Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Blood ; 140(20): 2113-2126, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-35704690

RESUMEN

The BCL2 inhibitor venetoclax has been approved to treat different hematological malignancies. Because there is no common genetic alteration causing resistance to venetoclax in chronic lymphocytic leukemia (CLL) and B-cell lymphoma, we asked if epigenetic events might be involved in venetoclax resistance. Therefore, we employed whole-exome sequencing, methylated DNA immunoprecipitation sequencing, and genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 screening to investigate venetoclax resistance in aggressive lymphoma and high-risk CLL patients. We identified a regulatory CpG island within the PUMA promoter that is methylated upon venetoclax treatment, mediating PUMA downregulation on transcript and protein level. PUMA expression and sensitivity toward venetoclax can be restored by inhibition of methyltransferases. We can demonstrate that loss of PUMA results in metabolic reprogramming with higher oxidative phosphorylation and adenosine triphosphate production, resembling the metabolic phenotype that is seen upon venetoclax resistance. Although PUMA loss is specific for acquired venetoclax resistance but not for acquired MCL1 resistance and is not seen in CLL patients after chemotherapy-resistance, BAX is essential for sensitivity toward both venetoclax and MCL1 inhibition. As we found loss of BAX in Richter's syndrome patients after venetoclax failure, we defined BAX-mediated apoptosis to be critical for drug resistance but not for disease progression of CLL into aggressive diffuse large B-cell lymphoma in vivo. A compound screen revealed TRAIL-mediated apoptosis as a target to overcome BAX deficiency. Furthermore, antibody or CAR T cells eliminated venetoclax resistant lymphoma cells, paving a clinically applicable way to overcome venetoclax resistance.


Asunto(s)
Neoplasias Hematológicas , Leucemia Linfocítica Crónica de Células B , Linfoma de Células B Grandes Difuso , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Resistencia a Antineoplásicos/genética , Proteínas Reguladoras de la Apoptosis/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Linfoma de Células B Grandes Difuso/patología , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/genética , Epigénesis Genética
2.
Internist (Berl) ; 62(6): 583-588, 2021 Jun.
Artículo en Alemán | MEDLINE | ID: mdl-33928413

RESUMEN

BACKGROUND: The observation that tumor-infiltrating lymphocytes (TIL) after ex vivo amplification can control tumors in the long term led to the concept of redirecting patients' cytolytic T­cells by a receptor with defined specificity against the tumor. OBJECTIVES: Development of a recombinant receptor-signal molecule (chimeric antigen receptor, CAR) to increase selectivity and enhance anti-tumor immunity. METHODS: Description of a prototype CAR, overview of the modular composition and further development of CAR technology for use in adoptive immune cell therapy. RESULTS: Intensive research over the last two decades has shown how CAR-mediated T­cell activation is influenced by factors such as binding affinity, the epitope of the target antigen, its expression density and accessibility on the tumor cells, as well as by the signaling domains and their combination to induce T­cell activation. The quality and duration of the T­cell response can be specifically modulated by modifying the modular composition of the CAR; CAR T­cells can act as "biopharmaceutical factories" (T-cells redirected for unrestricted cytokine-mediated killing, TRUCK) in the tissue by CAR-mediated release of transgenic therapeutic proteins. CONCLUSION: Adoptive CAR T­cell therapy has shown clinical efficacy in the treatment of hematological malignancies; the treatment of solid tumors, however, is more challenging. Allogeneic CAR T­cell technology is aimed at generating "off-the-shelf" CAR T­cells that are accessible for a large number of patients. A further promising approach is the use of CAR T­cells for other therapeutic applications such as the treatment of autoimmune diseases.


Asunto(s)
Receptores Quiméricos de Antígenos , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Linfocitos T
3.
Blood ; 136(24): 2786-2802, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33301031

RESUMEN

T-cell prolymphocytic leukemia (T-PLL) is a poor-prognostic neoplasm. Differentiation stage and immune-effector functions of the underlying tumor cell are insufficiently characterized. Constitutive activation of the T-cell leukemia 1A (TCL1A) oncogene distinguishes the (pre)leukemic cell from regular postthymic T cells. We assessed activation-response patterns of the T-PLL lymphocyte and interrogated the modulatory impact by TCL1A. Immunophenotypic and gene expression profiles revealed a unique spectrum of memory-type differentiation of T-PLL with predominant central-memory stages and frequent noncanonical patterns. Virtually all T-PLL expressed a T-cell receptor (TCR) and/or CD28-coreceptor without overrepresentation of specific TCR clonotypes. The highly activated leukemic cells also revealed losses of negative-regulatory TCR coreceptors (eg, CTLA4). TCR stimulation of T-PLL cells evoked higher-than-normal cell-cycle transition and profiles of cytokine release that resembled those of normal memory T cells. More activated phenotypes and higher TCL1A correlated with inferior clinical outcomes. TCL1A was linked to the marked resistance of T-PLL to activation- and FAS-induced cell death. Enforced TCL1A enhanced phospho-activation of TCR kinases, second-messenger generation, and JAK/STAT or NFAT transcriptional responses. This reduced the input thresholds for IL-2 secretion in a sensitizer-like fashion. Mice of TCL1A-initiated protracted T-PLL development resembled such features. When equipped with epitope-defined TCRs or chimeric antigen receptors, these Lckpr-hTCL1Atg T cells gained a leukemogenic growth advantage in scenarios of receptor stimulation. Overall, we propose a model of T-PLL pathogenesis in which TCL1A enhances TCR signals and drives the accumulation of death-resistant memory-type cells that use amplified low-level stimulatory input, and whose loss of negative coregulators additionally maintains their activated state. Treatment rationales are provided by combined interception in TCR and survival signaling.


Asunto(s)
Memoria Inmunológica , Leucemia Prolinfocítica de Células T/inmunología , Proteínas Proto-Oncogénicas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Animales , Humanos , Leucemia Prolinfocítica de Células T/genética , Leucemia Prolinfocítica de Células T/patología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Linfocitos T/patología
4.
Oncoimmunology ; 7(1): e1378842, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29296541

RESUMEN

Adoptive therapy with engineered T cells shows promising results in treating patients with malignant disease, but is challenged by incomplete responses and tumor recurrences. Here, we aimed to direct the tumor microenvironment in favor of a successful immune response by local secretion of interleukin (IL-) 12 and IL-18 by sadministered T cells. To this end, we engineered T cells with a melanoma-specific T cell receptor (TCR) and murine IL-12 and/or IL-18 under the control of a nuclear-factor of activated T-cell (NFAT)-sensitive promoter. These T cells produced IL-12 or IL-18, and consequently enhanced levels of IFNγ, following exposure to antigen-positive but not negative tumor cells. Adoptive transfer of T cells with a TCR and inducible (i)IL-12 to melanoma-bearing mice resulted in severe, edema-like toxicity that was accompanied by enhanced levels of IFNγ and TNFα in blood, and reduced numbers of peripheral TCR transgene-positive T cells. In contrast, transfer of T cells expressing a TCR and iIL-18 was without side effects, enhanced the presence of therapeutic CD8+ T cells within tumors, reduced tumor burden and prolonged survival. Notably, treatment with TCR+iIL-12 but not iIL-18 T cells resulted in enhanced intra-tumoral accumulation of macrophages, which was accompanied by a decreased frequency of therapeutic T cells, in particular of the CD8 subset. In addition, when administered to mice, iIL-18 but not iIL-12 demonstrated a favorable profile of T cell co-stimulatory and inhibitory receptors. In conclusion, we observed that treatment with T cells engineered with a TCR and iIL18 T cells is safe and able to skew the tumor microenvironment in favor of an improved anti-tumor T cell response.

5.
Gene Ther ; 23(10): 718-726, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27356950

RESUMEN

Adoptive cell therapy with chimeric antigen receptor (CAR)-modified T cells showed remarkable therapeutic efficacy in the treatment of leukaemia/lymphoma. However, the application to a variety of cancer entities is often constricted by the non-availability of a single chain antibody (scFv), which is usually the targeting domain in a CAR, while antibodies in the natural format are often available. To overcome the limitation, we designed a CAR that uses an antibody in its natural configuration for binding. Such CAR consists of two chains, the immunoglobulin light and heavy chain with their constant regions, whereby the heavy chain is anchored to the membrane and linked to an intracellular signalling domain for T-cell activation. The two chains form a stable heterodimer, a so-called dual chain CAR (dcCAR), and bind with high affinity and in a specific manner to their cognate antigen. By specific binding, the dcCAR activates engineered T cells for the release of pro-inflammatory cytokines and for target cell lysis. We provide evidence by three examples that the dcCAR format is universally applicable and thereby broadens the CAR cell therapy towards a larger variety of targets for which an scFv antibody is not available.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/genética , Animales , Afinidad de Anticuerpos , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Humanos , Ratones , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología
6.
Curr Mol Med ; 13(7): 1079-88, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23116267

RESUMEN

Adoptive cell therapy has shown impressive efficacy to combat cancer in early phase clinical trials, in particular when T cells engineered to specifically target tumor cells were applied. The patient's T cells are genetically equipped with a chimeric antigen receptor (CAR) which allows them to be redirected in a predefined manner towards virtually any target; by using an antibody-derived domain for binding, CAR T cells can be redirected in a major histocompatibility complex (MHC) dependent and independent fashion. The CAR also provides the stimuli required to induce and maintain T cell activation. Recent clinical data sustain the notion that strong costimulation in conjunction with the primary activation signal is crucial for lasting therapeutic efficacy of CAR T cells. However, costimulation is a double-edged sword and the impact of the individual costimuli to optimize T cell activation is still under debate; some general rules are emerging. The review summarizes how costimulation modulates, improves and prolongs the redirected anti-tumor T cell response and how the same costimulatory signals may contribute to unintended side effects including "cytokine storm" and T cell repression. Upcoming strategies to break the activation/repression circle by using CAR's with modified costimulatory signals are also discussed.


Asunto(s)
Inmunoterapia , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/citología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/uso terapéutico , Antígenos CD28/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Complejo Mayor de Histocompatibilidad/inmunología , Neoplasias/genética , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/uso terapéutico , Receptores OX40/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal , Linfocitos T/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
7.
Gene Ther ; 20(2): 177-86, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22378346

RESUMEN

A majority of cancer deaths are because of an uncontrolled relapse of the disease despite initial remission after therapy, asking for strategies to control tumour cells in the long term. Adoptive therapy with chimeric antigen receptor (CAR)-redirected T cells showed promising success in primary tumour elimination; the capacity of such engineered T cells to establish enduring tumour protection is currently a matter of discussion, in particular as most targeted 'tumour-associated antigens' are self-antigens. To address the issue in a clinically relevant model that closely mimics the human situation, we recorded rejection of carcinoembryonic antigen (CEA)-positive pancreatic tumours in the CEA transgenic mouse that expressed CEA as self-antigen in healthy cells of the gastrointestinal tract. Adoptive therapy with CD8(+) T cells, which were redirected by a CEA-specific, low-affinity CAR with CD3ζ endodomain, eliminated CEA(+) tumours in a primary response; cured mice produced an efficient recall response in the long term towards CEA(+) tumour cells upon rechallenge. Secondary tumour rejection was CEA specific, mediated by engineered T cells and did not require host T cells. No toxicity towards healthy tissues with CEA expression was recorded. Data indicate that adoptive therapy with engineered T cells can establish self-antigen-specific tumour protection in the long term without autoimmunity.


Asunto(s)
Carcinoma/prevención & control , Neoplasias Pancreáticas/prevención & control , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Linfocitos T/inmunología , Animales , Complejo CD3/química , Complejo CD3/genética , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Carcinoma/terapia , Línea Celular Tumoral , Terapia Genética , Células HEK293 , Humanos , Inmunoterapia Adoptiva , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/terapia , Estructura Terciaria de Proteína , Linfocitos T/trasplante
8.
Gene Ther ; 18(1): 62-72, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20944680

RESUMEN

Adoptive immunotherapy of cancer using chimeric antigen receptor (CAR)-engineered T cells with redirected specificity showed efficacy in recent trials. In preclinical models, 'second-generation' CARs with CD28 costimulatory domain in addition to CD3ζ performed superior in redirecting T-cell effector functions and survival. Whereas CD28 costimulation sustains physiological T-cell receptor (TCR)-CD3 activation of naïve T cells, the impact of CD28 cosignalling on the threshold of CAR-mediated activation of pre-stimulated T cells without B7-CD28 recruitment remained unclear. Using CARs of different binding affinities, but same epitope specificity, we demonstrate that CD28 cosignalling neither lowered the antigen threshold nor the binding affinity for redirected T-cell activation. 'Affinity ceiling' above which increase in affinity does not increase T-cell activation was not altered. Accordingly, redirected tumor cell killing depended on the binding affinity but was likewise effective for CD3ζ and CD28-CD3ζ CARs. In contrast to CD3ζ, CD28-CD3ζ CAR-driven activation was not increased further by CD28-B7 engagement. However, CD28 cosignalling, which is required for interleukin-2 induction could not be replaced by high-affinity CD3ζ CAR binding or high-density antigen engagement. We conclude that CD28 CAR cosignalling does not alter the activation threshold but redirects T-cell effector functions.


Asunto(s)
Antígenos CD28/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal , Linfocitos T/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Complejo CD3/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Células Tumorales Cultivadas
9.
Hepat Res Treat ; 2010: 901216, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21188203

RESUMEN

More than 350 million people worldwide are persistently infected with human heptatitis B virus (HBV) and at risk to develop liver cirrhosis and hepatocellular carcinoma making long-term treatment necessary. While a vaccine is available and new antiviral drugs are being developed, elimination of persistently infected cells is still a major issue. Recent efforts in adoptive cell therapy are experimentally exploring immunotherapeutic elimination of HBV-infected cells by means of a biological attack with genetically engineered "designer" T cells.

10.
Pathologe ; 31 Suppl 2: 215-20, 2010 Oct.
Artículo en Alemán | MEDLINE | ID: mdl-20730458

RESUMEN

AIMS: To improve survival of patients with advanced rhabdomyosarcomas (RMS), we aimed to adoptively transfer T-cells with redirected specificity for the fetal acetylcholine receptor (AChR), an RMS-specific cell surface antigen. METHODS: A "second generation" chimeric antigen receptor (CAR) with a combined CD28-CD3ζ signaling domain was derived from our previously described chimeric antigen receptor composed of an extracellular human anti-fAChR antibody fragment, an Fc hinge region, and the intracellular T-cell receptor zeta chain. Lymphocytes from the peripheral blood were modified by retroviral transduction and monitored by FACS analysis. Cytotoxicity of modified T-cells towards RMS cells was recorded by MTT-based viability tests; expression of co-stimulatory molecules and anti-apoptotic genes was studied by FACS and qRT-PCR analysis. RESULTS: Co-stimulatory molecules were expressed in low levels on RMS cells giving the rationale to generate a CD28-CD3ζ signalling CAR (chimeric antigen receptor) for redirecting T-cells. T-cells were successfully engineered with the "second generation" AChR-specific chimeric antigen receptor. Despite of high CAR expression engineered T-cells showed low killing efficiency towards RMS compared to redirected killing of CD20+ lymphoma or CEA-expressing adenocarcinoma cell lines when redirected by CD20- and/or CEA-specific CAR. CONCLUSIONS: Data suggest that RMS cells exhibit resistance to a T-cell attack redirected by a fAChR-specific CAR. Inhibition of anti-apoptotic pathways in those cells may improve sensitivity to conventional as well as T-cell-based therapeutics.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Rabdomiosarcoma/terapia , Linfocitos T/inmunología , Línea Celular Tumoral , Quimerismo , Pruebas Inmunológicas de Citotoxicidad , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Colinérgicos/inmunología , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/patología
11.
Gene Ther ; 17(10): 1206-13, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20555360

RESUMEN

Chimeric antigen receptors (CARs, immunoreceptors) are frequently used to redirect T cells with pre-defined specificity, in particular towards tumour cells for use in adoptive immunotherapy of malignant diseases. Specific targeting is mediated by an extracellularly located antibody-derived binding domain, which is joined to the transmembrane and intracellular CD3ζ moiety for T-cell activation. Stable CAR expression in T cells, however, requires a spacer domain interposed between the binding and the transmembrane domain and which is commonly the constant IgG1 Fc domain. We here revealed that CARs with Fc spacer domain bind to IgG Fc gamma receptors (FcγRs), thereby unintentionally activating innate immune cells, including monocytes and natural killer (NK) cells, which consequently secrete high amounts of pro-inflammatory cytokines. Engineered T cells, on the other hand, are likewise activated by FcγR binding resulting in cytokine secretion and lysis of monocytes and NK cells independently of the redirected specificity. To reduce FcγR binding, we modified the spacer domain without affecting CAR expression and antigen binding. Engineered with the modified CAR, T cells are not activated in presence of FcγR(+) cells, thereby minimizing the risk of off-target activation while preserving their redirected targeting specificity.


Asunto(s)
Ingeniería Genética , Inmunidad Innata , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/inmunología , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos/inmunología , Linfocitos T/inmunología , Células Cultivadas , Humanos , Inmunoglobulina G/química , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Estructura Terciaria de Proteína , Receptores de Antígenos/química , Receptores de Antígenos/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/metabolismo , Células Tumorales Cultivadas
12.
Horm Metab Res ; 42(4): 268-73, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20091460

RESUMEN

Low-grade inflammation is important in the development of obesity related pathologies such as insulin resistance and type 2 diabetes, and also cardiovascular disease. Visfatin/PBEF/Nampt and resistin are proinflammatory adipokines secreted from adipocytes, monocytes, and macrophages, and have been linked to atherosclerotic plaque formation, recently. The aim of the present study was to investigate if the expression of these molecules in circulating blood monocytes is altered in obese and/or type 2 diabetic human subjects. Monocytes were isolated by CD14-antibody based magnetic cell sorting from blood samples of 17 lean controls, 20 obese nondiabetic subjects, and 19 obese patients with type 2 diabetes. FACS analysis was performed to test purity of the cell preparations. Expression of the different adipokines was measured by multiplex real-time PCR on RNA-level. Visfatin/PBEF/Nampt was found to be very strongly expressed in monocytes, whereas resistin levels were significantly lower. Furthermore, visfatin/PBEF/Nampt expression was significantly upregulated in obese type 2 diabetic patients, whereas obese nondiabetics exhibited similar levels compared to lean controls, indicating that visfatin/PBEF/Nampt levels are related to type 2 diabetes rather than to obesity. In contrast, resistin expression displayed a different pattern being significantly increased in obese subjects compared to controls but not related to type 2 diabetes. These data suggest a differential role for these two proinflammatory adipokines in linking metabolic diseases to atherosclerosis with visfatin/PBEF/Nampt being more important in patients with type 2 diabetes and resistin in obese but nondiabetic human subjects.


Asunto(s)
Citocinas/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Monocitos/enzimología , Nicotinamida Fosforribosiltransferasa/sangre , Obesidad/sangre , Obesidad/complicaciones , Resistina/sangre , Antropometría , Citocinas/genética , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/genética , Femenino , Glucosa/farmacología , Humanos , Inflamación/sangre , Inflamación/complicaciones , Insulina/farmacología , Receptores de Lipopolisacáridos/metabolismo , Masculino , Persona de Mediana Edad , Nicotinamida Fosforribosiltransferasa/genética , Obesidad/enzimología , Obesidad/genética , Resistina/genética
13.
Gene Ther ; 16(9): 1088-96, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19554034

RESUMEN

Recent insight into the balance of self-tolerance and auto-aggression has raised interest in using human regulatory T (Treg) cells for adoptive immunotherapy of unlimited autoimmune diseases including type-1 diabetes, rhematoid arthritis and multiple sclerosis. The therapeutic use of Treg cells, however, is so far hampered by the inefficiency of current protocols in making them accessible for genetic manipulations. We report here that TCR/CD3 stimulation that is accompanied by extensive CD28 costimulation makes human Treg cells susceptible to retroviral gene transfer ex vivo while preserving their properties in vitro and in vivo. To show the power of genetic manipulation of human Treg cells, we engineered 'designer Treg cells' by retroviral expression of a chimeric immunoreceptor with defined specificity, which activates Treg cells in a ligand-dependent manner to proliferate, to secrete high amounts of interleukin-10 and to repress an ongoing cytolytic T-cell response in vivo. The procedure in genetically modifying human Treg cells ex vivo will open a panel of applications for their use in the adoptive therapy of deregulated immune responses.


Asunto(s)
Ingeniería Genética/métodos , Inmunoterapia Adoptiva/métodos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Apoptosis/inmunología , Línea Celular , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Humanos , Activación de Linfocitos/genética , Ratones , Ratones Desnudos , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Sarcoma Experimental/inmunología , Sarcoma Experimental/patología , Sarcoma Experimental/terapia
14.
Gene Ther ; 16(5): 596-604, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19158846

RESUMEN

Human T lymphocytes can be redirected with a new defined specificity by expression of a chimeric T-cell receptor (immunoreceptor) for the use in adoptive immunotherapy of cancer. Whereas standard procedures use retroviral gene transduction to constitutively express immunoreceptors in T cells, we here explored for the first time mRNA electroporation to achieve transient immunoreceptor expression, and thereby minimizing the risk of persistence of potential autoaggression. CD4(+) and CD8(+) T cells were efficiently transfected with immunoreceptors specific for ErbB2 and CEA. The immunoreceptor expression was transient with half-maximal expression at day 2 and no detectable immunoreceptor expression at day 9 after electroporation. Immunoreceptor-transfected T cells were specifically activated upon coincubation with ErbB2(+) and CEA(+) tumor cells, respectively, resulting in secretion of interferon-gamma (IFNgamma), interleukin-2 (IL-2), and tumor necrosis factor-alpha (TNFalpha). Furthermore, immunoreceptor-transfected CD8(+) T cells specifically lysed ErbB2(+) and CEA(+) tumor cells, respectively. The RNA-transfected T cells retained their cytotoxic function after 2 days of activation and exhibited cytolytic activities like retrovirally transduced T cells. RNA electroporation of T cells thereby provides a versatile tool for transient immunoreceptor expression, which may be of advantage in avoiding the persistence of unintended autoaggression.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Receptores Inmunológicos/biosíntesis , Citotoxicidad Inmunológica , Electroporación , Vectores Genéticos , Humanos , Activación de Linfocitos , Neoplasias/patología , ARN Mensajero/genética , Receptores Inmunológicos/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Retroviridae/genética , Transfección , Células Tumorales Cultivadas
15.
Gut ; 55(8): 1156-64, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16188919

RESUMEN

BACKGROUND AND AIMS: The prognosis of metastatic colorectal cancer is still poor, raising the need for alternative therapeutic approaches, particularly by manipulating the antitumour immune response. Advanced tumour stages, however, are frequently accompanied by functional T cell defects which may be critical for a T cell based anticancer immunotherapy. The aim of this study was to address whether T cells from colorectal cancer patients with advanced tumour stages can be specifically antigen activated against their autologous tumour cells. METHODS: T cells were isolated from colorectal cancer patients and retrovirally transduced to express a recombinant immunoreceptor that has an extracellular binding domain for carcinoembryonic antigen (CEA) and an intracellular CD3zeta signalling domain with and without CD28 costimulation for T cell activation. RESULTS: Peripheral blood T cells from colorectal cancer patients were successfully engineered to express the anti-CEA immunoreceptor on the cell surface. On coincubation with autologous CEA(+) tumour cells, T cells with anti-CEA immunoreceptor are specifically activated to secrete interferon gamma (IFN-gamma) and to lyse autologous tumour cells whereas T cells without immunoreceptor are not. T cells equipped with combined CD3zeta-CD28 signalling receptor are more efficiently activated to secrete IFN-gamma compared with T cells with CD3zeta signalling receptor. Induction of interleukin 2 secretion on targeting towards autologous tumour cells requires triggering of T cells by the CD3zeta-CD28 costimulatory receptor. CONCLUSIONS: T cells from advanced colorectal cancer patients can be tumour specifically activated with high efficiency by engraftment with a combined CD3zeta-CD28 immunoreceptor to break tolerance against autologous tumour cells.


Asunto(s)
Antígenos CD28/inmunología , Complejo CD3/inmunología , Antígeno Carcinoembrionario/inmunología , Neoplasias Colorrectales/inmunología , Linfocitos T/inmunología , Anciano , Técnicas de Cocultivo , Femenino , Humanos , Tolerancia Inmunológica/inmunología , Inmunoterapia/métodos , Interferón gamma/biosíntesis , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Receptores Inmunológicos/inmunología , Proteínas Recombinantes/inmunología , Retroviridae/genética , Transducción de Señal/inmunología , Transducción Genética , Células Tumorales Cultivadas
16.
Gut ; 53(3): 339-45, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14960512

RESUMEN

BACKGROUND AND AIMS: Interleukin-12 (IL-12), a p35/p40 heterodimer, plays a pivotal role in the immune response in Crohn's disease (CD). Since IL-12 p40 dimers act as IL-12 antagonists, we assayed p40 dimer proteins to modulate chronic intestinal inflammation. METHODS: We generated a fusion protein consisting of the IL-12(p40) subunit fused to the constant region of IgG2b. IL-12(p40)-IgG2b was tested in a murine 2,4,6,-trinitrobenzene sulphonic acid (TNBS) colitis model and in lamina propria mononuclear cells (LPMNC) from patients with CD in vitro. RESULTS: Dimeric IL-12(p40)-IgG2b fusion protein bound specifically to the IL-12 receptor. In concentrations <10(-7) M, it acted as an IL-12 antagonist as it inhibited interferon gamma (IFN-gamma) secretion, suppressed proliferation, and increased apoptosis of LPMNC from patients with CD. However, in concentrations >10(-6) M, IL-12(p40)-IgG2b increased IFN-gamma secretion and lymphocyte proliferation thereby acting as an IL-12 agonist. In TNBS colitic mice, IL-12(p40)-IgG2b decreased mortality (10% v 68%), prevented body weight loss, reduced tumour necrosis factor alpha, and increased IL-10 secretion. CONCLUSIONS: The IL-12(p40)-IgG2b fusion protein has dichotomic properties as a specific IL-12 antagonist and selective repressor of mucosal inflammation at low concentration and as an IL-12 agonist at high concentration.


Asunto(s)
Colitis/tratamiento farmacológico , Enfermedad de Crohn/inmunología , Inmunoglobulina G/uso terapéutico , Interleucina-12/uso terapéutico , Subunidades de Proteína/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Adulto , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Colitis/inducido químicamente , Colitis/inmunología , Enfermedad de Crohn/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunoglobulina G/metabolismo , Interferón gamma/biosíntesis , Interleucina-12/metabolismo , Subunidad p40 de la Interleucina-12 , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Estudios Prospectivos , Subunidades de Proteína/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12 , Proteínas Recombinantes de Fusión/metabolismo , Ácido Trinitrobencenosulfónico
17.
Br J Cancer ; 89(6): 1130-9, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12966437

RESUMEN

MUC1 mucin is aberrantly glycosylated and overexpressed in a number of epithelial malignancies and is therefore a promising tumour-associated antigen for target-directed immunotherapy of a panel of malignant diseases. In MUC1-positive tumours, MHC class I expression is frequently downregulated and MUC1-specific cytotoxic T cells (CTLs) are either not available or in a state of anergy allowing tumour growth without limitation by CTL control. To activate lymphocytes and natural killer (NK) cells, we here generated an anti-MUC1-scFv-IL2 fusion protein (C595scFv-Fc-IL2) that contains the C595 single-chain antibody for MUC1 binding, the human IgG1 CH2CH3 domain for protein dimerisation, and interleukin-2 (IL2) for activation of immunological effector cells. The fusion protein binds to MUC1-derived peptides and to MUC1-positive tumour cells with the same specificity as does the C595 monoclonal antibody. Bound to MUC1, the C595scFv-Fc-IL2 fusion protein stimulates proliferation of human activated lymphocytes in vitro. Upon binding to MUC1-positive MCF7 breast carcinoma cells, moreover, the fusion protein activates resting NK cells to tumour cell lysis. These properties make the C595scFv-Fc-IL2 fusion protein a suitable candidate for the immunotherapy of MUC1-positive tumours.


Asunto(s)
Neoplasias de la Mama/patología , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Mucina-1/inmunología , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T Citotóxicos/inmunología , Anticuerpos Monoclonales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Western Blotting , Neoplasias de la Mama/inmunología , División Celular/efectos de los fármacos , Cartilla de ADN/química , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunidad Celular , Activación de Linfocitos/inmunología , Fragmentos de Péptidos/inmunología , Plásmidos , Reacción en Cadena de la Polimerasa , Transfección , Células Tumorales Cultivadas , Vacunas Sintéticas/inmunología
18.
Gene Ther ; 10(17): 1408-19, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12900755

RESUMEN

Recombinant immunoreceptors are modularily composed of extracellular antigen binding and intracellular signalling domains that are preferentially derived from CD3zeta or Fc epsilon RIgamma. The impact of the signalling domain on the stability of immunoreceptor expression and function is not completely understood. To address this issue, we generated and expressed a panel of recombinant zeta- and gamma-chain immunoreceptors, respectively, in human peripheral blood T cells. The expression level of zeta-chain immunoreceptors in human T cells is significantly lower than those of the homologous gamma-chain receptors. Low zeta-chain receptor expression in peripheral T cells is because of the intracellular signalling domain and independent of the Fc epsilon RIgamma or CD3zeta transmembrane region. Expression of both receptors decreases upon prolonged cultivation. Shortly after receptor engraftment, target cell lysis and induction of IFN-gamma secretion are mediated with similar efficiency by zeta- and gamma-chain immunoreceptors. Upon prolonged propagation, however, T-cell activation mediated by zeta-chain immunoreceptors is more efficient than by gamma-chain receptors, indicating that the initial high expression level of gamma-chain immunoreceptors compensates its lower activation capacity. Consequently, gamma-chain immunoreceptors exhibit a higher threshold value for specific activation and are more pronouncedly inhibited by soluble ligand antigen compared to the homologous zeta-chain receptor. These findings have substantial consequences for the design of recombinant immunoreceptors for use in adoptive immunotherapy.


Asunto(s)
Terapia Genética/métodos , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Línea Celular , Células Cultivadas , Ingeniería Genética , Humanos , Cadenas epsilon de Inmunoglobulina/genética , Interferón gamma/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Proteínas Recombinantes/administración & dosificación , Linfocitos T/metabolismo , Células Tumorales Cultivadas
19.
Leukemia ; 16(5): 840-5, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11986945

RESUMEN

In early stages of cutaneous T cell lymphoma (Sézary syndrome) both CD4+CD7- and CD4+CD7+ T cells clonally expand whereas in late stages of the disease CD7- cells are predominant in number, giving rise to the question whether CD7- T cells have a survival advantage in the skin. Galectin-1, a cell-bound lectin, was recently reported to trigger apoptosis in activated CD7+ T cells. Here, we demonstrate that in contrast to activated CD7(+) T cells, quiescent and activated CD69+ CD7- T cells from healthy donors and from Sézary patients are resistant to galectin-1-mediated cell death. CD7- T cells are apoptosis-resistant even during coculture with IFN-gamma-stimulated endothelial cells that constitutively express galectin-1 in high amounts. These data imply that resistance of CD7- T cells to galectin-1-induced apoptosis may contribute to the accumulation of CD7- Sézary T cells during progression of the disease.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antígenos CD7/análisis , Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos , Hemaglutininas/farmacología , Síndrome de Sézary/patología , Adolescente , Adulto , Anciano , Antígenos CD7/efectos de los fármacos , Antígenos CD4/análisis , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Estudios de Casos y Controles , Niño , Femenino , Galectina 1 , Humanos , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Síndrome de Sézary/tratamiento farmacológico , Síndrome de Sézary/inmunología
20.
J Immunol ; 167(11): 6123-31, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11714771

RESUMEN

Recombinant immunoreceptors with specificity for the carcinoembryonic Ag (CEA) can redirect grafted T cells to a MHC/Ag-independent antitumor response. To analyze receptor-mediated cellular activation in the context of CD28 costimulation, we generated: 1) CEA+ colorectal tumor cells that express simultaneously B7-1 and B7-2, and 2) CEA-specific immunoreceptors that harbor intracellularly the signaling moities either of CD28 (BW431/26-scFv-Fc-CD28), CD3zeta (BW431/26-scFv-Fc-CD3zeta), or FcepsilonRIgamma (BW431/26-scFv-Fc-gamma). By retroviral gene transfer, we grafted activated T cells from the peripheral blood with these immunoreceptors. T cells that express the FcepsilonRIgamma or CD3zeta signaling receptor lysed specifically CEA+ tumor cells and secreted high amounts of IFN-gamma upon receptor cross-linking, whereas anti-CEA-CD28 receptor-grafted T cells did not, indicating that CD28 signaling alone is not sufficient for efficient T cell activation. CD28 costimulation did not affect cytolysis by T cells equipped with gamma- or zeta-signaling receptors, but enhanced both IFN-gamma secretion and proliferation. CD28 costimulation, however, was required for efficient IL-2 secretion of anti-CEA-gamma receptor-grafted T cells. Both purified CD4+ and CD8+ T cells grafted with immunoreceptors required CD28 costimulation for complete T cell activation. We integrated both CD28 and CD3zeta signaling domains into one combined immunoreceptor molecule (BW431/26-scFv-Fc-CD28/CD3zeta) with dual signaling properties. T cells grafted with the combined CD28/CD3zeta signaling receptor secreted high amounts of IL-2 upon Ag binding without exogenous B7/CD28 costimulation, demonstrating that both MHC-independent cellular activation and CD28 costimulation for complete T cell activation can be delivered by one recombinant receptor molecule.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos CD28/fisiología , Complejo CD3/fisiología , Interleucina-2/metabolismo , Activación de Linfocitos/genética , Receptores Inmunológicos/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Antígenos de Neoplasias/genética , Antígeno B7-1/biosíntesis , Antígenos CD28/genética , Complejo CD3/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Técnicas de Cocultivo , Epítopos de Linfocito T/genética , Humanos , Receptores de IgG/genética , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/fisiología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/genética , Linfocitos T/metabolismo , Transfección , Células Tumorales Cultivadas/inmunología , Células Tumorales Cultivadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...