Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 2020, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37037829

RESUMEN

Manufacturing autologous chimeric antigen receptor (CAR) T cell therapeutics is complex, and many patients experience treatment delays or cannot be treated at all. Although current allogeneic CAR products have the potential to overcome manufacturing bottlenecks, they are subject to immune rejection and failure to persist in the host, and thus do not provide the same level of efficacy as their autologous counterparts. Here, we aimed to develop universal allogeneic CAR T cells that evade the immune system and produce a durable response. We generated human hypoimmune (HIP) T cells with disrupted B2M, CIITA, and TRAC genes using CRISPR-Cas9 editing. In addition, CD47 and anti-CD19 CAR were expressed using lentiviral transduction. These allogeneic HIP CD19 CAR T cells were compared to allogeneic CD19 CAR T cells that only expressed the anti-CD19 CAR (allo CAR T). In vitro assays for cancer killing and exhaustion revealed no differences between allo CAR T and HIP CAR T cells, confirming that the HIP edits did not negatively affect T cell performance. Clearance of CD19+ tumors by HIP CAR T cells in immunodeficient NSG mice was comparable to that of allo CAR T cells. In fully immunocompetent humanized mice, HIP CAR T cells significantly outperformed allo CAR T cells, showed improved persistence and expansion, and provided lasting cancer clearance. Furthermore, CD47-targeting safety strategies reliably and specifically eliminated HIP CAR T cells. These findings suggest that universal allogeneic HIP CAR T cell-based therapeutics might overcome the limitations associated with poor persistence of allogeneic CAR T cells and exert durable anti-tumor responses.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Receptores Quiméricos de Antígenos/genética , Antígeno CD47 , Linfocitos T , Receptores de Antígenos de Linfocitos T/genética
2.
Mol Ther ; 29(2): 718-733, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33554868

RESUMEN

Allogeneic, off-the-shelf (OTS) chimeric antigen receptor (CAR) cell therapies have the potential to reduce manufacturing costs and variability while providing broader accessibility to cancer patients and those with other diseases. However, host-versus-graft reactivity can limit the durability and efficacy of OTS cell therapies requiring new strategies to evade adaptive and innate-immune responses. Human herpes virus-8 (HHV8) maintains infection, in part, by evading host T and natural killer (NK) cell attack. The viral K3 gene encodes a membrane-tethered E3 ubiquitin ligase that discretely targets major histocompatibility complex (MHC) class I components, whereas K5 encodes a similar E3 ligase with broader specificity, including MHC-II and the MHC-like MHC class I polypeptide-related sequence A (MIC-A)- and sequence B (MIC-B)-activating ligands of NK cells. We created γ-retroviruses encoding K3 and/or K5 transgenes that efficiently transduce primary human T cells. Expression of K3 or K5 resulted in dramatic downregulation of MHC-IA (human leukocyte antigen [HLA]-A, -B, and -C) and MHC class II (HLA-DR) cell-surface expression. K3 expression was sufficient for T cells to resist exogenously loaded peptide-MHC-specific cytotoxicity, as well as recognition in one-way allogeneic mixed lymphocyte reactions. Further, in immunodeficient mice engrafted with allogeneic T cells, K3-transduced T cells selectively expanded in vivo. Ectopic K5 expression in MHC class I-, MIC-A+/B+ K562 cells also reduced targeting by primary NK cells. Coexpression of K3 in prostate stem cell antigen (PSCA)-directed, inducible MyD88/CD40 (iMC)-enhanced CAR-T cells did not impact cytotoxicity, T cell growth, or cytokine production against HPAC pancreatic tumor target cells, whereas K5-expressing cells showed a modest reduction in interleukin (IL)-2 production without effect on cytotoxicity. Together, these results support application of these E3 ligases to advance development of OTS CAR-T cell products.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Ingeniería Genética , Herpesvirus Humano 8/inmunología , Antígenos de Histocompatibilidad/inmunología , Inmunoterapia Adoptiva , Proteínas Virales/inmunología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Humanos , Inmunoterapia Adoptiva/métodos , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Blood Adv ; 4(9): 1950-1964, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32384544

RESUMEN

Natural killer (NK) cells expressing chimeric antigen receptors (CARs) are a promising anticancer immunotherapy, leveraging both innate NK cell antitumor activity and target-specific cytotoxicity. Inducible MyD88/CD40 (iMC) is a potent, rimiducid-regulated protein switch that has been deployed previously as a T-cell activator to enhance proliferation and persistence of CAR-modified T cells. In this study, iMC was extended to CAR-NK cells to enhance their growth and augment cytotoxicity against tumor cells. iMC-activated NK cells substantially increased cytokine and chemokine secretion and displayed higher levels of perforin and granzyme B degranulation. In addition, iMC activation could be coupled with ectopic interleukin-15 (IL-15) to further enhance NK cell proliferation. When coexpressed with a target-specific CAR (CD123 or BCMA), this IL-15/iMC system showed further augmented antitumor activity through enhanced CAR-NK cell expansion and cytolytic activity. To protect against potential toxicity from engineered NK cells, an orthogonal rapamycin-regulated Caspase-9 (iRC9) was included in a 4-gene, dual-switch platform. After infusion of dual-switch NK cells, pharmacologic iRC9 dimerization led to rapid elimination of a majority of expanded transduced NK cells. Thus, CAR-NK cells utilizing dual molecular switches provide an innovative and effective approach to cancer immunotherapy with controlled specificity, efficacy, and safety.


Asunto(s)
Receptores Quiméricos de Antígenos , Interleucina-15/genética , Células Asesinas Naturales , Activación de Linfocitos , Factor 88 de Diferenciación Mieloide , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo
4.
Cytotherapy ; 21(12): 1246-1257, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31837737

RESUMEN

BACKGROUND: Gas Permeable Rapid Expansion (G-Rex) bioreactors have been shown to efficiently expand immune cells intended for therapeutic use, but do not address the complexity of the viral transduction step required for many engineered T-cell products. Here we demonstrate a novel method for transduction of activated T cells with Vectofusin-1 reagent. Transduction is accomplished in suspension, in G-Rex bioreactors. The simplified transduction step is integrated into a streamlined process that uses a single bioreactor with limited operator intervention. METHODS: Peripheral blood mononuclear cells (PBMCs) from healthy donors were thawed, washed and activated with soluble anti-CD3 and anti-CD28 antibodies either in cell culture bags or in G-Rex bioreactors. Cells were cultured in TexMACS GMP medium with interleukin (IL)-7 and IL-15 and transduced with RetroNectin in bags or Vectorfusin-1 in the G-Rex. Total viable cell number, fold expansion, viability, transduction efficiency, phenotype and function were compared between the two processes. RESULTS: The simplified process uses a single vessel from activation through harvest and achieves 56% transduction with 29-fold expansion in 11 days. The cells generated in the simplified process do not differ from cells produced in the conventional bag-based process functionally or phenotypically. DISCUSSION: This study demonstrates that T cells can be transduced in suspension. Further, the conventional method of generating engineered T cells in bags for clinical use can be streamlined to a much simpler, less-expensive process without compromising the quality or function of the cell product.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Organismos Modificados Genéticamente , Linfocitos T/fisiología , Ingeniería de Tejidos/métodos , Transducción Genética/métodos , Reactores Biológicos/normas , Técnicas de Cultivo de Célula/normas , Diferenciación Celular , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/instrumentación , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/normas , Células Cultivadas , Diseño de Equipo , Gases/farmacocinética , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/fisiología , Activación de Linfocitos/inmunología , Organismos Modificados Genéticamente/citología , Permeabilidad , Receptores Quiméricos de Antígenos/genética , Linfocitos T/citología , Transducción Genética/normas
5.
Leukemia ; 33(9): 2195-2207, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30816327

RESUMEN

Successful adoptive chimeric antigen receptor (CAR) T-cell therapies against hematological malignancies require CAR-T expansion and durable persistence following infusion. Balancing increased CAR-T potency with safety, including severe cytokine-release syndrome (sCRS) and neurotoxicity, warrants inclusion of safety mechanisms to control in vivo CAR-T activity. Here, we describe a novel CAR-T cell platform that utilizes expression of the toll-like receptor (TLR) adaptor molecule, MyD88, and tumor-necrosis factor family member, CD40 (MC), tethered to the CAR molecule through an intentionally inefficient 2A linker system, providing a constitutive signal that drives CAR-T survival, proliferation, and antitumor activity against CD19+ and CD123+ hematological cancers. Robust activity of MC-enhanced CAR-T cells was associated with cachexia in animal models that corresponded with high levels of human cytokine production. However, toxicity could be successfully resolved by using the inducible caspase-9 (iC9) safety switch to reduce serum cytokines, by administration of a neutralizing antibody against TNF-α, or by selecting "low" cytokine-producing CD8+ T cells, without loss of antitumor activity. Interestingly, high basal activity was essential for in vivo CAR-T expansion. This study shows that co-opting novel signaling elements (i.e., MyD88 and CD40) and development of a unique CAR-T architecture can drive T-cell proliferation in vivo to enhance CAR-T therapies.


Asunto(s)
Antígenos CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/terapia , Factor 88 de Diferenciación Mieloide/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Animales , Antígenos CD19/inmunología , Proliferación Celular/efectos de los fármacos , Células HEK293 , Humanos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Transducción de Señal/inmunología , Células THP-1
6.
Mol Ther Oncolytics ; 12: 124-137, 2019 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-30740516

RESUMEN

Use of chimeric antigen receptors (CARs) as the basis of targeted adoptive T cell therapies has enabled dramatic efficacy against multiple hematopoietic malignancies, but potency against bulky and solid tumors has lagged, potentially due to insufficient CAR-T cell expansion and persistence. To improve CAR-T cell efficacy, we utilized a potent activation switch based on rimiducid-inducible MyD88 and CD40 (iMC)-signaling elements. To offset potential toxicity risks by this enhanced CAR, an orthogonally regulated, rapamycin-induced, caspase-9-based safety switch (iRC9) was developed to allow in vivo elimination of CAR-T cells. iMC costimulation induced by systemic rimiducid administration enhanced CAR-T cell proliferation, cytokine secretion, and antitumor efficacy in both in vitro assays and xenograft tumor models. Conversely, rapamycin-mediated iRC9 dimerization rapidly induced apoptosis in a dose-dependent fashion as an approach to mitigate therapy-related toxicity. This novel, regulatable dual-switch system may promote greater CAR-T cell expansion and prolonged persistence in a drug-dependent manner while providing a safety switch to mitigate toxicity concerns.

7.
Mol Ther ; 25(9): 2176-2188, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28697888

RESUMEN

Anti-tumor efficacy of T cells engineered to express chimeric antigen receptors (CARs) is dependent on their specificity, survival, and in vivo expansion following adoptive transfer. Toll-like receptor (TLR) and CD40 signaling in T cells can improve persistence and drive proliferation of antigen-specific CD4+ and CD8+ T cells following pathogen challenge or in graft-versus-host disease (GvHD) settings, suggesting that these costimulatory pathways may be co-opted to improve CAR-T cell persistence and function. Here, we present a novel strategy to activate TLR and CD40 signaling in human T cells using inducible MyD88/CD40 (iMC), which can be triggered in vivo via the synthetic dimerizing ligand, rimiducid, to provide potent costimulation to CAR-modified T cells. Importantly, the concurrent activation of iMC (with rimiducid) and CAR (by antigen recognition) is required for interleukin (IL)-2 production and robust CAR-T cell expansion and may provide a user-controlled mechanism to amplify CAR-T cell levels in vivo and augment anti-tumor efficacy.


Asunto(s)
Antígenos CD28/metabolismo , Antígenos CD40/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos CD28/genética , Antígenos CD40/genética , Proliferación Celular , Supervivencia Celular , Análisis por Conglomerados , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inmunoterapia Adoptiva/métodos , Leucemia/genética , Leucemia/inmunología , Leucemia/metabolismo , Leucemia/terapia , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal , Linfocitos T/efectos de los fármacos , Receptores Toll-Like/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Nanobiotechnology ; 14: 24, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27029613

RESUMEN

BACKGROUND: Gold nanoparticles (AuNPs) have shown great promise as scaffolds for gene therapy vectors due to their attractive physiochemical properties which include biocompatibility, ease of functionalization via the nearly covalent gold-sulfur dative bond, and surface plasmon optical properties. Previously, we synthesized stable AuNP-polyamidoamine (AuPAMAM) conjugates and showed their success in vitro as non-viral gene delivery vectors. RESULTS: In this study, we systematically perturbed each component of the AuPAMAM conjugates and analyzed the resulting effect on transfection efficiency. Due to the modular, bottom-up nature of the AuPAMAM synthesis, we were able to probe each step of the fabrication process. The relationship between each conjugation parameter and the function of the final vector were investigated. More than fourfold enhanced transfection efficiency was achieved by modifying the PAMAM concentration, PAMAM core chemistry, PAMAM terminus chemistry, and self-assembled monolayer composition of the AuPAMAM conjugates. CONCLUSIONS: This work suggest that AuPAMAM synthesis platform is a promising non-viral gene therapy approach and highlights the importance of inspecting the role of each individual constituent in all nanotechnology hybrid materials.


Asunto(s)
Dendrímeros/química , Oro/química , Nanopartículas del Metal/química , Materiales Biocompatibles/química , Nanotecnología/métodos , Propiedades de Superficie , Transfección/métodos
9.
PLoS One ; 10(10): e0140744, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26473608

RESUMEN

Adoptive transfer of gene modified T cells provides possible immunotherapy for patients with cancers refractory to other treatments. We have previously used the non-viral piggyBac transposon system to gene modify human T cells for potential immunotherapy. However, these previous studies utilized adoptive transfer of modified human T cells to target cancer xenografts in highly immunodeficient (NOD-SCID) mice that do not recapitulate an intact immune system. Currently, only viral vectors have shown efficacy in permanently gene-modifying mouse T cells for immunotherapy applications. Therefore, we sought to determine if piggyBac could effectively gene modify mouse T cells to target cancer cells in a mouse cancer model. We first demonstrated that we could gene modify cells to express murine interleukin-12 (p35/p40 mIL-12), a transgene with proven efficacy in melanoma immunotherapy. The OT-I melanoma mouse model provides a well-established T cell mediated immune response to ovalbumin (OVA) positive B16 melanoma cells. B16/OVA melanoma cells were implanted in wild type C57Bl6 mice. Mouse splenocytes were isolated from C57Bl6 OT-I mice and were gene modified using piggyBac to express luciferase. Adoptive transfer of luciferase-modified OT-I splenocytes demonstrated homing to B16/OVA melanoma tumors in vivo. We next gene-modified OT-I cells to express mIL-12. Adoptive transfer of mIL-12-modified mouse OT-I splenocytes delayed B16/OVA melanoma tumor growth in vivo compared to control OT-I splenocytes and improved mouse survival. Our results demonstrate that the piggyBac transposon system can be used to gene modify splenocytes and mouse T cells for evaluating adoptive immunotherapy strategies in immunocompetent mouse tumor models that may more directly mimic immunotherapy applications in humans.


Asunto(s)
Traslado Adoptivo , Elementos Transponibles de ADN , Interleucina-12/biosíntesis , Melanoma/terapia , Neoplasias Experimentales/terapia , Bazo , Linfocitos T/trasplante , Animales , Células HeLa , Humanos , Interleucina-12/genética , Melanoma/genética , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Linfocitos T/metabolismo
10.
Methods Mol Biol ; 1139: 169-76, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24619679

RESUMEN

Human T cells can be genetically modified to express tumor-associated antigens (TAA) for the induction of tumor-specific immunity, suggesting that T cells may be alternative candidates of effective antigen-presenting cells (TAPC) and may be useful in vivo as cellular cancer vaccines. The effective induction of TAA-specific T cell immune responses requires activation of T cells by CD3/CD28 antibodies and the presence of proinflammatory cytokines such as interleukin-7 (IL-7) and interleukin-12 (IL-12). Here, we describe the technique of preparing activated human TAPC pulsed with TAA peptides for the induction of tumor antigen-specific T cell immunity in vitro.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Linfocitos T/metabolismo , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Expresión Génica , Antígeno HLA-A2/inmunología , Humanos , Activación de Linfocitos , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología
12.
Biomaterials ; 35(5): 1725-1734, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24286816

RESUMEN

The development of efficient and biocompatible non-viral vectors for gene therapy remains a great challenge, and exploiting the properties of both nanoparticle carriers and cationic polymers is an attractive approach. In this work, we have developed gold nanoparticle (AuNP) polyamidoamine (PAMAM) conjugates for use as non-viral transfection agents. AuPAMAM conjugates were prepared by crosslinking PAMAM dendrimers to carboxylic-terminated AuNPs via EDC and sulfo-NHS chemistry. EDC and sulfo-NHS have been utilized widely and in numerous applications such as amino acid coupling; however, their use in the coupling of PAMAM dendrimers to AuNPs presents new challenges to form effective and stable constructs for delivery that have not yet been examined. Enhanced colloidal stability and DNA condensation ability was established by probing two critical synthetic parameters: the reaction rate of the PAMAM crosslinking step, and the amine to carboxyl ratio. Based on this work, increasing the amine to carboxyl ratio during conjugation of PAMAM onto AuNPs yielded the optimal vector with respect to colloidal stability and transfection efficiency in vitro. AuPAMAM conjugates present attractive candidates for non-viral gene delivery due to their commercial availability, ease of fabrication and scale-up, high yield, high transfection efficiency and low cytotoxicity.


Asunto(s)
Dendrímeros/química , Terapia Genética , Oro/química , Nanopartículas del Metal , Línea Celular , Dendrímeros/síntesis química , Humanos , Microscopía Electrónica de Transmisión , Espectrofotometría Ultravioleta , Transfección
13.
PLoS One ; 8(7): e69073, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935927

RESUMEN

Ablative treatments such as photothermal therapy (PTT) are attractive anticancer strategies because they debulk accessible tumor sites while simultaneously priming antitumor immune responses. However, the immune response following thermal ablation is often insufficient to treat metastatic disease. Here we demonstrate that PTT induces the expression of proinflammatory cytokines and chemokines and promotes the maturation of dendritic cells within tumor-draining lymph nodes, thereby priming antitumor T cell responses. Unexpectedly, however, these immunomodulatory effects were not beneficial to overall antitumor immunity. We found that PTT promoted the infiltration of secondary tumor sites by CD11b(+)Ly-6G/C(+) myeloid-derived suppressor cells, consequently failing to slow the growth of poorly immunogenic B16-F10 tumors and enhancing the growth of distant lung metastases. To exploit the beneficial effects of PTT activity against local tumors and on antitumor immunity whilst avoiding the adverse consequences, we adoptively transferred gp100-specific pmel T cells following PTT. The combination of local control by PTT and systemic antitumor immune reactivity provided by adoptively transferred T cells prevented primary tumor recurrence post-ablation, inhibited tumor growth at distant sites, and abrogated the outgrowth of lung metastases. Hence, the combination of PTT and systemic immunotherapy prevented the adverse effects of PTT on metastatic tumor growth and optimized overall tumor control.


Asunto(s)
Oro/uso terapéutico , Hipertermia Inducida , Inmunoterapia Adoptiva , Melanoma/terapia , Nanocáscaras/uso terapéutico , Fototerapia , Linfocitos T/inmunología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Quimiocinas/metabolismo , Células Dendríticas/metabolismo , Mediadores de Inflamación/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ganglios Linfáticos/patología , Melanoma/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Células Mieloides/patología , Ovalbúmina , Recurrencia
14.
J Neurosci ; 33(33): 13560-8, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23946414

RESUMEN

Contemporary views of tumorigenesis regard its inception as a convergence of genetic mutation and developmental context. Glioma is the most common and deadly malignancy in the CNS; therefore, understanding how regulators of glial development contribute to its formation remains a key question. Previously we identified nuclear factor I-A (NFIA) as a key regulator of developmental gliogenesis, while miR-223 has been shown to repress NFIA expression in other systems. Using this relationship as a starting point, we found that miR-223 can suppress glial precursor proliferation via repression of NFIA during chick spinal cord development. This relationship is conserved in glioma, as miR-223 and NFIA expression is negatively correlated in human glioma tumors, and the miR-223/NFIA axis suppresses tumorigenesis in a human glioma cell line. Subsequent analysis of NFIA function revealed that it directly represses p21 and is required for tumorigenesis in a mouse neural stem cell model of glioma. These studies represent the first characterization of miR-223/NFIA axis function in glioma and demonstrate that it is a conserved proliferative mechanism across CNS development and tumorigenesis.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Glioma/metabolismo , MicroARNs/metabolismo , Factores de Transcripción NFI/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Transformación Celular Neoplásica/genética , Embrión de Pollo , Inmunoprecipitación de Cromatina , Regulación Neoplásica de la Expresión Génica/fisiología , Glioma/genética , Glioma/patología , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , MicroARNs/genética , Factores de Transcripción NFI/genética , Células Madre Neoplásicas/patología , Neuroglía/metabolismo , Neuroglía/patología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
15.
FASEB J ; 27(9): 3753-62, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23752206

RESUMEN

Transposons permit permanent cellular genome engineering in vivo. However, transgene expression falls rapidly postdelivery due to a variety of mechanisms, including immune responses. We hypothesized that delaying initial transgene expression would improve long-term transgene expression by using an engineered piggyBac transposon system that can regulate expression. We found that a 2-part nonviral Tet-KRAB inducible expression system repressed expression of a luciferase reporter in vitro. However, we also observed nonspecific promoter-independent repression. Thus, to achieve temporary transgene repression after gene delivery in vivo, we utilized a nonintegrating version of the repressor plasmid while the gene of interest was delivered in an integrating piggyBac transposon vector. When we delivered the luciferase transposon and repressor to immunocompetent mice by hydrodynamic injection, initial luciferase expression was repressed by 2 orders of magnitude. When luciferase expression was followed long term in vivo, we found that expression was increased >200-fold compared to mice that received only the luciferase transposon and piggyBac transposase. We found that repression of early transgene expression could prevent the priming of luciferase-specific T cells in vivo. Therefore, transient transgene repression postgene delivery is an effective strategy for inhibiting the antitransgene immune response and improving long-term expression in vivo without using immunosuppression.


Asunto(s)
Elementos Transponibles de ADN/genética , Transgenes/genética , Animales , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Células HeLa , Humanos , Immunoblotting , Ratones , Transposasas/genética , Transposasas/metabolismo
16.
Neuron ; 74(1): 79-94, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22500632

RESUMEN

Transcriptional cascades that operate over the course of lineage development are fundamental mechanisms that control cellular differentiation. In the developing central nervous system (CNS), these mechanisms are well characterized during neurogenesis, but remain poorly defined during neural stem cell commitment to the glial lineage. NFIA is a transcription factor that plays a crucial role in the onset of gliogenesis; we found that its induction is regulated by the transcription factor Sox9 and that this relationship mediates the initiation of gliogenesis. Subsequently, Sox9 and NFIA form a complex and coregulate a set of genes induced after glial initiation. Functional studies revealed that a subset of these genes, Apcdd1 and Mmd2, perform key migratory and metabolic roles during astro-gliogenesis, respectively. In sum, these studies delineate a transcriptional regulatory cascade that operates during the initiation of gliogenesis and identifies a unique set of genes that regulate key aspects of astro-glial precursor physiology during development.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción NFI/fisiología , Neuroglía/citología , Factor de Transcripción SOX9/fisiología , Animales , Linaje de la Célula/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Embrión de Pollo , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Ratones , Neuroglía/fisiología , Organogénesis/fisiología , Células Madre/citología , Células Madre/fisiología , Transcripción Genética
17.
J Biomed Biotechnol ; 2011: 417403, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22131805

RESUMEN

The success of cancer vaccines is dependent on the delivery of tumor-associated antigens (TAAs) within lymphoid tissue in the context of costimulatory molecules and immune stimulatory cytokines. Dendritic cells (DCs) are commonly utilized to elicit antitumor immune responses due to their attractive costimulatory molecule and cytokine expression profile. However, the efficacy of DC-based vaccines is limited by the poor viability and lymph-node migration of exogenously generated DCs in vivo. Alternatively, adoptively transferred T cells persist for long periods of time in vivo and readily migrate between the lymphoid and vascular compartments. In addition, T cells may be genetically modified to express both TAA and DC-activating molecules, suggesting that T cells may be ideal candidates to serve as cellular vehicles for antigen delivery to lymph node-resident DCs in vivo. This paper discusses the concept of using T cells to induce tumor-specific immunity for vaccination against cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Traslado Adoptivo , Antígenos de Neoplasias/genética , Citocinas/genética , Citocinas/inmunología , Perfilación de la Expresión Génica , Humanos , Ganglios Linfáticos/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección
18.
Nanoscale Res Lett ; 6(1): 283, 2011 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-21711861

RESUMEN

Gold nanoparticle-mediated photothermal therapy (PTT) has shown great potential for the treatment of cancer in mouse studies and is now being evaluated in clinical trials. For this therapy, gold nanoparticles (AuNPs) are injected intravenously and are allowed to accumulate within the tumor via the enhanced permeability and retention (EPR) effect. The tumor is then irradiated with a near infrared laser, whose energy is absorbed by the AuNPs and translated into heat. While reliance on the EPR effect for tumor targeting has proven adequate for vascularized tumors in small animal models, the efficiency and specificity of tumor delivery in vivo, particularly in tumors with poor blood supply, has proven challenging. In this study, we examine whether human T cells can be used as cellular delivery vehicles for AuNP transport into tumors. We first demonstrate that T cells can be efficiently loaded with 45 nm gold colloid nanoparticles without affecting viability or function (e.g. migration and cytokine production). Using a human tumor xenograft mouse model, we next demonstrate that AuNP-loaded T cells retain their capacity to migrate to tumor sites in vivo. In addition, the efficiency of AuNP delivery to tumors in vivo is increased by more than four-fold compared to injection of free PEGylated AuNPs and the use of the T cell delivery system also dramatically alters the overall nanoparticle biodistribution. Thus, the use of T cell chaperones for AuNP delivery could enhance the efficacy of nanoparticle-based therapies and imaging applications by increasing AuNP tumor accumulation.

19.
J Immunother ; 33(8): 780-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20842059

RESUMEN

For adoptive T-cell therapy to be effective against solid tumors, tumor-specific T cells must be able to migrate to the tumor site. One requirement for efficient migration is that the effector cells express chemokine receptors that match the chemokines produced either by tumor or tumor-associated cells. In this study, we investigated whether the tumor trafficking of activated T cells (ATCs) bearing a chimeric antigen receptor specific for the tumor antigen GD2 (GD2-CAR) could be enhanced by forced coexpression of the chemokine receptor CCR2b, as this receptor directs migration toward CCL2, a chemokine produced by many tumors, including neuroblastoma. Neuroblastoma cell lines (SK-N-SH and SK-N-AS) and primary tumor cells isolated from 6 patients all secreted high levels of CCL2, but GD2-CAR transduced ATCs lacked expression of CCR2 (<5%) and migrated poorly to recombinant CCL2 or tumor supernatants. After retroviral transduction, however, ATCs expressed high levels of CCR2b (>60%) and migrated well in vitro. We expressed firefly luciferase in CCR2b-expressing ATCs and observed improved homing (>10-fold) to CCL2-secreting neuroblastoma compared with CCR2-negative ATCs. As a result, ATCs co-modified with both CCR2b and GD2-CAR had greater antitumor activity in vivo.


Asunto(s)
Movimiento Celular , Inmunoterapia Adoptiva , Neuroblastoma/inmunología , Receptores CCR2/metabolismo , Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/inmunología , Quimiocina CCL2/metabolismo , Citotoxicidad Inmunológica/genética , Humanos , Ratones , Ratones SCID , Neuroblastoma/patología , Neuroblastoma/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CCR2/genética , Receptores CCR2/inmunología , Proteínas Recombinantes de Fusión/genética , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T/trasplante , Transgenes/genética
20.
Mol Ther ; 18(11): 2006-17, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20842106

RESUMEN

Adoptive transfer of antigen-specific cytotoxic T lymphocytes has shown promise for the therapy of cancer. However, tumor-specific T cells are susceptible to diverse inhibitory signals from the tumor microenvironment. The Akt/protein kinase B plays a central role in T-cell proliferation, function, and survival and we hypothesized that expression of constitutively active Akt (caAkt) in T cells could provide resistance to many of these tumor-associated inhibitory mechanisms. caAkt expression in activated human T cells increased proliferation and cytokine production, a likely result of their sustained expression of nuclear factor-κB (NF-κB) and provided resistance to apoptosis by upregulating antiapoptotic molecules. caAkt expressing T cells (caAkt-T-cells) were also relatively resistant to suppression by and conversion into regulatory T cells (Tregs). These characteristics provided a survival advantage to T cells cocultured with tumor cells in vitro; CD3/28-stimulated T cells expressing a chimeric antigen receptor (CAR) specific for disialoganglioside (GD2) that redirected their activity to the immunosuppressive, GD2-expressing neuroblastoma cell line, LAN-1, resisted tumor-induced apoptosis when co-expressing transgenic caAkt. In conclusion, caAkt-transduced T cells showed resistance to several evasion strategies employed by tumors and may therefore enhance the antitumor activity of adoptively transferred T lymphocytes.


Asunto(s)
Apoptosis , FN-kappa B/metabolismo , Neuroblastoma/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos T/metabolismo , Western Blotting , Proliferación Celular , Citometría de Flujo , Gangliósidos/metabolismo , Humanos , Activación de Linfocitos , FN-kappa B/genética , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transducción Genética , Factor de Crecimiento Transformador beta/farmacología , Carga Tumoral/inmunología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...