Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Ann Oncol ; 35(3): 276-284, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38061428

RESUMO

BACKGROUND: Persisting cancer-related fatigue impairs health-related quality of life (HRQoL) and social reintegration in patients with Hodgkin's lymphoma (HL). The GHSG HD18 trial established treatment de-escalation for advanced-stage HL guided by positron emission tomography after two cycles (PET-2) as new standard. Here, we investigate the impact of treatment de-escalation on long-term HRQoL, time to recovery from fatigue (TTR-F), and time to return to work (TTR-W). PATIENTS AND METHODS: Patients received European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire C30 (EORTC QLQ-C30) and life situation questionnaires at baseline, interim, end of treatment, and yearly follow-up. TTR-F was defined as time from the end of chemotherapy until the first fatigue score <30. TTR-W was analyzed in previously working or studying patients and measured from the end of treatment until the first documented work or education. We compared duration of treatment on TTR-F and TTR-W using Cox proportional hazards regression adjusted for confounding variables. RESULTS: HRQoL questionnaires at baseline were available in 1632 (83.9%) of all randomized patients. Overall, higher baseline fatigue and age were significantly associated with longer TTR-F and TTR-W and male sex with shorter TTR-W. Treatment reduction from eight to four chemotherapy cycles led to a significantly shorter TTR-F [hazard ratio (HR) 1.41, P = 0.008] and descriptively shorter TTR-W (HR 1.24, P = 0.084) in PET-2-negative patients. Reduction from six to four cycles led to non-significant but plausible intermediate accelerations. The addition of rituximab caused significantly slower TTR-F (HR 0.70, P = 0.0163) and TTR-W (HR 0.64, P = 0.0017) in PET-2-positive patients. HRQoL at baseline and age were the main determinants of 2-year HRQoL. CONCLUSIONS: Individualized first-line treatment in patients with advanced-stage HL considerably shortens TTR-F and TTR-W in PET-2-negative patients. Our results support the use of response-adapted shortened treatment duration for patients with HL.


Assuntos
Doença de Hodgkin , Humanos , Masculino , Doença de Hodgkin/patologia , Qualidade de Vida , Retorno ao Trabalho , Fadiga/etiologia , Sobreviventes , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
2.
Mol Cancer ; 21(1): 172, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-36045346

RESUMO

BACKGROUND: Anaplastic large cell lymphoma (ALCL) is an aggressive non-Hodgkin T cell lymphoma commonly driven by NPM-ALK. AP-1 transcription factors, cJUN and JUNb, act as downstream effectors of NPM-ALK and transcriptionally regulate PDGFRß. Blocking PDGFRß kinase activity with imatinib effectively reduces tumor burden and prolongs survival, although the downstream molecular mechanisms remain elusive. METHODS AND RESULTS: In a transgenic mouse model that mimics PDGFRß-driven human ALCL in vivo, we identify PDGFRß as a driver of aggressive tumor growth. Mechanistically, PDGFRß induces the pro-survival factor Bcl-xL and the growth-enhancing cytokine IL-10 via STAT5 activation. CRISPR/Cas9 deletion of both STAT5 gene products, STAT5A and STAT5B, results in the significant impairment of cell viability compared to deletion of STAT5A, STAT5B or STAT3 alone. Moreover, combined blockade of STAT3/5 activity with a selective SH2 domain inhibitor, AC-4-130, effectively obstructs tumor development in vivo. CONCLUSIONS: We therefore propose PDGFRß as a novel biomarker and introduce PDGFRß-STAT3/5 signaling as an important axis in aggressive ALCL. Furthermore, we suggest that inhibition of PDGFRß or STAT3/5 improve existing therapies for both previously untreated and relapsed/refractory ALK+ ALCL patients.


Assuntos
Linfoma Anaplásico de Células Grandes , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Quinase do Linfoma Anaplásico , Animais , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Humanos , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/patologia , Camundongos , Fosforilação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/genética , Transdução de Sinais
3.
Leukemia ; 31(3): 602-613, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27538486

RESUMO

Apart from its unique histopathological appearance with rare tumor cells embedded in an inflammatory background of bystander cells, classical Hodgkin lymphoma (cHL) is characterized by an unusual activation of a broad range of signaling pathways involved in cellular activation. This includes constitutive high-level activity of nuclear factor-κB (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), activator protein-1 (AP-1) and interferon regulatory factor (IRF) transcription factors (TFs) that are physiologically only transiently activated. Here, we demonstrate that inactivation of the putative ubiquitin E3-ligase PDLIM2 contributes to this TF activation. PDLIM2 expression is lost at the mRNA and protein levels in the majority of cHL cell lines and Hodgkin and Reed-Sternberg (HRS) cells of nearly all cHL primary samples. This loss is associated with PDLIM2 genomic alterations, promoter methylation and altered splicing. Reconstitution of PDLIM2 in HRS cell lines inhibits proliferation, blocks NF-κB transcriptional activity and contributes to cHL-specific gene expression. In non-Hodgkin B-cell lines, small interfering RNA-mediated PDLIM2 knockdown results in superactivation of TFs NF-κB and AP-1 following phorbol 12-myristate 13-acetate (PMA) stimulation. Furthermore, expression of PDLIM2 is lost in anaplastic large cell lymphoma (ALCL) that shares key biological aspects with cHL. We conclude that inactivation of PDLIM2 is a recurrent finding in cHL and ALCL, promotes activation of inflammatory signaling pathways and thereby contributes to their pathogenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Doença de Hodgkin/genética , Proteínas com Domínio LIM/genética , Linfoma Anaplásico de Células Grandes/genética , Proteínas dos Microfilamentos/genética , Sequência de Bases , Linhagem Celular Tumoral , Análise por Conglomerados , Metilação de DNA , Ativação Enzimática , Feminino , Inativação Gênica , Loci Gênicos , Doença de Hodgkin/metabolismo , Humanos , Proteínas com Domínio LIM/metabolismo , Linfoma Anaplásico de Células Grandes/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Mutação , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Proteólise , Sítios de Splice de RNA , Fatores de Transcrição , Ubiquitina-Proteína Ligases
5.
Br J Dermatol ; 167(2): 240-6, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22384858

RESUMO

BACKGROUND: Primary cutaneous T-cell lymphomas (CTCLs) are a heterogeneous group with Sézary syndrome (SS) as one of the most aggressive variants. Recently, we identified a loss of E2A as a recurrent event in SS, which enhanced proliferation via upregulation of the proto-oncogene MYC. MYC-induced transformation usually requires deleterious alterations of key apoptotic genes including p53; however, p53 functionality and mutation status in SS are unclear. OBJECTIVES: We investigated functionality of p53 signalling by pharmacological treatment with the MDM2 antagonist nutlin-3, which might result in p53 activation. Furthermore, we analysed the TP53 mutation status in CTCL cell lines and highly purified tumour cells from patients with SS by mRNA and DNA sequencing. METHODS: We analysed the apoptosis induction due to nutlin-3 treatment in various SS cell lines and primary patient samples by annexin V/propidium iodide staining. Induction of p53 target genes was analysed by immunoblotting, and TP53 was sequenced at the mRNA and DNA level. RESULTS: We identified various TP53 mutations and an impaired p53 signalling in the vast majority of the investigated cell lines and primary SS cells. CONCLUSIONS: In accordance with the importance of MYC deregulation in SS, p53 signalling is frequently nonfunctional in SS. However, although most likely ineffective as exclusive treatment in SS, it remains possible that pharmacological p53 activation could be beneficial in combination with other approaches including classical chemotherapeutics.


Assuntos
Genes p53/genética , Mutação/genética , Síndrome de Sézary/genética , Neoplasias Cutâneas/genética , Proteína Supressora de Tumor p53/fisiologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Immunoblotting , Perda de Heterozigosidade , Piperazinas/farmacologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Análise de Sequência de DNA , Análise de Sequência de RNA , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética
6.
Oncogene ; 30(28): 3198-206, 2011 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-21478911

RESUMO

Multiple myeloma (MM) is a paradigm for a malignant disease that exploits external stimuli of the microenvironment for growth and survival. A thorough understanding of the complex interactions between malignant plasma cells and their surrounding requires a detailed analysis of the transcriptional response of myeloma cells to environmental signals. We determined the changes in gene expression induced by interleukin (IL)-6, tumor necrosis factor-α, IL-21 or co-culture with bone marrow stromal cells in myeloma cell lines. Among a limited set of genes that were consistently activated in response to growth factors, a prominent transcriptional target of cytokine-induced signaling in myeloma cells was the gene encoding the serine/threonine kinase serum/glucocorticoid-regulated kinase 1 (SGK1), which is a down-stream effector of PI3-kinase. We could demonstrate a rapid, strong and sustained induction of SGK1 in the cell lines INA-6, ANBL-6, IH-1, OH-2 and MM.1S as well as in primary myeloma cells. Pharmacologic inhibition of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway abolished STAT3 phosphorylation and SGK1 induction. In addition, small hairpin RNA (shRNA)-mediated knock-down of STAT3 reduced basal and induced SGK1 levels. Furthermore, downregulation of SGK1 by shRNAs resulted in decreased proliferation of myeloma cell lines and reduced cell numbers. On the molecular level, this was reflected by the induction of cell cycle inhibitory genes, for example, CDKNA1/p21, whereas positively acting factors such as CDK6 and RBL2/p130 were downregulated. Our results indicate that SGK1 is a highly cytokine-responsive gene in myeloma cells promoting their malignant growth.


Assuntos
Citocinas/farmacologia , Proteínas Imediatamente Precoces/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Proteínas Serina-Treonina Quinases/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Citocinas/metabolismo , Regulação para Baixo/genética , Humanos , Proteínas Imediatamente Precoces/deficiência , Janus Quinases/metabolismo , Mieloma Múltiplo/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Interferência de RNA , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
7.
Oncogene ; 30(15): 1831-40, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21119597

RESUMO

Inappropriate activation of the NOTCH signaling pathway, for example, by activating mutations, contributes to the pathogenesis of various human malignancies. Here, we demonstrate that aberrant expression of an essential NOTCH coactivator of the Mastermind-like (MAML) family provides an alternative mechanism to activate NOTCH signaling in human lymphoma cells. We detected high-level MAML2 expression in several B cell-derived lymphoma types, including classical Hodgkin lymphoma (cHL) cells, relative to normal B cells. Inhibition of MAML-protein activity by a dominant negative form of MAML or by small hairpin RNAs targeting MAML2 in cHL cells resulted in downregulation of the NOTCH target genes HES7 and HEY1, which we identified as overexpressed in cHL cells, and in reduced proliferation. Furthermore, a NOTCH gene-expression signature in cHL cells confirmed their cell-autonomous NOTCH activity. Finally, in line with the essential role of MAML proteins for assembly and activity of the NOTCH transcriptional complex (NTC), we show that MAML-derived small-peptide constructs block NOTCH activity and disrupt NTC formation in vitro. These data strongly suggest direct targeting of the NTC as treatment strategy for NOTCH-dependent malignancies.


Assuntos
Proteínas de Ligação a DNA/genética , Linfoma/metabolismo , Proteínas Nucleares/genética , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Linhagem Celular , Humanos , Linfoma/patologia , Transativadores
8.
Dtsch Med Wochenschr ; 134(39): 1944-8, 2009 Sep.
Artigo em Alemão | MEDLINE | ID: mdl-19760557

RESUMO

Despite the fact that classical Hodgkin lymphoma (HL) has been described more than 170 years ago, only over the last 15 years significant advances regarding its molecular pathogenesis have been achieved. The lack of a specific lineage profile in combination with the low number of the malignant mononuclear Hodgkin- and multinucleated Reed-Sternberg- (HRS-) cells in the affected lymph nodes prevented for a long time both the identification of its cell of origin and of genomic and molecular defects. The development of methods for the analysis of micromanipulated single cells made it possible to demonstrate a B cell origin of HRS cells. However, it has become clear that the normal B cell-specific gene expression program in HRS cells is disrupted by various molecular lesions. Furthermore, molecular and genomic defects of various signaling pathways could be identified in HRS cells, including the NF-kappaB, JAK/STAT and MAPK-AP-1 signaling pathways, which protect HRS cells from apoptotic cell death. Despite significant advances in the treatment of HL, the considerable long term toxicity of conventional therapies requires the development of new non-genotoxic therapeutic strategies. Therefore, it will be a central aim to develop new treatment strategies based on these insights into HL pathogenesis.


Assuntos
Doença de Hodgkin/genética , Apoptose/fisiologia , Linfócitos B/fisiologia , Citocinas/fisiologia , Doença de Hodgkin/patologia , Doença de Hodgkin/terapia , Humanos , NF-kappa B/fisiologia , Células de Reed-Sternberg/patologia , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Fatores de Transcrição STAT/fisiologia , Transdução de Sinais
10.
Leukemia ; 22(8): 1587-94, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18449208

RESUMO

Plasticity of committed mouse B cells has been demonstrated by inactivation of the B-cell commitment transcription factor PAX5, resulting in loss of the B-cell phenotype and differentiation into various hematopoietic lineages. Furthermore, mature mouse B cells could be reprogrammed into macrophages by overexpression of myeloid-specific transcription factors. Here, we report that aberrant activity of the transmembrane receptor, Notch1, interferes with the B-lymphoid phenotype of mature human germinal center-derived B cells in Hodgkin lymphoma, so called Hodgkin and Reed-Sternberg cells. They have lost the B-cell phenotype despite their mature B-cell origin. Notch1 remodels the B-cell transcription factor network by antagonizing the key transcription factors E2A and early B-cell factor (EBF). Through this mechanism, B lineage-specific genes were suppressed and B lineage-inappropriate genes were induced. We provide evidence that absence of the Notch inhibitor Deltex1 contributes to deregulated Notch activity in Hodgkin and Reed-Sternberg cells. These data suggest that Notch activation interferes with dedifferentiation of neoplastic B cells in Hodgkin lymphoma.


Assuntos
Linfócitos B/imunologia , Doença de Hodgkin/patologia , Receptor Notch1/metabolismo , Linfócitos B/citologia , Sequência de Bases , Diferenciação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Primers do DNA , Doença de Hodgkin/imunologia , Doença de Hodgkin/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Fator de Transcrição PAX5/metabolismo , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Blood ; 97(9): 2798-807, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11313274

RESUMO

Constitutively activated nuclear factor (NF)-kappaB is observed in a variety of neoplastic diseases and is a hallmark of the malignant Hodgkin and Reed-Sternberg cells (H/RS) in Hodgkin lymphoma. Given the distinctive role of constitutive NF-kappaB for H/RS cell viability, NF-kappaB-dependent target genes were searched for by using adenoviral expression of the super-repressor IkappaBDeltaN. A surprisingly small but characteristic set of genes, including the cell-cycle regulatory protein cyclin D2, the antiapoptotic proteins Bfl-1/A1, c-IAP2, TRAF1, and Bcl-x(L), and the cell surface receptors CD86 and CD40 were identified. Thus, constitutive NF-kappaB activity maintains expression of a network of genes, which are known for frequent, marker-like expression in primary or cultured H/RS cells. Intriguingly, CD40, which is able to activate CD86 or Bcl-x(L) via NF-kappaB, is itself transcriptionally regulated by NF-kappaB through a promoter proximal binding site. NF-kappaB inhibition resulted in massive spontaneous and p53-independent apoptosis, which could be rescued by ectopic expression of Bcl-x(L), underscoring its dominant role in survival of H/RS cells. Hence, NF-kappaB controls a signaling network in H/RS cells, which promotes tumor cell growth and confers resistance to apoptosis.


Assuntos
Antígenos CD/genética , Apoptose/genética , Antígenos CD40/genética , Glicoproteínas de Membrana/genética , NF-kappa B/genética , Células de Reed-Sternberg/patologia , Células de Reed-Sternberg/fisiologia , Antígenos CD/metabolismo , Antígeno B7-2 , Antígenos CD40/metabolismo , Regulação Neoplásica da Expressão Gênica , Doença de Hodgkin/genética , Doença de Hodgkin/patologia , Humanos , Glicoproteínas de Membrana/metabolismo , NF-kappa B/biossíntese
12.
Cancer Res ; 60(24): 7170-6, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11156427

RESUMO

Clinical administration of the anti-CD20 antibody IDEC-C2B8 can induce remission of low-grade B-cell lymphoma. Whereas it has been suggested that the main mechanisms of action are complement-mediated and antibody-dependent cell-mediated cytotoxicity, we demonstrate that monoclonal antibody IDEC-C2B8 is a strong inducer of apoptosis in CD20-positive B-cell lymphoma cell lines reflecting different stages of lymphomagenesis. Thus, CD20-dependent apoptosis was inducible in human surface IgM-positive Burkitt's lymphoma cell lines as well as in more mature surface IgM-negative B-cell lymphoma cell lines carrying the t(14;18) translocation. Furthermore, in Burkitt's lymphoma cell lines, we observed a striking correlation between anti-CD20- and B-cell receptor-mediated apoptosis with regard to sensitivity toward the apoptotic stimuli and the execution of the apoptotic pathway. Thus, induction of anti-CD20- or B-cell receptor-mediated apoptosis involved rapid up-regulation of the proapoptotic protein Bax. In addition, we show similar changes in the mRNA expression level of two early response genes, c-myc and Berg36, as well as activation of the mitogen-activated protein kinase family members p44 (extracellular signal-regulated kinase 1) and p42 (extracellular signal-regulated kinase 2) and activation of activator protein 1 (AP-1) DNA binding activity. These data support our hypothesis that both pathways are mediated in part by the same signal-transducing molecules. These results might help explain the resistance and regression of lymphomas to IDEC-C2B8 and give new insights in the signaling cascade after CD20 ligation.


Assuntos
Antígenos CD20/metabolismo , Apoptose , Linfócitos B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Receptores de Antígenos de Linfócitos B/metabolismo , Northern Blotting , Linfoma de Burkitt/metabolismo , Fator 1 de Resposta a Butirato , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Proteínas Imediatamente Precoces/metabolismo , Immunoblotting , Imunoglobulina M/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator de Transcrição Sp1/metabolismo , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo , Translocação Genética , Células Tumorais Cultivadas , Regulação para Cima , Proteína X Associada a bcl-2
13.
Blood ; 94(9): 3129-34, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10556199

RESUMO

The transcription factor NF kappa B (NF-kappaB) mediates the expression of numerous genes involved in diverse functions such as inflammation, immune response, apoptosis, and cell proliferation. We recently identified constitutive activation of NF-kappaB (p50/p65) as a common feature of Hodgkin/Reed-Sternberg (HRS) cells preventing these cells from undergoing apoptosis and triggering proliferation. To examine possible alterations in the NF-kappaB/IkappaB system, which might be responsible for constitutive NF-kappaB activity, we have analyzed the inhibitor I kappa B alpha (IkappaBalpha) in primary and cultured HRS cells on protein, mRNA, and genomic levels. In lymph node biopsy samples from Hodgkin's disease patients, IkappaBalpha mRNA proved to be strongly overexpressed in the HRS cells. In 2 cell lines (L428 and KM-H2), we detected mutations in the IkappaBalpha gene, resulting in C-terminally truncated proteins, which are presumably not able to inhibit NF-kappaB-DNA binding activity. Furthermore, an analysis of the IkappaBalpha gene in single HRS cells micromanipulated from frozen tissue sections showed a monoallelic mutation in 1 of 10 patients coding for a comparable C-terminally truncated IkappaBalpha protein. We suggest that the observed IkappaBalpha mutations contribute to constitutive NF-kappaB activity in cultured and primary HRS cells and are therefore involved in the pathogenesis of these Hodgkin's disease (HD) patients. The demonstrated constitutive overexpression of IkappaBalpha in HRS cells evidences a deregulation of the NF-kappaB/IkappaB system also in the remaining cases, probably due to defects in other members of the IkappaB family.


Assuntos
Regulação Neoplásica da Expressão Gênica , Doença de Hodgkin/genética , NF-kappa B/genética , Células de Reed-Sternberg/metabolismo , Sequência de Bases , Doença de Hodgkin/metabolismo , Doença de Hodgkin/patologia , Humanos , Dados de Sequência Molecular , Mutação , NF-kappa B/biossíntese , RNA Mensageiro/análise , RNA Mensageiro/genética , Alinhamento de Sequência
14.
Anticancer Res ; 17(3C): 2025-32, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9216660

RESUMO

The poor prognosis of advanced gastric carcinoma requires new therapeutic approaches. Among these, the specific activation of resting lymphocytes by bispecific antibodies may be promising. Here we describe the generation and function of a bispecific monoclonal antibody (bi-mab) with specificity for CD3 and for the tumor antigen CA72-4 (TAG72) on various gastrointestinal tumors. We established a hybrid/hybridoma by somatic fusion of two hybridoma lines secreting antibodies against CA72-4 and CD3 respectively and characterized its bimab OKT3/B72.3. In combination with costimulatory anti-CD28 antibodies resting peripheral lymphocytes could be activated specifically by bi-mab OKT3/B72.3 with T cell proliferation and IL-2 secretion. The bi-mab OKT3/B72.3 could also trigger the cytotoxicity of these T cells toward, CA72-4+ cells in vitro. Our results indicate, that bi-mab OKT3/B72.3 in combination with an anti-CD28 mab can redirect T cell cytotoxicity specifically against CA72-4+ tumor cells implicating a novel strategy for the specific immunotherapy of CA72-4+ tumors.


Assuntos
Anticorpos Biespecíficos , Anticorpos Monoclonais , Antígenos Glicosídicos Associados a Tumores/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Citotoxicidade Imunológica , Ativação Linfocitária , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/isolamento & purificação , Anticorpos Monoclonais/isolamento & purificação , Células Cultivadas , Humanos , Hibridomas , Interleucina-2/biossíntese , Camundongos , Muromonab-CD3 , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA