Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Blood ; 140(20): 2113-2126, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-35704690

RESUMO

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.


Assuntos
Neoplasias Hematológicas , Leucemia Linfocítica Crônica de Células B , Linfoma Difuso de Grandes Células B , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Reguladoras de Apoptose/genética , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Linfoma Difuso de Grandes Células B/patologia , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/genética , Epigênese Genética
2.
Blood ; 136(24): 2786-2802, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33301031

RESUMO

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.


Assuntos
Memória Imunológica , Leucemia Prolinfocítica de Células T/imunologia , Proteínas Proto-Oncogênicas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Animais , Humanos , Leucemia Prolinfocítica de Células T/genética , Leucemia Prolinfocítica de Células T/patologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Receptores de Antígenos de Linfócitos T/genética , Transdução de Sinais/genética , Linfócitos T/patologia
3.
Internist (Berl) ; 62(6): 583-588, 2021 Jun.
Artigo em Alemão | MEDLINE | ID: mdl-33928413

RESUMO

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.


Assuntos
Receptores de Antígenos Quiméricos , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T
4.
Gene Ther ; 23(10): 718-726, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27356950

RESUMO

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.


Assuntos
Imunoterapia Adotiva/métodos , Ativação Linfocitária , Receptores de Antígenos de Linfócitos T/genética , Animais , Afinidade de Anticorpos , Linhagem Celular Tumoral , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/imunologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia
5.
Gene Ther ; 20(2): 177-86, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22378346

RESUMO

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.


Assuntos
Carcinoma/prevenção & controle , Neoplasias Pancreáticas/prevenção & controle , Complexo Receptor-CD3 de Antígeno de Linfócitos T/genética , Linfócitos T/imunologia , Animais , Complexo CD3/química , Complexo CD3/genética , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Carcinoma/terapia , Linhagem Celular Tumoral , Terapia Genética , Células HEK293 , Humanos , Imunoterapia Adotiva , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/terapia , Estrutura Terciária de Proteína , Linfócitos T/transplante
6.
Gene Ther ; 18(1): 62-72, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20944680

RESUMO

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.


Assuntos
Antígenos CD28/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Complexo CD3/metabolismo , Imunofluorescência , Humanos , Imunoterapia Adotiva , Ativação Linfocitária/imunologia , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Células Tumorais Cultivadas
7.
Gene Ther ; 17(10): 1206-13, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20555360

RESUMO

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.


Assuntos
Engenharia Genética , Imunidade Inata , Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/imunologia , Imunoterapia Adotiva/métodos , Receptores de Antígenos/imunologia , Linfócitos T/imunologia , Células Cultivadas , Humanos , Imunoglobulina G/química , Células Matadoras Naturais/metabolismo , Ativação Linfocitária , Estrutura Terciária de Proteína , Receptores de Antígenos/química , Receptores de Antígenos/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/metabolismo , Células Tumorais Cultivadas
8.
Horm Metab Res ; 42(4): 268-73, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20091460

RESUMO

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.


Assuntos
Citocinas/sangue , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Monócitos/enzimologia , Nicotinamida Fosforribosiltransferase/sangue , Obesidade/sangue , Obesidade/complicações , Resistina/sangue , Antropometria , Citocinas/genética , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/genética , Feminino , Glucose/farmacologia , Humanos , Inflamação/sangue , Inflamação/complicações , Insulina/farmacologia , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Nicotinamida Fosforribosiltransferase/genética , Obesidade/enzimologia , Obesidade/genética , Resistina/genética
9.
Pathologe ; 31 Suppl 2: 215-20, 2010 Oct.
Artigo em Alemão | MEDLINE | ID: mdl-20730458

RESUMO

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.


Assuntos
Imunoterapia Adotiva/métodos , Rabdomiossarcoma/terapia , Linfócitos T/imunologia , Linhagem Celular Tumoral , Quimerismo , Testes Imunológicos de Citotoxicidade , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores Colinérgicos/imunologia , Rabdomiossarcoma/imunologia , Rabdomiossarcoma/patologia
10.
Gene Ther ; 16(9): 1088-96, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19554034

RESUMO

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.


Assuntos
Engenharia Genética/métodos , Imunoterapia Adotiva/métodos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante , Animais , Apoptose/imunologia , Linhagem Celular , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Humanos , Ativação Linfocitária/genética , Camundongos , Camundongos Nus , Receptores Imunológicos/metabolismo , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Sarcoma Experimental/terapia
11.
Gene Ther ; 16(5): 596-604, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19158846

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Receptores Imunológicos/biossíntese , Citotoxicidade Imunológica , Eletroporação , Vetores Genéticos , Humanos , Ativação Linfocitária , Neoplasias/patologia , RNA Mensageiro/genética , Receptores Imunológicos/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Retroviridae/genética , Transfecção , Células Tumorais Cultivadas
12.
J Cell Biol ; 103(3): 795-805, 1986 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3745269

RESUMO

Fusion of mouse L929 cytoplasts with human peripheral blood lymphocytes induced lymphocyte proliferation that gave rise to lymphoid cell lines of B and T cell origin with unlimited growth potential. The immortalized cell lines were routinely grown in standard medium supplemented with fetal calf serum. Furthermore these cell lines could be propagated in chemically defined serum-free media. Each establishment of lymphoid cell lines was preceded by a proliferation phase 2 wk after cytoplast/cell fusion, which appears to be a necessary step in the immortalization process. The immortalized cells have a nearly normal human karyotype, do not form colonies in soft agar medium, and are not tumorigenic in nude mice. Cloned B cell lines produced human immunoglobulins of heavy and light chain types. No cross-reaction with DNA of herpes simplex virus, human cytomegalovirus, human T cell leukemia/lymphoma virus I and II, or polyoma virus was detected in the genome of immortalized cell lines by Southern blot hybridization. Furthermore B and T cell lines were established that appear to be free of Epstein-Barr virus genome.


Assuntos
Células Híbridas/fisiologia , Células L/fisiologia , Linfócitos/fisiologia , Animais , Fusão Celular , Sobrevivência Celular , Citoplasma/fisiologia , DNA Viral/análise , Humanos , Células Híbridas/imunologia , Células Híbridas/transplante , Imunoglobulinas/biossíntese , Camundongos , Camundongos Nus , Neoplasias Experimentais
13.
Oncoimmunology ; 7(1): e1378842, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29296541

RESUMO

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.

14.
Cancer Res ; 58(6): 1116-9, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9515791

RESUMO

Hodgkin's lymphoma patients fail to establish an efficient cellular response against CD30+ Hodgkin/Reed-Sternberg cells. An impaired T-cell receptor/CD3-zeta-mediated activation of T cells is thought to be involved in this situation. We here present a chimeric anti-CD30 receptor that mediates MHC and T-cell receptor/CD3-zeta-independent T-cell activation against CD30+ lymphoma cells even in the presence of soluble CD30. The receptor consists of the binding domain of the monoclonal antibody HRS3 and the signaling unit of the Fc epsilonRI-receptor gamma-chain. After expression in MD45 T cells, receptor cross-linking with immobilized anti-idiotypic monoclonal antibody and CD30+ cells, respectively, results in increased interleukin 2 secretion and specific cytolysis of CD30+ Hodgkin's lymphoma cells. Soluble CD30 in concentrations up to 6000 units/ml did not interfere with cellular activation induced by membrane-bound antigen. This demonstrates the feasibility of the chimeric anti-CD30-scFv-gamma receptor in CD30+ lymphoma cell targeting, even in the presence of as high concentrations of soluble CD30 as are found in patients during progression of the disease.


Assuntos
Complexo CD3/fisiologia , Doença de Hodgkin/imunologia , Antígeno Ki-1/imunologia , Citotoxicidade Imunológica , Relação Dose-Resposta Imunológica , Humanos , Fragmentos de Imunoglobulinas , Antígeno Ki-1/química , Ativação Linfocitária , Proteínas Recombinantes de Fusão , Solubilidade , Linfócitos T Citotóxicos/imunologia
15.
Cancer Res ; 61(5): 1976-82, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280755

RESUMO

Recombinant T-cell receptors with antibody-like specificity are successfully used to direct CTLs toward a MHC-independent immune response against target cells. Here we monitored the specific activation of receptor grafted CTLs in the context of CD28 costimulation. Peripheral blood T cells were retrovirally engrafted with recombinant anti-CD30 and anti-carcinoembryonic antigen receptors, respectively, that harbor either the Fc epsilonRI-gamma or the CD3-zeta intracellular signaling domain. Cross-linking of recombinant receptors by solid-phase bound ligand, i.e., CD30 and a carcinoembryonic antigen receptor-specific anti-idiotypic antibody, respectively, induces IFN-gamma secretion that is further enhanced by CD28 costimulation of grafted T cells. Induction of interleukin (IL)-2 secretion, in contrast, requires CD28 costimulation in addition to receptor cross-linking, irrespective of T-cell preactivation by anti-CD3 monoclonal antibody plus IL-2 or by anti-CD3 monoclonal antibody plus anti-CD28 monoclonal antibody. Accordingly, induction of IL-2 secretion upon receptor cross-linking by membrane-bound antigen requires CD28/B7 costimulation whereas IFN-gamma secretion and cell proliferation does not. The efficiency of cytolysis by receptor-grafted CTLs does not depend on and is not affected by CD28 costimulation. The data demonstrate that CTL proliferation, cytokine secretion, and cytolysis upon receptor cross-linking are differentially modulated by CD28 costimulation and that cytolysis does not require B7 expression on target cells.


Assuntos
Antígenos CD28/imunologia , Interleucina-2/metabolismo , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Superfície Celular , Linfócitos T/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígeno B7-2 , Células CHO , Antígeno Carcinoembrionário/imunologia , Proteínas de Transporte/imunologia , Divisão Celular/imunologia , Técnicas de Cocultura , Cricetinae , Citotoxicidade Imunológica/imunologia , Humanos , Interferon gama/metabolismo , Antígeno Ki-1/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Mutagênese Insercional , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T/biossíntese , Receptores de Antígenos de Linfócitos T/sangue , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Transfecção
16.
Cancer Res ; 57(9): 1704-9, 1997 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9135012

RESUMO

Bispecific CD3 x antitumor antibodies in combination with coactivating CD28 antibodies can induce resting T cells to proliferate and to lyse syngeneic tumor cells (M. Azuma et al., J. Immunol., 150: 2091-2101, 1993; M. Azuma et al., J. Exp. Med., 177: 845-850, 1993). This combination of antibodies may therefore be useful for active immunotherapy of malignant tumors. In this study, we present a preclinical model to evaluate CD3xCD19 bispecific antibodies. We investigated whether bispecific antibodies prevent the development of malignant EBV-induced lymphomas in severe combined immunodeficient (SCID) mice which lack functional B and T lymphocytes (G. C. Bosma et al., Immunogenetics, 29: 54-57, 1989). SCID mice were engrafted (i.p.) with peripheral blood lymphocytes and EBV and treated after 3 days with CD3xCD19 bispecific antibodies and CD28 antibodies. Our data demonstrate that the growth of B cell lymphomas can be prevented in SCID mice by treatment with CD3xCD19 bispecific antibodies and that B-lymphoma-specific T cells can be recruited. In contrast to in vitro experiments, there was no clear effect of CD28 administration which is due to high expression of B7-1 on the transplanted B cells. Lymphoma-bearing mice had elevated titers of interleukin10 in the serum, in contrast to tumor-free animals. As shown by PCR analysis, there was no evidence of dormant B-lymphoma cells in specimens from surviving mice. In the spleen of surviving mice, rearranged human T-cell receptor gamma gene segments were detectable. Furthermore, mice that were initially treated with CD3xCD19 and CD28 antibodies did not develop lymphomas upon rechallenge with EBV-infected mononuclear cells of the same donor, whereas control animals did. Our results obtained from this autologous human B-lymphoma model have implications for the design and evaluation of new immunotherapeutic modalities for the treatment of human B-cell lymphoma with bispecific antibodies.


Assuntos
Antígenos CD19/fisiologia , Antígenos CD28/fisiologia , Complexo CD3/fisiologia , Síndromes de Imunodeficiência/imunologia , Linfoma de Células B/prevenção & controle , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos , DNA Viral/análise , Citometria de Fluxo , Rearranjo Gênico da Cadeia gama dos Receptores de Antígenos dos Linfócitos T , Herpesvirus Humano 4 , Humanos , Imunoterapia/métodos , Linfoma de Células B/microbiologia , Camundongos , Camundongos SCID , Receptores de Antígenos de Linfócitos T gama-delta/genética , Análise de Sobrevida
17.
Cell Death Differ ; 4(8): 824-33, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16465295

RESUMO

Lymphocytes that proliferate autonomously are thought to be arrested at certain steps in differentiation. Here we demonstrate that autocrine proliferating B cells can be induced to terminal differentiation in the presence of ionomycin plus phorbol dibutyrate. The mature CD23high/CD38low B cell phenotype converts to the CD23low/CD38high plasma cell phenotype associated with increased immunoglobulin secretion and PC1 expression and a loss in surface immunoglobulin. Simultaneously, the cells arrest in proliferation and enter apoptosis at day 10. The cytokines IL-1alpha, IL-6, TNFalpha and TNFbeta that are required to sustain continuous growth are secreted in substantially increased amounts mediating entry into apoptosis of the proliferating cells. Decrease in IL-10 secretion sustains this process. Our results draw the concept that once plasmacytoid differentiation is initiated, the growth sustaining network of autocrine cytokines is disturbed in a particular fashion depriving appropriate signals to suppress the differentiation associated apoptotic program.

18.
Cell Death Differ ; 8(4): 395-402, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11550091

RESUMO

The CD7(-) subset of CD4(+) T cells reflects a stable differentiation state of post-thymic helper T cells with CD45R0(+)CD45RA(-) 'memory' phenotype. Here we report that CD4(+)CD7(-) T cells are prone to increased spontaneous apoptosis in vitro compared to CD4(+)CD7(+) T cells. Spontaneous apoptosis is prevented by IL-15, but not by IL-2. Moreover, IL-15 increases Bcl-2 and decreases CD95/Fas expression of CD7(-), but not of CD7(+) T cells. Because IL-15 is physiologically not secreted but expressed in a membrane-bound form, we cocultured T cells with TNF-alpha stimulated fibroblasts that expose membrane IL-15. TNF-alpha stimulated fibroblasts rescue CD4(+)CD7(-) T cells from apoptosis whereas unstimulated fibroblasts do not. Rescue from apoptosis requires cell-cell contact and is abolished by addition of neutralizing antibodies to IL-15. We conclude that membrane IL-15 prevents accelerated apoptosis of CD4(+)CD7(-) T cells. This mechanism may contribute to accumulation of CD7(-) T cells in chronic inflammatory skin lesions.


Assuntos
Antígenos CD7/análise , Apoptose , Linfócitos T CD4-Positivos/imunologia , Dermatite/imunologia , Interleucina-15/farmacologia , Subpopulações de Linfócitos T/imunologia , Apoptose/efeitos dos fármacos , Linfócitos T CD4-Positivos/citologia , Células Cultivadas , Derme/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Humanos , Memória Imunológica , Imunofenotipagem , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Interleucina-15 , Receptores de Interleucina-2/metabolismo , Subpopulações de Linfócitos T/classificação , Subpopulações de Linfócitos T/citologia , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/metabolismo
19.
Leukemia ; 16(5): 840-5, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11986945

RESUMO

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.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos CD7/análise , Apoptose/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Hemaglutininas/farmacologia , Síndrome de Sézary/patologia , Adolescente , Adulto , Idoso , Antígenos CD7/efeitos dos fármacos , Antígenos CD4/análise , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Estudos de Casos e Controles , Criança , Feminino , Galectina 1 , Humanos , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Síndrome de Sézary/tratamento farmacológico , Síndrome de Sézary/imunologia
20.
Eur J Cell Biol ; 65(1): 114-20, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7889982

RESUMO

Deregulated replication of the cellular genome is assumed to result in tumorigenic transformation of the cell. We tested this hypothesis by using mouse DNA elements that promote amplification of transfected DNA in mammalian cells when linked to a selectable marker gene that is driven by a truncated promoter (Holst et al., Cell 52, 355-365 (1988); Wegner et al., Nucl. Acids Res. 17, 9909-9932 (1989)). Here, the DNA elements muNTS-1, e-1, e-2, e-4, e-5, and e-12 were inserted into the plasmid p-hyg, which contains the gene for resistance to hygromycin B driven by a constitutive promoter. After transfer into established rat fibroblasts (208F), transfected DNA constructs persist in low copy numbers (1-10 copies per cell) integrated into high molecular weight DNA. We observed a neoplastically transformed phenotype in 40% to 70% of hygromycin-resistant colonies of 208F cells which is dependent on the DNA element transfected. 208F cells transfected with vector DNA exhibit a "normal" phenotype and are not tumorigenic. The transformed cells, on the other hand, induced malignant tumors after injection into immunodeficient NMRI nu/nu mice. In contrast to established cells, primary rat embryo fibroblasts (REF) with limited life span are neither neoplastically transformed nor immortalized after transfection of e-4 DNA.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Transformação Celular Neoplásica/genética , DNA de Neoplasias/genética , Regiões Promotoras Genéticas , Animais , Fibroblastos/fisiologia , Amplificação de Genes , Camundongos , Camundongos Nus , Ratos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA