Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EBioMedicine ; 39: 173-181, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30579863

RESUMO

BACKGROUND: Adoptive immunotherapy using T cells expressing chimeric antigen receptors (CARs) targeting CD19 has produced remarkable clinical outcomes. However, much of the mechanisms of action, such as the development of memory responses and sources of immune cytokines, remain elusive largely due to the challenge of characterizing human CAR T cell function in vivo. The lack of a suitable in vivo model also hinders the development of new CAR T cell therapies. METHODS: We established a humanized mouse (hu-mouse) model with a functional human immune system and genetically-matched (autologous) primary acute B-lymphoblastic leukemia (B-ALL) that permits modeling of CD19-targeted CAR T cell therapy in immunocompetent hosts without allogeneic or xenogeneic immune responses. FINDINGS: Anti-CD19 CAR T cells were detected in blood of leukemic hu-mice with kinetics and levels similar to those seen in patients receiving CAR T cell therapy. The levels of CAR T cells were correlated inversely with the burden of leukemia cells and positively with the survival times in anti-CD19 CAR T cell-treated leukemic hu-mice. Infusion of anti-CD19 CAR T cells also resulted in rapid production of T cell- and monocyte/macrophage-derived cytokines and an increase in frequency of regulatory T cells as reported in clinical studies. INTERPRETATION: These results provide a proof-of-principle that this novel preclinical model has the potential to be used to model human CAR T cell therapy and facilitate the design of new CARs with improved antitumor activity.


Assuntos
Antígenos CD19/imunologia , Leucemia de Células B/terapia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/transplante , Animais , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Imunoterapia Adotiva , Leucemia de Células B/imunologia , Camundongos , Linfócitos T/imunologia , Resultado do Tratamento , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Clin Cancer Res ; 22(14): 3440-50, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26847056

RESUMO

PURPOSE: The effectiveness of NK cell infusions to induce leukemic remission is limited by lack of both antigen specificity and in vivo expansion. To address the first issue, we previously generated a bispecific killer engager (BiKE) containing single-chain scFv against CD16 and CD33 to create an immunologic synapse between NK cells and CD33(+) myeloid targets. We have now incorporated a novel modified human IL15 crosslinker, producing a 161533 trispecific killer engager (TriKE) to induce expansion, priming, and survival, which we hypothesize will enhance clinical efficacy. EXPERIMENTAL DESIGN: Reagents were tested in proliferation and functional assays and in an in vivo xenograft model of AML. RESULTS: When compared with the 1633 BiKE, the 161533 TriKE induced superior NK cell cytotoxicity, degranulation, and cytokine production against CD33(+) HL-60 targets and increased NK survival and proliferation. Specificity was shown by the ability of a 1615EpCAM TriKE to kill CD33-EpCAM(+) targets. Using NK cells from patients after allogeneic stem cell transplantation when NK cell function is defective, the 161533 TriKE restored potent NK function against primary AML targets and induced specific NK cell proliferation. These results were confirmed in an immunodeficient mouse HL-60-Luc tumor model where the 161533 TriKE exhibited superior antitumor activity and induced in vivo persistence and survival of human NK cells for at least 3 weeks. CONCLUSIONS: Off-the-shelf 161533 TriKE imparts antigen specificity and promotes in vivo persistence, activation, and survival of NK cells. These qualities are ideal for NK cell therapy of myeloid malignancies or targeting antigens of solid tumors. Clin Cancer Res; 22(14); 3440-50. ©2016 AACRSee related commentary by Talmadge, p. 3419.


Assuntos
Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Animais , Proliferação de Células/fisiologia , Citotoxicidade Imunológica/imunologia , Molécula de Adesão da Célula Epitelial/imunologia , Células HL-60 , Células HT29 , Humanos , Ativação Linfocitária/imunologia , Camundongos , Células-Tronco/imunologia , Transplante Homólogo/métodos
3.
PLoS One ; 10(7): e0133152, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26173023

RESUMO

Patients with metastatic or recurrent and refractory sarcomas have a dismal prognosis. Therefore, new targeted therapies are urgently needed. This study was designed to evaluate chimeric antigen receptor (CAR) T cells targeting the type I insulin-like growth factor receptor (IGF1R) or tyrosine kinase-like orphan receptor 1 (ROR1) molecules for their therapeutic potential against sarcomas. Here, we report that IGF1R (15/15) and ROR1 (11/15) were highly expressed in sarcoma cell lines including Ewing sarcoma, osteosarcoma, alveolar or embryonal rhabdomyosarcoma, and fibrosarcoma. IGF1R and ROR1 CAR T cells derived from eight healthy donors using the Sleeping Beauty (SB) transposon system were cytotoxic against sarcoma cells and produced high levels of IFN-γ, TNF-α and IL-13 in an antigen-specific manner. IGF1R and ROR1 CAR T cells generated from three sarcoma patients released significant amounts of IFN-γ in response to sarcoma stimulation. The adoptive transfer of IGF1R and ROR1 CAR T cells derived from a sarcoma patient significantly reduced tumor growth in pre-established, systemically disseminated and localized osteosarcoma xenograft models in NSG mice. Infusion of IGF1R and ROR1 CAR T cells also prolonged animal survival in a localized sarcoma model using NOD/scid mice. Our data indicate that both IGF1R and ROR1 can be effectively targeted by SB modified CAR T cells and that such CAR T cells may be useful in the treatment of high risk sarcoma patients.


Assuntos
Neoplasias Ósseas/imunologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Somatomedina/metabolismo , Sarcoma/imunologia , Linfócitos T/imunologia , Transferência Adotiva/métodos , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/terapia , Linhagem Celular Tumoral , Elementos de DNA Transponíveis/genética , Humanos , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-13/imunologia , Interleucina-13/metabolismo , Células K562 , Células MCF-7 , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/imunologia , Receptor IGF Tipo 1 , Receptores de Antígenos de Linfócitos T/imunologia , Sarcoma/metabolismo , Sarcoma/terapia , Linfócitos T/metabolismo
4.
J Biol Chem ; 290(5): 2831-41, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25505246

RESUMO

We have previously reported 27 differentially expressed microRNAs (miRNAs) during human monocyte differentiation into immature dendritic cells (imDCs) and mature DCs (mDCs). However, their roles in DC differentiation and function remain largely elusive. Here, we report that microRNA (miR)-146a and miR-146b modulate DC apoptosis and cytokine production. Expression of miR-146a and miR-146b was significantly increased upon monocyte differentiation into imDCs and mDCs. Silencing of miR-146a and/or miR-146b in imDCs and mDCs significantly prevented DC apoptosis, whereas overexpressing miR-146a and/or miR-146b increased DC apoptosis. miR-146a and miR-146b expression in imDCs and mDCs was inversely correlated with TRAF6 and IRAK1 expression. Furthermore, siRNA silencing of TRAF6 and/or IRAK1 in imDCs and mDCs enhanced DC apoptosis. By contrast, lentivirus overexpression of TRAF6 and/or IRAK1 promoted DC survival. Moreover, silencing of miR-146a and miR-146b expression had little effect on DC maturation but enhanced IL-12p70, IL-6, and TNF-α production as well as IFN-γ production by IL-12p70-mediated activation of natural killer cells, whereas miR-146a and miR-146b overexpression in mDCs reduced cytokine production. Silencing of miR-146a and miR-146b in DCs also down-regulated NF-κB inhibitor IκBα and increased Bcl-2 expression. Our results identify a new negative feedback mechanism involving the miR-146a/b-TRAF6/IRAK1-NF-κB axis in promoting DC apoptosis.


Assuntos
Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , MicroRNAs/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , MicroRNAs/genética , Monócitos/citologia , Reação em Cadeia da Polimerase em Tempo Real , Fator 6 Associado a Receptor de TNF/genética
5.
Gastroenterology ; 146(1): 188-99, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24036151

RESUMO

BACKGROUND & AIMS: Variants in genes that regulate autophagy have been associated with Crohn's disease (CD). Defects in autophagy-mediated removal of pathogenic microbes could contribute to the pathogenesis of CD. We investigated the role of the microRNAs (miRs) MIR106B and MIR93 in induction of autophagy and bacterial clearance in human cell lines and the correlation between MIR106B and autophagy-related gene 16L1 (ATG16L1) expression in tissues from patients with CD. METHODS: We studied the ability of MIR106B and MIR93 to regulate ATG transcripts in human cancer cell lines (HCT116, SW480, HeLa, and U2OS) using luciferase report assays and bioinformatics analyses; MIR106B and MIR93 mimics and antagonists were transfected into cells to modify levels of miRs. Cells were infected with LF82, a CD-associated adherent-invasive strain of Escherichia coli, and monitored by confocal microscopy and for colony-forming units. Colon tissues from 41 healthy subjects (controls), 22 patients with active CD, 16 patients with inactive CD, and 7 patients with chronic inflammation were assessed for levels of MIR106B and ATG16L1 by in situ hybridization and immunohistochemistry. RESULTS: Silencing Dicer1, an essential processor of miRs, increased levels of ATG protein and formation of autophagosomes in cells, indicating that miRs regulate autophagy. Luciferase reporter assays indicated that MIR106B and MIR93 targeted ATG16L1 messenger RNA. MIR106B and MIR93 reduced levels of ATG16L1 and autophagy; these increased after expression of ectopic ATG16L1. In contrast, MIR106B and MIR93 antagonists increased formation of autophagosomes. Levels of MIR106B were increased in intestinal epithelia from patients with active CD, whereas levels of ATG16L1 were reduced compared with controls. Levels of c-Myc were also increased in intestinal epithelia of patients with active CD compared with controls. These alterations could impair removal of CD-associated bacteria by autophagy. CONCLUSIONS: In human cell lines, MIR106B and MIR93 reduce levels of ATG16L1 and autophagy and prevent autophagy-dependent eradication of intracellular bacteria. This process also appears to be altered in colon tissues from patients with active CD.


Assuntos
Autofagia/imunologia , Proteínas de Transporte/imunologia , Doença de Crohn/imunologia , Células Epiteliais/imunologia , Escherichia coli , MicroRNAs/imunologia , Autofagia/genética , Proteínas Relacionadas à Autofagia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Doença de Crohn/genética , RNA Helicases DEAD-box/imunologia , Células HCT116 , Células HeLa , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-myc/imunologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ribonuclease III/imunologia
6.
PLoS One ; 7(12): e51203, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251452

RESUMO

Genetic aberrations contribute to acute myeloid leukemia (AML). However, half of AML cases do not contain the well-known aberrations detectable mostly by cytogenetic analysis, and these cases are classified as normal karyotype AML. Different outcomes of normal karyotype AML suggest that this subgroup of AML could be genetically heterogeneous. But lack of genetic markers makes it difficult to further study this subgroup of AML. Using paired-end RNAseq method, we performed a transcriptome analysis in 45 AML cases including 29 normal karyotype AML, 8 abnormal karyotype AML and 8 AML without karyotype informaiton. Our study identified 134 fusion transcripts, all of which were formed between the partner genes adjacent in the same chromosome and distributed at different frequencies in the AML cases. Seven fusions are exclusively present in normal karyotype AML, and the rest fusions are shared between the normal karyotype AML and abnormal karyotype AML. CIITA, a master regulator of MHC class II gene expression and truncated in B-cell lymphoma and Hodgkin disease, is found to fuse with DEXI in 48% of normal karyotype AML cases. The fusion transcripts formed between adjacent genes highlight the possibility that certain such fusions could be involved in oncological process in AML, and provide a new source to identify genetic markers for normal karyotype AML.


Assuntos
Fusão Gênica , Cariotipagem , Leucemia Mieloide Aguda/genética , RNA Mensageiro/genética , Sequência de Aminoácidos , Linhagem Celular Tumoral , Humanos , Dados de Sequência Molecular
7.
J Gene Med ; 13(9): 487-96, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21850667

RESUMO

BACKGROUND: Epstein-Barr virus (EBV) causes a range of life-threatening B-lymphocyte malignancies but, despite the use of various strategies, treatment remains problematic. METHODS: In the present study, we developed a non-integrating lentiviral vector (NILV) that mediates specific killing of EBV nuclear antigen 1 (EBNA1)-expressing cells with minimal toxicity to EBNA1-negative cells. The EBV family of repeats (FR) was cloned intok the NILV genome upstream of various transgenes. RESULTS: The presence of the FR in the NILV genome induced transcriptional up-regulation and prolonged the expression of a transgene specifically in EBNA1-positive B cells. Transgene expression from an FR-containing NILV was also prolonged in EBV-transformed cells compared to an FR-negative NILV. We found that the delivery of an FR-containing NILV encoding herpes simplex virus 1 thymidine kinase (TK) lead to the killing of more than 99% of EBNA1-positive B cells with minimal toxicity to EBNA1-negative cells in the presence of gancyclovir. EBNA1-positive cells were not killed by an FR-negative vector containing the TK gene. An FR-TK-containing NILV also specifically killed EBNA1-containing cells in a mixed population of EBNA1-positive and EBNA1-negative cells, thus confirming that NILV-FR-TK-mediated killing is specific for EBNA1-expressing cells. CONCLUSIONS: Transgene expression from our NILVs is both EBNA1-specific and dependent upon the presence of the FR. The results obtained in the present study indicate that NILVs have potential use in the treatment of EBV-associated B cell malignancies.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Genes Transgênicos Suicidas , Vetores Genéticos/genética , HIV-1/genética , Linfoma de Células B/terapia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Ganciclovir/farmacologia , Regulação da Expressão Gênica , Ordem dos Genes , Técnicas de Transferência de Genes , Terapia Genética , Células HEK293 , Humanos , Linfoma de Células B/virologia , Timidina Quinase/genética , Transcrição Gênica , Transdução Genética , Regulação para Cima/genética
8.
Blood ; 117(16): 4293-303, 2011 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-21355095

RESUMO

Dendritic cells (DCs) are potent antigen-presenting cells derived from hematopoietic progenitor cells and circulating monocytes. To investigate the role of microRNAs (miRNAs) during DC differentiation, maturation, and function, we profiled miRNA expression in human monocytes, immature DCs (imDCs), and mature DCs (mDCs). Stage-specific, differential expression of 27 miRNAs was found during monocyte differentiation into imDCs and mDCs. Among them, decreased miR-221 and increased miR-155 expression correlated with p27(kip1) accumulation in DCs. Silencing of miR-221 or overexpressing of miR-155 in DCs resulted in p27(kip1) protein increase and DC apoptosis. Moreover, mDCs from miR-155(-/-) mice were less apoptotic than those from wild-type mice. Silencing of miR-155 expression had little effect on DC maturation but reduced IL-12p70 production, whereas miR-155 overexpression in mDCs enhanced IL-12p70 production. Kip1 ubiquitination-promoting complex 1, suppressor of cytokine signaling 1, and CD115 (M-CSFR) were functional targets of miR-155. Furthermore, we provide evidence that miR-155 indirectly regulated p27(kip1) protein level by targeting Kip1 ubiquitination-promoting complex 1. Thus, our study uncovered miRNA signatures during monocyte differentiation into DCs and the new regulatory role of miR-221 and miR-155 in DC apoptosis and IL-12p70 production.


Assuntos
Apoptose , Inibidor de Quinase Dependente de Ciclina p27/imunologia , Células Dendríticas/citologia , Interleucina-12/imunologia , MicroRNAs/imunologia , Proteínas Supressoras da Sinalização de Citocina/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Expressão Gênica , Humanos , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Proteína 1 Supressora da Sinalização de Citocina
9.
Nat Cell Biol ; 12(11): 1108-14, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20935635

RESUMO

The Polycomb group (PcG) protein, enhancer of zeste homologue 2 (EZH2), has an essential role in promoting histone H3 lysine 27 trimethylation (H3K27me3) and epigenetic gene silencing. This function of EZH2 is important for cell proliferation and inhibition of cell differentiation, and is implicated in cancer progression. Here, we demonstrate that under physiological conditions, cyclin-dependent kinase 1 (CDK1) and cyclin-dependent kinase 2 (CDK2) phosphorylate EZH2 at Thr 350 in an evolutionarily conserved motif. Phosphorylation of Thr 350 is important for recruitment of EZH2 and maintenance of H3K27me3 levels at EZH2-target loci. Blockage of Thr 350 phosphorylation not only diminishes the global effect of EZH2 on gene silencing, it also mitigates EZH2-mediated cell proliferation and migration. These results demonstrate that CDK-mediated phosphorylation is a key mechanism governing EZH2 function and that there is a link between the cell-cycle machinery and epigenetic gene silencing.


Assuntos
Proteína Quinase CDC2/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética/genética , Fatores de Transcrição/metabolismo , Proteína Quinase CDC2/genética , Quinase 2 Dependente de Ciclina/genética , Proteínas de Ligação a DNA/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Inativação Gênica , Células HEK293 , Humanos , Fosforilação , Complexo Repressor Polycomb 2 , Fatores de Transcrição/genética , Células Tumorais Cultivadas
10.
Mol Ther ; 18(10): 1803-13, 2010 10.
Artigo em Inglês | MEDLINE | ID: mdl-20606646

RESUMO

In this study, we compared the genomic integration efficiencies and transposition site preferences of Sleeping Beauty (SB or SB11), Tol2, and piggyBac (PB) transposon systems in primary T cells derived from peripheral blood lymphocytes (PBL) and umbilical cord blood (UCB). We found that PB demonstrated the highest efficiency of stable gene transfer in PBL-derived T cells, whereas SB11 and Tol2 mediated intermediate and lowest efficiencies, respectively. Southern hybridization analysis demonstrated that PB generated the highest number of integrants when compared to SB and Tol2 in both PBL and UCB T cells. Tol2 and PB appeared more likely to promote clonal expansion than SB, which may be in part due to the dysregulated expression of cancer-related genes near the insertion sites. Genome-wide integration analysis demonstrated that SB, Tol2, and PB integrations occurred in all the chromosomes without preference. Additionally, Tol2 and PB integration sites were mainly localized near transcriptional start sites (TSSs), CpG islands and DNaseI hypersensitive sites, whereas SB integrations were randomly distributed. These results suggest that SB may be a preferential choice of the delivery vector in T cells due to its random integration site preference and relatively high efficiency, and support continuing development of SB-mediated T-cell phase I trials.


Assuntos
Elementos de DNA Transponíveis/genética , Técnicas de Transferência de Genes , Linfócitos T/metabolismo , Células Cultivadas , Biologia Computacional , Humanos , Reação em Cadeia da Polimerase , Transposases/genética , Transposases/metabolismo
11.
Hum Gene Ther ; 21(11): 1577-90, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20528476

RESUMO

We have shown that the Sleeping Beauty (SB) transposon system can mediate stable expression of both reporter and therapeutic genes in human primary T cells and that trans delivery (i.e., transposon and transposase are on separate plasmids) is at least 3-fold more efficient than cis delivery. One concern about trans delivery is the potential for integration of the transposase-encoding sequence into the cell genome with the possibility of continued expression, transposon remobilization, and insertional mutagenesis. To address this concern, human peripheral blood lymphocytes were nucleofected with transposase plasmid and a DsRed transposon. Eighty-eight stable DsRed(+) T cell clones were generated and found to be negative for the transposase-encoding sequence by PCR analysis of genomic DNA. Genomic PCR was positive for transposase in 5 of 15 bulk T cell populations that were similarly transfected and selected for transgene expression where copy numbers were unexpectedly high (0.007-0.047 per cell) by quantitative PCR. Transposase-positive bulk T cells lacked transposase plasmid demonstrated by Hirt (episomal) extracted DNA and showed no detectable transposase by Southern hybridization, Western blot, and quantitative RT-PCR analyses. Cytogenetic and array comparative genomic hybridization analyses of the only identified transposase-positive clone (O56; 0.867 copies per cell) showed no chromosomal abnormality or tumor formation in nude mice although transposon remobilization was detected. Our data suggest that SB delivery via plasmid in T cells should be carried out with caution because of unexpectedly high copy numbers of randomly integrated SB transposase.


Assuntos
Dosagem de Genes , Linfócitos T/metabolismo , Transgenes , Transposases/genética , Transposases/metabolismo , Animais , Feminino , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutagênese Insercional , Plasmídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
12.
Hum Gene Ther ; 21(1): 75-86, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19719389

RESUMO

Umbilical cord blood (UCB) T cells can be redirected to kill leukemia and lymphoma cells by engineering with a single-chain chimeric antigen receptor (CAR) and thus may have general applications in adoptive cell therapy. However, the role of costimulatory molecules in UCB T-cell activation and effector functions in context with CAR remains elusive. To investigate the effect of costimulatory molecules (4-1BB and CD28) on UCB T cells, we transduced UCB T cells with lentiviral vectors expressing Green Fluorescent Protein (GFP) and CAR for CD19 containing an intracellular domain of the CD3zeta chain and either a 4-1BB (UCB-19BBzeta) or a CD28 intracellular domain (UCB-1928zeta), both (UCB-1928BBzeta), or neither (UCB-19zeta). We found that UCB-19BBzeta and UCB-28BBzeta T cells exhibited more cytotoxicity to CD19(+) leukemia and lymphoma cell lines than UCB-19zeta and UCB-1928zeta, although differences in secretion of interleukin-2 and interferon-gamma by these T cells were not evident. In vivo adoptive transfer of these T cells into intraperitoneal tumor-bearing mice demonstrated that UCB-19BBzeta and UCB-1928BBzeta T cells mounted the most potent antitumor response. The mice adoptively transferred with UCB-1928BBzeta cells survived longer than the mice with UCB-19BBzeta. Moreover, UCB-1928BBzeta T cells mounted a more robust antitumor response than UCB-19BBzeta in a systemic tumor model. Our data suggest a synergistic role of 4-1BB and CD28 costimulation in engineering antileukemia UCB effector cells and implicate a design for redirected UCB T-cell therapy for refractory leukemia.


Assuntos
Linfócitos B/patologia , Antígenos CD28/metabolismo , Sangue Fetal/citologia , Leucemia/terapia , Transdução de Sinais , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Animais , Antígenos CD19/metabolismo , Linfócitos B/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunofenotipagem , Interferon gama/biossíntese , Interleucina-2/biossíntese , Lentivirus/genética , Leucemia/imunologia , Leucemia/patologia , Linfoma/imunologia , Linfoma/patologia , Camundongos , Camundongos SCID , Proteínas Recombinantes/metabolismo , Linfócitos T/citologia , Transdução Genética
13.
Blood ; 114(7): 1319-30, 2009 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-19414858

RESUMO

Here we report stable gene transfer in cord blood-derived CD34(+) hematopoietic stem cells using a hyperactive nonviral Sleeping Beauty (SB) transposase (SB100X). In colony-forming assays, SB100X mediated the highest efficiency (24%) of stable Discosoma sp red fluorescent protein (DsRed) reporter gene transfer in committed hematopoietic progenitors compared with both the early-generation hyperactive SB11 transposase and the piggyBac transposon system (1.23% and 3.8%, respectively). In vitro differentiation assays further demonstrated that SB100X-transfected CD34(+) cells can develop into DsRed(+) CD4(+)CD8(+) T (3.17%-21.84%; median, 7.97%), CD19(+) B (3.83%-18.66%; median, 7.84%), CD56(+)CD3(-) NK (3.53%-79.98%; median, 7.88%), and CD33(+) myeloid (7.59%-15.63%; median, 9.48%) cells. SB100X-transfected CD34(+) cells achieved approximately 46% engraftment in NOD-scid IL2gammac(null) (NOG) mice. Twelve weeks after transplantation, 0.57% to 28.96% (median, 2.79%) and 0.49% to 34.50% (median, 5.59%) of total human CD45(+) cells in the bone marrow and spleen expressed DsRed, including CD19(+) B, CD14(+) monocytoid, and CD33(+) myeloid cell lineages. Integration site analysis revealed SB transposon sequences in the human chromosomes of in vitro differentiated T, B, NK, and myeloid cells, as well as in human CD45(+) cells isolated from bone marrow and spleen of transplanted NOG mice. Our results support the continuing development of SB-based gene transfer into human hematopoietic stem cells as a modality for gene therapy.


Assuntos
Antígenos CD34 , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Elementos de DNA Transponíveis , Sangue Fetal , Técnicas de Transferência de Genes , Terapia Genética/métodos , Células-Tronco Hematopoéticas , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Diferenciação Celular/genética , Feminino , Expressão Gênica , Sobrevivência de Enxerto/genética , Humanos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fatores de Tempo , Transplante Heterólogo , Proteína Vermelha Fluorescente
14.
Methods Mol Biol ; 506: 115-26, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19110623

RESUMO

Genetic modification of peripheral blood T lymphocytes (PBL) or hematopoietic stem cells (HSC) has been shown to be promising in the treatment of cancer (Nat Rev Cancer 3:35-45, 2003), transplant complications (Curr Opin Hematol 5:478-482, 1998), viral infections (Science 285:546-551, 1999), and immunodeficiencies (Nat Rev Immunol 2:615-621, 2002). There are also significant implications for the study of T cell biology (J Exp Med 191:2031-2037, 2000). Currently, there are three types of vectors that are commonly used for introducing genes into human primary T cells: oncoretroviral vectors, lentiviral vectors, and naked DNA. Oncoretroviral vectors transduce and integrate only in dividing cells. However, it has been shown that extended ex vivo culture, required by oncoretroviral-mediated gene transfer, may alter the biologic properties of T cells (Nat Med 4:775-780, 1998; Int Immunol 9:1073- 1083, 1997; Hum Gene Ther 11:1151-1164, 2001; Blood 15:1165-1173, 2002; Proc Natl Acad Sci U S A, 1994). HIV-1-derived lentiviral vectors have been shown to transduce a variety of slowly dividing or nondividing cells, including unstimulated T lymphocytes (Blood 96:1309-1316, 2000; Gene Ther 7:596-604, 2000; Blood 101:2167-2174, 2002; Hum Gene Ther 14:1089-1105, 2003). However, achieving effective gene transfer and expression using lentivirus vectors can be complex, and there is at least a perceived risk associated with clinical application of a vector based on a human pathogen (i.e., HIV-1). Recently it has been found that oncoretroviral and lentiviral vectors show a preference for integration into regulatory sequences and active genes, respectively (Cell 110:521-529, 2002; Science 300:1749-1751, 2003). Additionally, insertional mutagenesis has become a serious concern, after several patients treated with an oncoretroviral vector for X-linked SCID developed a leukemia-like syndrome associated with activation of the LMO2 oncogene (Science 302:415-419, 2003). Naked DNA-based genetic engineering of human T lymphocytes also requires T cells to be activated prior to gene transfer (Mol Ther 1:49-55, 2000; Blood 101:1637-1644, 2003; Blood 107:2643-2652, 2006). In addition, random integration by electroporation is of low efficiency. We have recently reported that the Sleeping Beauty transposon system can efficiently mediate stable transgene expression in human primary T cells without prior T cell activation (Blood 107:483-491, 2006). This chapter describes methodology for the introduction of SB transposons into human T cell cultures with subsequent integration and stable long-term expression at noticeably high efficiency for a nonviral gene transfer system.


Assuntos
Elementos de DNA Transponíveis , DNA/metabolismo , Linfócitos T/metabolismo , Citometria de Fluxo , Técnicas de Transferência de Genes , Humanos , Plasmídeos
15.
Mol Ther ; 16(3): 580-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18227839

RESUMO

We have reported earlier that the non-viral Sleeping Beauty (SB) transposon system can mediate genomic integration and long-term reporter gene expression in human primary peripheral blood (PB) T cells. In order to test whether this system can be used for genetically modifying both PB T cells and umbilical cord blood (UCB) T cells as graft-versus-leukemia effector cells, an SB transposon was constructed to coexpress a single-chain chimeric antigen receptor (CAR) for human CD19 and CD20. PB and UCB were nucleofected with the transposon and a transposase plasmid, activated and then expanded in culture using anti-CD3/CD28 beads. Stable dual-gene expression was confirmed in both T-cell types, permitting enrichment by positive selection with Rituxan. The engineered CD4(+) T cells and CD8(+) T cells both exhibited specific cytotoxicity against CD19(+) leukemia and lymphoma cell lines, as well as against CD19 transfectants, and produced high-levels of antigen-dependent Th1 (but not Th2) cytokines. The in vivo adoptive transfer of genetically engineered T cells significantly reduced tumor growth and prolonged the survival of the animal. Taken together, these data indicate that T cells from PB and UCB can be stably modified using a non-viral DNA transfer system, and that such modified T cells may be useful in the treatment of refractory leukemia and lymphoma.


Assuntos
Antígenos CD19/genética , Leucemia/terapia , Linfoma/terapia , Retroelementos/genética , Linfócitos T/metabolismo , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos , Antígenos CD20/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Imunoterapia Adotiva , Leucemia/genética , Leucemia/patologia , Linfoma/genética , Linfoma/patologia , Modelos Genéticos , Plasmídeos/genética , Reação em Cadeia da Polimerase , Rituximab , Linfócitos T/citologia , Transfecção/métodos
16.
Cancer Res ; 66(3): 1730-9, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16452233

RESUMO

The ability of cancers to evade immune surveillance and resist immunotherapy raises a fundamental question of how tumor cells survive in the presence of a competent immune system. Studies to address this question have primarily focused on mechanisms by which tumor cells avoid recognition by or induce tolerance in the immune system. However, little is known about whether cancer cells also acquire an intrinsic ability to resist killing by immune effectors. We find that cancer cells enhance their ability to withstand an attack by cytotoxic immune effector cells via acquisition of specific genetic alterations that interfere with the shared mitochondrial death signaling pathway entrained by granzyme B, IFN-gamma, and Apo2 ligand/tumor necrosis factor-related apoptosis inducing ligand (Apo2L/TRAIL), three key mediators of immunologic cell-mediated cytotoxicity. We show that the coexistence of specific mitochondrial signaling defects (either deletion of Bax, overexpression of Bcl-x(L), or deletion of Smac) with expression of X-linked inhibitor of apoptosis protein decreases the sensitivity of cancer cells to IFN-gamma/Apo2L/TRAIL- or granzyme B-induced apoptosis, lymphocyte-mediated cytotoxicity in vitro, and adoptive cellular immunotherapy in vivo. Conversely, negating X-linked inhibitor of apoptosis protein expression or function in tumor cells with defective mitochondrial signaling enables direct activation of caspase-3/-7 by granzyme B or Apo2L/TRAIL, and restores their susceptibility to immunologic cytotoxicity. These findings identify an important mechanism by which cancers evade elimination by immune effector cells and suggest that cancer immunotherapy might be improved by concurrent strategies to alleviate or circumvent the intrinsic mitochondrial death signaling defects that help cancer cells resist immunologic cytotoxicity.


Assuntos
Adenocarcinoma/imunologia , Neoplasias do Colo/imunologia , Imunoterapia Adotiva/métodos , Mitocôndrias/imunologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/imunologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/terapia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/farmacologia , Caspases/metabolismo , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/terapia , Citotoxicidade Imunológica , Ativação Enzimática , Feminino , Granzimas , Células HCT116 , Humanos , Interferon gama/imunologia , Interferon gama/farmacologia , Glicoproteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/enzimologia , Proteínas Recombinantes/farmacologia , Serina Endopeptidases/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/biossíntese , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Blood ; 107(2): 483-91, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16189271

RESUMO

The Sleeping Beauty (SB) transposon system is a nonviral DNA delivery system in which a transposase directs integration of an SB transposon into TA-dinucleotide sites in the genome. To determine whether the SB transposon system can mediate stable gene expression in human T cells, primary peripheral blood lymphocytes (PBLs) were nucleofected with SB vectors carrying a DsRed reporter gene. Plasmids containing the SB transposase on the same molecule as (cis) or on a molecule separate from (trans) the SB transposon mediated long-term and stable reporter gene expression in human primary T cells. Sequencing of transposon:chromosome junctions confirmed that stable gene expression was due to SB-mediated transposition. In other studies, PBLs were successfully transfected using the SB transposon system and shown to stably express a fusion protein consisting of (1) a surface receptor useful for positive T-cell selection and (2) a "suicide" gene useful for elimination of transfected T cells after chemotherapy. This study is the first report demonstrating that the SB transposon system can mediate stable gene transfer in human primary PBLs, which may be advantageous for T-cell-based gene therapies.


Assuntos
Elementos de DNA Transponíveis/genética , Vetores Genéticos , Plasmídeos/farmacologia , Transgenes/fisiologia , Transposases/genética , Transposases/metabolismo , Animais , Técnicas de Transferência de Genes , Genes Reporter/fisiologia , Humanos , Ativação Linfocitária , Suínos , Linfócitos T/citologia , Linfócitos T/metabolismo , Transfecção
18.
Cancer Res ; 65(3): 1079-88, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15705910

RESUMO

A phase I clinical trial with granulocyte-macrophage colony-stimulating factor tumor cell vaccines in patients with metastatic renal cell carcinoma (RCC) showed immune cell infiltration at vaccine sites and delayed-type hypersensitivity (DTH) responses to autologous tumor cells indicative of T-cell immunity. To further characterize RCC T-cell responses and identify relevant RCC-associated antigens, we did a detailed analysis of CD8+ T-cell responses in two vaccinated RCC patients who generated the greatest magnitude of DTH response and also displayed a strong clinical response to vaccination (>90% reduction in metastatic tumor volume). Three separate CD8+ T-cell lines (and subsequent derived clones) derived from patient 24 recognized distinct RCC-associated antigens. One recognized a shared HLA-A*0201-restricted antigen expressed by both renal cancer cells and normal kidney cells. This recognition pattern correlated with a positive DTH test to normal kidney cells despite no evidence of impairment of renal function by the patient's remaining kidney after vaccination. A second line recognized a shared HLA-C7-restricted antigen that was IFN-gamma inducible. A third line recognized a unique HLA-A*0101-restricted RCC antigen derived from a mutated KIAA1440 gene specific to the tumor. In addition, two independent CTL lines and three clones were also generated from patient 26 and they recognized autologous tumor cells restricted through HLA-A*0205, HLA-A/B/C, and HLA-B/C. These results show that paracrine granulocyte-macrophage colony-stimulating factor tumor vaccines may generate a diverse repertoire of tumor-reactive CD8+ T-cell responses and emphasize the importance of polyvalency in the design of cancer immunotherapies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Carcinoma de Células Renais/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Neoplasias Renais/imunologia , Proteínas Quinases Ativadas por Mitógeno/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias , Sequência de Bases , Sítios de Ligação , Linfócitos T CD8-Positivos/efeitos dos fármacos , Células COS , Carcinoma de Células Renais/terapia , Linhagem Celular Tumoral , Chlorocebus aethiops , Células Clonais , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Mapeamento de Epitopos , Antígeno HLA-A2/genética , Antígeno HLA-A2/imunologia , Antígeno HLA-A2/metabolismo , Humanos , Interferon gama/imunologia , Interferon gama/farmacologia , Neoplasias Renais/terapia , Proteínas Quinases Ativadas por Mitógeno/genética , Dados de Sequência Molecular , Transfecção
19.
Hum Gene Ther ; 14(11): 1089-105, 2003 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-12885348

RESUMO

In this report, we evaluated the efficiency of stable gene transfer into established CD8(+) human tumor antigen-specific cytotoxic T cell (CTL) lines and peripheral blood lymphocytes (PBL) by oncoretroviral and lentiviral vectors. In the oncoretroviral vector, the green fluorescent protein (GFP) reporter gene was regulated by the murine stem cell virus (MSCV) promoter. In three human immunodeficiency virus type 1 (HIV-1)-based lentiviral vectors, the GFP transgene was regulated by either a chimeric MSCV/HIV-1 promoter, or cellular promoters from human housekeeping genes PGK and EF1 alpha. We found that several lines of proliferating tumor-specific CTL were poorly (=2%) transduced by the oncoretroviral vector that transduced Jurkat T cell line efficiently (=80%). In contrast, three lentiviral vectors transduced 38-63% of these proliferating CTL. More interestingly, all lentiviral vectors packaged without the HIV-1 accessory proteins transduced human bulk PBL and purified CD4(+) and CD8(+) lymphocyte subsets without prior stimulation. Detailed analysis indicated that the lentiviral vectors containing the EF1 alpha or PGK ubiquitous promoter can transduce unstimulated PBL and achieve low-level transgene expression in the absence of any T-cell activation. However, T-cell activation subsequent to the transduction of unstimulated PBL is required for high-level transgene expression. Transduced PBL expressing transgene delivered by the lentiviral vectors still preserved resting and naïve cell phenotypes. Taken together, prior T cell stimulation and HIV-1 accessory proteins are dispensable for lentivirus-mediated gene transfer into resting naïve and memory T lymphocytes. These results will have significant implications for the study of T-cell biology and for the improvement of clinical gene therapies of acquired immune deficiency syndrome (AIDS) and cancer.


Assuntos
Antígenos de Neoplasias/imunologia , Vetores Genéticos , HIV-1/genética , Lentivirus/genética , Linfócitos T Citotóxicos/imunologia , Animais , Carcinoma de Células Renais/imunologia , Linhagem Celular , Vírus Defeituosos/genética , Citometria de Fluxo , Expressão Gênica , Proteínas de Fluorescência Verde , Humanos , Neoplasias Renais/imunologia , Cinética , Proteínas Luminescentes/genética , Ativação Linfocitária , Camundongos , Retroviridae/genética , Linfócitos T/imunologia , Transdução Genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...