Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 14(8)2022 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-35454906

RESUMEN

The hostile tumor microenvironment (TME) is a major challenge for the treatment of solid tumors with T-cell receptor (TCR)-modified T-cells (TCR-Ts), as it negatively influences T-cell efficacy, fitness, and persistence. These negative influences are caused, among others, by the inhibitory checkpoint PD-1/PD-L1 axis. The Preferentially Expressed Antigen in Melanoma (PRAME) is a highly relevant cancer/testis antigen for TCR-T immunotherapy due to broad expression in multiple solid cancer indications. A TCR with high specificity and sensitivity for PRAME was isolated from non-tolerized T-cell repertoires and introduced into T-cells alongside a chimeric PD1-41BB receptor, consisting of the natural extracellular domain of PD-1 and the intracellular signaling domain of 4-1BB, turning an inhibitory pathway into a T-cell co-stimulatory pathway. The addition of PD1-41BB to CD8+ T-cells expressing the transgenic PRAME-TCR enhanced IFN-γ secretion, improved cytotoxic capacity, and prevented exhaustion upon repetitive re-challenge with tumor cells in vitro without altering the in vitro safety profile. Furthermore, a single dose of TCR-Ts co-expressing PD1-41BB was sufficient to clear a hard-to-treat melanoma xenograft in a mouse model, whereas TCR-Ts without PD1-41BB could not eradicate the PD-L1-positive tumors. This cutting-edge strategy supports development efforts to provide more effective TCR-T immunotherapies for the treatment of solid tumors.

2.
Hum Gene Ther ; 28(12): 1158-1168, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28950731

RESUMEN

T-cell receptor (TCR) immunotherapy uses T cells engineered with new TCRs to enable detection and killing of cancer cells. Efficacy of TCR immunotherapy depends on targeting antigenic peptides that are efficiently presented by the best-suited major histocompatibility complex (MHC) molecules of cancer cells. However, efficient strategies are lacking to easily identify TCRs recognizing immunodominant peptide-MHC (pMHC) combinations utilizing any of the six possible MHC class I alleles of a cancer cell. We generated an MHC cell library and developed a platform approach to detect, isolate, and re-express TCRs specific for immunodominant pMHCs. The platform approach was applied to identify a human papillomavirus (HPV16) oncogene E5-specific TCR, recognizing a novel, naturally processed pMHC (HLA-B*15:01) and a cytomegalovirus-specific TCR targeting an immunodominant pMHC (HLA-B*07:02). The platform provides a useful tool to isolate in an unbiased manner TCRs specific for novel and immunodominant pMHC targets for use in TCR immunotherapy.


Asunto(s)
Traslado Adoptivo/métodos , Antígeno HLA-B15 , Antígeno HLA-B7 , Neoplasias , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T , Linfocitos T/inmunología , Antígeno HLA-B15/genética , Antígeno HLA-B15/inmunología , Antígeno HLA-B7/genética , Antígeno HLA-B7/inmunología , Humanos , Células K562 , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Péptidos/genética , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
3.
Cancer Res ; 77(13): 3577-3590, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28533272

RESUMEN

Inherent intermediate- to low-affinity T-cell receptors (TCR) that develop during the natural course of immune responses may not allow sufficient activation for tumor elimination, making the majority of T cells suboptimal for adoptive T-cell therapy (ATT). TCR affinity enhancement has been implemented to provide stronger T-cell activity but carries the risk of creating undesired cross-reactivity leading to potential serious adverse effects in clinical application. We demonstrate here that engineering of low-avidity T cells recognizing a naturally processed and presented tumor-associated antigen with a chimeric PD-1:28 receptor increases effector function to levels seen with high-avidity T cells of identical specificity. Upgrading the function of low-avidity T cells without changing the TCR affinity will allow a large arsenal of low-avidity T cells previously thought to be therapeutically inefficient to be considered for ATT. PD-1:28 engineering reinstated Th1 function in tumor-infiltrating lymphocytes that had been functionally disabled in the human renal cell carcinoma environment without unleashing undesired Th2 cytokines or IL10. Involved mechanisms may be correlated to restoration of ERK and AKT signaling pathways. In mouse tumor models of ATT, PD-1:28 engineering enabled low-avidity T cells to proliferate stronger and prevented PD-L1 upregulation and Th2 polarization in the tumor milieu. Engineered T cells combined with checkpoint blockade secreted significantly more IFNγ compared with T cells without PD-1:28, suggesting a beneficial combination with checkpoint blockade therapy or other therapeutic strategies. Altogether, the supportive effects of PD-1:28 engineering on T-cell function make it an attractive tool for ATT. Cancer Res; 77(13); 3577-90. ©2017 AACR.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Neoplasias/inmunología , Ingeniería de Proteínas , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Gastroenterology ; 149(4): 1042-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26052074

RESUMEN

BACKGROUND & AIMS: Cancer therapies are being developed based on our ability to direct T cells against tumor antigens. Glypican-3 (GPC3) is expressed by 75% of all hepatocellular carcinomas (HCC), but not in healthy liver tissue or other organs. We aimed to generate T cells with GPC3-specific receptors that recognize HCC and used them to eliminate GPC3-expressing xenograft tumors grown from human HCC cells in mice. METHODS: We used mass spectrometry to obtain a comprehensive peptidome from GPC3-expressing hepatoma cells after immune-affinity purification of human leukocyte antigen (HLA)-A2 and bioinformatics to identify immunodominant peptides. To circumvent GPC3 tolerance resulting from fetal expression, dendritic cells from HLA-A2-negative donors were cotransfected with GPC3 and HLA-A2 RNA to stimulate and expand antigen-specific T cells. RESULTS: Peptide GPC3367 was identified as a predominant peptide on HLA-A2. We used A2-GPC3367 multimers to detect, select for, and clone GPC3-specific T cells. These clones bound the A2-GPC3367 multimer and secreted interferon-γ when cultured with GPC3367, but not with control peptide-loaded cells. By genomic sequencing of these T-cell clones, we identified a gene encoding a dominant T-cell receptor. The gene was cloned and the sequence was codon optimized and expressed from a retroviral vector. Primary CD8(+) T cells that expressed the transgenic T-cell receptor specifically bound GPC3367 on HLA-A2. These T cells killed GPC3-expressing hepatoma cells in culture and slowed growth of HCC xenograft tumors in mice. CONCLUSIONS: We identified a GPC3367-specific T-cell receptor. Expression of this receptor by T cells allows them to recognize and kill GPC3-positive hepatoma cells. This finding could be used to advance development of adoptive T-cell therapy for HCC.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Carcinoma Hepatocelular/terapia , Citotoxicidad Inmunológica , Células Dendríticas/metabolismo , Genes Codificadores de los Receptores de Linfocitos T , Ingeniería Genética/métodos , Glipicanos/metabolismo , Antígeno HLA-A2/metabolismo , Inmunoterapia Adoptiva/métodos , Neoplasias Hepáticas/terapia , Activación de Linfocitos , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Supervivencia Celular , Técnicas de Cocultivo , Células Dendríticas/inmunología , Femenino , Glipicanos/genética , Glipicanos/inmunología , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Células Hep G2 , Humanos , Epítopos Inmunodominantes , Interferón gamma/inmunología , Interferón gamma/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones SCID , Factores de Tiempo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
5.
PLoS One ; 10(3): e0121633, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25799237

RESUMEN

Codon optimization of nucleotide sequences is a widely used method to achieve high levels of transgene expression for basic and clinical research. Until now, immunological side effects have not been described. To trigger T cell responses against human papillomavirus, we incubated T cells with dendritic cells that were pulsed with RNA encoding the codon-optimized E7 oncogene. All T cell receptors isolated from responding T cell clones recognized target cells expressing the codon-optimized E7 gene but not the wild type E7 sequence. Epitope mapping revealed recognition of a cryptic epitope from the +3 alternative reading frame of codon-optimized E7, which is not encoded by the wild type E7 sequence. The introduction of a stop codon into the +3 alternative reading frame protected the transgene product from recognition by T cell receptor gene-modified T cells. This is the first experimental study demonstrating that codon optimization can render a transgene artificially immunogenic through generation of a dominant cryptic epitope. This finding may be of great importance for the clinical field of gene therapy to avoid rejection of gene-corrected cells and for the design of DNA- and RNA-based vaccines, where codon optimization may artificially add a strong immunogenic component to the vaccine.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Codón , Papillomaviridae/inmunología , Proteínas E7 de Papillomavirus/inmunología , Células Cultivadas , Mapeo Epitopo , Terapia Genética/métodos , Humanos , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/genética
6.
Eur J Immunol ; 44(9): 2811-21, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24846220

RESUMEN

Immunity to tumor differentiation antigens, such as melanoma antigen recognized by T cells 1 (MART-1), has been comprehensively studied. Intriguingly, CD8(+) T cells specific for the MART-1(26(27)-35) epitope in the context of HLA-A0201 are about 100 times more abundant compared with T cells specific for other tumor-associated antigens. Moreover, MART-1-specific CD8(+) T cells show a highly biased usage of the Vα-region gene TRAV12-2. Here, we provide independent support for this notion, by showing that the combinatorial pairing of different TCRα- and TCRß- chains derived from HLA-A2-MART-1(26-35) -specific CD8(+) T-cell clones is unusually permissive in conferring MART-1 specificity, provided the CDR1α TRAV12-2 region is used. Whether TCR bias alone accounts for the unusual abundance of HLA-A2-MART-1(26-35) -specific CD8(+) T cells has remained conjectural. Here, we provide an alternative explanation: misinitiated transcription of the MART-1 gene resulting in truncated mRNA isoforms leads to lack of promiscuous transcription of the MART-1(26-35) epitope in human medullary thymic epithelial cells and, consequently, evasion of central self-tolerance toward this epitope. Thus, biased TCR usage and leaky central tolerance might act in an independent and additive manner to confer high frequency of MART-1(26-35) -specific CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Epiteliales/inmunología , Epítopos de Linfocito T/inmunología , Antígeno MART-1/inmunología , Timo/inmunología , Iniciación de la Transcripción Genética/inmunología , Linfocitos T CD8-positivos/citología , Línea Celular , Células Epiteliales/citología , Femenino , Antígeno HLA-A2/inmunología , Humanos , Masculino , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timo/citología
7.
J Immunother ; 36(2): 133-51, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23377668

RESUMEN

Nine cancer patients were treated with adoptive cell therapy using autologous anti-MAGE-A3 T-cell receptors (TCR)-engineered T cells. Five patients experienced clinical regression of their cancers including 2 on-going responders. Beginning 1-2 days postinfusion, 3 patients (#'s 5, 7, and 8) experienced mental status changes, and 2 patients (5 and 8) lapsed into comas and subsequently died. Magnetic resonance imagining analysis of patients 5 and 8 demonstrated periventricular leukomalacia, and examination of their brains at autopsy revealed necrotizing leukoencephalopathy with extensive white matter defects associated with infiltration of CD3(+)/CD8(+) T cells. Patient 7, developed Parkinson-like symptoms, which resolved over 4 weeks and fully recovered. Immunohistochemical staining of patient and normal brain samples demonstrated rare positively staining neurons with an antibody that recognizes multiple MAGE-A family members. The TCR used in this study recognized epitopes in MAGE-A3/A9/A12. Molecular assays of human brain samples using real-time quantitative-polymerase chain reaction, Nanostring quantitation, and deep-sequencing indicated that MAGE-A12 was expressed in human brain (and possibly MAGE-A1, MAGE-A8, and MAGE-A9). This previously unrecognized expression of MAGE-A12 in human brain was possibly the initiating event of a TCR-mediated inflammatory response that resulted in neuronal cell destruction and raises caution for clinical applications targeting MAGE-A family members with highly active immunotherapies.


Asunto(s)
Antígenos de Neoplasias , Terapia Genética/métodos , Inmunoterapia Adoptiva , Melanoma/terapia , Proteínas de Neoplasias , Receptores de Antígenos de Linfocitos T , Adulto , Anciano , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/uso terapéutico , Encéfalo/metabolismo , Células Dendríticas/inmunología , Femenino , Técnicas de Transferencia de Gen , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/mortalidad , Masculino , Melanoma/inmunología , Antígenos Específicos del Melanoma , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/uso terapéutico , ARN Mensajero/análisis , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/uso terapéutico , Vacunas de Subunidad/inmunología , Adulto Joven
8.
Oncoimmunology ; 1(9): 1643-1644, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23264922

RESUMEN

The adoptive transfer of lymphocytes expressing high-avidity T-cell receptors with antitumor specificity provides a promising therapy for cancer patients. Recently, we compared 12 HLA-A2-restricted, tyrosinase peptide-specific CD8(+) cytotoxic T-lymphocyte (CTL) clones and demonstrated that polyfunctional type 1 helper (Th1)-cytokine secretion serves to rapidly select high-quality, high-avidity CTLs.

9.
Oncoimmunology ; 1(2): 129-140, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22720234

RESUMEN

Designer T cells expressing transgenic T cell receptors (TCR) with anti-tumor specificity offer new treatment options for cancer patients. We developed a three phase procedure to identify T cells of high avidity based on the fact that T cells recognizing peptides presented by allogeneic MHC efficiently kill tumor cells. Autologous dendritic cells (DC) are co-transfected with ivt-RNA encoding an allogeneic MHC molecule and a selected antigen to allow them to express allogeneic MHC-peptide complexes that activate allo-restricted peptide-specific T cells. This approach provides great flexibility for obtaining high-avidity T cells as potential sources of TCR for adoptive T cell therapy.

10.
J Immunol ; 189(2): 598-605, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22689880

RESUMEN

Adoptive transfer of T cells expressing transgenic TCR with antitumor specificity provides a hopeful new therapy for patients with advanced cancer. To fulfill a large need for TCR with high affinity and specificity for various tumor entities, we sought to identify parameters for rapid selection of CTL clones with suitable characteristics. Twelve CTL clones displaying different Ag sensitivities for the same peptide-MHC epitope of the melanoma-associated Ag tyrosinase were analyzed in detail. Better MHC-multimer binding and slower multimer release are thought to reflect stronger TCR-peptide-MHC interactions; thus, these parameters would seem well suited to identify higher avidity CTL. However, large disparities were found comparing CTL multimer binding with peptide sensitivity. In contrast, CD8(+) CTL with superior Ag sensitivity mediated good tumor cytotoxicity and also secreted the triple combination of IFN-γ, IL-2, and TNF-α, representing a Th1 pattern often missing in lower avidity CTL. Furthermore, recipient lymphocytes were imbued with high Ag sensitivity, superior tumor recognition, as well as capacity for Th1 polycytokine secretion after transduction with the TCR of a high-avidity CTL. Thus, Th1 polycytokine secretion served as a suitable parameter to rapidly demark cytotoxic CD8(+) T cell clones for further TCR evaluation.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Células TH1/inmunología , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Adhesión Celular/inmunología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Células Clonales , Técnicas de Cocultivo , Citocinas/clasificación , Citocinas/metabolismo , Humanos , Células TH1/metabolismo , Células TH1/patología
11.
Blood ; 119(15): 3440-9, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22371883

RESUMEN

The hyaluronan-mediated motility receptor (HMMR/Rhamm) is overexpressed in numerous tumor types, including acute lymphoid leukemia and acute myeloid leukemia (AML). Several studies have reported the existence of T-cell responses directed against HMMR in AML patients that are linked to better clinical outcome. Therefore, we explored the use of HMMR-specific TCRs for transgenic expression in lymphocytes and their in vivo impact on HMMR(+) solid tumors and disseminated leukemia. We obtained TCRs via an in vitro priming approach in combination with CD137-mediated enrichment. Recipient lymphocytes expressing transgenic TCR revealed the specific tumor recognition pattern seen with the original T cells. Adoptive transfer experiments using a humanized xenograft mouse model resulted in significantly retarded solid tumor outgrowth, which was enhanced using IL-15-conditioned, TCR-transgenic effector memory cells. These cells also showed an increased potency to retard the outgrowth of disseminated AML, and this was further improved using CD8-enriched effector memory cells. To define a safe clinical setting for HMMR-TCR gene therapy, we analyzed transgenic T-cell recognition of hematopoietic stem cells (HSCs) and found on-target killing of HLA-A2(+) HSCs. Our findings clearly limit the use of HMMR-TCR therapy to MHC- mismatched HSC transplantation, in which HLA-A2 differences can be used to restrict recognition to patient HSCs and leukemia.


Asunto(s)
Procesos de Crecimiento Celular/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/inmunología , Receptores de Hialuranos/genética , Receptores de Hialuranos/inmunología , Linfocitos/fisiología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Animales , Procesos de Crecimiento Celular/inmunología , Células Cultivadas , Terapia Genética/métodos , Células HEK293 , Humanos , Inmunoterapia Adoptiva/métodos , Células K562 , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Ratones Transgénicos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Innate Immun ; 3(4): 420-34, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21178331

RESUMEN

The CD6 scavenger receptor is known to be expressed on virtually all T cells and is supposed to be involved in costimulation, synapse formation, thymic selection and leukocyte migration. Here, we demonstrate that CD6 is differentially expressed by a subpopulation of peripheral CD56(dim) natural killer (NK) cells and absent on CD56(bright) NK cells. CD56(dim)CD16(+) cells represent the major NK subset in the periphery, and most cells within this group are positive for CD6. Most killer immunoglobulin-like receptor- and immunoglobulin-like transcript-positive cells also belong to the CD6(+) subpopulation, as expected from their restricted expression on CD56(dim) NK cells. In addition, CD6(+) NK cells are similar to the newly identified CD94(low)CD56(dim) NK subpopulation and most distant from the recently defined CD27(+) NK subpopulation based on the reverse correlation of expression between CD6 and CD27, a marker associated primarily with CD56(bright) NK cells. With respect to CD6 function on NK cells, direct CD6 triggering did not result in degranulation but induced secretion of cytokines (interferon-γ and tumor necrosis factor-α) and chemokines [CXCL10 (IP-10), CXCL1 (GRO-α)]. Thus, CD6 expression on peripheral NK cells marks a novel CD56(dim) subpopulation associated with distinct patterns of cytokine and chemokine secretion.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígeno CD56/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Asesinas Naturales/metabolismo , Receptores Depuradores/metabolismo , Diferenciación Celular , Quimiocina CXCL1/metabolismo , Quimiocina CXCL10/metabolismo , Humanos , Interferón gamma/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Transl Med ; 8: 90, 2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20920165

RESUMEN

BACKGROUND: Antigen-loaded dendritic cells (DC) are capable of priming naïve T cells and therefore represent an attractive adjuvant for vaccine development in anti-tumor immunotherapy. Numerous protocols have been described to date using different maturation cocktails and time periods for the induction of mature DC (mDC) in vitro. For clinical application, the use of mDC that can be generated in only three days saves on the costs of cytokines needed for large scale vaccine cell production and provides a method to produce cells within a standard work-week schedule in a GMP facility. METHODS: In this study, we addressed the properties of antigen uptake, processing and presentation by monocyte-derived DC prepared in three days (3d mDC) compared with conventional DC prepared in seven days (7d mDC), which represent the most common form of DC used for vaccines to date. RESULTS: Although they showed a reduced capacity for spontaneous antigen uptake, 3d mDC displayed higher capacity for stimulation of T cells after loading with an extended synthetic peptide that requires processing for MHC binding, indicating they were more efficient at antigen processing than 7d DC. We found, however, that 3d DC were less efficient at expressing protein after introduction of in vitro transcribed (ivt)RNA by electroporation, based on published procedures. This deficit was overcome by altering electroporation parameters, which led to improved protein expression and capacity for T cell stimulation using low amounts of ivtRNA. CONCLUSIONS: This new procedure allows 3d mDC to replace 7d mDC for use in DC-based vaccines that utilize long peptides, proteins or ivtRNA as sources of specific antigen.


Asunto(s)
Antígenos/inmunología , Células Dendríticas/inmunología , Vacunas/biosíntesis , Línea Celular Tumoral , Humanos , Vacunas/inmunología
14.
J Clin Invest ; 120(11): 3869-77, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20978348

RESUMEN

The apoptosis inhibitor protein survivin is overexpressed in many tumors, making it a candidate target molecule for various forms of immunotherapy. To explore survivin as a target antigen for adoptive T cell therapy using lymphocytes expressing survivin-specific transgenic T cell receptors (Tg-TCRs), we isolated HLA-A2-allorestricted survivin-specific T cells with high functional avidity. Lymphocytes expressing Tg-TCRs were derived from these T cells and specifically recognized HLA-A2+ survivin+ tumor cells. Surprisingly, HLA-A2+ but not HLA-A2- lymphocytes expressing Tg-TCRs underwent extensive apoptosis over time. This demise was caused by HLA-A2-restricted fratricide that occurred due to survivin expression in lymphocytes, which created ligands for Tg-TCR recognition. Therefore, survivin-specific TCR gene therapy would be limited to application in HLA-A2-mismatched stem cell transplantation. We also noted that lymphocytes that expressed survivin-specific Tg-TCRs killed T cell clones of various specificities derived from HLA-A2+ but not HLA-A2- donors. These results raise a general question regarding the development of cancer vaccines that target proteins that are also expressed in activated lymphocytes, since induction of high-avidity T cells that expand in lymph nodes following vaccination or later accumulate at tumor sites might limit themselves by self-MHC-restricted fratricide while at the same time inadvertently eliminating neighboring T cells of other specificities.


Asunto(s)
Antígeno HLA-A2 , Linfocitos/inmunología , Complejo Mayor de Histocompatibilidad , Proteínas Asociadas a Microtúbulos/inmunología , Receptores de Antígenos de Linfocitos T , Transgenes , Animales , Muerte Celular/inmunología , Línea Celular , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Survivin , Linfocitos T/inmunología
15.
J Immunol ; 185(1): 738-47, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20511554

RESUMEN

In this paper, we describe a new method for preparation of human dendritic cells (DCs) that secrete bioactive IL-12(p70) using synthetic immunostimulatory compounds as TLR7/8 agonists. Monocyte-derived DCs were generated using a procedure that provided mature cells within 3 d. Several maturation mixtures that contained various cytokines, IFN-gamma, different TLR agonists, and PGE(2) were compared for impact on cell recovery, phenotype, cytokine secretion, migration, and lymphocyte activation. Mixtures that included the TLR7/8 agonists R848 or CL075, combined with the TLR3 agonist polyinosinic:polycytidylic acid, yielded 3-d mature DCs that secreted high levels of IL-12(p70), showed strong chemotaxis to CCR7 ligands, and had a positive costimulatory potential. They also had excellent capacity to activate NK cells, effectively polarized CD4(+) and CD8(+) T cells to secrete IFN-gamma and to induce T cell-mediated cytotoxic function. Thereby, mature DCs prepared within 3 d using such maturation mixtures displayed optimal functions required for vaccine development.


Asunto(s)
Diferenciación Celular/inmunología , Polaridad Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Quinolinas/farmacología , Células TH1/inmunología , Tiazoles/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/síntesis química , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Células Dendríticas/citología , Humanos , Imidazoles/agonistas , Imidazoles/farmacología , Inmunoterapia Adoptiva/métodos , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ligandos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Poli I-C/metabolismo , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Células TH1/efectos de los fármacos , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 8/fisiología
16.
Blood ; 114(10): 2131-9, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19587379

RESUMEN

Adoptive transfer of T cells expressing transgenic T-cell receptors (TCRs) with antitumor function is a hopeful new therapy for patients with advanced tumors; however, there is a critical bottleneck in identifying high-affinity TCR specificities needed to treat different malignancies. We have developed a strategy using autologous dendritic cells cotransfected with RNA encoding an allogeneic major histocompatibility complex molecule and a tumor-associated antigen to obtain allo-restricted peptide-specific T cells having superior capacity to recognize tumor cells and higher functional avidity. This approach provides maximum flexibility because any major histocompatibility complex molecule and any tumor-associated antigen can be combined in the dendritic cells used for priming of autologous T cells. TCRs of allo-restricted T cells, when expressed as transgenes in activated peripheral blood lymphocytes, transferred superior function compared with self-restricted TCR. This approach allows high-avidity T cells and TCR specific for tumor-associated self-peptides to be easily obtained for direct adoptive T-cell therapy or for isolation of therapeutic transgenic TCR sequences.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Antígenos HLA/inmunología , Isoantígenos/inmunología , Neoplasias/inmunología , Péptidos/inmunología , ARN/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Antígenos HLA/genética , Humanos , Isoantígenos/genética , Neoplasias/genética , Neoplasias/terapia , Péptidos/genética , ARN/genética , Receptores de Antígenos de Linfocitos T/genética , Transfección
17.
J Immunother ; 31(1): 52-62, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18157012

RESUMEN

Tumor cells that show downregulation of their tumor-associated antigens (TAAs) may be able to escape immune-mediated elimination. Therefore, efficient vaccine strategies attempt to target multiple TAAs simultaneously. This is easily achieved in dendritic cell (DC)-based vaccines by introducing antigens in the form of RNA. Although insufficient message may hinder adequate expression of individual TAAs when using total-tumor RNA, high amounts of individual RNAs as pools yield DCs presenting high numbers of specific peptide-major histocompatibility complex ligands with epitopes derived from different TAAs. We used the transfer of RNAs encoding the well-defined melanoma TAAs tyrosinase, Melan-A, CDK4mut, gp100, SNRP116mut, and GPNMBmut to characterize DCs at the levels of transfected RNA, expressed protein and peptide-major histocompatibility complex ligand presentation. TAA-encoding RNA was rapidly degraded in the DCs, allowing only a single surge in protein expression shortly after transfection. We compared the functional capacity of DCs transfected with pools of 3 versus 6 RNAs. Whereas functional assays demonstrated a decrease in stimulatory capacity of DCs transfected with a pool of 3 RNAs by only 30% as compared with single RNAs, a 60% loss was seen with 6 RNAs. We conclude that larger RNA pools result in diminished presentation of individual epitopes and suggest that smaller pools of RNA be transfected into separate DC populations which are then pooled to create multiplex vaccines.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , ARN/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/genética , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/inmunología , Quinasa 4 Dependiente de la Ciclina/metabolismo , Células Dendríticas/metabolismo , Citometría de Flujo , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Cinética , Antígeno MART-1 , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/inmunología , Monofenol Monooxigenasa/metabolismo , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/inmunología , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Transfección , Antígeno gp100 del Melanoma
18.
World J Urol ; 23(3): 166-74, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15997395

RESUMEN

Initial vaccine developments for renal cell carcinoma (RCC) have concentrated on cell-based approaches in which tumor cells themselves provide mixtures of unknown tumor-associated antigens as immunizing agents. Antigens derived from autologous tumors can direct responses to molecular composites characteristic of individual tumors, whereas antigens derived from allogeneic tumor cells must be commonly shared by RCC. Three types of cell-based vaccine for RCC have been investigated: isolated tumor cell suspensions, gene modified tumor cells and dendritic cells (DCs) expressing RCC-associated antigens. Approaches using genetic modification of autologous RCC have included ex vivo modification of tumor cells or modification of tumors in vivo. We have used gene-modification of allogeneic tumor cell lines to create generic RCC vaccines. More recently, emphasis has shifted to the use of DCs as cell-based vaccines for RCC. DCs have moved to a position of central interest because of their excellent stimulatory capacity, combined with their ability to process and present antigens to both naive CD4 and CD8 cells. The long impasse in identifying molecular targets for specific immunotherapy of RCC is now rapidly being overcome through the use of tools and information emerging from human genome research. Identification of candidate molecules expressed by RCC using cDNA arrays, combined with protein arrays and identification of peptides presented by MHC molecules, allow specific vaccines to be tailored to the antigenic profile of individual tumors, providing the basis for development of patient-specific vaccines.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Renales/terapia , Células Dendríticas/trasplante , Terapia Genética/métodos , Neoplasias Renales/terapia , Vacunas contra el Cáncer/genética , Células Dendríticas/citología , Células Dendríticas/fisiología , Ingeniería Genética , Humanos , Monocitos/citología
19.
DNA Repair (Amst) ; 3(10): 1345-53, 2004 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-15336629

RESUMEN

Homologous recombination between identical stretches of DNA depends on the coordinated action of many tightly regulated proteins. Cellular defects in homologous recombination are strongly associated with increased genomic instability and tumorigenesis. In cells of the cancer-prone syndrome ataxia telangiectasia (A-T), increased intrachromosomal recombination has been demonstrated, while extrachromosomal recombination has been discussed controversially. We constructed a novel, episomally replicating pGrec recombination vector containing two mutated alleles of the enhanced green fluorescent protein (eGFP) gene. Homologous recombination can reconstitute functional wildtype eGFP, thus allowing detection of recombination events based on cellular eGFP fluorescence. Using an isogenic cell pair of A-T fibroblasts and derivatives complemented by an ATM expression vector, we were able to demonstrate in A-T cells high extrachromosomal recombination rates, which are suppressed upon ectopic ATM expression. We thus found that ATM deficiency increases spontaneous recombination not only in intrachromosomal but also in extrachromosomal substrates, suggesting that lack of ATM increases homologous recombination independent of the chromatin structure.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , ADN , Proteínas Fluorescentes Verdes/fisiología , Plásmidos/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Recombinación Genética , Proteínas Supresoras de Tumor/deficiencia , Animales , Ataxia Telangiectasia/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Células CHO , Proteínas de Ciclo Celular , Cromosomas/genética , Cricetinae , ADN/genética , ADN/metabolismo , ADN Recombinante , Fibroblastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...