Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
BMC Cancer ; 16(1): 773, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27716204

RESUMO

BACKGROUND: The epidermal growth factor receptor (EGFR) is frequently overexpressed in head and neck squamous cell carcinoma (HNSCC) and several other human cancers. Monoclonal antibodies, such as cetuximab that block EGFR signaling, have emerged as valuable molecular targeting agents in clinical cancer therapy. Prolonged exposure to cetuximab can result in cells acquiring resistance by a process that remains incompletely understood. METHODS: In this study, we analyzed the immediate early molecular response of cetuximab on physical interactions between EGFR and Insulin growth factor 1 like receptor (IGF-1R) in head and neck cancer cells that are resistant to cetuximab. Co-immunoprecipitation, small molecule inhibitors against phospho-Src and IGF-1R, quantitative western blot of EGFR and Src phosphorylation, cell proliferation assays were used to suggest the role of IGF-1R mediated phosphorylation of specific tyrosine Y845 on EGFR via increased heterodimerization of EGFR and IGF-1R in cetuximab resistant cells. RESULTS: Heterodimerization of EGFR with IGF-1R was increased in cetuximab resistant HNSCC cell line UMSCC6. Basal levels of phosphorylated EGFR Y845 showed significant increase in the presence of cetuximab. Surprisingly, this activated Y845 level was not inhibited in the presence of Src inhibitor PP1. Instead, inhibition of IGF-1R by picropodophyllin (PPP) reduced the EGFR Y845 levels. Taken together, these results suggest that heterodimerization of EGFR with IGF-1R can lead to increased activity of EGFR and may be an important platform for cetuximab mediated signaling in head and neck tumors that have become resistant to anti-EGFR therapy. CONCLUSIONS: EGFR-IGF-1R interaction has a functional consequence of phosphorylation of EGFR Y845 in cetuximab resistant HNSCC cells and dual targeting of EGFR and IGF-1R is a promising therapeutic strategy.


Assuntos
Receptores de Somatomedina/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cetuximab/farmacologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Humanos , Fosforilação , Multimerização Proteica , Processamento de Proteína Pós-Traducional , Receptor IGF Tipo 1
2.
Recent Results Cancer Res ; 198: 45-87, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27318681

RESUMO

Ionizing radiation has been shown to activate and interact with multiple growth factor receptor pathways that can influence tumor response to therapy. Among these receptor interactions, the epidermal growth factor receptor (EGFR) has been the most extensively studied with mature clinical applications during the last decade. The combination of radiation and EGFR-targeting agents using either monoclonal antibody (mAb) or small-molecule tyrosine kinase inhibitor (TKI) offers a promising approach to improve tumor control compared to radiation alone. Several underlying mechanisms have been identified that contribute to improved anti-tumor capacity after combined treatment. These include effects on cell cycle distribution, apoptosis, tumor cell repopulation, DNA damage/repair, and impact on tumor vasculature. However, as with virtually all cancer drugs, patients who initially respond to EGFR-targeted agents may eventually develop resistance and manifest cancer progression. Several potential mechanisms of resistance have been identified including mutations in EGFR and downstream signaling molecules, and activation of alternative member-bound tyrosine kinase receptors that bypass the inhibition of EGFR signaling. Several strategies to overcome the resistance are currently being explored in preclinical and clinical models, including agents that target the EGFR T790 M resistance mutation or target multiple EGFR family members, as well as agents that target other receptor tyrosine kinase and downstream signaling sites. In this chapter, we focus primarily on the interaction of radiation with anti-EGFR therapies to summarize this promising approach and highlight newly developing opportunities.


Assuntos
Receptores ErbB/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Neoplasias/terapia , Radioterapia (Especialidade)/métodos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Antineoplásicos/uso terapêutico , Cetuximab/uso terapêutico , Quimiorradioterapia , Receptores ErbB/metabolismo , Humanos , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico
4.
Clin Cancer Res ; 15(5): 1585-92, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19190133

RESUMO

PURPOSE: The epidermal growth factor receptor (EGFR) is recognized as a key mediator of proliferation and progression in many human tumors. A series of EGFR-specific inhibitors have recently gained Food and Drug Administration approval in oncology. These strategies of EGFR inhibition have shown major tumor regressions in approximately 10% to 20% of advanced cancer patients. Many tumors, however, eventually manifest resistance to treatment. Efforts to better understand the underlying mechanisms of acquired resistance to EGFR inhibitors, and potential strategies to overcome resistance, are greatly needed. EXPERIMENTAL DESIGN: To develop cell lines with acquired resistance to EGFR inhibitors we utilized the human head and neck squamous cell carcinoma tumor cell line SCC-1. Cells were treated with increasing concentrations of cetuximab, gefitinib, or erlotinib, and characterized for the molecular changes in the EGFR inhibitor-resistant lines relative to the EGFR inhibitor-sensitive lines. RESULTS: EGFR inhibitor-resistant lines were able to maintain their resistant phenotype in both drug-free medium and in athymic nude mouse xenografts. In addition, EGFR inhibitor-resistant lines showed a markedly increased proliferation rate. EGFR inhibitor-resistant lines had elevated levels of phosphorylated EGFR, mitogen-activated protein kinase, AKT, and signal transducer and activator of transcription 3, which were associated with reduced apoptotic capacity. Subsequent in vivo experiments indicated enhanced angiogenic potential in EGFR inhibitor-resistant lines. Finally, EGFR inhibitor-resistant lines showed cross-resistance to ionizing radiation. CONCLUSIONS: We have developed EGFR inhibitor-resistant human head and neck squamous cell carcinoma cell lines. This model provides a valuable preclinical tool to investigate molecular mechanisms of acquired resistance to EGFR blockade.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Modelos Biológicos , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cetuximab , Receptores ErbB/genética , Cloridrato de Erlotinib , Gefitinibe , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Immunoblotting , Camundongos , Camundongos Nus , Neovascularização Patológica , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Doses de Radiação , Células Tumorais Cultivadas
5.
Cancer Res ; 67(3): 1155-62, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17283150

RESUMO

Signaling through the insulin-like growth factor-I receptor (IGF-IR) is implicated in cellular proliferation, apoptosis, carcinogenesis, metastasis, and resistance to cytotoxic cancer therapies. Targeted disruption of IGF-IR signaling combined with cytotoxic therapy may therefore yield improved anticancer efficacy over conventional treatments alone. In this study, a fully human anti-IGF-IR monoclonal antibody A12 (ImClone Systems, Inc., New York, NY) is examined as an adjunct to radiation therapy. IGF-IR expression is shown for a diverse cohort of cell lines, whereas targeted IGF-IR blockade by A12 inhibits IGF-IR phosphorylation and activation of the downstream effectors Akt and mitogen-activated protein kinase. Anchorage-dependent proliferation and xenograft growth is inhibited by A12 in a dose-dependent manner, particularly for non-small cell lung cancer lines. Clonogenic radiation survival of H226 and H460 cells grown under anchorage-dependent conditions is impaired by A12, demonstrating a radiation dose-enhancing effect for IGF-IR blockade. Postradiation anchorage-independent colony formation is inhibited by A12 in A549 and H460 cells. In the H460 xenograft model, combining A12 and radiation significantly enhances antitumor efficacy compared with either modality alone. These effects may be mediated by promotion of radiation-induced, double-stranded DNA damage and apoptosis as observed in cell culture. In summary, these results validate IGF-IR signal transduction blockade as a promising strategy to improve radiation therapy efficacy in human tumors, forming a basis for future clinical trials.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias/terapia , Receptor IGF Tipo 1/antagonistas & inibidores , Animais , Apoptose/fisiologia , Apoptose/efeitos da radiação , Adesão Celular/fisiologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Terapia Combinada , Dano ao DNA , DNA de Neoplasias/efeitos da radiação , Feminino , Humanos , Camundongos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/radioterapia , Fosforilação , Receptor IGF Tipo 1/biossíntese , Receptor IGF Tipo 1/imunologia , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Radiat Oncol Biol Phys ; 72(2): 534-42, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18793955

RESUMO

PURPOSE: To examine the interaction between panitumumab, a fully human anti-epidermal growth factor receptor monoclonal antibody, and radiation in head-and-neck squamous cell carcinoma and non-small-cell lung cancer cell lines and xenografts. METHODS AND MATERIALS: The head-and-neck squamous cell carcinoma lines UM-SCC1 and SCC-1483, as well as the non-small-cell lung cancer line H226, were studied. Tumor xenografts in athymic nude mice were used to assess the in vivo activity of panitumumab alone and combined with radiation. In vitro assays were performed to assess the effect of panitumumab on radiation-induced cell signaling, apoptosis, and DNA damage. RESULTS: Panitumumab increased the radiosensitivity as measured by the clonogenic survival assay. Radiation-induced epidermal growth factor receptor phosphorylation and downstream signaling through mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription 3 (STAT3) was inhibited by panitumumab. Panitumumab augmented radiation-induced DNA damage by 1.2-1.6-fold in each of the cell lines studied as assessed by residual gamma-H(2)AX foci after radiation. Radiation-induced apoptosis was increased 1.4-1.9-fold by panitumumab, as evidenced by Annexin V-fluorescein isothiocyanate staining and flow cytometry. In vivo, the combination therapy of panitumumab and radiation was superior to panitumumab or radiation alone in the H226 xenografts (p = 0.01) and showed a similar trend in the SCC-1483 xenografts (p = 0.08). In vivo, immunohistochemistry demonstrated the ability of panitumumab to augment the antiproliferative and antiangiogenic effects of radiation. CONCLUSION: These studies have identified a favorable interaction in the combination of radiation and panitumumab in upper aerodigestive tract tumor models, both in vitro and in vivo. These data suggest that clinical investigations examining the combination of radiation and panitumumab in the treatment of epithelial tumors warrant additional pursuit.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Carcinoma de Células Escamosas/radioterapia , Neoplasias Pulmonares/radioterapia , Neoplasias Bucais/radioterapia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Terapia Combinada/métodos , Dano ao DNA/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Soalho Bucal , Neoplasias Bucais/tratamento farmacológico , Panitumumabe , Fosforilação/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Fator de Transcrição STAT3/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transplante Heterólogo
7.
Cancer Res ; 65(8): 3328-35, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15833866

RESUMO

Erlotinib (Tarceva) is an orally available HER1 (epidermal growth factor receptor, EGFR) tyrosine kinase inhibitor advancing through clinical trials for the treatment of a range of human malignancies. In this study, we examine the capacity of erlotinib to modulate radiation response and investigate specific mechanisms underlying these interactions in human tumor cell lines and xenografts. The impact of erlotinib on cell cycle kinetics was analyzed using flow cytometry, and the impact on apoptosis was evaluated via fluorescein-labeled pan-caspase inhibition and poly(ADP-ribose) polymerase cleavage. Radiation-induced EGFR autophosphorylation and Rad51 expression were examined by Western blot analysis. Radiation survival was analyzed using a clonogenic assay and assessment of in vivo tumor growth was done using a mouse xenograft model system. Microarray studies were carried out using 20 K human cDNA microarray and select genes were validated using quantitative reverse transcription-PCR (RT-PCR). Independently, erlotinib and radiation induce accumulation of tumor cells in G(1) and G(2)-M phase, respectively, with a reduction of cells in S phase. When combined with radiation, erlotinib promotes a further reduction in S-phase fraction. Erlotinib enhances the induction of apoptosis, inhibits EGFR autophosphorylation and Rad51 expression following radiation exposure, and promotes an increase in radiosensitivity. Tumor xenograft studies confirm that systemic administration of erlotinib results in profound tumor growth inhibition when combined with radiation. cDNA microarray analysis assessing genes differentially regulated by erlotinib following radiation exposure identifies a diverse set of genes deriving from several functional classes. Validation is confirmed for several specific genes that may influence radiosensitization by erlotinib including Egr-1, CXCL1, and IL-1beta. These results identify the capacity of erlotinib to enhance radiation response at several levels, including cell cycle arrest, apoptosis induction, accelerated cellular repopulation, and DNA damage repair. Preliminary microarray data suggests additional mechanisms underlying the complex interaction between EGFR signaling and radiation response. These data suggest that the erlotinib/radiation combination represents a strategy worthy of further examination in clinical trials.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/efeitos da radiação , Neoplasias Pulmonares/radioterapia , Neoplasias da Próstata/radioterapia , Quinazolinas/farmacologia , Radiossensibilizantes/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/efeitos da radiação , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Antígeno Nuclear de Célula em Proliferação/biossíntese , Neoplasias da Próstata/tratamento farmacológico , Rad51 Recombinase , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Semin Radiat Oncol ; 16(1): 38-44, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16378905

RESUMO

Molecular inhibition of epidermal growth factor receptor (EGFR) signaling represents one of the most promising current arenas for the advancement of molecularly targeted cancer therapies. A series of EGFR inhibitors from both the monoclonal antibody (mAb) and tyrosine kinase inhibitor (TKI) class have shown clear clinical activity in the treatment of several common human cancers. Three EGFR inhibitors have recently gained Food and Drug Administration (FDA) approval for cancer therapy in the United States including the mAb cetuximab (Erbitux) and the small-molecule TKIs gefitinib (Iressa) and erlotinib (Tarceva). The rapidly expanding preclinical and clinical data contributing to these FDA approvals validate a central role of the EGFR as an important molecular target in epithelial malignancies. Indeed, one of the more striking clinical results in this field has been recently achieved by combining an EGFR inhibitor (cetuximab) with radiation in the treatment of advanced head and neck cancer patients. Nevertheless, the overall clinical gains realized to date with the EGFR inhibitors are modest for the global cancer population. Much remains to be learned regarding the rational integration of EGFR inhibitors into cancer treatment regimens as well as methods to optimize the selection of patients most likely to benefit from EGFR inhibitor strategies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Anticorpos Monoclonais Humanizados , Cetuximab , Ensaios Clínicos como Assunto , Terapia Combinada/métodos , Cloridrato de Erlotinib , Gefitinibe , Humanos , Inibidores de Proteínas Quinases , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/uso terapêutico
9.
Int J Radiat Oncol Biol Phys ; 64(5): 1458-65, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16488554

RESUMO

PURPOSE: To examine the antivascular and antitumor activity of the vascular targeting agent ZD6126 in combination with radiation in lung and head-and-neck (H and N) cancer models. The overall hypothesis was that simultaneous targeting of tumor cells (radiation) and tumor vasculature (ZD6126) might enhance tumor cell killing. METHODS AND MATERIALS: A series of in vitro studies using human umbilical vein endothelial cells (HUVEC) and in vivo studies in athymic mice bearing human lung (H226) and H and N (squamous cell carcinoma [SCC]1, SCC6) tumor xenografts treated with ZD6126 and/or radiation were performed. RESULTS: ZD6126 inhibited the capillary-like network formation in HUVEC. Treatment of HUVEC with ZD6126 resulted in cell cycle arrest in G2/M, with decrease of cells in S phase and proliferation inhibition in a dose-dependent manner. ZD6126 augmented the cell-killing effect of radiation and radiation-induced apoptosis in HUVEC. The combination of ZD6126 and radiation further decreased tumor vascularization in an in vivo Matrigel angiogenesis assay. In tumor xenografts, ZD6126 enhanced the antitumor activity of radiation, resulting in tumor growth delay. CONCLUSIONS: These preclinical studies suggest that ZD6126 can augment the radiation response of proliferating endothelial H and N and lung cancer cells. These results complement recent reports suggesting the potential value of combining radiation with vascular targeting/antiangiogenic agents.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Compostos Organofosforados/uso terapêutico , Animais , Caspases/análise , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Colágeno , Terapia Combinada/métodos , Combinação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/efeitos da radiação , Fase G2/efeitos dos fármacos , Humanos , Laminina , Camundongos , Camundongos Nus , Neovascularização Patológica/prevenção & controle , Proteoglicanas , Distribuição Aleatória , Fase S/efeitos dos fármacos , Veias Umbilicais/citologia
10.
Cancer Res ; 62(15): 4300-6, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12154033

RESUMO

ZD1839 ("Iressa") is an orally-active, selective epidermal growth factor receptor-tyrosinekinase inhibitor. We evaluated the antitumor activity of ZD1839 in combination with radiation in human squamous cell carcinomas (SCCs) of the head and neck. ZD1839 produced a dose-dependent inhibition of cellular proliferation in human SCCs grown in culture. Flow cytometry analysis of cell cycle progression confirmed the accumulation of cells in G(1) phase after exposure to ZD1839. Clonogenic analysis demonstrated that treatment of SCCs with ZD1839 reduced cell survival after radiation exposure. Flow cytometric analysis further demonstrated that treatment of SCCs with ZD1839 amplified radiation-induced apoptosis. Tumor xenograft studies confirmed that oral administration of ZD1839, or focal radiation, resulted in partial and transient tumor regression in both SCC-1 and SCC-6 xenografts. In contrast, profound tumor regression and regrowth delay was observed in mice treated with the combination of ZD1839 and radiation. To examine antiangiogenic effects, we studied the impact of ZD1839 on human umbilical vascular endothelial cells (HUVECs). In the presence of reconstituted Matrigel matrix, HUVECs established a capillary-like network structure (tube formation). Treatment with ZD1839 reduced the cell-to-cell interaction of HUVECs, resulting in disruption of tube formation. The effect of ZD1839 was further examined using an in vivo tumor xenograft model of angiogenesis (Matrigel plug) in athymic mice. Systemic treatment with ZD1839 significantly inhibited tumor-induced neovascularization across the Matrigel plug. Taken together, these results suggest that the antitumor activity of ZD1839 in combination with radiation appears to derive from not only proliferative growth inhibition (with associated cell cycle arrest and enhancement of radiation-induced apoptosis) but also from inhibition of tumor angiogenesis.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/terapia , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/terapia , Neovascularização Patológica/terapia , Quinazolinas/farmacologia , Radiossensibilizantes/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Terapia Combinada , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/efeitos da radiação , Feminino , Fase G1/efeitos dos fármacos , Fase G1/efeitos da radiação , Gefitinibe , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/radioterapia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Res ; 64(15): 5355-62, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15289342

RESUMO

Molecular inhibition of epidermal growth factor receptor (EGFR/HER1) signaling is under active investigation as a promising cancer treatment strategy. We examined the potency of EGFR inhibition achieved by combining anti-EGFR monoclonal antibody and tyrosine kinase inhibitor, which target extracellular and intracellular domains of the receptor, respectively. We specifically studied the combination of cetuximab (Erbitux, C225; ImClone Systems, New York, NY) with either gefitinib (Iressa, ZD1839; AstraZeneca, Macclesfield, UK) or erlotinib (Tarceva, OSI-774; Genentech, South San Francisco, CA) across a variety of human cancer cells. The combination of cetuximab plus gefitinib or erlotinib enhanced growth inhibition over that observed with either agent alone. As measured by immunostaining, inhibition of EGFR phosphorylation with the combination of cetuximab plus gefitinib or erlotinib was augmented over that obtained with single-agent therapy in head and neck (H&N) cancer cell lines. Phosphorylation inhibition of downstream effector molecules [mitogen-activated protein kinase (MAPK) and AKT] also was enhanced in tumor cells treated with the combination of cetuximab plus gefitinib or erlotinib. Flow cytometry and immunoblot analysis demonstrated that treatment of H&N tumor cells with cetuximab in combination with either gefitinib or erlotinib amplified the induction of apoptosis. Following establishment of cetuximab-resistant cell lines, we observed that gefitinib or erlotinib retained the capacity to inhibit growth of lung and H&N tumor cells that were highly resistant to cetuximab. Treatment with gefitinib or erlotinib, but not cetuximab, also could further inhibit the activation of downstream effectors of EGFR signaling in cetuximab-resistant cells, including MAPK and AKT. These data suggest that tyrosine kinase inhibitors may further modulate intracellular signaling that is not fully blocked by extracellular anti-EGFR antibody treatment. Finally, animal studies confirmed that single EGFR inhibitor treatment resulted in partial and transient tumor regression in human lung cancer xenografts. In contrast, more profound tumor regression and regrowth delay were observed in mice treated with the combination of cetuximab and gefitinib or erlotinib. Immunohistochemical staining, which demonstrated significant reduction of the proliferative marker proliferating cell nuclear antigen in mice treated with dual EGFR inhibitors, further supported this in vivo observation. Together, these data suggest that combined treatment with distinct EGFR inhibitory agents can augment the potency of EGFR signaling inhibition. This approach suggests potential new strategies to maximize effective target inhibition, which may improve the therapeutic ratio for anti-EGFR-targeted therapies in developing clinical trials.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Divisão Celular/efeitos dos fármacos , Cetuximab , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Feminino , Citometria de Fluxo , Gefitinibe , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Immunoblotting , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Quinazolinas/administração & dosagem , Transplante Heterólogo , Células Tumorais Cultivadas
12.
Clin Cancer Res ; 22(3): 633-43, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26420857

RESUMO

PURPOSE: Aberrant regulation of the EGF receptor family (EGFR, HER2, HER3, HER4) contributes to tumorigenesis and metastasis in epithelial cancers. Pan-HER represents a novel molecular targeted therapeutic composed of a mixture of six monoclonal antibodies against EGFR, HER2, and HER3. EXPERIMENTAL DESIGN: In the current study, we examine the capacity of Pan-HER to augment radiation response across a series of human lung and head and neck cancers, including EGFR inhibitor-resistant cell lines and xenografts. RESULTS: Pan-HER demonstrates superior antiproliferative and radiosensitizing impact when compared with cetuximab. The mechanisms underlying these effects appear to involve attenuation of DNA damage repair, enhancement of programmed cell death, cell-cycle redistribution, and induction of cellular senescence. Combined treatment of Pan-HER with single or fractionated radiation in human tumor xenografts reveals a potent antitumor and regrowth delay impact compared with Pan-HER or radiation treatment alone. CONCLUSIONS: These data highlight the capacity of Pan-HER to augment radiation response in lung and head and neck cancer models and support investigation of Pan-HER combined with radiation as a promising clinical therapeutic strategy.


Assuntos
Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias Pulmonares/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Senescência Celular/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Modelos Animais de Doenças , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inibidores , Receptor ErbB-3/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Res ; 76(13): 3929-41, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27197149

RESUMO

Interest in combining radiotherapy and immune checkpoint therapy is growing rapidly. In this study, we explored a novel combination of this type to augment antitumor immune responses in preclinical murine models of melanoma, neuroblastoma, and head and neck squamous cell carcinoma. Cooperative effects were observed with local radiotherapy and intratumoral injection of tumor-specific antibodies, arising in part from enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). We could improve this response by combining radiation with intratumoral injection of an IL2-linked tumor-specific antibody (termed here an immunocytokine), resulting in complete regression of established tumors in most animals associated with a tumor-specific memory T-cell response. Given the T-cell response elicited by combined local radiation and intratumoral immunocytokine, we tested the potential benefit of adding this treatment to immune checkpoint blockade. In mice bearing large primary tumors or disseminated metastases, the triple-combination of intratumoral immunocytokine, radiation, and systemic anti-CTLA-4 improved primary tumor response and animal survival compared with combinations of any two of these three interventions. Taken together, our results show how combining radiation and intratumoral immunocytokine in murine tumor models can eradicate large tumors and metastases, eliciting an in situ vaccination effect that can be leveraged further by T-cell checkpoint blockade, with immediate implications for clinical evaluation. Cancer Res; 76(13); 3929-41. ©2016 AACR.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígeno CTLA-4/imunologia , Interleucina-2/imunologia , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Neoplasias Pancreáticas/terapia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Western Blotting , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Quimiorradioterapia , Terapia Combinada , Feminino , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Linfócitos T/imunologia , Células Tumorais Cultivadas , Vacinação , Raios X , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Int J Radiat Oncol Biol Phys ; 62(1): 223-9, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15850925

RESUMO

PURPOSE: Histone deacetylase (HDAC) inhibitors, which modulate chromatin structure and gene expression, represent a class of anticancer agents that hold particular potential as radiation sensitizers. In this study, we examine the capacity of the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) to modulate radiation response in human tumor cell lines and explore potential mechanisms underlying these interactions. METHODS AND MATERIALS: Cell proliferation: Exponentially growing tumor cells were incubated in medium containing 0-10 microM of SAHA for 72 h. Cells were fixed/stained with crystal violet to estimate cell viability. Apoptosis: Caspase activity was analyzed by fluorescence spectroscopy using a fluorescein labeled pan-caspase inhibitor. Cells were harvested after 48 h of exposure to SAHA (1.0 microM), radiation (6 Gy), or the combination. Whole cell lysates were evaluated for poly(ADP-ribose) polymerase (PARP) cleavage by western blot analysis. Radiation survival: Cells were exposed to varying doses of radiation +/- 3 days pretreatment with SAHA (0.75-1.0 microM). After incubation intervals of 14-21 days, colonies were stained with crystal violet and manually counted. Immunocytochemistry: Cells were grown and treated in chamber slides. At specified times after treatment with SAHA, cells were fixed in paraformaldehyde, permeabilized in methanol, and probed with primary and secondary antibody solutions. Slides were analyzed using an epifluorescent microscope. RESULTS: SAHA induced a dose-dependent inhibition of proliferation in human prostate (DU145) and glioma (U373vIII) cancer cell lines. Exposure to SAHA enhanced radiation-induced apoptosis as measured by caspase activity (p < 0.05) and PARP cleavage. The impact of SAHA on radiation response was further characterized using clonogenic survival analysis, which demonstrated that treatment with SAHA reduced tumor survival after radiation exposure. We identified several oncoproteins and DNA damage repair proteins (epidermal growth factor receptor, AKT, DNA-PK, and Rad51) that show differential expression after exposure to SAHA. These proteins may contribute to mechanistic synergy between HDAC inhibition and radiation response. CONCLUSION: These preclinical results suggest that treatment with the HDAC inhibitor SAHA can enhance radiation-induced cytotoxicity in human prostate and glioma cells. We are examining the capacity of HDAC inhibitors to modulate radiation response and tumor control in animal xenograft model systems to strengthen the rationale for future clinical trial exploration.


Assuntos
Inibidores Enzimáticos/farmacologia , Glioma/radioterapia , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Neoplasias da Próstata/radioterapia , Radiossensibilizantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Glioma/enzimologia , Glioma/patologia , Humanos , Masculino , Poli(ADP-Ribose) Polimerases/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Tolerância a Radiação/efeitos dos fármacos , Células Tumorais Cultivadas , Vorinostat
15.
Int J Radiat Oncol Biol Phys ; 62(5): 1477-85, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16029810

RESUMO

The formation of new blood vessels (angiogenesis) represents a critical factor in the malignant growth of solid tumors and metastases. Vascular endothelial cell growth factor (VEGF) and its receptor VEGFR2 represent central molecular targets for antiangiogenic intervention, because of their integral involvement in endothelial cell proliferation and migration. In the current study, we investigated in vitro and in vivo effects of receptor blockade on various aspects of the angiogenic process using monoclonal antibodies against VEGFR2 (cp1C11, which is human specific, and DC101, which is mouse specific). Molecular blockade of VEGFR2 inhibited several critical steps involved in angiogenesis. VEGFR2 blockade in endothelial cells attenuated cellular proliferation, reduced cellular migration, and disrupted cellular differentiation and resultant formation of capillary-like networks. Further, VEGFR2 blockade significantly reduced the growth response of human squamous cell carcinoma xenografts in athymic mice. The growth-inhibitory effect of VEGFR2 blockade in tumor xenografts seems to reflect antiangiogenic influence as demonstrated by vascular growth inhibition in an in vivo angiogenesis assay incorporating tumor-bearing Matrigel plugs. Further, administration of VEGFR2-blocking antibodies in endothelial cell cultures, and in mouse xenograft models, increased their response to ionizing radiation, indicating an interactive cytotoxic effect of VEGFR2 blockade with radiation. These data suggest that molecular inhibition of VEGFR2 alone, and in combination with radiation, can enhance tumor response through molecular targeting of tumor vasculature.


Assuntos
Anticorpos Monoclonais/farmacologia , Endotélio Vascular/efeitos dos fármacos , Neovascularização Patológica/prevenção & controle , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/radioterapia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno , Combinação de Medicamentos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos da radiação , Feminino , Humanos , Laminina , Camundongos , Camundongos Nus , Proteoglicanas , Tolerância a Radiação , Especificidade da Espécie , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
16.
Mol Cancer Ther ; 1(7): 507-14, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12479268

RESUMO

Tumor metastasis represents a complex multistep process that requires migration, invasion, and angiogenesis. In this study, we examined the impact of molecular blockade of the epidermal growth factor receptor on the invasive and metastatic capacity of human squamous cell carcinoma (SCC) of the head and neck using in vitro and in vivo model systems. Treatment with the anti-epidermal growth factor receptor antibody C225 attenuated the migration of SCC-1 tumor cells through a chemotaxis chamber in a dose-dependent manner. Incubation of SCC cells with 10-100 nM C225 for 4 h resulted in 40-60% inhibition of cell migration. Furthermore, in the presence of C225, the capacity of SCC-1 to invade across a layer of extracellular matrix (Matrigel) was significantly inhibited. Using an in vivo orthotopic floor-of-mouth xenograft model, locoregional tumor invasion of SCC-1 into muscle, vessel, bone, and perineural tissues was inhibited in C225-treated mice. This inhibition was additionally characterized by down-regulation in the expression of matrix metalloproteinase-9. These data suggest that inhibition of metastatic potential by C225 may be mediated via decreased migration and invasion of SCC cells. Regarding angiogenesis in vitro, we first studied human umbilical vascular endothelial cells, which established a capillary-like network structure (tube formation) in the presence of reconstituted Matrigel. Treatment with C225 reduced cell-to-cell interaction of human umbilical vascular endothelial cells, resulting in disruption of tube formation. The effect of C225 was additionally examined using an in vivo tumor xenograft neovascularization model of angiogenesis. Systemic treatment with C225 not only reduced tumor growth and the number of blood capillaries but also hindered the growth of established vessels toward the tumor. Taken together, these results provide evidence that C225 can suppress tumor-induced neovascularization and metastasis in SCC of the head and neck.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Fator de Crescimento Epidérmico/metabolismo , Neovascularização Patológica , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Movimento Celular , Células Cultivadas , Cetuximab , Quimiotaxia , Colágeno/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Combinação de Medicamentos , Endotélio Vascular/citologia , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Imuno-Histoquímica , Laminina/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Proteoglicanas/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas
17.
Mol Cancer Ther ; 14(9): 2049-59, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26141946

RESUMO

Human epidermal growth factor receptor family members (EGFR, HER2, HER3, and HER4) play important roles in tumorigenesis and response to cancer therapeutics. In this study, we evaluated the capacity of the dual-target antibody MEHD7945A that simultaneously targets EGFR and HER3 to modulate radiation response in lung and head and neck cancer models. Antitumor effects of MEHD7945A in combination with radiation were evaluated in cell culture and tumor xenograft models. Mechanisms that may contribute to increased radiation killing by MEHD7945A, including DNA damage and inhibition of EGFR-HER signaling pathways, were analyzed. Immunohistochemical analysis of tumor xenografts was conducted to evaluate the effect of MEHD7945A in combination with radiation on tumor growth and microenvironment. MEHD7945A inhibited basal and radiation-induced EGFR and HER3 activation resulting in the inhibition of tumor cell growth and enhanced radiosensitivity. MEHD7945A was more effective in augmenting radiation response than treatment with individual anti-EGFR or anti-HER3 antibodies. An increase in DNA double-strand breaks associated γ-H2AX was observed in cells receiving combined treatment with MEHD7945A and radiation. Immunohistochemical staining evaluation in human tumor xenografts showed that MEHD7945A combined with radiation significantly reduced the expression of markers of tumor proliferation and tumor vasculature. These findings reveal the capacity of MEHD7945A to augment radiation response in lung and head and neck cancers. The dual EGFR/HER3-targeting action of MEHD7945A merits further investigation and clinical trial evaluation as a radiation sensitizer in cancer therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/metabolismo , Imunoglobulina G/farmacologia , Receptor ErbB-3/antagonistas & inibidores , Animais , Anticorpos Monoclonais/administração & dosagem , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Imunoglobulina G/administração & dosagem , Imuno-Histoquímica , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Radioterapia Adjuvante , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Mol Cancer Ther ; 14(9): 1994-2003, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26162687

RESUMO

MDM2-p53 interaction and downstream signaling affect cellular response to DNA damage. AMG 232 is a potent small molecule inhibitor that blocks the interaction of MDM2 and p53. We examined the capacity of AMG 232 to augment radiation response across a spectrum of human tumor cell lines and xenografts. AMG 232 effectively inhibited proliferation and enhanced radiosensitivity via inhibition of damage repair signaling. Combined AMG 232 and radiation treatment resulted in the accumulation of γH2AX-related DNA damage and induction of senescence with promotion of apoptotic and/or autophagic cell death. Several molecules involved in senescence, autophagy, and apoptosis were specifically modulated following the combined AMG 232/radiation treatment, including FoxM1, ULK-1, DRAM, and BAX. In vivo xenograft studies confirmed more potent antitumor and antiangiogenesis efficacy with combined AMG 232/radiation treatment than treatment with drug or radiation alone. Taken together, these data identify the capacity of AMG 232 to augment radiation response across a variety of tumor types harboring functional p53.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Acetatos/farmacologia , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Autofagia/efeitos dos fármacos , Autofagia/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Senescência Celular/efeitos dos fármacos , Senescência Celular/efeitos da radiação , Modelos Animais de Doenças , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/radioterapia , Piperidonas/farmacologia , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/genética , Radiação Ionizante , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Semin Radiat Oncol ; 12(3 Suppl 2): 21-6, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12174341

RESUMO

Approximately two thirds of all human solid tumors derive from epithelial tissues. The epidermal growth factor receptor (EGFR) serves as an important regulator of cellular growth in epithelial tumors. A spectrum of new anticancer agents have been specifically designed to target the EGFR in an effort to inhibit malignant growth. Although several of these new drugs show single-agent activity in early clinical trials, the predominant growth effect of EGFR signaling inhibition is cytostatic. However, the interaction of EGFR inhibition combined with conventional cytotoxic therapies (radiation and chemotherapy) is more potent, and shows great promise in the treatment of a variety of solid tumors that overexpress this receptor. This report focuses primarily on the capacity of EGFR inhibitors to modulate cellular and overall tumor response to ionizing radiation.


Assuntos
Receptores ErbB/biossíntese , Receptores ErbB/efeitos da radiação , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Ensaios Clínicos como Assunto , Terapia Combinada , Receptores ErbB/efeitos dos fármacos , Humanos , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/secundário , Neoplasias Epiteliais e Glandulares/terapia , Quinazolinas/antagonistas & inibidores , Quinazolinas/uso terapêutico , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/uso terapêutico , Resultado do Tratamento
20.
Int J Radiat Oncol Biol Phys ; 58(3): 976-83, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14967459

RESUMO

PURPOSE: To highlight some of the preclinical data that examine the interaction of epidermal growth factor receptor (EGFR) inhibitors with radiotherapy and chemotherapy. METHODS AND MATERIALS: Recognition of the EGFR as an important regulator of tumor cell growth in the early 1980s stimulated the development of a series of molecules specifically designed to inhibit EGFR signaling as anticancer agents. Many of these agents have now matured and are in advanced clinical trial investigations, with tumor response rates on the order of 10-20% identified across a variety of human malignancies. Initially designed primarily as "cytostatic" agents, as opposed to "cytotoxic" agents, it is possible that the EGFR inhibitors will realize their optimal clinical impact when delivered in concert with conventional cytotoxic modalities such as radiotherapy and/or chemotherapy. RESULTS: Despite very strong in vitro and in vivo preclinical results, several major gaps remain in our knowledge regarding the EGFR inhibitor mechanisms of interaction with radiotherapy and chemotherapy, with considerable selection bias in the publication of preclinical data available to date. CONCLUSION: By acknowledging the limitations of the available preclinical data and by expanding our mechanistic understanding of EGFR inhibitor function in representative tumor model systems, we should enhance our capacity to predict the most rational and successful methods to combine EGFR inhibitors with cytotoxic modalities in future clinical trials.


Assuntos
Antineoplásicos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Apoptose/fisiologia , Terapia Combinada , Reparo do DNA , Humanos , Invasividade Neoplásica , Neovascularização Patológica/prevenção & controle , Tolerância a Radiação , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA