Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 139(17): 2706-2711, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35134127

RESUMEN

Hematopoietic stem cell transplant (HSCT) is a curative option for patients with high-risk acute lymphoblastic leukemia (ALL), but relapse remains a major cause of treatment failure. To prevent disease relapse, we prepared and infused donor-derived multiple leukemia antigen-specific T cells (mLSTs) targeting PRAME, WT1, and survivin, which are leukemia-associated antigens frequently expressed in B- and T-ALL. Our goal was to maximize the graft-versus-leukemia effect while minimizing the risk of graft-versus-host disease (GVHD). We administered mLSTs (dose range, 0.5 × 107 to 2 × 107 cells per square meter) to 11 patients with ALL (8 pediatric, 3 adult), and observed no dose-limiting toxicity, acute GVHD or cytokine release syndrome. Six of 8 evaluable patients remained in long-term complete remission (median: 46.5 months; range, 9-51). In these individuals we detected an increased frequency of tumor-reactive T cells shortly after infusion, with activity against both targeted and nontargeted, known tumor-associated antigens, indicative of in vivo antigen spreading. By contrast, this in vivo amplification was absent in the 2 patients who experienced relapse. In summary, infusion of donor-derived mLSTs after allogeneic HSCT is feasible and safe and may contribute to disease control, as evidenced by in vivo tumor-directed T-cell expansion. Thus, this approach represents a promising strategy for preventing relapse in patients with ALL.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Leucemia , Adulto , Niño , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Leucemia/terapia , Recurrencia , Trasplante Homólogo/efectos adversos
2.
Blood ; 137(19): 2585-2597, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33270816

RESUMEN

Relapse after allogeneic hematopoietic stem cell transplantation (HCT) is the leading cause of death in patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Infusion of unselected donor lymphocytes (DLIs) enhances the graft-versus-leukemia (GVL) effect. However, because the infused lymphocytes are not selected for leukemia specificity, the GVL effect is often accompanied by life-threatening graft-versus-host disease (GVHD), related to the concurrent transfer of alloreactive lymphocytes. Thus, to minimize GVHD and maximize GVL, we selectively activated and expanded stem cell donor-derived T cells reactive to multiple antigens expressed by AML/MDS cells (PRAME, WT1, Survivin, and NY-ESO-1). Products that demonstrated leukemia antigen specificity were generated from 29 HCT donors. In contrast to DLIs, leukemia-specific T cells (mLSTs) selectively recognized and killed leukemia antigen-pulsed cells, with no activity against recipient's normal cells in vitro. We administered escalating doses of mLSTs (0.5 to 10 × 107 cells per square meter) to 25 trial enrollees, 17 with high risk of relapse and 8 with relapsed disease. Infusions were well tolerated with no grade >2 acute or extensive chronic GVHD seen. We observed antileukemia effects in vivo that translated into not-yet-reached median leukemia-free and overall survival at 1.9 years of follow-up and objective responses in the active disease cohort (1 complete response and 1 partial response). In summary, mLSTs are safe and promising for the prevention and treatment of AML/MDS after HCT. This trial is registered at www.clinicaltrials.com as #NCT02494167.


Asunto(s)
Efecto Injerto vs Leucemia , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/terapia , Transfusión de Linfocitos , Síndromes Mielodisplásicos/terapia , Terapia Recuperativa , Linfocitos T/trasplante , Adolescente , Adulto , Anciano , Aloinjertos , Antígenos de Neoplasias/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Terapia Combinada , Femenino , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Transfusión de Linfocitos/efectos adversos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/tratamiento farmacológico , Recurrencia , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T/inmunología , Donantes de Tejidos , Adulto Joven
3.
Mol Ther ; 25(1): 249-258, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28129119

RESUMEN

The adoptive transfer of T cells redirected to tumor-associated antigens via transgenic expression of chimeric antigen receptors (CARs) has produced tumor responses, even in patients with refractory diseases. To target pancreatic cancer, we generated CAR T cells directed against prostate stem cell antigen (PSCA) and demonstrated specific tumor lysis. However, pancreatic tumors employ immune evasion strategies such as the production of inhibitory cytokines, which limit CAR T cell persistence and function. Thus, to protect our cells from the immunosuppressive cytokine IL-4, we generated an inverted cytokine receptor in which the IL-4 receptor exodomain was fused to the IL-7 receptor endodomain (4/7 ICR). Transgenic expression of this molecule in CAR-PSCA T cells should invert the inhibitory effects of tumor-derived IL-4 and instead promote T cell proliferation. We now demonstrate the suppressed activity of CAR T cells in tumor-milieu conditions and the ability of CAR/ICR T cells to thrive in an IL-4-rich microenvironment, resulting in enhanced antitumor activity. Importantly, CAR/ICR T cells remained both antigen and cytokine dependent. These findings support the benefit of combining the 4/7 ICR with CAR-PSCA to treat pancreatic cancer, a PSCA-expressing tumor characterized by a dense immunosuppressive environment rich in IL-4.


Asunto(s)
Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Microambiente Tumoral/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Interleucina-4/metabolismo , Interleucina-4/farmacología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/efectos de los fármacos , Ratones , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética
4.
Immunol Rev ; 257(1): 39-55, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24329788

RESUMEN

The adoptive transfer of T cells specific for native tumor antigens (TAs) is an increasingly popular cancer treatment option because of the ability of these cells to discriminate between normal and tumor tissues and the corresponding lack of short or long-term toxicities. Infusions of antigen-specific CD4(+) and CD8(+) T cells targeting viral antigens derived from Epstein-Barr virus (EBV) induce sustained complete tumor remissions in patients with highly immunogenic tumors such as post-transplant lymphoproliferative disease, although resistance occurred when the infused T-cell population had restricted antigen specificity. T cells specific for EBV antigens have also produced complete remissions of EBV-positive nasopharyngeal carcinomas and lymphomas developing in immunocompetent individuals, even though in these patients tumor survival is dependent on their ability to evade T-cell immunity. Adapting this strategy to non-viral tumors is more challenging, as the target antigens expressed are less immunogenic and the tumors lack the potent danger signals that are characteristic of viruses. The goals of current studies are to define conditions that promote expansion of antigen-specific T cells ex vivo and to ensure their in vivo persistence and survival by combining with maneuvers such as lymphodepletion, checkpoint inhibition, cytokine infusions, or genetic manipulations. More pragmatic goals are to streamline manufacturing to facilitate the transition of these therapies to late phase trials and to evaluate closely histocompatibility antigen (HLA)-matched banked antigen-specific T cells so that T-cell therapies can be made more broadly available.


Asunto(s)
Inmunoterapia Adoptiva , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Antígenos Virales/inmunología , Técnicas de Cultivo de Célula , Epítopos de Linfocito T/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Herpesvirus Humano 4/inmunología , Humanos , Inmunoterapia Adoptiva/métodos , Leucemia/inmunología , Leucemia/patología , Leucemia/terapia , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfoma/inmunología , Linfoma/terapia , Linfoma/virología , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/terapia , Trastornos Linfoproliferativos/virología , Melanoma/inmunología , Melanoma/terapia , Subgrupos de Linfocitos T/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
5.
J Infect Dis ; 216(2): 153-161, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28472480

RESUMEN

Parainfluenza virus type 3 (PIV3) infections are a major cause of morbidity and mortality in immunocompromised individuals, with no approved therapies. Our group has demonstrated the safety and efficacy of adoptively transferred virus-specific T cells for the prevention and treatment of a broad range of viral infections including BK virus, cytomegalovirus, adenovirus, human herpesvirus 6, and Epstein-Barr virus. However, this approach is restricted to well-characterized viruses with known immunogenic/protective T-cell target antigens, precluding extension to PIV3. We now characterize the cellular immune response to all 7 PIV3-encoded antigens in 17 healthy donors and define a hierarchy of immunogenicity based on the frequency of responding donors and the magnitude of specific cells. We show that reactive populations of both CD4+ and CD8+ T cells are capable of producing Th1-polarized effector cytokines and killing PIV3-expressing targets. Furthermore, we confirm the clinical relevance of these cells by demonstrating a direct correlation between the presence of PIV3-specific T cells and viral control in allogeneic hematopoietic stem cell transplant recipients. Taken together, our findings support the clinical use of PIV3-specific T cells produced with our Good Manufacturing Practice-compliant manufacturing process, in immunocompromised patients with uncontrolled infections.


Asunto(s)
Antígenos Virales/inmunología , Inmunidad Celular , Leucocitos Mononucleares/virología , Virus de la Parainfluenza 3 Humana , Infecciones por Respirovirus/inmunología , Linfocitos T/inmunología , Preescolar , Citocinas/inmunología , Femenino , Humanos , Inmunoterapia , Lactante , Masculino , Persona de Mediana Edad
6.
J Infect Dis ; 216(6): 678-687, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28934427

RESUMEN

Human metapneumovirus (hMPV) is a respiratory virus detected in ≥9% of allogeneic hematopoietic stem cell transplant (HSCT) recipients, in whom it can cause significant morbidity and mortality. Given the lack of effective antivirals, we investigated the potential for immunotherapeutic intervention, using adoptively transferred T cells. Thus, we characterized the cellular immune response to the virus and identified F, N, M2-1, M, and P as immunodominant target antigens. Reactive T cells were polyclonal (ie, they expressed CD4 and CD8), T-helper type 1 polarized, and polyfunctional (ie, they produced interferon γ, tumor necrosis factor α, granulocyte-macrophage colony-stimulating factor, and granzyme B), and they were able to kill autologous antigen-loaded targets. The detection of hMPV-specific T cells in HSCT recipients who endogenously controlled active infections support the clinical importance of T-cell immunity in mediating protective antiviral effects. Our results demonstrate the feasibility of developing an immunotherapy for immunocompromised patients with uncontrolled infections.


Asunto(s)
Inmunoterapia Adoptiva , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/terapia , Adulto , Estudios de Factibilidad , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Granzimas/inmunología , Humanos , Inmunidad Celular , Huésped Inmunocomprometido/inmunología , Epítopos Inmunodominantes/inmunología , Interferón gamma/inmunología , Leucocitos Mononucleares/virología , Masculino , Metapneumovirus/aislamiento & purificación , Persona de Mediana Edad , Infecciones por Paramyxoviridae/inmunología , Linfocitos T/virología , Factor de Necrosis Tumoral alfa/inmunología , Adulto Joven
8.
Mol Ther ; 22(3): 623-633, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24213558

RESUMEN

The use of chimeric antigen receptor (CAR)-modified T cells as a therapy for hematologic malignancies and solid tumors is becoming more widespread. However, the infusion of a T-cell product targeting a single tumor-associated antigen may lead to target antigen modulation under this selective pressure, with subsequent tumor immune escape. With the purpose of preventing this phenomenon, we have studied the impact of simultaneously targeting two distinct antigens present on tumor cells: namely mucin 1 and prostate stem cell antigen, both of which are expressed in a variety of solid tumors, including pancreatic and prostate cancer. When used individually, CAR T cells directed against either tumor antigen were able to kill target-expressing cancer cells, but tumor heterogeneity led to immune escape. As a combination therapy, we demonstrate superior antitumor effects using both CARs simultaneously, but this was nevertheless insufficient to achieve a complete response. To understand the mechanism of escape, we studied the kinetics of T-cell killing and found that the magnitude of tumor destruction depended not only on the presence of target antigens but also on the intensity of expression-a feature that could be altered by administering epigenetic modulators that upregulated target expression and enhanced CAR T-cell potency.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Mucina-1/metabolismo , Neoplasias de la Próstata/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Células HEK293 , Humanos , Activación de Linfocitos/inmunología , Masculino , Ratones , Proteínas Recombinantes de Fusión/metabolismo
9.
Mol Ther ; 22(6): 1211-1220, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24732709

RESUMEN

The success of adoptively transferred tumor-directed T cells requires them to survive and expand in vivo. Most tumors, however, employ immune evasion mechanisms, including the production of inhibitory cytokines that limit in vivo T-cell persistence and effector function. To protect tumor-directed T cells from such negative influences, we generated a chimeric cytokine receptor in which the interleukin (IL) 4 receptor exodomain was fused to the IL7 receptor endodomain. We thereby inverted the effects of tumor-derived IL4 so that the proliferation and activation of tumor directed cytotoxic T cells was enhanced rather than inhibited in the tumor microenvironment, resulting in superior antitumor activity. These transgenic T cells were only activated in the tumor environment since triggering required exposure to both tumor antigen (signal 1) and tumor-derived IL4 (signal 2). This selectivity supports future clinical adaptation.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-4/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Receptores de Interleucina-7/genética , Linfocitos T/inmunología , Microambiente Tumoral , Traslado Adoptivo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Subunidad alfa del Receptor de Interleucina-4/inmunología , Activación de Linfocitos , Ratones SCID , Receptores de Interleucina-7/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/trasplante , Transgenes/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cytotherapy ; 16(6): 713-33, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24239105

RESUMEN

Adoptively transferred T cells have the capacity to traffic to distant tumor sites, infiltrate fibrotic tissue and kill antigen-expressing tumor cells. Various groups have investigated different genetic engineering strategies designed to enhance tumor specificity, increase T cell potency, improve proliferation, persistence or migratory capacity and increase safety. This review focuses on recent developments in T cell engineering, discusses the clinical application of these engineered cell products and outlines future prospects for this therapeutic modality.


Asunto(s)
Ingeniería Celular , Neoplasias/terapia , Proteínas Recombinantes de Fusión/genética , Linfocitos T/inmunología , Antígenos de Neoplasias/biosíntesis , Tratamiento Basado en Trasplante de Células y Tejidos , Citotoxicidad Inmunológica , Humanos , Inmunoterapia Adoptiva , Neoplasias/inmunología , Neoplasias/patología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T/citología
11.
Lancet Oncol ; 14(11): e447-e456, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24079872

RESUMEN

Gene therapy as a treatment for cancer is regarded as high in promise, but low in delivery, a deficiency that has become more obvious with ever-increasing reports of the successful correction of monogenic disorders by this approach. We review the commercial and scientific obstacles that have led to these delays and describe how they are progressively being overcome. Recent and striking successes and correspondingly increased commercial involvement suggest that gene transfer could finally become a powerful method for development of safe and effective cancer therapeutic drugs.


Asunto(s)
Genes Supresores de Tumor , Terapia Genética , Neoplasias/terapia , Animales , Humanos , Neoplasias/genética
12.
Haematologica ; 98(4): 533-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23242592

RESUMEN

The low frequency of naturally occurring regulatory T cells (nTregs) in peripheral blood and the suboptimal protocols available for their ex vivo expansion limit the development of clinical trials based on the adoptive transfer of these cells. We have, therefore, generated a simplified, robust and cost-effective platform for the large-scale expansion of nTregs using a gas permeable static culture flask (G-Rex) in compliance with Good Manufacturing Practice. More than 10(9) putative Tregs co-expressing CD25 and CD4 molecules (92 ± 5%) and FoxP3 (69 ± 19%) were obtained within 21 days of culture. Expanded Tregs showed potent regulatory activity in vitro (80 ± 13% inhibition of CD8(+) cell division) and in vivo (suppression or delay of graft-versus-host disease in a xenograft mouse model) indicating that the cost-effective and simplified production of nTregs we propose will facilitate the implementation of clinical trials based on their adoptive transfer.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Enfermedad Injerto contra Huésped/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Antígenos CD2/inmunología , Antígenos CD2/metabolismo , Técnicas de Cultivo de Célula/economía , Técnicas de Cultivo de Célula/instrumentación , Análisis Costo-Beneficio , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/terapia , Humanos , Inmunofenotipificación , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Trasplante Heterólogo
13.
Cytotherapy ; 14(1): 7-11, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22172091

RESUMEN

Infections with a range of common community viruses remain a major cause of mortality and morbidity after allogeneic hematopoietic stem cell transplantation. T cells specific for cytomegalovirus (CMV), Epstein-Barr virus (EBV) and adenoviruses can safely prevent and infections with these three most common culprits, but the manufacture of individual T cell lines for each virus would be prohibitive in terms of time and cost. We have demonstrated that T cells specific for all three viruses can be manufactured in a single culture using monocytes and EBV-transformed B lymphoblastoid cell lines (LCLs), both transduced with an adenovirus vector expressing pp65 of CMV, as antigen-presenting cells. Trivirus-specific T cell lines produced from healthy stem cell donors could prevent and treat infections with all three viruses, not only in the designated recipient, but in unrelated, partially-HLA-matched third party recipients. We now provide the details and logistics of T cell manufacture.


Asunto(s)
Adenoviridae/inmunología , Técnicas de Cultivo de Célula , Citomegalovirus/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Herpesvirus Humano 4/inmunología , Linfocitos T Citotóxicos/inmunología , Virosis/etiología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/trasplante , Línea Celular Transformada , Proliferación Celular , Citotoxicidad Inmunológica , Antígenos HLA/inmunología , Humanos , Fosfoproteínas/inmunología , Linfocitos T Citotóxicos/trasplante , Trasplante Homólogo/efectos adversos , Proteínas de la Matriz Viral/inmunología , Virosis/terapia
14.
Mol Ther ; 19(12): 2258-68, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21915103

RESUMEN

Although immunotherapy with Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes (CTLs) can treat EBV-associated Hodgkin and non-Hodgkin lymphoma (HL/NHL), more than 50% of such tumors are EBV negative. We now describe an approach that allows us to consistently generate, in a single line, CTLs that recognize a wide spectrum of nonviral tumor-associated antigens (TAAs) expressed by human HL/NHL, including Survivin, MAGE-A4, Synovial sarcoma X (SSX2), preferentially expressed antigen in melanoma (PRAME) and NY-ESO-1. We could generate these CTLs from nine of nine healthy donors and five of eight lymphoma patients, irrespective of human leukocyte antigen (HLA) type. We reactivated TAA-directed T cells ex vivo, by stimulation with dendritic cells (DCs) pulsed with overlapping peptide libraries spanning the chosen antigens in the presence of an optimized Th1-polarizing, prosurvival/proliferative and Treg inhibitory cytokine combination. The resultant lines of CD4(+) and CD8(+), polycytokine-producing T cells are directed against a multiplicity of epitopes expressed on the selected TAAs, with cytolytic activity against autologous tumor cells. Infusion of such multispecific monocultures may extend the benefits of CTL therapy to treatment even of EBV negative HL and NHL.


Asunto(s)
Antígenos de Neoplasias/inmunología , Epítopos/inmunología , Inmunoterapia , Linfoma/inmunología , Linfoma/terapia , Linfocitos T Citotóxicos/inmunología , Adulto , Antígenos de Neoplasias/genética , Células Cultivadas , Técnicas de Cocultivo , Infecciones por Virus de Epstein-Barr , Citometría de Flujo , Herpesvirus Humano 4 , Humanos , Técnicas para Inmunoenzimas , Activación de Linfocitos , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Fragmentos de Péptidos/inmunología , Biblioteca de Péptidos
15.
Ther Adv Med Oncol ; 14: 17588359221107113, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35860837

RESUMEN

Purpose: Adoptively transferred, ex vivo expanded multi-antigen-targeted T cells (multiTAA-T) represent a new, potentially effective, and nontoxic therapeutic approach for patients with breast cancer (BC). In this first-in-human trial, we investigated the safety and clinical effects of administering multiTAA T cells targeting the tumor-expressed antigens, Survivin, NY-ESO-1, MAGE-A4, SSX2, and PRAME, to patients with relapsed/refractory/metastatic BC. Materials and methods: MultiTAA T-cell products were generated from the peripheral blood of heavily pre-treated patients with metastatic or locally recurrent unresectable BC of all subtypes and infused at a fixed dose level of 2 × 107/m2. Patients received two infusions of cells 4 weeks apart and safety and clinical activity were determined. Cells were administered in an outpatient setting and without prior lymphodepleting chemotherapy. Results: All patients had estrogen receptor/progesterone receptor positive BC, with one patient also having human epidermal growth factor receptor 2-positive. There were no treatment-related toxicities and the infusions were well tolerated. Of the 10 heavily pre-treated patients enrolled and infused with multiTAA T cells, nine had disease progression while one patient with 10 lines of prior therapies experienced prolonged (5 months) disease stabilization that was associated with the in vivo expansion and persistence of T cells directed against the targeted antigens. Furthermore, antigen spreading and the endogenous activation of T cells directed against a spectrum of non-targeted tumor antigens were observed in 7/10 patients post-multiTAA infusion. Conclusion: MultiTAA T cells were well tolerated and induced disease stabilization in a patient with refractory BC. This was associated with in vivo T-cell expansion, persistence, and antigen spreading. Future directions of this approach may include additional strategies to enhance the therapeutic benefit of multiTAA T cells in patients with BC.

16.
HPB (Oxford) ; 13(9): 643-50, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21843265

RESUMEN

OBJECTIVE: Conventional chemotherapy and radiotherapy produce marginal survival benefits in pancreatic cancer, underscoring the need for novel therapies. The aim of this study is to develop an adoptive T cell transfer approach to target tumours expressing prostate stem cell antigen (PSCA), a tumour-associated antigen that is frequently expressed by pancreatic cancer cells. METHODS: Expression of PSCA on cell lines and primary tumour samples was confirmed by immunohistochemistry. Healthy donor- and patient-derived T cells were isolated, activated in vitro using CD3/CD28, and transduced with a retroviral vector encoding a chimeric antigen receptor (CAR) targeting PSCA. The ability of these cells to kill tumour cells was analysed by chromium-51 (Cr(51)) release. RESULTS: Prostate stem cell antigen was expressed on >70% of the primary tumour samples screened. Activated, CAR-modified T cells could be readily generated in clinically relevant numbers and were specifically able to kill PSCA-expressing pancreatic cancer cell lines with no non-specific killing of PSCA-negative target cells, thus indicating the potential efficacy and safety of this approach. CONCLUSIONS: Prostate stem cell antigen is frequently expressed on pancreatic cancer cells and can be targeted for immune-mediated destruction using CAR-modified, adoptively transferred T cells. The safety and efficacy of this approach indicate that it deserves further study and may represent a promising novel treatment for patients with pancreatic cancer.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Carcinoma Ductal Pancreático/terapia , Terapia Genética , Inmunoterapia Adoptiva , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/terapia , Receptores de Antígenos de Linfocitos T/genética , Anticuerpos de Cadena Única/genética , Linfocitos T/trasplante , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Estudios de Factibilidad , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Activación de Linfocitos , Muromonab-CD3/farmacología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Receptores de Antígenos de Linfocitos T/biosíntesis , Anticuerpos de Cadena Única/biosíntesis , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factores de Tiempo , Transducción Genética , Regulación hacia Arriba
17.
J Clin Oncol ; 39(13): 1415-1425, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33507803

RESUMEN

PURPOSE: Patients with relapsed lymphomas often fail salvage therapies including high-dose chemotherapy and mono-antigen-specific T-cell therapies, highlighting the need for nontoxic, novel treatments. To that end, we clinically tested an autologous T-cell product that targets multiple tumor-associated antigens (TAAs) expressed by lymphomas with the intent of treating disease and preventing immune escape. PATIENTS AND METHODS: We expanded polyclonal T cells reactive to five TAAs: PRAME, SSX2, MAGEA4, SURVIVIN, and NY-ESO-1. Products were administered to 32 patients with Hodgkin lymphomas (n = 14) or non-Hodgkin lymphomas (n = 18) in a two-part phase I clinical trial, where the objective of the first phase was to establish the safety of targeting all five TAAs (fixed dose, 0.5 × 107 cells/m2) simultaneously and the second stage was to establish the maximum tolerated dose. Patients had received a median of three prior lines of therapy and either were at high risk for relapse (adjuvant arm, n = 17) or had chemorefractory disease (n = 15) at enrollment. RESULTS: Infusions were safe with no dose-limiting toxicities observed in either the antigen- or dose-escalation phases. Although the maximum tolerated dose was not reached, the maximum tested dose at which efficacy was observed (two infusions, 2 × 107 cells/m2) was determined as the recommended phase II dose. Of the patients with chemorefractory lymphomas, two (of seven) with Hodgkin lymphomas and four (of eight) with non-Hodgkin lymphomas achieved durable complete remissions (> 3 years). CONCLUSION: T cells targeting five TAAs and administered at doses of up to two infusions of 2 × 107 cells/m2 are well-tolerated by patients with lymphoma both as adjuvant and to treat chemorefractory lymphoma. Preliminary indicators of antilymphoma activity were seen in the chemorefractory cohort across both antigen- and dose-escalation phases.


Asunto(s)
Antígenos de Neoplasias/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Linfoma/terapia , Terapia Recuperativa , Linfocitos T/trasplante , Adolescente , Adulto , Anciano , Femenino , Humanos , Linfoma/inmunología , Masculino , Persona de Mediana Edad , Pronóstico , Adulto Joven
18.
Mol Ther ; 17(9): 1616-25, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19584818

RESUMEN

Viral infections cause morbidity and mortality in allogeneic hematopoietic stem cell transplant (HSCT) recipients. To prevent and treat these, we have produced and infused cytotoxic T lymphocytes (CTLs) with specificity for Epstein-Barr virus (EBV), cytomegalovirus (CMV), and adenovirus (Adv), and shown that small numbers of infused cells proliferate in vivo and protect against all three viruses. Despite these encouraging results, broader implementation of this approach is limited by the need for infectious virus material (EBV), expensive production of clinical grade adenoviral vectors, and a prolonged (8-12 weeks) period of manufacture. There is also competition between virus-derived antigens within antigen-presenting cells (APCs), limiting extension to additional agents. We now describe an approach that uses DNA nucleofection of dendritic cells (DCs) with DNA plasmids that encode a range of immunodominant and subdominant viral antigens from CMV, EBV, BK, and Adv. Within 10 days, this methodology provides multivirus-reactive CTLs that lack alloreactivity. We further demonstrate that nucleofected DC stimulation can be combined with interferon-gamma (IFN-gamma) capture technology to produce even more rapid multivirus-CTL products for treatment of acute infection. These CTL generation procedures should increase the feasibility and applicability of T-cell therapy.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Huésped Inmunocomprometido , Inmunoterapia/métodos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Adenoviridae/genética , Adenoviridae/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Células Cultivadas , Citomegalovirus/genética , Citomegalovirus/inmunología , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/inmunología , Humanos , Leucocitos Mononucleares
19.
Mol Ther ; 17(5): 880-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19259067

RESUMEN

Adoptive transfer of antigen-specific cytotoxic T lymphocytes (CTLs) can induce objective clinical responses in patients with malignant diseases. The option of providing a proliferative and survival advantage to adoptively transferred CTLs remains a challenge to improve their efficacy. Host lymphodepletion and administration of recombinant interleukin-2 (IL-2) are currently used to improve CTL survival and expansion after adoptive transfer, but these approaches are frequently associated with significant side effects and may increase proliferation of T regulatory cells. IL-7 is a crucial homeostatic cytokine that has been safely administered as a recombinant protein. However, while IL-7 induces robust expansion of naive and memory T lymphocytes, the lack of expression of the IL-7 receptor alpha chain (IL-7Ralpha) by CTLs precludes their response to this cytokine. We found that CTLs can be genetically modified to re-express IL-7Ralpha, and that this manipulation restores the response of these cells to IL-7 without apparent modification of their antigen specificity or dependency, and without changing their response to other common gamma (gammac) chain cytokines. This approach may allow selective expansion of CTLs without the unwanted effects associated with IL-2.


Asunto(s)
Interleucina-7/inmunología , Receptores de Interleucina-7/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Animales , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-2/inmunología , Ratones , Ratones SCID , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-7/genética
20.
Sci Transl Med ; 12(554)2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32727914

RESUMEN

Multiple myeloma (MM) is an almost always incurable malignancy of plasma cells. Despite the advent of new therapies, most patients eventually relapse or become treatment-refractory. Consequently, therapies with nonoverlapping mechanisms of action that are nontoxic and provide long-term benefit to patients with MM are greatly needed. To this end, we clinically tested an autologous multitumor-associated antigen (mTAA)-specific T cell product for the treatment of patients with high-risk, relapsed or refractory MM. In this study, we expanded polyclonal T cells from 23 patients with MM. T cells whose native T cell receptors were reactive toward five myeloma-expressed target TAAs (PRAME, SSX2, MAGEA4, Survivin, and NY-ESO-1) were enriched ex vivo. To date, we have administered escalating doses of these nonengineered mTAA-specific T cells (0.5 × 107 to 2 × 107 cells/m2) to 21 patients with MM, 9 of whom were at high risk of relapse after a median of 3 lines of prior therapy and 12 with active, relapsed or refractory disease after a median of 3.5 prior lines. The cells were well tolerated, with only two transient, grade III infusion-related adverse events. Furthermore, patients with active relapsed or refractory myeloma enjoyed a longer than expected progression-free survival and responders included three patients who achieved objective responses concomitant with detection of functional TAA-reactive T cell clonotypes derived from the infused mTAA product.


Asunto(s)
Mieloma Múltiple , Antígenos de Neoplasias , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Mieloma Múltiple/terapia , Recurrencia Local de Neoplasia , Receptores de Antígenos de Linfocitos T
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA