Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Acta Oncol ; 54(9): 1263-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26248024

RESUMO

Molecular imaging of specific biomarkers can have prognostic, predictive or monitoring value in head and neck squamous cell carcinoma (HNSCC). The epidermal growth factor receptor (EGFR) is involved in various radiation resistance mechanisms as it steers the pathways related to DNA damage repair, proliferation, hypoxia and apoptosis. Radiolabeled labeled F(ab')2 fragments of the EGFR antibody cetuximab can be applied for non-invasive imaging of this receptor. Preclinical studies have shown that radioresistant tumors had a higher tracer uptake after irradiation, probably due to upregulation of membranous EGFR, thereby increasing target availability possibly as a compensation mechanism. Tumors with increased EGFR availability were also more responsive to the EGFR inhibitor cetuximab. Potentially, radionuclide imaging of the EGFR can be applied for monitoring treatment regimens in clinical practice.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Escamosas , Receptores ErbB/análise , Neoplasias de Cabeça e Pescoço , Imagem Molecular/métodos , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Anticorpos Monoclonais/farmacologia , Cetuximab/farmacologia , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço
2.
Contrast Media Mol Imaging ; 11(1): 65-70, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26242487

RESUMO

Overexpression of the epidermal growth factor receptor (EGFR) is linked to an adverse outcome in various solid tumors. Cetuximab is an EGFR inhibitor, which in combination with radiotherapy improves locoregional control and survival in a subgroup of patients with head and neck squamous cell carcinomas (HNSCCs). The aim of this study was to develop and characterize an EGFR-directed PET tracer, (64) Cu-cetuximab-F(ab')2, to determine the systemic accessibility of EGFR. Mice with HNSCC xenografts, UT-SCC-8 (n = 6) or UT-SCC-45 (n = 6), were imaged 24 h post injection with (64) Cu-NODAGA-cetuximab-F(ab')2 using PET/CT. One mouse for each tumor model was co-injected with excess unlabeled cetuximab 3 days before radiotracer injection to determine non-EGFR-mediated uptake. Ex vivo biodistribution of the tracer was determined and tumors were analyzed by autoradiography and immunohistochemistry. The SUVmax of UT-SCC-8 tumors was higher than that of UT-SCC-45: 1.5 ± 1.0 and 0.8 ± 0.2 (p < 0.05), respectively. SUVmax after in vivo blocking of EGFR with cetuximab was 0.4. Immunohistochemistry showed that UT-SCC-8 had a significantly higher EGFR expression than UT-SCC-45: 0.50 ± 0.19 versus 0.12 ± 0.08 (p < 0.005), respectively. Autoradiography indicated that (64) Cu-cetuximab-F(ab')2 uptake correlated with EGFR expression in both tumors: r = 0.86 ± 0.06 (UT-SCC-8) and 0.90 ± 0.06 (UT-SCC-45). (64) Cu-cetuximab-F(ab')2 is a promising PET tracer to determine expression of EGFR in vivo. Clinically, this tracer has the potential to be used to determine cetuximab targeting of tumors and possibly to non-invasively monitor the response to EGFR-inhibitor treatment.


Assuntos
Carcinoma de Células Escamosas/diagnóstico por imagem , Meios de Contraste/administração & dosagem , Receptores ErbB/isolamento & purificação , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Animais , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Cetuximab/administração & dosagem , Cetuximab/química , Meios de Contraste/química , Radioisótopos de Cobre/administração & dosagem , Radioisótopos de Cobre/química , Receptores ErbB/biossíntese , Receptores ErbB/química , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Radiografia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Clin Cancer Res ; 21(16): 3602-9, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25931452

RESUMO

Approximately 50,000 new cases of head and neck squamous cell carcinoma (HNSCC) are diagnosed worldwide each year and subsequently treated with surgery, chemotherapy, radiotherapy, and/or targeted therapy. The heterogeneity of the patient population in terms of treatment response drives the search for tumor-specific biomarkers. Imaging of biomarkers can reveal patient-specific responses to therapies and, if assessed early after the start of treatment, may allow adaptation of treatment regimens. In this review, tracers that have been tested to monitor treatment efficacy in HNSCC by PET scanning prior to and early after the onset of treatment are discussed. An important imaging target for this application in HNSCC patients is the EGFR. It steers the pathways related to proliferation, hypoxia, DNA damage repair, and apoptosis, all treatment-resistance mechanisms. The anti-EGFR antibody cetuximab has been labeled with various radionuclides and has been tested as an imaging biomarker in several HNSCC models. These studies suggest that EGFR-targeting tracers can be used to monitor EGFR receptor expression in HNSCC and have the potential to noninvasively monitor cetuximab treatment and steer individualized treatment regimens. Multiple factors can influence the uptake of EGFR-targeting tracers. Here, we discuss the relevance of gene and protein overexpression, mutations, and amplifications related to EGFR signaling. In addition, monoclonal antibody properties and the effect on the host immune system are reviewed in light of the future role of EGFR-targeted imaging in HNSCC.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Receptores ErbB/genética , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Imagem Molecular , Biomarcadores Tumorais , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Cetuximab/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Tomografia por Emissão de Pósitrons , Radiografia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Resultado do Tratamento
4.
J Nucl Med ; 56(2): 287-92, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25552666

RESUMO

UNLABELLED: Treatment of head and neck squamous cell carcinomas with radiotherapy and the epidermal growth factor receptor (EGFR) inhibitor cetuximab shows an improved response in a subgroup of patients. The aim of this study was to noninvasively monitor treatment response by visualizing systemically accessible EGFR with (111)In-cetuximab-F(ab')2 while simultaneously evaluating tumor metabolism with (18)F-FDG PET during combined-modality treatment. METHODS: Eighty mice with patient-derived head and neck squamous cell carcinomas xenografts, SCCNij202 or SCCNij185, were imaged with SPECT/CT using (111)In-cetuximab-F(ab')2 (5 µg, 28 ± 6.1 MBq, 24 h after injection), followed by PET imaging with (18)F-FDG (9.4 ± 2.9 MBq, 1 h after injection). Scans were acquired on mice 10 d before treatment with either single-dose irradiation (10 Gy), cetuximab alone, or cetuximab-plus-irradiation combined or on untreated control mice. Scans were repeated 18 d after treatment. Tumor growth was monitored up to 120 d after treatment. EGFR expression was evaluated immunohistochemically. RESULTS: SCCNij202 responded to combined treatment (P < 0.01) and cetuximab treatment alone (P < 0.05) but not to irradiation alone (P = 0.13). SCCNij185 responded to combined treatment (P < 0.05) and irradiation (P < 0.05) but not to cetuximab treatment alone (P = 0.34). (111)In-cetuximab-F(ab')2 uptake (tumor-to-liver ratio, scan 2 - scan 1) predicted response to therapy. A positive response to treatment significantly correlated with a reduced tracer uptake in the tumor in the second SPECT scan, compared with the first scan (P < 0.005 and <0.05 for SCCNij202 and SCCNij185, respectively). Resistance to therapy was characterized by a significantly increased (111)In-cetuximab-F(ab')2 tumor uptake; tumor-to-liver ratio was 2.2 ± 0.6 to 3.5 ± 1.2, P < 0.01, for (irradiated) SCCNij202 and 1.4 ± 0.4 to 2.0 ± 0.3, P < 0.05, for (cetuximab-treated) SCCNij185, respectively. (18)F-FDG PET tumor uptake (maximum standardized uptake value, scan 2 - scan 1) correlated with tumor response for SCCNij202 (P < 0.01) but not for SCCNij185 (P = 0.66). EGFR fractions were significantly different: 0.9 ± 0.1 (SCCNij202) and 0.5 ± 0.1 (SCCNij185) (P < 0.001). The EGFR fraction was significantly lower for irradiated SCCNij202 tumors than for controls (P < 0.005). CONCLUSION: (111)In-cetuximab-F(ab')2 predicted and monitored the effects of EGFR inhibition or irradiation during treatment in both head and neck carcinoma models investigated, whereas (18)F-FDG PET only correlated with tumor response in the SCCNij202 model. Thus, the additional value of the (111)In-cetuximab-F(ab')2 tracer is emphasized and the tracer can aid in evaluating future treatments with EGFR-targeted therapies.


Assuntos
Anticorpos Monoclonais Humanizados , Fragmentos de Imunoglobulinas/química , Radioisótopos de Índio , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada de Emissão de Fóton Único , Animais , Biomarcadores Tumorais/metabolismo , Cetuximab , Terapia Combinada , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Fluordesoxiglucose F18 , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Tomografia Computadorizada por Raios X , Resultado do Tratamento
5.
J Nucl Med ; 55(10): 1665-70, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25236350

RESUMO

UNLABELLED: Only a subset of patients with head and neck squamous cell carcinomas (HNSCCs) benefit from radiotherapy and concurrent epidermal growth factor receptor (EGFR) inhibitor therapy with cetuximab, indicating the need for patient selection. The aim of this study was to visualize the change in systemically accessible EGFR with (111)In-cetuximab-F(ab')2 SPECT before and after radiotherapy, while simultaneously evaluating (18)F-FDG PET uptake. METHODS: Mice with HNSCC xenografts, cetuximab-sensitive SCCNij202 and cetuximab-resistant SCCNij167, were imaged with SPECT/CT using (111)In-cetuximab-F(ab')2 as a tracer, directly followed by PET imaging with (18)F-FDG. Scans were acquired 7 d before radiotherapy (10 Gy) and 1, 7, and 14 d after treatment. Intratumoral localization of (111)In-cetuximab-F(ab')(2) was evaluated by autoradiography and histologic markers evaluated by immunofluorescence staining in the same tumor sections. RESULTS: Growth of irradiated SCCNij202 and SCCNij167 tumors was significantly delayed, compared with controls (P < 0.05). No changes in uptake of (18)F-FDG were observed in either of the xenografts after radiotherapy. SPECT images of tumor-bearing mice showed a significant increase in uptake of (111)In-cetuximab-F(ab')(2) in the SCCNij202 tumors after irradiation (tumor-to-liver ratio, 4.3 ± 1.1 vs. 10.5 ± 3.3, 7 d before and 14 d after treatment, respectively, P < 0.01) but not in SCCNij167 tumors. Immunohistochemical EGFR staining showed a translocation of the EGFR from the cytoplasm to the cell membrane in irradiated SCCNij202 xenografts. Intratumoral distribution of (111)In-cetuximab-F(ab')(2) as determined by autoradiography correlated well with the distribution of EGFR as determined immunohistochemically (r = 0.85; range, 0.69-0.95). CONCLUSION: EGFR accessibility can be visualized with (111)In-cetuximab-F(ab')(2). (111)In-cetuximab-F(ab')(2) uptake increased after irradiation only in cetuximab-sensitive SCCNij202 xenografts, implying that the tracer can be used to measure irradiation-induced changes of EGFR expression and can monitor the compensatory response of tumors to radiotherapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Carcinoma de Células Escamosas/diagnóstico por imagem , Fluordesoxiglucose F18 , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Animais , Linhagem Celular Tumoral , Cetuximab , Modelos Animais de Doenças , Receptores ErbB/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Fragmentos de Imunoglobulinas/química , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Imagem Multimodal/métodos , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons/métodos , Radioterapia/métodos , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tomografia Computadorizada por Raios X/métodos
6.
J Nucl Med ; 55(2): 281-6, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24408894

RESUMO

UNLABELLED: Arginine-glycine-aspartic acid (RGD)-based imaging tracers allow specific imaging of integrin αvß3, a protein overexpressed during angiogenesis, leading to the possibility that it might serve as a tool to stratify patients for antiangiogenic treatment. However, these tracers have generally been characterized in xenograft models in which integrin αvß3 was constitutively expressed by the tumor cells themselves. In the studies presented here, the use of (111)In-RGD2 as a tracer to image only integrin αvß3 expression on blood vessels in the tumor was determined using tumor xenografts in which tumor cells were integrin αvß3-negative. METHODS: DOTA-E-[c(RGDfK)]2 was radiolabeled with (111)In ((111)In-RGD2), and biodistribution studies were performed in squamous cell carcinoma of the head and neck (HNSCC) xenograft mouse models to determine the optimal peptide dose to image angiogenesis. Next, biodistribution and imaging studies were performed at the optimal peptide dose in 3 HNSCC mouse models, FaDu, SCCNij3, and SCCNij202. Immunohistochemical analysis of tumor vascular and cell surface expression of integrin αvß3 and correlation analysis of vascular integrin αvß3 and autoradiography were completed. RESULTS: All 3 HNSCC xenografts expressed integrin αvß3 on the vessels only. The optimal peptide dose of (111)In-RGD2 was 1 µg or less for specific integrin αvß3-mediated uptake of the tracer. SPECT/CT imaging showed clear uptake of the tracer in the periphery of the tumors, corresponding with well-vascularized areas of the tumor. Within the tumor, (111)In-RGD2 autoradiography coincided with vascular integrin αvß3 expression, as determined immunohistochemically. Integrin αvß3-mediated uptake was also detected in nontumor tissues, which, through immunohistochemical analysis, proved positive for integrin αvß3. CONCLUSION: (111)In-RGD2 allows the visualization of integrin αvß3 in xenograft models in which integrin αvß3 is expressed only on the neovasculature, such as in the HNSCC tumors. Thus, (111)In-RGD2 allows specific visualization of angiogenesis in tumor models lacking constitutive tumoral integrin αvß3 expression but may be less useful for this purpose in many tumors in which tumor cells express integrin αvß3.


Assuntos
Vasos Sanguíneos/metabolismo , Carcinoma de Células Escamosas/diagnóstico por imagem , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Radioisótopos de Índio/química , Integrina alfaVbeta3/metabolismo , Neovascularização Patológica , Oligopeptídeos/química , Animais , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Feminino , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Compostos Radiofarmacêuticos/química , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tomografia Computadorizada por Raios X/métodos
7.
Radiother Oncol ; 108(3): 484-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23932156

RESUMO

BACKGROUND AND PURPOSE: Immunohistochemical epidermal growth factor receptor (EGFR) expression does not correlate with treatment response in head and neck squamous cell carcinomas (HNSCC). Aim was to apply the tracer (111)In-cetuximab-F(ab')2 for EGFR microSPECT imaging and to investigate if tracer uptake correlated with response to EGFR-inhibition by cetuximab in HNSCC xenografts. Usage of F(ab)2 fragments allows for shorter interval between tracer injection and imaging. MATERIALS AND METHODS: Mice with HNSCC xenografts, SCCNij202, 153, 185 and 167 were imaged with microSPECT using (111)In-cetuximab-F(ab')2. Subsequently, tumors were analyzed by autoradiography and immunohistochemistry and tracer concentration was determined. Tumor uptake was correlated with previously assessed response to cetuximab treatment. RESULTS: MicroSPECT imaging showed preferential uptake in HNSCC xenografts. Tumor-to-liver ratios were 3.1 ± 0.2 (SCCNij202), 2.8 ± 0.4 (SCCNij153), 2.0 ± 0.8 (SCCNij185), 2.0 ± 0.4 (SCCNij167). Immunohistochemical EGFR fractions (fEGFR) differed significantly between xenografts; 0.77 ± 0.07 (SCCNij202), 0.66 ± 0.11 (SCCNij153), 0.57 ± 0.19 (SCCNij185), 0.16 ± 0.10 (SCCNij167) (p < 0.001). Tumor fEGFR correlated with (111)In-cetuximab-F(ab')2 tumor uptake (r = 0.6, p < 0.01) and tracer autoradiography (r = 0.7, p < 0.0001). Tumor uptake of (111)In-cetuximab-F(ab')2 was proportionally associated with cetuximab treatment response in three out of four xenograft models. CONCLUSION: (111)In-cetuximab-F(ab')2 showed good tumor-to-background contrast on microSPECT imaging, allowing noninvasive assessment of EGFR expression in vivo, and possibly evaluation of treatment response to EGFR-inhibition.


Assuntos
Anticorpos Monoclonais Humanizados/metabolismo , Carcinoma de Células Escamosas/química , Receptores ErbB/análise , Neoplasias de Cabeça e Pescoço/química , Fragmentos Fab das Imunoglobulinas , Radioisótopos de Índio , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Autorradiografia , Linhagem Celular Tumoral , Cetuximab , Receptores ErbB/antagonistas & inibidores , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Carcinoma de Células Escamosas de Cabeça e Pescoço
8.
J Nucl Med ; 54(12): 2118-24, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24136932

RESUMO

UNLABELLED: Combined treatment of advanced head and neck squamous cell carcinomas (HNSCC) with radiotherapy and the epidermal growth factor receptor (EGFR) inhibitor cetuximab improves clinical outcome in comparison to radiotherapy alone but is effective only in a few cases. To select those patients most likely to benefit from EGFR inhibition, it can be advantageous to quantify the tumor EGFR status before and possibly during therapy. The aim of this study was to develop and characterize the (111)In-cetuximab-F(ab')2 tracer to image EGFR targeting in vivo. METHODS: The affinity and internalization kinetics of (111)In-cetuximab-F(ab')2 were determined in vitro. The optimal protein-fragment dose for imaging was determined in nude mice with a subcutaneous head and neck carcinoma model (FaDu). Mice with FaDu tumors were imaged using ultra-high-resolution SPECT with (111)In-cetuximab-F(ab')2 or (111)In-cetuximab IgG at 4, 24, 48, and 168 h after injection. Tumor tracer uptake was determined on micro-SPECT and autoradiography images of tumor sections. Immunohistochemical staining was used to analyze EGFR expression in the tumor. RESULTS: In vitro, more than 50% of (111)In-cetuximab-F(ab')2 was internalized into FaDu cells within 24 h. The half maximal inhibitory concentration (IC50) of (111)In-cetuximab-F(ab')2 and (111)In-cetuximab was similar: 0.42 ± 0.16 nM versus 0.28 ± 0.14 nM, respectively. The protein dose-escalation study showed that the highest uptake of (111)In-cetuximab-F(ab')2 in tumors was obtained at doses of 10 µg/mouse or less (13.5 ± 5.2 percentage injected dose per gram [%ID/g]). Tumor uptake of (111)In-cetuximab was significantly higher (26.9 ± 3.3 %ID/g, P < 0.01). However, because of rapid blood clearance, tumor-to-blood ratios at 24 h after injection were significantly higher for (111)In-cetuximab-F(ab')2 (31.4 ± 3.8 vs. 1.7 ± 0.2, respectively; P < 0.001). The intratumoral distribution of (111)In-cetuximab-F(ab')2 correlated well with the immunohistochemical distribution of EGFR (r = 0.64 ± 0.06, P < 0.0001). micro-SPECT images of (111)In-cetuximab-F(ab')2 clearly showed preferential uptake in the tumor from 4 h onward, with superior tumor-to-background contrast at 24 h, compared with (111)In-cetuximab (107.0 ± 17.0 vs. 69.7 ± 3.9, respectively; P < 0.05). CONCLUSION: (111)In-cetuximab-F(ab')2 displays higher tumor-to-blood ratios early after injection than (111)In-cetuximab in an HNSCC model, making it more suitable for EGFR visualization and potentially for selecting patients for treatment with EGFR inhibitors.


Assuntos
Anticorpos Monoclonais Humanizados , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/diagnóstico , Fragmentos de Imunoglobulinas/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Animais , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais Humanizados/farmacocinética , Autorradiografia , Linhagem Celular Tumoral , Cetuximab , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Radioisótopos de Índio , Camundongos , Camundongos Endogâmicos BALB C , Imagem Multimodal , Doses de Radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA