Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Cell Death Dis ; 13(3): 274, 2022 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-35347108

RESUMO

Over the past decade, immunotherapy delivered novel treatments for many cancer types. However, lung cancer still leads cancer mortality, and non-small-cell lung carcinoma patients with mutant EGFR cannot benefit from checkpoint inhibitors due to toxicity, relying only on palliative chemotherapy and the third-generation tyrosine kinase inhibitor (TKI) osimertinib. This new drug extends lifespan by 9-months vs. second-generation TKIs, but unfortunately, cancers relapse due to resistance mechanisms and the lack of antitumor immune responses. Here we explored the combination of osimertinib with anti-HER3 monoclonal antibodies and observed that the immune system contributed to eliminate tumor cells in mice and co-culture experiments using bone marrow-derived macrophages and human PBMCs. Osimertinib led to apoptosis of tumors but simultaneously, it triggered inositol-requiring-enzyme (IRE1α)-dependent HER3 upregulation, increased macrophage infiltration, and activated cGAS in cancer cells to produce cGAMP (detected by a lentivirally transduced STING activity biosensor), transactivating STING in macrophages. We sought to target osimertinib-induced HER3 upregulation with monoclonal antibodies, which engaged Fc receptor-dependent tumor elimination by macrophages, and STING agonists enhanced macrophage-mediated tumor elimination further. Thus, by engaging a tumor non-autonomous mechanism involving cGAS-STING and innate immunity, the combination of osimertinib and anti-HER3 antibodies could improve the limited therapeutic and stratification options for advanced stage lung cancer patients with mutant EGFR.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Acrilamidas , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Endorribonucleases , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Mutação , Recidiva Local de Neoplasia/tratamento farmacológico , Nucleotidiltransferases , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases
3.
Oncogene ; 35(23): 2949-60, 2016 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-26434585

RESUMO

The epidermal growth factor receptor (EGFR) and a coreceptor denoted HER2/ERBB2 are frequently overexpressed or mutated in solid tumors, such as carcinomas and gliomas. In line with driver roles, cancer drugs intercepting EGFR or HER2 currently outnumber therapies targeting other hubs of signal transduction. To explain the roles for EGFR and HER2 as prime drivers and targets, we take lessons from invertebrates and refer to homeostatic regulation of several mammalian tissues. The model we infer ascribes to the EGFR-HER2 module pivotal functions in rapid clonal expansion of progenitors called transient amplifying cells (TACs). Accordingly, TACs of tumors suffer from replication stress, and hence accumulate mutations. In addition, several lines of evidence propose that in response to EGF and related mitogens, TACs might undergo dedifferentiation into tissue stem cells, which might enable entry of oncogenic mutations into the stem cell compartment. According to this view, antibodies or kinase inhibitors targeting EGFR-HER2 effectively retard some solid tumors because they arrest mutation-enriched TACs and possibly inhibit their dedifferentiation. Deeper understanding of the EGFR-HER2 module and relations between cancer stem cells and TACs will enhance our ability to control a broad spectrum of human malignancies.


Assuntos
Carcinoma/enzimologia , Receptores ErbB/metabolismo , Células-Tronco/enzimologia , Animais , Carcinoma/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Receptores ErbB/genética , Humanos , Transdução de Sinais
4.
Oncogene ; 35(4): 438-47, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-25915843

RESUMO

Growth factors of the epidermal growth factor (EGF)/neuregulin family are involved in tumor progression and, accordingly, antibodies that intercept a cognate receptor, epidermal growth factor receptor (EGFR)/ERBB1, or a co-receptor, HER2, have been approved for cancer therapy. Although they might improve safety and delay onset of chemoresistance, no anti-ligand antibodies have been clinically approved. To identify suitable ligands, we surveyed fluids from ovarian and lung cancer patients and found that amphiregulin (AREG) is the most abundant and generalized ligand secreted by advanced tumors. AREG is a low affinity EGFR ligand, which is upregulated following treatment with chemotherapeutic drugs. Because AREG depletion retarded growth of xenografted ovarian tumors in mice, we generated a neutralizing monoclonal anti-AREG antibody. The antibody inhibited growth of ovarian cancer xenografts and strongly enhanced chemotherapy efficacy. Taken together, these results raise the possibility that AREG and other low- or high-affinity binders of EGFR might serve as potential targets for cancer therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Família de Proteínas EGF/genética , Família de Proteínas EGF/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Anfirregulina , Animais , Anticorpos Monoclonais/imunologia , Antineoplásicos/farmacologia , Meios de Cultivo Condicionados/análise , Família de Proteínas EGF/imunologia , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Nus , Terapia de Alvo Molecular/métodos , Neoplasias Ovarianas/genética , Fator de Crescimento Transformador alfa/metabolismo , Fator de Crescimento Transformador alfa/farmacologia , Células Tumorais Cultivadas , Ubiquitinação , Ensaios Antitumorais Modelo de Xenoenxerto
5.
CPT Pharmacometrics Syst Pharmacol ; 4(12): 720-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26904386

RESUMO

We present a system of nonlinear ordinary differential equations used to quantify the complex dynamics of the interactions between tumor growth, vasculature generation, and antiangiogenic treatment. The primary dataset consists of longitudinal tumor size measurements (1,371 total observations) in 105 colorectal tumor-bearing mice. Mice received single or combination administration of sunitinib, an antiangiogenic agent, and/or irinotecan, a cytotoxic agent. Depending on the dataset, parameter estimation was performed either using a mixed-effect approach or by nonlinear least squares. Through a log-likelihood ratio test, we conclude that there is a potential synergistic interaction between sunitinib when administered in combination with irinotecan in preclinical settings. Model simulations were then compared to data from a follow-up preclinical experiment. We conclude that the model has predictive value in identifying the therapeutic window in which the timing between the administrations of these two drugs is most effective.

6.
Ann Oncol ; 25(4): 773-780, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24351401

RESUMO

Insights into tumour biology of breast cancer have led the path towards the introduction of targeted treatment approaches; still, breast cancer-related mortality remains relatively high. Efforts in the field of basic research revealed new druggable targets which now await validation within the context of clinical trials. Therefore, questions concerning the optimal design of future studies are becoming even more pertinent. Aspects such as the ideal end point, availability of predictive markers to identify the optimal cohort for drug testing, or potential mechanisms of resistance need to be resolved. An expert panel representing the academic community, the pharmaceutical industry, as well as European Regulatory Authorities met in Vienna, Austria, in November 2012, in order to discuss breast cancer biology, identification of novel biological targets and optimal drug development with the aim of treatment individualization. This article summarizes statements and perspectives provided by the meeting participants.


Assuntos
Receptor ErbB-2/genética , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/terapia , Ensaios Clínicos como Assunto , Feminino , Humanos , Terapia de Alvo Molecular , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/classificação , Neoplasias de Mama Triplo Negativas/genética
7.
Oncogene ; 31(43): 4599-608, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-22179831

RESUMO

Once stimulated, the epidermal growth factor receptor (EGFR) undergoes self-phosphorylation, which, on the one hand, instigates signaling cascades, and on the other hand, recruits CBL ubiquitin ligases, which mark EGFRs for degradation. Using RNA interference screens, we identified a deubiquitinating enzyme, Cezanne-1, that opposes receptor degradation and enhances EGFR signaling. These functions require the catalytic- and ubiquitin-binding domains of Cezanne-1, and they involve physical interactions and transphosphorylation of Cezanne-1 by EGFR. In line with the ability of Cezanne-1 to augment EGF-induced growth and migration signals, the enzyme is overexpressed in breast cancer. Congruently, the corresponding gene is amplified in approximately one third of mammary tumors, and high transcript levels predict an aggressive disease course. In conclusion, deubiquitination by Cezanne-1 curtails degradation of growth factor receptors, thereby promotes oncogenic growth signals.


Assuntos
Endopeptidases/metabolismo , Receptores ErbB/metabolismo , Neoplasias/patologia , Catálise , Progressão da Doença , Humanos , Neoplasias/metabolismo , Fosforilação , RNA Interferente Pequeno , Ubiquitina/metabolismo , Ubiquitinação
8.
Oncogene ; 31(30): 3505-15, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22105361

RESUMO

Epidermal growth factor (EGF)-like growth factors control tumor progression as well as evasion from the toxic effects of chemotherapy. Accordingly, antibodies targeting the cognate receptors, such as EGFR/ErbB-1 and the co-receptor HER2/ErbB-2, are widely used to treat cancer patients, but agents that target the EGF-like growth factors are not available. To circumvent the existence of 11 distinct ErbB ligands, we constructed a soluble fusion protein (hereinafter: TRAP-Fc) comprising truncated extracellular domains of EGFR/ErbB-1 and ErbB-4. The recombinant TRAP-Fc retained high-affinity ligand binding to EGF-like growth factors and partially inhibited growth of a variety of cultured tumor cells. Consistently, TRAP-Fc displayed an inhibitory effect in xenograft models of human cancer, as well as synergy with chemotherapy. Additionally, TRAP-Fc inhibited invasive growth of mammary tumor cells and reduced their metastatic seeding in the lungs of animals. Taken together, the activities displayed by TRAP-Fc reinforce critical roles of EGF-like growth factors in tumor progression, and they warrant further tests of TRAP-Fc in preclinical models.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Receptores ErbB/química , Neoplasias Pulmonares/tratamento farmacológico , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/secundário , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Camundongos SCID , Receptor ErbB-2/química , Receptor ErbB-4 , Proteínas Recombinantes de Fusão/química , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Oncogene ; 31(7): 907-17, 2012 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21743488

RESUMO

A large fraction of ductal carcinoma in situ (DCIS), a non-invasive precursor lesion of invasive breast cancer, overexpresses the HER2/neu oncogene. The ducts of DCIS are abnormally filled with cells that evade apoptosis, but the underlying mechanisms remain incompletely understood. We overexpressed HER2 in mammary epithelial cells and observed growth factor-independent proliferation. When grown in extracellular matrix as three-dimensional spheroids, control cells developed a hollow lumen, but HER2-overexpressing cells populated the lumen by evading apoptosis. We demonstrate that HER2 overexpression in this cellular model of DCIS drives transcriptional upregulation of multiple components of the Notch survival pathway. Importantly, luminal filling required upregulation of a signaling pathway comprising Notch3, its cleaved intracellular domain and the transcriptional regulator HES1, resulting in elevated levels of c-MYC and cyclin D1. In line with HER2-Notch3 collaboration, drugs intercepting either arm reverted the DCIS-like phenotype. In addition, we report upregulation of Notch3 in hyperplastic lesions of HER2 transgenic animals, as well as an association between HER2 levels and expression levels of components of the Notch pathway in tumor specimens of breast cancer patients. Therefore, it is conceivable that the integration of the Notch and HER2 signaling pathways contributes to the pathophysiology of DCIS.


Assuntos
Neoplasias da Mama/genética , Carcinoma Intraductal não Infiltrante/genética , Receptor ErbB-2/genética , Receptores Notch/genética , Animais , Neoplasias da Mama/metabolismo , Carcinoma Intraductal não Infiltrante/metabolismo , Linhagem Celular , Proliferação de Células , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Immunoblotting , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Receptor ErbB-2/metabolismo , Receptor Notch3 , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transfecção
10.
Oncogene ; 31(31): 3569-83, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22139081

RESUMO

The HER2/neu oncogene encodes a receptor-like tyrosine kinase whose overexpression in breast cancer predicts poor prognosis and resistance to conventional therapies. However, the mechanisms underlying aggressiveness of HER2 (human epidermal growth factor receptor 2)-overexpressing tumors remain incompletely understood. Because it assists epidermal growth factor (EGF) and neuregulin receptors, we overexpressed HER2 in MCF10A mammary cells and applied growth factors. HER2-overexpressing cells grown in extracellular matrix formed filled spheroids, which protruded outgrowths upon growth factor stimulation. Our transcriptome analyses imply a two-hit model for invasive growth: HER2-induced proliferation and evasion from anoikis generate filled structures, which are morphologically and transcriptionally analogous to preinvasive patients' lesions. In the second hit, EGF escalates signaling and transcriptional responses leading to invasive growth. Consistent with clinical relevance, a gene expression signature based on the HER2/EGF-activated transcriptional program can predict poorer prognosis of a subgroup of HER2-overexpressing patients. In conclusion, the integration of a three-dimensional cellular model and clinical data attributes progression of HER2-overexpressing lesions to EGF-like growth factors acting in the context of the tumor's microenvironment.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Modelos Biológicos , Receptor ErbB-2/fisiologia , Anoikis/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/patologia , Matriz Extracelular/fisiologia , Feminino , Perfilação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Invasividade Neoplásica , Lesões Pré-Cancerosas/patologia , Esferoides Celulares/fisiologia , Transcrição Gênica/fisiologia
11.
Oncogene ; 30(14): 1631-42, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21132012

RESUMO

Monoclonal antibodies (mAbs) to HER2 are currently used to treat breast cancer, but low clinical efficacy, along with primary and acquired resistance to therapy, commonly limit clinical applications. We previously reported that combinations of antibodies directed at non-overlapping epitopes of HER2 are endowed with enhanced antitumor effects, probably due to accelerated receptor degradation. Here, we extend these observations to three-dimensional mammary cell models, and compare the effects of single mAbs with the effects of antibody combinations. Collectively, our in vitro assays and computational image analyses indicate that combining mAbs against different epitopes of HER2 better inhibits invasive growth. Importantly, while growth factors are able to reduce intraluminal apoptosis and induce an invasive phenotype, combinations of mAbs better than single mAbs can reverse the growth factor-induced phenotypes of HER2-overexpressing spheroids. In conclusion, our studies propose that mAb combinations negate the biological effects of growth factors on invasive growth of HER2-overexpressing cells. Hence, combining mAbs offers a therapeutic strategy, potentially able to enhance clinical efficacy of existing antireceptor immunotherapeutics.


Assuntos
Anticorpos Monoclonais/imunologia , Antineoplásicos/imunologia , Neoplasias da Mama/terapia , Carcinoma Ductal de Mama/terapia , Epitopos/imunologia , Receptor ErbB-2/imunologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/imunologia , Carcinoma Ductal de Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas Quimioatraentes de Monócitos/imunologia
12.
Oncogene ; 29(43): 5850-60, 2010 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-20676128

RESUMO

Tumor cells often subvert normal regulatory mechanisms of signal transduction. This study shows this principle by studying yet uncharacterized mutants of the epidermal growth factor receptor (EGFR) previously identified in glioblastoma multiforme, which is the most aggressive brain tumor in adults. Unlike the well-characterized EGFRvIII mutant form, which lacks a portion of the ligand-binding cleft within the extracellular domain, EGFRvIVa and EGFRvIVb lack internal segments distal to the intracellular tyrosine kinase domain. By constructing the mutants and by ectopic expression in naive cells, we show that both mutants confer an oncogenic potential in vitro, as well as tumorigenic growth in animals. The underlying mechanisms entail constitutive receptor dimerization and basal activation of the kinase domain, likely through a mechanism that relieves a restraining molecular fold, along with stabilization due to association with HSP90. Phosphoproteomic analyses delineated the signaling pathways preferentially engaged by EGFRvIVb-identified unique substrates. This information, along with remarkable sensitivities to tyrosine kinase blockers and to a chaperone inhibitor, proposes strategies for pharmacological interception in brain tumors harboring EGFRvIV mutations.


Assuntos
Neoplasias Encefálicas/genética , Transformação Celular Neoplásica/genética , Genes erbB-1/genética , Glioblastoma/genética , Mutação , Transdução de Sinais/genética , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Feminino , Humanos , Imunoprecipitação , Espectrometria de Massas , Camundongos , Camundongos Nus , Células NIH 3T3
13.
IET Syst Biol ; 2(1): 1-4, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18248079

RESUMO

Receptor tyrosine kinases (RTKs) are transmembrane allosteric enzymes: binding of ligand growth factors to their ectodomains stimulates a cytoplasm-facing tyrosine kinase activity, which initiates a plethora of cellular processes. The enormous complexity of RTK signalling, along with rich involvement in pathologies (e.g. cancer and diabetes), motivated the establishment of the international, multi-disciplinary RTK consortium (http://www.rtkconsort.org/) in 2005. In collaboration with the British Society for Proteome Research and the European Bioinformatics Institute, the Consortium held on July 23rd and 24th a Workshop on Proteomics and Phosphoproteomics of RTK Signalling Networks (Hinxton Hall Conference Centre, Cambridge, UK). As highlighted below, systems control (a layered web of regulatory loops summarised in Fig.1) emerged throughout the workshop as a common theme of many presentations.


Assuntos
Retroalimentação/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Processamento de Proteína Pós-Traducional/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos
14.
Oncogene ; 26(48): 6905-14, 2007 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-17486069

RESUMO

Cleavable isoforms of the ErbB4 receptor tyrosine kinase release a soluble intracellular domain (ICD) that may translocate to the nucleus and regulate signaling. However, ErbB4 gene is alternatively spliced generating CYT-1 and CYT-2 isoforms with different cytoplasmic tails. Here, we addressed whether the two alternative ErbB4 ICDs of either CYT-1 (ICD1) or CYT-2 (ICD2) type differ in signaling to the nucleus. Confocal microscopy and extraction of nuclear cell fractions indicated that significantly more ICD2 translocated to the nuclei when compared to ICD1. Unlike the membrane-anchored 80 kDa fragments derived from full-length ErbB4 isoforms, the two ICDs did not differ from each other in metabolic stability or ubiquitylation. However, ICD2 was phosphorylated at tyrosine residues to a higher extent and demonstrated greater in vitro kinase activity than ICD1. Mutating the ATP-binding site within ICD2 kinase domain (ICD2 K751R) blocked its tyrosine phosphorylation and significantly reduced its nuclear translocation. When expressed in the context of full-length ErbB4, ICD2 was also more efficient than ICD1 in promoting transcriptional activation of the STAT5 target gene beta-casein. These findings indicate that the two alternative ICDs of ErbB4 differ in their nuclear accumulation, and that the mechanism involves differential kinase activity but not ubiquitin-regulated ICD stability.


Assuntos
Processamento Alternativo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Células COS , Caseínas/genética , Caseínas/metabolismo , Chlorocebus aethiops , Receptores ErbB/genética , Humanos , Rim/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Isoformas de Proteínas , Receptor ErbB-4 , Fator de Transcrição STAT5 , Transdução de Sinais , Tirosina/metabolismo , Ubiquitina/metabolismo
15.
Oncogene ; 26(49): 6968-78, 2007 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-17486068

RESUMO

Several distinct mutations within the kinase domain of the epidermal growth factor receptor (EGFR) are associated with non-small cell lung cancer, but mechanisms underlying their oncogenic potential are incompletely understood. Although normally ligand-induced kinase activation targets EGFR to Cbl-mediated receptor ubiquitinylation and subsequent degradation in lysosomes, we report that certain EGFR mutants escape this regulation. Defective endocytosis characterizes a deletion mutant of EGFR, as well as a point mutant (L858R-EGFR), whose association with c-Cbl and ubiquitinylation are impaired. Our data raise the possibility that refractoriness of L858R-EGFR to downregulation is due to enhanced heterodimerization with the oncogene product HER2, which leads to persistent stimulation.


Assuntos
Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Lisossomos/metabolismo , Transdução de Sinais/fisiologia , Ubiquitina/metabolismo , Biotinilação , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Dimerização , Regulação para Baixo , Receptores ErbB/genética , Humanos , Immunoblotting , Imunoprecipitação , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Mutagênese Sítio-Dirigida , Mutação , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Fator de Transcrição STAT3 , Transcrição Gênica , Ubiquitinação
16.
Oncology ; 61 Suppl 2: 1-13, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11694782

RESUMO

Human epidermal growth factor receptor-2 (HER2/erbB-2) belongs to a family of four transmembrane receptors involved in signal transduction pathways that regulate cell growth and differentiation. Overexpression/amplification of HER2 is associated with malignancy and a poor prognosis in breast cancer. HER2 acts as a networking receptor that mediates signaling to cancer cells, causing them to proliferate. HER receptors exist as monomers but dimerize on ligand binding. HER ligands are bivalent growth factor molecules whose low-affinity site binds to HER2. No HER2-specific ligand has been identified but HER2 is the preferred heterodimerization partner for other HER receptors. HER2-containing heterodimers are relatively long-lived and potent. HER3 has no inherent activity and is the major and most potent dimerization partner of HER2. HER2 overexpression biases the formation of HER2-containing heterodimers, leading to enhanced responsiveness to stromal growth factors and oncogenic transformation. Removal of HER2 from the cell surface or inhibition of its intrinsic enzymatic activity may reduce oncogenicity. Our research suggests that the antitumor efficacy of HER2-specific antibodies such as Herceptin relates to their ability to direct HER2 to a Cbl- dependent endocytosis and degradation pathway. The reported clinical therapeutic efficacy of anti-HER2 monoclonal antibodies in breast cancer highlights the importance of understanding the biology of HER2.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor ErbB-2/fisiologia , Ubiquitina-Proteína Ligases , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Mama/crescimento & desenvolvimento , Mama/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Dimerização , Progressão da Doença , Desenvolvimento Embrionário e Fetal/genética , Endocitose , Células Epiteliais/fisiologia , Evolução Molecular , Feminino , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Genes erbB-2 , Humanos , Ligantes , Mamíferos/metabolismo , Camundongos , Modelos Moleculares , Família Multigênica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Conformação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-cbl , Receptor ErbB-2/química , Receptor ErbB-2/imunologia , Receptor ErbB-3/química , Receptor ErbB-3/genética , Receptor ErbB-3/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais , Células Estromais/fisiologia , Trastuzumab
17.
Eur J Cancer ; 37 Suppl 4: S3-8, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11597398

RESUMO

Growth factors and their transmembrane receptor tyrosine kinases play important roles in cell proliferation, survival, migration and differentiation. One group of growth factors, comprising epidermal growth factor (EGF)-like proteins and neuregulins, stimulates cells to divide by activating members of the EGF receptor (EGFR) family, which consists of the EGFR itself and the receptors known as HER2-4. This highly conserved signalling module plays a fundamental role in the morphogenesis of a diverse spectrum of organisms, ranging from humans to nematodes, and has also been implicated in the development and growth of many types of human tumour cells. In humans, more than 30 ligands and the EGFR family of four receptors lie at the head of a complex, multi-layered signal-transduction network. Different activated receptor-ligand complexes vary in both the strength and type of cellular responses that they induce. Analysis of the multiple processes that modulate EGFR signal transduction, such as receptor heterodimerisation and endocytosis, has revealed new therapeutic opportunities and elucidated mechanisms contributing to the efficacy of existing anticancer treatments.


Assuntos
Receptores ErbB/fisiologia , Proteínas de Neoplasias/fisiologia , Transdução de Sinais , Humanos , Ligantes , Morfogênese , Neoplasias/fisiopatologia , Neoplasias/terapia , Vírus Oncogênicos/fisiologia
18.
Ann Oncol ; 12 Suppl 1: S3-8, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11521719

RESUMO

Human epidermal growth factor receptors (HER/erbB) constitute a family of four cell surface receptors involved in transmission of signals controlling normal cell growth and differentiation. A range of growth factors serve as ligands, but none is specific for the HER2 receptor. HER receptors exist as both monomers and dimers, either homo- or heterodimers. Ligand binding to HERI, HER3 or HER4 induces rapid receptor dimerization, with a marked preference for HER2 as a dimer partner. Moreover, HER2-containing heterodimers generate intracellular signals that are significantly stronger than signals emanating from other HER combinations. In normal cells, few HER2 molecules exist at the cell surface, so few heterodimers are formed and growth signals are relatively weak and controllable. When HER2 is overexpressed multiple HER2 heterodimers are formed and cell signaling is stronger, resulting in enhanced responsiveness to growth factors and malignant growth. This explains why HER2 overexpression is an indicator of poor prognosis in breast tumors and may be predictive of response to treatment. HER2 is a highly specific and promising target for new breast cancer treatments. The recombinant human anti-HER2 monoclonal antibody (rhuMAb-HER2, trastuzumab, Herceptin) induces rapid removal of HER2 from the cell surface, thereby reducing its availability to heterodimers and reducing oncogenicity.


Assuntos
Neoplasias da Mama/metabolismo , Receptor ErbB-2/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Genes erbB-2 , Humanos , Valor Preditivo dos Testes , Prognóstico , Receptor ErbB-2/genética , Transdução de Sinais , Trastuzumab
19.
J Biol Chem ; 276(29): 27677-84, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11375397

RESUMO

Cbl proteins function as ubiquitin protein ligases for the activated epidermal growth factor receptor and, thus, negatively regulate its activity. Here we show that Cbl-b is ubiquitinated and degraded upon activation of the receptor. Epidermal growth factor (EGF)-induced Cbl-b degradation requires intact RING finger and tyrosine kinase binding domains and requires binding of the Cbl-b protein to the activated EGF receptor (EGFR). Degradation of both the EGFR and the Cbl-b protein is blocked by lysosomal and proteasomal inhibitors. Other components of the EGFR-signaling complex (i.e. Grb2 and Shc) are also degraded in an EGF-induced Cbl-b-dependent fashion. Our results suggest that the ubiquitin protein ligase function of Cbl-b is regulated by coordinated degradation of the Cbl-b protein along with its substrate. Furthermore, the data demonstrate that Cbl-b mediates degradation of multiple proteins in the EGFR-signaling complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/fisiologia , Receptores ErbB/metabolismo , Fosfoproteínas/fisiologia , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases , Proteínas de Transporte/química , Humanos , Hidrólise , Fosfoproteínas/química , Conformação Proteica , Proteínas Proto-Oncogênicas c-cbl , Células Tumorais Cultivadas , Ubiquitinas/metabolismo
20.
Oncogene ; 20(2): 147-55, 2001 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-11313944

RESUMO

The anti-cancer agent paclitaxel (Taxol) stabilizes microtubules leading to G2/M cell cycle arrest and apoptotic cell death. In order to analyse the molecular mechanisms of Taxol-induced cytotoxicity, we studied the involvement of mitogen-activated protein kinases (MAPK) ERK and p38 as well as the p53 pathways in Taxol-induced apoptosis. The human breast carcinoma cell line MCF7 and its derivatives, MCF7/HER-2 and MDD2, were used in the study. We found that Taxol treatment strongly activated ERK, p38 MAP kinase and p53 in MAP kinase MCF7 cells prior to apoptosis. PD98059 or SB203580, specific inhibitors of ERK and p38 kinase activities, significantly decreased apoptosis, leaving the surviving cells arrested in G2/M. These inhibitors did not significantly affect Taxol-induced alterations in the cell cycle regulatory proteins Rb, p53, p21/Waf1 and Cdk-2. In addition, inactivation of p53 did not affect cellular sensitivity to Taxol killing. However, cells with inactivated p53, unlike cells harboring wild type p53, failed to arrest in G2/M after treatment with Taxol and continued to divide or go into apoptosis. Our data show that both ERK and p38 MAP kinase cascades are essential for apoptotic response to Taxol-induced cellular killing and are independent of p53 activity. However, p53 may serve as a survival factor in breast carcinoma cells treated with Taxol by blocking cells in G2/M phase of the cell cycle.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Quinases relacionadas a CDC2 e CDC28 , MAP Quinase Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Paclitaxel/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma/tratamento farmacológico , Carcinoma/metabolismo , Carcinoma/patologia , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/efeitos dos fármacos , Ciclinas/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Flavonoides/farmacologia , Fase G2/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitose/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/farmacologia , Ratos , Receptor ErbB-2/efeitos dos fármacos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Proteína do Retinoblastoma/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...