Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Prostate ; 76(14): 1257-70, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27324746

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR)-modified "designer" T cells (dTc, CAR-T) against PSMA selectively target antigen-expressing cells in vitro and eliminate tumors in vivo. Interleukin 2 (IL2), widely used in adoptive therapies, was proven essential in animal models for dTc to eradicate established solid tumors. METHODS: Patients under-went chemotherapy condi-tion-ing, followed by dTc dosing under a Phase I escalation with continuous infusion low dose IL2 (LDI). A target of dTc escalation was to achieve ≥20% engraftment of infused activated T cells. RESULTS: Six patients enrolled with doses prepared of whom five were treated. Patients received 10(9) or 10(10) autologous T cells, achieving expansions of 20-560-fold over 2 weeks and engraftments of 5-56%. Pharmacokinetic and pharmacodynamic analyses established the impact of conditioning to promote expansion and engraftment of the infused T cells. Unexpectedly, administered IL2 was depleted up to 20-fold with high engraftments of activated T cells (aTc) in an inverse correlation (P < 0.01). Clinically, no anti-PSMA toxicities were noted, and no anti-CAR reactivities were detected post-treatment. Two-of-five patients achieved clinical partial responses (PR), with PSA declines of 50% and 70% and PSA delays of 78 and 150 days, plus a minor response in a third patient. Responses were unrelated to dose size (P = 0.6), instead correlating inversely with engraftment (P = 0.06) and directly with plasma IL2 (P = 0.03), suggesting insufficient IL2 with our LDI protocol to support dTc anti-tumor activity under optimal (high) dTc engraftments. CONCLUSIONS: Under a Phase I dose escalation in prostate cancer, a 20% engraftment target was met or exceeded in three subjects with adequate safety, leading to study conclusion. Clinical responses were obtained but were suggested to be restrained by low plasma IL2 when depleted by high levels of engrafted activated T cells. This report presents a unique example of how the pharmaco-dynamics of "drug-drug" interactions may have a critical impact on the efficacy of their co-application. A new Pilot/Phase II trial is planned to test moderate dose IL2 (MDI) together with high dTc engraftments for anticipated improved therapeutic efficacy. Prostate 76:1257-1270, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Interleucina-2/administración & dosificación , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Receptores de Antígenos de Linfocitos T/administración & dosificación , Linfocitos T/trasplante , Anciano , Antígenos de Superficie/sangre , Glutamato Carboxipeptidasa II/sangre , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/sangre , Trasplante Autólogo/métodos , Resultado del Tratamiento
2.
Clin Cancer Res ; 21(14): 3149-59, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25850950

RESUMEN

PURPOSE: Chimeric antigen receptor-modified T cells (CAR-T) have demonstrated encouraging results in early-phase clinical trials. Successful adaptation of CAR-T technology for CEA-expressing adenocarcinoma liver metastases, a major cause of death in patients with gastrointestinal cancers, has yet to be achieved. We sought to test intrahepatic delivery of anti-CEA CAR-T through percutaneous hepatic artery infusions (HAIs). EXPERIMENTAL DESIGN: We conducted a phase I trial to test HAI of CAR-T in patients with CEA(+) liver metastases. Six patients completed the protocol, and 3 received anti-CEA CAR-T HAIs alone in dose-escalation fashion (10(8), 10(9), and 10(10) cells). We treated an additional 3 patients with the maximum planned CAR-T HAI dose (10(10) cells × 3) along with systemic IL2 support. RESULTS: Four patients had more than 10 liver metastases, and patients received a mean of 2.5 lines of conventional systemic therapy before enrollment. No patient suffered a grade 3 or 4 adverse event related to the CAR-T HAIs. One patient remains alive with stable disease at 23 months following CAR-T HAI, and 5 patients died of progressive disease. Among the patients in the cohort that received systemic IL2 support, CEA levels decreased 37% (range, 19%-48%) from baseline. Biopsies demonstrated an increase in liver metastasis necrosis or fibrosis in 4 of 6 patients. Elevated serum IFNγ levels correlated with IL2 administration and CEA decreases. CONCLUSIONS: We have demonstrated the safety of anti-CEA CAR-T HAIs with encouraging signals of clinical activity in a heavily pretreated population with large tumor burdens. Further clinical testing of CAR-T HAIs for liver metastases is warranted.


Asunto(s)
Adenocarcinoma/terapia , Quimioterapia del Cáncer por Perfusión Regional/métodos , Inmunoterapia/métodos , Neoplasias Hepáticas/terapia , Receptores de Antígenos de Linfocitos T/administración & dosificación , Linfocitos T/trasplante , Adenocarcinoma/secundario , Anciano , Femenino , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/terapia , Humanos , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad
3.
Cytokine ; 71(2): 339-47, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25516298

RESUMEN

T cells reactive to tumor antigens and viral antigens lose their reactivity when exposed to the antigen-rich environment of a larger tumor bed or viral load. Such non-responsive T cells are termed exhausted. T cell exhaustion affects both CD8+ and CD4+ T cells. T cell exhaustion is attributed to the functional impairment of T cells to produce cytokines, of which the most important may be Interleukin 2 (IL2). IL2 performs functions critical for the elimination of cancer cells and virus infected cells. In one such function, IL2 promotes CD8+ T cell and natural killer (NK) cell cytolytic activities. Other functions include regulating naïve T cell differentiation into Th1 and Th2 subsets upon exposure to antigens. Thus, the signaling pathways contributing to T cell exhaustion could be linked to the signaling pathways contributing to IL2 loss. This review will discuss the process of T cell exhaustion and the signaling pathways that could be contributing to T cell exhaustion.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Regulación hacia Abajo , Interleucina-2/metabolismo , Regiones no Traducidas 3' , Animales , Apoptosis , Infecciones Bacterianas/inmunología , Diferenciación Celular , Cromatina/metabolismo , Biología Computacional , Epigénesis Genética , Humanos , Inmunoterapia , Células Asesinas Naturales/citología , Enfermedades Parasitarias/inmunología , ARN Mensajero/metabolismo , Transducción de Señal
4.
Mol Ther Methods Clin Dev ; 1: 14022, 2014 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25419532

RESUMEN

Transduction and expression procedures in gene therapy protocols may optimally transfer more than a single gene to correct a defect and/or transmit new functions to recipient cells or organisms. This may be accomplished by transduction with two (or more) vectors, or, more efficiently, in a single vector. Occasionally, it may be useful to coexpress homologous genes or chimeric proteins with regions of shared homology. Retroviridae include the dominant vector systems for gene transfer (e.g., gamma-retro and lentiviruses) and are capable of such multigene expression. However, these same viruses are known for efficient recombination-deletion when domains are duplicated within the viral genome. This problem can be averted by resorting to two-vector strategies (two-chain two-vector), but at a penalty to cost, convenience, and efficiency. Employing a chimeric antigen receptor system as an example, we confirm that coexpression of two genes with homologous domains in a single gamma-retroviral vector (two-chain single-vector) leads to recombination-deletion between repeated sequences, excising the equivalent of one of the chimeric antigen receptors. Here, we show that a degenerate codon substitution strategy in the two-chain single-vector format efficiently suppressed intravector deletional loss with rescue of balanced gene coexpression by minimizing sequence homology between repeated domains and preserving the final protein sequence.

5.
Prostate ; 74(3): 286-96, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24174378

RESUMEN

BACKGROUND: Adoptive immunotherapy by infusion of designer T cells (dTc) engineered with chimeric antigen receptors (CARs) for tumoricidal activity represents a potentially highly specific modality for the treatment of cancer. In this study, 2nd generation (gen) anti-prostate specific membrane antigen (PSMA) dTc were developed for improving the efficacy of previously developed 1st gen dTc for prostate cancer immunotherapy. The 1st gen dTc are modified with chimeric immunoglobulin-T cell receptor (IgTCR) while the 2nd gen dTc are engineered with an immunoglobulin-CD28-T cell receptor (IgCD28TCR), which incorporates a CD28 costimulatory signal for optimal T cell activation. METHODS: A 2nd gen anti-PSMA IgCD28TCR CAR was constructed by inserting the CD28 signal domain into the 1st gen CAR. 1st and 2nd gen anti-PSMA dTc were created by transducing human T cells with anti-PSMA CARs and their antitumor efficacy was compared for specific activation on PSMA-expressing tumor contact, cytotoxicity against PSMA-expressing tumor cells in vitro, and suppression of tumor growth in an animal model. RESULTS: The 2nd gen dTc can be optimally activated to secrete larger amounts of cytokines such as IL2 and IFNγ than 1st gen and to proliferate more vigorously on PSMA-expressing tumor contact. More importantly, the 2nd gen dTc preserve the PSMA-specific cytotoxicity in vitro and suppress tumor growth in animal models with significant higher potency. CONCLUSIONS: Our results demonstrate that 2nd gen anti-PSMA designer T cells exhibit superior antitumor functions versus 1st gen, providing a rationale for advancing this improved agent toward clinical application in prostate cancer immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias de la Próstata/terapia , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales , Antígenos CD28/inmunología , Línea Celular Tumoral , Membrana Celular/inmunología , Citotoxicidad Inmunológica , Vectores Genéticos/genética , Humanos , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Células Jurkat , Activación de Linfocitos , Masculino , Ratones , Ratones Desnudos , Próstata/inmunología , Receptores de Antígenos/genética , Receptores de Antígenos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión , Retroviridae/genética , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Virology ; 446(1-2): 268-75, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24074590

RESUMEN

The current antiretroviral therapy (ART) can effectively reduce plasma HIV loads to undetectable levels, but cannot eliminate latently infected resting memory CD4 T cells that persist for the lifetime of infected patients. Therefore, designing new therapeutic approaches to eliminate these latently infected cells or the cells that produce HIV upon reactivation from latency is a priority in the ART era in order to progress to a cure of HIV. Here, we show that "designer" T cells expressing chimeric antigen receptor (CAR), CD4-CD28-CD3ζ, can target and kill HIV Env-expressing cells. Further, they secrete effector cytokines upon contact with HIV Env+ target cells that can reactivate latent HIV in a cell line model, thereby exposing those cells to recognition and killing by anti-HIV CAR+ T cells. Taken to the limit, this process could form the basis for an eventual functional or sterilizing cure for HIV in patients.


Asunto(s)
Antígenos CD28/biosíntesis , Complejo CD3/biosíntesis , Antígenos CD4/biosíntesis , VIH/inmunología , Receptores de Antígenos/biosíntesis , Linfocitos T/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Antígenos CD28/genética , Complejo CD3/genética , Antígenos CD4/genética , Línea Celular , Citotoxicidad Inmunológica , Infecciones por VIH/terapia , Humanos , Inmunoterapia/métodos , Receptores de Antígenos/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/biosíntesis
7.
Clin Cancer Res ; 18(21): 5949-60, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22966020

RESUMEN

PURPOSE: Glioblastoma multiforme (GBM) remains highly incurable, with frequent recurrences after standard therapies of maximal surgical resection, radiation, and chemotherapy. To address the need for new treatments, we have undertaken a chimeric antigen receptor (CAR) "designer T cell" (dTc) immunotherapeutic strategy by exploiting interleukin (IL)13 receptor α-2 (IL13Rα2) as a GBM-selective target. EXPERIMENTAL DESIGN: We tested a second-generation IL13 "zetakine" CAR composed of a mutated IL13 extracellular domain linked to intracellular signaling elements of the CD28 costimulatory molecule and CD3ζ. The aim of the mutation (IL13.E13K.R109K) was to enhance selectivity of the CAR for recognition and killing of IL13Rα2(+) GBMs while sparing normal cells bearing the composite IL13Rα1/IL4Rα receptor. RESULTS: Our aim was partially realized with improved recognition of tumor and reduced but persisting activity against normal tissue IL13Rα1(+) cells by the IL13.E13K.R109K CAR. We show that these IL13 dTcs were efficient in killing IL13Rα2(+) glioma cell targets with abundant secretion of cytokines IL2 and IFNγ, and they displayed enhanced tumor-induced expansion versus control unmodified T cells in vitro. In an in vivo test with a human glioma xenograft model, single intracranial injections of IL13 dTc into tumor sites resulted in marked increases in animal survivals. CONCLUSIONS: These data raise the possibility of immune targeting of diffusely invasive GBM cells either via dTc infusion into resection cavities to prevent GBM recurrence or via direct stereotactic injection of dTcs to suppress inoperable or recurrent tumors. Systemic administration of these IL13 dTc could be complicated by reaction against normal tissues expressing IL13Ra1.


Asunto(s)
Glioblastoma/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD28/genética , Antígenos CD28/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Orden Génico , Glioblastoma/mortalidad , Glioblastoma/terapia , Humanos , Inmunoterapia Adoptiva/métodos , Interleucina-13/genética , Interleucina-13/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Mutación , Multimerización de Proteína , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Wound Repair Regen ; 19(4): 498-504, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21649787

RESUMEN

Necrosis of surgically transferred flaps due to ischemia is a serious wound problem. We evaluated the improvement of flap survival and changes in angiogenic gene expression profiles after transfer of the VEGF gene by means of adeno-associated virus type 2 (AAV2) vector to rat ischemic flaps. Thirty rats were divided into one experimental group, one AAV2-GFP group, and one saline group. AAV2-VEGF or AAV2-GFP were injected intradermally into the rat dorsum in the AAV2-VEGF or AAV2-GFP group. The saline group received saline injection. A 3 × 10 cm flap was raised in each rat two weeks post-injection. One week after surgery, flap viability was evaluated. Angiogenesis real-time PCR array was performed to analyze the expression of angiogenesis-associated genes. The AAV2-VEGF treatment significantly improved flap survival (p<0.05). Immunohistochemical staining showed increased VEGF expression in AAV2-VEGF treated flaps. The PCR array identified remarkable changes in 6 out of the 84 angiogenesis-associated genes in AAV2-VEGF treated flaps. Particularly, EGF, PDGF-A and VEGF-B genes were up-regulated in these flaps. In contrast, FGF2 gene expression was down-regulated. In conclusion, AAV2-VEGF improves flap survival and affects the expression of a series of endogenous growth factor genes, which likely play critical roles in the enhancement of ischemic flap survival.


Asunto(s)
Supervivencia de Injerto , Isquemia/terapia , Neovascularización Fisiológica/genética , Colgajos Quirúrgicos/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Dependovirus , Regulación hacia Abajo , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos , Inmunohistoquímica , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba , Factor B de Crecimiento Endotelial Vascular/metabolismo
9.
J Biomed Biotechnol ; 2010: 386545, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20625484

RESUMEN

The successful ex vivo expansion of a large numbers of T cells is a prerequisite for adoptive immunotherapy. In this study, we found that cell density had important effects on the process of expansion of T cells in vitro. Resting T cells were activated to expand at high cell density but failed to be activated at low cell density. Activated T cells (ATCs) expanded rapidly at high cell density but underwent apoptosis at low cell density. Our studies indicated that low-cell-density related ATC death is mediated by oxidative stress. Antioxidants N-acetylcysteine, catalase, and albumin suppressed elevated reactive oxygen species (ROS) levels in low-density cultures and protected ATCs from apoptosis. The viability of ATCs at low density was preserved by conditioned medium from high-density cultures of ATCs in which the autocrine survival factor was identified as catalase. We also found that costimulatory signal CD28 increases T cell activation at lower cell density, paralleled by an increase in catalase secretion. Our findings highlight the importance of cell density in T cell activation, proliferation, survival and apoptosis and support the importance of maintaining T cells at high density for their successful expansion in vitro.


Asunto(s)
Inmunoterapia Adoptiva , Linfocitos T/citología , Linfocitos T/inmunología , Adulto , Apoptosis , Comunicación Autocrina/inmunología , Antígenos CD28/metabolismo , Catalasa/metabolismo , Recuento de Células , Linaje de la Célula , Proliferación Celular , Medios de Cultivo Condicionados , Humanos , Activación de Linfocitos/inmunología , Especies Reactivas de Oxígeno/metabolismo , Solubilidad , Linfocitos T/enzimología , Linfocitos T/metabolismo
10.
J Immunol Methods ; 359(1-2): 11-20, 2010 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-20460126

RESUMEN

Gene-engineered T cell therapy represents a promising strategy to treat cancers. To enable pre-selection of patients sensitive to this type of treatment we have setup and validated a T cell activation assay to test antigen expression on patient-derived tumor tissues. Chimeric antibody-based receptor (CAR) directed against CAIX, currently used in a clinical trial to treat RCC patients, was used as a model receptor. Primary human T cells expressing CAIX CAR were able to respond to CAIX-positive but not CAIX-negative tumor tissue and showed an increased production of IFNgamma, TNFalpha, IL-10 and IL-4, but not IL-2 or IL-5. Tumor tissue driven responses of primary T cells were paralleled by NFAT activation measured in CAR-transduced Jurkat T cells, which was shown to be triggered in a CAR and antigen-specific manner. Next, the reporter gene assay was applied to two independent PSMA CARs, which both mediated NFAT activation in response to tumor tissue. Taken together, a sensitive and donor-independent assay was established to measure T cell activation upon exposure to patient-derived tumor tissue, which may facilitate pre-selection of patients for clinical adoptive T cell therapy.


Asunto(s)
Traslado Adoptivo/métodos , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/terapia , Neoplasias Renales/inmunología , Neoplasias Renales/terapia , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Linfocitos T/trasplante , Reacciones Antígeno-Anticuerpo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/inmunología , Carcinoma de Células Renales/patología , Ingeniería Genética , Humanos , Células Jurkat , Neoplasias Renales/patología , Selección de Paciente , Receptores de Antígenos de Linfocitos T/inmunología , Células Tumorales Cultivadas
11.
Clin Cancer Res ; 16(10): 2769-80, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20460472

RESUMEN

PURPOSE: The aims of this study are to compare antitumor activities of two generations of GD3-specific chimeric antigen receptors (CAR) in human primary T lymphocytes in vitro and to evaluate the antitumor efficacy of using a combination of systemic infusion of interleukin-2 (IL2) and designer T cells to eradicate subcutaneous established GD3+ melanoma in nude mice. EXPERIMENTAL DESIGN: Antitumor activities were compared for two generations of designer T cells, the progenitor first-generation with immunoglobulin T-cell receptor (TCR) with Signal 1 and the second-generation designer T cells with Signal 1+2. Osmotic IL2 pumps were used to deliver the maximum tolerated dose of IL2 to enhance the antitumor effects of designer T cells on subcutaneous established melanoma in nude mice. RESULTS: Melanoma is associated with high expression of ganglioside GD3, which has been targeted with modest effect in antibody therapies. We previously showed that an anti-GD3 CAR (sFv-TCRzeta) will recruit T cells to target this non-T-dependent antigen, with potent killing of melanoma cells. Here, we report the addition of a CD28 costimulation domain to create a second-generation CAR, called Tandem for two signals. We show that this Tandem sFv-CD28/TCRzeta receptor on T cells confers advantages of improved cytokine secretion, cytotoxicity, proliferation, and clonal expansion on tumor contact versus the same CAR without costimulation. In an adoptive transfer model using established melanoma tumors, designer T cells with CD28 showed a 50% rate of complete remissions but only where IL2 was supplemented. CONCLUSIONS: As a reagent for clinical development, the second-generation product is shown to have superior properties to warrant its preference for clinical designer T-cell immunotherapy for melanoma and other tumors. Systemic IL2 was required for optimal activity in an established tumor model.


Asunto(s)
Antígenos CD28/inmunología , Gangliósidos/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Western Blotting , Línea Celular Tumoral , Separación Celular , Quimera , Citocinas/biosíntesis , Femenino , Citometría de Flujo , Humanos , Interleucina-2 , Melanoma Experimental/inmunología , Ratones , Ratones Desnudos , Tumores Neuroectodérmicos/inmunología , Tumores Neuroectodérmicos/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
AIDS Res Hum Retroviruses ; 21(10): 876-81, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16225415

RESUMEN

The F240 human monoclonal antibody specifically recognizes the disulfide loop-bonded immunodominant epitope of gp41 spanning residues 592-606 and expressed broadly on HIV-1 primary isolates. Despite broad reactivity with native virions and HIV-infected cells, the antibody fails to neutralize infection. However, cytoplasmic expression of single-chain antibody (scFv) directed against gp41 of HIV-1 provides a rationale means to inhibit the maturation of envelope protein. The variable regions of the heavy chain and light chain of human monoclonal antibody were amplified by PCR and linked by a 15 amino acid (GGSGS)3 linker in an orientation of VL-linker-VH and retroviral expression vectors were constructed to simultaneously express F240 scFv and eGFP to facilitate selection of scFv-producing cells. Incorporation of a human immunoglobulin signal sequence directed secretion of the F240 scFv (s-scFv) while an otherwise identical vector lacked this sequence (scFv) resulting in intracellular expression of scFv. Transduced human CD4+ H9 T cells were challenged with HIV. While both secreted and nonsecreted F240 scFv inhibited viral production, secretory F240 scFv was more potent. Thus, this novel approach to direct expression of a nonneutralizing scFv using the Ig signal sequence suggests that targeted therapy using antibodies to conserved, highly expressed epitopes may result in a decrease in viral production due to a reduction of viral assembly and/or transport and expression.


Asunto(s)
Anticuerpos Monoclonales/inmunología , VIH-1/fisiología , Región Variable de Inmunoglobulina/inmunología , Replicación Viral/inmunología , Animales , Secuencia de Bases , Cartilla de ADN , Proteína gp41 de Envoltorio del VIH/inmunología , Humanos , Ratones , Células 3T3 NIH , Pruebas de Neutralización
13.
Prostate ; 61(1): 12-25, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15287090

RESUMEN

BACKGROUND: Designer T cells are T lymphocytes engineered toward specific antibody-type membrane antigens through chimeric immunoglobulin-T-cell receptor (IgTCR) genes that have been used for adoptive cellular immunotherapy. We have extended this approach to prostate specific membrane antigen (PSMA) as a means to attack prostate cancer. METHODS: A chimeric anti-PSMA IgTCR gene was constructed based on an anti-PSMA monoclonal antibody, 3D8. Both T-cell lines and primary cultured human T lymphocytes were transduced with the chimeric anti-PSMA IgTCR construct and were analyzed for IgTCR expression, specific activation by PSMA, cytotoxicity against PSMA-expressing tumor cells in vitro, and retardation of tumor growth in an animal model. RESULTS: The IgTCR was incorporated into the TCR-CD3 complex and formed a functional chimeric complex. The IgTCR-modified T cells were specifically activated through the chimeric receptor with PSMA as measured by IL-2 production and increased CD25 expression and specifically lysed the PSMA-expressing prostate cancer cells in vitro as well as retarded tumor growth in an animal model. CONCLUSIONS: The anti-PSMA designer T cells exhibit an antibody-type specificity that can recognize PSMA expressing tumor cells in a MHC-independent fashion, resulting in T-cell activation, target cell lysis in vitro and inhibition of tumor growth in vivo.


Asunto(s)
Antígenos de Superficie/inmunología , Glutamato Carboxipeptidasa II/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Clonación Molecular , Pruebas Inmunológicas de Citotoxicidad , Citometría de Flujo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pruebas de Precipitina , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
14.
Cancer Gene Ther ; 11(4): 297-306, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15002034

RESUMEN

Chimeric immunoglobulin-T-cell receptor (IgTCR)-modified T cells ("designer T cells") kill tumor cells based on antibody-redirected recognition of tumor-associated antigen. Anti-carcinoembryonic antigen (CEA) designer T cells have been prepared and applied in adoptive cellular immunotherapy regimens for CEA-positive cancers. A CEA-immunoglobulin Fc (CEA-Fc) fusion protein was created from the A3B3 region of CEA and the Fc portion of human IgG for the purposes of activation and detection of anti-CEA designer T cells. CEA-Fc was expressed at high yield in CHO cells and purified to homogeneity in a single step on a protein A affinity column. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis revealed that CEA-Fc formed disulfide-linked dimers with a molecular weight of about 170 kDa and a monomer size of 85kDa. The A3B3 CEA component of the CEA-Fc bound to anti-CEA monoclonal antibody MN-14, as well as to the single-chain Fv (sFv) derived from this antibody that was expressed in the IgTCR on the surface of designer T cells. The Fc portion of CEA-Fc was recognized by anti-human IgG Fc antibody and bound by human monocyte Fc receptors. CEA-Fc activated the anti-CEA designer T cells as plate-bound or monocyte-bound form but not as soluble form, as measured by CD69 expression and T-cell proliferation. Our results indicate that the CEA-Fc fusion protein can be used to detect the expression of the anti-CEA IgTCR chimeric receptors on the modified T cells, as well as to serve as an antigen to activate the anti-CEA IgTCR modified T cells. CEA-Fc is the prototype for a new class of antigen-Fc molecules that may significantly augment the analytic and therapeutic goals of adoptive designer T-cell immunotherapies.


Asunto(s)
Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Células CHO , Cricetinae , Cricetulus , Citometría de Flujo , Vectores Genéticos/genética , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Células Jurkat , Activación de Linfocitos/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/análisis , Receptores de Antígenos de Linfocitos T/genética , Receptores Inmunológicos/análisis , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...