Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 10(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35768164

RESUMEN

BACKGROUND: Because of the shortage of ideal cell surface antigens, the development of T-cell receptor (TCR)-engineered T cells (TCR-T) that target intracellular antigens such as NY-ESO-1 is a promising approach for treating patients with solid tumors. However, endogenous TCRs in vector-transduced T cells have been suggested to impair cell-surface expression of transduced TCR while generating mispaired TCRs that can become self-reactive. METHODS: We conducted a first-in-human phase I clinical trial with the TCR-transduced T-cell product (TBI-1301) in patients with NY-ESO-1-expressing solid tumors. In manufacturing TCR-T cells, we used a novel affinity-enhanced NY-ESO-1-specific TCR that was transduced by a retroviral vector that enables siRNA (small interfering RNA)-mediated silencing of endogenous TCR. The patients were divided into two cohorts. Cohort 1 was given a dose of 5×108 cells (whole cells including TCR-T cells) preconditioned with 1500 mg/m2 cyclophosphamide. Cohort 2 was given 5× 109 cells preconditioned with 1500 mg/m2 cyclophosphamide. RESULTS: In vitro study showed that both the CD8+ and CD4+ T fractions of TCR-T cells exhibited cytotoxic effects against NY-ESO-1-expressing tumor cells. Three patients and six patients were allocated to cohort 1 and cohort 2, respectively. Three of the six patients who received 5×109 cells showed tumor response, while three patients developed early-onset cytokine release syndrome (CRS). One of the patients developed a grade 3 lung injury associated with the infiltration of the TCR-T cells. No siRNA-related adverse events other than CRS were observed. Cytokines including interleukin 6 I and monocyte chemotactic protein-1/chemokine (C-C motif) ligand (CCL2)increased in the sera of patients with CRS. In vitro analysis showed these cytokines were not secreted from the T cells infused. A significant fraction of the manufactured T cells in patients with CRS was found to express either CD244, CD39, or both at high levels. CONCLUSIONS: The trial showed that endogenous TCR-silenced and affinity-enhanced NY-ESO-1 TCR-T cells were safely administered except for grade 3 lung injury. The TCR-T cell infusion exhibited significant tumor response and early-onset CRS in patients with tumors that express NY-ESO-1 at high levels. The differentiation properties of the manufactured T cells may be prognostic for TCR-T-related CRS. TRIAL REGISTRATION NUMBER: NCT02366546.


Asunto(s)
Síndrome de Liberación de Citoquinas , Inmunoterapia , Neoplasias , Receptores de Antígenos de Linfocitos T , Linfocitos T , Antígenos de Neoplasias , Ciclofosfamida , Síndrome de Liberación de Citoquinas/terapia , Citocinas/metabolismo , Humanos , Proteínas de la Membrana , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología
2.
Blood ; 130(18): 1985-1994, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28860210

RESUMEN

Wilms' tumor 1 (WT1) is constantly expressed in leukemic cells of acute leukemia and myelodysplastic syndrome (MDS). A T-cell receptor (TCR) that specifically reacts with WT1 peptide in the context of HLA-A*24:02 has been identified. We conducted a first-in-human trial of TCR-gene transduced T-cell (TCR-T-cell) transfer in patients with refractory acute myeloblastic leukemia (AML) and high-risk MDS to investigate the safety and cell kinetics of the T cells. The WT1-specific TCR-gene was transduced to T cells using a retroviral vector encoding small interfering RNAs for endogenous TCR genes. The T cells were transferred twice with a 4-week interval in a dose-escalating design. After the second transfer, sequential WT1 peptide vaccines were given. Eight patients, divided into 2 dose cohorts, received cell transfer. No adverse events of normal tissue were seen. The TCR-T cells were detected in peripheral blood for 8 weeks at levels proportional to the dose administered, and in 5 patients, they persisted throughout the study period. The persisting cells maintained ex vivo peptide-specific immune reactivity. Two patients showed transient decreases in blast counts in bone marrow, which was associated with recovery of hematopoiesis. Four of 5 patients who had persistent T cells at the end of the study survived more than 12 months. These results suggest WT1-specific TCR-T cells manipulated by ex vivo culture of polyclonal peripheral lymphocytes survived in vivo and retained the capacity to mount an immune reaction to WT1. This trial was registered at www.umin.ac.jp as #UMIN000011519.


Asunto(s)
Genes Codificadores de los Receptores de Linfocitos T , Leucemia Mieloide Aguda/terapia , Síndromes Mielodisplásicos/terapia , Linfocitos T/metabolismo , Transducción Genética , Proteínas WT1/genética , Traslado Adoptivo , Anciano , Médula Ósea/patología , Femenino , Humanos , Cinética , Leucemia Mieloide Aguda/genética , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/genética , Péptidos/farmacología
3.
Cytotherapy ; 17(12): 1820-30, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26452983

RESUMEN

BACKGROUND AIMS: Haplo-identical hematopoietic stem cell transplantation (HSCT) with add-back of donor lymphocytes expressing the herpes simplex virus thymidine kinase suicide gene (TK cells) is one of the most widely applied promising new gene therapy approaches. However, the immunological status of added-back TK cells after HSCT has yet to be well characterized. METHODS: We investigated TK cells through the use of flow cytometry, T-cell receptor (TCR) Vß repertoire spectratyping and linear amplification-mediated polymerase chain reaction followed by insertion site analysis in a patient enrolled in our clinical trial. RESULTS: A comparison of onset with remission of acute graft-versus-host disease confirmed that TK cells were predominantly eliminated and that proliferative CD8(+) non-TK cells were also depleted in response to ganciclovir administration. The TCR Vß-chain repertoire of both TK cells and non-TK cells markedly changed after administration of ganciclovir, and, whereas the TCR repertoire of non-TK cells returned to a normal spectratype long after transplantation, that of TK cells remained skewed. With the long-term prophylactic administration of acyclovir, TK cells oligoclonally expanded and the frequency of spliced variants of TK cells increased. Known cancer-associated genes were not evident near the oligoclonally expanded herpes simplex virus (HSV)-TK insertion sites. CONCLUSIONS: We demonstrate obvious differences in immunological status between TK cells and non-TK cells. In addition, we speculate that long-term prophylactic administration of acyclovir increases the risk of oligoclonal expansion of spliced forms of TK cells.


Asunto(s)
Genes Transgénicos Suicidas , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas , Leucemia/terapia , Linfocitos T/metabolismo , Timidina Quinasa/genética , Femenino , Citometría de Flujo , Ganciclovir/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/inmunología , Humanos , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Simplexvirus/genética , Linfocitos T/inmunología , Donantes de Tejidos
5.
Int J Hematol ; 102(1): 101-10, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25948083

RESUMEN

The infusion of donor lymphocytes expressing the herpes simplex virus thymidine kinase suicide gene (TK-cells) is a promising strategy for the treatment of hematologic malignancies relapsing after allogeneic hematopoietic stem cell transplantation. Here we report the results of a phase I clinical trial designed to examine the feasibility, safety, and efficacy of donor lymphocyte infusion (DLI) of TK-cells. Three patients (two with malignant lymphomas, one with acute myeloid leukemia) were enrolled in the trial and received a single DLI of 1 × 10(7) or 5 × 10(7) TK-cells/kg. No local or systemic toxicity related to the gene-transfer procedure was observed. Two patients achieved stable disease. No patient had severe graft-versus-host disease requiring systemic steroid and/or ganciclovir administration. TK-cells were detected in the peripheral blood of all three patients by PCR, but did not persist longer than 28 days. Analysis of cytotoxic T lymphocyte activity detected no immune response against TK-cells by the recipient's own T cells. Flow cytometric analysis showed low proliferative activity and cytotoxic function of TK-cells. In conclusion, DLI of TK-cells was safely performed in all three patients. Our analysis suggests the probable cause of rapid disappearance of TK-cells to be insufficient in vivo expansion of TK-cells in these patients.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Ganciclovir/uso terapéutico , Neoplasias Hematológicas/terapia , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Simplexvirus/genética , Timidina Quinasa/genética , Adulto , Biomarcadores , Supervivencia Celular/efectos de los fármacos , Citotoxicidad Inmunológica , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/terapia , Neoplasias Hematológicas/diagnóstico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Inmunofenotipificación , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Recurrencia , Trasplante Homólogo , Resultado del Tratamiento
6.
Clin Cancer Res ; 21(10): 2268-77, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25855804

RESUMEN

PURPOSE: Preparative lymphodepletion, the temporal ablation of the immune system, has been reported to promote persistence of transferred cells along with increased rates of tumor regression in patients treated with adoptive T-cell therapy. However, it remains unclear whether lymphodepletion is indispensable for immunotherapy with T-cell receptor (TCR) gene-engineered T cells. EXPERIMENTAL DESIGN: We conducted a first-in-man clinical trial of TCR gene-transduced T-cell transfer in patients with recurrent MAGE-A4-expressing esophageal cancer. The patients were given sequential MAGE-A4 peptide vaccinations. The regimen included neither lymphocyte-depleting conditioning nor administration of IL2. Ten patients, divided into 3 dose cohorts, received T-cell transfer. RESULTS: TCR-transduced cells were detected in the peripheral blood for 1 month at levels proportional to the dose administered, and in 5 patients they persisted for more than 5 months. The persisting cells maintained ex vivo antigen-specific tumor reactivity. Despite the long persistence of the transferred T cells, 7 patients exhibited tumor progression within 2 months after the treatment. Three patients who had minimal tumor lesions at baseline survived for more than 27 months. CONCLUSIONS: These results suggest that TCR-engineered T cells created by relatively short-duration in vitro culture of polyclonal lymphocytes in peripheral blood retained the capacity to survive in a host. The discordance between T-cell survival and tumor regression suggests that multiple mechanisms underlie the benefits of preparative lymphodepletion in adoptive T-cell therapy.


Asunto(s)
Antígenos de Neoplasias/genética , Carcinoma de Células Escamosas/terapia , Neoplasias Esofágicas/terapia , Proteínas de Neoplasias/genética , Recurrencia Local de Neoplasia/terapia , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Traslado Adoptivo , Adulto , Anciano , Carcinoma de Células Escamosas/inmunología , Supervivencia Celular , Células Cultivadas , Neoplasias Esofágicas/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/inmunología , Linfocitos T/trasplante , Transducción Genética , Resultado del Tratamiento
7.
Eur J Immunol ; 44(6): 1747-58, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24723437

RESUMEN

T cells express multiple integrin molecules. The significance of signaling through these molecules on acquisition of T-cell effector functions and memory formation capacity remains largely unknown. Moreover, the impact of stimulation through these signals on the generation of T cells for adoptive immunotherapy has not been elucidated. In this study, using a recombinant fragment of fibronectin, CH-296, we demonstrated that stimulation via very late Ag (VLA)-4 and VLA-5 in human and BALB/c mouse CD8(+) T cells, in combination with TCR stimulation, enhances effector multifunctionality and in vivo memory formation. Using TCR-transgenic mouse-derived CD8(+) T cells expressing TCR specific for the syngeneic CMS5 fibrosarcoma-derived tumor Ag, we showed that stimulation by CH-296 improved the ability of tumor-specific CD8(+) T cells to inhibit CMS5 tumor growth when adoptively transferred into hosts with progressing tumors. Improved antitumor effects were associated with decreased infiltration of Foxp3(+) CD4(+) Treg cells in tumors. These results suggest that stimulation via VLA-4 and VLA-5 modulates the qualities of effector T cells and could potentially increase the efficacy of adoptive therapy against cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Fibrosarcoma/inmunología , Memoria Inmunológica , Integrina alfa4beta1/inmunología , Integrina alfa5beta1/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos , Línea Celular Tumoral , Femenino , Fibrosarcoma/genética , Fibrosarcoma/patología , Fibrosarcoma/terapia , Humanos , Integrina alfa4beta1/genética , Integrina alfa5beta1/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
8.
PLoS One ; 9(1): e86275, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24454964

RESUMEN

In retroviral vector-mediated gene transfer, transduction efficiency can be hampered by inhibitory molecules derived from the culture fluid of virus producer cell lines. To remove these inhibitory molecules to enable better gene transduction, we had previously developed a transduction method using a fibronectin fragment-coated vessel (i.e., the RetroNectin-bound virus transduction method). In the present study, we developed a method that combined RetroNectin-bound virus transduction with low-temperature shaking and applied this method in manufacturing autologous retroviral-engineered T cells for adoptive transfer gene therapy in a large-scale closed system. Retroviral vector was preloaded into a RetroNectin-coated bag and incubated at 4°C for 16 h on a reciprocating shaker at 50 rounds per minute. After the supernatant was removed, activated T cells were added to the bag. The bag transduction method has the advantage of increasing transduction efficiency, as simply flipping over the bag during gene transduction facilitates more efficient utilization of the retroviral vector adsorbed on the top and bottom surfaces of the bag. Finally, we performed validation runs of endoribonuclease MazF-modified CD4(+) T cell manufacturing for HIV-1 gene therapy and T cell receptor-modified T cell manufacturing for MAGE-A4 antigen-expressing cancer gene therapy and achieved over 200-fold (≥ 10(10)) and 100-fold (≥ 5 × 10(9)) expansion, respectively. In conclusion, we demonstrated that the large-scale closed transduction system is highly efficient for retroviral vector-based T cell manufacturing for adoptive transfer gene therapy, and this technology is expected to be amenable to automation and improve current clinical gene therapy protocols.


Asunto(s)
Fibronectinas/química , Retroviridae/genética , Transducción Genética/métodos , Adsorción , Linfocitos T CD4-Positivos , Técnicas de Cultivo de Célula , Línea Celular , Materiales Biocompatibles Revestidos , Terapia Genética , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Humanos , Regiones Promotoras Genéticas , Receptores de Antígenos de Linfocitos T/biosíntesis , Receptores de Antígenos de Linfocitos T/genética , Temperatura
9.
Neuro Oncol ; 15(6): 747-58, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23460320

RESUMEN

BACKGROUND: Meningiomas are the most commonly diagnosed primary intracranial neoplasms. Despite significant advances in modern therapies, the management of malignant meningioma and skull base meningioma remains a challenge. Thus, the development of new treatment modalities is urgently needed for these difficult-to-treat meningiomas. The goal of this study was to investigate the potential of build-in short interfering RNA-based Wilms' tumor protein (WT1)-targeted adoptive immunotherapy in a reproducible mouse model of malignant skull base meningioma that we recently established. METHODS: We compared WT1 mRNA expression in human meningioma tissues and gliomas by quantitative real-time reverse-transcription polymerase chain reaction. Human malignant meningioma cells (IOMM-Lee cells) were labeled with green fluorescent protein (GFP) and implanted at the skull base of immunodeficient mice by using the postglenoid foramen injection (PGFi) technique. The animals were sacrificed at specific time points for analysis of tumor formation. Two groups of animals received adoptive immunotherapy with control peripheral blood mononuclear cells (PBMCs) or WT1-targeted PBMCs. RESULTS: High levels of WT1 mRNA expression were observed in many meningioma tissues and all meningioma cell lines. IOMM-Lee-GFP cells were successfully implanted using the PGFi technique, and malignant skull base meningiomas were induced in all mice. The systemically delivered WT1-targeted PBMCs infiltrated skull base meningiomas and significantly delayed tumor growth and increased survival time. CONCLUSIONS: We have established a reproducible mouse model of malignant skull base meningioma. WT1-targeted adoptive immunotherapy appears to be a promising approach for the treatment of difficult-to-treat meningiomas.


Asunto(s)
Modelos Animales de Enfermedad , Ingeniería Genética , Inmunoterapia Adoptiva , Neoplasias Meníngeas/terapia , Meningioma/terapia , Neoplasias de la Base del Cráneo/terapia , Linfocitos T/inmunología , Proteínas WT1/metabolismo , Traslado Adoptivo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Western Blotting , Proliferación Celular , Femenino , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Masculino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/inmunología , Meningioma/genética , Meningioma/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Base del Cráneo/genética , Neoplasias de la Base del Cráneo/inmunología , Linfocitos T/trasplante , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Células Tumorales Cultivadas , Proteínas WT1/genética
10.
Cancer Sci ; 103(1): 17-25, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21951605

RESUMEN

Adoptive cell therapy with lymphocytes that have been genetically engineered to express tumor-reactive T-cell receptors (TCR) is a promising approach for cancer immunotherapy. We have been exploring the development of TCR gene therapy targeting cancer/testis antigens, including melanoma-associated antigen (MAGE) family antigens, that are ideal targets for adoptive T-cell therapy. The efficacy of TCR gene therapy targeting MAGE family antigens, however, has not yet been evaluated in vivo. Here, we demonstrate the in vivo antitumor activity in immunodeficient non-obese diabetic/SCID/γc(null) (NOG) mice of human lymphocytes genetically engineered to express TCR specific for the MAGE-A4 antigen. Polyclonal T cells derived from human peripheral blood mononuclear cells were transduced with the αß TCR genes specific for MAGE-A4, then adoptively transferred into NOG mice inoculated with MAGE-A4 expressing human tumor cell lines. The transferred T cells maintained their effector function in vivo, infiltrated into tumors, and inhibited tumor growth in an antigen-specific manner. The combination of adoptive cell therapy with antigen peptide vaccination enhanced antitumor activity, with improved multifunctionality of the transferred cells. These data suggest that TCR gene therapy with MAGE-A4-specific TCR is a promising strategy to treat patients with MAGE-A4-expressing tumors; in addition, the acquisition of multifunctionality in vivo is an important factor to predict the quality of the T-cell response during adoptive therapy with human lymphocytes.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Esofágicas/terapia , Terapia Genética , Inmunoterapia Adoptiva , Neoplasias Pulmonares/terapia , Proteínas de Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Animales , Antígenos de Neoplasias/genética , Terapia Combinada , Citotoxicidad Inmunológica/inmunología , Neoplasias Esofágicas/inmunología , Femenino , Citometría de Flujo , Vectores Genéticos/uso terapéutico , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Humanos , Técnicas para Inmunoenzimas , Leucocitos Mononucleares/inmunología , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/genética , Retroviridae , Transducción Genética , Vacunas de Subunidad/uso terapéutico
11.
J Biochem ; 149(3): 285-92, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21106541

RESUMEN

Large scale T-cell expansion and efficient gene transduction are required for adoptive T-cell gene therapy. Based on our previous observations, human peripheral blood mononuclear cells (PBMCs) can be expanded efficiently while conserving a naïve phenotype by stimulating with both recombinant human fibronectin fragment (CH-296) and anti-CD3 monoclonal antibodies. In this article, we explored the possibility of using this co-stimulation method to generate engineered T cells using lentiviral vector. Human PBMCs were stimulated with anti-CD3 together with immobilized CH-296 or anti-CD28 antibody as well as anti-CD3/anti-CD28 conjugated beads and transduced with lentiviral vector simultaneously. Co-stimulation with CH-296 gave superior transduction efficiency than with anti-CD28. Next, PBMCs were stimulated and transduced with anti-CD3/CH-296 or with anti-CD3/CD28 beads. T-cell expansion, gene transfer efficiencies and immunophenotypes were analysed. Stimulation with anti-CD3/CH-296 resulted in more than 10-times higher cell expansion and higher gene transfer efficiency with conservation of the naïve phenotype compared with anti-CD3/CD28 stimulation method. Thus, lentiviral transduction with anti-CD3/CH-296 co-stimulation is an efficient way to generate large numbers of genetically modified T cells and may be suitable for many gene therapy protocols that use adoptive T-cell transfer therapy.


Asunto(s)
Fibronectinas/farmacología , Vectores Genéticos/genética , Lentivirus/genética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Transducción Genética/métodos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Fibronectinas/química , Citometría de Flujo , Humanos , Inmunoterapia Adoptiva , Leucocitos Mononucleares/citología , Activación de Linfocitos/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
12.
J Transl Med ; 3(1): 4, 2005 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-15676080

RESUMEN

BACKGROUND: Metastatic, chemotherapy-resistant melanoma is an intractable cancer with a very poor prognosis. As to immunotherapy targeting metastatic melanoma, HLA-A2+ patients were mainly enrolled in the study in Western countries. However, HLA-A24+ melanoma patients-oriented immunotherapy has not been fully investigated. In the present study, we investigated the effect of dendritic cell (DC)-based immunotherapy on metastatic melanoma patients with HLA-A2 or A24 genotype. METHODS: Nine cases of metastatic melanoma were enrolled into a phase I study of monocyte-derived dendritic cell (DC)-based immunotherapy. HLA-genotype analysis revealed 4 cases of HLA-A*0201, 1 of A*0206 and 4 of A*2402. Enriched monocytes were obtained using OptiPreptrade mark from leukapheresis products, and then incubated with GM-CSF and IL-4 in a closed serum-free system. After pulsing with a cocktail of 5 melanoma-associated synthetic peptides (gp100, tyrosinase, MAGE-2, MAGE-3 and MART-1 or MAGE-1) restricted to HLA-A2 or A24 and KLH, cells were cryopreserved until used. Finally, thawed DCs were washed and injected subcutaneously (s.c.) into the inguinal region in a dose-escalation manner. RESULTS: The mean percentage of DCs rated as lin-HLA-DR+ in melanoma patients was 46.4 +/- 15.6 %. Most of DCs expressed high level of co-stimulatory molecules and type1 phenotype (CD11c+HLA-DR+), while a moderate number of mature DCs with CD83 and CCR7 positive were contained in DC products. DC injections were well tolerated except for transient liver dysfunction (elevation of transaminases, Grade I-II). All 6 evaluable cases except for early PD showed positive immunological responses to more than 2 melanoma peptides in an ELISPOT assay. Two representative responders demonstrated strong HLA-class I protein expression in the tumor and very high scores of ELISPOT that might correlate to the regression of metastatic tumors. Clinical response through DC injections was as follows : 1CR, 1 PR, 1SD and 6 PD. All 59 DC injections in the phase I study were tolerable in terms of safety, however, the maximal tolerable dose of DCs was not determined. CONCLUSIONS: These results suggested that peptide cocktail-treated DC-based immunotherapy had the potential for utilizing as one of therapeutic tools against metastatic melanoma in Japan.

13.
Oncol Rep ; 12(4): 725-31, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15375491

RESUMEN

Epstein-Barr virus (EBV) is associated with several types of malignancies including Burkitt's lymphoma, Hodgkin's disease, nasopharyngeal carcinoma, and gastric carcinoma. Previous reports have suggested that EBV-related antigen-targeting immunotherapy is one of the promising approaches for the treatment of these malignancies other than gastric carcinoma. EBV-associated gastric carcinoma (EBVaGC) has been shown to express Epstein-Barr virus nuclear antigen 1 (EBNA1) and latent membrane protein 2 (LMP2). In the present study, DNA and mRNA freshly isolated from tumors of patients with gastric cancer were subjected to polymerase chain reaction (PCR) using EBV-specific primers and reverse transcription (RT)-PCR specific for LMP2 transcripts. EBV-specific region was identified in genomic DNA isolated from cancerous tissues in 22% of gastric cancer patients. LMP2 mRNA was also detected in 3 out of these 5 DNA positive samples tested. To investigate the feasibility of specific immunotherapy for EBVaGC, we induced cytotoxic T lymphocytes (CTLs) from peripheral blood lymphocytes using two kinds of antigen-presenting cells (APCs) such as autologous lymphoblastoid cell line (LCL) and LMP2-derived peptide-pulsed dendritic cells (DCs). The cytotoxicity of these CTLs against peptide-pulsed targets was examined by standard 51Cr release assay and interferon (IFN)-gamma production assay. We further assessed the recognition of tumor cells endogenously expressing LMP2 by these T cells. T cells induced by peptide-loaded DCs and autologous LCL efficiently lysed peptide-pulsed targets. Furthermore, these T cells could recognize not only tumor cells transfected with LMP2, but also LMP2-positive gastric cancer cells which were successfully isolated and cultured from specimens obtained by surgery. Collectively, sensitization of peripheral blood lymphocytes with LMP2-derived peptide was able to induce CTL response against EBVaGC cells. Thus, EBVaGC is susceptible for the LMP2-targeting immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Infecciones por Virus de Epstein-Barr/terapia , Herpesvirus Humano 4/inmunología , Linfocitos/inmunología , Neoplasias Gástricas/etiología , Linfocitos T Citotóxicos/inmunología , Proteínas de la Matriz Viral/inmunología , Anciano , Anciano de 80 o más Años , Células Presentadoras de Antígenos , Cromo/metabolismo , ADN de Neoplasias , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Antígenos HLA-A/inmunología , Antígeno HLA-A24 , Humanos , Técnicas In Vitro , Interferón gamma/metabolismo , Linfocitos/metabolismo , Linfocitos/patología , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/inmunología , ARN Neoplásico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/terapia , Células Tumorales Cultivadas , Proteínas de la Matriz Viral/genética
14.
Anticancer Res ; 24(2B): 571-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15160996

RESUMEN

Many human leukocyte antigen (HLA)-class I (mainly A*0201)-restricted peptide-specific cytotoxic T cells (CTLs) have been derived from peripheral blood lymphocytes (PBLs) of melanoma patients. However, few studies regarding HLA-A*2402-restricted melanoma-associated peptides have been performed, because HLA-A24 is not a common allele in Caucasians. In this study, we investigated the specific CTL-inducing activity of 5 HLA-A*2402-restricted peptides derived from gp100, tyrosinase, MAGE1, MAGE2 and MAGE3. A CTL induction culture was performed using PBLs and cultured dendritic cell (DC) pulsed with HLA-A*2402-restricted melanoma peptide cocktail. The CTLs derived from volunteers killed the A24 peptide-pulsed TISI cells and even HLA-A*2402-positive melanoma cells, but not HLA-A*0201-positive cells. IFN-gamma levels produced by the melanoma patients' CTLs were obviously low in each peptide group compared with those produced by the volunteers' CTLs, which indicated the presence of immunosuppressive factors in metastatic melanoma. These results suggested that polyvalent immunotherapy using multiple epitopes from melanoma antigens might be a better way of improving the efficacy of treatment.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Antígenos HLA-A/inmunología , Melanoma/inmunología , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos de Neoplasias/biosíntesis , Células Dendríticas/inmunología , Antígeno HLA-A24 , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Activación de Linfocitos , Melanoma/metabolismo , Melanoma/terapia , Antígenos Específicos del Melanoma , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/inmunología , Monofenol Monooxigenasa/biosíntesis , Monofenol Monooxigenasa/inmunología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/inmunología , Linfocitos T Citotóxicos/metabolismo , Antígeno gp100 del Melanoma
15.
Int J Oncol ; 24(4): 909-17, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15010829

RESUMEN

We conducted a clinical study of cancer vaccine therapy with dendritic cells (DCs) and HLA-A24-restricted carcinoembryonic antigen (CEA)-derived peptide to assess the feasibility and efficacy of such therapy. Eighteen patients with CEA-expressing metastatic gastrointestinal or lung adenocarcinomas who were positive for human leukocyte antigen (HLA)-A24 were enrolled. DCs were generated from the patients' autologous monocyte-enriched fractions of granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cells in the presence of granulocyte/macrophage colony-stimulating factor and interleukin-4. The generated DCs were pulsed with CEA-derived, HLA-A24-restricted 9-mer peptide (CEA652) and injected into the patients intradermally and subcutaneously every 2 weeks. Toxicity and clinical and immunological responses were closely monitored in each patient. No severe toxicity directly attributable to the treatment was observed, and the vaccine was well tolerated. Although no definite tumor shrinkage occurred in any patient, long-term stable disease or marked decreases in the serum CEA level were observed in some patients after therapy. Most of the patients in whom treatment was clinically effective showed a positive skin response to CEA652-pulsed DCs (delayed-type hypersensitivity skin test) and a positive in vitro CTL response to CEA652 peptide after therapy. We conclude that active specific immunotherapy using DCs pulsed with CEA652 is a safe and feasible treatment that is clinically effective in some patients with metastatic gastrointestinal or lung adenocarcinomas. Our results will hopefully encourage further refinement and development of DC-based immunotherapy with HLA-A24-restricted CEA-derived peptide for refractory solid cancers that express CEA.


Asunto(s)
Adenocarcinoma/terapia , Células Dendríticas/inmunología , Neoplasias Gastrointestinales/terapia , Inmunoterapia , Neoplasias Pulmonares/terapia , Linfocitos T Citotóxicos/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/secundario , Adulto , Anciano , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/análisis , Antígeno Carcinoembrionario/inmunología , Estudios de Factibilidad , Femenino , Neoplasias Gastrointestinales/inmunología , Neoplasias Gastrointestinales/secundario , Factor Estimulante de Colonias de Granulocitos/farmacología , Antígenos HLA-A/inmunología , Antígeno HLA-A24 , Humanos , Hipersensibilidad Tardía/etiología , Neoplasias Pulmonares/inmunología , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
16.
Cancer Immunol Immunother ; 53(7): 609-16, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-14735319

RESUMEN

Carcinoembryonic antigen (CEA) is strongly expressed in a vast majority of gastrointestinal carcinomas. Recently, epitope peptides of CEA were identified. We have demonstrated HLA-A24-restricted peptide, CEA652[9] (TYACFVSNL), was capable of eliciting specific cytotoxic T lymphocytes (CTLs) which could lyse tumor cells expressing HLA-A24 and CEA. HLA-A24 is the most applicable MHC class I allele in the Japanese population. In this pilot study, we have used the peptide-pulsed dendritic cells (DCs) generated from peripheral blood mononuclear cells (PBMCs) supplemented with GM-CSF and IL-4 as the source of the vaccine. Eight patients with advanced CEA-expressing gastrointestinal malignancies received subcutaneous injections every 2 or 3 weeks. Immunomonitoring was performed by ELISpot (enzyme-linked immunosorbent spot) assay to measure the precursor frequency of CTLs and their capacity to elicit antitumor CTLs in vitro. Four of seven patients have developed their CTL response after vaccinations. DTH reaction was observed in one of eight patients at the DC-injected site. Skin biopsy at the injected site showed the infiltration of the lymphocytes. Furthermore, A24/CEA peptide tetramer assay revealed an increase in peptide-specific T-cell precursor frequency in vaccinated patients. No significant toxic adverse effects were observed, except for mild diarrhea in one case after three vaccinations. Three patients have shown stabilization of the disease after vaccinations. In conclusion, our results clearly demonstrated that our vaccination protocol was safe and might develop a CEA-specific CTL response in cancer patients.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Células Dendríticas/inmunología , Neoplasias Gastrointestinales/inmunología , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunación , Pruebas Inmunológicas de Citotoxicidad , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/secundario , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Antígenos HLA-A/metabolismo , Antígeno HLA-A24 , Humanos , Inyecciones Subcutáneas , Interleucina-4/uso terapéutico , Japón , Activación de Linfocitos , Proyectos Piloto
17.
Clin Cancer Res ; 8(12): 3885-92, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12473604

RESUMEN

PURPOSE: Prostate-specific membrane antigen (PSMA), which is a transmembrane glycoprotein predominantly expressed in prostate cancer, is an attractive target for tumor-specific immunotherapy. To identify human leukocyte antigen (HLA)-A24-restricted epitope peptides from PSMA for further application of the dendritic cell (DC)-based immunotherapy targeting prostate cancer, we have screened several PSMA-encoded HLA-A24-binding peptides for their capabilities to elicit specific antitumor CTL response in vitro. EXPERIMENTAL DESIGN: The amino acid sequence of PSMA was screened for peptides consisting of 9 or 10 amino acids, which possess the known HLA-A24-binding motif. Nine candidate peptides were screened for binding to HLA-A24 molecules. Then, each of these nine peptides was studied to determine whether CTL responses could be induced by primary in vitro immunization of CD8(+) T cells using peptide-pulsed autologous DCs derived from peripheral blood mononuclear cells of HLA-A24(+) healthy donor as antigen-presenting cells. The antigen specificity of the CTL lines was confirmed using several tumor cell lines as target cells, which were genetically modified to express both HLA-A24 and PSMA. RESULTS: Two peptides, LYSDPADYF and NYARTEDFF, were demonstrated to elicit CTL lines that lyse peptide-pulsed, HLA-A24(+) B-lymphoblastoid cells. Each of the CTL lines recognized their specific PSMA-expressing target cells in a HLA-A24-restricted manner. The capability to release IFN-gamma by the CTL lines was specifically inhibited by anti-MHC class I and anti-CD8 monoclonal antibodies but not by anti-MHC class II and anti-CD4 monoclonal antibodies. CONCLUSION: Two novel HLA-A24-restricted PSMA-derived epitopes were identified in this study. These epitopes can be used to further evaluate the clinical utility of DC-based immunotherapeutic strategies for treatment of hormone-refractory prostate cancers.


Asunto(s)
Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Antígenos de Superficie , Carboxipeptidasas/química , Epítopos/farmacología , Antígenos HLA-A/metabolismo , Oligopéptidos/farmacología , Neoplasias de la Próstata/inmunología , Linfocitos T Citotóxicos/inmunología , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Glutamato Carboxipeptidasa II , Antígeno HLA-A24 , Humanos , Inmunoterapia , Interferón gamma/farmacología , Linfoma de Células B/inmunología , Masculino , Neoplasias de la Próstata/terapia , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/inmunología
18.
Cancer Immunol Immunother ; 51(2): 99-106, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11904734

RESUMEN

Carcinoembryonic antigen (CEA), an oncofetal glycoprotein overexpressed in most gastrointestinal and lung cancers, is a candidate molecule for cancer immunotherapy. Recently, a CEA-derived 9-mer peptide, CEA652 (TYACFVSNL), has been identified as the epitope of cytotoxic T lymphocytes restricted with human leukocyte antigen (HLA)-A24, which is present in 60% of the Japanese population and in some Caucasians. The authors performed a clinical study of a vaccine using autologous dendritic cells (DCs) pulsed with CEA652 and adjuvant cytokines, natural human interferon alpha (nhuIFN-alpha), and natural human tumor necrosis factor alpha (nhuTNF-alpha), for the treatment of patients with CEA-expressing advanced metastatic malignancies. Ten HLA-A24 patients with advanced digestive tract or lung cancer were enrolled in the study to assess toxicity, tolerability and immune responses to the vaccine. DCs were generated from plastic adherent monocytes of granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood mononuclear cells (PBMCs) in the presence of granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin 4 (IL-4). Generated DCs showing an immature phenotype were loaded with CEA652 and injected into patients intradermally and subcutaneously with 50% of the dose administered by each route every 2 weeks for a total of ten vaccinations. The total dose of administered DCs ranged from 2.7x10(7)cells to 1.6x10(8)cells. Adjuvant cytokines, i.e., 1x10(6) U/body of nhuIFN-alpha and nhuTNF-alpha, were administered to patients twice a week during the vaccination period. No severe toxicity directly attributable to the treatment was observed, and the vaccine was well tolerated. In the delayed-type hypersensitivity (DTH) skin test, two patients showed a positive skin response to peptide-pulsed DCs after vaccination, although none of the patients tested positive prior to vaccination. In the two patients who showed a positive skin response disease remained stable for 6 and 9 months respectively. These results suggest that active immunization using DCs pulsed with CEA652 peptide in combination with the administration of adjuvant cytokines is a safe and feasible treatment procedure.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Células Dendríticas/inmunología , Neoplasias Gastrointestinales/terapia , Antígenos HLA-A/inmunología , Inmunoterapia/métodos , Neoplasias Pulmonares/terapia , Adulto , Antígeno Carcinoembrionario/análisis , Femenino , Neoplasias Gastrointestinales/inmunología , Factor Estimulante de Colonias de Granulocitos/farmacología , Antígeno HLA-A24 , Humanos , Hipersensibilidad Tardía/etiología , Interferón-alfa/administración & dosificación , Interferón gamma/biosíntesis , Neoplasias Pulmonares/inmunología , Masculino , Persona de Mediana Edad , Linfocitos T Citotóxicos/inmunología , Factor de Necrosis Tumoral alfa/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...