Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Imaging Biol ; 25(4): 648-658, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37193805

RESUMO

PURPOSE: Near-infrared photoimmunotherapy (NIR-PIT) is a new cancer phototherapy using an antibody-photosensitizer conjugate (Ab-IR700). By NIR light irradiation, Ab-IR700 forms a water-insoluble aggregation on the plasma membrane of cancer cells, leading to lethal membrane damage of cancer cells with high selectivity. However, IR700 produces singlet oxygen, which induces non-selective inflammatory responses such as edema in normal tissues around the tumor. Understanding such treatment-emergent responses is important to minimize side effects and improve clinical outcomes. Thus, in this study, we evaluated physiological responses during NIR-PIT by magnetic resonance imaging (MRI) and positron emission tomography (PET). PROCEDURES: Ab-IR700 was intravenously injected into tumor-bearing mice with two tumors on the right and left sides of the dorsum. At 24 h after injection, a tumor was irradiated with NIR light. Edema formation was examined by T1/T2/diffusion-weighted MRI and inflammation was investigated by PET with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG). Because inflammation can increase vascular permeability via inflammatory mediators, we evaluated changes in oxygen levels in tumors using a hypoxia imaging probe, [18F]fluoromisonidazole ([18F]FMISO). RESULTS: The uptake of [18F]FDG in the irradiated tumor was significantly decreased compared to the control tumor, indicating the impairment of glucose metabolism induced by NIR-PIT. MRI and [18F]FDG-PET images showed that inflammatory edema with [18F]FDG accumulation was present in the surrounding normal tissues of the irradiated tumor. Furthermore, [18F]FMISO accumulation in the center of the irradiated tumor was relatively low, indicating the enhancement of oxygen supply due to increased vascular permeability. In contrast, high [18F]FMISO accumulation was observed in the peripheral region, indicating enhancement of hypoxia in the region. This could be because inflammatory edema was formed in the surrounding normal tissues, which blocked blood flow to the tumor. CONCLUSIONS: We successfully monitored inflammatory edema and changes in oxygen levels during NIR-PIT. Our findings on the acute physiological responses after light irradiation will help to develop effective measures to minimize the side effects in NIR-PIT.


Assuntos
Imunoconjugados , Neoplasias , Animais , Camundongos , Fluordesoxiglucose F18 , Linhagem Celular Tumoral , Fototerapia/métodos , Imunoterapia/métodos , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias/terapia , Neoplasias/tratamento farmacológico
2.
Int J Pharm ; 609: 121135, 2021 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-34571072

RESUMO

Near-infrared photoimmunotherapy (NIR-PIT) is a cancer phototherapy that uses antibody-IR700 conjugate (Ab-IR700) and NIR light. Ab-IR700 forms aggregates on the plasma membranes of targeted cancer cells after light exposure, inducing lethal physical damage within the membrane. Low-molecular-weight (LMW) ligands are candidate targeting moieties instead of antibodies, but whether LMW-IR700 conjugates induce cell death by aggregation, the same mechanism as Ab-IR700, is unknown. Thus, we investigated differences in cytotoxicity and mechanisms between LMW-IR700 and Ab-IR700 targeting prostate-specific membrane antigen (PSMA). Both conjugates decreased cell viability to the same degree after light irradiation, but different morphological changes were observed in PSMA-positive LNCaP cells by microscopy. Cell swelling and bleb formation were induced by Ab-IR700, but only swelling was observed in cells treated with LMW-IR700, suggesting the cells were damaged via different cytotoxic mechanisms. However, LMW-IR700 induced bleb formation, a hallmark of NIR-PIT with Ab-IR700, when singlet oxygen was quenched or LMW-IR700 was localized only on the plasma membrane. Moreover, the water-soluble axial ligands of LMW-IR700 were cleaved, consistent with previous reports on Ab-IR700. Thus, the main cytotoxic mechanisms of Ab-IR700 and LMW-IR700 differ, although LMW-IR700 on the plasma membrane can cause aggregation-mediated cytotoxicity as well as Ab-IR700.


Assuntos
Antígenos de Superfície , Glutamato Carboxipeptidase II , Imunoterapia , Fármacos Fotossensibilizantes , Fototerapia , Neoplasias da Próstata , Linhagem Celular Tumoral , Humanos , Raios Infravermelhos , Ligantes , Masculino , Terapia de Alvo Molecular , Neoplasias da Próstata/terapia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Bioorg Med Chem ; 43: 116274, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34139484

RESUMO

Photoimmunotherapy (PIT) is a new molecular-targeted phototherapy in which administration of an antibody conjugated to IR700 (Ab-IR700, a phthalocyanine derivative) is followed by irradiation with near-infrared light. PIT induces cell death due to cell membrane damage, and the formation of IR700 aggregates on the cell membrane triggered by photochemical reactions is an important mechanism of cell killing. Specifically, water-soluble axial ligands of IR700 are cleaved by the photochemical reaction, and the phthalocyanine stacks up due to the π-π interaction, resulting in the formation of aggregates. In addition, the formation of IR700 radical anions and their protonation are essential for the progress of this photochemical reaction. The elucidation of these mechanisms may lead to the development of more effective compounds in the future. In addition, the optical properties of phthalocyanine are expected to expand the medical application of phthalocyanine derivatives in the future.


Assuntos
Antineoplásicos/uso terapêutico , Imunoterapia , Neoplasias/terapia , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia , Animais , Antineoplásicos/química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Fármacos Fotossensibilizantes/química , Relação Estrutura-Atividade
4.
Chempluschem ; 85(9): 1953, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32830450

RESUMO

Invited for this month's cover are the collaborating groups of Dr. Masato Kobayashi and Prof. Mikako Ogawa, both from Hokkaido University, Sapporo, Japan. The cover picture shows the photochemical reaction process of the near-infrared (NIR) photoimmunotherapy dye IR700, and subsequent cancer cell death. A computational study predicted that ligand dissociation, which is known to initiate cancer cell death, proceeds by the hydrolysis of the IR700 radical anion, rather than as a direct result of NIR irradiation. This mechanism has also been supported by experimental work. Read the full text of the Communication at 10.1002/cplu.202000338.


Assuntos
Imunoterapia , Indóis/química , Raios Infravermelhos , Modelos Teóricos , Compostos de Organossilício/química , Fármacos Fotossensibilizantes/farmacologia , Fototerapia , Ânions , Humanos , Hidrólise
5.
Chempluschem ; 85(9): 1959-1963, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32449613

RESUMO

Ligand release from IR700, a silicon phthalocyanine dye used in near-infrared (NIR) photoimmunotherapy, initiates cancer cell death after NIR absorption, although its photochemical mechanism has remained unclear. This theoretical study reveals that the direct Si-ligand dissociation by NIR light is difficult to activate because of the high dissociation energy even in excited states, i. e., >1.30 eV. Instead, irradiation generates the IR700 radical anion, leading to acid-base reactions with nearby water molecules (i. e., calculated pKb for the radical anion is 7.7) to produce hydrophobic ligand-released dyes. This suggests two possibilities: (1) water molecules participate in ligand release and (2) light is not required for Si-ligand dissociation as formation of the IR700 radical anion is sufficient. Experimental evidence confirmed possibility (1) by using 18 O-labeled water as the solvent, while (2) is supported by the pH dependence of ligand exchange, providing a complete description of the Si-ligand bond dissociation mechanism.


Assuntos
Imunoterapia , Indóis/química , Raios Infravermelhos , Modelos Teóricos , Compostos de Organossilício/química , Fármacos Fotossensibilizantes/farmacologia , Fototerapia , Ânions , Humanos , Hidrólise
6.
Cancer Sci ; 109(9): 2889-2896, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29949672

RESUMO

Near-infrared photoimmunotherapy (NIR-PIT) is a new cancer phototherapy modality using an antibody conjugated to a photosensitizer, IRDye700DX. When the conjugate binds to the plasma membrane and is exposed to NIR light, NIR-PIT-treated cells undergo swelling, and target-selective necrotic/immunogenic cell death is induced. However, the cytotoxic mechanism of NIR-PIT has not been elucidated. In order to understand the mechanism, it is important to elucidate how the damage to the plasma membrane induced by NIR light irradiation changes over time. Thus, in the present study, we investigated the changes in plasma membrane permeability using ions and molecules of various sizes. Na+ flowed into cells immediately after NIR light irradiation, even when the function of transporters or channels was blocked. Subsequently, fluorescent molecules larger than Na+ entered the cells, but the damage was not large enough for dextran to pass through at early time points. To assess these phenomena quantitatively, membrane permeability was estimated using radiolabeled ions and molecules: 111 InCl3 , 111 In-DTPA, and 3 H-H2 O, and comparable results were obtained. Although minute plasma membrane perforations usually do not induce cell death, our results suggest that the minute damage induced by NIR-PIT was irreversibly extended with time. In conclusion, minute plasma membrane damage is a trigger for the increase in plasma membrane permeability, cell swelling, and necrotic/immunogenic cell death in NIR-PIT. Our findings provide new insight into the cytotoxic mechanism of NIR-PIT.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Membrana Celular/patologia , Imunoterapia/efeitos adversos , Indóis/toxicidade , Transporte de Íons/efeitos dos fármacos , Compostos de Organossilício/toxicidade , Fototerapia/efeitos adversos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Imunoterapia/métodos , Indóis/uso terapêutico , Compostos de Organossilício/uso terapêutico , Fototerapia/métodos , Sódio/metabolismo , Trastuzumab/uso terapêutico
7.
Oncotarget ; 9(28): 20048-20057, 2018 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-29732002

RESUMO

PURPOSE: The aim of this study was to develop and assess a novel implantable, wireless-powered, light-emitting diode (LED) for near-infrared photoimmunotherapy (NIR-PIT). NIR-PIT is a recently developed cancer therapy that uses NIR light and antibody-photosensitizer conjugates and is able to induce cancer-specific cell death. Due to limited light penetration depth it is currently unable to treat tumors in deep tissues. Use of implanted LED might potentially overcome this limitation. RESULTS: The wireless LED system was able to emit NIR light up to a distance of 20 cm from the transmitter coil by using low magnetic fields as compliant with limits for use in humans. Results indicated that the LED system was able to kill tumor cells in vitro and to suppress tumor growth in implanted tumor-bearing mice. CONCLUSIONS: Results indicated that the proposed implantable wireless LED system was able to suppress tumor growth in vivo. These results are encouraging as wireless LED systems such as the one here developed might be a possible solution to treat tumors in deep regions in humans. Further research in this area would be important. MATERIALS AND METHODS: An implantable LED system was developed. It consisted of a LED capsule including two LED sources and a receiver coil coupled with an external coil and power source. Wireless power transmission was guaranteed by using electromagnetic induction. The system was tested in vitro by using EGFR-expressing cells and HER2-expressing cells. The system was also tested in vivo in tumor-bearing mice.

8.
Methods Mol Biol ; 1272: 257-70, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25563190

RESUMO

The family of G protein-coupled receptors (GPCRs) remains a central focus of basic pharmacology and drug discovery efforts. Convenient methods to assess the efficacy of potentially therapeutic reagents for GPCRs are strongly required for high-throughput screening (HTS) assay. We recently developed a rapid, sensitive, and quantitative method for detecting potential chemicals that act on GPCRs using split luciferase complementation. In principle, this is based on the detection of interactions of GPCR with ß-arrestin, which translocates to the activated GPCRs. This method can facilitate the construction of HTS systems in a multi-well plate format. Particularly, the method is compatible with single-cell imaging and animal models and even deeper tissues such as organs, because of its high sensitivity, suggesting that promising candidates from HTS assay can be moved easily to the next phase for additional analysis. This system can contribute to the effective evaluation of potentially therapeutic reagents and expedite the development of new drugs for GPCRs.


Assuntos
Arrestinas/metabolismo , Ensaios de Triagem em Larga Escala , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Somatostatina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Análise de Célula Única/métodos , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Arrestinas/genética , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Feminino , Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Isoproterenol/farmacologia , Luciferases/genética , Luciferases/metabolismo , Medições Luminescentes , Camundongos Endogâmicos BALB C , Plasmídeos/química , Plasmídeos/metabolismo , Propranolol/farmacologia , Receptores Adrenérgicos beta 2/genética , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Proteínas Recombinantes de Fusão/genética , Somatostatina/farmacologia , Transfecção , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA