Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 70(4): 1101-1113, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33123754

RESUMEN

Although immunotherapy has achieved impressive durable clinical responses, many cancers respond only temporarily or not at all to immunotherapy. To find novel, targetable mechanisms of resistance to immunotherapy, patient-derived melanoma cell lines were transduced with 576 open reading frames, or exposed to arrayed libraries of 850 bioactive compounds, prior to co-culture with autologous tumor-infiltrating lymphocytes (TILs). The synergy between the targets and TILs to induce apoptosis, and the mechanisms of inhibiting resistance to TILs were interrogated. Gene expression analyses were performed on tumor samples from patients undergoing immunotherapy for metastatic melanoma. Finally, the effect of inhibiting the top targets on the efficacy of immunotherapy was investigated in multiple preclinical models. Aurora kinase was identified as a mediator of melanoma cell resistance to T-cell-mediated cytotoxicity in both complementary screens. Aurora kinase inhibitors were validated to synergize with T-cell-mediated cytotoxicity in vitro. The Aurora kinase inhibition-mediated sensitivity to T-cell cytotoxicity was shown to be partially driven by p21-mediated induction of cellular senescence. The expression levels of Aurora kinase and related proteins were inversely correlated with immune infiltration, response to immunotherapy and survival in melanoma patients. Aurora kinase inhibition showed variable responses in combination with immunotherapy in vivo, suggesting its activity is modified by other factors in the tumor microenvironment. These data suggest that Aurora kinase inhibition enhances T-cell cytotoxicity in vitro and can potentiate antitumor immunity in vivo in some but not all settings. Further studies are required to determine the mechanism of primary resistance to this therapeutic intervention.


Asunto(s)
Aurora Quinasa A/metabolismo , Aurora Quinasa B/metabolismo , Resistencia a Antineoplásicos/inmunología , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Linfocitos T Citotóxicos/trasplante , Animales , Apoptosis , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/genética , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/genética , Proliferación Celular , Femenino , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/terapia , Ratones , Pronóstico , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Proc Natl Acad Sci U S A ; 113(48): E7788-E7797, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27849617

RESUMEN

Adoptive immunotherapy retargeting T cells to CD19 via a chimeric antigen receptor (CAR) is an investigational treatment capable of inducing complete tumor regression of B-cell malignancies when there is sustained survival of infused cells. T-memory stem cells (TSCM) retain superior potential for long-lived persistence, but challenges exist in manufacturing this T-cell subset because they are rare among circulating lymphocytes. We report a clinically relevant approach to generating CAR+ T cells with preserved TSCM potential using the Sleeping Beauty platform. Because IL-15 is fundamental to T-cell memory, we incorporated its costimulatory properties by coexpressing CAR with a membrane-bound chimeric IL-15 (mbIL15). The mbIL15-CAR T cells signaled through signal transducer and activator of transcription 5 to yield improved T-cell persistence independent of CAR signaling, without apparent autonomous growth or transformation, and achieved potent rejection of CD19+ leukemia. Long-lived T cells were CD45ROnegCCR7+CD95+, phenotypically most similar to TSCM, and possessed a memory-like transcriptional profile. Overall, these results demonstrate that CAR+ T cells can develop long-term persistence with a memory stem-cell phenotype sustained by signaling through mbIL15. This observation warrants evaluation in clinical trials.


Asunto(s)
Interleucina-15/metabolismo , Neoplasias Experimentales/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/fisiología , Animales , Antígenos CD19/metabolismo , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos , Ratones , Células Precursoras de Linfocitos T/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal
3.
Blood ; 127(24): 2980-90, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27118452

RESUMEN

Myeloablative autologous hematopoietic stem cell transplantation (HSCT) is a mainstay of therapy for relapsed intermediate-grade B-cell non-Hodgkin lymphoma (NHL); however, relapse rates are high. In phase 1 studies designed to improve long-term remission rates, we administered adoptive T-cell immunotherapy after HSCT, using ex vivo-expanded autologous central memory-enriched T cells (TCM) transduced with lentivirus expressing CD19-specific chimeric antigen receptors (CARs). We present results from 2 safety/feasibility studies, NHL1 and NHL2, investigating different T-cell populations and CAR constructs. Engineered TCM-derived CD19 CAR T cells were infused 2 days after HSCT at doses of 25 to 200 × 10(6) in a single infusion. In NHL1, 8 patients safely received T-cell products engineered from enriched CD8(+) TCM subsets, expressing a first-generation CD19 CAR containing only the CD3ζ endodomain (CD19R:ζ). Four of 8 patients (50%; 95% confidence interval [CI]: 16-84%) were progression free at both 1 and 2 years. In NHL2, 8 patients safely received T-cell products engineered from enriched CD4(+) and CD8(+) TCM subsets and expressing a second-generation CD19 CAR containing the CD28 and CD3ζ endodomains (CD19R:28ζ). Six of 8 patients (75%; 95% CI: 35-97%) were progression free at 1 year. The CD4(+)/CD8(+) TCM-derived CD19 CAR T cells (NHL2) exhibited improvement in expansion; however, persistence was ≤28 days, similar to that seen by others using CD28 CARs. Neither cytokine release syndrome nor delayed hematopoietic engraftment was observed in either trial. These data demonstrate the safety and feasibility of CD19 CAR TCM therapy after HSCT. Trials were registered at www.clinicaltrials.gov as #NCT01318317 and #NCT01815749.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Memoria Inmunológica , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/terapia , Linfocitos T/trasplante , Adulto , Anciano , Antígenos CD19/metabolismo , Recuento de Células , Terapia Combinada/efectos adversos , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Inmunoterapia Adoptiva/efectos adversos , Linfoma de Células B/inmunología , Linfoma no Hodgkin/inmunología , Linfoma no Hodgkin/terapia , Masculino , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Autólogo , Adulto Joven
4.
Immunol Rev ; 257(1): 181-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24329797

RESUMEN

The advent of efficient approaches to the genetic modification of T cells has provided investigators with clinically appealing methods to improve the potency of tumor-specific clinical grade T cells. For example, gene therapy has been successfully used to enforce expression of chimeric antigen receptors (CARs) that provide T cells with ability to directly recognize tumor-associated antigens without the need for presentation by human leukocyte antigen. Gene transfer of CARs can be undertaken using viral-based and non-viral approaches. We have advanced DNA vectors derived from the Sleeping Beauty (SB) system to avoid the expense and manufacturing difficulty associated with transducing T cells with recombinant viral vectors. After electroporation, the transposon/transposase improves the efficiency of integration of plasmids used to express CAR and other transgenes in T cells. The SB system combined with artificial antigen-presenting cells (aAPC) can selectively propagate and thus retrieve CAR(+) T cells suitable for human application. This review describes the translation of the SB system and aAPC for use in clinical trials and highlights how a nimble and cost-effective approach to developing genetically modified T cells can be used to implement clinical trials infusing next-generation T cells with improved therapeutic potential.


Asunto(s)
Antígenos CD19/inmunología , Terapia Genética , Vectores Genéticos/genética , Recombinación Homóloga , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Técnicas de Cultivo de Célula , Epítopos de Linfocito T/inmunología , Técnicas de Transferencia de Gen , Ingeniería Genética , Terapia Genética/métodos , Humanos , Inmunoterapia Adoptiva/métodos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
5.
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
6.
Pediatr Transplant ; 21(3)2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28160352

RESUMEN

We conducted a retrospective analysis of outcomes for children and young adults with sAML/sMDS who underwent HSCT at our institution. Thirty-two patients (median age 20 years) with sAML (n=24) and sMDS (n=8) received HSCT between 1990 and 2013. The median time from sAML/sMDS diagnosis to HSCT was 4.1 months (range: 1.2-27.2 months). The transplant regimens were primarily busulfan based (n=19). BM was the primary donor source (n=15). Eleven recipients were transplanted with residual disease. At a median follow-up of 62.3 months (range: 0.4-250.9 months), 14 patients had disease recurrence. Acute GVHD, grade III/IV, occurred in three patients. Causes of death were as follows: disease relapse (n=12), infection (n=2), pneumonia (n=1), pulmonary hemorrhage (n=1), acute GVHD (n=1), and graft failure (n=1). A PS of ≥90% at the time of HSCT had a significant impact on PFS (P=.02). Patients achieving pretransplant primary CR (n=8) and those with sMDS and RA (n=6) had prolonged PFS (P=.04). On multivariate analysis, shorter time to transplantation (≤6 months from diagnosis of sAML/sMDS) was associated with superior OS (P=.0018) and PFS (P=.0005).


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/terapia , Adolescente , Adulto , Busulfano/uso terapéutico , Niño , Femenino , Enfermedad Injerto contra Huésped , Humanos , Masculino , Análisis Multivariante , Recurrencia Local de Neoplasia , Recurrencia , Estudios Retrospectivos , Factores de Tiempo , Acondicionamiento Pretrasplante , Trasplante Homólogo , Resultado del Tratamiento , Adulto Joven
7.
J Pediatr Hematol Oncol ; 39(8): 609-613, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28859043

RESUMEN

INTRODUCTION: High rates of patients require readmission to the hospital within 6 months of hematopoietic stem cell transplantation (HSCT). We investigated the relationship between readmission rates and outcomes after HSCT in children, adolescents, and young adults (CAYA). MATERIALS AND METHODS: A retrospective analysis of patients (26 years or younger) treated with HSCT was conducted. RESULTS: A chart review of 435 CAYA who underwent HSCT from 2008 to 2015 revealed that 171 patients (39%) had at least 1 hospital readmission within 180 days of transplant; 87% received allogeneic and 13% received autologous HSCT. A total of 312 readmission events were reported. The median follow-up time was 31 months. Documented infection (n=99) and graft-versus-host disease complications (n=60) were the most common causes. Higher than 2 readmission rates were associated with lower overall survival (OS) (P=0.001) and disease-free survival (P<0.001) in patients who received allogeneic HSCT. These findings were not found in the autologous HSCT. In a multivariate analysis of those who received allogeneic HSCT, prior treatment with ≥2 chemotherapy regimens (P=0.03) was independent predictor of lower OS. There were also trends noted toward lower OS for patients with documented infections at index admission or subsequent readmissions (P=0.09). CONCLUSIONS: More than 2 hospital readmissions within 6 months of allogeneic HSCT in CAYA, who are either heavily pretreated or had documented infections at index admission or subsequent readmissions adversely affected the outcomes.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/estadística & datos numéricos , Readmisión del Paciente/estadística & datos numéricos , Adolescente , Adulto , Niño , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/epidemiología , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/mortalidad , Encuestas de Atención de la Salud , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Infecciones/epidemiología , Infecciones/etiología , Infecciones/mortalidad , Masculino , Estudios Retrospectivos , Análisis de Supervivencia , Texas/epidemiología , Factores de Tiempo , Trasplante Autólogo , Trasplante Homólogo , Adulto Joven
8.
Proc Natl Acad Sci U S A ; 111(29): 10660-5, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002471

RESUMEN

Clinical-grade T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) to redirect their specificity to target tumor-associated antigens in vivo. We now have developed this molecular strategy to render cytotoxic T cells specific for fungi. We adapted the pattern-recognition receptor Dectin-1 to activate T cells via chimeric CD28 and CD3-ζ (designated "D-CAR") upon binding with carbohydrate in the cell wall of Aspergillus germlings. T cells genetically modified with the Sleeping Beauty system to express D-CAR stably were propagated selectively on artificial activating and propagating cells using an approach similar to that approved by the Food and Drug Administration for manufacturing CD19-specific CAR(+) T cells for clinical trials. The D-CAR(+) T cells exhibited specificity for ß-glucan which led to damage and inhibition of hyphal growth of Aspergillus in vitro and in vivo. Treatment of D-CAR(+) T cells with steroids did not compromise antifungal activity significantly. These data support the targeting of carbohydrate antigens by CAR(+) T cells and provide a clinically appealing strategy to enhance immunity for opportunistic fungal infections using T-cell gene therapy.


Asunto(s)
Aspergilosis/inmunología , Aspergilosis/terapia , Bioingeniería/métodos , Carbohidratos/antagonistas & inhibidores , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/terapia , Linfocitos T/inmunología , Animales , Antígenos CD19/metabolismo , Aspergilosis/microbiología , Aspergilosis/patología , Aspergillus/efectos de los fármacos , Aspergillus/fisiología , Dexametasona/farmacología , Humanos , Hifa/efectos de los fármacos , Hifa/fisiología , Inmunofenotipificación , Lectinas Tipo C/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Infecciones Oportunistas/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos
9.
J Biol Chem ; 290(38): 22970-6, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26242737

RESUMEN

Methotrexate (MTX) is an anti-folate that inhibits de novo purine and thymidine nucleotide synthesis. MTX induces death in rapidly replicating cells and is used in the treatment of multiple cancers. MTX inhibits thymidine synthesis by targeting dihydrofolate reductase (DHFR) and thymidylate synthase (TYMS). The use of MTX to treat cancer also causes bone marrow suppression and inhibits the immune system. This has led to the development of an MTX-resistant DHFR, DHFR L22F, F31S (DHFR(FS)), to rescue healthy cells. 5-Fluorouracil-resistant TYMS T51S, G52S (TYMS(SS)) is resistant to MTX and improves MTX resistance of DHFR(FS) in primary T cells. Here we find that a known mechanism of MTX-induced increase in DHFR expression persists with DHFR(FS) and cis-expressed transgenes. We also find that TYMS(SS) expression of cis-expressed transgenes is similarly decreased in an MTX-inducible manner. MTX-inducible changes in DHFR(FS) and TYMS(SS) expression changes are lost when both genes are expressed together. In fact, expression of the DHFR(FS) and TYMS(SS) cis-expressed transgenes becomes correlated. These findings provide the basis for an unrecognized post-transcriptional mechanism that functionally links expression of DHFR and TYMS. These findings were made in genetically modified primary human T cells and have a clear potential for use in clinical applications where gene expression needs to be regulated by drug or maintained at a specific expression level. We demonstrate a potential application of this system in the controlled expression of systemically toxic cytokine IL-12.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Metotrexato/farmacología , Mutación Missense , Linfocitos T/enzimología , Tetrahidrofolato Deshidrogenasa , Timidilato Sintasa , Sustitución de Aminoácidos , Humanos , Células Jurkat , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Transgenes
10.
Bioinformatics ; 31(19): 3189-97, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26059718

RESUMEN

MOTIVATION: There is a need for effective automated methods for profiling dynamic cell-cell interactions with single-cell resolution from high-throughput time-lapse imaging data, especially, the interactions between immune effector cells and tumor cells in adoptive immunotherapy. RESULTS: Fluorescently labeled human T cells, natural killer cells (NK), and various target cells (NALM6, K562, EL4) were co-incubated on polydimethylsiloxane arrays of sub-nanoliter wells (nanowells), and imaged using multi-channel time-lapse microscopy. The proposed cell segmentation and tracking algorithms account for cell variability and exploit the nanowell confinement property to increase the yield of correctly analyzed nanowells from 45% (existing algorithms) to 98% for wells containing one effector and a single target, enabling automated quantification of cell locations, morphologies, movements, interactions, and deaths without the need for manual proofreading. Automated analysis of recordings from 12 different experiments demonstrated automated nanowell delineation accuracy >99%, automated cell segmentation accuracy >95%, and automated cell tracking accuracy of 90%, with default parameters, despite variations in illumination, staining, imaging noise, cell morphology, and cell clustering. An example analysis revealed that NK cells efficiently discriminate between live and dead targets by altering the duration of conjugation. The data also demonstrated that cytotoxic cells display higher motility than non-killers, both before and during contact. CONTACT: broysam@central.uh.edu or nvaradar@central.uh.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Comunicación Celular , Rastreo Celular/métodos , Células Asesinas Naturales/citología , Nanoestructuras/química , Linfocitos T/citología , Imagen de Lapso de Tiempo/métodos , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Procesamiento de Imagen Asistido por Computador , Células K562
11.
Biol Blood Marrow Transplant ; 21(10): 1714-20, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26172479

RESUMEN

Haploidentical transplantation can extend the opportunity for transplantation to almost all patients who lack an HLA-matched donor. Advances in the field of haploidentical transplantation have led to a marked decrease in treatment-related mortality, allowing investigators to focus on developing rationale pre- and peri-remission therapies aimed at preventing disease relapse after transplantation. Because of widespread availability, low treatment-related mortality, and cost, haploidentical donors may become the preferred "alternative" donors for allogeneic hematopoietic stem cell transplantation. One of the major advantages of using a related donor is the possibility of collecting or generating additional cellular products from the same immediately available donor, which will not be rejected. Infusion of these cells in the peri-transplantation period, derived from the same immune system, is opening the possibility of markedly enhancing the antitumor effects of the graft and hastening immunologic reconstitution after transplantation.


Asunto(s)
Antígenos HLA/inmunología , Haplotipos , Trasplante de Células Madre Hematopoyéticas/métodos , Histocompatibilidad , Inmunoterapia Adoptiva/métodos , Linfocitos T/trasplante , Ciclofosfamida/administración & dosificación , Ciclofosfamida/uso terapéutico , Genes Transgénicos Suicidas , Antígenos HLA/genética , Humanos , Células Asesinas Naturales/trasplante , Donadores Vivos/provisión & distribución , Depleción Linfocítica/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Receptores de Antígenos de Linfocitos T/uso terapéutico , Rodaminas/uso terapéutico , Inmunología del Trasplante
12.
N Engl J Med ; 367(24): 2305-15, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23234514

RESUMEN

BACKGROUND: Poor engraftment due to low cell doses restricts the usefulness of umbilical-cord-blood transplantation. We hypothesized that engraftment would be improved by transplanting cord blood that was expanded ex vivo with mesenchymal stromal cells. METHODS: We studied engraftment results in 31 adults with hematologic cancers who received transplants of 2 cord-blood units, 1 of which contained cord blood that was expanded ex vivo in cocultures with allogeneic mesenchymal stromal cells. The results in these patients were compared with those in 80 historical controls who received 2 units of unmanipulated cord blood. RESULTS: Coculture with mesenchymal stromal cells led to an expansion of total nucleated cells by a median factor of 12.2 and of CD34+ cells by a median factor of 30.1. With transplantation of 1 unit each of expanded and unmanipulated cord blood, patients received a median of 8.34×10(7) total nucleated cells per kilogram of body weight and 1.81×10(6) CD34+ cells per kilogram--doses higher than in our previous transplantations of 2 units of unmanipulated cord blood. In patients in whom engraftment occurred, the median time to neutrophil engraftment was 15 days in the recipients of expanded cord blood, as compared with 24 days in controls who received unmanipulated cord blood only (P<0.001); the median time to platelet engraftment was 42 days and 49 days, respectively (P=0.03). On day 26, the cumulative incidence of neutrophil engraftment was 88% with expansion versus 53% without expansion (P<0.001); on day 60, the cumulative incidence of platelet engraftment was 71% and 31%, respectively (P<0.001). CONCLUSIONS: Transplantation of cord-blood cells expanded with mesenchymal stromal cells appeared to be safe and effective. Expanded cord blood in combination with unmanipulated cord blood significantly improved engraftment, as compared with unmanipulated cord blood only. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT00498316.).


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Mesenquimatosas , Adolescente , Adulto , Recuento de Células Sanguíneas , Plaquetas , Causas de Muerte , Técnicas de Cultivo de Célula , Refuerzo Inmunológico de Injertos , Enfermedad Injerto contra Huésped , Neoplasias Hematológicas/mortalidad , Humanos , Células Madre Mesenquimatosas , Persona de Mediana Edad , Neutrófilos , Quimera por Trasplante , Trasplante Homólogo , Adulto Joven
13.
Mod Pathol ; 28(3): 373-82, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25216221

RESUMEN

Myelodysplastic syndromes are a heterogeneous group of clonal bone marrow hematopoietic stem cell disorders characterized by ineffective hematopoiesis and peripheral cytopenias. Chromosomal abnormalities and gene mutations have been shown to have essential roles in pathogenesis and correlate with prognosis. Molecular markers, however, are not integrated into currently used prognostic systems. The goal of this study is to identify plasma microRNAs useful for classification and risk stratification of myelodysplastic syndromes. We applied a novel, high-throughput digital quantification technology (NanoString) to profile microRNA expression in plasma samples of 72 patients with myelodysplastic syndromes and 12 healthy individuals. We correlated these results with overall survival. In patients with myelodysplastic syndromes associated with a diploid karyotype, we identified and validated a 7-microRNA signature as an independent predictor of survival with a predictive power of 75% accuracy (P=0.008), better than those of the International Prognostic Scoring Systems and the MD Anderson Prognostic Lower Risk Prognostic Model. We also identified differentially expressed plasma microRNAs in patients with myelodysplastic syndromes versus healthy individuals and between patients with myelodysplastic syndromes associated with different cytogenetic features. These results validate the utility of circulating-microRNA levels as noninvasive biomarkers that can inform the management of patients with myelodysplastic syndromes. Our findings also shed light on interactions of gene regulation pathways that are likely involved in the pathogenesis of myelodysplastic syndromes.


Asunto(s)
Biomarcadores/sangre , MicroARNs/sangre , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/genética , Adulto , Anciano , Anciano de 80 o más Años , Análisis Citogenético , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/mortalidad , Pronóstico
14.
Blood ; 122(8): 1341-9, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23741009

RESUMEN

Long-term engraftment of allogeneic cells necessitates eluding immune-mediated rejection, which is currently achieved by matching for human leukocyte antigen (HLA) expression, immunosuppression, and/or delivery of donor-derived cells to sanctuary sites. Genetic engineering provides an alternative approach to avoid clearance of cells that are recognized as "non-self" by the recipient. To this end, we developed designer zinc finger nucleases and employed a "hit-and-run" approach to genetic editing for selective elimination of HLA expression. Electro-transfer of mRNA species coding for these engineered nucleases completely disrupted expression of HLA-A on human T cells, including CD19-specific T cells. The HLA-A(neg) T-cell pools can be enriched and evade lysis by HLA-restricted cytotoxic T-cell clones. Recognition by natural killer cells of cells that had lost HLA expression was circumvented by enforced expression of nonclassical HLA molecules. Furthermore, we demonstrate that zinc finger nucleases can eliminate HLA-A expression from embryonic stem cells, which broadens the applicability of this strategy beyond infusing HLA-disparate immune cells. These findings establish that clinically appealing cell types derived from donors with disparate HLA expression can be genetically edited to evade an immune response and provide a foundation whereby cells from a single donor can be administered to multiple recipients.


Asunto(s)
Desoxirribonucleasas/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Trasplante de Células Madre/métodos , Trasplante Homólogo , Antígenos CD19/metabolismo , Secuencia de Bases , Diferenciación Celular , Citotoxicidad Inmunológica/inmunología , Electroporación , Células Madre Embrionarias/citología , Técnicas de Transferencia de Gen , Células HEK293 , Humanos , Leucocitos Mononucleares/citología , Datos de Secuencia Molecular , Ingeniería de Proteínas , Linfocitos T/inmunología , Dedos de Zinc
15.
Blood ; 119(22): 5164-72, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22498742

RESUMEN

Natural killer (NK) cells have gained significant attention in adoptive immunotherapy for cancer. Consequently, novel methods of clinical-grade expansion of NK cells have emerged. Subsets of NK cells express a variety of chemokine receptors. However, to expand the scope of adoptively transferred NK cell homing to various malignancies, expression of corresponding chemokine receptors on NK cells is essential. Here, we have explored the use of trogocytosis as a tool to transiently express the chemokine receptor CCR7 on expanded human NK cells with the aim to enhance their homing to lymph nodes. We generated a K562-based "donor" cell line expressing CCR7, Clone9.CCR7, to transfer CCR7 onto NK cells via trogocytosis. CCR7 expression occurred in 80% of expanded NK cells within 1 hour after coculture with Clone9.CCR7. After removal of the donor cells from the coculture, the CCR7 expression on NK cells steadily declined to baseline levels by 72 hours. The acquired CCR7 receptors mediated in vitro migration of NK cells toward CCL19 and CCL21 and increased the lymph node homing by 144% in athymic nude mice. This is the first report on exploiting trogocytosis to rapidly and transiently modify lymphocytes, without direct genetic intervention, for adoptive transfer.


Asunto(s)
Ingeniería Celular , Movimiento Celular , Expresión Génica , Células Asesinas Naturales/inmunología , Ganglios Linfáticos/inmunología , Receptores CCR7/inmunología , Traslado Adoptivo , Animales , Técnicas de Cocultivo , Femenino , Humanos , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/trasplante , Ganglios Linfáticos/citología , Masculino , Ratones , Ratones Desnudos , Receptores CCR7/genética , Transducción Genética , Trasplante Heterólogo
16.
Blood ; 119(24): 5697-705, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22535661

RESUMEN

Clinical-grade T cells are genetically modified ex vivo to express a chimeric antigen receptor (CAR) to redirect specificity to a tumor associated antigen (TAA) thereby conferring antitumor activity in vivo. T cells expressing a CD19-specific CAR recognize B-cell malignancies in multiple recipients independent of major histocompatibility complex (MHC) because the specificity domains are cloned from the variable chains of a CD19 monoclonal antibody. We now report a major step toward eliminating the need to generate patient-specific T cells by generating universal allogeneic TAA-specific T cells from one donor that might be administered to multiple recipients. This was achieved by genetically editing CD19-specific CAR(+) T cells to eliminate expression of the endogenous αß T-cell receptor (TCR) to prevent a graft-versus-host response without compromising CAR-dependent effector functions. Genetically modified T cells were generated using the Sleeping Beauty system to stably introduce the CD19-specific CAR with subsequent permanent deletion of α or ß TCR chains with designer zinc finger nucleases. We show that these engineered T cells display the expected property of having redirected specificity for CD19 without responding to TCR stimulation. CAR(+)TCR(neg) T cells of this type may potentially have efficacy as an off-the-shelf therapy for investigational treatment of B-lineage malignancies.


Asunto(s)
Antígenos CD19/inmunología , Epítopos/inmunología , Ingeniería Genética , Inmunoterapia/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes/inmunología , Linfocitos T/inmunología , Adulto , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/inmunología , Antígenos CD28/metabolismo , Complejo CD3/metabolismo , Células Cultivadas , Endonucleasas/metabolismo , Técnicas de Inactivación de Genes , Humanos , Células K562 , Activación de Linfocitos/inmunología , Dedos de Zinc
17.
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
18.
Mol Ther ; 21(3): 638-47, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23295945

RESUMEN

Even though other γδ T-cell subsets exhibit antitumor activity, adoptive transfer of γδ Tcells is currently limited to one subset (expressing Vγ9Vδ2 T-cell receptor (TCR)) due to dependence on aminobisphosphonates as the only clinically appealing reagent for propagating γδ T cells. Therefore, we developed an approach to propagate polyclonal γδ T cells and rendered them bispecific through expression of a CD19-specific chimeric antigen receptor (CAR). Peripheral blood mononuclear cells (PBMC) were electroporated with Sleeping Beauty (SB) transposon and transposase to enforce expression of CAR in multiple γδ T-cell subsets. CAR(+)γδ T cells were expanded on CD19(+) artificial antigen-presenting cells (aAPC), which resulted in >10(9) CAR(+)γδ T cells from <10(6) total cells. Digital multiplex assay detected TCR mRNA coding for Vδ1, Vδ2, and Vδ3 with Vγ2, Vγ7, Vγ8, Vγ9, and Vγ10 alleles. Polyclonal CAR(+)γδ T cells were functional when TCRγδ and CAR were stimulated and displayed enhanced killing of CD19(+) tumor cell lines compared with CAR(neg)γδ T cells. CD19(+) leukemia xenografts in mice were reduced with CAR(+)γδ T cells compared with control mice. Since CAR, SB, and aAPC have been adapted for human application, clinical trials can now focus on the therapeutic potential of polyclonal γδ T cells.


Asunto(s)
Antígenos CD19/inmunología , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD19/metabolismo , Línea Celular Tumoral , Electroporación , Humanos , Leucemia/terapia , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Transposasas/genética , Transposasas/metabolismo
19.
Chin J Cancer ; 33(9): 421-33, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25189715

RESUMEN

The adoptive transfer of T cells is a promising approach to treat cancers. Primary human T cells can be modified using viral and non-viral vectors to promote the specific targeting of cancer cells via the introduction of exogenous T-cell receptors (TCRs) or chimeric antigen receptors (CARs). This gene transfer displays the potential to increase the specificity and potency of the anticancer response while decreasing the systemic adverse effects that arise from conventional treatments that target both cancerous and healthy cells. This review highlights the generation of clinical-grade T cells expressing CARs for immunotherapy, the use of these cells to target B-cell malignancies and, particularly, the first clinical trials deploying the Sleeping Beauty gene transfer system, which engineers T cells to target CD19+ leukemia and non-Hodgkin's lymphoma.


Asunto(s)
Terapia Genética , Inmunoterapia Adoptiva , Linfoma de Células B/terapia , Linfoma no Hodgkin/terapia , Linfocitos T , Linfocitos B , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Leucemia , Neoplasias , Receptores de Antígenos , Receptores de Antígenos de Linfocitos T , Receptores CCR1 , Especificidad del Receptor de Antígeno de Linfocitos T
20.
Nat Cancer ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750245

RESUMEN

Chimeric antigen receptor (CAR) T cells used for the treatment of B cell malignancies can identify T cell subsets with superior clinical activity. Here, using infusion products of individuals with large B cell lymphoma, we integrated functional profiling using timelapse imaging microscopy in nanowell grids with subcellular profiling and single-cell RNA sequencing to identify a signature of multifunctional CD8+ T cells (CD8-fit T cells). CD8-fit T cells are capable of migration and serial killing and harbor balanced mitochondrial and lysosomal volumes. Using independent datasets, we validate that CD8-fit T cells (1) are present premanufacture and are associated with clinical responses in individuals treated with axicabtagene ciloleucel, (2) longitudinally persist in individuals after treatment with CAR T cells and (3) are tumor migrating cytolytic cells capable of intratumoral expansion in solid tumors. Our results demonstrate the power of multimodal integration of single-cell functional assessments for the discovery and application of CD8-fit T cells as a T cell subset with optimal fitness in cell therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA