Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Clin Cancer Res ; 26(23): 6230-6241, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32933998

RESUMO

PURPOSE: Ovarian cancer peritoneal metastases (OCPMs) are a pathophysiologically heterogeneous group of tumors that are rarely curable. αVß3 integrin (αVß3) is overexpressed on tumoral neovessels and frequently on ovarian cancer cells. Here, using two clinically relevant αVß3-positive OCPM mouse models, we studied the theranostic potential of an αVß3-specific radiopeptide, 64Cu-cyclam-RAFT-c(-RGDfK-)4 (64Cu-RaftRGD), and its intra- and intertumoral distribution in relation to the tumor microenvironment. EXPERIMENTAL DESIGN: αVß3-expressing peritoneal and subcutaneous models of ovarian carcinoma (IGR-OV1 and NIH:OVCAR-3) were established in nude mice. 64Cu-RaftRGD was administered either intravenously or intraperitoneally. We performed intratumoral distribution (ITD) studies, PET/CT imaging and quantification, biodistribution assay and radiation dosimetry, and therapeutic efficacy and toxicity studies. RESULTS: Intraperitoneal administration was an efficient route for targeting 64Cu-RaftRGD to OCPMs with excellent tumor penetration. Using the fluorescence surrogate, Cy5.5-RaftRGD, in our unique high-resolution multifluorescence analysis, we found that the ITD of 64Cu-RaftRGD was spatially distinct from, but complementary to, that of hypoxia. 64Cu-RaftRGD-based PET enabled clear visualization of multiple OCPM deposits and ascites and biodistribution analysis demonstrated an inverse correlation between tumor uptake and tumor size (1.2-17.2 mm). 64Cu-RaftRGD at a radiotherapeutic dose (148 MBq/0.357 nmol) showed antitumor activities by inhibiting tumor cell proliferation and inducing apoptosis, with negligible toxicity. CONCLUSIONS: Collectively, these results demonstrate the all-in-one potential of 64Cu-RaftRGD for imaging guided radiotherapy of OCPM by targeting both tumoral neovessels and cancerous cells. On the basis of the ITD finding, we propose that pairing αVß3- and hypoxia-targeted radiotherapies could improve therapeutic efficacy by overcoming the heterogeneity of ITD encountered with single-agent treatments.


Assuntos
Complexos de Coordenação/farmacologia , Radioisótopos de Cobre/farmacologia , Neoplasias Ovarianas/prevenção & controle , Peptídeos Cíclicos/farmacologia , Neoplasias Peritoneais/prevenção & controle , Compostos Radiofarmacêuticos/farmacologia , Animais , Apoptose , Proliferação de Células , Complexos de Coordenação/química , Radioisótopos de Cobre/química , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Peptídeos Cíclicos/química , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Compostos Radiofarmacêuticos/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Nat Commun ; 11(1): 2778, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32513979

RESUMO

The use of photothermal agents (PTAs) in cancer photothermal therapy (PTT) has shown promising results in clinical studies. The rapid degradation of PTAs may address safety concerns but usually limits the photothermal stability required for efficacious treatment. Conversely, PTAs with high photothermal stability usually degrade slowly. The solutions that address the balance between the high photothermal stability and rapid degradation of PTAs are rare. Here, we report that the inherent Cu2+-capturing ability of black phosphorus (BP) can accelerate the degradation of BP, while also enhancing photothermal stability. The incorporation of Cu2+ into BP@Cu nanostructures further enables chemodynamic therapy (CDT)-enhanced PTT. Moreover, by employing 64Cu2+, positron emission tomography (PET) imaging can be achieved for in vivo real-time and quantitative tracking. Therefore, our study not only introduces an "ideal" PTA that bypasses the limitations of PTAs, but also provides the proof-of-concept application of BP-based materials in PET-guided, CDT-enhanced combination cancer therapy.


Assuntos
Cobre/química , Hipertermia Induzida , Neoplasias/terapia , Fósforo/química , Fototerapia , Tomografia por Emissão de Pósitrons , Animais , Morte Celular , Linhagem Celular Tumoral , Terapia Combinada , Cobre/farmacocinética , Humanos , Íons , Camundongos , Nanoestruturas/química , Nanoestruturas/ultraestrutura , Oligopeptídeos/química , Fósforo/farmacocinética , Polietilenoglicóis/química , Espectrofotometria Ultravioleta , Nanomedicina Teranóstica
3.
J Nucl Med ; 60(10): 1437-1443, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30850497

RESUMO

Pancreatic cancer (PC) has a very poor prognosis. Surgery is the primary treatment for patients with resectable PC; however, local recurrence, hepatic metastasis, and peritoneal dissemination often occur even after extensive surgery. Adjuvant chemotherapy, typically with gemcitabine, has been used clinically but with only a modest survival benefit. To achieve a better outcome, we investigated the efficacy of 64Cu-intraperitoneal radioimmunotherapy (ipRIT) with 64Cu-labeled antiepidermal growth factor receptor antibody cetuximab as an adjuvant treatment after PC surgery using an orthotopic xenografted mouse model. Methods: The efficacy of adjuvant 64Cu-ipRIT was investigated in a human PC mouse model harboring orthotopic xenografts of xPA-1-DC cells. To reproduce the clinical situation, PC xenografts were surgically resected when pancreatic tumors were readily visible but not metastatic tumors. Increasing doses of 64Cu-cetuximab were intraperitoneally injected, and the mice were monitored for toxicity to determine the safe therapeutic dose. For adjuvant 64Cu-ipRIT, the day after tumor resection, the mice were intraperitoneally administered 22.2 MBq of 64Cu-PCTA-cetuximab and the survival was compared with that in surgery-only controls. For comparison, adjuvant chemotherapy with gemcitabine was also examined using the same model. Results: The mouse model not only developed primary tumors in the pancreas but also subsequently reproduced local recurrence, hepatic metastasis, and peritoneal dissemination after surgery, which is similar to the manifestations that occur with human PC. Adjuvant 64Cu-ipRIT with 64Cu-labeled cetuximab after surgery effectively suppressed local recurrence, hepatic metastasis, and peritoneal dissemination in this model. Significant improvement of the survival with minimal toxicity was achieved by adjuvant 64Cu-ipRIT compared with that in control mice that underwent surgery only. Adjuvant chemotherapy with gemcitabine nominally prolonged the survival, but the effect was not statistically significant. Conclusion:64Cu-ipRIT with cetuximab can be an effective adjuvant therapy after PC surgery.


Assuntos
Radioisótopos de Cobre/farmacologia , Injeções Intraperitoneais , Neoplasias Pancreáticas/radioterapia , Radioterapia Adjuvante , Animais , Linhagem Celular Tumoral , Cetuximab/farmacologia , Terapia Combinada , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Modelos Animais de Doenças , Receptores ErbB/química , Feminino , Células HCT116 , Humanos , Neoplasias Hepáticas/radioterapia , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Recidiva Local de Neoplasia , Transplante de Neoplasias , Radioimunoterapia , Resultado do Tratamento , Gencitabina , Neoplasias Pancreáticas
4.
Nucl Med Biol ; 39(8): 1281-5, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22727820

RESUMO

INTRODUCTION: As the use of metallic radionuclides increases, so does the demand for a simple production method. In this study, we demonstrated an in situ target processing concept for automated metallic radionuclide production without the use of any robotic device. METHODS: An alumina ceramic vessel for a vertical irradiation system was designed and developed. The ceramic vessel was evaluated by the production of Zr-89 using an yttrium powder target. The irradiated Y was dissolved remotely in HCl in the ceramic vessel and transferred as a solution to a hotcell through a Teflon tube. The crude Zr-89 was then purified by an automated apparatus. The Zr-89 was eluted with 100 µL of oxalic acid (solution) as the final product. RESULTS: The ceramic vessel gave a sufficient yield of Zr-89 (57±11MBq/µAh), showed good operability, and could be reused up to 10 times. With nominal irradiation (10µA×2h) in ~90 µL, the product (~940MBq) was obtained with >99.9% radionuclidic purity. CONCLUSION: The combination of the ceramic vessel and vertical irradiation has great potential for the remote production of various metallic radionuclides.


Assuntos
Óxido de Alumínio/química , Radioquímica/instrumentação , Radioisótopos/química , Zircônio/química , Soluções , Ítrio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA