Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cancer Cell ; 41(10): 1817-1828.e9, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37683639

RESUMEN

The dysregulated expression of immune checkpoint molecules enables cancer cells to evade immune destruction. While blockade of inhibitory immune checkpoints like PD-L1 forms the basis of current cancer immunotherapies, a deficiency in costimulatory signals can render these therapies futile. CD58, a costimulatory ligand, plays a crucial role in antitumor immune responses, but the mechanisms controlling its expression remain unclear. Using two systematic approaches, we reveal that CMTM6 positively regulates CD58 expression. Notably, CMTM6 interacts with both CD58 and PD-L1, maintaining the expression of these two immune checkpoint ligands with opposing functions. Functionally, the presence of CMTM6 and CD58 on tumor cells significantly affects T cell-tumor interactions and response to PD-L1-PD-1 blockade. Collectively, these findings provide fundamental insights into CD58 regulation, uncover a shared regulator of stimulatory and inhibitory immune checkpoints, and highlight the importance of tumor-intrinsic CMTM6 and CD58 expression in antitumor immune responses.


Asunto(s)
Antígeno B7-H1 , Proteínas con Dominio MARVEL , Proteínas de la Mielina , Neoplasias , Linfocitos T , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Inmunidad , Inmunoterapia , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Linfocitos T/inmunología , Proteínas de la Mielina/metabolismo , Proteínas con Dominio MARVEL/metabolismo
2.
J Immunother Cancer ; 8(2)2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32753545

RESUMEN

Treatment of metastatic melanoma with autologous tumor infiltrating lymphocytes (TILs) is currently applied in several centers. Robust and remarkably consistent overall response rates, of around 50% of treated patients, have been observed across hospitals, including a substantial fraction of durable, complete responses. PURPOSE: Execute a phase I/II feasibility study with TIL therapy in metastatic melanoma at the Netherlands Cancer Institute, with the goal to assess feasibility and potential value of a randomized phase III trial. EXPERIMENTAL: Ten patients were treated with TIL therapy. Infusion products and peripheral blood samples were phenotypically characterized and neoantigen reactivity was assessed. Here, we present long-term clinical outcome and translational data on neoantigen reactivity of the T cell products. RESULTS: Five out of 10 patients, who were all anti-PD-1 naïve at time of treatment, showed an objective clinical response, including two patients with a complete response that are both ongoing for more than 7 years. Immune monitoring demonstrated that neoantigen-specific T cells were detectable in TIL infusion products from three out of three patients analyzed. For six out of the nine neoantigen-specific T cell responses detected in these TIL products, T cell response magnitude increased significantly in the peripheral blood compartment after therapy, and neoantigen-specific T cells were detectable for up to 3 years after TIL infusion. CONCLUSION: The clinical results from this study confirm the robustness of TIL therapy in metastatic melanoma and the potential role of neoantigen-specific T cell reactivity. In addition, the data from this study supported the rationale to initiate an ongoing multicenter phase III TIL trial.


Asunto(s)
Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma/genética , Linfocitos T/metabolismo , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad
3.
Proc Natl Acad Sci U S A ; 117(12): 6686-6696, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32161126

RESUMEN

Cytotoxic CD8+ T cells can effectively kill target cells by producing cytokines, chemokines, and granzymes. Expression of these effector molecules is however highly divergent, and tools that identify and preselect CD8+ T cells with a cytotoxic expression profile are lacking. Human CD8+ T cells can be divided into IFN-γ- and IL-2-producing cells. Unbiased transcriptomics and proteomics analysis on cytokine-producing fixed CD8+ T cells revealed that IL-2+ cells produce helper cytokines, and that IFN-γ+ cells produce cytotoxic molecules. IFN-γ+ T cells expressed the surface marker CD29 already prior to stimulation. CD29 also marked T cells with cytotoxic gene expression from different tissues in single-cell RNA-sequencing data. Notably, CD29+ T cells maintained the cytotoxic phenotype during cell culture, suggesting a stable phenotype. Preselecting CD29-expressing MART1 TCR-engineered T cells potentiated the killing of target cells. We therefore propose that CD29 expression can help evaluate and select for potent therapeutic T cell products.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica/inmunología , Integrina beta1/metabolismo , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Melanoma/patología , Linfocitos T Citotóxicos/inmunología , Perfilación de la Expresión Génica , Humanos , Melanoma/inmunología , Melanoma/metabolismo , Pronóstico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Tasa de Supervivencia
5.
Cell ; 178(3): 585-599.e15, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31303383

RESUMEN

New opportunities are needed to increase immune checkpoint blockade (ICB) benefit. Whereas the interferon (IFN)γ pathway harbors both ICB resistance factors and therapeutic opportunities, this has not been systematically investigated for IFNγ-independent signaling routes. A genome-wide CRISPR/Cas9 screen to sensitize IFNγ receptor-deficient tumor cells to CD8 T cell elimination uncovered several hits mapping to the tumor necrosis factor (TNF) pathway. Clinically, we show that TNF antitumor activity is only limited in tumors at baseline and in ICB non-responders, correlating with its low abundance. Taking advantage of the genetic screen, we demonstrate that ablation of the top hit, TRAF2, lowers the TNF cytotoxicity threshold in tumors by redirecting TNF signaling to favor RIPK1-dependent apoptosis. TRAF2 loss greatly enhanced the therapeutic potential of pharmacologic inhibition of its interaction partner cIAP, another screen hit, thereby cooperating with ICB. Our results suggest that selective reduction of the TNF cytotoxicity threshold increases the susceptibility of tumors to immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Interferón gamma/metabolismo , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/mortalidad , Neoplasias/terapia , ARN Guía de Kinetoplastida/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Transducción de Señal/efectos de los fármacos , Factor 2 Asociado a Receptor de TNF/deficiencia , Factor 2 Asociado a Receptor de TNF/genética , Factor de Necrosis Tumoral alfa/farmacología , Receptor de Interferón gamma
6.
Nat Med ; 25(4): 612-619, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833751

RESUMEN

Cancer cells can evade immune surveillance through the expression of inhibitory ligands that bind their cognate receptors on immune effector cells. Expression of programmed death ligand 1 in tumor microenvironments is a major immune checkpoint for tumor-specific T cell responses as it binds to programmed cell death protein-1 on activated and dysfunctional T cells1. The activity of myeloid cells such as macrophages and neutrophils is likewise regulated by a balance between stimulatory and inhibitory signals. In particular, cell surface expression of the CD47 protein creates a 'don't eat me' signal on tumor cells by binding to SIRPα expressed on myeloid cells2-5. Using a haploid genetic screen, we here identify glutaminyl-peptide cyclotransferase-like protein (QPCTL) as a major component of the CD47-SIRPα checkpoint. Biochemical analysis demonstrates that QPCTL is critical for pyroglutamate formation on CD47 at the SIRPα binding site shortly after biosynthesis. Genetic and pharmacological interference with QPCTL activity enhances antibody-dependent cellular phagocytosis and cellular cytotoxicity of tumor cells. Furthermore, interference with QPCTL expression leads to a major increase in neutrophil-mediated killing of tumor cells in vivo. These data identify QPCTL as a novel target to interfere with the CD47 pathway and thereby augment antibody therapy of cancer.


Asunto(s)
Aminoaciltransferasas/metabolismo , Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Receptores Inmunológicos/metabolismo , Aminoaciltransferasas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Ratones Transgénicos , Neoplasias/patología , Proteínas Opsoninas/metabolismo , Ácido Pirrolidona Carboxílico/metabolismo
7.
PLoS One ; 14(2): e0212053, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30753225

RESUMEN

Experimental and clinical observations have highlighted the role of cytotoxic T cells in human tumor control. However, the parameters that control tumor cell sensitivity to T cell attack remain incompletely understood. To identify modulators of tumor cell sensitivity to T cell effector mechanisms, we performed a whole genome haploid screen in HAP1 cells. Selection of tumor cells by exposure to tumor-specific T cells identified components of the interferon-γ (IFN-γ) receptor (IFNGR) signaling pathway, and tumor cell killing by cytotoxic T cells was shown to be in large part mediated by the pro-apoptotic effects of IFN-γ. Notably, we identified schlafen 11 (SLFN11), a known modulator of DNA damage toxicity, as a regulator of tumor cell sensitivity to T cell-secreted IFN-γ. SLFN11 does not influence IFNGR signaling, but couples IFNGR signaling to the induction of the DNA damage response (DDR) in a context dependent fashion. In line with this role of SLFN11, loss of SLFN11 can reduce IFN-γ mediated toxicity. Collectively, our data indicate that SLFN11 can couple IFN-γ exposure of tumor cells to DDR and cellular apoptosis. Future work should reveal the mechanistic basis for the link between IFNGR signaling and DNA damage response, and identify tumor cell types in which SLFN11 contributes to the anti-tumor activity of T cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Interferón gamma/farmacología , Proteínas Nucleares/metabolismo , Linfocitos T Citotóxicos/inmunología , Clorometilcetonas de Aminoácidos/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Quinolinas/farmacología , Interferencia de ARN , ARN Guía de Kinetoplastida/metabolismo , ARN Interferente Pequeño/metabolismo , Linfocitos T Citotóxicos/metabolismo
8.
Cell ; 172(3): 549-563.e16, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29275860

RESUMEN

The immune system can mount T cell responses against tumors; however, the antigen specificities of tumor-infiltrating lymphocytes (TILs) are not well understood. We used yeast-display libraries of peptide-human leukocyte antigen (pHLA) to screen for antigens of "orphan" T cell receptors (TCRs) expressed on TILs from human colorectal adenocarcinoma. Four TIL-derived TCRs exhibited strong selection for peptides presented in a highly diverse pHLA-A∗02:01 library. Three of the TIL TCRs were specific for non-mutated self-antigens, two of which were present in separate patient tumors, and shared specificity for a non-mutated self-antigen derived from U2AF2. These results show that the exposed recognition surface of MHC-bound peptides accessible to the TCR contains sufficient structural information to enable the reconstruction of sequences of peptide targets for pathogenic TCRs of unknown specificity. This finding underscores the surprising specificity of TCRs for their cognate antigens and enables the facile indentification of tumor antigens through unbiased screening.


Asunto(s)
Adenocarcinoma/inmunología , Antígenos de Neoplasias/inmunología , Neoplasias Colorrectales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Anciano , Animales , Antígenos de Neoplasias/química , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Antígenos HLA-A/química , Antígenos HLA-A/inmunología , Humanos , Masculino , Persona de Mediana Edad , Biblioteca de Péptidos , Células Sf9 , Spodoptera
9.
Nature ; 549(7670): 106-110, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28813410

RESUMEN

The clinical benefit for patients with diverse types of metastatic cancers that has been observed upon blockade of the interaction between PD-1 and PD-L1 has highlighted the importance of this inhibitory axis in the suppression of tumour-specific T-cell responses. Notwithstanding the key role of PD-L1 expression by cells within the tumour micro-environment, our understanding of the regulation of the PD-L1 protein is limited. Here we identify, using a haploid genetic screen, CMTM6, a type-3 transmembrane protein of previously unknown function, as a regulator of the PD-L1 protein. Interference with CMTM6 expression results in impaired PD-L1 protein expression in all human tumour cell types tested and in primary human dendritic cells. Furthermore, through both a haploid genetic modifier screen in CMTM6-deficient cells and genetic complementation experiments, we demonstrate that this function is shared by its closest family member, CMTM4, but not by any of the other CMTM members tested. Notably, CMTM6 increases the PD-L1 protein pool without affecting PD-L1 (also known as CD274) transcription levels. Rather, we demonstrate that CMTM6 is present at the cell surface, associates with the PD-L1 protein, reduces its ubiquitination and increases PD-L1 protein half-life. Consistent with its role in PD-L1 protein regulation, CMTM6 enhances the ability of PD-L1-expressing tumour cells to inhibit T cells. Collectively, our data reveal that PD-L1 relies on CMTM6/4 to efficiently carry out its inhibitory function, and suggest potential new avenues to block this pathway.


Asunto(s)
Antígeno B7-H1/metabolismo , Proteínas con Dominio MARVEL/metabolismo , Antígeno B7-H1/biosíntesis , Antígeno B7-H1/química , Sistemas CRISPR-Cas , Línea Celular Tumoral , Células Dendríticas/metabolismo , Prueba de Complementación Genética , Haploidia , Humanos , Proteínas con Dominio MARVEL/genética , Melanoma/genética , Melanoma/metabolismo , Unión Proteica , Estabilidad Proteica , Ubiquitinación
10.
Mol Ther Nucleic Acids ; 5: e298, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27003756

RESUMEN

Recent years have seen major breakthroughs in genome-engineering systems, such as transposon-mediated gene delivery systems and CRISPR-Cas9-mediated genome-editing tools. In these systems, transient expression of auxiliary genes is responsible for permanent genomic modification. For both systems, it would be valuable to select for cells that are likely to undergo stable genome modification. Importantly, in particular for clinical applications of genome-engineered cell products, it will also be of importance to remove those cells that, due to random vector integration, display an unwanted stable expression of the auxiliary gene. Here, we develop a traceless selection system that on the one hand allows efficient enrichment of modified cells, and on the other hand can be used to select against cells that retain expression of the auxiliary gene. The value of this system to produce highly enriched-auxiliary gene-free cell products is demonstrated.

11.
Mol Ther ; 23(9): 1541-50, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25896248

RESUMEN

Here, we describe a fatal serious adverse event observed in a patient infused with autologous T-cell receptor (TCR) transduced T cells. This TCR, originally obtained from a melanoma patient, recognizes the well-described HLA-A*0201 restricted 26-35 epitope of MART-1, and was not affinity enhanced. Patient 1 with metastatic melanoma experienced a cerebral hemorrhage, epileptic seizures, and a witnessed cardiac arrest 6 days after cell infusion. Three days later, the patient died from multiple organ failure and irreversible neurologic damage. After T-cell infusion, levels of IL-6, IFN-γ, C-reactive protein (CRP), and procalcitonin increased to extreme levels, indicative of a cytokine release syndrome or T-cell-mediated inflammatory response. Infused T cells could be recovered from blood, broncho-alveolar lavage, ascites, and after autopsy from tumor sites and heart tissue. High levels of NT-proBNP indicate semi-acute heart failure. No cross reactivity of the modified T cells toward a beating cardiomyocyte culture was observed. Together, these observations suggest that high levels of inflammatory cytokines alone or in combination with semi-acute heart failure and epileptic seizure may have contributed substantially to the occurrence of the acute and lethal event. Protocol modifications to limit the risk of T-cell activation-induced toxicity are discussed.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Inmunoterapia Adoptiva/efectos adversos , Antígeno MART-1/inmunología , Receptores de Antígenos de Linfocitos T/genética , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Resultado Fatal , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Antígeno MART-1/metabolismo , Melanoma/diagnóstico , Melanoma/genética , Melanoma/inmunología , Melanoma/terapia , Estadificación de Neoplasias , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción Genética
12.
Hum Gene Ther Methods ; 25(5): 277-87, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25143008

RESUMEN

Advances in genetic engineering have made it possible to generate human T-cell products that carry desired functionalities, such as the ability to recognize cancer cells. The currently used strategies for the generation of gene-modified T-cell products lead to highly differentiated cells within the infusion product, and on the basis of data obtained in preclinical models, this is likely to impact the efficacy of these products. We set out to develop a good manufacturing practice (GMP) protocol that yields T-cell receptor (TCR) gene-modified T-cells with more favorable properties for clinical application. Here, we show the robust clinical-scale production of human peripheral blood T-cells with an early memory phenotype that express a MART-1-specific TCR. By combining selection and stimulation using anti-CD3/CD28 beads for retroviral transduction, followed by expansion in the presence of IL-7 and IL-15, production of a well-defined clinical-scale TCR gene-modified T-cell product could be achieved. A major fraction of the T-cells generated in this fashion were shown to coexpress CD62L and CD45RA, and express CD27 and CD28, indicating a central memory or memory stemlike phenotype. Furthermore, these cells produced IFNγ, TNFα, and IL-2 and displayed cytolytic activity against target cells expressing the relevant antigen. The T-cell products manufactured by this robust and validated GMP production process are now undergoing testing in a phase I/IIa clinical trial in HLA-A*02:01 MART-1-positive advanced stage melanoma patients. To our knowledge, this is the first clinical trial protocol in which the combination of IL-7 and IL-15 has been applied for the generation of gene-modified T-cell products.


Asunto(s)
Citotoxicidad Inmunológica/genética , Memoria Inmunológica/genética , Melanoma/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Neoplasias Cutáneas/terapia , Linfocitos T Citotóxicos/inmunología , Antígenos CD/genética , Antígenos CD/inmunología , Ingeniería Celular/métodos , Proliferación Celular , Ensayos Clínicos como Asunto , Expresión Génica , Vectores Genéticos , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-15/farmacología , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-7/farmacología , Antígeno MART-1/genética , Antígeno MART-1/inmunología , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Retroviridae/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/trasplante , Transducción Genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
13.
Nat Med ; 19(11): 1534-41, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24121928

RESUMEN

The transfer of T cell receptor (TCR) genes into patient T cells is a promising approach for the treatment of both viral infections and cancer. Although efficient methods exist to identify antibodies for the treatment of these diseases, comparable strategies to identify TCRs have been lacking. We have developed a high-throughput DNA-based strategy to identify TCR sequences by the capture and sequencing of genomic DNA fragments encoding the TCR genes. We establish the value of this approach by assembling a large library of cancer germline tumor antigen-reactive TCRs. Furthermore, by exploiting the quantitative nature of TCR gene capture, we show the feasibility of identifying antigen-specific TCRs in oligoclonal T cell populations from either human material or TCR-humanized mice. Finally, we demonstrate the ability to identify tumor-reactive TCRs within intratumoral T cell subsets without knowledge of antigen specificities, which may be the first step toward the development of autologous TCR gene therapy to target patient-specific neoantigens in human cancer.


Asunto(s)
Genes Codificadores de los Receptores de Linfocitos T , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Biblioteca de Genes , Terapia Genética , Humanos , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología
14.
Proc Natl Acad Sci U S A ; 109(48): 19739-44, 2012 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-23150545

RESUMEN

Recent work has demonstrated that following the clearance of infection a stable population of memory T cells remains present in peripheral organs and contributes to the control of secondary infections. However, little is known about how tissue-resident memory T cells behave in situ and how they encounter newly infected target cells. Here we demonstrate that antigen-specific CD8(+) T cells that remain in skin following herpes simplex virus infection show a steady-state crawling behavior in between keratinocytes. Spatially explicit simulations of the migration of these tissue-resident memory T cells indicate that the migratory dendritic behavior of these cells allows the detection of antigen-expressing target cells in physiologically relevant time frames of minutes to hours. Furthermore, we provide direct evidence for the identification of rare antigen-expressing epithelial cells by skin-patrolling memory T cells in vivo. These data demonstrate the existence of skin patrol by memory T cells and reveal the value of this patrol in the rapid detection of renewed infections at a previously infected site.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Epitelio/inmunología , Humanos
15.
Hum Gene Ther ; 21(10): 1335-42, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20486771

RESUMEN

Retroviral transduction is the most commonly used strategy to obtain long-term expression of therapeutic genes. To efficiently transduce mammalian cells, a recombinant fibronectin molecule, RetroNectin, is generally used to juxtapose viral particles and cells, and thereby enhance viral uptake. Although this strategy has become widely adopted, in particular for the genetic modification of hematopoietic cells, several limitations apply. For example, it requires the use of culture systems that allow protein coating, something that is not possible for many of the closed cell culture systems that are used in clinical trials. Furthermore, efficient transduction is obtained only when culture systems can be exposed to centrifugation, an approach termed spin transduction. Here, we describe a novel and more potent strategy for the transduction of T cells that can be applied on a clinical scale. We show that RetroNectin can efficiently be coated onto epoxy-modified paramagnetic beads. After a blocking step, these beads can subsequently bind retroviral particles from viral supernatants, rendering such supernatants largely devoid of functional viral particles. Addition of these virus-loaded beads to activated T cells results in efficient retroviral infection. Importantly, transduction does not require the use of culture systems that are compatible with protein coating, nor is it dependent on centrifugation of either the viral supernatant or the cells. Finally, cell growth, phenotype, and function of spin-transduced versus bead-transduced cells are comparable. Viral coating of microbeads should facilitate the production of genetically modified cells, in particular for use in clinical trials.


Asunto(s)
Vectores Genéticos , Retroviridae/genética , Linfocitos T , Transducción Genética/métodos , Adhesión Celular , Fibronectinas/genética , Fibronectinas/metabolismo , Citometría de Flujo , Expresión Génica , Humanos , Microesferas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T/virología
16.
J Immunol ; 184(9): 4936-46, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20351194

RESUMEN

Protective adaptive immune responses rely on TCR-mediated recognition of Ag-derived peptides presented by self-MHC molecules. However, self-Ag (tumor)-specific TCRs are often of too low affinity to achieve best functionality. To precisely assess the relationship between TCR-peptide-MHC binding parameters and T cell function, we tested a panel of sequence-optimized HLA-A(*)0201/NY-ESO-1(157-165)-specific TCR variants with affinities lying within physiological boundaries to preserve antigenic specificity and avoid cross-reactivity, as well as two outliers (i.e., a very high- and a low-affinity TCR). Primary human CD8 T cells transduced with these TCRs demonstrated robust correlations between binding measurements of TCR affinity and avidity and the biological response of the T cells, such as TCR cell-surface clustering, intracellular signaling, proliferation, and target cell lysis. Strikingly, above a defined TCR-peptide-MHC affinity threshold (K(D) < approximately 5 muM), T cell function could not be further enhanced, revealing a plateau of maximal T cell function, compatible with the notion that multiple TCRs with slightly different affinities participate equally (codominantly) in immune responses. We propose that rational design of improved self-specific TCRs may not need to be optimized beyond a given affinity threshold to achieve both optimal T cell function and avoidance of the unpredictable risk of cross-reactivity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citotoxicidad Inmunológica , Proteínas de Neoplasias/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Adhesión Celular/genética , Adhesión Celular/inmunología , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Citotoxicidad Inmunológica/genética , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/genética , Fragmentos de Péptidos/genética , Unión Proteica/genética , Unión Proteica/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética
17.
Blood ; 110(10): 3564-72, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17660381

RESUMEN

A recent phase 1 trial has demonstrated that the generation of tumor-reactive T lymphocytes by transfer of specific T-cell receptor (TCR) genes into autologous lymphocytes is feasible. However, compared with results obtained by infusion of tumor-infiltrating lymphocytes, the response rate observed in this first TCR gene therapy trial is low. One strategy that is likely to enhance the success rate of TCR gene therapy is the use of tumor-reactive TCRs with a higher capacity for tumor cell recognition. We therefore sought to develop standardized procedures for the selection of well-expressed, high-affinity, and safe human TCRs. Here we show that TCR surface expression can be improved by modification of TCR alpha and beta sequences and that such improvement has a marked effect on the in vivo function of TCR gene-modified T cells. From a panel of human, melanoma-reactive TCRs we subsequently selected the TCR with the highest affinity. Furthermore, a generally applicable assay was used to assess the lack of alloreactivity of this TCR against a large series of common human leukocyte antigen alleles. The procedures described in this study should be of general value for the selection of well- and stably expressed, high-affinity, and safe human TCRs for subsequent clinical testing.


Asunto(s)
Terapia Genética/métodos , Melanoma/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Inmunoterapia Adoptiva , Células Jurkat , Células K562 , Melanoma/genética , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Especificidad por Sustrato , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...