Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
FEBS J ; 285(4): 763-776, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28941174

RESUMO

Despite decades of research, cancer remains a devastating disease and new treatment options are needed. Today cancer is acknowledged as a multifactorial disease not only comprising of aberrant tumor cells but also the associated stroma including tumor vasculature, fibrotic plaques, and immune cells that interact in a complex heterotypic interplay. Myeloid cells represent one of the most abundant immune cell population within the tumor stroma and are equipped with a broad functional repertoire that promotes tumor growth by suppressing cytotoxic T cell activity, stimulating neoangiogenesis and tissue remodeling. Therefore, myeloid cells have become an attractive target for pharmacological intervention. In this review, we summarize the pharmacological approaches to therapeutically target tumor-associated myeloid cells with a focus on advanced programs that are clinically evaluated. In addition, for each therapeutic strategy, the preclinical rationale as well as advantages and challenges from a drug development perspective are discussed.


Assuntos
Antineoplásicos/farmacologia , Desenvolvimento de Medicamentos , Células Mieloides/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Antineoplásicos/síntese química , Antineoplásicos/química , Humanos , Neoplasias/metabolismo , Neoplasias/patologia
2.
PLoS One ; 12(7): e0182039, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28750018

RESUMO

Pancreatic cancer (PC) remains one of the most challenging solid tumors to treat with a high unmet medical need as patients poorly respond to standard-of-care-therapies. Prominent desmoplastic reaction involving cancer-associated fibroblasts (CAFs) and the immune cells in the tumor microenvironment (TME) and their cross-talk play a significant role in tumor immune escape and progression. To identify the key cellular mechanisms induce an immunosuppressive tumor microenvironment, we established 3D co-culture model with pancreatic cancer cells, CAFs and monocytes. Using this model, we analyzed the influence of tumor cells and fibroblasts on monocytes and their immune suppressive phenotype. Phenotypic characterization of the monocytes after 3D co-culture with tumor/fibroblast spheroids was performed by analyzing the expression of defined cell surface markers and soluble factors. Functionality of these monocytes and their ability to influence T cell phenotype and proliferation was investigated. 3D co-culture of monocytes with pancreatic cancer cells and fibroblasts induced the production of immunosuppressive cytokines which are known to promote polarization of M2 like macrophages and myeloid derived suppressive cells (MDSCs). These co-culture spheroid polarized monocyte derived macrophages (MDMs) were poorly differentiated and had an M2 phenotype. The immunosuppressive function of these co-culture spheroids polarized MDMs was demonstrated by their ability to inhibit CD4+ and CD8+ T cell activation and proliferation in vitro, which we could partially reverse by 3D co-culture spheroid treatment with therapeutic molecules that are able to re-activated spheroid polarized MDMs or block immune suppressive factors such as Arginase-I.


Assuntos
Polaridade Celular , Técnicas de Cocultura/métodos , Fibroblastos/patologia , Macrófagos/patologia , Modelos Biológicos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Quimiocinas/metabolismo , Humanos , Imunomodulação , Ativação Linfocitária/imunologia , Monócitos/patologia , Esferoides Celulares , Linfócitos T/citologia
3.
Mol Cancer Ther ; 15(5): 946-57, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27037412

RESUMO

Dysregulated cellular apoptosis and resistance to cell death are hallmarks of neoplastic initiation and disease progression. Therefore, the development of agents that overcome apoptosis dysregulation in tumor cells is an attractive therapeutic approach. Activation of the extrinsic apoptotic pathway is strongly dependent on death receptor (DR) hyperclustering on the cell surface. However, strategies to activate DR5 or DR4 through agonistic antibodies have had only limited clinical success. To pursue an alternative approach for tumor-targeted induction of apoptosis, we engineered a bispecific antibody (BsAb), which simultaneously targets fibroblast-activation protein (FAP) on cancer-associated fibroblasts in tumor stroma and DR5 on tumor cells. We hypothesized that bivalent binding to both FAP and DR5 leads to avidity-driven hyperclustering of DR5 and subsequently strong induction of apoptosis in tumor cells but not in normal cells. Here, we show that RG7386, an optimized FAP-DR5 BsAb, triggers potent tumor cell apoptosis in vitro and in vivo in preclinical tumor models with FAP-positive stroma. RG7386 antitumor efficacy was strictly FAP dependent, was independent of FcR cross-linking, and was superior to conventional DR5 antibodies. In combination with irinotecan or doxorubicin, FAP-DR5 treatment resulted in substantial tumor regression in patient-derived xenograft models. FAP-DR5 also demonstrated single-agent activity against FAP-expressing malignant cells, due to cross-binding of FAP and DR5 across tumor cells. Taken together, these data demonstrate that RG7386, a novel and potent antitumor agent in both mono- and combination therapies, overcomes limitations of previous DR5 antibodies and represents a promising approach to conquer tumor-associated resistance to apoptosis. Mol Cancer Ther; 15(5); 946-57. ©2016 AACR.


Assuntos
Anticorpos Biespecíficos/metabolismo , Anticorpos Biespecíficos/farmacologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Gelatinases/metabolismo , Proteínas de Membrana/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Serina Endopeptidases/metabolismo , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Afinidade de Anticorpos/imunologia , Antineoplásicos/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Endopeptidases , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Gelatinases/imunologia , Humanos , Proteínas de Membrana/imunologia , Camundongos , Ligação Proteica/imunologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Serina Endopeptidases/imunologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
PLoS One ; 10(6): e0127948, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26053043

RESUMO

In recent years, evidence has indicated that the tumor microenvironment (TME) plays a significant role in tumor progression. Fibroblasts represent an abundant cell population in the TME and produce several growth factors and cytokines. Fibroblasts generate a suitable niche for tumor cell survival and metastasis under the influence of interactions between fibroblasts and tumor cells. Investigating these interactions requires suitable experimental systems to understand the cross-talk involved. Most in vitro experimental systems use 2D cell culture and trans-well assays to study these interactions even though these paradigms poorly represent the tumor, in which direct cell-cell contacts in 3D spaces naturally occur. Investigating these interactions in vivo is of limited value due to problems regarding the challenges caused by the species-specificity of many molecules. Thus, it is essential to use in vitro models in which human fibroblasts are co-cultured with tumor cells to understand their interactions. Here, we developed a 3D co-culture model that enables direct cell-cell contacts between pancreatic, breast and or lung tumor cells and human fibroblasts/ or tumor-associated fibroblasts (TAFs). We found that co-culturing with fibroblasts/TAFs increases the proliferation in of several types of cancer cells. We also observed that co-culture induces differential expression of soluble factors in a cancer type-specific manner. Treatment with blocking antibodies against selected factors or their receptors resulted in the inhibition of cancer cell proliferation in the co-cultures. Using our co-culture model, we further revealed that TAFs can influence the response to therapeutic agents in vitro. We suggest that this model can be reliably used as a tool to investigate the interactions between a tumor and the TME.


Assuntos
Técnicas de Cocultura/métodos , Fibroblastos/citologia , Linhagem Celular , Sobrevivência Celular , Citocinas/metabolismo , Fibroblastos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Esferoides Celulares/citologia
5.
BMC Syst Biol ; 3: 1, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19118495

RESUMO

BACKGROUND: In breast cancer, overexpression of the transmembrane tyrosine kinase ERBB2 is an adverse prognostic marker, and occurs in almost 30% of the patients. For therapeutic intervention, ERBB2 is targeted by monoclonal antibody trastuzumab in adjuvant settings; however, de novo resistance to this antibody is still a serious issue, requiring the identification of additional targets to overcome resistance. In this study, we have combined computational simulations, experimental testing of simulation results, and finally reverse engineering of a protein interaction network to define potential therapeutic strategies for de novo trastuzumab resistant breast cancer. RESULTS: First, we employed Boolean logic to model regulatory interactions and simulated single and multiple protein loss-of-functions. Then, our simulation results were tested experimentally by producing single and double knockdowns of the network components and measuring their effects on G1/S transition during cell cycle progression. Combinatorial targeting of ERBB2 and EGFR did not affect the response to trastuzumab in de novo resistant cells, which might be due to decoupling of receptor activation and cell cycle progression. Furthermore, examination of c-MYC in resistant as well as in sensitive cell lines, using a specific chemical inhibitor of c-MYC (alone or in combination with trastuzumab), demonstrated that both trastuzumab sensitive and resistant cells responded to c-MYC perturbation. CONCLUSION: In this study, we connected ERBB signaling with G1/S transition of the cell cycle via two major cell signaling pathways and two key transcription factors, to model an interaction network that allows for the identification of novel targets in the treatment of trastuzumab resistant breast cancer. Applying this new strategy, we found that, in contrast to trastuzumab sensitive breast cancer cells, combinatorial targeting of ERBB receptors or of key signaling intermediates does not have potential for treatment of de novo trastuzumab resistant cells. Instead, c-MYC was identified as a novel potential target protein in breast cancer cells.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proteínas de Ligação a DNA/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Receptores ErbB/metabolismo , Fase G1/fisiologia , Modelos Biológicos , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Anticorpos Monoclonais , Anticorpos Monoclonais Humanizados , Western Blotting , Linhagem Celular Tumoral , Simulação por Computador , Feminino , Humanos , Engenharia de Proteínas/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trastuzumab
6.
J Biomol Screen ; 12(4): 510-20, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17478479

RESUMO

After sequencing the human genome, the challenge ahead is to systematically analyze the functions and disease relation of the proteins encoded. Here the authors describe the application of a flow cytometry-based high-throughput assay to screen for apoptosis-activating proteins in transiently transfected cells. The assay is based on the detection of activated caspase-3 with a specific antibody, in cells overexpressing proteins tagged C- or N-terminally with yellow fluorescent protein. Fluorescence intensities are measured using a flow cytometer integrated with a high-throughput autosampler. The applicability of this screen has been tested in a pilot screen with 200 proteins. The candidate proteins were all verified in an independent microscopy-based nuclear fragmentation assay, finally resulting in the identification of 6 apoptosis inducers.


Assuntos
Proteínas Reguladoras de Apoptose/análise , Proteínas Reguladoras de Apoptose/biossíntese , Apoptose/fisiologia , Citometria de Fluxo , Caspase 3/análise , Caspase 3/biossíntese , Linhagem Celular , Humanos , Projetos Piloto
7.
Genome Biol ; 7(8): R77, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16916453

RESUMO

Highthroughput cell-based assays with flow cytometric readout provide a powerful technique for identifying components of biologic pathways and their interactors. Interpretation of these large datasets requires effective computational methods. We present a new approach that includes data pre-processing, visualization, quality assessment, and statistical inference. The software is freely available in the Bioconductor package prada. The method permits analysis of large screens to detect the effects of molecular interventions in cellular systems.


Assuntos
Doenças Genéticas Inatas/epidemiologia , Software , Bases de Dados Factuais , Processamento Eletrônico de Dados/métodos , Processamento Eletrônico de Dados/normas , Perfilação da Expressão Gênica , Humanos , Modelos Genéticos , Modelos Estatísticos , Razão de Chances
8.
Int J Oncol ; 28(6): 1463-70, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16685447

RESUMO

Urokinase-type plasminogen activator (uPA) is implicated in various pathophysiological processes, including extracellular matrix turnover, cell migration and invasion. Our study aimed to determine the role of uPA in both proliferation and mitogen-activated protein kinase (MAPK) pathway. Hence, we analyzed the effects induced by exogeneous addition of domain-specific uPA antibodies and uPA-interacting molecules on proliferation of uPA-suppressed MDA-MB-231 breast cancer cells. uPA expression was reduced to 53% by stable transfection with an antisense/vector construct and to 65% by siRNA transfection. Immunocytochemical Ki67 staining and flow cytometry (S-phase) analysis indicated a strong decrease of cellular proliferation activity (35% and 38%, respectively). Exogenous addition of high molecular weight-uPA (HMW-uPA) or incubation with the amino terminal fragment (ATF), which lacks the enzymatic activity of uPA, lead to increased cell proliferation. A strong increase of proliferation was absent when the monoclonal anti-uPAR antibody IIIF10 (blocking uPA binding site), soluble uPAR (scavenger effect) and phosphatidyl-inositol-specific phospholipase C (PI-PLC, degrading uPAR) was added prior to the addition of HMW-uPA. In conclusion, HMW-uPA and ATF induce proliferation of breast cancer cells by binding to uPAR. Thereby, integrins situated adjacent to uPAR carry the signals into the cell, thus stimulating proliferation that is mediated via the MAPK pathway.


Assuntos
Neoplasias da Mama/patologia , Divisão Celular/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , RNA Interferente Pequeno/genética , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/genética
9.
Cancer Res ; 65(17): 7733-42, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140941

RESUMO

Cancer transcription microarray studies commonly deliver long lists of "candidate" genes that are putatively associated with the respective disease. For many of these genes, no functional information, even less their relevance in pathologic conditions, is established as they were identified in large-scale genomics approaches. Strategies and tools are thus needed to distinguish genes and proteins with mere tumor association from those causally related to cancer. Here, we describe a functional profiling approach, where we analyzed 103 previously uncharacterized genes in cancer relevant assays that probed their effects on DNA replication (cell proliferation). The genes had previously been identified as differentially expressed in genome-wide microarray studies of tumors. Using an automated high-throughput assay with single-cell resolution, we discovered seven activators and nine repressors of DNA replication. These were further characterized for effects on extracellular signal-regulated kinase 1/2 (ERK1/2) signaling (G1-S transition) and anchorage-independent growth (tumorigenicity). One activator and one inhibitor protein of ERK1/2 activation and three repressors of anchorage-independent growth were identified. Data from tumor and functional profiling make these proteins novel prime candidates for further in-depth study of their roles in cancer development and progression. We have established a novel functional profiling strategy that links genomics to cell biology and showed its potential for discerning cancer relevant modulators of the cell cycle in the candidate lists from microarray studies.


Assuntos
Genes cdc , Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Animais , Ciclo Celular/genética , Replicação do DNA , Perfilação da Expressão Gênica/métodos , Humanos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Células NIH 3T3 , Neoplasias/metabolismo , Neoplasias/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
10.
J Biol Chem ; 280(9): 8482-93, 2005 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-15611116

RESUMO

Mutations in activin receptor-like kinase 1 (ALK1), a transforming growth factor (TGF)-beta type I receptor, lead to the vascular disorder hereditary hemorrhagic telangiectasia caused by abnormal vascular remodeling. The underlying molecular cause of this disease is not well understood. Identifying binding partners for ALK1 will help to understand its cellular function. Using the two-hybrid system, we identified an ALK1-binding protein encoded by an ancient retroviral/retrotransposon element integrated as a single copy gene known as PEG10 on human chromosome 7q21. PEG10 contains two overlapping reading frames from which two proteins, PEG10-RF1 and PEG10-RF1/2, are translated by a typical retroviral -1 ribosomal frameshift mechanism. Reverse transcription-PCR and Northern blot analysis showed a broad range of PEG10 expression in different tissues and cell types, i.e. human placenta, brain, kidney, endothelial cells, lymphoblasts, and HepG2 and HEK293 cells. However, endogenous PEG10-RF1 and PEG10-RF1/2 proteins were only detected in HepG2 and HEK293 cells. PEG10-RF1, which is the major PEG10 protein product, represents a gag-like protein, and PEG10-RF1/2 represents a gag-pol-like protein. PEG10-RF1 also interacts with different members of TGF-beta superfamily type I and II receptors. PEG10-RF1 binding to ALK1 is mediated by a 200-amino acid domain with no recognized motif. PEG10-RF1 inhibits ALK1 as well as ALK5 signaling. Co-expression of ALK1 and PEG10-RF1 in different cell types induced morphological changes reminiscent of neuronal cells or sprouting cells. This is the first report of a human retroviral-like protein interacting with members of the TGF-beta receptor family.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Proteínas de Fusão gag-pol/metabolismo , Produtos do Gene gag/metabolismo , Retroviridae/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Receptores de Activinas Tipo II , Motivos de Aminoácidos , Animais , Northern Blotting , Western Blotting , Células CHO , Células COS , Linhagem Celular , Clonagem Molecular , Cricetinae , Citoplasma/metabolismo , Elementos de DNA Transponíveis , Biblioteca Gênica , Genes Reporter , Humanos , Imunoprecipitação , Luciferases/metabolismo , Microscopia de Fluorescência , Modelos Genéticos , Mutação , Neurônios/metabolismo , Fases de Leitura Aberta , Reação em Cadeia da Polimerase , Ligação Proteica , Estrutura Terciária de Proteína , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Distribuição Tecidual , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA