Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 111(29): 10660-5, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002471

RESUMEN

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


Asunto(s)
Aspergilosis/inmunología , Aspergilosis/terapia , Bioingeniería/métodos , Carbohidratos/antagonistas & inhibidores , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/terapia , Linfocitos T/inmunología , Animales , Antígenos CD19/metabolismo , Aspergilosis/microbiología , Aspergilosis/patología , Aspergillus/efectos de los fármacos , Aspergillus/fisiología , Dexametasona/farmacología , Humanos , Hifa/efectos de los fármacos , Hifa/fisiología , Inmunofenotipificación , Lectinas Tipo C/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Infecciones Oportunistas/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos
2.
Nat Med ; 12(10): 1160-6, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16998485

RESUMEN

Immunocompromised individuals are at high risk for life-threatening diseases, especially those caused by cytomegalovirus (CMV), Epstein-Barr virus (EBV) and adenovirus. Conventional therapeutics are primarily active only against CMV, and resistance is frequent. Adoptive transfer of polyclonal cytotoxic T lymphocytes (CTLs) specific for CMV or EBV seems promising, but it is unclear whether this strategy can be extended to adenovirus, which comprises many serotypes. In addition, the preparation of a specific CTL line for each virus in every eligible individual would be impractical. Here we describe genetic modification of antigen-presenting cell lines to facilitate the production of CD4(+) and CD8(+) T lymphocytes specific for CMV, EBV and several serotypes of adenovirus from a single cell culture. When administered to immunocompromised individuals, the single T lymphocyte line expands into multiple discrete virus-specific populations that supply clinically measurable antiviral activity. Monoculture-derived multispecific CTL infusion could provide a safe and efficient means to restore virus-specific immunity in the immunocompromised host.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Técnicas de Cultivo de Célula/métodos , Enfermedades del Sistema Inmune/sangre , Enfermedades del Sistema Inmune/terapia , Adenoviridae/metabolismo , Adolescente , Adulto , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Células Cultivadas , Niño , Preescolar , Citomegalovirus/metabolismo , Femenino , Herpesvirus Humano 4/metabolismo , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Linfocitos T/metabolismo
3.
Blood ; 113(11): 2442-50, 2009 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18971421

RESUMEN

Treatment of Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) with EBV-specific cytotoxic T cells (EBV-specific CTL) has been promising, producing clinical responses. However, infused EBV-specific CTL did not expand in vivo, likely limiting their antitumor activity. Lymphodepleting patients with chemotherapy before T-cell transfer enhances in vivo T-cell expansion, but results in nonspecific destruction of the resident immune system and can have significant toxicity. To evaluate if monoclonal antibodies (mAbs) can produce a more selective lymphodepletion, we conducted a clinical study in which NPC patients received a pair of lymphodepleting mAbs targeted to the CD45 antigen (CD45 mAbs) before EBV-specific CTL infusion. Eight patients with recurrent NPC received CD45 mAbs followed by escalating doses of autologous EBV-specific CTL. Infusion of CD45 mAbs resulted in transient lymphopenia in all patients and an increase in interleukin-15 (IL-15) levels in 6 out 8 patients. All patients had an increase in their peripheral blood frequency of EBV-specific T cells after CTL infusion. Three patients with a persistent increase had clinical benefits including 1 complete response (> 24 months) and 2 with stable disease (for 12 and 15 months). Lymphodepleting mAbs prior CTL transfer may represent an alternative to chemotherapy to enhance expansion of infused CTL. This study is registered at (http://www.clinialtrials.gov) as NCT00608257.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Carcinoma/terapia , Herpesvirus Humano 4/inmunología , Inmunoterapia Adoptiva/métodos , Antígenos Comunes de Leucocito/inmunología , Neoplasias Nasofaríngeas/terapia , Linfocitos T Citotóxicos/trasplante , Adolescente , Adulto , Anticuerpos Monoclonales/farmacología , Carcinoma/complicaciones , Carcinoma/inmunología , Infecciones por Virus de Epstein-Barr/complicaciones , Femenino , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Neoplasias Nasofaríngeas/complicaciones , Neoplasias Nasofaríngeas/inmunología , Linfocitos T Citotóxicos/inmunología , Factores de Tiempo , Acondicionamiento Pretrasplante/métodos
4.
J Exp Med ; 200(12): 1623-33, 2004 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-15611290

RESUMEN

Epstein Barr virus (EBV)+ Hodgkin's disease (HD) expresses clearly identified tumor antigens derived from the virus and could, in principle, be a target for adoptive immunotherapy with viral antigen-specific T cells. However, like most tumor-associated antigens in immunocompetent hosts, these potential targets are only weakly immunogenic, consisting primarily of the latent membrane protein (LMP)1 and LMP2 antigens. Moreover, Hodgkin tumors possess a range of tumor evasion strategies. Therefore, the likely value of immunotherapy with EBV-specific cytotoxic effector cells has been questioned. We have now used a combination of gene marking, tetramer, and functional analyses to track the fate and assess the activity of EBV cytotoxic T lymphocyte (CTL) lines administered to 14 patients treated for relapsed EBV+ HD. Gene marking studies showed that infused effector cells could further expand by several logs in vivo, contribute to the memory pool (persisting up to 12 mo), and traffic to tumor sites. Tetramer and functional analyses showed that T cells reactive with the tumor-associated antigen LMP2 were present in the infused lines, expanded in peripheral blood after infusion, and also entered tumor. Viral load decreased, demonstrating the biologic activity of the infused CTLs. Clinically, EBV CTLs were well tolerated, could control type B symptoms (fever, night sweats, and weight loss), and had antitumor activity. After CTL infusion, five patients were in complete remission at up to 40 mo, two of whom had clearly measurable tumor at the time of treatment. One additional patient had a partial response, and five had stable disease. The performance and fate of these human tumor antigen-specific T cells in vivo suggests that they might be of value for the treatment of EBV+ Hodgkin lymphoma.


Asunto(s)
Infecciones por Herpesviridae/terapia , Herpesvirus Humano 4/inmunología , Enfermedad de Hodgkin/terapia , Inmunoterapia Adoptiva , Linfocitos T Citotóxicos/trasplante , Adolescente , Adulto , Movimiento Celular/inmunología , Niño , Femenino , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/patología , Enfermedad de Hodgkin/virología , Humanos , Masculino , Pronóstico , Inducción de Remisión , Linfocitos T Citotóxicos/inmunología , Escape del Tumor/inmunología , Carga Viral , Proteínas de la Matriz Viral/inmunología
5.
Leuk Lymphoma ; 47(5): 837-42, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16753867

RESUMEN

Immunotherapy approaches with antigen-specific cytotoxic T lymphocytes (CTLs) have provided safe and effective prophylaxis and treatment of Epstein-Barr virus (EBV)-associated lymphomas arising after bone marrow transplantation. EBV is also associated with other malignancies including approximately 40% of cases of Hodgkin's disease, making this tumor another potential target for EBV-targeted immunotherapy. This study describes a patient with multiple relapsed EBV positive Hodgkin's Disease who received both autologous and allogeneic EBV CTL lines. After multiple chemotherapeutic and radiotherapy regimens including two autologous stem cell transplants, he received two doses of gene-marked autologous EBV-specific CTL which resulted in disease stabilization for 6 months. The gene-marked EBV-CTL persisted for 12 months in the peripheral blood after which he proceeded to unrelated donor stem cell transplant followed by immunotherapy with donor-derived EBV-specific CTL. Despite low levels of donor chimerism, the patient remains in complete remission 5 years post-allogeneic SCT. Comparison of the autologous and the donor-derived CTL lines showed that the donor line had specificity for two tumor-associated EBV antigens, latent membrane protein (LMP)1 and 2 compared to the autologous line, which only had specificity for LMP2 epitopes. Following infusion of the donor-derived CTL, functional analyses showed that T-cells reactive with both LMP1 and LMP2 epitopes expanded in the peripheral blood, suggesting that strategies to increase their frequency may result in a broader cytotoxic response against EBV+ Hodgkin tumors.


Asunto(s)
Herpesvirus Humano 4/inmunología , Enfermedad de Hodgkin/terapia , Enfermedad de Hodgkin/virología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T Citotóxicos/trasplante , Proteínas de la Matriz Viral/inmunología , Proliferación Celular , Epítopos/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Inmunoterapia/métodos , Transfusión de Linfocitos/métodos , Masculino , Linfocitos T Citotóxicos/inmunología , Trasplante Homólogo
6.
J Clin Invest ; 126(9): 3363-76, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27482888

RESUMEN

BACKGROUND: T cells expressing antigen-specific chimeric antigen receptors (CARs) improve outcomes for CD19-expressing B cell malignancies. We evaluated a human application of T cells that were genetically modified using the Sleeping Beauty (SB) transposon/transposase system to express a CD19-specific CAR. METHODS: T cells were genetically modified using DNA plasmids from the SB platform to stably express a second-generation CD19-specific CAR and selectively propagated ex vivo with activating and propagating cells (AaPCs) and cytokines. Twenty-six patients with advanced non-Hodgkin lymphoma and acute lymphoblastic leukemia safely underwent hematopoietic stem cell transplantation (HSCT) and infusion of CAR T cells as adjuvant therapy in the autologous (n = 7) or allogeneic settings (n = 19). RESULTS: SB-mediated genetic transposition and stimulation resulted in 2,200- to 2,500-fold ex vivo expansion of genetically modified T cells, with 84% CAR expression, and without integration hotspots. Following autologous HSCT, the 30-month progression-free and overall survivals were 83% and 100%, respectively. After allogeneic HSCT, the respective 12-month rates were 53% and 63%. No acute or late toxicities and no exacerbation of graft-versus-host disease were observed. Despite a low antigen burden and unsupportive recipient cytokine environment, CAR T cells persisted for an average of 201 days for autologous recipients and 51 days for allogeneic recipients. CONCLUSIONS: CD19-specific CAR T cells generated with SB and AaPC platforms were safe, and may provide additional cancer control as planned infusions after HSCT. These results support further clinical development of this nonviral gene therapy approach. TRIAL REGISTRATION: Autologous, NCT00968760; allogeneic, NCT01497184; long-term follow-up, NCT01492036. FUNDING: National Cancer Institute, private foundations, and institutional funds. Please see Acknowledgments for details.


Asunto(s)
Antígenos CD19/metabolismo , Elementos Transponibles de ADN , Linfoma no Hodgkin/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Linfocitos T/citología , Adulto , Células Presentadoras de Antígenos/inmunología , Citocinas/metabolismo , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Seguridad del Paciente , Plásmidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Trasplante Homólogo , Resultado del Tratamiento , Adulto Joven
7.
J Vis Exp ; (104)2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26484782

RESUMEN

The adoptive transfer of pathogen-specific T cells can be used to prevent and treat opportunistic infections such as cytomegalovirus (CMV) infection occurring after allogeneic hematopoietic stem-cell transplantation. Viral-specific T cells from allogeneic donors, including third party donors, can be propagated ex vivo in compliance with current good manufacturing practice (cGMP), employing repeated rounds of antigen-driven stimulation to selectively propagate desired T cells. The identification and isolation of antigen-specific T cells can also be undertaken based upon the cytokine capture system of T cells that have been activated to secrete gamma-interferon (IFN-γ). However, widespread human application of the cytokine capture system (CCS) to help restore immunity has been limited as the production process is time-consuming and requires a skilled operator. The development of a second-generation cell enrichment device such as CliniMACS Prodigy now enables investigators to generate viral-specific T cells using an automated, less labor-intensive system. This device separates magnetically labeled cells from unlabeled cells using magnetic activated cell sorting technology to generate clinical-grade products, is engineered as a closed system and can be accessed and operated on the benchtop. We demonstrate the operation of this new automated cell enrichment device to manufacture CMV pp65-specific T cells obtained from a steady-state apheresis product obtained from a CMV seropositive donor. These isolated T cells can then be directly infused into a patient under institutional and federal regulatory supervision. All the bio-processing steps including removal of red blood cells, stimulation of T cells, separation of antigen-specific T cells, purification, and washing are fully automated. Devices such as this raise the possibility that T cells for human application can be manufactured outside of dedicated good manufacturing practice (GMP) facilities and instead be produced in blood banking facilities where staff can supervise automated protocols to produce multiple products.


Asunto(s)
Eliminación de Componentes Sanguíneos/métodos , Citomegalovirus/inmunología , Inmunoterapia Adoptiva/métodos , Linfocitos T/inmunología , Linfocitos T/virología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Epítopos de Linfocito T/inmunología , Trasplante de Células Madre Hematopoyéticas , Humanos , Fosfoproteínas/inmunología , Linfocitos T/citología , Proteínas de la Matriz Viral/inmunología
8.
PLoS One ; 10(6): e0128151, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26030772

RESUMEN

T cells modified with chimeric antigen receptors (CARs) targeting CD19 demonstrated clinical activity against some B-cell malignancies. However, this is often accompanied by a loss of normal CD19+ B cells and humoral immunity. Receptor tyrosine kinase-like orphan receptor-1 (ROR1) is expressed on sub-populations of B-cell malignancies and solid tumors, but not by healthy B cells or normal post-partum tissues. Thus, adoptive transfer of T cells specific for ROR1 has potential to eliminate tumor cells and spare healthy tissues. To test this hypothesis, we developed CARs targeting ROR1 in order to generate T cells specific for malignant cells. Two Sleeping Beauty transposons were constructed with 2nd generation ROR1-specific CARs signaling through CD3ζ and either CD28 (designated ROR1RCD28) or CD137 (designated ROR1RCD137) and were introduced into T cells. We selected for T cells expressing CAR through co-culture with γ-irradiated activating and propagating cells (AaPC), which co-expressed ROR1 and co-stimulatory molecules. Numeric expansion over one month of co-culture on AaPC in presence of soluble interleukin (IL)-2 and IL-21 occurred and resulted in a diverse memory phenotype of CAR+ T cells as measured by non-enzymatic digital array (NanoString) and multi-panel flow cytometry. Such T cells produced interferon-γ and had specific cytotoxic activity against ROR1+ tumors. Moreover, such cells could eliminate ROR1+ tumor xenografts, especially T cells expressing ROR1RCD137. Clinical trials will investigate the ability of ROR1-specific CAR+ T cells to specifically eliminate tumor cells while maintaining normal B-cell repertoire.


Asunto(s)
Elementos Transponibles de ADN/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interferón gamma/biosíntesis , Leucemia/inmunología , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Fenotipo , Linfocitos T/citología , Linfocitos T/inmunología , Transcripción Genética
9.
Cancer Gene Ther ; 11(2): 81-91, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14685154

RESUMEN

Adoptive immunotherapy with Epstein-Barr virus (EBV)-specific cytotoxic T cells (CTL) is effective for the prophylaxis and treatment of EBV-induced lymphoma in hematopoietic stem cell recipients. However, in EBV-positive Hodgkin's disease (HD) the efficacy of adoptively transferred EBV-specific CTL may be limited by tumor-derived immunosuppressive factors, such as T-cell growth factor (TGF) beta, interleukin (IL)13 and the chemokine TARC. Local delivery of IL12 to tumor sites by tumor-specific CTL could provide direct antitumor effects and overcome the CTL-inhibitory effects of the Th2 tumor environment while avoiding the systemic toxicity of recombinant IL12. EBV-specific CTL transduced with a retrovirus vector expressing the p40 and p35 subunits of IL12 as a single molecule (Flexi-IL12), produced IL12 following antigenic stimulation. This resulted in an elevated production of Th1 cytokines, including interferon gamma and tumor necrosis factor alpha, and a reduction in the Th2 cytokines IL4 and IL5. Flexi-IL12-transduced CTL resisted the antiproliferative and anticytotoxic effects of exogenous TGFbeta, likely by antagonizing the TGFbeta-induced downregulation of the Th1 transcriptional factor T-bet. In addition, Flexi-IL12-transduced CTL demonstrated a proliferative advantage in the presence of inhibitory supernatants from HD-derived cell lines. Tumor-specific, Flexi-IL12-transduced EBV-specific CTL should have a functional advantage over unmodified CTL, particularly in the presence of the adverse Th2 cytokine environment produced by Hodgkin tumor cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Herpesvirus Humano 4/inmunología , Enfermedad de Hodgkin/terapia , Inmunoterapia Adoptiva , Interleucina-12/genética , Linfocitos T Citotóxicos/trasplante , Línea Celular , Citocinas/biosíntesis , Expresión Génica , Vectores Genéticos , Herpesvirus Humano 4/aislamiento & purificación , Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/virología , Humanos , Retroviridae/genética , Retroviridae/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Factor de Crecimiento Transformador beta/farmacología , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/farmacología
10.
Transplantation ; 78(5): 755-7, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15371682

RESUMEN

Epstein-Barr virus (EBV) lymphoproliferative disease (LPD) is a potentially fatal complication that may follow allogeneic hematopoietic stem-cell transplantation (HSCT). In this article, the authors report a 2-year-old girl with Hurler's syndrome who developed multiple central nervous system (CNS) EBV LPD lesions 1 year after unrelated donor HSCT. Before this CNS occurrence, the patient had a complete response to rituximab treatment for EBV LPD of the spleen and lymph nodes; however, treatment of the CNS disease with rituximab proved ineffective. Because of reported favorable response of primary CNS EBV LPD in two human immunodeficiency virus-positive patients, the authors treated this patient with low-dose oral hydroxyurea. The patient improved clinically, with a decrease in size of multiple EBV LPD brain lesions. Subsequently, the patient received EBV-specific cytotoxic T-cell lymphocytes and remains well. The benefit and limited toxicity of hydroxyurea therapy merit its further consideration as treatment for EBV LPD.


Asunto(s)
Infecciones por Virus de Epstein-Barr/etiología , Transfusión de Linfocitos/efectos adversos , Trastornos Linfoproliferativos/etiología , Trasplante de Células Madre/efectos adversos , Adulto , Encéfalo/diagnóstico por imagen , Preescolar , Humanos , Hidroxiurea , Mucopolisacaridosis I/terapia , Linfocitos T/trasplante , Linfocitos T Citotóxicos/inmunología , Tomografía Computarizada por Rayos X
11.
Clin Cancer Res ; 20(22): 5708-19, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24833662

RESUMEN

PURPOSE: To activate and propagate populations of γδ T cells expressing polyclonal repertoire of γ and δ T-cell receptor (TCR) chains for adoptive immunotherapy of cancer, which has yet to be achieved. EXPERIMENTAL DESIGN: Clinical-grade artificial antigen-presenting cells (aAPC) derived from K562 tumor cells were used as irradiated feeders to activate and expand human γδ T cells to clinical scale. These cells were tested for proliferation, TCR expression, memory phenotype, cytokine secretion, and tumor killing. RESULTS: γδ T-cell proliferation was dependent upon CD137L expression on aAPC and addition of exogenous IL2 and IL21. Propagated γδ T cells were polyclonal as they expressed TRDV1, TRDV2-2, TRDV3, TRDV5, TRDV7, and TRDV8 with TRGV2, TRGV3F, TRGV7, TRGV8, TRGV9*A1, TRGV10*A1, and TRGV11 TCR chains. IFNγ production by Vδ1, Vδ2, and Vδ1(neg)Vδ2(neg) subsets was inhibited by pan-TCRγδ antibody when added to cocultures of polyclonal γδ T cells and tumor cell lines. Polyclonal γδ T cells killed acute and chronic leukemia, colon, pancreatic, and ovarian cancer cell lines, but not healthy autologous or allogeneic normal B cells. Blocking antibodies demonstrated that polyclonal γδ T cells mediated tumor cell lysis through combination of DNAM1, NKG2D, and TCRγδ. The adoptive transfer of activated and propagated γδ T cells expressing polyclonal versus defined Vδ TCR chains imparted a hierarchy (polyclonal>Vδ1>Vδ1(neg)Vδ2(neg)>Vδ2) of survival of mice with ovarian cancer xenografts. CONCLUSIONS: Polyclonal γδ T cells can be activated and propagated with clinical-grade aAPCs and demonstrate broad antitumor activities, which will facilitate the implementation of γδ T-cell cancer immunotherapies in humans.


Asunto(s)
Activación de Linfocitos/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/farmacología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Interferón gamma/biosíntesis , Ratones , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/mortalidad , Neoplasias/terapia , ARN Mensajero , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Subgrupos de Linfocitos T/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Vis Exp ; (72): e50070, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23407473

RESUMEN

The potency of clinical-grade T cells can be improved by combining gene therapy with immunotherapy to engineer a biologic product with the potential for superior (i) recognition of tumor-associated antigens (TAAs), (ii) persistence after infusion, (iii) potential for migration to tumor sites, and (iv) ability to recycle effector functions within the tumor microenvironment. Most approaches to genetic manipulation of T cells engineered for human application have used retrovirus and lentivirus for the stable expression of CAR(1-3). This approach, although compliant with current good manufacturing practice (GMP), can be expensive as it relies on the manufacture and release of clinical-grade recombinant virus from a limited number of production facilities. The electro-transfer of nonviral plasmids is an appealing alternative to transduction since DNA species can be produced to clinical grade at approximately 1/10(th) the cost of recombinant GMP-grade virus. To improve the efficiency of integration we adapted Sleeping Beauty (SB) transposon and transposase for human application(4-8). Our SB system uses two DNA plasmids that consist of a transposon coding for a gene of interest (e.g. 2(nd) generation CD19-specific CAR transgene, designated CD19RCD28) and a transposase (e.g. SB11) which inserts the transgene into TA dinucleotide repeats(9-11). To generate clinically-sufficient numbers of genetically modified T cells we use K562-derived artificial antigen presenting cells (aAPC) (clone #4) modified to express a TAA (e.g. CD19) as well as the T cell costimulatory molecules CD86, CD137L, a membrane-bound version of interleukin (IL)-15 (peptide fused to modified IgG4 Fc region) and CD64 (Fc-γ receptor 1) for the loading of monoclonal antibodies (mAb)(12). In this report, we demonstrate the procedures that can be undertaken in compliance with cGMP to generate CD19-specific CAR(+) T cells suitable for human application. This was achieved by the synchronous electro-transfer of two DNA plasmids, a SB transposon (CD19RCD28) and a SB transposase (SB11) followed by retrieval of stable integrants by the every-7-day additions (stimulation cycle) of γ-irradiated aAPC (clone #4) in the presence of soluble recombinant human IL-2 and IL-21(13). Typically 4 cycles (28 days of continuous culture) are undertaken to generate clinically-appealing numbers of T cells that stably express the CAR. This methodology to manufacturing clinical-grade CD19-specific T cells can be applied to T cells derived from peripheral blood (PB) or umbilical cord blood (UCB). Furthermore, this approach can be harnessed to generate T cells to diverse tumor types by pairing the specificity of the introduced CAR with expression of the TAA, recognized by the CAR, on the aAPC.


Asunto(s)
Células Presentadoras de Antígenos/citología , Sangre Fetal/citología , Linfocitos T/fisiología , Transposasas/genética , Presentación de Antígeno , ADN Superhelicoidal/genética , Electroporación , Humanos , Células K562 , Plásmidos/genética , Receptores de Antígenos de Linfocitos T , Linfocitos T/inmunología , Transposasas/metabolismo
13.
PLoS One ; 7(1): e30264, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22279576

RESUMEN

NK cells have therapeutic potential for a wide variety of human malignancies. However, because NK cells expand poorly in vitro, have limited life spans in vivo, and represent a small fraction of peripheral white blood cells, obtaining sufficient cell numbers is the major obstacle for NK-cell immunotherapy. Genetically-engineered artificial antigen-presenting cells (aAPCs) expressing membrane-bound IL-15 (mbIL15) have been used to propagate clinical-grade NK cells for human trials of adoptive immunotherapy, but ex vivo proliferation has been limited by telomere shortening. We developed K562-based aAPCs with membrane-bound IL-21 (mbIL21) and assessed their ability to support human NK-cell proliferation. In contrast to mbIL15, mbIL21-expressing aAPCs promoted log-phase NK cell expansion without evidence of senescence for up to 6 weeks of culture. By day 21, parallel expansion of NK cells from 22 donors demonstrated a mean 47,967-fold expansion (median 31,747) when co-cultured with aAPCs expressing mbIL21 compared to 825-fold expansion (median 325) with mbIL15. Despite the significant increase in proliferation, mbIL21-expanded NK cells also showed a significant increase in telomere length compared to freshly obtained NK cells, suggesting a possible mechanism for their sustained proliferation. NK cells expanded with mbIL21 were similar in phenotype and cytotoxicity to those expanded with mbIL15, with retained donor KIR repertoires and high expression of NCRs, CD16, and NKG2D, but had superior cytokine secretion. The mbIL21-expanded NK cells showed increased transcription of the activating receptor CD160, but otherwise had remarkably similar mRNA expression profiles of the 96 genes assessed. mbIL21-expanded NK cells had significant cytotoxicity against all tumor cell lines tested, retained responsiveness to inhibitory KIR ligands, and demonstrated enhanced killing via antibody-dependent cell cytotoxicity. Thus, aAPCs expressing mbIL21 promote improved proliferation of human NK cells with longer telomeres and less senescence, supporting their clinical use in propagating NK cells for adoptive immunotherapy.


Asunto(s)
Proliferación Celular , Interleucinas/inmunología , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Células Artificiales/inmunología , Células Artificiales/metabolismo , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Interleucina-15/inmunología , Interleucina-15/metabolismo , Interleucinas/metabolismo , Células K562 , Células Asesinas Naturales/metabolismo , Proteínas de la Membrana/metabolismo , Receptores KIR/inmunología , Receptores KIR/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Telómero/genética , Células U937
15.
J Immunother ; 33(9): 983-90, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20948438

RESUMEN

Patients with recurrent or refractory Epstein Barr Virus (EBV)-positive nasopharyngeal carcinoma (NPC) continue to have poor outcomes. Our earlier Phase I dose escalation clinical study of 10 NPC patients showed that infusion of EBV-specific cytotoxic T cells (EBV-CTLs) was safe and had antitumor activity. To better define the overall response rate and discover whether disease status, EBV-antigen specificity, and/or in vivo expansion of infused EBV-CTLs predicted outcome, we treated 13 additional NPC patients with EBV-CTLs in a fixed-dose, Phase II component of the study. We assessed toxicity, efficacy, specificity, and expansion of infused CTLs for all 23 recurrent/refractory NPC patients treated on this Phase I/II clinical study. At the time of CTL infusion, 8 relapsed NPC patients were in remission and 15 had active disease. No significant toxicity was observed. Of the relapsed patients treated in their second or subsequent remission, 62% (5/8) remain disease free (at 17 to 75 mo), whereas 48.7% (7/15) of those with active disease had a CR/CRu (33.3%) or PR (15.4%). In contrast to locoregional disease, metastatic disease was associated with an increased risk of disease progression (HR: 3.91, P=0.015) and decreased overall survival (HR: 5.55, P=0.022). Neither the specificity of the infused CTLs for particular EBV antigens nor their measurable in vivo expansion discernibly influenced outcome. In conclusion, treatment of patients with relapsed/refractory EBV-positive NPC with EBV-CTLs is safe and can be associated with significant, long-term clinical benefit, particularly for patients with locoregional disease.


Asunto(s)
Carcinoma/diagnóstico , Carcinoma/inmunología , Herpesvirus Humano 4/inmunología , Inmunoterapia Adoptiva , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/inmunología , Linfocitos T Citotóxicos/metabolismo , Adolescente , Adulto , Antígenos Virales/inmunología , Carcinoma/patología , Carcinoma/fisiopatología , Carcinoma/terapia , Proliferación Celular , Niño , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Herpesvirus Humano 4/patogenicidad , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/fisiopatología , Neoplasias Nasofaríngeas/terapia , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Pronóstico , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Resultado del Tratamiento
16.
Nat Med ; 14(11): 1264-70, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18978797

RESUMEN

Cytotoxic T lymphocytes (CTLs) directed to nonviral tumor-associated antigens do not survive long term and have limited antitumor activity in vivo, in part because such tumor cells typically lack the appropriate costimulatory molecules. We therefore engineered Epstein-Barr virus (EBV)-specific CTLs to express a chimeric antigen receptor directed to the diasialoganglioside GD2, a nonviral tumor-associated antigen expressed by human neuroblastoma cells. We reasoned that these genetically engineered lymphocytes would receive optimal costimulation after engagement of their native receptors, enhancing survival and antitumor activity mediated through their chimeric receptors. Here we show in individuals with neuroblastoma that EBV-specific CTLs expressing a chimeric GD2-specific receptor indeed survive longer than T cells activated by the CD3-specific antibody OKT3 and expressing the same chimeric receptor but lacking virus specificity. Infusion of these genetically modified cells seemed safe and was associated with tumor regression or necrosis in half of the subjects tested. Hence, virus-specific CTLs can be modified to function as tumor-directed effector cells.


Asunto(s)
Herpesvirus Humano 4/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Recoverina/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Adolescente , Línea Celular , Proliferación Celular , Niño , Preescolar , Femenino , Humanos , Inmunoterapia , Activación de Linfocitos/inmunología , Masculino , Neuroblastoma/inmunología , Neuroblastoma/terapia , Fenotipo , Linfocitos T Citotóxicos/virología
17.
Blood ; 110(8): 2838-45, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17609424

RESUMEN

Epstein-Barr virus (EBV)-associated tumors developing in immunocompetent individuals present a challenge to immunotherapy, since they lack expression of immunodominant viral antigens. However, the tumors consistently express viral proteins including LMP2, which are immunologically "weak" but may nonetheless be targets for immune T cells. We previously showed that a majority of cytotoxic T lymphocytes (CTLs) reactivated using EBV-transformed B-lymphoblastoid cells lines (LCLs) contained minor populations of LMP2-specific T cells and homed to tumor sites. However, they did not produce remissions in patients with bulky disease. We have now used gene transfer into antigen-presenting cells (APCs) to augment the expression and immunogenicity of LMP2. These modified APCs increased the frequency of LMP2-specific CTLs by up to 100-fold compared with unmodified LCL-APCs. The LMP2-specific population expanded and persisted in vivo without adverse effects. Nine of 10 patients treated in remission of high-risk disease remain in remission, and 5 of 6 patients with active relapsed disease had a tumor response, which was complete in 4 and sustained for more than 9 months. It is therefore possible to generate immune responses to weak tumor antigens by ex vivo genetic modification of APCs and the CTLs so produced can have substantial antitumor activity. This study is registered at http://www.cancer.gov/clinicaltrials (protocol IDs: BCM-H-9936, NCT00062868, NCT00070226).


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Inmunoterapia Adoptiva/métodos , Linfoma/terapia , Recurrencia Local de Neoplasia/terapia , Linfocitos T Citotóxicos/trasplante , Proteínas de la Matriz Viral/inmunología , Adolescente , Adulto , Anciano , Niño , Infecciones por Virus de Epstein-Barr/complicaciones , Femenino , Técnicas de Transferencia de Gen , Herpesvirus Humano 4 , Humanos , Linfoma/patología , Linfoma/virología , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Citotóxicos/inmunología , Resultado del Tratamiento , Proteínas de la Matriz Viral/genética
18.
J Immunol ; 175(6): 4137-47, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16148164

RESUMEN

Viral proteins expressed by EBV-associated tumors provide target Ags for immunotherapy. Adoptive T cell therapy has proven effective for posttransplant EBV-associated lymphoma in which all EBV latent Ags are expressed (type III latency). Application of immunotherapeutic strategies to tumors such as nasopharyngeal carcinoma and Hodgkin's lymphoma that have a restricted pattern of EBV Ag expression (type II latency) is under investigation. Potential EBV Ag targets for T cell therapy expressed by these tumors include latent membrane proteins (LMP) 1 and 2. A broad panel of epitopes must be identified from these target Ags to optimize vaccination strategies and facilitate monitoring of tumor-specific T cell populations after immunotherapeutic interventions. To date, LMP2 epitopes have been identified for only a limited number of HLA alleles. Using a peptide library spanning the entire LMP2 sequence, 25 CTL lines from patients with EBV-positive malignancies expressing type II latency were screened for the presence of LMP2-specific T cell populations. In 21 of 25 lines, T cell responses against one to five LMP2 epitopes were identified. These included responses to previously described epitopes as well as to newly identified HLA-A*0206-, A*0204/17-, A29-, A68-, B*1402-, B27-, B*3501-, B53-, and HLA-DR-restricted epitopes. Seven of the nine newly identified epitopes were antigenically conserved among virus isolates from nasopharyngeal carcinoma tumors. These new LMP2 epitopes broaden the diversity of HLA alleles with available epitopes, and, in particular, those epitopes conserved between EBV strains provide valuable tools for immunotherapy and immune monitoring.


Asunto(s)
Herpesvirus Humano 4 , Enfermedad de Hodgkin/inmunología , Neoplasias Nasofaríngeas/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Citotóxicos/inmunología , Proteínas de la Matriz Viral/inmunología , Epítopos de Linfocito T , Antígenos HLA/inmunología , Enfermedad de Hodgkin/terapia , Enfermedad de Hodgkin/virología , Humanos , Inmunoterapia/métodos , Neoplasias Nasofaríngeas/terapia , Neoplasias Nasofaríngeas/virología , Células Tumorales Cultivadas
19.
Blood ; 105(5): 1898-904, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15542583

RESUMEN

Conventional treatment for nasopharyngeal carcinoma (NPC) frequently fails and is accompanied by severe long-term side effects. Since virtually all undifferentiated NPCs are associated with Epstein-Barr virus (EBV), this tumor is an attractive candidate for cellular immunotherapy targeted against tumor-associated viral antigens. We now demonstrate that EBV-specific cytotoxic T-cell (CTL) lines can readily be generated from individuals with NPC, notwithstanding the patients' prior exposure to chemotherapy/radiation. A total of 10 patients diagnosed with advanced NPC were treated with autologous CTLs. All patients tolerated the CTLs, although one developed increased swelling at the site of pre-existing disease. At 19 to 27 months after infusion, 4 patients treated in remission from locally advanced disease remain disease free. Of 6 patients with refractory disease prior to treatment, 2 had complete responses, and remain in remission over 11 to 23 months after treatment; 1 had a partial remission that persisted for 12 months; 1 has had stable disease for more than 14 months; and 2 had no response. These results demonstrate that administration of EBV-specific CTLs to patients with advanced NPC is feasible, appears to be safe, and can be associated with significant antitumor activity.


Asunto(s)
Herpesvirus Humano 4/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Nasofaríngeas/terapia , Linfocitos T Citotóxicos/trasplante , Antígenos Virales , Técnicas de Cultivo de Célula , Humanos , Inmunidad , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/virología , Inducción de Remisión , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Resultado del Tratamiento , Carga Viral
20.
Acta Haematol ; 110(2-3): 149-53, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14583675

RESUMEN

Immunotherapy approaches with antigen-specific cytotoxic T lymphocytes (CTLs) have proved safe and effective prophylaxis and treatment of Epstein-Barr virus (EBV)-associated lymphomas arising after bone marrow transplantation. EBV is also associated with other malignancies including about 40% of cases of Hodgkin's disease making this tumor another potential target for EBV-targeted immunotherapy. While studies with autologous EBV-specific CTLs have shown antiviral activity and immune effects, the clinical responses have been less impressive than those observed in post-transplant lymphomas. There are several possible reasons why the malignant cells in EBV-positive Hodgkin's disease may be less susceptible to immunotherapy approaches, including the fact that they express a more restricted array of EBV-encoded antigens and possess many immune evasion strategies. A number of approaches to overcome these tumor evasion strategies including targeting CTLs to the expressed antigens and genetic modification of CTLs are being evaluated.


Asunto(s)
Traslado Adoptivo , Infecciones por Virus de Epstein-Barr/complicaciones , Enfermedad de Hodgkin/terapia , Enfermedad de Hodgkin/virología , Infecciones por Virus de Epstein-Barr/inmunología , Enfermedad de Hodgkin/inmunología , Humanos , Linfocitos T
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA