific T-cell numbers and select cells to assess chimerism status in a subset of R+/D- and R+/seropositive donor patients. We show that in R+/D- patients, CMV-specific T cells are exclusively of recipient origin, can protect against recurrent CMV infections, and significantly influence the chimerism status toward recipients. The major findings were replicated in a separate validation cohort. T-cell depletion in the R+/D- setting may actually, therefore, foster more rapid reconstitution of protective antiviral immunity by reducing graft-vs-host directed alloreactivity and the associated elimination of the recipient T-cell compartment. Finally, conversion to donor chimerism after donor lymphocytes is associated with clinically occult transition to donor-derived immunity.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Trasplante de Células Madre Hematopoyéticas , Inmunidad Celular , Depleción Linfocítica , Quimera por Trasplante/inmunología , Aloinjertos , Femenino , Enfermedad Injerto contra Huésped/inmunología , Humanos , Masculino
10.
J Immunol ; 194(1): 125-133, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25404365

RESUMEN

Exhaustion of chronically stimulated CD8(+) T cells is a significant obstacle to immune control of chronic infections or tumors. Although coinhibitory checkpoint blockade with anti-programmed death ligand 1 (PD-L1) Ab can restore functions to exhausted T cell populations, recovery is often incomplete and dependent upon the pool size of a quiescent T-bet(high) subset that expresses lower levels of PD-1. In a model in which unhelped, HY-specific CD8(+) T cells gradually lose function following transfer to male bone marrow transplantation recipients, we have explored the effect of shifting the balance away from coinhibition and toward costimulation by combining anti-PD-L1 with agonistic Abs to the TNFR superfamily members, OX40 and CD27. Several weeks following T cell transfer, both agonistic Abs, but especially anti-CD27, demonstrated synergy with anti-PD-L1 by enhancing CD8(+) T cell proliferation and effector cytokine generation. Anti-CD27 and anti-PD-L1 synergized by downregulating the expression of multiple quiescence-related genes concomitant with a reduced frequency of T-bet(high) cells within the exhausted population. However, in the presence of persistent Ag, the CD8(+) T cell response was not sustained and the overall size of the effector cytokine-producing pool eventually contracted to levels below that of controls. Thus, CD27-mediated costimulation can synergize with coinhibitory checkpoint blockade to switch off molecular programs for quiescence in exhausted T cell populations, but at the expense of losing precursor cells required to maintain a response.


Asunto(s)
Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Glicoproteínas de Membrana/inmunología , Receptores OX40/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Factores de Necrosis Tumoral/inmunología , Traslado Adoptivo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Trasplante de Médula Ósea , Linfocitos T CD8-positivos/trasplante , Femenino , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ligando OX40 , Receptores OX40/genética , Proteínas de Dominio T Box/metabolismo
11.
J Immunol ; 194(3): 1080-9, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25539815

RESUMEN

Ag receptors used for cancer immunotherapy are often directed against tumor-associated Ags also expressed in normal tissues. Targeting of such Ags can result in unwanted autoimmune attack of normal tissues or induction of tolerance in therapeutic T cells. We used a murine model to study the phenotype and function of T cells redirected against the murine double minute protein 2 (MDM2), a tumor-associated Ag that shows low expression in many normal tissues. Transfer of MDM2-TCR-engineered T cells into bone marrow chimeric mice revealed that Ag recognition in hematopoietic tissues maintained T cell function, whereas presentation of MDM2 in nonhematopoietic tissues caused reduced effector function. TCR-engineered CD8(+) T cells underwent rapid turnover, downmodulated CD8 expression, and lost cytotoxic function. We found that MDM2-TCR-engineered CD4(+) T cells provided help and restored cytotoxic function of CD8(+) T cells bearing the same TCR. Although the introduction of the CD8 coreceptor enhanced the ability of CD4(+) T cells to recognize MDM2 in vitro, the improved self-antigen recognition abolished their ability to provide helper function in vivo. The data indicate that the same class I-restricted TCR responsible for Ag recognition and tolerance induction in CD8(+) T cells can, in the absence of the CD8 coreceptor, elicit CD4 T cell help and partially reverse tolerance. Thus MHC class I-restricted CD4(+) T cells may enhance the efficacy of therapeutic TCR-engineered CD8(+) T cells and can be readily generated with the same TCR.


Asunto(s)
Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Tolerancia Inmunológica , Receptores de Antígenos de Linfocitos T/genética , Traslado Adoptivo , Animales , Comunicación Celular , Citotoxicidad Inmunológica , Expresión Génica , Inmunofenotipificación , Ratones , Ratones Transgénicos , Fenotipo , Proteínas Proto-Oncogénicas c-mdm2/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción Genética
12.
Biol Blood Marrow Transplant ; 22(7): 1234-1241, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27095691

RESUMEN

Pretransplant (18)F-fluorodeoxyglucose (FDG) positron emission tomography status is an important prognostic factor for outcomes after autologous stem cell transplantation (SCT) in Hodgkin lymphoma (HL), but its impact on outcomes after allogeneic SCT remains unclear. We retrospectively evaluated outcomes after T cell-depleted allogeneic SCT of 116 patients with nonprogressive HL according to pretransplant Deauville scores. Endpoints were overall survival (OS), progression-free survival (PFS), relapse rate (RR), and nonrelapse-related mortality (NRM). OS, PFS, and RR did not differ significantly between the Deauville 1 to 2 and Deauville 3 to 5 cohorts (OS: 77.5% versus 67.3%, P = .49; PFS: 59.4% versus 55.7%, P = .43; RR: 20.9% versus 22.6%, P = .28 at 4 years). Differences in PFS remained statistically nonsignificant when comparisons were made between Deauville 1 to 3 and Deauville 4 to 5 cohorts (60.9% versus 51.4%, P = .10), and RR remained very similar (21.5% versus 23.8%, P = .42). Multivariate analyses demonstrated trends toward significance for an effect of Deauville score on PFS (hazard ratio 1.82 for Deauville 4 to 5, P = .06) and for number of lines of prior therapy on OS (hazard ratio 2.34 for >5 lines, P = .10). The latter effect appeared to be driven by higher NRM rather than increased RR. Our findings suggest that Deauville score before allogeneic SCT in patients with nonprogressive HL has a relatively modest impact on survival outcomes in comparison with the impact in autologous SCT and that predictive values for the individual patient remain low, indicating that residual FDG-avid disease should not preclude allogeneic SCT. Furthermore, our findings bring into question the importance of attainment of metabolic complete response in this setting if it is at the expense of increasing NRM risk.


Asunto(s)
Enfermedad de Hodgkin/terapia , Tomografía de Emisión de Positrones/mortalidad , Adulto , Toma de Decisiones Clínicas , Femenino , Fluorodesoxiglucosa F18 , Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedad de Hodgkin/diagnóstico por imagen , Enfermedad de Hodgkin/mortalidad , Humanos , Depleción Linfocítica , Masculino , Neoplasia Residual/diagnóstico por imagen , Neoplasia Residual/mortalidad , Neoplasia Residual/terapia , Tomografía de Emisión de Positrones/métodos , Recurrencia , Estudios Retrospectivos , Análisis de Supervivencia , Linfocitos T , Trasplante Autólogo , Trasplante Homólogo , Resultado del Tratamiento
13.
Blood ; 124(8): 1277-87, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-24970931

RESUMEN

A compact marker/suicide gene that utilizes established clinical-grade reagents and pharmaceuticals would be of considerable practical utility to T-cell cancer gene therapy. Marker genes enable measurement of transduction and allow selection of transduced cells, whereas suicide genes allow selective deletion of administered T cells in the face of toxicity. We have created a highly compact marker/suicide gene for T cells combining target epitopes from both CD34 and CD20 antigens (RQR8). This construct allows selection with the clinically approved CliniMACS CD34 system (Miltenyi). Further, the construct binds the widely used pharmaceutical antibody rituximab, resulting in selective deletion of transgene-expressing cells. We have tested the functionality of RQR8 in vitro and in vivo as well as in combination with T-cell engineering components. We predict that RQR8 will make T-cell gene therapy both safer and cheaper.


Asunto(s)
Antígenos CD20/inmunología , Antígenos CD34/inmunología , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Linfocitos T/trasplante , Aloinjertos , Animales , Antígenos CD20/genética , Antígenos CD20/metabolismo , Antígenos CD34/genética , Antígenos CD34/metabolismo , Epítopos , Ratones , Ratones Endogámicos BALB C , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
14.
Haematologica ; 101(4): 482-90, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26802053

RESUMEN

Due to the lack of specificity for tumor antigens, allogeneic T-cell therapy is associated with graft-versus-host disease. Enhancing the anti-tumor specificity while reducing the graft-versus-host disease risk of allogeneic T cells has remained a research focus. In this study, we demonstrate that the introduction of 'dominant' T-cell receptors into primary murine T cells can suppress the expression of endogenous T-cell receptors in a large proportion of the gene-modified T cells. Adoptive transfer of allogeneic T cells expressing a 'dominant' T-cell receptor significantly reduced the graft-versus-host toxicity in recipient mice. Using two bone marrow transplant models, enhanced anti-tumor activity was observed in the presence of reduced graft-versus-host disease. However, although transfer of T-cell receptor gene-modified allogeneic T cells resulted in the elimination of antigen-positive tumor cells and improved the survival of treated mice, it was associated with accumulation of T cells expressing endogenous T-cell receptors and the development of delayed graft-versus-host disease. The in-vivo deletion of the engineered T cells, mediated by endogenous mouse mammary tumor virus MTV8 and MTV9, abolished graft-versus-host disease while retaining significant anti-tumor activity of adoptively transferred T cells. Together, this study shows that the in-vitro selection of allogeneic T cells expressing high levels of a 'dominant' T-cell receptor can lower acute graft-versus-host disease and enhance anti-tumor activity of adoptive cell therapy, while the in-vivo outgrowth of T cells expressing endogenous T-cell receptors remains a risk factor for the delayed onset of graft-versus-host disease.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Inmunoterapia Adoptiva/métodos , Virus del Tumor Mamario del Ratón/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Trasplante de Médula Ósea/métodos , Línea Celular Tumoral , Femenino , Expresión Génica , Genes Dominantes , Vectores Genéticos/inmunología , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Depleción Linfocítica/métodos , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Receptores de Antígenos de Linfocitos T/genética , Análisis de Supervivencia , Linfocitos T/citología , Linfocitos T/inmunología , Transgenes , Trasplante Homólogo , Irradiación Corporal Total
15.
Blood ; 122(19): 3288-97, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24081658

RESUMEN

Although the inhibitory effects of therapeutic glucocorticoids (GCs) on dendritic cells (DCs) are well established, the roles of endogenous GCs in DC homeostasis are less clear. A critical element regulating endogenous GC concentrations involves local conversion of inactive substrates to active 11-hydroxyglucocorticoids, a reduction reaction catalyzed within the endoplasmic reticulum by an enzyme complex containing 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1) and hexose-6-phosphate dehydrogenase (H6PDH). In this study, we found that this GC amplification pathway operates both constitutively and maximally in steady state murine DC populations and is unaffected by additional inflammatory stimuli. Under physiologic conditions, 11ßHSD1-H6PDH increases the sensitivity of plasmacytoid DCs (pDCs) to GC-induced apoptosis and restricts the survival of this population through a cell-intrinsic mechanism. Upon CpG activation, the effects of enzyme activity are overridden, with pDCs becoming resistant to GCs and fully competent to release type I interferon. CD8α(+) DCs are also highly proficient in amplifying GC levels, leading to impaired maturation following toll-like receptor-mediated signaling. Indeed, pharmacologic inhibition of 11ßHSD1 synergized with CpG to enhance specific T-cell responses following vaccination targeted to CD8α(+) DCs. In conclusion, amplification of endogenous GCs is a critical cell-autonomous mechanism for regulating the survival and functions of DCs in vivo.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/inmunología , Deshidrogenasas de Carbohidratos/inmunología , Corticosterona/análogos & derivados , Células Dendríticas/inmunología , Receptores de Glucocorticoides/inmunología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Animales , Apoptosis/efectos de los fármacos , Trasplante de Médula Ósea , Antígenos CD8/genética , Antígenos CD8/inmunología , Deshidrogenasas de Carbohidratos/genética , Células Cultivadas , Corticosterona/metabolismo , Corticosterona/farmacología , Ciclopropanos/farmacología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Guanosina/análogos & derivados , Guanosina/farmacología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Receptores de Glucocorticoides/genética , Transducción de Señal , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Irradiación Corporal Total
16.
Trends Immunol ; 33(1): 8-13, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22030236

RESUMEN

Dendritic cells (DCs) prime and orchestrate naïve T cell immunity in lymphoid organs, but recent data also highlight the importance of DC-effector T cell interactions in tissues. These studies suggest that effector T cells require a second activating step in situ from tissue DCs to become fully competent for effector functions and/or proliferation and survival. DC stimulation of effector T cells within tissues has evolved as a mechanism to ensure that T cells are activated to their full potential only at the site of ongoing infection. Here, we propose that under conditions of uncontrolled inflammation and release of tissue antigens, the same DC-dependent checkpoint perpetuates a destructive response and immunopathology.


Asunto(s)
Células Dendríticas/inmunología , Animales , Humanos , Memoria Inmunológica , Inflamación/inmunología , Activación de Linfocitos , Linfocitos T/inmunología
18.
J Clin Invest ; 134(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38557497

RESUMEN

Refractory acute graft-versus-host disease (GVHD) occurs when the immune injury exceeds the capacity of injured tissues to regenerate and repair. While glucocorticoids have been used for decades to treat GVHD, Arnhold, Chang, and colleagues in this issue of the JCI question whether this approach can in fact be counterproductive. Using in vivo experimental models of GVHD and in vitro intestinal organoids, the study authors show that glucocorticoid exposure directly impeded small intestinal epithelial proliferation and survival, thus preventing the resolution of injury. These findings suggest that future treatment approaches for acute GVHD should include measures to reduce immune reactivity as well as interventions to actively promote tissue resilience.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Humanos , Trasplante Homólogo , Intención , Intestinos , Intestino Delgado , Enfermedad Injerto contra Huésped/terapia , Enfermedad Aguda
19.
Br J Haematol ; 160(5): 640-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23293871

RESUMEN

Following reduced intensity-conditioned allogeneic stem cell transplantation (RIC allo-SCT) for chronic lymphocytic leukaemia (CLL), there is an inverse relationship between relapse and extensive chronic graft-versus-host disease (GVHD). We evaluated outcomes in 50 consecutive patients with CLL using the approach of alemtuzumab-based RIC allo-SCT and pre-emptive donor lymphocyte infusions (DLI) for mixed chimerism or minimal residual disease (MRD), with the intention of reducing the risk of GVHD. Forty two patients had high-risk disease, including 30% with 17p deletion (17p-). Of patients who were not in complete remission (CR) entering transplant, 83% subsequently achieved MRD-negative CR. Both MRD detection and uncorrected mixed chimerism were associated with greater risks of treatment failure. Nine of sixteen patients receiving DLI for persistent or relapsed disease subsequently attained MRD-negative CR. With a median follow-up of 4.3 years, 4-year current progression-free survival was 65% and overall survival was 75% (60% and 61% in respectively, patients with 17p-). DLI was associated with a 29% cumulative incidence of severe GVHD and mortality of 6.4%. At last follow-up, 83% of patients in CR were off all immunosuppressive treatment. In conclusion, the directed delivery of allogeneic cellular therapy has the potential to induce durable remissions in high-risk CLL without incurring excessive GVHD.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Inmunoterapia Adoptiva , Leucemia Linfocítica Crónica de Células B/terapia , Adulto , Alemtuzumab , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ensayos Clínicos como Asunto/estadística & datos numéricos , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/epidemiología , Humanos , Inmunosupresores/uso terapéutico , Estimación de Kaplan-Meier , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/mortalidad , Leucemia Linfocítica Crónica de Células B/cirugía , Procedimientos de Reducción del Leucocitos , Transfusión de Linfocitos , Masculino , Persona de Mediana Edad , Estudios Multicéntricos como Asunto/estadística & datos numéricos , Neoplasia Residual , Pronóstico , Recurrencia , Estudios Retrospectivos , Medición de Riesgo , Terapia Recuperativa , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Resultado del Tratamiento
20.
Blood ; 117(26): 7063-9, 2011 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-21566096

RESUMEN

Langerhans cells (LCs) are a distinct population of dendritic cells that form a contiguous network in the epidermis of the skin. Although LCs possess many of the properties of highly proficient dendritic cells, recent studies have indicated that they are not necessary to initiate cutaneous immunity. In this study, we used a tractable model of cutaneous GVHD, induced by topical application of a Toll-like receptor agonist, to explore the role of LCs in the development of tissue injury. By adapting this model to permit inducible and selective depletion of host LCs, we found that GVHD was significantly reduced when LCs were absent. However, LCs were not required either for CD8 T-cell activation within the draining lymph node or subsequent homing of effector cells to the epidermis. Instead, we found that LCs were necessary for inducing transcription of IFN-γ and other key effector molecules by donor CD8 cells in the epidermis, indicating that they license CD8 cells to induce epithelial injury. These data demonstrate a novel regulatory role for epidermal LCs during the effector phase of an inflammatory immune response in the skin.


Asunto(s)
Comunicación Celular , Citotoxicidad Inmunológica , Epidermis/inmunología , Epidermis/patología , Células de Langerhans/inmunología , Linfocitos T Citotóxicos/inmunología , Aminoquinolinas/toxicidad , Animales , Células Cultivadas , Quimera , Epidermis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedad Injerto contra Huésped/inmunología , Granzimas/genética , Granzimas/metabolismo , Imiquimod , Interferón gamma/genética , Interferón gamma/metabolismo , Células de Langerhans/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/metabolismo , Linfocitos T Citotóxicos/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores Toll-Like/agonistas , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA