Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Biol Chem ; 402(10): 1247-1256, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34363384

RESUMO

Renal cell carcinoma (RCC) is the third most frequent malignancy within urological oncology. Sunitinib has been used as the standard of treatment for first-line RCC therapy. Understanding mechanisms of sunitinib resistance in RCC cell is important for clinical therapy and drug development. We established sunitinib resistant RCC cells by treating cells with increasing concentrations of sunitinib and named resistant cells as RCC/SR. Lefty A, an important embryonic morphogen, was increased in RCC/SR cells. Targeted inhibition of Lefty via its siRNAs restored the sensitivity of renal resistant cells to sunitinib treatment. It was due to that si-Lefty can decrease the expression of interleukin-8 (IL-8) in RCC/SR cells. Knockdown of IL-8 abolished Lefty-regulated sunitinib sensitivity of RCC cells. Mechanistically, Lefty can regulate IL-8 transcription via activation of p65, one major transcription factor of IL-8. Collectively, our present revealed that Lefty A can regulate sunitinib sensitivity of RCC cells of via NF-κB/IL-8 signals. It indicated that targeted inhibition of Lefty might be a potent approach to overcome sunitinib resistance of RCC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Sunitinibe , Humanos , Interleucina-8/metabolismo
2.
Theranostics ; 11(7): 3527-3539, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33537102

RESUMO

To investigate the utility of noninvasive µPET-CT with 64Cu-DOTA-anti-CD11b (64Cu-αCD11b) in assessing bone marrow status after anticancer therapies, and the protective role of anti-CSF-1 (αCSF-1) against bone marrow suppression induced by Abraxane. Methods: MDA-MB-435 tumor-bearing mice were treated with Abraxane, αCSF-1, or αCSF-1 plus Abraxane. µPET-CT and biodistribution of 64Cu-αCD11b were performed after intravenous injection of the radiotracer. Cells from mouse bone marrow and MDA-MB-435 tumor were analyzed by flow cytometry. A humanized αCSF-1 was investigated for its role in protecting bone marrow cells, using a transgenic mouse model that expresses functional human CSF-1. Results: µPET-CT showed that 64Cu-αCD11b had high uptake in the bone marrow and spleen of both normal and tumor-bearing mice. Abraxane significantly reduced 64Cu-αCD11b uptake in the bone marrow and spleen of treated mice compared to untreated mice. Interestingly, 64Cu-αCD11b µPET-CT revealed that αCSF-1 alleviated the depletion of bone marrow cells by Abraxane. These changes in the bone marrow population of CD11b+ myeloid cells were confirmed by flow cytometry. Moreover, αCSF-1 potently enhanced tolerance of bone marrow granulocytic myeloid cells to Abraxane, decreased cell migration, and suppressed recruitment of myeloid cells to the tumor microenvironment. The humanized αCSF-1 also alleviated the effects of Abraxane on bone marrow cells in transgenic mice expressing human CSF-1, suggesting clinical relevance of αCSF-1 in prevention of bone marrow suppression in addition to its role in reducing tumor-infiltrating myeloid cells. Conclusions: Abraxane-induced bone marrow CD11b+ myeloid cell depletion in tumor-bearing mice could be noninvasively assessed by µPET-CT with 64Cu-αCD11b and prevented by αCSF-1.


Assuntos
Paclitaxel Ligado a Albumina/toxicidade , Anticorpos/farmacologia , Antineoplásicos/toxicidade , Medula Óssea/diagnóstico por imagem , Neoplasias/diagnóstico por imagem , Baço/diagnóstico por imagem , Paclitaxel Ligado a Albumina/antagonistas & inibidores , Animais , Anticorpos/química , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Medula Óssea/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Linhagem Celular Tumoral , Radioisótopos de Cobre , Feminino , Expressão Gênica , Compostos Heterocíclicos com 1 Anel/química , Xenoenxertos , Humanos , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/imunologia , Camundongos , Camundongos Nus , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/patologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/química , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/química , Baço/efeitos dos fármacos , Baço/imunologia , Baço/patologia , Microambiente Tumoral/efeitos dos fármacos
3.
Nanomedicine ; 25: 102169, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32059873

RESUMO

Generation of durable tumor-specific immune response without isolation and expansion of dendritic cells or T cells ex vivo remains a challenge. In this study, we investigated the impact of nanoparticle-mediated photothermolysis in combination with checkpoint inhibition on the induction of systemic antitumor immunity. Photothermolysis based on near-infrared light-absorbing copper sulfide nanoparticles and 15-ns laser pulses combined with the immune checkpoint inhibitor anti-PD-1 antibody (αPD-1) increased tumor infiltration by antigen-presenting cells and CD8-positive T lymphocytes in the B16-OVA mouse model. Moreover, combined photothermolysis, polymeric conjugate of the Toll-like receptor 9 agonist CpG, and αPD-1 significantly prolonged mouse survival after re-inoculation of tumor cells at a distant site compared to individual treatments alone in the poorly immunogenic syngeneic ID8-ip1-Luc ovarian tumor model. Thus, photothermolysis is a promising interventional technique that synergizes with Toll-like receptor 9 agonists and immune checkpoint inhibitors to enhance the abscopal effect in tumors.


Assuntos
Melanoma Experimental/tratamento farmacológico , Terapia Fototérmica , Receptor de Morte Celular Programada 1/genética , Receptor Toll-Like 9/genética , Animais , Terapia Combinada , Modelos Animais de Doenças , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade Inata/efeitos dos fármacos , Imunoterapia/métodos , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptor Toll-Like 9/agonistas
4.
J Nucl Med ; 60(9): 1317-1324, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30796172

RESUMO

Current noninvasive imaging methods for monitoring immune response were largely developed for interrogation of the local reaction. This study developed the radiotracer 64Cu-labeled anti-CD11b (64Cu-αCD11b) for longitudinal assessment of local and systemic immune response involving mobilization of CD11b+ myeloid cells by small-animal PET/CT. Methods: Acute or chronic inflammation in the ears of BALB/c mice was induced by 12-o-tetradecanoylphorbol-13-acetate. Acute lung inflammation was induced by intratracheal lipopolysaccharide inoculation. αCD11b was conjugated with p-SCN-Bn-DOTA followed by labeling with 64Cu. PET/CT and biodistribution were evaluated at different times after intravenous injection of 64Cu-αCD11b. Cell populations from bone marrow (BM) and spleen were analyzed by flow cytometry. Results:64Cu-αCD11b was primarily taken up by BM and spleen in control mice. In comparison, 64Cu-αCD11b uptake was significantly reduced in the BM and spleen of CD11b-knockout mice, indicating that 64Cu-αCD11b selectively homed to CD11b+ myeloid cells in vivo. In mice with ear inflammation, for the local inflammatory response, 64Cu-αCD11b PET/CT revealed significantly higher 64Cu-αCD11b uptake in the inflamed ears in the acute inflammation phase than the chronic phase, consistent with markedly increased infiltration of CD11b+ cells into the inflammatory lesions at the acute phase. Moreover, imaging of 64Cu-αCD11b also showed the difference in mouse systemic response for different inflammatory stages. Compared with uptake in control mice, BM 64Cu-αCD11b uptake in mice with ear inflammation was significantly lower in the acute phase and higher in the chronic phase, reflecting an initial mobilization of CD11b+ cells from the BM to the inflammatory foci followed by a compensatory regeneration of CD11b+ myeloid cells in the BM. Similarly, in mice with lung inflammation, 64Cu-αCD11b PET/CT readily detected acute lung inflammation and recruitment of CD11b+ myeloid cells from the BM. Immunohistochemistry staining and flow cytometry results confirmed the noninvasive imaging of PET/CT. Conclusion:64Cu-αCD11b PET/CT successfully tracked ear and pulmonary inflammation in mice and differentiated acute from chronic inflammation at the local and systemic levels. 64Cu-αCD11b PET/CT is a robust quantitative method for imaging of local and systemic immune responses.


Assuntos
Antígeno CD11b/química , Radioisótopos de Cobre/química , Sistema Imunitário/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Animais , Medula Óssea/diagnóstico por imagem , Medula Óssea/metabolismo , Feminino , Citometria de Fluxo , Compostos Heterocíclicos/química , Imuno-Histoquímica , Isotiocianatos/química , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Pneumonia/diagnóstico por imagem , Compostos Radiofarmacêuticos , Ratos , Baço/diagnóstico por imagem , Baço/metabolismo , Distribuição Tecidual
5.
ACS Appl Bio Mater ; 2(8): 3203-3211, 2019 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-33907729

RESUMO

Although multifunctional inorganic nanoparticles have been extensively explored for effective cancer diagnosis and therapy, their clinical translation has been greatly impeded because of significant uptake in the reticuloendothelial system and concerns about potential toxicity. In this study, we uncovered the thermosensitive biodegradability of CuS nanoparticles, which have classically been considered as stable in bulk state. Polyethylene glycol (PEG)-coated CuS nanoparticles (CuS-PEG) were well preserved at 4 ºC but were rapidly degraded at 37 ºC within 1 week in both in vitro and in vivo tests. Furthermore, real-time multispectral optoacoustic tomography, which is more convenient and accurate than traditional ex vivo analysis, was successfully employed to noninvasively demonstrate the biodegradability of CuS-PEG nanoparticles and dynamically monitor their tumor imaging capacity. The temperature-dependent controllable degradation profile and excellent tumor retention of CuS-PEG nanoparticles endows them with great potential for clinical applications since it ensures that the nanoparticles remain intact during production, transportation, and storage but degrade and clear from the body at physiological temperature after accomplishing sufficient diagnosis and therapeutic operations.

6.
Biomaterials ; 152: 63-76, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29111494

RESUMO

As a result of therapy-induced apoptosis, peripheral blood monocytes are recruited to tumors, where they become tumor-associated macrophages (TAMs). To date, few studies have investigated noninvasive molecular imaging for assessment of macrophage infiltration in response to therapy-induced apoptosis. Here, noninvasive assessment of changes in tumor accumulation of TAMs was proposed as a new way to measure early tumor response to anticancer therapy. Three different nanoparticles, QD710-Dendron quantum dots (QD710-D), Ferumoxytol, and PG-Gd-NIR813, were used for near-infrared fluorescence imaging, T2-weighted magnetic resonance imaging, and dual optical/T1-weighted MR imaging, respectively, in the MDA-MB-435 tumor model. Treatment with Abraxane induced tumor apoptosis and infiltrating macrophages. In spite of markedly different physicochemical properties among the nanoparticles, in vivo imaging revealed increased uptake of all three nanoparticles in Abraxane-treated tumors compared with untreated tumors. Moreover, imaging visualized increased uptake of QD710-D in MDA-MB-435 tumors but not in drug-resistant MDA-MB-435R tumors grown in the mice treated with Abraxane. Our results suggest that infiltration of macrophages due to chemotherapy-induced apoptosis was partially responsible for increased nanoparticle uptake in treated tumors. Noninvasive imaging techniques in conjunction with systemic administration of imageable nanoparticles that are taken up by macrophages are a potentially useful tool for assessing early treatment response.


Assuntos
Antineoplásicos/química , Meios de Contraste/química , Macrófagos/metabolismo , Imageamento por Ressonância Magnética/métodos , Imagem Óptica/métodos , Animais , Antineoplásicos/farmacologia , Benzamidas/química , Linhagem Celular Tumoral , Óxido Ferroso-Férrico/química , Indóis/química , Macrófagos/patologia , Camundongos Nus , Nanopartículas/química , Tamanho da Partícula , Pontos Quânticos/química , Propriedades de Superfície , Microambiente Tumoral
7.
Radiology ; 281(2): 427-435, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27347765

RESUMO

Purpose To determine if combretastatin A-4 phosphate disodium (CA4P) can enhance the tumor uptake of doxorubicin (Dox)-loaded, polyethylene glycol (PEG)-coated hollow gold nanospheres (HAuNS) mixed with ethiodized oil for improved photothermal ablation (PTA)-chemoembolization therapy (CET) of hepatocellular carcinoma (HCC) in rats. Materials and Methods Animal experiments were approved by the institutional animal care and use committee and performed from February 2014 to April 2015. Male Sprague-Dawley rats (n = 45; age, 12 weeks) were inoculated with N1S1 HCC cells in the liver, and 8 days later, were randomly divided into two groups of 10 rats. Group 1 rats received intrahepatic arterial injection of PEG-HAuNS and ethiodized oil alone; group 2 received pretreatment with CA4P and injection of PEG-HAuNS and ethiodized oil 5 minutes later. The gold content of tumor and liver tissue at 1 hour or 24 hours after injection was quantified by using neutron activation analysis (n = 5 per time point). Five rats received pretreatment CA4P, PEG-copper 64-HAuNS, and ethiodized oil and underwent micro-positron emission tomography (PET)/computed tomography (CT). In a separate study, three groups of six rats with HCC were injected with saline solution (control group); CA4P, Dox-loaded PEG-coated HAuNS (Dox@PEG-HAuNS), and ethiodized oil (CET group); or CA4P, Dox@PEG-HAuNS, ethiodized oil, and near-infrared irradiation (PTA-CET group). Temperature was recorded during laser irradiation. Findings were verified at postmortem histopathologic and/or autoradiographic examination. Wilcoxon rank-sum test and Pearson correlation analyses were performed. Results PEG-HAuNS uptake in CA4P-pretreated HCC tumors was significantly higher than that in non-CA4P-pretreated tumors at both 1 hour (P < .03) and 24 hours (P < .01). Mean ± standard deviation of tumor-to-liver PEG-HAuNS uptake ratios at 1 hour and 24 hours, respectively, were 5.63 ± 3.09 and 1.68 ± 0.77 in the CA4P-treated group and 1.29 ± 2.40 and 0.14 ± 0.11 in the non-CA4P-treated group. Micro-PET/CT allowed clear delineation of tumors, enabling quantitative imaging analysis. Laser irradiation increased temperature to 60°C and 43°C in the tumor and adjacent liver, respectively. Mean HCC tumor volumes 10 days after therapy were 1.68 cm3 ± 1.01, 3.96 cm3 ± 1.75, and 6.13 cm3 ± 2.27 in the PTA-CET, CET, and control groups, respectively, with significant differences between the PTA-CET group and other groups (P < .05). Conclusion CA4P pretreatment caused a higher concentration of Dox@PEG-HAuNS to be trapped inside the tumor, thereby enhancing the efficacy of anti-HCC treatment with PTA-CET in rats. © RSNA, 2016 Online supplemental material is available for this article.


Assuntos
Carcinoma Hepatocelular/terapia , Quimioembolização Terapêutica/métodos , Doxorrubicina/farmacologia , Portadores de Fármacos/farmacocinética , Ouro/farmacocinética , Neoplasias Hepáticas/terapia , Animais , Carcinoma Hepatocelular/diagnóstico por imagem , Linhagem Celular Tumoral , Modelos Animais de Doenças , Doxorrubicina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Óleo Etiodado , Ouro/administração & dosagem , Hipertermia Induzida , Neoplasias Hepáticas/diagnóstico por imagem , Masculino , Nanosferas , Polietilenoglicóis , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Estilbenos/farmacologia
9.
Biomed Res Int ; 2013: 759057, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23710458

RESUMO

Hepatocellular carcinoma (HCC) is a highly aggressive and lethal cancer. It is typically asymptomatic at the early stage, with only 10%-20% of HCC patients being diagnosed early enough for appropriate surgical treatment. The delayed diagnosis of HCC is associated with limited treatment options and much lower survival rates. Therefore, the early and accurate detection of HCC is crucial to improve its currently dismal prognosis. The epidermal growth factor receptor (EGFR) has been reported to be involved in HCC tumorigenesis and to represent an attractive target for HCC imaging and therapy. In this study, an affibody molecule, Ac-Cys-ZEGFR:1907, targeting the extracellular domain of EGFR, was used for the first time to assess its potential to detect HCC xenografts. By evaluating radio- or fluorescent-labeled Ac-Cys-ZEGFR:1907 as a probe for positron emission tomography (PET) or optical imaging of HCC, subcutaneous EGFR-positive HCC xenografts were found to be successfully imaged by the PET probe. Thus, affibody-based PET imaging of EGFR provides a promising approach for detecting HCC in vivo.


Assuntos
Carcinoma Hepatocelular/patologia , Receptores ErbB/química , Neoplasias Hepáticas/patologia , Imagem Molecular , Animais , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Detecção Precoce de Câncer , Receptores ErbB/genética , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Camundongos , Estadiamento de Neoplasias , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Amino Acids ; 41(5): 1103-12, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20232090

RESUMO

Early evaluation of cancer response to a therapeutic regimen can help increase the effectiveness of treatment schemes and, by enabling early termination of ineffective treatments, minimize toxicity, and reduce expenses. Biomarkers that provide early indication of tumor therapy response are urgently needed. Solid tumors require blood vessels for growth, and new anti-angiogenic agents can act by preventing the development of a suitable blood supply to sustain tumor growth. The purpose of this study is to develop a class of novel molecular imaging probes that will predict tumor early response to an anti-angiogenic regimen with the humanized vascular endothelial growth factor antibody bevacizumab. Using a bevacizumab-sensitive LS174T colorectal cancer model and a 12-mer bacteriophage (phage) display peptide library, a bevacizumab-responsive peptide (BRP) was identified after six rounds of biopanning and tested in vitro and in vivo. This 12-mer peptide was metabolically stable and had low toxicity to both endothelial cells and tumor cells. Near-infrared dye IRDye800-labeled BRP phage showed strong binding to bevacizumab-treated tumors, but not to untreated control LS174T tumors. In addition, both IRDye800- and (18)F-labeled BRP peptide had significantly higher uptake in tumors treated with bevacizumab than in controls treated with phosphate-buffered saline. Ex vivo histopathology confirmed the specificity of the BRP peptide to bevacizumab-treated tumor vasculature. In summary, a novel 12-mer peptide BRP selected using phage display techniques allowed non-invasive visualization of early responses to anti-angiogenic treatment. Suitably labeled BRP peptide may be potentially useful pre-clinically and clinically for monitoring treatment response.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Monitoramento de Medicamentos/métodos , Sondas Moleculares , Peptídeos , Sequência de Aminoácidos , Animais , Bevacizumab , Linhagem Celular Tumoral , Neoplasias Colorretais/diagnóstico por imagem , Monitoramento de Medicamentos/instrumentação , Feminino , Humanos , Camundongos , Camundongos Nus , Sondas Moleculares/química , Sondas Moleculares/genética , Dados de Sequência Molecular , Biblioteca de Peptídeos , Peptídeos/química , Peptídeos/genética , Tomografia por Emissão de Pósitrons
11.
J Nucl Med ; 52(1): 140-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21149494

RESUMO

UNLABELLED: Abraxane (nanoparticle albumin-bound paclitaxel) is an anticancer drug approved by the Food and Drug Administration. However, the mechanism of action of Abraxane is complex, and no established biomarker is available to accurately monitor its treatment outcomes. The aim of this study was to investigate whether the integrin-specific PET tracer 18F-FPPRGD2 (investigational new drug 104150) can be used to monitor early response of tumors to Abraxane therapy. METHODS: Orthotopic MDA-MB-435 breast cancer mice were treated with Abraxane (25 mg/kg every other day, 3 doses) or phosphate-buffered saline. Tumor volume was monitored by caliper measurement. PET scans were obtained before and at different times after the start of treatment (days 0, 3, 7, 14, and 21) using 18F-FPPRGD2 and 18F-FDG. The tumoricidal effect was also assessed ex vivo by immunohistochemistry. RESULTS: Abraxane treatment inhibited the tumor growth, and a significant difference in tumor volume could be seen at day 5 after the initiation of treatment. The tumor uptake of 18F-FPPRGD2 in the Abraxane-treated group was significantly lower on days 3 and 7 than at baseline but returned to the baseline level at days 14 and 21, indicative of relapse of the tumors after the treatment was halted. Immunohistologic staining confirmed that the change of 18F-FPPRGD2 uptake correlated with the variation of integrin level in the tumor vasculature induced by Abraxane treatment. No significant change of tumor (rather than vascular) integrin expression was observed throughout the study. No significant decrease of 18F-FDG uptake was found between the treated and the control tumors on days 3, 14, and 21, although an increase in 18F-FDG tumor uptake of treated mice, as compared with the control mice, was found on day 7. The increase of 18F-FDG on day 7 was related to the inflammatory response during therapy. CONCLUSION: Abraxane-mediated downregulation of integrin αvß3 expression on tumor endothelial cells can be quantitatively visualized by PET. The change of integrin expression precedes that of tumor size. Consequently, 18F-FPPRGD2 PET is superior to 18F-FDG PET in monitoring early response to treatment, favoring its potential clinical translation.


Assuntos
Antineoplásicos/uso terapêutico , Radioisótopos de Flúor , Fluordesoxiglucose F18 , Integrina alfaVbeta3/análise , Neoplasias Mamárias Experimentais/tratamento farmacológico , Oligopeptídeos/metabolismo , Paclitaxel/uso terapêutico , Peptídeos Cíclicos/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos , Paclitaxel Ligado a Albumina , Albuminas/uso terapêutico , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Integrina alfaVbeta3/antagonistas & inibidores , Neoplasias Mamárias Experimentais/patologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Clin Cancer Res ; 16(7): 2095-105, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20215534

RESUMO

PURPOSE: To show the relationship between antibody delivery and therapeutic efficacy in head and neck cancers, in this study we evaluated the pharmacokinetics and pharmacodynamics of epidermal growth factor receptor (EGFR)-targeted immunotherapy and radioimmunotherapy by quantitative positron emission tomography (PET) imaging. EXPERIMENTAL DESIGN: EGFR expression on UM-SCC-22B and SCC1 human head and neck squamous cell cancer (HNSCC) cells were determined by flow cytometry and immunostaining. Tumor delivery and distribution of cetuximab in tumor-bearing nude mice were evaluated with small animal PET using (64)Cu-DOTA-cetuximab. The in vitro toxicity of cetuximab to HNSCC cells was evaluated by MTT assay. The tumor-bearing mice were then treated with four doses of cetuximab at 10 mg/kg per dose, and tumor growth was evaluated by caliper measurement. FDG PET was done after the third dose of antibody administration to evaluate tumor response. Apoptosis and tumor cell proliferation after cetuximab treatment were analyzed by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and Ki-67 staining. Radioimmunotherapy was done with (90)Y-DOTA-cetuximab. RESULTS: EGFR expression on UM-SCC-22B cells is lower than that on SCC1 cells. However, the UM-SCC-22B tumors showed much higher (64)Cu-DOTA-cetuximab accumulation than the SCC1 tumors. Cetuximab-induced apoptosis in SCC1 tumors and tumor growth was significantly inhibited, whereas an agonistic effect of cetuximab on UM-SCC-22B tumor growth was observed. After cetuximab treatment, the SCC1 tumors showed decreased FDG uptake, and the UM-SCC-22B tumors had increased FDG uptake. UM-SCC-22B tumors are more responsive to (90)Y-DOTA-cetuximab treatment than SCC1 tumors, partially due to the high tumor accumulation of the injected antibody. CONCLUSION: Cetuximab has an agonistic effect on the growth of UM-SCC-22B tumors, indicating that tumor response to cetuximab treatment is not necessarily related to EGFR expression and antibody delivery efficiency, as determined by PET imaging. Although PET imaging with antibodies as tracers has limited function in patient screening, it can provide guidance for targeted therapy using antibodies as delivery vehicles.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/terapia , Imunoterapia , Radioimunoterapia , Animais , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/radioterapia , Linhagem Celular Tumoral , Cetuximab , Radioisótopos de Cobre/uso terapêutico , Feminino , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Neoplasias de Cabeça e Pescoço/radioterapia , Compostos Heterocíclicos/uso terapêutico , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Nus , Compostos Organometálicos/uso terapêutico , Tomografia por Emissão de Pósitrons , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Nucl Med ; 50(7): 1168-77, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19525469

RESUMO

UNLABELLED: The overexpression of gastrin-releasing peptide receptor (GRPR) in various tumor types suggests that GRPR is an attractive target for cancer imaging and therapy with radiolabeled bombesin analogs. We recently reported the ability of (18)F-labeled RGD-bombesin heterodimer to be used for dual integrin alpha(v)beta(3)- and GRPR-targeted imaging. To further investigate the synergistic effect of the dual-receptor targeting of peptide heterodimers, we evaluated (64)Cu-labeled RGD-bombesin for PET imaging of tumors. METHODS: RGD-bombesin was coupled with 1,4,7,10-tetraazacyclododecane-N, N', N'', N'''-tetraacetic acid (DOTA) and 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA), and the conjugates were labeled with (64)Cu. The in vitro and in vivo characteristics of (64)Cu-NOTA-RGD-bombesin were compared with those of (64)Cu-NOTA-RGD, (64)Cu-NOTA-bombesin, and (64)Cu-DOTA-RGD-bombesin. RESULTS: (64)Cu-NOTA-RGD-bombesin and (64)Cu-DOTA-RGD-bombesin had comparable dual integrin alpha(v)beta(3)- and GRPR-binding affinities in vitro, both of which were slightly lower than RGD for integrin binding and bombesin for GRPR binding. (64)Cu-NOTA-RGD-bombesin possessed significantly higher tumor uptake than did (64)Cu-NOTA-RGD, (64)Cu-NOTA-bombesin, the mixture of (64)Cu-NOTA-RGD and (64)Cu-NOTA-bombesin, or (64)Cu-DOTA-RGD-bombesin in PC-3 prostate cancer. (64)Cu-NOTA-RGD-bombesin also showed improved in vivo kinetics such as lower liver and intestinal activity accumulation than did the bombesin tracers. (64)Cu-NOTA-RGD-bombesin also outperformed (64)Cu-NOTA-RGD in a 4T1 murine mammary carcinoma model that expresses integrin on tumor vasculature but no GRPR in tumor tissue, which had no uptake of (64)Cu-NOTA-bombesin. CONCLUSION: Compared with other tracers, (64)Cu-NOTA-RGD-bombesin showed favorable in vivo kinetics and enhanced tumor uptake, which warrants its further investigation for targeting tumors that express integrin or GRPR or that coexpress integrin and GRPR for imaging and therapeutic applications. The synergistic effect of RGD-bombesin heterodimers observed in this study also encourages further investigations of novel heterodimers recognizing other cell surface receptors for tumor targeting.


Assuntos
Bombesina/farmacocinética , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/metabolismo , Receptores da Bombesina/metabolismo , Animais , Bombesina/química , Linhagem Celular Tumoral , Radioisótopos de Cobre/farmacocinética , Dimerização , Humanos , Masculino , Taxa de Depuração Metabólica , Camundongos , Oligopeptídeos/farmacocinética , Especificidade de Órgãos , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
14.
Eur J Nucl Med Mol Imaging ; 36(9): 1510-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19440708

RESUMO

PURPOSE: 17-Dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), a heat-shock protein 90 (Hsp90) inhibitor, has been intensively investigated for cancer therapy and is undergoing clinical trials. Human epidermal growth factor receptor 2 (HER-2) is one of the client proteins of Hsp90 and its expression is decreased upon 17-DMAG treatment. In this study, we aimed to noninvasively monitor the HER-2 response to 17-DMAG treatment in xenografted mice. METHODS: The sensitivity of human ovarian cancer SKOV-3 cells to 17-DMAG in vitro was measured by MTT assay. HER-2 expression in SKOV-3 cells was determined by flow cytometry. Nude mice bearing SKOV-3 tumors were treated with 17-DMAG and the therapeutic efficacy was evaluated by tumor size measurement. Both treated and control mice were imaged with microPET using (64)Cu-DOTA-trastuzumab and (18)F-FDG. Biodistribution studies and immunofluorescence staining were performed to validate the microPET results. RESULTS: SKOV-3 cells are sensitive to 17-DMAG treatment, in a dose-dependent manner, with an IC(50) value of 24.72 nM after 72 h incubation. The tumor growth curve supported the inhibition effect of 17-DMAG on SKOV-3 tumors. Quantitative microPET imaging showed that (64)Cu-DOTA-trastuzumab had prominent tumor accumulation in untreated SKOV-3 tumors, which was significantly reduced in 17-DMAG-treated tumors. There was no uptake difference detected by FDG PET. Immunofluorescence staining confirmed the significant reduction in tumor HER-2 level upon 17-DMAG treatment. CONCLUSION: The early response to anti-Hsp90 therapy was successfully monitored by quantitative PET using (64)Cu-DOTA-trastuzumab. This approach may be valuable in monitoring the therapeutic response in HER-2-positive cancer patients under 17-DMAG treatment.


Assuntos
Anticorpos Monoclonais , Benzoquinonas/farmacologia , Lactamas Macrocíclicas/farmacologia , Compostos Organometálicos , Neoplasias Ovarianas/diagnóstico por imagem , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Benzoquinonas/uso terapêutico , Linhagem Celular Tumoral , Radioisótopos de Cobre , Feminino , Fluordesoxiglucose F18 , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Lactamas Macrocíclicas/uso terapêutico , Camundongos , Camundongos Nus , Transplante de Neoplasias , Compostos Organometálicos/farmacocinética , Neoplasias Ovarianas/tratamento farmacológico , Tomografia por Emissão de Pósitrons , Receptor ErbB-2/biossíntese , Distribuição Tecidual , Trastuzumab
15.
Clin Cancer Res ; 14(19): 6137-45, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829492

RESUMO

PURPOSE: Interleukin (IL)-18 plays important roles in cancer progression and metastasis. The goal of this study is to identify cell lines that are most sensitive to stand alone IL-18-binding protein (IL-18bp)-Fc treatment, to study the pharmacokinetics and tumor targeting efficiency of IL-18bp-Fc, and to evaluate the efficacy of IL-18bp-Fc in treating breast cancer experimental lung metastasis by multimodality imaging. EXPERIMENTAL DESIGN: Reverse transcription-PCR, ELISA, and other cell-based assays were done on murine 4T1, CT-26, and B16F10 cells. The most IL-18bp-Fc-sensitive 4T1 cells were stably transfected with firefly luciferase (fLuc) and injected i.v. into female BALB/C mice to establish the experimental lung metastasis model. Tumor targeting efficiency and pharmacokinetics of IL-18bp-Fc was assessed by (64)Cu-DOTA-IL-18bp-Fc positron emission tomography (PET) and biodistribution studies. Two groups of fLuc-4T1 experimental lung metastasis tumor-bearing mice were each given saline or IL-18bp-Fc (1 mg/kg) daily i.p. Bioluminescence imaging, (18)F-FDG PET, and computed tomography scans were done to evaluate the treatment efficacy. Ex vivo experiments were also carried out to validate the imaging results. RESULTS: IL-18bp-Fc had high and specific accumulation in the fLuc-4T1 lung metastasis tumor as evidenced by both PET and biodistribution studies. Bioluminescence imaging, (18)F-FDG PET, and computed tomography scans all revealed that IL-18bp-Fc treatment was effective in inhibiting the lung metastasis tumor progression, validated by ex vivo examination of the lung. CONCLUSIONS: IL-18bp-Fc therapy can inhibit 4T1 breast cancer experimental lung metastasis. Noninvasive multimodality molecular imaging is a powerful tool for evaluating the tumor targeting efficiency/pharmacokinetics of the drug and effective monitoring of the therapeutic response.


Assuntos
Fragmentos Fc das Imunoglobulinas/química , Imunoterapia/métodos , Peptídeos e Proteínas de Sinalização Intercelular/química , Interleucina-18/química , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Animais , Neoplasias da Mama , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Fragmentos Fc das Imunoglobulinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-18/metabolismo , Melanoma Experimental , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons/métodos , Tomografia Computadorizada por Raios X/métodos
16.
Cancer Res ; 68(16): 6652-60, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18701489

RESUMO

Chemically functionalized single-walled carbon nanotubes (SWNT) have shown promise in tumor-targeted accumulation in mice and exhibit biocompatibility, excretion, and little toxicity. Here, we show in vivo SWNT drug delivery for tumor suppression in mice. We conjugate paclitaxel (PTX), a widely used cancer chemotherapy drug, to branched polyethylene glycol chains on SWNTs via a cleavable ester bond to obtain a water-soluble SWNT-PTX conjugate. SWNT-PTX affords higher efficacy in suppressing tumor growth than clinical Taxol in a murine 4T1 breast cancer model, owing to prolonged blood circulation and 10-fold higher tumor PTX uptake by SWNT delivery likely through enhanced permeability and retention. Drug molecules carried into the reticuloendothelial system are released from SWNTs and excreted via biliary pathway without causing obvious toxic effects to normal organs. Thus, nanotube drug delivery is promising for high treatment efficacy and minimum side effects for future cancer therapy with low drug doses.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Nanotubos de Carbono/química , Paclitaxel/administração & dosagem , Animais , Feminino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Polietilenoglicóis/química , Análise Espectral Raman , Distribuição Tecidual , Células Tumorais Cultivadas
17.
Eur J Nucl Med Mol Imaging ; 35(8): 1489-98, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18373091

RESUMO

PURPOSE: Targeting drugs to receptors involved in tumor angiogenesis has been demonstrated as a novel and promising approach to improve cancer treatment. In this study, we evaluated the anti-tumor efficacy of a dimeric RGD peptide-paclitaxel conjugate (RGD2-PTX) in an orthotopic MDA-MB-435 breast cancer model. METHODS: To assess the effect of conjugation and the presence of drug moiety on the MDA-MB-435 tumor and normal tissue uptake, the biodistribution of (3)H-RGD2-PTX was compared with that of (3)H-PTX. The treatment effect of RGD2-PTX and RGD2+PTX was measured by tumor size, (18)F-FDG/PET, (18)F-FLT/PET, and postmortem histopathology. RESULTS: By comparing the biodistribution of (3)H-RGD2-PTX and (3)H-PTX, we found that (3)H-RGD2-PTX had higher initial tumor exposure dose and prolonged tumor retention than (3)H-PTX. Metronomic low-dose treatment of breast cancer indicated that RGD2-PTX is significantly more effective than PTX+RGD2 combination and solvent control. Although in vivo (18)F-FLT/PET imaging and ex vivo Ki67 staining indicated little effect of the PTX-based drug on cell proliferation, (18)F-FDG/PET imaging showed significantly reduced tumor metabolism in the RGD2-PTX-treated mice versus those treated with RGD2+PTX and solvent control. Terminal uridine deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining also showed that RGD2-PTX treatment also had significantly higher cell apoptosis ratio than the other two groups. Moreover, the microvessel density was significantly reduced after RGD2-PTX treatment as determined by CD31 staining. CONCLUSION: Our results demonstrate that integrin-targeted delivery of paclitaxel allows preferential cytotoxicity to integrin-expressing tumor cells and tumor vasculature. The targeted delivery strategies developed in this study may also be applied to other chemotherapeutics for selective tumor killing.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Oligopeptídeos/administração & dosagem , Oligopeptídeos/farmacocinética , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Neoplasias da Mama/diagnóstico por imagem , Sistemas de Liberação de Medicamentos/métodos , Feminino , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Especificidade de Órgãos , Cintilografia , Distribuição Tecidual
18.
Mol Imaging Biol ; 10(2): 92-98, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18097725

RESUMO

PURPOSE: 2-deoxy-2-[(18)F]fluoro-D-glucose (FDG) is the most commonly used positron emission tomography (PET) tracer for oncological and neurological imaging, but it has limitations on detecting tumor or inflammation in brain gray matter. In this study, we describe the development of 2-deoxy-2-[(18)F]fluorosorbitol ((18)F-FDS) and its possible application in lesion detection around brain area. PROCEDURES: (18)F-FDS was obtained by reduction of FDG using NaBH(4) (81 +/- 4% yield in 30 min). Cell uptake/efflux experiments in cell culture and small animal PET imaging on tumor and inflammation models were performed. RESULTS: Despite the low accumulation in cell culture, (18)F-FDS had good tumor uptake and contrast in the subcutaneous U87MG tumor model (4.54%ID/g at 30 min post-injection). Minimal uptake in the normal mouse brain facilitated good tumor contrast in both U87MG and GL-26 orthotopic tumor models. (18)F-FDS also had increased uptake in the inflamed foci of the TPA-induced acute inflammation model. CONCLUSIONS: Because of the ease of synthesis and favorable in vivo kinetics, (18)F-FDS may have potential applications in certain cases where FDG is inadequate (e.g., brain tumor).


Assuntos
Tomografia por Emissão de Pósitrons/métodos , Sorbitol/análogos & derivados , Sorbitol/síntese química , Animais , Linhagem Celular Tumoral , Feminino , Radioisótopos de Flúor , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Transplante de Neoplasias , Sorbitol/química , Imagem Corporal Total
19.
J Med Chem ; 50(21): 5057-69, 2007 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-17867662

RESUMO

This report presents synthesis and evaluation of the 64Cu-labeled triphenylphosphonium (TPP) cations as new radiotracers for imaging tumors by positron emission tomography. Biodistribution properties of 64Cu-L1, 64Cu-L2, 64Cu-L3, and 99mTc-Sestamibi were evaluated in athymic nude mice bearing U87MG human glioma xenografts. The most striking difference is that 64Cu-L1, 64Cu-L2, and 64Cu-L3 have much lower heart uptake (<0.6% ID/g) than 99mTc-Sestamibi ( approximately 18% ID/g) at >30 min p.i. Their tumor/heart ratios increase steadily from approximately 1 at 5 min p.i. to approximately 5 at 120 min p.i. The tumor/heart ratio of 64Cu-L3 is approximately 40 times better than that of 99mTc-Sestamibi at 120 min postinjection. Results from in vitro assays show that 64Cu-L1 is able to localize in tumor mitochondria. The tumor is clearly visualized in the tumor-bearing mice administered with 64Cu-L1 as 30 min postinjection. The 64Cu-labeled TPP/TPA cations are very selective radiotracers that are able to provide the information of mitochondrial bioenergetic function in tumors by monitoring mitochondrial potential in a noninvasive fashion.


Assuntos
Arsenicais/síntese química , Radioisótopos de Cobre , Neoplasias/metabolismo , Compostos Organofosforados/síntese química , Compostos Radiofarmacêuticos/síntese química , Animais , Arsenicais/química , Arsenicais/farmacologia , Compostos Aza/síntese química , Compostos Aza/química , Compostos Aza/farmacocinética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Cátions , Linhagem Celular Tumoral , Cicloexanos/síntese química , Cicloexanos/química , Cicloexanos/farmacocinética , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Transplante de Neoplasias , Compostos Organofosforados/química , Compostos Organofosforados/farmacocinética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Relação Estrutura-Atividade , Distribuição Tecidual , Transplante Heterólogo
20.
Eur J Nucl Med Mol Imaging ; 34(12): 2024-36, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17673999

RESUMO

PURPOSE: EphA2 receptor tyrosine kinase is significantly overexpressed in a wide variety of cancer types. High EphA2 expression has been correlated with increased metastatic potential and poor patient survival. Although many recent reports have focused on blocking the EphA2 signaling pathway in cancer, the in vivo imaging of EphA2 has not yet been investigated. METHODS: We labeled 1C1, a humanized monoclonal antibody against both human and murine EphA2, with (64)Cu through the chelating agent 1,4,7,10-tetraazacyclododecane N,N',N'',N'''-tetraacetic acid (DOTA) and carried out positron emission tomography (PET) imaging of eight tumor models with different EphA2 expression levels. Western blotting of tumor tissue lysate was performed to correlate the EphA2 expression level with (64)Cu-DOTA-1C1 uptake in the tumors. Immunofluorescence staining and biodistribution studies were also carried out to validate the in vivo results. RESULTS: The radiolabeling yield was 88.9 +/- 9.5% (n = 7) and the specific activity of (64)Cu-DOTA-1C1 was 1.32 +/- 0.14 GBq/mg of 1C1 mAb. The antibody retained antigen-binding affinity/specificity after DOTA conjugation as measured by FACS analysis. The uptake of (64)Cu-DOTA-1C1 in CT-26 tumors was as high as 25.1 +/- 2.5 %ID/g (n = 3) at 18 h post injection. (64)Cu-DOTA-IgG, an isotype-matched control, exhibited minimal non-specific uptake in all eight tumor models. In vivo EphA2 specificity of (64)Cu-DOTA-1C1 was confirmed by successful blocking of CT-26 tumor uptake by unlabeled 1C1. Most importantly, the tumor uptake value obtained from PET imaging had excellent linear correlation with the relative tumor tissue EphA2 expression level measured by Western blot, where r (2) equals 0.90 and 0.92 at 18 h and 42 h post injection, respectively. CONCLUSION: The tumor uptake of (64)Cu-DOTA-1C1 measured by microPET imaging reflects tumor EphA2 expression level in vivo. This is, to our knowledge, the first report of quantitative radioimmunoPET imaging of EphA2 in living subjects. Future clinical investigation of (64)Cu-DOTA-1C1 is warranted.


Assuntos
Anticorpos Monoclonais/farmacocinética , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Compostos Organometálicos/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Radioimunodetecção/métodos , Receptor EphA2/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Especificidade de Órgãos , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA