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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 17(1): 321, 2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31547819

RESUMEN

BACKGROUND: Medulloblastoma (MB), the most common pediatric brain cancer, presents with a poor prognosis in a subset of patients with high risk disease, or at recurrence, where current therapies are ineffective. Cord blood (CB) natural killer (NK) cells may be promising off-the-shelf effector cells for immunotherapy due to their recognition of malignant cells without the need for a known target, ready availability from multiple banks, and their potential to expand exponentially. However, they are currently limited by immune suppressive cytokines secreted in the MB tumor microenvironment including Transforming Growth Factor ß (TGF-ß). Here, we address this challenge in in vitro models of MB. METHODS: CB-derived NK cells were modified to express a dominant negative TGF-ß receptor II (DNRII) using retroviral transduction. The ability of transduced CB cells to maintain function in the presence of medulloblastoma-conditioned media was then assessed. RESULTS: We observed that the cytotoxic ability of nontransduced CB-NK cells was reduced in the presence of TGF-ß-rich, medulloblastoma-conditioned media (21.21 ± 1.19% killing at E:T 5:1 in the absence vs. 14.98 ± 2.11% in the presence of medulloblastoma-conditioned media, n = 8, p = 0.02), but was unaffected in CB-derived DNRII-transduced NK cells (21.11 ± 1.84% killing at E:T 5:1 in the absence vs. 21.81 ± 3.37 in the presence of medulloblastoma-conditioned media, n = 8, p = 0.85. We also observed decreased expression of CCR2 in untransduced NK cells (mean CCR2 MFI 826 ± 117 in untransduced NK + MB supernatant from mean CCR2 MFI 1639.29 ± 215 in no MB supernatant, n = 7, p = 0.0156), but not in the transduced cells. Finally, we observed that CB-derived DNRII-transduced NK cells may protect surrounding immune cells by providing a cytokine sink for TGF-ß (decreased TGF-ß levels of 610 ± 265 pg/mL in CB-derived DNRII-transduced NK cells vs. 1817 ± 342 pg/mL in untransduced cells; p = 0.008). CONCLUSIONS: CB NK cells expressing a TGF-ß DNRII may have a functional advantage over unmodified NK cells in the presence of TGF-ß-rich MB, warranting further investigation on its potential applications for patients with medulloblastoma.


Asunto(s)
Neoplasias Cerebelosas/inmunología , Células Asesinas Naturales/inmunología , Meduloblastoma/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Sangre Fetal/citología , Humanos , Células Asesinas Naturales/trasplante , Pruebas de Neutralización , Receptores CCR2/metabolismo , Trasplante Homólogo
2.
Br J Haematol ; 177(3): 457-466, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28295190

RESUMEN

Multiple myeloma (MM) is a disease with known immune dysregulation. Natural killer (NK) cells have shown preclinical activity in MM. We conducted a first-in-human study of umbilical cord blood-derived (CB) NK cells for MM patients undergoing high dose chemotherapy and autologous haematopoietic stem cell transplantation (auto-HCT). Patients received lenalidomide (10 mg) on days -8 to -2, melphalan 200 mg/m2 on day -7, CB-NK cells on day -5 and auto-HCT on day 0. Twelve patients were enrolled, three on each of four CB-NK cell dose levels: 5 × 106 , 1 × 107 , 5 × 107 and 1 × 108 CB-NK cells/kg. Ten patients had either high-risk chromosomal changes or a history of relapsed/progressed disease. There were no infusional toxicities and no graft-versus-host disease. One patient failed to engraft due to poor autologous graft quality and was rescued with a back-up autologous graft. Overall, 10 patients achieved at least a very good partial response as their best response, including eight with near complete response or better. With a median follow-up of 21 months, four patients have progressed or relapsed, two of whom have died. CB-NK cells were detected in vivo in six patients, with an activated phenotype (NKG2D+ /NKp30+ ). These data warrant further development of this novel cellular therapy.


Asunto(s)
Sangre Fetal/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Asesinas Naturales/trasplante , Mieloma Múltiple/terapia , Traslado Adoptivo/métodos , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Separación Celular/métodos , Terapia Combinada , Femenino , Citometría de Flujo/métodos , Supervivencia de Injerto , Humanos , Lenalidomida , Masculino , Melfalán/administración & dosificación , Persona de Mediana Edad , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Resultado del Tratamiento
3.
Blood ; 125(9): 1502-6, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25428215

RESUMEN

Adoptive therapy with regulatory T cells (Tregs) to prevent graft-versus-host disease (GVHD) would benefit from a strategy to improve homing to the sites of inflammation. We hypothesized that adding fucose to human Tregs, forming the Sialyl Lewis X moiety on P-selectin glycoprotein ligand-1, would improve their trafficking pattern. The selectin pathway recruiter, α-1,3-fucosyltransferase-VI enzyme, significantly increased Treg surface fucosylation (66% vs 8%). In a xenogenic GVHD mouse model, fucosylated Tregs showed prolonged periods of in vivo persistence. When given at a lower dose compared with the untreated Tregs, the murine recipients of fucosylated Tregs maintained weight, had ameliorated clinical GVHD, and improved survival (70% vs 30%; P < .0001). These preclinical data indicate that fucosylated human Tregs is an effective strategy for prevention of GVHD and, as such, warrants consideration for future clinical trials.


Asunto(s)
Modelos Animales de Enfermedad , Fucosa/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Linfocitos T Reguladores/inmunología , Animales , Western Blotting , Células Cultivadas , Selectina E/metabolismo , Femenino , Sangre Fetal/citología , Sangre Fetal/inmunología , Sangre Fetal/metabolismo , Citometría de Flujo , Fucosiltransferasas/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Oligosacáridos/metabolismo , Antígeno Sialil Lewis X
4.
Blood ; 125(19): 2885-92, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25778529

RESUMEN

Delayed engraftment is a major limitation of cord blood transplantation (CBT), due in part to a defect in the cord blood (CB) cells' ability to home to the bone marrow. Because this defect appears related to low levels of fucosylation of cell surface molecules that are responsible for binding to P- and E-selectins constitutively expressed by the marrow microvasculature, and thus for marrow homing, we conducted a first-in-humans clinical trial to correct this deficiency. Patients with high-risk hematologic malignancies received myeloablative therapy followed by transplantation with 2 CB units, one of which was treated ex vivo for 30 minutes with the enzyme fucosyltransferase-VI and guanosine diphosphate fucose to enhance the interaction of CD34(+) stem and early progenitor cells with microvessels. The results of enforced fucosylation for 22 patients enrolled in the trial were then compared with those for 31 historical controls who had undergone double unmanipulated CBT. The median time to neutrophil engraftment was 17 days (range, 12-34 days) compared with 26 days (range, 11-48 days) for controls (P = .0023). Platelet engraftment was also improved: median was 35 days (range, 18-100 days) compared with 45 days (range, 27-120 days) for controls (P = .0520). These findings support ex vivo fucosylation of multipotent CD34(+) CB cells as a clinically feasible means to improve engraftment efficiency in the double CBT setting. The trial is registered to www.clinicaltrials.gov as #NCT01471067.


Asunto(s)
Plaquetas/citología , Sangre Fetal/citología , Fucosa/metabolismo , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Neutrófilos/trasplante , Adolescente , Adulto , Anciano , Plaquetas/inmunología , Estudios de Cohortes , Selectina E/metabolismo , Estudios de Factibilidad , Femenino , Sangre Fetal/inmunología , Fucosiltransferasas/metabolismo , Enfermedad Injerto contra Huésped , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/mortalidad , Células Madre Hematopoyéticas/inmunología , Humanos , Masculino , Persona de Mediana Edad , Neutrófilos/citología , Neutrófilos/inmunología , Selectina-P/metabolismo , Transfusión de Plaquetas , Pronóstico , Tasa de Supervivencia , Adulto Joven
5.
Cytotherapy ; 19(3): 408-418, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28109751

RESUMEN

Cord blood (CB) natural killer (NK) cells are promising effector cells for tumor immunotherapy but are currently limited by immune-suppressive cytokines in the tumor microenvironment, such as transforming growth factor (TGF-ß). We observed that TGF-ß inhibits expression of activating receptors such as NKG2D and DNAM1 and decreases killing activity against glioblastoma tumor cells through inhibition of perforin secretion. To overcome the detrimental effects of TGF-ß, we engrafted a dominant negative TGF-ß receptor II (DNRII) on CB-derived NK cells by retroviral transduction and evaluated their ability to kill glioblastoma cells in the presence of TGF-ß. After manufacture using Good Manufacturing Practice-compliant methodologies and transduction with DNRII, CB-derived DNRII-transduced NK cells expanded to clinically relevant numbers and retained both their killing ability and their secretion of interferon-γ upon activation. More important, these cells maintained both perforin expression and NKG2D/DNMA1 expression in the presence of TGF-ß allowing for recognition and killing of glioblastoma tumor cells. Hence, NK cells expressing a DNRII should have a functional advantage over unmodified NK cells in the presence of TGF-ß-secreting tumors and may be an important therapeutic approach for patients with cancer.


Asunto(s)
Neoplasias Encefálicas/terapia , Sangre Fetal/citología , Terapia Genética/métodos , Glioblastoma/terapia , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/trasplante , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Neoplasias Encefálicas/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Sangre Fetal/inmunología , Sangre Fetal/trasplante , Genes Dominantes , Glioblastoma/inmunología , Humanos , Interferón gamma/metabolismo , Células Jurkat , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Perforina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/metabolismo
6.
Mol Ther ; 24(6): 1135-1149, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27019998

RESUMEN

Chimeric antigen receptor (CAR) T cells have shown great promise in the treatment of hematologic malignancies but more variable results in the treatment of solid tumors and the persistence and expansion of CAR T cells within patients has been identified as a key correlate of antitumor efficacy. Lack of immunological "space", functional exhaustion, and deletion have all been proposed as mechanisms that hamper CAR T-cell persistence. Here we describe the events following activation of third-generation CAR T cells specific for GD2. CAR T cells had highly potent immediate effector functions without evidence of functional exhaustion in vitro, although reduced cytokine production reversible by PD-1 blockade was observed after longer-term culture. Significant activation-induced cell death (AICD) of CAR T cells was observed after repeated antigen stimulation, and PD-1 blockade enhanced both CAR T-cell survival and promoted killing of PD-L1(+) tumor cell lines. Finally, we assessed CAR T-cell persistence in patients enrolled in the CARPETS phase 1 clinical trial of GD2-specific CAR T cells in the treatment of metastatic melanoma. Together, these data suggest that deletion also occurs in vivo and that PD-1-targeted combination therapy approaches may be useful to augment CAR T-cell efficacy and persistence in patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Melanoma/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/trasplante , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Gangliósidos/inmunología , Humanos , Activación de Linfocitos , Melanoma/inmunología , Ratones , Metástasis de la Neoplasia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Blood ; 124(18): 2824-33, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25049283

RESUMEN

Advances in the design of chimeric antigen receptors (CARs) have improved the antitumor efficacy of redirected T cells. However, functional heterogeneity of CAR T cells limits their therapeutic potential and is associated with toxicity. We proposed that CAR expression in Vα24-invariant natural killer T (NKT) cells can build on the natural antitumor properties of these cells while their restriction by monomorphic CD1d limits toxicity. Primary human NKT cells were engineered to express a CAR against the GD2 ganglioside (CAR.GD2), which is highly expressed by neuroblastoma (NB). We compared CAR.GD2 constructs that encoded the CD3ζ chain alone, with CD28, 4-1BB, or CD28 and 4-1BB costimulatory endodomains. CAR.GD2 expression rendered NKT cells highly cytotoxic against NB cells without affecting their CD1d-dependent reactivity. We observed a striking T helper 1-like polarization of NKT cells by 4-1BB-containing CARs. Importantly, expression of both CD28 and 4-1BB endodomains in the CAR.GD2 enhanced in vivo persistence of NKT cells. These CAR.GD2 NKT cells effectively localized to the tumor site had potent antitumor activity, and repeat injections significantly improved the long-term survival of mice with metastatic NB. Unlike T cells, CAR.GD2 NKT cells did not induce graft-versus-host disease. These results establish the potential of NKT cells to serve as a safe and effective platform for CAR-directed cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunoterapia , Células T Asesinas Naturales/inmunología , Neuroblastoma/inmunología , Neuroblastoma/terapia , Receptores de Antígenos/inmunología , Ligando 4-1BB/química , Ligando 4-1BB/metabolismo , Animales , Antígenos CD1d/metabolismo , Antígenos CD28/metabolismo , Línea Celular , Proliferación Celular , Citocinas/metabolismo , Citotoxicidad Inmunológica , Gangliósidos , Enfermedad Injerto contra Huésped/inmunología , Humanos , Activación de Linfocitos/inmunología , Macrófagos/metabolismo , Ratones , Metástasis de la Neoplasia , Neuroblastoma/patología , Estructura Terciaria de Proteína , Retroviridae/genética , Transducción Genética , Resultado del Tratamiento
8.
Blood ; 124(13): 2034-45, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25051962

RESUMEN

A subset of regulatory B cells (Bregs) in mice negatively regulate T-cell immune responses through the secretion of regulatory cytokines such as IL-10 and direct cell-cell contact and have been linked to experimental models of autoimmunity, inflammation, and cancer. However, the regulatory function of Bregs in human disease is much less clear. Here we demonstrate that B cells with immunoregulatory properties are enriched within both the CD19(+)IgM(+)CD27(+) memory and CD19(+)CD24(hi)CD38(hi) transitional B-cell subsets in healthy human donors. Both subsets suppressed the proliferation and interferon-γ production of CD3/CD28-stimulated autologous CD4(+) T cells in a dose-dependent manner, and both relied on IL-10 secretion as well as cell-cell contact, likely mediated through CD80 and CD86, to support their full suppressive function. Moreover, after allogeneic stem cell transplantation, Bregs from patients with chronic graft-versus-host disease (cGVHD) were less frequent and less likely to produce IL-10 than were Bregs from healthy donors and patients without cGVHD. These findings suggest that Bregs may be involved in the pathogenesis of cGVHD and support future investigation of regulatory B cell-based therapy in the treatment of this disease.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B Reguladores/inmunología , Enfermedad Injerto contra Huésped/inmunología , Inmunoglobulina M/inmunología , Memoria Inmunológica , ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD19/metabolismo , Subgrupos de Linfocitos B/metabolismo , Linfocitos B Reguladores/metabolismo , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Antígeno CD24/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Estudios de Casos y Controles , Comunicación Celular , Células Cultivadas , Enfermedad Crónica , Citocinas/biosíntesis , Enfermedad Injerto contra Huésped/metabolismo , Humanos , Inmunomodulación , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Interleucina-10/biosíntesis , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral
9.
Cytotherapy ; 18(11): 1410-1421, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27421740

RESUMEN

Natural killer (NK) cells are members of the innate immune system that recognize target cells via activating and inhibitory signals received through cell receptors. Derived from the lymphoid lineage, NK cells are able to produce cytokines and exert a cytotoxic effect on viral infected and malignant cells. It is their unique ability to lyse target cells rapidly and without prior education that renders NK cells a promising effector cell for adoptive cell therapy. However, both viruses and tumors employ evasion strategies to avoid attack by NK cells, which represent biological challenges that need to be harnessed to fully exploit the cytolytic potential of NK cells. Using genetic modification, the function of NK cells can be enhanced to improve their homing, cytolytic activity, in vivo persistence and safety. Examples include gene modification to express chemokine, high-affinity Fc receptor and chimeric antigen receptors, suicide genes and the forced expression of cytokines such as interleukin (IL)-2 and IL-15. Preclinical studies have clearly demonstrated that such approaches are effective in improving NK-cell function, homing and safety. In this review, we summarize the recent advances in the genetic manipulations of NK cells and their application for cellular immunotherapeutic strategies.


Asunto(s)
Ingeniería Genética , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Humanos , Evasión Inmune , Resultado del Tratamiento
10.
Cytotherapy ; 18(10): 1312-24, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27497700

RESUMEN

Regulatory T cells (Tregs) play a fundamental role in the maintenance of self-tolerance and immune homeostasis. Defects in Treg function and/or frequencies have been reported in multiple disease models. Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder affecting upper and lower motor neurons. Compelling evidence supports a neuroprotective role for Tregs in this disease. Indeed, rapid progression in ALS patients is associated with decreased FoxP3 expression and Treg frequencies. Thus, we propose that strategies to restore Treg number and function may slow disease progression in ALS. In this study, we developed a robust, Good Manufacturing Practice (GMP)-compliant procedure to enrich and expand Tregs from ALS patients. Tregs isolated from these patients were phenotypically similar to those from healthy individuals but were impaired in their ability to suppress T-cell effector function. In vitro expansion of Tregs for 4 weeks in the presence of GMP-grade anti-CD3/CD28 beads, interleukin (IL)-2 and rapamcyin resulted in a 25- to 200-fold increase in their number and restored their immunoregulatory activity. Collectively, our data facilitate and support the implementation of clinical trials of adoptive therapy with ex vivo expanded and highly suppressive Tregs in patients with ALS.


Asunto(s)
Traslado Adoptivo/normas , Esclerosis Amiotrófica Lateral/patología , Separación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/normas , Cultivo Primario de Células , Linfocitos T Reguladores/patología , Traslado Adoptivo/métodos , Esclerosis Amiotrófica Lateral/inmunología , Estudios de Casos y Controles , Separación Celular/métodos , Separación Celular/normas , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Adhesión a Directriz/normas , Humanos , Tolerancia Inmunológica , Interleucina-2/metabolismo , Cultivo Primario de Células/métodos , Cultivo Primario de Células/normas , Linfocitos T Reguladores/inmunología
11.
Cytotherapy ; 16(1): 90-100, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24480547

RESUMEN

BACKGROUND AIMS: Naturally occurring regulatory T cells (Treg) are emerging as a promising approach for prevention of graft-versus-host disease (GvHD), which remains an obstacle to the successful outcome of allogeneic hematopoietic stem cell transplantation. However, Treg only constitute 1-5% of total nucleated cells in cord blood (CB) (<3 × 106 cells), and therefore novel methods of Treg expansion to generate clinically relevant numbers are needed. METHODS: Several methodologies are currently being used for ex vivo Treg expansion. We report a new approach to expand Treg from CB and demonstrate their efficacy in vitro by blunting allogeneic mixed lymphocyte reactions and in vivo by preventing GvHD through the use of a xenogenic GvHD mouse model. RESULTS: With the use of magnetic cell sorting, naturally occurring Treg were isolated from CB by the positive selection of CD25⁺ cells. These were expanded to clinically relevant numbers by use of CD3/28 co-expressing Dynabeads and interleukin (IL)-2. Ex vivo-expanded Treg were CD4⁺25⁺ FOXP3⁺127(lo) and expressed a polyclonal T-cell receptor, Vß repertoire. When compared with conventional T-lymphocytes (CD4⁺25⁻ cells), Treg consistently showed demethylation of the FOXP3 TSDR promoter region and suppression of allogeneic proliferation responses in vitro. CONCLUSIONS: In our NOD-SCID IL-2Rγ(null) xenogeneic model of GvHD, prophylactic injection of third-party, CB-derived, ex vivo-expanded Treg led to the prevention of GvHD that translated into improved GvHD score, decreased circulating inflammatory cytokines and significantly superior overall survival. This model of xenogenic GvHD can be used to study the mechanism of action of CB Treg as well as other therapeutic interventions.


Asunto(s)
Sangre Fetal/trasplante , Enfermedad Injerto contra Huésped/terapia , Linfocitos T Reguladores/citología , Trasplante Homólogo/efectos adversos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Sangre Fetal/citología , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Ratones , Linfocitos T Reguladores/trasplante
12.
Mol Ther ; 21(11): 2087-101, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23939024

RESUMEN

Preclinical and early clinical studies have demonstrated that chimeric antigen receptor (CAR)-redirected T cells are highly promising in cancer therapy. We observed that targeting HER2 in a glioblastoma (GBM) cell line results in the emergence of HER2-null tumor cells that maintain the expression of nontargeted tumor-associated antigens. Combinational targeting of these tumor-associated antigens could therefore offset this escape mechanism. We studied the single-cell coexpression patterns of HER2, IL-13Rα2, and EphA2 in primary GBM samples using multicolor flow cytometry and immunofluorescence, and applied a binomial routine to the permutations of antigen expression and the related odds of complete tumor elimination. This mathematical model demonstrated that cotargeting HER2 and IL-13Rα2 could maximally expand the therapeutic reach of the T cell product in all primary tumors studied. Targeting a third antigen did not predict an added advantage in the tumor cohort studied. We therefore generated bispecific T cell products from healthy donors and from GBM patients by pooling T cells individually expressing HER2 and IL-13Rα2-specific CARs and by making individual T cells to coexpress both molecules. Both HER2/IL-13Rα2-bispecific T cell products offset antigen escape, producing enhanced effector activity in vitro immunoassays (against autologous glioma cells in the case of GBM patient products) and in an orthotopic xenogeneic murine model. Further, T cells coexpressing HER2 and IL-13Rα2-CARs exhibited accentuated yet antigen-dependent downstream signaling and a particularly enhanced antitumor activity.


Asunto(s)
Traslado Adoptivo , Antígenos de Neoplasias/metabolismo , Glioblastoma/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Terapia Combinada , Glioblastoma/inmunología , Glioblastoma/patología , Células HEK293 , Humanos , Subunidad alfa2 del Receptor de Interleucina-13/genética , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Ratones , Ratones SCID , Modelos Biológicos , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas , Escape del Tumor , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Blood ; 118(23): 6050-6, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21984804

RESUMEN

We generated MHC-independent chimeric antigen receptors (CARs) directed to the GD2 antigen expressed by neuroblastoma tumor cells and treated patients with this disease. Two distinguishable forms of this CAR were expressed in EBV-specific cytotoxic T lymphocytes (EBV-CTLs) and activated T cells (ATCs). We have previously shown that EBV-CTLs expressing GD2-CARs (CAR-CTLs) circulated at higher levels than GD2-CAR ATCs (CAR-ATCs) early after infusion, but by 6 weeks, both subsets became low or undetectable. We now report the long-term clinical and immunologic consequences of infusions in 19 patients with high-risk neuroblastoma: 8 in remission at infusion and 11 with active disease. Three of 11 patients with active disease achieved complete remission, and persistence of either CAR-ATCs or CAR-CTLs beyond 6 weeks was associated with superior clinical outcome. We observed persistence for up to 192 weeks for CAR-ATCs and 96 weeks for CAR-CTLs, and duration of persistence was highly concordant with the percentage of CD4(+) cells and central memory cells (CD45RO(+)CD62L(+)) in the infused product. In conclusion, GD2-CAR T cells can induce complete tumor responses in patients with active neuroblastoma; these CAR T cells may have extended, low-level persistence in patients, and such persistence was associated with longer survival. This study is registered at www.clinialtrials.gov as #NCT00085930.


Asunto(s)
Traslado Adoptivo/métodos , Neoplasias Óseas/terapia , Gangliósidos/inmunología , Terapia Genética/métodos , Neuroblastoma/terapia , Receptores de Antígenos de Linfocitos T/genética , Adolescente , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Neoplasias de la Médula Ósea/genética , Neoplasias de la Médula Ósea/inmunología , Neoplasias de la Médula Ósea/terapia , Neoplasias Óseas/genética , Neoplasias Óseas/inmunología , Línea Celular Tumoral , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Memoria Inmunológica/inmunología , Masculino , Neoplasia Residual/genética , Neoplasia Residual/inmunología , Neoplasia Residual/terapia , Neuroblastoma/genética , Neuroblastoma/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T Citotóxicos/inmunología , Adulto Joven
14.
Mol Ther ; 19(12): 2239-48, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21878902

RESUMEN

Despite activity as single agent cancer therapies, Rapamycin (rapa) and its rapalogs may have their greatest effects when combined with other therapeutic modalities. In addition to direct antitumor activity, rapalogs reverse multiple tumor-intrinsic immune evasion mechanisms. These should facilitate tumor-specific T cell activity, but since rapa directly inhibits effector T cells, this potential immune enhancement is lost. We hypothesized that if T cells were rendered resistant to rapa they could capitalize on its downregulation of tumor immune evasion. We therefore modified T cells with a rapa-resistant mutant of mTor, mTorRR, and directed them to B lymphomas by coexpressing a chimeric antigen receptor (CAR) for CD19 (CAR.CD19-28ζ). T cells expressing transgenic mTorRR from a piggyBac transposon maintain mTor signaling, proliferate in the presence of rapa and retain their cytotoxic function and ability to secrete interferon-γ (IFNγ) after stimulation, effector functions that were inhibited by rapa in control T cells. In combination, rapa and rapa-resistant-CAR.CD19-28ζ-expressing T cells produced greater antitumor activity against Burkitt's lymphoma and pre-B ALL cell lines in vitro than CAR.CD19-28ζ T cells or rapa alone. In conclusion, the combination of rapa and rapa-resistant, CAR.CD19-28ζ-expressing T cells may provide a novel therapy for the treatment of B cell malignancies and other cancers.


Asunto(s)
Linfoma de Burkitt/prevención & control , Resistencia a Antineoplásicos , Inmunosupresores/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/prevención & control , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antígenos CD19/genética , Antígenos CD19/metabolismo , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Western Blotting , Linfoma de Burkitt/genética , Linfoma de Burkitt/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Terapia Combinada , Femenino , Citometría de Flujo , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
15.
Nat Commun ; 13(1): 2155, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35444206

RESUMEN

T cell factor 1 (TCF1) is required for memory and stem-like CD8+ T cell functions. How TCF1 partners with other transcription factors to regulate transcription remains unclear. Here we show that negative elongation factor (NELF), an RNA polymerase II (Pol II) pausing factor, cooperates with TCF1 in T cell responses to cancer. Deletion of mouse Nelfb, which encodes the NELFB subunit, in mature T lymphocytes impairs immune responses to both primary tumor challenge and tumor antigen-mediated vaccination. Nelfb deletion causes more exhausted and reduced memory T cell populations, whereas its ectopic expression boosts antitumor immunity and efficacy of chimeric antigen receptor T-cell immunotherapy. Mechanistically, NELF is associated with TCF1 and recruited preferentially to the enhancers and promoters of TCF1 target genes. Nelfb ablation reduces Pol II pausing and chromatin accessibility at these TCF1-associated loci. Our findings thus suggest an important and rate-limiting function of NELF in anti-tumor immunity.


Asunto(s)
Linfocitos T CD8-positivos , ARN Polimerasa II , Animales , Ratones , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Mol Ther Methods Clin Dev ; 27: 415-430, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36381305

RESUMEN

Cord blood (CB)-derived natural killer (NK) cells that are genetically engineered to express a chimeric antigen receptor (CAR) are an attractive off-the-shelf therapy for the treatment of cancer, demonstrating a robust safety profile in vivo. For poor prognosis brain tumors such as glioblastoma multiforme (GBM), novel therapies are urgently needed. Although CAR-T cells demonstrate efficacy in preclinical GBM models, an off-the-shelf product may exhibit unwanted side effects like graft-versus-host disease. Hence, we developed an off-the-shelf CAR-NK cell approach using a B7H3 CAR and showed that CAR-transduced NK cells have robust cytolytic activity against GBM cells in vitro. However, transforming growth factor (TGF)-ß within the tumor microenvironment has devastating effects on the cytolytic activity of both unmodified and CAR-transduced NK cells. To overcome this potent immune suppression, we demonstrated that co-transducing NK cells with a B7H3 CAR and a TGF-ß dominant negative receptor (DNR) preserves cytolytic function in the presence of exogenous TGF-ß. This study demonstrates that a novel DNR and CAR co-expression strategy may be a promising therapeutic for recalcitrant CNS tumors like GBM.

17.
Nano Res ; 15(3): 2300-2314, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36089987

RESUMEN

Despite the promise of immunotherapy such as the immune checkpoint inhibitors (ICIs) anti-PD-1 and anti-CTLA-4 for advanced melanoma, only 26%-52% of patients respond, and many experience grade III/IV immune-related adverse events. Motivated by the need for an effective therapy for patients non-responsive to clinically approved ICIs, we have developed a novel nanoimmunotherapy that combines locally administered Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) with systemically administered agonistic anti-CD137 monoclonal antibody therapy (aCD137). PBNP-PTT was administered at various thermal doses to melanoma cells in vitro, and was combined with aCD137 in vivo to test treatment effects on melanoma tumor progression, animal survival, immunological protection against tumor rechallenge, and hepatotoxicity. When administered at a melanoma-specific thermal dose, PBNP-PTT elicits immunogenic cell death (ICD) in melanoma cells and upregulates markers associated with antigen presentation and immune cell co-stimulation in vitro. Consequently, PBNP-PTT eliminates primary melanoma tumors in vivo, yielding long-term tumor-free survival. However, the antitumor immune effects generated by PBNP-PTT cannot eliminate secondary tumors, despite significantly slowing their growth. The addition of aCD137 enables significant abscopal efficacy and improvement of survival, functioning through activated dendritic cells and tumor-infiltrating CD8+ T cells, and generates CD4+ and CD8+ T cell memory that manifests in the rejection of tumor rechallenge, with no long-term hepatotoxicity. This study describes for the first time a novel and effective nanoimmunotherapy combination of PBNP-PTT with aCD137 mAb therapy for melanoma.

18.
Stem Cells ; 28(6): 1107-15, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20506146

RESUMEN

Mesenchymal stromal cells (MSCs) have been infused in hundreds of patients to date, with minimal reported side effects. However, follow-up is limited and long-term side effects are unknown. Because several animal models have raised safety concerns, we sought to develop a system allowing control over the growth and survival of MSCs used therapeutically. We have previously described a suicide system based on an inducible caspase-9 (iCasp9) protein that is activated using a specific chemical inducer of dimerization (CID), analogs of which have been safely tested in a phase I study. Here, we show that MSCs can be easily transduced with this system and selected to high purity (greater than 97%) with clinical grade immunomagnetic procedures. The transduced cells maintain their basic physiology, including expression of surface antigens (such as positivity for CD73, CD90, and CD105, and negativity for hematopoietic markers) and their potential to differentiate into diverse connective tissue lineages (adipocytes, osteoblasts, and chondroblasts). Those cells and their differentiated progeny can be selectively eliminated in vitro or in vivo within 24 hours after exposure to pharmacological levels of CID, with evidence of apoptosis in more than 95% of iCasp9-positive cells. In conclusion, we have developed directed MSC killing to provide a necessary safety mechanism for therapies using progenitor cells. We believe that this approach will become of increasing value as clinical applications for MSCs develop further.


Asunto(s)
Caspasa 9/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Animales , Antígenos CD19/genética , Antígenos CD19/metabolismo , Apoptosis , Biomarcadores , Caspasa 9/genética , Diferenciación Celular , Células Cultivadas , Genes Transgénicos Suicidas , Humanos , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Ratones , Ratones SCID
19.
Cytotherapy ; 13(9): 1128-39, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21745159

RESUMEN

BACKGROUND AIMS: Several studies have demonstrated that the immunogenicity of chronic lymphocytic leukemia (CLL) cells can be increased by manipulation of the CD40/CD40-ligand (CD40L) pathway. Although immunologic, and perhaps clinical, benefits have been obtained with an autologous CLL tumor vaccine obtained by transgenic expression of CD40L and interleukin (IL)-2, there is little information about the optimal gene transfer strategies. METHODS: We compared two different CLL vaccines prepared by adenoviral gene transfer and plasmid electroporation, analyzing their phenotype and immunostimulatory activity. RESULTS: We found that higher expression of transgenic CD40L was mediated by adenoviral gene transfer than by plasmid transduction, and that adenoviral transfer of CD40L was associated with up-regulation of the co-stimulatory molecules CD80 and CD86 and adhesion molecule CD54. In contrast, transgenic IL-2 secretion was greater following plasmid transduction. These phenotypic differences in the vaccines were associated with different functionality, both ex vivo and following administration to patients. Thus adenoviral vaccines induced greater activation of leukemia-reactive T cells ex vivo than plasmid vaccines. In treated patients, specific T-cell (T helper 1 (Th1) and T helper 2 (Th2)) and humoral anti-leukemia responses were detected following administration of the adenoviral vaccine (n = 15), while recipients of the plasmid vaccine (n = 9) manifested only a low-level Th2 response. Progression-free survival at 2 years was 46.7% in the adenoviral vaccine recipients, versus 11.1 % in those receiving plasmid vaccine. CONCLUSIONS: CLL vaccines expressing the same transgenes but produced by distinct methods of gene transfer may differ in the polarity of the immune response they induce in patients.


Asunto(s)
Ligando de CD40/metabolismo , Vacunas contra el Cáncer , Técnicas de Transferencia de Gen , Interleucina-2/metabolismo , Leucemia Linfocítica Crónica de Células B/terapia , Adenoviridae/genética , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Antígenos de Neoplasias/inmunología , Ligando de CD40/genética , Adhesión Celular , Diferenciación Celular , Femenino , Humanos , Interleucina-2/genética , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Plásmidos/genética , Trasplante Autólogo
20.
Mol Ther Methods Clin Dev ; 23: 296-306, 2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34729377

RESUMEN

The synovial sarcoma X breakpoint 2 (SSX2) belongs to a multigene family of cancer-testis antigens and can be found overexpressed in multiple malignancies. Its restricted expression in immune-privileged normal tissues suggest that SSX2 may be a relevant target antigen for chimeric antigen receptor (CAR) therapy. We have developed a T cell receptor (TCR)-like antibody (Fab/3) that binds SSX2 peptide 41-49 (KASEKIFYV) in the context of HLA-A∗-0201. The sequence of Fab/3 was utilized to engineer a CAR with the CD3 zeta intra-cellular domain along with either a CD28 or 4-1BB costimulatory endodomain. Human T cells from HLA-A2+ donors were transduced to mediate anti-tumor activity against acute myeloid leukemia (AML) tumor cells. Upon challenge with HLA-A2+/SSX2+ AML tumor cells, CAR-expressing T cells released interferon-γ and eliminated the tumor cells in a long-term co-culture assay. Using the HLA-A2+ T2 cell line, we demonstrated a strong specificity of the single-chain variable fragment (scFv) for SSX2 p41-49 and the closely related SSX3 p41-49, with no response against the others SSX-homologous peptides or unrelated homologous peptides. Since SSX3 has not been observed in tumor cells and expression cannot be induced by pharmacological intervention, SSX241-49 represents an attractive target for CAR-based cellular therapy to treat multiple types of cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA